Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence

Information

  • Patent Grant
  • 11912985
  • Patent Number
    11,912,985
  • Date Filed
    Monday, November 7, 2022
    a year ago
  • Date Issued
    Tuesday, February 27, 2024
    2 months ago
Abstract
The present disclosure provides systems, compositions, and methods for simultaneously editing both strands of a double-stranded DNA sequence at a target site to be edited. Further provided herein are pharmaceutical compositions, polynucleotides, vectors, cells, and kits for simultaneously editing both strands of a double-stranded DNA sequence.
Description
REFERENCE TO AN ELECTRONIC SEQUENCE LISTING

The contents of the electronic sequence listing (B119570091US02-SUBSEQ-TNG.xml; Size: 5,535,149 bytes; and Date of Creation: Oct. 13, 2023) is herein incorporated by reference in its entirety.


BACKGROUND OF THE INVENTION

Pathogenic single nucleotide mutations contribute to approximately 50% of human diseases for which there is a genetic component,7 according to some estimates. Unfortunately, treatment options for patients with these genetic disorders remain extremely limited, despite decades of gene therapy exploration'. Perhaps the most parsimonious solution to this therapeutic challenge is direct correction of single nucleotide mutations in patient genomes, which would address the root cause of disease and would likely provide lasting benefit. Although such a strategy was previously unthinkable, recent improvements in genome editing capabilities brought about by the advent of the CRISPR/Cas system9 have now brought this therapeutic approach within reach. By straightforward design of a guide RNA (gRNA) sequence that contains ˜20 nucleotides complementary to the target DNA sequence, nearly any conceivable genomic site can be specifically accessed by CRISPR associated (Cas) nucleases1,2. To date, several monomeric bacterial Cas nuclease systems have been identified and adapted for genome editing applications10. This natural diversity of Cas nucleases, along with a growing collection of engineered variants11-14, offers fertile ground for developing new genome editing technologies.


While gene disruption with CRISPR is now a mature technique, precision editing of single base pairs in the human genome remains a major challenge3. Homology directed repair (HDR) has long been used in human cells and other organisms to insert, correct, or exchange DNA sequences at sites of double strand breaks (DSBs) using donor DNA repair templates that encode the desired edits15. However, traditional HDR has very low efficiency in most human cell types, particularly in non-dividing cells, and competing non-homologous end joining (NHEJ) leads predominantly to insertion-deletion (indel) byproducts16. Other issues relate to the generation of DSBs, which can give rise to large chromosomal rearrangements and deletions at target loci17, or activate the p53 axis leading to growth arrest and apoptosis18,19.


Several approaches have been explored to address these drawbacks of HDR. For example, repair of single-stranded DNA breaks (nicks) with oligonucleotide donors has been shown to reduce indel formation, but yields of desired repair products remain low20. Other strategies attempt to bias repair toward HDR over NHEJ using small molecule and biologic reagents21-23. However, the effectiveness of these methods is likely cell-type dependent, and perturbation of the normal cell state could lead to undesirable and unforeseeable effects.


Recently, the inventors, led by Prof. David Liu et al., developed base editing as a technology that edits target nucleotides without creating DSBs or relying on HDR4-6,24-27. Direct modification of DNA bases by Cas-fused deaminase enzymes allows for C•G to T•A, or A•T to G•C, base pair conversions in a short target window (˜5-7 bases) with very high efficiency. As a result, base editors have been rapidly adopted by the scientific community. However, the following factors limit their generality for precision genome editing: (1) “bystander editing” of non-target C or A bases within the target window are observed; (2) target nucleotide product mixtures are observed; (3) target bases must be located 15±2 nucleotides upstream of a PAM sequence; and (5) repair of small insertion and deletion mutations is not possible.


Therefore, the development of programmable editors that are flexibly capable of introducing any desired single nucleotide change and/or which could install base pair insertions or deletions (e.g., at least 1, 2, 3, 4, 5, 6, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50, 60, 70, 80, 90, 100, or more base pair insertions or deletions) and/or which could alter or modify the nucleotide sequence at a target site with high specificity and efficiency would substantially expand the scope and therapeutic potential of genome editing technologies based on CRISPR.


SUMMARY OF THE INVENTION

The present invention describes a new platform for genome editing called “multi-flap prime editing” (including, for example, “dual-flap prime editing” and “quadruple-flap prime editing”) and represents an innovative advancement of “prime editing” or “classical prime editing,” as described by the present inventors in Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149-157 (2019), which is incorporated herein by reference. Whereas classical prime editing in various embodiments polymerizes at a nick site a single 3′ flap which becomes integrated into the target nucleic acid on the same strand, the presently described multi-flap prime editing systems involve distinct constructs, systems, and methodologies that, in various embodiments, generate pairs or multiple pairs of 3′ flaps on different strands, which form duplexes comprising desired edits and which become incorporated into target nucleic acid molecules, e.g., at specific loci or edit sites in a genome. In various aspects, the pairs or multiple pairs of 3′ flaps form duplexes because they comprise reverse complementary sequences which anneal to one another once generated by the prime editors described herein. The duplexes become incorporated into the target site by cell-driven mechanisms that naturally replace the endogenous duplex sequences located between adjacent nick sites. In certain embodiments, the new duplex sequences may be introduced at one or more locations (e.g., at adjacent genomic loci or on two different chromosomal locations), and may comprise one or more sequences of interest, e.g., protein-encoding sequence, peptide-encoding sequence, or RNA-encoding sequence. In one embodiment, the new duplex sequences installed by the multi-flap prime editing systems may comprise a recombinase site, e.g., a Bxb1 recombinase attB (38 bp) and/or attP (50 bp) site, or a recombinase site recognized by Hin recombinase, Gin recombinase, Tn3 recombinase, β-six recombinase, CinH recombinase, ParA recombinase, γδ recombinase, ϕC31 recombinase, TP901 recombinase, TG1 recombinase, φBT1 recombinase, R4 recombinase, φRV1 recombinase, φFC1 recombinase, MR11 recombinase, A118 recombinase, U153 recombinase, and gp29 recombinase, Cre recombinase, FLP recombinase, R recombinase, Lambda recombinase, HK101 recombinase, HK022 recombinase, and pSAM2 recombinase.


The inventors recently developed prime editing which enables the insertion, deletion, or replacement of genomic DNA sequences without requiring error-prone double-strand DNA breaks. Prime editing uses an engineered Cas9 nickase—reverse transcriptase fusion protein (PE1 or PE2) paired with an engineered prime editing guide RNA (pegRNA) that both directs Cas9 to the target genomic site and encodes the information for installing the desired edit. Prime editing proceeds through a multi-step editing process: 1) the Cas9 domain binds and nicks the target genomic DNA site, which is specified by the pegRNA's spacer sequence; 2) the reverse transcriptase domain uses the nicked genomic DNA as a primer to initiate the synthesis of an edited DNA strand using an engineered extension on the pegRNA as a template for reverse transcription—this generates a single-stranded 3′ flap containing the edited DNA sequence; 3) cellular DNA repair resolves the 3′ flap intermediate by the displacement of a 5′ flap species that occurs via invasion by the edited 3′ flap, excision of the 5′ flap containing the original DNA sequence, and ligation of the new 3′ flap to incorporate the edited DNA strand, forming a heteroduplex of one edited and one unedited strand; and 4) cellular DNA repair replaces the unedited strand within the heteroduplex using the edited strand as a template for repair, completing the editing process.


Efficient incorporation of the desired edit requires that the newly synthesized 3′ flap contains a portion of sequence that is homologous to the genomic DNA site. This homology enables the edited 3′ flap to compete with the endogenous DNA strand (the corresponding 5′ flap) for incorporation into the DNA duplex. Because the edited 3′ flap will contain less sequence homology than the endogenous 5′ flap, the competition is expected to favor the 5′ flap strand. Thus, a potential limiting factor in the efficiency of prime editing may be the efficiency of the invasion of the 3′ flap of the endogenous DNA and the subsequent displacement and replacement of the 5′ flap strand. Moreover, successful 3′ flap invasion and removal of the 5′ flap only incorporates the edit on one strand of the double-stranded DNA genome. Permanent installation of the edit requires cellular DNA repair to replace the unedited complementary DNA strand using the edited strand as a template. While the cell can be made to favor replacement of the unedited strand over the edited strand (step 4 above) by the introduction of a nick in the unedited strand adjacent to the edit using a secondary sgRNA (the PE3 system), this process still relies on a second stage of DNA repair. These DNA repair steps may be particularly inefficient for edits which require equilibration of long 5′ and 3′ flap intermediates or contain long non-homologous regions, such as long insertions or long deletions. Further developments in prime editing would advance the art.


In various aspects, this Specification describes a multi-flap prime editing system (including, for example, dual prime editing systems and quadruple prime editing systems) that addresses the challenges associated with flap equilibration and subsequent incorporation of the edit into the non-edited complementary genomic DNA strand by simultaneously editing both DNA strands. In the dual-flap prime editing system, for example, two pegRNAs are used to target opposite strands of a genomic site and direct the synthesis of two complementary 3′ flaps containing edited DNA sequence (FIG. 91). Unlike classical prime editing, there is no requirement for the pair of edited DNA strands (3′ flaps) to directly compete with 5′ flaps in endogenous genomic DNA, as the complementary edited strand is available for hybridization instead. Since both strands of the duplex are synthesized as edited DNA, the dual-flap prime editing system obviates the need for the replacement of the non-edited complementary DNA strand required by classical prime editing. Instead, cellular DNA repair machinery need only excise the paired 5′ flaps (original genomic DNA) and ligate the paired 3′ flaps (edited DNA) into the locus. Therefore, there is also no need to include sequences homologous to genomic DNA in the newly synthesized DNA strands, allowing selective hybridization of the new strands and facilitating edits that contain minimal genomic homology. Nuclease-active versions of prime editors that cut both strands of DNA could also be used to accelerate the removal of the original DNA sequence. The quadruple-flap prime editing system, using four pegRNAs, provides similar advantages.


Like classical prime editing, multi-flap prime editing is a versatile and precise genome editing method that directly writes new genetic information into a specified DNA site using a nucleic acid programmable DNA binding protein (“napDNAbp”) working in association with a polymerase (i.e., in the form of a fusion protein or otherwise provided in trans with the napDNAbp), wherein the prime editing system is programmed with a prime editing (PE) guide RNA (“PEgRNA”) that both specifies the target site and templates the synthesis of the desired edit in the form of a replacement DNA strand by way of an extension (either DNA or RNA) engineered onto a guide RNA (e.g., at the 5′ or 3′ end, or at an internal portion of a guide RNA). The replacement strand containing the desired edit (e.g., a single nucleobase substitution) shares the same sequence as the endogenous strand of the target site to be edited (with the exception that it includes the desired edit). Through DNA repair and/or replication machinery, the endogenous strand of the target site is replaced by the newly synthesized replacement strand containing the desired edit. In some cases, prime editing may be thought of as a “search-and-replace” genome editing technology since the prime editors, as described herein, not only search and locate the desired target site to be edited, but at the same time, encode a replacement strand containing a desired edit, which is installed in place of the corresponding target site endogenous DNA strand.


The multi-flap prime editors of the present disclosure relate, in part, to the discovery that the mechanism of target-primed reverse transcription (TPRT) or “prime editing” can be leveraged or adapted for conducting precision CRISPR/Cas-based genome editing with high efficiency and genetic flexibility (e.g., as depicted in various embodiments of FIGS. 1A-1F). TPRT is naturally used by mobile DNA elements, such as mammalian non-LTR retrotransposons and bacterial Group II introns28,29. The inventors have herein used Cas protein-reverse transcriptase fusions or related systems to target a specific DNA sequence with a guide RNA, generate a single strand nick at the target site, and use the nicked DNA as a primer for reverse transcription of an engineered reverse transcriptase template that is integrated with the guide RNA. However, while the concept begins with prime editors that use reverse transcriptases as the DNA polymerase component, the multi-flap prime editors described herein are not limited to reverse transcriptases but may include the use of virtually any DNA polymerase. Indeed, while the application throughout may refer to multi-flap prime editors with “reverse transcriptases,” it is set forth here that reverse transcriptases are only one type of DNA polymerase that may work with multi-flap prime editing. Thus, whereever the specification mentions “reverse transcriptases,” the person having ordinary skill in the art should appreciate that any suitable DNA polymerase may be used in place of the reverse transcriptase. Thus, in one aspect, the multi-flap prime editors may comprise Cas9 (or an equivalent napDNAbp) which is programmed to target a DNA sequence by associating it with a specialized guide RNA (i.e., PEgRNA) containing a spacer sequence that anneals to a complement of a protospacer sequence in the target DNA. The specialized guide RNA also contains new genetic information in the form of an extension that encodes a replacement strand of DNA containing a desired genetic alteration which is used to replace a corresponding endogenous DNA strand at the target site. To transfer information from the PEgRNA to the target DNA, the mechanism of multi-flap prime editing involves nicking the target site in one strand of the DNA to expose a 3′-hydroxyl group. The exposed 3′-hydroxyl group can then be used to prime the DNA polymerization of the edit-encoding extension on PEgRNA directly into the target site. In various embodiments, the extension—which provides the template for polymerization of the replacement strand containing the edit—can be formed from RNA or DNA. In the case of an RNA extension, the polymerase of the prime editor can be an RNA-dependent DNA polymerase (such as, a reverse transcriptase). In the case of a DNA extension, the polymerase of the prime editor may be a DNA-dependent DNA polymerase.


In classical prime editing, the newly synthesized strand (i.e., the replacement DNA strand containing the desired edit) that is formed by the herein disclosed prime editors would be homologous to the genomic target sequence (i.e., have the same sequence as) except for the inclusion of a desired nucleotide change (e.g., a single nucleotide change, a deletion, or an insertion, or a combination thereof). The newly synthesized (or replacement) strand of DNA may also be referred to as a single strand DNA flap, which would compete for hybridization with the complementary homologous endogenous DNA strand, thereby displacing the corresponding endogenous strand. In certain embodiments, the system can be combined with the use of an error-prone reverse transcriptase enzyme (e.g., provided as a fusion protein with the Cas9 domain, or provided in trans to the Cas9 domain). The error-prone reverse transcriptase enzyme can introduce alterations during synthesis of the single strand DNA flap. Thus, in certain embodiments, error-prone reverse transcriptase can be utilized to introduce nucleotide changes to the target DNA. Depending on the error-prone reverse transcriptase that is used with the system, the changes can be random or non-random.


In classical prime editing, resolution of the hybridized intermediate (comprising the single strand DNA flap synthesized by the reverse transcriptase hybridized to the endogenous DNA strand) can include removal of the resulting displaced flap of endogenous DNA (e.g., with a 5′ end DNA flap endonuclease, FEN1), ligation of the synthesized single strand DNA flap to the target DNA, and assimilation of the desired nucleotide change as a result of cellular DNA repair and/or replication processes. Because templated DNA synthesis offers single nucleotide precision for the modification of any nucleotide, including insertions and deletions, the scope of this approach is very broad and could foreseeably be used for myriad applications in basic science and therapeutics.


In some aspects, the specification provides a pair of prime editors, each comprising a nucleic acid programmable DNA binding protein (napDNAbp) and a DNA polymerase. In some embodiments, each prime editor is capable of carrying out genome editing by target-primed reverse transcription in the presence of an extended guide RNA.


In some aspects, the specification provides a pair of prime editors, each comprising a nucleic acid programmable DNA binding protein (napDNAbp) and a DNA polymerase, wherein the DNA polymerase is provided in trans with the napDNAbp. In various embodiments, each prime editor is capable of carrying out genome editing by target-primed reverse transcription in the presence of an extended guide RNA.


In some aspects, the specification provides a pair of prime editors, each comprising a nucleic acid programmable DNA binding protein (napDNAbp) and a reverse transcriptase. In various embodiments, each prime editor is capable of carrying out genome editing by target-primed reverse transcription in the presence of an extended guide RNA.


In some aspects, the specification provides a pair of prime editors, each comprising a nucleic acid programmable DNA binding protein (napDNAbp) and a reverse transcriptase, wherein the reverse transcriptase is provided in trans with the napDNAbp. In various embodiments, each prime editor is capable of carrying out genome editing by target-primed reverse transcription in the presence of an extended guide RNA.


In certain embodiments, the napDNAbp has a nickase activity. The napDNAbp may also be a Cas9 protein or functional equivalent thereof, such as a nuclease active Cas9, a nuclease inactive Cas9 (dCas9), or a Cas9 nickase (nCas9).


In certain embodiments, the napDNAbp is selected from the group consisting of: Cas9, Cas12e, Cas12d, Cas12a, Cas12b1, Cas13a, Cas12c, and Argonaute and optionally has a nickase activity.


In other embodiments, each prime editor of the dual prime editors, when complexed with an extended guide RNA, is capable of binding to a target DNA sequence.


In still other embodiments, the target DNA sequence comprises a target strand and a complementary non-target strand.


In other embodiments, the binding of the prime editor complexed to the extended guide RNA forms an R-loop. The R-loop can comprise (i) an RNA-DNA hybrid comprising the extended guide RNA and the target strand, and (ii) the complementary non-target strand.


In still other embodiments, the complementary non-target strand is nicked to form a reverse transcriptase priming sequence having a free 3′ end.


In various embodiments, the extended guide RNA comprises (a) a guide RNA and (b) an RNA extension at the 5′ or the 3′ end of the guide RNA, or at an intramolecular location in the guide RNA. The RNA extension can comprise (i) a reverse transcription template sequence comprising a desired nucleotide change, (ii) a reverse transcription primer binding site, and (iii) optionally, a linker sequence. In various embodiments, the reverse transcription template sequence may encode a single-strand DNA flap that is complementary to an endogenous DNA sequence adjacent to the nick site, wherein the single-strand DNA flap comprises the desired nucleotide change.


In various embodiments, the RNA extension is at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, or at least 25 nucleotides in length.


In still other embodiments, the single-strand DNA flap may hybridize to the endogenous DNA sequence adjacent to the nick site, thereby installing the desired nucleotide change. In still other embodiments, the single-stranded DNA flap displaces the endogenous DNA sequence adjacent to the nick site and which has a free 5′ end. In certain embodiments, the displaced endogenous DNA having the 5′ end is excised by the cell.


In various embodiments, the cellular repair of the single-strand DNA flap results in installation of the desired nucleotide change, thereby forming a desired product.


In various other embodiments, the desired nucleotide change is installed in an editing window that is between about −4 to +10 of the PAM sequence.


In still other embodiments, the desired nucleotide change is installed in an editing window that is between about −5 to +5 of the nick site, or between about −10 to +10 of the nick site, or between about −20 to +20 of the nick site, or between about −30 to +30 of the nick site, or between about −40 to +40 of the nick site, or between about −50 to +50 of the nick site, or between about −60 to +60 of the nick site, or between about −70 to +70 of the nick site, or between about −80 to +80 of the nick site, or between about −90 to +90 of the nick site, or between about −100 to +100 of the nick site, or between about −200 to +200 of the nick site.


In various embodiments, the napDNAbp of the dual prime editors each comprise an amino acid sequence of SEQ ID NO: 18. In various other embodiments, the napDNAbp comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs: 26-39, 42-61, 75-76, 126, 130, 137, 141, 147, 153, 157, 445, 460, 467, and 482-487 (Cas9); SEQ ID NO: 77-86 (CP-Cas9); SEQ ID NO: 18-25 and 87-88 (SpCas9); and SEQ ID NOs: 62-72(Cas12)


In other embodiments, the reverse transcriptase of the disclosed prime editors and/or compositions of the dual prime editors may comprise any one of the amino acid sequences of SEQ ID NOs: 89-100, 105-122, 128-129, 132, 139, 143, 149, 154, 159, 235, 454, 471, 516, 662, 700-716, 739-742, and 766. In still other embodiments, the reverse transcriptase may comprise an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs: 89-100, 105-122, 128-129, 132, 139, 143, 149, 154, 159, 235, 454, 471, 516, 662, 700-716, 739-742, and 766. These sequences may be naturally occurring reverse transcriptase sequences, e.g., from a retrovirus or a retrotransposon, of the sequences may be recombinant.


In various other embodiments, the prime editors of the dual prime editors herein disclosed may comprise various structural configurations. For example, in embodiments in which the prime editors are provided as a fusion protein, each of the dual prime editor fusion proteins may comprise the structure NH2-[napDNAbp]-[reverse transcriptase]-COOH; or NH2-[reverse transcriptase]-[napDNAbp]-COOH, wherein each instance of “]-[” indicates the presence of an optional linker sequence.


In various embodiments, the linker sequence comprises an amino acid sequence of SEQ ID NOs: 127, 165-176, 446, 453, and 767-769, or an amino acid sequence that this at least 80%, 85%, or 90%, or 95%, or 99% identical to any one of the linker amino acid sequence of SEQ ID NOs: 127, 165-176, 446, 453, and 767-769.


In various embodiments, the desired nucleotide change that is incorporated into the target DNA can be a single nucleotide change (e.g., a transition or transversion), an insertion of one or more nucleotides, or a deletion of one or more nucleotides.


In certain cases, the insertion is at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 200, at least 300, at least 400, or at least 500 nucleotides in length.


In certain other cases, the deletion is at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 200, at least 300, at least 400, or at least 500 nucleotides in length.


In another aspect, the present disclosure provides an extended guide RNA comprising a guide RNA and at least one RNA extension. The RNA extension can be positioned at the 3′ end of the guide RNA. In other embodiments, the RNA extension can be positioned at the 5′ of the guide RNA. In still other embodiments, the RNA extension can be positioned at an intramolecular position within the guide RNA, however, preferable, the intramolecular positioning of the extended portion does not disrupt the functioning of the protospacer.


In various embodiments, the extended guide RNA is capable of binding to a napDNAbp and directing the napDNAbp to a target DNA sequence. The target DNA sequence can comprise a target strand and a complementary non-target strand, wherein the guide RNA hybridizes to the target strand to form an RNA-DNA hybrid and an R-loop.


In various embodiments of the extended guide RNA, the at least one RNA extension can comprise a reverse transcription template sequence. In various other embodiment, the RNA extension may further comprises a reverse transcription primer binding site. In still further embodiments, the RNA extension may comprise a linker or spacer sequence that joins the RNA extension to the guide RNA.


In various embodiments, the RNA extension can be at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides, at least 90 nucleotides, at least 100 nucleotides, at least 150 nucleotides, at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, or at least 500 nucleotides in length.


In other embodiments, the reverse transcription template sequence is at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides, at least 90 nucleotides, at least 100 nucleotides, at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, or at least 500 nucleotides in length.


In still other embodiments, wherein the reverse transcription primer binding site sequence is at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides, at least 90 nucleotides, at least 100 nucleotides, at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, or at least 500 nucleotides in length.


In other embodiments, the optional linker or spacer sequence is at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides, at least 90 nucleotides, at least 100 nucleotides, at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, or at least 500 nucleotides in length.


In various embodiments of the extended guide RNAs, the reverse transcription template sequence may encode a single-strand DNA flap that is complementary to an endogenous DNA sequence adjacent to a nick site, wherein the single-strand DNA flap comprises a desired nucleotide change. The single-stranded DNA flap may displace an endogenous single-strand DNA at the nick site. The displaced endogenous single-strand DNA at the nick site can have a 5′ end and form an endogenous flap, which can be excised by the cell. In various embodiments, excision of the 5′ end endogenous flap can help drive product formation since removing the 5′ end endogenous flap encourages hybridization of the single-strand 3′ DNA flap to the corresponding complementary DNA strand, and the incorporation or assimilation of the desired nucleotide change carried by the single-strand 3′ DNA flap into the target DNA.


In various embodiments of the extended guide RNAs, the cellular repair of the single-strand DNA flap results in installation of the desired nucleotide change, thereby forming a desired product.


In certain embodiments, the PEgRNA comprises the nucleotide sequence of SEQ ID NOs: 101-104, 181-183, 223-234, 237-244, 277, 324-330, 332, 334, 336, 338, 340, 342, 344, 346, 348, 350, 352, 354, 356, 358, 360, 362, 364, 366, 368, 394, 429-442, 499-505, 641-649, 678-692, 735-736, 757-761, 776-777, 2997-3103, 3113-3121, 3305-3455, 3479-3493, 3522-3540, 3549-3556, 3628-3698, 3755-3810, 3874, 3890-3901, 3905-3911, 3913-3929, and 3972-3989 or a nucleotide sequence having at least 85%, or at least 90%, or at least 95%, or at least 98%, or at least 99% sequence identity with any one of SEQ ID NOs: 101-104, 181-183, 223-234, 237-244, 277, 324-330, 332, 334, 336, 338, 340, 342, 344, 346, 348, 350, 352, 354, 356, 358, 360, 362, 364, 366, 368, 394, 429-442, 499-505, 641-649, 678-692, 735-736, 757-761, 776-777, 2997-3103, 3113-3121, 3305-3455, 3479-3493, 3522-3540, 3549-3556, 3628-3698, 3755-3810, 3874, 3890-3901, 3905-3911, 3913-3929, and 3972-3989.


In yet another aspect of the invention, the specification provides for complexes comprising a prime editor described herein and any extended guide RNA described above.


In still other aspects of the invention, the specification provides a complex comprising a napDNAbp and an extended guide RNA. The napDNAbp can be a Cas9 nickase, or can be an amino acid sequence of SEQ ID NOs: 42-57 (Cas9 nickase) and 65 (AsCas12a nickase), or an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs: 42-57 (Cas9 nickase) and 65 (AsCas12a nickase).


In various embodiments involving a complex, the extended guide RNA is capable of directing the napDNAbp to a target DNA sequence. In various embodiments, a reverse transcriptase may be provided in trans, i.e., provided from a different source than the complex itself. For example, a reverse transcriptase could be provided to the same cell having the complex by introducing a separate vector separately encoding the reverse transcriptase.


In another aspect, the disclosure provides a system comprising a first and a second prime editor complex, each complex comprising a prime editor and a prime editing guide RNA (PEgRNA). In some embodiments, each prime editor comprises a nucleic acid programmable DNA binding protein (napDNAbp) and a polypeptide having an RNA-dependent DNA polymerase activity, and each PEgRNA comprises a spacer sequence, a gRNA core, a DNA synthesis template, and a primer binding site. In certain embodiments, each DNA synthesis template encodes a single-stranded DNA sequence comprising an edited portion. Two single-stranded DNA sequences encoded may by complementary to one another and form a duplex that integrates into the target site to be edited. In some embodiments, the two single-stranded DNA sequences encoded may comprise a region of complementarity to one another. In certain embodiments, the two single-stranded DNA sequences encoded may comprise a region of complementarity to one another that is at least 2 bp, at least 3 bp, at least 4 bp, at least 5 bp, at least 10 bp, at least 20 bp, at least 30 bp, at least 40 bp, at least 50 bp, at least 100 bp, at least 200 bp, at least 300 bp, at least 400 bp, at least 500 bp, at least 600 bp, at least 700 bp, at least 800 bp, at least 900 bp, or at least 1000 bp long. In some embodiments, the prime editor is provided as a fusion protein. In certain embodiments, the components of the prime editor (i.e., the napDNAbp and the polypeptide having an RNA-dependent DNA polymerase activity) are provided in trans.


In another aspect, the disclosure provides a system comprising a first, a second, a third, and a fourth prime editor complex, each complex comprising a prime editor and a prime editing guide RNA (PEgRNA). In some embodiments, each prime editor comprises a nucleic acid programmable DNA binding protein (napDNAbp) and a polypeptide having an RNA-dependent DNA polymerase activity, and each PEgRNA comprises a spacer sequence, a gRNA core, a DNA synthesis template, and a primer binding site. In certain embodiments, each DNA synthesis template encodes a single-stranded DNA sequence comprising an edited portion. Two single-stranded DNA sequences encoded may by complementary to one another and form a duplex that integrates into the target site to be edited. In some embodiments, the two single-stranded DNA sequences encoded may comprise a region of complementarity to one another. In certain embodiments, the two single-stranded DNA sequences encoded may comprise a region of complementarity to one another that is at least 2 bp, at least 3 bp, at least 4 bp, at least 5 bp, at least 10 bp, at least 20 bp, at least 30 bp, at least 40 bp, at least 50 bp, at least 100 bp, at least 200 bp, at least 300 bp, at least 400 bp, at least 500 bp, at least 600 bp, at least 700 bp, at least 800 bp, at least 900 bp, or at least 1000 bp long. In some embodiments, the prime editor is provided as a fusion protein. In certain embodiments, the components of the prime editor (i.e., the napDNAbp and the polypeptide having an RNA-dependent DNA polymerase activity) are provided in trans.


In some embodiments, each napDNAbp is a Cas9 domain or variant thereof. In some embodiments, each napDNAbp is a nuclease active Cas9 domain, a nuclease inactive Cas9 domain, or a Cas9 nickase domain or a variant thereof. In certain embodiments, each napDNAbp is independently selected from the group consisting of: Cas9, Cas12e, Cas12d, Cas12a, Cas12b1, Cas13a, Cas12c, and Argonaute and optionally has a nickase activity. In various embodiments, each napDNAbp comprises an amino acid sequence of any one of SEQ ID NOs: 2-65, or an amino acid sequence at least 80%, 85%, 90%, 95%, or 99% identical to any one of SEQ ID NOs: 2-65.


In some embodiments, the polypeptide comprising an RNA-dependent DNA polymerase activity is a reverse transcriptase. In certain embodiments, the polypeptide comprising an RNA-dependent DNA polymerase activity comprises an amino acid sequence of any one of SEQ ID NOs: 37, 68-79, 82-98, 81, 98, and 110 or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 37, 68-79, 82-98, 81, 98, and 110.


In some embodiments, each prime editor may comprise a linker that joins the napDNAbp and the reverse transcriptase. In certain embodiments, the linker comprises an amino acid sequence of any one of SEQ ID NOs: 119-128, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 119-128. Each linker may be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 38, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 amino acids in length.


In some embodiments, each PEgRNA may independently comprise a nucleotide sequence of any one of SEQ ID NOs: 192-203, or a nucleotide sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 192-203.


In various embodiments, each spacer sequence of each PEgRNA may bind to a specific binding site of the double-stranded DNA sequence adjacent the target site to be edited. In some embodiments, the binding of a spacer sequence of one prime editor complex to one strand of the double-stranded DNA sequence and the binding of a spacer sequence of another prime editor complex to the opposite strand of the double-stranded DNA sequence results in the nicking of both DNA strands at a nick site proximal to the PAM sequences on each strand.


In another aspect, the present disclosure provides polynucleotides. In some embodiments, the polynucleotides may encode any of the complexes described herein. In certain embodiments, the polynucleotides may encode any of the PEgRNAs described herein.


In yet another aspect, the specification provides polynucleotides. In certain embodiments, the polynucleotides may encode any of the prime editors disclosed herein. In certain other embodiments, the polynucleotides may encode any of the napDNAbps disclosed herein. In still further embodiments, the polynucleotides may encode any of the reverse transcriptases disclosed herein. In yet other embodiments, the polynucleotides may encode any of the extended guide RNAs disclosed herein, any of the reverse transcription template sequences, or any of the reverse transcription primer sites, or any of the optional linker sequences.


In still other aspects, the specification provides vectors comprising the polynucleotides described herein. Thus, in certain embodiments, the vectors comprise polynucleotides for encoding the prime editors comprising a napDNAbp and a reverse transcriptase (i.e., as fusion protein, or expressed in trans). In certain embodiments, the vectors comprise polynucleotides for encoding any of the complexes described herein. In other embodiments, the vectors comprise polynucleotides that separately encode a napDNAbp and reverse transcriptase. In still other embodiments, the vectors may comprise polynucleotides that encode the extended guide RNAs. In various embodiments, the vectors may comprise one or more polynucleotides that encode napDNAbps, reverse transcriptase, and extended guide RNAs on the same or separate vectors. In some embodiments, the vectors comprise polynucleotides for encoding any of the pegRNAs described herein.


In still other aspects, the specification provides cells comprising a prime editor as described herein and an extended guide RNA. The cells may be transformed with the vectors comprising the prime editors, napDNAbps, reverse transcriptase, and extended guide RNAs. These genetic elements may be comprised on the same vector or on different vectors. In some embodiments, the cells comprise any of the systems or complexes described herein. The cells may be transformed with polynucleotides encoding the any of the systems, complexes, and/or pegRNAs disclosed herein, or vectors comprising polynucleotides encoding the any of the systems, complexes, or pegRNAs disclosed herein.


In yet another aspect, the specification provides pharmaceutical compositions. In certain embodiments, the pharmaceutical compositions comprise one or more of a napDNAbp, a prime editor, a reverse transcriptase, and an extended guide RNA. In certain embodiments, the pharmaceutical compositions comprise any of the systems and/or complexes described herein. In certain embodiments, the pharmaceutical compositions comprise any of the prime editors, systems, or complexes described herein and a pharmaceutically acceptable excipient. In other embodiments, the pharmaceutical compositions comprise any extend guide RNA described herein and a pharmaceutically acceptable excipient. In still other embodiments, the pharmaceutical compositions comprise any extend guide RNA described herein in combination with any prime editor described herein and a pharmaceutically acceptable excipient. In yet other embodiments, the pharmaceutical compositions comprise any polynucleotide sequence encoding one or more of a napDNAbp, a prime editor, a reverse transcriptase, and an extended guide RNA, or any of the vectors disclosed herein. In still other embodiments, the various components disclosed herein may be separated into one or more pharmaceutical compositions. For example, a first pharmaceutical composition may comprise a prime editor or a napDNAbp, a second pharmaceutical compositions may comprise a reverse transcriptase, and a third pharmaceutical composition may comprise an extended guide RNA.


In still a further aspect, the present disclosure provides kits. In one embodiment, the kit comprises one or more polynucleotides encoding one or more components, including a prime editor, a napDNAbp, a reverse transcriptase, and an extended guide RNA. The kits may also comprise vectors, cells, and isolated preparations of polypeptides, including any prime editor, napDNAbp, or reverse transcriptase disclosed herein.


In yet another aspect, the present disclosure provides for methods of using the disclosed compositions of matter, including methods of using any of the systems described herein for simultaneously editing both complementary strands of a double-stranded DNA sequence at a target site. In some embodiments, the method comprises contacting the double-stranded DNA sequence with any of the system disclosed herein.


In one aspect, the disclosure provides methods comprising contacting a double-stranded DNA sequence at a target site with a first and a second prime editor complex, each complex comprising a prime editor and a prime editing guide RNA (PEgRNA). In some embodiments, each prime editor comprises a nucleic acid programmable DNA binding protein (napDNAbp) and a polypeptide having an RNA-dependent DNA polymerase activity, and each PEgRNA comprises a spacer sequence, a gRNA core, a DNA synthesis template, and a primer binding site. In some embodiments, each prime editor is provided as a fusion protein. In some embodiments, the components of the prime editor are provided in trans. In certain embodiments, each DNA synthesis template encodes a single-stranded DNA sequence comprising an edited portion. Two single-stranded DNA sequences encoded may be complementary to one another and form a duplex that integrates into the target site to be edited. The various elements of the prime editor complexes may comprise any of the embodiments of the systems disclosed herein.


In another aspect, the disclosure provides methods comprising contacting a double-stranded DNA sequence at a target site with a first, a second, a third, and a fourth prime editor complex, each complex comprising a prime editor and a prime editing guide RNA (PEgRNA). In some embodiments, each prime editor comprises a nucleic acid programmable DNA binding protein (napDNAbp) and a polypeptide having an RNA-dependent DNA polymerase activity, and each PEgRNA comprises a spacer sequence, a gRNA core, a DNA synthesis template, and a primer binding site. In some embodiments, each prime editor is provided as a fusion protein. In some embodiments, the components of the prime editor are provided in trans. In certain embodiments, each DNA synthesis template encodes a single-stranded DNA sequence. Two single-stranded DNA sequences encoded may be complementary to one another and form a duplex that integrates into the target site to be edited. The various elements of the prime editor complexes may comprise any of the embodiments of the systems disclosed herein.


In some embodiments, the methods provided herein allow for inversion of a target DNA sequence. In some embodiments, the first single-stranded DNA sequence encoded by the first DNA synthesis template and the second single-stranded DNA sequence encoded by the second DNA synthesis template are on opposite ends of a target DNA sequence, and the third single-stranded DNA sequence encoded by the third DNA synthesis template and the fourth single-stranded DNA sequence encoded by the fourth DNA synthesis template are on opposite ends of the same target DNA sequence.


In some embodiments, the methods provided herein further comprise providing a circular DNA donor. In certain embodiments, the first single-stranded DNA sequence encoded by the first DNA synthesis template and the third single-stranded DNA sequence encoded by the third DNA synthesis template are on opposite ends of the target DNA sequence, and the second single-stranded DNA sequence encoded by the second DNA synthesis template and the fourth single-stranded DNA sequence encoded by the fourth DNA synthesis template are on the circular DNA donor. In some embodiments, the portion of the circular DNA donor between the second single-stranded DNA sequence and the fourth single-stranded DNA sequence replaces the target DNA sequence between the first single-stranded DNA sequence and the third single-stranded DNA sequence.


In some embodiments, the methods provided herein allow for translocation of a target DNA sequence from a first nucleic acid molecule (e.g., a first chromosome) to a second nucleic acid molecule (e.g., a second chromosome). In some embodiments, the first single-stranded DNA sequence encoded by the first DNA synthesis template and the third single-stranded DNA sequence encoded by the third single-stranded DNA synthesis template are on a first nucleic acid molecule, and the second single-stranded DNA sequence encoded by the second DNA synthesis template and the fourth single-stranded DNA sequence encoded by the fourth DNA synthesis template are on a second nucleic acid molecule. In certain embodiments, a portion of the first nucleic acid molecule between the first single-stranded DNA sequence and the third single-stranded DNA sequence is incorporated into the second nucleic acid molecule. In certain embodiments, a portion of the second nucleic acid molecule between the second single-stranded DNA sequence and the fourth single-stranded DNA sequence is incorporated into the first nucleic acid molecule.


In another aspect, the present disclosure provides a pair of PEgRNAs for use in multi-flap prime editing. In some embodiments, the pair comprises a first PEgRNA and a second PEgRNA, and each PEgRNA independently comprises a spacer sequence, a gRNA core, a DNA synthesis template, and a primer binding site. In certain embodiments, each DNA synthesis template encodes a single-stranded DNA sequence. In various embodiments, the multi-flap prime editors are used in connection with a pair of PEgRNAs which target separate prime editors to either side of a target site, wherein the pair of PEgRNA each encode 3′ nucleic acid flaps which comprise nucleic acid sequences which are reverse complements of each other. In various embodiments, the 3′ flaps comprising the reverse complement sequences may anneal to one another to form a duplex comprising the desired edit or nucleic acid sequence encoding by the PEgRNAs. The duplex then becomes integrated into the target site by replacement of the corresponding endogenous duplex positioned between adjacent nick sites.


In another aspect, the present disclosure provides a plurality of PEgRNAs for use in multi-flap prime editing. In some embodiments, the plurality comprises a first, a second, a third, and a fourth PEgRNA. In some embodiments, each of the four PEgRNA independently comprises a spacer sequence, a gRNA core, a DNA synthesis template, and a primer binding site. In certain embodiments, each DNA synthesis template encodes a single-stranded DNA sequence. Two single-stranded DNA sequences encoded may be complementary to one another.


In various aspects, the present disclosure provides polynucleotides encoding any of the pairs or pluralities of PEgRNAs described herein. In certain aspects, the present disclosure provides vectors encoding a polynucleotide encoding any of the pairs or pluralities of PEgRNAs described herein. In yet another aspect, the present disclosure provides cells comprising a vector encoding a polynucleotide encoding any of the pairs or pluralities of PEgRNAs described herein. In other aspects, the disclosure provides pharmaceutical compositions comprising any of the pairs or pluralities of PEgRNAs described herein, a vector encoding any of the pairs or pluralities of PEgRNAs described herein, or a cell comprising a vector encoding any of the pairs or pluralities of PEgRNAs described herein. In certain embodiments, the pharmaceutical compositions comprise a pharmaceutical excipient.


In one embodiment, the methods relate to a method for installing a desired nucleotide change in a double-stranded DNA sequence. The method first comprises contacting the double-stranded DNA sequence with a complex comprising a prime editor and an extended guide RNA, wherein the prime editor comprises a napDNAbp and a reverse transcriptase and wherein the extended guide RNA comprises a reverse transcription template sequence comprising the desired nucleotide change. In some embodiments, each prime editor is provided as a fusion protein. In some embodiments, the components of the prime editor are provided in trans. Next, the method involves nicking the double-stranded DNA sequence on the non-target strand, thereby generating a free single-strand DNA having a 3′ end. The method then involves hybridizing the 3′ end of the free single-strand DNA to the reverse transcription template sequence, thereby priming the reverse transcriptase domain. The method then involves polymerizing a strand of DNA from the 3′ end, thereby generating a single-strand DNA flap comprising the desired nucleotide change. Then, the method involves replacing an endogenous DNA strand adjacent the cut site with the single-strand DNA flap, thereby installing the desired nucleotide change in the double-stranded DNA sequence.


In other embodiments, the disclosure provides for a method for introducing one or more changes in the nucleotide sequence of a DNA molecule at a target locus, comprising contacting the DNA molecule with a nucleic acid programmable DNA binding protein (napDNAbp) and a guide RNA which targets the napDNAbp to the target locus, wherein the guide RNA comprises a reverse transcriptase (RT) template sequence comprising at least one desired nucleotide change. Next, the method involves forming an exposed 3′ end in a DNA strand at the target locus and then hybridizing the exposed 3′ end to the RT template sequence to prime reverse transcription. Next, a single strand DNA flap comprising the at least one desired nucleotide change based on the RT template sequence is synthesized or polymerized by reverse transcriptase. Lastly, the at least one desired nucleotide change is incorporated into the corresponding endogenous DNA, thereby introducing one or more changes in the nucleotide sequence of the DNA molecule at the target locus.


In still other embodiments, the disclosure provides a method for introducing one or more changes in the nucleotide sequence of a DNA molecule at a target locus by target-primed reverse transcription, the method comprising: (a) contacting the DNA molecule at the target locus with a (i) prime editor comprising a nucleic acid programmable DNA binding protein (napDNAbp) and a reverse transcriptase and (ii) a guide RNA comprising an RT template comprising a desired nucleotide change; (b) conducting target-primed reverse transcription of the RT template to generate a single strand DNA comprising the desired nucleotide change; and (c) incorporating the desired nucleotide change into the DNA molecule at the target locus through a DNA repair and/or replication process.


In certain embodiments, the step of replacing the endogenous DNA strand comprises: (i) hybridizing the single-strand DNA flap to the endogenous DNA strand adjacent the cut site to create a sequence mismatch; (ii) excising the endogenous DNA strand; and (iii) repairing the mismatch to form the desired product comprising the desired nucleotide change in both strands of DNA.


In various embodiments, the desired nucleotide change can be a single nucleotide substitution (e.g., and transition or a transversion change), a deletion, or an insertion. For example, the desired nucleotide change can be (1) a G to T substitution, (2) a G to A substitution, (3) a G to C substitution, (4) a T to G substitution, (5) a T to A substitution, (6) a T to C substitution, (7) a C to G substitution, (8) a C to T substitution, (9) a C to A substitution, (10) an A to T substitution, (11) an A to G substitution, or (12) an A to C substitution.


In other embodiments, the desired nucleoid change can convert (1) a G:C basepair to a T:A basepair, (2) a G:C basepair to an A:T basepair, (3) a G:C basepair to C:G basepair, (4) a T:A basepair to a G:C basepair, (5) a T:A basepair to an A:T basepair, (6) a T:A basepair to a C:G basepair, (7) a C:G basepair to a G:C basepair, (8) a C:G basepair to a T:A basepair, (9) a C:G basepair to an A:T basepair, (10) an A:T basepair to a T:A basepair, (11) an A:T basepair to a G:C basepair, or (12) an A:T basepair to a C:G basepair.


In still other embodiments, the method introduces a desired nucleotide change that is an insertion. In certain cases, the insertion is at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 200, at least 300, at least 400, or at least 500 nucleotides in length.


In other embodiments, the method introduces a desired nucleotide change that is a deletion. In certain other cases, the deletion is at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 200, at least 300, at least 400, or at least 500 nucleotides in length.


In various embodiments, the desired nucleotide change corrects a disease-associated gene. The disease-associated gene can be associated with a monogenetic disorder selected from the group consisting of: Adenosine Deaminase (ADA) Deficiency; Alpha-1 Antitrypsin Deficiency; Cystic Fibrosis; Duchenne Muscular Dystrophy; Galactosemia; Hemochromatosis; Huntington's Disease; Maple Syrup Urine Disease; Marfan Syndrome; Neurofibromatosis Type 1; Pachyonychia Congenita; Phenylkeotnuria; Severe Combined Immunodeficiency; Sickle Cell Disease; Smith-Lemli-Opitz Syndrome; and Tay-Sachs Disease. In other embodiments, the disease-associated gene can be associated with a polygenic disorder selected from the group consisting of: heart disease; high blood pressure; Alzheimer's disease; arthritis; diabetes; cancer; and obesity.


The methods disclosed herein may involve fusion proteins having a napDNAbp that is a nuclease dead Cas9 (dCas9), a Cas9 nickase (nCas9), or a nuclease active Cas9. In other embodiments, a napDNAbp and reverse transcriptase are not encoded as a single fusion protein, but rather can be provided in separate constructs. Thus, in some embodiments, the reverse transcriptase can be provided in trans relative to the napDNAbp (rather than by way of a fusion protein).


In various embodiments involving methods, the napDNAbp may comprise an amino acid sequence of SEQ ID NOs: 26-61, 75-76, 126, 130, 137, 141, 147, 153, 157, 445, 460, 467, and 482-487 (Cas9); (SpCas9); SEQ ID NO: 77-86 (CP-Cas9); SEQ ID NO: 18-25 and 87-88 (SpCas9); and SEQ ID NOs: 62-72(Cas12). The napDNAbp may also comprise an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs: 26-61, 75-76, 126, 130, 137, 141, 147, 153, 157, 445, 460, 467, and 482-487 (Cas9); (SpCas9); SEQ ID NO: 77-86 (CP-Cas9); SEQ ID NO: 18-25 and 87-88 (SpCas9); and SEQ ID NOs: 62-72(Cas12).


In various embodiments involving methods, the reverse transcriptase may comprise any one of the amino acid sequences of SEQ ID NOs: 89-100, 105-122, 128-129, 132, 139, 143, 149, 154, 159, 235, 454, 471, 516, 662, 700-716, 739-742, and 766. The reverse transcriptase may also comprise an amino acid sequence that is at least 80%, 85%, 90%, 95%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs: 89-100, 105-122, 128-129, 132, 139, 143, 149, 154, 159, 235, 454, 471, 516, 662, 700-716, 739-742, and 766.


The methods may involve the use of a PEgRNA comprising a nucleotide sequence of SEQ ID NOs: 101-104, 181-183, 223-234, 237-244, 277, 324-330, 332, 334, 336, 338, 340, 342, 344, 346, 348, 350, 352, 354, 356, 358, 360, 362, 364, 366, 368, 394, 429-442, 499-505, 641-649, 678-692, 735-736, 757-761, 776-777, 2997-3103, 3113-3121, 3305-3455, 3479-3493, 3522-3540, 3549-3556, 3628-3698, 3755-3810, 3874, 3890-3901, 3905-3911, 3913-3929, and 3972-3989, or a nucleotide sequence having at least 80%, or at least 85%, or at least 90%, or at least 95%, or at least 99% sequence identity thereto The methods may comprise the use of extended guide RNAs that comprise an RNA extension at the 3′ end, wherein the RNA extension comprises the reverse transcription template sequence.


The methods may comprise the use of extended guide RNAs that comprise an RNA extension at the 5′ end, wherein the RNA extension comprises the reverse transcription template sequence.


The methods may comprise the use of extended guide RNAs that comprise an RNA extension at an intramolecular location in the guide RNA, wherein the RNA extension comprises the reverse transcription template sequence.


The methods may comprise the use of extended guide RNAs having one or more RNA extensions that are at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 200, at least 300, at least 400, or at least 500 nucleotides in length.


It should be appreciated that the foregoing concepts, and additional concepts discussed below, may be arranged in any suitable combination, as the present disclosure is not limited in this respect. Further, other advantages and novel features of the present disclosure will become apparent from the following detailed description of various non-limiting embodiments when considered in conjunction with the accompanying figures.





BRIEF DESCRIPTION OF THE DRAWINGS

The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present disclosure, which can be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.



FIG. 1A provides a schematic of an exemplary process for introducing a single nucleotide change, and/or insertion, and/or deletion into a DNA molecule (e.g., a genome) using a fusion protein comprising a reverse transcriptase fused to a Cas9 protein in complex with an extended guide RNA molecule. In this embodiment, the guide RNA is extended at the 3′ end to include a reverse transcriptase template sequence. The schematic shows how a reverse transcriptase (RT) fused to a Cas9 nickase, in a complex with a guide RNA (gRNA), binds the DNA target site and nicks the PAM-containing DNA strand adjacent to the target nucleotide. The RT enzyme uses the nicked DNA as a primer for DNA synthesis from the gRNA, which is used as a template for the synthesis of a new DNA strand that encodes the desired edit. The editing process shown may be referred to as target-primed reverse transcription editing (TRT editing) or equivalently, “prime editing.”



FIG. 1B provides the same representation as in FIG. 1A, except that the prime editor complex is represented more generally as [napDNAbp]-[P]:PEgRNA or [P]-[napDNAbp]:PEgRNA, wherein “P” refers to any polymerase (e.g., a reverse transcriptase), “napDNAbp” refers to a nucleic acid programmable DNA binding protein (e.g., SpCas9), and “PEgRNA” refers to a prime editing guide RNA, and “]-[” refers to an optional linker. As described elsewhere, e.g., FIGS. 3A-3G, the PEgRNA comprises an 5′ extension arm comprising a primer binding site and a DNA synthesis template. Although not shown, it is contemplated that the extension arm of the PEgRNA (i.e., which comprises a primer binding site and a DNA synthesis template) can be DNA or RNA. The particular polymerase contemplated in this configuration will depend upon the nature of the DNA synthesis template. For instance, if the DNA synthesis template is RNA, then the polymerase case be an RNA-dependent DNA polymerase (e.g., reverse transcriptase). If the DNA synthesis template is DNA, then the polymerase can be a DNA-dependent DNA polymerase.



FIG. 1C provides a schematic of an exemplary process for introducing a single nucleotide change, and/or insertion, and/or deletion into a DNA molecule (e.g., a genome) using a fusion protein comprising a reverse transcriptase fused to a Cas9 protein in complex with an extended guide RNA molecule. In this embodiment, the guide RNA is extended at the 5′ end to include a reverse transcriptase template sequence. The schematic shows how a reverse transcriptase (RT) fused to a Cas9 nickase, in a complex with a guide RNA (gRNA), binds the DNA target site and nicks the PAM-containing DNA strand adjacent to the target nucleotide. The RT enzyme uses the nicked DNA as a primer for DNA synthesis from the gRNA, which is used as a template for the synthesis of a new DNA strand that encodes the desired edit. The editing process shown may be referred to as target-primed reverse transcription editing (TRT editing) or equivalently, “prime editing.”



FIG. 1D provides the same representation as in FIG. 1C, except that the prime editor complex is represented more generally as [napDNAbp]-[P]:PEgRNA or [P]-[napDNAbp]:PEgRNA, wherein “P” refers to any polymerase (e.g., a reverse transcriptase), “napDNAbp” refers to a nucleic acid programmable DNA binding protein (e.g., SpCas9), and “PEgRNA” refers to a prime editing guide RNA, and “]-[” refers to an optional linker. As described elsewhere, e.g., FIGS. 3A-3G, the PEgRNA comprises an 3′ extension arm comprising a primer binding site and a DNA synthesis template. Although not shown, it is contemplated that the extension arm of the PEgRNA (i.e., which comprises a primer binding site and a DNA synthesis template) can be DNA or RNA. The particular polymerase contemplated in this configuration will depend upon the nature of the DNA synthesis template. For instance, if the DNA synthesis template is RNA, then the polymerase case be an RNA-dependent DNA polymerase (e.g., reverse transcriptase). If the DNA synthesis template is DNA, then the polymerase can be a DNA-dependent DNA polymerase. In various embodiments, the PEgRNA can be engineered or synthesized to incorporate a DNA-based DNA synthesis template.



FIG. 1E is a schematic depicting an exemplary process of how the synthesized single strand of DNA (which comprises the desired nucleotide change) becomes resolved such that the desired nucleotide change is incorporated into the DNA. As shown, following synthesis of the edited strand (or “mutagenic strand”), equilibration with the endogenous strand, flap cleavage of the endogenous strand, and ligation leads to incorporation of the DNA edit after resolution of the mismatched DNA duplex through the action of endogenous DNA repair and/or replication processes.



FIG. 1F is a schematic showing that “opposite strand nicking” can be incorporated into the resolution method of FIG. 1E to help drive the formation of the desired product versus the reversion product. In opposite strand nicking, a second Cas9/gRNA complex is used to introduce a second nick on the opposite strand from the initial nicked strand. This induces the endogenous cellular DNA repair and/or replication processes to preferentially replace the unedited strand (i.e., the strand containing the second nick site).



FIG. 1G provides another schematic of an exemplary process for introducing a single nucleotide change, and/or insertion, and/or deletion into a DNA molecule (e.g., a genome) of a target locus using a nucleic acid programmable DNA binding protein (napDNAbp) complexed with an extended guide RNA. This process may be referred to as an embodiment of prime editing. The extended guide RNA comprises an extension at the 3′ or 5′ end of the guide RNA, or at an intramolecular location in the guide RNA. In step (a), the napDNAbp/gRNA complex contacts the DNA molecule and the gRNA guides the napDNAbp to bind to the target locus. In step (b), a nick in one of the strands of DNA (the R-loop strand, or the PAM-containing strand, or the non-target DNA strand, or the protospacer strand) of the target locus is introduced (e.g., by a nuclease or chemical agent), thereby creating an available 3′ end in one of the strands of the target locus. In certain embodiments, the nick is created in the strand of DNA that corresponds to the R-loop strand, i.e., the strand that is not hybridized to the guide RNA sequence. In step (c), the 3′ end DNA strand interacts with the extended portion of the guide RNA in order to prime reverse transcription. In certain embodiments, the 3′ ended DNA strand hybridizes to a specific RT priming sequence on the extended portion of the guide RNA. In step (d), a reverse transcriptase is introduced which synthesizes a single strand of DNA from the 3′ end of the primed site towards the 3′ end of the guide RNA. This forms a single-strand DNA flap comprising the desired nucleotide change (e.g., the single base change, insertion, or deletion, or a combination thereof). In step (e), the napDNAbp and guide RNA are released. Steps (f) and (g) relate to the resolution of the single strand DNA flap such that the desired nucleotide change becomes incorporated into the target locus. This process can be driven towards the desired product formation by removing the corresponding 5′ endogenous DNA flap that forms once the 3′ single strand DNA flap invades and hybridizes to the complementary sequence on the other strand. The process can also be driven towards product formation with second strand nicking, as exemplified in FIG. 1F. This process may introduce at least one or more of the following genetic changes: transversions, transitions, deletions, and insertions.



FIG. 1H is a schematic depicting the types of genetic changes that are possible with the prime editing processes described herein. The types of nucleotide changes achievable by prime editing include deletions (including short and long deletions), single-nucleotide changes (including transitions and transversions), inversions, and insertions (including short and long deletions).



FIG. 1I is a schematic depicting temporal second strand nicking exemplified by PE3b (PE3b=PE2 prime editor fusion protein+PEgRNA+second strand nicking guide RNA). Temporal second strand nicking is a variant of second strand nicking in order to facilitate the formation of the desired edited product. The “temporal” term refers to the fact that the second-strand nick to the unedited strand occurs only after the desired edit is installed in the edited strand. This avoids concurrent nicks on both strands to lead to double-stranded DNA breaks.



FIGS. 1J-1K depict a variation of prime editing contemplated herein that replaces the napDNAbp (e.g., SpCas9 nickase) with any programmable nuclease domain, such as zinc finger nucleases (ZFN) or transcription activator-like effector nucleases (TALEN). As such, it is contemplated that suitable nucleases do not necessarily need to be “programmed” by a nucleic acid targeting molecule (such as a guide RNA), but rather, may be programmed by defining the specificity of a DNA-binding domain, such as and in particular, a nuclease. Just as in prime editing with napDNAbp moieties, it is preferable that such alternative programmable nucleases be modified such that only one strand of a target DNA is cut. In other words, the programmable nucleases should function as nickases, preferably. Once a programmable nuclease is selected (e.g., a ZFN or a TALEN), then additional functionalities may be engineered into the system to allow it to operate in accordance with a prime editing-like mechanism. For example, the programmable nucleases may be modified by coupling (e.g., via a chemical linker) an RNA or DNA extension arm thereto, wherein the extension arm comprises a primer binding site (PBS) and a DNA synthesis template. The programmable nuclease may also be coupled (e.g., via a chemical or amino acid linker) to a polymerase, the nature of which will depend upon whether the extension arm is DNA or RNA. In the case of an RNA extension arm, the polymerase can be an RNA-dependent DNA polymerase (e.g., reverse transcriptase). In the case of a DNA extension arm, the polymerase can be a DNA-dependent DNA polymerase (e.g., a prokaryotic polymerase, including Pol I, Pol II, or Pol III, or a eukaryotic polymerase, including Pol a, Pol b, Pol g, Pol d, Pol e, or Pol z). The system may also include other functionalities added as fusions to the programmable nucleases, or added in trans to facilitate the reaction as a whole (e.g., (a) a helicase to unwind the DNA at the cut site to make the cut strand with the 3′ end available as a primer, (b) a flap endonuclease (e.g., FEN1) to help remove the endogenous strand on the cut strand to drive the reaction towards replacement of the endogenous strand with the synthesized strand, or (c) a nCas9:gRNA complex to create a second site nick on the opposite strand, which may help drive the integration of the synthesize repair through favored cellular repair of the non-edited strand). In an analogous manner to prime editing with a napDNAbp, such a complex with an otherwise programmable nuclease could be used to synthesize and then install a newly synthesized replacement strand of DNA carrying an edit of interest permanently into a target site of DNA.



FIG. 1L depicts, in one embodiment, the anatomical features of a target DNA that may be edited by prime editing. The target DNA comprises a “non-target strand” and a “target strand.” The target-strand is the strand that becomes annealed to the spacer of a PEgRNA of a prime editor complex that recognizes the PAM site (in this case, NGG, which is recognized by the canonical SpCas9-based prime editors) The target strand may also be referred to as the “non-PAM strand” or the “non-edit strand.” By contrast, the non-target strand (i.e., the strand containing the protospacer and the PAM sequence of NGG) may be referred to as the “PAM-strand” or the “edit strand.” In various embodiments, the nick site of the PE complex will be in the protospacer on the PAM-strand (e.g., with the SpCas9-based PE). The location of the nick will be characteristic of the particular Cas9 that forms the PE. For example, with an SpCas9-based PE, the nick site in the phosphodiester bond between bases three (“-3” position relative to the position 1 of the PAM sequence) and four (“-4” position relative to position 1 of the PAM sequence). The nick site in the protospacer forms a free 3′ hydroxyl group, which as seen in the following figures, complexes with the primer binding site of the extension arm of the PEgRNA and provides the substrate to begin polymerization of a single strand of DNA code for by the DNA synthesis template of the extension arm of the PEgRNA. This polymerization reaction is catalyzed by the polymerase (e.g., reverse transcriptase) of the PE fusion protein in the 5′ to 3′ direction. Polymerization terminates before reaching the gRNA core (e.g., by inclusion of a polymerization termination signal, or secondary structure, which functions to terminate the polymerization activity of PE), producing a single strand DNA flap that is extended from the original 3′ hydroxyl group of the nicked PAM strand. The DNA synthesis template codes for a single strand DNA that is homologous to the endogenous 5′-ended single strand of DNA that immediately follows the nick site on the PAM strand and incorporates the desired nucleotide change (e.g., single base substitution, insertion, deletion, inversion). The position of the desired edit can be in any position following downstream of the nick site on the PAM strand, which can include position +1, +2, +3, +4 (the start of the PAM site), +5 (position 2 of the PAM site), +6 (position 3 of the PAM site), +7, +8, +9, +10, +11, +12, +13, +14, +15, +16, +17, +18, +19, +20, +21, +22, +23, +24, +25, +26, +27, +28, +29, +30, +31, +32, +33, +34, +35, +36, +37, +38, +39, +40, +41, +42, +43, +44, +45, +46, +47, +48, +49, +50, +51, +52, +53, +54, +55, +56, +57, +58, +59, +60, +61, +62, +63, +64, +65, +66, +67, +68, +69, +70, +71, +72, +73, +74, +75, +76, +77, +78, +79, +80, +81, +82, +83, +84, +85, +86, +87, +88, +89, +90, +91, +92, +93, +94, +95, +96, +97, +98, +99, +100, +101, +102, +103, +104, +105, +106, +107, +108, +109, +110, +111, +112, +113, +114, +115, +116, +117, +118, +119, +120, +121, +122, +123, +124, +125, +126, +127, +128, +129, +130, +131, +132, +133, +134, +135, +136, +137, +138, +139, +140, +141, +142, +143, +144, +145, +146, +147, +148, +149, or +150, or more (relative to the downstream position of the nick site). Once the 3′ end single stranded DNA (containing the edit of interest) replaces the endogenous 5′ end single stranded DNA, the DNA repair and replication processes will result in permanent installation of the edit site on the PAM strand, and then correction of the mismatch on the non-PAM strand that will exist at the edit site. In this way, the edit will extend to both strands of DNA on the target DNA site. It will be appreciated that reference to “edited strand” and “non-edited” strand only intends to delineate the strands of DNA involved in the PE mechanism. The “edited strand” is the strand that first becomes edited by replacement of the 5′ ended single strand DNA immediately downstream of the nick site with the synthesized 3′ ended single stranded DNA containing the desired edit. The “non-edited” strand is the strand pair with the edited strand, but which itself also becomes edited through repair and/or replication to be complementary to the edited strand, and in particular, the edit of interest.



FIG. 1M depicts the mechanism of prime editing showing the anatomical features of the target DNA, prime editor complex, and the interaction between the PEgRNA and the target DNA. First, a prime editor comprising a fusion protein having a polymerase (e.g., reverse transcriptase) and a napDNAbp (e.g., SpCas9 nickase, e.g., a SpCas9 having a deactivating mutation in an HNH nuclease domain (e.g., H840A) or a deactivating mutation in a RuvC nuclease domain (D10A)) is complexed with a PEgRNA and DNA having a target DNA to be edited. The PEgRNA comprises a spacer, gRNA core (aka gRNA scaffold or gRNA backbone) (which binds to the napDNAbp), and an extension arm. The extension arm can be at the 3′ end, the 5′ end, or somewhere within the PEgRNA molecule. As shown, the extension arm is at the 3′ end of the PEgRNA. The extension arm comprises in the 3′ to 5′ direction a primer binding site and a DNA synthesis template (comprising both an edit of interest and regions of homology (i.e., homology arms) that are homologous with the 5′ ended single stranded DNA immediately following the nick site on the PAM strand. As shown, once the nick is introduced thereby producing a free 3′ hydroxyl group immediately upstream of the nick site, the region immediately upstream of the nick site on the PAM strand anneals to a complementary sequence at the 3′ end of the extension arm referred to as the “primer binding site,” creating a short double-stranded region with an available 3′ hydroxyl end, which forms a substrate for the polymerase of the prime editor complex. The polymerase (e.g., reverse transcriptase) then polymerase as strand of DNA from the 3′ hydroxyl end to the end of the extension arm. The sequence of the single stranded DNA is coded for by the DNA synthesis template, which is the portion of the extension arm (i.e., excluding the primer binding site) that is “read” by the polymerase to synthesize new DNA. This polymerization effectively extends the sequence of the original 3′ hydroxyl end of the initial nick site. The DNA synthesis template encodes a single strand of DNA that comprises not only the desired edit, but also regions that are homologous to the endogenous single strand of DNA immediately downstream of the nick site on the PAM strand. Next, the encoded 3′ ended single strand of DNA (i.e., the 3′ single strand DNA flap) displaces the corresponding homologous endogenous 5′-ended single strand of DNA immediately downstream of the nick site on the PAM strand, forming a DNA intermediate having a 5′-ended single strand DNA flap, which is removed by the cell (e.g., by a flap endonuclease). The 3′-ended single strand DNA flap, which anneals to the complement of the endogenous 5′-ended single strand DNA flap, is ligated to the endogenous strand after the 5′ DNA flap is removed. The desired edit in the 3′ ended single strand DNA flap, now annealed and ligate, forms a mismatch with the complement strand, which undergoes DNA repair and/or a round of replication, thereby permanently installing the desired edit on both strands.



FIG. 2 shows three Cas complexes (SpCas9, SaCas9, and LbCas12a) that can be used in the herein described prime editors and their PAM, gRNA, and DNA cleavage features. The figure shows designs for complexes involving SpCas9, SaCas9, and LbCas12a.



FIGS. 3A-3F show designs for engineered 5′ prime editor gRNA (FIG. 3A), 3′ prime editor gRNA (FIG. 3B), and an intramolecular extension (FIG. 3C). The extended guide RNA (or extended gRNA) may also be referred to herein as PEgRNA or “prime editing guide RNA.” FIG. 3D and FIG. 3E provide additional embodiments of 3′ and 5′ prime editor gRNAs (PEgRNAs), respectively. FIG. 3F illustrates the interaction between a 3′ end prime editor guide RNA with a target DNA sequence. The embodiments of FIGS. 3A-3C depict exemplary arrangements of the reverse transcription template sequence (i.e., or more broadly referred to as a DNA synthesis template, as indicated, since the RT is only one type of polymerase that may be used in the context of prime editors), the primer binding site, and an optional linker sequence in the extended portions of the 3′, 5′, and intramolecular versions, as well as the general arrangements of the spacer and core regions. The disclosed prime editing process is not limited to these configurations of extended guide RNAs. The embodiment of FIG. 3D provides the structure of an exemplary PEgRNA contemplated herein. The PEgRNA comprises three main component elements ordered in the 5′ to 3′ direction, namely: a spacer, a gRNA core, and an extension arm at the 3′ end. The extension arm may further be divided into the following structural elements in the 5′ to 3′ direction, namely: a primer binding site (A), an edit template (B), and a homology arm (C). In addition, the PEgRNA may comprise an optional 3′ end modifier region (e1) and an optional 5′ end modifier region (e2). Still further, the PEgRNA may comprise a transcriptional termination signal at the 3′ end of the PEgRNA (not depicted). These structural elements are further defined herein. The depiction of the structure of the PEgRNA is not meant to be limiting and embraces variations in the arrangement of the elements. For example, the optional sequence modifiers (e1) and (e2) could be positioned within or between any of the other regions shown, and not limited to being located at the 3′ and 5′ ends. The PEgRNA could comprise, in certain embodiments, secondary RNA structure, such as, but not limited to, hairpins, stem/loops, toe loops, RNA-binding protein recruitment domains (e.g., the MS2 aptamer which recruits and binds to the MS2cp protein). For instance, such secondary structures could be position within the spacer, the gRNA core, or the extension arm, and in particular, within the e1 and/or e2 modifier regions. In addition to secondary RNA structures, the PEgRNAs could comprise (e.g., within the e1 and/or e2 modifier regions) a chemical linker or a poly(N) linker or tail, where “N” can be any nucleobase. In some embodiments (e.g., as shown in FIG. 72(c)), the chemical linker may function to prevent reverse transcription of the sgRNA scaffold or core. In addition, in certain embodiments (e.g., see FIG. 72(c)), the extension arm (3) could be comprised of RNA or DNA, and/or could include one or more nucleobase analogs (e.g., which might add functionality, such as temperature resilience). Still further, the orientation of the extension arm (3) can be in the natural 5′-to-3′ direction, or synthesized in the opposite orientation in the 3′-to-5′ direction (relative to the orientation of the PEgRNA molecule overall). It is also noted that one of ordinary skill in the art will be able to select an appropriate DNA polymerase, depending on the nature of the nucleic acid materials of the extension arm (i.e., DNA or RNA), for use in prime editing that may be implemented either as a fusion with the napDNAbp or as provided in trans as a separate moiety to synthesize the desired template-encoded 3′ single-strand DNA flap that includes the desired edit. For example, if the extension arm is RNA, then the DNA polymerase could be a reverse transcriptase or any other suitable RNA-dependent DNA polymerase. However, if the extension arm is DNA, then the DNA polymerase could be a DNA-dependent DNA polymerase. In various embodiments, provision of the DNA polymerase could be in trans, e.g., through the use of an RNA-protein recruitment domain (e.g., an MS2 hairpin installed on the PEgRNA (e.g., in the e1 or e2 region, or elsewhere and an MS2cp protein fused to the DNA polymerase, thereby co-localizing the DNA polymerase to the PEgRNA). It is also noted that the primer binding site does not generally form a part of the template that is used by the DNA polymerase (e.g., reverse transcriptase) to encode the resulting 3′ single-strand DNA flap that includes the desired edit. Thus, the designation of the “DNA synthesis template” refers to the region or portion of the extension arm (3) that is used as a template by the DNA polymerase to encode the desired 3′ single-strand DNA flap containing the edit and regions of homology to the 5′ endogenous single strand DNA flap that is replaced by the 3′ single strand DNA strand product of prime editing DNA synthesis. In some embodiments, the DNA synthesis template includes the “edit template” and the “homology arm”, or one or more homology arms, e.g., before and after the edit template. The edit template can be as small as a single nucleotide substitution, or it may be an insertion, or an inversion of DNA. In addition, the edit template may also include a deletion, which can be engineered by encoding homology arm that contains a desired deletion. In other embodiments, the DNA synthesis template may also include the e2 region or a portion thereof. For instance, if the e2 region comprises a secondary structure that causes termination of DNA polymerase activity, then it is possible that DNA polymerase function will be terminated before any portion of the e2 region is actual encoded into DNA. It is also possible that some or even all of the e2 region will be encoded into DNA. How much of e2 is actually used as a template will depend on its constitution and whether that constitution interrupts DNA polymerase function.


The embodiment of FIG. 3E provides the structure of another PEgRNA contemplated herein. The PEgRNA comprises three main component elements ordered in the 5′ to 3′ direction, namely: a spacer, a gRNA core, and an extension arm at the 3′ end. The extension arm may further be divided into the following structural elements in the 5′ to 3′ direction, namely: a primer binding site (A), an edit template (B), and a homology arm (C). In addition, the PEgRNA may comprise an optional 3′ end modifier region (e1) and an optional 5′ end modifier region (e2). Still further, the PEgRNA may comprise a transcriptional termination signal on the 3′ end of the PEgRNA (not depicted). These structural elements are further defined herein. The depiction of the structure of the PEgRNA is not meant to be limiting and embraces variations in the arrangement of the elements. For example, the optional sequence modifiers (e1) and (e2) could be positioned within or between any of the other regions shown, and not limited to being located at the 3′ and 5′ ends. The PEgRNA could comprise, in certain embodiments, secondary RNA structures, such as, but not limited to, hairpins, stem/loops, toe loops, RNA-binding protein recruitment domains (e.g., the MS2 aptamer which recruits and binds to the MS2cp protein). These secondary structures could be positioned anywhere in the PEgRNA molecule. For instance, such secondary structures could be position within the spacer, the gRNA core, or the extension arm, and in particular, within the e1 and/or e2 modifier regions. In addition to secondary RNA structures, the PEgRNAs could comprise (e.g., within the e1 and/or e2 modifier regions) a chemical linker or a poly(N) linker or tail, where “N” can be any nucleobase. In some embodiments (e.g., as shown in FIG. 72(c)), the chemical linker may function to prevent reverse transcription of the sgRNA scaffold or core. In addition, in certain embodiments (e.g., see FIG. 72(c)), the extension arm (3) could be comprised of RNA or DNA, and/or could include one or more nucleobase analogs (e.g., which might add functionality, such as temperature resilience). Still further, the orientation of the extension arm (3) can be in the natural 5′-to-3′ direction, or synthesized in the opposite orientation in the 3′-to-5′ direction (relative to the orientation of the PEgRNA molecule overall). It is also noted that one of ordinary skill in the art will be able to select an appropriate DNA polymerase, depending on the nature of the nucleic acid materials of the extension arm (i.e., DNA or RNA), for use in prime editing that may be implemented either as a fusion with the napDNAbp or as provided in trans as a separate moiety to synthesize the desired template-encoded 3′ single-strand DNA flap that includes the desired edit. For example, if the extension arm is RNA, then the DNA polymerase could be a reverse transcriptase or any other suitable RNA-dependent DNA polymerase. However, if the extension arm is DNA, then the DNA polymerase could be a DNA-dependent DNA polymerase. In various embodiments, provision of the DNA polymerase could be in trans, e.g., through the use of an RNA-protein recruitment domain (e.g., an MS2 hairpin installed on the PEgRNA (e.g., in the e1 or e2 region, or elsewhere and an MS2cp protein fused to the DNA polymerase, thereby co-localizing the DNA polymerase to the PEgRNA). It is also noted that the primer binding site does not generally form a part of the template that is used by the DNA polymerase (e.g., reverse transcriptase) to encode the resulting 3′ single-strand DNA flap that includes the desired edit. Thus, the designation of the “DNA synthesis template” refers to the region or portion of the extension arm (3) that is used as a template by the DNA polymerase to encode the desired 3′ single-strand DNA flap containing the edit and regions of homology to the 5′ endogenous single strand DNA flap that is replaced by the 3′ single strand DNA strand product of prime editing DNA synthesis. In some embodiments, the DNA synthesis template includes the “edit template” and the “homology arm”, or one or more homology arms, e.g., before and after the edit template. The edit template can be as small as a single nucleotide substitution, or it may be an insertion, or an inversion of DNA. In addition, the edit template may also include a deletion, which can be engineered by encoding homology arm that contains a desired deletion. In other embodiments, the DNA synthesis template may also include the e2 region or a portion thereof. For instance, if the e2 region comprises a secondary structure that causes termination of DNA polymerase activity, then it is possible that DNA polymerase function will be terminated before any portion of the e2 region is actual encoded into DNA. It is also possible that some or even all of the e2 region will be encoded into DNA. How much of e2 is actually used as a template will depend on its constitution and whether that constitution interrupts DNA polymerase function.


The schematic of FIG. 3F depicts the interaction of a typical PEgRNA with a target site of a double stranded DNA and the concomitant production of a 3′ single stranded DNA flap containing the genetic change of interest. The double strand DNA is shown with the top strand (i.e., the target strand) in the 3′ to 5′ orientation and the lower strand (i.e., the PAM strand or non-target strand) in the 5′ to 3′ direction. The top strand comprises the complement of the “protospacer” and the complement of the PAM sequence and is referred to as the “target strand” because it is the strand that is target by and anneals to the spacer of the PEgRNA. The complementary lower strand is referred to as the “non-target strand” or the “PAM strand” or the “protospacer strand” since it contains the PAM sequence (e.g., NGG) and the protospacer. Although not shown, the PEgRNA depicted would be complexed with a Cas9 or equivalent domain of a prime editor fusion protein. As shown in the schematic, the spacer of the PEgRNA anneals to the complementary region of the protospacer on the target strand. This interaction forms as DNA/RNA hybrid between the spacer RNA and the complement of the protospacer DNA, and induces the formation of an R loop in the protospacer. As taught elsewhere herein, the Cas9 protein (not shown) then induces a nick in the non-target strand, as shown. This then leads to the formation of the 3′ ssDNA flap region immediately upstream of the nick site which, in accordance with *z*, interacts with the 3′ end of the PEgRNA at the primer binding site. The 3′ end of the ssDNA flap (i.e., the reverse transcriptase primer sequence) anneals to the primer binding site (A) on the PEgRNA, thereby priming reverse transcriptase. Next, reverse transcriptase (e.g., provided in trans or provided cis as a fusion protein, attached to the Cas9 construct) then polymerizes a single strand of DNA which is coded for by the DNA synthesis template (including the edit template (B) and homology arm (C)). The polymerization continues towards the 5′ end of the extension arm. The polymerized strand of ssDNA forms a ssDNA 3′ end flap which, as describe elsewhere (e.g., as shown in FIG. 1G), invades the endogenous DNA, displacing the corresponding endogenous strand (which is removed as a 5′ ended DNA flap of endogenous DNA), and installing the desired nucleotide edit (single nucleotide base pair change, deletions, insertions (including whole genes) through naturally occurring DNA repair/replication rounds.



FIG. 3G depicts yet another embodiment of prime editing contemplated herein. In particular, the top schematic depicts one embodiment of a prime editor (PE), which comprises a fusion protein of a napDNAbp (e.g., SpCas9) and a polymerase (e.g., a reverse transcriptase), which are joined by a linker. The PE forms a complex with a PEgRNA by binding to the gRNA core of the PEgRNA. In the embodiment shown, the PEgRNA is equipped with a 3′ extension arm that comprises, beginning at the 3′ end, a primer binding site (PBS) followed by a DNA synthesis template. The bottom schematic depicts a variant of a prime editor, referred to as a “trans prime editor (tPE).” In this embodiment, the DNA synthesis template and PBS are decoupled from the PEgRNA and presented on a separate molecule, referred to as a trans prime editor RNA template (“tPERT”), which comprises an RNA-protein recruitment domain (e.g., a MS2 hairpin). The PE itself is further modified to comprise a fusion to a rPERT recruiting protein (“RP”), which is a protein which specifically recognizes and binds to the RNA-protein recruitment domain. In the example where the RNA-protein recruitment domain is an MS2 hairpin, the corresponding rPERT recruiting protein can be MS2cp of the MS2 tagging system. The MS2 tagging system is based on the natural interaction of the MS2 bacteriophage coat protein (“MCP” or “MS2cp”) with a stem-loop or hairpin structure present in the genome of the phage, i.e., the “MS2 hairpin” or “MS2 aptamer.” In the case of trans prime editing, the RP-PE:gRNA complex “recruits” a tPERT having the appropriate RNA-protein recruitment domain to co-localize with the PE:gRNA complex, thereby providing the PBS and DNA synthesis template in trans for use in prime editing, as shown in the example depicted in FIG. 3H.



FIG. 3H depicts the process of trans prime editing. In this embodiment, the trans prime editor comprises a “PE2” prime editor (i.e., a fusion of a Cas9(H840A) and a variant MMLV RT) fused to an MS2cp protein (i.e., a type of recruiting protein that recognizes and binds to an MS2 aptamer) and which is complexed with an sgRNA (i.e., a standard guide RNA as opposed to a PEgRNA). The trans prime editor binds to the target DNA and nicks the nontarget strand. The MS2cp protein recruits a tPERT in trans through the specific interaction with the RNA-protein recruitment domain on the tPERT molecule. The tPERT becomes co-localized with the trans prime editor, thereby providing the PBS and DNA synthesis template functions in trans for use by the reverse transcriptase polymerase to synthesize a single strand DNA flap having a 3′ end and containing the desired genetic information encoded by the DNA synthesis template.



FIGS. 4A-4E demonstrate in vitro TPRT assays (i.e., prime editing assays). FIG. 4A is a schematic of fluorescently labeled DNA substrates gRNA templated extension by an RT enzyme, PAGE. FIG. 4B shows TPRT (i.e., prime editing) with pre-nicked substrates, dCas9, and 5′-extended gRNAs of differing synthesis template length. FIG. 4C shows the RT reaction with pre-nicked DNA substrates in the absence of Cas9. FIG. 4D shows TPRT (i.e., prime editing) on full dsDNA substrates with Cas9(H840A) and 5′-extended gRNAs. FIG. 4E shows a 3′-extended gRNA template with pre-nicked and full dsDNA substrates. All reactions are with M-MLV RT.



FIG. 5 shows in vitro validations using 5′-extended gRNAs with varying length synthesis templates. Fluorescently labeled (Cy5) DNA targets were used as substrates, and were pre-nicked in this set of experiments. The Cas9 used in these experiments is catalytically dead Cas9 (dCas9), and the RT used is Superscript III, a commercial RT derived from the Moloney-Murine Leukemia Virus (M-MLV). dCas9:gRNA complexes were formed from purified components. Then, the fluorescently labeled DNA substrate was added along with dNTPs and the RT enzyme. After 1 hour of incubation at 37° C., the reaction products were analyzed by denaturing urea-polyacrylamide gel electrophoresis (PAGE). The gel image shows extension of the original DNA strand to lengths that are consistent with the length of the reverse transcription template.



FIG. 6 shows in vitro validations using 5′-extended gRNAs with varying length synthesis templates, which closely parallels those shown in FIG. 5. However, the DNA substrates are not pre-nicked in this set of experiments. The Cas9 used in these experiments is a Cas9 nickase (SpyCas9 H840A mutant) and the RT used is Superscript III, a commercial RT derived from the Moloney-Murine Leukemia Virus (M-MLV). The reaction products were analyzed by denaturing urea-polyacrylamide gel electrophoresis (PAGE). As shown in the gel, the nickase efficiently cleaves the DNA strand when the standard gRNA is used (gRNA_0, lane 3).



FIG. 7 demonstrates that 3′ extensions support DNA synthesis and do not significantly effect Cas9 nickase activity. Pre-nicked substrates (black arrow) are near-quantitatively converted to RT products when either dCas9 or Cas9 nickase is used (lanes 4 and 5). Greater than 50% conversion to the RT product (red arrow) is observed with full substrates (lane 3). Cas9 nickase (SpyCas9 H840A mutant), catalytically dead Cas9 (dCas9) and Superscript III, a commercial RT derived from the Moloney-Murine Leukemia Virus (M-MLV) are used.



FIG. 8 demonstrates dual color experiments that were used to determine if the RT reaction preferentially occurs with the gRNA in cis (bound in the same complex). Two separate experiments were conducted for 5′-extended and 3′-extended gRNAs. Products were analyzed by PAGE. Product ratio calculated as (Cy3cis/Cy3trans)/(Cy5trans/Cy5cis).



FIGS. 9A-9D demonstrates a flap model substrate. FIG. 9A shows a dual-FP reporter for flap-directed mutagenesis. FIG. 9B shows stop codon repair in HEK cells. FIG. 9C shows sequenced yeast clones after flap repair. FIG. 9D shows testing of different flap features in human cells.



FIG. 10 demonstrates prime editing on plasmid substrates. A dual-fluorescent reporter plasmid was constructed for yeast (S. cerevisiae) expression. Expression of this construct in yeast produces only GFP. The in vitro prime editing reaction introduces a point mutation, and transforms the parent plasmid or an in vitro Cas9(H840A) nicked plasmid into yeast. The colonies are visualized by fluorescence imaging. Yeast dual-FP plasmid transformants are shown. Transforming the parent plasmid or an in vitro Cas9(H840A) nicked plasmid results in only green GFP expressing colonies. The prime editing reaction with 5′-extended or 3′-extended gRNAs produces a mix of green and yellow colonies. The latter express both GFP and mCherry. More yellow colonies are observed with the 3′-extended gRNA. A positive control that contains no stop codon is shown as well.



FIG. 11 shows prime editing on plasmid substrates similar to the experiment in FIG. 10, but instead of installing a point mutation in the stop codon, prime editing installs a single nucleotide insertion (left) or deletion (right) that repairs a frameshift mutation and allows for synthesis of downstream mCherry. Both experiments used 3′ extended gRNAs.



FIG. 12 shows editing products of prime editing on plasmid substrates, characterized by Sanger sequencing. Individually colonies from the TRT transformations were selected and analyzed by Sanger sequencing. Precise edits were observed by sequencing select colonies. Green colonies contained plasmids with the original DNA sequence, while yellow colonies contained the precise mutation designed by the prime editing gRNA. No other point mutations or indels were observed.



FIG. 13 shows the potential scope for the new prime editing technology is shown and compared to deaminase-mediated base editor technologies.



FIG. 14 shows a schematic of editing in human cells.



FIG. 15 demonstrates the extension of the primer binding site in gRNA.



FIG. 16 shows truncated gRNAs for adjacent targeting.



FIGS. 17A-17C are graphs displaying the % T to A conversion at the target nucleotide after transfection of components in human embryonic kidney (HEK) cells. FIG. 17A shows data, which presents results using an N-terminal fusion of wild type MLV reverse transcriptase to Cas9(H840A) nickase (32-amino acid linker). FIG. 17B is similar to FIG. 17A, but for C-terminal fusion of the RT enzyme. FIG. 17C is similar to FIG. 17A but the linker between the MLV RT and Cas9 is 60 amino acids long instead of 32 amino acids.



FIG. 18 shows high purity T to A editing at HEK3 site by high-throughput amplicon sequencing. The output of sequencing analysis displays the most abundant genotypes of edited cells.



FIG. 19 shows editing efficiency at the target nucleotide (blue bars) alongside indel rates (orange bars). WT refers to the wild type MLV RT enzyme. The mutant enzymes (M1 through M4) contain the mutations listed to the right. Editing rates were quantified by high throughput sequencing of genomic DNA amplicons.



FIG. 20 shows editing efficiency of the target nucleotide when a single strand nick is introduced in the complementary DNA strand in proximity to the target nucleotide. Nicking at various distances from the target nucleotide was tested (triangles). Editing efficiency at the target base pair (blue bars) is shown alongside the indel formation rate (orange bars). The “none” example does not contain a complementary strand nicking guide RNA. Editing rates were quantified by high throughput sequencing of genomic DNA amplicons.



FIG. 21 demonstrates processed high throughput sequencing data showing the desired T to A transversion mutation and general absence of other major genome editing byproducts.



FIG. 22 provides a schematic of an exemplary process for conducting targeted mutagenesis with an error-prone reverse transcriptase on a target locus using a nucleic acid programmable DNA binding protein (napDNAbp) complexed with an extended guide RNA, i.e., prime editing with an error-prone RT. This process may be referred to as an embodiment of prime editing for targeted mutagenesis. The extended guide RNA comprises an extension at the 3′ or 5′ end of the guide RNA, or at an intramolecular location in the guide RNA. In step (a), the napDNAbp/gRNA complex contacts the DNA molecule and the gRNA guides the napDNAbp to bind to the target locus to be mutagenized. In step (b), a nick in one of the strands of DNA of the target locus is introduced (e.g., by a nuclease or chemical agent), thereby creating an available 3′ end in one of the strands of the target locus. In certain embodiments, the nick is created in the strand of DNA that corresponds to the R-loop strand, i.e., the strand that is not hybridized to the guide RNA sequence. In step (c), the 3′ end DNA strand interacts with the extended portion of the guide RNA in order to prime reverse transcription. In certain embodiments, the 3′ ended DNA strand hybridizes to a specific RT priming sequence on the extended portion of the guide RNA. In step (d), an error-prone reverse transcriptase is introduced which synthesizes a mutagenized single strand of DNA from the 3′ end of the primed site towards the 3′ end of the guide RNA. Exemplary mutations are indicated with an asterisk “*”. This forms a single-strand DNA flap comprising the desired mutagenized region. In step (e), the napDNAbp and guide RNA are released. Steps (f) and (g) relate to the resolution of the single strand DNA flap (comprising the mutagenized region) such that the desired mutagenized region becomes incorporated into the target locus. This process can be driven towards the desired product formation by removing the corresponding 5′ endogenous DNA flap that forms once the 3′ single strand DNA flap invades and hybridizes to the complementary sequence on the other strand. The process can also be driven towards product formation with second strand nicking, as exemplified in FIG. 1F. Following endogenous DNA repair and/or replication processes, the mutagenized region becomes incorporated into both strands of DNA of the DNA locus.



FIG. 23 is a schematic of gRNA design for contracting trinucleotide repeat sequences and trinucleotide repeat contraction with TPRT genome editing (i.e., prime editing). Trinucleotide repeat expansion is associated with a number of human diseases, including Huntington's disease, Fragile X syndrome, and Friedreich's ataxia. The most common trinucleotide repeat contains CAG triplets, though GAA triplets (Friedreich's ataxia) and CGG triplets (Fragile X syndrome) also occur. Inheriting a predisposition to expansion, or acquiring an already expanded parental allele, increases the likelihood of acquiring the disease. Pathogenic expansions of trinucleotide repeats could hypothetically be corrected using prime editing. A region upstream of the repeat region can be nicked by an RNA-guided nuclease, then used to prime synthesis of a new DNA strand that contains a healthy number of repeats (which depends on the particular gene and disease). After the repeat sequence, a short stretch of homology is added that matches the identity of the sequence adjacent to the other end of the repeat (red strand). Invasion of the newly synthesized strand, and subsequent replacement of the endogenous DNA with the newly synthesized flap, leads to a contracted repeat allele.



FIG. 24 is a schematic showing precise 10-nucleotide deletion with prime editing. A guide RNA targeting the HEK3 locus was designed with a reverse transcription template that encodes a 10-nucleotide deletion after the nick site. Editing efficiency in transfected HEK cells was assessed using amplicon sequencing.



FIG. 25 is a schematic showing gRNA design for peptide tagging genes at endogenous genomic loci and peptide tagging with TPRT genome editing (i.e., prime editing). The FlAsH and ReAsH tagging systems comprise two parts: (1) a fluorophore-biarsenical probe, and (2) a genetically encoded peptide containing a tetracysteine motif, exemplified by the sequence FLNCCPGCCMEP (SEQ ID NO: 1). When expressed within cells, proteins containing the tetracysteine motif can be fluorescently labeled with fluorophore-arsenic probes (see ref: J. Am. Chem. Soc., 2002, 124 (21), pp 6063-6076. DOI: 10.1021/ja017687n). The “sortagging” system employs bacterial sortase enzymes that covalently conjugate labeled peptide probes to proteins containing suitable peptide substrates (see ref: Nat. Chem. Biol. 2007 November; 3(11):707-8. DOI: 10.1038/nchembio.2007.31). The FLAG-tag (DYKDDDDK (SEQ ID NO: 2)), V5-tag (GKPIPNPLLGLDST (SEQ ID NO: 3)), GCN4-tag (EELLSKNYHLENEVARLKK (SEQ ID NO: 4)), HA-tag (YPYDVPDYA (SEQ ID NO: 5)), and Myc-tag (EQKLISEEDL (SEQ ID NO: 6)) are commonly employed as epitope tags for immunoassays. The pi-clamp encodes a peptide sequence (FCPF (SEQ ID NO: 622)) that can by labeled with a pentafluoro-aromatic substrates (ref: Nat. Chem. 2016 February; 8(2):120-8. doi: 10.1038/nchem.2413).



FIG. 26A shows precise installation of a His6-tag and a FLAG-tag into genomic DNA. A guide RNA targeting the HEK3 locus was designed with a reverse transcription template that encodes either an 18-nt His-tag insertion or a 24-nt FLAG-tag insertion. Editing efficiency in transfected HEK cells was assessed using amplicon sequencing. Note that the full 24-nt sequence of the FLAG-tag is outside of the viewing frame (sequencing confirmed full and precise insertion). FIG. 26B shows a schematic outlining various applications involving protein/peptide tagging, including (a) rendering proteins soluble or insoluble, (b) changing or tracking the cellular localization of a protein, (c) extending the half-life of a protein, (d) facilitating protein purification, and (e) facilitating the detection of proteins.



FIG. 27 shows an overview of prime editing by installing a protective mutation in PRNP that prevents or halts the progression of prion disease. The PEgRNA sequences correspond to residue numbers 1-20 of SEQ ID NO: 810 on the left (i.e., 5′ of the sgRNA scaffold) and residue numbers 21-43 of SEQ ID NO: 810 on the right (i.e., 3′ of the sgRNA scaffold).



FIG. 28A is a schematic of PE-based insertion of sequences encoding RNA motifs. FIG. 28B is a list (not exhaustive) of some example motifs that could potentially be inserted, and their functions.



FIG. 29A is a depiction of a prime editor. FIG. 29B shows possible modifications to genomic, plasmid, or viral DNA directed by a PE. FIG. 29C shows an example scheme for insertion of a library of peptide loops into a specified protein (in this case GFP) via a library of PEgRNAs. FIG. 29D shows an example of possible programmable deletions of codons or N-, or C-terminal truncations of a protein using different PEgRNAs. Deletions would be predicted to occur with minimal generation of frameshift mutations.



FIG. 30 shows a possible scheme for iterative insertion of codons in a continual evolution system, such as PACE.



FIG. 31 is an illustration of an engineered gRNA showing the gRNA core, ˜20 nt spacer matching the sequence of the targeted gene, the reverse transcription template with immunogenic epitope nucleotide sequence and the primer binding site matching the sequence of the targeted gene.



FIG. 32 is a schematic showing using prime editing as a means to insert known immunogenicity epitopes into endogenous or foreign genomic DNA, resulting in modification of the corresponding proteins.



FIG. 33 is a schematic showing PEgRNA design for primer binding sequence insertions and primer binding insertion into genomic DNA using prime editing for determining off-target editing. In this embodiment, prime editing is conducted inside a living cell, a tissue, or an animal model. As a first step, an appropriate PEgRNA is designed. The top schematic shows an exemplary PEgRNA that may be used in this aspect. The spacer in the PEgRNA (labeled “protospacer”) is complementary to one of the strands of the genomic target. The PE:PEgRNA complex (i.e., the PE complex) installs a single stranded 3′ end flap at the nick site which contains the encoded primer binding sequence and the region of homology (coded by the homology arm of the PEgRNA) that is complementary to the region just downstream of the cut site (in red). Through flap invasion and DNA repair/replication processes, the synthesized strand becomes incorporated into the DNA, thereby installing the primer binding site. This process can occur at the desired genomic target, but also at other genomic sites that might interact with the PEgRNA in an off-target manner (i.e., the PEgRNA guides the PE complex to other off-target sites due to the complementarity of the spacer region to other genomic sites that are not the intended genomic site). Thus, the primer binding sequence may be installed not only at the desired genomic target, but at off-target genomic sites elsewhere in the genome. In order to detect the insertion of these primer binding sites at both the intended genomic target sites and the off-target genomic sites, the genomic DNA (post-PE) can be isolated, fragmented, and ligated to adapter nucleotides (shown in red). Next, PCR may be carried out with PCR oligonucleotides that anneal to the adapters and to the inserted primer binding sequence to amplify on-target and off-target genomic DNA regions into which the primer binding site was inserted by PE. High throughput sequencing then may be conducted and sequences aligned to identify the insertion points of PE-inserted primer binding sequences at either the on-target site or at off-target sites.



FIG. 34 is a schematic showing the precise insertion of a gene with PE.



FIG. 35A is a schematic showing the natural insulin signaling pathway. FIG. 35B is a schematic showing FKBP12-tagged insulin receptor activation controlled by FK1012.



FIG. 36 shows small-molecule monomers. References: humped FK506 mimic (2)107



FIGS. 37A-37B show small-molecule dimers. References: FK1012 495,96; FK1012 5108; FK1012 6107; AP1903 7107; cyclosporin A dimer 898; FK506-cyclosporin A dimer (FkCsA) 9100.



FIGS. 38A-38F provide an overview of prime editing and feasibility studies in vitro and in yeast cells. FIG. 38A shows the 75,122 known pathogenic human genetic variants in ClinVar (accessed July, 2019), classified by type. FIG. 38B shows that a prime editing complex consists of a prime editor (PE) protein containing an RNA-guided DNA-nicking domain, such as Cas9 nickase, fused to an engineered reverse transcriptase domain and complexed with a prime editing guide RNA (PEgRNA). The PE:PEgRNA complex binds the target DNA site and enables a large variety of precise DNA edits at a wide range of DNA positions before or after the target site's protospacer adjacent motif (PAM). FIG. 38C shows that upon DNA target binding, the PE:PEgRNA complex nicks the PAM-containing DNA strand. The resulting free 3′ end hybridizes to the primer-binding site of the PEgRNA. The reverse transcriptase domain catalyzes primer extension using the RT template of the PEgRNA, resulting in a newly synthesized DNA strand containing the desired edit (the 3′ flap).Equilibration between the edited 3′ flap and the unedited 5′ flap containing the original DNA, followed by cellular 5′ flap cleavage and ligation, and DNA repair or replication to resolve the heteroduplex DNA, results in stably edited DNA. FIG. 38D shows in vitro 5′-extended PEgRNA primer extension assays with pre-nicked dsDNA substrates containing 5′-Cy5 labeled PAM strands, dCas9, and a commercial M-MLV RT variant (RT, Superscript III). dCas9 was complexed with PEgRNAs containing RT template of varying lengths, then added to DNA substrates along with the indicated components. Reactions were incubated at 37° C. for 1 hour, then analyzed by denaturing urea PAGE and visualized for Cy5 fluorescence. FIG. 38E shows primer extension assays performed as in FIG. 38D using 3′-extended PEgRNAs pre-complexed with dCas9 or Cas9 H840A nickase, and pre-nicked or non-nicked 5′-Cy5-labeled dsDNA substrates. FIG. 38F shows yeast colonies transformed with GFP-mCherry fusion reporter plasmids edited in vitro with PEgRNAs, Cas9 nickase, and RT. Plasmids containing nonsense or frameshift mutations between GFP and mCherry were edited with 5′-extended or 3′-extended PEgRNAs that restore mCherry translation via transversion mutation, 1-bp insertion, or 1-bp deletion. GFP and mCherry double-positive cells (yellow) reflect successful editing.



FIGS. 39A-39D show prime editing of genomic DNA in human cells by PE1 and PE2. FIG. 39A shows PEgRNAs contain a spacer sequence, a sgRNA scaffold, and a 3′ extension containing a primer-binding site (green) and a reverse transcription (RT) template (purple), which contains the edited base(s) (red). The primer-binding site hybridizes to the PAM-containing DNA strand immediately upstream of the site of nicking. The RT template is homologous to the DNA sequence downstream of the nick, with the exception of the encoded edit. FIG. 39B shows an installation of a T•A-to-A•T transversion edit at the HEK3 site in HEK293T cells using Cas9 H840A nickase fused to wild-type M-MLV reverse transcriptase (PE1) and PEgRNAs of varying primer-binding site lengths. FIG. 39 C shows the use of an engineered pentamutant M-MLV reverse transcriptase (D200N, L603W, T306K, W313F, T330P) in PE2 substantially improves prime editing transversion efficiencies at five genomic sites in HEK293T cells, and small insertion and small deletion edits at HEK3. FIG. 39D is a comparison of PE2 editing efficiencies with varying RT template lengths at five genomic sites in HEK293T cells. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 40A-40C show PE3 and PE3b systems nick the non-edited strand to increase prime editing efficiency. FIG. 40A is an overview of the prime editing by PE3. After initial synthesis of the edited strand, DNA repair will remove either the newly synthesized strand containing the edit (3′ flap excision) or the original genomic DNA strand (5′ flap excision). 5′ flap excision leaves behind a DNA heteroduplex containing one edited strand and one non-edited strand. Mismatch repair machinery or DNA replication could resolve the heteroduplex to give either edited or non-edited products. Nicking the non-edited strand favors repair of that strand, resulting in preferential generation of stable duplex DNA containing the desired edit. FIG. 40B shows the effect of complementary strand nicking on PE3-mediated prime editing efficiency and indel formation. “None” refers to PE2 controls, which do not nick the complementary strand. FIG. 40C is a comparison of editing efficiencies with PE2 (no complementary strand nick), PE3 (general complementary strand nick), and PE3b (edit-specific complementary strand nick). All editing yields reflect the percentage of total sequencing reads that contain the intended edit and do not contain indels among all treated cells, with no sorting. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 41A-41K show targeted insertions, deletions, and all 12 types of point mutations with PE3 at seven endogenous human genomic loci in HEK293T cells. FIG. 41A is a graph showing all 12 types of single-nucleotide transition and transversion edits from position +1 to +8 (counting the location of the PEgRNA-induced nick as between position +1 and −1) of the HEK3 site using a 10-nt RT template. FIG. 41B is a graph showing long-range PE3 transversion edits at the HEK3 site using a 34-nt RT template. FIGS. 41C-41H are graphs showing all 12 types of transition and transversion edits at various positions in the prime editing window for (FIG. 41C) RNF2, (FIG. 41D) FANCF, (FIG. 41E) EMX1, (FIG. 41F) RUNX1, (FIG. 41G) VEGFA, and (FIG. 41H) DNMT1. FIG. 41I is a graph showing targeted 1- and 3-bp insertions, and 1- and 3-bp deletions with PE3 at seven endogenous genomic loci. FIG. 41J is a graph showing the targeted precise deletions of 5 to 80 bp at the HEK3 target site. FIG. 41K is a graph showing a combination edits of insertions and deletions, insertions and point mutations, deletions and point mutations, and double point mutations at three endogenous genomic loci. All editing yields reflect the percentage of total sequencing reads that contain the intended edit and do not contain indels among all treated cells, with no sorting. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 42A-42H show the comparison of prime editing and base editing, and off-target editing by Cas9 and PE3 at known Cas9 off-target sites. FIG. 42A shows total C•G-to-T•A editing efficiency at the same target nucleotides for PE2, PE3, BE2max, and BE4max at endogenous HEK3, FANCF, and EMX1 sites in HEK293T cells. FIG. 42B shows indel frequency from treatments in FIG. 42A. FIG. 42C shows the editing efficiency of precise C•G-to-T•A edits (without bystander edits or indels) for PE2, PE3, BE2max, and BE4max at HEK3, FANCF, and EMX1. For EMX1, precise PE combination edits of all possible combinations of C•G-to-T•A conversion at the three targeted nucleotides are also shown. FIG. 42D shows the total A•T-to-G•C editing efficiency for PE2, PE3, ABEdmax, and ABEmax at HEK3 and FANCF. FIG. 42E shows the precise A•T-to-G•C editing efficiency without bystander edits or indels for at HEK3 and FANCF. FIG. 42F shows indel frequency from treatments in FIG. 42D. FIG. 42G shows the average triplicate editing efficiencies (percentage sequencing reads with indels) in HEK293T cells for Cas9 nuclease at four on-target and 16 known off-target sites. The 16 off-target sites examined were the top four previously reported off-target sites118,159 for each of the four on-target sites. For each on-target site, Cas9 was paired with a sgRNA or with each of four PEgRNAs that recognize the same protospacer. FIG. 42H shows the average triplicate on-target and off-target editing efficiencies and indel efficiencies (below in parentheses) in HEK293T cells for PE2 or PE3 paired with each PEgRNA in (FIG. 42G). On-target editing yields reflect the percentage of total sequencing reads that contain the intended edit and do not contain indels among all treated cells, with no sorting. Off-target editing yields reflect off-target locus modification consistent with prime editing. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 43A-43I show prime editing in various human cell lines and primary mouse cortical neurons, installation and correction of pathogenic transversion, insertion, or deletion mutations, and comparison of prime editing and HDR. FIG. 43A is a graph showing the installation (via T•A-to-A•T transversion) and correction (via A•T-to-T•A transversion) of the pathogenic E6V mutation in HBB in HEK293T cells. Correction either to wild-type HBB, or to HBB containing a silent mutation that disrupts the PEgRNA PAM, is shown. FIG. 43B is a graph showing the installation (via 4-bp insertion) and correction (via 4-bp deletion) of the pathogenic HEXA 1278+TATC allele in HEK293T cells. Correction either to wild-type HEXA, or to HEXA containing a silent mutation that disrupts the PEgRNA PAM, is shown. FIG. 43C is a graph showing the installation of the protective G127V variant in PRNP in HEK293T cells via G•C-to-T•A transversion. FIG. 43D is a graph showing prime editing in other human cell lines including K562 (leukemic bone marrow cells), U2OS (osteosarcoma cells), and HeLa (cervical cancer cells). FIG. 43E is a graph showing the installation of a G•C-to-T•A transversion mutation in DNMT1 of mouse primary cortical neurons using a dual split-intein PE3 lentivirus system, in which the N-terminal half is Cas9 (1-573) fused to N-intein and through a P2A self-cleaving peptide to GFP-KASH, and the C-terminal half is the C-intein fused to the remainder of PE2. PE2 halves are expressed from a human synapsin promoter that is highly specific for mature neurons. Sorted values reflect editing or indels from GFP-positive nuclei, while unsorted values are from all nuclei. FIG. 43F is a comparison of PE3 and Cas9-mediated HDR editing efficiencies at endogenous genomic loci in HEK293T cells. FIG. 43G is a comparison of PE3 and Cas9-mediated HDR editing efficiencies at endogenous genomic loci in K562, U2OS, and HeLa cells. FIG. 43H is a comparison of PE3 and Cas9-mediated HDR indel byproduct generation in HEK293T, K562, U2OS, and HeLa cells. FIG. 43I shows targeted insertion of a His6 tag (18 bp), FLAG epitope tag (24 bp), or extended LoxP site (44 bp) in HEK293T cells by PE3. All editing yields reflect the percentage of total sequencing reads that contain the intended edit and do not contain indels among all treated cells. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 44A-44G show in vitro prime editing validation studies with fluorescently labeled DNA substrates. FIG. 44A shows electrophoretic mobility shift assays with dCas9, 5′-extended PEgRNAs and 5′-Cy5-labeled DNA substrates. PEgRNAs 1 through 5 contain a 15-nt linker sequence (linker A for PEgRNA 1, linker B for PEgRNAs 2 through 5) between the spacer and the PBS, a 5-nt PBS sequence, and RT templates of 7 nt (PEgRNAs 1 and 2), 8 nt (PEgRNA 3), 15 nt (PEgRNA 4), and 22 nt (PEgRNA 5). PEgRNAs are those used in FIGS. 44E and 44F; full sequences are listed in Tables 2A-2C. FIG. 44B shows in vitro nicking assays of Cas9 H840A using 5′-extended and 3′-extended PEgRNAs. FIG. 44C shows Cas9-mediated indel formation in HEK293T cells at HEK3 using 5′-extended and 3′-extended PEgRNAs. FIG. 44D shows an overview of prime editing in vitro biochemical assays. 5′-Cy5-labeled pre-nicked and non-nicked dsDNA substrates were tested. sgRNAs, 5′-extended PEgRNAs, or 3′-extended PEgRNAs were pre-complexed with dCas9 or Cas9 H840A nickase, then combined with dsDNA substrate, M-MLV RT, and dNTPs. Reactions were allowed to proceed at 37° C. for 1 hour prior to separation by denaturing urea PAGE and visualization by Cy5 fluorescence. FIG. 44E shows primer extension reactions using 5′-extended PEgRNAs, pre-nicked DNA substrates, and dCas9 lead to significant conversion to RT products. FIG. 44F shows primer extension reactions using 5′-extended PEgRNAs as in FIG. 44B, with non-nicked DNA substrate and Cas9 H840A nickase. Product yields are greatly reduced by comparison to pre-nicked substrate. FIG. 44G shows an in vitro primer extension reaction using a 3′-PEgRNA generates a single apparent product by denaturing urea PAGE. The RT product band was excised, eluted from the gel, then subjected to homopolymer tailing with terminal transferase (TdT) using either dGTP or dATP. Tailed products were extended by poly-T or poly-C primers, and the resulting DNA was sequenced. Sanger traces indicate that three nucleotides derived from the gRNA scaffold were reverse transcribed (added as the final 3′ nucleotides to the DNA product). Note that in mammalian cell prime editing experiments, PEgRNA scaffold insertion is much rarer than in vitro (FIGS. 56A-56D), potentially due to the inability of the tethered reverse transcriptase to access the Cas9-bound guide RNA scaffold, and/or cellular excision of mismatched 3′ ends of 3′ flaps containing PEgRNA scaffold sequences.



FIGS. 45A-45G show cellular repair in yeast of 3′ DNA flaps from in vitro prime editing reactions. FIG. 45A shows that dual fluorescent protein reporter plasmids contain GFP and mCherry open reading frames separated by a target site encoding an in-frame stop codon, a +1 frameshift, or a −1 frameshift. Prime editing reactions were carried out in vitro with Cas9 H840A nickase, PEgRNA, dNTPs, and M-MLV reverse transcriptase, and then transformed into yeast. Colonies that contain unedited plasmids produce GFP but not mCherry. Yeast colonies containing edited plasmids produce both GFP and mCherry as a fusion protein. FIG. 45B shows an overlay of GFP and mCherry fluorescence for yeast colonies transformed with reporter plasmids containing a stop codon between GFP and mCherry (unedited negative control, top), or containing no stop codon or frameshift between GFP and mCherry (pre-edited positive control, bottom). FIGS. 45C-45F show a visualization of mCherry and GFP fluorescence from yeast colonies transformed with in vitro prime editing reaction products. FIG. 45C shows a stop codon correction via T•A-to-A•T transversion using a 3′-extended PEgRNA, or a 5′-extended PEgRNA, as shown in FIG. 45D. FIG. 45E shows a +1 frameshift correction via a 1-bp deletion using a 3′-extended PEgRNA. FIG. 45F shows a −1 frameshift correction via a 1-bp insertion using a 3′-extended PEgRNA. FIG. 45G shows Sanger DNA sequencing traces from plasmids isolated from GFP-only colonies in FIG. 45B and GFP and mCherry double-positive colonies in FIG. 45C.



FIGS. 46A-46F show correct editing versus indel generation with PE1. FIG. 46A shows T•A-to-A•T transversion editing efficiency and indel generation by PE1 at the +1 position of HEK3 using PEgRNAs containing 10-nt RT templates and a PBS sequences ranging from 8-17 nt. FIG. 46B shows G•C-to-T•A transversion editing efficiency and indel generation by PE1 at the +5 position of EMX1 using PEgRNAs containing 13-nt RT templates and a PBS sequences ranging from 9-17 nt. FIG. 46C shows G•C-to-T•A transversion editing efficiency and indel generation by PE1 at the +5 position of FANCF using PEgRNAs containing 17-nt RT templates and a PBS sequences ranging from 8-17 nt. FIG. 46D shows C•G-to-A•T transversion editing efficiency and indel generation by PE1 at the +1 position of RNF2 using PEgRNAs containing 11-nt RT templates and a PBS sequences ranging from 9-17 nt. FIG. 46E shows G•C-to-T•A transversion editing efficiency and indel generation by PE1 at the +2 position of HEK4 using PEgRNAs containing 13-nt RT templates and a PBS sequences ranging from 7-15 nt. FIG. 46F shows PE1-mediated +1 T deletion, +1 A insertion, and +1 CTT insertion at the HEK3 site using a 13-nt PBS and 10-nt RT template. Sequences of PEgRNAs are those used in FIG. 39C (see Tables 3A-3R). Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 47A-47S show the evaluation of M-MLV RT variants for prime editing. FIG. 47A shows the abbreviations for prime editor variants used in this figure. FIG. 47B shows targeted insertion and deletion edits with PE1 at the HEK3 locus. FIGS. 47C-47H show a comparison of 18 prime editor constructs containing M-MLV RT variants for their ability to install a +2 G•C-to-C•G transversion edit at HEK3 as shown in FIG. 47C, a 24-bp FLAG insertion at HEK3 as shown in FIG. 47D, a +1 C•G-to-A•T transversion edit at RNF2 as shown in FIG. 47E, a +1 G•C-to-C•G transversion edit at EMX1 as shown in FIG. 47F, a +2 T•A-to-A•T transversion edit at HBB as shown in FIG. 47G, and a +1 G•C-to-C•G transversion edit at FANCF as shown in FIG. 47H. FIGS. 47I-47N show a comparison of four prime editor constructs containing M-MLV variants for their ability to install the edits shown in FIGS. 47C-47H in a second round of independent experiments. FIGS. 47O-47S show PE2 editing efficiency at five genomic loci with varying PBS lengths. FIG. 47O shows a +1 T•A-to-A•T variation at HEK3. FIG. 47P shows a +5 G•C-to-T•A variation at EMX1. FIG. 47Q shows a +5 G•C-to-T•A variation at FANCF. FIG. 47R shows a +1 C•G-to-A•T variation at RNF2. FIG. 47S shows a +2 G•C-to-T•A variation at HEK4. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 48A-48C show design features of PEgRNA PBS and RT template sequences. FIG. 48A shows PE2-mediated +5 G•C-to-T•A transversion editing efficiency (blue line) at VEGFA in HEK293T cells as a function of RT template length. Indels (gray line) are plotted for comparison. The sequence below the graph shows the last nucleotide templated for synthesis by the PEgRNA. G nucleotides (templated by a C in the PEgRNA) are highlighted; RT templates that end in C should be avoided during PEgRNA design to maximize prime editing efficiencies. FIG. 48B shows +5 G•C-to-T•A transversion editing and indels for DNMT1 as in FIG. 48A. FIG. 48C shows +5 G•C-to-T•A transversion editing and indels for RUNX1 as in FIG. 48A. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 49A-49B show the effects of PE2, PE2 R110S K103L, Cas9 H840A nickase, and dCas9 on cell viability. HEK293T cells were transfected with plasmids encoding PE2, PE2 R110S K103L, Cas9 H840A nickase, or dCas9, together with a HEK3-targeting PEgRNA plasmid. Cell viability was measured every 24 hours post-transfection for 3 days using the CellTiter-Glo 2.0 assay (Promega). FIG. 49A shows viability, as measured by luminescence, at 1, 2, or 3 days post-transfection. Values and error bars reflect the mean and s.e.m. of three independent biological replicates each performed in technical triplicate. FIG. 49B shows percent editing and indels for PE2, PE2 R110S K103L, Cas9 H840A nickase, or dCas9, together with a HEK3-targeting PEgRNA plasmid that encodes a +5 G to A edit. Editing efficiencies were measured on day 3 post-transfection from cells treated alongside of those used for assaying viability in FIG. 49A. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 50A-50B show PE3-mediated HBB E6V correction and HEXA 1278+TATC correction by various PEgRNAs. FIG. 50A shows a screen of 14 PEgRNAs for correction of the HBB E6V allele in HEK293T cells with PE3. All PEgRNAs evaluated convert the HBB E6V allele back to wild-type HBB without the introduction of any silent PAM mutation. FIG. 50B shows a screen of 41 PEgRNAs for correction of the HEXA 1278+TATC allele in HEK293T cells with PE3 or PE3b. Those PEgRNAs labeled HEXAs correct the pathogenic allele by a shifted 4-bp deletion that disrupts the PAM and leaves a silent mutation. Those PEgRNAs labeled HEXA correct the pathogenic allele back to wild-type. Entries ending in “b” use an edit-specific nicking sgRNA in combination with the PEgRNA (the PE3b system). Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 51A-51G show a PE3 activity in human cell lines and a comparison of PE3 and Cas9-initiated HDR. Efficiency of generating the correct edit (without indels) and indel frequency for PE3 and Cas9-initiated HDR in HEK293T cells as shown in FIG. 51A, K562 cells as shown in FIG. 51B, U2OS cells as shown in FIG. 51C, and HeLa cells as shown in FIG. 51D. Each bracketed editing comparison installs identical edits with PE3 and Cas9-initiated HDR. Non-targeting controls are PE3 and a PEgRNA that targets a non-target locus. FIG. 51E shows control experiments with non-targeting PEgRNA+PE3, and with dCas9+sgRNA, compared with wild-type Cas9 HDR experiments confirming that ssDNA donor HDR template, a common contaminant that artificially elevates apparent HDR efficiencies, does not contribute to the HDR measurements in FIGS. 51A-51D. FIGS. 51F-51G show example HEK3 site allele tables from genomic DNA samples isolated from K562 cells after editing with PE3 or with Cas9-initiated HDR. Alleles were sequenced on an Illumina MiSeq and analyzed with CRISPResso2178. The reference HEK3 sequence from this region is at the top. Allele tables are shown for a non-targeting PEgRNA negative control, a +1 CTT insertion at HEK3 using PE3, and a +1 CTT insertion at HEK3 using Cas9-initiated HDR. Allele frequencies and corresponding Illumina sequencing read counts are shown for each allele. All alleles observed with frequency ≥0.20% are shown. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 52A-52D show distribution by length of pathogenic insertions, duplications, deletions, and indels in the ClinVar database. The ClinVar variant summary was downloaded from NCBI Jul. 15, 2019. The lengths of reported insertions, deletions, and duplications were calculated using reference and alternate alleles, variant start and stop positions, or appropriate identifying information in the variant name. Variants that did not report any of the above information were excluded from the analysis. The lengths of reported indels (single variants that include both insertions and deletions relative to the reference genome) were calculated by determining the number of mismatches or gaps in the best pairwise alignment between the reference and alternate alleles.



FIGS. 53A-53E show FACS gating examples for GFP-positive cell sorting. Below are examples of original batch analysis files outlining the sorting strategy used for generating HEXA 1278+TATC and HBB E6V HEK293T cell lines. The image data was generated on a Sony LE-MA900 cytometer using Cell Sorter Software v. 3.0.5. Graphic 1 shows gating plots for cells that do not express GFP. Graphic 2 shows an example sort of P2A-GFP-expressing cells used for isolating the HBB E6V HEK293T cell lines. HEK293T cells were initially gated on population using FSC-A/BSC-A (Gate A), then sorted for singlets using FSC-A/FSC-H (Gate B). Live cells were sorted for by gating DAPI-negative cells (Gate C). Cells with GFP fluorescence levels that were above those of the negative-control cells were sorted for using EGFP as the fluorochrome (Gate D). FIG. 53A shows HEK293T cells (GFP-negative). FIG. 53B shows a representative plot of FACS gating for cells expressing PE2-P2A-GFP. FIG. 53C shows the genotypes for HEXA 1278+TATC homozygote HEK293T cells. FIGS. 53D-53E show allele tables for HBB E6V homozygote HEK293T cell lines.



FIG. 54 is a schematic which summarizes the PEgRNA cloning procedure.



FIGS. 55A-55G are schematics of PEgRNA designs. FIG. 55A shows a simple diagram of PEgRNA with domains labeled (left) and bound to nCas9 at a genomic site (right). FIG. 55B shows various types of modifications to PEgRNA which are anticipated to increase activity. FIG. 55C shows modifications to PEgRNA to increase transcription of longer RNAs via promoter choice and 5′, 3′ processing and termination. FIG. 55D shows the lengthening of the P1 system, which is an example of a scaffold modification. FIG. 55E shows that the incorporation of synthetic modifications within the template region, or elsewhere within the PEgRNA, could increase activity. FIG. 55F shows that a designed incorporation of minimal secondary structure within the template could prevent formation of longer, more inhibitory, secondary structure. FIG. 55G shows a split PEgRNA with a second template sequence anchored by an RNA element at the 3′ end of the PEgRNA (left). Incorporation of elements at the 5′ or 3′ ends of the PEgRNA could enhance RT binding.



FIGS. 56A-56D show the incorporation of PEgRNA scaffold sequence into target loci. HTS data were analyzed for PEgRNA scaffold sequence insertion as described in FIGS. 60A-60B. FIG. 56A shows an analysis for the EMX1 locus. Shown is the % of total sequencing reads containing one or more PEgRNA scaffold sequence nucleotides within an insertion adjacent to the RT template (left); the percentage of total sequencing reads containing a PEgRNA scaffold sequence insertion of the specified length (middle); and the cumulative total percentage of PEgRNA insertion up to and including the length specified on the X axis. FIG. 56B shows the same as FIG. 56A, but for FANCF. FIG. 56C shows the same as in FIG. 56A but for HEK3. FIG. 56D shows the same as FIG. 56A but for RNF2. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 57A-57I show the effects of PE2, PE2-dRT, and Cas9 H840A nickase on transcriptome-wide RNA abundance. Analysis of cellular RNA, depleted for ribosomal RNA, isolated from HEK293T cells expressing PE2, PE2-dRT, or Cas9 H840A nickase and a PRNP-targeting or HEXA-targeting PEgRNA. RNAs corresponding to 14,410 genes and 14,368 genes were detected in PRNP and HEXA samples, respectively. FIGS. 57A-57F show Volcano plot displaying the −log 10 FDR-adjusted p-value vs. log 2-fold change in transcript abundance for Aeach RNA, comparing (FIG. 57A) PE2 vs. PE2-dRT with PRNP-targeting PEgRNA, (FIG. 57B) PE2 vs. Cas9 H840A with PRNP-targeting PEgRNA, (FIG. 57C) PE2-dRT vs. Cas9 H840A with PRNP-targeting PEgRNA, (FIG. 57D) PE2 vs. PE2-dRT with HEXA-targeting PEgRNA, (FIG. 57E) PE2 vs. Cas9 H840A with HEXA-targeting PEgRNA, (FIG. 57F) PE2-dRT vs. Cas9 H840A with HEXA-targeting PEgRNA. Red dots indicate genes that show ≥2-d change in relative abundance that are statistically significant (FDR-adjusted p<0.05). FIGS. 57G-57I are Venn diagrams of upregulated and downregulated transcripts (≥2-fold change) comparing PRNP and HEXA samples for (FIG. 57G) PE2 vs PE2-dRT, (FIG. 57H) PE2 vs. Cas9 H840A, and (FIG. 57I) PE2-dRT vs. Cas9 H840A.



FIGS. 58A-58B show representative FACS gating for neuronal nuclei sorting. Nuclei were sequentially gated on the basis of DyeCycle Ruby signal, FSC/SSC ratio, SSC-Width/SSC-height ratio, and GFP/DyeCycle ratio.



FIGS. 59A-59G show the protocol for cloning 3′-extended PEgRNAs into mammalian U6 expression vectors by Golden Gate assembly. FIG. 59A shows the cloning overview. FIG. 59B shows ‘Step 1: Digest pU6-PEgRNA-GG-Vector plasmid (component 1)’. FIG. 59C shows ‘Steps 2 and 3: Order and anneal oligonucleotide parts (components 2, 3, and 4)’. FIG. 59D shows ‘Step 2.b.ii.: sgRNA scaffold phosphorylation (unnecessary if oligonucleotides were purchased phosphorylated)’. FIG. 59E shows ‘Step 4: PEgRNA assembly’. FIG. 59F shows ‘Steps 5 and 6: Transformation of assembled plasmids’. FIG. 59G shows a diagram summarizing the PEgRNA cloning protocol.



FIGS. 60A-60B show the Python script for quantifying PEgRNA scaffold integration. A custom python script was generated to characterize and quantify PEgRNA insertions at target genomic loci. The script iteratively matches text strings of increasing length taken from a reference sequence (guide RNA scaffold sequence) to the sequencing reads within fastq files, and counts the number of sequencing reads that match the search query. Each successive text string corresponds to an additional nucleotide of the guide RNA scaffold sequence. Exact length integrations and cumulative integrations up to a specified length were calculated in this manner. At the start of the reference sequence, 5 to 6 bases of the 3′ end of the new DNA strand synthesized by the reverse transcriptase are included to ensure alignment and accurate counting of short slices of the sgRNA.



FIG. 61 is a graph showing the percent of total sequencing reads with the specified edit for SaCas9(N580A)-MMLV RT HEK3+6 C>A. The values for the correct edits as well as indels are shown.



FIGS. 62A-62B show the importance of the protospacer for efficient installation of a desired edit at a precise location with prime editing. FIG. 62A is a graph showing the percent of total sequencing reads with target T•A base pairs converted to A•T for various HEK3 loci. FIG. 62B is a sequence analysis showing the same.



FIG. 63 is a graph showing SpCas9 PAM variants in PAM editing (N=3). The percent of total sequencing reads with the targeted PAM edit is shown for SpCas9(H840A)-VRQR-MMLV RT, where NGA>NTA, and for SpCas9(H840A)-VRER-MMLV RT, where NGCG>NTCG. The PEgRNA primer binding site (PBS) length, RT template (RT) length, and PE system used are listed.



FIGS. 64A-64F depict a schematic showing the introduction of various site-specific recombinase (SSR) targets into the genome using PE. FIG. 64A provides a general schematic of the insertion of a recombinase target sequence by a prime editor. FIG. 64B shows how a single SSR target inserted by PE can be used as a site for genomic integration of a DNA donor template. FIG. 64C shows how a tandem insertion of SSR target sites can be used to delete a portion of the genome. FIG. 64D shows how a tandem insertion of SSR target sites can be used to invert a portion of the genome. FIG. 64E shows how the insertion of two SSR target sites at two distal chromosomal regions can result in chromosomal translocation. FIG. 64F shows how the insertion of two different SSR target sites in the genome can be used to exchange a cassette from a DNA donor template.



FIG. 65 shows in 1) the PE-mediated synthesis of a SSR target site in a human cell genome and 2) the use of that SSR target site to integrate a DNA donor template comprising a GFP expression marker. Once successfully integrated, the GFP causes the cell to fluoresce.



FIG. 66 depicts one embodiment of a prime editor being provided as two PE half proteins which regenerate as whole prime editor through the self-splicing action of the split-intein halves located at the end or beginning of each of the prime editor half proteins.



FIGS. 67A-67B depict the mechanism of intein removal from a polypeptide sequence and the reformation of a peptide bond between the N-terminal and the C-terminal extein sequences. FIG. 67A depicts the general mechanism of two half proteins each containing half of an intein sequence, which when in contact within a cell result in a fully-functional intein which then undergoes self-spicing and excision. The process of excision results in the formation of a peptide bond between the N-terminal protein half (or the “N extein”) and the C-terminal protein half (or the “C extein”) to form a whole, single polypeptide comprising the N extein and the C extein portions. In various embodiments, the N extein may correspond to the N-terminal half of a split prime editor fusion protein and the C extein may correspond to the C-terminal half of a split prime editor. (b) shows a chemical mechanism of intein excision and the reformation of a peptide bond that joins the N extein half (the red-colored half) and the C extein half (the blue-colored half). Excision of the split inteins (i.e., the N intein and the C intein in the split intein configuration) may also be referred to as “trans splicing” as it involves the splicing action of two separate components provided in trans.



FIG. 68A demonstrates that delivery of both split intein halves of SpPE (SEQ ID NOs: 3875, 3876) at the linker maintains activity at three test loci when co-transfected into HEK293T cells.



FIG. 68B demonstrates that delivery of both split intein halves of SaPE2 (e.g., SEQ ID NO: 443 and SEQ ID NO: 450) recapitulate activity of full length SaPE2 (SEQ ID NO: 134) when co-transfected into HEK293T cells. Residues indicated in quotes are the sequence of amino acids 741-743 in SaCas9 (first residues of the C-terminal extein) which are important for the intein trans splicing reaction. ‘SMP’ are the native residues, which we also mutated to the ‘CFN’ consensus splicing sequence. The consensus sequence is shown to yield the highest reconstitution as measured by prime editing percentage.



FIG. 68C provides data showing that various disclosed PE ribonucleoprotein complexes (PE2 at high concentration, PE3 at high concentration and PE3 at low concentration) can be delivered in this manner



FIG. 69 shows a bacteriophage plaque assay to determine PE effectiveness in PANCE. Plaques (dark circles) indicate phage able to successfully infect E. coli. Increasing concentration of L-rhamnose results in increased expression of PE and an increase in plaque formation. Sequencing of plaques revealed the presence of the PE-installed genomic edit.



FIGS. 70A-70I provide an example of an edited target sequence as an illustration of a step-by-step instruction for designing PEgRNAs and nicking-sgRNAs for prime editing. FIG. 70A: Step 1. Define the target sequence and the edit. Retrieve the sequence of the target DNA region (˜200 bp) centered around the location of the desired edit (point mutation, insertion, deletion, or combination thereof). FIG. 70B: Step 2. Locate target PAMs. Identify PAMs in proximity to the edit location. Be sure to look for PAMs on both strands. While PAMs close to the edit position are preferred, it is possible to install edits using protospacers and PAMs that place the nick>30 nt from the edit position. FIG. 70C: Step 3. Locate the nick sites. For each PAM being considered, identify the corresponding nick site. For Sp Cas9 H840A nickase, cleavage occurs in the PAM-containing strand between the 3rd and 4th bases 5′ to the NGG PAM. All edited nucleotides must exist 3′ of the nick site, so appropriate PAMs must place the nick 5′ to the target edit on the PAM-containing strand. In the example shown below, there are two possible PAMs. For simplicity, the remaining steps will demonstrate the design of a PEgRNA using PAM 1 only. FIG. 70D: Step 4. Design the spacer sequence. The protospacer of Sp Cas9 corresponds to the 20 nucleotides 5′ to the NGG PAM on the PAM-containing strand. Efficient Pol III transcription initiation requires a G to be the first transcribed nucleotide. If the first nucleotide of the protospacer is a G, the spacer sequence for the PEgRNA is simply the protospacer sequence. If the first nucleotide of the protospacer is not a G, the spacer sequence of the PEgRNA is G followed by the protospacer sequence. FIG. 70E: Step 5. Design a primer binding site (PBS). Using the starting allele sequence, identify the DNA primer on the PAM-containing strand. The 3′ end of the DNA primer is the nucleotide just upstream of the nick site (i.e. the 4th base 5′ to the NGG PAM for Sp Cas9). As a general design principle for use with PE2 and PE3, a PEgRNA primer binding site (PBS) containing 12 to 13 nucleotides of complementarity to the DNA primer can be used for sequences that contain ˜40-60% GC content. For sequences with low GC content, longer (14- to 15-nt) PBSs should be tested. For sequences with higher GC content, shorter (8- to 11-nt) PBSs should be tested. Optimal PBS sequences should be determined empirically, regardless of GC content. To design a length-p PBS sequence, take the reverse complement of the first p nucleotides 5′ of the nick site in the PAM-containing strand using the starting allele sequence. FIG. 70F: Step 6. Design an RT template. The RT template encodes the designed edit and homology to the sequence adjacent to the edit. Optimal RT template lengths vary based on the target site. For short-range edits (positions +1 to +6), it is recommended to test a short (9 to 12 nt), a medium (13 to 16 nt), and a long (17 to 20 nt) RT template. For long-range edits (positions +7 and beyond), it is recommended to use RT templates that extend at least 5 nt (preferably 10 or more nt) past the position of the edit to allow for sufficient 3′ DNA flap homology. For long-range edits, several RT templates should be screened to identify functional designs. For larger insertions and deletions (>5 nt), incorporation of greater 3′ homology (˜20 nt or more) into the RT template is recommended. Editing efficiency is typically impaired when the RT template encodes the synthesis of a G as the last nucleotide in the reverse transcribed DNA product (corresponding to a C in the RT template of the PEgRNA). As many RT templates support efficient prime editing, avoidance of G as the final synthesized nucleotide is recommended when designing RT templates. To design a length-r RT template sequence, use the desired allele sequence and take the reverse complement of the first r nucleotides 3′ of the nick site in the strand that originally contained the PAM. Note that compared to SNP edits, insertion or deletion edits using RT templates of the same length will not contain identical homology. FIG. 70G: Step 7. Assemble the full PEgRNA sequence. Concatenate the PEgRNA components in the following order (5′ to 3′): spacer, scaffold, RT template and PBS. FIG. 70H: Step 8. Designing nicking-sgRNAs for PE3. Identify PAMs on the non-edited strand upstream and downstream of the edit. Optimal nicking positions are highly locus-dependent and should be determined empirically. In general, nicks placed 40 to 90 nucleotides 5′ to the position across from the PEgRNA-induced nick lead to higher editing yields and fewer indels. A nicking sgRNA has a spacer sequence that matches the 20-nt protospacer in the starting allele, with the addition of a 5′-G if the protospacer does not begin with a G. FIG. 70I: Step 9. Designing PE3b nicking-sgRNAs. If a PAM exists in the complementary strand and its corresponding protospacer overlaps with the sequence targeted for editing, this edit could be a candidate for the PE3b system. In the PE3b system, the spacer sequence of the nicking-sgRNA matches the sequence of the desired edited allele, but not the starting allele. The PE3b system operates efficiently when the edited nucleotide(s) falls within the seed region (˜10 nt adjacent to the PAM) of the nicking-sgRNA protospacer. This prevents nicking of the complementary strand until after installation of the edited strand, preventing competition between the PEgRNA and the sgRNA for binding the target DNA. PE3b also avoids the generation of simultaneous nicks on both strands, thus reducing indel formation significantly while maintaining high editing efficiency. PE3b sgRNAs should have a spacer sequence that matches the 20-nt protospacer in the desired allele, with the addition of a 5′ G if needed.



FIG. 71A shows the nucleotide sequence of a SpCas9 PEgRNA molecule (top) which terminates at the 3′ end in a “UUU” and does not contain a toeloop element. The lower portion of the figure depicts the same SpCas9 PEgRNA molecule but is further modified to contain a toeloop element having the sequence 5′-“GAAANNNNN”-3′ inserted immediately before the “UUU” 3′ end. The “N” can be any nucleobase.



FIG. 71B demonstrates that the efficiency of prime editing in HEK cells or EMX cells is increased using PEgRNA containing toeloop elements, whereas the percent of indel formation is largely unchanged.



FIGS. 72A-72C depict alternative PEgRNA configurations that can be used in prime editing. FIG. 72A depicts the PE2:PEgRNA embodiment of prime editing. This embodiment involves a PE2 (a fusion protein comprising a Cas9 and a reverse transcriptase) complexed with a PEgRNA (as also described in FIGS. 1A-1I and/or FIGS. 3A-3E). In this embodiment, the template for reverse transcription is incorporated into a 3′ extension arm on the sgRNA to make the PEgRNA, and the DNA polymerase enzyme is a reverse transcriptase (RT) fused directly to Cas9. FIG. 72B depict the MS2cp-PE2:sgRNA+tPERT embodiment. This embodiment comprises a PE2 fusion (Cas9+a reverse transcriptase) that is further fused to the MS2 bacteriophage coat protein (MS2cp) to form the MS2cp-PE2 fusion protein. To achieve prime editing, the MS2cp-PE2 fusion protein is complexed with an sgRNA that targets the complex to a specific target site in the DNA. The embodiment then involves the introduction of a trans prime editing RNA template (“tPERT”), which operates in place of a PEgRNA by providing a primer binding site (PBS) and an DNA synthesis template on separate molecule, i.e., the tPERT, which is also equipped with a MS2 aptamer (stem loop). The MS2cp protein recruits the tPERT by binding to the MS2 aptamer of the molecule. FIG. 72C depict alternative designs for PEgRNAs that can be achieved through known methods for chemical synthesis of nucleic acid molecules. For example, chemical synthesis can be used to synthesize a hybrid RNA/DNA PEgRNA molecule for use in prime editing, wherein the extension arm of the hybrid PEgRNA is DNA instead of RNA. In such an embodiment, a DNA-dependent DNA polymerase can be used in place of a reverse transcriptase to synthesize the 3′ DNA flap comprising the desired genetic change that is formed by prime editing. In another embodiment, the extension arm can be synthesized to include a chemical linker that prevents the DNA polymerase (e.g., a reverse transcriptase) from using the sgRNA scaffold or backbone as a template. In still another embodiment, the extension arm may comprise a DNA synthesis template that has the reverse orientation relative to the overall orientation of the PEgRNA molecule. For example, and as shown for a PEgRNA in the 5′-to-3′ orientation and with an extension attached to the 3′ end of the sgRNA scaffold, the DNA synthesis template is orientated in the opposite direction, i.e., the 3′-to-5′ direction. This embodiment may be advantageous for PEgRNA embodiments with extension arms positioned at the 3′ end of a gRNA. By reverse the orientation of the extension arm, the DNA synthesis by the polymerase (e.g., reverse transcriptase) will terminate once it reaches the newly orientated 5′ of the extension arm and will thus, not risk using the gRNA core as a template.



FIG. 73 demonstrates prime editing with tPERTs and the MS2 recruitment system (aka MS2 tagging technique). An sgRNA targeting the prime editor protein (PE2) to the target locus is expressed in combination with a tPERT containing a primer binding site (a13-nt or 17-nt PBS), an RT template encoding a His6 tag insertion and a homology arm, and an MS2 aptamer (located at the 5′ or 3′ end of the tPERT molecule). Either prime editor protein (PE2) or a fusion of the MS2cp to the N-terminus of PE2 was used. Editing was carried out with or without a complementary-strand nicking sgRNA, as in the previously developed PE3 system (designated in the x-axis as labels “PE2+nick” or “PE2”, respectively). This is also referred to and defined herein as “second-strand nicking.”



FIG. 74 demonstrates that the MS2 aptamer expression of the reverse transcriptase in trans and its recruitment with the MS2 aptamer system. The PEgRNAPEgRNA contains the MS2 RNA aptamer inserted into either one of two sgRNA scaffold hairpins. The wild-type M-MLV reverse transcriptase is expressed as an N-terminal or C-terminal fusion to the MS2 coat protein (MCP). Editing is at the HEK3 site in HEK293T cells.



FIG. 75 provides a bar graph comparing the efficiency (i.e., “% of total sequencing reads with the specified edit or indels”) of PE2, PE2-trunc, PE3, and PE3-trunc over different target sites in various cell lines. The data shows that the prime editors comprising the truncated RT variants were about as efficient as the prime editors comprising the non-truncated RT proteins.



FIG. 76 demonstrates the editing efficiency of intein-split prime editors. HEK239T cells were transfected with plasmids encoding full-length PE2 or intein-split PE2, PEgRNA and nicking guide RNA. Consensus sequence (most amino-terminal residues of C terminal extein) are indicated. Percent editing at two sites in shown: HEK3+1 CTT insertion and PRNP +6 G to T. Replicate n=3 independent transfections.



FIG. 77 demonstrates the editing efficiency of intein-split prime editors. Editing assessed by targeted deep sequencing in bulk cortex and GFP+ subpopulation upon delivery of 5E10vg per SpPE3 half and a small amount 1E10 of nuclear-localized GFP:KASH to P0 mice by ICV injection. Editors and GFP were packaged in AAV9 with EFS promoter. Mice were harvested three weeks post injection and GFP+ nuclei were isolated by flow cytometry. Individual data points are shown, with 1-2 mice per condition analyzed.



FIG. 78 demonstrates the editing efficiency of intein-split prime editors. Specifically, the figures depicts AAV split-SpPE3 constructs. Co-transduction by AAV particles separately expressing SpPE3-N and SpPE3-C recapitulates PE3 activity. Note N-terminal genome contains a U6-sgRNA cassette expressing the nicking sgRNA, and the C-terminal genome contains a U6-PEgRNA cassette expressing the PEgRNA.



FIG. 79 shows the editing efficiency of certain optimized linkers. In particular, the data shows the editing efficiency of the PE2 construct with the current linker (noted as PE2—white box) compared to various versions with the linker replaced with a sequence as indicated at the HEK3, EMX1, FANCF, RNF2 loci for representative PEgRNAs for transition, transversion, insertion, and deletion edits. The replacement linkers are referred to as ““1×SGGS” (SEQ ID NO: 174), “2×SGGS” (SEQ ID NO: 446), “3× SGGS” (SEQ ID NO: 3889), “1×XTEN” (SEQ ID NO: 171), “no linker”, “1× Gly”, “1× Pro”, “1× EAAAK” (SEQ ID NO: 3968), “2×EAAAK” “(SEQ ID NO: 3969), and “3×EAAAK” (SEQ ID NO: 3970). The editing efficiency is measured in bar graph format relative to the “control” editing efficiency of PE2. The linker of PE2 is SGGSSGGSSGSETPGTSESATPESSGGSSGGSS (SEQ ID NO: 127). All editing was done in the context of the PE3 system, i.e., which refers the PE2 editing construct plus the addition of the optimal secondary sgRNA nicking guide.



FIG. 80. Taking the average fold efficacy relative to PE2 yields the graph shown, indicating that use of a 1×XTEN (SEQ ID NO: 171) linker sequence improves editing efficiency by 1.14 fold on average (n=15).



FIG. 81 depicts the transcription level of PEgRNAs from different promoters.



FIG. 82 Depicts the impact of different types of modifications on PEgRNA structure on editing efficiency relative to unmodified PEgRNA.



FIG. 83 Depicts a PE experiment that targeted editing of the HEK3 gene, specifically targeting the insertion of a 10 nt insertion at position +1 relative to the nick site and using PE3.



FIG. 84A depicts an exemplary PEgRNA having a spacer, gRNA core, and an extension arm (RT template+primer binding site), which is modified at the 3′ end of the PEgRNA with a tRNA molecule, coupled through a UCU linker. The tRNA includes various post-transcriptional modifications. Said modification are not required, however.



FIG. 84B depicts structure of tRNA that can be used to modify PEgRNA structures. The P1 can be variable in length. The P1 can be extended to help prevent RNAseP processing of the PEgRNA-tRNA fusion.



FIG. 85 depicts a PE experiment that targeted editing of the FANCF gene, specifically targeting a G-to-T conversion at position +5 relative to the nick site and using PE3 construct.



FIG. 86 depicts a PE experiment that targeted editing of the HEK3 gene, specifically targeting the insertion of a 71 nt FLAG tag insertion at position +1 relative to the nick site and using PE3 construct.



FIG. 87 results from a screen in N2A cells where the pegRNA installs 1412Adel, with details about the primer binding site (PBS) length and reverse transcriptase (RT) template length. (Shown with and without indels).



FIG. 88 results from a screen in N2A cells where the pegRNA installs 1412Adel, with details about the primer binding site (PBS) length and reverse transcriptase (RT) template length. (Shown with and without indels).



FIG. 89 depicts results of editing at a proxy locus in the β-globin gene and at HEK3 in healthy HSCs, varying the concentration of editor to pegRNA and nicking gRNA.



FIG. 90 provides a schematic of an embodiment of dual-flap prime editing. A DNA target sequence is acted upon by two prime editing complexes (guided by pegRNA-A and pegRNA-B). The two pegRNAs target opposite strands of the double helix. Each prime editor (PE2•pegRNA) nicks a single DNA strand, then synthesizes a 3′ DNA flap using the pegRNA as a template. The action of the two prime editor complexes results in the production of an intermediate containing two 3′ flaps on opposite strands of the DNA. The two 3′ flaps are complementarity to one another at their 3′ ends. Annealing of the 3′ ends of the 3′ flaps results in the formation of a double-duplex structure, with one duplex made of paired 3′ flaps containing the new DNA sequence (red), and the other duplex made of paired 5′ flaps containing the original DNA sequence (black). Excision of the intervening original DNA duplex (black paired 5′ flaps) yields a double-nicked DNA species containing the desired new DNA sequence (red) having replaced the original DNA sequence. Ligation of both nicks completes the editing process.



FIGS. 91A-91B provide results from Example 7. FIG. 91A shows Crispresso2 output allele table (aligned for desired product) for the replacement of a 90-bp sequence with a new 22-bp sequence at the HEK3 site in HEK293T cells using dual prime editors. The desired product accounts for over 80% of the sequencing reads. The reference starting allele is shown above the sequenced alleles, for comparison. FIG. 91B shows Sequences of pegRNAs used to achieve the sequence replacement shown in FIG. 91A. pegRNA1 and pegRNA 2 target different strands of the DNA double helix, and generate 5′ displaced nicks as drawn in FIG. 90.



FIG. 92 shows design embodiments of pegRNA designs for dual-flap prime editing. Two pegRNAs are used for dual-flap prime editing, shown in the drawing as pegRNA A and pegRNA B. Each pegRNA contains a spacer sequence (dark blue) that guides the prime editing complex to the target DNA site. The two pegRNAs target opposite strands of the DNA double helix. Like other pegRNAs, dual-flap prime editing pegRNAs contain a 3′ extension with a primer binding sequence (PBS, green) that anneals to the nicked genomic DNA strand to initiate reverse transcription, and a reverse transcription template (RT template, light blue) that templates the synthesis of new DNA by the reverse transcriptase enzyme. Unlike pegRNAs used for classic prime editing, which require the newly synthesized edited 3′ flap to compete with the endogenous 5′ flap, there is no need to encode homology to the target site within the RT template. Instead, the 3′ ends of the two synthesized 3′ flaps need only contain complementarity to each other (i.e. the 3′ ends of the 3′ flaps are the reverse complement sequences of one another). This complementarity allows the two 3′ flaps to anneal and promote the formation of the desired edited DNA sequence.



FIGS. 93A-93B show the results of Example 7 of using dual prime editing to install Bxb1 attB and attP sites with dual-flap prime editing. FIG. 93A shows the installation of a 38-bp Bxb1 attB site at HEK3. Six pegRNAs were constructed, three targeting the (+) strand (A1, A2 and A3) and three targeting the (−) strand (B1, B2 and B3). These differ in the amounts of attB sequence encoded in the RT template, leading to a different number of complementary nucleotides between the two flaps. The 3×3 matrix of pegRNAs was evaluated for installation of the attB sequence at the target genomic location in HEK293T cells. FIG. 93A and FIG. 93B show the installation of a 50-bp Bxb1 attP site at HEK3. Six pegRNAs were constructed, three targeting the (+) strand (A1, A2 and A3) and three targeting the (−) strand (B1, B2 and B3). These differ in the amounts of attP sequence encoded in the RT template, leading to a different number of complementary nucleotides between the two flaps. The 3×3 matrix of pegRNAs was evaluated for installation of the attP sequence at the target genomic location in HEK293T cells. For both edits in FIG. 93A and FIG. 93B, the installation of the attB or attP site occurs with concomitant deletion of the 90-bp of genomic DNA sequence located between the two nick sites.



FIG. 94 shows the results of installation of Bxb1 attB and attP sites at human safe-harbor loci with dual-flap prime editing. Installation of Bxb1 attP sites at the AAVS1 locus (left) or Bxb1 attB sites at the CCR5 locus (right) in HEK293T cells. Correct edits are shown in blue (AAVS1) and red (CCR5), while indel byproducts are shown in gray.



FIG. 95 provides a schematic of genomic sequence inversion with quadruple-flap prime editing. A region of genomic DNA is targeted for inversion (green and orange segment). Four pegRNAs are delivered to cells with the PE2 prime editor. One pair of pegRNAs targets a single genomic DNA strand and templates the synthesis of two complementary DNA flaps (A and A′, blue), while the second pair targets the other genomic DNA strand and templates the synthesis of two complementary DNA flaps with an orthogonal DNA sequence (B and B′, pink). The complementary flaps anneal to form 3′ overhang duplexes. The 5′ overhang duplexes are excised by endogenous cellular repair enzymes. Nicks are ligated to produce the product allele containing an inverted DNA sequence (green and orange segments) and the pegRNA-templated sequences at the inversion junctions (blue and pink segments).



FIG. 96 shows the results of amplicon sequencing of AAVS1 inversion junctions. CRISPResso2 analysis output of a 2.7-kb inversion at the AAVS1 locus in HEK293T cells using the quadruple-flap primed editing strategy. PCR amplification and sequencing of the expected inversion junctions showed the desired products with Bxb1 attP or attB sequences inserted at the junctions of the inversion.



FIGS. 97A-97B show the results of targeted integration of a circular DNA plasmid into the genome using quadruple-flap prime editing. (FIG. 97A) A region of genomic DNA and a region of plasmid DNA are targeted for quadruple flap prime editing integration. Four pegRNAs are delivered to cells with the PE2 prime editor. Two pegRNAs template complementary sequences, one targeting a single genomic DNA strand, and the other targeting a single plasmid DNA strand (generating blue flaps). The other two pegRNAs target opposite genomic DNA and plasmid DNA strands from those of the first two pegRNAs, and they template the synthesis of two complementary DNA flaps (pink) that are orthogonal to the first pair. The complementary flaps anneal to form 3′ overhang duplexes. The 5′ overhang duplexes are excised by endogenous cellular repair enzymes. Nicks are ligated to produce the product allele containing an integrated plasmid DNA sequence (green and orange segments) and the pegRNA-templated sequences at the integration junctions (blue and pink segments). (FIG. 97B) CRISPResso2 analysis of amplicon sequencing of anticipated junction, showing plasmid backbone and genomic DNA sequence bridged by the pegRNA-templated attP sequence.



FIGS. 98A-98B show the results of targeted chromosomal translocation with quadruple-flap prime editing. (FIG. 98A) pegRNAs target two regions on different chromosomes. Complementary 3′ DNA flaps bridge the two chromosome sequences and direct the orientation of the translocation. (FIG. 98B) Targeted translocation between MYC and TIMM44 loci in HEK239T cells. CRISPResso2 analysis output from amplicon sequencing of the expected junctions from translocation between the MYC locus on chromosome 8 and the TIMM44 locus on chromosome 19. The majority of sequencing reads correspond to the desired allele sequence.



FIG. 99 shows installation of Bxb1 attB and attP sites with dual-flap prime editing at IDS locus. HEK293T cells were transfected with PE2 and different pairs of pegRNAs (e.g. in the first column pegRNA A1_a and pegRNA B2_a with templates for installing attP site in the forward direction). The efficiency was measured by HTS. This data shows that dual-flap editing can successfully insert the sequence of interest to the IDS locus with an efficiency up to ˜80%.



FIGS. 100A-100B describe dual-flap-mediated duplication. FIG. 100A is a schematic showing dual-flap-mediated duplication at AAVS1 locus in 293T cells. FIG. 100B shows results of using dual-flap pegRNA with PE2 for inducing duplication of genetic sequences at AAVS1.



FIG. 101 shows multi-flap induced new translocation MYC-CCR5. MYC-CCR5 translocation was induced by quad-flap pegRNAs and PE2. MYC-CCR5 translocation events were induced by quadruple-pegRNAs. Four different sets of pegRNAs were tested in HEK293T cells. The translocation junction products between derived chr8 and chr3 were amplified by junction primers. The % of reads aligned to the expected junction alleles are shown in the graph. The result shows that quadruple-flap can mediate translocation of MYC and CCR5 gene with product purity near 100% at junction 1 and ˜50% at junction 2. A representative allele plot shows the sequences aligned to the expected allele sequences at junction 1.



FIGS. 102A-102B show dual-flap and multi-flap editing in other human cell lines. FIG. 102A shows dual-flap editing in four different human cell lines. HEK293T and HeLa cells were transfected with dual pegRNAs and PE2 for editing three different genomic loci (IDS, MYC, and TIMM44). U2OS and K562 cells were nucleofected with the same components. Dual-flap has shown robust editing across all four human cells at the targeted loci, particularly at HEK293T and K562 cells. The cellular mechanism for enabling dual-flap editing are conserved across many human cell types. FIG. 102B shows multi-flap (quadruple-pegRNA) directed 2.7 kb inversion at the AAVS1 in HeLa cells.



FIG. 103A-103B shows results of inversion efficiency measurement by HTS at CCR5 locus. Percentage of the expected inversion edit allele was measured by HTS. Four quad-pegRNA sets PE2 were transfected in HEK293T cells, respectively. FIG. 103A shows dual-flap mediated sequence duplication (˜100 nt) at CCR5 locus in HEK293T cells. The editing efficiency achieves ˜1.5% via HTS 300-cycle pair-end sequencing analysis. FIG. 103B shows quadruple-flap-mediated sequence inversion (˜95-117 nt) at CCR5 locus in HeLa cells. The editing efficiency achieves ˜1.2% via HTS 300-cycle pair-end sequencing analysis. This result shows that multi-flap can successfully mediate duplication and inversion at CCR5 locus precisely. The editing specificity is high when the targeted sequence is duplicated (percentage of indels<2%).



FIGS. 104A-104E show targeted cellular repair pathway for dual-flap editing. In FIGS. 104A-104D, HEK293T cells were transfected with the plasmids that express Exo1, Fen1, Red Fluorescence Protein (control), DNA2, Mlh1 neg, and P53 inhibitor with pegRNA and PE2. The editing efficiency was measured by HTS. The editing efficiency was compared between the candidate and the RFP control. In FIG. 104E, HEK293T cells were transfected with the siRNA plasmids and pegRNA and PE2 for each target locus. Non-targeting siRNA (siNT) was used as the control. The editing efficiency was measured by HTS. The editing efficiency was compared between each siRNA knockdown and the siNT ctrl at each target locus. HEK293T cells were transfected with dual-pegRNAs, PE2, and the plasmids that express Exo1, Fen1, Red Fluorescence Protein (ctrl), DNA2, Mlh1 neg, and P53 inhibitor respectively. The editing efficiency was measured by HTS. The editing efficiency was compared between the candidate and the RFP ctrl. Two-tailed, paired Student t-test was used to measure statistical difference between each treatment and RFP control (P<0.05, *; P<0.01, **; P<0.001, ***). Overexpression of FEN1 improves dual flap editing efficiency in all four targeted loci (MYC, TIMM44, IDS, CCR5).



FIGS. 105A-105B show dual-flap-mediated sequence duplication at AAVS1 locus. FIG. 105A shows a schematic diagram of dual-flap-mediated sequence duplication at AAVS1 locus. FIG. 105B shows that by using dual pegRNAs that generate two unique 3′ flap structures, a ˜300 bp sequence duplication was induced at AAVS1 locus in 293T cells. Expected alleles are amplified with specific primers and are subjected for HTS. ˜94% reads are aligned to the expected alleles with duplication. Duplication products are not observed in the untreated samples.



FIG. 106 shows targeted IDS genomic sequence inversion with quadruple-flap prime editing. ˜13% of Hunter syndrome patients have been shown to have an inversion of the IDS gene sequences (Bondeson et al., Human Molecular Genetics, 1995). Quadruple-flap prime editing was applied to induce this pathogenic inversion of the ˜40 kb IDS genomic sequence in the HEK293T cells. Six sets of quadruple pegRNAs were tested by transfecting HEK293T cells with the pegRNAs and PE2. Primers were used to specifically amplify the inverted sequences at junction “ab” and junction “cd”. ˜95% of the expected inverted allele sequences were observed at both junctions with IDS_QF1. Other sets of pegRNAs also yield high percentage of the expected allele sequences at both junctions. Inverted junction products are not observed in the untreated samples.



FIG. 107 shows that PegRNA 3′ motif modification improves dual-flap editing efficiency at IDS locus. To further improve the dual flap editing efficiency, a pseudoknot evoPreQ1 motif was introduced to protect the pegRNA 3′ end. By comparing the editing efficiency generated by the unmodified and evoPreQ1-modified dual pegRNAs, there is an overall increase of the editing efficiency with modified pegRNAs at the targeted IDS locus. The improvement of dual-flap editing efficiency can reach up to 5.3-fold.



FIGS. 108A-108C show an overview of twinPE and twinPE-mediated sequence replacement. FIG. 108A shows that twinPE systems target genomic DNA sequences that contain two protospacer sequences on opposite strands of DNA. PE2•pegRNA complexes target each protospacer, generate a single-stranded nick, and reverse transcribe the pegRNA-encoded template containing the desired insertion sequence. After synthesis and release of the 3′ DNA flaps, a hypothetical intermediate exists possessing annealed 3′ flaps containing the edited DNA sequence and annealed 5′ flaps containing the original DNA sequence. Excision of the original DNA sequence contained in the 5′ flap, follow by ligation of the 3′ flaps to the corresponding excision site, generates the desired edited product. FIG. 108B shows an example of twinPE-mediated replacement of a 90-bp sequence in HEK site 3 with a 38-bp Bxb1 attB sequence. FIG. 108C shows an evaluation of twinPE in HEK293T cells for the installation of the 38-bp Bxb1 attB site as shown in FIG. 108B or the 50-bp Bxb1 attP site at HEK site 3 using pegRNAs that template varying lengths of the insertion sequence. Values and error bars reflect the mean and s.d. of three independent biological replicates.



FIGS. 109A-109E show targeted sequence insertion, deletion, and recoding with twinPE in human cells. FIG. 109A shows insertion of FKBP coding sequence fragments with PE3 (12 bp, 36 bp, 108 bp, or 321 bp) or twinPE (108 bp) at HEK site 3 in HEK293T cells. FIG. 109B shows recoding of sequence within exons 4 and 7 in PAH in HEK293T cells using twinPE. A 64-bp target sequence in exon 4 was edited using 24, 36, or 59 bp of overlapping flaps, a 46-bp target sequence in exon 7 was edited using 22 or 42 bp of overlapping flaps, or a 64-bp sequence in exon 7 was edited using 24 or 47 bp of overlapping flaps. Editing activity was compared using standard pegRNAs or epegRNAs containing 3′ evoPreQ1 motifs. FIG. 109C is a schematic diagram of three distinct dual-flap deletion strategies that were investigated for carrying out targeted deletions. The “Basic-Anchor (BA)” twinPE strategy allows for flexible deletion starting at an arbitrary position 3′ of one nick site and ending at the other nick site. The “Hybrid-Anchor (HA)” twinPE strategy allows for flexible deletion of sequence at arbitrarily chosen positions between the two nick sites. The “PrimeDel (PD)” strategy tested here allows for deletion of the sequence starting at one nick site and ending at another nick site. FIG. 109D shows deletion of sequences at HEK site 3 in HEK293T cells using the BA-twinPE, HA-twinPE, or PD strategies targeting the same protospacer pair. Editing activity was compared using standard pegRNAs or epegRNAs containing 3′ evoPreQ1 motifs. FIG. 109E shows deletion of exon 51 sequence at the DMD locus in HEK293T cells using BA-twinPE, PD, paired Cas9 nuclease, or twinPE-mediated attB sequence replacement. Values and error bars reflect the mean and s.d. of three independent biological replicates. In the DMD exon 51 skipping experiment, at least two independent biological replicates were performed.



FIGS. 110A-110E show site-specific genomic integration of DNA cargo with twinPE and Bxb1 recombinase in human cells. FIG. 110A shows screening of twinPE pegRNA pairs for installation of the Bxb1 attP sequence at the AAVS1 locus in HEK293T cells. FIG. 110B shows screening of twinPE pegRNA pairs for installation of the Bxb1 attB sequence at the CCR5 locus in HEK293T cells. FIG. 110C shows single transfection knock-in of 5.6-kb DNA donors using twinPE pegRNA pairs targeting CCR5 (four left-most bars) or AAVS1 (three right-most bars). The twinPE pegRNAs install attB at CCR5 or attP at AA VS/. Bxb1 then integrates a donor bearing the corresponding attachment site into the genomic attachment site. FIG. 110D shows optimization of single transfection knock-in at CCR5 using the 531/584 twinPE pegRNA pair. Identity of the templated edit (attB vs. attP), identity of the central dinucleotide (wild-type GT vs. orthogonal mutant GA), and length of the overlap between flaps were varied to identify the combination that supported the highest knock-in efficiency. FIG. 110E shows insertion of the Bxb1 attB sequence within intron 1 of ALB in HEK293T and Huh7 cells lines. FIG. 110F shows a comparison of single transfection knock-in efficiencies at CCR5 and ALB in HEK293T and Huh7 cell lines.



FIGS. 111A-111E show site-specific genomic sequence inversion with twinPE and Bxb1 recombinase in human cells. FIG. 111A is a schematic diagram of recombination hot spots in IDS and IDS2 that lead to pathogenic 39-kb inversions, and the combined twinPE-Bxb1 strategy for installing or correcting the IDS inversion mutation. FIG. 111B shows a screen of pegRNA pairs at IDS and IDS2 for installation of attP or attB recombination site insertion at IDS and IDS2 loci with specific DNA targets. FIG. 111C shows a DNA sequencing analysis of the attP or attB insertion with sequential DNA transfection. FIG. 111D shows inversion product purity at the inverted junction 1 and junction 2 (sequential transfection), indicating the successful inversion at the two junctions. FIG. 111E shows the quantification of inversion efficiency at the junctions (sequential transfection and “one-pot” RNA nucleofection).



FIG. 112 shows the recoding of sequences within exon 10, 11, and 12 in PAH in HEK293T cells via twinPE. A 64-bp target sequence in exon 10 was edited using 28 bp of overlapping flaps, a 61-bp and 55-bp target sequence in exon 11 was edited using 25 bp of overlapping flaps, or a 68-bp and 58-bp sequence in exon 12 was edited using 27 and 24 bp of overlapping flaps, respectively. Values and error bars reflect the mean and s.d. of three independent biological replicates



FIG. 113 shows transfection of HEK293T clonal cell line containing homozygous attB site insertion with BxBI plasmids and attP-containing donor DNA plasmids. The knock-in efficiency is between 12-17% at the target site as measured by ddPCR.



FIG. 114 shows HTS measurement of expected junction sequences containing attL and attR recombination products after twinPE and BxBI-mediated one-pot knock-in. The product purities range from 71-95%. Values and error bars reflect the mean and s.d. of three independent biological replicates



FIG. 115A shows twinPE mediated attB insertion efficiency with reduced flap overlap length in the dual pegRNAs.



FIG. 115B shows PCR products amplified by specific primer for capturing the recombination between donor DNA and pegRNA plasmids shown on the agarose gel. Recombination between donor DNA and pegRNA plasmid was reduced with smaller flap overlap.



FIG. 116 is a schematic diagram of the developed PCR strategies for quantifying IDS inversion efficiency.





DEFINITIONS

Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. The following references provide one of skill with a general definition of many of the terms used in this invention: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd ed. 1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th Ed., R. Rieger et al. (eds.), Springer Verlag (1991); and Hale & Marham, The Harper Collins Dictionary of Biology (1991). As used herein, the following terms have the meanings ascribed to them unless specified otherwise.


Antisense Strand


In genetics, the “antisense” strand of a segment within double-stranded DNA is the template strand, and which is considered to run in the 3′ to 5′ orientation. By contrast, the “sense” strand is the segment within double-stranded DNA that runs from 5′ to 3′, and which is complementary to the antisense strand of DNA, or template strand, which runs from 3′ to 5′. In the case of a DNA segment that encodes a protein, the sense strand is the strand of DNA that has the same sequence as the mRNA, which takes the antisense strand as its template during transcription, and eventually undergoes (typically, not always) translation into a protein. The antisense strand is thus responsible for the RNA that is later translated to protein, while the sense strand possesses a nearly identical makeup to that of the mRNA. Note that for each segment of dsDNA, there will possibly be two sets of sense and antisense, depending on which direction one reads (since sense and antisense is relative to perspective). It is ultimately the gene product, or mRNA, that dictates which strand of one segment of dsDNA is referred to as sense or antisense.


Bi-Specific Ligand


The term “bi-specific ligand” or “bi-specific moiety,” as used herein, refers to a ligand that binds to two different ligand-binding domains. In certain embodiments, the ligand is a small molecule compound, or a peptide, or a polypeptide. In other embodiments, ligand-binding domain is a “dimerization domain,” which can be install as a peptide tag onto a protein. In various embodiments, two proteins each comprising the same or different dimerization domains can be induced to dimerize through the binding of each dimerization domain to the bi-specific ligand. As used herein, “bi-specific ligands” may be equivalently refer to “chemical inducers of dimerization” or “CIDs”.


Cas9


The term “Cas9” or “Cas9 nuclease” refers to an RNA-guided nuclease comprising a Cas9 domain, or a fragment thereof (e.g., a protein comprising an active or inactive DNA cleavage domain of Cas9, and/or the gRNA binding domain of Cas9). A “Cas9 domain” as used herein, is a protein fragment comprising an active or inactive cleavage domain of Cas9 and/or the gRNA binding domain of Cas9. A “Cas9 protein” is a full length Cas9 protein. A Cas9 nuclease is also referred to sometimes as a casn1 nuclease or a CRISPR (Clustered Regularly Interspaced Short Palindromic Repeat)-associated nuclease. CRISPR is an adaptive immune system that provides protection against mobile genetic elements (viruses, transposable elements, and conjugative plasmids). CRISPR clusters contain spacers, sequences complementary to antecedent mobile elements, and target invading nucleic acids. CRISPR clusters are transcribed and processed into CRISPR RNA (crRNA). In type II CRISPR systems correct processing of pre-crRNA requires a trans-encoded small RNA (tracrRNA), endogenous ribonuclease 3 (rnc) and a Cas9 domain. The tracrRNA serves as a guide for ribonuclease 3-aided processing of pre-crRNA. Subsequently, Cas9/crRNA/tracrRNA endonucleolytically cleaves a linear or circular dsDNA target complementary to the spacer. The target strand not complementary to crRNA is first cut endonucleolytically, then trimmed 3′-5′ exonucleolytically. In nature, DNA-binding and cleavage typically requires protein and both RNAs. However, single guide RNAs (“sgRNA”, or simply “gNRA”) can be engineered to incorporate aspects of both the crRNA and tracrRNA into a single RNA species. See, e.g., Jinek M., Chylinski K., Fonfara I., Hauer M., Doudna J. A., Charpentier E. Science 337:816-821(2012), the entire contents of which are hereby incorporated by reference. Cas9 recognizes a short motif in the CRISPR repeat sequences (the PAM or protospacer adjacent motif) to help distinguish self versus non-self. Cas9 nuclease sequences and structures are well known to those of skill in the art (see, e.g., “Complete genome sequence of an M1 strain of Streptococcus pyogenes.” Ferretti et al., J. J., McShan W. M., Ajdic D. J., Savic D. J., Savic G., Lyon K., Primeaux C., Sezate S., Suvorov A. N., Kenton S., Lai H. S., Lin S. P., Qian Y., Jia H. G., Najar F. Z., Ren Q., Zhu H., Song L., White J., Yuan X., Clifton S. W., Roe B. A., McLaughlin R. E., Proc. Natl. Acad. Sci. U.S.A. 98:4658-4663(2001); “CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III.” Deltcheva E., Chylinski K., Sharma C. M., Gonzales K., Chao Y., Pirzada Z. A., Eckert M. R., Vogel J., Charpentier E., Nature 471:602-607(2011); and “A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity.” Jinek M., Chylinski K., Fonfara I., Hauer M., Doudna J. A., Charpentier E. Science 337:816-821(2012), the entire contents of each of which are incorporated herein by reference). Cas9 orthologs have been described in various species, including, but not limited to, S. pyogenes and S. thermophilus. Additional suitable Cas9 nucleases and sequences will be apparent to those of skill in the art based on this disclosure, and such Cas9 nucleases and sequences include Cas9 sequences from the organisms and loci disclosed in Chylinski, Rhun, and Charpentier, “The tracrRNA and Cas9 families of type II CRISPR-Cas immunity systems” (2013) RNA Biology 10:5, 726-737; the entire contents of which are incorporated herein by reference. In some embodiments, a Cas9 nuclease comprises one or more mutations that partially impair or inactivate the DNA cleavage domain.


A nuclease-inactivated Cas9 domain may interchangeably be referred to as a “dCas9” protein (for nuclease-“dead” Cas9). Methods for generating a Cas9 domain (or a fragment thereof) having an inactive DNA cleavage domain are known (see, e.g., Jinek et al., Science. 337:816-821(2012); Qi et al., “Repurposing CRISPR as an RNA-Guided Platform for Sequence-Specific Control of Gene Expression” (2013) Cell. 28; 152(5):1173-83, the entire contents of each of which are incorporated herein by reference). For example, the DNA cleavage domain of Cas9 is known to include two subdomains, the HNH nuclease subdomain and the RuvC1 subdomain. The HNH subdomain cleaves the strand complementary to the gRNA, whereas the RuvC1 subdomain cleaves the non-complementary strand. Mutations within these subdomains can silence the nuclease activity of Cas9. For example, the mutations D10A and H840A completely inactivate the nuclease activity of S. pyogenes Cas9 (Jinek et al., Science. 337:816-821(2012); Qi et al., Cell. 28; 152(5):1173-83 (2013)). In some embodiments, proteins comprising fragments of Cas9 are provided. For example, in some embodiments, a protein comprises one of two Cas9 domains: (1) the gRNA binding domain of Cas9; or (2) the DNA cleavage domain of Cas9. In some embodiments, proteins comprising Cas9 or fragments thereof are referred to as “Cas9 variants.” A Cas9 variant shares homology to Cas9, or a fragment thereof. For example, a Cas9 variant is at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, at least about 99.8% identical, or at least about 99.9% identical to wild type Cas9 (e.g., SpCas9 of SEQ ID NO: 18). In some embodiments, the Cas9 variant may have 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 21, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or more amino acid changes compared to wild type Cas9 (e.g., SpCas9 of SEQ ID NO: 18). In some embodiments, the Cas9 variant comprises a fragment of SEQ ID NO: 18 Cas9 (e.g., a gRNA binding domain or a DNA-cleavage domain), such that the fragment is at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to the corresponding fragment of wild type Cas9 (e.g., SpCas9 of SEQ ID NO: 18). In some embodiments, the fragment is at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% identical, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% of the amino acid length of a corresponding wild type Cas9 (e.g., SpCas9 of SEQ ID NO: 18).


cDNA


The term “cDNA” refers to a strand of DNA copied from an RNA template. cDNA is complementary to the RNA template.


Circular Permutant


As used herein, the term “circular permutant” refers to a protein or polypeptide (e.g., a Cas9) comprising a circular permutation, which is a change in the protein's structural configuration involving a change in the order of amino acids appearing in the protein's amino acid sequence. In other words, circular permutants are proteins that have altered N- and C-termini as compared to a wild-type counterpart, e.g., the wild-type C-terminal half of a protein becomes the new N-terminal half. Circular permutation (or CP) is essentially the topological rearrangement of a protein's primary sequence, connecting its N- and C-terminus, often with a peptide linker, while concurrently splitting its sequence at a different position to create new, adjacent N- and C-termini. The result is a protein structure with different connectivity, but which often can have the same overall similar three-dimensional (3D) shape, and possibly include improved or altered characteristics, including, reduced proteolytic susceptibility, improved catalytic activity, altered substrate or ligand binding, and/or improved thermostability. Circular permutant proteins can occur in nature (e.g., concanavalin A and lectin). In addition, circular permutation can occur as a result of posttranslational modifications or may be engineered using recombinant techniques.


Circularly Permuted Cas9


The term “circularly permuted Cas9” refers to any Cas9 protein, or variant thereof, that has been occurs as a circular permutant, whereby its N- and C-termini have been topically rearranged. Such circularly permuted Cas9 proteins (“CP-Cas9”), or variants thereof, retain the ability to bind DNA when complexed with a guide RNA (gRNA). See, Oakes et al., “Protein Engineering of Cas9 for enhanced function,” Methods Enzymol, 2014, 546: 491-511 and Oakes et al., “CRISPR-Cas9 Circular Permutants as Programmable Scaffolds for Genome Modification,” Cell, Jan. 10, 2019, 176: 254-267, each of which are incorporated herein by reference. The instant disclosure contemplates any previously known CP-Cas9 or use of a new CP-Cas9 so long as the resulting circularly permuted protein retains the ability to bind DNA when complexed with a guide RNA (gRNA). Exemplary CP-Cas9 proteins are SEQ ID NOs: 77-86.


CRISPR


CRISPR is a family of DNA sequences (i.e., CRISPR clusters) in bacteria and archaea that represent snippets of prior infections by a virus that have invaded the prokaryote. The snippets of DNA are used by the prokaryotic cell to detect and destroy DNA from subsequent attacks by similar viruses and effectively compose, along with an array of CRISPR-associated proteins (including Cas9 and homologs thereof) and CRISPR-associated RNA, a prokaryotic immune defense system. In nature, CRISPR clusters are transcribed and processed into CRISPR RNA (crRNA). In certain types of CRISPR systems (e.g., type II CRISPR systems), correct processing of pre-crRNA requires a trans-encoded small RNA (tracrRNA), endogenous ribonuclease 3 (mc) and a Cas9 protein. The tracrRNA serves as a guide for ribonuclease 3-aided processing of pre-crRNA. Subsequently, Cas9/crRNA/tracrRNA endonucleolytically cleaves a linear or circular dsDNA target complementary to the RNA. Specifically, the target strand not complementary to crRNA is first cut endonucleolytically, then trimmed 3′-5′ exonucleolytically. In nature, DNA-binding and cleavage typically requires protein and both RNAs. However, single guide RNAs (“sgRNA”, or simply “gNRA”) can be engineered so as to incorporate aspects of both the crRNA and tracrRNA into a single RNA species—the guide RNA. See, e.g., Jinek M., Chylinski K., Fonfara I., Hauer M., Doudna J. A., Charpentier E. Science 337:816-821(2012), the entire contents of which is hereby incorporated by reference. Cas9 recognizes a short motif in the CRISPR repeat sequences (the PAM or protospacer adjacent motif) to help distinguish self versus non-self. CRISPR biology, as well as Cas9 nuclease sequences and structures are well known to those of skill in the art (see, e.g., “Complete genome sequence of an M1 strain of Streptococcus pyogenes.” Ferretti et al., J. J., McShan W. M., Ajdic D. J., Savic D. J., Savic G., Lyon K., Primeaux C., Sezate S., Suvorov A. N., Kenton S., Lai H. S., Lin S. P., Qian Y., Jia H. G., Najar F. Z., Ren Q., Zhu H., Song L., White J., Yuan X., Clifton S. W., Roe B. A., McLaughlin R. E., Proc. Natl. Acad. Sci. U.S.A. 98:4658-4663(2001); “CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III.” Deltcheva E., Chylinski K., Sharma C. M., Gonzales K., Chao Y., Pirzada Z. A., Eckert M. R., Vogel J., Charpentier E., Nature 471:602-607(2011); and “A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity.” Jinek M., Chylinski K., Fonfara I., Hauer M., Doudna J. A., Charpentier E. Science 337:816-821(2012), the entire contents of each of which are incorporated herein by reference). Cas9 orthologs have been described in various species, including, but not limited to, S. pyogenes and S. thermophilus. Additional suitable Cas9 nucleases and sequences will be apparent to those of skill in the art based on this disclosure, and such Cas9 nucleases and sequences include Cas9 sequences from the organisms and loci disclosed in Chylinski, Rhun, and Charpentier, “The tracrRNA and Cas9 families of type II CRISPR-Cas immunity systems” (2013) RNA Biology 10:5, 726-737; the entire contents of which are incorporated herein by reference.


In certain types of CRISPR systems (e.g., type II CRISPR systems), correct processing of pre-crRNA requires a trans-encoded small RNA (tracrRNA), endogenous ribonuclease 3 (mc), and a Cas9 protein. The tracrRNA serves as a guide for ribonuclease 3-aided processing of pre-crRNA. Subsequently, Cas9/crRNA/tracrRNA endonucleolytically cleaves a linear or circular nucleic acid target complementary to the RNA. Specifically, the target strand not complementary to crRNA is first cut endonucleolytically, then trimmed 3′-5′ exonucleolytically. In nature, DNA-binding and cleavage typically requires protein and both RNAs. However, single guide RNAs (“sgRNA”, or simply “gRNA”) can be engineered to incorporate embodiments of both the crRNA and tracrRNA into a single RNA species—the guide RNA.


In general, a “CRISPR system” refers collectively to transcripts and other elements involved in the expression of or directing the activity of CRISPR-associated (“Cas”) genes, including sequences encoding a Cas gene, a tracr (trans-activating CRISPR) sequence (e.g. tracrRNA or an active partial tracrRNA), a tracr mate sequence (encompassing a “direct repeat” and a tracrRNA-processed partial direct repeat in the context of an endogenous CRISPR system), a guide sequence (also referred to as a “spacer” in the context of an endogenous CRISPR system), or other sequences and transcripts from a CRISPR locus. The tracrRNA of the system is complementary (fully or partially) to the tracr mate sequence present on the guide RNA.


DNA Synthesis Template


As used herein, the term “DNA synthesis template” refers to the region or portion of the extension arm of a PEgRNA that is utilized as a template strand by a polymerase of a prime editor to encode a 3′ single-strand DNA flap that contains the desired edit and which then, through the mechanism of prime editing, replaces the corresponding endogenous strand of DNA at the target site. In various embodiments, the DNA synthesis template is shown in FIG. 3A (in the context of a PEgRNA comprising a 5′ extension arm), FIG. 3B (in the context of a PEgRNA comprising a 3′ extension arm), FIG. 3C (in the context of an internal extension arm), FIG. 3D (in the context of a 3′ extension arm), and FIG. 3E (in the context of a 5′ extension arm). The extension arm, including the DNA synthesis template, may be comprised of DNA or RNA. In the case of RNA, the polymerase of the prime editor can be an RNA-dependent DNA polymerase (e.g., a reverse transcriptase). In the case of DNA, the polymerase of the prime editor can be a DNA-dependent DNA polymerase. In various embodiments (e.g., as depicted in FIGS. 3D-3E), the DNA synthesis template (4) may comprise the “edit template” and the “homology arm”, and all or a portion of the optional 5′ end modifier region, e2. That is, depending on the nature of the e2 region (e.g., whether it includes a hairpin, toeloop, or stem/loop secondary structure), the polymerase may encode none, some, or all of the e2 region, as well. Said another way, in the case of a 3′ extension arm, the DNA synthesis template (3) can include the portion of the extension arm (3) that spans from the 5′ end of the primer binding site (PBS) to 3′ end of the gRNA core that may operate as a template for the synthesis of a single-strand of DNA by a polymerase (e.g., a reverse transcriptase). In the case of a 5′ extension arm, the DNA synthesis template (3) can include the portion of the extension arm (3) that spans from the 5′ end of the PEgRNA molecule to the 3′ end of the edit template. Preferably, the DNA synthesis template excludes the primer binding site (PBS) of PEgRNAs either having a 3′ extension arm or a 5′ extension arm. Certain embodiments described here (e.g, FIG. 71A) refer to an “an RT template,” which is inclusive of the edit template and the homology arm, i.e., the sequence of the PEgRNA extension arm which is actually used as a template during DNA synthesis. The term “RT template” is equivalent to the term “DNA synthesis template.”


In the case of trans prime editing (e.g., FIG. 3G and FIG. 3H), the primer binding site (PBS) and the DNA synthesis template can be engineered into a separate molecule referred to as a trans prime editor RNA template (tPERT).


Dimerization Domain


The term “dimerization domain” refers to a ligand-binding domain that binds to a binding moiety of a bi-specific ligand. A “first” dimerization domain binds to a first binding moiety of a bi-specific ligand and a “second” dimerization domain binds to a second binding moiety of the same bi-specific ligand. When the first dimerization domain is fused to a first protein (e.g., via PE, as discussed herein) and the second dimerization domain (e.g., via PE, as discussed herein) is fused to a second protein, the first and second protein dimerize in the presence of a bi-specific ligand, wherein the bi-specific ligand has at least one moiety that binds to the first dimerization domain and at least another moiety that binds to the second dimerization domain.


Downstream


As used herein, the terms “upstream” and “downstream” are terms of relativity that define the linear position of at least two elements located in a nucleic acid molecule (whether single or double-stranded) that is orientated in a 5′-to-3′ direction. In particular, a first element is upstream of a second element in a nucleic acid molecule where the first element is positioned somewhere that is 5′ to the second element. For example, a SNP is upstream of a Cas9-induced nick site if the SNP is on the 5′ side of the nick site. Conversely, a first element is downstream of a second element in a nucleic acid molecule where the first element is positioned somewhere that is 3′ to the second element. For example, a SNP is downstream of a Cas9-induced nick site if the SNP is on the 3′ side of the nick site. The nucleic acid molecule can be a DNA (double or single stranded). RNA (double or single stranded), or a hybrid of DNA and RNA. The analysis is the same for single strand nucleic acid molecule and a double strand molecule since the terms upstream and downstream are in reference to only a single strand of a nucleic acid molecule, except that one needs to select which strand of the double stranded molecule is being considered. Often, the strand of a double stranded DNA which can be used to determine the positional relativity of at least two elements is the “sense” or “coding” strand. In genetics, a “sense” strand is the segment within double-stranded DNA that runs from 5′ to 3′, and which is complementary to the antisense strand of DNA, or template strand, which runs from 3′ to 5′. Thus, as an example, a SNP nucleobase is “downstream” of a promoter sequence in a genomic DNA (which is double-stranded) if the SNP nucleobase is on the 3′ side of the promoter on the sense or coding strand.


Edit Template


The term “edit template” refers to a portion of the extension arm that encodes the desired edit in the single strand 3′ DNA flap that is synthesized by the polymerase, e.g., a DNA-dependent DNA polymerase, RNA-dependent DNA polymerase (e.g., a reverse transcriptase). Certain embodiments described here (e.g., FIG. 71A) refer to “an RT template,” which refers to both the edit template and the homology arm together, i.e., the sequence of the PEgRNA extension arm which is actually used as a template during DNA synthesis. The term “RT edit template” is also equivalent to the term “DNA synthesis template,” but wherein the RT edit template reflects the use of a prime editor having a polymerase that is a reverse transcriptase, and wherein the DNA synthesis template reflects more broadly the use of a prime editor having any polymerase.


Effective Amount


The term “effective amount,” as used herein, refers to an amount of a biologically active agent that is sufficient to elicit a desired biological response. For example, in some embodiments, an effective amount of a prime editor (PE) may refer to the amount of the editor that is sufficient to edit a target site nucleotide sequence, e.g., a genome. In some embodiments, an effective amount of a prime editor (PE) provided herein, e.g., of a fusion protein comprising a nickase Cas9 domain and a reverse transcriptase may refer to the amount of the fusion protein that is sufficient to induce editing of a target site specifically bound and edited by the fusion protein. As will be appreciated by the skilled artisan, the effective amount of an agent, e.g., a fusion protein, a nuclease, a hybrid protein, a protein dimer, a complex of a protein (or protein dimer) and a polynucleotide, or a polynucleotide, may vary depending on various factors as, for example, on the desired biological response, e.g., on the specific allele, genome, or target site to be edited, on the cell or tissue being targeted, and on the agent being used.


Error-Prone Reverse Transcriptase


As used herein, the term “error-prone” reverse transcriptase (or more broadly, any polymerase) refers to a reverse transcriptase (or more broadly, any polymerase) that occurs naturally or which has been derived from another reverse transcriptase (e.g., a wild type M-MLV reverse transcriptase) which has an error rate that is less than the error rate of wild type M-MLV reverse transcriptase. The error rate of wild type M-MLV reverse transcriptase is reported to be in the range of one error in 15,000 (higher) to 27,000 (lower). An error rate of 1 in 15,000 corresponds with an error rate of 6.7×10−5. An error rate of 1 in 27,000 corresponds with an error rate of 3.7×105. See Boutabout et al. (2001) “DNA synthesis fidelity by the reverse transcriptase of the yeast retrotransposon Ty1,” Nucleic Acids Res 29(11):2217-2222, which is incorporated herein by reference. Thus, for purposes of this application, the term “error prone” refers to those RT that have an error rate that is greater than one error in 15,000 nucleobase incorporation (6.7×10−5 or higher), e.g., 1 error in 14,000 nucleobases (7.14×10−5 or higher), 1 error in 13,000 nucleobases or fewer (7.7×10−5 or higher), 1 error in 12,000 nucleobases or fewer (7.7×10−5 or higher), 1 error in 11,000 nucleobases or fewer (9.1×10−5 or higher), 1 error in 10,000 nucleobases or fewer (1×10 or 0.0001 or higher), 1 error in 9,000 nucleobases or fewer (0.00011 or higher), 1 error in 8,000 nucleobases or fewer (0.00013 or higher) 1 error in 7,000 nucleobases or fewer (0.00014 or higher), 1 error in 6,000 nucleobases or fewer (0.00016 or higher), 1 error in 5,000 nucleobases or fewer (0.0002 or higher), 1 error in 4,000 nucleobases or fewer (0.00025 or higher), 1 error in 3,000 nucleobases or fewer (0.00033 or higher), 1 error in 2,000 nucleobase or fewer (0.00050 or higher), or 1 error in 1,000 nucleobases or fewer (0.001 or higher), or 1 error in 500 nucleobases or fewer (0.002 or higher), or 1 error in 250 nucleobases or fewer (0.004 or higher).


Extein


The term “extein,” as used herein, refers to an polypeptide sequence that is flanked by an intein and is ligated to another extein during the process of protein splicing to form a mature, spliced protein. Typically, an intein is flanked by two extein sequences that are ligated together when the intein catalyzes its own excision. Exteins, accordingly, are the protein analog to exons found in mRNA. For example, a polypeptide comprising an intein may be of the structure extein(N)-intein-extein(C). After excision of the intein and splicing of the two exteins, the resulting structures are extein(N)-extein(C) and a free intein. In various configurations, the exteins may be separate proteins (e.g., half of a Cas9 or PE fusion protein), each fused to a split-intein, wherein the excision of the split inteins causes the splicing together of the extein sequences.


Extension Arm


The term “extension arm” refers to a nucleotide sequence component of a PEgRNA which provides several functions, including a primer binding site and an edit template for reverse transcriptase. In some embodiments, e.g., FIG. 3D, the extension arm is located at the 3′ end of the guide RNA. In other embodiments, e.g., FIG. 3E, the extension arm is located at the 5′ end of the guide RNA. In some embodiments, the extension arm also includes a homology arm. In various embodiments, the extension arm comprises the following components in a 5′ to 3′ direction: the homology arm, the edit template, and the primer binding site. Since polymerization activity of the reverse transcriptase is in the 5′ to 3′ direction, the preferred arrangement of the homology arm, edit template, and primer binding site is in the 5′ to 3′ direction such that the reverse transcriptase, once primed by an annealed primer sequence, polymerizes a single strand of DNA using the edit template as a complementary template strand. Further details, such as the length of the extension arm, are described elsewhere herein.


The extension arm may also be described as comprising generally two regions: a primer binding site (PBS) and a DNA synthesis template, as shown in FIG. 3G (top), for instance. The primer binding site binds to the primer sequence that is formed from the endogenous DNA strand of the target site when it becomes nicked by the prime editor complex, thereby exposing a 3′ end on the endogenous nicked strand. As explained herein, the binding of the primer sequence to the primer binding site on the extension arm of the PEgRNA creates a duplex region with an exposed 3′ end (i.e., the 3′ of the primer sequence), which then provides a substrate for a polymerase to begin polymerizing a single strand of DNA from the exposed 3′ end along the length of the DNA synthesis template. The sequence of the single strand DNA product is the complement of the DNA synthesis template. Polymerization continues towards the 5′ of the DNA synthesis template (or extension arm) until polymerization terminates. Thus, the DNA synthesis template represents the portion of the extension arm that is encoded into a single strand DNA product (i.e., the 3′ single strand DNA flap containing the desired genetic edit information) by the polymerase of the prime editor complex and which ultimately replaces the corresponding endogenous DNA strand of the target site that sits immediately downstream of the PE-induced nick site. Without being bound by theory, polymerization of the DNA synthesis template continues towards the 5′ end of the extension arm until a termination event. Polymerization may terminate in a variety of ways, including, but not limited to (a) reaching a 5′ terminus of the PEgRNA (e.g., in the case of the 5′ extension arm wherein the DNA polymerase simply runs out of template), (b) reaching an impassable RNA secondary structure (e.g., hairpin or stem/loop), or (c) reaching a replication termination signal, e.g., a specific nucleotide sequence that blocks or inhibits the polymerase, or a nucleic acid topological signal, such as, supercoiled DNA or RNA.


Flap Endonuclease (e.g., FEN1)


As used herein, the term “flap endonuclease” refers to an enzyme that catalyzes the removal of 5′ single strand DNA flaps. These are naturally occurring enzymes that process the removal of 5′ flaps formed during cellular processes, including DNA replication. The prime editing methods herein described may utilize endogenously supplied flap endonucleases or those provided in trans to remove the 5′ flap of endogenous DNA formed at the target site during prime editing. Flap endonucleases are known in the art and can be found described in Patel et al., “Flap endonucleases pass 5′-flaps through a flexible arch using a disorder-thread-order mechanism to confer specificity for free 5′-ends,” Nucleic Acids Research, 2012, 40(10): 4507-4519, Tsutakawa et al., “Human flap endonuclease structures, DNA double-base flipping, and a unified understanding of the FEN1 superfamily,” Cell, 2011, 145(2): 198-211, and Balakrishnan et al., “Flap Endonuclease 1,” Annu Rev Biochem, 2013, Vol 82: 119-138 (each of which are incorporated herein by reference). An exemplary flap endonuclease is FEN1, which can be represented by the following amino acid sequence:














DESCRIPTION
SEQUENCE
SEQ ID NO:







FEN1
MGIQGLAKLIADVAPSAIRENDIKSYFGRKVAIDASMSIYQFL
SEQ ID NO: 7


WILD TYPE
IAVRQGGDVLQNEEGETTSHLMGMFYRTIRMMENGIKPVY




VFDGKPPQLKSGELAKRSERRAEAEKQLQQAQAAGAEQEV




EKFTKRLVKVTKQHNDECKHLLSLMGIPYLDAPSEAEASCA




ALVKAGKVYAAATEDMDCLTFGSPVLMRHLTASEAKKLPIQ




EFHLSRILQELGLNQEQFVDLCILLGSDYCESIRGIGPKRAVD




LIQKHKSIEEIVRRLDPNKYPVPENWLHKEAHQLFLEPEVLD




PESVELKWSEPNEEELIKFMCGEKQFSEERIRSGVKRLSKSR




QGSTQGRLDDFFKVTGSLSSAKRKEPEPKGSTKKKAKTGAA




GKFKRGK










Functional Equivalent


The term “functional equivalent” refers to a second biomolecule that is equivalent in function, but not necessarily equivalent in structure to a first biomolecule. For example, a “Cas9 equivalent” refers to a protein that has the same or substantially the same functions as Cas9, but not necessarily the same amino acid sequence. In the context of the disclosure, the specification refers throughout to “a protein X, or a functional equivalent thereof.” In this context, a “functional equivalent” of protein X embraces any homolog, paralog, fragment, naturally occurring, engineered, mutated, or synthetic version of protein X which bears an equivalent function.


Fusion Protein


The term “fusion protein” as used herein refers to a hybrid polypeptide which comprises protein domains from at least two different proteins. One protein may be located at the amino-terminal (N-terminal) portion of the fusion protein or at the carboxy-terminal (C-terminal) protein thus forming an “amino-terminal fusion protein” or a “carboxy-terminal fusion protein,” respectively. A protein may comprise different domains, for example, a nucleic acid binding domain (e.g., the gRNA binding domain of Cas9 that directs the binding of the protein to a target site) and a nucleic acid cleavage domain or a catalytic domain of a nucleic-acid editing protein. Another example includes a Cas9 or equivalent thereof to a reverse transcriptase. Any of the proteins provided herein may be produced by any method known in the art. For example, the proteins provided herein may be produced via recombinant protein expression and purification, which is especially suited for fusion proteins comprising a peptide linker. Methods for recombinant protein expression and purification are well known, and include those described by Green and Sambrook, Molecular Cloning: A Laboratory Manual (4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2012)), the entire contents of which are incorporated herein by reference.


Gene of Interest (GOI)


The term “gene of interest” or “GOI” refers to a gene that encodes a biomolecule of interest (e.g., a protein or an RNA molecule). A protein of interest can include any intracellular protein, membrane protein, or extracellular protein, e.g., a nuclear protein, transcription factor, nuclear membrane transporter, intracellular organelle associated protein, a membrane receptor, a catalytic protein, and enzyme, a therapeutic protein, a membrane protein, a membrane transport protein, a signal transduction protein, or an immunological protein (e.g., an IgG or other antibody protein), etc. The gene of interest may also encode an RNA molecule, including, but not limited to, messenger RNA (mRNA), transfer RNA (tRNA), ribosomal RNA (rRNA), small nuclear RNA (snRNA), antisense RNA, guide RNA, microRNA (miRNA), small interfering RNA (siRNA), and cell-free RNA (cfRNA).


Guide RNA (“gRNA”)


As used herein, the term “guide RNA” is a particular type of guide nucleic acid which is mostly commonly associated with a Cas protein of a CRISPR-Cas9 and which associates with Cas9, directing the Cas9 protein to a specific sequence in a DNA molecule that includes complementarity to the protospacer sequence of the guide RNA. However, this term also embraces the equivalent guide nucleic acid molecules that associate with Cas9 equivalents, homologs, orthologs, or paralogs, whether naturally occurring or non-naturally occurring (e.g., engineered or recombinant), and which otherwise program the Cas9 equivalent to localize to a specific target nucleotide sequence. The Cas9 equivalents may include other napDNAbp from any type of CRISPR system (e.g., type II, V, VI), including Cpf1 (a type-V CRISPR-Cas systems), C2c1 (a type V CRISPR-Cas system), C2c2 (a type VI CRISPR-Cas system) and C2c3 (a type V CRISPR-Cas system). Further Cas-equivalents are described in Makarova et al., “C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector,” Science 2016; 353(6299), the contents of which are incorporated herein by reference. Exemplary sequences are and structures of guide RNAs are provided herein. In addition, methods for designing appropriate guide RNA sequences are provided herein. As used herein, the “guide RNA” may also be referred to as a “traditional guide RNA” to contrast it with the modified forms of guide RNA termed “prime editing guide RNAs” (or “PEgRNAs”) which have been invented for the prime editing methods and composition disclosed herein.


Guide RNAs or PEgRNAs may comprise various structural elements that include, but are not limited to:


Spacer sequence—the sequence in the guide RNA or PEgRNA (having about 20 nts in length) which binds to the protospacer in the target DNA.


gRNA core (or gRNA scaffold or backbone sequence)—refers to the sequence within the gRNA that is responsible for Cas9 binding, it does not include the 20 bp spacer/targeting sequence that is used to guide Cas9 to target DNA.


Extension arm—a single strand extension at the 3′ end or the 5′ end of the PEgRNA which comprises a primer binding site and a DNA synthesis template sequence that encodes via a polymerase (e.g., a reverse transcriptase) a single stranded DNA flap containing the genetic change of interest, which then integrates into the endogenous DNA by replacing the corresponding endogenous strand, thereby installing the desired genetic change.


Transcription terminator—the guide RNA or PEgRNA may comprise a transcriptional termination sequence at the 3′ of the molecule.


Homology Arm


The term “homology arm” refers to a portion of the extension arm that encodes a portion of the resulting reverse transcriptase-encoded single strand DNA flap that is to be integrated into the target DNA site by replacing the endogenous strand. The portion of the single strand DNA flap encoded by the homology arm is complementary to the non-edited strand of the target DNA sequence, which facilitates the displacement of the endogenous strand and annealing of the single strand DNA flap in its place, thereby installing the edit. This component is further defined elsewhere. The homology arm is part of the DNA synthesis template since it is by definition encoded by the polymerase of the prime editors described herein.


Host Cell


The term “host cell,” as used herein, refers to a cell that can host, replicate, and express a vector described herein, e.g., a vector comprising a nucleic acid molecule encoding a fusion protein comprising a Cas9 or Cas9 equivalent and a reverse transcriptase.


Inteins


As used herein, the term “intein” refers to auto-processing polypeptide domains found in organisms from all domains of life. An intein (intervening protein) carries out a unique auto-processing event known as protein splicing in which it excises itself out from a larger precursor polypeptide through the cleavage of two peptide bonds and, in the process, ligates the flanking extein (external protein) sequences through the formation of a new peptide bond. This rearrangement occurs post-translationally (or possibly co-translationally), as intein genes are found embedded in frame within other protein-coding genes. Furthermore, intein-mediated protein splicing is spontaneous; it requires no external factor or energy source, only the folding of the intein domain. This process is also known as cis-protein splicing, as opposed to the natural process of trans-protein splicing with “split inteins.” Inteins are the protein equivalent of the self-splicing RNA introns (see Perler et al., Nucleic Acids Res. 22:1125-1127 (1994)), which catalyze their own excision from a precursor protein with the concomitant fusion of the flanking protein sequences, known as exteins (reviewed in Perler et al., Curr. Opin. Chem. Biol. 1:292-299 (1997); Perler, F. B. Cell 92(1):1-4 (1998); Xu et al., EMBO J. 15(19):5146-5153 (1996)).


As used herein, the term “protein splicing” refers to a process in which an interior region of a precursor protein (an intein) is excised and the flanking regions of the protein (exteins) are ligated to form the mature protein. This natural process has been observed in numerous proteins from both prokaryotes and eukaryotes (Perler, F. B., Xu, M. Q., Paulus, H. Current Opinion in Chemical Biology 1997, 1, 292-299; Perler, F. B. Nucleic Acids Research 1999, 27, 346-347). The intein unit contains the necessary components needed to catalyze protein splicing and often contains an endonuclease domain that participates in intein mobility (Perler, F. B., Davis, E. O., Dean, G. E., Gimble, F. S., Jack, W. E., Neff, N., Noren, C. J., Thomer, J., Belfort, M. Nucleic Acids Research 1994, 22, 1127-1127). The resulting proteins are linked, however, not expressed as separate proteins. Protein splicing may also be conducted in trans with split inteins expressed on separate polypeptides spontaneously combine to form a single intein which then undergoes the protein splicing process to join to separate proteins.


The elucidation of the mechanism of protein splicing has led to a number of intein-based applications (Comb, et al., U.S. Pat. No. 5,496,714; Comb, et al., U.S. Pat. No. 5,834,247; Camarero and Muir, J. Amer. Chem. Soc., 121:5597-5598 (1999); Chong, et al., Gene, 192:271-281 (1997), Chong, et al., Nucleic Acids Res., 26:5109-5115 (1998); Chong, et al., J. Biol. Chem., 273:10567-10577 (1998); Cotton, et al. J. Am. Chem. Soc., 121:1100-1101 (1999); Evans, et al., J. Biol. Chem., 274:18359-18363 (1999); Evans, et al., J. Biol. Chem., 274:3923-3926 (1999); Evans, et al., Protein Sci., 7:2256-2264 (1998); Evans, et al., J. Biol. Chem., 275:9091-9094 (2000); Iwai and Pluckthun, FEBS Lett. 459:166-172 (1999); Mathys, et al., Gene, 231:1-13 (1999); Mills, et al., Proc. Natl. Acad. Sci. USA 95:3543-3548 (1998); Muir, et al., Proc. Natl. Acad. Sci. USA 95:6705-6710 (1998); Otomo, et al., Biochemistry 38:16040-16044 (1999); Otomo, et al., J. Biolmol. NMR 14:105-114 (1999); Scott, et al., Proc. Natl. Acad. Sci. USA 96:13638-13643 (1999); Severinov and Muir, J. Biol. Chem., 273:16205-16209 (1998); Shingledecker, et al., Gene, 207:187-195 (1998); Southworth, et al., EMBO J. 17:918-926 (1998); Southworth, et al., Biotechniques, 27:110-120 (1999); Wood, et al., Nat. Biotechnol., 17:889-892 (1999); Wu, et al., Proc. Natl. Acad. Sci. USA 95:9226-9231 (1998a); Wu, et al., Biochim Biophys Acta 1387:422-432 (1998b); Xu, et al., Proc. Natl. Acad. Sci. USA 96:388-393 (1999); Yamazaki, et al., J. Am. Chem. Soc., 120:5591-5592 (1998)). Each reference is incorporated herein by reference.


Ligand-Dependent Intein


The term “ligand-dependent intein,” as used herein refers to an intein that comprises a ligand-binding domain Typically, the ligand-binding domain is inserted into the amino acid sequence of the intein, resulting in a structure intein (N)—ligand-binding domain—intein (C). Typically, ligand-dependent inteins exhibit no or only minimal protein splicing activity in the absence of an appropriate ligand, and a marked increase of protein splicing activity in the presence of the ligand. In some embodiments, the ligand-dependent intein does not exhibit observable splicing activity in the absence of ligand but does exhibit splicing activity in the presence of the ligand. In some embodiments, the ligand-dependent intein exhibits an observable protein splicing activity in the absence of the ligand, and a protein splicing activity in the presence of an appropriate ligand that is at least 5 times, at least 10 times, at least 50 times, at least 100 times, at least 150 times, at least 200 times, at least 250 times, at least 500 times, at least 1000 times, at least 1500 times, at least 2000 times, at least 2500 times, at least 5000 times, at least 10000 times, at least 20000 times, at least 25000 times, at least 50000 times, at least 100000 times, at least 500000 times, or at least 1000000 times greater than the activity observed in the absence of the ligand. In some embodiments, the increase in activity is dose dependent over at least 1 order of magnitude, at least 2 orders of magnitude, at least 3 orders of magnitude, at least 4 orders of magnitude, or at least 5 orders of magnitude, allowing for fine-tuning of intein activity by adjusting the concentration of the ligand. Suitable ligand-dependent inteins are known in the art, and in include those provided below and those described in published U.S. Patent Application U.S. 2014/0065711 A1; Mootz et al., “Protein splicing triggered by a small molecule.” J. Am. Chem. Soc. 2002; 124, 9044-9045; Mootz et al., “Conditional protein splicing: a new tool to control protein structure and function in vitro and in vivo.” J. Am. Chem. Soc. 2003; 125, 10561-10569; Buskirk et al., Proc. Natl. Acad. Sci. USA. 2004; 101, 10505-10510); Skretas & Wood, “Regulation of protein activity with small-molecule-controlled inteins.” Protein Sci. 2005; 14, 523-532; Schwartz, et al., “Post-translational enzyme activation in an animal via optimized conditional protein splicing.” Nat. Chem. Biol. 2007; 3, 50-54; Peck et al., Chem. Biol. 2011; 18 (5), 619-630; the entire contents of each are hereby incorporated by reference. Exemplary sequences are as follows:













NAME
SEQUENCE OF LIGAND-DEPENDENT INTEIN







2-4 INTEIN:
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGAIVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 8)





3-2 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAVAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGAIVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYTNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 9)





30R3-1 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPIPYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEGLRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 10)





30R3-2 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 11)





30R3-3 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPIPYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 12)





37R3-1 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYNPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLERAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEGLRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC ((SEQ ID NO: 13)





37R3-2 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGAIVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLERAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEGLRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 14)





37R3-3 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAVAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLERAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 15)










Linker


The term “linker,” as used herein, refers to a molecule linking two other molecules or moieties. The linker can be an amino acid sequence in the case of a linker joining two fusion proteins. For example, a Cas9 can be fused to a reverse transcriptase by an amino acid linker sequence. The linker can also be a nucleotide sequence in the case of joining two nucleotide sequences together. For example, in the instant case, the traditional guide RNA is linked via a spacer or linker nucleotide sequence to the RNA extension of a prime editing guide RNA which may comprise a RT template sequence and an RT primer binding site. In other embodiments, the linker is an organic molecule, group, polymer, or chemical moiety. In some embodiments, the linker is 5-100 amino acids in length, for example, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 30-35, 35-40, 40-45, 45-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-150, or 150-200 amino acids in length. Longer or shorter linkers are also contemplated.


Isolated


“Isolated” means altered or removed from the natural state. For example, a nucleic 20 acid or a peptide naturally present in a living animal is not “isolated,” but the same nucleic acid or peptide partially or completely separated from the coexisting materials of its natural state is “isolated.” An isolated nucleic acid or protein can exist in substantially purified form, or can exist in a non-native environment such as, for example, a host cell.


In some embodiments, a gene of interest is encoded by an isolated nucleic acid. As used herein, the term “isolated,” refers to the characteristic of a material as provided herein being removed from its original or native environment (e.g., the natural environment if it is naturally occurring). Therefore, a naturally-occurring polynucleotide or protein or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated by human intervention from some or all of the coexisting materials in the natural system, is isolated. An artificial or engineered material, for example, a non-naturally occurring nucleic acid construct, such as the expression constructs and vectors described herein, are, accordingly, also referred to as isolated. A material does not have to be purified in order to be isolated. Accordingly, a material may be part of a vector and/or part of a composition, and still be isolated in that such vector or composition is not part of the environment in which the material is found in nature.


MS2 Tagging Technique


In various embodiments (e.g., as depicted in the embodiments of FIGS. 72-73 and in Example 19), the term “MS2 tagging technique” refers to the combination of an “RNA-protein interaction domain” (aka “RNA-protein recruitment domain or protein”) paired up with an RNA-binding protein that specifically recognizes and binds to the RNA-protein interaction domain, e.g., a specific hairpin structure. These types of systems can be leveraged to recruit a variety of functionalities to a prime editor complex that is bound to a target site. The MS2 tagging technique is based on the natural interaction of the MS2 bacteriophage coat protein (“MCP” or “MS2cp”) with a stem-loop or hairpin structure present in the genome of the phage, i.e., the “MS2 hairpin.” In the case of prime editing, the MS2 tagging technique comprises introducing the MS2 hairpin into a desired RNA molecule involved in prime editing (e.g., a PEgRNA or a tPERT), which then constitutes a specific interactable binding target for an RNA-binding protein that recognizes and binds to that structure. In the case of the MS2 hairpin, it is recognized and bound by the MS2 bacteriophage coat protein (MCP). And, if MCP is fused to another protein (e.g., a reverse transcriptase or other DNA polymerase), then the MS2 hairpin may be used to “recruit” that other protein in trans to the target site occupied by the prime editing complex.


The prime editors described herein may incorporate as an aspect any known RNA-protein interaction domain to recruit or “co-localize” specific functions of interest to a prime editor complex. A review of other modular RNA-protein interaction domains are described in the art, for example, in Johansson et al., “RNA recognition by the MS2 phage coat protein,” Sem Virol., 1997, Vol. 8(3): 176-185; Delebecque et al., “Organization of intracellular reactions with rationally designed RNA assemblies,” Science, 2011, Vol. 333: 470-474; Mali et al., “Cas9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering,” Nat. Biotechnol., 2013, Vol. 31: 833-838; and Zalatan et al., “Engineering complex synthetic transcriptional programs with CRISPR RNA scaffolds,” Cell, 2015, Vol. 160: 339-350, each of which are incorporated herein by reference in their entireties. Other systems include the PP7 hairpin, which specifically recruits the PCP protein, and the “com” hairpin, which specifically recruits the Com protein. See Zalatan et al.


The nucleotide sequence of the MS2 hairpin (or equivalently referred to as the “MS2 aptamer”) is: GCCAACATGAGGATCACCCATGTCTGCAGGGCC (SEQ ID NO: 763).


The amino acid sequence of the MCP or MS2cp is:









(SEQ ID NO: 764)


GSASNFTQFVLVDNGGTGDVTVAPSNFANGVAEWISSNSRSQAYKVTCS


VRQSSAQNRKYTIKVEVPKVATQTVGGEELPVAGWRSYLNMELTIPIFA


TNSDCELIVKAMQGLLKDGNPIPSAIAANSGIY.






The MS2 hairpin (or “MS2 aptamer”) may also be referred to as a type of “RNA effector recruitment domain” (or equivalently as “RNA-binding protein recruitment domain” or simply as “recruitment domain”) since it is a physical structure (e.g., a hairpin) that is installed into a PEgRNA or tPERT that effectively recruits other effector functions (e.g., RNA-binding proteins having various functions, such as DNA polymerases or other DNA-modifying enzymes) to the PEgRNA or rPERT that is so modified, and thus, co-localizing effector functions in trans to the prime editing machinery. This application is not intended to be limited in any way to any particular RNA effector recruitment domains and may include any available such domain, including the MS2 hairpin. Example 19 and FIG. 72(b) depicts the use of the MS2 aptamer joined to a DNA synthesis domain (i.e., the tPERT molecule) and a prime editor that comprises an MS2cp protein fused to a PE2 to cause the co-localization of the prime editor complex (MS2cp-PE2:sgRNA complex) bound to the target DNA site and the DNA synthesis domain of the tPERT molecule to effectuate the


napDNAbp


As used herein, the term “nucleic acid programmable DNA binding protein” or “napDNAbp,” of which Cas9 is an example, refer to proteins that use RNA:DNA hybridization to target and bind to specific sequences in a DNA molecule. Each napDNAbp is associated with at least one guide nucleic acid (e.g., guide RNA), which localizes the napDNAbp to a DNA sequence that comprises a DNA strand (i.e., a target strand) that is complementary to the guide nucleic acid, or a portion thereof (e.g., the protospacer of a guide RNA). In other words, the guide nucleic-acid “programs” the napDNAbp (e.g., Cas9 or equivalent) to localize and bind to a complementary sequence.


Without being bound by theory, the binding mechanism of a napDNAbp—guide RNA complex, in general, includes the step of forming an R-loop whereby the napDNAbp induces the unwinding of a double-strand DNA target, thereby separating the strands in the region bound by the napDNAbp. The guide RNA protospacer then hybridizes to the “target strand.” This displaces a “non-target strand” that is complementary to the target strand, which forms the single strand region of the R-loop. In some embodiments, the napDNAbp includes one or more nuclease activities, which then cut the DNA, leaving various types of lesions. For example, the napDNAbp may comprise a nuclease activity that cuts the non-target strand at a first location, and/or cuts the target strand at a second location. Depending on the nuclease activity, the target DNA can be cut to form a “double-stranded break” whereby both strands are cut. In other embodiments, the target DNA can be cut at only a single site, i.e., the DNA is “nicked” on one strand. Exemplary napDNAbp with different nuclease activities include “Cas9 nickase” (“nCas9”) and a deactivated Cas9 having no nuclease activities (“dead Cas9” or “dCas9”). Exemplary sequences for these and other napDNAbp are provided herein.


Nickase


The term “nickase” refers to a Cas9 with one of the two nuclease domains inactivated. This enzyme is capable of cleaving only one strand of a target DNA.


Nuclear Localization Sequence (NLS)


The term “nuclear localization sequence” or “NLS” refers to an amino acid sequence that promotes import of a protein into the cell nucleus, for example, by nuclear transport. Nuclear localization sequences are known in the art and would be apparent to the skilled artisan. For example, NLS sequences are described in Plank et al., international PCT application, PCT/EP2000/011690, filed Nov. 23, 2000, published as WO/2001/038547 on May 31, 2001, the contents of which are incorporated herein by reference for its disclosure of exemplary nuclear localization sequences. In some embodiments, a NLS comprises the amino acid sequence PKKKRKV (SEQ ID NO: 16) or MDSLLMNRRKFLYQFKNVRWAKGRRETYLC (SEQ ID NO: 17).


Nucleic Acid Molecule


The term “nucleic acid,” as used herein, refers to a polymer of nucleotides. The polymer may include natural nucleosides (i.e., adenosine, thymidine, guanosine, cytidine, uridine, deoxyadenosine, deoxythymidine, deoxyguanosine, and deoxycytidine), nucleoside analogs (e.g., 2-aminoadenosine, 2-thiothymidine, inosine, pyrrolo-pyrimidine, 3-methyl adenosine, 5-methylcytidine, C5 bromouridine, C5 fluorouridine, C5 iodouridine, C5 propynyl uridine, C5 propynyl cytidine, C5 methylcytidine, 7 deazaadenosine, 7 deazaguanosine, 8 oxoadenosine, 8 oxoguanosine, O(6) methylguanine, 4-acetylcytidine, 5-(carboxyhydroxymethyl)uridine, dihydrouridine, methylpseudouridine, 1-methyl adenosine, 1-methyl guanosine, N6-methyl adenosine, and 2-thiocytidine), chemically modified bases, biologically modified bases (e.g., methylated bases), intercalated bases, modified sugars (e.g., 2′-fluororibose, ribose, 2′-deoxyribose, 2′-O-methylcytidine, arabinose, and hexose), or modified phosphate groups (e.g., phosphorothioates and 5′ N phosphoramidite linkages).


PEgRNA


As used herein, the terms “prime editing guide RNA” or “PEgRNA” or “extended guide RNA” refer to a specialized form of a guide RNA that has been modified to include one or more additional sequences for implementing the prime editing methods and compositions described herein. As described herein, the prime editing guide RNA comprise one or more “extended regions” of nucleic acid sequence. The extended regions may comprise, but are not limited to, single-stranded RNA or DNA. Further, the extended regions may occur at the 3′ end of a traditional guide RNA. In other arrangements, the extended regions may occur at the 5′ end of a traditional guide RNA. In still other arrangements, the extended region may occur at an intramolecular region of the traditional guide RNA, for example, in the gRNA core region which associates and/or binds to the napDNAbp. The extended region comprises a “DNA synthesis template” which encodes (by the polymerase of the prime editor) a single-stranded DNA which, in turn, has been designed to be (a) homologous with the endogenous target DNA to be edited, and (b) which comprises at least one desired nucleotide change (e.g., a transition, a transversion, a deletion, or an insertion) to be introduced or integrated into the endogenous target DNA. The extended region may also comprise other functional sequence elements, such as, but not limited to, a “primer binding site” and a “spacer or linker” sequence, or other structural elements, such as, but not limited to aptamers, stem loops, hairpins, toe loops (e.g., a 3′ toeloop), or an RNA-protein recruitment domain (e.g., MS2 hairpin). As used herein the “primer binding site” comprises a sequence that hybridizes to a single-strand DNA sequence having a 3′ end generated from the nicked DNA of the R-loop.


In certain embodiments, the PEgRNAs are represented by FIG. 3A, which shows a PEgRNA having a 5′ extension arm, a spacer, and a gRNA core. The 5′ extension further comprises in the 5′ to 3′ direction a reverse transcriptase template, a primer binding site, and a linker. As shown, the reverse transcriptase template may also be referred to more broadly as the “DNA synthesis template” where the polymerase of a prime editor described herein is not an RT, but another type of polymerase.


In certain other embodiments, the PEgRNAs are represented by FIG. 3B, which shows a PEgRNA having a 5′ extension arm, a spacer, and a gRNA core. The 5′ extension further comprises in the 5′ to 3′ direction a reverse transcriptase template, a primer binding site, and a linker. As shown, the reverse transcriptase template may also be referred to more broadly as the “DNA synthesis template” where the polymerase of a prime editor described herein is not an RT, but another type of polymerase.


In still other embodiments, the PEgRNAs are represented by FIG. 3D, which shows a PEgRNA having in the 5′ to 3′ direction a spacer (1), a gRNA core (2), and an extension arm (3). The extension arm (3) is at the 3′ end of the PEgRNA. The extension arm (3) further comprises in the 5′ to 3′ direction a “primer binding site” (A), an “edit template” (B), and a “homology arm” (C). The extension arm (3) may also comprise an optional modifier region at the 3′ and 5′ ends, which may be the same sequences or different sequences. In addition, the 3′ end of the PEgRNA may comprise a transcriptional terminator sequence. These sequence elements of the PEgRNAs are further described and defined herein.


In still other embodiments, the PEgRNAs are represented by FIG. 3E, which shows a PEgRNA having in the 5′ to 3′ direction an extension arm (3), a spacer (1), and a gRNA core (2). The extension arm (3) is at the 5′ end of the PEgRNA. The extension arm (3) further comprises in the 3′ to 5′ direction a “primer binding site” (A), an “edit template” (B), and a “homology arm” (C). The extension arm (3) may also comprise an optional modifier region at the 3′ and 5′ ends, which may be the same sequences or different sequences. The PEgRNAs may also comprise a transcriptional terminator sequence at the 3′ end. These sequence elements of the PEgRNAs are further described and defined herein.


PE1


As used herein, “PE1” refers to a PE complex comprising a fusion protein comprising Cas9(H840A) and a wild type MMLV RT having the following structure: [NLS]-[Cas9(H840A)]-[linker]-[MMLV_RT(wt)]+a desired PEgRNA, wherein the PE fusion has the amino acid sequence of SEQ ID NO: 123, which is shown as follows;










(SEQ ID NO: 123)




MKRTADGSEFESPKKKRKV
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK








KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFL







VEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHF







LIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLPG







EKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLA







AKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQS







KNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLG







ELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEE







VVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLS







GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKD







KDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSR







KLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIAN







LAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEE







GIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSFL







KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGG







LSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKD







FQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIG







KATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQV







NIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGK







SKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLA







SAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFS







KRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTK







EVLDATLIHQSITGLYETRIDLSQLGGD

SGGSSGGSSGSETPGTSESATPESSGGSSGGSS

TLNI







EDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPMSQEARLGI







KPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSH







QWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFDEALHRDLADF







RIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQKQVKYLGYLLKEGQR







WLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPGFAEMAAPLYPLTKTGTLFNWGPDQQKAYQ







EIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMV







AAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQFGPWALNPAT







LLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTDGSSLLQEGQRKAGAAVFFETEVIWAK







ALPAGTSAQRAELIALTQALKMAEGKKLNVYTDSRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALL







KALFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP
SGGSKRTADGSEF







EPKKKRKV 



KEY:



NUCLEAR LOCALIZATION SEQUENCE (NLS) TOP: (SEQ ID NO: 124), BOTTOM: (SEQ ID NO: 133)




CAS9(H840A) (SEQ ID NO: 126)





33-AMINO ACID LINKER
(SEQ ID NO: 127)



M-MLV reverse transcriptase (SEQ ID NO: 128).







PE2


As used herein, “PE2” refers to a PE complex comprising a fusion protein comprising Cas9(H840A) and a variant MMLV RT having the following structure: [NLS]-[Cas9(H840A)]-[linker]-[MMLV_RT(D200N)(T330P)(L603W)(T306K)(W313F)]+a desired PEgRNA, wherein the PE fusion has the amino acid sequence of SEQ ID NO: 134, which is shown as follows:










(SEQ ID NO: 134)




MKRTADGSEFESPKKKRKV
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK








KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFL







VEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHF







LIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLPG







EKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLA







AKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQS







KNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLG







ELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEE







VVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLS







GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKD







KDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSR







KLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIAN







LAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEE







GIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSFL







KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGG







LSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKD







FQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIG







KATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQV







NIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGK







SKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLA







SAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFS







KRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTK







EVLDATLIHQSITGLYETRIDLSQLGGD

SGGSSGGSSGSETPGTSESATPESSGGSSGGSS

TLNI







EDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLnPLKATSTPVSIKQYPMSQEARLGI







KPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSH







QWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADF







RIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQKQVKYLGYLLKEGQR







WLTEARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPGFAEMAAPLYPLTKPGTLFNWGPDQQKAYQ







EIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMV







AAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQFGPWALNPAT







LLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTDGSSLLQEGQRKAGAAVFTETEVIWAK







ALPAGTSAQRAELIALTQALKMAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALL







KALFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP
SGGSKRTADGSEF







EPKKKRKV



KEY:



NUCLEAR LOCALIZATION SEQUENCE (NLS) TOP: (SEQ ID NO: 124), BOTTOM: (SEQ ID NO: 133)




CAS9(H840A) (SEQ ID NO: 137)





33-AMINO ACID LINKER
(SEQ ID NO: 127)



M-MLV reverse transcriptase (SEQ ID NO: 139).







PE3


As used herein, “PE3” refers to PE2 plus a second-strand nicking guide RNA that complexes with the PE2 and introduces a nick in the non-edited DNA strand in order to induce preferential replacement of the edited strand.


PE3b


As used herein, “PE3b” refers to PE3 but wherein the second-strand nicking guide RNA is designed for temporal control such that the second strand nick is not introduced until after the installation of the desired edit. This is achieved by designing a gRNA with a spacer sequence that matches only the edited strand, but not the original allele. Using this strategy, referred to hereafter as PE3b, mismatches between the protospacer and the unedited allele should disfavor nicking by the sgRNA until after the editing event on the PAM strand takes place.


PE-Short


As used herein, “PE-short” refers to a PE construct that is fused to a C-terminally truncated reverse transcriptase, and has the following amino acid sequence:










(SEQ ID NO: 765)




MKRTADGSEFESPKKKRKV
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK








KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFL







VEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGH







FLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLP







GEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFL







AAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFD







QSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIH







LGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWN







FEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKP







AFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLL







KIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGW







GRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLH







EHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKNSRERM







KRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIV







PQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTK







AERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVS







DFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAK







SEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKV







LSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVA







KVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENG







RKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEI







lEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDR







KRYTSTKEVLDATLIHOSITGLYETRIDLSOLGGD

SGGSSGGSSGSETPGTSESATPESSGGSS







GGSSTLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLHPLKATSTPVSIKQYP






MSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNPY







NLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNE







ALHRDLADFRIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQKQVKYL







GYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPGFAEMAAPLYPLTKPGTLFNW







GPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVA







AGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRV







QFGPWALNPATLLPLPEEGLQHNCLDNSRLIN
SGGSKRTADGSEFEPKKKRKV



KEY:



NUCLEAR LOCALIZATION SEQUENCE (NLS) TOP: (SEQ ID NO: 124), BOTTOM: (SEQ ID NO: 133)




CAS9(H840A) (SEQ ID NO: 157)





33-AMINO ACID LINKER 1
(SEQ ID NO: 127)



M-MLV TRUNCATED REVERSE TRANSCRIPTASE (SEQ ID NO: 766)







Peptide Tag


The term “peptide tag” refers to a peptide amino acid sequence that is genetically fused to a protein sequence to impart one or more functions onto the proteins that facilitate the manipulation of the protein for various purposes, such as, visualization, purification, solubilization, and separation, etc. Peptide tags can include various types of tags categorized by purpose or function, which may include “affinity tags” (to facilitate protein purification), “solubilization tags” (to assist in proper folding of proteins), “chromatography tags” (to alter chromatographic properties of proteins), “epitope tags” (to bind to high affinity antibodies), “fluorescence tags” (to facilitate visualization of proteins in a cell or in vitro).


Polymerase


As used herein, the term “polymerase” refers to an enzyme that synthesizes a nucleotide strand and that may be used in connection with the prime editor systems described herein. The polymerase can be a “template-dependent” polymerase (i.e., a polymerase that synthesizes a nucleotide strand based on the order of nucleotide bases of a template strand). The polymerase can also be a “template-independent” polymerase (i.e., a polymerase that synthesizes a nucleotide strand without the requirement of a template strand). A polymerase may also be further categorized as a “DNA polymerase” or an “RNA polymerase.” In various embodiments, the prime editor system comprises a DNA polymerase. In various embodiments, the DNA polymerase can be a “DNA-dependent DNA polymerase” (i.e., whereby the template molecule is a strand of DNA). In such cases, the DNA template molecule can be a PEgRNA, wherein the extension arm comprises a strand of DNA. In such cases, the PEgRNA may be referred to as a chimeric or hybrid PEgRNA which comprises an RNA portion (i.e., the guide RNA components, including the spacer and the gRNA core) and a DNA portion (i.e., the extension arm). In various other embodiments, the DNA polymerase can be an “RNA-dependent DNA polymerase” (i.e., whereby the template molecule is a strand of RNA). In such cases, the PEgRNA is RNA, i.e., including an RNA extension. The term “polymerase” may also refer to an enzyme that catalyzes the polymerization of nucleotide (i.e., the polymerase activity). Generally, the enzyme will initiate synthesis at the 3′-end of a primer annealed to a polynucleotide template sequence (e.g., such as a primer sequence annealed to the primer binding site of a PEgRNA) and will proceed toward the 5′ end of the template strand. A “DNA polymerase” catalyzes the polymerization of deoxynucleotides. As used herein in reference to a DNA polymerase, the term DNA polymerase includes a “functional fragment thereof”. A “functional fragment thereof” refers to any portion of a wild-type or mutant DNA polymerase that encompasses less than the entire amino acid sequence of the polymerase and which retains the ability, under at least one set of conditions, to catalyze the polymerization of a polynucleotide. Such a functional fragment may exist as a separate entity, or it may be a constituent of a larger polypeptide, such as a fusion protein.


Prime Editing and Multi-Flap Prime Editing


As used herein, the term “prime editing” or “classical prime editing” refers to an approach for gene editing using napDNAbps, a polymerase (e.g., a reverse transcriptase), and specialized guide RNAs that include a DNA synthesis template for encoding desired new genetic information (or deleting genetic information) that is then incorporated into a target DNA sequence. Certain embodiments of prime editing are described in the embodiments of FIGS. 1A-1H and FIG. 72(a)-72(c), among other figures. Classical prime editing is described in the inventors publication of Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149-157 (2019), which is incorporated herein by reference in its entirety.


Prime editing represents a platform for genome editing that is a versatile and precise genome editing method that directly writes new genetic information into a specified DNA site using a nucleic acid programmable DNA binding protein (“napDNAbp”) working in association with a polymerase (i.e., in the form of a fusion protein or otherwise provided in trans with the napDNAbp), wherein the prime editing system is programmed with a prime editing (PE) guide RNA (“PEgRNA”) that both specifies the target site and templates the synthesis of the desired edit in the form of a replacement DNA strand by way of an extension (either DNA or RNA) engineered onto a guide RNA (e.g., at the 5′ or 3′ end, or at an internal portion of a guide RNA). The replacement strand containing the desired edit (e.g., a single nucleobase substitution) shares the same (or is homologous to) sequence as the endogenous strand (immediately downstream of the nick site) of the target site to be edited (with the exception that it includes the desired edit). Through DNA repair and/or replication machinery, the endogenous strand downstream of the nick site is replaced by the newly synthesized replacement strand containing the desired edit. In some cases, prime editing may be thought of as a “search-and-replace” genome editing technology since the prime editors, as described herein, not only search and locate the desired target site to be edited, but at the same time, encode a replacement strand containing a desired edit which is installed in place of the corresponding target site endogenous DNA strand. The prime editors of the present disclosure relate, in part, to the discovery that the mechanism of target-primed reverse transcription (TPRT) or “prime editing” can be leveraged or adapted for conducting precision CRISPR/Cas-based genome editing with high efficiency and genetic flexibility (e.g., as depicted in various embodiments of FIGS. 1A-1F). TPRT is naturally used by mobile DNA elements, such as mammalian non-LTR retrotransposons and bacterial Group II introns28,29. The inventors have herein used Cas protein-reverse transcriptase fusions or related systems to target a specific DNA sequence with a guide RNA, generate a single strand nick at the target site, and use the nicked DNA as a primer for reverse transcription of an engineered reverse transcriptase template that is integrated with the guide RNA. However, while the concept begins with prime editors that use reverse transcriptase as the DNA polymerase component, the prime editors described herein are not limited to reverse transcriptases but may include the use of virtually any DNA polymerase. Indeed, while the application throughout may refer to prime editors with “reverse transcriptases,” it is set forth here that reverse transcriptases are only one type of DNA polymerase that may work with prime editing. Thus, whereever the specification mentions a “reverse transcriptase,” the person having ordinary skill in the art should appreciate that any suitable DNA polymerase may be used in place of the reverse transcriptase. Thus, in one aspect, the prime editors may comprise Cas9 (or an equivalent napDNAbp) which is programmed to target a DNA sequence by associating it with a specialized guide RNA (i.e., PEgRNA) containing a spacer sequence that anneals to a complement of a protospacer in the target DNA. The specialized guide RNA also contains new genetic information in the form of an extension that encodes a replacement strand of DNA containing a desired genetic alteration which is used to replace a corresponding endogenous DNA strand at the target site. To transfer information from the PEgRNA to the target DNA, the mechanism of prime editing involves nicking the target site in one strand of the DNA to expose a 3′-hydroxyl group. The exposed 3′-hydroxyl group can then be used to prime the DNA polymerization of the edit-encoding extension on PEgRNA directly into the target site. In various embodiments, the extension—which provides the template for polymerization of the replacement strand containing the edit—can be formed from RNA or DNA. In the case of an RNA extension, the polymerase of the prime editor can be an RNA-dependent DNA polymerase (such as, a reverse transcriptase). In the case of a DNA extension, the polymerase of the prime editor may be a DNA-dependent DNA polymerase. The newly synthesized strand (i.e., the replacement DNA strand containing the desired edit) that is formed by the herein disclosed prime editors would be homologous to the genomic target sequence (i.e., have the same sequence as) except for the inclusion of a desired nucleotide change (e.g., a single nucleotide change, a deletion, or an insertion, or a combination thereof). The newly synthesized (or replacement) strand of DNA may also be referred to as a single strand DNA flap, which would compete for hybridization with the complementary homologous endogenous DNA strand, thereby displacing the corresponding endogenous strand. In certain embodiments, the system can be combined with the use of an error-prone reverse transcriptase enzyme (e.g., provided as a fusion protein with the Cas9 domain, or provided in trans to the Cas9 domain). The error-prone reverse transcriptase enzyme can introduce alterations during synthesis of the single strand DNA flap. Thus, in certain embodiments, error-prone reverse transcriptase can be utilized to introduce nucleotide changes to the target DNA. Depending on the error-prone reverse transcriptase that is used with the system, the changes can be random or non-random. Resolution of the hybridized intermediate (comprising the single strand DNA flap synthesized by the reverse transcriptase hybridized to the endogenous DNA strand) can include removal of the resulting displaced flap of endogenous DNA (e.g., with a 5′ end DNA flap endonuclease, FEN1), ligation of the synthesized single strand DNA flap to the target DNA, and assimilation of the desired nucleotide change as a result of cellular DNA repair and/or replication processes. Because templated DNA synthesis offers single nucleotide precision for the modification of any nucleotide, including insertions and deletions, the scope of this approach is very broad and could foreseeably be used for myriad applications in basic science and therapeutics.


In various embodiments, prime editing operates by contacting a target DNA molecule (for which a change in the nucleotide sequence is desired to be introduced) with a nucleic acid programmable DNA binding protein (napDNAbp) complexed with a prime editing guide RNA (PEgRNA). In reference to FIG. 1G, the prime editing guide RNA (PEgRNA) comprises an extension at the 3′ or 5′ end of the guide RNA, or at an intramolecular location in the guide RNA and encodes the desired nucleotide change (e.g., single nucleotide change, insertion, or deletion). In step (a), the napDNAbp/extended gRNA complex contacts the DNA molecule and the extended gRNA guides the napDNAbp to bind to a target locus. In step (b), a nick in one of the strands of DNA of the target locus is introduced (e.g., by a nuclease or chemical agent), thereby creating an available 3′ end in one of the strands of the target locus. In certain embodiments, the nick is created in the strand of DNA that corresponds to the R-loop strand, i.e., the strand that is not hybridized to the guide RNA sequence, i.e., the “non-target strand.” The nick, however, could be introduced in either of the strands. That is, the nick could be introduced into the R-loop “target strand” (i.e., the strand hybridized to the protospacer of the extended gRNA) or the “non-target strand” (i.e., the strand forming the single-stranded portion of the R-loop and which is complementary to the target strand). In step (c), the 3′ end of the DNA strand (formed by the nick) interacts with the extended portion of the guide RNA in order to prime reverse transcription (i.e., “target-primed RT”). In certain embodiments, the 3′ end DNA strand hybridizes to a specific RT priming sequence on the extended portion of the guide RNA, i.e., the “reverse transcriptase priming sequence” or “primer binding site” on the PEgRNA. In step (d), a reverse transcriptase (or other suitable DNA polymerase) is introduced which synthesizes a single strand of DNA from the 3′ end of the primed site towards the 5′ end of the prime editing guide RNA. The DNA polymerase (e.g., reverse transcriptase) can be fused to the napDNAbp or alternatively can be provided in trans to the napDNAbp. This forms a single-strand DNA flap comprising the desired nucleotide change (e.g., the single base change, insertion, or deletion, or a combination thereof) and which is otherwise homologous to the endogenous DNA at or adjacent to the nick site. In step (e), the napDNAbp and guide RNA are released. Steps (f) and (g) relate to the resolution of the single strand DNA flap such that the desired nucleotide change becomes incorporated into the target locus. This process can be driven towards the desired product formation by removing the corresponding 5′ endogenous DNA flap that forms once the 3′ single strand DNA flap invades and hybridizes to the endogenous DNA sequence. Without being bound by theory, the cells endogenous DNA repair and replication processes resolves the mismatched DNA to incorporate the nucleotide change(s) to form the desired altered product. The process can also be driven towards product formation with “second strand nicking,” as exemplified in FIG. 1F. This process may introduce at least one or more of the following genetic changes: transversions, transitions, deletions, and insertions.


The term “prime editor (PE) system” or “prime editor (PE)” or “PE system” or “PE editing system” refers the compositions involved in the method of genome editing using target-primed reverse transcription (TPRT) describe herein, including, but not limited to the napDNAbps, reverse transcriptases, fusion proteins (e.g., comprising napDNAbps and reverse transcriptases), prime editing guide RNAs, and complexes comprising fusion proteins and prime editing guide RNAs, as well as accessory elements, such as second strand nicking components (e.g., second strand sgRNAs) and 5′ endogenous DNA flap removal endonucleases (e.g., FEN1) for helping to drive the prime editing process towards the edited product formation.


Although in the embodiments described thus far the PEgRNA constitutes a single molecule comprising a guide RNA (which itself comprises a spacer sequence and a gRNA core or scaffold) and a 5′ or 3′ extension arm comprising the primer binding site and a DNA synthesis template (e.g., see FIG. 3D, the PEgRNA may also take the form of two individual molecules comprised of a guide RNA and a trans prime editor RNA template (tPERT), which essentially houses the extension arm (including, in particular, the primer binding site and the DNA synthesis domain) and an RNA-protein recruitment domain (e.g., MS2 aptamer or hairpin) in the same molecule which becomes co-localized or recruited to a modified prime editor complex that comprises a tPERT recruiting protein (e.g., MS2cp protein, which binds to the MS2 aptamer). See FIG. 3G and FIG. 3H as an example of a tPERT that may be used with prime editing.


In the “dual-flap prime editing system”, two pegRNAs are used to target opposite strands of a genomic site and direct the synthesis of two complementary 3′ flaps containing edited DNA sequence (FIG. 91). Unlike classical prime editing, there is no requirement for the pair of edited DNA strands (3′ flaps) to directly compete with 5′ flaps in endogenous genomic DNA, as the complementary edited strand is available for hybridization instead. Since both strands of the duplex are synthesized as edited DNA, the dual-flap prime editing system obviates the need for the replacement of the non-edited complementary DNA strand required by classical prime editing. Instead, cellular DNA repair machinery need only excise the paired 5′ flaps (original genomic DNA) and ligate the paired 3′ flaps (edited DNA) into the locus. Therefore, there is also no need to include sequences homologous to genomic DNA in the newly synthesized DNA strands, allowing selective hybridization of the new strands and facilitating edits that contain minimal genomic homology. Nuclease-active versions of prime editors that cut both strands of DNA could also be used to accelerate the removal of the original DNA sequence.


Like classical prime editing, multi-flap prime editing (including dual-flap and quadruple-flap prime editing) is a versatile and precise genome editing method that directly writes new genetic information into a specified DNA site using a nucleic acid programmable DNA binding protein (“napDNAbp”) working in association with a polymerase (i.e., in the form of a fusion protein or otherwise provided in trans with the napDNAbp), wherein the prime editing system is programmed with a prime editing (PE) guide RNA (“PEgRNA”) that both specifies the target site and templates the synthesis of the desired edit in the form of a replacement DNA strand by way of an extension (either DNA or RNA) engineered onto a guide RNA (e.g., at the 5′ or 3′ end, or at an internal portion of a guide RNA). The replacement strand containing the desired edit (e.g., a single nucleobase substitution) shares the same sequence as the endogenous strand of the target site to be edited (with the exception that it includes the desired edit). Through DNA repair and/or replication machinery, the endogenous strand of the target site is replaced by the newly synthesized replacement strand containing the desired edit. In some cases, prime editing may be thought of as a “search-and-replace” genome editing technology since the prime editors, as described herein, not only search and locate the desired target site to be edited, but at the same time, encode a replacement strand containing a desired edit which is installed in place of the corresponding target site endogenous DNA strand.


Prime Editor


The dual prime editing system described herein comprises a pair of prime editors. The term “prime editor” refers to the herein described fusion constructs comprising a napDNAbp (e.g., Cas9 nickase) and a reverse transcriptase and is capable of carrying out prime editing on a target nucleotide sequence in the presence of a PEgRNA (or “extended guide RNA”). The term “prime editor” may refer to the fusion protein or to the fusion protein complexed with a PEgRNA, and/or further complexed with a second-strand nicking sgRNA. In some embodiments, the prime editor may also refer to the complex comprising a fusion protein (reverse transcriptase fused to a napDNAbp), a PEgRNA, and a regular guide RNA capable of directing the second-site nicking step of the non-edited strand as described herein. In other embodiments, the reverse transcriptase component of the “primer editor” may be provided in trans.


The dual-flap prime editing system described herein comprises a pair of prime editors. The quadruple-flap prime editing system described herein comprises four prime editors.


Primer Binding Site


The term “primer binding site” or “the PBS” refers to the nucleotide sequence located on a PEgRNA as a component of the extension arm (typically at the 3′ end of the extension arm) and serves to bind to the primer sequence that is formed after Cas9 nicking of the target sequence by the prime editor. As detailed elsewhere, when the Cas9 nickase component of a prime editor nicks one strand of the target DNA sequence, a 3′-ended ssDNA flap is formed, which serves a primer sequence that anneals to the primer binding site on the PEgRNA to prime reverse transcription.


Promoter


The term “promoter” is art-recognized and refers to a nucleic acid molecule with a sequence recognized by the cellular transcription machinery and able to initiate transcription of a downstream gene. A promoter can be constitutively active, meaning that the promoter is always active in a given cellular context, or conditionally active, meaning that the promoter is only active in the presence of a specific condition. For example, a conditional promoter may only be active in the presence of a specific protein that connects a protein associated with a regulatory element in the promoter to the basic transcriptional machinery, or only in the absence of an inhibitory molecule. A subclass of conditionally active promoters are inducible promoters that require the presence of a small molecule “inducer” for activity. Examples of inducible promoters include, but are not limited to, arabinose-inducible promoters, Tet-on promoters, and tamoxifen-inducible promoters. A variety of constitutive, conditional, and inducible promoters are well known to the skilled artisan, and the skilled artisan will be able to ascertain a variety of such promoters useful in carrying out the instant invention, which is not limited in this respect.


Protospacer


As used herein, the term “protospacer” refers to the sequence (˜20 bp) in DNA adjacent to the PAM (protospacer adjacent motif) sequence. The protospacer shares the same sequence as the spacer sequence of the guide RNA. The guide RNA anneals to the complement of the protospacer sequence on the target DNA (specifically, one strand thereof, i.e., the “target strand” versus the “non-target strand” of the target DNA sequence). In order for Cas9 to function it also requires a specific protospacer adjacent motif (PAM) that varies depending on the bacterial species of the Cas9 gene. The most commonly used Cas9 nuclease, derived from S. pyogenes, recognizes a PAM sequence of NGG that is found directly downstream of the target sequence in the genomic DNA, on the non-target strand. The skilled person will appreciate that the literature in the state of the art sometimes refers to the “protospacer” as the ˜20-nt target-specific guide sequence on the guide RNA itself, rather than referring to it as a “spacer.” Thus, in some cases, the term “protospacer” as used herein may be used interchangeably with the term “spacer.” The context of the description surrounding the appearance of either “protospacer” or “spacer” will help inform the reader as to whether the term is in reference to the gRNA or the DNA target.


Protospacer Adjacent Motif (PAM)


As used herein, the term “protospacer adjacent sequence” or “PAM” refers to an approximately 2-6 base pair DNA sequence that is an important targeting component of a Cas9 nuclease. Typically, the PAM sequence is on either strand, and is downstream in the 5′ to 3′ direction of the Cas9 cut site. The canonical PAM sequence (i.e., the PAM sequence that is associated with the Cas9 nuclease of Streptococcus pyogenes or SpCas9) is 5′-NGG-3′ wherein “N” is any nucleobase followed by two guanine (“G”) nucleobases. Different PAM sequences can be associated with different Cas9 nucleases or equivalent proteins from different organisms. In addition, any given Cas9 nuclease, e.g., SpCas9, may be modified to alter the PAM specificity of the nuclease such that the nuclease recognizes alternative PAM sequence.


For example, with reference to the canonical SpCas9 amino acid sequence is SEQ ID NO: 18, the PAM sequence can be modified by introducing one or more mutations, including (a) D1135V, R1335Q, and T1337R “the VQR variant”, which alters the PAM specificity to NGAN or NGNG, (b) D1135E, R1335Q, and T1337R “the EQR variant”, which alters the PAM specificity to NGAG, and (c) D1135V, G1218R, R1335E, and T1337R “the VRER variant”, which alters the PAM specificity to NGCG. In addition, the D1135E variant of canonical SpCas9 still recognizes NGG, but it is more selective compared to the wild type SpCas9 protein.


It will also be appreciated that Cas9 enzymes from different bacterial species (i.e., Cas9 orthologs) can have varying PAM specificities. For example, Cas9 from Staphylococcus aureus (SaCas9) recognizes NGRRT or NGRRN. In addition, Cas9 from Neisseria meningitis (NmCas) recognizes NNNNGATT. In another example, Cas9 from Streptococcus thermophilis (StCas9) recognizes NNAGAAW. In still another example, Cas9 from Treponema denticola (TdCas) recognizes NAAAAC. These are examples and are not meant to be limiting. It will be further appreciated that non-SpCas9s bind a variety of PAM sequences, which makes them useful when no suitable SpCas9 PAM sequence is present at the desired target cut site. Furthermore, non-SpCas9s may have other characteristics that make them more useful than SpCas9. For example, Cas9 from Staphylococcus aureus (SaCas9) is about 1 kilobase smaller than SpCas9, so it can be packaged into adeno-associated virus (AAV). Further reference may be made to Shah et al., “Protospacer recognition motifs: mixed identities and functional diversity,” RNA Biology, 10(5): 891-899 (which is incorporated herein by reference).


Recombinase


The term “recombinase,” as used herein, refers to a site-specific enzyme that mediates the recombination of DNA between recombinase recognition sequences, which results in the excision, integration, inversion, or exchange (e.g., translocation) of DNA fragments between the recombinase recognition sequences. Recombinases can be classified into two distinct families serine recombinases (e.g., resolvases and invertases) and tyrosine recombinases (e.g., integrases). Examples of serine recombinases include, without limitation, Hin, Gin, Tn3, β-six, CinH, ParA, γδ, Bxb1, ϕC31, TP901, TG1, φBT1, R4, φRV1, φFC1, MR11, A118, U153, and gp29. Examples of tyrosine recombinases include, without limitation, Cre, FLP, R, Lambda, HK101, HK022, and pSAM2. The serine and tyrosine recombinase names stem from the conserved nucleophilic amino acid residue that the recombinase uses to attack the DNA and which becomes covalently linked to the DNA during strand exchange. Recombinases have numerous applications, including the creation of gene knockouts/knock-ins and gene therapy applications. See, e.g., Brown et al., “Serine recombinases as tools for genome engineering.” Methods. 2011; 53(4):372-9; Hirano et al., “Site-specific recombinases as tools for heterologous gene integration.” Appl. Microbiol. Biotechnol. 2011; 92(2):227-39; Chavez and Calos, “Therapeutic applications of the ΦC31 integrase system.” Curr. Gene Ther. 2011; 11(5):375-81; Turan and Bode, “Site-specific recombinases: from tag-and-target- to tag-and-exchange-based genomic modifications.” FASEB J. 2011; 25(12):4088-107; Venken and Bellen, “Genome-wide manipulations of Drosophila melanogaster with transposons, Flp recombinase, and ΦC31 integrase.” Methods Mol. Biol. 2012; 859:203-28; Murphy, “Phage recombinases and their applications.” Adv. Virus Res. 2012; 83:367-414; Zhang et al., “Conditional gene manipulation: Cre-ating a new biological era.” J. Zhejiang Univ. Sci. B. 2012; 13(7):511-24; Karpenshif and Bernstein, “From yeast to mammals recent advances in genetic control of homologous recombination.” DNA Repair (Amst). 2012; 1; 11(10):781-8; the entire contents of each are hereby incorporated by reference in their entirety. The recombinases provided herein are not meant to be exclusive examples of recombinases that can be used in embodiments of the invention. The methods and compositions of the invention can be expanded by mining databases for new orthogonal recombinases or designing synthetic recombinases with defined DNA specificities (See, e.g., Groth et al., “Phage integrases: biology and applications.” J. Mol. Biol. 2004; 335, 667-678; Gordley et al., “Synthesis of programmable integrases.” Proc. Natl. Acad. Sci. USA. 2009; 106, 5053-5058; the entire contents of each are hereby incorporated by reference in their entirety). Other examples of recombinases that are useful in the methods and compositions described herein are known to those of skill in the art, and any new recombinase that is discovered or generated is expected to be able to be used in the different embodiments of the invention. In some embodiments, the catalytic domains of a recombinase are fused to a nuclease-inactivated RNA-programmable nuclease (e.g., dCas9, or a fragment thereof), such that the recombinase domain does not comprise a nucleic acid binding domain or is unable to bind to a target nucleic acid (e.g., the recombinase domain is engineered such that it does not have specific DNA binding activity). Recombinases lacking DNA binding activity and methods for engineering such are known, and include those described by Klippel et al., “Isolation and characterisation of unusual gin mutants.” EMBO J. 1988; 7: 3983-3989: Burke et al., “Activating mutations of Tn3 resolvase marking interfaces important in recombination catalysis and its regulation. Mol Microbiol. 2004; 51: 937-948; Olorunniji et al., “Synapsis and catalysis by activated Tn3 resolvase mutants.” Nucleic Acids Res. 2008; 36: 7181-7191; Rowland et al., “Regulatory mutations in Sin recombinase support a structure-based model of the synaptosome.” Mol Microbiol. 2009; 74: 282-298; Akopian et al., “Chimeric recombinases with designed DNA sequence recognition.” Proc Natl Acad Sci USA. 2003; 100: 8688-8691; Gordley et al., “Evolution of programmable zinc finger-recombinases with activity in human cells. J Mol Biol. 2007; 367: 802-813; Gordley et al., “Synthesis of programmable integrases.” Proc Natl Acad Sci USA. 2009; 106: 5053-5058; Arnold et al., “Mutants of Tn3 resolvase which do not require accessory binding sites for recombination activity.” EMBO J. 1999; 18: 1407-1414; Gaj et al., “Structure-guided reprogramming of serine recombinase DNA sequence specificity.” Proc Natl Acad Sci USA. 2011; 108(2):498-503; and Proudfoot et al., “Zinc finger recombinases with adaptable DNA sequence specificity.” PLoS One. 2011; 6(4):e19537; the entire contents of each are hereby incorporated by reference. For example, serine recombinases of the resolvase-invertase group, e.g., Tn3 and γδ resolvases and the Hin and Gin invertases, have modular structures with autonomous catalytic and DNA-binding domains (See, e.g., Grindley et al., “Mechanism of site-specific recombination.” Ann Rev Biochem. 2006; 75: 567-605, the entire contents of which are incorporated by reference). The catalytic domains of these recombinases are thus amenable to being recombined with nuclease-inactivated RNA-programmable nucleases (e.g., dCas9, or a fragment thereof) as described herein, e.g., following the isolation of ‘activated’ recombinase mutants which do not require any accessory factors (e.g., DNA binding activities) (See, e.g., Klippel et al., “Isolation and characterisation of unusual gin mutants.” EMBO J. 1988; 7: 3983-3989: Burke et al., “Activating mutations of Tn3 resolvase marking interfaces important in recombination catalysis and its regulation. Mol Microbiol. 2004; 51: 937-948; Olorunniji et al., “Synapsis and catalysis by activated Tn3 resolvase mutants.” Nucleic Acids Res. 2008; 36: 7181-7191; Rowland et al., “Regulatory mutations in Sin recombinase support a structure-based model of the synaptosome.” Mol Microbiol. 2009; 74: 282-298; Akopian et al., “Chimeric recombinases with designed DNA sequence recognition.” Proc Natl Acad Sci USA. 2003; 100: 8688-8691). Additionally, many other natural serine recombinases having an N-terminal catalytic domain and a C-terminal DNA binding domain are known (e.g., phiC31 integrase, TnpX transposase, IS607 transposase), and their catalytic domains can be co-opted to engineer programmable site-specific recombinases as described herein (See, e.g., Smith et al., “Diversity in the serine recombinases.” Mol Microbiol. 2002; 44: 299-307, the entire contents of which are incorporated by reference). Similarly, the core catalytic domains of tyrosine recombinases (e.g., Cre, λ integrase) are known, and can be similarly co-opted to engineer programmable site-specific recombinases as described herein (See, e.g., Guo et al., “Structure of Cre recombinase complexed with DNA in a site-specific recombination synapse.” Nature. 1997; 389:40-46; Hartung et al., “Cre mutants with altered DNA binding properties.” J Biol Chem 1998; 273:22884-22891; Shaikh et al., “Chimeras of the Flp and Cre recombinases: Tests of the mode of cleavage by Flp and Cre. J Mol Biol. 2000; 302:27-48; Rongrong et al., “Effect of deletion mutation on the recombination activity of Cre recombinase.” Acta Biochim Pol. 2005; 52:541-544; Kilbride et al., “Determinants of product topology in a hybrid Cre-Tn3 resolvase site-specific recombination system.” J Mol Biol. 2006; 355:185-195; Warren et al., “A chimeric cre recombinase with regulated directionality.” Proc Natl Acad Sci USA. 2008 105:18278-18283; Van Duyne, “Teaching Cre to follow directions.” Proc Natl Acad Sci USA. 2009 Jan. 6; 106(1):4-5; Numrych et al., “A comparison of the effects of single-base and triple-base changes in the integrase arm-type binding sites on the site-specific recombination of bacteriophage λ.” Nucleic Acids Res. 1990; 18:3953-3959; Tirumalai et al., “The recognition of core-type DNA sites by λ integrase.” J Mol Biol. 1998; 279:513-527; Aihara et al., “A conformational switch controls the DNA cleavage activity of λ integrase.” Mol Cell. 2003; 12:187-198; Biswas et al., “A structural basis for allosteric control of DNA recombination by λ integrase.” Nature. 2005; 435:1059-1066; and Warren et al., “Mutations in the amino-terminal domain of λ-integrase have differential effects on integrative and excisive recombination.” Mol Microbiol. 2005; 55:1104-1112; the entire contents of each are incorporated by reference).


Recombinase Recognition Sequence


The term “recombinase recognition sequence”, or equivalently as “RRS” or “recombinase target sequence” or “recombinase site,” as used herein, refers to a nucleotide sequence target recognized by a recombinase and which undergoes strand exchange with another DNA molecule having a the RRS that results in excision, integration, inversion, or exchange of DNA fragments between the recombinase recognition sequences. In various embodiments, the multi-strand prime editors may install one or more recombinase sites in a target sequence, or in more than one target sequence. When more than one recombinase site is installed by a multi-strand prime editor, the recombinase sites can be installed at adjacent target sites or non-adjacent target sites (e.g., separate chromosomes). In various embodiments, single installed recombinase sites can be used as “landing sites” for a recombinase-mediated reaction between the genomic recombinase site and a second recombinase site within an exogenously supplied nucleic acid molecule, e.g., a plasmid. This enables the targeted integration of a desired nucleic acid molecule. In other embodiments, where two recombinase sites are inserted in adjacent regions of DNA (e.g., separated by 25-50 bp, 50-100 bp, 100-200 bp, 200-300 bp, 300-400 bp, 400-500 bp, 500-600 bp, 600-700 bp, 700-800 bp, 800-900 bp, 900-1000 bp, 1000-2000 bp, 2000-3000 bp, 3000-4000 bp, 4000-5000 bp, or more), the recombinase sites can be used for recombinase-mediated excision or inversion of the intervening sequence, or for recombinase-mediated cassette exchange with exogenous DNA having the same recombinase sites. When the two or more recombinase sites are installed by multi-flap prime editors on two different chromosomes, translocation of the intervening sequence can occur from a first chromosomal location to the second.


Recombine or Recombination


The term “recombine,” or “recombination,” in the context of a nucleic acid modification (e.g., a genomic modification), is used to refer to the process by which two or more nucleic acid molecules, or two or more regions of a single nucleic acid molecule, are modified by the action of a recombinase protein (e.g., an inventive recombinase fusion protein provided herein). Recombination can result in, inter alia, the insertion, inversion, excision, or translocation of nucleic acids, e.g., in or between one or more nucleic acid molecules.


Reverse Transcriptase


The term “reverse transcriptase” describes a class of polymerases characterized as RNA-dependent DNA polymerases. All known reverse transcriptases require a primer to synthesize a DNA transcript from an RNA template. Historically, reverse transcriptase has been used primarily to transcribe mRNA into cDNA which can then be cloned into a vector for further manipulation. Avian myoblastosis virus (AMV) reverse transcriptase was the first widely used RNA-dependent DNA polymerase (Verma, Biochim. Biophys. Acta 473:1 (1977)). The enzyme has 5′-3′ RNA-directed DNA polymerase activity, 5′-3′ DNA-directed DNA polymerase activity, and RNase H activity. RNase H is a processive 5′ and 3′ ribonuclease specific for the RNA strand for RNA-DNA hybrids (Perbal, A Practical Guide to Molecular Cloning, New York: Wiley & Sons (1984)). Errors in transcription cannot be corrected by reverse transcriptase because known viral reverse transcriptases lack the 3′-5′ exonuclease activity necessary for proofreading (Saunders and Saunders, Microbial Genetics Applied to Biotechnology, London: Croom Helm (1987)). A detailed study of the activity of AMV reverse transcriptase and its associated RNase H activity has been presented by Berger et al., Biochemistry 22:2365-2372 (1983). Another reverse transcriptase which is used extensively in molecular biology is reverse transcriptase originating from Moloney murine leukemia virus (M-MLV). See, e.g., Gerard, G. R., DNA 5:271-279 (1986) and Kotewicz, M. L., et al., Gene 35:249-258 (1985). M-MLV reverse transcriptase substantially lacking in RNase H activity has also been described. See, e.g., U.S. Pat. No. 5,244,797. The invention contemplates the use of any such reverse transcriptases, or variants or mutants thereof.


In addition, the invention contemplates the use of reverse transcriptases that are error-prone, i.e., that may be referred to as error-prone reverse transcriptases or reverse transcriptases that do not support high fidelity incorporation of nucleotides during polymerization. During synthesis of the single-strand DNA flap based on the RT template integrated with the guide RNA, the error-prone reverse transcriptase can introduce one or more nucleotides which are mismatched with the RT template sequence, thereby introducing changes to the nucleotide sequence through erroneous polymerization of the single-strand DNA flap. These errors introduced during synthesis of the single strand DNA flap then become integrated into the double strand molecule through hybridization to the corresponding endogenous target strand, removal of the endogenous displaced strand, ligation, and then through one more round of endogenous DNA repair and/or sequencing processes.


Reverse Transcription


As used herein, the term “reverse transcription” indicates the capability of an enzyme to synthesize a DNA strand (that is, complementary DNA or cDNA) using RNA as a template. In some embodiments, the reverse transcription can be “error-prone reverse transcription,” which refers to the properties of certain reverse transcriptase enzymes which are error-prone in their DNA polymerization activity.


PACE


The term “phage-assisted continuous evolution (PACE),” as used herein, refers to continuous evolution that employs phage as viral vectors. The general concept of PACE technology has been described, for example, in International PCT Application, PCT/US2009/056194, filed Sep. 8, 2009, published as WO 2010/028347 on Mar. 11, 2010; International PCT Application, PCT/US2011/066747, filed Dec. 22, 2011, published as WO 2012/088381 on Jun. 28, 2012; U.S. Application, U.S. Pat. No. 9,023,594, issued May 5, 2015, International PCT Application, PCT/US2015/012022, filed Jan. 20, 2015, published as WO 2015/134121 on Sep. 11, 2015, and International PCT Application, PCT/US2016/027795, filed Apr. 15, 2016, published as WO 2016/168631 on Oct. 20, 2016, the entire contents of each of which are incorporated herein by reference.


Phage


The term “phage,” as used herein interchangeably with the term “bacteriophage,” refers to a virus that infects bacterial cells. Typically, phages consist of an outer protein capsid enclosing genetic material. The genetic material can be ssRNA, dsRNA, ssDNA, or dsDNA, in either linear or circular form. Phages and phage vectors are well known to those of skill in the art and non-limiting examples of phages that are useful for carrying out the PACE methods provided herein are λ (Lysogen), T2, T4, T7, T12, R17, M13, MS2, G4, P1, P2, P4, Phi X174, N4, Φ6, and Φ29. In certain embodiments, the phage utilized in the present invention is M13. Additional suitable phages and host cells will be apparent to those of skill in the art and the invention is not limited in this aspect. For an exemplary description of additional suitable phages and host cells, see Elizabeth Kutter and Alexander Sulakvelidze: Bacteriophages: Biology and Applications. CRC Press; 1st edition (December 2004), ISBN: 0849313368; Martha R. J. Clokie and Andrew M. Kropinski: Bacteriophages: Methods and Protocols, Volume 1: Isolation, Characterization, and Interactions (Methods in Molecular Biology) Humana Press; 1st edition (December, 2008), ISBN: 1588296822; Martha R. J. Clokie and Andrew M. Kropinski: Bacteriophages: Methods and Protocols, Volume 2: Molecular and Applied Aspects (Methods in Molecular Biology) Humana Press; 1st edition (December 2008), ISBN: 1603275649; all of which are incorporated herein in their entirety by reference for disclosure of suitable phages and host cells as well as methods and protocols for isolation, culture, and manipulation of such phages).


Protein, Peptide, and Polypeptide


The terms “protein,” “peptide,” and “polypeptide” are used interchangeably herein, and refer to a polymer of amino acid residues linked together by peptide (amide) bonds. The terms refer to a protein, peptide, or polypeptide of any size, structure, or function. Typically, a protein, peptide, or polypeptide will be at least three amino acids long. A protein, peptide, or polypeptide may refer to an individual protein or a collection of proteins. One or more of the amino acids in a protein, peptide, or polypeptide may be modified, for example, by the addition of a chemical entity such as a carbohydrate group, a hydroxyl group, a phosphate group, a farnesyl group, an isofarnesyl group, a fatty acid group, a linker for conjugation, functionalization, or other modification, etc. A protein, peptide, or polypeptide may also be a single molecule or may be a multi-molecular complex. A protein, peptide, or polypeptide may be just a fragment of a naturally occurring protein or peptide. A protein, peptide, or polypeptide may be naturally occurring, recombinant, or synthetic, or any combination thereof. Any of the proteins provided herein may be produced by any method known in the art. For example, the proteins provided herein may be produced via recombinant protein expression and purification, which is especially suited for fusion proteins comprising a peptide linker. Methods for recombinant protein expression and purification are well known, and include those described by Green and Sambrook, Molecular Cloning: A Laboratory Manual (4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2012)), the entire contents of which are incorporated herein by reference.


Protein Splicing


The term “protein splicing,” as used herein, refers to a process in which a sequence, an intein (or split inteins, as the case may be), is excised from within an amino acid sequence, and the remaining fragments of the amino acid sequence, the exteins, are ligated via an amide bond to form a continuous amino acid sequence. The term “trans” protein splicing refers to the specific case where the inteins are split inteins and they are located on different proteins.


Second-Strand Nicking


The resolution of heteroduplex DNA (i.e., containing one edited and one non-edited strand) formed as a result of prime editing determines long-term editing outcomes. In words, a goal of prime editing is to resolve the heteroduplex DNA (the edited strand paired with the endogenous non-edited strand) formed as an intermediate of PE by permanently integrating the edited strand into the complement, endogenous strand. The approach of “second-strand nicking” can be used herein to help drive the resolution of heteroduplex DNA in favor of permanent integration of the edited strand into the DNA molecule. As used herein, the concept of “second-strand nicking” refers to the introduction of a second nick at a location downstream of the first nick (i.e., the initial nick site that provides the free 3′ end for use in priming of the reverse transcriptase on the extended portion of the guide RNA), preferably on the unedited strand. In certain embodiments, the first nick and the second nick are on opposite strands. In other embodiments, the first nick and the second nick are on opposite strands. In yet another embodiment, the first nick is on the non-target strand (i.e., the strand that forms the single strand portion of the R-loop), and the second nick is on the target strand. In still other embodiments, the first nick is on the edited strand, and the second nick is on the unedited strand. The second nick can be positioned at least 5 nucleotides downstream of the first nick, or at least 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, or 150 or more nucleotides downstream of the first nick. The second nick, in certain embodiments, can be introduced between about 5-150 nucleotides on the unedited strand away from the site of the PEgRNA-induced nick, or between about 5-140, or between about 5-130, or between about 5-120, or between about 5-110, or between about 5-100, or between about 5-90, or between about 5-80, or between about 5-70, or between about 5-60, or between about 5-50, or between about 5-40, or between about 5-30, or between about 5-20, or between about 5-10. In one embodiment, the second nick is introduced between 14-116 nucleotides away from the PEgRNA-induced nick. Without being bound by theory, the second nick induces the cell's endogenous DNA repair and replication processes towards replacement or editing of the unedited strand, thereby permanently installing the edited sequence on both strands and resolving the heteroduplex that is formed as a result of PE. In some embodiments, the edited strand is the non-target strand and the unedited strand is the target strand. In other embodiments, the edited strand is the target strand, and the unedited strand is the non-target strand.


Sense Strand


In genetics, a “sense” strand is the segment within double-stranded DNA that runs from 5′ to 3′, and which is complementary to the antisense strand of DNA, or template strand, which runs from 3′ to 5′. In the case of a DNA segment that encodes a protein, the sense strand is the strand of DNA that has the same sequence as the mRNA, which takes the antisense strand as its template during transcription, and eventually undergoes (typically, not always) translation into a protein. The antisense strand is thus responsible for the RNA that is later translated to protein, while the sense strand possesses a nearly identical makeup to that of the mRNA. Note that for each segment of dsDNA, there will possibly be two sets of sense and antisense, depending on which direction one reads (since sense and antisense is relative to perspective). It is ultimately the gene product, or mRNA, that dictates which strand of one segment of dsDNA is referred to as sense or antisense.


In the context of a PEgRNA, the first step is the synthesis of a single-strand complementary DNA (i.e., the 3′ ssDNA flap, which becomes incorporated) oriented in the 5′ to 3′ direction which is templated off of the PEgRNA extension arm. Whether the 3′ ssDNA flap should be regarded as a sense or antisense strand depends on the direction of transcription since it well accepted that both strands of DNA may serve as a template for transcription (but not at the same time). Thus, in some embodiments, the 3′ ssDNA flap (which overall runs in the 5′ to 3′ direction) will serve as the sense strand because it is the coding strand. In other embodiments, the 3′ ssDNA flap (which overall runs in the 5′ to 3′ direction) will serve as the antisense strand and thus, the template for transcription.


Sequence Homology


A “homologous sequence” or a sequence exhibiting “homology” to another sequence means a sequence of a nucleic acid molecule exhibiting at least about 65%, 70%, 75%, 80%, 85%, or 90% sequence identity to another nucleic acid molecule. In other embodiments, a “homologous sequence” of nucleic acids may exhibit 93%, 95% or 98% sequence identity to the reference nucleic acid.


When a percentage of sequence homology or identity is specified, in the context of two nucleic acid sequences or two polypeptide sequences, the percentage of homology or identity generally refers to the alignment of two or more sequences across a portion of their length when, compared and aligned for maximum correspondence. Unless stated otherwise, sequence homology or identity is assessed over the specified length of the nucleic acid, polypeptide, or portion thereof. In some embodiments, the homology or identity is assessed over a functional portion or specified portion of the length.


Alignment of sequences for assessment of sequence homology can be conducted by algorithms known in the art, such as the Basic Local Alignment Search Tool (BLAST) algorithm, which is described in Altschul et al, J. Mol. Biol. 215:403-410, 1990. A publicly available internet interface for performing BLAST analyses is accessible through the National Center for Biotechnology Information. Additional known algorithms include those published in: Smith & Waterman, “Comparison of Biosequences”, Adv. Appl. Math. 2:482, 1981; Needleman & Wunsch, “A general method applicable to the search for similarities in the amino acid sequence of two proteins” J. Mol. Biol. 48:443, 1970; Pearson & Lipman “Improved tools for biological sequence comparison”, Proc. Natl. Acad. Sci. USA 85:2444, 1988; or by automated implementation of these or similar algorithms. Global alignment programs may also be used to align similar sequences of roughly equal size. Examples of global alignment programs include NEEDLE (available at www.ebi.ac.uk/Tools/psa/emboss_needle/) which is part of the EMBOSS package (Rice P et al., Trends Genet., 2000; 16: 276-277), and the GGSEARCH program fasta.bioch. Virginia. edu/fasta_www2/fasta_www.cgi?rm=compare&pgm=gnw), which is part of the FASTA package (Pearson W and Lipman D, 1988, Proc. Natl. Acad. Sci. USA, 85: 2444-2448). Both of these programs are based on the Needleman-Wunsch algorithm which is used to find the optimum alignment (including gaps) of two sequences along their entire length. A detailed discussion of sequence analysis can also be found in Unit 19.3 of Ausubel et al (“Current Protocols in Molecular Biology” John Wiley & Sons Inc, 1994-1998, Chapter 15, 1998).


Spacer Sequence


As used herein, the term “spacer sequence” in connection with a guide RNA or a PEgRNA refers to the portion of the guide RNA or PEgRNA of about 20 nucleotides which contains a nucleotide sequence that shares the same sequence as the protospacer sequence in the target DNA sequence. The spacer sequence anneals to the complement of the protospacer sequence to form a ssRNA/ssDNA hybrid structure at the target site and a corresponding R loop ssDNA structure of the endogenous DNA strand.


Subject


The term “subject,” as used herein, refers to an individual organism, for example, an individual mammal. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human mammal. In some embodiments, the subject is a non-human primate. In some embodiments, the subject is a rodent. In some embodiments, the subject is a sheep, a goat, a cattle, a cat, or a dog. In some embodiments, the subject is a vertebrate, an amphibian, a reptile, a fish, an insect, a fly, or a nematode. In some embodiments, the subject is a research animal. In some embodiments, the subject is genetically engineered, e.g., a genetically engineered non-human subject. The subject may be of either sex and at any stage of development.


Split Intein


Although inteins are most frequently found as a contiguous domain, some exist in a naturally split form. In this case, the two fragments are expressed as separate polypeptides and must associate before splicing takes place, so-called protein trans-splicing.


An exemplary split intein is the Ssp DnaE intein, which comprises two subunits, namely, DnaE-N and DnaE-C. The two different subunits are encoded by separate genes, namely dnaE-n and dnaE-c, which encode the DnaE-N and DnaE-C subunits, respectively. DnaE is a naturally occurring split intein in Synechocytis sp. PCC6803 and is capable of directing trans-splicing of two separate proteins, each comprising a fusion with either DnaE-N or DnaE-C.


Additional naturally occurring or engineered split-intein sequences are known in the or can be made from whole-intein sequences described herein or those available in the art. Examples of split-intein sequences can be found in Stevens et al., “A promiscuous split intein with expanded protein engineering applications,” PNAS, 2017, Vol. 114: 8538-8543; Iwai et al., “Highly efficient protein trans-splicing by a naturally split DnaE intein from Nostc punctiforme, FEBS Lett, 580: 1853-1858, each of which are incorporated herein by reference. Additional split intein sequences can be found, for example, in WO 2013/045632, WO 2014/055782, WO 2016/069774, and EP2877490, the contents each of which are incorporated herein by reference.


In addition, protein splicing in trans has been described in vivo and in vitro (Shingledecker, et al., Gene 207:187 (1998), Southworth, et al., EMBO J. 17:918 (1998); Mills, et al., Proc. Natl. Acad. Sci. USA, 95:3543-3548 (1998); Lew, et al., J. Biol. Chem., 273:15887-15890 (1998); Wu, et al., Biochim. Biophys. Acta 35732:1 (1998b), Yamazaki, et al., J. Am. Chem. Soc. 120:5591 (1998), Evans, et al., J. Biol. Chem. 275:9091 (2000); Otomo, et al., Biochemistry 38:16040-16044 (1999); Otomo, et al., J. Biolmol. NMR 14:105-114 (1999); Scott, et al., Proc. Natl. Acad. Sci. USA 96:13638-13643 (1999)) and provides the opportunity to express a protein as to two inactive fragments that subsequently undergo ligation to form a functional product, e.g., as shown in FIGS. 66 and 67 with regard to the formation of a complete PE fusion protein from two separately-expressed halves.


Target Site


The term “target site” refers to a sequence within a nucleic acid molecule that is edited by a prime editor (PE) disclosed herein. The target site further refers to the sequence within a nucleic acid molecule to which a complex of the prime editor (PE) and gRNA binds.


tPERT


See definition for “trans prime editor RNA template (tPERT).”


Temporal Second-Strand Nicking


As used herein, the term “temporal second-strand nicking” refers to a variant of second strand nicking whereby the installation of the second nick in the unedited strand occurs only after the desired edit is installed in the edited strand. This avoids concurrent nicks on both strands that could lead to double-stranded DNA breaks. The second-strand nicking guide RNA is designed for temporal control such that the second strand nick is not introduced until after the installation of the desired edit. This is achieved by designing a gRNA with a spacer sequence that matches only the edited strand, but not the original allele. Using this strategy, mismatches between the protospacer and the unedited allele should disfavor nicking by the sgRNA until after the editing event on the PAM strand takes place.


Trans Prime Editing


As used herein, the term “trans prime editing” or similarly, “trans dual prime editing,” refers to a modified form of prime editing or dual prime editing that utilizes a split PEgRNA, i.e., wherein the PEgRNA is separated into two separate molecules: an sgRNA and a trans prime editing RNA template (tPERT). The sgRNA serves to target the prime editor (or more generally, to target the napDNAbp component of the prime editor) to the desired genomic target site, while the tPERT is used by the polymerase (e.g., a reverse transcriptase) to write new DNA sequence into the target locus once the tPERT is recruited in trans to the prime editor by the interaction of binding domains located on the prime editor and on the tPERT. In one embodiment, the binding domains can include RNA-protein recruitment moieties, such as a MS2 aptamer located on the tPERT and an MS2cp protein fused to the prime editor. An advantage of trans prime editing is that by separating the DNA synthesis template from the guide RNA, one can potentially use longer length templates.


An embodiment of trans prime editing is shown in FIGS. 3G and 3H. FIG. 3G shows the composition of the trans prime editor complex on the left (“RP-PE:gRNA complex), which comprises an napDNAbp fused to each of a polymerase (e.g., a reverse transcriptase) and a rPERT recruiting protein (e.g., MS2sc), and which is complexed with a guide RNA. FIG. 3G further shows a separate tPERT molecule, which comprises the extension arm features of a PEgRNA, including the DNA synthesis template and the primer binding sequence. The tPERT molecule also includes an RNA-protein recruitment domain (which, in this case, is a stem loop structure and can be, for example, MS2 aptamer). As depicted in the process described in FIG. 3H, the RP-PE:gRNA complex binds to and nicks the target DNA sequence. Then, the recruiting protein (RP) recruits a tPERT to co-localize to the prime editor complex bound to the DNA target site, thereby allowing the primer binding site to bind to the primer sequence on the nicked strand, and subsequently, allowing the polymerase (e.g., RT) to synthesize a single strand of DNA against the DNA synthesis template up through the 5′ of the tPERT.


While the tPERT is shown in FIG. 3G and FIG. 3H as comprising the PBS and DNA synthesis template on the 5′ end of the RNA-protein recruitment domain, the tPERT in other configurations may be designed with the PBS and DNA synthesis template located on the 3′ end of the RNA-protein recruitment domain. However, the tPERT with the 5′ extension has the advantage that synthesis of the single strand of DNA will naturally terminate at the 5′ end of the tPERT and thus, does not risk using any portion of the RNA-protein recruitment domain as a template during the DNA synthesis stage of prime editing.


Trans Prime Editor RNA Template (tPERT)


As used herein, a “trans prime editor RNA template (tPERT)” refers to a component used in trans prime editing or trans dual prime editing, a modified version of prime editing which operates by separating the PEgRNA into two distinct molecules: a guide RNA and a tPERT molecule. The tPERT molecule is programmed to co-localize with the prime editor complex at a target DNA site, bringing the primer binding site and the DNA synthesis template to the prime editor in trans. For example, see FIG. 3G for an embodiment of a trans prime editor (tPE) which shows a two-component system comprising (1) an RP-PE:gRNA complex and (2) a tPERT that includes the primer binding site and the DNA synthesis template joined to an RNA-protein recruitment domain, wherein the RP (recruiting protein) component of the RP-PE:gRNA complex recruits the tPERT to a target site to be edited, thereby associating the PBS and DNA synthesis template with the prime editor in trans. Said another way, the tPERT is engineered to contain (all or part of) the extension arm of a PEgRNA, which includes the primer binding site and the DNA synthesis template.


Transitions


As used herein, “transitions” refer to the interchange of purine nucleobases (A↔G) or the interchange of pyrimidine nucleobases (C↔T). This class of interchanges involves nucleobases of similar shape. The compositions and methods disclosed herein are capable of inducing one or more transitions in a target DNA molecule. The compositions and methods disclosed herein are also capable of inducing both transitions and transversion in the same target DNA molecule. These changes involve A↔G, G↔A, C↔T, or T↔C. In the context of a double-strand DNA with Watson-Crick paired nucleobases, transversions refer to the following base pair exchanges: A:T↔G:C, G:G↔A:T, C:G↔T:A, or C:G. The compositions and methods disclosed herein are capable of inducing one or more transitions in a target DNA molecule. The compositions and methods disclosed herein are also capable of inducing both transitions and transversion in the same target DNA molecule, as well as other nucleotide changes, including deletions and insertions.


Transversions


As used herein, “transversions” refer to the interchange of purine nucleobases for pyrimidine nucleobases, or in the reverse and thus, involve the interchange of nucleobases with dissimilar shape. These changes involve T↔A, T↔G, C↔G, C↔A, A↔T, A↔C, G↔C, and G↔T. In the context of a double-strand DNA with Watson-Crick paired nucleobases, transversions refer to the following base pair exchanges: T:A↔A:T, T:A↔G:C, C:G↔G:C, C:G↔A:T, A:T↔T:A, A:T↔C:G, G:C↔C:G, and G:C↔T:A. The compositions and methods disclosed herein are capable of inducing one or more transversions in a target DNA molecule. The compositions and methods disclosed herein are also capable of inducing both transitions and transversion in the same target DNA molecule, as well as other nucleotide changes, including deletions and insertions.


Treatment


The terms “treatment,” “treat,” and “treating,” refer to a clinical intervention aimed to reverse, alleviate, delay the onset of, or inhibit the progress of a disease or disorder, or one or more symptoms thereof, as described herein. As used herein, the terms “treatment,” “treat,” and “treating” refer to a clinical intervention aimed to reverse, alleviate, delay the onset of, or inhibit the progress of a disease or disorder, or one or more symptoms thereof, as described herein. In some embodiments, treatment may be administered after one or more symptoms have developed and/or after a disease has been diagnosed. In other embodiments, treatment may be administered in the absence of symptoms, e.g., to prevent or delay onset of a symptom or inhibit onset or progression of a disease. For example, treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example, to prevent or delay their recurrence.


Trinucleotide Repeat Disorder


As used herein, a “trinucleotide repeat disorder” (or alternatively, “expansion repeat disorder” or “repeat expansion disorder”) refers to a set of genetic disorders which are cause by “trinucleotide repeat expansion,” which is a kind of mutation where a certain trinucleotide repeats in certain genes or introns. Trinucleotide repeats were once thought to be commonplace iterations in the genome, but the 1990s clarified these disorders. These apparently ‘benign’ stretches of DNA can sometimes expand and cause disease. Several defining features are shared amongst disorders caused by trinucleotide repeat expansions. First, the mutant repeats show both somatic and germline instability and, more frequently, they expand rather than contract in successive transmissions. Secondly, an earlier age of onset and increasing severity of phenotype in subsequent generations (anticipation) generally are correlated with larger repeat length. Finally, the parental origin of the disease allele can often influence anticipation, with paternal transmissions carrying a greater risk of expansion for many of these disorders.


Triplet expansion is thought to be caused by slippage during DNA replication. Due to the repetitive nature of the DNA sequence in these regions ‘loop out’ structures may form during DNA replication while maintaining complementary base pairing between the parent strand and daughter strand being synthesized. If the loop out structure is formed from sequence on the daughter strand this will result in an increase in the number of repeats. However, if the loop out structure is formed on the parent strand a decrease in the number of repeats occurs. It appears that expansion of these repeats is more common than reduction. Generally the larger the expansion the more likely they are to cause disease or increase the severity of disease. This property results in the characteristic of anticipation seen in trinucleotide repeat disorders. Anticipation describes the tendency of age of onset to decrease and severity of symptoms to increase through successive generations of an affected family due to the expansion of these repeats.


Nucleotide repeat disorders may include those in which the triplet repeat occurs in a non-coding region (i.e., a non-coding trinucleotide repeat disorder) or in a coding region


The prime editor (PE) system described herein may use to treat nucleotide repeat disorders, which may include fragile X syndrome (FRAXA), fragile XE MR (FRAXE), Freidreich ataxia (FRDA), myotonic dystrophy (DM), spinocerebellar ataxia type 8 (SCAB), and spinocerebellar ataxia type 12 (SCA12), among others.


Upstream


As used herein, the terms “upstream” and “downstream” are terms of relativity that define the linear position of at least two elements located in a nucleic acid molecule (whether single or double-stranded) that is orientated in a 5′-to-3′ direction. In particular, a first element is upstream of a second element in a nucleic acid molecule where the first element is positioned somewhere that is 5′ to the second element. For example, a SNP is upstream of a Cas9-induced nick site if the SNP is on the 5′ side of the nick site. Conversely, a first element is downstream of a second element in a nucleic acid molecule where the first element is positioned somewhere that is 3′ to the second element. For example, a SNP is downstream of a Cas9-induced nick site if the SNP is on the 3′ side of the nick site. The nucleic acid molecule can be a DNA (double or single stranded). RNA (double or single stranded), or a hybrid of DNA and RNA. The analysis is the same for single strand nucleic acid molecule and a double strand molecule since the terms upstream and downstream are in reference to only a single strand of a nucleic acid molecule, except that one needs to select which strand of the double stranded molecule is being considered. Often, the strand of a double stranded DNA which can be used to determine the positional relativity of at least two elements is the “sense” or “coding” strand. In genetics, a “sense” strand is the segment within double-stranded DNA that runs from 5′ to 3′, and which is complementary to the antisense strand of DNA, or template strand, which runs from 3′ to 5′. Thus, as an example, a SNP nucleobase is “downstream” of a promoter sequence in a genomic DNA (which is double-stranded) if the SNP nucleobase is on the 3′ side of the promoter on the sense or coding strand.


Variant


As used herein the term “variant” should be taken to mean the exhibition of qualities that have a pattern that deviates from what occurs in nature, e.g., a variant Cas9 is a Cas9 comprising one or more changes in amino acid residues as compared to a wild type Cas9 amino acid sequence. The term “variant” encompasses homologous proteins having at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or at least 99% percent identity with a reference sequence and having the same or substantially the same functional activity or activities as the reference sequence. The term also encompasses mutants, truncations, or domains of a reference sequence, and which display the same or substantially the same functional activity or activities as the reference sequence.


Vector


The term “vector,” as used herein, refers to a nucleic acid that can be modified to encode a gene of interest and that is able to enter into a host cell, mutate and replicate within the host cell, and then transfer a replicated form of the vector into another host cell. Exemplary suitable vectors include viral vectors, such as retroviral vectors or bacteriophages and filamentous phage, and conjugative plasmids. Additional suitable vectors will be apparent to those of skill in the art based on the instant disclosure.


Wild Type


As used herein the term “wild type” is a term of the art understood by skilled persons and means the typical form of an organism, strain, gene or characteristic as it occurs in nature as distinguished from mutant or variant forms.


5′ Endogenous DNA Flap


As used herein, the term “5′ endogenous DNA flap” refers to the strand of DNA situated immediately downstream of the PE-induced nick site in the target DNA. The nicking of the target DNA strand by PE exposes a 3′ hydroxyl group on the upstream side of the nick site and a 5′ hydroxyl group on the downstream side of the nick site. The endogenous strand ending in the 3′ hydroxyl group is used to prime the DNA polymerase of the prime editor (e.g., wherein the DNA polymerase is a reverse transcriptase). The endogenous strand on the downstream side of the nick site and which begins with the exposed 5′ hydroxyl group is referred to as the “5′ endogenous DNA flap” and is ultimately removed and replaced by the newly synthesized replacement strand (i.e., “3′ replacement DNA flap”) the encoded by the extension of the PEgRNA.


5′ Endogenous DNA Flap Removal


As used herein, the term “5′ endogenous DNA flap removal” or “5′ flap removal” refers to the removal of the 5′ endogenous DNA flap that forms when the RT-synthesized single-strand DNA flap competitively invades and hybridizes to the endogenous DNA, displacing the endogenous strand in the process. Removing this endogenous displaced strand can drive the reaction towards the formation of the desired product comprising the desired nucleotide change. The cell's own DNA repair enzymes may catalyze the removal or excision of the 5′ endogenous flap (e.g., a flap endonuclease, such as EXO1 or FEN1). Also, host cells may be transformed to express one or more enzymes that catalyze the removal of said 5′ endogenous flaps, thereby driving the process toward product formation (e.g., a flap endonuclease). Flap endonucleases are known in the art and can be found described in Patel et al., “Flap endonucleases pass 5′-flaps through a flexible arch using a disorder-thread-order mechanism to confer specificity for free 5′-ends,” Nucleic Acids Research, 2012, 40(10): 4507-4519 and Tsutakawa et al., “Human flap endonuclease structures, DNA double-base flipping, and a unified understanding of the FEN1 superfamily,” Cell, 2011, 145(2): 198-211 (each of which are incorporated herein by reference).


3′ Replacement DNA Flap


As used herein, the term “3′ replacement DNA flap” or simply, “replacement DNA flap,” refers to the strand of DNA that is synthesized by the prime editor and which is encoded by the extension arm of the prime editor PEgRNA. More in particular, the 3′ replacement DNA flap is encoded by the polymerase template of the PEgRNA. The 3′ replacement DNA flap comprises the same sequence as the 5′ endogenous DNA flap except that it also contains the edited sequence (e.g., single nucleotide change). The 3′ replacement DNA flap anneals to the target DNA, displacing or replacing the 5′ endogenous DNA flap (which can be excised, for example, by a 5′ flap endonuclease, such as FEN1 or EXO1) and then is ligated to join the 3′ end of the 3′ replacement DNA flap to the exposed 5′ hydoxyl end of endogenous DNA (exposed after excision of the 5′ endogenous DNA flap, thereby reforming a phosophodiester bond and installing the 3′ replacement DNA flap to form a heteroduplex DNA containing one edited strand and one unedited strand. DNA repair processes resolve the heteroduplex by copying the information in the edited strand to the complementary strand permanently installs the edit in to the DNA. This resolution process can be driven further to completion by nicking the unedited strand, i.e., by way of “second-strand nicking,” as described herein.


DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS

Adoption of the clustered regularly interspaced short palindromic repeat (CRISPR) system for genome editing has revolutionized the life sciences1-3. Although gene disruption using CRISPR is now routine, the precise installation of single nucleotide edits remains a major challenge, despite being necessary for studying or correcting a large number of disease-causative mutations. Homology directed repair (HDR) is capable of achieving such edits, but suffers from low efficiency (often <5%), a requirement for donor DNA repair templates, and deleterious effects of double-stranded DNA break (DSB) formation. Recently, Prof. David Liu et al.'s laboratory developed base editing, which achieves efficient single nucleotide editing without DSBs. Base editors (BEs) combine the CRISPR system with base-modifying deaminase enzymes to convert target C•G or A•T base pairs to A•T or G•C, respectively4-6. Although already widely used by researchers worldwide, current BEs enable only four of the twelve possible base pair conversions and are unable to correct small insertions or deletions. Moreover, the targeting scope of base editing is limited by the editing of non-target C or A bases adjacent to the target base (“bystander editing”) and by the requirement that a PAM sequence exist 15±2 bp from the target base. Overcoming these limitations would therefore greatly broaden the basic research and therapeutic applications of genome editing.


New precision editing approaches (e.g., classical prime editing) have been developed that offer many of the benefits of base editing—namely, avoidance of double strand breaks and donor DNA repair templates—while overcoming its major limitations. The proposed approach described herein achieves the direct installation of edited DNA strands at target genomic sites using target-primed reverse transcription (TPRT). In the design discussed herein, CRISPR guide RNA (gRNA) will be engineered to carry a reverse transcriptase (RT) template sequence encoding a single-stranded DNA comprising a desired nucleotide change. The CRISPR nuclease (Cas9)-nicked target site DNA will serve as the primer for reverse transcription of the template sequence on the modified gRNA, allowing for direct incorporation of any desired nucleotide edit.


The mechanism of target-primed reverse transcription (TPRT) can be leveraged or adapted for conducting precision CRISPR/Cas-based genome editing with high efficiency and genetic flexibility (e.g., as depicted in various embodiments of FIGS. 1A-1F). Cas protein-reverse transcriptase fusions are used to target a specific DNA sequence with a modified guide RNA (“an extended guide RNA”), generate a single strand nick at the target site, and use the nicked DNA as a primer for reverse transcription of an engineered reverse transcriptase template that is integrated into the extended guide RNA. The newly synthesized strand would be homologous to the genomic target sequence except for the inclusion of a desired nucleotide change (e.g., a single nucleotide change, a deletion, or an insertion, or a combination thereof). The newly synthesize strand of DNA may be referred to as a single strand DNA flap, which would compete for hybridization with the complementary homologous endogenous DNA strand, thereby displacing the corresponding endogenous strand. Resolution of this hybridized intermediate can include removal of the resulting displaced flap of endogenous DNA (e.g., with a 5′ end DNA flap endonuclease, FEN1), ligation of the synthesized single strand DNA flap to the target DNA, and assimilation of the desired nucleotide change as a result of cellular DNA repair and/or replication processes. Because templated DNA synthesis offers single nucleotide precision, the scope of this approach is very broad and could foreseeably be used for myriad applications in basic science and therapeutics.


The present invention describes a new platform for genome editing called “multi-flap prime editing” (including, for example, “dual-flap prime editing” and “quadruple-flap prime editing”) and represents an innovative advancement of “prime editing” or “classical prime editing,” as described by the present inventors in Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149-157 (2019), which is incorporated herein by reference. Whereas classical prime editing in various embodiments polymerizes at a nick site a single 3′ flap which becomes integrated into the target nucleic acid on the same strand, the presently described multi-flap prime editing systems involve distinct constructs, systems, and methodologies that, in various embodiments, generate pairs or multiple pairs of 3′ flaps on different strands, which form duplexes comprising desired edits and which become incorporated into target nucleic acid molecules, e.g., at specific loci or edit sites in a genome. In various aspects, the pairs or multiple pairs of 3′ flaps form duplexes because they comprise reverse complementary sequences which anneal to one another once generated by the prime editors described herein. The duplexes become incorporated into the target site by cell-driven mechanisms that naturally replace the endogenous duplex sequences located between adjacent nick sites. In certain embodiments, the new duplex sequences may be introduced at one or more locations (e.g., at adjacent genomic loci or on two different chromosomal locations), and may comprise one or more sequences of interest, e.g., protein-encoding sequence, peptide-encoding sequence, or RNA-encoding sequence. In one embodiment, the new duplex sequences installed by the multi-flap prime editing systems may comprise a recombinase site, e.g., a Bxb1 recombinase attB (38 bp) and/or attP (50 bp) site, or a recombinase site recognized by Hin recombinase, Gin recombinase, Tn3 recombinase, β-six recombinase, CinH recombinase, ParA recombinase, γδ recombinase, ϕC31 recombinase, TP901 recombinase, TG1 recombinase, φBT1 recombinase, R4 recombinase, φRV1 recombinase, φFC1 recombinase, MR11 recombinase, A118 recombinase, U153 recombinase, and gp29 recombinase, Cre recombinase, FLP recombinase, R recombinase, Lambda recombinase, HK101 recombinase, HK022 recombinase, and pSAM2 recombinase.


In various aspects, this Specification describes a multi-flap prime editing system (including, for example, dual prime editing systems and quadruple prime editing systems) that addresses the challenges associated with flap equilibration and subsequent incorporation of the edit into the non-edited complementary genomic DNA strand by simultaneously editing both DNA strands. In the dual-flap prime editing system, for example, two PEgRNAs are used to target opposite strands of a genomic site and direct the synthesis of two complementary 3′ flaps containing edited DNA sequence (FIG. 90). Unlike classical prime editing, there is no requirement for the pair of edited DNA strands (3′ flaps) to directly compete with 5′ flaps in endogenous genomic DNA, as the complementary edited strand is available for hybridization instead. Since both strands of the duplex are synthesized as edited DNA, the dual-flap prime editing system obviates the need for the replacement of the non-edited complementary DNA strand required by classical prime editing. Instead, cellular DNA repair machinery need only excise the paired 5′ flaps (original genomic DNA) and ligate the paired 3′ flaps (edited DNA) into the locus. Therefore, there is also no need to include sequences homologous to genomic DNA in the newly synthesized DNA strands, allowing selective hybridization of the new strands and facilitating edits that contain minimal genomic homology. Nuclease-active versions of prime editors that cut both strands of DNA could also be used to accelerate the removal of the original DNA sequence. The quadruple-flap prime editing system, using four PEgRNAs, provides similar advantages.


[1] napDNAbp

The multi-flap prime editors described herein may comprise a nucleic acid programmable DNA binding protein (napDNAbp).


In one aspect, a napDNAbp can be associated with or complexed with at least one guide nucleic acid (e.g., guide RNA or a PEgRNA), which localizes the napDNAbp to a DNA sequence that comprises a DNA strand (i.e., a target strand) that is complementary to the guide nucleic acid, or a portion thereof (e.g., the spacer of a guide RNA which anneals to the protospacer of the DNA target). In other words, the guide nucleic-acid “programs” the napDNAbp (e.g., Cas9 or equivalent) to localize and bind to complementary sequence of the protospacer in the DNA.


Any suitable napDNAbp may be used in the multi-flap prime editors described herein. In various embodiments, the napDNAbp may be any Class 2 CRISPR-Cas system, including any type II, type V, or type VI CRISPR-Cas enzyme. Given the rapid development of CRISPR-Cas as a tool for genome editing, there have been constant developments in the nomenclature used to describe and/or identify CRISPR-Cas enzymes, such as Cas9 and Cas9 orthologs. This application references CRISPR-Cas enzymes with nomenclature that may be old and/or new. The skilled person will be able to identify the specific CRISPR-Cas enzyme being referenced in this Application based on the nomenclature that is used, whether it is old (i.e., “legacy”) or new nomenclature. CRISPR-Cas nomenclature is extensively discussed in Makarova et al., “Classification and Nomenclature of CRISPR-Cas Systems: Where from Here?,” The CRISPR Journal, Vol. 1. No. 5, 2018, the entire contents of which are incorporated herein by reference. The particular CRISPR-Cas nomenclature used in any given instance in this Application is not limiting in any way and the skilled person will be able to identify which CRISPR-Cas enzyme is being referenced.


For example, the following type II, type V, and type VI Class 2 CRISPR-Cas enzymes have the following art-recognized old (i.e., legacy) and new names. Each of these enzymes, and/or variants thereof, may be used with the multi-flap prime editors described herein:
















Legacy nomenclature
Current nomenclature*
















type II CRISPR-Cas enzymes










Cas9
same







type V CRISPR-Cas enzymes










Cpf1
Cas12a



CasX
Cas12e



C2c1
Cas12b1



Cas12b2
same



C2c3
Cas12c



CasY
Cas12d



C2c4
same



C2c8
same



C2c5
same



C2c10
same



C2c9
same







type VI CRISPR-Cas enzymes










C2c2
Cas13a



Cas13d
same



C2c7
Cas13c



C2c6
Cas13b







*See Makarova et al., The CRISPR Journal, Vol. 1, No. 5, 2018






Without being bound by theory, the mechanism of action of certain napDNAbp contemplated herein includes the step of forming an R-loop whereby the napDNAbp induces the unwinding of a double-strand DNA target, thereby separating the strands in the region bound by the napDNAbp. The guide RNA spacer then hybridizes to the “target strand” at the protospacer sequence. This displaces a “non-target strand” that is complementary to the target strand, which forms the single strand region of the R-loop. In some embodiments, the napDNAbp includes one or more nuclease activities, which then cut the DNA leaving various types of lesions. For example, the napDNAbp may comprises a nuclease activity that cuts the non-target strand at a first location, and/or cuts the target strand at a second location. Depending on the nuclease activity, the target DNA can be cut to form a “double-stranded break” whereby both strands are cut. In other embodiments, the target DNA can be cut at only a single site, i.e., the DNA is “nicked” on one strand. Exemplary napDNAbp with different nuclease activities include “Cas9 nickase” (“nCas9”) and a deactivated Cas9 having no nuclease activities (“dead Cas9” or “dCas9”).


The below description of various napDNAbps which can be used in connection with the presently disclosed multi-flap prime editors is not meant to be limiting in any way. The multi-flap prime editors may comprise the canonical SpCas9, or any ortholog Cas9 protein, or any variant Cas9 protein—including any naturally occurring variant, mutant, or otherwise engineered version of Cas9—that is known or that can be made or evolved through a directed evolutionary or otherwise mutagenic process. In various embodiments, the Cas9 or Cas9 variants have a nickase activity, i.e., only cleave one strand of the target DNA sequence. In other embodiments, the Cas9 or Cas9 variants have inactive nucleases, i.e., are “dead” Cas9 proteins. Other variant Cas9 proteins that may be used are those having a smaller molecular weight than the canonical SpCas9 (e.g., for easier delivery) or having modified or rearranged primary amino acid structure (e.g., the circular permutant formats).


The multi-flap prime editors described herein may also comprise Cas9 equivalents, including Cas12a (Cpf1) and Cas12b1 proteins which are the result of convergent evolution. The napDNAbps used herein (e.g., SpCas9, Cas9 variant, or Cas9 equivalents) may also contain various modifications that alter/enhance their PAM specifities. Lastly, the application contemplates any Cas9, Cas9 variant, or Cas9 equivalent which has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.9% sequence identity to a reference Cas9 sequence, such as a reference SpCas9 canonical sequence or a reference Cas9 equivalent (e.g., Cas12a (Cpf1)).


The napDNAbp can be a CRISPR (clustered regularly interspaced short palindromic repeat)-associated nuclease. As outlined above, CRISPR is an adaptive immune system that provides protection against mobile genetic elements (viruses, transposable elements and conjugative plasmids). CRISPR clusters contain spacers, sequences complementary to antecedent mobile elements, and target invading nucleic acids. CRISPR clusters are transcribed and processed into CRISPR RNA (crRNA). In type II CRISPR systems, correct processing of pre-crRNA requires a trans-encoded small RNA (tracrRNA), endogenous ribonuclease 3 (mc), and a Cas9 protein. The tracrRNA serves as a guide for ribonuclease 3-aided processing of pre-crRNA. Subsequently, Cas9/crRNA/tracrRNA endonucleolytically cleaves a linear or circular dsDNA target complementary to the spacer. The target strand not complementary to crRNA is first cut endonucleolytically, then trimmed 3′-5′ exonucleolytically. In nature, DNA-binding and cleavage typically requires protein and both RNAs. However, single guide RNAs (“sgRNA”, or simply “gRNA”) can be engineered so as to incorporate aspects of both the crRNA and tracrRNA into a single RNA species. See, e.g., Jinek M. et al., Science 337:816-821(2012), the entire contents of which is hereby incorporated by reference.


In some embodiments, the napDNAbp directs cleavage of one or both strands at the location of a target sequence, such as within the target sequence and/or within the complement of the target sequence. In some embodiments, the napDNAbp directs cleavage of one or both strands within about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 50, 100, 200, 500, or more base pairs from the first or last nucleotide of a target sequence. In some embodiments, a vector encodes a napDNAbp that is mutated to with respect to a corresponding wild-type enzyme such that the mutated napDNAbp lacks the ability to cleave one or both strands of a target polynucleotide containing a target sequence. For example, an aspartate-to-alanine substitution (D10A) in the RuvC I catalytic domain of Cas9 from S. pyogenes converts Cas9 from a nuclease that cleaves both strands to a nickase (cleaves a single strand). Other examples of mutations that render Cas9 a nickase include, without limitation, H840A, N854A, and N863A in reference to the canonical SpCas9 sequence, or to equivalent amino acid positions in other Cas9 variants or Cas9 equivalents.


As used herein, the term “Cas protein” refers to a full-length Cas protein obtained from nature, a recombinant Cas protein having a sequences that differs from a naturally occurring Cas protein, or any fragment of a Cas protein that nevertheless retains all or a significant amount of the requisite basic functions needed for the disclosed methods, i.e., (i) possession of nucleic-acid programmable binding of the Cas protein to a target DNA, and (ii) ability to nick the target DNA sequence on one strand. The Cas proteins contemplated herein embrace CRISPR Cas 9 proteins, as well as Cas9 equivalents, variants (e.g., Cas9 nickase (nCas9) or nuclease inactive Cas9 (dCas9)) homologs, orthologs, or paralogs, whether naturally occurring or non-naturally occurring (e.g., engineered or recombinant), and may include a Cas9 equivalent from any Class 2 CRISPR system (e.g., type II, V, VI), including Cas12a (Cpf1), Cas12e (CasX), Cas12b1 (C2c1), Cas12b2, Cas12c (C2c3), C2c4, C2c8, C2c5, C2c10, C2c9 Cas13a (C2c2), Cas13d, Cas13c (C2c7), Cas13b (C2c6), and Cas13b. Further Cas-equivalents are described in Makarova et al., “C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector,” Science 2016; 353(6299) and Makarova et al., “Classification and Nomenclature of CRISPR-Cas Systems: Where from Here?,” The CRISPR Journal, Vol. 1. No. 5, 2018, the contents of which are incorporated herein by reference.


The terms “Cas9” or “Cas9 nuclease” or “Cas9 moiety” or “Cas9 domain” embrace any naturally occurring Cas9 from any organism, any naturally-occurring Cas9 equivalent or functional fragment thereof, any Cas9 homolog, ortholog, or paralog from any organism, and any mutant or variant of a Cas9, naturally-occurring or engineered. The term Cas9 is not meant to be particularly limiting and may be referred to as a “Cas9 or equivalent.” Exemplary Cas9 proteins are further described herein and/or are described in the art and are incorporated herein by reference. The present disclosure is unlimited with regard to the particular Cas9 that is employed in the multi-flap prime editors described herein.


As noted herein, Cas9 nuclease sequences and structures are well known to those of skill in the art (see, e.g., “Complete genome sequence of an M1 strain of Streptococcus pyogenes.” Ferretti et al., J. J., McShan W. M., Ajdic D. J., Savic D. J., Savic G., Lyon K., Primeaux C., Sezate S., Suvorov A. N., Kenton S., Lai H. S., Lin S. P., Qian Y., Jia H. G., Najar F. Z., Ren Q., Zhu H., Song L., White J., Yuan X., Clifton S. W., Roe B. A., McLaughlin R. E., Proc. Natl. Acad. Sci. U.S.A. 98:4658-4663(2001); “CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III.” Deltcheva E., Chylinski K., Sharma C. M., Gonzales K., Chao Y., Pirzada Z. A., Eckert M. R., Vogel J., Charpentier E., Nature 471:602-607(2011); and “A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity.” Jinek M., Chylinski K., Fonfara I., Hauer M., Doudna J. A., Charpentier E. Science 337:816-821(2012), the entire contents of each of which are incorporated herein by reference).


Examples of Cas9 and Cas9 equivalents are provided as follows; however, these specific examples are not meant to be limiting. The multi-flap prime editors of the present disclosure may use any suitable napDNAbp, including any suitable Cas9 or Cas9 equivalent.


A. Wild Type Canonical SpCas9


In one embodiment, the multi-flap prime editor constructs described herein may comprise the “canonical SpCas9” nuclease from S. pyogenes, which has been widely used as a tool for genome engineering and is categorized as the type II subgroup of enzymes of the Class 2 CRISPR-Cas systems. This Cas9 protein is a large, multi-domain protein containing two distinct nuclease domains. Point mutations can be introduced into Cas9 to abolish one or both nuclease activities, resulting in a nickase Cas9 (nCas9) or dead Cas9 (dCas9), respectively, that still retains its ability to bind DNA in a sgRNA-programmed manner. In principle, when fused to another protein or domain, Cas9, or a variant thereof (e.g., nCas9) can target that protein to virtually any DNA sequence simply by co-expression with an appropriate sgRNA. As used herein, the canonical SpCas9 protein refers to the wild type protein from Streptococcus pyogenes having the following amino acid sequence:














Description
Sequence
SEQ ID NO:







SpCas9
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDR
SEQ ID NO: 18



Streptococcus

HSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYL




pyogenes

QEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVD



M1
EVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFR



SwissProt
GHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVD



Accession
AKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTP



No. Q99ZW2
NFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLF



Wild type
LAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDL




TLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFY




KFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHL




GELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGN




SRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDK




NLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLS




GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGV




EDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLF




EDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKL




INGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQ




KAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKV




MGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKEL




GSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDIN




RLSDYDVDHIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPS




EEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELD




KAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREV




KVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVV




GTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATA




KYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKG




RDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDK




LIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLK




SVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHY




EKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADA




NLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYF




DTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






SpCas9
ATGGATAAAAAATATAGCATTGGCCTGGATATTGGCACC
SEQ ID NO: 19


Reverse
AACAGCGTGGGCTGGGCGGTGATTACCGATGAATATAAA



translation of
GTGCCGAGCAAAAAATTTAAAGTGCTGGGCAACACCGAT



SwissProt
CGCCATAGCATTAAAAAAAACCTGATTGGCGCGCTGCTG



Accession
TTTGATAGCGGCGAAACCGCGGAAGCGACCCGCCTGAAA



No. Q99ZW2
CGCACCGCGCGCCGCCGCTATACCCGCCGCAAAAACCGC




Streptococcus

ATTTGCTATCTGCAGGAAATTTTTAGCAACGAAATGGCGA




pyogenes

AAGTGGATGATAGCTTTTTTCATCGCCTGGAAGAAAGCTT




TCTGGTGGAAGAAGATAAAAAACATGAACGCCATCCGAT




TTTTGGCAACATTGTGGATGAAGTGGCGTATCATGAAAA




ATATCCGACCATTTATCATCTGCGCAAAAAACTGGTGGAT




AGCACCGATAAAGCGGATCTGCGCCTGATTTATCTGGCG




CTGGCGCATATGATTAAATTTCGCGGCCATTTTCTGATTG




AAGGCGATCTGAACCCGGATAACAGCGATGTGGATAAAC




TGTTTATTCAGCTGGTGCAGACCTATAACCAGCTGTTTGA




AGAAAACCCGATTAACGCGAGCGGCGTGGATGCGAAAGC




GATTCTGAGCGCGCGCCTGAGCAAAAGCCGCCGCCTGGA




AAACCTGATTGCGCAGCTGCCGGGCGAAAAAAAAAACGG




CCTGTTTGGCAACCTGATTGCGCTGAGCCTGGGCCTGACC




CCGAACTTTAAAAGCAACTTTGATCTGGCGGAAGATGCG




AAACTGCAGCTGAGCAAAGATACCTATGATGATGATCTG




GATAACCTGCTGGCGCAGATTGGCGATCAGTATGCGGAT




CTGTTTCTGGCGGCGAAAAACCTGAGCGATGCGATTCTGC




TGAGCGATATTCTGCGCGTGAACACCGAAATTACCAAAG




CGCCGCTGAGCGCGAGCATGATTAAACGCTATGATGAAC




ATCATCAGGATCTGACCCTGCTGAAAGCGCTGGTGCGCC




AGCAGCTGCCGGAAAAATATAAAGAAATTTTTTTTGATC




AGAGCAAAAACGGCTATGCGGGCTATATTGATGGCGGCG




CGAGCCAGGAAGAATTTTATAAATTTATTAAACCGATTCT




GGAAAAAATGGATGGCACCGAAGAACTGCTGGTGAAACT




GAACCGCGAAGATCTGCTGCGCAAACAGCGCACCTTTGA




TAACGGCAGCATTCCGCATCAGATTCATCTGGGCGAACT




GCATGCGATTCTGCGCCGCCAGGAAGATTTTTATCCGTTT




CTGAAAGATAACCGCGAAAAAATTGAAAAAATTCTGACC




TTTCGCATTCCGTATTATGTGGGCCCGCTGGCGCGCGGCA




ACAGCCGCTTTGCGTGGATGACCCGCAAAAGCGAAGAAA




CCATTACCCCGTGGAACTTTGAAGAAGTGGTGGATAAAG




GCGCGAGCGCGCAGAGCTTTATTGAACGCATGACCAACT




TTGATAAAAACCTGCCGAACGAAAAAGTGCTGCCGAAAC




ATAGCCTGCTGTATGAATATTTTACCGTGTATAACGAACT




GACCAAAGTGAAATATGTGACCGAAGGCATGCGCAAACC




GGCGTTTCTGAGCGGCGAACAGAAAAAAGCGATTGTGGA




TCTGCTGTTTAAAACCAACCGCAAAGTGACCGTGAAACA




GCTGAAAGAAGATTATTTTAAAAAAATTGAATGCTTTGAT




AGCGTGGAAATTAGCGGCGTGGAAGATCGCTTTAACGCG




AGCCTGGGCACCTATCATGATCTGCTGAAAATTATTAAAG




ATAAAGATTTTCTGGATAACGAAGAAAACGAAGATATTC




TGGAAGATATTGTGCTGACCCTGACCCTGTTTGAAGATCG




CGAAATGATTGAAGAACGCCTGAAAACCTATGCGCATCT




GTTTGATGATAAAGTGATGAAACAGCTGAAACGCCGCCG




CTATACCGGCTGGGGCCGCCTGAGCCGCAAACTGATTAA




CGGCATTCGCGATAAACAGAGCGGCAAAACCATTCTGGA




TTTTCTGAAAAGCGATGGCTTTGCGAACCGCAACTTTATG




CAGCTGATTCATGATGATAGCCTGACCTTTAAAGAAGAT




ATTCAGAAAGCGCAGGTGAGCGGCCAGGGCGATAGCCTG




CATGAACATATTGCGAACCTGGCGGGCAGCCCGGCGATT




AAAAAAGGCATTCTGCAGACCGTGAAAGTGGTGGATGAA




CTGGTGAAAGTGATGGGCCGCCATAAACCGGAAAACATT




GTGATTGAAATGGCGCGCGAAAACCAGACCACCCAGAAA




GGCCAGAAAAACAGCCGCGAACGCATGAAACGCATTGA




AGAAGGCATTAAAGAACTGGGCAGCCAGATTCTGAAAGA




ACATCCGGTGGAAAACACCCAGCTGCAGAACGAAAAACT




GTATCTGTATTATCTGCAGAACGGCCGCGATATGTATGTG




GATCAGGAACTGGATATTAACCGCCTGAGCGATTATGAT




GTGGATCATATTGTGCCGCAGAGCTTTCTGAAAGATGATA




GCATTGATAACAAAGTGCTGACCCGCAGCGATAAAAACC




GCGGCAAAAGCGATAACGTGCCGAGCGAAGAAGTGGTG




AAAAAAATGAAAAACTATTGGCGCCAGCTGCTGAACGCG




AAACTGATTACCCAGCGCAAATTTGATAACCTGACCAAA




GCGGAACGCGGCGGCCTGAGCGAACTGGATAAAGCGGG




CTTTATTAAACGCCAGCTGGTGGAAACCCGCCAGATTACC




AAACATGTGGCGCAGATTCTGGATAGCCGCATGAACACC




AAATATGATGAAAACGATAAACTGATTCGCGAAGTGAAA




GTGATTACCCTGAAAAGCAAACTGGTGAGCGATTTTCGC




AAAGATTTTCAGTTTTATAAAGTGCGCGAAATTAACAACT




ATCATCATGCGCATGATGCGTATCTGAACGCGGTGGTGG




GCACCGCGCTGATTAAAAAATATCCGAAACTGGAAAGCG




AATTTGTGTATGGCGATTATAAAGTGTATGATGTGCGCAA




AATGATTGCGAAAAGCGAACAGGAAATTGGCAAAGCGA




CCGCGAAATATTTTTTTTATAGCAACATTATGAACTTTTTT




AAAACCGAAATTACCCTGGCGAACGGCGAAATTCGCAAA




CGCCCGCTGATTGAAACCAACGGCGAAACCGGCGAAATT




GTGTGGGATAAAGGCCGCGATTTTGCGACCGTGCGCAAA




GTGCTGAGCATGCCGCAGGTGAACATTGTGAAAAAAACC




GAAGTGCAGACCGGCGGCTTTAGCAAAGAAAGCATTCTG




CCGAAACGCAACAGCGATAAACTGATTGCGCGCAAAAAA




GATTGGGATCCGAAAAAATATGGCGGCTTTGATAGCCCG




ACCGTGGCGTATAGCGTGCTGGTGGTGGCGAAAGTGGAA




AAAGGCAAAAGCAAAAAACTGAAAAGCGTGAAAGAACT




GCTGGGCATTACCATTATGGAACGCAGCAGCTTTGAAAA




AAACCCGATTGATTTTCTGGAAGCGAAAGGCTATAAAGA




AGTGAAAAAAGATCTGATTATTAAACTGCCGAAATATAG




CCTGTTTGAACTGGAAAACGGCCGCAAACGCATGCTGGC




GAGCGCGGGCGAACTGCAGAAAGGCAACGAACTGGCGC




TGCCGAGCAAATATGTGAACTTTCTGTATCTGGCGAGCCA




TTATGAAAAACTGAAAGGCAGCCCGGAAGATAACGAACA




GAAACAGCTGTTTGTGGAACAGCATAAACATTATCTGGA




TGAAATTATTGAACAGATTAGCGAATTTAGCAAACGCGT




GATTCTGGCGGATGCGAACCTGGATAAAGTGCTGAGCGC




GTATAACAAACATCGCGATAAACCGATTCGCGAACAGGC




GGAAAACATTATTCATCTGTTTACCCTGACCAACCTGGGC




GCGCCGGCGGCGTTTAAATATTTTGATACCACCATTGATC




GCAAACGCTATACCAGCACCAAAGAAGTGCTGGATGCGA




CCCTGATTCATCAGAGCATTACCGGCCTGTATGAAACCCG




CATTGATCTGAGCCAGCTGGGCGGCGAT









The multi-flap prime editors described herein may include canonical SpCas9, or any variant thereof having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity with a wild type Cas9 sequence provided above. These variants may include SpCas9 variants containing one or more mutations, including any known mutation reported with the SwissProt Accession No. Q99ZW2 (SEQ ID NO: 18) entry, which include:













SpCas9 mutation (relative to



the amino acid sequence of the
Function/Characteristic (as reported) (see


canonical SpCas9 sequence,
UniProtKB-Q99ZW2 (CAS9_STRPT1)


SEQ ID NO: 18)
entry-incorporated herein by reference)







D10A
Nickase mutant which cleaves the



protospacer strand (but no cleavage of



non-protospacer strand)


S15A
Decreased DNA cleavage activity


R66A
Decreased DNA cleavage activity


R70A
No DNA cleavage


R74A
Decreased DNA cleavage


R78A
Decreased DNA cleavage


  97-150 deletion
No nuclease activity


R165A
Decreased DNA cleavage


 175-307 deletion
About 50% decreased DNA cleavage


 312-409 deletion
No nuclease activity


E762A
Nickase


H840A
Nickase mutant which cleaves the



non-protospacer strand but does not



cleave the protospacer strand


N854A
Nickase


N863A
Nickase


H982A
Decreased DNA cleavage


D986A
Nickase


1099-1368 deletion
No nuclease activity


R1333A
Reduced DNA binding









Other wild type SpCas9 sequences that may be used in the present disclosure, include:














Description
Sequence
SEQ ID NO:







SpCas9
ATGGATAAGAAATACTCAATAGGCTTAGATATCGGCACAAAT
SEQ ID NO: 20



Streptococcus

AGCGTCGGATGGGCGGTGATCACTGATGATTATAAGGTTCCG




pyogenes

TCTAAAAAGTTCAAGGTTCTGGGAAATACAGACCGCCACAGT



MGAS1882
ATCAAAAAAAATCTTATAGGGGCTCTTTTATTTGGCAGTGGAG



wild type
AGACAGCGGAAGCGACTCGTCTCAAACGGACAGCTCGTAGAA



NC_017053.1
GGTATACACGTCGGAAGAATCGTATTTGTTATCTACAGGAGAT




TTTTTCAAATGAGATGGCGAAAGTAGATGATAGTTTCTTTCAT




CGACTTGAAGAGTCTTTTTTGGTGGAAGAAGACAAGAAGCAT




GAACGTCATCCTATTTTTGGAAATATAGTAGATGAAGTTGCTT




ATCATGAGAAATATCCAACTATCTATCATCTGCGAAAAAAATT




GGCAGATTCTACTGATAAAGCGGATTTGCGCTTAATCTATTTG




GCCTTAGCGCATATGATTAAGTTTCGTGGTCATTTTTTGATTG




AGGGAGATTTAAATCCTGATAATAGTGATGTGGACAAACTAT




TTATCCAGTTGGTACAAATCTACAATCAATTATTTGAAGAAAA




CCCTATTAACGCAAGTAGAGTAGATGCTAAAGCGATTCTTTCT




GCACGATTGAGTAAATCAAGACGATTAGAAAATCTCATTGCT




CAGCTCCCCGGTGAGAAGAGAAATGGCTTGTTTGGGAATCTC




ATTGCTTTGTCATTGGGATTGACCCCTAATTTTAAATCAAATTT




TGATTTGGCAGAAGATGCTAAATTACAGCTTTCAAAAGATACT




TACGATGATGATTTAGATAATTTATTGGCGCAAATTGGAGATC




AATATGCTGATTTGTTTTTGGCAGCTAAGAATTTATCAGATGC




TATTTTACTTTCAGATATCCTAAGAGTAAATAGTGAAATAACT




AAGGCTCCCCTATCAGCTTCAATGATTAAGCGCTACGATGAAC




ATCATCAAGACTTGACTCTTTTAAAAGCTTTAGTTCGACAACA




ACTTCCAGAAAAGTATAAAGAAATCTTTTTTGATCAATCAAAA




AACGGATATGCAGGTTATATTGATGGGGGAGCTAGCCAAGAA




GAATTTTATAAATTTATCAAACCAATTTTAGAAAAAATGGATG




GTACTGAGGAATTATTGGTGAAACTAAATCGTGAAGATTTGCT




GCGCAAGCAACGGACCTTTGACAACGGCTCTATTCCCCATCA




AATTCACTTGGGTGAGCTGCATGCTATTTTGAGAAGACAAGA




AGACTTTTATCCATTTTTAAAAGACAATCGTGAGAAGATTGAA




AAAATCTTGACTTTTCGAATTCCTTATTATGTTGGTCCATTGGC




GCGTGGCAATAGTCGTTTTGCATGGATGACTCGGAAGTCTGA




AGAAACAATTACCCCATGGAATTTTGAAGAAGTTGTCGATAA




AGGTGCTTCAGCTCAATCATTTATTGAACGCATGACAAACTTT




GATAAAAATCTTCCAAATGAAAAAGTACTACCAAAACATAGT




TTGCTTTATGAGTATTTTACGGTTTATAACGAATTGACAAAGG




TCAAATATGTTACTGAGGGAATGCGAAAACCAGCATTTCTTTC




AGGTGAACAGAAGAAAGCCATTGTTGATTTACTCTTCAAAAC




AAATCGAAAAGTAACCGTTAAGCAATTAAAAGAAGATTATTT




CAAAAAAATAGAATGTTTTGATAGTGTTGAAATTTCAGGAGTT




GAAGATAGATTTAATGCTTCATTAGGCGCCTACCATGATTTGC




TAAAAATTATTAAAGATAAAGATTTTTTGGATAATGAAGAAA




ATGAAGATATCTTAGAGGATATTGTTTTAACATTGACCTTATT




TGAAGATAGGGGGATGATTGAGGAAAGACTTAAAACATATGC




TCACCTCTTTGATGATAAGGTGATGAAACAGCTTAAACGTCGC




CGTTATACTGGTTGGGGACGTTTGTCTCGAAAATTGATTAATG




GTATTAGGGATAAGCAATCTGGCAAAACAATATTAGATTTTTT




GAAATCAGATGGTTTTGCCAATCGCAATTTTATGCAGCTGATC




CATGATGATAGTTTGACATTTAAAGAAGATATTCAAAAAGCA




CAGGTGTCTGGACAAGGCCATAGTTTACATGAACAGATTGCT




AACTTAGCTGGCAGTCCTGCTATTAAAAAAGGTATTTTACAGA




CTGTAAAAATTGTTGATGAACTGGTCAAAGTAATGGGGCATA




AGCCAGAAAATATCGTTATTGAAATGGCACGTGAAAATCAGA




CAACTCAAAAGGGCCAGAAAAATTCGCGAGAGCGTATGAAAC




GAATCGAAGAAGGTATCAAAGAATTAGGAAGTCAGATTCTTA




AAGAGCATCCTGTTGAAAATACTCAATTGCAAAATGAAAAGC




TCTATCTCTATTATCTACAAAATGGAAGAGACATGTATGTGGA




CCAAGAATTAGATATTAATCGTTTAAGTGATTATGATGTCGAT




CACATTGTTCCACAAAGTTTCATTAAAGACGATTCAATAGACA




ATAAGGTACTAACGCGTTCTGATAAAAATCGTGGTAAATCGG




ATAACGTTCCAAGTGAAGAAGTAGTCAAAAAGATGAAAAACT




ATTGGAGACAACTTCTAAACGCCAAGTTAATCACTCAACGTA




AGTTTGATAATTTAACGAAAGCTGAACGTGGAGGTTTGAGTG




AACTTGATAAAGCTGGTTTTATCAAACGCCAATTGGTTGAAAC




TCGCCAAATCACTAAGCATGTGGCACAAATTTTGGATAGTCGC




ATGAATACTAAATACGATGAAAATGATAAACTTATTCGAGAG




GTTAAAGTGATTACCTTAAAATCTAAATTAGTTTCTGACTTCC




GAAAAGATTTCCAATTCTATAAAGTACGTGAGATTAACAATT




ACCATCATGCCCATGATGCGTATCTAAATGCCGTCGTTGGAAC




TGCTTTGATTAAGAAATATCCAAAACTTGAATCGGAGTTTGTC




TATGGTGATTATAAAGTTTATGATGTTCGTAAAATGATTGCTA




AGTCTGAGCAAGAAATAGGCAAAGCAACCGCAAAATATTTCT




TTTACTCTAATATCATGAACTTCTTCAAAACAGAAATTACACT




TGCAAATGGAGAGATTCGCAAACGCCCTCTAATCGAAACTAA




TGGGGAAACTGGAGAAATTGTCTGGGATAAAGGGCGAGATTT




TGCCACAGTGCGCAAAGTATTGTCCATGCCCCAAGTCAATATT




GTCAAGAAAACAGAAGTACAGACAGGCGGATTCTCCAAGGA




GTCAATTTTACCAAAAAGAAATTCGGACAAGCTTATTGCTCGT




AAAAAAGACTGGGATCCAAAAAAATATGGTGGTTTTGATAGT




CCAACGGTAGCTTATTCAGTCCTAGTGGTTGCTAAGGTGGAAA




AAGGGAAATCGAAGAAGTTAAAATCCGTTAAAGAGTTACTAG




GGATCACAATTATGGAAAGAAGTTCCTTTGAAAAAAATCCGA




TTGACTTTTTAGAAGCTAAAGGATATAAGGAAGTTAAAAAAG




ACTTAATCATTAAACTACCTAAATATAGTCTTTTTGAGTTAGA




AAACGGTCGTAAACGGATGCTGGCTAGTGCCGGAGAATTACA




AAAAGGAAATGAGCTGGCTCTGCCAAGCAAATATGTGAATTT




TTTATATTTAGCTAGTCATTATGAAAAGTTGAAGGGTAGTCCA




GAAGATAACGAACAAAAACAATTGTTTGTGGAGCAGCATAAG




CATTATTTAGATGAGATTATTGAGCAAATCAGTGAATTTTCTA




AGCGTGTTATTTTAGCAGATGCCAATTTAGATAAAGTTCTTAG




TGCATATAACAAACATAGAGACAAACCAATACGTGAACAAGC




AGAAAATATTATTCATTTATTTACGTTGACGAATCTTGGAGCT




CCCGCTGCTTTTAAATATTTTGATACAACAATTGATCGTAAAC




GATATACGTCTACAAAAGAAGTTTTAGATGCCACTCTTATCCA




TCAATCCATCACTGGTCTTTATGAAACACGCATTGATTTGAGT




CAGCTAGGAGGTGACTGA






SpCas9
MDKKYSIGLDIGTNSVGWAVITDDYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 21



Streptococcus

KNLIGALLFGSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



MGAS1882
TIYHLRKKLADSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



wild type
SDVDKLFIQLVQIYNQLFEENPINASRVDAKAILSARLSKSRRLEN



NC_017053.1
LIAQLPGEKRNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKD




TYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNSEITK




APLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNG




YAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQR




TFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPY




YVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIER




MTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKP




AFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISG




VEDRFNASLGAYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFED




RGMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGHSLHEQIANLAGSPAIKKGILQTVKIVDELVKVMGHKPENIVI




EMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQ




LQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFIKD




DSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLI




TQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDS




RMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNY




HHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAK




SEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETG




EIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKR




NSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKL




KSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLF




ELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKG




SPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSA




YNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTST




KEVLDATLIHQSITGLYETRIDLSQLGGD






SpCas9
ATGGATAAAAAGTATTCTATTGGTTTAGACATCGGCACTAATT
SEQ ID NO: 22



Streptococcus

CCGTTGGATGGGCTGTCATAACCGATGAATACAAAGTACCTTC




pyogenes wild

AAAGAAATTTAAGGTGTTGGGGAACACAGACCGTCATTCGAT



type
TAAAAAGAATCTTATCGGTGCCCTCCTATTCGATAGTGGCGAA



SWBC2D7W014
ACGGCAGAGGCGACTCGCCTGAAACGAACCGCTCGGAGAAG




GTATACACGTCGCAAGAACCGAATATGTTACTTACAAGAAAT




TTTTAGCAATGAGATGGCCAAAGTTGACGATTCTTTCTTTCAC




CGTTTGGAAGAGTCCTTCCTTGTCGAAGAGGACAAGAAACAT




GAACGGCACCCCATCTTTGGAAACATAGTAGATGAGGTGGCA




TATCATGAAAAGTACCCAACGATTTATCACCTCAGAAAAAAG




CTAGTTGACTCAACTGATAAAGCGGACCTGAGGTTAATCTACT




TGGCTCTTGCCCATATGATAAAGTTCCGTGGGCACTTTCTCAT




TGAGGGTGATCTAAATCCGGACAACTCGGATGTCGACAAACT




GTTCATCCAGTTAGTACAAACCTATAATCAGTTGTTTGAAGAG




AACCCTATAAATGCAAGTGGCGTGGATGCGAAGGCTATTCTT




AGCGCCCGCCTCTCTAAATCCCGACGGCTAGAAAACCTGATC




GCACAATTACCCGGAGAGAAGAAAAATGGGTTGTTCGGTAAC




CTTATAGCGCTCTCACTAGGCCTGACACCAAATTTTAAGTCGA




ACTTCGACTTAGCTGAAGATGCCAAATTGCAGCTTAGTAAGG




ACACGTACGATGACGATCTCGACAATCTACTGGCACAAATTG




GAGATCAGTATGCGGACTTATTTTTGGCTGCCAAAAACCTTAG




CGATGCAATCCTCCTATCTGACATACTGAGAGTTAATACTGAG




ATTACCAAGGCGCCGTTATCCGCTTCAATGATCAAAAGGTAC




GATGAACATCACCAAGACTTGACACTTCTCAAGGCCCTAGTCC




GTCAGCAACTGCCTGAGAAATATAAGGAAATATTCTTTGATC




AGTCGAAAAACGGGTACGCAGGTTATATTGACGGCGGAGCGA




GTCAAGAGGAATTCTACAAGTTTATCAAACCCATATTAGAGA




AGATGGATGGGACGGAAGAGTTGCTTGTAAAACTCAATCGCG




AAGATCTACTGCGAAAGCAGCGGACTTTCGACAACGGTAGCA




TTCCACATCAAATCCACTTAGGCGAATTGCATGCTATACTTAG




AAGGCAGGAGGATTTTTATCCGTTCCTCAAAGACAATCGTGA




AAAGATTGAGAAAATCCTAACCTTTCGCATACCTTACTATGTG




GGACCCCTGGCCCGAGGGAACTCTCGGTTCGCATGGATGACA




AGAAAGTCCGAAGAAACGATTACTCCATGGAATTTTGAGGAA




GTTGTCGATAAAGGTGCGTCAGCTCAATCGTTCATCGAGAGG




ATGACCAACTTTGACAAGAATTTACCGAACGAAAAAGTATTG




CCTAAGCACAGTTTACTTTACGAGTATTTCACAGTGTACAATG




AACTCACGAAAGTTAAGTATGTCACTGAGGGCATGCGTAAAC




CCGCCTTTCTAAGCGGAGAACAGAAGAAAGCAATAGTAGATC




TGTTATTCAAGACCAACCGCAAAGTGACAGTTAAGCAATTGA




AAGAGGACTACTTTAAGAAAATTGAATGCTTCGATTCTGTCGA




GATCTCCGGGGTAGAAGATCGATTTAATGCGTCACTTGGTACG




TATCATGACCTCCTAAAGATAATTAAAGATAAGGACTTCCTGG




ATAACGAAGAGAATGAAGATATCTTAGAAGATATAGTGTTGA




CTCTTACCCTCTTTGAAGATCGGGAAATGATTGAGGAAAGACT




AAAAACATACGCTCACCTGTTCGACGATAAGGTTATGAAACA




GTTAAAGAGGCGTCGCTATACGGGCTGGGGACGATTGTCGCG




GAAACTTATCAACGGGATAAGAGACAAGCAAAGTGGTAAAA




CTATTCTCGATTTTCTAAAGAGCGACGGCTTCGCCAATAGGAA




CTTTATGCAGCTGATCCATGATGACTCTTTAACCTTCAAAGAG




GATATACAAAAGGCACAGGTTTCCGGACAAGGGGACTCATTG




CACGAACATATTGCGAATCTTGCTGGTTCGCCAGCCATCAAAA




AGGGCATACTCCAGACAGTCAAAGTAGTGGATGAGCTAGTTA




AGGTCATGGGACGTCACAAACCGGAAAACATTGTAATCGAGA




TGGCACGCGAAAATCAAACGACTCAGAAGGGGCAAAAAAAC




AGTCGAGAGCGGATGAAGAGAATAGAAGAGGGTATTAAAGA




ACTGGGCAGCCAGATCTTAAAGGAGCATCCTGTGGAAAATAC




CCAATTGCAGAACGAGAAACTTTACCTCTATTACCTACAAAAT




GGAAGGGACATGTATGTTGATCAGGAACTGGACATAAACCGT




TTATCTGATTACGACGTCGATCACATTGTACCCCAATCCTTTTT




GAAGGACGATTCAATCGACAATAAAGTGCTTACACGCTCGGA




TAAGAACCGAGGGAAAAGTGACAATGTTCCAAGCGAGGAAG




TCGTAAAGAAAATGAAGAACTATTGGCGGCAGCTCCTAAATG




CGAAACTGATAACGCAAAGAAAGTTCGATAACTTAACTAAAG




CTGAGAGGGGTGGCTTGTCTGAACTTGACAAGGCCGGATTTA




TTAAACGTCAGCTCGTGGAAACCCGCCAAATCACAAAGCATG




TTGCACAGATACTAGATTCCCGAATGAATACGAAATACGACG




AGAACGATAAGCTGATTCGGGAAGTCAAAGTAATCACTTTAA




AGTCAAAATTGGTGTCGGACTTCAGAAAGGATTTTCAATTCTA




TAAAGTTAGGGAGATAAATAACTACCACCATGCGCACGACGC




TTATCTTAATGCCGTCGTAGGGACCGCACTCATTAAGAAATAC




CCGAAGCTAGAAAGTGAGTTTGTGTATGGTGATTACAAAGTTT




ATGACGTCCGTAAGATGATCGCGAAAAGCGAACAGGAGATAG




GCAAGGCTACAGCCAAATACTTCTTTTATTCTAACATTATGAA




TTTCTTTAAGACGGAAATCACTCTGGCAAACGGAGAGATACG




CAAACGACCTTTAATTGAAACCAATGGGGAGACAGGTGAAAT




CGTATGGGATAAGGGCCGGGACTTCGCGACGGTGAGAAAAGT




TTTGTCCATGCCCCAAGTCAACATAGTAAAGAAAACTGAGGT




GCAGACCGGAGGGTTTTCAAAGGAATCGATTCTTCCAAAAAG




GAATAGTGATAAGCTCATCGCTCGTAAAAAGGACTGGGACCC




GAAAAAGTACGGTGGCTTCGATAGCCCTACAGTTGCCTATTCT




GTCCTAGTAGTGGCAAAAGTTGAGAAGGGAAAATCCAAGAAA




CTGAAGTCAGTCAAAGAATTATTGGGGATAACGATTATGGAG




CGCTCGTCTTTTGAAAAGAACCCCATCGACTTCCTTGAGGCGA




AAGGTTACAAGGAAGTAAAAAAGGATCTCATAATTAAACTAC




CAAAGTATAGTCTGTTTGAGTTAGAAAATGGCCGAAAACGGA




TGTTGGCTAGCGCCGGAGAGCTTCAAAAGGGGAACGAACTCG




CACTACCGTCTAAATACGTGAATTTCCTGTATTTAGCGTCCCA




TTACGAGAAGTTGAAAGGTTCACCTGAAGATAACGAACAGAA




GCAACTTTTTGTTGAGCAGCACAAACATTATCTCGACGAAATC




ATAGAGCAAATTTCGGAATTCAGTAAGAGAGTCATCCTAGCT




GATGCCAATCTGGACAAAGTATTAAGCGCATACAACAAGCAC




AGGGATAAACCCATACGTGAGCAGGCGGAAAATATTATCCAT




TTGTTTACTCTTACCAACCTCGGCGCTCCAGCCGCATTCAAGT




ATTTTGACACAACGATAGATCGCAAACGATACACTTCTACCA




AGGAGGTGCTAGACGCGACACTGATTCACCAATCCATCACGG




GATTATATGAAACTCGGATAGATTTGTCACAGCTTGGGGGTG




ACGGATCCCCCAAGAAGAAGAGGAAAGTCTCGAGCGACTACA




AAGACCATGACGGTGATTATAAAGATCATGACATCGATTACA




AGGATGACGATGACAAGGCTGCAGGA






SpCas9
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 23



Streptococcus

KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE



pyogenes wild
MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



type
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



Encoded
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE



product of
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK



SWBC2D7W014
DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT




KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGDGSPKKKRKVSSD




YKDHDGDYKDHDIDYKDDDDKAAG






SpCas9
ATGGATAAGAAATACTCAATAGGCTTAGATATCGGCACAAAT
SEQ ID NO: 24



Streptococcus

AGCGTCGGATGGGCGGTGATCACTGATGAATATAAGGTTCCG




pyogenes

TCTAAAAAGTTCAAGGTTCTGGGAAATACAGACCGCCACAGT



M1GAS wild
ATCAAAAAAAATCTTATAGGGGCTCTTTTATTTGACAGTGGAG



type
AGACAGCGGAAGCGACTCGTCTCAAACGGACAGCTCGTAGAA



NC_002737.2
GGTATACACGTCGGAAGAATCGTATTTGTTATCTACAGGAGAT




TTTTTCAAATGAGATGGCGAAAGTAGATGATAGTTTCTTTCAT




CGACTTGAAGAGTCTTTTTTGGTGGAAGAAGACAAGAAGCAT




GAACGTCATCCTATTTTTGGAAATATAGTAGATGAAGTTGCTT




ATCATGAGAAATATCCAACTATCTATCATCTGCGAAAAAAATT




GGTAGATTCTACTGATAAAGCGGATTTGCGCTTAATCTATTTG




GCCTTAGCGCATATGATTAAGTTTCGTGGTCATTTTTTGATTG




AGGGAGATTTAAATCCTGATAATAGTGATGTGGACAAACTAT




TTATCCAGTTGGTACAAACCTACAATCAATTATTTGAAGAAAA




CCCTATTAACGCAAGTGGAGTAGATGCTAAAGCGATTCTTTCT




GCACGATTGAGTAAATCAAGACGATTAGAAAATCTCATTGCT




CAGCTCCCCGGTGAGAAGAAAAATGGCTTATTTGGGAATCTC




ATTGCTTTGTCATTGGGTTTGACCCCTAATTTTAAATCAAATTT




TGATTTGGCAGAAGATGCTAAATTACAGCTTTCAAAAGATACT




TACGATGATGATTTAGATAATTTATTGGCGCAAATTGGAGATC




AATATGCTGATTTGTTTTTGGCAGCTAAGAATTTATCAGATGC




TATTTTACTTTCAGATATCCTAAGAGTAAATACTGAAATAACT




AAGGCTCCCCTATCAGCTTCAATGATTAAACGCTACGATGAAC




ATCATCAAGACTTGACTCTTTTAAAAGCTTTAGTTCGACAACA




ACTTCCAGAAAAGTATAAAGAAATCTTTTTTGATCAATCAAAA




AACGGATATGCAGGTTATATTGATGGGGGAGCTAGCCAAGAA




GAATTTTATAAATTTATCAAACCAATTTTAGAAAAAATGGATG




GTACTGAGGAATTATTGGTGAAACTAAATCGTGAAGATTTGCT




GCGCAAGCAACGGACCTTTGACAACGGCTCTATTCCCCATCA




AATTCACTTGGGTGAGCTGCATGCTATTTTGAGAAGACAAGA




AGACTTTTATCCATTTTTAAAAGACAATCGTGAGAAGATTGAA




AAAATCTTGACTTTTCGAATTCCTTATTATGTTGGTCCATTGGC




GCGTGGCAATAGTCGTTTTGCATGGATGACTCGGAAGTCTGA




AGAAACAATTACCCCATGGAATTTTGAAGAAGTTGTCGATAA




AGGTGCTTCAGCTCAATCATTTATTGAACGCATGACAAACTTT




GATAAAAATCTTCCAAATGAAAAAGTACTACCAAAACATAGT




TTGCTTTATGAGTATTTTACGGTTTATAACGAATTGACAAAGG




TCAAATATGTTACTGAAGGAATGCGAAAACCAGCATTTCTTTC




AGGTGAACAGAAGAAAGCCATTGTTGATTTACTCTTCAAAAC




AAATCGAAAAGTAACCGTTAAGCAATTAAAAGAAGATTATTT




CAAAAAAATAGAATGTTTTGATAGTGTTGAAATTTCAGGAGTT




GAAGATAGATTTAATGCTTCATTAGGTACCTACCATGATTTGC




TAAAAATTATTAAAGATAAAGATTTTTTGGATAATGAAGAAA




ATGAAGATATCTTAGAGGATATTGTTTTAACATTGACCTTATT




TGAAGATAGGGAGATGATTGAGGAAAGACTTAAAACATATGC




TCACCTCTTTGATGATAAGGTGATGAAACAGCTTAAACGTCGC




CGTTATACTGGTTGGGGACGTTTGTCTCGAAAATTGATTAATG




GTATTAGGGATAAGCAATCTGGCAAAACAATATTAGATTTTTT




GAAATCAGATGGTTTTGCCAATCGCAATTTTATGCAGCTGATC




CATGATGATAGTTTGACATTTAAAGAAGACATTCAAAAAGCA




CAAGTGTCTGGACAAGGCGATAGTTTACATGAACATATTGCA




AATTTAGCTGGTAGCCCTGCTATTAAAAAAGGTATTTTACAGA




CTGTAAAAGTTGTTGATGAATTGGTCAAAGTAATGGGGCGGC




ATAAGCCAGAAAATATCGTTATTGAAATGGCACGTGAAAATC




AGACAACTCAAAAGGGCCAGAAAAATTCGCGAGAGCGTATG




AAACGAATCGAAGAAGGTATCAAAGAATTAGGAAGTCAGATT




CTTAAAGAGCATCCTGTTGAAAATACTCAATTGCAAAATGAA




AAGCTCTATCTCTATTATCTCCAAAATGGAAGAGACATGTATG




TGGACCAAGAATTAGATATTAATCGTTTAAGTGATTATGATGT




CGATCACATTGTTCCACAAAGTTTCCTTAAAGACGATTCAATA




GACAATAAGGTCTTAACGCGTTCTGATAAAAATCGTGGTAAA




TCGGATAACGTTCCAAGTGAAGAAGTAGTCAAAAAGATGAAA




AACTATTGGAGACAACTTCTAAACGCCAAGTTAATCACTCAA




CGTAAGTTTGATAATTTAACGAAAGCTGAACGTGGAGGTTTG




AGTGAACTTGATAAAGCTGGTTTTATCAAACGCCAATTGGTTG




AAACTCGCCAAATCACTAAGCATGTGGCACAAATTTTGGATA




GTCGCATGAATACTAAATACGATGAAAATGATAAACTTATTC




GAGAGGTTAAAGTGATTACCTTAAAATCTAAATTAGTTTCTGA




CTTCCGAAAAGATTTCCAATTCTATAAAGTACGTGAGATTAAC




AATTACCATCATGCCCATGATGCGTATCTAAATGCCGTCGTTG




GAACTGCTTTGATTAAGAAATATCCAAAACTTGAATCGGAGTT




TGTCTATGGTGATTATAAAGTTTATGATGTTCGTAAAATGATT




GCTAAGTCTGAGCAAGAAATAGGCAAAGCAACCGCAAAATAT




TTCTTTTACTCTAATATCATGAACTTCTTCAAAACAGAAATTA




CACTTGCAAATGGAGAGATTCGCAAACGCCCTCTAATCGAAA




CTAATGGGGAAACTGGAGAAATTGTCTGGGATAAAGGGCGAG




ATTTTGCCACAGTGCGCAAAGTATTGTCCATGCCCCAAGTCAA




TATTGTCAAGAAAACAGAAGTACAGACAGGCGGATTCTCCAA




GGAGTCAATTTTACCAAAAAGAAATTCGGACAAGCTTATTGC




TCGTAAAAAAGACTGGGATCCAAAAAAATATGGTGGTTTTGA




TAGTCCAACGGTAGCTTATTCAGTCCTAGTGGTTGCTAAGGTG




GAAAAAGGGAAATCGAAGAAGTTAAAATCCGTTAAAGAGTTA




CTAGGGATCACAATTATGGAAAGAAGTTCCTTTGAAAAAAAT




CCGATTGACTTTTTAGAAGCTAAAGGATATAAGGAAGTTAAA




AAAGACTTAATCATTAAACTACCTAAATATAGTCTTTTTGAGT




TAGAAAACGGTCGTAAACGGATGCTGGCTAGTGCCGGAGAAT




TACAAAAAGGAAATGAGCTGGCTCTGCCAAGCAAATATGTGA




ATTTTTTATATTTAGCTAGTCATTATGAAAAGTTGAAGGGTAG




TCCAGAAGATAACGAACAAAAACAATTGTTTGTGGAGCAGCA




TAAGCATTATTTAGATGAGATTATTGAGCAAATCAGTGAATTT




TCTAAGCGTGTTATTTTAGCAGATGCCAATTTAGATAAAGTTC




TTAGTGCATATAACAAACATAGAGACAAACCAATACGTGAAC




AAGCAGAAAATATTATTCATTTATTTACGTTGACGAATCTTGG




AGCTCCCGCTGCTTTTAAATATTTTGATACAACAATTGATCGT




AAACGATATACGTCTACAAAAGAAGTTTTAGATGCCACTCTTA




TCCATCAATCCATCACTGGTCTTTATGAAACACGCATTGATTT




GAGTCAGCTAGGAGGTGACTGA






SpCas9
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 25


Streptococcus
KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



M1GAS wild
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



type
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE



Encoded
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK



product of
DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT



NC_002737.2
KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN



(100% identical
GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ



to the canonical
RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP



Q99ZW2
YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE



wild type)
RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD









The multi-flap prime editors described herein may include any of the above SpCas9 sequences, or any variant thereof having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.


B. Wild Type Cas9 Orthologs


In other embodiments, the Cas9 protein can be a wild type Cas9 ortholog from another bacterial species different from the canonical Cas9 from S. pyogenes. For example, the following Cas9 orthologs can be used in connection with the multi-flap prime editor constructs described in this specification. In addition, any variant Cas9 orthologs having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity to any of the below orthologs may also be used with the multi-flap present prime editors.













Description
Sequence







LfCas9
MKEYHIGLDIGTSSIGWAVTDSQFKLMRIKGKTAIGVRLFEEGKTAAERRTFRT



Lactobacillus

TRRRLKRRKWRLHYLDEIFAPHLQEVDENFLRRLKQSNIHPEDPTKNQAFIGKL



fermentum

LFPDLLKKNERGYPTLIKMRDELPVEQRAHYPVMNIYKLREAMINEDRQFDLRE


wild type
VYLAVHHIVKYRGHFLNNASVDKFKVGRIDFDKSFNVLNEAYEELQNGEGSFTI


GenBank:
EPSKVEKIGQLLLDTKMRKLDRQKAVAKLLEVKVADKEETKRNKQIATAMSK


SNX31424.11
LVLGYKADFATVAMANGNEWKIDLSSETSEDEIEKFREELSDAQNDILTEITSLF



SQIMLNEIVPNGMSISESMMDRYWTHERQLAEVKEYLATQPASARKEFDQVYN



KYIGQAPKERGFDLEKGLKKILSKKENWKEIDELLKAGDFLPKQRTSANGVIPH



QMHQQELDRIIEKQAKYYPWLATENPATGERDRHQAKYELDQLVSFRIPYYVG



PLVTPEVQKATSGAKFAWAKRKEDGEITPWNLWDKIDRAESAEAFIKRMTVKD



TYLLNEDVLPANSLLYQKYNVLNELNNVRVNGRRLSVGIKQDIYTELFKKKKT



VKASDVASLVMAKTRGVNKPSVEGLSDPKKFNSNLATYLDLKSIVGDKVDDN



RYQTDLENIIEWRSVFEDGEIFADKLTEVEWLTDEQRSALVKKRYKGWGRLSK



KLLTGIVDENGQRIIDLMWNTDQNFKEIVDQPVFKEQIDQLNQKAITNDGMTLR



ERVESVLDDAYTSPQNKKAIWQVVRVVEDIVKAVGNAPKSISIEFARNEGNKGE



ITRSRRTQLQKLFEDQAHELVKDTSLTEELEKAPDLSDRYYFYFTQGGKDMYT



GDPINFDEISTKYDIDHILPQSFVKDNSLDNRVLTSRKENNKKSDQVPAKLYAA



KMKPYWNQLLKQGLITQRKFENLTKDVDQNIKYRSLGFVKRQLVETRQVIKLT



ANILGSMYQEAGTEIIETRAGLTKQLREEFDLPKVREVNDYHHAVDAYLTTFAG



QYLNRRYPKLRSFFVYGEYMKFKHGSDLKLRNFNFFHELMEGDKSQGKVVDQ



QTGELITTRDEVAKSFDRLLNMKYMLVSKEVHDRSDQLYGATIVTAKESGKLT



SPIEIKKNRLVDLYGAYTNGTSAFMTIIKFTGNKPKYKVIGIPTTSAASLKRAGKP



GSESYNQELHRIIKSNPKVKKGFEIVVPHVSYGQLIVDGDCKFTLASPTVQHPAT



QLVLSKKSLETISSGYKILKDKPAIANERLIRVFDEVVGQMNRYFTIFDQRSNRQ



KVADARDKFLSLPTESKYEGAKKVQVGKTEVITNLLMGLHANATQGDLKVLG



LATFGFFQSTTGLSLSEDTMIVYQSPTGLFERRICLKDI (SEQ ID NO: 26)





SaCas9
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFD



Staphylococcus

SGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEE



aureus

DKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIK


wild type
FRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSK


GenBank:
SRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYD


AYD60528.1
DDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDE



HHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEK



MDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDN



REKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSF



IERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQ



KKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLL



KIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKR



RRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDI



QKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIE



MARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYY



LQNGRDMYVDQELDINRLSDYDVDHIVPQSFLKDDSIDNKVLTRSDKNRGKSD



NVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQL



VETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKV



REINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQ



EIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFAT



VRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFD



SPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKE



VKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYE



KLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKH



RDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSIT



GLYETRIDLSQLGGD (SEQ ID NO: 27)





SaCas9
MGKRNYILGLDIGITSVGYGIIDYETRDVIDAGVRLFKEANVENNEGRRSKRGA



Staphylococcus

RRLKRRRRHRIQRVKKLLFDYNLLTDHSELSGINPYEARVKGLSQKLSEEEFSA



aureus

ALLHLAKRRGVHNVNEVEEDTGNELSTKEQISRNSKALEEKYVAELQLERLKK



DGEVRGSINRFKTSDYVKEAKQLLKVQKAYHQLDQSFIDTYIDLLETRRTYYEG



PGEGSPFGWKDIKEWYEMLMGHCTYFPEELRSVKYAYNADLYNALNDLNNLV



ITRDENEKLEYYEKFQIIENVFKQKKKPTLKQIAKEILVNEEDIKGYRVTSTGKPE



FTNLKVYHDIKDITARKEIIENAELLDQIAKILTIYQSSEDIQEELTNLNSELTQEEI



EQISNLKGYTGTHNLSLKAINLILDELWHTNDNQIAIFNRLKLVPKKVDLSQQKE



IPTTLVDDFILSPVVKRSFIQSIKVINAIIKKYGLPNDIIIELAREKNSKDAQKMINE



MQKRNRQTNERIEEIIRTTGKENAKYLIEKIKLHDMQEGKCLYSLEAIPLEDLLN



NPFNYEVDHIIPRSVSFDNSFNNKVLVKQEENSKKGNRTPFQYLSSSDSKISYETF



KKHILNLAKGKGRISKTKKEYLLEERDINRFSVQKDFINRNLVDTRYATRGLMN



LLRSYFRVNNLDVKVKSINGGFTSFLRRKWKFKKERNKGYKHHAEDALIIANA



DFIFKEWKKLDKAKKVMENQMFEEKQAESMPEIETEQEYKEIFITPHQIKHIKDF



KDYKYSHRVDKKPNRKLINDTLYSTRKDDKGNTLIVNNLNGLYDKDNDKLKK



LINKSPEKLLMYHHDPQTYQKLKLIMEQYGDEKNPLYKYYEETGNYLTKYSKK



DNGPVIKKIKYYGNKLNAHLDITDDYPNSRNKVVKLSLKPYRFDVYLDNGVYK



FVTVKNLDVIKKENYYEVNSKCYEEAKKLKKISNQAEFIASFYKNDLIKINGEL



YRVIGVNNDLLNRIEVNMIDITYREYLENMNDKRPPHIIKTIASKTQSIKKYSTDI



LGNLYEVKSKKHPQIIKK



(SEQ ID NO: 28)





StCas9
MLFNKCIIISINLDFSNKEKCMTKPYSIGLDIGTNSVGWAVITDNYKVPSKKMKV



LGNTSKKYIKKNLLGVLLFDSGITAEGRRLKRTARRRYTRRRNRILYLQEIFSTE



MATLDDAFFQRLDDSFLVPDDKRDSKYPIFGNLVEEKVYHDEFPTIYHLRKYLA



Streptococcus

DSTKKADLRLVYLALAHMIKYRGHFLIEGEFNSKNNDIQKNFQDFLDTYNAIFE



thermophilus

SDLSLENSKQLEEIVKDKISKLEKKDRILKLFPGEKNSGIFSEFLKLIVGNQADFR


UniProtKB/
KCFNLDEKASLHFSKESYDEDLETLLGYIGDDYSDVFLKAKKLYDAILLSGFLT


Swiss-Prot:
VTDNETEAPLSSAMIKRYNEHKEDLALLKEYIRNISLKTYNEVFKDDTKNGYAG


G3ECR1.2
YIDGKTNQEDFYVYLKNLLAEFEGADYFLEKIDREDFLRKQRTFDNGSIPYQIHL


Wild type
QEMRAILDKQAKFYPFLAKNKERIEKILTFRIPYYVGPLARGNSDFAWSIRKRNE



KITPWNFEDVIDKESSAEAFINRMTSFDLYLPEEKVLPKHSLLYETFNVYNELTK



VRFIAESMRDYQFLDSKQKKDIVRLYFKDKRKVTDKDIIEYLHAIYGYDGIELK



GIEKQFNSSLSTYHDLLNIINDKEFLDDSSNEAIIEEIIHTLTIFEDREMIKQRLSKF



ENIFDKSVLKKLSRRHYTGWGKLSAKLINGIRDEKSGNTILDYLIDDGISNRNFM



QLIHDDALSFKKKIQKAQIIGDEDKGNIKEVVKSLPGSPAIKKGILQSIKIVDELV



KVMGGRKPESIVVEMARENQYTNQGKSNSQQRLKRLEKSLKELGSKILKENIPA



KLSKIDNNALQNDRLYLYYLQNGKDMYTGDDLDIDRLSNYDIDHIIPQAFLKDN



SIDNKVLVSSASNRGKSDDFPSLEVVKKRKTFWYQLLKSKLISQRKFDNLTKAE



RGGLLPEDKAGFIQRQLVETRQITKHVARLLDEKFNNKKDENNRAVRTVKIITL



KSTLVSQFRKDFELYKVREINDFHHAHDAYLNAVIASALLKKYPKLEPEFVYGD



YPKYNSFRERKSATEKVYFYSNIMNIFKKSISLADGRVIERPLIEVNEETGESVW



NKESDLATVRRVLSYPQVNVVKKVEEQNHGLDRGKPKGLFNANLSSKPKPNSN



ENLVGAKEYLDPKKYGGYAGISNSFAVLVKGTIEKGAKKKITNVLEFQGISILD



RINYRKDKLNFLLEKGYKDIELIIELPKYSLFELSDGSRRMLASILSTNNKRGEIH



KGNQIFLSQKFVKLLYHAKRISNTINENHRKYVENHKKEFEELFYYILEFNENYV



GAKKNGKLLNSAFQSWQNHSIDELCSSFIGPTGSERKGLFELTSRGSAADFEFLG



VKIPRYRDYTPSSLLKDATLIHQSVTGLYETRIDLAKLGEG (SEQ ID NO: 29)





LcCas9
MKIKNYNLALTPSTSAVGHVEVDDDLNILEPVHHQKAIGVAKFGEGETAEARR



Lactobacillus

LARSARRTTKRRANRINHYFNEIMKPEIDKVDPLMFDRIKQAGLSPLDERKEFRT



crispatus

VIFDRPNIASYYHNQFPTIWHLQKYLMITDEKADIRLIYWALHSLLKHRGHFFNT


NCBI
TPMSQFKPGKLNLKDDMLALDDYNDLEGLSFAVANSPEIEKVIKDRSMHKKEK


Reference
IAELKKLIVNDVPDKDLAKRNNKIITQIVNAIMGNSFHLNFIFDMDLDKLTSKA


Sequence:
WSFKLDDPELDTKFDAISGSMTDNQIGIFETLQKIYSAISLLDILNGSSNVVDAKN


WP_133478044.1
ALYDKHKRDLNLYFKFLNTLPDEIAKTLKAGYTLYIGNRKKDLLAARKLLKVN


Wild type
VAKNFSQDDFYKLINKELKSIDKQGLQTRFSEKVGELVAQNNFLPVQRSSDNVF



IPYQLNAITFNKILENQGKYYDFLVKPNPAKKDRKNAPYELSQLMQFTIPYYVG



PLVTPEEQVKSGIPKTSRFAWMVRKDNGAITPWNFYDKVDIEATADKFIKRSIA



KDSYLLSELVLPKHSLLYEKYEVFNELSNVSLDGKKLSGGVKQILFNEVFKKTN



KVNTSRILKALAKHNIPGSKITGLSNPEEFTSSLQTYNAWKKYFPNQIDNFAYQQ



DLEKMIEWSTVFEDHKILAKKLDEIEWLDDDQKKFVANTRLRGWGRLSKRLLT



GLKDNYGKSIMQRLETTKANFQQIVYKPEFREQIDKISQAAAKNQSLEDILANS



YTSPSNRKAIRKTMSVVDEYIKLNHGKEPDKIFLMFQRSEQEKGKQTEARSKQL



NRILSQLKADKSANKLFSKQLADEFSNAIKKSKYKLNDKQYFYFQQLGRDALT



GEVIDYDELYKYTVLHIIPRSKLTDDSQNNKVLTKYKIVDGSVALKFGNSYSDA



LGMPIKAFWTELNRLKLIPKGKLLNLTTDFSTLNKYQRDGYIARQLVETQQIVK



LLATIMQSRFKHTKIIEVRNSQVANIRYQFDYFRIKNLNEYYRGFDAYLAAVVG



TYLYKVYPKARRLFVYGQYLKPKKTNQENQDMHLDSEKKSQGFNFLWNLLYG



KQDQIFVNGTDVIAFNRKDLITKMNTVYNYKSQKISLAIDYHNGAMFKATLFPR



NDRDTAKTRKLIPKKKDYDTDIYGGYTSNVDGYMLLAEIIKRDGNKQYGFYGV



PSRLVSELDTLKKTRYTEYEEKLKEIIKPELGVDLKKIKKIKILKNKVPFNQVIID



KGSKFFITSTSYRWNYRQLILSAESQQTLMDLVVDPDFSNHKARKDARKNADE



RLIKVYEEILYQVKNYMPMFVELHRCYEKLVDAQKTFKSLKISDKAMVLNQILI



LLHSNATSPVLEKLGYHTRFTLGKKHNLISENAVLVTQSITGLKENHVSIKQML



(SEQ ID NO: 30)





PdCas9
MTNEKYSIGLDIGTSSIGFAVVNDNNRVIRVKGKNAIGVRLFDEGKAAADRRSF



Pedicoccus

RTTRRSFRTTRRRLSRRRWRLKLLREIFDAYITPVDEAFFIRLKESNLSPKDSKKQ



damnosus

YSGDILFNDRSDKDFYEKYPTIYHLRNALMTEHRKFDVREIYLAIHHIMKFRGH


NCBI
FLNATPANNFKVGRLNLEEKFEELNDIYQRVFPDESIEFRTDNLEQIKEVLLDNK


Reference
RSRADRQRTLVSDIYQSSEDKDIEKRNKAVATEILKASLGNKAKLNVITNVEVD


Sequence:
KEAAKEWSITFDSESIDDDLAKIEGQMTDDGHEIIEVLRSLYSGITLSAIVPENHT


WP_062913273.1
LSQSMVAKYDLHKDHLKLFKKLINGMTDTKKAKNLRAAYDGYIDGVKGKVLP


Wild type
QEDFYKQVQVNLDDSAEANEIQTYIDQDIFMPKQRTKANGSIPHQLQQQELDQI



IENQKAYYPWLAELNPNPDKKRQQLAKYKLDELVTFRVPYYVGPMITAKDQK



NQSGAEFAWMIRKEPGNITPWNFDQKVDRMATANQFIKRMTTTDTYLLGEDV



LPAQSLLYQKFEVLNELNKIRIDHKPISIEQKQQIFNDLFKQFKNVTIKHLQDYLV



SQGQYSKRPLIEGLADEKRFNSSLSTYSDLCGIFGAKLVEENDRQEDLEKIIEWS



TIFEDKKIYRAKLNDLTWLTDDQKEKLATKRYQGWGRLSRKLLVGLKNSEHR



NIMDILWITNENFMQIQAEPDFAKLVTDANKGMLEKTDSQDVINDLYTSPQNK



KAIRQILLVVHDIQNAMHGQAPAKIHVEFARGEERNPRRSVQRQRQVEAAYEK



VSNELVSAKVRQEFKEAINNKRDFKDRLFLYFMQGGIDIYTGKQLNIDQLSSYQI



DHILPQAFVKDDSLTNRVLTNENQVKADSVPIDIFGKKMLSVWGRMKDQGLIS



KGKYRNLTMNPENISAHTENGFINRQLVETRQVIKLAVNILADEYGDSTQIISVK



ADLSHQMREDFELLKNRDVNDYHHAFDAYLAAFIGNYLLKRYPKLESYFVYG



DFKKFTQKETKMRRFNFIYDLKHCDQVVNKETGEILWTKDEDIKYIRHLFAYK



KILVSHEVREKRGALYNQTIYKAKDDKGSGQESKKLIRIKDDKETKIYGGYSGK



SLAYMTIVQITKKNKVSYRVIGIPTLALARLNKLENDSTENNGELYKIIKPQFTH



YKVDKKNGEIIETTDDFKIVVSKVRFQQLIDDAGQFFMLASDTYKNNAQQLVIS



NNALKAINNTNITDCPRDDLERLDNLRLDSAFDEIVKKMDKYFSAYDANNFRE



KIRNSNLIFYQLPVEDQWENNKITELGKRTVLTRILQGLHANATTTDMSIFKIKT



PFGQLRQRSGISLSENAQLIYQSPTGLFERRVQLNKIK (SEQ ID NO: 31)





FnCas9
MKKQKFSDYYLGFDIGTNSVGWCVTDLDYNVLRFNKKDMWGSRLFEEAKTA



Fusobaterium

AERRVQRNSRRRLKRRKWRLNLLEEIFSNEILKIDSNFFRRLKESSLWLEDKSSK



nucleatum

EKFTLFNDDNYKDYDFYKQYPTIFHLRNELIKNPEKKDIRLVYLAIHSIFKSRGH


NCBI
FLFEGQNLKEIKNFETLYNNLIAFLEDNGINKIIDKNNIEKLEKIVCDSKKGLKDK


Reference
EKEFKEIFNSDKQLVAIFKLSVGSSVSLNDLFDTDEYKKGEVEKEKISFREQIYED


Sequence:
DKPIYYSILGEKIELLDIAKTFYDFMVLNNILADSQYISEAKVKLYEEHKKDLKN


WP_060798984.1
LKYIIRKYNKGNYDKLFKDKNENNYSAYIGLNKEKSKKEVIEKSRLKIDDLIKNI



KGYLPKVEEIEEKDKAIFNKILNKIELKTILPKQRISDNGTLPYQIHEAELEKILEN



QSKYYDFLNYEENGIITKDKLLMTFKFRIPYYVGPLNSYHKDKGGNSWIVRKEE



GKILPWNFEQKVDIEKSAEEFIKRMTNKCTYLNGEDVIPKDTFLYSEYVILNELN



KVQVNDEFLNEENKRKIIDELFKENKKVSEKKFKEYLLVKQIVDGTIELKGVKD



SFNSNYISYIRFKDIFGEKLNLDIYKEISEKSILWKCLYGDDKKIFEKKIKNEYGDI



LTKDEIKKINTFKFNNWGRLSEKLLTGIEFINLETGECYSSVMDALRRTNYNLM



ELLSSKFTLQESINNENKEMNEASYRDLIEESYVSPSLKRAIFQTLKIYEEIRKITG



RVPKKVFIEMARGGDESMKNKKIPARQEQLKKLYDSCGNDIANFSIDIKEMKNS



LISYDNNSLRQKKLYLYYLQFGKCMYTGREIDLDRLLQNNDTYDIDHIYPRSKV



IKDDSFDNLVLVLKNENAEKSNEYPVKKEIQEKMKSFWRFLKEKNFISDEKYKR



LTGKDDFELRGFMARQLVNVRQTTKEVGKILQQIEPEIKIVYSKAEIASSFREMF



DFIKVRELNDTHHAKDAYLNIVAGNVYNTKFTEKPYRYLQEIKENYDVKKIYN



YDIKNAWDKENSLEIVKKNMEKNTVNITRFIKEKKGQLFDLNPIKKGETSNEIISI



KPKVYNGKDDKLNEKYGYYKSLNPAYFLYVEHKEKNKRIKSFERVNLVDVNN



IKDEKSLVKYLIENKKLVEPRVIKKVYKRQVILINDYPYSIVTLDSNKLMDFENL



KPLFLENKYEKILKNVIKFLEDNQGKSEENYKFIYLKKKDRYEKNETLESVKDR



YNLEFNEMYDKFLEKLDSKDYKNYMNNKKYQELLDVKEKFIKLNLFDKAFTL



KSFLDLFNRKTMADFSKVGLTKYLGKIQKISSNVLSKNELYLLEESVTGLFVKKI



KL (SEQ ID NO: 32)





EcCas9
RRKQRIQILQELLGEEVLKTDPGFFHRMKESRYVVEDKRTLDGKQVELPYALFV



Enterococcus

DKDYTDKEYYKQFPTINHLIVYLMTTSDTPDIRLVYLALHYYMKNRGNFLHSG



cecorum

DINNVKDINDILEQLDNVLETFLDGWNLKLKSYVEDIKNIYNRDLGRGERKKAF


NCBI
VNTLGAKTKAEKAFCSLISGGSTNLAELFDDSSLKEIETPKIEFASSSLEDKIDGIQ


Reference
EALEDRFAVIEAAKRLYDWKTLTDILGDSSSLAEARVNSYQMHHEQLLELKSL


Sequence:
VKEYLDRKVFQEVFVSLNVANNYPAYIGHTKINGKKKELEVKRTKRNDFYSYV


WP_047338501.1
KKQVIEPIKKKVSDEAVLTKLSEIESLIEVDKYLPLQVNSDNGVIPYQVKLNELT


Wild type
RIFDNLENRIPVLRENRDKIIKTFKFRIPYYVGSLNGVVKNGKCTNWMVRKEEG



KIYPWNFEDKVDLEASAEQFIRRMTNKCTYLVNEDVLPKYSLLYSKYLVLSELN



NLRIDGRPLDVKIKQDIYENVFKKNRKVTLKKIKKYLLKEGIITDDDELSGLADD



VKSSLTAYRDFKEKLGHLDLSEAQMENIILNITLFGDDKKLLKKRLAALYPFIDD



KSLNRIATLNYRDWGRLSERFLSGITSVDQETGELRTIIQCMYETQANLMQLLA



EPYHFVEAIEKENPKVDLESISYRIVNDLYVSPAVKRQIWQTLLVIKDIKQVMKH



DPERIFIEMAREKQESKKTKSRKQVLSEVYKKAKEYEHLFEKLNSLTEEQLRSK



KIYLYFTQLGKCMYSGEPIDFENLVSANSNYDIDHIYPQSKTIDDSFNNIVLVKK



SLNAYKSNHYPIDKNIRDNEKVKTLWNTLVSKGLITKEKYERLIRSTPFSDEELA



GFIARQLVETRQSTKAVAEILSNWFPESEIVYSKAKNVSNFRQDFEILKVRELND



CHHAHDAYLNIVVGNAYHTKFTNSPYRFIKNKANQEYNLRKLLQKVNKIESNG



VVAWVGQSENNPGTIATVKKVIRRNTVLISRMVKEVDGQLFDLTLMKKGKGQ



VPIKSSDERLTDISKYGGYNKATGAYFTFVKSKKRGKVVRSFEYVPLHLSKQFE



NNNELLKEYIEKDRGLTDVEILIPKVLINSLFRYNGSLVRITGRGDTRLLLVHEQP



LYVSNSFVQQLKSVSSYKLKKSENDNAKLTKTATEKLSNIDELYDGLLRKLDLP



IYSYWFSSIKEYLVESRTKYIKLSIEEKALVIFEILHLFQSDAQVPNLKILGLSTKP



SRIRIQKNLKDTDKMSIIHQSPSGIFEHEIELTSL (SEQ ID NO: 33)





AhCas9
MQNGFLGITVSSEQVGWAVTNPKYELERASRKDLWGVRLFDKAETAEDRRMF



Anaerostipes

RTNRRLNQRKKNRIHYLRDIFHEEVNQKDPNFFQQLDESNFCEDDRTVEFNFDT



hadrus

NLYKNQFPTVYHLRKYLMETKDKPDIRLVYLAFSKFMKNRGHFLYKGNLGEV


NCBI
MDFENSMKGFCESLEKFNIDFPTLSDEQVKEVRDILCDHKIAKTVKKKNIITITK


Reference
VKSKTAKAWIGLFCGCSVPVKVLFQDIDEEIVTDPEKISFEDASYDDYIANIEKG


Sequence:
VGIYYEAIVSAKMLFDWSILNEILGDHQLLSDAMIAEYNKHHDDLKRLQKIIKG


WP_044924278.1
TGSRELYQDIFINDVSGNYVCYVGHAKTMSSADQKQFYTFLKNRLKNVNGISSE


Wild type
DAEWIDTEIKNGTLLPKQTKRDNSVIPHQLQLREFELILDNMQEMYPFLKENRE



KLLKIFNFVIPYYVGPLKGVVRKGESTNWMVPKKDGVIHPWNFDEMVDKEAS



AECFISRMTGNCSYLFNEKVLPKNSLLYETFEVLNELNPLKINGEPISVELKQRIY



EQLFLTGKKVTKKSLTKYLIKNGYDKDIELSGIDNEFHSNLKSHIDFEDYDNLSD



EEVEQIILRITVFEDKQLLKDYLNREFVKLSEDERKQICSLSYKGWGNLSEMLLN



GITVTDSNGVEVSVMDMLWNTNLNLMQILSKKYGYKAEIEHYNKEHEKTIYNR



EDLMDYLNIPPAQRRKVNQLITIVKSLKKTYGVPNKIFFKISREHQDDPKRTSSR



KEQLKYLYKSLKSEDEKHLMKELDELNDHELSNDKVYLYFLQKGRCIYSGKKL



NLSRLRKSNYQNDIDYIYPLSAVNDRSMNNKVLTGIQENRADKYTYFPVDSEIQ



KKMKGFWMELVLQGFMTKEKYFRLSRENDFSKSELVSFIEREISDNQQSGRMIA



SVLQYYFPESKIVFVKEKLISSFKRDFHLISSYGHNHLQAAKDAYITIVVGNVYH



TKFTMDPAIYFKNHKRKDYDLNRLFLENISRDGQIAWESGPYGSIQTVRKEYAQ



NHIAVTKRVVEVKGGLFKQMPLKKGHGEYPLKTNDPRFGNIAQYGGYTNVTG



SYFVLVESMEKGKKRISLEYVPVYLHERLEDDPGHKLLKEYLVDHRKLNHPKIL



LAKVRKNSLLKIDGFYYRLNGRSGNALILTNAVELIMDDWQTKTANKISGYMK



RRAIDKKARVYQNEFHIQELEQLYDFYLDKLKNGVYKNRKNNQAELIHNEKEQ



FMELKTEDQCVLLTEIKKLFVCSPMQADLTLIGGSKHTGMIAMSSNVTKADFA



VIAEDPLGLRNKVIYSHKGEK (SEQ ID NO: 34)





KvCas9
MSQNNNKIYNIGLDIGDASVGWAVVDEHYNLLKRHGKHMWGSRLFTQANTA



Kandleria

VERRSSRSTRRRYNKRRERIRLLREIMEDMVLDVDPTFFIRLANVSFLDQEDKK



vitulina

DYLKENYHSNYNLFIDKDFNDKTYYDKYPTIYHLRKHLCESKEKEDPRLIYLAL


NCBI
HHIVKYRGNFLYEGQKFSMDVSNIEDKMIDVLRQFNEINLFEYVEDRKKIDEVL


Reference
NVLKEPLSKKHKAEKAFALFDTTKDNKAAYKELCAALAGNKFNVTKMLKEAE


Sequence:
LHDEDEKDISFKFSDATFDDAFVEKQPLLGDCVEFIDLLHDIYSWVELQNILGSA


WP_031589969.1
HTSEPSISAAMIQRYEDHKNDLKLLKDVIRKYLPKKYFEVFRDEKSKKNNYCNY


Wild type
INHPSKTPVDEFYKYIKKLIEKIDDPDVKTILNKIELESFMLKQNSRTNGAVPYQ



MQLDELNKILENQSVYYSDLKDNEDKIRSILTFRIPYYFGPLNITKDRQFDWIIKK



EGKENERILPWNANEIVDVDKTADEFIKRMRNFCTYFPDEPVMAKNSLTVSKY



EVLNEINKLRINDHLIKRDMKDKMLHTLFMDHKSISANAMKKWLVKNQYFSN



TDDIKIEGFQKENACSTSLTPWIDFTKIFGKINESNYDFIEKIIYDVTVFEDKKILR



RRLKKEYDLDEEKIKKILKLKYSGWSRLSKKLLSGIKTKYKDSTRTPETVLEVM



ERTNMNLMQVINDEKLGFKKTIDDANSTSVSGKFSYAEVQELAGSPAIKRGIWQ



ALLIVDEIKKIMKHEPAHVYIEFARNEDEKERKDSFVNQMLKLYKDYDFEDETE



KEANKHLKGEDAKSKIRSERLKLYYTQMGKCMYTGKSLDIDRLDTYQVDHIVP



QSLLKDDSIDNKVLVLSSENQRKLDDLVIPSSIRNKMYGFWEKLFNNKIISPKKF



YSLIKTEFNEKDQERFINRQIVETRQITKHVAQIIDNHYENTKVVTVRADLSHQF



RERYHIYKNRDINDFHHAHDAYIATILGTYIGHRFESLDAKYIYGEYKRIFRNQK



NKGKEMKKNNDGFILNSMRNIYADKDTGEIVWDPNYIDRIKKCFYYKDCFVTK



KLEENNGTFFNVTVLPNDTNSDKDNTLATVPVNKYRSNVNKYGGFSGVNSFIV



AIKGKKKKGKKVIEVNKLTGIPLMYKNADEEIKINYLKQAEDLEEVQIGKEILK



NQLIEKDGGLYYIVAPTEIINAKQLILNESQTKLVCEIYKAMKYKNYDNLDSEKI



IDLYRLLINKMELYYPEYRKQLVKKFEDRYEQLKVISIEEKCNIIKQILATLHCNS



SIGKIMYSDFKISTTIGRLNGRTISLDDISFIAESPTGMYSKKYKL (SEQ ID NO:



35)





EfCas9
MRLFEEGHTAEDRRLKRTARRRISRRRNRLRYLQAFFEEAMTDLDENFFARLQE



Enterococcus

SFLVPEDKKWHRHPIFAKLEDEVAYHETYPTIYHLRKKLADSSEQADLRLIYLA



faecalis

LAHIVKYRGHFLIEGKLSTENTSVKDQFQQFMVIYNQTFVNGESRLVSAPLPESV


NCBI
LIEEELTEKASRTKKSEKVLQQFPQEKANGLFGQFLKLMVGNKADFKKVFGLE


Reference
EEAKITYASESYEEDLEGILAKVGDEYSDVFLAAKNVYDAVELSTILADSDKKS


Sequence:
HAKLSSSMIVRFTEHQEDLKKFKRFIRENCPDEYDNLFKNEQKDGYAGYIAHAG


WP_016631044.1
KVSQLKFYQYVKKIIQDIAGAEYFLEKIAQENFLRKQRTFDNGVIPHQIHLAELQ


Wild type
AIIHRQAAYYPFLKENQEKIEQLVTFRIPYYVGPLSKGDASTFAWLKRQSEEPIRP



WNLQETVDLDQSATAFIERMTNFDTYLPSEKVLPKHSLLYEKFMVFNELTKISY



TDDRGIKANFSGKEKEKIFDYLFKTRRKVKKKDIIQFYRNEYNTEIVTLSGLEED



QFNASFSTYQDLLKCGLTRAELDHPDNAEKLEDIIKILTIFEDRQRIRTQLSTFKG



QFSAEVLKKLERKHYTGWGRLSKKLINGIYDKESGKTILDYLVKDDGVSKHYN



RNFMQLINDSQLSFKNAIQKAQSSEHEETLSETVNELAGSPAIKKGIYQSLKIVD



ELVAIMGYAPKRIVVEMARENQTTSTGKRRSIQRLKIVEKAMAEIGSNLLKEQP



TTNEQLRDTRLFLYYMQNGKDMYTGDELSLHRLSHYDIDHIIPQSFMKDDSLD



NLVLVGSTENRGKSDDVPSKEVVKDMKAYWEKLYAAGLISQRKFQRLTKGEQ



GGLTLEDKAHFIQRQLVETRQITKNVAGILDQRYNAKSKEKKVQIITLKASLTSQ



FRSIFGLYKVREVNDYHHGQDAYLNCVVATTLLKVYPNLAPEFVYGEYPKFQT



FKENKATAKAIIYTNLLRFFTEDEPRFTKDGEILWSNSYLKTIKKELNYHQMNIV



KKVEVQKGGFSKESIKPKGPSNKLIPVKNGLDPQKYGGFDSPVVAYTVLFTHEK



GKKPLIKQEILGITIMEKTRFEQNPILFLEEKGFLRPRVLMKLPKYTLYEFPEGRR



RLLASAKEAQKGNQMVLPEHLLTLLYHAKQCLLPNQSESLAYVEQHQPEFQEI



LERVVDFAEVHTLAKSKVQQIVKLFEANQTADVKEIAASFIQLMQFNAMGAPS



TFKFFQKDIERARYTSIKEIFDATIIYQSPTGLYETRRKVVD (SEQ ID NO: 36)






Staphylococcus

KRNYILGLDIGITSVGYGIIDYETRDVIDAGVRLFKEANVENNEGRRSKRGARRL



aureus

KRRRRHRIQRVKKLLFDYNLLTDHSELSGINPYEARVKGLSQKLSEEEFSAALL


Cas9
HLAKRRGVHNVNEVEEDTGNELSTKEQISRNSKALEEKYVAELQLERLKKDGE



VRGSINRFKTSDYVKEAKQLLKVQKAYHQLDQSFIDTYIDLLETRRTYYEGPGE



GSPFGWKDIKEWYEMLMGHCTYFPEELRSVKYAYNADLYNALNDLNNLVITR



DENEKLEYYEKFQIIENVFKQKKKPTLKQIAKEILVNEEDIKGYRVTSTGKPEFT



NLKVYHDIKDITARKEIIENAELLDQIAKILTIYQSSEDIQEELTNLNSELTQEEIE



QISNLKGYTGTHNLSLKAINLILDELWHTNDNQIAIFNRLKLVPKKVDLSQQKEI



PTTLVDDFILSPVVKRSFIQSIKVINAIIKKYGLPNDIIIELAREKNSKDAQKMINE



MQKRNRQTNERIEEIIRTTGKENAKYLIEKIKLHDMQEGKCLYSLEAIPLEDLLN



NPFNYEVDHIIPRSVSFDNSFNNKVLVKQEENSKKGNRTPFQYLSSSDSKISYETF



KKHILNLAKGKGRISKTKKEYLLEERDINRFSVQKDFINRNLVDTRYATRGLMN



LLRSYFRVNNLDVKVKSINGGFTSFLRRKWKFKKERNKGYKHHAEDALIIANA



DFIFKEWKKLDKAKKVMENQMFEEKQAESMPEIETEQEYKEIFITPHQIKHIKDF



KDYKYSHRVDKKPNRELINDTLYSTRKDDKGNTLIVNNLNGLYDKDNDKLKK



LINKSPEKLLMYHHDPQTYQKLKLIMEQYGDEKNPLYKYYEETGNYLTKYSKK



DNGPVIKKIKYYGNKLNAHLDITDDYPNSRNKVVKLSLKPYRFDVYLDNGVYK



FVTVKNLDVIKKENYYEVNSKCYEEAKKLKKISNQAEFIASFYNNDLIKINGEL



YRVIGVNNDLLNRIEVNMIDITYREYLENMNDKRPPRIIKTIASKTQSIKKYSTDI



LGNLYEVKSKKHPQIIKKG



(SEQ ID NO: 37)






Geobacillus

MKYKIGLDIGITSIGWAVINLDIPRIEDLGVRIFDRAENPKTGESLALPRRLARSA



thermo-

RRRLRRRKHRLERIRRLFVREGILTKEELNKLFEKKHEIDVWQLRVEALDRKLN



denitrificans

NDELARILLHLAKRRGFRSNRKSERTNKENSTMLKHIEENQSILSSYRTVAEMV


Cas9
VKDPKFSLHKRNKEDNYTNTVARDDLEREIKLIFAKQREYGNIVCTEAFEHEYIS



IWASQRPFASKDDIEKKVGFCTFEPKEKRAPKATYTFQSFTVWEHINKLRLVSP



GGIRALTDDERRLIYKQAFHKNKITFHDVRTLLNLPDDTRFKGLLYDRNTTLKE



NEKVRFLELGAYHKIRKAIDSVYGKGAAKSFRPIDFDTFGYALTMFKDDTDIRS



YLRNEYEQNGKRMENLADKVYDEELIEELLNLSFSKFGHLSLKALRNILPYMEQ



GEVYSTACERAGYTFTGPKKKQKTVLLPNIPPIANPVVMRALTQARKVVNAIIK



KYGSPVSIHIELARELSQSFDERRKMQKEQEGNRKKNETAIRQLVEYGLTLNPT



GLDIVKFKLWSEQNGKCAYSLQPIEIERLLEPGYTEVDHVIPYSRSLDDSYTNKV



LVLTKENREKGNRTPAEYLGLGSERWQQFETFVLTNKQFSKKKRDRLLRLHYD



ENEENEFKNRNLNDTRYISRFLANFIREHLKFADSDDKQKVYTVNGRITAHLRS



RWNFNKNREESNLHHAVDAAIVACTTPSDIARVTAFYQRREQNKELSKKTDPQ



FPQPWPHFADELQARLSKNPKESIKALNLGNYDNEKLESLQPVFVSRMPKRSIT



GAAHQETLRRYIGIDERSGKIQTVVKKKLSEIQLDKTGHFPMYGKESDPRTYEAI



RQRLLEHNNDPKKAFQEPLYKPKKNGELGPIIRTIKIIDTTNQVIPLNDGKTVAY



NSNIVRVDVFEKDGKYYCVPIYTIDMMKGILPNKAIEPNKPYSEWKEMTEDYTF



RFSLYPNDLIRIEFPREKTIKTAVGEEIKIKDLFAYYQTIDSSNGGLSLVSHDNNFS



LRSIGSRTLKRFEKYQVDVLGNIYKVRGEKRVGVASSSHSKAGETIRPL



(SEQ ID NO: 38)





ScCas9
MEKKYSIGLDIGTNSVGWAVITDDYKVPSKKFKVLGNTNRKSIKKNLMGALLF



S. canis

DSGETAEATRLKRTARRRYTRRKNRIRYLQEIFANEMAKLDDSFFQRLEESFLV


1375 AA
EEDKKNERHPIFGNLADEVAYHRNYPTIYHLRKKLADSPEKADLRLIYLALAHII


159.2 kDa
KFRGHFLIEGKLNAENSDVAKLFYQLIQTYNQLFEESPLDEIEVDAKGILSARLS



KSKRLEKLIAVFPNEKKNGLFGNIIALALGLTPNFKSNFDLTEDAKLQLSKDTYD



DDLDELLGQIGDQYADLFSAAKNLSDAILLSDILRSNSEVTKAPLSASMVKRYD



EHHQDLALLKTLVRQQFPEKYAEIFKDDTKNGYAGYVGIGIKHRKRTTKLATQ



EEFYKFIKPILEKMDGAEELLAKLNRDDLLRKQRTFDNGSIPHQIHLKELHAILR



RQEEFYPFLKENREKIEKILTFRIPYYVGPLARGNSRFAWLTRKSEEAITPWNFEE



VVDKGASAQSFIERMTNFDEQLPNKKVLPKHSLLYEYFTVYNELTKVKYVTER



MRKPEFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIIGVEDRFN



ASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFD



DKVMKQLKRRHYTGWGRLSRKMINGIRDKQSGKTILDFLKSDGFSNRNFMQLI



HDDSLTFKEEIEKAQVSGQGDSLHEQIADLAGSPAIKKGILQTVKIVDELVKVM



GHKPENIVIEMARENQTTTKGLQQSRERKKRIEEGIKELESQILKENPVENTQLQ



NEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFIKDDSIDNKVLTRSV



ENRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSEADK



AGFIKRQLVETRQITKHVARILDSRMNTKRDKNDKPIREVKVITLKSKLVSDFRK



DFQLYKVRDINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRK



MIAKSEQEIGKATAKRFFYSNIMNFFKTEVKLANGEIRKRPLIETNGETGEVVW



NKEKDFATVRKVLAMPQVNIVKKTEVQTGGFSKESILSKRESAKLIPRKKGWD



TRKYGGFGSPTVAYSILVVAKVEKGKAKKLKSVKVLVGITIMEKGSYEKDPIGF



LEAKGYKDIKKELIFKLPKYSLFELENGRRRMLASATELQKANELVLPQHLVRL



LYYTQNISATTGSNNLGYIEQHREEFKEIFEKIIDFSEKYILKNKVNSNLKSSFDE



QFAVSDSILLSNSFVSLLKYTSFGASGGFTFLDLDVKQGRLRYQTVTEVLDATLI



YQSITGLYETRTDLSQLGGD (SEQ ID NO: 39)









The multi-flap prime editors described herein may include any of the above Cas9 ortholog sequences, or any variants thereof having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.


The napDNAbp may include any suitable homologs and/or orthologs or naturally occurring enzymes, such as, Cas9. Cas9 homologs and/or orthologs have been described in various species, including, but not limited to, S. pyogenes and S. thermophilus. Preferably, the Cas moiety is configured (e.g, mutagenized, recombinantly engineered, or otherwise obtained from nature) as a nickase, i.e., capable of cleaving only a single strand of the target double-stranded DNA. Additional suitable Cas9 nucleases and sequences will be apparent to those of skill in the art based on this disclosure, and such Cas9 nucleases and sequences include Cas9 sequences from the organisms and loci disclosed in Chylinski, Rhun, and Charpentier, “The tracrRNA and Cas9 families of type II CRISPR-Cas immunity systems” (2013) RNA Biology 10:5, 726-737; the entire contents of which are incorporated herein by reference. In some embodiments, a Cas9 nuclease has an inactive (e.g., an inactivated) DNA cleavage domain; that is, the Cas9 is a nickase. In some embodiments, the Cas9 protein comprises an amino acid sequence that is at least 80% identical to the amino acid sequence of a Cas9 protein as provided by any one of the variants of Table 3. In some embodiments, the Cas9 protein comprises an amino acid sequence that is at least 85%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to the amino acid sequence of a Cas9 protein as provided by any one of the Cas9 orthologs in the above tables.


C. Dead Cas9 Variant


In certain embodiments, the multi-flap prime editors described herein may include a dead Cas9, e.g., dead SpCas9, which has no nuclease activity due to one or more mutations that inactive both nuclease domains of Cas9, namely the RuvC domain (which cleaves the non-protospacer DNA strand) and HNH domain (which cleaves the protospacer DNA strand). The nuclease inactivation may be due to one or mutations that result in one or more substitutions and/or deletions in the amino acid sequence of the encoded protein, or any variants thereof having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.


As used herein, the term “dCas9” refers to a nuclease-inactive Cas9 or nuclease-dead Cas9, or a functional fragment thereof, and embraces any naturally occurring dCas9 from any organism, any naturally-occurring dCas9 equivalent or functional fragment thereof, any dCas9 homolog, ortholog, or paralog from any organism, and any mutant or variant of a dCas9, naturally-occurring or engineered. The term dCas9 is not meant to be particularly limiting and may be referred to as a “dCas9 or equivalent.” Exemplary dCas9 proteins and method for making dCas9 proteins are further described herein and/or are described in the art and are incorporated herein by reference.


In other embodiments, dCas9 corresponds to, or comprises in part or in whole, a Cas9 amino acid sequence having one or more mutations that inactivate the Cas9 nuclease activity. In other embodiments, Cas9 variants having mutations other than D10A and H840A are provided which may result in the full or partial inactivation of the endogeneous Cas9 nuclease activity (e.g., nCas9 or dCas9, respectively). Such mutations, by way of example, include other amino acid substitutions at D10 and H840, or other substitutions within the nuclease domains of Cas9 (e.g., substitutions in the HNH nuclease subdomain and/or the RuvC1 subdomain) with reference to a wild type sequence such as Cas9 from Streptococcus pyogenes (NCBI Reference Sequence: NC_017053.1). In some embodiments, variants or homologues of Cas9 (e.g., variants of Cas9 from Streptococcus pyogenes (NCBI Reference Sequence: NC_017053.1 (SEQ ID NO: 20))) are provided which are at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to NCBI Reference Sequence: NC_017053.1. In some embodiments, variants of dCas9 (e.g., variants of NCBI Reference Sequence: NC_017053.1 (SEQ ID NO: 20)) are provided having amino acid sequences which are shorter, or longer than NC_017053.1 (SEQ ID NO: 20) by about 5 amino acids, by about 10 amino acids, by about 15 amino acids, by about 20 amino acids, by about 25 amino acids, by about 30 amino acids, by about 40 amino acids, by about 50 amino acids, by about 75 amino acids, by about 100 amino acids or more.


In one embodiment, the dead Cas9 may be based on the canonical SpCas9 sequence of Q99ZW2 and may have the following sequence, which comprises a D10X and an H810X, wherein X may be any amino acid, substitutions (underlined and bolded), or a variant be variant of SEQ ID NO: 40 having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.


In one embodiment, the dead Cas9 may be based on the canonical SpCas9 sequence of Q99ZW2 and may have the following sequence, which comprises a D10A and an H810A substitutions (underlined and bolded), or be a variant of SEQ ID NO: 41 having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.














Description
Sequence
SEQ ID NO:







dead Cas9 or
MDKKYSIGLXIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 40


dCas9
KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




Streptococcus

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP




pyogenes

TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



Q99ZW2 Cas9
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE



with D10X and
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK



H810X
DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT



Where “X” is
KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN



any amino acid
GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDXIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






dead Cas9 or
MDKKYSIGLAIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 41


dCas9
KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




Streptococcus

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP




pyogenes

TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



Q99ZW2 Cas9
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE



with D10A and
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK



H810A
DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT




KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD









D. Cas9 Nickase Variant


In one embodiment, the multi-flap prime editors described herein comprise a Cas9 nickase. The term “Cas9 nickase” or “nCas9” refers to a variant of Cas9 which is capable of introducing a single-strand break in a double strand DNA molecule target. In some embodiments, the Cas9 nickase comprises only a single functioning nuclease domain. The wild type Cas9 (e.g., the canonical SpCas9) comprises two separate nuclease domains, namely, the RuvC domain (which cleaves the non-protospacer DNA strand) and HNH domain (which cleaves the protospacer DNA strand). In one embodiment, the Cas9 nickase comprises a mutation in the RuvC domain which inactivates the RuvC nuclease activity. For example, mutations in aspartate (D) 10, histidine (H) 983, aspartate (D) 986, or glutamate (E) 762, have been reported as loss-of-function mutations of the RuvC nuclease domain and the creation of a functional Cas9 nickase (e.g., Nishimasu et al., “Crystal structure of Cas9 in complex with guide RNA and target DNA,” Cell 156(5), 935-949, which is incorporated herein by reference). Thus, nickase mutations in the RuvC domain could include D10X, H983X, D986X, or E762X, wherein X is any amino acid other than the wild type amino acid. In certain embodiments, the nickase could be D10A, H983A, D986A, or E762A, or a combination thereof.


In various embodiments, the Cas9 nickase can have a mutation in the RuvC nuclease domain and have one of the following amino acid sequences, or a variant thereof having an amino acid sequence that has at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.














Description
Sequence
SEQ ID NO:







Cas9 nickase
MDKKYSIGLXIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 42



Streptococcus

KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with D10X,
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE



wherein X is
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK



any alternate
DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT



amino acid
KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






Cas9 nickase
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 43



Streptococcus

KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with E762X,
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE



wherein X is
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK



any alternate
DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT



amino acid
KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIXMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






Cas9 nickase
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 44


Streptococcus
KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with H983X,
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE



wherein X is
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK



any alternate
DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT



amino acid
KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHXAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






Cas9 nickase
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 45



Streptococcus

KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with D986X,
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE



wherein X is
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK



any alternate
DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT



amino acid
KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHXAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






Cas9 nickase
MDKKYSIGLAIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 46



Streptococcus

KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with D10A
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE




NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK




DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT




KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






Cas9 nickase
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 47



Streptococcus

KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with E762A
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE




NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK




DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT




KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIAMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






Cas9 nickase
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 48



Streptococcus

KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with H983A
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE




NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK




DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT




KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHAAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






Cas9 nickase
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 49



Streptococcus

KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with D986A
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE




NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK




DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT




KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHAAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD









In another embodiment, the Cas9 nickase comprises a mutation in the HNH domain which inactivates the HNH nuclease activity. For example, mutations in histidine (H) 840 or asparagine (R) 863 have been reported as loss-of-function mutations of the HNH nuclease domain and the creation of a functional Cas9 nickase (e.g., Nishimasu et al., “Crystal structure of Cas9 in complex with guide RNA and target DNA,” Cell 156(5), 935-949, which is incorporated herein by reference). Thus, nickase mutations in the HNH domain could include H840X and R863X, wherein X is any amino acid other than the wild type amino acid. In certain embodiments, the nickase could be H840A or R863A or a combination thereof.


In various embodiments, the Cas9 nickase can have a mutation in the HNH nuclease domain and have one of the following amino acid sequences, or a variant thereof having an amino acid sequence that has at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.














Description
Sequence
SEQ ID NO:







Cas9 nickase
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 50



Streptococcus

KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with H840X,
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE



wherein X is
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK



any alternate
DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT



amino acid
KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDXIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






Cas9 nickase
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 51



Streptococcus

KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE




pyogenes

MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with H840A
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE




NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK




DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT




KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSFL




KDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






Cas9 nickase
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 52


Streptococcus
KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE



pyogenes
MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with R863X,
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE



wherein X is
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK



any alternate
DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT



amino acid
KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNXGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






Cas9 nickase
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK
SEQ ID NO: 53


Streptococcus
KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNE



pyogenes
MAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYP



Q99ZW2 Cas9
TIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDN



with R863A
SDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE




NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSK




DTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEIT




KAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKN




GYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ




RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIP




YYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIE




RMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEIS




GVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFE




DREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIR




DKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENI




VIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENT




QLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFL




KDDSIDNKVLTRSDKNAGKSDNVPSEEVVKKMKNYWRQLLNA




KLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQI




LDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREIN




NYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI




AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGE




TGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILP




KRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSK




KLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKL




KGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV




LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR




YTSTKEVLDATLIHQSITGLYETRIDLSQLGGD









In some embodiments, the N-terminal methionine is removed from a Cas9 nickase, or from any Cas9 variant, ortholog, or equivalent disclosed or contemplated herein. For example, methionine-minus Cas9 nickases include the following sequences, or a variant thereof having an amino acid sequence that has at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity thereto.













Description
Sequence







Cas9 nickase
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSG


(Met minus)
ETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDK



Streptococcus

KHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRG



pyogenes

HFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRL


Q99ZW2 Cas9
ENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDN


with H840X,
LLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDL


wherein X is
TLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTE


any alternate
ELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKI


amino acid
LTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNF



DKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLL



FKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFL



DNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGR



LSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQ



GDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTT



QKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMY



VDQELDINRLSDYDVDXIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVK



KMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKH



VAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHA



HDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYF



FYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQV



NIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVV



AKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYS



LFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQK



QLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIH



LFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGG



D (SEQ ID NO: 54)





Cas9 nickase
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSG


(Met minus)
ETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDK



Streptococcus

KHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRG



pyogenes

HFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRL


Q99ZW2 Cas9
ENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDN


with H840A
LLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDL



TLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTE



ELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKI



LTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNF



DKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLL



FKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFL



DNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGR



LSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQ



GDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTT



QKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMY



VDQELDINRLSDYDVDAIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVK



KMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKH



VAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHA



HDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYF



FYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQV



NIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVV



AKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYS



LFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQK



QLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIH



LFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGG



D (SEQ ID NO: 55)





Cas9 nickase
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSG


(Met minus)
ETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDK



Streptococcus

KHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRG



pyogenes

HFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRL


Q99ZW2 Cas9
ENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDN


with R863X,
LLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDL


wherein X is
TLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTE


any alternate
ELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKI


amino acid
LTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNF



DKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLL



FKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFL



DNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGR



LSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQ



GDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTT



QKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMY



VDQELDINRLSDYDVDHIVPQSFLKDDSIDNKVLTRSDKNXGKSDNVPSEEVVK



KMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKH



VAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHA



HDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYF



FYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQV



NIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVV



AKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYS



LFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQK



QLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIH



LFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGG



D (SEQ ID NO: 56)





Cas9 nickase
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSG


(Met minus)
ETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDK



Streptococcus

KHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRG



pyogenes

HFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRL


Q99ZW2 Cas9
ENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDN


with R863A
LLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDL



TLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTE



ELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKI



LTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNF



DKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLL



FKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFL



DNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGR



LSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQ



GDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTT



QKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMY



VDQELDINRLSDYDVDHIVPQSFLKDDSIDNKVLTRSDKNAGKSDNVPSEEVVK



KMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKH



VAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHA



HDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYF



FYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQV



NIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVV



AKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYS



LFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQK



QLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIH



LFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGG



D (SEQ ID NO: 57)









E. Other Cas9 Variants


Besides dead Cas9 and Cas9 nickase variants, the Cas9 proteins used herein may also include other “Cas9 variants” having at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to any reference Cas9 protein, including any wild type Cas9, or mutant Cas9 (e.g., a dead Cas9 or Cas9 nickase), or fragment Cas9, or circular permutant Cas9, or other variant of Cas9 disclosed herein or known in the art. In some embodiments, a Cas9 variant may have 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 21, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 or more amino acid changes compared to a reference Cas9. In some embodiments, the Cas9 variant comprises a fragment of a reference Cas9 (e.g., a gRNA binding domain or a DNA-cleavage domain), such that the fragment is at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to the corresponding fragment of wild type Cas9. In some embodiments, the fragment is at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% identical, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% of the amino acid length of a corresponding wild type Cas9 (e.g., SEQ ID NO: 18).


In some embodiments, the disclosure also may utilize Cas9 fragments that retain their functionality and that are fragments of any herein disclosed Cas9 protein. In some embodiments, the Cas9 fragment is at least 100 amino acids in length. In some embodiments, the fragment is at least 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000, 1050, 1100, 1150, 1200, 1250, or at least 1300 amino acids in length.


In various embodiments, the multi-flap prime editors disclosed herein may comprise one of the Cas9 variants described as follows, or a Cas9 variant thereof having at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to any reference Cas9 variants.


F. Small-Sized Cas9 Variants


In some embodiments, the multi-flap prime editors contemplated herein can include a Cas9 protein that is of smaller molecular weight than the canonical SpCas9 sequence. In some embodiments, the smaller-sized Cas9 variants may facilitate delivery to cells, e.g., by an expression vector, nanoparticle, or other means of delivery. In certain embodiments, the smaller-sized Cas9 variants can include enzymes categorized as type II enzymes of the Class 2 CRISPR-Cas systems. In some embodiments, the smaller-sized Cas9 variants can include enzymes categorized as type V enzymes of the Class 2 CRISPR-Cas systems. In other embodiments, the smaller-sized Cas9 variants can include enzymes categorized as type VI enzymes of the Class 2 CRISPR-Cas systems.


The canonical SpCas9 protein is 1368 amino acids in length and has a predicted molecular weight of 158 kilodaltons. The term “small-sized Cas9 variant”, as used herein, refers to any Cas9 variant—naturally occurring, engineered, or otherwise—that is less than at least 1300 amino acids, or at least less than 1290 amino acids, or than less than 1280 amino acids, or less than 1270 amino acid, or less than 1260 amino acid, or less than 1250 amino acids, or less than 1240 amino acids, or less than 1230 amino acids, or less than 1220 amino acids, or less than 1210 amino acids, or less than 1200 amino acids, or less than 1190 amino acids, or less than 1180 amino acids, or less than 1170 amino acids, or less than 1160 amino acids, or less than 1150 amino acids, or less than 1140 amino acids, or less than 1130 amino acids, or less than 1120 amino acids, or less than 1110 amino acids, or less than 1100 amino acids, or less than 1050 amino acids, or less than 1000 amino acids, or less than 950 amino acids, or less than 900 amino acids, or less than 850 amino acids, or less than 800 amino acids, or less than 750 amino acids, or less than 700 amino acids, or less than 650 amino acids, or less than 600 amino acids, or less than 550 amino acids, or less than 500 amino acids, but at least larger than about 400 amino acids and retaining the required functions of the Cas9 protein. The Cas9 variants can include those categorized as type II, type V, or type VI enzymes of the Class 2 CRISPR-Cas system.


In various embodiments, the multi-flap prime editors disclosed herein may comprise one of the small-sized Cas9 variants described as follows, or a Cas9 variant thereof having at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to any reference small-sized Cas9 protein.














Description
Sequence
SEQ ID NO:







SaCas9
MGKRNYILGLDIGITSVGYGIIDYETRDVIDAGVRLFKEANVENN
SEQ ID NO: 58



Staphylococcus

EGRRSKRGARRLKRRRRHRIQRVKKLLFDYNLLTDHSELSGINPY




aureus

EARVKGLSQKLSEEEFSAALLHLAKRRGVHNVNEVEEDTGNELS



1053 AA
TKEQISRNSKALEEKYVAELQLERLKKDGEVRGSINRFKTSDYV



123 kDa
KEAKQLLKVQKAYHQLDQSFIDTYIDLLETRRTYYEGPGEGSPFG




WKDIKEWYEMLMGHCTYFPEELRSVKYAYNADLYNALNDLNN




LVITRDENEKLEYYEKFQIIENVFKQKKKPTLKQIAKEILVNEEDI




KGYRVTSTGKPEFTNLKVYHDIKDITARKEIIENAELLDQIAKILTI




YQSSEDIQEELTNLNSELTQEEIEQISNLKGYTGTHNLSLKAINLIL




DELWHTNDNQIAIFNRLKLVPKKVDLSQQKEIPTTLVDDFILSPV




VKRSFIQSIKVINAIIKKYGLPNDIIIELAREKNSKDAQKMINEMQK




RNRQTNERIEEIIRTTGKENAKYLIEKIKLHDMQEGKCLYSLEAIP




LEDLLNNPFNYEVDHIIPRSVSFDNSFNNKVLVKQEENSKKGNRT




PFQYLSSSDSKISYETFKKHILNLAKGKGRISKTKKEYLLEERDIN




RFSVQKDFINRNLVDTRYATRGLMNLLRSYFRVNNLDVKVKSIN




GGFTSFLRRKWKFKKERNKGYKHHAEDALIIANADFIFKEWKKL




DKAKKVMENQMFEEKQAESMPEIETEQEYKEIFITPHQIKHIKDF




KDYKYSHRVDKKPNRKLINDTLYSTRKDDKGNTLIVNNLNGLY




DKDNDKLKKLINKSPEKLLMYHHDPQTYQKLKLIMEQYGDEKN




PLYKYYEETGNYLTKYSKKDNGPVIKKIKYYGNKLNAHLDITDD




YPNSRNKVVKLSLKPYRFDVYLDNGVYKFVTVKNLDVIKKENY




YEVNSKCYEEAKKLKKISNQAEFIASFYKNDLIKINGELYRVIGV




NNDLLNRIEVNMIDITYREYLENMNDKRPPHIIKTIASKTQSIKKY




STDILGNLYEVKSKKHPQIIKK






NmeCas9
MAAFKPNSINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVF
SEQ ID NO: 59



N. meningitidis

ERAEVPKTGDSLAMARRLARSVRRLTRRRAHRLLRTRRLLKREG



1083 AA
VLQAANFDENGLIKSLPNTPWQLRAAALDRKLTPLEWSAVLLHL



124.5 kDa
IKHRGYLSQRKNEGETADKELGALLKGVAGNAHALQTGDFRTP




AELALNKFEKESGHIRNQRSDYSHTFSRKDLQAELILLFEKQKEF




GNPHVSGGLKEGIETLLMTQRPALSGDAVQKMLGHCTFEPAEPK




AAKNTYTAERFIWLTKLNNLRILEQGSERPLTDTERATLMDEPYR




KSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYH




AISRALEKEGLKDKKSPLNLSPELQDEIGTAFSLFKTDEDITGRLK




DRIQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRYDEACA




EIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGV




VRRYGSPARIHIETAREVGKSFKDRKEIEKRQEENRKDREKAAA




KFREYFPNFVGEPKSKDILKLRLYEQQHGKCLYSGKEINLGRLNE




KGYVEIDAALPFSRTWDDSFNNKVLVLGSENQNKGNQTPYEYF




NGKDNSREWQEFKARVETSRFPRSKKQRILLQKFDEDGFKERNL




NDTRYVNRFLCQFVADRMRLTGKGKKRVFASNGQITNLLRGFW




GLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRYKEMNA




FDGKTIDKETGEVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPE




FEEADTLEKLRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGQ




GHMETVKSAKRLDEGVSVLRVPLTQLKLKDLEKMVNREREPKL




YEALKARLEAHKDDPAKAFAEPFYKYDKAGNRTQQVKAVRVE




QVQKTGVWVRNHNGIADNATMVRVDVFEKGDKYYLVPIYSWQ




VAKGILPDRAVVQGKDEEDWQLIDDSFNFKFSLHPNDLVEVITK




KARMFGYFASCHRGTGNINIRIHDLDHKIGKNGILEGIGVKTALS




FQKYQIDELGKEIRPCRLKKRPPVR






CjCas9
MARILAFDIGISSIGWAFSENDELKDCGVRIFTKVENPKTGESLAL
SEQ ID NO: 60



C. jejuni

PRRLARSARKRLARRKARLNHLKHLIANEFKLNYEDYQSFDESL



984 AA
AKAYKGSLISPYELRFRALNELLSKQDFARVILHIAKRRGYDDIK



114.9 kDa
NSDDKEKGAILKAIKQNEEKLANYQSVGEYLYKEYFQKFKENSK




EFTNVRNKKESYERCIAQSFLKDELKLIFKKQREFGFSFSKKFEEE




VLSVAFYKRALKDFSHLVGNCSFFTDEKRAPKNSPLAFMFVALT




RIINLLNNLKNTEGILYTKDDLNALLNEVLKNGTLTYKQTKKLLG




LSDDYEFKGEKGTYFIEFKKYKEFIKALGEHNLSQDDLNEIAKDI




TLIKDEIKLKKALAKYDLNQNQIDSLSKLEFKDHLNISFKALKLV




TPLMLEGKKYDEACNELNLKVAINEDKKDFLPAFNETYYKDEVT




NPVVLRAIKEYRKVLNALLKKYGKVHKINIELAREVGKNHSQRA




KIEKEQNENYKAKKDAELECEKLGLKINSKNILKLRLFKEQKEFC




AYSGEKIKISDLQDEKMLEIDHIYPYSRSFDDSYMNKVLVFTKQN




QEKLNQTPFEAFGNDSAKWQKIEVLAKNLPTKKQKRILDKNYK




DKEQKNFKDRNLNDTRYIARLVLNYTKDYLDFLPLSDDENTKLN




DTQKGSKVHVEAKSGMLTSALRHTWGFSAKDRNNHLHHAIDA




VIIAYANNSIVKAFSDFKKEQESNSAELYAKKISELDYKNKRKFF




EPFSGFRQKVLDKIDEIFVSKPERKKPSGALHEETFRKEEEFYQSY




GGKEGVLKALELGKIRKVNGKIVKNGDMFRVDIFKHKKTNKFY




AVPIYTMDFALKVLPNKAVARSKKGEIKDWILMDENYEFCFSLY




KDSLILIQTKDMQEPEFVYYNAFTSSTVSLIVSKHDNKFETLSKN




QKILFKNANEKEVIAKSIGIQNLKVFEKYIVSALGEVTKAEFRQRE




DFKK






GeoCas9
MRYKIGLDIGITSVGWAVMNLDIPRIEDLGVRIFDRAENPQTGES
SEQ ID NO: 61



G.

LALPRRLARSARRRLRRRKHRLERIRRLVIREGILTKEELDKLFEE




stearo-

KHEIDVWQLRVEALDRKLNNDELARVLLHLAKRRGFKSNRKSE




thermophilus

RSNKENSTMLKHIEENRAILSSYRTVGEMIVKDPKFALHKRNKG



1087 AA
ENYTNTIARDDLEREIRLIFSKQREFGNMSCTEEFENEYITIWASQ



127 kDa
RPVASKDDIEKKVGFCTFEPKEKRAPKATYTFQSFIAWEHINKLR




LISPSGARGLTDEERRLLYEQAFQKNKITYHDIRTLLHLPDDTYFK




GIVYDRGESRKQNENIRFLELDAYHQIRKAVDKVYGKGKSSSFLP




IDFDTFGYALTLFKDDADIHSYLRNEYEQNGKRMPNLANKVYD




NELIEELLNLSFTKFGHLSLKALRSILPYMEQGEVYSSACERAGY




TFTGPKKKQKTMLLPNIPPIANPVVMRALTQARKVVNAIIKKYGS




PVSIHIELARDLSQTFDERRKTKKEQDENRKKNETAIRQLMEYGL




TLNPTGHDIVKFKLWSEQNGRCAYSLQPIEIERLLEPGYVEVDHV




IPYSRSLDDSYTNKVLVLTRENREKGNRIPAEYLGVGTERWQQF




ETFVLTNKQFSKKKRDRLLRLHYDENEETEFKNRNLNDTRYISRF




FANFIREHLKFAESDDKQKVYTVNGRVTAHLRSRWEFNKNREES




DLHHAVDAVIVACTTPSDIAKVTAFYQRREQNKELAKKTEPHFP




QPWPHFADELRARLSKHPKESIKALNLGNYDDQKLESLQPVFVS




RMPKRSVTGAAHQETLRRYVGIDERSGKIQTVVKTKLSEIKLDAS




GHFPMYGKESDPRTYEAIRQRLLEHNNDPKKAFQEPLYKPKKNG




EPGPVIRTVKIIDTKNQVIPLNDGKTVAYNSNIVRVDVFEKDGKY




YCVPVYTMDIMKGILPNKAIEPNKPYSEWKEMTEDYTFRFSLYP




NDLIRIELPREKTVKTAAGEEINVKDVFVYYKTIDSANGGLELISH




DHRFSLRGVGSRTLKRFEKYQVDVLGNIYKVRGEKRVGLASSAH




SKPGKTIRPLQSTRD






LbaCas12a
MSKLEKFTNCYSLSKTLRFKAIPVGKTQENIDNKRLLVEDEKRAE
SEQ ID NO: 62



L. bacterium

DYKGVKKLLDRYYLSFINDVLHSIKLKNLNNYISLFRKKTRTEKE



1228 AA
NKELENLEINLRKEIAKAFKGNEGYKSLFKKDIIETILPEFLDDKD



143.9 kDa
EIALVNSFNGFTTAFTGFFDNRENMFSEEAKSTSIAFRCINENLTR




YISNMDIFEKVDAIFDKHEVQEIKEKILNSDYDVEDFFEGEFFNFV




LTQEGIDVYNAIIGGFVTESGEKIKGLNEYINLYNQKTKQKLPKF




KPLYKQVLSDRESLSFYGEGYTSDEEVLEVFRNTLNKNSEIFSSIK




KLEKLFKNFDEYSSAGIFVKNGPAISTISKDIFGEWNVIRDKWNA




EYDDIHLKKKAVVTEKYEDDRRKSFKKIGSFSLEQLQEYADADL




SVVEKLKEIIIQKVDEIYKVYGSSEKLFDADFVLEKSLKKNDAVV




AIMKDLLDSVKSFENYIKAFFGEGKETNRDESFYGDFVLAYDILL




KVDHIYDAIRNYVTQKPYSKDKFKLYFQNPQFMGGWDKDKETD




YRATILRYGSKYYLAIMDKKYAKCLQKIDKDDVNGNYEKINYK




LLPGPNKMLPKVFFSKKWMAYYNPSEDIQKIYKNGTFKKGDMF




NLNDCHKLIDFFKDSISRYPKWSNAYDFNFSETEKYKDIAGFYRE




VEEQGYKVSFESASKKEVDKLVEEGKLYMFQIYNKDFSDKSHGT




PNLHTMYFKLLFDENNHGQIRLSGGAELFMRRASLKKEELVVHP




ANSPIANKNPDNPKKTTTLSYDVYKDKRFSEDQYELHIPIAINKCP




KNIFKINTEVRVLLKHDDNPYVIGIDRGERNLLYIVVVDGKGNIV




EQYSLNEIINNFNGIRIKTDYHSLLDKKEKERFEARQNWTSIENIK




ELKAGYISQVVHKICELVEKYDAVIALEDLNSGFKNSRVKVEKQ




VYQKFEKMLIDKLNYMVDKKSNPCATGGALKGYQITNKFESFK




SMSTQNGFIFYIPAWLTSKIDPSTGFVNLLKTKYTSIADSKKFISSF




DRIMYVPEEDLFEFALDYKNFSRTDADYIKKWKLYSYGNRIRIFR




NPKKNNVFDWEEVCLTSAYKELFNKYGINYQQGDIRALLCEQSD




KAFYSSFMALMSLMLQMRNSITGRTDVDFLISPVKNSDGIFYDSR




NYEAQENAILPKNADANGAYNIARKVLWAIGQFKKAEDEKLDK




VKIAISNKEWLEYAQTSVKH






BhCas12b
MATRSFILKIEPNEEVKKGLWKTHEVLNHGIAYYMNILKLIRQEA
SEQ ID NO: 63



B. hisashii

IYEHHEQDPKNPKKVSKAEIQAELWDFVLKMQKCNSFTHEVDK



1108 AA
DEVFNILRELYEELVPSSVEKKGEANQLSNKFLYPLVDPNSQSGK



130.4 kDa
GTASSGRKPRWYNLKIAGDPSWEEEKKKWEEDKKKDPLAKILG




KLAEYGLIPLFIPYTDSNEPIVKEIKWMEKSRNQSVRRLDKDMFI




QALERFLSWESWNLKVKEEYEKVEKEYKTLEERIKEDIQALKAL




EQYEKERQEQLLRDTLNTNEYRLSKRGLRGWREIIQKWLKMDE




NEPSEKYLEVFKDYQRKHPREAGDYSVYEFLSKKENHFIWRNHP




EYPYLYATFCEIDKKKKDAKQQATFTLADPINHPLWVRFEERSG




SNLNKYRILTEQLHTEKLKKKLTVQLDRLIYPTESGGWEEKGKV




DIVLLPSRQFYNQIFLDIEEKGKHAFTYKDESIKFPLKGTLGGARV




QFDRDHLRRYPHKVESGNVGRIYFNMTVNIEPTESPVSKSLKIHR




DDFPKVVNFKPKELTEWIKDSKGKKLKSGIESLEIGLRVMSIDLG




QRQAAAASIFEVVDQKPDIEGKLFFPIKGTELYAVHRASFNIKLPG




ETLVKSREVLRKAREDNLKLMNQKLNFLRNVLHFQQFEDITERE




KRVTKWISRQENSDVPLVYQDELIQIRELMYKPYKDWVAFLKQL




HKRLEVEIGKEVKHWRKSLSDGRKGLYGISLKNIDEIDRTRKFLL




RWSLRPTEPGEVRRLEPGQRFAIDQLNHLNALKEDRLKKMANTII




MHALGYCYDVRKKKWQAKNPACQIILFEDLSNYNPYEERSRFE




NSKLMKWSRREIPRQVALQGEIYGLQVGEVGAQFSSRFHAKTGS




PGIRCSVVTKEKLQDNRFFKNLQREGRLTLDKIAVLKEGDLYPD




KGGEKFISLSKDRKCVTTHADINAAQNLQKRFWTRTHGFYKVY




CKAYQVDGQTVYIPESKDQKQKIIEEFGEGYFILKDGVYEWVNA




GKLKIKKGSSKQSSSELVDSDILKDSFDLASELKGEKLMLYRDPS




GNVFPSDKWMAAGVFFGKLERILISKLTNQYSISTIEDDSSKQSM









G. Cas9 Equivalents


In some embodiments, the multi-flap prime editors described herein can include any Cas9 equivalent. As used herein, the term “Cas9 equivalent” is a broad term that encompasses any napDNAbp protein that serves the same function as Cas9 in the present multi-flap prime editors despite that its amino acid primary sequence and/or its three-dimensional structure may be different and/or unrelated from an evolutionary standpoint. Thus, while Cas9 equivalents include any Cas9 ortholog, homolog, mutant, or variant described or embraced herein that are evolutionarily related, the Cas9 equivalents also embrace proteins that may have evolved through convergent evolution processes to have the same or similar function as Cas9, but that do not necessarily have any similarity with regard to amino acid sequence and/or three-dimensional structure. The multi-flapprime editors described here embrace any Cas9 equivalent that would provide the same or similar function as Cas9 despite that the Cas9 equivalent may be based on a protein that arose through convergent evolution. For instance, if Cas9 refers to a type II enzyme of the CRISPR-Cas system, a Cas9 equivalent can refer to a type V or type VI enzyme of the CRISPR-Cas system.


For example, Cas12e (CasX) is a Cas9 equivalent that reportedly has the same function as Cas9 but which evolved through convergent evolution. Thus, the Cas12e (CasX) protein described in Liu et al., “CasX enzymes comprises a distinct family of RNA-guided genome editors,” Nature, 2019, Vol. 566: 218-223, is contemplated to be used with the multi-flap prime editors described herein. In addition, any variant or modification of Cas12e (CasX) is conceivable and within the scope of the present disclosure.


Cas9 is a bacterial enzyme that evolved in a wide variety of species. However, the Cas9 equivalents contemplated herein may also be obtained from archaea, which constitute a domain and kingdom of single-celled prokaryotic microbes different from bacteria.


In some embodiments, Cas9 equivalents may refer to Cas12e (CasX) or Cas12d (CasY), which have been described in, for example, Burstein et al., “New CRISPR-Cas systems from uncultivated microbes.” Cell Res. 2017 February 21. doi: 10.1038/cr.2017.21, the entire contents of which is hereby incorporated by reference. Using genome-resolved metagenomics, a number of CRISPR-Cas systems were identified, including the first reported Cas9 in the archaeal domain of life. This divergent Cas9 protein was found in little-studied nanoarchaea as part of an active CRISPR-Cas system. In bacteria, two previously unknown systems were discovered, CRISPR-Cas12e and CRISPR-Cas12d, which are among the most compact systems yet discovered. In some embodiments, Cas9 refers to Cas12e, or a variant of Cas12e. In some embodiments, Cas9 refers to a Cas12d, or a variant of Cas12d. It should be appreciated that other RNA-guided DNA binding proteins may be used as a nucleic acid programmable DNA binding protein (napDNAbp) and are within the scope of this disclosure. Also see Liu et al., “CasX enzymes comprises a distinct family of RNA-guided genome editors,” Nature, 2019, Vol. 566: 218-223. Any of these Cas9 equivalents are contemplated.


In some embodiments, the Cas9 equivalent comprises an amino acid sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to a naturally-occurring Cas12e (CasX) or Cas12d (CasY) protein. In some embodiments, the napDNAbp is a naturally-occurring Cas12e (CasX) or Cas12d (CasY) protein. In some embodiments, the napDNAbp comprises an amino acid sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to a wild-type Cas moiety or any Cas moiety provided herein.


In various embodiments, the nucleic acid programmable DNA binding proteins include, without limitation, Cas9 (e.g., dCas9 and nCas9), Cas12e (CasX), Cas12d (CasY), Cas12a (Cpf1), Cas12b1 (C2c1), Cas13a (C2c2), Cas12c (C2c3), Argonaute, and Cas12b1. One example of a nucleic acid programmable DNA-binding protein that has different PAM specificity than Cas9 is Clustered Regularly Interspaced Short Palindromic Repeats from Prevotella and Francisella 1 (i.e, Cas12a (Cpf1)). Similar to Cas9, Cas12a (Cpf1) is also a Class 2 CRISPR effector, but it is a member of type V subgroup of enzymes, rather than the type II subgroup. It has been shown that Cas12a (Cpf1) mediates robust DNA interference with features distinct from Cas9. Cas12a (Cpf1) is a single RNA-guided endonuclease lacking tracrRNA, and it utilizes a T-rich protospacer-adjacent motif (TTN, TTTN, or YTN). Moreover, Cpf1 cleaves DNA via a staggered DNA double-stranded break. Out of 16 Cpf1-family proteins, two enzymes from Acidaminococcus and Lachnospiraceae are shown to have efficient genome-editing activity in human cells. Cpf1 proteins are known in the art and have been described previously, for example Yamano et al., “Crystal structure of Cpf1 in complex with guide RNA and target DNA.” Cell (165) 2016, p. 949-962; the entire contents of which is hereby incorporated by reference.


In still other embodiments, the Cas protein may include any CRISPR associated protein, including but not limited to, Cas12a, Cas12b1, Cas1, Cas1B, Cas2, Cas3, Cas4, Cas5, Cas6, Cas7, Cas8, Cas9 (also known as Csn1 and Csx12), Cas10, Csy1, Csy2, Csy3, Cse1, Cse2, Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3, Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csx1, Csx15, Csf1, Csf2, Csf3, Csf4, homologs thereof, or modified versions thereof, and preferably comprising a nickase mutation (e.g., a mutation corresponding to the D10A mutation of the wild type Cas9 polypeptide of SEQ ID NO: 18).


In various other embodiments, the napDNAbp can be any of the following proteins: a Cas9, a Cas12a (Cpf1), a Cas12e (CasX), a Cas12d (CasY), a Cas12b1 (C2c1), a Cas13a (C2c2), a Cas12c (C2c3), a GeoCas9, a CjCas9, a Cas12g, a Cas12h, a Cas12i, a Cas13b, a Cas13c, a Cas13d, a Cas14, a Csn2, an xCas9, an SpCas9-NG, a circularly permuted Cas9, or an Argonaute (Ago) domain, or a variant thereof.


Exemplary Cas9 equivalent protein sequences can include the following:













Description
Sequence







AsCas12a
MTQFEGFTNLYQVSKTLRFELIPQGKTLKHIQEQGFIEEDKARNDHYKELKPIIDRI


(previously
YKTYADQCLQLVQLDWENLSAAIDSYRKEKTEETRNALIEEQATYRNAIHDYFIGR


known as Cpf1)
TDNLTDAINKRHAEIYKGLFKAELFNGKVLKQLGTVTTTEHENALLRSFDKFTTYF



Acidaminococcus

SGFYENRKNVFSAEDISTAIPHRIVQDNFPKFKENCHIFTRLITAVPSLREHFENVKK


sp.
AIGIFVSTSIEEVFSFPFYNQLLTQTQIDLYNQLLGGISREAGTEKIKGLNEVLNLAIQ


(strain
KNDETAHIIASLPHRFIPLFKQILSDRNTLSFILEEFKSDEEVIQSFCKYKTLLRNENV


BV3L6)
LETAEALFNELNSIDLTHIFISHKKLETISSALCDHWDTLRNALYERRISELTGKITKS


UniProtKB
AKEKVQRSLKHEDINLQEIISAAGKELSEAFKQKTSEILSHAHAALDQPLPTTLKKQ


U2UMQ6
EEKEILKSQLDSLLGLYHLLDWFAVDESNEVDPEFSARLTGIKLEMEPSLSFYNKA



RNYATKKPYSVEKFKLNFQMPTLASGWDVNKEKNNGAILFVKNGLYYLGIMPKQ



KGRYKALSFEPTEKTSEGFDKMYYDYFPDAAKMIPKCSTQLKAVTAHFQTHTTPIL



LSNNFIEPLEITKEIYDLNNPEKEPKKFQTAYAKKTGDQKGYREALCKWIDFTRDFL



SKYTKTTSIDLSSLRPSSQYKDLGEYYAELNPLLYHISFQRIAEKEIMDAVETGKLY



LFQIYNKDFAKGHHGKPNLHTLYWTGLFSPENLAKTSIKLNGQAELFYRPKSRMK



RMAHRLGEKMLNKKLKDQKTPIPDTLYQELYDYVNHRLSHDLSDEARALLPNVIT



KEVSHEIIKDRRFTSDKFFFHVPITLNYQAANSPSKFNQRVNAYLKEHPETPIIGIDR



GERNLIYITVIDSTGKILEQRSLNTIQQFDYQKKLDNREKERVAARQAWSVVGTIK



DLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGFKSKRTGIAEKAVYQQFEKMLI



DKLNCLVLKDYPAEKVGGVLNPYQLTDQFTSFAKMGTQSGFLFYVPAPYTSKIDP



LTGFVDPFVWKTIKNHESRKHFLEGFDFLHYDVKTGDFILHFKMNRNLSFQRGLP



GFMPAWDIVFEKNETQFDAKGTPFIAGKRIVPVIENHRFTGRYRDLYPANELIALLE



EKGIVFRDGSNILPKLLENDDSHAIDTMVALIRSVLQMRNSNAATGEDYINSPVRD



LNGVCFDSRFQNPEWPMDADANGAYHIALKGQLLLNHLKESKDLKLQNGISNQD



WLAYIQELRN (SEQ ID NO: 64)





AsCas12a
MTQFEGFTNLYQVSKTLRFELIPQGKTLKHIQEQGFIEEDKARNDHYKELKPIIDRI


nickase (e.g.,
YKTYADQCLQLVQLDWENLSAAIDSYRKEKTEETRNALIEEQATYRNAIHDYFIGR


R1226A)
TDNLTDAINKRHAEIYKGLFKAELFNGKVLKQLGTVTTTEHENALLRSFDKFTTYF



SGFYENRKNVFSAEDISTAIPHRIVQDNFPKFKENCHIFTRLITAVPSLREHFENVKK



AIGIFVSTSIEEVFSFPFYNQLLTQTQIDLYNQLLGGISREAGTEKIKGLNEVLNLAIQ



KNDETAHIIASLPHRFIPLFKQILSDRNTLSFILEEFKSDEEVIQSFCKYKTLLRNENV



LETAEALFNELNSIDLTHIFISHKKLETISSALCDHWDTLRNALYERRISELTGKITKS



AKEKVQRSLKHEDINLQEIISAAGKELSEAFKQKTSEILSHAHAALDQPLPTTLKKQ



EEKEILKSQLDSLLGLYHLLDWFAVDESNEVDPEFSARLTGIKLEMEPSLSFYNKA



RNYATKKPYSVEKFKLNFQMPTLASGWDVNKEKNNGAILFVKNGLYYLGIMPKQ



KGRYKALSFEPTEKTSEGFDKMYYDYFPDAAKMIPKCSTQLKAVTAHFQTHTTPIL



LSNNFIEPLEITKEIYDLNNPEKEPKKFQTAYAKKTGDQKGYREALCKWIDFTRDFL



SKYTKTTSIDLSSLRPSSQYKDLGEYYAELNPLLYHISFQRIAEKEIMDAVETGKLY



LFQIYNKDFAKGHHGKPNLHTLYWTGLFSPENLAKTSIKLNGQAELFYRPKSRMK



RMAHRLGEKMLNKKLKDQKTPIPDTLYQELYDYVNHRLSHDLSDEARALLPNVIT



KEVSHEIIKDRRFTSDKFFFHVPITLNYQAANSPSKFNQRVNAYLKEHPETPIIGIDR



GERNLIYITVIDSTGKILEQRSLNTIQQFDYQKKLDNREKERVAARQAWSVVGTIK



DLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGFKSKRTGIAEKAVYQQFEKMLI



DKLNCLVLKDYPAEKVGGVLNPYQLTDQFTSFAKMGTQSGFLFYVPAPYTSKIDP



LTGFVDPFVWKTIKNHESRKHFLEGFDFLHYDVKTGDFILHFKMNRNLSFQRGLP



GFMPAWDIVFEKNETQFDAKGTPFIAGKRIVPVIENHRFTGRYRDLYPANELIALLE



EKGIVFRDGSNILPKLLENDDSHAIDTMVALIRSVLQMANSNAATGEDYINSPVRD



LNGVCFDSRFQNPEWPMDADANGAYHIALKGQLLLNHLKESKDLKLQNGISNQD



WLAYIQELRN (SEQ ID NO: 65)





LbCas12a
MNYKTGLEDFIGKESLSKTLRNALIPTESTKIHMEEMGVIRDDELRAEKQQELKEI


(previously
MDDYYRTFIEEKLGQIQGIQWNSLFQKMEETMEDISVRKDLDKIQNEKRKEICCYF


known as Cpf1)
TSDKRFKDLFNAKLITDILPNFIKDNKEYTEEEKAEKEQTRVLFQRFATAFTNYFNQ



Lachnospiraceae

RRNNFSEDNISTAISFRIVNENSEIHLQNMRAFQRIEQQYPEEVCGMEEEYKDMLQE



bacterium

WQMKHIYSVDFYDRELTQPGIEYYNGICGKINEHMNQFCQKNRINKNDFRMKKL


GAM79
HKQILCKKSSYYEIPFRFESDQEVYDALNEFIKTMKKKEIIRRCVHLGQECDDYDLG


Ref Seq.
KIYISSNKYEQISNALYGSWDTIRKCIKEEYMDALPGKGEKKEEKAEAAAKKEEYR


WP_119623382.1
SIADIDKIISLYGSEMDRTISAKKCITEICDMAGQISIDPLVCNSDIKLLQNKEKTTEIK



TILDSFLHVYQWGQTFIVSDIIEKDSYFYSELEDVLEDFEGITTLYNHVRSYVTQKP



YSTVKFKLHFGSPTLANGWSQSKEYDNNAILLMRDQKFYLGIFNVRNKPDKQIIKG



HEKEEKGDYKKMIYNLLPGPSKMLPKVFITSRSGQETYKPSKHILDGYNEKRHIKS



SPKFDLGYCWDLIDYYKECIHKHPDWKNYDFHFSDTKDYEDISGFYREVEMQGY



QIKWTYISADEIQKLDEKGQIFLFQIYNKDFSVHSTGKDNLHTMYLKNLFSEENLK



DIVLKLNGEAELFFRKASIKTPIVHKKGSVLVNRSYTQTVGNKEIRVSIPEEYYTEIY



NYLNHIGKGKLSSEAQRYLDEGKIKSFTATKDIVKNYRYCCDHYFLHLPITINFKA



KSDVAVNERTLAYIAKKEDIHIIGIDRGERNLLYISVVDVHGNIREQRSFNIVNGYD



YQQKLKDREKSRDAARKNWEEIEKIKELKEGYLSMVIHYIAQLVVKYNAVVAME



DLNYGFKTGRFKVERQVYQKFETMLIEKLHYLVFKDREVCEEGGVLRGYQLTYIP



ESLKKVGKQCGFIFYVPAGYTSKIDPTTGFVNLFSFKNLTNRESRQDFVGKFDEIRY



DRDKKMFEFSFDYNNYIKKGTILASTKWKVYTNGTRLKRIVVNGKYTSQSMEVEL



TDAMEKMLQRAGIEYHDGKDLKGQIVEKGIEAEIIDIFRLTVQMRNSRSES EDREY



DRLISPVLNDKGEFFDTATADKTLPQDADANGAYCIALKGLYEVKQIKENWKENE



QFPRNKLVQDNKTWFDFMQKKRYL (SEQ ID NO: 66)





PcCas12a-
MAKNFEDFKRLYSLSKTLRFEAKPIGATLDNIVKSGLLDEDEHRAASYVKVKKLID


previously
EYHKVFIDRVLDDGCLPLENKGNNNSLAEYYESYVSRAQDEDAKKKFKEIQQNLR


known at
SVIAKKLTEDKAYANLFGNKLIESYKDKEDKKKIIDSDLIQFINTAESTQLDSMSQD


Cpf1
EAKELVKEFWGFVTYFYGFFDNRKNMYTAEEKSTGIAYRLVNENLPKFIDNIEAFN



Prevotella

RAITRPEIQENMGVLYSDFSEYLNVESIQEMFQLDYYNMLLTQKQIDVYNAIIGGK



copri

TDDEHDVKIKGINEYINLYNQQHKDDKLPKLKALFKQILSDRNAISWLPEEFNSDQ


Ref Seq.
EVLNAIKDCYERLAENVLGDKVLKSLLGSLADYSLDGIFIRNDLQLTDISQKMFGN


WP_119227726.1
WGVIQNAIMQNIKRVAPARKHKESEEDYEKRIAGIFKKADSFSISYINDCLNEADPN



NAYFVENYFATFGAVNTPTMQRENLFALVQNAYTEVAALLHSDYPTVKHLAQDK



ANVSKIKALLDAIKSLQHFVKPLLGKGDESDKDERFYGELASLWAELDTVTPLYN



MIRNYMTRKPYSQKKIKLNFENPQLLGGWDANKEKDYATIILRRNGLYYLAIMDK



DSRKLLGKAMPSDGECYEKMVYKFFKDVTTMIPKCSTQLKDVQAYFKVNTDDYV



LNSKAFNKPLTITKEVFDLNNVLYGKYKKFQKGYLTATGDNVGYTHAVNVWIKF



CMDFLNSYDSTCIYDFSSLKPESYLSLDAFYQDANLLLYKLSFARASVSYINQLVEE



GKMYLFQIYNKDFSEYSKGTPNMHTLYWKALFDERNLADVVYKLNGQAEMFYR



KKSIENTHPTHPANHPILNKNKDNKKKESLFDYDLIKDRRYTVDKFMFHVPITMNF



KSVGSENINQDVKAYLRHADDMHIIGIDRGERHLLYLVVIDLQGNIKEQYSLNEIV



NEYNGNTYHTNYHDLLDVREEERLKARQSWQTIENIKELKEGYLSQVIHKITQLM



VRYHAIVVLEDLSKGFMRSRQKVEKQVYQKFEKMLIDKLNYLVDKKTDVSTPGG



LLNAYQLTCKSDSSQKLGKQSGFLFYIPAWNTSKIDPVTGFVNLLDTHSLNSKEKI



KAFFSKFDAIRYNKDKKWFEFNLDYDKFGKKAEDTRTKWTLCTRGMRIDTFRNK



EKNSQWDNQEVDLTTEMKSLLEHYYIDIHGNLKDAISAQTDKAFFTGLLHILKLTL



QMRNSITGTETDYLVSPVADENGIFYDSRSCGNQLPENADANGAYNIARKGLMLIE



QIKNAEDLNNVKFDISNKAWLNFAQQKPYKNG (SEQ ID NO: 67)





ErCas12a-
MFSAKLISDILPEFVIHNNNYSASEKEEKTQVIKLFSRFATSFKDYFKNRANCFSAN


previously
DISSSSCHRIVNDNAEIFFSNALVYRRIVKNLSNDDINKISGDMKDSLKEMSLEEIYS


known at
YEKYGEFITQEGISFYNDICGKVNLFMNLYCQKNKENKNLYKLRKLHKQILCIADT


Cpf1
SYEVPYKFESDEEVYQSVNGFLDNISSKHIVERLRKIGENYNGYNLDKIYIVSKFYE



Eubacterium

SVSQKTYRDWETINTALEIHYNNILPGNGKSKADKVKKAVKNDLQKSITEINELVS



rectale

NYKLCPDDNIKAETYIHEISHILNNFEAQELKYNPEIHLVESELKASELKNVLDVIM


Ref Seq.
NAFHWCSVFMTEELVDKDNNFYAELEEIYDEIYPVISLYNLVRNYVTQKPYSTKKI


WP_119223642.1
KLNFGIPTLADGWSKSKEYSNNAIILMRDNLYYLGIFNAKNKPDKKIIEGNTSENK



GDYKKMIYNLLPGPNKMIPKVFLSSKTGVETYKPSAYILEGYKQNKHLKSSKDFDI



TFCHDLIDYFKNCIAIHPEWKNFGFDFSDTSTYEDISGFYREVELQGYKIDWTYISE



KDIDLLQEKGQLYLFQIYNKDFSKKSSGNDNLHTMYLKNLFSEENLKDIVLKLNGE



AEIFFRKSSIKNPIIHKKGSILVNRTYEAEEKDQFGNIQIVRKTIPENIYQELYKYFND



KSDKELSDEAAKLKNVVGHHEAATNIVKDYRYTYDKYFLHMPITINFKANKTSFI



NDRILQYIAKEKDLHVIGIDRGERNLIYVSVIDTCGNIVEQKSFNIVNGYDYQIKLK



QQEGARQIARKEWKEIGKIKEIKEGYLSLVIHEISKMVIKYNAIIAMEDLSYGFKKG



RFKVERQVYQKFETMLINKLNYLVFKDISITENGGLLKGYQLTYIPDKLKNVGHQC



GCIFYVPAAYTSKIDPTTGFVNIFKFKDLTVDAKREFIKKFDSIRYDSDKNLFCFTFD



YNNFITQNTVMSKSSWSVYTYGVRIKRRFVNGRFSNESDTIDITKDMEKTLEMTDI



NWRDGHDLRQDIIDYEIVQHIFEIFKLTVQMRNSLSELEDRDYDRLISPVLNENNIF



YDSAKAGDALPKDADANGAYCIALKGLYEIKQITENWKEDGKFSRDKLKISNKD



WFDFIQNKRYL (SEQ ID NO: 68)





CsCas12a-
MNYKTGLEDFIGKESLSKTLRNALIPTESTKIHMEEMGVIRDDELRAEKQQELKEI


previously
MDDYYRAFIEEKLGQIQGIQWNSLFQKMEETMEDISVRKDLDKIQNEKRKEICCYF


known at
TSDKRFKDLFNAKLITDILPNFIKDNKEYTEEEKAEKEQTRVLFQRFATAFTNYFNQ


Cpf1
RRNNFSEDNISTAISFRIVNENSEIHLQNMRAFQRIEQQYPEEVCGMEEEYKDMLQE



Clostridium

WQMKHIYLVDFYDRVLTQPGIEYYNGICGKINEHMNQFCQKNRINKNDFRMKKL


sp. AF34-
HKQILCKKSSYYEIPFRFESDQEVYDALNEFIKTMKEKEIICRCVHLGQKCDDYDLG


10BH
KIYISSNKYEQISNALYGSWDTIRKCIKEEYMDALPGKGEKKEEKAEAAAKKEEYR


Ref Seq.
SIADIDKIISLYGSEMDRTISAKKCITEICDMAGQISTDPLVCNSDIKLLQNKEKTTEI


WP_118538418.1
KTILDSFLHVYQWGQTFIVSDIIEKDSYFYSELEDVLEDFEGITTLYNHVRSYVTQK



PYSTVKFKLHFGSPTLANGWSQSKEYDNNAILLMRDQKFYLGIFNVRNKPDKQIIK



GHEKEEKGDYKKMIYNLLPGPSKMLPKVFITSRSGQETYKPSKHILDGYNEKRHIK



SSPKFDLGYCWDLIDYYKECIHKHPDWKNYDFHFSDTKDYEDISGFYREVEMQGY



QIKWTYISADEIQKLDEKGQIFLFQIYNKDFSVHSTGKDNLHTMYLKNLFSEENLK



DIVLKLNGEAELFFRKASIKTPVVHKKGSVLVNRSYTQTVGDKEIRVSIPEEYYTEI



YNYLNHIGRGKLSTEAQRYLEERKIKSFTATKDIVKNYRYCCDHYFLHLPITINFKA



KSDIAVNERTLAYIAKKEDIHIIGIDRGERNLLYISVVDVHGNIREQRSFNIVNGYDY



QQKLKDREKSRDAARKNWEEIEKIKELKEGYLSMVIHYIAQLVVKYNAVVAMED



LNYGFKTGRFKVERQVYQKFETMLIEKLHYLVFKDREVCEEGGVLRGYQLTYIPE



SLKKVGKQCGFIFYVPAGYTSKIDPTTGFVNLFSFKNLTNRESRQDFVGKFDEIRYD



RDKKMFEFSFDYNNYIKKGTMLASTKWKVYTNGTRLKRIVVNGKYTSQSMEVEL



TDAMEKMLQRAGIEYHDGKDLKGQIVEKGIEAEIIDIFRLTVQMRNSRSES EDREY



DRLISPVLNDKGEFFDTATADKTLPQDADANGAYCIALKGLYEVKQIKENWKENE



QFPRNKLVQDNKTWFDFMQKKRYL (SEQ ID NO: 69)





BhCas12b
MATRSFILKIEPNEEVKKGLWKTHEVLNHGIAYYMNILKLIRQEAIYEHHEQDPKN



Bacillus

PKKVSKAEIQAELWDFVLKMQKCNSFTHEVDKDEVFNILRELYEELVPSSVEKKG



hisashii

EANQLSNKFLYPLVDPNSQSGKGTASSGRKPRWYNLKIAGDPSWEEEKKKWEED


Ref Seq.
KKKDPLAKILGKLAEYGLIPLFIPYTDSNEPIVKEIKWMEKSRNQSVRRLDKDMFIQ


WP_095142515.1
ALERFLSWESWNLKVKEEYEKVEKEYKTLEERIKEDIQALKALEQYEKERQEQLL



RDTLNTNEYRLSKRGLRGWREIIQKWLKMDENEPSEKYLEVFKDYQRKHPREAG



DYSVYEFLSKKENHFIWRNHPEYPYLYATFCEIDKKKKDAKQQATFTLADPINHPL



WVRFEERSGSNLNKYRILTEQLHTEKLKKKLTVQLDRLIYPTESGGWEEKGKVDIV



LLPSRQFYNQIFLDIEEKGKHAFTYKDESIKFPLKGTLGGARVQFDRDHLRRYPHK



VESGNVGRIYFNMTVNIEPTESPVSKSLKIHRDDFPKVVNFKPKELTEWIKDSKGK



KLKSGIESLEIGLRVMSIDLGQRQAAAASIFEVVDQKPDIEGKLFFPIKGTELYAVH



RASFNIKLPGETLVKSREVLRKAREDNLKLMNQKLNFLRNVLHFQQFEDITEREKR



VTKWISRQENSDVPLVYQDELIQIRELMYKPYKDWVAFLKQLHKRLEVEIGKEVK



HWRKSLSDGRKGLYGISLKNIDEIDRTRKFLLRWSLRPTEPGEVRRLEPGQRFAIDQ



LNHLNALKEDRLKKMANTIIMHALGYCYDVRKKKWQAKNPACQIILFEDLSNYN



PYEERSRFENSKLMKWSRREIPRQVALQGEIYGLQVGEVGAQFSSRFHAKTGSPGI



RCSVVTKEKLQDNRFFKNLQREGRLTLDKIAVLKEGDLYPDKGGEKFISLSKDRKC



VTTHADINAAQNLQKRFWTRTHGFYKVYCKAYQVDGQTVYIPESKDQKQKIIEEF



GEGYFILKDGVYEWVNAGKLKIKKGSSKQSSSELVDSDILKDSFDLASELKGEKLM



LYRDPSGNVFPSDKWMAAGVFFGKLERILISKLTNQYSISTIEDDSSKQSM (SEQ ID



NO: 70)





ThCas12b
MSEKTTQRAYTLRLNRASGECAVCQNNSCDCWHDALWATHKAVNRGAKAFGD



Thermomonas

WLLTLRGGLCHTLVEMEVPAKGNNPPQRPTDQERRDRRVLLALSWLSVEDEHGA



hydrothermalis

PKEFIVATGRDSADDRAKKVEEKLREILEKRDFQEHEIDAWLQDCGPSLKAHIRED


Ref Seq.
AVWVNRRALFDAAVERIKTLTWEEAWDFLEPFFGTQYFAGIGDGKDKDDAEGPA


WP_072754838
RQGEKAKDLVQKAGQWLSARFGIGTGADFMSMAEAYEKIAKWASQAQNGDNGK



ATIEKLACALRPSEPPTLDTVLKCISGPGHKSATREYLKTLDKKSTVTQEDLNQLRK



LADEDARNCRKKVGKKGKKPWADEVLKDVENSCELTYLQDNSPARHREFSVML



DHAARRVSMAHSWIKKAEQRRRQFESDAQKLKNLQERAPSAVEWLDRFCESRSM



TTGANTGSGYRIRKRAIEGWSYVVQAWAEASCDTEDKRIAAARKVQADPEIEKFG



DIQLFEALAADEAICVWRDQEGTQNPSILIDYVTGKTAEHNQKRFKVPAYRHPDEL



RHPVFCDFGNSRWSIQFAIHKEIRDRDKGAKQDTRQLQNRHGLKMRLWNGRSMT



DVNLHWSSKRLTADLALDQNPNPNPTEVTRADRLGRAASSAFDHVKIKNVFNEKE



WNGRLQAPRAELDRIAKLEEQGKTEQAEKLRKRLRWYVSFSPCLSPSGPFIVYAG



QHNIQPKRSGQYAPHAQANKGRARLAQLILSRLPDLRILSVDLGHRFAAACAVWE



TLSSDAFRREIQGLNVLAGGSGEGDLFLHVEMTGDDGKRRTVVYRRIGPDQLLDN



TPHPAPWARLDRQFLIKLQGEDEGVREASNEELWTVHKLEVEVGRTVPLIDRMVR



SGFGKTEKQKERLKKLRELGWISAMPNEPSAETDEKEGEIRSISRSVDELMSSALGT



LRLALKRHGNRARIAFAMTADYKPMPGGQKYYFHEAKEASKNDDETKRRDNQIE



FLQDALSLWHDLFSSPDWEDNEAKKLWQNHIATLPNYQTPEEISAELKRVERNKK



RKENRDKLRTAAKALAENDQLRQHLHDTWKERWESDDQQWKERLRSLKDWIFP



RGKAEDNPSIRHVGGLSITRINTISGLYQILKAFKMRPEPDDLRKNIPQKGDDELEN



FNRRLLEARDRLREQRVKQLASRIIEAALGVGRIKIPKNGKLPKRPRTTVDTPCHAV



VIESLKTYRPDDLRTRRENRQLMQWSSAKVRKYLKEGCELYGLHFLEVPANYTSR



QCSRTGLPGIRCDDVPTGDFLKAPWWRRAINTAREKNGGDAKDRFLVDLYDHLN



NLQSKGEALPATVRVPRQGGNLFIAGAQLDDTNKERRAIQADLNAAANIGLRALL



DPDWRGRWWYVPCKDGTSEPALDRIEGSTAFNDVRSLPTGDNSSRRAPREIENLW



RDPSGDSLESGTWSPTRAYWDTVQSRVIELLRRHAGLPTS (SEQ ID NO: 71)





LsCas12b
MSIRSFKLKLKTKSGVNAEQLRRGLWRTHQLINDGIAYYMNWLVLLRQEDLFIRN



Laceyella

KETNEIEKRSKEEIQAVLLERVHKQQQRNQWSGEVDEQTLLQALRQLYEEIVPSVI



sacchari

GKSGNASLKARFFLGPLVDPNNKTTKDVSKSGPTPKWKKMKDAGDPNWVQEYE


WP_132221894.1
KYMAERQTLVRLEEMGLIPLFPMYTDEVGDIHWLPQASGYTRTWDRDMFQQAIE



RLLSWESWNRRVRERRAQFEKKTHDFASRFSESDVQWMNKLREYEAQQEKSLEE



NAFAPNEPYALTKKALRGWERVYHSWMRLDSAASEEAYWQEVATCQTAMRGEF



GDPAIYQFLAQKENHDIWRGYPERVIDFAELNHLQRELRRAKEDATFTLPDSVDHP



LWVRYEAPGGTNIHGYDLVQDTKRNLTLILDKFILPDENGSWHEVKKVPFSLAKS



KQFHRQVWLQEEQKQKKREVVFYDYSTNLPHLGTLAGAKLQWDRNFLNKRTQQ



QIEETGEIGKVFFNISVDVRPAVEVKNGRLQNGLGKALTVLTHPDGTKIVTGWKAE



QLEKWVGESGRVSSLGLDSLSEGLRVMSIDLGQRTSATVSVFEITKEAPDNPYKFF



YQLEGTEMFAVHQRSFLLALPGENPPQKIKQMREIRWKERNRIKQQVDQLSAILRL



HKKVNEDERIQAIDKLLQKVASWQLNEEIATAWNQALSQLYSKAKENDLQWNQA



IKNAHHQLEPVVGKQISLWRKDLSTGRQGIAGLSLWSIEELEATKKLLTRWSKRSR



EPGVVKRIERFETFAKQIQHHINQVKENRLKQLANLIVMTALGYKYDQEQKKWIE



VYPACQVVLFENLRSYRFSFERSRRENKKLMEWSHRSIPKLVQMQGELFGLQVAD



VYAAYSSRYHGRTGAPGIRCHALTEADLRNETNIIHELIEAGFIKEEHRPYLQQGDL



VPWSGGELFATLQKPYDNPRILTLHADINAAQNIQKRFWHPSMWFRVNCESVMEG



EIVTYVPKNKTVHKKQGKTFRFVKVEGSDVYEWAKWSKNRNKNTFSSITERKPPS



SMILFRDPSGTFFKEQEWVEQKTFWGKVQSMIQAYMKKTIVQRMEE (SEQ ID NO:



72)





DtCas12b
MVLGRKDDTAELRRALWTTHEHVNLAVAEVERVLLRCRGRSYWTLDRRGDPVH



Dsulfonatronum

VPESQVAEDALAMAREAQRRNGWPVVGEDEEILLALRYLYEQIVPSCLLDDLGKP



thiodismutans

LKGDAQKIGTNYAGPLFDSDTCRRDEGKDVACCGPFHEVAGKYLGALPEWATPIS


WP_031386437
KQEFDGKDASHLRFKATGGDDAFFRVSIEKANAWYEDPANQDALKNKAYNKDD



WKKEKDKGISSWAVKYIQKQLQLGQDPRTEVRRKLWLELGLLPLFIPVFDKTMVG



NLWNRLAVRLALAHLLSWESWNHRAVQDQALARAKRDELAALFLGMEDGFAGL



REYELRRNESIKQHAFEPVDRPYVVSGRALRSWTRVREEWLRHGDTQESRKNICN



RLQDRLRGKFGDPDVFHWLAEDGQEALWKERDCVTSFSLLNDADGLLEKRKGYA



LMTFADARLHPRWAMYEAPGGSNLRTYQIRKTENGLWADVVLLSPRNESAAVEE



KTFNVRLAPSGQLSNVSFDQIQKGSKMVGRCRYQSANQQFEGLLGGAEILFDRKRI



ANEQHGATDLASKPGHVWFKLTLDVRPQAPQGWLDGKGRPALPPEAKHFKTALS



NKSKFADQVRPGLRVLSVDLGVRSFAACSVFELVRGGPDQGTYFPAADGRTVDDP



EKLWAKHERSFKITLPGENPSRKEEIARRAAMEELRSLNGDIRRLKAILRLSVLQED



DPRTEHLRLFMEAIVDDPAKSALNAELFKGFGDDRFRSTPDLWKQHCHFFHDKAE



KVVAERFSRWRTETRPKSSSWQDWRERRGYAGGKSYWAVTYLEAVRGLILRWN



MRGRTYGEVNRQDKKQFGTVASALLHHINQLKEDRIKTGADMIIQAARGFVPRKN



GAGWVQVHEPCRLILFEDLARYRFRTDRSRRENSRLMRWSHREIVNEVGMQGEL



YGLHVDTTEAGFSSRYLASSGAPGVRCRHLVEEDFHDGLPGMHLVGELDWLLPK



DKDRTANEARRLLGGMVRPGMLVPWDGGELFATLNAASQLHVIHADINAAQNLQ



RRFWGRCGEAIRIVCNQLSVDGSTRYEMAKAPKARLLGALQQLKNGDAPFHLTSI



PNSQKPENSYVMTPTNAGKKYRAGPGEKSSGEEDELALDIVEQAEELAQGRKTFF



RDPSGVFFAPDRWLPSEIYWSRIRRRIWQVTLERNSSGRQERAEMDEMPY (SEQ ID



NO: 73)









The multi-flap prime editors described herein may also comprise Cas12a (Cpf1) (dCpf1) variants that may be used as a guide nucleotide sequence-programmable DNA-binding protein domain. The Cas12a (Cpf1) protein has a RuvC-like endonuclease domain that is similar to the RuvC domain of Cas9 but does not have a HNH endonuclease domain, and the N-terminal of Cas12a (Cpf1) does not have the alfa-helical recognition lobe of Cas9. It was shown in Zetsche et al., Cell, 163, 759-771, 2015 (which is incorporated herein by reference) that, the RuvC-like domain of Cas12a (Cpf1) is responsible for cleaving both DNA strands and inactivation of the RuvC-like domain inactivates Cas12a (Cpf1) nuclease activity.


In some embodiments, the napDNAbp is a single effector of a microbial CRISPR-Cas system. Single effectors of microbial CRISPR-Cas systems include, without limitation, Cas9, Cas12a (Cpf1), Cas12b1 (C2c1), Cas13a (C2c2), and Cas12c (C2c3). Typically, microbial CRISPR-Cas systems are divided into Class 1 and Class 2 systems. Class 1 systems have multi-subunit effector complexes, while Class 2 systems have a single protein effector. For example, Cas9 and Cas12a (Cpf1) are Class 2 effectors. In addition to Cas9 and Cas12a (Cpf1), three distinct Class 2 CRISPR-Cas systems (Cas12b1, Cas13a, and Cas12c) have been described by Shmakov et al., “Discovery and Functional Characterization of Diverse Class 2 CRISPR Cas Systems”, Mol. Cell, 2015 Nov. 5; 60(3): 385-397, the entire contents of which are hereby incorporated by reference.


Effectors of two of the systems, Cas12b1 and Cas12c, contain RuvC-like endonuclease domains related to Cas12a. A third system, Cas13a contains an effector with two predicted HEPN RNase domains. Production of mature CRISPR RNA is tracrRNA-independent, unlike production of CRISPR RNA by Cas12b1. Cas12b1 depends on both CRISPR RNA and tracrRNA for DNA cleavage. Bacterial Cas13a has been shown to possess a unique RNase activity for CRISPR RNA maturation distinct from its RNA-activated single-stranded RNA degradation activity. These RNase functions are different from each other and from the CRISPR RNA-processing behavior of Cas12a. See, e.g., East-Seletsky, et al., “Two distinct RNase activities of CRISPR-Cas13a enable guide-RNA processing and RNA detection”, Nature, 2016 Oct. 13; 538(7624):270-273, the entire contents of which are hereby incorporated by reference. In vitro biochemical analysis of Cas13a in Leptotrichia shahii has shown that Cas13a is guided by a single CRISPR RNA and can be programed to cleave ssRNA targets carrying complementary protospacers. Catalytic residues in the two conserved HEPN domains mediate cleavage. Mutations in the catalytic residues generate catalytically inactive RNA-binding proteins. See e.g., Abudayyeh et al., “C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector”, Science, 2016 Aug. 5; 353(6299), the entire contents of which are hereby incorporated by reference.


The crystal structure of Alicyclobaccillus acidoterrastris Cas12b1 (AacC2c1) has been reported in complex with a chimeric single-molecule guide RNA (sgRNA). See e.g., Liu et al., “C2c1-sgRNA Complex Structure Reveals RNA-Guided DNA Cleavage Mechanism”, Mol. Cell, 2017 Jan. 19; 65(2):310-322, the entire contents of which are hereby incorporated by reference. The crystal structure has also been reported in Alicyclobacillus acidoterrestris C2c1 bound to target DNAs as ternary complexes. See e.g., Yang et al., “PAM-dependent Target DNA Recognition and Cleavage by C2C1 CRISPR-Cas endonuclease”, Cell, 2016 Dec. 15; 167(7):1814-1828, the entire contents of which are hereby incorporated by reference. Catalytically competent conformations of AacC2c1, both with target and non-target DNA strands, have been captured independently positioned within a single RuvC catalytic pocket, with C2c1-mediated cleavage resulting in a staggered seven-nucleotide break of target DNA. Structural comparisons between C2c1 ternary complexes and previously identified Cas9 and Cpf1 counterparts demonstrate the diversity of mechanisms used by CRISPR-Cas9 systems.


In some embodiments, the napDNAbp may be a C2c1, a C2c2, or a C2c3 protein. In some embodiments, the napDNAbp is a C2c1 protein. In some embodiments, the napDNAbp is a Cas13a protein. In some embodiments, the napDNAbp is a Cas12c protein. In some embodiments, the napDNAbp comprises an amino acid sequence that is at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to a naturally-occurring Cas12b1 (C2c1), Cas13a (C2c2), or Cas12c (C2c3) protein. In some embodiments, the napDNAbp is a naturally-occurring Cas12b1 (C2c1), Cas13a (C2c2), or Cas12c (C2c3) protein.


H. Cas9 Circular Permutants


In various embodiments, the multi-flap prime editors disclosed herein may comprise a circular permutant of Cas9.


The term “circularly permuted Cas9” or “circular permutant” of Cas9 or “CP-Cas9”) refers to any Cas9 protein, or variant thereof, that occurs or has been modify to engineered as a circular permutant variant, which means the N-terminus and the C-terminus of a Cas9 protein (e.g., a wild type Cas9 protein) have been topically rearranged. Such circularly permuted Cas9 proteins, or variants thereof, retain the ability to bind DNA when complexed with a guide RNA (gRNA). See, Oakes et al., “Protein Engineering of Cas9 for enhanced function,” Methods Enzymol, 2014, 546: 491-511 and Oakes et al., “CRISPR-Cas9 Circular Permutants as Programmable Scaffolds for Genome Modification,” Cell, Jan. 10, 2019, 176: 254-267, each of are incorporated herein by reference. The instant disclosure contemplates any previously known CP-Cas9 or use a new CP-Cas9 so long as the resulting circularly permuted protein retains the ability to bind DNA when complexed with a guide RNA (gRNA).


Any of the Cas9 proteins described herein, including any variant, ortholog, or naturally occurring Cas9 or equivalent thereof, may be reconfigured as a circular permutant variant.


In various embodiments, the circular permutants of Cas9 may have the following structure: N-terminus-[original C-terminus]-[optional linker]-[original N-terminus]-C-terminus.


As an example, the present disclosure contemplates the following circular permutants of canonical S. pyogenes Cas9 (1368 amino acids of UniProtKB-Q99ZW2 (CAS9_STRP1) (numbering is based on the amino acid position in SEQ ID NO: 18)):

    • N-terminus-[1268-1368]-[optional linker]-[1-1267]-C-terminus;
    • N-terminus-[1168-1368]-[optional linker]-[1-1167]-C-terminus;
    • N-terminus-[1068-1368]-[optional linker]-[1-1067]-C-terminus;
    • N-terminus-[968-1368]-[optional linker]-[1-967]-C-terminus;
    • N-terminus-[868-1368]-[optional linker]-[1-867]-C-terminus;
    • N-terminus-[768-1368]-[optional linker]-[1-767]-C-terminus;
    • N-terminus-[668-1368]-[optional linker]-[1-667]-C-terminus;
    • N-terminus-[568-1368]-[optional linker]-[1-567]-C-terminus;
    • N-terminus-[468-1368]-[optional linker]-[1-467]-C-terminus;
    • N-terminus-[368-1368]-[optional linker]-[1-367]-C-terminus;
    • N-terminus-[268-1368]-[optional linker]-[1-267]-C-terminus;
    • N-terminus-[168-1368]-[optional linker]-[1-167]-C-terminus;
    • N-terminus-[68-1368]-[optional linker]-[1-67]-C-terminus; or
    • N-terminus-[10-1368]-[optional linker]-[1-9]-C-terminus, or the corresponding circular permutants of other Cas9 proteins (including other Cas9 orthologs, variants, etc).


In particular embodiments, the circular permuant Cas9 has the following structure (based on S. pyogenes Cas9 (1368 amino acids of UniProtKB-Q99ZW2 (CAS9_STRP1) (numbering is based on the amino acid position in SEQ ID NO: 18):

    • N-terminus-[102-1368]-[optional linker]-[1-101]-C-terminus;
    • N-terminus-[1028-1368]-[optional linker]-[1-1027]-C-terminus;
    • N-terminus-[1041-1368]-[optional linker]-[1-1043]-C-terminus;
    • N-terminus-[1249-1368]-[optional linker]-[1-1248]-C-terminus; or
    • N-terminus-[1300-1368]-[optional linker]-[1-1299]-C-terminus, or the corresponding circular permutants of other Cas9 proteins (including other Cas9 orthologs, variants, etc).


In still other embodiments, the circular permuant Cas9 has the following structure (based on S. pyogenes Cas9 (1368 amino acids of UniProtKB-Q99ZW2 (CAS9_STRP1) (numbering is based on the amino acid position in SEQ ID NO: 18):

    • N-terminus-[103-1368]-[optional linker]-[1-102]-C-terminus;
    • N-terminus-[1029-1368]-[optional linker]-[1-1028]-C-terminus;
    • N-terminus-[1042-1368]-[optional linker]-[1-1041]-C-terminus;
    • N-terminus-[1250-1368]-[optional linker]-[1-1249]-C-terminus; or
    • N-terminus-[1301-1368]-[optional linker]-[1-1300]-C-terminus, or the corresponding circular permutants of other Cas9 proteins (including other Cas9 orthologs, variants, etc).


In some embodiments, the circular permutant can be formed by linking a C-terminal fragment of a Cas9 to an N-terminal fragment of a Cas9, either directly or by using a linker, such as an amino acid linker. In some embodiments, The C-terminal fragment may correspond to the C-terminal 95% or more of the amino acids of a Cas9 (e.g., amino acids about 1300-1368), or the C-terminal 90%, 85%, 80%, 75%, 70%, 65%, 60%, 55%, 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, or 5% or more of a Cas9 (e.g., any one of SEQ ID NOs: 77-86). The N-terminal portion may correspond to the N-terminal 95% or more of the amino acids of a Cas9 (e.g., amino acids about 1-1300), or the N-terminal 90%, 85%, 80%, 75%, 70%, 65%, 60%, 55%, 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, or 5% or more of a Cas9 (e.g., of SEQ ID NO: 18).


In some embodiments, the circular permutant can be formed by linking a C-terminal fragment of a Cas9 to an N-terminal fragment of a Cas9, either directly or by using a linker, such as an amino acid linker. In some embodiments, the C-terminal fragment that is rearranged to the N-terminus includes or corresponds to the C-terminal 30% or less of the amino acids of a Cas9 (e.g., amino acids 1012-1368 of SEQ ID NO: 18). In some embodiments, the C-terminal fragment that is rearranged to the N-terminus, includes or corresponds to the C-terminal 30%, 29%, 28%, 27%, 26%, 25%, 24%, 23%, 22%, 21%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% of the amino acids of a Cas9 (e.g., the Cas9 of SEQ ID NO: 18). In some embodiments, the C-terminal fragment that is rearranged to the N-terminus, includes or corresponds to the C-terminal 410 residues or less of a Cas9 (e.g., the Cas9 of SEQ ID NO: 18). In some embodiments, the C-terminal portion that is rearranged to the N-terminus, includes or corresponds to the C-terminal 410, 400, 390, 380, 370, 360, 350, 340, 330, 320, 310, 300, 290, 280, 270, 260, 250, 240, 230, 220, 210, 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 90, 80, 70, 60, 50, 40, 30, 20, or 10 residues of a Cas9 (e.g., the Cas9 of SEQ ID NO: 18). In some embodiments, the C-terminal portion that is rearranged to the N-terminus includes or corresponds to the C-terminal 357, 341, 328, 120, or 69 residues of a Cas9 (e.g., the Cas9 of SEQ ID NO: 18).


In other embodiments, circular permutant Cas9 variants may be defined as a topological rearrangement of a Cas9 primary structure based on the following method, which is based on S. pyogenes Cas9 of SEQ ID NO: 18: (a) selecting a circular permutant (CP) site corresponding to an internal amino acid residue of the Cas9 primary structure, which dissects the original protein into two halves: an N-terminal region and a C-terminal region; (b) modifying the Cas9 protein sequence (e.g., by genetic engineering techniques) by moving the original C-terminal region (comprising the CP site amino acid) to precede the original N-terminal region, thereby forming a new N-terminus of the Cas9 protein that now begins with the CP site amino acid residue. The CP site can be located in any domain of the Cas9 protein, including, for example, the helical-II domain, the RuvCIII domain, or the CTD domain. For example, the CP site may be located (relative the S. pyogenes Cas9 of SEQ ID NO: 18) at original amino acid residue 181, 199, 230, 270, 310, 1010, 1016, 1023, 1029, 1041, 1247, 1249, or 1282. Thus, once relocated to the N-terminus, original amino acid 181, 199, 230, 270, 310, 1010, 1016, 1023, 1029, 1041, 1247, 1249, or 1282 would become the new N-terminal amino acid. Nomenclature of these CP-Cas9 proteins may be referred to as Cas9-CP181, Cas9-CP199, Cas9-CP230, Cas9-CP270, Cas9-CP310, Cas9-CP1010, Cas9-CP1016, Cas9-CP1023, Cas9-CP1029, Cas9-CP1041, Cas9-CP1247, Cas9-CP1249, and Cas9-CP1282, respectively. This description is not meant to be limited to making CP variants from SEQ ID NO: 18, but may be implemented to make CP variants in any Cas9 sequence, either at CP sites that correspond to these positions, or at other CP sites entirely. This description is not meant to limit the specific CP sites in any way. Virtually any CP site may be used to form a CP-Cas9 variant.


Exemplary CP-Cas9 amino acid sequences, based on the Cas9 of SEQ ID NO: 18, are provided below in which linker sequences are indicated by underlining and optional methionine (M) residues are indicated in bold. It should be appreciated that the disclosure provides CP-Cas9 sequences that do not include a linker sequence or that include different linker sequences. It should be appreciated that CP-Cas9 sequences may be based on Cas9 sequences other than that of SEQ ID NO: 18 and any examples provided herein are not meant to be limiting. Exemplary CP-Cas9 sequences are as follows:














CP name
Sequence
SEQ ID NO:







CP1012
DYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLA
SEQ ID NO: 77



NGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIV




KKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDS




PTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPI




DFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGEL




QKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQ




HKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIRE




QAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATL




IHQSITGLYETRIDLSQLGGDGGSGGSGGSGGSGGSGGSGGD




KKYSIGLAIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIK




KNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFS




NEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYH




EKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIE




GDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILS




ARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNF




DLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNL




SDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALV




RQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEK




MDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILR




RQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMT




RKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKV




LPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAI




VDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNAS




LGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIE




ERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDK




QSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSG




QGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKP




ENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKE




HPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDV




DHIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKM




KNYWRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQ




LVETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKSKL




VSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPK




LESEFVYG






CP1028
EIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETG
SEQ ID NO: 78



EIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESIL




PKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEK




GKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKD




LIIKLPKYSLFELENGRKRMLASAGELQKGNELALPSKYVNF




LYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFS




KRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLG




APAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDL




SQLGGDGGSGGSGGSGGSGGSGGSGGMDKKYSIGLAIGTNS




VGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSG




ETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSF




FHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLR




KKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSD




VDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRL




ENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKL




QLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDI




LRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEK




YKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEE




LLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFY




PFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETI




TPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLL




YEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLFKT




NRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHD




LLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTY




AHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTIL




DFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLH




EHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIE




MARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVEN




TQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVP




QSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYW




RQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVET




RQITKHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDF




RKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLES




EFVYGDYKVYDVRKMIAKSEQ






CP1041
NIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFAT
SEQ ID NO: 79



VRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKK




DWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLKSVKELL




GITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELE




NGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKG




SPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDK




VLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTI




DRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDGGSGG





SGGSGGSGGSGGSGGDKKYSIGLAIGTNSVGWAVITDEYKV





PSKKFKVLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTAR




RRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDK




KHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLR




LIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQ




LFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKNG




LFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDN




LLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSAS




MIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAG




YIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQR




TFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFR




IPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASA




QSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKY




VTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYF




KKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFLDNEE




NEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKR




RRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFM




QLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKG




ILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKN




SRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQN




GRDMYVDQELDINRLSDYDVDHIVPQSFLKDDSIDNKVLTR




SDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDN




LTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMN




TKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNY




HHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRK




MIAKSEQEIGKATAKYFFYS






CP1249
PEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV
SEQ ID NO: 80



LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDR




KRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDGGSGGSG





GSGGSGGSGGSGG
MDKKYSIGLAIGTNSVGWAVITDEYKV





PSKKFKVLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTAR




RRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDK




KHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLR




LIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQ




LFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKNG




LFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDN




LLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSAS




MIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAG




YIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQR




TFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFR




IPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASA




QSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKY




VTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYF




KKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFLDNEE




NEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKR




RRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFM




QLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKG




ILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKN




SRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQN




GRDMYVDQELDINRLSDYDVDHIVPQSFLKDDSIDNKVLTR




SDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDN




LTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMN




TKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNY




HHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRK




MIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLI




ETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTG




GFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVL




VVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKG




YKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNELA




LPSKYVNFLYLASHYEKLKGS






CP1300
KPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEV
SEQ ID NO: 81



LDATLIHQSITGLYETRIDLSQLGGDGGSGGSGGSGGSGGSG





GSGGDKKYSIGLAIGTNSVGWAVITDEYKVPSKKFKVLGNT





DRHSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRIC




YLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNI




VDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIK




FRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASG




VDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLG




LTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYA




DLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHH




QDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQE




EFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPH




QIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPL




ARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMT




NFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK




PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDS




VEISGVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIV




LTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWG




RLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSL




TFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVV




DELVKVMGRHKPENIVIEMARENQTTQKGQKNSRERMKRI




EEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVD




QELDINRLSDYDVDHIVPQSFLKDDSIDNKVLTRSDKNRGKS




DNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGG




LSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDK




LIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYL




NAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIG




KATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIV




WDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKR




NSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKS




KKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIK




LPKYSLFELENGRKRMLASAGELQKGNELALPSKYVNFLYL




ASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVI




LADANLDKVLSAYNKHRD









The Cas9 circular permutants may be useful in the multi-flap prime editing constructs described herein. Exemplary C-terminal fragments of Cas9, based on the Cas9 of SEQ ID NO: 18, which may be rearranged to an N-terminus of Cas9, are provided below. It should be appreciated that such C-terminal fragments of Cas9 are exemplary and are not meant to be limiting. These exemplary CP-Cas9 fragments have the following sequences:














CP name
Sequence
SEQ ID NO:







CP1012 C-
DYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITL
SEQ ID NO: 82


terminal
ANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVN



fragment
IVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGF




DSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEK




NPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAG




ELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFV




EQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKP




IREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLD




ATLIHQSITGLYETRIDLSQLGGD






CP1028 C-
EIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETG
SEQ ID NO: 83


terminal
EIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESIL



fragment
PKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEK




GKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKD




LIIKLPKYSLFELENGRKRMLASAGELQKGNELALPSKYVNF




LYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFS




KRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLG




APAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDL




SQLGGD






CP1041 C-
NIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFAT
SEQ ID NO: 84


terminal
VRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKK



fragment
DWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLKSVKELL




GITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELE




NGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKG




SPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDK




VLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTI




DRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






CP1249 C-
PEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKV
SEQ ID NO: 85


terminal
LSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDR



fragment
KRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGD






CP1300 C-
KPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEV
SEQ ID NO: 86


terminal
LDATLIHQSITGLYETRIDLSQLGGD



fragment









I. Cas9 Variants with Modified PAM Specificities


The multi-flap prime editors of the present disclosure may also comprise Cas9 variants with modified PAM specificities. Some aspects of this disclosure provide Cas9 proteins that exhibit activity on a target sequence that does not comprise the canonical PAM (5′-NGG-3′, where N is A, C, G, or T) at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NGG-3′ PAM sequence at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NNG-3′ PAM sequence at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NNA-3′ PAM sequence at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NNC-3′ PAM sequence at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NNT-3′ PAM sequence at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NGT-3′ PAM sequence at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NGA-3′ PAM sequence at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NGC-3′ PAM sequence at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NAA-3′ PAM sequence at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NAC-3′ PAM sequence at its 3′-end. In some embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NAT-3′ PAM sequence at its 3′-end. In still other embodiments, the Cas9 protein exhibits activity on a target sequence comprising a 5′-NAG-3′ PAM sequence at its 3′-end.


It should be appreciated that any of the amino acid mutations described herein, (e.g., A262T) from a first amino acid residue (e.g., A) to a second amino acid residue (e.g., T) may also include mutations from the first amino acid residue to an amino acid residue that is similar to (e.g., conserved) the second amino acid residue. For example, mutation of an amino acid with a hydrophobic side chain (e.g., alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, or tryptophan) may be a mutation to a second amino acid with a different hydrophobic side chain (e.g., alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, or tryptophan). For example, a mutation of an alanine to a threonine (e.g., a A262T mutation) may also be a mutation from an alanine to an amino acid that is similar in size and chemical properties to a threonine, for example, serine. As another example, mutation of an amino acid with a positively charged side chain (e.g., arginine, histidine, or lysine) may be a mutation to a second amino acid with a different positively charged side chain (e.g., arginine, histidine, or lysine). As another example, mutation of an amino acid with a polar side chain (e.g., serine, threonine, asparagine, or glutamine) may be a mutation to a second amino acid with a different polar side chain (e.g., serine, threonine, asparagine, or glutamine). Additional similar amino acid pairs include, but are not limited to, the following: phenylalanine and tyrosine; asparagine and glutamine; methionine and cysteine; aspartic acid and glutamic acid; and arginine and lysine. The skilled artisan would recognize that such conservative amino acid substitutions will likely have minor effects on protein structure and are likely to be well tolerated without compromising function. In some embodiments, any amino of the amino acid mutations provided herein from one amino acid to a threonine may be an amino acid mutation to a serine. In some embodiments, any amino of the amino acid mutations provided herein from one amino acid to an arginine may be an amino acid mutation to a lysine. In some embodiments, any amino of the amino acid mutations provided herein from one amino acid to an isoleucine, may be an amino acid mutation to an alanine, valine, methionine, or leucine. In some embodiments, any amino of the amino acid mutations provided herein from one amino acid to a lysine may be an amino acid mutation to an arginine. In some embodiments, any amino of the amino acid mutations provided herein from one amino acid to an aspartic acid may be an amino acid mutation to a glutamic acid or asparagine. In some embodiments, any amino of the amino acid mutations provided herein from one amino acid to a valine may be an amino acid mutation to an alanine, isoleucine, methionine, or leucine. In some embodiments, any amino of the amino acid mutations provided herein from one amino acid to a glycine may be an amino acid mutation to an alanine. It should be appreciated, however, that additional conserved amino acid residues would be recognized by the skilled artisan and any of the amino acid mutations to other conserved amino acid residues are also within the scope of this disclosure.


In some embodiments, the Cas9 protein comprises a combination of mutations that exhibit activity on a target sequence comprising a 5′-NAA-3′ PAM sequence at its 3′-end. In some embodiments, the combination of mutations are present in any one of the clones listed in Table 1. In some embodiments, the combination of mutations are conservative mutations of the clones listed in Table 1. In some embodiments, the Cas9 protein comprises the combination of mutations of any one of the Cas9 clones listed in Table 1.









TABLE 1





NAA PAM Clones


Mutations from wild-type SpCas9 (e.g., SEQ ID NO: 18)















D177N, K218R, D614N, D1135N, P1137S, E1219V, A1320V, A1323D, R1333K


D177N, K218R, D614N, D1135N, E1219V, Q1221H, H1264Y, A1320V, R1333K


A10T, I322V, S409I, E427G, G715C, D1135N, E1219V, Q1221H, H1264Y, A1320V, R1333K


A367T, K710E, R1114G, D1135N, P1137S, E1219V, Q1221H, H1264Y, A1320V, R1333K


A10T, I322V, S409I, E427G, R753G, D861N, D1135N, K1188R, E1219V, Q1221H, H1264H,


A1320V, R1333K


A10T, I322V, S409I, E427G, R654L, V743I, R753G, M1021T, D1135N,


D1180G, K1211R, E1219V, Q1221H, H1264Y, A1320V, R1333K


A10T, I322V, S409I, E427G, V743I, R753G, E762G, D1135N, D1180G,


K1211R, E1219V, Q1221H, H1264Y, A1320V, R1333K


A10T, I322V, S409I, E427G, R753G, D1135N, D1180G, K1211R, E1219V, Q1221H, H1264Y,


S1274R, A1320V, R1333K


A10T, I322V, S409I, E427G, A589S, R753G, D1135N, E1219V, Q1221H, H1264H, A1320V,


R1333K


A10T, I322V, S409I, E427G, R753G, E757K, G865G, D1135N, E1219V, Q1221H, H1264Y,


A1320V, R1333K


A10T, I322V, S409I, E427G, R654L, R753G, E757K, D1135N, E1219V, Q1221H,


H1264Y, A1320V, R1333K


A10T, I322V, S409I, E427G, K599R, M631A, R654L, K673E, V743I, R753G, N758H, E762G,


D1135N, D1180G, E1219V, Q1221H, Q1256R, H1264Y, A1320V, A1323D, R1333K


A10T, I322V, S409I, E427G, R654L, K673E, V743I, R753G, E762G, N869S, N1054D, R1114G,


D1135N, D1180G, E1219V, Q1221H, H1264Y, A1320V, A1323D, R1333K


A10T, I322V, S409I, E427G, R654L, L727I, V743I, R753G, E762G, R859S, N946D, F1134L,


D1135N, D1180G, E1219V, Q1221H, H1264Y, N1317T, A1320V, A1323D, R1333K


A10T, I322V, S409I, E427G, R654L, K673E, V743I, R753G, E762G, N803S, N869S, Y1016D,


G1077D, R1114G, F1134L, D1135N, D1180G, E1219V, Q1221H, H1264Y, V1290G, L1318S,


A1320V, A1323D, R1333K


A10T, I322V, S409I, E427G, R654L, K673E, V743I, R753G, E762G, N803S, N869S, Y1016D,


G1077D, R1114G, F1134L, D1135N, K1151E, D1180G, E1219V, Q1221H, H1264Y, V1290G,


L1318S, A1320V, R1333K


A10T, I322V, S409I, E427G, R654L, K673E, V743I, R753G, E762G, N803S, N869S, Y1016D,


G1077D, R1114G, F1134L, D1135N, D1180G, E1219V, Q1221H, H1264Y, V1290G, L1318S,


A1320V, A1323D, R1333K


A10T, I322V, S409I, E427G, R654L, K673E, F693L, V743I, R753G, E762G, N803S, N869S,


L921P, Y1016D, G1077D, F1080S, R1114G, D1135N, D1180G, E1219V, Q1221H,


H1264Y, L1318S, A1320V, A1323D, R1333K


A10T, I322V, S409I, E427G, E630K, R654L, K673E, V743I, R753G, E762G, Q768H, N803S,


N869S, Y1016D, G1077D, R1114G, F1134L, D1135N, D1180G, E1219V, Q1221H, H1264Y,


L1318S, A1320V, R1333K


A10T, I322V, S409I, E427G, R654L, K673E, F693L, V743I, R753G, E762G,


Q768H, N803S, N869S, Y1016D, G1077D, R1114G, F1134L, D1135N, D1180G, E1219V,


Q1221H, G1223S, H1264Y, L1318S, A1320V, R1333K


A10T, I322V, S409I, E427G, R654L, K673E, F693L, V743I, R753G, E762G, N803S,


N869S, L921P, Y1016D, G1077D, F1801S, R1114G, D1135N, D1180G, E1219V,


Q1221H, H1264Y, L1318S, A1320V, A1323D, R1333K


A10T, I322V, S409I, E427G, R654L, V743I, R753G, M1021T, D1135N, D1180G,


K1211R, E1219V, Q1221H, H1264Y, A1320V, R1333K


A10T, I322V, S409I, E427G, R654L, K673E, V743I, R753G, E762G, M673I, N803S, N869S,


G1077D, R1114G, D1135N, V1139A, D1180G, E1219V, Q1221H, A1320V, R1333K


A10T, I322V, S409I, E427G, R654L, K673E, V743I, R753G, E762G, N803S, N869S, R1114G,


D1135N, E1219V, Q1221H, A1320V, R1333K









In some embodiments, the Cas9 protein comprises an amino acid sequence that is at least 80% identical to the amino acid sequence of a Cas9 protein as provided by any one of the variants of Table 1. In some embodiments, the Cas9 protein comprises an amino acid sequence that is at least 85%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to the amino acid sequence of a Cas9 protein as provided by any one of the variants of Table 1.


In some embodiments, the Cas9 protein exhibits an increased activity on a target sequence that does not comprise the canonical PAM (5′-NGG-3′) at its 3′ end as compared to Streptococcus pyogenes Cas9 as provided by SEQ ID NO: 18. In some embodiments, the Cas9 protein exhibits an activity on a target sequence having a 3′ end that is not directly adjacent to the canonical PAM sequence (5′-NGG-3′) that is at least 5-fold increased as compared to the activity of Streptococcus pyogenes Cas9 as provided by SEQ ID NO: 18 on the same target sequence. In some embodiments, the Cas9 protein exhibits an activity on a target sequence that is not directly adjacent to the canonical PAM sequence (5′-NGG-3′) that is at least 10-fold, at least 50-fold, at least 100-fold, at least 500-fold, at least 1,000-fold, at least 5,000-fold, at least 10,000-fold, at least 50,000-fold, at least 100,000-fold, at least 500,000-fold, or at least 1,000,000-fold increased as compared to the activity of Streptococcus pyogenes as provided by SEQ ID NO: 18 on the same target sequence. In some embodiments, the 3′ end of the target sequence is directly adjacent to an AAA, GAA, CAA, or TAA sequence. In some embodiments, the Cas9 protein comprises a combination of mutations that exhibit activity on a target sequence comprising a 5′-NAC-3′ PAM sequence at its 3′-end. In some embodiments, the combination of mutations are present in any one of the clones listed in Table 2. In some embodiments, the combination of mutations are conservative mutations of the clones listed in Table 2. In some embodiments, the Cas9 protein comprises the combination of mutations of any one of the Cas9 clones listed in Table 2.









TABLE 2





NAC PAM Clones


MUTATIONS FROM WILD-TYPE SPCAS9 (E.G., SEQ ID NO: 18)















T472I, R753G, K890E, D1332N, R1335Q, T1337N


I1057S, D1135N, P1301S, R1335Q, T1337N


T472I, R753G, D1332N, R1335Q, T1337N


D1135N, E1219V, D1332N, R1335Q, T1337N


T472I, R753G, K890E, D1332N, R1335Q, T1337N


I1057S, D1135N, P1301S, R1335Q, T1337N


T472I, R753G, D1332N, R1335Q, T1337N


T472I, R753G, Q771H, D1332N, R1335Q, T1337N


E627K, T638P, K652T, R753G, N803S, K959N, R1114G, D1135N, E1219V, D1332N, R1335Q,


T1337N


E627K, T638P, K652T, R753G, N803S, K959N, R1114G, D1135N, K1156E, E1219V, D1332N,


R1335Q, T1337N


E627K, T638P, V647I, R753G, N803S, K959N, G1030R, I1055E, R1114G, D1135N, E1219V,


D1332N, R1335Q, T1337N


E627K, E630G, T638P, V647A, G687R, N767D, N803S, K959N, R1114G, D1135N,


E1219V, D1332G, R1335Q, T1337N


E627K, T638P, R753G, N803S, K959N, R1114G, D1135N, E1219V, N1266H, D1332N, R1335Q,


T1337N


E627K, T638P, R753G, N803S, K959N, I1057T, R1114G, D1135N, E1219V, D1332N, R1335Q,


T1337N


E627K, T638P, R753G, N803S, K959N, R1114G, D1135N, E1219V, D1332N, R1335Q, T1337N


E627K, M631I, T638P, R753G, N803S, K959N, Y1036H, R1114G, D1135N, E1219V, D1251G,


D1332G, R1335Q, T1337N


E627K, T638P, R753G, N803S, V875I, K959N, Y1016C, R1114G, D1135N, E1219V, D1251G,


D1332G, R1335Q, T1337N, I1348V


K608R, E627K, T638P, V647I, R654L, R753G, N803S, T804A, K848N, V922A, K959N, R1114G,


D1135N, E1219V, D1332N, R1335Q, T1337N


K608R, E627K, T638P, V647I, R753G, N803S, V922A, K959N, K1014N, V1015A,


R1114G, D1135N, K1156N, E1219V, N1252D, D1332N, R1335Q, T1337N


K608R, E627K, R629G, T638P, V647I, A711T, R753G, K775R, K789E, N803S, K959N, V1015A,


Y1036H, R1114G, D1135N, E1219V, N1286H, D1332N, R1335Q, T1337N


K608R, E627K, T638P, V647I, T740A, R753G, N803S, K948E, K959N, Y1016S,


R1114G, D1135N, E1219V, N1286H, D1332N, R1335Q, T1337N


K608R, E627K, T638P, V647I, T740A, N803S, K948E, K959N, Y1016S, R1114G,


D1135N, E1219V, N1286H, D1332N, R1335Q, T1337N


I670S, K608R, E627K, E630G, T638P, V647I, R653K, R753G, I795L, K797N, N803S, K866R,


K890N, K959N, Y1016C, R1114G, D1135N, E1219V, D1332N, R1335Q, T1337N


K608R, E627K, T638P, V647I, T740A, G752R, R753G, K797N, N803S, K948E, K959N, V1015A,


Y1016S, R1114G, D1135N, E1219V, N1266H, D1332N, R1335Q, T1337N


I570T, A589V, K608R, E627K, T638P, V647I, R654L, Q716R, R753G, N803S, K948E, K959N,


Y1016S, R1114G, D1135N, E1207G, E1219V, N1234D, D1332N, R1335Q, T1337N


K608R, E627K, R629G, T638R V647I, R654L, Q740R, R753G, N803S, K959N, N990S, T995S,


V1015A, Y1036D, R1114G, D1135N, E1207G, E1219V, N1234D, N1266H, D1332N, R1335Q,


T1337N


I562F, V565D, I570T, K608R, L625S, E627K, T638P, V647I, R654I, G752R, R753G, N803S,


N808D, K959N, M1021L, R1114G, D1135N, N1177S, N1234D, D1332N, R1335Q, T1337N


I562F, I570T, K608R, E627K, T638P, V647I, R753G, E790A, N803S, K959N, V1015A, Y1036H,


R1114G, D1135N, D1180E, A1184T, E1219V, D1332N, R1335Q, T1337N


I570T, K608R, E627K, T638P, V647I, R654H, R753G, E790A, N803S, K959N, V1015A, R1114G,


D1127A, D1135N, E1219V, D1332N, R1335Q, T1337N


I570T, K608R, L625S, E627K, T638P, V647I, R654I, T703P, R753G, N803S, N808D, K959N,


M1021L, R1114G, D1135N, E1219V, D1332N, R1335Q, T1337N


I570S, K608R, E627K, E630G, T638P, V647I, R653K, R753G, I795L, N803S, K866R, K890N,


K959N, Y1016C, R1114G, D1135N, E1219V, D1332N, R1335Q, T1337N


I570T, K608R, E627K, T638P, V647I, R654H, R753G, E790A, N803S, K959N, V1016A, R1114G,


D1135N, E1219V, K1246E, D1332N, R1335Q, T1337N


K608R, E627K, T638P, V647I, R654L, K673E, R753G, E790A, N803S, K948E, K959N, R1114G,


D1127G, D1135N, D1180E, E1219V, N1286H, D1332N, R1335Q, T1337N


K608R, L625S, E627K, T638P, V647I, R654I, I670T, R753G, N803S, N808D, K959N, M1021L,


R1114G, D1135N, E1219V, N1286H, D1332N, R1335Q, T1337N


E627K, M631V, T638P, V647I, K710E, R753G, N803S, N808D, K948E, M1021L,


R1114G, D1135N, E1219V, D1332N, R1335Q, T1337N, S1338T, H1349R









In some embodiments, the Cas9 protein comprises an amino acid sequence that is at least 80% identical to the amino acid sequence of a Cas9 protein as provided by any one of the variants of Table 2. In some embodiments, the Cas9 protein comprises an amino acid sequence that is at least 85%, at least 90%, at least 92%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% identical to the amino acid sequence of a Cas9 protein as provided by any one of the variants of Table 2.


In some embodiments, the Cas9 protein exhibits an increased activity on a target sequence that does not comprise the canonical PAM (5′-NGG-3′) at its 3′ end as compared to Streptococcus pyogenes Cas9 as provided by SEQ ID NO: 18. In some embodiments, the Cas9 protein exhibits an activity on a target sequence having a 3′ end that is not directly adjacent to the canonical PAM sequence (5′-NGG-3′) that is at least 5-fold increased as compared to the activity of Streptococcus pyogenes Cas9 as provided by SEQ ID NO: 18 on the same target sequence. In some embodiments, the Cas9 protein exhibits an activity on a target sequence that is not directly adjacent to the canonical PAM sequence (5′-NGG-3′) that is at least 10-fold, at least 50-fold, at least 100-fold, at least 500-fold, at least 1,000-fold, at least 5,000-fold, at least 10,000-fold, at least 50,000-fold, at least 100,000-fold, at least 500,000-fold, or at least 1,000,000-fold increased as compared to the activity of Streptococcus pyogenes as provided by SEQ ID NO: 18 on the same target sequence. In some embodiments, the 3′ end of the target sequence is directly adjacent to an AAC, GAC, CAC, or TAC sequence.


In some embodiments, the Cas9 protein comprises a combination of mutations that exhibit activity on a target sequence comprising a 5′-NAT-3′ PAM sequence at its 3′-end. In some embodiments, the combination of mutations are present in any one of the clones listed in Table 3. In some embodiments, the combination of mutations are conservative mutations of the clones listed in Table 3. In some embodiments, the Cas9 protein comprises the combination of mutations of any one of the Cas9 clones listed in Table 3.









TABLE 3





NAT PAM Clones


MUTATIONS FROM WILD-TYPE SPCAS9 (E.G., SEQ ID NO: 18)















K961E, H985Y, D1135N, K1191N, E1219V, Q1221H, A1320A, P1321S, R1335L


D1135N, G1218S, E1219V, Q1221H, P1249S, P1321S, D1322G, R1335L


V743I, R753G, E790A, D1135N, G1218S, E1219V, Q1221H, A1227V, P1249S, N1286K, A1293T,


P1321S, D1322G, R1335L, T1339I


F575S, M631L, R654L, V748I, V743I, R753G, D853E, V922A, R1114G D1135N, G1218S,


E1219V, Q1221H, A1227V, P1249S, N1286K, A1293T, P1321S, D1322G, R1335L, T1339I


F575S, M631L, R654L, R664K, R753G, D853E, V922A, R1114G D1135N, D1180G, G1218S,


E1219V, Q1221H, P1249S, N1286K, P1321S, D1322G, R1335L


M631L, R654L, R753G, K797E, D853E, V922A, D1012A, R1114G D1135N, G1218S, E1219V,


Q1221H, P1249S, N1317K, P1321S, D1322G, R1335L


F575S, M631L, R654L, R664K, R753G, D853E, V922A, R1114G, Y1131C, D1135N, D1180G,


G1218S, E1219V, Q1221H, P1249S, P1321S, D1322G, R1335L


F575S, M631L, R654L, R664K, R753G, D853E, V922A, R1114G, Y1131C, D1135N, D1180G,


G1218S, E1219V, Q1221H, P1249S, P1321S, D1322G, R1335L


F575S, D596Y, M631L, R654L, R664K, R753G, D853E, V922A, R1114G, Y1131C,


D1135N, D1180G, G1218S, E1219V, Q1221H, P1249S, Q1256R, P1321S, D1322G, R1335L


F575S, M631L, R654L, R664K, K710E, V750A, R753G, D853E, V922A, R1114G,


Y1131C, D1135N, D1180G, G1218S, E1219V, Q1221H, P1249S, P1321S, D1322G, R1335L


F575S, M631L, K649R, R654L, R664K, R753G, D853E, V922A, R1114G, Y1131C,


D1135N, K1156E, D1180G, G1218S, E1219V, Q1221H, P1249S, P1321S, D1322G, R1335L


F575S, M631L, R654L, R664K, R753G, D853E, V922A, R1114G, Y1131C, D1135N, D1180G,


G1218S, E1219V, Q1221H, P1249S, P1321S, D1322G, R1335L


F575S, M631L, R654L, R664K, R753G, D853E, V922A, I1057G, R1114G, Y1131C, D1135N,


D1180G, G1218S, E1219V, Q1221H, P1249S, N1308D, P1321S, D1322G, R1335L


M631L, R654L, R753G, D853E, V922A, R1114G, Y1131C, D1135N, E1150V, D1180G, G1218S,


E1219V, Q1221H, P1249S, P1321S, D1332G, R1335L


M631L, R654L, R664K, R753G, D853E, 11057V, Y1131C, D1135N, D1180G, G1218S, E1219V,


Q1221H, P1249S, P1321S, D1332G, R1335L


M631L, R654L, R664K, R753G, 11057V, R1114G, Y1131C, D1135N, D1180G, G1218S, E1219V,


Q1221H, P1249S, P1321S, D1332G, R1335L









The above description of various napDNAbps which can be used in connection with the presently disclosed multi-flap prime editors is not meant to be limiting in any way. The multi-flap prime editors may comprise the canonical SpCas9, or any ortholog Cas9 protein, or any variant Cas9 protein—including any naturally occurring variant, mutant, or otherwise engineered version of Cas9—that is known or which can be made or evolved through a directed evolutionary or otherwise mutagenic process. In various embodiments, the Cas9 or Cas9 varants have a nickase activity, i.e., only cleave of strand of the target DNA sequence. In other embodiments, the Cas9 or Cas9 variants have inactive nucleases, i.e., are “dead” Cas9 proteins. Other variant Cas9 proteins that may be used are those having a smaller molecular weight than the canonical SpCas9 (e.g., for easier delivery) or having modified or rearranged primary amino acid structure (e.g., the circular permutant formats). The multi-flap prime editors described herein may also comprise Cas9 equivalents, including Cas12a/Cpf1 and Cas12b proteins which are the result of convergent evolution. The napDNAbps used herein (e.g., SpCas9, Cas9 variant, or Cas9 equivalents) may also contain various modifications that alter/enhance their PAM specifities. Lastly, the application contemplates any Cas9, Cas9 variant, or Cas9 equivalent which has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.9% sequence identity to a reference Cas9 sequence, such as a references SpCas9 canonical sequences or a reference Cas9 equivalent (e.g., Cas12a/Cpf1).


In a particular embodiment, the Cas9 variant having expanded PAM capabilities is SpCas9 (H840A) VRQR (SEQ ID NO: 87), which has the following amino acid sequence (with the V, R, Q, R substitutions relative to the SpCas9 (H840A) of SEQ ID NO: 51 being show in bold underline. In addition, the methionine residue in SpCas9 (H840) was removed for SpCas9 (H840A) VRQR):










(SEQ ID NO: 87)



DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSGETAEATRLK






RTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHE





KYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQ





LFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLA





EDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIK





RYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTE





ELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVG





PLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYE





YFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVE





ISGVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDD





KVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDI





QKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQ





KGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLS





DYDVDAIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRK





FDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKS





KLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI





AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVL





SMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFVSPTVAYSVLVVAKVE





KGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRML





ASARELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSK





RVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKQYRSTKEV





LDATLIHQSITGLYETRIDLSQLGGD






In another particular embodiment, the Cas9 variant having expanded PAM capabilities is SpCas9 (H840A) VRER, which has the following amino acid sequence (with the V, R, E, R substitutions relative to the SpCas9 (H840A) of SEQ ID NO: 51 being shown in bold underline. In addition, the methionine residue in SpCas9 (H840) was removed for SpCas9 (H840A) VRER):










(SEQ ID NO: 88)



DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSGETAEATRLK






RTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHE





KYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQ





LFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLA





EDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIK





RYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTE





ELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVG





PLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYE





YFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVE





ISGVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDD





KVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDI





QKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQ





KGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLS





DYDVDAIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRK





FDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKS





KLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMI





AKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVL





SMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFVSPTVAYSVLVVAKVE





KGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRML





ASARELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSK





RVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKEYRSTKEV





LDATLIHQSITGLYETRIDLSQLGGD






In some embodiments, the napDNAbp that functions with a non-canonical PAM sequence is an Argonaute protein. One example of such a nucleic acid programmable DNA binding protein is an Argonaute protein from Natronobacterium gregoryi (NgAgo). NgAgo is a ssDNA-guided endonuclease. NgAgo binds 5′ phosphorylated ssDNA of ˜24 nucleotides (gDNA) to guide it to its target site and will make DNA double-strand breaks at the gDNA site. In contrast to Cas9, the NgAgo-gDNA system does not require a protospacer-adjacent motif (PAM). Using a nuclease inactive NgAgo (dNgAgo) can greatly expand the bases that may be targeted. The characterization and use of NgAgo have been described in Gao et al., Nat Biotechnol., 2016 July; 34(7):768-73. PubMed PMID: 27136078; Swarts et al., Nature. 507(7491) (2014):258-61; and Swarts et al., Nucleic Acids Res. 43(10) (2015):5120-9, each of which is incorporated herein by reference.


In some embodiments, the napDNAbp is a prokaryotic homolog of an Argonaute protein. Prokaryotic homologs of Argonaute proteins are known and have been described, for example, in Makarova K., et al., “Prokaryotic homologs of Argonaute proteins are predicted to function as key components of a novel system of defense against mobile genetic elements”, Biol Direct. 2009 Aug. 25; 4:29. doi: 10.1186/1745-6150-4-29, the entire contents of which is hereby incorporated by reference. In some embodiments, the napDNAbp is a Marinitoga piezophila Argunaute (MpAgo) protein. The CRISPR-associated Marinitoga piezophila Argunaute (MpAgo) protein cleaves single-stranded target sequences using 5′-phosphorylated guides. The 5′ guides are used by all known Argonautes. The crystal structure of an MpAgo-RNA complex shows a guide strand binding site comprising residues that block 5′ phosphate interactions. This data suggests the evolution of an Argonaute subclass with noncanonical specificity for a 5′-hydroxylated guide. See, e.g., Kaya et al., “A bacterial Argonaute with noncanonical guide RNA specificity”, Proc Natl Acad Sci USA. 2016 Apr. 12; 113(15):4057-62, the entire contents of which are hereby incorporated by reference). It should be appreciated that other argonaute proteins may be used, and are within the scope of this disclosure.


Some aspects of the disclosure provide Cas9 domains that have different PAM specificities. Typically, Cas9 proteins, such as Cas9 from S. pyogenes (spCas9), require a canonical NGG PAM sequence to bind a particular nucleic acid region. This may limit the ability to edit desired bases within a genome. In some embodiments, the base editing fusion proteins provided herein may need to be placed at a precise location, for example where a target base is placed within a 4 base region (e.g., a “editing window”), which is approximately 15 bases upstream of the PAM. See Komor, A. C., et al., “Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage” Nature 533, 420-424 (2016), the entire contents of which are hereby incorporated by reference. Accordingly, in some embodiments, any of the fusion proteins provided herein may contain a Cas9 domain that is capable of binding a nucleotide sequence that does not contain a canonical (e.g., NGG) PAM sequence. Cas9 domains that bind to non-canonical PAM sequences have been described in the art and would be apparent to the skilled artisan. For example, Cas9 domains that bind non-canonical PAM sequences have been described in Kleinstiver, B. P., et al., “Engineered CRISPR-Cas9 nucleases with altered PAM specificities” Nature 523, 481-485 (2015); and Kleinstiver, B. P., et al., “Broadening the targeting range of Staphylococcus aureus CRISPR-Cas9 by modifying PAM recognition” Nature Biotechnology 33, 1293-1298 (2015); the entire contents of each are hereby incorporated by reference.


For example, a napDNAbp domain with altered PAM specificity, such as a domain with at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity with wild type Francisella novicida Cpf1 (D917, E1006, and D1255) (SEQ ID NO: 74), which has the following amino acid sequence:










(SEQ ID NO: 74)



MSIYQEFVNKYSLSKTLRFELIPQGKTLENIKARGLILDDEKRAKDYKKAKQIIDKYHQFFIEEI






LSSVCISEDLLQNYSDVYFKLKKSDDDNLQKDFKSAKDTIKKQISEYIKDSEKFKNLFNQNLID





AKKGQESDLILWLKQSKDNGIELFKANSDITDIDEALEIIKSFKGWTTYFKGFHENRKNVYSSN





DIPTSIIYRIVDDNLPKFLENKAKYESLKDKAPEAINYEQIKKDLAEELTFDIDYKTSEVNQRVFS





LDEVFEIANFNNYLNQSGITKFNTIIGGKFVNGENTKRKGINEYINLYSQQINDKTLKKYKMSV





LFKQILSDTESKSFVIDKLEDDSDVVTTMQSFYEQIAAFKTVEEKSIKETLSLLFDDLKAQKLDL





SKIYFKNDKSLTDLSQQVFDDYSVIGTAVLEYITQQIAPKNLDNPSKKEQELIAKKTEKAKYLSL





ETIKLALEEFNKHRDIDKQCRFEEILANFAAIPMIFDEIAQNKDNLAQISIKYQNQGKKDLLQAS





AEDDVKAIKDLLDQTNNLLHKLKIFHISQSEDKANILDKDEHFYLVFEECYFELANIVPLYNKIR





NYITQKPYSDEKFKLNFENSTLANGWDKNKEPDNTAILFIKDDKYYLGVMNKKNNKIFDDKAI





KENKGEGYKKIVYKLLPGANKMLPKVFFSAKSIKFYNPSEDILRIRNHSTHTKNGSPQKGYEKF





EFNIEDCRKFIDFYKQSISKHPEWKDFGFRFSDTQRYNSIDEFYREVENQGYKLTFENISESYIDS





VVNQGKLYLFQIYNKDFSAYSKGRPNLHTLYWKALFDERNLQDVVYKLNGEAELFYRKQSIP





KKITHPAKEAIANKNKDNPKKESVFEYDLIKDKRFTEDKFFFHCPITINFKSSGANKFNDEINLL





LKEKANDVHILSIDRGERHLAYYTLVDGKGNIIKQDTFNIIGNDRMKTNYHDKLAAIEKDRDS





ARKDWKKINNIKEMKEGYLSQVVHEIAKLVIEYNAIVVFEDLNFGFKRGRFKVEKQVYQKLE





KMLIEKLNYLVFKDNEFDKTGGVLRAYQLTAPFETFKKMGKQTGIIYYVPAGFTSKICPVTGFV





NQLYPKYESVSKSQEFFSKFDKICYNLDKGYFEFSFDYKNFGDKAAKGKWTIASFGSRLINFRN





SDKNHNWDTREVYPTKELEKLLKDYSIEYGHGECIKAAICGESDKKFFAKLTSVLNTILQMRN





SKTGTELDYLISPVADVNGNFFDSRQAPKNMPQDADANGAYHIGLKGLMLLGRIKNNQEGKK





LNLVIKNEEYFEFVQNRNN






An additional napDNAbp domain with altered PAM specificity, such as a domain having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity with wild type Geobacillus thermodenitrificans Cas9 (SEQ ID NO: 75), which has the following amino acid sequence:










(SEQ ID NO: 75)



MKYKIGLDIGITSIGWAVINLDIPRIEDLGVRIFDRAENPKTGESLALPRRLARSARRRLRRRKHR






LERIRRLFVREGILTKEELNKLFEKKHEIDVWQLRVEALDRKLNNDELARILLHLAKRRGFRSN





RKSERTNKENSTMLKHIEENQSILSSYRTVAEMVVKDPKFSLHKRNKEDNYTNTVARDDLEREI





KLIFAKQREYGNIVCTEAFEHEYISIWASQRPFASKDDIEKKVGFCTFEPKEKRAPKATYTFQSF





TVWEHINKLRLVSPGGIRALTDDERRLIYKQAFHKNKITFHDVRTLLNLPDDTRFKGLLYDRNT





TLKENEKVRFLELGAYHKIRKAIDSVYGKGAAKSFRPIDFDTFGYALTMFKDDTDIRSYLRNEY





EQNGKRMENLADKVYDEELIEELLNLSFSKFGHLSLKALRNILPYMEQGEVYSTACERAGYTF





TGPKKKQKTVLLPNIPPIANPVVMRALTQARKVVNAIIKKYGSPVSIHIELARELSQSFDERRK





MQKEQEGNRKKNETAIRQLVEYGLTLNPTGLDIVKFKLWSEQNGKCAYSLQPIEIERLLEPGYT





EVDHVIPYSRSLDDSYTNKVLVLTKENREKGNRTPAEYLGLGSERWQQFETFVLTNKQFSKKK





RDRLLRLHYDENEENEFKNRNLNDTRYISRFLANFIREHLKFADSDDKQKVYTVNGRITAHLRS





RWNFNKNREESNLHHAVDAAIVACTTPSDIARVTAFYQRREQNKELSKKTDPQFPQPWPHFAD





ELQARLSKNPKESIKALNLGNYDNEKLESLQPVFVSRMPKRSITGAAHQETLRRYIGIDERSGKI





QTVVKKKLSEIQLDKTGHFPMYGKESDPRTYEAIRQRLLEHNNDPKKAFQEPLYKPKKNGELG





PIIRTIKIIDTTNQVIPLNDGKTVAYNSNIVRVDVFEKDGKYYCVPIYTIDMMKGILPNKAIEPNK





PYSEWKEMTEDYTFRFSLYPNDLIRIEFPREKTIKTAVGEEIKIKDLFAYYQTIDSSNGGLSLVSH





DNNFSLRSIGSRTLKRFEKYQVDVLGNIYKVRGEKRVGVASSSHSKAGETIRPL






In some embodiments, the nucleic acid programmable DNA binding protein (napDNAbp) is a nucleic acid programmable DNA binding protein that does not require a canonical (NGG) PAM sequence. In some embodiments, the napDNAbp is an argonaute protein. One example of such a nucleic acid programmable DNA binding protein is an Argonaute protein from Natronobacterium gregoryi (NgAgo). NgAgo is a ssDNA-guided endonuclease. NgAgo binds 5′ phosphorylated ssDNA of ˜24 nucleotides (gDNA) to guide it to its target site and will make DNA double-strand breaks at the gDNA site. In contrast to Cas9, the NgAgo-gDNA system does not require a protospacer-adjacent motif (PAM). Using a nuclease inactive NgAgo (dNgAgo) can greatly expand the bases that may be targeted. The characterization and use of NgAgo have been described in Gao et al., Nat Biotechnol., 34(7): 768-73 (2016), PubMed PMID: 27136078; Swarts et al., Nature, 507(7491): 258-61 (2014); and Swarts et al., Nucleic Acids Res. 43(10) (2015): 5120-9, each of which is incorporated herein by reference. The sequence of Natronobacterium gregoryi Argonaute is provided in SEQ ID NO: 76.


The disclosed fusion proteins may comprise a napDNAbp domain having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity with wild type Natronobacterium gregoryi Argonaute (SEQ ID NO: 76), which has the following amino acid sequence:










(SEQ ID NO: 76)



MTVIDLDSTTTADELTSGHTYDISVTLTGVYDNTDEQHPRMSLAFEQDNGERRYITLWKNTTP






KDVFTYDYATGSTYIFTNIDYEVKDGYENLTATYQTTVENATAQEVGTTDEDETFAGGEPLDHH





LDDALNETPDDAETESDSGHVMTSFASRDQLPEWTLHTYTLTATDGAKTDTEYARRTLAYTVR





QELYTDHDAAPVATDGLMLLTPEPLGETPLDLDCGVRVEADETRTLDYTTAKDRLLARELVEE





GLKRSLWDDYLVRGIDEVLSKEPVLTCDEFDLHERYDLSVEVGHSGRAYLHINFRHRFVPKLTL





ADIDDDNIYPGLRVKTTYRPRRGHIVWGLRDECATDSLNTLGNQSVVAYHRNNQTPINTDLLD





AIEAADRRVVETRRQGHGDDAVSFPQELLAVEPNTHQIKQFASDGFHQQARSKTRLSASRCSE





KAQAFAERLDPVRLNGSTVEFSSEFFTGNNEQQLRLLYENGESVLTFRDGARGAHPDETFSKGI





VNPPESFEVAVVLPEQQADTCKAQWDTMADLLNQAGAPPTRSETVQYDAFSSPESISLNVAGA





IDPSEVDAAFVVLPPDQEGFADLASPTETYDELKKALANMGIYSQMAYFDRFRDAKIFYTRNV





ALGLLAAAGGVAFTTEHAMPGDADMFIGIDVSRSYPEDGASGQINIAATATAVYKDGTILGHSS





TRPQLGEKLQSTDVRDIMKNAILGYQQVTGESPTHIVIHRDGFMNEDLDPATEFLNEQGVEYDI





VEIRKQPQTRLLAVSDVQYDTPVKSIAAINQNEPRATVATFGAPEYLATRDGGGLPRPIQIERVA





GETDIETLTRQVYLLSQSHIQVHNSTARLPITTAYADQASTHATKGYLVQTGAFESNVGFL






In addition, any available methods may be utilized to obtain or construct a variant or mutant Cas9 protein. The term “mutation,” as used herein, refers to a substitution of a residue within a sequence, e.g., a nucleic acid or amino acid sequence, with another residue, or a deletion or insertion of one or more residues within a sequence. Mutations are typically described herein by identifying the original residue followed by the position of the residue within the sequence and by the identity of the newly substituted residue. Various methods for making the amino acid substitutions (mutations) provided herein are well known in the art, and are provided by, for example, Green and Sambrook, Molecular Cloning: A Laboratory Manual (4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2012)). Mutations can include a variety of categories, such as single base polymorphisms, microduplication regions, indel, and inversions, and is not meant to be limiting in any way. Mutations can include “loss-of-function” mutations which is the normal result of a mutation that reduces or abolishes a protein activity. Most loss-of-function mutations are recessive, because in a heterozygote the second chromosome copy carries an unmutated version of the gene coding for a fully functional protein whose presence compensates for the effect of the mutation. Mutations also embrace “gain-of-function” mutations, which is one which confers an abnormal activity on a protein or cell that is otherwise not present in a normal condition. Many gain-of-function mutations are in regulatory sequences rather than in coding regions, and can therefore have a number of consequences. For example, a mutation might lead to one or more genes being expressed in the wrong tissues, these tissues gaining functions that they normally lack. Because of their nature, gain-of-function mutations are usually dominant.


Mutations can be introduced into a reference Cas9 protein using site-directed mutagenesis. Older methods of site-directed mutagenesis known in the art rely on sub-cloning of the sequence to be mutated into a vector, such as an M13 bacteriophage vector, that allows the isolation of single-stranded DNA template. In these methods, one anneals a mutagenic primer (i.e., a primer capable of annealing to the site to be mutated but bearing one or more mismatched nucleotides at the site to be mutated) to the single-stranded template and then polymerizes the complement of the template starting from the 3′ end of the mutagenic primer. The resulting duplexes are then transformed into host bacteria and plaques are screened for the desired mutation. More recently, site-directed mutagenesis has employed PCR methodologies, which have the advantage of not requiring a single-stranded template. In addition, methods have been developed that do not require sub-cloning. Several issues must be considered when PCR-based site-directed mutagenesis is performed. First, in these methods it is desirable to reduce the number of PCR cycles to prevent expansion of undesired mutations introduced by the polymerase. Second, a selection must be employed in order to reduce the number of non-mutated parental molecules persisting in the reaction. Third, an extended-length PCR method is preferred in order to allow the use of a single PCR primer set. And fourth, because of the non-template-dependent terminal extension activity of some thermostable polymerases it is often necessary to incorporate an end-polishing step into the procedure prior to blunt-end ligation of the PCR-generated mutant product.


Mutations may also be introduced by directed evolution processes, such as phage-assisted continuous evolution (PACE) or phage-assisted noncontinuous evolution (PANCE). The term “phage-assisted continuous evolution (PACE),” as used herein, refers to continuous evolution that employs phage as viral vectors. The general concept of PACE technology has been described, for example, in International PCT Application, PCT/US2009/056194, filed Sep. 8, 2009, published as WO 2010/028347 on Mar. 11, 2010; International PCT Application, PCT/US2011/066747, filed Dec. 22, 2011, published as WO 2012/088381 on Jun. 28, 2012; U.S. Application, U.S. Pat. No. 9,023,594, issued May 5, 2015, International PCT Application, PCT/US2015/012022, filed Jan. 20, 2015, published as WO 2015/134121 on Sep. 11, 2015, and International PCT Application, PCT/US2016/027795, filed Apr. 15, 2016, published as WO 2016/168631 on Oct. 20, 2016, the entire contents of each of which are incorporated herein by reference. Variant Cas9s may also be obtain by phage-assisted non-continuous evolution (PANCE),” which as used herein, refers to non-continuous evolution that employs phage as viral vectors. PANCE is a simplified technique for rapid in vivo directed evolution using serial flask transfers of evolving ‘selection phage’ (SP), which contain a gene of interest to be evolved, across fresh E. coli host cells, thereby allowing genes inside the host E. coli to be held constant while genes contained in the SP continuously evolve. Serial flask transfers have long served as a widely-accessible approach for laboratory evolution of microbes, and, more recently, analogous approaches have been developed for bacteriophage evolution. The PANCE system features lower stringency than the PACE system.


Any of the references noted above which relate to Cas9 or Cas9 equivalents are hereby incorporated by reference in their entireties, if not already stated so.


J. Divided napDNAbp Domains for Split PE Delivery


In various embodiments, the prime editors described herein may be delivered to cells as two or more fragments which become assembled inside the cell (either by passive assembly, or by active assembly, such as using split intein sequences) into a reconstituted prime editor. In some cases, the self assembly may be passive whereby the two or more prime editor fragments associate inside the cell covalently or non-covalently to reconstitute the prime editor. In other cases, the self-assembly may be catalzyed by dimerization domains installed on each of the fragments. Examples of dimerization domains are described herein. In still other cases, the self-assembly may be catalyzed by split intein sequences installed on each of the prime editor fragments.


Split PE delivery may be advantageous to address various size constraints of different delivery approaches. For example, delivery approaches may include virus-based delivery methods, messenger RNA-based delivery methods, or RNP-based delivery (ribonucleoprotein-based delivery). And, each of these methods of delivery may be more efficient and/or effective by dividing up the prime editor into smaller pieces. Once inside the cell, the smaller pieces can assemble into a functional prime editor. Depending on the means of splitting, the divided prime editor fragments can be reassembled in a non-covalent manner or a covalent manner to reform the prime editor. In one embodiment, the prime editor can be split at one or more split sites into two or more fragments. The fragments can be unmodified (other than being split). Once the fragments are delivered to the cell (e.g., by direct delivery of a ribonucleoprotein complex or by nucleic delivery—e.g., mRNA delivery or virus vector based delivery), the fragments can reassociate covalently or non-covalently to reconstitute the prime editor. In another embodiment, the prime editor can be split at one or more split sites into two or more fragments. Each of the fragments can be modified to comprise a dimerization domain, whereby each fragment that is formed is coupled to a dimerization domain Once delivered or expressed within a cell, the dimerization domains of the different fragments associate and bind to one another, bringing the different prime editor fragments together to reform a functional prime editor. In yet another embodiment, the prime editor fragment may be modified to comprise a split intein. Once delivered or expressed within a cell, the split intein domains of the different fragments associate and bind to one another, and then undergo trans-splicing, which results in the excision of the split-intein domains from each of the fragments, and a concomitant formation of a peptide bond between the fragments, thereby restoring the prime editor.


In one embodiment, the prime editor can be delivered using a split-intein approach.


The location of the split site can be positioned between any one or more pair of residues in the prime editor and in any domains therein, including within the napDNAbp domain, the polymerase domain (e.g., RT domain), linker domain that joins the napDNAbp domain and the polymerase domain


In one embodiment, depicted in FIG. 66, the prime editor (PE) is divided at a split site within the napDNAbp.


In certain embodiments, the napDNAbp is a canonical SpCas9 polypeptide of SEQ ID NO: 18, as follows:
















SpCas9
MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDR
SEQ ID NO: 18



Streptococcus

HSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYL




pyogenes

QEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVD



M1
EVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFR



SwissProt
GHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVD



Accession
AKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTP



No. Q99ZW2
NFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLF



Wild type
LAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDL



1368 AA
TLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFY




KFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHL




GELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGN




SRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDK




NLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLS




GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGV




EDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLF




EDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKL




INGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQ




KAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKV




MGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKEL




GSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDIN




RLSDYDVDHIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPS




EEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELD




KAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREV




KVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVV




GTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATA




KYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKG




RDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDK




LIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLK




SVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHY




EKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADA




NLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYF




DTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGD









In certain embodiments, the SpCas9 is split into two fragments at a split site located between residues 1 and 2, or 2 and 3, or 3 and 4, or 4 and 5, or 5 and 6, or 6 and 7, or 7 and 8, or 8 and 9, or 9 and 10, or between any two pair of residues located anywhere between residues 1-10, 10-20, 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-200, 200-300, 300-400, 400-500, 500-600, 600-700, 700-800, 800-900, 1000-1100, 1100-1200, 1200-1300, or 1300-1368 of canonical SpCas9 of SEQ ID NO: 18.


In certain embodiments, a napDNAbp is split into two fragments at a split site that is located at a pair of residue that corresponds to any two pair of residues located anywhere between positions 1-10, 10-20, 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-200, 200-300, 300-400, 400-500, 500-600, 600-700, 700-800, 800-900, 1000-1100, 1100-1200, 1200-1300, or 1300-1368 of canonical SpCas9 of SEQ ID NO: 18.


In certain embodiments, the SpCas9 is split into two fragments at a split site located between residues 1 and 2, or 2 and 3, or 3 and 4, or 4 and 5, or 5 and 6, or 6 and 7, or 7 and 8, or 8 and 9, or 9 and 10, or between any two pair of residues located anywhere between residues 1-10, 10-20, 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-200, 200-300, 300-400, 400-500, 500-600, 600-700, 700-800, 800-900, 1000-1100, 1100-1200, 1200-1300, or 1300-1368 of canonical SpCas9 of SEQ ID NO: 18. In certain embodiments, the split site is located one or more polypeptide bond sites (i.e., a “split site or split-intein split site”), fused to a split intein, and then delivered to cells as separately-encoded fusion proteins. Once the split-intein fusion proteins (i.e., protein halves) are expressed within a cell, the proteins undergo trans-splicing to form a complete or whole PE with the concomitant removal of the joined split-intein sequences.


For example, as shown in FIG. 66, the N-terminal extein can be fused to a first split-intein (e.g., N intein) and the C-terminal extein can be fused to a second split-intein (e.g., C intein). The N-terminal extein becomes fused to the C-terminal extein to reform a whole prime editor fusion protein comprising an napDNAbp domain and a polymerase domain (e.g., RT domain) upon the self-association of the N intein and the C intein inside the cell, followed by their self-excision, and the concomitant formation of a peptide bond between the N-terminal extein and C-terminal extein portions of a whole prime editor (PE).


To take advantage of a split-PE delivery strategy using split-inteins, the prime editor needs to be divided at one or more split sites to create at least two separate halves of a prime editor, each of which may be rejoined inside a cell if each half is fused to a split-intein sequence.


In certain embodiments, the prime editor is split at a single split site. In certain other embodiments, the prime editor is split at two split sites, or three split sites, or four split sites, or more.


In a preferred embodiment, the prime editor is split at a single split site to create two separate halves of a prime editor, each of which can be fused to a split intein sequence


An exemplary split intein is the Ssp DnaE intein, which comprises two subunits, namely, DnaE-N and DnaE-C. The two different subunits are encoded by separate genes, namely dnaE-n and dnaE-c, which encode the DnaE-N and DnaE-C subunits, respectively. DnaE is a naturally occurring split intein in Synechocytis sp. PCC6803 and is capable of directing trans-splicing of two separate proteins, each comprising a fusion with either DnaE-N or DnaE-C.


Additional naturally occurring or engineered split-intein sequences are known in the or can be made from whole-intein sequences described herein or those available in the art. Examples of split-intein sequences can be found in Stevens et al., “A promiscuous split intein with expanded protein engineering applications,” PNAS, 2017, Vol. 114: 8538-8543; Iwai et al., “Highly efficient protein trans-splicing by a naturally split DnaE intein from Nostc punctiforme, FEBS Lett, 580: 1853-1858, each of which are incorporated herein by reference. Additional split intein sequences can be found, for example, in WO 2013/045632, WO 2014/055782, WO 2016/069774, and EP2877490, the contents each of which are incorporated herein by reference.


In addition, protein splicing in trans has been described in vivo and in vitro (Shingledecker, et al., Gene 207:187 (1998), Southworth, et al., EMBO J. 17:918 (1998); Mills, et al., Proc. Natl. Acad. Sci. USA, 95:3543-3548 (1998); Lew, et al., J. Biol. Chem., 273:15887-15890 (1998); Wu, et al., Biochim. Biophys. Acta 35732:1 (1998b), Yamazaki, et al., J. Am. Chem. Soc. 120:5591 (1998), Evans, et al., J. Biol. Chem. 275:9091 (2000); Otomo, et al., Biochemistry 38:16040-16044 (1999); Otomo, et al., J. Biolmol. NMR 14:105-114 (1999); Scott, et al., Proc. Natl. Acad. Sci. USA 96:13638-13643 (1999)) and provides the opportunity to express a protein as to two inactive fragments that subsequently undergo ligation to form a functional product, e.g., as shown in FIGS. 66 and 67 with regard to the formation of a complete PE fusion protein from two separately-expressed halves.


In various embodiments described herein, the continuous evolution methods (e.g., PACE) may be used to evolve a first portion of a base editor. A first portion could include a single component or domain, e.g., a Cas9 domain, a deaminase domain, or a UGI domain. The separately evolved component or domain can be then fused to the remaining portions of the base editor within a cell by separately express both the evolved portion and the remaining non-evolved portions with split-intein polypeptide domains. The first portion could more broadly include any first amino acid portion of a base editor that is desired to be evolved using a continuous evolution method described herein. The second portion would in this embodiment refer to the remaining amino acid portion of the base editor that is not evolved using the herein methods. The evolved first portion and the second portion of the base editor could each be expressed with split-intein polypeptide domains in a cell. The natural protein splicing mechanisms of the cell would reassemble the evolved first portion and the non-evolved second portion to form a single fusion protein evolved base editor. The evolved first portion may comprise either the N- or C-terminal part of the single fusion protein. In an analogous manner, use of a second orthogonal trans-splicing intein pair could allow the evolved first portion to comprise an internal part of the single fusion protein.


Thus, any of the evolved and non-evolved components of the base editors herein described may be expressed with split-intein tags in order to facilitate the formation of a complete base editor comprising the evolved and non-evolved component within a cell.


The mechanism of the protein splicing process has been studied in great detail (Chong, et al., J. Biol. Chem. 1996, 271, 22159-22168; Xu, M-Q & Perler, F. B. EMBO Journal, 1996, 15, 5146-5153) and conserved amino acids have been found at the intein and extein splicing points (Xu, et al., EMBO Journal, 1994, 13 5517-522). The constructs described herein contain an intein sequence fused to the 5′-terminus of the first gene (e.g., the evolved portion of the base editor). Suitable intein sequences can be selected from any of the proteins known to contain protein splicing elements. A database containing all known inteins can be found on the World Wide Web (Perler, F. B. Nucleic Acids Research, 1999, 27, 346-347). The intein sequence is fused at the 3′ end to the 5′ end of a second gene. For targeting of this gene to a certain organelle, a peptide signal can be fused to the coding sequence of the gene. After the second gene, the intein-gene sequence can be repeated as often as desired for expression of multiple proteins in the same cell. For multi-intein containing constructs, it may be useful to use intein elements from different sources. After the sequence of the last gene to be expressed, a transcription termination sequence must be inserted. In one embodiment, a modified intein splicing unit is designed so that it can both catalyze excision of the exteins from the inteins as well as prevent ligation of the exteins. Mutagenesis of the C-terminal extein junction in the Pyrococcus species GB-D DNA polymerase was found to produce an altered splicing element that induces cleavage of exteins and inteins but prevents subsequent ligation of the exteins (Xu, M-Q & Perler, F. B. EMBO Journal, 1996, 15, 5146-5153). Mutation of serine 538 to either an alanine or glycine induced cleavage but prevented ligation. Mutation of equivalent residues in other intein splicing units should also prevent extein ligation due to the conservation of amino acids at the C-terminal extein junction to the intein. A preferred intein not containing an endonuclease domain is the Mycobacterium xenopi GyrA protein (Telenti, et al. J. Bacteriol. 1997, 179, 6378-6382). Others have been found in nature or have been created artificially by removing the endonuclease domains from endonuclease containing inteins (Chong, et al. J. Biol. Chem. 1997, 272, 15587-15590). In a preferred embodiment, the intein is selected so that it consists of the minimal number of amino acids needed to perform the splicing function, such as the intein from the Mycobacterium xenopi GyrA protein (Telenti, A., et al., J. Bacteriol. 1997, 179, 6378-6382). In an alternative embodiment, an intein without endonuclease activity is selected, such as the intein from the Mycobacterium xenopi GyrA protein or the Saccharaomyces cerevisiae VMA intein that has been modified to remove endonuclease domains (Chong, 1997). Further modification of the intein splicing unit may allow the reaction rate of the cleavage reaction to be altered allowing protein dosage to be controlled by simply modifying the gene sequence of the splicing unit.


Inteins can also exist as two fragments encoded by two separately transcribed and translated genes. These so-called split inteins self-associate and catalyze protein-splicing activity in trans. Split inteins have been identified in diverse cyanobacteria and archaea (Caspi et al, Mol Microbiol. 50: 1569-1577 (2003); Choi J. et al, J Mol Biol. 556: 1093-1106 (2006.); Dassa B. et al, Biochemistry. 46:322-330 (2007.); Liu X. and Yang J., J Biol Chem. 275:26315-26318 (2003); Wu H. et al.


Proc Natl Acad Sci USA. £5:9226-9231 (1998.); and Zettler J. et al, FEBS Letters. 553:909-914 (2009)), but have not been found in eukaryotes thus far. Recently, a bioinformatic analysis of environmental metagenomic data revealed 26 different loci with a novel genomic arrangement. At each locus, a conserved enzyme coding region is interrupted by a split intein, with a freestanding endonuclease gene inserted between the sections coding for intein subdomains Among them, five loci were completely assembled: DNA helicases (gp41-1, gp41-8); Inosine-5′-monophosphate dehydrogenase (IMPDH-1); and Ribonucleotide reductase catalytic subunits (NrdA-2 and NrdJ-1). This fractured gene organization appears to be present mainly in phages (Dassa et al, Nucleic Acids Research. 57:2560-2573 (2009)).


The split intein Npu DnaE was characterized as having the highest rate reported for the protein trans-splicing reaction. In addition, the Npu DnaE protein splicing reaction is considered robust and high-yielding with respect to different extein sequences, temperatures from 6 to 37° C., and the presence of up to 6M Urea (Zettler J. et al, FEBS Letters. 553:909-914 (2009); Iwai I. et al, FEBS Letters 550: 1853-1858 (2006)). As expected, when the Cysl Ala mutation at the N-domain of these inteins was introduced, the initial N to S-acyl shift and therefore protein splicing was blocked. Unfortunately, the C-terminal cleavage reaction was also almost completely inhibited. The dependence of the asparagine cyclization at the C-terminal splice junction on the acyl shift at the N-terminal scissile peptide bond seems to be a unique property common to the naturally split DnaE intein alleles (Zettler J. et al. FEBS Letters. 555:909-914 (2009)).


The mechanism of protein splicing typically has four steps [29-30]: 1) an N—S or N—O acyl shift at the intein N-terminus, which breaks the upstream peptide bond and forms an ester bond between the N-extein and the side chain of the intein's first amino acid (Cys or Ser); 2) a transesterification relocating the N-extein to the intein C-terminus, forming a new ester bond linking the N-extein to the side chain of the C-extein's first amino acid (Cys, Ser, or Thr); 3) Asn cyclization breaking the peptide bond between the intein and the C-extein; and 4) a S—N or O—N acyl shift that replaces the ester bond with a peptide bond between the N-extein and C-extein.


Protein trans-splicing, catalyzed by split inteins, provides an entirely enzymatic method for protein ligation [31]. A split-intein is essentially a contiguous intein (e g a mini-intein) split into two pieces named N-intein and C-intein, respectively. The N-intein and C-intein of a split intein can associate non-covalently to form an active intein and catalyze the splicing reaction essentially in same way as a contiguous intein does. Split inteins have been found in nature and also engineered in laboratories [31-35]. As used herein, the term “split intein” refers to any intein in which one or more peptide bond breaks exists between the N-terminal and C-terminal amino acid sequences such that the N-terminal and C-terminal sequences become separate molecules that can non-covalently reassociate, or reconstitute, into an intein that is functional for trans-splicing reactions. Any catalytically active intein, or fragment thereof, may be used to derive a split intein for use in the methods of the invention. For example, in one aspect the split intein may be derived from a eukaryotic intein. In another aspect, the split intein may be derived from a bacterial intein. In another aspect, the split intein may be derived from an archaeal intein. Preferably, the split intein so-derived will possess only the amino acid sequences essential for catalyzing trans-splicing reactions.


As used herein, the “N-terminal split intein (In)” refers to any intein sequence that comprises an N-terminal amino acid sequence that is functional for trans-splicing reactions. An In thus also comprises a sequence that is spliced out when trans-splicing occurs. An In can comprise a sequence that is a modification of the N-terminal portion of a naturally occurring intein sequence. For example, an In can comprise additional amino acid residues and/or mutated residues so long as the inclusion of such additional and/or mutated residues does not render the In non-functional in trans-splicing. Preferably, the inclusion of the additional and/or mutated residues improves or enhances the trans-splicing activity of the In.


As used herein, the “C-terminal split intein (Ic)” refers to any intein sequence that comprises a C-terminal amino acid sequence that is functional for trans-splicing reactions. In one aspect, the Ic comprises 4 to 7 contiguous amino acid residues, at least 4 amino acids of which are from the last β-strand of the intein from which it was derived. An Ic thus also comprises a sequence that is spliced out when trans-splicing occurs. An Ic can comprise a sequence that is a modification of the C-terminal portion of a naturally occurring intein sequence. For example, an Ic can comprise additional amino acid residues and/or mutated residues so long as the inclusion of such additional and/or mutated residues does not render the In non-functional in trans-splicing. Preferably, the inclusion of the additional and/or mutated residues improves or enhances the trans-splicing activity of the Ic.


In some embodiments of the invention, a peptide linked to an Ic or an In can comprise an additional chemical moiety including, among others, fluorescence groups, biotin, polyethylene glycol (PEG), amino acid analogs, unnatural amino acids, phosphate groups, glycosyl groups, radioisotope labels, and pharmaceutical molecules. In other embodiments, a peptide linked to an Ic can comprise one or more chemically reactive groups including, among others, ketone, aldehyde, Cys residues and Lys residues. The N-intein and C-intein of a split intein can associate non-covalently to form an active intein and catalyze the splicing reaction when an “intein-splicing polypeptide (ISP)” is present. As used herein, “intein-splicing polypeptide (ISP)” refers to the portion of the amino acid sequence of a split intein that remains when the Ic, In, or both, are removed from the split intein. In certain embodiments, the In comprises the ISP. In another embodiment, the Ic comprises the ISP. In yet another embodiment, the ISP is a separate peptide that is not covalently linked to In nor to Ic.


Split inteins may be created from contiguous inteins by engineering one or more split sites in the unstructured loop or intervening amino acid sequence between the −12 conserved beta-strands found in the structure of mini-inteins [25-28]. Some flexibility in the position of the split site within regions between the beta-strands may exist, provided that creation of the split will not disrupt the structure of the intein, the structured beta-strands in particular, to a sufficient degree that protein splicing activity is lost.


In protein trans-splicing, one precursor protein consists of an N-extein part followed by the N-intein, another precursor protein consists of the C-intein followed by a C-extein part, and a trans-splicing reaction (catalyzed by the N- and C-inteins together) excises the two intein sequences and links the two extein sequences with a peptide bond. Protein trans-splicing, being an enzymatic reaction, can work with very low (e.g. micromolar) concentrations of proteins and can be carried out under physiological conditions.


[2] Other Programmable Nucleases

In various embodiments described herein, the multi-flap prime editors comprise a napDNAbp, such as a Cas9 protein. These proteins are “programmable” by way of their becoming complexed with a guide RNA (or a PEgRNA, as the case may be), which guides the Cas9 protein to a target site on the DNA which possess a sequence that is complementary to the spacer portion of the gRNA (or PEgRNA) and also which possesses the required PAM sequence. However, in certain embodiment envisioned here, the napDNAbp may be substituted with a different type of programmable protein, such as a zinc finger nuclease or a transcription activator-like effector nuclease (TALEN).



FIG. 1J depicts such a variation of prime editing contemplated herein that replaces the napDNAbp (e.g., SpCas9 nickase) with any programmable nuclease domain, such as zinc finger nucleases (ZFN) or transcription activator-like effector nucleases (TALEN). As such, it is contemplated that suitable nucleases do not necessarily need to be “programmed” by a nucleic acid targeting molecule (such as a guide RNA), but rather, may be programmed by defining the specificity of a DNA-binding domain, such as and in particular, a nuclease. Just as in prime editing with napDNAbp moities, it is preferable that such alternative programmable nucleases be modified such that only one strand of a target DNA is cut. In other words, the programmable nucleases should function as nickases, preferably. Once a programmable nuclease is selected (e.g., a ZFN or a TALEN), then additional functionalities may be engineered into the system to allow it to operate in accordance with a prime editing-like mechanism. For example, the programmable nucleases may be modified by coupling (e.g., via a chemical linker) an RNA or DNA extension arm thereto, wherein the extension arm comprises a primer binding site (PBS) and a DNA synthesis template. The programmable nuclease may also be coupled (e.g., via a chemical or amino acid linker) to a polymerase, the nature of which will depend upon whether the extension arm is DNA or RNA. In the case of an RNA extension arm, the polymerase can be an RNA-dependent DNA polymerase (e.g., reverse transcriptase). In the case of a DNA extension arm, the polymerase can be a DNA-dependent DNA polymerase (e.g., a prokaryotic polymerase, including Pol I, Pol II, or Pol III, or a eukaryotic polymerase, including Pol a, Pol b, Pol g, Pol d, Pol e, or Pol z). The system may also include other functionalities added as fusions to the programmable nucleases, or added in trans to facilitate the reaction as a whole (e.g., (a) a helicase to unwind the DNA at the cut site to make the cut strand with the 3′ end available as a primer, (b) a FEN1 to help remove the endogenous strand on the cut strand to drive the reaction towards replacement of the endogenous strand with the synthesized strand, or (c) a nCas9:gRNA complex to create a second site nick on the opposite strand, which may help drive the integration of the synthesize repair through favored cellular repair of the non-edited strand). In an analogous manner to prime editing with a napDNAbp, such a complex with an otherwise programmable nuclease could be used to synthesize and then install a newly synthesized replacement strand of DNA carrying an edit of interest permanently into a target site of DNA.


Suitable alternative programmable nucleases are well known in the art which may be used in place of a napDNAbp:gRNA complex to construct an alternative prime editor system that can be programmed to selectively bind a target site of DNA, and which can be further modified in the manner described above to co-localize a polymerase and an RNA or DNA extension arm comprising a primer binding site and a DNA synthesis template to specific nick site. For example, and as represented in FIG. 1J, Transcription Activator-Like Effector Nucleases (TALENs) may be used as the programmable nuclease in the multi-flap prime editing methods and compositions of matter described herein. TALENS are artificial restriction enzymes generated by fusing the TAL effector DNA binding domain to a DNA cleavage domain. These reagents enable efficient, programmable, and specific DNA cleavage and represent powerful tools for genome editing in situ. Transcription activator-like effectors (TALEs) can be quickly engineered to bind practically any DNA sequence. The term TALEN, as used herein, is broad and includes a monomeric TALEN that can cleave double stranded DNA without assistance from another TALEN. The term TALEN is also used to refer to one or both members of a pair of TALENs that are engineered to work together to cleave DNA at the same site. TALENs that work together may be referred to as a left-TALEN and a right-TALEN, which references the handedness of DNA. See U.S. Ser. No. 12/965,590; U.S. Ser. No. 13/426,991 (U.S. Pat. No. 8,450,471); U.S. Ser. No. 13/427,040 (U.S. Pat. No. 8,440,431); U.S. Ser. No. 13/427,137 (U.S. Pat. No. 8,440,432); and U.S. Ser. No. 13/738,381, all of which are incorporated by reference herein in their entirety. In addition, TALENS are described in WO 2015/027134, U.S. Pat. No. 9,181,535, Boch et al., “Breaking the Code of DNA Binding Specificity of TAL-Type III Effectors”, Science, vol. 326, pp. 1509-1512 (2009), Bogdanove et al., TAL Effectors: Customizable Proteins for DNA Targeting, Science, vol. 333, pp. 1843-1846 (2011), Cade et al., “Highly efficient generation of heritable zebrafish gene mutations using homo- and heterodimeric TALENs”, Nucleic Acids Research, vol. 40, pp. 8001-8010 (2012), and Cermak et al., “Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting”, Nucleic Acids Research, vol. 39, No. 17, e82 (2011), each of which are incorporated herein by reference.


As represented in FIG. 1J, zinc finger nucleases may also be used as alternative programmable nucleases for use in multi-flap prime editing in place of napDNAbps, such as Cas9 nickases. Like with TALENS, the ZFN proteins may be modified such that they function as nickases, i.e., engineering the ZFN such that it cleaves only one strand of the target DNA in a manner similar to the napDNAbp used with the multi-flap prime editors described herein. ZFN proteins have been extensively described in the art, for example, in Carroll et al., “Genome Engineering with Zinc-Finger Nucleases,” Genetics, August 2011, Vol. 188: 773-782; Durai et al., “Zinc finger nucleases: custom-designed molecular scissors for genome engineering of plant and mammalian cells,” Nucleic Acids Res, 2005, Vol. 33: 5978-90; and Gaj et al., “ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering,” Trends Biotechnol. 2013, Vol. 31: 397-405, each of which are incorporated herein by reference in their entireties.


[3] Polymerases (e.g., Reverse Transcriptases)

In various embodiments, the multi-flap prime editor system disclosed herein includes a polymerase (e.g., DNA-dependent DNA polymerase or RNA-dependent DNA polymerase, such as, reverse transcriptase), or a variant thereof, which can be provided as a fusion protein with a napDNAbp or other programmable nuclease, or provide in trans.


Any polymerase may be used in the multi-flap prime editors disclosed herein. The polymerases may be wild type polymerases, functional fragments, mutants, variants, or truncated variants, and the like. The polymerases may include wild type polymerases from eukaryotic, prokaryotic, archael, or viral organisms, and/or the polymerases may be modified by genetic engineering, mutagenesis, directed evolution-based processes. The polymerases may include T7 DNA polymerase, T5 DNA polymerase, T4 DNA polymerase, Klenow fragment DNA polymerase, DNA polymerase III and the like. The polymerases may also be thermostable, and may include Taq, Tne, Tma, Pfu, Tfl, Tth, Stoffel fragment, VENT® and DEEPVENT® DNA polymerases, KOD, Tgo, JDF3, and mutants, variants and derivatives thereof (see U.S. Pat. Nos. 5,436,149; 4,889,818; 4,965,185; 5,079,352; 5,614,365; 5,374,553; 5,270,179; 5,047,342; 5,512,462; WO 92/06188; WO 92/06200; WO 96/10640; Barnes, W. M., Gene 112:29-35 (1992); Lawyer, F. C., et al., PCR Meth. Appl. 2:275-287 (1993); Flaman, J.-M, et al., Nuc. Acids Res. 22(15):3259-3260 (1994), each of which are incorporated by reference). For synthesis of longer nucleic acid molecules (e.g, nucleic acid molecules longer than about 3-5 Kb in length), at least two DNA polymerases can be employed. In certain embodiments, one of the polymerases can be substantially lacking a 3′ exonuclease activity and the other may have a 3′ exonuclease activity. Such pairings may include polymerases that are the same or different. Examples of DNA polymerases substantially lacking in 3′ exonuclease activity include, but are not limited to, Taq, Tne(exo-), Tma(exo-), Pfu(exo-), Pwo(exo-), exo-KOD and Tth DNA polymerases, and mutants, variants and derivatives thereof.


Preferably, the polymerase usable in the multi-flap prime editors disclosed herein are “template-dependent” polymerase (since the polymerases are intended to rely on the DNA synthesis template to specify the sequence of the DNA strand under synthesis during prime editing. As used herein, the term “template DNA molecule” refers to that strand of a nucleic acid from which a complementary nucleic acid strand is synthesized by a DNA polymerase, for example, in a primer extension reaction of the DNA synthesis template of a PEgRNA.


As used herein, the term “template dependent manner” is intended to refer to a process that involves the template dependent extension of a primer molecule (e.g., DNA synthesis by DNA polymerase). The term “template dependent manner” refers to polynucleotide synthesis of RNA or DNA wherein the sequence of the newly synthesized strand of polynucleotide is dictated by the well-known rules of complementary base pairing (see, for example, Watson, J. D. et al., In: Molecular Biology of the Gene, 4th Ed., W. A. Benjamin, Inc., Menlo Park, Calif. (1987)). The term “complementary” refers to the broad concept of sequence complementarity between regions of two polynucleotide strands or between two nucleotides through base-pairing. It is known that an adenine nucleotide is capable of forming specific hydrogen bonds (“base pairing”) with a nucleotide which is thymine or uracil. Similarly, it is known that a cytosine nucleotide is capable of base pairing with a guanine nucleotide. As such, in the case of prime editing, it can be said that the single strand of DNA synthesized by the polymerase of the prime editor against the DNA synthesis template is said to be “complementary” to the sequence of the DNA synthesis template.


A. Exemplary Polymerases


In various embodiments, the multi-flap prime editors described herein comprise a polymerase. The disclosure contemplates any wild type polymerase obtained from any naturally-occurring organism or virus, or obtained from a commercial or non-commercial source. In addition, the polymerases usable in the multi-flap prime editors of the disclosure can include any naturally-occurring mutant polymerase, engineered mutant polymerase, or other variant polymerase, including truncated variants that retain function. The polymerases usable herein may also be engineered to contain specific amino acid substitutions, such as those specifically disclosed herein. In certain preferred embodiments, the polymerases usable in the multi-flap prime editors of the disclosure are template-based polymerases, i.e., they synthesize nucleotide sequences in a template-dependent manner.


A polymerase is an enzyme that synthesizes a nucleotide strand and which may be used in connection with the multi-flap prime editor systems described herein. The polymerases are preferrably “template-dependent” polymerases (i.e., a polymerase which synthesizes a nucleotide strand based on the order of nucleotide bases of a template strand). In certain configurations, the polymerases can also be a “template-independent” (i.e., a polymerase which synthesizes a nucleotide strand without the requirement of a template strand). A polymerase may also be further categorized as a “DNA polymerase” or an “RNA polymerase.” In various embodiments, the multi-flap prime editor systems comprise a DNA polymerase. In various embodiments, the DNA polymerase can be a “DNA-dependent DNA polymerase” (i.e., whereby the template molecule is a strand of DNA). In such cases, the DNA template molecule can be a PEgRNA, wherein the extension arm comprises a strand of DNA. In such cases, the PEgRNA may be referred to as a chimeric or hybrid PEgRNA which comprises an RNA portion (i.e., the guide RNA components, including the spacer and the gRNA core) and a DNA portion (i.e., the extension arm). In various other embodiments, the DNA polymerase can be an “RNA-dependent DNA polymerase” (i.e., whereby the template molecule is a strand of RNA). In such cases, the PEgRNA is RNA, i.e., including an RNA extension. The term “polymerase” may also refer to an enzyme that catalyzes the polymerization of nucleotide (i.e., the polymerase activity). Generally, the enzyme will initiate synthesis at the 3′-end of a primer annealed to a polynucleotide template sequence (e.g., such as a primer sequence annealed to the primer binding site of a PEgRNA), and will proceed toward the 5′ end of the template strand. A “DNA polymerase” catalyzes the polymerization of deoxynucleotides. As used herein in reference to a DNA polymerase, the term DNA polymerase includes a “functional fragment thereof”. A “functional fragment thereof” refers to any portion of a wild-type or mutant DNA polymerase that encompasses less than the entire amino acid sequence of the polymerase and which retains the ability, under at least one set of conditions, to catalyze the polymerization of a polynucleotide. Such a functional fragment may exist as a separate entity, or it may be a constituent of a larger polypeptide, such as a fusion protein.


In some embodiments, the polymerases can be from bacteriophage. Bacteriophage DNA polymerases are generally devoid of 5′ to 3′ exonuclease activity, as this activity is encoded by a separate polypeptide. Examples of suitable DNA polymerases are T4, T7, and phi29 DNA polymerase. The enzymes available commercially are: T4 (available from many sources e.g., Epicentre) and T7 (available from many sources, e.g. Epicentre for unmodified and USB for 3′ to 5′ exo T7 “Sequenase” DNA polymerase).


The other embodiments, the polymerases are archaeal polymerases. There are 2 different classes of DNA polymerases which have been identified in archaea: 1. Family B/pol I type (homologs of Pfu from Pyrococcus furiosus) and 2. pol II type (homologs of P. furiosus DP1/DP2 2-subunit polymerase). DNA polymerases from both classes have been shown to naturally lack an associated 5′ to 3′ exonuclease activity and to possess 3′ to 5′ exonuclease (proofreading) activity. Suitable DNA polymerases (pol I or pol II) can be derived from archaea with optimal growth temperatures that are similar to the desired assay temperatures.


Thermostable archaeal DNA polymerases are isolated from Pyrococcus species (furiosus, species GB-D, woesii, abysii, horikoshii), Thermococcus species (kodakaraensis KOD1, litoralis, species 9 degrees North-7, species JDF-3, gorgonarius), Pyrodictium occultum, and Archaeoglobus fulgidus.


Polymerases may also be from eubacterial species. There are 3 classes of eubacterial DNA polymerases, pol I, II, and III. Enzymes in the Pol I DNA polymerase family possess 5′ to 3′ exonuclease activity, and certain members also exhibit 3′ to 5′ exonuclease activity. Pol II DNA polymerases naturally lack 5′ to 3′ exonuclease activity, but do exhibit 3′ to 5′ exonuclease activity. Pol III DNA polymerases represent the major replicative DNA polymerase of the cell and are composed of multiple subunits. The pol III catalytic subunit lacks 5′ to 3′ exonuclease activity, but in some cases 3′ to 5′ exonuclease activity is located in the same polypeptide.


There are a variety of commercially available Pol I DNA polymerases, some of which have been modified to reduce or abolish 5′ to 3′ exonuclease activity.


Suitable thermostable pol I DNA polymerases can be isolated from a variety of thermophilic eubacteria, including Thermus species and Thermotoga maritima such as Thermus aquaticus (Taq), Thermus thermophilus (Tth) and Thermotoga maritima (Tma UlTma).


Additional eubacteria related to those listed above are described in Thermophilic Bacteria (Kristjansson, J. K., ed.) CRC Press, Inc., Boca Raton, Fla., 1992.


The invention further provides for chimeric or non-chimeric DNA polymerases that are chemically modified according to methods disclosed in U.S. Pat. Nos. 5,677,152, 6,479,264 and 6,183,998, the contents of which are hereby incorporated by reference in their entirety.


Additional archaea DNA polymerases related to those listed above are described in the following references: Archaea: A Laboratory Manual (Robb, F. T. and Place, A. R., eds.), Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1995 and Thermophilic Bacteria (Kristjansson, J. K., ed.) CRC Press, Inc., Boca Raton, Fla., 1992.


B. Exemplarily Reverse Transcriptases


In various embodiments, the multi-flap prime editors described herein comprise a reverse transcriptase as the polymerase. The disclosure contemplates any wild type reverse transcriptase obtained from any naturally-occurring organism or virus, or obtained from a commercial or non-commercial source. In addition, the reverse transcriptases usable in the multi-flap prime editors of the disclosure can include any naturally-occurring mutant RT, engineered mutant RT, or other variant RT, including truncated variants that retain function. The RTs may also be engineered to contain specific amino acid substitutions, such as those specifically disclosed herein.


Reverse transcriptases are multi-functional enzymes typically with three enzymatic activities including RNA- and DNA-dependent DNA polymerization activity, and an RNaseH activity that catalyzes the cleavage of RNA in RNA-DNA hybrids. Some mutants of reverse transcriptases have disabled the RNaseH moiety to prevent unintended damage to the mRNA. These enzymes that synthesize complementary DNA (cDNA) using mRNA as a template were first identified in RNA viruses. Subsequently, reverse transcriptases were isolated and purified directly from virus particles, cells or tissues. (e.g., see Kacian et al., 1971, Biochim. Biophys. Acta 46: 365-83; Yang et al., 1972, Biochem. Biophys. Res. Comm 47: 505-11; Gerard et al., 1975, J. Virol. 15: 785-97; Liu et al., 1977, Arch. Virol. 55 187-200; Kato et al., 1984, J. Virol. Methods 9: 325-39; Luke et al., 1990, Biochem. 29: 1764-69 and Le Grice et al., 1991, J. Virol. 65: 7004-07, each of which are incorporated by reference). More recently, mutants and fusion proteins have been created in the quest for improved properties such as thermostability, fidelity and activity. Any of the wild type, variant, and/or mutant forms of reverse transcriptase which are known in the art or which can be made using methods known in the art are contemplated herein.


The reverse transcriptase (RT) gene (or the genetic information contained therein) can be obtained from a number of different sources. For instance, the gene may be obtained from eukaryotic cells which are infected with retrovirus, or from a number of plasmids which contain either a portion of or the entire retrovirus genome. In addition, messenger RNA-like RNA which contains the RT gene can be obtained from retroviruses. Examples of sources for RT include, but are not limited to, Moloney murine leukemia virus (M-MLV or MLVRT); human T-cell leukemia virus type 1 (HTLV-1); bovine leukemia virus (BLV); Rous Sarcoma Virus (RSV); human immunodeficiency virus (HIV); yeast, including Saccharomyces, Neurospora, Drosophila; primates; and rodents. See, for example, Weiss, et al., U.S. Pat. No. 4,663,290 (1987); Gerard, G. R., DNA:271-79 (1986); Kotewicz, M. L., et al., Gene 35:249-58 (1985); Tanese, N., et al., Proc. Natl. Acad. Sci. (USA):4944-48 (1985); Roth, M. J., at al., J. Biol. Chem. 260:9326-35 (1985); Michel, F., et al., Nature 316:641-43 (1985); Akins, R. A., et al., Cell 47:505-16 (1986), EMBO J. 4:1267-75 (1985); and Fawcett, D. F., Cell 47:1007-15 (1986) (each of which are incorporated herein by reference in their entireties).


Wild Type RTs


Exemplary enzymes for use with the herein disclosed multi-flap prime editors can include, but are not limited to, M-MLV reverse transcriptase and RSV reverse transcriptase. Enzymes having reverse transcriptase activity are commercially available. In certain embodiments, the reverse transcriptase provided in trans to the other components of the multi-flap prime editor (PE) system. That is, the reverse transcriptase is expressed or otherwise provided as an individual component, i.e., not as a fusion protein with a napDNAbp.


A person of ordinary skill in the art will recognize that wild type reverse transcriptases, including but not limited to, Moloney Murine Leukemia Virus (M-MLV); Human Immunodeficiency Virus (HIV) reverse transcriptase and avian Sarcoma-Leukosis Virus (ASLV) reverse transcriptase, which includes but is not limited to Rous Sarcoma Virus (RSV) reverse transcriptase, Avian Myeloblastosis Virus (AMV) reverse transcriptase, Avian Erythroblastosis Virus (AEV) Helper Virus MCAV reverse transcriptase, Avian Myelocytomatosis Virus MC29 Helper Virus MCAV reverse transcriptase, Avian Reticuloendotheliosis Virus (REV-T) Helper Virus REV-A reverse transcriptase, Avian Sarcoma Virus UR2 Helper Virus UR2AV reverse transcriptase, Avian Sarcoma Virus Y73 Helper Virus YAV reverse transcriptase, Rous Associated Virus (RAV) reverse transcriptase, and Myeloblastosis Associated Virus (MAV) reverse transcriptase may be suitably used in the subject methods and composition described herein.


Exemplary wild type RT enzymes are as follows:













DESCRIPTION
SEQUENCE







REVERSE
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQA


TRANSCRIPTASE (M-
PLIIPLKATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWN


MLV RT) WILD TYPE
TPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPS


MOLONEY MURINE
HQWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRL


LEUKEMIA VIRUS
PQGFKNSPTLFDEALHRDLADFRIQHPDLILLQYVDDLLLAATSELD


USED IN PE1 (PRIME
CQQGTRALLQTLGNLGYRASAKKAQICQKQVKYLGYLLKEGQRW


EDITOR 1 FUSION
LTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPGFAEMAAPLYP


PROTEIN DISCLOSED
LTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEK


HEREIN)
QGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAV



LTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALL



LDTDRVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTD



QPLPDADHTWYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTS



AQRAELIALTQALKMAEGKKLNVYTDSRYAFATAHIHGEIYRRRGLL



TSEGKEIKNKDEILALLKALFLPKRLSIIHCPGHQKGHSAEARGNRM



ADQAARKAAITETPDTSTLLIENSSP (SEQ ID NO: 700)





REVERSE
AFPLERPDWDYTTQAGRNHLVHYRQLLLAGLQNAGRSPTNLAKVK


TRANSCRIPTASE
GITQGPNESPSAFLERLKEAYRRYTPYDPEDPGQETNVSMSFIWQSA


MOLONEY MURINE
PDIGRKLGRLEDLKSKTLGDLVREAEKIFNKRETPEEREERIRRETEE


LEUKEMIA VIRUS
KEERRRTVDEQKEKERDRRRHREMSKLLATVVIGQEQDRQEGERK


REF SEQ. AAA66622.1
RPQLDKDQCAYCKEKGHWAKDCPKKPRGPRGPRPQTSLLTLGDXG



GQGQDPPPEPRITLKVGGQPVTFLVDTGAQHSVLTQNPGPLSDKSA



WVQGATGGKRYRWTTDRKVHLATGKVTHSFLHVPDCPYPLLGRDL



LTKLKAQIHFEGSGAQVVGPMGQPLQVLTLNIEDEYRLHETSKEPDV



SLGFTWLSDFPQAWAESGGMGLAVRQAPLIIPLKATSTPVSIKQYPM



SQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQ



DLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR



LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFDEALHRD



LADFR (SEQ ID NO: 90)





REVERSE
TLQLEEEYRLFEPESTQKQEMDIWLKNFPQAWAETGGMGTAHCQA


TRANSCRIPTASE
PVLIQLKATATPISIRQYPMPHEAYQGIKPHIRRMLDQGILKPCQSPW


FELINE LEUKEMIA
NTPLLPVKKPGTEDYRPVQDLREVNKRVEDIHPTVPNPYNLLSTLPP


VIRUS
SHPWYTVLDLKDAFFCLRLHSESQLLFAFEWRDPEIGLSGQLTWTRL


REF SEQ. NP955579.1
PQGFKNSPTLFDEALHSDLADFRVRYPALVLLQYVDDLLLAAATRTE



CLEGTKALLETLGNKGYRASAKKAQICLQEVTYLGYSLKDGQRWL



TKARKEAILSIPVPKNSRQVREFLGTAGYCRLWIPGFAELAAPLYPLT



RPGTLFQWGTEQQLAFEDIKKALLSSPALGLPDITKPFELFIDENSGF



AKGVLVQKLGPWKRPVAYLSKKLDTVASGWPPCLRMVAAIAILVKD



AGKLTLGQPLTILTSHPVEALVRQPPNKWLSNARMTHYQAMLLDAE



RVHFGPTVSLNPATLLPLPSGGNHHDCLQILAETHGTRPDLTDQPLPD



ADLTWYTDGSSFIRNGEREAGAAVTTESEVIWAAPLPPGTSAQRAEL



IALTQALKMAEGKKLTVYTDSRYAFATTHVHGEIYRRRGLLTSEGKE



IKNKNEILALLEALFLPKRLSIIHCPGHQKGDSPQAKGNRLADDTAK



KAATETHSSLTVL



SEQ ID NO: 91)





REVERSE
PISPIETVPVKLKPGMDGPKVKQWPLTEEKIKALVEICTEMEKEGKIS


TRANSCRIPTASE
KIGPENPYNTPVFAIKKKDSTKWRKLVDFRELNKRTQDFWEVQLGIP


HIV-1 RT, CHAIN A
HPAGLKKKKSVTVLDVGDAYFSVPLDEDFRKYTAFTIPSINNETPGIR


REF SEQ. ITL3-A
YQYNVLPQGWKGSPAIFQSSMTKILEPFRKQNPDIVIYQYMDDLYV



GSDLEIGQHRTKIEELRQHLLRWGLTTPDKKHQKEPPFLWMGYELH



PDKWTVQPIVLPEKDSWTVNDIQKLVGKLNWASQIYPGIKVRQLXK



LLRGTKALTEVIPLTEEAELELAENREILKEPVHGVYYDPSKDLIAEI



QKQGQGQWTYQIYQEPFKNLKTGKYARMRGAHTNDVKQLTEAVQ



KITTESIVIWGKTPKFKLPIQKETWETWWTEYWQATWIPEWEFVNT



PPLVKLWYQLEKEPIVGAETFYVDGAANRETKLGKAGYVTNRGRQ



KVVTLTDTTNQKTELQAIYLALQDSGLEVNIVTDSQYALGIIQAQPD



QSESELVNQIIEQLIKKEKVYLAWVPAHKGIGGNEQVDKLVSAGIRK



V (SEQ ID NO: 92)



SEE MARTINELLI ET AL., VIROLOGY, 1990, 174(1): 135-



144, WHICH IS INCORPORATED BY REFERENCE





REVERSE
PISPIETVPVKLKPGMDGPKVKQWPLTEEKIKALVEICTEMEKEGKIS


TRANSCRIPTASE
KIGPENPYNTPVFAIKKKDSTKWRKLVDFRELNKRTQDFWEVQLGIP


HIV-1 RT, CHAIN B
HPAGLKKKKSVTVLDVGDAYFSVPLDEDFRKYTAFTIPSINNETPGIR


REF SEQ. ITL3-B
YQYNVLPQGWKGSPAIFQSSMTKILEPFRKQNPDIVIYQYMDDLYV



GSDLEIGQHRTKIEELRQHLLRWGLTTPDKKHQKEPPFLWMGYELH



PDKWTVQPIVLPEKDSWTVNDIQKLVGKLNWASQIYPGIKVRQLCK



LLRGTKALTEVIPLTEEAELELAENREILKEPVHGVYYDPSKDLIAEI



QKQGQGQWTYQIYQEPFKNLKTGKYARMRGAHTNDVKQLTEAVQ



KITTESIVIWGKTPKFKLPIQKETWETWWTEYWQATWIPEWEFVNT



PPLVKLWYQLEKEPIVGAETF (SEQ ID NO: 93)



SEE STAMMERS ET AL., J. MOL. BIOL., 1994, 242(4): 586-



588, WHICH IS INCORPORATED BY REFERENCE





REVERSE
TVALHLAIPLKWKPNHTPVWIDQWPLPEGKLVALTQLVEKELQLGHI


TRANSCRIPTASE
EPSLSCWNTPVFVIRKASGSYRLLHDLRAVNAKLVPFGAVQQGAPV


ROUS SARCOMA
LSALPRGWPLMVLDLKDCFFSIPLAEQDREAFAFTLPSVNNQAPARR


VIRUS RT
FQWKVLPQGMTCSPTICQLIVGQILEPLRLKHPSLRMLHYMDDLLL


REF SEQ. ACL14945
AASSHDGLEAAGEEVISTLERAGFTISPDKVQKEPGVQYLGYKLGST



YAAPVGLVAEPRIATLWDVQKLVGSLQWLRPALGIPPRLRGPFYEQL



RGSDPNEAREWNLDMKMAWREIVQLSTTAALERWDPALPLEGAVA



RCEQGAIGVLGQGLSTHPRPCLWLFSTQPTKAFTAWLEVLTLLITKL



RASAVRTFGKEVDILLLPACFRDELPLPEGILLALRGFAGKIRSSDTPS



IFDIARPLHVSLKVRVTDHPVPGPTVFTDASSSTHKGVVVWREGPR



WEIKEIADLGASVQQLEARAVAMALLLWPTTPTNVVTDSAFVAKML



LKMGQEGVPSTAAAFILEDALSQRSAMAAVLHVRSHSEVPGFFTEG



NDVADSQATFQAYPLREAKDLHTALHIGPRALSKACNISMQQAREV



VQTCPHCNSAPALEAGVNPRGLGPLQIWQTDFTLEPRMAPRSWLAV



TVDTASSAIVVTQHGRVTSVAAQHHWATVIAVLGRPKAIKTDNGSCF



TSKSTREWLARWGIAHTTGIPGNSQGQAMVERANRLLKDKIRVLAE



GDGFMKRIPTSKQGELLAKAMYALNHFERGENTKTPIQKHWRPTVL



TEGPPVKIRIETGEWEKGWNVLVWGRGYAAVKNRDTDKVIWVPSR



KVKPDIAQKDEVTKKDEASPLFA (SEQ ID NO: 94)



SEE YASUKAWA ET AL., J. BIOCHEM. 2009, 145(3): 315-



324, WHICH IS INCORPORATED BY REFERENCE





REVERSE
MMDHLLQKTQIQNQTEQVMNITNPNSIYIKGRLYFKGYKKIELHCF


TRANSCRIPTASE
VDTGASLCIASKFVIPEEHWINAERPIMVKIADGSSITINKVCRDIDLII


CAULIFLOWER
AGEIFHIPTVYQQESGIDFIIGNNFCQLYEPFIQFTDRVIFTKDRTYPVH


MOSAIC VIRUS RT
IAKLTRAVRVGTEGFLESMKKRSKTQQPEPVNISTNKIAILSEGRRLS


REF SEQ. AGT42196
EEKLFITQQRMQKIEELLEKVCSENPLDPNKTKQWMKASIKLSDPSK



AIKVKPMKYSPMDREEFDKQIKELLDLKVIKPSKSPHMAPAFLVNNE



AEKRRGKKRMVVNYKAMNKATVGDAYNLPNKDELLTLIRGKKIFS



SFDCKSGFWQVLLDQDSRPLTAFTCPQGHYEWNVVPFGLKQAPSIF



QRHMDEAFRVFRKFCCVYVDDILVFSNNEEDHLLHVAMILQKCNQH



GIILSKKKAQLFKKKINFLGLEIDEGTHKPQGHILEHINKFPDTLEDK



KQLQRFLGILTYASDYIPKLAQIRKPLQAKLKENVPWKWTKEDTLY



MQKVKKNLQGFPPLHHPLPEEKLIIETDASDDYWGGMLKAIKINEG



TNTELICRYASGSFKAAEKNYHSNDKETLAVINTIKKFSIYLTPVHFLI



RTDNTHFKSFVNLNYKGDSKLGRNIRWQAWLSHYSFDVEHIKGTD



NHFADFLSREFNRVNS (SEQ ID NO: 95)



SEE FARZADFAR ET AL., VIRUS GENES, 2013, 47(2): 347-



356, WHICH IS INCORPORATED BY REFERENCE





REVERSE
MKEKISKIDKNFYTDIFIKTSFQNEFEAGGVIPPIAKNQVSTISNKNKT


TRANSCRIPTASE
FYSLAHSSPHYSIQTRIEKFLLKNIPLSASSFAFRKERSYLHYLEPHTQ


KLEBSIELLA
NVKYCHLDIVSFFHSIDVNIVRDTFSVYFSDEFLVKEKQSLLDAFMA


PNEUMONIA
SVTLTAELDGVEKTFIPMGFKSSPSISNIIFRKIDILIQKFCDKNKITYT


REF SEQ. RFF81513.1
RYADDLLFSTKKENNILSSTFFINEISSILSINKFKLNKSKYLYKEGTIS



LGGYVIENILKDNSSGNIRLSSSKLNPLYKALYEIKKGSSSKHICIKVF



NLKLKRFIYKKNKEKFEAKFYSSQLKNKLLGYRSYLLSFVIFHKKYK



CINPIFLEKCVFLISEIESIMNRKF(SEQ ID NO: 96)





REVERSE
MKITSNNVTAVINGKGWHSINWKKCHQHVKTIQTRIAKAACNQQW


TRANSCRIPTASE
RTVGRLQRLLVRSFSARALAVKRVTENSGRKTPGVDGQIWSTPESK



ESCHERICHIA COLI

WEAIFKLRRKGYKPLPLKRVFIPKSNGKKRPLGIPVMLDRAMQALH


RT
LLGLEPVSETNADHNSYGFRPARCTADAIQQVCNMYSSRNASKWVL


REF SEQ. TGH57013
EGDIKGCFEHISHEWLLENIPMDKQILRNWLKAGIIEKSIFSKTLSGTP



QGGIISPVLANMALDGLERLLQNRFGRNRLI (SEQ ID NO: 97)





REVERSE
MSKIKINYEKYHIKPFPHFDQRIKVNKKVKENLQNPFYIAAHSFYPFI


TRANSCRIPTASE
HYKKISYKFKNGTLSSPKERDIFYSGHMDGYIYKHYGEILNHKYNN



BACILLUS SUBTILIS

TCIGKGIDHVSLAYRNNKMGKSNIHFAAEVINFISEQQQAFIFVSDFS


RT
SYFDSLDHAILKEKLIEVLEEQDKLSKDWWNVFKHITRYNWVEKEE


REF SEQ. QBJ66766
VISDLECTKEKIARDKKSRERYYTPAEFREFRKRVNIKSNDTGVGIPQ



GTAISAVLANVYAIDLDQKLNQYALKYGGIYRRYSDDIIMVLPMTSD



GQDPSNDHVSFIKSVVKRNKVTMGDSKTSVLYYANNNIYEDYQRK



RESKMDYLGFSFDGMTVKIREKSLFKYYHRTYKKINSINWASVKKE



KKVGRKKLYLLYSHLGRNYKGHGNFISYCKKAHAVFEGNKKIESLI



NQQIKRHWKKIQKRLVDV(SEQ ID NO: 98)






EUBACTERIUM

DTSNLMEQILSSDNLNRAYLQVVRNKGAEGVDGMKYTELKEHLAK


RECTALE GROUP II
NGETIKGQLRTRKYKPQPARRVEIPKPDGGVRNLGVPTVTDRFIQQA


INTRON RT
IAQVLTPIYEEQFHDHSYGFRPNRCAQQAILTALNIMNDGNDWIVDI



DLEKFFDTVNHDKLMTLIGRTIKDGDVISIVRKYLVSGIMIDDEYEDS



IVGTPQGGNLSPLLANIMLNELDKEMEKRGLNFVRYADDCIIMVGSE



MSANRVMRNISRFIEEKLGLKVNMTKSKVDRPSGLKYLGFGFYFDP



RAHQFKAKPHAKSVAKFKKRMKELTCRSWGVSNSYKVEKLNQLIR



GWINYFKIGSMKTLCKELDSRIRYRLRMCIWKQWKTPQNQEKNLV



KLGIDRNTARRVAYTGKRIAYVCNKGAVNVAISNKRLASFGLISMLD



YYIEKCVTC (SEQ ID NO: 99)






GEOBACILLUS

ALLERILARDNLITALKRVEANQGAPGIDGVSTDQLRDYIRAHWSTI



STEAROTHERMOPHIL

HAQLLAGTYRPAPVRRVEIPKPGGGTRQLGIPTVVDRLIQQAILQELT


US GROUP II INTRON
PIFDPDFSSSSFGFRPGRNAHDAVRQAQGYIQEGYRYVVDMDLEKFF


RT
DRVNHDILMSRVARKVKDKRVLKLIRAYLQAGVMIEGVKVQTEEGT



PQGGPLSPLLANILLDDLDKELEKRGLKFCRYADDCNIYVKSLRAGQ



RVKQSIQRFLEKTLKLKVNEEKSAVDRPWKRAFLGFSFTPERKARIR



LAPRSIQRLKQRIRQLTNPNWSISMPERIHRVNQYVMGWIGYFRLVE



TPSVLQTIEGWIRRRLRLCQWLQWKRVRTRIRELRALGLKETAVMEI



ANTRKGAWRTTKTPQLHQALGKTYWTAQGLKSLTQR(SEQ ID NO:



100)










Variant and Error-Prone RTs


Reverse transcriptases are essential for synthesizing complementary DNA (cDNA) strands from RNA templates. Reverse transcriptases are enzymes composed of distinct domains that exhibit different biochemical activities. The enzymes catalyze the synthesis of DNA from an RNA template, as follows: In the presence of an annealed primer, reverse transcriptase binds to an RNA template and initiates the polymerization reaction. RNA-dependent DNA polymerase activity synthesizes the complementary DNA (cDNA) strand, incorporating dNTPs. RNase H activity degrades the RNA template of the DNA:RNA complex. Thus, reverse transcriptases comprise (a) a binding activity that recognizes and binds to a RNA/DNA hybrid, (b) an RNA-dependent DNA polymerase activity, and (c) an RNase H activity. In addition, reverse transcriptases generally are regarded as having various attributes, including their thermostability, processivity (rate of dNTP incorporation), and fidelity (or error-rate). The reverse transcriptase variants contemplated herein may include any mutations to reverse transcriptase that impacts or changes any one or more of these enzymatic activities (e.g., RNA-dependent DNA polymerase activity, RNase H activity, or DNA/RNA hybrid-binding activity) or enzyme properties (e.g., thermostability, processivity, or fidelity). Such variants may be available in the art in the public domain, available commercially, or may be made using known methods of mutagenesis, including directed evolutionary processes (e.g., PACE or PANCE).


In various embodiments, the reverse transcriptase may be a variant reverse transcriptase. As used herein, a “variant reverse transcriptase” includes any naturally occurring or genetically engineered variant comprising one or more mutations (including singular mutations, inversions, deletions, insertions, and rearrangements) relative to a reference sequences (e.g., a reference wild type sequence). RT naturally have several activities, including an RNA-dependent DNA polymerase activity, ribonuclease H activity, and DNA-dependent DNA polymerase activity. Collectively, these activities enable the enzyme to convert single-stranded RNA into double-stranded cDNA. In retroviruses and retrotransposons, this cDNA can then integrate into the host genome, from which new RNA copies can be made via host-cell transcription. Variant RT's may comprise a mutation which impacts one or more of these activities (either which reduces or increases these activities, or which eliminates these activities all together). In addition, variant RTs may comprise one or more mutations which render the RT more or less stable, less prone to aggregation, and facilitates purification and/or detection, and/or other the modification of properties or characteristics.


A person of ordinary skill in the art will recognize that variant reverse transcriptases derived from other reverse transcriptases, including but not limited to Moloney Murine Leukemia Virus (M-MLV); Human Immunodeficiency Virus (HIV) reverse transcriptase and avian Sarcoma-Leukosis Virus (ASLV) reverse transcriptase, which includes but is not limited to Rous Sarcoma Virus (RSV) reverse transcriptase, Avian Myeloblastosis Virus (AMV) reverse transcriptase, Avian Erythroblastosis Virus (AEV) Helper Virus MCAV reverse transcriptase, Avian Myelocytomatosis Virus MC29 Helper Virus MCAV reverse transcriptase, Avian Reticuloendotheliosis Virus (REV-T) Helper Virus REV-A reverse transcriptase, Avian Sarcoma Virus UR2 Helper Virus UR2AV reverse transcriptase, Avian Sarcoma Virus Y73 Helper Virus YAV reverse transcriptase, Rous Associated Virus (RAV) reverse transcriptase, and Myeloblastosis Associated Virus (MAV) reverse transcriptase may be suitably used in the subject methods and composition described herein.


One method of preparing variant RTs is by genetic modification (e.g., by modifying the DNA sequence of a wild-type reverse transcriptase). A number of methods are known in the art that permit the random as well as targeted mutation of DNA sequences (see for example, Ausubel et. al. Short Protocols in Molecular Biology (1995) 3.sup.rd Ed. John Wiley & Sons, Inc.). In addition, there are a number of commercially available kits for site-directed mutagenesis, including both conventional and PCR-based methods. Examples include the QuikChange Site-Directed Mutagenesis Kits (AGILENT®), the Q5® Site-Directed Mutagenesis Kit (NEW ENGLAND BIOLABS®), and GeneArt™ Site-Directed Mutagenesis System (THERMOFISHER SCIENTIFIC®).


In addition, mutant reverse transcriptases may be generated by insertional mutation or truncation (N-terminal, internal, or C-terminal insertions or truncations) according to methodologies known to one skilled in the art. The term “mutation,” as used herein, refers to a substitution of a residue within a sequence, e.g., a nucleic acid or amino acid sequence, with another residue, or a deletion or insertion of one or more residues within a sequence. Mutations are typically described herein by identifying the original residue followed by the position of the residue within the sequence and by the identity of the newly substituted residue. Various methods for making the amino acid substitutions (mutations) provided herein are well known in the art, and are provided by, for example, Green and Sambrook, Molecular Cloning: A Laboratory Manual (4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2012)). Mutations can include a variety of categories, such as single base polymorphisms, microduplication regions, indel, and inversions, and is not meant to be limiting in any way. Mutations can include “loss-of-function” mutations which is the normal result of a mutation that reduces or abolishes a protein activity. Most loss-of-function mutations are recessive, because in a heterozygote the second chromosome copy carries an unmutated version of the gene coding for a fully functional protein whose presence compensates for the effect of the mutation. Mutations also embrace “gain-of-function” mutations, which is one which confers an abnormal activity on a protein or cell that is otherwise not present in a normal condition. Many gain-of-function mutations are in regulatory sequences rather than in coding regions, and can therefore have a number of consequences. For example, a mutation might lead to one or more genes being expressed in the wrong tissues, these tissues gaining functions that they normally lack. Because of their nature, gain-of-function mutations are usually dominant.


Older methods of site-directed mutagenesis known in the art rely on sub-cloning of the sequence to be mutated into a vector, such as an M13 bacteriophage vector, that allows the isolation of single-stranded DNA template. In these methods, one anneals a mutagenic primer (i.e., a primer capable of annealing to the site to be mutated but bearing one or more mismatched nucleotides at the site to be mutated) to the single-stranded template and then polymerizes the complement of the template starting from the 3′ end of the mutagenic primer. The resulting duplexes are then transformed into host bacteria and plaques are screened for the desired mutation.


More recently, site-directed mutagenesis has employed PCR methodologies, which have the advantage of not requiring a single-stranded template. In addition, methods have been developed that do not require sub-cloning. Several issues must be considered when PCR-based site-directed mutagenesis is performed. First, in these methods it is desirable to reduce the number of PCR cycles to prevent expansion of undesired mutations introduced by the polymerase. Second, a selection must be employed in order to reduce the number of non-mutated parental molecules persisting in the reaction. Third, an extended-length PCR method is preferred in order to allow the use of a single PCR primer set. And fourth, because of the non-template-dependent terminal extension activity of some thermostable polymerases it is often necessary to incorporate an end-polishing step into the procedure prior to blunt-end ligation of the PCR-generated mutant product.


Methods of random mutagenesis, which will result in a panel of mutants bearing one or more randomly situated mutations, exist in the art. Such a panel of mutants may then be screened for those exhibiting the desired properties, for example, increased stability, relative to a wild-type reverse transcriptase.


An example of a method for random mutagenesis is the so-called “error-prone PCR method.” As the name implies, the method amplifies a given sequence under conditions in which the DNA polymerase does not support high fidelity incorporation. Although the conditions encouraging error-prone incorporation for different DNA polymerases vary, one skilled in the art may determine such conditions for a given enzyme. A key variable for many DNA polymerases in the fidelity of amplification is, for example, the type and concentration of divalent metal ion in the buffer. The use of manganese ion and/or variation of the magnesium or manganese ion concentration may therefore be applied to influence the error rate of the polymerase.


In various aspects, the RT of the multi-flap prime editors may be an “error-prone” reverse transcriptase variant. Error-prone reverse transcriptases that are known and/or available in the art may be used. It will be appreciated that reverse transcriptases naturally do not have any proofreading function; thus the error rate of reverse transcriptase is generally higher than DNA polymerases comprising a proofreading activity. The error-rate of any particular reverse transcriptase is a property of the enzyme's “fidelity,” which represents the accuracy of template-directed polymerization of DNA against its RNA template. An RT with high fidelity has a low-error rate. Conversely, an RT with low fidelity has a high-error rate. The fidelity of M-MLV-based reverse transcriptases are reported to have an error rate in the range of one error in 15,000 to 27,000 nucleotides synthesized. See Boutabout et al., “DNA synthesis fidelity by the reverse transcriptase of the yeast retrotransposon Ty1,” Nucleic Acids Res, 2001, 29: 2217-2222, which is incorporated by reference. Thus, for purposes of this application, those reverse transcriptases considered to be “error-prone” or which are considered to have an “error-prone fidelity” are those having an error rate that is less than one error in 15,000 nucleotides synthesized.


Error-prone reverse transcriptase also may be created through mutagenesis of a starting RT enzyme (e.g., a wild type M-MLV RT). The method of mutagenesis is not limited and may include directed evolution processes, such as phage-assisted continuous evolution (PACE) or phage-assisted noncontinuous evolution (PANCE). The term “phage-assisted continuous evolution (PACE),” as used herein, refers to continuous evolution that employs phage as viral vectors. The general concept of PACE technology has been described, for example, in International PCT Application, PCT/US2009/056194, filed Sep. 8, 2009, published as WO 2010/028347 on Mar. 11, 2010; International PCT Application, PCT/US2011/066747, filed Dec. 22, 2011, published as WO 2012/088381 on Jun. 28, 2012; U.S. Application, U.S. Pat. No. 9,023,594, issued May 5, 2015, International PCT Application, PCT/US2015/012022, filed Jan. 20, 2015, published as WO 2015/134121 on Sep. 11, 2015, and International PCT Application, PCT/US2016/027795, filed Apr. 15, 2016, published as WO 2016/168631 on Oct. 20, 2016, the entire contents of each of which are incorporated herein by reference.


Error-prone reverse transcriptases may also be obtain by phage-assisted non-continuous evolution (PANCE),” which as used herein, refers to non-continuous evolution that employs phage as viral vectors. PANCE is a simplified technique for rapid in vivo directed evolution using serial flask transfers of evolving ‘selection phage’ (SP), which contain a gene of interest to be evolved, across fresh E. coli host cells, thereby allowing genes inside the host E. coli to be held constant while genes contained in the SP continuously evolve. Serial flask transfers have long served as a widely-accessible approach for laboratory evolution of microbes, and, more recently, analogous approaches have been developed for bacteriophage evolution. The PANCE system features lower stringency than the PACE system.


Other error-prone reverse transcriptases have been described in the literature, each of which are contemplated for use in the herein methods and compositions. For example, error-prone reverse transcriptases have been described in Bebenek et al., “Error-prone Polymerization by HIV-1 Reverse Transcriptase,” J Biol Chem, 1993, Vol. 268: 10324-10334 and Sebastian-Martin et al., “Transcriptional inaccuracy threshold attenuates differences in RNA-dependent DNA synthesis fidelity between retroviral reverse transcriptases,” Scientific Reports, 2018, Vol. 8: 627, each of which are incorporated by reference. Still further, reverse transcriptases, including error-prone reverse transcriptases can be obtained from a commercial supplier, including ProtoScript® (II) Reverse Transcriptase, AMV Reverse Transcriptase, WarmStart® Reverse Transcriptase, and M-MuLV Reverse Transcriptase, all from NEW ENGLAND BIOLABS®, or AMV Reverse Transcriptase XL, SMARTScribe Reverse Transcriptase, GPR ultra-pure MMLV Reverse Transcriptase, all from TAKARA BIO USA, INC. (formerly CLONTECH).


The herein disclosure also contemplates reverse transcriptases having mutations in RNaseH domain. As mentioned above, one of the intrinsic properties of reverse transcriptases is the RNase H activity, which cleaves the RNA template of the RNA:cDNA hybrid concurrently with polymerization. The RNase H activity can be undesirable for synthesis of long cDNAs because the RNA template may be degraded before completion of full-length reverse transcription. The RNase H activity may also lower reverse transcription efficiency, presumably due to its competition with the polymerase activity of the enzyme. Thus, the present disclosure contemplates any reverse transcriptase variants that comprise a modified RNaseH activity.


The herein disclosure also contemplates reverse transcriptases having mutations in the RNA-dependent DNA polymerase domain. As mentioned above, one of the intrinsic properties of reverse transcriptases is the RNA-dependent DNA polymerase activity, which incorporates the nucleobases into the nascent cDNA strand as coded by the template RNA strand of the RNA:cDNA hybrid. The RNA-dependent DNA polymerase activity can be increased or decreased (i.e., in terms of its rate of incorporation) to either increase or decrease the processivity of the enzyme. Thus, the present disclosure contemplates any reverse transcriptase variants that comprise a modified RNA-dependent DNA polymerase activity such that the processivity of the enzyme of either increased or decreased relative to an unmodified version.


Also contemplated herein are reverse transcriptase variants that have altered thermostability characteristics. The ability of a reverse transcriptase to withstand high temperatures is an important aspect of cDNA synthesis. Elevated reaction temperatures help denature RNA with strong secondary structures and/or high GC content, allowing reverse transcriptases to read through the sequence. As a result, reverse transcription at higher temperatures enables full-length cDNA synthesis and higher yields, which can lead to an improved generation of the 3′ flap ssDNA as a result of the multi-flap prime editing process. Wild type M-MLV reverse transcriptase typically has an optimal temperature in the range of 37-48° C.; however, mutations may be introduced that allow for the reverse transcription activity at higher temperatures of over 48° C., including 49° C., 50° C., 51° C., 52° C., 53° C., 54° C., 55° C., 56° C., 57° C., 58° C., 59° C., 60° C., 61° C., 62° C., 63° C., 64° C., 65° C., 66° C., and higher.


The variant reverse transcriptases contemplated herein, including error-prone RTs, thermostable RTs, increase-processivity RTs, can be engineered by various routine strategies, including mutagenesis or evolutionary processes. In some cases, the variants can be produced by introducing a single mutation. In other cases, the variants may require more than one mutation. For those mutants comprising more than one mutation, the effect of a given mutation may be evaluated by introduction of the identified mutation to the wild-type gene by site-directed mutagenesis in isolation from the other mutations borne by the particular mutant. Screening assays of the single mutant thus produced will then allow the determination of the effect of that mutation alone.


Variant RT enzymes used herein may also include other “RT variants” having at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to any reference RT protein, including any wild type RT, or mutant RT, or fragment RT, or other variant of RT disclosed or contemplated herein or known in the art.


In some embodiments, an RT variant may have 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 21, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, or up to 100, or up to 200, or up to 300, or up to 400, or up to 500 or more amino acid changes compared to a reference RT. In some embodiments, the RT variant comprises a fragment of a reference RT, such that the fragment is at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to the corresponding fragment of the reference RT. In some embodiments, the fragment is at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% identical, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% of the amino acid length of a corresponding wild type RT (M-MLV reverse transcriptase) (e.g., SEQ ID NO: 89) or to any of the reverse transcriptases of SEQ ID NOs: 90-100.


In some embodiments, the disclosure also may utilize RT fragments which retain their functionality and which are fragments of any herein disclosed RT proteins. In some embodiments, the RT fragment is at least 100 amino acids in length. In some embodiments, the fragment is at least 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, or up to 600 or more amino acids in length.


In still other embodiments, the disclosure also may utilize RT variants which are truncated at the N-terminus or the C-terminus, or both, by a certain number of amino acids which results in a truncated variant which still retains sufficient polymerase function. In some embodiments, the RT truncated variant has a truncation of at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, or 250 amino acids at the N-terminal end of the protein. In other embodiments, the RT truncated variant has a truncation of at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, or 250 amino acids at the C-terminal end of the protein. In still other embodiments, the RT truncated variant has a truncation at the N-terminal and the C-terminal end which are the same or different lengths.


For example, the multi-flap prime editors disclosed herein may include a truncated version of M-MLV reverse transcriptase. In this embodiment, the reverse transcriptase contains 4 mutations (D200N, T306K, W313F, T330P; noting that the L603W mutation present in PE2 is no longer present due to the truncation). The DNA sequence encoding this truncated editor is 522 bp smaller than PE2, and therefore makes its potentially useful for applications where delivery of the DNA sequence is challenging due to its size (i.e., adeno-associated virus and lentivirus delivery). This embodiment is referred to as MMLV-RT(trunc) and has the following amino acid sequence:















MMLV-RT(TRUNC)

TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIP





LKATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVK





KPGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLK





DAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEA





LHRDLADFRIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGY





RASAKKAQICQKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLR





EFLGKAGFCRLFIPGFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQA





LLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLD





PVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPD





RWLSNARMTHYQALLLDTDRVQFGPWALNPATLLPLPEEGLQHNCLDN





SRLIN (SEQ ID NO: 766)










In various embodiments, the multi-flap prime editors disclosed herein may comprise one of the RT variants described herein, or a RT variant thereof having at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to any reference Cas9 variants.


In still other embodiments, the present methods and compositions may utilize a DNA polymerase that has been evolved into a reverse transcriptase, as described in Effefson et al., “Synthetic evolutionary origin of a proofreading reverse transcriptase,” Science, Jun. 24, 2016, Vol. 352: 1590-1593, the contents of which are incorporated herein by reference.


In certain other embodiments, the reverse transcriptase is provided as a component of a fusion protein also comprising a napDNAbp. In other words, in some embodiments, the reverse transcriptase is fused to a napDNAbp as a fusion protein.


In various embodiments, variant reverse transcriptases can be engineered from wild type M-MLV reverse transcriptase as represented by SEQ ID NO: 89.


In various embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising one or more of the following mutations: P51L, S67K, E69K, L139P, T197A, D200N, H204R, F209N, E302K, E302R, T306K, F309N, W313F, T330P, L345G, L435G, N454K, D524G, E562Q, D583N, H594Q, L603W, E607K, or D653N in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence.


Some exemplary reverse transcriptases that can be fused to napDNAbp proteins or provided as individual proteins according to various embodiments of this disclosure are provided below. Exemplary reverse transcriptases include variants with at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity to the following wild-type enzymes or partial enzymes:













Description
Sequence (variant substitutions relative to wild type)







Reverse
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


transcriptase
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


(M-MLV RT)
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


wild type
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFDEALHRDLADFRI


moloney
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


murine
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP


leukemia virus
GFAEMAAPLYPLTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE


Used in PE1
LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA


(prime editor 1
VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD


fusion protein
RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT


disclosed
WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK


herein)
MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKAL



FLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 89)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN



DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR



LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI



QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP



GFAEMAAPLYPLTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKAL



FLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 701)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR



LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI



QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP



GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKAL



FLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 702)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI



QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP



GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKA



LFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 740)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQKARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


E69K
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP



GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKA



LFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 703)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
atstpvsikqypmsqearlgikphiqrlldqgilvpcqspwntpllpvkkpgtn


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


E302R
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLRRFLGTAGFCRLWIP



GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKA



LFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 704)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


E607K
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP



GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSKGKEIKNKDEILALLKA



LFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 705)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGPPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


L139P
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP



GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKA



LFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 706)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


L435G
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP



GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVIGAPHAVEALVKQPPDRWLSNARMTHYQALLLDT



DRVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKA



LFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 707)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


N454K
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP



GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSKARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKA



LFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 708)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


T306K
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKAGFCRLWIP



GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKA



LFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 709)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


W313F
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLFIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFEL



FVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAV



LTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDR



VQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTW



YTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMA



EGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFL



PKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 710)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


D524G
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


E562Q
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP


D583N
GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTGGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAQLIALTQALK



MAEGKKLNVYTNSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKA



LFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 711)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


E302R
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


W313F
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLRRFLGTAGFCRLFIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFEL



FVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAV



LTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDR



VQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTW



YTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMA



EGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFL



PKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 712)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGPPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


E607K
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


L139P
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIP



GFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFE



LFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIA



VLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTD



RVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT



WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSKGKEIKNKDEILALLKA



LFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 713)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIILLK


P51L S67K
ATSTPVSIKQYPMKQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGT


T197A H204R
NDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCL


E302K F309N
RLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPALFDEALRRDLADFR


W313F T330P
IQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQ


L435G N454K
KQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLRKFLGTAGNCRLFI


D524G D583N
PGFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPF


H594Q D653N
ELFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAI



AVLTKDAGKLTMGQPLVIGAPHAVEALVKQPPDRWLSKARMTHYQALLLD



TDRVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADH



TWYTGGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTNSRYAFATAHIQGEIYRRRGLLTSEGKEIKNKDEILALLKAL



FLPKRLSIIHCPGHQKGHSAEARGNRMANQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 714)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIILLK


D200N P51L
ATSTPVSIKQYPMKQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGT


S67K T197A
NDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCL


H204R E302K
RLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPALFNEALRRDLADFR


F309N W313F
IQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQ


T330P L345G
KQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLRKFLGTAGNCRLFI


N454K D524G
PGFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPF


D583N H594Q
ELFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAI


D653N
AVLTKDAGKLTMGQPLVIGAPHAVEALVKQPPDRWLSKARMTHYQALLLD



TDRVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADH



TWYTGGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALK



MAEGKKLNVYTNSRYAFATAHIQGEIYRRRGLLTSEGKEIKNKDEILALLKAL



FLPKRLSIIHCPGHQKGHSAEARGNRMANQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 715)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N T330P
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


L603W T306K
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


W313F
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


in PE2
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFEL



FVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAV



LTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDR



VQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTW



YTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMA



EGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFL



PKRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP



(SEQ ID NO: 716)









In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising one or more of the following mutations: P51X, S67X, E69X, L139X, T197X, D200X, H204X, F209X, E302X, T306X, F309X, W313X, T330X, L345X, L435X, N454X, D524X, E562X, D583X, H594X, L603X, E607X, or D653X in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a P51X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is L.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a S67X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is K.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a E69X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is K.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a L139X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is P.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a T197X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is A.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a D200X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is N.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a H204X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is R.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a F209X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is N.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a E302X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is K.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a E302X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is R.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a T306X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is K.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a F309X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is N.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a W313X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is F.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a T330X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is P.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a L345X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is G.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a L435X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is G.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a N454X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is K.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a D524X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is G.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a E562X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is Q.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a D583X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is N.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a H594X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is Q.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a L603X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is W.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a E607X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is K.


In various other embodiments, the multi-flap prime editors described herein (with RT provided as either a fusion partner or in trans) can include a variant RT comprising a D653X mutation in the wild type M-MLV RT of SEQ ID NO: 89 or at a corresponding amino acid position in another wild type RT polypeptide sequence, wherein “X” can be any amino acid. In certain embodiments, X is N.


Some exemplary reverse transcriptases that can be fused to napDNAbp proteins or provided as individual proteins according to various embodiments of this disclosure are provided below. Exemplary reverse transcriptases include variants with at least 80%, at least 85%, at least 90%, at least 95% or at least 99% sequence identity to the following wild-type enzymes or partial enzymes:













DESCRIPTION
SEQUENCE (VARIANT SUBSTITUTIONS RELATIVE TO WILD TYPE)







REVERSE
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


TRANSCRIPTASE
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


(M-MLV
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


RT) WILD
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFDEALHRDLADFRI


TYPE
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


MOLONEY
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG


MURINE
FAEMAAPLYPLTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF


LEUKEMIA
VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT


VIRUS
KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ


USED IN PE1
FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD


(PRIME
GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK


EDITOR 1
KLNVYTDSRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLS


FUSION
IIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 89)





PROTEIN



DISCLOSED



HEREIN)



M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN



DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR



LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI



QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG



FAEMAAPLYPLTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLS



IIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 106)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR



LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI



QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLS



IIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 107)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI



QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 108)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


E69K
ATSTPVSIKQYPMSQKARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


D200N
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


T330P
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


L603W
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 109)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


E302R
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLRRFLGTAGFCRLWIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP(SEQ ID



NO: 110)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


E607K
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSKGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 111)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGPPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


L139P
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 112)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


L435G
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVIGAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 113)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


N454K
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSKARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 114)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


T306K
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKAGFCRLWIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 115)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


W313F
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLFIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 116)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


D524G
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


E562Q
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG


D583N
FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTG



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAQLIALTQALKMAEGK



KLNVYTNSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 117)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


E302R
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


W313F
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLRRFLGTAGFCRLFIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 118)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T330P
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGPPPSHQWYTVLDLKDAFFCLR


L603W
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


E607K
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


L139P
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSKGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 119)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIILLK


P51L S67K
ATSTPVSIKQYPMKQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T197A H204R
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


E302K F309N
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPALFDEALRRDLADFRI


W313F T330P
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


L435G N454K
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLRKFLGTAGNCRLFIPG


D524G D583N
FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF


H594Q D653N
VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVIGAPHAVEALVKQPPDRWLSKARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTG



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTNSRYAFATAHIQGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLS



IIHCPGHQKGHSAEARGNRMANQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 120)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIILLK


P51L S67K
ATSTPVSIKQYPMKQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


T197A D200N
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


H204R E302K
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPALFNEALRRDLADFRI


F309N W313F
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


T330P L345G
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLRKFLGTAGNCRLFIPG


N454K D524G
FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF


D583N H594Q
VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT


D653N
KDAGKLTMGQPLVIGAPHAVEALVKQPPDRWLSKARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTG



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTNSRYAFATAHIQGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLS



IIHCPGHQKGHSAEARGNRMANQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 121)





M-MLV RT
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


D200N T330P
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


L603W T306K
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


W313F
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


IN PE2
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPG



FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 122)









The multi-flap prime editor system described here contemplates any publicly-available reverse transcriptase described or disclosed in any of the following U.S. patents (each of which are incorporated by reference in their entireties): U.S. Pat. Nos. 10,202,658; 10,189,831; 10,150,955; 9,932,567; 9,783,791; 9,580,698; 9,534,201; and 9,458,484, and any variant thereof that can be made using known methods for installing mutations, or known methods for evolving proteins. The following references describe reverse transcriptases in art. Each of their disclosures are incorporated herein by reference in their entireties.

  • Herzig, E., Voronin, N., Kucherenko, N. & Hizi, A. A Novel Leu92 Mutant of HIV-1 Reverse Transcriptase with a Selective Deficiency in Strand Transfer Causes a Loss of Viral Replication. J. Virol. 89, 8119-8129 (2015).
  • Mohr, G. et al. A Reverse Transcriptase-Casl Fusion Protein Contains a Cas6 Domain Required for Both CRISPR RNA Biogenesis and RNA Spacer Acquisition. Mol. Cell 72, 700-714.e8 (2018).
  • Zhao, C., Liu, F. & Pyle, A. M. An ultraprocessive, accurate reverse transcriptase encoded by a metazoan group II intron. RNA 24, 183-195 (2018).
  • Zimmerly, S. & Wu, L. An Unexplored Diversity of Reverse Transcriptases in Bacteria. Microbiol Spectr 3, MDNA3-0058-2014 (2015).
  • Ostertag, E. M. & Kazazian Jr, H. H. Biology of Mammalian L1 Retrotransposons. Annual Review of Genetics 35, 501-538 (2001).
  • Perach, M. & Hizi, A. Catalytic Features of the Recombinant Reverse Transcriptase of Bovine Leukemia Virus Expressed in Bacteria. Virology 259, 176-189 (1999).
  • Lim, D. et al. Crystal structure of the moloney murine leukemia virus RNase H domain. J. Virol. 80, 8379-8389 (2006).
  • Zhao, C. & Pyle, A. M. Crystal structures of a group II intron maturase reveal a missing link in spliceosome evolution. Nature Structural & Molecular Biology 23, 558-565 (2016).
  • Griffiths, D. J. Endogenous retroviruses in the human genome sequence. Genome Biol. 2, REVIEWS1017 (2001).
  • Baranauskas, A. et al. Generation and characterization of new highly thermostable and processive M-MuLV reverse transcriptase variants. Protein Eng Des Sel 25, 657-668 (2012).
  • Zimmerly, S., Guo, H., Perlman, P. S. & Lambowltz, A. M. Group II intron mobility occurs by target DNA-primed reverse transcription. Cell 82, 545-554 (1995).
  • Feng, Q., Moran, J. V., Kazazian, H. H. & Boeke, J. D. Human L1 retrotransposon encodes a conserved endonuclease required for retrotransposition. Cell 87, 905-916 (1996).
  • Berkhout, B., Jebbink, M. & Zsíros, J. Identification of an Active Reverse Transcriptase Enzyme Encoded by a Human Endogenous HERV-K Retrovirus. Journal of Virology 73, 2365-2375 (1999).
  • Kotewicz, M. L., Sampson, C. M., D'Alessio, J. M. & Gerard, G. F. Isolation of cloned Moloney murine leukemia virus reverse transcriptase lacking ribonuclease H activity. Nucleic Acids Res 16, 265-277 (1988).
  • Arezi, B. & Hogrefe, H. Novel mutations in Moloney Murine Leukemia Virus reverse transcriptase increase thermostability through tighter binding to template-primer. Nucleic Acids Res 37, 473-481 (2009).
  • Blain, S. W. & Goff, S. P. Nuclease activities of Moloney murine leukemia virus reverse transcriptase. Mutants with altered substrate specificities. J. Biol. Chem. 268, 23585-23592 (1993).
  • Xiong, Y. & Eickbush, T. H. Origin and evolution of retroelements based upon their reverse transcriptase sequences. EMBO J 9, 3353-3362 (1990).
  • Herschhorn, A. & Hizi, A. Retroviral reverse transcriptases. Cell. Mol. Life Sci. 67, 2717-2747 (2010).
  • Taube, R., Loya, S., Avidan, O., Perach, M. & Hizi, A. Reverse transcriptase of mouse mammary tumour virus: expression in bacteria, purification and biochemical characterization. Biochem. J. 329 (Pt 3), 579-587 (1998).
  • Liu, M. et al. Reverse Transcriptase-Mediated Tropism Switching in Bordetella Bacteriophage. Science 295, 2091-2094 (2002).
  • Luan, D. D., Korman, M. H., Jakubczak, J. L. & Eickbush, T. H. Reverse transcription of R2Bm RNA is primed by a nick at the chromosomal target site: a mechanism for non-LTR retrotransposition. Cell 72, 595-605 (1993).
  • Nottingham, R. M. et al. RNA-seq of human reference RNA samples using a thermostable group II intron reverse transcriptase. RNA 22, 597-613 (2016).
  • Telesnitsky, A. & Goff, S. P. RNase H domain mutations affect the interaction between Moloney murine leukemia virus reverse transcriptase and its primer-template. Proc. Natl. Acad. Sci. U.S.A. 90, 1276-1280 (1993).
  • Halvas, E. K., Svarovskaia, E. S. & Pathak, V. K. Role of Murine Leukemia Virus Reverse Transcriptase Deoxyribonucleoside Triphosphate-Binding Site in Retroviral Replication and In Vivo Fidelity. Journal of Virology 74, 10349-10358 (2000).
  • Nowak, E. et al. Structural analysis of monomeric retroviral reverse transcriptase in complex with an RNA/DNA hybrid. Nucleic Acids Res 41, 3874-3887 (2013).
  • Stamos, J. L., Lentzsch, A. M. & Lambowitz, A. M. Structure of a Thermostable Group II Intron Reverse Transcriptase with Template-Primer and Its Functional and Evolutionary Implications. Molecular Cell 68, 926-939.e4 (2017).
  • Das, D. & Georgiadis, M. M. The Crystal Structure of the Monomeric Reverse Transcriptase from Moloney Murine Leukemia Virus. Structure 12, 819-829 (2004).
  • Avidan, O., Meer, M. E., Oz, I. & Hizi, A. The processivity and fidelity of DNA synthesis exhibited by the reverse transcriptase of bovine leukemia virus. European Journal of Biochemistry 269, 859-867 (2002).
  • Gerard, G. F. et al. The role of template-primer in protection of reverse transcriptase from thermal inactivation. Nucleic Acids Res 30, 3118-3129 (2002).
  • Monot, C. et al. The Specificity and Flexibility of L1 Reverse Transcription Priming at Imperfect T-Tracts. PLOS Genetics 9, e1003499 (2013).
  • Mohr, S. et al. Thermostable group II intron reverse transcriptase fusion proteins and their use in cDNA synthesis and next-generation RNA sequencing. RNA 19, 958-970 (2013).


Any of the references noted above which relate to reverse transriptases are hereby incorporated by reference in their entireties, if not already stated so.


[4] Prime Editors

The disclosure provides systems comprising prime editors. In one aspect, this disclosure provides systems for simultaneously editing both strands of a double-stranded DNA sequence at a target site to be edited comprising a first prime editor complex and a second prime editor complex, wherein each of the first and second prime editor complexes comprises (1) a prime editor comprising (i) a nucleic acid programmable DNA binding protein (napDNAbp), and (ii) a polypeptide having an RNA-dependent DNA polymerase activity; and (2) a pegRNA comprising a spacer sequence, gRNA core, a DNA synthesis template, and a primer binding site, wherein the DNA synthesis template of the pegRNA of the first prime editor complex encodes a first single-stranded DNA sequence and the DNA synthesis template of the pegRNA of the second prime editor complex encodes a second single-stranded DNA sequence, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence each comprises a region of complementarity to the other, and wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence form a duplex comprising an edited portion as compared to the DNA sequence at the target site to be edited, which integrates into the target site to be edited.


In another aspect, the present disclosure provides systems for editing one or more double-stranded DNA sequences, the system comprising:

    • a) a first prime editor complex comprising:
      • i. a first prime editor comprising a first nucleic acid programmable DNA binding protein (first napDNAbp) and a first polypeptide comprising an RNA-dependent DNA polymerase activity; and
      • ii. a first prime editing guide RNA (first PEgRNA) that binds to a first binding site on a first strand of a first double-stranded DNA sequence at a first target site to be edited;
    • b) a second prime editor complex comprising:
      • i. a second prime editor comprising a second nucleic acid programmable DNA binding protein (second napDNAbp) and a second polypeptide comprising an RNA-dependent DNA polymerase activity; and
      • ii. a second prime editing guide RNA (second PEgRNA) that binds to a second binding site on a second strand of the first double-stranded DNA sequence at the first target site to be edited;
    • c) a third prime editor complex comprising:
      • i. a third prime editor comprising a third nucleic acid programmable DNA binding protein (third napDNAbp) and a third polypeptide comprising an RNA-dependent DNA polymerase activity; and
      • ii. a third prime editing guide RNA (third PEgRNA) that binds to a first binding site on a first strand of a second double-stranded DNA sequence at a second target site to be edited;
    • d) a fourth prime editor complex comprising:
      • i. a fourth prime editor comprising a fourth nucleic acid programmable DNA binding protein (fourth napDNAbp) and a fourth polypeptide comprising an RNA-dependent DNA polymerase activity; and
      • ii. a fourth prime editing guide RNA (fourth PEgRNA) that binds to a second binding site on a second strand of the second double-stranded DNA sequence at the second target site to be edited;
        • wherein the first PEgRNA comprises a first DNA synthesis template encoding a first single-stranded DNA sequence, the second PEgRNA comprises a second DNA synthesis template encoding a second single-stranded DNA sequence, the third PEgRNA comprises a third DNA synthesis template encoding a third single-stranded DNA sequence, and the fourth PEgRNA comprises a fourth DNA synthesis template encoding a fourth single-stranded DNA sequence;
        • wherein the first and the third single-stranded DNA sequence each comprise a region of complementarity to the other; and wherein, wherein the second and the fourth single-stranded DNA sequence each comprise a region of complementarity to the other.


In some embodiments, the prime editors used in the systems described herein (e.g., the first prime editor, second prime editor, third prime editor, and/or fourth prime editor in the systems described above) are provided as fusion proteins. In some embodiments, the prime editor fusion proteins comprise a napDNAbp and a polymerase (e.g., DNA-dependent DNA polymerase or RNA-dependent DNA polymerase, such as, reverse transcriptase). In some embodiments, the napDNAbp and the polymerase are optionally joined by linker to form the fusion protein. Various configurations of the prime editor fusion proteins and additional domains of the fusion proteins are described further herein. In various embodiments, the systems and methods provided by the present disclosure contemplate the use of any of the prime editor fusion proteins described herein.


In some embodiments, the prime editor complexes used in the systems described herein comprise a prime editor (e.g., the first prime editor, second prime editor, third prime editor, and/or fourth prime editor in the systems described above) where the components of one or more of the prime editors are provided in trans, as is described in additional detail throughout the present specification. In some embodiments, the prime editor comprises a napDNAbp and a polymerase expressed in trans. In some embodiments, the napDNAbp and the polymerase are expressed from one or more vectors (e.g., both components are expressed from the same vector, or each component is expressed from a different vector). In certain embodiments, the prime editors comprise additional components as described herein expressed in trans. In some embodiments, the prime editors used in the systems described herein may comprise both one or more prime editors provided as fusion proteins and one or more prime editors whose components are provided in trans.


In some embodiments, the multi-flap prime editor systems described herein contemplate fusion proteins comprising a napDNAbp and a polymerase (e.g., DNA-dependent DNA polymerase or RNA-dependent DNA polymerase, such as, reverse transcriptase), and optionally joined by a linker. The application contemplates any suitable napDNAbp and polymerase (e.g., DNA-dependent DNA polymerase or RNA-dependent DNA polymerase, such as, reverse transcriptase) to be combined in a single fusion protein. Examples of napDNAbps and polymerases (e.g., DNA-dependent DNA polymerase or RNA-dependent DNA polymerase, such as, reverse transcriptase) are each defined herein. Since polymerases are well-known in the art, and the amino acid sequences are readily available, this disclosure is not meant in any way to be limited to those specific polymerases identified herein.


In various embodiments, the fusion proteins may comprise any suitable structural configuration. For example, the fusion protein may comprise from the N-terminus to the C-terminus direction, a napDNAbp fused to a polymerase (e.g., DNA-dependent DNA polymerase or RNA-dependent DNA polymerase, such as, reverse transcriptase). In other embodiments, the fusion protein may comprise from the N-terminus to the C-terminus direction, a polymerase (e.g., a reverse transcriptase) fused to a napDNAbp. The fused domain may optionally be joined by a linker, e.g., an amino acid sequence. In other embodiments, the fusion proteins may comprise the structure NH2-[napDNAbp]-[polymerase]-COOH; or NH[polymerase]-[napDNAbp]-COOH, wherein each instance of “]-[” indicates the presence of an optional linker sequence. In embodiments wherein the polymerase is a reverse transcriptase, the fusion proteins may comprise the structure NH2-[napDNAbp]-[RT]-COOH; or NH2-[RT]-[napDNAbp]-COOH, wherein each instance of “]-[” indicates the presence of an optional linker sequence.


An exemplary fusion protein is depicted in FIG. 14, which shows a fusion protein comprising an MLV reverse transcriptase (“MLV-RT”) fused to a nickase Cas9 (“Cas9(H840A)”) via a linker sequence. This example is not intended to limit scope of fusion proteins that may be utilized for the prime editor (PE) system described herein.


In various embodiments, the multi-flap prime editor fusion protein may have the following amino acid sequence (referred to herein as “PE1”), which includes a Cas9 variant comprising an H840A mutation (i.e., a Cas9 nickase) and an M-MLV RT wild type, as well as an N-terminal NLS sequence (19 amino acids) and an amino acid linker (32 amino acids) that joins the C-terminus of the Cas9 nickase domain to the N-terminus of the RT domain. The PE1 fusion protein has the following structure: [NLS]-[Cas9(H840A)]-[linker]-[MMLV_RT(wt)]. The amino acid sequence of PE1 and its individual components are as follows:













DESCRIPTION
SEQUENCE







PE1 FUSION

MKRTADGSEFESPKKKRKV
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFK



PROTEIN

VLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQ



CAS9(H840A)-

EIFSNEMAKYDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPT



MMLV_RT(WT)

IYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKL





FIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKN





GLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQ





YADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLK





ALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGT





EELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDN





REKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGAS





AQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK





PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDR





FNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKT





YAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDG





FANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGIL





QTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEE





GIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDY





DVDAIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQ





LLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQIL





DSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAH





DAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAK





YFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKV





LSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFD





SPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKG





YKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNELALPSKYVNFL





YLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANL





DKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTS





TKEVLDATLIHOSITGLYETRIDLSQLGGD

SGGSSGGSSGSETPGTSESATP







ESSGGSSGGSS

TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVR





QAPLIIPLKATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVK





KPGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFC





LRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFDEALHRDLADFRI





QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQKQVKY





LGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPGFAEMAAP





LYPLTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAK





GVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLV





ILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQFGPWALNPATLLPLPEE





GLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTDGSSLLQEGQRKAGAAVTTET





EVIWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTDSRYAFATAHIHGEIYRRRG





LLTSEGKEIKNKDEILALLKALFLPKRLSHHCPGHQKGHSAEARGNRMADQAARK





AAITETPDTSTLLIENSSP
SGGSKRTADGSEFEPKKKRKV (SEQ ID NO: 123)




KEY:



NUCLEAR LOCALIZATION SEQUENCE (NLS) TOP:(SEQ ID NO: 124),



BOTTOM: (SEQ ID NO: 133)



CAS9(H840A) (SEQ ID NO: 126)



33-AMINO ACID LINKER (SEQ ID NO: 127)



M-MLVREVERSE TRANSCRIPTASE (SEQ ID NO: 128)





PE1-N-
MKRTADGSEFESPKKKRKV (SEQ ID NO: 124)


TERMINAL



NLS






PE1-CAS9
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDS


(H840A)(MET
GETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEE


MINUS)
DKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMI



KFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARL



SKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDT



YDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKR



YDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIK



PILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYP



FLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKG



ASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKP



AFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNAS



LGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDD



KVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLI



HDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKV



MGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVEN



TQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSFLKDDSIDNK



VLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERG



GLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKS



KLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGD



YKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETN



GETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKL



IARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERS



SFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNE



LALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKR



VILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDR



KRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGD (SEQ ID NO: 130)





PE1-LINKER
SGGSSGGSSGSETPGTSESATPESSGGSSGGSS (SEQ ID NO: 127)


BETWEEN



CAS9



DOMAIN



ANDRT



DOMAIN (33



AMINO



ACIDS)






PE1-M-MLV
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


RT
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN



DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR



LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFDEALHRDLADFRI



QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK



QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG



FAEMAAPLYPLTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLS



IIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 132)





PE1-C-
SGGSKRTADGSEFEPKKKRKV (SEQ ID NO: 133)


TERMINAL



NLS









In another embodiment, the multi-flap prime editor fusion protein may have the following amino acid sequence (referred to herein as “PE2”), which includes a Cas9 variant comprising an H840A mutation (i.e., a Cas9 nickase) and an M-MLV RT comprising mutations D200N, T330P, L603W, T306K, and W313F, as well as an N-terminal NLS sequence (19 amino acids) and an amino acid linker (33 amino acids) that joins the C-terminus of the Cas9 nickase domain to the N-terminus of the RT domain. The PE2 fusion protein has the following structure: [NLS]-[Cas9(H840A)]-[linker]-[MMLV_RT(D200N)(T330P)(L603W)(T306K)(W313F)]. The amino acid sequence of PE2 is as follows:















PE2 FUSION

MKRTADGSEFESPKKKRKV
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFK



PROTEIN

VLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQ



CAS9(H840A)-

EIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPT



MMLV_RT

IYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKL



D200N

FIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKN



T330P

GLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQ



L603W

YADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLK



T306K

ALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGT



W313F

EELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDN





REKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGAS





AQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK





PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDR





FNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKT





YAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDG





FANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGIL





QTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEE





GIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDY





DVDAIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQ





LLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQIL





DSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAH





DAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAK





YFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKV





LSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFD





SPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKG





YKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNELALPSKYVNFL





YLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANL





DKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTS





TKEVLDATLIHOSITGLYETRIDLSQLGGD

SGGSSGGSSGSETPGTSESATP







ESSGGSSGGSS

TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQ





APLIIPLKATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPV





KKPGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFC





LRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI





QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQKQVKY





LGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPGFAEMAAP





LYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAK





GVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLV





ILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQFGPWALNPATLLPLPEEG





LQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTDGSSLLQEGQRKAGAAVTTETEV





IWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTDSRYAFATAHIHGEIYRRRG





WLTSEGKEIKNKDEILALLKALFLPKRLSIIHCPGHQKGHSAEARGNRMADQAARK





AAITETPDTSTLLIENSSP
SGGSKRTADGSEFEPKKKRKV (SEQ ID NO: 134)




KEY:



NUCLEAR LOCALIZATION SEQUENCE (NLS) TOP:(SEQ ID NO: 124),



BOTTOM: (SEQ ID NO: 133)



CAS9(H840A) (SEQ ID NO: 137)



33-AMINO ACID LINKER (SEQ ID NO: 127)



M-MLVREVERSE TRANSCRIPTASE (SEQ ID NO: 139)





PE2-N-
MKRTADGSEFESPKKKRKV (SEQ ID NO: 124)


TERMINAL



NLS






PE2-CAS9
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDS


(H840A)
GETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEE


(MET MINUS)
DKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMI



KFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARL



SKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDT



YDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKR



YDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIK



PILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYP



FLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKG



ASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKP



AFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNAS



LGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDD



KVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLI



HDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKV



MGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVEN



TQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSFLKDDSIDNK



VLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERG



GLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKS



KLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGD



YKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETN



GETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKL



IARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERS



SFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNE



LALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKR



VILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDR



KRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGD (SEQ ID NO: 141)





PE2-LINKER
SGGSSGGSSGSETPGTSESATPESSGGSSGGSS (SEQ ID NO: 127)


BETWEEN



CAS9



DOMAIN



ANDRT



DOMAIN 



(33 AMINO



ACIDS)






PE2-
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLK


MMLV_RT
ATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTN


D200N
DYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLR


T330P
LHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRI


L603W
QHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQK


T306K
QVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPG


W313F
FAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELF



VDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLT



KDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQ



FGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTD



GSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGK



KLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRL



SIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP (SEQ ID



NO: 143)





PE2-C-
SGGSKRTADGSEFEPKKKRKV (SEQ ID NO: 133)


TERMINAL



NLS









In still other embodiments, the prime editor fusion protein may have the following amino acid sequences:















PE FUSION

MKRYADGSEEESPKKKRKV
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETG



PROTEIN

GMGLAVRQAPLIIPLKATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSP



MMLV_RT

WNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTV



(WT)-32AA-

LDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFDEAL



CAS9(H840A)

HRDLADFRIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKA





QICQKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWI





PGFAEMAAPLYPLTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFV





DEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGK





LTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQFGPWALNP





ATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTDGSSLLQEGQRKA





GAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTDSRYAFATAHI





HGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLSHHCPGHQKGHSAEARGNR





MADQAARKAAITETPDTSTLLIENSSP

SGGSSGGSSGSETPGTSESATPESSGGS







SGGSS

DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNL





IGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFF





HRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKA





DLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENP





INASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPN





FKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAIL





LSDILRYNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFF





DQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQ





RTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGP





LARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLP





NEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLF





KTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKD





KDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKR





RRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLT





FKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGR





HKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVEN





TQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSFLKDDSID





NKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLT





KAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIR





EVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKY





PKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITL





ANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQT





GGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKG





KSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSL





FELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDN





EQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIR





EQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGL





YETRIDLSQLGGD
SGGSKRTADGSEFEPKKKRKV (SEQ ID NO: 145)




KEY:



NUCLEAR LOCALIZATION SEQUENCE (NLS) TOP:(SEQ ID NO: 124),



BOTTOM: (SEQ ID NO: 133)



CAS9(H840A) (SEQ ID NO: 147)



33-AMINO ACID LINKER (SEQ ID NO: 127)



M-MLVREVERSE TRANSCRIPTASE (SEQ ID NO: 149)





PE FUSION

MKRTADGSEEESPKKKRKV
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAET



PROTEIN

GGMGLAVRQAPLIIPLKATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSP



MMLV_RT

WNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTV



(WT)-60AA-

LDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFDEAL



CAS9(H840A)

HRDLADFRIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKA





QICQKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWI





PGFAEMAAPLYPLTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFV





DEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGK





LTMGQPLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQFGPWALNP





ATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTDGSSLLQEGQRKA





GAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTDSRYAFATAHI





HGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLSHHCPGHQKGHSAEARGNR





MADQAARKAAITETPDTSTLLIENSSPS

GGSSGGSSGSETVGTSESATVESAGSN







PYDVPDYAGSAAPAAKKKKLDGSGSGGSSGGS

DKKYSIGLDIGTNSVGWA





VITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTARR





RYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFG





NIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIE





GDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRL





ENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYD





DDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIK





RYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFY





KFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILR





RQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITP





WNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELT





KVKYVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIEC





FDSVEISGVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLF





EDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQS





GKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIA





NLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQ





KNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMY





VDQELDINRLSDYDVDAIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEE





VVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLV





ETRQITKHVAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQF





YKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKM





IAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIV





WDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARK





KDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSS





FEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKG





NELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQIS





EFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFK





YFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGD
SGGSKRTAD





GSEFEPKKKRKV (SEQ ID NO: 150)




KEY:



NUCLEAR LOCALIZATION SEQUENCE (NLS) TOP:(SEQ ID NO: 124),



BOTTOM: (SEQ ID NO: 133)



CAS9(H840A)(SEQ ID NO: 153)



AMINO ACID LINKER (SEQ ID NO: 175)



M-MLVREVERSE TRANSCRIPTASE (SEQ ID NO: 149)





PE FUSION

MKRTADGSEFESPKKKRKV
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFK



PROTEIN

VLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQ



CAS9(H840A)-

EIFSNEMAKYDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPT



FEN1-

IYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKL



MMLV_RT

FIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKN



D200N

GLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQ



T330P

YADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLK



L603W

ALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGT



T306K

EELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDN



W313F

REKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGAS





AQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRK





PAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDR





FNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKT





YAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDG





FANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGIL





QTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEE





GIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDY





DVDAIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQ





LLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQIL





DSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAH





DAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAK





YFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKV





LSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFD





SPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKG





YKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNELALPSKYVNFL





YLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANL





DKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTS





TKEVLDATLIHQSITGLYETRIDLSQLGGD

SGGSSGGSSGSETPGTSESATP







ESSGGSSGGSS
GIOGLAKLIADVAPSAIRENDIKSYFGRKVAIDASMSIYQFLIA




VRQGGDVLQNEEGETTSHLMGMFYRTIRMMENGIKPVYVFDGKPPQLKSGE



LAKRSERRAEAEKQLQQAQAAGAEQEVEKFTKRLVKVTKQHNDECKHLLSL



MGIPYLDAPSEAEASCAALVKAGKVYAAATEDMDCLTFGSPVLMRHLTASEA



KKLPIQEFHLSRILQELGLNQEQFVDLCILLGSDYCESIRGIGPKRAVDLIQKHK



SIEEIVRRLDPNKYPVPENWLHKEAHQLFLEPEVLDPESVELKWSEPNEEELIK



FMCGEKQFSEERIRSGVKRLSKSRQGSTQGRLDDFFKVTGSLSSAKRKEPEPK



GSTKKKAKTGAAGKFKRGKSGGSSGGSSGSETPGTSESATPESSGGSSGGSS




TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLKATST





PVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQ





DLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQPLFA





FEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRIQHPDLILLQYVD





DLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQKQVKYLGYLLKEGQRW





LTEARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPGFAEMAAPLYPLTKPGTLFN





WGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRR





PVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQ





PPDRWLSNARMTHYQALLLDTDRVQFGPWALNPATLLPLPEEGLQHNCLDILAEA





HGTRPDLTDQPLPDADHTWYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSA





QRAELIALTQALKMAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNK





DEILALLKALFLPKRLSHHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLL





IENSSP
SGGSKRTADGSEFEPKKKRKV (SEQIDNO: 154)




KEY:



NUCLEAR LOCALIZATION SEQUENCE (NLS) TOP:(SEQ ID NO: 124),



BOTTOM: (SEQ ID NO: 133)



CAS9(H840A) (SEQ ID NO: 157)



33-AMINO ACID LINKER 1 (SEQ ID NO: 127)



M-MLVREVERSE TRANSCRIPTASE (SEQ ID NO: 159)



33-AMINO ACID LINKER 2 (SEQ ID NO: 127)



FEN1 (SEQ ID NO: 161)









In other embodiments, the multi-flap prime editor fusion proteins can be based on SaCas9 or on SpCas9 nickases with altered PAM specificities, such as the following exemplary sequences:















SACAS9-M-MLV RT
MKRTADGSEFESPKKKRKVGKRNYILGLDIGITSVGYGIIDYETRDVID


PRIME EDITOR
AGVRLFKEANVENNEGRRSKRGARRLKRRRRHRIQRVKKLLFDYNLL



TDHSELSGINPYEARVKGLSQKLSEEEFSAALLHLAKRRGVHNVNEVE



EDTGNELSTKEQISRNSKALEEKYVAELQLERLKKDGEVRGSINRFKT



SDYVKEAKQLLKVQKAYHQLDQSFIDTYIDLLETRRTYYEGPGEGSPF



GWKDIKEWYEMLMGHCTYFPEELRSVKYAYNADLYNALNDLNNLVI



TRDENEKLEYYEKFQIIENVFKQKKKPTLKQIAKEILVNEEDIKGYRVT



STGKPEFTNLKVYHDIKDITARKEIIENAELLDQIAKILTIYQSSEDIQEE



LTNLNSELTQEEIEQISNLKGYTGTHNLSLKAINLILDELWHTNDNQIAI



FNRLKLVPKKVDLSQQKEIPTTLVDDFILSPVVKRSFIQSIKVINAIIKKY



GLPNDIIIELAREKNSKDAQKMINEMQKRNRQTNERIEEIIRTTGKENA



KYLIEKIKLHDMQEGKCLYSLEAIPLEDLLNNPFNYEVDHIIPRSVSFD



NSFNNKVLVKQEEASKKGNRTPFQYLSSSDSKISYETFKKHILNLAKG



KGRISKTKKEYLLEERDINRFSVQKDFINRNLVDTRYATRGLMNLLRS



YFRVNNLDVKVKSINGGFTSFLRRKWKFKKERNKGYKHHAEDALIIA



NADFIFKEWKKLDKAKKVMENQMFEEKQAESMPEIETEQEYKEIFITP



HQIKHIKDFKDYKYSHRVDKKPNRELINDTLYSTRKDDKGNTLIVNNL



NGLYDKDNDKLKKLINKSPEKLLMYHHDPQTYQKLKLIMEQYGDEK



NPLYKYYEETGNYLTKYSKKDNGPVIKKIKYYGNKLNAHLDITDDYP



NSRNKVVKLSLKPYRFDVYLDNGVYKFVTVKNLDVIKKENYYEVNS



KCYEEAKKLKKISNQAEFIASFYNNDLIKINGELYRVIGVNNDLLNRIE



VNMIDITYREYLENMNDKRPPRIIKTIASKTQSIKKYSTDILGNLYEVK



SKKHPQIIKKGSGGSSGGSSGSETPGTSESATPESSGGSSGGSSTLNIED



EYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLKA



TSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKK



PGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLD



LKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTL



FDEALHRDLADFRIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQT



LGNLGYRASAKKAQICQKQVKYLGYLLKEGQRWLTEARKETVMGQP



TPKTPRQLREFLGTAGFCRLWIPGFAEMAAPLYPLTKTGTLFNWGPDQ



QKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPW



RRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVIL



APHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQFGPVVALNPAT



LLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHTWYTDGSSLLQ



EGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGKK



LNVYTDSRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKALFLP



KRLSIIHCPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENS



SPSGGSKRTADGSEFEPKKKRKV (SEQ ID NO: 162)





SPCAS9(H840A)-
MKRTADGSEFESPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSK


VRQR-MALONEY
KFKVLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNR


MURINE
ICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVA


LEUKEMIA VIRUS
YHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLN


REVERSE
PDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE


TRANSCRIPTASE
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTY


PRIME EDITOR
DDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSAS



MIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGA



SQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHL



GELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWM



TRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSL



LYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVT



VKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFLD



NEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRY



TGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTF



KEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVM



GRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEH



PVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSF



LKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLI



TQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMN



TKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDA



YLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATA



KYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATV



RKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKK



YGGFVSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPI



DFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASARELQKGNEL



ALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQIS



EFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAA



FKYFDTTIDRKQYRSTKEVLDATLIHQSITGLYETRIDLSQLGGDSGGS



SGGSSGSETPGTSESATPESSGGSSGGSSTLNIEDEYRLHETSKEPDVSL



GSTWLSDFPQAWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPMSQE



ARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLRE



VNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQ



PLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRIQ



HPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQ



ICQKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKA



GFCRLFIPGFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAP



ALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPV



AAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPD



RWLSNARMTHYQALLLDTDRVQFGPVVALNPATLLPLPEEGLQHNCL



DILAEAHGTRPDLTDQPLPDADHTWYTDGSSLLQEGQRKAGAAVTTE



TEVIWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTDSRYAFAT



AHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRLSIIHCPGHQ



KGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSPSGGSKRTADG



SEFEPKKKRKV (SEQ ID NO: 163)





SPCAS9(H840A)-
MKRTADGSEFESPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSK


VRER-MALONEY
KFKVLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNR


MURINE
ICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVA


LEUKEMIA VIRUS
YHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLN


REVERSE
PDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLE


TRANSCRIPTASE
NLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTY


PRIME EDITOR
DDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSAS



MIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGA



SQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHL



GELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWM



TRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSL



LYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVT



VKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFLD



NEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRY



TGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTF



KEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVM



GRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEH



PVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSF



LKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLI



TQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMN



TKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDA



YLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATA



KYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATV



RKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKK



YGGFVSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPI



DFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASARELQKGNEL



ALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQIS



EFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAA



FKYFDTTIDRKEYRSTKEVLDATLIHQSITGLYETRIDLSQLGGDSGGSS



GGSSGSETPGTSESATPESSGGSSGGSSTLNIEDEYRLHETSKEPDVSLG



STWLSDFPQAWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPMSQEA



RLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLREV



NKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQP



LFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFRIQH



PDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQI



CQKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKA



GFCRLFIPGFAEMAAPLYPLTKPGTLFNWGPDQQKAYQEIKQALLTAP



ALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPV



AAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPD



RWLSNARMTHYQALLLDTDRVQFGPVVALNPATLLPLPEEGLQHNCL



DILAEAHGTRPDLTDQPLPDADHTWYTDGSSLLQEGQRKAGAAVTTE



TEVIWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTDSRYAFAT



AHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRLSIIHCPGHQ



KGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSPSGGSKRTADG



SEFEPKKKRKV (SEQ ID NO: 164)









In yet other embodiments, the multi-flap prime editor fusion proteins contemplated herein may include a Cas9 nickase (e.g., Cas9 (H840A)) fused to a truncated version of M-MLV reverse transcriptase. In this embodiment, the reverse transcriptase also contains 4 mutations (D200N, T306K, W313F, T330P; noting that the L603W mutation present in PE2 is no longer present due to the truncation). The DNA sequence encoding this truncated editor is 522 bp smaller than PE2, and therefore makes its potentially useful for applications where delivery of the DNA sequence is challenging due to its size (i.e. adeno-associated virus and lentivirus delivery). This embodiment is referred to as Cas9(H840A)-MMLV-RT(trunc) or “PE2-short” or “PE2-trunc” and has the following amino acid sequence:















CAS9(H840A)-

MKRTADGSEFESPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVP



MMLV-RT(TRUNC)
SKKFKYLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTARRRYT


OR PE2-SHORT
RRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIF



GNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKF



RGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAK



AILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSN



FDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAI



LLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPE



KYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELL



VKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKD



NREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEV



VDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTK



VKYVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFK



KIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILE



DIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGR



LSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKED



IQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMG



RHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILK



EHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAI



VPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQ



LLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKH



VAQILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKV



REINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVR



KMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIET



NGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKES



ILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGK



SKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLP



KYSLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYE



KLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDK



VLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKR



YTSTKEVLDATLIHQSITGLYETRIDLSQLGGDSGGSSGGSSGSETP




GTSESATPESSGGSSGGSSTLNIEDEYRLHETSKEPDVSLGSTWLSDFPQ




AWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPMSQEARLGIKPHIQRLL



DQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNP



YNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQ



LTWTRLPQGFKNSPTLFNEALHRDLADFRIQHPDLILLQYVDDLLLAATSE



LDCQQGTRALLQTLGNLGYRASAKKAQICQKQVKYLGYLLKEGQRWLTE



ARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPGFAEMAAPLYPLTKPGT



LFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVL



TQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQ



PLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQFGPWALNP



ATLLPLPEEGLQHNCLDNSRLINSGGSKRTADGSEFEPKKKRKV (SEO



ID NO: 765)



KEY:



NUCLEAR LOCALIZATION SEQUENCE (NLS) TOP:(SEQ ID NO: 124),



BOTTOM: (SEQ ID NO: 133)



CAS9(H840A) (SEQ ID NO: 157)



33-AMINO ACID LINKER 1 (SEQ ID NO: 127)



M-MLV TRUNCATED REVERSE TRANSCRIPTASE (SEQ ID NO: 766)









See FIG. 75, which provides a bar graph comparing the efficiency (i.e., “% of total sequencing reads with the specified eidt or indels”) of PE2, PE2-trunc, PE3, and PE3-trunc over different target sites in various cell lines. The data shows that the prime editors comprising the truncated RT variants were about as efficient as the prime editors comprising the non-trunctated RT proteins.


In various embodiments, the multi-flap prime editor fusion proteins contemplated herein may also include any variants of the above-disclosed sequences having an amino acid sequence that is at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to PE1, PE2, or any of the above indicated prime editor fusion sequences.


In certain embodiments, linkers may be used to link any of the peptides or peptide domains or moieties of the invention (e.g., a napDNAbp linked or fused to a reverse transcriptase).


[5] Linkers and Other Domains

The PE fusion proteins may comprise various other domains besides the napDNAbp (e.g., Cas9 domain) and the polymerase domain (e.g., RT domain) For example, in the case where the napDNAbp is a Cas9 and the polymerase is a RT, the PE fusion proteins may comprise one or more linkers that join the Cas9 domain with the RT domain. The linkers may also join other functional domains, such as nuclear localization sequences (NLS) or a FEN1 (or other flap endonuclease) to the PE fusion proteins or a domain thereof.


In addition, in embodiments involving trans prime editing, linkers may be used to link tPERT recruitment protein to a prime editor, e.g., between the tPERt recruitment protein and the napDNAbp. See e.g., FIG. 3G for an exemplary schematic of a trans prime editor (tPE) that includes linkers to separately fuse a polymerase domain and a recruiting protein domain to a napDNAbp.


A. Linkers


As defined above, the term “linker,” as used herein, refers to a chemical group or a molecule linking two molecules or moieties, e.g., a binding domain and a cleavage domain of a nuclease. In some embodiments, a linker joins a gRNA binding domain of an RNA-programmable nuclease and the catalytic domain of a polymerase (e.g., a reverse transcriptase). In some embodiments, a linker joins a dCas9 and reverse transcriptase. Typically, the linker is positioned between, or flanked by, two groups, molecules, or other moieties and connected to each one via a covalent bond, thus connecting the two. In some embodiments, the linker is an amino acid or a plurality of amino acids (e.g., a peptide or protein). In some embodiments, the linker is an organic molecule, group, polymer, or chemical moiety. In some embodiments, the linker is 5-100 amino acids in length, for example, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 30-35, 35-40, 40-45, 45-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-150, or 150-200 amino acids in length. Longer or shorter linkers are also contemplated.


The linker may be as simple as a covalent bond, or it may be a polymeric linker many atoms in length. In certain embodiments, the linker is a polpeptide or based on amino acids. In other embodiments, the linker is not peptide-like. In certain embodiments, the linker is a covalent bond (e.g., a carbon-carbon bond, disulfide bond, carbon-heteroatom bond, etc.). In certain embodiments, the linker is a carbon-nitrogen bond of an amide linkage. In certain embodiments, the linker is a cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic or heteroaliphatic linker. In certain embodiments, the linker is polymeric (e.g., polyethylene, polyethylene glycol, polyamide, polyester, etc.). In certain embodiments, the linker comprises a monomer, dimer, or polymer of aminoalkanoic acid. In certain embodiments, the linker comprises an aminoalkanoic acid (e.g., glycine, ethanoic acid, alanine, beta-alanine, 3-aminopropanoic acid, 4-aminobutanoic acid, 5-pentanoic acid, etc.). In certain embodiments, the linker comprises a monomer, dimer, or polymer of aminohexanoic acid (Ahx). In certain embodiments, the linker is based on a carbocyclic moiety (e.g., cyclopentane, cyclohexane). In other embodiments, the linker comprises a polyethylene glycol moiety (PEG). In other embodiments, the linker comprises amino acids. In certain embodiments, the linker comprises a peptide. In certain embodiments, the linker comprises an aryl or heteroaryl moiety. In certain embodiments, the linker is based on a phenyl ring. The linker may included funtionalized moieties to facilitate attachment of a nucleophile (e.g., thiol, amino) from the peptide to the linker. Any electrophile may be used as part of the linker. Exemplary electrophiles include, but are not limited to, activated esters, activated amides, Michael acceptors, alkyl halides, aryl halides, acyl halides, and isothiocyanates.


In some other embodiments, the linker comprises the amino acid sequence (GGGGS)n (SEQ ID NO: 165), (G)n (SEQ ID NO: 166), (EAAAK)n (SEQ ID NO: 167), (GGS)n (SEQ ID NO: 168), (SGGS))n (SEQ ID NO: 169), (XP)n (SEQ ID NO: 170), or any combination thereof, wherein n is independently an integer between 1 and 30, and wherein X is any amino acid. In some embodiments, the linker comprises the amino acid sequence (GGS)N (SEQ ID NO: 176), wherein n is 1, 3, or 7. In some embodiments, the linker comprises the amino acid sequence SGSETPGTSESATPES (SEQ ID NO: 171). In some embodiments, the linker comprises the amino acid sequence SGGSSGGSSGSETPGTSESATPESSGGSSGGS (SEQ ID NO: 172). In some embodiments, the linker comprises the amino acid sequence SGGSGGSGGS (SEQ ID NO: 173). In some embodiments, the linker comprises the amino acid sequence SGGS (SEQ ID NO: 174). In other embodiments, the linker comprises the amino acid sequence SGGSSGGSSGSETPGTSESATPESAGSYPYDVPDYAGSAAPAAKKKKLDGSGSGGSSGGS (SEQ ID NO: 175, 60AA).


In certain embodiments, linkers may be used to link any of the peptides or peptide domains or moieties of the invention (e.g., a napDNAbp linked or fused to a reverse transcriptase).


As defined above, the term “linker,” as used herein, refers to a chemical group or a molecule linking two molecules or moieties, e.g., a binding domain and a cleavage domain of a nuclease. In some embodiments, a linker joins a gRNA binding domain of an RNA-programmable nuclease and the catalytic domain of a recombinase. In some embodiments, a linker joins a dCas9 and reverse transcriptase. Typically, the linker is positioned between, or flanked by, two groups, molecules, or other moieties and connected to each one via a covalent bond, thus connecting the two. In some embodiments, the linker is an amino acid or a plurality of amino acids (e.g., a peptide or protein). In some embodiments, the linker is an organic molecule, group, polymer, or chemical moiety. In some embodiments, the linker is 5-100 amino acids in length, for example, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 30-35, 35-40, 40-45, 45-50, 50-60, 60-70, 70-80, 80-90, 90-100, 100-150, or 150-200 amino acids in length. Longer or shorter linkers are also contemplated.


The linker may be as simple as a covalent bond, or it may be a polymeric linker many atoms in length. In certain embodiments, the linker is a polypeptide or based on amino acids. In other embodiments, the linker is not peptide-like. In certain embodiments, the linker is a covalent bond (e.g., a carbon-carbon bond, disulfide bond, carbon-heteroatom bond, etc.). In certain embodiments, the linker is a carbon-nitrogen bond of an amide linkage. In certain embodiments, the linker is a cyclic or acyclic, substituted or unsubstituted, branched or unbranched aliphatic or heteroaliphatic linker. In certain embodiments, the linker is polymeric (e.g., polyethylene, polyethylene glycol, polyamide, polyester, etc.). In certain embodiments, the linker comprises a monomer, dimer, or polymer of aminoalkanoic acid. In certain embodiments, the linker comprises an aminoalkanoic acid (e.g., glycine, ethanoic acid, alanine, beta-alanine, 3-aminopropanoic acid, 4-aminobutanoic acid, 5-pentanoic acid, etc.). In certain embodiments, the linker comprises a monomer, dimer, or polymer of aminoHEXAnoic acid (Ahx). In certain embodiments, the linker is based on a carbocyclic moiety (e.g., cyclopentane, cycloHEXAne). In other embodiments, the linker comprises a polyethylene glycol moiety (PEG). In other embodiments, the linker comprises amino acids. In certain embodiments, the linker comprises a peptide. In certain embodiments, the linker comprises an aryl or heteroaryl moiety. In certain embodiments, the linker is based on a phenyl ring. The linker may included funtionalized moieties to facilitate attachment of a nucleophile (e.g., thiol, amino) from the peptide to the linker. Any electrophile may be used as part of the linker. Exemplary electrophiles include, but are not limited to, activated esters, activated amides, Michael acceptors, alkyl halides, aryl halides, acyl halides, and isothiocyanates.


In some other embodiments, the linker comprises the amino acid sequence (GGGGS)n (SEQ ID NO: 165), (G)n (SEQ ID NO: 166), (EAAAK)n (SEQ ID NO: 167), (GGS)n (SEQ ID NO: 168), (SGGS)n (SEQ ID NO: 169), (XP)n (SEQ ID NO: 170), or any combination thereof, wherein n is independently an integer between 1 and 30, and wherein X is any amino acid. In some embodiments, the linker comprises the amino acid sequence (GGS)N (SEQ ID NO: 176), wherein n is 1, 3, or 7. In some embodiments, the linker comprises the amino acid sequence SGSETPGTSESATPES (SEQ ID NO: 171). In some embodiments, the linker comprises the amino acid sequence SGGSSGGSSGSETPGTSESATPESSGGSSGGS (SEQ ID NO: 172). In some embodiments, the linker comprises the amino acid sequence SGGSGGSGGS (SEQ ID NO: 173). In some embodiments, the linker comprises the amino acid sequence SGGS (SEQ ID NO: 174).


In particular, the following linkers can be used in various embodiments to join prime editor domains with one another:









(SEQ ID NO: 767)


GGS;





(SEQ ID NO: 768)


GGSGGS;





(SEQ ID NO: 769)


GGSGGSGGS;





(SEQ ID NO: 127)


SGGSSGGSSGSETPGTSESATPESSGGSSGGSS;





(SEQ ID NO: 171)


SGSETPGTSESATPES;





(SEQ ID NO: 175)


SGGSSGGSSGSETPGTSESATPESAGSYPYDVPDYAGSAAPAAKKKKL





DGSGSGGSSGG S.






B. Nuclear Localization Sequence (NLS)


In various embodiments, the PE fusion proteins may comprise one or more nuclear localization sequences (NLS), which help promote translocation of a protein into the cell nucleus. Such sequences are well-known in the art and can include the following examples:














DESCRIPTION
SEQUENCE
SEQ ID NO:







NLS OF
PKKKRKV
SEQ ID NO: 16


SV40




LARGE T-




AG







NLS
MKRTADGSEFESPKKKRKV
SEQ ID NO: 124





NLS
MDSLLMNRRKFLYQFKNVRW
SEQ ID NO: 17



AKGRRETYLC






NLS OF
AVKRPAATKKAGQAKKKKLD
SEQ ID NO: 190


NUCLEOPL




ASMIN







NLS OF
MSRRRKANPTKLSENAKKLA
SEQ ID NO: 191


EGL-13
KEVEN






NLS OF C-
PAAKRVKLD
SEQ ID NO: 192


MYC







NLS OF
KLKIKRPVK
SEQ ID NO: 193


TUS-




PROTEIN







NLS OF
VSRKRPRP
SEQ ID NO: 194


POLYOMA




LARGE T-




AG







NLS OF
EGAPPAKRAR
SEQ ID NO: 195


HEPATITIS




D VIRUS




ANTIGEN







NLS OF
PPQPKKKPLDGE
SEQ ID NO: 196


MURINE




P53







NLS OF PE1
SGGSKRTADGSEFEPKKKRKV
SEQ ID NO: 133


AND PE2









The NLS examples above are non-limiting. The PE fusion proteins may comprise any known NLS sequence, including any of those described in Cokol et al., “Finding nuclear localization signals,” EMBO Rep., 2000, 1(5): 411-415 and Freitas et al., “Mechanisms and Signals for the Nuclear Import of Proteins,” Current Genomics, 2009, 10(8): 550-7, each of which are incorporated herein by reference.


In various embodiments, the multi-flap prime editors and constructs encoding the prime editors disclosed herein further comprise one or more, preferably, at least two nuclear localization signals. In certain embodiments, the multi-flap prime editors comprise at least two NLSs. In embodiments with at least two NLSs, the NLSs can be the same NLSs or they can be different NLSs. In addition, the NLSs may be expressed as part of a fusion protein with the remaining portions of the multi-flap prime editors. In some embodiments, one or more of the NLSs are bipartite NLSs (“bpNLS”). In certain embodiments, the disclosed fusion proteins comprise two bipartite NLSs. In some embodiments, the disclosed fusion proteins comprise more than two bipartite NLSs.


The location of the NLS fusion can be at the N-terminus, the C-terminus, or within a sequence of a prime editor (e.g., inserted between the encoded napDNAbp component (e.g., Cas9) and a polymerase domain (e.g., a reverse transcriptase domain).


The NLSs may be any known NLS sequence in the art. The NLSs may also be any future-discovered NLSs for nuclear localization. The NLSs also may be any naturally-occurring NLS, or any non-naturally occurring NLS (e.g., an NLS with one or more desired mutations).


The term “nuclear localization sequence” or “NLS” refers to an amino acid sequence that promotes import of a protein into the cell nucleus, for example, by nuclear transport. Nuclear localization sequences are known in the art and would be apparent to the skilled artisan. For example, NLS sequences are described in Plank et al., International PCT application PCT/EP2000/011690, filed Nov. 23, 2000, published as WO/2001/038547 on May 31, 2001, the contents of which are incorporated herein by reference. In some embodiments, an NLS comprises the amino acid sequence PKKKRKV (SEQ ID NO: 16), MDSLLMNRRKFLYQFKNVRWAKGRRETYLC (SEQ ID NO: 17), KRTADGSEFESPKKKRKV (SEQ ID NO: 3864), or KRTADGSEFEPKKKRKV (SEQ ID NO: 125). In other embodiments, NLS comprises the amino acid sequences NLSKRPAAIKKAGQAKKKK (SEQ ID NO: 136), PAAKRVKLD (SEQ ID NO: 192), RQRRNELKRSF (SEQ ID NO: 3934), NQSSNFGPMKGGNFGGRSSGPYGGGGQYFAKPRNQGGY (SEQ ID NO: 3935).


In one aspect of the disclosure, a multi-flap prime editor may be modified with one or more nuclear localization signals (NLS), preferably at least two NLSs. In certain embodiments, the multi-flap prime editors are modified with two or more NLSs. The disclosure contemplates the use of any nuclear localization signal known in the art at the time of the disclosure, or any nuclear localization signal that is identified or otherwise made available in the state of the art after the time of the instant filing. A representative nuclear localization signal is a peptide sequence that directs the protein to the nucleus of the cell in which the sequence is expressed. A nuclear localization signal is predominantly basic, can be positioned almost anywhere in a protein's amino acid sequence, generally comprises a short sequence of four amino acids (Autieri & Agrawal, (1998) J. Biol. Chem. 273: 14731-37, incorporated herein by reference) to eight amino acids, and is typically rich in lysine and arginine residues (Magin et al., (2000) Virology 274: 11-16, incorporated herein by reference). Nuclear localization signals often comprise proline residues. A variety of nuclear localization signals have been identified and have been used to effect transport of biological molecules from the cytoplasm to the nucleus of a cell. See, e.g., Tinland et al., (1992) Proc. Natl. Acad. Sci. U.S.A. 89:7442-46; Moede et al., (1999) FEBS Lett. 461:229-34, which is incorporated by reference. Translocation is currently thought to involve nuclear pore proteins.


Most NLSs can be classified in three general groups: (i) a monopartite NLS exemplified by the SV40 large T antigen NLS (PKKKRKV (SEQ ID NO: 16)); (ii) a bipartite motif consisting of two basic domains separated by a variable number of spacer amino acids and exemplified by the Xenopus nucleoplasmin NLS (KRXXXXXXXXXXKKKL (SEQ ID NO: 3936))); and (iii) noncanonical sequences such as M9 of the hnRNP Al protein, the influenza virus nucleoprotein NLS, and the yeast Gal4 protein NLS (Dingwall and Laskey 1991).


Nuclear localization signals appear at various points in the amino acid sequences of proteins. NLS's have been identified at the N-terminus, the C-terminus, and in the central region of proteins. Thus, the disclosure provides multi-flap prime editors that may be modified with one or more NLSs at the C-terminus, the N-terminus, as well as at an internal region of the multi-flap prime editor. The residues of a longer sequence that do not function as component NLS residues should be selected so as not to interfere, for example tonically or sterically, with the nuclear localization signal itself. Therefore, although there are no strict limits on the composition of an NLS-comprising sequence, in practice, such a sequence can be functionally limited in length and composition.


The present disclosure contemplates any suitable means by which to modify a multi-flap prime editor to include one or more NLSs. In one aspect, the multi-flap prime editors may be engineered to express a prime editor protein that is translationally fused at its N-terminus or its C-terminus (or both) to one or more NLSs, i.e., to form a prime editor-NLS fusion construct. In other embodiments, the prime editor-encoding nucleotide sequence may be genetically modified to incorporate a reading frame that encodes one or more NLSs in an internal region of the encoded prime editor. In addition, the NLSs may include various amino acid linkers or spacer regions encoded between the prime editor and the N-terminally, C-terminally, or internally-attached NLS amino acid sequence, e.g, and in the central region of proteins. Thus, the present disclosure also provides for nucleotide constructs, vectors, and host cells for expressing fusion proteins that comprise a prime editor and one or more NLSs.


The multi-flap prime editors described herein may also comprise nuclear localization signals which are linked to a prime editor through one or more linkers, e.g., and polymeric, amino acid, nucleic acid, polysaccharide, chemical, or nucleic acid linker element. The linkers within the contemplated scope of the disclosure are not intented to have any limitations and can be any suitable type of molecule (e.g., polymer, amino acid, polysaccharide, nucleic acid, lipid, or any synthetic chemical linker domain) and be joined to the prime editor by any suitable strategy that effectuates forming a bond (e.g., covalent linkage, hydrogen bonding) between the prime editor and the one or more NLSs.


C. Flap Endonucleases (e.g., FEN1)


In various embodiments, the PE fusion proteins may comprise one or more flap endonulceases (e.g., FEN1), which refers to an enzyme that catalyzes the removal of 5′ single strand DNA flaps. These are naturally occurring enzymes that process the removal of 5′ flaps formed during cellular processes, including DNA replication. The multi-flap prime editing methods herein described may utilize endogenously supplied flap endonucleases or those provided in trans to remove the 5′ flap of endogenouse DNA formed at the target site during multi-flap prime editing. Flap endonucleases are known in the art and can be found described in Patel et al., “Flap endonucleases pass 5′-flaps through a flexible arch using a disorder-thread-order mechanism to confer specificity for free 5′-ends,” Nucleic Acids Research, 2012, 40(10): 4507-4519 and Tsutakawa et al., “Human flap endonuclease structures, DNA double-base flipping, and a unified understanding of the FEN1 superfamily,” Cell, 2011, 145(2): 198-211 (each of which are incorporated herein by reference). An exemplary flap endonuclease is FEN1, which can be represented by the following amino acid sequence:














Description
Sequence
SEQ ID NO:







FEN1
MGIQGLAKLIADVAPSAIRENDIKSYF
SEQ ID NO:


Wild type
GRKVAIDASMSIYQFLIAVRQGGDVLQ
198


(wt)
NEEGETTSHLMGMFYRTIRMMENGIKP




VYVFDGKPPQLKSGELAKRSERRAEAE




KQLQQAQAAGAEQEVEKFTKRLVKVTK




QHNDECKHLLSLMGIPYLDAPSEAEAS




CAALVKAGKVYAAATEDMDCLTFGSPV




LMRHLTASEAKKLPIQEFHLSRILQEL




GLNQEQFVDLCILLGSDYCESIRGIGP




KRAVDLIQKHKSIEEIVRRLDPNKYPV




PENWLHKEAHQLFLEPEVLDPESVELK




WSEPNEEELIKFMCGEKQFSEERIRSG




VKRLSKSRQGSTQGRLDDFFKVTGSLS




SAKRKEPEPKGSTKKKAKTGAAGKFKR




GK









The flap endonucleases may also include any FEN1 variant, mutant, or other flap endonuclease ortholog, homolog, or variant. Non-limiting FEN1 variant examples are as follows:














Description
Sequence
SEQ ID NO:







FEN1
MGIQGLAKLIADVAPSAIRENDIKSYFGRKVAIDASMSIYQF
SEQ ID NO:


K168R
LIAVRQGGDVLQNEEGETTSHLMGMFYRTIRMMENGIKPV
199


(relative to
YVFDGKPPQLKSGELAKRSERRAEAEKQLQQAQAAGAEQE



FEN1 wt)
VEKFTKRLVKVTKQHNDECKHLLSLMGIPYLDAPSEAEASC




AALVRAGKVYAAATEDMDCLTFGSPVLMRHLTASEAKKLP




IQEFHLSRILQELGLNQEQFVDLCILLGSDYCESIRGIGPKRA




VDLIQKHKSIEEIVRRLDPNKYPVPENWLHKEAHQLFLEPEV




LDPESVELKWSEPNEEELIKFMCGEKQFSEERIRSGVKRLSK




SRQGSTQGRLDDFFKVTGSLSSAKRKEPEPKGSTKKKAKTG




AAGKFKRGK






FEN1
MGIQGLAKLIADVAPSAIRENDIKSYFGRKVAIDASMSIYQF
SEQ ID NO:


S187 A
LIAVRQGGDVLQNEEGETTSHLMGMFYRTIRMMENGIKPV
200


(relative to
YVFDGKPPQLKSGELAKRSERRAEAEKQLQQAQAAGAEQE



FEN1 wt)
VEKFTKRLVKVTKQHNDECKHLLSLMGIPYLDAPSEAEASC




AALVKAGKVYAAATEDMDCLTFGAPVLMRHLTASEAKKL




PIQEFHLSRILQELGLNQEQFVDLCILLGSDYCESIRGIGPKRA




VDLIQKHKSIEEIVRRLDPNKYPVPENWLHKEAHQLFLEPEV




LDPESVELKWSEPNEEELIKFMCGEKQFSEERIRSGVKRLSK




SRQGSTQGRLDDFFKVTGSLSSAKRKEPEPKGSTKKKAKTG




AAGKFKRGK






FEN1
MGIQGLAKLIADVAPSAIRENDIKSYFGRKVAIDASMSIYQF
SEQ ID NO:


K354R
LIAVRQGGDVLQNEEGETTSHLMGMFYRTIRMMENGIKPV
201


(relative to
YVFDGKPPQLKSGELAKRSERRAEAEKQLQQAQAAGAEQE



FEN1 wt)
VEKFTKRLVKVTKQHNDECKHLLSLMGIPYLDAPSEAEASC




AALVKAGKVYAAATEDMDCLTFGSPVLMRHLTASEAKKLP




IQEFHLSRILQELGLNQEQFVDLCILLGSDYCESIRGIGPKRA




VDLIQKHKSIEEIVRRLDPNKYPVPENWLHKEAHQLFLEPEV




LDPESVELKWSEPNEEELIKFMCGEKQFSEERIRSGVKRLSK




SRQGSTQGRLDDFFKVTGSLSSARRKEPEPKGSTKKKAKTG




AAGKFKRGK






GEN1
MGVNDLWQILEPVKQHIPLRNLGGKTIAVDLSLWVCEAQT
SEQ ID NO:



VKKMMGSVMKPHLRNLFFRISYLTQMDVKLVFVMEGEPPK
202



LKADVISKRNQSRYGSSGKSWSQKTGRSHFKSVLRECLHML




ECLGIPWVQAAGEAEAMCAYLNAGGHVDGCLTNDGDTFL




YGAQTVYRNFTMNTKDPHVDCYTMSSIKSKLGLDRDALVG




LAILLGCDYLPKGVPGVGKEQALKLIQILKGQSLLQRFNRW




NETSCNSSPQLLVTKKLAHCSVCSHPGSPKDHERNGCRLCK




SDKYCEPHDYEYCCPCEWHRTEHDRQLSEVENNIKKKACC




CEGFPFHEVIQEFLLNKDKLVKVIRYQRPDLLLFQRFTLEKM




EWPNHYACEKLLVLLTHYDMIERKLGSRNSNQLQPIRIVKT




RIRNGVHCFEIEWEKPEHYAMEDKQHGEFALLTIEEESLFEA




AYPEIVAVYQKQKLEIKGKKQKRIKPKENNLPEPDEVMSFQ




SHMTLKPTCEIFHKQNSKLNSGISPDPTLPQESISASLNSLLLP




KNTPCLNAQEQFMSSLRPLAIQQIKAVSKSLISESSQPNTSSH




NISVIADLHLSTIDWEGTSFSNSPAIQRNTFSHDLKSEVESELS




AIPDGFEN1PEQLSCESERYTANIKKVLDEDSDGISPEEHLLS




GITDLCLQDLPLKERIFTKLSYPQDNLQPDVNLKTLSILSVKE




SCIANSGSDCTSHLSKDLPGIPLQNESRDSKILKGDQLLQEDY




KVNTSVPYSVSNTVVKTCNVRPPNTALDHSRKVDMQTTRKI




LMKKSVCLDRHSSDEQSAPVFGKAKYTTQRMKHSSQKHNS




SHFKESGHNKLSSPKIHIKETEQCVRSYETAENEESCFPDSTK




SSLSSLQCHKKENNSGTCLDSPLPLRQRLKLRFQST






ERCC5
MGVQGLWKLLECSGRQVSPEALEGKILAVDISIWLNQALKG
SEQ ID NO:



VRDRHGNSIENPHLLTLFHRLCKLLFFRIRPIFVFDGDAPLLK
203



KQTLVKRRQRKDLASSDSRKTTEKLLKTFLKRQAIKTAFRS




KRDEALPSLTQVRRENDLYVLPPLQEEEKHSSEEEDEKEWQ




ERMNQKQALQEEFFHNPQAIDIESEDFSSLPPEVKHEILTDM




KEFTKRRRTLFEAMPEESDDFSQYQLKGLLKKNYLNQHIEH




VQKEMNQQHSGHIRRQYEDEGGFLKEVESRRVVSEDTSHYI




LIKGIQAKTVAEVDSESLPSSSKMHGMSFDVKSSPCEKLKTE




KEPDATPPSPRTLLAMQAALLGSSSEEELESENRRQARGRN




APAAVDEGSISPRTLSAIKRALDDDEDVKVCAGDDVQTGGP




GAEEMRINSSTENSDEGLKVRDGKGIPFTATLASSSVNSAEE




HVASTNEGREPTDSVPKEQMSLVHVGTEAFPISDESMIKDR




KDRLPLESAVVRHSDAPGLPNGRELTPASPTCTNSVSKNETH




AEVLEQQNELCPYESKFDSSLLSSDDETKCKPNSASEVIGPV




SLQETSSIVSVPSEAVDNVENVVSFNAKEHENFLETIQEQQT




TESAGQDLISIPKAVEPMEIDSEESESDGSFIEVQSVISDEELQ




AEFPETSKPPSEQGEEELVGTREGEAPAESESLLRDNSERDD




VDGEPQEAEKDAEDSLHEWQDINLEELETLESNLLAQQNSL




KAQKQQQERIAATVTGQMFLESQELLRLFGIPYIQAPMEAE




AQCAILDLTDQTSGTITDDSDIWLFGARHVYRNFFNKNKFV




EYYQYVDFHNQLGLDRNKLINLAYLLGSDYTEGIPTVGCVT




AMEILNEFPGHGLEPLLKFSEWWHEAQKNPKIRPNPHDTKV




KKKLRTLQLTPGFPNPAVAEAYLKPVVDDSKGSFLWGKPD




LDKIREFCQRYFGWNRTKTDESLFPVLKQLDAQQTQLRIDSF




FRLAQQEKEDAKRIKSQRLNRAVTCMLRKEKEAAASEIEAV




SVAMEKEFELLDKAKRKTQKRGITNTLEESSSLKRKRLSDS




KRKNTCGGFLGETCLSESSDGSSSEDAESSSLMNVQRRTAA




KEPKTSASDSQNSVKEAPVKNGGATTSSSSDSDDDGGKEK




MVLVTARSVFGKKRRKLRRARGRKRKT









In various embodiments, the multi-flap prime editor fusion proteins contemplated herein may include any flap endonulcease variant of the above-disclosed sequences having an amino acid sequence that is at least about 70% identical, at least about 80% identical, at least about 90% identical, at least about 95% identical, at least about 96% identical, at least about 97% identical, at least about 98% identical, at least about 99% identical, at least about 99.5% identical, or at least about 99.9% identical to any of the above sequences.


Other endonucleases that may be utilized by the instant methods to facilitate removal of the 5′ end single strand DNA flap include, but are not limited to (1) trex 2, (2) exol endonuclease (e.g., Keijzers et al., Biosci Rep. 2015, 35(3): e00206)


Trex 2


3′ three prime repair exonuclease 2 (TREX2)—human









Accession No. NM_080701


(SEQ ID NO: 3865)


MSEAPRAETFVFLDLEATGLPSVEPEIAELSLFAVHRSSLENPEHDESG





ALVLPRVLDKLTLCMCPERPFTAKASEITGLSSEGLARCRKAGFDGAVV





RTLQAFLSRQAGPICLVAHNGFDYDFPLLCAELRRLGARLPRDTVCLDT





LPALRGLDRAHSHGTRARGRQGYSLGSLFHRYFRAEPSAAHSAEGDVHT





LLLIFLHRAAELLAWADEQARGWAHIEPMYLPPDDPSLEA.






3′ three prime repair exonuclease 2 (TREX2)—mouse









Accession No. NM_011907


(SEQ ID NO: 3866)


MSEPPRAETFVFLDLEATGLPNMDPEIAEISLFAVHRSSLENPERDDSG





SLVLPRVLDKLTLCMCPERPFTAKASEITGLSSESLMHCGKAGFNGAVV





RTLQGFLSRQEGPICLVAHNGFDYDFPLLCTELQRLGAHLPQDTVCLDT





LPALRGLDRAHSHGTRAQGRKSYSLASLFHRYFQAEPSAAHSAEGDVHT





LLLIFLHRAPELLAWADEQARSWAHIEPMYVPPDGPSLEA.






3′ three prime repair exonuclease 2 (TREX2)—rat









Accession No. NM_001107580


(SEQ ID NO: 3867)


MSEPLRAETFVFLDLEATGLPNMDPEIAEISLFAVHRSSLENPERDDSG





SLVLPRVLDKLTLCMCPERPFTAKASEITGLSSEGLMNCRKAAFNDAVV





RTLQGFLSRQEGPICLVAHNGFDYDFPLLCTELQRLGAHLPRDTVCLDT





LPALRGLDRVHSHGTRAQGRKSYSLASLFHRYFQAEPSAAHSAEGDVNT





LLLIFLHRAPELLAWADEQARSWAHIEPMYVPPDGPSLEA.







ExoI


Human exonuclease 1 (EXO1) has been implicated in many different DNA metabolic processes, including DNA mismatch repair (MMR), micro-mediated end-joining, homologous recombination (HR), and replication. Human EXO1 belongs to a family of eukaryotic nucleases, Rad2/XPG, which also include FEN1 and GEN1. The Rad2/XPG family is conserved in the nuclease domain through species from phage to human. The EXO1 gene product exhibits both 5′ exonuclease and 5′ flap activity. Additionally, EXO1 contains an intrinsic 5′ RNase H activity. Human EXO1 has a high affinity for processing double stranded DNA (dsDNA), nicks, gaps, pseudo Y structures and can resolve Holliday junctions using its inherit flap activity. Human EXO1 is implicated in MMR and contain conserved binding domains interacting directly with MLH1 and MSH2. EXO1 nucleolytic activity is positively stimulated by PCNA, MutSα (MSH2/MSH6 complex), 14-3-3, MRN and 9-1-1 complex.


exonuclease 1 (EXO1) Accession No. NM_003686 (Homo sapiens exonuclease 1 (EXO1), transcript variant 3)—isoform A









(SEQ ID NO: 3868)


MGIQGLLQFIKEASEPIHVRKYKGQVVAVDTYCWLHKGAIACAEKLAKG





EPTDRYVGFCMKFVNMLLSHGIKPILVFDGCTLPSKKEVERSRRERRQA





NLLKGKQLLREGKVSEARECFTRSINITHAMAHKVIKAARSQGVDCLVA





PYEADAQLAYLNKAGIVQAIITEDSDLLAFGCKKVILKMDQFGNGLEID





QARLGMCRQLGDVFTEEKFRYMCILSGCDYLSSLRGIGLAKACKVLRLA





NNPDIVKVIKKIGHYLKMNITVPEDYINGFIRANNTFLYQLVFDPIKRK





LIPLNAYEDDVDPETLSYAGQYVDDSIALQIALGNKDINTFEQIDDYNP





DTAMPAHSRSHSWDDKTCQKSANVSSIWHRNYSPRPESGTVSDAPQLKE





NPSTVGVERVISTKGLNLPRKSSIVKRPRSAELSEDDLLSQYSLSFTKK





TKKNSSEGNKSLSFSEVFVPDLVNGPTNKKSVSTPPRTRNKFATFLQRK





NEESGAVVVPGTRSRFFCSSDSTDCVSNKVSIQPLDETAVTDKENNLHE





SEYGDQEGKRLVDTDVARNSSDDIPNNHIPGDHIPDKATVFTDEESYSF





ESSKFTRTISPPTLGTLRSCFSWSGGLGDFSRTPSPSPSTALQQFRRKS





DSPTSLPENNMSDVSQLKSEESSDDESHPLREEACSSQSQESGEFSLQS





SNASKLSQCSSKDSDSEESDCNIKLLDSQSDQTSKLRLSHFSKKDTPLR





NKVPGLYKSSSADSLSTTKIKPLGPARASGLSKKPASIQKRKHHNAENK





PGLQIKLNELWKNFGFKKF.






exonuclease 1 (EXO1) Accession No. NM_006027 (Homo sapiens exonuclease 1 (EXO1), transcript variant 3)—isoform B









(SEQ ID NO: 3869)


MGIQGLLQFIKEASEPIHVRKYKGQVVAVDTYCWLHKGAIACAEKLAKG





EPTDRYVGFCMKFVNMLLSHGIKPILVFDGCTLPSKKEVERSRRERRQA





NLLKGKQLLREGKVSEARECFTRSINITHAMAHKVIKAARSQGVDCLVA





PYEADAQLAYLNKAGIVQAIITEDSDLLAFGCKKVILKMDQFGNGLEID





QARLGMCRQLGDVFTEEKFRYMCILSGCDYLSSLRGIGLAKACKVLRLA





NNPDIVKVIKKIGHYLKMNITVPEDYINGFIRANNTFLYQLVFDPIKRK





LIPLNAYEDDVDPETLSYAGQYVDDSIALQIALGNKDINTFEQIDDYNP





DTAMPAHSRSHSWDDKTCQKSANVSSIWHRNYSPRPESGTVSDAPQLKE





NPSTVGVERVISTKGLNLPRKSSIVKRPRSAELSEDDLLSQYSLSFTKK





TKKNSSEGNKSLSFSEVFVPDLVNGPTNKKSVSTPPRTRNKFATFLQRK





NEESGAVVVPGTRSRFFCSSDSTDCVSNKVSIQPLDETAVTDKENNLHE





SEYGDQEGKRLVDTDVARNSSDDIPNNHIPGDHIPDKATVFTDEESYSF





ESSKFTRTISPPTLGTLRSCFSWSGGLGDFSRTPSPSPSTALQQFRRKS





DSPTSLPENNMSDVSQLKSEESSDDESHPLREEACSSQSQESGEFSLQS





SNASKLSQCSSKDSDSEESDCNIKLLDSQSDQTSKLRLSHFSKKDTPLR





NKVPGLYKSSSADSLSTTKIKPLGPARASGLSKKPASIQKRKHHNAENK





PGLQIKLNELWKNFGFKKDSEKLPPCKKPLSPVRDNIQLTPEAEEDIFN





KPECGRVQRAIFQ.






exonuclease 1 (EXO1) Accession No. NM_001319224 (Homo sapiens exonuclease 1 (EXO1), transcript variant 4)—isoform C









(SEQ ID NO: 3870)


MGIQGLLQFIKEASEPIHVRKYKGQVVAVDTYCWLHKGAIACAEKLAKG





EPTDRYVGFCMKFVNMLLSHGIKPILVFDGCTLPSKKEVERSRRERRQA





NLLKGKQLLREGKVSEARECFTRSINITHAMAHKVIKAARSQGVDCLVA





PYEADAQLAYLNKAGIVQAIITEDSDLLAFGCKKVILKMDQFGNGLEID





QARLGMCRQLGDVFTEEKFRYMCILSGCDYLSSLRGIGLAKACKVLRLA





NNPDIVKVIKKIGHYLKMNITVPEDYINGFIRANNTFLYQLVFDPIKRK





LIPLNAYEDDVDPETLSYAGQYVDDSIALQIALGNKDINTFEQIDDYNP





DTAMPAHSRSHSWDDKTCQKSANVSSIWHRNYSPRPESGTVSDAPQLKE





NPSTVGVERVISTKGLNLPRKSSIVKRPRSELSEDDLLSQYSLSFTKKT





KKNSSEGNKSLSFSEVFVPDLVNGPTNKKSVSTPPRTRNKFATFLQRKN





EESGAVVVPGTRSRFFCSSDSTDCVSNKVSIQPLDETAVTDKENNLHES





EYGDQEGKRLVDTDVARNSSDDIPNNHIPGDHIPDKATVFTDEESYSFE





SSKFTRTISPPTLGTLRSCFSWSGGLGDFSRTPSPSPSTALQQFRRKSD





SPTSLPENNMSDVSQLKSEESSDDESHPLREEACSSQSQESGEFSLQSS





NASKLSQCSSKDSDSEESDCNIKLLDSQSDQTSKLRLSHFSKKDTPLRN





KVPGLYKSSSADSLSTTKIKPLGPARASGLSKKPASIQKRKHHNAENKP





GLQIKLNELWKNFGFKKDSEKLPPCKKPLSPVRDNIQLTPEAEEDIFNK





PECGRVQRAIFQ.






D. Inteins and Split-Inteins


It will be understood that in some embodiments (e.g., delivery of a multi-flap prime editor in vivo using AAV particles), it may be advantageous to split a polypeptide (e.g., a deaminase or a napDNAbp) or a fusion protein (e.g., a multi-flap prime editor) into an N-terminal half and a C-terminal half, delivery them separately, and then allow their colocalization to reform the complete protein (or fusion protein as the case may be) within the cell. Separate halves of a protein or a fusion protein may each comprise a split-intein tag to facilitate the reformation of the complete protein or fusion protein by the mechanism of protein trans splicing.


Protein trans-splicing, catalyzed by split inteins, provides an entirely enzymatic method for protein ligation. A split-intein is essentially a contiguous intein (e g a mini-intein) split into two pieces named N-intein and C-intein, respectively. The N-intein and C-intein of a split intein can associate non-covalently to form an active intein and catalyze the splicing reaction essentially in same way as a contiguous intein does. Split inteins have been found in nature and also engineered in laboratories. As used herein, the term “split intein” refers to any intein in which one or more peptide bond breaks exists between the N-terminal and C-terminal amino acid sequences such that the N-terminal and C-terminal sequences become separate molecules that can non-covalently reassociate, or reconstitute, into an intein that is functional for trans-splicing reactions. Any catalytically active intein, or fragment thereof, may be used to derive a split intein for use in the methods of the invention. For example, in one aspect the split intein may be derived from a eukaryotic intein. In another aspect, the split intein may be derived from a bacterial intein. In another aspect, the split intein may be derived from an archaeal intein. Preferably, the split intein so-derived will possess only the amino acid sequences essential for catalyzing trans-splicing reactions.


As used herein, the “N-terminal split intein (In)” refers to any intein sequence that comprises an N-terminal amino acid sequence that is functional for trans-splicing reactions. An In thus also comprises a sequence that is spliced out when trans-splicing occurs. An In can comprise a sequence that is a modification of the N-terminal portion of a naturally occurring intein sequence. For example, an In can comprise additional amino acid residues and/or mutated residues so long as the inclusion of such additional and/or mutated residues does not render the In non-functional in trans-splicing. Preferably, the inclusion of the additional and/or mutated residues improves or enhances the trans-splicing activity of the In.


As used herein, the “C-terminal split intein (k)” refers to any intein sequence that comprises a C-terminal amino acid sequence that is functional for trans-splicing reactions. In one aspect, the Ic comprises 4 to 7 contiguous amino acid residues, at least 4 amino acids of which are from the last β-strand of the intein from which it was derived. An Ic thus also comprises a sequence that is spliced out when trans-splicing occurs. An Ic can comprise a sequence that is a modification of the C-terminal portion of a naturally occurring intein sequence. For example, an Ic can comprise additional amino acid residues and/or mutated residues so long as the inclusion of such additional and/or mutated residues does not render the In non-functional in trans-splicing. Preferably, the inclusion of the additional and/or mutated residues improves or enhances the trans-splicing activity of the Ic.


In some embodiments of the invention, a peptide linked to an Ic or an In can comprise an additional chemical moiety including, among others, fluorescence groups, biotin, polyethylene glycol (PEG), amino acid analogs, unnatural amino acids, phosphate groups, glycosyl groups, radioisotope labels, and pharmaceutical molecules. In other embodiments, a peptide linked to an Ic can comprise one or more chemically reactive groups including, among others, ketone, aldehyde, Cys residues and Lys residues. The N-intein and C-intein of a split intein can associate non-covalently to form an active intein and catalyze the splicing reaction when an “intein-splicing polypeptide (ISP)” is present. As used herein, “intein-splicing polypeptide (ISP)” refers to the portion of the amino acid sequence of a split intein that remains when the Ic, In, or both, are removed from the split intein. In certain embodiments, the In comprises the ISP. In another embodiment, the Ic comprises the ISP. In yet another embodiment, the ISP is a separate peptide that is not covalently linked to In nor to Ic.


Split inteins may be created from contiguous inteins by engineering one or more split sites in the unstructured loop or intervening amino acid sequence between the −12 conserved beta-strands found in the structure of mini-inteins. Some flexibility in the position of the split site within regions between the beta-strands may exist, provided that creation of the split will not disrupt the structure of the intein, the structured beta-strands in particular, to a sufficient degree that protein splicing activity is lost.


In protein trans-splicing, one precursor protein consists of an N-extein part followed by the N-intein, another precursor protein consists of the C-intein followed by a C-extein part, and a trans-splicing reaction (catalyzed by the N- and C-inteins together) excises the two intein sequences and links the two extein sequences with a peptide bond. Protein trans-splicing, being an enzymatic reaction, can work with very low (e.g. micromolar) concentrations of proteins and can be carried out under physiological conditions.


Exemplary sequences are as follows:













NAME
SEQUENCE OF LIGAND-DEPENDENT INTEIN







2-4 INTEIN:
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGAIVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 8)





3-2 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAVAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGAIVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYTNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 9)





30R3-1 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPIPYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEGLRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 10)





30R3-2 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 11)





30R3-3 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPIPYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 12)





37R3-1 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYNPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLERAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEGLRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC ((SEQ ID NO: 13)





37R3-2 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGAIVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLERAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEGLRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 14)





37R3-3 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAVAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLERAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 15)









Although inteins are most frequently found as a contiguous domain, some exist in a naturally split form. In this case, the two fragments are expressed as separate polypeptides and must associate before splicing takes place, so-called protein trans-splicing.


An exemplary split intein is the Ssp DnaE intein, which comprises two subunits, namely, DnaE-N and DnaE-C. The two different subunits are encoded by separate genes, namely dnaE-n and dnaE-c, which encode the DnaE-N and DnaE-C subunits, respectively. DnaE is a naturally occurring split intein in Synechocytis sp. PCC6803 and is capable of directing trans-splicing of two separate proteins, each comprising a fusion with either DnaE-N or DnaE-C.


Additional naturally occurring or engineered split-intein sequences are known in the or can be made from whole-intein sequences described herein or those available in the art. Examples of split-intein sequences can be found in Stevens et al., “A promiscuous split intein with expanded protein engineering applications,” PNAS, 2017, Vol. 114: 8538-8543; Iwai et al., “Highly efficient protein trans-splicing by a naturally split DnaE intein from Nostc punctiforme, FEBS Lett, 580: 1853-1858, each of which are incorporated herein by reference. Additional split intein sequences can be found, for example, in WO 2013/045632, WO 2014/055782, WO 2016/069774, and EP2877490, the contents each of which are incorporated herein by reference.


In addition, protein splicing in trans has been described in vivo and in vitro (Shingledecker, et al., Gene 207:187 (1998), Southworth, et al., EMBO J. 17:918 (1998); Mills, et al., Proc. Natl. Acad. Sci. USA, 95:3543-3548 (1998); Lew, et al., J. Biol. Chem., 273:15887-15890 (1998); Wu, et al., Biochim. Biophys. Acta 35732:1 (1998b), Yamazaki, et al., J. Am. Chem. Soc. 120:5591 (1998), Evans, et al., J. Biol. Chem. 275:9091 (2000); Otomo, et al., Biochemistry 38:16040-16044 (1999); Otomo, et al., J. Biolmol. NMR 14:105-114 (1999); Scott, et al., Proc. Natl. Acad. Sci. USA 96:13638-13643 (1999)) and provides the opportunity to express a protein as to two inactive fragments that subsequently undergo ligation to form a functional product, e.g., as shown in FIGS. 66 and 67 with regard to the formation of a complete PE fusion protein from two separately-expressed halves.


E. RNA-Protein Interaction Domain


In various embodiments, two separate protein domains (e.g., a Cas9 domain and a polymerase domain) may be colocalized to one another to form a functional complex (akin to the function of a fusion protein comprising the two separate protein domains) by using an “RNA-protein recruitment system,” such as the “MS2 tagging technique.” Such systems generally tag one protein domain with an “RNA-protein interaction domain” (aka “RNA-protein recruitment domain”) and the other with an “RNA-binding protein” that specifically recognizes and binds to the RNA-protein interaction domain, e.g., a specific hairpin structure. These types of systems can be leveraged to colocalize the domains of a multi-flap prime editor, as well as to recruitment additional functionalities to a multi-flap prime editor, such as a UGI domain. In one example, the MS2 tagging technique is based on the natural interaction of the MS2 bacteriophage coat protein (“MCP” or “MS2cp”) with a stem-loop or hairpin structure present in the genome of the phage, i.e., the “MS2 hairpin.” In the case of the MS2 hairpin, it is recognized and bound by the MS2 bacteriophage coat protein (MCP). Thus, in one exemplarily scenario a deaminase-MS2 fusion can recruit a Cas9-MCP fusion.


A review of other modular RNA-protein interaction domains are described in the art, for example, in Johansson et al., “RNA recognition by the MS2 phage coat protein,” Sem Virol., 1997, Vol. 8(3): 176-185; Delebecque et al., “Organization of intracellular reactions with rationally designed RNA assemblies,” Science, 2011, Vol. 333: 470-474; Mali et al., “Cas9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering,” Nat. Biotechnol., 2013, Vol. 31: 833-838; and Zalatan et al., “Engineering complex synthetic transcriptional programs with CRISPR RNA scaffolds,” Cell, 2015, Vol. 160: 339-350, each of which are incorporated herein by reference in their entireties. Other systems include the PP7 hairpin, which specifically recruits the PCP protein, and the “coin” hairpin, which specifically recruits the Com protein. See Zalatan et al.


The nucleotide sequence of the MS2 hairpin (or equivalently referred to as the “MS2 aptamer”) is: GCCAACATGAGGATCACCCATGTCTGCAGGGCC (SEQ ID NO: 763).


The amino acid sequence of the MCP or MS2cp is:









(SEQ ID NO: 764)


GSASNFTQFVLVDNGGTGDVTVAPSNFANGVAEWISSNSRSQAYKVTCS


VRQSSAQNRKYTIKVEVPKVATQTVGGEELPVAGWRSYLNMELTIPIFA


TNSDCELIVKAMQGLLKDGNPIPSAIAANSGIY.






F. UGI Domain


In other embodiments, the multi-flap prime editors described herein may comprise one or more uracil glycosylase inhibitor domains. The term “uracil glycosylase inhibitor (UGI)” or “UGI domain,” as used herein, refers to a protein that is capable of inhibiting a uracil-DNA glycosylase base-excision repair enzyme. In some embodiments, a UGI domain comprises a wild-type UGI or a UGI as set forth in SEQ ID NO: 3873. In some embodiments, the UGI proteins provided herein include fragments of UGI and proteins homologous to a UGI or a UGI fragment. For example, in some embodiments, a UGI domain comprises a fragment of the amino acid sequence set forth in SEQ ID NO: 3873. In some embodiments, a UGI fragment comprises an amino acid sequence that comprises at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 99.5% of the amino acid sequence as set forth in SEQ ID NO: 3873. In some embodiments, a UGI comprises an amino acid sequence homologous to the amino acid sequence set forth in SEQ ID NO: 3873, or an amino acid sequence homologous to a fragment of the amino acid sequence set forth in SEQ ID NO: 3873. In some embodiments, proteins comprising UGI or fragments of UGI or homologs of UGI or UGI fragments are referred to as “UGI variants.” A UGI variant shares homology to UGI, or a fragment thereof. For example a UGI variant is at least 70% identical, at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, at least 99% identical, at least 99.5% identical, or at least 99.9% identical to a wild type UGI or a UGI as set forth in SEQ ID NO: 3873. In some embodiments, the UGI variant comprises a fragment of UGI, such that the fragment is at least 70% identical, at least 80% identical, at least 90% identical, at least 95% identical, at least 96% identical, at least 97% identical, at least 98% identical, at least 99% identical, at least 99.5% identical, or at least 99.9% to the corresponding fragment of wild-type UGI or a UGI as set forth in SEQ ID NO: 3873. In some embodiments, the UGI comprises the following amino acid sequence:


Uracil-DNA glycosylase inhibitor:









>sp|P14739|UNGI_BPPB2


(SEQ ID NO: 3873)


MTNLSDIIEKETGKQLVIQESILMLPEEVEEVIGNKPESDILVHTAYDE


STDENVMLLTSDAPEYKPWALVIQDSNGENKIKML.






The multi-flap prime editors described herein may comprise more than one UGI domain, which may be separated by one or more linkers as described herein.


G. Additional PE Elements


In certain embodiments, the multi-flap prime editors described herein may comprise an inhibitor of base repair. The term “inhibitor of base repair” or “IBR” refers to a protein that is capable in inhibiting the activity of a nucleic acid repair enzyme, for example a base excision repair enzyme. In some embodiments, the IBR is an inhibitor of OGG base excision repair. In some embodiments, the IBR is an inhibitor of base excision repair (“iBER”). Exemplary inhibitors of base excision repair include inhibitors of APE1, Endo III, Endo IV, Endo V, Endo VIII, Fpg, hOGG1, hNEIL1, T7 Endol, T4PDG, UDG, hSMUG1, and hAAG. In some embodiments, the IBR is an inhibitor of Endo V or hAAG. In some embodiments, the IBR is an iBER that may be a catalytically inactive glycosylase or catalytically inactive dioxygenase or a small molecule or peptide inhibitor of an oxidase, or variants thereof. In some embodiments, the IBR is an iBER that may be a TDG inhibitor, MBD4 inhibitor or an inhibitor of an AlkBH enzyme. In some embodiments, the IBR is an iBER that comprises a catalytically inactive TDG or catalytically inactive MBD4. An exemplary catalytically inactive TDG is an N140A mutant of SEQ ID NO: 3872 (human TDG).


Some exemplary glycosylases are provided below. The catalytically inactivated variants of any of these glycosylase domains are iBERs that may be fused to the napDNAbp or polymerase domain of the multi-flap prime editors provided in this disclosure.









OGG (human)


(SEQ ID NO: 3937)


MPARALLPRRMGHRTLASTPALWASIPCPRSELRLDLVLPSGQSFRWRE





QSPAHWSGVLADQVWTLTQTEEQLHCTVYRGDKSQASRPTPDELEAVRK





YFQLDVTLAQLYHHWGSVDSHFQEVAQKFQGVRLLRQDPIECLFSFICS





SNNNIARITGMVERLCQAFGPRLIQLDDVTYHGFPSLQALAGPEVEAHL





RKLGLGYRARYVSASARAILEEQGGLAWLQQLRESSYEEAHKALCILPG





VGTKVADCICLMALDKPQAVPVDVHMWHIAQRDYSWHPTTSQAKGPSPQ 





TNKELGNFFRSLWGPYAGWAQAVLFSADLRQSRHAQEPPAKRRKGSKGP





EG





MPG (human)


(SEQ ID NO: 3938)


MVTPALQMKKPKQFCRRMGQKKQRPARAGQPHSSSDAAQAPAEQPHSSS





DAAQAPCPRERCLGPPTTPGPYRSIYFSSPKGHLTRLGLEFFDQPAVPL





ARAFLGQVLVRRLPNGTELRGRIVETEAYLGPEDEAAHSRGGRQTPRNR





GMFMKPGTLYVYIIYGMYFCMNISSQGDGACVLLRALEPLEGLETMRQL





RSTLRKGTASRVLKDRELCSGPSKLCQALAINKSFDQRDLAQDEAVWLE





RGPLEPSEPAVVAAARVGVGHAGEWARKPLRFYVRGSPWVSVVDRVAEQ





DTQA





MBD4 (human)


(SEQ ID NO: 3871)


MGTTGLESLSLGDRGAAPTVTSSERLVPDPPNDLRKEDVAMELERVGED





EEQMMIKRSSECNPLLQEPIASAQFGATAGTECRKSVPCGWERVVKQRL





FGKTAGRFDVYFISPQGLKFRSKSSLANYLHKNGETSLKPEDFDFTVLS





KRGIKSRYKDCSMAALTSHLQNQSNNSNWNLRTRSKCKKDVFMPPSSSS





ELQESRGLSNFTSTHLLLKEDEGVDDVNFRKVRKPKGKVTILKGIPIKK





TKKGCRKSCSGFVQSDSKRESVCNKADAESEPVAQKSQLDRTVCISDAG





ACGETLSVTSEENSLVKKKERSLSSGSNFCSEQKTSGIINKFCSAKDSE





HNEKYEDTFLESEEIGTKVEVVERKEHLHTDILKRGSEMDNNCSPTRKD





FTGEKIFQEDTIPRTQIERRKTSLYFSSKYNKEALSPPRRKAFKKWTPP





RSPFNLVQETLFHDPWKLLIATIFLNRTSGKMAIPVLWKFLEKYPSAEV





ARTADWRDVSELLKPLGLYDLRAKTIVKFSDEYLTKQWKYPIELHGIGK





YGNDSYRIFCVNEWKQVHPEDHKLNKYHDWLWENHEKLSLS





TDG (human)


(SEQ ID NO: 3872)


MEAENAGSYSLQQAQAFYTFPFQQLMAEAPNMAVVNEQQMPEEVPAPAP





AQEPVQEAPKGRKRKPRTTEPKQPVEPKKPVESKKSGKSAKSKEKQEKI





TDTFKVKRKVDRFNGVSEAELLTKTLPDILTFNLDIVIIGINPGLMAAY





KGHHYPGPGNHFWKCLFMSGLSEVQLNHMDDHTLPGKYGIGFTNMVERT





TPGSKDLSSKEFREGGRILVQKLQKYQPRIAVFNGKCIYEIFSKEVFGV





KVKNLEFGLQPHKIPDTETLCYVMPSSSARCAQFPRAQDKVHYYIKLKD





LRDQLKGIERNMDVQEVQYTFDLQLAQEDAKKMAVKEEKYDPGYEAAYG





GAYGENPCSSEPCGFSSNGLIESVELRGESAFSGIPNGQWMTQSFTDQI





PSFSNHCGTQEQEEESHA






In some embodiments, the fusion proteins described herein may comprise one or more heterologous protein domains (e.g., about or more than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more domains in addition to the prime editor components). A fusion protein may comprise any additional protein sequence, and optionally a linker sequence between any two domains. Other exemplary features that may be present are localization sequences, such as cytoplasmic localization sequences, export sequences, such as nuclear export sequences, or other localization sequences, as well as sequence tags that are useful for solubilization, purification, or detection of the fusion proteins.


Examples of protein domains that may be fused to a multi-flap prime editor or component thereof (e.g., the napDNAbp domain, the polymerase domain, or the NLS domain) include, without limitation, epitope tags, and reporter gene sequences. Non-limiting examples of epitope tags include histidine (His) tags, V5 tags, FLAG tags, influenza hemagglutinin (HA) tags, Myc tags, VSV-G tags, and thioredoxin (Trx) tags. Examples of reporter genes include, but are not limited to, glutathione-5-transferase (GST), horseradish peroxidase (HRP), chloramphenicol acetyltransferase (CAT), beta-galactosidase, beta-glucuronidase, luciferase, green fluorescent protein (GFP), HcRed, DsRed, cyan fluorescent protein (CFP), yellow fluorescent protein (YFP), and autofluorescent proteins including blue fluorescent protein (BFP). A multi-flap prime editor may be fused to a gene sequence encoding a protein or a fragment of a protein that bind DNA molecules or bind other cellular molecules, including, but not limited to, maltose binding protein (MBP), S-tag, Lex A DNA binding domain (DBD) fusions, GAL4 DNA binding domain fusions, and herpes simplex virus (HSV) BP16 protein fusions. Additional domains that may form part of a prime editor are described in US Patent Publication No. 2011/0059502, published Mar. 10, 2011 and incorporated herein by reference in its entirety.


In an aspect of the disclosure, a reporter gene which includes, but is not limited to, glutathione-5-transferase (GST), horseradish peroxidase (HRP), chloramphenicol acetyltransferase (CAT) beta-galactosidase, beta-glucuronidase, luciferase, green fluorescent protein (GFP), HcRed, DsRed, cyan fluorescent protein (CFP), yellow fluorescent protein (YFP), and autofluorescent proteins including blue fluorescent protein (BFP), may be introduced into a cell to encode a gene product which serves as a marker by which to measure the alteration or modification of expression of the gene product. In certain embodiments of the disclosure the gene product is luciferase. In a further embodiment of the disclosure the expression of the gene product is decreased.


Suitable protein tags provided herein include, but are not limited to, biotin carboxylase carrier protein (BCCP) tags, myc-tags, calmodulin-tags, FLAG-tags, hemagglutinin (HA)-tags, polyhistidine tags, also referred to as histidine tags or His-tags, maltose binding protein (MBP)-tags, nus-tags, glutathione-S-transferase (GST)-tags, green fluorescent protein (GFP)-tags, thioredoxin-tags, S-tags, Softags (e.g., Softag 1, Softag 3), strep-tags, biotin ligase tags, FlAsH tags, V5 tags, and SBP-tags. Additional suitable sequences will be apparent to those of skill in the art. In some embodiments, the fusion protein comprises one or more His tags.


In some embodiments of the present disclosure, the activity of the multi-flap prime editing system may be temporally regulated by adjusting the residence time, the amount, and/or the activity of the expressed components of the PE system. For example, as described herein, the PE may be fused with a protein domain that is capable of modifying the intracellular half-life of the PE. In certain embodiments involving two or more vectors (e.g., a vector system in which the components described herein are encoded on two or more separate vectors), the activity of the PE system may be temporally regulated by controlling the timing in which the vectors are delivered. For example, in some embodiments a vector encoding the nuclease system may deliver the PE prior to the vector encoding the template. In other embodiments, the vector encoding the PEgRNA may deliver the guide prior to the vector encoding the PE system. In some embodiments, the vectors encoding the PE system and PEgRNA are delivered simultaneously. In certain embodiments, the simultaneously delivered vectors temporally deliver, e.g., the PE, PEgRNA, and/or second strand guide RNA components. In further embodiments, the RNA (such as, e.g., the nuclease transcript) transcribed from the coding sequence on the vectors may further comprise at least one element that is capable of modifying the intracellular half-life of the RNA and/or modulating translational control. In some embodiments, the half-life of the RNA may be increased. In some embodiments, the half-life of the RNA may be decreased. In some embodiments, the element may be capable of increasing the stability of the RNA. In some embodiments, the element may be capable of decreasing the stability of the RNA. In some embodiments, the element may be within the 3′ UTR of the RNA. In some embodiments, the element may include a polyadenylation signal (PA). In some embodiments, the element may include a cap, e.g., an upstream mRNA or PEgRNA end. In some embodiments, the RNA may comprise no PA such that it is subject to quicker degradation in the cell after transcription. In some embodiments, the element may include at least one AU-rich element (ARE). The AREs may be bound by ARE binding proteins (ARE-BPs) in a manner that is dependent upon tissue type, cell type, timing, cellular localization, and environment. In some embodiments the destabilizing element may promote RNA decay, affect RNA stability, or activate translation. In some embodiments, the ARE may comprise 50 to 150 nucleotides in length. In some embodiments, the ARE may comprise at least one copy of the sequence AUUUA. In some embodiments, at least one ARE may be added to the 3′ UTR of the RNA. In some embodiments, the element may be a Woodchuck Hepatitis Virus (WHP).


Posttranscriptional Regulatory Element (WPRE), which creates a tertiary structure to enhance expression from the transcript. In further embodiments, the element is a modified and/or truncated WPRE sequence that is capable of enhancing expression from the transcript, as described, for example in Zufferey et al., J Virol, 73(4): 2886-92 (1999) and Flajolet et al., J Virol, 72(7): 6175-80 (1998). In some embodiments, the WPRE or equivalent may be added to the 3′ UTR of the RNA. In some embodiments, the element may be selected from other RNA sequence motifs that are enriched in either fast- or slow-decaying transcripts.


In some embodiments, the vector encoding the PE or the PEgRNA may be self-destroyed via cleavage of a target sequence present on the vector by the PE system. The cleavage may prevent continued transcription of a PE or a PEgRNA from the vector. Although transcription may occur on the linearized vector for some amount of time, the expressed transcripts or proteins subject to intracellular degradation will have less time to produce off-target effects without continued supply from expression of the encoding vectors.


[6] PEgRNAs

The multi-flap prime editing system described herein contemplates the use of any suitable PEgRNAs. The mechanism of target-primed reverse transcription (TPRT) can be leveraged or adapted for conducting precision and versatile CRISPR/Cas-based genome editing through the use of a specially configured guide RNA comprising a reverse transcription (RT) template sequence that codes for the desired nucleotide change. The application refers to this specially configured guide RNA as an “extended guide RNA” or a “PEgRNA” since the RT template sequence can be provided as an extension of a standard or traditional guide RNA molecule. The application contemplates any suitable configuration or arrangement for the extended guide RNAs for use in dual-flap and quadruple-flap prime editing.


The general designs of pegRNAs used for dual-flap and multi-flap prime editing are shown in FIG. 92. pegRNAs used for dual-flap and multi-flap prime editing have a similar design to those used for classic prime editing, however it is not necessary for the RT template region to encode any homology to the target locus. Instead, the two pegRNAs can in various embodiments contain RT templates that encode the synthesis of 3′ flaps whose 3′ ends are reverse complement sequences of one another. This complementarity between the 3′ flaps promotes their annealing and replacement of the endogenous DNA sequence with the intended new DNA sequence. This necessitates that the 5′ regions of the RT templates in the two pegRNAs are reverse complement sequences to one another, and this amount of complementarity can vary (FIG. 92).


PEgRNA Architecture



FIG. 3A shows one embodiment of an extended guide RNA usable in the multi-flap prime editing system disclosed herein whereby a traditional guide RNA (the green portion) includes a ˜20 nt protospacer sequence and a gRNA core region, which binds with the napDNAbp. In this embodiment, the guide RNA includes an extended RNA segment at the 5′ end, i.e., a 5′ extension. In this embodiment, the 5′ extension includes a reverse transcription template sequence, a reverse transcription primer binding site, and an optional 5-20 nucleotide linker sequence. As shown in FIGS. 1A-1B, the RT primer binding site hybrizes to the free 3′ end that is formed after a nick is formed in the non-target strand of the R-loop, thereby priming reverse transcriptase for DNA polymerization in the 5′-3′ direction.



FIG. 3B shows another embodiment of an extended guide RNA usable in the prime editing system disclosed herein whereby a traditional guide RNA (the green portion) includes a ˜20 nt protospacer sequence and a gRNA core, which binds with the napDNAbp. In this embodiment, the guide RNA includes an extended RNA segment at the 3′ end, i.e., a 3′ extension. In this embodiment, the 3′ extension includes a reverse transcription template sequence, and a reverse transcription primer binding site. As shown in FIGS. 1C-1D, the RT primer binding site hybrizes to the free 3′ end that is formed after a nick is formed in the non-target strand of the R-loop, thereby priming reverse transcriptase for DNA polymerization in the 5′-3′ direction.



FIG. 3C shows another embodiment of an extend guide RNA usable in the prime editing system disclosed herein whereby a traditional guide RNA (the green portion) includes a ˜20 nt protospacer sequence and a gRNA core, which binds with the napDNAbp. In this embodiment, the guide RNA includes an extended RNA segment at an intermolecular position within the gRNA core, i.e., an intramolecular extension. In this embodiment, the intramolecular extension includes a reverse transcription template sequence, and a reverse transcription primer binding site. The RT primer binding site hybrizes to the free 3′ end that is formed after a nick is formed in the non-target strand of the R-loop, thereby priming reverse transcriptase for DNA polymerization in the 5′-3′ direction.


In one embodiment, the position of the intermolecular RNA extension is not in the protospacer sequence of the guide RNA. In another embodiment, the position of the intermolecular RNA extension in the gRNA core. In still another embodiment, the position of the intermolecular RNA extension is any with the guide RNA molecule except within the protospacer sequence, or at a position which disrupts the protospacer sequence.


In one embodiment, the intermolecular RNA extension is inserted downstream from the 3′ end of the protospacer sequence. In another embodiment, the intermolecular RNA extension is inserted at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides downstream of the 3′ end of the protospacer sequence.


In other embodiments, the intermolecular RNA extension is inserted into the gRNA, which refers to the portion of the guide RNA corresponding or comprising the tracrRNA, which binds and/or interacts with the Cas9 protein or equivalent thereof (i.e, a different napDNAbp). Preferably the insertion of the intermolecular RNA extension does not disrupt or minimally disrupts the interaction between the tracrRNA portion and the napDNAbp.


The length of the RNA extension (which includes at least the RT template and primer binding site, e.g., see FIG. 92) can be any useful length. In various embodiments, the RNA extension is at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides, at least 90 nucleotides, at least 100 nucleotides, at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, or at least 500 nucleotides in length.


The RT template sequence can also be any suitable length. For example, the RT template sequence can be at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides, at least 90 nucleotides, at least 100 nucleotides, at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, or at least 500 nucleotides in length.


In still other embodiments, wherein the reverse transcription primer binding site sequence is at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides, at least 90 nucleotides, at least 100 nucleotides, at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, or at least 500 nucleotides in length.


In other embodiments, the optional linker or spacer sequence is at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides, at least 90 nucleotides, at least 100 nucleotides, at least 200 nucleotides, at least 300 nucleotides, at least 400 nucleotides, or at least 500 nucleotides in length.


The RT template sequence, in certain embodiments, encodes a single-stranded DNA molecule which is homologous to the non-target strand (and thus, complementary to the corresponding site of the target strand) but includes one or more nucleotide changes. The least one nucleotide change may include one or more single-base nucleotide changes, one or more deletions, and one or more insertions.


As depicted in FIG. 1G, the synthesized single-stranded DNA product of the RT template sequence is homologous to the non-target strand and contains one or more nucleotide changes. The single-stranded DNA product of the RT template sequence hybridizes in equilibrium with the complementary target strand sequence, thereby displacing the homologous endogenous target strand sequence. The displaced endogenous strand may be referred to in some embodiments as a 5′ endogenous DNA flap species (e.g., see FIG. 1E). This 5′ endogenous DNA flap species can be removed by a 5′ flap endonuclease (e.g., FEN1) and the single-stranded DNA product, now hybridized to the endogenous target strand, may be ligated, thereby creating a mismatch between the endogenous sequence and the newly synthesized strand. The mismatch may be resolved by the cell's innate DNA repair and/or replication processes.


In various embodiments, the nucleotide sequence of the RT template sequence corresponds to the nucleotide sequence of the non-target strand which becomes displaced as the 5′ flap species and which overlaps with the site to be edited.


In various embodiments of the extended guide RNAs, the reverse transcription template sequence may encode a single-strand DNA flap that is complementary to an endogenous DNA sequence adjacent to a nick site, wherein the single-strand DNA flap comprises a desired nucleotide change. The single-stranded DNA flap may displace an endogenous single-strand DNA at the nick site. The displaced endogenous single-strand DNA at the nick site can have a 5′ end and form an endogenous flap, which can be excised by the cell. In various embodiments, excision of the 5′ end endogenous flap can help drive product formation since removing the 5′ end endogenous flap encourages hybridization of the single-strand 3′ DNA flap to the corresponding complementary DNA strand, and the incorporation or assimilation of the desired nucleotide change carried by the single-strand 3′ DNA flap into the target DNA.


In various embodiments of the extended guide RNAs, the cellular repair of the single-strand DNA flap results in installation of the desired nucleotide change, thereby forming a desired product.


In still other embodiments, the desired nucleotide change is installed in an editing window that is between about −5 to +5 of the nick site, or between about −10 to +10 of the nick site, or between about −20 to +20 of the nick site, or between about −30 to +30 of the nick site, or between about −40 to +40 of the nick site, or between about −50 to +50 of the nick site, or between about −60 to +60 of the nick site, or between about −70 to +70 of the nick site, or between about −80 to +80 of the nick site, or between about −90 to +90 of the nick site, or between about −100 to +100 of the nick site, or between about −200 to +200 of the nick site. In other embodiments, the desired nucleotide change is installed in an editing window that is between about +1 to +2 from the nick site, or about +1 to +3, +1 to +4, +1 to +5, +1 to +6, +1 to +7, +1 to +8, +1 to +9, +1 to +10, +1 to +11, +1 to +12, +1 to +13, +1 to +14, +1 to +15, +1 to +16, +1 to +17, +1 to +18, +1 to +19, +1 to +20, +1 to +21, +1 to +22, +1 to +23, +1 to +24, +1 to +25, +1 to +26, +1 to +27, +1 to +28, +1 to +29, +1 to +30, +1 to +31, +1 to +32, +1 to +33, +1 to +34, +1 to +35, +1 to +36, +1 to +37, +1 to +38, +1 to +39, +1 to +40, +1 to +41, +1 to +42, +1 to +43, +1 to +44, +1 to +45, +1 to +46, +1 to +47, +1 to +48, +1 to +49, +1 to +50, +1 to +51, +1 to +52, +1 to +53, +1 to +54, +1 to +55, +1 to +56, +1 to +57, +1 to +58, +1 to +59, +1 to +60, +1 to +61, +1 to +62, +1 to +63, +1 to +64, +1 to +65, +1 to +66, +1 to +67, +1 to +68, +1 to +69, +1 to +70, +1 to +71, +1 to +72, +1 to +73, +1 to +74, +1 to +75, +1 to +76, +1 to +77, +1 to +78, +1 to +79, +1 to +80, +1 to +81, +1 to +82, +1 to +83, +1 to +84, +1 to +85, +1 to +86, +1 to +87, +1 to +88, +1 to +89, +1 to +90, +1 to +90, +1 to +91, +1 to +92, +1 to +93, +1 to +94, +1 to +95, +1 to +96, +1 to +97, +1 to +98, +1 to +99, +1 to +100, +1 to +101, +1 to +102, +1 to +103, +1 to +104, +1 to +105, +1 to +106, +1 to +107, +1 to +108, +1 to +109, +1 to +110, +1 to +111, +1 to +112, +1 to +113, +1 to +114, +1 to +115, +1 to +116, +1 to +117, +1 to +118, +1 to +119, +1 to +120, +1 to +121, +1 to +122, +1 to +123, +1 to +124, or +1 to +125 from the nick site.


In still other embodiments, the desired nucleotide change is installed in an editing window that is between about +1 to +2 from the nick site, or about +1 to +5, +1 to +10, +1 to +15, +1 to +20, +1 to +25, +1 to +30, +1 to +35, +1 to +40, +1 to +45, +1 to +50, +1 to +55, +1 to +100, +1 to +105, +1 to +110, +1 to +115, +1 to +120, +1 to +125, +1 to +130, +1 to +135, +1 to +140, +1 to +145, +1 to +150, +1 to +155, +1 to +160, +1 to +165, +1 to +170, +1 to +175, +1 to +180, +1 to +185, +1 to +190, +1 to +195, or +1 to +200, from the nick site.


In various aspects, the extended guide RNAs are modified versions of a guide RNA. Guide RNAs maybe naturally occurring, expressed from an encoding nucleic acid, or synthesized chemically. Methods are well known in the art for obtaining or otherwise synthesizing guide RNAs and for determining the appropriate sequence of the guide RNA, including the protospacer sequence which interacts and hybridizes with the target strand of a genomic target site of interest.


In various embodiments, the particular design aspects of a guide RNA sequence will depend upon the nucleotide sequence of a genomic target site of interest (i.e., the desired site to be edited) and the type of napDNAbp (e.g., Cas9 protein) present in the multi-flap prime editing systems described herein, among other factors, such as PAM sequence locations, percent G/C content in the target sequence, the degree of microhomology regions, secondary structures, etc.


In general, a guide sequence is any polynucleotide sequence having sufficient complementarity with a target polynucleotide sequence to hybridize with the target sequence and direct sequence-specific binding of a napDNAbp (e.g., a Cas9, Cas9 homolog, or Cas9 variant) to the target sequence. In some embodiments, the degree of complementarity between a guide sequence and its corresponding target sequence, when optimally aligned using a suitable alignment algorithm, is about or more than about 50%, 60%, 75%, 80%, 85%, 90%, 95%, 97.5%, 99%, or more. Optimal alignment may be determined with the use of any suitable algorithm for aligning sequences, non-limiting example of which include the Smith-Waterman algorithm, the Needleman-Wunsch algorithm, algorithms based on the Burrows-Wheeler Transform (e.g., the Burrows Wheeler Aligner), ClustalW, Clustal X, BLAT, Novoalign (Novocraft Technologies, ELAND (Illumina, San Diego, Calif.), SOAP (available at soap.genomics.org.cn), and Maq (available at maq.sourceforge.net). In some embodiments, a guide sequence is about or more than about 5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, 75, or more nucleotides in length.


In some embodiments, a guide sequence is less than about 75, 50, 45, 40, 35, 30, 25, 20, 15, 12, or fewer nucleotides in length. The ability of a guide sequence to direct sequence-specific binding of a multi-flap prime editor to a target sequence may be assessed by any suitable assay. For example, the components of a multi-flap prime editor, including the guide sequence to be tested, may be provided to a host cell having the corresponding target sequence, such as by transfection with vectors encoding the components of a multi-flap prime editor disclosed herein, followed by an assessment of preferential cleavage within the target sequence, such as by Surveyor assay as described herein. Similarly, cleavage of a target polynucleotide sequence may be evaluated in a test tube by providing the target sequence, components of a multi-flap prime editor, including the guide sequence to be tested and a control guide sequence different from the test guide sequence, and comparing binding or rate of cleavage at the target sequence between the test and control guide sequence reactions. Other assays are possible, and will occur to those skilled in the art.


A guide sequence may be selected to target any target sequence. In some embodiments, the target sequence is a sequence within a genome of a cell. Exemplary target sequences include those that are unique in the target genome. For example, for the S. pyogenes Cas9, a unique target sequence in a genome may include a Cas9 target site of the form MMMMMMMMNNNNNNNNNNNNXGG (SEQ ID NO: 204) where NNNNNNNNNNNNXGG (SEQ ID NO: 205) (N is A, G, T, or C; and X can be anything). A unique target sequence in a genome may include an S. pyogenes Cas9 target site of the form MMMMMMMMMNNNNNNNNNNNXGG (SEQ ID NO: 206) where NNNNNNNNNNNXGG (SEQ ID NO: 207) (N is A, G, T, or C; and X can be anything). For the S. thermophilus CRISPR1Cas9, a unique target sequence in a genome may include a Cas9 target site of the form MMMMMMMMNNNNNNNNNNNNXXAGAAW (SEQ ID NO: 208) where NNNNNNNNNNNNXXAGAAW (SEQ ID NO: 209) (N is A, G, T, or C; X can be anything; and W is A or T). A unique target sequence in a genome may include an S. thermophilus CRISPR 1 Cas9 target site of the form MMMMMMMMMNNNNNNNNNNNXXAGAAW (SEQ ID NO: 210) where NNNNNNNNNNNXXAGAAW (SEQ ID NO: 211) (N is A, G, T, or C; X can be anything; and W is A or T). For the S. pyogenes Cas9, a unique target sequence in a genome may include a Cas9 target site of the form MMMMMMMMNNNNNNNNNNNNXGGXG (SEQ ID NO: 212) where NNNNNNNNNNNNXGGXG (SEQ ID NO: 213) (N is A, G, T, or C; and X can be anything). A unique target sequence in a genome may include an S. pyogenes Cas9 target site of the form MMMMMMMMMNNNNNNNNNNNXGGXG (SEQ ID NO: 214) where NNNNNNNNNNNXGGXG (SEQ ID NO: 215) (N is A, G, T, or C; and X can be anything). In each of these sequences “M” may be A, G, T, or C, and need not be considered in identifying a sequence as unique.


As used herein in a PEgRNA or guide RNA sequence, unless indicated otherwise, it should be appreciated that the letter “T” or “thymine” indicates a nucleobase in a DNA sequence that encodes the PEgRNA or guide RNA sequence, and is intended to refer to a uracil (U) nucleobase of the PEgRNA or guide RNA or any chemically modified uracil nucleobase known in the art, such as 5-methoxyuracil.


In some embodiments, a guide sequence is selected to reduce the degree of secondary structure within the guide sequence. Secondary structure may be determined by any suitable polynucleotide folding algorithm. Some programs are based on calculating the minimal Gibbs free energy. An example of one such algorithm is mFold, as described by Zuker and Stiegler (Nucleic Acids Res. 9 (1981), 133-148). Another example folding algorithm is the online webserver RNAfold, developed at Institute for Theoretical Chemistry at the University of Vienna, using the centroid structure prediction algorithm (see e.g. A. R. Gruber et al., 2008, Cell 106(1): 23-24; and P A Can and G M Church, 2009, Nature Biotechnology 27(12): 1151-62). Further algorithms may be found in U.S. application Ser. No. 61/836,080; Broad Reference BI-2013/004A); incorporated herein by reference.


In general, a tracr mate sequence includes any sequence that has sufficient complementarity with a tracr sequence to promote one or more of: (1) excision of a guide sequence flanked by tracr mate sequences in a cell containing the corresponding tracr sequence; and (2) formation of a complex at a target sequence, wherein the complex comprises the tracr mate sequence hybridized to the tracr sequence. In general, degree of complementarity is with reference to the optimal alignment of the tracr mate sequence and tracr sequence, along the length of the shorter of the two sequences. Optimal alignment may be determined by any suitable alignment algorithm, and may further account for secondary structures, such as self-complementarity within either the tracr sequence or tracr mate sequence. In some embodiments, the degree of complementarity between the tracr sequence and tracr mate sequence along the length of the shorter of the two when optimally aligned is about or more than about 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 97.5%, 99%, or higher. In some embodiments, the tracr sequence is about or more than about 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, or more nucleotides in length. In some embodiments, the tracr sequence and tracr mate sequence are contained within a single transcript, such that hybridization between the two produces a transcript having a secondary structure, such as a hairpin. Preferred loop forming sequences for use in hairpin structures are four nucleotides in length, and most preferably have the sequence GAAA. However, longer or shorter loop sequences may be used, as may alternative sequences. The sequences preferably include a nucleotide triplet (for example, AAA), and an additional nucleotide (for example C or G). Examples of loop forming sequences include CAAA and AAAG. In an embodiment of the invention, the transcript or transcribed polynucleotide sequence has at least two or more hairpins. In preferred embodiments, the transcript has two, three, four or five hairpins. In a further embodiment of the invention, the transcript has at most five hairpins. In some embodiments, the single transcript further includes a transcription termination sequence; preferably this is a polyT sequence, for example six T nucleotides. Further non-limiting examples of single polynucleotides comprising a guide sequence, a tracr mate sequence, and a tracr sequence are as follows (listed 5′ to 3′), where “N” represents a base of a guide sequence, the first block of lower case letters represent the tracr mate sequence, and the second block of lower case letters represent the tracr sequence, and the final poly-T sequence represents the transcription terminator:









(1)


(SEQ ID NO: 216)


NNNNNNNNGTTTTTGTACTCTCAAGATTTAGAAATAAATCTTGCAGAAG


CTACAAAGATAAGGCTTCATGCCGAAATCAACACCCTGTCATTTTATGG


CAGGGTGTTTTCGTTATTTAATTTTTT;





(2)


(SEQ ID NO: 217)


NNNNNNNNNNNNNNNNNNGTTTTTGTACTCTCAGAAATGCAGAAGCTAC


AAAGATAAGGCTTCATGCCGAAATCAACACCCTGTCATTTTATGGCAGG


GTGTTTTCGTTATTTAATTTTTT;





(3)


(SEQ ID NO: 218)


NNNNNNNNNNNNNNNNNNNNGTTTTTGTACTCTCAGAAATGCAGAAGCT


ACAAAGATAAGGCTTCATGCCGAAATCAACACCCTGTCATTTTATGGCA


GGGTGTTTTTT;





(4)


(SEQ ID NO: 219)


NNNNNNNNNNNNNNNNNNNNGTTTTAGAGCTAGAAATAGCAAGTTAAAA


TAAGGCTAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGTCGGTGCTT


TTTT;





(5)


(SEQ ID NO: 220)


NNNNNNNNNNNNNNNNNNNNGTTTTAGAGCTAGAAATAGCAAGTTAAAA


TAAGGCTAGTCCGTTATCAACTTGAAAAAGTGTTTTTTT;


AND





(6)


(SEQ ID NO: 221)


NNNNNNNNNNNNNNNNNNNNGTTTTAGAGCTAGAAATAGCAAGTTAAAA


TAAGGCTAGTCCGTTATCATTTTTTTT.






In some embodiments, sequences (1) to (3) are used in combination with Cas9 from S. thermophilus CRISPR1. In some embodiments, sequences (4) to (6) are used in combination with Cas9 from S. pyogenes. In some embodiments, the tracr sequence is a separate transcript from a transcript comprising the tracr mate sequence.


It will be apparent to those of skill in the art that in order to target any of the fusion proteins comprising a Cas9 domain and a single-stranded DNA binding protein, as disclosed herein, to a target site, e.g., a site comprising a point mutation to be edited, it is typically necessary to co-express the fusion protein together with a guide RNA, e.g., an sgRNA. As explained in more detail elsewhere herein, a guide RNA typically comprises a tracrRNA framework allowing for Cas9 binding, and a guide sequence, which confers sequence specificity to the Cas9:nucleic acid editing enzyme/domain fusion protein.


In some embodiments, the guide RNA comprises a structure 5′-[guide sequence]-GUUUUAGAGCUAGAAAUAGCAAGUUAAAAUAAAGGCUAGUCCGUUAUCAACUUGAAAAA GUGGCACCGAGUCGGUGCUUUUU-3′ (SEQ ID NO: 222), wherein the guide sequence comprises a sequence that is complementary to the target sequence. The guide sequence is typically 20 nucleotides long. The sequences of suitable guide RNAs for targeting Cas9:nucleic acid editing enzyme/domain fusion proteins to specific genomic target sites will be apparent to those of skill in the art based on the instant disclosure. Such suitable guide RNA sequences typically comprise guide sequences that are complementary to a nucleic sequence within 50 nucleotides upstream or downstream of the target nucleotide to be edited. Some exemplary guide RNA sequences suitable for targeting any of the provided fusion proteins to specific target sequences are provided herein. Additional guide sequences are well known in the art and can be used with the multi-flap prime editor described herein.


In other embodiments, the PEgRNAs include those depicted in FIG. 3D.


In still other embodiments, the PEgRNAs may include those depicted in FIG. 3E.



FIG. 3D provides the structure of an embodiment of a PEgRNA contemplated herein and which may be designed in accordance with the methodology defined in Example 2. The PEgRNA comprises three main component elements ordered in the 5′ to 3′ direction, namely: a spacer, a gRNA core, and an extension arm at the 3′ end. The extension arm may further be divided into the following structural elements in the 5′ to 3′ direction, namely: a primer binding site (A), an edit template (B), and a homology arm (C). In addition, the PEgRNA may comprise an optional 3′ end modifier region (e1) and an optional 5′ end modifier region (e2). Still further, the PEgRNA may comprise a transcriptional termination signal at the 3′ end of the PEgRNA (not depicted). These structural elements are further defined herein. The depiction of the structure of the PEgRNA is not meant to be limiting and embraces variations in the arrangement of the elements. For example, the optional sequence modifiers (e1) and (e2) could be positioned within or between any of the other regions shown, and not limited to being located at the 3′ and 5′ ends.



FIG. 3E provides the structure of another embodiment of a PEgRNA contemplated herein may be designed in accordance with the methodology defined in Example 2. The PEgRNA comprises three main component elements ordered in the 5′ to 3′ direction, namely: a spacer, a gRNA core, and an extension arm at the 3′ end. The extension arm may further be divided into the following structural elements in the 5′ to 3′ direction, namely: a primer binding site (A), an edit template (B), and a homology arm (C). In addition, the PEgRNA may comprise an optional 3′ end modifier region (e1) and an optional 5′ end modifier region (e2). Still further, the PEgRNA may comprise a transcriptional termination signal on the 3′ end of the PEgRNA (not depicted). These structural elements are further defined herein. The depiction of the structure of the PEgRNA is not meant to be limiting and embraces variations in the arrangement of the elements. For example, the optional sequence modifiers (e1) and (e2) could be positioned within or between any of the other regions shown, and not limited to being located at the 3′ and 5′ ends.


PEgRNA Improvements


The PEgRNAs may also include additional design improvements that may modify the properties and/or characteristics of PEgRNAs thereby improving the efficacy of multi-flap prime editing. In various embodiments, these improvements may belong to one or more of a number of different categories, including but not limited to: (1) designs to enable efficient expression of functional PEgRNAs from non-polymerase III (pol III) promoters, which would enable the expression of longer PEgRNAs without burdensome sequence requirements; (2) improvements to the core, Cas9-binding PEgRNA scaffold, which could improve efficacy; (3) modifications to the PEgRNA to improve RT processivity, enabling the insertion of longer sequences at targeted genomic loci; and (4) addition of RNA motifs to the 5′ or 3′ termini of the PEgRNA that improve PEgRNA stability, enhance RT processivity, prevent misfolding of the PEgRNA, or recruit additional factors important for genome editing.


In one embodiment, PEgRNA could be designed with polIII promoters to improve the expression of longer-length PEgRNA with larger extension arms. sgRNAs are typically expressed from the U6 snRNA promoter. This promoter recruits pol III to express the associated RNA and is useful for expression of short RNAs that are retained within the nucleus. However, pol III is not highly processive and is unable to express RNAs longer than a few hundred nucleotides in length at the levels required for efficient genome editing. Additionally, pol III can stall or terminate at stretches of U's, potentially limiting the sequence diversity that could be inserted using a PEgRNA. Other promoters that recruit polymerase II (such as pCMV) or polymerase I (such as the U1 snRNA promoter) have been examined for their ability to express longer sgRNAs. However, these promoters are typically partially transcribed, which would result in extra sequence 5′ of the spacer in the expressed PEgRNA, which has been shown to result in markedly reduced Cas9:sgRNA activity in a site-dependent manner. Additionally, while pol III-transcribed PEgRNAs can simply terminate in a run of 6-7 U's, PEgRNAs transcribed from pol II or pol I would require a different termination signal. Often such signals also result in polyadenylation, which would result in undesired transport of the PEgRNA from the nucleus. Similarly, RNAs expressed from pol II promoters such as pCMV are typically 5′-capped, also resulting in their nuclear export.


Previously, Rinn and coworkers screened a variety of expression platforms for the production of long-noncoding RNA-(lncRNA) tagged sgRNAs183. These platforms include RNAs expressed from pCMV and that terminate in the ENE element from the MALAT1 ncRNA from humans', the PAN ENE element from KSHV185, or the 3′ box from U1 snRNA186. Notably, the MALAT1 ncRNA and PAN ENEs form triple helices protecting the polyA-tail184,187 These constructs could also enhance RNA stability. It is contemplated that these expression systems will also enable the expression of longer PEgRNAs.


In addition, a series of methods have been designed for the cleavage of the portion of the pol II promoter that would be transcribed as part of the PEgRNA, adding either a self-cleaving ribozyme such as the hammerhead188, pistol189, hatchet189, hairpin190, VS191, twister192, or twister sister192 ribozymes, or other self-cleaving elements to process the transcribed guide, or a hairpin that is recognized by Csy4193 and also leads to processing of the guide. Also, it is hypothesized that incorporation of multiple ENE motifs could lead to improved PEgRNA expression and stability, as previously demonstrated for the KSHV PAN RNA and element185. It is also anticipated that circularizing the PEgRNA in the form of a circular intronic RNA (ciRNA) could also lead to enhanced RNA expression and stability, as well as nuclear localization194.


In various embodiments, the PEgRNA may include various above elements, as exemplified by the following sequence.


Non-limiting example 1—PEgRNA expression platform consisting of pCMV, Csy4 hairpin, the PEgRNA, and MALAT1 ENE









(SEQ ID NO: 223)


TAGTTATTAATAGTAATCAATTACGGGGTCATTAGTTCATAGCCCATAT





ATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGAC





CGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCAT





AGTAACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTA





CGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCATATGCCAAGTA





CGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGC





CCAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACATCTACGTA





TTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATG





GGCGTGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCAT





TGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCA





AAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTG





TACGGTGGGAGGTCTATATAAGCAGAGCTGGTTTAGTGAACCGTCAGAT





CGTTCACTGCCGTATAGGCAGGGCCCAGACTGAGCACGTGAGTTTTAGA





GCTAGAAATAGCAAGTTAAAATAAGGCTAGTCCGTTATCAACTTGAAAA





AGTGGGACCGAGTCGGTCCTCTGCCATCAAAGCGTGCTCAGTCTGTTTT





AGGGTCATGAAGGTTTTTCTTTTCCTGAGAAAACAACACGTATTGTTTT





CTCAGGTTTTGCTTTTTGGCCTTTTTCTAGCTTAAAAAAAAAAAAAGCA





AAAGATGCTGGTGGTTGGCACTCCTGGTTTCCAGGACGGGGTTCAAATC





CCTGCGGCGTCTTTGCTTTGACT






Non-limiting example 2—PEgRNA expression platform consisting of pCMV, Csy4 hairing, the PEgRNA, and PAN ENE









(SEQ ID NO: 224)


TAGTTATTAATAGTAATCAATTACGGGGTCATTAGTTCATAGCCCATAT





ATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGAC





CGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCAT





AGTAACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTA





CGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCATATGCCAAGTA





CGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGC





CCAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACATCTACGTA





TTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATG





GGCGTGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCAT





TGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCA





AAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTG





TACGGTGGGAGGTCTATATAAGCAGAGCTGGTTTAGTGAACCGTCAGAT





CGTTCACTGCCGTATAGGCAGGGCCCAGACTGAGCACGTGAGTTTTAGA





GCTAGAAATAGCAAGTTAAAATAAGGCTAGTCCGTTATCAACTTGAAAA





AGTGGGACCGAGTCGGTCCTCTGCCATCAAAGCGTGCTCAGTCTGTTTT





GTTTTGGCTGGGTTTTTCCTTGTTCGCACCGGACACCTCCAGTGACCAG





ACGGCAAGGTTTTTATCCCAGTGTATATTGGAAAAACATGTTATACTTT





TGACAATTTAACGTGCCTAGAGCTCAAATTAAACTAATACCATAACGTA





ATGCAACTTACAACATAAATAAAGGTCAATGTTTAATCCATAAAAAAAA





AAAAAAAAAAA






Non-limiting example 3—PEgRNA expression platform consisting of pCMV, Csy4 hairing, the PEgRNA, and 3×PAN ENE









(SEQ ID NO: 225)


TAGTTATTAATAGTAATCAATTACGGGGTCATTAGTTCATAGCCCATAT





ATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGAC





CGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCAT





AGTAACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTA





CGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCATATGCCAAGTA





CGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGC





CCAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACATCTACGTA





TTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATG





GGCGTGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCAT





TGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCA





AAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTG





TACGGTGGGAGGTCTATATAAGCAGAGCTGGTTTAGTGAACCGTCAGAT





CGTTCACTGCCGTATAGGCAGGGCCCAGACTGAGCACGTGAGTTTTAGA





GCTAGAAATAGCAAGTTAAAATAAGGCTAGTCCGTTATCAACTTGAAAA





AGTGGGACCGAGTCGGTCCTCTGCCATCAAAGCGTGCTCAGTCTGTTTT





GTTTTGGCTGGGTTTTTCCTTGTTCGCACCGGACACCTCCAGTGACCAG





ACGGCAAGGTTTTTATCCCAGTGTATATTGGAAAAACATGTTATACTTT





TGACAATTTAACGTGCCTAGAGCTCAAATTAAACTAATACCATAACGTA





ATGCAACTTACAACATAAATAAAGGTCAATGTTTAATCCATAAAAAAAA





AAAAAAAAAAAACACACTGTTTTGGCTGGGTTTTTCCTTGTTCGCACCG





GACACCTCCAGTGACCAGACGGCAAGGTTTTTATCCCAGTGTATATTGG





AAAAACATGTTATACTTTTGACAATTTAACGTGCCTAGAGCTCAAATTA





AACTAATACCATAACGTAATGCAACTTACAACATAAATAAAGGTCAATG





TTTAATCCATAAAAAAAAAAAAAAAAAAATCTCTCTGTTTTGGCTGGGT





TTTTCCTTGTTCGCACCGGACACCTCCAGTGACCAGACGGCAAGGTTTT





TATCCCAGTGTATATTGGAAAAACATGTTATACTTTTGACAATTTAACG





TGCCTAGAGCTCAAATTAAACTAATACCATAACGTAATGCAACTTACAA





CATAAATAAAGGTCAATGTTTAATCCATAAAAAAAAAAAAAAAAAAA






Non-limiting example 4—PEgRNA expression platform consisting of pCMV, Csy4 hairing, the PEgRNA, and 3′ box









(SEQ ID NO: 226)


TAGTTATTAATAGTAATCAATTACGGGGTCATTAGTTCATAGCCCATAT





ATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGAC





CGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCAT





AGTAACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTA





CGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCATATGCCAAGTA





CGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGC





CCAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACATCTACGTA





TTAGTCATCGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATG





GGCGTGGATAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCAT





TGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCA





AAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTG





TACGGTGGGAGGTCTATATAAGCAGAGCTGGTTTAGTGAACCGTCAGAT





CGTTCACTGCCGTATAGGCAGGGCCCAGACTGAGCACGTGAGTTTTAGA





GCTAGAAATAGCAAGTTAAAATAAGGCTAGTCCGTTATCAACTTGAAAA





AGTGGGACCGAGTCGGTCCTCTGCCATCAAAGCGTGCTCAGTCTGTTTG





TTTCAAAAGTAGACTGTACGCTAAGGGTCATATCTTTTTTTGTTTGGTT





TGTGTCTTGGTTGGCGTCTTAAA






Non-limiting example 5—PEgRNA expression platform consisting of pU1, Csy4 hairping, the PEgRNA, and 3′ box









(SEQ ID NO: 227)


CTAAGGACCAGCTTCTTTGGGAGAGAACAGACGCAGGGGCGGGAGGGAA





AAAGGGAGAGGCAGACGTCACTTCCCCTTGGCGGCTCTGGCAGCAGATT





GGTCGGTTGAGTGGCAGAAAGGCAGACGGGGACTGGGCAAGGCACTGTC





GGTGACATCACGGACAGGGCGACTTCTATGTAGATGAGGCAGCGCAGAG





GCTGCTGCTTCGCCACTTGCTGCTTCACCACGAAGGAGTTCCCGTGCCC





TGGGAGCGGGTTCAGGACCGCTGATCGGAAGTGAGAATCCCAGCTGTGT





GTCAGGGCTGGAAAGGGCTCGGGAGTGCGCGGGGCAAGTGACCGTGTGT





GTAAAGAGTGAGGCGTATGAGGCTGTGTCGGGGCAGAGGCCCAAGATCT





CAGTTCACTGCCGTATAGGCAGGGCCCAGACTGAGCACGTGAGTTTTAG





AGCTAGAAATAGCAAGTTAAAATAAGGCTAGTCCGTTATCAACTTGAAA





AAGTGGGACCGAGTCGGTCCTCTGCCATCAAAGCGTGCTCAGTCTGTTT





CAGCAAGTTCAGAGAAATCTGAACTTGCTGGATTTTTGGAGCAGGGAGA





TGGAATAGGAGCTTGCTCCGTCCACTCCACGCATCGACCTGGTATTGCA





GTACCTCCAGGAACGGTGCACCCACTTTCTGGAGTTTCAAAAGTAGACT





GTACGCTAAGGGTCATATCTTTTTTTGTTTGGTTTGTGTCTTGGTTGGC





GTCTTAAA.






In various other embodiments, the PEgRNA may be improved by introducing improvements to the scaffold or core sequences. This can be done by introducing known


The core, Cas9-binding PEgRNA scaffold can likely be improved to enhance PE activity. Several such approaches have already been demonstrated. For instance, the first pairing element of the scaffold (P1) contains a GTTTT-AAAAC (SEQ ID NO: 3939) pairing element. Such runs of Ts have been shown to result in pol III pausing and premature termination of the RNA transcript. Rational mutation of one of the T-A pairs to a G-C pair in this portion of P1 has been shown to enhance sgRNA activity, suggesting this approach would also be feasible for PEgRNAs195. Additionally, increasing the length of P1 has also been shown to enhance sgRNA folding and lead to improved activity195, suggesting it as another avenue for the improvement of PEgRNA activity. Example improvements to the core can include:


PEgRNA containing a 6 nt extension to P1









(SEQ ID NO: 228)


GGCCCAGACTGAGCACGTGAGTTTTAGAGCTAGCTCATGAAAATGAGCT


AGCAAGTTAAAATAAGGCTAGTCCGTTATCAACTTGAAAAAGTGGGACC


GAGTCGGTCCTCTGCCATCAAAGCGTGCTCAGTCTGTTTTTTT






PEgRNA containing a T-A to G-C mutation within P1









(SEQ ID NO: 229)


GGCCCAGACTGAGCACGTGAGTTTGAGAGCTAGAAATAGCAAGTTTAAA


TAAGGCTAGTCCGTTATCAACTTGAAAAAGTGGGACCGAGTCGGTCCTC


TGCCATCAAAGCGTGCTCAGTCTGTTTTTTT






In various other embodiments, the PEgRNA may be improved by introducing modifications to the edit template region. As the size of the insertion templated by the PEgRNA increases, it is more likely to be degraded by endonucleases, undergo spontaneous hydrolysis, or fold into secondary structures unable to be reverse-transcribed by the RT or that disrupt folding of the PEgRNA scaffold and subsequent Cas9-RT binding. Accordingly, it is likely that modification to the template of the PEgRNA might be necessary to affect large insertions, such as the insertion of whole genes. Some strategies to do so include the incorporation of modified nucleotides within a synthetic or semi-synthetic PEgRNA that render the RNA more resistant to degradation or hydrolysis or less likely to adopt inhibitory secondary structures196. Such modifications could include 8-aza-7-deazaguanosine, which would reduce RNA secondary structure in G-rich sequences; locked-nucleic acids (LNA) that reduce degradation and enhance certain kinds of RNA secondary structure; 2′-O-methyl, 2′-fluoro, or 2′-O-methoxyethoxy modifications that enhance RNA stability. Such modifications could also be included elsewhere in the PEgRNA to enhance stability and activity. Alternatively or additionally, the template of the PEgRNA could be designed such that it both encodes for a desired protein product and is also more likely to adopt simple secondary structures that are able to be unfolded by the RT. Such simple structures would act as a thermodynamic sink, making it less likely that more complicated structures that would prevent reverse transcription would occur. Finally, one could also split the template into two, separate PEgRNAs. In such a design, a PE would be used to initiate transcription and also recruit a separate template RNA to the targeted site via an RNA-binding protein fused to Cas9 or an RNA recognition element on the PEgRNA itself such as the MS2 aptamer. The RT could either directly bind to this separate template RNA, or initiate reverse transcription on the original PEgRNA before swapping to the second template. Such an approach could enable long insertions by both preventing misfolding of the PEgRNA upon addition of the long template and also by not requiring dissociation of Cas9 from the genome for long insertions to occur, which could possibly be inhibiting PE-based long insertions.


In still other embodiments, the PEgRNA may be improved by introducing additional RNA motifs at the 5′ and 3′ termini of the PEgRNAs, or even at positions therein between (e.g., in the gRNA core region, or the spacer). Several such motifs—such as the PAN ENE from KSHV and the ENE from MALAT1 were discussed above as possible means to terminate expression of longer PEgRNAs from non-pol III promoters. These elements form RNA triple helices that engulf the polyA tail, resulting in their being retained within the nucleus184,187. However, by forming complex structures at the 3′ terminus of the PEgRNA that occlude the terminal nucleotide, these structures would also likely help prevent exonuclease-mediated degradation of PEgRNAs.


Other structural elements inserted at the 3′ terminus could also enhance RNA stability, albeit without enabling termination from non-pol III promoters. Such motifs could include hairpins or RNA quadruplexes that would occlude the 3′ terminus197, or self-cleaving ribozymes such as HDV that would result in the formation of a 2′-3′-cyclic phosphate at the 3′ terminus and also potentially render the PEgRNA less likely to be degraded by exonucleases198. Inducing the PEgRNA to cyclize via incomplete splicing—to form a ciRNA—could also increase PEgRNA stability and result in the PEgRNA being retained within the nucleus194.


Additional RNA motifs could also improve RT processivity or enhance PEgRNA activity by enhancing RT binding to the DNA-RNA duplex. Addition of the native sequence bound by the RT in its cognate retroviral genome could enhance RT activity199. This could include the native primer binding site (PBS), polypurine tract (PPT), or kissing loops involved in retroviral genome dimerization and initiation of transcription199.


Addition of dimerization motifs—such as kissing loops or a GNRA tetraloop/tetraloop receptor pair200—at the 5′ and 3′ termini of the PEgRNA could also result in effective circularization of the PEgRNA, improving stability. Additionally, it is envisioned that addition of these motifs could enable the physical separation of the PEgRNA spacer and primer, prevention occlusion of the spacer which would hinder PE activity. Short 5′ extensions or 3′ extensions to the PEgRNA that form a small toehold hairpin in the spacer region or along the primer binding site could also compete favorably against the annealing of intracomplementary regions along the length of the PEgRNA, e.g., the interaction between the spacer and the primer binding site that can occur. Finally, kissing loops could also be used to recruit other template RNAs to the genomic site and enable swapping of RT activity from one RNA to the other. As exemplary embodiments of various secondary structures, the PEgRNA depicted in FIG. 3D and FIG. 3E list a number secondary RNA structures that may be engineered into any region of the PEgRNA, including in the terminal portions of the extension arm (i.e., e1 and e2), as shown.


Example improvements include, but are not limited to:









PEgRNA-HDV fusion


(SEQ ID NO: 230)


GGCCCAGACTGAGCACGTGAGTTTTAGAGCTAGAAATAGCAAGTTAAAA





TAAGGCTAGTCCGTTATCAACTTGAAAAAGTGGGACCGAGTCGGTCCTC





TGCCATCAAAGCGTGCTCAGTCTGGGCCGGCATGGTCCCAGCCTCCTCG





CTGGCGCCGGCTGGGCAACATGCTTCGGCATGGCGAATGGGACTTTTTT 





T





PEgRNA-MMLV kissing loop


(SEQ ID NO: 231)


GGTGGGAGACGTCCCACCGGCCCAGACTGAGCACGTGAGTTTTAGAGCT





AGAAATAGCAAGTTAAAATAAGGCTAGTCCGTTATCAACTTGAAAAAGT





GGGACCGAGTCGGTCCTCTGCCATCAAAGCTTCGACCGTGCTCAGTCTG





GTGGGAGACGTCCCACCTTTTTTT





PEgRNA-VS ribozyme kissing loop


(SEQ ID NO: 232)


GAGCAGCATGGCGTCGCTGCTCACGGCCCAGACTGAGCACGTGAGTTTT





AGAGCTAGAAATAGCAAGTTAAAATAAGGCTAGTCCGTTATCAACTTGA





AAAAGTGGGACCGAGTCGGTCCTCTGCCATCAAAGCTTCGACCGTGCTC





AGTCTCCATCAGTTGACACCCTGAGGTTTTTTT





PEgRNA-GNRA tetraloop/tetraloop receptor


(SEQ ID NO: 233)


GCAGACCTAAGTGGUGACATATGGTCTGGGCCCAGACTGAGCACGTGAG





TTTTAGAGCTAUACGTAGCAAGTTAAAATAAGGCTAGTCCGTTATCAAC





TTUACGAAGTGGGACCGAGTCGGTCCTCTGCCATCAAAGCTTCGACCGT





GCTCAGTCTGCATGCGATTAGAAATAATCGCATGTTTTTTT





PEgRNA template switching secondary RNA-HDV fusion


(SEQ ID NO: 234)


TCTGCCATCAAAGCTGCGACCGTGCTCAGTCTGGTGGGAGACGTCCCAC





CGGCCGGCATGGTCCCAGCCTCCTCGCTGGCGCCGGCTGGGCAACATGC





TTCGGCATGGCGAATGGGACTTTTTTT






PEgRNA scaffolds could be further improved via directed evolution, in an analogous fashion to how SpCas9 and prime editors (PE) have been improved. Directed evolution could enhance PEgRNA recognition by Cas9 or evolved Cas9 variants. Additionally, it is likely that different PEgRNA scaffold sequences would be optimal at different genomic loci, either enhancing PE activity at the site in question, reducing off-target activities, or both. Finally, evolution of PEgRNA scaffolds to which other RNA motifs have been added would almost certainly improve the activity of the fused PEgRNA relative to the unevolved, fusion RNA. For instance, evolution of allosteric ribozymes composed of c-di-GMP-I aptamers and hammerhead ribozymes led to dramatically improved activity202, suggesting that evolution would improve the activity of hammerhead-PEgRNA fusions as well. In addition, while Cas9 currently does not generally tolerate 5′ extension of the sgRNA, directed evolution will likely generate enabling mutations that mitigate this intolerance, allowing additional RNA motifs to be utilized.


The present disclosure contemplates any such ways to further improve the efficacy of the multi-flap prime editing systems disclosed here.


In various embodiments, it may be advantageous to limit the appearance of consecutive sequence of Ts from the extension arm as consecutive series of T's may limit the capacity of the PEgRNA to be transcribed. For example, strings of at least consecutive three T's, at least consecutive four T's, at least consecutive five T's, at least consecutive six T's, at least consecutive seven T's, at least consecutive eight T's, at least consecutive nine T's, at least consecutive ten T's, at least consecutive eleven T's, at least consecutive twelve T's, at least consecutive thirteen T's, at least consecutive fourteen T's, or at least consecutive fifteen T's should be avoided when designing the PEgRNA, or should be at least removed from the final designed sequence. In one embodiment, one can avoid the includes of unwanted strings of consecutive T's in PEgRNA extension arms but avoiding target sites that are rich in consecutive A:T nucleobase pairs.


Split PEgRNA Designs for Trans Prime Editing


The instant disclosure also contemplates trans prime editing, which refers to a modified version of prime editing which operates by separating the PEgRNA into two distinct molecules: a guide RNA and a tPERT molecule. The tPERT molecule is programmed to co-localize with the prime editor complex at a target DNA site, bringing the primer binding site and the DNA synthesis template to the prime editor in trans. For example, see FIG. 3G for an embodiment of a trans prime editor (tPE) which shows a two-component system comprising (1) an recruiting protein (RP)-PE:gRNA complex and (2) a tPERT that includes a primer binding site and a DNA synthesis template joined to an RNA-protein recruitment domain (e.g., stem loop or hairpin), wherein the recruiting protein component of the RP-PE:gRNA complex recruits the tPERT to a target site to be edited, thereby associating the PBS and DNA synthesis template with the prime editor in trans. Said another way, the tPERT is engineered to contain (all or part of) the extension arm of a PEgRNA, which includes the primer binding site and the DNA synthesis template. One advantage of this approach is to separate the extension arm of a PEgRNA from the guide RNA, thereby minimizing annealing interactions that tend to occur between the PBS of the extension arm and the spacer sequence of the guide RNA.


A key feature of trans prime editing is the ability of the trans prime editor to recruit the tPERT to the site of DNA editing, thereby effectively co-localizing all of the functions of a PEgRNA at the site of prime editing. Recruitment can be achieve by installing an RNA-protein recruitment domain, such as a MS2 aptamer, into the tPERT and fusing a corresponding recruiting protein to the prime editor (e.g., via a linker to the napDNAbp or via a linker to the polymerase) that is capable of specifically binding to the RNA-protein recruitment domain, thereby recruiting the tPERT molecule to the prime editor complex. As depicted in the process described in FIG. 3H, the RP-PE:gRNA complex binds to and nicks the target DNA sequence. Then, the recruiting protein (RP) recruits a tPERT to co-localize to the prime editor complex bound to the DNA target site, thereby allowing the primer binding site, located on the tPERT, to bind to the primer sequence on the nicked strand, and subsequently, allowing the polymerase (e.g., RT) to synthesize a single strand of DNA against the DNA synthesis template, located on the tPERT, up through the 5′ end of the tPERT.


While the tPERT is shown in FIG. 3G and FIG. 3H as comprising the PBS and DNA synthesis template on the 5′ end of the RNA-protein recruitment domain, the tPERT in other configurations may be designed with the PBS and DNA synthesis template located on the 3′ end of the RNA-protein recruitment domain. However, the tPERT with the 5′ extension has the advantage that synthesis of the single strand of DNA will naturally terminate at the 5′ end of the tPERT and thus, does not risk using any portion of the RNA-protein recruitment domain as a template during the DNA synthesis stage of prime editing.


PEgRNA Design Method


The present disclosure also relates to methods for designing PEgRNAs.


In one aspect of design, the design approach can take into account the particular application for which prime editing is being used. For instance, and as exemplied and discussed herein, prime editing can be used, without limitation, to (a) install mutation-correcting changes to a nucleotide sequence, (b) install protein and RNA tags, (c) install immunoepitopes on proteins of interest, (d) install inducible dimerization domains in proteins, (e) install or remove sequences to alter that activity of a biomolecule, (f) install recombinase target sites to direct specific genetic changes, and (g) mutagenesis of a target sequence by using an error-prone RT. In addition to these methods which, in general, insert, change, or delete nucleotide sequences at target sites of interest, prime editors can also be used to construct highly programmable libraries, as well as to conduct cell data recording and lineage tracing studies. In these various uses, there may be as described herein particular design aspects pertaining to the preparation of a PEgRNA that is particularly useful for any given of these applications.


When designing a PEgRNA for any particular application or use of prime editing, a number of considerations may be taken into account, which include, but are not limited to:

    • (a) the target sequence, i.e., the nucleotide sequence in which one or more nucleobase modifications are desired to be installed by the prime editor;
    • (b) the location of the cut site within the target sequence, i.e., the specific nucleobase position at which the prime editor will induce a single-stand nick to create a 3′ end RT primer sequence on one side of the nick and the 5′ end endogenous flap on the other side of the nick (which ultimately is removed by FEN1 or equivalent thereto and replaced by the 3′ ssDNA flap. The cut site is analogous to the “edit location” since this what creates the 3′ end RT primer sequence which becomes extended by the RT during RNA-depending DNA polymerization to create the 3′ ssDNA flap containing the desired edit, which then replaces the 5′ endogenous DNA flap in the target sequence.
    • (c) the available PAM sequences (including the canonical SpCas9 PAM sites, as well as non-canonical PAM sites recognized by Cas9 variants and equivalents with expanded or differing PAM specificities);
    • (d) the spacing between the available PAM sequences and the location of the cut site in the target sequence;
    • (e) the particular Cas9, Cas9 variant, or Cas9 equivalent of the prime editor being used;
    • (f) the sequence and length of the primer binding site;
    • (g) the sequence and length of the edit template;
    • (h) the sequence and length of the homology arm;
    • (i) the spacer sequence and length; and
    • (j) the core sequence.


The instant disclosure discusses these aspects above.


In one embodiment, an approach to designing a suitable PEgRNA, and optionally a nicking-sgRNA design guide for second-site nicking, is hereby provided. This embodiment provides a step-by-step set of instructions for designing PEgRNAs and nicking-sgRNAs for prime editing which takes into account one or more of the above considerations. The steps reference the examples shown in FIGS. 70A-70I.

    • 1. Define the target sequence and the edit. Retrieve the sequence of the target DNA region (˜200 bp) centered around the location of the desired edit (point mutation, insertion, deletion, or combination thereof). See FIG. 70A.
    • 2. Locate target PAMs. Identify PAMs in the proximity to the desired edit location. PAMs can be identified on either strand of DNA proximal to the desired edit location. While PAMs close to the edit position are preferred (i.e., wherein the nick site is less than 30 nt from the edit position, or less than 29 nt, 28 nt, 27 nt, 26 nt, 25 nt, 24 nt, 23 nt, 22 nt, 21 nt, 20 nt, 19 nt, 18 nt, 17 nt, 16 nt, 15 nt, 14 nt, 13 nt, 12 nt, 11 nt, 10 nt, 9 nt, 8 nt, 7 nt, 6 nt, 5 nt, 4 nt, 3 nt, or 2 nt from the edit position to the nick site), it is possible to install edits using protospacers and PAMs that place the nick ≥30 nt from the edit position. See FIG. 70B.
    • 3. Locate the nick sites. For each PAM being considered, identify the corresponding nick site and on which strand. For Sp Cas9 H840A nickase, cleavage occurs in the PAM-containing strand between the 3rd and 4th bases 5′ to the NGG PAM. All edited nucleotides must exist 3′ of the nick site, so appropriate PAMs must place the nick 5′ to the target edit on the PAM-containing strand. In the example shown below, there are two possible PAMs. For simplicity, the remaining steps will demonstrate the design of a PEgRNA using PAM 1 only. See FIG. 70C.
    • 4. Design the spacer sequence. The protospacer of Sp Cas9 corresponds to the 20 nucleotides 5′ to the NGG PAM on the PAM-containing strand. Efficient Pol III transcription initiation requires a G to be the first transcribed nucleotide. If the first nucleotide of the protospacer is a G, the spacer sequence for the PEgRNA is simply the protospacer sequence. If the first nucleotide of the protospacer is not a G, the spacer sequence of the PEgRNA is G followed by the protospacer sequence. See FIG. 70D.
    • 5. Design a primer binding site (PBS). Using the starting allele sequence, identify the DNA primer on the PAM-containing strand. The 3′ end of the DNA primer is the nucleotide just upstream of the nick site (i.e. the 4th base 5′ to the NGG PAM for Sp Cas9). As a general design principle for use with PE2 and PE3, a PEgRNA primer binding site (PBS) containing 12 to 13 nucleotides of complementarity to the DNA primer can be used for sequences that contain ˜40-60% GC content. For sequences with low GC content, longer (14- to 15-nt) PBSs should be tested. For sequences with higher GC content, shorter (8- to 11-nt) PBSs should be tested. Optimal PBS sequences should be determined empirically, regardless of GC content. To design a length-p PBS sequence, take the reverse complement of the first p nucleotides 5′ of the nick site in the PAM-containing strand using the starting allele sequence. See FIG. 70E.
    • 6. Design an RT template (or DNA synthesis template). The RT template (or DNA synthesis template where the polymerase is not reverse transcriptase) encodes the designed edit and homology to the sequence adjacent to the edit. In one embodiment, these regions correspond to the DNA synthesis template of FIG. 3D and FIG. 3E, wherein the DNA synthesis template comprises the “edit template” and the “homology arm.” Optimal RT template lengths vary based on the target site. For short-range edits (positions +1 to +6), it is recommended to test a short (9 to 12 nt), a medium (13 to 16 nt), and a long (17 to 20 nt) RT template. For long-range edits (positions +7 and beyond), it is recommended to use RT templates that extend at least 5 nt (preferably 10 or more nt) past the position of the edit to allow for sufficient 3′ DNA flap homology. For long-range edits, several RT templates should be screened to identify functional designs. For larger insertions and deletions (≥5 nt), incorporation of greater 3′ homology (˜20 nt or more) into the RT template is recommended. Editing efficiency is typically impaired when the RT template encodes the synthesis of a G as the last nucleotide in the reverse transcribed DNA product (corresponding to a C in the RT template of the PEgRNA). As many RT templates support efficient prime editing, avoidance of G as the final synthesized nucleotide is recommended when designing RT templates. To design a length-r RT template sequence, use the desired allele sequence and take the reverse complement of the first r nucleotides 3′ of the nick site in the strand that originally contained the PAM. Note that compared to SNP edits, insertion or deletion edits using RT templates of the same length will not contain identical homology. See FIG. 70F.
    • 7. Assemble the full PEgRNA sequence. Concatenate the PEgRNA components in the following order (5′ to 3′): spacer, scaffold, RT template and PBS. See FIG. 70G.
    • 8. Designing nicking-sgRNAs for PE3. Identify PAMs on the non-edited strand upstream and downstream of the edit. Optimal nicking positions are highly locus-dependent and should be determined empirically. In general, nicks placed 40 to 90 nucleotides 5′ to the position across from the PEgRNA-induced nick lead to higher editing yields and fewer indels. A nicking sgRNA has a spacer sequence that matches the 20-nt protospacer in the starting allele, with the addition of a 5′-G if the protospacer does not begin with a G. See FIG. 70H.
    • 9. Designing PE3b nicking-sgRNAs. If a PAM exists in the complementary strand and its corresponding protospacer overlaps with the sequence targeted for editing, this edit could be a candidate for the PE3b system. In the PE3b system, the spacer sequence of the nicking-sgRNA matches the sequence of the desired edited allele, but not the starting allele. The PE3b system operates efficiently when the edited nucleotide(s) falls within the seed region (˜10 nt adjacent to the PAM) of the nicking-sgRNA protospacer. This prevents nicking of the complementary strand until after installation of the edited strand, preventing competition between the PEgRNA and the sgRNA for binding the target DNA. PE3b also avoids the generation of simultaneous nicks on both strands, thus reducing indel formation significantly while maintaining high editing efficiency. PE3b sgRNAs should have a spacer sequence that matches the 20-nt protospacer in the desired allele, with the addition of a 5′ G if needed. See FIG. 70I.


The above step-by-step process for designing a suitable PEgRNA and a second-site nicking sgRNA is not meant to be limiting in any way. The disclosure contemplates variations of the above-described step-by-step process which would be derivable therefrom by a person of ordinary skill in the art.


[7] Applications Utilizing Multi-Flap Prime Editing

Dual-flap and quadruple-flap prime editing (i.e., multi-flap prime editing) have many potential applications, such as installing peptide tags, RNA tags, immunoepitopes, dimerization domains, and recombinase target sites. One such application of dual-flap prime editing is the installation of recombinase or integrase sequences at user-specified locations in the genome. FIG. 93 illustrates the installation of Bxb1 recombinase attB (38 bp) and attP (50 bp) sites into a targeted region of the human genome (HEK293T site 3, or HEK3) with simultaneous deletion of 90 bp of intervening sequence between the two nick sites. Various degrees of complementarity between 3′ flaps allow for successful editing, though longer sequences of complementarity produce more favorable ratios of desired edits to indels. Other dual-flap prime editing applications include endogenous tagging of genes with peptide or protein sequences, or the replacement of exons with new DNA sequences that have the potential to substitute for multiple variants for which the mutation falls within the exon sequence.


Dual-flap prime editing can be used to introduce one or two recombinase sites at targeted positions in the human genome. If single recombinase sites are inserted, these can be used as landing sites for a recombinase-mediated reaction between the genomic recombinase site and a second recombinase site within an exogenously supplied DNA, such as a plasmid. This enables the targeted integration of DNA cargo. If two recombinase sites are inserted in adjacent regions of DNA, depending on the orientation of the recombinase sites, these can be used for recombinase-mediated excision or inversion of the intervening sequence, or for recombinase-mediated cassette exchange with exogenous DNA for cargo integration. Integration of compatible recombinase sites on different chromosomes enables targeted and directional chromosomal translocation. Dual-flap prime editing can be used to efficiently introduce recombinase sites at a number of loci in the human genome (FIG. 94). Thus, the pairing of recombinase site integration by dual-flap prime editing with DNA recombinase enzymes represents a powerful approach for achieving many types of SV edits and target integration of DNA cargo.


The multi-flap prime editors (e.g., the embodiment depicted in FIG. 90) described herein may be used for the precise insertion of new DNA sequence, the precise deletion of endogenous genomic DNA sequence, or the replacement of an endogenous genomic DNA sequence with a new DNA sequence.


For example, and as exemplified and discussed herein, dual prime editing can be used to (a) install mutation-correcting changes to a nucleotide sequence, (b) install protein and RNA tags, (c) installation of immunoepitopes on proteins of interest, (d) install inducible dimerization domains in proteins, (e) install recombinase target sites to direct specific genetic changes. In addition to these methods which, in general, insert, change, or delete nucleotide sequences at target sites of interest, dual prime editors can also be used to construct highly programmable libraries, as well as to conduct cell data recording and lineage tracing studies.


These specific exemplary uses of dual prime editing are in no way intended to be limiting. The present Application contemplates any use for dual prime editing which involves, in general, some form of the installation, deletion, and/or replacement of one or more nucleobases at a target site in a nucleotide sequence, e.g., a genomic DNA.


For any of the exemplified uses for dual prime editing, one may use any prime editor disclosed herein, including PE1, PE2, PE3, and PE3b, or PE-short.


A. Prime Editing Versus Multi-Flap Prime Editing


Classical Prime Editing


In various embodiments, prime editing (or “prime editing”) operates by contacting a target DNA molecule (for which a change in the nucleotide sequence is desired to be introduced) with a nucleic acid programmable DNA binding protein (napDNAbp) complexed with an extended guide RNA. In reference to FIG. 1G, the extended guide RNA comprises an extension at the 3′ or 5′ end of the guide RNA, or at an intramolecular location in the guide RNA and encodes the desired nucleotide change (e.g., single nucleotide change, insertion, or deletion). In step (a), the napDNAbp/extended gRNA complex contacts the DNA molecule and the extended gRNA guides the napDNAbp to bind to a target locus. In step (b), a nick in one of the strands of DNA of the target locus is introduced (e.g., by a nuclease or chemical agent), thereby creating an available 3′ end in one of the strands of the target locus. In certain embodiments, the nick is created in the strand of DNA that corresponds to the R-loop strand, i.e., the strand that is not hybridized to the guide RNA sequence, i.e., the “non-target strand.” The nick, however, could be introduced in either of the strands. That is, the nick could be introduced into the R-loop “target strand” (i.e., the strand hybridized to the protospacer sequence of the extended gRNA) or the “non-target strand” (i.e, the strand forming the single-stranded portion of the R-loop and which is complementary to the target strand). In step (c), the 3′ end of the DNA strand (formed by the nick) interacts with the extended portion of the guide RNA in order to prime reverse transcription (i.e, “target-primed RT”). In certain embodiments, the 3′ end DNA strand hybridizes to a specific RT priming sequence on the extended portion of the guide RNA, i.e, the “reverse transcriptase priming sequence.” In step (d), a reverse transcriptase is introduced (as a fusion protein with the napDNAbp or in trans) which synthesizes a single strand of DNA from the 3′ end of the primed site towards the 5′ end of the extended guide RNA. This forms a single-strand DNA flap comprising the desired nucleotide change (e.g., the single base change, insertion, or deletion, or a combination thereof) and which is otherwise homologous to the endogenous DNA at or adjacent to the nick site. In step (e), the napDNAbp and guide RNA are released. Steps (f) and (g) relate to the resolution of the single strand DNA flap such that the desired nucleotide change becomes incorporated into the target locus. This process can be driven towards the desired product formation by removing the corresponding 5′ endogenous DNA flap (e.g., by FEN1 or similar enzyme that is provide in trans, as a fusion with the prime editor, or endogenously provided) that forms once the 3′ single strand DNA flap invades and hybridizes to the endogenous DNA sequence. Without being bound by theory, the cells endogenous DNA repair and replication processes resolves the mismatched DNA to incorporate the nucleotide change(s) to form the desired altered product. The process can also be driven towards product formation with “second strand nicking,” as exemplified in FIG. 1G, or “termporal second strand nicking,” as exemplified in FIG. 1I and discussed herein.


The process of prime editing may introduce at least one or more of the following genetic changes: transversions, transitions, deletions, and insertions. In addition, prime editing may be implemented for specific applications. For example, and as exemplied and discussed herein, prime editing can be used to (a) install mutation-correcting changes to a nucleotide sequence, (b) install protein and RNA tags, (c) installation of immunoepitopes on proteins of interest, (d) install inducible dimerization domains in proteins, (e) install or remove sequences to alter that activity of a biomolecule, (f) install recombinase target sites to direct specific genetic changes, and (g) mutagenesis of a target sequence by using an error-prone RT. In addition to these methods which, in general, insert, change, or delete nucleotide sequences at target sites of interest, prime editors can also be used to construct highly programmable libraries, as well as to conduct cell data recording and lineage tracing studies. The inventors have also contemplated additional design features of PEgRNAs that are aimed to improve the efficacy of prime editing. Still further, the inventors have conceived of methods for successfully delivering prime editors using vector delivery systems and which involve splitting the napDNAbp using intein domains.


The term “prime editing system” or “prime editor (PE)” refers the compositions involved in the method of genome editing using target-primed reverse transcription (TPRT) describe herein, including, but not limited to the napDNAbps, reverse transcriptases, fusion proteins (e.g., comprising napDNAbps and reverse transcriptases), extended guide RNAs, and complexes comprising fusion proteins and extended guide RNAs, as well as accessory elements, such as second strand nicking components and 5′ endogenous DNA flap removal endonucleases (e.g., FEN1) for helping to drive the prime editing process towards the edited product formation.


In another embodiment, the schematic of FIG. 3F depicts the interaction of a typical PEgRNA with a target site of a double stranded DNA and the concomitant production of a 3′ single stranded DNA flap containing the genetic change of interest. The double strand DNA is shown with the top strand in the 3′ to 5′ orientation and the lower strand in the 5′ to 3′ direction. The top strand comprises the “protospacer” and the PAM sequence and is referred to as the “target strand.” The complementary lower strand is referred to as the “non-target strand.” Although not shown, the PEgRNA depicted would be complexed with a Cas9 or equivalent. As shown in the schematic, the spacer of the PEgRNA anneals to a complementary region on the target strand, which is referred to as the protospacer, which is located just downstream of the PAM sequence is approximately 20 nucleotides in length. This interaction forms as DNA/RNA hybrid between the spacer RNA and the protospacer DNA, and induces the formation of an R loop in the region opposite the protospacer. As taught elsewhere herein, the Cas9 protein (not shown) then induces a nick in the non-target strand, as shown. This then leads to the formation of the 3′ ssDNA flap region which, in accordance with *z*, interacts with the 3′ end of the PEgRNA at the primer binding site. The 3′ end of the ssDNA flap (i.e., the reverse transcriptase primer sequence) anneals to the primer binding site (A) on the PEgRNA, thereby priming reverse transcriptase. Next, reverse transcriptase (e.g., provided in trans or provided cis as a fusion protein, attached to the Cas9 construct) then polymerizes a single strand of DNA which is coded for by the edit template (B) and homology arm (C). The polymerization continues towards the 5′ end of the extension arm. The polymerized strand of ssDNA forms a ssDNA 3′ end flap which, as describe elsewhere (e.g., as shown in FIG. 1G), invades the endogenous DNA, displacing the corresponding endogenous strand (which is removed as a 5′ DNA flap of endogenous DNA), and installing the desired nucleotide edit (single nucleotide base pair change, deletions, insertions (including whole genes) through naturally occurring DNA repair/replication rounds.


This application of prime editing can be further described in Example 1.


Dual Prime Editing


This Specification describes a dual prime editing system (or a dual-flap prime editing system) that addresses the challenges associated with flap equilibration and subsequent incorporation of the edit into the non-edited complementary genomic DNA strand by simultaneously editing both DNA strands. In the dual-flap prime editing system, two pegRNAs are used to target opposite strands of a genomic site and direct the synthesis of two complementary 3′ flaps containing edited DNA sequence (FIG. 91). Unlike classical prime editing, there is no requirement for the pair of edited DNA strands (3′ flaps) to directly compete with 5′ flaps in endogenous genomic DNA, as the complementary edited strand is available for hybridization instead. Since both strands of the duplex are synthesized as edited DNA, the dual-flap prime editing system obviates the need for the replacement of the non-edited complementary DNA strand required by classical prime editing. Instead, cellular DNA repair machinery need only excise the paired 5′ flaps (original genomic DNA) and ligate the paired 3′ flaps (edited DNA) into the locus. Therefore, there is also no need to include sequences homologous to genomic DNA in the newly synthesized DNA strands, allowing selective hybridization of the new strands and facilitating edits that contain minimal genomic homology. Nuclease-active versions of prime editors that cut both strands of DNA could also be used to accelerate the removal of the original DNA sequence. Accordingly, in certain embodiments, a dual flap prime editing system involves a pair of newly synthesized DNA strands (e.g. 3′ flaps) that share homology with the endogeneous DNA sequence at a target site to be edited. In some embodiments, a dual flap prime editing system involves a pair of newly synthesized DNA strands, where at least one of the newly synthesized DNA strands does not share homology with the endogeneous DNA sequence at the target site to be edited. In some embodiments, one of the newly synthesized DNA strands comprises homology with the endogenous DNA sequence at the target site and not with the other newly synthesized DNA strand. In some embodiments, each of the newly synthesized DNA strands comprises homology with the endogenous DNA sequence at the target site and not with the other newly synthesized DNA strand. For example, a newly synthesized 3′ flap encoded by one of the dual-flap pegRNAs may comprise a region of complementarity to a protospacer sequence of the other dual-flap pegRNA. Accordingly, in some embodiments, a pair of dual-flap pegRNAs each having complementarity to a spacer sequence of the other pegRNA may result in deletion of the endogenous DNA sequence positioned between protospacer sequences of the pair of dual-flap pegRNAs. In some embodiments, a dual flap prime editing system involves a pair of newly synthesized DNA strands, where neither of the newly synthesized DNA strands share homology with the endogeneous DNA sequence at the target site to be edited. Rather, the two newly synthesized DNA strands (e.g. 3′ flaps) each comprises a region of complementarity to each other and may form a duplex by the complementarity. A desired edited portion as compared to the endogeneous DNA sequence target site to be edited in the duplex may then be incorporated at the target site. Like classical prime editing, dual prime editing is a versatile and precise genome editing method that directly writes new genetic information into a specified DNA site using a nucleic acid programmable DNA binding protein (“napDNAbp”) working in association with a polymerase (i.e., in the form of a fusion protein or otherwise provided in trans with the napDNAbp), wherein the prime editing system is programmed with a prime editing (PE) guide RNA (“PEgRNA”) that both specifies the target site and templates the synthesis of the desired edit in the form of a replacement DNA strand by way of an extension (either DNA or RNA) engineered onto a guide RNA (e.g., at the 5′ or 3′ end, or at an internal portion of a guide RNA). The replacement strand containing the desired edit (e.g., a single nucleobase substitution) shares the same sequence as the endogenous strand of the target site to be edited (with the exception that it includes the desired edit). Through DNA repair and/or replication machinery, the endogenous strand of the target site is replaced by the newly synthesized replacement strand containing the desired edit. In some cases, prime editing may be thought of as a “search-and-replace” genome editing technology since the prime editors, as described herein, not only search and locate the desired target site to be edited, but at the same time, encode a replacement strand containing a desired edit which is installed in place of the corresponding target site endogenous DNA strand.


B. Use of Dual Prime Editing for Peptide Tagging


In another aspect, the disclosure provides a method of using the herein described dual prime editors for genetically grafting one or more peptide tags onto a protein using prime editing (such as the embodiment shown in FIG. 90). More in particular, the disclosure provides a method for genetically installing one or more peptide tags onto a protein comprising: contacting a target nucleotide sequence encoding the protein with a prime editor configured to insert therein a second nucleotide sequence encoding the one or more peptide tags to result in a recombinant nucleotide sequence that encodes a fusion protein comprising the protein fused to the protein tag.


In other embodiments, the disclosure provides a method for making a fusion protein comprising a peptide of interest and one or more peptide tags, the method comprising: contacting a target nucleotide sequence encoding the protein with a prime editor configured to insert therein a second nucleotide sequence encoding the one or more peptide tags to result in a recombinant nucleotide sequence that encodes the fusion protein comprising the protein fused to the protein tag.


In various embodiments, the target nucleotide sequence is a specific gene of interest in a genomic DNA. The gene of interest may encode a protein of interest (e.g., a receptor, an enzyme, a therapeutic protein, a membrane protein, a transport protein, a signal transduction protein, or an immunological protein, etc.). The gene of interest may also encode an RNA molecule, including, but not limited to, messenger RNA (mRNA), transfer RNA (tRNA), ribosomal RNA (rRNA), small nuclear RNA (snRNA), antisense RNA, guide RNA, microRNA (miRNA), small interfering RNA (siRNA), and cell-free RNA (cfRNA).


The peptide tag may be any peptide tag or variant thereof which imparts one or more functions onto a protein for purposes such as separation, purification, visualization, solubilization, or detection. The peptides tags can include “affinity tags” (to facilitate protein purification), “solubilization tags” (to assist in proper folding of proteins), “chromatography tags” (to alter chromatographic properties of proteins), “epitope tags” (to bind to high affinity antibodies), and “fluorescence tags” (to facilitate visualization of proteins in a cell or in vitro). Examples of peptide tags include, but are not limited to the following tags:














NAME
AMINO ACID SEQUENCE
SEQ ID NO:







AVITAG™
GLNDIFEAQKIEWHE
SEQ ID NO: 245





C-TAG
EPEA
SEQ ID NO: 246





CALMODULIN-
KRRWKKNFIAVSAAN
SEQ ID NO: 247


TAG
RFKKISSSGAL






POLYGLUTAMATE
EEEEEE
SEQ ID NO: 248


TAG







E-TAG
GAPVPYPDPLEPR
SEQ ID NO: 249





FLAG-TAG
DYKDDDDK
SEQ ID NO: 250





HA-TAG
YPYDVPDYA
SEQ ID NO: 251





HIS-TAG
H (HIS1)
SEQ ID NO: 252



HH (HIS2)
SEQ ID NO: 253



HHH (HIS3)
SEQ ID NO: 254



HHHH (HIS4)
SEQ ID NO: 255



HHHHH (HIS5)
SEQ ID NO: 256



HHHHHH (HIS6)
SEQ ID NO: 257



HHHHHHH (HIS7)
SEQ ID NO: 258



HHHHHHHH (HIS8)
SEQ ID NO: 259



HHHHHHHHH (HIS9)
SEQ ID NO: 260



HHHHHHHHHH (HIS10)
SEQ ID NO: 261



HHHHHHHHHH . . .
SEQ ID NO: 262



H . . .(HISN, 




WHEREIN N = 1-25)






MYC-TAG
EQKLISEEDL
SEQ ID NO: 263





NE-TAG
TKENPRSNQEESYDDNES
SEQ ID NO: 264





RHO1D4-TAG
TETSQVAPA
SEQ ID NO: 265





S-TAG
KETAAAKFERQHMDS
SEQ ID NO: 266





SBP-TAG
MDEKTTGWRGGHVVEGLAG
SEQ ID NO: 267



ELEQLRARLEHHPQGQREP






SOFTAG-1
SLAELLNAGLGGS
SEQ ID NO: 268





SOFTAG-2
TQDPSRVG
SEQ ID NO: 269





SPOT-TAG
PDRVRAVSHWSS
SEQ ID NO: 270





STREP-TAG
WSHPQFEK
SEQ ID NO: 271





TC TAG
CCPGCC
SEQ ID NO: 272





TY TAG
EVHTNQDPLD
SEQ ID NO: 273





V5 TAG
GKPIPNPLLGLDST
SEQ ID NO: 274





VSV-TAG
YTDIEMNRLGK
SEQ ID NO: 275





XPRESS TAG
DLYDDDDK
SEQ ID NO: 276









Peptide tags may also be the following affinity tags (for separation and/or purification of proteins) (as described in Table 9.9.1 of Kimple et al., “Overview of Affinity Tags for Protein Purification,” Curr Protoc Protein Sci, 2013, 73:Unit-9.9, which is incorporated herein by reference).















AMINO ACID



NAME
SEQUENCE







AU1 EPITOPE
DTYRYI
SEQ ID NO: 278





AU5 EPITOPE
TDFYLK
SEQ ID NO: 279





BACTERIOPHAGE T7
MASMTGGQQMG
SEQ ID NO: 280


EPITOPE (T7-TAG)







BLUETONGUE VIRUS
QYPALT
SEQ ID NO: 281


TAG (B-TAG)







E2 EPITOPE
SSTSSDFRDR
SEQ ID NO: 282





HISTIDINE AFFINITY
KDHLIHNVHK
SEQ ID NO: 283


TAG (HAT)
EFHAHAHNK






HSV EPITOPE
QPELAPED
SEQ ID NO: 284





POLYARGININE
RRRRR
SEQ ID NO: 285


(ARG-TAG)







POLYASPARTATE
CCCC
SEQ ID NO: 286


(ASP-TAG)







POLYPHENYLALANINE
FFFFFFFFFFF
SEQ ID NO: 287


(PHE-TAG)







S1-TAG
NANNPDWDF
SEQ ID NO: 288





S-TAG
KETAAAKFE
SEQ ID NO: 289



RQHMDS






VSV-G
YTDIEMNRLGK
SEQ ID NO: 290









In particular embodiments, the peptide tags may include a His6 tag, FLAG-tag, V5-tag, GCN4-tag, HA-tag, Myc-Tag, FIAsH/ReAsH-tag, Sortase substrate, pi-clamp.


In various embodiments, the peptide tags may be used for applications that include protein fluorescent labeling, immunoprecipitation, immunoblotting, immunohistochemistry, protein recruitment, inducible protein degrons, and genome-wide screening.


In various other embodiments, the peptide tag may include an intein sequence to install protein self-splicing function. As used herein, the term “intein” refers to auto-processing polypeptide domains found in organisms from all domains of life. An intein (intervening protein) carries out a unique auto-processing event known as protein splicing in which it excises itself out from a larger precursor polypeptide through the cleavage of two peptide bonds and, in the process, ligates the flanking extein (external protein) sequences through the formation of a new peptide bond. This rearrangement occurs post-translationally (or possibly co-translationally), as intein genes are found embedded in frame within other protein-coding genes. Furthermore, intein-mediated protein splicing is spontaneous; it requires no external factor or energy source, only the folding of the intein domain. This process is also known as cis-protein splicing, as opposed to the natural process of trans-protein splicing with “split inteins.” Inteins are the protein equivalent of the self-splicing RNA introns (see Perler et al., Nucleic Acids Res. 22:1125-1127 (1994)), which catalyze their own excision from a precursor protein with the concomitant fusion of the flanking protein sequences, known as exteins (reviewed in Perler et al., Curr. Opin. Chem. Biol. 1:292-299 (1997); Perler, F. B. Cell 92(1):1-4 (1998); Xu et al., EMBO J. 15(19):5146-5153 (1996)).


The mechanism of the protein splicing process has been studied in great detail (Chong, et al., J. Biol. Chem. 1996, 271, 22159-22168; Xu, M-Q & Perler, F. B. EMBO Journal, 1996, 15, 5146-5153) and conserved amino acids have been found at the intein and extein splicing points (Xu, et al., EMBO Journal, 1994, 13 5517-522).


Inteins can also exist as two fragments encoded by two separately transcribed and translated genes. These so-called split inteins self-associate and catalyze protein-splicing activity in trans. Split inteins have been identified in diverse cyanobacteria and archaea (Caspi et al, Mol Microbiol. 50: 1569-1577 (2003); Choi J. et al, J Mol Biol. 556: 1093-1106 (2006.); Dassa B. et al, Biochemistry. 46:322-330 (2007.); Liu X. and Yang J., J Biol Chem. 275:26315-26318 (2003); Wu H. et al. Proc Natl Acad Sci USA. £5:9226-9231 (1998.); and Zettler J. et al, FEBS Letters. 553:909-914 (2009)), but have not been found in eukaryotes thus far. Recently, a bioinformatic analysis of environmental metagenomic data revealed 26 different loci with a novel genomic arrangement. At each locus, a conserved enzyme coding region is interrupted by a split intein, with a freestanding endonuclease gene inserted between the sections coding for intein subdomains. Among them, five loci were completely assembled: DNA helicases (gp41-1, gp41-8); Inosine-5′-monophosphate dehydrogenase (IMPDH-1); and Ribonucleotide reductase catalytic subunits (NrdA-2 and NrdJ-1). This fractured gene organization appears to be present mainly in phages (Dassa et al, Nucleic Acids Research. 57:2560-2573 (2009)).


In certain embodiments, the prime editors described herein can be used to insert split-intein tags in two different proteins, causing their intracellular ligation when co-expressed to form a fusion protein. In protein trans-splicing, one precursor protein consists of an N-extein part followed by the N-intein, another precursor protein consists of the C-intein followed by a C-extein part, and a trans-splicing reaction (catalyzed by the N- and C-inteins together) excises the two intein sequences and links the two extein sequences with a peptide bond. Protein trans-splicing, being an enzymatic reaction, can work with very low (e.g., micromolar) concentrations of proteins and can be carried out under physiological conditions.


The split intein Npu DnaE was characterized as having the highest rate reported for the protein trans-splicing reaction. In addition, the Npu DnaE protein splicing reaction is considered robust and high-yielding with respect to different extein sequences, temperatures from 6 to 37° C., and the presence of up to 6M Urea (Zettler J. et al, FEBS Letters. 553:909-914 (2009); Iwai I. et al, FEBS Letters 550: 1853-1858 (2006)). As expected, when the Cysl Ala mutation at the N-domain of these inteins was introduced, the initial N to S-acyl shift and therefore protein splicing was blocked. Unfortunately, the C-terminal cleavage reaction was also almost completely inhibited. The dependence of the asparagine cyclization at the C-terminal splice junction on the acyl shift at the N-terminal scissile peptide bond seems to be a unique property common to the naturally split DnaE intein alleles (Zettler J. et al. FEBS Letters. 555:909-914 (2009)).


Protein trans-splicing, catalyzed by split inteins, provides an entirely enzymatic method for protein ligation. A split-intein is essentially a contiguous intein (e g a mini-intein) split into two pieces named N-intein and C-intein, respectively. The N-intein and C-intein of a split intein can associate non-covalently to form an active intein and catalyze the splicing reaction essentially in same way as a contiguous intein does. Split inteins have been found in nature and also engineered in laboratories. As used herein, the term “split intein” refers to any intein in which one or more peptide bond breaks exists between the N-terminal and C-terminal amino acid sequences such that the N-terminal and C-terminal sequences become separate molecules that can non-covalently reassociate, or reconstitute, into an intein that is functional for trans-splicing reactions. Any catalytically active intein, or fragment thereof, may be used to derive a split intein for use in the methods of the invention. For example, in one aspect the split intein may be derived from a eukaryotic intein. In another aspect, the split intein may be derived from a bacterial intein. In another aspect, the split intein may be derived from an archaeal intein. Preferably, the split intein so-derived will possess only the amino acid sequences essential for catalyzing trans-splicing reactions.


Split inteins may be created from contiguous inteins by engineering one or more split sites in the unstructured loop or intervening amino acid sequence between the −12 conserved beta-strands found in the structure of mini-inteins. Some flexibility in the position of the split site within regions between the beta-strands may exist, provided that creation of the split will not disrupt the structure of the intein, the structured beta-strands in particular, to a sufficient degree that protein splicing activity is lost.


The prime editors described herein may incorporate peptide tags (including inteins) into the C-terminal end of a protein of interest. In other embodiments, the peptide tags (including inteins) may be incorporated into the N-terminal end of a protein of interest. The peptide tags may also be incorporated into the interior of a protein of interest. The resulting fusion proteins created by the herein described prime editors may have the following structures:

    • [protein of interest]-[peptide tag];
    • [peptide tag]-[protein of interest]; or
    • [protein of interest-N-terminal region]-[peptide tag]-[protein of interest—C-terminal region].


The principles of guide RNA design for use in peptide tagging throughout may be applied to peptide tagging. For example, in one embodiment, the PEgRNA structure for peptide tagging may have the following structure: 5′-[spacer sequence]-[gRNA core or scaffold]-[extension arm]-3′, wherein the extension arm comprises in the 5′ to 3′ direction, a homology arm, edit template (comprising the sequence that encodes the peptide tag), and a primer binding site. This configuration is depicted in FIG. 3D and in FIG. 24.


In another embodiment, the PEgRNA structure for peptide tagging may have the following structure: 5′-[extension arm]-[spacer sequence]-[gRNA core or scaffold]-3′, wherein the extension arm comprises in the 5′ to 3′ direction, a homology arm, edit template (comprising the sequence that encodes the peptide tag), and a primer binding site. This configuration is depicted in FIG. 3E.


Embodiments of peptide tagging using prime editing is depicted in FIGS. 25 and 26 and described in Example 4.


C. Use of Dual Prime Editing for RNA Tagging


Dual prime editing may also be used to manipulate, alter, and otherwise modify the sequences of DNA encoding RNA functions through RNA tagging, and in this way provides a means to indirectly modify the structure and function of RNA. For example, dual PE can be used to insert motifs that are functional at the RNA level (hereafter RNA motifs) to tag or otherwise manipulate non-coding RNAs or mRNAs. These motifs could serve to increase gene expression, decrease gene expression, alter splicing, change post-transcriptional modification, affect the sub-cellular location of the RNA, enable isolation or determination of the intra- or extra-cellular location of the RNA (using, for instance, fluorescent RNA aptamers such as Spinach, Spinach2, Baby Spinach, or Broccoli), recruit endogenous or exogenous protein or RNA binders, introduce sgRNAs, or induce processing of the RNA, by either self-cleavage or RNAses (see FIG. 28B and Example 6 for further details).


The following RNA tags or motifs may be inserted into a gene of interest using dual prime editing (e.g., with the dual PE methodology of FIG. 90) with an appropriate PEgRNA (designed using the guidance provided herein) to affect various properties of RNA, including RNA transport, expression level, splicing, and detection.


















EXEMPLARY PEGRNA





FOR PRIME EDITING





INSERTION OF RNA





MOTIF INTO THE




FUNCTION/
EXEMPLARY HEXA


RNA MOTIF
SEQUENCE OF RNA MOTIF
EFFECT
GENE*







POLYOMAVIRUS
AACTTGTTTATTGCAGCTTAT
TERMINATION
ATCCTTCCAGTCAGG


SIMIAN
AATGGTTACAAATAAAGCAA
OF
GCCATGTTTGAGAGC


VIRUS 40
TAGCATCACAAATTTCACAA
TRANSCRIPTION

TAGAAATAGCAAGTT



(SV40) TYPE1
ATAAAGCATTTTTTTCACTGC
OF THE

TAAATAAGGCTAGTC




ATTCTAGTTGTGGTTTGTCCA
TAGGED

CGTTATCAACTTGAA




AACTCATCAATGTATCTTA
GENE;

AAAGTGGGACCGAG




(SEQ ID NO: 331)
TRANSPORT

TCGGTCC
ACCTGAACC





OF MRNA INTO

GTATATC
TAAGATACA





CYTOSOL;

TTGATGAGTTTGGA





INCREASED

CAAACCACAACTAG





RNA

AATGCAGTGAAAAA





STABILITY

AATGCTTTATTTGTG





AND

AAATTTGTGATGCTA





EXPRESSION

TTGCTTTATTTGTAA





OF ENCODED

CCATTATAAGCTGCA





PROTEIN

ATAAACAAGTTCTAT






GGCCCTGACTGGAA





(SEQ ID NO: 332)





POLYOMAVIRUS
CCATGGCCCAACTTGTTTATT
TERMINATION
ATCCTTCCAGTCAGG


SIMIAN
GCAGCTTATAATGGTTACAA
OF
GCCATGTTTGAGAGC


VIRUS 40
ATAAAGCAATAGCATCACAA
TRANSCRIPTION

TAGAAATAGCAAGTT



(SV40) TYPE2
ATTTCACAAATAAAGCATTTT
OF THE

TAAATAAGGCTAGTC




TTTCACTGCATTCTAGTTGTG
TAGGED

CGTTATCAACTTGAA




GTTTGTCCAAACTCATCAAT
GENE;

AAAGTGGGACCGAG




GTATCTTATCATGTCTGGATC
TRANSPORT

TCGGTCC
ACCTGAACC




TC (SEQ ID NO: 333)
OF MRNA INTO

GTATATC
GAGATCCA





CYTOSOL;

GACATGATAAGATAC





INCREASED

ATTGATGAGTTTGG





RNA

ACAAACCACAACTA





STABILITY

GAATGCAGTGAAAA





AND

AAATGCTTTATTTGT





EXPRESSION

GAAATTTGTGATGC





OF ENCODED

TATTGCTTTATTTGT





PROTEIN

AACCATTATAAGCTG







CAATAAACAAGTTG







GGCCATGGCTATGGC






CCTGACTGGAA





(SEQ ID NO: 334)





POLYOMAVIRUS
TGATCATAATCAAGCCATATC
TERMINATION
ATCCTTCCAGTCAGG


SIMIAN
ACATCTGTAGAGGTTTACTT
OF
GCCATGTTTGAGAGC


VIRUS 40
GCTTTAAAAAACCTCCACAC
TRANSCRIPTION

TAGAAATAGCAAGTT



(SV40) TYPE3
CTCCCCCTGAACCTGAAACA
OF THE

TAAATAAGGCTAGTC




TAAAATGAATGCAATTGTTG
TAGGED

CGTTATCAACTTGAA




TTGTTAACTTGTTTATTGCAG
GENE;

AAAGTGGGACCGAG




CTTATAATGGTTACAAATAAA
TRANSPORT

TCGGTCC
ACCTGAACC




GCAATAGCATCACAAATTTC
OF MRNA INTO

GTATATC
GCAGATCC




ACAAATAAAGCATTTTTTTC
CYTOSOL;

AGACATGATAAGATA




ACTGCATTCTAGTTGTGGTTT
INCREASED

CATTGATGAGTTTG




GTCCAAACTCATCAATGTAT
RNA

GACAAACCACAACT




CTTATCATGTCTGGATCTGC
STABILITY

AGAATGCAGTGAAA




(SEQ ID NO: 335)
AND

AAAATGCTTTATTTG





EXPRESSION

TGAAATTTGTGATG





OF ENCODED

CTATTGCTTTATTTG





PROTEIN

TAACCATTATAAGCT







GCAATAAACAAGTT







AACAACAACAATTG







CATTCATTTTATGTT







TCAGGTTCAGGGGG







AGGTGTGGAGGTTT







TTTAAAGCAAGTAA







ACCTCTACAGATGT







GATATGGCTTGATTA







TGATCACTATGGCCC







TGACTGGAA






(SEQ ID NO: 336)





HUMAN
ACGGGTGGCATCCCTGTGAC
TRANSPORT
ATCCTTCCAGTCAGG


GROWTH
CCCTCCCCAGTGCCTCTCCT
OF RNA INTO
GCCATGTTTGAGAGC


HORMONE
GGCCCTGGAAGTTGCCACTC
CYTOPLASM;

TAGAAATAGCAAGTT



(HGH)
CAGTGCCCACCAGCCTTGTC
ENHANCED

TAAATAAGGCTAGTC




CTAATAAAATTAAGTTGCATC
RNA

CGTTATCAACTTGAA




ATTTTGTCTGACTAGGTGTC
STABILITY

AAAGTGGGACCGAG




CTTCTATAATATTATGGGGTG
AND

TCGGTCC
ACCTGAACC




GAGGGGGGTGGTATGGAGC
EXPRESSION

GTATATC
AAGGACAG




AAGGGGCAAGTTGGGAAGA
OF ENCODED

GGAAGGGAGCAGT




CAACCTGTAGGGCCTGCGGG
PROTEIN

GGTTCACGCCTGTA




GTCTATTGGGAACCAAGCTG


ATCCCAGCAATTTG




GAGTGCAGTGGCACAATCTT


GGAGGCCAAGGTG




GGCTCACTGCAATCTCCGCC


GGTAGATCACCTGA




TCCTGGGTTCAAGCGATTCT


GATTAGGAGTTGGA




CCTGCCTCAGCCTCCCGAGT


GACCAGCCTGGCCA




TGTTGGGATTCCAGGCATGC


ATATGGTGAAACCC




ATGACCAGGCTCAGCTAATT


CGTCTCTACCAAAA




TTTGTTTTTTTGGTAGAGAC


AAACAAAAATTAGC




GGGGTTTCACCATATTGGCC


TGAGCCTGGTCATG




AGGCTGGTCTCCAACTCCTA


CATGCCTGGAATCC




ATCTCAGGTGATCTACCCAC


CAACAACTCGGGAG




CTTGGCCTCCCAAATTGCTG


GCTGAGGCAGGAGA




GGATTACAGGCGTGAACCAC


ATCGCTTGAACCCA




TGCTCCCTTCCCTGTCCTT


GGAGGCGGAGATTG




(SEQ ID NO: 337)


CAGTGAGCCAAGAT







TGTGCCACTGCACT







CCAGCTTGGTTCCC







AATAGACCCCGCAG







GCCCTACAGGTTGT







CTTCCCAACTTGCC







CCTTGCTCCATACC







ACCCCCCTCCACCC







CATAATATTATAGAA







GGACACCTAGTCAG







ACAAAATGATGCAA







CTTAATTTTATTAGG







ACAAGGCTGGTGGG







CACTGGAGTGGCAA







CTTCCAGGGCCAGG







AGAGGCACTGGGGA







GGGGTCACAGGGAT







GCCACCCGTCTATGG







CCCTGACTGGAA






(SEQ ID NO: 338)





BOVINE
CGACTGTGCCTTCTAGTTGC
TRANSPORT
ATCCTTCCAGTCAGG


GROWTH
CAGCCATCTGTTGTTTGCCC
OF RNA INTO
GCCATGTTTGAGAGC


HORMONE
CTCCCCCGTGCCTTCCTTGA
CYTOPLASM;

TAGAAATAGCAAGTT



(BGH)
CCCTGGAAGGTGCCACTCCC
ENHANCED

TAAATAAGGCTAGTC




ACTGTCCTTTCCTAATAAAAT
RNA

CGTTATCAACTTGAA




GAGGAAATTGCATCGCATTG
STABILITY

AAAGTGGGACCGAG




TCTGAGTAGGTGTCATTCTAT
AND

TCGGTCC
ACCTGAACC




TCTGGGGGGTGGGGTGGGG
EXPRESSION

GTATATC
CCATAGAGC




CAGGACAGCAAGGGGGAGG
OF ENCODED

CCACCGCATCCCCA




ATTGGGAAGACAATAGCAGG
PROTEIN

GCATGCCTGCTATT




CATGCTGGGGATGCGGTGGG


GTCTTCCCAATCCT




CTCTATGG (SEQ ID NO: 339)


CCCCCTTGCTGTCC







TGCCCCACCCCACC







CCCCAGAATAGAAT







GACACCTACTCAGA







CAATGCGATGCAAT







TTCCTCATTTTATTA







GGAAAGGACAGTGG







GAGTGGCACCTTCC







AGGGTCAAGGAAGG







CACGGGGGAGGGG







CAAACAACAGATGG







CTGGCAACTAGAAG







GCACAGTCGCTATG







GCCCTGACTGGAA






(SEQ ID NO: 340)





RABBIT BETA-
TTCACTCCTCAGGTGCAGGC
TRANSPORT
ATCCTTCCAGTCAGG


GLOBIN
TGCCTATCAGAAGGTGGTGG
OF RNA INTO
GCCATGTTTGAGAGC


(RBGLOB)
CTGGTGTGGCCAATGCCCTG
CYTOPLASM;

TAGAAATAGCAAGTT




GCTCACAAATACCACTGAGA
ENHANCED

TAAATAAGGCTAGTC




TCTTTTTCCCTCTGCCAAAA
RNA

CGTTATCAACTTGAA




ATTATGGGGACATCATGAAG
STABILITY

AAAGTGGGACCGAG




CCCCTTGAGCATCTGACTTC
AND

TCGGTCC
ACCTGAACC




TGGCTAATAAAGGAAATTTA
EXPRESSION

GTATATC
GATCTCCAT




TTTTCATTGCAATAGTGTGTT
OF ENCODED

AAGAGAAGAGGGAC




GGAATTTTTTGTGTCTCTCAC
PROTEIN

AGCTATGACTGGGA




TCGGAAGGACATATGGGAGG


GTAGTCAGGAGAGG




GCAAATCATTTAAAACATCA


AGGAAAAATCTGGC




GAATGAGTATTTGGTTTAGA


TAGTAAAACATGTAA




GTTTGGCAACATATGCCCATA


GGAAAATTTTAGGG




TGCTGGCTGCCATGAACAAA


ATGTTAAAGAAAAA




GGTTGGCTATAAAGAGGTCA


AATAACACAAAACA




TCAGTATATGAAACAGCCCC


AAATATAAAAAAAAT




CTGCTGTCCATTCCTTATTCC


CTAACCTCAAGTCA




ATAGAAAAGCCTTGACTTGA


AGGCTTTTCTATGG




GGTTAGATTTTTTTTATATTTT


AATAAGGAATGGAC




GTTTTGTGTTATTTTTTTCTT


AGCAGGGGGCTGTT




TAACATCCCTAAAATTTTCCT


TCATATACTGATGAC




TACATGTTTTACTAGCCAGAT


CTCTTTATAGCCAAC




TTTTCCTCCTCTCCTGACTAC


CTTTGTTCATGGCA




TCCCAGTCATAGCTGTCCCT


GCCAGCATATGGGC




CTTCTCTTATGGAGATC (SEQ


ATATGTTGCCAAACT




ID NO: 341)


CTAAACCAAATACTC







ATTCTGATGTTTTAA







ATGATTTGCCCTCC







CATATGTCCTTCCGA







GTGAGAGACACAAA







AAATTCCAACACAC







TATTGCAATGAAAAT







AAATTTCCTTTATTA







GCCAGAAGTCAGAT







GCTCAAGGGGCTTC







ATGATGTCCCCATAA







TTTTTGGCAGAGGG







AAAAAGATCTCAGT







GGTATTTGTGAGCC







AGGGCATTGGCCAC







ACCAGCCACCACCT







TCTGATAGGCAGCC







TGCACCTGAGGAGT







GAACTATGGCCCTGA







CTGGAA






(SEQ ID NO: 342)





THYMIDINE
GGGGGAGGCTAACTGAAAC
TRANSPORT
ATCCTTCCAGTCAGG


KINASE (TK)
ACGGAAGGAGACAATACCG
OF RNA INTO
GCCATGTTTGAGAGC



GAAGGAACCCGCGCTATGAC
CYTOPLASM;

TAGAAATAGCAAGTT




GGCAATAAAAAGACAGAATA
ENHANCED

TAAATAAGGCTAGTC




AAACGCACGGGTGTTGGGT
RNA

CGTTATCAACTTGAA




CGTTTGTTCATAAACGCGGG
STABILITY

AAAGTGGGACCGAG




GTTCGGTCCCAGGGCTGGCA
AND

TCGGTCC
ACCTGAACC




CTCTGTCGATACCCCACCGA
EXPRESSION

GTATATC
CTATGGCAG




GACCCCATTGGGGCCAATAC
OF ENCODED

GGCCTGCCGCCCCG




GCCCGCGTTTCTTCCTTTTCC
PROTEIN

ACGTTGGCTGCGAG




CCACCCCACCCCCCAAGTTC


CCCTGGGCCTTCAC




GGGTGAAGGCCCAGGGCTC


CCGAACTTGGGGGG




GCAGCCAACGTCGGGGCGG


TGGGGTGGGGAAAA




CAGGCCCTGCCATAG (SEQ


GGAAGAAACGCGG




ID NO: 343)


GCGTATTGGCCCCA







ATGGGGTCTCGGTG







GGGTATCGACAGAG







TGCCAGCCCTGGGA







CCGAACCCCGCGTT







TATGAACAAACGAC







CCAACACCCGTGCG







TTTTATTCTGTCTTT







TTATTGCCGTCATAG







CGCGGGTTCCTTCC







GGTATTGTCTCCTTC







CGTGTTTCAGTTAG







CCTCCCCCCTATGGC







CCTGACTGGAA






(SEQ ID NO: 344)





MALAT1 ENE-
TAGGGTCATGAAGGTTTTTC
RESULTS IN
ATCCTTCCAGTCAGG


MASCRNA
TTTTCCTGAGAAAACAACAC
RETENTION OF
GCCATGTTTGAGAGC



GTATTGTTTTCTCAGGTTTTG
RNA IN

TAGAAATAGCAAGTT




CTTTTTGGCCTTTTTCTAGCT
NUCLEUS,

TAAATAAGGCTAGTC




TAAAAAAAAAAAAAGCAAA
TRANSCRIPT

CGTTATCAACTTGAA




AGATGCTGGTGGTTGGCACT
TERMINATION

AAAGTGGGACCGAG




CCTGGTTTCCAGGACGGGGT
AND

TCGGTCC
ACCTGAACC




TCAAATCCCTGCGGCGTCTT
STABILIZATION

GTATATC
AGTCAAAG




TGCTTTGACT (SEQ ID NO:


CAAAGACGCCGCAG




345)


GGATTTGAACCCCG







TCCTGGAAACCAGG







AGTGCCAACCACCA







GCATCTTTTGCTTTT







TTTTTTTTTAAGCTA







GAAAAAGGCCAAAA







AGCAAAACCTGAGA







AAACAATACGTGTT







GTTTTCTCAGGAAA







AGAAAAACCTTCAT







GACCCTACTATGGCC






CTGACTGGAA





(SEQ ID NO: 346)





KSHV PAN ENE
TGTTTTGGCTGGGTTTTTCCT
RESULTS IN
ATCCTTCCAGTCAGG



TGTTCGCACCGGACACCTCC
RETENTION OF
GCCATGTTTGAGAGC



AGTGACCAGACGGCAAGGT
RNA IN

TAGAAATAGCAAGTT




TTTTATCCCAGTGTATATTGG
NUCLEUS,

TAAATAAGGCTAGTC




AAAAACATGTTATACTTTTG
TRANSCRIPT

CGTTATCAACTTGAA




ACAATTTAACGTGCCTAGAG
TERMINATION

AAAGTGGGACCGAG




CTCAAATTAAACTAATACCAT
AND

TCGGTCC
ACCTGAACC




AACGTAATGCAACTTACAAC
STABILIZATION

GTATATC
TTTTTTTTT




ATAAATAAAGGTCAATGTTT


TTTTTTTTTTATGGA




AATCCATAAAAAAAAAAAA


TTAAACATTGACCTT




AAAAAAA (SEQ ID NO: 347)


TATTTATGTTGTAAG







TTGCATTACGTTATG







GTATTAGTTTAATTT







GAGCTCTAGGCACG







TTAAATTGTCAAAA







GTATAACATGTTTTT







CCAATATACACTGG







GATAAAAACCTTGC







CGTCTGGTCACTGG







AGGTGTCCGGTGCG







AACAAGGAAAAACC







CAGCCAAAACACTAT







GGCCCTGACTGGAA






(SEQ ID NO: 348)





THREE,
TGTTTTGGCTGGGTTTTTCCT
RESULTS IN
ATCCTTCCAGTCAGG


SEQUENTIAL
TGTTCGCACCGGACACCTCC
RETENTION OF
GCCATGTTTGAGAGC


KSHV PAN
AGTGACCAGACGGCAAGGT
RNA IN

TAGAAATAGCAAGTT



ENES WITH
TTTTATCCCAGTGTATATTGG
NUCLEUS,

TAAATAAGGCTAGTC



SHORT,
AAAAACATGTTATACTTTTG
TRANSCRIPT

CGTTATCAACTTGAA



UNCONSERVED
ACAATTTAACGTGCCTAGAG
TERMINATION

AAAGTGGGACCGAG



RNA
CTCAAATTAAACTAATACCAT
AND

TCGGTCC
ACCTGAACC



LINKERS
AACGTAATGCAACTTACAAC
STABILIZATION,

GTATATC
TTTTTTTTT




ATAAATAAAGGTCAATGTTT
PREDICTED

TTTTTTTTTTATGGA




AATCCATAAAAAAAAAAAA
TOBE

TTAAACATTGACCTT




AAAAAAAACACACTGTTTTG
GREATER

TATTTATGTTGTAAG




GCTGGGTTTTTCCTTGTTCG
THANA

TTGCATTACGTTATG




CACCGGACACCTCCAGTGAC
SINGLE PAN

GTATTAGTTTAATTT




CAGACGGCAAGGTTTTTATC
ENE

GAGCTCTAGGCACG




CCAGTGTATATTGGAAAAAC


TTAAATTGTCAAAA




ATGTTATACTTTTGACAATTT


GTATAACATGTTTTT




AACGTGCCTAGAGCTCAAAT


CCAATATACACTGG




TAAACTAATACCATAACGTA


GATAAAAACCTTGC




ATGCAACTTACAACATAAAT


CGTCTGGTCACTGG




AAAGGTCAATGTTTAATCCA


AGGTGTCCGGTGCG




TAAAAAAAAAAAAAAAAAA


AACAAGGAAAAACC




ATCTCTCTGTTTTGGCTGGGT


CAGCCAAAACAGAG




TTTTCCTTGTTCGCACCGGA


AGATTTTTTTTTTTT




CACCTCCAGTGACCAGACG


TTTTTTTATGGATTA




GCAAGGTTTTTATCCCAGTG


AACATTGACCTTTAT




TATATTGGAAAAACATGTTAT


TTATGTTGTAAGTTG




ACTTTTGACAATTTAACGTG


CATTACGTTATGGTA




CCTAGAGCTCAAATTAAACT


TTAGTTTAATTTGAG




AATACCATAACGTAATGCAA


CTCTAGGCACGTTA




CTTACAACATAAATAAAGGT


AATTGTCAAAAGTAT




CAATGTTTAATCCATAAAAA


AACATGTTTTTCCA




AAAAAAAAAAAAAA (SEQ


ATATACACTGGGATA




ID NO: 349)


AAAACCTTGCCGTC







TGGTCACTGGAGGT







GTCCGGTGCGAACA







AGGAAAAACCCAGC







CAAAACAGTGTGTT







TTTTTTTTTTTTTTT







TTTATGGATTAAACA







TTGACCTTTATTTAT







GTTGTAAGTTGCAT







TACGTTATGGTATTA







GTTTAATTTGAGCTC







TAGGCACGTTAAAT







TGTCAAAAGTATAA







CATGTTTTTCCAATA







TACACTGGGATAAA







AACCTTGCCGTCTG







GTCACTGGAGGTGT







CCGGTGCGAACAAG







GAAAAACCCAGCCA







AAACACTATGGCCCT







GACTGGAA






(SEQ ID NO: 350)





SMBOX/U1
CAGCAAGTTCAGAGAAATCT
RESULTS IN
ATCCTTCCAGTCAGG


SNRNABOX
GAACTTGCTGGATTTTTGGA
RETENTION OF
GCCATGTTTGAGAGC



GCAGGGAGATGGAATAGGA
RNA IN

TAGAAATAGCAAGTT




GCTTGCTCCGTCCACTCCAC
NUCLEUS AND

TAAATAAGGCTAGTC




GCATCGACCTGGTATTGCAG
TRANSCRIPT

CGTTATCAACTTGAA




TACCTCCAGGAACGGTGCAC
TERMINATION

AAAGTGGGACCGAG




CCACTTTCTGGAGTTTCAAA


TCGGTCC
ACCTGAACC




AGTAGACTGTACGCTAAGGG


GTATATC
TTTAAGACG




TCATATCTTTTTTTGTTTGGT


CCAACCAAGACACA




TTGTGTCTTGGTTGGCGTCT


AACCAAACAAAAAA




TAAA(SEQ ID NO: 351)


AGATATGACCCTTA







GCGTACAGTCTACT







TTTGAAACTCCAGA







AAGTGGGTGCACCG







TTCCTGGAGGTACT







GCAATACCAGGTCG







ATGCGTGGAGTGGA







CGGAGCAAGCTCCT







ATTCCATCTCCCTGC







TCCAAAAATCCAGC







AAGTTCAGATTTCT







CTGAACTTGCTGCT






ATGGCCCTGACTGGAA





(SEQ ID NO: 352)





U1 SNRNA 3′
GTTTCAAAAGTAGACTGTAC
RESULTS IN
ATCCTTCCAGTCAGG


BOX
GCTAAGGGTCATATCTTTTTT
RETENTION OF
GCCATGTTTGAGAGC



TGTTTGGTTTGTGTCTTGGTT
RNA IN

TAGAAATAGCAAGTT




GGCGTCTTAAA (SEQ ID NO:
NUCLEUS AND

TAAATAAGGCTAGTC




353)
TRANSCRIPT

CGTTATCAACTTGAA





TERMINATION

AAAGTGGGACCGAG







TCGGTCC
ACCTGAACC







GTATATC
TTTAAGACG







CCAACCAAGACACA







AACCAAACAAAAAA







AGATATGACCCTTA







GCGTACAGTCTACT







TTTGAAACCTATGGC







CCTGACTGGAA






(SEQ ID NO: 354)





TRNA-LYSINE
GCCCGGCTAGCTCAGTCGGT
REPORTED TO
ATCCTTCCAGTCAGG



AGAGCATGAGACTCTTAATC
ENABLE
GCCATGTTTGAGAGC



TCAGGGTCGTGGGTTCGAGC
TRANSPORT

TAGAAATAGCAAGTT




CCCACGTTGGGCG (SEQ ID
OF RNA TO

TAAATAAGGCTAGTC




NO: 355)
MITOCHONDRIA

CGTTATCAACTTGAA







AAAGTGGGACCGAG







TCGGTCC
ACCTGAACC







GTATATC







CGCCCAACGTGGGG







CTCGAACCCACGAC







CCTGAGATTAAGAG







TCTCATGCTCTACC







GACTGAGCTAGCCG







GGCCTATGGCCCTGA







CTGGAA






(SEQ ID NO: 356)





BROCCOLI
GAGACGGTCGGGTCCAGATA
VISUALIZATION
ATCCTTCCAGTCAGG


APTAMER
TTCGTATCTGTCGAGTAGAG
(FLUORESCENCE)
GCCATGTTTGAGAGC



TGTGGGCTC (SEQ ID NO:


TAGAAATAGCAAGTT




357)


TAAATAAGGCTAGTC







CGTTATCAACTTGAA







AAAGTGGGACCGAG







TCGGTCC
ACCTGAACC







GTATATC
GAGCCCAC







ACTCTACTCGACAG







ATACGAATATCTGGA







CCCGACCGTCTCCT






ATGGCCCTGACTGGAA





(SEQ ID NO: 358)





SPINACH
GACGCAACTGAATGAAATGG
VISUALIZATION
ATCCTTCCAGTCAGG


APTAMER
TGAAGGACGGGTCCAGGTG
(FLUORESCENCE)
GCCATGTTTGAGAGC



TGGCTGCTTCGGCAGTGCAG


TAGAAATAGCAAGTT




CTTGTTGAGTAGAGTGTGAG


TAAATAAGGCTAGTC




CTCCGTAACTAGTCGCGTC


CGTTATCAACTTGAA




(SEQ ID NO: 359)


AAAGTGGGACCGAG







TCGGTCC
ACCTGAACC







GTATATC
GACGCGAC







TAGTTACGGAGCTC







ACACTCTACTCAAC







AAGCTGCACTGCCG







AAGCAGCCACACCT







GGACCCGTCCTTCA







CCATTTCATTCAGTT







GCGTCCTATGGCCCT







GACTGGAA






(SEQ ID NO: 360)





SPINACH2
GATGTAACTGAATGAAATGG
VISUALIZATION
ATCCTTCCAGTCAGG


APTAMER
TGAAGGACGGGTCCAGTAG
(FLUORESCENCE)
GCCATGTTTGAGAGC



GCTGCTTCGGCAGCCTACTT


TAGAAATAGCAAGTT




GTTGAGTAGAGTGTGAGCTC


TAAATAAGGCTAGTC




CGTAACTAGTTACATC (SEQ


CGTTATCAACTTGAA




ID NO: 361)


AAAGTGGGACCGAG







TCGGTCC
ACCTGAACC







GTATATC
GATGTAACT







AGTTACGGAGCTCA







CACTCTACTCAACA







AGTAGGCTGCCGAA







GCAGCCTACTGGAC







CCGTCCTTCACCAT







TTCATTCAGTTACAT







CCTATGGCCCTGACTG







GAA






(SEQ ID NO: 362)





MANGO
GGCACGTACGAAGGGACGG
VISUALIZATION
ATCCTTCCAGTCAGG


APTAMER
TGCGGAGAGGAGAGTACGT
(FLUORESCENCE)
GCCATGTTTGAGAGC



GC (SEQ ID NO: 363)


TAGAAATAGCAAGTT







TAAATAAGGCTAGTC







CGTTATCAACTTGAA







AAAGTGGGACCGAG







TCGGTCC
ACCTGAACC







GTATATC
GCACGTAC







TCTCCTCTCCGCAC







CGTCCCTTCGTACG







TGCCCTATGGCCCTG







ACTGGAA






(SEQ ID NO: 364)





HDV
GGCCGGCATGGTCCCAGCCT
3′ END RNA
ATCCTTCCAGTCAGG


RIBOZYME
CCTCGCTGGCGCCGGCTGGG
PROCESSING
GCCATGTTTGAGAGC



CAACATGCTTCGGCATGGCG


TAGAAATAGCAAGTT




AATGGGAC (SEQ ID NO: 365)


TAAATAAGGCTAGTC







CGTTATCAACTTGAA







AAAGTGGGACCGAG







TCGGTCC
ACCTGAACC







GTATATC
GTCCCATTC







GCCATGCCGAAGCA







TGTTGCCCAGCCGG







CGCCAGCGAGGAG







GCTGGGACCATGCC







GGCCCTATGGCCCTG







ACTGGAA






(SEQ ID NO: 366)





N6-
GGACTCTAGGACTGGACTTT
TARGET FOR
ATCCTTCCAGTCAGG


METHYLADENOSINE
GGACT (SEQ ID NO: 367)
METHYLATION
GCCATGTTTGAGAGC


MARKER (M6A)

(UNDERLINED

TAGAAATAGCAAGTT





A'S ARE

TAAATAAGGCTAGTC





METHYLATED).

CGTTATCAACTTGAA





M6A

AAAGTGGGACCGAG





METHYLATION

TCGGTCC
ACCTGAACC





CAN RESULT

GTATATC
AGTCCAAA





IN ENHANCED

GTCCAGTCCTAGAG





RNA

TCCCTATGGCCCTGA





STABILITY

CTGGAA





AND
(SEQ ID NO: 368)




EXPRESSION,





BUT IS NOT





YET FULLY





UNDERSTOOD





*each PEgRNA is shown in the 5′ to 3′ direction and has the following structural elements of FIG. 3F as designated by font type, as follows: 5′-spacer sequence (normal font)-gRNA core (underlined sequence)-homology arm (italicized)-RT template (bolded font)-primer binding site (italicized)-3′.






The PEgRNAs of the above table are designed to site-specifically insert examples of the above motifs into the HEXA gene (defective in Tay-Sachs disease) (e.g., GenBank No. KR710351.1 (SEQ ID NO: 369), however, this is only for purposes of illustration. The use of prime editing in RNA tagging is not limited to the HEXA gene and indeed may be any. The HEXA mRNA has the following nucleotide sequence:











(SEQ ID NO: 369)



GTTCGTTGCAACAAATTGATGAGCAATGCTTTTTT







ATAATGCCAACTTTGTACAAAAAAGTTGGCATGAC







AAGTTCCAGGCTTTGGTTTTCGCTGCTGCTGGCGG







CAGCGTTCGCAGGACGGGCGACGGCCCTCTGGCCC







TGGCCTCAGAACTTCCAAACCTCCGACCAGCGCTA







CGTCCTTTACCCGAACAACTTTCAATTCCAGTACG







ATGTCAGCTCGGCCGCGCAGCCCGGCTGCTCAGTC







CTCGACGAGGCCTTCCAGCGCTATCGTGACCTGCT







TTTCGGTTCCGGGTCTTGGCCCCGTCCTTACCTCA







CAGGGAAACGGCATACACTGGAGAAGAATGTGTTG







GTTGTCTCTGTAGTCACACCTGGATGTAACCAGCT







TCCTACTTTGGAGTCAGTGGAGAATTATACCCTGA







CCATAAATGATGACCAGTGTTTACTCCTCTCTGAG







ACTGTCTGGGGAGCTCTCCGAGGTCTGGAGACTTT







TAGCCAGCTTGTTTGGAAATCTGCTGAGGGCACAT







TCTTTATCAACAAGACTGAGATTGAGGACTTTCCC







CGCTTTCCTCACCGGGGCTTGCTGTTGGATACATC







TCGCCATTACCTGCCACTCTCTAGCATCCTGGACA







CTCTGGATGTCATGGCGTACAATAAATTGAACGTG







TTCCACTGGCATCTGGTAGATGATCCTTCCTTCCC







ATATGAGAGCTTCACTTTTCCAGAGCTCATGAGAA







AGGGGTCCTACAACCCTGTCACCCACATCTACACA







GCACAGGATGTGAAGGAGGTCATTGAATACGCACG







GCTCCGGGGTATCCGTGTGCTTGCAGAGTTTGACA







CTCCTGGCCACACTTTGTCCTGGGGACCAGGTATC







CCTGGATTACTGACTCCTTGCTACCCTGGGTCTGA







GCCCTCTGGCACCTTTGGACCAGTGAATCCCAGTC







TCAATAATACCTATGAGTTCATGAGCACATTCTTC







TTAGAAGTCAGCTCTGTCTTCCCAGATTTTTATCT







TCATCTTGGAGGAGATGAGGTTGATTTCACCTGCT







GGAAGTCCAACCCAGAGATCCAGGACTTTATGAGG







AAGAAAGGCTTCGGTGAGGACTTCAAGCAGCTGGA







GTCCTTCTACATCCAGACGCTGCTGGACATCGTCT







CTTCTTATGGCAAGGGCTATGTGGTGTGGCAGGAG







GTGTTTGATAATAAAGTAAAGATTCAGCCAGACAC







AATCATACAGGTGTGGCGAGAGGATATTCCAGTGA







ACTATATGAAGGAGCTGGAACTGGTCACCAAGGCC







GGCTTCCGGGCCCTTCTCTCTGCCCCCTGGTACCT







GAACCGTATATCCTATGGCCCTGACTGGAAGGATT







TCTACGTAGTGGAACCCCTGGCATTTGAAGGTACC







CCTGAGCAGAAGGCTCTGGTGATTGGTGGAGAGGC







TTGTATGTGGGGAGAATATGTGGACAACACAAACC







TGGTCCCCAGGCTCTGGCCCAGAGCAGGGGCTGTT







GCCGAAAGGCTGTGGAGCAACAAGTTGACATCTGA







CCTGACATTTGCCTATGAACGTTTGTCACACTTCC







GCTGTGAGTTGCTGAGGCGAGGTGTCCAGGCCCAA







CCCCTCAATGTAGGCTTCTGTGAGCAGGAGTTTGA







ACAGACCTGCCCAACTTTCTTGTACAAAGTTGGCA







TTATAAGAAAGCATTGCTTATCAATTTGTTGCAAC







GAAC.






The corresponding HEXA protein has the following amino acid sequence:











(SEQ ID NO: 370)



MTSSRLWFSLLLAAAFAGRATALWPWPQNFQTSDQ






RYVLYPNNFQFQYDVSSAAQPGCSVLDEAFQRYRD






LLFGSGSWPRPYLTGKRHTLEKNVLVVSVVTPGCN






QLPTLESVENYTLTINDDQCLLLSETVWGALRGLE






TFSQLVWKSAEGTFFINKTEIEDFPRFPHRGLLLD






TSRHYLPLSSILDTLDVMAYNKLNVFHWHLVDDPS






FPYESFTFPELMRKGSYNPVTHIYTAQDVKEVIEY






ARLRGIRVLAEFDTPGHTLSWGPGIPGLLTPCYPG






SEPSGTFGPVNPSLNNTYEFMSTFFLEVSSVFPDF






YLHLGGDEVDFTCWKSNPEIQDFMRKKGFGEDFKQ






LESFYIQTLLDIVSSYGKGYVVWQEVFDNKVKIQP






DTIIQVWREDIPVNYMKELELVTKAGFRALLSAPW






YLNRISYGPDWKDFYVVEPLAFEGTPEQKALVIGG






EACMWGEYVDNTNLVPRLWPRAGAVAERLWSNKLT






SDLTFAYERLSHFRCELLRRGVQAQPLNVGFCEQE






FEQT.






Notably, the resulting RNA motifs would be included within the translated region of the HEXA gene, disrupting the function of the protein coding gene. Inserted polyadenylation motifs would result in premature transcript termination. This site is merely illustrative of the potential PEgRNAs that could result in insertion of the listed RNA motifs of the above table within a genomic site that is transcribed and thus which would produce an RNA product.


PEgRNAs for use with PE for RNA tagging could be expressed from a U6 promoter (in which case a single guanosine would be added to the 5′ end of the PEgRNA for guides that include protospacers that do not begin with a G and 6-7 thymine would be added to the 3′ end) or a pol II promoter such as pCMV (in which case it might be necessary to remove the intrinsically transcribed sequence of this promoter from the 5′ end of the RNA via a self-cleaving element or Csy4 motif, and a termination motif would need to be added to the 3′ end of the RNA that does not result in export of the RNA from the nucleus, such as the 3′ box motif listed above. Note that this motif would not be inserted into the genome as a result of PE, as it would be 3′ of the annealing region). The core PEgRNA scaffold is underlined, the homology and annealing regions are italicized, and the inserted sequence is bolded. Note that the sequence inserted is the reverse complement of the above examples—as described below and therefore these PEgRNAs would need to be targeted to the coding strand.


Also, note that self-cleaving ribozymes other than HDV need in some embodiments to be tailored to the given target site; that is, while HDV cleaves the encoded transcript immediately 5′ to itself, the cut sites for all other self-cleaving ribozymes are within the ribozyme itself. Therefore, the first and last roughly 5-10 nucleotides (and in some instances potentially more than 10) would actually be a part of the encoded sequence. As an example, to cleave the sequence 5′NNNNNTCATCCTGATAAACTGCAAA3′ (SEQ ID NO: 371) after the 5 Ns, where N is any nucleotide, using a hammerhead self-cleaving ribozyme, the following sequence would be inserted, where the underlined sequences form an imperfect RNA pairing element.











(SEQ ID NO: 372)



5′NNNNNCAGTTTGTACGGATGACTGATGAGTCCCAAATAGGA







CGAAACGCGCTTCGGTGCGTCTCATCCTGATAAACTGCAAA-3′.






There is significant flexibility in terms of the length and nature of this pairing element, and this would be true for any of the non-HDV self-cleaving ribozymes listed in the original submission. To install a hammerhead ribozyme to cleave the hexA mRNA using a PEgRNA with the same protospacer as the above listed constructs, the following PEgRNA sequence could be used (labels same as above):











5′ACCTGAACCGTATATCGACGCACCGAAGCGCGTTT






CGTCCTATTTGGGACTCATCAGGATATACGGTTCA






GGTGATATACGGTTCAGGTGACGCACCGAAGCGCG






TTTCGTCCTATTTGGGACTCATCAGACCTGAACCG






TATATCATCCTTCCAGTCAGGGCCATGTTTGAGAG







CTAGAAATAGCAAGTTTAAATAAGGCTAGTCCGTT








ATCAACTTGAAAAAGTGGGACCGAGTCGGTCCACC







TGAACCGTATATCGACGCACCGAAGCGCGTTTCGT







CCTATTTGGGACTCATCAGGATATACGGTTCAGGT







CTATGGCCCTGACTGGAA-3′







(wherein the core PEgRNA scaffold is underlined, the homology and annealing regions are italicized, and the inserted sequence is bolded) (SEQ ID NO: 373).


Designing other PEgRNA for insertion of RNA motifs may follow the general principle described herein. However, it is noted that many of RNA motifs are potentially highly structured, which could make it difficult for them to be reverse-transcribed and inserted into the genome. Although for some RNA sequences, such as simple hairpins, both the RNA sequence itself and its complement are structured. However, that is unlikely to be true for the sequences noted above. Therefore, when inserting these motifs, it would most likely be best for the PEgRNA to encode the reverse complement of these sequences, resulting in the insertion of the DNA sequence actually encoding the motif into the genome. Similarly, inclusion of a self-cleaving ribozyme in the PEgRNA template region would result in processing and inefficient activity, while inclusion of its reverse complement would not. Thus, these PEgRNAs will likely have to target the coding strand, whereas PEgRNAs encoding other types of insertions (such as therapeutic correction) would be able to theoretically target either strand.


Also, note that for many of the inserted motifs, the resulting PEgRNA might not be able to be transcribed from the U6 promoter, necessitating use of other promoters, such as pCMV. Similarly, longer PEgRNAs could also be less stable. Shorter motifs, such as m6A markers, would not have this challenge.


D. Use of Dual Prime Editing for Insertion of Immunoepitopes


Dual prime editing (e.g., such as the embodiment shown in FIG. 90) may also be used as a means to insert known immunogenicity epitopes into endogenous or foreign genomic DNA, resulting in modification of the corresponding proteins for therapeutic or biotechnological applications (see FIGS. 31 and 32). Prior to the invention of classical prime editing or dual prime editing, such insertions could be achieved only inefficiently and with high rates of indel formation from DSBs. prime editing solves the problem of high indel formation from insertion edits while generally offering higher efficiency than HDR. This lower rate of indel formation presents a major advantage of prime editing over HDR as a method for targeted DNA insertions, especially in the described application of inserting immunogenicity epitopes. The length of epitopes is in a range from few bases to hundreds of bases. Prime editor is an efficient approach to achieve such targeted insertions in mammalian cells.


The key concept of the invention is the use of dual prime editors to insert a nucleotide sequence containing previously described immunogenicity epitopes into endogenous or foreign genomic DNA for the downregulation and/or destruction of their protein products and/or expressing cell types. Nucleotide sequences for immunogenic epitope insertion would be targeted to genes in a manner to produce fusion proteins of the targeted gene's coded protein and the inserted immunogenic epitope's corresponding protein translation. Patient's immune systems will have been previously trained to recognize these epitopes as a result of standard prior immunization from routine vaccination against, for example, tetanus or diphtheria or measles. As a result of the immunogenic nature of the fused epitopes, patient's immune systems would be expected to recognize and disable the prime edited protein (not just the inserted epitope) and potentially the cells from which it was expressed.


Precise genome targeting technologies using the CRISPR/Cas system have recently been explored in a wide range of applications, including the insertion of engineered DNA sequences into targeted genomic loci. Previously, homology-directed repair (HDR) has been used for this application, requiring an ssDNA donor template and repair initiation by means of a double-stranded DNA break (DSB). This strategy offers the broadest range of possible changes to be made in cells and is the only method available to insert large DNA sequences into mammalian cells. However, HDR is hampered by undesired cellular side effects stemming from its initiating DSB, such as high levels of indel formation, DNA translocations, large deletions, and P53 activation. In addition to these drawbacks, HDR is limited by low efficiency in many cell types (T cells are a notable exception to this observation). Recent efforts to overcome these drawbacks include fusing human Rad51 mutants to a Cas9 D10A nickase (RDN), resulting in a DSB-free HDR system that features improved HDR product:indel ratios and lower off target editing, but is still hampered by cell-type dependencies and only modest HDR editing efficiency.


Recently developed fusions of Cas9 to reverse transcriptases (“Prime editors”) coupled with PEgRNAs represent a novel genome editing technology that offers a number of advantages over existing genome editing methods, including the ability to install any single nucleotide substitution, and to insert or delete any short stretch of nucleotides (up to at least several dozen bases) in a site-specific manner Notably, PE edits are achieved with generally low rates of unintended indels. As such, PE enables targeted insertion-based editing applications that have been previously impossible or impractical.


This particular aspect describes a method for using prime editing as a means to insert known immunogenicity epitopes into endogenous or foreign genomic DNA, resulting in modification of the corresponding proteins for therapeutic or biotechnological applications (see FIGS. 31 and 32). Prior to the invention of prime editing, such insertions could be achieved only inefficiently and with high rates of indel formation from DSBs. prime editing solves the problem of high indel formation from insertion edits while generally offering higher efficiency than HDR. This lower rate of indel formation presents a major advantage of prime editing over HDR as a method for targeted DNA insertions, especially in the described application of inserting immunogenicity epitopes. The length of epitopes is in a range from few bases to hundreds of bases. Prime editor is the most efficient and cleanest technology to achieve such targeted insertions in mammalian cells.


The key concept of this aspect is the use of prime editors to insert a nucleotide sequence containing previously described immunogenicity epitopes into endogenous or foreign genomic DNA for the downregulation and/or destruction of their protein products and/or expressing cell types. Nucleotide sequences for immunogenic epitope insertion would be targeted to genes in a manner to produce fusion proteins of the targeted gene's coded protein and the inserted immunogenic epitope's corresponding protein translation. Patient's immune systems will have been previously trained to recognize these epitopes as a result of standard prior immunization from routine vaccination against, for example, tetanus or diphtheria or measles. As a result of the immunogenic nature of the fused epitopes, patient's immune systems would be expected to recognize and disable the prime edited protein (not just the inserted epitope) and potentially the cells from which it was expressed.


Fusions to targeted genes would be engineered as needed to ensure the inserted epitope protein translation is exposed for immune system recognition. This could include targeted nucleotide insertions resulting in protein translations yielding C-terminal fusions of immunogenicity epitopes to targeted genes, N-terminal fusions of immunogenicity epitopes to targeted genes, or the insertion of nucleotides into genes so that immunogenicity epitopes are coded within surfaced-exposed regions of protein structure.


Protein linkers encoded as nucleotides inserted between the target gene sequence and the inserted immunogenicity epitope nucleotide sequence may need to be engineered as part of this invention to facilitate immune system recognition, cellular trafficking, protein function, or protein folding of the targeted gene. These inserted nucleotide-encoded protein linkers may include (but are not limited to) variable lengths and sequences of the XTEN linker or variable lengths and sequences of Glycine-Serine linkers. These engineered linkers have been previous used to successfully facilitate protein fusions. Examplary linkers may include any of those described herein, including the amino acid sequence (GGGGS)n (SEQ ID NO: 165), (G)n (SEQ ID NO: 166), (EAAAK)n (SEQ ID NO: 167), (GGS)n (SEQ ID NO: 168), (SGGS)n (SEQ ID NO: 169), (XP)n (SEQ ID NO: 170), or any combination thereof, wherein n is independently an integer between 1 and 30, and wherein X is any amino acid. In some embodiments, the linker comprises the amino acid sequence (GGS)n (SEQ ID NO: 176), wherein n is 1, 3, or 7. In some embodiments, the linker comprises the amino acid sequence SGSETPGTSESATPES (SEQ ID NO: 171). In some embodiments, the linker comprises the amino acid sequence SGGSSGGSSGSETPGTSESATPESSGGSSGGS (SEQ ID NO: 172). In some embodiments, the linker comprises the amino acid sequence SGGSGGSGGS (SEQ ID NO: 173). In some embodiments, the linker comprises the amino acid sequence SGGS (SEQ ID NO: 174).


Distinguishing features of this aspect include the ability to use previously acquired immune responses to specific amino acid sequences as a means to induce an immune response against otherwise non-immunogenic proteins. Another distinguishing feature is the ability to insert the nucleotide sequences of these immunogenic epitopes in a targets manner that does not induce high levels of unwanted indels as a by-product editing and is efficient in its insertion. This specific application of PE has the ability to combine cell type-specific delivery methods (such as AAV serotypes) to insert epitopes in cell types that are of interest to trigger an immune response to.


Prime editing as a means of inserting immunogenic epitopes into pathogenic genes could be used to program the patient's immune system to fight a wide variety of diseases (not limited to cancer as with immuno-oncology strategies). An immediately relevant use of this technology would be as a cancer therapeutic as it could undermine a tumor's immune escape mechanism by causing an immune response to a relevant oncogene like HER2 or growth factors like EGFR. Such an approach could seem similar to T-cell engineering, but one novel advance of this approach is that it can be utilized in many cell types and for diseases beyond cancer, without needed to generate and introduce engineered T-cells into patients.


Using PE to insert an immunogenicity epitope which most people are already vaccinated against (tetanus, pertussis, diphtheria, measles, mumps, rubella, etc.) into a foreign or endogenous gene that drives a disease, so the patient's immune system learns to disable that protein.


Diseases that stand to have a potential therapeutic benefit from the aforementioned strategy include those caused by aggregation of toxic proteins, such as in fatal familial insomnia. Other diseases that could benefit include those caused by pathogenic overexpression of an otherwise nontoxic endogenous protein, and those caused by foreign pathogens.


Primary therapeutic indications include those mentioned above such as therapeutics for cancer, prion and other neurodegenerative diseases, infectious diseases, and preventative medicine. Secondary therapeutic indications may include preventative care for patients with late-onset genetic diseases. It is expected that current standard of care medicines may be used in conjunction with prime editing for some diseases, like particularly aggressive cancers, or in cases where medications help alleviate disease symptoms until the disease completely cured. Below are examples of immunoepitopes that may be inserted into genes by the herein disclosed prime editors:

















EPITOPE AMINO



VACCINE
DISEASE
ACID SEQUENCE
EXAMPLE NUCLEIC ACID SEQUENCE (8)


















1
TETANUS
QYIKANSKFIGITEL
CATGATATAAAAGCAAATTCTAAATTTATA



TOXOID
(SEQ ID NO: 396)
GGTATAACTGAACTA (SEQ ID NO: 397)





2
DIPHTHERIA
GADDVVDSSKSFV
GGCGCCGACGACGTGGTGGACAGCAGCA



TOXIN
MENFSSYHGTKPG
AGAGCTTCGTGATGGAGAACTTCAGCAGC



MUTANT
YVDSIQKGIQKPKS
TACCACGGCACCAAGCCCGGCTACGTGGA



CRM197
GTQGNYDDDWKEF
CAGCATCCAGAAGGGCATCCAGAAGCCCA




YSTDNKYDAAGYS
AGAGCGGCACCCAGGGCAACTACGACGA




VDNENPLSGKAGG
CGACTGGAAGGAGTTCTACAGCACCGAC




VVKVTYPGLTKVL
AACAAGTACGACGCCGCCGGCTACAGCGT




ALKVDNAETIKKEL
GGACAACGAGAACCCCCTGAGCGGCAAG




GLSLTEPLMEQVGT
GCCGGCGGCGTGGTGAAGGTGACCTACCC




EEFIKRFGDGASRV
CGGCCTGACCAAGGTGCTGGCCCTGAAG




VLSLPFAEGSSSVEY
GTGGACAACGCCGAGACCATCAAGAAGG




INNWEQAKALSVE
AGCTGGGCCTGAGCCTGACCGAGCCCCTG




LEINFETRGKRGQD
ATGGAGCAGGTGGGCACCGAGGAGTTCAT




AMYEYMAQACAG
CAAGAGGTTCGGCGACGGCGCCAGCAGG




NRVRRSVGSSLSCI
GTGGTGCTGAGCCTGCCCTTCGCCGAGGG




NLDWDVIRDKTKT
CAGCAGCAGCGTGGAGTACATCAACAACT




KIESLKEHGPIKNK
GGGAGCAGGCCAAGGCCCTGAGCGTGGA




MSESPNKTVSEEKA
GCTGGAGATCAACTTCGAGACCAGGGGC




KQYLEEFHQTALEH
AAGAGGGGCCAGGACGCCATGTACGAGT




PELSELKTVTGTNP
ACATGGCCCAGGCCTGCGCCGGCAACAG




VFAGANYAAWAVN
GGTGAGGAGGAGCGTGGGCAGCAGCCTG




VAQVIDSETADNLE
AGCTGCATCAACCTGGACTGGGACGTGAT




KTTAALSILPGIGSV
CAGGGACAAGACCAAGACCAAGATCGAG




MGIADGAVHHNTE
AGCCTGAAGGAGCACGGCCCCATCAAGA




EIVAQSIALSSLMVA
ACAAGATGAGCGAGAGCCCCAACAAGAC




QAIPLVGELVDIGFA
CGTGAGCGAGGAGAAGGCCAAGCAGTAC




AYNFVESIINLFQVV
CTGGAGGAGTTCCACCAGACCGCCCTGGA




HNSYNRPAYSPGHK
GCACCCCGAGCTGAGCGAGCTGAAGACC




TQPFLHDGYAVSW
GTGACCGGCACCAACCCCGTGTTCGCCGG




NTVEDSIIRTGFQGE
CGCCAACTACGCCGCCTGGGCCGTGAACG




SGHDIKITAENTPLP
TGGCCCAGGTGATCGACAGCGAGACCGCC




IAGVLLPTIPGKLDV
GACAACCTGGAGAAGACCACCGCCGCCC




NKSKTHISVNGRKI
TGAGCATCCTGCCCGGCATCGGCAGCGTG




RMRCRAIDGDVTF
ATGGGCATCGCCGACGGCGCCGTGCACCA




CRPKSPVYVGNGV
CAACACCGAGGAGATCGTGGCCCAGAGC




HANLHVAFHRSSSE
ATCGCCCTGAGCAGCCTGATGGTGGCCCA




KIHSNEISSDSIGVL
GGCCATCCCCCTGGTGGGCGAGCTGGTGG




GYQKTVDHTKVNS
ACATCGGCTTCGCCGCCTACAACTTCGTG




KLSLFFEIKS (SEQ
GAGAGCATCATCAACCTGTTCCAGGTGGT




ID NO: 398)
GCACAACAGCTACAACAGGCCCGCCTACA





GCCCCGGCCACAAGACCCAGCCCTTCCTG





CACGACGGCTACGCCGTGAGCTGGAACA





CCGTGGAGGACAGCATCATCAGGACCGGC





TTCCAGGGCGAGAGCGGCCACGACATCA





AGATCACCGCCGAGAACACCCCCCTGCCC





ATCGCCGGCGTGCTGCTGCCCACCATCCC





CGGCAAGCTGGACGTGAACAAGAGCAAG





ACCCACATCAGCGTGAACGGCAGGAAGAT





CAGGATGAGGTGCAGGGCCATCGACGGC





GACGTGACCTTCTGCAGGCCCAAGAGCCC





CGTGTACGTGGGCAACGGCGTGCACGCCA





ACCTGCACGTGGCCTTCCACAGGAGCAGC





AGCGAGAAGATCCACAGCAACGAGATCA





GCAGCGACAGCATCGGCGTGCTGGGCTAC





CAGAAGACCGTGGACCACACCAAGGTGA





ACAGCAAGCTGAGCCTGTTCTTCGAGATC





AAGAGC (SEQ ID NO: 399)





3
MUMPS
GTYRLIPNARANLT
GGCACCTACAGGCTGATCCCCAACGCCAG



IMMUNO-
A (SEQ ID NO: 400)
GGCCAACCTGACCGCC (SEQ ID NO: 401)



EPITOPE 1







4
MUMPS
PSKFFTISDSATFAP
CCGAGCAAATTTTTTACCATTAGCGATAGC



IMMUNO-
GPVSNA (SEQ ID
GCGACCTTTGCGCCGGGCCCGGTGAGCAA



EPITOPE 2
NO: 402)
CGCG (SEQ ID NO: 403)



MUMPS
PSKLFIMLDNATFAP
CCGAGCAAACTGTTTATTATGCTGGATAAC



IMMUNO-
GPVVNA (SEQ ID
GCGACCTTTGCGCCGGGCCCGGTGGTGAA



EPITOPE 1
NO: 404)
CGCG (SEQ ID NO: 405)





SELECTED EXAMPLES FROM





HEMAGGLUTININ-NEURAMINIDASE (HN)





DIVERSITY AMONG OUTBREAK STRAINS





(TABLE1) DIVERGENCE BETWEEN





VACCINE STRAIN JL5 AND OUTBREAK





STRAINS (TABLE2)





5
RUBELLA
TPPPYQVSCGGESD
ACCCCCCCCCCCTACCAGGTGAGCTGCGG



VIRUS (RV)
RASARVIDPAAQS
CGGCGAGAGCGACAGGGCCAGCGCCAGG




(SEQ ID NO: 406)
GTGATCGACCCCGCCGCCCAGAGC (SEQ





ID NO: 407)





6
HEMAGGLUTININ
PEYAYKIVKNKKME
CCCGAGTACGCCTACAAGATCGTGAAGAA




DGFLQGMVDGWY
CAAGAAGATGGAGGACGGCTTCCTGCAG




GHHSNEQGSGLME
GGCATGGTGGACGGCTGGTACGGCCACCA




NERTLDKANPNND
CAGCAACGAGCAGGGCAGCGGCCTGATG




LCSWSDHEASSNNT
GAGAACGAGAGGACCCTGGACAAGGCCA




NQEDLLQRESRRK
ACCCCAACAACGACCTGTGCAGCTGGAG




KRIGTSTLNQRGNC
CGACCACGAGGCCAGCAGCAACAACACC




NTKCQTEEARLKRE
AACCAGGAGGACCTGCTGCAGAGGGAGA




EVSLVKSDQCSNGS
GCAGGAGGAAGAAGAGGATCGGCACCAG




LQCRANNSTEQVD
CACCCTGAACCAGAGGGGCAACTGCAAC




(SEQ ID NO: 408)
ACCAAGTGCCAGACCGAGGAGGCCAGGC





TGAAGAGGGAGGAGGTGAGCCTGGTGAA





GAGCGACCAGTGCAGCAACGGCAGCCTG





CAGTGCAGGGCCAACAACAGCACCGAGC





AGGTGGAC (SEQ ID NO: 409)





7
NEURAMINIDASE
TKSTNSRSGGISGP
ACCAAGAGCACCAACAGCAGGAGCGGCG




DNEAPVGEAPSPYG
GCATCAGCGGCCCCGACAACGAGGCCCCC




DNPRPNDGNNIRIG
GTGGGCGAGGCCCCCAGCCCCTACGGCG




SKGYNGIITDTIEES
ACAACCCCAGGCCCAACGACGGCAACAA




CSCYPDAKVVKSV
CATCAGGATCGGCAGCAAGGGCTACAACG




ELDSTIWTSGSSPN
GCATCATCACCGACACCATCGAGGAGAGC




QKIITIGWDPNGWT
TGCAGCTGCTACCCCGACGCCAAGGTGGT




GTPMSPNGAYGTD
GAAGAGCGTGGAGCTGGACAGCACCATC




GPSNGQANQHQAE
TGGACCAGCGGCAGCAGCCCCAACCAGA




SISAGNSSLCPIRDN
AGATCATCACCATCGGCTGGGACCCCAAC




WHGSNRSWSWPD
GGCTGGACCGGCACCCCCATGAGCCCCAA




GAE (SEQ ID NO:
CGGCGCCTACGGCACCGACGGCCCCAGC




410)
AACGGCCAGGCCAACCAGCACCAGGCCG





AGAGCATCAGCGCCGGCAACAGCAGCCT





GTGCCCCATCAGGGACAACTGGCACGGCA





GCAACAGGAGCTGGAGCTGGCCCGACGG





CGCCGAG (SEQ ID NO: 411)





8
TAP1
EKIVLLLAMMEKIV
GAGAAGATCGTGCTGCTGCTGGCCATGAT



(TRANSPORT
LLLAKCQTPMGAIK
GGAGAAGATCGTGCTGCTGCTGGCCAAGT



ANTIGEN
AVDGVTNKCPYLG
GCCAGACCCCCATGGGCGCCATCAAGGCC



PRESENTATION)
SPSF (SEQ ID NO:
GTGGACGGCGTGACCAACAAGTGCCCCTA



ON
412)
CCTGGGCAGCCCCAGCTTC (SEQ ID NO:



H5N1

413)



VIRUS





HEMAGGLUTININ







9
TAP2
IRPCFWVELNPNQK
ATCAGGCCCTGCTTCTGGGTGGAGCTGAA



(TRANSPOR
IITIRPCFWVELICYP
CCCCAACCAGAAGATCATCACCATCAGGC



T ANTIGEN
DAGEIT (SEQ ID
CCTGCTTCTGGGTGGAGCTGATCTGCTAC



PRESENTATION)
NO: 414)
CCCGACGCCGGCGAGATCACC (SEQ ID



ON

NO: 415)



H5N1





VIRUS





NEURAMINIDASE




10
HEMAGGLUTININ
MEKIVLLLAEKIVL
ATGGAGAAGATCGTGCTGCTGCTGGCCGA



EPITOPES
LLAMCPYLGSPSFK
GAAGATCGTGCTGCTGCTGGCCATGTGCC



TOWARD
CQTPMGAIKAVDG
CCTACCTGGGCAGCCCCAGCTTCAAGTGC



CLASS I
VTNK (SEQ ID NO:
CAGACCCCCATGGGCGCCATCAAGGCCGT



HLA
416)
GGACGGCGTGACCAACAAG (SEQ ID NO:





417)





11
NEURAMINIDASE
NPNQKIITICYPDAGE
AACCCCAACCAGAAGATCATCACCATCTGC



EPITOPES
ITIRPCFWVELRPCFW
TACCCCGACGCCGGCGAGATCACCATCAGG



TOWARD
VELI (SEQ ID NO:
CCCTGCTTCTGGGTGGAGCTGAGGCCCTGC



CLASS I
418)
TTCTGGGTGGAGCTGATC (SEQ ID NO: 419)



HLA







12
HEMAGGLUTININ
MVSLVKSDQIGTSTL
ATGGTGAGCCTGGTGAAGAGCGACCAGATC



EPITOPES
NQR (SEQ ID NO: 420)
GGCACCAGCACCCTGAACCAGAGG (SEQ ID



TOWARD

NO: 421)



CLASS II





HLA







13
NEURAMINIDASE
YNGIITDTI (SEQ ID
TACAACGGCATCATCACCGACACCATC (SEQ



EPITOPES
NO: 422)
ID NO: 423)



TOWARD





CLASS II





HLA







14
HEMAGGLUTININ
MEKIVLLLAEKIVLL
ATGGAGAAGATCGTGCTGCTGCTGGCCGAG



EPITOPE
LAMMVSLVKSDQCP
AAGATCGTGCTGCTGCTGGCCATGATGGTG



H5N1-
YLGSPSFIGTSTLNQR
AGCCTGGTGAAGAGCGACCAGTGCCCCTAC



BOUND
KCQTPMGAIKAVDG
CTGGGCAGCCCCAGCTTCATCGGCACCAGC



CLASS I
VTNK (SEQ ID NO:
ACCCTGAACCAGAGG (SEQ ID NO: 425)



AND CLASS
424)




II HLA







15
NEURAMINIDASE
NPNQKIITIYNGIITDT
AACCCCAACCAGAAGATCATCACCATCTAC



EPITOPE
ICYPDAGEITIRPCFW
AACGGCATCATCACCGACACCATCTGCTAC



H5N1-
VELRPCFWVELI
CCCGACGCCGGCGAGATCACCATCAGGCCC



BOUND
(SEQ ID NO: 426)
TGCTTCTGGGTGGAGCTGAGGCCCTGCTTC



CLASS I

TGGGTGGAGCTGATC (SEQ ID NO: 427)



AND





CLASS II 





HLA









Additional immunoepitopes may also be installed which are known in the art. Any of the immunoepitopes available from the Immune Epitope Database and Analysis Resource (iedb.org/epitopedetails_v3.php) (the contents of which are incorporated herein by reference) may be installed by the prime editors disclosed herein.


In some embodiments, the immunoepitopes which may be installed by the prime editors disclosed herein may include any of the following epitopes:



















SEQ






ID
ACCESS.


NO.
DISEASE
EPITOPE AMINO ACID SEQUENCE
NO
NO.



















1
TETANUS TOXOID
QYIKANSKFIGITEL
396
NA





2
DIPHTHERIA
GADDVVDSSKSFVMENFSSYHGTKPGYVDS
428
NA



TOXIN MUTANT
IQKGIQKPKSGTQGNYDDDWKEFYSTDNKY





CRM 197
DAAGYSVDNENPLSGKAGGVVKVTYPGLTK






VLALKVDNAETIKKELGLSLTEPLMEQVGT






EEFIKRFGDGASRVVLSLPFAEGSSSVEYI






NNWEQAKALSVELEINFETRGKRGQDAMYE






YMAQACAGNRVRRSVGSSLSCINLDWDVIR






DKTKTKIESLKEHGPIKNKMSESPNKTVSE






EKAKQYLEEFHQTALEHPELSELKTVTGTN






PVFAGANYAAWAVNVAQVIDSETADNLEKT






TAALSILPGIGSVMGIADGAVHHNTEEIVA






QSIALSSLMVAQAIPLVGELVDIGFAAYNF






VESIINLFQVVHNSYNRPAYSPGHKTQPFL






HDGYAVSWNTVEDSIIRTGFQGESGHDIKI






TAENTPLPIAGVLLPTIPGKLDVNKSKTHI






SV






NGRKIRMRCRAIDGDVTFCRPKSPVYVGNG






VHANLHVAFHRSSSEKIHSNEISSDSIGVL






GYQKTVDHTKVNSKLSLFFEIKS







3
MUMPS
GTYRLIPNARANLTA
400
NA





4
MUMPS
PSKFFTISDSATFAPGPVSNA;
402;
NA




PSKLFIMLDNATFAPGPVVNA
404






5
RUBELLA VIRUS
TPPPYQVSCGGESDRASARVIDPAAQS
406
NA



(RV)








6
HEMAGGLUTININ
PEYAYKIVKNKKMEDGFLQGMVDGWYGHHS
408
NA




NEQGSGLMENERTLDKANPNNDLCSWSDHE






ASSNNTNQEDLLQRESRRKKRIGTSTLNQR






GNCNTKCQTEEARLKREEVSLVKSDQCSNG






SLQCRANNSTEQVD







7
NEURAMINIDASE
TKSTNSRSGGISGPDNEAPVGEAPSPYGDN
410
NA




PRPNDGNNIRIGSKGYNGIITDTIEESCSC






YPDAKVVKSVELDSTIWTSGSSPNQKIITI






GWDPNGWTGTPMSPNGAYGTDGPSNGQANQ






HQAESISAGNSSLCPIRDNWHGSNRSWSWP






DGAE







8
TAP (TRANSPORT
EKIVLLLAMMEKIVLLLAKCQTPMGAIKAV
412
NA



ANTIGEN
DGVTNKCPYLGSPSF





PRESENTATION)






ON H5N1 VIRUS






HEMAGGLUTININ








9
TAP (TRANSPORT
IRPCFWVELNPNQKIITIRPCFWVELICYP
414
NA



ANTIGEN
DAGEIT





PRESENTATION)






ON H5N1 VIRUS






NEURAMINIDASE








10
HEMAGGLUTININ
MEKIVLLLAEKIVLLLAMCPYLGSPSFKCQ
416
NA



EPITOPES TOWARD
TPMGAIKAVDGVTNK





CLASS I HLA








11
NEURAMINIDASE
NPNQKIITICYPDAGEITIRPCFWVELRPC
418
NA



EPITOPES TOWARD
FWVELI





CLASS I HLA








12
HEMAGGLUTININ
MVSLVKSDQIGTSTLNQR
420
NA



EPITOPES TOWARD






CLASS II HLA








13
NEURAMINIDASE
YNGIITDTI
422
NA



EPITOPES TOWARD






CLASS II HLA








14
HEMAGGLUTININ
MEKIVLLLAEKIVLLLAMMVSLVKSDQCPY
424
NA



EPITOPE H5N1-
LGSPSFIGTSTLNQRKCQTPMGAIKAVDGV





BOUND CLASS I
TNK





AND CLASS II






HLA








15
NEURAMINIDASE
NPNQKIITIYNGIITDTICYPDAGEITIRP
426
NA



EPITOPE H5N1-
CFWVELRPCFWVELI





BOUND CLASS I






AND CLASS II






HLA








16

CORYNEBACTERIUM

AACAGNRVRRSVGSSLKC
899
SRC280292




DIPHTHERIAE









17
MEASLES VIRUS
AADHCPVVEVNGVTI
900
P69353.1



STRAIN






EDMONSTON








18
MEASLES VIRUS
AAHLPTGTPLDID
901
P04851.1



STRAIN






EDMONSTON








19

BORDETELLA

AALAVWAGLAVQ
902
Q00879.1




PERTUSSIS









20
MEASLES VIRUS
AALGVATAAQITAGI
903
P69353.1



STRAIN






EDMONSTON








21
RUBELLA VIRUS
AALLNTPPPYQVSCGGESDRATAR
904
P07566.1



STRAIN THERIEN








22
RUBELLA VIRUS
AAQSFTGVVYGTHTT
905
BAA28178.1





23
RUBELLA VIRUS
ACEVEPAFGHSDAAC
906
BAA28178.1





24
RUBELLA VIRUS
ACTFWAVNAYSSGGY
907
BAA28178.1





25
RUBELLA VIRUS
ADDPLLR
908
BAA19893.1





26
RUBELLA VIRUS
ADDPLLRT
909
CAJ88851.1





27
MEASLES VIRUS
AEMICDIDTYIVEAG
910
P04851.1



STRAIN






EDMONSTON








28
MEASLES VIRUS
AEMICDIDTYIVEAGLASFI
911
P04851.1



STRAIN






EDMONSTON








29
MEASLES VIRUS
AEPLLSC
912
P04851.1



STRAIN






EDMONSTON








30

BORDETELLA

AFVSTSSSRRYTEVY
913
CAD44970.1




PERTUSSIS









31

BORDETELLA

AGFIYRETFCITTIYKTGQPAADHYYSKVT
914
P04979.1




PERTUSSIS

A







32
RUBELLA VIRUS
AGLLACCAKCLYYLR
915
BAA28178.1





33
RUBELLA VIRUS
AHTTSDPWHPPG
916
BAA19893.1





34
MEASLES VIRUS
AIAKLEDAKELLESS
917
P69353.1



STRAIN






EDMONSTON








35
MEASLES VIRUS
AIDNLRASLETTNQA
918
P69353.1



STRAIN






EDMONSTON








36

BORDETELLA

AKGVEFR
919
ACI16088.1




PERTUSSIS









37
MEASLES VIRUS
AKWAVPTTRTDDKLR
920
P08362.1



STRAIN






EDMONSTON








38
MEASLES VIRUS
ALAEVLKKPV
921
ABO69699.1



STRAIN






EDMONSTON








39
MEASLES VIRUS
ALGVINTLEWIPRFK
922
P08362.1



STRAIN






EDMONSTON








40
MEASLES VIRUS
ALHQSMLNSQAIDNL
923
P69353.1



STRAIN






EDMONSTON








41
MEASLES VIRUS
ALIGILSLFV
924
ABI5411O.1



STRAIN






EDMONSTON








42
RUBELLA VIRUS
ALLNTPPPYQVSCGGESDRA
925
CAJ88851.1





43
RUBELLA VIRUS
ALLNTPPPYQVSCGGESDRASARV
926
CAJ88851.1



STRAIN M33








44
RUBELLA VIRUS
ALVEGLAPGGGNCHL
927
BAA28178.1





45

BORDETELLA

AMAAWSERAGEA
928
P04977.1




PERTUSSIS









46
MEASLES VIRUS
ANCASILCKCYTTGT
929
P69353.1



STRAIN






EDMONSTON








47

BORDETELLA

ANPNPYTSRRSV
930
P04977.1




PERTUSSIS









48
RUBELLA VIRUS
APGPGEVW
931
CAJ88851.1





49
RUBELLA VIRUS
APLPPHTTERIETRSARHPWRIR
932
ABD64214.1





50
RUBELLA VIRUS
APPMPPQPPRAHGQHYGHHHHQLPFLG
933
CAA33016.1



VACCINE STRAIN






RA27/3








51
RUBELLA VIRUS
APPTLPQPPCAHGQHYGHHHHQLPFLG
934
P07566.1



STRAIN THERIEN








52
RUBELLA VIRUS
APPTLPQPPRAHGQHYGHHHHQLPFLG
935
ABD64214.1





53

BORDETELLA

APQPGPQPPQPPQPQPEAPAPQ
936
P14283.3




PERTUSSIS









54
RUBELLA VIRUS
AQLASYFNPGGSYYK
937
BAA28178.1





55
MEASLES VIRUS
ARAAHLPTGTPLD
938
P04851.1



STRAIN






EDMONSTON








56
MEASLES VIRUS
ARLVSEIAMHTTEDK
939
P04851.1



STRAIN






EDMONSTON








57
MEASLES VIRUS
ARLVSEIAMHTTEDKISRAV
940
P04851.1



STRAIN






EDMONSTON








58
MEASLES VIRUS
ASDVETAEGGEIHELLR
941
PO3422.1



STRAIN






EDMONSTON-B








59
MEASLES
ASDVETAEGGEIHELLRLQ
942
ABO69699.1



MORBILLIVIRUS








60
MEASLES VIRUS
ASDVETAEGGEIHELLRLQSR
943
P03422.1



STRAIN






EDMONSTON-B








61
MEASLES
ASDVETAEGGEIHKLLRLQ
944
AAA63285.1



MORBILLIVIRUS








62
RUBELLA VIRUS
ASDVLPGHWLQG
945
NP_740663.1



STRAIN M33








63
MEASLES VIRUS
ASELGITAEDARLVS
946
P04851.1



STRAIN






EDMONSTON








64
MEASLES VIRUS
ASELGITAEDARLVSEIAMH
947
P04851.1



STRAIN






EDMONSTON








65
MEASLES VIRUS
ASESSQDPQDSRR
948
P04851.1



STRAIN






EDMONSTON








66
MEASLES VIRUS
ASILCKCYTTGTIIN
949
P69353.1



STRAIN






EDMONSTON








67
RUBELLA VIRUS
ASPVCQRHSPDCSRL
950
BAA28178.1





68

BORDETELLA

ASQARWTGATRA
951
BAF35031.1




PERTUSSIS









69
RUBELLA VIRUS
ASYFNPGGSYYKQYHPTACEVEPAFGHS
952
P07566.1



STRAIN THERIEN








70
MEASLES VIRUS
ASYKVMTRSSHQSLV
953
P69353.1



STRAIN






EDMONSTON








71

BORDETELLA

ASYVKKPKEDVD
954
ACI16088.1




PERTUSSIS









72
MEASLES VIRUS
ATAAQITAGIALHQS
955
P69353.1



STRAIN






EDMONSTON








73
RUBELLA VIRUS
ATPERPRL
956
CAJ88851.1





74
MEASLES VIRUS
AVCLGGLIGIPALIC
957
P69353.1



STRAIN






EDMONSTON








75
MEASLES VIRUS
AVGPRQAQVSF
958
P04851.1



STRAIN






EDMONSTON








76
RUBELLA VIRUS
AVNAYSSGGYAQLAS
959
BAA28178.1





77
RUBELLA VIRUS
AVSETRQTWAEWAAA
960
BAA28178.1





78
MEASLES VIRUS
AVTAPDTAADSELRR
961
P04851.1



STRAIN






EDMONSTON








79
MEASLES VIRUS
AVTAPDTAADSELRRWIKYT
962
P04851.1



STRAIN






EDMONSTON








80

BORDETELLA

AYGGIIKDAPPGAGFIYRETFC
963
P04979.1




PERTUSSIS









81
RUBELLA VIRUS
CALPLAGLLACCAKC
964
BAA28178.1





82
RUBELLA VIRUS
CARIWNGTQRACTFW
965
BAA28178.1





83
MEASLES VIRUS
CARTLVSGSFGNRFI
966
P69353.1



STRAIN






EDMONSTON








84

BORDETELLA

CASPYEGRYRDMYDALRBRLLY
967
SRC280066




PERTUSSIS









85

BORDETELLA

CAVFVRSGQPVIGA
968
AAA83981.1




PERTUSSIS









86
RUBELLA VIRUS
CCAKCLYYLRGAIAPR
969
BAA28178.1





87
MEASLES VIRUS
CCRGRCNKKGEQVGM
970
P69353.1



STRAIN






EDMONSTON








88
RUBELLA VIRUS
CEIPTDVSCEGLGAW
971
BAA28178.1





89

BORDETELLA

CFGKDLKRPGSSPMEV
972
P0A3R5.1




PERTUSSIS









90
MEASLES
CFQQACKGKIQALCE
973
P06830.1



MORBILLIVIRUS








91
MEASLES
CFQQACKGKIQALCENPEWAPLKDNRIPS
974
AAR89413.1



MORBILLIVIRUS








92
RUBELLA VIRUS
CGGESDRASARVIDP
975
BAA28178.1





93

BORDETELLA

CITTIYKTGQPAADHYYSKVTA
976
P04979.1




PERTUSSIS









94
MEASLES
CKGKIQALCENPEWA
977
AAR89413.1



MORBILLIVIRUS








95
MEASLES VIRUS
CKPWQESRKNKAQ
978
P04851.1



STRAIN






EDMONSTON








96
MEASLES VIRUS
CNKKGEQVGMSRPGL
979
P69353.1



STRAIN






EDMONSTON








97
RUBELLA VIRUS
CNVTTEHPFCNTPHG
980
BAA28178.1





98

BORDETELLA

CQVGSSNSAF
981
P04977.1




PERTUSSIS









99
MEASLES
CSGPTTIRGQFS
982
P08362.1



MORBILLIVIRUS








100

BORDETELLA

CTSPYDGKYWSMYSRL
983
AAA83981.1




PERTUSSIS









101
MEASLES VIRUS
CVLADSESGGHITHS
984
P08362.1



STRAIN






EDMONSTON








102
MEASLES
CYTTGTIINQDPDKILTYIAADHC
985
AAF02706.1



MORBILLIVIRUS








103
RUBELLA VIRUS
DADDPLLR
986
CAJ88851.1





104
MEASLES VIRUS
DARAAHLPTGTPLDI
987
P04851.1



STRAIN






EDMONSTON








105
MEASLES VIRUS
DARAAHLPTGTPLDIDTASE
988
P04851.1



STRAIN






EDMONSTON








106
MEASLES VIRUS
DCHAPTYLPAEVDGD
989
P08362.1



STRAIN






EDMONSTON








107
RUBELLA VIRUS
DCSRLVGATPERPRL
990
BAA28178.1





108
MEASLES VIRUS
DDKLRMETCFQQACK
991
P08362.1



STRAIN






EDMONSTON








109
RUBELLA VIRUS
DDPLLRTA
992
CAJ88851.1





110
RUBELLA VIRUS
DDPLLRTAPGPGEVW
993
BAA28178.1





111

BORDETELLA

DDPPATVYRYD
994
P04977.1




PERTUSSIS









112

BORDETELLA

DDPPATVYRYDSRPPED
995
CAD44970.1




PERTUSSIS









113

BORDETELLA

DDPPATVYRYDSRPPEDV
996
ACI04548.1




PERTUSSIS









114
MEASLES
DEVGLRTPQRFTDLV
997
P06830.1



MORBILLIVIRUS








115
MEASLES VIRUS
DHCPVVEVNGVTIQV
998
P69353.1



STRAIN






EDMONSTON








116
MEASLES VIRUS
DIDTASESSQDPQ
999
P04851.1



STRAIN






EDMONSTON








117
MEASLES VIRUS
DINKVLEKLGYSGGD
1000
P69353.1



STRAIN






EDMONSTON








118

BORDETELLA

DLIAYKQ
1001
ACI16088.1




PERTUSSIS









119
MEASLES VIRUS
DLIGQKLGLKLLRYY
1002
P69353.1



STRAIN






EDMONSTON








120
RUBELLA VIRUS
DLQKALEAQSRALRAELAA
1003
P07566.1





121
MEASLES
DLQYVLATYDTSRVE
1004
P06830.1



MORBILLIVIRUS








122
MEASLES VIRUS
DLSLRRFMV
1005
P04851.1



STRAIN






EDMONSTON








123
MEASLES VIRUS
DLSNCMVALGELKLA
1006
P08362.1



STRAIN






EDMONSTON








124
RUBELLA VIRUS
DLVEYIMNYTGNQQSRWGLGSPNC
1007
CAJ88851.1





125
MEASLES
DLVKFISDKIKFLNP
1008
AAR89413.1



MORBILLIVIRUS








126
MEASLES
DLVKFISTKIKFLNP
1009
SRC280117



MORBILLIVIRUS








127
MEASLES VIRUS
DLYKSNHNNV
1010
P08362.1



STRAIN






EDMONSTON








128

BORDETELLA

DNVLDHLTGR
1011
ACI04548.1




PERTUSSIS









129

BORDETELLA

DNVLDHLTGRSC
1012
P04977.1




PERTUSSIS









130

BORDETELLA

DNVLDHLTGRSCQ
1013
P04977.1




PERTUSSIS









131
MEASLES
DPDKILTYIAA
1014
AAF02706.1



MORBILLIVIRUS








132
RUBELLA VIRUS
DPGDLVEYIMNYTGNQQSR
1015
P07566.1



STRAIN THERIEN








133
RUBELLA VIRUS
DPLLRTAP
1016
CAJ88851.1





134
RUBELLA VIRUS
DPLLRTAPGPGEVWVTPVIGSQ
1017
CAJ88851.1





135
MEASLES VIRUS
DPQDSRRSAEPLL
1018
P04851.1



STRAIN






EDMONSTON








136
MEASLES VIRUS
DPVIDRLYLSSHRGV
1019
P08362.1



STRAIN






EDMONSTON








137
MEASLES VIRUS
DQILRSMKGLSSTSI
1020
P69353.1



STRAIN






EDMONSTON








138
MEASLES
DQYCADVAAEELMNA
1021
P06830.1



MORBILLIVIRUS








139
MEASLES VIRUS
DSESGGHITH
1022
P08362.1



STRAIN






EDMONSTON








140
MEASLES VIRUS
DTASESSQDPQDS
1023
P04851.1



STRAIN






EDMONSTON








141
RUBELLA VIRUS
DTVMSVFALASYVQH
1024
BAA28178.1





142

BORDETELLA

DVFQNGFTAWGNND
1025
P04977.1




PERTUSSIS









143
RUBELLA VIRUS
DVGAVPPGKFVTAAL
1026
BAA28178.1





144

CORYNEBACTERIUM

DVNKSKTHISVNGRKI
1027
CAE11230.1




DIPHTHERIAE









145
RUBELLA VIRUS
DVSCEGLGAWVPAAP
1028
BAA28178.1





146
RUBELLA VIRUS
DWASPVCQRHSPDCSRLVGATPERPRL
1029
P07566.1



STRAIN THERIEN








147
MEASLES VIRUS
EARESYRETGPSR
1030
P04851.1



STRAIN






EDMONSTON








148

BORDETELLA

EAVEAERAGRGTG
1031
ACI04548.1




PERTUSSIS









149
MEASLES VIRUS
EDAKELLESSDQILR
1032
P69353.1



STRAIN






EDMONSTON








150
MEASLES VIRUS
EDRRVKQSRGEAR
1033
P04851.1



STRAIN






EDMONSTON








151
MEASLES VIRUS
EDSITIPYQGSGKGV
1034
P08362.1



STRAIN






EDMONSTON








152
RUBELLA VIRUS
EEAFTYLCTAPGCAT
1035
BAA28178.1





153
MEASLES VIRUS
EGFNMILGTILAQIW
1036
P04851.1



STRAIN






EDMONSTON








154
MEASLES VIRUS
EGFNMILGTILAQIWVLLAK
1037
P04851.1



STRAIN






EDMONSTON








155
RUBELLA VIRUS
EHPFCNTPHGQLEVQ
1038
BAA28178.1





156
MEASLES VIRUS
EISDIEVQDPEGFNM
1039
P04851.1



STRAIN






EDMONSTON








157
MEASLES VIRUS
EISDIEVQDPEGFNMILGTI
1040
P04851.1



STRAIN






EDMONSTON








158
MEASLES VIRUS
EKPNLSSKRSE
1041
P08362.1



STRAIN






EDMONSTON








159
MEASLES VIRUS
ELKLAALCHGEDSIT
1042
P08362.1



STRAIN






EDMONSTON








160
MEASLES
ELMNALVNSTLLETR
1043
P06830.1



MORBILLIVIRUS








161
MEASLES VIRUS
ELPRL
1044
P04851.1



STRAIN






EDMONSTON








162
MEASLES
ENPEWAPLKDNRIPSYGVLSVDL
1045
AAR89413.1



MORBILLIVIRUS








163
MEASLES VIRUS
EPIRD ALNAMTQNIR
1046
P69353.1



STRAIN






EDMONSTON








164
MEASLES VIRUS
EQVGMSRPGLKPDLT
1047
P69353.1



STRAIN






EDMONSTON








165
RUBELLA VIRUS
ERPRLRLV
1048
CAJ88851.1





166
RUBELLA VIRUS
ERPRLRLVDADDPLL
1049
BAA28178.1





167
MEASLES VIRUS
ESPGQLIQRITDDPDVS
1050
P04851.1



CAM/RB








168
MEASLES VIRUS
ESRGIKARITHVDTE
1051
P69353.1



STRAIN






EDMONSTON








169
MEASLES VIRUS
ESSCTFMPEGTVCSQ
1052
P69353.1



STRAIN






EDMONSTON








170
MEASLES VIRUS
ESSQDPQDSRRSA
1053
P04851.1



STRAIN






EDMONSTON








171
MEASLES VIRUS
ETRTTNQFLAVSKGN
1054
P08362.1



STRAIN






EDMONSTON








172
MEASLES VIRUS
EVDGDVKLSSNLVIL
1055
P08362.1



STRAIN






EDMONSTON








173
MEASLES VIRUS
EVNGVTIQV
1056
P26031.1



STRAIN






EDMONSTON-B








174
RUBELLA VIRUS
EVWVTPVI
1057
CAJ88851.1





175
RUBELLA VIRUS
EVWVTPVIGSQA
1058
BAA19893.1





176
RUBELLA VIRUS
EWAAAHWWQLTLGAT
1059
BAA28178.1





177
MEASLES
EWIPRFKVSPYLFTV
1060
P06830.1



MORBILLIVIRUS








178

BORDETELLA

FEYVDTYGDNAG
1061
P04977.1




PERTUSSIS









179
MEASLES
FGPLITHGSGMDLYK
1062
P06830.1



MORBILLIVIRUS








180
MEASLES VIRUS
FIFD ALAEV
1063
ABK40531.1



STRAIN






EDMONSTON








181
MEASLES VIRUS
FISDKIKFL
1064
P08362.1



STRAIN






EDMONSTON








182
MEASLES VIRUS
FKRNKDKPPITSGSG
1065
P04851.1



STRAIN






EDMONSTON








183
MEASLES VIRUS
FKRNKDKPPITSGSGGAIRG
1066
P04851.1



STRAIN






EDMONSTON








184
RUBELLA VIRUS
FKTVRPVALPRTLAP
1067
BAA28178.1





185
MEASLES VIRUS
FLMDRHIIV
1068
ABK40531.1



STRAIN






EDMONSTON








186
MEASLES VIRUS
FMAVLLTLQTPTGQI
1069
P69353.1



STRAIN






EDMONSTON








187
MEASLES VIRUS
FMPEGTVCSQNALYP
1070
P69353.1



STRAIN






EDMONSTON








188
MEASLES
FMYMSLLGV
1071
AAN09804.1



MORBILLIVIRUS








189
MEASLES VIRUS
FNVPIKEAGEDCHAP
1072
P08362.1



STRAIN






EDMONSTON








190
MEASLES
FRDLTWCINPPERIK
1073
AAC35876.2



MORBILLIVIRUS








191
MEASLES VIRUS
FSHDDPISSDQSRFG
1074
P04851.1



STRAIN






EDMONSTON








192
MEASLES VIRUS
FSHDDPISSDQSRFGWFENK
1075
P04851.1



STRAIN






EDMONSTON








193
MEASLES
FTDLVKFISDKIKFL
1076
P06830.1



MORBILLIVIRUS








194
MEASLES
FTWDQKLWCRHFCVL
1077
P06830.1



MORBILLIVIRUS








195

BORDETELLA

FVRDGQSVIGACASPYEGRYRDLYDALRRL
1078
SRC280066




PERTUSSIS

LY







196

BORDETELLA

FVRSGQPVIGACTSPYDGKYWSILYSRLRK
1079
SRC280066




PERTUSSIS

MLY







197
MEASLES
FYKDNPHPKGSRIVI
1080
P06830.1



MORBILLIVIRUS








198

BORDETELLA

GAASSYFEYVDTYG
1081
ACI04548.1




PERTUSSIS









199

BORDETELLA

GAFDLKTTFCIMTTRNTGQPA
1082
AAA83981.1




PERTUSSIS









200
MEASLES VIRUS
GALIGILSLFVESPG
1083
P04851.1



STRAIN






EDMONSTON








201
MEASLES VIRUS
GALIGILSLFVESPGQLIQR
1084
P04851.1



STRAIN






EDMONSTON








202
RUBELLA VIRUS
GATPERPR
1085
CAJ88851.1





203

BORDETELLA

GAYGRCPNGTRALTVAELRGNAEL
1086
P04979.1




PERTUSSIS









204
RUBELLA VIRUS
GCFAPWDLEATGACI
1087
BAA28178.1





205
MEASLES
GDINKVLEKLGYS
1088
BAB60865.1



MORBILLIVIRUS








206
MEASLES
GDINKVLEKLGYSGGDLLG
1089
AAL29688.1



MORBILLIVIRUS








207
RUBELLA VIRUS
GDLRAVHHRPVPA
1090
CAA28880.1





208
RUBELLA VIRUS
GDLVEYIMNYTGNQQ
1091
BAA28178.1





209
MEASLES VIRUS
GDSSITTRSRLLDRL
1092
P04851.1



STRAIN






EDMONSTON








210
MEASLES VIRUS
GDSSITTRSRLLDRLVRLIG
1093
P04851.1



STRAIN






EDMONSTON








211
MEASLES
GEDCHAPTYLPAEVD
1094
P06830.1



MORBILLIVIRUS








212
MEASLES VIRUS
GELSTLESLMNLYQQ
1095
P04851.1



STRAIN






EDMONSTON








213
MEASLES VIRUS
GELSTLESLMNLYQQMGKPA
1096
P04851.1



STRAIN






EDMONSTON








214
MUMPS
GEQARYLALLEA
1097
P21186.1



RUBULAVIRUS








215
RUBELLA VIRUS
GEVWVT
1098
BAA19893.1





216
RUBELLA VIRUS
GEVWVTPV
1099
CAJ88851.1





217
RUBELLA VIRUS
GEVWVTPVIGSQAR
1100
BAA19893.1





218

BORDETELLA

GEYGGVIKDGTPGGA
1101
AAA83981.1




PERTUSSIS









219
RUBELLA VIRUS
GFLSGVGPMRLRHGADT
1102
SRC265968





220
MEASLES VIRUS
GFRASDVETAEGGEIHELLRLQ
1103
PO3422.1



STRAIN






EDMONSTON-B








221

BORDETELLA

GGAVPGGAVPGGAVPGGFGPGGFGP
1104
P14283.3




PERTUSSIS









222

BORDETELLA

GGAVPGGAVPGGFGPGGFGPGGFGP
1105
CAA09475.1




PERTUSSIS









223

BORDETELLA

GGAVPGGAVPGGFGPGGFGPGGFGPGGFGP
1106
CAA09474.1




PERTUSSIS









224
MEASLES
GGHITHSGMVGMGVS
1107
P06830.1



MORBILLIVIRUS








225
MEASLES
GILESRGIKARITHVDTESY
1108
P26032.1



MORBILLIVIRUS








226

BORDETELLA

GITGETTTTEYSNARYV
1109
CAD44970.1




PERTUSSIS









227
MEASLES VIRUS
GKEDRRVKQSRGE
1110
P04851.1



STRAIN






EDMONSTON








228

BORDETELLA

GKVTNGS
1111
ACI16088.1




PERTUSSIS









229
RUBELLA VIRUS
GLGAWVPAAPCARIW
1112
BAA28178.1





230
MEASLES VIRUS
GLIGIPALICCCRGR
1113
P69353.1



STRAIN






EDMONSTON








231
RUBELLA VIRUS
GLLACCAKCLYYLRGAIAPR
1114
P07566.1



STRAIN THERIEN








232
MEASLES
GLLAIAGIRLHRAAI
1115
P06830.1



MORBILLIVIRUS








233
RUBELLA VIRUS
GLQPRADMAAPPTLPQ
1116
NP_740663.1





234
MEASLES
GMGVSCTVTREDGTNRR
1117
AAR89413.1



MORBILLIVIRUS








235
MEASLES
GMYGGTYLVEKP
1118
AAR89413.1



MORBILLIVIRUS








236

BORDETELLA

GNAELQTYLRQITPGWSIYGLYDGTYLG
1119
P04979.1




PERTUSSIS









237
RUBELLA VIRUS
GNCHLTVNGEDVGAV
1120
BAA28178.1





238

BORDETELLA

GNNDNVLDHLTGR
1121
P04977.1




PERTUSSIS









239

BORDETELLA

GNNDNVLDHLTGRSC
1122
P04977.1




PERTUSSIS









240
MEASLES VIRUS
GNRFILSQGNLIANC
1123
P69353.1



STRAIN






EDMONSTON








241
RUBELLA VIRUS
GNRGRGQRRDWSRAPPPPEERQETRSQTPA
1124
P07566.1




PKPS







242
RUBELLA VIRUS
GPGEVWVT
1125
CAJ88851.1





243
RUBELLA VIRUS
GPMRLRHGADTRCGRLI
1126
P07566.1



STRAIN THERIEN








244

BORDETELLA

GPNHTKV
1127
ACI16083.1




PERTUSSIS









245
MEASLES VIRUS
GPRQAQVSF
1128
P10050.1



STRAIN HALLE








246
MEASLES VIRUS
GPRQAQVSFLQGDQS
1129
P04851.1



STRAIN






EDMONSTON








247
MEASLES VIRUS
GPRQAQVSFLQGDQSENELP
1130
P04851.1



STRAIN






EDMONSTON








248
MEASLES
GRGYNVSSIVTMTSQ
1131
P06830.1



MORBILLIVIRUS








249

BORDETELLA

GRTPFII
1132
ACI16083.1




PERTUSSIS









250
RUBELLA VIRUS
GSPNCHGPDWASPVC
1133
BAA28178.1





251
RUBELLA VIRUS
GSQARKCGLHIRAGP
1134
BAA28178.1





252

BORDETELLA

GSSNSAFVSTSSSRR
1135
P04977.1




PERTUSSIS









253
MEASLES VIRUS
GSTKSCARTLVSGSF
1136
P69353.1



STRAIN






EDMONSTON








254
RUBELLA VIRUS
GSYYKQYHPTACEVE
1137
BAA28178.1





255
RUBELLA VIRUS
GTHTTAVSETRQTWA
1138
BAA28178.1





256
MEASLES VIRUS
GTIINQDPDKILTYI
1139
P69353.1



STRAIN






EDMONSTON








257

BORDETELLA

GTLVRMAPVIG
1140
ADA85124.1




PERTUSSIS









258
MEASLES VIRUS
GTPLDIDTASESS
1141
P04851.1



STRAIN






EDMONSTON








259

BORDETELLA

GTYLGQAYGGIIKDAPPGAGFIYRETFC
1142
P04979.1




PERTUSSIS









260

BORDETELLA

GVATKGLGVHAKSSDWG
1143
P15318.2




PERTUSSIS









261

CORYNEBACTERIUM

GVLLPTIPGKLDVNKSKTHI
1144
AAV70486.1




DIPHTHERIAE









262
MEASLES
GVLSVDLSLTVELKI
1145
P06830.1



MORBILLIVIRUS








263
MEASLES
GVPIELQVECFTWDQ
1146
P06830.1



MORBILLIVIRUS








264
MEASLES VIRUS
GVSCTVTREDGTNRR
1147
P08362.1



STRAIN






EDMONSTON








265
MEASLES VIRUS
GVSYNIGSQEWYTTV
1148
P69353.1



STRAIN






EDMONSTON








266
MEASLES VIRUS
GYNVSSIVTMTSQGM
1149
P08362.1



STRAIN






EDMONSTON








267
MEASLES
HFCVLADSESGGHIT
1150
P06830.1



MORBILLIVIRUS








268
MEASLES
HGEDSITIPYQGSGK
1151
P06830.1



MORBILLIVIRUS








269
RUBELLA VIRUS
HGPDWASP
1152
BAA19893.1





270
RUBELLA VIRUS
HGPDWASPVCQRHSP
1153
BAA28178.1





271
RUBELLA VIRUS
HGPDWASPVCQRHSPDCSRLVG
1154
CAJ88851.1





272
RUBELLA VIRUS
HGPDWASPVCQRHSPDCSRLVGATPERPRL
1155
CAJ88851.1



STRAIN M33
RLV







273
MEASLES VIRUS
HITHSGMEGMGVSCT
1156
P08362.1



STRAIN






EDMONSTON








274
MEASLES
HKSLSTNLDVTNSIE
1157
P06830.1



MORBILLIVIRUS








275
MEASLES VIRUS
HLMIDRPYV
1158
P08362.1



STRAIN






EDMONSTON








276
MEASLES VIRUS
HLPTGTPLDIDTA
1159
P04851.1



STRAIN






EDMONSTON








277
MEASLES VIRUS
HLPTGTPLDIDTATESSQDPQDSR
1160
Q77M43.1



STRAIN






EDMONSTON-B








278
MEASLES
HMTNYLEQPVSNDLS
1161
P06830.1



MORBILLIVIRUS








279
MEASLES VIRUS
HQSLVIKLMPNITLL
1162
P69353.1



STRAIN






EDMONSTON








280
MEASLES
HRAAIYTAEIHKSLS
1163
P06830.1



MORBILLIVIRUS








281

BORDETELLA

HRMQEAVEAERAGRGTGH
1164
P04977.1




PERTUSSIS









282
MEASLES VIRUS
HVDTESYFIVLSIAY
1165
P69353.1



STRAIN






EDMONSTON








283
MEASLES VIRUS
HWGNLSKIGVVGIGS
1166
P69353.1



STRAIN






EDMONSTON








284
RUBELLA VIRUS
HWWQLTLGATCALPL
1167
BAA28178.1





285
RUBELLA VIRUS
HYRNASDVLPGHWLQGGWGCYNL
1168
NP_740663.1





286
MEASLES VIRUS
IDLGPPISLERLDVG
1169
P69353.1



STRAIN






EDMONSTON








287
MEASLES VIRUS
IEAIRQAGQEMILAV
1170
P69353.1



STRAIN






EDMONSTON








288
RUBELLA VIRUS
IETRSARHP
1171
CAA28880.1



STRAIN M33








289
MEASLES VIRUS
IGSQEWYTTVPKYVA
1172
P69353.1



STRAIN






EDMONSTON








290
MEASLES VIRUS
IKGVIVHRLEGVSYN
1173
P69353.1



STRAIN






EDMONSTON








291
MEASLES VIRUS
IKHIIIVPIPGDSSI
1174
P04851.1



STRAIN






EDMONSTON








292
MEASLES VIRUS
IKHIIIVPIPGDSSITTRSR
1175
P04851.1



STRAIN






EDMONSTON








293

BORDETELLA

IKLKDCP
1176
ACI16083.1




PERTUSSIS









294
MEASLES VIRUS
IKLMPNITLLNNCTR
1177
P69353.1



STRAIN






EDMONSTON








295
MEASLES
ILLERLDVGT
1178
AAF85664.1



MORBILLIVIRUS








296
MEASLES
ILPGQDLQYV
1179
P08362.1



MORBILLIVIRUS








297
MEASLES VIRUS
ILTYIAADHCPVVEV
1180
P69353.1



STRAIN






EDMONSTON








298
MEASLES
INQDPDKILTY
1181
AAL29688.1



MORBILLIVIRUS








299
MEASLES VIRUS
IPRFKVSPYLFNVPI
1182
P08362.1



STRAIN






EDMONSTON








300
MEASLES VIRUS
IQALSYALGGDINKV
1183
P69353.1



STRAIN






EDMONSTON








301
RUBELLA VIRUS
IRAGPYGHATVEMPE
1184
BAA28178.1





302

BORDETELLA

IRMGTDK
1185
ACI16088.1




PERTUSSIS









303
MEASLES VIRUS
ISNFDESSCTFMPEG
1186
P69353.1



STRAIN






EDMONSTON








304
MEASLES VIRUS
ITAGIALHQSMLNSQ
1187
P69353.1



STRAIN






EDMONSTON








305
MEASLES VIRUS
ITDDPDVSIRLLEVV
1188
P04851.1



STRAIN






EDMONSTON








306
MEASLES VIRUS
ITDDPDVSIRLLEVVQSDQS
1189
P04851.1



STRAIN






EDMONSTON








307

BORDETELLA

ITTYV
1190
ACI16083.1




PERTUSSIS









308
MEASLES VIRUS
IVEAGLASFILTIKF
1191
P04851.1



STRAIN






EDMONSTON








309
MEASLES VIRUS
IVEAGLASFILTIKFGIETM
1192
P04851.1



STRAIN






EDMONSTON








310
MEASLES VIRUS
IVEAGLASFILTIKFGIETMYPALG
1193
P04851.1



STRAIN






EDMONSTON








311
RUBELLA VIRUS
KALEAQSRALRAELAA
1194
P07566.1





312
MEASLES VIRUS
KARITHVDTESYFIV
1195
P69353.1



STRAIN






EDMONSTON








313
RUBELLA VIRUS
KCGLHIRAGPYGHAT
1196
BAA28178.1





314
MEASLES VIRUS
KCYTTGTIINQDPDK
1197
P69353.1



STRAIN






EDMONSTON








315
MEASLES VIRUS
KDNPHPKGSR
1198
P08362.1



STRAIN






EDMONSTON








316
MEASLES VIRUS
KDNRIPSYGVLSVDL
1199
P08362.1



STRAIN






EDMONSTON








317
MEASLES
KFLNPDREYDFRDLT
1200
AAC35876.2



MORBILLIVIRUS








318
RUBELLA VIRUS
KFVTAALLN
1201
BAA28178.1





319
MEASLES VIRUS
KGNCSGPTTIR
1202
P08362.1



STRAIN






EDMONSTON








320
MEASLES
KIKFLNPDREYDFRD
1203
P06830.1



MORBILLIVIRUS








321
RUBELLA VIRUS
KIVDGGCFAPWDLEA
1204
BAA28178.1





322
MEASLES VIRUS
KLGLKLLRYYTEILS
1205
P69353.1



STRAIN






EDMONSTON








323
MEASLES VIRUS
KLGVWKSPTDMQSWV
1206
P08362.1



STRAIN






EDMONSTON








324

BORDETELLA

KLKECPQ
1207
ACI16088.1




PERTUSSIS









325
MEASLES
KLLRYYTEI
1208
P26031.1



MORBILLIVIRUS








326
MEASLES VIRUS
KLMPFSGDFV
1209
ABK40531.1



STRAIN






EDMONSTON








327
MEASLES
KLMPNITLL
1210
P26031.1



MORBILLIVIRUS








328
MEASLES
KLRMETCFQQACKGKIQALCENPEWA
1211
AAR89413.1



MORBILLIVIRUS








329
MEASLES
KLWCRHFCV
1212
P08362.1



MORBILLIVIRUS








330
MEASLES VIRUS
KLWCRHFCVL
1213
P08362.1



STRAIN






EDMONSTON








331
MEASLES
KLWESPQEI
1214
BAB60863.1



MORBILLIVIRUS








332
MEASLES VIRUS
KMSSAVGFV
1215
ABO69699.1



STRAIN






EDMONSTON








333
MEASLES VIRUS
KMSSAVGFVPDTGPASR
1216
P03422.1



STRAIN






EDMONSTON-B








334

BORDETELLA

KMVYATN
1217
ACI16083.1




PERTUSSIS









335
MEASLES VIRUS
KPDLTGTSKSYVRSL
1218
P69353.1



STRAIN






EDMONSTON








336
MEASLES
KPNLSSKRSELSQLS
1219
P08362.1



MORBILLIVIRUS








337
MEASLES VIRUS
KPNLSSKRSELSQLSMYRVF
1220
P08362.1



STRAIN






EDMONSTON








338
MEASLES VIRUS
KQSRGEARESYRETG
1221
P04851.1



STRAIN






EDMONSTON








339
MEASLES VIRUS
KQSRGEARESYRETGPSRAS
1222
P04851.1



STRAIN






EDMONSTON








340
MEASLES VIRUS
KRFAGVVLAGAALGV
1223
P69353.1



STRAIN






EDMONSTON








341
MEASLES VIRUS
KRTPGNKPRIAEMIC
1224
P04851.1



STRAIN






EDMONSTON








342
MEASLES VIRUS
KRTPGNKPRIAEMICDIDTY
1225
P04851.1



STRAIN






EDMONSTON








343
MEASLES
KSNHNNVYWLTIPPMKNLALGVINTL
1226
AAR89413.1



MORBILLIVIRUS








344
MEASLES
KVSPYLFNV
1227
P08362.1



MORBILLIVIRUS








345

BORDETELLA

KVVQLPKISKNALKANG
1228
ACI16083.1




PERTUSSIS









346

BORDETELLA

KVVQLPKISKNALRNDG
1229
ACI16087.1




PERTUSSIS









347

BORDETELLA

LAHRRIPPENIR
1230
P04977.1




PERTUSSIS









348

BORDETELLA

LALALWAGFALS
1231
P11092.1




PERTUSSIS









349
RUBELLA VIRUS
LAPGGGNCHLTVNGE
1232
BAA28178.1





350
MEASLES VIRUS
LAQIWVLLAKAVTAP
1233
P04851.1



STRAIN






EDMONSTON








351
MEASLES VIRUS
LAQIWVLLAKAVTAPDTAAD
1234
P04851.1



STRAIN






EDMONSTON








352
RUBELLA VIRUS
LASYFNPGGSYYKQYHPTACEVEPAFGHS
1235
BAA19893.1





353
MEASLES
LAVSKGNCSGPTTIR
1236
P06830.1



MORBILLIVIRUS








354
MEASLES VIRUS
LCENPEWAPLKDNRI
1237
P08362.1



STRAIN






EDMONSTON








355
MEASLES
LDRLVRLIG
1238
ABI5411O.1



MORBILLIVIRUS








356

CORYNEBACTERIUM

LEEEGVTPL
1239
P33120.2




DIPHTHERIAE









357

BORDETELLA

LEHRMQEAVEAERAGRGTGHFI
1240
CAD44970.1




PERTUSSIS









358
MEASLES VIRUS
LEKLGYSGGDLLGIL
1241
P69353.1



STRAIN






EDMONSTON








359
MEASLES VIRUS
LEQPVSNDLS
1242
P08362.1



STRAIN






EDMONSTON








360
MEASLES VIRUS
LERKWLDVVRNIIAE
1243
P04851.1



STRAIN






EDMONSTON








361
MEASLES VIRUS
LERKWLDVVRNIIAEDLSLR
1244
P04851.1



STRAIN






EDMONSTON








362
MEASLES VIRUS
LFGPSLRDPISAEIS
1245
P69353.1



STRAIN






EDMONSTON








363
MEASLES
LGELKLAALCHGEDS
1246
P06830.1



MORBILLIVIRUS








364
MEASLES VIRUS
LGGKEDRRVKQSR
1247
P04851.1



STRAIN






EDMONSTON








365
RUBELLA VIRUS
LGHDGHHGGTLRVGQHHRNASDVL
1248
ABD64214.1





366
RUBELLA VIRUS
LGSPNCHGPDWASPVCQRHS
1249
P07566.1



STRAIN THERIEN








367
RUBELLA VIRUS
LGSPNCHGPDWASPVCQRHSPDCSRLV
1250
P07566.1



STRAIN THERIEN








368
RUBELLA VIRUS
LHDPDTEAPTEACVTSWL
1251
ABD64214.1





369
MEASLES VIRUS
LIANCASILCKCYTT
1252
P69353.1



STRAIN






EDMONSTON








370
MEASLES
LIGLLAIAGIRLHRAAIYTAEIHK
1253
AAR89413.1



MORBILLIVIRUS








371
MEASLES VIRUS
LIPSMNQLSCDLIGQ
1254
P69353.1



STRAIN






EDMONSTON








372
MEASLES VIRUS
LKIKIASGFGPLITH
1255
P08362.1



STRAIN






EDMONSTON








373
MEASLES
LKIKIASGFGPLITHGSGMDLYK
1256
AAR89413.1



MORBILLIVIRUS








374

BORDETELLA

LKLYFEP
1257
ACI16088.1




PERTUSSIS









375
MEASLES VIRUS
LLAVLFVMFL
1258
P08362.1



STRAIN






EDMONSTON








376
MEASLES VIRUS
LLDRLVRLIGNPDVS
1259
P04851.1



STRAIN






EDMONSTON








377
MEASLES VIRUS
LLDRLVRLIGNPDVSGPKLT
1260
P04851.1



STRAIN






EDMONSTON








378
MEASLES VIRUS
LLESSDQILRSMKGL
1261
P69353.1



STRAIN






EDMONSTON








379
MEASLES
LLETRTTNQFLAVSK
1262
P06830.1



MORBILLIVIRUS








380
MEASLES VIRUS
LLEVVQSDQSQSGLT
1263
P04851.1



STRAIN






EDMONSTON








381
MEASLES VIRUS
LLEVVQSDQSQSGLTFASR
1264
P04851.1



STRAIN






EDMONSTON








382
MEASLES VIRUS
LLEVVQSDQSQSGLTFASRG
1265
P04851.1



STRAIN






EDMONSTON








383
MEASLES
LLGILESRGIKARIT
1266
AAL29688.1



MORBILLIVIRUS








384
RUBELLA VIRUS
LLRTAPGP
1267
CAJ88851.1





385
MEASLES VIRUS
LLRYYTEILSLFGPS
1268
P69353.1



STRAIN






EDMONSTON








386
RUBELLA VIRUS
LLVPWVLIFMVCRRACRRRG
1269
P07566.1



STRAIN THERIEN








387
MEASLES VIRUS
LLWRSRCKIV
1270
ABK40528.1



STRAIN






EDMONSTON








388
MEASLES
LLWSYAMGV
1271
P04851.1



MORBILLIVIRUS








389
MEASLES VIRUS
LLWSYAMGVGVELEN
1272
P04851.1



STRAIN






EDMONSTON








390
MEASLES VIRUS
LLWSYAMGVGVELENSMGGL
1273
P04851.1



STRAIN






EDMONSTON








391
MEASLES
LMIDRPYVL
1274
P08362.1



MORBILLIVIRUS








392
MEASLES VIRUS
LNAMTQNIRPVQSVA
1275
P69353.1



STRAIN






EDMONSTON








393
RUBELLA VIRUS
LNTPPPYQVSCGGES
1276
BAA28178.1





394
MEASLES VIRUS
LRDPISAEISIQALS
1277
P69353.1



STRAIN






EDMONSTON








395

BORDETELLA

LRGSGDLQEYLRHVTR
1278
AAA83981.1




PERTUSSIS









396
RUBELLA VIRUS
LRLVDADD
1279
CAJ88851.1





397
RUBELLA VIRUS
LRLVDADDPLLR
1280
BAA19893.1





398
RUBELLA VIRUS
LRLVDADDPLLRTAPGPGEVWVTPVIGSQA
1281
BAA19893.1




R







399

BORDETELLA

LRRLLYMIYMSGLAVRVHVSKEEQYYDY
1282
P04979.1




PERTUSSIS









400
RUBELLA VIRUS
LRTAPGPG
1283
CAJ88851.1





401
RUBELLA VIRUS
LRVGQHYRNASDVLPGHWLQ
1284
NP_740663.1





402

BORDETELLA

LRYLA
1285
ACI16088.1




PERTUSSIS









403
MEASLES VIRUS
LSCKPWQESRKNK
1286
P04851.1



STRAIN






EDMONSTON








404
MEASLES
LSEIKGVIVHRLEGV
1287
AAL29688.1



MORBILLIVIRUS








405
MEASLES VIRUS
LSIAYPTLSEIKGVI
1288
P69353.1



STRAIN






EDMONSTON








406
MEASLES VIRUS
LSLLDLYLGRGYNVS
1289
P08362.1



STRAIN






EDMONSTON








407
MEASLES VIRUS
LSQGNLIANCASILC
1290
P69353.1



STRAIN






EDMONSTON








408
MEASLES
LSSHRGVIADNQAKW
1291
P06830.1



MORBILLIVIRUS








409
MEASLES VIRUS
LSVDLSLTVELKIKI
1292
P08362.1



STRAIN






EDMONSTON








410

BORDETELLA

LTGISICNPGSSLC
1293
AAA83981.1




PERTUSSIS









411
MEASLES VIRUS
LTIKFGIETMYPALG
1294
P04851.1



STRAIN






EDMONSTON








412
MEASLES VIRUS
LTIKFGIETMYPALGLHEFA
1295
P04851.1



STRAIN






EDMONSTON








413
MEASLES VIRUS
LTLQTPTGQIHWGNL
1296
P69353.1



STRAIN






EDMONSTON








414
RUBELLA VIRUS
LVD ADDPL
1297
CAJ88851.1





415
RUBELLA VIRUS
LVDADDPLLR
1298
BAA19893.1





416
MEASLES VIRUS
LVEKPNLSSKRSELS
1299
P08362.1



STRAIN






EDMONSTON








417
RUBELLA VIRUS
LVGATPE
1300
BAA19893.1





418
RUBELLA VIRUS
LVGATPER
1301
CAJ88851.1





419
MEASLES
LVKLGVWKSPTGMQS
1302
P06830.1



MORBILLIVIRUS








420
MEASLES VIRUS
LVSGSFGNRFILSQGNLI
1303
P26031.1



STRAIN






EDMONSTON-B








421
MEASLES VIRUS
LYKSNHNNVYWLTIP
1304
P08362.1



STRAIN






EDMONSTON








422
MEASLES VIRUS
LYPMSPLLQECLRGSTKSCARTLVS
1305
P69353.1



STRAIN






EDMONSTON








423
RUBELLA VIRUS
MASTTPITMEDLQKALEA
1306
P07566.1





424
RUBELLA VIRUS
MASTTPITMEDLQKALEAQSR
1307
ABD64200.1





425
RUBELLA VIRUS
MASTTPITMEDLQKALEAQSRALRAELAA
1308
P07566.1



STRAIN THERIEN








426
RUBELLA VIRUS
MASTTPITMEDLQKALEAQSRALRAGLAA
1309
ABD64200.1





427
RUBELLA VIRUS
MASTTPITMEDLQKALETQSRVLRAGLTA
1310
CAA33016.1



VACCINE STRAIN






RA27/3








428
MEASLES VIRUS
MATLLRSLALFKRNK
1311
P04851.1



STRAIN






EDMONSTON








429
MEASLES VIRUS
MATLLRSLALFKRNKDKPPI
1312
P04851.1



STRAIN






EDMONSTON








430
MEASLES
MDLYKSNHNNVYWLT
1313
P06830.1



MORBILLIVIRUS








431
RUBELLA VIRUS
MEDLQKALEAQSRA
1314
P07566.1





432
RUBELLA VIRUS
MEDLQKALEAQSRALRAELAA
1315
P07566.1





433
MEASLES VIRUS
MGLKVNVSAIFMAVL
1316
P69353.1



STRAIN






EDMONSTON








434
MEASLES
MIDRPYVLLAVLFVM
1317
P06830.1



MORBILLIVIRUS








435
MEASLES VIRUS
MILAVQGVQDYINNE
1318
P69353.1



STRAIN






EDMONSTON








436
MEASLES VIRUS
MLNSQAIDNLRASLE
1319
P69353.1



STRAIN






EDMONSTON








437
MEASLES VIRUS
MNALVNSTLLETRTT
1320
P08362.1



STRAIN






EDMONSTON








438
RUBELLA VIRUS
MNYTGNQQSRWGLGSPNCHGPDWASPVCQR
1321
BAA19893.1




HS







439
MEASLES VIRUS
MQSWVPLSTDDPVID
1322
P08362.1



STRAIN






EDMONSTON








440
MEASLES
MSLSLLDLYLGRGYN
1323
P06830.1



MORBILLIVIRUS








441
MEASLES VIRUS
MSPLLQECLRGSTKS
1324
P69353.1



STRAIN






EDMONSTON








442
MEASLES
MSPQRDRINAFYKDN
1325
P06830.1



MORBILLIVIRUS








443
MEASLES
MYRVFEVSVIRNPGL
1326
P06830.1



MORBILLIVIRUS








444
MEASLES VIRUS
NALYPMSPLLQECLR
1327
P69353.1



STRAIN






EDMONSTON








445
RUBELLA VIRUS
NCHGPDWASPVCQRHSPDCSRLVGAT
1328
P07566.1



STRAIN THERIEN








446
MEASLES VIRUS
NFGRSYFDPAYFRLG
1329
P04851.1



STRAIN






EDMONSTON








447
MEASLES VIRUS
NFGRSYFDPAYFRLGQEMVR
1330
P04851.1



STRAIN






EDMONSTON








448
RUBELLA VIRUS
NGTQRACTFWAVNAY
1331
BAA28178.1





449
MEASLES VIRUS
NGVTIQVGSRRYPDA
1332
P69353.1



STRAIN






EDMONSTON








450
MEASLES VIRUS
NIIAEDLSLRRFMVA
1333
P04851.1



STRAIN






EDMONSTON








451
MEASLES VIRUS
NIIAEDLSLRRFMVALILDI
1334
P04851.1



STRAIN






EDMONSTON








452
MEASLES VIRUS
NITLLNNCTRVEIAE
1335
P69353.1



STRAIN






EDMONSTON








453
MEASLES
NLALGVINTLEWIPR
1336
P06830.1



MORBILLIVIRUS








454

CORYNEBACTERIUM

NLFQVVHWSYNRPAYSPG
1337
SRC280292




DIPHTHERIAE









455
MEASLES VIRUS
NLVILPGQDLQYVLA
1338
P08362.1



STRAIN






EDMONSTON








456
MEASLES VIRUS
NLYQQMGKPAPYMVN
1339
P04851.1



STRAIN






EDMONSTON








457
MEASLES VIRUS
NLYQQMGKPAPYMVNLENSI
1340
P04851.1



STRAIN






EDMONSTON








458
MEASLES VIRUS
NNCTRVEIAEYRRLL
1341
P69353.1



STRAIN






EDMONSTON








459
MEASLES VIRUS
NPDREYDFRD
1342
P08362.1



STRAIN






EDMONSTON








460
MEASLES VIRUS
NPDVSGPKL
1343
P10050.1



STRAIN HALLE








461
MEASLES VIRUS
NPDVSGPKLTGALIG
1344
P04851.1



STRAIN






EDMONSTON








462
MEASLES VIRUS
NPDVSGPKLTGALIGILSLF
1345
P04851.1



STRAIN






EDMONSTON








463
MEASLES
NPPERIKLDYDQYCA
1346
P06830.1



MORBILLIVIRUS








464
MEASLES
NQAKWAVPTTRTDDK
1347
P06830.1



MORBILLIVIRUS








465
MEASLES
NQDPDKILTYIAADH
1348
AAF02706.1



MORBILLIVIRUS








466
MEASLES VIRUS
NQLSCDLIGQKLGLK
1349
P69353.1



STRAIN






EDMONSTON








467
RUBELLA VIRUS
NQQSRWGLGSPNCHGPDWASPVCQRHS
1350
ABD64214.1





468
MUMPS
NSTLGVKSAREF
1351
ABP48111.1



RUBULAVIRUS








469
RUBELLA VIRUS
NTPHGQLEVQVPPDP
1352
BAA28178.1





470
MEASLES VIRUS
NVSAIFMAVLLTLQT
1353
P69353.1



STRAIN






EDMONSTON








471
MEASLES
PAEVDGDVKLSSNLV
1354
P06830.1



MORBILLIVIRUS








472
RUBELLA VIRUS
PAFGHSDAACWGFPT
1355
BAA28178.1





473
MEASLES VIRUS
PALICCCRGRCNKKG
1356
P69353.1



STRAIN






EDMONSTON








474
MEASLES
PDKILTYIAADHC
1357
AAF02706.1



MORBILLIVIRUS








475
MEASLES VIRUS
PERIKLDYDQYCADV
1358
P08362.1



STRAIN






EDMONSTON








476
RUBELLA VIRUS
PERPRLRL
1359
CAJ88851.1





477
RUBELLA VIRUS
PGCATQAPVPVRLAG
1360
BAA28178.1





478
RUBELLA VIRUS
PGCATQAPVPVRLAGVRFESKIVDGGCFA
1361
CAJ88851.1



VACCINE STRAIN






RA27/3








479
RUBELLA VIRUS
PGEVWVTP
1362
CAJ88851.1





480
RUBELLA VIRUS
PGEVWVTPVIGSQAR
1363
BAA28178.1





481

CORYNEBACTERIUM

PGKLDVNKSKTHISVN
1364
CAE11230.1




DIPHTHERIAE









482
MEASLES VIRUS
PGLGAPVFHMTNYLE
1365
P08362.1



STRAIN






EDMONSTON








483
RUBELLA VIRUS
PGPGEVWV
1366
CAJ88851.1





484
RUBELLA VIRUS
PHKTVRVKFHTETRT
1367
BAA28178.1





485
MEASLES
PIELQVECFTWDQKL
1368
AAR89413.1



MORBILLIVIRUS








486
MEASLES VIRUS
PISLERLDVG
1369
P26031.1



STRAIN






EDMONSTON-B








487
MEASLES VIRUS
PISLERLDVGTNLGN
1370
P69353.1



STRAIN






EDMONSTON








488

BORDETELLA

PKALFTQQGGAYGRC
1371
P04979.1




PERTUSSIS









489
MEASLES VIRUS
PKYVATQGYLISNFD
1372
P69353.1



STRAIN






EDMONSTON








490
MEASLES VIRUS
PLDIDTASESSQD
1373
P04851.1



STRAIN






EDMONSTON








491
RUBELLA VIRUS
PLGLKFKTVRPVALP
1374
BAA28178.1





492
MEASLES VIRUS
PLITHGSGMDLYKSN
1375
P08362.1



STRAIN






EDMONSTON








493
MEASLES
PLKDNRIPSYGVLSV
1376
P06830.1



MORBILLIVIRUS








494
RUBELLA VIRUS
PLLRTAPG
1377
CAJ88851.1





495
MEASLES VIRUS
PLLSCKPWQESRK
1378
P04851.1



STRAIN






EDMONSTON








496

BORDETELLA

PPATVYRYDSRPPE
1379
P04977.1




PERTUSSIS









497
MEASLES
PPISLERLDVGT
1380
AAL29688.1



MORBILLIVIRUS








498
RUBELLA VIRUS
PPPPEERQETRSQTPAPKPS
1381
P07566.1





499

BORDETELLA

PQEQITQHGSPYGRC
1382
AAA83981.1




PERTUSSIS









500

BORDETELLA

PQEQITQHGSPYGRCANK
1383
AAA83981.1




PERTUSSIS









501

BORDETELLA

PQPGPQPPQPPQPQPEAPAPQPPAG
1384
P14283.3




PERTUSSIS









502
MEASLES VIRUS
PRLGGKEDRRVKQ
1385
P04851.1



STRAIN






EDMONSTON








503
RUBELLA VIRUS
PRLRLVDA
1386
CAJ88851.1





504
RUBELLA VIRUS
PRNVRVTGCYQCGTP
1387
BAA28178.1





505
MEASLES VIRUS
PSRASDARAAHLP
1388
P04851.1



STRAIN






EDMONSTON








506
MEASLES VIRUS
PTGQIHWGNLSKIGV
1389
P69353.1



STRAIN






EDMONSTON








507
MEASLES VIRUS
PTGTPLDIDTASE
1390
P04851.1



STRAIN






EDMONSTON








508
MEASLES VIRUS
PTLSEIKGVIVHRLE
1391
P69353.1



STRAIN






EDMONSTON








509
MEASLES
PTTIRGQFSNMSLSL
1392
P06830.1



MORBILLIVIRUS








510
MEASLES
PTTRTDDKLR
1393
AAR89413.1



MORBILLIVIRUS








511
MEASLES
PTTRTDDKLRMETCFQQACKG
1394
AAR89413.1



MORBILLIVIRUS








512
RUBELLA VIRUS
PVALPRTLAPPRNVR
1395
BAA28178.1





513

CORYNEBACTERIUM

PVFAGANYAAWAVNVAQVI
1396
AAV70486.1




DIPHTHERIAE









514
RUBELLA VIRUS
PVIGSQAR
1397
CAJ88851.1





515
MEASLES
PVVEVNGVTIQVGSR
1398
AAL29688.1



MORBILLIVIRUS








516
RUBELLA VIRUS
PWELVVLTARPEDGWTCRGV
1399
P07566.1



STRAIN THERIEN








517
MEASLES VIRUS
PWQESRKNKAQTR
1400
P04851.1



STRAIN






EDMONSTON








518
MEASLES VIRUS
PYMVNLENSIQNKFS
1401
P04851.1



STRAIN






EDMONSTON








519
MEASLES VIRUS
PYMVNLENSIQNKFSAGSYP
1402
P04851.1



STRAIN






EDMONSTON








520
MEASLES VIRUS
PYQGSGKGVS
1403
P08362.1



STRAIN






EDMONSTON








521
RUBELLA VIRUS
PYQVSCGGESDRASA
1404
BAA28178.1





522
MEASLES
QACKGKIQALCEN
1405
P08362.1



MORBILLIVIRUS








523
MEASLES VIRUS
QAGQEMILAVQGVQD
1406
P69353.1



STRAIN






EDMONSTON








524
MEASLES
QALCENPECVPLKDN
1407
P06830.1



MORBILLIVIRUS








525

BORDETELLA

QALGALK
1408
ACI16088.1




PERTUSSIS









526
RUBELLA VIRUS
QAPVPVRLAGVRFES
1409
BAA28178.1





527
RUBELLA VIRUS
QCGTPALVEGLAPGG
1410
BAA28178.1





528
MEASLES VIRUS
QDPDKILTYIAADHC
1411
P69353.1



STRAIN






EDMONSTON








529
MEASLES VIRUS
QECLRGSTKSCARTL
1412
P69353.1



STRAIN






EDMONSTON








530

BORDETELLA

QEQITQHGSPYGRC
1413
AAA83981.1




PERTUSSIS









531
MEASLES VIRUS
QESRKNKAQTRTP
1414
P04851.1



STRAIN






EDMONSTON








532
MEASLES VIRUS
QGDQSENELPRLGGK
1415
P04851.1



STRAIN






EDMONSTON








533
MEASLES VIRUS
QGDQSENELPRLGGKEDRRV
1416
P04851.1



STRAIN






EDMONSTON








534

CORYNEBACTERIUM

QGESGHDIKITAENTPLPIA
1417
AAV70486.1




DIPHTHERIAE









535
MEASLES
QGSGKGVSFQLVKLG
1418
P06830.1



MORBILLIVIRUS








536
MEASLES VIRUS
QGVQDYINNELIPSM
1419
P69353.1



STRAIN






EDMONSTON








537
RUBELLA VIRUS
QLEVQVPPDPGDLVE
1420
BAA28178.1





538
MEASLES
QLPEATFMV
1421
ABK40528.1



MORBILLIVIRUS








539
RUBELLA VIRUS
QLPFLGHDGHHGGTLRVGQHYRNAS
1422
NP_740663.1





540
MEASLES VIRUS
QLSMYRVFEV
1423
P08362.1



STRAIN






EDMONSTON








541

BORDETELLA

QLSNIT
1424
ACI16083.1




PERTUSSIS









542
MEASLES VIRUS
QNIRPVQSVASSRRH
1425
P69353.1



STRAIN






EDMONSTON








543
MEASLES VIRUS
QNKFSAGSYPLLWSY
1426
P04851.1



STRAIN






EDMONSTON








544
MEASLES VIRUS
QNKFSAGSYPLLWSYAMGVG
1427
P04851.1



STRAIN






EDMONSTON








545
MEASLES VIRUS
QNKFSAGSYPLLWSYAMGVGVELEN
1428
P04851.1



STRAIN






EDMONSTON








546
MEASLES VIRUS
QQACKGKIQALCENP
1429
P08362.1



STRAIN






EDMONSTON








547
MEASLES VIRUS
QQRRVVGEFRLERKW
1430
P04851.1



STRAIN






EDMONSTON








548
MEASLES VIRUS
QQRRVVGEFRLERKWLDVVR
1431
P04851.1



STRAIN






EDMONSTON








549

BORDETELLA

QQTRANPNPYTSRRSVAS
1432
P04977.1




PERTUSSIS









550
RUBELLA VIRUS
QRHSPDCSRLVGATP
1433
BAA28178.1





551
MEASLES VIRUS
QSGLTFASRGTNMED
1434
P04851.1



STRAIN






EDMONSTON








552
MEASLES VIRUS
QSGLTFASRGTNMEDEADQY
1435
P04851.1



STRAIN






EDMONSTON








553

CORYNEBACTERIUM

QSIALSSLMVAQAIPLVGEL
1436
AAV70486.1




DIPHTHERIAE









554
MEASLES VIRUS
QSRFGWFENKEISDI
1437
P04851.1



STRAIN






EDMONSTON








555
MEASLES VIRUS
QSRFGWFENKEISDIEVQDP
1438
P04851.1



STRAIN






EDMONSTON








556
MEASLES VIRUS
QSRGEAR
1439
P04851.1



STRAIN






EDMONSTON








557
MEASLES VIRUS
QSRGEARESYRETGPSRA
1440
P04851.1



STRAIN






EDMONSTON








558
RUBELLA VIRUS
QTGRGGSAPRPELGPPTN
1441
P07566.1



STRAIN THERIEN








559
RUBELLA VIRUS
QTPAPKPSRAPPQQPQPPRMQTGRG
1442
P07566.1



STRAIN THERIEN








560
MEASLES VIRUS
QVGSRRYPDAVYLHR
1443
P69353.1



STRAIN






EDMONSTON








561
MEASLES VIRUS
QVSFLQGDQSENE
1444
P04851.1



STRAIN






EDMONSTON








562
RUBELLA VIRUS
QYHPTACEVEPAFGH
1445
BAA28178.1





563
MEASLES VIRUS
QYVLATYDTSRVEHA
1446
P08362.1



STRAIN






EDMONSTON








564

BORDETELLA

RANPNPYTSRRSV
1447
ACI04548.1




PERTUSSIS









565
MEASLES VIRUS
RASDARAAHLPTG
1448
P04851.1



STRAIN






EDMONSTON








566
MEASLES VIRUS
RASLETTNQAIEAIR
1449
P69353.1



STRAIN






EDMONSTON








567
RUBELLA VIRUS
RCGRLICGLSTTAQYPPTRF
1450
P07566.1



STRAIN THERIEN








568

BORDETELLA

RDGQSVIGACASPYEGRYR
1451
P04979.1




PERTUSSIS









569
MEASLES VIRUS
RESYRETGPSRAS
1452
P04851.1



STRAIN






EDMONSTON








570
MEASLES VIRUS
RETGPSRASDARA
1453
P04851.1



STRAIN






EDMONSTON








571
MUMPS
RFAKYQQQGRLEAR
1454
P21186.1



RUBULAVIRUS








572
RUBELLA VIRUS
RFGAPQAFLAGLLLATVAVGTARA
1455
P07566.1



STRAIN THERIEN








573
MEASLES VIRUS
RFMVALILDIKRTPG
1456
P04851.1



STRAIN






EDMONSTON








574
MEASLES VIRUS
RFMVALILDIKRTPGNKPRI
1457
P04851.1



STRAIN






EDMONSTON








575
MEASLES VIRUS
RGEARESYRETGP
1458
P04851.1



STRAIN






EDMONSTON








576
RUBELLA VIRUS
RGTTPPAYG
1459
CAA28880.1





577
RUBELLA VIRUS
RIETRSARH
1460
ABD64214.1



STRAIN M33








578

BORDETELLA

RILAGALATYQ
1461
P04977.1




PERTUSSIS









579

BORDETELLA

RIPPENIRRVT
1462
ACI04548.1




PERTUSSIS









580

BORDETELLA

RISNLND
1463
ACI16083.1




PERTUSSIS









581

BORDETELLA

RLANLNG
1464
ACI16088.1




PERTUSSIS









582
MEASLES VIRUS
RLDVGTNLGNAIAKL
1465
P69353.1



STRAIN






EDMONSTON








583
MEASLES VIRUS
RLERKWLDV
1466
P04851.1



STRAIN






EDMONSTON








584
MEASLES VIRUS
RLGGKEDRRVKQSRG
1467
P04851.1



STRAIN






EDMONSTON








585
MEASLES VIRUS
RLGGKEDRRVKQSRGEARES
1468
P04851.1



STRAIN






EDMONSTON








586
MEASLES VIRUS
RLLDRLVRL
1469
ABI54110.1



STRAIN






EDMONSTON








587
RUBELLA VIRUS
RLRLVDAD
1470
CAJ88851.1





588
RUBELLA VIRUS
RLRLVDADDPLLR
1471
BAA19893.1





589
RUBELLA VIRUS
RLRLVDADDPLLRTAPGPGEVWVTPVIGSQ
1472
BAA19893.1




A







590
RUBELLA VIRUS
RLRLVQDADDPLLRIAPGPGEVWVTPVIGS
1473
SRC265968




QA







591
MEASLES VIRUS
RLSDNGYYTV
1474
ABK40528.1



STRAIN






EDMONSTON








592
RUBELLA VIRUS
RLVDADDP
1475
CAJ88851.1





593
RUBELLA VIRUS
RLVDADDPLLRTAPG
1476
BAA28178.1





594
RUBELLA VIRUS
RLVGATPE
1477
CAJ88851.1





595
RUBELLA VIRUS
RMQTGRGGSAPRPELGPPTNPFQAAVA
1478
ABD64216.1





596
MEASLES
RMSKGVFKV
1479
ABY21184.1



MORBILLIVIRUS








597
MEASLES
RNPGLGAPVFHMTNY
1480
P06830.1



MORBILLIVIRUS








598
RUBELLA VIRUS
RPRLRLVD
1481
CAJ88851.1





599

BORDETELLA

RQAESSEAMAAWSERAGEA
1482
P04977.1




PERTUSSIS









600
RUBELLA VIRUS
RQTWAEWAAAHWWQL
1483
BAA28178.1





601
MEASLES VIRUS
RRVKQSRGEARES
1484
P04851.1



STRAIN






EDMONSTON








602
MEASLES
RRYPDAVYL
1485
ACA09725.1



MORBILLIVIRUS








603
MEASLES VIRUS
RRYPDAVYLHRIDLG
1486
P69353.1



STRAIN






EDMONSTON








604
MEASLES VIRUS
RSAGKVSSTLASELG
1487
P04851.1



STRAIN






EDMONSTON








605
MEASLES VIRUS
RSAGKVSSTLASELGITAED
1488
P04851.1



STRAIN






EDMONSTON








606
MEASLES VIRUS
RSELSQLS
1489
P08362.1



STRAIN






EDMONSTON








607
MEASLES VIRUS
RSELSQLSMYRVFEV
1490
P08362.1



STRAIN






EDMONSTON








608
RUBELLA VIRUS
RSQTPAPKPSRAPPQQPQPPRMQT
1491
ABD64214.1





609
RUBELLA VIRUS
RTAPGPGE
1492
CAJ88851.1





610
RUBELLA VIRUS
RTAPGPGEVWVTPVI
1493
BAA28178.1





611
MEASLES
RTDDKLRMETCFQQA
1494
P06830.1



MORBILLIVIRUS








612
RUBELLA VIRUS
RTLAPPRNVRVTGCY
1495
BAA28178.1





613
MEASLES VIRUS
RTVLEPIRDALNAMT
1496
P69353.1



STRAIN






EDMONSTON








614
MEASLES VIRUS
RVEHAVVYYVYSPSR
1497
P08362.1



STRAIN






EDMONSTON








615
MEASLES VIRUS
RVFEVGVIRNPGLGA
1498
P08362.1



STRAIN






EDMONSTON








616

BORDETELLA

RVHVSKEEQYYDYEDATFE
1499
P04978.2




PERTUSSIS









617
RUBELLA VIRUS
RVIDPAAQ
1500
BAA28178.1





618
RUBELLA VIRUS
RVKFHTETRTVWQLS
1501
BAA28178.1





619

BORDETELLA

RVYHNGITGET
1502
ACI04548.1




PERTUSSIS









620

BORDETELLA

RYDSRPPEDVF
1503
ACI04548.1




PERTUSSIS









621
MEASLES VIRUS
RYPDAVYLHRIDLGP
1504
P69353.1



STRAIN






EDMONSTON








622
MEASLES VIRUS
SAEISIQALSYALGG
1505
P69353.1



STRAIN






EDMONSTON








623
MEASLES VIRUS
SAEPLLSCKPWQESR
1506
P04851.1



STRAIN






EDMONSTON








624
MEASLES VIRUS
SAEPLLSCKPWQESRKNKAQ
1507
P04851.1



STRAIN






EDMONSTON








625
MEASLES
SAGKVSSTLASELG
1508
P04851.1



MORBILLIVIRUS








626
MEASLES VIRUS
SAGKVSSTLASELGITAEDARLVS
1509
P04851.1



STRAIN






EDMONSTON








627
MEASLES VIRUS
SCTVTREDGT
1510
P08362.1



STRAIN






EDMONSTON








628
RUBELLA VIRUS
SDAACWGFPTDTVMS
1511
BAA28178.1





629
MEASLES VIRUS
SDARAAHLPTGTP
1512
P04851.1



STRAIN






EDMONSTON








630
RUBELLA VIRUS
SDWHQGTHVCHTKHMDFWCVEHD
1513
P07566.1



STRAIN THERIEN








631
MEASLES VIRUS
SELRRWIKYTQQRRV
1514
P04851.1



STRAIN






EDMONSTON








632
MEASLES VIRUS
SELRRWIKYTQQRRVVGEFR
1515
P04851.1



STRAIN






EDMONSTON








633
MEASLES VIRUS
SELSQL
1516
P08362.1



STRAIN






EDMONSTON








634
MEASLES VIRUS
SELSQLS
1517
P08362.1



STRAIN






EDMONSTON








635

BORDETELLA

SEYLAHRRIPPENIRRVTRV
1518
CAD44970.1




PERTUSSIS









636
MEASLES VIRUS
SFLQGDQSENELP
1519
P04851.1



STRAIN






EDMONSTON








637
MEASLES VIRUS
SGKGVSFQLVKLGVW
1520
P08362.1



STRAIN






EDMONSTON








638
MEASLES VIRUS
SHRGVIADNQAKWAV
1521
P08362.1



STRAIN






EDMONSTON








639
MEASLES VIRUS
SIEHQVKDVLTPLFK
1522
P08362.1



STRAIN






EDMONSTON








640
MEASLES VIRUS
SKIGVVGIGSASYKV
1523
P69353.1



STRAIN






EDMONSTON








641
MEASLES
SKRSELSQLSMYRVF
1524
P06830.1



MORBILLIVIRUS








642
MEASLES
SLFVESPGQLIQRITDDPDVS
1525
ABI54110.1



MORBILLIVIRUS








643
MEASLES VIRUS
SLSTNLDVTNSIEHQ
1526
P08362.1



STRAIN






EDMONSTON








644
MEASLES VIRUS
SLSTNLDVTNSIEHQVKDVLTPLFK
1527
P08362.1



STRAIN






EDMONSTON








645
MEASLES VIRUS
SLWGSGLLML
1528
BAE98296.1



STRAIN






EDMONSTON








646
MEASLES VIRUS
SMKGLSSTSIVYILI
1529
P69353.1



STRAIN






EDMONSTON








647
MEASLES
SMYRVFEVGV
1530
P08362.1



MORBILLIVIRUS








648
MEASLES
SNDLSNCMVALGELK
1531
P06830.1



MORBILLIVIRUS








649
MEASLES VIRUS
SPGQLIQR
1532
P10050.1



STRAIN HALLE








650
MEASLES VIRUS
SQDPQDSRRSAEP
1533
P04851.1



STRAIN






EDMONSTON








651
MEASLES
SRIVINREHLMIDRP
1534
P06830.1



MORBILLIVIRUS








652
MEASLES VIRUS
SRKNKAQTRTPLQ
1535
P04851.1



STRAIN






EDMONSTON








653
RUBELLA VIRUS
SRLVGATP
1536
CAJ88851.1





654
RUBELLA VIRUS
SRLVGATPERPRLRLVDADDPLLR
1537
CAJ88851.1





655
MEASLES VIRUS
SRPGLKPDLTGTSKS
1538
P69353.1



STRAIN






EDMONSTON








656

BORDETELLA

SRRSVASIVGTLVRM
1539
CAD44970.1




PERTUSSIS









657
RUBELLA VIRUS
SRWGLGSPNCHGPDW
1540
BAA28178.1





658

BORDETELLA

SSATK
1541
ACI16088.1




PERTUSSIS









659
RUBELLA VIRUS
SSGGYAQLASYFNPG
1542
BAA28178.1





660

BORDETELLA

SSLGNGV
1543
ACI16083.1




PERTUSSIS









661
MEASLES
SSNLVILPGQDLQYV
1544
P06830.1



MORBILLIVIRUS








662
MEASLES VIRUS
SSQDPQDSRRSAEPL
1545
P04851.1



STRAIN






EDMONSTON








663
MEASLES VIRUS
SSQDPQDSRRSAEPLLSCKP
1546
P04851.1



STRAIN






EDMONSTON








664
MEASLES VIRUS
SSRRHKRFAGVVLAG
1547
P69353.1



STRAIN






EDMONSTON








665
MEASLES VIRUS
SSTSIVYILIAVCLG
1548
P69353.1



STRAIN






EDMONSTON








666

BORDETELLA

STPGIVI
1549
AAA83981.1




PERTUSSIS









667

BORDETELLA

STPGIVIPPQEQITQHGSPYGRC
1550
AAA83981.1




PERTUSSIS









668

BORDETELLA

STSSSRRYTEVY
1551
P04977.1




PERTUSSIS









669
MEASLES VIRUS
SYFIVLSIAYPTLSE
1552
P69353.1



STRAIN






EDMONSTON








670
MEASLES VIRUS
SYRETGPSRASDA
1553
P04851.1



STRAIN






EDMONSTON








671

BORDETELLA

SYVK
1554
ACI16083.1




PERTUSSIS









672
RUBELLA VIRUS
SYVQHPHKTVRVKFH
1555
BAA28178.1





673
RUBELLA VIRUS
TAPGPGEV
1556
CAJ88851.1





674

BORDETELLA

TATRLLSSTNSRLC
1557
AAA83981.1




PERTUSSIS









675
MEASLES
TDDPVIDRLYLSSHR
1558
P06830.1



MORBILLIVIRUS








676
MEASLES VIRUS
TEILSLFGPSLRDPI
1559
P69353.1



STRAIN






EDMONSTON








677
RUBELLA VIRUS
TETRTVWQLSVAGVS
1560
BAA28178.1





678

BORDETELLA

TEVYLEHRMQEAVE
1561
P04977.1




PERTUSSIS









679
MEASLES VIRUS
TFMPEGTVCSQNALY
1562
P69353.1



STRAIN






EDMONSTON








680
RUBELLA VIRUS
TGACICEIPTDVSCE
1563
BAA28178.1





681

BORDETELLA

TGDLRAY
1564
ACI16083.1




PERTUSSIS









682
MEASLES
TGMQSWVPLSTDDPV
1565
P06830.1



MORBILLIVIRUS








683
RUBELLA VIRUS
TGNQQSRWGLGSPNC
1566
BAA28178.1





684
MEASLES VIRUS
TGPSRASDARAAH
1567
P04851.1



STRAIN






EDMONSTON








685
MEASLES
TGTIINQDPDKILTY
1568
AAF02706.1



MORBILLIVIRUS








686
RUBELLA VIRUS
TGVVYGTHTTAVSET
1569
BAA28178.1





687
MEASLES VIRUS
TIRGQFSNMSLSLLD
1570
P08362.1



STRAIN






EDMONSTON








688
RUBELLA VIRUS
TLGATCALPLAGLLA
1571
BAA28178.1





689
MEASLES
TLLNNCTRV
1572
P26031.1



MORBILLIVIRUS








690
MEASLES VIRUS
TLNVPPPPDPGR
1573
P03422.1



STRAIN






EDMONSTON-B








691
MEASLES VIRUS
TLNVPPPPDPGRASTSGTPIKK
1574
P03422.1



STRAIN






EDMONSTON-B








692
MEASLES
TMTSQGMYGGTYPVE
1575
P06830.1



MORBILLIVIRUS








693
MEASLES VIRUS
TNLGNAIAKLEDAKE
1576
P69353.1



STRAIN






EDMONSTON








694
MEASLES VIRUS
TNMEDEADQYFSHDD
1577
P04851.1



STRAIN






EDMONSTON








695
MEASLES VIRUS
TNMEDEADQYFSHDDPISSD
1578
P04851.1



STRAIN






EDMONSTON








696
RUBELLA VIRUS
TNPFQAAVARGLRPP
1579
CAA28880.1



STRAIN M33








697
MEASLES
TNSIEHQVKDVLTPL
1580
P06830.1



MORBILLIVIRUS








698
MEASLES VIRUS
TNYLEQPVSNDLSNC
1581
P08362.1



STRAIN






EDMONSTON








699
MEASLES
TNYLEQPVSNDLSNCMVALGELKLAAL
1582
AAR89413.1



MORBILLIVIRUS








700
RUBELLA VIRUS
TPERPRLR
1583
CAJ88851.1





701
RUBELLA VIRUS
TPERPRLRLVDADDPLLRTA
1584
P07566.1



STRAIN THERIEN








702
MEASLES VIRUS
TPGNKPRIA
1585
P10050.1



STRAIN HALLE








703
MEASLES VIRUS
TPLDIDTASESSQDP
1586
P04851.1



STRAIN






EDMONSTON








704
MEASLES VIRUS
TPLDIDTASESSQDPQDSRR
1587
P04851.1



STRAIN






EDMONSTON








705
MEASLES VIRUS
TPLFKIIGDEVGLRT
1588
P08362.1



STRAIN






EDMONSTON








706
MEASLES VIRUS
TPLQCTM
1589
P04851.1



STRAIN






EDMONSTON








707
RUBELLA VIRUS
TPVIGSQA
1590
CAJ88851.1





708
RUBELLA VIRUS
TPVIGSQARK
1591
BAA19893.1





709
MEASLES VIRUS
TQGYLISNFDESSCT
1592
P69353.1



STRAIN






EDMONSTON








710

BORDETELLA

TRANPNPYTSRRSVASIVGTLVRM
1593
P04977.1




PERTUSSIS









711
RUBELLA VIRUS
TRFGCAMRWGLPP
1594
NP_740663.1





712

BORDETELLA

TRNTGQPATDHYYSNV
1595
AAA83981.1




PERTUSSIS









713
MEASLES VIRUS
TRTPLQCTMTEIF
1596
P04851.1



STRAIN






EDMONSTON








714
RUBELLA VIRUS
TRWHRLLRMPVR
1597
ABD64216.1





715
MEASLES VIRUS
TSGSGGAIRGIKHII
1598
P04851.1



STRAIN






EDMONSTON








716
MEASLES VIRUS
TSGSGGAIRGIKHIIIVPIP
1599
P04851.1



STRAIN






EDMONSTON








717
MEASLES VIRUS
TSQGMYGGTYLVEKP
1600
P08362.1



STRAIN






EDMONSTON








718
MEASLES
TSRVEHAVVYYVYSP
1601
P06830.1



MORBILLIVIRUS








719

BORDETELLA

TSSSRRYTEVYL
1602
ACI04548.1




PERTUSSIS









720

BORDETELLA

TSYVG
1603
ACI16088.1




PERTUSSIS









721
MEASLES VIRUS
TTEDKISRAVGPRQA
1604
P04851.1



STRAIN






EDMONSTON








722
MEASLES VIRUS
TTEDKISRAVGPRQAQVSFL
1605
P04851.1



STRAIN






EDMONSTON








723
RUBELLA VIRUS
TTERIETRSARHP
1606
ABD64214.1



STRAIN M33








724
MEASLES VIRUS
TTNQAIEAIRQAGQE
1607
P69353.1



STRAIN






EDMONSTON








725
RUBELLA VIRUS
TTSDPWHPPGPLGLK
1608
BAA28178.1





726
MEASLES VIRUS
TVCSQNALYPMSPLL
1609
P69353.1



STRAIN






EDMONSTON








727
RUBELLA VIRUS
TVNGEDVGAVPPGKF
1610
BAA28178.1





728
MEASLES
TYPVEKPNLSSKRSE
1611
P06830.1



MORBILLIVIRUS








729
RUBELLA VIRUS
VAGVSCNVTTEHPFC
1612
BAA28178.1





730

BORDETELLA

VAPGIVIPPKALFTQQGGAYGRC
1613
P04979.1




PERTUSSIS









731
RUBELLA VIRUS
VCHTKHMDFWCVEHDRPPPATPTPL
1614
NP_740663.1





732
RUBELLA VIRUS
VCQRHSPDCSRLVGATPER
1615
BAA19893.1





733
RUBELLA VIRUS
VDADDPLL
1616
CAJ88851.1





734
RUBELLA VIRUS
VDADDPLLRTAPGPGEVWVT
1617
BAA19893.1





735

CORYNEBACTERIUM

VDIGFAAYNFVESIINLFQV
1618
AAV70486.1




DIPHTHERIAE









736
MEASLES VIRUS
VEIAEYRRLLRTVLE
1619
P69353.1



STRAIN






EDMONSTON








737
MEASLES VIRUS
VELENSMGGLNFGRS
1620
P04851.1



STRAIN






EDMONSTON








738
MEASLES VIRUS
VELENSMGGLNFGRSYFDPA
1621
P04851.1



STRAIN






EDMONSTON








739
MEASLES
VELKIKIASGFGPLI
1622
P06830.1



MORBILLIVIRUS








740
RUBELLA VIRUS
VEMDEWIHAHTTSD
1623
SRC265968





741
RUBELLA VIRUS
VEMPEWIHAHTTSDP
1624
BAA28178.1





742

CORYNEBACTERIUM

VERRLVKVL
1625
P33120.2




DIPHTHERIAE









743
MEASLES
VESPGQLI
1626
ABI54110.1



MORBILLIVIRUS








744
MEASLES VIRUS
VESPGQLIQRITDDP
1627
P04851.1



STRAIN






EDMONSTON








745
MEASLES VIRUS
VESPGQLIQRITDDPDVSIR
1628
P04851.1



STRAIN






EDMONSTON








746
RUBELLA VIRUS
VFALASYVQHPHKTV
1629
BAA28178.1





747
RUBELLA VIRUS
VGATPERP
1630
CAJ88851.1





748
RUBELLA VIRUS
VGATPERPRL
1631
BAA19893.1





749
RUBELLA VIRUS
VGATPERPRLRLVDA
1632
BAA28178.1





750
MEASLES VIRUS
VGIGSASYKVMTRSS
1633
P69353.1



STRAIN






EDMONSTON








751
MEASLES VIRUS
VGLRTPQRFTDLVKF
1634
P08362.1



STRAIN






EDMONSTON








752

CORYNEBACTERIUM

VHHNTEEIVAQSIALSSLMV
1635
AAV70486.1




DIPHTHERIAE









753
MEASLES VIRUS
VHRLEGVSYNIGSQE
1636
P69353.1



STRAIN






EDMONSTON








754
RUBELLA VIRUS
VIGSQARK
1637
CAJ88851.1





755

BORDETELLA

VITGSI
1638
ACI16088.1




PERTUSSIS









756

BORDETELLA

VITGTI
1639
ACI16083.1




PERTUSSIS









757
MEASLES VIRUS
VKQSRGEA
1640
P04851.1



STRAIN






EDMONSTON








758
MEASLES VIRUS
VLFVMFLSLI
1641
P08362.1



STRAIN






EDMONSTON








759
MEASLES
VLFVMFLSLIGLLAI
1642
P06830.1



MORBILLIVIRUS








760
MEASLES
VLTPLFKIIGDEVGL
1643
P06830.1



MORBILLIVIRUS








761
MEASLES VIRUS
VMTRSSHQSLVIKLM
1644
P69353.1



STRAIN






EDMONSTON








762
RUBELLA VIRUS
VPAAPCARIWNGTQR
1645
BAA28178.1





763
RUBELLA VIRUS
VPPDPGDLVEYIMNY
1646
BAA28178.1





764
MEASLES VIRUS
VQSVASSRRHKRFAG
1647
P69353.1



STRAIN






EDMONSTON








765

BORDETELLA

VQTGGTSRTVTMRYLAS
1648
ACI16083.1




PERTUSSIS









766

BORDETELLA

VQVRI
1649
ACI16083.1




PERTUSSIS









767
RUBELLA VIRUS
VRAYNQPAGDV
1650
NP_740662.1





768
RUBELLA VIRUS
VRFESKIVDGGCFAP
1651
BAA28178.1





769
RUBELLA VIRUS
VRLAGVRFESKIVDG
1652
BAA28178.1





770
MEASLES VIRUS
VSGSFGNRFILSQGN
1653
P69353.1



STRAIN






EDMONSTON








771

BORDETELLA

VSKEEQYYDYEDAT
1654
AAA83981.1




PERTUSSIS









772
MEASLES VIRUS
VSKGNCSGPTTIRGQ
1655
P08362.1



STRAIN






EDMONSTON








773
RUBELLA VIRUS
VTAALLNTPPPYQVS
1656
BAA28178.1





774
RUBELLA VIRUS
VTGCYQCGTPALVEG
1657
BAA28178.1





775
RUBELLA VIRUS
VTPVIGSQ
1658
CAJ88851.1





776
RUBELLA VIRUS
VTTEHPFCNTPHGQLEVQVPPD
1659
P07566.1



STRAIN THERIEN








777
MEASLES VIRUS
VVLAGAALGVATAAQ
1660
P69353.1



STRAIN






EDMONSTON








778
RUBELLA VIRUS
VWQLSVAGVSCNVTT
1661
BAA28178.1





779
RUBELLA VIRUS
VWVTPVIG
1662
CAJ88851.1





780
RUBELLA VIRUS
VWVTPVIGSQAR
1663
BAA19893.1





781
MEASLES VIRUS
VYILIAVCLGGLIGI
1664
P69353.1



STRAIN






EDMONSTON








782
MEASLES VIRUS
VYLHRIDLGPPISLE
1665
P69353.1



STRAIN






EDMONSTON








783

BORDETELLA

VYRYDSRP
1666
P04977.1




PERTUSSIS









784

BORDETELLA

VYRYDSRPPEDV
1667
P04977.1




PERTUSSIS









785
MEASLES
VYWLTIPPMKNLALG
1668
P06830.1



MORBILLIVIRUS








786
RUBELLA VIRUS
WDLEATGACICEIPT
1669
BAA28178.1





787
MEASLES
WDQKLWCRHFCVLAD
1670
AAR89413.1



MORBILLIVIRUS








788
RUBELLA VIRUS
WGFPTDTVMSVFALA
1671
BAA28178.1





789
RUBELLA VIRUS
WHPPGPLGLKFKTVR
1672
BAA28178.1





790
RUBELLA VIRUS
WIHAHTTSDPWHPPG
1673
BAA28178.1





791
MEASLES VIRUS
WLTIPPMKNLALGVI
1674
P08362.1



STRAIN






EDMONSTON








792
MEASLES VIRUS
WQESRKNKAQTRTPLQCTMT
1675
P04851.1



STRAIN






EDMONSTON








793
RUBELLA VIRUS
WVCIFMVCRRACR
1676
SRC265968





794
RUBELLA VIRUS
WVTPVIGS
1677
CAJ88851.1





795
MEASLES VIRUS
WYTTVPKYVATQGYL
1678
P69353.1



STRAIN






EDMONSTON








796
MEASLES VIRUS
YALGGDINKVLEKLG
1679
P69353.1



STRAIN






EDMONSTON








797
MEASLES VIRUS
YAMGVGVELE
1680
P04851.1



STRAIN






EDMONSTON








798
MEASLES
YAMGVGVELEN
1681
ABI54110.1



MORBILLIVIRUS








799
MEASLES
YCADVAAEELMNALV
1682
AAR89413.1



MORBILLIVIRUS








800
MEASLES
YDFRDLTWCINPPER
1683
P06830.1



MORBILLIVIRUS








801

BORDETELLA

YFEPGPT
1684
ACI16083.1




PERTUSSIS









802
RUBELLA VIRUS
YFNPGGSYYKQYHPT
1685
BAA28178.1





803
MEASLES VIRUS
YFRLGQEMVRRSAGK
1686
P04851.1



STRAIN






EDMONSTON








804
MEASLES VIRUS
YFRLGQEMVRRSAGKVSSTL
1687
P04851.1



STRAIN






EDMONSTON








805
MEASLES
YFYPFRLPIKGVPIE
1688
P06830.1



MORBILLIVIRUS








806

BORDETELLA

YGDNAGRILAGALAT
1689
P04977.1




PERTUSSIS









807
RUBELLA VIRUS
YGHATVEMPEWIHAH
1690
BAA28178.1





808
RUBELLA VIRUS
YIMNYTGNQQSRWGL
1691
BAA28178.1





809
MEASLES VIRUS
YINNELIPSMNQLSC
1692
P69353.1



STRAIN






EDMONSTON








810
RUBELLA VIRUS
YLCTAPGCATQAPVP
1693
BAA28178.1





811
MEASLES
YLFTVPIKEAGEDCH
1694
P06830.1



MORBILLIVIRUS








812
MEASLES VIRUS
YLHDPEFNL
1695
ABK40531.1



STRAIN






EDMONSTON








813
MEASLES VIRUS
YLNMSRLFV
1696
ABK40531.1



STRAIN






EDMONSTON








814
MEASLES VIRUS
YPALGLHEF
1697
P10050.1



STRAIN HALLE








815
MEASLES
YPALGLHEFA
1698
ABI54110.1



MORBILLIVIRUS








816
MEASLES VIRUS
YPALGLHEFAGELST
1699
P04851.1



STRAIN






EDMONSTON








817
MEASLES VIRUS
YPALGLHEFAGELSTLESLM
1700
P04851.1



STRAIN






EDMONSTON








818
MEASLES VIRUS
YPFRLPIKGVPIELQ
1701
P08362.1



STRAIN






EDMONSTON








819
MEASLES VIRUS
YPLLWSYAM
1702
P10050.1



STRAIN HALLE








820
MUMPS
YQQQGRL
1703
P21186.1



RUBULAVIRUS








821

BORDETELLA

YQSEYLAHRR
1704
P04977.1




PERTUSSIS









822
MEASLES VIRUS
YRETGPSRASDARAA
1705
P04851.1



STRAIN






EDMONSTON








823
MEASLES VIRUS
YRETGPSRASDARAAHLPTG
1706
P04851.1



STRAIN






EDMONSTON








824
RUBELLA VIRUS
YRNASDVLPGHWLQGGWGCYNLSDW
1707
NP_740663.1





825
MEASLES VIRUS
YRRLLRTVLEPIRDA
1708
P69353.1



STRAIN






EDMONSTON








826

BORDETELLA

YRYDSRPP
1709
P04977.1




PERTUSSIS









827
MEASLES VIRUS
YSGGDLLGILESRGI
1710
P69353.1



STRAIN






EDMONSTON








828

BORDETELLA

YSKVTATBLLASTNSRLCAVFVRDG
1711
SRC280066




PERTUSSIS









829
MEASLES VIRUS
YSPSRSFSYFYPFRL
1712
P08362.1



STRAIN






EDMONSTON








830
RUBELLA VIRUS
YTGNQQSRWGLGSPNCHGPDWASPV
1713
P07566.1



STRAIN THERIEN








831
MEASLES
YVLLAVLFV
1714
P08362.1



MORBILLIVIRUS








832
MEASLES
YVYSPGRSFSYFYPF
1715
P06830.1



MORBILLIVIRUS








833

BORDETELLA

YYDYEDATFQTYALTGISLCNPAASIC
1716
P04979.1




PERTUSSIS









834
MEASLES
GDLLGILESRGIKAR
1717
AAF02706.1



MORBILLIVIRUS








835
MEASLES
TVPKYVATQGYLISN
1718
AAL29688.1



MORBILLIVIRUS








836
MEASLES
KPWDSPQEI
1719
P26035.1



MORBILLIVIRUS








837
MEASLES
KPWESPQEI
1720
CAA34579.1



MORBILLIVIRUS








838

BORDETELLA

ATYQSEYLAHRRIPP
1721
ACI04548.1




PERTUSSIS









839

BORDETELLA

CMARQAESSEAMAAWSERAGEAMVLVYYES
1722
ACI04548.1




PERTUSSIS

IAYSF







840

BORDETELLA

CQVGSSNSAFVSTSSSRRYTEVYL
1723
ACI04548.1




PERTUSSIS









841

BORDETELLA

DDPPATVYRYDSRPP
1724
ACI04548.1




PERTUSSIS









842

BORDETELLA

GALATYQSEYLAHRRIPP
1725
ACI04548.1




PERTUSSIS









843

BORDETELLA

MAAWSERAGEAMVLVYYESIAYSF
1726
ACI04548.1




PERTUSSIS









844

BORDETELLA

MVLVYYESIAYSF
1727
ACI04548.1




PERTUSSIS









845

BORDETELLA

PATVYRYDSRPPEDV
1728
ACI04548.1




PERTUSSIS









846

BORDETELLA

YDSRPPEDV
1729
ACI04548.1




PERTUSSIS









847

BORDETELLA

EPGITTNYDT
1730
ACI16087.1




PERTUSSIS









848

BORDETELLA

GDLRAYKMVYATNPQTQLSN
1731
ACI16083.1




PERTUSSIS









849

BORDETELLA

KNGDVEASAITTYVGFSVVYP
1732
ACI16083.1




PERTUSSIS









850

BORDETELLA

KVTNGSKSYTLRYLASYVK
1733
ACI 1608 8.1




PERTUSSIS









851

BORDETELLA

QALGALKLYFEPGITTNYDTGDLIAYKQTY
1734
ACI 1608 8.1




PERTUSSIS

NASGN







852

BORDETELLA

YATNPQTQLS
1735
ACI16083.1




PERTUSSIS









853

CORYNEBACTERIUM

DNENPLSGKAGGVVKVTYPGLTKV
1736
AAV70486.1




DIPHTHERIAE









854

CORYNEBACTERIUM

ENFSSYHGTKPGYVDSI
1737
AAV70486.1




DIPHTHERIAE









855

CORYNEBACTERIUM

KVDNAETIKKELGLSLTEP
1738
AAV70486.1




DIPHTHERIAE









856
RUBELLA VIRUS
MEDLQKALEAQSRALRAGLAA
1739
CAA28880.1



STRAIN M33








857

CORYNEBACTERIUM

QKGIQKPKSGTQGNYDDDWKGFY
1740
AAV70486.1




DIPHTHERIAE









858
RUBELLA VIRUS
RTGAWQRKDWSRAPPPPEERQESRSQTPAP
1741
CAA28880.1



STRAIN M33
KPSR







859
RUBELLA VIRUS
AAGASQSRRPRPPRHARAQHLPEMTPAVT
1742
SRC265968





860
RUBELLA VIRUS
CVTSWLWSEGEGAVFYRVDLHFINLGTP
1743
CAA28880.1





861
RUBELLA VIRUS
FRVGGTRWHRLLRMPVRGLDGDTAPLP
1744
CAA28880.1





862

CORYNEBACTERIUM

GRKIRMRCRAIDGDVTFCRPKSPVYVGN
1745
1007216A




DIPHTHERIAE









863
RUBELLA VIRUS
GTPPLDEDGRWDPALMYNPCGPEPPAHV
1746
CAA28880.1





864

CORYNEBACTERIUM

GVHANLHVAFHRSSSEKIHSNEISSDSIGV
1747
AAV70486.1




DIPHTHERIAE

LGYQKTVDHTKVNSKLSLFFEIKS







865
RUBELLA VIRUS
MASTTPITMEDLQKALEAQSRALRAGLAAG
1748
ABD64200.1





866
RUBELLA VIRUS
PELGPPTNPFQAAVARGLRPPLHDPDTEAP
1749
CAA28880.1




TEAC







867
RUBELLA VIRUS
PLPPHTTERIETRSARHPWRIRFGAP
1750
CAA28880.1





868
RUBELLA VIRUS
SRAPPPPEERQESRSQTPAPKPSRAPP
1751
CAA28880.1





869
RUBELLA VIRUS
SRAPPQQPQPPRMQTGRGGSAPRPELGP
1752
CAA28880.1





870
RUBELLA VIRUS
TPAVTPEGPAPPRTGAWQRKDWSRAPP
1753
CAA28880.1





871
RUBELLA VIRUS
VRAYNQPAGDVRGVWGKGERTYAEQDFRV
1754
CAA28880.1





872
RUBELLA VIRUS
AFGHSDAACWGFPTDTVMSV
1755
CAA28880.1





873
RUBELLA VIRUS
CARIWNGTQRACTFWAVNAYS
1756
CAA28880.1





874
RUBELLA VIRUS
EEAFTYLCTAPGCATQTPVPVR
1757
CAA28880.1





875
RUBELLA VIRUS
FAPWDLEATGACICEIPTDV
1758
CAA28880.1





876
RUBELLA VIRUS
GEDVGAFPPGKFVTAAL
1759
CAA28880.1





877
RUBELLA VIRUS
GEVWVTPVIGSQARKCGLHI
1760
CAA28880.1





878
RUBELLA VIRUS
GQLEVQVPPDPGDLVEYIMN
1761
CAA28880.1





879
RUBELLA VIRUS
GSYYKQYHPTACEVEPAFGH
1762
CAA28880.1





880
RUBELLA VIRUS
IHAHTTSDPWHPPGPLGLKF
1763
CAA28880.1





881
RUBELLA VIRUS
IMNYTGNQQSRWGLGSPNCH
1764
CAA28880.1





882
RUBELLA VIRUS
LHIRAGPYGHATVEMPEWIH
1765
CAA28880.1





883
RUBELLA VIRUS
LKFKTVRPVALPRALAPPRN
1766
CAA28880.1





884
RUBELLA VIRUS
LNTPPPYQVSCGGESDRASAGH
1767
CAA28880.1





885
RUBELLA VIRUS
NCHGPDWASPVCQRHSPDCS
1768
CAA28880.1





886
RUBELLA VIRUS
PDCSRLVGATPERPRLRLVD
1769
CAA28880.1





887
RUBELLA VIRUS
PRNVRVTGCYQCGTPALVEG
1770
CAA28880.1





888
RUBELLA VIRUS
PTDVSCEGLGAWVPTAPCARI
1771
CAA28880.1





889
RUBELLA VIRUS
RLVDADDPLLRTAPGPGEVW
1772
CAA28880.1





890
RUBELLA VIRUS
SVFALASYVQHPHKTVRVKF
1773
CAA28880.1





891
RUBELLA VIRUS
VEGLAPGGGNCHLTVNGEDV
1774
CAA28880.1





892
RUBELLA VIRUS
VKFHTETRTVWQLSVAGVSC
1775
CAA28880.1





893
RUBELLA VIRUS
VPVRLAGVGFESKIVDGGCF
1776
CAA28880.1





894
RUBELLA VIRUS
VSCNVTTEHPFCNTPHGQLE
1777
CAA28880.1





895

BORDETELLA

AAASSPDAHVPF
1778
AAA22983.1




PERTUSSIS









896

BORDETELLA

AASSPDA
1779
AAA22983.1




PERTUSSIS









897

BORDETELLA

AKLGAAASSPDA
1780
AAA22983.1




PERTUSSIS









898

BORDETELLA

AMKPYEVTPTRM
1781
AAA22983.1




PERTUSSIS









899

BORDETELLA

AMTHLSPALADVPYVLVKTNMVVTS
1782
AAA22983.1




PERTUSSIS









900

BORDETELLA

ASSPDAHVPFCF
1783
AAA22983.1




PERTUSSIS









901

BORDETELLA

ASSPDAHVPFCFGKDLKRPGSSPME
1784
AAA22983.1




PERTUSSIS









902

BORDETELLA

CFGKDLKRPGSS
1785
AAA22983.1




PERTUSSIS









903

BORDETELLA

CFGKDLKRPGSSPMEVMLRAVFMQQ
1786
AAA22983.1




PERTUSSIS









904

BORDETELLA

CGIAAKLGAAAS
1787
AAA22983.1




PERTUSSIS









905

BORDETELLA

CGIAAKLGAAASSPDAHVPFCFGKD
1788
AAA22983.1




PERTUSSIS









906

BORDETELLA

DAHVPFCFGKDL
1789
AAA22983.1




PERTUSSIS









907

BORDETELLA

DLKRPGSSPMEV
1790
AAA22983.1




PERTUSSIS









908

BORDETELLA

DVPYVLVKTNMV
1791
AAA22983.1




PERTUSSIS









909

BORDETELLA

DVPYVLVKTNMVVTSVAMKPYEVTPT
1792
AAA22983.1




PERTUSSIS









910

BORDETELLA

EVMLRAVFMQQR
1793
AAA22983.1




PERTUSSIS









911

BORDETELLA

FEGKPALELIRM
1794
AAA22983.1




PERTUSSIS









912

BORDETELLA

FLGPKQLTFEGK
1795
AAA22983.1




PERTUSSIS









913

BORDETELLA

FLGPKQLTFEGKPALELIRMVECSG
1796
AAA22983.1




PERTUSSIS









914

BORDETELLA

FMQQRPLRMFLGPKQLT
1797
AAA22983.1




PERTUSSIS









915

BORDETELLA

GKDLKRPGSSPM
1798
AAA22983.1




PERTUSSIS









916

BORDETELLA

GKDLKRPGSSPME
1799
AAA22983.1




PERTUSSIS









917

BORDETELLA

GKPALELIRMVE
1800
AAA22983.1




PERTUSSIS









918

BORDETELLA

GPKQLTFEGKPA
1801
AAA22983.1




PERTUSSIS









919

BORDETELLA

HVPFCFGKDLKR
1802
AAA22983.1




PERTUSSIS









920

BORDETELLA

IAAKLGAAASSP
1803
AAA22983.1




PERTUSSIS









921

BORDETELLA

KPYEVTPTRMLV
1804
AAA22983.1




PERTUSSIS









922

BORDETELLA

KQLTFEGKPALE
1805
AAA22983.1




PERTUSSIS









923

BORDETELLA

KRPGSSPMEVML
1806
AAA22983.1




PERTUSSIS









924

BORDETELLA

LELIRMVECSGK
1807
AAA22983.1




PERTUSSIS









925

BORDETELLA

LGAAASSPDAHV
1808
AAA22983.1




PERTUSSIS









926

BORDETELLA

LIRMVECSGKQD
1809
AAA22983.1




PERTUSSIS









927

BORDETELLA

LVCGIAAKLGAA
1810
AAA22983.1




PERTUSSIS









928

BORDETELLA

MKPYEVTPTRM
1811
AAA22983.1




PERTUSSIS









929

BORDETELLA

MLRAVFMQQRPL
1812
AAA22983.1




PERTUSSIS









930

BORDETELLA

MQQRPLRM
1813
AAA22983.1




PERTUSSIS









931

BORDETELLA

MQQRPLRMFLGP
1814
AAA22983.1




PERTUSSIS









932

BORDETELLA

MVVTSVAMKPYE
1815
AAA22983.1




PERTUSSIS









933

BORDETELLA

MVVTSVAMKPYEVTPTRMLVCGIAA
1816
AAA22983.1




PERTUSSIS









934

BORDETELLA

PALELIRMVECS
1817
AAA22983.1




PERTUSSIS









935

BORDETELLA

PALELIRMVECSGK
1818
AAA22983.1




PERTUSSIS









936

BORDETELLA

PFCFGKDLKRPG
1819
AAA22983.1




PERTUSSIS









937

BORDETELLA

PGSSPMEVMLRA
1820
AAA22983.1




PERTUSSIS









938

BORDETELLA

PGSSPMEVMLRAVF
1821
AAA22983.1




PERTUSSIS









939

BORDETELLA

PKQLTFEGK
1822
AAA22983.1




PERTUSSIS









940

BORDETELLA

PLRMFLGPKQLT
1823
AAA22983.1




PERTUSSIS









941

BORDETELLA

PTRMLVCGIAAK
1824
AAA22983.1




PERTUSSIS









942

BORDETELLA

PYVLVKTNMVVT
1825
AAA22983.1




PERTUSSIS









943

BORDETELLA

QLTFEGKPALELIRMVECSGKQDCP
1826
AAA22983.1




PERTUSSIS









944

BORDETELLA

QRPLRMFLGPKQ
1827
AAA22983.1




PERTUSSIS









945

BORDETELLA

RAVFMQQRPLRM
1828
AAA22983.1




PERTUSSIS









946

BORDETELLA

RMFLGPKQLTFE
1829
AAA22983.1




PERTUSSIS









947

BORDETELLA

RMLVCGIAAKLG
1830
AAA22983.1




PERTUSSIS









948

BORDETELLA

RMVECSGKQDCP
1831
AAA22983.1




PERTUSSIS









949

BORDETELLA

SPDAHVPFCFGK
1832
AAA22983.1




PERTUSSIS









950

BORDETELLA

SSPMEVMLRAVF
1833
AAA22983.1




PERTUSSIS









951

BORDETELLA

SSPMEVMLRAVFMQQRPLRMFLGPK
1834
AAA22983.1




PERTUSSIS









952

BORDETELLA

SVAMKPYEVTPT
1835
AAA22983.1




PERTUSSIS









953

BORDETELLA

VECSGKQDCP
1836
AAA22983.1




PERTUSSIS









954

BORDETELLA

VFMQQRPLRMFL
1837
AAA22983.1




PERTUSSIS









955

BORDETELLA

VFMQQRPLRMFLGPKQLTFEGKPAL
1838
AAA22983.1




PERTUSSIS









956

BORDETELLA

VKTNMVVTSVAM
1839
AAA22983.1




PERTUSSIS









957

BORDETELLA

VLVKTNMVVTSV
1840
AAA22983.1




PERTUSSIS









958

BORDETELLA

VTPTRMLVCGIA
1841
AAA22983.1




PERTUSSIS









959

BORDETELLA

VTSVAMKPYEVT
1842
AAA22983.1




PERTUSSIS









960

BORDETELLA

YEVTPTRMLVCG
1843
AAA22983.1




PERTUSSIS









961

BORDETELLA

YEVTPTRMLVCGIAAKLGAAASSPD
1844
AAA22983.1




PERTUSSIS









962

BORDETELLA

CASPYEGRYRDMYDALR
1845
P04979.1




PERTUSSIS









963

BORDETELLA

CAVFVRDGQSV
1846
P04979.1




PERTUSSIS









964

BORDETELLA

CITTIYKTG
1847
P04979.1




PERTUSSIS









965

BORDETELLA

CPNGTRALTV
1848
P04979.1




PERTUSSIS









966

BORDETELLA

DALRRLLYMIYMSG
1849
P04979.1




PERTUSSIS









967
RUBELLA VIRUS
GNRGRGQRRDWSRAPPPPEERQETRS
1850
P07566.1



STRAIN THERIEN








968

BORDETELLA

GQPAADHYYSKVT
1851
P04979.1




PERTUSSIS









969
RUBELLA VIRUS
GSPNCHGPDWASPVCQRHS
1852
ABD64214.1





970
MEASLES VIRUS
HKSLSTNLDVTNSIEHQ
1853
P08362.1



STRAIN






EDMONSTON








971

BORDETELLA

LFTQQGGAYGRC
1854
P04979.1




PERTUSSIS









972
MEASLES VIRUS
LIGLLAIAGIRLHRAAIYTAEI
1855
P08362.1



STRAIN






EDMONSTON








973
MEASLES
PDTAADSELRRWIKY
1856
ABI54110.1



MORBILLIVIRUS








974
RUBELLA VIRUS
PNCHGPDWASPVCQRHS
1857
P07566.1



STRAIN THERIEN








975
RUBELLA VIRUS
QTPAPKPSRAPPQQPQPPRMQTGR
1858
ABD64216.1





976
RUBELLA VIRUS
RAGLTAGASQSRRPRPPR
1859
CAA33016.1



VACCINE STRAIN






RA27/3








977
RUBELLA VIRUS
RFGAPQAFLAGLLLAAVAVGTARA
1860
ABD64214.1



VACCINE STRAIN






RA27/3








978

BORDETELLA

RGNAELQTYLRQITPG
1861
P04979.1




PERTUSSIS









979

BORDETELLA

RVHVSKEEQYYDYED
1862
P04979.1




PERTUSSIS









980

BORDETELLA

SIYGLYDGTYL
1863
P04979.1




PERTUSSIS









981

BORDETELLA

SKVTATRLLASTNS
1864
P04979.1




PERTUSSIS









982

BORDETELLA

TQQGGAYGRCPNGTRA
1865
P04979.1




PERTUSSIS









983

BORDETELLA

VAPGIVIPPKAL
1866
P04979.1




PERTUSSIS









984

BORDETELLA

DSRPPEDVFQNGFTAWG
1867
ACI04548.1




PERTUSSIS









985

BORDETELLA

EHRMQEAVEAERAGR
1868
ACI04548.1




PERTUSSIS









986
MEASLES VIRUS
ETCFQQACKGKIQALCENPEWA
1869
P08362.1



STRAIN






EDMONSTON








987

BORDETELLA

EYVDTYGDNAGRILAGALATYQ
1870
ACI04548.1




PERTUSSIS









988

BORDETELLA

HRRIPPENIRRVTR
1871
ACI04548.1




PERTUSSIS









989

BORDETELLA

MARQAESSE
1872
ACI04548.1




PERTUSSIS









990

BORDETELLA

MQEAVEAERAGR
1873
ACI04548.1




PERTUSSIS









991

BORDETELLA

SQQTRANPNPYTSRR
1874
ACI04548.1




PERTUSSIS









992

BORDETELLA

TRANPNPYTSRRSVASIVGTLVHG
1875
SRC280066




PERTUSSIS









993

BORDETELLA

TVYRYDSRPPED
1876
ACI04548.1




PERTUSSIS









994
MEASLES
NDRNLLD
1877
P10050.1



MORBILLIVIRUS








995
MEASLES
NMEDEADQYFSHDDPISSDQSRFGWFENK
1878
P04851.1



MORBILLIVIRUS








996
MEASLES
SRASDARAAHLPTGTPLDID
1879
P04851.1



MORBILLIVIRUS








997

BORDETELLA

EDVFQNGFTAW
1880
ACI04548.1




PERTUSSIS









998

CORYNEBACTERIUM

AEGSSSVEYINNWEQAK
1881
AAV70486.1




DIPHTHERIAE









999

CORYNEBACTERIUM

GPIKNKMSESPNKT
1882
AAV70486.1




DIPHTHERIAE









1000
MEASLES VIRUS
GPKLTGALIGILSLFVESPGQLIQRITDDP
1883
P10050.1



STRAIN HALLE
DV







1001

CORYNEBACTERIUM

GYQKTVDHTKVNSK
1884
AAV70486.1




DIPHTHERIAE









1002

CORYNEBACTERIUM

KTVDH
1885
AAV70486.1




DIPHTHERIAE









1003

CORYNEBACTERIUM

SESPNK
1886
AAV70486.1




DIPHTHERIAE









1004

CORYNEBACTERIUM

AEGSSSVEYINNWEQAKALS
1887
AAV70486.1




DIPHTHERIAE









1005

CORYNEBACTERIUM

AQAIPLVGELVDIGFAAYNF
1888
AAV70486.1




DIPHTHERIAE









1006

CORYNEBACTERIUM

ASRVVLSLPFAEGSSSVEYI
1889
AAV70486.1




DIPHTHERIAE









1007

CORYNEBACTERIUM

CINLDWDVIRDKTKTKIESL
1890
AAV70486.1




DIPHTHERIAE









1008

CORYNEBACTERIUM

CRAIDGDVTFCRPKSPVYVG
1891
AAV70486.1




DIPHTHERIAE









1009

CORYNEBACTERIUM

CRPKSPVYVGNGVHANLHVA
1892
AAV70486.1




DIPHTHERIAE









1010

CORYNEBACTERIUM

DAAGYSVDNENPLSGKAGGV
1893
AAV70486.1




DIPHTHERIAE









1011

CORYNEBACTERIUM

DKTKTKIESLKEHGPIKNKM
1894
AAV70486.1




DIPHTHERIAE









1012

CORYNEBACTERIUM

EEFIKRFGDGASRVVLSLPF
1895
AAV70486.1




DIPHTHERIAE









1013

CORYNEBACTERIUM

EKAKQYLEEFHQTALEHPEL
1896
AAV70486.1




DIPHTHERIAE









1014

CORYNEBACTERIUM

FHRSSSEKIHSNEISSDSIG
1897
AAV70486.1




DIPHTHERIAE









1015

CORYNEBACTERIUM

GADDVVDSSKSFVMENFSSY
1898
CAE11230.1




DIPHTHERIAE









1016

CORYNEBACTERIUM

GKRGQDAMYEYMAQACAGNR
1899
AAV70486.1




DIPHTHERIAE









1017

CORYNEBACTERIUM

GSVMGIADGAVHHNTEEIVA
1900
AAV70486.1




DIPHTHERIAE









1018

CORYNEBACTERIUM

GTQGNYDDDWKGFYSTDNKY
1901
AAV70486.1




DIPHTHERIAE









1019

CORYNEBACTERIUM

HDGYAVSWNTVEDSIIRTGF
1902
AAV70486.1




DIPHTHERIAE









1020

CORYNEBACTERIUM

HGTKPGYVDSIQKGIQKPKS
1903
AAV70486.1




DIPHTHERIAE









1021

CORYNEBACTERIUM

HQTALEHPELSELKTVTGTN
1904
AAV70486.1




DIPHTHERIAE









1022

CORYNEBACTERIUM

IQKGIQKPKSGTQGNYDDDW
1905
AAV70486.1




DIPHTHERIAE









1023

CORYNEBACTERIUM

KEHGPIKNKMSESPNKTVSE
1906
AAV70486.1




DIPHTHERIAE









1024

CORYNEBACTERIUM

KGFYSTDNKYDAAGYSVDNE
1907
AAV70486.1




DIPHTHERIAE









1025

CORYNEBACTERIUM

LDVNKSKTHISVNGRKIRMR
1908
AAV70486.1




DIPHTHERIAE









1026
RUBELLA VIRUS
MASTIPITMEDLQKALEA
1909
SRC265968





1027

CORYNEBACTERIUM

NGVHANLHVAFHRSSSEKIH
1910
AAV70486.1




DIPHTHERIAE









1028

CORYNEBACTERIUM

NNWEQAKALSVELEINFETR
1911
AAV70486.1




DIPHTHERIAE









1029

CORYNEBACTERIUM

NPLSGKAGGVVKVTYPGLTK
1912
AAV70486.1




DIPHTHERIAE









1030

CORYNEBACTERIUM

PVFAGANYAAWAVNVAQVID
1913
AAV70486.1




DIPHTHERIAE









1031

CORYNEBACTERIUM

SELKTVTGTNPVFAGANYAA
1914
AAV70486.1




DIPHTHERIAE









1032

CORYNEBACTERIUM

SESPNKTVSEEKAKQYLEEF
1915
AAV70486.1




DIPHTHERIAE









1033

CORYNEBACTERIUM

SETADNLEKTTAALSILPGI
1916
AAV70486.1




DIPHTHERIAE









1034

CORYNEBACTERIUM

SFVMENFSSYHGTKPGYVDS
1917
AAV70486.1




DIPHTHERIAE









1035

CORYNEBACTERIUM

SNEISSDSIGVLGYQKTVDH
1918
AAV70486.1




DIPHTHERIAE









1036

CORYNEBACTERIUM

SPGHKTQPFLHDGYAVSWNT
1919
AAV70486.1




DIPHTHERIAE









1037

CORYNEBACTERIUM

SVNGRKIRMRCRAIDGDVTF
1920
AAV70486.1




DIPHTHERIAE









1038

CORYNEBACTERIUM

TAALSILPGIGSVMGIADGA
1921
AAV70486.1




DIPHTHERIAE









1039

CORYNEBACTERIUM

TAENTPLPIAGVLLPTIPGK
1922
AAV70486.1




DIPHTHERIAE









1040

CORYNEBACTERIUM

TEPLMEQVGTEEFIKRFGDG
1923
AAV70486.1




DIPHTHERIAE









1041

CORYNEBACTERIUM

TIKKELGLSLTEPLMEQVGT
1924
AAV70486.1




DIPHTHERIAE









1042

CORYNEBACTERIUM

TKVNSKLSLFFEIKS
1925
AAV70486.1




DIPHTHERIAE









1043

CORYNEBACTERIUM

VEDSIIRTGFQGESGHDIKI
1926
AAV70486.1




DIPHTHERIAE









1044

CORYNEBACTERIUM

VELEINFETRGKRGQDAMYE
1927
AAV70486.1




DIPHTHERIAE









1045

CORYNEBACTERIUM

VESIINLFQVVHNSYNRPAY
1928
AAV70486.1




DIPHTHERIAE









1046

CORYNEBACTERIUM

VHNSYNRPAYSPGHKTQPFL
1929
AAV70486.1




DIPHTHERIAE









1047

CORYNEBACTERIUM

VKVTYPGLTKVLALKVDNAE
1930
AAV70486.1




DIPHTHERIAE









1048

CORYNEBACTERIUM

VLALKVDNAETIKKELGLSL
1931
AAV70486.1




DIPHTHERIAE









1049

CORYNEBACTERIUM

VLGYQKTVDHTKVNSKLSLF
1932
AAV70486.1




DIPHTHERIAE









1050

CORYNEBACTERIUM

VRRSVGSSLSCINLDWDVIR
1933
AAV70486.1




DIPHTHERIAE









1051

CORYNEBACTERIUM

WAVNVAQVIDSETADNLEKT
1934
AAV70486.1




DIPHTHERIAE









1052

CORYNEBACTERIUM

YMAQACAGNRVRRSVGSSLS
1935
AAV70486.1




DIPHTHERIAE









1053

BORDETELLA

AKAPPAPKPAPQPGP
1936
ABO77783.1




PERTUSSIS









1054

BORDETELLA

APKPAPQPGP
1937
ABO77783.1




PERTUSSIS









1055

BORDETELLA

APKPAPQPGPQPPQP
1938
ABO77783.1




PERTUSSIS









1056
MEASLES VIRUS
AQTRTPLQCTMTEIF
1939
P04851.1



STRAIN






EDMONSTON








1057
MEASLES VIRUS
ASRGTNMEDEADQYFSHDD
1940
P04851.1



STRAIN






EDMONSTON








1058

BORDETELLA

ATIRR
1941
ABO77783.1




PERTUSSIS









1059

BORDETELLA

DNRAG
1942
ABO77783.1




PERTUSSIS









1060

BORDETELLA

EAPAPQPPAGRELSA
1943
ABO77783.1




PERTUSSIS









1061
MEASLES VIRUS
EMVRRSAGKVSSTLASELGI
1944
P04851.1



STRAIN






EDMONSTON








1062

BORDETELLA

GASEL
1945
ABO77783.1




PERTUSSIS









1063

BORDETELLA

GDALAGGAVP
1946
AAA22980.1




PERTUSSIS









1064

BORDETELLA

GDAPAGGAVP
1947
ABO77783.1




PERTUSSIS









1065

BORDETELLA

GDTWDDD
1948
ABO77783.1




PERTUSSIS









1066

BORDETELLA

GERQH
1949
ABO77783.1




PERTUSSIS









1067

BORDETELLA

GGAVP
1950
ABO77783.1




PERTUSSIS









1068

BORDETELLA

GGFGP
1951
P14283.3




PERTUSSIS









1069

BORDETELLA

GGFGPGGFGP
1952
BAF35031.1




PERTUSSIS









1070

BORDETELLA

GGFGPVLDGW
1953
ABO77783.1




PERTUSSIS









1071
MEASLES VIRUS
GGKEDRRVKQSRGEARESYR
1954
P04851.1



STRAIN






EDMONSTON








1072

BORDETELLA

GILLEN
1955
ABO77783.1




PERTUSSIS









1073

BORDETELLA

GIRRFL
1956
ABO77783.1




PERTUSSIS









1074
MEASLES VIRUS
HDDPISSDQSRFGWFENKEI
1957
P04851.1



STRAIN






EDMONSTON








1075
MEASLES VIRUS
HEFAGELSTLESLMNLY
1958
P04851.1



STRAIN






EDMONSTON








1076

BORDETELLA

HLGGLAGY
1959
ABO77783.1




PERTUSSIS









1077
MEASLES VIRUS
HTTEDKISRAVGPRQAQVSFL
1960
P04851.1



STRAIN






EDMONSTON








1078
MEASLES VIRUS
ICDIDTYIVEAGLASFILTI
1961
P04851.1



STRAIN






EDMONSTON








1079
MEASLES VIRUS
IIIVPIPGDSSITTRSRLLD
1962
P04851.1



STRAIN






EDMONSTON








1080
MEASLES VIRUS
ILDIKRTPGNKPRIAEMICD
1963
P04851.1



STRAIN






EDMONSTON








1081

BORDETELLA

KALLYR
1964
ABO77783.1




PERTUSSIS









1082
MEASLES VIRUS
KPPITSGSGGAIRGIKHIII
1965
P04851.1



STRAIN






EDMONSTON








1083

BORDETELLA

LAGSGL
1966
ABO77783.1




PERTUSSIS









1084
MEASLES VIRUS
LGITAEDARLVSEIAMHTTE
1967
P04851.1



STRAIN






EDMONSTON








1085
MEASLES VIRUS
LGTILAQIWVLLAKAVTA
1968
P04851.1



STRAIN






EDMONSTON








1086
MEASLES VIRUS
LPTGTPLDIDTASESSQD
1969
P04851.1



STRAIN






EDMONSTON








1087
MEASLES VIRUS
MATLLRSLALFKRNKDK
1970
P04851.1



STRAIN






EDMONSTON








1088

BORDETELLA

PAPQPP
1971
ABO77783.1




PERTUSSIS









1089
MEASLES VIRUS
PDTAADSELRRWIKYTQQRR
1972
P04851.1



STRAIN






EDMONSTON








1090
MEASLES VIRUS
PKLTGALIGILSLFVESPGQ
1973
P04851.1



STRAIN






EDMONSTON








1091

BORDETELLA

PQP
1974
ABO77783.1




PERTUSSIS









1092

BORDETELLA

PQPGP
1975
ABO77783.1




PERTUSSIS









1093

BORDETELLA

PQPGPQPPQPPQPQP
1976
ABO77783.1




PERTUSSIS









1094
MEASLES VIRUS
QDPQDSRRSAEPLLSCKPWQ
1977
P04851.1



STRAIN






EDMONSTON








1095
MEASLES VIRUS
QRRVVGEFRLERKWLDVVR
1978
P04851.1



STRAIN






EDMONSTON








1096

BORDETELLA

RELSA
1979
ABO77783.1




PERTUSSIS









1097

BORDETELLA

RFAPQ
1980
ABO77783.1




PERTUSSIS









1098
MEASLES VIRUS
SIQNKFSAGSYPLLWSYAMG
1981
P04851.1



STRAIN






EDMONSTON








1099

BORDETELLA

SITLQAGAH
1982
ABO77783.1




PERTUSSIS









1100

BORDETELLA

SLQPED
1983
ABO77783.1




PERTUSSIS









1101

BORDETELLA

SNALSKRL
1984
ABO77783.1




PERTUSSIS









1102
MEASLES VIRUS
SPGQLIQRITDDPDVSIRLL
1985
P04851.1



STRAIN






EDMONSTON








1103

BORDETELLA

TELPSIPG
1986
ABO77783.1




PERTUSSIS









1104

BORDETELLA

TFTLANK
1987
ABO77783.1




PERTUSSIS









1105

BORDETELLA

TWDDD
1988
ABO77783.1




PERTUSSIS









1106
MEASLES VIRUS
VSFLQGDQSENELPRLGGKE
1989
P04851.1



STRAIN






EDMONSTON








1107
MEASLES VIRUS
WQESRKNKAQTRTPLQC
1990
P04851.1



STRAIN






EDMONSTON








1108
MEASLES VIRUS
YAMGVGVELENSMGGLNFGR
1991
P04851.1



STRAIN






EDMONSTON








1109
MEASLES VIRUS
YQQMGKPAPYMVNLENSI
1992
P04851.1



STRAIN






EDMONSTON








1110
MEASLES VIRUS
MTRSSHQSLVIKLMP
1993
P69355.1



STRAIN HALLE








1111
MEASLES VIRUS
PIRDALNAMTQNIRP
1994
P69355.1



STRAIN HALLE








1112
RUBELLA VIRUS
ALLNTPPPYQVSCGGESDRASAGH
1995
CAA28880.1



STRAIN M33








1113
RUBELLA VIRUS
GLGSPNCHGPDWASPVCQRHS
1996
P07566.1



STRAIN THERIEN








1114
RUBELLA VIRUS
GLGSPNCHGPDWASPVCQRHSPDCSRLV
1997
P07566.1



STRAIN THERIEN








1115
RUBELLA VIRUS
NYTGNQQSRWGLGSPNCHGPDWASPV
1998
P07566.1



STRAIN THERIEN








1116
RUBELLA VIRUS
TLPQPPCAHGQHYGHHHHQL
1999
P07566.1



STRAIN THERIEN








1117
RUBELLA VIRUS
TVRVKFHTETRTVWQLSVAGVSCNVT
2000
P07566.1



STRAIN THERIEN








1118
RUBELLA VIRUS
ASYFNPGGSYYKQYH
2001
BAA28178.1





1119
RUBELLA VIRUS
FALASYVQHPHKTVR
2002
BAA28178.1





1120
RUBELLA VIRUS
GGESDRASARVIDPAAQSFTG
2003
BAA28178.1





1121
RUBELLA VIRUS
GPGEVWVTPVIGSQARKC
2004
BAA28178.1





1122
RUBELLA VIRUS
GSQARKCGLHIRAG
2005
BAA28178.1





1123
RUBELLA VIRUS
LVGATPERPRLRLVDADDPLLRTAP
2006
BAA28178.1





1124
RUBELLA VIRUS
TAPGPGEVWVTPVI
2007
BAA28178.1





1125
MEASLES
FIVLSIAYPTLSEIK
2008
AAL29688.1



MORBILLIVIRUS








1126
MEASLES VIRUS
ETCFQQACKGKIQALCENPEWAPLKDNRIP
2009
P08362.1



STRAIN
SY





EDMONSTON-






ZAGREB








1127
MEASLES
LPRLGGKEDR
2010
P04851.1



MORBILLIVIRUS








1128
MEASLES
MSKTEWNASQ
2011
SRC280080



MORBILLIVIRUS








1129
MEASLES
SRFGWFENKE
2012
P04851.1



MORBILLIVIRUS








1130
RUBELLA VIRUS
EACVTSWLWSEGEGAVFYRVDLHFINLGT
2013
CAA28880.1





1131
RUBELLA VIRUS
MDFWCVEHDRPPPATPTSLTT
2014
CAA33016.1





1132
RUBELLA VIRUS
PFLGHDGHHGGTLRVGQHHRNASDV
2015
CAA33016.1





1133
RUBELLA VIRUS
RVKFHTETRTVWQLSVAGVSC
2016
BAA19893.1





1134
MEASLES
AEEQARHVKNGLE
2017
ABO69699.1



MORBILLIVIRUS








1135
MEASLES
ESPQEISKHQALG
2018
SRC280080



MORBILLIVIRUS








1136
MEASLES
GVGVELENSMGGLNF
2019
ABI54110.1



MORBILLIVIRUS








1137
MEASLES
IKGANDLAKFHQMLMKIIMK
2020
ABO69699.1



MORBILLIVIRUS








1138
MEASLES
MSKTLHAQLGFKKT
2021
ABK40528.1



MORBILLIVIRUS








1139
MEASLES
NASGLSRPSPSAH
2022
BAE98296.1



MORBILLIVIRUS








1140
MEASLES
VRVIDPSLGDRKDE
2023
SRC280148



MORBILLIVIRUS








1141

BORDETELLA

DLSDGDLLV
2024
AAC31207.1




PERTUSSIS









1142

BORDETELLA

EAERAGRGTG
2025
ACI04548.1




PERTUSSIS









1143

BORDETELLA

YRYDSRPPEDV
2026
ACI04548.1




PERTUSSIS









1144

BORDETELLA

YVDTYGDNAG
2027
ACI04548.1




PERTUSSIS









1145
MEASLES
SFSYFYPFR
2028
CAB43772.1



MORBILLIVIRUS








1146
MUMPS
DIFIVSPR
2029
ADF49557.1



RUBULAVIRUS








1147
MEASLES
QDSRRSADALLRLQAMAGISEEQGSDTDTP
2030
CAA59302.1



MORBILLIVIRUS
IVYNDRN







1148
MEASLES
SAEALLRLQA
2031
BAH22350.1



MORBILLIVIRUS








1149
MEASLES
RIVINREHL
2032
BAB39835.1



MORBILLIVIRUS








1150
MEASLES VIRUS
IPRFK
2033
BAB39848.1



GENOTYPE A








1151

BORDETELLA

AAALSPMEI
2034
P15318.2




PERTUSSIS









1152

BORDETELLA

AAASVVGAPV
2035
P15318.2




PERTUSSIS









1153

BORDETELLA

AALGRQDSI
2036
P15318.2




PERTUSSIS









1154

BORDETELLA

AAQRLVHAIA
2037
P15318.2




PERTUSSIS









1155

BORDETELLA

AAVEAAEL
2038
P15318.2




PERTUSSIS









1156

BORDETELLA

AGANVLNGL
2039
P15318.2




PERTUSSIS









1157

BORDETELLA

AGYANAAD
2040
P15318.2




PERTUSSIS









1158

BORDETELLA

AGYEQFEFRV
2041
P15318.2




PERTUSSIS









1159

BORDETELLA

AITGNADNL
2042
P15318.2




PERTUSSIS









1160

BORDETELLA

AKEKNATLM
2043
P15318.2




PERTUSSIS









1161

BORDETELLA

AKGVFLSL
2044
P15318.2




PERTUSSIS









1162

BORDETELLA

APHEYGFGI
2045
P15318.2




PERTUSSIS









1163

BORDETELLA

ARQGNDLEI
2046
P15318.2




PERTUSSIS









1164

BORDETELLA

ASVVGAPV
2047
P15318.2




PERTUSSIS









1165

BORDETELLA

ATLMFRLV
2048
P15318.2




PERTUSSIS









1166

BORDETELLA

AVAAAQRL
2049
P15318.2




PERTUSSIS









1167

BORDETELLA

DAGANVLNGL
2050
P15318.2




PERTUSSIS









1168

BORDETELLA

DALLAQLYR
2051
P15318.2




PERTUSSIS









1169

BORDETELLA

DANGVLKHSI
2052
P15318.2




PERTUSSIS









1170

BORDETELLA

DGDMNIGVI
2053
P15318.2




PERTUSSIS









1171

BORDETELLA

DHVKNIENL
2054
P15318.2




PERTUSSIS









1172

BORDETELLA

DIDMFAIM
2055
P15318.2




PERTUSSIS









1173

BORDETELLA

DMFAIMPHL
2056
P15318.2




PERTUSSIS









1174

BORDETELLA

DNVRNVENV
2057
P15318.2




PERTUSSIS









1175

BORDETELLA

DNVRNVENVI
2058
P15318.2




PERTUSSIS









1176

BORDETELLA

DTVDYSAM
2059
P15318.2




PERTUSSIS









1177

BORDETELLA

DTVDYSAMI
2060
P15318.2




PERTUSSIS









1178

BORDETELLA

DYLRQAGL
2061
P15318.2




PERTUSSIS









1179

BORDETELLA

DYYDNVRNV
2062
P15318.2




PERTUSSIS









1180

BORDETELLA

EFTTFVEI
2063
P15318.2




PERTUSSIS









1181

BORDETELLA

EFTTFVEIV
2064
P15318.2




PERTUSSIS









1182

BORDETELLA

EGYVFYEN
2065
P15318.2




PERTUSSIS









1183

BORDETELLA

ENVQYRHV
2066
P15318.2




PERTUSSIS









1184

BORDETELLA

EQLANSDGL
2067
P15318.2




PERTUSSIS









1185

BORDETELLA

FGVGYGHDTI
2068
P15318.2




PERTUSSIS









1186

BORDETELLA

FSPDVLETVP
2069
P15318.2




PERTUSSIS









1187

BORDETELLA

FSVDHVKNI
2070
P15318.2




PERTUSSIS









1188

BORDETELLA

GDDTYLFGV
2071
P15318.2




PERTUSSIS









1189

BORDETELLA

GDDVFLQDL
2072
P15318.2




PERTUSSIS









1190

BORDETELLA

GEDGNDIFL
2073
P15318.2




PERTUSSIS









1191

BORDETELLA

GERFNVRKQL
2074
P15318.2




PERTUSSIS









1192

BORDETELLA

GGAGNDTLV
2075
P15318.2




PERTUSSIS









1193

BORDETELLA

GGDDFEAV
2076
P15318.2




PERTUSSIS









1194

BORDETELLA

GKSEFTTFV
2077
P15318.2




PERTUSSIS









1195

BORDETELLA

GKSLFDDGL
2078
P15318.2




PERTUSSIS









1196

BORDETELLA

GNADNLKSV
2079
P15318.2




PERTUSSIS









1197

BORDETELLA

GQLVEVDTL
2080
P15318.2




PERTUSSIS









1198

BORDETELLA

GRSKFSPDV
2081
P15318.2




PERTUSSIS









1199

BORDETELLA

GSSAYDTV
2082
P15318.2




PERTUSSIS









1200

BORDETELLA

GTVEKWPAL
2083
P15318.2




PERTUSSIS









1201

BORDETELLA

GVDYYDNV
2084
P15318.2




PERTUSSIS









1202

BORDETELLA

GYEQFEFRV
2085
P15318.2




PERTUSSIS









1203

BORDETELLA

HAVGAQDVV
2086
P15318.2




PERTUSSIS









1204

BORDETELLA

IAAGRIGLGI
2087
P15318.2




PERTUSSIS









1205

BORDETELLA

IGDAQANTL
2088
P15318.2




PERTUSSIS









1206

BORDETELLA

IGLGILADL
2089
P15318.2




PERTUSSIS









1207

BORDETELLA

IGNAAGIPL
2090
P15318.2




PERTUSSIS









1208

BORDETELLA

IGTSMKDVL
2091
P15318.2




PERTUSSIS









1209

BORDETELLA

IGVITDFEL
2092
P15318.2




PERTUSSIS









1210

BORDETELLA

IPLTADIDM
2093
P15318.2




PERTUSSIS









1211

BORDETELLA

ISKSALEL
2094
P15318.2




PERTUSSIS









1212

BORDETELLA

ITGNADNL
2095
P15318.2




PERTUSSIS









1213

BORDETELLA

KIFVVSAT
2096
P15318.2




PERTUSSIS









1214

BORDETELLA

KQLNNANVYR
2097
P15318.2




PERTUSSIS









1215

BORDETELLA

KVIGNAAGI
2098
P15318.2




PERTUSSIS









1216

BORDETELLA

LAKVVSQL
2099
P15318.2




PERTUSSIS









1217

BORDETELLA

LANDYARKI
2100
P15318.2




PERTUSSIS









1218

BORDETELLA

LDYLRQAGL
2101
P15318.2




PERTUSSIS









1219

BORDETELLA

LGKGFASL
2102
P15318.2




PERTUSSIS









1220

BORDETELLA

LGKGFASLM
2103
P15318.2




PERTUSSIS









1221

BORDETELLA

LGVDYYDN
2104
P15318.2




PERTUSSIS









1222

BORDETELLA

LGVDYYDNV
2105
P15318.2




PERTUSSIS









1223

BORDETELLA

LKHSIKLDVI
2106
P15318.2




PERTUSSIS









1224

BORDETELLA

LQAGYIPV
2107
P15318.2




PERTUSSIS









1225

BORDETELLA

LQLTGGTVE
2108
P15318.2




PERTUSSIS









1226

BORDETELLA

LSAAVFGL
2109
P15318.2




PERTUSSIS









1227

BORDETELLA

LSLGKGFASL
2110
P15318.2




PERTUSSIS









1228

BORDETELLA

LSPMEIYGL
2111
P15318.2




PERTUSSIS









1229

BORDETELLA

NAHDNFLAGG
2112
P15318.2




PERTUSSIS









1230

BORDETELLA

NANVYREGV
2113
P15318.2




PERTUSSIS









1231

BORDETELLA

NDTLYGGL
2114
P15318.2




PERTUSSIS









1232

BORDETELLA

NGLAGNDVL
2115
P15318.2




PERTUSSIS









1233

BORDETELLA

NNANVYREGV
2116
P15318.2




PERTUSSIS









1234

BORDETELLA

NTVSYAAL
2117
P15318.2




PERTUSSIS









1235

BORDETELLA

NVLRNIENAV
2118
P15318.2




PERTUSSIS









1236

BORDETELLA

PALTFITPL
2119
P15318.2




PERTUSSIS









1237

BORDETELLA

PETSNVLRNI
2120
P15318.2




PERTUSSIS









1238

BORDETELLA

PMEIYGLV
2121
P15318.2




PERTUSSIS









1239

BORDETELLA

PQAYFEKNL
2122
P15318.2




PERTUSSIS









1240

BORDETELLA

PVNPNLSKL
2123
P15318.2




PERTUSSIS









1241

BORDETELLA

QAGWNASSV
2124
P15318.2




PERTUSSIS









1242

BORDETELLA

QAGWNASSVI
2125
P15318.2




PERTUSSIS









1243

BORDETELLA

QDAANAGNL
2126
P15318.2




PERTUSSIS









1244

BORDETELLA

QDAANAGNLL
2127
P15318.2




PERTUSSIS









1245

BORDETELLA

QDSGYDSL
2128
P15318.2




PERTUSSIS









1246

BORDETELLA

QQSHYADQL
2129
P15318.2




PERTUSSIS









1247

BORDETELLA

RALQGAQAV
2130
P15318.2




PERTUSSIS









1248

BORDETELLA

RGGLGLDTL
2131
P15318.2




PERTUSSIS









1249

BORDETELLA

RKQLNNANV
2132
P15318.2




PERTUSSIS









1250

BORDETELLA

RQDSGYDSL
2133
P15318.2




PERTUSSIS









1251

BORDETELLA

RQFRYDGDM
2134
P15318.2




PERTUSSIS









1252

BORDETELLA

RSKFSPDVL
2135
P15318.2




PERTUSSIS









1253

BORDETELLA

SAGAAAGAL
2136
P15318.2




PERTUSSIS









1254

BORDETELLA

SAHWGQRAL
2137
P15318.2




PERTUSSIS









1255

BORDETELLA

SAMIHPGRI
2138
P15318.2




PERTUSSIS









1256

BORDETELLA

SAMIHPGRIV
2139
P15318.2




PERTUSSIS









1257

BORDETELLA

SAYDTVSGI
2140
P15318.2




PERTUSSIS









1258

BORDETELLA

SAYGYEGD
2141
P15318.2




PERTUSSIS









1259

BORDETELLA

SGGAGDDVL
2142
P15318.2




PERTUSSIS









1260

BORDETELLA

SGLQVAGA
2143
P15318.2




PERTUSSIS









1261

BORDETELLA

SGYDSLDGV
2144
P15318.2




PERTUSSIS









1262

BORDETELLA

SLLTGALNGI
2145
P15318.2




PERTUSSIS









1263

BORDETELLA

SPMEIYGL
2146
P15318.2




PERTUSSIS









1264

BORDETELLA

SQMLTRGQL
2147
P15318.2




PERTUSSIS









1265

BORDETELLA

SSAYDTVSGI
2148
P15318.2




PERTUSSIS









1266

BORDETELLA

SSLAHGHTA
2149
P15318.2




PERTUSSIS









1267

BORDETELLA

SSVTSGDSV
2150
P15318.2




PERTUSSIS









1268

BORDETELLA

SVIGVQTTEI
2151
P15318.2




PERTUSSIS









1269

BORDETELLA

TNTVSYAAL
2152
P15318.2




PERTUSSIS









1270

BORDETELLA

TSLIAEGV
2153
P15318.2




PERTUSSIS









1271

BORDETELLA

TSLLTGAL
2154
P15318.2




PERTUSSIS









1272

BORDETELLA

TVPASPGL
2155
P15318.2




PERTUSSIS









1273

BORDETELLA

VAKEKNATL
2156
P15318.2




PERTUSSIS









1274

BORDETELLA

VAPHEYGFGI
2157
P15318.2




PERTUSSIS









1275

BORDETELLA

VAVVTSLL
2158
P15318.2




PERTUSSIS









1276

BORDETELLA

VFYENRAYG
2159
P15318.2




PERTUSSIS









1277

BORDETELLA

VFYENRAYGV
2160
P15318.2




PERTUSSIS









1278

BORDETELLA

VITDFELEV
2161
P15318.2




PERTUSSIS









1279

BORDETELLA

VNPHSTSL
2162
P15318.2




PERTUSSIS









1280

BORDETELLA

VNPHSTSLI
2163
P15318.2




PERTUSSIS









1281

BORDETELLA

VNPNLSKL
2164
P15318.2




PERTUSSIS









1282

BORDETELLA

VNPNLSKLF
2165
P15318.2




PERTUSSIS









1283

BORDETELLA

VQQPIIEKL
2166
P15318.2




PERTUSSIS









1284

BORDETELLA

VQYRHVEL
2167
P15318.2




PERTUSSIS









1285

BORDETELLA

VSIAAAASV
2168
P15318.2




PERTUSSIS









1286

BORDETELLA

VSIAAAASVV
2169
P15318.2




PERTUSSIS









1287

BORDETELLA

VTSLLTGAL
2170
P15318.2




PERTUSSIS









1288

BORDETELLA

VVLANASRI
2171
P15318.2




PERTUSSIS









1289

BORDETELLA

WPALNLFSV
2172
P15318.2




PERTUSSIS









1290

BORDETELLA

WVRKASAL
2173
P15318.2




PERTUSSIS









1291

BORDETELLA

YAVQYRRKGG
2174
P15318.2




PERTUSSIS









1292

BORDETELLA

YGGLGDDTL
2175
P15318.2




PERTUSSIS









1293

BORDETELLA

YGLVQQSHYA
2176
P15318.2




PERTUSSIS









1294

BORDETELLA

YGYEGDALL
2177
P15318.2




PERTUSSIS









1295

BORDETELLA

YIPVNPNL
2178
P15318.2




PERTUSSIS









1296

BORDETELLA

YSAMIHPGRI
2179
P15318.2




PERTUSSIS









1297

BORDETELLA

YSQTGAHAGI
2180
P15318.2




PERTUSSIS









1298

CORYNEBACTERIUM

AYNFVESIINLFQVVHNSYN
2181
CAE11230.1




DIPHTHERIAE









1299

BORDETELLA

SGTTIK
2182
BAF35031.1




PERTUSSIS









1300

BORDETELLA

RGHTLESAEGRKIFG
2183
AAA22974.1




PERTUSSIS









1301

BORDETELLA

AGAMTVRDVAAAADLALQAGDA
2184
AAA22974.1




PERTUSSIS









1302

BORDETELLA

AGAMTVRDVAAAADLALQAGDAL
2185
AAA22974.1




PERTUSSIS









1303

BORDETELLA

ALAAVLVNPHIFTRIGAAQTSLADGAAGPA
2186
AAA22974.1




PERTUSSIS









1304

BORDETELLA

ALSIDSMTALGA
2187
AAA22974.1




PERTUSSIS









1305

BORDETELLA

DLSAARGADISGEGR
2188
AAA22974.1




PERTUSSIS









1306

BORDETELLA

DQNRYEYIWGLY
2189
AAA22974.1




PERTUSSIS









1307

BORDETELLA

DYTVSADAIALA
2190
AAA22974.1




PERTUSSIS









1308

BORDETELLA

GPIVVEAGELVSHAGG
2191
AAA22974.1




PERTUSSIS









1309

BORDETELLA

GRPEGLKIGAHSATSVSGSFDAL
2192
AAA22974.1




PERTUSSIS









1310

BORDETELLA

ITVTSRGGFDNEGKMESNK
2193
AAA22974.1




PERTUSSIS









1311

BORDETELLA

LDQNRYEYIWGLYP
2194
AAA22974.1




PERTUSSIS









1312

BORDETELLA

LSAARGADISG
2195
AAA22974.1




PERTUSSIS









1313

BORDETELLA

NKIRLMGPLQ
2196
AAA22974.1




PERTUSSIS









1314

BORDETELLA

NKLGRIRAGEDM
2197
AAA22974.1




PERTUSSIS









1315

BORDETELLA

NKLGRIRAGEDMHLDAPRIE
2198
AAA22974.1




PERTUSSIS









1316

BORDETELLA

PHLRNTGQVVAG
2199
AAA22974.1




PERTUSSIS









1317

BORDETELLA

QVDLHDLSAARGADISG
2200
AAA22974.1




PERTUSSIS









1318

BORDETELLA

RDVAAAADLALQ
2201
AAA22974.1




PERTUSSIS









1319

BORDETELLA

SAARGADISGEG
2202
AAA22974.1




PERTUSSIS









1320

BORDETELLA

TKGEMQIAGKGGGSP
2203
AAA22974.1




PERTUSSIS









1321

BORDETELLA

TVSADAIALAAQ
2204
AAA22974.1




PERTUSSIS









1322

BORDETELLA

VVAGHDIHI
2205
AAA22974.1




PERTUSSIS









1323

BORDETELLA

PSGPNHTKVVQLPKISKNALKANG
2206
CAD12823.1




PERTUSSIS









1324
RUBELLA VIRUS
LVGATPERPRLRLVDADDPLLRTAPGPGEV
2207
BAA 19902.1




WVTPVIGSQAR







1325
RUBELLA VIRUS
QQSRWGLGSPNCHGPDWASPVCQRHSP
2208
BAA 19902.1





1326

BORDETELLA

AGEAMVLVYYESIAYSF
2209
ACI04548.1




PERTUSSIS









1327

BORDETELLA

GGVGLASTLWYAESNALSKRLGEL
2210
AAZ74322.1




PERTUSSIS









1328

BORDETELLA

GTLVRIAPVIGACMARQA
2211
ACI04548.1




PERTUSSIS









1329

BORDETELLA

IRRVTRVYHNGITGETTT
2212
ACI04548.1




PERTUSSIS









1330

BORDETELLA

IVKTGERQHGIHIQGSDP
2213
AAZ74322.1




PERTUSSIS









1331

BORDETELLA

IVKTGERQHGIHIQGSDPGGVRTA
2214
AAZ74338.1




PERTUSSIS









1332

BORDETELLA

LRDTNVTAVPASGAPAAVSVLGAS
2215
AAZ74338.1




PERTUSSIS









1333

BORDETELLA

PEAPAPQPPAGRELSAAANAAVNT
2216
AAZ74322.1




PERTUSSIS









1334

BORDETELLA

AAADFAHAE
2217
WP_019247158.1




PERTUSSIS









1335

BORDETELLA

AAAEVAGAL
2218
WP_019249248.1




PERTUSSIS









1336

BORDETELLA

AAESTFESY
2219
WP_019247158.1




PERTUSSIS









1337

BORDETELLA

AAGFDPEVQ
2220
WP_019248145.1




PERTUSSIS









1338

BORDETELLA

AALGRGHSL
2221
AGS56996.1




PERTUSSIS









1339

BORDETELLA

AAMQGAVVH
2222
AGT50936.1




PERTUSSIS









1340

BORDETELLA

AAPAAHADW
2223
AGS56996.1




PERTUSSIS









1341

BORDETELLA

AAQATVVQR
2224
WP_019247158.1




PERTUSSIS









1342

BORDETELLA

AARVAGDNY
2225
WP_019249248.1




PERTUSSIS









1343

BORDETELLA

AAVALLNKL
2226
WP_019249248.1




PERTUSSIS









1344

BORDETELLA

ADDPPATVY
2227
AAW72734.1




PERTUSSIS









1345

BORDETELLA

AEAGRFKVL
2228
AGS56996.1




PERTUSSIS









1346

BORDETELLA

AEATQLVTA
2229
WP_019247158.1




PERTUSSIS









1347

BORDETELLA

AEGGATLGA
2230
WP_019249248.1




PERTUSSIS









1348

BORDETELLA

AEHGEVSIQ
2231
WP_019249248.1




PERTUSSIS









1349

BORDETELLA

AEIAFYPKE
2232
WP_019249248.1




PERTUSSIS









1350

BORDETELLA

AEKVTTPAV
2233
WP_019247158.1




PERTUSSIS









1351

BORDETELLA

AELQTYLRQ
2234
1BCP_C




PERTUSSIS









1352

BORDETELLA

AEQSLIEVG
2235
WP_019249248.1




PERTUSSIS









1353

BORDETELLA

AESNALSKR
2236
AGS56996.1




PERTUSSIS









1354

BORDETELLA

AESSEAMAA
2237
AFK26302.1




PERTUSSIS









1355

BORDETELLA

AEVKVGYRA
2238
WP_019247158.1




PERTUSSIS









1356

BORDETELLA

AEVTDTSPS
2239
WP_019249248.1




PERTUSSIS









1357

BORDETELLA

AGKSLKKKN
2240
WP_019247158.1




PERTUSSIS









1358

BORDETELLA

AGLAGPSAV
2241
WP_019249248.1




PERTUSSIS









1359

BORDETELLA

AIRVGRGAR
2242
AGT50936.1




PERTUSSIS









1360

BORDETELLA

ALAAIASAA
2243
WP_019248145.1




PERTUSSIS









1361

BORDETELLA

ALADVPYVL
2244
AAA22983.1




PERTUSSIS









1362

BORDETELLA

ALANDGTIV
2245
WP_019248658.1




PERTUSSIS









1363

BORDETELLA

ALGRGHSLY
2246
AGS56996.1




PERTUSSIS









1364

BORDETELLA

ALILAASPV
2247
WP_019248658.1




PERTUSSIS









1365

BORDETELLA

ALMLACTGL
2248
AAA22974.1




PERTUSSIS









1366

BORDETELLA

AMQGAVVHL
2249
AGT50936.1




PERTUSSIS









1367

BORDETELLA

AMTHLSPAL
2250
AAA22983.1




PERTUSSIS









1368

BORDETELLA

AMYGKHITL
2251
WP_019249248.1




PERTUSSIS









1369

BORDETELLA

ANEANALLW
2252
WP_019249248.1




PERTUSSIS









1370

BORDETELLA

APLSITLQA
2253
AGT50936.1




PERTUSSIS









1371

BORDETELLA

APNALAWAL
2254
AAA22974.1




PERTUSSIS









1372

BORDETELLA

APPAPKPAP
2255
AGS56996.1




PERTUSSIS









1373

BORDETELLA

APPGAGFIY
2256
1BCP_C




PERTUSSIS









1374

BORDETELLA

APQAAPLSI
2257
AGT50936.1




PERTUSSIS









1375

BORDETELLA

APRIENTAK
2258
WP_019249248.1




PERTUSSIS









1376

BORDETELLA

AQGKALLYR
2259
AGT50936.1




PERTUSSIS









1377

BORDETELLA

AQITSYVGF
2260
WP_019248658.1




PERTUSSIS









1378

BORDETELLA

AQLEVRGQR
2261
WP_019249248.1




PERTUSSIS









1379

BORDETELLA

AQQLKQADR
2262
WP_019247699.1




PERTUSSIS









1380

BORDETELLA

AQVTVAGRY
2263
WP_019249248.1




PERTUSSIS









1381

BORDETELLA

ARRSRVRAL
2264
NP_882284.1




PERTUSSIS









1382

BORDETELLA

ASPRRARRA
2265
WP_019249248.1




PERTUSSIS









1383

BORDETELLA

ASSPDAHVP
2266
AAA22983.1




PERTUSSIS









1384

BORDETELLA

ASVSNPGTF
2267
WP_019249248.1




PERTUSSIS









1385

BORDETELLA

ATWNFQSTY
2268
WP_019249248.1




PERTUSSIS









1386

BORDETELLA

ATYIADSGF
2269
AGS56996.1




PERTUSSIS









1387

BORDETELLA

AVAAPAVGA
2270
WP_019249248.1




PERTUSSIS









1388

BORDETELLA

AVFMQQRPL
2271
AAA22983.1




PERTUSSIS









1389

BORDETELLA

AVLVNPHIF
2272
WP_019249248.1




PERTUSSIS









1390

BORDETELLA

CFGKDLKRP
2273
AAA22983.1




PERTUSSIS









1391

BORDETELLA

CPSSLGNGV
2274
WP_019248145.1




PERTUSSIS









1392

BORDETELLA

DAGHEHDTW
2275
AAA22984.1




PERTUSSIS









1393

BORDETELLA

DAKHDLTVT
2276
WP_019249248.1




PERTUSSIS









1394

BORDETELLA

DASGQHRLW
2277
AGS56996.1




PERTUSSIS









1395

BORDETELLA

DATFETYAL
2278
YP_006628018.1




PERTUSSIS









1396

BORDETELLA

DATFQTYAL
2279
1BCP_C




PERTUSSIS









1397

BORDETELLA

DATLVGAKF
2280
WP_019247158.1




PERTUSSIS









1398

BORDETELLA

DDEVDVSGR
2281
AAA22974.1




PERTUSSIS









1399

BORDETELLA

DENGKPQTY
2282
WP_019247158.1




PERTUSSIS









1400

BORDETELLA

DGPPSRPTT
2283
WP_019247158.1




PERTUSSIS









1401

BORDETELLA

DHLTGRSCQ
2284
AAW72734.1




PERTUSSIS









1402

BORDETELLA

DNEGKMESN
2285
WP_019249248.1




PERTUSSIS









1403

BORDETELLA

DPPATVYRY
2286
AAW72734.1




PERTUSSIS









1404

BORDETELLA

EATEGDATL
2287
WP_019247158.1




PERTUSSIS









1405

BORDETELLA

EATQQAAGF
2288
WP_019248145.1




PERTUSSIS









1406

BORDETELLA

ECSGKQDCP
2289
AAA22983.1




PERTUSSIS









1407

BORDETELLA

EGGKLRGKD
2290
AAA22974.1




PERTUSSIS









1408

BORDETELLA

EGKMESNKD
2291
WP_019249248.1




PERTUSSIS









1409

BORDETELLA

EHRMQEAVE
2292
AAW72734.1




PERTUSSIS









1410

BORDETELLA

EKRLDIDDA
2293
WP_019249248.1




PERTUSSIS









1411

BORDETELLA

EPASANTLL
2294
AGS56996.1




PERTUSSIS









1412

BORDETELLA

EPQAELAVF
2295
AGS56996.1




PERTUSSIS









1413

BORDETELLA

EPVKLTLTG
2296
AGT50936.1




PERTUSSIS









1414

BORDETELLA

ESAEGRKIF
2297
WP_019249248.1




PERTUSSIS









1415

BORDETELLA

ESYSESHNF
2298
WP_019247158.1




PERTUSSIS









1416

BORDETELLA

ETFCITTIY
2299
1BCP_C




PERTUSSIS









1417

BORDETELLA

EVAGALELS
2300
WP_019249248.1




PERTUSSIS









1418

BORDETELLA

EVAKVEVVP
2301
WP_019247158.1




PERTUSSIS









1419

BORDETELLA

EVDGIIQEF
2302
WP_019249248.1




PERTUSSIS









1420

BORDETELLA

EVRADNNFY
2303
WP_019248344.1




PERTUSSIS









1421

BORDETELLA

FAILSSTTE
2304
WP_019247158.1




PERTUSSIS









1422

BORDETELLA

FAISAYALK
2305
AAA22984.1




PERTUSSIS









1423

BORDETELLA

FALYDGTYL
2306
AFK26303.1




PERTUSSIS









1424

BORDETELLA

FDTMLGFAI
2307
AAA22984.1




PERTUSSIS









1425

BORDETELLA

FEGKPALEL
2308
AAA22983.1




PERTUSSIS









1426

BORDETELLA

FELGADHAV
2309
AGS56996.1




PERTUSSIS









1427

BORDETELLA

FEPGITTNY
2310
WP_019248658.1




PERTUSSIS









1428

BORDETELLA

FETYALTGI
2311
YP_006628018.1




PERTUSSIS









1429

BORDETELLA

FIYRETFCI
2312
1BCP_C




PERTUSSIS









1430

BORDETELLA

FPTRTTAPG
2313
NP_882284.1




PERTUSSIS









1431

BORDETELLA

FQTYALTGI
2314
1BCP_C




PERTUSSIS









1432

BORDETELLA

FTHADGWFL
2315
AGS56996.1




PERTUSSIS









1433

BORDETELLA

FVRDGQSVI
2316
1BCP_C




PERTUSSIS









1434

BORDETELLA

FVRSGQPVI
2317
YP_006628018.1




PERTUSSIS









1435

BORDETELLA

FVWYVDTVI
2318
WP_019248866.1




PERTUSSIS









1436

BORDETELLA

GAASSRQAL
2319
WP_019249248.1




PERTUSSIS









1437

BORDETELLA

GAASSYFEY
2320
AAW72734.1




PERTUSSIS









1438

BORDETELLA

GAFDLKTTF
2321
AFK26303.1




PERTUSSIS









1439

BORDETELLA

GAPAAVSVL
2322
AGS56996.1




PERTUSSIS









1440

BORDETELLA

GATRAVDSL
2323
AGS56996.1




PERTUSSIS









1441

BORDETELLA

GAVPGGAVP
2324
AGT50936.1




PERTUSSIS









1442

BORDETELLA

GEAMVLVYY
2325
AFK26302.1




PERTUSSIS









1443

BORDETELLA

GEIALGDAT
2326
WP_019249248.1




PERTUSSIS









1444

BORDETELLA

GELMAAQVA
2327
WP_019247158.1




PERTUSSIS









1445

BORDETELLA

GGVPGGAVP
2328
AAZ74338.1




PERTUSSIS









1446

BORDETELLA

GHEHDTWFD
2329
AAA22984.1




PERTUSSIS









1447

BORDETELLA

GIGALKAGA
2330
WP_019249248.1




PERTUSSIS









1448

BORDETELLA

GIVIPPKAL
2331
1BCP_C




PERTUSSIS









1449

BORDETELLA

GKDLKRPGS
2332
AAA22983.1




PERTUSSIS









1450

BORDETELLA

GKLPKPVTV
2333
WP_019247158.1




PERTUSSIS









1451

BORDETELLA

GKSLKKKNQ
2334
WP_019247158.1




PERTUSSIS









1452

BORDETELLA

GLDVQQGTV
2335
WP_019249248.1




PERTUSSIS









1453

BORDETELLA

GLTDGVSRI
2336
WP_019249248.1




PERTUSSIS









1454

BORDETELLA

GLYPTYTEW
2337
WP_019249248.1




PERTUSSIS









1455

BORDETELLA

GLYQTYTEW
2338
YP_006626470.1




PERTUSSIS









1456

BORDETELLA

GPPSRPTTP
2339
WP_019247158.1




PERTUSSIS









1457

BORDETELLA

GPSAVAAPA
2340
WP_019249248.1




PERTUSSIS









1458

BORDETELLA

GVAPTAQQL
2341
WP_019248866.1




PERTUSSIS









1459

BORDETELLA

GVGLASTLW
2342
AGS56996.1




PERTUSSIS









1460

BORDETELLA

GYEAGFSLG
2343
WP_019247158.1




PERTUSSIS









1461

BORDETELLA

HADDGTIVI
2344
WP_019248145.1




PERTUSSIS









1462

BORDETELLA

HADWNNQSI
2345
AGS56996.1




PERTUSSIS









1463

BORDETELLA

HAEHEKDVR
2346
WP_019247158.1




PERTUSSIS









1464

BORDETELLA

HANHYGTRI
2347
WP_019247158.1




PERTUSSIS









1465

BORDETELLA

HAQGKALLY
2348
AGT50936.1




PERTUSSIS









1466

BORDETELLA

HFIGYIYEV
2349
AAW72734.1




PERTUSSIS









1467

BORDETELLA

HLSPALADV
2350
AAA22983.1




PERTUSSIS









1468

BORDETELLA

HSLYASYEY
2351
AGS56996.1




PERTUSSIS









1469

BORDETELLA

HVRGMLVPV
2352
AAA22974.1




PERTUSSIS









1470

BORDETELLA

HVSKEEQYY
2353
YP_006628018.1




PERTUSSIS









1471

BORDETELLA

HVTRGWSIF
2354
AFK26303.1




PERTUSSIS









1472

BORDETELLA

IADSGFYLD
2355
AGS56996.1




PERTUSSIS









1473

BORDETELLA

IAHRTELRG
2356
AGS56996.1




PERTUSSIS









1474

BORDETELLA

IENTAKLSG
2357
WP_019249248.1




PERTUSSIS









1475

BORDETELLA

IESKISQSV
2358
WP_019249248.1




PERTUSSIS









1476

BORDETELLA

IETGGARRF
2359
AGT50936.1




PERTUSSIS









1477

BORDETELLA

IIKDAPPGA
2360
1BCP_C




PERTUSSIS









1478

BORDETELLA

IIQEFAADL
2361
WP_019249248.1




PERTUSSIS









1479

BORDETELLA

ILAGALATY
2362
AAW72734.1




PERTUSSIS









1480

BORDETELLA

ILLENPAAE
2363
AGS56996.1




PERTUSSIS









1481

BORDETELLA

ILPILVLAL
2364
NP_882286.1




PERTUSSIS









1482

BORDETELLA

IPFQRALRL
2365
WP_019248145.1




PERTUSSIS









1483

BORDETELLA

ISVRVHVSK
2366
YP_006628018.1




PERTUSSIS









1484

BORDETELLA

ITNETGKTY
2367
WP_019247158.1




PERTUSSIS









1485

BORDETELLA

ITNKRAALI
2368
WP_019249248.1




PERTUSSIS









1486

BORDETELLA

ITSYVGFSV
2369
WP_019248658.1




PERTUSSIS









1487

BORDETELLA

ITVTSRGGF
2370
WP_019249248.1




PERTUSSIS









1488

BORDETELLA

IVIPPKALF
2371
1BCP_C




PERTUSSIS









1489

BORDETELLA

IVVEAGELV
2372
WP_019249248.1




PERTUSSIS









1490

BORDETELLA

KAAKSVNLM
2373
WP_019247158.1




PERTUSSIS









1491

BORDETELLA

KAAPLRRTT
2374
AGS56996.1




PERTUSSIS









1492

BORDETELLA

KAGKLSATG
2375
WP_019249248.1




PERTUSSIS









1493

BORDETELLA

KAGTIAAPW
2376
WP_019249248.1




PERTUSSIS









1494

BORDETELLA

KAKSLTTEI
2377
WP_019249248.1




PERTUSSIS









1495

BORDETELLA

KATVTTVQV
2378
WP_019247158.1




PERTUSSIS









1496

BORDETELLA

KDYRDKDGG
2379
WP_019247158.1




PERTUSSIS









1497

BORDETELLA

KEAATIVAA
2380
WP_019249248.1




PERTUSSIS









1498

BORDETELLA

KEDVDAAQI
2381
WP_019248658.1




PERTUSSIS









1499

BORDETELLA

KEVDGIIQE
2382
WP_019249248.1




PERTUSSIS









1500

BORDETELLA

KGPKLAMPW
2383
AGS56996.1




PERTUSSIS









1501

BORDETELLA

KLASGGGAV
2384
WP_019249248.1




PERTUSSIS









1502

BORDETELLA

KLKGKNQEF
2385
AAA22984.1




PERTUSSIS









1503

BORDETELLA

KLLHHILPI
2386
NP_882286.1




PERTUSSIS









1504

BORDETELLA

KPAPQPGPQ
2387
AGS56996.1




PERTUSSIS









1505

BORDETELLA

KPAPTAPPM
2388
WP_019249248.1




PERTUSSIS









1506

BORDETELLA

KPAVSVKVA
2389
WP_019249248.1




PERTUSSIS









1507

BORDETELLA

KPDRAARVA
2390
WP_019249248.1




PERTUSSIS









1508

BORDETELLA

KPLADIAVI
2391
YP_006626470.1




PERTUSSIS









1509

BORDETELLA

KPLADIAVV
2392
WP_019249248.1




PERTUSSIS









1510

BORDETELLA

KPLPKPLPV
2393
WP_019247158.1




PERTUSSIS









1511

BORDETELLA

KQADRDFVW
2394
WP_019247699.1




PERTUSSIS









1512

BORDETELLA

KSLPGGKLP
2395
WP_019247158.1




PERTUSSIS









1513

BORDETELLA

KSYTLRYLA
2396
WP_019248658.1




PERTUSSIS









1514

BORDETELLA

KTNMVVTSV
2397
AAA22983.1




PERTUSSIS









1515

BORDETELLA

KVLAPRLYL
2398
AAA22974.1




PERTUSSIS









1516

BORDETELLA

KVLSTKTTL
2399
WP_019247158.1




PERTUSSIS









1517

BORDETELLA

LAAGAGLTL
2400
WP_019249248.1




PERTUSSIS









1518

BORDETELLA

LAAIASAAH
2401
WP_019248145.1




PERTUSSIS









1519

BORDETELLA

LAANGNGQW
2402
AGS56996.1




PERTUSSIS









1520

BORDETELLA

LAAQVTQRG
2403
WP_019249248.1




PERTUSSIS









1521

BORDETELLA

LAARGDGAL
2404
AAA22974.1




PERTUSSIS









1522

BORDETELLA

LAAVLVNPH
2405
WP_019249248.1




PERTUSSIS









1523

BORDETELLA

LAGSGLFRM
2406
AGS56996.1




PERTUSSIS









1524

BORDETELLA

LAKALSAAL
2407
WP_019249248.1




PERTUSSIS









1525

BORDETELLA

LALQAGDAL
2408
WP_019249248.1




PERTUSSIS









1526

BORDETELLA

LAMPWTFHA
2409
AGS56996.1




PERTUSSIS









1527

BORDETELLA

LANDGTIVI
2410
WP_019248658.1




PERTUSSIS









1528

BORDETELLA

LAPTVGVAF
2411
WP_019247158.1




PERTUSSIS









1529

BORDETELLA

LASDGSVDF
2412
AGS56996.1




PERTUSSIS









1530

BORDETELLA

LATYQSEYL
2413
AAW72734.1




PERTUSSIS









1531

BORDETELLA

LAWALMLAC
2414
AAA22974.1




PERTUSSIS









1532

BORDETELLA

LEAGRRFTH
2415
AGS56996.1




PERTUSSIS









1533

BORDETELLA

LFTQQGGAY
2416
1BCP_C




PERTUSSIS









1534

BORDETELLA

LKRPGSSPM
2417
AAA22983.1




PERTUSSIS









1535

BORDETELLA

LLGSHVARA
2418
AFK26303.1




PERTUSSIS









1536

BORDETELLA

LLHHILPIL
2419
NP_882286.1




PERTUSSIS









1537

BORDETELLA

LLNAGGTLL
2420
WP_019249248.1




PERTUSSIS









1538

BORDETELLA

LMGPLQVNA
2421
WP_019249248.1




PERTUSSIS









1539

BORDETELLA

LNDSKITMG
2422
WP_019248145.1




PERTUSSIS









1540

BORDETELLA

LPEPVKLTL
2423
AGT50936.1




PERTUSSIS









1541

BORDETELLA

LPILVLALL
2424
NP_882286.1




PERTUSSIS









1542

BORDETELLA

LPKISKNAL
2425
WP_019248145.1




PERTUSSIS









1543

BORDETELLA

LPKPVTVKL
2426
WP_019247158.1




PERTUSSIS









1544

BORDETELLA

LPLKANPMH
2427
NP_882285.1




PERTUSSIS









1545

BORDETELLA

LPPRPVVAE
2428
WP_019247158.1




PERTUSSIS









1546

BORDETELLA

LPSIPGTSI
2429
AGS56996.1




PERTUSSIS









1547

BORDETELLA

LPTHLYKNF
2430
AAA22984.1




PERTUSSIS









1548

BORDETELLA

LPVRGVALV
2431
WP_019249248.1




PERTUSSIS









1549

BORDETELLA

LPVSLTALD
2432
WP_019249248.1




PERTUSSIS









1550

BORDETELLA

LQGGNKVPV
2433
WP_019249248.1




PERTUSSIS









1551

BORDETELLA

LSAALGADW
2434
WP_019249248.1




PERTUSSIS









1552

BORDETELLA

LSDAGHEHD
2435
AAA22984.1




PERTUSSIS









1553

BORDETELLA

LSGEVQRKG
2436
WP_019249248.1




PERTUSSIS









1554

BORDETELLA

LSSPSAITV
2437
WP_019249248.1




PERTUSSIS









1555

BORDETELLA

LTWLAILAV
2438
AAW72734.1




PERTUSSIS









1556

BORDETELLA

LVFSHVRGM
2439
AAA22974.1




PERTUSSIS









1557

BORDETELLA

LVSDAGADL
2440
WP_019249248.1




PERTUSSIS









1558

BORDETELLA

LVYYESIAY
2441
AFK26302.1




PERTUSSIS









1559

BORDETELLA

MAAESTFES
2442
WP_019247158.1




PERTUSSIS









1560

BORDETELLA

MAAGHDATL
2443
WP_019249248.1




PERTUSSIS









1561

BORDETELLA

MAAWSERAG
2444
AFK26302.1




PERTUSSIS









1562

BORDETELLA

MALGALGAA
2445
AGS56996.1




PERTUSSIS









1563

BORDETELLA

MAPVMGACM
2446
ADA85123.1




PERTUSSIS









1564

BORDETELLA

MATKGEMQI
2447
WP_019249248.1




PERTUSSIS









1565

BORDETELLA

MDAKGGTLL
2448
WP_019249248.1




PERTUSSIS









1566

BORDETELLA

MESNKDIVI
2449
WP_019249248.1




PERTUSSIS









1567

BORDETELLA

MEVMLRAVF
2450
AAA22983.1




PERTUSSIS









1568

BORDETELLA

MEYFKTPLP
2451
WP_019249248.1




PERTUSSIS









1569

BORDETELLA

MHTIASILL
2452
AAA22984.1




PERTUSSIS









1570

BORDETELLA

MIYMSGLAV
2453
1BCP_C




PERTUSSIS









1571

BORDETELLA

MLACTGLPL
2454
AAA22974.1




PERTUSSIS









1572

BORDETELLA

MPIDRKTLC
2455
AFK26303.1




PERTUSSIS









1573

BORDETELLA

MPKAPELDL
2456
WP_019249248.1




PERTUSSIS









1574

BORDETELLA

MQRQAGLPL
2457
NP_882285.1




PERTUSSIS









1575

BORDETELLA

NALAWALML
2458
AAA22974.1




PERTUSSIS









1576

BORDETELLA

NITNKRAAL
2459
WP_019249248.1




PERTUSSIS









1577

BORDETELLA

NLMAAESTF
2460
WP_019247158.1




PERTUSSIS









1578

BORDETELLA

NNETMSGRQ
2461
WP_019249248.1




PERTUSSIS









1579

BORDETELLA

NPGSLIAEV
2462
WP_019249248.1




PERTUSSIS









1580

BORDETELLA

NPMHTIASI
2463
NP_882285.1




PERTUSSIS









1581

BORDETELLA

NPQTQLSNI
2464
WP_019248145.1




PERTUSSIS









1582

BORDETELLA

NPYTSRRSV
2465
AFK26302.1




PERTUSSIS









1583

BORDETELLA

PAPTAPPMP
2466
WP_019249248.1




PERTUSSIS









1584

BORDETELLA

PASANTLLL
2467
AGS56996.1




PERTUSSIS









1585

BORDETELLA

PAVALPRPL
2468
WP_019249248.1




PERTUSSIS









1586

BORDETELLA

PDAHVPFCF
2469
AAA22983.1




PERTUSSIS









1587

BORDETELLA

PELGAAIRV
2470
AGT50936.1




PERTUSSIS









1588

BORDETELLA

PFIIKLKDC
2471
WP_019248145.1




PERTUSSIS









1589

BORDETELLA

PGPQPPQPP
2472
AGS56996.1




PERTUSSIS









1590

BORDETELLA

PGPQPPQPQ
2473
AAZ74338.1




PERTUSSIS









1591

BORDETELLA

PGPTTDYST
2474
WP_019248145.1




PERTUSSIS









1592

BORDETELLA

PGTFTAGKD
2475
WP_019249248.1




PERTUSSIS









1593

BORDETELLA

PGTPGDLLE
2476
AAA22984.1




PERTUSSIS









1594

BORDETELLA

PKPKPKAER
2477
WP_019247158.1




PERTUSSIS









1595

BORDETELLA

PKPKPKPKA
2478
WP_019247158.1




PERTUSSIS









1596

BORDETELLA

PKPKPKPKP
2479
WP_019247158.1




PERTUSSIS









1597

BORDETELLA

PLPPRPVVA
2480
WP_019247158.1




PERTUSSIS









1598

BORDETELLA

PPAPKPAPQ
2481
AGS56996.1




PERTUSSIS









1599

BORDETELLA

PPKPAPVAK
2482
WP_019247158.1




PERTUSSIS









1600

BORDETELLA

PPRPVAAQV
2483
WP_019247158.1




PERTUSSIS









1601

BORDETELLA

PPRPVVAEK
2484
WP_019247158.1




PERTUSSIS









1602

BORDETELLA

PPSRPTTPP
2485
WP_019247158.1




PERTUSSIS









1603

BORDETELLA

PRRARRALR
2486
WP_019249248.1




PERTUSSIS









1604

BORDETELLA

QAAPLSITL
2487
AGT50936.1




PERTUSSIS









1605

BORDETELLA

QADRDFVWY
2488
WP_019247699.1




PERTUSSIS









1606

BORDETELLA

QAIVVGKDL
2489
WP_019249248.1




PERTUSSIS









1607

BORDETELLA

QALGALKLY
2490
ACI16088.1




PERTUSSIS









1608

BORDETELLA

QELALKLKG
2491
AAA22984.1




PERTUSSIS









1609

BORDETELLA

QITQHGGPY
2492
NP_882283.1




PERTUSSIS









1610

BORDETELLA

QITQHGSPY
2493
AFK26303.1




PERTUSSIS









1611

BORDETELLA

QPLPPRPVA
2494
WP_019247158.1




PERTUSSIS









1612

BORDETELLA

QPPAGRELS
2495
AGS56996.1




PERTUSSIS









1613

BORDETELLA

QQLKQADRD
2496
WP_019247699.1




PERTUSSIS









1614

BORDETELLA

QQVQVLQRQ
2497
WP_019247158.1




PERTUSSIS









1615

BORDETELLA

QSIVEAPEL
2498
AGT50936.1




PERTUSSIS









1616

BORDETELLA

QVGSSNSAF
2499
AAW72734.1




PERTUSSIS









1617

BORDETELLA

RAGLSPATW
2500
WP_019249248.1




PERTUSSIS









1618

BORDETELLA

RARRALRQD
2501
WP_019249248.1




PERTUSSIS









1619

BORDETELLA

RASASRARI
2502
WP_019249248.1




PERTUSSIS









1620

BORDETELLA

RELSAAANA
2503
AGS56996.1




PERTUSSIS









1621

BORDETELLA

RETFCITTI
2504
1BCP_C




PERTUSSIS









1622

BORDETELLA

RGFAQRQQL
2505
AGS56996.1




PERTUSSIS









1623

BORDETELLA

RGSAATFTL
2506
WP_019248295.1




PERTUSSIS









1624

BORDETELLA

RGWSIFALY
2507
AFK26303.1




PERTUSSIS









1625

BORDETELLA

RKMLYLIYV
2508
YP_006628018.1




PERTUSSIS









1626

BORDETELLA

RLRKMLYLI
2509
YP_006628018.1




PERTUSSIS









1627

BORDETELLA

RPQITDAVT
2510
WP_019249248.1




PERTUSSIS









1628

BORDETELLA

RPSVNGGRI
2511
WP_019249248.1




PERTUSSIS









1629

BORDETELLA

RRFTHADGW
2512
AGS56996.1




PERTUSSIS









1630

BORDETELLA

RSGARATSL
2513
AAA22974.1




PERTUSSIS









1631

BORDETELLA

RSRVRALAW
2514
NP_882284.1




PERTUSSIS









1632

BORDETELLA

RSRVRALSW
2515
YP_006628019.1




PERTUSSIS









1633

BORDETELLA

RTHGVGASL
2516
AGS56996.1




PERTUSSIS









1634

BORDETELLA

RTRGQARSG
2517
AAA22974.1




PERTUSSIS









1635

BORDETELLA

RVAPPAVAL
2518
WP_019249248.1




PERTUSSIS









1636

BORDETELLA

RVLPEPVKL
2519
AGT50936.1




PERTUSSIS









1637

BORDETELLA

RVRALAWLL
2520
NP_882284.1




PERTUSSIS









1638

BORDETELLA

RVRALSWLL
2521
YP_006628019.1




PERTUSSIS









1639

BORDETELLA

RVTVSGGSL
2522
AGT50936.1




PERTUSSIS









1640

BORDETELLA

SEAMAAWSE
2523
AFK26302.1




PERTUSSIS









1641

BORDETELLA

SESHNFHAS
2524
WP_019247158.1




PERTUSSIS









1642

BORDETELLA

SGEGRVNIG
2525
WP_019249248.1




PERTUSSIS









1643

BORDETELLA

SGLAVRVHV
2526
1BCP_C




PERTUSSIS









1644

BORDETELLA

SLADISLGA
2527
WP_019249248.1




PERTUSSIS









1645

BORDETELLA

SLFAILSST
2528
WP_019247158.1




PERTUSSIS









1646

BORDETELLA

SLFAPHGNV
2529
AAZ74338.1




PERTUSSIS









1647

BORDETELLA

SLSIDNATW
2530
AGS56996.1




PERTUSSIS









1648

BORDETELLA

SPMEVMLRA
2531
AAA22983.1




PERTUSSIS









1649

BORDETELLA

SPQPIRATV
2532
WP_019247158.1




PERTUSSIS









1650

BORDETELLA

SPRRARRAL
2533
WP_019249248.1




PERTUSSIS









1651

BORDETELLA

SPSRLAGTL
2534
WP_019249248.1




PERTUSSIS









1652

BORDETELLA

SSTPLGSLF
2535
WP_019247158.1




PERTUSSIS









1653

BORDETELLA

STYELLDYL
2536
WP_019249248.1




PERTUSSIS









1654

BORDETELLA

SVAMKPYEV
2537
AAA22983.1




PERTUSSIS









1655

BORDETELLA

SVAPNALAW
2538
AAA22974.1




PERTUSSIS









1656

BORDETELLA

SVKVAKKLF
2539
WP_019249248.1




PERTUSSIS









1657

BORDETELLA

TAFMSGRSL
2540
AAA22984.1




PERTUSSIS









1658

BORDETELLA

TAGATPFDI
2541
WP_019248658.1




PERTUSSIS









1659

BORDETELLA

TAPVTSPAW
2542
AAW72734.1




PERTUSSIS









1660

BORDETELLA

TARTGWLTW
2543
AAW72734.1




PERTUSSIS









1661

BORDETELLA

TEAQGVQVR
2544
WP_019248145.1




PERTUSSIS









1662

BORDETELLA

TEVYLEHRM
2545
AAW72734.1




PERTUSSIS









1663

BORDETELLA

TFEGKPALE
2546
AAA22983.1




PERTUSSIS









1664

BORDETELLA

TFTGKVTNG
2547
WP_019248658.1




PERTUSSIS









1665

BORDETELLA

TGDGGGHTD
2548
AGS56996.1




PERTUSSIS









1666

BORDETELLA

TLAKALSAA
2549
WP_019249248.1




PERTUSSIS









1667

BORDETELLA

TLANVGDTW
2550
AGS56996.1




PERTUSSIS









1668

BORDETELLA

TLNASNLTL
2551
WP_019249248.1




PERTUSSIS









1669

BORDETELLA

TLSSAHGNV
2552
WP_019249248.1




PERTUSSIS









1670

BORDETELLA

TPFDIKLKE
2553
WP_019248658.1




PERTUSSIS









1671

BORDETELLA

TPFIIKLKD
2554
WP_019248145.1




PERTUSSIS









1672

BORDETELLA

TPGWSIYGL
2555
1BCP_C




PERTUSSIS









1673

BORDETELLA

TPLGSAATF
2556
AGS56996.1




PERTUSSIS









1674

BORDETELLA

TPLGSLFAI
2557
WP_019247158.1




PERTUSSIS









1675

BORDETELLA

TPLPVSLTA
2558
WP_019249248.1




PERTUSSIS









1676

BORDETELLA

TRQGIMDQY
2559
YP_006626873.1




PERTUSSIS









1677

BORDETELLA

TSKQDERNY
2560
WP_019247158.1




PERTUSSIS









1678

BORDETELLA

TSPYDGKYW
2561
YP_006628018.1




PERTUSSIS









1679

BORDETELLA

TSRRSVASI
2562
AFK26302.1




PERTUSSIS









1680

BORDETELLA

TSRTVTMRY
2563
NP_879898.1




PERTUSSIS









1681

BORDETELLA

TTEYPNARY
2564
ADA85123.1




PERTUSSIS









1682

BORDETELLA

TTEYSNARY
2565
AFK26302.1




PERTUSSIS









1683

BORDETELLA

TTLGLEQTF
2566
WP_019247158.1




PERTUSSIS









1684

BORDETELLA

TVLAAGAGL
2567
WP_019249248.1




PERTUSSIS









1685

BORDETELLA

TVQELALKL
2568
AAA22984.1




PERTUSSIS









1686

BORDETELLA

TVVQRNKHW
2569
WP_019247158.1




PERTUSSIS









1687

BORDETELLA

VAAAADLAL
2570
WP_019249248.1




PERTUSSIS









1688

BORDETELLA

VALASQARW
2571
AGS56996.1




PERTUSSIS









1689

BORDETELLA

VAMKPYEVT
2572
AAA22983.1




PERTUSSIS









1690

BORDETELLA

VARLVKLQG
2573
WP_019249248.1




PERTUSSIS









1691

BORDETELLA

VAVAGGRWH
2574
AGS56996.1




PERTUSSIS









1692

BORDETELLA

VEASAITTY
2575
WP_019248145.1




PERTUSSIS









1693

BORDETELLA

VEDIGGKNY
2576
WP_019247158.1




PERTUSSIS









1694

BORDETELLA

VEVSSPPPV
2577
WP_019247158.1




PERTUSSIS









1695

BORDETELLA

VGAGGYEAG
2578
WP_019247158.1




PERTUSSIS









1696

BORDETELLA

VGGGEHGRW
2579
WP_019249248.1




PERTUSSIS









1697

BORDETELLA

VHVSKEEQY
2580
YP_006628018.1




PERTUSSIS









1698

BORDETELLA

VIDGQKVLA
2581
AAA22974.1




PERTUSSIS









1699

BORDETELLA

VIGACTSPY
2582
YP_006628018.1




PERTUSSIS









1700

BORDETELLA

VKLGGVYEA
2583
AAA22974.1




PERTUSSIS









1701

BORDETELLA

VLAPRLYLT
2584
AAA22974.1




PERTUSSIS









1702

BORDETELLA

VLVKTNMVV
2585
AAA22983.1




PERTUSSIS









1703

BORDETELLA

VPASGAPAA
2586
AGS56996.1




PERTUSSIS









1704

BORDETELLA

VPFCFGKDL
2587
AAA22983.1




PERTUSSIS









1705

BORDETELLA

VPVSEHCTV
2588
AAA22974.1




PERTUSSIS









1706

BORDETELLA

VPVTPPKVE
2589
WP_019247158.1




PERTUSSIS









1707

BORDETELLA

VRTVSAMEY
2590
WP_019249248.1




PERTUSSIS









1708

BORDETELLA

VSGGSLSAP
2591
AGT50936.1




PERTUSSIS









1709

BORDETELLA

VSSATKAKG
2592
WP_019248658.1




PERTUSSIS









1710

BORDETELLA

VSSPPPVSV
2593
WP_019247158.1




PERTUSSIS









1711

BORDETELLA

VSVKVAKKL
2594
WP_019249248.1




PERTUSSIS









1712

BORDETELLA

VTMRYLASY
2595
WP_019248145.1




PERTUSSIS









1713

BORDETELLA

VTSVAMKPY
2596
AAA22983.1




PERTUSSIS









1714

BORDETELLA

VVAEKVTTP
2597
WP_019247158.1




PERTUSSIS









1715

BORDETELLA

VVDGPPSRP
2598
WP_019247158.1




PERTUSSIS









1716

BORDETELLA

VVETAQPLP
2599
WP_019247158.1




PERTUSSIS









1717

BORDETELLA

WLTWLAILA
2600
AAW72734.1




PERTUSSIS









1718

BORDETELLA

WTFHAGYRY
2601
AGS56996.1




PERTUSSIS









1719

BORDETELLA

WVMTDNSNV
2602
AGS56996.1




PERTUSSIS









1720

BORDETELLA

YAEHGEVSI
2603
WP_019249248.1




PERTUSSIS









1721

BORDETELLA

YAIDGTAAG
2604
WP_019249248.1




PERTUSSIS









1722

BORDETELLA

YALKSRIAL
2605
AAA22984.1




PERTUSSIS









1723

BORDETELLA

YATNPQTQL
2606
WP_019248145.1




PERTUSSIS









1724

BORDETELLA

YDTGDLIAY
2607
WP_019248658.1




PERTUSSIS









1725

BORDETELLA

YEAGFSLGS
2608
WP_019247158.1




PERTUSSIS









1726

BORDETELLA

YEDATFETY
2609
YP_006628018.1




PERTUSSIS









1727

BORDETELLA

YENKSSTPL
2610
WP_019247158.1




PERTUSSIS









1728

BORDETELLA

YEVTPTRML
2611
AAA22983.1




PERTUSSIS









1729

BORDETELLA

YEYIWGLYP
2612
WP_019249248.1




PERTUSSIS









1730

BORDETELLA

YEYIWGLYQ
2613
YP_006626470.1




PERTUSSIS









1731

BORDETELLA

YEYSKGPKL
2614
AGS56996.1




PERTUSSIS









1732

BORDETELLA

YFEPGPTTD
2615
WP_019248145.1




PERTUSSIS









1733

BORDETELLA

YIWGLYPTY
2616
WP_019249248.1




PERTUSSIS









1734

BORDETELLA

YIWGLYQTY
2617
YP_006626470.1




PERTUSSIS









1735

BORDETELLA

YLRQITPGW
2618
1BCP_C




PERTUSSIS









1736

BORDETELLA

YMIYMSGLA
2619
1BCP_C




PERTUSSIS









1737

BORDETELLA

YPALRAALI
2620
WP_019248658.1




PERTUSSIS









1738

BORDETELLA

YPGTPGDLL
2621
AAA22984.1




PERTUSSIS









1739

BORDETELLA

YPTYTEWSV
2622
WP_019249248.1




PERTUSSIS









1740

BORDETELLA

YQTYTEWSV
2623
YP_006626470.1




PERTUSSIS









1741

BORDETELLA

YSTGDLRAY
2624
WP_019248145.1




PERTUSSIS









1742

BORDETELLA

YTLRYLASY
2625
WP_019248658.1




PERTUSSIS









1743

BORDETELLA

YVLVKTNMV
2626
AAA22983.1




PERTUSSIS









1744

BORDETELLA

YYDYEDATF
2627
YP_006628018.1




PERTUSSIS









1745

BORDETELLA

AAFIALYPNSQLAPT
2628
Q7VU05




PERTUSSIS 509









1746

BORDETELLA

GGAEYNLALGQRRA
2629
Q7VU04




PERTUSSIS 509









1747

BORDETELLA

GGAEYNLALGQRRADA
2630
Q7VU04




PERTUSSIS 509









1748

BORDETELLA

IALYPNSQLAPT
2631
Q7VU05




PERTUSSIS 509









1749

BORDETELLA

KPDQGEVVAVGPGKKTED
2632
POA339.1




PERTUSSIS 509









1750

BORDETELLA

KPDQGEVVAVGPGKKTEDG
2633
POA339.1




PERTUSSIS 509









1751

BORDETELLA

LAEVLDYHNFVLTQ
2634
Q7VWM1.1




PERTUSSIS 509









1752

CORYNEBACTERIUM

QSIALSSLMVAQAIP
2635
AAV70486.1




DIPHTHERIAE









1753

CORYNEBACTERIUM

SIGVLGYQKTVDHTKVNSKLSLF
2636
AAV70486.1




DIPHTHERIAE









1754

BORDETELLA

AAHADWNNQSIVKT
2637
ABO77783.1




PERTUSSIS









1755

BORDETELLA

AALGRG
2638
ABO77783.1




PERTUSSIS









1756

BORDETELLA

AALGRGHSLYASYE
2639
ABO77783.1




PERTUSSIS









1757

BORDETELLA

AAPLRRTTLAMALG
2640
ABO77783.1




PERTUSSIS









1758

BORDETELLA

AAPLSITLQAGAHA
2641
ABO77783.1




PERTUSSIS









1759

BORDETELLA

ADAQGDIVATELPS
2642
ABO77783.1




PERTUSSIS









1760

BORDETELLA

ADSGFYLDATLRAS
2643
ABO77783.1




PERTUSSIS









1761

BORDETELLA

AELA
2644
ABO77783.1




PERTUSSIS









1762

BORDETELLA

AELAVFRAGGGAYR
2645
ABO77783.1




PERTUSSIS









1763

BORDETELLA

AELQFRNGSVTSSG
2646
ABO77783.1




PERTUSSIS









1764

BORDETELLA

AGGRWHLGGLAGYT
2647
ABO77783.1




PERTUSSIS









1765

BORDETELLA

AGVAAMQGAVVHLQ
2648
ABO77783.1




PERTUSSIS









1766

BORDETELLA

AGYTRGDRGFTGDG
2649
ABO77783.1




PERTUSSIS









1767

BORDETELLA

ALASQARWTGATRA
2650
ABO77783.1




PERTUSSIS









1768

BORDETELLA

AMPWTFHAGYRYSW
2651
ABO77783.1




PERTUSSIS









1769

BORDETELLA

AMQGAVVHLQRATIRRGDAP
2652
ABO77783.1




PERTUSSIS









1770

BORDETELLA

ANGLRVRDE
2653
ABO77783.1




PERTUSSIS









1771

BORDETELLA

ANGLRVRDEGGSSV
2654
ABO77783.1




PERTUSSIS









1772

BORDETELLA

ANKDGKVDIGTYRY
2655
ABO77783.1




PERTUSSIS









1773

BORDETELLA

APAAVSVLGASELT
2656
ABO77783.1




PERTUSSIS









1774

BORDETELLA

APPAPKPAPQPGPQ
2657
ABO77783.1




PERTUSSIS









1775

BORDETELLA

AQGILLENPAAELQ
2658
ABO77783.1




PERTUSSIS









1776

BORDETELLA

ARWTGATRAVDSLS
2659
ABO77783.1




PERTUSSIS









1777

BORDETELLA

ASLEAGRRFTHADG
2660
ABO77783.1




PERTUSSIS









1778

BORDETELLA

ASYEYSKGPKLAMP
2661
ABO77783.1




PERTUSSIS









1779

BORDETELLA

ATFTLANKD
2662
ABO77783.1




PERTUSSIS









1780

BORDETELLA

ATFTLANKDGKVDI
2663
ABO77783.1




PERTUSSIS









1781

BORDETELLA

ATRAVDSLSIDNAT
2664
ABO77783.1




PERTUSSIS









1782

BORDETELLA

DDDGIALYVAGEQAQ
2665
ABO77783.1




PERTUSSIS









1783

BORDETELLA

DDGIALYVAGEQAQ
2666
ABO77783.1




PERTUSSIS









1784

BORDETELLA

DGGHITGGRAAGVA
2667
ABO77783.1




PERTUSSIS









1785

BORDETELLA

DGIRRFLGTVTVKAGK
2668
ABO77783.1




PERTUSSIS









1786

BORDETELLA

DGSVDFQQPAEAGR
2669
ABO77783.1




PERTUSSIS









1787

BORDETELLA

DGYAVKGKYRTHGV
2670
ABO77783.1




PERTUSSIS









1788

BORDETELLA

DIVATELPSIPGTS
2671
ABO77783.1




PERTUSSIS









1789

BORDETELLA

DKLVVMQDASGQHR
2672
ABO77783.1




PERTUSSIS









1790

BORDETELLA

DLGLSDKLVVMQDA
2673
ABO77783.1




PERTUSSIS









1791

BORDETELLA

DNATWVMTDNSNVGA
2674
ABO77783.1




PERTUSSIS









1792

BORDETELLA

DNATWVMTDNSNVGALRLA
2675
ABO77783.1




PERTUSSIS









1793

BORDETELLA

DNRAGRRFDQKVAG
2676
ABO77783.1




PERTUSSIS









1794

BORDETELLA

EAGRFKVLTVNTLA
2677
ABO77783.1




PERTUSSIS









1795

BORDETELLA

ELAQSIVEAPELGA
2678
ABO77783.1




PERTUSSIS









1796

BORDETELLA

ELGAAIRVGRGARV
2679
ABO77783.1




PERTUSSIS









1797

BORDETELLA

ELGADHAVAVAGGR
2680
ABO77783.1




PERTUSSIS









1798

BORDETELLA

ELPSIPGTSIGPLD
2681
ABO77783.1




PERTUSSIS









1799

BORDETELLA

EPVKLTLTGGADAQ
2682
ABO77783.1




PERTUSSIS









1800

BORDETELLA

EQAQASIADSTLQG
2683
ABO77783.1




PERTUSSIS









1801

BORDETELLA

ERGANVTVQRSAIV
2684
ABO77783.1




PERTUSSIS









1802

BORDETELLA

ERQHGIHIQGSDPG
2685
ABO77783.1




PERTUSSIS









1803

BORDETELLA

EVGKRIELAGGRQV
2686
ABO77783.1




PERTUSSIS









1804

BORDETELLA

FDGAGTVHTNGIAH
2687
ABO77783.1




PERTUSSIS









1805

BORDETELLA

FQQPAEAGRFKVLT
2688
ABO77783.1




PERTUSSIS









1806

BORDETELLA

FRAGGGAYRAANGL
2689
ABO77783.1




PERTUSSIS









1807

BORDETELLA

GAHAQGKALLYRVL
2690
ABO77783.1




PERTUSSIS









1808

BORDETELLA

GARVTVSGGSLSAP
2691
ABO77783.1




PERTUSSIS









1809

BORDETELLA

GAYRAANGLRVRDE
2692
ABO77783.1




PERTUSSIS









1810

BORDETELLA

GDAPAGGAVPGGAV
2693
ABO77783.1




PERTUSSIS









1811

BORDETELLA

GGAVPGGAVPGGFG
2694
ABO77783.1




PERTUSSIS









1812

BORDETELLA

GGAVPGGFGPVLDG
2695
ABO77783.1




PERTUSSIS









1813

BORDETELLA

GGFGPVLDGWYGVD
2696
ABO77783.1




PERTUSSIS









1814

BORDETELLA

GGLHIGALQSLQPE
2697
ABO77783.1




PERTUSSIS









1815

BORDETELLA

GGVQIERGANVTVQ
2698
ABO77783.1




PERTUSSIS









1816

BORDETELLA

GHSLYASYEYSKGP
2699
ABO77783.1




PERTUSSIS









1817

BORDETELLA

GHTDSVHVGGYATY
2700
ABO77783.1




PERTUSSIS









1818

BORDETELLA

GIAHRTELRGTRAE
2701
ABO77783.1




PERTUSSIS









1819

BORDETELLA

GKALLYRVLPEPVK
2702
ABO77783.1




PERTUSSIS









1820

BORDETELLA

GLGMAAALGRGHSL
2703
ABO77783.1




PERTUSSIS









1821

BORDETELLA

GNVIETGGARRFAP
2704
ABO77783.1




PERTUSSIS









1822

BORDETELLA

GPLDVALASQARWT
2705
ABO77783.1




PERTUSSIS









1823

BORDETELLA

GQHRLWVRN
2706
ABO77783.1




PERTUSSIS









1824

BORDETELLA

GRGFAQRQQLDNRA
2707
ABO77783.1




PERTUSSIS









1825

BORDETELLA

GRLGLEVGKRIELA
2708
ABO77783.1




PERTUSSIS









1826

BORDETELLA

GRQVQPYIKASVLQ
2709
ABO77783.1




PERTUSSIS









1827

BORDETELLA

GRRFTHADGWFLEPQAELA
2710
ABO77783.1




PERTUSSIS









1828

BORDETELLA

GSEPASANTLLLVQ
2711
ABO77783.1




PERTUSSIS









1829

BORDETELLA

GSSVLGRLGLEVGK
2712
ABO77783.1




PERTUSSIS









1830

BORDETELLA

GTTIKVSGRQAQGI
2713
ABO77783.1




PERTUSSIS









1831

BORDETELLA

GTVTVKAGKLVADH
2714
ABO77783.1




PERTUSSIS









1832

BORDETELLA

HAVAVAGGRWHLGG
2715
ABO77783.1




PERTUSSIS









1833

BORDETELLA

IELAGGRQVQPYIK
2716
ABO77783.1




PERTUSSIS









1834

BORDETELLA

IHIQGSDPGGVRTA
2717
ABO77783.1




PERTUSSIS









1835

BORDETELLA

IRRFLGTVTVKAGK
2718
ABO77783.1




PERTUSSIS









1836

BORDETELLA

IRVGRGARVTVSGG
2719
ABO77783.1




PERTUSSIS









1837

BORDETELLA

ITLQAGAHA
2720
ABO77783.1




PERTUSSIS









1838

BORDETELLA

ITLQAGAHAQGKAL
2721
ABO77783.1




PERTUSSIS









1839

BORDETELLA

IVEAPELGAAIRVG
2722
ABO77783.1




PERTUSSIS









1840

BORDETELLA

IVKTGERQHGIHIQ
2723
ABO77783.1




PERTUSSIS









1841

BORDETELLA

KAGKLVADHATLAN
2724
ABO77783.1




PERTUSSIS









1842

BORDETELLA

KGKYRTHGVGASLE
2725
ABO77783.1




PERTUSSIS









1843

BORDETELLA

KPAPQPGPQPPQPP
2726
ABO77783.1




PERTUSSIS









1844

BORDETELLA

KVAGFELGADHAVA
2727
ABO77783.1




PERTUSSIS









1845

BORDETELLA

KVAGSDGYAVKGKY
2728
ABO77783.1




PERTUSSIS









1846

BORDETELLA

KVDIGTYRYRLAAN
2729
ABO77783.1




PERTUSSIS









1847

BORDETELLA

KVLTVNTLAGSGLF
2730
ABO77783.1




PERTUSSIS









1848

BORDETELLA

LAANGNGQWSLVGA
2731
ABO77783.1




PERTUSSIS









1849

BORDETELLA

LAMPWTFHAGYRYS
2732
ABO77783.1




PERTUSSIS









1850

BORDETELLA

LASTLWYAESNALS
2733
ABO77783.1




PERTUSSIS









1851

BORDETELLA

LENDFKVAGSDGYA
2734
ABO77783.1




PERTUSSIS









1852

BORDETELLA

LENPAAELQFRNGS
2735
ABO77783.1




PERTUSSIS









1853

BORDETELLA

LGAAPAAHADWNNQ
2736
ABO77783.1




PERTUSSIS









1854

BORDETELLA

LGGLAGYTRGDRGFTGDG
2737
ABO77783.1




PERTUSSIS









1855

BORDETELLA

LLENP
2738
ABO77783.1




PERTUSSIS









1856

BORDETELLA

LLVQTPLGSAATFT
2739
ABO77783.1




PERTUSSIS









1857

BORDETELLA

LPPSRVVLRDTNVT
2740
ABO77783.1




PERTUSSIS









1858

BORDETELLA

LQPEDLPPS
2741
ABO77783.1




PERTUSSIS









1859

BORDETELLA

LQPEDLPPSRVVLR
2742
ABO77783.1




PERTUSSIS









1860

BORDETELLA

LRASRLENDFKVAG
2743
ABO77783.1




PERTUSSIS









1861

BORDETELLA

LRLASDGSVDFQQP
2744
ABO77783.1




PERTUSSIS









1862

BORDETELLA

LSAAANAAVNTGGV
2745
ABO77783.1




PERTUSSIS









1863

BORDETELLA

LSAPHGNVIETGGA
2746
ABO77783.1




PERTUSSIS









1864

BORDETELLA

LSDDGIRRFLGTVT
2747
ABO77783.1




PERTUSSIS









1865

BORDETELLA

LVGAKAPPAPKPAP
2748
ABO77783.1




PERTUSSIS









1866

BORDETELLA

LYVAGEQAQASIAD
2749
ABO77783.1




PERTUSSIS









1867

BORDETELLA

MALGALGAAPAAHA
2750
ABO77783.1




PERTUSSIS









1868

BORDETELLA

MNMSLSRIVKAAPL
2751
ABO77783.1




PERTUSSIS









1869

BORDETELLA

MQDASGQHR
2752
ABO77783.1




PERTUSSIS









1870

BORDETELLA

MQGAVVHLQRATIR
2753
ABO77783.1




PERTUSSIS









1871

BORDETELLA

NAAVNTGGVGLAST
2754
ABO77783.1




PERTUSSIS









1872

BORDETELLA

NALSKRLGELRLNP
2755
ABO77783.1




PERTUSSIS









1873

BORDETELLA

NGQWSLVGAKAPPA
2756
ABO77783.1




PERTUSSIS









1874

BORDETELLA

NTLAGSGLFRMNVF
2757
ABO77783.1




PERTUSSIS









1875

BORDETELLA

PAGRELSAAANAAV
2758
ABO77783.1




PERTUSSIS









1876

BORDETELLA

PAPQPPAGRELSAA
2759
ABO77783.1




PERTUSSIS









1877

BORDETELLA

PGPQPPQPPQPQPE
2760
ABO77783.1




PERTUSSIS









1878

BORDETELLA

PGTSIGPLDVALAS
2761
ABO77783.1




PERTUSSIS









1879

BORDETELLA

PLGSAATFTLANKD
2762
ABO77783.1




PERTUSSIS









1880

BORDETELLA

PQPEAPAPQPPAGR
2763
ABO77783.1




PERTUSSIS









1881

BORDETELLA

PQPPQPQPEAPAPQ
2764
ABO77783.1




PERTUSSIS









1882

BORDETELLA

PYIKASVLQEFDGA
2765
ABO77783.1




PERTUSSIS









1883

BORDETELLA

RFAPQAAPLSITLQ
2766
ABO77783.1




PERTUSSIS









1884

BORDETELLA

RLGELRLNPDAGGA
2767
ABO77783.1




PERTUSSIS









1885

BORDETELLA

RLNPDAGGAWGRGF
2768
ABO77783.1




PERTUSSIS









1886

BORDETELLA

RNGSVTSSGQLSDD
2769
ABO77783.1




PERTUSSIS









1887

BORDETELLA

RRFDQKVAGFELGA
2770
ABO77783.1




PERTUSSIS









1888

BORDETELLA

RTTLAMALGALGAA
2771
ABO77783.1




PERTUSSIS









1889

BORDETELLA

SAIVDGGLHIGALQ
2772
ABO77783.1




PERTUSSIS









1890

BORDETELLA

SANTLLLVQTPLGS
2773
ABO77783.1




PERTUSSIS









1891

BORDETELLA

SDPGGVRTASGTTI
2774
ABO77783.1




PERTUSSIS









1892

BORDETELLA

SELTLDGGHITGGR
2775
ABO77783.1




PERTUSSIS









1893

BORDETELLA

SGLFRMNVF
2776
ABO77783.1




PERTUSSIS









1894

BORDETELLA

SGLFRMNVFADLGL
2777
ABO77783.1




PERTUSSIS









1895

BORDETELLA

SGSSVELAQSIVEA
2778
ABO77783.1




PERTUSSIS









1896

BORDETELLA

SIADSTLQGAGGVQ
2779
ABO77783.1




PERTUSSIS









1897

BORDETELLA

SKGPKLAMPWTFHA
2780
ABO77783.1




PERTUSSIS









1898

BORDETELLA

SNVGALRLASDGSV
2781
ABO77783.1




PERTUSSIS









1899

BORDETELLA

SRIVKAAPLRRTTL
2782
ABO77783.1




PERTUSSIS









1900

BORDETELLA

SVLGASELTLDGGH
2783
ABO77783.1




PERTUSSIS









1901

BORDETELLA

SVLQEFDGA
2784
ABO77783.1




PERTUSSIS









1902

BORDETELLA

SVLQEFDGAGTVHT
2785
ABO77783.1




PERTUSSIS









1903

BORDETELLA

TELR
2786
ABO77783.1




PERTUSSIS









1904

BORDETELLA

TELRGTRAELGLGM
2787
ABO77783.1




PERTUSSIS









1905

BORDETELLA

TGDGGGHTDSVHVG
2788
ABO77783.1




PERTUSSIS









1906

BORDETELLA

TGGARRFAPQAAPL
2789
ABO77783.1




PERTUSSIS









1907

BORDETELLA

TGGRAAGVAAMQGA
2790
ABO77783.1




PERTUSSIS









1908

BORDETELLA

TGGVGLASTLWYAE
2791
ABO77783.1




PERTUSSIS









1909

BORDETELLA

THGVGASLEAGRRF
2792
ABO77783.1




PERTUSSIS









1910

BORDETELLA

TIRRGDAPA
2793
ABO77783.1




PERTUSSIS









1911

BORDETELLA

TLANVGDTWDDDGI
2794
ABO77783.1




PERTUSSIS









1912

BORDETELLA

TLQGAGGVQIERGA
2795
ABO77783.1




PERTUSSIS









1913

BORDETELLA

TLTGGADAQGDIVA
2796
ABO77783.1




PERTUSSIS









1914

BORDETELLA

TNVTAVPASGAPAA
2797
ABO77783.1




PERTUSSIS









1915

BORDETELLA

TRAELGLGMAAALG
2798
ABO77783.1




PERTUSSIS









1916

BORDETELLA

TSSGQLSDDGIRRF
2799
ABO77783.1




PERTUSSIS









1917

BORDETELLA

TVHTNGIAHRTELR
2800
ABO77783.1




PERTUSSIS









1918

BORDETELLA

TYRYRLAANGNGQW
2801
ABO77783.1




PERTUSSIS









1919

BORDETELLA

VADHATLANVGDTW
2802
ABO77783.1




PERTUSSIS









1920

BORDETELLA

VHVGGYATYIADSG
2803
ABO77783.1




PERTUSSIS









1921

BORDETELLA

VLDGWYGVD
2804
ABO77783.1




PERTUSSIS









1922

BORDETELLA

VPASGAPAAVSVLG
2805
ABO77783.1




PERTUSSIS









1923

BORDETELLA

VRDEGGSSVLGRLG
2806
ABO77783.1




PERTUSSIS









1924

BORDETELLA

VRTASGTTIKVSGR
2807
ABO77783.1




PERTUSSIS









1925

BORDETELLA

VSGGSLSAPHGNVI
2808
ABO77783.1




PERTUSSIS









1926

BORDETELLA

VSGRQAQGILLENP
2809
ABO77783.1




PERTUSSIS









1927

BORDETELLA

VTVQRSAIVDGGLH
2810
ABO77783.1




PERTUSSIS









1928

BORDETELLA

VVLRDTNVTAVPAS
2811
ABO77783.1




PERTUSSIS









1929

BORDETELLA

WNNQSIVKTGERQH
2812
ABO77783.1




PERTUSSIS









1930

BORDETELLA

WVRNSGSEPASANT
2813
ABO77783.1




PERTUSSIS









1931

BORDETELLA

WYAESNALSKRLGE
2814
ABO77783.1




PERTUSSIS









1932

BORDETELLA

YATYIADSGFYLDA
2815
ABO77783.1




PERTUSSIS









1933

BORDETELLA

YGVDVSGSS
2816
ABO77783.1




PERTUSSIS









1934

BORDETELLA

YGVDVSGSSVELAQ
2817
ABO77783.1




PERTUSSIS









1935

BORDETELLA

YLDATLRASRLEND
2818
ABO77783.1




PERTUSSIS









1936

BORDETELLA

YRVLPEPVKLTLTG
2819
ABO77783.1




PERTUSSIS









1937

BORDETELLA

VKAQNITNKRAALIEA
2820
AAA22974.1




PERTUSSIS









1938

BORDETELLA

YYSNVTATRLLSSTNS
2821
AAA83981.1




PERTUSSIS









1939

BORDETELLA

SPNLTDERAAQAGVT
2822
CPP72976.1




PERTUSSIS









1940
MEASLES
SSRASDERAAHLPTS
2823
BAA33867.1



MORBILLIVIRUS








1941

CORYNEBACTERIUM

QVVHNSYNRPAYSPG
2824
1007216A




DIPHTHERIAE









1942
MEASLES VIRUS
AEGGEIHEL
2825
AAF85692.1



STRAIN






EDMONSTON-B








1943
MEASLES VIRUS
AENLISNGIGKY
2826
AAF85698.1



STRAIN






EDMONSTON-B








1944
MEASLES VIRUS
AEVDGDVKL
2827
CAB43772.1



STRAIN






EDMONSTON-B








1945
MEASLES VIRUS
AIYTAEIHK
2828
AAF85697.1



STRAIN






EDMONSTON-B








1946
MEASLES VIRUS
APVFHMTNY
2829
CAB43772.1



STRAIN






EDMONSTON-B








1947
MEASLES VIRUS
APVFHMTNYLEQPVSN
2830
AAR89413.1



STRAIN






EDMONSTON-B








1948
MEASLES VIRUS
AQRLNEIY
2831
AAF85698.1



STRAIN






EDMONSTON-B








1949
MEASLES VIRUS
ARVPHAYSL
2832
AAF85698.1



STRAIN






EDMONSTON-B








1950
MEASLES VIRUS
AVRDLERAM
2833
PO3424.1



STRAIN






EDMONSTON-B








1951
MEASLES VIRUS
AVRDLERAMTTLK
2834
PO3424.1



STRAIN






EDMONSTON-B








1952
MEASLES VIRUS
DALLRLQAM
2835
Q89933.1



STRAIN






EDMONSTON-B








1953
MEASLES VIRUS
DIKEKVINL
2836
AAF85698.1



STRAIN






EDMONSTON-B








1954
MEASLES VIRUS
DQGLFKVL
2837
AAF85695.1



STRAIN






EDMONSTON-B








1955
MEASLES VIRUS
DTGVDTRIW
2838
Q9EMA9.1



STRAIN






EDMONSTON-B








1956
MEASLES VIRUS
EPIGSLAIEEAM
2839
AAF85692.1



STRAIN






EDMONSTON-B








1957
MEASLES VIRUS
EPIRD ALNAM
2840
P69354.1



STRAIN






EDMONSTON-B








1958
MEASLES VIRUS
FPKLGKTL
2841
AAF85692.1



STRAIN






EDMONSTON-B








1959
MEASLES VIRUS
FRSVNAVAF
2842
AAF85695.1



STRAIN






EDMONSTON-B








1960
MEASLES VIRUS
GKIIDNTEQL
2843
AAF85695.1



STRAIN






EDMONSTON-B








1961
MEASLES VIRUS
GLNEKLVFY
2844
AAF85695.1



STRAIN






EDMONSTON-B








1962
MEASLES VIRUS
GMYGGTYLVEK
2845
AAC35876.2



STRAIN






EDMONSTON-B








1963
MEASLES VIRUS
GPPISLERLDVGTN
2846
P69354.1



STRAIN






EDMONSTON-B








1964
MEASLES VIRUS
GPRQAQVSFL
2847
Q89933.1



STRAIN






EDMONSTON-B








1965
MEASLES VIRUS
GRLVPQVRVID
2848
AAF85695.1



STRAIN






EDMONSTON-B








1966
MEASLES VIRUS
GSAPISMGFR
2849
AAF85692.1



STRAIN






EDMONSTON-B








1967
MEASLES VIRUS
HILAKSTAL
2850
AAF85698.1



STRAIN






EDMONSTON-B








1968
MEASLES VIRUS
HYREVNLVY
2851
AAF85698.1



STRAIN






EDMONSTON-B








1969
MEASLES VIRUS
IPPMKNLAL
2852
AAC35876.2



STRAIN






EDMONSTON-B








1970
MEASLES VIRUS
IPYQGSGKGVSF
2853
CAB43772.1



STRAIN






EDMONSTON-B








1971
MEASLES VIRUS
ISKESQHVY
2854
AAF85698.1



STRAIN






EDMONSTON-B








1972
MEASLES VIRUS
IVSSHFFVY
2855
AAF85698.1



STRAIN






EDMONSTON-B








1973
MEASLES VIRUS
KEIKETGRLF
2856
AAF85698.1



STRAIN






EDMONSTON-B








1974
MEASLES VIRUS
KESQHVYYL
2857
AAF85698.1



STRAIN






EDMONSTON-B








1975
MEASLES VIRUS
KIIDNTEQL
2858
AAF85695.1



STRAIN






EDMONSTON-B








1976
MEASLES VIRUS
KKQINRQN
2859
AAA63285.1



STRAIN






EDMONSTON-B








1977
MEASLES VIRUS
KKVDTNFIYQ
2860
AAF85698.1



STRAIN






EDMONSTON-B








1978
MEASLES VIRUS
KLIDGFFPA
2861
AAF85698.1



STRAIN






EDMONSTON-B








1979
MEASLES VIRUS
KPNLSSKRSEL
2862
BAB39848.1



STRAIN






EDMONSTON-B








1980
MEASLES VIRUS
KVSPYLFTV
2863
AAR89413.1



STRAIN






EDMONSTON-B








1981
MEASLES VIRUS
LETRTTNQFL
2864
CAB43772.1



STRAIN






EDMONSTON-B








1982
MEASLES VIRUS
LLKEATEL
2865
AAF85695.1



STRAIN






EDMONSTON-B








1983
MEASLES VIRUS
LLKKGNSLY
2866
AAF85698.1



STRAIN






EDMONSTON-B








1984
MEASLES VIRUS
LPAPIGGMNY
2867
AAF85698.1



STRAIN






EDMONSTON-B








1985
MEASLES VIRUS
MPEETLHQVM
2868
AAF85698.1



STRAIN






EDMONSTON-B








1986
MEASLES VIRUS
PTTIRGQFS
2869
CAB43772.1



STRAIN






EDMONSTON-B








1987
MEASLES VIRUS
QEISRHQALGY
2870
PO3424.1



STRAIN






EDMONSTON-B








1988
MEASLES VIRUS
RITHVDTESY
2871
P69354.1



STRAIN






EDMONSTON-B








1989
MEASLES VIRUS
RPGLKPDL
2872
P69354.1



STRAIN






EDMONSTON-B








1990
MEASLES VIRUS
RPIYGLEV
2873
AAF85698.1



STRAIN






EDMONSTON-B








1991
MEASLES VIRUS
RQAGQEMILAV
2874
P69354.1



STRAIN






EDMONSTON-B








1992
MEASLES VIRUS
SAVRIATVY
2875
AAF85698.1



STRAIN






EDMONSTON-B








1993
MEASLES VIRUS
SLMPEETLHQV
2876
AAF85698.1



STRAIN






EDMONSTON-B








1994
MEASLES VIRUS
SMIDLVTKF
2877
AAF85698.1



STRAIN






EDMONSTON-B








1995
MEASLES VIRUS
SMLNSQAIDNLRA
2878
P69354.1



STRAIN






EDMONSTON-B








1996
MEASLES VIRUS
SMYRVFEV
2879
CAB43772.1



STRAIN






EDMONSTON-B








1997
MEASLES VIRUS
SQQGMFHAY
2880
AAF85698.1



STRAIN






EDMONSTON-B








1998
MEASLES VIRUS
TDTPIVYNDRNLLD
2881
Q89933.1



STRAIN






EDMONSTON-B








1999
MEASLES VIRUS
VIINDDQGLFKV
2882
AAF85695.1



STRAIN






EDMONSTON-B








2000
MEASLES VIRUS
YESGVRIASL
2883
AAF85698.1



STRAIN






EDMONSTON-B








2001
MEASLES VIRUS
YLKDKALA
2884
AAF85698.1



STRAIN






EDMONSTON-B








2002
MEASLES VIRUS
YVYDHSGEAVK
2885
AAF85692.1



STRAIN






EDMONSTON-B








2003
RUBELLA VIRUS
ARVIDPAAQSFTGVV
2886
BAA28178.1





2004
RUBELLA VIRUS
SDRASARVIDPAAQS
2887
BAA28178.1





2005
RUBELLA VIRUS
VPPGKFVTAALLNTP
2888
BAA28178.1





2006
RUBELLA VIRUS
WVTPVIGSQARKCGL
2889
BAA28178.1





2007
MUMPS
GTYRLIPNARANLTA
400
AGC97176.1



RUBULAVIRUS









E. Delivery of Multi-Flap Prime Editors


In another aspect, the present disclosure provides for the delivery of multi-flap prime editors in vitro and in vivo using various strategies, including on separate vectors using split inteins and as well as direct delivery strategies of the ribonucleoprotein complex (i.e., the prime editor complexed to the PEgRNA and/or the second-site gRNA) using techniques such as electroporation, use of cationic lipid-mediated formulations, and induced endocytosis methods using receptor ligands fused to the ribonucleotprotein complexes. Any such methods are contemplated herein.


Overview of Delivery Options


In some aspects, the invention provides methods comprising delivering one or more multi-flap prime editor-encoding polynucleotides, such as or one or more vectors as described herein encoding one or more components of the multi-flap prime editing system described herein, one or more transcripts thereof, and/or one or proteins transcribed therefrom, to a host cell. In some aspects, the invention further provides cells produced by such methods, and organisms (such as animals, plants, or fungi) comprising or produced from such cells. In some embodiments, a multi-flap prime editor as described herein in combination with (and optionally complexed with) a guide sequence is delivered to a cell. Conventional viral and non-viral based gene transfer methods can be used to introduce nucleic acids in mammalian cells or target tissues. Such methods can be used to administer nucleic acids encoding components of a multi-flap prime editor system to cells in culture, or in a host organism. Non-viral vector delivery systems include DNA plasmids, RNA (e.g. a transcript of a vector described herein), naked nucleic acid, and nucleic acid complexed with a delivery vehicle, such as a liposome. Viral vector delivery systems include DNA and RNA viruses, which have either episomal or integrated genomes after delivery to the cell. For a review of gene therapy procedures, see Anderson, Science 256:808-813 (1992); Nabel & Felgner, TIBTECH 11:211-217 (1993); Mitani & Caskey, TIBTECH 11:162-166 (1993); Dillon, TIBTECH 11:167-175 (1993); Miller, Nature 357:455-460 (1992); Van Brunt, Biotechnology 6(10):1149-1154 (1988); Vigne, Restorative Neurology and Neuroscience 8:35-36 (1995); Kremer & Perricaudet, British Medical Bulletin 51(1):31-44 (1995); Haddada et al., in Current Topics in Microbiology and Immunology Doerfler and Bihm (eds) (1995); and Yu et al., Gene Therapy 1:13-26 (1994).


Methods of non-viral delivery of nucleic acids include lipofection, nucleofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipid:nucleic acid conjugates, naked DNA, artificial virions, and agent-enhanced uptake of DNA. Lipofection is described in e.g., U.S. Pat. Nos. 5,049,386, 4,946,787; and 4,897,355) and lipofection reagents are sold commercially (e.g., Transfectam™ and Lipofectin™). Cationic and neutral lipids that are suitable for efficient receptor-recognition lipofection of polynucleotides include those of Feigner, WO 91/17424; WO 91/16024. Delivery can be to cells (e.g. in vitro or ex vivo administration) or target tissues (e.g. in vivo administration).


The preparation of lipid:nucleic acid complexes, including targeted liposomes such as immunolipid complexes, is well known to one of skill in the art (see, e.g., Crystal, Science 270:404-410 (1995); Blaese et al., Cancer Gene Ther. 2:291-297 (1995); Behr et al., Bioconjugate Chem. 5:382-389 (1994); Remy et al., Bioconjugate Chem. 5:647-654 (1994); Gao et al., Gene Therapy 2:710-722 (1995); Ahmad et al., Cancer Res. 52:4817-4820 (1992); U.S. Pat. Nos. 4,186,183, 4,217,344, 4,235,871, 4,261,975, 4,485,054, 4,501,728, 4,774,085, 4,837,028, and 4,946,787).


The use of RNA or DNA viral based systems for the delivery of nucleic acids take advantage of highly evolved processes for targeting a virus to specific cells in the body and trafficking the viral payload to the nucleus. Viral vectors can be administered directly to patients (in vivo) or they can be used to treat cells in vitro, and the modified cells may optionally be administered to patients (ex vivo). Conventional viral based systems could include retroviral, lentivirus, adenoviral, adeno-associated and herpes simplex virus vectors for gene transfer. Integration in the host genome is possible with the retrovirus, lentivirus, and adeno-associated virus gene transfer methods, often resulting in long term expression of the inserted transgene. Additionally, high transduction efficiencies have been observed in many different cell types and target tissues.


The tropism of a viruses can be altered by incorporating foreign envelope proteins, expanding the potential target population of target cells. Lentiviral vectors are retroviral vectors that are able to transduce or infect non-dividing cells and typically produce high viral titers. Selection of a retroviral gene transfer system would therefore depend on the target tissue. Retroviral vectors are comprised of cis-acting long terminal repeats with packaging capacity for up to 6-10 kb of foreign sequence. The minimum cis-acting LTRs are sufficient for replication and packaging of the vectors, which are then used to integrate the therapeutic gene into the target cell to provide permanent transgene expression. Widely used retroviral vectors include those based upon murine leukemia virus (MuLV), gibbon ape leukemia virus (GaLV), Simian Immuno deficiency virus (SIV), human immuno deficiency virus (HIV), and combinations thereof (see, e.g., Buchscher et al., J. Virol. 66:2731-2739 (1992); Johann et al., J. Virol. 66:1635-1640 (1992); Sommnerfelt et al., Virol. 176:58-59 (1990); Wilson et al., J. Virol. 63:2374-2378 (1989); Miller et al., J. Virol. 65:2220-2224 (1991); PCT/US94/05700). In applications where transient expression is preferred, adenoviral based systems may be used. Adenoviral based vectors are capable of very high transduction efficiency in many cell types and do not require cell division. With such vectors, high titer and levels of expression have been obtained. This vector can be produced in large quantities in a relatively simple system. Adeno-associated virus (“AAV”) vectors may also be used to transduce cells with target nucleic acids, e.g., in the in vitro production of nucleic acids and peptides, and for in vivo and ex vivo gene therapy procedures (see, e.g., West et al., Virology 160:38-47 (1987); U.S. Pat. No. 4,797,368; WO 93/24641; Kotin, Human Gene Therapy 5:793-801 (1994); Muzyczka, J. Clin. Invest. 94:1351 (1994). Construction of recombinant AAV vectors are described in a number of publications, including U.S. Pat. No. 5,173,414; Tratschin et al., Mol. Cell. Biol. 5:3251-3260 (1985); Tratschin, et al., Mol. Cell. Biol. 4:2072-2081 (1984); Hermonat & Muzyczka, PNAS 81:6466-6470 (1984); and Samulski et al., J. Virol. 63:03822-3828 (1989).


Packaging cells are typically used to form virus particles that are capable of infecting a host cell. Such cells include 293 cells, which package adenovirus, and ψ2 cells or PA317 cells, which package retrovirus. Viral vectors used in gene therapy are usually generated by producing a cell line that packages a nucleic acid vector into a viral particle. The vectors typically contain the minimal viral sequences required for packaging and subsequent integration into a host, other viral sequences being replaced by an expression cassette for the polynucleotide(s) to be expressed. The missing viral functions are typically supplied in trans by the packaging cell line. For example, AAV vectors used in gene therapy typically only possess ITR sequences from the AAV genome which are required for packaging and integration into the host genome. Viral DNA is packaged in a cell line, which contains a helper plasmid encoding the other AAV genes, namely rep and cap, but lacking ITR sequences. The cell line may also be infected with adenovirus as a helper. The helper virus promotes replication of the AAV vector and expression of AAV genes from the helper plasmid. The helper plasmid is not packaged in significant amounts due to a lack of ITR sequences. Contamination with adenovirus can be reduced by, e.g., heat treatment to which adenovirus is more sensitive than AAV. Additional methods for the delivery of nucleic acids to cells are known to those skilled in the art. See, for example, US20030087817, incorporated herein by reference.


In various embodiments, the dual PE constructs (including, the split-constructs) may be engineered for delivery in one or more rAAV vectors. An rAAV as related to any of the methods and compositions provided herein may be of any serotype including any derivative or pseudotype (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 2/1, 2/5, 2/8, 2/9, 3/1, 3/5, 3/8, or 3/9). An rAAV may comprise a genetic load (i.e., a recombinant nucleic acid vector that expresses a gene of interest, such as a whole or split PE fusion protein that is carried by the rAAV into a cell) that is to be delivered to a cell. An rAAV may be chimeric.


As used herein, the serotype of an rAAV refers to the serotype of the capsid proteins of the recombinant virus. Non-limiting examples of derivatives and pseudotypes include rAAV2/1, rAAV2/5, rAAV2/8, rAAV2/9, AAV2-AAV3 hybrid, AAVrh.10, AAVhu.14, AAV3a/3b, AAVrh32.33, AAV-HSC15, AAV-HSC17, AAVhu.37, AAVrh.8, CHt-P6, AAV2.5, AAV6.2, AAV2i8, AAV-HSC15/17, AAVM41, AAV9.45, AAV6(Y445F/Y731F), AAV2.5T, AAV-HAE1/2, AAV clone 32/83, AAVShH10, AAV2 (Y→F), AAV8 (Y733F), AAV2.15, AAV2.4, AAVM41, and AAVr3.45. A non-limiting example of derivatives and pseudotypes that have chimeric VP1 proteins is rAAV2/5-1VP1u, which has the genome of AAV2, capsid backbone of AAV5 and VP1u of AAV1. Other non-limiting example of derivatives and pseudotypes that have chimeric VP1 proteins are rAAV2/5-8VP1u, rAAV2/9-1VP1u, and rAAV2/9-8VP1u.


AAV derivatives/pseudotypes, and methods of producing such derivatives/pseudotypes are known in the art (see, e.g., Mol Ther. 2012 April; 20(4):699-708. doi: 10.1038/mt.2011.287. Epub 2012 Jan. 24. The AAV vector toolkit: poised at the clinical crossroads. Asokan A1, Schaffer D V, Samulski R J.). Methods for producing and using pseudotyped rAAV vectors are known in the art (see, e.g., Duan et al., J. Virol., 75:7662-7671, 2001; Halbert et al., J. Virol., 74:1524-1532, 2000; Zolotukhin et al., Methods, 28:158-167, 2002; and Auricchio et al., Hum. Molec. Genet., 10:3075-3081, 2001).


Methods of making or packaging rAAV particles are known in the art and reagents are commercially available (see, e.g., Zolotukhin et al. Production and purification of serotype 1, 2, and 5 recombinant adeno-associated viral vectors. Methods 28 (2002) 158-167; and U.S. Patent Publication Numbers US20070015238 and US20120322861, which are incorporated herein by reference; and plasmids and kits available from ATCC and Cell Biolabs, Inc.). For example, a plasmid comprising a gene of interest may be combined with one or more helper plasmids, e.g., that contain a rep gene (e.g., encoding Rep78, Rep68, Rep52 and Rep40) and a cap gene (encoding VP1, VP2, and VP3, including a modified VP2 region as described herein), and transfected into a recombinant cells such that the rAAV particle can be packaged and subsequently purified.


Recombinant AAV may comprise a nucleic acid vector, which may comprise at a minimum: (a) one or more heterologous nucleic acid regions comprising a sequence encoding a protein or polypeptide of interest or an RNA of interest (e.g., a siRNA or microRNA), and (b) one or more regions comprising inverted terminal repeat (ITR) sequences (e.g., wild-type ITR sequences or engineered ITR sequences) flanking the one or more nucleic acid regions (e.g., heterologous nucleic acid regions). Herein, heterologous nucleic acid regions comprising a sequence encoding a protein of interest or RNA of interest are referred to as genes of interest.


Any one of the rAAV particles provided herein may have capsid proteins that have amino acids of different serotypes outside of the VP1u region. In some embodiments, the serotype of the backbone of the VP1 protein is different from the serotype of the ITRs and/or the Rep gene. In some embodiments, the serotype of the backbone of the VP1 capsid protein of a particle is the same as the serotype of the ITRs. In some embodiments, the serotype of the backbone of the VP1 capsid protein of a particle is the same as the serotype of the Rep gene. In some embodiments, capsid proteins of rAAV particles comprise amino acid mutations that result in improved transduction efficiency.


In some embodiments, the nucleic acid vector comprises one or more regions comprising a sequence that facilitates expression of the nucleic acid (e.g., the heterologous nucleic acid), e.g., expression control sequences operatively linked to the nucleic acid. Numerous such sequences are known in the art. Non-limiting examples of expression control sequences include promoters, insulators, silencers, response elements, introns, enhancers, initiation sites, termination signals, and poly(A) tails. Any combination of such control sequences is contemplated herein (e.g., a promoter and an enhancer).


Final AAV constructs may incorporate a sequence encoding the PEgRNA. In other embodiments, the AAV constructs may incorporate a sequence encoding the second-site nicking guide RNA. In still other embodiments, the AAV constructs may incorporate a sequence encoding the second-site nicking guide RNA and a sequence encoding the PEgRNA.


In various embodiments, the PEgRNAs and the second-site nicking guide RNAs can be expressed from an appropriate promoter, such as a human U6 (hU6) promoter, a mouse U6 (mU6) promoter, or other appropriate promoter. The PEgRNAs and the second-site nicking guide RNAs can be driven by the same promoters or different promoters.


In some embodiments, a rAAV constructs or the herein compositions are administered to a subject enterally. In some embodiments, a rAAV constructs or the herein compositions are administered to the subject parenterally. In some embodiments, a rAAV particle or the herein compositions are administered to a subject subcutaneously, intraocularly, intravitreally, subretinally, intravenously (IV), intracerebro-ventricularly, intramuscularly, intrathecally (IT), intracisternally, intraperitoneally, via inhalation, topically, or by direct injection to one or more cells, tissues, or organs. In some embodiments, a rAAV particle or the herein compositions are administered to the subject by injection into the hepatic artery or portal vein.


Split Multi-Flap PE Vector-Based Strategies


In this aspect, the multi-flap prime editors can be divided at a split site and provided as two halves of a whole/complete prime editor. The two halves can be delivered to cells (e.g., as expressed proteins or on separate expression vectors) and once in contact inside the cell, the two halves form the complete prime editor through the self-splicing action of the inteins on each prime editor half. Split intein sequences can be engineered into each of the halves of the encoded prime editor to facilitate their transplicing inside the cell and the concomitant restoration of the complete, functioning PE.


These split intein-based methods overcome several barriers to in vivo delivery. For example, the DNA encoding prime editors is larger than the rAAV packaging limit, and so requires special solutions. One such solution is formulating the editor fused to split intein pairs that are packaged into two separate rAAV particles that, when co-delivered to a cell, reconstitute the functional editor protein. Several other special considerations to account for the unique features of multi-flap prime editing are described, including the optimization of second-site nicking targets and properly packaging multi-flap prime editors into virus vectors, including lentiviruses and rAAV.


In this aspect, the multi-flap prime editors can be divided at a split site and provided as two halves of a whole/complete prime editor. The two halves can be delivered to cells (e.g., as expressed proteins or on separate expression vectors) and once in contact inside the cell, the two halves form the complete prime editor through the self-splicing action of the inteins on each prime editor half. Split intein sequences can be engineered into each of the halves of the encoded prime editor to facilitate their transplicing inside the cell and the concomitant restoration of the complete, functioning PE.



FIG. 66 depicts one embodiment of a prime editor being provided as two PE half proteins which regenerate as whole prime editor through the self-splicing action of the split-intein halves located at the end or beginning of each of the prime editor half proteins. The multi-flap prime editors described herein may also be provided in the same manner. As used herein, the term “PE N-terminal half” refers to the N-terminal half of a complete prime editor and which comprises the “N intein” at the C-terminal end of the PE N-terminal half (i.e., the N-terminal extein) of the complete prime editor. The “N intein” refers to the N-terminal half of a complete, fully-formed split-intein moiety. As used herein, the term “PE C-terminal half” refers to the C-terminal half of a complete prime editor and which comprises the “C intein” at the N-terminal end of the C-terminal half (i.e., the C-terminal extein) of a complete prime editor. When the two half proteins, i.e., the PE N-terminal half and the PE C-terminal half, come into contact with one another, e.g., within the cell, the N intein and the C intein undergo the simultaneous process of self-excision and the formation of a peptide bond between the C-terminal end of the PE N-terminal half and the N-terminal end of the PE C-terminal half to reform the complete prime editor protein comprising the complete napDNAbp domain (e.g., Cas9 nickase) and the RT domain Although not shown in the drawing, the prime editor may also comprise additional sequences including NLS at the N-terminus and/or C-terminus, as well as amino acid linkers sequences joining each domain.


In various embodiments, the multi-flap prime editors may be engineered as two half proteins (i.e., a PE N-terminal half and a PE C-terminal half) by “splitting” the whole prime editor as a “split site.” The “split site” refers to the location of insertion of split intein sequences (i.e., the N intein and the C intein) between two adjacent amino acid residues in the prime editor. More specifically, the “split site” refers to the location of dividing the whole prime editor into two separate halves, wherein in each halve is fused at the split site to either the N intein or the C intein motifs. The split site can be at any suitable location in the prime editor fusion protein, but preferably the split site is located at a position that allows for the formation of two half proteins which are appropriately sized for delivery (e.g., by expression vector) and wherein the inteins, which are fused to each half protein at the split site termini, are available to sufficiently interact with one another when one half protein contacts the other half protein inside the cell.


In some embodiments, the split site is located in the napDNAbp domain. In other embodiments, the split site is located in the RT domain. In other embodiments, the split site is located in a linker that joins the napDNAbp domain and the RT domain.


In various embodiments, split site design requires finding sites to split and insert an N- and C-terminal intein that are both structurally permissive for purposes of packaging the two half prime editor domains into two different AAV genomes. Additionally, intein residues necessary for trans splicing can be incorporated by mutating residues at the N terminus of the C terminal extein or inserting residues that will leave an intein “scar.”


Exemplary split configurations of split multi-flap (e.g., dual flap or quadruple flap) prime editors comprising either the SpCas9 nickase or the SaCas9 nickase are as follows.











S. PYOGENES PE, SPLIT AT LINKER, N TERMINAL PORTION




STRUCTURE: [N EXTEIN]-[N INTEIN]


(SEQ ID NO: 443)




MKRTADGSEFESPKKKRKV
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSI





KKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEES




FLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFR




GHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIA




QLPGEKKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYA




DLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYK




EIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNG




SIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKS




EETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKY




VTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNA




SLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQ




LKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQK




AQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTT




QKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQEL




DINRLSDYDVDAIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLL




NAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDEND




KLIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEF




VYGDYKYYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGET




GEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDP




KKYGGFDSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKE




VKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSP




EDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQAENII




HLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGD

SGGS






SGGS
CLSYETEILTVEYGLLPIGKIVEKRIECTVYSVDNNGNIYTQPVAQWHDRGEQEVFEYCL



EDGSLIRATKDHKFMTVDGQMLPIDEIFERELDLMRVDNLPNSGGSKRrADGSEFEPKKKRKV





KEY:



NLS (SEQ ID NO: 124, 125)



CAS9 (SEQ ID NO: 445)




LINKER
 (SEQ ID NO: 446)



NPUN INTEIN (SEQ ID NO: 447)






S. PYOGENES PE, SPLIT AT LINKER, C TERMINAL PORTION



STRUCTURE: [C INTEIN]-[C EXTEIN]


(SEQ ID NO: 450)




MKRTADGSEFESPKKKRKV
IKIATRKYLGKQNVYDIGVERDHNFALKNGFIASN
CFNSGSETPGTS






ESATPESSGGSSGGSS
TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIP






LKATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLREV






NKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLT






WTRLPQGFKNSPTLFDEALHRDLADFRIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNL






GYRASAKKAQICQKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPG






FAEMAAPLYPLTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQ






KLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPDR






WLSNARMTHYQALLLDTDRVQFGPWALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPD






ADHTWYTDGSSLLQEGQRKAGAAVFPETEVIWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTD






SRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLSIIHCPGHQKGHSAEARGNRMA






DQAARKAAITETPDTSTLLIENSSP
SGGSKRYADGSEFEPKKKRKV







KEY:



NLS (SEQ ID NO: 124, 125)





LINKER 1
 (SEQ ID NO: 453)





LINKER 2
 (SEQ ID NO: 174)




NPUC INTEIN (SEQ ID NO: 452)





RT
 (SEQ ID NO: 454)







S. AUREUS PE, SPLIT BETWEEN RESIDUES 740/741, N TERMINAL PORTION



STRUCTURE: [N EXTEIN]-[N INTEIN]


(SEQ ID NO: 458)




MKRTADGSEFESPKKKRKV
GKRNYILGLAIGITSVGYGIIDYETRDVIDAGVRLFKEANVEN





NEGRRSKRGARRLKRRRRHRIQRVKKLLFDYNLLTDHSELSGINPYEARVKGLSQKLSEE




EFSAALLHLAKRRGVHNVNEVEEDTGNELSTKEQISRNSKALEEKYVAELQLERLKKDG




EVRGSINRFKTSDYVKEAKQLLKVQKAYHQLDQSFIDTYIDLLETRRTYYEGPGEGSPFG




WKDIKEWYEMLMGHCTYFPEELRSVKYAYNADLYNALNDLNNLVITRDENEKLEYYEK




FQIIENVFKQKKKPTLKQIAKEILVNEEDIKGYRVTSTGKPEFTNLKVYHDIKDITARKEII




ENAELLDQIAKILTIYQSSEDIQEELTNLNSELTQEEIEQISNLKGYTGTHNLSLKAINLILD




ELWHTNDNQIAIFNRLKLVPKKVDLSQQKEIPTTLVDDFILSPVVKRSFIQSIKVINAIIKKY




GLPNDIIIELAREKNSKDAQKMINEMQKRNRQTNERIEEIIRTTGKENAKYLIEKIKLHDM




QEGKCLYSLEAIPLEDLLNNPFNYEVDHIIPRSVSFDNSFNNKVLVKQEENSKKGNRTPFQ




YLSSSDSKISYETFKKHILNLAKGKGRISKTKKEYLLEERDINRFSVQKDFINRNLVDTRYA




TRGLMNLLRSYFRVNNLDVKVKSINGGFTSFLRRKWKFKKERNKGYKHHAEDALIIANA




DFIFKEWKKLDKAKKVMENQMFEEKQAECLSYETEILTVEYGLLPIGKIVEKRIECTVYSVD



NNGNIYTQPVAQWHDRGEQEVFEYCLEDGSLIRATKDHKFMTVDGQMLPIDEIFERELDLMRV


DNLPNSGGSKRTADGSEFEPKKKRKV





KEY:



NLS (SEQ ID NO: 124, 125)




CAS9 (SEQ ID NO: 460)





LINKER
 (SEQ ID NO: 174)



NPUN INTEIN (SEQ ID NO: 462)






S. AUREUS PE, SPLIT BETWEEN RESIDUES 740/741, C TERMINAL PORTION



STRUCTURE: [C INTEIN]-[C EXTEIN]


(SEQ ID NO: 465)




MKRTADGSEFESPKKKRKV
IKIATRKYLGKQNVYDIGVERDHNFALKNGFIASN

CFN

EIETEQEY





KEIFITPHQIKHIKDFKDYKYSHRVDKKPNRKLINDTLYSTRKDDKGNTLIVNNLNGLYDK




DNDKLKKLINKSPEKLLMYHHDPQTYQKLKLIMEQYGDEKNPLYKYYEETGNYLTKYS




KKDNGPVIKKIKYYGNKLNAHLDITDDYPNSRNKVVKLSLKPYRFDVYLDNGVYKFVTV




KNLDVIKKENYYEVNSKCYEEAKKLKKISNQAEFIASFYKNDLIKINGELYRVIGVNNDLL




NRIEVNMIDITYREYLENMNDKRPPHIIKTIASKTQSIKKYSTDILGNLYEVKSKKHPQIIK




KG

SGGSSGGSSGSETPGTSESATPFSSGGSSGGSS
TLNIEDEYRLHETSKEPDVSLGSTWLSDFP






QAWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVPCQSPWNTPL






LPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTS






QPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFDEALHRDLADFRIQHPDLILLQYVDDLLLAAT






SELDCQQGTRALLQTLGNLGYRASAKKAQICQKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTP






RQLREFLGTAGFCRLWIPGFAEMAAPLYPLTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTK






PFELFVDEKQGYAKGVLTQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQ






PLVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQFGPWALNPATLLPLPEEGLQHNCLD






ILAEAHGTRPDLTDQPLPDADHTWYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELI






ALTQALKMAEGKKLNVYTDSRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLSIIH






CPGHQKGHSAEARGNRMADQAARKAAITETPDTSTLLIENSSP
SGGSKRTADGSEFEPKKKRKV







KEY:



NLS (SEQ ID NO: 124, 125)




CAS9 (SEQ ID NO: 467)





LINKER 1
 (SEQ ID NO: 127)





LINKER 2
 (SEQ ID NO: 174)




NPUC INTEIN (SEQ ID NO: 452)





RT
 (SEQ ID NO: 471)







In various embodiments, using SpCas9 nickase (SEQ ID NO: 18, 1368 amino acids) as an example, the split can between any two amino acids between 1 and 1368. Preferred splits, however, will be located between the central region of the protein, e.g., from amino acids 50-1250, or from 100-1200, or from 150-1150, or from 200-1100, or from 250-1050, or from 300-1000, or from 350-950, or from 400-900, or from 450-850, or from 500-800, or from 550-750, or from 600-700 of SEQ ID NO: 18. In specific exemplary embodiments, the split site may be between 740/741, or 801/802, or 1010/1011, or 1041/1042. In other embodiments the split site may be between 1/2, 2/3, 3/4, 4/5, 5/6, 6/7, 7/8, 8/9, 9/10, 10/11, 12/13, 14/15, 15/16, 17/18, 19/20, 20/21, 21/22, 22/23, 23/24, 24/25, 25/26, 26/27, 27/28, 28/29, 29/30, 30/31, 31/32, 32/33, 33/34, 34/35, 35/36, 36/37, 38/39, 39/40, 41/42, 42/43, 43/44, 44/45, 45/46, 46/47, 47/48, 48/49, 49/50, 51/52, 52/53, 53/54, 54/55, 55/56, 56/57, 57/58, 58/59, 59/60, 61/62, 62/63, 63/64, 64/65, 65/66, 66/67, 67/68, 68/69, 69/70, 71/72, 72/73, 73/74, 74/75, 75/76, 76/77, 77/78, 78/79, 79/80, 81/82, 82/83, 83/84, 84/85, 85/86, 86/87, 87/88, 88/89, 89/90, or between any two pairs of adjacent residues between 90-100, 100-150, 150-200, 200-250, 250-300, 300-350, 350-400, 450-500, 500-550, 550-600, 600-650, 650-700, 700-750, 750-800, 800-850, 850-900, 900-950, 950-1000, 1000-1050, 1050-1100, 1100-1150, 1150-1200, 1200-1250, 1250-1300, 1300-1350, and 1350-1368, relative to SpCas9 of SEQ ID NO: 18, at between any two corresponding residues in an amino acid sequence having at least 80%, 85%, 90%, 95%, 98%, 99%, or 99.9% sequence identity with SEQ ID NO: 18, or between any two corresponding residues in a variant or equivalent of SpCas9 of any of amino acid sequences SEQ ID NOs. 19-88, or an amino acid sequence having at least 80%, 85%, 90%, 95%, 98%, 99%, or 99.9% sequence identity with any of SEQ ID NOs: 19-88.


In various embodiments, the split intein sequences can be engineered by from the following intein sequences.













NAME
SEQUENCE OF LIGAND-DEPENDENT INTEIN







2-4 INTEIN:
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGAIVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 472)





3-2 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAVAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGAIVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYTNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 473)





30R3-1 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPIPYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEGLRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 474)





30R3-2 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 475)





30R3-3 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPIPYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLECAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 476)





37R3-1 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYNPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLERAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEGLRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC ((SEQ ID NO: 477)





37R3-2 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAAAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGAIVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLERAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEGLRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 478)





37R3-3 INTEIN
CLAEGTRIFDPVTGTTHRIEDVVDGRKPIHVVAVAKDGTLLARPVVSWFD



QGTRDVIGLRIAGGATVWATPDHKVLTEYGWRAAGELRKGDRVAGPGGS



GNSLALSLTADQMVSALLDAEPPILYSEYDPTSPFSEASMMGLLTNLADRE



LVHMINWAKRVPGFVDLTLHDQAHLLERAWLEILMIGLVWRSMEHPGKL



LFAPNLLLDRNQGKCVEGMVEIFDMLLATSSRFRMMNLQGEEFVCLKSII



LLNSGVYTFLSSTLKSLEEKDHIHRALDKITDTLIHLMAKAGLTLQQQHQR



LAQLLLILSHIRHMSNKGMEHLYSMKYKNVVPLYDLLLEMLDAHRLHAG



GSGASRVQAFADALDDKFLHDMLAEELRYSVIREVLPTRRARTFDLEVEE



LHTLVAEGVVVHNC (SEQ ID NO: 479)









In various other embodiments, the split intein sequences can be used as follows:













INTEIN-N
INTEIN-C







NPU-N
NPU-C


CLSYETEILTVEYGLLPIGKIVEKRIEC
IKIATRKYLGKQNVYDIGVE


TVYSVDNNGNIYTQPVAQWHDRGEQEVF
RDHNFALKNGFIASN


EYCLEDGSLIRATKDHKFMTVDGQMLPI
(SEQ ID NO: 452)


DEIFERELDLMRVDNLPNSGGS



(SEQ ID NO: 447)









In various embodiments, the split inteins can be used to separately deliver separate portions of a complete PE fusion protein to a cell, which upon expression in a cell, become reconstituted as a complete PE fusion protein through the trans splicing.


In some embodiments, the disclosure provides a method of delivering a PE fusion protein to a cell, comprising:

    • (a) constructing a first expression vector encoding an N-terminal fragment of the PE fusion protein fused to a first split intein sequence;
    • (b) constructing a second expression vector encoding a C-terminal fragment of the PE fusion protein fused to a second split intein sequence;
    • (c) delivering the first and second expression vectors to a cell,


      wherein the N-terminal and C-terminal fragment are reconstituted as the PE fusion protein in the cell as a result of trans splicing activity causing self-excision of the first and second split intein sequences.


The split site in some embodiments can be anywhere in the prime editor fusion, including the napDNAbp domain, the linker, or the reverse transcriptase domain.


In other embodiments, the split site is in the napDNAbp domain.


In still other embodiments, the split site is in the reverse transcriptase or polymerase domain.


In yet other embodiments, the split site is in the linker.


In various embodiments, the present disclosure provides multi-flap prime editors comprising a napDNAbp (e.g., a Cas9 domain) and a reverse transcriptase wherein one or both of the napDNAbp and/or the reverse transcriptase comprise an intein, for example, a ligand-dependent intein. Typically the intein is a ligand-dependent intein which exhibits no or minimal protein splicing activity in the absence of ligand (e.g., small molecules such as 4-hydroxytamoxifen, peptides, proteins, polynucleotides, amino acids, and nucleotides). Ligand-dependent inteins are known, and include those described in U.S. patent application, U.S. Ser. No. 14/004,280, published as U.S. 2014/0065711 A1, the entire contents of which are incorporated herein by reference. In addition, use of split-Cas9 architecture In some embodiments, the intein comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 8-15, 447, 452, 462, and 472-479.


In various embodiments, the napDNAbp domains are smaller-sized napDNAbp domains as compared to the canonical SpCas9 domain of SEQ ID NO: 18.


The canonical SpCas9 protein is 1368 amino acids in length and has a predicted molecular weight of 158 kilodaltons. The term “small-sized Cas9 variant”, as used herein, refers to any Cas9 variant—naturally occurring, engineered, or otherwise—that is less than at least 1300 amino acids, or at least less than 1290 amino acids, or than less than 1280 amino acids, or less than 1270 amino acid, or less than 1260 amino acid, or less than 1250 amino acids, or less than 1240 amino acids, or less than 1230 amino acids, or less than 1220 amino acids, or less than 1210 amino acids, or less than 1200 amino acids, or less than 1190 amino acids, or less than 1180 amino acids, or less than 1170 amino acids, or less than 1160 amino acids, or less than 1150 amino acids, or less than 1140 amino acids, or less than 1130 amino acids, or less than 1120 amino acids, or less than 1110 amino acids, or less than 1100 amino acids, or less than 1050 amino acids, or less than 1000 amino acids, or less than 950 amino acids, or less than 900 amino acids, or less than 850 amino acids, or less than 800 amino acids, or less than 750 amino acids, or less than 700 amino acids, or less than 650 amino acids, or less than 600 amino acids, or less than 550 amino acids, or less than 500 amino acids, but at least larger than about 400 amino acids and retaining the required functions of the Cas9 protein.


In one embodiment, as depicted in Example 20, the specification embraces the following split-intein PE constructs, which are split between residues 1024 and 1025 of the canonical SpCas9 (SEQ ID NO: 18) (or which may be referred to as residues 1023 and 1024, respectively, relative to a Met-minus SEQ ID NO: 18).


First, the amino acid sequence of SEQ ID NO: 18 is shown as follows, indicating the location of the split site between 1024 (“K”) and 1025 (“S”) residues:














Description
Sequence
SEQ ID NO:







SpCas9


M
DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDR

SEQ ID NO: 18,



Streptococcus

HSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYL
indicated with



pyogenes

QEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVD
split site


M1
EVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFR
1024/1025 in


SwissProt
GHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVD
bold


Accession
AKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTP
The M at


No. Q99ZW2
NFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLF
position 1 is not


Wild type
LAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDL
necessarily



TLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFY
present in the



KFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHL
PE fusion



GELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLARGN
protein in



SRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDK
certain



NLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLS
embodiments.



GEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISGV
Thus, the



EDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLF
numbering of



EDREMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKL
the split site is



INGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKEDIQ
1023/1024 in



KAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKV
the case that the



MGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKEL
amino acid



GSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDIN
sequence



RLSDYDVDHIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPS
excludes Met at



EEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGGLSELD
position 1.



KAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREV




KVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVV




GTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATA




KYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKG




RDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDK




LIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLK




SVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKY




SLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHY




EKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADA




NLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYF




DTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGD









In this configuration, the amino acid sequence of N-terminal half (amino acids 1-1024) is as follows:









(SEQ ID NO: 3877)


MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGA





LLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHR





LEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKAD





LRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENP





INASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTP





NFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAI





LLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEI





FFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLR





KQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPY





YVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDK





NLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVD





LLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKI





IKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQ





LKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDD





SLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKV





MGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHP





VENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSFLKDD





SIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNL





TKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLI





REVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKK





YPKLESEFVYGDYKVYDVRKMIAK.






In this configuration, the amino acid sequence of N-terminal half (amino acids 1-1023) (where the protein is Met-minus at position 1) is as follows:









(SEQ ID NO: 3878)


DKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGAL





LFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRL





EESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADL





RLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPI





NASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPN





FKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAIL





LSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIF





FDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRK





QRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYY





VGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKN





LPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDL





LFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKII





KDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQL





KRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDS





LTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVM





GRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPV





ENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDAIVPQSFLKDDS





IDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLT





KAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIR





EVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKY





PKLESEFVYGDYKVYDVRKMIAK.






In this configuration, the amino acid sequence of C-terminal half (amino acids 1024-1368 (or counted as amino acids 1023-1367 in a Met-minus Cas9) is as follows:









(SEQ ID NO: 3879)




S
EQEIGKATAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVW






DKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKK





DWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSF





EKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGN





ELALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQIS





EFSKRVILADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAF





KYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGD.






As shown in Example 20, the PE2 (which is based on SpCas9 of SEQ ID NO: 18) construct was split at position 1023/1024 (relative to a Met-minus SEQ ID NO: 18) into two separate constructs, as follows:










SpPE2 split at 1023/1024 N terminal half



(SEQ ID NO: 3875)




MKRTADGSEFESPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRH







SIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFLV





EEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEG





DLNPDNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLF





GNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLS





DILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGAS





QEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLK





DNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFD





KNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLFKTNRKVTVK





QLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDR





EMIEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNF





MQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKVMGRHKPE





NIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRD





MYVDQELDINRLSDYDVDAIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWR





QLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDK





LIREVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESEFVYGD







embedded image






embedded image






embedded image




SpPE2 split at 1023/1024 C terminal half


(SEQ ID NO: 3876)





embedded image





FYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQT





GGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKLKSVKELLGI





TIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNELALPSKY





VNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKH





RDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYETRIDLSQL







embedded image





DFPQAWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPMSQEARLGIKPHIQRLLDQGILVP







CQSPWNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNPYNLLSGLPPSHQWYTVLD







LKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPTLFNEALHRDLADFR







IQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQICQKQVKYLGY







LLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPGFAEMAAPLYPLTKPG







TLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRR







PVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPDR







WLSNARMTHYQALLLDTDRVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLT







DQPLPDADHTWYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQAL







KMAEGKKLNVYTDSRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRLS








embedded image




KRK 




embedded image








The present disclosure also contemplates methods of delivering split-intein multi-flap prime editors to cells and/or treating cells with split-intein multi-flap prime editors.


In some embodiments, the disclosure provides a method of delivering a PE fusion protein to a cell, comprising:

    • (a) constructing a first expression vector encoding an N-terminal fragment of the PE fusion protein fused to a first split intein sequence;
    • (b) constructing a second expression vector encoding a C-terminal fragment of the PE fusion protein fused to a second split intein sequence;
    • (c) delivering the first and second expression vectors to a cell,


      wherein the N-terminal and C-terminal fragment are reconstituted as the PE fusion protein in the cell as a result of trans splicing activity causing self-excision of the first and second split intein sequences.


In certain embodiments, the N-terminal fragment of the PE fusion protein fused to a first split intein sequence is SEQ ID NO: 3875, or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99.9% sequence identity with SEQ ID NO: 3875.


In other embodiments, the C-terminal fragment of the PE fusion protein fused to a first split intein sequence is SEQ ID NO: 3876, or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99.9% sequence identity with SEQ ID NO: 3876.


In other embodiments, the disclosure provides a method of editing a target DNA sequence within a cell, comprising:

    • (a) constructing a first expression vector encoding an N-terminal fragment of the PE fusion protein fused to a first split intein sequence;
    • (b) constructing a second expression vector encoding a C-terminal fragment of the PE fusion protein fused to a second split intein sequence;
    • (c) delivering the first and second expression vectors to a cell,


      wherein the N-terminal and C-terminal fragment are reconstituted as the PE fusion protein in the cell as a result of trans splicing activity causing self-excision of the first and second split intein sequences.


In certain embodiments, the N-terminal fragment of the PE fusion protein fused to a first split intein sequence is SEQ ID NO: 3875, or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99.9% sequence identity with SEQ ID NO: 3875.


In other embodiments, the C-terminal fragment of the PE fusion protein fused to a first split intein sequence is SEQ ID NO: 3876, or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99.9% sequence identity with SEQ ID NO: 3876.


Delivery of PE Ribonucleoprotein Complexes


In this aspect, the multi-flap prime editors may be delivered by non-viral delivery strategies involving delivery of a multi-flap prime editor complexed with PEgRNA (i.e., a PE ribonucleoprotein complex) by various methods, including electroporation and lipid nanoparticles. Methods of non-viral delivery of nucleic acids include lipofection, nucleofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipid:nucleic acid conjugates, naked DNA, artificial virions, and agent-enhanced uptake of DNA. Lipofection is described in e.g., U.S. Pat. Nos. 5,049,386, 4,946,787; and 4,897,355) and lipofection reagents are sold commercially (e.g., Transfectam™ and Lipofectin™) Cationic and neutral lipids that are suitable for efficient receptor-recognition lipofection of polynucleotides include those of Feigner, WO 91/17424; WO 91/16024. Delivery can be to cells (e.g. in vitro or ex vivo administration) or target tissues (e.g. in vivo administration).


The preparation of lipid:nucleic acid complexes, including targeted liposomes such as immunolipid complexes, is well known to one of skill in the art (see, e.g., Crystal, Science 270:404-410 (1995); Blaese et al., Cancer Gene Ther. 2:291-297 (1995); Behr et al., Bioconjugate Chem. 5:382-389 (1994); Remy et al., Bioconjugate Chem. 5:647-654 (1994); Gao et al., Gene Therapy 2:710-722 (1995); Ahmad et al., Cancer Res. 52:4817-4820 (1992); U.S. Pat. Nos. 4,186,183, 4,217,344, 4,235,871, 4,261,975, 4,485,054, 4,501,728, 4,774,085, 4,837,028, and 4,946,787).


Additional reference may be made to the following references that discuss approaches for non-viral delivery of ribonucleoprotein complexes, each of which are incorporated herein by reference.

  • Chen, Sean, et al. “Highly efficient mouse genome editing by CRISPR ribonucleoprotein electroporation of zygotes.” Journal of Biological Chemistry (2016): jbc-M116. PubMed
  • Zuris, John A., et al. “Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo.” Nature biotechnology 33.1 (2015): 73. PubMed
  • Rouet, Romain, et al. “Receptor-Mediated Delivery of CRISPR-Cas9 Endonuclease for Cell-Type-Specific Gene Editing.” Journal of the American Chemical Society 140.21 (2018): 6596-6603. PubMed.



FIG. 68C provides data showing that various disclosed PE ribonucleoprotein complexes (PE2 at high concentration, PE3 at high concentration and PE3 at low concentration) can be delivered in this manner.


Delivery of PE by mRNA


Another method that may be employed to deliver multi-flap prime editors and/or PEgRNAs to cells in which multi-flap prime editing-based genome editing is desired is by employing the use of messenger RNA (mRNA) delivery methods and technologies. Examples of mRNA delivery methods and compositions that may be utilized in the present disclosure including, for example, PCT/US2014/028330, U.S. Pat. No. 8,822,663B2, NZ700688A, ES2740248T3, EP2755693A4, EP2755986A4, WO2014152940A1, EP3450553B1, BR112016030852A2, and EP3362461A1, each of which are incorporated herein by reference in their entireties. Additional disclosure hereby incorporated by reference can be found in Kowalski et al., “Delivering the Messenger: Advances in Technologies for Therapeutic mRNA Delivery,” Mol Therap., 2019; 27(4): 710-728.


In contrast to DNA vector encoding multi-flap prime editors, the use of RNA as a delivery agent for multi-flap prime editors has the advantage that the genetic material does not have to enter the nucleus to perform its function. The delivered mRNA may be directly translated in the cytoplasm into the desired protein (e.g., prime editor fusion protein) and nucleic acid products (e.g., PEgRNA). However, in order to be more stable (e.g., resist RNA-degrading enzymes in the cytoplasm), it is in some embodiments necessary to stabilize the mRNA to improve delivery efficiency. Certain delivery carriers such as cationic lipids or polymeric delivery carriers can also help protect the transfected mRNA from endogenous RNase enzymes that might otherwise degrade the therapeutic mRNA encoding the desired prime editor fusion proteins. In addition, despite the increased stability of modified mRNA, delivery of mRNA, particularly mRNA encoding full-length protein, to cells in vivo in a manner that allows therapeutic levels of protein production remains a challenge.


With some exceptions, the intracellular delivery of mRNA is generally more challenging than that of small oligonucleotides, and it requires encapsulation into a delivery nanoparticle, in part due to the significantly larger size of mRNA molecules (300-5,000 kDa, ˜1-15 kb) as compared to other types of RNAs (small interfering RNAs [siRNAs], ˜14 kDa; antisense oligonucleotides [ASOs], 4-10 kDa).


mRNA must cross the cell membrane in order to reach the cytoplasm. The cell membrane is a dynamic and formidable barrier to intracellular delivery. It is made up primarily of a lipid bilayer of zwitterionic and negatively charged phospholipids, where the polar heads of the phospholipids point toward the aqueous environment and the hydrophobic tails form a hydrophobic core.


In some embodiments, the mRNA compositions of the disclosure comprise mRNA (encoding a prime editor and/or PEgRNA), a transport vehicle, and optionally an agent that facilitates contact with the target cell and subsequent transfection.


In some embodiments, the mRNA can include one or more modifications that confer stability to the mRNA (eg, compared to the wild-type or native version of the mRNA) and is involved in the associated abnormal expression of the protein. One or more modifications to the wild type that correct the defect may also be included. For example, the nucleic acids of the invention can include modifications of one or both of a 5′ untranslated region or a 3′ untranslated region. Such modifications may include the inclusion of sequences encoding a partial sequence of the cytomegalovirus (CMV) immediate early 1 (IE1) gene, poly A tail, Cap1 structure, or human growth hormone (hGH). In some embodiments, the mRNA is modified to reduce mRNA immunogenicity.


In one embodiment, the multi-flap prime editor mRNA in the composition of the invention can be formulated in a liposome transfer vehicle to facilitate delivery to target cells. Contemplated transfer vehicles can include one or more cationic lipids, non-cationic lipids, and/or PEG-modified lipids. For example, the transfer vehicle can include at least one of the following cationic lipids: C12-200, DLin-KC2-DMA, DODAP, HGT4003, ICE, HGT5000, or HGT5001. In embodiments, the transfer vehicle comprises cholesterol (chol) and/or PEG modified lipids. In some embodiments, the transfer vehicle comprises DMG-PEG2K. In certain embodiments, the transfer vehicle has the following lipid formulation: C12-200, DOPE, chol, DMG-PEG2K; DODAP, DOPE, cholesterol, DMG-PEG2K; HGT5000, DOPE, chol, DMG-PEG2K, HGT5001, DOPE, chol, one of DMG-PEG2K.


The present disclosure also provides compositions and methods useful for facilitating transfection of target cells with one or more PE-encoding mRNA molecules. For example, the compositions and methods of the present invention contemplate the use of targeting ligands that can increase the affinity of the composition for one or more target cells. In one embodiment, the targeting ligand is apolipoprotein B or apolipoprotein E, and the corresponding target cells express low density lipoprotein receptors and thus promote recognition of the targeting ligand. A vast number of target cells can be preferentially targeted using the methods and compositions of the present disclosure. For example, contemplated target cells include hepatocytes, epithelial cells, hematopoietic cells, epithelial cells, endothelial cells, lung cells, bone cells, stem cells, mesenchymal cells, nerve cells, heart cells, adipocytes, vascular smooth muscle Includes cells, cardiomyocytes, skeletal muscle cells, beta cells, pituitary cells, synovial lining cells, ovarian cells, testis cells, fibroblasts, B cells, T cells, reticulocytes, leukocytes, granulocytes, and tumor cells However, it is not limited to these.


In some embodiments, the PE-encoding mRNA may optionally have chemical or biological modifications which, for example, improve the stability and/or half-life of such mRNA or which improve or otherwise facilitate protein production. Upon transfection, a natural mRNA in the compositions of the invention may decay with a half-life of between 30 minutes and several days. The mRNAs in the compositions of the disclosure may retain at least some ability to be translated, thereby producing a functional protein or enzyme. Accordingly, the invention provides compositions comprising and methods of administering a stabilized mRNA. In some embodiments, the activity of the mRNA is prolonged over an extended period of time. For example, the activity of the mRNA may be prolonged such that the compositions of the present disclosure are administered to a subject on a semi-weekly or bi-weekly basis, or more preferably on a monthly, bi-monthly, quarterly or an annual basis. The extended or prolonged activity of the mRNA of the present invention is directly related to the quantity of protein or enzyme produced from such mRNA. Similarly, the activity of the compositions of the present disclosure may be further extended or prolonged by modifications made to improve or enhance translation of the mRNA. Furthermore, the quantity of functional protein or enzyme produced by the target cell is a function of the quantity of mRNA delivered to the target cells and the stability of such mRNA. To the extent that the stability of the mRNA of the present invention may be improved or enhanced, the half-life, the activity of the produced protein or enzyme and the dosing frequency of the composition may be further extended.


Accordingly, in some embodiments, the mRNA in the compositions of the disclosure comprise at least one modification which confers increased or enhanced stability to the nucleic acid, including, for example, improved resistance to nuclease digestion in vivo. As used herein, the terms “modification” and “modified” as such terms relate to the nucleic acids provided herein, include at least one alteration which preferably enhances stability and renders the mRNA more stable (e.g., resistant to nuclease digestion) than the wild-type or naturally occurring version of the mRNA. As used herein, the terms “stable” and “stability” as such terms relate to the nucleic acids of the present invention, and particularly with respect to the mRNA, refer to increased or enhanced resistance to degradation by, for example nucleases (i.e., endonucleases or exonucleases) which are normally capable of degrading such mRNA. Increased stability can include, for example, less sensitivity to hydrolysis or other destruction by endogenous enzymes (e.g., endonucleases or exonucleases) or conditions within the target cell or tissue, thereby increasing or enhancing the residence of such mRNA in the target cell, tissue, subject and/or cytoplasm. The stabilized mRNA molecules provided herein demonstrate longer half-lives relative to their naturally occurring, unmodified counterparts (e.g. the wild-type version of the mRNA). Also contemplated by the terms “modification” and “modified” as such terms related to the mRNA of the present invention are alterations which improve or enhance translation of mRNA nucleic acids, including for example, the inclusion of sequences which function in the initiation of protein translation (e.g., the Kozak consensus sequence). (Kozak, M., Nucleic Acids Res 15 (20): 8125-48 (1987)).


In some embodiments, the mRNAs used in the compositions of the disclosure have undergone a chemical or biological modification to render them more stable. Exemplary modifications to an mRNA include the depletion of a base (e.g., by deletion or by the substitution of one nucleotide for another) or modification of a base, for example, the chemical modification of a base. The phrase “chemical modifications” as used herein, includes modifications which introduce chemistries which differ from those seen in naturally occurring mRNA, for example, covalent modifications such as the introduction of modified nucleotides, (e.g., nucleotide analogs, or the inclusion of pendant groups which are not naturally found in such mRNA molecules).


Other suitable polynucleotide modifications that may be incorporated into the PE-encoding mRNA used in the compositions of the disclosure include, but are not limited to, 4′-thio-modified bases: 4′-thio-adenosine, 4′-thio-guanosine, 4′-thio-cytidine, 4′-thio-uridine, 4′-thio-5-methyl-cytidine, 4′-thio-pseudouridine, and 4′-thio-2-thiouridine, pyridin-4-one ribonucleoside, 5-aza-uridine, 2-thio-5-aza-uridine, 2-thiouridine, 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxyuridine, 3-methyluridine, 5-carboxymethyl-uridine, 1-carboxymethyl-pseudouridine, 5-propynyl-uridine, 1-propynyl-pseudouridine, 5-taurinomethyluridine, 1-taurinomethyl-pseudouridine, 5-taurinomethyl-2-thio-uridine, 1-taurinomethyl-4-thio-uridine, 5-methyl-uridine, 1-methyl-pseudouridine, 4-thio-1-methyl-pseudouridine, 2-thio-1-methyl-pseudouridine, 1-methyl-1-deaza-pseudouridine, 2-thio-1-methyl-1-deaza-pseudouridine, dihydrouridine, dihydropseudouridine, 2-thio-dihydrouridine, 2-thio-dihydropseudouridine, 2-methoxyuridine, 2-methoxy-4-thio-uridine, 4-methoxy-pseudouridine, 4-methoxy-2-thio-pseudouridine, 5-aza-cytidine, pseudoisocytidine, 3-methyl-cytidine, N4-acetylcytidine, 5-formylcytidine, N4-methylcytidine, 5-hydroxymethylcytidine, 1-methyl-pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine, 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine, 4-thio-1-methyl-pseudoisocytidine, 4-thio-1-methyl-1-deaza-pseudoisocytidine, 1-methyl-1-deaza-pseudoisocytidine, zebularine, 5-aza-zebularine, 5-methyl-zebularine, 5-aza-2-thio-zebularine, 2-thio-zebularine, 2-methoxy-cytidine, 2-methoxy-5-methyl-cytidine, 4-methoxy-pseudoisocytidine, 4-methoxy-1-methyl-pseudoisocytidine, 2-aminopurine, 2,6-diaminopurine, 7-deaza-adenine, 7-deaza-8-aza-adenine, 7-deaza-2-aminopurine, 7-deaza-8-aza-2-aminopurine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyladenosine, N6-methyladenosine, N6-isopentenyladenosine, N6-(cis-hydroxyisopentenyl)adenosine, 2-methylthio-N6-(cis-hydroxyisopentenyl)adenosine, N6-glycinylcarbamoyladenosine, N6-threonylcarbamoyladenosine, 2-methylthio-N6-threonyl carbamoyladenosine, N6,N6-dimethyladenosine, 7-methyladenine, 2-methylthio-adenine, and 2-methoxy-adenine, inosine, 1-methyl-inosine, wyosine, wybutosine, 7-deaza-guanosine, 7-deaza-8-aza-guanosine, 6-thio-guanosine, 6-thio-7-deaza-guanosine, 6-thio-7-deaza-8-aza-guanosine, 7-methyl-guanosine, 6-thio-7-methyl-guanosine, 7-methylinosine, 6-methoxy-guanosine, 1-methylguanosine, N2-methylguanosine, N2,N2-dimethylguanosine, 8-oxo-guanosine, 7-methyl-8-oxo-guanosine, 1-methyl-6-thio-guanosine, N2-methyl-6-thio-guanosine, and N2,N2-dimethyl-6-thio-guanosine, and combinations thereof. The term modification also includes, for example, the incorporation of non-nucleotide linkages or modified nucleotides into the mRNA sequences of the present invention (e.g., modifications to one or both of the 3′ and 5′ ends of an mRNA molecule encoding a functional protein or enzyme). Such modifications include the addition of bases to an mRNA sequence (e.g., the inclusion of a poly A tail or a longer poly A tail), the alteration of the 3′ UTR or the 5′ UTR, complexing the mRNA with an agent (e.g., a protein or a complementary nucleic acid molecule), and inclusion of elements which change the structure of an mRNA molecule (e.g., which form secondary structures).


In some embodiments, PE-encoding mRNAs include a 5′ cap structure. A 5′ cap is typically added as follows: first, an RNA terminal phosphatase removes one of the terminal phosphate groups from the 5′ nucleotide, leaving two terminal phosphates; guanosine triphosphate (GTP) is then added to the terminal phosphates via a guanylyl transferase, producing a 5′5′5 triphosphate linkage; and the 7-nitrogen of guanine is then methylated by a methyltransferase. Examples of cap structures include, but are not limited to, m7G(5′)ppp (5′(A,G(5′)ppp(5′)A and G(5′)ppp(5′)G. Naturally occurring cap structures comprise a 7-methyl guanosine that is linked via a triphosphate bridge to the 5′-end of the first transcribed nucleotide, resulting in a dinucleotide cap of m7G(5′)ppp(5′)N, where N is any nucleoside. In vivo, the cap is added enzymatically. The cap is added in the nucleus and is catalyzed by the enzyme guanylyl transferase. The addition of the cap to the 5′ terminal end of RNA occurs immediately after initiation of transcription. The terminal nucleoside is typically a guanosine, and is in the reverse orientation to all the other nucleotides, i.e., G(5′)ppp(5′)GpNpNp.


Additional cap analogs include, but are not limited to, a chemical structures selected from the group consisting of m7GpppG, m7GpppA, m7GpppC; unmethylated cap analogs (e.g., GpppG); dimethylated cap analog (e.g., m2,7GpppG), trimethylated cap analog (e.g., m2,2,7GpppG), dimethylated symmetrical cap analogs (e.g., m7Gpppm7G), or anti reverse cap analogs (e.g., ARCA; m7,2′OmeGpppG, m72′dGpppG, m7,3′OmeGpppG, m7,3′dGpppG and their tetraphosphate derivatives) (see, e.g., Jemielity, J. et al., “Novel ‘anti-reverse’ cap analogs with superior translational properties”, RNA, 9: 1108-1122 (2003)).


Typically, the presence of a “tail” serves to protect the mRNA from exonuclease degradation. A poly A or poly U tail is thought to stabilize natural messengers and synthetic sense RNA. Therefore, in certain embodiments a long poly A or poly U tail can be added to an mRNA molecule thus rendering the RNA more stable. Poly A or poly U tails can be added using a variety of art-recognized techniques. For example, long poly A tails can be added to synthetic or in vitro transcribed RNA using poly A polymerase (Yokoe, et al. Nature Biotechnology. 1996; 14: 1252-1256). A transcription vector can also encode long poly A tails. In addition, poly A tails can be added by transcription directly from PCR products. Poly A may also be ligated to the 3′ end of a sense RNA with RNA ligase (see, e.g., Molecular Cloning A Laboratory Manual, 2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1991 edition)).


Typically, the length of a poly A or poly U tail can be at least about 10, 50, 100, 200, 300, 400 at least 500 nucleotides. In some embodiments, a poly-A tail on the 3′ terminus of mRNA typically includes about 10 to 300 adenosine nucleotides (e.g., about 10 to 200 adenosine nucleotides, about 10 to 150 adenosine nucleotides, about 10 to 100 adenosine nucleotides, about 20 to 70 adenosine nucleotides, or about 20 to 60 adenosine nucleotides). In some embodiments, mRNAs include a 3′ poly(C) tail structure. A suitable poly-C tail on the 3′ terminus of mRNA typically include about 10 to 200 cytosine nucleotides (e.g., about 10 to 150 cytosine nucleotides, about 10 to 100 cytosine nucleotides, about 20 to 70 cytosine nucleotides, about 20 to 60 cytosine nucleotides, or about 10 to 40 cytosine nucleotides). The poly-C tail may be added to the poly-A or poly U tail or may substitute the poly-A or poly U tail.


PE-encoding mRNAs according to the present disclosure may be synthesized according to any of a variety of known methods. For example, mRNAs according to the present invention may be synthesized via in vitro transcription (IVT). Briefly, IVT is typically performed with a linear or circular DNA template containing a promoter, a pool of ribonucleotide triphosphates, a buffer system that may include DTT and magnesium ions, and an appropriate RNA polymerase (e.g., T3, T7 or SP6 RNA polymerase), DNAse I, pyrophosphatase, and/or RNAse inhibitor. The exact conditions will vary according to the specific application.


In embodiments involving mRNA delivery, the ratio of the mRNA encoding the PE fusion protein to the PEgRNA may be important for efficient editing. In certain embodiments, the weight ratio of mRNA (encoding the PE fusion protein) to PEgRNA is 1:1. In certain other embodiments, the weight ratio of mRNA (encoding the PE fusion protein) to PEgRNA is 2:1. In still other embodiments, the weight ratio of mRNA (encoding the PE fusion protein) to PEgRNA is 1:2. In still further embodiments, the weight ratio of mRNA (encoding the PE fusion protein) to PEgRNA is selected from the group consisting of about 1:1000, 1:900; 1:800; 1:700; 1:600; 1:500; 1:400; 1:300; 1:200; 1:100; 1:90; 1:80; 1:70; 1:60; 1:50; 1:40; 1:30; 1:20; 1:10; and 1:1. In other embodiments, the weight ratio of mRNA (encoding the PE fusion protein) to PEgRNA is selected from the group consisting of about 1:1000, 1:900; 800:1; 700:1; 600:1; 500:1; 400:1; 300:1; 200:1; 100:1; 90:1; 80:1; 70:1; 60:1; 50:1; 40:1; 30:1; 20:1; 10:1; and 1:1.


F. Use of Multi-Flap Prime Editing for Insertion of Inducible Dimerization Domains


The multi-flap prime editors (as represented by FIGS. 90, 95, 97A, 98A) described herein may also be used to install dimerization domains into one or more protein targets. The dimerization domains may facilitate inducible regulation of the activity associated with the dimerization of the one or more protein targets via a linking moiety (e.g., a small molecule, peptide, or protein) that binds in a bi-specific manner. In various aspects, the dimerization domains, when installed on different proteins (e.g., the same type or different proteins), each bind to the same bi-specific moiety (e.g., a bi-specific small molecule, peptide, or polypeptide having a least two regions that separately bind to the dimerization domains), thereby causing the dimerization of the proteins through their common interaction to the bi-specific ligand. In this manner, the bi-specific ligand functions as an “inducer” of dimerization of two proteins. In some cases, the bi-specific ligand or compound will have two targeting moieties that are the same. In other embodiments, the bi-specific ligand or compound will have targeting moieties that are each different from the other. The bi-specific ligand or compound having the same two targeting moieties will be able to target the same dimerization domain installed on different protein targets. The bi-specific ligand or compound having different targeting moieties will be able to target different dimerization domains installed on different protein targets.


As used herein, the term “dimerization domain” refers to a ligand-binding domain that binds to a binding moiety of a bi-specific ligand. A “first” dimerization domain binds to a first binding moiety of a bi-specific ligand and a “second” dimerization domain binds to a second binding moiety of the same bi-specific ligand. When the first dimerization domain is fused to a first protein (e.g., via PE, as discussed herein) and the second dimerization domain (e.g., via PE, as discussed herein) is fused to a second protein, the first and second protein dimerize in the presence of a bi-specific ligand, wherein the bi-specific ligand has at least one moeity that binds to the first dimerization domain and at least another moiety that binds to the second dimerization domain.


The term “bi-specific ligand” or “bi-specific moiety,” as used herein, refers to a ligand that binds to two different ligand-binding domains. In various embodiments, the bi-specific moiety itself is a dimer of two of same or two different chemical moieties, wherein each moiety specifically and tightly binds to a dimerization domain. In certain embodiments, the ligand is a small molecule compound, or a peptide, or a polypeptide. In other embodiments, ligand-binding domain is a “dimerization domain,” which can be install as a peptide tag onto a protein. In various embodiments, two proteins each comprising the same or different dimerization domains can be induced to dimerize through the binding of each dimerization domain to the bi-specific ligand. These molecules may also be referred to as “chemical inducers of dimerization” or CIDs. In addition, the bi-specific ligands may be prepared by coupling (e.g., through standardize chemical linkages) two of the same moieties together, or two different moieties together, wherein each moiety tightly and specifically binds to a dimerization domain.


In various aspects, the dimerization domains installed by multi-flap PE can be the same or different.


For example, the dimerization domains can be FKBP12, which has the following amino acid sequence:











FKBP12



(SEQ ID NO: 488)



MGVQVETISPGDGRTFPKRGQTCVVHYTGML







EDGKKFDSSRDRNKPFKFMLGKQEVIRGWEE







GVAQMSVGQRAKLTISPDYAYGATGHPGIIPPH







ATLVFDVELLKLE






In another example, the dimerization domain can be a mutant of FKBP12 referred to as FKBP12-F36V, a mutant of FKBP12 with an engineered hole that binds a synthetic bumped FK506 mimic (2, FIG. 3)107:











FKBP12-F36V



(SEQ ID NO: 489)



MGVQVETISPGDGRTFPKRGQTCVVHYTGML







EDGKKVDSSRDRNKPFKFMLGKQEVIRGWEE







GVAQMSVGQRAKLTISPDYAYGATGHPGIIPPH







ATLVFDVELLKLE






In another example, the dimerization domain can be cyclophilin, as follows:











CYCLOPHILIN



(SEQ ID NO: 490)



MVNPTVFFDIAVDGEPLGRVSFELFADKVPKT







AENFRALSTGEKGFGYKGSCFHRIIPGFMCQG







GDFTRHNGTGGKSIYGEKFEDENFILKHTGPGI







LSMANAGPNTNGSQFFICTAKTEWLDGKHVV







FGKVKEGMNIVEAMERFGSRNGKTSKKITIAD







CGQLE






In various embodiments, the amino acid sequences of these dimerization domains may be altered in order to optimize binding or to improve binding orthogonality to native targets. The nucleic acid sequences of the genes encoding small-molecule binding proteins may be altered in order to optimize the efficiency of the PE process, such as by reducing PEgRNA secondary structure.


Other examples of suitable dimerization domains and a cognate small molecule compound which binds thereto are provided as follows. Note that the cognate small molecule compound could be coupled (e.g., via a chemical linker) to a second small molecule compound (either the same compound or a different compound) in order to form a bi-specific ligand that may bind two dimerization domains. In some cases, such as FK506 and cyclosporin A, dimerization of each (e.g., FK506-FK506 or cyclosporin A-cyclosporin A) reduces or eliminates immunosuppressive activity of the monomeric compounds.













SMALL MOLECULE-BINDS TO THE DIMERIZATION DOMAIN-



A DIMER OF THESE MOLECULES WOULD CONSTITUTE A BI-SPECIFIC



LIGAND THAT WOULD BIND TWO DIMERIZATION DOMAINS
DIMERIZATION DOMAIN(S)









embedded image


AMINO ACID SEQUENCE OF FKBP12: MGVQVETISPGDGRTFPKRGQTCVVHYTG MLEDGKKFDSSRDRNKPFKFMLGKQEVIR GWEEGVAQMSVGQRAKLTISPDYAYGATG HPGIIPPHATLVFDVELLKLE (SEQ ID NO: 491) CALCINEURIN:







embedded image


AMINO ACID SEQUENCE OF HUMAN CYCLOPHILIN A: MVNPTVFFDIAVDGEPLGRVSFELFAD KVPKTAENFRALSTGEKGFGYKGSCFH RIIPGFMCQGGDFTRHNGTGGKSIYGE KFEDENFILKHTGPGILSMANAGPNTN GSQFFICTAKTEWLDGKHVVFGKVKE GMNIVEAMERFGSRNGKTSKKITIADC GQLE (SEQ ID NO: 490) AMINO ACID SEQUENCE OF HUMAN CYCLOPHILIN B: MLRLSERNMKVLLAAALIAGSVFFLLL PGPSAADEKKKGPKVTVKVYFDLRIG DEDVGRVIFGLFGKTVPKTVDNFVALA TGEKGFGYKNSKFHRVIKDFMIQGGDF TRGDGTGGKSIYGERFPDENFKLKHYG PGWVSMANAGKDTNGSQFFITTVKTA WLDGKHVVFGKVLEGMEVVRKVEST KTDSRDKPLKDVIIADCGKIEVEKPFAI AKE (SEQ ID NO: 493) AMINO ACID SEQUENCE OF MURINE CYCLOPHILIN C: MSPGPRLLLPAVLCLGLGALVSSSGSSG VRKRGPSVTDKVFFDVRIGDKDVGRIV IGLFGNVVPKTVENFVALATGEKGYGY KGSIFHRVIKDFMIQGGDFTARDGTGG MSIYGETFPDENFKLKHYGIGWVSMA NAGPDTNGSQFFITLTKPTWLDGKHVV FGKVLDGMTVVHSIELQATDGHDRPLT DCTIVNSGKIDVKTPFVVEVPDW (SEQ ID NO: 494)




embedded image


AMINO ACID SEQUENCE OF FKBP12-F36V MGVQVETISPGDGRTFPKRGQTCVVHYTG MLEDGKKVDSSRDRNKPFKFMLGKQEVIR GWEEGVAQMSVGQRAKLT ISPDYAYGATGHPGIIPPHATLVFDVELLKL E (SEQ ID NO: 495)







embedded image


AMINO ACID SEQUENCE OF HUMAN DIHYDROFOLATE REDUCTASE MVGSLNCIVAVSQNMGIGKNGDLPWPPLR NEFRYFQRMTTTSSVEGKQNLVIMGKKTW FSIPEKNRPLKGRINLVLSRELKEPPQGAHF LSRSLDDALKLTEQPELANKVDMVWIVGG SSVYKEAMNHPGHLKLFVTRIMQDFESDT FFPEIDLEKYKLLPEYPGVLSDVQEEKGIK YKFEVYEKND (SEQ ID NO: 496)







embedded image


AMINO ACID SEQUENCE FOR E. COLI DIHYDROFOLATE REDUCTASE MISLIAALAVDRVIGMENAMPWNLPADLA WFKRNTLNKPVIMGRHTWESIGRPLPGRK NIILSSQPGTDDRVTWVKSVDEAIAACGDV PEIMVIGGGRVYEQFLPKAQKLYLTHIDAE VEGDTHFPDYEPDDWESVFSEFHDADAQN SHSYCFEILERR (SEQ ID NO: 497)







embedded image


AMINO ACID SEQUENCE FOR HUMAN GLUCOCORTICOID RECEPTOR MDSKESLTPGREENPSSVLAQERGDVMDF YKTLRGGATVKVSASSPSLAVASQSDSKQR RLLVDFPKGSVSNAQQPDLSKAVSLSMGL YMGETETKVMGNDLGFPQQGQISLSSGET DLKLLEESIANLNRSTSVPENPKSSASTAVS AAPTEKEF PKTHSDVSSEQQHLKGQTGTNGGNVKLYT TDQSTFDILQDLEFSSGSPGKETNESPWRS DLLIDENCLLSPLAGEDDSFLLEGNSNEDC KPLILPDTKPKIKDNGDLVLSSPSNVTLPQV KTEKEDFIELCTPGVIKQEKLGTVYCQASF PGANIIGNKMSAISVHGVSTSGGQMYHYD MNTASLSQQQDQKPIFNVIPPIPVGSENWN RCQGSGDDNLTSLGTLNFPGRTVFSNGYSS PSMRPDVSSPPSSSSTATTGPPPKLCLVCSD EASGCHYGVLTCGSCKVFFKRAVEGQHNY LCAGRNDCIIDKIRRKNCPACRYRKCLQAG



MNLEARKTKKKIKGIQQATTGVSQETSEN



PGNKTIVPATLPQLTPTLVSLLEVIEPEVLYA



GYDSSVPDSTWRIMTTLNMLGGRQVIAAV



KWAKAIPGFRNLHLDDQMTLLQYSWMFL



MAFALGWRSYRQSSANLLCFAPDLIINEQR



MTLPCMYDQCKHMLYVSSELHRLQVSYE



EYLCMKTLLLLSSVPKDGLKSQELFDEIRM



TYIKELGKAIVKREGNSSQNWQRFYQLTK



LLDSMHEVVENLLNYCFQTFLDKTMSIEFP



EMLAEIITNQIPKYSNGNIKKLLFHQK (SEQ



ID NO: 498)




embedded image


AMINO ACID SEQUENCE OF FKBP12: MGVQVETISPGDGRTFPKRGQTCVVHYTG MLEDGKKFDSSRDRNKPFKFMLGKQEVIR GWEEGVAQMSVGQRAKLTISPDYAYGATG HPGIIPPHATLVFDVELLKLE (SEQ ID NO: 491)









Other examples of naturally occurring bifunctional molecules and their dual target receptors are as follows. Prime editing may be used to install the dual target receptors into different proteins. Once the different proteins are modified by PE to contain a bifunctional molecule receptor, the bifunctional molecules may be introduced, thereby causing the dimerization of the proteins modified to comprise the different dimerization domains. Examples of pairings of (1) a biofunctional molecule and (2) their dual target receptors are as follows:

















TARGET 



NATURALLY 
RECEPTORS



OCCURRING 
OF THE 



BIOFUNCTIONAL
BIOFUNCTIONAL 



MOLECULES
MOLECULE











embedded image


Target receptor 1: auxin receptor Target receptor 2: TIR1 E3 ligase









embedded image


Target receptor 1:  JAZ receptor Target receptor 2:  Col1 E3 ligase









embedded image


target receptor 1:  GBF1 target receptor 2:  GTPase Arf1p









embedded image


target receptor 1:  PYR receptor target receptor 2:  phosphoprotein phosphatase 2C









embedded image


target receptor 1:  adenylyl cyclase  monomers target receptor 2:  adenylyl cyclase  monomers









embedded image


target receptor 1:  14-3-3 proteins target receptor 2:  H+-ATPase









embedded image


target receptor 1:  FKBP12 target receptor 2:  mTOR









embedded image


target receptor 1:  cyclophilin target receptor 2:  IMP dehydrogenase 2









embedded image


target receptor 1:  cyclophilin target receptor 2:  calcineurin










Examples of other bifunctional molecules that can be used with this aspect of prime editing are as follows:


Synstab A:




embedded image


Paclitaxel:




embedded image


Discodermolide:




embedded image


Synstab A, paclitaxel, and discodermolide are microtubule stabilizers. Thus, these compounds could be used to dimerize proteins modified by PE to comprise microtubule proteins. GNE-0011, ARV-825, and dBET1 comprise a BRD4 binding motif and a CRBN binding motif. Thus, these compounds could be used to dimerize proteins modified by PE to comprise these targeting domains.


The PEgRNAs for installing dimerization domains may comprising the following structures (in reference to FIG. 3D):

    • 5′-[spacer]-[gRNA core]-[extension arm]-3′, wherein the extension arm comprises 5′-[homology arm]-[edit template]-[primer binding site]-3′; or
    • 5′-[extension arm]-[spacer]-[gRNA core]-3′, wherein the extension arm comprises 5′-[homology arm]-[edit template]-[primer binding site]-3′, and wherein with either configuration the “edit template” comprises a nucleotide sequence of a dimerization domain.


In one example, the PEgRNA for insertion of the FKBP12 dimerization domain at the C-terminal end of human insulin receptor (spacer underlined, gRNA core plain, flap homology bold, FKBP12 insertion in italics, annealing region bold italics):











PEGRNA FOR INSTALLING FKBP12 IN HUMAN INSULIN



RECEPTOR



(SEQ ID NO: 499)




CACGGUAGGCACUGUUAGGAGUUUUAGAG








CUAGAAAUAGCAAGUUAAAAUAAGGCUAG







UCCGUUAUCAACUUGAAAAAGUGGCACCG







AGUCGGUGCUUGCCUCGGUCCAAUCCUUC








C
GGAGUGCAGGUGGAAACCAUCUCCCCAGGA









GACGGGCGCACCUUCCCCAAGCGCGGCCAGA









CCUGCGUGGUGCACUACACCGGGAUGCUUG









AAGAUGGAAAGAAAUUUGAUUCCUCCCGGGA









CAGAAACAAGCCCUUUAAGUUUAUGCUAGGC









AAGCAGGAGGUGAUCCGAGGCUGGGAAGAAG









GGGUUGCCCAGAUGAGUGUGGGUCAGAGAG









CCAAACUGACUAUAUCUCCAGAUUAUGCCUA









UGGUGCCACUGGGCACCCAGGCAUCAUCCCA









CCACAUGCCACUCUCGUCUUCGAUGUGGAG









CUUCUAAAACUGGAAcustom character







In another example, the PEgRNA for insertion of the FKBP12 dimerization domain at the HEK3 locus (for optimization):











PEGRNA FOR INSTALLING FKBP12 IN HEK3



(SEQ ID NO: 500)



GGCCCAGACTGAGCACGTGAGTTTTAGAGC







TAGAAATAGCAAGTTAAAATAAGGCTAGTC







CGTTATCAACTTGAAAAAGTGGCACCGAGT







CGGTGCTGGAGGAAGCAGGGCTTCCTTTCCT







CTGCCATCATTCCAGTTTTAGAAGCTCCACA







TCGAAGACGAGAGTGGCATGTGGTGGGATG







ATGCCTGGGTGCCCAGTGGCACCATAGGCA







TAATCTGGAGATATAGTCAGTTTGGCTCTCT







GACCCACACTCATCTGGGCAACCCCTTCTTC







CCAGCCTCGGATCACCTCCTGCTTGCCTAGC







ATAAACTTAAAGGGCTTGTTTCTGTCCCGGG







AGGAATCAAATTTCTTTCCATCTTCAAGCAT







CCCGGTGTAGTGCACCACGCAGGTCTGGCC







GCGCTTGGGGAAGGTGCGCCCGTCTCCTGG







GGAGATGGTTTCCACCTGCACTCCCGTGCTC







AGTCTG






The target proteins for installing dimerization domains are not particularly limited; however, it is advantageous their dimerization (once modified by PE) in the presence of a bi-specific ligand produces some advantageous biological effect, e.g., a signaling pathway, decreased immunoresponsiveness, etc. In various aspects, the target proteins that are to be dimerized through the PE-dependent installation of dimerization domains can be the same protein or different proteins. Preferably, the proteins, when dimerized, trigger one or more downstream biological cascades, e.g., a signal transduction cascade, phorsphorylation, etc. Exemplary target proteins into which PE may be used to install dimerization domains, include, but are not limited to:
















MEMBRANDE-
KINASE





BOUND
DOMAIN
CID
SIGNALING



RECEPTOR
FUSED TO
EMPLOYED
CASCADE
REFERENCE







T-CELL
FKBP12
FK1012
T-CELL

SCIENCE 262, 1019-24



RECEPTOR

(FK506
RECEPTOR
(1993)




DIMER)
SIGNALING

CHEM. BIOL. 1, 163-172







(1994).


FAS RECEPTOR
MURINE
CYCLO-
FAS PATHWAY

CHEM. BIOL. 3, 731-738




CYCLO-
SPORIN A
FOR
(1996).



PHILIN C
DIMER
APOPTOSIS



INSULIN
FKBP12
FK1012
INSULIN

CURR. BIOL. 8, 11-18



RECEPTOR

(FK506
SIGNALING
(1998).




DIMER)




PLATELET-
FKBP12
FK1012
PDGF

CURR. BIOL. 8, 11-18



DERIVED

(FK506
MESODERM
(1998).


GROWTH

DIMER)
FORMATION



FACTOR (PDGF)


SIGNALING



BETA






ERYTHRO-
FKBP12
FK1012
EPOR-

PROC. NATL. ACAD.



POIETIN

(FK506
MEDIATED

SCI. 94, 3076-3081 (2002).



RECEPTOR

DIMER)
PROLIFERATIVE



(EPOR)


SIGNALING









In one aspect, the multi-flap prime editor systems described herein may be used to install sequences encoding dimerization domains into one or more genes encoding target proteins of interest in a living cell or patient. This may be referred to as the “multi-flap prime editing—CID system,” wherein the CID is the bi-specific ligand that induced dimerization of target proteins, each fused to a dimerization domain installed by multi-flap PE. This edit alone should have no physiological effect. Upon administration of a bi-specific ligand, which typically is a dimeric small molecule that can simultaneously bind to two dimerization domains, each of which is fused to a copy of the target protein, the bi-specific ligand causes dimerization of the targeted protein. This target protein dimerization event then induces a biological signaling event, such as erythropoiesis or insulin signaling. A new method to place dimerization-induced biological processes, such as receptor signaling, under control of a convenient small-molecule drug (i.e., the bi-specific ligand) by the genomic integration of genes encoding small-molecule binding proteins (i.e., the dimerization domains) with multi-flap prime editing is described herein.


Protein dimerization is a ubiquitous biological process. Notably, homodimerization of many membrane-bound receptors is known to initiate signaling cascades, often with profound biological consequences. A number of small-molecule natural products approved for use as drugs act as chemical inducers of protein dimerization as part of their mechanism of action.92 For example, FK506 binds tightly to FKBP12, and the resulting small molecule—protein complex then binds the phosphatase calcineurin, thereby inhibiting a step in T cell receptor signaling.93 Likewise, cyclosporin A induces dimerization of cyclophilin and calcineurin, and rapamycin induces dimerization of FKBP and mTOR.93,94


In one embodiment, leveraging the selective, high-affinity binding of the FK506: FKBP12 and cyclosporin A:cyclophilin small molecule:protein binding interaction, synthetic chemical inducers of dimerization have also been developed. In an example, a small molecule comprised of two units of FK506, termed FK1012, was shown to effect signal transduction when the cytoplasmic domains of signaling receptors were tagged with FKBP12.95 Chemical inducers of dimerization (CIDs) have since been used to control a number of signaling pathways.96-103


While useful tools for studying biological processes, one challenge facing synthetic CIDs for therapeutic applications is that introduction of the FKBP12—or cyclophilin—target protein chimeras into patients is challenging.


The present disclosure brings together multiple concepts to create a previously inaccessible therapeutic process. The first concept is multi-flap prime editing (e.g., dual flap or quadruple flap prime editing), described herein, which allows for precise genome editing, including targeted insertions, in living cells. The second concept is chemical-induced dimerization, a powerful tool that has enabled small-molecule control over signaling and oligomerization processes in cell culture.


Specific cases in which chemical control over protein dimerization may have had a beneficial therapeutic effect have been identified.


The insulin receptor is a heterotetrameric transmembrane protein that responds to insulin binding to the extracellular domain by phosphorylation of the cytoplasmic kinase domain.104 An engineered chimeric protein composed of a membrane-localization component, the C-terminal kinase domain of the insulin receptor, and three copies of FKBP12 responds to FK1012 and initiates the insulin response in cell culture.99 Similarly, it is expected that the fusion of FKBP12 to the C-terminal end of the kinase domain of the native insulin receptor in patient cells should allow for FK1012-dependent phosphorylation and initiation of the insulin signaling cascade. This system could replace or complement insulin use in patients who cannot make insulin (e.g., type-1 diabetics), or who respond weakly to insulin (e.g., type-2 diabetics).


Additionally, erythropoietin stimulates erythrocyte proliferation by binding to the erythropoietin receptor (EpoR), either inducing dimerization or a conformational change in a preformed receptor dimer which results in activation of the Jak/STAT signaling cascade.105 It has been demonstrated that FK1012-induced oligomerization of the membrane-anchored cytoplasmic domain of EpoR tagged with FKBP12 is sufficient to initiate the Jak/STAT signaling cascade and promote cell proliferation.106 It is anticipated that fusing FKBP12 to native EpoR by prime editing in patient cells will allow for FK1012-induced control over erythrocyte proliferation (erythropoiesis). This system could be used to trigger red blood cell growth in anemic patients. FK1012-inducible EpoR could also be employed as an in vivo selectable marker for blood cells that have undergone ex vivo engineering.


In principle, any receptor tyrosine kinase could be viable target for a multi-flap prime editing—CID therapeutic. The table below includes a list of all receptor tyrosine kinases in the human genome.110




















NT
PROT



Family
Receptor
Synonyms
Accession
Accession
Chromosome







ALK
ALK
Ki1
NM_004304
NP_004295
2p23


family
LTK
TYK1
NM_002344
NP_002335
15q15.1-q21.1


AXL
AXL
UFO, Tyro7(r) Ark(m)
NM_001699
NP_001690
19q13.1


family
MER
MERTK, NYK, Eyk(ch)
NM_006343
NP_006334
2q14.1



TYRO3
RSE, SKY, BRT, DTK,
NM_006293
NP_006284
15q15.1-q21.1




TIF





DDR
DDR1
CAK, TRKE, NEP
NM_013993
NP_001945
6p21.3


family

NTRK4, EDDR1, PTK3






DDR2
TKT, TYRO10,
NM_006182
NP_006173
1q21-q22




NTRKR3





EGFR
EGFR
ERBB, ERBB1
NM_005228
NP_005219
7p12


family
ERBB2
HER2, Neu(r), NGL
NM_004448
NP_004439
17q11.2-q12



ERBB3
HER3
NM_001982
NP_001973
12q13



ERBB4
HER4
NM_005235
NP_005226
2q33.3-q34


EPH
EPHA1
EPH, EPHT
NM_005232
NP_005223
7q32-q36


family
EPHA2
ECK, Sek(m), Myk2(m)
NM_004431
NP_004422
1p34



EPHA3
HEK, ETK1, Tyro4(r),
NM_005233
NP_005224
3p11.2




Mek4(m), Cek4(ch)






EPHA4
HEK8, Tyro1(r),
NM_004438
NP_004429
2q36qter




Sek1(m), Cek8(ch)






EPHA5
HEK7, Ehk(r), Bsk(r),
L36644
P54756





Cek7(ch)






EPHA6
DKFZp434C1418,
AL133666






Ehk2(r)






EPHA7
HEK11, Mdk1(m),
NM_004440
NP_004431
6q21




Ebk(m), Ehk3(r),







Cek11(ch)






EPHA8
HEK3, KIAA1459,
AB040892
CAB81612
1q23-q24




Eek(r), Cek10(ch)






EPHB1
NET, EPHT2, HEK6,
NM_004441
NP_004432
3q21-q23




Elk(r), Cek6(ch)






EPHB2
HEK5, ERK, DRT,
AF025304
AAB94602
1p36.1-p35




EPHT3, Tyro5(r),







Nuk(m), Sek3(m),







Cek5(ch)






EPHB3
HEK2, Tyro6, Mdk5(m),
NM_004443
NP_004434
3q21-qter




Sek4(m)






EPHB4
HTK, Tyro11(r),
NM_004444
NP_004435





Mdk2(m), Myk1(m)






EPHB6
HEP, Mep(m), Cek1(ch)
NM_004445
NP_004436
7q33-q35


FGFR
FGFR1
FLT2, bFGFR, FLG, N-
M34641
AAA35835
8p11.2


family

SAM






FGFR2
KGFR, K-SAM, Bek(m),
NM_000141
NP_000132
10q26




CFD1, JWS, Cek3(ch)






FGFR3
HBGFR, ACH, Cek2(ch)
NM_000142
NP_000133
4p16.3



FGFR4

NM_002011
NP_002002
5q35.1-qter


INSR
IGF1R
JTK13
NM_000875
NP_000866
15q25-q26


family
INSR
IR
NM_000208
NP_000199
19p13.3-p13.2



INSRR
IRR
J05046
AAC31759
1q21-q23


MET
MET
HGFR
NM_000245
NP_000236
7q31


Family
RON
MST1R, CDw136,
NM_002447
NP_002438
3p21.3




Fv2(m), STK(m),







SEA(ch)





MUSK
MUSK
Nsk2(m), Mlk1(m),
NM_005592
NP_005583
9q31.3-q32


family

Mk2(m)





PDGFR
CSF1R
FMS, C-FMS, CD115
NM_005211
NP_005202
5q31-q32


family
FLT3
FLK2, STK1, CD135
NM_004119
NP_0041110
13q12



KIT
Sfr(m), CKIT
NM_000222
NP_000213
4q11-q12



PDGFRA

NM_006206
NP_006197
4q11-q13



PDGFRB
PDGFR, JTK12
NM_002609
NP_002600
5q31-q32


PTK7
PTK7
CCK4, KLG(ch)
NM_002821
NP_002812
6p21.2-p12.2


family







RET
RET
MEN2A/B, HSCR1,
X12949
P07949
10q11.2


family

MTC1





ROR
ROR1
NTRKR1
NM_005012
NP_005003
1p32-p31


family
ROR2
NTRKR2
NM_004560
NP_004551



ROS
ROS1
MCF3
NM_002944
NP_002935
6q22


family







RYK
RYK
Vlk(m), Mrk(m)
S59184
AAB263411
3q22


family







TIE family
TEK
TIE2
NM_000459
NP_000450
9p21



TIE
TIE1, JTK14
NM_005424
NP_005415
1p34-p33


TRK
NTRK1
TRK, TRKA
NM_002529
NP_002520
1q21-q22


family
NTRK2
TRKB
NM_006180
NP_006171
9q22.1



NTRK3
TRKC
NM_002530
NP_002521
15q25


VEGFR
VEGFR1
FLT1
NM_002019
NP_002010
13q12


family
VEGFR2
KDR, FLK1

AAB88005
4q11-q12



VEGFR3
FLT4, PCL
NM_002020
NP_002011
5q34-q35


AATYK
AATYK
AATK, KIAA0641
NM_004920
NP_004911
17q25.3


family
AATYK2
KIAA1079
NM_014916
NP_055731
7q21-q22



AATYK3



19q13.2-q13.3









There are numerous advantages to the multi-flap prime editing—CID system. One such advantage is that it can replace endogenous ligands, which are typically proteins that pose complications in manufacturing, cost, delivery, production, or storage, with drug-like small-molecules that can be orally administered instead of administered by IV or injection, are readily prepared from FDA-approved drugs (or are themselves already drugs), and do not incur special production or storage costs typically associated with protein drugs. Another advantage is that the edit alone should have no physiological effect. The amount of target protein dimerization can be controlled by dosing the small-molecule CID. Further, target protein dimerization is readily and rapidly reversible by adding the monomeric form of the CID. Yet another advantage is that in instances where a single ligand targets multiple receptors, selectivity can be achieved by prime-editing only one receptor. Finally, depending on the delivery method used for prime editing, it may also be possible to restrict editing to a localized tissue or organ, allowing for inducible receptor activation only in specific areas.


If editing efficiencies are high enough with multi-flap prime editing that two separate editing events could occur at high levels, it would also be possible to tag two proteins of interest with different small-molecule binding domains (such as FKBP and cyclophilin) and induce heterodimerizations with small molecule heterodimers (such as an FK506-cyclosporin A dimer).


The fusion of FKBP12 or other small-molecule binding proteins to native proteins has been accomplished, generally by overexpression from plasmid in tissue culture. Subsequent chemical-induced dimerization has been demonstrated to induce phenotypic changes to cells producing the fusion proteins.


The following references are cited in the above section and are incorporated herein by reference.

  • 1. Crabtree, G. R. & Schreiber, S. L. Three-part inventions: intracellular signaling and induced proximity Trends Biochem. Sci. 21, 418-22 (1996).
  • 2. Liu, J. et al. Calcineurin Is a Common Target of A and FKBP-FK506 Complexes. Cell 66, 807-815 (1991).
  • 3. Keith, C. T. et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature 369, 756-758 (2003).
  • 4. Spencer, D. M., Wandless, T. J., Schreiber, S. L. S. & Crabtree, G. R. Controlling signal transduction with synthetic ligands. Science 262, 1019-24 (1993).
  • 5. Pruschy, M. N. et al. Mechanistic studies of a signaling pathway activated by the organic dimerizer FK1012. Chem. Biol. 1, 163-172 (1994).
  • 6. Spencer, D. M. et al. Functional analysis of Fas signaling in vivo using synthetic inducers of dimerization. Curr. Biol. 6, 839-847 (1996).
  • 7. Belshaw, P. J., Spencer, D. M., Crabtree, G. R. & Schreiber, S. L. Controlling programmed cell death with a cyclophilin-cyclosporin-based chemical inducer of dimerization. Chem. Biol. 3, 731-738 (1996).
  • 8. Yang, J. X., Symes, K., Mercola, M. & Schreiber, S. L Small-molecule control of insulin and PDGF receptor signaling and the role of membrane attachment. Curr. Biol. 8, 11-18 (1998).
  • 9. Belshaw, P. J., Ho, S. N., Crabtree, G. R. & Schreiber, S. L. Controlling protein association and subcellular localization with a synthetic ligand that induces heterodimerization of proteins. Proc.Natl. Acad. Sci. 93, 4604-4607 (2002).
  • 10. Stockwell, B. R. & Schreiber, S. L. Probing the role of homomeric and heteromeric receptor interactions in TGF-β signaling using small molecule dimerizers. Curr. Biol. 8, 761-773 (2004).
  • 11. Spencer, D. M., Graef, I., Austin, D. J., Schreiber, S. L. & Crabtree, G. R. A general strategy for producing conditional alleles of Src-like tyrosine kinases. Proc. Natl. Acad. Sci. 92, 9805-9809 (2006).
  • 12. Holsinger, L. J., Spencer, D. M., Austin, D. J., Schreiber, S. L. & Crabtree, G. R. Signal transduction in T lymphocytes using a conditional allele of Sos. Proc. Natl. Acad. Sci. 92, 9810-9814 (2006).
  • 13. Myers, M. G. Insulin Signal Transduction and the IRS Proteins. Annu. Rev. Pharmacol. Toxicol. 36, 615-658 (1996).
  • 14. Watowich, S. S. The erythropoietin receptor: Molecular structure and hematopoietic signaling pathways. J. Investig. Med. 59, 1067-1072 (2011).
  • 15. Blau, C. A., Peterson, K. R., Drachman, J. G. & Spencer, D. M. A proliferation switch for genetically modified cells. Proc. Natl. Acad. Sci. 94, 3076-3081 (2002).
  • 16. Clackson, T. et al. Redesigning an FKBP-ligand interface to generate chemical dimerizers with novel specificity. Proc. Natl. Acad. Sci. 95, 10437-10442 (1998).
  • 17. Diver, S. T. & Schreiber, S. L. Single-step synthesis of cell-permeable protein dimerizers that activate signal transduction and gene expression. J. Am. Chem. Soc. 119, 5106-5109 (1997).
  • 18. Guo, Z. F., Zhang, R. & Liang, F. Sen. Facile functionalization of FK506 for biological studies by the thiol-ene ‘click’ reaction. RSC Adv. 4, 11400-11403 (2014).
  • 19. Robinson, D. R., Wu, Y.-M. & Lin, S.-F. The protein tyrosine kinase family of the human genome. Oncogene 19, 5548-5557 (2000).


G. Use of Multi-Flap Prime Editing for Inserting Recombinase Target Sites


In another aspect, multi-flap prime editing (e.g., as represented by FIGS. 90, 95, 97A, 98A) may be used to insert recombinase sites (or “recombinase recognition sequences”) into a desired genomic site. Insertion of recombinase sites provides a programmed location for effecting site-specific genetic changes in a genome. Such genetic changes can include, for example, genomic integration of a plasmid, genomic deletion or insertion, chromosomal translocations, and cassette exchanges, among other genetic changes. These exemplary types of genetic changes are illustrated in FIG. 64(b)-(f). The installed recombinase recognition sequences may then be used to conduct site-specific recombination at that site to effectuate a variety of recombination outcomes, such as, excision, integration, inversion, or exchange of DNA fragments. For example, FIG. 65 illustrates the installation of a recombinase site that can then be used to integrate a DNA donor template comprising a GFP expression marker. Cells containing the integrated GFP expression system into the recombinase site will fluoresce.


The mechanism of installing a recombinase site into the genome is analogous to installing other sequences, such as peptide/protein and RNA tags, into the genome. A schematic exemplifying the installation of a recombinase target sequence is shows in FIG. 64(a). The process begins with selecting a desired target locus into which the recombinase target sequence will be introduced. Next, a prime editor fusion is provided (“RT-Cas9:gRNA”). Here, the “gRNA” refers to a PEgRNA, which can be designed using the principles described herein. The PEgRNA in various embodiments will comprise an architecture corresponding to FIG. 3D (5′-[˜20-nt spacer]-[gRNA core]-[extension arm]-3′, wherein the extension arm comprises in the 3′ to 5′ direction, a primer binding site (“A”), an edit template (“B”), and a homology arm (“C”). The edit template (“B”) will comprise a sequence corresponding to a recombinase site, i.e., a single strand RNA of the PEgRNA that codes for a complementary single strand DNA that is either the sense or the antisense strand of the recombinase site and which is incorporated into the genomic DNA target locus through the multi-flap prime editing process.


In various aspects, the present disclosure provides for the use of a multi-flap PE to introduce recombinase recognition sequences at high-value loci in human or other genomes, which, after exposure to site-specific recombinase(s), will direct precise and efficient genomic modifications (FIG. 64). In various embodiments show in FIG. 64, PE may be used to (b) insert a single SSR target for use as a site for genomic integration of a DNA donor template. (c) shows how a tandem insertion of SSR target sites can be used to delete a portion of the genome. (d) shows how a tandem insertion of SSR target sites can be used to invert a portion of the genome. (e) shows how the insertion of two SSR target sites at two distal chromosomal regions can result in chromosomal translocation. (f) shows how the insertion of two different SSR target sites in the genome can be used to exchange a cassette from a DNA donor template. Each of the types of genome modifications are envisioned by using PE to insert SSR targets, but this list also is not meant to be limiting.


Multi-flap PE-mediated introduction of recombinase recognition sequences could be particularly useful for the treatment of genetic diseases which are caused by large-scale genomic defects, such as gene loss, inversion, or duplication, or chromosomal translocation1-7 (Table 6). For example, Williams-Beuren syndrome is a developmental disorder caused by a deletion of 24 in chromosome 721. No technology exists currently for the efficient and targeted insertion of multiple entire genes in living cells (the potential of PE to do such a full-length gene insertion is currently being explored but has not yet been established); however, recombinase-mediated integration at a target inserted by PE offers one approach towards a permanent cure for this and other diseases. In addition, targeted introduction of recombinase recognition sequences could be highly enabling for applications including generation of transgenic plants, animal research models, bioproduction cell lines, or other custom eukaryotic cell lines. For example, recombinase-mediated genomic rearrangement in transgenic plants at multi-flap PE-specific targets could overcome one of the bottlenecks to generating agricultural crops with improved properties8,9.









TABLE 6







Examples of genetic diseases linked to large-scale


genomic modifications that could be repaired through


multi-flap PE-based installation of recombinase


recognition sequences.










DISEASE
CAUSE







TRISOMY 17P
GENE DUPLICATION



CHARCOT-MARIE-TOOTH
GENE DUPLICATION



DISEASE TYPE I




SMITH-MAGENIS
GENE DELETION



SYNDROME




WILLIAMS-BEUREN
GENE DELETION



SYNDROME




DE LA CHAPELLE
CHROMOSOMAL



SYNDROME
TRANSLOCATION



DOWN SYNDROME
CHROMOSOMAL



(SOME FORMS)
TRANSLOCATION



HEMOPHILIA A
GENE INVERSION



HUNTER SYNDROME
GENE INVERSION










A number of SSR family members have been characterized and their recombinase recognition sequences described, including natural and engineered tyrosine recombinases (Table 7), large serine integrases (Table 8), serine resolvases (Table 9), and tyrosine integrases (Table 10). Modified target sequences that demonstrate enhanced rates of genomic integration have also been described for several SSRs22-33. In addition to natural recombinases, programmable recombinases with distinct specificities have been developed31-40. Using PE, one or more of these recognition sequences could be introduced into the genomic at a specified location, such as a safe harbor locus41-43, depending on the desired application.


For example, introduction of a single recombinase recognition sequence in the genome by prime editing would result in integrative recombination with a DNA donor template (FIG. 64b). Serine integrases, which operate robustly in human cells, may be especially well-suited for gene integration44,45.


Additionally, introduction of two recombinase recognition sequences could result in deletion of the intervening sequence, inversion of the intervening sequence, chromosomal translocation, or cassette exchange, depending on the identity and orientation of the targets (FIG. 64c-f). By choosing endogenous sequences that already closely resemble recombinase targets, the scope of editing required to introduce the complete recombinase target would be reduced.


Finally, several recombinases have been demonstrated to integrate into human or eukaryotic genomes at natively occurring pseudosites46,64. Multi-flap prime editing could be used to modify these loci to enhance rates of integration at these natural pseudosites, or alternatively, to eliminate pseudosites that may serve as unwanted off-target sequences.


This disclosure describes a general methodology for introducing recombinase target sequences in eukaryotic genomes using multi-flap PE, the applications of which are nearly limitless. The genome editing reactions are intended for use with “prime editors,” a chimeric fusion of a CRISPR/Cas9 protein and a reverse-transcriptase domain, which utilizes a custom prime editing guide RNA (PEgRNA). By extension, Cas9 tools and homology-directed repair (HDR) pathways may also be exploited to introduce recombinase recognition sequences through DNA templates by lowering the rates of indels using several techniques65-67. A proof-of-concept experiment in human cell culture is shown in FIG. 65. In some embodiments, the prime editors are multi-flap prime editors.


The following several tables are cited in the above description relating to PE-directed installation of recombinase recognition sequences and provide a listing of exemplary recombinases that may be used, and their cognate recombinase recognition sequences that may be installed by multi-flap PE.









TABLE 7







Tyrosine recombinases and SSR target sequences.









Recombinase
Recombinase recognition sequence
Name





Cre
ATAACTTCGTATAGCATACATTATACGAAGTTAT (SEQ
loxP



ID NO: 517)






Dre
TAACTTTAAATAATGCCAATTATTTAAAGTTA (SEQ ID
rox



NO: 518)






VCre
TCAATTTCTGAGAACTGTCATTCTCGGAAATTGA (SEQ
loxV



ID NO: 519)






SCre
CTCGTGTCCGATAACTGTAATTATCGGACATGAT (SEQ
loxS



ID NO: 520)






Flp
GAAGTTCCTATTCTCTAGAAAGTATAGGAACTTC (SEQ
FRT



ID NO: 521)






B2
GAGTTTCATTAAGGAATAACTAATTCCCTAATGAAACT
loxB



C (SEQ ID NO: 522)






B3
GGTTGCTTAAGAATAAGTAATTCTTAAGCAACC (SEQ ID
loxB3



NO: 523)






Kw
ACGAAAAATGGTAAGGAATAGACCATTCCTTACCATTT




TTGGT (SEQ ID NO: 524)






R
TTGATGAAAGAATAACGTATTCTTTCATCAA (SEQ ID
RS



NO: 525)






TD1-40
GTGCGTCAAATAATAACGTATTATTTGACACTT (SEQ ID
TDRS



NO: 526)






Vika
AATAGGTCTGAGAACGCCCATTCTCAGACGTATT (SEQ
vox



ID NO: 527)






Nigri
TGAATGTCCTATAATTACACTTATAGGACATTCA (SEQ
nox



ID NO: 528)






Panto
GAAACTTTAAATAATAAGTCTTATTTAAAGTTTC (SEQ
pox



ID NO: 529)






Kd
AAACGATATCAGACATTTGTCTGATAATGCTTCATTA
loxK



TCAGACAAATGTCTGATATCGTTT (SEQ ID NO: 530)






Fre
ATATATACGTATATAGACATATATACGTATATAT (SEQ
loxH



ID NO: 531)






CreALSHG
ATAACTCTATATAATGTATGCTATATAGAGTTAT (SEQ
loxM7



ID NO: 532)






Tre
ACAACATCCTATTACACCCTATATGCCAACATGG (SEQ
loxLTR



ID NO: 533)






Brec1
AACCCACTGCTTAAGCCTCAATAAAGCTTGCCTT (SEQ
loxBTR



ID NO: 534)






Cre-R3M3
GATACAACGTATATACCTTTCTATACGTTGTTTA (SEQ
loxK2



ID NO: 535)
















TABLE 8







Large serine integrases and SSR target sequences.










Recombinase recognition sequence
Recombinase recognition sequence


Recombinase
Left
Right





Bxb1
GGTTTGTCTGGTCAACCACCGCGG
GGCTTGTCGACGACGGCGGTCTCCGTC



TCTCAGTGGTGTACGGTACAAACC
GTCAGGATCAT (SEQ ID NO: 537)



(SEQ ID NO: 536)






phiC31
GTGCCCCAACTGGGGTAACCTTTG
TGCGGGTGCCAGGGCGTGCCCTTGGG



AGTTCTCTCAGTTGGGGG (SEQ ID
CTCCCCGGGCGCGTACTCC (SEQ ID



NO: 538)
NO: 539)





R4
TGTTCCCCAAAGCGATACCACTTG
GCATGTTCCCCAAAGCGATACCACTTG



AAGCAGTGGTACTGCTTGTGGGT
AAGCAGTGGTACTGCTTGTGGGTACA



ACA (SEQ ID NO: 540)
CTCTGCGGGTG (SEQ ID NO: 541)





phiBT1
GGTGCTGGGTTGTTGTCTCTGGAC
CAGGTTTTTGACGAAAGTGATCCAGAT



AGTGATCCATGGGAAACTACTCA
GATCCAG (SEQ ID NO: 543)



GCACC (SEQ ID NO: 542)






MJ1 (phiFC1)
ATTTTAGGTATATGATTTTGTTTAT
CAAAGGATCACTGAATCAAAAGTATT



TAGTGTAAATAACACTATGTACCT
GCTCATCCACGCGAAA (SEQ ID NO:



AAAAT (SEQ ID NO: 544)
545)





MR11
TTTGTGCGGAACTACGAACAGTTC
CGAAAATGTATGGAGGCACTTGTATC



ATTAATACGAAGTGTACAAACTTC
AATATAGGATGTATACCTTCGAAGAC



CATACAA (SEQ ID NO: 546)
ACTT (SEQ ID NO: 547)





TP901-1
GAGTTTTTATTTCGTTTATTTCAAT
ATGCCAACACAATTAACATCTCAATCA



TAAGGTAACTAAAAAACTCCTTTT
AGGTAAATGCTTTTTGCTTTTTTTGC



AAGG (SEQ ID NO: 548)
(SEQ ID NO: 549)





A118
TTCCTCGTTTTCTCTCGTTGGAAG
TTTCGGATCAAGCTATGAAGGACGCA



AAGAAGAAACGAGAAA (SEQ ID
AAGAGGGAACTAAA(SEQ ID NO: 551)



NO: 550)






U153
TTCCTCGTTTTCTCTCGTTGGACG
TTTCGGATCAAGCTATGAAGGACGCA



GAAACGAATCGAGAAA (SEQ ID
AAGAGGGAACTAAA (SEQ ID NO: 553)



NO: 552)






phiRV1
GTAGTGTATCTCACAGGTCCACGG
GAAGGTGTTGGTGCGGGGTTGGCCGT



TTGGCCGTGGACTGCTGAAGAAC
GGTCGAGGTGGGGT (SEQ ID NO: 555)



ATTCC (SEQ ID NO: 554)






phi370.1
AAAAAAATACAGCGTTTTTCATGT
TTGTAAAGGAGACTGATAATGGCATG



ACAACTATACTAGTTGTAGTGCCT
TACAACTATACTCGTCGGTAAAAAGG



AAAA (SEQ ID NO: 556)
CA (SEQ ID NO: 557)





TG1
TCCAGCCCAACAGTGTTAGTCTTT
GATCAGCTCCGCGGGCAAGACCTTTCT



GCTCTTACCCAGTTGGGCGGGA
CCTTCACGGGGTGGAAGGTC (SEQ ID



(SEQ ID NO: 558)
NO: 559)





WB
CTAGTTTTAAAGTTGGTTATTAGT
CGGAAGGTAGCGTCAACGATAGGTGT



TACTGTGATATTTATCACGGTACC
AACTGTCGTGTTTGTAACGGTACTTCC



CAATAACCAATGAAT (SEQ ID NO:
AACAGCTGGCGCCGCCAC (SEQ ID NO:






560)
561)


BL3
CAATGAAAAACTAGGCATGTAGA
TTTCCACAGACAACTCACGTGGAGGT



AGTTGTTTGT (SEQ ID NO: 562)
AGTCAC (SEQ ID NO: 563)





SprA
TGTAGTAAGTATCTTAATATACAG
CACCCATTGTGTTCACAGGAGATACA



CTTTATCTGTTTTTTAAGATACTTA
GCTTTATCTGTACTGATATTAATGACA



CTACTTT (SEQ ID NO: 564)
TGCTG (SEQ ID NO: 565)





phiJoe
AGTTGTGGCCATGTGTCCATCTGG
ATCTGGATGTGGGTGTCCATCTGCGGG



GGGCAGATGGAGACGGGGTCACA
CAGACGCCGCAGTCGAAGCACGG



(SEQ ID NO: 566)
(SEQ ID NO: 567)





 —

ACCTTGATCTCGGTGTCCATCGCCGGG




CAGACGCCGCAGTCGAAGCACGG




(SEQ ID NO: 568)





phiK38
CCCTAATACGCAAGTCGATAACTC
GAGCGCCGGATCAGGGAGTGGACGGC



TCCTGGGAGCGTTGACAACTTGCG
CTGGGAGCGCTACACGCTGTGGCTGC



CACCCTGATCTG (SEQ ID NO: 569)
GGTCGGTGC (SEQ ID NO: 570)





Int2
GCTCATGTATGTGTCTACGCGAGA
GGACGGCGCAGAAGGGGAGTAGCTCT



TTCTCGCCCGAGAACTTCTGCAAG
TCGCCGGACCGTCGACATACTGCTCAG



GCACTGCTCTTGGCT (SEQ ID NO:
CTCGTC (SEQ ID NO: 572)



571)






Int3
ATGGATAAAAAAATACAGCGTTT
GTTTGTAAAGGAGACTGATAATGGCA



TTCATGTACAACTATACTAGTTGT
TGTACAACTATACTCGTCGGTAAAAA



AGTGCCTAAATAATGCTT (SEQ ID
GGCATCTTAT (SEQ ID NO: 574)



NO: 573)






Int4
AAAAATTACAAAGTTTTCAACCCT
TTCCAAAGAGCGCCCAACGCGACCTG



TGATTTGAATTAGCGGTCAAATAA
AAATTTGAATAAGACTGCTGCTTGTGT



TTTGTAATTCGTTT (SEQ ID NO:
AAAGGCGATGATT (SEQ ID NO: 576)



575)






Int7
GTGTTATAAACCTGTGTGAGAGTT
AGACGAGAAACGTTCCGTCCGTCTGG



AAGTTTACATGCCTAACCTTAACT
GTCAGTTGGGCAAAGTTGATGACCGG



TTTACGCAGGTTCAGCTT(SEQ ID
GTCGTCCGTT (SEQ ID NO: 578)



NO: 577)






Int8
TTAATAAACTATGGAAGTATGTAC
CAATCATCAGATAACTATGGCGGCAC



AGTCTTGCAATGTTGAGTGAACA
GTGCATTAACCACGGTTGTATCCCGTC



AACTTCCATAATAAAAT (SEQ ID
TAAAGTACTCGT (SEQ ID NO: 580)



NO: 579)






Int9
GTGGTTGTTTTTGTTGGAAGTGTG
TTTATATTGCGAAAAATAATTGGCGAA



TATCAGGTATCTGCATAGTTATTC
CGAGGTAACTGGATACCTCATCCGCC



CGAACTTCCAATTA (SEQ ID NO:
AATTAAAATTTG (SEQ ID NO: 582)



581)






Int10
GGAAAATATAAATAATTTTAGTA
AGCACGCTGATAATCAGCAAGACCAC



ACCTACATCTCAATCAAGGATAGT
CAACATTTCCACCAATGTAAAAGCTTT



AAAACTCTCACTCTT (SEQ ID NO:
AACCTTAGC (SEQ ID NO: 584)



583)






Int11
GTTTATATGTTTACTAATAAGACG
ATGGATTTTGCAGATTCCCAGATGCCC



CTCTCAACCCATAAAGTCTTATTA
CTACAGAAAGAGGTACAAAACATTTA



GTAAACATATTTCAACT (SEQ ID
TTGGAATTAATT (SEQ ID NO: 586)



NO: 585)






Int12
TTTTTGTATGTTAGTTGTGTCACTG
GTTCGTGGTAACTATGGGTGGTACAG



GGTAGACCTAAATAGTGACACAA
GTGCCACATTAGTTGTACCATTTATGT



CTGCTATTAAAATTTAA (SEQ ID
TTATGTGGTTAAC (SEQ ID NO: 588)



NO: 587)






Int13
CAATAACGGTTGTATTTGTAGAAC
GCATACATTGTTGTTGTTTTTCCAGAT



TTGACCAGTTGTTTTAGTAACATA
CCAGTTGGTCCTGTAAATATAAGCAAT



AATACAACTCCGAATA (SEQ ID
CCATGTGAGT (SEQ ID NO: 590)



NO: 589)






LI
GTTTAGTATCTCGTTATCTCTCGTT
TAACTTTTTCGGATCGAGTTATGATGG



GGAGGGAGAAGAAACGGGATACC
ACGTAAAGAGGGAACAAAGCATCTA



AAAA (SEQ ID NO: 591)
(SEQ ID NO: 592)





Peaches
TAGTTTCCAATGTTACAGGAACTG
CGGTCTCCATCGGGATCTGCTGATCGA



CTGGCAGAATCCAACACATTGGA
GCAGCATGCCGACCA (SEQ ID NO: 594)



AGTCG (SEQ ID NO: 593)






Bxz2
TAACCGCAAGTGTACATCCCTCGG
CGGTCTCCATCGGGATCTGCTGATCGA



CTGGCCGAGACAAGTACAGTTGC
GCAGCATGCCGACCA (SEQ ID NO: 596)



GACAG (SEQ ID NO: 595)






SV1
ATGTGGTCCTTTAGATCCACTGAC
CATCAGGGCGGTCAGGCCGTAGATGT



GTGGGTCAGTGTCTCTAAAGGACT
GGAAGAAACGGCAGCACGGCGAGGA



CGCG (SEQ ID NO: 597)
CG (SEQ ID NO: 598)
















TABLE 9







Serine resolvases and SSR target sequences.










Recombinase recognition sequence
Recombinase recognition sequence


Resolvase
Left
Right





Gin
CGTTTCCTGTAAACCGAGGTTTTGGA
CGTTTCCTGTAAACCGAGGTTTTGGA



TAAACA (SEQ ID NO: 599)
TAATGG (SEQ ID NO: 600)





Cin
GAGTTCTCTTAAACCAAGGTTTAGG
GAGTTCTCTTAAACCAAGGTATTGG



ATTGAAA (SEQ ID NO: 601)
ATAACAG (SEQ ID NO: 602)





Hin
TGGTTCTTGAAAACCAAGGTTTTTGA
AAATTTTCCTTTTTGGAAGGTTTTTG



TAAAGC (SEQ ID NO: 603)
ATAACCA (SEQ ID NO: 604)





Min
GCCTTCCCCTAAACCAACGTTTTTAT
GCCTTCCCCCAAACCAAGGTAATCA



GCCGCC (SEQ ID NO: 605)
AGAACGC (SEQ ID NO: 606)





Sin
TTGTGAAATTTGGGTACACCCTAATC
CGTATGATTAGGGTGTATATTAATTT



ATACAA (SEQ ID NO: 607)
(SEQ ID NO: 608)
















TABLE 10







Tyrosine integrases and target sequences.









Integrase
attP
attB





HK022
CAAATGATTTTATTTTGACTAATAATGA
GCACTTTAGGTGAAAAAGGTT



CCTACTTACATTAATTTACTGATAATTA
(SEQ ID NO: 610)



AAGAGATTTTAAATATACAACTTATTC




ACCTAAAGGATGACAAAA (SEQ ID NO:




609)




TAACATTAATCACTTAAAAATCATCGC




ATTACACTAATCTGTGGTTAAATGATA




GACTACATAATGCGACAAAACGCAACA




TATCCAGTCACTATGAATCAACTACTT




AGATAGTATTAGTGACCT (SEQ ID NO:




611)






P22
CTAAGTGGTTTGGGACAAAAATGGGAC
GCAGCGCATTCGTAATGCGAAG



ATACAAATCTTTGCATCGGTTTGCAAG
GTCGT (SEQ ID NO: 613)



GCTTTGCATGTCTTTCGAAGATGGGAC




GTGTGAGCGCAGGTATGACGTGGTATG




TGTTGACTTAAAAGGTAGTTCTTATAAT




TCGTAATGCGAAGGTCGTAGGTTCGAC




TCCTATTATCGGCACCAGTTAAATCAA




ATACTTACGTATTATTCGTGCCTTCCTT




ATTTTTACTGTGGGACATATTTGGGACA




GAAGTACCAAAAA (SEQ ID NO: 612)






L5
GCGATCCCCATCCGCGACGTGCCAACT
GAGCGGGCGACGGGAATCGAA



AGGTCTCCTCTCGTCGTGAACAAGGCT
CCCGCGTAGCTAGTTTGGAAGA



ACCGGGTTGCAACTCCTGTGCAACTCT
(SEQ ID NO: 615)



CAGGCTTCAACGCGCTTCTACGACCTG




CAATTTCTTTCCACTTAGAGGATGCAG




CCGAGAGGGGTAAAAACCTATCTTGAC




CGGCCCATATGTGGTCGGCAGACACCC




ATTCTTCCAAACTAGCTACGCGGGTTC




GATTCCCGTCGCCCGCTCCGCTGGTCA




GAGGGTGTTTTCGCCCTCTGGCCATTTT




TCTTTCCAGGGGTCTGCAACTCTTGTGC




GACTCTTCTGACCTGGGCATACGCGGT




TGCAACGCATCCCTGATCTGGCTACTTT




CGATGCTGACAAACGAATAGAGCCCCC




CGCCTGCGCGAACAGACGAGGGGCATT




CACA (SEQ ID NO: 614)









In various other aspects, the present disclosure relates to methods of using multi-flap PE to install one or more recombinase recognition sequences and their use in site-specific recombination.


In some embodiments, the site-specific recombination may effectuate a variety of recombination outcomes, such as, excision, integration, inversion, or exchange of DNA fragments.


In some embodiments, the methods are useful for inducing recombination of or between two or more regions of two or more nucleic acid (e.g., DNA) molecules. In other embodiments, the methods are useful for inducing recombination of or between two or more regions in a single nucleic acid molecule (e.g., DNA).


In some embodiments, the disclosure provides a method for integrating a donor DNA template by site-specific recombination, comprising: (a) installing a recombinase recognition sequence at a genomic locus by multi-flap prime editing; (b) contacting the genomic locus with a DNA donor template that also comprises the recombinase recognition sequence in the presence of a recombinase.


In other embodiments, the disclosure provides a method for deleting a genomic region by site-specific recombination, comprising: (a) installing a pair of recombinase recognition sequences at a genomic locus by multi-flap prime editing; (b) contacting the genomic locus with a recombinase, thereby catalyzing the deletion of the genomic region between the pair of recombinase recognition sequences.


In yet other embodiments, the disclosure provides a method for inverting a genomic region by site-specific recombination, comprising: (a) installing a pair of recombinase recognition sequences at a genomic locus by multi-flap prime editing; (b) contacting the genomic locus with a recombinase, thereby catalyzing the inversion of the genomic region between the pair of recombinase recognition sequences.


In still other embodiments, the disclosure provides a method for inducing chromosomal translocation between a first genomic site and a second genomic site, comprising: (a) installing a first recombinase recognition sequence at a first genomic locus by prime editing; (b) installing a second recombinase recognition sequence at a second genomic locus by multi-flap prime editing; (c) contacting the first and the second genomic loci with a recombinase, thereby catalyzing the chromosomal translocation of the first and second genomic loci.


In other embodiments, the disclosure provides a method for inducing cassette exchange between a genomic locus and a donor DNA comprising a cassette, comprising: (a) installing a first recombinase recognition sequence at a first genomic locus by multi-flap prime editing; (b) installing a second recombinase recognition sequence at a second genomic locus by multi-flap prime editing; (c) contacting the first and the second genomic loci with a donor DNA comprising a cassette that is flanked by the first and second recombinase recognition sequences and a recombinase, thereby catalyzing the exchange of the flanked genomic locus and the cassette in the DNA donor.


In various embodiments involving the insertion of more than one recombinase recognition sequences in the genome, the recombinase recognition sequences can be the same or different. In some embodiments, the recombinase recognition sequences are the same. In other embodiments, that recombinase recognition sequences are different.


In various embodiments, the recombinase can be a tyrosine recombinase, such as Cre, Dre, Vcre, Scre, Flp, B2, B3, Kw, R, TD1-40, Vika, Nigri, Panto, Kd, Fre, Cre(ALSHG), Tre, Brec1, or Cre-R3M3, as shown in Table 7. In such embodiments, the recombinase recognition sequence may be an RRS of Table 7 that corresponds to the recombinase under use.


In various other embodiments, the recombinase can be a large serine recombinase, such as Bxb1, PhiC31, R4, phiBT1, MJ1, MR11, TP901-1, A118, V153, phiRV1, phi370.1, TG1, WB, BL3, SprA, phiJoe, phiK38, Int2, Int3, Int4, Int7, Int8, Int9, Int10, Int11, Int12, Int13, L1, peaches, Bxz2, or SV1, as shown in Table 8. In such embodiments, the recombinase recognition sequence may be an RRS of Table 8 that corresponds to the recombinase under use.


In still other embodiments, the recombinase can be a serine recombinase, such as Bxb1, PhiC31, R4, phiBT1, MJ1, MR11, TP901-1, A118, V153, phiRV1, phi370.1, TG1, WB, BL3, SprA, phiJoe, phiK38, Int2, Int3, Int4, Int7, Int8, Int9, Int10, Int11, Int12, Int13, L1, peaches, Bxz2, or SV1, as shown in Table 8. In such embodiments, the recombinase recognition sequence may be an RRS of Table 8 that corresponds to the recombinase under use.


In other embodiments, the recombinase can be a serine resolvase, such as Gin, Cin, Hin, Min, or Sin, as shown in Table 9. In such embodiments, the recombinase recognition sequence may be an RRS of Table 9 that corresponds to the recombinase under use.


In various other embodiments, the recombinase can be a tyrosine integrase, such as HK022, P22, or L5, as shown in Table 10. In such embodiments, the recombinase recognition sequence may be an RRS of Table 10 that corresponds to the recombinase under use.


In some embodiments, any of the methods for site-specific recombination with multi-flap PE can be performed in vivo or in vitro. In some embodiments, any of the methods for site-specific recombination are performed in a cell (e.g., recombine genomic DNA in a cell). The cell can be prokaryotic or eukaryotic. The cell, such as a eukaryotic cell, can be in an individual, such as a subject, as described herein (e.g., a human subject). The methods described herein are useful for the genetic modification of cells in vitro and in vivo, for example, in the context of the generation of transgenic cells, cell lines, or animals, or in the alteration of genomic sequence, e.g., the correction of a genetic defect, in a cell in a subject.


[8] Embodiments of the Multi-Flap Prime Editors Described Herein

The following non-limiting embodiments describe aspects of the dual prime editors and their use as described in this application.

    • 1. A system for simultaneously editing both strands of a double-stranded DNA sequence at a target site to be edited, wherein the system comprises a first prime editor complex and a second prime editor complex, wherein each of the first and second prime editor complexes comprises (1) a prime editor comprising (i) a nucleic acid programmable DNA binding protein (napDNAbp), and (ii) a polypeptide having an RNA-dependent DNA polymerase activity; and (2) a pegRNA comprising a spacer sequence, gRNA core, a DNA synthesis template, and a primer binding site, wherein the DNA synthesis template of the pegRNA of the first prime editor complex encodes a first single-stranded DNA sequence and the DNA synthesis template of the pegRNA of the second prime editor complex encodes a second single-stranded DNA sequence, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence each comprises a region of complementarity to the other, and wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence form a duplex comprising an edited portion as compared to the DNA sequence at the target site to be edited, which integrates into the target site to be edited.
    • 2. The system of embodiment 1, wherein the prime editor of the first prime editor complex, the second prime editor complex, or both the first prime editor complex and the second prime editor complex is a fusion protein comprising the napDNAbp and the polypeptide having an RNA-dependent DNA polymerase activity.
    • 3. The system of embodiment 1 or 2, wherein the first prime editor complex and the second prime editor complex comprise the same prime editor.
    • 4. The system of embodiment 1 or 2, wherein the first prime editor complex and the second prime editor complex comprise different prime editors.
    • 5. The system of any one of embodiments 1-4, wherein the napDNAbp is a Cas9 domain or variant thereof.
    • 6. The system of any one of embodiments 1-5, wherein the napDNAbp is a nuclease active Cas9 domain, a nuclease inactive Cas9 domain, a Cas9 nickase domain, or variant thereof.
    • 7. The system of any one of embodiments 1-4, wherein the napDNAbp is selected from the group consisting of: Cas9, Cas12e, Cas12d, Cas12a, Cas12b1, Cas13a, Cas12c, CasX, CasY, and Argonaute.
    • 8. The system of any one of embodiments 1-7, wherein the napDNAbp has a nickase activity.
    • 9. The system of any one of embodiments 1-4, wherein the napDNAbp comprises an amino acid sequence of any one of SEQ ID NOs: 18-88, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 18-88.
    • 10. The system of any one of embodiments 1-9, wherein the polypeptide having an RNA-dependent DNA polymerase activity is a reverse transcriptase.
    • 11. The system of any one of embodiments 1-10, wherein the polypeptide having an RNA-dependent DNA polymerase activity comprises an amino acid sequence of any one of SEQ ID NOs: 89-100, 105-122, 128-129, 132, 139, 143, 149, 154, 159, and 700-766 or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 89-100, 105-122, 128-129, and 132.
    • 12. The system of any one of embodiments 1-11, wherein the prime editor further comprises a linker that joins the napDNAbp and the polypeptide having the RNA-dependent DNA polymerase activity.
    • 13. The system of embodiment 12, wherein the linker comprises an amino acid sequence of any one of SEQ ID NOs: 166-177, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 166-177.
    • 14. The system of embodiment 12 or 13, wherein the linker is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 38, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 amino acids in length.
    • 15. The system of any one of embodiments 1-14, wherein the pegRNA comprises a nucleotide sequence of any one of SEQ ID NOs: 325-330, 499-505, 101-104, 181-183, and 223-244, or a nucleotide sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 325-330, 499-505, 101-104, 181-183, and 223-244.
    • 16. The system of any one of embodiments 1-15, wherein the spacer sequence of the pegRNA of the first prime editor complex binds to a first binding site on a first strand of the double-stranded DNA sequence upstream from the target site to be edited.
    • 17. The system of any one of embodiments 1-16, wherein the spacer sequence of the pegRNA of the second prime editor complex binds to a second binding site on a second strand of the double-stranded DNA sequence downstream from the target site to be edited.
    • 18. The system of embodiment 17, whereby the binding of the spacer sequence of the pegRNAs of the first and second prime editor complexes in the presence of a PAM sequence results in the nicking of the first and second strands, respectively, at a nick site proximal to the PAM sequences on each strand by the napDNAbps of each of the first and second prime editor complex.
    • 19. The system of embodiment 18, wherein the first and second binding sites define two ends of a contiguous region of double-stranded DNA comprising the target site to be edited.
    • 20. The system of embodiment 19, wherein the contiguous region between the first and second binding sites is at least 10 nucleobase pairs, at least 11 nucleobase pairs, at least 12 nucleobase pairs, at least 13 nucleobase pairs, at least 14 nucleobase pairs, at least 15 nucleobase pairs, at least 16 nucleobase pairs, at least 17 nucleobase pairs, at least 18 nucleobase pairs, at least 19 nucleobase pairs, at least 20 nucleobase pairs, at least 21 nucleobase pairs, at least 22 nucleobase pairs, at least 23 nucleobase pairs, at least 24 nucleobase pairs, at least 25 nucleobase pairs, at least 26 nucleobase pairs, at least 27 nucleobase pairs, at least 28 nucleobase pairs, at least 29 nucleobase pairs, at least 30 nucleobase pairs, at least 31 nucleobase pairs, at least 32 nucleobase pairs, at least 33 nucleobase pairs, at least 34 nucleobase pairs, at least 35 nucleobase pairs, at least 36 nucleobase pairs, at least 37 nucleobase pairs, at least 37 nucleobase pairs, at least 38 nucleobase pairs, at least 39 nucleobase pairs, or at least 40 nucleobase pairs, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 21. The system of embodiment 19, wherein the contiguous region becomes replaced by the duplex comprising the edited portion.
    • 22. The system of any one of embodiments 1-21, wherein the DNA synthesis template is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 23. The system of any one of embodiments 1-22, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence have the same lengths.
    • 24. The system of any one of embodiments 1-22, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence have different lengths.
    • 25. The system of any one of embodiments 1-24, wherein the region of complementarity is at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, at least 50%, at least 45%, at least 40%, at least 35%, at least 30%, at least 25%, at least 20%, at least 15%, or at least 10% of the total length of the first single-stranded DNA sequence or the second single-stranded DNA sequence.
    • 26. The system of embodiment 25, wherein the region of complementarity is at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length, and is either less than or equal to the length of the desired DNA sequence and complement thereof.
    • 27. The system of embodiment 25, wherein the region of complementarity of the first single-stranded DNA sequence and the second single-stranded DNA sequence form the duplex comprising the edited portion.
    • 28. The system of any one of embodiments 1-27, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence are each single-stranded DNA strands having 3′ ends.
    • 29. The system of embodiment 21, wherein the replacement of the contiguous region by the duplex comprising the edited portion results in an insertion, deletion, or replacement of DNA at the target site.
    • 30. A system for simultaneously editing both strands of a double-stranded DNA sequence at a target site to be edited, said system comprising:
      • (a) a first prime editor complex comprising:
        • i. a first prime editor comprising a first nucleic acid programmable DNA binding protein (first napDNAbp) and a first polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a first prime editing guide RNA (first PEgRNA) that binds to a first binding site on a first strand of the double-stranded DNA sequence upstream of the target site to be edited;
      • (b) a second prime editor complex comprising:
        • i. a second prime editor comprising a second nucleic acid programmable DNA binding protein (second napDNAbp) and a second polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a second prime editing guide RNA (second PEgRNA) that binds to a second binding site on a second strand of the double-stranded DNA sequence downstream of the target site to be edited;
      • wherein the first PEgRNA comprises a first DNA synthesis template encoding a first single-stranded DNA sequence and the second PEgRNA comprises a second DNA synthesis template encoding a second single-stranded DNA sequence, and wherein the first strand and the second strand of the double-stranded DNA sequence are complementary to each other.
    • 31. The system of embodiment 30, wherein the prime editor of the first prime editor complex, the second prime editor complex, or both the first prime editor complex and the second prime editor complex is a fusion protein comprising the napDNAbp and the polypeptide having an RNA-dependent DNA polymerase activity.
    • 32. The system of embodiment 30, wherein the first and the second single-stranded DNA sequence each comprise a region of complementarity to the other.
    • 33. The system of embodiment 30, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence form a duplex comprising the edited portion which integrates into the target site to be edited.
    • 34. The system of embodiment 30, wherein the first single-stranded DNA sequence and/or the second single stranded sequence does not comprise sequence homology as compared to the DNA sequence at the target site.
    • 35. The system of embodiment 30, wherein the first PEgRNA further comprises a first spacer sequence, a first gRNA core, and a first primer binding site, and the second PEgRNA further comprises a second spacer sequence, a second gRNA core, and a second primer binding site.
    • 36. The system of any one of embodiments 30-35, wherein the first prime editor complex and the second prime editor complex comprise the same prime editor.
    • 37. The system of any one of embodiments 30-35, wherein the first polypeptide comprising an RNA-dependent DNA polymerase activity and the second polypeptide comprising an RNA-dependent DNA polymerase activity are the same.
    • 38. The system of any one of embodiments 30-37, wherein the first and/or second napDNAbp is a Cas9 domain or variant thereof.
    • 39. The system of any one of embodiments 30-38, wherein the first and/or second napDNAbp is a nuclease active Cas9 domain, a nuclease inactive Cas9 domain, a Cas9 nickase domain, or a variant thereof.
    • 40. The system of any one of embodiments 30-39, wherein the first and/or second napDNAbp is selected from the group consisting of Cas9, Cas12e, Cas12d, Cas12a, Cas12b1, Cas13a, Cas12c, CasX, CasY, and Argonaute.
    • 41. The system of any one of embodiments 30-40, wherein the napDNAbp has a nickase activity.
    • 42. The system of any one of embodiments 30-41, wherein the first and/or second napDNAbp comprises an amino acid sequence of any one of SEQ ID NOs: 18-88, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 18-88.
    • 43. The system of any one of embodiments 30-42, wherein the first and/or second polypeptide having an RNA-dependent DNA polymerase activity is a reverse transcriptase.
    • 44. The system of any one of embodiments 30-43, wherein the first and/or second polypeptide having an RNA-dependent DNA polymerase activity comprises an amino acid sequence of any one of SEQ ID NOs: 89-100, 105-122, 128-129, 132, 139, 143, 149, 154, 159, and 700-766 or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 89-100, 105-122, 128-129, and 132.
    • 45. The system of any one of embodiments 30-44, wherein the first prime editor further comprises a linker that joins the first napDNAbp and the first polypeptide having the RNA-dependent DNA polymerase activity; and/or the second prime editor further comprises a linker that joins the second napDNAbp and the second polypeptide having the RNA-dependent DNA polymerase activity.
    • 46. The system of embodiment 45, wherein the linker comprises an amino acid sequence of any one of SEQ ID NOs: 166-177, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 166-177.
    • 47. The system of embodiment 45 or 46, wherein the linker is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 38, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 amino acid in length.
    • 48. The system of any one of embodiments 30-47, wherein the first and/or second pegRNA comprises a nucleotide sequence of any one of SEQ ID NOs: 325-330, 499-505, 101-104, 181-183, and 223-244, or a nucleotide sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 325-330, 499-505, 101-104, 181-183, and 223-244.
    • 49. The system of any one of embodiments 30-48, wherein the first spacer sequence binds to a first binding site on a first strand of the double-stranded DNA sequence upstream from the target site to be edited.
    • 50. The system of embodiment 49, wherein the second spacer sequence of the second prime editor complex binds to a second binding site on a second strand of the double-stranded DNA sequence downstream from the target site to be edited.
    • 51. The system of embodiment 50, whereby the binding of the spacer sequence of the first and second prime editor complexes in the presence of a PAM sequence results in the nicking of the first and second strands, respectively, at a nick site proximal to the PAM sequences on each strand by the first and/or second napDNAbps of each of the first and second prime editor complexes.
    • 52. The system of embodiment 51, wherein the first and second binding sites define two ends of a contiguous region of double-stranded DNA comprising the target site to be edited.
    • 53. The system of embodiment 52, wherein the contiguous region between the first and second binding sites is least 10 nucleobase pairs, at least 11 nucleobase pairs, at least 12 nucleobase pairs, at least 13 nucleobase pairs, at least 14 nucleobase pairs, at least 15 nucleobase pairs, at least 16 nucleobase pairs, at least 17 nucleobase pairs, at least 18 nucleobase pairs, at least 19 nucleobase pairs, at least 20 nucleobase pairs, at least 21 nucleobase pairs, at least 22 nucleobase pairs, at least 23 nucleobase pairs, at least 24 nucleobase pairs, at least 25 nucleobase pairs, at least 26 nucleobase pairs, at least 27 nucleobase pairs, at least 28 nucleobase pairs, at least 29 nucleobase pairs, at least 30 nucleobase pairs, at least 31 nucleobase pairs, at least 32 nucleobase pairs, at least 33 nucleobase pairs, at least 34 nucleobase pairs, at least 35 nucleobase pairs, at least 36 nucleobase pairs, at least 37 nucleobase pairs, at least 37 nucleobase pairs, at least 38 nucleobase pairs, at least 39 nucleobase pairs, or at least 40 nucleobase pairs, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 54. The system of embodiment 53, wherein the contiguous region becomes replaced by the duplex comprising the edited portion.
    • 55. The system of any one of embodiments 30-54, wherein the first and/or second DNA synthesis template is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 56. The system of any one of embodiments 30-55, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence have the same lengths.
    • 57. The system of any one of embodiments 30-55, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence have different lengths.
    • 58. The system of any one of embodiments 30-57, wherein the region of complementarity is at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, at least 50%, at least 45%, at least 40%, at least 35%, at least 30%, at least 25%, at least 20%, at least 15%, or at least 10% of the total length of the first single-stranded DNA sequence or the second single-stranded DNA sequence.
    • 59. The system of any one of embodiments 30-58, wherein the region of complementarity is at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 60. The system of any one of embodiments 30-59, wherein the region of complementarity of the first single-stranded DNA sequence and the second single-stranded DNA sequence form a duplex.
    • 61. The system of any one of embodiments 30-60, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence are each single-stranded DNA strands having 3′ ends.
    • 62. The system of embodiment 54, wherein the replacement of the contiguous region by the duplex comprising the edited portion results in an insertion, deletion, or replacement of DNA at the target site.
    • 63. A polynucleotide encoding the system of any of embodiments 1-62.
    • 64. A polynucleotide encoding the first and second prime editors of any one of embodiments 1-62.
    • 65. A polynucleotide encoding the first and/or second pegRNA of any one of embodiments 1-29.
    • 66. A polynucleotide encoding the first and/or second pegRNA of any one of embodiments 30-62.
    • 67. A polynucleotide encoding the first and second pegRNA of any one of embodiments 1-29.
    • 68. A polynucleotide encoding the first and second pegRNA of any one of embodiments 30-62.
    • 69. A vector comprising a polynucleotide of any one of embodiments 63-68.
    • 70. A cell comprising a polynucleotide of any one of embodiments 63-69 or a vector of embodiment 64.
    • 71. A pharmaceutical composition comprising a polynucleotide of any one of embodiments 63-68, a vector of embodiment 69, or a cell of embodiment 70, and a pharmaceutical excipient.
    • 72. A kit comprising a polynucleotide of any of embodiments 63-69 or a vector of embodiment 70 and instructions for simultaneously editing both strands of a double-stranded DNA sequence at a target site to be edited.
    • 73. A method for simultaneously editing a first and a second complementary strands of a double-stranded DNA sequence at a target site, said method comprising contacting the double-stranded DNA sequence with a pair of prime editor complexes, said pair comprising:
      • a. a first prime editor complex, comprising:
        • i. a first prime editor comprising a first nucleic acid programmable DNA binding protein (napDNAbp) and a first polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a first prime editing guide RNA (first PEgRNA) that binds to a first binding site on the first strand of the genomic DNA sequence upstream of the target site;
      • b. a second prime editor complex, comprising:
        • i. a second prime editor comprising a second nucleic acid programmable DNA binding protein (second napDNAbp) and a second polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a second prime editing guide RNA (second PEgRNA) that binds to a second binding site on the second strand of the genomic DNA sequence downstream of the target site;
      • wherein the first prime editor complex causes a first nick at a sequence complementary to the first binding site and the subsequent polymerization of a first single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the first nick;
      • wherein the second prime editor complex causes a second nick at a sequence complementary to the second binding site and the subsequent polymerization of a second single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the second nick;
      • wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence are reverse complements over at least a region of complementarity and form a duplex comprising an edit; and
      • wherein the duplex replaces the nicked first and second complementary strands of the double-stranded DNA sequence.
    • 74. The method of embodiment 73, wherein the prime editor of the first prime editor complex, the second prime editor complex, or both the first prime editor complex and the second prime editor complex is a fusion protein comprising the napDNAbp and the polypeptide having an RNA-dependent DNA polymerase activity
    • 75. The method of embodiment 73, wherein the first prime editor complex and the second prime editor complex comprise the same prime editor.
    • 76. The method of embodiment 73, wherein the first prime editor complex and the second prime editor complex comprise a different prime editor.
    • 77. The method of any one of embodiments 73-76, wherein the first and/or second napDNAbp is a Cas9 domain or variant thereof.
    • 78. The method of any one of embodiments 73-77, wherein the first and/or second napDNAbp is a nuclease active Cas9 domain, a nuclease inactive Cas9 domain, a Cas9 nickase domain, or variant thereof.
    • 79. The method of any one of embodiments 73-76, wherein the first and/or second napDNAbp is selected from the group consisting of: Cas9, Cas12e, Cas12d, Cas12a, Cas12b1, Cas13a, Cas12c, CasX, CasY, and Argonaute.
    • 80. The method of any one of embodiments 73-79, wherein the first and/or second napDNAbp has a nickase activity.
    • 81. The method of any one of embodiments 73-80, wherein the first and/or second napDNAbp comprises an amino acid sequence of any one of SEQ ID NOs: 18-88, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 18-88.
    • 82. The method of any one of embodiments 73-81, wherein the first and/or second polypeptide having an RNA-dependent DNA polymerase activity is a reverse transcriptase.
    • 83. The method of any one of embodiments 73-82, wherein the first and/or second polypeptide having an RNA-dependent DNA polymerase activity comprises an amino acid sequence of any one of SEQ ID NOs: 89-100, 105-122, 128-129, 132, 139, 143, 149, 154, 159, and 700-766 or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 89-100, 105-122, 128-129, and 132.
    • 84. The method of any one of embodiments 73-83, wherein the first prime editor further comprises a linker that joins the first napDNAbp and the first polypeptide having the RNA-dependent DNA polymerase activity, and/or the second prime editor further comprises a linker that joins the second napDNAbp and the second polypeptide having the RNA-dependent DNA polymerase activity.
    • 85. The method of embodiment 84, wherein the linker comprises an amino acid sequence of any one of SEQ ID NOs: 166-177, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 166-177.
    • 86. The method of embodiment 84 or 85, wherein the linker is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 38, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 amino acids in length.
    • 87. The method of any one of embodiments 73-86, wherein the first and/or second pegRNA comprises a nucleotide sequence of any one of SEQ ID NOs: 325-330, 499-505, 101-104, 181-183, and 223-244, or a nucleotide sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 325-330, 499-505, 101-104, 181-183, and 223-244.
    • 88. The method of any one of embodiments 73-87, wherein the first and/or second PEgRNA comprises a nucleotide sequence of any one of SEQ ID NOs: 325-330, 499-505, 101-104, 181-183, and 223-244, or a nucleotide sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 325-330, 499-505, 101-104, 181-183, and 223-244.
    • 89. The method of any one of embodiments 73-88, wherein the first spacer sequence of the first prime editor complex binds to a first binding site on a first strand of the double-stranded DNA sequence upstream from the target site to be edited.
    • 90. The method of any one of embodiments 73-89, wherein the second spacer sequence of the second prime editor complex binds to a second binding site on a second strand of the double-stranded DNA sequence downstream from the target site to be edited.
    • 91. The method of embodiment 90, wherein the binding of the first and second spacer sequences of the first and second prime editor complexes in the presence of a PAM sequence results in the nicking of the first and second strands, respectively, at a nick site proximal to the PAM sequence on each strand by the napDNAbps of each first and second prime editor complex.
    • 92. The method of embodiment 91, wherein the first and second binding sites define two ends of a contiguous region of double-stranded DNA comprising the target site to be edited.
    • 93. The method of embodiment 92, wherein the contiguous region between the first and second binding sites is least 10 nucleobase pairs, at least 11 nucleobase pairs, at least 12 nucleobase pairs, at least 13 nucleobase pairs, at least 14 nucleobase pairs, at least 15 nucleobase pairs, at least 16 nucleobase pairs, at least 17 nucleobase pairs, at least 18 nucleobase pairs, at least 19 nucleobase pairs, at least 20 nucleobase pairs, at least 21 nucleobase pairs, at least 22 nucleobase pairs, at least 23 nucleobase pairs, at least 24 nucleobase pairs, at least 25 nucleobase pairs, at least 26 nucleobase pairs, at least 27 nucleobase pairs, at least 28 nucleobase pairs, at least 29 nucleobase pairs, at least 30 nucleobase pairs, at least 31 nucleobase pairs, at least 32 nucleobase pairs, at least 33 nucleobase pairs, at least 34 nucleobase pairs, at least 35 nucleobase pairs, at least 36 nucleobase pairs, at least 37 nucleobase pairs, at least 37 nucleobase pairs, at least 38 nucleobase pairs, at least 39 nucleobase pairs, or at least 40 nucleobase pairs, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 94. The method of embodiment 92 or 93, wherein the contiguous region becomes replaced by the duplex comprising the edited portion.
    • 95. The method of any one of embodiments 73-94, wherein the first and/or second DNA synthesis template is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 96. The method of any one of embodiments 73-95, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence have the same lengths.
    • 97. The method of any one of embodiments 73-95, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence have different lengths.
    • 98. The method of any one of embodiments 73-97, wherein the region of complementarity is at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, at least 50%, at least 45%, at least 40%, at least 35%, at least 30%, at least 25%, at least 20%, at least 15%, or at least 10% of the total length of the first single-stranded DNA sequence or the second single-stranded DNA sequence.
    • 99. The method of any one of embodiments 73-98, wherein the region of complementarity is at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length, and is either less than or equal to the length of the desired DNA sequence and complement thereof.
    • 100. The method of any one of embodiments 73-99, wherein the region of complementarity of the first single-stranded DNA sequence and the second single-stranded DNA sequence form the duplex.
    • 101. The method of any one of embodiments 73-100, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence are each single-stranded DNA strands having 3′ ends.
    • 102. The method of embodiment 94, wherein the replacement of the contiguous region by the duplex comprising the edited portion results in an insertion, deletion, or replacement of DNA at the target site.
    • 103. The method of any one of embodiments 73-102, wherein the duplex comprising an edit comprising one or more recombinase sites.
    • 104. The method of embodiment 103 further comprising providing a recombinase to promote recombinase-mediated editing of the DNA sequence at the target site to be edited.
    • 105. A pair of pegRNAs for use in dual prime editing, wherein the pair comprises:
      • a. a first prime editing guide RNA (first PEgRNA) that binds to a first binding site on a first strand of a double-stranded DNA sequence upstream of a target site to be edited, wherein the first PEgRNA comprises a first spacer sequence, a first gRNA core, a first DNA synthesis template, and a first primer binding site, wherein the first DNA synthesis template encodes a first single-stranded DNA sequence comprising an edited portion;
      • b. a second prime editing guide RNA (second PEgRNA) that binds to a second binding site on a second strand of the double-stranded DNA sequence downstream of the target site to be edited, wherein the second PEgRNA comprises a second spacer sequence, a second gRNA core, a second DNA synthesis template, and a second primer binding site, wherein the second DNA synthesis template encodes a second single-stranded DNA sequence comprising the edited portion, wherein the second single-stranded DNA sequence comprises a region of complementarity to the first single-stranded DNA sequence.
    • 106. The pair of pegRNAs of embodiment 105, wherein the first PEgRNA comprises the structure:
      • 5′-[first gRNA spacer sequence]-[first gRNA core]-[first DNA synthesis template]-[first primer binding site]-3′; and
      • wherein the second PEgRNA comprises the structure:
      • 5′-[second gRNA spacer sequence]-[second gRNA core]-[second DNA synthesis template]-[second primer binding site]-3′.
    • 107. The pair of pegRNAs of embodiment 105 or 106, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence form a duplex that integrates into the double-stranded DNA sequence at the target site to be edited.
    • 108. The pair of pegRNAs of embodiment 107, wherein the integration of the duplex results in an insertion, deletion, or replacement of DNA at the target site.
    • 109. The pair of pegRNAs of embodiment 108, wherein the insertion is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 110. The pair of pegRNAs of embodiment 109, wherein the deletion is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides, or more in length.
    • 111. The pair of pegRNAs of embodiment 110, wherein the replacement is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides, or more in length.
    • 112. A polynucleotide encoding a pair of pegRNAs of any one of embodiments 105-111.
    • 113. A vector comprising the polynucleotide of embodiment 112 for expressing the pair of pegRNAs in a cell.
    • 114. A cell comprising the vector of embodiment 113.
    • 115. A pharmaceutical composition comprising a pair of pegRNAs of any one of embodiments 105-111, a vector of embodiment 113, or a cell of embodiment 114, and a pharmaceutical excipient.
    • 116. A pair of prime editor complexes, said pair comprises:
      • a. a first prime editor complex, comprising:
        • i. a first prime editor comprising a first nucleic acid programmable DNA binding protein (napDNAbp) and a first polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a first prime editing guide RNA (first PEgRNA) that binds to a first target sequence on the strand of the genomic DNA sequence upstream of the target site;
      • b. a second prime editor complex, comprising:
        • i. a second prime editor comprising a second nucleic acid programmable DNA binding protein (second napDNAbp) and a second polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a second prime editing guide RNA (second PEgRNA) that binds to a second target sequence on the second strand of the genomic DNA sequence downstream of the target site.
    • 117. The pair of prime editor complexes of embodiment 116, wherein the prime editor of the first prime editor complex, the second prime editor complex, or both the first prime editor complex and the second prime editor complex is a fusion protein comprising the napDNAbp and the polypeptide having an RNA-dependent DNA polymerase activity.
    • 118. A polynucleotide encoding a pair of prime editor complexes of embodiment 116 or 117.
    • 119. A vector comprising the polynucleotide of embodiment 118 for expressing the pair of prime editor complexes in a cell.
    • 120. A cell comprising the vector of embodiment 119.
    • 121. A pharmaceutical composition comprising a pair of prime editor complexes of embodiment 116 or 117, a vector of embodiment 119, or a cell of embodiment 120, and a pharmaceutical excipient.
    • 122. The system of any one of embodiments 1-62, or the method of any one of embodiments 68-98, wherein integration of the duplex comprising the edited portion results in an insertion, deletion, or replacement of DNA at the target site.
    • 123. The system or method of embodiment 122, wherein the insertion is a peptide tag.
    • 124. The system or method of embodiment 123, wherein the peptide tag is a poly-histidine (e.g., HHHHHH) (SEQ ID NO: 252-262), FLAG (e.g., DYKDDDDK) (SEQ ID NO: 2), V5 (e.g., GKPIPNPLLGLDST) (SEQ ID NO: 3), GCN4, HA (e.g., YPYDVPDYA) (SEQ ID NO: 5), Myc (e.g. EQKLISEED) (SEQ ID NO: 6), or GST.
    • 125. The system or method of embodiment 123, wherein the peptide tag has an amino acid sequence selected from the group consisting of SEQ ID NO: 1-6, 245-249, 252-262, 264-273, 275-276, 281, 278-288, and 622.
    • 126. The system or method of embodiment 122, wherein the insertion is an immunoepitope tag.
    • 127. The system or method of embodiment 126, wherein the immunoepitope tag is selected from the group consisting of: tetanus toxoid (SEQ ID NO: 396); diphtheria toxin mutant CRM197 (SEQ ID NO: 630); mumps immunoepitope 1 (SEQ ID NO: 400); mumps immunoepitope 2 (SEQ ID NO: 402); mumps immunoeptitope 3 (SEQ ID NO: 404); rubella virus (SEQ ID NO: 406); hemagglutinin (SEQ ID NO: 408); neuraminidase (SEQ ID NO: 410); TAP1 (SEQ ID NO: 412); TAP2 (SEQ ID NO: 414); hemagglutinin epitopes toward class I HLA (SEQ ID NO: 416); neuraminidase epitopes toward class I HLA (SEQ ID NO: 418); hemagglutinin epitopes toward class II HLA (SEQ ID NO: 420); neuraminidase epitopes toward class II HLA (SEQ ID NO: 422); hemagglutinin epitope H5N1-bound class I and class II HLA (SEQ ID NO: 424); and neuraminidase epitope H5N1-bound class I and class II HLA (SEQ ID NO: 426).
    • 128. The system or method of embodiment 122, wherein the insertion is a dimerization domain
    • 129. The system or method of embodiment 128, wherein the dimerization domain is small molecule binding domain of FKBP12 of SEQ ID NO: 488, FKBP12-F36V of SEQ ID NO: 489, or cyclophilin of SEQ ID NOs: 490 or 493-494.
    • 130. A system for simultaneously editing both strands of a double-stranded DNA sequence at a target site to be edited, the system comprising:
      • a) a first prime editor complex comprising:
        • i. a first prime editor comprising a first nucleic acid programmable DNA binding protein (first napDNAbp) and a first polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a first prime editing guide RNA (first PEgRNA) that binds to a first binding site on a first strand of the double-stranded DNA sequence upstream of the target site to be edited;
      • b) a second prime editor complex comprising:
        • iii. a second prime editor comprising a second nucleic acid programmable DNA binding protein (second napDNAbp) and a second polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • iv. a second prime editing guide RNA (second PEgRNA) that binds to a second binding site on a second strand of the double-stranded DNA sequence upstream of the target site to be edited;
      • c) a third prime editor complex comprising:
        • iii. a third prime editor comprising a third nucleic acid programmable DNA binding protein (third napDNAbp) and a third polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • iv. a third prime editing guide RNA (third PEgRNA) that binds to a third binding site on the first strand of the double-stranded DNA sequence downstream of the target site to be edited;
      • d) a fourth prime editor complex comprising:
        • iii. a fourth prime editor comprising a fourth nucleic acid programmable DNA binding protein (fourth napDNAbp) and a fourth polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • iv. a fourth prime editing guide RNA (fourth PEgRNA) that binds to a fourth binding site on the second strand of the double-stranded DNA sequence downstream of the target site to be edited;
          • wherein the first PEgRNA comprises a first DNA synthesis template encoding a first single-stranded DNA sequence, the second PEgRNA comprises a second DNA synthesis template encoding a second single-stranded DNA sequence, the third PEgRNA comprises a third DNA synthesis template encoding a third single-stranded DNA sequence, and the fourth PEgRNA comprises a fourth DNA synthesis template encoding a fourth single-stranded DNA sequence;
          • wherein the first and the third single-stranded DNA sequence each comprise a region of complementarity to the other; and wherein, wherein the second and the fourth single-stranded DNA sequence each comprise a region of complementarity to the other.
    • 131. A system for editing one or more double-stranded DNA sequences, the system comprising:
      • e) a first prime editor complex comprising:
        • iii. a first prime editor comprising a first nucleic acid programmable DNA binding protein (first napDNAbp) and a first polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • iv. a first prime editing guide RNA (first PEgRNA) that binds to a first binding site on a first strand of a first double-stranded DNA sequence at a first target site to be edited;
      • f) a second prime editor complex comprising:
        • v. a second prime editor comprising a second nucleic acid programmable DNA binding protein (second napDNAbp) and a second polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • vi. a second prime editing guide RNA (second PEgRNA) that binds to a second binding site on a second strand of the first double-stranded DNA sequence at the first target site to be edited;
      • g) a third prime editor complex comprising:
        • v. a third prime editor comprising a third nucleic acid programmable DNA binding protein (third napDNAbp) and a third polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • vi. a third prime editing guide RNA (third PEgRNA) that binds to a first binding site on a first strand of a second double-stranded DNA sequence at a second target site to be edited;
      • h) a fourth prime editor complex comprising:
        • v. a fourth prime editor comprising a fourth nucleic acid programmable DNA binding protein (fourth napDNAbp) and a fourth polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • vi. a fourth prime editing guide RNA (fourth PEgRNA) that binds to a second binding site on a second strand of the second double-stranded DNA sequence at the second target site to be edited;
          • wherein the first PEgRNA comprises a first DNA synthesis template encoding a first single-stranded DNA sequence, the second PEgRNA comprises a second DNA synthesis template encoding a second single-stranded DNA sequence, the third PEgRNA comprises a third DNA synthesis template encoding a third single-stranded DNA sequence, and the fourth PEgRNA comprises a fourth DNA synthesis template encoding a fourth single-stranded DNA sequence;
          • wherein the first and the third single-stranded DNA sequence each comprise a region of complementarity to the other; and wherein, wherein the second and the fourth single-stranded DNA sequence each comprise a region of complementarity to the other.
    • 132. The system of embodiment 130 or 131, wherein the prime editor of the first prime editor complex, the second prime editor complex, the third prime editor complex, an/or the fourth prime editor complex is a fusion protein comprising the napDNAbp and the polypeptide having an RNA-dependent DNA polymerase activity
    • 133. The system of any one of embodiments 129-132, wherein
      • (i) the first PEgRNA further comprises a first spacer sequence, first gRNA core, and a first primer binding site;
      • (ii) the second PEgRNA further comprises a second spacer sequence, a second gRNA core, and a second primer binding site;
      • (iii) the third PEgRNA further comprises a third spacer sequence, a third gRNA core, and a third primer binding site; and
      • (iv) the fourth PEgRNA comprises a fourth spacer sequence, a fourth gRNA core, a fourth DNA synthesis template, and a fourth primer binding site.
    • 134. The system of embodiment 131, wherein the region of complementarity of the first and third single-stranded DNA sequence, and/or the region of complementarity of the second and fourth single-stranded DNA sequence, form a duplex.
    • 135. the system of embodiment 134, wherein the first spacer sequence binds to the first binding site of the first double stranded DNA sequence upstream of the first target site to be edited and the second spacer sequence binds to the second binding site on the first double stranded DNA sequence downstream of the first target site to be edited.
    • 136. the system of embodiment 134 or 135, wherein the third spacer sequence binds to the first binding site of the second double stranded DNA sequence upstream of the second target site to be edited and the forth spacer sequence binds to the second binding site on the second double stranded DNA sequence downstream of the second target site to be edited.
    • 137. the system of any one of embodiments 134-136, wherein the first and second binding sites define two ends of a first contiguous region of the first double-stranded DNA.
    • 138. The system of any one of embodiments 134-137, wherein the third and fourth binding sites define two ends of a second contiguous region of the second double-stranded DNA.
    • 139. The system of embodiment 138, wherein the contiguous region between the first and second binding sites or the third and fourth binding sites is least 10 nucleobase pairs, at least 11 nucleobase pairs, at least 12 nucleobase pairs, at least 13 nucleobase pairs, at least 14 nucleobase pairs, at least 15 nucleobase pairs, at least 16 nucleobase pairs, at least 17 nucleobase pairs, at least 18 nucleobase pairs, at least 19 nucleobase pairs, at least 20 nucleobase pairs, at least 21 nucleobase pairs, at least 22 nucleobase pairs, at least 23 nucleobase pairs, at least 24 nucleobase pairs, at least 25 nucleobase pairs, at least 26 nucleobase pairs, at least 27 nucleobase pairs, at least 28 nucleobase pairs, at least 29 nucleobase pairs, at least 30 nucleobase pairs, at least 31 nucleobase pairs, at least 32 nucleobase pairs, at least 33 nucleobase pairs, at least 34 nucleobase pairs, at least 35 nucleobase pairs, at least 36 nucleobase pairs, at least 37 nucleobase pairs, at least 37 nucleobase pairs, at least 38 nucleobase pairs, at least 39 nucleobase pairs, or at least 40 nucleobase pairs, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 140. The system of any one of embodiments 134-140, wherein the region of complementarity of the first single-stranded DNA sequence and the second single-stranded DNA sequence, and/or the region of complementarity of the second single-stranded DNA sequence and the fourth single-stranded DNA sequence, form a duplex.
    • 141. The system of embodiment 140, wherein the duplex is incorporated in the first double stranded DNA and/or the second double stranded DNA.
    • 142. The system of any one of embodiments 134-141, wherein the system results in replacement of the first contiguous region by the second contiguous region into the first double-stranded DNA.
    • 143. The system of any one of embodiments 134-141, wherein the system results in exchange of the first contiguous region and the second contiguous region between the first double-stranded DNA and the second double-stranded DNA.
    • 144. The system of any one of embodiments 130-143, wherein the first prime editor complex, the second prime editor complex, the third prime editor complex, and the fourth prime editor complex comprise the same prime editor.
    • 145. The system of any one of embodiments 130-144, wherein the first, second, third, and fourth polypeptide comprising an RNA-dependent DNA polymerase activity are the same.
    • 146. The system of any one of embodiments 130-145, wherein the first, second, third, and/or fourth napDNAbp is a Cas9 domain or variant thereof.
    • 147. The system of any one of embodiments 130-146, wherein the first, second, third, and/or fourth napDNAbp is a nuclease active Cas9 domain, a nuclease inactive Cas9 domain, or a Cas9 nickase domain or a variant thereof.
    • 148. The system of any one of embodiments 130-147, wherein the first, second, third, and/or fourth napDNAbp is selected from the group consisting of: Cas9, Cas12e, Cas12d, Cas12a, Cas12b1, Cas13a, Cas12c, and Argonaute.
    • 149. The system of any one of embodiments 130-148, wherein the first, second, third, and/or fourth napDNAbp has nickase activity.
    • 150. The system of any one of embodiments 130-149, wherein the first, second, third, and/or fourth napDNAbp comprises an amino acid sequence of any one of SEQ ID NOs: 2-65, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 2-65.
    • 151. The system of any one of embodiments 130-150, wherein the first, second, third, and/or fourth polypeptide having an RNA-dependent DNA polymerase activity is a reverse transcriptase.
    • 152. The system of any one of embodiments 130-151, wherein the first, second, third, and/or fourth polypeptide having an RNA-dependent DNA polymerase activity comprises an amino acid sequence of any one of SEQ ID NOs: 37, 68-79, 82-98, 81, 98, and 110 or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 37, 68-79, 82-98, 81, 98, and 110.
    • 153. The system of any one of embodiments 130-152, wherein
      • (i) the first prime editor further comprises a linker that joins the first napDNAbp and the first polypeptide having the RNA-dependent DNA polymerase activity;
      • (ii) the second prime editor further comprises a linker that joins the second napDNAbp and the second polypeptide having the RNA-dependent DNA polymerase activity;
      • (iii) the third prime editor further comprises a linker that joins the third napDNAbp and the third polypeptide having the RNA-dependent DNA polymerase activity; and/or
      • (iv) the fourth prime editor further comprises a linker that joins the fourth napDNAbp and the fourth polypeptide having the RNA-dependent DNA polymerase activity.
    • 154. The system of embodiment 153, wherein the linker comprises an amino acid sequence of any one of SEQ ID NOs: 119-128, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 119-128.
    • 155. The system of embodiment 153 or 154, wherein the linker is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 38, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 amino acids in length.
    • 156. The system of any one of embodiments 130-155, wherein the first, second, third, and/or fourth PEgRNA comprises a nucleotide sequence of any one of SEQ ID NOs: 192-203, or a nucleotide sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 192-203.
    • 157. The system of any one of embodiments 130-156, wherein the first spacer sequence binds to a first binding site on a first strand of the double-stranded DNA sequence upstream from the target site to be edited.
    • 158. The system of any one of embodiments 130-157, wherein the second spacer sequence of the second prime editor complex binds to a second binding site on a second strand of the double-stranded DNA sequence upstream from the target site to be edited.
    • 159. The system of any one of embodiments 130-158, wherein the third spacer sequence binds to a third binding site on a first strand of the double-stranded DNA sequence downstream from the target site to be edited.
    • 160. The system of any one of embodiments 130-159, wherein the fourth spacer sequence of the fourth prime editor complex binds to a fourth binding site on a second strand of the double-stranded DNA sequence downstream from the target site to be edited.
    • 161. The system of embodiment 160, wherein the binding of the spacer sequence of the first and second prime editor complexes in the presence of a PAM sequence results in the nicking of the first and second strands, respectively, at a nick site proximal to the PAM sequences on each strand by the first and/or second napDNAbps of each first and second prime editor complexes.
    • 162. The system of embodiment 161, wherein the binding of the spacer sequence of the third and fourth prime editor complexes in the presence of a PAM sequence results in the nicking of the first and second strands, respectively, at a nick site proximal to the PAM sequences on each strand by the third and/or fourth napDNAbps of each third and fourth prime editor complexes.
    • 163. The system of embodiment 160 or 162, wherein the first and third binding sites, or the second and fourth binding sites, define two ends of a contiguous region of double-stranded DNA.
    • 164. The system of embodiment 163, wherein the contiguous region between the first and third binding sites or the second and fourth binding sites is least 10 nucleobase pairs, at least 11 nucleobase pairs, at least 12 nucleobase pairs, at least 13 nucleobase pairs, at least 14 nucleobase pairs, at least 15 nucleobase pairs, at least 16 nucleobase pairs, at least 17 nucleobase pairs, at least 18 nucleobase pairs, at least 19 nucleobase pairs, at least 20 nucleobase pairs, at least 21 nucleobase pairs, at least 22 nucleobase pairs, at least 23 nucleobase pairs, at least 24 nucleobase pairs, at least 25 nucleobase pairs, at least 26 nucleobase pairs, at least 27 nucleobase pairs, at least 28 nucleobase pairs, at least 29 nucleobase pairs, at least 30 nucleobase pairs, at least 31 nucleobase pairs, at least 32 nucleobase pairs, at least 33 nucleobase pairs, at least 34 nucleobase pairs, at least 35 nucleobase pairs, at least 36 nucleobase pairs, at least 37 nucleobase pairs, at least 37 nucleobase pairs, at least 38 nucleobase pairs, at least 39 nucleobase pairs, or at least 40 nucleobase pairs, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 165. The system of any one of embodiments 130-164, wherein the region of complementarity of the first single-stranded DNA sequence and the third single-stranded DNA sequence, and/or the region of complementarity of the second single-stranded DNA sequence and the fourth single-stranded DNA sequence, form a duplex.
    • 166. The system of embodiment 164 or 165, wherein the contiguous region becomes replaced by the duplex.
    • 167. The system of embodiment 164 or 165, wherein the duplex is incorporated in the double-stranded DNA at the target site.
    • 168. The system of embodiment any one of embodiments 130-167, wherein the first, second, third, and/or fourth DNA synthesis template is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 169. The system of any one of embodiments 130-168, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence, and/or the third single-stranded DNA sequence and the fourth single-stranded DNA sequence, have the same lengths.
    • 170. The system of any one of embodiments 130-168, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence, and/or the third single-stranded DNA sequence and the fourth single-stranded DNA sequence, have different lengths.
    • 171. The system of any one of embodiments 130-170, wherein the region of complementarity of the first, second, third, and/or fourth single-stranded DNA sequence is at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, at least 50%, at least 45%, at least 40%, at least 35%, at least 30%, at least 25%, at least 20%, at least 15%, or at least 10% of the total length of the first single-stranded DNA sequence and the third single-stranded DNA sequence, or the second single-stranded DNA sequence and the fourth single-stranded DNA sequence.
    • 172. The system of any one of embodiments 130-171, wherein the region of complementarity of the first, second, third, and/or fourth single-stranded DNA sequence is at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length, and is either less than or equal to the length of the first single-stranded DNA sequence and the third single-stranded DNA sequence, and the second single-stranded DNA sequence and the fourth single-stranded DNA sequence.
    • 173. The system of any one of embodiments 130-172, wherein the first, second, third, and fourth single-stranded DNA sequence are each single-stranded DNA strands having 3′ ends.
    • 174. The system of embodiment 167, wherein the incorporation of the duplex comprising the edited portion results in an insertion, deletion, or replacement of DNA at the target site.
    • 175. A polynucleotide encoding the first, second, third, and fourth prime editors of any one of embodiments 130-174.
    • 176. A polynucleotide encoding the first, second, third, and fourth PEgRNA of any of embodiments 130-174.
    • 177. A vector comprising a polynucleotide of embodiment 175 or 176.
    • 178. A cell comprising a polynucleotide of embodiment 175 or 176 or a vector of embodiment 177.
    • 179. A pharmaceutical composition comprising a polynucleotide of embodiment 175 or 176, a vector of embodiment 177, or a cell of embodiment 178, and a pharmaceutical excipient.
    • 180. A kit comprising a polynucleotide of embodiment 175 or 176 or a vector of embodiment 177 and instructions for simultaneously editing both strands of a double-stranded DNA sequence at a target site to be edited.
    • 181. A method for simultaneously editing first and second complementary strands of a double-stranded DNA sequence at a target site, the method comprising contacting the double-stranded DNA sequence with a pair of prime editor complexes, the pair comprising:
      • (a) a first prime editor complex, comprising:
        • i. a first prime editor comprising a first nucleic acid programmable DNA binding protein (first napDNAbp) and a first polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a first prime editing guide RNA (first PEgRNA) that binds to a first target sequence on the first strand of the genomic DNA sequence upstream of the target site;
      • (b) a second prime editor complex, comprising:
        • i. a second prime editor comprising a second nucleic acid programmable DNA binding protein (second napDNAbp) and a second polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a second prime editing guide RNA (second PEgRNA) that binds to a second target sequence on the second strand of the genomic DNA sequence upstream of the target site;
      • (c) a third prime editor complex, comprising:
        • i. a third prime editor comprising a third nucleic acid programmable DNA binding protein (third napDNAbp) and a third polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a third prime editing guide RNA (third PEgRNA) that binds to a third target sequence on the first strand of the genomic DNA sequence downstream of the target site;
      • (d) a fourth prime editor complex, comprising:
        • i. a fourth prime editor comprising a second nucleic acid programmable DNA binding protein (fourth napDNAbp) and a fourth polypeptide comprising an RNA-dependent DNA polymerase activity; and
        • ii. a fourth prime editing guide RNA (fourth PEgRNA) that binds to a fourth target sequence on the second strand of the genomic DNA sequence downstream of the target site;
      • wherein the first prime editor complex causes a first nick at the first target sequence and the subsequent polymerization of a first single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the first nick;
      • wherein the second prime editor complex causes a second nick at the second target sequence and the subsequent polymerization of a second single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the second nick;
      • wherein the third prime editor complex causes a third nick at the third target sequence and the subsequent polymerization of a third single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the third nick;
      • wherein the fourth prime editor complex causes a fourth nick at the fourth target sequence and the subsequent polymerization of a fourth single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the fourth nick;
      • wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence are reverse complements over at least a region of complementarity and form a duplex, wherein the duplex replaces the nicked first and second complementary strands of the double-stranded DNA sequence; and
      • wherein the third single-stranded DNA sequence and the fourth single-stranded DNA sequence are reverse complements over at least a region of complementarity and form a duplex, wherein the duplex replaces the nicked first and second complementary strands of the double-stranded DNA sequence.
    • 182. The method of embodiment 181, wherein the prime editor of the first prime editor complex, the second prime editor complex, the third prime editor complex, and/or the fourth prime editor complex is a fusion protein comprising the napDNAbp and the polypeptide having an RNA-dependent DNA polymerase activity.
    • 183. The method of embodiment 181 or 182, wherein the first prime editor complex, the second prime editor complex, the third prime editor complex, and the fourth prime editor complex comprise the same prime editor.
    • 184. The method of embodiment 181 or 182, wherein the first, second, third, and/or fourth prime editor complex comprise a different prime editor.
    • 185. The method of any one of embodiments 181-184, wherein first, second, third, and/or fourth napDNAbp is a Cas9 domain or variant thereof.
    • 186. The method of any one of embodiments 181-185, wherein first, second, third, and/or fourth napDNAbp is a nuclease active Cas9 domain, a nuclease inactive Cas9 domain, or a Cas9 nickase domain or variant thereof.
    • 187. The method of any one of embodiments 181-184, wherein the first, second, third, and/or fourth napDNAbp is selected from the group consisting of: Cas9, Cas12e, Cas12d, Cas12a, Cas12b1, Cas13a, Cas12c.
    • 188. The method of any one of embodiments 181-187, wherein the first, second, third, and/or fourth napDNAbp has a nickase activity.
    • 189. The method of any one of embodiments 181-188, wherein the first, second, third, and/or fourth napDNAbp comprises an amino acid sequence of any one of SEQ ID NOs: 2-65, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 2-65.
    • 190. The method of any one of embodiments 181-189, wherein the first, second, third, and/or fourth polypeptide having an RNA-dependent DNA polymerase activity is a reverse transcriptase.
    • 191. The method of any one of embodiments 181-190, wherein the first, second, third, and/or fourth polypeptide having an RNA-dependent DNA polymerase activity comprises an amino acid sequence of any one of SEQ ID NOs: 37, 68-79, 82-98, 81, 98, and 110 or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 37, 68-79, 82-98, 81, 98, and 110.
    • 192. The method of any one of embodiments 181-191, wherein
      • (i) the first prime editor further comprises a linker that joins the first napDNAbp and the first polypeptide having the RNA-dependent DNA polymerase activity;
      • (ii) the second prime editor further comprises a linker that joins the second napDNAbp and the second polypeptide having the RNA-dependent DNA polymerase activity;
      • (iii) the third prime editor further comprises a linker that joins the third napDNAbp and the third polypeptide having the RNA-dependent DNA polymerase activity; and/or
      • (iv) the fourth prime editor further comprises a linker that joins the fourth napDNAbp and the fourth polypeptide having the RNA-dependent DNA polymerase activity.
    • 193. The method of embodiment 192, wherein the linker comprises an amino acid sequence of any one of SEQ ID NOs: 119-128, or an amino acid sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 119-128.
    • 194. The method of embodiment 192 or 193, wherein the linker is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 38, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 amino acids in length.
    • 195. The method of any one of embodiments 181-191, wherein the first, second, third, and/or fourth PEgRNA comprises a nucleotide sequence of any one of SEQ ID NOs: 192-203, or a nucleotide sequence having at least an 80%, 85%, 90%, 95%, or 99% sequence identity with any one of SEQ ID NOs: 192-203.
    • 196. The method of any one of embodiments 181-195, wherein the first spacer sequence of the first prime editor complex binds to a first binding site on a first strand of the double-stranded DNA sequence upstream from the target site to be edited.
    • 197. The method of any one of embodiments 181-196, wherein the second spacer sequence of the second prime editor complex binds to a second binding site on a second strand of the double-stranded DNA sequence downstream from the target site to be edited.
    • 198. The method of any one of embodiments 181-197, wherein the third spacer sequence of the third prime editor complex binds to a third binding site on a first strand of the double-stranded DNA sequence upstream from the target site to be edited.
    • 199. The method of any one of embodiments 181-198, wherein the fourth spacer sequence of the fourth prime editor complex binds to a fourth binding site on a second strand of the double-stranded DNA sequence downstream from the target site to be edited.
    • 200. The method of embodiment 197, wherein the binding of the first and second spacer sequences of the first and second prime editor complexes in the presence of a PAM sequence results in the nicking of the first and second strands, respectively, at a nick site proximal to the PAM sequence on each strand by the napDNAbps of each first and second prime editor complex.
    • 201. The method of embodiment 199, wherein the binding of the third and fourth spacer sequences of the third and fourth prime editor complexes in the presence of a PAM sequence results in the nicking of the first and second strands, respectively, at a nick site proximal to the PAM sequence on each strand by the napDNAbps of each third and fourth prime editor complex.
    • 202. The method of embodiment 200 or 201, wherein the first and second binding sites, and/or the third and fourth binding sites, define two ends of a contiguous region of double-stranded DNA comprising the target site to be edited.
    • 203. The method of embodiment 202, wherein the contiguous region between the first and second binding sites, and/or the contiguous region between the third and fourth binding sites, is least 10 nucleobase pairs, at least 11 nucleobase pairs, at least 12 nucleobase pairs, at least 13 nucleobase pairs, at least 14 nucleobase pairs, at least 15 nucleobase pairs, at least 16 nucleobase pairs, at least 17 nucleobase pairs, at least 18 nucleobase pairs, at least 19 nucleobase pairs, at least 20 nucleobase pairs, at least 21 nucleobase pairs, at least 22 nucleobase pairs, at least 23 nucleobase pairs, at least 24 nucleobase pairs, at least 25 nucleobase pairs, at least 26 nucleobase pairs, at least 27 nucleobase pairs, at least 28 nucleobase pairs, at least 29 nucleobase pairs, at least 30 nucleobase pairs, at least 31 nucleobase pairs, at least 32 nucleobase pairs, at least 33 nucleobase pairs, at least 34 nucleobase pairs, at least 35 nucleobase pairs, at least 36 nucleobase pairs, at least 37 nucleobase pairs, at least 37 nucleobase pairs, at least 38 nucleobase pairs, at least 39 nucleobase pairs, or at least 40 nucleobase pairs, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 204. The method of embodiment 202 or 203, wherein the contiguous region becomes replaced by the duplex.
    • 205. The method of any one of embodiments 181-204, wherein the first, second, third, and/or fourth DNA synthesis template is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 206. The method of any one of embodiments 181-205, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence, and/or the third single-stranded-DNA sequence and the fourth single-stranded DNA sequence, have the same lengths.
    • 207. The method of any one of embodiments 181-206, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence, and/or the third single-stranded-DNA sequence and the fourth single-stranded DNA sequence, have different lengths.
    • 208. The method of any one of embodiments 181-207, wherein the region of complementarity is at least 99%, at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, at least 50%, at least 45%, at least 40%, at least 35%, at least 30%, at least 25%, at least 20%, at least 15%, or at least 10% of the total length of the first single-stranded DNA sequence and the second single-stranded DNA sequence, or the third single-stranded DNA sequence and the fourth single-stranded DNA sequence.
    • 209. The method of any one of embodiments 181-208, wherein the region of complementarity is at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 210. The method of any one of embodiments 181-209, wherein the region of complementarity of the first single-stranded DNA sequence and the second single-stranded DNA sequence, and/or the third single-stranded-DNA sequence and the fourth single-stranded DNA sequence, form the duplex.
    • 211. The method of any one of embodiments 181-210, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence, and/or the third single-stranded-DNA sequence and the fourth single-stranded DNA sequence, are each single-stranded DNA strands having 3′ ends.
    • 212. The method of embodiment 204, wherein the replacement of the contiguous region by the duplex comprising the edited portion results in an insertion, deletion, or replacement of DNA at the target site.
    • 213. The method of any one of embodiments 181-212, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence are on ends of a target DNA sequence, and wherein the third single-stranded DNA sequence and the fourth single-stranded DNA sequence are on opposite ends of the same target DNA sequence.
    • 214. The method of embodiment 213, wherein the target DNA sequence is inverted.
    • 215. The method of any one of embodiments 181-214 further comprising providing a circular DNA donor.
    • 216. The method of embodiment 215, wherein the first single-stranded DNA sequence and the third single-stranded DNA sequence are on opposite ends of the target DNA sequence, and wherein the second single-stranded DNA sequence and the fourth single-stranded DNA sequence are on the circular DNA donor.
    • 217. The method of embodiment 216, wherein a portion of the circular DNA donor between the second single-stranded DNA sequence and the fourth single-stranded DNA sequence replaces the target sequence between the first single-stranded DNA sequence and the third single-stranded DNA sequence.
    • 218. The method of any one of embodiments 181-214, wherein the first single-stranded DNA sequence and the third single-stranded DNA sequence are on a first nucleic acid molecule, and wherein the second single-stranded DNA sequence and the fourth single-stranded DNA sequence are on a second nucleic acid molecule.
    • 219. The method of embodiment 218, wherein a portion of the first nucleic acid molecule between the first single-stranded DNA sequence and the third single-stranded DNA sequence is incorporated into the second nucleic acid molecule, and wherein a portion of the second nucleic acid molecule between the second single-stranded DNA sequence and the fourth single-stranded DNA sequence is incorporated into the first nucleic acid molecule.
    • 220. The method of embodiment 218 or 219, wherein the first nucleic acid molecule is a first chromosome, and the second nucleic acid molecule is a second chromosome.
    • 221. A set of PEgRNAs for use in multi-flap prime editing, wherein the PEgRNAs comprise:
      • (a) a first prime editing guide RNA (first PEgRNA) that binds to a first binding site on a first strand of a double-stranded DNA sequence upstream of a target site to be edited, wherein the first PEgRNA comprises a first spacer sequence, first gRNA core, a first DNA synthesis template, and a first primer binding site, wherein the first DNA synthesis template encodes a first single-stranded DNA sequence;
      • (b) a second prime editing guide RNA (second PEgRNA) that binds to a second binding site on a second strand of the double-stranded DNA sequence upstream of the target site to be edited, wherein the second PEgRNA comprises a second spacer sequence, second gRNA core, a second DNA synthesis template, and a second primer binding site, wherein the second DNA synthesis template encodes a second single-stranded DNA sequence comprising the edited portion, wherein the second single-stranded DNA sequence is complementary to the first single-stranded DNA sequence.
      • (c) a third prime editing guide RNA (third PEgRNA) that binds to a third binding site on a first strand of a double-stranded DNA sequence downstream of a target site to be edited, wherein the third PEgRNA comprises a third spacer sequence, third gRNA core, a third DNA synthesis template, and a third primer binding site, wherein the third DNA synthesis template encodes a third single-stranded DNA sequence;
      • (d) a fourth prime editing guide RNA (fourth PEgRNA) that binds to a fourth binding site on a second strand of the double-stranded DNA sequence downstream of the target site to be edited, wherein the fourth PEgRNA comprises a fourth spacer sequence, fourth gRNA core, a fourth DNA synthesis template, and a fourth primer binding site, wherein the fourth DNA synthesis template encodes a fourth single-stranded DNA sequence, wherein the fourth single-stranded DNA sequence comprises a region of complementarity to the third single-stranded DNA sequence.
    • 222. The set of PEgRNAs of embodiment 221, wherein
      • the first PEgRNA comprises the structure:
      • 5′-[first gRNA spacer sequence]-[first gRNA core]-[first DNA synthesis template]-[first primer binding site]-3′;
      • the second PEgRNA comprises the structure:
      • 5′-[second gRNA spacer sequence]-[second gRNA core]-[second DNA synthesis template]-[second primer binding site]-3′;
      • the third PEgRNA comprises the structure:
      • 5′-[third gRNA spacer sequence]-[third gRNA core]-[third DNA synthesis template]-[third primer binding site]-3′; and
      • the fourth PEgRNA comprises the structure:
      • 5′-[fourth gRNA spacer sequence]-[fourth gRNA core]-[fourth DNA synthesis template]-[fourth primer binding site]-3′.
    • 223. The set of PEgRNAs of embodiment 221 or 222, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence, and/or the third single-stranded DNA sequence and the fourth single-stranded DNA sequence, form a duplex that becomes integrated into the double-stranded DNA sequence at the target site to be edited.
    • 224. The set of PEgRNAs of any one of embodiments 221-223, wherein the integration of the duplex results in an insertion, deletion, or replacement of DNA at the target site.
    • 225. The set of PEgRNAs of embodiment 224, wherein the insertion is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides, at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides in length.
    • 226. The set of PEgRNAs of embodiment 224, wherein the deletion is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides is at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides, or more in length.
    • 227. The set of PEgRNAs of embodiment 224, wherein the replacement is at least 1 nucleotide, at least 2 nucleotides, at least 3 nucleotides, at least 4 nucleotides, at least 5 nucleotides, at least 6 nucleotides, at least 7 nucleotides, at least 8 nucleotides, at least 9 nucleotides, at least 10 nucleotides, at least 11 nucleotides, at least 12 nucleotides, at least 13 nucleotides, at least 14 nucleotides, at least 15 nucleotides, at least 16 nucleotides, at least 17 nucleotides, at least 18 nucleotides, at least 19 nucleotides at least 20 nucleotides, at least 21 nucleotides, at least 22 nucleotides, at least 23 nucleotides, at least 24 nucleotides, at least 25 nucleotides, at least 26 nucleotides, at least 27 nucleotides, at least 28 nucleotides, at least 29 nucleotides, at least 30 nucleotides, at least 31 nucleotides, at least 32 nucleotides, at least 33 nucleotides, at least 34 nucleotides, at least 35 nucleotides, at least 36 nucleotides, at least 37 nucleotides, at least 38 nucleotides, at least 39 nucleotides, at least 40 nucleotides, at least 41 nucleotides, at least 42 nucleotides, at least 43 nucleotides, at least 44 nucleotides, at least 45 nucleotides, at least 46 nucleotides, at least 47 nucleotides, at least 48 nucleotides, at least 49 nucleotides, at least 50 nucleotides, at least 51 nucleotides, at least 52 nucleotides, at least 53 nucleotides, at least 54 nucleotides, at least 55 nucleotides, at least 56 nucleotides, at least 57 nucleotides, at least 58 nucleotides, at least 59 nucleotides, at least 60 nucleotides, at least 61 nucleotides, at least 62 nucleotides, at least 63 nucleotides, at least 64 nucleotides, at least 65 nucleotides, at least 66 nucleotides, at least 67 nucleotides, at least 68 nucleotides, at least 69 nucleotides, at least 70 nucleotides, at least 71 nucleotides, at least 72 nucleotides, at least 73 nucleotides, at least 74 nucleotides, at least 75 nucleotides, at least 76 nucleotides, at least 77 nucleotides, at least 78 nucleotides, at least 79 nucleotides, at least 80 nucleotides, at least 81 nucleotides, at least 82 nucleotides, at least 83 nucleotides, at least 84 nucleotides, at least 85 nucleotides, at least 86 nucleotides, at least 87 nucleotides, at least 88 nucleotides, at least 89 nucleotides, at least 90 nucleotides, at least 91 nucleotides, at least 92 nucleotides, at least 93 nucleotides, at least 94 nucleotide at least 95 nucleotides, at least 96 nucleotides, at least 97 nucleotides, at least 98 nucleotides, at least 99 nucleotides, at least 100 nucleotides, at least 101 nucleotides, at least 102 nucleotides, at least 103 nucleotides, at least 104 nucleotides, at least 105 nucleotides, at least 106 nucleotides, at least 107 nucleotides, at least 108 nucleotides, at least 109 nucleotides, or at least 110 nucleotides, or more in length.
    • 228. A polynucleotide encoding a set of PEgRNAs of any of embodiments 221-227.
    • 229. A vector comprising the polynucleotide of embodiment 228 for expressing the plurality of PEgRNAs in a cell.
    • 230. A cell comprising the vector of embodiment 229.
    • 231. A pharmaceutical composition comprising the plurality of PEgRNAs of any one of embodiments 221-227, a vector of embodiment 229, or a cell of embodiment 230, and a pharmaceutical excipient.
    • 232. A method for modifying a genome in a cell, comprising:
      • (i) contacting a genome with a dual-flap prime editing system, said system comprising a first prime editor/pegRNA complex capable of installing a first 3′ nucleic acid flap at a first nick site on a first strand of a target site and a second prime editor/pegRNA complex capable of installing a second 3′ nucleic acid flap at a second nick site on a second strand of a target site,
        • wherein the first 3′ nucleic acid flap and the second 3′ nucleic acid flap are reverse complement sequences that form a duplex, and
        • wherein the duplex comprises a first recombinase site, and
        • wherein the cell installs the duplex comprising the first recombinase site at the target site in place of an endogenous duplex positioned between the first and second nick sites;
      • (ii) contacting the first recombinase site with a donor nucleic acid comprising a second recombinase site; and
      • (iii) contacting the genome with a recombinase capable recombination between the first and second recombinase sites, thereby integrating the donor nucleic acid at the target site in the genome.


[9] Methods

Methods for Multi-Flap Prime Editing


In one aspect, the present disclosure provides methods for simultaneously editing a first and a second complementary strands of a double-stranded DNA sequence at a target site, said method comprising contacting the double-stranded DNA sequence with a pair of prime editor complexes, said pair comprising:

    • a. a first prime editor complex, comprising:
      • i. a first prime editor comprising a first nucleic acid programmable DNA binding protein (napDNAbp) and a first polypeptide comprising an RNA-dependent DNA polymerase activity; and
      • ii. a first prime editing guide RNA (first PEgRNA) that binds to a first binding site on the first strand of the genomic DNA sequence upstream of the target site;
    • b. a second prime editor complex, comprising:
      • i. a second prime editor comprising a second nucleic acid programmable DNA binding protein (second napDNAbp) and a second polypeptide comprising an RNA-dependent DNA polymerase activity; and
      • ii. a second prime editing guide RNA (second PEgRNA) that binds to a second binding site on the second strand of the genomic DNA sequence downstream of the target site;
    • wherein the first prime editor complex causes a first nick at a sequence complementary to the first binding site and the subsequent polymerization of a first single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the first nick;
    • wherein the second prime editor complex causes a second nick at a sequence complementary to the second binding site and the subsequent polymerization of a second single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the second nick;
    • wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence are reverse complements over at least a region of complementarity and form a duplex comprising an edit; and
    • wherein the duplex replaces the nicked first and second complementary strands of the double-stranded DNA sequence.


In another aspect, the present disclosure provides methods for simultaneously editing first and second complementary strands of a double-stranded DNA sequence at a target site, the method comprising contacting the double-stranded DNA sequence with a pair of prime editor complexes, the pair comprising:

    • (a) a first prime editor complex, comprising:
      • i. a first prime editor comprising a first nucleic acid programmable DNA binding protein (first napDNAbp) and a first polypeptide comprising an RNA-dependent DNA polymerase activity; and
      • ii. a first prime editing guide RNA (first PEgRNA) that binds to a first target sequence on the first strand of the genomic DNA sequence upstream of the target site;
    • (b) a second prime editor complex, comprising:
      • i. a second prime editor comprising a second nucleic acid programmable DNA binding protein (second napDNAbp) and a second polypeptide comprising an RNA-dependent DNA polymerase activity; and
      • ii. a second prime editing guide RNA (second PEgRNA) that binds to a second target sequence on the second strand of the genomic DNA sequence upstream of the target site;
    • (c) a third prime editor complex, comprising:
      • i. a third prime editor comprising a third nucleic acid programmable DNA binding protein (third napDNAbp) and a third polypeptide comprising an RNA-dependent DNA polymerase activity; and
      • ii. a third prime editing guide RNA (third PEgRNA) that binds to a third target sequence on the first strand of the genomic DNA sequence downstream of the target site;
    • (d) a fourth prime editor complex, comprising:
      • i. a fourth prime editor comprising a second nucleic acid programmable DNA binding protein (fourth napDNAbp) and a fourth polypeptide comprising an RNA-dependent DNA polymerase activity; and
      • ii. a fourth prime editing guide RNA (fourth PEgRNA) that binds to a fourth target sequence on the second strand of the genomic DNA sequence downstream of the target site;
    • wherein the first prime editor complex causes a first nick at the first target sequence and the subsequent polymerization of a first single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the first nick;
    • wherein the second prime editor complex causes a second nick at the second target sequence and the subsequent polymerization of a second single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the second nick;
    • wherein the third prime editor complex causes a third nick at the third target sequence and the subsequent polymerization of a third single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the third nick;
    • wherein the fourth prime editor complex causes a fourth nick at the fourth target sequence and the subsequent polymerization of a fourth single-stranded DNA sequence having a 3′-end from the available 5′-end formed by the fourth nick;
    • wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence are reverse complements over at least a region of complementarity and form a duplex, wherein the duplex replaces the nicked first and second complementary strands of the double-stranded DNA sequence; and
    • wherein the third single-stranded DNA sequence and the fourth single-stranded DNA sequence are reverse complements over at least a region of complementarity and form a duplex, wherein the duplex replaces the nicked first and second complementary strands of the double-stranded DNA sequence.


This Specification describes multi-flap prime editing systems (including, for example, a dual-flap prime editing system and a quadruple-flap prime editing system) that address the challenges associated with flap equilibration and subsequent incorporation of the edit into the non-edited complementary genomic DNA strand by simultaneously editing both DNA strands. In the dual-flap prime editing system, two PEgRNAs are used to target opposite strands of a genomic site and direct the synthesis of two complementary 3′ flaps containing edited DNA sequence (FIG. 90). In the quadruple-flap prime editing system, four PEgRNAs are used and direct the synthesis of four 3′ flaps, two of which are complementary to one another and the other two of which are complementary to one another (FIG. 95, FIG. 96A, and FIG. 98A). Unlike classical prime editing, there is no requirement for the pair of edited DNA strands (3′ flaps) to directly compete with 5′ flaps in endogenous genomic DNA, as the complementary edited strand is available for hybridization instead. Since both strands of the duplex are synthesized as edited DNA, the multi-flap prime editing system obviates the need for the replacement of the non-edited complementary DNA strand required by classical prime editing. Instead, cellular DNA repair machinery need only excise the paired 5′ flaps (original genomic DNA) and ligate the paired 3′ flaps (edited DNA) into the locus. Therefore, there is also no need to include sequences homologous to genomic DNA in the newly synthesized DNA strands, allowing selective hybridization of the new strands and facilitating edits that contain minimal genomic homology. Nuclease-active versions of multi-flap prime editors that cut both strands of DNA could also be used to accelerate the removal of the original DNA sequence.


Like classical prime editing, multi-flap prime editing is a versatile and precise genome editing method that directly writes new genetic information into a specified DNA site using a nucleic acid programmable DNA binding protein (“napDNAbp”) working in association with a polymerase (i.e., in the form of a fusion protein or otherwise provided in trans with the napDNAbp), wherein the prime editing system is programmed with a prime editing (PE) guide RNA (“PEgRNA”) that both specifies the target site and templates the synthesis of the desired edit in the form of a replacement DNA strand by way of an extension (either DNA or RNA) engineered onto a guide RNA (e.g., at the 5′ or 3′ end, or at an internal portion of a guide RNA). The replacement strand containing the desired edit (e.g., a single nucleobase substitution) shares the same sequence as the endogenous strand of the target site to be edited (with the exception that it includes the desired edit). Through DNA repair and/or replication machinery, the endogenous strand of the target site is replaced by the newly synthesized replacement strand containing the desired edit. In some cases, prime editing may be thought of as a “search-and-replace” genome editing technology since the dual prime editors, as described herein, not only search and locate the desired target site to be edited, but at the same time, encode a replacement strand containing a desired edit which is installed in place of the corresponding target site endogenous DNA strand.


In dual-flap prime editing, a double-stranded DNA sequence is contacted at a target site with a first and a second prime editor complex. Each complex comprises a fusion protein and a PEgRNA. In some embodiments, each fusion protein comprises a nucleic acid programmable DNA binding protein (napDNAbp) and a polypeptide having an RNA-dependent DNA polymerase activity (e.g., a reverse transcriptase), and each PEgRNA comprises a spacer sequence, a gRNA core, a DNA synthesis template, and a primer binding site. Each DNA synthesis template can encode a single-stranded DNA sequence, which may comprise an edited portion of one or more nucleotides. The two single-stranded DNA sequences encoded may be complementary to one another and form a duplex, which can integrate into the target site to be edited. The various elements of the prime editor complexes (e.g., fusion proteins, napDNAbp, polymerase, PEgRNAs, etc.) may comprise any of the embodiments of the systems disclosed herein.


In quadruple-flap prime editing, a double-stranded DNA sequence is contacted at a target site with a first, a second, a third, and a fourth prime editor complex. Each complex comprises a fusion protein and a PEgRNA. In some embodiments, each fusion protein comprises a nucleic acid programmable DNA binding protein (napDNAbp) and a polypeptide having an RNA-dependent DNA polymerase activity (e.g., a reverse transcriptase), and each PEgRNA comprises a spacer sequence, a gRNA core, a DNA synthesis template, and a primer binding site. Each DNA synthesis template encodes a single-stranded DNA sequence. The two single-stranded DNA sequences encoded may be complementary to one another and form a duplex, which can integrate into the target site to be edited. The various elements of the prime editor complexes (e.g., fusion proteins, napDNAbp, polymerase, PEgRNAs, etc.) may comprise any of the embodiments of the systems disclosed herein.


The methods for multi-flap prime editing provided herein can be used for numerous applications. For example, they can be used to facilitate the inversion of a target DNA sequence. In this application, a first single-stranded DNA sequence encoded by the DNA synthesis template of the first PEgRNA and a second single-stranded DNA sequence encoded by the DNA synthesis template of the second PEgRNA are on opposite ends of a target DNA sequence, and a third single-stranded DNA sequence encoded by the DNA synthesis template of the third PEgRNA and a fourth single-stranded DNA sequence encoded by the DNA synthesis template of a fourth PEgRNA are on opposite ends of the same target DNA sequence.


In some embodiments, the methods for multi-flap prime editing provided herein further comprise providing a circular DNA donor, part of which can be integrated into a double-stranded nucleic acid at a target site. In this application, a first single-stranded DNA sequence encoded by the DNA synthesis template of the first PEgRNA and a third single-stranded DNA sequence encoded by the DNA synthesis template of the third PEgRNA are on opposite ends of the target DNA sequence, and a second single-stranded DNA sequence encoded by the DNA synthesis template of the second PEgRNA and a fourth single-stranded DNA sequence encoded by the DNA synthesis template of the fourth PEgRNA are on the circular DNA donor. The portion of the circular DNA donor between the second single-stranded DNA sequence and the fourth single-stranded DNA sequence can form a duplex, which replaces the target DNA sequence between the first single-stranded DNA sequence and the third single-stranded DNA sequence.


In another application, the methods for multi-flap prime editing provided herein allow for translocation of a target DNA sequence from a first nucleic acid molecule (e.g., a first chromosome) to a second nucleic acid molecule (e.g., a second chromosome). In this application, a first single-stranded DNA sequence encoded by the DNA synthesis template of the first PEgRNA and a third single-stranded DNA sequence encoded by the DNA synthesis template of the third PEgRNA are on a first nucleic acid molecule, and a second single-stranded DNA sequence encoded by the DNA synthesis template of the second PEgRNA and a fourth single-stranded DNA sequence encoded by the DNA synthesis template of the fourth PEgRNA are on a second nucleic acid molecule. The portion of the first nucleic acid molecule between the first single-stranded DNA sequence and the third single-stranded DNA sequence can be incorporated into the second nucleic acid molecule, and the portion of the second nucleic acid molecule between the second single-stranded DNA sequence and the fourth single-stranded DNA sequence is incorporated into the first nucleic acid molecule.


Other Methods


The instant disclosure provides methods for the treatment of a subject diagnosed with a disease associated with or caused by a point mutation, or other mutations (e.g., deletion, insertion, inversion, duplication, etc.) that can be corrected by the multi-flap prime editing system provided herein, as exemplified, but not limited to prion disease (e.g., Example 5 herein), trinucleotide repeat expansion disease (e.g., Example 3 herein), or CDKL5 Deficiency Disorder (CDD) (e.g., Example 23 herein).


Virtually any disease-causing genetic defect may be repaired by using multi-flap prime editing, which includes the selection of an appropriate prime editor fusion protein (including a napDNAbp and a polymerase (e.g., a reverse transcriptase), and designing of an appropriate PEgRNA designed to (a) target the appropriate target DNA containing an edit site, and (b) provide a template for the synthesis of a single strand of DNA from the 3′ end of the nick site that includes the desired edit which displaces and replaces the endogenous strand immediately downstream of the nick site. Multi-flap prime editing can be used, without limitation, to (a) install mutation-correcting changes to a nucleotide sequence, (b) install protein and RNA tags, (c) install immunoepitopes on proteins of interest, (d) install inducible dimerization domains in proteins, (e) install or remove sequences to alter that activity of a biomolecule, (f) install recombinase target sites to direct specific genetic changes, and (g) mutagenesis of a target sequence by using an error-prone RT.


The method of treating a disorder can involve as an early step the design of an appropriate PEgRNA and prime editor fusion protein in accordance with the methods described herein, which include a number of considerations that may be taken into account, such as:

    • (a) the target sequence, i.e., the nucleotide sequence in which one or more nucleobase modifications are desired to be installed by the prime editor;
    • (b) the location of the cut site within the target sequence, i.e., the specific nucleobase position at which the prime editor will induce a single-stand nick to create a 3′ end RT primer sequence on one side of the nick and the 5′ end endogenous flap on the other side of the nick (which ultimately is removed by FEN1 or equivalent thereto and replaced by the 3′ ssDNA flap. The cut site creates the 3′ end primer sequence which becomes extended by the polymerase of the PE fusion protein (e.g., a RT enzyme) during RNA-dependent DNA polymerization to create the 3′ ssDNA flap containing the desired edit, which then replaces the 5′ endogenous DNA flap in the target sequence.
    • (c) the available PAM sequences (including the canonical SpCas9 PAM sites, as well as non-canonical PAM sites recognized by Cas9 variants and equivalents with expanded or differing PAM specificities);
    • (d) the spacing between the available PAM sequences and the location of the cut site in the PAM strand;
    • (e) the particular Cas9, Cas9 variant, or Cas9 equivalent of the prime editor available to be used (which in part is dictated by the available PAM);
    • (f) the sequence and length of the primer binding site;
    • (g) the sequence and length of the edit template;
    • (h) the sequence and length of the homology arm;
    • (i) the spacer sequence and length; and
    • (j) the gRNA core sequence.


A suitable PEgRNA, and optionally a nicking-sgRNA design guide for second-site nicking, can be designed by way of the following exemplarily step-by-step set of instructions which takes into account one or more of the above considerations. The steps reference the examples shown in FIGS. 70A-70I.

    • 1. Define the target sequence and the edit. Retrieve the sequence of the target DNA region (˜200 bp) centered around the location of the desired edit (point mutation, insertion, deletion, or combination thereof). See FIG. 70A.
    • 2. Locate target PAMs. Identify PAMs in the proximity to the desired edit location. PAMs can be identified on either strand of DNA proximal to the desired edit location. While PAMs close to the edit position are preferred (i.e., wherein the nick site is less than 30 nt from the edit position, or less than 29 nt, 28 nt, 27 nt, 26 nt, 25 nt, 24 nt, 23 nt, 22 nt, 21 nt, 20 nt, 19 nt, 18 nt, 17 nt, 16 nt, 15 nt, 14 nt, 13 nt, 12 nt, 11 nt, 10 nt, 9 nt, 8 nt, 7 nt, 6 nt, 5 nt, 4 nt, 3 nt, or 2 nt from the edit position to the nick site), it is possible to install edits using protospacers and PAMs that place the nick ≥30 nt from the edit position. See FIG. 70B.
    • 3. Locate the nick sites. For each PAM being considered, identify the corresponding nick site and on which strand. For Sp Cas9 H840A nickase, cleavage occurs in the PAM-containing strand between the 3′ and 4th bases 5′ to the NGG PAM. All edited nucleotides must exist 3′ of the nick site, so appropriate PAMs must place the nick 5′ to the target edit on the PAM-containing strand. In the example shown below, there are two possible PAMs. For simplicity, the remaining steps will demonstrate the design of a PEgRNA using PAM 1 only. See FIG. 70C.
    • 4. Design the spacer sequence. The protospacer of SpCas9 corresponds to the 20 nucleotides 5′ to the NGG PAM on the PAM-containing strand. Efficient Pol III transcription initiation requires a G to be the first transcribed nucleotide. If the first nucleotide of the protospacer is a G, the spacer sequence for the PEgRNA is simply the protospacer sequence. If the first nucleotide of the protospacer is not a G, the spacer sequence of the PEgRNA is G followed by the protospacer sequence. See FIG. 70D.
    • 5. Design a primer binding site (PBS). Using the starting allele sequence, identify the DNA primer on the PAM-containing strand. The 3′ end of the DNA primer is the nucleotide just upstream of the nick site (i.e. the 4th base 5′ to the NGG PAM for Sp Cas9). As a general design principle for use with PE2 and PE3, a PEgRNA primer binding site (PBS) containing 12 to 13 nucleotides of complementarity to the DNA primer can be used for sequences that contain ˜40-60% GC content. For sequences with low GC content, longer (14- to 15-nt) PBSs should be tested. For sequences with higher GC content, shorter (8- to 11-nt) PBSs should be tested. Optimal PBS sequences should be determined empirically, regardless of GC content. To design a length-p PBS sequence, take the reverse complement of the first p nucleotides 5′ of the nick site in the PAM-containing strand using the starting allele sequence. See FIG. 70E.
    • 6. Design an RT template (or DNA synthesis template). The RT template (or DNA synthesis template where the polymerase is not reverse transcriptase) encodes the designed edit and homology to the sequence adjacent to the edit. In one embodiment, these regions correspond to the DNA synthesis template of FIG. 3D and FIG. 3E, wherein the DNA synthesis template comprises the “edit template” and the “homology arm.” Optimal RT template lengths vary based on the target site. For short-range edits (positions +1 to +6), it is recommended to test a short (9 to 12 nt), a medium (13 to 16 nt), and a long (17 to 20 nt) RT template. For long-range edits (positions +7 and beyond), it is recommended to use RT templates that extend at least 5 nt (preferably 10 or more nt) past the position of the edit to allow for sufficient 3′ DNA flap homology. For long-range edits, several RT templates should be screened to identify functional designs. For larger insertions and deletions (≥5 nt), incorporation of greater 3′ homology (˜20 nt or more) into the RT template is recommended. Editing efficiency is typically impaired when the RT template encodes the synthesis of a G as the last nucleotide in the reverse transcribed DNA product (corresponding to a C in the RT template of the PEgRNA). As many RT templates support efficient prime editing, avoidance of G as the final synthesized nucleotide is recommended when designing RT templates. To design a length-r RT template sequence, use the desired allele sequence and take the reverse complement of the first r nucleotides 3′ of the nick site in the strand that originally contained the PAM. Note that compared to SNP edits, insertion or deletion edits using RT templates of the same length will not contain identical homology. See FIG. 70F.
    • 7. Assemble the full PEgRNA sequence. Concatenate the PEgRNA components in the following order (5′ to 3′): spacer, scaffold, RT template and PBS. See FIG. 70G.
    • 8. Designing nicking-sgRNAs for PE3. Identify PAMs on the non-edited strand upstream and downstream of the edit. Optimal nicking positions are highly locus-dependent and should be determined empirically. In general, nicks placed 40 to 90 nucleotides 5′ to the position across from the PEgRNA-induced nick lead to higher editing yields and fewer indels. A nicking sgRNA has a spacer sequence that matches the 20-nt protospacer in the starting allele, with the addition of a 5′-G if the protospacer does not begin with a G. See FIG. 70H.
    • 9. Designing PE3b nicking-sgRNAs. If a PAM exists in the complementary strand and its corresponding protospacer overlaps with the sequence targeted for editing, this edit could be a candidate for the PE3b system. In the PE3b system, the spacer sequence of the nicking-sgRNA matches the sequence of the desired edited allele, but not the starting allele. The PE3b system operates efficiently when the edited nucleotide(s) falls within the seed region (˜10 nt adjacent to the PAM) of the nicking-sgRNA protospacer. This prevents nicking of the complementary strand until after installation of the edited strand, preventing competition between the PEgRNA and the sgRNA for binding the target DNA. PE3b also avoids the generation of simultaneous nicks on both strands, thus reducing indel formation significantly while maintaining high editing efficiency. PE3b sgRNAs should have a spacer sequence that matches the 20-nt protospacer in the desired allele, with the addition of a 5′ G if needed. See FIG. 70I.


The above step-by-step process for designing a suitable PEgRNA and a second-site nicking sgRNA is not meant to be limiting in any way. The disclosure contemplates variations of the above-described step-by-step process which would be derivable therefrom by a person of ordinary skill in the art.


Once a suitable PEgRNA and PE fusion protein are selected/designed, they may be administered by a suitable methodology, such as by vector-based transfection (in which one or more vectors comprising DNA encoding the PEgRNA and the PE fusion protein and which are expressed within a cell upon transfection with the vectors), direct delivery of the PE fusion protein complexed with the PEgRNA (e.g., RNP delivery) in a delivery format (e.g., lipid particles, nanoparticles), or by a mRNA-based delivery system. Such methods are described herein in the present disclosure and any know method may be utilized.


The PEgRNA and PE fusion protein (or together, referred to as the PE complex) can be delivered to a cell in a therapeutically effective amount such that upon contacting the target DNA of interest, the desired edit becomes installed therein.


Any disease is conceivably treatable by such methods so long as delivery to the appropriate cells is feasible. The person having ordinary skill in the art will be able to choose and/or select a PE delivery methodology to suit the intended purpose and the intended target cells.


For example, in some embodiments, a method is provided that comprises administering to a subject having such a disease, e.g., a cancer associated with a point mutation as described above, an effective amount of the multi-flap prime editing system described herein that corrects the point mutation or introduces a deactivating mutation into a disease-associated gene as mediated by homology-directed repair in the presence of a donor DNA molecule comprising desired genetic change. In some embodiments, a method is provided that comprises administering to a subject having such a disease, e.g., a cancer associated with a point mutation as described above, an effective amount of the multi-flap prime editing system described herein that corrects the point mutation or introduces a deactivating mutation into a disease-associated gene. In some embodiments, the disease is a proliferative disease. In some embodiments, the disease is a genetic disease. In some embodiments, the disease is a neoplastic disease. In some embodiments, the disease is a metabolic disease. In some embodiments, the disease is a lysosomal storage disease. Other diseases that can be treated by correcting a point mutation or introducing a deactivating mutation into a disease-associated gene will be known to those of skill in the art, and the disclosure is not limited in this respect.


The instant disclosure provides methods for the treatment of additional diseases or disorders, e.g., diseases or disorders that are associated or caused by a point mutation that can be corrected by TPRT-mediated gene editing. Some such diseases are described herein, and additional suitable diseases that can be treated with the strategies and fusion proteins provided herein will be apparent to those of skill in the art based on the instant disclosure. Exemplary suitable diseases and disorders are listed below. It will be understood that the numbering of the specific positions or residues in the respective sequences depends on the particular protein and numbering scheme used. Numbering might be different, e.g., in precursors of a mature protein and the mature protein itself, and differences in sequences from species to species may affect numbering. One of skill in the art will be able to identify the respective residue in any homologous protein and in the respective encoding nucleic acid by methods well known in the art, e.g., by sequence alignment and determination of homologous residues. Exemplary suitable diseases and disorders include, without limitation: 2-methyl-3-hydroxybutyric aciduria; 3 beta-Hydroxysteroid dehydrogenase deficiency; 3-Methylglutaconic aciduria; 3-Oxo-5 alpha-steroid delta 4-dehydrogenase deficiency; 46,XY sex reversal, type 1, 3, and 5; 5-Oxoprolinase deficiency; 6-pyruvoyl-tetrahydropterin synthase deficiency; Aarskog syndrome; Aase syndrome; Achondrogenesis type 2; Achromatopsia 2 and 7; Acquired long QT syndrome; Acrocallosal syndrome, Schinzel type; Acrocapitofemoral dysplasia; Acrodysostosis 2, with or without hormone resistance; Acroerythrokeratoderma; Acromicric dysplasia; Acth-independent macronodular adrenal hyperplasia 2; Activated PI3K-delta syndrome; Acute intermittent porphyria; deficiency of Acyl-CoA dehydrogenase family, member 9; Adams-Oliver syndrome 5 and 6; Adenine phosphoribosyltransferase deficiency; Adenylate kinase deficiency; hemolytic anemia due to Adenylosuccinate lyase deficiency; Adolescent nephronophthisis; Renal-hepatic-pancreatic dysplasia; Meckel syndrome type 7; Adrenoleukodystrophy; Adult junctional epidermolysis bullosa; Epidermolysis bullosa, junctional, localisata variant; Adult neuronal ceroid lipofuscinosis; Adult neuronal ceroid lipofuscinosis; Adult onset ataxia with oculomotor apraxia; ADULT syndrome; Afibrinogenemia and congenital Afibrinogenemia; autosomal recessive Agammaglobulinemia 2; Age-related macular degeneration 3, 6, 11, and 12; Aicardi Goutieres syndromes 1, 4, and 5; Chilbain lupus 1; Alagille syndromes 1 and 2; Alexander disease; Alkaptonuria; Allan-Herndon-Dudley syndrome; Alopecia universalis congenital; Alpers encephalopathy; Alpha-1-antitrypsin deficiency; autosomal dominant, autosomal recessive, and X-linked recessive Alport syndromes; Alzheimer disease, familial, 3, with spastic paraparesis and apraxia; Alzheimer disease, types, 1, 3, and 4; hypocalcification type and hypomaturation type, IIA1 Amelogenesis imperfecta; Aminoacylase 1 deficiency; Amish infantile epilepsy syndrome; Amyloidogenic transthyretin amyloidosis; Amyloid Cardiomyopathy, Transthyretin-related; Cardiomyopathy; Amyotrophic lateral sclerosis types 1, 6, 15 (with or without frontotemporal dementia), 22 (with or without frontotemporal dementia), and 10; Frontotemporal dementia with TDP43 inclusions, TARDBP-related; Andermann syndrome; Andersen Tawil syndrome; Congenital long QT syndrome; Anemia, nonspherocytic hemolytic, due to G6PD deficiency; Angelman syndrome; Severe neonatal-onset encephalopathy with microcephaly; susceptibility to Autism, X-linked 3; Angiopathy, hereditary, with nephropathy, aneurysms, and muscle cramps; Angiotensin i-converting enzyme, benign serum increase; Aniridia, cerebellar ataxia, and mental retardation; Anonychia; Antithrombin III deficiency; Antley-Bixler syndrome with genital anomalies and disordered steroidogenesis; Aortic aneurysm, familial thoracic 4, 6, and 9; Thoracic aortic aneurysms and aortic dissections; Multisystemic smooth muscle dysfunction syndrome; Moyamoya disease 5; Aplastic anemia; Apparent mineralocorticoid excess; Arginase deficiency; Argininosuccinate lyase deficiency; Aromatase deficiency; Arrhythmogenic right ventricular cardiomyopathy types 5, 8, and 10; Primary familial hypertrophic cardiomyopathy; Arthrogryposis multiplex congenita, distal, X-linked; Arthrogryposis renal dysfunction cholestasis syndrome; Arthrogryposis, renal dysfunction, and cholestasis 2; Asparagine synthetase deficiency; Abnormality of neuronal migration; Ataxia with vitamin E deficiency; Ataxia, sensory, autosomal dominant; Ataxia-telangiectasia syndrome; Hereditary cancer-predisposing syndrome; Atransferrinemia; Atrial fibrillation, familial, 11, 12, 13, and 16; Atrial septal defects 2, 4, and 7 (with or without atrioventricular conduction defects); Atrial standstill 2; Atrioventricular septal defect 4; Atrophia bulborum hereditaria; ATR-X syndrome; Auriculocondylar syndrome 2; Autoimmune disease, multisystem, infantile-onset; Autoimmune lymphoproliferative syndrome, type 1a; Autosomal dominant hypohidrotic ectodermal dysplasia; Autosomal dominant progressive external ophthalmoplegia with mitochondrial DNA deletions 1 and 3; Autosomal dominant torsion dystonia 4; Autosomal recessive centronuclear myopathy; Autosomal recessive congenital ichthyosis 1, 2, 3, 4A, and 4B; Autosomal recessive cutis laxa type IA and 1B; Autosomal recessive hypohidrotic ectodermal dysplasia syndrome; Ectodermal dysplasia 11b; hypohidrotic/hair/tooth type, autosomal recessive; Autosomal recessive hypophosphatemic bone disease; Axenfeld-Rieger syndrome type 3; Bainbridge-Ropers syndrome; Bannayan-Riley-Ruvalcaba syndrome; PTEN hamartoma tumor syndrome; Baraitser-Winter syndromes 1 and 2; Barakat syndrome; Bardet-Biedl syndromes 1, 11, 16, and 19; Bare lymphocyte syndrome type 2, complementation group E; Bartter syndrome antenatal type 2; Bartter syndrome types 3, 3 with hypocalciuria, and 4; Basal ganglia calcification, idiopathic, 4; Beaded hair; Benign familial hematuria; Benign familial neonatal seizures 1 and 2; Seizures, benign familial neonatal, 1, and/or myokymia; Seizures, Early infantile epileptic encephalopathy 7; Benign familial neonatal-infantile seizures; Benign hereditary chorea; Benign scapuloperoneal muscular dystrophy with cardiomyopathy; Bernard-Soulier syndrome, types A1 and A2 (autosomal dominant); Bestrophinopathy, autosomal recessive; beta Thalassemia; Bethlem myopathy and Bethlem myopathy 2; Bietti crystalline corneoretinal dystrophy; Bile acid synthesis defect, congenital, 2; Biotinidase deficiency; Birk Bard mental retardation dysmorphism syndrome; Blepharophimosis, ptosis, and epicanthus inversus; Bloom syndrome; Borjeson-Forssman-Lehmann syndrome; Boucher Neuhauser syndrome; Brachydactyly types A1 and A2; Brachydactyly with hypertension; Brain small vessel disease with hemorrhage; Branched-chain ketoacid dehydrogenase kinase deficiency; Branchiootic syndromes 2 and 3; Breast cancer, early-onset; Breast-ovarian cancer, familial 1, 2, and 4; Brittle cornea syndrome 2; Brody myopathy; Bronchiectasis with or without elevated sweat chloride 3; Brown-Vialetto-Van laere syndrome and Brown-Vialetto-Van Laere syndrome 2; Brugada syndrome; Brugada syndrome 1; Ventricular fibrillation; Paroxysmal familial ventricular fibrillation; Brugada syndrome and Brugada syndrome 4; Long QT syndrome; Sudden cardiac death; Bull eye macular dystrophy; Stargardt disease 4; Cone-rod dystrophy 12; Bullous ichthyosiform erythroderma; Burn-Mckeown syndrome; Candidiasis, familial, 2, 5, 6, and 8; Carbohydrate-deficient glycoprotein syndrome type I and II; Carbonic anhydrase VA deficiency, hyperammonemia due to; Carcinoma of colon; Cardiac arrhythmia; Long QT syndrome, LQT1 subtype; Cardioencephalomyopathy, fatal infantile, due to cytochrome c oxidase deficiency; Cardiofaciocutaneous syndrome; Cardiomyopathy; Danon disease; Hypertrophic cardiomyopathy; Left ventricular noncompaction cardiomyopathy; Carnevale syndrome; Carney complex, type 1; Carnitine acylcarnitine translocase deficiency; Carnitine palmitoyltransferase I, II, II (late onset), and II (infantile) deficiency; Cataract 1, 4, autosomal dominant, autosomal dominant, multiple types, with microcornea, coppock-like, juvenile, with microcornea and glucosuria, and nuclear diffuse nonprogressive; Catecholaminergic polymorphic ventricular tachycardia; Caudal regression syndrome; Cd8 deficiency, familial; Central core disease; Centromeric instability of chromosomes 1,9 and 16 and immunodeficiency; Cerebellar ataxia infantile with progressive external ophthalmoplegi and Cerebellar ataxia, mental retardation, and dysequilibrium syndrome 2; Cerebral amyloid angiopathy, APP-related; Cerebral autosomal dominant and recessive arteriopathy with subcortical infarcts and leukoencephalopathy; Cerebral cavernous malformations 2; Cerebrooculofacioskeletal syndrome 2; Cerebro-oculo-facio-skeletal syndrome; Cerebroretinal microangiopathy with calcifications and cysts; Ceroid lipofuscinosis neuronal 2, 6, 7, and 10; Ch\xc3\xa9diak-Higashi syndrome, Chediak-Higashi syndrome, adult type; Charcot-Marie-Tooth disease types 1B, 2B2, 2C, 2F, 21, 2U (axonal), 1C (demyelinating), dominant intermediate C, recessive intermediate A, 2A2, 4C, 4D, 4H, IF, IVF, and X; Scapuloperoneal spinal muscular atrophy; Distal spinal muscular atrophy, congenital nonprogressive; Spinal muscular atrophy, distal, autosomal recessive, 5; CHARGE association; Childhood hypophosphatasia; Adult hypophosphatasia; Cholecystitis; Progressive familial intrahepatic cholestasis 3; Cholestasis, intrahepatic, of pregnancy 3; Cholestanol storage disease; Cholesterol monooxygenase (side-chain cleaving) deficiency; Chondrodysplasia Blomstrand type; Chondrodysplasia punctata 1, X-linked recessive and 2 X-linked dominant; CHOPS syndrome; Chronic granulomatous disease, autosomal recessive cytochrome b-positive, types 1 and 2; Chudley-McCullough syndrome; Ciliary dyskinesia, primary, 7, 11, 15, 20 and 22; Citrullinemia type I; Citrullinemia type I and II; Cleidocranial dysostosis; C-like syndrome; Cockayne syndrome type A; Coenzyme Q10 deficiency, primary 1, 4, and 7; Coffin Siris/Intellectual Disability; Coffin-Lowry syndrome; Cohen syndrome; Cold-induced sweating syndrome 1; COLE-CARPENTER SYNDROME 2; Combined cellular and humoral immune defects with granulomas; Combined d-2- and 1-2-hydroxyglutaric aciduria; Combined malonic and methylmalonic aciduria; Combined oxidative phosphorylation deficiencies 1, 3, 4, 12, 15, and 25; Combined partial and complete 17-alpha-hydroxylase/17,20-lyase deficiency; Common variable immunodeficiency 9; Complement component 4, partial deficiency of, due to dysfunctional c1 inhibitor; Complement factor B deficiency; Cone monochromatism; Cone-rod dystrophy 2 and 6; Cone-rod dystrophy amelogenesis imperfecta; Congenital adrenal hyperplasia and Congenital adrenal hypoplasia, X-linked; Congenital amegakaryocytic thrombocytopenia; Congenital aniridia; Congenital central hypoventilation; Hirschsprung disease 3; Congenital contractural arachnodactyly; Congenital contractures of the limbs and face, hypotonia, and developmental delay; Congenital disorder of glycosylation types 1B, 1D, 1G, 1H, 1J, 1K, 1N, 1P, 2C, 2J, 2K, IIm; Congenital dyserythropoietic anemia, type I and II; Congenital ectodermal dysplasia of face; Congenital erythropoietic porphyria; Congenital generalized lipodystrophy type 2; Congenital heart disease, multiple types, 2; Congenital heart disease; Interrupted aortic arch; Congenital lipomatous overgrowth, vascular malformations, and epidermal nevi; Non-small cell lung cancer; Neoplasm of ovary; Cardiac conduction defect, nonspecific; Congenital microvillous atrophy; Congenital muscular dystrophy; Congenital muscular dystrophy due to partial LAMA2 deficiency; Congenital muscular dystrophy-dystroglycanopathy with brain and eye anomalies, types A2, A7, A8, A11, and A14; Congenital muscular dystrophy-dystroglycanopathy with mental retardation, types B2, B3, B5, and B15; Congenital muscular dystrophy-dystroglycanopathy without mental retardation, type B5; Congenital muscular hypertrophy-cerebral syndrome; Congenital myasthenic syndrome, acetazolamide-responsive; Congenital myopathy with fiber type disproportion; Congenital ocular coloboma; Congenital stationary night blindness, type 1A, 1B, 1C, 1E, 1F, and 2A; Coproporphyria; Cornea plana 2; Corneal dystrophy, Fuchs endothelial, 4; Corneal endothelial dystrophy type 2; Corneal fragility keratoglobus, blue sclerae and joint hypermobility; Cornelia de Lange syndromes 1 and 5; Coronary artery disease, autosomal dominant 2; Coronary heart disease; Hyperalphalipoproteinemia 2; Cortical dysplasia, complex, with other brain malformations 5 and 6; Cortical malformations, occipital; Corticosteroid-binding globulin deficiency; Corticosterone methyloxidase type 2 deficiency; Costello syndrome; Cowden syndrome 1; Coxa plana; Craniodiaphyseal dysplasia, autosomal dominant; Craniosynostosis 1 and 4; Craniosynostosis and dental anomalies; Creatine deficiency, X-linked; Crouzon syndrome; Cryptophthalmos syndrome; Cryptorchidism, unilateral or bilateral; Cushing symphalangism; Cutaneous malignant melanoma 1; Cutis laxa with osteodystrophy and with severe pulmonary, gastrointestinal, and urinary abnormalities; Cyanosis, transient neonatal and atypical nephropathic; Cystic fibrosis; Cystinuria; Cytochrome c oxidase i deficiency; Cytochrome-c oxidase deficiency; D-2-hydroxyglutaric aciduria 2; Darier disease, segmental; Deafness with labyrinthine aplasia microtia and microdontia (LAMM); Deafness, autosomal dominant 3a, 4, 12, 13, 15, autosomal dominant nonsyndromic sensorineural 17, 20, and 65; Deafness, autosomal recessive 1A, 2, 3, 6, 8, 9, 12, 15, 16, 18b, 22, 28, 31, 44, 49, 63, 77, 86, and 89; Deafness, cochlear, with myopia and intellectual impairment, without vestibular involvement, autosomal dominant, X-linked 2; Deficiency of 2-methylbutyryl-CoA dehydrogenase; Deficiency of 3-hydroxyacyl-CoA dehydrogenase; Deficiency of alpha-mannosidase; Deficiency of aromatic-L-amino-acid decarboxylase; Deficiency of bisphosphoglycerate mutase; Deficiency of butyryl-CoA dehydrogenase; Deficiency of ferroxidase; Deficiency of galactokinase; Deficiency of guanidinoacetate methyltransferase; Deficiency of hyaluronoglucosaminidase; Deficiency of ribose-5-phosphate isomerase; Deficiency of steroid 11-beta-monooxygenase; Deficiency of UDPglucose-hexose-1-phosphate uridylyltransferase; Deficiency of xanthine oxidase; Dejerine-Sottas disease; Charcot-Marie-Tooth disease, types ID and IVF; Dejerine-Sottas syndrome, autosomal dominant; Dendritic cell, monocyte, B lymphocyte, and natural killer lymphocyte deficiency; Desbuquois dysplasia 2; Desbuquois syndrome; DFNA 2 Nonsyndromic Hearing Loss; Diabetes mellitus and insipidus with optic atrophy and deafness; Diabetes mellitus, type 2, and insulin-dependent, 20; Diamond-Blackfan anemia 1, 5, 8, and 10; Diarrhea 3 (secretory sodium, congenital, syndromic) and 5 (with tufting enteropathy, congenital); Dicarboxylic aminoaciduria; Diffuse palmoplantar keratoderma, Bothnian type; Digitorenocerebral syndrome; Dihydropteridine reductase deficiency; Dilated cardiomyopathy 1A, 1AA, 1C, 1G, 1BB, 1DD, 1FF, 1HH, 1I, 1KK, 1N, 1S, 1Y, and 3B; Left ventricular noncompaction 3; Disordered steroidogenesis due to cytochrome p450 oxidoreductase deficiency; Distal arthrogryposis type 2B; Distal hereditary motor neuronopathy type 2B; Distal myopathy Markesbery-Griggs type; Distal spinal muscular atrophy, X-linked 3; Distichiasis-lymphedema syndrome; Dominant dystrophic epidermolysis bullosa with absence of skin; Dominant hereditary optic atrophy; Donnai Barrow syndrome; Dopamine beta hydroxylase deficiency; Dopamine receptor d2, reduced brain density of; Dowling-degos disease 4; Doyne honeycomb retinal dystrophy; Malattia leventinese; Duane syndrome type 2; Dubin-Johnson syndrome; Duchenne muscular dystrophy; Becker muscular dystrophy; Dysfibrinogenemia; Dyskeratosis congenita autosomal dominant and autosomal dominant, 3; Dyskeratosis congenita, autosomal recessive, 1, 3, 4, and 5; Dyskeratosis congenita X-linked; Dyskinesia, familial, with facial myokymia; Dysplasminogenemia; Dystonia 2 (torsion, autosomal recessive), 3 (torsion, X-linked), 5 (Dopa-responsive type), 10, 12, 16, 25, 26 (Myoclonic); Seizures, benign familial infantile, 2; Early infantile epileptic encephalopathy 2, 4, 7, 9, 10, 11, 13, and 14; Atypical Rett syndrome; Early T cell progenitor acute lymphoblastic leukemia; Ectodermal dysplasia skin fragility syndrome; Ectodermal dysplasia-syndactyly syndrome 1; Ectopia lentis, isolated autosomal recessive and dominant; Ectrodactyly, ectodermal dysplasia, and cleft lip/palate syndrome 3; Ehlers-Danlos syndrome type 7 (autosomal recessive), classic type, type 2 (progeroid), hydroxylysine-deficient, type 4, type 4 variant, and due to tenascin-X deficiency; Eichsfeld type congenital muscular dystrophy; Endocrine-cerebroosteodysplasia; Enhanced s-cone syndrome; Enlarged vestibular aqueduct syndrome; Enterokinase deficiency; Epidermodysplasia verruciformis; Epidermolysa bullosa simplex and limb girdle muscular dystrophy, simplex with mottled pigmentation, simplex with pyloric atresia, simplex, autosomal recessive, and with pyloric atresia; Epidermolytic palmoplantar keratoderma; Familial febrile seizures 8; Epilepsy, childhood absence 2, 12 (idiopathic generalized, susceptibility to) 5 (nocturnal frontal lobe), nocturnal frontal lobe type 1, partial, with variable foci, progressive myoclonic 3, and X-linked, with variable learning disabilities and behavior disorders; Epileptic encephalopathy, childhood-onset, early infantile, 1, 19, 23, 25, 30, and 32; Epiphyseal dysplasia, multiple, with myopia and conductive deafness; Episodic ataxia type 2; Episodic pain syndrome, familial, 3; Epstein syndrome; Fechtner syndrome; Erythropoietic protoporphyria; Estrogen resistance; Exudative vitreoretinopathy 6; Fabry disease and Fabry disease, cardiac variant; Factor H, VII, X, v and factor viii, combined deficiency of 2, xiii, a subunit, deficiency; Familial adenomatous polyposis 1 and 3; Familial amyloid nephropathy with urticaria and deafness; Familial cold urticarial; Familial aplasia of the vermis; Familial benign pemphigus; Familial cancer of breast; Breast cancer, susceptibility to; Osteosarcoma; Pancreatic cancer 3; Familial cardiomyopathy; Familial cold autoinflammatory syndrome 2; Familial colorectal cancer; Familial exudative vitreoretinopathy, X-linked; Familial hemiplegic migraine types 1 and 2; Familial hypercholesterolemia; Familial hypertrophic cardiomyopathy 1, 2, 3, 4, 7, 10, 23 and 24; Familial hypokalemia-hypomagnesemia; Familial hypoplastic, glomerulocystic kidney; Familial infantile myasthenia; Familial juvenile gout; Familial Mediterranean fever and Familial mediterranean fever, autosomal dominant; Familial porencephaly; Familial porphyria cutanea tarda; Familial pulmonary capillary hemangiomatosis; Familial renal glucosuria; Familial renal hypouricemia; Familial restrictive cardiomyopathy 1; Familial type 1 and 3 hyperlipoproteinemia; Fanconi anemia, complementation group E, I, N, and O; Fanconi-Bickel syndrome; Favism, susceptibility to; Febrile seizures, familial, 11; Feingold syndrome 1; Fetal hemoglobin quantitative trait locus 1; FG syndrome and FG syndrome 4; Fibrosis of extraocular muscles, congenital, 1, 2, 3a (with or without extraocular involvement), 3b; Fish-eye disease; Fleck corneal dystrophy; Floating-Harbor syndrome; Focal epilepsy with speech disorder with or without mental retardation; Focal segmental glomerulosclerosis 5; Forebrain defects; Frank Ter Haar syndrome; Borrone Di Rocco Crovato syndrome; Frasier syndrome; Wilms tumor 1; Freeman-Sheldon syndrome; Frontometaphyseal dysplasia 1 and 3; Frontotemporal dementia; Frontotemporal dementia and/or amyotrophic lateral sclerosis 3 and 4; Frontotemporal Dementia Chromosome 3-Linked and Frontotemporal dementia ubiquitin-positive; Fructose-biphosphatase deficiency; Fuhrmann syndrome; Gamma-aminobutyric acid transaminase deficiency; Gamstorp-Wohlfart syndrome; Gaucher disease type 1 and Subacute neuronopathic; Gaze palsy, familial horizontal, with progressive scoliosis; Generalized dominant dystrophic epidermolysis bullosa; Generalized epilepsy with febrile seizures plus 3, type 1, type 2; Epileptic encephalopathy Lennox-Gastaut type; Giant axonal neuropathy; Glanzmann thrombasthenia; Glaucoma 1, open angle, e, F, and G; Glaucoma 3, primary congenital, d; Glaucoma, congenital and Glaucoma, congenital, Coloboma; Glaucoma, primary open angle, juvenile-onset; Glioma susceptibility 1; Glucose transporter type 1 deficiency syndrome; Glucose-6-phosphate transport defect; GLUT1 deficiency syndrome 2; Epilepsy, idiopathic generalized, susceptibility to, 12; Glutamate formiminotransferase deficiency; Glutaric acidemia IIA and IIB; Glutaric aciduria, type 1; Gluthathione synthetase deficiency; Glycogen storage disease 0 (muscle), II (adult form), IXa2, IXc, type 1A; type II, type IV, IV (combined hepatic and myopathic), type V, and type VI; Goldmann-Favre syndrome; Gordon syndrome; Gorlin syndrome; Holoprosencephaly sequence; Holoprosencephaly 7; Granulomatous disease, chronic, X-linked, variant; Granulosa cell tumor of the ovary; Gray platelet syndrome; Griscelli syndrome type 3; Groenouw corneal dystrophy type I; Growth and mental retardation, mandibulofacial dysostosis, microcephaly, and cleft palate; Growth hormone deficiency with pituitary anomalies; Growth hormone insensitivity with immunodeficiency; GTP cyclohydrolase I deficiency; Hajdu-Cheney syndrome; Hand foot uterus syndrome; Hearing impairment; Hemangioma, capillary infantile; Hematologic neoplasm; Hemochromatosis type 1, 2B, and 3; Microvascular complications of diabetes 7; Transferrin serum level quantitative trait locus 2; Hemoglobin H disease, nondeletional; Hemolytic anemia, nonspherocytic, due to glucose phosphate isomerase deficiency; Hemophagocytic lymphohistiocytosis, familial, 2; Hemophagocytic lymphohistiocytosis, familial, 3; Heparin cofactor II deficiency; Hereditary acrodermatitis enteropathica; Hereditary breast and ovarian cancer syndrome; Ataxia-telangiectasia-like disorder; Hereditary diffuse gastric cancer; Hereditary diffuse leukoencephalopathy with spheroids; Hereditary factors II, IX, VIII deficiency disease; Hereditary hemorrhagic telangiectasia type 2; Hereditary insensitivity to pain with anhidrosis; Hereditary lymphedema type I; Hereditary motor and sensory neuropathy with optic atrophy; Hereditary myopathy with early respiratory failure; Hereditary neuralgic amyotrophy; Hereditary Nonpolyposis Colorectal Neoplasms; Lynch syndrome I and II; Hereditary pancreatitis; Pancreatitis, chronic, susceptibility to; Hereditary sensory and autonomic neuropathy type IIB and IIA; Hereditary sideroblastic anemia; Hermansky-Pudlak syndrome 1, 3, 4, and 6; Heterotaxy, visceral, 2, 4, and 6, autosomal; Heterotaxy, visceral, X-linked; Heterotopia; Histiocytic medullary reticulosis; Histiocytosis-lymphadenopathy plus syndrome; Holocarboxylase synthetase deficiency; Holoprosencephaly 2, 3, 7, and 9; Holt-Oram syndrome; Homocysteinemia due to MTHFR deficiency, CBS deficiency, and Homocystinuria, pyridoxine-responsive; Homocystinuria-Megaloblastic anemia due to defect in cobalamin metabolism, cblE complementation type; Howel-Evans syndrome; Hurler syndrome; Hutchinson-Gilford syndrome; Hydrocephalus; Hyperammonemia, type III; Hypercholesterolaemia and Hypercholesterolemia, autosomal recessive; Hyperekplexia 2 and Hyperekplexia hereditary; Hyperferritinemia cataract syndrome; Hyperglycinuria; Hyperimmunoglobulin D with periodic fever; Mevalonic aciduria; Hyperimmunoglobulin E syndrome; Hyperinsulinemic hypoglycemia familial 3, 4, and 5; Hyperinsulinism-hyperammonemia syndrome; Hyperlysinemia; Hypermanganesemia with dystonia, polycythemia and cirrhosis; Hyperornithinemia-hyperammonemia-homocitrullinuria syndrome; Hyperparathyroidism 1 and 2; Hyperparathyroidism, neonatal severe; Hyperphenylalaninemia, bh4-deficient, a, due to partial pts deficiency, BH4-deficient, D, and non-pku; Hyperphosphatasia with mental retardation syndrome 2, 3, and 4; Hypertrichotic osteochondrodysplasia; Hypobetalipoproteinemia, familial, associated with apob32; Hypocalcemia, autosomal dominant 1; Hypocalciuric hypercalcemia, familial, types 1 and 3; Hypochondrogenesis; Hypochromic microcytic anemia with iron overload; Hypoglycemia with deficiency of glycogen synthetase in the liver; Hypogonadotropic hypogonadism 11 with or without anosmia; Hypohidrotic ectodermal dysplasia with immune deficiency; Hypohidrotic X-linked ectodermal dysplasia; Hypokalemic periodic paralysis 1 and 2; Hypomagnesemia 1, intestinal; Hypomagnesemia, seizures, and mental retardation; Hypomyelinating leukodystrophy 7; Hypoplastic left heart syndrome; Atrioventricular septal defect and common atrioventricular junction; Hypospadias 1 and 2, X-linked; Hypothyroidism, congenital, nongoitrous, 1; Hypotrichosis 8 and 12; Hypotrichosis-lymphedema-telangiectasia syndrome; I blood group system; Ichthyosis bullosa of Siemens; Ichthyosis exfoliativa; Ichthyosis prematurity syndrome; Idiopathic basal ganglia calcification 5; Idiopathic fibrosing alveolitis, chronic form; Dyskeratosis congenita, autosomal dominant, 2 and 5; Idiopathic hypercalcemia of infancy; Immune dysfunction with T-cell inactivation due to calcium entry defect 2; Immunodeficiency 15, 16, 19, 30, 31C, 38, 40, 8, due to defect in cd3-zeta, with hyper IgM type 1 and 2, and X-Linked, with magnesium defect, Epstein-Barr virus infection, and neoplasia; Immunodeficiency-centromeric instability-facial anomalies syndrome 2; Inclusion body myopathy 2 and 3; Nonaka myopathy; Infantile convulsions and paroxysmal choreoathetosis, familial; Infantile cortical hyperostosis; Infantile GM1 gangliosidosis; Infantile hypophosphatasia; Infantile nephronophthisis; Infantile nystagmus, X-linked; Infantile Parkinsonism-dystonia; Infertility associated with multi-tailed spermatozoa and excessive DNA; Insulin resistance; Insulin-resistant diabetes mellitus and acanthosis nigricans; Insulin-dependent diabetes mellitus secretory diarrhea syndrome; Interstitial nephritis, karyomegalic; Intrauterine growth retardation, metaphyseal dysplasia, adrenal hypoplasia congenita, and genital anomalies; lodotyrosyl coupling defect; IRAK4 deficiency; Iridogoniodysgenesis dominant type and type 1; Iron accumulation in brain; Ischiopatellar dysplasia; Islet cell hyperplasia; Isolated 17,20-lyase deficiency; Isolated lutropin deficiency; Isovaleryl-CoA dehydrogenase deficiency; Jankovic Rivera syndrome; Jervell and Lange-Nielsen syndrome 2; Joubert syndrome 1, 6, 7, 9/15 (digenic), 14, 16, and 17, and Orofaciodigital syndrome xiv; Junctional epidermolysis bullosa gravis of Herlitz; Juvenile GM>1< gangliosidosis; Juvenile polyposis syndrome; Juvenile polyposis/hereditary hemorrhagic telangiectasia syndrome; Juvenile retinoschisis; Kabuki make-up syndrome; Kallmann syndrome 1, 2, and 6; Delayed puberty; Kanzaki disease; Karak syndrome; Kartagener syndrome; Kenny-Caffey syndrome type 2; Keppen-Lubinsky syndrome; Keratoconus 1; Keratosis follicularis; Keratosis palmoplantaris striata 1; Kindler syndrome; L-2-hydroxyglutaric aciduria; Larsen syndrome, dominant type; Lattice corneal dystrophy Type III; Leber amaurosis; Zellweger syndrome; Peroxisome biogenesis disorders; Zellweger syndrome spectrum; Leber congenital amaurosis 11, 12, 13, 16, 4, 7, and 9; Leber optic atrophy; Aminoglycoside-induced deafness; Deafness, nonsyndromic sensorineural, mitochondrial; Left ventricular noncompaction 5; Left-right axis malformations; Leigh disease; Mitochondrial short-chain Enoyl-CoA Hydratase 1 deficiency; Leigh syndrome due to mitochondrial complex I deficiency; Leiner disease; Leri Weill dyschondrosteosis; Lethal congenital contracture syndrome 6; Leukocyte adhesion deficiency type I and III; Leukodystrophy, Hypomyelinating, 11 and 6; Leukoencephalopathy with ataxia, with Brainstem and Spinal Cord Involvement and Lactate Elevation, with vanishing white matter, and progressive, with ovarian failure; Leukonychia totalis; Lewy body dementia; Lichtenstein-Knorr Syndrome; Li-Fraumeni syndrome 1; Lig4 syndrome; Limb-girdle muscular dystrophy, type 1B, 2A, 2B, 2D, C1, C5, C9, C14; Congenital muscular dystrophy-dystroglycanopathy with brain and eye anomalies, type A14 and B14; Lipase deficiency combined; Lipid proteinosis; Lipodystrophy, familial partial, type 2 and 3; Lissencephaly 1, 2 (X-linked), 3, 6 (with microcephaly), X-linked; Subcortical laminar heterotopia, X-linked; Liver failure acute infantile; Loeys-Dietz syndrome 1, 2, 3; Long QT syndrome 1, 2, 2/9, 2/5, (digenic), 3, 5 and 5, acquired, susceptibility to; Lung cancer; Lymphedema, hereditary, id; Lymphedema, primary, with myelodysplasia; Lymphoproliferative syndrome 1, 1 (X-linked), and 2; Lysosomal acid lipase deficiency; Macrocephaly, macrosomia, facial dysmorphism syndrome; Macular dystrophy, vitelliform, adult-onset; Malignant hyperthermia susceptibility type 1; Malignant lymphoma, non-Hodgkin; Malignant melanoma; Malignant tumor of prostate; Mandibuloacral dysostosis; Mandibuloacral dysplasia with type A or B lipodystrophy, atypical; Mandibulofacial dysostosis, Treacher Collins type, autosomal recessive; Mannose-binding protein deficiency; Maple syrup urine disease type 1A and type 3; Marden Walker like syndrome; Marfan syndrome; Marinesco-Sj\xc3\xb6gren syndrome; Martsolf syndrome; Maturity-onset diabetes of the young, type 1, type 2, type 11, type 3, and type 9; May-Hegglin anomaly; MYH9 related disorders; Sebastian syndrome; McCune-Albright syndrome; Somatotroph adenoma; Sex cord-stromal tumor; Cushing syndrome; McKusick Kaufman syndrome; McLeod neuroacanthocytosis syndrome; Meckel-Gruber syndrome; Medium-chain acyl-coenzyme A dehydrogenase deficiency; Medulloblastoma; Megalencephalic leukoencephalopathy with subcortical cysts 1 and 2a; Megalencephaly cutis marmorata telangiectatica congenital; PIK3CA Related Overgrowth Spectrum; Megalencephaly-polymicrogyria-polydactyly-hydrocephalus syndrome 2; Megaloblastic anemia, thiamine-responsive, with diabetes mellitus and sensorineural deafness; Meier-Gorlin syndromes 1 and 4; Melnick-Needles syndrome; Meningioma; Mental retardation, X-linked, 3, 21, 30, and 72; Mental retardation and microcephaly with pontine and cerebellar hypoplasia; Mental retardation X-linked syndromic 5; Mental retardation, anterior maxillary protrusion, and strabismus; Mental retardation, autosomal dominant 12, 13, 15, 24, 3, 30, 4, 5, 6, and 9; Mental retardation, autosomal recessive 15, 44, 46, and 5; Mental retardation, stereotypic movements, epilepsy, and/or cerebral malformations; Mental retardation, syndromic, Claes-Jensen type, X-linked; Mental retardation, X-linked, nonspecific, syndromic, Hedera type, and syndromic, wu type; Merosin deficient congenital muscular dystrophy; Metachromatic leukodystrophy juvenile, late infantile, and adult types; Metachromatic leukodystrophy; Metatrophic dysplasia; Methemoglobinemia types I and 2; Methionine adenosyltransferase deficiency, autosomal dominant; Methylmalonic acidemia with homocystinuria; Methylmalonic aciduria cblB type; Methylmalonic aciduria due to methylmalonyl-CoA mutase deficiency; METHYLMALONIC ACIDURIA, mut(0) TYPE; Microcephalic osteodysplastic primordial dwarfism type 2; Microcephaly with or without chorioretinopathy, lymphedema, or mental retardation; Microcephaly, hiatal hernia and nephrotic syndrome; Microcephaly; Hypoplasia of the corpus callosum; Spastic paraplegia 50, autosomal recessive; Global developmental delay; CNS hypomyelination; Brain atrophy; Microcephaly, normal intelligence and immunodeficiency; Microcephaly-capillary malformation syndrome; Microcytic anemia; Microphthalmia syndromic 5, 7, and 9; Microphthalmia, isolated 3, 5, 6, 8, and with coloboma 6; Microspherophakia; Migraine, familial basilar; Miller syndrome; Minicore myopathy with external ophthalmoplegia; Myopathy, congenital with cores; Mitchell-Riley syndrome; mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase deficiency; Mitochondrial complex I, II, III, III (nuclear type 2, 4, or 8) deficiency; Mitochondrial DNA depletion syndrome 11, 12 (cardiomyopathic type), 2, 4B (MNGIE type), 8B (MNGIE type); Mitochondrial DNA-depletion syndrome 3 and 7, hepatocerebral types, and 13 (encephalomyopathic type); Mitochondrial phosphate carrier and pyruvate carrier deficiency; Mitochondrial trifunctional protein deficiency; Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency; Miyoshi muscular dystrophy 1; Myopathy, distal, with anterior tibial onset; Mohr-Tranebjaerg syndrome; Molybdenum cofactor deficiency, complementation group A; Mowat-Wilson syndrome; Mucolipidosis III Gamma; Mucopolysaccharidosis type VI, type VI (severe), and type VII; Mucopolysaccharidosis, MPS-I-H/S, MPS-II, MPS-III-A, MPS-III-B, MPS-III-C, MPS-IV-A, MPS-IV-B; Retinitis Pigmentosa 73; Gangliosidosis GM1 type1 (with cardiac involvement) 3; Multicentric osteolysis nephropathy; Multicentric osteolysis, nodulosis and arthropathy; Multiple congenital anomalies; Atrial septal defect 2; Multiple congenital anomalies-hypotonia-seizures syndrome 3; Multiple Cutaneous and Mucosal Venous Malformations; Multiple endocrine neoplasia, types 1 and 4; Multiple epiphyseal dysplasia 5 or Dominant; Multiple gastrointestinal atresias; Multiple pterygium syndrome Escobar type; Multiple sulfatase deficiency; Multiple synostoses syndrome 3; Muscle AMP guanine oxidase deficiency; Muscle eye brain disease; Muscular dystrophy, congenital, megaconial type; Myasthenia, familial infantile, 1; Myasthenic Syndrome, Congenital, 11, associated with acetylcholine receptor deficiency; Myasthenic Syndrome, Congenital, 17, 2A (slow-channel), 4B (fast-channel), and without tubular aggregates; Myeloperoxidase deficiency; MYH-associated polyposis; Endometrial carcinoma; Myocardial infarction 1; Myoclonic dystonia; Myoclonic-Atonic Epilepsy; Myoclonus with epilepsy with ragged red fibers; Myofibrillar myopathy 1 and ZASP-related; Myoglobinuria, acute recurrent, autosomal recessive; Myoneural gastrointestinal encephalopathy syndrome; Cerebellar ataxia infantile with progressive external ophthalmoplegia; Mitochondrial DNA depletion syndrome 4B, MNGIE type; Myopathy, centronuclear, 1, congenital, with excess of muscle spindles, distal, 1, lactic acidosis, and sideroblastic anemia 1, mitochondrial progressive with congenital cataract, hearing loss, and developmental delay, and tubular aggregate, 2; Myopia 6; Myosclerosis, autosomal recessive; Myotonia congenital; Congenital myotonia, autosomal dominant and recessive forms; Nail-patella syndrome; Nance-Horan syndrome; Nanophthalmos 2; Navajo neurohepatopathy; Nemaline myopathy 3 and 9; Neonatal hypotonia; Intellectual disability; Seizures; Delayed speech and language development; Mental retardation, autosomal dominant 31; Neonatal intrahepatic cholestasis caused by citrin deficiency; Nephrogenic diabetes insipidus, Nephrogenic diabetes insipidus, X-linked; Nephrolithiasis/osteoporosis, hypophosphatemic, 2; Nephronophthisis 13, 15 and 4; Infertility; Cerebello-oculo-renal syndrome (nephronophthisis, oculomotor apraxia and cerebellar abnormalities); Nephrotic syndrome, type 3, type 5, with or without ocular abnormalities, type 7, and type 9; Nestor-Guillermo progeria syndrome; Neu-Laxova syndrome 1; Neurodegeneration with brain iron accumulation 4 and 6; Neuroferritinopathy; Neurofibromatosis, type 1 and type 2; Neurofibrosarcoma; Neurohypophyseal diabetes insipidus; Neuropathy, Hereditary Sensory, Type IC; Neutral 1 amino acid transport defect; Neutral lipid storage disease with myopathy; Neutrophil immunodeficiency syndrome; Nicolaides-Baraitser syndrome; Niemann-Pick disease type C1, C2, type A, and type C1, adult form; Non-ketotic hyperglycinemia; Noonan syndrome 1 and 4, LEOPARD syndrome 1; Noonan syndrome-like disorder with or without juvenile myelomonocytic leukemia; Normokalemic periodic paralysis, potassium-sensitive; Norum disease; Epilepsy, Hearing Loss, And Mental Retardation Syndrome; Mental Retardation, X-Linked 102 and syndromic 13; Obesity; Ocular albinism, type I; Oculocutaneous albinism type 1B, type 3, and type 4; Oculodentodigital dysplasia; Odontohypophosphatasia; Odontotrichomelic syndrome; Oguchi disease; Oligodontia-colorectal cancer syndrome; Opitz G/BBB syndrome; Optic atrophy 9; Oral-facial-digital syndrome; Ornithine aminotransferase deficiency; Orofacial cleft 11 and 7, Cleft lip/palate-ectodermal dysplasia syndrome; Orstavik Lindemann Solberg syndrome; Osteoarthritis with mild chondrodysplasia; Osteochondritis dissecans; Osteogenesis imperfecta type 12, type 5, type 7, type 8, type I, type III, with normal sclerae, dominant form, recessive perinatal lethal; Osteopathia striata with cranial sclerosis; Osteopetrosis autosomal dominant type 1 and 2, recessive 4, recessive 1, recessive 6; Osteoporosis with pseudoglioma; Oto-palato-digital syndrome, types I and II; Ovarian dysgenesis 1; Ovarioleukodystrophy; Pachyonychia congenita 4 and type 2; Paget disease of bone, familial; Pallister-Hall syndrome; Palmoplantar keratoderma, nonepidermolytic, focal or diffuse; Pancreatic agenesis and congenital heart disease; Papillon-Lef\xc3\xa8vre syndrome; Paragangliomas 3; Paramyotonia congenita of von Eulenburg; Parathyroid carcinoma; Parkinson disease 14, 15, 19 (juvenile-onset), 2, 20 (early-onset), 6, (autosomal recessive early-onset, and 9; Partial albinism; Partial hypoxanthine-guanine phosphoribosyltransferase deficiency; Patterned dystrophy of retinal pigment epithelium; PC-K6a; Pelizaeus-Merzbacher disease; Pendred syndrome; Peripheral demyelinating neuropathy, central dysmyelination; Hirschsprung disease; Permanent neonatal diabetes mellitus; Diabetes mellitus, permanent neonatal, with neurologic features; Neonatal insulin-dependent diabetes mellitus; Maturity-onset diabetes of the young, type 2; Peroxisome biogenesis disorder 14B, 2A, 4A, 5B, 6A, 7A, and 7B; Perrault syndrome 4; Perry syndrome; Persistent hyperinsulinemic hypoglycemia of infancy; familial hyperinsulinism; Phenotypes; Phenylketonuria; Pheochromocytoma; Hereditary Paraganglioma-Pheochromocytoma Syndromes; Paragangliomas 1; Carcinoid tumor of intestine; Cowden syndrome 3; Phosphoglycerate dehydrogenase deficiency; Phosphoglycerate kinase 1 deficiency; Photosensitive trichothiodystrophy; Phytanic acid storage disease; Pick disease; Pierson syndrome; Pigmentary retinal dystrophy; Pigmented nodular adrenocortical disease, primary, 1; Pilomatrixoma; Pitt-Hopkins syndrome; Pituitary dependent hypercortisolism; Pituitary hormone deficiency, combined 1, 2, 3, and 4; Plasminogen activator inhibitor type 1 deficiency; Plasminogen deficiency, type I; Platelet-type bleeding disorder 15 and 8; Poikiloderma, hereditary fibrosing, with tendon contractures, myopathy, and pulmonary fibrosis; Polycystic kidney disease 2, adult type, and infantile type; Polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy; Polyglucosan body myopathy 1 with or without immunodeficiency; Polymicrogyria, asymmetric, bilateral frontoparietal; Polyneuropathy, hearing loss, ataxia, retinitis pigmentosa, and cataract; Pontocerebellar hypoplasia type 4; Popliteal pterygium syndrome; Porencephaly 2; Porokeratosis 8, disseminated superficial actinic type; Porphobilinogen synthase deficiency; Porphyria cutanea tarda; Posterior column ataxia with retinitis pigmentosa; Posterior polar cataract type 2; Prader-Willi-like syndrome; Premature ovarian failure 4, 5, 7, and 9; Primary autosomal recessive microcephaly 10, 2, 3, and 5; Primary ciliary dyskinesia 24; Primary dilated cardiomyopathy; Left ventricular noncompaction 6; 4, Left ventricular noncompaction 10; Paroxysmal atrial fibrillation; Primary hyperoxaluria, type I, type, and type III; Primary hypertrophic osteoarthropathy, autosomal recessive 2; Primary hypomagnesemia; Primary open angle glaucoma juvenile onset 1; Primary pulmonary hypertension; Primrose syndrome; Progressive familial heart block type 1B; Progressive familial intrahepatic cholestasis 2 and 3; Progressive intrahepatic cholestasis; Progressive myoclonus epilepsy with ataxia; Progressive pseudorheumatoid dysplasia; Progressive sclerosing poliodystrophy; Prolidase deficiency; Proline dehydrogenase deficiency; Schizophrenia 4; Properdin deficiency, X-linked; Propionic academia; Proprotein convertase 1/3 deficiency; Prostate cancer, hereditary, 2; Protan defect; Proteinuria; Finnish congenital nephrotic syndrome; Proteus syndrome; Breast adenocarcinoma; Pseudoachondroplastic spondyloepiphyseal dysplasia syndrome; Pseudohypoaldosteronism type 1 autosomal dominant and recessive and type 2; Pseudohypoparathyroidism type 1A, Pseudopseudohypoparathyroidism; Pseudoneonatal adrenoleukodystrophy; Pseudoprimary hyperaldosteronism; Pseudoxanthoma elasticum; Generalized arterial calcification of infancy 2; Pseudoxanthoma elasticum-like disorder with multiple coagulation factor deficiency; Psoriasis susceptibility 2; PTEN hamartoma tumor syndrome; Pulmonary arterial hypertension related to hereditary hemorrhagic telangiectasia; Pulmonary Fibrosis And/Or Bone Marrow Failure, Telomere-Related, 1 and 3; Pulmonary hypertension, primary, 1, with hereditary hemorrhagic telangiectasia; Purine-nucleoside phosphorylase deficiency; Pyruvate carboxylase deficiency; Pyruvate dehydrogenase E1-alpha deficiency; Pyruvate kinase deficiency of red cells; Raine syndrome; Rasopathy; Recessive dystrophic epidermolysis bullosa; Nail disorder, nonsyndromic congenital, 8; Reifenstein syndrome; Renal adysplasia; Renal carnitine transport defect; Renal coloboma syndrome; Renal dysplasia; Renal dysplasia, retinal pigmentary dystrophy, cerebellar ataxia and skeletal dysplasia; Renal tubular acidosis, distal, autosomal recessive, with late-onset sensorineural hearing loss, or with hemolytic anemia; Renal tubular acidosis, proximal, with ocular abnormalities and mental retardation; Retinal cone dystrophy 3B; Retinitis pigmentosa; Retinitis pigmentosa 10, 11, 12, 14, 15, 17, and 19; Retinitis pigmentosa 2, 20, 25, 35, 36, 38, 39, 4, 40, 43, 45, 48, 66, 7, 70, 72; Retinoblastoma; Rett disorder; Rhabdoid tumor predisposition syndrome 2; Rhegmatogenous retinal detachment, autosomal dominant; Rhizomelic chondrodysplasia punctata type 2 and type 3; Roberts-SC phocomelia syndrome; Robinow Sorauf syndrome; Robinow syndrome, autosomal recessive, autosomal recessive, with brachy-syn-polydactyly; Rothmund-Thomson syndrome; Rapadilino syndrome; RRM2B-related mitochondrial disease; Rubinstein-Taybi syndrome; Salla disease; Sandhoff disease, adult and infantil types; Sarcoidosis, early-onset; Blau syndrome; Schindler disease, type 1; Schizencephaly; Schizophrenia 15; Schneckenbecken dysplasia; Schwannomatosis 2; Schwartz Jampel syndrome type 1; Sclerocornea, autosomal recessive; Sclerosteosis; Secondary hypothyroidism; Segawa syndrome, autosomal recessive; Senior-Loken syndrome 4 and 5; Sensory ataxic neuropathy, dysarthria, and ophthalmoparesis; Sepiapterin reductase deficiency; SeSAME syndrome; Severe combined immunodeficiency due to ADA deficiency, with microcephaly, growth retardation, and sensitivity to ionizing radiation, atypical, autosomal recessive, T cell-negative, B cell-positive, NK cell-negative of NK-positive; Severe congenital neutropenia; Severe congenital neutropenia 3, autosomal recessive or dominant; Severe congenital neutropenia and 6, autosomal recessive; Severe myoclonic epilepsy in infancy; Generalized epilepsy with febrile seizures plus, types 1 and 2; Severe X-linked myotubular myopathy; Short QT syndrome 3; Short stature with nonspecific skeletal abnormalities; Short stature, auditory canal atresia, mandibular hypoplasia, skeletal abnormalities; Short stature, onychodysplasia, facial dysmorphism, and hypotrichosis; Primordial dwarfism; Short-rib thoracic dysplasia 11 or 3 with or without polydactyly; Sialidosis type I and II; Silver spastic paraplegia syndrome; Slowed nerve conduction velocity, autosomal dominant; Smith-Lemli-Opitz syndrome; Snyder Robinson syndrome; Somatotroph adenoma; Prolactinoma; familial, Pituitary adenoma predisposition; Sotos syndrome 1 or 2; Spastic ataxia 5, autosomal recessive, Charlevoix-Saguenay type, 1, 10, or 11, autosomal recessive; Amyotrophic lateral sclerosis type 5; Spastic paraplegia 15, 2, 3, 35, 39, 4, autosomal dominant, 55, autosomal recessive, and 5A; Bile acid synthesis defect, congenital, 3; Spermatogenic failure 11, 3, and 8; Spherocytosis types 4 and 5; Spheroid body myopathy; Spinal muscular atrophy, lower extremity predominant 2, autosomal dominant; Spinal muscular atrophy, type II; Spinocerebellar ataxia 14, 21, 35, 40,and 6; Spinocerebellar ataxia autosomal recessive 1 and 16; Splenic hypoplasia; Spondylocarpotarsal synostosis syndrome; Spondylocheirodysplasia, Ehlers-Danlos syndrome-like, with immune dysregulation, Aggrecan type, with congenital joint dislocations, short limb-hand type, Sedaghatian type, with cone-rod dystrophy, and Kozlowski type; Parastremmatic dwarfism; Stargardt disease 1; Cone-rod dystrophy 3; Stickler syndrome type 1; Kniest dysplasia; Stickler syndrome, types 1(nonsyndromic ocular) and 4; Sting-associated vasculopathy, infantile-onset; Stormorken syndrome; Sturge-Weber syndrome, Capillary malformations, congenital, 1; Succinyl-CoA acetoacetate transferase deficiency; Sucrase-isomaltase deficiency; Sudden infant death syndrome; Sulfite oxidase deficiency, isolated; Supravalvar aortic stenosis; Surfactant metabolism dysfunction, pulmonary, 2 and 3; Symphalangism, proximal, 1b; Syndactyly Cenani Lenz type; Syndactyly type 3; Syndromic X-linked mental retardation 16; Talipes equinovarus; Tangier disease; TARP syndrome; Tay-Sachs disease, B1 variant, Gm2-gangliosidosis (adult), Gm2-gangliosidosis (adult-onset); Temtamy syndrome; Tenorio Syndrome; Terminal osseous dysplasia; Testosterone 17-beta-dehydrogenase deficiency; Tetraamelia, autosomal recessive; Tetralogy of Fallot; Hypoplastic left heart syndrome 2; Truncus arteriosus; Malformation of the heart and great vessels; Ventricular septal defect 1; Thiel-Behnke corneal dystrophy; Thoracic aortic aneurysms and aortic dissections; Marfanoid habitus; Three M syndrome 2; Thrombocytopenia, platelet dysfunction, hemolysis, and imbalanced globin synthesis; Thrombocytopenia, X-linked; Thrombophilia, hereditary, due to protein C deficiency, autosomal dominant and recessive; Thyroid agenesis; Thyroid cancer, follicular; Thyroid hormone metabolism, abnormal; Thyroid hormone resistance, generalized, autosomal dominant; Thyrotoxic periodic paralysis and Thyrotoxic periodic paralysis 2; Thyrotropin-releasing hormone resistance, generalized; Timothy syndrome; TNF receptor-associated periodic fever syndrome (TRAPS); Tooth agenesis, selective, 3 and 4; Torsades de pointes; Townes-Brocks-branchiootorenal-like syndrome; Transient bullous dermolysis of the newborn; Treacher collins syndrome 1; Trichomegaly with mental retardation, dwarfism and pigmentary degeneration of retina; Trichorhinophalangeal dysplasia type I; Trichorhinophalangeal syndrome type 3; Trimethylaminuria; Tuberous sclerosis syndrome; Lymphangiomyomatosis; Tuberous sclerosis 1 and 2; Tyrosinase-negative oculocutaneous albinism; Tyrosinase-positive oculocutaneous albinism; Tyrosinemia type I; UDPglucose-4-epimerase deficiency; Ullrich congenital muscular dystrophy; Ulna and fibula absence of with severe limb deficiency; Upshaw-Schulman syndrome; Urocanate hydratase deficiency; Usher syndrome, types 1, 1B, 1D, 1G, 2A, 2C, and 2D; Retinitis pigmentosa 39; UV-sensitive syndrome; Van der Woude syndrome; Van Maldergem syndrome 2; Hennekam lymphangiectasia-lymphedema syndrome 2; Variegate porphyria; Ventriculomegaly with cystic kidney disease; Verheij syndrome; Very long chain acyl-CoA dehydrogenase deficiency; Vesicoureteral reflux 8; Visceral heterotaxy 5, autosomal; Visceral myopathy; Vitamin D-dependent rickets, types 1 and 2; Vitelliform dystrophy; von Willebrand disease type 2M and type 3; Waardenburg syndrome type 1, 4C, and 2E (with neurologic involvement); Klein-Waardenberg syndrome; Walker-Warburg congenital muscular dystrophy; Warburg micro syndrome 2 and 4; Warts, hypogammaglobulinemia, infections, and myelokathexis; Weaver syndrome; Weill-Marchesani syndrome 1 and 3; Weill-Marchesani-like syndrome; Weissenbacher-Zweymuller syndrome; Werdnig-Hoffmann disease; Charcot-Marie-Tooth disease; Werner syndrome; WFS1-Related Disorders; Wiedemann-Steiner syndrome; Wilson disease; Wolfram-like syndrome, autosomal dominant; Worth disease; Van Buchem disease type 2; Xeroderma pigmentosum, complementation group b, group D, group E, and group G; X-linked agammaglobulinemia; X-linked hereditary motor and sensory neuropathy; X-linked ichthyosis with steryl-sulfatase deficiency; X-linked periventricular heterotopia; Oto-palato-digital syndrome, type I; X-linked severe combined immunodeficiency; Zimmermann-Laband syndrome and Zimmermann-Laband syndrome 2; and Zonular pulverulent cataract 3.


The target nucleotide sequence may comprise a target sequence (e.g., a point mutation) associated with a disease, disorder, or condition. The target sequence may comprise a T to C (or A to G) point mutation associated with a disease, disorder, or condition, and wherein the deamination of the mutant C base results in mismatch repair-mediated correction to a sequence that is not associated with a disease, disorder, or condition. The target sequence may comprise a G to A (or C to T) point mutation associated with a disease, disorder, or condition, and wherein the deamination of the mutant A base results in mismatch repair-mediated correction to a sequence that is not associated with a disease, disorder, or condition. The target sequence may encode a protein, and where the point mutation is in a codon and results in a change in the amino acid encoded by the mutant codon as compared to a wild-type codon. The target sequence may also be at a splice site, and the point mutation results in a change in the splicing of an mRNA transcript as compared to a wild-type transcript. In addition, the target may be at a non-coding sequence of a gene, such as a promoter, and the point mutation results in increased or decreased expression of the gene.


Thus, in some aspects, the deamination of a mutant C results in a change of the amino acid encoded by the mutant codon, which in some cases can result in the expression of a wild-type amino acid. In other aspects, the deamination of a mutant A results in a change of the amino acid encoded by the mutant codon, which in some cases can result in the expression of a wild-type amino acid.


The methods described herein involving contacting a cell with a composition or rAAV particle can occur in vitro, ex vivo, or in vivo. In certain embodiments, the step of contacting occurs in a subject. In certain embodiments, the subject has been diagnosed with a disease, disorder, or condition.


In some embodiments, the methods disclosed herein involve contacting a mammalian cell with a composition or rAAV particle. In particular embodiments, the methods involve contacting a retinal cell, cortical cell or cerebellar cell.


The split Cas9 protein or split prime editor delivered using the methods described herein preferably have comparable activity compared to the original Cas9 protein or prime editor (i.e., unsplit protein delivered to a cell or expressed in a cell as a whole). For example, the split Cas9 protein or split prime editor retains at least 50% (e.g., at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, at least 99%, or 100%) of the activity of the original Cas9 protein or prime editor. In some embodiments, the split Cas9 protein or split prime editor is more active (e.g., 2-fold, 5-fold, 10-fold, 100-fold, 1000-fold, or more) than that of an original Cas9 protein or prime editor.


The compositions described herein may be administered to a subject in need thereof in a therapeutically effective amount to treat and/or prevent a disease or disorder the subject is suffering from. Any disease or disorder that maybe treated and/or prevented using CRISPR/Cas9-based genome-editing technology may be treated by the split Cas9 protein or the split multi-flap prime editor described herein. It is to be understood that, if the nucleotide sequences encoding the split Cas9 protein or the multi-flap prime editor does not further encode a gRNA, a separate nucleic acid vector encoding the gRNA may be administered together with the compositions described herein.


Exemplary suitable diseases, disorders or conditions include, without limitation the disease or disorder is selected from the group consisting of: cystic fibrosis, phenylketonuria, epidermolytic hyperkeratosis (EHK), chronic obstructive pulmonary disease (COPD), Charcot-Marie-Toot disease type 4J, neuroblastoma (NB), von Willebrand disease (vWD), myotonia congenital, hereditary renal amyloidosis, dilated cardiomyopathy, hereditary lymphedema, familial Alzheimer's disease, prion disease, chronic infantile neurologic cutaneous articular syndrome (CINCA), congenital deafness, Niemann-Pick disease type C (NPC) disease, and desmin-related myopathy (DRM). In particular embodiments, the disease or condition is Niemann-Pick disease type C (NPC) disease.


In some embodiments, the disease, disorder or condition is associated with a point mutation in an NPC gene, a DNMT1 gene, a PCSK9 gene, or a TMC1 gene. In certain embodiments, the point mutation is a T3182C mutation in NPC, which results in an I1061T amino acid substitution.


In certain embodiments, the point mutation is an A545G mutation in TMC1, which results in a Y182C amino acid substitution. TMC1 encodes a protein that forms mechanosensitive ion channels in sensory hair cells of the inner ear and is required for normal auditory function. The Y182C amino acid substitution is associated with congenital deafness.


In some embodiments, the disease, disorder or condition is associated with a point mutation that generates a stop codon, for example, a premature stop codon within the coding region of a gene.


Additional exemplary diseases, disorders and conditions include cystic fibrosis (see, e.g., Schwank et al., Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell stem cell. 2013; 13: 653-658; and Wu et. al., Correction of a genetic disease in mouse via use of CRISPR-Cas9. Cell stem cell. 2013; 13: 659-662, neither of which uses a deaminase fusion protein to correct the genetic defect); phenylketonuria—e.g., phenylalanine to serine mutation at position 835 (mouse) or 240 (human) or a homologous residue in phenylalanine hydroxylase gene (T>C mutation)—see, e.g., McDonald et al., Genomics. 1997; 39:402-405; Bernard-Soulier syndrome (BSS)—e.g., phenylalanine to serine mutation at position 55 or a homologous residue, or cysteine to arginine at residue 24 or a homologous residue in the platelet membrane glycoprotein IX (T>C mutation)—see, e.g., Noris et al., British Journal of Haematology. 1997; 97: 312-320, and Ali et al., Hematol. 2014; 93: 381-384; epidermolytic hyperkeratosis (EHK)—e.g., leucine to proline mutation at position 160 or 161 (if counting the initiator methionine) or a homologous residue in keratin 1 (T>C mutation)—see, e.g., Chipev et al., Cell. 1992; 70: 821-828, see also accession number P04264 in the UNIPROT database at www[dot]uniprot[dot]org; chronic obstructive pulmonary disease (COPD)—e.g., leucine to proline mutation at position 54 or 55 (if counting the initiator methionine) or a homologous residue in the processed form of α1-antitrypsin or residue 78 in the unprocessed form or a homologous residue (T>C mutation)—see, e.g., Poller et al., Genomics. 1993; 17: 740-743, see also accession number P01011 in the UNIPROT database; Charcot-Marie-Toot disease type 4J—e.g., isoleucine to threonine mutation at position 41 or a homologous residue in FIG4 (T>C mutation)—see, e.g., Lenk et al., PLoS Genetics. 2011; 7: e1002104; neuroblastoma (NB)—e.g., leucine to proline mutation at position 197 or a homologous residue in Caspase-9 (T>C mutation)—see, e.g., Kundu et al., 3 Biotech. 2013, 3:225-234; von Willebrand disease (vWD)—e.g., cysteine to arginine mutation at position 509 or a homologous residue in the processed form of von Willebrand factor, or at position 1272 or a homologous residue in the unprocessed form of von Willebrand factor (T>C mutation)—see, e.g., Lavergne et al., Br. J. Haematol. 1992, see also accession number P04275 in the UNIPROT database; 82: 66-72; myotonia congenital—e.g., cysteine to arginine mutation at position 277 or a homologous residue in the muscle chloride channel gene CLCN1 (T>C mutation)—see, e.g., Weinberger et al., The J. of Physiology. 2012; 590: 3449-3464; hereditary renal amyloidosis—e.g., stop codon to arginine mutation at position 78 or a homologous residue in the processed form of apolipoprotein AII or at position 101 or a homologous residue in the unprocessed form (T>C mutation)—see, e.g., Yazaki et al., Kidney Int. 2003; 64: 11-16; dilated cardiomyopathy (DCM)—e.g., tryptophan to Arginine mutation at position 148 or a homologous residue in the FOXD4 gene (T>C mutation), see, e.g., Minoretti et. al., Int. J. of Mol. Med. 2007; 19: 369-372; hereditary lymphedema—e.g., histidine to arginine mutation at position 1035 or a homologous residue in VEGFR3 tyrosine kinase (A>G mutation), see, e.g., Irrthum et al., Am. J. Hum. Genet. 2000; 67: 295-301; familial Alzheimer's disease—e.g., isoleucine to valine mutation at position 143 or a homologous residue in presenilin1 (A>G mutation), see, e.g., Gallo et. al., J. Alzheimer's disease. 2011; 25: 425-431; Prion disease—e.g., methionine to valine mutation at position 129 or a homologous residue in prion protein (A>G mutation)—see, e.g., Lewis et. al., J. of General Virology. 2006; 87: 2443-2449; chronic infantile neurologic cutaneous articular syndrome (CINCA)—e.g., Tyrosine to Cysteine mutation at position 570 or a homologous residue in cryopyrin (A>G mutation)—see, e.g., Fujisawa et. al. Blood. 2007; 109: 2903-2911; and desmin-related myopathy (DRM)—e.g., arginine to glycine mutation at position 120 or a homologous residue in αβ crystallin (A>G mutation)—see, e.g., Kumar et al., J. Biol. Chem. 1999; 274: 24137-24141. The entire contents of all references and database entries is incorporated herein by reference.


Trinucleotide Repeat Expansion Disease


Trinucleotide repeat expansion is associated with a number of human diseases, including Huntington's Disease, Fragile X syndrome, and Friedreich's ataxia. The most common trinucleotide repeat contains CAG triplets, though GAA triplets (Friedreich's ataxia) and CGG triplets (Fragile X syndrome) also occur. Inheriting a predisposition to expansion, or acquiring an already expanded parental allele, increases the likelihood of acquiring the disease. Pathogenic expansions of trinucleotide repeats could hypothetically be corrected using multi-flap prime editing.


A region upstream of the repeat region can be nicked by an RNA-guided nuclease, then used to prime synthesis of a new DNA strand that contains a healthy number of repeats (which depends on the particular gene and disease), in accordance with the general mechanism outlined in FIG. 1G or FIG. 22. After the repeat sequence, a short stretch of homology is added that matches the identity of the sequence adjacent to the other end of the repeat (red strand). Invasion of the newly synthesized strand by the TPRT system, and subsequent replacement of the endogenous DNA with the newly synthesized flap, leads to a contracted repeat allele. The term “contracted” refers to a shortening of the length of the nucleotide repeat region, thereby resulting in repairing the trinucleotide repeat region.


The multi-flap prime editing systems (e.g., dual-flap and quadruple-flap prime editing systems) described herein may be used to contract trinucleotide repeat mutations (or “triplet expansion diseases”) to treating conditions such as Huntington's disease and other trinucleotide repeat disorders. Trinucleotide repeat expansion disorders are complex, progressive disorders that involve developmental neurobiology and often affect cognition as well as sensori-motor functions. The disorders show genetic anticipation (i.e. increased severity with each generation). The DNA expansions or contractions usually happen meiotically (i.e. during the time of gametogenesis, or early in embryonic development), and often have sex-bias meaning that some genes expand only when inherited through the female, others only through the male. In humans, trinucleotide repeat expansion disorders can cause gene silencing at either the transcriptional or translational level, which essentially knocks out gene function. Alternatively, trinucleotide repeat expansion disorders can cause altered proteins generated with large repetitive amino acid sequences that either abrogate or change protein function, often in a dominant-negative manner (e.g. poly-glutamine diseases).


Without wishing to be bound by theory, triplet expansion is caused by slippage during DNA replication or during DNA repair synthesis. Because the tandem repeats have identical sequence to one another, base pairing between two DNA strands can take place at multiple points along the sequence. This may lead to the formation of “loop out” structures during DNA replication or DNA repair synthesis. This may lead to repeated copying of the repeated sequence, expanding the number of repeats. Additional mechanisms involving hybrid RNA:DNA intermediates have been proposed. Multi-flap prime editing may be used to reduce or eliminate these triplet expansion regions by deletion one or more or the offending repeat codon triplets. In an embodiment of this use, FIG. 23, provides a schematic of a PEgRNA design for contracting or reducing trinucleotide repeat sequences with prime editing.


Multi-flap prime editing may be implemented to contract triplet expansion regions by nicking a region upstream of the triplet repeat region with the prime editor comprising a PEgRNA appropriated targeted to the cut site. The prime editor then synthesizes a new DNA strand (ssDNA flap) based on the PEgRNA as a template (i.e., the edit template thereof) that codes for a healthy number of triplet repeats (which depends on the particular gene and disease). The newly synthesized ssDNA strand comprising the healthy triplet repeat sequence also is synthesized to include a short stretch of homology (i.e., the homology arm) that matches the sequence adjacent to the other end of the repeat (red strand). Invasion of the newly synthesized strand, and subsequent replacement of the endogenous DNA with the newly synthesized ssDNA flap, leads to a contracted repeat allele.


Depending on the particular trinucleotide expansion disorder, the defect-inducing triplet expansions may occur in “trinucleotide repeat expansion proteins.” Trinucleotide repeat expansion proteins are a diverse set of proteins associated with susceptibility for developing a trinucleotide repeat expansion disorder, the presence of a trinucleotide repeat expansion disorder, the severity of a trinucleotide repeat expansion disorder or any combination thereof. Trinucleotide repeat expansion disorders are divided into two categories determined by the type of repeat. The most common repeat is the triplet CAG, which, when present in the coding region of a gene, codes for the amino acid glutamine (Q). Therefore, these disorders are referred to as the polyglutamine (polyQ) disorders and comprise the following diseases: Huntington Disease (HD); Spinobulbar Muscular Atrophy (SBMA); Spinocerebellar Ataxias (SCA types 1, 2, 3, 6, 7, and 17); and Dentatorubro-Pallidoluysian Atrophy (DRPLA). The remaining trinucleotide repeat expansion disorders either do not involve the CAG triplet or the CAG triplet is not in the coding region of the gene and are, therefore, referred to as the non-polyglutamine disorders. The non-polyglutamine disorders comprise Fragile X Syndrome (FRAXA); Fragile XE Mental Retardation (FRAXE); Friedreich Ataxia (FRDA); Myotonic Dystrophy (DM); and Spinocerebellar Ataxias (SCA types 8, and 12).


The proteins associated with trinucleotide repeat expansion disorders can be selected based on an experimental association of the protein associated with a trinucleotide repeat expansion disorder to a trinucleotide repeat expansion disorder. For example, the production rate or circulating concentration of a protein associated with a trinucleotide repeat expansion disorder may be elevated or depressed in a population having a trinucleotide repeat expansion disorder relative to a population lacking the trinucleotide repeat expansion disorder. Differences in protein levels may be assessed using proteomic techniques including but not limited to Western blot, immunohistochemical staining, enzyme linked immunosorbent assay (ELISA), and mass spectrometry. Alternatively, the proteins associated with trinucleotide repeat expansion disorders may be identified by obtaining gene expression profiles of the genes encoding the proteins using genomic techniques including but not limited to DNA microarray analysis, serial analysis of gene expression (SAGE), and quantitative real-time polymerase chain reaction (Q-PCR).


Non-limiting examples of proteins associated with trinucleotide repeat expansion disorders which can be corrected by multi-flap prime editing include AR (androgen receptor), FMR1 (fragile X mental retardation 1), HTT (huntingtin), DMPK (dystrophia myotonica-protein kinase), FXN (frataxin), ATXN2 (ataxin 2), ATN1 (atrophin 1), FEN1 (flap structure-specific endonuclease 1), TNRC6A (trinucleotide repeat containing 6A), PABPN1 (poly(A) binding protein, nuclear 1), JPH3 (junctophilin 3), MED15 (mediator complex subunit 15), ATXN1 (ataxin 1), ATXN3 (ataxin 3), TBP (TATA box binding protein), CACNA1A (calcium channel, voltage-dependent, P/Q type, alpha 1A subunit), ATXN8OS (ATXN8 opposite strand (non-protein coding)), PPP2R2B (protein phosphatase 2, regulatory subunit B, beta), ATXN7 (ataxin 7), TNRC6B (trinucleotide repeat containing 6B), TNRC6C (trinucleotide repeat containing 6C), CELF3 (CUGBP, Elav-like family member 3), MAB21L1 (mab-21-like 1 (C. elegans)), MSH2 (mutS homolog 2, colon cancer, nonpolyposis type 1 (E. coli)), TMEM185A (transmembrane protein 185A), SIX5 (SIX homeobox 5), CNPY3 (canopy 3 homolog (zebrafish)), FRAXE (fragile site, folic acid type, rare, fra(X)(q28) E), GNB2 (guanine nucleotide binding protein (G protein), beta polypeptide 2), RPL14 (ribosomal protein L14), ATXN8 (ataxin 8), INSR (insulin receptor), TTR (transthyretin), EP400 (E1A binding protein p400), GIGYF2 (GRB10 interacting GYF protein 2), OGG1 (8-oxoguanine DNA glycosylase), STC1 (stanniocalcin 1), CNDP1 (carnosine dipeptidase 1 (metallopeptidase M20 family)), C10orf2 (chromosome 10 open reading frame 2), MAML3 mastermind-like 3 (Drosophila), DKC1 (dyskeratosis congenita 1, dyskerin), PAXIP1 (PAX interacting (with transcription-activation domain) protein 1), CASK (calcium/calmodulin-dependent serine protein kinase (MAGUK family)), MAPT (microtubule-associated protein tau), SP1 (Sp1 transcription factor), POLG (polymerase (DNA directed), gamma), AFF2 (AF4/FMR2 family, member 2), THB S1 (thrombospondin 1), TP53 (tumor protein p53), ESR1 (estrogen receptor 1), CGGBP1 (CGG triplet repeat binding protein 1), ABT1 (activator of basal transcription 1), KLK3 (kallikrein-related peptidase 3), PRNP (prion protein), JUN (jun oncogene), KCNN3 (potassium intermediate/small conductance calcium-activated channel, subfamily N, member 3), BAX (BCL2-associated X protein), FRAXA (fragile site, folic acid type, rare, fra(X)(q27.3) A (macroorchidism, mental retardation)), KBTBD10 (kelch repeat and BTB (POZ) domain containing 10), MBNL1 (muscleblind-like (Drosophila)), RAD51 (RAD51 homolog (RecA homolog, E. coli) (S. cerevisiae)), NCOA3 (nuclear receptor coactivator 3), ERDA1 (expanded repeat domain, CAG/CTG 1), TSC1 (tuberous sclerosis 1), COMP (cartilage oligomeric matrix protein), GCLC (glutamate-cysteine ligase, catalytic subunit), RRAD (Ras-related associated with diabetes), MSH3 (mutS homolog 3 (E. coli)), DRD2 (dopamine receptor D2), CD44 (CD44 molecule (Indian blood group)), CTCF (CCCTC-binding factor (zinc finger protein)), CCND1 (cyclin D1), CLSPN (claspin homolog (Xenopus laevis)), MEF2A (myocyte enhancer factor 2A), PTPRU (protein tyrosine phosphatase, receptor type, U), GAPDH (glyceraldehyde-3-phosphate dehydrogenase), TRIM22 (tripartite motif-containing 22), WT1 (Wilms tumor 1), AHR (aryl hydrocarbon receptor), GPX1 (glutathione peroxidase 1), TPMT (thiopurine S-methyltransferase), NDP (Norrie disease (pseudoglioma)), ARX (aristaless related homeobox), MUS81 (MUS81 endonuclease homolog (S. cerevisiae)), TYR (tyrosinase (oculocutaneous albinism IA)), EGR1 (early growth response 1), UNG (uracil-DNA glycosylase), NUMBL (numb homolog (Drosophila)-like), FABP2 (fatty acid binding protein 2, intestinal), EN2 (engrailed homeobox 2), CRYGC (crystallin, gamma C), SRP14 (signal recognition particle 14 kDa (homologous Alu RNA binding protein)), CRYGB (crystallin, gamma B), PDCD1 (programmed cell death 1), HOXA1 (homeobox A1), ATXN2L (ataxin 2-like), PMS2 (PMS2 postmeiotic segregation increased 2 (S. cerevisiae)), GLA (galactosidase, alpha), CBL (Cas-Br-M (murine) ecotropic retroviral transforming sequence), FTH1 (ferritin, heavy polypeptide 1), IL12RB2 (interleukin 12 receptor, beta 2), OTX2 (orthodenticle homeobox 2), HOXA5 (homeobox A5), POLG2 (polymerase (DNA directed), gamma 2, accessory subunit), DLX2 (distal-less homeobox 2), SIRPA (signal-regulatory protein alpha), OTX1 (orthodenticle homeobox 1), AHRR (aryl-hydrocarbon receptor repressor), MANF (mesencephalic astrocyte-derived neurotrophic factor), TMEM158 (transmembrane protein 158 (gene/pseudogene)), and ENSG00000078687.


In a particular aspect, the instant disclosure provides TPRT-based methods for the treatment of a subject diagnosed with an expansion repeat disorder (also known as a repeat expansion disorder or a trinucleotide repeat disorder). Expansion repeat disorders occur when microsatellite repeats expand beyond a threshold length. Currently, at least 30 genetic diseases are believed to be caused by repeat expansions. Scientific understanding of this diverse group of disorders came to lights in the early 1990's with the discovery that trinucleotide repeats underlie several major inherited conditions, including Fragile X, Spinal and Bulbar Muscular Atrophy, Myotonic Dystrophy, and Huntington's disease (Nelson et al, “The unstable repeats—three evolving faces of neurological disease,” Neuron, Mar. 6, 2013, Vol. 77; 825-843, which is incorporated herein by reference), as well as Haw River Syndrome, Jacobsen Syndrome, Dentatorubral-pallidoluysian atrophy (DRPLA), Machado-Joseph disease, Synpolydactyly (SPD II), Hand-foot genital syndrome (HFGS), Cleidocranial dysplasia (CCD), Holoprosencephaly disorder (HPE), Congenital central hypventilation syndrome (CCHS), ARX-nonsyndromic X-linked mental retardation (XLMR), and Oculopharyngeal muscular dystrophy (OPMD) (see. Microsatellite repeat instability was found to be a hallmark of these conditions, as was anticipation—the phenomenon in which repeat expansion can occur with each successive generation, which leads to a more severe phenotype and earlier age of onset in the offspring. Repeat expansions are believed to cause diseases via several different mechanisms. Namely, expansions may interfere with cellular functioning at the level of the gene, the mRNA transcript, and/or the encoded protein. In some conditions, mutations act via a loss-of-function mechanism by silencing repeat-containing genes. In others, disease results from gain-of-function mechanisms, whereby either the mRNA transcript or protein takes on new, aberrant functions.


In one embodiment, a method of treating a trinucleotide repeat disorder is depicted in FIG. 23. In general, the approach involves using TPRT genome editing (i.e., multi-flap prime editing) in combination with an extended gRNA that comprises a region that encodes a desired and healthy replacement trinucleotide repeat sequence that is intended to replace the endogenous diseased trinucleotide repeat sequence through the mechanism of the prime editing process. A schematic of an exemplary gRNA design for contracting trinucleotide repeat sequences and trinucleotide repeat contraction with TPRT genome editing (i.e., multi-flap prime editing) is shown in FIG. 23.


Prion Disease


Multi-flap prime editing can also be used to prevent or halt the progression of prion disease through the installation of one or more protective mutations into prion proteins (PRNP) which become misfolded during the course of disease. Prion diseases or transmissible spongiform encephalopathies (TSEs) are a family of rare progressive neurodegenerative disorders that affect both humans and animals They are distinguished by long incubation periods, characteristic spongiform changes associated with neuronal loss, and a failure to induce inflammatory response.


In humans, prion disease includes Creutzfeldt-Jakob Disease (CJD), Variant Creutzfeldt-Jakob Disease (vCJD), Gerstmann-Straussler-Scheinker Syndrome, Fatal Familial Insomnia, and Kuru. In animals, prion disease includes Bovine Spongiform Encephalopathy (BSE or “mad cow disease”), Chronic Wasting Disease (CWD), Scrapie, Transmissible Mink Encephalopathy, Feline Spongiform Encephalopathy, and Ungulate Spongiform Encephalopathy. Multi-flap prime editing may be used to install protective point mutations into a prion protein in order to prevent or halt the progression of any one of these prion diseases.


Classic CJD is a human prion disease. It is a neurodegenerative disorder with characteristic clinical and diagnostic features. This disease is rapidly progressive and always fatal. Infection with this disease leads to death usually within 1 year of onset of illness. CJD is a rapidly progressive, invariably fatal neurodegenerative disorder believed to be caused by an abnormal isoform of a cellular glycoprotein known as the prion protein. CJD occurs worldwide and the estimated annual incidence in many countries, including the United States, has been reported to be about one case per million population. The vast majority of CJD patients usually die within 1 year of illness onset. CJD is classified as a transmissible spongiform encephalopathy (TSE) along with other prion diseases that occur in humans and animals. In about 85% of patients, CJD occurs as a sporadic disease with no recognizable pattern of transmission. A smaller proportion of patients (5 to 15%) develop CJD because of inherited mutations of the prion protein gene. These inherited forms include Gerstmann-Straussler-Scheinker syndrome and fatal familial insomnia. No treatment is currently known for CJD.


Variant Creutzfeldt-Jakob disease (vCJD) is a prion disease that was first described in 1996 in the United Kingdom. There is now strong scientific evidence that the agent responsible for the outbreak of prion disease in cows, bovine spongiform encephalopathy (BSE or ‘mad cow’ disease), is the same agent responsible for the outbreak of vCJD in humans. Variant CJD (vCJD) is not the same disease as classic CJD (often simply called CJD). It has different clinical and pathologic characteristics from classic CJD. Each disease also has a particular genetic profile of the prion protein gene. Both disorders are invariably fatal brain diseases with unusually long incubation periods measured in years, and are caused by an unconventional transmissible agent called a prion. No treatment is currently known for vCJD.


BSE (bovine spongiform encephalopathy or “mad cow disease”) is a progressive neurological disorder of cattle that results from infection by an unusual transmissible agent called a prion. The nature of the transmissible agent is not well understood. Currently, the most accepted theory is that the agent is a modified form of a normal protein known as prion protein. For reasons that are not yet understood, the normal prion protein changes into a pathogenic (harmful) form that then damages the central nervous system of cattle. There is increasing evidence that there are different strains of BSE: the typical or classic BSE strain responsible for the outbreak in the United Kingdom and two atypical strains (H and L strains). No treatment is currently known for BSE.


Chronic wasting disease (CWD) is a prion disease that affects deer, elk, reindeer, sika deer and moose. It has been found in some areas of North America, including Canada and the United States, Norway and South Korea. It may take over a year before an infected animal develops symptoms, which can include drastic weight loss (wasting), stumbling, listlessness and other neurologic symptoms. CWD can affect animals of all ages and some infected animals may die without ever developing the disease. CWD is fatal to animals and there are no treatments or vaccines.


The causative agents of TSEs are believed to be prions. The term “prions” refers to abnormal, pathogenic agents that are transmissible and are able to induce abnormal folding of specific normal cellular proteins called prion proteins that are found most abundantly in the brain. The functions of these normal prion proteins are still not completely understood. The abnormal folding of the prion proteins leads to brain damage and the characteristic signs and symptoms of the disease. Prion diseases are usually rapidly progressive and always fatal.


As used herein, the term “prion” shall mean an infectious particle known to cause diseases (spongiform encephalopathies) in humans and animals. The term “prion” is a contraction of the words “protein” and “infection” and the particles are comprised largely if not exclusively of PRNPSc molecules encoded by a PRNP gene which expresses PRNPC which changes conformation to become PRNPSc. Prions are distinct from bacteria, viruses and viroids. Known prions include those which infect animals to cause scrapie, a transmissible, degenerative disease of the nervous system of sheep and goats as well as bovine spongiform encephalopathies (BSE) or mad cow disease and feline spongiform encephalopathies of cats. Four prion diseases, as discussed above, known to affect humans are (1) kuru, (2) Creutzfeldt-Jakob Disease (CJD), (3) Gerstmann-Strassler-Scheinker Disease (GSS), and (4) fatal familial insomnia (FFI). As used herein prion includes all forms of prions causing all or any of these diseases or others in any animals used—and in particular in humans and in domesticated farm animals.


In general, and without wishing to be bound by theory, prior diseases are caused by misfolding of prion proteins. Such diseases—often called deposition diseases—the misfolding of the prion proteins can be accounted for as follows. If A is the normally synthesized gene product that carries out an intended physiologic role in a monomeric or oligomeric state, A* is a conformationally activated form of A that is competent to undergo a dramatic conformational change, B is the conformationally altered state that prefers multimeric assemblies (i.e., the misfolded form which forms depositions) and Bn is the multimeric material that is pathogenic and relatively difficult to recycle. For the prion diseases, PRNPC and PRNPSc correspond to states A and Bn where A is largely helical and monomeric and Bn is β-rich and multimeric.


It is known that certain mutations in prion proteins can be associated with increased risk of prior disease. Conversely, certain mutations in prion proteins can be protective in nature. See Bagynszky et al., “Characterization of mutations in PRNP (prion) gene and their possible roles in neurodegenerative diseases,” Neuropsychiatr Dis Treat., 2018; 14: 2067-2085, the contents of which are incorporated herein by reference.


PRNP (NCBI RefSeq No. NP_000302.1 (SEQ ID NO: 291))—the human prion protein—is encoded by a 16 kb long gene, located on chromosome 20 (4686151-4701588). It contains two exons, and the exon 2 carries the open reading frame which encodes the 253 amino acid (AA) long PrP protein. Exon 1 is a noncoding exon, which may serve as transcriptional initiation site. The post-translational modifications result in the removal of the first 22 AA N-terminal fragment (NTF) and the last 23 AA C-terminal fragment (CTF). The NTF is cleaved after PrP transport to the endoplasmic reticulum (ER), while the CTF (glycosylphosphatidylinositol [GPI] signal peptide [GPI-SP]) is cleaved by the GPI anchor. GPI anchor could be involved in PrP protein transport. It may also play a role of attachment of prion protein into the outer surface of cell membrane. Normal PrP is composed of a long N-terminal loop (which contains the octapeptide repeat region), two short β sheets, three a helices, and a C-terminal region (which contains the GPI anchor). Cleavage of PrP results in a 208 AA long glyocoprotein, anchored in the cell membrane.


The 253 amino acid sequence of PRNP (NP_000302.1) is as follows:









(SEQ ID NO: 291)


MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYP





PQGGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWN





KPSKPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYY





RENMHRYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTE





TDVKMMERVVEQMCITQYERESQAYYQRGSSMVLFSSPPVILLISFLIFL





IVG.






The 253 amino acid sequence of PRNP (NP_000302.1) is encoded by the following nucleotide sequence (NCBI Ref. Seq No. NM_000311.5, “Homo sapiens prion protein (PRNP), transcript variant 1, mRNA), is as follows:









(SEQ ID NO: 292)


GCGAACCTTGGCTGCTGGATGCTGGTTCTCTTTGTGGCCACATGGAGTGA





CCTGGGCCTCTGCAAGAAGCGCCCGAAGCCTGGAGGATGGAACACTGGGG





GCAGCCGATACCCGGGGCAGGGCAGCCCTGGAGGCAACCGCTACCCACCT





CAGGGCGGTGGTGGCTGGGGGCAGCCTCATGGTGGTGGCTGGGGGCAGCC





TCATGGTGGTGGCTGGGGGCAGCCCCATGGTGGTGGCTGGGGACAGCCTC





ATGGTGGTGGCTGGGGTCAAGGAGGTGGCACCCACAGTCAGTGGAACAAG





CCGAGTAAGCCAAAAACCAACATGAAGCACATGGCTGGTGCTGCAGCAGC





TGGGGCAGTGGTGGGGGGCCTTGGCGGCTACATGCTGGGAAGTGCCATGA





GCAGGCCCATCATACATTTCGGCAGTGACTATGAGGACCGTTACTATCGT





GAAAACATGCACCGTTACCCCAACCAAGTGTACTACAGGCCCATGGATGA





GTACAGCAACCAGAACAACTTTGTGCACGACTGCGTCAATATCACAATCA





AGCAGCACACGGTCACCACAACCACCAAGGGGGAGAACTTCACCGAGACC





GACGTTAAGATGATGGAGCGCGTGGTTGAGCAGATGTGTATCACCCAGTA





CGAGAGGGAATCTCAGGCCTATTACCAGAGAGGATCGAGCATGGTCCTCT





TCTCCTCTCCACCTGTGATCCTCCTGATCTCTTTCCTCATCTTCCTGATA





GTGGGATGAGGAAGGTCTTCCTGTTTTCACCATCTTTCTAATCTTTTTCC





AGCTTGAGGGAGGCGGTATCCACCTGCAGCCCTTTTAGTGGTGGTGTCTC





ACTCTTTCTTCTCTCTTTGTCCCGGATAGGCTAATCAATACCCTTGGCAC





TGATGGGCACTGGAAAACATAGAGTAGACCTGAGATGCTGGTCAAGCCCC





CTTTGATTGAGTTCATCATGAGCCGTTGCTAATGCCAGGCCAGTAAAAGT





ATAACAGCAAATAACCATTGGTTAATCTGGACTTATTTTTGGACTTAGTG





CAACAGGTTGAGGCTAAAACAAATCTCAGAACAGTCTGAAATACCTTTGC





CTGGATACCTCTGGCTCCTTCAGCAGCTAGAGCTCAGTATACTAATGCCC





TATCTTAGTAGAGATTTCATAGCTATTTAGAGATATTTTCCATTTTAAGA





AAACCCGACAACATTTCTGCCAGGTTTGTTAGGAGGCCACATGATACTTA





TTCAAAAAAATCCTAGAGATTCTTAGCTCTTGGGATGCAGGCTCAGCCCG





CTGGAGCATGAGCTCTGTGTGTACCGAGAACTGGGGTGATGTTTTACTTT





TCACAGTATGGGCTACACAGCAGCTGTTCAACAAGAGTAAATATTGTCAC





AACACTGAACCTCTGGCTAGAGGACATATTCACAGTGAACATAACTGTAA





CATATATGAAAGGCTTCTGGGACTTGAAATCAAATGTTTGGGAATGGTGC





CCTTGGAGGCAACCTCCCATTTTAGATGTTTAAAGGACCCTATATGTGGC





ATTCCTTTCTTTAAACTATAGGTAATTAAGGCAGCTGAAAAGTAAATTGC





CTTCTAGACACTGAAGGCAAATCTCCTTTGTCCATTTACCTGGAAACCAG





AATGATTTTGACATACAGGAGAGCTGCAGTTGTGAAAGCACCATCATCAT





AGAGGATGATGTAATTAAAAAATGGTCAGTGTGCAAAGAAAAGAACTGCT





TGCATTTCTTTATTTCTGTCTCATAATTGTCAAAAACCAGAATTAGGTCA





AGTTCATAGTTTCTGTAATTGGCTTTTGAATCAAAGAATAGGGAGACAAT





CTAAAAAATATCTTAGGTTGGAGATGACAGAAATATGATTGATTTGAAGT





GGAAAAAGAAATTCTGTTAATGTTAATTAAAGTAAAATTATTCCCTGAAT





TGTTTGATATTGTCACCTAGCAGATATGTATTACTTTTCTGCAATGTTAT





TATTGGCTTGCACTTTGTGAGTATTCTATGTAAAAATATATATGTATATA





AAATATATATTGCATAGGACAGACTTAGGAGTTTTGTTTAGAGCAGTTAA





CATCTGAAGTGTCTAATGCATTAACTTTTGTAAGGTACTGAATACTTAAT





ATGTGGGAAACCCTTTTGCGTGGTCCTTAGGCTTACAATGTGCACTGAAT





CGTTTCATGTAAGAATCCAAAGTGGACACCATTAACAGGTCTTTGAAATA





TGCATGTACTTTATATTTTCTATATTTGTAACTTTGCATGTTCTTGTTTT





GTTATATAAAAAAATTGTAAATGTTTAATATCTGACTGAAATTAAACGAG





CGAAGATGAGCACCA






Mutation sites relative to PRNP (NP_000302.1) which are linked to CJD and FFI are reported are as follows. These mutations can be removed or installed using the multi-flap prime editors disclosed herein.














AMINO ACID SEQUENCE OF MUTANT PRNP LINKED TO CJD PRION DISEASE



(SEE TABLE 1 OF BAGYNSZKY ET AL., 2018)


MUTATION
(RELATIVE TO SEQ ID NO: 291 OF PRNP NP_000302.1)







D178N
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHNCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3940)





T188K
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHKVTTTTKGENFTETDVKMMER



VVEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3941)





E196K
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGKNFTETDVKMMER



VVEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3942)





E196A
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGANFTETDVKMMER



VVEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 296)





E200K
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTKTDVKMMER



VVEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3943)





E200G
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTGTDVKMMER



VVEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 298)





V203I
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDIKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3944)





R208H
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMEH



VVEQMCITQYERESQAYYQRGSSMVLFSSPPV(SEQ ID NO: 3945)





V210I
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



IEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3946)





E211Q
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VQQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3947)





M232R
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSRVLFSSPPV (SEQ ID NO: 3948)









Mutation sites relative to PRNP (NP_000302.1) (SEQ ID NO: 291) which are linked to GSS are reported, as follows:














AMINO ACID SEQUENCE OF MUTANT PRNP LINKED TO GSS PRION DISEASE



(SEE TABLE 2 OF BAGYNSZKY ET AL., 2018)


MUTATION
(RELATIVE TO SEQ ID NO: 291 OF PRNP NP_000302.1)







P102L
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKLS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3949)





P105L
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KLKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3950)





A117V
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAVAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMHR



YPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3951)





G131V
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLVSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3952)





V176G
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFGHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3953)





H187R
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQRTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3954)






MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 487)





F198S
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENSTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3955)





D202N
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETNVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3956)





Q212P
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEPMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3957)





Q217R
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITRYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3958)





M232T
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSTVLFSSPPV (SEQ ID NO: 3959)









Mutation sites relative to PRNP (NP_000302.1) (SEQ ID NO: 291) which are linked to a possible protective nature against prion disease, as follows:














AMINO ACID SEQUENCE OF MUTANT PRNP LINKED TO A PROTECTIVE



NATURE AGAINST PRION DISEASE (SEE TABLE 4 OF BAGYNSZKY ET AL.,


MUTATION
2018) (RELATIVE TO SEQ ID NO: 291 OF PRNP NP_000302.1)







G127S
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGSYMLGSAMSRPIIHFGSDYEDRYYRENMHR



YPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3960)





G127V
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGVYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3961)





M129V
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYVLGSAMSRPIIHFGSDYEDRYYRENMHR



YPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3962)





D167G
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMGEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3963)





D167N
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMNEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3964)





N171S
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSSQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3965)





E219K
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYKRESQAYYQRGSSMVLFSSPPV (SEQ ID NO: 3966)





P238S
MANLGCWMLVLFVATWSDLGLCKKRPKPGGWNTGGSRYPGQGSPGGNRYPPQG



GGGWGQPHGGGWGQPHGGGWGQPHGGGWGQPHGGGWGQGGGTHSQWNKPS



KPKTNMKHMAGAAAAGAVVGGLGGYMLGSAMSRPIIHFGSDYEDRYYRENMH



RYPNQVYYRPMDEYSNQNNFVHDCVNITIKQHTVTTTTKGENFTETDVKMMERV



VEQMCITQYERESQAYYQRGSSMVLFSSSPV (SEQ ID NO: 3967)









Thus, in various embodiments, multi-flap prime editing may be used to remove a mutation in PRNP that is linked to prion disease or install a mutation in PRNP that is considered to be protective against prion disease. For example, multi-flap prime editing may be use to remove or restore a D178N, V1801, T188K, E196K, E196A, E200K, E200G, V203I, R208H, V210I, E211Q, I215V, or M232R mutation in the PRNP protein (relative to PRNP of NP_000302.1) (SEQ ID NO: 291). In other embodiments, multi-flap prime editing may be use to remove or restore a P102L, P105L, A117V, G131V, V176G, H187R, F198S, D202N, Q212P, Q217R, or M232T mutation in the PRNP protein (relative to PRNP of NP_000302.1) (SEQ ID NO: 291). By removing or correcting for the presence of such mutations in PRNP using prime editing, the risk of prion disease may be reduced or eliminated.


In other embodiments, multi-flap prime editing may be used to install a protective mutation in PRNP that is linked to a protective effect against one or more prion diseases. For example, multi-flap prime editing may be used to install a G127S, G127V, M129V, D167G, D167N, N171S, E219K, or P238S protective mutation in PRNP (relative to PRNP of NP_000302.1) (SEQ ID NO: 291). In still other embodiments, the protective mutation may be any alternate amino acid installed at G127, G127, M129, D167, D167, N171, E219, or P238 in PRNP (relative to PRNP of NP_000302.1) (SEQ ID NO: 291).


In particular embodiments, multi-flap prime editing may be used to install a G127V protective mutation in PRNP, as illustrated in FIG. 27 and discussed in Example 5.


In another embodiment, prime editing may be used to install an E219K protective mutation in PRNP.


The PRNP protein and the protective mutation site are conserved in mammals, so in addition to treating human disease it could also be used to generate cows and sheep that are immune to prion disease, or even help cure wild populations of animals that are suffering from prion disease. Prime editing has already been used to achieve ˜25% installation of a naturally occurring protective allele in human cells, and previous mouse experiments indicate that this level of installation is sufficient to cause immunity from most prion diseases. This method is the first and potentially only current way to install this allele with such high efficiency in most cell types. Another possible strategy for treatment is to use prime editing or multi-flap prime editing to reduce or eliminate the expression of PRNP by installing an early stop codon in the gene.


Using the principles described herein for PEgRNA design, appropriate PEgRNAs may be designed for installing desired protective mutations, or for removing prion disease-associated mutations from PRNP. For example, the below list of PEgRNAs can be used to install the G127V protective allele and the E219K protective allele in human PRNP, as well as the G127V protective allele in PRNP of various animals.


[10] Pharmaceutical Compositions

Other aspects of the present disclosure relate to pharmaceutical compositions comprising any of the various components of the multi-flap prime editing system described herein (e.g., including, but not limited to, the napDNAbps, reverse transcriptases, fusion proteins (e.g., comprising napDNAbps and reverse transcriptases), extended guide RNAs, and complexes comprising fusion proteins and extended guide RNAs, as well as accessory elements, such as second strand nicking components and 5′ endogenous DNA flap removal endonucleases for helping to drive the multi-flap prime editing process towards the edited product formation).


The term “pharmaceutical composition”, as used herein, refers to a composition formulated for pharmaceutical use. In some embodiments, the pharmaceutical composition further comprises a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition comprises additional agents (e.g. for specific delivery, increasing half-life, or other therapeutic compounds).


As used here, the term “pharmaceutically-acceptable carrier” means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the compound from one site (e.g., the delivery site) of the body, to another site (e.g., organ, tissue or portion of the body). A pharmaceutically acceptable carrier is “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the tissue of the subject (e.g., physiologically compatible, sterile, physiologic pH, etc.). Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) pH buffered solutions; (21) polyesters, polycarbonates and/or polyanhydrides; (22) bulking agents, such as polypeptides and amino acids (23) serum component, such as serum albumin, HDL and LDL; (22) C2-C12 alcohols, such as ethanol; and (23) other non-toxic compatible substances employed in pharmaceutical formulations. Wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation. The terms such as “excipient”, “carrier”, “pharmaceutically acceptable carrier” or the like are used interchangeably herein.


In some embodiments, the pharmaceutical composition is formulated for delivery to a subject, e.g., for gene editing. Suitable routes of administrating the pharmaceutical composition described herein include, without limitation: topical, subcutaneous, transdermal, intradermal, intralesional, intraarticular, intraperitoneal, intravesical, transmucosal, gingival, intradental, intracochlear, transtympanic, intraorgan, epidural, intrathecal, intramuscular, intravenous, intravascular, intraosseus, periocular, intratumoral, intracerebral, and intracerebroventricular administration.


In some embodiments, the pharmaceutical composition described herein is administered locally to a diseased site (e.g., tumor site). In some embodiments, the pharmaceutical composition described herein is administered to a subject by injection, by means of a catheter, by means of a suppository, or by means of an implant, the implant being of a porous, non-porous, or gelatinous material, including a membrane, such as a sialastic membrane, or a fiber.


In other embodiments, the pharmaceutical composition described herein is delivered in a controlled release system. In one embodiment, a pump may be used (see, e.g., Langer, 1990, Science 249:1527-1533; Sefton, 1989, CRC Crit. Ref. Biomed. Eng. 14:201; Buchwald et al., 1980, Surgery 88:507; Saudek et al., 1989, N. Engl. J. Med. 321:574). In another embodiment, polymeric materials can be used. (See, e.g., Medical Applications of Controlled Release (Langer and Wise eds., CRC Press, Boca Raton, Fla., 1974); Controlled Drug Bioavailability, Drug Product Design and Performance (Smolen and Ball eds., Wiley, New York, 1984); Ranger and Peppas, 1983, Macromol. Sci. Rev. Macromol. Chem. 23:61. See also Levy et al., 1985, Science 228:190; During et al., 1989, Ann. Neurol. 25:351; Howard et al., 1989, J. Neurosurg. 71:105). Other controlled release systems are discussed, for example, in Langer, supra.


In some embodiments, the pharmaceutical composition is formulated in accordance with routine procedures as a composition adapted for intravenous or subcutaneous administration to a subject, e.g., a human. In some embodiments, pharmaceutical composition for administration by injection are solutions in sterile isotonic aqueous buffer. Where necessary, the pharmaceutical can also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the pharmaceutical is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the pharmaceutical composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.


A pharmaceutical composition for systemic administration may be a liquid, e.g., sterile saline, lactated Ringer's or Hank's solution. In addition, the pharmaceutical composition can be in solid forms and re-dissolved or suspended immediately prior to use. Lyophilized forms are also contemplated.


The pharmaceutical composition can be contained within a lipid particle or vesicle, such as a liposome or microcrystal, which is also suitable for parenteral administration. The particles can be of any suitable structure, such as unilamellar or plurilamellar, so long as compositions are contained therein. Compounds can be entrapped in “stabilized plasmid-lipid particles” (SPLP) containing the fusogenic lipid dioleoylphosphatidylethanolamine (DOPE), low levels (5-10 mol %) of cationic lipid, and stabilized by a polyethyleneglycol (PEG) coating (Zhang Y. P. et al., Gene Ther. 1999, 6:1438-47). Positively charged lipids such as N-[1-(2,3-dioleoyloxi)propyl]-N,N,N-trimethyl-amoniummethylsulfate, or “DOTAP,” are particularly preferred for such particles and vesicles. The preparation of such lipid particles is well known. See, e.g., U.S. Pat. Nos. 4,880,635; 4,906,477; 4,911,928; 4,917,951; 4,920,016; and 4,921,757; each of which is incorporated herein by reference.


The pharmaceutical composition described herein may be administered or packaged as a unit dose, for example. The term “unit dose” when used in reference to a pharmaceutical composition of the present disclosure refers to physically discrete units suitable as unitary dosage for the subject, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required diluent; i.e., carrier, or vehicle.


Further, the pharmaceutical composition can be provided as a pharmaceutical kit comprising (a) a container containing a compound of the invention in lyophilized form and (b) a second container containing a pharmaceutically acceptable diluent (e.g., sterile water) for injection. The pharmaceutically acceptable diluent can be used for reconstitution or dilution of the lyophilized compound of the invention. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.


In another aspect, an article of manufacture containing materials useful for the treatment of the diseases described above is included. In some embodiments, the article of manufacture comprises a container and a label. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers may be formed from a variety of materials such as glass or plastic. In some embodiments, the container holds a composition that is effective for treating a disease described herein and may have a sterile access port. For example, the container may be an intravenous solution bag or a vial having a stopper pierce-able by a hypodermic injection needle. The active agent in the composition is a compound of the invention. In some embodiments, the label on or associated with the container indicates that the composition is used for treating the disease of choice. The article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution, or dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.


[11] Kits, Cells, Vectors, and Delivery

Kits


The compositions of the present disclosure may be assembled into kits. In some embodiments, the kit comprises nucleic acid vectors for the expression of the multi-flap prime editors described herein. In other embodiments, the kit further comprises appropriate guide nucleotide sequences (e.g., PEgRNAs and second-site gRNAs) or nucleic acid vectors for the expression of such guide nucleotide sequences, to target the Cas9 protein or prime editor to the desired target sequence.


The kit described herein may include one or more containers housing components for performing the methods described herein and optionally instructions for use. Any of the kit described herein may further comprise components needed for performing the assay methods. Each component of the kits, where applicable, may be provided in liquid form (e.g., in solution) or in solid form, (e.g., a dry powder). In certain cases, some of the components may be reconstitutable or otherwise processible (e.g., to an active form), for example, by the addition of a suitable solvent or other species (for example, water), which may or may not be provided with the kit.


In some embodiments, the kits may optionally include instructions and/or promotion for use of the components provided. As used herein, “instructions” can define a component of instruction and/or promotion, and typically involve written instructions on or associated with packaging of the disclosure. Instructions also can include any oral or electronic instructions provided in any manner such that a user will clearly recognize that the instructions are to be associated with the kit, for example, audiovisual (e.g., videotape, DVD, etc.), Internet, and/or web-based communications, etc. The written instructions may be in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals or biological products, which can also reflect approval by the agency of manufacture, use or sale for animal administration. As used herein, “promoted” includes all methods of doing business including methods of education, hospital and other clinical instruction, scientific inquiry, drug discovery or development, academic research, pharmaceutical industry activity including pharmaceutical sales, and any advertising or other promotional activity including written, oral and electronic communication of any form, associated with the disclosure. Additionally, the kits may include other components depending on the specific application, as described herein.


The kits may contain any one or more of the components described herein in one or more containers. The components may be prepared sterilely, packaged in a syringe and shipped refrigerated. Alternatively, it may be housed in a vial or other container for storage. A second container may have other components prepared sterilely. Alternatively, the kits may include the active agents premixed and shipped in a vial, tube, or other container.


The kits may have a variety of forms, such as a blister pouch, a shrink-wrapped pouch, a vacuum sealable pouch, a sealable thermoformed tray, or a similar pouch or tray form, with the accessories loosely packed within the pouch, one or more tubes, containers, a box or a bag. The kits may be sterilized after the accessories are added, thereby allowing the individual accessories in the container to be otherwise unwrapped. The kits can be sterilized using any appropriate sterilization techniques, such as radiation sterilization, heat sterilization, or other sterilization methods known in the art. The kits may also include other components, depending on the specific application, for example, containers, cell media, salts, buffers, reagents, syringes, needles, a fabric, such as gauze, for applying or removing a disinfecting agent, disposable gloves, a support for the agents prior to administration, etc. Some aspects of this disclosure provide kits comprising a nucleic acid construct comprising a nucleotide sequence encoding the various components of the multi-flap prime editing systems (e.g., dual prime editing and quadruple prime editing systems) described herein (e.g., including, but not limited to, the napDNAbps, reverse transcriptases, polymerases, fusion proteins (e.g., comprising napDNAbps and reverse transcriptases (or more broadly, polymerases), extended guide RNAs, and complexes comprising fusion proteins and extended guide RNAs, as well as accessory elements, such as second strand nicking components (e.g., second strand nicking gRNA) and 5′ endogenous DNA flap removal endonucleases for helping to drive the multi-flap prime editing process towards the edited product formation). In some embodiments, the nucleotide sequence(s) comprises a heterologous promoter (or more than a single promoter) that drives expression of the multi-flap prime editing system components.


Other aspects of this disclosure provide kits comprising one or more nucleic acid constructs encoding the various components of the multi-flap prime editing systems described herein, e.g., comprising a nucleotide sequence encoding the components of the multi-flap prime editing system capable of modifying a target DNA sequence. In some embodiments, the nucleotide sequence comprises a heterologous promoter that drives expression of the multi-flap prime editing system components.


Some aspects of this disclosure provide kits comprising a nucleic acid construct, comprising (a) a nucleotide sequence encoding a napDNAbp (e.g., a Cas9 domain) fused to a reverse transcriptase and (b) a heterologous promoter that drives expression of the sequence of (a).


Cells


Cells that may contain any of the compositions described herein include prokaryotic cells and eukaryotic cells. The methods described herein are used to deliver a Cas9 protein or a multi-flap prime editor into a eukaryotic cell (e.g., a mammalian cell, such as a human cell). In some embodiments, the cell is in vitro (e.g., cultured cell. In some embodiments, the cell is in vivo (e.g., in a subject such as a human subject). In some embodiments, the cell is ex vivo (e.g., isolated from a subject and may be administered back to the same or a different subject).


Mammalian cells of the present disclosure include human cells, primate cells (e.g., vero cells), rat cells (e.g., GH3 cells, 0C23 cells) or mouse cells (e.g., MC3T3 cells). There are a variety of human cell lines, including, without limitation, human embryonic kidney (HEK) cells, HeLa cells, cancer cells from the National Cancer Institute's 60 cancer cell lines (NCI60), DU145 (prostate cancer) cells, Lncap (prostate cancer) cells, MCF-7 (breast cancer) cells, MDA-MB-438 (breast cancer) cells, PC3 (prostate cancer) cells, T47D (breast cancer) cells, THP-1 (acute myeloid leukemia) cells, U87 (glioblastoma) cells, SHSY5Y human neuroblastoma cells (cloned from a myeloma) and Saos-2 (bone cancer) cells. In some embodiments, rAAV vectors are delivered into human embryonic kidney (HEK) cells (e.g., HEK 293 or HEK 293T cells). In some embodiments, rAAV vectors are delivered into stem cells (e.g., human stem cells) such as, for example, pluripotent stem cells (e.g., human pluripotent stem cells including human induced pluripotent stem cells (hiPSCs)). A stem cell refers to a cell with the ability to divide for indefinite periods in culture and to give rise to specialized cells. A pluripotent stem cell refers to a type of stem cell that is capable of differentiating into all tissues of an organism, but not alone capable of sustaining full organismal development. A human induced pluripotent stem cell refers to a somatic (e.g., mature or adult) cell that has been reprogrammed to an embryonic stem cell-like state by being forced to express genes and factors important for maintaining the defining properties of embryonic stem cells (see, e.g., Takahashi and Yamanaka, Cell 126 (4): 663-76, 2006, incorporated by reference herein). Human induced pluripotent stem cell cells express stem cell markers and are capable of generating cells characteristic of all three germ layers (ectoderm, endoderm, mesoderm).


Additional non-limiting examples of cell lines that may be used in accordance with the present disclosure include 293-T, 293-T, 3T3, 4T1, 721, 9L, A-549, A172, A20, A253, A2780, A2780ADR, A2780cis, A431, ALC, B16, B35, BCP-1, BEAS-2B, bEnd.3, BHK-21, BR 293, BxPC3, C2C12, C3H-10T1/2, C6, C6/36, Cal-27, CGR8, CHO, CML Tl, CMT, COR-L23, COR-L23/5010, COR-L23/CPR, COR-L23/R23, COS-7, COV-434, CT26, D17, DH82, DU145, DuCaP, E14Tg2a, EL4, EM2, EM3, EMT6/AR1, EMT6/AR10.0, FM3, H1299, H69, HB54, HB55, HCA2, Hepa1c1c7, High Five cells, HL-60, HMEC, HT-29, HUVEC, J558L cells, Jurkat, JY cells, K562 cells, KCL22, KG1, Ku812, KYO1, LNCap, Ma-Mel 1, 2, 3 . . . 48, MC-38, MCF-10A, MCF-7, MDA-MB-231, MDA-MB-435, MDA-MB-468, MDCK II, MG63, MONO-MAC 6, MOR/0.2R, MRCS, MTD-1A, MyEnd, NALM-1, NCI-H69/CPR, NCI-H69/LX10, NCI-H69/LX20, NCI-H69/LX4, NIH-3T3, NW-145, OPCN/OPCT Peer, PNT-1A/PNT 2, PTK2, Raji, RBL cells, RenCa, RIN-5F, RMA/RMAS, S2, Saos-2 cells, Sf21, Sf9, SiHa, SKBR3, SKOV-3, T-47D, T2, T84, THP1, U373, U87, U937, VCaP, WM39, WT-49, X63, YAC-1 and YAR cells.


Some aspects of this disclosure provide cells comprising any of the constructs disclosed herein. In some embodiments, a host cell is transiently or non-transiently transfected with one or more vectors described herein. In some embodiments, a cell is transfected as it naturally occurs in a subject. In some embodiments, a cell that is transfected is taken from a subject. In some embodiments, the cell is derived from cells taken from a subject, such as a cell line. A wide variety of cell lines for tissue culture are known in the art. Examples of cell lines include, but are not limited to, C8161, CCRF-CEM, MOLT, mIMCD-3, NHDF, HeLa-S3, Huh1, Huh4, Huh7, HUVEC, HASMC, HEKn, HEKa, MiaPaCell, Panc1, PC-3, TF1, CTLL-2, C1R, Rat6, CV1, RPTE, A10, T24, J82, A375, ARH-77, Calu1, SW480, SW620, SKOV3, SK-UT, CaCo2, P388D1, SEM-K2, WEHI-231, HB56, TIB55, Jurkat, J45.01, LRMB, Bcl-1, BC-3, IC21, DLD2, Raw264.7, NRK, NRK-52E, MRCS, MEF, Hep G2, HeLa B, HeLa T4, COS, COS-1, COS-6, COS-M6A, BS-C-1 monkey kidney epithelial, BALB/3T3 mouse embryo fibroblast, 3T3 Swiss, 3T3-L1, 132-d5 human fetal fibroblasts; 10.1 mouse fibroblasts, 293-T, 3T3, 721, 9L, A2780, A2780ADR, A2780cis, A 172, A20, A253, A431, A-549, ALC, B16, B35, BCP-1 cells, BEAS-2B, bEnd.3, BHK-21, BR 293. BxPC3. C3H-10T1/2, C6/36, Cal-27, CHO, CHO-7, CHO-IR, CHO-K1, CHO-K2, CHO-T, CHO Dhfr−/−, COR-L23, COR-L23/CPR, COR-L23/5010, COR-L23/R23, COS-7, COV-434, CML T1, CMT, CT26, D17, DH82, DU145, DuCaP, EL4, EM2, EM3, EMT6/AR1, EMT6/AR10.0, FM3, H1299, H69, HB54, HB55, HCA2, HEK-293, HeLa, Hepa1c1c7, HL-60, HMEC, HT-29, Jurkat, JY cells, K562 cells, Ku812, KCL22, KG1, KYO1, LNCap, Ma-Mel 1-48, MC-38, MCF-7, MCF-10A, MDA-MB-231, MDA-MB-468, MDA-MB-435, MDCK II, MDCK 11, MOR/0.2R, MONO-MAC 6, MTD-1A, MyEnd, NCI-H69/CPR, NCI-H69/LX10, NCI-H69/LX20, NCI-H69/LX4, NIH-3T3, NALM-1, NW-145, OPCN/OPCT cell lines, Peer, PNT-1A/PNT 2, RenCa, RIN-5F, RMA/RMAS, Saos-2 cells, Sf-9, SkBr3, T2, T-47D, T84, THP1 cell line, U373, U87, U937, VCaP, Vero cells, WM39, WT-49, X63, YAC-1, YAR, and transgenic varieties thereof.


Cell lines are available from a variety of sources known to those with skill in the art (see, e.g., the American Type Culture Collection (ATCC) (Manassus, Va.)). In some embodiments, a cell transfected with one or more vectors described herein is used to establish a new cell line comprising one or more vector-derived sequences. In some embodiments, a cell transiently transfected with the components of a CRISPR system as described herein (such as by transient transfection of one or more vectors, or transfection with RNA), and modified through the activity of a CRISPR complex, is used to establish a new cell line comprising cells containing the modification but lacking any other exogenous sequence. In some embodiments, cells transiently or non-transiently transfected with one or more vectors described herein, or cell lines derived from such cells are used in assessing one or more test compounds.


Vectors


Some aspects of the present disclosure relate to using recombinant virus vectors (e.g., adeno-associated virus vectors, adenovirus vectors, or herpes simplex virus vectors) for the delivery of the multi-flap prime editors or components thereof described herein, e.g., the split Cas9 protein or a split nucleobase multi-flap prime editors, into a cell. In the case of a split-PE approach, the N-terminal portion of a PE fusion protein and the C-terminal portion of a PE fusion are delivered by separate recombinant virus vectors (e.g., adeno-associated virus vectors, adenovirus vectors, or herpes simplex virus vectors) into the same cell, since the full-length Cas9 protein or multi-flap prime editors exceeds the packaging limit of various virus vectors, e.g., rAAV (˜4.9 kb).


Thus, in one embodiment, the disclosure contemplates vectors capable of delivering split multi-flap prime editor fusion proteins, or split components thereof. In some embodiments, a composition for delivering the split Cas9 protein or split prime editor into a cell (e.g., a mammalian cell, a human cell) is provided. In some embodiments, the composition of the present disclosure comprises: (i) a first recombinant adeno-associated virus (rAAV) particle comprising a first nucleotide sequence encoding a N-terminal portion of a Cas9 protein or prime editor fused at its C-terminus to an intein-N; and (ii) a second recombinant adeno-associated virus (rAAV) particle comprising a second nucleotide sequence encoding an intein-C fused to the N-terminus of a C-terminal portion of the Cas9 protein or prime editor. The rAAV particles of the present disclosure comprise a rAAV vector (i.e., a recombinant genome of the rAAV) encapsidated in the viral capsid proteins.


In some embodiments, the rAAV vector comprises: (1) a heterologous nucleic acid region comprising the first or second nucleotide sequence encoding the N-terminal portion or C-terminal portion of a split Cas9 protein or a split multi-flap prime editor in any form as described herein, (2) one or more nucleotide sequences comprising a sequence that facilitates expression of the heterologous nucleic acid region (e.g., a promoter), and (3) one or more nucleic acid regions comprising a sequence that facilitate integration of the heterologous nucleic acid region (optionally with the one or more nucleic acid regions comprising a sequence that facilitates expression) into the genome of a cell. In some embodiments, viral sequences that facilitate integration comprise Inverted Terminal Repeat (ITR) sequences. In some embodiments, the first or second nucleotide sequence encoding the N-terminal portion or C-terminal portion of a split Cas9 protein or a split multi-flap prime editor is flanked on each side by an ITR sequence. In some embodiments, the nucleic acid vector further comprises a region encoding an AAV Rep protein as described herein, either contained within the region flanked by ITRs or outside the region. The ITR sequences can be derived from any AAV serotype (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) or can be derived from more than one serotype. In some embodiments, the ITR sequences are derived from AAV2 or AAV6.


Thus, in some embodiments, the rAAV particles disclosed herein comprise at least one rAAV2 particle, rAAV6 particle, rAAV8 particle, rPHP.B particle, rPHP.eB particle, or rAAV9 particle, or a variant thereof. In particular embodiments, the disclosed rAAV particles are rPHP.B particles, rPHP.eB particles, rAAV9 particles.


ITR sequences and plasmids containing ITR sequences are known in the art and commercially available (see, e.g., products and services available from Vector Biolabs, Philadelphia, PA; Cellbiolabs, San Diego, CA; Agilent Technologies, Santa Clara, Ca; and Addgene, Cambridge, MA; and Gene delivery to skeletal muscle results in sustained expression and systemic delivery of a therapeutic protein. Kessler P D, Podsakoff G M, Chen X, McQuiston S A, Colosi P C, Matelis L A, Kurtzman G J, Byrne B J. Proc Natl Acad Sci USA. 1996 Nov. 26; 93(24):14082-7; and Curtis A. Machida. Methods in Molecular Medicine™. Viral Vectors for Gene Therapy Methods and Protocols. 10.1385/1-59259-304-6:201© Humana Press Inc. 2003. Chapter 10. Targeted Integration by Adeno-Associated Virus. Matthew D. Weitzman, Samuel M. Young Jr., Toni Cathomen and Richard Jude Samulski; U.S. Pat. Nos. 5,139,941 and 5,962,313, all of which are incorporated herein by reference).


In some embodiments, the rAAV vector of the present disclosure comprises one or more regulatory elements to control the expression of the heterologous nucleic acid region (e.g., promoters, transcriptional terminators, and/or other regulatory elements). In some embodiments, the first and/or second nucleotide sequence is operably linked to one or more (e.g., 1, 2, 3, 4, 5, or more) transcriptional terminators. Non-limiting examples of transcriptional terminators that may be used in accordance with the present disclosure include transcription terminators of the bovine growth hormone gene (bGH), human growth hormone gene (hGH), SV40, CW3, ϕ, or combinations thereof. The efficiencies of several transcriptional terminators have been tested to determine their respective effects in the expression level of the split Cas9 protein or the split multi-flap prime editor. In some embodiments, the transcriptional terminator used in the present disclosure is a bGH transcriptional terminator. In some embodiments, the rAAV vector further comprises a Woodchuck Hepatitis Virus Posttranscriptional Regulatory Element (WPRE). In certain embodiments, the WPRE is a truncated WPRE sequence, such as “W3.” In some embodiments, the WPRE is inserted 5′ of the transcriptional terminator. Such sequences, when transcribed, create a tertiary structure which enhances expression, in particular, from viral vectors.


In some embodiments, the vectors used herein may encode the PE fusion proteins, or any of the components thereof (e.g., napDNAbp, linkers, or polymerases). In addition, the vectors used herein may encode the PEgRNAs, and/or the accessory gRNA for second strand nicking. The vectors may be capable of driving expression of one or more coding sequences in a cell. In some embodiments, the cell may be a prokaryotic cell, such as, e.g., a bacterial cell. In some embodiments, the cell may be a eukaryotic cell, such as, e.g., a yeast, plant, insect, or mammalian cell. In some embodiments, the eukaryotic cell may be a mammalian cell. In some embodiments, the eukaryotic cell may be a rodent cell. In some embodiments, the eukaryotic cell may be a human cell. Suitable promoters to drive expression in different types of cells are known in the art. In some embodiments, the promoter may be wild-type. In other embodiments, the promoter may be modified for more efficient or efficacious expression. In yet other embodiments, the promoter may be truncated yet retain its function. For example, the promoter may have a normal size or a reduced size that is suitable for proper packaging of the vector into a virus.


In some embodiments, the promoters that may be used in the prime editor vectors may be constitutive, inducible, or tissue-specific. In some embodiments, the promoters may be a constitutive promoters. Non-limiting exemplary constitutive promoters include cytomegalovirus immediate early promoter (CMV), simian virus (SV40) promoter, adenovirus major late (MLP) promoter, Rous sarcoma virus (RSV) promoter, mouse mammary tumor virus (MMTV) promoter, phosphoglycerate kinase (PGK) promoter, elongation factor-alpha (EF1a) promoter, ubiquitin promoters, actin promoters, tubulin promoters, immunoglobulin promoters, a functional fragment thereof, or a combination of any of the foregoing. In some embodiments, the promoter may be a CMV promoter. In some embodiments, the promoter may be a truncated CMV promoter. In other embodiments, the promoter may be an EF1a promoter. In some embodiments, the promoter may be an inducible promoter. Non-limiting exemplary inducible promoters include those inducible by heat shock, light, chemicals, peptides, metals, steroids, antibiotics, or alcohol. In some embodiments, the inducible promoter may be one that has a low basal (non-induced) expression level, such as, e.g., the Tet-On® promoter (Clontech). In some embodiments, the promoter may be a tissue-specific promoter. In some embodiments, the tissue-specific promoter is exclusively or predominantly expressed in liver tissue. Non-limiting exemplary tissue-specific promoters include B29 promoter, CD14 promoter, CD43 promoter, CD45 promoter, CD68 promoter, desmin promoter, elastase-1 promoter, endoglin promoter, fibronectin promoter, Flt-1 promoter, GFAP promoter, GPIIb promoter, ICAM-2 promoter, INF-β promoter, Mb promoter, Nphs1 promoter, OG-2 promoter, SP-B promoter, SYN1 promoter, and WASP promoter.


In some embodiments, the prime editor vectors (e.g., including any vectors encoding the prime editor fusion protein and/or the PEgRNAs, and/or the accessory second strand nicking gRNAs) may comprise inducible promoters to start expression only after it is delivered to a target cell. Non-limiting exemplary inducible promoters include those inducible by heat shock, light, chemicals, peptides, metals, steroids, antibiotics, or alcohol. In some embodiments, the inducible promoter may be one that has a low basal (non-induced) expression level, such as, e.g., the Tet-On® promoter (Clontech).


In additional embodiments, the prime editor vectors (e.g., including any vectors encoding the prime editor fusion protein and/or the PEgRNAs, and/or the accessory second strand nicking gRNAs) may comprise tissue-specific promoters to start expression only after it is delivered into a specific tissue. Non-limiting exemplary tissue-specific promoters include B29 promoter, CD14 promoter, CD43 promoter, CD45 promoter, CD68 promoter, desmin promoter, elastase-1 promoter, endoglin promoter, fibronectin promoter, Flt-1 promoter, GFAP promoter, GPIIb promoter, ICAM-2 promoter, INF-β promoter, Mb promoter, Nphs1 promoter, OG-2 promoter, SP-B promoter, SYN1 promoter, and WASP promoter.


In some embodiments, the nucleotide sequence encoding the PEgRNA (or any guide RNAs used in connection with multi-flap prime editing) may be operably linked to at least one transcriptional or translational control sequence. In some embodiments, the nucleotide sequence encoding the guide RNA may be operably linked to at least one promoter. In some embodiments, the promoter may be recognized by RNA polymerase III (Pol III). Non-limiting examples of Pol III promoters include U6, HI and tRNA promoters. In some embodiments, the nucleotide sequence encoding the guide RNA may be operably linked to a mouse or human U6 promoter. In other embodiments, the nucleotide sequence encoding the guide RNA may be operably linked to a mouse or human HI promoter. In some embodiments, the nucleotide sequence encoding the guide RNA may be operably linked to a mouse or human tRNA promoter. In embodiments with more than one guide RNA, the promoters used to drive expression may be the same or different. In some embodiments, the nucleotide encoding the crRNA of the guide RNA and the nucleotide encoding the tracr RNA of the guide RNA may be provided on the same vector. In some embodiments, the nucleotide encoding the crRNA and the nucleotide encoding the tracr RNA may be driven by the same promoter. In some embodiments, the crRNA and tracr RNA may be transcribed into a single transcript. For example, the crRNA and tracr RNA may be processed from the single transcript to form a double-molecule guide RNA. Alternatively, the crRNA and tracr RNA may be transcribed into a single-molecule guide RNA.


In some embodiments, the nucleotide sequence encoding the guide RNA may be located on the same vector comprising the nucleotide sequence encoding the PE fusion protein. In some embodiments, expression of the guide RNA and of the PE fusion protein may be driven by their corresponding promoters. In some embodiments, expression of the guide RNA may be driven by the same promoter that drives expression of the PE fusion protein. In some embodiments, the guide RNA and the PE fusion protein transcript may be contained within a single transcript. For example, the guide RNA may be within an untranslated region (UTR) of the Cas9 protein transcript. In some embodiments, the guide RNA may be within the 5′ UTR of the PE fusion protein transcript. In other embodiments, the guide RNA may be within the 3′ UTR of the PE fusion protein transcript. In some embodiments, the intracellular half-life of the PE fusion protein transcript may be reduced by containing the guide RNA within its 3′ UTR and thereby shortening the length of its 3′ UTR. In additional embodiments, the guide RNA may be within an intron of the PE fusion protein transcript. In some embodiments, suitable splice sites may be added at the intron within which the guide RNA is located such that the guide RNA is properly spliced out of the transcript. In some embodiments, expression of the Cas9 protein and the guide RNA in close proximity on the same vector may facilitate more efficient formation of the CRISPR complex.


The multi-flap prime editor vector system may comprise one vector, or two vectors, or three vectors, or four vectors, or five vector, or more. In some embodiments, the vector system may comprise one single vector, which encodes both the PE fusion protein and PEgRNA. In other embodiments, the vector system may comprise two vectors, wherein one vector encodes the PE fusion protein and the other encodes the PEgRNA. In additional embodiments, the vector system may comprise three vectors, wherein the third vector encodes the second strand nicking gRNA used in the herein methods.


In some embodiments, the composition comprising the rAAV particle (in any form contemplated herein) further comprises a pharmaceutically acceptable carrier. In some embodiments, the composition is formulated in appropriate pharmaceutical vehicles for administration to human or animal subjects.


Some examples of materials which can serve as pharmaceutically-acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) pH buffered solutions; (21) polyesters, polycarbonates and/or polyanhydrides; (22) bulking agents, such as polypeptides and amino acids (23) serum component, such as serum albumin, HDL and LDL; (22) C2-C12 alcohols, such as ethanol; and (23) other non-toxic compatible substances employed in pharmaceutical formulations. Wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation. The terms such as “excipient”, “carrier”, “pharmaceutically acceptable carrier” or the like are used interchangeably herein.


Delivery Methods


In some aspects, the invention provides methods comprising delivering one or more polynucleotides encoding the various components of the multi-flap prime editors described herein, such as or one or more vectors as described herein, one or more transcripts thereof, and/or one or proteins transcribed therefrom, to a host cell. In some aspects, the invention further provides cells produced by such methods, and organisms (such as animals, plants, or fungi) comprising or produced from such cells. In some embodiments, a base editor as described herein in combination with (and optionally complexed with) a guide sequence is delivered to a cell.


Exemplary delivery strategies are described herein elsewhere, which include vector-based strategies, PE ribonucleoprotein complex delivery, and delivery of PE by mRNA methods.


In some embodiments, the method of delivery provided comprises nucleofection, microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipid:nucleic acid conjugates, naked DNA, artificial virions, and agent-enhanced uptake of DNA.


Exemplary methods of delivery of nucleic acids include lipofection, nucleofection, electroporation, stable genome integration (e.g., piggybac), microinjection, biolistics, virosomes, liposomes, immunoliposomes, polycation or lipid:nucleic acid conjugates, naked DNA, artificial virions, and agent-enhanced uptake of DNA. Lipofection is described in e.g., U.S. Pat. Nos. 5,049,386, 4,946,787; and 4,897,355) and lipofection reagents are sold commercially (e.g., Transfectam™, Lipofectin™ and SF Cell Line 4D-Nucleofector X Kit™ (Lonza)). Cationic and neutral lipids that are suitable for efficient receptor-recognition lipofection of polynucleotides include those of Feigner, WO 91/17424; WO 91/16024. Delivery may be to cells (e.g. in vitro or ex vivo administration) or target tissues (e.g. in vivo administration). Delivery may be achieved through the use of RNP complexes.


The preparation of lipid:nucleic acid complexes, including targeted liposomes such as immunolipid complexes, is well known to one of skill in the art (see, e.g., Crystal, Science 270:404-410 (1995); Blaese et al., Cancer Gene Ther. 2:291-297 (1995); Behr et al., Bioconjugate Chem. 5:382-389 (1994); Remy et al., Bioconjugate Chem. 5:647-654 (1994); Gao et al., Gene Therapy 2:710-722 (1995); Ahmad et al., Cancer Res. 52:4817-4820 (1992); U.S. Pat. Nos. 4,186,183, 4,217,344, 4,235,871, 4,261,975, 4,485,054, 4,501,728, 4,774,085, 4,837,028, and 4,946,787).


In other embodiments, the method of delivery and vector provided herein is an RNP complex. RNP delivery of fusion proteins markedly increases the DNA specificity of base editing. RNP delivery of fusion proteins leads to decoupling of on- and off-target DNA editing. RNP delivery ablates off-target editing at non-repetitive sites while maintaining on-target editing comparable to plasmid delivery, and greatly reduces off-target DNA editing even at the highly repetitive VEGFA site 2. See Rees, H. A. et al., Improving the DNA specificity and applicability of base editing through protein engineering and protein delivery, Nat. Commun. 8, 15790 (2017), U.S. Pat. No. 9,526,784, issued Dec. 27, 2016, and U.S. Pat. No. 9,737,604, issued Aug. 22, 2017, each of which is incorporated by reference herein.


Additional methods for the delivery of nucleic acids to cells are known to those skilled in the art. See, for example, US 2003/0087817, incorporated herein by reference.


Other aspects of the present disclosure provide methods of delivering the multi-flap prime editor constructs into a cell to form a complete and functional prime editor within a cell. For example, in some embodiments, a cell is contacted with a composition described herein (e.g., compositions comprising nucleotide sequences encoding the split Cas9 or the split prime editor or AAV particles containing nucleic acid vectors comprising such nucleotide sequences). In some embodiments, the contacting results in the delivery of such nucleotide sequences into a cell, wherein the N-terminal portion of the Cas9 protein or the prime editor and the C-terminal portion of the Cas9 protein or the prime editor are expressed in the cell and are joined to form a complete Cas9 protein or a complete prime editor.


It should be appreciated that any rAAV particle, nucleic acid molecule or composition provided herein may be introduced into the cell in any suitable way, either stably or transiently. In some embodiments, the disclosed proteins may be transfected into the cell. In some embodiments, the cell may be transduced or transfected with a nucleic acid molecule. For example, a cell may be transduced (e.g., with a virus encoding a split protein), or transfected (e.g., with a plasmid encoding a split protein) with a nucleic acid molecule that encodes a split protein, or an rAAV particle containing a viral genome encoding one or more nucleic acid molecules. Such transduction may be a stable or transient transduction. In some embodiments, cells expressing a split protein or containing a split protein may be transduced or transfected with one or more guide RNA sequences, for example in delivery of a split Cas9 (e.g., nCas9) protein. In some embodiments, a plasmid expressing a split protein may be introduced into cells through electroporation, transient (e.g., lipofection) and stable genome integration (e.g., piggybac) and viral transduction or other methods known to those of skill in the art.


In certain embodiments, the compositions provided herein comprise a lipid and/or polymer. In certain embodiments, the lipid and/or polymer is cationic. The preparation of such lipid particles is well known. See, e.g. U.S. Pat. Nos. 4,880,635; 4,906,477; 4,911,928; 4,917,951; 4,920,016; 4,921,757; and 9,737,604, each of which is incorporated herein by reference.


The guide RNA sequence may be 15-100 nucleotides in length and comprise a sequence of at least 10, at least 15, or at least 20 contiguous nucleotides that is complementary to a target nucleotide sequence. The guide RNA may comprise a sequence of 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40 contiguous nucleotides that is complementary to a target nucleotide sequence. The guide RNA may be 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleotides in length.


In some embodiments, the target nucleotide sequence is a DNA sequence in a genome, e.g. a eukaryotic genome. In certain embodiments, the target nucleotide sequence is in a mammalian (e.g. a human) genome.


The compositions of this disclosure may be administered or packaged as a unit dose, for example. The term “unit dose” when used in reference to a pharmaceutical composition of the present disclosure refers to physically discrete units suitable as unitary dosage for the subject, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required diluent, i.e., a carrier or vehicle.


Treatment of a disease or disorder includes delaying the development or progression of the disease, or reducing disease severity. Treating the disease does not necessarily require curative results.


As used therein, “delaying” the development of a disease means to defer, hinder, slow, retard, stabilize, and/or postpone progression of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individuals being treated. A method that “delays” or alleviates the development of a disease, or delays the onset of the disease, is a method that reduces probability of developing one or more symptoms of the disease in a given time frame and/or reduces extent of the symptoms in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a number of subjects sufficient to give a statistically significant result.


“Development” or “progression” of a disease means initial manifestations and/or ensuing progression of the disease. Development of the disease can be detectable and assessed using standard clinical techniques as well known in the art. However, development also refers to progression that may be undetectable. For purpose of this disclosure, development or progression refers to the biological course of the symptoms. “Development” includes occurrence, recurrence, and onset.


As used herein “onset” or “occurrence” of a disease includes initial onset and/or recurrence. Conventional methods, known to those of ordinary skill in the art of medicine, can be used to administer the isolated polypeptide or pharmaceutical composition to the subject, depending upon the type of disease to be treated or the site of the disease.


Without further elaboration, it is believed that one skilled in the art can, based on the above description, utilize the present disclosure to its fullest extent. The following specific embodiments are, therefore, to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever. All publications cited herein are incorporated by reference for the purposes or subject matter referenced herein.


EXAMPLES
Example 1. Prime Editing (PE) for Installing Precise Nucleotide Changes in the Genome

The objective is to develop a transformative genome editing technology for precise and general installation of single nucleotide changes in mammalian genomes. This technology would allow investigators to study the effects of single nucleotide variations in virtually any mammalian gene, and potentially enable therapeutic interventions for correcting pathogenic point mutations in human patients.


Adoption of the clustered regularly interspaced short palindromic repeat (CRISPR) system for genome editing has revolutionized the life sciences1-3. Although gene disruption using CRISPR is now routine, the precise installation of single nucleotide edits remains a major challenge, despite being necessary for studying or correcting a large number of disease-causative mutations. Homology directed repair (HDR) is capable of achieving such edits, but suffers from low efficiency (often <5%), a requirement for donor DNA repair templates, and deleterious effects of double-stranded DNA break (DSB) formation. Recently, the Liu laboratory developed base editing, which achieves efficient single nucleotide editing without DSBs. Base editors (BEs) combine the CRISPR system with base-modifying deaminase enzymes to convert target C•G or A•T base pairs to A•T or G•C, respectively4-6. Although already widely used by researchers worldwide (>5,000 Liu lab BE constructs distributed by Addgene), current BEs enable only four of the twelve possible base pair conversions and are unable to correct small insertions or deletions. Moreover, the targeting scope of base editing is limited by the editing of non-target C or A bases adjacent to the target base (“bystander editing”) and by the requirement that a PAM sequence exist 15±2 bp from the target base. Overcoming these limitations would therefore greatly broaden the basic research and therapeutic applications of genome editing.


Here, it is proposed to develop a new precision editing approach that offers many of the benefits of base editing—namely, avoidance of double strand breaks and donor DNA repair templates—while overcoming its major limitations. To achieve this ambitious goal, it is aimed to directly install edited DNA strands at target genomic sites using target-primed reverse transcription (TPRT). In the design discussed herein, CRISPR guide RNA (gRNA) will be engineered to carry a template encoding mutagenic DNA strand synthesis, to be executed by an associated reverse transcriptase (RT) enzyme. The CRISPR nuclease (Cas9)-nicked target site DNA will serve as the primer for reverse transcription, allowing for direct incorporation of any desired nucleotide edit.


Section 1


Establish guide RNA-templated reverse transcription of mutagenic DNA strands. Prior studies have shown that, following DNA cleavage but prior to complex dissociation, Cas9 releases the non-target DNA strand to expose a free 3′ terminus. It is hypothesized that this DNA strand is accessible to extension by polymerase enzymes, and that gRNAs can be engineered through extension of their 5′ or 3′ terminus to serve as templates for DNA synthesis. In preliminary in vitro studies, it was established that nicked DNA strands within Cas9:gRNA-bound complexes can indeed prime reverse transcription using the bound gRNA as a template (RT enzyme in trans). Next, different gRNA linkers, primer binding sites, and synthesis templates will be explored to determine optimal design rules in vitro. Then, different RT enzymes, acting in trans or as fusions to Cas9, will be evaluated in vitro. Finally, engineered gRNA designs will be identified that retain efficient binding and cutting activity in cells. Successful demonstration of this aim will provide a foundation for carrying out mutagenic strand synthesis in cells.


Section 2


Establish prime editing in human cells. Based on DNA processing and repair mechanisms, it is hypothesized that mutagenic DNA strands (single stranded flaps) can be used to direct specific and efficient editing of target nucleotides. In encouraging preliminary studies, feasibility for this strategy was established by demonstrating editing with model plasmid substrates containing mutagenic flaps. Concurrent with Aim 1, repair outcomes will be further evaluated by systematically varying the mutagenic flap's length, sequence composition, target nucleotide identity, and 3′ terminus. Small 1 to 3 nucleotide insertions and deletions will also be tested. In parallel, and building from Aim 1, Cas9-RT architectures will be evaluated, including fusion proteins and non-covalent recruitment strategies. Cas9-RT architectures and extended gRNAs will be assayed for cellular editing at multiple target sites in the human genome, and will then be optimized for high efficiency. If successful, this aim would immediately establish TPRT genome editing (i.e., prime editing) for basic science applications.


Section 3


Achieve site-specific editing of pathogenic mutations in cultured human cells. The potential generality of this technology could enable editing of transversion mutations and indels that are not currently correctable by BEs. Guided by the results of Aim 1 and Aim 2, pathogenic transversion mutations will be targeted in cultured human cells, including the sickle cell disease founder mutation in beta globin (requires an A•T to T•A transversion to correct) and the most prevalent Wilson's disease mutation in ATP7B (requires a G•C to T•A transversion to correct). The correction of small insertion and deletion mutations will also be examined, including the 3-nucleotide AF508 deletion in CFTR that causes cystic fibrosis. If successful, this would lay the foundation for developing powerful therapeutic approaches that address these important human diseases.


Approach


The objective is to develop a genome editing strategy that directly installs point mutations at targeted genomic sites. In the technology development phase, efforts will focus on protein and RNA engineering to incorporate TPRT functionality into the CRISPR/Cas system. In vitro assays will be used to carefully probe the function of each step of TPRT, building from the ground up (Aim 1). The second focus area will evaluate editing outcomes in mammalian cells using a combination of model substrates and engineered CRISPR/Cas systems (Aim 2). Finally, the application phase will use the technology to correct mutations that have been intractable to genome editing by other methods (Aim 3).


The general editing design is shown in FIGS. 1A-1B. Cas9 nickases contain inactivating mutations to the HNH nuclease domain (Spy Cas9 H840A or N863A), restricting DNA cleavage to the PAM containing strand (non-target strand). Guide RNAs (gRNAs) are engineered to contain a template for reverse transcription (designs detailed on slide 5). Shown is a 5′ extension of the gRNA, but 3′ extensions can also be implemented. The Cas9 nickase is fused to a reverse transcriptase (RT) enzyme, either through the C-terminus or N-terminus. The gRNA:Cas9-RT complex targets the DNA region of interest and forms an R loop after displacing the non-target strand. Cas9 nicks the non-target DNA strand. Release of the nicked strand exposes a free 3′-OH terminus that is competent to prime reverse transcription using the extended gRNA as a template. This DNA synthesis reaction is carried out by the fused RT enzyme. The gRNA template encodes a DNA sequence that is homologous to the original DNA duplex, with the exception of the nucleotide that is targeted for editing. The product of reverse transcription is a single stranded DNA flap that encodes the desired edit. This flap, which contains a free 3′ terminus, can equilibrate with the adjacent DNA strand, resulting in a 5′ flap species. The latter species is hypothesized to serve as an efficient substrate for FEN1 (flap endonuclease 1), an enzyme that naturally excises 5′ flaps from Okazaki fragments during lagging strand DNA synthesis, and removes 5′ flaps following strand displacement synthesis that occurs during long-patch base excision repair. Ligation of the nicked DNA produces a mismatched base pair. This intermediate could either undergo reversion to the original base pair or conversion to the desired edited base pair via mismatch repair (MMR) processes. Alternatively, semiconservative DNA replication could give rise to one copy each of the reversion and edit.


Example 2—Prime Editing: Highly Versatile and Precise Search-and-Replace Genome Editing in Human Cells without Double-Stranded DNA Breaks

Current genome editing methods can disrupt, delete, or insert target genes with accompanying byproducts of double-stranded DNA breaks using programmable nucleases, and install the four transition point mutations at target loci using base editors Small insertions, small deletions, and the eight transversion point mutations, however, collectively represent most pathogenic genetic variants but cannot be corrected efficiently and without an excess of byproducts in most cell types. Described herein is prime editing, a highly versatile and precise genome editing method that directly writes new genetic information into a specified DNA site using a catalytically impaired Cas9 fused to an engineered reverse transcriptase, programmed with an engineered prime editing guide RNA (PEgRNA) that both specifies the target site and encodes the desired edit. Greater than 175 distinct edits in human cells were performed to establish that prime editing can make targeted insertions, deletions, all 12 possible types of point mutations, and combinations thereof efficiently (typically 20-60%, up to 77% in unsorted cells) and with low byproducts (typically 1-10%), without requiring double-stranded breaks or donor DNA templates. Prime editing was applied in human cells to correct the primary genetic causes of sickle cell disease (requiring an A•T-to-T•A transversion in HBB) and Tay-Sachs disease (requiring a 4-base deletion in HEXA), in both cases efficiently reverting the pathogenic genomic alleles to wild-type with minimal byproducts. Prime editing was also used to create human cell lines with these pathogenic HBB transversion and HEXA insertion mutations, to install the G127V mutation in PRNP that confers resistance to prion disease (requiring a G•C-to-T•A transversion), and to efficiently insert a His6 tag, a FLAG epitope tag, and an extended LoxP site into target loci in human cells. Prime editing offers efficiency and product purity advantages over HDR, and complementary strengths and weaknesses compared to base editing. Consistent with its search-and-replace mechanism, which requires three distinct base-pairing events, prime editing is much less prone to off-target DNA modification at known Cas9 off-target sites than Cas9. Prime editing substantially expands the scope and capabilities of genome editing, and in principle can correct ˜89% of known pathogenic human genetic variants.


The ability to make virtually any targeted change in the genome of any living cell or organism is a longstanding aspiration of the life sciences. Despite rapid advances in genome editing technologies, the majority of the >75,000 known human genetic variants associated with diseases111 cannot be corrected or installed in most therapeutically relevant cells (FIG. 38A). Programmable nucleases such as CRISPR-Cas9 make double-stranded DNA breaks (DSBs) that can disrupt genes by inducing mixtures of insertions and deletions (indels) at target sites112-114. Nucleases can also be used to delete target genes115,116, or insert exogenous genes117-119, through homology-independent processes. Double-stranded DNA breaks, however, are also associated with undesired outcomes including complex mixtures of products, translocations120, and p53 activation121,122. Moreover, the vast majority of pathogenic alleles differ from their non-pathogenic counterparts by small insertions, deletions, or base substitutions that require much more precise editing technologies to correct (FIG. 38A). Homology-directed repair (HDR) stimulated by nuclease-induced DSBs123 has been widely used to install a variety of precise DNA changes. HDR, however, relies on exogenous donor DNA repair templates, typically generates an excess of indel byproducts from end-joining repair of DSBs, and is inefficient in most therapeutically relevant cell types (T cells and some stem cells being important exceptions)124,125. While enhancing the efficiency and precision of DSB-mediated genome editing remains the focus of promising efforts126-130, these challenges necessitate the exploration of alternative precision genome editing strategies.


Base editing can efficiently install or correct the four types of transition mutations (C to T, G to A, A to G, and T to C) without requiring DSBs in a wide variety of cell types and organisms, including mammals128-131, but cannot currently achieve any of the eight transversion mutations (C to A, C to G, G to C, G to T, A to C, A to T, T to A, and T to G), such as the T•A-to-A•T mutation needed to directly correct the most common cause of sickle cell disease (HBB E6V)132. In addition, no DSB-free method has been reported to perform target deletions, such as the removal of the 4-base duplication that causes Tay-Sachs disease (HEXA 1278+TATC)133, or targeted insertions, such as the precise 3-base insertion required to directly correct the most common cause of cystic fibrosis (CFTR ΔF508)134. Targeted transversion point mutations, insertions, and deletions thus are difficult to install or correct efficiently and without excess byproducts in most cell types, even though they collectively account for most known pathogenic alleles (FIG. 38A).


Described herein is the development of prime editing, a new “search-and-replace” genome editing technology that mediates targeted insertions, deletions, and all 12 possible base-to-base conversions at targeted loci in human cells without requiring double-stranded DNA breaks, or donor DNA templates. Prime editors, initially exemplified by PE1, use a reverse transcriptase fused to a programmable nickase and a prime editing extended guide RNA (PEgRNA) to directly copy genetic information from the extension on the PEgRNA into the target genomic locus. A second-generation prime editor (PE2) uses an engineered reverse transcriptase to substantially increase editing efficiencies with minimal (typically <2%) indel formation, while a third-generation PE3 system adds a second guide RNA to nick the non-edited strand, thereby favoring replacement of the non-edited strand and further increasing editing efficiency, typically, to about 20-50% in human cells with about 1-10% indel formation. PE3 offers far fewer byproducts and higher or similar efficiency compared to optimized Cas9 nuclease-initiated HDR, and offers complementary strengths and weaknesses compared to current-generation base editors.


PE3 was applied at genomic loci in human HEK293T cells to achieve efficient conversion of HBB E6V to wild-type HBB, deletion of the inserted TATC to restore HEXA 1278+TATC to wild-type HEXA, installation in PRNP of the G127V mutation that confers resistance to prion disease135 (requiring a G•C-to-T•A transversion), and targeted insertion of a His6 tag (18 bp), FLAG epitope tag (24 bp), and extended LoxP site for Cre-mediated recombination (44 bp). Prime editing was also successful in three other human cell lines, as well as in post-mitotic primary mouse cortical neurons, with varying efficiencies. Due to a high degree of flexibility in the distance between the initial nick and location of the edit, prime editing is not substantially constrained by the PAM requirement of Cas9 and in principle can target the vast majority of genomic loci. Off-target prime editing is much rarer than off-target Cas9 editing at known Cas9 off-target loci, likely due to the requirement of three distinct DNA base pairing events in order for productive prime editing to take place. By enabling precise targeted insertions, deletions, and all 12 possible classes of point mutations at a wide variety of genomic loci without the need for DSBs or donor DNA templates, prime editing has the potential to advance the study and correction of many gene variants.


Results


Strategy for Transferring Information from an Extended Guide RNA into a Target DNA Locus


Cas9 targets DNA using a guide RNA containing a spacer sequence that hybridizes to the target DNA site112-114,136,137. The aim was to engineer guide RNAs to both specify the DNA target as in natural CRISPR systems138,139, and also to contain new genetic information that replaces the corresponding DNA nucleotides at the target locus. The direct transfer of genetic information from an extended guide RNA into a specified DNA site, followed by replacement of the original unedited DNA, in principle could provide a general means of installing targeted DNA sequence changes in living cells, without dependence on DSBs or donor DNA templates. To achieve this direct information transfer, the aim was to use genomic DNA, nicked at the target site to expose a 3′-hydroxyl group, to prime the reverse transcription of the genetic information from an extension on the engineered guide RNA (hereafter referred to as the prime editing guide RNA, or PEgRNA) directly into the target site (FIG. 38A).


These initial steps of nicking and reverse transcription, which resemble mechanisms used by some natural mobile genetic elements140, result in a branched intermediate with two redundant single-stranded DNA flaps on one strand: a 5′ flap that contains the unedited DNA sequence, and a 3′ flap that contains the edited sequence copied from the PEgRNA (FIG. 38B). To achieve a successful edit, this branched intermediate must be resolved so that the edited 3′ flap replaces the unedited 5′ flap. While hybridization of the 5′ flap with the unedited strand is likely to be thermodynamically favored since the edited 3′ flap can make fewer base pairs with the unedited strand, 5′ flaps are the preferred substrate for structure-specific endonucleases such as FEN1141, which excises 5′ flaps generated during lagging-strand DNA synthesis and long-patch base excision repair. It was reasoned that preferential 5′ flap excision and 3′ flap ligation could drive the incorporation of the edited DNA strand, creating heteroduplex DNA containing one edited strand and one unedited strand (FIG. 38B).


Permanent installation of the edit could arise from subsequent DNA repair that resolves the mismatch between the two DNA strands in a manner that copies the information in the edited strand to the complementary DNA strand (FIG. 38C). Based on a similar strategy developed to maximize the efficiency of DNA base editing131-133, it was envisioned that nicking the non-edited DNA strand, far enough from the site of the initial nick to minimize double-strand break formation, might bias DNA repair to preferentially replace the non-edited strand.


Validation of Prime Editing Steps In Vitro and in Yeast Cells


Following cleavage of the PAM-containing DNA strand by the RuvC nuclease domain of Cas9, the PAM-distal fragment of this strand can dissociate from otherwise stable Cas9:sgRNA:DNA complexes143. It was hypothesized that the 3′ end of this liberated strand might be sufficiently accessible to prime DNA polymerization. Guide RNA engineering efforts144-146 and crystal structures of Cas9:sgRNA:DNA complexes147-149 suggest that the 5′ and 3′ termini of the sgRNA can be extended without abolishing Cas9:sgRNA activity. PEgRNAs were designed by extending sgRNAs to include two critical components: a primer binding site (PBS) that allows the 3′ end of the nicked DNA strand to hybridize to the PEgRNA, and a reverse transcriptase (RT) template containing the desired edit that would be directly copied into the genomic DNA site as the 3′ end of the nicked DNA strand is extended across the RNA template by a polymerase (FIG. 38C).


These hypotheses were tested in vitro using purified S. pyogenes Cas9 protein. A series of PEgRNA candidates were constructed by extending sgRNAs on either terminus with a PBS sequence (5 to 6 nucleotides, nt) and an RT template (7 to 22 nt). It was confirmed that 5′-extended PEgRNAs direct Cas9 binding to target DNA, and that both 5′-extended PEgRNAs and 3′-extended PEgRNAs support Cas9-mediated target nicking in vitro and DNA cleavage activities in mammalian cells (FIGS. 44A-44C). These candidate PEgRNA designs were tested using pre-nicked 5′-Cy5-labeled dsDNA substrates, catalytically dead Cas9 (dCas9), and a commercial variant of Moloney murine leukemia virus (M-MLV) reverse transcriptase (FIG. 44D). When all components were present, efficient conversion of the fluorescently labeled DNA strand into longer DNA products with gel mobilities, consistent with reverse transcription along the RT template, (FIG. 38D, FIGS. 44D-44E) was observed. Products of desired length were formed with either 5′-extended or 3′-extended PEgRNAs (FIGS. 38D-38E). Omission of dCas9 led to nick translation products derived from reverse transcriptase-mediated DNA polymerization on the DNA template, with no PEgRNA information transfer (FIG. 38D). No DNA polymerization products were observed when the PEgRNA was replaced by a conventional sgRNA, confirming the necessity of the PBS and RT template components of the PEgRNA (FIG. 38D). These results demonstrate that Cas9-mediated DNA melting exposes a single-stranded R-loop that, if nicked, is competent to prime reverse transcription from either a 5′-extended or 3′-extended PEgRNA.


Next, non-nicked dsDNA substrates were tested with a Cas9 nickase (H840A mutant) that exclusively nicks the PAM-containing strand112. In these reactions, 5′-extended PEgRNAs generated reverse transcription products inefficiently, possibly due to impaired Cas9 nickase activity (FIG. 44F). However, 3′-extended PEgRNAs enabled robust Cas9 nicking and efficient reverse transcription (FIG. 38E). The use of 3′-extended PEgRNAs generated only a single apparent product, despite the potential, in principle, for reverse transcription to terminate anywhere within the remainder of the PEgRNA. DNA sequencing of the products of reactions with Cas9 nickase, RT, and 3′-extended PEgRNAs revealed that the complete RT template sequence was reverse transcribed into the DNA substrate (FIG. 44G). These experiments established that 3′-extended PEgRNAs can template the reverse transcription of new DNA strands while retaining the ability to direct Cas9 nickase activity.


To evaluate the eukaryotic cell DNA repair outcomes of 3′ flaps produced by PEgRNA-programmed reverse transcription in vitro, DNA nicking and reverse transcription using PEgRNAs, Cas9 nickase, and RT in vitro on reporter plasmid substrates were performed, and the reaction products were then transformed into yeast (S. cerevisiae) cells (FIG. 45A). Encouragingly, when plasmids were edited in vitro with 3′-extended PEgRNAs encoding a T•A-to-A•T transversion that corrects the premature stop codon, 37% of yeast transformants expressed both GFP and mCherry proteins (FIG. 38F, FIG. 45C). Consistent with the results in FIG. 38E and FIG. 44F, editing reactions carried out in vitro with 5′-extended PEgRNAs yielded fewer GFP and mCherry double-positive colonies (9%) than those with 3′-extended PEgRNAs (FIG. 38F and FIG. 45D). Productive editing was also observed using 3′-extended PEgRNAs that insert a single nucleotide (15% double-positive transformants) or delete a single nucleotide (29% double-positive transformants) to correct frameshift mutations (FIG. 38F and FIGS. 45E-45F). DNA sequencing of edited plasmids recovered from double-positive yeast colonies confirmed that the encoded transversion edit occurred at the desired sequence position (FIG. 45G). These results demonstrate that DNA repair in eukaryotic cells can resolve 3′ DNA flaps arising from prime editing to incorporate precise DNA edits including transversions, insertions, and deletions.


Design of Prime Editor 1 (PE1)


Encouraged by the results in vitro and in yeast, a prime editing system with a minimum number of components capable of editing genomic DNA in mammalian cells was sought for development. It was hypothesized that 3′-extended PEgRNAs (hereafter referred to simply as PEgRNAs, FIG. 39A) and direct fusions of Cas9 H840A to reverse transcriptase via a flexible linker may constitute a functional two-component prime editing system. HEK293T (immortalized human embryonic kidney) cells were transfected with one plasmid encoding a fusion of wild-type M-MLV reverse transcriptase to either terminus of Cas9 H840A nickase as well as a second plasmid encoding a PEgRNA. Initial attempts led to no detectable T•A-to-A•T conversion at the HEK3 target locus.


Extension of the PBS in the PEgRNA to 8-15 bases (FIG. 39A), however, led to detectable T•A-to-A•T editing at the HEK3 target site (FIG. 39B), with higher efficiencies for prime editor constructs in which the RT was fused to the C-terminus of Cas9 nickase (3.7% maximal T•A-to-A•T conversion with PBS lengths ranging from 8-15 nt) compared to N-terminal RT-Cas9 nickase fusions (1.3% maximal T•A-to-A•T conversion) (FIG. 39B; all mammalian cell data herein reports values for the entire treated cell population, without selection or sorting, unless otherwise specified). These results suggest that wild-type M-MLV RT fused to Cas9 requires longer PBS sequences for genome editing in human cells compared to what is required in vitro using the commercial variant of M-MLV RT supplied in trans. This first-generation wild-type M-MLV reverse transcriptase fused to the C-terminus of Cas9 H840A nickase was designated as PE1.


The ability of PE1 to precisely introduce transversion point mutations at four additional genomic target sites specified by the PEgRNA (FIG. 39C) was tested. Similar to editing at the HEK3 locus, efficiency at these genomic sites was dependent on PBS length, with maximal editing efficiencies ranging from 0.7-5.5% (FIG. 39C). Indels from PE1 were low, averaging 0.2±0.1% for the five sites under conditions that maximized each site's editing efficiency (FIG. 46A). PE1 was also able to install targeted insertions and deletions, exemplified by a single-nucleotide deletion (4.0% efficiency), a single-nucleotide insertion (9.7%), and a three-nucleotide insertion (17%) at the HEK3 locus (FIG. 39C). These results establish the ability of PE1 to directly install targeted transversions, insertions, and deletions without requiring double-stranded DNA breaks or DNA templates.


Design of Prime Editor 2 (PE2)


While PE1 can install a variety of edits at several loci in HEK293T cells, editing efficiencies were generally low (typically ≤5%) (FIG. 39C). It was hypothesized that engineering the reverse transcriptase in PE1 might improve the efficiency of DNA synthesis within the unique conformational constraints of the prime editing complex, resulting in higher genome editing yields. M-MLV RT mutations have been previously reported that increase enzyme thermostability150,151, processivity150, and DNA:RNA heteroduplex substrate affinity152, and that inactivate RNaseH activity153. 19 PE1 variants were constructed containing a variety of reverse transcriptase mutations to evaluate their prime editing efficiency in human cells.


First, a series of M-MLV RT variants that previously emerged from laboratory evolution for their ability to support reverse transcription at elevated temperatures150 were investigated. Successive introduction of three of these amino acid substitutions (D200N, L603W, and T330P) into M-MLV RT, hereafter referred to as M3, led to a 6.8-fold average increase in transversion and insertion editing efficiency across five genomic loci in HEK293T cells compared to that of PE1 (FIGS. 47A-47S).


Next, in combination with M3, additional reverse transcriptase mutations that were previously shown to enhance binding to template:PBS complex, enzyme processivity, and thermostability152, were tested. Among the 14 additional mutants analyzed, a variant with T306K and W313F substitutions, in addition to the M3 mutations, improved editing efficiency an additional 1.3-fold to 3.0-fold compared to M3 for six transversion or insertion edits across five genomic sites in human cells (FIGS. 47A-47S). This pentamutant of M-MLV reverse transcriptase incorporated into the PE1 architecture (Cas9 H840A—M-MLV RT (D200N L603W T330P T306K W313F)) is hereafter referred to as PE2.


PE2 installs single-nucleotide transversion, insertion, and deletion mutations with substantially higher efficiency than PE1 (FIG. 39C), and is compatible with shorter PBS PEgRNA sequences (FIG. 39C), consistent with an enhanced ability to productively engage transient genomic DNA:PBS complexes. On average, PE2 led to a 1.6- to 5.1-fold improvement in prime editing point mutation efficiency over PE1 (FIG. 39C), and in some cases dramatically improved editing yields up to 46-fold (FIG. 47F and FIG. 47I). PE2 also effected targeted insertions and deletions more efficiently than PE1, achieving the targeted insertion of the 24-bp FLAG epitope tag at the HEK3 locus with 4.5% efficiency, a 15-fold improvement over the efficiency of installing this insertion with PE1 (FIG. 47D), and mediated a 1-bp deletion in HEK3 with 8.6% efficiency, 2.1-fold higher than that of PE1 (FIG. 39C). These results establish PE2 as a more efficient prime editor than PE1.


Optimization of PEgRNA Features


The relationship between PEgRNA architecture and prime editing efficiency was systematically probed at five genomic loci in HEK293T cells with PE2 (FIG. 39C). In general, priming sites with lower GC content required longer PBS sequences (EMX1 and RNF2, containing 40% and 30% GC content, respectively, in the first 10 nt upstream of the nick), whereas those with greater GC content supported prime editing with shorter PBS sequences (HEK4 and FANCF, containing 80% and 60% GC content, respectively, in the first 10 nt upstream of the nick) (FIG. 39C), consistent with the energetic requirements for hybridization of the nicked DNA strand to the PEgRNA PBS. No PBS length or GC content level was strictly predictive of prime editing efficiency, and other factors such as secondary structure in the DNA primer or PEgRNA extension may also influence editing activity. It is recommended to start with a PBS length of ˜13 nt for a typical target sequence, and exploring different PBS lengths if the sequence deviates from ˜40-60% GC content. When necessary, optimal PBS sequences should be determined empirically.


Next, the performance determinants of the RT template portion of the PEgRNA were studied. PEgRNAs with RT templates ranging from 10-20 nt in length were systemically evaluated at five genomic target sites using PE2 (FIG. 39D) and with longer RT templates as long as 31 nt at three genomic sites (FIGS. 48A-48C). As with PBS length, RT template length also could be varied to maximize prime editing efficiency, although in general many RT template lengths ≥10 nt long support more efficient prime editing (FIG. 39D). Since some target sites preferred longer RT templates (>15 nt) to achieve higher editing efficiencies (FANCF, EMX1), while other loci preferred short RT templates (HEK3, HEK4) (FIG. 39D), it is recommend both short and long RT templates be tested when optimizing a PEgRNA, starting with ˜10-16 nt.


Importantly, RT templates that place a C as the nucleotide adjacent to the terminal hairpin of the sgRNA scaffold generally resulted in lower editing efficiency compared to other PEgRNAs with RT templates of similar length (FIGS. 48A-48C). Based on the structure of sgRNAs bound to Cas9148,149, it was speculated that the presence of a C as the first nucleotide of the 3′ extension of a canonical sgRNA can disrupt the sgRNA scaffold fold by pairing with G81, a nucleotide that natively forms a pi stack with Tyr 1356 in Cas9 and a non-canonical base pair with sgRNA A68. Since many RT template lengths support prime editing, it is recommended to choose PEgRNAs in which the first base of the 3′ extension (the last reverse-transcribed base of the RT template) is not C.


Design of Prime Editor 3 Systems (PE3 and PE3b)


While PE2 can transfer genetic information from the PEgRNA to the target locus more efficiently than PE1, the manner in which the cell resolves the resulting heteroduplex DNA created by one edited strand and one unedited strand determines if the edit is durable. A previous development of base editing faced a similar challenge since the initial product of cytosine or adenine deamination is heteroduplex DNA containing one edited and one non-edited strand. To increase the efficiency of base editing, a Cas9 D10A nickase was used to introduce a nick into the non-edited strand and to direct DNA repair to that strand, using the edited strand as a template129,130,142. To exploit this principle to enhance prime editing efficiencies, a similar strategy of nicking the non-edited strand using the Cas9 H840A nickase already present in PE2 and a simple sgRNA to induce preferential replacement of the non-edited strand by the cell (FIG. 40A) was tested. Since the edited DNA strand was also nicked to initiate prime editing, a variety of sgRNA-programmed nick locations were tested on the non-edited strand to minimize the production of double-stranded DNA breaks that lead to indels.


This PE3 strategy was first tested at five genomic sites in HEK293T cells by screening sgRNAs that induce nicks located 14 to 116 bases from the site of the PEgRNA-induced nick, either 5′ or 3′ of the PAM. In four of the five sites tested, nicking the non-edited strand increased the amount of indel-free prime editing products compared to the PE2 system by 1.5- to 4.2-fold, to as high as 55% (FIG. 40B). While the optimal nicking position varied depending on the genomic site, nicks positioned 3′ of the PAM (positive distances in FIG. 40B) approximately 40-90 bp from the PEgRNA-induced nick generally produced favorable increases in prime editing efficiency (averaging 41%) without excess indel formation (6.8% average indels for the sgRNA resulting in the highest editing efficiency for each of the five sites tested) (FIG. 40B). As expected, at some sites, placement of the non-edited strand nick within 40 bp of the PEgRNA-induced nick led to large increases in indel formation up to 22% (FIG. 40B), presumably due to the formation of a double-strand break from nicking both strands close together. At other sites, however, nicking as close as 14 bp away from the PEgRNA-induced nick produced only 5% indels (FIG. 40B), suggesting that locus-dependent factors control conversion of proximal dual nicks into double-strand DNA breaks. At one tested site (HEK4), complementary strand nicks either provided no benefit or led to indel levels that surpassed editing efficiency (up to 26%), even when placed at distances >70 bp from the PEgRNA-induced nick, consistent with an unusual propensity of the edited strand at that site to be nicked by the cell, or to be ligated inefficiently. It is recommend to start with non-edited strand nicks approximately 50 bp from the PEgRNA-mediated nick, and to test alternative nick locations if indel frequencies exceed acceptable levels.


This model for how complementary strand nicking improved prime editing efficiency (FIG. 40A) predicted that nicking the non-edited strand only after edited strand flap resolution could minimize the presence of concurrent nicks, decreasing the frequency of double-strand breaks that go on to form indels. To achieve temporal control over non-edited strand nicking, sgRNAs with spacer sequences that match the edited strand, but not the original allele, were designed. Using this strategy, referred to hereafter as PE3b, mismatches between the spacer and the unedited allele should disfavor nicking by the sgRNA until after the editing event on the PAM strand takes place. This PE3b approach was tested with five different edits at three genomic sites in HEK293T cells and compared outcomes to those achieved with PE2 and PE3 systems. In all cases, PE3b was associated with substantially lower levels of indels compared to PE3 (3.5- to 30-fold, averaging 12-fold lower indels, or 0.85%), without any evident decrease in overall editing efficiency compared to PE3 (FIG. 40C). Therefore, when the edit lay within a second protospacer, the PE3b system could decrease indels while still improving editing efficiency compared to PE2, often to levels similar to those of PE3 (FIG. 40C).


Together, these findings established that PE3 systems (Cas9 nickase—optimized reverse transcriptase+PEgRNA+sgRNA) improved editing efficiencies ˜3-fold compared with PE2 (FIGS. 40B-40C). PE3 was accompanied by wider ranges of indels than PE2, as expected given the additional nicking activity of PE3. The use of PE3 is recommended when prioritizing prime editing efficiency. When minimization of indels is critical, PE2 offers ˜10-fold lower indel frequencies. When it is possible to use a sgRNA that recognizes the installed edit to nick the non-edited strand, the PE3b system can achieve PE3-like editing levels while greatly reducing indel formation.


To demonstrate the targeting scope and versatility of prime editing with PE3, the installation of all possible single nucleotide substitutions across the +1 to +8 positions (counting the first base 3′ of the PEgRNA-induced nick as position +1) of the HEK3 target site using PE3 and PEgRNAs with 10-nucleotide RT templates (FIG. 41A) was explored. Collectively, these 24 distinct edits cover all four transition mutations and all eight transversion mutations, and proceed with editing efficiencies (containing no indels) averaging 33±7.9% (ranging between 14% and 48%), with an average of 7.5±1.8% indels.


Importantly, long-distance RT templates could also give rise to efficient prime editing with PE3. For example, using PE3 with a 34-nt RT template, point mutations were installed at positions +12, +14, +17, +20, +23, +24, +26, +30, and +33 (12 to 33 bases from the PEgRNA-induced nick) in the HEK3 locus with an average of 36±8.7% efficiency and 8.6±2.0% indels (FIG. 41B). Although edits beyond the +10 position at other loci were not attempted, other RT templates ≥30 nt at three alternative sites also support efficient editing (FIGS. 48A-C). The viability of long RT templates enabled efficient prime editing for dozens of nucleotides from the initial nick site. Since an NGG PAM on either DNA strand occurs on average every ˜8 bp, far less than maximum distances between the edit and the PAM that support efficient prime editing, prime editing is not substantially constrained by the availability of a nearby PAM sequence, in contrast with other precision genome editing methods125,142,154. Given the presumed relationship between RNA secondary structure and prime editing efficiency, when designing PEgRNAs for long-range edits it is prudent to test RT templates of various lengths and, if necessary, sequence compositions (e.g., synonymous codons) to optimize editing efficiency.


To further test the scope and limitations of the PE3 system for introducing transition and transversion point mutations, 72 additional edits covering all 12 possible types of point mutations across six additional genomic target sites (FIG. 41C-41H) were tested. Overall, indel-free editing efficiency averaged 25±14%, while indel formation averaged 8.3±7.5%. Since the PEgRNA RT template included the PAM sequence, prime editing could induce changes to the PAM sequence. In these cases, higher editing efficiency (averaging 39±9.7%) and lower indel generation (averaging 5.0±2.9%) were observed (FIGS. 41A-41K, point mutations at positions +5 or +6). This increase in efficiency and decrease in indel formation for PAM edits may arise from the inability of the Cas9 nickase to re-bind and nick the edited strand prior to the repair of the complementary strand. Since prime editing supports combination edits with no apparent loss of editing efficiency, editing the PAM, in addition to other desired changes, when possible, is recommended.


Next, 14 targeted small insertions and 14 targeted small deletions at seven genomic sites using PE3 (FIG. 41I) were performed. Targeted 1-bp insertions proceeded with an average efficiency of 32±9.8%, while 3-bp insertions were installed with an average efficiency of 39±16%. Targeted 1-bp and 3-bp deletions were also efficient, proceeding with an average yield of 29±14% and 32±11%, respectively. Indel generation (beyond the targeted insertion or deletion) averaged 6.8±5.4%. Since insertions and deletions introduced between positions +1 and +6 alter the position or the structure of the PAM, it was speculated that insertion and deletion edits in this range are typically more efficient due to the inability of Cas9 nickase to re-bind and nick the edited DNA strand prior to repair of the complementary strand, similar to point mutations that edit the PAM.


PE3 was also tested for its ability to mediate larger precise deletions of 5 bp to 80 bp at the HEK3 site (FIG. 41J). Very high editing efficiencies (52 to 78%) were observed for 5-, 10-, and 15-bp deletions when using a 13-nt PBS and an RT template that contained 29, 24, or 19 bp of homology to the target locus, respectively. Using a 26-nt RT template supported a larger deletion of 25 bp with 72±4.2% efficiency, while a 20-nt RT template enabled an 80-bp deletion with an efficiency of 52±3.8%. These targeted deletions were accompanied by indel frequencies averaging 11±4.8% (FIG. 41J).


Finally, the ability of PE3 to mediate 12 combinations of multiple edits at the same target locus consisting of insertions and deletions, insertions and point mutations, deletions and point mutations, or two point mutations across three genomic sites was tested. These combination edits were very efficient, averaging 55% of the target edit with 6.4% indels (FIG. 41K), and demonstrating the ability of prime editing to make combinations of precision insertions, deletions, and point mutations at individual target sites with high efficiency and low indel frequencies.


Together, the examples in FIGS. 41A-41K represent 156 distinct transition, transversion, insertion, deletion, and combination edits across seven human genomic loci. These findings establish the versatility, precision, and targeting flexibility of prime editing.


Prime Editing Compared with Base Editing


Current-generation cytidine base editors (CBEs) and adenine base editors (ABEs) can install C•G-to-T•A transition mutations and A•T-to-G•C transition mutations with high efficiency and low indels129,130,142. The application of base editing can be limited by the presence of multiple cytidine or adenine bases within the base editing activity window (typically ˜5-bp wide), which gives rise to unwanted bystander edits129,130,142,155, or by the absence of a PAM positioned approximately 15±2 nt from the target nucleotide142,156. It was anticipated that prime editing could be particularly useful for precise installation of transitions mutations without bystander edits, or when the lack of suitably positioned PAMs precludes favorable positioning the target nucleotide within the CBE or ABE activity window.


Prime editing and cytosine base editing was compared by editing three genomic loci that contain multiple target cytidines in the canonical base editing window (protospacer positions 4-8, counting the PAM as positions 21-23) using optimized CBEs157 without nickase activity (BE2max) or with nickase activity (BE4max), or using the analogous PE2 and PE3 prime editing systems. Among the nine total target cytosines within the base editing windows of the three sites, BE4max yielded 2.2-fold higher average total C•G-to-T•A conversion than PE3 for bases in the center of the base editing window (protospacer positions 5-7, FIG. 42A). Likewise, non-nicking BE2max outperformed PE2 by 1.4-fold on average at these well-positioned bases (FIG. 42A). However, PE3 outperformed BE4max by 2.7-fold, and PE2 outperformed BE2max by 2.0-fold, for cytosines beyond the center of the base editing window (average editing of 40±17% for PE3 vs. 15±18% for BE4max, and 22±11% for PE2 vs. 11±13% for BE2max). Overall, indel frequencies for PE2 were very low (averaging 0.86±0.47%), and for PE3 were similar to or modestly higher than that of BE4max (BE4max range: 2.5% to 14%; PE3 range: 2.5% to 21%) (FIG. 42B).


When comparing the efficiency of base editing to prime editing for installation of precise C•G-to-T•A edits (without any bystander editing), the efficiency of prime editing greatly exceeded that of base editing at the above sites, which like most genomic DNA sites, contain multiple cytosines within the ˜5-bp base editing window (FIG. 42C). At these sites, such as EMX1, which contains cytosines at protospacer positions C5, C6, and C7, BE4max generated few products containing only the single target base pair conversion with no bystander edits. In contrast, prime editing at this site could be used to selectively install a C•G-to-T•A edit at any position or combination of positions (C5, C6, C7, C5+C6, C6+C7, C5+C7, or C5+C6+C7) (FIG. 42C). All precise one-base or two-base edits (that is, edits that do not modify any other nearby bases) were much more efficient with PE3 or PE2 than with BE4max or BE2, respectively, while the three-base C•G-to-T•A edit was more efficient with BE4max (FIG. 42C), reflecting the propensity of base editors to edit all target bases within the activity window. Taken together, these results demonstrate that cytosine base editors can result in higher levels of editing at optimally positioned target bases than PE2 or PE3, but prime editing can outperform base editing at non-optimally positioned target bases, and can edit with much higher precision with multiple editable bases.


A•T-to-G•C editing was compared at two genomic loci by an optimized non-nicking ABE (ABEmax152 with a dCas9 instead of a Cas9 nickase, hereafter referred to as ABEdmax) versus PE2, and by the optimized nicking adenine base editor ABEmax versus PE3. At a site that contains two target adenines in the base editing window (HEK3), ABEs were more efficient than PE2 or PE3 for conversion of A5, but PE3 was more efficient for conversion of A8, which lies at the edge of the ABEmax editing window (FIG. 42D). When comparing the efficiency of precision edits in which only a single adenine is converted, PE3 outperformed ABEmax at both A5 and A8 (FIG. 42E). Overall, ABEs produced far fewer indels at HEK3 than prime editors (0.19±0.02% for ABEdmax vs. 1.5±0.46% for PE2, and 0.53±0.16% for ABEmax vs. 11±2.3% for PE3, FIG. 42F). At FANCF, in which only a single A is present within the base editing window, ABE2 and ABEmax outperformed their prime editing counterparts in total target base pair conversion by 1.8- to 2.9-fold, with virtually all edited products from both base editing and prime editing containing only the precise edit (FIGS. 42D-42E). As with the HEK3 site, ABEs produced far fewer indels at the FANCF site (FIG. 42F).


Collectively, these results indicate that base editing and prime editing offer complementary strengths and weaknesses for making targeted transition mutations. For cases in which a single target nucleotide is present within the base editing window, or when bystander edits are acceptable, current base editors are typically more efficient and result in fewer indels than prime editors. When multiple cytosines or adenines are present and bystander edits are undesirable, or when target bases are poorly positioned for base editing relative to available PAMs, prime editors offer substantial advantages.


Off-Target Prime Editing


To result in productive editing, prime editing requires target locus:PEgRNA spacer complementary for the Cas9 domain to bind, target locus:PEgRNA PBS complementarity for PEgRNA-primed reverse transcription to initiate, and target locus:reverse transcriptase product complementarity for flap resolution. It was hypothesized that these three distinct DNA hybridization requirements may minimize off-target prime editing compared to that of other genome editing methods. To test this possibility, HEK293T cells were treated with PE3 or PE2 and 16 total PEgRNAs designed to target four on-target genomic loci, with Cas9 and the four corresponding sgRNAs targeting the same protospacers, or with Cas9 and the same 16 PEgRNAs. These four target loci were chosen because each has at least four well-characterized off-target sites for which Cas9 and the corresponding on-target sgRNA in HEK293T cells is known to cause substantial off-target DNA modification118,159. Following treatment, the four on-target loci and the top four known Cas9 off-target sites for each on-target spacer, were sequenced, for a total of 16 off-target sites (Table 1).


Consistent with previous studies', Cas9 and the four target sgRNAs modified all 16 of the previously reported off-target loci (FIG. 42G). Cas9 off-target modification efficiency among the four off-target sites for the HEK3 target locus averaged 16%. Cas9 and the sgRNA targeting HEK4 resulted in an average of 60% modification of the four tested known off-target sites. Likewise, off-target sites for EMX1 and FANCF were modified by Cas9:sgRNA at an average frequency of 48% and 4.3%, respectively (FIG. 42G). It was noted that PEgRNAs with Cas9 nuclease modified on-target sites at similar (1- to 1.5-fold lower) efficiency on average compared to sgRNAs, while PEgRNAs with Cas9 nuclease modified off-target sites at ˜4-fold lower average efficiency than sgRNAs.


Strikingly, PE3 or PE2 with the same 16 tested PEgRNAs containing these four target spacers resulted in much lower off-target editing (FIG. 42H). Of the 16 sites known to undergo off-target editing by Cas9+sgRNA, PE3+PEgRNAs or PE2+PEgRNAs resulted in detectable off-target prime editing at only 3 of 16 off-target sites, with only 1 of 16 showing off-target editing efficiency ≥1% (FIG. 42H). Average off-target prime editing for the PEgRNAs targeting HEK3, HEK4, EMX1, and FANCF at these 16 known Cas9 off-target sites was <0.1%, <2.2±5.2%, <0.1%, and <0.13±0.11%, respectively (FIG. 42H). Notably, at the HEK4 off-target 3 site that Cas9+PEgRNA1 edits with 97% efficiency, PE2+PEgRNA1 results in only 0.7% off-target editing despite sharing the same spacer sequence, demonstrating how the two additional DNA hybridization events required for prime editing compared to Cas9 editing can greatly reduce off-target editing. Taken together, these results suggest that PE3 and PEgRNAs induce much lower off-target DNA editing in human cells than Cas9 and sgRNAs that target the same protospacers.


Reverse transcription of 3′-extended PEgRNAs in principle can proceed into the guide RNA scaffold. If the resulting 3′ flap, despite a lack of complementary at its 3′ end with the unedited DNA strand, is incorporated into the target locus, the outcome is insertion of PEgRNA scaffold nucleotides that contributes to indel frequency. We analyzed sequencing data from 66 PE3-mediated editing experiments at four loci in HEK293T cells and observed PEgRNA scaffold insertion at a low frequency, averaging 1.7±1.5% total insertion of any number of PEgRNA scaffold nucleotides (FIGS. 56A-56D). It is speculated that inaccessibility of the guide RNA scaffold to the reverse transcriptase due to Cas9 domain binding, as well as cellular excision during flap resolution of the mismatched 3′ end of the 3′ flap that results from PEgRNA scaffold reverse transcription, minimizes products that incorporate PEgRNA scaffold nucleotides. While such events are rare, future efforts to engineer PEgRNAs or prime editor proteins that minimize PEgRNA scaffold incorporation may further decrease indel frequencies.


Deaminases in some base editors can act in a Cas9-independent manner, resulting in low-level but widespread off-target DNA editing among first-generation CBEs (but not ABEs)160-162 and off-target RNA editing among first-generation CBEs and ABEs163-165, although newer CBE and ABE variants with engineered deaminases greatly reduce Cas9-independent off-target DNA and RNA editing163-165. Prime editors lack base-modification enzymes such as deaminases, and therefore have no inherent ability to modify DNA or RNA bases in a Cas9-independent manner.


While the reverse transcriptase domain in prime editors in principle could process properly primed RNA or DNA templates in cells, it was noted that retrotransposons such as those in the LINE-1 family166, endogenous retroviruses167,168, and human telomerase all provided active endogenous human reverse transcriptases. Their natural presence in human cells suggests that reverse transcriptase activity itself is not substantially toxic. Indeed, no PE3-dependent differences were observed in HEK293T cell viability compared to that of controls expressing dCas9, Cas9 H840A nickase, or PE2 with R110S+K103L (PE2-dRT) mutations that inactivate the reverse transcriptase169 (FIGS. 49A-49B).


The above data and analyses notwithstanding, additional studies are needed to assess off-target prime editing in an unbiased, genome-wide manner, as well as to characterize the extent to which the reverse transcriptase variants in prime editors, or prime editing intermediates, may affect cells.


Prime Editing Pathogenic Transversion, Insertion, and Deletion Mutations in Human Cells


The ability of PE3 to directly install or correct in human cells transversion, small insertion, and small deletion mutations that cause genetic diseases, was tested. Sickle cell disease is most commonly caused by an A•T-to-T•A transversion mutation in HBB, resulting in the mutation of Glu6→Val in beta-globin. Treatment of hematopoietic stem cells ex vivo with Cas9 nuclease and a donor DNA template for HDR, followed by enrichment of edited cells, transplantation, and engraftment is a promising potential strategy for the treatment of sickle-cell disease170. However, this approach still generates many indel-containing byproducts in addition to the correctly edited HBB allele170-171. While base editors generally produce far fewer indels, they cannot currently make the T•A-to-A•T transversion mutation needed to directly restore the normal sequence of HBB.


PE3 was used to install the HBB E6V mutation in HEK293T cells with 44% efficiency and 4.8% indels (FIG. 43A. From the mixture of PE3-treated cells, we isolated six HEK293T cell lines that are homozygous (triploid) for the HBB E6V allele (FIGS. 53A-53D), demonstrating the ability of prime editing to generate human cell lines with pathogenic mutations. To correct the HBB E6V allele to wild-type HBB, we treated homozygous HBB E6V HEK293T cells with PE3 and a PEgRNA programmed to directly revert the HBB E6V mutation to wild-type HBB. In total, 14 PEgRNA designs were tested. After three days, DNA sequencing revealed that all 14 PEgRNAs when combined with PE3 gave efficient correction of HBB E6V to wild-type HBB (≥26% wild-type HBB without indels), and indel levels averaging 2.8±0.70% (FIG. 50A). The best PEgRNA resulted in 52% correction of HBB E6V to wild-type with 2.4% indels (FIG. 43A). Introduction of a silent mutation that modifies the PAM recognized by the PEgRNA modestly improved editing efficiency and product purity, to 58% correction with 1.4% indels (FIG. 43A). These results establish that prime editing can install and correct a pathogenic transversion point mutation in a human cell line with high efficiency and minimal byproducts.


Tay-Sachs disease is most often caused by a 4-bp insertion into the HEXA gene (HEXA 1278+TATC)136. PE3 was used to install this 4-bp insertion into HEK293T cells with 31% efficiency and 0.8% indels (FIG. 43B), and isolated two HEK293T cell lines that are homozygous for the HEXA 1278+TATC allele (FIGS. 53A-53D). These cells were used to test 43 PEgRNAs and three nicking sgRNAs with PE3 or PE3b systems for correction of the pathogenic insertion in HEXA (FIG. 50B), either by perfect reversion to the wild-type allele or by a shifted 4-bp deletion that disrupts the PAM and installs a silent mutation. Nineteen of the 43 PEgRNAs tested resulted in ≥20% editing. Perfect correction to wild-type HEXA with PE3 or PE3b and the best PEgRNA proceeded with similar average efficiencies (30% for PE3 vs. 33% for PE3b), but the PE3b system was accompanied by 5.3-fold fewer indel products (1.7% for PE3 vs. 0.32% for PE3b) (FIG. 43B and FIG. 50B). These findings demonstrate the ability of prime editing to make precise small insertions and deletions that install or correct a pathogenic allele in mammalian cells efficiently and with a minimum of byproducts.


Finally, the installation of a protective SNP into PRNP, the gene encoding the human prion protein (PrP), was tested. PrP misfolding causes progressive and fatal neurodegenerative prion disease that can arise spontaneously, through inherited dominant mutations in the PRNP gene, or through exposure to misfolded PrP172. A naturally occurring PRNP G127V mutant allele confers resistance to prion disease in humans138 and mice173. PE3 was used to install G127V into the human PRNP allele in HEK293T cells, which requires a G•C-to-T•A transversion. Four PEgRNAs and three nicking sgRNAs were evaluated with the PE3 system. After three days of exposure to the most effective PE3 and PEgRNA, DNA sequencing revealed 53±11% efficiency of installing the G127V mutation and indel levels of 1.7±0.7% (FIG. 43C). Taken together, these results establish the ability of prime editing in human cells to install or correct transversion, insertion, or deletion mutations that cause or confer resistance to disease efficiently, and with a minimum of byproducts.


Prime Editing in Various Human Cell Lines and Primary Mouse Neurons


Next, prime editing was tested for its ability to edit endogenous sites in three additional human cell lines. In K562 (leukemic bone marrow) cells, PE3 was used to perform transversion edits in the HEK3, EMX1, and FANCF sites, as well as the 18-bp insertion of a 6×His tag in HEK3. An average editing efficiency of 15-30% was observed for each of these four PE3-mediated edits, with indels averaging 0.85-2.2% (FIG. 43A). In U2OS (osteosarcoma) cells, transversion mutations in HEK3 and FANCF were installed, as well as a 3-bp insertion and 6×His tag insertion into HEK3, with 7.9-22% editing efficiency that exceeded indel formation 10- to 76 fold (FIG. 43A). Finally, in HeLa (cervical cancer) cells, a 3-bp insertion into HEK3 was performed, with 12% average efficiency and 1.3% indels (FIG. 43A). Collectively, these data indicate that multiple cell lines beyond HEK293T cells support prime editing, although editing efficiencies vary by cell type and are generally less efficient than in HEK293T cells. Editing:indel ratios remained high in all tested human cell lines.


To determine if prime editing is possible in post-mitotic, terminally differentiated primary cells, primary cortical neurons harvested from E18.5 mice were transduced with a dual split-PE3 lentiviral delivery system in which split-intein splicing203 reconstitutes PE2 protein from N-terminal and C-terminal halves, each delivered from a separate virus. To restrict editing to post-mitotic neurons, the human synapsin promoter, which is highly specific for mature neurons204, was used to drive expression of both PE2 protein components. GFP was fused through a self-cleaving P2A peptide205 to the N-terminal half of PE2. Nuclei from neurons were isolated two weeks following dual viral transduction and were sequenced directly, or sorted for GFP expression before sequencing. A 7.1±1.2% average prime editing to install a transversion at the DNMT1 locus with 0.58±0.14% average indels in sorted nuclei (FIG. 43D was observed. Cas9 nuclease in the same split-intein dual lentivirus system resulted in 31±5.5% indels among sorted cortical neuron nuclei (FIG. 43D. These data indicate that post-mitotic, terminally differentiated primary cells can support prime editing, and thus establish that prime editing does not require cell replication.


Prime Editing Compared with Cas9-Initiated HDR


The performance of PE3 was compared with that of optimized Cas9-initiated HDR128,125 in mitotic cell lines that support HDR128. HEK293T, HeLa, K562 and U2OS cells were treated with Cas9 nuclease, a sgRNA, and an ssDNA donor oligonucleotide template designed to install a variety of transversion and insertion edits (FIGS. 43E-43G, and FIGS. 51A-51F). Cas9-initiated HDR in all cases successfully installed the desired edit, but with far higher levels of byproducts (predominantly indels), as expected from treatments that cause double-stranded breaks. Using PE3 in HEK293T cells, HBB E6V installation and correction proceeded with 42% and 58% average editing efficiency with 2.6% and 1.4% average indels, respectively (FIG. 43E and FIG. 43G). In contrast, the same edits with Cas9 nuclease and an HDR template resulted in 5.2% and 6.7% average editing efficiency, with 79% and 51% average indel frequency (FIG. 43E and FIG. 43G). Similarly, PE3 installed PRNP G127V with 53% efficiency and 1.7% indels, whereas Cas9-initiated HDR installed this mutation with 6.9% efficiency and 53% indels (FIG. 43E and FIG. 43G). Thus, the ratio of editing:indels for HBB E6V installation, HBB E6V correction, and PRNP G127V installation on average was 270-fold higher for PE3 than for Cas9-initiated HDR.


Comparisons between PE3 and HDR in human cell lines other than HEK293T showed similar results, although with lower PE3 editing efficiencies. For example, in K562 cells, PE3-mediated 3-bp insertion into HEK3 proceeded with 25% efficiency and 2.8% indels, compared with 17% editing and 72% indels for Cas9-initiated HDR, a 40-fold editing:indel ratio advantage favoring PE3 (FIGS. 43F-43G). In U2OS cells, PE3 performed this 3-bp insertion with 22% efficiency and 2.2% indels, while Cas9-initiated HDR resulted in 15% editing with 74% indels, a 49-fold lower editing:indel ratio (FIGS. 43F-43G). In HeLa cells, PE3 made this insertion with 12% efficiency and 1.3% indels, versus 3.0% editing and 69% indels for Cas9-initiated HDR, a 210-fold editing:indel ratio difference (FIGS. 43F-43G). Collectively, these data indicated that HDR typically results in similar or lower editing efficiencies and far higher indels than PE3 in the four cell lines tested (FIGS. 51A-51F).


Discussion and Future Directions


The ability to insert DNA sequences with single-nucleotide precision is an especially enabling prime editing capability. For example, PE3 was used to precisely insert into the HEK3 locus in HEK293T cells a His6tag (18 bp, 65% average efficiency), a FLAG epitope tag (24 bp, 18% average efficiency), and an extended LoxP site (44 bp, 23% average efficiency) that is the native substrate for Cre recombinase. Average indels ranged between 3.0% and 5.9% for these examples (FIG. 43H). Many biotechnological, synthetic biology, and therapeutic applications are envisioned to arise from the ability to efficiently and precisely introduce new DNA sequences into target sites of interest in living cells.


Collectively, the prime editing experiments described herein installed 18 insertions up to 44 bp, 22 deletions up to 80 bp, 113 point mutations including 77 transversions, and 18 combination edits, across 12 endogenous loci in the human and mouse genomes at locations ranging from 3 bp upstream to 29 bp downstream of the start of a PAM without making explicit double-stranded DNA breaks. These results establish prime editing as a remarkably versatile genome editing method. Because the overwhelming majority (85-99%) of insertions, deletions, indels, and duplications in ClinVar are ≤30 bp (FIGS. 52A-52D), in principle prime editing can correct up to ˜89% of the 75,122 currently known pathogenic human genetic variants in ClinVar (transitions, transversions, insertions, deletions, indels, and duplications in FIG. 38A), with additional potential to ameliorate diseases caused by copy number gain or loss.


Importantly, for any desired edit the flexibility of prime editing offers many possible choices of PEgRNA-induced nick locations, sgRNA-induced second nick locations, PBS lengths, RT template lengths, and which strand to edit first, as demonstrated extensively herein. This flexibility, which contrasts with more limited options typically available for other precision genome editing methods125,142,154, allows editing efficiency, product purity, DNA specificity, or other parameters to be optimized to suit the needs of a given application, as shown in FIGS. 50A-50B in which testing 14 and 43 PEgRNAs covering a range of prime editing strategies optimized correction of pathogenic HBB and HEXA alleles, respectively.


Much additional research is needed to further understand and improve prime editing. Additional modifications of prime editor systems may be required to expand their compatibility to include other cell types, such as post-mitotic cells. Interfacing prime editing with viral and non-viral in vitro and in vivo delivery strategies is needed to fully explore the potential of prime editing to enable a wide range of applications including the study and treatment of genetic diseases. By enabling highly precise targeted transitions, transversions, small insertions, and small deletions in the genomes of mammalian cells without requiring double-stranded breaks or HDR, however, prime editing provides a new “search-and-replace” capability that substantially expands the scope of genome editing.


Methods


General Methods


DNA amplification was conducted by PCR using Phusion U Green Multiplex PCR Master Mix (ThermoFisher Scientific) or Q5 Hot Start High-Fidelity 2× Master Mix (New England BioLabs) unless otherwise noted. DNA oligonucleotides, including Cy5-labeled DNA oligonucleotides, dCas9 protein, and Cas9 H840A protein were obtained from Integrated DNA Technologies. Yeast reporter plasmids were derived from previously described plasmids64 and cloned by the Gibson assembly method. All mammalian editor plasmids used herein were assembled using the USER cloning method as previously described175. Plasmids expressing sgRNAs were constructed by ligation of annealed oligonucleotides into BsmBI-digested acceptor vector. Plasmids expressing PEgRNAs were constructed by Gibson assembly or Golden Gate assembly using a custom acceptor plasmid (see supplemental ‘Golden Gate assembly’ outline). Sequences of sgRNA and PEgRNA constructs used herein are listed in Tables 2A-2C and Tables 3A-3R. All vectors for mammalian cell experiments were purified using Plasmid Plus Midiprep kits (Qiagen) or PureYield plasmid miniprep kits (Promega), which include endotoxin removal steps. All experiments using live animals were approved by the Broad Institute Institutional and Animal Care and Use Committees. Wild-type C57BL/6 mice were obtained from Charles River (#027).


In Vitro Biochemical Assays


PEgRNAs and sgRNAs were transcribed in vitro using the HiScribe T7 in vitro transcription kit (New England Biolabs) from PCR-amplified templates containing a T7 promoter sequence. RNA was purified by denaturing urea PAGE and quality-confirmed by an analytical gel prior to use. 5′-Cy5-labeled DNA duplex substrates were annealed using two oligonucleotides (Cy5-AVA024 and AVA025; 1:1.1 ratio) for the non-nicked substrate or three oligonucleotides (Cy5-AVA023, AVA025 and AVA026; 1:1.1:1.1) for the pre-nicked substrate by heating to 95° C. for 3 minutes followed by slowly cooling to room temperature (Tables 2A-2C). Cas9 cleavage and reverse transcription reactions were carried out in 1× cleavage buffer205 supplemented with dNTPs (20 mM HEPES-K, pH 7.5; 100 mM KC1; 5% glycerol; 0.2 mM EDTA, pH 8.0; 3 mM MgCl2; 0.5 mM dNTP mix; 5 mM DTT). dCas9 or Cas9 H840A (5 μM final) and the sgRNA or PEgRNA (5 μM final) were pre-incubated at room temperature in a 5 μL reaction mixture for 10 minutes prior to the addition of duplex DNA substrate (400 nM final), followed by the addition of Superscript III reverse transcriptase (ThermoFisher Scientific), an undisclosed M-MLV RT variant, when applicable. Reactions were carried out at 37° C. for 1 hour, then diluted to a volume of 10 μL with water, treated with 0.2 μL of proteinase K solution (20 mg/mL, ThermoFisher Scientific), and incubated at room temperature for 30 minutes. Following heat inactivation at 95° C. for 10 minutes, reaction products were combined with 2× formamide gel loading buffer (90% formamide; 10% glycerol; 0.01% bromophenol blue), denatured at 95° C. for 5 minutes, and separated by denaturing urea-PAGE gel (15% TBE-urea, 55° C., 200V). DNA products were visualized by Cy5 fluorescence signal using a Typhoon FLA 7000 biomolecular imager.


Electrophoretic mobility shift assays were carried out in 1× binding buffer (1× cleavage buffer+10 μg/mL heparin) using pre-incubated dCas9:sgRNA or dCas9:PEgRNA complexes (concentration range between 5 nM and 1 μM final) and Cy5-labeled duplex DNA (Cy5-AVA024 and AVA025; 20 nM final). After 15 minutes of incubation at 37° C., the samples were analyzed by native PAGE gel (10% TBE) and imaged for Cy5 fluorescence.


For DNA sequencing of reverse transcription products, fluorescent bands were excised and purified from urea-PAGE gels, then 3′ tailed with terminal transferase (TdT; New England Biolabs) in the presence of dGTP or dATP according to the manufacturer's protocol. Tailed DNA products were diluted 10-fold with binding buffer (40% saturated aqueous guanidinium chloride+60% isopropanol) and purified by QIAquick spin column (Qiagen), then used as templates for primer extension by Klenow fragment (New England Biolabs) using primer AVA134 (A-tailed products) or AVA135 (G-tailed products) (Tables 2A-2C). Extension were amplified by PCR for 10 cycles using primers AVA110 and AVA122, then sequenced with AVA037 using the Sanger method (Tables 2A-2C).


Yeast Fluorescent Reporter Assays


Dual fluorescent reporter plasmids containing an in-frame stop codon, a +1 frameshift, or a −1 frameshift were subjected to 5′-extended PEgRNA or 3′-extended PEgRNA prime editing reactions in vitro as described above. Following incubation at 37° C. for 1 hour, the reactions were diluted with water and plasmid DNA was precipitated with 0.3 M sodium acetate and 70% ethanol. Resuspended DNA was transformed into S. cerevisiae by electroporation as previously described67 and plated on synthetic complete media without leucine (SC(glucose), L-). GFP and mCherry fluorescence signals were visualized from colonies with the Typhoon FLA 7000 biomolecular imager.


General Mammalian Cell Culture Conditions


HEK293T (ATCC CRL-3216), U2OS (ATTC HTB-96), K562 (CCL-243), and HeLa (CCL-2) cells were purchased from ATCC and cultured and passaged in Dulbecco's Modified Eagle's Medium (DMEM) plus GlutaMAX (ThermoFisher Scientific), McCoy's 5A Medium (Gibco), RPMI Medium 1640 plus GlutaMAX (Gibco), or Eagle's Minimal Essential Medium (EMEM, ATCC), respectively, each supplemented with 10% (v/v) fetal bovine serum (Gibco, qualified) and 1× Penicillin Streptomycin (Corning). All cell types were incubated, maintained, and cultured at 37° C. with 5% CO2. Cell lines were authenticated by their respective suppliers and tested negative for mycoplasma.


HEK293T Tissue Culture Transfection Protocol and Genomic DNA Preparation


HEK293T cells grown were seeded on 48-well poly-D-lysine coated plates (Corning). 16 to 24 hours post-seeding, cells were transfected at approximately 60% confluency with 1 μL of Lipofectamine 2000 (Thermo Fisher Scientific) according to the manufacturer's protocols and 750 ng of PE plasmid, 250 ng of PEgRNA plasmid, and 83 ng of sgRNA plasmid (for PE3 and PE3b). Unless otherwise stated, cells were cultured 3 days following transfection, after which the media was removed, the cells were washed with 1×PBS solution (Thermo Fisher Scientific), and genomic DNA was extracted by the addition of 150 μL of freshly prepared lysis buffer (10 mM Tris-HCl, pH 7.5; 0.05% SDS; 25 μg/mL Proteinase K (ThermoFisher Scientific)) directly into each well of the tissue culture plate. The genomic DNA mixture was incubated at 37° C. for 1 to 2 hours, followed by an 80° C. enzyme inactivation step for 30 minutes. Primers used for mammalian cell genomic DNA amplification are listed in Table 4. For HDR experiments in HEK293T cells, 231 ng of nuclease-expression plasmid, 69 ng of sgRNA expression plasmid, 50 ng (1.51 pmol) 100-nt ssDNA donor template (PAGE-purified; Integrated DNA Technologies) was lipofected using 1.4 μL Lipofectamine 2000 (ThermoFisher) per well. Genomic DNA from all HDR experiments was purified using the Agencourt DNAdvance Kit (Beckman Coulter), according to the manufacturer's protocol.


High-Throughput DNA Sequencing of Genomic DNA Samples


Genomic sites of interest were amplified from genomic DNA samples and sequenced on an Illumina MiSeq as previously described with the following modifications129,130. Briefly, amplification primers containing Illumina forward and reverse adapters (Table 4) were used for a first round of PCR (PCR 1) amplifying the genomic region of interest. 25 μL PCR 1 reactions were performed with 0.5 μM of each forward and reverse primer, 1 μL of genomic DNA extract and 12.5 μL of Phusion U Green Multiplex PCR Master Mix. PCR reactions were carried out as follows: 98° C. for 2 minutes, then 30 cycles of [98° C. for 10 seconds, 61° C. for 20 seconds, and 72° C. for 30 seconds], followed by a final 72° C. extension for 2 minutes. Unique Illumina barcoding primer pairs were added to each sample in a secondary PCR reaction (PCR 2). Specifically, 25 μL of a given PCR 2 reaction contained 0.5 μM of each unique forward and reverse illumina barcoding primer pair, 1 μL of unpurified PCR 1 reaction mixture, and 12.5 μL of Phusion U Green Multiplex PCR 2× Master Mix. The barcoding PCR 2 reactions were carried out as follows: 98° C. for 2 minutes, then 12 cycles of [98° C. for 10 seconds, 61° C. for 20 seconds, and 72° C. for 30 seconds], followed by a final 72° C. extension for 2 minutes. PCR products were evaluated analytically by electrophoresis in a 1.5% agarose gel. PCR 2 products (pooled by common amplicons) were purified by electrophoresis with a 1.5% agarose gel using a QIAquick Gel Extraction Kit (Qiagen), eluting with 40 μL of water. DNA concentration was measured by fluorometric quantification (Qubit, ThermoFisher Scientific) or qPCR (KAPA Library Quantification Kit-Illumina, KAPA Biosystems) and sequenced on an Illumina MiSeq instrument according to the manufacturer's protocols.


Sequencing reads were demultiplexed using MiSeq Reporter (Illumina). Alignment of amplicon sequences to a reference sequence was performed using CRISPResso2178. For quantification of point mutation editing, CRISPResso2 was run in standard mode with “discard_indel_reads” on. Editing efficiency was calculated as: (frequency of specified point mutation in non-discarded reads)×(# of non-discarded reads)÷total reads. For insertion or deletion edits, CRISPResso2 was run in HDR mode using the desired allele as the expected allele (e flag), and with “discard_indel_reads” ON. Editing yield was calculated as the number of HDR aligned reads divided by total reads. For all edits, indel yields were calculated as the number of discarded reads divided by total reads.


Nucleofection of U205, K562, and HeLa Cells


Nucleofection was performed in all experiments using K562, HeLa, and U2OS cells. For PE conditions in these cell types, 800 ng prime editor-expression plasmid, 200 ng PEgRNA-expression plasmid, and 83 ng nicking plasmid was nucleofected in a final volume of 20 μL in a 16-well nucleocuvette strip (Lonza). For HDR conditions in these three cell types, 350 ng nuclease-expression plasmid, 150 ng sgRNA-expression plasmid and 200 pmol (6.6 μg) 100-nt ssDNA donor template (PAGE-purified; Integrated DNA Technologies) was nucleofected in a final volume of 20 μL per sample in a 16-well Nucleocuvette strip (Lonza). K562 cells were nucleofected using the SF Cell Line 4D-Nucleofector X Kit (Lonza) with 5×105 cells per sample (program FF-120), according to the manufacturers protocol. U2OS cells were nucleofected using the SE Cell Line 4D-Nucleofector X Kit (Lonza) with 3-4×105 cells per sample (program DN-100), according to the manufacturers protocol. HeLa cells were nucleofected using the SE Cell Line 4D-Nucleofector X Kit (Lonza) with 2×105 cells per sample (program CN-114), according to the manufacturers protocol. Cells were harvested 72 hours after nucleofection for genomic DNA extraction.


Genomic DNA Extraction for HDR Experiments


Genomic DNA from all HDR comparison experiments in HEK293T, HEK293T HBB E6V, K562, U205, and HeLa cells was purified using the Agencourt DNAdvance Kit (Beckman Coulter), according to the manufacturer's protocol.


Comparison Between PE2, PE3, BE2, BE4max, ABEdmax, and ABEmax


HEK293T cells were seeded on 48-well poly-D-lysine coated plates (Corning). After 16 to 24 hours, cells were transfected at approximately 60% confluency. For base editing with CBE or ABE constructs, cells were transfected with 750 ng of base editor plasmid, 250 ng of sgRNA expression plasmid, and 1 μL of Lipofectamine 2000 (Thermo Fisher Scientific). PE transfections were performed as described above. Genomic DNA extraction for PE and BE was performed as described above.


Determination of PE3 Activity at Known Cas9 Off-Target Sites


To evaluate PE3 off-target editing activity at known Cas9 off-target sites, genomic DNA extracted from HEK293T cells 3 days after transfection with PE3 was used as template for PCR amplification of 16 previously reported Cas9 off-target genomic sites118,159 (the top four off-target sites each for the HEK3, EMX1, FANCF, and HEK4 spacers; primer sequences are listed in Table 4). These genomic DNA samples were identical to those used for quantifying on-target PE3 editing activities shown in FIGS. 41A-41K; PEgRNA and nicking sgRNA sequences are listed in Tables 3A-3R. Following PCR amplification of off-target sites, amplicons were sequenced on the Illumina MiSeq platform as described above (HTS analysis). For determining Cas9 nuclease, Cas9 H840A nickase, dCas9, and PE2-dRT on-target and off-target editing activity, HEK293T cells were transfected with 750 ng of editor plasmid (Cas9 nuclease, Cas9 H840A nickase, dCas9, or PE2-dRT), 250 ng of PEgRNA or sgRNA plasmid, and 1 μL of Lipofectamine 2000. Genomic DNA was isolated from cells 3 days after transfection as described above. On-target and off-target genomic loci were amplified by PCR using primer sequences in Table 4 and sequenced on an Illumina MiSeq.


HTS data analysis was performed using CRISPResso2178. The editing efficiencies of Cas9 nuclease, Cas9 H840A nickase, and dCas9 were quantified as the percent of total sequencing reads containing indels. For quantification of PE3 and PE3-dRT off-targets, aligned sequencing reads were examined for point mutations, insertions, or deletions that were consistent with the anticipated product of PEgRNA reverse transcription initiated at the Cas9 nick site. Single nucleotide variations occurring at <0.1% overall frequency among total reads within a sample were excluded from analysis. For reads containing single nucleotide variations that both occurred at frequencies ≥0.1% and were partially consistent with the PEgRNA-encoded edit, t-tests (unpaired, one-tailed, α=0.5) were used to determine if the variants occurred at significantly higher levels compared to samples treated with PEgRNAs that contained the same spacer but encoded different edits. To avoid differences in sequencing errors, comparisons were made between samples that were sequenced simultaneously within the same MiSeq run. Variants that did not meet the criteria of p-value >0.05 were excluded. Off-target PE3 editing activity was then calculated as the percentage of total sequencing reads that met the above criteria.


Generation of a HEK293T Cell Line Containing the HBB E6V Mutation Using Cas9-Initiated HDR


HEK293T cells were seeded in a 48-well plate and transfected at approximately 60% confluency with 1.5 μL of Lipofectamine 2000, 300 ng of Cas9 D10A nickase plasmid, 100 ng of sgRNA plasmid, and 200 ng of 100-mer ssDNA donor template (Table 5). Three days after transfection, media was exchanged for fresh media. Four days after transfection, cells were dissociated using 30 μL of TrypLE solution and suspended in 1.5 mL of media. Single cells were isolated into individual wells of two 96-well plates by fluorescence-activated cell sorting (FACS) (Beckman-Coulter Astrios). See FIGS. 53A-53B for representative FACS sorting examples. Cells were expanded for 14 days prior to genomic DNA sequencing as described above. Of the isolated clonal populations, none was found to be homozygous for the HBB E6V mutation, so a second round of editing by lipofection, sorting, and outgrowth was repeated in a partially edited cell line to yield a cell line homozygous for the E6V allele.


Generation of a HEK293T Cell Line Containing the HBB E6V Mutation Using PE3


2.5×104 HEK293T cells grown in the absence of antibiotic were seeded on 48-well poly-D-lysine coated plates (Corning). 16 to 24 hours post-seeding, cells were transfected at approximately 70% confluency with 1 μL of Lipofectamine 2000 (Thermo Fisher Scientific) according to the manufacturer's protocols and 750 ng of PE2-P2A-GFP plasmid, 250 ng of PEgRNA plasmid, and 83 ng of sgRNA plasmid. After 3 days post transfection, cells were washed with phosphate-buffered saline (Gibco) and dissociated using TrypLE Express (Gibco). Cells were then diluted with DMEM plus GlutaMax (Thermo Fisher Scientific) supplemented with 10% (v/v) FBS (Gibco) and passed through a 35-μm cell strainer (Corning) prior to sorting. Flow cytometry was carried out on a LE-MA900 cell sorter (Sony). Cells were treated with 3 nM DAPI (BioLegend) 15 minutes prior to sorting. After gating for doublet exclusion, single DAPI-negative cells with GFP fluorescence above that of a GFP-negative control cell population were sorted into 96-well flat-bottom cell culture plates (Corning) filled with pre-chilled DMEM with GlutaMax supplemented with 10% FBS. See FIGS. 53A-53B for representative FACS sorting examples. Cells were cultured for 10 days prior to genomic DNA extraction and characterization by HTS, as described above. A total of six clonal cell lines were identified that are homozygous for the E6V mutation in HBB.


Generation of a HEK293T Cell Line Containing the HEXA 1278+TATC Insertion Using PE3


HEK293T cells containing the HEXA 1278+TATC allele were generated following the protocol described above for creation of the HBB E6V cell line; PEgRNA and sgRNA sequences are listed in Tables 2A-2C under the FIGS. 43A-43H subheading. After transfection and sorting, cells were cultured for 10 days prior to genomic DNA extraction and characterization by HTS, as described above. Two heterozygous cell lines were isolated that contained 50% HEXA 1278+TATC alleles, and two homozygous cell lines containing 100% HEXA 1278+TATC alleles were recovered.


Cell Viability Assays


HEK293T cells were seeded in 48-well plates and transfected at approximately 70% confluency with 750 ng of editor plasmid (PE3, PE3 R110S K103L, Cas9 H840A nickase, or dCas9), 250 ng of HEK3-targeting PEgRNA plasmid, and 1 μL of Lipofectamine 2000, as described above. Cell viability was measured every 24 hours post-transfection for 3 days using the CellTiter-Glo 2.0 assay (Promega) according to the manufacturer's protocol. Luminescence was measured in 96-well flat-bottomed polystyrene microplates (Corning) using a M1000 Pro microplate reader (Tecan) with a 1-second integration time.


Lentivirus Production


Lentivirus was produced as previously described206. T-75 flasks of rapidly dividing HEK293T cells (ATCC; Manassas, VA, USA) were transfected with lentivirus production helper plasmids pVSV-G and psPAX2 in combination with modified lentiCRISPR_v2 genomes carrying intein-split PE2 editor using FuGENE HD (Promega, Madison, WI, USA) according to the manufacturer's directions. Four split-intein editor constructs were designed: 1) a viral genome encoding a U6-PEgRNA expression cassette and the N-terminal portion (1-573) of Cas9 H840A nickase fused to the Npu N-intein, a self-cleaving P2A peptide, and GFP-KASH; 2) a viral genome encoding the Npu C-intein fused to the C-terminal remainder of PE2; 3) a viral genome encoding the Npu C-intein fused to the C-terminal remainder of Cas9 for the Cas9 control; and 4) a nicking sgRNA for DNMT1. The split-intein mediates trans splicing to join the two halves of PE2 or Cas9, while the P2A GFP-KASH enables co-translational production of a nuclear membrane-localized GFP. After 48 hours, supernatant was collected, centrifuged at 500 g for 5 minutes to remove cellular debris, and filtered using a 0.45 μm filter. Filtered supernatant was concentrated using the PEG-it Virus Precipitation Solution (System Biosciences, Palo Alto, CA, USA) according to the manufacturer's directions. The resulting pellet was resuspended in Opti-MEM (Thermo Fisher Scientific, Waltham, MA, USA) using 1% of the original media volume. Resuspended pellet was flash-frozen and stored at −80° C. until use.


Mouse Primary Cortical Neuron Dissection and Culture


E18.5 dissociated cortical cultures were harvested from timed-pregnant C57BL/6 mice (Charles River). Embryos were harvested from pregnant mice after euthanasia by CO2 followed by decapitation. Cortical caps were dissected in ice-cold Hibernate-E supplemented with penicillin/streptomycin (Life Technologies). Following a rinse with ice-cold Hibernate-E, tissue was digested at 37° C. for 8 minutes in papain/DNase (Worthington/Sigma). Tissue was triturated in NBActiv4 (BrainBits) supplemented with DNase. Cells were counted and plated in 24-well plates at 100,000 cells per well. Half of the media was changed twice per week.


Prime Editing in Primary Neurons and Nuclei Isolation


At DIV 1, 15 μL of lentivirus was added at 10:10:1 ratio of N-terminal:C-terminal:nicking sgRNA. At DIV 14, neuronal nuclei were isolated using the EZ-PREP buffer (Sigma D8938) following the manufacturer's protocol. All steps were performed on ice or at 4° C. Media was removed from dissociated cultures, and cultures were washed with ice-cold PBS. PBS was aspirated and replaced with 200 μL EZ-PREP solution. Following a 5-minute incubation on ice, EZ-PREP was pipetted across the surface of the well to dislodge remaining cells. The sample was centrifuged at 500 g for 5 minutes, and the supernatant removed. Samples were washed with 200 μL EZ-PREP and centrifuged again at 500 g for 5 minutes. Samples were resuspended with gentle pipetting in 200 μL ice-cold Nuclei Suspension Buffer (NSB) consisting of 100 μg/mL BSA and 3.33 μM Vybrant DyeCycle Ruby (Thermo Fisher) in 1×PBS, then centrifuged at 500 g for 5 minutes. The supernatant was removed and nuclei were resuspended in 100 μL NSB and sorted into 100 μL Agencourt DNAdvance lysis buffer using a MoFlo Astrios (Beckman Coulter) at the Broad Institute flow cytometry facility. Genomic DNA was purified according to the manufacturer's Agencourt DNAdvance instructions.


RNA-Sequencing and Data Analysis


HEK293T cells were co-transfected with PRNP-targeting or HEXA-targeting PEgRNAs and PE2, PE2-dRT, or Cas9 H840A nickase. 72 hours following transfection, total RNA was harvested from cells using TRIzol reagent (Thermo Fisher) and purified with RNeasy Mini kit (Qiagen) including on-column DNaseI treatment. Ribosomes were depleted from total RNA using the rRNA removal protocol of the TruSeq Stranded Total RNA library prep kit (Illumina) and subsequently washed with RNAClean XP beads (Beckman Coulter). Sequencing libraries were prepared using ribo-depleted RNA on a SMARTer PrepX Apollo NGS library prep system (Takara) following the manufacturer's protocol. Resulting libraries were visualized on a 2200 TapeStation (Agilent Technologies), normalized using a Qubit dsDNA HS assay (Thermo Fisher), and sequenced on a NextSeq 550 using high output v2 flow cell (Illumina) as 75-bp paired-end reads. Fastq files were generated with bcl2fastq2 version 2.20 and trimmed using TrimGalore version 0.6.2 (github.com/FelixKrueger/TrimGalore) to remove low-quality bases, unpaired sequences, and adaptor sequences. Trimmed reads were aligned to a Homo sapiens genome assembly GRCh148 with a custom Cas9 H840A gene entry using RSEM version 1.3.1207. The limma-voom208 package was used to normalize gene expression levels and perform differential expression analysis with batch effect correction. Differentially expressed genes were called with FDR-corrected p-value <0.05 and fold-change >2 cutoffs, and results were visualized in R.


ClinVar Analysis


The ClinVar variant summary was downloaded from NCBI (accessed Jul. 15, 2019), and the information contained therein was used for all downstream analysis. The list of all reported variants was filtered by allele ID in order to remove duplicates and by clinical significance in order to restrict the analysis to pathogenic variants. The list of pathogenic variants was filtered sequentially by variant type in order to calculate the fraction of pathogenic variants that are insertions, deletions, etc. Single nucleotide variants (SNVs) were separated into two categories (transitions and transversions) based on the reported reference and alternate alleles. SNVs that did not report reference or alternate alleles were excluded from the analysis.


The lengths of reported insertions, deletions, and duplications were calculated using reference/alternate alleles, variant start/stop positions, or appropriate identifying information in the variant name. Variants that did not report any of the above information were excluded from the analysis. The lengths of reported indels (single variants that include both insertions and deletions relative to the reference genome) were calculated by determining the number of mismatches or gaps in the best pairwise alignment between the reference and alternate alleles. Frequency distributions of variant lengths were calculated using GraphPad Prism 8.


Data Availability


High-throughput sequencing data are deposited to the NCBI Sequence Read Archive database. Plasmids encoding PE1, PE2/PE3, and PEgRNA expression vectors will be available from Addgene.


Code Availability


The script used to quantify PEgRNA scaffold insertion is provided in FIGS. 60A-60B.


Supplemental Information: Tables and Sequences









TABLE 1





Activities of prime editors, Cas9 nuclease, Cas9 H840A nickase, and PE2-dRT at


HEK3, HEK4, EMX1, and FANCF on-target and off-target sites. PE2/PE3 editing is shown as % prime


editing alongside % indels (in parentheses). % indels are shown for Cas9, Cas9 H840A nickase (nCas9),


and PE2-dRT at the top four previously characterized off-target sites179, 180. sgRNA and PEgRNA


sequences can be found in Tables 3A-3R, under the FIGs. 42A-42H heading. All values are the average of


three independent biological replicates.

















PE pegRNA












HEK3 (PE3)
HEK4 (PE2)
EXM1 (PE3)
FANCF (PE3)



























Site

1
2
3
4

1
2
3
4

1
2
3
4

1
2
3
4





On-target

44.2
61.2
40.4
48.4

18.2
14.4
9.8
7.9

28.6
14.1
35.7
15.4

56.8
32.4
42.8
47.6




(11.9)
(8.8)
(16.5)
(3.3)

(0.9)
(1.8)
(2.0)
(2.2)

(3.5)
(2.4)
(3.3)
(2.9)

(9.3)
(16.7)
(13.6)
(12.0)


Off-target 1

<0.1
<0.1
<0.1
<0.1

<0.1
0.4
<0.1
0.4

<0.1
<0.1
<0.1
<0.1

<0.1
0.6
<0.1
<0.1




(<0.1)
(<0.1)
(<0.1)
(<0.1)

(<0.1)
(<0.1)
(<0.1)
(0.1)

(0.1)
(0.1)
(0.1)
(0.1)

(0.1)
(0.1)
(0.1)
(<0.1)


Off-target 2

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1




(<0.1)
(<0.1)
(<0.1)
(<0.1)

(0.1)
(0.1)
(0.1)
(0.1)

(<0.1)
(0.1)
(0.1)
(0.1)

(<0.1)
(<0.1)
(0.1)
(<0.1)


Off-target 3

<0.1
<0.1
<0.1
<0.1

0.2
6.8
19.2
7.9

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1




(<0.1)
(<0.1)
(<0.1)
(<0.1)

(0.5)
(1.9)
(0.5)
(3.5)

(0.3)
(0.3)
(0.3)
(0.3)

(<0.1)
(<0.1)
(0.1)
(<0.1)


Off-target 4

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1




(0.1)
(0.1)
(0.1)
(<0.1)

(<0.1)
(0.1)
(0.2)
(<0.1)

(0.1)
(0.1)
(0.2)
(0.1)

(<0.1)
(<0.1)
(<0.1)
(<0.1)









Cas9 pegRNA












HEK3
HEK4
EXM1
FANCF



























Site
SgRNA
1
2
3
4
SgRNA
1
2
3
4
SgRNA
1
2
3
4
SgRNA
1
2
3
4





On-target
91.8
87.5
89.2
89.1
86.8
71.8
68.6
72.8
72.8
70.9
85.6
79.7
70.6
76.6
76.0
78.7
55.9
58.3
51.8
52.0


Off-target 1
17.2
1.9
5.5
5.2
1.8
54.2
39.5
48.4
49.7
49.2
81.1
63.5
48.1
53.0
59.6
12.6
1.9
1.9
1.7
1.7


Off-target 2
38.0
6.5
12.6
11.8
4.7
42.5
19.5
29.4
27.3
30.3
58.3
12.0
6.0
8.2
12.9
1.1
0.2
0.2
0.2
0.1


Off-target 3
8.8
0.6
1.7
1.5
0.5
98.1
96.9
97.3
97.6
97.5
14.8
4.2
3.1
3.6
4.8
2.4
0.2
<0.1
0.2
0.2


Off-target 4
0.3
<0.1
<0.1
0.1
<0.1
45.3
16.9
28.0
27.5
29.7
39.5
1.3
0.9
0.6
1.3
1.0
0.2
0.2
0.2
0.2












nCas9 pegRNA












HEK3
HEK4
EXM1
FANCF



























Site

1
2
3
4

1
2
3
4

1
2
3
4

1
2
3
4





Off-target 1

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1

0.1
0.1
<0.1
0.1

<0.1
<0.1
<0.1
<0.1


Off-target 2

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1

0.3
0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1


Off-target 3

<0.1
<0.1
<0.1
<0.1

0.3
0.5
0.7
0.7

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1


Off-target 4

<0.1
<0.1
<0.1
<0.1

<0.1
0.1
<0.1
<0.1

1.5
0.1
0.1
0.4

<0.1
<0.1
<0.1
<0.1












PE2-dRT pegRNA












HEK3
HEK4
EXM1
FANCF



























Site

1
2
3
4

1
2
3
4

1
2
3
4

1
2
3
4





Off-target 1

<0.1
<0.1
<0.1
<0.1

0.1
0.2
<0.1
<0.1

0.1
0.1
0.1
0.1

<0.1
<0.1
<0.1
<0.1


Off-target 2

<0.1
<0.1
<0.1
<0.1

<0.1
0.1
<0.1
<0.1

<0.1
0.3
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1


Off-target 3

<0.1
<0.1
<0.1
<0.1

<0.1
0.1
1.4
0.9

<0.1
<0.1
<0.1
<0.1

<0.1
<0.1
<0.1
<0.1


Off-target 4

<0.1
<0.1
<0.1
<0.1

0.1
0.1
0.1
0.2

0.9
0.1
0.1
0.2

<0.1
<0.1
<0.1
<0.1









Tables 2A-2C: Sequences of DNA oligonucleotides, PEgRNAs, and sgRNAs used for in vitro experiments.









TABLE 2A







DNA oligonucleotides








OLIGO-



NUCLEOTIDE
SEQUENCE





AVA023
5CY5-CCTGGGTCAATCCTTGGGGCCCAGACTGAGCACG (SEQ ID NO:



374)





AVA024
5CY5-



CCTGGGTCAATCCTTGGGGCCCAGACTGAGCACGTGATGGCAGAGG



AAAGG (SEQ ID NO: 375)





AVA025
5PHOS-



CCTTTCCTCTGCCATCACGTGCTCAGTCTGGGCCCCAAGGATTGACC



CAGG (SEQ ID NO: 376)





AVA026
5PHOS-TGATGGCAGAGGAAAGG (SEQ ID NO: 377)





AVA037
GCAGGCTTTAAAGGAACCAATTC (SEQ ID NO: 378)





AVA110
GCAGGCTTTAAAGGAACCAATTCCCTGGGTCAATCCTTGGGGC (SEQ



ID NO: 379)





AVA122
CTCTGGAGGATCTAGCGGAG (SEQ ID NO: 380)





AVA134
CTCTGGAGGATCTAGCGGAGTTTTTTTTTTTTTTTTTTTT (SEQ ID NO:



381)





AVA135
CTCTGGAGGATCTAGCGGAGCCCCCCCCCCCCCC (SEQ ID NO: 382)
















TABLE 2B







5′-extended PEgRNAs

















RT





LINKER
PBS
TEMPLATE



SPACER
5′ EXTENSION
LENGTH
LENGTH
LENGTH


PEGRNA
SEQUENCE
SEQUENCE
(NT)
(NT)
(NT)





PEGRNA1
GGCCCAGACTGA
GGCTAACCGTGCCATTT
15
5
 7



GCACGTGA (SEQ
GATCAGGTCA (SEQ ID






ID NO: 383)
NO: 429)








PEGRNA 2
GGCCCAGACTGA
GGCTAACCGTGCAAATT
15
5
 7



GCACGTGA (SEQ
AACAAACTAA (SEQ ID






ID NO: 384)
NO: 430)








PEGRNA 3
GGCCCAGACTGA
GGCCATCTCGTGCAAAT
15
5
 8



GCACGTGA (SEQ
TAACAAACTAA (SEQ ID






ID NO: 385)
NO: 431)








PEGRNA 4
GGCCCAGACTGA
GGTCCTCTGCCATCTCG
15
5
15



GCACGTGA (SEQ
TGCAAATTAACAAACTA






ID NO: 386)
A (SEQ ID NO: 432)








PEGRNA 5
GGCCCAGACTGA
GGCTTCCTTTCCTCTGC
15
5
22



GCACGTGA (SEQ
CATCTCGTGCAAATTAA






ID NO: 387)
CAAACTAA (SEQ ID NO:







433)








5′-
GGCCCAGACTGA
GGCTAACCGTGCCATTT
15
5
 7


PEGRNA_RT_7_
GCACGTGA (SEQ
GATCAGGTCA (SEQ ID





A
ID NO: 388)
NO: 434)








5′-
GGCCCAGACTGA
GGCTAACCGTGCAAATT
15
5
 7


PEGRNA_RT_7_
GCACGTGA (SEQ
AACAAACTAA (SEQ ID





B
ID NO: 389)
NO: 435)








5′-PEGRNA_RT_8
GGCCCAGACTGA
GGCCATCTCGTGCAAAT
15
5
 8



GCACGTGA (SEQ
TAACAAACTAA (SEQ ID






ID NO: 390)
NO: 436)








5′-
GGCCCAGACTGA
GGTCCTCTGCCATCTCG
15
5
15


PEGRNA_RT_15
GCACGTGA (SEQ
TGCAAATTAACAAACTA






ID NO: 391)
A (SEQ ID NO: 437)








5′-
GGCCCAGACTGA
GGCTTCCTTTCCTCTGC
15
5
22


PEGRNA_RT_22
GCACGTGA (SEQ
CATCTCGTGCAAATTAA






ID NO: 392)
CAAACTAA (SEQ ID NO:







438)
















TABLE 2C







3′-extended PEgRNAs















RT





PBS
TEMPLATE



SPACER
3′ EXTENSION
LENGTH
LENGTH


PEGRNA
SEQUENCE
SEQUENCE
(NT)
(NT)





3′-PEGRNA_10
GGCCCAGACTGAGC
TCTGCCATCTCGT
7
10



ACGTGA (SEQ ID NO:
GCTC (SEQ ID NO:





506)
439)







3′-
GGCCCAGACTGAGC
TCTGCCATCTCGT
7
10


PEGRNA_YEAST_
ACGTGA (SEQ ID NO:
GCTC(SEQ ID NO:




TTOA
507)
440)







3′-
GGCCCAGACTGAGC
TCTGCCATCATCG
7
11


PEGRNA_YEAST_
ACGTGA (SEQ ID NO:
TGCTC(SEQ ID NO:




+1AINS
508)
441)







3′-
GGCCCAGACTGAGC
TCTGCCATCCGTG
7
 9


PEGRNA_YEAST_
ACGTGA (SEQ ID NO:
CTC(SEQ ID NO:




+1TDEL
509)
442)









Tables 3A-3R: Sequences of PEgRNAs and sgRNAs used in mammalian cell experiments. All sequences are shown in 5′ to 3′ orientation. To construct PEgRNAs, spacer sequences listed below were added to the 5′ end of the sgRNA scaffold and the 3′ extensions listed below containing the primer binding site and RT template were added to the 3′ end of the sgRNA scaffold. The sgRNA scaffold sequence is









(SEQ ID NO: 131)


GTTTTAGAGCTAGAAATAGCAAGTTAAAATAAGGCTAGTCCGTTATCAA


CTTGAAAAAGTGGCACCGAGTCGGTGC.













TABLE 3A







FIGS. 39A-39D PEgRNA















RT



SPACER SEQUENCE

PBS
TEMPLATE



(SEQ ID NOS:
3′ EXTENSION (SEQ ID
LENGTH
LENGTH


PEGRNA
2890-2996)
NOS: 2997-3103)
(NT)
(NT)





HEK3_2B-
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCA
 8
10


C_8
ACGTGA








HEK3_2B-
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCAG
 9
10


C_9
ACGTGA








HEK3_2B-
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCAG
10
10


C_10
ACGTGA
T







HEK3_2B-
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCAG
11
10


C_11
ACGTGA
TC







HEK3_2B-
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCAG
12
10


C_12
ACGTGA
TCT







HEK3_2B-
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCAG
13
10


C_13
ACGTGA
TCTG







HEK3_2B-
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCAG
14
10


C_14
ACGTGA
TCTGG







HEK3_2B-
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCAG
15
10


C_15
ACGTGA
TCTGGG







HEK3_2C_16
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCAG
16
10



ACGTGA
TCTGGGC







HEK3_2C_17
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCAG
17
10



ACGTGA
TCTGGGCC







EMX1_2C_9
GAGTCCGAGCAGAA
ATGGGAGCACTTCTTCTTC
 9
13



GAAGAA
TGC







EMX1_2C_10
GAGTCCGAGCAGAA
ATGGGAGCACTTCTTCTTC
10
13



GAAGAA
TGCT







EMX1_2C_11
GAGTCCGAGCAGAA
ATGGGAGCACTTCTTCTTC
11
13



GAAGAA
TGCTC







EMX1_2C_12
GAGTCCGAGCAGAA
ATGGGAGCACTTCTTCTTC
12
13



GAAGAA
TGCTCG







EMX1_2C_13
GAGTCCGAGCAGAA
ATGGGAGCACTTCTTCTTC
13
13



GAAGAA
TGCTCGG







EMX1_2C_14
GAGTCCGAGCAGAA
ATGGGAGCACTTCTTCTTC
14
13



GAAGAA
TGCTCGGA







EMX1_2C_15
GAGTCCGAGCAGAA
ATGGGAGCACTTCTTCTTC
15
13



GAAGAA
TGCTCGGAC







EMX1_2C_16
GAGTCCGAGCAGAA
ATGGGAGCACTTCTTCTTC
16
13



GAAGAA
TGCTCGGACT







EMX1_2C_17
GAGTCCGAGCAGAA
ATGGGAGCACTTCTTCTTC
17
13



GAAGAA
TGCTCGGACTC







FANCF_2C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
 8
17


8
GCACC
CTGCAGA







FANCF_2C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
 9
17


9
GCACC
CTGCAGAA







FANCF_2C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
10
17


10
GCACC
CTGCAGAAG







FANCF_2C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
11
17


11
GCACC
CTGCAGAAGG







FANCF_2C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
12
17


12
GCACC
CTGCAGAAGGG







FANCF_2C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
13
17


13
GCACC
CTGCAGAAGGGA







FANCF_2C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
14
17


14
GCACC
CTGCAGAAGGGAT







FANCF_2C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
15
17


15
GCACC
CTGCAGAAGGGATT







FANCF_2C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
16
17


16
GCACC
CTGCAGAAGGGATTC







FANCF_2C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
17
17


17
GCACC
CTGCAGAAGGGATTCC







RNF2_2C_9
GTCATCTTAGTCATT
GAACACCTCATGTAATGAC
 9
11



ACCTG
T







RNF2_2C_10
GTCATCTTAGTCATT
GAACACCTCATGTAATGAC
10
11



ACCTG
TA







RNF2_2C_11
GTCATCTTAGTCATT
GAACACCTCATGTAATGAC
11
11



ACCTG
TAA







RNF2_2C_12
GTCATCTTAGTCATT
GAACACCTCATGTAATGAC
12
11



ACCTG
TAAG







RNF2_2C_13
GTCATCTTAGTCATT
GAACACCTCATGTAATGAC
13
11



ACCTG
TAAGA







RNF2_2C_14
GTCATCTTAGTCATT
GAACACCTCATGTAATGAC
14
11



ACCTG
TAAGAT







RNF2_2C_15
GTCATCTTAGTCATT
GAACACCTCATGTAATGAC
15
11



ACCTG
TAAGATG







RNF2_2C_16
GTCATCTTAGTCATT
GAACACCTCATGTAATGAC
16
11



ACCTG
TAAGATGA







RNF2_2C_17
GTCATCTTAGTCATT
GAACACCTCATGTAATGAC
17
11



ACCTG
TAAGATGAC







HEK4_2C_7
GGCACTGCGGCTGG
GCTTTAACCCCAACCTCCA
 7
13



AGGTGG
G







HEK4_2C_8
GGCACTGCGGCTGG
GCTTTAACCCCAACCTCCA
 8
13



AGGTGG
GC







HEK4_2C_9
GGCACTGCGGCTGG
GCTTTAACCCCAACCTCCA
 9
13



AGGTGG
GCC







HEK4_2C_10
GGCACTGCGGCTGG
GCTTTAACCCCAACCTCCA
10
13



AGGTGG
GCCG







HEK4_2C_11
GGCACTGCGGCTGG
GCTTTAACCCCAACCTCCA
11
13



AGGTGG
GCCGC







HEK4_2C_12
GGCACTGCGGCTGG
GCTTTAACCCCAACCTCCA
12
13



AGGTGG
GCCGCA







HEK4_2C_13
GGCACTGCGGCTGG
GCTTTAACCCCAACCTCCA
13
13



AGGTGG
GCCGCAG







HEK4_2C_14
GGCACTGCGGCTGG
GCTTTAACCCCAACCTCCA
14
13



AGGTGG
GCCGCAGT







HEK4_2C_15
GGCACTGCGGCTGG
GCTTTAACCCCAACCTCCA
15
13



AGGTGG
GCCGCAGTG







HEK3_2C_
GGCCCAGACTGAGC
TCTGCCATCCGTGCTCAGT
13
10


1TDEL
ACGTGA
CTG







HEK3_2C_
GGCCCAGACTGAGC
TCTGCCATCATCGTGCTCA
13
10


1AINS
ACGTGA
GTCTG







HEK3_2C_
GGCCCAGACTGAGC
TCTGCCATCAAAGCGTGCT
13
10


1CTTINS
ACGTGA
CAGTCTG







HEK3_2D_10
GGCCCAGACTGAGC
TCTGCCATCTCGTGCTCAG
13
10



ACGTGA
TCTG







HEK3_2D_11
GGCCCAGACTGAGC
CTCTGCCATCTCGTGCTCA
13
11



ACGTGA
GTCTG







HEK3_2D_12
GGCCCAGACTGAGC
CCTCTGCCATCTCGTGCTC
13
12



ACGTGA
AGTCTG







HEK3_2D_13
GGCCCAGACTGAGC
TCCTCTGCCATCTCGTGCTC
13
13



ACGTGA
AGTCTG







HEK3_2D_14
GGCCCAGACTGAGC
TTCCTCTGCCATCTCGTGCT
13
14



ACGTGA
CAGTCTG







HEK3_2D_15
GGCCCAGACTGAGC
TTTCCTCTGCCATCTCGTGC
13
15



ACGTGA
TCAGTCTG







HEK3_2D_16
GGCCCAGACTGAGC
CTTTCCTCTGCCATCTCGTG
13
16



ACGTGA
CTCAGTCTG







HEK3_2D_17
GGCCCAGACTGAGC
CCTTTCCTCTGCCATCTCGT
13
17



ACGTGA
GCTCAGTCTG







HEK3_2D_18
GGCCCAGACTGAGC
TCCTTTCCTCTGCCATCTCG
13
18



ACGTGA
TGCTCAGTCTG







HEK3_2D_19
GGCCCAGACTGAGC
TTCCTTTCCTCTGCCATCTC
13
19



ACGTGA
GTGCTCAGTCTG







HEK3_2D_20
GGCCCAGACTGAGC
CTTCCTTTCCTCTGCCATCT
13
20



ACGTGA
CGTGCTCAGTCTG







EMX1_2D_10
GAGTCCGAGCAGAA
GGAGCCCTTGTTCTTCTGC
13
10



GAAGAA
TCGG







EMX1_2D_11
GAGTCCGAGCAGAA
GGGAGCCCTTGTTCTTCTG
13
11



GAAGAA
CTCGG







EMX1_2D_12
GAGTCCGAGCAGAA
TGGGAGCCCTTGTTCTTCT
13
12



GAAGAA
GCTCGG







EMX1_2D_13
GAGTCCGAGCAGAA
ATGGGAGCCCTTGTTCTTC
13
13



GAAGAA
TGCTCGG







EMX1_2D_14
GAGTCCGAGCAGAA
GATGGGAGCCCTTGTTCTT
13
14



GAAGAA
CTGCTCGG







EMX1_2D_15
GAGTCCGAGCAGAA
TGATGGGAGCCCTTGTTCT
13
15



GAAGAA
TCTGCTCGG







EMX1_2D_16
GAGTCCGAGCAGAA
GTGATGGGAGCCCTTGTTC
13
16



GAAGAA
TTCTGCTCGG







EMX1_2D_17
GAGTCCGAGCAGAA
TGTGATGGGAGCCCTTGTT
13
17



GAAGAA
CTTCTGCTCGG







EMX1_2D_18
GAGTCCGAGCAGAA
ATGTGATGGGAGCCCTTGT
13
18



GAAGAA
TCTTCTGCTCGG







EMX1_2D_19
GAGTCCGAGCAGAA
GATGTGATGGGAGCCCTTG
13
19



GAAGAA
TTCTTCTGCTCGG







EMX1_2D_20
GAGTCCGAGCAGAA
TGATGTGATGGGAGCCCTT
13
20



GAAGAA
GTTCTTCTGCTCGG







FANCF_2D_
GGAATCCCTTCTGCA
CGATCAAGGTGCTGCAGAA
13
10


10
GCACC
GGGA







FANCF_2D_
GGAATCCCTTCTGCA
GCGATCAAGGTGCTGCAGA
13
11


11
GCACC
AGGGA







FANCF_2D_
GGAATCCCTTCTGCA
AGCGATCAAGGTGCTGCAG
13
12


12
GCACC
AAGGGA







FANCF_2D_
GGAATCCCTTCTGCA
AAGCGATCAAGGTGCTGCA
13
13


13
GCACC
GAAGGGA







FANCF_2D_
GGAATCCCTTCTGCA
AAAGCGATCAAGGTGCTGC
13
14


14
GCACC
AGAAGGGA







FANCF_2D_
GGAATCCCTTCTGCA
AAAAGCGATCAAGGTGCT
13
15


15
GCACC
GCAGAAGGGA







FANCF_2D_
GGAATCCCTTCTGCA
GAAAAGCGATCAAGGTGC
13
16


16
GCACC
TGCAGAAGGGA







FANCF_2D_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
13
17


17
GCACC
CTGCAGAAGGGA







FANCF_2D_
GGAATCCCTTCTGCA
CGGAAAAGCGATCAAGGT
13
18


18
GCACC
GCTGCAGAAGGGA







FANCF_2D_
GGAATCCCTTCTGCA
TCGGAAAAGCGATCAAGG
13
19


19
GCACC
TGCTGCAGAAGGGA







FANCF_2D_
GGAATCCCTTCTGCA
CTCGGAAAAGCGATCAAG
13
20


20
GCACC
GTGCTGCAGAAGGGA







RNF2_2D_10
GTCATCTTAGTCATT
AACACCTCATGTAATGACT
15
10



ACCTG
AAGATG







RNF2_2D_11
GTCATCTTAGTCATT
GAACACCTCATGTAATGAC
15
11



ACCTG
TAAGATG







RNF2_2D_12
GTCATCTTAGTCATT
CGAACACCTCATGTAATGA
15
12



ACCTG
CTAAGATG







RNF2_2D_13
GTCATCTTAGTCATT
ACGAACACCTCATGTAATG
15
13



ACCTG
ACTAAGATG







RNF2_2D_14
GTCATCTTAGTCATT
AACGAACACCTCATGTAAT
15
14



ACCTG
GACTAAGATG







RNF2_2D_15
GTCATCTTAGTCATT
CAACGAACACCTCATGTAA
15
15



ACCTG
TGACTAAGATG







RNF2_2D_16
GTCATCTTAGTCATT
ACAACGAACACCTCATGTA
15
16



ACCTG
ATGACTAAGATG







RNF2_2D_17
GTCATCTTAGTCATT
TACAACGAACACCTCATGT
15
17



ACCTG
AATGACTAAGATG







RNF2_2D_18
GTCATCTTAGTCATT
TTACAACGAACACCTCATG
15
18



ACCTG
TAATGACTAAGATG







RNF2_2D_19
GTCATCTTAGTCATT
GTTACAACGAACACCTCAT
15
19



ACCTG
GTAATGACTAAGATG







RNF2_2D_20
GTCATCTTAGTCATT
AGTTACAACGAACACCTCA
15
20



ACCTG
TGTAATGACTAAGATG







HEK4_2D_7
GGCACTGCGGCTGG
ACCCCAACCTCCAGCCGC
11
 7



AGGTGG








HEK4_2D_8
GGCACTGCGGCTGG
AACCCCAACCTCCAGCCGC
11
 8



AGGTGG








HEK4_2D_9
GGCACTGCGGCTGG
TAACCCCAACCTCCAGCCG
11
 9



AGGTGG
C







HEK4_2D_10
GGCACTGCGGCTGG
TTAACCCCAACCTCCAGCC
11
10



AGGTGG
GC







HEK4_2D_11
GGCACTGCGGCTGG
TTTAACCCCAACCTCCAGC
11
11



AGGTGG
CGC







HEK4_2D_12
GGCACTGCGGCTGG
CTTTAACCCCAACCTCCAG
11
12



AGGTGG
CCGC







HEK4_2D_13
GGCACTGCGGCTGG
GCTTTAACCCCAACCTCCA
11
13



AGGTGG
GCCGC







HEK4_2D_14
GGCACTGCGGCTGG
CGCTTTAACCCCAACCTCC
11
14



AGGTGG
AGCCGC







HEK4_2D_15
GGCACTGCGGCTGG
CCGCTTTAACCCCAACCTC
11
15



AGGTGG
CAGCCGC







HEK4_2D_16
GGCACTGCGGCTGG
TCCGCTTTAACCCCAACCT
11
16



AGGTGG
CCAGCCGC







HEK4_2D_17
GGCACTGCGGCTGG
CTCCGCTTTAACCCCAACC
11
17



AGGTGG
TCCAGCCGC







HEK4_2D_18
GGCACTGCGGCTGG
CTCCGCTTTAACCCCAACC
11
18



AGGTGG
TCCAGCCGC







HEK4_2D_19
GGCACTGCGGCTGG
CTCCGCTTTAACCCCAACC
11
19



AGGTGG
TCCAGCCGC
















TABLE 3B







FIGS. 40A-40C PEgRNA















RT





PBS
TEMPLATE



SPACER SEQUENCE
3′ EXTENSION (SEQ 
LENGTH
LENGTH


PEGRNA
3104-3112)
ID NOS: 3113-3121)
(NT)
(NT)





RNF2_3B
GTCATCTTAGTCATT
AACGAACACCTCATGTAAT
15
14



ACCTG
GACTAAGATG







EMX1_3B
GAGTCCGAGCAGAA
ATGGGAGCACTTCTTCTTC
15
13



GAAGAA
TGCTCGGAC







FANCF_3B
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
15
17



GCACC
CTGCAGAAGGGATT







HE3_3B
GGCCCAGACTGAGC
TCTGCCATGACGTGCTCAG
13
10



ACGTGA
TCTG







HEK4_3B
GGCACTGCGGCTGG
TTAACCCCAACCTCCAGCC
 9
10



AGGTGG








RNF2_3C_
GTCATCTTAGTCATT
AACGAACACCGCAGGTAA
15
14


4ATOC
ACCTG
TGACTAAGATG







RNF2_3C_
GTCATCTTAGTCATT
AACGAACACCCCAGGTAA
15
14


4ATOG
ACCTG
TGACTAAGATG







FANCF_3C_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGGTG
13
17


5GTOT
GCACC
CTGCAGAAGGGA







FANCF_3C_
GGAATCCCTTCTGCA
GGAAAAGCGAGCCAGGTG
14
17


7ATOC
GCACC
CTGCAGAAGGGAT
















TABLE 3C







FIGS. 40A-40C nicking sgRNA sequences











SEQ


NICKING SGRNA
SPACER SEQUENCE
ID NO:





RNF2_2B_+41
GTCAACCATTAAGCAAAACAT
3122





RNF2_2B_+67
GTCTCAGGCTGTGCAGACAAA
3123





EMX1_2B_-116
GGGGCACAGATGAGAAACTC
3124





EMX1_2B_-57
GCCGTTTGTACTTTGTCCTC
3125





EMX1_2B_+14
GCGCCACCGGTTGATGTGAT
3126





EMX1_2B_+27
GCTTCGTGGCAATGCGCCAC
3127





EMX1_2B_+53
GACATCGATGTCCTCCCCAT
3128





EMX1_2B_+80
GTGGTTGCCCACCCTAGTCAT
3129





FANCF_2B_-78
GCGACTCTCTGCGTACTGAT
3130





FANCF_2B_-50
GCCCTACTTCCGCTTTCACCT
3131





FANCF_2B_-27
GGATTCCATGAGGTGCGCGA
3132





FANCF_2B_-17
GCTGCAGAAGGGATTCCATG
3133





FANCF_2B_+21
GCTTGAGACCGCCAGAAGCT
3134





FANCF_2B_+48
GGGGTCCCAGGTGCTGACGT
3135





HEK3_2B_-108
GCAGAAATAGACTAATTGCA
3136





HEK3_2B_-38
GGATTGACCCAGGCCAGGGC
3137





HEK3_2B_+26
GACGCCCTCTGGAGGAAGCA
3138





HEK3_2B_+37
GCTGTCCTGCGACGCCCTC
3139





HEK3_2B_+63
GCACATACTAGCCCCTGTCT
3140





HEK3_2B_+90
GTCAACCAGTATCCCGGTGC
3141





HEK4_2B_-95
TCCCTTCCTTCCACCCAGCC
3142





HEK4_2B_-51
CCCTGCCTGTCATCCTGCTT
3143





HEK4_2B_-26
GCAGTGCCACCGGGGCGCCG
3144





HEK4_2B_+52
GCGGGGGCTCAGAGAGGGCA
3145





HEK4_2B_+74
GAGACACACACACAGGCCTGG
3146





RNF2_2C_+41
GTCAACCATTAAGCAAAACAT
3147





RNF2_2C_4ATOC_+5
GTGAGTTACAACGAACACCGC
3148





RNF2_2C_4ATOG_+5
GTGAGTTACAACGAACACCCC
3149





FANCF_2C_+48
GGGGTCCCAGGTGCTGACGT
3150





FANCF_2C_5GTOT_+7
GAAGCTCGGAAAAGCGATCA
3151





FANCF_2C_7ATOC_+7
GAAGCTCGGAAAAGCGAGCC
3152





HEK3_2C_+90
GTCAACCAGTATCCCGGTGC
3153
















TABLE 3D







FIGS. 41A-41K PEgRNA















RT





PBS
TEMPLATE



SPACER SEQUENCE
3′ EXTENSION (SEQ
LENGTH
LENGTH


PEGRNA
(SEQ ID NO: 3154-3304)
ID NO: 3305-3455)
(NT)
(NT)





HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCATCTCGTGCTCAGT
13
10


1TTOA
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCATCGCGTGCTCAGT
13
10


1TTOC
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCATCGCGTGCTCAGT
13
10


1TTOG
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCATTACGTGCTCAGT
13
10


2GTOA
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCATGACGTGCTCAGT
13
10


2GTOC
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCATAACGTGCTCAGT
13
10


2GTOT
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCAGCACGTGCTCAG
13
10


3ATOC
CGTGA
TCTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCACCACGTGCTCAG
13
10


3ATOG
CGTGA
TCTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCAACACGTGCTCAG
13
10


3ATOT
CGTGA
TCTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCTTCACGTGCTCAGT
13
10


4TTOA
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCGTCACGTGCTCAG
13
10


4TTOC
CGTGA
TCTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCCCTCACGTGCTCAGT
13
10


4TTOG
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCTATCACGTGCTCAGT
13
10


5GTOA
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCGATCACGTGCTCAGT
13
10


5GTOC
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGCAATCACGTGCTCAGT
13
10


5GTOT
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGTCATCACGTGCTCAGT
13
10


6GTOA
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGGCATCACGTGCTCAGT
13
10


6GTOC
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTGACATCACGTGCTCAGT
13
10


6GTOT
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTTCCATCACGTGCTCAGT
13
10


7CTOA
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTCCCATCACGTGCTCAGT
13
10


7CTOG
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCTACCATCACGTGCTCAGT
13
10


7CTOT
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCGGCCATCACGTGCTCAG
13
10


8ATOC
CGTGA
TCTG







HEK3_4A_
GGCCCAGACTGAGCA
TCCGCCATCACGTGCTCAGT
13
10


8ATOG
CGTGA
CTG







HEK3_4A_
GGCCCAGACTGAGCA
TCAGCCATCACGTGCTCAG
13
10


8ATOT
CGTGA
TCTG







HEK3_4B_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
34


1TTOA
CGTGA
TTTCCTCTGCCATCTCGTGC






TCAGTCTG







HEK3_4B_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
34


12GTOC
CGTGA
TTTGCTCTGCCATCACGTGC






TCAGTCTG







HEK3_4B_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
34


14ATOT
CGTGA
TATCCTCTGCCATCACGTGC






TCAGTCTG







HEK3_4B_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
34


17GTOC
CGTGA
TTTCCTCTGCCATCACGTGC






TCAGTCTG







HEK3_4B_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
34


20GTOC
CGTGA
TTTCCTCTGCCATCACGTGC






TCAGTCTG







HEK3_4B_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
34


23CTOG
CGTGA
TTTCCTCTGCCATCACGTGC






TCAGTCTG







HEK3_4B_
GGCCCAGACTGAGCA
TGGAGGAAGCTGGGCTTCC
13
34


24TTOA
CGTGA
TTTCCTCTGCCATCACGTGC






TCAGTCTG







HEK3_4B_
GGCCCAGACTGAGCA
TGGAGGAACCAGGGCTTCC
13
34


26CTOG
CGTGA
TTTCCTCTGCCATCACGTGC






TCAGTCTG







HEK3_4B_
GGCCCAGACTGAGCA
TGGACGAAGCAGGGCTTCC
13
34


30CTOG
CGTGA
TTTCCTCTGCCATCACGTGC






TCAGTCTG







HEK3_4B_
GGCCCAGACTGAGCA
TCGAGGAAGCAGGGCTTCC
13
34


33CTOG
CGTGA
TTTCCTCTGCCATCACGTGC






TCAGTCTG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACACCTCATGTAAT
15
14


1CTOA
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACACCTCACGTAAT
15
14


1CTOG
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACACCTCAAGTAAT
15
14


1CTOT
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACACCTCTGGTAAT
15
14


2TTOA
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACACCTCCGGTAAT
15
14


2TTOG
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACACCTGAGGTAAT
15
14


3GTOC
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACACCGCAGGTAAT
15
14


4ATOC
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACACCACAGGTAAT
15
14


4ATOT
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACACCCCAGGTAAT
15
14


4ATOG
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACACATCAGGTAAT
15
14


5GTOT
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACATCTCAGGTAAT
15
14


6GTOA
CCTG
GACTAAGATG







RNF2_4C_
GTCATCTTAGTCATTA
AACGAACGCCTCAGGTAAT
15
14


7TTOC
CCTG
GACTAAGATG







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGATCCAGGCGC
14
17


1ATOG
GCACC
TGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGATCCAGGAGC
14
17


1ATOT
GCACC
TGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGATCCAGTTGC
14
17


2CTOA
GCACC
TGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGATCCACGTGC
14
17


3CTOG
GCACC
TGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGATCCAAGTGC
14
17


3CTOT
GCACC
TGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGATCCTGGTGC
14
17


4TTOA
GCACC
TGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGATCCCGGTGC
14
17


4TTOG
GCACC
TGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGATCTAGGTGC
14
17


5GTOA
GCACC
TGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGATGCAGGTGC
14
17


6GTOC
GCACC
TGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGAGCCAGGTG
14
17


7ATOC
GCACC
CTGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGCGGTCCAGGTGC
14
17


8TTOC
GCACC
TGCAGAAGGGAT







FANCF_4D_
GGAATCCCTTCTGCA
GGAAAAGAGATCCAGGTGC
14
17


10GTOT
GCACC
TGCAGAAGGGAT







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGGAGCCCTGCTTC
14
16


2ATOC
GAAGAA
TTCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGGAGCCCTACTTCT
14
16


2ATOT
GAAGAA
TCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGGAGCCCCTCTTCT
14
16


3ATOG
GAAGAA
TCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGGAGCCGTTCTTCT
14
16


4GTOC
GAAGAA
TCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGGAGCTCTTCTTCT
14
16


5GTOA
GAAGAA
TCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGGAGCACTTCTTCT
14
16


5GTOT
GAAGAA
TCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGGATCCCTTCTTCT
14
16


7CTOA
GAAGAA
TCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGGTGCCCTTCTTCT
14
16


8TTOA
GAAGAA
TCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGGGGCCCTTCTTCT
14
16


8TTOC
GAAGAA
TCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGGCGCCCTTCTTCT
14
16


8TTOG
GAAGAA
TCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGCAGCCCTTCTTCT
14
16


9CTOG
GAAGAA
TCTGCTCGGA







EMX1_4E_
GAGTCCGAGCAGAA
GTGATGGAAGCCCTTCTTCT
14
16


9CTOT
GAAGAA
TCTGCTCGGA







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCCATCTCTTCC
15
15


1CTOA
AGCGA
TCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCCATCCCTTCC
15
15


1CTOG
AGCGA
TCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCCATCACTTCC
15
15


1CTOT
AGCGA
TCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCCATTGCTTCC
15
15


2GTOA
AGCGA
TCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCCAGCGCTTC
15
15


3ATOC
AGCGA
CTCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCCACCGCTTC
15
15


3ATOG
AGCGA
CTCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCCAACGCTTC
15
15


3ATOT
AGCGA
CTCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCCTTCGCTTCC
15
15


4TTOA
AGCGA
TCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCCGTCGCTTC
15
15


4TTOC
AGCGA
CTCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCCCTCGCTTCC
15
15


4TTOG
AGCGA
TCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGCAATCGCTTCC
15
15


5GTOT
AGCGA
TCCTGAAAAT







RUNX1_4F_
GCATTTTCAGGAGGA
TGTCTGAAGGCATCGCTTCC
15
15


6GTOC
AGCGA
TCCTGAAAAT







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCCT
13
22


1TTOA
GATGA
CTTCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCCT
13
22


1TTOC
GATGA
CGTCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCCT
13
22


1TTOG
GATGA
CCTCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCCT
13
22


2GTOA
GATGA
TATCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCC
13
22


3ATOC
GATGA
GCATCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCC
13
22


3ATOG
GATGA
CCATCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCC
13
22


3ATOT
GATGA
ACATCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCAC
13
22


5GTOT
GATGA
TCATCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGGCC
13
22


6GTOC
GATGA
TCATCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGATCCCT
13
22


7CTOA
GATGA
CATCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAACCCT
13
22


7CTOT
GATGA
CATCTGGCCTGCAGA







VEGFA_4G_
GATGTCTGCAGGCCA
AATGTGCCATCTGCAGCCCT
13
22


9CTOG
GATGA
CATCTGGCCTGCAGA







DNMT1_4H_
GATTCCTGGTGCCAG
GTCACCCCTGGTTCTGGCA
13
11


1ATOC
AAACA
CCAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GTCACCCCTGCTTCTGGCA
13
11


1ATOG
AAACA
CCAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GTCACCCCTTTTTCTGGCAC
13
11


2CTOA
AAACA
CAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GTCACCCCTCTTTCTGGCAC
13
11


2CTOG
AAACA
CAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GTCACCCCTATTTCTGGCAC
13
11


2CTOT
AAACA
CAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GTCACCCCAGTTTCTGGCA
13
11


3ATOT
AAACA
CCAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GTCACCCTTGTTTCTGGCAC
13
11


4GTOA
AAACA
CAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GTCACCACTGTTTCTGGCA
13
11


5GTOT
AAACA
CCAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GTCACGCCTGTTTCTGGCA
13
11


6GTOC
AAACA
CCAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GCCCTCCCGTCTCCCCTGTT
13
19


8TTOA
AAACA
TCTGGCACCAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GCCCTCCCGTCGCCCCTGTT
13
19


8TTOC
AAACA
TCTGGCACCAGG







DNMT1_4H_
GATTCCTGGTGCCAG
GCCCTCCCGTCGCCCCTGTT
13
19


8TTOG
AAACA
TCTGGCACCAGG







HEK3_4J_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
29


DEL1-5
CGTGA
TTTCCTCTGCCGTGCTCAGT






CTG







HEK3_4J_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
24


DEL1-10
CGTGA
TTTCCCGTGCTCAGTCTG







HEK3_4J_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
19


DEL1-15
CGTGA
CGTGCTCAGTCTG







HEK3_4J_
GGCCCAGACTGAGCA
TGTCCTGCGACGCCCTCTG
13
26


DEL1-25
CGTGA
GAGGAAGCGTGCTCAGTCT






G







HEK3_4J_
GGCCCAGACTGAGCA
TGTCCTGCGACGCCCTCTG
13
21


DEL1-30
CGTGA
GACGTGCTCAGTCTG







HEK3_4J_
GGCCCAGACTGAGCA
AGTATCCCGGTGCAGGAGC
13
20


DEL1-80
CGTGA
TCGTGCTCAGTCTG







HEK3_4I_
GGCCCAGACTGAGCA
TCTGCCATCATCGTGCTCAG
13
11


1AINS
CGTGA
TCTG







HEK3_4I_
GGCCCAGACTGAGCA
TCTGCCATCAAAGCGTGCT
13
13


1CTTINS
CGTGA
CAGTCTG







HEK3_4I_
GGCCCAGACTGAGCA
TCTGCCATCCGTGCTCAGTC
13
9


1TDEL
CGTGA
TG







HEK3_4I_1-
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
31


3TGADEL
CGTGA
TTTCCTCTGCCACGTGCTCA






GTCTG







RNF2_4I_
GTCATCTTAGTCATTA
AACGAACACCTCAGAGTAA
15
15


1TINS
CCTG
TGACTAAGATG







RNF2_4I_
GTCATCTTAGTCATTA
AACGAACACCTCAGTACGT
15
17


1GTAINS
CCTG
AATGACTAAGATG







RNF2_4I_
GTCATCTTAGTCATTA
AACGAACACCCAGGTAATG
15
13


4ADEL
CCTG
ACTAAGATG







RNF2_4I_3-
GTCATCTTAGTCATTA
AACGAACACAGGTAATGAC
15
11


5GAGDEL
CCTG
TAAGATG







FANCF_4I_
GGAATCCCTTCTGCA
GGAAAAGCGATCCAGGGTG
14
18


3CINS
GCACC
CTGCAGAAGGGAT







FANCF_4I_
GGAATCCCTTCTGCA
GGAAAAGCGATCCAATCGG
14
20


4GATINS
GCACC
TGCTGCAGAAGGGAT







FANCF_4I_
GGAATCCCTTCTGCA
GGAAAAGCGATCAGGTGCT
14
16


6GDEL
GCACC
GCAGAAGGGAT







FANCF_4I_
GGAATCCCTTCTGCA
GGAAAAGCGAAGGTGCTGC
14
14


5-7GGADEL
GCACC
AGAAGGGAT







EMX1_4I_
GAGTCCGAGCAGAA
GTGATGGGAGCACCTTCTT
14
17


6TINS
GAAGAA
CTTCTGCTCGGA







EMX1_4I_
GAGTCCGAGCAGAA
GTGATGGGAGCCCTTCGCA
14
19


1TGCINS
GAAGAA
TTCTTCTGCTCGGA







EMX1_4I_
GAGTCCGAGCAGAA
GTGATGGGAGCCTTCTTCTT
14
15


5GDEL
GAAGAA
CTGCTCGGA







EMX1_4I_4-
GAGTCCGAGCAGAA
GTGATGGGAGTTCTTCTTCT
14
13


6GGGDEL
GAAGAA
GCTCGGA







RUNX1_4I_
GCATTTTCAGGAGGA
TGTCTGAAGCCATCGGCTTC
15
16


1CINS
AGCGA
CTCCTGAAAAT







RUNX1_4I_
GCATTTTCAGGAGGA
TGTCTGAAGCCATCCATGCT
15
18


1ATGINS
AGCGA
TCCTCCTGAAAAT







RUNX1_4I_
GCATTTTCAGGAGGA
TGTCTGAAGCCATGCTTCCT
15
14


2GDEL
AGCGA
CCTGAAAAT







RUNX1_4I_
GCATTTTCAGGAGGA
TGTCTGAAGCCGCTTCCTCC
15
12


2-4GATDEL
AGCGA
TGAAAAT







VEGFA_4I_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCG
13
23


4CINS
GATGA
CTCATCTGGCCTGCAGA







VEGFA_4I_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCCT
13
25


2ACAINS
GATGA
TGTCATCTGGCCTGCAGA







VEGFA_4I_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCC
13
21


3ADEL
GATGA
CATCTGGCCTGCAGA







VEGFA_4I_
GATGTCTGCAGGCCA
AATGTGCCATCTGGAGCCAT
13
19


2-4GAGDEL
GATGA
CTGGCCTGCAGA







DNMT1_4I_
GATTCCTGGTGCCAG
TCCCGTCACCCGCTGTTTCT
13
16


4CINS
AAACA
GGCACCAGG







DNMT1_4I_
GATTCCTGGTGCCAG
TCCCGTCACCCCTGTGATTT
13
18


1TCAINS
AAACA
CTGGCACCAGG







DNMT1_4I_
GATTCCTGGTGCCAG
TCCCGTCACCCCGTTTCTGG
13
14


3ADEL
AAACA
CACCAGG







DNMT1_4I_
GATTCCTGGTGCCAG
TCCCGTCACCGTTTCTGGCA
13
12


3-5AGGDEL
AAACA
CCAGG







HEK3_4K_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
36


1CTTINS_
CGTGA
TTTCCTCTGCATCAAAGCGT




5GDEL

GCTCAGTCTG







HEK3_4K_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
37


1CTTINS_
CGTGA
TTTCCTCTGCCATGAAAGCG




2GTOC

TGCTCAGTCTG







HEK3_4K_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
33


3TDEL_
CGTGA
TTTCCTCTGCGATCCGTGCT




5GTOC

CAGTCTG







HEK3_4K_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
34


3GTOC_
CGTGA
TTTCCTCTGACATGACGTGC




6GTOT

TCAGTCTG







RNF2_4K_
GTCATCTTAGTCATTA
AACGAACACCATTGGTAAT
15
14


2AAINS_3-
CCTG
GACTAAGATG




4GADEL









RNF2_4K_
GTCATCTTAGTCATTA
AACGAACACGTCAGTGTAA
15
15


1AINS_
CCTG
TGACTAAGATG




5GTOC









RNF2_4K_1-
GTCATCTTAGTCATTA
AACGAACAACTCGTAATGA
15
12


2CTDEL_
CCTG
CTAAGATG




6GTOT









RNF2_4K_
GTCATCTTAGTCATTA
AACGAACACATCATGTAAT
15
14


1CTOA_
CCTG
GACTAAGATG




5GTOT









FANCF_4K_
GGAATCCCTTCTGCA
GGAAAAGCGATCGGTAGCT
14
16


1TINS_3-
GCACC
GCAGAAGGGAT




4TGDEL









FANCF_4K_
GGAATCCCTTCTGCA
GGAAAAGCGATTCAGGTAG
14
18


1TINS_
GCACC
CTGCAGAAGGGAT




6GTOA









FANCF_4K_
GGAATCCCTTCTGCA
GGAAAAGCGATCAAGTGCT
14
16


2CDEL_
GCACC
GCAGAAGGGAT




5GTOT
















TABLE 3E







FIGS. 41A-41K nicking sgRNA









SPACER SEQUENCE (SEQ ID


NICKING SGRNA
NOS: 3456-3463)





HEK3_4A_+90
GTCAACCAGTATCCCGGTGC





HEK3_4B_+90
GTCAACCAGTATCCCGGTGC





RNF2_4C_+41
GTCAACCATTAAGCAAAACAT





FANCF_4D_+48
GGGGTCCCAGGTGCTGACGT





EMX1_4E_+53
GACATCGATGTCCTCCCCAT





RUNX1_4F_+38
GATGAAGCACTGTGGGTACGA





VEGFA_4G_+57
GATGTACAGAGAGCCCAGGGC





DNMTl_4H_+49
GCCCTTCAGCTAAAATAAAGG
















TABLE 3F







FIGS. 42A-42H PEgRNA















RT



SPACER SEQUENCE

PBS
TEMPLATE



(SEQ ID NOS:
3′ EXTENSION (SEQ ID NOS:
LENGTH
LENGTH


PEGRNA
3464-3478)
3479-3493)
(NT)
(NT)





HEK3_5A_
GGCCCAGACTGAGC
TCTGTCATCACGTGCTCAGTCTG
13
10


C3
ACGTGA








HEK3_5A_
GGCCCAGACTGAGC
TCTGCTATCACGTGCTCAGTCTG
13
10


C4
ACGTGA








HEK3_5A_
GGCCCAGACTGAGC
TCTGCCATTACGTGCTCAGTCTG
13
10


C7
ACGTGA








FANCF_5A_
GGAATCCCTTCTGCA
GGAAAAGTGATCCAGGTGCTGC
14
17


C3
GCACC
AGAAGGGAT







FANCF_5A_
GGAATCCCTTCTGCA
GGAAAAGCGATTCAGGTGCTGC
14
17


C7
GCACC
AGAAGGGAT







FANCF_5A_
GGAATCCCTTCTGCA
GGAAAAGCGATCTAGGTGCTGC
14
17


C8
GCACC
AGAAGGGAT







EMX1_5A_
GAGTCCGAGCAGAA
GTGATGGGAGTCCTTCTTCTTCT
14
16


C5
GAAGAA
GCTCGGA







EMX1_5A_
GAGTCCGAGCAGAA
GTGATGGGAGCTCTTCTTCTTCT
14
16


C6
GAAGAA
GCTCGGA







EMX1_5A_
GAGTCCGAGCAGAA
GTGATGGGAGCCTTTCTTCTTCT
14
16


C7
GAAGAA
GCTCGGA







EMX1_5C_
GAGTCCGAGCAGAA
GTGATGGGAGTTCTTCTTCTTCT
14
16


C5_6
GAAGAA
GCTCGGA







EMX1_5C_
GAGTCCGAGCAGAA
GTGATGGGAGTCTTTCTTCTTCT
14
16


C5_7
GAAGAA
GCTCGGA







EMX1_5C_
GAGTCCGAGCAGAA
GTGATGGGAGCTTTTCTTCTTCT
14
16


C6_7
GAAGAA
GCTCGGA







EMX1_5C_
GAGTCCGAGCAGAA
GTGATGGGAGTTTTTCTTCTTCT
14
16


C5_6_7
GAAGAA
GCTCGGA







HEK3_5D_
GGCCCAGACTGAGC
TCTGCCGTCACGTGCTCAGTCT
13
10


A5
ACGTGA
G







HEK3_5D_
GGCCCAGACTGAGC
TCTGCCATCGCGTGCTCAGTCT
13
10


A8
ACGTGA
G
















TABLE 3G







FIGS. 42A-42H nicking sgRNA










POSSIBLE SPACER SEQUENCE
SEQUENCE(SEQ ID NOS:


NICKING SGRNA
(SEQ ID NOS: 616-618)
619-621)





HEK3_5A-F_+90
GTCAACCAGTATCCCGGTGC
GTCAACCAGTATCCCGGTGC





FANCF_5A-F_+48
GGGGTCCCAGGTGCTGACGT
GATGTACAGAGAGCCCAGGGC





EMX1_5A-F_+57
GATGTACAGAGAGCCCAGGGC
GGGGTCCCAGGTGCTGACGT
















TABLE 3H







FIGS. 42A-42H base editing sgRNA








BASE EDITING



SGRNA
SPACER SEQUENCE





HEK3_5A-F_BE
GTGCCATCACGTGCTCAGTCT (SEQ



ID NO: 455)





FANCF_5A-F_BE
GAGCGATCCAGGTGCTGCAGA (SEQ



ID NO: 456)





EMX1_5A-F_BE
GGAGCCCTTCTTCTTCTGCT (SEQ ID



NO: 457)
















TABLE 3I







FIGS. 42A-42H on-target sgRNA








ON-TARGET



SGRNA
SPACER SEQUENCE





HEK3_5G
GGCCCAGACTGAGCACGTGA (SEQ ID



NO: 510)





HEK4_5G
GGCACTGCGGCTGGAGGTGG (SEQ ID



NO: 511)





EMX1_5G
GAGTCCGAGCAGAAGAAGAA (SEQ



ID NO: 512)





FANCF_5G
GGAATCCCTTCTGCAGCACC (SEQ ID



NO: 513)
















TABLE 3J







FIGS. 42A-42H on-target PEgRNA















RT





PBS
TEMPLATE


ON-TARGET
SPACER SEQUENCE
3′ EXTENSION 
LENGTH
LENGTH


PEGRNA
(SEQ ID NO: 663-677)
(SEQ ID NO: 678-692)
(NT)
(NT)





HEK3_5G-
GGCCCAGACTGAGCAC
TCTGCCATCTCGTGCTCAG
13
10


H_PEGRNA_1
GTGA
TCTG







HEK3_5G-
GGCCCAGACTGAGCAC
TCTGCCATCAAAGCGTGCT
13
13


H_PEGRNA_2
GTGA
CAGTCTG







HEK3_5G-
GGCCCAGACTGAGCAC
TCTGCCATCCGTGCTCAGT
13
 9


H_PEGRNA_3
GTGA
CTG







HEK3_5G-
GGCCCAGACTGAGCAC
TCTGCGATCACGTGCTCAG
13
10


H_PEGRNA_4
GTGA
TCTG







HEK4_5G-
GGCACTGCGGCTGGAG
TTAACGCCCACCTCCAGCC
 9
10


H_PEGRNA_1
GTGG








HEK4_5G-
GGCACTGCGGCTGGAG
TTAACCCCCCCCTCCAGCC
 9
10


H_PEGRNA_2
GTGG








HEK4_5G-
GGCACTGCGGCTGGAG
TTAACCCCTTACACCTCCA
 9
13


H_PEGRNA_3
GTGG
GCC







HEK4_5G-
GGCACTGCGGCTGGAG
TTAACCCCCCCTCCAGCC
 9
 9


H_PEGRNA_4
GTGG








EMX1_5G-
GAGTCCGAGCAGAAGA
GTGATGGGAGCACTTCTTC
14
16


H_PEGRNA_1
AGAA
TTCTGCTCGGA







EMX1_5G-
GAGTCCGAGCAGAAGA
GTGATGGGAGCCCTGCTTC
14
16


H_PEGRNA_2
AGAA
TTCTGCTCGGA







EMX1_5G-
GAGTCCGAGCAGAAGA
GTGATGGGAGCCCTTCGCA
14
19


H_PEGRNA_3
AGAA
TTCTTCTGCTCGGA







EMX1_5G-
GAGTCCGAGCAGAAGA
GTGATGGGAGTTCTTCTTC
14
13


H_PEGRNA_4
AGAA
TGCTCGGA







FANCF_5G-
GGAATCCCTTCTGCAG
GGAAAAGCGATGCAGGTG
14
17


H_PEGRNA_1
CACC
CTGCAGAAGGGAT







FANCF_5G-
GGAATCCCTTCTGCAG
GGAAAAGCGATCCAGGCG
14
17


H_PEGRNA_2
CACC
CTGCAGAAGGGAT







FANCF_5G-
GGAATCCCTTCTGCAG
GGAAAAGCGATCCAATCG
14
20


H_PEGRNA_3
CACC
GTGCTGCAGAAGGGAT
















TABLE 3K







FIGS. 49A-49BPEgRNA















RT



SPACER

PBS
TEMPLATE



SEQUENCE (SEQ
3′ EXTENSION (SEQ 
LENGTH
LENGTH


PEGRNA
ID NO: 3494-3521)
ID NO: 3522-3540)
(NT)
(NT)





HEK3_6A_
GGCCCAGACTGA
TCTGCCATGACGTGCTCAG
13
10


2GTOC
GCACGTGA
TCTG







HEK3_6A_
GGCCCAGACTGA





2GTOC
GCACGTGA








EMX1_6A_
GAGTCCGAGCAG
ATGGGAGCCCTTGTTCTTC
13
13


3GTOC
AAGAAGAA
TGCTCGG







EMX1_6A_
GAGTCCGAGCAG





3GTOC
AAGAAGAA








FANCF_6A_
GGAATCCCTTCTG
AAAAGCGATCAAGGTGCT
13
15


5GTOT
CAGCACC
GCAGAAGGGA







FANCF_6A_
GGAATCCCTTCTG





5GTOT
CAGCACC








HEK3_6A_
GGCCCAGACTGA
TGGAGGAAGCAGGGCTTC
13
52


1HIS6INS
GCACGTGA
CTTTCCTCTGCCATCAATG






ATGGTGATGATGGTGCGTG






CTCAGTCTG







HEK3_6A_
GGCCCAGACTGA





1HIS6INS
GCACGTGA








HEK3_6A_
GGCCCAGACTGA
TCTGCAATCACGTGCTCAG
13
10


5GTOT
GCACGTGA
TCTG







HEK3_6A_
GGCCCAGACTGA





5GTOT
GCACGTGA








HEK3_6A_
GGCCCAGACTGA
TCTGCCATCAAAGCGTGCT
13
10


1CTTINS
GCACGTGA
CAGTCTG







HEK3_6A_
GGCCCAGACTGA





1CTTINS
GCACGTGA








HBB_6B_
GCATGGTGCACCT
AGACTTCTCCACAGGAGT
13
14


INSALL
GACTCCTG
CAGGTGCAC







HBB_6B_
GCATGGTGCACCT





INSALL
GACTCCTG








HBB_6B_
GCATGGTGCACCT
AGACTTCTCCTCAGGAGTC
13
14


CORRECT
GACTCCTG
AGGTGCAC







HBB_6B_
GCATGGTGCACCT





CORRECT
GACTCCTG








HBB_6B_COR
GCATGGTGCACCT
AGACTTCTCTTCAGGAGTC
13
14


RECT_W_
GACTCCTG
AGGTGCAC




SILENT









HBB_6B_COR
GCATGGTGCACCT





RECT_W_
GACTCCTG





SILENT









HEXA_6B_
GTACCTGAACCGT
AGTCAGGGCCATAGGATAG
12
14


INSTALL
ATATCCTA
ATATACGGTTC







HEXA_6B_
GATCCTTCCAGTC
ACCTGAACCGTATATCCTAT
10
21


CORRECT
AGGGCCAT
GGCCCTGACTG







HEXA_6B_
GATCCTTCCAGTC
GTACCTGAACCGTATATCTT
 9
27


CORRECT_W_
AGGGCCAT
ATGGCCCTGACT




SILENT









PRNP_6C
GCAGTGGTGGGG
ATGTAGACGCCAAGGCCCC
12
12



GGCCTTGG
CCACC







HEK3_6E-
GGCCCAGACTGA
TCTGCCATCCCGTGCTCAG
13
10


G_1TTOG
GCACGTGA
TCTG







HEK3_6E-
GGCCCAGACTGA
TCTGCCATCAAAGCGTGCT
13
10


G_1CTTINS
GCACGTGA
CAGTCTG







RNF2_6E-
GTCATCTTAGTCA
AACGAACACCTCACGTAAT
15
14


G_1CTOG
TTACCTG
GACTAAGATG







HBB_6E-
GCATGGTGCACCT
AGACTTCTCCACAGGAGT
13
14


G_4ATOT
GACTCCTG
CAGGTGCAC







HEK3_6H_
GGCCCAGACTGA
TGGAGGAAGCAGGGCTTC
13
52


1HIS6INS
GCACGTGA
CTTTCCTCTGCCATCAATG






ATGGTGATGATGGTGCGTG






CTCAGTCTG







HEK3_6H_
GGCCCAGACTGA
TGGAGGAAGCAGGGCTTC
13
58


1FLAGINS
GCACGTGA
CTTTCCTCTGCCATCACTTA






TCGTCGTCATCCTTGTAATC






CGTGCTCAGTCTG
















TABLE 3L







FIGS. 47A-74D PEgRNA















RT





PBS
TEMPLATE



SPACER SEQUENCE
3′ EXTENSION SEQUENCE
LENGTH
LENGTH


PEGRNA
(SEQ ID NO: 3541-3547)
(SEQ ID NO: 3549-3556)
(NT)
(NT)





HEK3_ED4B_
GGCCCAGACTGAGCA
TCTGCCATCCGTGCTCAGTC
13
 9


1TDEL
CGTGA
TG







HEK3_ED4B_
GGCCCAGACTGAGCA
TCTGCCATCATCGTGCTCAG
13
11


1AINS
CGTGA
TCTG







HEK3_ED4B_
GGCCCAGACTGAGCA
TCTGCCATCAAAGCGTGCT
13
13


1CTTINS
CGTGA
CAGTCTG







HEK3_ED4C_
GGCCCAGACTGAGCA
TCTGCCATGACGTGCTCAGT
13
10


2GTOC
CGTGA
CTG







HEK3_ED4D_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
58


1FLAGINS
CGTGA
TTTCCTCTGCCATCACTTAT






CGTCGTCATCCTTGTAATCC






GTGCTCAGTCTG







RNF2_ED4E_
GTCATCTTAGTCATTA
AACGAACACCTCATGTAAT
15
14


1CTOA
CCTG
GACTAAGATG







EMX1_ED4F_
GAGTCCGAGCAGAAG
ATGGGAGCCCTTGTTCTTCT
13
13


1GTOC
AAGAA
GCTCGG







HBB_ED4G_
GTAACGGCAGACTTCT
ATCTGACTCCTGTGGAGAA
12
14


2TTOA
CCTC
GTCTGCC
















TABLE 3M







FIGS. 48A-48C PEgRNA















RT



SPACER SEQUENCE

PBS
TEMPLATE



(SEQ ID NO:
3′ EXTENSION SEQUENCE (SEQ
LENGTH
LENGT


PEGRNA
3557-3627)
ID NO: 3628-3698)
(NT)
H(NT)





VEGFA_
GATGTCTGCAGGCC
CCCTCTGACAATGTGCCATCTG
13
31


ED5A_31
AGATGA
GAGCACTCATCTGGCCTGCAG






A




VEGFA_
GATGTCTGCAGGCC
CCTCTGACAATGTGCCATCTGG
13
30


ED5A_30
AGATGA
AGCACTCATCTGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
CTCTGACAATGTGCCATCTGGA
13
29


ED5A_29
AGATGA
GCACTCATCTGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
TCTGACAATGTGCCATCTGGAG
13
28


ED5A_28
AGATGA
CACTCATCTGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
CTGACAATGTGCCATCTGGAGC
13
27


ED5A_27
AGATGA
ACTCATCTGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
TGACAATGTGCCATCTGGAGCA
13
26


ED5A_26
AGATGA
CTCATCTGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
GACAATGTGCCATCTGGAGCAC
13
25


ED5A_25
AGATGA
TCATCTGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
ACAATGTGCCATCTGGAGCACT
13
24


ED5A_24
AGATGA
CATCTGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
CAATGTGCCATCTGGAGCACTC
13
23


ED5A_23
AGATGA
ATCTGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
AATGTGCCATCTGGAGCACTCA
13
22


ED5A_22
AGATGA
TCTGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
ATGTGCCATCTGGAGCACTCAT
13
21


ED5A_21
AGATGA
CTGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
TGTGCCATCTGGAGCACTCATC
13
20


ED5A_20
AGATGA
TGGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
GTGCCATCTGGAGCACTCATCT
13
19


ED5A_19
AGATGA
GGCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
TGCCATCTGGAGCACTCATCTG
13
18


ED5A_18
AGATGA
GCCTGCAGA







VEGFA_
GATGTCTGCAGGCC
GCCATCTGGAGCACTCATCTGG
13
17


ED5A_17
AGATGA
CCTGCAGA







VEGFA_
GATGTCTGCAGGCC
CCATCTGGAGCACTCATCTGGC
13
16


ED5A_16
AGATGA
CTGCAGA







VEGFA_
GATGTCTGCAGGCC
CATCTGGAGCACTCATCTGGCC
13
15


ED5A_15
AGATGA
TGCAGA







VEGFA_
GATGTCTGCAGGCC
ATCTGGAGCACTCATCTGGCCT
13
14


ED5A_14
AGATGA
GCAGA







VEGFA_
GATGTCTGCAGGCC
TCTGGAGCACTCATCTGGCCTG
13
13


ED5A_13
AGATGA
CAGA







VEGFA_
GATGTCTGCAGGCC
CTGGAGCACTCATCTGGCCTGC
13
12


ED5A_12
AGATGA
AGA







VEGFA_
GATGTCTGCAGGCC
TGGAGCACTCATCTGGCCTGCA
13
11


ED5A_11
AGATGA
GA







VEGFA_
GATGTCTGCAGGCC
GGAGCACTCATCTGGCCTGCA
13
10


ED5A_10
AGATGA
GA







VEGFA_
GATGTCTGCAGGCC
GAGCACTCATCTGGCCTGCAG
13
 9


ED5A_9
AGATGA
A







VEGFA_
GATGTCTGCAGGCC
AGCACTCATCTGGCCTGCAGA
13
 8


ED5A_8
AGATGA








DNMT1_
GATTCCTGGTGCCA
AGGACTAGTTCTGCCCTCCCGT
13
31


ED5B_31
GAAACA
CACCACTGTTTCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
GGACTAGTTCTGCCCTCCCGTC
13
30


ED5B_30
GAAACA
ACCACTGTTTCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
GACTAGTTCTGCCCTCCCGTCA
13
29


ED5B_29
GAAACA
CCACTGTTTCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
ACTAGTTCTGCCCTCCCGTCAC
13
28


ED5B_28
GAAACA
CACTGTTTCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
CTAGTTCTGCCCTCCCGTCACC
13
27


ED5B_27
GAAACA
ACTGTTTCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
TAGTTCTGCCCTCCCGTCACCA
13
26


ED5B_26
GAAACA
CTGTTTCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
AGTTCTGCCCTCCCGTCACCAC
13
25


ED5B_25
GAAACA
TGTTTCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
GTTCTGCCCTCCCGTCACCACT
13
24


ED5B_24
GAAACA
GTTTCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
TTCTGCCCTCCCGTCACCACTG
13
23


ED5B_23
GAAACA
TTTCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
TCTGCCCTCCCGTCACCACTGT
13
22


ED5B_22
GAAACA
TTCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
CTGCCCTCCCGTCACCACTGTT
13
21


ED5B_21
GAAACA
TCTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
TGCCCTCCCGTCACCACTGTTT
13
20


ED5B_20
GAAACA
CTGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
GCCCTCCCGTCACCACTGTTTC
13
19


ED5B_19
GAAACA
TGGCACCAGG







DNMT1_
GATTCCTGGTGCCA
CCCTCCCGTCACCACTGTTTCT
13
18


ED5B_18
GAAACA
GGCACCAGG







DNMT1_
GATTCCTGGTGCCA
CCTCCCGTCACCACTGTTTCTG
13
17


ED5B_17
GAAACA
GCACCAGG







DNMT1_
GATTCCTGGTGCCA
CTCCCGTCACCACTGTTTCTGG
13
16


ED5B_16
GAAACA
CACCAGG







DNMT1_
GATTCCTGGTGCCA
TCCCGTCACCACTGTTTCTGGC
13
15


ED5B_15
GAAACA
ACCAGG







DNMT1_
GATTCCTGGTGCCA
CCCGTCACCACTGTTTCTGGCA
13
14


ED5B_14
GAAACA
CCAGG







DNMT1_
GATTCCTGGTGCCA
CCGTCACCACTGTTTCTGGCAC
13
13


ED5B_13
GAAACA
CAGG







DNMT1_
GATTCCTGGTGCCA
CGTCACCACTGTTTCTGGCACC
13
12


ED5B_12
GAAACA
AGG







DNMT1_
GATTCCTGGTGCCA
GTCACCACTGTTTCTGGCACCA
13
11


ED5B_11
GAAACA
GG







DNMT1_
GATTCCTGGTGCCA
TCACCACTGTTTCTGGCACCAG
13
10


ED5B_10
GAAACA
G







DNMT1_
GATTCCTGGTGCCA
CACCACTGTTTCTGGCACCAGG
13
 9


ED5B_9
GAAACA








DNMT1_
GATTCCTGGTGCCA
ACCACTGTTTCTGGCACCAGG
13
 8


ED5B_8
GAAACA








RUNX1_
GCATTTTCAGGAGG
AATGACTCAAATATGCTGTCTG
15
31


ED5C_31
AAGCGA
AAGCAATCGCTTCCTCCTGAAA






AT







RUNX1_
GCATTTTCAGGAGG
ATGACTCAAATATGCTGTCTGA
15
30


ED5C_30
AAGCGA
AGCAATCGCTTCCTCCTGAAAA






T







RUNX1_
GCATTTTCAGGAGG
TGACTCAAATATGCTGTCTGAA
15
29


ED5C_29
AAGCGA
GCAATCGCTTCCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
GACTCAAATATGCTGTCTGAAG
15
28


ED5C_28
AAGCGA
CAATCGCTTCCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
ACTCAAATATGCTGTCTGAAGC
15
27


ED5C_27
AAGCGA
AATCGCTTCCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
CTCAAATATGCTGTCTGAAGCA
15
26


ED5C_26
AAGCGA
ATCGCTTCCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
TCAAATATGCTGTCTGAAGCAA
15
25


ED5C_25
AAGCGA
TCGCTTCCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
CAAATATGCTGTCTGAAGCAAT
15
24


ED5C_24
AAGCGA
CGCTTCCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
AAATATGCTGTCTGAAGCAATC
15
23


ED5C_23
AAGCGA
GCTTCCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
AATATGCTGTCTGAAGCAATCG
15
22


ED5C_22
AAGCGA
CTTCCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
ATATGCTGTCTGAAGCAATCGC
15
21


ED5C_21
AAGCGA
TTCCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
TATGCTGTCTGAAGCAATCGCT
15
20


ED5C_20
AAGCGA
TCCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
ATGCTGTCTGAAGCAATCGCTT
15
19


ED5C_19
AAGCGA
CCTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
TGCTGTCTGAAGCAATCGCTTC
15
18


ED5C_18
AAGCGA
CTCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
GCTGTCTGAAGCAATCGCTTCC
15
17


ED5C_17
AAGCGA
TCCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
CTGTCTGAAGCAATCGCTTCCT
15
16


ED5C_16
AAGCGA
CCTGAAAAT







RUNX1_
GCATTTTCAGGAGG
TGTCTGAAGCAATCGCTTCCTC
15
15


ED5C_15
AAGCGA
CTGAAAAT







RUNX1_
GCATTTTCAGGAGG
GTCTGAAGCAATCGCTTCCTCC
15
14


ED5C_14
AAGCGA
TGAAAAT







RUNX1_
GCATTTTCAGGAGG
TCTGAAGCAATCGCTTCCTCCT
15
13


ED5C_13
AAGCGA
GAAAAT







RUNX1_
GCATTTTCAGGAGG
CTGAAGCAATCGCTTCCTCCTG
15
12


ED5C_12
AAGCGA
AAAAT







RUNX1_
GCATTTTCAGGAGG
TGAAGCAATCGCTTCCTCCTGA
15
11


ED5C_11
AAGCGA
AAAT







RUNX1_
GCATTTTCAGGAGG
GAAGCAATCGCTTCCTCCTGAA
15
10


ED5C_10
AAGCGA
AAT







RUNX1_
GCATTTTCAGGAGG
AAGCAATCGCTTCCTCCTGAAA
15
9


ED5C_9
AAGCGA
AT
















TABLE 3N







FIGS. 48A-48C PEgRNA















RT





PBS
TEMPLATE





LENGTH
LENGTH


PEGRNA
SPACER SEQUENCE
3′ EXTENSION SEQUENCE
(NT)
(NT)





HEK3_ED6_
GGCCCAGACTGAGCAC
TCTGCTATCACGTGCTCA
13
10


5GTOA
GTGA (SEQ ID NO: 393)
GTCTG (SEQ ID NO: 394)
















TABLE 30







FIGS. 48A-48C nicking sgRNA








NICKING SGRNA
SPACER SEQUENCE





HEK3_ED6_+63
GCACATACTAGCCCCTGTCT (SEQ ID



NO: 395)
















TABLE 3P







FIGS. 50A-50B PEgRNA















RT





PBS
TEMPLATE



SPACER (SEQ ID
3′ EXTENSION (5′ TO 3′)
LENGTH
LENGTH


PEGRNA
NO: 3699-3754)
(SEQ ID NO: 3755-3810)
(NT)
(NT)





HBB
GTAACGGCAGACTT
AGACTTCTCCTCAGGAGTCAGGTGCA
12
14


3.5
CTCCAC
C







HBB
GCATGGTGCACCTG
AGACTTCTCTTCAGGAGTCAGGTGCA
13
14


3.7
ACTCCTG
C







HBB
GCATGGTGCACCTG
TAACGGCAGACTTCTCCTCAGGAGTC
13
19


5.2
ACTCCTG
AGGTGCAC







HBB
GCATGGTGCACCTG
ACGGCAGACTTCTCCTCAGGAGTCAG
13
17


5.3
ACTCCTG
GTGCAC







HBB
GCATGGTGCACCTG
GGCAGACTTCTCCTCAGGAGTCAGGT
13
16


5.4
ACTCCTG
GCAC







HBB
GCATGGTGCACCTG
GCAGACTTCTCCTCAGGAGTCAGGTG
13
13


5.5
ACTCCTG
CAC







HBB
GCATGGTGCACCTG
GACTTCTCCTCAGGAGTCAGGTGCAC
13
12


5.6
ACTCCTG








HBB
GCATGGTGCACCTG
ACTTCTCCTCAGGAGTCAGGTGCAC
13
21


5.7
ACTCCTG








HBB
GCATGGTGCACCTG
TAACGGCAGACTTCTCCTCAGGAGTC
12
19


5.8
ACTCCTG
AGGTGCA







HBB
GCATGGTGCACCTG
ACGGCAGACTTCTCCTCAGGAGTCAG
12
17


5.9
ACTCCTG
GTGCA







HBB
GCATGGTGCACCTG
GGCAGACTTCTCCTCAGGAGTCAGGT
12
16


5.10
ACTCCTG
GCA







HBB
GCATGGTGCACCTG
GCAGACTTCTCCTCAGGAGTCAGGTG
12
13


5.11
ACTCCTG
CA







HBB
GCATGGTGCACCTG
GACTTCTCCTCAGGAGTCAGGTGCA
12
12


5.12
ACTCCTG








HBB
GCATGGTGCACCTG
ACTTCTCCTCAGGAGTCAGGTGCA
12
14


5.13
ACTCCTG








HEXA
ATCCTTCCAGTCAGG
ATATCTTATGGCCCTGACTGGAA
13
14


S1
GCCAT








HEXA
ATCCTTCCAGTCAGG
TATATCTTATGGCCCTGACTGGAA
13
15


S2
GCCAT








HEXA
ATCCTTCCAGTCAGG
GTATATCTTATGGCCCTGACTGGAA
13
16


S3
GCCAT








HEXA
ATCCTTCCAGTCAGG
ACCGTATATCTTATGGCCCTGACTGGA
13
19


S4
GCCAT
A







HEXA
ATCCTTCCAGTCAGG
AACCGTATATCTTATGGCCCTGACTGG
13
20


S5
GCCAT
AA







HEXA
ATCCTTCCAGTCAGG
GAACCGTATATCTTATGGCCCTGACTG
13
21


S6
GCCAT
GAA







HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCTTATGGCCCTGACT
13
22


S7
GCCAT
GGAA







HEXA
ATCCTTCCAGTCAGG
ATATCTTATGGCCCTGACT
 9
14


S8
GCCAT








HEXA
ATCCTTCCAGTCAGG
TATATCTTATGGCCCTGACT
 9
15


S9
GCCAT








HEXA
ATCCTTCCAGTCAGG
GTATATCTTATGGCCCTGACT
 9
16


S10
GCCAT








HEXA
ATCCTTCCAGTCAGG
ACCGTATATCTTATGGCCCTGACT
 9
19


S11
GCCAT








HEXA
ATCCTTCCAGTCAGG
AACCGTATATCTTATGGCCCTGACT
 9
20


S12
GCCAT








HEXA
ATCCTTCCAGTCAGG
GAACCGTATATCTTATGGCCCTGACT
 9
21


S13
GCCAT








HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCTTATGGCCCTGACT
 9
22


S14
GCCAT








HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCTTATGGCCCTGAC
 8
22


S15
GCCAT








HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCTTATGGCCCTGACT
10
22


S16
GCCAT
G







HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCTTATGGCCCTGACT
11
22


S17
GCCAT
GG







HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCTTATGGCCCTGACT
12
22


S18
GCCAT
GGA







HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCTTATGGCCCTGACT
13
22


S19
GCCAT
GGAA







HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCTTATGGCCCTGACT
14
22


S20
GCCAT
GGAAG







HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCTTATGGCCCTGACT
15
22


S21
GCCAT
GGAAGG







HEXA
ATCCTTCCAGTCAGG
ACCTGAACCGTATATCTTATGGCCCTG
 9
25


S22
GCCAT
ACT







HEXA
ATCCTTCCAGTCAGG
TACCTGAACCGTATATCTTATGGCCCT
 9
26


S23
GCCAT
GACT







HEXA
ATCCTTCCAGTCAGG
GTACCTGAACCGTATATCTTATGGCCC
 9
27


S24
GCCAT
TGACT







HEXA
ATCCTTCCAGTCAGG
GGTACCTGAACCGTATATCTTATGGCC
 9
28


S25
GCCAT
CTGACT







HEXA
ATCCTTCCAGTCAGG
TGGTACCTGAACCGTATATCTTATGGC
 9
29


S26
GCCAT
CCTGACT







HEXA
ATCCTTCCAGTCAGG
ACCTGAACCGTATATCCTATGGCCCTG
13
21


5
GCCAT
ACTGGAA







HEXA
ATCCTTCCAGTCAGG
ACCGTATATCCTATGGCCCTGACTGGA
13
15


6
GCCAT
A







HEXA
ATCCTTCCAGTCAGG
ACCTGAACCGTATATCCTATGGCCCTG
15
21


7
GCCAT
ACTGGAAGG







HEXA
ATCCTTCCAGTCAGG
ACCTGAACCGTATATCCTATGGCCCTG
14
21


8
GCCAT
ACTGGAAG







HEXA
ATCCTTCCAGTCAGG
ACCTGAACCGTATATCCTATGGCCCTG
12
21


9
GCCAT
ACTGGA







HEXA
ATCCTTCCAGTCAGG
ACCTGAACCGTATATCCTATGGCCCTG
11
21


10
GCCAT
ACTGG







HEXA
ATCCTTCCAGTCAGG
ACCTGAACCGTATATCCTATGGCCCTG
10
21


11
GCCAT
ACTG







HEXA
ATCCTTCCAGTCAGG
AACCGTATATCCTATGGCCCTGACTGG
13
16


12
GCCAT
AA







HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCCTATGGCCCTGACT
13
18


13
GCCAT
GGAA







HEXA
ATCCTTCCAGTCAGG
TACCTGAACCGTATATCCTATGGCCCT
13
22


14
GCCAT
GACTGGAA







HEXA
ATCCTTCCAGTCAGG
TGGTACCTGAACCGTATATCCTATGGC
13
25


15
GCCAT
CCTGACTGGAA







HEXA
ATCCTTCCAGTCAGG
GTACCTGAACCGTATATCCTATGGCCC
13
23


16
GCCAT
TGACTGGAA







HEXA
ATCCTTCCAGTCAGG
AACCGTATATCCTATGGCCCTGACTG
10
16


17
GCCAT








HEXA
ATCCTTCCAGTCAGG
TGAACCGTATATCCTATGGCCCTGACT
10
18


18
GCCAT
G







HEXA
ATCCTTCCAGTCAGG
TACCTGAACCGTATATCCTATGGCCCT
10
22


19
GCCAT
GACTG







HEXA
ATCCTTCCAGTCAGG
TGGTACCTGAACCGTATATCCTATGGC
10
25


20
GCCAT
CCTGACTG
















TABLE 3Q







FIGS. 50A-50B nicking sgRNA








NICKING SGRNA
SPACER SEQUENCE





HBB_ED7A_+72
GCCTTGATACCAACCTGCCCA



(SEQ ID NO: 626)





HEXA_ED7B_+60
GCTGGAACTGGTCACCAAGGC



(SEQ ID NO: 627)





HEXA_ED7B_CORRECT_
GTACCTGAACCGTATATCCTA


WT_PE3B
(SEQ ID NO: 628)





HEXA_ED7B_CORRECT_
GTACCTGAACCGTATATCTTA


SILENT_PE3B
(SEQ ID NO: 629)
















TABLE 3R







FIGS. 51A-51G PEgRNA















RT





PBS
TEMPLATE



SPACER SEQUENCE
3′ EXTENSION (SEQ 
LENGTH
LENGTH


PEGRNA
(SEQ ID NO: 632-640)
ID NO: 641-649)
(NT)
(NT)





HEK3_ED8_
GGCCCAGACTGAGCA
TCTGCCATCCCGTGCTCAGT
13
10


1TTOG
CGTGA
CTG







HEK3_ED8_
GGCCCAGACTGAGCA
TCTGCCAGCACGTGCTCAGT
13
10


3ATOC
CGTGA
CTG







HEK3_ED8_
GGCCCAGACTGAGCA
TCTGCCAACACGTGCTCAGT
13
10


3ATOT
CGTGA
CTG







HEK3_ED8_
GGCCCAGACTGAGCA
TGGAGGAAGCAGGGCTTCC
13
34


3ATOT_5-
CGTGA
TTTCCTCTGAAAACACGTGC




6GGTOTT

TCAGTCTG







HEK3_ED8_
GGCCCAGACTGAGCA
TCTGCCATCAAAGCGTGCTC
13
10


1CTTINS
CGTGA
AGTCTG







RNF2_ED8_
GTCATCTTAGTCATTA
AACGAACACCTCATGTAATG
15
14


1CTOA
CCTG
ACTAAGATG







RNF2_ED8_
GTCATCTTAGTCATTA
AACGAACACCTCACGTAATG
15
14


1CTOG
CCTG
ACTAAGATG







RNF2_ED8_
GTCATCTTAGTCATTA
AACGAACACCTCAGTACGTA
15
17


1GTAINS
CCTG
ATGACTAAGATG







HBB_ED8_
GCATGGTGCACCTGAC
AGACTTCTCCACAGGAGTCA
13
14


4ATOT
TCCTG
GGTGCAC
















TABLE 4







Sequences of primers used for mammalian cell genomic DNA amplification


and HTS181.








DESCRIPTION
SEQUENCE (SEQ ID NOS: 3811-3863)





HEK3 FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNATGTGGGCT



GCCTAGAAAGG





HEK3 REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTCCCAGCCAAACTTGTCA



ACC





RNF2 FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNACGTCTCAT



ATGCCCCTTGG





RNF2 REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTACGTAGGAATTTTGGTG



GGACA





HEK4 FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGAACCCAG



GTAGCCAGAGAC





HEK4 REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTTCCTTTCAACCCGAACG



GAG





EMX1 FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNCAGCTCAGC



CTGAGTGTTGA





EMX1 REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTCTCGTGGGTTTGTGGTT



GC





FANCF FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNCATTGCAGA



GAGGCGTATCA





FANCF REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTGGGGTCCCAGGTGCTG



AC





HBB FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNAGGGTTGGC



CAATCTACTCCC





HBB REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTGTCTTCTCTGTCTCCAC



ATGCC





PRNP FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGTCAGTGGA



ACAAGCCGAGT





PRNP REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTACTTGGTTGGGGTAACG



GTG





HEXA FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNCATACAGGT



GTGGCGAGAGG





HEXA REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTCCAGCCTCCTTTGGTTA



GCA





RUNX1 FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNTCACAAACA



AGACAGGGAACTG





RUNX1 REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTAGATGTAGGGCTAGAGG



GGTG





VEGFA FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNACTTGGTGC



CAAATTCTTCTCC





VEGFA REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTAAAGAGGGAATGGGCT



TTGGA





DNMT FWD
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNCACAACAG



CTTCATGTCAGCC





DNMT REV
TGGAGTTCAGACGTGTGCTCTTCCGATCTACGTTAATGTTTCCTGAT



GGTCC


HEK3 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNTCCCCTGTT


SITE 1 FWD
GACCTGGAGAA





HEK3 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTCACTGTACTTGCCCTGA


SITE 1 REV
CCA





HEK3 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNTTGGTGTTG


SITE 2 FWD
ACAGGGAGCAA





HEK3 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTCTGAGATGTGGGCAGA


SITE 2 REV
AGGG





HEK3 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNTGAGAGGG


SITE 3 FWD
AACAGAAGGGCT





HEK3 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTGTCCAAAGGCCCAAGA


SITE 3 REV
ACCT





HEK3 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNTCCTAGCAC


SITE 4 FWD
TTTGGAAGGTCG





HEK3 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTGCTCATCTTAATCTGCT


SITE 4 REV
CAGCC





HEK4 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGGCATGGCT


SITE 1 FWD
TCTGAGACTCA





HEK4 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTGTCTCCCTTGCACTCCC


SITE 1 REV
TGTCTTT





HEK4 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNTTTGGCAAT


SITE 2 FWD
GGAGGCATTGG





HEK4 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTGAAGAGGCTGCCCATG


SITE 2 REV
AGAG





HEK4 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGGTCTGAGG


SITE 3 FWD
CTCGAATCCTG





HEK4 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTCTGTGGCCTCCATATCC


SITE 3 REV
CTG





HEK4 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNTTTCCACCA


SITE 4 FWD
GAACTCAGCCC





HEK4 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTCCTCGGTTCCTCCACAA


SITE 4 REV
CAC





EMX1 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGTGGGGAG


SITE 1 FWD
ATTTGCATCTGTGGAGG





EMX1 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTGCTTTTATACCATCTTGG


SITE 1 REV
GGTTACAG





EMX1 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNCAATGTGCT


SITE 2 FWD
TCAACCCATCACGGC





EMX1 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTCCATGAATTTGTGATGG


SITE 2 REV
ATGCAGTCTG





EMX1 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGAGAAGGA


SITE 3 FWD
GGTGCAGGAGCTAGAC





EMX1 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTCATCCCGACCTTCATCC


SITE 3 REV
CTCCTGG





EMX1 OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGTAGTTCTG


SITE 4 FWD
ACATTCCTCCTGAGGG





EMX1 OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTTCAAACAAGGTGCAGA


SITE 4 REV
TACAGCA





FANCF OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGCGGGCAG


SITE 1 FWD
TGGCGTCTTAGTCG





FANCF OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTCCCTGGGTTTGGTTGGC


SITE 1 REV
TGCTC





FANCF OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNCTCCTTGCC


SITE 2 FWD
GCCCAGCCGGTC





FANCF OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTCACTGGGGAAGAGGCG


SITE 2 REV
AGGACAC





FANCF OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNCCAGTGTTT


SITE 3 FWD
CCCATCCCCAACAC





FANCF OFF-TARGET
TGGAGTTCAGACGTGTGCTCTTCCGATCTGAATGGATCCCCCCCTA


SITE 3 REV
GAGCTC





FANCF OFF-TARGET
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNCAGGCCCAC


SITE 4 FWD
AGGTCCTTCTGGA
















TABLE 5





Sequences of 100-mer single-stranded DNA oligonucleotide donor templates used


in HDR experiments and in the creation of the HBB E6V HEK293T cell line.


Oligonucleotides are 100-103 nt in length with homology arms centered around


the site of the edit. Oligonucleotides were from Integrated DNA Technologies,


purified by PAGE.
















HEK3 +3 A TOT
GCTTCTCCAGCCCTGGCCTGGGTCAATCCTTGGGGCCCAGACTG



AGCACGTGTTGGCAGAGGAAAGGAAGCCCTGCTTCCTCCAGAG



GGCGTCGCAGGAC (SEQ ID NO: 717)





HEK3 +3 A TO T, +5,6 GG
GCTTCTCCAGCCCTGGCCTGGGTCAATCCTTGGGGCCCAGACTG


TO TT
AGCACGTGTTTTCAGAGGAAAGGAAGCCCTGCTTCCTCCAGAG



GGCGTCGCAGGAC (SEQ ID NO: 718)





HEK3 +1 TTOG
GCTTCTCCAGCCCTGGCCTGGGTCAATCCTTGGGGCCCAGACTG



AGCACGGGATGGCAGAGGAAAGGAAGCCCTGCTTCCTCCAGA



GGGCGTCGCAGGAC (SEQ ID NO: 719)





HEK3 +3 A TOC
GCTTCTCCAGCCCTGGCCTGGGTCAATCCTTGGGGCCCAGACTG



AGCACGTGCTGGCAGAGGAAAGGAAGCCCTGCTTCCTCCAGA



GGGCGTCGCAGGAC (SEQ ID NO: 720)





HEK3 +1 CTT
GCTTCTCCAGCCCTGGCCTGGGTCAATCCTTGGGGCCCAGACTG


INSERTION
AGCACGCTTTGATGGCAGAGGAAAGGAAGCCCTGCTTCCTCCA



GAGGGCGTCGCAGGAC (SEQ ID NO: 721)





RNF2 +1 CTOA
CCCAGTTTACACGTCTCATATGCCCCTTGGCAGTCATCTTAGTCA



TTACATGAGGTGTTCGTTGTAACTCATATAAACTGAGTTCCCATG



TTTTGCTTAA (SEQ ID NO: 722)





RNF2 +1 CTOG
CCCAGTTTACACGTCTCATATGCCCCTTGGCAGTCATCTTAGTCA



TTACGTGAGGTGTTCGTTGTAACTCATATAAACTGAGTTCCCATG



TTTTGCTTAA (SEQ ID NO: 723)





RNF2 +1 GTA
CAGTTTACACGTCTCATATGCCCCTTGGCAGTCATCTTAGTCATT


INSERTION
ACGTACTGAGGTGTTCGTTGTAACTCATATAAACTGAGTTCCCAT



GTTTTGCTTA (SEQ ID NO: 724)





HBB E6V INSTALLATION
ACTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAG


(ALSO USED FOR
ACTTCTCCACAGGAGTCAGATGCACCATGGTGTCTGTTTGAGGT


CREATION OF THE HBB
TGCTAGTGAACAC (SEQ ID NO: 725)


E6V HEK293T CELL



LINE)






HBB E6V CORRECTION
ACTTCATCCACGTTCACCTTGCCCCACAGGGCAGTAACGGCAG


PROTOSPACER A
ACTTCTCCTCAGGAGTCAGGTGCACCATGGTGTCTGTTTGAGGT



TGCTAGTGAACAC (SEQ ID NO: 726)





HBB E6V CORRECTION
GTGTTCACTAGCAACCTCAAACAGACACCATGGTGCACCTGAC


PROTOSPACER B
TCCTGAGGAGAAGTCTGCCGTTACTGCCCTGTGGGGCAAGGTG



AACGTGGATGAAGT (SEQ ID NO: 727)





HBB E6V CORRECTION
GTGTTCACTAGCAACCTCAAACAGACACCATGGTGCACCTGAC


PROTOSPACER B,
TCCTGATGAGAAGTCTGCCGTTACTGCCCTGTGGGGCAAGGTG


SILENT PAM MUTATION
AACGTGGATGAAGT (SEQ ID NO: 728)





PRNP G127V
CACATGGCTGGTGCTGCAGCAGCTGGGGCAGTGGTGGGGGGCC



TTGGCGTCTACATGCTGGGAAGTGCCATGAGCAGGCCCATCATA



CATTTCGGCAGTG (SEQ ID NO: 729)









Additional Sequences


Sequences of Yeast Dual Fluorescent Reporter Plasmids Used Herein










p425-GFP_stop_mCherry:



(SEQ ID NO: 730)



ATGTCTAAAGGTGAAGAATTATTCACTGGTGTTGTCCCAATTTTGGTTGAATTAGATGGTGATG






TTAATGGTCACAAATTTTCTGTCTCCGGTGAAGGTGAAGGTGATGCTACTTACGGTAAATTGA





CCTTAAAATTTATTTGTACTACTGGTAAATTGCCAGTTCCATGGCCAACCTTAGTCACTACTTTC





GGTTATGGTGTTCAATGTTTTGCTAGATACCCAGATCATATGAAACAACATGACTTTTTCAAGT





CTGCCATGCCAGAAGGTTATGTTCAAGAAAGAACTATTTTTTTCAAAGATGACGGTAACTACA





AGACCAGAGCTGAAGTCAAGTTTGAAGGTGATACCTTAGTTAATAGAATCGAATTAAAAGGTA





TTGATTTTAAAGAAGATGGTAACATTTTAGGTCACAAATTGGAATACAACTATAACTCTCACAA





TGTTTACATCATGGCTGACAAACAAAAGAATGGTATCAAAGTTAACTTCAAAATTAGACACAA





CATTGAAGATGGTTCTGTTCAATTAGCTGACCATTATCAACAAAATACTCCAATTGGTGATGGT





CCAGTCTTGTTACCAGACAACCATTACTTATCCACTCAATCTGCCTTATCCAAAGATCCAAACG





AAAAGAGAGACCACATGGTCTTGTTAGAATTTGTTACTGCTGCTGGTATTACCCATGGTATGGA





TGAATTGTACAAAGCTAGCAACCTGGGTCAATCCTTGGGGCCCAGACTGAGCACGTGATGG





CAGAGCACAGGAGACGTCATGGTTTCAAAAGGTGAAGAAGATAATATGGCTATTATTAAAGAATTTA






TGAGATTTAAAGTTCATATGGAAGGTTCAGTTAATGGTCATGAATTTGAAATTGAAGGTGAAGGTGAA







GGTAGACCATATGAAGGTACTCAAACTGCTAAATTGAAAGTTACTAAAGGTGGTCCATTACCATTTGC







TTGGGATATTTTGTCACCACAATTTATGTATGGTTCAAAAGCTTATGTTAAACATCCAGCTGATATTCCA







GATTATTTAAAATTGTCATTTCCAGAAGGTTTTAAATGGGAAAGAGTTATGAATTTTGAAGATGGTGGT







GTTGTTACTGTTACTCAAGATTCATCATTACAAGATGGTGAATTTATTTATAAAGTTAAATTGAGAGGTA







CTAATTTTCCATCAGATGGTCCAGTTATGCAAAAAAAAACTATGGGTTGGGAAGCTTCATCAGAAAGA







ATGTATCCAGAAGATGGTGCTTTAAAAGGTGAAATTAAACAAAGATTGAAATTAAAAGATGGTGGTCA







TTATGATGCTGAAGTTAAAACTACTTATAAAGCTAAAAAACCAGTTCAATTACCAGGTGCTTATAATGTT







AATATTAAATTGGATATTACTTCACATAATGAAGATTATACTATTGTTGAACAATATGAAAGAGCTGAAG







GTAGACATTCAACTGGTGGTATGGATGAATTATATAAAGGTACCGCTCGAGCAGCTGTGATTGATTGA






GTCGACTTGGTTGAACACGTTGCCAAGGCTTAAGTGAATTTACTTTAAATCTTGCATTTAAATA





AATTTTCTTTTTATAGCTTTATGACTTAGTTTCAATTTATATACTATTTTAATGACATTTTCGATTC





GGATCCACTAGTTCTAGAGCGGCCGCCACCGCGGTGGAGCTCCAGCTTTTGTTCCCTTTAGTG





AGGGTTAATTGCGCGCTTGGCGTAATCATGGTCATAGCTGTTTCCTGTGTGAAATTGTTATCCG





CTCACAATTCCACACAACATAGGAGCCGGAAGCATAAAGTGTAAAGCCTGGGGTGCCTAATG





AGTGAGGTAACTCACATTAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTC





GTGCCAGCTGCATTAATGAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCT





CTTCCGCTTCCTCGCTCACTGACTCGCTGCGCTCGGTCGTTCGGCTGCGGCGAGCGGTATCAG





CTCACTCAAAGGCGGTAATACGGTTATCCACAGAATCAGGGGATAACGCAGGAAAGAACATG





TGAGCAAAAGGCCAGCAAAAGGCCAGGAACCGTAAAAAGGCCGCGTTGCTGGCGTTTTTCC





ATAGGCTCCGCCCCCCTGACGAGCATCACAAAAATCGACGCTCAAGTCAGAGGTGGCGAAAC





CCGACAGGACTATAAAGATACCAGGCGTTTCCCCCTGGAAGCTCCCTCGTGCGCTCTCCTGTT





CCGACCCTGCCGCTTACCGGATACCTGTCCGCCTTTCTCCCTTCGGGAAGCGTGGCGCTTTCT





CATAGCTCACGCTGTAGGTATCTCAGTTCGGTGTAGGTCGTTCGCTCCAAGCTGGGCTGTGTG





CACGAACCCCCCGTTCAGCCCGACCGCTGCGCCTTATCCGGTAACTATCGTCTTGAGTCCAAC





CCGGTAAGACACGACTTATCGCCACTGGCAGCAGCCACTGGTAACAGGATTAGCAGAGCGAG





GTATGTAGGCGGTGCTACAGAGTTCTTGAAGTGGTGGCCTAACTACGGCTACACTAGAAGGAC





AGTATTTGGTATCTGCGCTCTGCTGAAGCCAGTTACCTTCGGAAAAAGAGTTGGTAGCTCTTG





ATCCGGCAAACAAACCACCGCTGGTAGCGGTGGTTTTTTTGTTTGCAAGCAGCAGATTACGCG





CAGAAAAAAAGGATCTCAAGAAGATCCTTTGATCTTTTCTACGGGGTCTGACGCTCAGTGGA





ACGAAAACTCACGTTAAGGGATTTTGGTCATGAGATTATCAAAAAGGATCTTCACCTAGATCC





TTTTAAATTAAAAATGAAGTTTTAAATCAATCTAAAGTATATATGAGTAAACTTGGTCTGACAG





TTACCAATGCTTAATCAGTGAGGCACCTATCTCAGCGATCTGTCTATTTCGTTCATCCATAGTTG





CCTGACTCCCCGTCGTGTAGATAACTACGATACGGGAGGGCTTACCATCTGGCCCCAGTGCTG





CAATGATACCGCGAGACCCACGCTCACCGGCTCCAGATTTATCAGCAATAAACCAGCCAGCCG





GAAGGGCCGAGCGCAGAAGTGGTCCTGCAACTTTATCCGCCTCCATCCAGTCTATTAATTGTT





GCCGGGAAGCTAGAGTAAGTAGTTCGCCAGTTAATAGTTTGCGCAACGTTGTTGCCATTGCTA





CAGGCATCGTGGTGTCACGCTCGTCGTTTGGTATGGCTTCATTCAGCTCCGGTTCCCAACGATC





AAGGCGAGTTACATGATCCCCCATGTTGTGCAAAAAAGCGGTTAGCTCCTTCGGTCCTCCGAT





CGTTGTCAGAAGTAAGTTGGCCGCAGTGTTATCACTCATGGTTATGGCAGCACTGCATAATTCT





CTTACTGTCATGCCATCCGTAAGATGCTTTTCTGTGACTGGTGAGTACTCAACCAAGTCATTCT





GAGAATAGTGTATGCGGCGACCGAGTTGCTCTTGCCCGGCGTCAATACGGGATAATACCGCGC





CACATAGCAGAACTTTAAAAGTGCTCATCATTGGAAAACGTTCTTCGGGGCGAAAACTCTCA





AGGATCTTACCGCTGTTGAGATCCAGTTCGATGTAACCCACTCGTGCACCCAACTGATCTTCA





GCATCTTTTACTTTCACCAGCGTTTCTGGGTGAGCAAAAACAGGAAGGCAAAATGCCGCAAA





AAAGGGAATAAGGGCGACACGGAAATGTTGAATACTCATACTCTTCCTTTTTCAATATTATTGA





AGCATTTATCAGGGTTATTGTCTCATGAGCGGATACATATTTGAATGTATTTAGAAAAATAAACA





AATAGGGGTTCCGCGCACATTTCCCCGAAAAGTGCCACCTGAACGAAGCATCTGTGCTTCATT





TTGTAGAACAAAAATGCAACGCGAGAGCGCTAATTTTTCAAACAAAGAATCTGAGCTGCATTT





TTACAGAACAGAAATGCAACGCGAAAGCGCTATTTTACCAACGAAGAATCTGTGCTTCATTTT





TGTAAAACAAAAATGCAACGCGAGAGCGCTAATTTTTCAAACAAAGAATCTGAGCTGCATTTT





TACAGAACAGAAATGCAACGCGAGAGCGCTATTTTACCAACAAAGAATCTATACTTCTTTTTT





GTTCTACAAAAATGCATCCCGAGAGCGCTATTTTTCTAACAAAGCATCTTAGATTACTTTTTTTC





TCCTTTGTGCGCTCTATAATGCAGTCTCTTGATAACTTTTTGCACTGTAGGTCCGTTAAGGTTAG





AAGAAGGCTACTTTGGTGTCTATTTTCTCTTCCATAAAAAAAGCCTGACTCCACTTCCCGCGTT





TACTGATTACTAGCGAAGCTGCGGGTGCATTTTTTCAAGATAAAGGCATCCCCGATTATATTCT





ATACCGATGTGGATTGCGCATACTTTGTGAACAGAAAGTGATAGCGTTGATGATTCTTCATTGG





TCAGAAAATTATGAACGGTTTCTTCTATTTTGTCTCTATATACTACGTATAGGAAATGTTTACATT





TTCGTATTGTTTTCGATTCACTCTATGAATAGTTCTTACTACAATTTTTTTGTCTAAAGAGTAATA





CTAGAGATAAACATAAAAAATGTAGAGGTCGAGTTTAGATGCAAGTTCAAGGAGCGAAAGGT





GGATGGGTAGGTTATATAGGGATATAGCACAGAGATATATAGCAAAGAGATACTTTTGAGCAAT





GTTTGTGGAAGCGGTATTCGCAATATTTTAGTAGCTCGTTACAGTCCGGTGCGTTTTTGGTTTT





TTGAAAGTGCGTCTTCAGAGCGCTTTTGGTTTTCAAAAGCGCTCTGAAGTTCCTATACTTTCTA





GAGAATAGGAACTTCGGAATAGGAACTTCAAAGCGTTTCCGAAAACGAGCGCTTCCGAAAAT





GCAACGCGAGCTGCGCACATACAGCTCACTGTTCACGTCGCACCTATATCTGCGTGTTGCCTG





TATATATATATACATGAGAAGAACGGCATAGTGCGTGTTTATGCTTAAATGCGTACTTATATGCGT





CTATTTATGTAGGATGAAAGGTAGTCTAGTACCTCCTGTGATATTATCCCATTCCATGCGGGGTA





TCGTATGCTTCCTTCAGCACTACCCTTTAGCTGTTCTATATGCTGCCACTCCTCAATTGGATTAG





TCTCATCCTTCAATGCTATCATTTCCTTTGATATTGGATCATACTAAGAAACCATTATTATCATGA





CATTAACCTATAAAAATAGGCGTATCACGAGGCCCTTTCGTCTCGCGCGTTTCGGTGATGACGG





TGAAAACCTCTGACACATGCAGCTCCCGGAGACGGTCACAGCTTGTCTGTAAGCGGATGCCG





GGAGCAGACAAGCCCGTCAGGGCGCGTCAGCGGGTGTTGGCGGGTGTCGGGGCTGGCTTAA





CTATGCGGCATCAGAGCAGATTGTACTGAGAGTGCACCATATCGACTACGTCGTAAGGCCGTT





TCTGACAGAGTAAAATTCTTGAGGGAACTTTCACCATTATGGGAAATGCTTCAAGAAGGTATT





GACTTAAACTCCATCAAATGGTCAGGTCATTGAGTGTTTTTTATTTGTTGTATTTTTTTTTTTTTA





GAGAAAATCCTCCAATATCAAATTAGGAATCGTAGTTTCATGATTTTCTGTTACACCTAACTTTT





TGTGTGGTGCCCTCCTCCTTGTCAATATTAATGTTAAAGTGCAATTCTTTTTCCTTATCACGTTG





AGCCATTAGTATCAATTTGCTTACCTGTATTCCTTTACTATCCTCCTTTTTCTCCTTCTTGATAAA





TGTATGTAGATTGCGTATATAGTTTCGTCTACCCTATGAACATATTCCATTTTGTAATTTCGTGTC





GTTTCTATTATGAATTTCATTTATAAAGTTTATGTACAAATATCATAAAAAAAGAGAATCTTTTTA





AGCAAGGATTTTCTTAACTTCTTCGGCGACAGCATCACCGACTTCGGTGGTACTGTTGGAACC





ACCTAAATCACCAGTTCTGATACCTGCATCCAAAACCTTTTTAACTGCATCTTCAATGGCCTTA





CCTTCTTCAGGCAAGTTCAATGACAATTTCAACATCATTGCAGCAGACAAGATAGTGGCGATA





GGGTCAACCTTATTCTTTGGCAAATCTGGAGCAGAACCGTGGCATGGTTCGTACAAACCAAAT





GCGGTGTTCTTGTCTGGCAAAGAGGCCAAGGACGCAGATGGCAACAAACCCAAGGAACCTG





GGATAACGGAGGCTTCATCGGAGATGATATCACCAAACATGTTGCTGGTGATTATAATACCATT





TAGGTGGGTTGGGTTCTTAACTAGGATCATGGCGGCAGAATCAATCAATTGATGTTGAACCTTC





AATGTAGGGAATTCGTTCTTGATGGTTTCCTCCACAGTTTTTCTCCATAATCTTGAAGAGGCCA





AAAGATTAGCTTTATCCAAGGACCAAATAGGCAATGGTGGCTCATGTTGTAGGGCCATGAAAG





CGGCCATTCTTGTGATTCTTTGCACTTCTGGAACGGTGTATTGTTCACTATCCCAAGCGACACC





ATCACCATCGTCTTCCTTTCTCTTACCAAAGTAAATACCTCCCACTAATTCTCTGACAACAACG





AAGTCAGTACCTTTAGCAAATTGTGGCTTGATTGGAGATAAGTCTAAAAGAGAGTCGGATGCA





AAGTTACATGGTCTTAAGTTGGCGTACAATTGAAGTTCTTTACGGATTTTTAGTAAACCTTGTT





CAGGTCTAACACTACCGGTACCCCATTTAGGACCAGCCACAGCACCTAACAAAACGGCATCA





ACCTTCTTGGAGGCTTCCAGCGCCTCATCTGGAAGTGGGACACCTGTAGCATCGATAGCAGCA





CCACCAATTAAATGATTTTCGAAATCGAACTTGACATTGGAACGAACATCAGAAATAGCTTTA





AGAACCTTAATGGCTTCGGCTGTGATTTCTTGACCAACGTGGTCACCTGGCAAAACGACGATC





TTCTTAGGGGCAGACATAGGGGCAGACATTAGAATGGTATATCCTTGAAATATATATATATATTG





CTGAAATGTAAAAGGTAAGAAAAGTTAGAAAGTAAGACGATTGCTAACCACCTATTGGAAAA





AACAATAGGTCCTTAAATAATATTGTCAACTTCAAGTATTGTGATGCAAGCATTTAGTCATGAA





CGCTTCTCTATTCTATATGAAAAGCCGGTTCCGGCCTCTCACCTTTCCTTTTTCTCCCAATTTTT





CAGTTGAAAAAGGTATATGCGTCAGGCGACCTCTGAAATTAACAAAAAATTTCCAGTCATCGA





ATTTGATTCTGTGCGATAGCGCCCCTGTGTGTTCTCGTTATGTTGAGGAAAAAAATAATGGTTG





CTAAGAGATTCGAACTCTTGCATCTTACGATACCTGAGTATTCCCACAGTTAACTGCGGTCAAG





ATATTTCTTGAATCAGGCGCCTTAGACCGCTCGGCCAAACAACCAATTACTTGTTGAGAAATA





GAGTATAATTATCCTATAAATATAACGTTTTTGAACACACATGAACAAGGAAGTACAGGACAAT





TGATTTTGAAGAGAATGTGGATTTTGATGTAATTGTTGGGATTCCATTTTTAATAAGGCAATAAT





ATTAGGTATGTGGATATACTAGAAGTTCTCCTCGACCGTCGATATGCGGTGTGAAATACCGCAC





AGATGCGTAAGGAGAAAATACCGCATCAGGAAATTGTAAACGTTAATATTTTGTTAAAATTCGC





GTTAAATTTTTGTTAAATCAGCTCATTTTTTAACCAATAGGCCGAAATCGGCAAAATCCCTTATA





AATCAAAAGAATAGACCGAGATAGGGTTGAGTGTTGTTCCAGTTTGGAACAAGAGTCCACTAT





TAAAGAACGTGGACTCCAACGTCAAAGGGCGAAAAACCGTCTATCAGGGCGATGGCCCACTA





CGTGAACCATCACCCTAATCAAGTTTTTTGGGGTCGAGGTGCCGTAAAGCACTAAATCGGAAC





CCTAAAGGGAGCCCCCGATTTAGAGCTTGACGGGGAAAGCCGGCGAACGTGGCGAGAAAGG





AAGGGAAGAAAGCGAAAGGAGCGGGCGCTAGGGCGCTGGCAAGTGTAGCGGTCACGCTGC





GCGTAACCACCACACCCGCCGCGCTTAATGCGCCGCTACAGGGCGCGTCGCGCCATTCGCCAT





TCAGGCTGCGCAACTGTTGGGAAGGGCGATCGGTGCGGGCCTCTTCGCTATTACGCCAGCTG





GCGAAAGGGGGATGTGCTGCAAGGCGATTAAGTTGGGTAACGCCAGGGTTTTCCCAGTCACG





ACGTTGTAAAACGACGGCCAGTGAGCGCGCGTAATACGACTCACTATAGGGCGAATTGGGTA





CCGGGCCCCCCCTCGAGGTCGACGGTATCGATAAGCTTGATATCGAATTCCTGCAGCCCGGGG





GATCCGTTAGAATCATTTTGAATAAAAAACACGCTTTTTCAGTTCGAGTTTATCATTATCAATAC





TGCCATTTCAAAGAATACGTAAATAATTAATAGTAGTGATTTTCCTAACTTTATTTAGTCAAAAA





ATTAGCCTTTTAATTCTGCTGTAACCCGTACATGCCCAAAATAGGGGGCGGGTTACACAGAATA





TATAACATCGTAGGTGTCTGGGTGAACAGTTTATTCCTGGCATCCACTAAATATAATGGAGCCC





GCTTTTTAAGCTGGCATCCAGAAAAAAAAAGAATCCCAGCACCAAAATATTGTTTTCTTCACC





AACCATCAGTTCATAGGTCCATTCTCTTAGCGCAACTACAGAGAACAGGGGCACAAACAGGC





AAAAAACGGGCACAACCTCAATGGAGTGATGCAACCTGCCTGGAGTAAATGATGACACAAGG





CAATTGACCCACGCATGTATCTATCTCATTTTCTTACACCTTCTATTACCTTCTGCTCTCTCTGAT





TTGGAAAAAGCTGAAAAAAAAGGTTGAAACCAGTTCCCTGAAATTATTCCCCTACTTGACTA





ATAAGTATATAAAGACGGTAGGTATTGATTGTAATTCTGTAAATCTATTTCTTAAACTTCTTAAAT





TCTACTTTTATAGTTAGTCTTTTTTTTAGTTTTAAAACACCAAGAACTTAGTTTCGAATAAACAC





ACATAAACAAACAAAGAATTC





p425-GFP_+1fs_mCherry:


(SEQ ID NO: 731)



ATGTCTAAAGGTGAAGAATTATTCACTGGTGTTGTCCCAATTTTGGTTGAATTAGATGGTGATG






TTAATGGTCACAAATTTTCTGTCTCCGGTGAAGGTGAAGGTGATGCTACTTACGGTAAATTGA





CCTTAAAATTTATTTGTACTACTGGTAAATTGCCAGTTCCATGGCCAACCTTAGTCACTACTTTC





GGTTATGGTGTTCAATGTTTTGCTAGATACCCAGATCATATGAAACAACATGACTTTTTCAAGT





CTGCCATGCCAGAAGGTTATGTTCAAGAAAGAACTATTTTTTTCAAAGATGACGGTAACTACA





AGACCAGAGCTGAAGTCAAGTTTGAAGGTGATACCTTAGTTAATAGAATCGAATTAAAAGGTA





TTGATTTTAAAGAAGATGGTAACATTTTAGGTCACAAATTGGAATACAACTATAACTCTCACAA





TGTTTACATCATGGCTGACAAACAAAAGAATGGTATCAAAGTTAACTTCAAAATTAGACACAA





CATTGAAGATGGTTCTGTTCAATTAGCTGACCATTATCAACAAAATACTCCAATTGGTGATGGT





CCAGTCTTGTTACCAGACAACCATTACTTATCCACTCAATCTGCCTTATCCAAAGATCCAAACG





AAAAGAGAGACCACATGGTCTTGTTAGAATTTGTTACTGCTGCTGGTATTACCCATGGTATGGA





TGAATTGTACAAAGCTAGCAAACCTGGGTCAATCCTTGGGGCCCAGACTGAGCACGTGATG






GCAGAGCACAGGAGACGTCATGGTTTCAAAAGGTGAAGAAGATAATATGGCTATTATTAAAGAATTT







ATGAGATTTAAAGTTCATATGGAAGGTTCAGTTAATGGTCATGAATTTGAAATTGAAGGTGAAGGTGA







AGGTAGACCATATGAAGGTACTCAAACTGCTAAATTGAAAGTTACTAAAGGTGGTCCATTACCATTTG







CTTGGGATATTTTGTCACCACAATTTATGTATGGTTCAAAAGCTTATGTTAAACATCCAGCTGATATTCC







AGATTATTTAAAATTGTCATTTCCAGAAGGTTTTAAATGGGAAAGAGTTATGAATTTTGAAGATGGTGG







TGTTGTTACTGTTACTCAAGATTCATCATTACAAGATGGTGAATTTATTTATAAAGTTAAATTGAGAGGT







ACTAATTTTCCATCAGATGGTCCAGTTATGCAAAAAAAAACTATGGGTTGGGAAGCTTCATCAGAAAG







AATGTATCCAGAAGATGGTGCTTTAAAAGGTGAAATTAAACAAAGATTGAAATTAAAAGATGGTGGTC







ATTATGATGCTGAAGTTAAAACTACTTATAAAGCTAAAAAACCAGTTCAATTACCAGGTGCTTATAATGT







TAATATTAAATTGGATATTACTTCACATAATGAAGATTATACTATTGTTGAACAATATGAAAGAGCTGAAG







GTAGACATTCAACTGGTGGTATGGATGAATTATATAAAGGTACCGCTCGAGCAGCTGTGATTGATTGA






GTCGACTTGGTTGAACACGTTGCCAAGGCTTAAGTGAATTTACTTTAAATCTTGCATTTAAATA





AATTTTCTTTTTATAGCTTTATGACTTAGTTTCAATTTATATACTATTTTAATGACATTTTCGATTC





GGATCCACTAGTTCTAGAGCGGCCGCCACCGCGGTGGAGCTCCAGCTTTTGTTCCCTTTAGTG





AGGGTTAATTGCGCGCTTGGCGTAATCATGGTCATAGCTGTTTCCTGTGTGAAATTGTTATCCG





CTCACAATTCCACACAACATAGGAGCCGGAAGCATAAAGTGTAAAGCCTGGGGTGCCTAATG





AGTGAGGTAACTCACATTAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTC





GTGCCAGCTGCATTAATGAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCT





CTTCCGCTTCCTCGCTCACTGACTCGCTGCGCTCGGTCGTTCGGCTGCGGCGAGCGGTATCAG





CTCACTCAAAGGCGGTAATACGGTTATCCACAGAATCAGGGGATAACGCAGGAAAGAACATG





TGAGCAAAAGGCCAGCAAAAGGCCAGGAACCGTAAAAAGGCCGCGTTGCTGGCGTTTTTCC





ATAGGCTCCGCCCCCCTGACGAGCATCACAAAAATCGACGCTCAAGTCAGAGGTGGCGAAAC





CCGACAGGACTATAAAGATACCAGGCGTTTCCCCCTGGAAGCTCCCTCGTGCGCTCTCCTGTT





CCGACCCTGCCGCTTACCGGATACCTGTCCGCCTTTCTCCCTTCGGGAAGCGTGGCGCTTTCT





CATAGCTCACGCTGTAGGTATCTCAGTTCGGTGTAGGTCGTTCGCTCCAAGCTGGGCTGTGTG





CACGAACCCCCCGTTCAGCCCGACCGCTGCGCCTTATCCGGTAACTATCGTCTTGAGTCCAAC





CCGGTAAGACACGACTTATCGCCACTGGCAGCAGCCACTGGTAACAGGATTAGCAGAGCGAG





GTATGTAGGCGGTGCTACAGAGTTCTTGAAGTGGTGGCCTAACTACGGCTACACTAGAAGGAC





AGTATTTGGTATCTGCGCTCTGCTGAAGCCAGTTACCTTCGGAAAAAGAGTTGGTAGCTCTTG





ATCCGGCAAACAAACCACCGCTGGTAGCGGTGGTTTTTTTGTTTGCAAGCAGCAGATTACGCG





CAGAAAAAAAGGATCTCAAGAAGATCCTTTGATCTTTTCTACGGGGTCTGACGCTCAGTGGA





ACGAAAACTCACGTTAAGGGATTTTGGTCATGAGATTATCAAAAAGGATCTTCACCTAGATCC





TTTTAAATTAAAAATGAAGTTTTAAATCAATCTAAAGTATATATGAGTAAACTTGGTCTGACAG





TTACCAATGCTTAATCAGTGAGGCACCTATCTCAGCGATCTGTCTATTTCGTTCATCCATAGTTG





CCTGACTCCCCGTCGTGTAGATAACTACGATACGGGAGGGCTTACCATCTGGCCCCAGTGCTG





CAATGATACCGCGAGACCCACGCTCACCGGCTCCAGATTTATCAGCAATAAACCAGCCAGCCG





GAAGGGCCGAGCGCAGAAGTGGTCCTGCAACTTTATCCGCCTCCATCCAGTCTATTAATTGTT





GCCGGGAAGCTAGAGTAAGTAGTTCGCCAGTTAATAGTTTGCGCAACGTTGTTGCCATTGCTA





CAGGCATCGTGGTGTCACGCTCGTCGTTTGGTATGGCTTCATTCAGCTCCGGTTCCCAACGATC





AAGGCGAGTTACATGATCCCCCATGTTGTGCAAAAAAGCGGTTAGCTCCTTCGGTCCTCCGAT





CGTTGTCAGAAGTAAGTTGGCCGCAGTGTTATCACTCATGGTTATGGCAGCACTGCATAATTCT





CTTACTGTCATGCCATCCGTAAGATGCTTTTCTGTGACTGGTGAGTACTCAACCAAGTCATTCT





GAGAATAGTGTATGCGGCGACCGAGTTGCTCTTGCCCGGCGTCAATACGGGATAATACCGCGC





CACATAGCAGAACTTTAAAAGTGCTCATCATTGGAAAACGTTCTTCGGGGCGAAAACTCTCA





AGGATCTTACCGCTGTTGAGATCCAGTTCGATGTAACCCACTCGTGCACCCAACTGATCTTCA





GCATCTTTTACTTTCACCAGCGTTTCTGGGTGAGCAAAAACAGGAAGGCAAAATGCCGCAAA





AAAGGGAATAAGGGCGACACGGAAATGTTGAATACTCATACTCTTCCTTTTTCAATATTATTGA





AGCATTTATCAGGGTTATTGTCTCATGAGCGGATACATATTTGAATGTATTTAGAAAAATAAACA





AATAGGGGTTCCGCGCACATTTCCCCGAAAAGTGCCACCTGAACGAAGCATCTGTGCTTCATT





TTGTAGAACAAAAATGCAACGCGAGAGCGCTAATTTTTCAAACAAAGAATCTGAGCTGCATTT





TTACAGAACAGAAATGCAACGCGAAAGCGCTATTTTACCAACGAAGAATCTGTGCTTCATTTT





TGTAAAACAAAAATGCAACGCGAGAGCGCTAATTTTTCAAACAAAGAATCTGAGCTGCATTTT





TACAGAACAGAAATGCAACGCGAGAGCGCTATTTTACCAACAAAGAATCTATACTTCTTTTTT





GTTCTACAAAAATGCATCCCGAGAGCGCTATTTTTCTAACAAAGCATCTTAGATTACTTTTTTTC





TCCTTTGTGCGCTCTATAATGCAGTCTCTTGATAACTTTTTGCACTGTAGGTCCGTTAAGGTTAG





AAGAAGGCTACTTTGGTGTCTATTTTCTCTTCCATAAAAAAAGCCTGACTCCACTTCCCGCGTT





TACTGATTACTAGCGAAGCTGCGGGTGCATTTTTTCAAGATAAAGGCATCCCCGATTATATTCT





ATACCGATGTGGATTGCGCATACTTTGTGAACAGAAAGTGATAGCGTTGATGATTCTTCATTGG





TCAGAAAATTATGAACGGTTTCTTCTATTTTGTCTCTATATACTACGTATAGGAAATGTTTACATT





TTCGTATTGTTTTCGATTCACTCTATGAATAGTTCTTACTACAATTTTTTTGTCTAAAGAGTAATA





CTAGAGATAAACATAAAAAATGTAGAGGTCGAGTTTAGATGCAAGTTCAAGGAGCGAAAGGT





GGATGGGTAGGTTATATAGGGATATAGCACAGAGATATATAGCAAAGAGATACTTTTGAGCAAT





GTTTGTGGAAGCGGTATTCGCAATATTTTAGTAGCTCGTTACAGTCCGGTGCGTTTTTGGTTTT





TTGAAAGTGCGTCTTCAGAGCGCTTTTGGTTTTCAAAAGCGCTCTGAAGTTCCTATACTTTCTA





GAGAATAGGAACTTCGGAATAGGAACTTCAAAGCGTTTCCGAAAACGAGCGCTTCCGAAAAT





GCAACGCGAGCTGCGCACATACAGCTCACTGTTCACGTCGCACCTATATCTGCGTGTTGCCTG





TATATATATATACATGAGAAGAACGGCATAGTGCGTGTTTATGCTTAAATGCGTACTTATATGCGT





CTATTTATGTAGGATGAAAGGTAGTCTAGTACCTCCTGTGATATTATCCCATTCCATGCGGGGTA





TCGTATGCTTCCTTCAGCACTACCCTTTAGCTGTTCTATATGCTGCCACTCCTCAATTGGATTAG





TCTCATCCTTCAATGCTATCATTTCCTTTGATATTGGATCATACTAAGAAACCATTATTATCATGA





CATTAACCTATAAAAATAGGCGTATCACGAGGCCCTTTCGTCTCGCGCGTTTCGGTGATGACGG





TGAAAACCTCTGACACATGCAGCTCCCGGAGACGGTCACAGCTTGTCTGTAAGCGGATGCCG





GGAGCAGACAAGCCCGTCAGGGCGCGTCAGCGGGTGTTGGCGGGTGTCGGGGCTGGCTTAA





CTATGCGGCATCAGAGCAGATTGTACTGAGAGTGCACCATATCGACTACGTCGTAAGGCCGTT





TCTGACAGAGTAAAATTCTTGAGGGAACTTTCACCATTATGGGAAATGCTTCAAGAAGGTATT





GACTTAAACTCCATCAAATGGTCAGGTCATTGAGTGTTTTTTATTTGTTGTATTTTTTTTTTTTTA





GAGAAAATCCTCCAATATCAAATTAGGAATCGTAGTTTCATGATTTTCTGTTACACCTAACTTTT





TGTGTGGTGCCCTCCTCCTTGTCAATATTAATGTTAAAGTGCAATTCTTTTTCCTTATCACGTTG





AGCCATTAGTATCAATTTGCTTACCTGTATTCCTTTACTATCCTCCTTTTTCTCCTTCTTGATAAA





TGTATGTAGATTGCGTATATAGTTTCGTCTACCCTATGAACATATTCCATTTTGTAATTTCGTGTC





GTTTCTATTATGAATTTCATTTATAAAGTTTATGTACAAATATCATAAAAAAAGAGAATCTTTTTA





AGCAAGGATTTTCTTAACTTCTTCGGCGACAGCATCACCGACTTCGGTGGTACTGTTGGAACC





ACCTAAATCACCAGTTCTGATACCTGCATCCAAAACCTTTTTAACTGCATCTTCAATGGCCTTA





CCTTCTTCAGGCAAGTTCAATGACAATTTCAACATCATTGCAGCAGACAAGATAGTGGCGATA





GGGTCAACCTTATTCTTTGGCAAATCTGGAGCAGAACCGTGGCATGGTTCGTACAAACCAAAT





GCGGTGTTCTTGTCTGGCAAAGAGGCCAAGGACGCAGATGGCAACAAACCCAAGGAACCTG





GGATAACGGAGGCTTCATCGGAGATGATATCACCAAACATGTTGCTGGTGATTATAATACCATT





TAGGTGGGTTGGGTTCTTAACTAGGATCATGGCGGCAGAATCAATCAATTGATGTTGAACCTTC





AATGTAGGGAATTCGTTCTTGATGGTTTCCTCCACAGTTTTTCTCCATAATCTTGAAGAGGCCA





AAAGATTAGCTTTATCCAAGGACCAAATAGGCAATGGTGGCTCATGTTGTAGGGCCATGAAAG





CGGCCATTCTTGTGATTCTTTGCACTTCTGGAACGGTGTATTGTTCACTATCCCAAGCGACACC





ATCACCATCGTCTTCCTTTCTCTTACCAAAGTAAATACCTCCCACTAATTCTCTGACAACAACG





AAGTCAGTACCTTTAGCAAATTGTGGCTTGATTGGAGATAAGTCTAAAAGAGAGTCGGATGCA





AAGTTACATGGTCTTAAGTTGGCGTACAATTGAAGTTCTTTACGGATTTTTAGTAAACCTTGTT





CAGGTCTAACACTACCGGTACCCCATTTAGGACCAGCCACAGCACCTAACAAAACGGCATCA





ACCTTCTTGGAGGCTTCCAGCGCCTCATCTGGAAGTGGGACACCTGTAGCATCGATAGCAGCA





CCACCAATTAAATGATTTTCGAAATCGAACTTGACATTGGAACGAACATCAGAAATAGCTTTA





AGAACCTTAATGGCTTCGGCTGTGATTTCTTGACCAACGTGGTCACCTGGCAAAACGACGATC





TTCTTAGGGGCAGACATAGGGGCAGACATTAGAATGGTATATCCTTGAAATATATATATATATTG





CTGAAATGTAAAAGGTAAGAAAAGTTAGAAAGTAAGACGATTGCTAACCACCTATTGGAAAA





AACAATAGGTCCTTAAATAATATTGTCAACTTCAAGTATTGTGATGCAAGCATTTAGTCATGAA





CGCTTCTCTATTCTATATGAAAAGCCGGTTCCGGCCTCTCACCTTTCCTTTTTCTCCCAATTTTT





CAGTTGAAAAAGGTATATGCGTCAGGCGACCTCTGAAATTAACAAAAAATTTCCAGTCATCGA





ATTTGATTCTGTGCGATAGCGCCCCTGTGTGTTCTCGTTATGTTGAGGAAAAAAATAATGGTTG





CTAAGAGATTCGAACTCTTGCATCTTACGATACCTGAGTATTCCCACAGTTAACTGCGGTCAAG





ATATTTCTTGAATCAGGCGCCTTAGACCGCTCGGCCAAACAACCAATTACTTGTTGAGAAATA





GAGTATAATTATCCTATAAATATAACGTTTTTGAACACACATGAACAAGGAAGTACAGGACAAT





TGATTTTGAAGAGAATGTGGATTTTGATGTAATTGTTGGGATTCCATTTTTAATAAGGCAATAAT





ATTAGGTATGTGGATATACTAGAAGTTCTCCTCGACCGTCGATATGCGGTGTGAAATACCGCAC





AGATGCGTAAGGAGAAAATACCGCATCAGGAAATTGTAAACGTTAATATTTTGTTAAAATTCGC





GTTAAATTTTTGTTAAATCAGCTCATTTTTTAACCAATAGGCCGAAATCGGCAAAATCCCTTATA





AATCAAAAGAATAGACCGAGATAGGGTTGAGTGTTGTTCCAGTTTGGAACAAGAGTCCACTAT





TAAAGAACGTGGACTCCAACGTCAAAGGGCGAAAAACCGTCTATCAGGGCGATGGCCCACTA





CGTGAACCATCACCCTAATCAAGTTTTTTGGGGTCGAGGTGCCGTAAAGCACTAAATCGGAAC





CCTAAAGGGAGCCCCCGATTTAGAGCTTGACGGGGAAAGCCGGCGAACGTGGCGAGAAAGG





AAGGGAAGAAAGCGAAAGGAGCGGGCGCTAGGGCGCTGGCAAGTGTAGCGGTCACGCTGC





GCGTAACCACCACACCCGCCGCGCTTAATGCGCCGCTACAGGGCGCGTCGCGCCATTCGCCAT





TCAGGCTGCGCAACTGTTGGGAAGGGCGATCGGTGCGGGCCTCTTCGCTATTACGCCAGCTG





GCGAAAGGGGGATGTGCTGCAAGGCGATTAAGTTGGGTAACGCCAGGGTTTTCCCAGTCACG





ACGTTGTAAAACGACGGCCAGTGAGCGCGCGTAATACGACTCACTATAGGGCGAATTGGGTA





CCGGGCCCCCCCTCGAGGTCGACGGTATCGATAAGCTTGATATCGAATTCCTGCAGCCCGGGG





GATCCGTTAGAATCATTTTGAATAAAAAACACGCTTTTTCAGTTCGAGTTTATCATTATCAATAC





TGCCATTTCAAAGAATACGTAAATAATTAATAGTAGTGATTTTCCTAACTTTATTTAGTCAAAAA





ATTAGCCTTTTAATTCTGCTGTAACCCGTACATGCCCAAAATAGGGGGCGGGTTACACAGAATA





TATAACATCGTAGGTGTCTGGGTGAACAGTTTATTCCTGGCATCCACTAAATATAATGGAGCCC





GCTTTTTAAGCTGGCATCCAGAAAAAAAAAGAATCCCAGCACCAAAATATTGTTTTCTTCACC





AACCATCAGTTCATAGGTCCATTCTCTTAGCGCAACTACAGAGAACAGGGGCACAAACAGGC





AAAAAACGGGCACAACCTCAATGGAGTGATGCAACCTGCCTGGAGTAAATGATGACACAAGG





CAATTGACCCACGCATGTATCTATCTCATTTTCTTACACCTTCTATTACCTTCTGCTCTCTCTGAT





TTGGAAAAAGCTGAAAAAAAAGGTTGAAACCAGTTCCCTGAAATTATTCCCCTACTTGACTA





ATAAGTATATAAAGACGGTAGGTATTGATTGTAATTCTGTAAATCTATTTCTTAAACTTCTTAAAT





TCTACTTTTATAGTTAGTCTTTTTTTTAGTTTTAAAACACCAAGAACTTAGTTTCGAATAAACAC





ACATAAACAAACAAAGAATTC





p425-GFP_-1fs_mCherry.


(SEQ ID NO: 732)



ATGTCTAAAGGTGAAGAATTATTCACTGGTGTTGTCCCAATTTTGGTTGAATTAGATGGTGATG






TTAATGGTCACAAATTTTCTGTCTCCGGTGAAGGTGAAGGTGATGCTACTTACGGTAAATTGA





CCTTAAAATTTATTTGTACTACTGGTAAATTGCCAGTTCCATGGCCAACCTTAGTCACTACTTTC





GGTTATGGTGTTCAATGTTTTGCTAGATACCCAGATCATATGAAACAACATGACTTTTTCAAGT





CTGCCATGCCAGAAGGTTATGTTCAAGAAAGAACTATTTTTTTCAAAGATGACGGTAACTACA





AGACCAGAGCTGAAGTCAAGTTTGAAGGTGATACCTTAGTTAATAGAATCGAATTAAAAGGTA





TTGATTTTAAAGAAGATGGTAACATTTTAGGTCACAAATTGGAATACAACTATAACTCTCACAA





TGTTTACATCATGGCTGACAAACAAAAGAATGGTATCAAAGTTAACTTCAAAATTAGACACAA





CATTGAAGATGGTTCTGTTCAATTAGCTGACCATTATCAACAAAATACTCCAATTGGTGATGGT





CCAGTCTTGTTACCAGACAACCATTACTTATCCACTCAATCTGCCTTATCCAAAGATCCAAACG





AAAAGAGAGACCACATGGTCTTGTTAGAATTTGTTACTGCTGCTGGTATTACCCATGGTATGGA





TGAATTGTACAAAGCTAGCAAACCTGGGTCAATCCTTGGGGCCCAGACTGAGCACGTGATG






GCAGAGCACAGGACGTCATGGTTTCAAAAGGTGAAGAAGATAATATGGCTATTATTAAAGAATTTAT







GAGATTTAAAGTTCATATGGAAGGTTCAGTTAATGGTCATGAATTTGAAATTGAAGGTGAAGGTGAAG







GTAGACCATATGAAGGTACTCAAACTGCTAAATTGAAAGTTACTAAAGGTGGTCCATTACCATTTGCTT







GGGATATTTTGTCACCACAATTTATGTATGGTTCAAAAGCTTATGTTAAACATCCAGCTGATATTCCAG







ATTATTTAAAATTGTCATTTCCAGAAGGTTTTAAATGGGAAAGAGTTATGAATTTTGAAGATGGTGGTG







TTGTTACTGTTACTCAAGATTCATCATTACAAGATGGTGAATTTATTTATAAAGTTAAATTGAGAGGTAC







TAATTTTCCATCAGATGGTCCAGTTATGCAAAAAAAAACTATGGGTTGGGAAGCTTCATCAGAAAGAA







TGTATCCAGAAGATGGTGCTTTAAAAGGTGAAATTAAACAAAGATTGAAATTAAAAGATGGTGGTCAT







TATGATGCTGAAGTTAAAACTACTTATAAAGCTAAAAAACCAGTTCAATTACCAGGTGCTTATAATGTTA







ATATTAAATTGGATATTACTTCACATAATGAAGATTATACTATTGTTGAACAATATGAAAGAGCTGAAGG







TAGACATTCAACTGGTGGTATGGATGAATTATATAAAGGTACCGCTCGAGCAGCTGTGATTGATTGAG






TCGACTTGGTTGAACACGTTGCCAAGGCTTAAGTGAATTTACTTTAAATCTTGCATTTAAATAA





ATTTTCTTTTTATAGCTTTATGACTTAGTTTCAATTTATATACTATTTTAATGACATTTTCGATTCG





GATCCACTAGTTCTAGAGCGGCCGCCACCGCGGTGGAGCTCCAGCTTTTGTTCCCTTTAGTGA





GGGTTAATTGCGCGCTTGGCGTAATCATGGTCATAGCTGTTTCCTGTGTGAAATTGTTATCCGC





TCACAATTCCACACAACATAGGAGCCGGAAGCATAAAGTGTAAAGCCTGGGGTGCCTAATGA





GTGAGGTAACTCACATTAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTCG





TGCCAGCTGCATTAATGAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCTCT





TCCGCTTCCTCGCTCACTGACTCGCTGCGCTCGGTCGTTCGGCTGCGGCGAGCGGTATCAGCT





CACTCAAAGGCGGTAATACGGTTATCCACAGAATCAGGGGATAACGCAGGAAAGAACATGTG





AGCAAAAGGCCAGCAAAAGGCCAGGAACCGTAAAAAGGCCGCGTTGCTGGCGTTTTTCCAT





AGGCTCCGCCCCCCTGACGAGCATCACAAAAATCGACGCTCAAGTCAGAGGTGGCGAAACC





CGACAGGACTATAAAGATACCAGGCGTTTCCCCCTGGAAGCTCCCTCGTGCGCTCTCCTGTTC





CGACCCTGCCGCTTACCGGATACCTGTCCGCCTTTCTCCCTTCGGGAAGCGTGGCGCTTTCTC





ATAGCTCACGCTGTAGGTATCTCAGTTCGGTGTAGGTCGTTCGCTCCAAGCTGGGCTGTGTGC





ACGAACCCCCCGTTCAGCCCGACCGCTGCGCCTTATCCGGTAACTATCGTCTTGAGTCCAACC





CGGTAAGACACGACTTATCGCCACTGGCAGCAGCCACTGGTAACAGGATTAGCAGAGCGAGG





TATGTAGGCGGTGCTACAGAGTTCTTGAAGTGGTGGCCTAACTACGGCTACACTAGAAGGACA





GTATTTGGTATCTGCGCTCTGCTGAAGCCAGTTACCTTCGGAAAAAGAGTTGGTAGCTCTTGA





TCCGGCAAACAAACCACCGCTGGTAGCGGTGGTTTTTTTGTTTGCAAGCAGCAGATTACGCG





CAGAAAAAAAGGATCTCAAGAAGATCCTTTGATCTTTTCTACGGGGTCTGACGCTCAGTGGA





ACGAAAACTCACGTTAAGGGATTTTGGTCATGAGATTATCAAAAAGGATCTTCACCTAGATCC





TTTTAAATTAAAAATGAAGTTTTAAATCAATCTAAAGTATATATGAGTAAACTTGGTCTGACAG





TTACCAATGCTTAATCAGTGAGGCACCTATCTCAGCGATCTGTCTATTTCGTTCATCCATAGTTG





CCTGACTCCCCGTCGTGTAGATAACTACGATACGGGAGGGCTTACCATCTGGCCCCAGTGCTG





CAATGATACCGCGAGACCCACGCTCACCGGCTCCAGATTTATCAGCAATAAACCAGCCAGCCG





GAAGGGCCGAGCGCAGAAGTGGTCCTGCAACTTTATCCGCCTCCATCCAGTCTATTAATTGTT





GCCGGGAAGCTAGAGTAAGTAGTTCGCCAGTTAATAGTTTGCGCAACGTTGTTGCCATTGCTA





CAGGCATCGTGGTGTCACGCTCGTCGTTTGGTATGGCTTCATTCAGCTCCGGTTCCCAACGATC





AAGGCGAGTTACATGATCCCCCATGTTGTGCAAAAAAGCGGTTAGCTCCTTCGGTCCTCCGAT





CGTTGTCAGAAGTAAGTTGGCCGCAGTGTTATCACTCATGGTTATGGCAGCACTGCATAATTCT





CTTACTGTCATGCCATCCGTAAGATGCTTTTCTGTGACTGGTGAGTACTCAACCAAGTCATTCT





GAGAATAGTGTATGCGGCGACCGAGTTGCTCTTGCCCGGCGTCAATACGGGATAATACCGCGC





CACATAGCAGAACTTTAAAAGTGCTCATCATTGGAAAACGTTCTTCGGGGCGAAAACTCTCA





AGGATCTTACCGCTGTTGAGATCCAGTTCGATGTAACCCACTCGTGCACCCAACTGATCTTCA





GCATCTTTTACTTTCACCAGCGTTTCTGGGTGAGCAAAAACAGGAAGGCAAAATGCCGCAAA





AAAGGGAATAAGGGCGACACGGAAATGTTGAATACTCATACTCTTCCTTTTTCAATATTATTGA





AGCATTTATCAGGGTTATTGTCTCATGAGCGGATACATATTTGAATGTATTTAGAAAAATAAACA





AATAGGGGTTCCGCGCACATTTCCCCGAAAAGTGCCACCTGAACGAAGCATCTGTGCTTCATT





TTGTAGAACAAAAATGCAACGCGAGAGCGCTAATTTTTCAAACAAAGAATCTGAGCTGCATTT





TTACAGAACAGAAATGCAACGCGAAAGCGCTATTTTACCAACGAAGAATCTGTGCTTCATTTT





TGTAAAACAAAAATGCAACGCGAGAGCGCTAATTTTTCAAACAAAGAATCTGAGCTGCATTTT





TACAGAACAGAAATGCAACGCGAGAGCGCTATTTTACCAACAAAGAATCTATACTTCTTTTTT





GTTCTACAAAAATGCATCCCGAGAGCGCTATTTTTCTAACAAAGCATCTTAGATTACTTTTTTTC





TCCTTTGTGCGCTCTATAATGCAGTCTCTTGATAACTTTTTGCACTGTAGGTCCGTTAAGGTTAG





AAGAAGGCTACTTTGGTGTCTATTTTCTCTTCCATAAAAAAAGCCTGACTCCACTTCCCGCGTT





TACTGATTACTAGCGAAGCTGCGGGTGCATTTTTTCAAGATAAAGGCATCCCCGATTATATTCT





ATACCGATGTGGATTGCGCATACTTTGTGAACAGAAAGTGATAGCGTTGATGATTCTTCATTGG





TCAGAAAATTATGAACGGTTTCTTCTATTTTGTCTCTATATACTACGTATAGGAAATGTTTACATT





TTCGTATTGTTTTCGATTCACTCTATGAATAGTTCTTACTACAATTTTTTTGTCTAAAGAGTAATA





CTAGAGATAAACATAAAAAATGTAGAGGTCGAGTTTAGATGCAAGTTCAAGGAGCGAAAGGT





GGATGGGTAGGTTATATAGGGATATAGCACAGAGATATATAGCAAAGAGATACTTTTGAGCAAT





GTTTGTGGAAGCGGTATTCGCAATATTTTAGTAGCTCGTTACAGTCCGGTGCGTTTTTGGTTTT





TTGAAAGTGCGTCTTCAGAGCGCTTTTGGTTTTCAAAAGCGCTCTGAAGTTCCTATACTTTCTA





GAGAATAGGAACTTCGGAATAGGAACTTCAAAGCGTTTCCGAAAACGAGCGCTTCCGAAAAT





GCAACGCGAGCTGCGCACATACAGCTCACTGTTCACGTCGCACCTATATCTGCGTGTTGCCTG





TATATATATATACATGAGAAGAACGGCATAGTGCGTGTTTATGCTTAAATGCGTACTTATATGCGT





CTATTTATGTAGGATGAAAGGTAGTCTAGTACCTCCTGTGATATTATCCCATTCCATGCGGGGTA





TCGTATGCTTCCTTCAGCACTACCCTTTAGCTGTTCTATATGCTGCCACTCCTCAATTGGATTAG





TCTCATCCTTCAATGCTATCATTTCCTTTGATATTGGATCATACTAAGAAACCATTATTATCATGA





CATTAACCTATAAAAATAGGCGTATCACGAGGCCCTTTCGTCTCGCGCGTTTCGGTGATGACGG





TGAAAACCTCTGACACATGCAGCTCCCGGAGACGGTCACAGCTTGTCTGTAAGCGGATGCCG





GGAGCAGACAAGCCCGTCAGGGCGCGTCAGCGGGTGTTGGCGGGTGTCGGGGCTGGCTTAA





CTATGCGGCATCAGAGCAGATTGTACTGAGAGTGCACCATATCGACTACGTCGTAAGGCCGTT





TCTGACAGAGTAAAATTCTTGAGGGAACTTTCACCATTATGGGAAATGCTTCAAGAAGGTATT





GACTTAAACTCCATCAAATGGTCAGGTCATTGAGTGTTTTTTATTTGTTGTATTTTTTTTTTTTTA





GAGAAAATCCTCCAATATCAAATTAGGAATCGTAGTTTCATGATTTTCTGTTACACCTAACTTTT





TGTGTGGTGCCCTCCTCCTTGTCAATATTAATGTTAAAGTGCAATTCTTTTTCCTTATCACGTTG





AGCCATTAGTATCAATTTGCTTACCTGTATTCCTTTACTATCCTCCTTTTTCTCCTTCTTGATAAA





TGTATGTAGATTGCGTATATAGTTTCGTCTACCCTATGAACATATTCCATTTTGTAATTTCGTGTC





GTTTCTATTATGAATTTCATTTATAAAGTTTATGTACAAATATCATAAAAAAAGAGAATCTTTTTA





AGCAAGGATTTTCTTAACTTCTTCGGCGACAGCATCACCGACTTCGGTGGTACTGTTGGAACC





ACCTAAATCACCAGTTCTGATACCTGCATCCAAAACCTTTTTAACTGCATCTTCAATGGCCTTA





CCTTCTTCAGGCAAGTTCAATGACAATTTCAACATCATTGCAGCAGACAAGATAGTGGCGATA





GGGTCAACCTTATTCTTTGGCAAATCTGGAGCAGAACCGTGGCATGGTTCGTACAAACCAAAT





GCGGTGTTCTTGTCTGGCAAAGAGGCCAAGGACGCAGATGGCAACAAACCCAAGGAACCTG





GGATAACGGAGGCTTCATCGGAGATGATATCACCAAACATGTTGCTGGTGATTATAATACCATT





TAGGTGGGTTGGGTTCTTAACTAGGATCATGGCGGCAGAATCAATCAATTGATGTTGAACCTTC





AATGTAGGGAATTCGTTCTTGATGGTTTCCTCCACAGTTTTTCTCCATAATCTTGAAGAGGCCA





AAAGATTAGCTTTATCCAAGGACCAAATAGGCAATGGTGGCTCATGTTGTAGGGCCATGAAAG





CGGCCATTCTTGTGATTCTTTGCACTTCTGGAACGGTGTATTGTTCACTATCCCAAGCGACACC





ATCACCATCGTCTTCCTTTCTCTTACCAAAGTAAATACCTCCCACTAATTCTCTGACAACAACG





AAGTCAGTACCTTTAGCAAATTGTGGCTTGATTGGAGATAAGTCTAAAAGAGAGTCGGATGCA





AAGTTACATGGTCTTAAGTTGGCGTACAATTGAAGTTCTTTACGGATTTTTAGTAAACCTTGTT





CAGGTCTAACACTACCGGTACCCCATTTAGGACCAGCCACAGCACCTAACAAAACGGCATCA





ACCTTCTTGGAGGCTTCCAGCGCCTCATCTGGAAGTGGGACACCTGTAGCATCGATAGCAGCA





CCACCAATTAAATGATTTTCGAAATCGAACTTGACATTGGAACGAACATCAGAAATAGCTTTA





AGAACCTTAATGGCTTCGGCTGTGATTTCTTGACCAACGTGGTCACCTGGCAAAACGACGATC





TTCTTAGGGGCAGACATAGGGGCAGACATTAGAATGGTATATCCTTGAAATATATATATATATTG





CTGAAATGTAAAAGGTAAGAAAAGTTAGAAAGTAAGACGATTGCTAACCACCTATTGGAAAA





AACAATAGGTCCTTAAATAATATTGTCAACTTCAAGTATTGTGATGCAAGCATTTAGTCATGAA





CGCTTCTCTATTCTATATGAAAAGCCGGTTCCGGCCTCTCACCTTTCCTTTTTCTCCCAATTTTT





CAGTTGAAAAAGGTATATGCGTCAGGCGACCTCTGAAATTAACAAAAAATTTCCAGTCATCGA





ATTTGATTCTGTGCGATAGCGCCCCTGTGTGTTCTCGTTATGTTGAGGAAAAAAATAATGGTTG





CTAAGAGATTCGAACTCTTGCATCTTACGATACCTGAGTATTCCCACAGTTAACTGCGGTCAAG





ATATTTCTTGAATCAGGCGCCTTAGACCGCTCGGCCAAACAACCAATTACTTGTTGAGAAATA





GAGTATAATTATCCTATAAATATAACGTTTTTGAACACACATGAACAAGGAAGTACAGGACAAT





TGATTTTGAAGAGAATGTGGATTTTGATGTAATTGTTGGGATTCCATTTTTAATAAGGCAATAAT





ATTAGGTATGTGGATATACTAGAAGTTCTCCTCGACCGTCGATATGCGGTGTGAAATACCGCAC





AGATGCGTAAGGAGAAAATACCGCATCAGGAAATTGTAAACGTTAATATTTTGTTAAAATTCGC





GTTAAATTTTTGTTAAATCAGCTCATTTTTTAACCAATAGGCCGAAATCGGCAAAATCCCTTATA





AATCAAAAGAATAGACCGAGATAGGGTTGAGTGTTGTTCCAGTTTGGAACAAGAGTCCACTAT





TAAAGAACGTGGACTCCAACGTCAAAGGGCGAAAAACCGTCTATCAGGGCGATGGCCCACTA





CGTGAACCATCACCCTAATCAAGTTTTTTGGGGTCGAGGTGCCGTAAAGCACTAAATCGGAAC





CCTAAAGGGAGCCCCCGATTTAGAGCTTGACGGGGAAAGCCGGCGAACGTGGCGAGAAAGG





AAGGGAAGAAAGCGAAAGGAGCGGGCGCTAGGGCGCTGGCAAGTGTAGCGGTCACGCTGC





GCGTAACCACCACACCCGCCGCGCTTAATGCGCCGCTACAGGGCGCGTCGCGCCATTCGCCAT





TCAGGCTGCGCAACTGTTGGGAAGGGCGATCGGTGCGGGCCTCTTCGCTATTACGCCAGCTG





GCGAAAGGGGGATGTGCTGCAAGGCGATTAAGTTGGGTAACGCCAGGGTTTTCCCAGTCACG





ACGTTGTAAAACGACGGCCAGTGAGCGCGCGTAATACGACTCACTATAGGGCGAATTGGGTA





CCGGGCCCCCCCTCGAGGTCGACGGTATCGATAAGCTTGATATCGAATTCCTGCAGCCCGGGG





GATCCGTTAGAATCATTTTGAATAAAAAACACGCTTTTTCAGTTCGAGTTTATCATTATCAATAC





TGCCATTTCAAAGAATACGTAAATAATTAATAGTAGTGATTTTCCTAACTTTATTTAGTCAAAAA





ATTAGCCTTTTAATTCTGCTGTAACCCGTACATGCCCAAAATAGGGGGCGGGTTACACAGAATA





TATAACATCGTAGGTGTCTGGGTGAACAGTTTATTCCTGGCATCCACTAAATATAATGGAGCCC





GCTTTTTAAGCTGGCATCCAGAAAAAAAAAGAATCCCAGCACCAAAATATTGTTTTCTTCACC





AACCATCAGTTCATAGGTCCATTCTCTTAGCGCAACTACAGAGAACAGGGGCACAAACAGGC





AAAAAACGGGCACAACCTCAATGGAGTGATGCAACCTGCCTGGAGTAAATGATGACACAAGG





CAATTGACCCACGCATGTATCTATCTCATTTTCTTACACCTTCTATTACCTTCTGCTCTCTCTGAT





TTGGAAAAAGCTGAAAAAAAAGGTTGAAACCAGTTCCCTGAAATTATTCCCCTACTTGACTA





ATAAGTATATAAAGACGGTAGGTATTGATTGTAATTCTGTAAATCTATTTCTTAAACTTCTTAAAT





TCTACTTTTATAGTTAGTCTTTTTTTTAGTTTTAAAACACCAAGAACTTAGTTTCGAATAAACAC





ACATAAACAAACAAAGAATTC


KEY:


GFP open reading frame


Linker containing stop codon +1 frameshift, or -1 frameshift


mCherry open reading frame


Plasmid backbone (containing the GPD promoter, Leu2 marker, and AmpR)


Protospacer (underlined)


PAM (boldfaced)






DNA Sequences of Mammalian Prime Editor Plasmids and Example PEgRNA Plasmid










pCMV-PE1:



(SEQ ID NO: 733)



ATGAAACGGACAGCCGACGGAAGCGAGTTCGAGTCACCAAAGAAGAAGCGGAAAGTCGAC



AAGAAGTACAGCATCGGCCTGGACATCGGCACCAACTCTGTGGGCTGGGCCGTGATCACCGA


CGAGTACAAGGTGCCCAGCAAGAAATTCAAGGTGCTGGGCAACACCGACCGGCACAGCATC


AAGAAGAACCTGATCGGAGCCCTGCTGTTCGACAGCGGCGAAACAGCCGAGGCCACCCGGC


TGAAGAGAACCGCCAGAAGAAGATACACCAGACGGAAGAACCGGATCTGCTATCTGCAAGA


GATCTTCAGCAACGAGATGGCCAAGGTGGACGACAGCTTCTTCCACAGACTGGAAGAGTCCT


TCCTGGTGGAAGAGGATAAGAAGCACGAGCGGCACCCCATCTTCGGCAACATCGTGGACGAG


GTGGCCTACCACGAGAAGTACCCCACCATCTACCACCTGAGAAAGAAACTGGTGGACAGCAC


CGACAAGGCCGACCTGCGGCTGATCTATCTGGCCCTGGCCCACATGATCAAGTTCCGGGGCCA


CTTCCTGATCGAGGGCGACCTGAACCCCGACAACAGCGACGTGGACAAGCTGTTCATCCAGC


TGGTGCAGACCTACAACCAGCTGTTCGAGGAAAACCCCATCAACGCCAGCGGCGTGGACGCC


AAGGCCATCCTGTCTGCCAGACTGAGCAAGAGCAGACGGCTGGAAAATCTGATCGCCCAGCT


GCCCGGCGAGAAGAAGAATGGCCTGTTCGGAAACCTGATTGCCCTGAGCCTGGGCCTGACCC


CCAACTTCAAGAGCAACTTCGACCTGGCCGAGGATGCCAAACTGCAGCTGAGCAAGGACAC


CTACGACGACGACCTGGACAACCTGCTGGCCCAGATCGGCGACCAGTACGCCGACCTGTTTC


TGGCCGCCAAGAACCTGTCCGACGCCATCCTGCTGAGCGACATCCTGAGAGTGAACACCGAG


ATCACCAAGGCCCCCCTGAGCGCCTCTATGATCAAGAGATACGACGAGCACCACCAGGACCT


GACCCTGCTGAAAGCTCTCGTGCGGCAGCAGCTGCCTGAGAAGTACAAAGAGATTTTCTTCG


ACCAGAGCAAGAACGGCTACGCCGGCTACATTGACGGCGGAGCCAGCCAGGAAGAGTTCTA


CAAGTTCATCAAGCCCATCCTGGAAAAGATGGACGGCACCGAGGAACTGCTCGTGAAGCTGA


ACAGAGAGGACCTGCTGCGGAAGCAGCGGACCTTCGACAACGGCAGCATCCCCCACCAGAT


CCACCTGGGAGAGCTGCACGCCATTCTGCGGCGGCAGGAAGATTTTTACCCATTCCTGAAGG


ACAACCGGGAAAAGATCGAGAAGATCCTGACCTTCCGCATCCCCTACTACGTGGGCCCTCTG


GCCAGGGGAAACAGCAGATTCGCCTGGATGACCAGAAAGAGCGAGGAAACCATCACCCCCT


GGAACTTCGAGGAAGTGGTGGACAAGGGCGCTTCCGCCCAGAGCTTCATCGAGCGGATGAC


CAACTTCGATAAGAACCTGCCCAACGAGAAGGTGCTGCCCAAGCACAGCCTGCTGTACGAGT


ACTTCACCGTGTATAACGAGCTGACCAAAGTGAAATACGTGACCGAGGGAATGAGAAAGCCC


GCCTTCCTGAGCGGCGAGCAGAAAAAGGCCATCGTGGACCTGCTGTTCAAGACCAACCGGA


AAGTGACCGTGAAGCAGCTGAAAGAGGACTACTTCAAGAAAATCGAGTGCTTCGACTCCGTG


GAAATCTCCGGCGTGGAAGATCGGTTCAACGCCTCCCTGGGCACATACCACGATCTGCTGAA


AATTATCAAGGACAAGGACTTCCTGGACAATGAGGAAAACGAGGACATTCTGGAAGATATCG


TGCTGACCCTGACACTGTTTGAGGACAGAGAGATGATCGAGGAACGGCTGAAAACCTATGCC


CACCTGTTCGACGACAAAGTGATGAAGCAGCTGAAGCGGCGGAGATACACCGGCTGGGGCA


GGCTGAGCCGGAAGCTGATCAACGGCATCCGGGACAAGCAGTCCGGCAAGACAATCCTGGAT


TTCCTGAAGTCCGACGGCTTCGCCAACAGAAACTTCATGCAGCTGATCCACGACGACAGCCT


GACCTTTAAAGAGGACATCCAGAAAGCCCAGGTGTCCGGCCAGGGCGATAGCCTGCACGAGC


ACATTGCCAATCTGGCCGGCAGCCCCGCCATTAAGAAGGGCATCCTGCAGACAGTGAAGGTG


GTGGACGAGCTCGTGAAAGTGATGGGCCGGCACAAGCCCGAGAACATCGTGATCGAAATGG


CCAGAGAGAACCAGACCACCCAGAAGGGACAGAAGAACAGCCGCGAGAGAATGAAGCGGA


TCGAAGAGGGCATCAAAGAGCTGGGCAGCCAGATCCTGAAAGAACACCCCGTGGAAAACAC


CCAGCTGCAGAACGAGAAGCTGTACCTGTACTACCTGCAGAATGGGCGGGATATGTACGTGG


ACCAGGAACTGGACATCAACCGGCTGTCCGACTACGATGTGGACGCTATCGTGCCTCAGAGC


TTTCTGAAGGACGACTCCATCGACAACAAGGTGCTGACCAGAAGCGACAAGAACCGGGGCA


AGAGCGACAACGTGCCCTCCGAAGAGGTCGTGAAGAAGATGAAGAACTACTGGCGGCAGCT


GCTGAACGCCAAGCTGATTACCCAGAGAAAGTTCGACAATCTGACCAAGGCCGAGAGAGGC


GGCCTGAGCGAACTGGATAAGGCCGGCTTCATCAAGAGACAGCTGGTGGAAACCCGGCAGAT


CACAAAGCACGTGGCACAGATCCTGGACTCCCGGATGAACACTAAGTACGACGAGAATGACA


AGCTGATCCGGGAAGTGAAAGTGATCACCCTGAAGTCCAAGCTGGTGTCCGATTTCCGGAAG


GATTTCCAGTTTTACAAAGTGCGCGAGATCAACAACTACCACCACGCCCACGACGCCTACCTG


AACGCCGTCGTGGGAACCGCCCTGATCAAAAAGTACCCTAAGCTGGAAAGCGAGTTCGTGTA


CGGCGACTACAAGGTGTACGACGTGCGGAAGATGATCGCCAAGAGCGAGCAGGAAATCGGC


AAGGCTACCGCCAAGTACTTCTTCTACAGCAACATCATGAACTTTTTCAAGACCGAGATTACC


CTGGCCAACGGCGAGATCCGGAAGCGGCCTCTGATCGAGACAAACGGCGAAACCGGGGAGA


TCGTGTGGGATAAGGGCCGGGATTTTGCCACCGTGCGGAAAGTGCTGAGCATGCCCCAAGTG


AATATCGTGAAAAAGACCGAGGTGCAGACAGGCGGCTTCAGCAAAGAGTCTATCCTGCCCAA


GAGGAACAGCGATAAGCTGATCGCCAGAAAGAAGGACTGGGACCCTAAGAAGTACGGCGGC


TTCGACAGCCCCACCGTGGCCTATTCTGTGCTGGTGGTGGCCAAAGTGGAAAAGGGCAAGTC


CAAGAAACTGAAGAGTGTGAAAGAGCTGCTGGGGATCACCATCATGGAAAGAAGCAGCTTC


GAGAAGAATCCCATCGACTTTCTGGAAGCCAAGGGCTACAAAGAAGTGAAAAAGGACCTGA


TCATCAAGCTGCCTAAGTACTCCCTGTTCGAGCTGGAAAACGGCCGGAAGAGAATGCTGGCC


TCTGCCGGCGAACTGCAGAAGGGAAACGAACTGGCCCTGCCCTCCAAATATGTGAACTTCCT


GTACCTGGCCAGCCACTATGAGAAGCTGAAGGGCTCCCCCGAGGATAATGAGCAGAAACAGC


TGTTTGTGGAACAGCACAAGCACTACCTGGACGAGATCATCGAGCAGATCAGCGAGTTCTCC


AAGAGAGTGATCCTGGCCGACGCTAATCTGGACAAAGTGCTGTCCGCCTACAACAAGCACCG


GGATAAGCCCATCAGAGAGCAGGCCGAGAATATCATCCACCTGTTTACCCTGACCAATCTGGG


AGCCCCTGCCGCCTTCAAGTACTTTGACACCACCATCGACCGGAAGAGGTACACCAGCACCA


AAGAGGTGCTGGACGCCACCCTGATCCACCAGAGCATCACCGGCCTGTACGAGACACGGATC


GACCTGTCTCAGCTGGGAGGTGACTCTGGAGGATCTAGCGGAGGATCCTCTGGCAGCGAGACAC



CAGGAACAAGCGAGTCAGCAACACCAGAGAGCAGTGGCGGCAGCAGCGGCGGCAGCAGC

ACCC






TAAATATAGAAGATGAGTATCGGCTACATGAGACCTCAAAAGAGCCAGATGTTTCTCTAG






GGTCCACATGGCTGTCTGATTTTCCTCAGGCCTGGGCGGAAACCGGGGGCATGGGACT






GGCAGTTCGCCAAGCTCCTCTGATCATACCTCTGAAAGCAACCTCTACCCCCGTGTCCA






TAAAACAATACCCCATGTCACAAGAAGCCAGACTGGGGATCAAGCCCCACATACAGAGA






CTGTTGGACCAGGGAATACTGGTACCCTGCCAGTCCCCCTGGAACACGCCCCTGCTACC






CGTTAAGAAACCAGGGACTAATGATTATAGGCCTGTCCAGGATCTGAGAGAAGTCAACA






AGCGGGTGGAAGACATCCACCCCACCGTGCCCAACCCTTACAACCTCTTGAGCGGGCT






CCCACCGTCCCACCAGTGGTACACTGTGCTTGATTTAAAGGATGCCTTTTTCTGCCTGA






GACTCCACCCCACCAGTCAGCCTCTCTTCGCCTTTGAGTGGAGAGATCCAGAGATGGG






AATCTCAGGACAATTGACCTGGACCAGACTCCCACAGGGTTTCAAAAACAGTCCCACCC






TGTTTGATGAGGCACTGCACAGAGACCTAGCAGACTTCCGGATCCAGCACCCAGACTT






GATCCTGCTACAGTACGTGGATGACTTACTGCTGGCCGCCACTTCTGAGCTAGACTGCC






AACAAGGTACTCGGGCCCTGTTACAAACCCTAGGGAACCTCGGGTATCGGGCCTCGGC






CAAGAAAGCCCAAATTTGCCAGAAACAGGTCAAGTATCTGGGGTATCTTCTAAAAGAGG






GTCAGAGATGGCTGACTGAGGCCAGAAAAGAGACTGTGATGGGGCAGCCTACTCCGAA






GACCCCTCGACAACTAAGGGAGTTCCTAGGGACGGCAGGCTTCTGTCGCCTCTGGATC






CCTGGGTTTGCAGAAATGGCAGCCCCCCTGTACCCTCTCACCAAAACGGGGACTCTGTT






TAATTGGGGCCCAGACCAACAAAAGGCCTATCAAGAAATCAAGCAAGCTCTTCTAACTG






CCCCAGCCCTGGGGTTGCCAGATTTGACTAAGCCCTTTGAACTCTTTGTCGACGAGAAG






CAGGGCTACGCCAAAGGTGTCCTAACGCAAAAACTGGGACCTTGGCGTCGGCCGGTGG






CCTACCTGTCCAAAAAGCTAGACCCAGTAGCAGCTGGGTGGCCCCCTTGCCTACGGATG






GTAGCAGCCATTGCCGTACTGACAAAGGATGCAGGCAAGCTAACCATGGGACAGCCAC






TAGTCATTCTGGCCCCCCATGCAGTAGAGGCACTAGTCAAACAACCCCCCGACCGCTGG






CTTTCCAACGCCCGGATGACTCACTATCAGGCCTTGCTTTTGGACACGGACCGGGTCCA






GTTCGGACCGGTGGTAGCCCTGAACCCGGCTACGCTGCTCCCACTGCCTGAGGAAGGG






CTGCAACACAACTGCCTTGATATCCTGGCCGAAGCCCACGGAACCCGACCCGACCTAAC






GGACCAGCCGCTCCCAGACGCCGACCACACCTGGTACACGGATGGAAGCAGTCTCTTA






CAAGAGGGACAGCGTAAGGCGGGAGCTGCGGTGACCACCGAGACCGAGGTAATCTGG






GCTAAAGCCCTGCCAGCCGGGACATCCGCTCAGCGGGCTGAACTGATAGCACTCACCC






AGGCCCTAAAGATGGCAGAAGGTAAGAAGCTAAATGTTTATACTGATAGCCGTTATGCTT






TTGCTACTGCCCATATCCATGGAGAAATATACAGAAGGCGTGGGTTGCTCACATCAGAA






GGCAAAGAGATCAAAAATAAAGACGAGATCTTGGCCCTACTAAAAGCCCTCTTTCTGCC






CAAAAGACTTAGCATAATCCATTGTCCAGGACATCAAAAGGGACACAGCGCCGAGGCTA






GAGGCAACCGGATGGCTGACCAAGCGGCCCGAAAGGCAGCCATCACAGAGACTCCAGA






CACCTCTACCCTCCTCATAGAAAATTCATCACCCTCTGGCGGCTCAAAAAGAACCGCCG






ACGGCAGCGAATTCGAGCCCAAGAAGAAGAGGAAAGTCTAA
CCGGTCATCATCACCATCA



CCATTGAGTTTAAACCCGCTGATCAGCCTCGACTGTGCCTTCTAGTTGCCAGCCATCTGTTGTT


TGCCCCTCCCCCGTGCCTTCCTTGACCCTGGAAGGTGCCACTCCCACTGTCCTTTCCTAATAA


AATGAGAAAATTGCATCGCATTGTCTGAGTAGGTGTCATTCTATTCTGGGGGGTGGGGTGGGG


CAGGACAGCAAGGGGGAGGATTGGGAAGACAATAGCAGGCATGCTGGGGATGCGGTGGGCT


CTATGGCTTCTGAGGCGGAAAGAACCAGCTGGGGCTCGATACCGTCGACCTCTAGCTAGAGC


TTGGCGTAATCATGGTCATAGCTGTTTCCTGTGTGAAATTGTTATCCGCTCACAATTCCACACA


ACATACGAGCCGGAAGCATAAAGTGTAAAGCCTAGGGTGCCTAATGAGTGAGCTAACTCACAT


TAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTCGTGCCAGCTGCATTAAT


GAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCTCTTCCGCTTCCTCGCTC


ACTGACTCGCTGCGCTCGGTCGTTCGGCTGCGGCGAGCGGTATCAGCTCACTCAAAGGCGGT


AATACGGTTATCCACAGAATCAGGGGATAACGCAGGAAAGAACATGTGAGCAAAAGGCCAGC


AAAAGGCCAGGAACCGTAAAAAGGCCGCGTTGCTGGCGTTTTTCCATAGGCTCCGCCCCCCT


GACGAGCATCACAAAAATCGACGCTCAAGTCAGAGGTGGCGAAACCCGACAGGACTATAAA


GATACCAGGCGTTTCCCCCTGGAAGCTCCCTCGTGCGCTCTCCTGTTCCGACCCTGCCGCTTA


CCGGATACCTGTCCGCCTTTCTCCCTTCGGGAAGCGTGGCGCTTTCTCATAGCTCACGCTGTAG


GTATCTCAGTTCGGTGTAGGTCGTTCGCTCCAAGCTGGGCTGTGTGCACGAACCCCCCGTTCA


GCCCGACCGCTGCGCCTTATCCGGTAACTATCGTCTTGAGTCCAACCCGGTAAGACACGACTT


ATCGCCACTGGCAGCAGCCACTGGTAACAGGATTAGCAGAGCGAGGTATGTAGGCGGTGCTA


CAGAGTTCTTGAAGTGGTGGCCTAACTACGGCTACACTAGAAGAACAGTATTTGGTATCTGCG


CTCTGCTGAAGCCAGTTACCTTCGGAAAAAGAGTTGGTAGCTCTTGATCCGGCAAACAAACC


ACCGCTGGTAGCGGTGGTTTTTTTGTTTGCAAGCAGCAGATTACGCGCAGAAAAAAAGGATC


TCAAGAAGATCCTTTGATCTTTTCTACGGGGTCTGACGCTCAGTGGAACGAAAACTCACGTTA


AGGGATTTTGGTCATGAGATTATCAAAAAGGATCTTCACCTAGATCCTTTTAAATTAAAAATGA


AGTTTTAAATCAATCTAAAGTATATATGAGTAAACTTGGTCTGACAGTTACCAATGCTTAATCA


GTGAGGCACCTATCTCAGCGATCTGTCTATTTCGTTCATCCATAGTTGCCTGACTCCCCGTCGT


GTAGATAACTACGATACGGGAGGGCTTACCATCTGGCCCCAGTGCTGCAATGATACCGCGAGA


CCCACGCTCACCGGCTCCAGATTTATCAGCAATAAACCAGCCAGCCGGAAGGGCCGAGCGCA


GAAGTGGTCCTGCAACTTTATCCGCCTCCATCCAGTCTATTAATTGTTGCCGGGAAGCTAGAGT


AAGTAGTTCGCCAGTTAATAGTTTGCGCAACGTTGTTGCCATTGCTACAGGCATCGTGGTGTC


ACGCTCGTCGTTTGGTATGGCTTCATTCAGCTCCGGTTCCCAACGATCAAGGCGAGTTACATG


ATCCCCCATGTTGTGCAAAAAAGCGGTTAGCTCCTTCGGTCCTCCGATCGTTGTCAGAAGTAA


GTTGGCCGCAGTGTTATCACTCATGGTTATGGCAGCACTGCATAATTCTCTTACTGTCATGCCAT


CCGTAAGATGCTTTTCTGTGACTGGTGAGTACTCAACCAAGTCATTCTGAGAATAGTGTATGC


GGCGACCGAGTTGCTCTTGCCCGGCGTCAATACGGGATAATACCGCGCCACATAGCAGAACTT


TAAAAGTGCTCATCATTGGAAAACGTTCTTCGGGGCGAAAACTCTCAAGGATCTTACCGCTGT


TGAGATCCAGTTCGATGTAACCCACTCGTGCACCCAACTGATCTTCAGCATCTTTTACTTTCAC


CAGCGTTTCTGGGTGAGCAAAAACAGGAAGGCAAAATGCCGCAAAAAAGGGAATAAGGGCG


ACACGGAAATGTTGAATACTCATACTCTTCCTTTTTCAATATTATTGAAGCATTTATCAGGGTTA


TTGTCTCATGAGCGGATACATATTTGAATGTATTTAGAAAAATAAACAAATAGGGGTTCCGCGC


ACATTTCCCCGAAAAGTGCCACCTGACGTCGACGGATCGGGAGATCGATCTCCCGATCCCCTA


GGGTCGACTCTCAGTACAATCTGCTCTGATGCCGCATAGTTAAGCCAGTATCTGCTCCCTGCTT


GTGTGTTGGAGGTCGCTGAGTAGTGCGCGAGCAAAATTTAAGCTACAACAAGGCAAGGCTTG


ACCGACAATTGCATGAAGAATCTGCTTAGGGTTAGGCGTTTTGCGCTGCTTCGCGATGTACGG


GCCAGATATACGCGTTGACATTGATTATTGACTAGTTATTAATAGTAATCAATTACGGGGTCATT


AGTTCATAGCCCATATATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGA


CCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATA


GGGACTTTCCATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCCCACTTGGCAGTACAT


CAAGTGTATCATATGCCAAGTACGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGG


CATTATGCCCAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACATCTACGTATTAGTCAT


CGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCA


CGGGGATTTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAA


CGGGACTTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTA


CGGTGGGAGGTCTATATAAGCAGAGCTGGTTTAGTGAACCGTCAGATCCGCTAGAGATCCGCG


GCCGCTAATACGACTCACTATAGGGAGAGCCGCCACC





pCMV-PE2:


(SEQ ID NO: 734)



ATGAAACGGACAGCCGACGGAAGCGAGTTCGAGTCACCAAAGAAGAAGCGGAAAGTCGAC



AAGAAGTACAGCATCGGCCTGGACATCGGCACCAACTCTGTGGGCTGGGCCGTGATCACCGA


CGAGTACAAGGTGCCCAGCAAGAAATTCAAGGTGCTGGGCAACACCGACCGGCACAGCATC


AAGAAGAACCTGATCGGAGCCCTGCTGTTCGACAGCGGCGAAACAGCCGAGGCCACCCGGC


TGAAGAGAACCGCCAGAAGAAGATACACCAGACGGAAGAACCGGATCTGCTATCTGCAAGA


GATCTTCAGCAACGAGATGGCCAAGGTGGACGACAGCTTCTTCCACAGACTGGAAGAGTCCT


TCCTGGTGGAAGAGGATAAGAAGCACGAGCGGCACCCCATCTTCGGCAACATCGTGGACGAG


GTGGCCTACCACGAGAAGTACCCCACCATCTACCACCTGAGAAAGAAACTGGTGGACAGCAC


CGACAAGGCCGACCTGCGGCTGATCTATCTGGCCCTGGCCCACATGATCAAGTTCCGGGGCCA


CTTCCTGATCGAGGGCGACCTGAACCCCGACAACAGCGACGTGGACAAGCTGTTCATCCAGC


TGGTGCAGACCTACAACCAGCTGTTCGAGGAAAACCCCATCAACGCCAGCGGCGTGGACGCC


AAGGCCATCCTGTCTGCCAGACTGAGCAAGAGCAGACGGCTGGAAAATCTGATCGCCCAGCT


GCCCGGCGAGAAGAAGAATGGCCTGTTCGGAAACCTGATTGCCCTGAGCCTGGGCCTGACCC


CCAACTTCAAGAGCAACTTCGACCTGGCCGAGGATGCCAAACTGCAGCTGAGCAAGGACAC


CTACGACGACGACCTGGACAACCTGCTGGCCCAGATCGGCGACCAGTACGCCGACCTGTTTC


TGGCCGCCAAGAACCTGTCCGACGCCATCCTGCTGAGCGACATCCTGAGAGTGAACACCGAG


ATCACCAAGGCCCCCCTGAGCGCCTCTATGATCAAGAGATACGACGAGCACCACCAGGACCT


GACCCTGCTGAAAGCTCTCGTGCGGCAGCAGCTGCCTGAGAAGTACAAAGAGATTTTCTTCG


ACCAGAGCAAGAACGGCTACGCCGGCTACATTGACGGCGGAGCCAGCCAGGAAGAGTTCTA


CAAGTTCATCAAGCCCATCCTGGAAAAGATGGACGGCACCGAGGAACTGCTCGTGAAGCTGA


ACAGAGAGGACCTGCTGCGGAAGCAGCGGACCTTCGACAACGGCAGCATCCCCCACCAGAT


CCACCTGGGAGAGCTGCACGCCATTCTGCGGCGGCAGGAAGATTTTTACCCATTCCTGAAGG


ACAACCGGGAAAAGATCGAGAAGATCCTGACCTTCCGCATCCCCTACTACGTGGGCCCTCTG


GCCAGGGGAAACAGCAGATTCGCCTGGATGACCAGAAAGAGCGAGGAAACCATCACCCCCT


GGAACTTCGAGGAAGTGGTGGACAAGGGCGCTTCCGCCCAGAGCTTCATCGAGCGGATGAC


CAACTTCGATAAGAACCTGCCCAACGAGAAGGTGCTGCCCAAGCACAGCCTGCTGTACGAGT


ACTTCACCGTGTATAACGAGCTGACCAAAGTGAAATACGTGACCGAGGGAATGAGAAAGCCC


GCCTTCCTGAGCGGCGAGCAGAAAAAGGCCATCGTGGACCTGCTGTTCAAGACCAACCGGA


AAGTGACCGTGAAGCAGCTGAAAGAGGACTACTTCAAGAAAATCGAGTGCTTCGACTCCGTG


GAAATCTCCGGCGTGGAAGATCGGTTCAACGCCTCCCTGGGCACATACCACGATCTGCTGAA


AATTATCAAGGACAAGGACTTCCTGGACAATGAGGAAAACGAGGACATTCTGGAAGATATCG


TGCTGACCCTGACACTGTTTGAGGACAGAGAGATGATCGAGGAACGGCTGAAAACCTATGCC


CACCTGTTCGACGACAAAGTGATGAAGCAGCTGAAGCGGCGGAGATACACCGGCTGGGGCA


GGCTGAGCCGGAAGCTGATCAACGGCATCCGGGACAAGCAGTCCGGCAAGACAATCCTGGAT


TTCCTGAAGTCCGACGGCTTCGCCAACAGAAACTTCATGCAGCTGATCCACGACGACAGCCT


GACCTTTAAAGAGGACATCCAGAAAGCCCAGGTGTCCGGCCAGGGCGATAGCCTGCACGAGC


ACATTGCCAATCTGGCCGGCAGCCCCGCCATTAAGAAGGGCATCCTGCAGACAGTGAAGGTG


GTGGACGAGCTCGTGAAAGTGATGGGCCGGCACAAGCCCGAGAACATCGTGATCGAAATGG


CCAGAGAGAACCAGACCACCCAGAAGGGACAGAAGAACAGCCGCGAGAGAATGAAGCGGA


TCGAAGAGGGCATCAAAGAGCTGGGCAGCCAGATCCTGAAAGAACACCCCGTGGAAAACAC


CCAGCTGCAGAACGAGAAGCTGTACCTGTACTACCTGCAGAATGGGCGGGATATGTACGTGG


ACCAGGAACTGGACATCAACCGGCTGTCCGACTACGATGTGGACGCTATCGTGCCTCAGAGC


TTTCTGAAGGACGACTCCATCGACAACAAGGTGCTGACCAGAAGCGACAAGAACCGGGGCA


AGAGCGACAACGTGCCCTCCGAAGAGGTCGTGAAGAAGATGAAGAACTACTGGCGGCAGCT


GCTGAACGCCAAGCTGATTACCCAGAGAAAGTTCGACAATCTGACCAAGGCCGAGAGAGGC


GGCCTGAGCGAACTGGATAAGGCCGGCTTCATCAAGAGACAGCTGGTGGAAACCCGGCAGAT


CACAAAGCACGTGGCACAGATCCTGGACTCCCGGATGAACACTAAGTACGACGAGAATGACA


AGCTGATCCGGGAAGTGAAAGTGATCACCCTGAAGTCCAAGCTGGTGTCCGATTTCCGGAAG


GATTTCCAGTTTTACAAAGTGCGCGAGATCAACAACTACCACCACGCCCACGACGCCTACCTG


AACGCCGTCGTGGGAACCGCCCTGATCAAAAAGTACCCTAAGCTGGAAAGCGAGTTCGTGTA


CGGCGACTACAAGGTGTACGACGTGCGGAAGATGATCGCCAAGAGCGAGCAGGAAATCGGC


AAGGCTACCGCCAAGTACTTCTTCTACAGCAACATCATGAACTTTTTCAAGACCGAGATTACC


CTGGCCAACGGCGAGATCCGGAAGCGGCCTCTGATCGAGACAAACGGCGAAACCGGGGAGA


TCGTGTGGGATAAGGGCCGGGATTTTGCCACCGTGCGGAAAGTGCTGAGCATGCCCCAAGTG


AATATCGTGAAAAAGACCGAGGTGCAGACAGGCGGCTTCAGCAAAGAGTCTATCCTGCCCAA


GAGGAACAGCGATAAGCTGATCGCCAGAAAGAAGGACTGGGACCCTAAGAAGTACGGCGGC


TTCGACAGCCCCACCGTGGCCTATTCTGTGCTGGTGGTGGCCAAAGTGGAAAAGGGCAAGTC


CAAGAAACTGAAGAGTGTGAAAGAGCTGCTGGGGATCACCATCATGGAAAGAAGCAGCTTC


GAGAAGAATCCCATCGACTTTCTGGAAGCCAAGGGCTACAAAGAAGTGAAAAAGGACCTGA


TCATCAAGCTGCCTAAGTACTCCCTGTTCGAGCTGGAAAACGGCCGGAAGAGAATGCTGGCC


TCTGCCGGCGAACTGCAGAAGGGAAACGAACTGGCCCTGCCCTCCAAATATGTGAACTTCCT


GTACCTGGCCAGCCACTATGAGAAGCTGAAGGGCTCCCCCGAGGATAATGAGCAGAAACAGC


TGTTTGTGGAACAGCACAAGCACTACCTGGACGAGATCATCGAGCAGATCAGCGAGTTCTCC


AAGAGAGTGATCCTGGCCGACGCTAATCTGGACAAAGTGCTGTCCGCCTACAACAAGCACCG


GGATAAGCCCATCAGAGAGCAGGCCGAGAATATCATCCACCTGTTTACCCTGACCAATCTGGG


AGCCCCTGCCGCCTTCAAGTACTTTGACACCACCATCGACCGGAAGAGGTACACCAGCACCA


AAGAGGTGCTGGACGCCACCCTGATCCACCAGAGCATCACCGGCCTGTACGAGACACGGATC


GACCTGTCTCAGCTGGGAGGTGACTCTGGAGGATCTAGCGGAGGATCCTCTGGCAGCGAGAC


ACCAGGAACAAGCGAGTCAGCAACACCAGAGAGCAGTGGCGGCAGCAGCGGCGGCAGCAG


CACCCTAAATATAGAAGATGAGTATCGGCTACATGAGACCTCAAAAGAGCCAGATGTTTCTCT


AGGGTCCACATGGCTGTCTGATTTTCCTCAGGCCTGGGCGGAAACCGGGGGCATGGGACTGG


CAGTTCGCCAAGCTCCTCTGATCATACCTCTGAAAGCAACCTCTACCCCCGTGTCCATAAAAC


AATACCCCATGTCACAAGAAGCCAGACTGGGGATCAAGCCCCACATACAGAGACTGTTGGAC


CAGGGAATACTGGTACCCTGCCAGTCCCCCTGGAACACGCCCCTGCTACCCGTTAAGAAACC


AGGGACTAATGATTATAGGCCTGTCCAGGATCTGAGAGAAGTCAACAAGCGGGTGGAAGACA


TCCACCCCACCGTGCCCAACCCTTACAACCTCTTGAGCGGGCTCCCACCGTCCCACCAGTGGT


ACACTGTGCTTGATTTAAAGGATGCCTTTTTCTGCCTGAGACTCCACCCCACCAGTCAGCCTC


TCTTCGCCTTTGAGTGGAGAGATCCAGAGATGGGAATCTCAGGACAATTGACCTGGACCAGA


CTCCCACAGGGTTTCAAAAACAGTCCCACCCTGTTTAATGAGGCACTGCACAGAGACCTAGC


AGACTTCCGGATCCAGCACCCAGACTTGATCCTGCTACAGTACGTGGATGACTTACTGCTGGC


CGCCACTTCTGAGCTAGACTGCCAACAAGGTACTCGGGCCCTGTTACAAACCCTAGGGAACC


TCGGGTATCGGGCCTCGGCCAAGAAAGCCCAAATTTGCCAGAAACAGGTCAAGTATCTGGGG


TATCTTCTAAAAGAGGGTCAGAGATGGCTGACTGAGGCCAGAAAAGAGACTGTGATGGGGCA


GCCTACTCCGAAGACCCCTCGACAACTAAGGGAGTTCCTAGGGAAGGCAGGCTTCTGTCGCC


TCTTCATCCCTGGGTTTGCAGAAATGGCAGCCCCCCTGTACCCTCTCACCAAACCGGGGACTC


TGTTTAATTGGGGCCCAGACCAACAAAAGGCCTATCAAGAAATCAAGCAAGCTCTTCTAACTG


CCCCAGCCCTGGGGTTGCCAGATTTGACTAAGCCCTTTGAACTCTTTGTCGACGAGAAGCAG


GGCTACGCCAAAGGTGTCCTAACGCAAAAACTGGGACCTTGGCGTCGGCCGGTGGCCTACCT


GTCCAAAAAGCTAGACCCAGTAGCAGCTGGGTGGCCCCCTTGCCTACGGATGGTAGCAGCCA


TTGCCGTACTGACAAAGGATGCAGGCAAGCTAACCATGGGACAGCCACTAGTCATTCTGGCC


CCCCATGCAGTAGAGGCACTAGTCAAACAACCCCCCGACCGCTGGCTTTCCAACGCCCGGAT


GACTCACTATCAGGCCTTGCTTTTGGACACGGACCGGGTCCAGTTCGGACCGGTGGTAGCCCT


GAACCCGGCTACGCTGCTCCCACTGCCTGAGGAAGGGCTGCAACACAACTGCCTTGATATCC


TGGCCGAAGCCCACGGAACCCGACCCGACCTAACGGACCAGCCGCTCCCAGACGCCGACCA


CACCTGGTACACGGATGGAAGCAGTCTCTTACAAGAGGGACAGCGTAAGGCGGGAGCTGCG


GTGACCACCGAGACCGAGGTAATCTGGGCTAAAGCCCTGCCAGCCGGGACATCCGCTCAGCG


GGCTGAACTGATAGCACTCACCCAGGCCCTAAAGATGGCAGAAGGTAAGAAGCTAAATGTTT


ATACTGATAGCCGTTATGCTTTTGCTACTGCCCATATCCATGGAGAAATATACAGAAGGCGTGG


GTGGCTCACATCAGAAGGCAAAGAGATCAAAAATAAAGACGAGATCTTGGCCCTACTAAAAG


CCCTCTTTCTGCCCAAAAGACTTAGCATAATCCATTGTCCAGGACATCAAAAGGGACACAGCG


CCGAGGCTAGAGGCAACCGGATGGCTGACCAAGCGGCCCGAAAGGCAGCCATCACAGAGAC


TCCAGACACCTCTACCCTCCTCATAGAAAATTCATCACCCTCTGGCGGCTCAAAAAGAACCGC


CGACGGCAGCGAATTCGAGCCCAAGAAGAAGAGGAAAGTCTAACCGGTCATCATCACCATCA


CCATTGAGTTTAAACCCGCTGATCAGCCTCGACTGTGCCTTCTAGTTGCCAGCCATCTGTTGTT


TGCCCCTCCCCCGTGCCTTCCTTGACCCTGGAAGGTGCCACTCCCACTGTCCTTTCCTAATAA


AATGAGAAAATTGCATCGCATTGTCTGAGTAGGTGTCATTCTATTCTGGGGGGTGGGGTGGGG


CAGGACAGCAAGGGGGAGGATTGGGAAGACAATAGCAGGCATGCTGGGGATGCGGTGGGCT


CTATGGCTTCTGAGGCGGAAAGAACCAGCTGGGGCTCGATACCGTCGACCTCTAGCTAGAGC


TTGGCGTAATCATGGTCATAGCTGTTTCCTGTGTGAAATTGTTATCCGCTCACAATTCCACACA


ACATACGAGCCGGAAGCATAAAGTGTAAAGCCTAGGGTGCCTAATGAGTGAGCTAACTCACAT


TAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTCGTGCCAGCTGCATTAAT


GAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCTCTTCCGCTTCCTCGCTC


ACTGACTCGCTGCGCTCGGTCGTTCGGCTGCGGCGAGCGGTATCAGCTCACTCAAAGGCGGT


AATACGGTTATCCACAGAATCAGGGGATAACGCAGGAAAGAACATGTGAGCAAAAGGCCAGC


AAAAGGCCAGGAACCGTAAAAAGGCCGCGTTGCTGGCGTTTTTCCATAGGCTCCGCCCCCCT


GACGAGCATCACAAAAATCGACGCTCAAGTCAGAGGTGGCGAAACCCGACAGGACTATAAA


GATACCAGGCGTTTCCCCCTGGAAGCTCCCTCGTGCGCTCTCCTGTTCCGACCCTGCCGCTTA


CCGGATACCTGTCCGCCTTTCTCCCTTCGGGAAGCGTGGCGCTTTCTCATAGCTCACGCTGTAG


GTATCTCAGTTCGGTGTAGGTCGTTCGCTCCAAGCTGGGCTGTGTGCACGAACCCCCCGTTCA


GCCCGACCGCTGCGCCTTATCCGGTAACTATCGTCTTGAGTCCAACCCGGTAAGACACGACTT


ATCGCCACTGGCAGCAGCCACTGGTAACAGGATTAGCAGAGCGAGGTATGTAGGCGGTGCTA


CAGAGTTCTTGAAGTGGTGGCCTAACTACGGCTACACTAGAAGAACAGTATTTGGTATCTGCG


CTCTGCTGAAGCCAGTTACCTTCGGAAAAAGAGTTGGTAGCTCTTGATCCGGCAAACAAACC


ACCGCTGGTAGCGGTGGTTTTTTTGTTTGCAAGCAGCAGATTACGCGCAGAAAAAAAGGATC


TCAAGAAGATCCTTTGATCTTTTCTACGGGGTCTGACGCTCAGTGGAACGAAAACTCACGTTA


AGGGATTTTGGTCATGAGATTATCAAAAAGGATCTTCACCTAGATCCTTTTAAATTAAAAATGA


AGTTTTAAATCAATCTAAAGTATATATGAGTAAACTTGGTCTGACAGTTACCAATGCTTAATCA


GTGAGGCACCTATCTCAGCGATCTGTCTATTTCGTTCATCCATAGTTGCCTGACTCCCCGTCGT


GTAGATAACTACGATACGGGAGGGCTTACCATCTGGCCCCAGTGCTGCAATGATACCGCGAGA


CCCACGCTCACCGGCTCCAGATTTATCAGCAATAAACCAGCCAGCCGGAAGGGCCGAGCGCA


GAAGTGGTCCTGCAACTTTATCCGCCTCCATCCAGTCTATTAATTGTTGCCGGGAAGCTAGAGT


AAGTAGTTCGCCAGTTAATAGTTTGCGCAACGTTGTTGCCATTGCTACAGGCATCGTGGTGTC


ACGCTCGTCGTTTGGTATGGCTTCATTCAGCTCCGGTTCCCAACGATCAAGGCGAGTTACATG


ATCCCCCATGTTGTGCAAAAAAGCGGTTAGCTCCTTCGGTCCTCCGATCGTTGTCAGAAGTAA


GTTGGCCGCAGTGTTATCACTCATGGTTATGGCAGCACTGCATAATTCTCTTACTGTCATGCCAT


CCGTAAGATGCTTTTCTGTGACTGGTGAGTACTCAACCAAGTCATTCTGAGAATAGTGTATGC


GGCGACCGAGTTGCTCTTGCCCGGCGTCAATACGGGATAATACCGCGCCACATAGCAGAACTT


TAAAAGTGCTCATCATTGGAAAACGTTCTTCGGGGCGAAAACTCTCAAGGATCTTACCGCTGT


TGAGATCCAGTTCGATGTAACCCACTCGTGCACCCAACTGATCTTCAGCATCTTTTACTTTCAC


CAGCGTTTCTGGGTGAGCAAAAACAGGAAGGCAAAATGCCGCAAAAAAGGGAATAAGGGCG


ACACGGAAATGTTGAATACTCATACTCTTCCTTTTTCAATATTATTGAAGCATTTATCAGGGTTA


TTGTCTCATGAGCGGATACATATTTGAATGTATTTAGAAAAATAAACAAATAGGGGTTCCGCGC


ACATTTCCCCGAAAAGTGCCACCTGACGTCGACGGATCGGGAGATCGATCTCCCGATCCCCTA


GGGTCGACTCTCAGTACAATCTGCTCTGATGCCGCATAGTTAAGCCAGTATCTGCTCCCTGCTT


GTGTGTTGGAGGTCGCTGAGTAGTGCGCGAGCAAAATTTAAGCTACAACAAGGCAAGGCTTG


ACCGACAATTGCATGAAGAATCTGCTTAGGGTTAGGCGTTTTGCGCTGCTTCGCGATGTACGG


GCCAGATATACGCGTTGACATTGATTATTGACTAGTTATTAATAGTAATCAATTACGGGGTCATT


AGTTCATAGCCCATATATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGA


CCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATA


GGGACTTTCCATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCCCACTTGGCAGTACAT


CAAGTGTATCATATGCCAAGTACGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGG


CATTATGCCCAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACATCTACGTATTAGTCAT


CGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCA


CGGGGATTTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAA


CGGGACTTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTA


CGGTGGGAGGTCTATATAAGCAGAGCTGGTTTAGTGAACCGTCAGATCCGCTAGAGATCCGCG


GCCGCTAATACGACTCACTATAGGGAGAGCCGCCACC






N-terminal NLS + Cas9 H840A




Flexible linker





M-MLV reverse transcriptase + C-terminal NLS




Plasmid backbone (containing CMV promoter and AmpR)





pU6-HEK3_PEgRNA_CTTins:


(SEQ ID NO: 735)



GAGGGCCTATTTCCCATGATTCCTTCATATTTGCATATACGATACAAGGCTGTTAGAGAGATAAT



TAGAATTAATTTGACTGTAAACACAAAGATATTAGTACAAAATACGTGACGTAGAAAGTAATAA


TTTCTTGGGTAGTTTGCAGTTTTAAAATTATGTTTTAAAATGGACTATCATATGCTTACCGTAAC


TTGAAAGTATTTCGATTTCTTGGCTTTATATATCTTGTGGAAAGGACGAAACACCGGCCCAGAC




TGAGCACGTGA
GTTTTAGAGCTAGAAATAGCAAGTTAAAATAAGGCTAGTCCGTTATCAACTTG



AAAAAGTGGGACCGAGTCGGTCCTCTGCCATCAAAGCGTGCTCAGTCTGTTTTTTTAAGCT


TGGGCCGCTCGAGGTACCTCTCTACATATGACATGTGAGCAAAAGGCCAGCAAAAGGCCAGG


AACCGTAAAAAGGCCGCGTTGCTGGCGTTTTTCCATAGGCTCCGCCCCCCTGACGAGCATCAC


AAAAATCGACGCTCAAGTCAGAGGTGGCGAAACCCGACAGGACTATAAAGATACCAGGCGTT


TCCCCCTGGAAGCTCCCTCGTGCGCTCTCCTGTTCCGACCCTGCCGCTTACCGGATACCTGTC


CGCCTTTCTCCCTTCGGGAAGCGTGGCGCTTTCTCATAGCTCACGCTGTAGGTATCTCAGTTCG


GTGTAGGTCGTTCGCTCCAAGCTGGGCTGTGTGCACGAACCCCCCGTTCAGCCCGACCGCTG


CGCCTTATCCGGTAACTATCGTCTTGAGTCCAACCCGGTAAGACACGACTTATCGCCACTGGC


AGCAGCCACTGGTAACAGGATTAGCAGAGCGAGGTATGTAGGCGGTGCTACAGAGTTCTTGA


AGTGGTGGCCTAACTACGGCTACACTAGAAGAACAGTATTTGGTATCTGCGCTCTGCTGAAGC


CAGTTACCTTCGGAAAAAGAGTTGGTAGCTCTTGATCCGGCAAACAAACCACCGCTGGTAGC


GGTGGTTTTTTTGTTTGCAAGCAGCAGATTACGCGCAGAAAAAAAGGATCTCAAGAAGATCC


TTTGATCTTTTCTACGGGGTCTGACGCTCAGTGGAACGAAAACTCACGTTAAGGGATTTTGGT


CATGAGATTATCAAAAAGGATCTTCACCTAGATCCTTTTAAATTAAAAATGAAGTTTTAAATCA


ATCTAAAGTATATATGAGTAAACTTGGTCTGACAGTTACCAATGCTTAATCAGTGAGGCACCTA


TCTCAGCGATCTGTCTATTTCGTTCATCCATAGTTGCCTGACTCCCCGTCGTGTAGATAACTACG


ATACGGGAGGGCTTACCATCTGGCCCCAGTGCTGCAATGATACCGCGAGACCCACGCTCACCG


GCTCCAGATTTATCAGCAATAAACCAGCCAGCCGGAAGGGCCGAGCGCAGAAGTGGTCCTGC


AACTTTATCCGCCTCCATCCAGTCTATTAATTGTTGCCGGGAAGCTAGAGTAAGTAGTTCGCCA


GTTAATAGTTTGCGCAACGTTGTTGCCATTGCTACAGGCATCGTGGTGTCACGCTCGTCGTTTG


GTATGGCTTCATTCAGCTCCGGTTCCCAACGATCAAGGCGAGTTACATGATCCCCCATGTTGTG


CAAAAAAGCGGTTAGCTCCTTCGGTCCTCCGATCGTTGTCAGAAGTAAGTTGGCCGCAGTGTT


ATCACTCATGGTTATGGCAGCACTGCATAATTCTCTTACTGTCATGCCATCCGTAAGATGCTTTT


CTGTGACTGGTGAGTACTCAACCAAGTCATTCTGAGAATAGTGTATGCGGCGACCGAGTTGCT


CTTGCCCGGCGTCAATACGGGATAATACCGCGCCACATAGCAGAACTTTAAAAGTGCTCATCA


TTGGAAAACGTTCTTCGGGGCGAAAACTCTCAAGGATCTTACCGCTGTTGAGATCCAGTTCGA


TGTAACCCACTCGTGCACCCAACTGATCTTCAGCATCTTTTACTTTCACCAGCGTTTCTGGGTG


AGCAAAAACAGGAAGGCAAAATGCCGCAAAAAAGGGAATAAGGGCGACACGGAAATGTTG


AATACTCATACTCTTCCTTTTTCAATATTATTGAAGCATTTATCAGGGTTATTGTCTCATGAGCGG


ATACATATTTGAATGTATTTAGAAAAATAAACAAATAGGGGTTCCGCGCACATTTCCCCGAAAA


GTGCCACCTGACGTCGCTAGCTGTACAAAAAAGCAGGCTTTAAAGGAACCAATTCAGTCGAC


TGGATCCGGTACCAAGGTCGGGCAGGAA





U6 Promoter sequence




Spacer sequence




sgRNA scaffold




3

 extension (contains PBS and RT template)




Backbone (contains AmpR)





pLenti-hSyn-N-PE2-NpuN-P2A-GFP-KASH_U6-DNMT1-PEgRNA:


(SEQ ID NO: 736)



GTCGACGGATCGGGAGATCTCCCGATCCCCTATGGTGCACTCTCAGTACAATCTGCTCTGATGCCGC



ATAGTTAAGCCAGTATCTGCTCCCTGCTTGTGTGTTGGAGGTCGCTGAGTAGTGCGCGAGCAAAATT


TAAGCTACAACAAGGCAAGGCTTGACCGACAATTGCATGAAGAATCTGCTTAGGGTTAGGCGTTTT


GCGCTGCTTCGCGATGTACGGGCCAGATATACGCGTTGACATTGATTATTGACTAGTTATTAATAGTA


ATCAATTACGGGGTCATTAGTTCATAGCCCATATATGGAGTTCCGCGTTACATAACTTACGGTAAATG


GCCCGCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGT


AACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCCCACTTGGC


AGTACATCAAGTGTATCATATGCCAAGTACGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCC


TGGCATTATGCCCAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACATCTACGTATTAGTCAT


CGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCACGG


GGATTTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGAC


TTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGAG


GTCTATATAAGCAGCGCGTTTTGCCTGTACTGGGTCTCTCTGGTTAGACCAGATCTGAGCCTGGGAG


CTCTCTGGCTAACTAGGGAACCCACTGCTTAAGCCTCAATAAAGCTTGCCTTGAGTGCTTCAAGTA


GTGTGTGCCCGTCTGTTGTGTGACTCTGGTAACTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGGA


AAATCTCTAGCAGTGGCGCCCGAACAGGGACTTGAAAGCGAAAGGGAAACCAGAGGAGCTCTCT


CGACGCAGGACTCGGCTTGCTGAAGCGCGCACGGCAAGAGGCGAGGGGCGGCGACTGGTGAGTA


CGCCAAAAATTTTGACTAGCGGAGGCTAGAAGGAGAGAGATGGGTGCGAGAGCGTCAGTATTAAG


CGGGGGAGAATTAGATCGCGATGGGAAAAAATTCGGTTAAGGCCAGGGGGAAAGAAAAAATATAA


ATTAAAACATATAGTATGGGCAAGCAGGGAGCTAGAACGATTCGCAGTTAATCCTGGCCTGTTAGAA


ACATCAGAAGGCTGTAGACAAATACTGGGACAGCTACAACCATCCCTTCAGACAGGATCAGAAGA


ACTTAGATCATTATATAATACAGTAGCAACCCTCTATTGTGTGCATCAAAGGATAGAGATAAAAGACA


CCAAGGAAGCTTTAGACAAGATAGAGGAAGAGCAAAACAAAAGTAAGACCACCGCACAGCAAGC


GGCCGCTGATCTTCAGACCTGGAGGAGGAGATATGAGGGACAATTGGAGAAGTGAATTATATAAAT


ATAAAGTAGTAAAAATTGAACCATTAGGAGTAGCACCCACCAAGGCAAAGAGAAGAGTGGTGCAG


AGAGAAAAAAGAGCAGTGGGAATAGGAGCTTTGTTCCTTGGGTTCTTGGGAGCAGCAGGAAGCAC


TATGGGCGCAGCGTCAATGACGCTGACGGTACAGGCCAGACAATTATTGTCTGGTATAGTGCAGCA


GCAGAACAATTTGCTGAGGGCTATTGAGGCGCAACAGCATCTGTTGCAACTCACAGTCTGGGGCAT


CAAGCAGCTCCAGGCAAGAATCCTGGCTGTGGAAAGATACCTAAAGGATCAACAGCTCCTGGGGA


TTTGGGGTTGCTCTGGAAAACTCATTTGCACCACTGCTGTGCCTTGGAATGCTAGTTGGAGTAATAA


ATCTCTGGAACAGATTTGGAATCACACGACCTGGATGGAGTGGGACAGAGAAATTAACAATTACAC


AAGCTTAATACACTCCTTAATTGAAGAATCGCAAAACCAGCAAGAAAAGAATGAACAAGAATTATT


GGAATTAGATAAATGGGCAAGTTTGTGGAATTGGTTTAACATAACAAATTGGCTGTGGTATATAAAA


TTATTCATAATGATAGTAGGAGGCTTGGTAGGTTTAAGAATAGTTTTTGCTGTACTTTCTATAGTGAAT


AGAGTTAGGCAGGGATATTCACCATTATCGTTTCAGACCCACCTCCCAACCCCGAGGGGACCCGAC


AGGCCCGAAGGAATAGAAGAAGAAGGTGGAGAGAGAGACAGAGACAGATCCATTCGATTAGTGA


ACGGATCGGCACTGCGTGCGCCAATTCTGCAGACAAATGGCAGTATTCATCCACAATTTTAAAAGA


AAAGGGGGGATTGGGGGGTACAGTGCAGGGGAAAGAATAGTAGACATAATAGCAACAGACATACA


AACTAAAGAATTACAAAAACAAATTACAAAAATTCAAAATTTTCGGGTTTATTACAGGGACAGCAG


AGATCCAGTTTGGTTAATTAAGGTACCAAGGTCGGGCAGGAAGAGGGCCTATTTCCCATGATTCCTT



CATATTTGCATATACGATACAAGGCTGTTAGAGAGATAATTAGAATTAATTTGACTGTAAACACAAAG




ATATTAGTACAAAATACGTGACGTAGAAAGTAATAATTTCTTGGGTAGTTTGCAGTTTTAAAATTATG




TTTTAAAATGGACTATCATATGCTTACCGTAACTTGAAAGTATTTCGATTTCTTGGCTTTATATATCTTG




TGGAAAGGACGAAACACC
GCGGGCTGGAGCTGTTCGCGCGTTTTAGAGCTAGAAATAGCAAG




TTAAAATAAGGCTAGTCCGTTATCAACTTGAAAAAGTGGCACCGAGTCGGTGCAAGATGCAA




GCGCGAACAGCTCCAG

TTTTTTTGAATTC

AGTGCAAGTGGGTTTTAGGACCAGGATGAGGCGGGG




TGGGGGTGCCTACCTGACGACCGACCCCGACCCACTGGACAAGCACCCAACCCCCATTCCCCAAATTGC




GCATCCCCTATCAGAGAGGGGGAGGGGAAACAGGATGCGGCGAGGCGCGTGCGCACTGCCAGCTTCA




GCACCGCGGACAGTGCCTTCGCCCCCGCCTGGCGGCGCGCGCCACCGCCGCCTCAGCACTGAAGGCG




CGCTGACGTCACTCGCCGGTCCCCCGCAAACTCCCCTTCCCGGCCACCTTGGTCGCGTCCGCGCCGCC




GCCGGCCCAGCCGGACCGCACCACGCGAGGCGCGAGATAGGGGGGCACGGGCGCGACCATCTGCGC




TGCGGCGCCGGCGACTCAGCGCTGCCTCAGTCTGCGGTGGGCAGCGGAGGAGTCGTGTCGTGCCTGA




GAGCGCAGcustom character ATGAAACGGACAGCCGACGGAAGCGAGTTCGAG



TCACCAAAGAAGAAGCGGAAAGTCGACAAGAAGTACAGCATCGGCCTGGACATCGGCACCAACT


CTGTGGGCTGGGCCGTGATCACCGACGAGTACAAGGTGCCCAGCAAGAAATTCAAGGTGCTGGGC


AACACCGACCGGCACAGCATCAAGAAGAACCTGATCGGAGCCCTGCTGTTCGACAGCGGCGAAAC


AGCCGAGGCCACCCGGCTGAAGAGAACCGCCAGAAGAAGATACACCAGACGGAAGAACCGGATC


TGCTATCTGCAAGAGATCTTCAGCAACGAGATGGCCAAGGTGGACGACAGCTTCTTCCACAGACTG


GAAGAGTCCTTCCTGGTGGAAGAGGATAAGAAGCACGAGCGGCACCCCATCTTCGGCAACATCGT


GGACGAGGTGGCCTACCACGAGAAGTACCCCACCATCTACCACCTGAGAAAGAAACTGGTGGACA


GCACCGACAAGGCCGACCTGCGGCTGATCTATCTGGCCCTGGCCCACATGATCAAGTTCCGGGGCC


ACTTCCTGATCGAGGGCGACCTGAACCCCGACAACAGCGACGTGGACAAGCTGTTCATCCAGCTG


GTGCAGACCTACAACCAGCTGTTCGAGGAAAACCCCATCAACGCCAGCGGCGTGGACGCCAAGGC


CATCCTGTCTGCCAGACTGAGCAAGAGCAGACGGCTGGAAAATCTGATCGCCCAGCTGCCCGGCG


AGAAGAAGAATGGCCTGTTCGGAAACCTGATTGCCCTGAGCCTGGGCCTGACCCCCAACTTCAAG


AGCAACTTCGACCTGGCCGAGGATGCCAAACTGCAGCTGAGCAAGGACACCTACGACGACGACCT


GGACAACCTGCTGGCCCAGATCGGCGACCAGTACGCCGACCTGTTTCTGGCCGCCAAGAACCTGT


CCGACGCCATCCTGCTGAGCGACATCCTGAGAGTGAACACCGAGATCACCAAGGCCCCCCTGAGC


GCCTCTATGATCAAGAGATACGACGAGCACCACCAGGACCTGACCCTGCTGAAAGCTCTCGTGCGG


CAGCAGCTGCCTGAGAAGTACAAAGAGATTTTCTTCGACCAGAGCAAGAACGGCTACGCCGGCTA


CATTGACGGCGGAGCCAGCCAGGAAGAGTTCTACAAGTTCATCAAGCCCATCCTGGAAAAGATGG


ACGGCACCGAGGAACTGCTCGTGAAGCTGAACAGAGAGGACCTGCTGCGGAAGCAGCGGACCTT


CGACAACGGCAGCATCCCCCACCAGATCCACCTGGGAGAGCTGCACGCCATTCTGCGGCGGCAGG


AAGATTTTTACCCATTCCTGAAGGACAACCGGGAAAAGATCGAGAAGATCCTGACCTTCCGCATCC


CCTACTACGTGGGCCCTCTGGCCAGGGGAAACAGCAGATTCGCCTGGATGACCAGAAAGAGCGAG


GAAACCATCACCCCCTGGAACTTCGAGGAAGTGGTGGACAAGGGCGCTTCCGCCCAGAGCTTCAT


CGAGCGGATGACCAACTTCGATAAGAACCTGCCCAACGAGAAGGTGCTGCCCAAGCACAGCCTGC


TGTACGAGTACTTCACCGTGTATAACGAGCTGACCAAAGTGAAATACGTGACCGAGGGAATGAGAA


AGCCCGCCTTCCTGAGCGGCGAGCAGAAAAAGGCCATCGTGGACCTGCTGTTCAAGACCAACCGG


AAAGTGACCGTGAAGCAGCTGAAAGAGGACTACTTCAAGAAAATCGAGTGCCTGTCCTACGAGACA




GAGATCCTGACAGTGGAGTATGGCCTGCTGCCAATCGGCAAGATCGTGGAGAAGAGGATCGAGTGTACC






GTGTACTCTGTGGATAACAATGGCAACATCTATACACAGCCCGTGGCACAGTGGCACGATAGGGGAGAGC






AGGAGGTGTTCGAGTATTGCCTGGAGGACGGCAGCCTGATCAGGGCAACCAAGGACCACAAGTTCATGA






CAGTGGATGGCCAGATGCTGCCCATCGACGAGATTTTCGAGCGGGAGCTGGACCTGATGAGAGTGGATA






ACCTGCCTAAT

custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character




custom character
custom character
custom character ACGC



GTTAAGTCGACAATCAACCTCTGGATTACAAAATTTGTGAAAGATTGACTGGTATTCTTAACTATGTT


GCTCCTTTTACGCTATGTGGATACGCTGCTTTAATGCCTTTGTATCATGCTATTGCTTCCCGTATGGCT


TTCATTTTCTCCTCCTTGTATAAATCCTGGTTGCTGTCTCTTTATGAGGAGTTGTGGCCCGTTGTCAG


GCAACGTGGCGTGGTGTGCACTGTGTTTGCTGACGCAACCCCCACTGGTTGGGGCATTGCCACCAC


CTGTCAGCTCCTTTCCGGGACTTTCGCTTTCCCCCTCCCTATTGCCACGGCGGAACTCATCGCCGCC


TGCCTTGCCCGCTGCTGGACAGGGGCTCGGCTGTTGGGCACTGACAATTCCGTGGTGTTGTCGGGG


AAATCATCGTCCTTTCCTTGGCTGCTCGCCTGTGTTGCCACCTGGATTCTGCGCGGGACGTCCTTCT


GCTACGTCCCTTCGGCCCTCAATCCAGCGGACCTTCCTTCCCGCGGCCTGCTGCCGGCTCTGCGGC


CTCTTCCGCGTCTTCGCCTTCGCCCTCAGACGAGTCGGATCTCCCTTTGGGCCGCCTCCCCGCGTCG


ACTTTAAGACCAATGACTTACAAGGCAGCTGTAGATCTTAGCCACTTTTTAAAAGAAAAGGGGGGA


CTGGAAGGGCTAATTCACTCCCAACGAAGACAAGATCTGCTTTTTGCTTGTACTGGGTCTCTCTGGT


TAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAACTAGGGAACCCACTGCTTAAGCCTCAATAAA


GCTTGCCTTGAGTGCTTCAAGTAGTGTGTGCCCGTCTGTTGTGTGACTCTGGTAACTAGAGATCCCT


CAGACCCTTTTAGTCAGTGTGGAAAATCTCTAGCAGGGCCCGTTTAAACCCGCTGATCAGCCTCGA


CTGTGCCTTCTAGTTGCCAGCCATCTGTTGTTTGCCCCTCCCCCGTGCCTTCCTTGACCCTGGAAGG


TGCCACTCCCACTGTCCTTTCCTAATAAAATGAGGAAATTGCATCGCATTGTCTGAGTAGGTGTCAT


TCTATTCTGGGGGGTGGGGTGGGGCAGGACAGCAAGGGGGAGGATTGGGAAGACAATAGCAGGC


ATGCTGGGGATGCGGTGGGCTCTATGGCTTCTGAGGCGGAAAGAACCAGCTGGGGCTCTAGGGGG


TATCCCCACGCGCCCTGTAGCGGCGCATTAAGCGCGGCGGGTGTGGTGGTTACGCGCAGCGTGACC


GCTACACTTGCCAGCGCCCTAGCGCCCGCTCCTTTCGCTTTCTTCCCTTCCTTTCTCGCCACGTTCG


CCGGCTTTCCCCGTCAAGCTCTAAATCGGGGGCTCCCTTTAGGGTTCCGATTTAGTGCTTTACGGCA


CCTCGACCCCAAAAAACTTGATTAGGGTGATGGTTCACGTAGTGGGCCATCGCCCTGATAGACGGT


TTTTCGCCCTTTGACGTTGGAGTCCACGTTCTTTAATAGTGGACTCTTGTTCCAAACTGGAACAACA


CTCAACCCTATCTCGGTCTATTCTTTTGATTTATAAGGGATTTTGCCGATTTCGGCCTATTGGTTAAAA


AATGAGCTGATTTAACAAAAATTTAACGCGAATTAATTCTGTGGAATGTGTGTCAGTTAGGGTGTGG


AAAGTCCCCAGGCTCCCCAGCAGGCAGAAGTATGCAAAGCATGCATCTCAATTAGTCAGCAACCAG


GTGTGGAAAGTCCCCAGGCTCCCCAGCAGGCAGAAGTATGCAAAGCATGCATCTCAATTAGTCAGC


AACCATAGTCCCGCCCCTAACTCCGCCCATCCCGCCCCTAACTCCGCCCAGTTCCGCCCATTCTCCG


CCCCATGGCTGACTAATTTTTTTTATTTATGCAGAGGCCGAGGCCGCCTCTGCCTCTGAGCTATTCCA


GAAGTAGTGAGGAGGCTTTTTTGGAGGCCTAGGCTTTTGCAAAAAGCTCCCGGGAGCTTGTATATC


CATTTTCGGATCTGATCAGCACGTGTTGACAATTAATCATCGGCATAGTATATCGGCATAGTATAATAC


GACAAGGTGAGGAACTAAACCATGGCCAAGTTGACCAGTGCCGTTCCGGTGCTCACCGCGCGCGA


CGTCGCCGGAGCGGTCGAGTTCTGGACCGACCGGCTCGGGTTCTCCCGGGACTTCGTGGAGGACG


ACTTCGCCGGTGTGGTCCGGGACGACGTGACCCTGTTCATCAGCGCGGTCCAGGACCAGGTGGTG


CCGGACAACACCCTGGCCTGGGTGTGGGTGCGCGGCCTGGACGAGCTGTACGCCGAGTGGTCGGA


GGTCGTGTCCACGAACTTCCGGGACGCCTCCGGGCCGGCCATGACCGAGATCGGCGAGCAGCCGT


GGGGGCGGGAGTTCGCCCTGCGCGACCCGGCCGGCAACTGCGTGCACTTCGTGGCCGAGGAGCA


GGACTGACACGTGCTACGAGATTTCGATTCCACCGCCGCCTTCTATGAAAGGTTGGGCTTCGGAAT


CGTTTTCCGGGACGCCGGCTGGATGATCCTCCAGCGCGGGGATCTCATGCTGGAGTTCTTCGCCCA


CCCCAACTTGTTTATTGCAGCTTATAATGGTTACAAATAAAGCAATAGCATCACAAATTTCACAAATA


AAGCATTTTTTTCACTGCATTCTAGTTGTGGTTTGTCCAAACTCATCAATGTATCTTATCATGTCTGTA


TACCGTCGACCTCTAGCTAGAGCTTGGCGTAATCATGGTCATAGCTGTTTCCTGTGTGAAATTGTTAT


CCGCTCACAATTCCACACAACATACGAGCCGGAAGCATAAAGTGTAAAGCCTGGGGTGCCTAATGA


GTGAGCTAACTCACATTAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTCGTGCC


AGCTGCATTAATGAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCTCTTCCGCTT


CCTCGCTCACTGACTCGCTGCGCTCGGTCGTTCGGCTGCGGCGAGCGGTATCAGCTCACTCAAAGG


CGGTAATACGGTTATCCACAGAATCAGGGGATAACGCAGGAAAGAACATGTGAGCAAAAGGCCAG


CAAAAGGCCAGGAACCGTAAAAAGGCCGCGTTGCTGGCGTTTTTCCATAGGCTCCGCCCCCCTGAC


GAGCATCACAAAAATCGACGCTCAAGTCAGAGGTGGCGAAACCCGACAGGACTATAAAGATACCA


GGCGTTTCCCCCTGGAAGCTCCCTCGTGCGCTCTCCTGTTCCGACCCTGCCGCTTACCGGATACCTG


TCCGCCTTTCTCCCTTCGGGAAGCGTGGCGCTTTCTCATAGCTCACGCTGTAGGTATCTCAGTTCGG


TGTAGGTCGTTCGCTCCAAGCTGGGCTGTGTGCACGAACCCCCCGTTCAGCCCGACCGCTGCGCCT


TATCCGGTAACTATCGTCTTGAGTCCAACCCGGTAAGACACGACTTATCGCCACTGGCAGCAGCCA


CTGGTAACAGGATTAGCAGAGCGAGGTATGTAGGCGGTGCTACAGAGTTCTTGAAGTGGTGGCCTA


ACTACGGCTACACTAGAAGAACAGTATTTGGTATCTGCGCTCTGCTGAAGCCAGTTACCTTCGGAA


AAAGAGTTGGTAGCTCTTGATCCGGCAAACAAACCACCGCTGGTAGCGGTGGTTTTTTTGTTTGCA


AGCAGCAGATTACGCGCAGAAAAAAAGGATCTCAAGAAGATCCTTTGATCTTTTCTACGGGGTCTG


ACGCTCAGTGGAACGAAAACTCACGTTAAGGGATTTTGGTCATGAGATTATCAAAAAGGATCTTCA


CCTAGATCCTTTTAAATTAAAAATGAAGTTTTAAATCAATCTAAAGTATATATGAGTAAACTTGGTCT


GACAGTTACCAATGCTTAATCAGTGAGGCACCTATCTCAGCGATCTGTCTATTTCGTTCATCCATAGT


TGCCTGACTCCCCGTCGTGTAGATAACTACGATACGGGAGGGCTTACCATCTGGCCCCAGTGCTGCA


ATGATACCGCGAGACCCACGCTCACCGGCTCCAGATTTATCAGCAATAAACCAGCCAGCCGGAAGG


GCCGAGCGCAGAAGTGGTCCTGCAACTTTATCCGCCTCCATCCAGTCTATTAATTGTTGCCGGGAAG


CTAGAGTAAGTAGTTCGCCAGTTAATAGTTTGCGCAACGTTGTTGCCATTGCTACAGGCATCGTGGT


GTCACGCTCGTCGTTTGGTATGGCTTCATTCAGCTCCGGTTCCCAACGATCAAGGCGAGTTACATGA


TCCCCCATGTTGTGCAAAAAAGCGGTTAGCTCCTTCGGTCCTCCGATCGTTGTCAGAAGTAAGTTG


GCCGCAGTGTTATCACTCATGGTTATGGCAGCACTGCATAATTCTCTTACTGTCATGCCATCCGTAAG


ATGCTTTTCTGTGACTGGTGAGTACTCAACCAAGTCATTCTGAGAATAGTGTATGCGGCGACCGAGT


TGCTCTTGCCCGGCGTCAATACGGGATAATACCGCGCCACATAGCAGAACTTTAAAAGTGCTCATCA


TTGGAAAACGTTCTTCGGGGCGAAAACTCTCAAGGATCTTACCGCTGTTGAGATCCAGTTCGATGT


AACCCACTCGTGCACCCAACTGATCTTCAGCATCTTTTACTTTCACCAGCGTTTCTGGGTGAGCAAA


AACAGGAAGGCAAAATGCCGCAAAAAAGGGAATAAGGGCGACACGGAAATGTTGAATACTCATAC


TCTTCCTTTTTCAATATTATTGAAGCATTTATCAGGGTTATTGTCTCATGAGCGGATACATATTTGAAT


GTATTTAGAAAAATAAACAAATAGGGGTTCCGCGCACATTTCCCCGAAAAGTGCCACCTGAC







U6promoter





PEgRNA




hSynpromoter



N-termPE2




N-termNpu





custom character






pLenti-hSyn-C-PE2-NpuC:


(SEQ ID NO: 737)



GTCGACGGATCGGGAGATCTCCCGATCCCCTATGGTGCACTCTCAGTACAATCTGCTCTGATGCCGC



ATAGTTAAGCCAGTATCTGCTCCCTGCTTGTGTGTTGGAGGTCGCTGAGTAGTGCGCGAGCAAAATT


TAAGCTACAACAAGGCAAGGCTTGACCGACAATTGCATGAAGAATCTGCTTAGGGTTAGGCGTTTT


GCGCTGCTTCGCGATGTACGGGCCAGATATACGCGTTGACATTGATTATTGACTAGTTATTAATAGTA


ATCAATTACGGGGTCATTAGTTCATAGCCCATATATGGAGTTCCGCGTTACATAACTTACGGTAAATG


GCCCGCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGT


AACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGCCCACTTGGC


AGTACATCAAGTGTATCATATGCCAAGTACGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCC


TGGCATTATGCCCAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACATCTACGTATTAGTCAT


CGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTCACGG


GGATTTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCAACGGGAC


TTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGAG


GTCTATATAAGCAGCGCGTTTTGCCTGTACTGGGTCTCTCTGGTTAGACCAGATCTGAGCCTGGGAG


CTCTCTGGCTAACTAGGGAACCCACTGCTTAAGCCTCAATAAAGCTTGCCTTGAGTGCTTCAAGTA


GTGTGTGCCCGTCTGTTGTGTGACTCTGGTAACTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGGA


AAATCTCTAGCAGTGGCGCCCGAACAGGGACTTGAAAGCGAAAGGGAAACCAGAGGAGCTCTCT


CGACGCAGGACTCGGCTTGCTGAAGCGCGCACGGCAAGAGGCGAGGGGCGGCGACTGGTGAGTA


CGCCAAAAATTTTGACTAGCGGAGGCTAGAAGGAGAGAGATGGGTGCGAGAGCGTCAGTATTAAG


CGGGGGAGAATTAGATCGCGATGGGAAAAAATTCGGTTAAGGCCAGGGGGAAAGAAAAAATATAA


ATTAAAACATATAGTATGGGCAAGCAGGGAGCTAGAACGATTCGCAGTTAATCCTGGCCTGTTAGAA


ACATCAGAAGGCTGTAGACAAATACTGGGACAGCTACAACCATCCCTTCAGACAGGATCAGAAGA


ACTTAGATCATTATATAATACAGTAGCAACCCTCTATTGTGTGCATCAAAGGATAGAGATAAAAGACA


CCAAGGAAGCTTTAGACAAGATAGAGGAAGAGCAAAACAAAAGTAAGACCACCGCACAGCAAGC


GGCCGCTGATCTTCAGACCTGGAGGAGGAGATATGAGGGACAATTGGAGAAGTGAATTATATAAAT


ATAAAGTAGTAAAAATTGAACCATTAGGAGTAGCACCCACCAAGGCAAAGAGAAGAGTGGTGCAG


AGAGAAAAAAGAGCAGTGGGAATAGGAGCTTTGTTCCTTGGGTTCTTGGGAGCAGCAGGAAGCAC


TATGGGCGCAGCGTCAATGACGCTGACGGTACAGGCCAGACAATTATTGTCTGGTATAGTGCAGCA


GCAGAACAATTTGCTGAGGGCTATTGAGGCGCAACAGCATCTGTTGCAACTCACAGTCTGGGGCAT


CAAGCAGCTCCAGGCAAGAATCCTGGCTGTGGAAAGATACCTAAAGGATCAACAGCTCCTGGGGA


TTTGGGGTTGCTCTGGAAAACTCATTTGCACCACTGCTGTGCCTTGGAATGCTAGTTGGAGTAATAA


ATCTCTGGAACAGATTTGGAATCACACGACCTGGATGGAGTGGGACAGAGAAATTAACAATTACAC


AAGCTTAATACACTCCTTAATTGAAGAATCGCAAAACCAGCAAGAAAAGAATGAACAAGAATTATT


GGAATTAGATAAATGGGCAAGTTTGTGGAATTGGTTTAACATAACAAATTGGCTGTGGTATATAAAA


TTATTCATAATGATAGTAGGAGGCTTGGTAGGTTTAAGAATAGTTTTTGCTGTACTTTCTATAGTGAAT


AGAGTTAGGCAGGGATATTCACCATTATCGTTTCAGACCCACCTCCCAACCCCGAGGGGACCCGAC


AGGCCCGAAGGAATAGAAGAAGAAGGTGGAGAGAGAGACAGAGACAGATCCATTCGATTAGTGA


ACGGATCGGCACTGCGTGCGCCAATTCTGCAGACAAATGGCAGTATTCATCCACAATTTTAAAAGA


AAAGGGGGGATTGGGGGGTACAGTGCAGGGGAAAGAATAGTAGACATAATAGCAACAGACATACA


AACTAAAGAATTACAAAAACAAATTACAAAAATTCAAAATTTTCGGGTTTATTACAGGGACAGCAG


AGATCCAGTTTGGTTAATTAAGGTACCAGTGCAAGTGGGTTTTAGGACCAGGATGAGGCGGGGTGG



GGGTGCCTACCTGACGACCGACCCCGACCCACTGGACAAGCACCCAACCCCCATTCCCCAAATTGC




GCATCCCCTATCAGAGAGGGGGAGGGGAAACAGGATGCGGCGAGGCGCGTGCGCACTGCCAGCTT




CAGCACCGCGGACAGTGCCTTCGCCCCCGCCTGGCGGCGCGCGCCACCGCCGCCTCAGCACTGAA




GGCGCGCTGACGTCACTCGCCGGTCCCCCGCAAACTCCCCTTCCCGGCCACCTTGGTCGCGTCCGC




GCCGCCGCCGGCCCAGCCGGACCGCACCACGCGAGGCGCGAGATAGGGGGGCACGGGCGCGACC




ATCTGCGCTGCGGCGCCGGCGACTCAGCGCTGCCTCAGTCTGCGGTGGGCAGCGGAGGAGTCGTG




TCGTGCCTGAGAGCGCAG
TCGAGAATCTAGAGCGCTGCCACCATGAAACGGACAGCCGACGGAAGCG




AGTTCGAGTCACCAAAGAAGAAGCGGAAAGTC

ATCAAGATTGCTACACGGAAATACCTGGGAAAGCAGAA






CGTGTACGACATCGGCGTGGAGCGGGATCACAACTTCGCCCTGAAGAATGGCTTTATCGCCAGCAAT
TG






CTTCGACTCCGTGGAAATCTCCGGCGTGGAAGATCGGTTCAACGCCTCCCTGGGCACATACC






ACGATCTGCTGAAAATTATCAAGGACAAGGACTTCCTGGACAATGAGGAAAACGAGGACATT






CTGGAAGATATCGTGCTGACCCTGACACTGTTTGAGGACAGAGAGATGATCGAGGAACGGC






TGAAAACCTATGCCCACCTGTTCGACGACAAAGTGATGAAGCAGCTGAAGCGGCGGAGATA






CACCGGCTGGGGCAGGCTGAGCCGGAAGCTGATCAACGGCATCCGGGACAAGCAGTCCGG






CAAGACAATCCTGGATTTCCTGAAGTCCGACGGCTTCGCCAACAGAAACTTCATGCAGCTGA






TCCACGACGACAGCCTGACCTTTAAAGAGGACATCCAGAAAGCCCAGGTGTCCGGCCAGGG






CGATAGCCTGCACGAGCACATTGCCAATCTGGCCGGCAGCCCCGCCATTAAGAAGGGCATCC






TGCAGACAGTGAAGGTGGTGGACGAGCTCGTGAAAGTGATGGGCCGGCACAAGCCCGAGA






ACATCGTGATCGAAATGGCCAGAGAGAACCAGACCACCCAGAAGGGACAGAAGAACAGCCG






CGAGAGAATGAAGCGGATCGAAGAGGGCATCAAAGAGCTGGGCAGCCAGATCCTGAAAGAA






CACCCCGTGGAAAACACCCAGCTGCAGAACGAGAAGCTGTACCTGTACTACCTGCAGAATG






GGCGGGATATGTACGTGGACCAGGAACTGGACATCAACCGGCTGTCCGACTACGATGTGGA






CCATATCGTGCCTCAGAGCTTTCTGAAGGACGACTCCATCGACAACAAGGTGCTGACCAGAA






GCGACAAGAACCGGGGCAAGAGCGACAACGTGCCCTCCGAAGAGGTCGTGAAGAAGATGA






AGAACTACTGGCGGCAGCTGCTGAACGCCAAGCTGATTACCCAGAGAAAGTTCGACAATCTG






ACCAAGGCCGAGAGAGGCGGCCTGAGCGAACTGGATAAGGCCGGCTTCATCAAGAGACAGC






TGGTGGAAACCCGGCAGATCACAAAGCACGTGGCACAGATCCTGGACTCCCGGATGAACAC






TAAGTACGACGAGAATGACAAGCTGATCCGGGAAGTGAAAGTGATCACCCTGAAGTCCAAG






CTGGTGTCCGATTTCCGGAAGGATTTCCAGTTTTACAAAGTGCGCGAGATCAACAACTACCA






CCACGCCCACGACGCCTACCTGAACGCCGTCGTGGGAACCGCCCTGATCAAAAAGTACCCTA






AGCTGGAAAGCGAGTTCGTGTACGGCGACTACAAGGTGTACGACGTGCGGAAGATGATCGC






CAAGAGCGAGCAGGAAATCGGCAAGGCTACCGCCAAGTACTTCTTCTACAGCAACATCATGA






ACTTTTTCAAGACCGAGATTACCCTGGCCAACGGCGAGATCCGGAAGCGGCCTCTGATCGAG






ACAAACGGCGAAACCGGGGAGATCGTGTGGGATAAGGGCCGGGATTTTGCCACCGTGCGGA






AAGTGCTGAGCATGCCCCAAGTGAATATCGTGAAAAAGACCGAGGTGCAGACAGGCGGCTT






CAGCAAAGAGTCTATCCTGCCCAAGAGGAACAGCGATAAGCTGATCGCCAGAAAGAAGGAC






TGGGACCCTAAGAAGTACGGCGGCTTCGACAGCCCCACCGTGGCCTATTCTGTGCTGGTGG






TGGCCAAAGTGGAAAAGGGCAAGTCCAAGAAACTGAAGAGTGTGAAAGAGCTGCTGGGGAT






CACCATCATGGAAAGAAGCAGCTTCGAGAAGAATCCCATCGACTTTCTGGAAGCCAAGGGCT






ACAAAGAAGTGAAAAAGGACCTGATCATCAAGCTGCCTAAGTACTCCCTGTTCGAGCTGGAA






AACGGCCGGAAGAGAATGCTGGCCTCTGCCGGCGAACTGCAGAAGGGAAACGAACTGGCC






CTGCCCTCCAAATATGTGAACTTCCTGTACCTGGCCAGCCACTATGAGAAGCTGAAGGGCTC






CCCCGAGGATAATGAGCAGAAACAGCTGTTTGTGGAACAGCACAAGCACTACCTGGACGAG






ATCATCGAGCAGATCAGCGAGTTCTCCAAGAGAGTGATCCTGGCCGACGCTAATCTGGACAA






AGTGCTGTCCGCCTACAACAAGCACCGGGATAAGCCCATCAGAGAGCAGGCCGAGAATATCA






TCCACCTGTTTACCCTGACCAATCTGGGAGCCCCTGCCGCCTTCAAGTACTTTGACACCACC






ATCGACCGGAAGAGGTACACCAGCACCAAAGAGGTGCTGGACGCCACCCTGATCCACCAGA






GCATCACCGGCCTGTACGAGACACGGATCGACCTGTCTCAGCTGGGAGGTGACTCTGGCGG






CTCAAAAAGAACCGCCGACGGCAGCGAATTCGAGCCCAAGAAGAAGAGGAAAGTCTAA
ACG



CGTTAAGTCGACAATCAACCTCTGGATTACAAAATTTGTGAAAGATTGACTGGTATTCTTAACTATG


TTGCTCCTTTTACGCTATGTGGATACGCTGCTTTAATGCCTTTGTATCATGCTATTGCTTCCCGTATGG


CTTTCATTTTCTCCTCCTTGTATAAATCCTGGTTGCTGTCTCTTTATGAGGAGTTGTGGCCCGTTGTC


AGGCAACGTGGCGTGGTGTGCACTGTGTTTGCTGACGCAACCCCCACTGGTTGGGGCATTGCCACC


ACCTGTCAGCTCCTTTCCGGGACTTTCGCTTTCCCCCTCCCTATTGCCACGGCGGAACTCATCGCCG


CCTGCCTTGCCCGCTGCTGGACAGGGGCTCGGCTGTTGGGCACTGACAATTCCGTGGTGTTGTCGG


GGAAATCATCGTCCTTTCCTTGGCTGCTCGCCTGTGTTGCCACCTGGATTCTGCGCGGGACGTCCTT


CTGCTACGTCCCTTCGGCCCTCAATCCAGCGGACCTTCCTTCCCGCGGCCTGCTGCCGGCTCTGCG


GCCTCTTCCGCGTCTTCGCCTTCGCCCTCAGACGAGTCGGATCTCCCTTTGGGCCGCCTCCCCGCGT


CGACTTTAAGACCAATGACTTACAAGGCAGCTGTAGATCTTAGCCACTTTTTAAAAGAAAAGGGGG


GACTGGAAGGGCTAATTCACTCCCAACGAAGACAAGATCTGCTTTTTGCTTGTACTGGGTCTCTCT


GGTTAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAACTAGGGAACCCACTGCTTAAGCCTCAAT


AAAGCTTGCCTTGAGTGCTTCAAGTAGTGTGTGCCCGTCTGTTGTGTGACTCTGGTAACTAGAGAT


CCCTCAGACCCTTTTAGTCAGTGTGGAAAATCTCTAGCAGGGCCCGTTTAAACCCGCTGATCAGCC


TCGACTGTGCCTTCTAGTTGCCAGCCATCTGTTGTTTGCCCCTCCCCCGTGCCTTCCTTGACCCTGG


AAGGTGCCACTCCCACTGTCCTTTCCTAATAAAATGAGGAAATTGCATCGCATTGTCTGAGTAGGTG


TCATTCTATTCTGGGGGGTGGGGTGGGGCAGGACAGCAAGGGGGAGGATTGGGAAGACAATAGCA


GGCATGCTGGGGATGCGGTGGGCTCTATGGCTTCTGAGGCGGAAAGAACCAGCTGGGGCTCTAGG


GGGTATCCCCACGCGCCCTGTAGCGGCGCATTAAGCGCGGCGGGTGTGGTGGTTACGCGCAGCGTG


ACCGCTACACTTGCCAGCGCCCTAGCGCCCGCTCCTTTCGCTTTCTTCCCTTCCTTTCTCGCCACGT


TCGCCGGCTTTCCCCGTCAAGCTCTAAATCGGGGGCTCCCTTTAGGGTTCCGATTTAGTGCTTTACG


GCACCTCGACCCCAAAAAACTTGATTAGGGTGATGGTTCACGTAGTGGGCCATCGCCCTGATAGAC


GGTTTTTCGCCCTTTGACGTTGGAGTCCACGTTCTTTAATAGTGGACTCTTGTTCCAAACTGGAACA


ACACTCAACCCTATCTCGGTCTATTCTTTTGATTTATAAGGGATTTTGCCGATTTCGGCCTATTGGTTA


AAAAATGAGCTGATTTAACAAAAATTTAACGCGAATTAATTCTGTGGAATGTGTGTCAGTTAGGGTG


TGGAAAGTCCCCAGGCTCCCCAGCAGGCAGAAGTATGCAAAGCATGCATCTCAATTAGTCAGCAAC


CAGGTGTGGAAAGTCCCCAGGCTCCCCAGCAGGCAGAAGTATGCAAAGCATGCATCTCAATTAGTC


AGCAACCATAGTCCCGCCCCTAACTCCGCCCATCCCGCCCCTAACTCCGCCCAGTTCCGCCCATTCT


CCGCCCCATGGCTGACTAATTTTTTTTATTTATGCAGAGGCCGAGGCCGCCTCTGCCTCTGAGCTATT


CCAGAAGTAGTGAGGAGGCTTTTTTGGAGGCCTAGGCTTTTGCAAAAAGCTCCCGGGAGCTTGTAT


ATCCATTTTCGGATCTGATCAGCACGTGTTGACAATTAATCATCGGCATAGTATATCGGCATAGTATAA


TACGACAAGGTGAGGAACTAAACCATGGCCAAGTTGACCAGTGCCGTTCCGGTGCTCACCGCGCG


CGACGTCGCCGGAGCGGTCGAGTTCTGGACCGACCGGCTCGGGTTCTCCCGGGACTTCGTGGAGG


ACGACTTCGCCGGTGTGGTCCGGGACGACGTGACCCTGTTCATCAGCGCGGTCCAGGACCAGGTG


GTGCCGGACAACACCCTGGCCTGGGTGTGGGTGCGCGGCCTGGACGAGCTGTACGCCGAGTGGTC


GGAGGTCGTGTCCACGAACTTCCGGGACGCCTCCGGGCCGGCCATGACCGAGATCGGCGAGCAGC


CGTGGGGGCGGGAGTTCGCCCTGCGCGACCCGGCCGGCAACTGCGTGCACTTCGTGGCCGAGGAG


CAGGACTGACACGTGCTACGAGATTTCGATTCCACCGCCGCCTTCTATGAAAGGTTGGGCTTCGGA


ATCGTTTTCCGGGACGCCGGCTGGATGATCCTCCAGCGCGGGGATCTCATGCTGGAGTTCTTCGCCC


ACCCCAACTTGTTTATTGCAGCTTATAATGGTTACAAATAAAGCAATAGCATCACAAATTTCACAAAT


AAAGCATTTTTTTCACTGCATTCTAGTTGTGGTTTGTCCAAACTCATCAATGTATCTTATCATGTCTGT


ATACCGTCGACCTCTAGCTAGAGCTTGGCGTAATCATGGTCATAGCTGTTTCCTGTGTGAAATTGTTA


TCCGCTCACAATTCCACACAACATACGAGCCGGAAGCATAAAGTGTAAAGCCTGGGGTGCCTAATG


AGTGAGCTAACTCACATTAATTGCGTTGCGCTCACTGCCCGCTTTCCAGTCGGGAAACCTGTCGTG


CCAGCTGCATTAATGAATCGGCCAACGCGCGGGGAGAGGCGGTTTGCGTATTGGGCGCTCTTCCGC


TTCCTCGCTCACTGACTCGCTGCGCTCGGTCGTTCGGCTGCGGCGAGCGGTATCAGCTCACTCAAA


GGCGGTAATACGGTTATCCACAGAATCAGGGGATAACGCAGGAAAGAACATGTGAGCAAAAGGCC


AGCAAAAGGCCAGGAACCGTAAAAAGGCCGCGTTGCTGGCGTTTTTCCATAGGCTCCGCCCCCCT


GACGAGCATCACAAAAATCGACGCTCAAGTCAGAGGTGGCGAAACCCGACAGGACTATAAAGATA


CCAGGCGTTTCCCCCTGGAAGCTCCCTCGTGCGCTCTCCTGTTCCGACCCTGCCGCTTACCGGATAC


CTGTCCGCCTTTCTCCCTTCGGGAAGCGTGGCGCTTTCTCATAGCTCACGCTGTAGGTATCTCAGTT


CGGTGTAGGTCGTTCGCTCCAAGCTGGGCTGTGTGCACGAACCCCCCGTTCAGCCCGACCGCTGCG


CCTTATCCGGTAACTATCGTCTTGAGTCCAACCCGGTAAGACACGACTTATCGCCACTGGCAGCAGC


CACTGGTAACAGGATTAGCAGAGCGAGGTATGTAGGCGGTGCTACAGAGTTCTTGAAGTGGTGGCC


TAACTACGGCTACACTAGAAGAACAGTATTTGGTATCTGCGCTCTGCTGAAGCCAGTTACCTTCGGA


AAAAGAGTTGGTAGCTCTTGATCCGGCAAACAAACCACCGCTGGTAGCGGTGGTTTTTTTGTTTGC


AAGCAGCAGATTACGCGCAGAAAAAAAGGATCTCAAGAAGATCCTTTGATCTTTTCTACGGGGTCT


GACGCTCAGTGGAACGAAAACTCACGTTAAGGGATTTTGGTCATGAGATTATCAAAAAGGATCTTC


ACCTAGATCCTTTTAAATTAAAAATGAAGTTTTAAATCAATCTAAAGTATATATGAGTAAACTTGGTC


TGACAGTTACCAATGCTTAATCAGTGAGGCACCTATCTCAGCGATCTGTCTATTTCGTTCATCCATAG


TTGCCTGACTCCCCGTCGTGTAGATAACTACGATACGGGAGGGCTTACCATCTGGCCCCAGTGCTGC


AATGATACCGCGAGACCCACGCTCACCGGCTCCAGATTTATCAGCAATAAACCAGCCAGCCGGAAG


GGCCGAGCGCAGAAGTGGTCCTGCAACTTTATCCGCCTCCATCCAGTCTATTAATTGTTGCCGGGAA


GCTAGAGTAAGTAGTTCGCCAGTTAATAGTTTGCGCAACGTTGTTGCCATTGCTACAGGCATCGTGG


TGTCACGCTCGTCGTTTGGTATGGCTTCATTCAGCTCCGGTTCCCAACGATCAAGGCGAGTTACATG


ATCCCCCATGTTGTGCAAAAAAGCGGTTAGCTCCTTCGGTCCTCCGATCGTTGTCAGAAGTAAGTTG


GCCGCAGTGTTATCACTCATGGTTATGGCAGCACTGCATAATTCTCTTACTGTCATGCCATCCGTAAG


ATGCTTTTCTGTGACTGGTGAGTACTCAACCAAGTCATTCTGAGAATAGTGTATGCGGCGACCGAGT


TGCTCTTGCCCGGCGTCAATACGGGATAATACCGCGCCACATAGCAGAACTTTAAAAGTGCTCATCA


TTGGAAAACGTTCTTCGGGGCGAAAACTCTCAAGGATCTTACCGCTGTTGAGATCCAGTTCGATGT


AACCCACTCGTGCACCCAACTGATCTTCAGCATCTTTTACTTTCACCAGCGTTTCTGGGTGAGCAAA


AACAGGAAGGCAAAATGCCGCAAAAAAGGGAATAAGGGCGACACGGAAATGTTGAATACTCATAC


TCTTCCTTTTTCAATATTATTGAAGCATTTATCAGGGTTATTGTCTCATGAGCGGATACATATTTGAA


TGTATTTAGAAAAATAAACAAATAGGGGTTCCGCGCACATTTCCCCGAAAAGTGCCACCTGAC



hSynpromoter



C-termNpu




C-termwtCas9







pLenti-U6-DNMT1_nicking_sgRNA:


(SEQ ID NO: 738)



TAACAAATTGGCTGTGGTATATAAAATTATTCATAATGATAGTAGGAGGCTTGGTAGGTTTAAGAATA



GTTTTTGCTGTACTTTCTATAGTGAATAGAGTTAGGCAGGGATATTCACCATTATCGTTTCAGACCCA


CCTCCCAACCCCGAGGGGACCCAGAGAGGGCCTATTTCCCATGATTCCTTCATATTTGCATATACGATAC



AAGGCTGTTAGAGAGATAATTAGAATTAATTTGACTGTAAACACAAAGATATTAGTACAAAATACGTGACGTA




GAAAGTAATAATTTCTTGGGTAGTTTGCAGTTTTAAAATTATGTTTTAAAATGGACTATCATATGCTTACCGTA




ACTTGAAAGTATTTCGATTTCTTGGCTTTATATATCTTGTGGAAAGGACGAAACACCcustom character




custom character
custom character
custom character




custom character TTTTTTAAGCTTGGCGTAACTAGATCTTGAGACAAATGGCAGTATTCATCCA



CAATTTTAAAAGAAAAGGGGGGATTGGGGGGTACAGTGCAGGGGAAAGAATAGTAGACATAATAG


CAACAGACATACAAACTAAAGAATTACAAAAACAAATTACAAAAATTCAAAATTTTCGGGTTTATTA


CAGGGACAGCAGAGATCCACTTTGGCGCCGGCTCGAGGGGGCCCGGGTGCAAAGATGGATAAAGT


TTTAAACAGAGAGGAATCTTTGCAGCTAATGGACCTTCTAGGTCTTGAAAGGAGTGGGAATTGGCT


CCGGTGCCCGTCAGTGGGCAGAGCGCACATCGCCCACAGTCCCCGAGAAGTTGGGGGGAGGGGTC


GGCAATTGATCCGGTGCCTAGAGAAGGTGGCGCGGGGTAAACTGGGAAAGTGATGTCGTGTACTG


GCTCCGCCTTTTTCCCGAGGGTGGGGGAGAACCGTATATAAGTGCAGTAGTCGCCGTGAACGTTCT


TTTTCGCAACGGGTTTGCCGCCAGAACACAGGTAAGTGCCGTGTGTGGTTCCCGCGGGCCTGGCCT


CTTTACGGGTTATGGCCCTTGCGTGCCTTGAATTACTTCCACCTGGCTGCAGTACGTGATTCTTGATC


CCGAGCTTCGGGTTGGAAGTGGGTGGGAGAGTTCGAGGCCTTGCGCTTAAGGAGCCCCTTCGCCT


CGTGCTTGAGTTGAGGCCTGGCCTGGGCGCTGGGGCCGCCGCGTGCGAATCTGGTGGCACCTTCG


CGCCTGTCTCGCTGCTTTCGATAAGTCTCTAGCCATTTAAAATTTTTGATGACCTGCTGCGACGCTTT


TTTTCTGGCAAGATAGTCTTGTAAATGCGGGCCAAGATCTGCACACTGGTATTTCGGTTTTTGGGGC


CGCGGGCGGCGACGGGGCCCGTGCGTCCCAGCGCACATGTTCGGCGAGGCGGGGCCTGCGAGCG


CGGCCACCGAGAATCGGACGGGGGTAGTCTCAAGCTGGCCGGCCTGCTCTGGTGCCTGGCCTCGC


GCCGCCGTGTATCGCCCCGCCCTGGGCGGCAAGGCTGGCCCGGTCGGCACCAGTTGCGTGAGCGG


AAAGATGGCCGCTTCCCGGCCCTGCTGCAGGGAGCTCAAAATGGAGGACGCGGCGCTCGGGAGAG


CGGGCGGGTGAGTCACCCACACAAAGGAAAAGGGCCTTTCCGTCCTCAGCCGTCGCTTCATGTGA


CTCCACGGAGTACCGGGCGCCGTCCAGGCACCTCGATTAGTTCTCGAGCTTTTGGAGTACGTCGTC


TTTAGGTTGGGGGGAGGGGTTTTATGCGATGGAGTTTCCCCACACTGAGTGGGTGGAGACTGAAGT


TAGGCCAGCTTGGCACTTGATGTAATTCTCCTTGGAATTTGCCCTTTTTGAGTTTGGATCTTGGTTCA


TTCTCAAGCCTCAGACAGTGGTTCAAAGTTTTTTTCTTCCATTTCAGGTGTCGTGACGTACGGCCAC


CATGACCGAGTACAAGCCCACGGTGCGCCTCGCCACCCGCGACGACGTCCCCAGGGCCGTACGCA


CCCTCGCCGCCGCGTTCGCCGACTACCCCGCCACGCGCCACACCGTCGATCCGGACCGCCACATCG


AGCGGGTCACCGAGCTGCAAGAACTCTTCCTCACGCGCGTCGGGCTCGACATCGGCAAGGTGTGG


GTCGCGGACGACGGCGCCGCCGTGGCGGTCTGGACCACGCCGGAGAGCGTCGAAGCGGGGGCGG


TGTTCGCCGAGATCGGCCCGCGCATGGCCGAGTTGAGCGGTTCCCGGCTGGCCGCGCAGCAACAG


ATGGAAGGCCTCCTGGCGCCGCACCGGCCCAAGGAGCCCGCGTGGTTCCTGGCCACCGTCGGAGT


CTCGCCCGACCACCAGGGCAAGGGTCTGGGCAGCGCCGTCGTGCTCCCCGGAGTGGAGGCGGCCG


AGCGCGCCGGGGTGCCCGCCTTCCTGGAGACCTCCGCGCCCCGCAACCTCCCCTTCTACGAGCGG


CTCGGCTTCACCGTCACCGCCGACGTCGAGGTGCCCGAAGGACCGCGCACCTGGTGCATGACCCG


CAAGCCCGGTGCCTGAACGCGTTAAGTCGACAATCAACCTCTGGATTACAAAATTTGTGAAAGATT


GACTGGTATTCTTAACTATGTTGCTCCTTTTACGCTATGTGGATACGCTGCTTTAATGCCTTTGTATCA


TGCTATTGCTTCCCGTATGGCTTTCATTTTCTCCTCCTTGTATAAATCCTGGTTGCTGTCTCTTTATGA


GGAGTTGTGGCCCGTTGTCAGGCAACGTGGCGTGGTGTGCACTGTGTTTGCTGACGCAACCCCCA


CTGGTTGGGGCATTGCCACCACCTGTCAGCTCCTTTCCGGGACTTTCGCTTTCCCCCTCCCTATTGC


CACGGCGGAACTCATCGCCGCCTGCCTTGCCCGCTGCTGGACAGGGGCTCGGCTGTTGGGCACTG


ACAATTCCGTGGTGTTGTCGGGGAAATCATCGTCCTTTCCTTGGCTGCTCGCCTGTGTTGCCACCTG


GATTCTGCGCGGGACGTCCTTCTGCTACGTCCCTTCGGCCCTCAATCCAGCGGACCTTCCTTCCCGC


GGCCTGCTGCCGGCTCTGCGGCCTCTTCCGCGTCTTCGCCTTCGCCCTCAGACGAGTCGGATCTCC


CTTTGGGCCGCCTCCCCGCGTCGACTTTAAGACCAATGACTTACAAGGCAGCTGTAGATCTTAGCC


ACTTTTTAAAAGAAAAGGGGGGACTGGAAGGGCTAATTCACTCCCAACGAAGACAAGATCTGCTT


TTTGCTTGTACTGGGTCTCTCTGGTTAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAACTAGGGA


ACCCACTGCTTAAGCCTCAATAAAGCTTGCCTTGAGTGCTTCAAGTAGTGTGTGCCCGTCTGTTGTG


TGACTCTGGTAACTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGGAAAATCTCTAGCAGTACGTAT


AGTAGTTCATGTCATCTTATTATTCAGTATTTATAACTTGCAAAGAAATGAATATCAGAGAGTGAGAG


GAACTTGTTTATTGCAGCTTATAATGGTTACAAATAAAGCAATAGCATCACAAATTTCACAAATAAA


GCATTTTTTTCACTGCATTCTAGTTGTGGTTTGTCCAAACTCATCAATGTATCTTATCATGTCTGGCTC


TAGCTATCCCGCCCCTAACTCCGCCCATCCCGCCCCTAACTCCGCCCAGTTCCGCCCATTCTCCGCC


CCATGGCTGACTAATTTTTTTTATTTATGCAGAGGCCGAGGCCGCCTCGGCCTCTGAGCTATTCCAG


AAGTAGTGAGGAGGCTTTTTTGGAGGCCTAGGGACGTACCCAATTCGCCCTATAGTGAGTCGTATTA


CGCGCGCTCACTGGCCGTCGTTTTACAACGTCGTGACTGGGAAAACCCTGGCGTTACCCAACTTAA


TCGCCTTGCAGCACATCCCCCTTTCGCCAGCTGGCGTAATAGCGAAGAGGCCCGCACCGATCGCCC


TTCCCAACAGTTGCGCAGCCTGAATGGCGAATGGGACGCGCCCTGTAGCGGCGCATTAAGCGCGGC


GGGTGTGGTGGTTACGCGCAGCGTGACCGCTACACTTGCCAGCGCCCTAGCGCCCGCTCCTTTCGC


TTTCTTCCCTTCCTTTCTCGCCACGTTCGCCGGCTTTCCCCGTCAAGCTCTAAATCGGGGGCTCCCT


TTAGGGTTCCGATTTAGTGCTTTACGGCACCTCGACCCCAAAAAACTTGATTAGGGTGATGGTTCAC


GTAGTGGGCCATCGCCCTGATAGACGGTTTTTCGCCCTTTGACGTTGGAGTCCACGTTCTTTAATAG


TGGACTCTTGTTCCAAACTGGAACAACACTCAACCCTATCTCGGTCTATTCTTTTGATTTATAAGGG


ATTTTGCCGATTTCGGCCTATTGGTTAAAAAATGAGCTGATTTAACAAAAATTTAACGCGAATTTTAA


CAAAATATTAACGCTTACAATTTAGGTGGCACTTTTCGGGGAAATGTGCGCGGAACCCCTATTTGTT


TATTTTTCTAAATACATTCAAATATGTATCCGCTCATGAGACAATAACCCTGATAAATGCTTCAATAAT


ATTGAAAAAGGAAGAGTATGAGTATTCAACATTTCCGTGTCGCCCTTATTCCCTTTTTTGCGGCATTT


TGCCTTCCTGTTTTTGCTCACCCAGAAACGCTGGTGAAAGTAAAAGATGCTGAAGATCAGTTGGGT


GCACGAGTGGGTTACATCGAACTGGATCTCAACAGCGGTAAGATCCTTGAGAGTTTTCGCCCCGAA


GAACGTTTTCCAATGATGAGCACTTTTAAAGTTCTGCTATGTGGCGCGGTATTATCCCGTATTGACGC


CGGGCAAGAGCAACTCGGTCGCCGCATACACTATTCTCAGAATGACTTGGTTGAGTACTCACCAGT


CACAGAAAAGCATCTTACGGATGGCATGACAGTAAGAGAATTATGCAGTGCTGCCATAACCATGAG


TGATAACACTGCGGCCAACTTACTTCTGACAACGATCGGAGGACCGAAGGAGCTAACCGCTTTTTT


GCACAACATGGGGGATCATGTAACTCGCCTTGATCGTTGGGAACCGGAGCTGAATGAAGCCATACC


AAACGACGAGCGTGACACCACGATGCCTGTAGCAATGGCAACAACGTTGCGCAAACTATTAACTG


GCGAACTACTTACTCTAGCTTCCCGGCAACAATTAATAGACTGGATGGAGGCGGATAAAGTTGCAG


GACCACTTCTGCGCTCGGCCCTTCCGGCTGGCTGGTTTATTGCTGATAAATCTGGAGCCGGTGAGCG


TGGGTCTCGCGGTATCATTGCAGCACTGGGGCCAGATGGTAAGCCCTCCCGTATCGTAGTTATCTAC


ACGACGGGGAGTCAGGCAACTATGGATGAACGAAATAGACAGATCGCTGAGATAGGTGCCTCACT


GATTAAGCATTGGTAACTGTCAGACCAAGTTTACTCATATATACTTTAGATTGATTTAAAACTTCATTT


TTAATTTAAAAGGATCTAGGTGAAGATCCTTTTTGATAATCTCATGACCAAAATCCCTTAACGTGAGT


TTTCGTTCCACTGAGCGTCAGACCCCGTAGAAAAGATCAAAGGATCTTCTTGAGATCCTTTTTTTCT


GCGCGTAATCTGCTGCTTGCAAACAAAAAAACCACCGCTACCAGCGGTGGTTTGTTTGCCGGATCA


AGAGCTACCAACTCTTTTTCCGAAGGTAACTGGCTTCAGCAGAGCGCAGATACCAAATACTGTTCT


TCTAGTGTAGCCGTAGTTAGGCCACCACTTCAAGAACTCTGTAGCACCGCCTACATACCTCGCTCTG


CTAATCCTGTTACCAGTGGCTGCTGCCAGTGGCGATAAGTCGTGTCTTACCGGGTTGGACTCAAGA


CGATAGTTACCGGATAAGGCGCAGCGGTCGGGCTGAACGGGGGGTTCGTGCACACAGCCCAGCTT


GGAGCGAACGACCTACACCGAACTGAGATACCTACAGCGTGAGCTATGAGAAAGCGCCACGCTTC


CCGAAGGGAGAAAGGCGGACAGGTATCCGGTAAGCGGCAGGGTCGGAACAGGAGAGCGCACGAG


GGAGCTTCCAGGGGGAAACGCCTGGTATCTTTATAGTCCTGTCGGGTTTCGCCACCTCTGACTTGA


GCGTCGATTTTTGTGATGCTCGTCAGGGGGGCGGAGCCTATGGAAAAACGCCAGCAACGCGGCCTT


TTTACGGTTCCTGGCCTTTTGCTGGCCTTTTGCTCACATGTTCTTTCCTGCGTTATCCCCTGATTCTGT


GGATAACCGTATTACCGCCTTTGAGTGAGCTGATACCGCTCGCCGCAGCCGAACGACCGAGCGCAG


CGAGTCAGTGAGCGAGGAAGCGGAAGAGCGCCCAATACGCAAACCGCCTCTCCCCGCGCGTTGGC


CGATTCATTAATGCAGCTGGCACGACAGGTTTCCCGACTGGAAAGCGGGCAGTGAGCGCAACGCA


ATTAATGTGAGTTAGCTCACTCATTAGGCACCCCAGGCTTTACACTTTATGCTTCCGGCTCGTATGTT


GTGTGGAATTGTGAGCGGATAACAATTTCACACAGGAAACAGCTATGACCATGATTACGCCAAGCG


CGCAATTAACCCTCACTAAAGGGAACAAAAGCTGGAGCTGCAAGCTTAATGTAGTCTTATGCAATA


CTCTTGTAGTCTTGCAACATGGTAACGATGAGTTAGCAACATGCCTTACAAGGAGAGAAAAAGCAC


CGTGCATGCCGATTGGTGGAAGTAAGGTGGTACGATCGTGCCTTATTAGGAAGGCAACAGACGGGT


CTGACATGGATTGGACGAACCACTGAATTGCCGCATTGCAGAGATATTGTATTTAAGTGCCTAGCTC


GATACATAAACGGGTCTCTCTGGTTAGACCAGATCTGAGCCTGGGAGCTCTCTGGCTAACTAGGGA


ACCCACTGCTTAAGCCTCAATAAAGCTTGCCTTGAGTGCTTCAAGTAGTGTGTGCCCGTCTGTTGTG


TGACTCTGGTAACTAGAGATCCCTCAGACCCTTTTAGTCAGTGTGGAAAATCTCTAGCAGTGGCGC


CCGAACAGGGACTTGAAAGCGAAAGGGAAACCAGAGGAGCTCTCTCGACGCAGGACTCGGCTTG


CTGAAGCGCGCACGGCAAGAGGCGAGGGGCGGCGACTGGTGAGTACGCCAAAAATTTTGACTAGC


GGAGGCTAGAAGGAGAGAGATGGGTGCGAGAGCGTCAGTATTAAGCGGGGGAGAATTAGATCGCG


ATGGGAAAAAATTCGGTTAAGGCCAGGGGGAAAGAAAAAATATAAATTAAAACATATAGTATGGGC


AAGCAGGGAGCTAGAACGATTCGCAGTTAATCCTGGCCTGTTAGAAACATCAGAAGGCTGTAGACA


AATACTGGGACAGCTACAACCATCCCTTCAGACAGGATCAGAAGAACTTAGATCATTATATAATACA


GTAGCAACCCTCTATTGTGTGCATCAAAGGATAGAGATAAAAGACACCAAGGAAGCTTTAGACAAG


ATAGAGGAAGAGCAAAACAAAAGTAAGACCACCGCACAGCAAGCGGCCGCTGATCTTCAGACCTG


GAGGAGGAGATATGAGGGACAATTGGAGAAGTGAATTATATAAATATAAAGTAGTAAAAATTGAAC


CATTAGGAGTAGCACCCACCAAGGCAAAGAGAAGAGTGGTGCAGAGAGAAAAAAGAGCAGTGGG


AATAGGAGCTTTGTTCCTTGGGTTCTTGGGAGCAGCAGGAAGCACTATGGGCGCAGCGTCAATGAC


GCTGACGGTACAGGCCAGACAATTATTGTCTGGTATAGTGCAGCAGCAGAACAATTTGCTGAGGGC


TATTGAGGCGCAACAGCATCTGTTGCAACTCACAGTCTGGGGCATCAAGCAGCTCCAGGCAAGAAT


CCTGGCTGTGGAAAGATACCTAAAGGATCAACAGCTCCTGGGGATTTGGGGTTGCTCTGGAAAACT


CATTTGCACCACTGCTGTGCCTTGGAATGCTAGTTGGAGTAATAAATCTCTGGAACAGATTTGGAAT


CACACGACCTGGATGGAGTGGGACAGAGAAATTAACAATTACACAAGCTTAATACACTCCTTAATT


GAAGAATCGCAAAACCAGCAAGAAAAGAATGAACAAGAATTATTGGAATTAGATAAATGGGCAA


GTTTGTGGAATTGGTTTAACA


U6promoter



custom character







Amino acid sequences of Maloney murine leukemia virus reverse transcriptase (M-MLV RT) variants used herein.










PE1 M-MLV RT:



(SEQ ID NO: 739)



TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPM



SQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNP


YNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPT


LFDEALHRDLADFRIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQIC


QKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPGFAEMAAPLYP


LTKTGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRP


VAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNA


RMTHYQALLLDTDRVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT


WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTD


SRYAFATAHIHGEIYRRRGLLTSEGKEIKNKDEILALLKALFLPKRLSIIHCPGHQKGHSAEARGNR


MADQAARKAAITETPDTSTLLIENSSP





M3 M-MLV RT (D200N, T330P, L603W)(see Baranauskas et al.182):


(SEQ ID NO: 740)



TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPM



SQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNP


YNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPT


LFNEALHRDLADFRIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQIC


QKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPGFAEMAAPLYP


LTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRP


VAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNA


RMTHYQALLLDTDRVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT


WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTD


SRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRLSIIHCPGHQKGHSAEARGNR


MADQAARKAAITETPDTSTLLIENSSP





PE2 M-MLVRT (D200N, T306K, W313F, T330P, L603W):


(SEQ ID NO: 741)



TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPM



SQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPNP


YNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPT


LFNEALHRDLADFRIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQIC


QKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPGFAEMAAPLYP


LTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRP


VAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNA


RMTHYQALLLDTDRVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT


WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTD


SRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRLSIIHCPGHQKGHSAEARGNR


MADQAARKAAITETPDTSTLLIENSSP





M3-deadRT M-MLV RT:


(SEQ ID NO: 742)



TLNIEDEYRLHETSKEPDVSLGSTWLSDFPQAWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPM



SQEARLGIKPHIQRLLDQGILVPCQSPWNTPLLPVKLPGTNDYSPVQDLREVNKRVEDIHPTVPNP


YNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISGQLTWTRLPQGFKNSPT


LFNEALHRDLADFRIQHPDLILLQYVDDLLLAATSELDCQQGTRALLQTLGNLGYRASAKKAQIC


QKQVKYLGYLLKEGQRWLTEARKETVMGQPTPKTPRQLREFLGTAGFCRLWIPGFAEMAAPLYP


LTKPGTLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVLTQKLGPWRRP


VAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQPLVILAPHAVEALVKQPPDRWLSNA


RMTHYQALLLDTDRVQFGPVVALNPATLLPLPEEGLQHNCLDILAEAHGTRPDLTDQPLPDADHT


WYTDGSSLLQEGQRKAGAAVTTETEVIWAKALPAGTSAQRAELIALTQALKMAEGKKLNVYTD


SRYAFATAHIHGEIYRRRGWLTSEGKEIKNKDEILALLKALFLPKRLSIIHCPGHQKGHSAEARGNR


MADQAARKAAITETPDTSTLLIENSSP






REFERENCES FOR EXAMPLE 2

Each of the following references are cited in Example 12, each of which are incorporated herein by reference.

  • 1. Landrum, M. J. et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 44, D862—D868 (2016).
  • 2. Jinek, M. et al. A Programmable Dual-RNA-Guided DNA Endonuclease in Adaptive Bacterial Immunity. Science 337, 816-821 (2012).
  • 3. Cong, L. et al. Multiplex Genome Engineering Using CRISPR/Cas Systems. Science 339, 819-823 (2013).
  • 4. Mali, P. et al. RNA-Guided Human Genome Engineering via Cas9. Science 339, 823-826 (2013).
  • 5. Yang, H. et al. One-Step Generation of Mice Carrying Reporter and Conditional Alleles by CRISPR/Cas-Mediated Genome Engineering. Cell 154, 1370-1379 (2013).
  • 6. Kim, S., Kim, D., Cho, S. W., Kim, J. & Kim, J.-S. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res. 24, 1012-1019 (2014).
  • 7. Orlando, S. J. et al. Zinc-finger nuclease-driven targeted integration into mammalian genomes using donors with limited chromosomal homology. Nucleic Acids Res. 38, e152-e152 (2010).
  • 8. Tsai, S. Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPRCas nucleases. Nat. Biotechnol. 33, 187-197 (2015).
  • 9. Suzuki, K. et al. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature 540, 144-149 (2016).
  • 10. Kosicki, M., Tomberg, K. & Bradley, A. Repair of double-strand breaks induced by CRISPR-Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 36, 765-771 (2018).
  • 11. Haapaniemi, E., Botla, S., Persson, J., Schmierer, B. & Taipale, J. CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response. Nat. Med. 24, 927-930 (2018).
  • 12. Ihry, R. J. et al. p53 inhibits CRISPR-Cas9 engineering in human pluripotent stem cells. Nat. Med. 24, 939-946 (2018).
  • 13. Rouet, P., Smih, F. & Jasin, M. Expression of a site-specific endonuclease stimulates homologous recombination in mammalian cells. Proc. Natl. Acad. Sci. 91, 6064-6068 (1994).
  • 14. Chapman, J. R., Taylor, M. R. G. & Boulton, S. J. Playing the end game: DNA double-strand break repair pathway choice. Mol. Cell 47, 497-510 (2012).
  • 15. Cox, D. B. T., Platt, R. J. & Zhang, F. Therapeutic genome editing: prospects and challenges. Nat. Med. 21, 121-131 (2015).
  • 16. Paquet, D. et al. Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature 533, 125-129 (2016).
  • 17. Chu, V. T. et al. Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat. Biotechnol. 33, 543-548 (2015).
  • 18. Maruyama, T. et al. Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat. Biotechnol. 33, 538-542 (2015).
  • 19. Rees, H. A., Yeh, W.-H. & Liu, D. R. Development of hRad51-Cas9 nickase fusions that mediate HDR without double-stranded breaks. Nat. Commun. 10, 1-12 (2019).
  • 20. Shen, M. W. et al. Predictable and precise template-free CRISPR editing of pathogenic variants. Nature 563, 646-651 (2018).
  • 21. Rees, H. A. & Liu, D. R. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 19, 770 (2018).
  • 22. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420-424 (2016).
  • 23. Gaudelli, N. M. et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551, 464-471 (2017).
  • 24. Gao, X. et al. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature 553, 217-221 (2018).
  • 25. Ingram, V. M. A specific chemical difference between the globins of normal human and sickle-cell anaemia haemoglobin. Nature 178, 792-794 (1956).
  • 26. Myerowitz, R. & Costigan, F. C. The major defect in Ashkenazi Jews with Tay-Sachs disease is an insertion in the gene for the alpha-chain of beta-hexosaminidase. J. Biol. Chem. 263, 18587-18589 (1988).
  • 27. Zielenski, J. Genotype and Phenotype in Cystic Fibrosis. Respiration 67, 117-133 (2000).
  • 28. Mead, S. et al. A Novel Protective Prion Protein Variant that Colocalizes with Kuru Exposure. N. Engl. J. Med. 361, 2056-2065 (2009).
  • 29. Marraffini, L. A. & Sontheimer, E. J. CRISPR interference limits horizontal gene transfer in staphylococci by targeting DNA. Science 322, 1843-1845 (2008).
  • 30. Barrangou, R. et al. CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709-1712 (2007).
  • 31. Jiang, F. & Doudna, J. A. CRISPR-Cas9 Structures and Mechanisms. Annu. Rev. Biophys. 46, 505-529 (2017).
  • 32. Hille, F. et al. The Biology of CRISPR-Cas: Backward and Forward. Cell 172, 1239-1259 (2018).
  • 33. Luan, D. D., Korman, M. H., Jakubczak, J. L. & Eickbush, T. H. Reverse transcription of R2Bm RNA is primed by a nick at the chromosomal target site: a mechanism for non-LTR retrotransposition. Cell 72, 595-605 (1993).
  • 34. Liu, Y., Kao, H.-I. & Bambara, R. A. Flap endonuclease 1: a central component of DNA metabolism. Annu. Rev. Biochem. 73, 589-615 (2004).
  • 35. Richardson, C. D., Ray, G. J., DeWitt, M. A., Curie, G. L. & Corn, J. E. Enhancing homology directed genome editing by catalytically active and inactive CRISPR-Cas9 using asymmetric donor DNA. Nat. Biotechnol. 34, 339-344 (2016).
  • 36. Qi, L. S. et al. Repurposing CRISPR as an RNA-Guided Platform for Sequence-Specific Control of Gene Expression. Cell 152, 1173-1183 (2013).
  • 37. Shechner, D. M., Hacisuleyman, E., Younger, S. T. & Rinn, J. L. Multiplexable, locus-specific targeting of long RNAs with CRISPR-Display. Nat. Methods 12, 664-670 (2015).
  • 38. Tang, W., Hu, J. H. & Liu, D. R. Aptazyme-embedded guide RNAs enable ligand-responsive genome editing and transcriptional activation. Nat. Commun. 8, 15939 (2017).
  • 39. Jinek, M. et al. Structures of Cas9 Endonucleases Reveal RNA-Mediated Conformational Activation. Science 343, 1247997 (2014).
  • 40. Nishimasu, H. et al. Crystal Structure of Cas9 in Complex with Guide RNA and Target DNA. Cell 156, 935-949 (2014).
  • 41. Jiang, F., Zhou, K., Ma, L., Gressel, S. & Doudna, J. A. A Cas9-guide RNA complex preorganized for target DNA recognition. Science 348, 1477-1481 (2015).
  • 42. Baranauskas, A. et al. Generation and characterization of new highly thermostable and processive M-MuLV reverse transcriptase variants. Protein Eng. Des. Sel. 25, 657-668 (2012).
  • 43. Gerard, G. F. et al. The role of template-primer in protection of reverse transcriptase from thermal inactivation. Nucleic Acids Res. 30, 3118-3129 (2002).
  • 44. Arezi, B. & Hogrefe, H. Novel mutations in Moloney Murine Leukemia Virus reverse transcriptase increase thermostability through tighter binding to template-primer. Nucleic Acids Res. 37, 473-481 (2009).
  • 45. Kotewicz, M. L., Sampson, C. M., D'Alessio, J. M. & Gerard, G. F. Isolation of cloned Moloney murine leukemia virus reverse transcriptase lacking ribonuclease H activity. Nucleic Acids Res. 16, 265-277 (1988).
  • 46. Thuronyi, B. W. et al. Continuous evolution of base editors with expanded target compatibility and improved activity. Nat. Biotechnol. (2019). doi:10.1038/s41587-019-0193-0
  • 47. Kim, Y. B. et al. Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat. Biotechnol. 35, 371-376 (2017).
  • 48. Koblan, L. W. et al. Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat. Biotechnol. (2018). doi:10.1038/nbt.4172
  • 49. Kleinstiver, B. P. et al. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off target effects. Nature 529, 490-495 (2016).
  • 50. Zuo, E. et al. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science 364, 289-292 (2019).
  • 51. Jin, S. et al. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science 364, 292-295 (2019).
  • 52. Kim, D., Kim, D., Lee, G., Cho, S.-I. & Kim, J.-S. Genome-wide target specificity of CRISPR RNA guided adenine base editors. Nat. Biotechnol. 37, 430-435 (2019).
  • 53. Grunewald, J. et al. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 569, 433-437 (2019).
  • 54. Zhou, C. et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature 571, 275-278 (2019).
  • 55. Rees, H. A., Wilson, C., Doman, J. L. & Liu, D. R. Analysis and minimization of cellular RNA editing by DNA adenine base editors. Sci. Adv. 5, eaax5717 (2019).
  • 56. Ostertag, E. M. & Kazazian Jr, H. H. Biology of Mammalian L1 Retrotransposons. Annu. Rev. Genet. 35, 501-538 (2001).
  • 57. Griffiths, D. J. Endogenous retroviruses in the human genome sequence. Genome Biol. 2, REVIEWS1017 (2001).
  • 58. Berkhout, B., Jebbink, M. & Zsiros, J. Identification of an Active Reverse Transcriptase Enzyme Encoded by a Human Endogenous HERV-K Retrovirus. J. Virol. 73, 2365-2375 (1999).
  • 59. Halvas, E. K., Svarovskaia, E. S. & Pathak, V. K. Role of Murine Leukemia Virus Reverse Transcriptase Deoxyribonucleoside Triphosphate-Binding Site in Retroviral Replication and In Vivo Fidelity. J. Virol. 74, 10349-10358 (2000).
  • 60. Dever, D. P. et al. CRISPR/Cas9 Beta-globin Gene Targeting in Human Hematopoietic Stem Cells. Nature 539, 384-389 (2016).
  • 61. Park, S. H. et al. Highly efficient editing of the β-globin gene in patient-derived hematopoietic stem and progenitor cells to treat sickle cell disease. Nucleic Acids Res. doi:10.1093/nar/gkz475
  • 62. Collinge, J. Prion diseases of humans and animals their causes and molecular basis. Annu. Rev. Neurosci. 24, 519-550 (2001).
  • 63. Asante, E. A. et al. A naturally occurring variant of the human prion protein completely prevents prion disease. Nature 522, 478-481 (2015).
  • 64. Zettler, J., Schutz, V. & Mootz, H. D. The naturally split Npu DnaE intein exhibits an extraordinarily high rate in the protein trans-splicing reaction. FEBS Lett. 583, 909-914 (2009).
  • 65. Kugler, S., Kilic, E. & Bahr, M. Human synapsin 1 gene promoter confers highly neuron-specific long-term transgene expression from an adenoviral vector in the adult rat brain depending on the transduced area. Gene Ther. 10, 337-347 (2003).
  • 66. de Felipe, P., Hughes, L. E., Ryan, M. D. & Brown, J. D. Co-translational, intraribosomal cleavageof polypeptides by the foot-and-mouth disease virus 2A peptide. J. Biol. Chem. 278, 11441-11448 (2003).


    1. Establish Guide RNA-Templated Reverse Transcription of Mutagenic DNA Strands.


Background and Rationale


In the proposed genome editing strategy, the Cas9-nicked non-target DNA strand (PAM-containing strand that forms the R-loop) acts as the primer for DNA synthesis. It is hypothesized that this is possible based on several pieces of biochemical and structural data. Nuclease protection experiments32, crystallographic studies33, and base editing windows4,24 have demonstrated a large degree of flexibility and disorder for the non-target strand nucleotides −20 through −10 within the so-called R-loop of the Cas9-bound complex (numbering indicates distance 5′ from first PAM nucleotide). Moreover, the PAM-distal portion of the cleaved non-target strand can be displaced from tightly bound ternary complexes when complementary ssDNA is added in trans20. These studies support that the non-target strand is highly flexible, is accessible to enzymes, and that after nicking, the 3′ terminus of the PAM-distal fragment is released prior to Cas9 dissociation. Furthermore, it is hypothesized that gRNAs can be extended to template DNA synthesis. Prior studies have shown that gRNAs for SpCas9, SaCas9, and LbCas12a (formerly Cpf1) tolerate gRNA extensions with RNA aptamers34, ligand-inducible self-cleaving ribozymes35, and long non-coding RNAs36. This literature establishes precedent for two major features that will be exploited. In assessing this strategy, several CRISPR-Cas systems will be evaluated in conjunction with 5′ and 3′ extended gRNA designs using a combination of in vitro and cellular assays (FIGS. 2A-2C).


Designs for engineered gRNAs for TRT editing are shown in FIGS. 3A-3B. DNA synthesis proceeds 5′ to 3′, and thus copies the RNA template in the 3′ to 5′ direction. The design for the 5′ extension contains a linker region, a primer binding site where the nicked DNA strand anneals, and a template for DNA synthesis by reverse transcription. The 3′ extended gRNA contains a primer binding site and a reverse transcription template. In some cases, the 3′ RNA hairpin of the gRNA core is modified to match the DNA target sequence, as in vitro experiments showed that reverse transcription extends ˜3 nucleotides into the gRNA core for the 3′ extended gRNA constructs (modification of the hairpin sequence appears well tolerated so long as compensatory changes are made that maintain the hairpin RNA structure). DNA synthesis proceed 5′ to 3′, with nucleotides added to the 3′ OH of the growing DNA strand.


Preliminary Results


Cas9 nicked DNA primes reverse transcription of gRNA templates. To evaluate the accessibility of the nicked non-target DNA strand, in vitro biochemical assays were performed using the Cas9 nuclease from S. pyogenes (SpCas9) and Cy5 fluorescently labeled duplex DNA substrates (51 base pairs). First, a series of gRNAs containing 5′ extensions with varying synthesis template lengths were prepared by in vitro transcription (overall design shown in FIG. 2B). Electrophoretic mobility shift assays (EMSA) with nuclease dead Cas9 (dCas9) established that 5′ extended gRNAs maintain target binding affinity (data not shown). Next, TPRT activity was tested on pre-nicked Cy5-labeled duplex DNA substrates using dCas9, 5′-extended gRNAs, and Molony-Murine Leukemia Virus (M-MLV) reverse transcriptase (Superscript III). After 1 hour of incubation at 37° C., products were evaluated by denaturing polyacrylamide gel electrophoresis (PAGE) and imaged using Cy5 fluorescence (FIG. 4A). Each 5′-extended gRNA variant led to significant product formation, with the observed DNA product sizes being consistent with the length of the extension template (FIG. 4B). Importantly, in the absence of dCas9, pre-nicked substrates were extended to the full 51-bp length of the DNA substrate, strongly suggesting that the complementary DNA strand, and not the gRNA, was used as the template for DNA synthesis when dCas9 was not present (FIG. 4C). Of note, the system was designed such that the newly synthesized DNA strand mirrors the product that would be required for target site editing (a homologous strand with a single nucleotide change). This result establishes that Cas9:gRNA binding exposes the nicked non-target strand's 3′ end, and that the non-target strand is accessible to reverse transcription.


Next, non-nicked dsDNA substrates were evaluated using the Cas9(H840A) mutant, which nicks the non-target DNA strand. First, to test Cas9(H840A) nickase activity with 5′-extended gRNAs, in vitro cleavage assays were performed as previously described'. Although nicking was impaired by comparison to the standard gRNA, appreciable cleavage products were formed (FIG. 4D). Importantly, RT products were also observed when TPRT reactions were carried out with 5′-extended gRNAs and Cas9(H840A), albeit at lower yields that are likely explained by the decreased nicking activity (FIG. 4D). This result establishes that 5′-extended gRNA:Cas9(H840A) complexes can nick DNA and template reverse transcription.


Finally, 3′ gRNA extensions were evaluated for Cas9(H840A) nicking and TPRT. By comparison to 5′-extended gRNAs, DNA cleavage by 3′-extended gRNAs was not impaired to any detectable extent compared to the standard gRNA. Significantly, 3′-extended gRNA templates also supported efficient reverse transcription with both pre-nicked and intact duplex DNA substrates when M-MLV RT was supplied in trans (FIG. 4E). Surprisingly, only a single product was observed for 3′-extended templates, indicating that reverse transcription terminates at a specific location along the gRNA scaffold. Homopolymer tailing of the product with terminal transferase followed by Klenow extension and Sanger sequencing revealed that the full gRNA synthesis template was copied in addition to the terminal 3 nucleotides of the gRNA core. In the future, the flap terminus will be reprogrammed by modifying the terminal gRNA sequence38,39. This result demonstrates that 3′-extended gRNAs can serve as efficient nuclease targeting guides and can template reverse transcription.


Cas9-TPRT uses nicked DNA and gRNA in cis. Dual color experiments were used to determine if the RT reaction preferentially occurs with the gRNA in cis (bound in the same complex) (see FIG. 8). Two separate experiments were conducted for 5′-extended and 3′-extended gRNAs. For a given experiment, ternary complexes of dCas9, gRNA, and DNA substrate were formed in separate tubes. In one tube, the gRNA encodes a long RT product and the DNA substrate is labeled with Cy3 (red); in the other, the gRNA encodes a short RT product and the DNA substrate is labeled with Cy5 (blue). After short incubations, the complexes were mixed and then treated with RT enzyme and dNTPs. Products were separated by urea-denaturing PAGE and visualized by fluorescence in the Cy3 and Cy5 channels. Reaction products were found to preferentially form using the gRNA template that was pre-complexed with the DNA substrate, indicating that the RT reaction likely can occur in cis. This results supports that a single Cas9:gRNA complex can target a DNA site and template reverse transcription of a mutagenic DNA strand.


Testing TPRT with Other Cas Systems


Similar experiments to those presented in the previous sections will be carried out using other Cas systems, including Cas9 from S. aureus and Cas12a from L. bacterium (see FIGS. 2A-2C). If TRPT can also be demonstrated for these Cas variants, the potential editing scope and likelihood of overall success in cells would increase.


Testing TPRT with RT-Cas9 Fusion Proteins


A series of commercially available or purifiable RT enzymes will first be evaluated in trans for TPRT activity. In addition to the already tested RT from M-MLV, the RT from Avian Myeloblastosis Virus (AMV), the Geobacillus stearothermophilus Group II Intron (GsI-IIC)41,42, and the Eubacterium rectale group II intron (Euse.I2)43,44 will be evaluated. Significantly, the latter two RTs perform TPRT in their natural biological contexts. Where relevant, RNAse inactivating mutations and other potentially beneficial RT enzyme modifications will be tested. Once functional RTs are identified when supplied in trans, each will be evaluated as a fusion protein to Cas9 variants. Both N-terminus and C-terminus fusion orientations will be tested, along with various linker lengths and architectures. Kinetic time course experiments will be used to determine whether TPRT can occur using the RT enzyme in cis. If an RT-Cas9 fusion architecture can be constructed that allows for efficient TPRT chemistry, this will greatly increase the likelihood of functional editing in the context of a cell.


Cas9 Targeting with Engineered gRNAs in Cells


Candidate engineered gRNAs developed in the previous sub-aims will be evaluated in human cell culture experiments (HEK293) to confirm Cas9 targeting efficiency. Using established indel formation assays employing wild type SpCas945, engineered gRNAs will be compared side-by-side with standard gRNAs across 5 or more sites in the human genome. Genome editing efficiency will be characterized by amplicon sequencing in multiplex using the Illumina MiSeq platform housed in the laboratory. It is anticipated that results from this and the preceding sections will generate insights that inform subsequent iterations of the design-build-test cycle, where gRNAs can be optimized for both templating reverse transcription and efficient Cas9 targeting in cells.


Results of in vitro validations are shown in FIGS. 5-7. In vitro experiments demonstrated that the nicked non-target DNA strand is flexible and available for priming DNA synthesis, and that the gRNA extension can serve as a template for reverse transcription (see FIG. 5). This set of experiments used 5′-extended gRNAs (designed as shown in FIGS. 3A-3B) with varying length synthesis templates (listed to the left). Fluorescently labeled (Cy5) DNA targets were used as substrates, and were pre-nicked in this set of experiments. The Cas9 used in these experiments is catalytically dead Cas9 (dCas9), so cannot cut DNA but can still bind efficiently. Superscript III, a commercial RT derived from the Moloney-Murine Leukemia Virus (M-MLV), was supplied in trans. First, dCas9:gRNA complexes were formed from purified components. Then, the fluorescently labeled DNA substrate was added along with dNTPs and the RT enzyme. After 1 hour of incubation at 37 C, the reaction products were analyzed by denaturing urea-polyacrylamide gel electrophoresis (PAGE). The gel image shows extension of the original DNA strand to lengths that are consistent with the length of the reverse transcription template. Of note, reactions carried out in the absence of dCas9 produced DNA products of length 51 nucleotides, regardless of the gRNA used. This product corresponds to use of the complementary DNA strand as the template for DNA synthesis and not the RNA (data not shown). Thus, Cas9 binding is required for directing DNA synthesis to the RNA template. This set of in vitro experiments closely parallels those shown in FIG. 5, except that the DNA substrate is not pre-nicked, and a Cas9 nickase (SpyCas9 H840A mutant) is used. As shown in the gel, the nickase efficiently cleaves the DNA strand when the standard gRNA is used (gRNA_0, lane 3). Multiple cleavage products are observed, consistent with prior biochemical studies of SpyCas9. The 5′ extension impairs nicking activity (lanes 4-8), but some RT product is still observed. FIG. 7 shows that 3′ extensions support DNA synthesis and do not significantly effect Cas9 nickase activity. Pre-nicked substrates (black arrow) are near-quantitatively converted to RT products when either dCas9 or Cas9 nickase is used (lanes 4 and 5). Greater than 50% conversion to the RT product (red arrow) is observed with full substrates (lane 3). To determine the length and sequence of the RT product, the product band was excised from the gel, extracted, and sequenced. This revealed that RT extended 3 nucleotides into the gRNA core's 3′ terminal hairpin. Subsequent experiments (not shown) demonstrated that these three nucleotides could be changed to match target DNA sequences, so long as complementary changes were made that maintain the hairpin RNA structure.


Potential Difficulties and Alternatives


(1) RT does not function as a fusion: molecular crowding and/or unfavorable geometries could encumber polymerase extension by Cas9-fused RT enzymes. First, linker optimization can be tested. Circularly permutated variants of Cas9, which could re-orient the spatial relationship between the DNA primer, gRNA, and RT enzyme, will be evaluated. Non-covalent RT recruitment strategies as detailed in Aim 2 can be tested. (2) Decreased Cas targeting efficiency by extended gRNA variants: this is most likely to be an issue for 5′-extended gRNAs. Based on structural data', Cas9 mutants can be designed and screened to identify variants with greater tolerance to gRNA extension. In addition, gRNA libraries could be screened in cells for linkers that improve targeting activity.


Significance


These preliminary results establish that Cas9 nickases and extended gRNAs can initiate target-primed reverse transcription on bound DNA targets using a reverse transcriptase supplied in trans. Importantly, Cas9 binding was found to be critically important for product formation. Though perhaps not an absolute requirement for genome editing in cells, further development of the system that incorporates RT enzyme function in cis would significantly increase the likelihood of success in cell-based applications. Achievement of the remaining aspects of this Aim would provide a molecular foundation for carrying out precision genome editing in the context of the human genome.


2. Establish Prime Editing in Human Cells.


Background and Rationale


In the proposed strategy, an engineered RT-Cas9:gRNA complex will introduce mutagenic 3′ DNA flaps at genomic target sites. It is hypothesized that mutagenic 3′ flaps containing a single mismatch will be incorporated by the DNA repair machinery through energetically accessible equilibration with adjacent 5′ flaps, which would be preferentially removed (FIGS. 1C-1D). The DNA replication and repair machineries encounter 5′ ssDNA flaps when processing Okazaki fragments46 and during long-patch base excision repair (LP-BER)47. 5′ flaps are the preferred substrates for the widely expressed flap endonuclease FEN1, which is recruited to DNA repair sites by the homotrimeric sliding clamp complex PCNA48. PCNA also serves as a scaffold for simultaneous recruitment of other repair factors including the DNA ligase Lig149. Acting as a ‘toolbelt’, PCNA accelerates serial flap cleavage and ligation, which is essential to processing the millions of Okazaki fragments generated during every cell division50,51. Based on resemblance to these natural DNA intermediates, it is hypothesized that mutagenic strands would be incorporated through equilibration with 5′ flaps, followed by coordinated 5′ flap excision and ligation. Mismatch repair (MMR) should then occur on either strand with equal probability, leading to editing or reversion (FIGS. 1C-1D). Alternatively, DNA replication could occur first and lead directly to the incorporation of the edit in the newly synthesized daughter strand. While the highest expected yield from this process is 50%, multiple substrate editing attempts could drive the reaction toward completion due to the irreversibility of editing repair.


Preliminary Result


DNA flaps induce site-specific mutagenesis in plasmid model substrates in yeast and HEK cells. To test the proposed editing strategy, studies were initiated with model plasmid substrates containing mutagenic 3′ flaps that resemble the product of TPRT. A dual fluorescent protein reporter was created that encodes a stop codon between GFP and mCherry. Mutagenic flaps encode a correction to the stop codon (FIG. 9A), enabling mCherry synthesis. Thus, mutagenesis efficiency can be quantified by GFP:mCherry ratios. Plasmid substrates were prepared in vitro and introduced into yeast (S. cerevisiae) or human cells (HEK293). High frequency mutagenesis was observed in both systems (FIG. 9B), and isolated yeast colonies contained either the reverted base, the mutated base, or a mixture of both products (FIG. 9C). Detection of the latter suggests that plasmid replication occurred prior to MMR in these cases, and further suggests that flap excision and ligation precede MMR. This result establishes the feasibility of DNA editing using 3′ mutagenic strands.


Systematic Studies with Model Flap Substrates


Based on the preliminary results described above, a broader spectrum of flap substrates will be evaluated in HEK cells to infer principles of efficient editing. 3′ ssDNA flaps will be systematically varied to determine the influence of mismatch pairings, the location of the mutagenic nucleotide along the flap, and the identity of the terminal nucleotide (FIG. 9D). Single nucleotide insertions and deletions will also be tested. Amplicon sequencing will be used to analyze editing precision. These results will help inform the design of gRNA reverse transcription templates.


In vitro TPRT on plasmid substrates leads to efficient editing outcomes. TPRT reactions developed in Aim 1 were used to induce mutagenesis within a plasmid substrate. The reaction was carried out on circular DNA plasmid substrates (see FIG. 10). This rules out the possibility of DNA strand dissociation as the mechanism for RT extension in the previous in vitro experiments. It also allowed for the testing of DNA repair of flap substrates in cells. A dual-fluorescent reporter plasmid was constructed for yeast (S. cerevisiae) expression. This plasmid encodes GFP (green fluorescent protein) and mCherry (red fluorescent protein) with an intervening stop codon (TGA). Expression of this construct in yeast produces only GFP. The plasmid was used as a substrate for in vitro TRT [Cas9(H840A) nickase, engineered gRNA, MLV RT enzyme, dNTPS]. The gRNA extension encodes a mutation to the stop codon. The flap strand is used for repair of the stop codon and it is anticipated to produce a plasmid that expresses both GFP and mCherry as a fusion protein. Yeast dual-FP plasmid transformants are shown in FIG. 10. Transforming the parent plasmid or an in vitro Cas9(H840A) nicked plasmid results in only green GFP expressing colonies. TRT reaction with 5′-extended or 3′-extended gRNAs produces a mix of green and yellow colonies. The latter express both GFP and mCherry. More yellow colonies are observed with the 3′-extended gRNA. A positive control that contains no stop codon is shown as well.


This result establishes that long double stranded substrates can undergo TPRT, and that TPRT products induce editing in eukaryotic cells.


Another experiment similar to the foregoing prime editing experiment was carried out, but instead of installing a point mutation in the stop codon, TRT editing installs a single nucleotide insertion (left) or deletion (right) that repairs a frameshift mutation and allows for synthesis of downstream mCherry (see FIG. 11). Both experiments used 3′ extended gRNAs. Individual colonies from the TRT transformations were selected and analyzed by Sanger sequencing (see FIG. 12). Green colonies contained plasmids with the original DNA sequence, while yellow colonies contained the precise mutation designed by the TRT editing gRNA. No other point mutations or indels were observed.


Establish Prime Editing in HEK Cells Using RT-Cas9 Architectures


The optimized constructs from previous aims will be adapted for mammalian expression and editing at targeted sites in the human genome. Multiple RT enzymes and fusion architectures will be tested, in addition to adjacent targeting with secondary gRNAs (truncated to prevent nicking). Non-covalent RT recruitment will also be evaluated using the Sun-Tag system52 and MS2 aptamer system53. Indel formation assays will be used to evaluate targeting efficiency with standard gRNAs and RT-Cas9 fusions (as above). Then, for each genomic site, extended gRNAs and RT-Cas9 pairs will be assayed for single nucleotide editing. Editing outcomes will be evaluated with MiSeq.


Initial experiments in HEK cells were performed using Cas9-RT fusions. Editing by components expressed within cells requires a Cas9(H840A) nickase, a reverse transcriptase (expressed as a fusion or supplied in trans), and an engineered gRNA with a 3′ extension (see FIG. 14). Preliminary studies indicated that the length of the primer binding site within the gRNA extension was important for increasing the efficiency of editing in human cells (see FIG. 15).


Optimize Prime Editing Parameters in HEK Cells


After identifying Cas9-RT architectures that can perform prime editing in cells, the components and design will be optimized to achieve high efficiency editing. The location and nucleotide identity of the encoded point mutation, and the total length of the newly synthesized DNA strand, will be varied to evaluate editing scope and potential limitations. Short insertion and deletion mutations will also be evaluated. Protein expression constructs will be codon optimized. If successful, this would establish efficient prime editing in mammalian cells.


Preliminary Result. Additional gRNAs were designed to bring the RT enzyme to a higher local concentration at the editing locus, in the event that intramolecular reverse transcription by the fused RT enzyme were not possible. These auxiliary guides are truncated at the 5′ end (14-15 nt spacer), which has previously been shown to prevent Cas9 cutting but retain binding (see FIG. 16). The HEK3 locus was chosen to explore this strategy.


Potential Difficulties and Alternatives


1) gRNA degradation in cells: if extended gRNA termini are truncated in cells, stabilizing secondary structures could be installed, or synthetic gRNAs with stabilizing modifications could be tested. (2) No observed editing in human cells: additional strategies will be explored, including secondary targeting of RT-Cas9 fusions to adjacent genomic sites54. In addition, potential directed evolution strategies in E. coli or S. cerevisiae could be explored.


Significance


If prime editing could be established in experimental cell lines, this would have an immediate impact for basic biomedical research by enabling the rapid generation and characterization of a large number of point mutations in human genes. The generality of the method, and its orthogonal editing window with respect to base editors, would provide an approach to installing many currently inaccessible mutations. Moreover, if prime editing could be optimized for high efficiency and product purity, its potential applicability to correcting disease mutations in other human cell types would be significant.


3. Achieve Site-Specific Editing of Pathogenic Mutations in Cultured Human Cells.


Background and Rationale.


A large number of pathogenic mutations cannot be corrected by current base editors due to PAM restrictions, or a need for transversion or indel mutation correction. With prime editing, all transitions and transversions are theoretically possible, as may be small insertions and deletions. Moreover, in relation to the PAM, the prime editing window (anticipated −3 to +4) is distinct from that of base editors (−18 to −12) (FIG. 13). Mendelian conditions not currently correctable by base editors include: (1) the sickle cell disease Glu6Val founder mutation in hemoglobin beta (requires A•T to T•A transversion); (2) the most common Wilson's disease variant His1069Gln in ATP7B (requires G•C to T•A transversion); and (3) the ΔPhe508 mutation in CFTR that causes cystic fibrosis (requires 3-nucleotide insertion). Each of these targets contains an appropriately positioned PAM for SpCas9 targeting and prime editing.


Preliminary Results.


T to A Editing in HEK3 Cells is not Achievable by Current Base Editing but is Achievable by TRPT Editing (see FIGS. 17A-17C).



FIG. 17A shows a graph displaying the % T to A conversion at the target nucleotide after transfection of components in human embryonic kidney (HEK) cells. This data presents results using an N-terminal fusion of wild type MLV reverse transcriptase to Cas9(H840A) nickase (32-amino acid linker). Editing efficiency was improved dramatically when the length of the primer binding site is extended from 7 nucleotides to 11 or 12 nucleotides. Additionally, the auxiliary guide A, which is positioned just upstream of the editing locus (see FIG. 16), significantly improves editing activity, particularly for shorter length primer binding sites. Editing efficiency was quantified by amplicon sequencing using the Illumina MiSeq platform. FIG. 17B also shows % T to A conversion at the target nucleotide after transfection of components in human embryonic kidney (HEK) cells, but this data presents results using a C-terminal fusion of the RT enzyme. Here, the auxiliary guide A does not have as much of an effect, and editing efficiency is overall higher. FIG. 17C shows data presenting results using an N-terminal fusion of wild type MLV reverse transcriptase to Cas9(H840A) nickase similar to that used in FIG. 17A; however the linker between the MLV RT and Cas9 is 60 amino acids long instead of 32 amino acids.


T to A Editing at HEK3 Site by TRPT Editing Results Displays High Purity.



FIG. 18 shows the output of sequencing analysis by high-throughput amplicon sequencing. The output displays the most abundant genotypes of edited cells. Of note, no major indel products are obtained, and the desired point mutation (T to A) is cleanly installed without bystander edits. The first sequence shows the reference genotype. The top two products are the starting genotype containing an endogenous polymorphism (G or A). The bottom two products represent the correctly edited genotypes.


MLV RT Mutants Improve Editing.


Mutant reverse transcriptases, described in Baranauskas, et al (doi:10.1093/protein/gzs034), were tested as C-terminal fusions to the Cas9(H840A) nickase for target nucleotide editing in human embryonic kidney (HEK) cells. Cas9-RT editor plasmid was co-transfected with a plasmid encoding a 3′-prime editing guide RNA that templates reverse transcription. Editing efficiency at the target nucleotide (blue bars) is shown alongside indel rates (orange bars) in FIG. 19. WT refers to the wild type MLV RT enzyme. The mutant enzymes (M1 through M4) contain the mutations listed to the right. Editing rates were quantified by high throughput sequencing of genomic DNA amplicons.


Complementary Strand Nicking with a Second gRNA Improves Editing.


This experiment evaluates editing efficiency of the target nucleotide when a single strand nick is introduced in the complementary DNA strand in proximity to the target nucleotide, with the hypothesis being that this would direct mismatch repair to preferentially remove the original nucleotide and convert the base pair to the desired edit. The Cas9(H840A)-RT editing construct was co-transfected with two guide RNA encoding plasmids, one of which templates the reverse transcription reaction, while the other targets the complementary DNA strand for nicking. Nicking at various distances from the target nucleotide was tested (orange triangles) (see FIG. 20). Editing efficiency at the target base pair (blue bars) is shown alongside the indel formation rate (orange bars). The “none” example does not contain a complementary strand nicking guide RNA. Editing rates were quantified by high throughput sequencing of genomic DNA amplicons.



FIG. 21 shows processed high throughput sequencing data showing the desired T to A transversion mutation and general absence of other major genome editing byproducts.


Scope. The potential scope for the new editing technology is shown in FIG. 13 and is compared to deaminase-mediated base editor technologies. Previously developed base editors target a region ˜15±2 bp upstream of the PAM. By converting target C or A nucleotides to T or G, respectively, previously developed base editors enable all transition mutations (A:T to G:C conversions). However, previously developed base editors are unable to install transversion mutations (A to T, A to C, G to T, G to C, T to A, T to G, C to A, C to G). Moreover, if there are multiple target nucleotides in the editing window, additional undesired edits can result.


The new prime editing technology could theoretically install any nucleotide and base pair conversion, and potentially small insertion and deletion edits as well. With respect to the PAM, prime editing windows start at the site of DNA nicking (3 bases upstream of the PAM) and end at an as-of-yet undetermined position downstream of the PAM. Of note, this editing window is distinct from that of deaminase base editors. Because the TPRT systems performs editing using DNA polymerase enzymes, it potentially has all of their benefits including generality, precision, and fidelity.


Correct Pathogenic Mutations in Patient-Derived Cell Lines.


Cell lines harboring the relevant mutations (sickle cell disease: CD34+ hematopoietic stem cells; Wilson's disease: cultured fibroblasts; cystic fibrosis: cultured bronchial epithelia) will be obtained from ATCC, the Coriell Biobank, or collaborating Harvard/Broad affiliate laboratories. Editing efficiency will be evaluated by high throughput sequencing, and the efficacy of the corrected genotype will be tested using phenotypic assays (hemoglobin HPLC, ATP7B immunostaining, and CFTR membrane potential assays).


Characterize Off-Target Editing Activity.


Potential off-target editing will be screened with established methods such as GUIDE-seq55 and CIRCLE-seq56 using target gRNAs paired with wild type Cas9. If potential off-targets are identified, these loci will be probed in TPRT edited cells to identify true off-target editing events.


Potential Difficulties and Alternatives.


(1) Low editing efficiency: prime editor (PE)s may require optimization for each target. In this case, gRNA libraries can be tested to identify the highest functioning variants for specific applications. RT-Cas fusion expression and nuclear localization can be optimized. Liposomal RNP delivery could be used to limit off-target editing.


Upcoming Experiments.


Optimization of gRNA designs can be achieved by further exploration of the primer binding site length and extension of synthesis template. Testing scope and generality will include different nucleotide conversions, small insertions and deletions, as well as, different editing positions with respect to PAM, and multiple sites in the human genome. Optimization of RT component will include exploring mutations in MLV RT to enhance activity (Rnase H inactivation, increase primer-template binding affinity, adjustments to processivity), and new RT enzymes (group II intro RTs, other retroviral RTs).


Significance.


Myriad genetic disorders result from single nucleotide changes in individual genes. Developing the genome editing technology described here, and applying it in disease-relevant cell types, would establish a foundation for translation to the clinic. For some diseases, such as Sickle Cell Disease, a single point mutation represents the dominant genotype throughout the population. However, for many other genetic disorders, a large heterogeneity of different point mutations within a single gene is observed throughout the patient population, each of which gives rise to a similar disease phenotype. Therefore, as a general genome editing method that could in theory target a large number of such mutations, this technology could provide enormous potential benefit to many of these patients and their families. If proof of principle for these applications could be established in cells, it would establish the foundation to studies in animal models of disease.


Advantages


Precision: the desired edit is encoded directed in nucleic acid sequence. Generality: in theory, could be possible to make any base pair conversion, including transversion edits, as well as small insertions or deletions. There is a distinct editing window from that of base editors with respect to Cas9 protospacer adjacent motif (PAM) sequence. This method achieves many of the editing capabilities of homology-directed repair (HDR), but without the major limitations of HDR (inefficient in most cell types, and is usually accompanied by an excess of undesired byproducts such as indels). Also, it does not make double-stranded DNA breaks (DSBs, so few indels, translocations, large deletions, p53 activation, etc.


Example 3—Peptide Tagging with PE

The prime editing systems (i.e., PE systems) described herein may also be used to introduce various peptide tags into protein coding genes. Such tags can include HEXAhistidine tags, FLAG-tag, V5-tag, GCN4-tag, HA-tag, Myc-tag and others. This approach may be useful in applications such as protein fluorescent labeling, immunoprecipitation, immunoblotting, immunohistochemistry, protein recruitment, inducible protein degrons, and genome-wide screening. Embodiments are depicted in FIGS. 25 and 26.



FIG. 25 is a schematic showing gRNA design for peptide tagging genes at endogenous genomic loci and peptide tagging with TPRT genome editing (i.e., prime editing). The FlAsH and ReAsH tagging systems comprise two parts: (1) a fluorophore-biarsenical probe, and (2) a genetically encoded peptide containing a tetracysteine motif, exemplified by the sequence FLNCCPGCCMEP (SEQ ID NO: 1). When expressed within cells, proteins containing the tetracysteine motif can be fluorescently labeled with fluorophore-arsenic probes (see ref: J. Am. Chem. Soc., 2002, 124 (21), pp 6063-6076. DOI: 10.1021/ja017687n). The “sortagging” system employs bacterial sortase enzymes that covalently conjugate labeled peptide probes to proteins containing suitable peptide substrates (see ref: Nat. Chem. Biol. 2007 November; 3(11):707-8. DOI: 10.1038/nchembio.2007.31). The FLAG-tag (DYKDDDDK (SEQ ID NO: 2)), V5-tag (GKPIPNPLLGLDST (SEQ ID NO: 3)), GCN4-tag (EELLSKNYHLENEVARLKK (SEQ ID NO: 4)), HA-tag (YPYDVPDYA (SEQ ID NO: 5)), and Myc-tag (EQKLISEEDL (SEQ ID NO: 6)) are commonly employed as epitope tags for immunoassays. The pi-clamp encodes a peptide sequence (FCPF) (SEQ ID NO: 622) that can by labeled with a pentafluoro-aromatic substrates (ref: Nat. Chem. 2016 February; 8(2):120-8. doi: 10.1038/nchem.2413).



FIG. 26 shows precise installation of a His6-tag and a FLAG-tag into genomic DNA. A guide RNA targeting the HEK3 locus was designed with a reverse transcription template that encodes either an 18-nt His-tag insertion or a 24-nt FLAG-tag insertion. Editing efficiency in transfected HEK cells was assessed using amplicon sequencing. Note that the full 24-nt sequence of the FLAG-tag is outside of the viewing frame (sequencing confirmed full and precise insertion).


Example 4—RNA Tagging and Manipulation Using PE

A new method for the insertion of motifs into genetic sequences that tag or otherwise manipulate RNA within mammalian, eukaryotic, and bacterial cells is described herein. While it is estimated that only 1% of the human genome encodes proteins, virtually all of the genome is transcribed at some level. It is an open question how much of the resulting non-coding RNA (ncRNA) plays a functional role, let alone what the roles of most of these putative RNAs are. “Tagging” of these RNA molecules via the insertion of a novel RNA-encoding sequence with a useful property into genes of interest is a useful method for studying the biological functions of RNA molecules in cells. It can also be useful install tags onto protein-encoding mRNAs as a means to perturb and thus better understand how mRNA modifications can affect cellular function. For instance, a ubiquitous natural RNA tag—polyadenylation—is used by cells to affect transport of mRNA into the cytoplasm. Different types of polyadenylation signals result in different transport rates and different mRNA lifespans and—thus—differences in the levels to which the encoded protein is expressed.


A common approach for expressing tagged RNAs within cells is to exogenously introduce a synthetic construct using either (i) transient plasmid transfection that produces a short-term burst of gene expression, often at supraphysiologic levels; or (ii) permanent integration of the tagged RNA gene into the genome (at random sites) using lentiviral integration or transposons, which enables prolonged expression. Both of these approaches are limited by production of altered expression levels, and by the absence of natural mechanisms that regulate the expression or activity of the gene. An alternative strategy is to directly tag a gene of interest at its endogenous locus using homology-directed repair (HDR) of double-stranded DNA breaks induced by Cas9 or other targeted DNA nucleases. While this approach enables the generation of a wide range of endogenously tagged genes, HDR is markedly inefficient and so requires significant screening to identify the desired clonal population of cells that have been successfully tagged. Moreover, HDR is typically very inefficient or entirely inactive in a large number of cell types, most notably in post-mitotic cells. The low efficiency of HDR is further complicated by the generation of undesired indel products, would could be especially problematic in the case of RNA genes as they might lead to the production of an RNA whose activities interfere with the function of normal alleles. Finally, researchers often need to screen various tagging positions within an RNA molecule to achieve optimal performance Combined, these drawbacks make HDR a less desirable method for installation of tags in RNA.


Prime editing is a new genome editing technology that enables targeted editing of genomic loci via the transfer of genetic information from RNA to DNA. Using prime editing, RNA genes could be tagged with a variety of components such as RNA aptamers, ribozymes, or other RNA motifs. Prime editing has the potential to be faster, cheaper, and effective in a greater variety of cell types by comparison to HDR strategies. As such, the described invention represents a novel, useful, and non-obvious tool for investigating the biology of RNA genes in health and disease. A new method for the insertion of RNA motifs into genetic sequences that tag or otherwise manipulate RNA using prime editors (PEs) is described herein. PEs are capable of site-specifically inserting, mutating, and/or deleting multiple nucleotides at a desired genomic locus that is targetable by a CRISPR/Cas system. PEs are composed of fusions between Cas9 nuclease domains and reverse transcriptase domains. They are guided to their genomic target by engineered PEgRNAs (prime editing guide RNAs), which contain a guide spacer portion for DNA targeting, as well as a template for reverse transcription that encodes the desired genome edit (see FIG. 28A). It is envisioned that PE can be used to insert motifs that are functional at the RNA level (hereafter RNA motifs) to tag or otherwise manipulate non-coding RNAs or mRNAs. These motifs could serve to increase gene expression, decrease gene expression, alter splicing, change post-transcriptional modification, affect the sub-cellular location of the RNA, enable isolation or determination of the intra- or extra-cellular location of the RNA (using, for instance, fluorescent RNA aptamers such as Spinach, Spinach2, Baby Spinach, or Broccoli), recruit endogenous or exogenous protein or RNA binders, introduce sgRNAs, or induce processing of the RNA, by either self-cleavage or RNAses (see FIG. 28B). Due to the flexibility of prime editing, it is not possible to provide a comprehensive list of RNA motifs that could be installed within the genome. A series of examples are shown here that broadly illustrate the predicted scope of PE-installed RNA motifs that could be used to tag RNA genes. It is currently not possible to efficiently and fairly cleanly make these changes in most types of cells (including the many that do not support HDR) using any other reported genome editing method besides PE.


Gene expression could be affected by encoding a 3′ untranslated region (UTR) that results in changes in nuclear transport or retention or mRNA lifespan. For instance, the polyA tail from polyomavirus simian virus 40 (SV40) has additional helper sequences that enable efficient transcription termination and can increase gene expression relative to other 3′ UTRs57,58. Example sequence of SV40 polyA tail:









SV40 POLYA TAIL


(SEQ ID NO: 331)


AACTTGTTTATTGCAGCTTATAATGGTTACAAATAAAGCAATAGCATCA





CAAATTTCACAAATAAAGCATTTTTTTCACTGCATTCTAGTTGTGGTTT





GTCCAAACTCATCAATGTATCTTA






Post-translational modification signals, besides polyadenylation signals, could also be encoded by PE. These include signals incorporate N6-methyladenosine, N1-methyladenosine, 5-methylcytosine, and pseudouridine modifications59. By using PE to include sequences bound by enzymes that write or remove these modifications within an RNA transcript, it would be possible to induce their writing or erasing. This could be used as a tool to study the effects of these markers, to induce cellular differentiation, affect stress responses, or, given the function of these markers are as yet underexplored, affect targeted cells in other fashions.


PE could encode mutations that affect subcellular localization. For instance, incorporation of tRNA-Lys within an mRNA can theoretically result in transport to the mitochondria60, while various 3′ UTRs can result in nuclear retention or transport61.


Examples

SV40 polyA signal results in transport.









SV40 POLYA TAIL


(SEQ ID NO: 331)


AACTTGTTTATTGCAGCTTATAATGGTTACAAATAAAGCAATAGCATCA





CAAATTTCACAAATAAAGCATTTTTTTCACTGCATTCTAGTTGTGGTTT





GTCCAAACTCATCAATGTATCTTA






U1 snRNA 3′ box results in retention.









U1 SNRNA 3′ BOX


(SEQ ID NO: 625)


TTCATTCAGCAAGTTCAGAGAAATCTGAACTTGCTGGATTTTTGGAGC





AGGGAGATGGAATAGGAGCTTGCTCCGTCCACTCCACGCATCGACCTG





GTATTGCAGTACCTCCAGGAACGGTGCACCCACTTTCTGGAGTTTCAA





AAGTAGACTGTACGCTAAGGGTCATATCTTTTTTTGTTT





GGTTTGTGTCTTGGTTGGCGTCTTAA






Determining the sub-cellular localization of endogenous RNA can be challenging and requires the addition of exogenous, fluorescently-tagged nucleotide probes, as in the case of FISH, or time-consuming and potentially inaccurate cell fractionation followed by RNA detection. Encoding a probe within the endogenous RNA would obviate many of these issues. One example would be to encode a fluorescent RNA aptamer, such as Spinach62 or Broccoli within an endogenous RNA, thereby visualizing the presence of that RNA via addition of a small molecule proto-fluorophore.


Broccoli Aptamer:









BROCCOLI APTAMER


(SEQ ID NO: 357)


GAGACGGTCGGGTCCAGATATTCGTATCTGTCGAGTAGAGTGTGGGCT





C






PE could insert or remove sequences that encode RNA that are recognized by RNA binding proteins, affecting RNA stability, expression, localization, or modification (for instance, see proteins listed63).


PE could insert sequences that encode sgRNAs within the genome, as a viral or cancer defense mechanism. Similarly, it could be used to insert microRNAs (e.g., pre-microRNAs) to direct silencing of targeted genes.


PE could insert sequences resulting in processing of the RNA, either by itself, or by external factors, either as a therapy or tool for studying the function of various portions of the RNA. For instance, the HDV ribozyme64, when included within an RNA sequence, results in processing of the RNA immediately 5′ of the ribozyme, while the hammerhead ribozyme cleaves prior to the third stem within the ribozyme65. Other self-cleaving ribozymes include pistol66, hatchet67, hairpin67, Neuropora Varkud satellite68, glmS69, twister70, and twister sister66. These sequences could include wild-type or engineered or evolved versions of ribozymes. The majority of these ribozymes could have different sequences depending on the region of RNA into which they were associated, depending on where the ribozyme cut site is located. Sequences that would direct the processing of the RNA by external factors, such as sequence specific RNAses71, RNAses that recognize specific structures72—such as Dicer73 and Drosha74, could also be achieved.


HDV ribozyme:















HDV
GGCCGGCATGGTCCCAGCCTCCTCGCTGGCGCCGG


RIBOZYME
CTGGGCAACATGCTTCGGCATGGCGAATGGGAC 



(SEQ ID NO: 365)









REFERENCES FOR EXAMPLE 4

The following references are incorporated herein by reference in their entireties.

  • 1. Schek N, Cooke C, Alwine J C. Molecular and Cellular Biology. 1992.
  • 2. Gil A, Proudfoot N J. Cell. 1987.
  • 3. Zhao, B. S., Roundtree, I. A., He, C. Nat Rev Mol Cell Biol. 2017.
  • 4. Rubio, M. A. T., Hopper, A. K. Wiley Interdiscip Rev RNA 2011.
  • 5. Shechner, D. M., Hacisuleyman E, Younger, S. T., Rinn, J. L. Nat Methods. 2015.
  • 6. Paige, J. S., Wu, K. Y., Jaffrey, S. R. Science 2011.
  • 7. Ray D., . . . Hughes T R. Nature 2013.
  • 8. Chadalavada, D. M., Cerrone-Szakal, A. L., Bevilacqua, P.C. RNA 2007.
  • 9. Forster A C, Symons R H. Cell. 1987.
  • 10. Weinberg Z, Kim P B, Chen T H, Li S, Harris K A, Lünse C E, Breaker R R. Nat. Chem. Biol. 2015.
  • 11. Feldstein P A, Buzayan J M, Bruening G. Gene 1989.
  • 12. Saville B J, Collins R A. Cell. 1990.
  • 13. Winkler W C, Nahvi A, Roth A, Collins J A, Breaker R R. Nature 2004.
  • 14. Roth A, Weinberg Z, Chen A G, Kim P G, Ames T D, Breaker R R. Nat Chem Biol. 2013.
  • 15. Choudhury R, Tsai Y S, Dominguez D, Wang Y, Wang Z. Nat Commun. 2012.
  • 16. MacRae I J, Doudna J A. Curr Opin Struct Biol. 2007.
  • 17. Bernstein E, Caudy A A, Hammond S M, Hannon G J Nature 2001.
  • 18. Filippov V, Solovyev V, Filippova M, Gill S S. Gene 2000.


Example 5—Immunoepitope Insertion by PE

Precise genome targeting technologies using the CRISPR/Cas system have recently been explored in a wide range of applications, including the insertion of engineered DNA sequences into targeted genomic loci. Previously, homology-directed repair (HDR) has been used for this application, requiring an ssDNA donor template and repair initiation by means of a double-stranded DNA break (DSB). This strategy offers the broadest range of possible changes to be made in cells and is the only method available to insert large DNA sequences into mammalian cells. However, HDR is hampered by undesired cellular side effects stemming from its initiating DSB, such as high levels of indel formation, DNA translocations, large deletions, and P53 activation. In addition to these drawbacks, HDR is limited by low efficiency in many cell types (T cells are a notable exception to this observation). Recent efforts to overcome these drawbacks include fusing human Rad51 mutants to a Cas9 D10A nickase (RDN), resulting in a DSB-free HDR system that features improved HDR product:indel ratios and lower off target editing, but is still hampered by cell-type dependencies and only modest HDR editing efficiency.


Recently developed fusions of Cas9 to reverse transcriptases (“Prime editors”) coupled with PEgRNAs represent a novel genome editing technology that offers a number of advantages over existing genome editing methods, including the ability to install any single nucleotide substitution, and to insert or delete any short stretch of nucleotides (up to at least several dozen bases) in a site-specific manner Notably, PE edits are achieved with generally low rates of unintended indels. As such, PE enables targeted insertion-based editing applications that have been previously impossible or impractical.


This particular invention describes a method for using prime editing as a means to insert known immunogenicity epitopes into endogenous or foreign genomic DNA, resulting in modification of the corresponding proteins for therapeutic or biotechnological applications (see FIGS. 31 and 32). Prior to the invention of prime editing, such insertions could be achieved only inefficiently and with high rates of indel formation from DSBs. prime editing solves the problem of high indel formation from insertion edits while generally offering higher efficiency than HDR. This lower rate of indel formation presents a major advantage of prime editing over HDR as a method for targeted DNA insertions, especially in the described application of inserting immunogenicity epitopes. The length of epitopes is in a range from few bases to hundreds of bases. Prime editor is the most efficient and cleanest technology to achieve such targeted insertions in mammalian cells.


The key concept of the invention is the use of prime editors to insert a nucleotide sequence containing previously described immunogenicity epitopes into endogenous or foreign genomic DNA for the downregulation and/or destruction of their protein products and/or expressing cell types. Nucleotide sequences for immunogenic epitope insertion would be targeted to genes in a manner to produce fusion proteins of the targeted gene's coded protein and the inserted immunogenic epitope's corresponding protein translation. Patient's immune systems will have been previously trained to recognize these epitopes as a result of standard prior immunization from routine vaccination against, for example, tetanus or diphtheria or measles. As a result of the immunogenic nature of the fused epitopes, patient's immune systems would be expected to recognize and disable the prime edited protein (not just the inserted epitope) and potentially the cells from which it was expressed.


Fusions to targeted genes would be engineered as needed to ensure the inserted epitope protein translation is exposed for immune system recognition. This could include targeted nucleotide insertions resulting in protein translations yielding C-terminal fusions of immunogenicity epitopes to targeted genes, N-terminal fusions of immunogenicity epitopes to targeted genes, or the insertion of nucleotides into genes so that immunogenicity epitopes are coded within surfaced-exposed regions of protein structure.


Protein linkers encoded as nucleotides inserted between the target gene sequence and the inserted immunogenicity epitope nucleotide sequence may need to be engineered as part of this invention to facilitate immune system recognition, cellular trafficking, protein function, or protein folding of the targeted gene. These inserted nucleotide-encoded protein linkers may include (but are not limited to) variable lengths and sequences of the XTEN linker or variable lengths and sequences of Glycine-Serine linkers. These engineered linkers have been previous used to successfully facilitate protein fusions.


Distinguishing features of this invention include the ability to use previously acquired immune responses to specific amino acid sequences as a means to induce an immune response against otherwise non-immunogenic proteins. Another distinguishing feature is the ability to insert the nucleotide sequences of these immunogenic epitopes in a targets manner that does not induce high levels of unwanted indels as a by-product editing and is efficient in its insertion. The invention discussed herein also has the ability to combine cell type-specific delivery methods (such as AAV serotypes) to insert epitopes in cell types that are of interest to trigger an immune response to.


Prime editing as a means of inserting immunogenic epitopes into pathogenic genes could be used to program the patient's immune system to fight a wide variety of diseases (not limited to cancer as with immuno-oncology strategies). An immediately relevant use of this technology would be as a cancer therapeutic as it could undermine a tumor's immune escape mechanism by causing an immune response to a relevant oncogene like HER2 or growth factors like EGFR. Such an approach could seem similar to T-cell engineering, but one novel advance of this approach is that it can be utilized in many cell types and for diseases beyond cancer, without needed to generate and introduce engineered T-cells into patients.


Using PE to insert an immunogenicity epitope which most people are already vaccinated against (tetanus, pertussis, diphtheria, measles, mumps, rubella, etc.) into a foreign or endogenous gene that drives a disease, so the patient's immune system learns to disable that protein.


Diseases that stand to have a potential therapeutic benefit from the aforementioned strategy include those caused by aggregation of toxic proteins, such as in fatal familial insomnia. Other diseases that could benefit include those caused by pathogenic overexpression of an otherwise nontoxic endogenous protein, and those caused by foreign pathogens.


Primary therapeutic indications include those mentioned above such as therapeutics for cancer, prion and other neurodegenerative diseases, infectious diseases, and preventative medicine. Secondary therapeutic indications may include preventative care for patients with late-onset genetic diseases. It is expected that current standard of care medicines may be used in conjunction with prime editing for some diseases, like particularly aggressive cancers, or in cases where medications help alleviate disease symptoms until the disease completely cured.


Below are Examples of Immunogenic Epitopes that can by Inserted by Prime Editing can be Used to Achieve:

















Epitope Amino
Example Nucleic


Vaccine
Disease
Acid Sequence
Acid Sequence (8)







 1
Tetanus toxoid
QYIKANSKFIGITEL
CATGATATAAAAGCAAATTCTAAATTTA




(SEQ ID NO: 396)
TAGGTATAACTGAACTA (SEQ ID NO:





397)





 2
Diphtheria
GADDVVDSSKSFV
GGCGCCGACGACGTGGTGGACAGCAGC



toxin mutant
MENFSSYHGTKPG
AAGAGCTTCGTGATGGAGAACTTCAGC



CRM197
YVDSIQKGIQKPKS
AGCTACCACGGCACCAAGCCCGGCTAC




GTQGNYDDDWKEF
GTGGACAGCATCCAGAAGGGCATCCAG




YSTDNKYDAAGYS
AAGCCCAAGAGCGGCACCCAGGGCAAC




VDNENPLSGKAGG
TACGACGACGACTGGAAGGAGTTCTAC




VVKVTYPGLTKVL
AGCACCGACAACAAGTACGACGCCGCC




ALKVDNAETIKKEL
GGCTACAGCGTGGACAACGAGAACCCC




GLSLTEPLMEQVGT
CTGAGCGGCAAGGCCGGCGGCGTGGTG




EEFIKRFGDGASRV
AAGGTGACCTACCCCGGCCTGACCAAG




VLSLPFAEGSSSVE
GTGCTGGCCCTGAAGGTGGACAACGCC




YINNWEQAKALSV
GAGACCATCAAGAAGGAGCTGGGCCTG




ELEINFETRGKRGQ
AGCCTGACCGAGCCCCTGATGGAGCAG




DAMYEYMAQACA
GTGGGCACCGAGGAGTTCATCAAGAGG




GNRVRRSVGSSLSC
TTCGGCGACGGCGCCAGCAGGGTGGTG




INLDWDVIRDKTKT
CTGAGCCTGCCCTTCGCCGAGGGCAGC




KIESLKEHGPIKNK
AGCAGCGTGGAGTACATCAACAACTGG




MSESPNKTVSEEKA
GAGCAGGCCAAGGCCCTGAGCGTGGAG




KQYLEEFHQTALEH
CTGGAGATCAACTTCGAGACCAGGGGC




PELSELKTVTGTNP
AAGAGGGGCCAGGACGCCATGTACGAG




VFAGANYAAWAV
TACATGGCCCAGGCCTGCGCCGGCAAC




NVAQVIDSETADNL
AGGGTGAGGAGGAGCGTGGGCAGCAGC




EKTTAALSILPGIGS
CTGAGCTGCATCAACCTGGACTGGGAC




VMGIADGAVHHNT
GTGATCAGGGACAAGACCAAGACCAAG




EEIVAQSIALSSLMV
ATCGAGAGCCTGAAGGAGCACGGCCCC




AQAIPLVGELVDIG
ATCAAGAACAAGATGAGCGAGAGCCCC




FAAYNFVESIINLFQ
AACAAGACCGTGAGCGAGGAGAAGGCC




VVHNSYNRPAYSP
AAGCAGTACCTGGAGGAGTTCCACCAG




GHKTQPFLHDGYA
ACCGCCCTGGAGCACCCCGAGCTGAGC




VSWNTVEDSIIRTG
GAGCTGAAGACCGTGACCGGCACCAAC




FQGESGHDIKITAE
CCCGTGTTCGCCGGCGCCAACTACGCCG




NTPLPIAGVLLPTIP
CCTGGGCCGTGAACGTGGCCCAGGTGA




GKLDVNKSKTHISV
TCGACAGCGAGACCGCCGACAACCTGG




NGRKIRMRCRAIDG
AGAAGACCACCGCCGCCCTGAGCATCC




DVTFCRPKSPVYVG
TGCCCGGCATCGGCAGCGTGATGGGCA




NGVHANLHVAFHR
TCGCCGACGGCGCCGTGCACCACAACA




SSSEKIHSNEISSDSI
CCGAGGAGATCGTGGCCCAGAGCATCG




GVLGYQKTVDHTK
CCCTGAGCAGCCTGATGGTGGCCCAGG




VNSKLSLFFEIKS
CCATCCCCCTGGTGGGCGAGCTGGTGG




(SEQ ID NO: 630)
ACATCGGCTTCGCCGCCTACAACTTCGT





GGAGAGCATCATCAACCTGTTCCAGGT





GGTGCACAACAGCTACAACAGGCCCGC





CTACAGCCCCGGCCACAAGACCCAGCC





CTTCCTGCACGACGGCTACGCCGTGAGC





TGGAACACCGTGGAGGACAGCATCATC





AGGACCGGCTTCCAGGGCGAGAGCGGC





CACGACATCAAGATCACCGCCGAGAAC





ACCCCCCTGCCCATCGCCGGCGTGCTGC





TGCCCACCATCCCCGGCAAGCTGGACGT





GAACAAGAGCAAGACCCACATCAGCGT





GAACGGCAGGAAGATCAGGATGAGGTG





CAGGGCCATCGACGGCGACGTGACCTT





CTGCAGGCCCAAGAGCCCCGTGTACGT





GGGCAACGGCGTGCACGCCAACCTGCA





CGTGGCCTTCCACAGGAGCAGCAGCGA





GAAGATCCACAGCAACGAGATCAGCAG





CGACAGCATCGGCGTGCTGGGCTACCA





GAAGACCGTGGACCACACCAAGGTGAA





CAGCAAGCTGAGCCTGTTCTTCGAGATC





AAGAGC (SEQ ID NO: 399)





 3
mumps
GTYRLIPNARANLT
GGCACCTACAGGCTGATCCCCAACGCC




A (SEQ ID NO: 400)
AGGGCCAACCTGACCGCC (SEQ ID NO:





401)





 4
mumps
PSKFFTISDSATFAP
Ccgagcaaattttttaccattagcgat




GPVSNA (SEQ ID
agcgcgacctttgcgccgggcccggtg




NO: 402)
agcaacgcg 




PSKLFIMLDNATFA
(SEQ ID NO: 403)




PGPVVNA (SEQ ID
Ccgagcaaactgtttattatgctggat




NO: 404)
aacgcgacctttgcgccgggcccggtg





gtgaacgcg (SEQ ID NO: 405)





Selected examples from 





Hemagglutinin-neuraminidase





(HN) diversity among outbreak





strains (table1) Divergence





between vaccine strain JL5





and outbreak strains (table2)





 5
Rubella virus
TPPPYQVSCGGESD
ACCCCCCCCCCCTACCAGGTGAGCTGCG



(RV)
RASARVIDPAAQS
GCGGCGAGAGCGACAGGGCCAGCGCCA




(SEQ ID NO: 406)
GGGTGATCGACCCCGCCGCCCAGAGC





(SEQ ID NO: 407)





 6
Hemagglutinin
PEYAYKIVKNKKM
CCCGAGTACGCCTACAAGATCGTGAAG




EDGFLQGMVDGW
AACAAGAAGATGGAGGACGGCTTCCTG




YGHHSNEQGSGLM
CAGGGCATGGTGGACGGCTGGTACGGC




ENERTLDKANPNN
CACCACAGCAACGAGCAGGGCAGCGGC




DLCSWSDHEASSN
CTGATGGAGAACGAGAGGACCCTGGAC




NTNQEDLLQRESRR
AAGGCCAACCCCAACAACGACCTGTGC




KKRIGTSTLNQRGN
AGCTGGAGCGACCACGAGGCCAGCAGC




CNTKCQTEEARLK
AACAACACCAACCAGGAGGACCTGCTG




REEVSLVKSDQCSN
CAGAGGGAGAGCAGGAGGAAGAAGAG




GSLQCRANNSTEQ
GATCGGCACCAGCACCCTGAACCAGAG




VD 
GGGCAACTGCAACACCAAGTGCCAGAC




(SEQ ID NO: 408)
CGAGGAGGCCAGGCTGAAGAGGGAGG





AGGTGAGCCTGGTGAAGAGCGACCAGT





GCAGCAACGGCAGCCTGCAGTGCAGGG





CCAACAACAGCACCGAGCAGGTGGAC





(SEQ ID NO: 409)





 7
Neuraminidase
TKSTNSRSGGISGP
ACCAAGAGCACCAACAGCAGGAGCGGC




DNEAPVGEAPSPYG
GGCATCAGCGGCCCCGACAACGAGGCC




DNPRPNDGNNIRIG
CCCGTGGGCGAGGCCCCCAGCCCCTAC




SKGYNGIITDTIEES
GGCGACAACCCCAGGCCCAACGACGGC




CSCYPDAKVVKSV
AACAACATCAGGATCGGCAGCAAGGGC




ELDSTIWTSGSSPN
TACAACGGCATCATCACCGACACCATC




QKIITIGWDPNGWT
GAGGAGAGCTGCAGCTGCTACCCCGAC




GTPMSPNGAYGTD
GCCAAGGTGGTGAAGAGCGTGGAGCTG




GPSNGQANQHQAE
GACAGCACCATCTGGACCAGCGGCAGC




SISAGNSSLCPIRDN
AGCCCCAACCAGAAGATCATCACCATC




WHGSNRSWSWPD
GGCTGGGACCCCAACGGCTGGACCGGC




GAE (SEQ ID NO:
ACCCCCATGAGCCCCAACGGCGCCTAC




410)
GGCACCGACGGCCCCAGCAACGGCCAG





GCCAACCAGCACCAGGCCGAGAGCATC





AGCGCCGGCAACAGCAGCCTGTGCCCC





ATCAGGGACAACTGGCACGGCAGCAAC





AGGAGCTGGAGCTGGCCCGACGGCGCC





GAG (SEQ ID NO: 411)





 8
TAP (transport
EKIVLLLAMMEKIV
GAGAAGATCGTGCTGCTGCTGGCCATG



antigen
LLLAKCQTPMGAIK
ATGGAGAAGATCGTGCTGCTGCTGGCC



presentation)
AVDGVTNKCPYLG
AAGTGCCAGACCCCCATGGGCGCCATC



on H5N1 virus
SPSF (SEQ ID NO:
AAGGCCGTGGACGGCGTGACCAACAAG



hemagglutinin
412)
TGCCCCTACCTGGGCAGCCCCAGCTTC





(SEQ ID NO: 413)





 9
TAP (transport
IRPCFWVELNPNQK
ATCAGGCCCTGCTTCTGGGTGGAGCTGA



antigen
IITIRPCFWVELICYP
ACCCCAACCAGAAGATCATCACCATCA



presentation)
DAGEIT (SEQ ID
GGCCCTGCTTCTGGGTGGAGCTGATCTG



on h5n1 virus
NO: 414)
CTACCCCGACGCCGGCGAGATCACC



neuraminidase

(SEQ ID NO: 415)





10
hemagglutinin
MEKIVLLLAEKIVL
ATGGAGAAGATCGTGCTGCTGCTGGCC



epitopes toward
LLAMCPYLGSPSFK
GAGAAGATCGTGCTGCTGCTGGCCATGT



class I HLA
CQTPMGAIKAVDG
GCCCCTACCTGGGCAGCCCCAGCTTCAA




VTNK (SEQ ID NO:
GTGCCAGACCCCCATGGGCGCCATCAA




416)
GGCCGTGGACGGCGTGACCAACAAG





(SEQ ID NO: 417)





11
neuraminidase
NPNQKIITICYPDAGE
AACCCCAACCAGAAGATCATCACCATCT



epitopes toward
ITIRPCFWVELRPCFW
GCTACCCCGACGCCGGCGAGATCACCAT



class I HLA
VELI (SEQ ID NO:
CAGGCCCTGCTTCTGGGTGGAGCTGAGGC




418)
CCTGCTTCTGGGTGGAGCTGATC (SEQ ID





NO: 419)





12
hemagglutinin
MVSLVKSDQIGTSTL
ATGGTGAGCCTGGTGAAGAGCGACCAGA



epitopes toward
NQR 
TCGGCACCAGCACCCTGAACCAGAGG



class II HLA
(SEQ ID NO: 420)
(SEQ ID NO: 421)





13
neuraminidase
YNGIITDTI (SEQ ID
TACAACGGCATCATCACCGACACCATC



epitopes toward
NO: 422)
(SEQ ID NO: 423)



class II HLA







14
hemagglutinin
MEKIVLLLAEKIVLL
ATGGAGAAGATCGTGCTGCTGCGGCCG



epitope H5N1-
LAMMVSLVKSDQCP
AGAAGATCGTGCTGCTGCTGGCCATGATG



bound class I
YLGSPSFIGTSTLNQR
GTGAGCCTGGTGAAGAGCGACCAGTGCC



and class II
KCQTPMGAIKAVDG
CCTACCTGGGCAGCCCCAGCTTCATCGGC



HLA
VTNK (SEQ ID NO:
ACCAGCACCCTGAACCAGAGG (SEQ ID




424)
NO: 425)





15
neuraminidase
NPNQKIITIYNGIITDT
AACCCCAACCAGAAGATCATCACCATCT



epitope H5N1-
ICYPDAGEITIRPCFW
ACAACGGCATCATCACCGACACCATCTGC



bound class I
VELRPCFWVELI
TACCCCGACGCCGGCGAGATCACCATCA



and class II
(SEQ ID NO: 426)
GGCCCTGCTTCTGGGTGGAGCTGAGGCCC



HLA

TGCTTCTGGGTGGAGCTGATC (SEQ ID





NO: 427)









Below are additional examples of epitopes that may be integrated into a target gene for immunoepitope taggin:


REFERENCES CITED IN EXAMPLE 5

The following references are incorporated by reference in their entireties.

  • 1. X. Wen, K. Wen, D. Cao, G. Li, R. W. Jones, J. Li, S. Szu, Y. Hoshino, L. Yuan, Inclusion of a universal tetanus toxoid CD4(+) T cell epitope P2 significantly enhanced the immunogenicity of recombinant rotavirus ΔVP8* subunit parenteral vaccines. Vaccine 32, 4420-4427 (2014).
  • 2. G. Ada, D. Isaacs, Carbohydrate-protein conjugate vaccines. Clin Microbiol Infect 9, 79-85 (2003).
  • 3. E. Malito, B. Bursulaya, C. Chen, P. L. Surdo, M. Picchianti, E. Balducci, M. Biancucci, A. Brock, F. Berti, M. J. Bottomley, M. Nissum, P. Costantino, R. Rappuoli, G. Spraggon, Structural basis for lack of toxicity of the diphtheria toxin mutant CRM197. Proceedings of the National Academy of Sciences 109, 5229 (2012).
  • 4. J. de Wit, M. E. Emmelot, M. C. M. Poelen, J. Lanfermeijer, W. G. H. Han, C. van Els, P. Kaaijk, The Human CD4(+) T Cell Response against Mumps Virus Targets a Broadly Recognized Nucleoprotein Epitope. J Virol 93, (2019).
  • 5. M. May, C. A. Rieder, R. J. Rowe, Emergent lineages of mumps virus suggest the need for a polyvalent vaccine. Int J Infect Dis 66, 1-4 (2018).
  • 6. M Ramamurthy, P. Rajendiran, N. Saravanan, S. Sankar, S. Gopalan, B. Nandagopal, Identification of immunogenic B-cell epitope peptides of rubella virus E1 glycoprotein towards development of highly specific immunoassays and/or vaccine. Conference Abstract, (2019).
  • 7. U. S. F. Tambunan, F. R. P. Sipahutar, A. A. Parikesit, D. Kerami, Vaccine Design for H5N1 Based on B- and T-cell Epitope Predictions. Bioinform Biol Insights 10, 27-35 (2016).


Example 6—Use of PE to Enable Chemical-Induced Dimerization of Target Proteins In Vivo

The prime editors described herein may also be used to place dimerization-induced biological processes, such as receptor signaling, under control of a convenient small-molecule drug by the genomic integration of genes encoding small-molecule binding proteins with prime editing is described herein. Using the prime editors described herein, the gene sequence encoding a small-molecule binding protein may be inserted within a gene encoding a target protein of interest in a living cell or patient. This edit alone should have no physiological effect. Upon administration of the small-molecule drug, which typically is a dimeric small molecule that can simultaneously bind to two drug-binding protein domains each of which is fused to a copy of the target protein, the small-molecule induces dimerization of the targeted protein. This target protein dimerization event then induces a biological signaling event, such as erythropoiesis or insulin signaling.


Example 7—DNA Sequence Insertion, Deletion, and Replacement with Dual Prime Editors

This Example describes the use of multi-flap prime editing, specifically using dual prime editors, for the precise insertion of new DNA sequence, the precise deletion of endogenous genomic DNA sequence, or the replacement of an endogenous genomic DNA sequence with a new DNA sequence. See FIG. 90. This invention addresses a long-standing objective in genome engineering: the programmable insertion, deletion, or replacement of DNA sequences in the genomes of living cells. This technology could be applied to modify or edit the genome of any living cell for research, therapeutic, or industrial applications.


Previously, homology directed repair (HDR) of nuclease-induced double-strand DNA breaks (DSBs) has been used to insert, delete, or replace DNA sequences in the genomes of living cells. While versatile, the low efficiency of HDR limits its application in many mammalian cell types, especially therapeutically relevant human cell types1-4. Moreover, the generation of DSBs leads to preferential repair by error-prone end-joining mechanisms that produce insertion and deletion (indel) repair products which cannot be subsequently converted to the desired edited DNA sequence5-10. As a result, indels typically represent the majority of modified genomic DNA products when nucleases are used in mammalian cells. Moreover, DSBs generated by nucleases can lead to undesired genomic changes such as large deletions and chromosomal translocations11, and the activation of p5312,13. Structural variants (SVs) can also be generated or corrected using programmable nucleases without relying on homologous recombination, though nuclease-based strategies suffer similar drawbacks for these applications. For example, dual-cutting strategies can be used to invert a targeted genomic DNA sequence, but most product alleles contain large deletions between the cut sites instead of the desired inverted sequence, or indels at the individual cut sites.


Prime editing is a recently reported genome editing technology that enables the insertion, deletion, or replacement of genomic DNA sequences without requiring error-prone double-strand DNA breaks14. Prime editing uses an engineered Cas9 nickase-reverse transcriptase fusion protein (PE1 or PE2) paired with an engineered prime editing guide RNA (pegRNA) that both directs Cas9 to the target genomic site and encodes the information for installing the desired edit. Prime editing proceeds through a multi-step editing process: 1) the Cas9 domain binds and nicks the target genomic DNA site, which is specified by the pegRNA's spacer sequence; 2) the reverse transcriptase domain uses the nicked genomic DNA as a primer to initiate the synthesis of an edited DNA strand using an engineered extension on the pegRNA as a template for reverse transcription—this generates a single-stranded 3′ flap containing the edited DNA sequence; 3) cellular DNA repair resolves the 3′ flap intermediate by the displacement of a 5′ flap species that occurs via invasion by the edited 3′ flap, excision of the 5′ flap containing the original DNA sequence, and ligation of the new 3′ flap to incorporate the edited DNA strand, forming a heteroduplex of one edited and one unedited strand; and 4) cellular DNA repair replaces the unedited strand within the heteroduplex using the edited strand as a template for repair, completing the editing process.


Efficient incorporation of the desired edit requires that the newly synthesized 3′ flap contains a portion of sequence that is homologous to the genomic DNA site. This homology enables the edited 3′ flap to compete with the endogenous DNA strand (the corresponding 5′ flap) for incorporation into the DNA duplex. Because the edited 3′ flap will contain less sequence homology than the endogenous 5′ flap, the competition is expected to favor the 5′ flap strand. Thus, the 3′ flap, which contains the edit, may fail to effectively invade and displace the 5′ flap strand. Moreover, successful 3′ flap invasion and removal of the 5′ flap only incorporates the edit on one strand of the double-stranded DNA genome. Permanent installation of the edit requires cellular DNA repair to replace the unedited complementary DNA strand using the edited strand as a template. While the cell can be made to favor replacement of the unedited strand over the edited strand (step 4 above) by the introduction of a nick in the unedited strand adjacent to the edit using a secondary sgRNA (the PE3 system), this process still relies on a second stage of DNA repair. These DNA repair steps may be particularly inefficient for edits which require equilibration of long 5′ and 3′ flap intermediates or contain long non-homologous regions, such as long insertions or long deletions.


This Example describes a dual prime editing system (or a dual-flap prime editing system) that addresses the challenges associated with flap equilibration and subsequent incorporation of the edit into the non-edited complementary genomic DNA strand by simultaneously editing both DNA strands. In the dual-flap prime editing system, two pegRNAs are used to target opposite strands of a genomic site and direct the synthesis of two complementary 3′ flaps containing edited DNA sequence (FIG. 91). Unlike classical prime editing, there is no requirement for the pair of edited DNA strands (3′ flaps) to directly compete with 5′ flaps in endogenous genomic DNA, as the complementary edited strand is available for hybridization instead. Since both strands of the duplex are synthesized as edited DNA, the dual-flap prime editing system obviates the need for the replacement of the non-edited complementary DNA strand required by classical prime editing. Instead, cellular DNA repair machinery need only excise the paired 5′ flaps (original genomic DNA) and ligate the paired 3′ flaps (edited DNA) into the locus. Therefore, there is also no need to include sequences homologous to genomic DNA in the newly synthesized DNA strands, allowing selective hybridization of the new strands and facilitating edits that contain minimal genomic homology. Nuclease-active versions of prime editors that cut both strands of DNA could also be used to accelerate the removal of the original DNA sequence.


Depending on the orientation of the staggered DNA nicks generated by dual prime editors, the system can either replace the sequence between the two nicks with a new desired sequence (5′ overhang staggered orientation), or insert new DNA sequence with concomitant target site duplication (3′ overhang staggered orientation). FIG. 90 shows an orientation (5′ displaced nicks) where the sequence between the nicks is replaced. Of note, the new 3′ flaps need not overlap completely along the length of the new DNA sequence, but instead can be complementary in just a portion of the new sequence. In this way, each flap can be shorter than the full length of the new DNA sequence.


Dual-Flap Prime Editing at the HEK293 Site 3 Locus


Dual-flap prime editing at the HEK293 site 3 locus in human cells (HEK293T) was used to achieve highly efficient introduction of heterologous DNA sequences. For example, replacement of a 90-bp sequence in the locus with a new 22-bp sequence that encodes a 6×His tag occurred with greater than 80% efficiency (FIG. 92 allele table). Similarly, >60% yield was achieved for replacing this same 90-bp sequence with a GFP-11 peptide sequence (52 bp). The GFP-11 tag insertion was also performed at the C-terminus of the MYC gene with an efficiency of >20%, occurring with concomitant programmed deletion of a portion of the Myc 3′ UTR sequence.


Dual-Flap Prime Editing pegRNAs


The general designs of pegRNAs used for dual-flap prime editing are shown in FIG. 93. pegRNAs used for dual-flap prime editing have a similar design to those used for classic prime editing, however it is not necessary for the RT template region to encode any homology to the target locus. Instead, the two pegRNAs may contain RT templates that encode the synthesis of 3′ flaps whose 3′ ends are reverse complement sequences of one another. This complementarity between the 3′ flaps promotes their annealing and replacement of the endogenous DNA sequence with the intended new DNA sequence. This necessitates that the 5′ regions of the RT templates in the two pegRNAs are reverse complement sequences to one another, and this amount of complementarity can vary (FIG. 93).


Use of Dual-Flap Prime Editing to Install Recombination Sites


Dual-flap prime editing has many potential applications, such as installing peptide tags, RNA tags, immunoepitopes, dimerization domains, and recombinase target sites.


One such application is the installation of recombinase or integrase sequences at user-specified locations in the genome. FIG. 94 illustrates the installation of Bxb1 recombinase attB (38 bp) and attP (50 bp) sites into a targeted region of the human genome (HEK293T site 3, or HEK3) with simultaneous deletion of 90 bp of intervening sequence between the two nick sites. Various degrees of complementarity between 3′ flaps allow for successful editing, though longer sequences of complementarity produce more favorable ratios of desired edits to indels. Other dual-flap prime editing applications include endogenous tagging of genes with peptide or protein sequences, or the replacement of exons with new DNA sequences that have the potential to substitute for multiple variants for which the mutation falls within the exon sequence.


Dual-flap prime editing can be used to introduce one or two recombinase sites at targeted positions in the human genome. If single recombinase sites are inserted, these can be used as landing sites for a recombinase-mediated reaction between the genomic recombinase site and a second recombinase site within an exogenously supplied DNA, such as a plasmid. This enables the targeted integration of DNA cargo. If two recombinase sites are inserted in adjacent regions of DNA, depending on the orientation of the recombinase sites, these can be used for recombinase-mediated excision or inversion of the intervening sequence, or for recombinase-mediated cassette exchange with exogenous DNA for cargo integration. Integration of compatible recombinase sites on different chromosomes enables targeted and directional chromosomal translocation. Dual-flap prime editing can be used to efficiently introduce recombinase sites at a number of loci in the human genome (FIG. 94). Thus, the pairing of recombinase site integration by dual-flap prime editing with DNA recombinase enzymes represents a powerful approach for achieving many types of SV edits and target integration of DNA cargo.


Example 8—DNA Sequence Insertion, Deletion, Inversion, Translocation, and Integration Using Quadruple-Flap Prime Editing

This Example describes the use of multi-flap prime editing, specifically quadruple-flap prime editing, for targeted insertion, deletion, duplication, and replacement of endogenous genomic DNA sequences, and for targeted genomic DNA sequence inversion, targeted chromosomal translocation, and targeted integration of exogenous DNA. This technology has the potential to address long-standing objectives in genome engineering: the programmable installation or correction of structural variants (generally defined as >50 bp of genomic sequence change, often represented by large deletions, insertions/duplications, inversion, and translocations), and the targeted integration of DNA cargo at a specific location in the human genome. The latter could enable gene augmentation therapeutic strategies and other biotechnology applications. This technology could potentially be applied to modify or edit the genome of any living cell for research, therapeutic, or industrial applications.


This invention describes a multi-flap prime editing system that addresses the challenges associated with flap equilibration and subsequent incorporation of the edit into the non-edited complementary genomic DNA strand by simultaneously editing both DNA strands. It also expands upon the capabilities of traditional prime editing by allowing the newly synthesized 3′ flaps to direct larger sequence rearrangements.


A quadruple-flap prime editing approach was developed that can directly carry out desired sequence alterations without the need for a recombinase enzyme (as in dual-flap prime editing) but with similar versatility. In quadruple-flap prime editing, four different pegRNA sequences are delivered to cells along with the PE2 editor. One pair of pegRNAs template the synthesis of complementary DNA flaps, as with dual-flap prime editing, while the other pair of pegRNAs template the synthesis of an orthogonal pair of complementary DNA flaps. The location of these flaps, their relative orientation, and the pairing of complementarity dictates which class of rearrangement will occur. The junctions of the rearranged DNA contain the sequence encoded by the pegRNA-templated 3′ flaps, which direct the orientation of the rearrangement and can serve as useful DNA sequence for downstream processes. Targeted genomic sequence inversion was performed with the quadruple-flap prime editing strategy (FIG. 95). An inversion of a 2.7-kb sequence at the AAVS1 locus in HEK293T cells was achieved using four pegRNAs that place Bxb1 attB and attP sequences at the inversion junctions. Verification of the expected junctions was performed using amplicon sequencing, which demonstrated the correct junctions containing the attB or attP site sequences in ≥90% of the sequencing reads (FIG. 96).


Targeted integration of exogenous DNA plasmids was also achieved with quadruple-flap prime editing. In this approach, two pegRNAs target the endogenous genomic DNA site while two pegRNAs target a region on the cargo plasmid. 3′ DNA flaps are synthesized on both the genomic DNA and plasmid DNA so that the plasmid and genomic DNA will be bridged by hybridized 3′ flaps (FIG. 97A). The excision of 5′ overhangs and ligation of the nicks results in an integrated plasmid product. The orientation of integration can be controlled by the pairing of pegRNA template sequences.


A plasmid encoding the expression of a H2B-GFP fusion protein was integrated at the AAVS1 locus in HEK293T cells using quadruple-flap prime editing. This resulted in 1.6% of cells obtaining long-lasting, nuclear localized GFP fluorescence, as assessed by flow cytometry. PCR of a predicted junction showed the expected product containing the pegRNA-templated BxB1 attP sequence (FIG. 97B).


Chromosomal translocations were also performed using quadruple flap prime editing (FIG. 98A) to direct a chromosomal translocation between the MYC locus on chromosome 8 and the TIMM44 locus on chromosome 19 (FIG. 98B).


Dual flap prime editing at the IDS locus was performed with 12 pairs of pegRNAs and PE2. HEK293T cells were transfected with PE2 and different pairs of pegRNAs (e.g., in the first column pegRNA A1_a and pegRNA B2_a with templates for installing attP site in the forward direction). The efficiency was measured by HTS. This data showed that dual-flap editing can successfully insert the sequence of interest to the IDS locus with an efficiency up to ˜80% (FIG. 99).


Dual-flap-mediated duplication was also performed at the AAVS1 locus in 293T cells (FIG. 100A). Dual-flap pegRNA with PE2 was used to induce duplication of genetic sequences at AAVS1 (FIG. 100B).


Multi-flap prime editing induced translocation of MYC-CCR5 was also performed (FIG. 101). MYC-CCR5 translocation was induced by quad-flap pegRNAs and PE2. MYC-CCR5 translocation events were induced by quadruple-pegRNAs. Four different sets of pegRNAs were tested in HEK293T cells. The translocation junction products between derived chr8 and chr3 were amplified by junction primers. The percent of reads aligned to the expected junction alleles are shown in the graph. This shows that quadruple-flap prime editing can mediate translocation of MYC and CCR5 gene with product purity near 100% at junction 1 and ˜50% at junction 2. A representative allele plot shows the sequences aligned to the expected allele sequences at junction 1.


Dual-flap and quadruple-flap prime editing were also performed in other human cell lines (FIGS. 102A-102B). Dual-flap prime editing was performed in four different human cell lines (FIG. 102A). HEK293T and HeLa cells were transfected with dual pegRNAs and PE2 for editing three different genomic loci (IDS, MYC, and TIMM44). U2OS and K562 cells were nucleofected with the same components. Dual-flap prime editing showed robust editing efficiency across all four human cells at the targeted loci, particularly at HEK293T and K562 cells. The cellular mechanisms for enabling dual-flap prime editing are conserved across many human cell types. Multi-flap (quadruple-pegRNA) directed inversion of a 2.7 kb sequence was also performed at the AAVS1 locus in HeLa cells (FIG. 102B).


Inversion efficiency was also measured by HTS at the CCR5 locus (FIG. 103A-103B). Percentage of the expected inversion edit allele was measured by HTS. Four quad-pegRNA sets of PE2 were transfected in HEK293T cells, respectively. Dual-flap mediated sequence duplication (˜100 nt) at the CCR5 locus in HEK293T cells was performed (FIG. 103A). The editing efficiency achieves ˜1.5% via HTS 300-cycle pair-end sequencing analysis. Quadruple-flap-mediated sequence inversion (˜95-117 nt) was also performed at the CCR5 locus in HeLa cells (FIG. 103B). The editing efficiency achieves ˜1.2% via HTS 300-cycle pair-end sequencing analysis. This result shows that multi-flap prime editing can successfully mediate duplication and inversion at the CCR5 locus precisely. The editing specificity is high when the targeted sequence is duplicated (percentage of indels <2%).


Next, HEK293T cells were transfected with the plasmids that express Exo1, Fen1, Red Fluorescence Protein (control), DNA2, Mlh1 neg, and P53 inhibitor with pegRNA and PE2 (FIGS. 104A-104D). The editing efficiency was measured by HTS. The editing efficiency was compared between the candidate and the RFP control. HEK293T cells were also transfected with the siRNA plasmids and pegRNA and PE2 for each target locus (FIG. 104E). Non-targeting siRNA (siNT) was used as the control. The editing efficiency was measured by HTS. The editing efficiency was compared between each siRNA knockdown and the siNT ctrl at each target locus. HEK293T cells were transfected with dual-pegRNAs, PE2, and the plasmids that express Exo1, Fen1, Red Fluorescence Protein (ctrl), DNA2, Mlh1 neg, and P53 inhibitor, respectively. The editing efficiency was measured by HTS. The editing efficiency was compared between the candidate and the RFP control. It was found that overexpression of FEN1 improves dual flap editing efficiency in all four targeted loci (MYC, TIMM44, IDS, CCR5).


Dual-flap-mediated sequence duplication was also performed at the AAVS1 locus (FIG. 105A). By using dual pegRNAs that generate two unique 3′ flap structures, a ˜300 bp sequence duplication was induced at AAVS1 locus in 293T cells (FIG. 105B). Expected alleles were amplified with specific primers and subjected for HTS. Approximately 94% of reads were aligned to the expected alleles with duplication. Duplication products were not observed in the untreated samples.


Targeted IDS genomic sequence inversion was also performed with quadruple-flap prime editing (FIG. 106). Approximately 13% of Hunter syndrome patients have been shown to have an inversion of the IDS gene sequences (Bondeson et al., Human Molecular Genetics, 1995). Quadruple-flap prime editing was applied to induce this pathogenic inversion of the approximately 40 kb IDS genomic sequence in the HEK293T cells. Six sets of quadruple pegRNAs were tested by transfecting HEK293T cells with the pegRNAs and PE2. Primers were used to specifically amplify the inverted sequences at junction “ab” and junction “cd”. Approximately 95% of the expected inverted allele sequences were observed at both junctions with IDS_QF1. Other sets of pegRNAs also yield high percentage of the expected allele sequences at both junctions. Inverted junction products are not observed in the untreated samples.


It was also found that PegRNA 3′ motif modification improves dual-flap editing efficiency at the IDS locus (FIG. 107). To further improve the dual flap editing efficiency, a pseudoknot evoPreQ1 motif was introduced to protect the pegRNA 3′ end. In comparing the editing efficiency generated by the unmodified and evoPreQ1-modified dual pegRNAs, there was an overall increase of the editing efficiency with modified pegRNAs at the targeted IDS locus. The improvement of dual-flap editing efficiency can reach up to 5.3-fold.


REFERENCES (EXAMPLES 7 AND 8)



  • 1. Rouet, P., Smih, F. & Jasin, M. Expression of a site-specific endonuclease stimulates homologous recombination in mammalian cells. PNAS 91, 6064-6068 (1994).

  • 2. Heyer, W.-D., Ehmsen, K. T. & Liu, J. Regulation of homologous recombination in eukaryotes. Annu. Rev. Genet. 44, 113-139 (2010).

  • 3. Chapman, J. R., Taylor, M. R. G. & Boulton, S. J. Playing the end game: DNA double-strand break repair pathway choice. Mol. Cell 47, 497-510 (2012).

  • 4. Cox, D. B. T., Platt, R. J. & Zhang, F. Therapeutic genome editing: prospects and challenges. Nature Medicine 21, 121-131 (2015).

  • 5. Rouet, P., Smih, F. & Jasin, M. Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol Cell Biol 14, 8096-8106 (1994).

  • 6. Bibikova, M., Golic, M., Golic, K. G. & Carroll, D. Targeted chromosomal cleavage and mutagenesis in Drosophila using zinc-finger nucleases. Genetics 161, 1169-1175 (2002).

  • 7. Urnov, F. D., Rebar, E. J., Holmes, M. C., Zhang, H. S. & Gregory, P. D. Genome editing with engineered zinc finger nucleases. Nat Rev Genet 11, 636-646 (2010).

  • 8. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823-826 (2013).

  • 9. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819-823 (2013).

  • 10. Jinek, M. et al. RNA-programmed genome editing in human cells. eLife 2, e00471 (2013).

  • 11. Kosicki, M., Tomberg, K. & Bradley, A. Repair of double-strand breaks induced by CRISPR-Cas9 leads to large deletions and complex rearrangements. Nature Biotechnology 36, 765-771 (2018).

  • 12. Ihry, R. J. et al. p53 inhibits CRISPR-Cas9 engineering in human pluripotent stem cells. Nature Medicine 24, 939-946 (2018).

  • 13. Haapaniemi, E., Botla, S., Persson, J., Schmierer, B. & Taipale, J. CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response. Nature Medicine 24, 927-930 (2018).

  • 14. Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 576, 149-157 (2019).



Example 9—Programmable Large DNA Deletion, Replacement, Integration, and Inversion with Twin Prime Editing and Site-Specific Recombinases

The targeted deletion, replacement, integration, or inversion of DNA sequences at specified locations in the genome can in principle be used to study or treat many human genetic diseases. However, it remains challenging to precisely and efficiently carry out these DNA sequence transformations in the context of a mammalian cell genome. Presented herein is twin prime editing (twinPE), a method for the programmable replacement or excision of DNA sequence at endogenous human genomic sites without requiring double-strand DNA breaks. twinPE employs a prime editor (PE) protein and two prime editing guide RNAs (pegRNAs) that template the synthesis of complementary DNA flaps on opposing strands of genomic DNA, resulting in the replacement of endogenous DNA sequence between the PE-induced nick sites with pegRNA-encoded sequences. It is shown herein that twinPE can perform precise deletions of up to 780 bp and precise replacements of genomic DNA sequence with new sequences of up to 108 bp. By combining single or multiplexed twinPE with site-specific serine recombinases, it is demonstrated that targeted integration of gene-sized DNA plasmids at safe-harbor loci including AAVS1, CCR5, and ALB, and further, a 39-kb inversion at IDS that could be used to correct a common Hunter syndrome allele is also demonstrated. TwinPE substantially expands genome editing capabilities without requiring double-strand DNA breaks and, in combination with other genome editing tools, enables the correction or complementation of complex pathogenic allele variants in human cells.


Disease-associated human genetic variants arise through a variety of sequence changes, ranging from single-base pair substitutions to mega base-scale deletions and rearrangements. Genome editing approaches that can install, correct, or complement these pathogenic variants in human cells have the potential to advance understanding of human genetic disease and could lead to the development of novel therapeutics. Recently, several genome editing approaches based on CRISPR-Cas systems have been developed for genome editing, including nucleases, base editors, and prime editors. CRISPR-Cas nucleases, when used with one guide RNA, can be used to disrupt genes through the generation of double-strand DNA breaks (DSBs) that lead to stochastic indels. Base editors can be used to precisely install C•G-to-T•A or A•T-to-G•C transition base pair edits, or C•G-to-G•C base pair edits. Prime editors can precisely install of any one of the twelve base pair substitution edits as well as small to medium-sized insertions and deletions. However, these strategies alone are not directly applicable for making more substantial genome edits, such as large deletions, insertions, replacements, or inversions.


Previously, paired Cas9 nuclease strategies have been developed for the targeted deletion of genomic DNA sequences ranging from ˜50 to >100,000 base pairs in length. Moreover, by providing a linear donor DNA sequence, targeted insertion of new DNA sequences can be performed at single cut sites or between paired cut sites through end-joining or homology-directed DNA repair processes. While versatile in many ways, single-nuclease and paired-nuclease approaches are accompanied by several drawbacks. First, the use of paired nucleases for deletions can generate multiple on-target byproducts, including the desired deletion with indels, indels at individual DSB sites without the desired deletion, undesired inversion of the DNA sequence between DSB sites, and unintended integration of exogenous DNA sequence at DSB sites. Furthermore, the precise location of the deletions is restricted by PAM availability and the corresponding cut sites generated by the nuclease. Similar restrictions and byproducts exist for DNA donor knock-in, which often occurs without control of sequence orientation when homology-independent approaches are used. Finally, the simultaneous generation of multiple DSBs at on-target and off-target sites, which becomes more likely with additional guide RNAs, can lead to off-target genome modification, chromosomal rearrangements, or chromothripsis. Therefore, considerable challenges exist relating to product control and purity when applying nuclease-based editing strategies for the excision, replacement, or insertion of larger DNA sequences.


Prime editing has been shown to be capable of making precise insertions of up to ˜40 bp and deletions of up to ˜80 bp in human cells with high desired product to byproduct ratios. In principle, prime editing could be used to circumvent many of the challenges associated with nucleases by avoiding the generation of DSBs. However, prime editing has not yet been shown to mediate larger insertions and deletions of the size of typical gene coding sequences, and the presumed mechanism of simple prime editing reactions makes these larger DNA changes difficult by requiring long pegRNA reverse transcription templates. Site-specific DNA recombinase enzymes have the ability to perform several DNA transformations, including excision, inversion, integration, or exchange of large DNA sequences. However, challenges with reprogramming site-specific recombinases currently makes it challenging to apply for genome engineering and editing applications.


Presented herein is the development of twin prime editing (twinPE), which enables the deletion, substitution, or insertion of DNA sequence at endogenous genomic sites with high efficiencies in human cells. In addition to these changes, it is shown that twinPE can be used to integrate one or more recombinase recognition sites with high efficiency at targeted sites in the human genome, which can subsequently be used as substrates for site-specific serine recombinase enzymes that enable the targeted integration of genetic cargo or the targeted inversion of genomic DNA sequence.


Results


Prime editing uses a prime editor protein comprising a fusion of a catalytically impaired Cas9 nickase and a wild-type (PE1) or engineered (PE2) MMLV reverse transcriptase enzyme, and a prime editing guide RNA (pegRNA) that both specifies the target genomic site and encodes the desired edit. Upon target site recognition, PE•pegRNA complexes nick the PAM-containing DNA strand and directly reverse transcribe the pegRNA's RT template into genomic DNA using the nicked strand as a primer. Following reverse transcription, the newly synthesized 3′ flap invades the adjacent DNA, to which it is largely homologous, to replace the redundant 5′ flap sequence. The opposing nonedited strand is then repaired using the edited DNA strand as a template. This proposed editing pathway therefore presents two opportunities at which cellular DNA repair can reject the desired edit and revert the DNA sequence to its original form.


It was predicted that bypassing potentially disfavored steps in DNA repair could allow prime editing to occur with increased efficiency and enable distinct classes of genome edits. Furthermore, a twin prime editing (twinPE) strategy that uses a pair of pegRNAs, each of which targets one or the other DNA strand and templates the synthesis of a 3′ flap DNA sequence that is complementary to the sequence templated by the other pegRNA was envisioned (FIG. 108A). It was hypothesized that if the newly synthesized DNA strands were highly dissimilar to the endogenous target site, the 3′ flaps would preferentially hybridize to create an intermediate possessing annealed 3′ overhangs of new DNA sequence and annealed 5′ overhangs of original DNA sequence (FIG. 108A). As both edited strands are synthesized by prime editor complexes, there is no requirement for invasion of the target site by edit-containing flap strands, or for the edit to be copied into the complementary DNA strand. Excision of the original DNA sequence (annealed 5′ overhangs), filling in of gaps by polymerases, and ligation of the pair of nicks would result in the replacement of the endogenous sequence between the nick sites with the paired 3′ flap sequences (FIG. 108A). Due to the flexibility in template design, the edit could in principle introduce a new DNA sequence, replace a portion of DNA sequence, or delete a portion of DNA sequence.


To evaluate the twinPE strategy, the HEK site 3 locus (previously referred to as HEK3) in HEK293T was targeted to replace 90 bp of endogenous sequence with a 38-bp Bxb1 attB attachment sequence (FIG. 108B). For each protospacer, three pegRNAs were designed with RT templates that contained 30, 34, or 38 nt of the 38-bp attB sequence (FIG. 108C). Pairwise combinations of these pegRNA are predicted to generate 3′ flaps with overlapping complementarity ranging from 22 to 38 bp (FIG. 108C). Excitingly, when both pegRNAs were delivered to cells along with PE2 in a plasmid DNA transfection, high efficiency of attB site insertion was observed, with some combinations of pegRNAs yielding >80% conversion to the desired product based on amplicon sequencing (FIG. 108C). A similar strategy for the insertion of the 50-bp Bxb1 attP attachment sequence achieved editing with efficiencies reaching up to 58% (FIG. 108C). Notably, it was not necessary for each pegRNA to encode the full insertion sequence, since partially overlapping complementary flaps enabled full-length attB or attP sequence incorporation. However, 3′ flaps with greater overlap led to slightly higher editing efficiencies and fewer indels for insertion of the Bxb1 attB and attP sequences at HEK site 3.


Next, twinPE was tested to determine if it could support the insertion of DNA sequences larger than that which has been demonstrated using the PE2 or PE3 systems. First, PE3-mediated insertions of FKBP12 coding sequence fragments ranging from 12 to 321 bp were examined Targeting the HEK site 3 locus with PE3, modest efficiencies were achieved for the shorter 12-bp and 36-bp insertions (31.8% and 17.3%, respectively), but very little desired product was observed for the 108-bp insertion (0.8%) and no full-length product for the 321-bp insertion (FIG. 109A). By contrast, twinPE enabled 16% insertion efficiency for the 108-bp fragment, amounting to a 20-fold improvement over PE3. Furthermore, 113-bp and 103-bp insertions containing paired Bxb1 recombinase sites were achieved with similar efficiencies (10.7% and 9.7%, respectively) at the CCR5 locus (data not shown). These results illustrate the potential for larger sequence insertion using twinPE compared to PE3.


One potential application of the twinPE strategy is the replacement of exonic coding sequence with recoded DNA sequence that maintains the protein sequence and has the potential to correct any mutation between target twinPE-induced nick sites. To test this approach, the PAH gene was targeted, mutations in which cause the genetic metabolic disorder phenylketonuria (PKU). Recoding of portions of exon 4 and exon 7 was tested in PAH of wild type HEK293T cells, where corrective editing is detectable. By testing different flap overlap lengths and with the addition of evoPreQ1 motifs to the 3′ end of the pegRNAs, the desired sequence recoding was achieved with modest efficiencies reaching up to 9.4% average efficiency for a 64-bp recoding in exon 4, up to 22.7% average efficiency for a 46-bp recoding in exon 7, and up to 27.4% average efficiency for a 64-bp recoding in exon 7 (FIG. 109B). Based on mutations annotated in bioPKU database, twinPE editing of these regions collectively allow for the correction of distinct mutations implicated in PKU. Additional exons examined could also be recoded, albeit with lower efficiency (FIG. 112). These results demonstrate that twinPE could in principle be used to correct multiple mutations with a single pair of pegRNAs.


In addition to insertion and replacement of DNA sequences, twinPE should also be able to make precise deletion edits. Paired-nuclease deletion strategies generate deletions that span the two DSB sites and are thus restricted by PAM availability, and desired deletions are often accompanied by undesired indel byproducts. By contrast, twinPE has the potential to make deletions with greater flexibility and precision due to the lack of DSB generation and the ability to write in additional DNA sequence at the twinPE-induced nick sites. To assess twinPE for precise deletions, three strategies using paired pegRNAs were compared: a “single-anchor” twinPE strategy, a “hybrid-anchor” twinPE strategy, and the recently reported “PrimeDel” strategy (FIG. 109C). Each strategy differs in the sequence that is encoded in the complementary flaps, allowing for flexibility in the positioning of the deletion with respect to the nick sites.


Using the single-anchor strategy, 13-nt complementary flaps were used to delete 77 bp of sequence adjacent to one of the pegRNA-induced nick sites with 14.8% efficiency, and 34-nt complementary flaps were used to precisely excise 56 bp of sequence with 18.8% efficiency (FIG. 109D). Using the hybrid-anchor strategy, 64 bp were deleted between the pegRNA-induced nick sites such that the product retains 13 bp of sequence 3′ of each nick, with 11.7% efficiency (FIG. 109D). Finally, the PrimeDel strategy was tested for the deletion of 90 bp between the pegRNA-induced nick sites, which occurred with 40.4% efficiency (FIG. 109D). Of note, the PrimeDel strategy disrupts the PAM sequences on both strands, which likely increases efficiency while also decreasing indels. Editing efficiencies could be improved 1.5-fold to 2.5-fold by the addition of the evoPreQ1 motif the 3′ end of the pegRNAs, albeit with accompanied increases in indels in some cases (FIG. 109D). Together, these data show that twinPE offers a strategy for performing targeted deletions with high flexibility that does not rely on availability of perfectly positioned nuclease cut sites.


Lastly, a therapeutically relevant locus, DMD, was targeted to apply twinPE for larger deletions. Pathogenic DMD alleles, which are responsible for Duchenne muscular dystrophy, commonly contain large deletions in regions containing exons that result in frameshifted mRNA transcripts. Because production of full-length dystrophin protein without replacement of deleted exons can lead to partial rescue of protein function, disruption of a second exon that restores the reading frame has been proposed as a potential therapeutic strategy. Three twinPE deletion strategies were examined along with a previously reported Cas9 nuclease deletion strategy for excising exon 51 in DMD. Using single-anchor twinPE deletion approaches, 14.4% to 24% efficiency was observed for the deletion of a 780-bp sequence containing exon 51 in DMD (FIG. 109E). While the paired Cas9 nuclease strategy achieved higher deletion efficiency (averaging 49.8%) compared to the twinPE strategies (averaging 14% to 24%), paired Cas9 nuclease-mediated deletion was also accompanied by much higher indel levels (33% desired deletion with indels, 11% indels without the desired deletion) compared to twinPE (1% to 12% total indels) (FIG. 109E). Exon 51 of DMD could also be excised using alternative spacer pairs that generate a 627-bp deletion with the PrimeDel strategy (28.8% average efficiency) or a 590-bp deletion with the single-anchor twinPE strategy (26.4% average efficiency), or by using twinPE to replace a 589-bp sequence with a 38-bp Bxb1 attB sequence (up to 40.4% average efficiency) (FIG. 109E). These experiments show that twinPE and PrimeDel are capable of generating large deletions at therapeutically relevant loci in human cells with superior product purity compared to paired Cas9 nuclease strategies, albeit with lower average efficiencies.


Targeted DNA Integration at Safe Harbor Loci with TwinPE and Bxb1 Integrase


Although twinPE is able to perform larger insertion edits than PE3, the upper limits of sequence insertion size were still not sufficient to support the integration of gene-sized DNA fragments. Therefore, having successfully inserted Bxb1 attachment sequences into endogenous human genomic sites with high efficiency using twinPE, twinPE was combined with serine recombinases for the site-specific integration of DNA cargo. To identify locations for DNA cargo integration, the twinPE-mediated insertion of Bxb1 attB and attP attachment sequences at established safe harbor loci in HEK293T cells. 32 spacer pairs targeting the AAVS1 locus were screened for insertion of the 50-bp Bxb1 attP sequence (data not shown). Of the evaluated spacer pairs, optimal pegRNAs for 18 out of 32 spacer combinations achieved >50% correct insertion efficiency by amplicon sequencing (FIG. 110A). Additionally, 19 spacer pairs targeting the CCR5 locus were screened for insertion of the 38-bp Bxb1 attB sequence (data not shown), of which 6 spacer pairs achieved >50% desired editing efficiency by amplicon sequencing (FIG. 110B). These results demonstrate that twinPE can be used to insert recombinase attachment sequences at safe harbor loci in human cells with high efficiency.


Next, twinPE-incorporated Bxb1 attB and attP sequences were examined to determine if they could serve as target sites for the integration of plasmid DNA cargo harboring partner Bxb1 attachment sites. First, twinPE was used to generate single-cell clones bearing homozygous attB site insertions at the CCR5 locus (data not shown). Transfection of this clonal cell line with a plasmid expressing Bxb1 recombinase and a 5.6-kB attP-containing donor DNA plasmid yielded 12-17% knock-in at the target site as measured by ddPCR (FIG. 113), consistent with previously reported Bxb1 integration efficiencies. Encouraged by these results, twinPE-mediated attachment site insertion and BxB1-mediated DNA donor integration were explored to determine if they could be achieved in a single transfection step. Excitingly, transfection of HEK293T cells with plasmids encoding PE2, both pegRNAs, BxB1, and donor DNA resulted in 1.4-6.8% knock-in efficiency as measured by ddPCR (FIG. 110C). The expected junction sequences containing the expected attL and attR recombination products were confirmed by amplicon sequencing, with product purities ranging from 71-95% (FIG. 114).


In an effort to improve the “one-pot” knock-in efficiency, the twinPE-mediated incorporation of either attB or attP attachment sites, the canonical (GT) Bxb1 core sequence, an alternative (GA) Bxb1 core sequence, and varying flap overlap lengths were tested. It was found that the incorporation of the attB site with twinPE performed better than the incorporation of the attP site for one-pot knock-in transfections (3.3% vs. 0.5% knock-in, FIG. 110D). In addition, it was observed that insertions of attachment sites with the canonical GT core sequence generally led to higher knock-in efficiencies than insertions of attachment sites with the alternative GA sequence (3.3% vs. 0% in the attB context, FIG. 110D). Of note, however, the canonical and alternative core sequences allow for orthogonal recombination with corresponding attP attachment sites. Reducing overlap between pegRNA-encoded recombinase sequences also improved knock-in efficiency, from 3.3% with 38 base pairs of overlap to 5.5% with 20 base pairs of overlap when inserting attB (FIG. 110D), possibly due to the reduced extent of attachment sequence within the pegRNA-expressing plasmid DNA sequence. Although similar twinPE efficiency was observed across overlap lengths, recombination between donor DNA and pegRNA plasmid was reduced (FIGS. 115A-115B).


Next, donor integration was evaluated at the ALB locus, which can be coopted for therapeutic protein secretion from hepatocytes. Albumin is actively secreted by the liver and constitutes approximately 60% of human plasma proteins. Therefore, therapeutic transgene integration at the ALB locus in a relatively small percentage of hepatocytes could achieve therapeutic levels of protein expression for many diseases. A strategy targeting intron 1 of ALB was devised, wherein Bxb1 would mediate the integration of circular DNA containing a splice acceptor sequence followed by the cDNA for a protein of interest. Integration would in principle allow for splicing of the albumin secretion signal, encoded in exon 1, to the therapeutic transgene coding sequence. First, pegRNA pairs were screened for the insertion of the Bxb1 attB sequence within intron 1 of ALB. After screening pegRNA pairs, a spacer combination that achieved 43% correct insertion of the attB sequence by amplicon sequencing was identified (FIG. 110E). Single transfection knock-in at this locus was achieved with 1.3% efficiency in HEK293T (FIG. 110F). In Huh7 cells, 34% correct insertion of the attB sequence, with single transfection knock-in efficiency of 2.6% was observed. Interestingly, knock-in at ALB in Huh7 was more efficient than knock-in at CCR5 (2.6% vs. 1.1%), despite CCR5 knock-in being more efficient than ALB knock-in in HEK293T cells.









TABLE 14







DNA donor sequences used in this Example











SEQ




ID


DNA donor
Sequence
NO:





attB_
gatgccagctcattcctcccactcatgatctatagatcccccgg
4282


Puro_GA
gctgcaggaattctacccactctgtcgataccccaccgagaccc



donor
cattggggccaatacgcccgcgtttcttccttttccccacccca



DNA:
ccccccaagttcgggtgaaggcccagggctcgcagccaacgtc




ggggcggcaagcttacatcgagatcccggcttgtcgacgacgg




cggactccgtcgtcaggatcatccgtgagcaagggcgagga




gctgttcaccggggtggtgcccatcctggtcgagctggacgg




cgacgtaaacggccacaagttcagcgtgtccggcgagggcga




gggcgatgccacctacggcaagctgaccctgaagttcatctgc




accaccggcaagctgcccgtgccctggcccaccctcgtgacc




accctgacctacggcgtgcagtgcttcagccgctaccccgac




cacatgaagcagcacgacttcttcaagtccgccatgcccgaag




gctacgtccaggagcgcaccatcttcttcaaggacgacggca




actacaagacccgcgccgaggtgaagttcgagggcgacaccc




tggtgaaccgcatcgagctgaagggcatcgacttcaaggagg




acggcaacatcctggggcacaagctggagtacaactacaaca




gccacaacgtctatatcatggccgacaagcagaagaacggcat




caaggtgaacttcaagatccgccacaacatcgaggacggca




gcgtgcagctcgccgaccactaccagcagaacacccccatcgg




cgacggccccgtgctgctgcccgacaaccactacctgagca




cccagtccgccctgagcaaagaccccaacgagaagcgcgatc




acatggtcctgctggagttcgtgaccgccgccgggatcactct




cggcatggacgagctgtacaagagcggcctgaggagcagag




cccaggcgagcaacagcgccgtggacgccaccatgggcga




tcgcccgggaattgactagtgcggccgcctaggatccatgccg




atagcgttggttgagtggataaccgtattaccgccaagcttatg




catgtgcccgtcagtgggcagagcgcacatcgcccacagtcc




ccgagaagttggggggaggggtcggcaattgaaccggtgcct




agagaaggtggcgcggggtaaactgggaaagtgatgtcgtgta




ctggctccgcctttttcccgagggtgggggagaaccgtatata




agtgcagtagtcgccgtgaacgttctttttcgcaacgggtttg




ccgccagaacacaggtaagtgccgtgtgtggttcccgcgggcct




ggcctctttacgggttatggcccttgcgtgccttgaattacttcc




acctggctgcagtacgtgattcttgatcccgagcttcgggttgga




agtgggtgggagagttcgaggccttgcgcttaaggagcccctt




cgcctcgtgcttgagttgaggcctggcctgggcgctggggcc




gccgcgtgcgaatctggtggcaccttcgcgcctgtctcgctgctt




tcgataagtctctagccatttaaaatttttgatgacctgctgcga




cgctttttttctggcaagatagtcttgtaaatgcgggccaagatc




tgcacactggtatttcggtttttggggccgcgggcggcgacggg




gcccgtgcgtcccagcgcacatgttcggcgaggcggggcct




gcgagcgcggccaccgagaatcggacgggggtagtctcaag




ctggccggcctgctctggtgcctggcctcgcgccgccgtgtat




cgccccgccctgggcggcaaggctggcccggtcggcaccag




ttgcgtgagcggaaagatggccgcttcccggccctgctgca




gggagctcaaaatggaggacgcggcgctcgggagagcgggc




gggtgagtcacccacacaaaggaaaagggcctttccgtcctca




gccgtcgcttcatgtgactccacggagtaccgggcgccgtcca




ggcacctcgattagttctcgcgcttttggagtacgtcgtctttag




gttggggggaggggttttatgcgatggagtttccccacactgagt




gggtggagactgaagttaggccagcttggcacttgatgtaattct




ccttggaatttgccctttttgagtttggatcttggttcattctca




agcctcagacagtggttcaaagtttttttcttccatttcaggtgt




cgtgagctagcccaccatgaccgagtacaagcccacggtgcgcct




cgccacccgcgacgacgtcccccgggccgtacgcaccctcgc




cgccgcgttcgccgactaccccgccacgcgccacaccgtcg




acccggaccgccacatcgagcgggtcaccgagctgcaagaac




tcttcctcacgcgcgtcgggctcgacatcggcaaggtgtggg




tcgcggacgacggcgccgcggtggcggtctggaccacgccg




gagagcgtcgaagcgggggcggtgttcgccgagatcggcc




cgcgcatggccgagttgagcggttcccggctggccgcgcag




caacagatggaaggcctcctggcgccgcaccggcccaagga




gcccgcgtggttcctggccaccgtcggcgtctcgcccgacc




accagggcaagggtctgggcagcgccgtcgtgctccccggagt




ggaggcggccgagcgcgccggggtgcccgccttcctggaga




cctccgcgccccgcaacctccccttctacgagcggctcggctt




caccgtcaccgccgacgtcgaggtgcccgaaggaccgcgcacc




tggtgcatgacccgcaagcccggtgccggatcgggagag




ggcagaggaagtctgctaacatgcggtgacgtcgaggagaa




tcctggcccaccggtcgccaccagcgagctgattaaggagaac




atgcacatgaagctgtacatggagggcaccgtggacaaccat




cacttcaagtgcacatccgagggcgaaggcaagccctacgag




ggcacccagaccatgagaatcaaggtggtcgagggcggccctc




tccccttcgccttcgacatcctggctactagcttcctctacggc




agcaagaccttcatcaaccacacccagggcatccccgacttct




tcaagcagtccttccctgagggcttcacatgggagagagtcac




cacatacgaagacgggggcgtgctgaccgctacccaggacac




cagcctccaggacggctgcctcatctacaacgtcaagatcag




aggggtgaacttcacatccaacggccctgtgatgcagaagaa




aacactcggctgggaggccttcaccgagacgctgtaccccgct




gacggcggcctggaaggcagaaacgacatggccctgaagctc




gtgggcgggagccatctgatcgcaaacatcaagaccacatat




agatccaagaaacccgctaagaacctcaagatgcctggcgtct




actatgtggactacagactggaaagaatcaaggaggccaaca




acgagacctacgtcgagcagcacgaggtggcagtggccagatac




tgcgacctccctagcaaactggggcacaagcttaattaagaatt




ctctagaggatccagacatgataagatacattgatgagtttggac




aaaccacaactagaatgcagtgaaaaaaatgctttatttgtgaaa




tttgtgatgctattgctttatttgtaaccattataagctgcaata




aacaagttaacaacaacaattgcattcattttatgtttcaggttc




agggggaggtgtgggaggttttttaaagcaagtaaaacctctaca




aatgtggtatggctgattatgatcctgcaagcctcgtcgccgcgg




tttattctgttgacaattaatcatcggcatagtatatcggcatag




tataatacgacaaggtgaggaactaaaccatgggatcggccattg




aacaagatggattgcacgcaggttctccggccgcttgggtgga




gaggctattcggctatgactgggcacaacagacgatcggctg




ctctgatgccgccgtgttccggctgtcagcgcaggggcgcccg




gttctttttgtcaagaccgacctgtccggtgccctgaatgaact




gcaggacgaggcagcgcggctatcgtggctggccacgacggg




cgttccttgcgcagctgtgctcgacgttgtcactgaagcggg




aagggactggctgctattgggcgaagtgccggggcaggatctcc




tgtcatctcaccttgctcctgccgagaaagtatccatcatggc




tgatgcaatgcggcggctgcatacgcttgatccggctacctgc




ccattcgaccaccaagcgaaacatcgcatcgagcgagcacgta




ctcggatggaagccggtcttgtcgatcaggatgatctggacga




agagcatcaggggctcgcgccagccgaactgttcgccaggct




caaggcgcgcatgcccgacggcgaggatctcgtcgtgaccca




tggcgatgcctgcttgccgaatatcatggtggaaaatggccgc




ttttctggattcatcgactgtggccggctgggtgtggcggaccg




ctatcaggacatagcgttggctacccgtgatattgctgaagagc




ttggcggcgaatgggctgaccgcttcctcgtgctttacggtatcg




ccgcccccgattcgcagcgcatcgccttctatcgccttcttgac




gagttcttctgagcgggactctggggttcgaataaagaccgacc




aagcgacgtctgagagctccctggcgaattcggtaccaataaa




agagctttattttcatgatctgtgtgttggtttttggccgcgttg




ctggcgtttttccataggctccgcccccctgacgagcatcacaaa




aatcgacgctcaagtcagaggtggcgaaacccgacaggactat




aaagataccaggcgtttccccctggaagctccctcgtgcgctc




tcctgttccgaccctgccgcttaccggatacctgtccgcctttct




cccttcgggaagcgtggcgctttctcatagctcacgctgtaggta




tctcagttcggtgtaggtcgttcgctccaagctgggctgtgtgc




acgaaccccccgttcagcccgaccgctgcgccttatccggtaac




tatcgtcttgagtccaacccggtaagacacgacttatcgccact




ggcagcagccactggtaacaggattagcagagcgaggtatgta




ggcggtgctacagagttcttgaagtggtggcctaactacggct




acactagaagaacagtatttggtatctgcgctctgctgaagccag




ttaccttcggaaaaagagttggtagctcttgatccggcaaacaaa




ccaccgctggtagcggtggtttttttgtttgcaagcagcagatt




acgcgcagaaaaaaaggatctcaagaagatcctttgatctttt




ctagtgtgcg






attB_Puro
gatgccagctcattcctcccactcatgatctatagatcccccgg
4283


donor DNA:
gctgcaggaattctacccactctgtcgataccccaccgagaccc




cattggggccaatacgcccgcgtttcttccttttccccacccc




accccccaagttcgggtgaaggcccagggctcgcagccaacgtc




ggggcggcaagcttacatcgagatcccggcttgtcgacgacg




gcggtctccgtcgtcaggatcatccgtgagcaagggcgagga




gctgttcaccggggtggtgcccatcctggtcgagctggacggc




gacgtaaacggccacaagttcagcgtgtccggcgagggcga




gggcgatgccacctacggcaagctgaccctgaagttcatctg




caccaccggcaagctgcccgtgccctggcccaccctcgtgacc




accctgacctacggcgtgcagtgcttcagccgctaccccgacc




acatgaagcagcacgacttcttcaagtccgccatgcccgaag




gctacgtccaggagcgcaccatcttcttcaaggacgacggca




actacaagacccgcgccgaggtgaagttcgagggcgacaccc




tggtgaaccgcatcgagctgaagggcatcgacttcaaggag




gacggcaacatcctggggcacaagctggagtacaactacaaca




gccacaacgtctatatcatggccgacaagcagaagaacggca




tcaaggtgaacttcaagatccgccacaacatcgaggacggca




gcgtgcagctcgccgaccactaccagcagaacacccccatcg




gcgacggccccgtgctgctgcccgacaaccactacctgagca




cccagtccgccctgagcaaagaccccaacgagaagcgcgatc




acatggtcctgctggagttcgtgaccgccgccgggatcactct




cggcatggacgagctgtacaagagcggcctgaggagcaga




gcccaggcgagcaacagcgccgtggacgccaccatgggcga




tcgcccgggaattgactagtgcggccgcctaggatccatgccg




atagcgttggttgagtggataaccgtattaccgccaagcttatg




catgtgcccgtcagtgggcagagcgcacatcgcccacagtcc




ccgagaagttggggggaggggtcggcaattgaaccggtgcct




agagaaggtggcgcggggtaaactgggaaagtgatgtcgtgta




ctggctccgcctttttcccgagggtgggggagaaccgtatata




agtgcagtagtcgccgtgaacgttctttttcgcaacgggtttg




ccgccagaacacaggtaagtgccgtgtgtggttcccgcgggcct




ggcctctttacgggttatggcccttgcgtgccttgaattacttcc




acctggctgcagtacgtgattcttgatcccgagcttcgggttgga




agtgggtgggagagttcgaggccttgcgcttaaggagcccctt




cgcctcgtgcttgagttgaggcctggcctgggcgctggggcc




gccgcgtgcgaatctggtggcaccttcgcgcctgtctcgctgctt




tcgataagtctctagccatttaaaatttttgatgacctgctgcga




cgctttttttctggcaagatagtcttgtaaatgcgggccaagat




ctgcacactggtatttcggtttttggggccgcgggcggcgacggg




gcccgtgcgtcccagcgcacatgttcggcgaggcggggcctg




cgagcgcggccaccgagaatcggacgggggtagtctcaag




ctggccggcctgctctggtgcctggcctcgcgccgccgtgta




tcgccccgccctgggcggcaaggctggcccggtcggcaccag




ttgcgtgagcggaaagatggccgcttcccggccctgctgca




gggagctcaaaatggaggacgcggcgctcgggagagcgggc




gggtgagtcacccacacaaaggaaaagggcctttccgtcctca




gccgtcgcttcatgtgactccacggagtaccgggcgccgtcc




aggcacctcgattagttctcgcgcttttggagtacgtcgtcttta




ggttggggggaggggttttatgcgatggagtttccccacactgag




tgggtggagactgaagttaggccagcttggcacttgatgtaattc




tccttggaatttgccctttttgagtttggatcttggttcattctc




aagcctcagacagtggttcaaagtttttttcttccatttcaggtg




tcgtgagctagcccaccatgaccgagtacaagcccacggtgcgcc




tcgccacccgcgacgacgtcccccgggccgtacgcaccctcgc




cgccgcgttcgccgactaccccgccacgcgccacaccgtcg




acccggaccgccacatcgagcgggtcaccgagctgcaagaac




tcttcctcacgcgcgtcgggctcgacatcggcaaggtgtggg




tcgcggacgacggcgccgcggtggcggtctggaccacgccg




gagagcgtcgaagcgggggcggtgttcgccgagatcggcc




cgcgcatggccgagttgagcggttcccggctggccgcgcagc




aacagatggaaggcctcctggcgccgcaccggcccaagga




gcccgcgtggttcctggccaccgtcggcgtctcgcccgacca




ccagggcaagggtctgggcagcgccgtcgtgctccccggagt




ggaggcggccgagcgcgccggggtgcccgccttcctggagac




ctccgcgccccgcaacctccccttctacgagcggctcggctt




caccgtcaccgccgacgtcgaggtgcccgaaggaccgcgca




cctggtgcatgacccgcaagcccggtgccggatcgggagag




ggcagaggaagtctgctaacatgcggtgacgtcgaggagaat




cctggcccaccggtcgccaccagcgagctgattaaggagaac




atgcacatgaagctgtacatggagggcaccgtggacaacca




tcacttcaagtgcacatccgagggcgaaggcaagccctacgag




ggcacccagaccatgagaatcaaggtggtcgagggcggccctc




tccccttcgccttcgacatcctggctactagcttcctctacggc




agcaagaccttcatcaaccacacccagggcatccccgacttct




tcaagcagtccttccctgagggcttcacatgggagagagtcac




cacatacgaagacgggggcgtgctgaccgctacccaggaca




ccagcctccaggacggctgcctcatctacaacgtcaagatcag




aggggtgaacttcacatccaacggccctgtgatgcagaagaa




aacactcggctgggaggccttcaccgagacgctgtaccccgct




gacggcggcctggaaggcagaaacgacatggccctgaagctc




gtgggcgggagccatctgatcgcaaacatcaagaccacatat




agatccaagaaacccgctaagaacctcaagatgcctggcgtc




tactatgtggactacagactggaaagaatcaaggaggccaaca




acgagacctacgtcgagcagcacgaggtggcagtggccagatac




tgcgacctccctagcaaactggggcacaagcttaattaagaatt




ctctagaggatccagacatgataagatacattgatgagtttgga




caaaccacaactagaatgcagtgaaaaaaatgctttatttgtgaa




atttgtgatgctattgctttatttgtaaccattataagctgcaat




aaacaagttaacaacaacaattgcattcattttatgtttcaggtt




cagggggaggtgtgggaggttttttaaagcaagtaaaacctctac




aaatgtggtatggctgattatgatcctgcaagcctcgtcgccgcg




gtttattctgttgacaattaatcatcggcatagtatatcggcata




gtataatacgacaaggtgaggaactaaaccatgggatcggccatt




gaacaagatggattgcacgcaggttctccggccgcttgggtgga




gaggctattcggctatgactgggcacaacagacgatcggctg




ctctgatgccgccgtgttccggctgtcagcgcaggggcgcccg




gttctttttgtcaagaccgacctgtccggtgccctgaatgaact




gcaggacgaggcagcgcggctatcgtggctggccacgacggg




cgttccttgcgcagctgtgctcgacgttgtcactgaagcggg




aagggactggctgctattgggcgaagtgccggggcaggatctc




ctgtcatctcaccttgctcctgccgagaaagtatccatcatggc




tgatgcaatgcggcggctgcatacgcttgatccggctacctgc




ccattcgaccaccaagcgaaacatcgcatcgagcgagcacgta




ctcggatggaagccggtcttgtcgatcaggatgatctggacg




aagagcatcaggggctcgcgccagccgaactgttcgccaggct




caaggcgcgcatgcccgacggcgaggatctcgtcgtgacccat




ggcgatgcctgcttgccgaatatcatggtggaaaatggccgc




ttttctggattcatcgactgtggccggctgggtgtggcggaccg




ctatcaggacatagcgttggctacccgtgatattgctgaagagc




ttggcggcgaatgggctgaccgcttcctcgtgctttacggtatc




gccgcccccgattcgcagcgcatcgccttctatcgccttcttgac




gagttcttctgagcgggactctggggttcgaataaagaccgac




caagcgacgtctgagagctccctggcgaattcggtaccaataaa




agagctttattttcatgatctgtgtgttggtttttggccgcgttg




ctggcgtttttccataggctccgcccccctgacgagcatcacaaa




aatcgacgctcaagtcagaggtggcgaaacccgacaggactat




aaagataccaggcgtttccccctggaagctccctcgtgcgctc




tcctgttccgaccctgccgcttaccggatacctgtccgcctttct




cccttcgggaagcgtggcgctttctcatagctcacgctgtaggta




tctcagttcggtgtaggtcgttcgctccaagctgggctgtgtgc




acgaaccccccgttcagcccgaccgctgcgccttatccggtaac




tatcgtcttgagtccaacccggtaagacacgacttatcgccact




ggcagcagccactggtaacaggattagcagagcgaggtatgta




ggcggtgctacagagttcttgaagtggtggcctaactacggcta




cactagaagaacagtatttggtatctgcgctctgctgaagccag




ttaccttcggaaaaagagttggtagctcttgatccggcaaacaa




accaccgctggtagcggtggtttttttgtttgcaagcagcagatt




acgcgcagaaaaaaaggatctcaagaagatcctttgatctttt




ctagtgtgcg






attP_
gatgccagctcattcctcccactcatgatctatagatcccccgg
4284


Puro_GA 
gctgcaggaattctacccactctgtcgataccccaccgagaccc



donor
cattggggccaatacgcccgcgtttcttccttttccccacccc



DNA:
accccccaagttcgggtgaaggcccagggctcgcagccaacgtc




ggggcggcaagcttacatcgagatcccggtttgtctggtcaa




ccaccgcggactcagtggtgtacggtacaaaccccgtgagcaa




gggcgaggagctgttcaccggggtggtgcccatcctggtcga




gctggacggcgacgtaaacggccacaagttcagcgtgtccgg




cgagggcgagggcgatgccacctacggcaagctgaccctgaa




gttcatctgcaccaccggcaagctgcccgtgccctggcccac




cctcgtgaccaccctgacctacggcgtgcagtgcttcagccgc




taccccgaccacatgaagcagcacgacttcttcaagtccgcca




tgcccgaaggctacgtccaggagcgcaccatcttcttcaagg




acgacggcaactacaagacccgcgccgaggtgaagttcgagg




gcgacaccctggtgaaccgcatcgagctgaagggcatcgac




ttcaaggaggacggcaacatcctggggcacaagctggagtac




aactacaacagccacaacgtctatatcatggccgacaagcag




aagaacggcatcaaggtgaacttcaagatccgccacaacatcg




aggacggcagcgtgcagctcgccgaccactaccagcagaac




acccccatcggcgacggccccgtgctgctgcccgacaaccac




tacctgagcacccagtccgccctgagcaaagaccccaacgag




aagcgcgatcacatggtcctgctggagttcgtgaccgccgcc




gggatcactctcggcatggacgagctgtacaagagcggcct




gaggagcagagcccaggcgagcaacagcgccgtggacgcca




ccatgggcgatcgcccgggaattgactagtgcggccgcctag




gatccatgccgatagcgttggttgagtggataaccgtattaccg




ccaagcttatgcatgtgcccgtcagtgggcagagcgcacatc




gcccacagtccccgagaagttggggggaggggtcggcaattg




aaccggtgcctagagaaggtggcgcggggtaaactgggaaag




tgatgtcgtgtactggctccgcctttttcccgagggtggggga




gaaccgtatataagtgcagtagtcgccgtgaacgttctttttcg




caacgggtttgccgccagaacacaggtaagtgccgtgtgtggtt




cccgcgggcctggcctctttacgggttatggcccttgcgtgcct




tgaattacttccacctggctgcagtacgtgattcttgatcccgag




cttcgggttggaagtgggtgggagagttcgaggccttgcgctt




aaggagccccttcgcctcgtgcttgagttgaggcctggcctgg




gcgctggggccgccgcgtgcgaatctggtggcaccttcgcgcctg




tctcgctgctttcgataagtctctagccatttaaaatttttgatg




acctgctgcgacgctttttttctggcaagatagtcttgtaaatgc




gggccaagatctgcacactggtatttcggtttttggggccgcggg




cggcgacggggcccgtgcgtcccagcgcacatgttcggcg




aggcggggcctgcgagcgcggccaccgagaatcggacgggg




gtagtctcaagctggccggcctgctctggtgcctggcctcgc




gccgccgtgtatcgccccgccctgggcggcaaggctggcccg




gtcggcaccagttgcgtgagcggaaagatggccgcttcccg




gccctgctgcagggagctcaaaatggaggacgcggcgctcgg




gagagcgggcgggtgagtcacccacacaaaggaaaagggcct




ttccgtcctcagccgtcgcttcatgtgactccacggagtaccg




ggcgccgtccaggcacctcgattagttctcgcgcttttggagtac




gtcgtctttaggttggggggaggggttttatgcgatggagtttcc




ccacactgagtgggtggagactgaagttaggccagcttggcactt




gatgtaattctccttggaatttgccctttttgagtttggatcttg




gttcattctcaagcctcagacagtggttcaaagtttttttcttcc




atttcaggtgtcgtgagctagcccaccatgaccgagtacaagccc




acggtgcgcctcgccacccgcgacgacgtcccccgggccgt




acgcaccctcgccgccgcgttcgccgactaccccgccacgcgc




cacaccgtcgacccggaccgccacatcgagcgggtcaccgag




ctgcaagaactcttcctcacgcgcgtcgggctcgacatcggc




aaggtgtgggtcgcggacgacggcgccgcggtggcggtctg




gaccacgccggagagcgtcgaagcgggggcggtgttcgcc




gagatcggcccgcgcatggccgagttgagcggttcccggctg




gccgcgcagcaacagatggaaggcctcctggcgccgcaccg




gcccaaggagcccgcgtggttcctggccaccgtcggcgtctc




gcccgaccaccagggcaagggtctgggcagcgccgtcgtgc




tccccggagtggaggcggccgagcgcgccggggtgcccgcct




tcctggagacctccgcgccccgcaacctccccttctacgag




cggctcggcttcaccgtcaccgccgacgtcgaggtgcccga




aggaccgcgcacctggtgcatgacccgcaagcccggtgccgg




atcgggagagggcagaggaagtctgctaacatgcggtgacg




tcgaggagaatcctggcccaccggtcgccaccagcgagctga




ttaaggagaacatgcacatgaagctgtacatggagggcaccg




tggacaaccatcacttcaagtgcacatccgagggcgaaggcaa




gccctacgagggcacccagaccatgagaatcaaggtggtcgag




ggcggccctctccccttcgccttcgacatcctggctactagct




tcctctacggcagcaagaccttcatcaaccacacccagggcat




ccccgacttcttcaagcagtccttccctgagggcttcacatggg




agagagtcaccacatacgaagacgggggcgtgctgaccgct




acccaggacaccagcctccaggacggctgcctcatctacaacg




tcaagatcagaggggtgaacttcacatccaacggccctgtgat




gcagaagaaaacactcggctgggaggccttcaccgagacgct




gtaccccgctgacggcggcctggaaggcagaaacgacatggc




cctgaagctcgtgggcgggagccatctgatcgcaaacatca




agaccacatatagatccaagaaacccgctaagaacctcaagat




gcctggcgtctactatgtggactacagactggaaagaatcaag




gaggccaacaacgagacctacgtcgagcagcacgaggtggcagt




ggccagatactgcgacctccctagcaaactggggcacaagctt




aattaagaattctctagaggatccagacatgataagatacattga




tgagtttggacaaaccacaactagaatgcagtgaaaaaaatgctt




tatttgtgaaatttgtgatgctattgctttatttgtaaccattat




aagctgcaataaacaagttaacaacaacaattgcattcattttat




gtttcaggttcagggggaggtgtgggaggttttttaaagcaagt




aaaacctctacaaatgtggtatggctgattatgatcctgcaagcc




tcgtcgccgcggtttattctgttgacaattaatcatcggcatag




tatatcggcatagtataatacgacaaggtgaggaactaaaccatg




ggatcggccattgaacaagatggattgcacgcaggttctccgg




ccgcttgggtggagaggctattcggctatgactgggcacaaca




gacgatcggctgctctgatgccgccgtgttccggctgtcagcg




caggggcgcccggttctttttgtcaagaccgacctgtccggtgc




cctgaatgaactgcaggacgaggcagcgcggctatcgtggct




ggccacgacgggcgttccttgcgcagctgtgctcgacgttgtc




actgaagcgggaagggactggctgctattgggcgaagtgccgg




ggcaggatctcctgtcatctcaccttgctcctgccgagaaagt




atccatcatggctgatgcaatgcggcggctgcatacgcttgat




ccggctacctgcccattcgaccaccaagcgaaacatcgcatcga




gcgagcacgtactcggatggaagccggtcttgtcgatcagga




tgatctggacgaagagcatcaggggctcgcgccagccgaact




gttcgccaggctcaaggcgcgcatgcccgacggcgaggatctc




gtcgtgacccatggcgatgcctgcttgccgaatatcatggtg




gaaaatggccgcttttctggattcatcgactgtggccggctgg




gtgtggcggaccgctatcaggacatagcgttggctacccgtgat




attgctgaagagcttggcggcgaatgggctgaccgcttcctcgt




gctttacggtatcgccgcccccgattcgcagcgcatcgccttct




atcgccttcttgacgagttcttctgagcgggactctggggttcg




aataaagaccgaccaagcgacgtctgagagctccctggcgaat




tcggtaccaataaaagagctttattttcatgatctgtgtgttggt




ttttggccgcgttgctggcgtttttccataggctccgcccccctg




acgagcatcacaaaaatcgacgctcaagtcagaggtggcgaaac




ccgacaggactataaagataccaggcgtttccccctggaagct




ccctcgtgcgctctcctgttccgaccctgccgcttaccggatacc




tgtccgcctttctcccttcgggaagcgtggcgctttctcatagct




cacgctgtaggtatctcagttcggtgtaggtcgttcgctccaa




gctgggctgtgtgcacgaaccccccgttcagcccgaccgctgcg




ccttatccggtaactatcgtcttgagtccaacccggtaagaca




cgacttatcgccactggcagcagccactggtaacaggattagca




gagcgaggtatgtaggcggtgctacagagttcttgaagtggtgg




cctaactacggctacactagaagaacagtatttggtatctgcg




ctctgctgaagccagttaccttcggaaaaagagttggtagctctt




gatccggcaaacaaaccaccgctggtagcggtggtttttttgttt




gcaagcagcagattacgcgcagaaaaaaaggatctcaagaaga




tcctttgatcttttctagtgtgcg






attP_Puro
gatgccagctcattcctcccactcatgatctatagatcccccgg
4285


donor DNA:
gctgcaggaattctacccactctgtcgataccccaccgagaccc




cattggggccaatacgcccgcgtttcttccttttccccacccca




ccccccaagttcgggtgaaggcccagggctcgcagccaacgtc




ggggcggcaagcttacatcgagatcccggtttgtctggtcaac




caccgcggtctcagtggtgtacggtacaaaccccgtgagcaag




ggcgaggagctgttcaccggggtggtgcccatcctggtcga




gctggacggcgacgtaaacggccacaagttcagcgtgtccggc




gagggcgagggcgatgccacctacggcaagctgaccctgaag




ttcatctgcaccaccggcaagctgcccgtgccctggcccacc




ctcgtgaccaccctgacctacggcgtgcagtgcttcagccgct




accccgaccacatgaagcagcacgacttcttcaagtccgccat




gcccgaaggctacgtccaggagcgcaccatcttcttcaagga




cgacggcaactacaagacccgcgccgaggtgaagttcgaggg




cgacaccctggtgaaccgcatcgagctgaagggcatcgact




tcaaggaggacggcaacatcctggggcacaagctggagtacaa




ctacaacagccacaacgtctatatcatggccgacaagcagaa




gaacggcatcaaggtgaacttcaagatccgccacaacatcgag




gacggcagcgtgcagctcgccgaccactaccagcagaacac




ccccatcggcgacggccccgtgctgctgcccgacaaccacta




cctgagcacccagtccgccctgagcaaagaccccaacgagaa




gcgcgatcacatggtcctgctggagttcgtgaccgccgccgg




gatcactctcggcatggacgagctgtacaagagcggcctga




ggagcagagcccaggcgagcaacagcgccgtggacgccacc




atgggcgatcgcccgggaattgactagtgcggccgcctaggat




ccatgccgatagcgttggttgagtggataaccgtattaccgcc




aagcttatgcatgtgcccgtcagtgggcagagcgcacatcgc




ccacagtccccgagaagttggggggaggggtcggcaattgaa




ccggtgcctagagaaggtggcgcggggtaaactgggaaagtg




atgtcgtgtactggctccgcctttttcccgagggtgggggaga




accgtatataagtgcagtagtcgccgtgaacgttctttttcgca




acgggtttgccgccagaacacaggtaagtgccgtgtgtggttcc




cgcgggcctggcctctttacgggttatggcccttgcgtgccttga




attacttccacctggctgcagtacgtgattcttgatcccgagctt




cgggttggaagtgggtgggagagttcgaggccttgcgcttaag




gagccccttcgcctcgtgcttgagttgaggcctggcctgggcg




ctggggccgccgcgtgcgaatctggtggcaccttcgcgcctgtct




cgctgctttcgataagtctctagccatttaaaatttttgatgacc




tgctgcgacgctttttttctggcaagatagtcttgtaaatgcgg




gccaagatctgcacactggtatttcggtttttggggccgcgggcg




gcgacggggcccgtgcgtcccagcgcacatgttcggcgagg




cggggcctgcgagcgcggccaccgagaatcggacgggggt




agtctcaagctggccggcctgctctggtgcctggcctcgcg




ccgccgtgtatcgccccgccctgggcggcaaggctggcccggt




cggcaccagttgcgtgagcggaaagatggccgcttcccggc




cctgctgcagggagctcaaaatggaggacgcggcgctcggga




gagcgggcgggtgagtcacccacacaaaggaaaagggcctttc




cgtcctcagccgtcgcttcatgtgactccacggagtaccgggcg




ccgtccaggcacctcgattagttctcgcgcttttggagtacgtc




gtctttaggttggggggaggggttttatgcgatggagtttcccca




cactgagtgggtggagactgaagttaggccagcttggcacttgat




gtaattctccttggaatttgccctttttgagtttggatcttggtt




cattctcaagcctcagacagtggttcaaagtttttttcttccatt




tcaggtgtcgtgagctagcccaccatgaccgagtacaagcccacg




gtgcgcctcgccacccgcgacgacgtcccccgggccgtacgc




accctcgccgccgcgttcgccgactaccccgccacgcgcca




caccgtcgacccggaccgccacatcgagcgggtcaccgagct




gcaagaactcttcctcacgcgcgtcgggctcgacatcggcaa




ggtgtgggtcgcggacgacggcgccgcggtggcggtctgg




accacgccggagagcgtcgaagcgggggcggtgttcgccga




gatcggcccgcgcatggccgagttgagcggttcccggctgg




ccgcgcagcaacagatggaaggcctcctggcgccgcaccggc




ccaaggagcccgcgtggttcctggccaccgtcggcgtctcgc




ccgaccaccagggcaagggtctgggcagcgccgtcgtgctcc




ccggagtggaggcggccgagcgcgccggggtgcccgccttc




ctggagacctccgcgccccgcaacctccccttctacgagcgg




ctcggcttcaccgtcaccgccgacgtcgaggtgcccgaagg




accgcgcacctggtgcatgacccgcaagcccggtgccggatc




gggagagggcagaggaagtctgctaacatgcggtgacgtc




gaggagaatcctggcccaccggtcgccaccagcgagctgatta




aggagaacatgcacatgaagctgtacatggagggcaccgtgg




acaaccatcacttcaagtgcacatccgagggcgaaggcaagc




cctacgagggcacccagaccatgagaatcaaggtggtcgaggg




cggccctctccccttcgccttcgacatcctggctactagcttc




ctctacggcagcaagaccttcatcaaccacacccagggcatc




cccgacttcttcaagcagtccttccctgagggcttcacatgggag




agagtcaccacatacgaagacgggggcgtgctgaccgctacc




caggacaccagcctccaggacggctgcctcatctacaacgtc




aagatcagaggggtgaacttcacatccaacggccctgtgatg




cagaagaaaacactcggctgggaggccttcaccgagacgctgt




accccgctgacggcggcctggaaggcagaaacgacatggcc




ctgaagctcgtgggcgggagccatctgatcgcaaacatcaag




accacatatagatccaagaaacccgctaagaacctcaagatgc




ctggcgtctactatgtggactacagactggaaagaatcaagga




ggccaacaacgagacctacgtcgagcagcacgaggtggcagt




ggccagatactgcgacctccctagcaaactggggcacaagcttaa




ttaagaattctctagaggatccagacatgataagatacattgatg




agtttggacaaaccacaactagaatgcagtgaaaaaaatgcttta




tttgtgaaatttgtgatgctattgctttatttgtaaccattataa




gctgcaataaacaagttaacaacaacaattgcattcattttatgt




ttcaggttcagggggaggtgtgggaggttttttaaagcaagtaaa




acctctacaaatgtggtatggctgattatgatcctgcaagcctcg




tcgccgcggtttattctgttgacaattaatcatcggcatagtat




atcggcatagtataatacgacaaggtgaggaactaaaccatggga




tcggccattgaacaagatggattgcacgcaggttctccggccg




cttgggtggagaggctattcggctatgactgggcacaacagac




gatcggctgctctgatgccgccgtgttccggctgtcagcgcag




gggcgcccggttctttttgtcaagaccgacctgtccggtgccct




gaatgaactgcaggacgaggcagcgcggctatcgtggctggc




cacgacgggcgttccttgcgcagctgtgctcgacgttgtcact




gaagcgggaagggactggctgctattgggcgaagtgccggggc




aggatctcctgtcatctcaccttgctcctgccgagaaagtatc




catcatggctgatgcaatgcggcggctgcatacgcttgatccg




gctacctgcccattcgaccaccaagcgaaacatcgcatcgagc




gagcacgtactcggatggaagccggtcttgtcgatcaggatg




atctggacgaagagcatcaggggctcgcgccagccgaactgtt




cgccaggctcaaggcgcgcatgcccgacggcgaggatctcgt




cgtgacccatggcgatgcctgcttgccgaatatcatggtggaa




aatggccgcttttctggattcatcgactgtggccggctgggtgt




ggcggaccgctatcaggacatagcgttggctacccgtgatattg




ctgaagagcttggcggcgaatgggctgaccgcttcctcgtgct




ttacggtatcgccgcccccgattcgcagcgcatcgccttctatc




gccttcttgacgagttcttctgagcgggactctggggttcgaat




aaagaccgaccaagcgacgtctgagagctccctggcgaattcggt




accaataaaagagctttattttcatgatctgtgtgttggtttttg




gccgcgttgctggcgtttttccataggctccgcccccctgacgag




catcacaaaaatcgacgctcaagtcagaggtggcgaaacccga




caggactataaagataccaggcgtttccccctggaagctccct




cgtgcgctctcctgttccgaccctgccgcttaccggatacctgt




ccgcctttctcccttcgggaagcgtggcgctttctcatagctcac




gctgtaggtatctcagttcggtgtaggtcgttcgctccaagctg




ggctgtgtgcacgaaccccccgttcagcccgaccgctgcgcctt




atccggtaactatcgtcttgagtccaacccggtaagacacgac




ttatcgccactggcagcagccactggtaacaggattagcagag




cgaggtatgtaggcggtgctacagagttcttgaagtggtggcct




aactacggctacactagaagaacagtatttggtatctgcgctct




gctgaagccagttaccttcggaaaaagagttggtagctcttgat




ccggcaaacaaaccaccgctggtagcggtggtttttttgtttgca




agcagcagattacgcgcagaaaaaaaggatctcaagaagatcc




tttgatcttttctagtgtgcg










Multiplex Editing and Recombinase-Mediated Inversion


Large structural variants are found in many human pathogenic alleles, such as the large 0.6-Mb inversion at the F8 locus that causes ˜50% of severe hemophilia A cases. Inspired by the high efficiency of recombinase attachments site insertions using twinPE, it was reasoned that multiplexing the insertion of both attB and attP Bxb1 attachment sites could be used to correct more complex genetic variants by unidirectional deletion or inversion of the intervening DNA sequence. To test this approach on a therapeutically relevant locus, a 39-kb inversion between IDS and its pseudogene IDS2 was attempted. Inversions between these sites have been observed in ˜13% of Hunter syndrome cases, and characterization of the breakpoints in pathogenic alleles has revealed that the inversion often occurs within a recombination hotspot of intron 7 that is present in both IDS and IDS2. Therefore, regions flanking these recombination hotspots were targeted to insert Bxb1 attP and attB attachment sequences, which when oriented in opposite directions, could be used as substrated for unidirectional inversion by Bxb1 for the correction of the pathogenic allele sequence in patient cells (FIG. 111A). After screening 12 pegRNA spacer pairs and optimizing pegRNA sequence features, it was found that pegRNA pairs that are capable of inserting attP or attB attachment sites at the left and right targeted sites with 73.7% and 69.8% efficiency, respectively (FIG. 111B).


Next, multiplexing twinPE-mediated insertion of both attP and attB attachment sites were explored with Bxb1 recombinase-mediated inversion of the 39-kb sequence in IDS and IDS2. Initially, sequential DNA transfections were explored with twinPE components followed by Bxb1 recombinase. A first set of pegRNAs (set 1) was tested that installs a forward-oriented attP sequence in intron 3 of IDS2 and a reverse-oriented attB sequence in intron 7 of IDS. In addition, a second set of pegRNAs (set 2) was used to install a reverse-oriented attP sequence in IDS2 and a forward-oriented attB sequence in IDS in a first transfection. 52.3% or 54.5% attP sequence insertion and 27.6% or 30.9% attB sequence insertion was observed for multiplex editing with pegRNA set 1 and set 2, respectively (FIG. 111C). When edited cells were subsequently transfected with Bxb1 recombinase, a significantly decreased percentage of amplified alleles containing attP and attB sequence was observed compared to mock transfection controls (p-value<0.05), suggesting that some edited alleles were being used as substrates for Bxb1-mediated recombination (FIG. 111C) Amplification of the anticipated inversion junctions followed by amplicon sequencing showed both the attL and attR sequence signatures of Bxb1-mediated recombination, with product purity at or above 80% at both junctions (FIG. 111D).


To carry out one-pot twinPE and Bxb1-mediated inversion and circumvent unwanted recombination between pegRNA plasmid DNA, all-RNA components comprising PE2 mRNA, synthetic pegRNAs from set 1, and Bxb1 mRNA were nucleofected. Using amplicon sequencing, the expected inverted allele junctions containing attR and attL sequences were captured. Furthermore, to quantify the inversion efficiency, a reverse primer that can bind to an identical sequence in both the non-inverted and inverted alleles and therefore amplify both edits using the same primer pair was designed (FIG. 116). ˜4.6-7.2% and ˜1.5-2.3% inversion efficiency for “sequential” and “one-pot” strategy was observed, respectively (FIG. 111E). Collectively, these data support that combining twinPE with site-specific serine recombinases could be used to correct a common 39-kb inversion found in Hunter syndrome alleles and could serve as a promising therapeutic platform for targeting other complex pathogenic variants.


Discussion


As presented herein, a twin prime editing approach that can be used to delete, replace, or insert DNA sequences at targeted location in the genomes of human cells was developed. When combined with site-specific serine recombinase enzymes, twinPE can also support the integration of DNA cargo, or the inversion of DNA sequence.


Methods


General Methods


DNA amplification was conducted by PCR using Phusion U Green Multiplex PCR Master Mix (ThermoFisher Scientific) or Q5 Hot Start High-Fidelity 2× Master Mix (New England BioLabs) unless otherwise noted. DNA oligonucleotides were obtained from Integrated DNA Technologies. Plasmids expressing sgRNAs were constructed by ligation of annealed oligonucleotides into BsmBI-digested acceptor vector. Plasmids expressing pegRNAs were constructed by Gibson assembly or Golden Gate assembly as previously described. Sequences of sgRNA and pegRNA constructs used in this Example are listed in Tables 11-12.









TABLE 11







Sequences of sgRNA used in this Example











SEQ


sgRNA
Spacer Sequence
ID NO:





HEK3_3b_+90_
GTCAACCAGTATCCCGGTGC
 616


nicking







pU6-Sp-sgRNA-
GATTGGCTTTGATTTCCCTA
3971


DMDExo51-A1







pU6-Sp-sgRNA-
GTATATGATTGTTACTGAGA
3972


DMDExo51-A3







pU6-Sp-sgRNA-
Gcagttgcctaagaactggt
3973


DMDExo51-B1
















TABLE 12







Sequences of pegRNA used in this Example













Spacer

Extension



Spacer
SEQ

SEQ


pegRNA
Sequence
ID NO:
3′ Extension
ID NO:





HEK3_attB_A_38
ggcccagactgagc
 383
ATGATCCTGACGACGGAGACCGCCGT
4029



acgtga

CGTCGACAAGCCCGTGCTCAGTCTG






HEK3_attB_A_34
ggcccagactgagc
 383
TCCTGACGACGGAGACCGCCGTCGTC
4030



acgtga

GACAAGCCCGTGCTCAGTCTG






HEK3_attB_A_30
ggcccagactgagc
 383
GACGACGGAGACCGCCGTCGTCGAC
4031



acgtga

AAGCCCGTGCTCAGTCTG






HEK3_attB_B_38
gtcaaccagtatccc
 616
GGCTTGTCGACGACGGCGGTCTCCGT
4032



ggtgc

CGTCAGGATCATCCGGGATACTGG






HEK3_attB_B_34
gtcaaccagtatccc
 616
TGTCGACGACGGCGGTCTCCGTCGTC
4033



ggtgc

AGGATCATCCGGGATACTGG






HEK3_attB_B_30
gtcaaccagtatccc
 616
GACGACGGCGGTCTCCGTCGTCAGGA
4034



ggtgc

TCATCCGGGATACTGG






HEK3_attP_A_43
ggcccagactgagc
 383
TACCGTACACCACTGAGACCGCGGTG
4035



acgtga

GTTGACCAGACAAACCTCGTGCTCAG






TCTG






HEK3_attP_A_39
ggcccagactgagc
 383
GTACACCACTGAGACCGCGGTGGTTG
4036



acgtga

ACCAGACAAACCTCGTGCTCAGTCTG






HEK3_attP_A_35
ggcccagactgagc
 383
ACCACTGAGACCGCGGTGGTTGACCA
4037



acgtga

GACAAACCTCGTGCTCAGTCTG






HEK3_attP_B_44
gtcaaccagtatccc
 616
GTCTGGTCAACCACCGCGGTCTCAGT
4038



ggtgc

GGTGTACGGTACAAACCTCCGGGATA






CTGG






HEK3_attP_B_40
gtcaaccagtatccc
 616
GGTCAACCACCGCGGTCTCAGTGGTG
4039



ggtgc

TACGGTACAAACCTCCGGGATACTGG






HEK3_attP_B_36
gtcaaccagtatccc
 616
AACCACCGCGGTCTCAGTGGTGTACG
4040



ggtgc

GTACAAACCTCCGGGATACTGG






PE3_HEK3_FKBP_ins12bp
ggcccagactgagc
 383
tggaggaagcagggcttcctttcctctgccatcacacctgcac
4041



acgtga

tcccgtgctcagtctg






PE3_HEK3_FKBP_ins36bp
ggcccagactgagc
 383
tggaggaagcagggcttcctttcctctgccatcacccgtctcc
4042



acgtga

tggggagatggtttccacctgcactcccgtgctcagtctg






PE3_HEK3_FKBP_ins108bp
ggcccagactgagc
 383
tggaggaagcagggcttcctttcctctgccatcaaaatttctttc
4043



acgtga

catcttcaagcatcccggtgtagtgcaccacgcaggtctggc






cgcgcttggggaaggtgcgcccgtctcctggggagatggttt






ccacctgcactcccgtgctcagtctg






PE3_HEK3_FKBP_ins321bp
ggcccagactgagc
 383
tggaggaagcagggcttcctttcctctgccatcattccagtttt
4044



acgtga

agaagctccacatcgaagacgagagtggcatgtggtgggat






gatgcctgggtgcccagtggcaccataggcataatctggag






atatagtcagtttggctctctgacccacactcatctgggcaacc






ccttcttcccagcctcggatcacctcctgcttgcctagcataaa






cttaaagggcttgtttctgtcccgggaggaatcaaatttctttcc






atcttcaagcatcccggtgtagtgcaccacgcaggtctggcc






gcgcttggggaaggtgcgcccgtctcctggggagatggtttc






cacctgcactcccgtgctcagtctg






twinPE_HEK3_FKBP_
ggcccagactgagc
 383
accacgcaggtctggccgcgcttggggaaggtgcgcccgt
4045


ins108bp_32bp_
acgtga

ctcctggggagatggtttccacctgcactcccgtgctcagtct



overhang_A


g






twinPE_HEK3_FKBP_
gtcaaccagtatccc
 616
accttccccaagcgcggccagacctgcgtggtgcactacac
4046


ins108bp_32bp_
ggtgc

cgggatgcttgaagatggaaagaaatttccgggatactgg



overhang_B









pU6-Sp-gRNA-
ggcccagactgagc
 383
tcctctgccatcacgtgctcagtctg
4047


HEK3_del_DF_A_13nt
acgtga





TEMP_Basic









pU6-Sp-gRNA-
gtcaaccagtatccc
 616
tgatggcagaggaccgggatactgg
4048


HEK3_del_DF_B_13nt
ggtgc





TEMP_Basic









pU6-Sp-gRNA-
ggcccagactgagc
 383
tggaggaagcagggcttcctttcctctgccatcacgtgctcag
4049


HEK3_del_DF_A_34nt
acgtga

tctg



TEMP_Basic









pU6-Sp-gRNA-
gtcaaccagtatccc
 616
tgatggcagaggaaaggaagccctgcttcctccaccgggat
4050


HEK3_del_DF_B_34nt
ggtgc

actgg



TEMP_Basic









pU6-Sp-gRNA-
ggcccagactgagc
 383
tgcaggagctgcatcctctgccatcacgtgctcagtctg
4051


HEK3_del_DF_A_13-
acgtga





13TEMP_Hybrid









pU6-Sp-gRNA-
gtcaaccagtatccc
 616
tgatggcagaggatgcagctcctgcaccgggatactgg
4052


HEK3_del_DF_B_13-
ggtgc





13TEMP_Hybrid









pU6-Sp-gRNA-
ggcccagactgagc
 383
gcccagccaaacttgtcaaccagtatcccggcgtgctcagtc
4053


HEK3_del_DF_A_31nt
acgtga

tg



TEMP_Shendure









pU6-Sp-gRNA-
gtcaaccagtatccc
 616
gggtcaatccttggggcccagactgagcacgccgggatact
4054


HEK3_del_DF_B_31nt
ggtgc

gg



TEMP_Shendure









pU6-Sp-gRNA-
ggcccagactgagc
 383
tcctctgccatcacgtgctcagtctgTTAAATAACGC
4055


HEK3_del_DF_A_13nt
acgtga

GGTTCTATCTAGTTACGCGTTAAACC



TEMP_Basic_EvoPreQ1


AACTAGAA






pU6-Sp-gRNA-
gtcaaccagtatccc
 616
tgatggcagaggaccgggatactggAAAATATACG
4056


HEK3_del_DF_B_13nt
ggtgc

CGGTTCTATCTAGTTACGCGTTAAAC



TEMP_Basic_EvoPreQ1


CAACTAGAA






pU6-Sp-gRNA-
ggcccagactgagc
 383
tggaggaagcagggcttcctttcctctgccatcacgtgctcag
4057


HEK3_del_DF_A_34nt
acgtga

tctgTTAATAATCGCGGTTCTATCTAGT



TEMP_Basic_EvoPreQ1


TACGCGTTAAACCAACTAGAA






pU6-Sp-gRNA-
gtcaaccagtatccc
 616
tgatggcagaggaaaggaagccctgcttcctccaccgggat
4058


HEK3_del_DF_B_34nt
ggtgc

actggAAAAAAAACGCGGTTCTATCTAG



TEMP_Basic_EvoPreQ1


TTACGCGTTAAACCAACTAGAA






pU6-Sp-gRNA-
ggcccagactgagc
 383
tgcaggagctgcatcctctgccatcacgtgctcagtctgAT
4059


HEK3_del_DF_A_13-
acgtga

AAATAACGCGGTTCTATCTAGTTACG



13TEMP_Hybrid_


CGTTAAACCAACTAGAA



EvoPreQ1









pU6-Sp-gRNA-
gtcaaccagtatccc
 616
tgatggcagaggatgcagctcctgcaccgggatactggAA
4060


HEK3_del_DF_B_13-
ggtgc

AAAAGGCGCGGTTCTATCTAGTTACG



13TEMP_Hybrid_


CGTTAAACCAACTAGAA



EvoPreQ1









pU6-Sp-gRNA-
ggcccagactgagc
 383
gcccagccaaacttgtcaaccagtatcccggcgtgctcagtc
4061


HEK3_del_DF_A_31nt
acgtga

tgTTAAATACCGCGGTTCTATCTAGTT



TEMP_Shendure_


ACGCGTTAAACCAACTAGAA



EvoPreQ1









pU6-Sp-gRNA-
gtcaaccagtatccc
 616
gggtcaatccttggggcccagactgagcacgccgggatact
4062


HEK3_del_DF_B_31nt
ggtgc

ggAAAATAATCGCGGTTCTATCTAGTT



TEMP_Shendure_


ACGCGTTAAACCAACTAGAA



EvoPreQ1









pU6-Sp-pegRNA-
GATTGGCTTT
3971
ttgaataggaagtaaattaatttgaagctggaccctagGGA
4063


DMDExo51-
GATTTCCCTA

AATCAA



A1_a_Basic









pU6-Sp-pegRNA-
GATTGGCTTT
3971
ttgaataggaagtaaattaatttgaagctggaccctagGGA
4064


DMDExo51-
GATTTCCCTA

AATCAAAG



A1_b_Basic









pU6-Sp-pegRNA-
GATTGGCTTT
3971
ttgaataggaagtaaattaatttgaagctggaccctagGGA
4065


DMDExo51-
GATTTCCCTA

AATCAAAGCC



A1_c_Basic









pU6-Sp-pegRNA-
Gcagttgcctaagaa
3973
tagggtccagcttcaaattaatttacttcctattcaaagttcttag
4066


DMDExo51-
ctggt

gc



B1_A1_b_Basic









pU6-Sp-pegRNA-
Gcagttgcctaagaa
3973
tagggtccagcttcaaattaatttacttcctattcaaagttcttag
4067


DMDExo51-
ctggt

gcaa



B1_A1_c_Basic









pU6-Sp-pegRNA-
GTATATGATT
3972
atgcctggacaagtaacttaagttaaataagccttctCAGT
4068


DMDExo51-
GTTACTGAG

AACAATCAT



A3_c_Basic
A








pU6-Sp-pegRNA-
Gcagttgcctaagaa
3973
agaaggcttatttaacttaagttacttgtccaggcatagttctta
4069


DMDExo51-
ctggt

g



B1_A3_a_Basic









pu6-sp-pegrna-
GTATATGATT
3972
tgctgagagagaaacagttgcctaagaactCAGTAAC
4070


dmdexo51-a3-
GTTACTGAG

AAT



b1_a_PrimeDel
A








pu6-sp-pegrna-
Gcagttgcctaagaa
3973
accacttccacaatgtatatgattgttactgagttcttagg
4071


dmdexo51-b1-a3_a-
ctggt





PrimeDel









pu6-sp-pegrna-
Gcagttgcctaagaa
3973
accacttccacaatgtatatgattgttactgagttcttaggcaa
4072


dmdexo51-b1-a3_b-
ctggt





PrimeDel









pU6-Sp-pegRNA-
GTATATGATT
3972
ATGATCCTGACGACGGAGACCGCCGT
4073


DMDExo51-
GTTACTGAG

CGTCGACAAGCCCAGTAACAATC



A3_b_attB_fwd
A








pU6-Sp-pegRNA-
GTATATGATT
3972
ATGATCCTGACGACGGAGACCGCCGT
4074


DMDExo51-
GTTACTGAG

CGTCGACAAGCCCAGTAACAATCATA



A3_c_attB_fwd
A








pU6-Sp-pegRNA-
Gcagttgcctaagaa
3973
GGCTTGTCGACGACGGCGGTCTCCGT
4075


DMDExo51-
ctggt

CGTCAGGATCATagttcttaggc



B1_b_attB_fwd









pU6-Sp-pegRNA-
Gcagttgcctaagaa
3973
GGCTTGTCGACGACGGCGGTCTCCGT
4076


DMDExo51-
ctggt

CGTCAGGATCATagttcttaggcaac



B1_c_attB_fwd









pU6-Sp-pegRNA-
Gaggagagtaaagt
3974
GGCTTGTCGACGACGGCGGTCTCCGT
4077


DMDExo51-
gattgg

CGTCAGGATCATatcactttactc



B2_b_attB_fwd









pU6-Sp-pegRNA-
Gaggagagtaaagt
3974
GGCTTGTCGACGACGGCGGTCTCCGT
4078


DMDExo51-
gattgg

CGTCAGGATCATatcactttactctcc



B2_c_attB_fwd









A_1077a
GCAGAGCCA
3975
TACCGTACACCACTGAGACCGCGGTG
4079



GGAACCCCT

GTTGACCAGACAAACCTGGGGTTCCT




GT








A_1077b
GCAGAGCCA
3975
TACCGTACACCACTGAGACCGCGGTG
4080



GGAACCCCT

GTTGACCAGACAAACCTGGGGTTCCT




GT

GG






A_1077c
GCAGAGCCA
3975
TACCGTACACCACTGAGACCGCGGTG
4081



GGAACCCCT

GTTGACCAGACAAACCTGGGGTTCCT




GT

GGCT






A_1098a
GGGAAGGGG
3976
TACCGTACACCACTGAGACCGCGGTG
4082



CAGGAGAGC

GTTGACCAGACAAACCTCTCTCCTGC




CA








A_1098b
GGGAAGGGG
3976
TACCGTACACCACTGAGACCGCGGTG
4083



CAGGAGAGC

GTTGACCAGACAAACCTCTCTCCTGC




CA

CC






A_1098c
GGGAAGGGG
3976
TACCGTACACCACTGAGACCGCGGTG
4084



CAGGAGAGC

GTTGACCAGACAAACCTCTCTCCTGC




CA

CCCT






A_1246a
GAATATGTC
3977
TACCGTACACCACTGAGACCGCGGTG
4085



CCAGATAGC

GTTGACCAGACAAACCTCTATCTGGG




AC








A_1246b
GAATATGTC
3977
TACCGTACACCACTGAGACCGCGGTG
4086



CCAGATAGC

GTTGACCAGACAAACCTCTATCTGGG




AC

AC






A_1246c
GAATATGTC
3977
TACCGTACACCACTGAGACCGCGGTG
4087



CCAGATAGC

GTTGACCAGACAAACCTCTATCTGGG




AC

ACAT






A_1267a
GGGGACTCT
3978
TACCGTACACCACTGAGACCGCGGTG
4088



TTAAGGAAA

GTTGACCAGACAAACCTTTCCTTAAA




GA








A_1267b
GGGGACTCT
3978
TACCGTACACCACTGAGACCGCGGTG
4089



TTAAGGAAA

GTTGACCAGACAAACCTTTCCTTAAA




GA

GA






A_1267c
GGGGACTCT
3978
TACCGTACACCACTGAGACCGCGGTG
4090



TTAAGGAAA

GTTGACCAGACAAACCTTTCCTTAAA




GA

GAGT






A_1293a
GAGAAAGAG
3979
TACCGTACACCACTGAGACCGCGGTG
4091



AAAGGGAGT

GTTGACCAGACAAACCTCTCCCTTTC




AG








A_1293b
GAGAAAGAG
3979
TACCGTACACCACTGAGACCGCGGTG
4092



AAAGGGAGT

GTTGACCAGACAAACCTCTCCCTTTC




AG

TC






A_1293c
GAGAAAGAG
3979
TACCGTACACCACTGAGACCGCGGTG
4093



AAAGGGAGT

GTTGACCAGACAAACCTCTCCCTTTC




AG

TCTT






A_1307a
GAGTAGAGG
3980
TACCGTACACCACTGAGACCGCGGTG
4094



CGGCCACGA

GTTGACCAGACAAACCTCGTGGCCGC




CC








A_1307b
GAGTAGAGG
3980
TACCGTACACCACTGAGACCGCGGTG
4095



CGGCCACGA

GTTGACCAGACAAACCTCGTGGCCGC




CC

CT






A_1307c
GAGTAGAGG
3980
TACCGTACACCACTGAGACCGCGGTG
4096



CGGCCACGA

GTTGACCAGACAAACCTCGTGGCCGC




CC

CTCT






A_1582a
GATCAGTGA
3981
TACCGTACACCACTGAGACCGCGGTG
4097



AACGCACCA

GTTGACCAGACAAACCTGGTGCGTTT




GA








A_1582b
GATCAGTGA
3981
TACCGTACACCACTGAGACCGCGGTG
4098



AACGCACCA

GTTGACCAGACAAACCTGGTGCGTTT




GA

CA






A_1582c
GATCAGTGA
3981
TACCGTACACCACTGAGACCGCGGTG
4099



AACGCACCA

GTTGACCAGACAAACCTGGTGCGTTT




GA

CACT






A_1615a
GCAGCTCAG
3982
TACCGTACACCACTGAGACCGCGGTG
4100



GTTCTGGGA

GTTGACCAGACAAACCTCCCAGAACC




GA








A_1615b
GCAGCTCAG
3982
TACCGTACACCACTGAGACCGCGGTG
4101



GTTCTGGGA

GTTGACCAGACAAACCTCCCAGAACC




GA

TG






A_1615c
GCAGCTCAG
3982
TACCGTACACCACTGAGACCGCGGTG
4102



GTTCTGGGA

GTTGACCAGACAAACCTCCCAGAACC




GA

TGAG






A_1647a
GTGGCCACT
3983
TACCGTACACCACTGAGACCGCGGTG
4103



GAGAACCGG

GTTGACCAGACAAACCTCGGTTCTCA




GC








A_1647b
GTGGCCACT
3983
TACCGTACACCACTGAGACCGCGGTG
4104



GAGAACCGG

GTTGACCAGACAAACCTCGGTTCTCA




GC

GT






A_1647c
GTGGCCACT
3983
TACCGTACACCACTGAGACCGCGGTG
4105



GAGAACCGG

GTTGACCAGACAAACCTCGGTTCTCA




GC

GTGG






A_1810a
GAATCTGCCT
3984
TACCGTACACCACTGAGACCGCGGTG
4106



AACAGGAGG

GTTGACCAGACAAACCTTCCTGTTAG




T








A_1810b
GAATCTGCCT
3984
TACCGTACACCACTGAGACCGCGGTG
4107



AACAGGAGG

GTTGACCAGACAAACCTTCCTGTTAG




T

GC






A_1810c
GAATCTGCCT
3984
TACCGTACACCACTGAGACCGCGGTG
4108



AACAGGAGG

GTTGACCAGACAAACCTTCCTGTTAG




T

GCAG






A_1890a
GTCACCAAT
3985
TACCGTACACCACTGAGACCGCGGTG
4109



CCTGTCCCTA

GTTGACCAGACAAACCTGGGACAGG




G

A






A_1890b
GTCACCAAT
3985
TACCGTACACCACTGAGACCGCGGTG
4110



CCTGTCCCTA

GTTGACCAGACAAACCTGGGACAGG




G

ATT






A_1890c
GTCACCAAT
3985
TACCGTACACCACTGAGACCGCGGTG
4111



CCTGTCCCTA

GTTGACCAGACAAACCTGGGACAGG




G

ATTGG






A_3786a
GCTGGCCCC
3986
TACCGTACACCACTGAGACCGCGGTG
4112



CCACCGCCC

GTTGACCAGACAAACCTGGCGGTGGG




CA








A_3786b
GCTGGCCCC
3986
TACCGTACACCACTGAGACCGCGGTG
4113



CCACCGCCC

GTTGACCAGACAAACCTGGCGGTGGG




CA

GG






A_3786c
GCTGGCCCC
3986
TACCGTACACCACTGAGACCGCGGTG
4114



CCACCGCCC

GTTGACCAGACAAACCTGGCGGTGGG




CA

GGGC






A_3835a
GACGTCACG
3987
TACCGTACACCACTGAGACCGCGGTG
4115



GCGCTGCCC

GTTGACCAGACAAACCTGGCAGCGCC




CA








A_3835b
GACGTCACG
3987
TACCGTACACCACTGAGACCGCGGTG
4116



GCGCTGCCC

GTTGACCAGACAAACCTGGCAGCGCC




CA

GT






A_3835c
GACGTCACG
3987
TACCGTACACCACTGAGACCGCGGTG
4117



GCGCTGCCC

GTTGACCAGACAAACCTGGCAGCGCC




CA

GTGA






A_3856a
GGTGTGCTG
3988
TACCGTACACCACTGAGACCGCGGTG
4118



GGCAGGTCG

GTTGACCAGACAAACCTGACCTGCCC




CG








A_3856b
GGTGTGCTG
3988
TACCGTACACCACTGAGACCGCGGTG
4119



GGCAGGTCG

GTTGACCAGACAAACCTGACCTGCCC




CG

AG






A_3856c
GGTGTGCTG
3988
TACCGTACACCACTGAGACCGCGGTG
4120



GGCAGGTCG

GTTGACCAGACAAACCTGACCTGCCC




CG

AGCA






B_1154a
GTCCTTGGCA
3989
GTCTGGTCAACCACCGCGGTCTCAGT
4121



AGCCCAGGA

GGTGTACGGTACAAACCTCTGGGCTT




G

G






B_1154b
GTCCTTGGCA
3989
GTCTGGTCAACCACCGCGGTCTCAGT
4122



AGCCCAGGA

GGTGTACGGTACAAACCTCTGGGCTT




G

GCC






B_1154c
GTCCTTGGCA
3989
cTCTGGTCAACCACCGCGGTCTCAGT
4123



AGCCCAGGA

GGTGTACGGTACAAACCTCTGGGCTT




G

GCCAA






B_1314a
GTGCGTCCTA
3990
GTCTGGTCAACCACCGCGGTCTCAGT
4124



GGTGTTCACC

GGTGTACGGTACAAACCTGAACACCT






A






B_1314b
GTGCGTCCTA
3990
GTCTGGTCAACCACCGCGGTCTCAGT
4125



GGTGTTCACC

GGTGTACGGTACAAACCTGAACACCT






AGG






B_1314c
GTGCGTCCTA
3990
TCTGGTCAACCACCGCGGTCTCAGTG
4126



GGTGTTCACC

GTGTACGGTACAAACCTGAACACCTA






GGAC






B_1376a
GTCCTGGCA
3991
GTCTGGTCAACCACCGCGGTCTCAGT
4127



GGGCTGTGG

GGTGTACGGTACAAACCTCACAGCCC




TG

T






B_1376b
GTCCTGGCA
3991
GTCTGGTCAACCACCGCGGTCTCAGT
4128



GGGCTGTGG

GGTGTACGGTACAAACCTCACAGCCC




TG

TGC






B_1376c
GTCCTGGCA
3991
TCTGGTCAACCACCGCGGTCTCAGTG
4129



GGGCTGTGG

GTGTACGGTACAAACCTCACAGCCCT




TG

GCCA






B_1640a
GTGACCTGC
3992
GTCTGGTCAACCACCGCGGTCTCAGT
4130



CCGGTTCTCA

GGTGTACGGTACAAACCTAGAACCGG




G

G






B_1640b
GTGACCTGC
3992
GTCTGGTCAACCACCGCGGTCTCAGT
4131



CCGGTTCTCA

GGTGTACGGTACAAACCTAGAACCGG




G

GCA






B_1640c
GTGACCTGC
3992
TCTGGTCAACCACCGCGGTCTCAGTG
4132



CCGGTTCTCA

GTGTACGGTACAAACCTAGAACCGGG




G

CAGG






B_1676a
GAGCTTGGC
3993
GTCTGGTCAACCACCGCGGTCTCAGT
4133



AGGGGGTGG

GGTGTACGGTACAAACCTCACCCCCT




GA

G






B_1676b
GAGCTTGGC
3993
GTCTGGTCAACCACCGCGGTCTCAGT
4134



AGGGGGTGG

GGTGTACGGTACAAACCTCACCCCCT




GA

GCC






B_1676c
GAGCTTGGC
3993
TCTGGTCAACCACCGCGGTCTCAGTG
4135



AGGGGGTGG

GTGTACGGTACAAACCTCACCCCCTG




GA

CCAA






B_1701a
GAGCCAGAG
3994
GTCTGGTCAACCACCGCGGTCTCAGT
4136



AGGATCCTG

GGTGTACGGTACAAACCTAGGATCCT




GG

C






B_1701b
GAGCCAGAG
3994
GTCTGGTCAACCACCGCGGTCTCAGT
4137



AGGATCCTG

GGTGTACGGTACAAACCTAGGATCCT




GG

CTC






B_1701c
GAGCCAGAG
3994
TCTGGTCAACCACCGCGGTCTCAGTG
4138



AGGATCCTG

GTGTACGGTACAAACCTAGGATCCTC




GG

TCTG






B_1705a
GATGGAGCC
3995
GTCTGGTCAACCACCGCGGTCTCAGT
4139



AGAGAGGAT

GGTGTACGGTACAAACCTTCCTCTCT




CC

G






B_1705b
GATGGAGCC
3995
GTCTGGTCAACCACCGCGGTCTCAGT
4140



AGAGAGGAT

GGTGTACGGTACAAACCTTCCTCTCT




CC

GGC






B_1705c
GATGGAGCC
3995
TCTGGTCAACCACCGCGGTCTCAGTG
4141



AGAGAGGAT

GTGTACGGTACAAACCTTCCTCTCTG




CC

GCTC






B_1883a
GGGGCCACT
3996
GTCTGGTCAACCACCGCGGTCTCAGT
4142



AGGGACAGG

GGTGTACGGTACAAACCTCTGTCCCT




AT

A






B_1883b
GGGGCCACT
3996
GTCTGGTCAACCACCGCGGTCTCAGT
4143



AGGGACAGG

GGTGTACGGTACAAACCTCTGTCCCT




AT

AGT






B_1883c
GGGGCCACT
3996
TCTGGTCAACCACCGCGGTCTCAGTG
4144



AGGGACAGG

GTGTACGGTACAAACCTCTGTCCCTA




AT

GTGG






B_1902a
GTCCCCTCCA
3997
GTCTGGTCAACCACCGCGGTCTCAGT
4145



CCCCACAGT

GGTGTACGGTACAAACCTTGTGGGGT




G

G






B_1902b
GTCCCCTCCA
3997
GTCTGGTCAACCACCGCGGTCTCAGT
4146



CCCCACAGT

GGTGTACGGTACAAACCTTGTGGGGT




G

GGA






B_1902c
GTCCCCTCCA
3997
TCTGGTCAACCACCGCGGTCTCAGTG
4147



CCCCACAGT

GTGTACGGTACAAACCTTGTGGGGTG




G

GAGG






B_1962a
GGGACCACC
3998
GTCTGGTCAACCACCGCGGTCTCAGT
4148



TTATATTCCC

GGTGTACGGTACAAACCTGAATATAA




A

G






B_1962b
GGGACCACC
3998
GTCTGGTCAACCACCGCGGTCTCAGT
4149



TTATATTCCC

GGTGTACGGTACAAACCTGAATATAA




A

GGT






B_1962c
GGGACCACC
3998
TCTGGTCAACCACCGCGGTCTCAGTG
4150



TTATATTCCC

GTGTACGGTACAAACCTGAATATAAG




A

GTGG






B_3839a
GACCTGCCC
3999
GTCTGGTCAACCACCGCGGTCTCAGT
4151



AGCACACCC

GGTGTACGGTACAAACCTGGTGTGCT




TG

G






B_3839b
GACCTGCCC
3999
GTCTGGTCAACCACCGCGGTCTCAGT
4152



AGCACACCC

GGTGTACGGTACAAACCTGGTGTGCT




TG

GGG






B_3839c
GACCTGCCC
3999
TCTGGTCAACCACCGCGGTCTCAGTG
4153



AGCACACCC

GTGTACGGTACAAACCTGGTGTGCTG




TG

GGCA






B_3903a
GCGACTCCT
4000
GTCTGGTCAACCACCGCGGTCTCAGT
4154



GGAAGTGGC

GGTGTACGGTACAAACCTCCACTTCC




CA

A






B_3903b
GCGACTCCT
4000
GTCTGGTCAACCACCGCGGTCTCAGT
4155



GGAAGTGGC

GGTGTACGGTACAAACCTCCACTTCC




CA

AGG






B_3903c
GCGACTCCT
4000
TCTGGTCAACCACCGCGGTCTCAGTG
4156



GGAAGTGGC

GTGTACGGTACAAACCTCCACTTCCA




CA

GGAG






B_3930a
GGACTTCCC
4001
GTCTGGTCAACCACCGCGGTCTCAGT
4157



AGTGTGCAT

GGTGTACGGTACAAACCTTGCACACT




CG

G






B_3930b
GGACTTCCC
4001
GTCTGGTCAACCACCGCGGTCTCAGT
4158



AGTGTGCAT

GGTGTACGGTACAAACCTTGCACACT




CG

GGG






B_3930c
GGACTTCCC
4001
TCTGGTCAACCACCGCGGTCTCAGTG
4159



AGTGTGCAT

GTGTACGGTACAAACCTTGCACACTG




CG

GGAA






A_1_223_a
TTTGCAGTTT
4002
ATGATCCTGACGACGGAGACCGCCGT
4160



ATCAGGATG

CGTCGACAAGCCGCAGTTTATC




A








A_1_223_b
TTTGCAGTTT
4002
ATGATCCTGACGACGGAGACCGCCGT
4161



ATCAGGATG

CGTCGACAAGCCGCAGTTTATCAG




A








A_1_223_c
TTTGCAGTTT
4002
ATGATCCTGACGACGGAGACCGCCGT
4162



ATCAGGATG

CGTCGACAAGCCGCAGTTTATCAGG




A








A_2_260_a
GGTTGAGCA
4003
ATGATCCTGACGACGGAGACCGCCGT
4163



GGTAGATGT

CGTCGACAAGCCTGAGCAGGT




CA








A_2_260_b
GGTTGAGCA
4003
ATGATCCTGACGACGGAGACCGCCGT
4164



GGTAGATGT

CGTCGACAAGCCTGAGCAGGTAG




CA








A_2_260_c
GGTTGAGCA
4003
ATGATCCTGACGACGGAGACCGCCGT
4165



GGTAGATGT

CGTCGACAAGCCTGAGCAGGTAGAT




CA








A_3_325_a
CTGGGCGGC
4004
ATGATCCTGACGACGGAGACCGCCGT
4166



AGCATAGTG

CGTCGACAAGCCGGCGGCAGC




AG








A_3_325_b
CTGGGCGGC
4004
ATGATCCTGACGACGGAGACCGCCGT
4167



AGCATAGTG

CGTCGACAAGCCGGCGGCAGCAT




AG








A_3_325_c
CTGGGCGGC
4004
ATGATCCTGACGACGGAGACCGCCGT
4168



AGCATAGTG

CGTCGACAAGCCGGCGGCAGCATAG




AG








A_4_360_a
GTCAAGAGT
4005
ATGATCCTGACGACGGAGACCGCCGT
4169



TGACACATT

CGTCGACAAGCCAAGAGTTGAC




GT








A_4_360_b
GTCAAGAGT
4005
ATGATCCTGACGACGGAGACCGCCGT
4170



TGACACATT

CGTCGACAAGCCAAGAGTTGACAC




GT








A_4_360_c
GTCAAGAGT
4005
ATGATCCTGACGACGGAGACCGCCGT
4171



TGACACATT

CGTCGACAAGCCAAGAGTTGACACA




GT








A_5_507_a
CCCAAGTGA
4006
ATGATCCTGACGACGGAGACCGCCGT
4172



TCACACTTGT

CGTCGACAAGCCAAGTGATCAC




C








A_5_507_b
CCCAAGTGA
4006
ATGATCCTGACGACGGAGACCGCCGT
4173



TCACACTTGT

CGTCGACAAGCCAAGTGATCACAC




C








A_5_507_c
CCCAAGTGA
4006
ATGATCCTGACGACGGAGACCGCCGT
4174



TCACACTTGT

CGTCGACAAGCCAAGTGATCACACTT




C








A_6_510_a
CCACCCAAG
4007
ATGATCCTGACGACGGAGACCGCCGT
4175



TGATCACACT

CGTCGACAAGCCCCCAAGTGA




T








A_6_510_b
CCACCCAAG
4007
ATGATCCTGACGACGGAGACCGCCGT
4176



TGATCACACT

CGTCGACAAGCCCCCAAGTGATC




T








A_6_510_c
CCACCCAAG
4007
ATGATCCTGACGACGGAGACCGCCGT
4177



TGATCACACT

CGTCGACAAGCCCCCAAGTGATCAC




T








A_7_532_a
GGGAGAGAC
4008
ATGATCCTGACGACGGAGACCGCCGT
4178



GCAAACACA

CGTCGACAAGCCAGAGACGCA




GC








A_7_532_b
GGGAGAGAC
4008
ATGATCCTGACGACGGAGACCGCCGT
4179



GCAAACACA

CGTCGACAAGCCAGAGACGCAAA




GC








A_7_532_c
GGGAGAGAC
4008
ATGATCCTGACGACGGAGACCGCCGT
4180



GCAAACACA

CGTCGACAAGCCAGAGACGCAAACA




GC








B_1_272_a
TCTCTGACCT
4009
GGCTTGTCGACGACGGCGGTCTCCGT
4181



GTTTTTCCTT

CGTCAGGATCATCTGACCTG






B_1_272_b
TCTCTGACCT
4009
GGCTTGTCGACGACGGCGGTCTCCGT
4182



GTTTTTCCTT

CGTCAGGATCATCTGACCTGT






B_1_272_c
TCTCTGACCT
4009
GGCTTGTCGACGACGGCGGTCTCCGT
4183



GTTTTTCCTT

CGTCAGGATCATCTGACCTGTTT






B_2_291_a
TCTTACTGTC
4010
GGCTTGTCGACGACGGCGGTCTCCGT
4184



CCCTTCTGGG

CGTCAGGATCATTACTGTCCC






B_2_291_b
TCTTACTGTC
4010
GGCTTGTCGACGACGGCGGTCTCCGT
4185



CCCTTCTGGG

CGTCAGGATCATTACTGTCCCCT






B_2_291_c
TCTTACTGTC
4010
GGCTTGTCGACGACGGCGGTCTCCGT
4186



CCCTTCTGGG

CGTCAGGATCATTACTGTCCCCTTC






B_3_305_a
TCTGGGCTCA
4011
GGCTTGTCGACGACGGCGGTCTCCGT
4187



CTATGCTGCC

CGTCAGGATCATGGGCTCACT






B_3_305_b
TCTGGGCTCA
4011
GGCTTGTCGACGACGGCGGTCTCCGT
4188



CTATGCTGCC

CGTCAGGATCATGGGCTCACTAT






B_3_305_c
TCTGGGCTCA
4011
GGCTTGTCGACGACGGCGGTCTCCGT
4189



CTATGCTGCC

CGTCAGGATCATGGGCTCACTATGC






B_4_326_a
CCCAGTGGG
4012
GGCTTGTCGACGACGGCGGTCTCCGT
4190



ACTTTGGAA

CGTCAGGATCATAGTGGGACT




AT








B_4_326_b
CCCAGTGGG
4012
GGCTTGTCGACGACGGCGGTCTCCGT
4191



ACTTTGGAA

CGTCAGGATCATAGTGGGACTTTG




AT








B_4_326_c
CCCAGTGGG
4012
GGCTTGTCGACGACGGCGGTCTCCGT
4192



ACTTTGGAA

CGTCAGGATCATAGTGGGACTTTGG




AT








B_5_330_a
GTGGGACTTT
4013
GGCTTGTCGACGACGGCGGTCTCCGT
4193



GGAAATACA

CGTCAGGATCATGGACTTTGG




A








B_5_330_b
GTGGGACTTT
4013
GGCTTGTCGACGACGGCGGTCTCCGT
4194



GGAAATACA

CGTCAGGATCATGGACTTTGGAA




A








B_5_330_c
GTGGGACTTT
4013
GGCTTGTCGACGACGGCGGTCTCCGT
4195



GGAAATACA

CGTCAGGATCATGGACTTTGGAAAT




A








B_6_414_a
CCTGACAAT
4014
GGCTTGTCGACGACGGCGGTCTCCGT
4196



CGATAGGTA

CGTCAGGATCATGACAATCGA




CC








B_6_414_b
CCTGACAAT
4014
GGCTTGTCGACGACGGCGGTCTCCGT
4197



CGATAGGTA

CGTCAGGATCATGACAATCGATA




CC








B_6_414_c
CCTGACAAT
4014
GGCTTGTCGACGACGGCGGTCTCCGT
4198



CGATAGGTA

CGTCAGGATCATGACAATCGATAGG




CC








B_7_538_a
GGAATCATC
4015
GGCTTGTCGACGACGGCGGTCTCCGT
4199



TTTACCAGAT

CGTCAGGATCATATCATCTTTA




C








B_7_538_b
GGAATCATC
4015
GGCTTGTCGACGACGGCGGTCTCCGT
4200



TTTACCAGAT

CGTCAGGATCATATCATCTTTACC




C








B_7_538_c
GGAATCATC
4015
GGCTTGTCGACGACGGCGGTCTCCGT
4201



TTTACCAGAT

CGTCAGGATCATATCATCTTTACCAG




C








B_8_584_a
GCAGCTCTC
4016
GGCTTGTCGACGACGGCGGTCTCCGT
4202



ATTTTCCATA

CGTCAGGATCATGCTCTCATT




C








B_8_584_b
GCAGCTCTC
4016
GGCTTGTCGACGACGGCGGTCTCCGT
4203



ATTTTCCATA

CGTCAGGATCATGCTCTCATTTTC




C








B_8_584_c
GCAGCTCTC
4016
GGCTTGTCGACGACGGCGGTCTCCGT
4204



ATTTTCCATA

CGTCAGGATCATGCTCTCATTTTCCA




C








B_9_601_a
TACAGTCAG
4017
GGCTTGTCGACGACGGCGGTCTCCGT
4205



TATCAATTCT

CGTCAGGATCATAGTCAGTAT




G








B_9_601_b
TACAGTCAG
4017
GGCTTGTCGACGACGGCGGTCTCCGT
4206



TATCAATTCT

CGTCAGGATCATAGTCAGTATCA




G








B_9_601_c
TACAGTCAG
4017
GGCTTGTCGACGACGGCGGTCTCCGT
4207



TATCAATTCT

CGTCAGGATCATAGTCAGTATCAAT




G








A_3_325_a
CTGGGCGGC
4004
ATGATCCTGACGACGGAGACCGCCGT
4166



AGCATAGTG

CGTCGACAAGCCGGCGGCAGC




AG








B_6_414_b
CCTGACAAT
4014
GGCTTGTCGACGACGGCGGTCTCCGT
4197



CGATAGGTA

CGTCAGGATCATGACAATCGATA




CC








A_5_507_c
CCCAAGTGA
4006
ATGATCCTGACGACGGAGACCGCCGT
4174



TCACACTTGT

CGTCGACAAGCCAAGTGATCACACTT




C








A_6_510_b
CCACCCAAG
4007
ATGATCCTGACGACGGAGACCGCCGT
4176



TGATCACACT

CGTCGACAAGCCCCCAAGTGATC




T








A_7_532_b
GGGAGAGAC
4008
ATGATCCTGACGACGGAGACCGCCGT
4179



GCAAACACA

CGTCGACAAGCCAGAGACGCAAA




GC








B_8_584_b
GCAGCTCTC
4016
GGCTTGTCGACGACGGCGGTCTCCGT
4203



ATTTTCCATA

CGTCAGGATCATGCTCTCATTTTC




C








A_1077c
GCAGAGCCA
3975
TACCGTACACCACTGAGACCGCGGTG
4081



GGAACCCCT

GTTGACCAGACAAACCTGGGGTTCCT




GT

GGCT






B_1154c
GTCCTTGGCA
3989
cTCTGGTCAACCACCGCGGTCTCAGT
4123



AGCCCAGGA

GGTGTACGGTACAAACCTCTGGGCTT




G

GCCAA






A_3786c
GCTGGCCCC
3986
TACCGTACACCACTGAGACCGCGGTG
4114



CCACCGCCC

GTTGACCAGACAAACCTGGCGGTGGG




CA

GGGC






B_3903c
GCGACTCCT
4000
TCTGGTCAACCACCGCGGTCTCAGTG
4156



GGAAGTGGC

GTGTACGGTACAAACCTCCACTTCCA




CA

GGAG






B_3930c
GGACTTCCC
4001
TCTGGTCAACCACCGCGGTCTCAGTG
4159



AGTGTGCAT

GTGTACGGTACAAACCTTGCACACTG




CG

GGAA






A_7_532_b
GGGAGAGAC
4008
ATGATCCTGACGACGGAGACCGCCGT
4179



GCAAACACA

CGTCGACAAGCCAGAGACGCAAA




GC








B_8_584_b
GCAGCTCTC
4016
GGCTTGTCGACGACGGCGGTCTCCGT
4203



ATTTTCCATA

CGTCAGGATCATGCTCTCATTTTC




C








A7_attB_30
GGGAGAGAC
4008
TCCTGACGACGGAGACCGCCGTCGTC
4208



GCAAACACA

GACAAGCCAGAGACGCAAA




GC








B8_attB_30
GCAGCTCTC
4016
TGTCGACGACGGCGGTCTCCGTCGTC
4209



ATTTTCCATA

AGGATCATGCTCTCATTTTC




C








A7_attB_20
GGGAGAGAC
4008
ACGACGGAGACCGCCGTCGTCGACA
4210



GCAAACACA

AGCCAGAGACGCAAA




GC








B8_attB_20
GCAGCTCTC
4016
ACGACGGCGGTCTCCGTCGTCAGGAT
4211



ATTTTCCATA

CATGCTCTCATTTTC




C








A7_attB_GA_38
GGGAGAGAC
4008
ATGATCCTGACGACGGAGTCCGCCGT
4212



GCAAACACA

CGTCGACAAGCCAGAGACGCAAA




GC








B8_attB_GA_38
GCAGCTCTC
4016
GGCTTGTCGACGACGGCGGACTCCGT
4213



ATTTTCCATA

CGTCAGGATCATGCTCTCATTTTC




C








A7_attB_GA_30
GGGAGAGAC
4008
TCCTGACGACGGAGTCCGCCGTCGTC
4214



GCAAACACA

GACAAGCCAGAGACGCAAA




GC








B8_attB_GA_30
GCAGCTCTC
4016
TGTCGACGACGGCGGACTCCGTCGTC
4215



ATTTTCCATA

AGGATCATGCTCTCATTTTC




C








A7_attB_GA_20
GGGAGAGAC
4008
ACGACGGAGTCCGCCGTCGTCGACAA
4216



GCAAACACA

GCCAGAGACGCAAA




GC








B8_attB_GA_20
GCAGCTCTC
4016
ACGACGGCGGACTCCGTCGTCAGGAT
4217



ATTTTCCATA

CATGCTCTCATTTTC




C








A7_attP_50
GGGAGAGAC
4008
GGGTTTGTACCGTACACCACTGAGAC
4218



GCAAACACA

CGCGGTGGTTGACCAGACAAACCAA




GC

GAGACGCAAA






B8_attP_50
GCAGCTCTC
4016
TGGTTTGTCTGGTCAACCACCGCGGT
4219



ATTTTCCATA

CTCAGTGGTGTACGGTACAAACCCGC




C

TCTCATTTTC






A7_attP_40
GGGAGAGAC
4008
TGTACCGTACACCACTGAGACCGCGG
4220



GCAAACACA

TGGTTGACCAGACAAACCAAGAGAC




GC

GCAAA






B8_attP_40
GCAGCTCTC
4016
TGTCTGGTCAACCACCGCGGTCTCAG
4221



ATTTTCCATA

TGGTGTACGGTACAAACCCGCTCTCA




C

TTTTC






A7_attP_30
GGGAGAGAC
4008
CGTACACCACTGAGACCGCGGTGGTT
4222



GCAAACACA

GACCAGACAAACCAAGAGACGCAAA




GC








B8_attP_30
GCAGCTCTC
4016
GGTCAACCACCGCGGTCTCAGTGGTG
4223



ATTTTCCATA

TACGGTACAAACCCGCTCTCATTTTC




C








A7_attP_GA_50
GGGAGAGAC
4008
GGGTTTGTACCGTACACCACTGAGTC
4224



GCAAACACA

CGCGGTGGTTGACCAGACAAACCAA




GC

GAGACGCAAA






B8_attP_GA_50
GCAGCTCTC
4016
TGGTTTGTCTGGTCAACCACCGCGGA
4225



ATTTTCCATA

CTCAGTGGTGTACGGTACAAACCCGC




C

TCTCATTTTC






A7_attP_GA_40
GGGAGAGAC
4008
TGTACCGTACACCACTGAGTCCGCGG
4226



GCAAACACA

TGGTTGACCAGACAAACCAAGAGAC




GC

GCAAA






B8_attP_GA_40
GCAGCTCTC
4016
TGTCTGGTCAACCACCGCGGACTCAG
4227



ATTTTCCATA

TGGTGTACGGTACAAACCCGCTCTCA




C

TTTTC






A7_attP_GA_30
GGGAGAGAC
4008
CGTACACCACTGAGTCCGCGGTGGTT
4228



GCAAACACA

GACCAGACAAACCAAGAGACGCAAA




GC








B8_attP_GA_30
GCAGCTCTC
4016
GGTCAACCACCGCGGACTCAGTGGTG
4229



ATTTTCCATA

TACGGTACAAACCCGCTCTCATTTTC




C








A277_b
GACTGAAAC
4018
AACTTCACAGAGGCTTGTCGACGACG
4230



TTCACAGAA

GCGGTCTCCGTCGTCAGGATCAT




TA








A277_c
GACTGAAAC
4018
GAAACTTCACAGAGGCTTGTCGACGA
4231



TTCACAGAA

CGGCGGTCTCCGTCGTCAGGATCAT




TA








B358_b
GATTTATGA
4019
GGCTTGTCGACGACGGCGGTCTCCGT
4232



GATCAACAG

CGTCAGGATCATCTGTTGATCTC




CAC








B358_c
GATTTATGA
4019
GGCTTGTCGACGACGGCGGTCTCCGT
4233



GATCAACAG

CGTCAGGATCATCTGTTGATCTCAT




CAC








A7_attB_20
GGGAGAGAC
4008
ACGACGGAGACCGCCGTCGTCGACA
4210



GCAAACACA

AGCCAGAGACGCAAA




GC








B8_attB_20
GCAGCTCTC
4016
ACGACGGCGGTCTCCGTCGTCAGGAT
4211



ATTTTCCATA

CATGCTCTCATTTTC




C








A277_c
GACTGAAAC
4018
GAAACTTCACAGAGGCTTGTCGACGA
4231



TTCACAGAA

CGGCGGTCTCCGTCGTCAGGATCAT




TA








B358_c
GATTTATGA
4019
GGCTTGTCGACGACGGCGGTCTCCGT
4233



GATCAACAG

CGTCAGGATCATCTGTTGATCTCAT




CAC








pU6-Sp-gRNA-
GACACCAAA
4020
TACCGTACACCACTGAGACCGCGGTG
4234


IDS_DF_A1_a_attP_
AACTGCCAC

GTTGACCAGACAAACCTGTGGCAGTT



fwd.dna
AGG

A






pU6-Sp-gRNA-
GACACCAAA
4020
GTCTGGTCAACCACCGCGGTCTCAGT
4235


IDS_DF_A1_a_attP_
AACTGCCAC

GGTGTACGGTACAAACCTGTGGCAGT



rev.dna
AGG

TA






pU6-Sp-gRNA-
GACACCAAA
4020
TACCGTACACCACTGAGACCGCGGTG
4236


IDS_DF_A1_c_attP_
AACTGCCAC

GTTGACCAGACAAACCTGTGGCAGTT



fwd.dna
AGG

TTA






pU6-Sp-gRNA-
GACACCAAA
4020
GTCTGGTCAACCACCGCGGTCTCAGT
4237


IDS_DF_A1_c_attP_
AACTGCCAC

GGTGTACGGTACAAACCTGTGGCAGT



rev.dna
AGG

TTTA






pU6-Sp-gRNA-
GCACTCATTT
4021
TACCGTACACCACTGAGACCGCGGTG
4238


IDS_DF_A4_b_attP_
CCTCCAAGCT

GTTGACCAGACAAACCTCTTGGAGGA



fwd.dna
C

AA






pU6-Sp-gRNA-
GCACTCATTT
4021
GTCTGGTCAACCACCGCGGTCTCAGT
4239


IDS_DF_A4_b_attP_
CCTCCAAGCT

GGTGTACGGTACAAACCTCTTGGAGG



rev.dna
C

AAA






pU6-Sp-gRNA-
GTAGGTACA
4022
GTCTGGTCAACCACCGCGGTCTCAGT
4240


IDS_DF_B2_a_attP_
GGACAGGGC

GGTGTACGGTACAAACCTCCCTGTCC



fwd.dna
AG








pU6-Sp-gRNA-
GTAGGTACA
4022
TACCGTACACCACTGAGACCGCGGTG
4241


IDS_DF_B2_a_attP_
GGACAGGGC

GTTGACCAGACAAACCTCCCTGTCC



rev.dna
AG








pU6-Sp-gRNA-
GAGATAGGT
4023
TCTGGTCAACCACCGCGGTCTCAGTG
4242


IDS_DF_B3_a_attP_
AGGTACAGG

GTGTACGGTACAAACCTCCTGTACCT



fwd.dna
ACA








pU6-Sp-gRNA-
GAGATAGGT
4023
TACCGTACACCACTGAGACCGCGGTG
4243


IDS_DF_B3_a_attP_
AGGTACAGG

GTTGACCAGACAAACCTCCTGTACCT



rev.dna
ACA








pU6-Sp-gRNA-
GTGAAAAGA
4024
GTCTGGTCAACCACCGCGGTCTCAGT
4244


IDS_DF_B4_b_attP_
TAGGTAGGT

GGTGTACGGTACAAACCTCCTACCTA



fwd.dna
AC

TCTA






pU6-Sp-gRNA-
GTGAAAAGA
4024
TACCGTACACCACTGAGACCGCGGTG
4245


IDS_DF_B4_b_attP_
TAGGTAGGT

GTTGACCAGACAAACCTCCTACCTAT



rev.dna
AC

CTA






pU6-Sp-gRNA-
GTTATGGTTT
4025
GTCTGGTCAACCACCGCGGTCTCAGT
4246


IDS_DF_B7_b_attP_
ACTCCATCTA

GGTGTACGGTACAAACCTATGGAGTA



fwd.dna


AACCT






pU6-Sp-gRNA-
GTTATGGTTT
4025
TACCGTACACCACTGAGACCGCGGTG
4247


IDS_DF_B7_b_attP_
ACTCCATCTA

GTTGACCAGACAAACCTATGGAGTAA



rev.dna


ACC






pU6-Sp-gRNA-
GTTTTGGTTT
4026
ATGATCCTGACGACGGAGACCGCCGT
4248


IDS_DF_C2_c_attB_
ACCCTATCTA

CGTCGACAAGCCATAGGGTAAACCA



fwd.dna









pU6-Sp-gRNA-
GTTTTGGTTT
4026
GGCTTGTCGACGACGGCGGTCTCCGT
4249


IDS_DF_C2_c_attB_
ACCCTATCTA

CGTCAGGATCATATAGGGTAAACCA



rev.dna









pU6-Sp-gRNA-
GCTGTGGAA
4027
GGCTTGTCGACGACGGCGGTCTCCGT
4250


IDS_DF_D1_b_attB_
CTGCAACAC

CGTCAGGATCATGTGTTGCAGT



fwd.dna
ACT








pU6-Sp-gRNA-
GCTGTGGAA
4027
ATGATCCTGACGACGGAGACCGCCGT
4251


IDS_DF_D1_b_attB_
CTGCAACAC

CGTCGACAAGCCGTGTTGCAGT



rev.dna
ACT








pU6-Sp-gRNA-
GTGCCACCT
4028
GGCTTGTCGACGACGGCGGTCTCCGT
4252


IDS_DF_D2_c_attB_
AACAGTGAG

CGTCAGGATCATCTCACTGTTAGGT



fwd.dna
CTG








pU6-Sp-gRNA-
GTGCCACCT
4028
ATGATCCTGACGACGGAGACCGCCGT
4253


IDS_DF_D2_c_attB_
AACAGTGAG

CGTCGACAAGCCCTCACTGTTAGGT



rev.dna
CTG









All vectors for mammalian cell experiments were purified using Plasmid Plus Midiprep kits (Qiagen) or PureYield plasmid miniprep kits (Promega), or QIAprep Spin Miniprep kits.


General Mammalian Cell Culture Conditions


HEK293T (ATCC CRL-3216), U2OS (ATTC HTB-96), K562 (CCL-243), and HeLa (CCL-2) cells were purchased from ATCC and cultured and passaged in Dulbecco's Modified Eagle's Medium (DMEM) plus GlutaMAX (ThermoFisher Scientific), McCoy's 5A Medium (Gibco), RPMI Medium 1640 plus GlutaMAX (Gibco), or Eagle's Minimal Essential Medium (EMEM, ATCC), respectively, each supplemented with 10% (v/v) fetal bovine serum (Gibco, qualified) and 1× Penicillin Streptomycin (Corning). All cell types were incubated, maintained, and cultured at 37° C. with 5% CO2. Cell lines were authenticated by their respective suppliers and tested negative for mycoplasma.


HEK293T, HeLa, and Huh7 Tissue Culture Transfection Protocol and Genomic DNA Preparation


HEK293T cells grown were seeded on 48-well poly-D-lysine coated plates (Corning). 16-24 h post-seeding, cells were transfected at approximately 60% confluency with 1 μL of Lipofectamine 2000 (Thermo Fisher Scientific) according to the manufacturer's protocols and either: 750 ng of PE2 plasmid, 125 ng of pegRNA 1, and 125 ng of pegRNA 2 (for twinPE transfections); 750 ng of PE2 plasmid, 250 ng of pegRNA plasmid, and 83 ng of sgRNA plasmid (for PE3 transfections); or, 750 ng of Cas9 and 125 ng of sgRNA 1, and 125 ng of sgRNA 2 (for paired Cas9 nuclease transfections). Unless otherwise stated, cells were cultured 3 days following transfection, after which the media was removed, the cells were washed with 1×PBS solution (Thermo Fisher Scientific), and genomic DNA was extracted by the addition of 150 μL of freshly prepared lysis buffer (10 mM Tris-HCl, pH 7.5; 0.05% SDS; 25 μg/mL Proteinase K (ThermoFisher Scientific)) directly into each well of the tissue culture plate. The genomic DNA mixture was incubated at 37° C. for 1-2 hrs, followed by an 80° C. enzyme inactivation step for 30 min. Primers used for mammalian cell genomic DNA amplification are listed in Table 13.









TABLE 13







Sequences of primers used in this Example











SEQ


Primers
Sequence
ID NO:





HEK3_fwd
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNATGTGGGC
3811



TGCCTAGAAAGG






HEK3_rev
TGGAGTTCAGACGTGTGCTCTTCCGATCTCCCAGCCAAACTTGTC
3812



AACC






HEK3_fwd
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNATGTGGGC
3811



TGCCTAGAAAGG






HEK3_rev
TGGAGTTCAGACGTGTGCTCTTCCGATCTCCCAGCCAAACTTGTC
3812



AACC






DMD_UMI_fwd1
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNNNNNNNN
4254



NNNNtgctggccagtttactaacaat






DMD_UMI_fwd2
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNNNNNNNN
4255



NNNNcagaaagaagatcttatcccatcttg






DMD_rev0
TGGAGTTCAGACGTGTGCTCTTCCGATCTggctacttttgttat
4256



ttgcatt






AAVS1_AVA1713
TGGAGTTCAGACGTGTGCTCTTCCGATCTCCAGAGCAGGGTCCC
4257



GCTTC






AAVS1_AVA1717
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNACGGGGCT
4258



CAGTCTGAAGAG






AAVS1_AVA1651
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGCCAAGG
4259



ACTCAAACCCAGA






AAVS1_AVA1652
TGGAGTTCAGACGTGTGCTCTTCCGATCTTCCGTGCGTCAGTTTT
4260



ACCT






AAVS1_AVA1653
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNAACTGCTT
4261



CTCCTCTTGGGAA






AAVS1_AVA1715
TGGAGTTCAGACGTGTGCTCTTCCGATCTTCCTTGCCAGAACCTC
4262



TAAGGT






AAVS1_AVA1655
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNATCCTCTC
4263



TGGCTCCATCGTA






AAVS1_AVA1656
TGGAGTTCAGACGTGTGCTCTTCCGATCTTCCACTTCAGGACAGC
4264



ATGTTT






AAVS1_AVA1707
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNCGCCGGG
4265



AACTGCCGCTGGC






AAVS1_AVA1710
TGGAGTTCAGACGTGTGCTCTTCCGATCTGAGGAGGCCCTCATCT
4266



GGCG






CCR5_AVA1678
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNAATCAATG
4267



TGAAGCAAATCGCAGC






CCR5_AVA1679
TGGAGTTCAGACGTGTGCTCTTCCGATCTTCGATTGTCAGGAGG
4268



ATGATGAA






CCR5_AVA1680
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNTCCTTCTT
4269



ACTGTCCCCTTCTGGGC






CCR5_AVA1681
TGGAGTTCAGACGTGTGCTCTTCCGATCTGCAAACACAGCATGG
4270



ACGAC






CCR5_AVA1682
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNACAATCGA
4271



TAGGTACCTGGCTGTC






CCR5_AVA1683
TGGAGTTCAGACGTGTGCTCTTCCGATCTACCAGCCCCAAGATG
4272



ACTAT






IDS_AVA1763
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNCTGAAAAC
4273



CTGAGCTTGGAGG






IDS_AVA1764
TGGAGTTCAGACGTGTGCTCTTCCGATCTGTCTACTCCAGCTTAA
4274



TGGAAGTGG






IDS_AVA1765
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGAGAAGA
4275



TGTGGAAATGCCTCAC






IDS_AVA1766
TGGAGTTCAGACGTGTGCTCTTCCGATCTAATCAACATGAAGGG
4276



TTGTGTTGT






IDS_AVA1769
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNGTTCCCAC
4277



ACATGCGTTCCTC






IDS_AVA1770
TGGAGTTCAGACGTGTGCTCTTCCGATCTGGCATGAAGGGTTGTT
4278



TTTAATTGA






IDS_UMI_junc1_
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNNNNNNNN
4279


fwd
NNNNGCCCTGTCCTGTACCTACCT






IDS_junc2_fwd
ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNTTCAGTTT
4280



GGGGTATGTGCCA






IDS_universal_
TGGAGTTCAGACGTGTGCTCTTCCGATCTCTCAAATTTACCCGTG
4281


rev
GCAGC









For HeLa cells, they were grown and seeded with a density similar to HEK293T cells and each well of cells were transfected with 190 ng PE2_P2A_Blast (blasticidin resistence gene), 31.5 ng of pegRNA1, and 31.5 ng of pegRNA 2 (for twinPE transfections) with 0.75 uL of HeLa monster reagent (Mirus). Cells were then treated with 10 ug/mL for the selection 24 h after transfection. The rest of genomic DNA collection steps are identical to HEK293T cells. Huh7 cells were seeded 150 k/well on 24-well poly-D-lysine coated plates (Corning). 16-24 h post-seeding, cells were transfected at high confluency with 2 μL of Lipofectamine 2000 (Thermo Fisher Scientific) according to the manufacturer's protocols and up to 800 ng of plasmid DNA. Genomic DNA collection proceeded as above for HEK293T.


High-Throughput DNA Sequencing of Genomic DNA Samples


Genomic sites of interest were amplified from genomic DNA samples and sequenced on an Illumina MiSeq as previously described with the following modifications. Briefly, amplification primers containing Illumina forward and reverse adapters (Table 13) were used for a first round of PCR (PCR 1) amplifying the genomic region of interest. 25-μL PCR 1 reactions were performed with 0.5 μM of each forward and reverse primer, 1 μL of genomic DNA extract and 12.5 μL of Phusion U Green Multiplex PCR Master Mix. PCR reactions were carried out as follows: 98° C. for 2 min, then 30 cycles of [98° C. for 10 s, 61° C. for 20 s, and 72° C. for 30 s], followed by a final 72° C. extension for 2 min. Unique Illumina barcoding primer pairs were added to each sample in a secondary PCR reaction (PCR 2). Specifically, 25 μL of a given PCR 2 reaction contained 0.5 μM of each unique forward and reverse Illumina barcoding primer pair, 1 μL of unpurified PCR 1 reaction mixture, and 12.5 μL of Phusion U Green Multiplex PCR 2× Master Mix. The barcoding PCR 2 reactions were carried out as follows: 98° C. for 2 min, then 12 cycles of [98° C. for 10 s, 61° C. for 20 s, and 72° C. for 30 s], followed by a final 72° C. extension for 2 min. PCR products were evaluated analytically by electrophoresis in a 1.5% agarose gel. PCR 2 products (pooled by common amplicons) were purified by electrophoresis with a 1.5% agarose gel using a QIAquick Gel Extraction Kit (Qiagen), eluting with 40 μL of water. DNA concentration was measured by fluorometric quantification (Qubit, ThermoFisher Scientific) or qPCR (KAPA Library Quantification Kit-Illumina, KAPA Biosystems) and sequenced on an Illumina MiSeq instrument according to the manufacturer's protocols.


Sequencing reads were demultiplexed using MiSeq Reporter (Illumina). Alignment of amplicon sequences to a reference sequence was performed using CRISPResso2. For all prime editing yield quantification, prime editing efficiency was calculated as: % of [# of reads with the desired edit that do not contain indels]÷[# of total reads]. For quantification of point mutation editing, CRISPResso2 was run in standard mode with “discard_indel_reads” on. Prime editing for installation of point mutations was then explicitly calculated as: [frequency of specified point mutation in non-discarded reads]×[# of non-discarded reads]÷[total reads]. For quantification of editing, CRISPResso2 was run in HDR mode using the desired allele as the expected allele (e flag), and with “discard_indel_reads” on. Editing yield was calculated as: [# of non-discarded HDR aligned reads]÷[total reads]. Indel yields were calculated as: [# of indel-containing discarded reads]÷ [total reads]. Unique molecular identifiers (UMIs) were applied to quantify the deletion efficiency and assess PCR bias in this three-step PCR protocol. Briefly, in the first step of linear amplification, 1 uL of genomic DNA extract was linearly amplified by 0.1 uM of single forward UMI primer that contains 15-bp or 16-bp unique molecular identifiers and 12.5 uL Phusion U Green Multiplex PCR Master Mix in a 25-uL reaction. The PCR products were then purified by 1.6× AmPure beads (Beckman Coulter) and eluted in 20 uL QIAGEN elution buffer. In the second step, 1 or 2 uL of linearly amplified PCR eluents were then amplified for 30 cycles with 0.5 uM of each forward and reverse primer, Phusion U Green Multiplex PCR mix in a 25-uL reaction. In this case, the forward primer anneals to the P5 Illumina adaptor sequence that locates at the 5′ of the UMI primer and upstream of UMI. The PCR products were purified by 1× ampure beads and eluted in 25 uL elution buffer. In the third step, 1 uL eluent was amplified for 12 cycles as mentioned above for adding unique Illumina barcoding. To assess the large deletion at DMD locus, the top band (unedited large amplicon) and bottom band (edited amplicons with deletions) were excised separately from the 1.5% agarose and the final library (60 pM of top bands v.s. 20 pM of bottom bands) was loaded into two separate Miseq kits to avoid weak clustering of large amplicons (˜1 kb) compared to short ones (˜300 bp)


Raw sequencing reads were UMI deduplicated using AmpUMI. For paired-end reads, SeqKit was used to concatenate (merge without overlap) R1s with the reverse complement of R2s. The concatenated R1+R2s were UMI deduplicated using the UMI at the 5′ end of R1. UMI-deduplicated R1s or concatenated R1+R2s were analyzed using CRISPResso2. For analyzing concatenated R1+R2s, an appropriate concatenated reference amplicon sequence was provided to minimize sequencing alignment artifacts due to the concatenation.


Nucleofection of U2OS and K562


Nucleofection was performed in all experiments using K562 and U2OS cells. 200,000 cells were used per nucleofection. Counted cells were spun down and washed with PBS and then resuspended in nucleofection solution following the recommendation of Lonza SE Cell Line 4D-Nucleofector Kit. After nucleofection cells in the device, the cells were allowed to incubate in the cuvette at room temperature for 10 minutes. The contents of the cuvette were transferred to a 48 well plate filled with pre-warmed media. Genomic DNA was extracted and prepared for Illumina Miseq preparation as mentioned above.


One-Pot TwinPE and Bxb1-Mediated Cargo Knock-In and Large Inversion:


For the one-pot mediated large inversion experiment, sequential plasmid transfection and one-pot mRNA nucleofection were performed. In the sequential plasmid transfection experiment, HEK293T cells were transfected with 750 ng PE2, 62.5 ng of each pegRNA (four in total) using Lipofectamine 2000 as mentioned above. After three days, cells were trypsinized and plated into 24-well plate and passaged for about seven days. 20K cells were then seeded again and transfected with 500 ng BxBI plasmid. Genomic DNA was extracted and prepared for Illumina Miseq preparation as mentioned above. In the one-pot mRNA nucleofection experiment, 200,000 cells were nucleofected with 1 ug PE2 mRNA, 30 pmol each synthetic pegRNA (4 pegRNAs in total), and 750 ng BxBI mRNA. Synthetic pegRNAs were order from IDT and mRNA transcripts were in vitro transcribed following the published protocol. 20 uL mixture of Lonza buffer and cells were electroporated with program CM-130 and recovered with 80 uL warmed media for five minutes. 25 uL samples from the cuvette was then added to each well of the 48-well plate and incubated at 37° C. for 72 hours.


OTHER REFERENCES MENTIONED THROUGHOUT THIS DISCLOSURE

The following references are each incorporated herein by reference in their entireties.




  • 1. Jinek, M. et al. A Programmable Dual-RNA-Guided DNA Endonuclease in Adaptive Bacterial Immunity. Science 337, 816-821 (2012).

  • 2. Cong, L. et al. Multiplex Genome Engineering Using CRISPR/Cas Systems. Science 339, 819-823 (2013).

  • 3. Komor, A. C., Badran, A. H. & Liu, D. R. CRISPR-Based Technologies for the Manipulation of Eukaryotic Genomes. Cell 168, 20-36 (2017).

  • 4. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420-424 (2016).

  • 5. Nishida, K. et al. Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science 353, aaf8729 (2016).

  • 6. Gaudelli, N. M. et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551, 464-471 (2017).

  • 7. ClinVar, July 2019.

  • 8. Dunbar, C. E. et al. Gene therapy comes of age. Science 359, eaan4672 (2018).

  • 9. Cox, D. B. T., Platt, R. J. & Zhang, F. Therapeutic genome editing: prospects and challenges. Nat. Med. 21, 121-131 (2015).

  • 10. Adli, M. The CRISPR tool kit for genome editing and beyond. Nat. Commun. 9, 1911 (2018).

  • 11. Kleinstiver, B. P. et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 523, 481-485 (2015).

  • 12. Kleinstiver, B. P. et al. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature 529, 490-495 (2016).

  • 13. Hu, J. H. et al. Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature 556, 57-63 (2018).

  • 14. Nishimasu, H. et al. Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science 361, 1259-1262 (2018).

  • 15. Jasin, M. & Rothstein, R. Repair of strand breaks by homologous recombination. Cold Spring Harb. Perspect. Biol. 5, a012740 (2013).

  • 16. Paquet, D. et al. Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature 533, 125-129 (2016).

  • 17. Kosicki, M., Tomberg, K. & Bradley, A. Repair of double-strand breaks induced by CRISPR-Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 36, 765-771 (2018).

  • 18. Haapaniemi, E., Botla, S., Persson, J., Schmierer, B. & Taipale, J. CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response. Nat. Med. 24, 927-930 (2018).

  • 19. Ihry, R. J. et al. p53 inhibits CRISPR-Cas9 engineering in human pluripotent stem cells. Nat. Med. 24, 939-946 (2018).

  • 20. Richardson, C. D., Ray, G. J., DeWitt, M. A., Curie, G. L. & Corn, J. E. Enhancing homology-directed genome editing by catalytically active and inactive CRISPR-Cas9 using asymmetric donor DNA. Nat. Biotechnol. 34, 339-344 (2016).

  • 21. Srivastava, M. et al. An Inhibitor of Nonhomologous End-Joining Abrogates Double-Strand Break Repair and Impedes Cancer Progression. Cell 151, 1474-1487 (2012).

  • 22. Chu, V. T. et al. Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat. Biotechnol. 33, 543-548 (2015).

  • 23. Maruyama, T. et al. Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat. Biotechnol. 33, 538-542 (2015).

  • 24. Kim, Y. B. et al. Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat. Biotechnol. 35, 371-376 (2017).

  • 25. Li, X. et al. Base editing with a Cpf1-cytidine deaminase fusion. Nat. Biotechnol. 36, 324-327 (2018).

  • 26. Gehrke, J. M. et al. An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities. Nat. Biotechnol. (2018). doi:10.1038/nbt.4199

  • 27. Rees, H. A. & Liu, D. R. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 1 (2018). doi:10.1038/s41576-018-0059-1.

  • 28. Ostertag, E. M. & Kazazian Jr, H. H. Biology of Mammalian L1 Retrotransposons. Annu. Rev. Genet. 35, 501-538 (2001).

  • 29. Zimmerly, S., Guo, H., Perlman, P. S. & Lambowltz, A. M. Group II intron mobility occurs by target DNA-primed reverse transcription. Cell 82, 545-554 (1995).

  • 30. Luan, D. D., Korman, M. H., Jakubczak, J. L. & Eickbush, T. H. Reverse transcription of R2Bm RNA is primed by a nick at the chromosomal target site: a mechanism for non-LTR retrotransposition. Cell 72, 595-605 (1993).

  • 31. Feng, Q., Moran, J. V., Kazazian, H. H. & Boeke, J. D. Human L1 retrotransposon encodes a conserved endonuclease required for retrotransposition. Cell 87, 905-916 (1996).

  • 32. Jinek, M. et al. Structures of Cas9 Endonucleases Reveal RNA-Mediated Conformational Activation. Science 343, 1247997 (2014).

  • 33. Jiang, F. et al. Structures of a CRISPR-Cas9 R-loop complex primed for DNA cleavage. Science aad8282 (2016). doi:10.1126/science.aad8282

  • 34. Qi, L. S. et al. Repurposing CRISPR as an RNA-Guided Platform for Sequence-Specific Control of Gene Expression. Cell 152, 1173-1183 (2013).

  • 35. Tang, W., Hu, J. H. & Liu, D. R. Aptazyme-embedded guide RNAs enable ligand-responsive genome editing and transcriptional activation. Nat. Commun. 8, 15939 (2017).

  • 36. Shechner, D. M., Hacisuleyman, E., Younger, S. T. & Rinn, J. L. Multiplexable, locus-specific targeting of long RNAs with CRISPR-Display. Nat. Methods 12, 664-670 (2015).

  • 37. Anders, C. & Jinek, M. Chapter One—In vitro Enzymology of Cas9. in Methods in Enzymology (eds. Doudna, J. A. & Sontheimer, E. J.) 546, 1-20 (Academic Press, 2014).

  • 38. Briner, A. E. et al. Guide RNA Functional Modules Direct Cas9 Activity and Orthogonality. Mol. Cell 56, 333-339 (2014).

  • 39. Nowak, C. M., Lawson, S., Zerez, M. & Bleris, L. Guide RNA engineering for versatile Cas9 functionality. Nucleic Acids Res. 44, 9555-9564 (2016).

  • 40. Sternberg, S. H., Redding, S., Jinek, M., Greene, E. C. & Doudna, J. A. DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature 507, 62-67 (2014).

  • 41. Mohr, S. et al. Thermostable group II intron reverse transcriptase fusion proteins and their use in cDNA synthesis and next-generation RNA sequencing. RNA 19, 958-970 (2013).

  • 42. Stamos, J. L., Lentzsch, A. M. & Lambowitz, A. M. Structure of a Thermostable Group II Intron Reverse Transcriptase with Template-Primer and Its Functional and Evolutionary Implications. Mol. Cell 68, 926-939.e4 (2017).

  • 43. Zhao, C. & Pyle, A. M. Crystal structures of a group II intron maturase reveal a missing link in spliceosome evolution. Nat. Struct. Mol. Biol. 23, 558-565 (2016).

  • 44. Zhao, C., Liu, F. & Pyle, A. M. An ultraprocessive, accurate reverse transcriptase encoded by a metazoan group II intron. RNA 24, 183-195 (2018).

  • 45. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281-2308 (2013).

  • 46. Liu, Y., Kao, H.-I. & Bambara, R. A. Flap endonuclease 1: a central component of DNA metabolism. Annu. Rev. Biochem. 73, 589-615 (2004).

  • 47. Krokan, H. E. & Bjørås, M. Base Excision Repair. Cold Spring Harb. Perspect. Biol. 5, (2013).

  • 48. Kelman, Z. PCNA: structure, functions and interactions. Oncogene 14, 629-640 (1997).

  • 49. Choe, K. N. & Moldovan, G.-L. Forging Ahead through Darkness: PCNA, Still the Principal Conductor at the Replication Fork. Mol. Cell 65, 380-392 (2017).

  • 50. Li, X., Li, J., Harrington, J., Lieber, M. R. & Burgers, P. M. Lagging strand DNA synthesis at the eukaryotic replication fork involves binding and stimulation of FEN-1 by proliferating cell nuclear antigen. J. Biol. Chem. 270, 22109-22112 (1995).

  • 51. Tom, S., Henricksen, L. A. & Bambara, R. A. Mechanism whereby proliferating cell nuclear antigen stimulates flap endonuclease 1. J. Biol. Chem. 275, 10498-10505 (2000).

  • 52. Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S. & Vale, R. D. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635-646 (2014).

  • 53. Bertrand, E. et al. Localization of ASH1 mRNA particles in living yeast. Mol. Cell 2, 437-445 (1998).

  • 54. Dahlman, J. E. et al. Orthogonal gene knockout and activation with a catalytically active Cas9 nuclease. Nat. Biotechnol. 33, 1159-1161 (2015).

  • 55. Tsai, S. Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187-197 (2015).

  • 56. Tsai, S. Q. et al. CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR-Cas9 nuclease off-targets. Nat. Methods 14, 607-614 (2017).

  • 57. Schek N, Cooke C, Alwine J C. Molecular and Cellular Biology. (1992).

  • 58. Gil A, Proudfoot N J. Cell. (1987).

  • 59. Zhao, B. S., Roundtree, I. A., He, C. Nat Rev Mol Cell Biol. (2017).

  • 60. Rubio, M. A. T., Hopper, A. K. Wiley Interdiscip Rev RNA (2011).

  • 61. Shechner, D. M., Hacisuleyman E, Younger, S. T., Rinn, J. L. Nat Methods. (2015).

  • 62. Paige, J. S., Wu, K. Y., Jaffrey, S. R. Science (2011).

  • 63. Ray D., . . . Hughes T R. Nature (2013).

  • 64. Chadalavada, D. M., Cerrone-Szakal, A. L., Bevilacqua, P. C. RNA (2007).

  • 65. Forster A C, Symons R H. Cell. (1987).

  • 66. Weinberg Z, Kim P B, Chen T H, Li S, Harris K A, Lünse C E, Breaker R R. Nat. Chem. Biol. (2015).

  • 67. Feldstein P A, Buzayan J M, Bruening G. Gene (1989).

  • 68. Saville B J, Collins R A. Cell. (1990).

  • 69. Winkler W C, Nahvi A, Roth A, Collins J A, Breaker R R. Nature (2004).

  • 70. Roth A, Weinberg Z, Chen A G, Kim P G, Ames T D, Breaker R R. Nat Chem Biol. (2013).

  • 71. Choudhury R, Tsai Y S, Dominguez D, Wang Y, Wang Z. Nat Commun. (2012).

  • 72. MacRae I J, Doudna J A. Curr Opin Struct Biol. (2007).

  • 73. Bernstein E, Caudy A A, Hammond S M, Hannon G J Nature (2001).

  • 74. Filippov V, Solovyev V, Filippova M, Gill S S. Gene (2000).

  • 75. Cadwell R C and Joyce G F. PCR Methods Appl. (1992).

  • 76. McInerney P, Adams P, and Hadi M Z. Mol Biol Int. (2014).

  • 77. Esvelt K M, Carlson J C, and Liu D R. Nature. (2011).

  • 78. Naorem S S, Hin J, Wang S, Lee W R, Heng X, Miller J F, Guo H. Proc Natl Acad Sci USA (2017).

  • 79. Martinez M A, Vartanian J P, Wain-Hobson S. Proc Natl Acad Sci USA (1994).

  • 80. Meyer A J, Ellefson J W, Ellington A D. Curr Protoc Mol Biol. (2014).

  • 81. Wang H H, Isaacs F J, Carr P A, Sun Z Z, Xu G, Forest C R, Church G M. Nature. (2009).

  • 82. Nyerges A et al. Proc Natl Acad Sci USA. (2016).

  • 83. Mascola J R, Haynes B F. Immunol Rev. (2013).

  • 84. X. Wen, K. Wen, D. Cao, G. Li, R. W. Jones, J. Li, S. Szu, Y. Hoshino, L. Yuan, Inclusion of a universal tetanus toxoid CD4(+) T cell epitope P2 significantly enhanced the immunogenicity of recombinant rotavirus ΔVP8* subunit parenteral vaccines. Vaccine 32, 4420-4427 (2014).

  • 85. G. Ada, D. Isaacs, Carbohydrate-protein conjugate vaccines. Clin Microbiol Infect 9, 79-85 (2003).

  • 86. E. Malito, B. Bursulaya, C. Chen, P. L. Surdo, M. Picchianti, E. Balducci, M. Biancucci, A. Brock, F. Berti, M. J. Bottomley, M. Nissum, P. Costantino, R. Rappuoli, G. Spraggon, Structural basis for lack of toxicity of the diphtheria toxin mutant CRM197. Proceedings of the National Academy of Sciences 109, 5229 (2012).

  • 87. J. de Wit, M. E. Emmelot, M. C. M. Poelen, J. Lanfermeijer, W. G. H. Han, C. van Els, P. Kaaijk, The Human CD4(+) T Cell Response against Mumps Virus Targets a Broadly Recognized Nucleoprotein Epitope. J Virol 93, (2019).

  • 88. M. May, C. A. Rieder, R. J. Rowe, Emergent lineages of mumps virus suggest the need for a polyvalent vaccine. Int J Infect Dis 66, 1-4 (2018).

  • 89. M Ramamurthy, P. Rajendiran, N. Saravanan, S. Sankar, S. Gopalan, B. Nandagopal, Identification of immunogenic B-cell epitope peptides of rubella virus E1 glycoprotein towards development of highly specific immunoassays and/or vaccine. Conference Abstract, (2019).

  • 90. U. S. F. Tambunan, F. R. P. Sipahutar, A. A. Parikesit, D. Kerami, Vaccine Design for H5N1 Based on B- and T-cell Epitope Predictions. Bioinform Biol Insights 10, 27-35 (2016).

  • 91. Asante, E A. et. al. “A naturally occurring variant of the human prion protein completely prevents prion disease”. Nature. (2015).

  • 92. Crabtree, G. R. & Schreiber, S. L. Three-part inventions: intracellular signaling and induced proximity Trends Biochem. Sci. 21, 418-22 (1996).

  • 93. Liu, J. et al. Calcineurin Is a Common Target of A and FKBP-FK506 Complexes. Cell 66, 807-815 (1991).

  • 94. Keith, C. T. et al. A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature 369, 756-758 (2003).

  • 95. Spencer, D. M., Wandless, T. J., Schreiber, S. L. S. & Crabtree, G. R. Controlling signal transduction with synthetic ligands. Science 262, 1019-24 (1993).

  • 96. Pruschy, M. N. et al. Mechanistic studies of a signaling pathway activated by the organic dimerizer FK1012. Chem. Biol. 1, 163-172 (1994).

  • 97. Spencer, D. M. et al. Functional analysis of Fas signaling in vivo using synthetic inducers of dimerization. Curr. Biol. 6, 839-847 (1996).

  • 98. Belshaw, P. J., Spencer, D. M., Crabtree, G. R. & Schreiber, S. L. Controlling programmed cell death with a cyclophilin-cyclosporin-based chemical inducer of dimerization. Chem. Biol. 3, 731-738 (1996).

  • 99. Yang, J. X., Symes, K., Mercola, M. & Schreiber, S. L Small-molecule control of insulin and PDGF receptor signaling and the role of membrane attachment. Curr. Biol. 8, 11-18 (1998).

  • 100. Belshaw, P. J., Ho, S. N., Crabtree, G. R. & Schreiber, S. L. Controlling protein association and subcellular localization with a synthetic ligand that induces heterodimerization of proteins. Proc. Natl. Acad. Sci. 93, 4604-4607 (2002).

  • 101. Stockwell, B. R. & Schreiber, S. L. Probing the role of homomeric and heteromeric receptor interactions in TGF-β signaling using small molecule dimerizers. Curr. Biol. 8, 761-773 (2004).

  • 102. Spencer, D. M., Graef, I., Austin, D. J., Schreiber, S. L. & Crabtree, G. R. A general strategy for producing conditional alleles of Src-like tyrosine kinases. Proc. Natl. Acad. Sci. 92, 9805-9809 (2006).

  • 103. Holsinger, L. J., Spencer, D. M., Austin, D. J., Schreiber, S. L. & Crabtree, G. R. Signal transduction in T lymphocytes using a conditional allele of Sos. Proc. Natl. Acad. Sci. 92, 9810-9814 (2006).

  • 104. Myers, M. G. Insulin Signal Transduction and the IRS Proteins. Annu. Rev. Pharmacol. Toxicol.

  • 36, 615-658 (1996).

  • 105. Watowich, S. S. The erythropoietin receptor: Molecular structure and hematopoietic signaling pathways. J. Investig. Med. 59, 1067-1072 (2011).

  • 106. Blau, C. A., Peterson, K. R., Drachman, J. G. & Spencer, D. M. A proliferation switch for genetically modified cells. Proc. Natl. Acad. Sci. 94, 3076-3081 (2002).

  • 107. Clackson, T. et al. Redesigning an FKBP-ligand interface to generate chemical dimerizers with novel specificity. Proc. Natl. Acad. Sci. 95, 10437-10442 (1998).

  • 108. Diver, S. T. & Schreiber, S. L. Single-step synthesis of cell-permeable protein dimerizers that activate signal transduction and gene expression. J. Am. Chem. Soc. 119, 5106-5109 (1997).

  • 109. Guo, Z. F., Zhang, R. & Liang, F. Sen. Facile functionalization of FK506 for biological studies by the thiol-ene ‘click’ reaction. RSC Adv. 4, 11400-11403 (2014).

  • 110. Robinson, D. R., Wu, Y.-M. & Lin, S.-F. The protein tyrosine kinase family of the human genome. Oncogene 19, 5548-5557 (2000).

  • 111. Landrum, M. J. et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 44, D862-D868 (2016).

  • 112. Jinek, M. et al. A Programmable Dual-RNA-Guided DNA Endonuclease in Adaptive Bacterial Immunity. Science 337, 816-821 (2012).

  • 113. Cong, L. et al. Multiplex Genome Engineering Using CRISPR/Cas Systems. Science 339, 819-823 (2013).

  • 114. Mali, P. et al. RNA-Guided Human Genome Engineering via Cas9. Science 339, 823-826 (2013).

  • 115. Yang, H. et al. One-Step Generation of Mice Carrying Reporter and Conditional Alleles by CRISPR/Cas-Mediated Genome Engineering. Cell 154, 1370-1379 (2013).

  • 116. Kim, S., Kim, D, Cho, S. W., Kim, J. & Kim, J.-S. Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res. 24, 1012-1019 (2014).

  • 117. Orlando, S. J. et al. Zinc-finger nuclease-driven targeted integration into mammalian genomes using donors with limited chromosomal homology. Nucleic Acids Res. 38, e152-e152 (2010).

  • 118. Tsai, S. Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187-197 (2015).

  • 119. Suzuki, K. et al. In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature 540, 144-149 (2016).

  • 120. Kosicki, M., Tomberg, K. & Bradley, A. Repair of double-strand breaks induced by CRISPR-Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 36, 765-771 (2018).

  • 121. Haapaniemi, E., Botla, S., Persson, J., Schmierer, B. & Taipale, J. CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response. Nat. Med. 24, 927-930 (2018).

  • 122. Ihry, R. J. et al. p53 inhibits CRISPR-Cas9 engineering in human pluripotent stem cells. Nat. Med. 24, 939-946 (2018).

  • 123. Chapman, J. R., Taylor, M. R. G. & Boulton, S. J. Playing the end game: DNA double-strand break repair pathway choice. Mol. Cell 47, 497-510 (2012).

  • 124. Cox, D. B. T., Platt, R. J. & Zhang, F. Therapeutic genome editing: prospects and challenges. Nat. Med. 21, 121-131 (2015).

  • 125. Paquet, D. et al. Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature 533, 125-129 (2016).

  • 126. Chu, V. T. et al. Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat. Biotechnol. 33, 543-548 (2015).

  • 127. Maruyama, T. et al. Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat. Biotechnol. 33, 538-542 (2015).

  • 128. Rees, H. A., Yeh, W.-H. & Liu, D. R. Development of hRad51-Cas9 nickase fusions that mediate HDR without double-stranded breaks. Nat. Commun. 10, 1-12 (2019).

  • 129. Komor, A. C., Kim, Y. B., Packer, M. S., Zuris, J. A. & Liu, D. R. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420-424 (2016).

  • 130. Gaudelli, N. M. et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551, 464-471 (2017).

  • 131. Gao, X. et al. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature 553, 217-221 (2018).

  • 132. Ingram, V. M. A specific chemical difference between the globins of normal human and sickle-cell anaemia haemoglobin. Nature 178, 792-794 (1956).

  • 133. Myerowitz, R. & Costigan, F. C. The major defect in Ashkenazi Jews with Tay-Sachs disease is an insertion in the gene for the alpha-chain of beta-hexosaminidase. J. Biol. Chem. 263, 18587-18589 (1988).

  • 134. Zielenski, J. Genotype and Phenotype in Cystic Fibrosis. Respiration 67, 117-133 (2000).

  • 135. Mead, S. et al. A Novel Protective Prion Protein Variant that Colocalizes with Kuru Exposure. N. Engl. J. Med. 361, 2056-2065 (2009).

  • 136. Marraffini, L. A. & Sontheimer, E. J. CRISPR interference limits horizontal gene transfer in staphylococci by targeting DNA. Science 322, 1843-1845 (2008).

  • 137. Barrangou, R. et al. CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709-1712 (2007).

  • 138. Jiang, F. & Doudna, J. A. CRISPR-Cas9 Structures and Mechanisms. Annu. Rev. Biophys. 46, 505-529 (2017).

  • 139. Hille, F. et al. The Biology of CRISPR-Cas: Backward and Forward. Cell 172, 1239-1259 (2018).

  • 140. Luan, D. D., Korman, M. H., Jakubczak, J. L. & Eickbush, T. H. Reverse transcription of R2Bm RNA is primed by a nick at the chromosomal target site: a mechanism for non-LTR retrotransposition. Cell 72, 595-605 (1993).

  • 141. Liu, Y., Kao, H.-I. & Bambara, R. A. Flap endonuclease 1: a central component of DNA metabolism. Annu. Rev. Biochem. 73, 589-615 (2004).

  • 142. Rees, H. A. & Liu, D. R. Base editing: precision chemistry on the genome and transcriptome of living cells. Nat. Rev. Genet. 19, 770 (2018).

  • 143. Richardson, C. D., Ray, G. J., DeWitt, M. A., Curie, G. L. & Corn, J. E. Enhancing homology-directed genome editing by catalytically active and inactive CRISPR-Cas9 using asymmetric donor DNA. Nat. Biotechnol. 34, 339-344 (2016).

  • 144. Qi, L. S. et al. Repurposing CRISPR as an RNA-Guided Platform for Sequence-Specific Control of Gene Expression. Cell 152, 1173-1183 (2013).

  • 145. Shechner, D. M., Hacisuleyman, E., Younger, S. T. & Rinn, J. L. Multiplexable, locus-specific targeting of long RNAs with CRISPR-Display. Nat. Methods 12, 664-670 (2015).

  • 146. Tang, W., Hu, J. H. & Liu, D. R. Aptazyme-embedded guide RNAs enable ligand-responsive genome editing and transcriptional activation. Nat. Commun. 8, 15939 (2017).

  • 147. Jinek, M. et al. Structures of Cas9 Endonucleases Reveal RNA-Mediated Conformational Activation. Science 343, 1247997 (2014).

  • 148. Nishimasu, H. et al. Crystal Structure of Cas9 in Complex with Guide RNA and Target DNA. Cell 156, 935-949 (2014).

  • 149. Jiang, F., Zhou, K., Ma, L., Gressel, S. & Doudna, J. A. A Cas9-guide RNA complex preorganized for target DNA recognition. Science 348, 1477-1481 (2015).

  • 150. Baranauskas, A. et al. Generation and characterization of new highly thermostable and processive M-MuLV reverse transcriptase variants. Protein Eng. Des. Sel. 25, 657-668 (2012).

  • 151. Gerard, G. F. et al. The role of template-primer in protection of reverse transcriptase from thermal inactivation. Nucleic Acids Res. 30, 3118-3129 (2002).

  • 152. Arezi, B. & Hogrefe, H. Novel mutations in Moloney Murine Leukemia Virus reverse transcriptase increase thermostability through tighter binding to template-primer. Nucleic Acids Res. 37, 473-481 (2009).

  • 153. Kotewicz, M. L., Sampson, C. M., D'Alessio, J. M. & Gerard, G. F. Isolation of cloned Moloney murine leukemia virus reverse transcriptase lacking ribonuclease H activity. Nucleic Acids Res. 16, 265-277 (1988).

  • 154. Shen, M. W. et al. Predictable and precise template-free CRISPR editing of pathogenic variants. Nature 563, 646-651 (2018).

  • 155. Thuronyi, B. W. et al. Continuous evolution of base editors with expanded target compatibility and improved activity. Nat. Biotechnol. (2019). doi:10.1038/s41587-019-0193-0

  • 156. Kim, Y. B. et al. Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat. Biotechnol. 35, 371-376 (2017).

  • 157. Koblan, L. W. et al. Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat. Biotechnol. (2018). doi:10.1038/nbt.4172

  • 158. Komor, A. C. et al. Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T:A base editors with higher efficiency and product purity. Sci. Adv. 3, eaao4774 (2017).

  • 159. Kleinstiver, B. P. et al. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature 529, 490-495 (2016).

  • 160. Zuo, E. et al. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science 364, 289-292 (2019).

  • 161. Jin, S. et al. Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science 364, 292-295 (2019).

  • 162. Kim, D., Kim, D., Lee, G., Cho, S.-I. & Kim, J.-S. Genome-wide target specificity of CRISPR RNA-guided adenine base editors. Nat. Biotechnol. 37, 430-435 (2019).

  • 163. Grunewald, J. et al. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 569, 433-437 (2019).

  • 164. Zhou, C. et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature 571, 275-278 (2019).

  • 165. Rees, H. A., Wilson, C., Doman, J. L. & Liu, D. R. Analysis and minimization of cellular RNA editing by DNA adenine base editors. Sci. Adv. 5, eaax5717 (2019).

  • 166. Ostertag, E. M. & Kazazian Jr, H. H. Biology of Mammalian L1 Retrotransposons. Annu. Rev. Genet. 35, 501-538 (2001).

  • 167. Griffiths, D. J. Endogenous retroviruses in the human genome sequence. Genome Biol. 2, REVIEWS1017 (2001).

  • 168. Berkhout, B., Jebbink, M. & Zsíros, J. Identification of an Active Reverse Transcriptase Enzyme Encoded by a Human Endogenous HERV-K Retrovirus. J. Virol. 73, 2365-2375 (1999).

  • 169. Halvas, E. K., Svarovskaia, E. S. & Pathak, V. K. Role of Murine Leukemia Virus Reverse Transcriptase Deoxyribonucleoside Triphosphate-Binding Site in Retroviral Replication and In Vivo Fidelity. J. Virol. 74, 10349-10358 (2000).

  • 170. Dever, D. P. et al. CRISPR/Cas9 Beta-globin Gene Targeting in Human Hematopoietic Stem Cells. Nature 539, 384-389 (2016).

  • 171. Park, S. H. et al. Highly efficient editing of the β-globin gene in patient-derived hematopoietic stem and progenitor cells to treat sickle cell disease. Nucleic Acids Res. doi:10.1093/nar/gkz475

  • 172. Collinge, J. Prion diseases of humans and animals their causes and molecular basis. Annu. Rev. Neurosci. 24, 519-550 (2001).

  • 173. Asante, E. A. et al. A naturally occurring variant of the human prion protein completely prevents prion disease. Nature 522, 478-481 (2015).

  • 174. Anzalone, A. V., Lin, A. J., Zairis, S., Rabadan, R. & Cornish, V. W. Reprogramming eukaryotic translation with ligand-responsive synthetic RNA switches. Nat. Methods 13, 453-458 (2016).

  • 175. Badran, A. H. et al. Continuous evolution of Bacillus thuringiensis toxins overcomes insect resistance. Nature 533, 58-63 (2016).

  • 176. Anders, C. & Jinek, M. Chapter One—In Vitro Enzymology of Cas9. in Methods in Enzymology (eds. Doudna, J. A. & Sontheimer, E. J.) 546, 1-20 (Academic Press, 2014).

  • 177. Pirakitikulr, N., Ostrov, N., Peralta-Yahya, P. & Cornish, V. W. PCRless library mutagenesis via oligonucleotide recombination in yeast. Protein Sci. Publ. Protein Soc. 19, 2336-2346 (2010).

  • 178. Clement, K. et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat. Biotechnol. 37, 224-226 (2019).

  • 179. Tsai, S. Q. et al. GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol. 33, 187-197 (2015).

  • 180. Kleinstiver, B. P. et al. High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature 529, 490-495 (2016).

  • 181. Koblan, L. W. et al. Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat. Biotechnol. (2018). doi:10.1038/nbt.4172

  • 182. Baranauskas, A. et al. Generation and characterization of new highly thermostable and processive M-MuLV reverse transcriptase variants. Protein Eng. Des. Sel. 25, 657-668 (2012).

  • 183. Schechner, D M, Hacisuleyman E., Younger S T, Rinn J L. Nat Methods 664-70 (2015).

  • 184. Brown J A, et al. Nat Struct Mol Biol 633-40 (2014).

  • 185. Conrad N A and Steitz J A. EMBO J 1831-41 (2005).

  • 186. Bartlett J S, et al. Proc Natl Acad Sci USA 8852-7 (1996).

  • 187. Mitton-Fry R M, DeGregorio S J, Wang J, Steitz T A, Steitz J A. Science 1244-7 (2010).

  • 188. Forster A C, Symons R H. Cell. 1987.

  • 189. Weinberg Z, Kim P B, Chen T H, Li S, Harris K A, Lünse C E, Breaker R R. Nat. Chem. Biol. 2015.

  • 190. Feldstein P A, Buzayan J M, Bruening G. Gene 1989.

  • 191. Saville B J, Collins R A. Cell. 1990.

  • 192. Roth A, Weinberg Z, Chen A G, Kim P G, Ames T D, Breaker R R. Nat Chem Biol. 2013.

  • 193. Borchardt E K, et al. RNA 1921-30 (2015).

  • 194. Zhang Y, et al. Mol Cell 792-806 (2013).

  • 195. Dang Y, et al. Genome Biol 280 (2015).

  • 196. Schaefer M, Kapoor U, and Jantsch M F. Open Biol 170077 (2017).

  • 197. Nahar S, et al. Chem Comm 2377-80 (2018).

  • 198. Gao Y and Zhao Y. J Integr Plant Biol 343-9 (2014).

  • 199. Dubois N, Marquet R, Paillart J, Bernacchi S. Front Microbiol 527 (2018).

  • 200. Costa M and Michel F. EMBO J 1276-85 (1995).

  • 201. Hu J H, et al. Nature 57-63 (2018).

  • 202. Furukawa K, Gu H, Breaker R R. Methods Mol Biol 209-20 (2014).

  • 203. Zettler, J., Schutz, V. & Mootz, H. D. The naturally split Npu DnaE intein exhibits an extraordinarily high rate in the protein trans-splicing reaction. FEBS Lett. 583, 909-914 (2009).

  • 204. Kugler, S., Kilic, E. & Bahr, M. Human synapsin 1 gene promoter confers highly neuron-specific long-term transgene expression from an adenoviral vector in the adult rat brain depending on the transduced area. Gene Ther. 10, 337-347 (2003).

  • 205. de Felipe, P., Hughes, L. E., Ryan, M. D. & Brown, J. D. Co-translational, intraribosomal cleavage of polypeptides by the foot-and-mouth disease virus 2A peptide. J. Biol. Chem. 278, 11441-11448 (2003).

  • 206. Levy, J. M. & Nicoll, R. A. Membrane-associated guanylate kinase dynamics reveal regional and developmental specificity of synapse stability. J. Physiol. 595, 1699-1709 (2017).

  • 207. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12,323 (2011).

  • 208. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47-e47 (2015).



Equivalents and Scope

In the articles such as “a,” “an,” and “the” may mean one or more than one unless indicated to the contrary or otherwise evident from the context. Embodiments or descriptions that include “or” between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The invention includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The invention includes embodiments in which more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process.


Furthermore, the disclosure encompasses all variations, combinations, and permutations in which one or more limitations, elements, clauses, and descriptive terms from one or more of the listed claims is introduced into another claim. For example, any claim that is dependent on another claim can be modified to include one or more limitations found in any other claims that is dependent on the same base claim. Where elements are presented as lists, e.g., in Markush group format, each subgroup of the elements is also disclosed, and any element(s) can be removed from the group. It should it be understood that, in general, where the invention, or aspects of the invention, is/are referred to as comprising particular elements and/or features, certain embodiments of the disclosure or aspects of the disclosure consist, or consist essentially of, such elements and/or features. For purposes of simplicity, those embodiments have not been specifically set forth in haec verba herein. It is also noted that the terms “comprising” and “containing” are intended to be open and permits the inclusion of additional elements or steps. Where ranges are given, endpoints are included. Furthermore, unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or sub-range within the stated ranges in different embodiments of the invention, to the tenth of the unit of the lower limit of the range, unless the context clearly dictates otherwise.


This application refers to various issued patents, published patent applications, journal articles, and other publications, all of which are incorporated herein by reference. If there is a conflict between any of the incorporated references and the instant specification, the specification shall control. In addition, any particular embodiment of the present invention that falls within the prior art may be explicitly excluded from any one or more of the embodiments. Because such embodiments are deemed to be known to one of ordinary skill in the art, they may be excluded even if the exclusion is not set forth explicitly herein. Any particular embodiment of the invention can be excluded from any embodiment, for any reason, whether or not related to the existence of prior art.


Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation many equivalents to the specific embodiments described herein. The scope of the present embodiments described herein is not intended to be limited to the above Description, but rather is as set forth in the appended embodiments. Those of ordinary skill in the art will appreciate that various changes and modifications to this description may be made without departing from the spirit or scope of the present invention, as defined in the following embodiments.

Claims
  • 1. A system for simultaneously editing both strands of a double-stranded DNA sequence at a target site to be edited, said system comprising: (a) a prime editor or one or more polynucleotides encoding the prime editor, wherein the prime editor comprises a nucleic acid programmable DNA binding protein (napDNAbp) and a polypeptide comprising an RNA-dependent DNA polymerase activity, wherein the napDNAbp comprises a RuvC nuclease domain and a HNH nuclease domain, and wherein the HNH nuclease domain comprises one or more mutations that decrease or eliminate its nuclease activity;(b) a first prime editing guide RNA (first PEgRNA) or one or more polynucleotides encoding the first PEgRNA, wherein the first PEgRNA comprises (i) a first spacer sequence that is complementary to a first binding site on a first strand of the double-stranded DNA sequence upstream of the target site relative to the second strand,(ii) a first gRNA core that is capable of complexing with the prime editor,(iii) a first DNA synthesis template that encodes a first single-stranded DNA sequence, and(iv) a first primer binding site complementary to a region of the second strand upstream of a first cut site; and(c) a second prime editing guide RNA (second PEgRNA) or one or more polynucleotides encoding the second PEgRNA, wherein the second PEgRNA comprises (i) a second spacer sequence that is complementary to a second binding site on a second strand of the double-stranded DNA sequence downstream of the target site relative to the second strand;(ii) a second gRNA core that is capable of complexing with the prime editor,(iii) a second DNA synthesis template that encodes a second single-stranded DNA sequence, and(iv) a second primer binding site complementary to a region of the first strand upstream of a second cut site;wherein the prime editor is capable of cleaving the first strand at the a-first cut site when complexed with the second PEgRNA and cleaving the second strand at the a-second cut site when complexed with the first PEgRNA,wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence are reverse complements over a region of complementarity of each single-stranded DNA sequence of
  • 2. The system of claim 1, wherein the second single-stranded DNA sequence comprises a second edit compared to the first strand of the target site that starts at a position no more than 3 nucleotides from the second cut site.
  • 3. The system of claim 2, wherein the region of complementarity of the first single-stranded DNA sequence and the second single-stranded DNA sequence forms a duplex comprising the first edit and the second edit.
  • 4. The system of claim 2, wherein the second edit comprises an insertion, a deletion, a substitution, or a combination thereof.
  • 5. The system of claim 2, wherein the first edit or the second edit comprises a recombinase recognition sequence.
  • 6. The system of claim 5, further comprising a recombinase capable of recognizing the recombinase recognition sequence, or one or more polynucleotides encoding the recombinase.
  • 7. The system of claim 6, wherein the recombinase is a serine recombinase.
  • 8. The system of claim 6, wherein the recombinase is Bxb1.
  • 9. The system of claim 6, wherein the recombinase recognition sequence comprises SEQ ID NO: 536 or SEQ ID NO: 537.
  • 10. The system of claim 6 further comprising a donor template or a polynucleotide encoding the donor template.
  • 11. The system of claim 10, wherein the recombinase recognition sequence comprises SEQ ID NO: 536 and the donor template comprises SEQ ID NO: 537, or wherein the recombinase recognition sequence comprises SEQ ID NO: 537 and the donor template comprises SEQ ID NO: 536.
  • 12. The system of claim 2, wherein the second edit starts at a position no more than 2 nucleotides from the second cut site.
  • 13. The system of claim 2, wherein the second edit starts at the second cut site.
  • 14. The system of claim 1, wherein the region of complementarity encompasses the 3′ ends of the first and second single-stranded DNA sequences.
  • 15. The system of claim 14, wherein the region of complementarity is from 22 to 38 nucleotides in length.
  • 16. The system of claim 15, wherein the first edit comprises a recombinase recognition sequence.
  • 17. The system of claim 16, wherein the recombinase recognition sequence comprises SEQ ID NO: 536 or SEQ ID NO: 537.
  • 18. The system of claim 1, wherein the region of complementarity is at least 10 nucleotides in length.
  • 19. The system of claim 1, wherein the region of complementarity is from 22 to 38 nucleotides in length.
  • 20. The system of claim 14, wherein the region of complementarity is at least 10 nucleotides in length.
  • 21. The system of claim 1, wherein the first single-stranded DNA sequence is the reverse complement of the second single-stranded DNA sequence.
  • 22. The system of claim 1, wherein the first edit comprises an insertion, a deletion, a substitution, or a combination thereof.
  • 23. The system of claim 1, wherein the first single-stranded DNA sequence and the second single-stranded DNA sequence have the same length.
  • 24. The system of claim 1, wherein the first edit starts at a position no more than 2 nucleotides from the first cut site.
  • 25. The system of claim 1, wherein the first edit starts at the first cut site.
  • 26. The system of claim 1, wherein the first single-stranded DNA sequence and/or the second single-stranded DNA sequence does not comprise sequence homology as compared to the DNA sequence at the target site.
  • 27. The system of claim 1, wherein the napDNAbp comprises an amino acid sequence of any one of SEQ ID NOs: 18, 21, 23, 25-39, 42-61, 75, and 77-88, or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity with any one of SEQ ID NOs: 18, 21, 23, 25-39, 42-61, 75, and 77-88.
  • 28. The system of claim 1, wherein the polypeptide comprising the RNA-dependent DNA polymerase activity is a reverse transcriptase.
  • 29. The system of claim 1, wherein the polypeptide comprising the RNA-dependent DNA polymerase activity comprises an amino acid sequence of any one of SEQ ID NOs: 89-100, 106-122, 128, 132, 139, 143, 149, 154, 159, 700-736, 738-742, and 763-766, or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% sequence identity with any one of SEQ ID NOs: 89-100, 106-122, 128, 132, 139, 143, 149, 154, 159, 700-736, 738-742, and 763-766.
RELATED APPLICATIONS AND INCORPORATION BY REFERENCE

This application claims priority under 35 U.S.C. §§ 120 and 365(c) to and is a continuation of international PCT Application, PCT/US2021/031439, filed on May 7, 2021, which claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Application, U.S. Ser. No. 63/022,397, filed on May 8, 2020, and U.S. Provisional Application U.S. Ser. No. 63/116,785 filed on Nov. 20, 2020, each of which is incorporated herein by reference. This U.S. Provisional Application also refers to and incorporates by reference the following applications, namely, U.S. Provisional Application No. 62/820,813, filed Mar. 19, 2019, U.S. Provisional Application No. 62/858,958, filed Jun. 7, 2019, U.S. Provisional Application No. 62/889,996, filed Aug. 21, 2019, U.S. Provisional Application No. 62/922,654, filed Aug. 21, 2019, U.S. Provisional Application No. 62/913,553, filed Oct. 10, 2019, U.S. Provisional Application No. 62/973,558, filed Oct. 10, 2019, U.S. Provisional Application No. 62/931,195, filed Nov. 5, 2019, U.S. Provisional Application No. 62/944,231, filed Dec. 5, 2019, U.S. Provisional Application No. 62/974,537, filed Dec. 5, 2019, U.S. Provisional Application No. 62/991,069, filed Mar. 17, 2020, and U.S. Provisional Application No. 63/100,548, filed Mar. 17, 2020. In addition, this U.S. Provisional Application refers to and incorporates by reference International PCT Application Nos.: PCT/US20/23721; PCT/US20/23730; PCT/US20/23713; PCT/US20/23712; PCT/US20/23727; PCT/US20/23724; PCT/US20/23725; PCT/US20/23728; PCT/US20/23732; PCT/US20/23723; PCT/US20/23553; and PCT/US20/23583, each filed on Mar. 19, 2020.

GOVERNMENT SUPPORT

This invention was made with government support under grant numbers U01AI142756, RM1HG009490, R01EB022376, and R35GM118062 awarded by the National Institutes of Health. The government has certain rights in the invention.

US Referenced Citations (601)
Number Name Date Kind
4182449 Kozlow Jan 1980 A
4186183 Steck et al. Jan 1980 A
4217344 Vanlerberghe et al. Aug 1980 A
4235871 Papahadjopoulos et al. Nov 1980 A
4261975 Fullerton et al. Apr 1981 A
4485054 Mezei et al. Nov 1984 A
4501728 Geho et al. Feb 1985 A
4663290 Weis et al. May 1987 A
4737323 Martin et al. Apr 1988 A
4774085 Fidler Sep 1988 A
4797368 Carter et al. Jan 1989 A
4837028 Allen Jun 1989 A
4873316 Meade et al. Oct 1989 A
4880635 Janoff et al. Nov 1989 A
4889818 Gelfand et al. Dec 1989 A
4897355 Eppstein et al. Jan 1990 A
4906477 Kurono et al. Mar 1990 A
4911928 Wallach Mar 1990 A
4917951 Wallach Apr 1990 A
4920016 Allen et al. Apr 1990 A
4921757 Wheatley et al. May 1990 A
4946787 Eppstein et al. Aug 1990 A
4965185 Grischenko et al. Oct 1990 A
5017492 Kotewicz et al. May 1991 A
5047342 Chatterjee Sep 1991 A
5049386 Eppstein et al. Sep 1991 A
5079352 Gelfand et al. Jan 1992 A
5139941 Muzyczka et al. Aug 1992 A
5173414 Lebkowski et al. Dec 1992 A
5223409 Ladner et al. Jun 1993 A
5244797 Kotewicz et al. Sep 1993 A
5270179 Chatterjee Dec 1993 A
5374553 Gelfand et al. Dec 1994 A
5405776 Kotewicz et al. Apr 1995 A
5436149 Barnes Jul 1995 A
5449639 Wei et al. Sep 1995 A
5496714 Comb et al. Mar 1996 A
5512462 Cheng Apr 1996 A
5580737 Polisky et al. Dec 1996 A
5614365 Tabor et al. Mar 1997 A
5652094 Usman et al. Jul 1997 A
5658727 Barbas et al. Aug 1997 A
5668005 Kotewicz et al. Sep 1997 A
5677152 Birch et al. Oct 1997 A
5767099 Harris et al. Jun 1998 A
5780053 Ashley et al. Jul 1998 A
5830430 Unger et al. Nov 1998 A
5834247 Comb et al. Nov 1998 A
5835699 Kimura Nov 1998 A
5844075 Kawakami et al. Dec 1998 A
5849548 Haseloff et al. Dec 1998 A
5851548 Dattagupta et al. Dec 1998 A
5855910 Ashley et al. Jan 1999 A
5856463 Blankenborg et al. Jan 1999 A
5962313 Podsakoff et al. Oct 1999 A
5981182 Jacobs, Jr. et al. Nov 1999 A
6015794 Haseloff et al. Jan 2000 A
6057153 George et al. May 2000 A
6063608 Kotewicz et al. May 2000 A
6077705 Duan et al. Jun 2000 A
6156509 Schellenberger Dec 2000 A
6183998 Ivanov et al. Feb 2001 B1
6355415 Wagner et al. Mar 2002 B1
6429298 Ellington et al. Aug 2002 B1
6453242 Eisenberg et al. Sep 2002 B1
6479264 Louwrier Nov 2002 B1
6503717 Case et al. Jan 2003 B2
6534261 Cox, III et al. Mar 2003 B1
6589768 Kotewicz et al. Jul 2003 B1
6599692 Case et al. Jul 2003 B1
6607882 Cox, III et al. Aug 2003 B1
6610522 Kotewicz et al. Aug 2003 B1
6689558 Case et al. Feb 2004 B2
6716973 Baskerville et al. Apr 2004 B2
6824978 Cox, III et al. Nov 2004 B1
6933113 Case et al. Aug 2005 B2
6979539 Cox, III et al. Dec 2005 B2
7013219 Case et al. Mar 2006 B2
7045337 Schultz et al. May 2006 B2
7067650 Tanaka Jun 2006 B1
7070928 Liu et al. Jul 2006 B2
7078208 Smith et al. Jul 2006 B2
7083970 Schultz et al. Aug 2006 B2
7163824 Cox, III et al. Jan 2007 B2
7192739 Liu et al. Mar 2007 B2
7223545 Liu et al. May 2007 B2
7354761 Schultz et al. Apr 2008 B2
7368275 Schultz et al. May 2008 B2
7442160 Liu et al. Oct 2008 B2
7476500 Liu et al. Jan 2009 B1
7476734 Liu Jan 2009 B2
7479573 Chu et al. Jan 2009 B2
7491494 Liu et al. Feb 2009 B2
7541450 Liu et al. Jun 2009 B2
7557068 Liu et al. Jul 2009 B2
7595179 Chen et al. Sep 2009 B2
7638300 Schultz et al. Dec 2009 B2
7670807 Lampson et al. Mar 2010 B2
7678554 Liu et al. Mar 2010 B2
7713721 Schultz et al. May 2010 B2
7771935 Liu et al. Aug 2010 B2
7794931 Breaker et al. Sep 2010 B2
7807408 Liu et al. Oct 2010 B2
7851658 Liu et al. Dec 2010 B2
7915025 Schultz et al. Mar 2011 B2
7919277 Russell et al. Apr 2011 B2
7993672 Huang et al. Aug 2011 B2
7998904 Liu et al. Aug 2011 B2
8012739 Schultz et al. Sep 2011 B2
8017323 Liu et al. Sep 2011 B2
8017755 Liu et al. Sep 2011 B2
8030074 Schultz et al. Oct 2011 B2
8067556 Hogrefe et al. Nov 2011 B2
8114648 Schultz et al. Feb 2012 B2
8173364 Schultz et al. May 2012 B2
8173392 Schultz et al. May 2012 B2
8183012 Schultz et al. May 2012 B2
8183178 Liu et al. May 2012 B2
8206914 Liu et al. Jun 2012 B2
8361725 Russell et al. Jan 2013 B2
8394604 Liu et al. Mar 2013 B2
8440431 Voytas et al. May 2013 B2
8440432 Voytas et al. May 2013 B2
8450471 Voytas et al. May 2013 B2
8492082 De Franciscis et al. Jul 2013 B2
8546553 Terns et al. Oct 2013 B2
8569256 Heyes et al. Oct 2013 B2
8586363 Voytas et al. Nov 2013 B2
8680069 de Fougerolles et al. Mar 2014 B2
8691729 Liu et al. Apr 2014 B2
8691750 Constien et al. Apr 2014 B2
8697359 Zhang Apr 2014 B1
8697853 Voytas et al. Apr 2014 B2
8709466 Coady et al. Apr 2014 B2
8728526 Heller May 2014 B2
8748667 Budzik et al. Jun 2014 B2
8758810 Okada et al. Jun 2014 B2
8759103 Kim et al. Jun 2014 B2
8759104 Unciti-Broceta et al. Jun 2014 B2
8771728 Huang et al. Jul 2014 B2
8790664 Pitard et al. Jul 2014 B2
8795965 Zhang Aug 2014 B2
8822663 Schrum et al. Sep 2014 B2
8835148 Janulaitis et al. Sep 2014 B2
8846578 McCray et al. Sep 2014 B2
8871445 Cong et al. Oct 2014 B2
8889418 Zhang et al. Nov 2014 B2
8900814 Yasukawa et al. Dec 2014 B2
8945839 Zhang Feb 2015 B2
8975232 Liu et al. Mar 2015 B2
8993233 Zhang et al. Mar 2015 B2
8999641 Zhang et al. Apr 2015 B2
9023594 Liu et al. May 2015 B2
9023649 Mali et al. May 2015 B2
9034650 Padidam May 2015 B2
9068179 Liu et al. Jun 2015 B1
9150626 Liu et al. Oct 2015 B2
9163271 Schultz et al. Oct 2015 B2
9163284 Liu et al. Oct 2015 B2
9181535 Liu et al. Nov 2015 B2
9200045 Liu et al. Dec 2015 B2
9221886 Liu et al. Dec 2015 B2
9228207 Liu et al. Jan 2016 B2
9234213 Wu Jan 2016 B2
9243038 Liu et al. Jan 2016 B2
9267127 Liu et al. Feb 2016 B2
9322006 Liu et al. Apr 2016 B2
9322037 Liu et al. Apr 2016 B2
9340799 Liu et al. May 2016 B2
9340800 Liu et al. May 2016 B2
9359599 Liu et al. Jun 2016 B2
9388430 Liu et al. Jul 2016 B2
9394537 Liu et al. Jul 2016 B2
9434774 Liu et al. Sep 2016 B2
9458484 Ma et al. Oct 2016 B2
9512446 Joung et al. Dec 2016 B1
9526784 Liu et al. Dec 2016 B2
9534210 Park et al. Jan 2017 B2
9580698 Xu et al. Feb 2017 B1
9610322 Liu et al. Apr 2017 B2
9637739 Siksnys et al. May 2017 B2
9663770 Rogers et al. May 2017 B2
9737604 Jin et al. Aug 2017 B2
9738693 Telford et al. Aug 2017 B2
9753340 Saitou Sep 2017 B2
9771574 Liu et al. Sep 2017 B2
9783791 Hogrefe et al. Oct 2017 B2
9816093 Donohoue et al. Nov 2017 B1
9840538 Telford et al. Dec 2017 B2
9840690 Karli et al. Dec 2017 B2
9840699 Liu et al. Dec 2017 B2
9840702 Collingwood et al. Dec 2017 B2
9850521 Braman et al. Dec 2017 B2
9873907 Zeiner et al. Jan 2018 B2
9879270 Hittinger et al. Jan 2018 B2
9914939 Church et al. Mar 2018 B2
9932567 Xu et al. Apr 2018 B1
9938288 Kishi et al. Apr 2018 B1
9944933 Storici et al. Apr 2018 B2
9982279 Gill et al. May 2018 B1
9999671 Liu et al. Jun 2018 B2
10011868 Liu et al. Jul 2018 B2
10053725 Liu et al. Aug 2018 B2
10059940 Zhong Aug 2018 B2
10077453 Liu et al. Sep 2018 B2
10113163 Liu et al. Oct 2018 B2
10150955 Lambowitz et al. Dec 2018 B2
10167457 Liu et al. Jan 2019 B2
10179911 Liu et al. Jan 2019 B2
10189831 Arrington et al. Jan 2019 B2
10202593 Liu et al. Feb 2019 B2
10202658 Parkin et al. Feb 2019 B2
10227581 Liu et al. Mar 2019 B2
10323236 Liu et al. Jun 2019 B2
10336997 Liu et al. Jul 2019 B2
10358670 Janulaitis et al. Jul 2019 B2
10392674 Liu et al. Aug 2019 B2
10407474 Liu et al. Sep 2019 B2
10407697 Doudna et al. Sep 2019 B2
10465176 Liu et al. Nov 2019 B2
10508298 Liu et al. Dec 2019 B2
10583201 Chen et al. Mar 2020 B2
10597679 Liu et al. Mar 2020 B2
10612011 Liu et al. Apr 2020 B2
10640767 Maianti et al. May 2020 B2
10682410 Liu et al. Jun 2020 B2
10704062 Liu et al. Jul 2020 B2
10745677 Maianti et al. Aug 2020 B2
10858639 Liu et al. Dec 2020 B2
10912833 Liu et al. Feb 2021 B2
10930367 Zhang et al. Feb 2021 B2
10947530 Liu et al. Mar 2021 B2
10954548 Liu et al. Mar 2021 B2
11046948 Liu et al. Jun 2021 B2
11053481 Liu et al. Jul 2021 B2
11124782 Liu et al. Sep 2021 B2
11214780 Liu et al. Jan 2022 B2
11268082 Liu et al. Mar 2022 B2
11299755 Liu et al. Apr 2022 B2
11306324 Liu et al. Apr 2022 B2
11319532 Liu et al. May 2022 B2
11447770 Liu et al. Sep 2022 B1
11542496 Liu et al. Jan 2023 B2
11542509 Maianti et al. Jan 2023 B2
11560566 Liu et al. Jan 2023 B2
11578343 Liu et al. Feb 2023 B2
11643652 Liu et al. May 2023 B2
11661590 Liu et al. May 2023 B2
11702651 Liu et al. Jul 2023 B2
20030082575 Schultz et al. May 2003 A1
20030087817 Cox et al. May 2003 A1
20030096337 Hillman et al. May 2003 A1
20030108885 Schultz et al. Jun 2003 A1
20030119764 Loeb et al. Jun 2003 A1
20030167533 Yadav et al. Sep 2003 A1
20030203480 Kovesdi et al. Oct 2003 A1
20040003420 Kuhn et al. Jan 2004 A1
20040115184 Smith et al. Jun 2004 A1
20040197892 Moore et al. Oct 2004 A1
20040203109 Lal et al. Oct 2004 A1
20050136429 Guarente et al. Jun 2005 A1
20050222030 Allison Oct 2005 A1
20050260626 Lorens et al. Nov 2005 A1
20060088864 Smolke et al. Apr 2006 A1
20060104984 Littlefield et al. May 2006 A1
20060246568 Honjo et al. Nov 2006 A1
20070015238 Snyder et al. Jan 2007 A1
20070264692 Liu et al. Nov 2007 A1
20070269817 Shapero Nov 2007 A1
20080008697 Mintier et al. Jan 2008 A1
20080051317 Church et al. Feb 2008 A1
20080124725 Barrangou et al. May 2008 A1
20080182254 Hall et al. Jul 2008 A1
20080220502 Schellenberger et al. Sep 2008 A1
20080241917 Akita et al. Oct 2008 A1
20080268516 Perreault et al. Oct 2008 A1
20090130718 Short May 2009 A1
20090215878 Tan et al. Aug 2009 A1
20090234109 Han et al. Sep 2009 A1
20100076057 Sontheimer et al. Mar 2010 A1
20100093617 Barrangou et al. Apr 2010 A1
20100104690 Barrangou et al. Apr 2010 A1
20100273857 Thakker et al. Oct 2010 A1
20100305197 Che Dec 2010 A1
20100316643 Eckert et al. Dec 2010 A1
20110016540 Weinstein et al. Jan 2011 A1
20110059160 Essner et al. Mar 2011 A1
20110059502 Chalasani Mar 2011 A1
20110104787 Church et al. May 2011 A1
20110177495 Liu et al. Jul 2011 A1
20110189775 Ainley et al. Aug 2011 A1
20110189776 Terns et al. Aug 2011 A1
20110217739 Terns et al. Sep 2011 A1
20110301073 Gregory et al. Dec 2011 A1
20120129759 Liu et al. May 2012 A1
20120141523 Castado et al. Jun 2012 A1
20120244601 Bertozzi et al. Sep 2012 A1
20120270273 Zhang et al. Oct 2012 A1
20120322861 Byrne et al. Dec 2012 A1
20130022980 Nelson et al. Jan 2013 A1
20130059931 Petersen-Mahrt et al. Mar 2013 A1
20130117869 Duchateau et al. May 2013 A1
20130130248 Haurwitz et al. May 2013 A1
20130158245 Russell et al. Jun 2013 A1
20130165389 Schellenberger et al. Jun 2013 A1
20130212725 Kuhn et al. Aug 2013 A1
20130309720 Schultz et al. Nov 2013 A1
20130344117 Mirosevich et al. Dec 2013 A1
20130345064 Liu et al. Dec 2013 A1
20140004280 Loomis Jan 2014 A1
20140005269 Ngwuluka et al. Jan 2014 A1
20140017214 Cost Jan 2014 A1
20140018404 Chen et al. Jan 2014 A1
20140044793 Goll et al. Feb 2014 A1
20140065711 Liu et al. Mar 2014 A1
20140068797 Doudna et al. Mar 2014 A1
20140127752 Zhou et al. May 2014 A1
20140128449 Liu et al. May 2014 A1
20140141094 Smyth et al. May 2014 A1
20140141487 Feldman et al. May 2014 A1
20140179770 Zhang et al. Jun 2014 A1
20140186843 Zhang et al. Jul 2014 A1
20140186919 Zhang Jul 2014 A1
20140186958 Zhang et al. Jul 2014 A1
20140201858 Ostertag et al. Jul 2014 A1
20140234289 Liu et al. Aug 2014 A1
20140248702 Zhang et al. Sep 2014 A1
20140273037 Wu Sep 2014 A1
20140273226 Wu Sep 2014 A1
20140273230 Chen et al. Sep 2014 A1
20140273234 Zhang et al. Sep 2014 A1
20140283156 Zador et al. Sep 2014 A1
20140295556 Joung et al. Oct 2014 A1
20140295557 Joung et al. Oct 2014 A1
20140342456 Mali et al. Nov 2014 A1
20140342457 Mali et al. Nov 2014 A1
20140342458 Mali et al. Nov 2014 A1
20140349400 Jakimo et al. Nov 2014 A1
20140356867 Peter et al. Dec 2014 A1
20140356956 Church et al. Dec 2014 A1
20140356958 Mali et al. Dec 2014 A1
20140356959 Church et al. Dec 2014 A1
20140357523 Zeiner et al. Dec 2014 A1
20140377868 Joung et al. Dec 2014 A1
20150010526 Liu et al. Jan 2015 A1
20150031089 Lindstrom Jan 2015 A1
20150031132 Church et al. Jan 2015 A1
20150031133 Church et al. Jan 2015 A1
20150044191 Liu et al. Feb 2015 A1
20150044192 Liu et al. Feb 2015 A1
20150044772 Zhao Feb 2015 A1
20150050699 Siksnys et al. Feb 2015 A1
20150056177 Liu et al. Feb 2015 A1
20150056629 Guthrie-Honea Feb 2015 A1
20150064138 Lu et al. Mar 2015 A1
20150064789 Paschon et al. Mar 2015 A1
20150071898 Liu et al. Mar 2015 A1
20150071899 Liu et al. Mar 2015 A1
20150071900 Liu et al. Mar 2015 A1
20150071901 Liu et al. Mar 2015 A1
20150071902 Liu et al. Mar 2015 A1
20150071903 Liu et al. Mar 2015 A1
20150071906 Liu et al. Mar 2015 A1
20150079680 Bradley et al. Mar 2015 A1
20150079681 Zhang Mar 2015 A1
20150098954 Hyde et al. Apr 2015 A1
20150118216 Liu et al. Apr 2015 A1
20150128300 Warming et al. May 2015 A1
20150132269 Orkin et al. May 2015 A1
20150140664 Byrne et al. May 2015 A1
20150159172 Miller et al. Jun 2015 A1
20150165054 Liu et al. Jun 2015 A1
20150166980 Liu et al. Jun 2015 A1
20150166981 Liu et al. Jun 2015 A1
20150166982 Liu et al. Jun 2015 A1
20150166983 Liu et al. Jun 2015 A1
20150166984 Liu et al. Jun 2015 A1
20150166985 Liu et al. Jun 2015 A1
20150191744 Wolfe et al. Jul 2015 A1
20150197759 Xu et al. Jul 2015 A1
20150211058 Carstens Jul 2015 A1
20150218573 Loque et al. Aug 2015 A1
20150225773 Farmer et al. Aug 2015 A1
20150252358 Maeder et al. Sep 2015 A1
20150275202 Liu et al. Oct 2015 A1
20150291965 Zhang et al. Oct 2015 A1
20150307889 Petolino et al. Oct 2015 A1
20150315252 Haugwitz et al. Nov 2015 A1
20150344549 Muir et al. Dec 2015 A1
20160017393 Jacobson et al. Jan 2016 A1
20160017396 Cann et al. Jan 2016 A1
20160032292 Storici et al. Feb 2016 A1
20160032353 Braman et al. Feb 2016 A1
20160040155 Maizels et al. Feb 2016 A1
20160046952 Hittinger et al. Feb 2016 A1
20160046961 Jinek et al. Feb 2016 A1
20160046962 May et al. Feb 2016 A1
20160053272 Wurtzel et al. Feb 2016 A1
20160053304 Wurtzel et al. Feb 2016 A1
20160074535 Ranganathan et al. Mar 2016 A1
20160076093 Shendure et al. Mar 2016 A1
20160090603 Carnes et al. Mar 2016 A1
20160090622 Liu et al. Mar 2016 A1
20160115488 Zhang et al. Apr 2016 A1
20160138046 Wu May 2016 A1
20160153003 Joung et al. Jun 2016 A1
20160186214 Brouns et al. Jun 2016 A1
20160200779 Liu et al. Jul 2016 A1
20160201040 Liu et al. Jul 2016 A1
20160201089 Gersbach et al. Jul 2016 A1
20160206566 Lu et al. Jul 2016 A1
20160208243 Zhang et al. Jul 2016 A1
20160208288 Liu et al. Jul 2016 A1
20160215275 Zhong Jul 2016 A1
20160215276 Liu et al. Jul 2016 A1
20160215300 May et al. Jul 2016 A1
20160244784 Jacobson et al. Aug 2016 A1
20160244829 Bang et al. Aug 2016 A1
20160264934 Giallourakis et al. Sep 2016 A1
20160272593 Ritter et al. Sep 2016 A1
20160272965 Zhang et al. Sep 2016 A1
20160281072 Zhang Sep 2016 A1
20160298136 Chen et al. Oct 2016 A1
20160304846 Liu et al. Oct 2016 A1
20160304855 Stark et al. Oct 2016 A1
20160312304 Sorrentino et al. Oct 2016 A1
20160319262 Doudna et al. Nov 2016 A1
20160333389 Liu et al. Nov 2016 A1
20160340622 Abdou Nov 2016 A1
20160340662 Zhang et al. Nov 2016 A1
20160345578 Barrangou et al. Dec 2016 A1
20160346360 Quake et al. Dec 2016 A1
20160346361 Quake et al. Dec 2016 A1
20160346362 Quake et al. Dec 2016 A1
20160348074 Quake et al. Dec 2016 A1
20160348096 Liu et al. Dec 2016 A1
20160350476 Quake et al. Dec 2016 A1
20160355796 Davidson et al. Dec 2016 A1
20160369262 Reik et al. Dec 2016 A1
20170009224 Liu et al. Jan 2017 A1
20170009242 McKinley et al. Jan 2017 A1
20170014449 Bangera et al. Jan 2017 A1
20170020922 Wagner et al. Jan 2017 A1
20170037432 Donohoue et al. Feb 2017 A1
20170044520 Liu et al. Feb 2017 A1
20170044592 Peter et al. Feb 2017 A1
20170053729 Kotani et al. Feb 2017 A1
20170058271 Joung et al. Mar 2017 A1
20170058272 Carter et al. Mar 2017 A1
20170058298 Kennedy et al. Mar 2017 A1
20170073663 Wang et al. Mar 2017 A1
20170073670 Nishida et al. Mar 2017 A1
20170087224 Quake Mar 2017 A1
20170087225 Quake Mar 2017 A1
20170088587 Quake Mar 2017 A1
20170088828 Quake Mar 2017 A1
20170107536 Zhang et al. Apr 2017 A1
20170107560 Peter et al. Apr 2017 A1
20170114367 Hu et al. Apr 2017 A1
20170121693 Liu et al. May 2017 A1
20170145394 Yeo et al. May 2017 A1
20170145405 Tang et al. May 2017 A1
20170145438 Kantor May 2017 A1
20170152528 Zhang Jun 2017 A1
20170152787 Kubo et al. Jun 2017 A1
20170159033 Kamtekar et al. Jun 2017 A1
20170166928 Vyas et al. Jun 2017 A1
20170175104 Doudna et al. Jun 2017 A1
20170175142 Zhang et al. Jun 2017 A1
20170191047 Terns et al. Jul 2017 A1
20170191078 Zhang et al. Jul 2017 A1
20170198269 Zhang et al. Jul 2017 A1
20170198277 Kmiec et al. Jul 2017 A1
20170198302 Feng et al. Jul 2017 A1
20170211061 Weiss Jul 2017 A1
20170226522 Hu et al. Aug 2017 A1
20170233703 Xie et al. Aug 2017 A1
20170233708 Liu et al. Aug 2017 A1
20170233756 Begemann et al. Aug 2017 A1
20170247671 Yung et al. Aug 2017 A1
20170247703 Sloan et al. Aug 2017 A1
20170268022 Liu et al. Sep 2017 A1
20170275648 Barrangou et al. Sep 2017 A1
20170275665 Silas et al. Sep 2017 A1
20170283797 Robb et al. Oct 2017 A1
20170283831 Zhang et al. Oct 2017 A1
20170306306 Potter et al. Oct 2017 A1
20170314016 Kim et al. Nov 2017 A1
20170362635 Chamberlain et al. Dec 2017 A1
20180023062 Lamb et al. Jan 2018 A1
20180064077 Dunham et al. Mar 2018 A1
20180066258 Powell Mar 2018 A1
20180068062 Zhang et al. Mar 2018 A1
20180073012 Liu et al. Mar 2018 A1
20180080051 Sheikh et al. Mar 2018 A1
20180087046 Badran et al. Mar 2018 A1
20180100147 Yates et al. Apr 2018 A1
20180105867 Xiao et al. Apr 2018 A1
20180119118 Lu et al. May 2018 A1
20180127759 Lu et al. May 2018 A1
20180127780 Liu et al. May 2018 A1
20180155708 Church et al. Jun 2018 A1
20180155720 Donohoue et al. Jun 2018 A1
20180163213 Aneja et al. Jun 2018 A1
20180170984 Harris et al. Jun 2018 A1
20180179503 Maianti et al. Jun 2018 A1
20180179547 Zhang et al. Jun 2018 A1
20180201921 Malcolm Jul 2018 A1
20180230464 Zhong Aug 2018 A1
20180230471 Storici et al. Aug 2018 A1
20180236081 Liu et al. Aug 2018 A1
20180237787 Maianti et al. Aug 2018 A1
20180245066 Yao et al. Aug 2018 A1
20180245075 Khalil et al. Aug 2018 A1
20180258418 Kim Sep 2018 A1
20180265864 Li et al. Sep 2018 A1
20180273939 Yu et al. Sep 2018 A1
20180282722 Jakimo et al. Oct 2018 A1
20180298391 Jakimo et al. Oct 2018 A1
20180305688 Zhong Oct 2018 A1
20180305704 Zhang Oct 2018 A1
20180312822 Lee et al. Nov 2018 A1
20180312825 Liu et al. Nov 2018 A1
20180312828 Liu et al. Nov 2018 A1
20180312835 Yao et al. Nov 2018 A1
20180327756 Zhang et al. Nov 2018 A1
20180346927 Doudna et al. Dec 2018 A1
20180371497 Gill et al. Dec 2018 A1
20190010481 Joung et al. Jan 2019 A1
20190055543 Tran et al. Feb 2019 A1
20190055549 Capurso et al. Feb 2019 A1
20190062734 Cotta-Ramusino et al. Feb 2019 A1
20190093099 Liu et al. Mar 2019 A1
20190185883 Liu et al. Jun 2019 A1
20190218547 Lee et al. Jul 2019 A1
20190225955 Liu et al. Jul 2019 A1
20190233847 Savage et al. Aug 2019 A1
20190241633 Fotin-Mleczek et al. Aug 2019 A1
20190256842 Liu et al. Aug 2019 A1
20190264202 Church et al. Aug 2019 A1
20190276816 Liu et al. Sep 2019 A1
20190309290 Neuteboom et al. Oct 2019 A1
20190322992 Liu et al. Oct 2019 A1
20190330619 Smith et al. Oct 2019 A1
20190352632 Liu et al. Nov 2019 A1
20190367891 Liu et al. Dec 2019 A1
20200010818 Liu et al. Jan 2020 A1
20200010835 Maianti et al. Jan 2020 A1
20200063127 Lu et al. Feb 2020 A1
20200071722 Liu et al. Mar 2020 A1
20200109398 Rubens et al. Apr 2020 A1
20200172931 Liu et al. Jun 2020 A1
20200181619 Tang et al. Jun 2020 A1
20200190493 Liu et al. Jun 2020 A1
20200216833 Liu et al. Jul 2020 A1
20200255868 Liu et al. Aug 2020 A1
20200277587 Liu et al. Sep 2020 A1
20200323984 Liu et al. Oct 2020 A1
20200399619 Maianti et al. Dec 2020 A1
20200399626 Liu et al. Dec 2020 A1
20210054416 Liu et al. Feb 2021 A1
20210115428 Maianti et al. Apr 2021 A1
20210196809 Maianti et al. Jul 2021 A1
20210198330 Liu et al. Jul 2021 A1
20210214698 Liu et al. Jul 2021 A1
20210230577 Liu et al. Jul 2021 A1
20210254127 Liu et al. Aug 2021 A1
20210292753 Halperin Sep 2021 A1
20210315994 Liu et al. Oct 2021 A1
20210317440 Liu et al. Oct 2021 A1
20220033785 Liu et al. Feb 2022 A1
20220119785 Liu et al. Apr 2022 A1
20220170013 Liu et al. Jun 2022 A1
20220177877 Church et al. Jun 2022 A1
20220204975 Liu et al. Jun 2022 A1
20220213507 Liu et al. Jul 2022 A1
20220220462 Liu et al. Jul 2022 A1
20220238182 Shen et al. Jul 2022 A1
20220249697 Liu et al. Aug 2022 A1
20220282275 Liu et al. Sep 2022 A1
20220290115 Liu et al. Sep 2022 A1
20220307001 Liu et al. Sep 2022 A1
20220307003 Liu et al. Sep 2022 A1
20220315906 Liu et al. Oct 2022 A1
20220356469 Liu et al. Nov 2022 A1
20220380740 Liu et al. Dec 2022 A1
20220389395 Liu et al. Dec 2022 A1
20230002745 Liu et al. Jan 2023 A1
20230021641 Liu et al. Jan 2023 A1
20230056852 Liu et al. Feb 2023 A1
20230058176 Liu et al. Feb 2023 A1
20230078265 Liu et al. Mar 2023 A1
20230086199 Liu et al. Mar 2023 A1
20230090221 Liu et al. Mar 2023 A1
20230108687 Liu et al. Apr 2023 A1
20230123669 Liu et al. Apr 2023 A1
20230127008 Liu et al. Apr 2023 A1
20230159913 Liu et al. May 2023 A1
20230193295 Maianti et al. Jun 2023 A1
20230272425 Liu et al. Aug 2023 A1
20230279443 Liu et al. Sep 2023 A1
Foreign Referenced Citations (1684)
Number Date Country
2012244264 Nov 2012 AU
2015252023 Nov 2015 AU
2015101792 Jan 2016 AU
2012354062 Sep 2017 AU
112015013786 Jul 2017 BR
2894668 Jun 2014 CA
2894681 Jun 2014 CA
2894684 Jun 2014 CA
2852593 Nov 2015 CA
3193022 Mar 2022 CA
2865578 Jan 2023 CA
1069962 Mar 1993 CN
104004778 Aug 2001 CN
101460619 Jun 2009 CN
101873862 Oct 2010 CN
102892777 Jan 2013 CN
103224947 Jul 2013 CN
103233028 Aug 2013 CN
103388006 Nov 2013 CN
103614415 Mar 2014 CN
103642836 Mar 2014 CN
103668472 Mar 2014 CN
103820441 May 2014 CN
103820454 May 2014 CN
103911376 Jul 2014 CN
103923911 Jul 2014 CN
103088008 Aug 2014 CN
103981211 Aug 2014 CN
103981212 Aug 2014 CN
104004782 Aug 2014 CN
104017821 Sep 2014 CN
104109687 Oct 2014 CN
104178461 Dec 2014 CN
104342457 Feb 2015 CN
104404036 Mar 2015 CN
104450774 Mar 2015 CN
104480144 Apr 2015 CN
104498493 Apr 2015 CN
104504304 Apr 2015 CN
104531704 Apr 2015 CN
104531705 Apr 2015 CN
104560864 Apr 2015 CN
104561095 Apr 2015 CN
104593418 May 2015 CN
104593422 May 2015 CN
104611370 May 2015 CN
104651392 May 2015 CN
104651398 May 2015 CN
104651399 May 2015 CN
104651401 May 2015 CN
104673816 Jun 2015 CN
104725626 Jun 2015 CN
104726449 Jun 2015 CN
104726494 Jun 2015 CN
104745626 Jul 2015 CN
104762321 Jul 2015 CN
104805078 Jul 2015 CN
104805099 Jul 2015 CN
104805118 Jul 2015 CN
104846010 Aug 2015 CN
104894068 Sep 2015 CN
104894075 Sep 2015 CN
104928321 Sep 2015 CN
105039339 Nov 2015 CN
105039399 Nov 2015 CN
105063061 Nov 2015 CN
105087620 Nov 2015 CN
105112422 Dec 2015 CN
105112445 Dec 2015 CN
105112519 Dec 2015 CN
105121648 Dec 2015 CN
105132427 Dec 2015 CN
105132451 Dec 2015 CN
105177038 Dec 2015 CN
105177126 Dec 2015 CN
105210981 Jan 2016 CN
105219799 Jan 2016 CN
105238806 Jan 2016 CN
105255937 Jan 2016 CN
105274144 Jan 2016 CN
105296518 Feb 2016 CN
105296537 Feb 2016 CN
105316324 Feb 2016 CN
105316327 Feb 2016 CN
105316337 Feb 2016 CN
105331607 Feb 2016 CN
105331608 Feb 2016 CN
105331609 Feb 2016 CN
105331627 Feb 2016 CN
105400773 Mar 2016 CN
105400779 Mar 2016 CN
105400810 Mar 2016 CN
105441451 Mar 2016 CN
105462968 Apr 2016 CN
105463003 Apr 2016 CN
105463027 Apr 2016 CN
105492608 Apr 2016 CN
105492609 Apr 2016 CN
105505976 Apr 2016 CN
105505979 Apr 2016 CN
105518134 Apr 2016 CN
105518135 Apr 2016 CN
105518137 Apr 2016 CN
105518138 Apr 2016 CN
105518139 Apr 2016 CN
105518140 Apr 2016 CN
105543228 May 2016 CN
105543266 May 2016 CN
105543270 May 2016 CN
105567688 May 2016 CN
105567689 May 2016 CN
105567734 May 2016 CN
105567735 May 2016 CN
105567738 May 2016 CN
105593367 May 2016 CN
105594664 May 2016 CN
105602987 May 2016 CN
105624146 Jun 2016 CN
105624187 Jun 2016 CN
105646719 Jun 2016 CN
105647922 Jun 2016 CN
105647962 Jun 2016 CN
105647968 Jun 2016 CN
105647969 Jun 2016 CN
105671070 Jun 2016 CN
105671083 Jun 2016 CN
105695485 Jun 2016 CN
105779448 Jul 2016 CN
105779449 Jul 2016 CN
105802980 Jul 2016 CN
105821039 Aug 2016 CN
105821040 Aug 2016 CN
105821049 Aug 2016 CN
105821072 Aug 2016 CN
105821075 Aug 2016 CN
105821116 Aug 2016 CN
105838733 Aug 2016 CN
105861547 Aug 2016 CN
105861552 Aug 2016 CN
105861554 Aug 2016 CN
105886498 Aug 2016 CN
105886534 Aug 2016 CN
105886616 Aug 2016 CN
105907758 Aug 2016 CN
105907785 Aug 2016 CN
105925608 Sep 2016 CN
105934516 Sep 2016 CN
105950560 Sep 2016 CN
105950626 Sep 2016 CN
105950633 Sep 2016 CN
105950639 Sep 2016 CN
105985985 Oct 2016 CN
106011104 Oct 2016 CN
106011150 Oct 2016 CN
106011167 Oct 2016 CN
106011171 Oct 2016 CN
106032540 Oct 2016 CN
106047803 Oct 2016 CN
106047877 Oct 2016 CN
106047930 Oct 2016 CN
106086008 Nov 2016 CN
106086028 Nov 2016 CN
106086061 Nov 2016 CN
106086062 Nov 2016 CN
106109417 Nov 2016 CN
106119275 Nov 2016 CN
106119283 Nov 2016 CN
106148286 Nov 2016 CN
106148370 Nov 2016 CN
106148416 Nov 2016 CN
106167525 Nov 2016 CN
106167808 Nov 2016 CN
106167810 Nov 2016 CN
106167821 Nov 2016 CN
106172238 Dec 2016 CN
106190903 Dec 2016 CN
106191057 Dec 2016 CN
106191061 Dec 2016 CN
106191062 Dec 2016 CN
106191064 Dec 2016 CN
106191071 Dec 2016 CN
106191099 Dec 2016 CN
106191107 Dec 2016 CN
106191113 Dec 2016 CN
106191114 Dec 2016 CN
106191116 Dec 2016 CN
106191124 Dec 2016 CN
106222177 Dec 2016 CN
106222193 Dec 2016 CN
106222203 Dec 2016 CN
106244555 Dec 2016 CN
106244557 Dec 2016 CN
106244591 Dec 2016 CN
106244609 Dec 2016 CN
106282241 Jan 2017 CN
106318934 Jan 2017 CN
106318973 Jan 2017 CN
106350540 Jan 2017 CN
106367435 Feb 2017 CN
106399306 Feb 2017 CN
106399311 Feb 2017 CN
106399360 Feb 2017 CN
106399367 Feb 2017 CN
106399375 Feb 2017 CN
106399377 Feb 2017 CN
106434651 Feb 2017 CN
106434663 Feb 2017 CN
106434688 Feb 2017 CN
106434737 Feb 2017 CN
106434748 Feb 2017 CN
106434752 Feb 2017 CN
106434782 Feb 2017 CN
106446600 Feb 2017 CN
106479985 Mar 2017 CN
106480027 Mar 2017 CN
106480036 Mar 2017 CN
106480067 Mar 2017 CN
106480080 Mar 2017 CN
106480083 Mar 2017 CN
106480097 Mar 2017 CN
106544351 Mar 2017 CN
106544353 Mar 2017 CN
106544357 Mar 2017 CN
106554969 Apr 2017 CN
106566838 Apr 2017 CN
106701763 May 2017 CN
106701808 May 2017 CN
106701818 May 2017 CN
106701823 May 2017 CN
106701830 May 2017 CN
106754912 May 2017 CN
106755026 May 2017 CN
106755077 May 2017 CN
106755088 May 2017 CN
106755091 May 2017 CN
106755097 May 2017 CN
106755424 May 2017 CN
106801056 Jun 2017 CN
106834323 Jun 2017 CN
106834341 Jun 2017 CN
106834347 Jun 2017 CN
106845151 Jun 2017 CN
106868008 Jun 2017 CN
106868031 Jun 2017 CN
106906240 Jun 2017 CN
106906242 Jun 2017 CN
106916820 Jul 2017 CN
106916852 Jul 2017 CN
106939303 Jul 2017 CN
106947750 Jul 2017 CN
106947780 Jul 2017 CN
106957830 Jul 2017 CN
106957831 Jul 2017 CN
106957844 Jul 2017 CN
106957855 Jul 2017 CN
106957858 Jul 2017 CN
106967697 Jul 2017 CN
106967726 Jul 2017 CN
106978428 Jul 2017 CN
106987570 Jul 2017 CN
106987757 Jul 2017 CN
107012164 Aug 2017 CN
107012174 Aug 2017 CN
107012213 Aug 2017 CN
107012250 Aug 2017 CN
107022562 Aug 2017 CN
107034188 Aug 2017 CN
107034218 Aug 2017 CN
107034229 Aug 2017 CN
107043775 Aug 2017 CN
107043779 Aug 2017 CN
107043787 Aug 2017 CN
107058320 Aug 2017 CN
107058328 Aug 2017 CN
107058358 Aug 2017 CN
107058372 Aug 2017 CN
107083392 Aug 2017 CN
107099533 Aug 2017 CN
107099850 Aug 2017 CN
107119053 Sep 2017 CN
107119071 Sep 2017 CN
107129999 Sep 2017 CN
107130000 Sep 2017 CN
107142272 Sep 2017 CN
107142282 Sep 2017 CN
107177591 Sep 2017 CN
107177595 Sep 2017 CN
107177625 Sep 2017 CN
107177631 Sep 2017 CN
107190006 Sep 2017 CN
107190008 Sep 2017 CN
107217042 Sep 2017 CN
107217075 Sep 2017 CN
107227307 Oct 2017 CN
107227352 Oct 2017 CN
107236737 Oct 2017 CN
107236739 Oct 2017 CN
107236741 Oct 2017 CN
107245502 Oct 2017 CN
107254485 Oct 2017 CN
107266541 Oct 2017 CN
107267515 Oct 2017 CN
107287245 Oct 2017 CN
107298701 Oct 2017 CN
107299114 Oct 2017 CN
107304435 Oct 2017 CN
107312785 Nov 2017 CN
107312793 Nov 2017 CN
107312795 Nov 2017 CN
107312798 Nov 2017 CN
107326042 Nov 2017 CN
107326046 Nov 2017 CN
107354156 Nov 2017 CN
107354173 Nov 2017 CN
107356793 Nov 2017 CN
107362372 Nov 2017 CN
107365786 Nov 2017 CN
107365804 Nov 2017 CN
107384894 Nov 2017 CN
107384922 Nov 2017 CN
107384926 Nov 2017 CN
107400677 Nov 2017 CN
107418974 Dec 2017 CN
107435051 Dec 2017 CN
107435069 Dec 2017 CN
107446922 Dec 2017 CN
107446923 Dec 2017 CN
107446924 Dec 2017 CN
107446932 Dec 2017 CN
107446951 Dec 2017 CN
107446954 Dec 2017 CN
107460196 Dec 2017 CN
107474129 Dec 2017 CN
107475300 Dec 2017 CN
107488649 Dec 2017 CN
107502608 Dec 2017 CN
107502618 Dec 2017 CN
107513531 Dec 2017 CN
107519492 Dec 2017 CN
107523567 Dec 2017 CN
107523583 Dec 2017 CN
107541525 Jan 2018 CN
107557373 Jan 2018 CN
107557378 Jan 2018 CN
107557381 Jan 2018 CN
107557390 Jan 2018 CN
107557393 Jan 2018 CN
107557394 Jan 2018 CN
107557455 Jan 2018 CN
107574179 Jan 2018 CN
107586777 Jan 2018 CN
107586779 Jan 2018 CN
107604003 Jan 2018 CN
107619829 Jan 2018 CN
107619837 Jan 2018 CN
107630006 Jan 2018 CN
107630041 Jan 2018 CN
107630042 Jan 2018 CN
107630043 Jan 2018 CN
107641631 Jan 2018 CN
107653256 Feb 2018 CN
107686848 Feb 2018 CN
206970581 Feb 2018 CN
107760652 Mar 2018 CN
107760663 Mar 2018 CN
107760684 Mar 2018 CN
107760715 Mar 2018 CN
107784200 Mar 2018 CN
107794272 Mar 2018 CN
107794276 Mar 2018 CN
107815463 Mar 2018 CN
107828738 Mar 2018 CN
107828794 Mar 2018 CN
107828826 Mar 2018 CN
107828874 Mar 2018 CN
107858346 Mar 2018 CN
107858373 Mar 2018 CN
107880132 Apr 2018 CN
107881184 Apr 2018 CN
107893074 Apr 2018 CN
107893075 Apr 2018 CN
107893076 Apr 2018 CN
107893080 Apr 2018 CN
107893086 Apr 2018 CN
107904261 Apr 2018 CN
107937427 Apr 2018 CN
107937432 Apr 2018 CN
107937501 Apr 2018 CN
107974466 May 2018 CN
107988229 May 2018 CN
107988246 May 2018 CN
107988256 May 2018 CN
107988268 May 2018 CN
108018316 May 2018 CN
108034656 May 2018 CN
108048466 May 2018 CN
108102940 Jun 2018 CN
108103090 Jun 2018 CN
108103092 Jun 2018 CN
108103098 Jun 2018 CN
108103586 Jun 2018 CN
108148835 Jun 2018 CN
108148837 Jun 2018 CN
108148873 Jun 2018 CN
108192956 Jun 2018 CN
108251423 Jul 2018 CN
108251451 Jul 2018 CN
108251452 Jul 2018 CN
108342480 Jul 2018 CN
108359691 Aug 2018 CN
108359712 Aug 2018 CN
108384784 Aug 2018 CN
108396027 Aug 2018 CN
108410877 Aug 2018 CN
108410906 Aug 2018 CN
108410907 Aug 2018 CN
108410911 Aug 2018 CN
108424931 Aug 2018 CN
108441519 Aug 2018 CN
108441520 Aug 2018 CN
108486108 Sep 2018 CN
108486111 Sep 2018 CN
108486145 Sep 2018 CN
108486146 Sep 2018 CN
108486154 Sep 2018 CN
108486159 Sep 2018 CN
108486234 Sep 2018 CN
108504657 Sep 2018 CN
108504685 Sep 2018 CN
108504693 Sep 2018 CN
108546712 Sep 2018 CN
108546717 Sep 2018 CN
108546718 Sep 2018 CN
108559730 Sep 2018 CN
108559732 Sep 2018 CN
108559745 Sep 2018 CN
108559760 Sep 2018 CN
108570479 Sep 2018 CN
108588071 Sep 2018 CN
108588123 Sep 2018 CN
108588128 Sep 2018 CN
108588182 Sep 2018 CN
108610399 Oct 2018 CN
108611364 Oct 2018 CN
108624622 Oct 2018 CN
108642053 Oct 2018 CN
108642055 Oct 2018 CN
108642077 Oct 2018 CN
108642078 Oct 2018 CN
108642090 Oct 2018 CN
108690844 Oct 2018 CN
108707604 Oct 2018 CN
108707620 Oct 2018 CN
108707621 Oct 2018 CN
108707628 Oct 2018 CN
108707629 Oct 2018 CN
108715850 Oct 2018 CN
108728476 Nov 2018 CN
108728486 Nov 2018 CN
108753772 Nov 2018 CN
108753783 Nov 2018 CN
108753813 Nov 2018 CN
108753817 Nov 2018 CN
108753832 Nov 2018 CN
108753835 Nov 2018 CN
108753836 Nov 2018 CN
108795902 Nov 2018 CN
108822217 Nov 2018 CN
108823248 Nov 2018 CN
108823249 Nov 2018 CN
108823291 Nov 2018 CN
108841845 Nov 2018 CN
108853133 Nov 2018 CN
108866093 Nov 2018 CN
108893529 Nov 2018 CN
108913664 Nov 2018 CN
108913691 Nov 2018 CN
108913714 Nov 2018 CN
108913717 Nov 2018 CN
208034188 Nov 2018 CN
109517841 Mar 2019 CN
0264166 Apr 1988 EP
0321201 Jun 1989 EP
0519463 Dec 1992 EP
2604255 Jun 2013 EP
2840140 Feb 2015 EP
2877490 Jun 2015 EP
2966170 Jan 2016 EP
3009511 Apr 2016 EP
3031921 Jun 2016 EP
3045537 Jul 2016 EP
3115457 Jan 2017 EP
3144390 Mar 2017 EP
3199632 Aug 2017 EP
3216867 Sep 2017 EP
3252160 Dec 2017 EP
3450553 Dec 2019 EP
3856898 Aug 2021 EP
2740248 Feb 2020 ES
2528177 Jan 2016 GB
2531454 Apr 2016 GB
2542653 Mar 2017 GB
1208045 Feb 2016 HK
2007-501626 Feb 2007 JP
2008-515405 May 2008 JP
2010-033344 Feb 2010 JP
2010-535744 Nov 2010 JP
2010-539929 Dec 2010 JP
2011-081011 Apr 2011 JP
2011-523353 Aug 2011 JP
2012-525146 Oct 2012 JP
2012-210172 Nov 2012 JP
2012-531909 Dec 2012 JP
2015-523856 Aug 2015 JP
2015-532654 Nov 2015 JP
2016-525888 Sep 2016 JP
2016-534132 Nov 2016 JP
2017-500035 Jan 2017 JP
20125-521045 Aug 2018 JP
6629734 Jan 2020 JP
6633524 Jan 2020 JP
101584933 Jan 2016 KR
2016-0050069 May 2016 KR
20160133380 Nov 2016 KR
20170037025 Apr 2017 KR
20170037028 Apr 2017 KR
101748575 Jun 2017 KR
20170128137 Nov 2017 KR
2018-0022465 Mar 2018 KR
2016104674 Aug 2017 RU
2634395 Oct 2017 RU
2652899 May 2018 RU
2015128057 Mar 2019 RU
2015128098 Mar 2019 RU
2687451 May 2019 RU
2019112514 Jun 2019 RU
2019127300 Sep 2019 RU
2701850 Oct 2019 RU
10201707569 Oct 2017 SG
10201710486 Jan 2018 SG
10201710487 Jan 2018 SG
10201710488 Jan 2018 SG
I608100 Dec 2017 TW
2018-29773 Aug 2018 TW
WO 1990002809 Mar 1990 WO
WO 1991003162 Mar 1991 WO
WO 1991016024 Oct 1991 WO
WO 1991017271 Nov 1991 WO
WO 1991017424 Nov 1991 WO
WO 1992006188 Apr 1992 WO
WO 1992006200 Apr 1992 WO
WO 1992007065 Apr 1992 WO
WO 1993015187 Aug 1993 WO
WO 1993024641 Dec 1993 WO
WO 1994018316 Aug 1994 WO
WO 1994026877 Nov 1994 WO
WO 1996004403 Feb 1996 WO
WO 1996010640 Apr 1996 WO
WO 1998032845 Jul 1998 WO
WO 2001036452 May 2001 WO
WO 2001038547 May 2001 WO
WO 2002059296 Aug 2002 WO
WO 2002068676 Sep 2002 WO
WO 2002103028 Dec 2002 WO
WO 2004007684 Jan 2004 WO
WO 2005014791 Feb 2005 WO
WO 2005019415 Mar 2005 WO
WO 2006002547 Jan 2006 WO
WO 2006042112 Apr 2006 WO
WO 2007025097 Mar 2007 WO
WO 2007037444 Apr 2007 WO
WO 2007066923 Jun 2007 WO
WO 2007136815 Nov 2007 WO
WO 2007143574 Dec 2007 WO
WO 2008005529 Jan 2008 WO
WO 2008108989 Sep 2008 WO
WO 2009002418 Dec 2008 WO
WO 2009098290 Aug 2009 WO
WO 2009134808 Nov 2009 WO
WO 2010011961 Jan 2010 WO
WO 2010012902 Feb 2010 WO
WO 2010028347 Mar 2010 WO
WO 2010054108 May 2010 WO
WO 2010054154 May 2010 WO
WO 2010068289 Jun 2010 WO
WO 2010075424 Jul 2010 WO
WO 2010102257 Sep 2010 WO
WO 2010104749 Sep 2010 WO
WO 2010129019 Nov 2010 WO
WO 2010129023 Nov 2010 WO
WO 2010132092 Nov 2010 WO
WO 2010144150 Dec 2010 WO
WO 2011002503 Jan 2011 WO
WO 2011017293 Feb 2011 WO
WO 2011053868 May 2011 WO
WO 2011053982 May 2011 WO
WO 2011068810 Jun 2011 WO
WO 2011075627 Jun 2011 WO
WO 2011091311 Jul 2011 WO
WO 2011091396 Jul 2011 WO
WO 2011109031 Sep 2011 WO
WO 2011143124 Nov 2011 WO
WO 2011147590 Dec 2011 WO
WO 2011159369 Dec 2011 WO
WO 2012054726 Apr 2012 WO
WO 2012065043 May 2012 WO
WO 2012088381 Jun 2012 WO
WO 2012125445 Sep 2012 WO
WO 2012138927 Oct 2012 WO
WO 2012149470 Nov 2012 WO
WO 2012158985 Nov 2012 WO
WO 2012158986 Nov 2012 WO
WO 2012164565 Dec 2012 WO
WO 2012170930 Dec 2012 WO
WO 2013012674 Jan 2013 WO
WO 2013013105 Jan 2013 WO
WO 2013039857 Mar 2013 WO
WO 2013039861 Mar 2013 WO
WO 2013045632 Apr 2013 WO
WO 2013047844 Apr 2013 WO
WO 2013066438 May 2013 WO
WO 2013086441 Jun 2013 WO
WO 2013086444 Jun 2013 WO
WO 2013098244 Jul 2013 WO
WO 2013119602 Aug 2013 WO
WO 2013126794 Aug 2013 WO
WO 2013130824 Sep 2013 WO
WO 2013141680 Sep 2013 WO
WO 2013142578 Sep 2013 WO
WO 2013152359 Oct 2013 WO
WO 2013160230 Oct 2013 WO
WO 2013166315 Nov 2013 WO
WO 2013169398 Nov 2013 WO
WO 2013169802 Nov 2013 WO
WO 2013176772 Nov 2013 WO
WO 2013176915 Nov 2013 WO
WO 2013176916 Nov 2013 WO
WO 2013181440 Dec 2013 WO
WO 2013186754 Dec 2013 WO
WO 2013188037 Dec 2013 WO
WO 2013188522 Dec 2013 WO
WO 2013188638 Dec 2013 WO
WO 2013192278 Dec 2013 WO
WO 2013142378 Jan 2014 WO
WO 2014004336 Jan 2014 WO
WO 2014005042 Jan 2014 WO
WO 2014011237 Jan 2014 WO
WO 2014011901 Jan 2014 WO
WO 2014018423 Jan 2014 WO
WO 2014020608 Feb 2014 WO
WO 2014022120 Feb 2014 WO
WO 2014022702 Feb 2014 WO
WO 2014036219 Mar 2014 WO
WO 2014039513 Mar 2014 WO
WO 2014039523 Mar 2014 WO
WO 2014039585 Mar 2014 WO
WO 2014039684 Mar 2014 WO
WO 2014039692 Mar 2014 WO
WO 2014039702 Mar 2014 WO
WO 2014039872 Mar 2014 WO
WO 2014039970 Mar 2014 WO
WO 2014041327 Mar 2014 WO
WO 2014043143 Mar 2014 WO
WO 2014047103 Mar 2014 WO
WO 2014055782 Apr 2014 WO
WO 2014059173 Apr 2014 WO
WO 2014059255 Apr 2014 WO
WO 2014065596 May 2014 WO
WO 2014066505 May 2014 WO
WO 2014068346 May 2014 WO
WO 2014070887 May 2014 WO
WO 2014071006 May 2014 WO
WO 2014071219 May 2014 WO
WO 2014071235 May 2014 WO
WO 2014072941 May 2014 WO
WO 2014081729 May 2014 WO
WO 2014081730 May 2014 WO
WO 2014081855 May 2014 WO
WO 2014082644 Jun 2014 WO
WO 2014085261 Jun 2014 WO
WO 2014085593 Jun 2014 WO
WO 2014085830 Jun 2014 WO
WO 2014089212 Jun 2014 WO
WO 2014089290 Jun 2014 WO
WO 2014089348 Jun 2014 WO
WO 2014089513 Jun 2014 WO
WO 2014089533 Jun 2014 WO
WO 2014089541 Jun 2014 WO
WO 2014093479 Jun 2014 WO
WO 2014093595 Jun 2014 WO
WO 2014093622 Jun 2014 WO
WO 2014093635 Jun 2014 WO
WO 2014093655 Jun 2014 WO
WO 2014093661 Jun 2014 WO
WO 2014093694 Jun 2014 WO
WO 2014093701 Jun 2014 WO
WO 2014093709 Jun 2014 WO
WO 2014093712 Jun 2014 WO
WO 2014093718 Jun 2014 WO
WO 2014093736 Jun 2014 WO
WO 2014093768 Jun 2014 WO
WO 2014093852 Jun 2014 WO
WO 2014096972 Jun 2014 WO
WO 2014099744 Jun 2014 WO
WO 2014099750 Jun 2014 WO
WO 2014104878 Jul 2014 WO
WO 2014110006 Jul 2014 WO
WO 2014110552 Jul 2014 WO
WO 2014113493 Jul 2014 WO
WO 2014123967 Aug 2014 WO
WO 2014124226 Aug 2014 WO
WO 2014125668 Aug 2014 WO
WO 2014127287 Aug 2014 WO
WO 2014128324 Aug 2014 WO
WO 2014128659 Aug 2014 WO
WO 2014130706 Aug 2014 WO
WO 2014130955 Aug 2014 WO
WO 2014131833 Sep 2014 WO
WO 2014138379 Sep 2014 WO
WO 2014143381 Sep 2014 WO
WO 2014144094 Sep 2014 WO
WO 2014144155 Sep 2014 WO
WO 2014144288 Sep 2014 WO
WO 2014144592 Sep 2014 WO
WO 2014144761 Sep 2014 WO
WO 2014144951 Sep 2014 WO
WO 2014145599 Sep 2014 WO
WO 2014145736 Sep 2014 WO
WO 2014150624 Sep 2014 WO
WO 2014152432 Sep 2014 WO
WO 2014152940 Sep 2014 WO
WO 2014153118 Sep 2014 WO
WO 2014153470 Sep 2014 WO
WO 2014158593 Oct 2014 WO
WO 2014161821 Oct 2014 WO
WO 2014164466 Oct 2014 WO
WO 2014165177 Oct 2014 WO
WO 2014165349 Oct 2014 WO
WO 2014165612 Oct 2014 WO
WO 2014165707 Oct 2014 WO
WO 2014165825 Oct 2014 WO
WO 2014172458 Oct 2014 WO
WO 2014172470 Oct 2014 WO
WO 2014172489 Oct 2014 WO
WO 2014173955 Oct 2014 WO
WO 2014182700 Nov 2014 WO
WO 2014183071 Nov 2014 WO
WO 2014184143 Nov 2014 WO
WO 2014184741 Nov 2014 WO
WO 2014184744 Nov 2014 WO
WO 2014186585 Nov 2014 WO
WO 2014186686 Nov 2014 WO
WO 2014190181 Nov 2014 WO
WO 2014191128 Dec 2014 WO
WO 2014191518 Dec 2014 WO
WO 2014191521 Dec 2014 WO
WO 2014191525 Dec 2014 WO
WO 2014191527 Dec 2014 WO
WO 2014193583 Dec 2014 WO
WO 2014194190 Dec 2014 WO
WO 2014197568 Dec 2014 WO
WO 2014197748 Dec 2014 WO
WO 2014199358 Dec 2014 WO
WO 2014200659 Dec 2014 WO
WO 2014201015 Dec 2014 WO
WO 2014204578 Dec 2014 WO
WO 2014204723 Dec 2014 WO
WO 2014204724 Dec 2014 WO
WO 2014204725 Dec 2014 WO
WO 2014204726 Dec 2014 WO
WO 2014204727 Dec 2014 WO
WO 2014204728 Dec 2014 WO
WO 2014204729 Dec 2014 WO
WO 2014205192 Dec 2014 WO
WO 2014207043 Dec 2014 WO
WO 2015002780 Jan 2015 WO
WO 2015004241 Jan 2015 WO
WO 2015006290 Jan 2015 WO
WO 2015006294 Jan 2015 WO
WO 2015006437 Jan 2015 WO
WO 2015006498 Jan 2015 WO
WO 2015006747 Jan 2015 WO
WO 2015007194 Jan 2015 WO
WO 2015010114 Jan 2015 WO
WO 2015011483 Jan 2015 WO
WO 2015013583 Jan 2015 WO
WO 2015017866 Feb 2015 WO
WO 2015018503 Feb 2015 WO
WO 2015021353 Feb 2015 WO
WO 2015021426 Feb 2015 WO
WO 2015021990 Feb 2015 WO
WO 2015024017 Feb 2015 WO
WO 2015024986 Feb 2015 WO
WO 2015026883 Feb 2015 WO
WO 2015026885 Feb 2015 WO
WO 2015026886 Feb 2015 WO
WO 2015026887 Feb 2015 WO
WO 2015027134 Feb 2015 WO
WO 2015028969 Mar 2015 WO
WO 2015030881 Mar 2015 WO
WO 2015031619 Mar 2015 WO
WO 2015031775 Mar 2015 WO
WO 2015032494 Mar 2015 WO
WO 2015033293 Mar 2015 WO
WO 2015034872 Mar 2015 WO
WO 2015034885 Mar 2015 WO
WO 2015035136 Mar 2015 WO
WO 2015035139 Mar 2015 WO
WO 2015035162 Mar 2015 WO
WO 2015040075 Mar 2015 WO
WO 2015040402 Mar 2015 WO
WO 2015042393 Mar 2015 WO
WO 2015042585 Mar 2015 WO
WO 2015048577 Apr 2015 WO
WO 2015048690 Apr 2015 WO
WO 2015048707 Apr 2015 WO
WO 2015048801 Apr 2015 WO
WO 2015049897 Apr 2015 WO
WO 2015051191 Apr 2015 WO
WO 2015052133 Apr 2015 WO
WO 2015052231 Apr 2015 WO
WO 2015052335 Apr 2015 WO
WO 2015053995 Apr 2015 WO
WO 2015054253 Apr 2015 WO
WO 2015054315 Apr 2015 WO
WO 2015057671 Apr 2015 WO
WO 2015057834 Apr 2015 WO
WO 2015057852 Apr 2015 WO
WO 2015057976 Apr 2015 WO
WO 2015057980 Apr 2015 WO
WO 2015059265 Apr 2015 WO
WO 2015065964 May 2015 WO
WO 2015066119 May 2015 WO
WO 2015066634 May 2015 WO
WO 2015066636 May 2015 WO
WO 2015066637 May 2015 WO
WO 2015066638 May 2015 WO
WO 2015066643 May 2015 WO
WO 2015069682 May 2015 WO
WO 2015070083 May 2015 WO
WO 2015070193 May 2015 WO
WO 2015070212 May 2015 WO
WO 2015071474 May 2015 WO
WO 2015073683 May 2015 WO
WO 2015073867 May 2015 WO
WO 2015073990 May 2015 WO
WO 2015075056 May 2015 WO
WO 2015075154 May 2015 WO
WO 2015075175 May 2015 WO
WO 2015075195 May 2015 WO
WO 2015075557 May 2015 WO
WO 2015077058 May 2015 WO
WO 2015077290 May 2015 WO
WO 2015077318 May 2015 WO
WO 2015079056 Jun 2015 WO
WO 2015079057 Jun 2015 WO
WO 2015086795 Jun 2015 WO
WO 2015086798 Jun 2015 WO
WO 2015088643 Jun 2015 WO
WO 2015089046 Jun 2015 WO
WO 2015089077 Jun 2015 WO
WO 2015089277 Jun 2015 WO
WO 2015089351 Jun 2015 WO
WO 2015089354 Jun 2015 WO
WO 2015089364 Jun 2015 WO
WO 2015089406 Jun 2015 WO
WO 2015089419 Jun 2015 WO
WO 2015089427 Jun 2015 WO
WO 2015089462 Jun 2015 WO
WO 2015089465 Jun 2015 WO
WO 2015089473 Jun 2015 WO
WO 2015089486 Jun 2015 WO
WO 2015095804 Jun 2015 WO
WO 2015099850 Jul 2015 WO
WO 2015100929 Jul 2015 WO
WO 2015103057 Jul 2015 WO
WO 2015103153 Jul 2015 WO
WO 2015105928 Jul 2015 WO
WO 2015108993 Jul 2015 WO
WO 2015109752 Jul 2015 WO
WO 2015110474 Jul 2015 WO
WO 2015112790 Jul 2015 WO
WO 2015112896 Jul 2015 WO
WO 2015113063 Jul 2015 WO
WO 2015114365 Aug 2015 WO
WO 2015115903 Aug 2015 WO
WO 2015116686 Aug 2015 WO
WO 2015116969 Aug 2015 WO
WO 2015117021 Aug 2015 WO
WO 2015117041 Aug 2015 WO
WO 2015117081 Aug 2015 WO
WO 2015118156 Aug 2015 WO
WO 2015119941 Aug 2015 WO
WO 2015121454 Aug 2015 WO
WO 2015122967 Aug 2015 WO
WO 2015123339 Aug 2015 WO
WO 2015124715 Aug 2015 WO
WO 2015124718 Aug 2015 WO
WO 2015126927 Aug 2015 WO
WO 2015127428 Aug 2015 WO
WO 2015127439 Aug 2015 WO
WO 2015129686 Sep 2015 WO
WO 2015131101 Sep 2015 WO
WO 2015133554 Sep 2015 WO
WO 2015134121 Sep 2015 WO
WO 2015134812 Sep 2015 WO
WO 2015136001 Sep 2015 WO
WO 2015138510 Sep 2015 WO
WO 2015138739 Sep 2015 WO
WO 2015138855 Sep 2015 WO
WO 2015138870 Sep 2015 WO
WO 2015139008 Sep 2015 WO
WO 2015139139 Sep 2015 WO
WO 2015143046 Sep 2015 WO
WO 2015143177 Sep 2015 WO
WO 2015145417 Oct 2015 WO
WO 2015148431 Oct 2015 WO
WO 2015148670 Oct 2015 WO
WO 2015148680 Oct 2015 WO
WO 2015148761 Oct 2015 WO
WO 2015148860 Oct 2015 WO
WO 2015148863 Oct 2015 WO
WO 2015153760 Oct 2015 WO
WO 2015153780 Oct 2015 WO
WO 2015153789 Oct 2015 WO
WO 2015153791 Oct 2015 WO
WO 2015153889 Oct 2015 WO
WO 2015153940 Oct 2015 WO
WO 2015155341 Oct 2015 WO
WO 2015155686 Oct 2015 WO
WO 2015157070 Oct 2015 WO
WO 2015157534 Oct 2015 WO
WO 2015159068 Oct 2015 WO
WO 2015159086 Oct 2015 WO
WO 2015159087 Oct 2015 WO
WO 2015160683 Oct 2015 WO
WO 2015161276 Oct 2015 WO
WO 2015163733 Oct 2015 WO
WO 2015164740 Oct 2015 WO
WO 2015164748 Oct 2015 WO
WO 2015165274 Nov 2015 WO
WO 2015165275 Nov 2015 WO
WO 2015165276 Nov 2015 WO
WO 2015166272 Nov 2015 WO
WO 2015167766 Nov 2015 WO
WO 2015167956 Nov 2015 WO
WO 2015168125 Nov 2015 WO
WO 2015168158 Nov 2015 WO
WO 2015168404 Nov 2015 WO
WO 2015168547 Nov 2015 WO
WO 2015168800 Nov 2015 WO
WO 2015171603 Nov 2015 WO
WO 2015171894 Nov 2015 WO
WO 2015171932 Nov 2015 WO
WO 2015172128 Nov 2015 WO
WO 2015173436 Nov 2015 WO
WO 2015175642 Nov 2015 WO
WO 2015179540 Nov 2015 WO
WO 2015183025 Dec 2015 WO
WO 2015183026 Dec 2015 WO
WO 2015183885 Dec 2015 WO
WO 2015184259 Dec 2015 WO
WO 2015184262 Dec 2015 WO
WO 2015184268 Dec 2015 WO
WO 2015188056 Dec 2015 WO
WO 2015188065 Dec 2015 WO
WO 2015188094 Dec 2015 WO
WO 2015188109 Dec 2015 WO
WO 2015188132 Dec 2015 WO
WO 2015188135 Dec 2015 WO
WO 2015188191 Dec 2015 WO
WO 2015189693 Dec 2015 WO
WO 2015191693 Dec 2015 WO
WO 2015191899 Dec 2015 WO
WO 2015191911 Dec 2015 WO
WO 2015193858 Dec 2015 WO
WO 2015195547 Dec 2015 WO
WO 2015195621 Dec 2015 WO
WO 2015195798 Dec 2015 WO
WO 2015198020 Dec 2015 WO
WO 2015200334 Dec 2015 WO
WO 2015200378 Dec 2015 WO
WO 2015200555 Dec 2015 WO
WO 2015200805 Dec 2015 WO
WO 2016001978 Jan 2016 WO
WO 2016004010 Jan 2016 WO
WO 2016004318 Jan 2016 WO
WO 2016007347 Jan 2016 WO
WO 2016007604 Jan 2016 WO
WO 2016007948 Jan 2016 WO
WO 2016011080 Jan 2016 WO
WO 2016011210 Jan 2016 WO
WO 2016011428 Jan 2016 WO
WO 2016012544 Jan 2016 WO
WO 2016012552 Jan 2016 WO
WO 2016014409 Jan 2016 WO
WO 2016014565 Jan 2016 WO
WO 2016014794 Jan 2016 WO
WO 2016014837 Jan 2016 WO
WO 2016016119 Feb 2016 WO
WO 2016016358 Feb 2016 WO
WO 2016019144 Feb 2016 WO
WO 2016020399 Feb 2016 WO
WO 2016021972 Feb 2016 WO
WO 2016021973 Feb 2016 WO
WO 2016022363 Feb 2016 WO
WO 2016022866 Feb 2016 WO
WO 2016022931 Feb 2016 WO
WO 2016025131 Feb 2016 WO
WO 2016025469 Feb 2016 WO
WO 2016025759 Feb 2016 WO
WO 2016026444 Feb 2016 WO
WO 2016028682 Feb 2016 WO
WO 2016028843 Feb 2016 WO
WO 2016028887 Feb 2016 WO
WO 2016033088 Mar 2016 WO
WO 2016033230 Mar 2016 WO
WO 2016033246 Mar 2016 WO
WO 2016033298 Mar 2016 WO
WO 2016035044 Mar 2016 WO
WO 2016036754 Mar 2016 WO
WO 2016037157 Mar 2016 WO
WO 2016040030 Mar 2016 WO
WO 2016040594 Mar 2016 WO
WO 2016044182 Mar 2016 WO
WO 2016044416 Mar 2016 WO
WO 2016046635 Mar 2016 WO
WO 2016049024 Mar 2016 WO
WO 2016049163 Mar 2016 WO
WO 2016049230 Mar 2016 WO
WO 2016049251 Mar 2016 WO
WO 2016049258 Mar 2016 WO
WO 2016053397 Apr 2016 WO
WO 2016054326 Apr 2016 WO
WO 2016057061 Apr 2016 WO
WO 2016057821 Apr 2016 WO
WO 2016057835 Apr 2016 WO
WO 2016057850 Apr 2016 WO
WO 2016057951 Apr 2016 WO
WO 2016057961 Apr 2016 WO
WO 2016061073 Apr 2016 WO
WO 2016061374 Apr 2016 WO
WO 2016061481 Apr 2016 WO
WO 2016061523 Apr 2016 WO
WO 2016064894 Apr 2016 WO
WO 2016065364 Apr 2016 WO
WO 2016069282 May 2016 WO
WO 2016069283 May 2016 WO
WO 2016069591 May 2016 WO
WO 2016069774 May 2016 WO
WO 2016069910 May 2016 WO
WO 2016069912 May 2016 WO
WO 2016070037 May 2016 WO
WO 2016070070 May 2016 WO
WO 2016070129 May 2016 WO
WO 2016072399 May 2016 WO
WO 2016072936 May 2016 WO
WO 2016073433 May 2016 WO
WO 2016073559 May 2016 WO
WO 2016073990 May 2016 WO
WO 2016075662 May 2016 WO
WO 2016076672 May 2016 WO
WO 2016077273 May 2016 WO
WO 2016077350 May 2016 WO
WO 2016080097 May 2016 WO
WO 2016080795 May 2016 WO
WO 2016081923 May 2016 WO
WO 2016081924 May 2016 WO
WO 2016082135 Jun 2016 WO
WO 2016083811 Jun 2016 WO
WO 2016084084 Jun 2016 WO
WO 2016084088 Jun 2016 WO
WO 2016086177 Jun 2016 WO
WO 2016089433 Jun 2016 WO
WO 2016089866 Jun 2016 WO
WO 2016089883 Jun 2016 WO
WO 2016090385 Jun 2016 WO
WO 2016094679 Jun 2016 WO
WO 2016094845 Jun 2016 WO
WO 2016094867 Jun 2016 WO
WO 2016094872 Jun 2016 WO
WO 2016094874 Jun 2016 WO
WO 2016094880 Jun 2016 WO
WO 2016094888 Jun 2016 WO
WO 2016097212 Jun 2016 WO
WO 2016097231 Jun 2016 WO
WO 2016097751 Jun 2016 WO
WO 2016099887 Jun 2016 WO
WO 2016100272 Jun 2016 WO
WO 2016100389 Jun 2016 WO
WO 2016100568 Jun 2016 WO
WO 2016100571 Jun 2016 WO
WO 2016100951 Jun 2016 WO
WO 2016100955 Jun 2016 WO
WO 2016100974 Jun 2016 WO
WO 2016103233 Jun 2016 WO
WO 2016104716 Jun 2016 WO
WO 2016106236 Jun 2016 WO
WO 2016106239 Jun 2016 WO
WO 2016106244 Jun 2016 WO
WO 2016106338 Jun 2016 WO
WO 2016108926 Jul 2016 WO
WO 2016109255 Jul 2016 WO
WO 2016109840 Jul 2016 WO
WO 2016110214 Jul 2016 WO
WO 2016110453 Jul 2016 WO
WO 2016110511 Jul 2016 WO
WO 2016110512 Jul 2016 WO
WO 2016111546 Jul 2016 WO
WO 2016112242 Jul 2016 WO
WO 2016112351 Jul 2016 WO
WO 2016112963 Jul 2016 WO
WO 2016113357 Jul 2016 WO
WO 2016114972 Jul 2016 WO
WO 2016115179 Jul 2016 WO
WO 2016115326 Jul 2016 WO
WO 2016115355 Jul 2016 WO
WO 2016116032 Jul 2016 WO
WO 2016120480 Aug 2016 WO
WO 2016123071 Aug 2016 WO
WO 2016123230 Aug 2016 WO
WO 2016123243 Aug 2016 WO
WO 2016123578 Aug 2016 WO
WO 2016126747 Aug 2016 WO
WO 2016130600 Aug 2016 WO
WO 2016130697 Aug 2016 WO
WO 2016131009 Aug 2016 WO
WO 2016132122 Aug 2016 WO
WO 2016133165 Aug 2016 WO
WO 2016135507 Sep 2016 WO
WO 2016135557 Sep 2016 WO
WO 2016135558 Sep 2016 WO
WO 2016135559 Sep 2016 WO
WO 2016137774 Sep 2016 WO
WO 2016137949 Sep 2016 WO
WO 2016141224 Sep 2016 WO
WO 2016141893 Sep 2016 WO
WO 2016142719 Sep 2016 WO
WO 2016145150 Sep 2016 WO
WO 2016148994 Sep 2016 WO
WO 2016149484 Sep 2016 WO
WO 2016149547 Sep 2016 WO
WO 2016150336 Sep 2016 WO
WO 2016150855 Sep 2016 WO
WO 2016154016 Sep 2016 WO
WO 2016154579 Sep 2016 WO
WO 2016154596 Sep 2016 WO
WO 2016155482 Oct 2016 WO
WO 2016161004 Oct 2016 WO
WO 2016161207 Oct 2016 WO
WO 2016161260 Oct 2016 WO
WO 2016161380 Oct 2016 WO
WO 2016161446 Oct 2016 WO
WO 2016164356 Oct 2016 WO
WO 2016164797 Oct 2016 WO
WO 2016166340 Oct 2016 WO
WO 2016167300 Oct 2016 WO
WO 2016168631 Oct 2016 WO
WO 2016170484 Oct 2016 WO
WO 2016172359 Oct 2016 WO
WO 2016172727 Oct 2016 WO
WO 2016174056 Nov 2016 WO
WO 2016174151 Nov 2016 WO
WO 2016174250 Nov 2016 WO
WO 2016176191 Nov 2016 WO
WO 2016176404 Nov 2016 WO
WO 2016176690 Nov 2016 WO
WO 2016177682 Nov 2016 WO
WO 2016178207 Nov 2016 WO
WO 2016179038 Nov 2016 WO
WO 2016179112 Nov 2016 WO
WO 2016181357 Nov 2016 WO
WO 2016182893 Nov 2016 WO
WO 2016182917 Nov 2016 WO
WO 2016182959 Nov 2016 WO
WO 2016183236 Nov 2016 WO
WO 2016183298 Nov 2016 WO
WO 2016183345 Nov 2016 WO
WO 2016183402 Nov 2016 WO
WO 2016183438 Nov 2016 WO
WO 2016183448 Nov 2016 WO
WO 2016184955 Nov 2016 WO
WO 2016184989 Nov 2016 WO
WO 2016185411 Nov 2016 WO
WO 2016186745 Nov 2016 WO
WO 2016186772 Nov 2016 WO
WO 2016186946 Nov 2016 WO
WO 2016186953 Nov 2016 WO
WO 2016187717 Dec 2016 WO
WO 2016187904 Dec 2016 WO
WO 2016191684 Dec 2016 WO
WO 2016191869 Dec 2016 WO
WO 2016196273 Dec 2016 WO
WO 2016196282 Dec 2016 WO
WO 2016196308 Dec 2016 WO
WO 2016196361 Dec 2016 WO
WO 2016196499 Dec 2016 WO
WO 2016196539 Dec 2016 WO
WO 2016196655 Dec 2016 WO
WO 2016196805 Dec 2016 WO
WO 2016196887 Dec 2016 WO
WO 2016197132 Dec 2016 WO
WO 2016197133 Dec 2016 WO
WO 2016197354 Dec 2016 WO
WO 2016197355 Dec 2016 WO
WO 2016197356 Dec 2016 WO
WO 2016197357 Dec 2016 WO
WO 2016197358 Dec 2016 WO
WO 2016197359 Dec 2016 WO
WO 2016197360 Dec 2016 WO
WO 2016197361 Dec 2016 WO
WO 2016197362 Dec 2016 WO
WO 2016198361 Dec 2016 WO
WO 2016198500 Dec 2016 WO
WO 2016200263 Dec 2016 WO
WO 2016201047 Dec 2016 WO
WO 2016201138 Dec 2016 WO
WO 2016201152 Dec 2016 WO
WO 2016201153 Dec 2016 WO
WO 2016201155 Dec 2016 WO
WO 2016205276 Dec 2016 WO
WO 2016205613 Dec 2016 WO
WO 2016205623 Dec 2016 WO
WO 2016205680 Dec 2016 WO
WO 2016205688 Dec 2016 WO
WO 2016205703 Dec 2016 WO
WO 2016205711 Dec 2016 WO
WO 2016205728 Dec 2016 WO
WO 2016205745 Dec 2016 WO
WO 2016205749 Dec 2016 WO
WO 2016205759 Dec 2016 WO
WO 2016205764 Dec 2016 WO
WO 2017001572 Jan 2017 WO
WO 2017001988 Jan 2017 WO
WO 2017004261 Jan 2017 WO
WO 2017004279 Jan 2017 WO
WO 2017004616 Jan 2017 WO
WO 2017005807 Jan 2017 WO
WO 2017009399 Jan 2017 WO
WO 2017010556 Jan 2017 WO
WO 2017011519 Jan 2017 WO
WO 2017011721 Jan 2017 WO
WO 2017011804 Jan 2017 WO
WO 2017015015 Jan 2017 WO
WO 2017015101 Jan 2017 WO
WO 2017015545 Jan 2017 WO
WO 2017015567 Jan 2017 WO
WO 2017015637 Jan 2017 WO
WO 2017017016 Feb 2017 WO
WO 2017019867 Feb 2017 WO
WO 2017019895 Feb 2017 WO
WO 2017023803 Feb 2017 WO
WO 2017023974 Feb 2017 WO
WO 2017024047 Feb 2017 WO
WO 2017024319 Feb 2017 WO
WO 2017024343 Feb 2017 WO
WO 2017024602 Feb 2017 WO
WO 2017025323 Feb 2017 WO
WO 2017027423 Feb 2017 WO
WO 2017028768 Feb 2017 WO
WO 2017029664 Feb 2017 WO
WO 2017031360 Feb 2017 WO
WO 2017031483 Feb 2017 WO
WO 2017035416 Mar 2017 WO
WO 2017040348 Mar 2017 WO
WO 2017040511 Mar 2017 WO
WO 2017040709 Mar 2017 WO
WO 2017040786 Mar 2017 WO
WO 2017040793 Mar 2017 WO
WO 2017040813 Mar 2017 WO
WO 2017043573 Mar 2017 WO
WO 2017043656 Mar 2017 WO
WO 2017044419 Mar 2017 WO
WO 2017044776 Mar 2017 WO
WO 2017044857 Mar 2017 WO
WO 2017048390 Mar 2017 WO
WO 2017049129 Mar 2017 WO
WO 2017050963 Mar 2017 WO
WO 2017053312 Mar 2017 WO
WO 2017053431 Mar 2017 WO
WO 2017053713 Mar 2017 WO
WO 2017053729 Mar 2017 WO
WO 2017053753 Mar 2017 WO
WO 2017053762 Mar 2017 WO
WO 2017053879 Mar 2017 WO
WO 2017054721 Apr 2017 WO
WO 2017058658 Apr 2017 WO
WO 2017059241 Apr 2017 WO
WO 2017062605 Apr 2017 WO
WO 2017062723 Apr 2017 WO
WO 2017062754 Apr 2017 WO
WO 2017062855 Apr 2017 WO
WO 2017062886 Apr 2017 WO
WO 2017062983 Apr 2017 WO
WO 2017064439 Apr 2017 WO
WO 2017064546 Apr 2017 WO
WO 2017064566 Apr 2017 WO
WO 2017066175 Apr 2017 WO
WO 2017066497 Apr 2017 WO
WO 2017066588 Apr 2017 WO
WO 2017066707 Apr 2017 WO
WO 2017066781 Apr 2017 WO
WO 2017068077 Apr 2017 WO
WO 2017068377 Apr 2017 WO
WO 2017069829 Apr 2017 WO
WO 2017070029 Apr 2017 WO
WO 2017070032 Apr 2017 WO
WO 2017070169 Apr 2017 WO
WO 2017070284 Apr 2017 WO
WO 2017070598 Apr 2017 WO
WO 2017070605 Apr 2017 WO
WO 2017070632 Apr 2017 WO
WO 2017070633 Apr 2017 WO
WO 2017072590 May 2017 WO
WO 2017074526 May 2017 WO
WO 2017074962 May 2017 WO
WO 2017075261 May 2017 WO
WO 2017075335 May 2017 WO
WO 2017075475 May 2017 WO
WO 2017077135 May 2017 WO
WO 2017077329 May 2017 WO
WO 2017078751 May 2017 WO
WO 2017079400 May 2017 WO
WO 2017079428 May 2017 WO
WO 2017079673 May 2017 WO
WO 2017079724 May 2017 WO
WO 2017081097 May 2017 WO
WO 2017081288 May 2017 WO
WO 2017083368 May 2017 WO
WO 2017083722 May 2017 WO
WO 2017083766 May 2017 WO
WO 2017087395 May 2017 WO
WO 2017090724 Jun 2017 WO
WO 2017091510 Jun 2017 WO
WO 2017091630 Jun 2017 WO
WO 2017092201 Jun 2017 WO
WO 2017093370 Jun 2017 WO
WO 2017093969 Jun 2017 WO
WO 2017095111 Jun 2017 WO
WO 2017096041 Jun 2017 WO
WO 2017096237 Jun 2017 WO
WO 2017100158 Jun 2017 WO
WO 2017100431 Jun 2017 WO
WO 2017104404 Jun 2017 WO
WO 2017105251 Jun 2017 WO
WO 2017105350 Jun 2017 WO
WO 2017105991 Jun 2017 WO
WO 2017106414 Jun 2017 WO
WO 2017106528 Jun 2017 WO
WO 2017106537 Jun 2017 WO
WO 2017106569 Jun 2017 WO
WO 2017106616 Jun 2017 WO
WO 2017106657 Jun 2017 WO
WO 2017106767 Jun 2017 WO
WO 2017109134 Jun 2017 WO
WO 2017109757 Jun 2017 WO
WO 2017112620 Jun 2017 WO
WO 2017115268 Jul 2017 WO
WO 2017117395 Jul 2017 WO
WO 2017118598 Jul 2017 WO
WO 2017118720 Jul 2017 WO
WO 2017123609 Jul 2017 WO
WO 2017123910 Jul 2017 WO
WO 2017124086 Jul 2017 WO
WO 2017124100 Jul 2017 WO
WO 2017124652 Jul 2017 WO
WO 2017126987 Jul 2017 WO
WO 2017127807 Jul 2017 WO
WO 2017131237 Aug 2017 WO
WO 2017132112 Aug 2017 WO
WO 2017132580 Aug 2017 WO
WO 2017136520 Aug 2017 WO
WO 2017136629 Aug 2017 WO
WO 2017136794 Aug 2017 WO
WO 2017139264 Aug 2017 WO
WO 2017139505 Aug 2017 WO
WO 2017141173 Aug 2017 WO
WO 2017142835 Aug 2017 WO
WO 2017142999 Aug 2017 WO
WO 2017143042 Aug 2017 WO
WO 2017147056 Aug 2017 WO
WO 2017147278 Aug 2017 WO
WO 2017147432 Aug 2017 WO
WO 2017147446 Aug 2017 WO
WO 2017147555 Aug 2017 WO
WO 2017151444 Sep 2017 WO
WO 2017151719 Sep 2017 WO
WO 2017152015 Sep 2017 WO
WO 2017155717 Sep 2017 WO
WO 2017157422 Sep 2017 WO
WO 2017158153 Sep 2017 WO
WO 2017160689 Sep 2017 WO
WO 2017160752 Sep 2017 WO
WO 2017160890 Sep 2017 WO
WO 2017161068 Sep 2017 WO
WO 2017165826 Sep 2017 WO
WO 2017165862 Sep 2017 WO
WO 2017167712 Oct 2017 WO
WO 2017172644 Oct 2017 WO
WO 2017172645 Oct 2017 WO
WO 2017172860 Oct 2017 WO
WO 2017173004 Oct 2017 WO
WO 2017173054 Oct 2017 WO
WO 2017173092 Oct 2017 WO
WO 2017174329 Oct 2017 WO
WO 2017176529 Oct 2017 WO
WO 2017176806 Oct 2017 WO
WO 2017178590 Oct 2017 WO
WO 2017180694 Oct 2017 WO
WO 2017180711 Oct 2017 WO
WO 2017180915 Oct 2017 WO
WO 2017180926 Oct 2017 WO
WO 2017181107 Oct 2017 WO
WO 2017181735 Oct 2017 WO
WO 2017182468 Oct 2017 WO
WO 2017184334 Oct 2017 WO
WO 2017184768 Oct 2017 WO
WO 2017184786 Oct 2017 WO
WO 2017186550 Nov 2017 WO
WO 2017189308 Nov 2017 WO
WO 2017189336 Nov 2017 WO
WO 2017190041 Nov 2017 WO
WO 2017190257 Nov 2017 WO
WO 2017190664 Nov 2017 WO
WO 2017191210 Nov 2017 WO
WO 2017191274 Nov 2017 WO
WO 2017192172 Nov 2017 WO
WO 2017192512 Nov 2017 WO
WO 2017192544 Nov 2017 WO
WO 2017192573 Nov 2017 WO
WO 2017193029 Nov 2017 WO
WO 2017193053 Nov 2017 WO
WO 2017196768 Nov 2017 WO
WO 2017197038 Nov 2017 WO
WO 2017197238 Nov 2017 WO
WO 2017197301 Nov 2017 WO
WO 2017201476 Nov 2017 WO
WO 2017205290 Nov 2017 WO
WO 2017205423 Nov 2017 WO
WO 2017207589 Dec 2017 WO
WO 2017208247 Dec 2017 WO
WO 2017209809 Dec 2017 WO
WO 2017213896 Dec 2017 WO
WO 2017213898 Dec 2017 WO
WO 2017214460 Dec 2017 WO
WO 2017216392 Dec 2017 WO
WO 2017216771 Dec 2017 WO
WO 2017218185 Dec 2017 WO
WO 2017219027 Dec 2017 WO
WO 2017219033 Dec 2017 WO
WO 2017220751 Dec 2017 WO
WO 2017222370 Dec 2017 WO
WO 2017222773 Dec 2017 WO
WO 2017222834 Dec 2017 WO
WO 2017223107 Dec 2017 WO
WO 2017223330 Dec 2017 WO
WO 2018000657 Jan 2018 WO
WO 2018002719 Jan 2018 WO
WO 2018005117 Jan 2018 WO
WO 2018005289 Jan 2018 WO
WO 2018005691 Jan 2018 WO
WO 2018005782 Jan 2018 WO
WO 2018005873 Jan 2018 WO
WO 201806693 Jan 2018 WO
WO 2018009520 Jan 2018 WO
WO 2018009562 Jan 2018 WO
WO 2018009822 Jan 2018 WO
WO 2018013821 Jan 2018 WO
WO 2018013932 Jan 2018 WO
WO 2018013990 Jan 2018 WO
WO 2018014384 Jan 2018 WO
WO 2018015444 Jan 2018 WO
WO 2018015936 Jan 2018 WO
WO 2018017754 Jan 2018 WO
WO 2018018979 Feb 2018 WO
WO 2018020248 Feb 2018 WO
WO 2018021878 Feb 2018 WO
WO 2018022480 Feb 2018 WO
WO 2018022634 Feb 2018 WO
WO 2018025206 Feb 2018 WO
WO 2018026723 Feb 2018 WO
WO 2018026976 Feb 2018 WO
WO 2018027078 Feb 2018 WO
WO 2018030608 Feb 2018 WO
WO 2018031683 Feb 2018 WO
WO 2018035250 Feb 2018 WO
WO 2018035300 Feb 2018 WO
WO 2018035423 Feb 2018 WO
WO 2018035503 Feb 2018 WO
WO 2018039145 Mar 2018 WO
WO 2018039438 Mar 2018 WO
WO 2018039440 Mar 2018 WO
WO 2018039448 Mar 2018 WO
WO 2018045630 Mar 2018 WO
WO 2018048827 Mar 2018 WO
WO 2018049073 Mar 2018 WO
WO 2018049168 Mar 2018 WO
WO 2018051347 Mar 2018 WO
WO 2018058064 Mar 2018 WO
WO 2018062866 Apr 2018 WO
WO 2018064352 Apr 2018 WO
WO 2018064371 Apr 2018 WO
WO 2018064516 Apr 2018 WO
WO 2018067546 Apr 2018 WO
WO 2018067846 Apr 2018 WO
WO 2018068053 Apr 2018 WO
WO 2018069474 Apr 2018 WO
WO 2018071623 Apr 2018 WO
WO 2018071663 Apr 2018 WO
WO 2018071868 Apr 2018 WO
WO 2018071892 Apr 2018 WO
WO 2018074979 Apr 2018 WO
WO 2018079134 May 2018 WO
WO 2018080573 May 2018 WO
WO 2018081504 May 2018 WO
WO 2018081535 May 2018 WO
WO 2018081728 May 2018 WO
WO 2018083128 May 2018 WO
WO 2018083606 May 2018 WO
WO 2018085288 May 2018 WO
WO 2018085414 May 2018 WO
WO 2018086623 May 2018 WO
WO 2018089664 May 2018 WO
WO 2018093990 May 2018 WO
WO 2018098383 May 2018 WO
WO 2018098480 May 2018 WO
WO 2018098587 Jun 2018 WO
WO 2018099256 Jun 2018 WO
WO 2018103686 Jun 2018 WO
WO 2018106268 Jun 2018 WO
WO 2018107028 Jun 2018 WO
WO 2018107103 Jun 2018 WO
WO 2018107129 Jun 2018 WO
WO 2018108272 Jun 2018 WO
WO 2018109101 Jun 2018 WO
WO 2018111946 Jun 2018 WO
WO 2018111947 Jun 2018 WO
WO 2018112336 Jun 2018 WO
WO 2018112446 Jun 2018 WO
WO 2018119354 Jun 2018 WO
WO 2018119359 Jun 2018 WO
WO 2018120283 Jul 2018 WO
WO 2018130830 Jul 2018 WO
WO 2018135838 Jul 2018 WO
WO 2018136396 Jul 2018 WO
WO 2018138385 Aug 2018 WO
WO 2018142364 Aug 2018 WO
WO 2018148246 Aug 2018 WO
WO 2018148256 Aug 2018 WO
WO 2018148647 Aug 2018 WO
WO 2018149418 Aug 2018 WO
WO 2018149888 Aug 2018 WO
WO 2018149915 Aug 2018 WO
WO 2018152197 Aug 2018 WO
WO 2018152418 Aug 2018 WO
WO 2018154380 Aug 2018 WO
WO 2018154387 Aug 2018 WO
WO 2018154412 Aug 2018 WO
WO 2018154413 Aug 2018 WO
WO 2018154418 Aug 2018 WO
WO 2018154439 Aug 2018 WO
WO 2018154459 Aug 2018 WO
WO 2018154462 Aug 2018 WO
WO 2018156372 Aug 2018 WO
WO 2018156824 Aug 2018 WO
WO 2018161009 Sep 2018 WO
WO 2018165504 Sep 2018 WO
WO 2018165629 Sep 2018 WO
WO 2018170015 Sep 2018 WO
WO 2018170340 Sep 2018 WO
WO 2018175502 Sep 2018 WO
WO 2018176009 Sep 2018 WO
WO 2018177351 Oct 2018 WO
WO 2018179578 Oct 2018 WO
WO 2018183403 Oct 2018 WO
WO 2018189184 Oct 2018 WO
WO 2018191388 Oct 2018 WO
WO 2018195402 Oct 2018 WO
WO 2018195545 Oct 2018 WO
WO 2018195555 Oct 2018 WO
WO 2018197020 Nov 2018 WO
WO 2018197495 Nov 2018 WO
WO 2018202800 Nov 2018 WO
WO 2018204493 Nov 2018 WO
WO 2018208755 Nov 2018 WO
WO 2018208998 Nov 2018 WO
WO 2018209158 Nov 2018 WO
WO 2018209320 Nov 2018 WO
WO 2018213351 Nov 2018 WO
WO 2018213708 Nov 2018 WO
WO 2018213726 Nov 2018 WO
WO 2018213771 Nov 2018 WO
WO 2018213791 Nov 2018 WO
WO 2018217852 Nov 2018 WO
WO 2018217981 Nov 2018 WO
WO 2018218166 Nov 2018 WO
WO 2018218188 Nov 2018 WO
WO 2018218206 Nov 2018 WO
WO 2018226855 Dec 2018 WO
WO 2019005884 Jan 2019 WO
WO 2019005886 Jan 2019 WO
WO 2019010384 Jan 2019 WO
WO 2019023680 Jan 2019 WO
WO 2019051097 Mar 2019 WO
WO 2019075357 Apr 2019 WO
WO 2019079347 Apr 2019 WO
WO 2019084062 May 2019 WO
WO 2019090367 May 2019 WO
WO 2019092042 May 2019 WO
WO 2019118935 Jun 2019 WO
WO 2019118949 Jun 2019 WO
WO 2019123430 Jun 2019 WO
WO 2019139645 Jul 2019 WO
WO 2019139951 Jul 2019 WO
WO 2019147014 Aug 2019 WO
WO 2019161251 Aug 2019 WO
WO 2019168953 Sep 2019 WO
WO 2019183641 Sep 2019 WO
WO 2019204369 Oct 2019 WO
WO 2019217942 Nov 2019 WO
WO 2019226593 Nov 2019 WO
WO 2019226953 Nov 2019 WO
WO 2019236566 Dec 2019 WO
WO 2019241649 Dec 2019 WO
WO 2020014261 Jan 2020 WO
WO 2020028555 Feb 2020 WO
WO 2020028823 Feb 2020 WO
WO 2020041751 Feb 2020 WO
WO 2020047124 Mar 2020 WO
WO 2020051360 Mar 2020 WO
WO 2020081568 Apr 2020 WO
WO 2020086908 Apr 2020 WO
WO 2020092453 May 2020 WO
WO 2020102659 May 2020 WO
WO 2020102709 May 2020 WO
WO 2020154500 Jul 2020 WO
WO 2020157008 Aug 2020 WO
WO 2020160071 Aug 2020 WO
WO 2020160517 Aug 2020 WO
WO 2020180975 Sep 2020 WO
WO 2020181178 Sep 2020 WO
WO 2020181180 Sep 2020 WO
WO 2020181193 Sep 2020 WO
WO 2020181195 Sep 2020 WO
WO 2020181202 Sep 2020 WO
WO 2020191153 Sep 2020 WO
WO 2020191171 Sep 2020 WO
WO 2020191233 Sep 2020 WO
WO 2020191234 Sep 2020 WO
WO 2020191239 Sep 2020 WO
WO 2020191241 Sep 2020 WO
WO 2020191242 Sep 2020 WO
WO 2020191243 Sep 2020 WO
WO 2020191245 Sep 2020 WO
WO 2020191246 Sep 2020 WO
WO 2020191248 Sep 2020 WO
WO 2020191249 Sep 2020 WO
WO 2020210751 Oct 2020 WO
WO 2020214842 Oct 2020 WO
WO 2020236982 Nov 2020 WO
WO 2020247587 Dec 2020 WO
WO 2021022043 Feb 2021 WO
WO 2021025750 Feb 2021 WO
WO 2021030666 Feb 2021 WO
WO 2021042047 Mar 2021 WO
WO 2021042062 Mar 2021 WO
WO 2021072328 Apr 2021 WO
WO 2021080922 Apr 2021 WO
WO 2021081264 Apr 2021 WO
WO 2021087182 May 2021 WO
WO 2021108717 Jun 2021 WO
WO 2021138469 Jul 2021 WO
WO 2021155065 Aug 2021 WO
WO 2021158921 Aug 2021 WO
WO 2021158995 Aug 2021 WO
WO 2021158999 Aug 2021 WO
WO 2021178709 Sep 2021 WO
WO 2021178717 Sep 2021 WO
WO 2021178720 Sep 2021 WO
WO 2021178898 Sep 2021 WO
WO 2021222318 Nov 2021 WO
WO 2021226558 Nov 2021 WO
WO 2023039440 Mar 2023 WO
WO 2023039447 Mar 2023 WO
Non-Patent Literature Citations (1947)
Entry
U.S. Appl. No. 61/716,256, filed Oct. 19, 2012, Jinek et al.
U.S. Appl. No. 61/717,324, filed Oct. 23, 2012, Cho et al.
U.S. Appl. No. 61/734,256, filed Dec. 6, 2012, Chen et al.
U.S. Appl. No. 61/758,624, filed Jan. 30, 2013, Chen et al.
U.S. Appl. No. 61/761,046, filed Feb. 5, 2013, Knight et al.
U.S. Appl. No. 61/794,422, filed Mar. 15, 2013, Knight et al.
U.S. Appl. No. 61/803,599, filed Mar. 20, 2013, Kim et al.
U.S. Appl. No. 61/837,481, filed Jun. 20, 2013, Cho et al.
U.S. Appl. No. 61/838,178, filed Jun. 21, 2013, Joung et al.
U.S. Appl. No. 61/874,682, filed Sep. 6, 2013, Liu et al.
U.S. Appl. No. 61/874,746, filed Sep. 6, 2013, Liu et al.
U.S. Appl. No. 62/288,661, filed Jan. 29, 2016, Muir et al.
U.S. Appl. No. 62/357,332, filed Jun. 30, 2016, Liu et al.
International Search Report and Written Opinion for Application No. PCT/US2021/031439, dated Sep. 22, 2021.
Anonymous Third Party Observations for Application No. PCT/US2022/031439, dated Sep. 8, 2022.
International Preliminary Report on Patentability for Application No. PCT/US2021/031439, dated Nov. 17, 2022.
[No Author Listed] “Nucleic Acids Sizes and Molecular Weights.” Printed Mar. 19, 2021. 2 pages.
[No Author Listed] “Zinc Finger Nuclease” from Wikipedia. Retrieved from https://en.wikipedia.org/w/index.php?title=Zinc_finger_nuclease&oldid=1007053318. Page last edited Feb. 16, 2021. Printed on Mar. 19, 2021.
[No Author Listed] Beast2: Bayesian evolutionary analysis by sampling trees. http://www.beast2.org/ Last accessed Apr. 28, 2021.
[No Author Listed] HyPhy—Hypothesis testing using Phylogenies. Last modified Apr. 21, 2017. Accessed online via http://hyphy.org/w/index.php/Main_Page on Apr. 28, 2021.
[No Author Listed] NCBI Accession No. XP_015843220.1. C ->U editing enzyme APOBEC-1 [Peromyscus maniculatus bairdii], XP002793540. Mar. 21, 2016.
[No Author Listed] NCBI Accession No. XP_021505673.1. C ->U editing enzyme APOBEC-1 [Meriones unguiculatus], XP002793541. Jun. 27, 2017.
[No Author Listed] NCBI Reference Sequence: WP_001516895.1. Mar. 13, 2021. 2 pages.
[No Author Listed] NCBI Reference Sequence: WP_087959824.1. Oct. 9, 2019. 2 pages.
[No Author Listed] Nucleic Acid Data from New England Biolabs. Printed Sep. 28, 2021. 1 page.
[No Author Listed] Score result for SEQ 355 to W02017032580. Muir et al. 2016.
[No Author Listed] Theoretical Biochemistry Group. Institute for Theoretical Chemistry. The ViennaRNA Package. Universitat Wien. https://www.tbi.univie.ac.at/RNA/. Last accessed Apr. 28, 2021.
[No Author Listed] Transcription activator-like effector nuclease. Wikipedia. Last edited Sep. 27, 2021. Accessed via https://en.wikipedia.org/w/index.php?title=Transcription_activator-like_effector_nuclease&oldid=1046813325 on Sep. 28, 2021. 9 pages.
[No Author Listed], “Enediyne” from Wikipedia. Retrieved from https://en.wikipedia.org/w/index.php?title=Enediyne&oldid=1114086576. Page last edited Oct. 4, 2022. Printed on Oct. 7, 2022. 8 pages.
[No Author Listed], “FokI” from New England Biolabs Inc. Last accessed online via https://www.neb.com/products/r0109-foki#Product%20Information on Mar. 19, 2021. 1 page.
[No Author Listed], “Human genome.” Encyclopedia Britannica. Encyclopedia Brittanica, Inc. Published Feb. 15, 2019. Last accessed online via https://www.britannica.com/science/human-genome on Mar. 19, 2021. 2 pages.
[No Author Listed], “Lambda DNA” from Catalog & Technical Reference. New England Biolabs Inc. 2002/2003. pp. 133 and 270-273.
[No Author Listed], Adenine deaminase polypeptide SEQ: 49. EBI Acc. No. BJG44493. Jun. 10, 2021. 1 page.
[No Author Listed], EMBL Accession No. Q99ZW2. Nov. 2012. 2 pages.
[No Author Listed], Gag-Pol polyprotein. UniProtKB/Swiss-Prot No. P03355.5. Sep. 18, 2019. 18 pages.
[No Author Listed], Homo sapiens signal transducer and activator of transcription 3 (STAT3), transcript variant 1, mRNA. NCBI Ref Seq No. NM_139276.2. Retrived from https://www.ncbi.nlm.nih.gov/nuccore/nm_139276.2. Feb. 26, 2020. 8 pages.
[No Author Listed], Homo sapiens survival of motor neuron 2, centromeric (SMN2), RefSeqGene (LRG_677) on chromosome 5. NCBI Ref Seq No. NG_008728.1. Aug. 21, 2022. Accessible at https://www.ncbi.nlm.nih.gov/nuccore/NG_008728.1/. 14 pages.
[No Author Listed], Invitrogen Lipofectamine™ 2000 product sheets, 2002. 2 pages.
[No Author Listed], Invitrogen Lipofectamine™ 2000 product sheets, 2005. 3 pages.
[No Author Listed], Invitrogen Lipofectamine™ LTX product sheets, 2011. 4 pages.
[No Author Listed], Multispecies: tRNA adenosine(34) deaminase TadA [Enterobacteriaceae]. NCBI Ref Seq No. WP_001297409.1. Feb. 28, 2022. Retrieved from https://www.ncbi.nlm.nih.gov/protein/WP_001297409.1/. 2 pages.
[No Author Listed], Mus musculus (Mouse). UniProtKB Accession No. P51908 (ABEC1_MOUSE). Oct. 1, 1996. 10 pages.
[No Author Listed], signal transducer and activator of transcription 3 isoform 1 [Homo sapiens]. NCBI Ref Seq No. NP_644805.1. Retrieved from https://www.ncbi.nlm.nih.gov/protein/NP_644805.1/. Nov. 27, 2022. 5 pages.
[No Author Listed], Streptococcus pyogenes Cas9 protein. EBI Acc. No. BIR16744. Jan. 21, 2021. 1 page.
[No Author Listed], Streptococcus pyogenes Cas9 protein. EBI Acc. No. BIR16747. Jan. 21, 2021. 1 page.
[No Author Listed], Thermo Fisher Scientific—How Cationic Lipid Mediated Transfection Works, retrieved from the internet Aug. 27, 2015. 2 pages.
[No Author Listed], tumor-specific antigen. Retrieved from http://www.cancer.gov/publications/dictionaries/cancer-terms/def/tumor-specific-antigen. Retrieved on Oct. 7, 2022. 1 page.
Abremski et al., Bacteriophage P1 site-specific recombination. Purification and properties of the Cre recombinase protein. J Biol Chem. Feb. 10, 1984;259(3):1509-14.
Abudayyeh et al., A cytosine deaminase for programmable single-base RNA editing. Science. Jul. 26, 2019;365(6451):382-386. doi: 10.1126/science.aax7063. Epub Jul. 11, 2019.
Abudayyeh et al., C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector. Science. Aug. 5, 2016;353(6299):aaf5573. doi: 10.1126/science.aaf5573. Epub Jun. 2, 2016.
Abudayyeh et al., RNA targeting with CRISPR-Cas13. Nature. Oct. 12, 2017;550(7675):280-284. doi: 10.1038/nature24049. Epub Oct. 4, 2017.
Ada et al., Carbohydrate-protein conjugate vaccines. Clin Microbiol Infect. Feb. 2003;9(2):79-85. doi: 10.1046/j.1469-0691.2003.00530.x.
Adamala et al., Programmable RNA-binding protein composed of repeats of a single modular unit. Proc Natl Acad Sci U S A. May 10, 2016;113(19):E2579-88. doi: 10.1073/pnas.1519368113. Epub Apr. 26, 2016.
Adams et al., New biarsenical ligands and tetracysteine motifs for protein labeling in vitro and in vivo: synthesis and biological applications. J Am Chem Soc. May 29, 2002;124(21):6063-76. doi: 10.1021/ja017687n.
Addgene Plasmid # 44246. pdCas9-humanized, 2017, Stanley Qi.
Addgene Plasmid # 73021. PCMV-BE3, 2017, David Liu.
Addgene Plasmid # 79620. pcDNA3.1_pCMV-nCas-PmCDA1-ugi pH1-gRNA(HPRT), 2017, Akihiko Kondo.
Adli, The CRISPR tool kit for genome editing and beyond. Nat Commun. May 15, 2018;9(1):1911. doi: 10.1038/s41467-018-04252-2.
Aguilo et al., Coordination of m(6)A mRNA Methylation and Gene Transcription by ZFP217 Regulates Pluripotency and Reprogramming. Cell Stem Cell. Dec. 3, 2015;17(6):689-704. doi: 10.1016/j.stem.2015.09.005. Epub Oct. 29, 2015.
Ahmad et al., Antibody-mediated specific binding and cytotoxicity of liposome-entrapped doxorubicin to lung cancer cells in vitro. Cancer Res. Sep. 1, 1992;52(17):4817-20.
Ai et al., C-terminal Loop Mutations Determine Folding and Secretion Properties of PCSK9. iMedPub J: Biochem Mol Biol J. Nov. 5, 2016;2(3):17. doi: 10.21767/2471-8084.100026. 12 pages.
Aida et al., Prime editing primarily incudes undesired outcomes in mice. bioRxiv preprint and Supplemental Information. Aug. 6, 2020. Retrieved from www.biorxiv.org. doi: 10.1101/2020.08.06.239723. 40 pages.
Aihara et al., A conformational switch controls the DNA cleavage activity of lambda integrase. Mol Cell. Jul. 2003;12(1):187-98.
Aik et al., Structure of human RNA N6-methyladenine demethylase ALKBH5 provides insights into its mechanisms of nucleic acid recognition and demethylation. Nucleic Acids Res. Apr. 2014;42(7):4741-54. doi: 10.1093/nar/gku085. Epub Jan. 30, 2014.
Aird et al., Increasing Cas9-mediated homology-directed repair efficiency through covalent tethering of DNA repair template. Commun Biol. May 31, 2018;1:54. doi: 10.1038/s42003-018-0054-2.
Akcakaya et al., In vivo CRISPR editing with No. detectable genome-wide off-target mutations. Nature. Sep. 2018;561(7723):416-419. doi: 10.1038/s41586-018-0500-9. Epub Sep. 12, 2018. PMID: 30209390; PMCID: PMC6194229.
Akins et al., Mitochondrial plasmids of Neurospora: integration into mitochondrial DNA and evidence for reverse transcription in mitochondria. Cell. Nov. 21, 1986;47(4):505-16. doi: 10.1016/0092-8674(86)90615-x.
Akinsheye et al., Fetal hemoglobin in sickle cell anemia. Blood. Jul. 7, 2011;118(1):19-27. doi: 10.1182/blood-2011-03-325258. Epub Apr. 13, 2011.
Akopian et al., Chimeric recombinases with designed DNA sequence recognition. Proc Natl Acad Sci U S A. Jul. 22, 2003;100(15):8688-91. Epub Jul. 1, 2003.
Alarcón et al., HNRNPA2B1 Is a Mediator of m(6)A-Dependent Nuclear RNA Processing Events. Cell. Sep. 10, 2015;162(6):1299-308. doi: 10.1016/j.cell.2015.08.011. Epub Aug. 27, 2015.
Alarcón et al., N6-methyladenosine marks primary microRNAs for processing. Nature. Mar. 26, 2015;519(7544):482-5. doi: 10.1038/nature14281. Epub Mar. 18, 2015.
Alexander, HFE-associated hereditary hemochromatosis. Genet Med. May 2009;11(5):307-13. doi: 10.1097/GIM.0b013e31819d30f2.
Alexandrov et al., Signatures of mutational processes in human cancer. Nature. Aug. 22, 2013;500(7463):415-21. doi: 10.1038/nature12477. Epub Aug. 14, 2013.
Ali et al., Novel genetic abnormalities in Bernard-Soulier syndrome in India. Ann Hematol. Mar. 2014;93(3):381-4. doi: 10.1007/s00277-013-1895-x. Epub Sep. 1, 2013.
Altschul et al., Basic local alignment search tool. J Mol Biol. Oct. 5, 1990;215(3):403-10. doi: 10.1016/S0022-2836(05)80360-2.
Amato et al., Interpreting elevated fetal hemoglobin in pathology and health at the basic laboratory level: new and known γ-gene mutations associated with hereditary persistence of fetal hemoglobin. Int J Lab Hematol. Feb. 2014;36(1):13-9. doi: 10.1111/ijlh.12094. Epub Apr. 29, 2013.
Ames et al., A eubacterial riboswitch class that senses the coenzyme tetrahydrofolate. Chem Biol. Jul. 30, 2010;17(7):681-5. doi: 10.1016/j.chembiol.2010.05.020.
Amrann et al., Tightly regulated tac promoter vectors useful for the expression of unfused and fused proteins in Escherichia coli. Gene. Sep. 30, 1988;69(2):301-15.
Anders et al., Chapter One: In Vitro Enzymology of Cas9. in Methods in Enzymology, eds Doudna et al. 2014: 546:1-20.
Anders et al., Structural basis of PAM-dependent target DNA recognition by the Cas9 endonuclease. Nature. Sep. 25, 2014;513(7519):569-73. doi: 10.1038/nature13579. Epub Jul. 27, 2014.
Anderson, Human gene therapy. Science. May 8, 1992;256(5058):808-13. doi: 10.1126/science.1589762.
André et al., Axotomy-induced expression of calcium-activated chloride current in subpopulations of mouse dorsal root ganglion neurons. J Neurophysiol. Dec. 2003;90(6):3764-73. doi: 10.1152/jn.00449.2003. Epub Aug. 27, 2003.
Anzalone et al., Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol. Jul. 2020;38(7):824-844. doi: 10.1038/s41587-020-0561-9. Epub Jun. 22, 2020.
Anzalone et al., Reprogramming eukaryotic translation with ligand-responsive synthetic RNA switches. Nat Methods. May 2016;13(5):453-8. doi: 10.1038/nmeth.3807. Epub Mar. 21, 2016.
Anzalone et al., Search-and-replace genome editing without double-strand breaks or donor DNA. Nature. Dec. 2019;576(7785):149-157 and Suppl Info. doi: 10.1038/s41586-019-1711-4. Epub Oct. 21, 2019. 72 pages.
Anzalone et al., Search-and-replace genome editing without double-strand breaks or donor DNA. Nature. Dec. 2019;576(7785):149-157. doi: 10.1038/s41586-019-1711-4. Epub Oct. 21, 2019.
Aplan, Causes of oncogenic chromosomal translocation. Trends Genet. Jan. 2006;22(1):46-55. doi: 10.1016/j.tig.2005.10.002. Epub Oct. 28, 2005.
Arakawa et al., A method to convert mRNA into a gRNA library for CRISPR/Cas9 editing of any organism. Sci Adv. Aug. 24, 2016;2(8):e1600699. doi: 10.1126/sciadv.1600699.
Araki et al., Comparative analysis of right element mutant lox sites on recombination efficiency in embryonic stem cells. BMC Biotechnol. Mar. 31, 2010;10:29. doi: 10.1186/1472-6750-10-29.
Araki et al., Site-specific recombinase, R, encoded by yeast plasmid pSR1. J Mol Biol. May 5, 1992;225(1):25-37. doi: 10.1016/0022-2836(92)91023-i.
Araki et al., Targeted integration of DNA using mutant lox sites in embryonic stem cells. Nucleic Acids Res. Feb. 15, 1997;25(4):868-72. doi: 10.1093/nar/25.4.868.
Arambula et al., Surface display of a massively variable lipoprotein by a Legionella diversity-generating retroelement. Proc Natl Acad Sci U S A. May 14, 2013;110(20):8212-7. doi: 10.1073/pnas.1301366110. Epub Apr. 30, 2013.
Arazoe et al., Targeted Nucleotide Editing Technologies for Microbial Metabolic Engineering. Biotechnol J. Sep. 2018;13(9):e1700596. doi: 10.1002/biot.201700596. Epub Jun. 19, 2018.
Arbab et al., Cloning-free CRISPR. Stem Cell Reports. Nov. 10, 2015;5(5):908-917. doi: 10.1016/j.stemcr.2015.09.022. Epub Oct. 29, 2015.
Arbab et al., Determinants of Base Editing Outcomes from Target Library Analysis and Machine Learning. Cell. Jul. 23, 2020;182(2):463-480.e30. doi: 10.1016/j.cell.2020.05.037. Epub Jun. 12, 2020.
Arezi et al., Novel mutations in Moloney Murine Leukemia Virus reverse transcriptase increase thermostability through tighter binding to template-primer. Nucleic Acids Res. Feb. 2009;37(2):473-81. doi: 10.1093/nar/gkn952. Epub Dec. 4, 2008.
Arnold et al., Mutants of Tn3 resolvase which do not require accessory binding sites for recombination activity. EMBO J. Mar. 1, 1999;18(5):1407-14.
Asante et al., A naturally occurring variant of the human prion protein completely prevents prion disease. Nature. Jun. 25, 2015;522(7557):478-81. doi: 10.1038/nature14510. Epub Jun. 10, 2015.
Asokan et al., The AAV vector toolkit: poised at the clinical crossroads. Mol Ther. Apr. 2012;20(4):699-708. doi: 10.1038/mt.2011.287. Epub Jan. 24, 2012.
Atkins et al., Ribosomal frameshifting and transcriptional slippage: From genetic steganography and cryptography to adventitious use. Nucleic Acids Res. Sep. 6, 2016;44(15):7007-78. doi: 10.1093/nar/gkw530. Epub Jul. 19, 2016.
Auer et al., Highly efficient CRISPR/Cas9-mediated knock-in in zebrafish by homology-independent DNA repair. Genome Res. Jan. 2014;24(1):142-53. doi: 10.1101/gr.161638.113. Epub Oct. 31, 2013.
Auricchio et al., Exchange of surface proteins impacts on viral vector cellular specificity and transduction characteristics: the retina as a model. Hum Mol Genet. Dec. 15, 2001;10(26):3075-81. doi: 10.1093/hmg/10.26.3075.
Autieri et al., IRT-1, a novel interferon-gamma-responsive transcript encoding a growth-suppressing basic leucine zipper protein. J Biol Chem. Jun. 12, 1998;273(24):14731-7. doi: 10.1074/jbc.273.24.14731.
Avidan et al., The processivity and fidelity of DNA synthesis exhibited by the reverse transcriptase of bovine leukemia virus. Eur J Biochem. Feb. 2002;269(3):859-67. doi: 10.1046/j.0014-2956.2001.02719.x.
Baba et al., Construction of Escherichia coli K-12 in-frame, single-gene knockout mutants: the Keio collection. Mol Syst Biol. 2006;2:2006.0008. doi: 10.1038/msb4100050. Epub Feb. 21, 2006.
Babacic et al., CRISPR-cas gene-editing as plausible treatment of neuromuscular and nucleotide-repeat-expansion diseases: A systematic review. PLoS One. Feb. 22, 2019;14(2):e0212198. doi: 10.1371/journal.pone.0212198.
Bacman et al., Specific elimination of mutant mitochondrial genomes in patient-derived cells by mitoTALENs. Nat Med. Sep. 2013;19(9):1111-3. doi: 10.1038/nm.3261. Epub Aug. 4, 2013.
Badran et al., Continuous evolution of Bacillus thuringiensis toxins overcomes insect resistance. Nature. May 5, 2016;533(7601):58-63. doi: 10.1038/nature17938. Epub Apr. 27, 2016.
Badran et al., Development of potent in vivo mutagenesis plasmids with broad mutational spectra. Nat Commun. Oct. 7, 2015;6:8425. doi: 10.1038/ncomms9425.
Badran et al., In vivo continuous directed evolution. Curr Opin Chem Biol. Feb. 2015;24:1-10. doi: 10.1016/j.cbpa.2014.09.040. Epub Nov. 7, 2014.
Bae et al., Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics. May 15, 2014;30(10):1473-5. doi: 10.1093/bioinformatics/btu048. Epub Jan. 24, 2014.
Bae et al., Microhomology-based choice of Cas9 nuclease target sites. Nat Methods. Jul. 2014;11(7):705-6. doi: 10.1038/nmeth.3015.
Bagal et al., Recent progress in sodium channel modulators for pain. Bioorg Med Chem Lett. Aug. 15, 2014;24(16):3690-9. doi: 10.1016/j.bmcl.2014.06.038. Epub Jun. 21, 2014.
Bagyinszky et al., Characterization of mutations in PRNP (prion) gene and their possible roles in neurodegenerative diseases. Neuropsychiatr Dis Treat. Aug. 14, 2018;14:2067-2085. doi: 10.2147/NDT.S165445.
Balakrishnan et al., Flap endonuclease 1. Annu Rev Biochem. 2013;82:119-38. doi: 10.1146/annurev-biochem-072511-122603. Epub Feb. 28, 2013.
Baldari et al., A novel leader peptide which allows efficient secretion of a fragment of human interleukin 1 beta in Saccharomyces cerevisiae. EMBO J. Jan. 1987;6(1):229-34.
Banerjee et al., Cadmium inhibits mismatch repair by blocking the ATPase activity of the MSH2-MSH6 complex [published correction appears in Nucleic Acids Res. 2005;33(5):1738]. Nucleic Acids Res. 2005;33(4): 1410-1419. Published Mar. 3, 2005. doi: 10.1093/nar/gki291.
Banerji et al., A lymphocyte-specific cellular enhancer is located downstream of the joining region in immunoglobulin heavy chain genes. Cell. Jul. 1983;33(3):729-40. doi: 10.1016/0092-8674(83)90015-6.
Bannert et al., Retroelements and the human genome: new perspectives on an old relation. Proc Natl Acad Sci U S A. Oct. 5, 2004;101 Suppl 2(Suppl 2):14572-9. doi: 10.1073/pnas.0404838101. Epub Aug. 13, 2004.
Banno et al., Deaminase-mediated multiplex genome editing in Escherichia coli. Nat Microbiol. Apr. 2018;3(4):423-429. doi: 10.1038/s41564-017-0102-6. Epub Feb. 5, 2018.
Baranauskas et al., Generation and characterization of new highly thermostable and processive M-MuLV reverse transcriptase variants. Protein Eng Des Sel. Oct. 2012;25(10):657-68. doi: 10.1093/protein/gzs034. Epub Jun. 12, 2012.
Barmania et al., C-C chemokine receptor type five (CCR5): An emerging target for the control of HIV infection. Appl Transl Genom. May 26, 2013;2:3-16. doi: 10.1016/j.atg.2013.05.004.
Barnes et al., Repair and genetic consequences of endogenous DNA base damage in mammalian cells. Annu Rev Genet. 2004;38:445-76.
Barnes et al., The fidelity of Taq polymerase catalyzing PCR is improved by an N-terminal deletion. Gene. Mar. 1, 1992;112(1):29-35. doi: 10.1016/0378-1119(92)90299-5.
Barrangou et al., CRISPR provides acquired resistance against viruses in prokaryotes. Science. Mar. 23, 2007;315(5819):1709-12.
Barrangou, RNA-mediated programmable DNA cleavage. Nat Biotechnol. Sep. 2012;30(9):836-8. doi: 10.1038/nbt.2357.
Bartlett et al., Efficient expression of protein coding genes from the murine U1 small nuclear RNA promoters. Proc Natl Acad Sci U S A. Aug. 20, 1996;93(17):8852-7. doi: 10.1073/pnas.93.17.8852.
Bartosovic et al., N6-methyladenosine demethylase FTO targets pre-mRNAs and regulates alternative splicing and 3′-end processing. Nucleic Acids Res. Nov. 2, 2017;45(19):11356-11370. doi: 10.1093/nar/gkx778.
Basha et al., Influence of cationic lipid composition on gene silencing properties of lipid nanoparticle formulations of siRNA in antigen-presenting cells. Mol Ther. Dec. 2011;19(12):2186-200. doi: 10.1038/mt.2011.190. Epub Oct. 4, 2011.
Bass, B.L., RNA editing by adenosine deaminases that act on RNA. Annu Rev Biochem. 2002;71:817-46. doi: 10.1146/annurev.biochem.71.110601.135501. Epub Nov. 9, 2001.
Basturea et al., Substrate specificity and properties of the Escherichia coli 16S rRNA methyltransferase, RsmE. RNA. Nov. 2007;13(11):1969-76. doi: 10.1261/rna.700507. Epub Sep. 13, 2007.
Batey et al., Structure of a natural guanine-responsive riboswitch complexed with the metabolite hypoxanthine. Nature. Nov. 18, 2004;432(7015):411-5.
Beale et al., Comparison of the differential context-dependence of DNA deamination by APOBEC enzymes: correlation with mutation spectra in vivo. J Mol Biol. Mar. 26, 2004;337(3):585-96.
Beaudry et al., Directed evolution of an RNA enzyme. Science. Jul. 31, 1992;257(5070):635-41. doi: 10.1126/science.1496376.
Bebenek et al., Error-prone polymerization by HIV-1 reverse transcriptase. Contribution of template-primer misalignment, miscoding, and termination probability to mutational hot spots. J Biol Chem. May 15, 1993;268(14):10324-34.
Bedell et al., In vivo genome editing using a high-efficiency TALEN system. Nature. Nov. 1, 2012;491(7422):114-8. Doi: 10.1038/nature11537. Epub Sep. 23, 2012.
Begley, Scientists unveil the ‘most clever CRISPR gadget’ so far. STAT, Apr. 20, 2016. https://www.statnews.com/2016/04/20/clever-crispr-advance-unveiled/.
Behr, Gene transfer with synthetic cationic amphiphiles: prospects for gene therapy. Bioconjug Chem. Sep.-Oct. 1994;5(5):382-9. doi: 10.1021/bc00029a002.
Bell et al., Ribozyme-catalyzed excision of targeted sequences from within RNAs. Biochemistry. Dec. 24, 2002;41(51):15327-33. doi: 10.1021/bi0267386.
Belshaw et al., Controlling programmed cell death with a cyclophilin-cyclosporin-based chemical inducer of dimerization. Chem Biol. Sep. 1996;3(9):731-8. doi: 10.1016/s1074-5521(96)90249-5.
Belshaw et al., Controlling protein association and subcellular localization with a synthetic ligand that induces heterodimerization of proteins. Proc Natl Acad Sci U S A. May 14, 1996;93(10):4604-7. doi: 10.1073/pnas.93.10.4604.
Benarroch, Hcn channels: function and clinical implications. Neurology. Jan. 15, 2013;80(3):304-10. doi: 10.1212/WNL.0b013e31827dec42.
Bennett et al., Painful and painless channelopathies. Lancet Neurol. Jun. 2014;13(6):587-99. doi: 10.1016/S1474-4422(14)70024-9. Epub May 6, 2014.
Bentin, T., A ribozyme transcribed by a ribozyme. Artif DNA PNA XNA. Apr. 2011;2(2):40-42. doi: 10.4161/adna.2.2.16852.
Berger et al., Reverse transcriptase and its associated ribonuclease H: interplay of two enzyme activities controls the yield of single-stranded complementary deoxyribonucleic acid. Biochemistry. May 10, 1983;22(10):2365-72. doi: 10.1021/bi00279a010.
Berges et al., Transduction of brain by herpes simplex virus vectors. Mol Ther. Jan. 2007;15(1):20-9. doi: 10.1038/sj.mt.6300018.
Berkhout et al., Identification of an active reverse transcriptase enzyme encoded by a human endogenous HERV-K retrovirus. J Virol. Mar. 1999;73(3):2365-75. doi: 10.1128/JVI.73.3.2365-2375.1999.
Bernhart et al., Local RNA base pairing probabilities in large sequences. Bioinformatics. Mar. 1, 2006;22(5):614-5. doi: 10.1093/bioinformatics/btk014. Epub Dec. 20, 2005.
Bernstein et al., Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature. Jan. 18, 2001;409(6818):363-6. doi: 10.1038/35053110.
Bershtein et al., Advances in laboratory evolution of enzymes. Curr Opin; Chem Biol. Apr. 2008; 12(2):151-8. doi: 10.1016/j.cbpa.2008.01.027. Epub Mar. 7, 2008. Review.
Bertolotti et al., Toward genosafe endonuclease-boosted gene targeting using breakthrough CRISP/Cas9 for next generation stem cell gene therapy culminating in efficient ex VIVO in VIVO gene repair/genomic editing. Molecular Therapy. May 2015;23(Suppl1):S139. Abstract 350. 18th Ann Meeting of the American Society of Gene and Cell Therapy. ASGCT 2015. New Orleans, LA. May 13, 2015-May 16, 2015.
Bertrand et al., Localization of ASH1 mRNA particles in living yeast. Mol Cell. Oct. 1998;2(4):437-45. doi: 10.1016/s1097-2765(00)80143-4.
Bessen et al., High-resolution specificity profiling and off-target prediction for site-specific DNA recombinases. Nat Commun. Apr. 26, 2019;10(1):1937. doi: 10.1038/s41467-019-09987-0.
Beumer et al., Efficient gene targeting in Drosophila with zinc-finger nucleases. Genetics. Apr. 2006; 172(4):2391-403. Epub Feb. 1, 2006.
Bhagwat, DNA-cytosine deaminases: from antibody maturation to antiviral defense. DNA Repair (Amst). Jan. 5, 2004;3(1):85-9.
Bi et al., Pseudo attP sites in favor of transgene integration and expression in cultured porcine cells identified by Streptomyces phage phiC31 integrase. BMC Mol Biol. Sep. 8, 2013;14:20. doi: 10.1186/1471-2199-14-20.
Bibb et al., Integration and excision by the large serine recombinase phiRv1 integrase. Mol Microbiol. Mar. 2005;55(6):1896-910. doi: 10.1111/j.1365-2958.2005.04517.x.
Bibikova et al., Targeted chromosomal cleavage and mutagenesis in Drosophila using zinc-finger nucleases. Genetics. Jul. 2002;161(3):1169-75. doi: 10.1093/genetics/161.3.1169.
Biehs et al., DNA Double-Strand Break Resection Occurs during Non-homologous End Joining in G1 but Is Distinct from Resection during Homologous Recombination. Mol Cell. Feb. 16, 2017;65(4):671-684.e5. doi: 10.1016/j.molcel.2016.12.016. Epub Jan. 26, 2017.
Billon et al., CRISPR-Mediated Base Editing Enables Efficient Disruption of Eukaryotic Genes through Induction of STOP Codons. Mol Cell. Sep. 21, 2017;67(6):1068-1079.e4. doi: 10.1016/j.molcel.2017.08.008. Epub Sep. 7, 2017.
Birling et al., Site-specific recombinases for manipulation of the mouse genome. Methods Mol Biol. 2009;561:245-63. doi: 10.1007/978-1-60327-019-9_16.
Biswas et al., A structural basis for allosteric control of DNA recombination by lambda integrase. Nature. Jun. 23, 2005;435(7045):1059-66. doi: 10.1038/nature03657.
Bitinaite et al., FokI dimerization is required for DNA cleavage. Proc Natl Acad Sci U S A. Sep. 1, 1998;95(18):10570-5.
Blaese et al., Vectors in cancer therapy: how will they deliver? Cancer Gene Ther. Dec. 1995;2(4):291-7.
Blain et al., Nuclease activities of Moloney murine leukemia virus reverse transcriptase. Mutants with altered substrate specificities. J Biol Chem. Nov. 5, 1993;268(31):23585-92.
Blaisonneau et al., A circular plasmid from the yeast Torulaspora delbrueckii. Plasmid. 1997;38(3):202-9. doi: 10.1006/plas.1997.1315.
Blau et al., A proliferation switch for genetically modified cells. PNAS Apr. 1, 1997 94 (7) 3076-3081; https://doi.org/10.1073/pnas.94.7.3076.
Blauw et al., SMN1 gene duplications are associated with sporadic ALS. Neurology. Mar. 13, 2012;78(11):776-80. doi: 10.1212/WNL.0b013e318249f697. Epub Feb. 8, 2012.
Bloom et al., Evolving strategies for enzyme engineering. Curr Opin Struct Biol. Aug. 2005;15(4):447-52.
Boch, TALEs of genome targeting. Nat Biotechnol. Feb. 2011;29(2):135-6. Doi: 10.1038/nbt.1767.
Bodi et al., Yeast m6A Methylated mRNAs Are Enriched on Translating Ribosomes during Meiosis, and under Rapamycin Treatment. PLoS One. Jul. 17, 2015;10(7):e0132090. doi: 10.1371/journal.pone.0132090.
Boeckle et al., Melittin analogs with high lytic activity at endosomal pH enhance transfection with purified targeted PEI polyplexes. J Control Release. May 15, 2006;112(2):240-8. Epub Mar. 20, 2006.
Boersma et al., Selection strategies for improved biocatalysts. FEBS J. May 2007;274(9):2181-95.
Bogdanove et al., Engineering altered protein-DNA recognition specificity. Nucleic Acids Res. Jun. 1, 2018;46(10):4845-4871. doi: 10.1093/nar/gky289.
Bogdanove et al., TAL effectors: customizable proteins for DNA targeting. Science. Sep. 30, 2011;333(6051):1843-6. doi: 10.1126/science.1204094.
Bohlke et al., Sense codon emancipation for proteome-wide incorporation of noncanonical amino acids: rare isoleucine codon AUA as a target for genetic code expansion. FEMS Microbiol Lett. Feb. 2014;351(2):133-44. doi: 10.1111/1574-6968.12371. Epub Jan. 27, 2014.
Bolotin et al., Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology. Aug. 2005;151(Pt 8):2551-61.
Bolusani et al., Evolution of variants of yeast site-specific recombinase Flp that utilize native genomic sequences as recombination target sites. Nucleic Acids Res. 2006;34(18):5259-69. Epub Sep. 26, 2006.
Bondeson et al., Inversion of the IDS gene resulting from recombination with IDS-related sequences is a common cause of the Hunter syndrome. Hum Mol Genet. Apr. 1995;4(4):615-21. doi: 10.1093/hmg/4.4.615.
Borchardt et al., Controlling mRNA stability and translation with the CRISPR endoribonuclease Csy4. RNA. Nov. 2015;21(11):1921-30. doi: 10.1261/rna.051227.115. Epub Sep. 9, 2015.
Borman, Improved route to single-base genome editing. Chemical & Engineering News, Apr. 25, 2016;94(17)p5. http://cen.acs.org/articles/94/i17/Improved-route-single-base-genome.html.
Bothmer et al., Characterization of the interplay between DNA repair and CRISPR/Cas9-induced DNA lesions at an endogenous locus. Nat Commun. Jan. 9, 2017;8:13905. doi: 10.1038/ncomms13905.
Bourinet et al., Silencing of the Cav3.2 T-type calcium channel gene in sensory neurons demonstrates its major role in nociception. EMBO J. Jan. 26, 2005;24(2):315-24. doi: 10.1038/sj.emboj.7600515. Epub Dec. 16, 2004.
Boutabout et al., DNA synthesis fidelity by the reverse transcriptase of the yeast retrotransposon Ty1. Nucleic Acids Res. Jun. 1, 2001;29(11):2217-22. doi: 10.1093/nar/29.11.2217.
Box et al., A multi-domain protein system based on the HC fragment of tetanus toxin for targeting DNA to neuronal cells. J Drug Target. Jul. 2003;11(6):333-43. doi: 10.1080/1061186310001634667.
Branden and Tooze, Introduction to Protein Structure. 1999; 2nd edition. Garland Science Publisher: 3-12.
Braun et al., Immunogenic duplex nucleic acids are nuclease resistant. J Immunol. Sep. 15, 1988;141(6):2084-9.
Brierley et al., Viral RNA pseudoknots: versatile motifs in gene expression and replication. Nat Rev Microbiol. Aug. 2007;5(8):598-610. doi: 10.1038/nrmicro1704.
Briner et al., Guide RNA functional modules direct Cas9 activity and orthogonality. Mol Cell. Oct. 23, 2014;56(2):333-339. doi: 10.1016/j.molcel.2014.09.019.
Britt et al., Re-engineering plant gene targeting. Trends Plant Sci. Feb. 2003;8(2):90-5.
Brouns et al., Small CRISPR RNAs guide antiviral defense in prokaryotes. Science. Aug. 15, 2008;321(5891):960-4. doi: 10.1126/science. 1159689.
Brown et al., A mammalian protein targeted by G1-arresting rapamycin-receptor complex. Nature. Jun. 30, 1994;369(6483):756-8. doi: 10.1038/369756a0.
Brown et al., Characterization of the genetic elements required for site-specific integration of plasmid pSE211 in Saccharopolyspora erythraea. J Bacteriol. Apr. 1990; 172(4):1877-88. doi: 10.1128/jb.172.4.1877-1888.1990.
Brown et al., Serine recombinases as tools for genome engineering. Methods. Apr. 2011;53(4):372-9. doi: 10.1016/j.ymeth.2010.12.031. Epub Dec. 30, 2010.
Brown et al., Structural insights into the stabilization of MALATI noncoding RNA by a bipartite triple helix. Nat Struct Mol Biol. Jul. 2014;21(7):633-40. doi: 10.1038/nsmb.2844. Epub Jun. 22, 2014.
Brusse et al., Spinocerebellar ataxia associated with a mutation in the fibroblast growth factor 14 gene (SCA27): A new phenotype. Mov Disord. Mar. 2006;21(3):396-401.
Brutlag et al., Improved sensitivity of biological sequence database searches. Comput Appl Biosci. Jul. 1990;6(3):237-45. doi: 10.1093/bioinformatics/6.3.237.
Brzezicha et al., Identification of human tRNA:m5C methyltransferase catalysing intron- dependent m5C formation in the first position of the anticodon of the pre-tRNA Leu (CAA). Nucleic Acids Res. 2006;34(20):6034-43. doi: 10.1093/nar/gk1765. Epub Oct. 27, 2006.
Buchholz et al., Alteration of Cre recombinase site specificity by substrate-linked protein evolution. Nat Biotechnol. Nov. 2001;19(11):1047-52.
Buchschacher et al., Human immunodeficiency virus vectors for inducible expression of foreign genes. J Virol. May 1992;66(5):2731-9. doi: 10.1128/JVI.66.5.2731-2739.1992.
Buchwald et al., Long-term, continuous intravenous heparin administration by an implantable infusion pump in ambulatory patients with recurrent venous thrombosis. Surgery. Oct. 1980;88(4):507-16.
Buckley et al., Targeting the von Hippel-Lindau E3 ubiquitin ligase using small molecules to disrupt the VHL/HIF-1 α interaction. J Am Chem Soc. Mar. 14, 2012;134(10):4465-8. doi: 10.1021/ja209924v. Epub Feb. 27, 2012.
Budisa et al., Residue-specific bioincorporation of non-natural, biologically active amino acids into proteins as possible drug carriers: structure and stability of the per-thiaproline mutant of annexin V. Proc Natl Acad Sci U S A. Jan. 20, 1998;95(2):455-9.
Budker et al., Protein/amphipathic polyamine complexes enable highly efficient transfection with minimal toxicity. Biotechniques. Jul. 1997;23(1):139, 142-7. doi: 10.2144/97231rr02.
Budworth et al., A brief history of triplet repeat diseases. Methods Mol Biol. 2013; 1010:3-17. doi: 10.1007/978-1-62703-411-1_1.
Bulow et al., Multienzyme systems obtained by gene fusion. Trends Biotechnol. Jul. 1991;9(7):226-31.
Burke et al., Activating mutations of Tn3 resolvase marking interfaces important in recombination catalysis and its regulation. Mol Microbiol. Feb. 2004;51(4):937-48.
Burke et al., RNA Aptamers to the Adenosine Moiety of S-adenosyl Methionine: Structural Inferences From Variations on a Theme and the Reproducibility of SELEX. Nucleic Acids Res. May 15, 1997;25(10):2020-4. doi: 10.1093/nar/25.10.2020.
Burstein et al., New CRISPR-Cas systems from uncultivated microbes. Nature Feb. 2017;542(7640):237-240.
Burton et al., Gene delivery using herpes simplex virus vectors. DNA Cell Biol. Dec. 2002;21(12):915-36. doi: 10.1089/104454902762053864.
Buskirk et al., Directed evolution of ligand dependence: small-molecule-activated protein splicing. Proc Natl Acad Sci U S A. Jul. 20, 2004;101(29): 10505-10. Epub Jul. 9, 2004.
Buskirk et al., In vivo evolution of an RNA-based transcriptional activator. Chem Biol. Jun. 2003;10(6):533-40. doi: 10.1016/s1074-5521(03)00109-1.
Butt et al., Efficient CRISPR/Cas9-Mediated Genome Editing Using a Chimeric Single-Guide RNA Molecule. Front Plant Sci. Aug. 24, 2017;8:1441(1-8). doi: 10.3389/fpls.2017.01441.
Byrne et al., Multiplex gene regulation: a two-tiered approach to transgene regulation in transgenic mice. Proc Natl Acad Sci U S A. Jul. 1989;86(14):5473-7. doi: 10.1073/pnas.86.14.5473.
Böck et al., Selenocysteine: the 21st amino acid. Mol Microbiol. Mar. 1991;5(3):515-20.
Cade et al., Highly efficient generation of heritable zebrafish gene mutations using homo- and heterodimeric TALENs. Nucleic Acids Res. Sep. 2012;40(16):8001-10. Doi: 10.1093/nar/gks518. Epub Jun. 7, 2012.
Cadwell et al., Randomization of genes by PCR mutagenesis. PCR Methods Appl. Aug. 1992;2(1):28-33. doi: 10.1101/gr.2.1.28.
Cai et al., Reconstruction of ancestral protein sequences and its applications. BMC Evol Biol. Sep. 17, 2004;4:33. doi: 10.1186/1471-2148-4-33.
Calame et al., Transcriptional controlling elements in the immunoglobulin and T cell receptor loci. Adv Immunol. 1988;43:235-75. doi: 10.1016/s0065-2776(08)60367-3.
Caldecott et al., Single-strand break repair and genetic disease. Nat Rev Genet. Aug. 2008;9(8):619-31. doi: 10.1038/nrg2380.
Camarero et al., Biosynthesis of a Head-to-Tail Cyclized Protein with Improved Biological Activity. J. Am. Chem. Soc. May 29, 1999; 121(23):5597-5598. https://doi.org/10.1021/ja990929n.
Cameron, Recent advances in transgenic technology. Mol Biotechnol. Jun. 1997;7(3):253-65.
Camper et al., Postnatal repression of the alpha-fetoprotein gene is enhancer independent. Genes Dev. Apr. 1989;3(4):537-46. doi: 10.1101/gad.3.4.537.
Camps et al., Targeted gene evolution in Escherichia coli using a highly error-prone DNA polymerase I. Proc Natl Acad Sci U S A. Aug. 19, 2003;100(17):9727-32. Epub Aug. 8, 2003.
Canchaya et al., Genome analysis of an inducible prophage and prophage remnants integrated in the Streptococcus pyogenes strain SF370. Virology. Oct. 25, 2002;302(2):245-58. doi: 10.1006/viro.2002.1570.
Canny et al., Inhibition of 53BP1 Favors Homology-Dependent DNA Repair and Increases CRISPR-Cas9 Genome-Editing Efficiency. Nat Biotechnol. Jan. 2018;36(1):95-102. doi: 10.1038/nbt.4021. Epub Nov. 27, 2017.
Canver et al., Customizing the genome as therapy for the β-hemoglobinopathies. Blood. May 26, 2016;127(21):2536-45. doi: 10.1182/blood-2016-01-678128. Epub Apr. 6, 2016.
Cao et al., Rapamycin reverses cellular phenotypes and enhances mutant protein clearance in Hutchinson-Gilford progeria syndrome cells. Sci Transl Med. Jun. 29, 2011;3(89):89ra58. doi: 10.1126/scitranslmed.3002346.
Cargill et al., Characterization of single-nucleotide polymorphisms in coding regions of human genes. Nat Genet. Jul. 1999;22(3):231-8.
Carlier et al., Burkholderia cenocepacia H111 Rhy-family protein. Apr. 16, 2015. Retrieved from the Internet via https://www.ebi.ac.uk/ena/browser/api/embl/CDN65395.1?lineLimit=1000. Last retrieved Apr. 26, 2021.
Carlier et al., Genome Sequence of Burkholderia cenocepacia H111, a Cystic Fibrosis Airway Isolate. Genome Announc. Apr. 10, 2014;2(2):e00298-14. doi: 10.1128/genomeA.00298-14.
Carlson et al., Negative selection and stringency modulation in phage-assisted continuous evolution. Nat Chem Biol. Mar. 2014; 10(3):216-22. doi: 10.1038/nchembio.1453. Epub Feb. 2, 2014. With Supplementary Results.
Caron et al., Intracellular delivery of a Tat-eGFP fusion protein into muscle cells. Mol Ther. Mar. 2001;3(3):310-8.
Carr et al., Genome engineering. Nat Biotechnol. Dec. 2009;27(12):1151-62. doi: 10.1038/nbt.1590.
Carroll et al., Gene targeting in Drosophila and Caenorhabditis elegans with zinc-finger nucleases. Methods Mol Biol. 2008;435:63-77. doi: 10.1007/978-1-59745-232-8_5.
Carroll et al., Progress and prospects: zinc-finger nucleases as gene therapy agents. Gene Ther. Nov. 2008;15(22):1463-8. doi: 10.1038/gt.2008.145. Epub Sep. 11, 2008.
Carroll, a Crispr approach to gene targeting. Mol Ther. Sep. 2012;20(9):1658-60. doi: 10.1038/mt.2012.171.
Carroll, Genome engineering with zinc-finger nucleases. Genetics. Aug. 2011;188(4):773-82. doi: 10.1534/genetics.111.131433. Review.
Cartegni et al., Determinants of exon 7 splicing in the spinal muscular atrophy genes, SMN1 and SMN2. Am J Hum Genet. Jan. 2006;78(1):63-77. doi: 10.1086/498853. Epub Nov. 16, 2005.
Carvalho et al., Evolution in health and medicine Sackler colloquium: Genomic disorders: a window into human gene and genome evolution. Proc Natl Acad Sci U S A. Jan. 26, 2010;107 Suppl 1(Suppl 1):1765-71. doi: 10.1073/pnas.0906222107. Epub Jan. 13, 2010.
Caspi et al., Distribution of split DnaE inteins in cyanobacteria. Mol Microbiol. Dec. 2003;50(5):1569-77. doi: 10.1046/j.1365-2958.2003.03825.x.
Cattaneo et al., SELIL affects human pancreatic cancer cell cycle and invasiveness through modulation of PTEN and genes related to cell-matrix interactions. Neoplasia. 2005;7(11):1030-1038.
Ceccaldi et al., Repair Pathway Choices and Consequences at the Double-Strand Break. Trends Cell Biol. Jan. 2016;26(1):52-64. doi: 10.1016/j.tcb.2015.07.009. Epub Oct. 1, 2015.
Cermak et al., Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. Jul. 2011;39(12):e82. Doi: 10.1093/nar/gkr218. Epub Apr. 14, 2011.
Chadalavada et al., Wild-type is the optimal sequence of the HDV ribozyme under cotranscriptional conditions. RNA. Dec. 2007;13(12):2189-201. doi: 10.1261/rna.778107. Epub Oct. 23, 2007.
Chadwick et al., In Vivo Base Editing of PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) as a Therapeutic Alternative to Genome Editing. Arterioscler Thromb Vasc Biol. Sep. 2017;37(9):1741-1747. doi: 10.1161/ATVBAHA.117.309881. Epub Jul. 27, 2017.
Chaikind et al., A programmable Cas9-serine recombinase fusion protein that operates on DNA sequences in mammalian cells. Nucleic Acids Res. Nov. 16, 2016;44(20):9758-9770. Epub Aug. 11, 2016.
Chalberg et al., Integration specificity of phage phiC31 integrase in the human genome. J Mol Biol. Mar. 17, 2006;357(1):28-48. doi: 10.1016/j.jmb.2005.11.098. Epub Dec. 22, 2005.
Chalberg et al., phiC31 integrase confers genomic integration and long-term transgene expression in rat retina. Invest Ophthalmol Vis Sci. Jun. 2005;46(6):2140-6. doi: 10.1167/iovs.04-1252.
Chan et al., Molecular recording of mammalian embryogenesis. Nature. Jun. 2019;570(7759):77-82. doi: 10.1038/s41586-019-1184-5. Epub May 13, 2019.
Chan et al., Novel selection methods for DNA-encoded chemical libraries. Curr Opin Chem Biol. 2015;26:55-61. doi:10.1016/j.cbpa.2015.02.010.
Chan et al., The choice of nucleotide inserted opposite abasic sites formed within chromosomal DNA reveals the polymerase activities participating in translesion DNA synthesis. DNA Repair (Amst). Nov. 2013;12(11):878-89. doi: 10.1016/j.dnarep.2013.07.008. Epub Aug. 26, 2013.
Chang et al., Degradation of survival motor neuron (SMN) protein is mediated via the ubiquitin/proteasome pathway. Neurochem Int. Dec. 2004;45(7):1107-12. doi: 10.1016/j.neuint.2004.04.005.
Chapman et al., Playing the end game: DNA double-strand break repair pathway choice. Mol Cell. Aug. 24, 2012;47(4):497-510. doi: 10.1016/j.molcel.2012.07.029.
Chari et al., Unraveling CRISPR-Cas9 genome engineering parameters via a library-on-library approach. Nat Methods. Sep. 2015;12(9):823-6. doi: 10.1038/nmeth.3473. Epub Jul. 13, 2015.
Charpentier et al., Biotechnology: Rewriting a genome. Nature. Mar. 7, 2013;495(7439):50-1. doi: 10.1038/495050a.
Chatterjee et al., A Cas9 with PAM recognition for adenine dinucleotides. Nat Commun. May 18, 2020;11(1):2474. doi: 10.1038/s41467-020-16117-8.
Chatterjee et al., Robust Genome Editing of Single-Base PAM Targets; with Engineered ScCas9 Variants. bioRxiv. doi: 10.1101/620351. Posted Apr. 26, 2019.
Chaturvedi et al., Stabilization of triple-stranded oligonucleotide complexes: use of probes containing alternating phosphodiester and stereo-uniform cationic phosphoramidate linkages. Nucleic Acids Res. Jun. 15, 1996;24(12):2318-23.
Chavez et al., Highly efficient Cas9-mediated transcriptional programming. Nat Methods. Apr. 2015;12(4):326-8. doi: 10.1038/nmeth.3312. Epub Mar. 2, 2015.
Chavez et al., Precise Cas9 targeting enables genomic mutation prevention. bioRxiv. Jun. 14, 2016; http://dx/doi.oreg/10.1101/058974. 6 pages. bioRxiv preprint first posted online Jun. 14, 2016.
Chavez et al., Therapeutic applications of the ΦC31 integrase system. Curr Gene Ther. Oct. 2011; 11(5):375-81. Review.
Chawla et al., An atlas of RNA base pairs involving modified nucleobases with optimal geometries and accurate energies. Nucleic Acids Res. Aug. 18, 2015;43(14):6714-29. doi: 10.1093/nar/gkv606. Epub Jun. 27, 2015.
Chelico et al., Biochemical basis of immunological and retroviral responses to DNA-targeted cytosine deamination by activation-induced cytidine deaminase and APOBEC3G. J Biol Chem. Oct. 9, 2009;284(41):27761-5. doi: 10.1074/jbc.R109.052449. Epub Aug. 13, 2009.
Chelico et al., Stochastic properties of processive cytidine DNA deaminases AID and APOBEC3G. Philos Trans R Soc Lond B Biol Sci. Mar. 12, 2009;364(1517):583-93. doi: 10.1098/rstb.2008.0195.
Chen et al., A general strategy for the evolution of bond-forming enzymes using yeast display. Proc Natl Acad Sci U S A. Jul. 12, 2011;108(28):11399-404. doi: 10.1073/pnas.1101046108. Epub Jun. 22, 2011.
Chen et al., Alterations in PMS2, MSH2 and MLH1 expression in human prostate cancer. Int J Oncol. May 2003;22(5):1033-43.
Chen et al., Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell. Oct. 28, 2021;184(22):5635-5652.e29. doi: 10.1016/j.cell.2021.09.018. Epub Oct. 14, 2021.
Chen et al., Enhanced proofreading governs CRISPR-Cas9 targeting accuracy. Nature. Oct. 19, 2017;550(7676):407-410. doi: 10.1038/nature24268. Epub Sep. 20, 2017.
Chen et al., Fusion protein linkers: property, design and functionality. Adv Drug Deliv Rev. Oct. 2013;65(10):1357-69. doi: 10.1016/j.addr.2012.09.039. Epub Sep. 29, 2012.
Chen et al., Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell. Mar. 12, 2015;160(6):1246-60. doi: 10.1016/j.cell.2015.02.038. Epub Mar. 5, 2015.
Chen et al., Highly Efficient Mouse Genome Editing by CRISPR Ribonucleoprotein Electroporation of Zygotes. J Biol Chem. Jul. 8, 2016;291(28):14457-67. doi: 10.1074/jbc.M116.733154. Epub May 5, 2016.
Chen et al., m(6)A RNA methylation is regulated by microRNAs and promotes reprogramming to pluripotency. Cell Stem Cell. Mar. 5, 2015;16(3):289-301. doi: 10.1016/j.stem.2015.01.016. Epub Feb. 12, 2015.
Chen et al., Structure of the DNA deaminase domain of the HIV-1 restriction factor APOBEC3G. Nature. Mar. 6, 2008;452(7183):116-9. doi: 10.1038/nature06638. Epub Feb. 20, 2008.
Chen et al., Targeting genomic rearrangements in tumor cells through Cas9-mediated insertion of a suicide gene. Nat Biotechnol. Jun. 2017;35(6):543-550. doi: 10.1038/nbt.3843. Epub May 1, 2017.
Cheng et al., Multiplexed activation of endogenous genes by CRISPR-on, an RNA-guided transcriptional activator system. Cell Res. Oct. 2013;23(10):1163-71. doi: 10.1038/cr.2013.122. Epub Aug. 27, 2013.
Chesnoy et al., Structure and function of lipid-DNA complexes for gene delivery. Annu Rev Biophys Biomol Struct. 2000;29:27-47.
Chester et al., The apolipoprotein B mRNA editing complex performs a multifunctional cycle and suppresses nonsense-mediated decay. EMBO J. Aug. 1, 2003;22(15):3971-82. doi: 10.1093/emboj/cdg369.
Chew et al., A multifunctional AAV-CRISPR-Cas9 and its host response. Nat Methods. Oct. 2016; 13(10):868-74. doi: 10.1038/nmeth.3993. Epub Sep. 5, 2016.
Chew et al., A multifunctional AAV-CRISPR-Cas9 and its host response. Nat Methods. Oct. 2016; 13(10):868-74. doi: 10.1038/nmeth.3993. Epub Sep. 5, 2016. Supplementary Information.
Chichili et al., Linkers in the structural biology of protein-protein interactions. Protein Science. 2013;22:153-67.
Chin, Expanding and reprogramming the genetic code of cells and animals. Annu Rev Biochem. 2014;83:379-408. doi: 10.1146/annurev-biochem-060713-035737. Epub Feb. 10, 2014.
Chipev et al., A leucine—proline mutation in the H1 subdomain of keratin 1 causes epidermolytic hyperkeratosis. Cell. Sep. 4, 1992;70(5):821-8.
Cho et al., A degron created by SMN2 exon 7 skipping is a principal contributor to spinal muscular atrophy severity. Genes Dev. Mar. 1, 2010;24(5):438-42. doi: 10.1101/gad.1884910.
Cho et al., Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res. Jan. 2014;24(1):132-41. doi: 10.1101/gr.162339.113. Epub Nov. 19, 2013.
Cho et al., Site-specific recombination of bacteriophage P22 does not require integration host factor. J Bacteriol. Jul. 1999; 181(14):4245-9. doi: 10.1128/JB.181.14.4245-4249.1999.
Cho et al., Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat Biotechnol. Mar. 2013;31(3):230-2. doi: 10.1038/nbt.2507. Epub Jan. 29, 2013.
Cho et al., The calcium-activated chloride channel anoctamin 1 acts as a heat sensor in nociceptive neurons. Nat Neurosci. May 27, 2012;15(7):1015-21. doi: 10.1038/nn.3111.
Choe et al., Forging Ahead through Darkness: PCNA, Still the Principal Conductor at the Replication Fork. Mol Cell. Feb. 2, 2017;65(3):380-392. doi: 10.1016/j.molcel.2016.12.020.
Choi et al., N(6)-methyladenosine in mRNA disrupts tRNA selection and translation-elongation dynamics. Nat Struct Mol Biol. Feb. 2016;23(2):110-5. doi: 10.1038/nsmb.3148. Epub Jan. 11, 2016.
Choi et al., Protein trans-splicing and characterization of a split family B-type DNA polymerase from the hyperthermophilic archaeal parasite Nanoarchaeum equitans. J Mol Biol. Mar. 10, 2006;356(5):1093-106. doi: 10.1016/j.jmb.2005.12.036. Epub Dec. 27, 2005.
Choi et al., Translesion synthesis across abasic lesions by human B-family and Y-family DNA polymerases α, δ, η, ι, κ, and REV1. J Mol Biol. Nov. 19, 2010;404(1):34-44. doi: 10.1016/j.jmb.2010.09.015. Epub Oct. 1, 2010.
Chong et al., Utilizing the C-terminal cleavage activity of a protein splicing element to purify recombinant proteins in a single chromatographic step. Nucleic Acids Res. Nov. 15, 1998;26(22):5109-15. doi: 10.1093/nar/26.22.5109.
Chong et al., Modulation of protein splicing of the Saccharomyces cerevisiae vacuolar membrane ATPase intein. J Biol Chem. Apr. 24, 1998;273(17):10567-77. doi: 10.1074/jbc.273.17.10567.
Chong et al., Protein splicing involving the Saccharomyces cerevisiae VMA intein. The steps in the splicing pathway, side reactions leading to protein cleavage, and establishment of an in vitro splicing system. J Biol Chem. Sep. 6, 1996;271(36):22159-68. doi: 10.1074/jbc.271.36.22159.
Chong et al., Protein splicing of the Saccharomyces cerevisiae VMA intein without the endonuclease motifs. J Biol Chem. Jun. 20, 1997;272(25):15587-90. doi: 10.1074/jbc.272.25.15587.
Chong et al., Single-column purification of free recombinant proteins using a self-cleavable affinity tag derived from a protein splicing element. Gene. Jun. 19, 1997;192(2):271-81. doi: 10.1016/s0378-1119(97)00105-4.
Choudhury et al., CRISPR-dCas9 mediated TET1 targeting for selective DNA demethylation at BRCA1 promoter. Oncotarget. Jul. 19, 2016;7(29):46545-46556. doi: 10.18632/oncotarget.10234.
Choudhury et al., CRISPR/Cas9 recombineering-mediated deep mutational scanning of essential genes in Escherichia coli. Mol Syst Biol. Mar. 2020; 16(3):e9265. doi: 10.15252/msb.20199265.
Choudhury et al., Engineering RNA endonucleases with customized sequence specificities. Nat Commun. 2012;3:1147. doi: 10.1038/ncomms2154.
Choulika et al., Induction of homologous recombination in mammalian chromosomes by using the I-Scel system of Saccharomyces cerevisiae. Mol Cell Biol. Apr. 1995;15(4):1968-73. doi: 10.1128/MCB.15.4.1968.
Christian et al., Targeting G with TAL effectors: a comparison of activities of TALENs constructed with NN and NK repeat variable di-residues. PLoS One. 2012;7(9):e45383. doi: 10.1371/journal.pone.0045383. Epub Sep. 24, 2012.
Christian et al., Targeting DNA double-strand breaks with TAL effector nucleases. Genetics. Oct. 2010;186(2):757-61. Doi: 10.1534/genetics.110.120717. Epub Jul. 26, 2010.
Christiansen et al., Characterization of the lactococcal temperate phage TP901-1 and its site-specific integration. J Bacteriol. Feb. 1994;176(4):1069-76. doi: 10.1128/jb.176.4.1069-1076.1994.
Chu et al., Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat Biotech. Feb. 13, 2015;33:543-8. doi: 10.1038/nbt.3198. Epub Mar. 24, 2015.
Chuai et al., DeepCRISPR: optimized CRISPR guide RNA design by deep learning. Genome Biol. Jun. 26, 2018;19(1):80. doi: 10.1186/s13059-018-1459-4.
Chuai et al., In Silico Meets In Vivo: Towards Computational CRISPR-Based sgRNA Design. Trends Biotechnol. Jan. 2017;35(1):12-21. doi: 10.1016/j.tibtech.2016.06.008. Epub Jul. 11, 2016.
Chuang et al., Novel Heterotypic Rox Sites for Combinatorial Dre Recombination Strategies. G3 (Bethesda). Dec. 29, 2015;6(3):559-71. doi: 10.1534/g3.115.025841.
Chujo et al., Trmt61B is a methyltransferase responsible for 1-methyladenosine at position 58 of human mitochondrial tRNAs. RNA. Dec. 2012;18(12):2269-76. doi: 10.1261/rna.035600.112. Epub Oct. 24, 2012.
Chung-Il et al., Artificial control of gene expression in mammalian cells by modulating RNA interference through aptamer-small molecule interaction. RNA. May 2006;12(5):710-6. Epub Apr. 10, 2006.
Chylinski et al., The tracrRNA and Cas9 families of type II CRISPR-Cas immunity systems. RNA Biol. May 2013;10(5):726-37. doi: 10.4161/rna.24321. Epub Apr. 5, 2013.
Clackson et al., Redesigning an FKBP-ligand interface to generate chemical dimerizers with novel specificity. Proc Natl Acad Sci U S A. Sep. 1, 1998;95(18):10437-42. doi: 10.1073/pnas.95.18.10437.
Clement et al., CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat Biotechnol. Mar. 2019;37(3):224-226. doi: 10.1038/s41587-019-0032-3.
Cobb et al., Directed evolution as a powerful synthetic biology tool. Methods. Mar. 15, 2013;60(1):81-90. doi: 10.1016/j.ymeth.2012.03.009. Epub Mar. 23, 2012.
Cokol et al., Finding nuclear localization signals. EMBO Rep. Nov. 2000;1(5):411-5. doi: 10.1093/embo-reports/kvd092.
Cole et al., Reconstructing evolutionary adaptive paths for protein engineering. Methods Mol Biol. 2013;978:115-25. doi: 10.1007/978-1-62703-293-3_8.
Cole-Strauss et al., Correction of the mutation responsible for sickle cell anemia by an RNA-DNA oligonucleotide. Science. Sep. 6, 1996;273(5280):1386-9.
Collinge, Prion diseases of humans and animals: their causes and molecular basis. Annu Rev Neurosci. 2001;24:519-50. doi: 10.1146/annurev.neuro.24.1.519.
Cong et al., Multiplex genome engineering using CRISPR/Cas systems. Science. Feb. 15, 2013;339(6121):819-23. doi: 10.1126/science.1231143. Epub Jan. 3, 2013.
Conrad et al., A Kaposi's sarcoma virus RNA element that increases the nuclear abundance of intronless transcripts. EMBO J. May 18, 2005;24(10):1831-41. doi: 10.1038/sj.emboj.7600662. Epub Apr. 28, 2005.
Conticello, The AID/APOBEC family of nucleic acid mutators. Genome Biol. 2008;9(6):229. doi: 10.1186/GB-2008-9-6-229. Epub Jun. 17, 2008.
Corcia et al., The importance of the SMN genes in the genetics of sporadic ALS. Amyotroph Lateral Scler. Oct.-Dec. 2009; 10(5-6):436-40. doi: 10.3109/17482960902759162.
Cornu et al., Refining strategies to translate genome editing to the clinic. Nat Med. Apr. 3, 2017;23(4):415-423. doi: 10.1038/nm.4313.
Corti et al., Genetic correction of human induced pluripotent stem cells from patients with spinal muscular atrophy. Sci Transl Med. Dec. 19, 2012;4(165):165ra162. doi: 10.1126/scitranslmed.3004108.
Costa et al., Frequent use of the same tertiary motif by self-folding RNAs. EMBO J. Mar. 15, 1995;14(6):1276-85.
Cotton et al., Insertion of a Synthetic Peptide into a Recombinant Protein Framework: A Protein Biosensor. J. Am. Chem. Soc. Jan. 22, 1999; 121(5):1100-1. https://doi.org/10.1021/ja983804b.
Covino et al., The CCL2/CCR2 Axis in the Pathogenesis of HIV-1 Infection: A New Cellular Target for Therapy? Current Drug Targets Dec. 2016;17(1):76-110. DOI : 10.2174/138945011701151217110917.
Cox et al., An SCN9A channelopathy causes congenital inability to experience pain. Nature. Dec. 14, 2006;444(7121):894-8. doi: 10.1038/nature05413.
Cox et al., Conditional gene expression in the mouse inner ear using Cre-loxP. J Assoc Res Otolaryngol. Jun. 2012;13(3):295-322. doi: 10.1007/s10162-012-0324-5. Epub Apr. 24, 2012.
Cox et al., Congenital insensitivity to pain: novel SCN9A missense and in-frame deletion mutations. Hum Mutat. Sep. 2010;31(9):E1670-86. doi: 10.1002/humu.21325.
Cox et al., RNA editing with CRISPR-Cas13. Science. Nov. 24, 2017;358(6366):1019-1027. doi: 10.1126/science.aaq0180. Epub Oct. 25, 2017.
Cox et al., Therapeutic genome editing: prospects and challenges. Nat Med. Feb. 2015;21(2):121-31. doi: 10.1038/nm.3793.
Cox, Proteins pinpoint double strand breaks. Elife. Oct. 29, 2013;2:e01561. doi: 10.7554/eLife.01561.
Crabtree et al., Three-part inventions: intracellular signaling and induced proximity. Trends Biochem Sci. Nov. 1996;21(11):418-22. doi: 10.1016/s0968-0004(96)20027-1.
Cradick et al., CRISPR/Cas9 systems targeting ?-globin and CCR5 genes have substantial off-target activity. Nucleic Acids Res. Nov. 1, 2013;41(20):9584-92. doi: 10.1093/nar/gkt714. Epub Aug. 11, 2013.
Cradick et al., ZFN-site searches genomes for zinc finger nuclease target sites and off-target sites. BMC Bioinformatics. May 13, 2011;12:152. doi: 10.1186/1471-2105-12-152.
Cradick et al., Zinc-finger nucleases as a novel therapeutic strategy for targeting hepatitis B virus DNAs. Mol Ther. May 2010;18(5):947-54. Doi: 10.1038/mt.2010.20. Epub Feb. 16, 2010.
Crick, On protein synthesis. Symp Soc Exp Biol. 1958;12:138-63.
Cronican et al., A class of human proteins that deliver functional proteins into mammalian cells in vitro and in vivo. Chem Biol. Jul. 29, 2011;18(7):833-8. doi: 10.1016/j.chembiol.2011.07.003.
Cronican et al., Potent delivery of functional proteins into Mammalian cells in vitro and in vivo using a supercharged protein. ACS Chem Biol. Aug. 20, 2010;5(8):747-52. doi:10.1021/cb1001153.
Crystal, Transfer of genes to humans: early lessons and obstacles to success. Science. Oct. 20, 1995;270(5235):404-10. doi: 10.1126/science.270.5235.404.
Cucchiarini et al., Enhanced expression of the central survival of motor neuron (SMN) protein during the pathogenesis of osteoarthritis. J Cell Mol Med. Jan. 2014;18(1):115-24. doi: 10.1111/jcmm.12170. Epub Nov. 17, 2013.
Cui et al., Consequences of Cas9 cleavage in the chromosome of Escherichia coli. Nucleic Acids Res. May 19, 2016;44(9):4243-51. doi: 10.1093/nar/gkw223. Epub Apr. 8, 2016.
Cui et al., m6A RNA Methylation Regulates the Self-Renewal and Tumorigenesis of Glioblastoma Stem Cells. Cell Rep. Mar. 14, 2017;18(11):2622-2634. doi: 10.1016/j.celrep.2017.02.059.
Cui et al., Review of CRISPR/Cas9 sgRNA Design Tools. Interdiscip Sci. Jun. 2018;10(2):455-465. doi: 10.1007/s12539-018-0298-z. Epub Apr. 11, 2018.
Cui et al., Targeted integration in rat and mouse embryos with zinc-finger nucleases. Nat Biotechnol. Jan. 2011;29(1):64-7. Doi: 10.1038/nbt.1731. Epub Dec. 12, 2010.
Cunningham et al., Ensembl 2015. Nucleic Acids Res. Jan. 2015;43(Database issue):D662-9. doi: 10.1093/nar/gku1010. Epub Oct. 28, 2014.
Cupples et al., A set of lacZ mutations in Escherichia coli that allow rapid detection of each of the six base substitutions. Proc Natl Acad Sci U S A. Jul. 1989;86(14):5345-9.
D'Adda di Fagagna et al., The Gam protein of bacteriophage Mu is an orthologue of eukaryotic Ku. EMBO Rep. Jan. 2003;4(1):47-52.
D'YDEWALLE et al., The Antisense Transcript SMN-AS1 Regulates SMN Expression and Is a Novel Therapeutic Target for Spinal Muscular Atrophy. Neuron. Jan. 4, 2017;93(1):66-79 and Supplemental Information. doi: 10.1016/j.neuron.2016.11.033. Epub Dec. 22, 2016.
Dahlem et al., Simple methods for generating and detecting locus-specific mutations induced with TALENs in the zebrafish genome. PLoS Genet. 2012;8(8):e1002861. doi: 10.1371/journal.pgen.1002861. Epub Aug. 16, 2012.
Dahlgren et al., A novel mutation in ribosomal protein S4 that affects the function of a mutated RF1. Biochimie. Aug. 2000;82(8):683-91.
Dahlman et al., Orthogonal gene knockout and activation with a catalytically active Cas9 nuclease. Nat Biotechnol. Nov. 2015;33(11):1159-61. doi: 10.1038/nbt.3390.
Dandage et al., beditor: A Computational Workflow for Designing Libraries of Guide RNAs for CRISPR-Mediated Base Editing. Genetics. Jun. 2019;212(2):377-385. doi: 10.1534/genetics.119.302089. Epub Apr. 1, 2019.
Dang et al., Optimizing sgRNA structure to improve CRISPR-Cas9 knockout efficiency. Genome Biol. Dec. 15, 2015;16:280. doi: 10.1186/s13059-015-0846-3.
Das et al., The crystal structure of the monomeric reverse transcriptase from Moloney murine leukemia virus. Structure. May 2004;12(5):819-29. doi: 10.1016/j.str.2004.02.032.
Dassa et al., Fractured genes: a novel genomic arrangement involving new split inteins and a new homing endonuclease family. Nucleic Acids Res. May 2009;37(8):2560-73. doi: 10.1093/nar/gkp095. Epub Mar. 5, 2009.
Dassa et al., Trans protein splicing of cyanobacterial split inteins in endogenous and exogenous combinations. Biochemistry. Jan. 9, 2007;46(1):322-30. doi: 10.1021/bi0611762.
Database EBI Accession No. ADE34233 Jan. 29, 2004.
Database EBI Accession No. BFF09785. May 31, 2018. 2 pages.
Database EBI Accession No. BGE38086. Jul. 25, 2019. 2 pages.
Database UniProt Accession No. G8I3E0. Jan. 14, 2012.
Datsenko et al., One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc Natl Acad Sci U S A. Jun. 6, 2000;97(12):6640-5.
Davidson et al., Viral vectors for gene delivery to the nervous system. Nat Rev Neurosci. May 2003;4(5):353-64. doi: 10.1038/nrn1104.
Davis et al., Assaying Repair at DNA Nicks. Methods Enzymol. 2018;601:71-89. doi: 10.1016/bs.mie.2017.12.001. Epub Feb. 1, 2018.
Davis et al., DNA double strand break repair via non-homologous end-joining. Transl Cancer Res. Jun. 2013;2(3):130-143.
Davis et al., Homology-directed repair of DNA nicks via pathways distinct from canonical double-strand break repair. Proc Natl Acad Sci U S A. Mar. 11, 2014;111(10):E924-32. doi: 10.1073/pnas.1400236111. Epub Feb. 20, 2014.
Davis et al., Small molecule-triggered Cas9 protein with improved genome-editing specificity. Nat Chem Biol. May 2015;11(5):316-8. doi: 10.1038/nchembio.1793. Epub Apr. 6, 2015.
Davis et al., Two Distinct Pathways Support Gene Correction by Single-Stranded Donors at DNA Nicks. Cell Rep. Nov. 8, 2016;17(7):1872-1881. doi: 10.1016/j.celrep.2016.10.049.
De Felipe et al., Co-translational, intraribosomal cleavage of polypeptides by the foot-and-mouth disease virus 2A peptide. J Biol Chem. Mar. 28, 2003;278(13):11441-8. doi: 10.1074/jbc.M211644200. Epub Jan. 8, 2003.
De La Peña et al., The Hammerhead Ribozyme: A Long History for a Short RNA. Molecules. Jan. 4, 2017;22(1):78. doi: 10.3390/molecules22010078.
De Sandre-Giovannoli et al., Lamin a truncation in Hutchinson-Gilford progeria. Science. Jun. 27, 2003;300(5628):2055. doi: 10.1126/science.1084125. Epub Apr. 17, 2003.
De Souza, Primer: genome editing with engineered nucleases. Nat Methods. Jan. 2012;9(1):27.
De Wit et al., The Human CD4+ T Cell Response against Mumps Virus Targets a Broadly Recognized Nucleoprotein Epitope. J Virol. Mar. 5, 2019;93(6):e01883-18. doi: 10.1128/JVI.01883-18.
Dean et al., Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Francisco City Cohort, ALIVE Study. Science. Sep. 27, 1996;273(5283):1856-62. doi: 10.1126/science.273.5283.1856.
Dekosky et al., Large-scale sequence and structural comparisons of human naive and antigen-experienced antibody repertoires. Proc Natl Acad Sci U S A. May 10, 2016;113(19):E2636-45. doi: 10.1073/pnas.1525510113. Epub Apr. 25, 2016.
Delebecque et al., Organization of intracellular reactions with rationally designed RNA assemblies. Science. Jul. 22, 2011;333(6041):470-4. doi: 10.1126/science.1206938. Epub Jun. 23, 2011.
Deltcheva et al., Crispr RNA maturation by trans-encoded small RNA and host factor RNase III. Nature. Mar. 31, 2011;471(7340):602-7. doi: 10.1038/nature09886.
Deng et al., Widespread occurrence of N6-methyladenosine in bacterial mRNA. Nucleic Acids Res. Jul. 27, 2015;43(13):6557-67. doi: 10.1093/nar/gkv596. Epub Jun. 11, 2015.
Denizio et al., Harnessing natural DNA modifying activities for editing of the genome and epigenome. Curr Opin Chem Biol. Aug. 2018;45:10-17. doi: 10.1016/j.cbpa.2018.01.016. Epub Feb. 13, 2018.
Deriano et al., Modernizing the nonhomologous end-joining repertoire: alternative and classical NHEJ share the stage. Annu Rev Genet. 2013;47:433-55. doi: 10.1146/annurev-genet-110711-155540. Epub Sep. 11, 2013.
Deussing, Targeted mutagenesis tools for modelling psychiatric disorders. Cell Tissue Res. Oct. 2013;354(1):9-25. doi: 10.1007/s00441-013-1708-5. Epub Sep. 10, 2013.
Dever et al., CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells. Nature. Nov. 17, 2016;539(7629):384-389. doi: 10.1038/nature20134. Epub Nov. 7, 2016.
Deverman et al., Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat Biotechnol. Feb. 2016;34(2):204-9. doi: 10.1038/nbt.3440. Epub Feb. 1, 2016.
Devigili et al., Paroxysmal itch caused by gain-of-function Nav1.7 mutation. Pain. Sep. 2014;155(9):1702-1707. doi: 10.1016/j.pain.2014.05.006. Epub May 10, 2014.
Dianov et al., Mammalian base excision repair: the forgotten archangel. Nucleic Acids Res. Apr. 1, 2013;41(6):3483-90. doi: 10.1093/nar/gkt076. Epub Feb. 13, 2013.
Dicarlo et al., Genome engineering in Saccharomyces cerevisiae using CRISPR-Cas systems. Nucleic Acids Res. Apr. 2013;41(7):4336-43. doi: 10.1093/nar/gkt135. Epub Mar. 4, 2013.
Dicarlo et al., Safeguarding CRISPR-Cas9 gene drives in yeast. Nat Biotechnol. Dec. 2015;33(12):1250-1255. doi: 10.1038/nbt.3412. Epub Nov. 16, 2015.
Dickey et al., Single-stranded DNA-binding proteins: multiple domains for multiple functions. Structure. Jul. 2, 2013;21(7):1074-84. doi: 10.1016/j.str.2013.05.013.
Dickinson et al., A system for the continuous directed evolution of proteases rapidly reveals drug-resistance mutations. Nat Commun. Oct. 30, 2014;5:5352. doi: 10.1038/ncomms6352.
Dickinson et al., Experimental interrogation of the path dependence and stochasticity of protein evolution using phage-assisted continuous evolution. Proc Natl Acad Sci USA. May 2013;110(22):9007-12.
Dillon, Regulating gene expression in gene therapy. Trends Biotechnol. May 1993;11(5):167-73. doi: 10.1016/0167-7799(93)90109-M.
Ding et al., A Talen genome-editing system for generating human stem cell-based disease models. Cell Stem Cell. Feb. 7, 2013;12(2):238-51. Doi: 10.1016/j.stem.2012.11.011. Epub Dec. 13, 2012.
Ding et al., Permanent alteration of PCSK9 with in vivo CRISPR-Cas9 genome editing. Circ Res. Aug. 15, 2014;115(5):488-92. doi: 10.1161/CIRCRESAHA.115.304351. Epub Jun. 10, 2014.
Dingwall et al., Nuclear targeting sequences—a consensus? Trends Biochem Sci. Dec. 1991;16(12):478-81. doi: 10.1016/0968-0004(91)90184-w.
Diver et al., Single-Step Synthesis of Cell-Permeable Protein Dimerizers That Activate Signal Transduction and Gene Expression. J. Am. Chem. Soc. Jun. 4, 1997;119(22):5106-5109. https://doi.org/10.1021/ja963891c.
Dixon et al., Reengineering orthogonally selective riboswitches. Proc Natl Acad Sci U S A. Feb. 16, 2010;107(7):2830-5. doi: 10.1073/pnas.0911209107. Epub Jan. 26, 2010.
Doench et al., Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat Biotechnol. Feb. 2016;34(2):184-191. doi: 10.1038/nbt.3437.
Doench et al., Rational design of highly active sgRNAs for CRISPR-Cas9-mediated gene inactivation. Nat Biotechnol. Dec. 2014;32(12):1262-7. doi: 10.1038/nbt.3026. Epub Sep. 3, 2014.
Dolan et al., Trans-splicing with the group I intron ribozyme from Azoarcus. RNA. Feb. 2014;20(2):202-13. doi: 10.1261/rna.041012.113. Epub Dec. 16, 2013.
Doman et al., Evaluation and minimization of Cas9-independent off-target DNA editing by cytosine base editors. Nat Biotechnol. May 2020;38(5):620-628. doi: 10.1038/s41587-020-0414-6. Epub Feb. 10, 2020.
Dominissini et al., Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. Apr. 29, 2012;485(7397):201-6. doi: 10.1038/nature11112.
Dorgan et al., An enzyme-coupled continuous spectrophotometric assay for S-adenosylmethionine-dependent methyltransferases. Anal Biochem. Mar. 15, 2006;350(2):249-55. doi: 10.1016/j.ab.2006.01.004. Epub Feb. 7, 2006.
Dormiani et al., Long-term and efficient expression of human β-globin gene in a hematopoietic cell line using a new site-specific integrating non-viral system. Gene Ther. Aug. 2015;22(8):663-74. doi: 10.1038/gt.2015.30. Epub Apr. 1, 2015.
Dorr et al., Reprogramming the specificity of sortase enzymes. Proc Natl Acad Sci U S A. Sep. 16, 2014;111(37):13343-8. doi: 10.1073/pnas.1411179111. Epub Sep. 3, 2014.
Doudna et al., Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science. Nov. 28, 2014;346(6213):1258096. doi: 10.1126/science.1258096.
Doudna, The promise and challenge of therapeutic genome editing. Nature. Feb. 2020;578(7794):229-236. doi: 10.1038/s41586-020-1978-5. Epub Feb. 12, 2020.
Dove et al., Conversion of the omega subunit of Escherichia coli RNA polymerase into a transcriptional activator or an activation target. Genes Dev. Mar. 1, 1998;12(5):745-54.
Doyon et al., Directed evolution and substrate specificity profile of homing endonuclease I-SceI. J Am Chem Soc. Feb. 22, 2006;128(7):2477-84.
Doyon et al., Heritable targeted gene disruption in zebrafish using designed zinc-finger nucleases. Nat Biotechnol. Jun. 2008;26(6):702-8. Doi: 10.1038/nbt1409. Epub May 25, 2008.
Drake, A constant rate of spontaneous mutation in DNA-based microbes. Proc Natl Acad Sci USA. Aug. 15, 1991;88(16):7160-4.
Drenth et al., Mutations in sodium-channel gene SCN9A cause a spectrum of human genetic pain disorders. J Clin Invest. Dec. 2007;117(12):3603-9. doi: 10.1172/JCI33297.
Drost et al., Inactivation of DNA mismatch repair by variants of uncertain significance in the PMS2 gene. Hum Mutat. Nov. 2013;34(11):1477-80. doi: 10.1002/humu.22426. Epub Sep. 11, 2013.
Duan et al., Enhancement of muscle gene delivery with pseudotyped adeno-associated virus type 5 correlates with myoblast differentiation. J Virol. Aug. 2001;75(16):7662-71. doi: 10.1128/JVI.75.16.7662-7671.2001.
Dubois et al., Retroviral RNA Dimerization: From Structure to Functions. Front Microbiol. Mar. 22, 2018;9:527. doi: 10.3389/fmicb.2018.00527.
Dugar et al., CRISPR RNA-Dependent Binding and Cleavage of Endogenous RNAs by the Campylobacter jejuni Cas9. Mol Cell. Mar. 1, 2018;69(5):893-905.e7. doi: 10.1016/j.molcel.2018.01.032.
Dumas et al., Designing logical codon reassignment—Expanding the chemistry in biology. Chem Sci. Jan. 1, 2015;6(1):50-69. doi: 10.1039/c4sc01534g. Epub Jul. 14, 2014. Review.
Dunaime, Breakthrough method means CRISPR just got a lot more relevant to human health. The Verge. Apr. 20, 2016. http://www.theverge.com/2016/4/20/11450262/crispr-base-editing-single-nucleotides-dna-gene-liu-harvard.
Dunbar et al., Gene therapy comes of age. Science. Jan. 12, 2018;359(6372):eaan4672. doi: 10.1126/science.aan4672.
Dupuy et al., Le syndrome de De La Chapelle [De La Chapelle syndrome]. Presse Med. Mar. 3, 2001;30(8):369-72. French.
Durai et al., A bacterial one-hybrid selection system for interrogating zinc finger-DNA interactions. Comb Chem High Throughput Screen. May 2006;9(4):301-11.
Durai et al., Zinc finger nucleases: custom-designed molecular scissors for genome engineering of plant and mammalian cells. Nucleic Acids Res. Oct. 26, 2005;33(18):5978-90. doi: 10.1093/nar/gki912.
During et al., Controlled release of dopamine from a polymeric brain implant: in vivo characterization. Ann Neurol. Apr. 1989;25(4):351-6.
East-Seletsky et al., Two distinct RNase activities of CRISPR-C2c2 enable guide-RNA processing and RNA detection. Nature Oct. 2016;538(7624):270-3.
Edlund et al., Cell-specific expression of the rat insulin gene: evidence for role of two distinct 5′ flanking elements. Science. Nov. 22, 1985;230(4728):912-6. doi: 10.1126/science.3904002.
Edwards et al., An Escherichia coli tyrosine transfer RNA is a leucine-specific transfer RNA in the yeast Saccharomyces cerevisiae. Proc Natl Acad Sci U S A. Feb. 15, 1991;88(4):1153-6.
Edwards et al., Crystal structures of the thi-box riboswitch bound to thiamine pyrophosphate analogs reveal adaptive RNA-small molecule recognition. Structure. Sep. 2006;14(9):1459-68.
Eick et al., Robustness of Reconstructed Ancestral Protein Functions to Statistical Uncertainty. Mol Biol Evol. Feb. 1, 2017;34(2):247-261. doi: 10.1093/molbev/msw223.
Eiler et al., Structural Basis for the Fast Self-Cleavage Reaction Catalyzed by the Twister Ribozyme. Proc Natl Acad Sci U S A. Sep. 9, 2014;111(36):13028-33. doi: 10.1073/pnas.1414571111. Epub Aug. 25, 2014.
Eisenberg et al., A-to-I RNA editing—immune protector and transcriptome diversifier. Nat Rev Genet. Aug. 2018; 19(8):473-490. doi: 10.1038/s41576-018-0006-1.
Ekstrand et al., Frequent alterations of the PI3K/AKT/mTOR pathways in hereditary nonpolyposis colorectal cancer. Fam Cancer. Jun. 2010;9(2):125-9. doi: 10.1007/s10689-009-9293-1.
Eltoukhy et al., Nucleic acid-mediated intracellular protein delivery by lipid-like nanoparticles. Biomaterials. Aug. 2014;35(24):6454-61. doi: 10.1016/j.biomaterials.2014.04.014. Epub May 13, 2014.
Emery et al., HCN2 ion channels play a central role in inflammatory and neuropathic pain. Science. Sep. 9, 2011;333(6048):1462-6. doi: 10.1126/science.1206243.
Endo et al., Toward establishing an efficient and versatile gene targeting system in higher plants. Biocatalysis and Agricultural Biotechnology 2014;3,(1):2-6.
Engel et al., The emerging role of mRNA methylation in normal and pathological behavior. Genes Brain Behav. Mar. 2018;17(3):e12428. doi: 10.1111/gbb.12428. Epub Nov. 17, 2017.
Engelward et al., Base excision repair deficient mice lacking the Aag alkyladenine DNA glycosylase. Proc Natl Acad Sci U S A. Nov. 25, 1997;94(24):13087-92.
England, Unnatural amino acid mutagenesis: a precise tool for probing; protein structure and function. Biochemistry. Sep. 21, 2004;43(37):11623-9.
Entin-Meer et al., The role of phenylalanine-119 of the reverse transcriptase of mouse mammary tumour virus in DNA synthesis, ribose selection and drug resistance. Biochem J. Oct. 15, 2002;367(Pt 2):381-91. doi: 10.1042/BJ20020712.
Enyeart et al., Biotechnological applications of mobile group II introns and their reverse transcriptases: gene targeting, RNA-seq, and non-coding RNA analysis. Mobile DNA 5, 2(2014). https://doi.org/10.1186/1759-8753-5-2. https://doi.org/10.1186/1759-8753-5-2.
Epstein, HSV-1-based amplicon vectors: design and applications. Gene Ther. Oct. 2005;12 Suppl 1:S154-8. doi: 10.1038/sj.gt.3302617.
Eriksson et al., Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature. May 15, 2003;423(6937):293-8. doi: 10.1038/nature01629. Epub Apr. 25, 2003. PMID: 12714972.
Estacion et al., A sodium channel gene SCN9A polymorphism that increases nociceptor excitability. Ann Neurol. Dec. 2009;66(6):862-6. doi: 10.1002/ana.21895.
Esvelt et al., A system for the continuous directed evolution of biomolecules. Nature. Apr. 28, 2011;472(7344):499-503. doi: 10.1038/nature09929. Epub Apr. 10, 2011.
Esvelt et al., Genome-scale engineering for systems and synthetic biology. Mol Syst Biol. 2013;9:641. doi: 10.1038/msb.2012.66.
Esvelt et al., Orthogonal Cas9 proteins for RNA-guided gene regulation and editing. Nat Methods. Nov. 2013;10(11):1116-21. doi: 10.1038/nmeth.2681. Epub Sep. 29, 2013.
Evans et al., Protein trans-splicing and cyclization by a naturally split intein from the dnaE gene of Synechocystis species PCC6803. J Biol Chem. Mar. 31, 2000;275(13):9091-4. doi: 10.1074/jbc.275.13.9091.
Evans et al., Semisynthesis of cytotoxic proteins using a modified protein splicing element. Protein Sci. Nov. 1998;7(11):2256-64. doi: 10.1002/pro.5560071103.
Evans et al., The cyclization and polymerization of bacterially expressed proteins using modified self-splicing inteins. J Biol Chem. Jun. 25, 1999;274(26):18359-63. doi: 10.1074/jbc.274.26.18359.
Evans et al., The in vitro ligation of bacterially expressed proteins using an intein from Methanobacterium thermoautotrophicum. J Biol Chem. Feb. 12, 1999;274(7):3923-6. doi: 10.1074/jbc.274.7.3923.
Evers et al., CRISPR knockout screening outperforms shRNA and CRISPRi in identifying essential genes. Nat Biotechnol. Jun. 2016;34(6):631-3. doi: 10.1038/nbt.3536. Epub Apr. 25, 2016.
Fagerlund et al., The Cpf1 CRISPR-Cas protein expands genome-editing tools. Genome Biology Nov. 17, 2015;16:251. https://doi.org/10.1186/s13059-015-0824-9.
Falnes et al., DNA repair by bacterial AlkB proteins. Res Microbiol. Oct. 2003;154(8):531-8. doi: 10.1016/S0923-2508(03)00150-5.
Falnes et al., Repair of methyl lesions in DNA and RNA by oxidative demethylation. Neuroscience. Apr. 14, 2007;145(4):1222-32. doi: 10.1016/j.neuroscience.2006.11.018. Epub Dec. 18, 2006.
Fang et al., Synthetic Studies Towards Halichondrins: Synthesis of the Left Halves of Norhalichondrins and Homohalichondrins. Tetrahedron Letters 1992;33(12):1557-1560.
Farboud et al., Dramatic enhancement of genome editing by CRISPR/Cas9 through improved guide RNA design. Genetics. Apr. 2015;199(4):959-71. doi: 10.1534/genetics.115.175166. Epub Feb. 18, 2015.
Farhood et al., Codelivery to mammalian cells of a transcriptional factor with cis-acting element using cationic liposomes. Anal Biochem. Feb. 10, 1995;225(1):89-93.
Fawcett et al., Transposable elements controlling I-R hybrid dysgenesis in D. melanogaster are similar to mammalian LINEs. Cell. Dec. 26, 1986;47(6):1007-15. doi: 10.1016/0092-8674(86)90815-9.
Feldstein et al., Two sequences participating in the autolytic processing of satellite tobacco ringspot virus complementary RNA. Gene. Oct. 15, 1989;82(1):53-61. doi: 10.1016/0378-1119(89)90029-2.
Felletti et al., Twister Ribozymes as Highly Versatile Expression Platforms for Artificial Riboswitches. Nat Commun. Sep. 27, 2016;7:12834. doi: 10.1038/ncomms12834.
Feng et al., Crystal structures of the human RNA demethylase Alkbh5 reveal basis for substrate recognition. J Biol Chem. Apr. 25, 2014;289(17):11571-11583. doi: 10.1074/jbc.M113.546168. Epub Mar. 10, 2014.
Feng et al., Human L1 retrotransposon encodes a conserved endonuclease required for retrotransposition. Cell. Nov. 29, 1996;87(5):905-16. doi: 10.1016/s0092-8674(00)81997-2.
Ferreira Da Silva et al., Prime editing efficiency and fidelity are enhanced in the absence of mismatch repair. Nat Commun. Feb. 9, 2022;13(1):760. doi: 10.1038/s41467-022-28442-1.
Ferretti et al., Complete genome sequence of an M1 strain of Streptococcus pyogenes. Proc Natl Acad Sci U S A. Apr. 10, 2001;98(8):4658-63.
Ferry et al., Rational design of inducible CRISPR guide RNAs for de novo assembly of transcriptional programs. Nat Commun. Mar. 3, 2017;8:14633. doi: 10.1038/ncomms14633.
Feuk, Inversion variants in the human genome: role in disease and genome architecture. Genome Med. Feb. 12, 2010;2(2):11. doi: 10.1186/gm132.
Filippov et al., A novel type of RNase III family proteins in eukaryotes. Gene. Mar. 7, 2000;245(1):213-21. doi: 10.1016/s0378-1119(99)00571-5.
Filippova et al., Guide RNA modification as a way to improve CRISPR/Cas9-based genome-editing systems. Biochimie. Dec. 2019;167:49-60. doi: 10.1016/j.biochi.2019.09.003. Epub Sep. 4, 2019.
Fine et al., Trans-spliced Cas9 allows cleavage of HBB and CCR5 genes in human cells using compact expression cassettes. Scientific Reports 2015;5(1):Article No. 10777. doi:10.1038/srep10777. With Supplementary Information.
Fire et al., Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. Feb. 19, 1998;391(6669):806-11. doi: 10.1038/35888.
Fischbach et al., Directed evolution can rapidly improve the activity of chimeric assembly-line enzymes. Proc Natl Acad Sci U S A. Jul. 17, 2007;104(29):11951-6. doi: 10.1073/pnas.0705348104. Epub Jul. 9, 2007.
Fischer et al., Cryptic epitopes induce high-titer humoral immune response in patients with cancer. J Immunol. Sep. 1, 2010;185(5):3095-102. doi: 10.4049/jimmunol.0902166. Epub Jul. 26, 2010.
Fitzjohn, Diversitree: comparative phylogenetic analyses of diversification in R. Methods in Evology and Evolution. Dec. 2012;3(6):1084-92 .doi: 10.1111/j.2041-210X.2012.00234.x.
Flajolet et al., Woodchuck hepatitis virus enhancer I and enhancer II are both involved in N-myc2 activation in woodchuck liver tumors. J Virol. Jul. 1998;72(7):6175-80. doi: 10.1128/JVI.72.7.6175-6180.1998.
Flaman et al., A rapid PCR fidelity assay. Nucleic Acids Res. Aug. 11, 1994;22(15):3259-60. doi: 10.1093/nar/22.15.3259.
Flynn et al., CRISPR-mediated genotypic and phenotypic correction of a chronic granulomatous disease mutation in human iPS cells. Exp Hematol. Oct. 2015;43(10):838-848.e3. doi: 10.1016/j.exphem.2015.06.002. Epub Jun. 19, 2015. Including supplementary figures and data.
Fogg et al., Genome Integration and Excision by a New Streptomyces bacteriophage, øJoe. Appl Environ Microbiol. Feb. 15, 2017;83(5):e02767-16. doi: 10.1128/AEM.02767-16.
Fogg et al., New applications for phage integrases. J Mol Biol. Jul. 29, 2014;426(15):2703-16. doi: 10.1016/j.jmb.2014.05.014. Epub May 22, 2014.
Fonfara et al., Phylogeny of Cas9 determines functional exchangeability of dual-RNA and Cas9 among orthologous type II CRISPR-Cas systems. Nucleic Acids Res. Feb. 2014;42(4):2577-90. doi: 10.1093/nar/gkt1074. Epub Nov. 22, 2013. Including Supplementary Information.
Forster et al., Self-cleavage of virusoid RNA is performed by the proposed 55-nucleotide active site. Cell. Jul. 3, 1987;50(1):9-16. doi: 10.1016/0092-8674(87)90657-x.
Fortini et al., Different DNA polymerases are involved in the short- and long-patch base excision repair in mammalian cells. Biochemistry. Mar. 17, 1998;37(11):3575-80. doi: 10.1021/bi972999h.
Fouts et al., Sequencing Bacillus anthracis typing phages gamma and cherry reveals a common ancestry. J Bacteriol. May 2006;188(9):3402-8. doi: 10.1128/JB.188.9.3402-3408.2006.
Freitas et al., Mechanisms and signals for the nuclear import of proteins. Curr Genomics. Dec. 2009;10(8):550-7. doi: 10.2174/138920209789503941.
Freshney, Culture of Animal Cells. A Manual of Basic Technique. Alan R. Liss, Inc. New York. 1983;4.
Friedman, J. H., Greedy function approximation: A gradient boosting machine. Ann. Statist. Oct. 2001;29(5):1189-232. doi: 10.1214/aos/1013203451.
Fu et al., High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol. Sep. 2013;31(9):822-6. doi: 10.1038/nbt.2623. Epub Jun. 23, 2013.
Fu et al., Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat Biotechnol. Mar. 2014;32(3):279-84. doi: 10.1038/nbt.2808. Epub Jan. 26, 2014.
Fu et al., Promises and Pitfalls of Intracellular Delivery of Proteins. Bioconjugate Chemistry. Aug. 2014;25:1602-8.
Fuchs et al., Polyarginine as a multifunctional fusion tag. Protein Sci. Jun. 2005; 14(6):1538-44.
Fujisawa et al., Disease-associated mutations in CIAS1 induce cathepsin B-dependent rapid cell death of human THP-1 monocytic cells. Blood. Apr. 1, 2007;109(7):2903-11.
Fukui et al., DNA Mismatch Repair in Eukaryotes and Bacteria. J Nucleic Acids. Jul. 27, 2010;2010. pii: 260512. doi: 10.4061/2010/260512.
Fung et al., Repair at single targeted DNA double-strand breaks in pluripotent and differentiated human cells. PLoS One. 2011;6(5):e20514. doi: 10.1371/journal.pone.0020514. Epub May 25, 2011.
Furukawa et al., In vitro selection of allosteric ribozymes that sense the bacterial second messenger c-di-GMP. Methods Mol Biol. 2014;1111:209-20. doi: 10.1007/978-1-62703-755-6_15.
Fusi et al., In Silico Predictive Modeling of CRISPR/Cas9 guide efficiency. Jun. 26, 2015; bioRxiv. http://dx.doi.org/10.1101/021568.
Gaj et al., 3rd. Genome engineering with custom recombinases. Methods Enzymol. 2014;546:79-91. doi: 10.1016/B978-0-12-801185-0.00004-0.
Gaj et al., A comprehensive approach to zinc-finger recombinase customization enables genomic targeting in human cells. Nucleic Acids Res. Feb. 6, 2013;41(6):3937-46.
Gaj et al., Enhancing the specificity of recombinase-mediated genome engineering through dimer interface redesign. J Am Chem Soc. Apr. 2, 2014;136(13):5047-56. doi: 10.1021/ja4130059. Epub Mar. 20, 2014.
Gaj et al., Expanding the scope of site-specific recombinases for genetic and metabolic engineering. Biotechnol Bioeng. Jan. 2014; 111(1):1-15. doi: 10.1002/bit.25096. Epub Sep. 13, 2013.
Gaj et al., Structure-guided reprogramming of serine recombinase DNA sequence specificity. Proc Natl Acad Sci U S A. Jan. 11, 2011;108(2):498-503. doi: 10.1073/pnas.1014214108. Epub Dec. 27, 2010.
Gaj et al., ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Biotechnol. Jul. 2013;31(7):397-405. doi: 10.1016/j.tibtech.2013.04.004. Epub May 9, 2013.
Gajula, Designing an Elusive C⋅G→G⋅C Crispr Base Editor. Trends Biochem Sci. Feb. 2019;44(2):91-94. doi: 10.1016/j.tibs.2018.10.004. Epub Nov. 13, 2018.
Gallo et al., A novel pathogenic PSEN1 mutation in a family with Alzheimer's disease: phenotypical and neuropathological features. J Alzheimers Dis. 2011;25(3):425-31. doi: 10.3233/JAD-2011-110185.
Gangopadhyay et al., Precision Control of CRISPR-Cas9 Using Small Molecules and Light. Biochemistry. Jan. 29, 2019;58(4):234-244. doi: 10.1021/acs.biochem.8b01202. Epub Jan. 22, 2019.
Gao et al., Cationic liposome-mediated gene transfer. Gene Ther. Dec. 1995;2(10):710-22.
Gao et al., DNA-guided genome editing using the Natronobacterium gregoryi Argonaute. Nat Biotechnol. Jul. 2016;34(7):768-73. doi: 10.1038/nbt.3547. Epub May 2, 2016.
Gao et al., Prime editing in mice reveals the essentiality of a single base in driving tissue-specific gene expression. Genome Biol. Mar. 16, 2021;22(1):83. doi: 10.1186/s13059-021-02304-3.
Gao et al., Self-processing of ribozyme-flanked RNAs into guide RNAs in vitro and in vivo for CRISPR-mediated genome editing. J Integr Plant Biol. Apr. 2014;56(4):343-9. doi: 10.1111/jipb.12152. Epub Mar. 6, 2014.
Gao et al., Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature. Jan. 11, 2018;553(7687):217-221. doi: 10.1038/nature25164. Epub Dec. 20, 2017.
Gapinske et al., CRISPR-SKIP: programmable gene splicing with single base editors. Genome Biol. Aug. 15, 2018;19(1):107. doi: 10.1186/s13059-018-1482-5.
Garcia et al., Transglycosylation: a mechanism for RNA modification (and editing?). Bioorg Chem. Jun. 2005;33(3):229-51. doi: 10.1016/j.bioorg.2005.01.001. Epub Feb. 23, 2005.
Gardlik et al., Vectors and delivery systems in gene therapy. Med Sci Monit. Apr. 2005; 11(4):RA110-21. Epub Mar. 24, 2005.
Garibyan et al., Use of the rpoB gene to determine the specificity of base substitution mutations on the Escherichia coli chromosome. DNA Repair (Amst). May 13, 2003;2(5):593-608.
Garneau et al., The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature. Nov. 4, 2010;468(7320):67-71. doi: 10.1038/nature09523.
Gasiunas et al., Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc Natl Acad Sci U S A. Sep. 25, 2012;109(39):E2579-86. Epub Sep. 4, 2012. Supplementary materials included.
Gasiunas et al., RNA-dependent DNA endonuclease Cas9 of the CRISPR system: Holy Grail of genome editing? Trends Microbiol. Nov. 2013;21(11):562-7. doi: 10.1016/j.tim.2013.09.001. Epub Oct. 1, 2013.
Gaudelli et al., Directed evolution of adenine base editors with increased activity and therapeutic application. Nat Biotechnol. Jul. 2020;38(7):892-900. doi: 10.1038/s41587-020-0491-6. Epub Apr. 13, 2020.
Gaudelli et al., Programmable base editing of A⋅T to G⋅C in genomic DNA without DNA cleavage. Nature. Nov. 23, 2017;551(7681):464-471. doi: 10.1038/nature24644. Epub Oct. 25, 2017. Erratum in: Nature. May 2, 2018.
Gearing, Addgene blog. CRISPR 101: Cas9 nickase design and homology directed repair. 2018. pp. 1-12. https://blog.addgene.org/crispr-101-cas9-nickase-design-and-homlogy-directed-repair. Last retrieved online Jun. 25, 2021.
Gehrke et al., An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities. Nat Biotechnol. Nov. 2018;36(10):977-982. doi: 10.1038/nbt.4199. Epub Jul. 30, 2018.
GenBank Accession No. J01600.1. Brooks et al., E.coli dam gene coding for DNA adenine methylase. Apr. 26, 1993.
GenBank Accession No. U07651.1. Lu, Escherichia coli K12 negative regulator of replication initiation (seqA) gene, complete cds. Jul. 19, 1994.
GENBANK Submission; NIH/NCBI Accession No. 4UN5_B. Anders et al., Jul. 23, 2014. 5 pages.
GENBANK Submission; NIH/NCBI Accession No. NM_001319224.2. Umar et al., Apr. 21, 2021. 7 pages.
GENBANK Submission; NIH/NCBI Accession No. NM_006027.4. Umar et al., Apr. 10, 2021. 7 pages.
GENBANK Submission; NIH/NCBI, Accession No. AAA66622.1. Martinelli et al., May 18, 1995. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. AGT42196. Farzadfar et al., Nov. 2, 2013. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. AIT42264.1. Hyun et al., Oct. 15, 2014. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. AKA60242.1. Tong et al., Apr. 5, 2015. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. AKQ21048.1. Gilles et al., Jul. 19, 2015. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. AKS40380.1. Nodvig et al., Aug. 2, 2015. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. APG80656.1. Burstein et al., Dec. 10, 2016. 1 pages.
GENBANK Submission; NIH/NCBI, Accession No. AYD60528.1. Ram et al., Oct. 2, 2018. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. BDB43378. Zhang et al., Aug. 11, 2016. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. J04623. Kita et al., Apr. 26, 1993. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. KR710351.1. Sahni et al., Jun. 1, 2015. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. NC 002737.2. Nasser et al., Feb. 7, 2021. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. NC_000001.11. Gregory et al., Jun. 6, 2016. 3 pages.
GENBANK Submission; NIH/NCBI, Accession No. NC_002737.1. Ferretti et al., Jun. 27, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. NC_015683.1. Trost et al., Jul. 6, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. NC_016782.1. Trost et al., Jun. 11, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. NC_016786.1. Trost et al., Aug. 28, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. NC_017053.1. Fittipaldi et al., Jul. 6, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. NC_017317.1. Trost et al., Jun. 11, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. NC_017861.1. Heidelberg et al., Jun. 11, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. NC_018010.1. Lucas et al., Jun. 11, 2013. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. NC_018721.1. Feng et al., Jun. 11, 2013. 1 pages.
GENBANK Submission; NIH/NCBI, Accession No. NC_021284.1. Ku et al., Jul. 12, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. NC_021314.1. Zhang et al., Jul. 15, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. NC_021846.1. Lo et al., Jul. 22, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. NG_008692.2. McClintock et al., Aug. 27, 2018. 33 pages.
GENBANK Submission; NIH/NCBI, Accession No. NM_000311.5. Alves et al., Mar. 7, 2021. 5 pages.
GENBANK Submission; NIH/NCBI, Accession No. NM_001319224. Umar et al., Apr. 21, 2021. 7 pages.
GENBANK Submission; NIH/NCBI, Accession No. NM_002945.3. Weiser et al., Sep. 3, 2017. 5 pages.
GENBANK Submission; NIH/NCBI, Accession No. NM_002946.5. Kavli et al., Jun. 26, 2021. 5 pages.
GENBANK Submission; NIH/NCBI, Accession No. NM_002947.4. Xiao et al., May 1, 2019. 4 pages.
GENBANK Submission; NIH/NCBI, Accession No. NM_003686. Umar et al., Apr. 9, 2021. 7 pages.
GENBANK Submission; NIH/NCBI, Accession No. NM_003686.4. Umar et al., Apr. 9, 2021. 7 pages.
GENBANK Submission; NIH/NCBI, Accession No. NM_006027. Umar et al., Apr. 10, 2021. 7 pages.
GENBANK Submission; NIH/NCBI, Accession No. NM_174936. Guo et al., Oct. 28, 2015. 6 pages.
GENBANK Submission; NIH/NCBI, Accession No. NM_206933.2. Khalaileh et al., Sep. 16, 2018. 12 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_000302.1. Alves et al., Mar. 7, 2021. 4 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_001075493.1. Schiaffella et al., Jun. 24, 2018. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_001157741.1. Zeng et al., Sep. 17, 2018. 3 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_001157742.1. Zeng et al., Oct. 21, 2018. 3 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_001243439.1. Lee et al., Jul. 26, 2021. 4 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_033040.2. Liu et al., Jun. 23, 2018. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_076161.2. Wade et al., Jun. 20, 2021. 4 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_358988.1. Hoskins et al., Jan. 11, 2017. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_391970.1. Borriss et al., Feb. 12, 2021. 3 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_472073.1. Glaser et al., Jun. 27, 2013. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_628093.1. Hsiao et al., Aug. 3, 2016. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_955579.1. Chen et al., Aug. 13, 2018. 5 pages.
GENBANK Submission; NIH/NCBI, Accession No. NP_996816.2. Fu et al., Sep. 22, 2019. 9 pages.
GENBANK Submission; NIH/NCBI, Accession No. P42212. Prasher et al., Mar. 19, 2014. 7 pages.
GENBANK Submission; NIH/NCBI, Accession No. QBJ66766. Duan et al. Aug. 12, 2020. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. RFF81513.1. Zhou et al., Aug. 21, 2018. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. SNX31424.1. Weckx, S., Feb. 16, 2018. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. TGH57013. Xu et al., Apr. 9, 2019. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. WP_002989955.1. No Author Listed, May 6, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_010922251.1. No Author Listed, May 15, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_011054416.1. No Author Listed, May 15, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_011284745.1. No Author Listed, May 16, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_011285506.1. No Author Listed, May 16, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_011527619.1. No Author Listed, May 16, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_012560673.1. No Author Listed, May 17, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_014407541.1. No Author Listed, May 18, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_016631044.1. Haft et al., Sep. 22, 2020. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_020905136.1. No Author Listed, Jul. 25, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_023080005.1. No Author Listed, Oct. 27, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_023610282.1. No Author Listed, Nov. 27, 2013. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_030125963.1. No Author Listed, Jul. 9, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_030126706.1. No Author Listed, Jul. 9, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_031386437.1. No Author Listed., Sep. 23, 2019. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_031488318.1. No Author Listed., Aug. 5, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_031589969.1. Haft et al., Oct. 9, 2019. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. WP_032460140.1. No Author Listed, Oct. 4, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_032461047.1. No Author Listed, Oct. 4, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_032462016.1. Haft et al., Oct. 4, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_032462936.1. No Author Listed, Oct. 4, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_032464890.1. No Author Listed, Oct. 4, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_038431314.1. No Author Listed, Dec. 26, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_038432938.1. No Author Listed, Dec. 26, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_038434062.1. No Author Listed, Dec. 26, 2014. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_044924278.1. Haft et al., Oct. 9, 2019. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. WP_047338501.1. Haft et al., Oct. 9, 2019. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. WP_048327215.1. No Author Listed, Jun. 26, 2015. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_049519324.1. No Author Listed, Jul. 20, 2015. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_060798984.1. Haft et al., Oct. 9, 2019. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. WP_062913273.1. Haft et al., Oct. 9, 2019, 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. WP_072754838. No Author Listed., Sep. 23, 2019. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_095142515.1. No Author Listed., Sep. 23, 2019. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_118538418.1. No Author Listed., Oct. 13, 2019. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_119223642.1. No Author Listed., Oct. 13, 2019. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_119227726.1. No Author Listed., Oct. 13, 2019. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_119623382.1. No Author Listed., Oct. 13, 2019. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_132221894.1. No Author Listed., Sep. 23, 2019. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. WP_133478044.1. Haft et al., Oct. 9, 2019. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. XP_003314669.1. No Author Listed, Mar. 20, 2018. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. XP_026671085.1. No Author Listed, Oct. 17, 2018. 1 page.
GENBANK Submission; NIH/NCBI, Accession No. YP_002004532.1. Villegas et al., Oct. 11, 2021. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. YP_002342100.1. Bernardini et al., Jun. 10, 2013. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. YP_002344900.1. Gundogdu et al., Mar. 19, 2014. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. YP_006589943.1. Lynch et al., Oct. 15, 2021. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. YP_009137104.1. Davison, Aug. 13, 2018. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. YP_009283008.1. Bernardini et al., Sep. 23, 2016. 2 pages.
GENBANK Submission; NIH/NCBI, Accession No. YP_820832.1. Makarova et al., Aug. 27, 2013. 2 pages.
George et al., Adenosine deaminases acting on RNA, RNA editing, and interferon action. J Interferon Cytokine Res. Jan. 2011;31(1):99-117. doi: 10.1089/jir.2010.0097. Epub Dec. 23, 2010. PMID: 21182352; PMCID: PMC3034097.
Gerard et al., Influence on stability in Escherichia coli of the carboxy-terminal structure of cloned Moloney murine leukemia virus reverse transcriptase. DNA. Aug. 1986;5(4):271-9. doi: 10.1089/dna.1986.5.271.
Gerard et al., Purification and characterization of the DNA polymerase and RNase H activities in Moloney murine sarcoma-leukemia virus. J Virol. Apr. 1975;15(4):785-97. doi: 10.1128/JVI.15.4.785-797.1975.
Gerard et al., The role of template-primer in protection of reverse transcriptase from thermal inactivation. Nucleic Acids Res. Jul. 15, 2002;30(14):3118-29. doi: 10.1093/nar/gkf417.
Gerber et al., An adenosine deaminase that generates inosine at the wobble position of tRNAs. Science. Nov. 5, 1999;286(5442):1146-9. doi: 10.1126/science.286.5442.1146.
Gerber et al., RNA editing by base deamination: more enzymes, more targets, new mysteries. Trends Biochem Sci. Jun. 2001;26(6):376-84.
Gersbach et al., Directed evolution of recombinase specificity by split gene reassembly. Nucleic Acids Res. Jul. 2010;38(12):4198-206. doi: 10.1093/nar/gkq125. Epub Mar. 1, 2010.
Gersbach et al., Targeted plasmid integration into the human genome by an engineered zinc-finger recombinase. Nucleic Acids Res. Sep. 1, 2011;39(17):7868-78. doi: 10.1093/nar/gkr421. Epub Jun. 7, 2011.
Gete et al., Mechanisms of angiogenic incompetence in Hutchinson-Gilford progeria via downregulation of endothelial Nos. Aging Cell. Jul. 2021;20(7):e13388. doi: 10.1111/acel.13388. Epub Jun. 4, 2021.
Ghahfarokhi et al., Blastocyst Formation Rate and Transgene Expression are Associated with Gene Insertion into Safe and Non-Safe Harbors in the Cattle Genome. Sci Rep. Nov. 13, 2017;7(1):15432. doi: 10.1038/s41598-017-15648-3.
Gibson et al., Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat Methods. May 2009;6(5):343-5. doi: 10.1038/nmeth.1318. Epub Apr. 12, 2009.
Gil, Position-dependent sequence elements downstream of AAUAAA are required for efficient rabbit beta-globin mRNA 3′ end formation. Cell. May 8, 1987;49(3):399-406. doi: 10.1016/0092-8674(87)90292-3.
Gilbert et al., CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell. 2013 154(2):442-51.
Gilleron et al., Image-based analysis of lipid nanoparticle-mediated siRNA delivery, intracellular trafficking and endosomal escape. Nat Biotechnol. Jul. 2013;31(7):638-46. doi: 10.1038/nbt.2612. Epub Jun. 23, 2013.
Glasgow et al.,DNA-binding properties of the Hin recombinase. J Biol Chem. Jun. 15, 1989;264(17):10072-82.
Glassner et al., Generation of a strong mutator phenotype in yeast by imbalanced base excision repair. Proc Natl Acad Sci U S A. Aug. 18, 1998;95(17):9997-10002.
Goldberg et al., Epigenetics: a landscape takes shape. Cell. Feb. 23, 2007;128(4):635-8. doi: 10.1016/j.cell.2007.02.006.
Goldberg et al., Loss-of-function mutations in the Nav1.7 gene underlie congenital indifference to pain in multiple human populations. Clin Genet. Apr. 2007;71(4):311-9. doi: 10.1111/j.1399-0004.2007.00790.x.
Gong et al., Active DNA demethylation by oxidation and repair. Cell Res. Dec. 2011;21(12):1649-51. doi: 10.1038/cr.2011.140. Epub Aug. 23, 2011.
Gonzalez et al., An iCRISPR platform for rapid, multiplexable, and inducible genome editing in human pluripotent stem cells. Cell Stem Cell. Aug. 7, 2014;15(2):215-26. doi: 10.1016/j.stem.2014.05.018. Epub Jun. 12, 2014.
Goodnough et al., Development of a delivery vehicle for intracellular transport of botulinum neurotoxin antagonists. FEBS Lett. Feb. 27, 2002;513(2-3):163-8.
Gordley et al., Synthesis of programmable integrases. Proc Natl Acad Sci U S A. Mar. 31, 2009;106(13):5053-8. doi: 10.1073/pnas.0812502106. Epub Mar. 12, 2009.
Gordley et al., Evolution of programmable zinc finger-recombinases with activity in human cells. J Mol Biol. Mar. 30, 2007;367(3):802-13. Epub Jan. 12, 2007.
Gou et al., Designing single guide RNA for CIRSPR-Cas9 base editor by deep learning. Peer reviewed Thesis/Dissertation. UCLA Electronic Theses and Dissertations. Jan. 1, 2019. Retrieved from the Internet via https://escholarship.org/uc/item/7vf9z54t. Last accessed on Apr. 29, 2021.
Grainge et al., The integrase family of recombinase: organization and function of the active site. Mol Microbiol. Aug. 1999;33(3):449-56.
Grati et al., Localization of PDZD7 to the stereocilia ankle-link associates this scaffolding protein with the Usher syndrome protein network. J Neurosci. Oct. 10, 2012;32(41):14288-93. doi: 10.1523/JNEUROSCI.3071-12.2012.
Gregory et al., Integration site for Streptomyces phage phiBT1 and development of site-specific integrating vectors. J Bacteriol. Sep. 2003;185(17):5320-3. doi: 10.1128/jb.185.17.5320-5323.2003.
Griffiths, Endogenous retroviruses in the human genome sequence. Genome Biol. 2001;2(6):REVIEWS1017. doi: 10.1186/GB-2001-2-6-reviews1017. Epub Jun. 5, 2001.
Grindley et al., Mechanisms of site-specific recombination. Annu Rev Biochem. 2006;75:567-605. doi: 10.1146/annurev.biochem.73.011303.073908.
Grishok et al., Genes and Mechanisms Related to RNA Interference Regulate Expression of the Small Temporal RNAs that Control C. elegans Developmental Timing. Jul. 13, 2001:106(1):P23-4.
Groher et al., Synthetic riboswitches—A tool comes of age. Biochim Biophys Acta. Oct. 2014; 1839(10):964-973. doi: 10.1016/j.bbagrm.2014.05.005. Epub May 17, 2014.
Groth et al., Construction of transgenic Drosophila by using the site-specific integrase from phage phiC31. Genetics. Apr. 2004; 166(4):1775-82. doi: 10.1534/genetics.166.4.1775.
Groth et al., Phage integrases: biology and applications. J Mol Biol. Jan. 16, 2004;335(3):667-78.
Gruber et al., Strategies for measuring evolutionary conservation of RNA secondary structures. BMC Bioinformatics. Feb. 26, 2008;9:122. doi: 10.1186/1471-2105-9-122.
Gruber et al., The Vienna RNA websuite. Nucleic Acids Res. Jul. 1, 2008;36(Web Server issue): W70-4. doi: 10.1093/nar/gkn188. Epub Apr. 19, 2008.
Grunebaum et al., Recent advances in understanding and managing adenosine deaminase and purine nucleoside phosphorylase deficiencies. Curr Opin Allergy Clin Immunol. Dec. 2013;13(6):630-8. doi: 10.1097/ACI.0000000000000006.
GRÜNEWALD et al., Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature. May 2019;569(7756):433-437. doi: 10.1038/s41586-019-1161-z. Epub Apr. 17, 2019.
Guedon et al., Current gene therapy using viral vectors for chronic pain. Mol Pain. May 13, 2015;11:27. doi: 10.1186/s12990-015-0018-1.
Guilinger et al., Broad specificity profiling of TALENs results in engineered nucleases with improved DNA-cleavage specificity. Nat Methods. Apr. 2014; 11(4):429-35. doi: 10.1038/nmeth.2845. Epub Feb. 16, 2014.
Guilinger et al., Fusion of catalytically inactive Cas9 to FokI nuclease improves the specificity of genome modification. Nat Biotechnol. Jun. 2014;32(6):577-82. doi: 10.1038/nbt.2909. Epub Apr. 25, 2014.
Gumulya et al., Exploring the past and the future of protein evolution with ancestral sequence reconstruction: the ‘retro’ approach to protein engineering. Biochem J. Jan. 1, 2017;474(1):1-19. doi: 10.1042/BCJ20160507.
Guo et al., Evolution of Tetrahymena ribozyme mutants with increased structural stability. Nat Struct Biol. Nov. 2002;9(11):855-61. doi: 10.1038/nsb850.
Guo et al., Facile functionalization of FK506 for biological studies by the thiol-ene ‘click’ reaction. RSC Advances. 2014;22:11400-3.
Guo et al., Protein tolerance to random amino acid change. Proc Natl Acad Sci U S A. Jun. 22, 2004;101(25):9205-10. Epub Jun. 14, 2004.
Guo et al., Structure of Cre recombinase complexed with DNA in a site-specific recombination synapse. Nature. Sep. 4, 1997;389(6646):40-6.
Gupta et al., Cross-talk between cognate and noncognate RpoE sigma factors and Zn(2+)-binding anti-sigma factors regulates photooxidative stress response in Azospirillum brasilense. Antioxid Redox Signal. Jan. 1, 2014;20(1):42-59. doi: 10.1089/ars.2013.5314. Epub Jul. 19, 2013.
Gupta et al., Sequences in attB that affect the ability of phiC31 integrase to synapse and to activate DNA cleavage. Nucleic Acids Res. 2007;35(10):3407-19. doi: 10.1093/nar/gkm206. Epub May 3, 2007.
Gutschner et al., Post-translational Regulation of Cas9 during G1 Enhances Homology-Directed Repair. Cell Rep. Feb. 16, 2016;14(6):1555-1566. doi: 10.1016/j.celrep.2016.01.019. Epub Feb. 4, 2016.
Guzman et al., Tight regulation, modulation, and high-level expression by vectors containing the arabinose PBAD promoter. J Bacteriol. 1995;177(14):4121-4130.
Haapaniemi et al., CRISPR-Cas9 genome editing induces a p53-mediated DNA damage response. Nat Med. Jul. 2018;24(7):927-930. doi: 10.1038/s41591-018-0049-z. Epub Jun. 11, 2018.
Haddada et al., Gene therapy using adenovirus vectors. Curr Top Microbiol Immunol. 1995;199 ( Pt 3):297-306. doi: 10.1007/978-3-642-79586-2_14.
Haeussler et al., Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR. Genome Biol. Jul. 5, 2016;17(1):148. doi: 10.1186/s13059-016-1012-2.
Hagen et al., A high rate of polymerization during synthesis of mouse mammary tumor virus DNA alleviates hypermutation by APOBEC3 proteins. PLoS Pathog. Feb. 15, 2019;15(2):e1007533. doi: 10.1371/journal.ppat.1007533.
Halbert et al., Repeat transduction in the mouse lung by using adeno-associated virus vectors with different serotypes. J Virol. Feb. 2000;74(3): 1524-32. doi: 10.1128/jvi.74.3.1524-1532.2000.
Hale et al., RNA-guided RNA cleavage by a CRISPR RNA-Cas protein complex. Cell. Nov. 25, 2009;139(5):945-56. doi: 10.1016/j.cell.2009.07.040.
Halmai et al., Targeted CRIPSR/dCas9-mediated reactivation of epigenetically silenced genes suggests limited escape from the inactive X chromosome. 2nd Intl Conf on Epigenetics and Bioengineering. Oct. 4, 2018; Retrieved from the Internet: https://aiche.confex.com/aiche/epibiol8/webprogram/paper544785.html. Retrieved Jun. 29, 2020.
Halperin et al., CRISPR-guided DNA polymerases enable diversification of all nucleotides in a tunable window. Nature. Aug. 2018;560(7717):248-252. doi: 10.1038/s41586-018-0384-8. Epub Aug. 1, 2018.
Halvas et al., Role of murine leukemia virus reverse transcriptase deoxyribonucleoside triphosphate-binding site in retroviral replication and in vivo fidelity. J Virol. Nov. 2000;74(22): 10349-58. doi: 10.1128/jvi.74.22.10349-10358.2000.
Hamano-Takaku et al., A mutant Escherichia coli tyrosyl-tRNA synthetase utilizes the unnatural amino acid azatyrosine more efficiently than tyrosine. J Biol Chem. Dec. 22, 2000;275(51):40324-8.
Han, New CRISPR/Cas9-based Tech Edits Single Nucleotides Without Breaking DNA. Genome Web, Apr. 20, 2016. https://www.genomeweb.com/gene-silencinggene-editing/new-crisprcas9-based-tech-edits-single-nucleotides-without-breaking-dna.
Handa et al., Template-assisted synthesis of adenine-mutagenized cDNA by a retroelement protein complex. Nucleic Acids Res. Oct. 12, 2018;46(18):9711-9725. doi: 10.1093/nar/gky620.
Hanna et al., Massively parallel assessment of human variants with base editor screens. Cell. Feb. 18, 2021;184(4):1064-1080.e20. doi: 10.1016/j.cell.2021.01.012.
Hanson et al., Codon optimality, bias and usage in translation and mRNA decay. Nat Rev Mol Cell Biol. Jan. 2018;19(1):20-30. doi: 10.1038/nrm.2017.91. Epub Oct. 11, 2017.
Hardt et al.,Missense variants in hMLH1 identified in patients from the German HNPCC consortium and functional studies. Fam Cancer. Jun. 2011; 10(2):273-84. doi: 10.1007/s10689-011-9431-4.
Harms et al., Evolutionary biochemistry: revealing the historical and physical causes of protein properties. Nat Rev Genet. Aug. 2013;14(8):559-71. doi: 10.1038/nrg3540.
Harmsen et al., DNA mismatch repair and oligonucleotide end-protection promote base-pair substitution distal from a CRISPR/Cas9-induced DNA break. Nucleic Acids Res. Apr. 6, 2018;46(6):2945-2955. doi: 10.1093/nar/gky076.
Harrington et al., A thermostable Cas9 with increased lifetime in human plasma. Nat Commun. Nov. 10, 2017;8(1):1424. doi: 10.1038/s41467-017-01408-4. Posted May 16, 2017 as bioRxiv preprint. Doi.org/10.1101/138867.
Harrington et al., Programmed DNA destruction by miniature CRISPR-Cas14 enzymes. Science. Nov. 16, 2018;362(6416):839-842. doi: 10.1126/science.aav4294. Epub Oct. 18, 2018.
Harris et al., RNA Editing Enzyme APOBEC1 and Some of Its Homologs Can Act as DNA Mutators. Mol Cell. Nov. 2002; 10(5):1247-53.
Hart et al., High-Resolution CRISPR Screens Reveal Fitness Genes and Genotype-Specific Cancer Liabilities. Cell. Dec. 3, 2015;163(6):1515-26. doi: 10.1016/j.cell.2015.11.015. Epub Nov. 25, 2015.
Hartung et al., Correction of metabolic, craniofacial, and neurologic abnormalities in MPS I mice treated at birth with adeno-associated virus vector transducing the human alpha-L-iduronidase gene. Mol Ther. Jun. 2004;9(6):866-75.
Hartung et al., Cre mutants with altered DNA binding properties. J Biol Chem. Sep. 4, 1998;273(36):22884-91.
Hasadsri et al., Functional protein delivery into neurons using polymeric nanoparticles. J Biol Chem. Mar. 13, 2009;284(11):6972-81. doi: 10.1074/jbc.M805956200. Epub Jan. 7, 2009.
Hasegawa et al., Spontaneous mutagenesis associated with nucleotide excision repair in Escherichia coli. Genes Cells. May 2008;13(5):459-69. doi: 10.1111/j.1365-2443.2008.01185.x.
Hayes et al., Stop codons preceded by rare arginine codons are efficient determinants of SsrA tagging in Escherichia coli. Proc Natl Acad Sci U S A. Mar. 19, 2002;99(6):3440-5. Epub Mar. 12, 2002.
Hector et al., CDKL5 variants: Improving our understanding of a rare neurologic disorder. Neurol Genet. Dec. 15, 2017;3(6):e200. doi: 10.1212/NXG.0000000000000200.
Heidenreich et al., Non-homologous end joining as an important mutagenic process in cell cycle-arrested cells. EMBO J. May 1, 2003;22(9):2274-83. doi: 10.1093/emboj/cdg203.
Held et al., In vivo correction of murine hereditary tyrosinemia type I by phiC31 integrase-mediated gene delivery. Mol Ther. Mar. 2005;11(3):399-408. doi: 10.1016/j.ymthe.2004.11.001.
Heller et al., Replisome assembly and the direct restart of stalled replication forks. Nat Rev Mol Cell Biol. Dec. 2006;7(12):932-43. Epub Nov. 8, 2006.
Hendel et al., Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells. Nat Biotechnol. Sep. 2015;33(9):985-989. doi: 10.1038/nbt.3290. Epub Jun. 29, 2015. Author Manuscript. 14 pages.
Hendricks et al., The S. cerevisiae Mag1 3-methyladenine DNA glycosylase modulates susceptibility to homologous recombination. DNA Repair (Amst). 2002;1(8):645-659.
Hermonat et al., Use of adeno-associated virus as a mammalian DNA cloning vector: transduction of neomycin resistance into mammalian tissue culture cells. Proc Natl Acad Sci U S A. Oct. 1984;81(20):6466-70. doi: 10.1073/pnas.81.20.6466.
Herschhorn et al., Retroviral reverse transcriptases. Cell Mol Life Sci. Aug. 2010;67(16):2717-47. doi: 10.1007/s00018-010-0346-2. Epub Apr. 1, 2010.
Herzig et al., A Novel Leu92 Mutant of HIV-1 Reverse Transcriptase with a Selective Deficiency in Strand Transfer Causes a Loss of Viral Replication. J Virol. Aug. 2015;89(16):8119-29. doi: 10.1128/JVI.00809-15. Epub May 20, 2015.
Hess et al., Directed evolution using dCas9-targeted somatic hypermutation in mammalian cells. Nat Methods. Dec. 2016; 13(12):1036-1042. doi: 10.1038/nmeth.4038. Epub Oct. 31, 2016.
Heyer et al., Regulation of homologous recombination in eukaryotes. Annu Rev Genet. 2010;44:113-39. doi: 10.1146/annurev-genet-051710-150955. Author Manuscript. 33 pages.
Hickford et al., Antitumour polyether macrolides: four new halichondrins from the New Zealand deep-water marine sponge Lissodendoryx sp. Bioorg Med Chem. Mar. 15, 2009;17(6):2199-203. doi: 10.1016/j.bmc.2008.10.093. Epub Nov. 19, 2008.
Hida et al., Directed evolution for drug and nucleic acid; delivery. Adv Drug Deliv Rev. Dec. 22, 2007;59(15):1562-78. Epub Aug. 28, 2007.; Review.
Higgs et al., Genetic complexity in sickle cell disease. Proc Natl Acad Sci U S A. Aug. 19, 2008;105(33):11595-6. doi: 10.1073/pnas.0806633105. Epub Aug. 11, 2008.
Hilbers et al., New developments in structure determination of pseudoknots. Biopolymers. 1998;48(2-3):137-53. doi: 10.1002/(SICI)1097-0282(1998)48:2<137::AID-BIP4>3.0.CO;2-H.
Hill et al., Functional analysis of conserved histidines in ADP-glucose pyrophosphorylase from Escherichia coli.Biochem Biophys Res Commun. Mar. 17, 1998;244(2):573-7.
Hille et al., The Biology of CRISPR-Cas: Backward and Forward. Cell. Mar. 8, 2018;172(6):1239-1259. doi: 10.1016/j.cell.2017.11.032.
Hilton et al., Enabling functional genomics with genome engineering. Genome Res. Oct. 2015;25(10):1442-55. doi: 10.1101/gr.190124.115.
Hirano et al., Site-specific recombinases as tools for heterologous gene integration. Appl Microbiol Biotechnol. Oct. 2011;92(2):227-39. doi: 10.1007/s00253-011-3519-5. Epub Aug. 7, 2011. Review.
Hirano et al., Structural Basis for the Altered PAM Specificities of Engineered CRISPR-Cas9. Mol Cell. Mar. 17, 2016;61(6):886-94. doi: 10.1016/j.molcel.2016.02.018.
Hoang et al., UFBoot2: Improving the Ultrafast Bootstrap Approximation. Mol Biol Evol. Feb. 1, 2018;35(2):518-522. doi: 10.1093/molbev/msx281.
Hockemeyer et al., Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nat Biotechnol. Sep. 2009;27(9):851-7. doi: 10.1038/nbt.1562. Epub Aug. 13, 2009.
Hockemeyer et al., Genetic engineering of human pluripotent cells using TALE nucleases. Nat Biotechnol. Jul. 7, 2011;29(8):731-4. doi: 10.1038/nbt.1927.
Hoernes et al., Translating the epitranscriptome. Wiley Interdiscip Rev RNA. Jan. 2017;8(1):e1375. doi: 10.1002/wrna.1375. Epub Jun. 27, 2016.
Hoess et al., DNA specificity of the Cre recombinase resides in the 25 kDa carboxyl domain of the protein. J Mol Biol. Dec. 20, 1990;216(4):873-82. doi: 10.1016/S0022-2836(99)80007-2.
Holden et al., Crystal structure of the anti-viral APOBEC3G catalytic domain and functional implications. Nature. Nov. 6, 2008;456(7218):121-4. doi: 10.1038/nature07357. Epub Oct. 12, 2008.
Hollis et al., Phage integrases for the construction and manipulation of transgenic mammals. Reprod Biol Endocrinol. Nov. 7, 2003;1:79. doi: 10.1186/1477-7827-1-79.
Holsinger et al., Signal transduction in T lymphocytes using a conditional allele of Sos. Proc Natl Acad Sci U S A. Oct. 10, 1995;92(21):9810-4. doi: 10.1073/pnas.92.21.9810.
Holt et al., Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo. Nat Biotechnol. Aug. 2010;28(8):839-47. doi: 10.1038/nbt.1663. Epub Jul. 2, 2010.
Hondares et al., Peroxisome Proliferator-activated Receptor α (PPARα) Induces PPARγCoactivator 1α (PGC-1α) Gene Expression and Contributes to Thermogenic Activation of Brown Fat. J Biol. Chem Oct. 2011; 286(50):43112-22. doi: 10.1074/jbc.M111.252775.
Hoogenboom et al., Natural and designer binding sites made by phage display technology. Immunol Today. Aug. 2000;21(8):371-8.
Horvath et al., CRISPR/Cas, the immune system of bacteria and archaea. Science. Jan. 8, 2010;327(5962):167-70. doi: 10.1126/science.1179555.
Horvath et al., Diversity, Activity, and Evolution of CRISPR Loci in Streptococcus thermophilus. J Bacteriol. Feb. 2008;190(4):1401-12. doi: 10.1128/JB.01415-07. Epub Dec. 7, 2007.
Hotta et al., [Neurotropic viruses—classification, structure and characteristics]. Nihon Rinsho. Apr. 1997;55(4):777-82. Japanese.
Hou et al., Efficient genome engineering in human pluripotent stem cells using Cas9 from Neisseria meningitidis. Proc Natl Acad Sci U S A. Sep. 24, 2013;110(39):15644-9. doi: 10.1073/pnas.1313587110. Epub Aug. 12, 2013.
Houdebine, The methods to generate transgenic animals and to control transgene expression. J Biotechnol. Sep. 25, 2002;98(2-3):145-60.
Housden et al., Identification of potential drug targets for tuberous sclerosis complex by synthetic screens combining CRISPR-based knockouts with RNAi. Sci Signal. Sep. 8, 2015;8(393):rs9. doi: 10.1126/scisignal.aab3729.
Howard et al., Intracerebral drug delivery in rats with lesion-induced memory deficits. J Neurosurg. Jul. 1989;71(1):105-12.
Hower et al., Shape-based peak identification for ChIP-Seq. BMC Bioinformatics. Jan. 12, 2011;12:15. doi: 10.1186/1471-2105-12-15.
Hsu et al., DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol. Sep. 2013;31(9):827-32. doi: 10.1038/nbt.2647. Epub Jul. 21, 2013.
Hsu et al., DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol. Sep. 2013;31(9):827-32. doi: 10.1038/nbt.2647. Epub Jul. 21, 2013. Supplementary Information. 27 pages.
Hsu et al., PrimeDesign software for rapid and simplified design of prime editing guide RNAs. Nat Commun. Feb. 15, 2021;12(1):1034. doi: 10.1038/s41467-021-21337-7.
Hu et al., Chemical Biology Approaches to Genome Editing: Understanding, Controlling, and Delivering Programmable Nucleases. Cell Chem Biol. Jan. 21, 2016;23(1):57-73. doi: 10.1016/j.chembiol.2015.12.009.
Hu et al., Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature. Apr. 5, 2018;556(7699):57-63 and Extended/Supplementary Data. doi: 10.1038/nature26155. Epub Feb. 28, 2018. 21 pages.
Hu et al., Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature. Apr. 5, 2018;556(7699):57-63. doi: 10.1038/nature26155. Epub Feb. 28, 2018.
Hua et al., Expanding the base editing scope in rice by using Cas9 variants. Plant Biotechnol J. Feb. 2019;17(2):499-504. doi: 10.1111/pbi.12993. Epub Oct. 5, 2018.
Hua et al., Precise A⋅T to G⋅C Base Editing in the Rice Genome. Mol Plant. Apr. 2, 2018;11(4):627-630. doi: 10.1016/j.molp.2018.02.007. Epub Feb. 21, 2018.
Huang et al., Circularly permuted and PAM-modified Cas9 variants broaden the targeting scope of base editors. Nat Biotechnol. Jun. 2019;37(6):626-631. doi: 10.1038/s41587-019-0134-y. Epub May 20, 2019. Including Supplementary Information.
Huang et al., Gain-of-function mutations in sodium channel Na(v)1.9 in painful neuropathy. Brain. Jun. 2014; 137(Pt 6):1627-42. doi: 10.1093/brain/awu079. Epub Apr. 27, 2014.
Huang et al., Heritable gene targeting in zebrafish using customized TALENs. Nat Biotechnol. Aug. 5, 2011;29(8):699-700. doi: 10.1038/nbt.1939.
Huang et al., Precision genome editing using cytosine and adenine base editors in mammalian cells. Nat Protoc. Feb. 2021;16(2):1089-1128. doi: 10.1038/s41596-020-00450-9. Epub Jan. 18, 2021.
Huggins et al., Flap endonuclease 1 efficiently cleaves base excision repair and DNA replication intermediates assembled into nucleosomes. Mol Cell. Nov. 2002;10(5):1201-11. doi: 10.1016/s1097-2765(02)00736-0.
Humbert et al., Targeted gene therapies: tools, applications, optimization. Crit Rev Biochem Mol Biol. May-Jun. 2012;47(3):264-81. doi: 10.3109/10409238.2012.658112.
Hung et al., Protein localization in disease and therapy. J Cell Sci. Oct. 15, 2011;124(Pt 20):3381-92. doi: 10.1242/jcs.089110.
Hurt et al., Highly specific zinc finger proteins obtained by directed domain shuffling and cell-based selection. Proc Natl Acad Sci U S A. Oct. 14, 2003; 100(21):12271-6. Epub Oct. 3, 2003.
Husimi, Selection and evolution of bacteriophages in cellstat. Adv Biophys. ; 1989;25:1-43. Review.
Hwang et al., Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat Biotechnol. Mar. 2013;31(3):227-9. doi: 10.1038/nbt.2501. Epub Jan. 29, 2013.
Hwang et al., Efficient In Vivo Genome Editing Using RNA-Guided Nucleases. Nat Biotechnol. Mar. 2013; 31(3): 227-229. doi: 10.1038/nbt.2501. Epub Jan. 29, 2013.
Hwang et al., Web-based design and analysis tools for CRISPR base editing. BMC Bioinformatics. Dec. 27, 2018;19(1):542. doi: 10.1186/s12859-018-2585-4.
Ibba et al., Relaxing the substrate specificity of an aminoacyl-tRNA; synthetase allows in vitro and in vivo synthesis of proteins containing unnatural amino acids. FEBS Lett. May 1, 1995;364(3):272-5.
Ibba et al., Substrate specificity is determined by amino acid binding pocket size in Escherichia coli phenylalanyl-tRNA synthetase. Biochemistry. Jun. 14, 1994;33(23):7107-12.
Ihry et al., p53 inhibits CRISPR-Cas9 engineering in human pluripotent stem cells. Nat Med. Jul. 2018;24(7):939-946. doi: 10.1038/s41591-018-0050-6. Epub Jun. 11, 2018.
Iida et al., A site-specific, conservative recombination system carried by bacteriophage P1. Mapping the recombinase gene cin and the cross-over sites cix for the inversion of the C segment. EMBO J. 1982;1(11):1445-53.
Iida et al., The Min DNA inversion enzyme of plasmid p15B of Escherichia coli 15T-: a new member of the Din family of site-specific recombinases. Mol Microbiol. Jun. 1990;4(6):991-7. doi: 10.1111/j.1365-2958.1990.tb00671.x.
Ikediobi et al., Mutation analysis of 24 known cancer genes in the NCI-60 cell line set. Mol Cancer Ther. Nov. 2006;5(11):2606-12. Epub Nov. 6, 2006.
Imanishi et al., Detection of N6-methyladenosine based on the methyl-sensitivity of MazF RNA endonuclease. Chem Commun (Camb). Nov. 30, 2017;53(96):12930-12933. doi: 10.1039/c7cc07699a.
Imburgio et al., Studies of promoter recognition and start site selection by T7 RNA polymerase using a comprehensive collection of promoter variants. Biochemistry. Aug. 29, 2000;39(34):10419-30.
Ingram, A specific chemical difference between the globins of normal human and sickle-cell anaemia haemoglobin. Nature. Oct. 13, 1956;178(4537):792-4. doi: 10.1038/178792a0.
Irion et al., Identification and targeting of the ROSA26 locus in human embryonic stem cells. Nat Biotechnol. Dec. 2007;25(12):1477-82. doi: 10.1038/nbt1362. Epub Nov. 25, 2007.
Irrthum et al., Congenital hereditary lymphedema caused by a mutation that inactivates VEGFR3 tyrosine kinase. Am J Hum Genet. Aug. 2000;67(2):295-301. Epub Jun. 9, 2000.
Isaacs et al., Engineered riboregulators enable post-transcriptional control of gene expression. Nat Biotechnol. Jul. 2004;22(7):841-7. doi: 10.1038/nbt986. Epub Jun. 20, 2004.
Ishino et al., Nucleotide sequence of the iap gene, responsible for alkaline phosphatase isozyme conversion in Escherichia coli, and identification of the gene product. J Bacteriol. Dec. 1987;169(12):5429-33.
Ishizuka et al., Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature. Jan. 2019;565(7737):43-48. doi: 10.1038/s41586-018-0768-9. Epub Dec. 17, 2018.
Iwai et al., Circular beta-lactamase: stability enhancement by cyclizing the backbone. FEBS Lett. Oct. 8, 1999;459(2):166-72. doi: 10.1016/s0014-5793(99)01220-x.
Iwai et al., Highly efficient protein trans-splicing by a naturally split DnaE intein from Nostoc punctiforme. FEBS Lett. Mar. 20, 2006;580(7):1853-8. doi: 10.1016/j.febslet.2006.02.045. Epub Feb. 24, 2006.
Iyama et al., DNA repair mechanisms in dividing and non-dividing cells. DNA Repair (Amst). Aug. 2013;12(8):620-36. doi: 10.1016/j.dnarep.2013.04.015. Epub May 16, 2013.
Jaffrey et al., Emerging links between m6A and misregulated mRNA methylation in cancer. Genome Med. Jan. 12, 2017;9(1):2. doi: 10.1186/s13073-016-0395-8.
Jakimo et al., A Cas9 with Complete PAM Recognition for Adenine Dinucleotides. bioRxiv preprint. Sep. 27, 2018. doi.org/10.1101/429654. 29 pages.
Jamieson et al., Drug discovery with engineered zinc-finger proteins. Nat Rev Drug Discov. May 2003;2(5):361-8.
Jansen et al., Backbone and nucleobase contacts to glucosamine-6-phosphate in the glmS ribozyme. Nat Struct Mol Biol. Jun. 2006;13(6):517-23. Epub May 14, 2006.
Jansen et al., Identification of genes that are associated with DNA repeats in prokaryotes. Mol Microbiol. Mar. 2002;43(6):1565-75.
Jardine et al., HIV-1 Vaccines. Priming a broadly neutralizing antibody response to HIV-1 using a germline-targeting immunogen. Science. Jul. 10, 2015;349(6244):156-61. doi: 10.1126/science.aac5894. Epub Jun. 18, 2015.
Jasin et al., Repair of strand breaks by homologous recombination. Cold Spring Harb Perspect Biol. Nov. 1, 2013;5(11):a012740. doi: 10.1101/cshperspect.a012740.
Jeggo, DNA breakage and repair. Adv Genet. 1998;38:185-218. doi: 10.1016/s0065-2660(08)60144-3.
Jemielity et al., Novel “anti-reverse” cap analogs with superior translational properties. RNA. Sep. 2003;9(9):1108-22. doi: 10.1261/rna.5430403.
Jenkins et al., Comparison of a preQ1 riboswitch aptamer in metabolite-bound and free states with implications for gene regulation. J Biol Chem. Jul. 15, 2011;286(28):24626-37. doi: 10.1074/jbc.M111.230375. Epub May 18, 2011.
Jeong et al., Measurement of deoxyinosine adduct: Can it be a reliable tool to assess oxidative or nitrosative DNA damage? Toxicol Lett. Oct. 17, 2012;214(2):226-33. doi: 10.1016/j.toxlet.2012.08.013. Epub Aug. 23, 2012.
Jia et al., The MLH1 ATPase domain is needed for suppressing aberrant formation of interstitial telomeric sequences. DNA Repair (Amst). May 2018;65:20-25. doi: 10.1016/j.dnarep.2018.03.002. Epub Mar. 7, 2018.
Jiang et al., Chemical modifications of adenine base editor mRNA and guide RNA expand its application scope. Nat Commun. Apr. 24, 2020;11(1):1979. doi: 10.1038/s41467-020-15892-8.
Jiang et al., CRISPR-Cas9 Structures and Mechanisms. Annu Rev Biophys. May 22, 2017;46:505-529. doi: 10.1146/annurev-biophys-062215-010822. Epub Mar. 30, 2017.
Jiang et al., Prime editing efficiently generates W542L and S621I double mutations in two ALS genes of maize. bioRxiv preprint. Jul. 6, 2020. Retrieved from www.biorxiv.org. doi: 10.1101/2020.07.06.188896. 15 pages.
Jiang et al., RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nat Biotechnol. Mar. 2013;31(3):233-9. doi: 10.1038/nbt.2508. Epub Jan. 29, 2013.
Jiang et al., Structural Biology. A Cas9-guide RNA Complex Preorganized for Target DNA Recognition. Science. Jun. 26, 2015;348(6242):1477-81. doi: 10.1126/science.aab1452.
Jiang et al., Structures of a CRISPR-Cas9 R-loop complex primed for DNA cleavage. Science. Feb. 19, 2016;351(6275):867-71. doi: 10.1126/science.aad8282. Epub Jan. 14, 2016.
Jin et al., Cytosine, but not adenine, base editors induce genome-wide off-target mutations in rice. Science. Apr. 19, 2019;364(6437):292-295. doi: 10.1126/science.aaw7166. Epub Feb. 28, 2019.
Jinek et al., A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. Aug. 17, 2012;337(6096):816-21. doi: 10.1126/science.1225829. Epub Jun. 28, 2012.
Jinek et al., RNA-programmed genome editing in human cells. Elife. Jan. 29, 2013;2:e00471. doi: 10.7554/eLife.00471.
Jinek et al., Structures of Cas9 endonucleases reveal RNA-mediated conformational activation. Science. Mar. 14, 2014;343(6176):1247997. doi: 10.1126/science.1247997. Epub Feb. 6, 2014.
Jiricny, The multifaceted mismatch-repair system. Nat Rev Mol Cell Biol. May 2006;7(5):335-46. doi: 10.1038/nrm1907.
Johann et al., GLVR1, a receptor for gibbon ape leukemia virus, is homologous to a phosphate permease of Neurospora crassa and is expressed at high levels in the brain and thymus. J Virol. Mar. 1992;66(3):1635-40. doi: 10.1128/JVI.66.3.1635-1640.1992.
Johansson et al., RNA Recognition by the MS2 Phage Coat Protein. Seminars in Virology. 1997;8(3):176-85. https://doi.org/10.1006/smvy.1997.0120.
Johansson et al., Selenocysteine in proteins-properties and biotechnological use. Biochim Biophys Acta. Oct. 30, 2005;1726(1):1-13. Epub Jun. 1, 2005.
Johns et al., The promise and peril of continuous in vitro evolution. J Mol Evol. Aug. 2005;61(2):253-63. Epub Jun. 27, 2005.
Johnson et al., Trans insertion-splicing: ribozyme-catalyzed insertion of targeted sequences into RNAs. Biochemistry. Aug. 9, 2005;44(31):10702-10. doi: 10.1021/bi0504815.
Joho et al., Identification of a region of the bacteriophage T3 and T7 RNA polymerases that determines promoter specificity. J Mol Biol. Sep. 5, 1990;215(1):31-9.
Jore et al., Structural basis for CRISPR RNA-guided DNA recognition by Cascade. Nat Struct Mol Biol. May 2011;18(5):529-36. doi: 10.1038/nsmb.2019. Epub Apr. 3, 2011.
Joung et al., TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol. Jan. 2013;14(1):49-55. doi: 10.1038/nrm3486. Epub Nov. 21, 2012.
Joyce et al., Amplification, mutation and selection of catalytic RNA. Gene. Oct. 15, 1989;82(1):83-7. doi: 10.1016/0378-1119(89)90033-4.
Jusiak et al., Comparison of Integrases Identifies Bxb1-GA Mutant as the Most Efficient Site-Specific Integrase System in Mammalian Cells. ACS Synth Biol. Jan. 18, 2019;8(1):16-24. doi: 10.1021/acssynbio.8b00089. Epub Jan. 9, 2019.
Jyothy et al., Translocation Down syndrome. Indian J Med Sci. Mar. 2002;56(3):122-6.
Kacian et al., Purification of the DNA polymerase of avian myeloblastosis virus. Biochim Biophys Acta. Sep. 24, 1971;246(3):365-83. doi: 10.1016/0005-2787(71)90773-8.
Kaczmarczyk et al., Manipulating the Prion Protein Gene Sequence and Expression Levels with CRISPR/Cas9. PLoS One. Apr. 29, 2016;11(4):e0154604. doi: 10.1371/journal.pone.0154604.
Kadoch et al., Reversible disruption of mSWI/SNF (BAF) complexes by the SS18-SSX oncogenic fusion in synovial sarcoma. Cell. Mar. 28, 2013;153(1):71-85. doi: 10.1016/j.cell.2013.02.036.
Kahmann et al., G inversion in bacteriophage Mu DNA is stimulated by a site within the invertase gene and a host factor. Cell. Jul. 1985;41(3):771-80. doi: 10.1016/s0092- 8674(85)80058-1.
Kaiser et al., Gene therapy. Putting the fingers on gene repair. Science. Dec. 23, 2005;310(5756):1894-6.
Kakiyama et al., A peptide release system using a photo-cleavable linker in a cell array format for cell-toxicity analysis. Polymer J. Feb. 27, 2013;45:535-9.
Kalyaanamoorthy et al., ModelFinder: fast model selection for accurate phylogenetic estimates. Nat Methods. Jun. 2017; 14(6):587-589. doi: 10.1038/nmeth.4285. Epub May 8, 2017.
Kan et al., Mechanisms of precise genome editing using oligonucleotide donors. Genome Res. Jul. 2017;27(7):1099-1111. doi: 10.1101/gr.214775.116. Epub Mar. 29, 2017.
Kandavelou et al., Targeted manipulation of mammalian genomes using designed zinc finger nucleases. Biochem Biophys Res Commun. Oct. 9, 2009;388(1):56-61. doi: 10.1016/j.bbrc.2009.07.112. Epub Jul. 25, 2009.
Kang et al., Structural Insights into riboswitch control of the biosynthesis of queuosine, a modified nucleotide found in the anticodon of tRNA. Mol Cell. Mar. 27, 2009;33(6):784-90. doi: 10.1016/j.molcel.2009.02.019. Epub Mar. 12, 2009.
Kang et al., Precision genome engineering through adenine base editing in plants. Nat Plants. Jul. 2018;4(7):427-431. doi: 10.1038/s41477-018-0178-x. Epub Jun. 4, 2018. Erratum in: Nat Plants. Sep. 2018;4(9):730.
Kao et al., Cleavage specificity of Saccharomyces cerevisiae flap endonuclease 1 suggests a double-flap structure as the cellular substrate. J Biol Chem. Apr. 26, 2002;277(17):14379-89. doi: 10.1074/jbc.M110662200. Epub Feb. 1, 2002.
Kappel et al., Regulating gene expression in transgenic animals.Curr Opin Biotechnol. Oct. 1992;3(5):548-53.
Karimova et al., Discovery of Nigri/nox and Panto/pox site-specific recombinase systems facilitates advanced genome engineering. Sci Rep. Jul. 22, 2016;6:30130. doi: 10.1038/srep30130.
Karimova et al., Vika/vox, a novel efficient and specific Cre/loxP-like site-specific recombination system. Nucleic Acids Res. Jan. 2013;41(2):e37. doi: 10.1093/nar/gks1037. Epub Nov. 9, 2012.
Karpenshif et al., From yeast to mammals: recent advances in genetic control of homologous recombination. DNA Repair (Amst). Oct. 1, 2012;11(10):781-8. doi: 10.1016/j.dnarep.2012.07.001. Epub Aug. 11, 2012. Review.
Karpinsky et al., Directed evolution of a recombinase that excises the provirus of most HIV-1 primary isolates with high specificity. Nat Biotechnol. Apr. 2016;34(4):401-9. doi: 10.1038/nbt.3467. Epub Feb. 22, 2016.
Katafuchi et al., DNA polymerases involved in the incorporation of oxidized nucleotides into DNA: their efficiency and template base preference. Mutat Res. Nov. 28, 2010;703(1):24-31. doi: 10.1016/j.mrgentox.2010.06.004. Epub Jun. 11, 2010.
Kato et al., Improved purification and enzymatic properties of three forms of reverse transcriptase from avian myeloblastosis virus. J Virol Methods. Dec. 1984;9(4):325-39. doi: 10.1016/0166-0934(84)90058-2.
Katoh et al., MAFFT multiple sequence alignment software version 7: improvements in performance and usability. Mol Biol Evol. Apr. 2013;30(4):772-80. doi: 10.1093/molbev/mst010. Epub Jan. 16, 2013.
Kaufman et al., Translational efficiency of polycistronic mRNAs and their utilization to express heterologous genes in mammalian cells. EMBO J. Jan. 1987;6(1):187-93.
Kavli et al., Excision of cytosine and thymine from DNA by mutants of human uracil-DNA glycosylase. EMBO J. Jul. 1, 1996;15(13):3442-7.
Kawarasaki et al., Enhanced crossover SCRATCHY: construction and high-throughput screening of a combinatorial library containing multiple non-homologous crossovers. Nucleic Acids Res. Nov. 1, 2003;31(21):e126.
Kay et al., Viral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeutics. Nat Med. Jan. 2001;7(1):33-40.
Kaya et al., A bacterial Argonaute with noncanonical guide RNA specificity. Proc. Natl. Acad. Sci. USA Apr. 2016;113(15):4057-62.
Keijzers et al., Human exonuclease 1 (EXO1) activity characterization and its function on flap structures. Biosci Rep. Apr. 25, 2015;35(3):e00206. doi: 10.1042/BSR20150058.
Kellendonk et al., Regulation of Cre recombinase activity by the synthetic steroid RU 486. Nucleic Acids Res. Apr. 15, 1996;24(8):1404-11.
Kelman, PCNA: structure, functions and interactions. Oncogene. Feb. 13, 1997;14(6):629-40. doi: 10.1038/sj.onc.1200886.
Keravala et al., A diversity of serine phage integrases mediate site-specific recombination in mammalian cells. Mol Genet Genomics. Aug. 2006;276(2):135-46. doi: 10.1007/s00438-006-0129-5. Epub May 13, 2006.
Kessel et al., Murine developmental control genes. Science. Jul. 27, 1990;249(4967):374-9. doi: 10.1126/science.1974085.
Kessler et al., Gene delivery to skeletal muscle results in sustained expression and systemic delivery of a therapeutic protein. Proc Natl Acad Sci U S A. Nov. 26, 1996;93(24):14082-7. doi: 10.1073/pnas.93.24.14082.
Ketha et al., Application of bioinformatics-coupled experimental analysis reveals a new transport-competent nuclear localization signal in the nucleoprotein of Influenza A virus strain. BMC Cell Biol. Apr. 28, 2008; 9:22. https://doi.org/10.1186/1471-2121-9-22.
Kiga et al., An engineered Escherichia coli tyrosyl-tRNA synthetase for site-specific incorporation of an unnatural amino acid into proteins in eukaryotic translation and its application in a wheat germ cell-free system. Proc Natl Acad Sci U S A. Jul. 23, 2002;99(15):9715-20. Epub Jul. 3, 2002.
Kilbride et al., Determinants of product topology in a hybrid Cre-Tn3 resolvase site-specific recombination system. J Mol Biol. Jan. 13, 2006;355(2):185-95. Epub Nov. 9, 2005.
Kilcher et al., Brochothrix thermosphacta bacteriophages feature heterogeneous and highly mosaic genomes and utilize unique prophage insertion sites. J Bacteriol. Oct. 2010; 192(20):5441-53. doi: 10.1128/JB.00709-10. Epub Aug. 13, 2010.
Kim et al., DJ-1, a novel regulator of the tumor suppressor PTEN. Cancer Cell. 2005;7(3):263-273.
Kim et al., Genome-wide target specificity of CRISPR RNA-guided adenine base editors. Nat Biotechnol. Apr. 2019;37(4):430-435. doi: 10.1038/s41587-019-0050-1. Epub Mar. 4, 2019.
Kim et al., A library of TAL effector nucleases spanning the human genome. Nat Biotechnol. Mar. 2013;31(3):251-8. Doi: 10.1038/nbt.2517. Epub Feb. 17, 2013.
Kim et al., Adenine base editors catalyze cytosine conversions in human cells. Nat Biotechnol. Oct. 2019;37(10):1145-1148. doi: 10.1038/s41587-019-0254-4. Epub Sep. 23, 2019.
Kim et al., An anionic human protein mediates cationic liposome delivery of genome editing proteins into mammalian cells. Nat Commun. Jul. 2, 2019;10(1):2905. doi: 10.1038/s41467-019-10828-3.
Kim et al., Evaluating and Enhancing Target Specificity of Gene-Editing Nucleases and Deaminases. Annu Rev Biochem. Jun. 20, 2019;88:191-220. doi: 10.1146/annurev-biochem-013118-111730. Epub Mar. 18, 2019.
Kim et al., Genome-wide target specificities of CRISPR RNA-guided programmable deaminases. Nat Biotechnol. May 2017;35(5):475-480. doi: 10.1038/nbt.3852. Epub Apr. 10, 2017.
Kim et al., High cleavage efficiency of a 2A peptide derived from porcine teschovirus-1 in human cell lines, zebrafish and mice. PLoS One. 2011;6(4):e18556. doi: 10.1371/journal.pone.0018556. Epub Apr. 29, 2011.
Kim et al., High-throughput analysis of the activities of xCas9, SpCas9-NG and SpCas9 at matched and mismatched target sequences in human cells. Nat Biomed Eng. Jan. 2020;4(1):111-124. doi: 10.1038/s41551-019-0505-1. Epub Jan. 14, 2020.
Kim et al., Highly efficient RNA-guided base editing in mouse embryos. Nat Biotechnol. May 2017;35(5):435-437. doi: 10.1038/nbt.3816. Epub Feb. 27, 2017.
Kim et al., Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res. Jun. 2014;24(6):1012-9. doi: 10.1101/gr.171322.113. Epub Apr. 2, 2014.
Kim et al., In vivo genome editing with a small Cas9 orthologue derived from Campylobacter jejuni. Nat Commun. Feb. 21, 2017;8:14500. doi: 10.1038/ncomms14500. PMID: 28220790; PMCID: PMC5473640.
Kim et al., In vivo high-throughput profiling of CRISPR-Cpf1 activity. Nat Methods. Feb. 2017;14(2): 153-159. doi: 10.1038/nmeth.4104. Epub Dec. 19, 2016.
Kim et al., Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nat Biotechnol. Apr. 2017;35(4):371-376. doi: 10.1038/nbt.3803. Epub Feb. 13, 2017.
Kim et al., Mycobacteriophage Bxb1 integrates into the Mycobacterium smegmatis groEL1 gene. Mol Microbiol. Oct. 2003;50(2):463-73. doi: 10.1046/j.1365-2958.2003.03723.x.
Kim et al., Predicting the efficiency of prime editing guide RNAs in human cells. Nat Biotechnol. Feb. 2021;39(2):198-206. doi: 10.1038/s41587-020-0677-y. Epub Sep. 21, 2020.
Kim et al., RAD51 mutants cause replication defects and chromosomal instability. Mol Cell Biol. Sep. 2012;32(18):3663-80. doi: 10.1128/MCB.00406-12. Epub Jul. 9, 2012.
Kim et al., Rescue of high-specificity Cas9 variants using sgRNAs with matched 5′ nucleotides. Genome Biol. Nov. 15, 2017;18(1):218. doi: 10.1186/s13059-017-1355-3.
Kim et al., Structural and kinetic characterization of Escherichia coli TadA, the wobble-specific tRNA deaminase. Biochemistry. May 23, 2006;45(20):6407-16. doi: 10.1021/bi0522394. PMID: 16700551.
Kim et al., TALENs and ZFNs are associated with different mutationsignatures. Nat Methods. Mar. 2013;10(3):185. doi: 10.1038/nmeth.2364. Epub Feb. 10, 2013.
Kim et al., Targeted genome editing in human cells with zinc finger nucleases constructed via modular assembly. Genome Res. Jul. 2009; 19(7):1279-88. doi: 10.1101/gr.089417.108. Epub May 21, 2009.
Kim et al., The role of apolipoprotein E in Alzheimer's disease. Neuron. Aug. 13, 2009;63(3):287-303. doi: 10.1016/j.neuron.2009.06.026.
Kim et al., Transcriptional repression by zinc finger peptides. Exploring the potential for applications in gene therapy. J Biol Chem. Nov. 21, 1997;272(47):29795-800.
King et al., No gain, No pain: NaV1.7 as an analgesic target. ACS Chem Neurosci. Sep. 17, 2014;5(9):749-51. doi: 10.1021/cn500171p. Epub Aug. 11, 2014.
Kitamura et al., Uracil DNA glycosylase counteracts APOBEC3G-induced hypermutation of hepatitis B viral genomes: excision repair of covalently closed circular DNA. PloS Pathog. 2013;9(5):e1003361. doi: 10.1371/journal.ppat.1003361. Epub May 16, 2013.
Klapacz et al., Frameshift mutagenesis and microsatellite instability induced by human alkyladenine DNA glycosylase. Mol Cell. Mar. 26, 2010;37(6):843-53. doi: 10.1016/j.molcel.2010.01.038.
Klauser et al., An engineered small RNA-mediated genetic switch based on a ribozyme expression platform. Nucleic Acids Res. May 1, 2013;41(10):5542-52. doi: 10.1093/nar/gkt253. Epub Apr. 12, 2013.
Klein et al., Cocrystal structure of a class I preQ1 riboswitch reveals a pseudoknot recognizing an essential hypermodified nucleobase. Nat Struct Mol Biol. Mar. 2009; 16(3):343-4. doi: 10.1038/nsmb.1563.Epub Feb. 22, 2009.
Kleiner et al., In vitro selection of a DNA-templated small-molecule library reveals a class of macrocyclic kinase inhibitors. J Am Chem Soc. Aug. 25, 2010;132(33):11779-91. doi: 10.1021/ja104903x.
Kleinstiver et al., Broadening the targeting range of Staphylococcus aureus CRISPR-Cas9 by modifying PAM recognition. Nat Biotechnol. Dec. 2015;33(12):1293-1298. doi: 10.1038/nbt.3404. Epub Nov. 2, 2015.
Kleinstiver et al., Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature. Jul. 23, 2015;523(7561):481-5 and Supplementary Materials. doi: 10.1038/nature14592. Epub Jun. 22, 2015. 27 pages.
Kleinstiver et al., Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature. Jul. 23, 2015;523(7561):481-5. doi: 10.1038/nature14592. Epub Jun. 22, 2015.
Kleinstiver et al., High-fidelity CRISPR-Cas9 nucleases with no detectable genome-wide off-target effects. Nature. Jan. 28, 2016;529(7587):490-5. doi: 10.1038/nature16526. Epub Jan. 6, 2016.
Kleinstiver et al., Monomeric site-specific nucleases for genome editing. Proc Natl Acad Sci U S A. May 22, 2012;109(21):8061-6. doi: 10.1073/pnas.1117984109. Epub May 7, 2012.
Klement et al., Discrimination between bacteriophage T3 and T7 promoters by the T3 and T7 RNA polymerases depends primarily upon a three base-pair region located 10 to 12 base-pairs upstream from the start site. J Mol Biol. Sep. 5, 1990;215(1):21-9.
Klippel et al., Isolation and characterization of unusual gin mutants. EMBO J. Dec. 1, 1988;7(12):3983-9.
Klippel et al., The DNA invertase Gin of phage Mu: formation of a covalent complex with DNA via a phosphoserine at amino acid position 9. EMBO J. Apr. 1988;7(4):1229-37.
Klompe et al., Transposon-encoded CRISPR-Cas systems direct RNA-guided DNA integration. Nature. Jul. 2019;571(7764):219-225. doi: 10.1038/s41586-019-1323-z. Epub Jun. 12, 2019.
Kluesner et al., CRISPR-Cas9 cytidine and adenosine base editing of splice-sites mediates highly-efficient disruption of proteins in primary and immortalized cells. Nat Commun. Apr. 23, 2021;12(1):2437. doi: 10.1038/s41467-021-22009-2.
Knott et al., CRISPR-Cas guides the future of genetic engineering. Science. Aug. 31, 2018;361(6405):866-869. doi: 10.1126/science.aat5011.
Knott et al., Guide-bound structures of an RNA-targeting A-cleaving CRISPR-Cas13a enzyme. Nat Struct Mol Biol. Oct. 2017;24(10):825-833. doi: 10.1038/nsmb.3466. Epub Sep. 11, 2017.
Koblan et al., In vivo base editing rescues Hutchinson-Gilford progeria syndrome in mice. Nature. Jan. 2021;589(7843):608-614. doi: 10.1038/s41586-020-03086-7. Epub Jan. 6, 2021.
Koblan et al., Efficient C⋅G-to-G⋅C base editors developed using CRISPRi screens, target-library analysis, and machine learning. Nature Biotechnol. Jun. 28, 2021. https://doi.org/10.1038/s41587-021-00938-z.
Koblan et al., Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat Biotechnol. Oct. 2018;36(9):843-846. doi: 10.1038/nbt.4172. Epub May 29, 2018.
Kobori et al., Deep Sequencing Analysis of Aptazyme Variants Based on a Pistol Ribozyme. ACS Synth Biol. Jul. 21, 2017;6(7):1283-1288. doi: 10.1021/acssynbio.7b00057. Epub Apr. 14, 2017.
Kohli et al., A portable hot spot recognition loop transfers sequence preferences from APOBEC family members to activation-induced cytidine deaminase. J Biol Chem. Aug. 21, 2009;284(34):22898-904. doi: 10.1074/jbc.M109.025536. Epub Jun. 26, 2009.
Kohli et al., Local sequence targeting in the AID/APOBEC family differentially impacts retroviral restriction and antibody diversification. J Biol Chem. Dec. 24, 2010;285(52):40956-64. doi: 10.1074/jbc.M110.177402. Epub Oct. 6, 2010.
Koike-Yusa et al., Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library. Nat Biotechnol. Mar. 2014;32(3):267-73. doi: 10.1038/nbt.2800. Epub Dec. 23, 2013.
Kolot et al., Site promiscuity of coliphage HK022 integrase as a tool for gene therapy. Gene Ther. Jul. 2015;22(7):521-7. doi: 10.1038/gt.2015.9. Epub Mar. 12, 2015.
Kolot et al., Site-specific recombination in mammalian cells expressing the Int recombinase of bacteriophage HK022. Mol Biol Rep. Aug. 1999;26(3):207-13. doi: 10.1023/a:1007096701720.
Komor et al., CRISPR-Based Technologies for the Manipulation of Eukaryotic Genomes. Cell. Jan. 12, 2017;168(1-2):20-36. doi: 10.1016/j.cell.2016.10.044.
Komor et al., Improved base excision repair inhibition and bacteriophage Mu Gam protein yields C:G-to-T:A base editors with higher efficiency and product purity. Sci Adv. Aug. 30, 2017;3(8):eaao4774. doi: 10.1126/sciadv.aao4774. eCollection Aug. 2017.
Komor et al., Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature. Apr. 20, 2016;533(7603):420-4. doi: 10.1038/nature17946.
Komor, Editing the Genome Without Double-Stranded DNA Breaks. ACS Chem Biol. Feb. 16, 2018;13(2):383-388. doi: 10.1021/acschembio.7b00710. Epub Oct. 9, 2017.
Konermann et al., Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature. Jan. 29, 2015;517(7536):583-8. doi: 10.1038/nature14136. Epub Dec. 10, 2014.
Konishi et al., Amino acid substitutions away from the RNase H catalytic site increase the thermal stability of Moloney murine leukemia virus reverse transcriptase through RNase H inactivation. Biochem Biophys Res Commun. Nov. 14, 2014;454(2):269-74. doi: 10.1016/j.bbrc.2014.10.044. Epub Oct. 17, 2014.
Koonin et al., Diversity, classification and evolution of CRISPR-Cas systems. Curr Opin Microbiol. 2017;37:67?78. doi:10.1016/j.mib.2017.05.008.
Kosicki et al., Repair of double-strand breaks induced by CRISPR-Cas9 leads to large deletions and complex rearrangements. Nat Biotechnol. Sep. 2018;36(8):765-771. doi: 10.1038/nbt.4192. Epub Jul. 16, 2018.
Kotewicz et al., Cloning and overexpression of Moloney murine leukemia virus reverse transcriptase in Escherichia coli. Gene. 1985;35(3):249-58. doi: 10.1016/0378-1119(85)90003-4.
Kotewicz et al., Isolation of cloned Moloney murine leukemia virus reverse transcriptase lacking ribonuclease H activity. Nucleic Acids Res. Jan. 11, 1988;16(1):265-77. doi: 10.1093/nar/16.1.265.
Kotin, Prospects for the use of adeno-associated virus as a vector for human gene therapy. Hum Gene Ther. Jul. 1994;5(7):793-801. doi: 10.1089/hum.1994.5.7-793.
Kouzminova et al., Patterns of chromosomal fragmentation due to uracil-DNA incorporation reveal a novel mechanism of replication-dependent double-stranded breaks. Mol Microbiol. Apr. 2008;68(1):202-15. doi: 10.1111/j.1365-2958.2008.06149.x.
Kowal et al., Exploiting unassigned codons in Micrococcus luteus for tRNA-based amino acid mutagenesis. Nucleic Acids Res. Nov. 15, 1997;25(22):4685-9.
Kowalski et al., Delivering the Messenger: Advances in Technologies for Therapeutic mRNA Delivery. Mol Ther. Apr. 10, 2019;27(4):710-728. doi: 10.1016/j.ymthe.2019.02.012. Epub Feb. 19, 2019.
Kozak, An analysis of 5′-noncoding sequences from 699 vertebrate messenger RNAs. Nucleic Acids Res. Oct. 26, 1987;15(20):8125-48. doi: 10.1093/nar/15.20.8125.
Kraft et al., Deletions, Inversions, Duplications: Engineering of Structural Variants using CRISPR/Cas in Mice. Cell Rep. Feb. 10, 2015;10(5):833-839. doi: 10.1016/j.celrep.2015.01.016. Epub Feb. 7, 2015.
Kremer et al., Adenovirus and adeno-associated virus mediated gene transfer. Br Med Bull. Jan. 1995;51(1):31-44. doi: 10.1093/oxfordjournals.bmb.a072951.
Krokan et al., Uracil in DNA—occurrence, consequences and repair. Oncogene. Dec. 16, 2002;21(58):8935-48. doi: 10.1038/sj.onc.1205996.
Krokan et al., Base excision repair. Cold Spring Harb Perspect Biol. Apr. 1, 2013;5(4):a012583. doi: 10.1101/cshperspect.a012583.
Krzywkowski et al., Limited reverse transcriptase activity of phi29 DNA polymerase. Nucleic Acids Res. Apr. 20, 2018;46(7):3625-3632. doi: 10.1093/nar/gky190.
Kumar et al., Gene therapy for chronic neuropathic pain: how does it work and where do we stand today? Pain Med. May 2011;12(5):808-22. doi: 10.1111/j.1526-4637.2011.01120.x.
Kumar et al., Structural and functional consequences of the mutation of a conserved arginine residue in alphaA and alphaB crystallins. J Biol Chem. Aug. 20, 1999;274(34):24137-41.
Kundu et al., Leucine to proline substitution by SNP at position 197 in Caspase-9 gene expression leads to neuroblastoma: a bioinformatics analysis. 3 Biotech. 2013; 3:225-34.
Kunkel et al., Eukaryotic Mismatch Repair in Relation to DNA Replication. Annu Rev Genet. 2015;49:291-313. doi: 10.1146/annurev-genet-112414-054722.
Kunz et al., DNA Repair in mammalian cells: Mismatched repair: variations on a theme. Cell Mol Life Sci. Mar. 2009;66(6):1021-38. doi: 10.1007/s00018-009-8739-9.
Kurjan et al., Structure of a yeast pheromone gene (MF alpha): a putative alpha-factor precursor contains four tandem copies of mature alpha-factor. Cell. Oct. 1982;30(3):933-43. doi: 10.1016/0092-8674(82)90298-7.
Kury et al., De Novo Disruption of the Proteasome Regulatory Subunit PSMD12 Causes a Syndromic Neurodevelopmental Disorder. Am J Hum Genet. Feb. 2, 2017;100(2):352-363. doi: 10.1016/j.ajhg.2017.01.003. Epub Jan. 26, 2017.
Kuscu et al., CRISPR-Cas9-AID base editor is a powerful gain-of-function screening tool. Nat Methods. Nov. 29, 2016;13(12):983-984. doi: 10.1038/nmeth.4076.
Kuscu et al., CRISPR-STOP: gene silencing through base-editing-induced nonsense mutations. Nat Methods. Jul. 2017; 14(7):710-712. doi: 10.1038/nmeth.4327. Epub Jun. 5, 2017.
Kuscu et al., Genome-wide analysis reveals characteristics of off-target sites bound by the Cas9 endonuclease. Nat Biotechnol. Jul. 2014;32(7):677-83. doi: 10.1038/nbt.2916. Epub May 18, 2014.
Kwart et al., Precise and efficient scarless genome editing in stem cells using CORRECT. Nat Protoc. Feb. 2017; 12(2):329-354. doi: 10.1038/nprot.2016.171. Epub Jan. 19, 2017.
Kweon et al., A CRISPR-based base-editing screen for the functional assessment of BRCA1 variants. Oncogene. Jan. 2020;39(1):30-35. doi: 10.1038/s41388-019-0968-2. Epub Aug. 29, 2019.
Kweon et al., Fusion guide RNAs for orthogonal gene manipulation with Cas9 and Cpf1. Nat Commun. Nov. 23, 2017;8(1):1723. doi: 10.1038/s41467-017-01650-w. Erratum in: Nat Commun. Jan. 16, 2018;9(1):303.
Kwon et al., Chemical basis of glycine riboswitch cooperativity. RNA. Jan. 2008;14(1):25-34. Epub Nov. 27, 2007.
Köhrer et al., A possible approach to site-specific insertion of two different unnatural amino acids into proteins in mammalian cells via nonsense suppression. Chem Biol. Nov. 2003;10(11):1095-102.
Köhrer et al., Complete set of orthogonal 21st aminoacyl-tRNA synthetase-amber, ochre and opal suppressor tRNA pairs: concomitant suppression of three different termination codons in an mRNA in mammalian cells. Nucleic Acids Res. Dec. 1, 2004;32(21):6200-11. Print 2004.
Kügler et al., Human synapsin 1 gene promoter confers highly neuron-specific long-term transgene expression from an adenoviral vector in the adult rat brain depending on the transduced area. Gene Ther. Feb. 2003; 10(4):337-47. doi: 10.1038/sj.gt.3301905.
Lada et al., Mutator effects and mutation signatures of editing deaminases produced in bacteria and yeast. Biochemistry (Mosc). Jan. 2011;76(1):131-46.
Lakich et al., Inversions disrupting the factor VIII gene are a common cause of severe haemophilia A. Nat Genet. Nov. 1993;5(3):236-41. doi: 10.1038/ng1193-236.
Lancaster et al., Limited trafficking of a neurotropic virus through inefficient retrograde axonal transport and the type I interferon response. PLoS Pathog. Mar. 5, 2010;6(3):e1000791. doi: 10.1371/journal.ppat.1000791.
Landrum et al., ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. Jan. 4, 2016;44(D1):D862-8. doi: 10.1093/nar/gkv1222. Epub Nov. 17, 2015.
Landrum et al., ClinVar: public archive of relationships among sequence variation and human phenotype. Nucleic Acids Res. Jan. 2014;42(Database issue):D980-5. doi: 10.1093/nar/gkt1113. Epub Nov. 14, 2013.
Langer et al., Chemical and Physical Structure of Polymers as Carriers for Controlled Release of Bioactive Agents: A Review. J Macromol Sci, Part C, 1983;23(1):61-126. doi: 10.1080/07366578308079439.
Langer et al., Chemical and Physical Structure of Polymers as Carriers for Controlled Release of Bioactive Agents: A Review. Journal of Macromolecular Science, 2006;23(1):61-126. DOI: 10.1080/07366578308079439.
Langer et al., New methods of drug delivery. Science. Sep. 28, 1990;249(4976):1527-33.
Lapinaite et al., DNA capture by a CRISPR-Cas9-guided adenine base editor. Science. Jul. 31, 2020;369(6503):566-571. doi: 10.1126/science.abb1390.
Larson et al., CRISPR interference (CRISPRi) for sequence-specific control of gene expression. Nat Protoc. Nov. 2013;8(11):2180-96. doi: 10.1038/nprot.2013.132. Epub Oct. 17, 2013.
Lau et al., Molecular basis for discriminating between normal and damaged bases by the human alkyladenine glycosylase, AAG. Proc Natl Acad Sci U S A. Dec. 5, 2000;97(25):13573-8.
Lauer et al., Construction, characterization, and use of two Listeria monocytogenes site-specific phage integration vectors. J Bacteriol. Aug. 2002; 184(15):4177-86. doi: 10.1128/jb.184.15.4177-4186.2002.
Lavergne et al., Defects in type IIA von Willebrand disease: a cysteine 509 to arginine substitution in the mature von Willebrand factor disrupts a disulphide loop involved in the interaction with platelet glycoprotein Ib-IX. Br J Haematol. Sep. 1992;82(1):66-72.
Lawrence et al., Supercharging proteins can impart unusual resilience. J Am Chem Soc. Aug. 22, 2007;129(33):10110-2. Epub Aug. 1, 2007.
Lawyer et al., High-level expression, purification, and enzymatic characterization of full-length Thermus aquaticus DNA polymerase and a truncated form deficient in 5′ to 3′ exonuclease activity. PCR Methods Appl. May 1993;2(4):275-87. doi: 10.1101/gr.2.4.275.
Lazar et al., Transforming growth factor alpha: mutation of aspartic acid 47 and leucine 48 results in different biological activities. Mol Cell Biol. Mar. 1988;8(3):1247-52.
Lazarevic et al., Nucleotide sequence of the Bacillus subtilis temperate bacteriophage SPbetac2. Microbiology (Reading). May 1999;145 ( Pt 5):1055-1067. doi: 10.1099/13500872-145-5-1055.
Le et al., SMNDelta7, the major product of the centromeric survival motor neuron (SMN2) gene, extends survival in mice with spinal muscular atrophy and associates with full-length SMN. Hum Mol Genet. Mar. 15, 2005;14(6):845-57. doi: 10.1093/hmg/ddi078. Epub Feb. 9, 2005.
Le Grice et al., Purification and characterization of recombinant equine infectious anemia virus reverse transcriptase. J Virol. Dec. 1991;65(12):7004-7. doi: 10.1128/JVI.65.12.7004-7007.1991.
Leaver-Fay et al., ROSETTA3: an object-oriented software suite for the simulation and design of macromolecules. Methods Enzymol. 2011;487:545-74. doi: 10.1016/B978-0-12-381270-4.00019-6.
Leconte et al., A population-based experimental model for protein evolution: effects of mutation rate and selection stringency on evolutionary outcomes. Biochemistry. Feb. 26, 2013;52(8):1490-9. doi: 10.1021/bi3016185. Epub Feb. 14, 2013.
Ledford, Gene-editing hack yields pinpoint precision. Nature, Apr. 20, 2016. http://www.nature.com/news/gene-editing-hack-yields-pinpoint-precision-1.19773.
Lee et al., A chimeric thyroid hormone receptor constitutively bound to DNA requires retinoid X receptor for hormone-dependent transcriptional activation in yeast. Mol Endocrinol. Sep. 1994;8(9):1245-52.
Lee et al., A monoclonal antibody that targets a NaV1.7 channel voltage sensor for pain and itch relief. Cell. Jun. 5, 2014;157(6):1393-1404. doi: 10.1016/j.cell.2014.03.064. Epub May 22, 2014. Retraction in: Cell. Jun. 2, 20205;181(7):1695.
Lee et al., An allosteric self-splicing ribozyme triggered by a bacterial second messenger. Science. Aug. 13, 2010;329(5993):845-8. doi: 10.1126/science.1190713.
Lee et al., Failure to detect DNA-guided genome editing using Natronobacterium gregoryi Argonaute. Nat Biotechnol. Nov. 28, 2016;35(1):17-18. doi: 10.1038/nbt.3753.
Lee et al., Group I Intron-Based Therapeutics Through Trans-Splicing Reaction. Prog Mol Biol Transl Sci. 2018;159:79-100. doi: 10.1016/bs.pmbts.2018.07.001. Epub Aug. 9, 2018.
Lee et al., PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas. Oncogene. Feb. 17, 2005;24(8):1477-80.
Lee et al., Recognition of liposomes by cells: in vitro binding and endocytosis mediated by specific lipid headgroups and surface charge density. Biochim Biophys Acta. Jan. 31, 1992;1103(2):185-97.
Lee et al., Ribozyme Mediated gRNA Generation for In Vitro and In Vivo CRISPR/Cas9 Mutagenesis. PLoS One. Nov. 10, 2016;11(11):e0166020. doi: 10.1371/journal.pone.0166020.eCollection 2016.
Lee et al., Simultaneous targeting of linked loci in mouse embryos using base editing. Sci Rep. Feb. 7, 2019;9(1):1662. doi: 10.1038/s41598-018-33533-5.
Lee et al., Single C-to-T substitution using engineered APOBEC3G-nCas9 base editors with minimum genome- and transcriptome-wide off-target effects. Sci Adv. Jul. 15, 2020;6(29):eaba1773. doi: 10.1126/sciadv.aba1773.
Lee et al., Single C-to-T substitution using engineered APOBEC3G-nCas9 base editors with minimum genome- and transcriptome-wide off-target effects. Sci Adv. Jul. 15, 2020;6(29):eaba1773. doi: 10.1126/sciadv.aba1773. 13 pages.
Lee et al., Site-specific integration of mycobacteriophage L5: integration-proficient vectors for Mycobacterium smegmatis, Mycobacterium tuberculosis, and bacille Calmette-Guérin. Proc Natl Acad Sci U S A. Apr. 15, 1991;88(8):3111-5. doi: 10.1073/pnas.88.8.3111.
Lee et al., Synthetically modified guide RNA and donor DNA are a versatile platform for CRISPR-Cas9 engineering. Elife. May 2, 2017;6:e25312. doi: 10.7554/eLife.25312.
Lee et al., Targeted chromosomal deletions in human cells using zinc finger nucleases. Genome Res. Jan. 2010 20: 81-89; Published in Advance Dec. 1, 2009, doi:10.1101/gr.099747.109.
Lee et al., Targeting fidelity of adenine and cytosine base editors in mouse embryos. Nat Commun. Nov. 15, 2018;9(1):4804. doi: 10.1038/s41467-018-07322-7.
Lee et al., Transcriptional regulation and its misregulation in disease. Cell. Mar. 14, 2013;152(6):1237-51. doi: 10.1016/j.cell.2013.02.014.
Lefebvre et al., Identification and characterization of a spinal muscular atrophy-determining gene. Cell. Jan. 13, 1995;80(1):155-65. doi: 10.1016/0092-8674(95)90460-3.
Lei et al., Efficient targeted gene disruption in Xenopus embryos using engineered transcription activator-like effector nucleases (TALENs). Proc Natl Acad Sci U S A. Oct. 23, 2012;109(43):17484-9. Doi: 10.1073/pnas.1215421109. Epub Oct. 8, 2012.
Lei et al., Site-specificity of serine integrase demonstrated by the attB sequence preference of ΦBT1 integrase. FEBS Lett. Apr. 2018;592(8):1389-1399. doi: 10.1002/1873-3468.13023. Epub Mar. 25, 2018.
Leipold et al., A de novo gain-of-function mutation in SCN11A causes loss of pain perception. Nat Genet. Nov. 2013;45(11):1399-404. doi: 10.1038/ng.2767. Epub Sep. 15, 2013.
Lemos et al., CRISPR/Cas9 cleavages in budding yeast reveal templated insertions and strand-specific insertion/deletion profiles. Proc Natl Acad Sci U S A. Feb. 27, 2018;115(9):E2040-E2047. doi: 10.1073/pnas.1716855115. Epub Feb. 13, 2018.
Lenk et al., Pathogenic mechanism of the FIG4 mutation responsible for Charcot-Marie-Tooth disease CMT4J. Plos Genet. Jun. 2011;7(6):e1002104. doi: 10.1371/journal.pgen.1002104. Epub Jun. 2, 2011.
Lesinski et al., The potential for targeting the STAT3 pathway as a novel therapy for melanoma. Future Oncol. Jul. 2013;9(7):925-7. doi: 10.2217/fon.13.83. Author Manuscript. 4 pages.
Levy et al., Cytosine and adenine base editing of the brain, liver, retina, heart and skeletal muscle of mice via adeno-associated viruses. Nat Biomed Eng. 2020;4(1):97-110. doi:10.1038/s41551-019-0501-5.
Levy et al., Inhibition of calcification of bioprosthetic heart valves by local controlled-release diphosphonate. Science. Apr. 12, 1985;228(4696):190-2.
Levy et al., Membrane-associated guanylate kinase dynamics reveal regional and developmental specificity of synapse stability. J Physiol. Mar. 1, 2017;595(5):1699-1709. doi: 10.1113/JP273147. Epub Jan. 18, 2017.
Lew et al., Protein splicing in vitro with a semisynthetic two-component minimal intein. J Biol Chem. Jun. 26, 1998;273(26):15887-90. doi: 10.1074/jbc.273.26.15887.
Lewis et al., A serum-resistant cytofectin for cellular delivery of antisense oligodeoxynucleotides and plasmid DNA. Proc Natl Acad Sci U S A. Apr. 16, 1996;93(8):3176-81.
Lewis et al., Building the Class 2 CRISPR-Cas Arsenal. Mol Cell 2017;65(3);377-379.
Lewis et al., Codon 129 polymorphism of the human prion protein influences the kinetics of amyloid formation. J Gen Virol. Aug. 2006;87(Pt 8):2443-9.
Lewis et al., Cytosine deamination and the precipitous decline of spontaneous mutation during Earth's history. Proc Natl Acad Sci U S A. Jul. 19, 2016;113(29):8194-9. doi: 10.1073/pnas.1607580113. Epub Jul. 5, 2016.
Lewis et al., RNA modifications and structures cooperate to guide RNA-protein interactions. Nat Rev Mol Cell Biol. Mar. 2017;18(3):202-210. doi: 10.1038/nrm.2016.163. Epub Feb. 1, 2017.
Li et al., A Radioactivity-Based Assay for Screening Human m6A-RNA Methyltransferase, METTL3-METTL14 Complex, and Demethylase ALKBH5. J Biomol Screen. Mar. 2016;21(3):290-7. doi: 10.1177/1087057115623264. Epub Dec. 23, 2015.
Li et al., Base editing with a Cpf1-cytidine deaminase fusion. Nat Biotechnol. Apr. 2018;36(4):324-327. doi: 10.1038/nbt.4102. Epub Mar. 19, 2018.
Li et al., Current approaches for engineering proteins with diverse biological properties. Adv Exp Med Biol. 2007;620:18-33.
Li et al., Disruption of splicing-regulatory elements using CRISPR/Cas9 to rescue spinal muscular atrophy in human iPSCs and mice. National Science Review. Jan. 1, 2020:92-101. DOI: 10.1093/nsr/nwz131. Retrieved from the Internet via https://academic.oup.com/nsr/article-pdf/7/92/33321439/nwz131.pdf. Last accessed Apr. 28, 2021.
Li et al., Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. Jul. 15, 2009;25(14):1754-60. doi: 10.1093/bioinformatics/btp324. Epub May 18, 2009.
Li et al., Generation of Targeted Point Mutations in Rice by a Modified CRISPR/Cas9 System. Mol Plant. Mar. 6, 2017;10(3):526-529. doi: 10.1016/j.molp.2016.12.001. Epub Dec. 8, 2016.
Li et al., Highly efficient and precise base editing in discarded human tripronuclear embryos. Protein Cell. Aug. 19, 2017. doi: 10.1007/s13238-017-0458-7. [Epub ahead of print].
Li et al., Lagging strand DNA synthesis at the eukaryotic replication fork involves binding and stimulation of FEN-1 by proliferating cell nuclear antigen. J Biol Chem. Sep. 22, 1995;270(38):22109-12. doi: 10.1074/jbc.270.38.22109.
Li et al., Loss of post-translational modification sites in disease. Pac Symp Biocomput. 2010:337-47. doi: 10.1142/9789814295291_0036.
Li et al., Modularly assembled designer TAL effector nucleases for targeted gene knockout and gene replacement in eukaryotes. Nucleic Acids Res. Aug. 2011;39(14):6315-25. doi: 10.1093/nar/gkr188. Epub Mar. 31, 2011.
Li et al., Multiplex and homologous recombination-mediated genome editing in Arabidopsis and Nicotiana benthamiana using guide RNA and Cas9. Nat Biotechnol. Aug. 2013;31(8):688-91. doi: 10.1038/nbt.2654.
Li et al., Precise Modifications of Both Exogenous and Endogenous Genes in Rice by Prime Editing. Mol Plant. May 4, 2020;13(5):671-674. doi: 10.1016/j.molp.2020.03.011. Epub Mar. 25, 2020.
Li et al., Programmable Single and Multiplex Base-Editing in Bombyx mori Using RNA-Guided Cytidine Deaminases. G3 (Bethesda). May 4, 2018;8(5):1701-1709. doi: 10.1534/g3.118.200134.
Li et al., Protein trans-splicing as a means for viral vector-mediated in vivo gene therapy. Hum Gene Ther. Sep. 2008;19(9):958-64. doi: 10.1089/hum.2008.009.
Li et al., RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. Aug. 4, 2011;12:323. doi: 10.1186/1471-2105-12-323.
Li et al., TAL nucleases (TALNs): hybrid proteins composed of TAL effectors and FokI DNA-cleavage domain. Nucleic Acids Res. Jan. 2011;39(1):359-72. doi: 10.1093/nar/gkq704. Epub Aug. 10, 2010.
Li, Mechanisms and functions of DNA mismatch repair. Cell Res. Jan. 2008;18(1):85-98. doi: 10.1038/cr.2007.115.
Liang et al., Correction of β-thalassemia mutant by base editor in human embryos. Protein Cell. Nov. 2017;8(11):811-822. doi: 10.1007/s13238-017-0475-6. Epub Sep. 23, 2017.
Liang et al., Homology-directed repair is a major double-strand break repair pathway in mammalian cells. Proc Natl Acad Sci U S A. Apr. 28, 1998;95(9):5172-7. doi: 10.1073/pnas.95.9.5172.
Liang et al., Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection. Send to; J Biotechnol. Aug. 20, 2015;208:44-53. doi: 10.1016/j.jbiotec.2015.04.024.
Liao et al., One-step assembly of large CRISPR arrays enables multi-functional targeting and reveals constraints on array design. bioRxiv. May 2, 2018. doi: 10.1101/312421. 45 pages.
Lieber et al., Mechanism and regulation of human non-homologous DNA end-joining. Nat Rev Mol Cell Biol. Sep. 2003;4(9):712-20.
Liefke et al., The oxidative demethylase ALKBH3 marks hyperactive gene promoters in human cancer cells. Genome Med. Jun. 30, 2015;7(1):66. doi: 10.1186/s13073-015-0180-0.
Lienert et al., Two- and three-input TALE-based AND logic computation in embryonic stem cells. Nucleic Acids Res. Nov. 2013;41(21):9967-75. doi: 10.1093/nar/gkt758. Epub Aug. 27, 2013.
Lilley, D.M. The Varkud Satellite Ribozyme. RNA. Feb. 2004;10(2):151-8.doi: 10.1261/rna.5217104.
Lim et al., Crystal structure of the moloney murine leukemia virus RNase H domain. J Virol. Sep. 2006;80(17):8379-89. doi: 10.1128/JVI.00750-06.
Lim et al., Viral vectors for neurotrophic factor delivery: a gene therapy approach for neurodegenerative diseases of the CNS. Pharmacol Res. Jan. 2010;61(1):14-26. doi: 10.1016/j.phrs.2009.10.002. Epub Oct. 17, 2009.
Lin et al., [Construction and evaluation of DnaB split intein high expression vector and a six amino acids cyclic peptide library]. Sheng Wu Gong Cheng Xue Bao. Nov. 2008;24(11):1924-30. Chinese.
Lin et al., Base editing-mediated splicing correction therapy for spinal muscular atrophy. Cell Res. Jun. 2020;30(6):548-550. doi: 10.1038/s41422-020-0304-y. Epub Mar. 24, 2020.
Lin et al., Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery. Elife. Dec. 15, 2014;3:e04766. doi: 10.7554/eLife.04766.
Lin et al., High-efficiency prime editing with optimized, paired pegRNAs in plants. Nat Biotechnol. Aug. 2021;39(8):923-927. doi: 10.1038/s41587-021-00868-w. Epub Mar. 25, 2021.
Lin et al., Prime genome editing in rice and wheat. Nat Biotechnol. May 2020;38(5):582-585 and Supplemental Info. doi: 10.1038/s41587-020-0455-x. Epub Mar. 16, 2020. 8 pages.
Lin et al., Prime genome editing in rice and wheat. Nat Biotechnol. May 2020;38(5):582-585. doi: 10.1038/s41587-020-0455-x. Epub Mar. 16, 2020.
Lin et al., The human REV1 gene codes for a DNA template-dependent dCMP transferase. Nucleic Acids Res. Nov. 15, 1999;27(22):4468-75. doi: 10.1093/nar/27.22.4468.
Lindahl, T., Instability and decay of the primary structure of DNA. Nature. Apr. 22, 1993;362(6422):709-15. doi: 10.1038/362709a0.
Link et al., Engineering ligand-responsive gene-control elements: lessons learned from natural riboswitches. Gene Ther. Oct. 2009;16(10):1189-201. doi: 10.1038/gt.2009.81. Epub Jul. 9, 2009. Review.
Liu et al., C2c1-sgRNA Complex Structure Reveals RNA-Guided DNA Cleavage Mechanism. Molecular Cell Jan. 2017;65(2):310-22.
Liu et al., Split dnaE genes encoding multiple novel inteins in Trichodesmium erythraeum. J Biol Chem. Jul. 18, 2003;278(29):26315-8. doi: 10.1074/jbc.C300202200. Epub May 24, 2003.
Liu et al., A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. Feb. 2014;10(2):93-5. doi: 10.1038/nchembio.1432. Epub Dec. 6, 2013.
Liu et al., Adding new chemistries to the genetic code. Annu Rev Biochem. 2010;79:413-44. doi: 10.1146/annurev.biochem.052308.105824.
Liu et al., Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol. Feb. 2013;9(2):106-18. doi: 10.1038/nrneurol.2012.263. Epub Jan. 8, 2013.
Liu et al., Balancing AID and DNA repair during somatic hypermutation. Trends Immunol. Apr. 2009;30(4):173-81. doi: 10.1016/j.it.2009.01.007.
Liu et al., Calcineurin is a common target of cyclophilin-cyclosporin A and FKBP-FK506 complexes. Cell. Aug. 23, 1991;66(4):807-15. doi: 10.1016/0092-8674(91)90124-h.
Liu et al., CasX enzymes comprise a distinct family of RNA-guided genome editors. Nature. Feb. 2019;566(7743):218-223. doi: 10.1038/s41586-019-0908-x. Epub Feb. 4, 2019. Author manuscript entitled CRISPR-CasX is an RNA-dominated enzyme active for human genome editing.
Liu et al., Cell-penetrating peptide-mediated delivery of TALEN proteins via bioconjugation for genome engineering. PLoS One. Jan. 20, 2014;9(1):e85755. doi: 10.1371/journal.pone.0085755. eCollection 2014.
Liu et al., Computational approaches for effective CRISPR guide RNA design and evaluation. Comput Struct Biotechnol J. Nov. 29, 2019;18:35-44. doi: 10.1016/j.csbj.2019.11.006.
Liu et al., Design of polydactyl zinc-finger proteins for unique addressing within complex genomes. Proc Natl Acad Sci U S A. May 27, 1997;94(11):5525-30.
Liu et al., Direct Promoter Repression by BCL11A Controls the Fetal to Adult Hemoglobin Switch. Cell. Apr. 5, 2018;173(2):430-442.e17. doi: 10.1016/j.cell.2018.03.016. Epub Mar. 29, 2018.
Liu et al., Distance determination by GIY-YIG intron endonucleases: discrimination between repression and cleavage functions. Nucleic Acids Res. Mar. 31, 2006;34(6):1755-64. Print 2006.
Liu et al., Editing DNA Methylation in the Mammalian Genome. Cell. Sep. 22, 2016;167(1):233-247.e17. doi: 10.1016/j.cell.2016.08.056.
Liu et al., Engineering a tRNA and aminoacyl-tRNA synthetase for the site-specific incorporation of unnatural amino acids into proteins in vivo. Proc Natl Acad Sci U S A. Sep. 16, 1997;94(19):10092-7.
Liu et al., Fast Colorimetric Sensing of Adenosine and Cocaine Based on a General Sensor Design Involving Aptamers and Nanoparticles. Angew Chem. Dec. 16, 2006;45(1):90-4. DOI: 10.1002/anie.200502589.
Liu et al., Fast Colorimetric Sensing of Adenosine and Cocaine Based on a General Sensor Design Involving Aptamers and Nanoparticles. Angew Chem. 2006;118(1):96-100.
Liu et al., Flap endonuclease 1: a central component of DNA metabolism. Annu Rev Biochem. 2004;73:589-615. doi:10.1146/annurev.biochem.73.012803.092453.
Liu et al., Functional Nucleic Acid Sensors. Chem Rev. May 2009; 109(5):1948-98. doi: 10.1021/cr030183i.
Liu et al., Genetic incorporation of unnatural amino acids into proteins in mammalian cells. Nat Methods. Mar. 2007;4(3):239-44. Epub Feb. 25, 2007.
Liu et al., Highly efficient RNA-guided base editing in rabbit. Nat Commun. Jul. 13, 2018;9(1):2717. doi: 10.1038/s41467-018-05232-2.
Liu et al., Human BRCA2 protein promotes RAD51 filament formation on RPA-covered single-stranded DNA. Nat Struct Mol Biol. Oct. 2010; 17(10):1260-2. doi: 10.1038/nsmb.1904. Epub Aug. 22, 2010.
Liu et al., Improving Editing Efficiency for the Sequences with NGH PAM Using xCas9-Derived Base Editors. Mol Ther Nucleic Acids. Sep. 6, 2019;17:626-635. doi: 10.1016/j.omtn.2019.06.024. Epub Jul. 12, 2019.
Liu et al., Intrinsic Nucleotide Preference of Diversifying Base Editors Guides Antibody Ex Vivo Affinity Maturation. Cell Rep. Oct. 23, 2018;25(4):884-892.e3. doi: 10.1016/j.celrep.2018.09.090.
Liu et al., N(6)-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature. Feb. 26, 2015;518(7540):560-4. doi: 10.1038/nature14234.
Liu et al., Probing N6-methyladenosine RNA modification status at single nucleotide resolution in mRNA and long noncoding RNA. RNA. Dec. 2013;19(12):1848-56. doi: 10.1261/rna.041178.113. Epub Oct. 18, 2013.
Liu et al., Reverse transcriptase of foamy virus. Purification of the enzymes and immunological identification. Arch Virol. 1977;55(3):187-200. doi: 10.1007/BF01319905.
Liu et al., Reverse transcriptase-mediated tropism switching in Bordetella bacteriophage. Science. Mar. 15, 2002;295(5562):2091-4. doi: 10.1126/science.1067467.
Liu et al., Saccharomyces cerevisiae flap endonuclease 1 uses flap equilibration to maintain triplet repeat stability. Mol Cell Biol. May 2004;24(9):4049-64. doi: 10.1128/MCB.24.9.4049-4064.2004.
Liu et al., The Molecular Architecture for RNA-Guided RNA Cleavage by Cas13a. Cell. Aug. 10, 2017;170(4):714-726.e10. doi: 10.1016/j.cell.2017.06.050. Epub Jul. 27, 2017.
Liu et al., Usherin is required for maintenance of retinal photoreceptors and normal development of cochlear hair cells. Proc Natl Acad Sci U S A. Mar. 13, 2007;104(11):4413-8. doi: 10.1073/pnas.0610950104. Epub Mar. 5, 2007.
Loessner et al., Complete nucleotide sequence, molecular analysis and genome structure of bacteriophage A118 of Listeria monocytogenes: implications for phage evolution. Mol Microbiol. Jan. 2000;35(2):324-40. doi: 10.1046/j.1365-2958.2000.01720.x.
Lombardo et al., Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery. Nat Biotechnol. Nov. 2007;25(11):1298-306. Epub Oct. 28, 2007.
Long et al., Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy. Science. Jan. 22, 2016;351(6271):400-3. doi: 10.1126/science.aad5725. Epub Dec. 31, 2015.
Lopez-Girona et al., Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide. Leukemia. Nov. 2012;26(11):2326-35. doi: 10.1038/leu.2012.119. Epub May 3, 2012.
Lorenz et al., ViennaRNA Package 2.0. Algorithms Mol Biol. Nov. 24, 2011;6:26. doi: 10.1186/1748-7188-6-26.
Lorson et al., A single nucleotide in the SMN gene regulates splicing and is responsible for spinal muscular atrophy. Proc Natl Acad Sci U S A. May 25, 1999;96(11):6307-11. doi: 10.1073/pnas.96.11.6307.
Losey et al., Crystal structure of Staphylococcus sureus tRNA adenosine deaminase tadA in complex with RNA. Nature Struct. Mol. Biol. Feb. 2006;13(2): 153-9.
Lu et al., Precise Editing of a Target Base in the Rice Genome Using a Modified CRISPR/Cas9 System. Mol Plant. Mar. 6, 2017;10(3):523-525. doi: 10.1016/j.molp.2016.11.013. Epub Dec. 6, 2016.
Luan et al., Reverse transcription of R2Bm RNA is primed by a nick at the chromosomal target site: a mechanism for non-LTR retrotransposition. Cell. Feb. 26, 1993;72(4):595-605. doi: 10.1016/0092-8674(93)90078-5.
Luckow et al., High level expression of nonfused foreign genes with Autographa californica nuclear polyhedrosis virus expression vectors. Virology. May 1989;170(1):31-9. doi: 10.1016/0042-6822(89)90348-6.
Lukacsovich et al., Repair of a specific double-strand break generated within a mammalian chromosome by yeast endonuclease I-SceI. Nucleic Acids Res. Dec. 25, 1994;22(25):5649-57. doi: 10.1093/nar/22.25.5649.
Lundberg et al., Delivery of short interfering RNA using endosomolytic cell-penetrating peptides. FASEB J. Sep. 2007;21(11):2664-71. Epub Apr. 26, 2007.
Lundquist et al., Site-directed mutagenesis and characterization of uracil-DNA glycosylase inhibitor protein. Role of specific carboxylic amino acids in complex formation with Escherichia coli uracil-DNA glycosylase. J Biol Chem. Aug. 22, 1997;272(34):21408-19.
Lutz et al., Postsymptomatic restoration of SMN rescues the disease phenotype in a mouse model of severe spinal muscular atrophy. J Clin Invest. Aug. 2011;121(8):3029-41. doi: 10.1172/JCI57291. Epub Jul. 25, 2011.
Lynch, Evolution of the mutation rate. Trends Genet. Aug. 2010;26(8):345-52. doi: 10.1016/j.tig.2010.05.003. Epub Jun. 30, 2010.
Lyons et al., Efficient Recognition of an Unpaired Lesion by a DNA Repair Glycosylase. J. Am. Chem. Soc., 2009;131(49):17742-3. DOI: 10.1021/ja908378y.
Lüke et al., Partial purification and characterization of the reverse transcriptase of the simian immunodeficiency virus TYO-7 isolated from an African green monkey. Biochemistry. Feb. 20, 1990;29(7):1764-9. doi: 10.1021/bi00459a015.
Ma et al., Human RAD52 interactions with replication protein A and the RAD51 presynaptic complex. J Biol Chem. Jul. 14, 2017;292(28):11702-11713. doi: 10.1074/jbc.M117.794545. Epub May 27, 2017.
Ma et al., Identification of pseudo attP sites for phage phiC31 integrase in bovine genome. Biochem Biophys Res Commun. Jul. 7, 2006;345(3):984-8. doi: 10.1016/j.bbrc.2006.04.145. Epub May 3, 2006.
Ma et al., In vitro protein engineering using synthetic tRNA(Ala) with different anticodons. Biochemistry. Aug. 10, 1993;32(31):7939-45.
Ma et al., PhiC31 integrase induces efficient site-specific recombination in the Capra hircus genome. DNA Cell Biol. Aug. 2014;33(8):484-91. doi: 10.1089/dna.2013.2124. Epub Apr. 22, 2014.
Ma et al., Single-Stranded DNA Cleavage by Divergent CRISPR-Cas9 Enzymes. Mol Cell. Nov. 5, 2015;60(3):398-407. doi: 10.1016/j.molcel.2015.10.030.
Ma et al., Targeted AID-mediated mutagenesis (TAM) enables efficient genomic diversification in mammalian cells. Nature Methods. Oct. 2016; 13:1029-35. doi: 10.1038/nmeth.4027 .
Maas et al., Identification and characterization of a human tRNA-specific adenosine deaminase related to the ADAR family of pre-mRNA editing enzymes. Proc Natl Acad Sci U S A. Aug. 3, 1999;96(16):8895-900. doi: 10.1073/pnas.96.16.8895.
Macbeth et al., Inositol hexakisphosphate is bound in the ADAR2 core and required for RNA editing. Science. Sep. 2, 2005;309(5740):1534-9. doi: 10.1126/science.1113150.
Macrae et al., Ribonuclease revisited: structural insights into ribonuclease III family enzymes. Curr Opin Struct Biol. Feb. 2007;17(1):138-45. doi: 10.1016/j.sbi.2006.12.002. Epub Dec. 27, 2006.
Madura et al., Structural basis for ineffective T-cell responses to MHC anchor residue-improved “heteroclitic” peptides. Eur J Immunol. Feb. 2015;45(2):584-91. doi: 10.1002/eji.201445114. Epub Dec. 28, 2014.
Maeder et al., CRISPR RNA-guided activation of endogenous human genes. Nat Methods. Oct. 2013;10(10):977-9. doi: 10.1038/nmeth.2598. Epub Jul. 25, 2013.
Maeder et al., Rapid “open-source” engineering of customized zinc-finger nucleases for highly efficient gene modification. Mol Cell. Jul. 25, 2008;31(2):294-301. doi:10.1016/j.molcel.2008.06.016.
Maeder et al., Robust, synergistic regulation of human gene expression using TALE activators. Nat Methods. Mar. 2013;10(3):243-5. doi: 10.1038/nmeth.2366. Epub Feb. 10, 2013.
Maerker et al., A novel Usher protein network at the periciliary reloading point between molecular transport machineries in vertebrate photoreceptor cells. Hum Mol Genet. Jan. 1, 2008;17(1):71-86. doi: 10.1093/hmg/ddm285. Epub Sep. 28, 2007.
Magin et al., Corf, the Rev/Rex homologue of HTDV/HERV-K, encodes an arginine-rich nuclear localization signal that exerts a trans-dominant phenotype when mutated. Virology. Aug. 15, 2000;274(1):11-6. doi: 10.1006/viro.2000.0438.
Mahfouz et al., De novo-engineered transcription activator-like effector (TALE) hybrid nuclease with novel DNA binding specificity creates double-strand breaks. Proc Natl Acad Sci U S A. Feb. 8, 2011;108(6):2623-8. doi: 10.1073/pnas.1019533108. Epub Jan. 24, 2011.
Maizels et al., Initiation of homologous recombination at DNA nicks. Nucleic Acids Res. Aug. 21, 2018;46(14):6962-6973. doi: 10.1093/nar/gky588.
Maji et al., A High-Throughput Platform to Identify Small-Molecule Inhibitors of CRISPR-Cas9. Cell. May 2, 2019;177(4):1067-1079.e19. doi: 10.1016/j.cell.2019.04.009.
Makarova et al., An updated evolutionary classification of CRISPR-Cas systems. Nat Rev Microbiol. Nov. 2015;13(11):722-36. doi: 10.1038/nrmicro3569. Epub Sep. 28, 2015.
Makarova et al., Classification and Nomenclature of CRISPR-Cas Systems: Where from Here? Crispr J. Oct. 2018;1(5):325-336. doi: 10.1089/crispr.2018.0033.
Makarova et al., Evolution and classification of the CRISPR-Cas systems. Nat Rev Microbiol. Jun. 2011;9(6):467-77. doi: 10.1038/nrmicro2577. Epub May 9, 2011.
Makarova et al., Prokaryotic homologs of Argonaute proteins are predicted to function as key components of a novel system of defense against mobile genetic elements. Biology Direct 2009;4:29. doi: 10.1186/1745-6150-4-29.
Makeyev et al., Evolutionary potential of an RNA virus. J Virol. Feb. 2004;78(4):2114-20.
Malashkevich et al., Crystal structure of tRNA adenosine deaminase TadA from Escherichia coli. Deposited: Mar. 10, 2005 Released: Feb. 21, 2006 doi:10.2210/pdb1z3a/pdb (2006).
Mali et al., Cas9 as a versatile tool for engineeringbiology. Nat Methods. Oct. 2013;10(10):957-63. doi: 10.1038/nmeth.2649.
Mali et al., CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat Biotechnol. Sep. 2013;31(9):833-8. doi: 10.1038/nbt.2675. Epub Aug. 1, 2013.
Mali et al., RNA-guided human genome engineering via Cas9. Science. Feb. 15, 2013;339(6121):823-6. doi: 10.1126/science.1232033. Epub Jan. 3, 2013.
Malito et al., Structural basis for lack of toxicity of the diphtheria toxin mutant CRM197. Proc Natl Acad Sci U S A. Apr. 3, 2012;109(14):5229-34. doi: 10.1073/pnas. 1201964109. Epub Mar. 19, 2012.
Mandal et al., Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. Cell Stem Cell. Nov. 6, 2014;15(5):643-52. doi: 10.1016/j.stem.2014.10.004. Epub Nov. 6, 2014.
Mandal et al., Riboswitches Control Fundamental Biochemical Pathways in Bacillus Subtilis and Other Bacteria. Cell. May 30, 2003;113(5):577-86. doi: 10.1016/s0092-8674(03)00391-x.
Mani et al., Design, engineering, and characterization of zinc finger nucleases. Biochem Biophys Res Commun. Sep. 23, 2005;335(2):447-57.
Marceau, Functions of single-strand DNA-binding proteins in DNA replication, recombination, and repair. Methods Mol Biol. 2012;922:1-21. doi: 10.1007/978-1-62703-032-8_1.
Maresca et al., Obligate ligation-gated recombination (ObLiGaRe): custom-designed nuclease-mediated targeted integration through nonhomologous end joining. Genome Res. Mar. 2013;23(3):539-46. Doi: 10.1101/gr.145441.112. Epub Nov. 14, 2012.
Marioni et al., DNA methylation age of blood predicts all-cause mortality in later life. Genome Biol. Jan. 30, 2015;16:25. doi: 10.1186/s13059-015-0584-6.
Marquart et al., Predicting base editing outcomes with an attention-based deep learning algorithm trained on high-throughput target library screeen. bioRxiv. Jul. 5, 2020. DOI: 10.1101/2020.07.05.186544. Retrieved from the Internet via https://www.biorxiv.org/content/10.1101/2020.07.05.186544v1.full.pdf lased accessed on Apr. 28, 2021.
Marraffini et al., CRISPR interference limits horizontal gene transfer in staphylococci by targeting DNA. Science. Dec. 19, 2008;322(5909):1843-5. doi: 10.1126/science.1165771.
Marsden et al., The Tumor-Associated Variant RAD51 G151D Induces a Hyper-Recombination Phenotype. PLoS Genet. Aug. 11, 2016;12(8):e1006208. doi: 10.1371/journal.pgen.1006208.
Martinez et al., Hypermutagenesis of RNA using human immunodeficiency virus type 1 reverse transcriptase and biased dNTP concentrations. Proc Natl Acad Sci U S A. Dec. 6, 1994;91(25):11787-91. doi: 10.1073/pnas.91.25.11787.
Martsolf et al., Complete trisomy 17p a relatively new syndrome. Ann Genet. 1988;31(3):172-4.
Martz, L., Nav-i-gating antibodies for pain. Science-Business eXchange. Jun. 12, 2014;7(662):1-2. doi: 10.1038/scibx.2014.662.
Maruyama et al., Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat Biotechnol. May 2015;33(5):538-42. doi: 10.1038/nbt.3190. Epub Mar. 23, 2015.
Marzec et al., Prime Editing: A New Way for Genome Editing. Trends Cell Biol. Apr. 2020;30(4):257-259. doi: 10.1016/j.tcb.2020.01.004. Epub Jan. 27, 2020.
Mascola et al., HIV-1 neutralizing antibodies: understanding nature's pathways. Immunol Rev. Jul. 2013;254(1):225-44. doi: 10.1111/imr.12075.
Mason et al., Non-enzymatic roles of human RAD51 at stalled replication forks. bioRxiv. Jul. 31, 2019; doi.org/10.1101/359380. 36 pages. bioRxiv preprint first posted online Jul. 31, 2019.
Mathys et al., Characterization of a self-splicing mini-intein and its conversion into autocatalytic N- and C-terminal cleavage elements: facile production of protein building blocks for protein ligation. Gene. Apr. 29, 1999;231(1-2):1-13. doi: 10.1016/s0378-1119(99)00103-1.
Matsuura et al., A gene essential for the site-specific excision of actinophage r4 prophage genome from the chromosome of a lysogen. J Gen Appl Microbiol. 1995;41(1):53-61.
Matthews, Structures of human ADAR2 bound to dsRNA reveal base-flipping mechanism and basis for site selectivity. Nat Struct Mol Biol. May 2016;23(5):426-33. doi: 10.1038/nsmb.3203. Epub Apr. 11, 2016.
May et al., Emergent lineages of mumps virus suggest the need for a polyvalent vaccine. Int J Infect Dis. Jan. 2018;66:1-4. doi: 10.1016/j.ijid.2017.09.024. Epub Oct. 4, 2017.
McCarroll et al., Copy-number variation and association studies of human disease. Nat Genet. Jul. 2007;39(7 Suppl):S37-42. doi: 10.1038/ng2080.
McDonald et al., Characterization of mutations at the mouse phenylalanine hydroxylase locus. Genomics. Feb. 1, 1997;39(3):402-5. doi: 10.1006/geno.1996.4508.
McInerney et al., Error Rate Comparison during Polymerase Chain Reaction by DNA Polymerase. Mol Biol Int. 2014;2014:287430. doi: 10.1155/2014/287430. Epub Aug. 17, 2014.
McKenna et al., Recording development with single cell dynamic lineage tracing. Development. Jun. 27, 2019;146(12):dev169730. doi: 10.1242/dev.169730.
McKenna et al., Whole-organism lineage tracing by combinatorial and cumulative genome editing. Science. Jul. 29, 2016;353(6298):aaf7907. doi: 10.1126/science.aaf7907. Epub May 26, 2016.
McNaughton et al., Mammalian cell penetration, siRNA transfection, and DNA transfection by supercharged proteins. Proc Natl Acad Sci U S A. Apr. 14, 2009;106(15):6111-6. doi: 10.1073/pnas.0807883106. Epub Mar. 23, 2009.
McVey et al., MMEJ repair of double-strand breaks (director's cut): deleted sequences and alternative endings. Trends Genet. Nov. 2008;24(11):529-38. doi: 10.1016/j.tig.2008.08.007. Epub Sep. 21, 2008.
Mead et al., A novel protective prion protein variant that colocalizes with kuru exposure. N Engl J Med. Nov. 19, 2009;361(21):2056-65. doi: 10.1056/NEJMoa0809716.
Mei et al., Recent Progress in CRISPR/Cas9 Technology. J Genet Genomics. Feb. 2, 2016;43(2):63-75. doi: 10.1016/j.jgg.2016.01.001. Epub Jan. 18, 2016.
Meinke et al., Cre Recombinase and Other Tyrosine Recombinases. Chem Rev. Oct. 26, 2016;116(20):12785-12820. doi: 10.1021/acs.chemrev.6b00077. Epub May 10, 2016.
Mendell et al., Single-Dose Gene-Replacement Therapy for Spinal Muscular Atrophy. N Engl J Med. Nov. 2, 2017;377(18): 1713-1722. doi: 10.1056/NEJMoa1706198.
Meng et al., Targeted gene inactivation in zebrafish using engineered zinc-finger nucleases. Nat Biotechnol. Jun. 2008;26(6):695-701. doi: 10.1038/nbt1398. Epub May 25, 2008.
Menéndez-Arias, Mutation rates and intrinsic fidelity of retroviral reverse transcriptases. Viruses. Dec. 2009;1(3):1137-65. doi: 10.3390/v1031137. Epub Dec. 4, 2009.
Mercer et al., Chimeric TALE recombinases with programmable DNA sequence specificity. Nucleic Acids Res. Nov. 2012;40(21):11163-72. doi: 10.1093/nar/gks875. Epub Sep. 26, 2012.
Mertens et al., Site-specific recombination in bacteriophage Mu: characterization of binding sites for the DNA invertase Gin. EMBO J. Apr. 1988;7(4):1219-27.
Meyer et al., Breathing life into polycations: functionalization with pH-responsive endosomolytic peptides and polyethylene glycol enables siRNA delivery. J Am Chem Soc. Mar. 19, 2008;130(11):3272-3. doi: 10.1021/ja710344v. Epub Feb. 21, 2008.
Meyer et al., Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell. Jun. 22, 2012;149(7):1635-46. doi: 10.1016/j.cell.2012.05.003. Epub May 17, 2012.
Meyer et al., Confirmation of a second natural preQ1 aptamer class in Streptococcaceae bacteria. RNA. Apr. 2008;14(4):685-95. doi: 10.1261/rna.937308. Epub Feb. 27, 2008.
Meyer et al., Library generation by gene shuffling. Curr Protoc Mol Biol. Jan. 6, 2014;105: Unit 15.12 . . . doi: 10.1002/0471142727.mb1512s105.
Meyer et al., Ribosome biogenesis factor Tsr3 is the aminocarboxypropyl transferase responsible for 18S rRNA hypermodification in yeast and humans. Nucleic Acids Res. May 19, 2016;44(9):4304-16. doi: 10.1093/nar/gkw244. Epub Apr. 15, 2016.
Meyer et al., The dynamic epitranscriptome: N6-methyladenosine and gene expression control. Nat Rev Mol Cell Biol. May 2014; 15(5):313-26. doi: 10.1038/nrm3785. Epub Apr. 9, 2014.
Michel et al., Mitochondrial class II introns encode proteins related to the reverse transcriptases of retroviruses. Nature. Aug. 15-21, 1985;316(6029):641-3. doi: 10.1038/316641a0.
Midoux et al., Chemical vectors for gene delivery: a current review on polymers, peptides and lipids containing histidine or imidazole as nucleic acids carriers. Br J Pharmacol. May 2009;157(2):166-78. doi: 10.1111/j.1476-5381.2009.00288.x.
Mihai et al., PTEN inhibition improves wound healing in lung epithelia through changes in cellular mechanics that enhance migration. Am J Physiol Lung Cell Mol Physiol. 2012;302(3):L287-L299.
Mijakovic et al., Bacterial single-stranded DNA-binding proteins are phosphorylated on tyrosine. Nucleic Acids Res. Mar. 20, 2006;34(5):1588-96. doi: 10.1093/nar/gkj514.
Miller et al., A Tale nuclease architecture for efficient genome editing. Nat Biotechnol. Feb. 2011;29(2):143-8. doi:10.1038/nbt.1755. Epub Dec. 22, 2010.
Miller et al., An improved zinc-finger nuclease architecture for highly specific genome editing. Nat Biotechnol. Jul. 2007;25(7):778-85. Epub Jul. 1, 2007.
Miller et al., Construction and properties of retrovirus packaging cells based on gibbon ape leukemia virus. J Virol. May 1991;65(5):2220-4. doi: 10.1128/JVI.65.5.2220-2224.1991.
Miller et al., Continuous evolution of SpCas9 variants compatible with non-G PAMs. Nat Biotechnol. Apr. 2020;38(4):471-481. doi: 10.1038/s41587-020-0412-8. Epub Feb. 10, 2020.
Miller et al., Phage-assisted continuous and non-continuous evolution. Nat Protoc. Dec. 2020; 15(12):4101-4127. doi: 10.1038/s41596-020-00410-3. Epub Nov. 16, 2020.
Miller, Human gene therapy comes of age. Nature. Jun. 11, 1992;357(6378):455-60. doi: 10.1038/357455a0.
Mills et al., Protein splicing in trans by purified N- and C-terminal fragments of the Mycobacterium tuberculosis RecA intein. Proc Natl Acad Sci U S A. Mar. 31, 1998;95(7):3543-8. doi: 10.1073/pnas.95.7.3543.
Minoche et al., Evaluation of genomic high-throughput sequencing data generated on Illumina HiSeq and genome analyzer systems. Genome Biol. Nov. 8, 2011;12(11):R112. doi: 10.1186/gb-2011-12-11-r112.
Minoretti et al., A W148R mutation in the human FOXD4 gene segregating with dilated cardiomyopathy, obsessive-compulsive disorder, and suicidality. Int J Mol Med. Mar. 2007;19(3):369-72.
Mir et al., Two Active Site Divalent Ions in the Crystal Structure of the Hammerhead Ribozyme Bound to a Transition State Analogue. Biochemistry . . . Feb. 2, 2016;55(4):633-6. doi: 10.1021/acs.biochem.5b01139. Epub Jan. 19, 2016.
Mir et al., Type II-C CRISPR-Cas9 Biology, Mechanism, and Application. ACS Chem Biol. Feb. 16, 2018;13(2):357-365. doi: 10.1021/acschembio.7b00855. Epub Dec. 20, 2017.
Mishina et al., Conditional gene targeting on the pure C57BL/6 genetic background. Neurosci Res. Jun. 2007;58(2):105-12. doi: 10.1016/j.neures.2007.01.004. Epub Jan. 18, 2007.
Mitani et al., Delivering therapeutic genes—matching approach and application. Trends Biotechnol. May 1993;11(5):162-6. doi: 10.1016/0167-7799(93)90108-L.
Mitton-Fry et al., Poly(A) tail recognition by a viral RNA element through assembly of a triple helix. Science. Nov. 26, 2010;330(6008):1244-7. doi: 10.1126/science.1195858.
Miyaoka et al., Systematic quantification of HDR and NHEJ reveals effects of locus, nuclease, and cell type on genome-editing. Sci Rep. Mar. 31, 2016;6:23549. doi: 10.1038/srep23549.
Moede et al., Identification of a nuclear localization signal, RRMKWKK, in the homeodomain transcription factor PDX-1. FEBS Lett. Nov. 19, 1999;461(3):229-34. doi: 10.1016/s0014-5793(99)01446-5.
Mohr et al., A Reverse Transcriptase-Cas1 Fusion Protein Contains a Cas6 Domain Required for Both CRISPR RNA Biogenesis and RNA Spacer Acquisition. Mol Cell. Nov. 15, 2018;72(4):700-714.e8. doi: 10.1016/j.molcel.2018.09.013. Epub Oct. 18, 2018. Including Supplemental Information.
Mohr et al., Thermostable group II intron reverse transcriptase fusion proteins and their use in cDNA synthesis and next-generation RNA sequencing. RNA. Jul. 2013;19(7):958-70. doi: 10.1261/rna.039743.113. Epub May 22, 2013.
Mojica et al., Intervening sequences of regularly spaced prokaryotic repeats derive from foreign genetic elements. J Mol Evol. Feb. 2005;60(2):174-82.
Mok et al., A bacterial cytidine deaminase toxin enables CRISPR-free mitochondrial base editing. Nature. Jul. 2020;583(7817):631-637. doi: 10.1038/s41586-020-2477-4. Epub Jul. 8, 2020.
Mol et al., Crystal structure and mutational analysis of human uracil-DNA glycosylase: structural basis for specificity and catalysis. Cell. Mar. 24, 1995;80(6):869-78. doi: 10.1016/0092-8674(95)90290-2.
Mol et al., Crystal structure of human uracil-DNA glycosylase in complex with a protein inhibitor: protein mimicry of DNA. Cell. Sep. 8, 1995;82(5):701-8.
Molla et al., CRISPR/Cas-Mediated Base Editing: Technical Considerations and Practical Applications. Trends Biotechnol. Oct. 2019;37(10):1121-1142. doi: 10.1016/j.tibtech.2019.03.008. Epub Apr. 14, 2019.
Monahan et al., Site-specific incorporation of unnatural amino acids into receptors expressed in Mammalian cells. Chem Biol. Jun. 2003;10(6):573-80.
Monani et al., A single nucleotide difference that alters splicing patterns distinguishes the SMA gene SMN1 from the copy gene SMN2. Hum Mol Genet. Jul. 1999;8(7):1177-83. doi: 10.1093/hmg/8.7.1177.
Monot et al., The specificity and flexibility of 11 reverse transcription priming at imperfect T-tracts. PLoS Genet. May 2013;9(5):e1003499. doi: 10.1371/journal.pgen.1003499. Epub May 9, 2013.
Montange et al., Structure of the S-adenosylmethionine riboswitch regulatory mRNA element. Nature. Jun. 29, 2006;441(7097):1172-5.
Moore et al., Improved somatic mutagenesis in zebrafish using transcription activator-like effector nucleases (TALENs). PloS One. 2012;7(5):e37877. Doi: 10.1371/journal.pone.0037877. Epub May 24, 2012.
Mootz et al., Conditional protein splicing: a new tool to control protein structure and function in vitro and in vivo. J Am Chem Soc. Sep. 3, 2003;125(35):10561-9.
Mootz et al., Protein splicing triggered by a small molecule. J Am Chem Soc. Aug. 7, 2002;124(31):9044-5 and Supporting Information. doi: 10.1021/ja026769o. 4 pages.
Mootz et al., Protein splicing triggered by a small molecule. J Am Chem Soc. Aug. 7, 2002;124(31):9044-5.
Morbitzer et al., Assembly of custom TALE-type DNA binding domains by modular cloning. Nucleic Acids Res. Jul. 2011;39(13):5790-9. doi: 10.1093/nar/gkr151. Epub Mar. 18, 2011.
Moreno-Mateos et al., CRISPRscan: designing highly efficient sgRNAs for CRISPR-Cas9 targeting in vivo. Nat Methods. Oct. 2015;12(10):982-8. doi: 10.1038/nmeth.3543. Epub Aug. 31, 2015.
Morita et al., The site-specific recombination system of actinophage TG1. FEMS Microbiol Lett. Aug. 2009;297(2):234-40. doi: 10.1111/j.1574-6968.2009.01683.x.
Morris et al., A peptide carrier for the delivery of biologically active proteins into mammalian cells. Nat Biotechnol. Dec. 2001;19(12):1173-6.
Moscou et al., A simple cipher governs DNA recognition by TAL effectors. Science. Dec. 11, 2009;326(5959):1501. doi: 10.1126/science.1178817.
Mougiakos et al., Characterizing a thermostable Cas9 for bacterial genome editing and silencing. Nat Commun. Nov. 21, 2017;8(1):1647. doi: 10.1038/s41467-017-01591-4.
Muir et al., Expressed protein ligation: a general method for protein engineering. Proc Natl Acad Sci U S A. Jun. 9, 1998;95(12):6705-10. doi: 10.1073/pnas.95.12.6705.
Muller et al., Nucleotide exchange and excision technology (NExT) DNA shuffling: a robust method for DNA fragmentation and directed evolution. Nucleic Acids Res. Aug. 1, 2005;33(13):e117. doi: 10.1093/nar/gni116. PMID: 16061932; PMCID: PMC1182171.
Muller, U.F., Design and Experimental Evolution of trans-Splicing Group I Intron Ribozymes. Molecules. Jan. 2, 2017;22(1):75. doi: 10.3390/molecules22010075.
Mullins et al., Transgenesis in nonmurine species. Hypertension. Oct. 1993;22(4):630-3.
Mumtsidu et al., Structural features of the single-stranded DNA-binding protein of Epstein-Barr virus. J Struct Biol. Feb. 2008;161(2):172-87. doi: 10.1016/j.jsb.2007.10.014. Epub Nov. 1, 2007.
Murphy, Phage recombinases and their applications. Adv Virus Res. 2012;83:367-414. doi: 10.1016/B978-0-12-394438-2.00008-6. Review.
Murray et al., Selective vulnerability of motor neurons and dissociation of pre- and post-synaptic pathology at the neuromuscular junction in mouse models of spinal muscular atrophy. Hum Mol Genet. Apr. 1, 2008;17(7):949-62. doi: 10.1093/hmg/ddm367. Epub Dec. 8, 2007.
Murugan et al., The Revolution Continues: Newly Discovered Systems Expand the CRISPR-Cas Toolkit. Mol Cell. Oct. 5, 2017;68(1):15-25. doi: 10.1016/j.molcel.2017.09.007.
Mussolino et al., A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. Nov. 2011;39(21):9283-93. Doi: 10.1093/nar/gkr597. Epub Aug. 3, 2011.
Mussolino et al., TALE nucleases: tailored genome engineering made easy. Curr Opin Biotechnol. Oct. 2012;23(5):644-50. doi: 10.1016/j.copbio.2012.01.013. Epub Feb. 17, 2012.
Muzyczka et al., Adeno-associated virus (AAV) vectors: will they work? J Clin Invest. Oct. 1994;94(4):1351. doi: 10.1172/JCI117468.
Myerowitz et al., The major defect in Ashkenazi Jews with Tay-Sachs disease is an insertion in the gene for the alpha-chain of beta-hexosaminidase. J Biol Chem. Dec. 15, 1988;263(35):18587-9.
Myers et al., Insulin signal transduction and the IRS proteins. Annu Rev Pharmacol Toxicol. 1996;36:615-58. doi: 10.1146/annurev.pa.36.040196.003151.
Nabel et al., Direct gene transfer for immunotherapy and immunization. Trends Biotechnol. May 1993;11(5):211-5. doi: 10.1016/0167-7799(93)90117-R.
Nahar et al., A G-quadruplex motif at the 3′ end of sgRNAs improves CRISPR-Cas9 based genome editing efficiency. Chem Commun (Camb). Mar. 7, 2018;54(19):2377-2380. doi: 10.1039/c7cc08893k. Epub Feb. 16, 2018.
Nahvi et al., Coenzyme B12 riboswitches are widespread genetic control elements in prokaryotes. Nucleic Acids Res. Jan. 2, 2004;32(1):143-50.
Nakade et al., Microhomology-mediated end-joining-dependent integration of donor DNA in cells and animals using TALENs and CRISPR/Cas9. Nat Commun. Nov. 20, 2014;5:5560. doi: 10.1038/ncomms6560.
Nakamura et al., Codon usage tabulated from international DNA sequence databases: status for the year 2000. Nucleic Acids Res. Jan. 1, 2000;28(1):292. doi: 10.1093/nar/28.1.292.
Naorem et al., DGR mutagenic transposition occurs via hypermutagenic reverse transcription primed by nicked template RNA. Proc Natl Acad Sci U S A. Nov. 21, 2017;114(47):E10187-E10195. doi: 10.1073/pnas.1715952114. Epub Nov. 6, 2017.
Narayanan et al., Clamping down on weak terminal base pairs: oligonucleotides with molecular caps as fidelity-enhancing elements at the 5′-and 3′-terminal residues. Nucleic Acids Res. May 20, 2004;32(9):2901-11. Print 2004.
Navaratnam et al., An overview of cytidine deaminases. Int J Hematol. Apr. 2006;83(3):195-200.
NCBI Reference Sequence: NM_002427.3. Wu et al., May 3, 2014. 5 pages.
Neel et al., Riboswitches: Classification, function and in silico approach, International Journal of Pharma Sciences and Research. 2010;1(9):409-420.
Nelson et al., Engineered pegRNAs improve prime editing efficiency. Nat Biotechnol. Oct. 4, 2021. doi: 10.1038/s41587-021-01039-7. Epub ahead of print. Erratum in: Nat Biotechnol. Dec. 8, 2021. 14 pages.
Nelson et al., Filamentous phage DNA cloning vectors: a noninfective mutant with a nonpolar deletion in gene III. Virology. 1981; 108(2): 338-50.
Nelson et al., In vivo genome editing improves muscle function in a mouse model of Duchenne muscular dystrophy. Science. Jan. 22, 2016;351(6271):403-7. doi: 10.1126/science.aad5143. Epub Dec. 31, 2015.
Nelson et al., The unstable repeats—three evolving faces of neurological disease. Neuron. Mar. 6, 2013;77(5):825-43. doi: 10.1016/j.neuron.2013.02.022.
Nern et al., Multiple new site-specific recombinases for use in manipulating animal genomes. Proc Natl Acad Sci U S A. Aug. 23, 2011;108(34):14198-203. doi: 10.1073/pnas.1111704108. Epub Aug. 9, 2011.
Newby et al., Base editing of haematopoietic stem cells rescues sickle cell disease in mice. Nature. Jun. 2, 2021. doi: 10.1038/s41586-021-03609-w. Epub ahead of print.
Nguyen et al., Evolutionary drivers of thermoadaptation in enzyme catalysis. Science. Jan. 20, 2017;355(6322):289-294. doi: 10.1126/science.aah3717. Epub Dec. 22, 2016.
Nguyen et al., IQ-TREE: a fast and effective stochastic algorithm for estimating maximum-likelihood phylogenies. Mol Biol Evol. Jan. 2015;32(1):268-74. doi: 10.1093/molbev/msu300. Epub Nov. 3, 2014.
Ni et al., A PCSK9-binding antibody that structurally mimics the EGF(A) domain of LDL-receptor reduces LDL cholesterol in vivo. J Lipid Res. 2011;52:76-86.
Ni et al., Nucleic acid aptamers: clinical applications and promising new horizons. Curr Med Chem. 2011;18(27):4206-14. Review.
Nishida et al., Targeted nucleotide editing using hybrid prokaryotic and vertebrate adaptive immune systems. Science. Sep. 16, 2016;353(6305):1248. pii: aaf8729. doi: 10.1126/science.aaf8729. Epub Aug. 4, 2016.
Nishikura, Functions and regulation of RNA editing by ADAR deaminases. Annu Rev Biochem. 2010;79:321-349. doi:10.1146/annurev-biochem-060208-105251.
Nishimasu et al., Crystal structure of Cas9 in complex with guide RNA and target DNA. Cell. Feb. 27, 2014;156(5):935-49. doi: 10.1016/j.cell.2014.02.001. Epub Feb. 13, 2014.
Nishimasu et al., Crystal Structure of Staphylococcus aureus Cas9. Cell. Aug. 27, 2015;162(5):1113-26. doi: 10.1016/j.cell.2015.08.007.
Nishimasu et al., Engineered CRISPR-Cas9 nuclease with expanded targeting space. Science. Sep. 21, 2018;361(6408):1259-1262. doi: 10.1126/science.aas9129. Epub Aug. 30, 2018.
Noack et al., Epitranscriptomics: A New Regulatory Mechanism of Brain Development and Function. Front Neurosci. Feb. 20, 2018;12:85. doi: 10.3389/fnins.2018.00085. 9 pages.
Nomura et al., Controlling Mammalian Gene Expression by Allosteric Hepatitis Delta Virus Ribozymes. ACS Synth Biol. Dec. 20, 2013;2(12):684-9. doi: 10.1021/sb400037a. Epub May 22, 2013.
Nomura et al., Synthetic mammalian riboswitches based on guanine aptazyme. Chem Commun (Camb). Jul. 21, 2012;48(57):7215-7. doi: 10.1039/c2cc33140c. Epub Jun. 13, 2012.
Noris et al., A phenylalanine-55 to serine amino-acid substitution in the human glycoprotein IX leucine-rich repeat is associated with Bernard-Soulier syndrome. Br J Haematol. May 1997;97(2):312-20.
Nottingham et al., RNA-seq of human reference RNA samples using a thermostable group II intron reverse transcriptase. RNA. Apr. 2016;22(4):597-613. doi: 10.1261/rna.055558.115. Epub Jan. 29, 2016.
Nowak et al., Characterization of single-stranded DNA-binding proteins from the psychrophilic bacteria Desulfotalea psychrophila, Flavobacterium psychrophilum, Psychrobacter arcticus, Psychrobacter cryohalolentis, Psychromonas ingrahamii, Psychroflexus torquis, and Photobacterium profundum. BMC Microbiol. Apr. 14, 2014;14:91. doi: 10.1186/1471-2180-14-91.
Nowak et al., Guide RNA Engineering for Versatile Cas9 Functionality. Nucleic Acids Res. Nov. 16, 2016;44(20):9555-9564. doi: 10.1093/nar/gkw908. Epub Oct. 12, 2016.
Nowak et al., Structural analysis of monomeric retroviral reverse transcriptase in complex with an RNA/DNA hybrid. Nucleic Acids Res. Apr. 1, 2013;41(6):3874-87. doi: 10.1093/nar/gkt053. Epub Feb. 4, 2013.
Numrych et al., A comparison of the effects of single-base and triple-base changes in the integrase arm-type binding sites on the site-specific recombination of bacteriophage lambda. Nucleic Acids Res. Jul. 11, 1990;18(13):3953-9. doi: 10.1093/nar/18.13.3953.
Nyerges et al., A highly precise and portable genome engineering method allows comparison of mutational effects across bacterial species. Proc Natl Acad Sci U S A. Mar. 1, 2016;113(9):2502-7. doi: 10.1073/pnas.1520040113. Epub Feb. 16, 2016.
O'Connell et al., Programmable RNA recognition and cleavage by CRISPR/Cas9. Nature. Dec. 11, 2014;516(7530):263-6. doi: 10.1038/nature13769. Epub Sep. 28, 2014.
O'Maille et al., Structure-based combinatorial protein engineering (SCOPE). J Mol Biol. Aug. 23, 2002;321(4):677-91.
Oakes et al., CRISPR-Cas9 Circular Permutants as Programmable Scaffolds for Genome Modification. Cell. Jan. 10, 2019;176(1-2):254-267.e16. doi: 10.1016/j.cell.2018.11.052.
Oakes et al., Profiling of engineering hotspots identifies an allosteric CRISPR-Cas9 switch. Nat Biotechnol. Jun. 2016;34(6):646-51. doi: 10.1038/nbt.3528. Epub May 2, 2016.
Oakes et al., Protein engineering of Cas9 for enhanced function. Methods Enzymol. 2014;546:491-511.
Odsbu et al., Specific N-terminal interactions of the Escherichia coli SeqA protein are required to form multimers that restrain negative supercoils and form foci. Genes Cells. Nov. 2005; 10(11):1039-49.
Oeemig et al., Solution structure of DnaE intein from Nostoc punctiforme: structural basis for the design of a new split intein suitable for site-specific chemical modification. FEBS Lett. May 6, 2009;583(9):1451-6.
Offord, Advances in Genome Editing. The Scientist, Apr. 20, 2016. http://www.the-scientist.com/?articles.view/articleNo/45903/title/Advances-in-Genome-Editing/.
Oh et al., Positional cloning of a gene for Hermansky-Pudlak syndrome, a disorder of cytoplasmic organelles. Nat Genet. Nov. 1996;14(3):300-6. doi: 10.1038/ng1196-300.
Ohe et al., Purification and properties of xanthine dehydrogenase from Streptomyces cyanogenus. J Biochem. Jul. 1979;86(1):45-53.
Olivares et al., Site-specific genomic integration produces therapeutic Factor IX levels in mice. Nat Biotechnol. Nov. 2002;20(11):1124-8. doi: 10.1038/nbt753. Epub Oct. 15, 2002.
Olorunniji et al., Purification and In Vitro Characterization of Zinc Finger Recombinases. Methods Mol Biol. 2017;1642:229-245. doi: 10.1007/978-1-4939-7169-5_15.
Olorunniji et al., Site-specific recombinases: molecular machines for the Genetic Revolution. Biochem J. Mar. 15, 2016;473(6):673-84. doi: 10.1042/BJ20151112.
Olorunniji et al., Synapsis and catalysis by activated Tn3 resolvase mutants. Nucleic Acids Res. Dec. 2008;36(22):7181-91. doi: 10.1093/nar/gkn885. Epub Nov. 10, 2008.
Orlando et al., Zinc-finger nuclease-driven targeted integration into mammalian genomes using donors with limited chromosomal homology. Nucleic Acids Res. Aug. 2010;38(15):e152. doi: 10.1093/nar/gkq512. Epub Jun. 8, 2010.
Orthwein et al., A mechanism for the suppression of homologous recombination in G1 cells. Nature. Dec. 17, 2015;528(7582):422-6. doi: 10.1038/nature16142. Epub Dec. 9, 2015.
Ortiz-Urda et al., Stable nonviral genetic correction of inherited human skin disease. Nat Med. Oct. 2002;8(10):1166-70. doi: 10.1038/nm766. Epub Sep. 16, 2002. Erratum in: Nat Med. Feb. 2003;9(2):237.
Osborn et al., Base Editor Correction of COL7A1 in Recessive Dystrophic Epidermolysis Bullosa Patient-Derived Fibroblasts and iPSCs. J Invest Dermatol. Feb. 2020;140(2):338-347.e5. doi: 10.1016/j.jid.2019.07.701. Epub Aug. 19, 2019.
Osborn et al., TALEN-based gene correction for epidermolysis bullosa. Mol Ther. Jun. 2013;21(6):1151-9. doi: 10.1038/mt.2013.56. Epub Apr. 2, 2013.
Ostermeier et al., A combinatorial approach to hybrid enzymes independent of DNA homology. Nat Biotechnol. Dec. 1999;17(12):1205-9.
Ostertag et al., Biology of mammalian L1 retrotransposons. Annu Rev Genet. 2001;35:501-38. doi: 10.1146/annurev.genet.35.102401.091032.
Otomo et al., Improved segmental isotope labeling of proteins and application to a larger protein. J Biomol NMR. Jun. 1999;14(2):105-14. doi: 10.1023/a:1008308128050.
Otomo et al., NMR observation of selected segments in a larger protein: central-segment isotope labeling through intein-mediated ligation. Biochemistry. Dec. 7, 1999;38(49):16040-4. doi: 10.1021/bi991902j.
Ottesen, ISS-N1 makes the First FDA-approved Drug for Spinal Muscular Atrophy. Transl Neurosci. Jan. 26, 2017;8:1-6. doi: 10.1515/tnsci-2017-0001.
Otto et al., The probability of fixation in populations of changing size. Genetics. Jun. 1997; 146(2):723-33.
Packer et al., Methods for the directed evolution of proteins. Nat Rev Genet. Jul. 2015;16(7):379-94. doi: 10.1038/nrg3927. Epub Jun. 9, 2015.
Packer et al., Phage-assisted continuous evolution of proteases with altered substrate specificity. Nat Commun. Oct. 16, 2017;8(1):956. doi: 10.1038/s41467-017-01055-9.
Paige et al., RNA mimics of green fluorescent protein. Science. Jul. 29, 2011;333(6042):642-6. doi:10.1126/science.1207339.
Paiva et al., Targeted protein degradation: elements of PROTAC design. Curr Opin Chem Biol. Jun. 2019;50:111-119. doi: 10.1016/j.cbpa.2019.02.022. Epub Apr. 17, 2019.
Pan et al., Biological and biomedical applications of engineered nucleases. Mol Biotechnol. Sep. 2013;55(1):54-62. doi: 10.1007/s12033-012-9613-9.
Paquet et al., Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature. May 5, 2016;533(7601):125-9. doi: 10.1038/nature17664. Epub Apr. 27, 2016.
Parente et al., Advances in spinal muscular atrophy therapeutics. Ther Adv Neurol Disord. Feb. 5, 2018;11:1756285618754501. doi: 10.1177/1756285618754501. 13 pages.
Park et al., Digenome-seq web tool for profiling CRISPR specificity. Nat Methods. May 30, 2017;14(6):548-549. doi: 10.1038/nmeth.4262.
Park et al., Highly efficient editing of the β-globin gene in patient-derived hematopoietic stem and progenitor cells to treat sickle cell disease. Nucleic Acids Res. Sep. 5, 2019;47(15):7955-7972. doi: 10.1093/nar/gkz475.
Park et al., Sendai virus, an RNA virus with no risk of genomic integration, delivers CRISPR/Cas9 for efficient gene editing. Mol Ther Methods Clin Dev. Aug. 24, 2016;3:16057. doi: 10.1038/mtm.2016.57.
Parker et al., Admixture mapping identifies a quantitative trait locus associated with FEV1/FVC in the COPDGene Study. Genet Epidemiol. Nov. 2014;38(7):652-9. doi: 10.1002/gepi.21847. Epub Aug. 11, 2014.
Passini et al., Antisense oligonucleotides delivered to the mouse CNS ameliorate symptoms of severe spinal muscular atrophy. Sci Transl Med. Mar. 2, 2011;3(72):72ra18. doi: 10.1126/scitranslmed.3001777.
Patel et al., Flap endonucleases pass 5′-flaps through a flexible arch using a disorder-thread-order mechanism to confer specificity for free 5′-ends. Nucleic Acids Res. May 2012;40(10):4507-19. doi: 10.1093/nar/gks051. Epub Feb. 8, 2012.
Pattanayak et al., Determining the specificities of TALENs, Cas9, and other genome-editing enzymes. Methods Enzymol. 2014;546:47-78. doi: 10.1016/B978-0-12-801185-0.00003-9.
Pattanayak et al., High-throughput profiling of off-target DNA cleavage reveals RNA-programmed Cas9 nuclease specificity. Nat Biotechnol. Sep. 2013;31(9):839-43. doi: 10.1038/nbt.2673. Epub Aug. 11, 2013.
Pattanayak et al., Revealing off-target cleavage specificities of zinc-finger nucleases by in vitro selection. Nat Methods. Aug. 7, 2011;8(9):765-70. doi: 10.1038/nmeth.1670.
Pavletich et al., Zinc finger-DNA recognition: crystal structure of a Zif268-DNA complex at 2.1 A. Science. May 10, 1991;252(5007):809-17.
Pawson et al., Protein phosphorylation in signaling—50 years and counting. Trends Biochem Sci. Jun. 2005;30(6):286-90. doi: 10.1016/j.tibs.2005.04.013.
Pearl, Structure and function in the uracil-DNA glycosylase superfamily. Mutat Res. Aug. 30, 2000;460(3-4):165-81.
Peck et al., Directed evolution of a small-molecule-triggered intein with improved splicing properties in mammalian cells. Chem Biol. May 27, 2011;18(5):619-30. doi: 10.1016/j.chembiol.2011.02.014.
Pellegrini et al., Insights into DNA recombination from the structure of a RAD51-BRCA2 complex. Nature. Nov. 21, 2002;420(6913):287-93. doi: 10.1038/nature01230. Epub Nov. 10, 2002.
Pellenz et al., New human chromosomal safe harbor sites for genome engineering with CRISPR/Cas9, TAL effector and homing endonucleases. Aug. 20, 2018. bioRxiv doi: https://doi.org/10.1101/396390.
Pelletier, CRISPR-Cas systems for the study of the immune function. Nov. 15, 2016. https://doi.org/10.1002/9780470015902.a0026896.
Pendse et al., Exon 13-skipped USH2A protein retains functional integrity in mice, suggesting an exo-skipping therapeutic approach to treat USH2A-associated disease. bioRxiv preprint. Feb. 4, 2020. Retrieved from www.biorxiv.org. doi: 10.1101/2020.02.04.934240. 34 pages.
Pendse et al., In Vivo Assessment of Potential Therapeutic Approaches for USH2A-Associated Diseases. Adv Exp Med Biol. 2019;1185:91-96. doi: 10.1007/978-3-030-27378-1_15.
Pennisi et al., The CRISPR craze. Science. Aug. 23, 2013;341(6148):833-6. doi: 10.1126/science.341.6148.833.
Pennisi et al., The tale of the TALEs. Science. Dec. 14, 2012;338(6113):1408-11. doi: 10.1126/science.338.6113.1408.
Perach et al., Catalytic features of the recombinant reverse transcriptase of bovine leukemia virus expressed in bacteria. Virology. Jun. 20, 1999;259(1): 176-89. doi: 10.1006/viro.1999.9761.
Perez et al., Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol. Jul. 2008;26(7):808-16. Doi: 10.1038/nbt1410. Epub Jun. 29, 2008.
Perez-Palma et al., Simple ClinVar: an interactive web server to explore and retrieve gene and disease variants aggregated in ClinVar database. Nucleic Acids Res. Jul. 2, 2019;47(W1):W99-W105. doi: 10.1093/nar/gkz411.
Perez-Pinera et al., Advances in targeted genome editing. Curr Opin Chem Biol. Aug. 2012; 16(3-4):268-77. doi: 10.1016/j.cbpa.2012.06.007. Epub Jul. 20, 2012.
Perez-Pinera et al., RNA-guided gene activation by CRISPR-Cas9-based transcription factors. Nat Methods. Oct. 2013;10(10):973-6. doi: 10.1038/nmeth.2600. Epub Jul. 25, 2013.
Perler et al., Protein splicing and autoproteolysis mechanisms. Curr Opin Chem Biol. Oct. 1997; 1(3):292-9. doi: 10.1016/s1367-5931(97)80065-8.
Perler et al., Protein splicing elements: inteins and exteins—a definition of terms and recommended nomenclature. Nucleic Acids Res. Apr. 11, 1994;22(7):1125-7. doi: 10.1093/nar/22.7.1125.
Perler, InBase, the New England Biolabs Intein Database. Nucleic Acids Res. Jan. 1, 1999;27(1):346-7. doi: 10.1093/nar/27.1.346.
Perler, Protein splicing of inteins and hedgehog autoproteolysis: structure, function, and evolution. Cell. Jan. 9, 1998;92(1):1-4. doi: 10.1016/s0092-8674(00)80892-2.
Perreault et al., Mixed deoxyribo- and ribo-oligonucleotides with catalytic activity. Nature. Apr. 5, 1990;344(6266):565-7. doi: 10.1038/344565a0.
Petek et al., Frequent endonuclease cleavage at off-target locations in vivo. Mol Ther. May 2010; 18(5):983-6. Doi: 10.1038/mt.2010.35. Epub Mar. 9, 2010.
Petersen-Mahrt et al., AID mutates E. coli suggesting a DNA deamination mechanism for antibody diversification. Nature. Jul. 4, 2002;418(6893):99-103.
Petolino et al., Editing Plant Genomes: a new era of crop improvement. Plant Biotechnol J. Feb. 2016;14(2):435-6. doi: 10.1111/pbi.12542.
Peyrottes et al., Oligodeoxynucleoside phosphoramidates (P-NH2): synthesis and thermal stability of duplexes with DNA and RNA targets. Nucleic Acids Res. May 15, 1996;24(10):1841-8.
Pfeiffer et al., Mechanisms of DNA double-strand break repair and their potential to induce chromosomal aberrations. Mutagenesis. Jul. 2000;15(4):289-302. doi: 10.1093/mutage/15.4.289.
Phillips, The challenge of gene therapy and DNA delivery. J Pharm Pharmacol. Sep. 2001;53(9):1169-74.
Pickart et al., Ubiquitin: structures, functions, mechanisms. Biochim Biophys Acta. Nov. 29, 2004;1695(1-3):55-72. doi: 10.1016/j.bbamcr.2004.09.019.
Pieken et al., Kinetic characterization of ribonuclease-resistant 2′-modified hammerhead ribozymes. Science. Jul. 19, 1991;253(5017):314-7. doi: 10.1126/science.1857967.
Pinkert et al., An albumin enhancer located 10 kb upstream functions along with its promoter to direct efficient, liver-specific expression in transgenic mice. Genes Dev. May 1987;1(3):268-76. doi: 10.1101/gad.1.3.268.
Pirakitikulr et al., PCRless library mutagenesis via oligonucleotide recombination in yeast. Protein Sci. Dec. 2010; 19(12):2336-46. doi: 10.1002/pro.513.
Plasterk et al., DNA inversions in the chromosome of Escherichia coli and in bacteriophage Mu: relationship to other site-specific recombination systems. Proc Natl Acad Sci U S A. Sep. 1983;80(17):5355-8.
Plosky et al., CRISPR-Mediated Base Editing without DNA Double-Strand Breaks. Mol Cell. May 19, 2016;62(4):477-8. doi: 10.1016/j.molcel.2016.05.006.
Pluciennik et al., PCNA function in the activation and strand direction of MutLα endonuclease in mismatch repair. Proc Natl Acad Sci U S A. Sep. 14, 2010;107(37):16066-71. doi: 10.1073/pnas.1010662107. Epub Aug. 16, 2010.
Poller et al., A leucine-to-proline substitution causes a defective alpha 1-antichymotrypsin allele associated with familial obstructive lung disease. Genomics. Sep. 1993;17(3):740-3.
Popp et al., Sortagging: a versatile method for protein labeling. Nat Chem Biol. Nov. 2007;3(11):707-8. doi: 10.1038/nchembio.2007.31. Epub Sep. 23, 2007.
Porensky et al., A single administration of morpholino antisense oligomer rescues spinal muscular atrophy in mouse. Hum Mol Genet. Apr. 1, 2012;21(7):1625-38. doi: 10.1093/hmg/ddr600. Epub Dec. 20, 2011.
Porteus, Design and testing of zinc finger nucleases for use in mammalian cells. Methods Mol Biol. 2008;435:47-61. doi: 10.1007/978-1-59745-232-8_4.
Posnick et al., Imbalanced base excision repair increases spontaneous mutation and alkylation sensitivity in Escherichia coli. J Bacteriol. Nov. 1999;181(21):6763-71.
Pospísilová et al., Hydrolytic cleavage of N6-substituted adenine derivatives by eukaryotic adenine and adenosine deaminases. Biosci Rep. 2008;28(6):335-347. doi:10.1042/BSR20080081.
Pourcel et al., CRISPR elements in Yersinia pestis acquire new repeats by preferential uptake of bacteriophage DNA, and provide additional tools for evolutionary studies. Microbiology. Mar. 2005;151(Pt 3):653-63.
Prasad et al., Rev1 is a base excision repair enzyme with 5′-deoxyribose phosphate lyase activity. Nucleic Acids Res. Dec. 15, 2016;44(22):10824-10833. doi: 10.1093/nar/gkw869. Epub Sep. 28, 2016.
Prasad et al., Visualizing the assembly of human Rad51 filaments on double-stranded DNA. J Mol Biol. Oct. 27, 2006;363(3):713-28. doi: 10.1016/j.jmb.2006.08.046. Epub Aug. 22, 2006.
Prashant et al., CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nature Biotechnology 2013;31(9):833-8.
Prorocic et al., Zinc-finger recombinase activities in vitro. Nucleic Acids Res. Nov. 2011;39(21):9316-28. doi: 10.1093/nar/gkr652. Epub Aug. 17, 2011.
Proudfoot et al., Zinc finger recombinases with adaptable DNA sequence specificity. PLoS One. Apr. 29, 2011;6(4):e19537. doi: 10.1371/journal.pone.0019537.
Pruschy et al., Mechanistic studies of a signaling pathway activated by the organic dimerizer FK1012. Chem Biol. Nov. 1994;1(3):163-72. doi: 10.1016/1074-5521(94)90006-x.
Prykhozhij et al., CRISPR multitargeter: a web tool to find common and unique CRISPR single guide RNA targets in a set of similar sequences. PLoS One. Mar. 5, 2015;10(3):e0119372. doi: 10.1371/journal.pone.0119372. eCollection 2015.
Pu et al., Evolution of a split RNA polymerase as a versatile biosensor platform. Nat Chem Biol. Apr. 2017;13(4):432-438. doi: 10.1038/nchembio.2299. Epub Feb. 13, 2017.
Putnam et al., Protein mimicry of DNA from crystal structures of the uracil-DNA glycosylase inhibitor protein and its complex with Escherichia coli uracil-DNA glycosylase. J Mol Biol. Mar. 26, 1999;287(2):331-46.
Qi et al., Engineering naturally occurring trans-acting non-coding RNAs to sense molecular signals. Nucleic Acids Res. Jul. 2012;40(12):5775-86. doi: 10.1093/nar/gks168. Epub Mar. 1, 2012.
Qi et al., Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell. Feb. 28, 2013;152(5):1173-83. doi: 10.1016/j.cell.2013.02.022.
Qu et al., Global mapping of binding sites for phic31 integrase in transgenic maden-darby bovine kidney cells using ChIP-seq. Hereditas. Jan. 14, 2019;156:3. doi: 10.1186/s41065-018-0079-z.
Queen et al., Immunoglobulin gene transcription is activated by downstream sequence elements. Cell. Jul. 1983;33(3):741-8. doi: 10.1016/0092-8674(83)90016-8.
Radany et al., Increased spontaneous mutation frequency in human cells expressing the phage PBS2-encoded inhibitor of uracil-DNA glycosylase. Mutat Res. Sep. 15, 2000;461(1):41-58. doi: 10.1016/s0921-8777(00)00040-9.
Raghavan et al., Abstract 27: Therapeutic Targeting of Human Lipid Genes with in vivo CRISPR-Cas9 Genome Editing. Oral Abstract Presentations: Lipoprotein Metabolism and Therapeutic Targets. Arterioscler THromb Vasc Biol. 2015;35(Suppl. 1):Abstract 27. 5 pages.
Raillard et al., Targeting sites within HIV-1 cDNA with a DNA-cleaving ribozyme. Biochemistry. Sep. 10, 1996;35(36):11693-701. doi: 10.1021/bi960845g.
Raina et al., PROTAC-induced BET protein degradation as a therapy for castration-resistant prostate cancer. Proc Natl Acad Sci U S A. Jun. 28, 2016;113(26):7124-9. doi: 10.1073/pnas.1521738113. Epub Jun. 6, 2016.
Rajagopal et al., High-throughput mapping of regulatory DNA. Nat Biotechnol. Feb. 2016;34(2):167-74. doi: 10.1038/nbt.3468. Epub Jan. 25, 2016.
Rakonjac et al., Roles of PIII in filamentous phage assembly. J Mol Biol. 1998; 282(1)25-41.
Ramakrishna et al., Gene disruption by cell-penetrating peptide-mediated delivery of Cas9 protein and guide RNA. Genome Res. Jun. 2014;24(6):1020-7. doi: 10.1101/gr.171264.113. Epub Apr. 2, 2014.
Ramamurthy et al., Identification of immunogenic B-cell epitope peptides of rubella virus El glycoprotein towards development of highly specific immunoassays and/or vaccine. Conference Abstract. 2019.
Ramirez et al., Engineered zinc finger nickases induce homology-directed repair with reduced mutagenic effects. Nucleic Acids Res. Jul. 2012;40(12):5560-8. doi: 10.1093/nar/gks179. Epub Feb. 28, 2012.
Ramirez et al., Unexpected failure rates for modular assembly of engineered zinc fingers. Nat Methods. May 2008;5(5):374-5. Doi: 10.1038/nmeth0508-374.
Ramos et al., Age-dependent SMN expression in disease-relevant tissue and implications for SMA treatment. J Clin Invest. Nov. 1, 2019;129(11):4817-4831. doi: 10.1172/JCI124120.
Ran et al., Double Nicking by RNA-guided CRISPR Cas9 for Enhanced Genome Editing Specificity. Cell. Sep. 12, 2013;154(6):1380-9. doi: 10.1016/j.cell.2013.08.021. Epub Aug. 29, 2013.
Ran et al., Genome engineering using the CRISPR-Cas9 system. Nat Protoc. Nov. 2013;8(11):2281-308. doi: 10.1038/nprot.2013.143. Epub Oct. 24, 2013.
Ran et al., In vivo genome editing using Staphylococcus aureus Cas9. Nature. Apr. 9, 2015;520(7546):186-91. doi: 10.1038/nature14299. Epub Apr. 1, 2015.
Ranzau et al., Genome, Epigenome, and Transcriptome Editing via Chemical Modification of Nucleobases in Living Cells. Biochemistry. Feb. 5, 2019;58(5):330-335. doi: 10.1021/acs.biochem.8b00958. Epub Dec. 12, 2018.
Rashel et al., A novel site-specific recombination system derived from bacteriophage phiMR11. Biochem Biophys Res Commun. Apr. 4, 2008;368(2):192-8. doi: 10.1016/j.bbrc.2008.01.045. Epub Jan. 22, 2008.
Rasila et al., Critical evaluation of random mutagenesis by error-prone polymerase chain reaction protocols, Escherichia coli mutator strain, and hydroxylamine treatment. Anal Biochem. May 1, 2009;388(1):71-80. doi: 10.1016/j.ab.2009.02.008. Epub Feb. 10, 2009.
Raskin et al., Substitution of a single bacteriophage T3 residue in bacteriophage T7 RNA polymerase at position 748 results in a switch in promoter specificity. J Mol Biol. Nov. 20, 1992;228(2):506-15.
Raskin et al., T7 RNA polymerase mutants with altered promoter specificities. Proc Natl Acad Sci U S A. Apr. 15, 1993;90(8):3147-51.
Rath et al., Fidelity of end joining in mammalian episomes and the impact of Metnase on joint processing. BMC Mol Biol. Mar. 22, 2014;15:6. doi: 10.1186/1471-2199-15-6.
Rauch et al., Programmable RNA Binding Proteins for Imaging and Therapeutics. Biochemistry. Jan. 30, 2018;57(4):363-364. doi: 10.1021/acs.biochem.7b01101. Epub Nov. 17, 2017.
Ravishankar et al., X-ray analysis of a complex of Escherichia coli uracil DNA glycosylase (EcUDG) with a proteinaceous inhibitor. The structure elucidation of a prokaryotic UDG. Nuclei Acids Res. 26 (21): 4880-4887 (1998).
Ray et al., A compendium of RNA-binding motifs for decoding gene regulation. Nature. Jul. 11, 2013;499(7457):172-7. doi: 10.1038/nature12311.
Ray et al., Homologous recombination: ends as the means. Trends Plant Sci. Oct. 2002;7(10):435-40.
Rebar et al., Phage display methods for selecting zinc finger proteins with novel DNA-binding specificities. Methods Enzymol. 1996;267:129-49.
Rebuzzini et al., New mammalian cellular systems to study mutations introduced at the break site by non-homologous end-joining. DNA Repair (Amst). May 2, 2005;4(5):546-55.
Rees et al., Analysis and minimization of cellular RNA editing by DNA adenine base editors. Sci Adv. May 8, 2019;5(5):eaax5717. doi: 10.1126/sciadv.aax5717.
Rees et al., Base editing: precision chemistry on the genome and transcriptome of living cells. Nat Rev Genet. Dec. 2018;19(12):770-788. doi: 10.1038/s41576-018-0059-1.
Rees et al., Development of hRad51-Cas9 nickase fusions that mediate HDR without double-stranded breaks. Nat Commun. May 17, 2019;10(1):2212. doi: 10.1038/s41467-019-09983-4.
Rees et al., Improving the DNA specificity and applicability of base editing through protein engineering and protein delivery. Nat Commun. Jun. 6, 2017;8:15790. doi: 10.1038/ncomms15790.
Reiners et al., Scaffold protein harmonin (USH1C) provides molecular links between Usher syndrome type 1 and type 2. Hum Mol Genet. Dec. 15, 2005;14(24):3933-43. doi: 10.1093/hmg/ddi417. Epub Nov. 21, 2005.
Relph et al., Recent developments and current status of gene therapy using viral vectors in the United Kingdom. BMJ. 2004;329(7470):839-842. doi:10.1136/bmj.329.7470.839.
Remy et al., Gene transfer with a series of lipophilic DNA-binding molecules. Bioconjug Chem. Nov.-Dec. 1994;5(6):647-54. doi: 10.1021/bc00030a021.
Ren et al., In-line Alignment and Mg2+ Coordination at the Cleavage Site of the env22 Twister Ribozyme. Nat Commun. Nov. 20, 2014;5:5534. doi: 10.1038/ncomms6534.
Ren et al., Pistol Ribozyme Adopts a Pseudoknot Fold Facilitating Site-Specific In-Line Cleavage. Nat Chem Biol. Sep. 2016;12(9):702-8. doi: 10.1038/nchembio.2125. Epub Jul. 11, 2016.
Reynaud et al., What role for AID: mutator, or assembler of the immunoglobulin mutasome? Nat Immunol. Jul. 2003;4(7):631-8.
Reyon et al., FLASH assembly of TALENs for high-throughput genome editing. Nat Biotechnol. May 2012;30(5):460-5. doi: 10.1038/nbt.2170.
Ribeiro et al., Protein Engineering Strategies to Expand CRISPR-Cas9 Applications. Int J Genomics. Aug. 2, 2018;2018:1652567. doi: 10.1155/2018/1652567.
Richardson et al., CRISPR-Cas9 genome editing in human cells occurs via the Fanconi anemia pathway. Nat Genet. Aug. 2018;50(8):1132-1139. doi: 10.1038/s41588-018-0174-0. Epub Jul. 27, 2018.
Richardson et al., Enhancing homology-directed genome editing by catalytically active and inactive CRISPR-Cas9 using asymmetric donor DNA. Nat Biotechnol. Mar. 2016;34(3):339-44. doi: 10.1038/nbt.3481. Epub Jan. 20, 2016.
Richardson et al., Frequent chromosomal translocations induced by DNA double-strand breaks. Nature. Jun. 8, 2000;405(6787):697-700. doi: 10.1038/35015097.
Richter et al., Function and regulation of clustered regularly interspaced short palindromic repeats (CRISPR) / CRISPR associated (Cas) systems. Viruses. Oct. 19, 2012;4(10):2291-311. doi: 10.3390/v4102291.
Richter et al., Phage-assisted evolution of an adenine base editor with improved Cas domain compatibility and activity. Nat Biotechnol. Jul. 2020;38(7):883-891. doi: 10.1038/s41587-020-0453-z. Epub Mar. 16, 2020.
Riechmann et al., The C-terminal domain of TolA is the coreceptor for filamentous phage infection of E. coli. Cell. 1997; 90(2):351-60. PMID:9244308.
Ringrose et al., The Kw recombinase, an integrase from Kluyveromyces waltii. Eur J Biochem. Sep. 15, 1997;248(3):903-12. doi: 10.1111/j.1432-1033.1997.00903.x.
Risso et al., Hyperstability and substrate promiscuity in laboratory resurrections of Precambrian β-lactamases. J Am Chem Soc. Feb. 27, 2013;135(8):2899-902. doi: 10.1021/ja311630a. Epub Feb. 14, 2013.
Ritchie et al., limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. Apr. 20, 2015;43(7):e47. doi: 10.1093/nar/gkv007. Epub Jan. 20, 2015.
Robertson et al., DNA repair in mammalian cells: Base excision repair: the long and short of it. Cell Mol Life Sci. Mar. 2009;66(6):981-93. doi: 10.1007/s00018-009-8736-z.
Robertson et al., Selection in vitro of an RNA enzyme that specifically cleaves single-stranded DNA. Nature. Mar. 29, 1990;344(6265):467-8. doi: 10.1038/344467a0.
Robinson et al., The protein tyrosine kinase family of the human genome. Oncogene. Nov. 20, 2000;19(49):5548-57. doi: 10.1038/sj.onc.1203957.
Rodriguez-Muela et al., Single-Cell Analysis of SMN Reveals Its Broader Role in Neuromuscular Disease. Cell Rep. Feb. 7, 2017;18(6):1484-1498 and Supplemental Information. doi: 10.1016/j.celrep.2017.01.035.
Rogozin et al., Evolution and diversification of lamprey antigen receptors: evidence for involvement of an AID-APOBEC family cytosine deaminase. Nat Immunol. Jun. 2007;8(6):647-56. doi: 10.1038/ni1463. Epub Apr. 29, 2007.
Rong et al., Homologous recombination in human embryonic stem cells using CRISPR/Cas9 nickase and a long DNA donor template. Protein Cell. Apr. 2014;5(4):258-60. doi: 10.1007/s13238-014-0032-5.
Rongrong et al., Effect of deletion mutation on the recombination activity of Cre recombinase. Acta Biochim Pol. 2005;52(2):541-4. Epub May 15, 2005.
Roth et al., A widespread self-cleaving ribozyme class is revealed by bioinformatics. Nat Chem Biol. Jan. 2014;10(1):56-60. doi: 10.1038/nchembio.1386. Epub Nov. 17, 2013.
Roth et al., Purification and characterization of murine retroviral reverse transcriptase expressed in Escherichia coli. J Biol Chem. Aug. 5, 1985;260(16):9326-35.
Rouet et al., Expression of a site-specific endonuclease stimulates homologous recombination in mammalian cells. Proc Natl Acad Sci U S A. Jun. 21, 1994;91(13):6064-8. doi: 10.1073/pnas.91.13.6064.
Rouet et al., Introduction of double-strand breaks into the genome of mouse cells by expression of a rare-cutting endonuclease. Mol Cell Biol. Dec. 1994; 14(12):8096-106. doi: 10.1128/mcb.14.12.8096.
Rouet et al., Receptor-Mediated Delivery of CRISPR-Cas9 Endonuclease for Cell-Type-Specific Gene Editing. J Am Chem Soc. May 30, 2018;140(21):6596-6603. doi: 10.1021/jacs.8b01551. Epub May 18, 2018.
Roundtree et al., YTHDC1 mediates nuclear export of N6-methyladenosine methylated mRNAs. Elife. Oct. 6, 2017;6:e31311. doi: 10.7554/eLife.31311.
Rowland et al., Regulatory mutations in Sin recombinase support a structure-based model of the synaptosome. Mol Microbiol. Oct. 2009;74(2):282-98. doi: 10.1111/j.1365-2958.2009.06756.x. Epub Jun. 8, 2009.
Rowland et al., Sin recombinase from Staphylococcus aureus: synaptic complex architecture and transposon targeting. Mol Microbiol. May 2002;44(3):607-19. doi: 10.1046/j.1365-2958.2002.02897.x.
Rowley, Chromosome translocations: dangerous liaisons revisited. Nat Rev Cancer. Dec. 2001;1(3):245-50. doi: 10.1038/35106108.
Rubio et al., An adenosine-to-inosine tRNA-editing enzyme that can perform C-to-U deamination of DNA. Proc Natl Acad Sci U S A. May 8, 2007;104(19):7821-6. doi: 10.1073/pnas.0702394104. Epub May 1, 2007. PMID: 17483465; PMCID: PMC1876531.
Rubio et al., Transfer RNA travels from the cytoplasm to organelles. Wiley Interdiscip Rev RNA. Nov.-Dec. 2011;2(6):802-17. doi: 10.1002/wrna.93. Epub Jul. 11, 2011.
Rudolph et al., Synthetic riboswitches for the conditional control of gene expression in Streptomyces coelicolor. Microbiology. Jul. 2013;159(Pt 7):1416-22. doi: 10.1099/mic.0.067322-0. Epub May 15, 2013.
Rutherford et al., Attachment site recognition and regulation of directionality by the serine integrases. Nucleic Acids Res. Sep. 2013;41(17):8341-56. doi: 10.1093/nar/gkt580. Epub Jul. 2, 2013.
Ryu et al., Adenine base editing in mouse embryos and an adult mouse model of Duchenne muscular dystrophy. Nat Biotechnol. Jul. 2018;36(6):536-539. doi: 10.1038/nbt.4148. Epub Apr. 27, 2018.
Rüfer et al., Non-contact positions impose site selectivity on Cre recombinase. Nucleic Acids Res. Jul. 1, 2002;30(13):2764-71. doi: 10.1093/nar/gkf399.
Saayman et al., The therapeutic application of CRISPR/Cas9 technologies for HIV. Expert Opin Biol Ther. Jun. 2015;15(6):819-30. doi: 10.1517/14712598.2015.1036736. Epub Apr. 12, 2015.
Sadelain et al., Safe harbours for the integration of new DNA in the human genome. Nat Rev Cancer. Dec. 1, 2011;12(1):51-8. doi: 10.1038/nrc3179.
Sadowski, The Flp recombinase of the 2-microns plasmid of Saccharomyces cerevisiae. Prog Nucleic Acid Res Mol Biol. 1995;51:53-91.
Safari et al., CRISPR Cpf1 proteins: structure, function and implications for genome editing. Cell Biosci. May 9, 2019;9:36. doi: 10.1186/s13578-019-0298-7.
Sage et al., Proliferation of functional hair cells in vivo in the absence of the retinoblastoma protein. Science. Feb. 18, 2005;307(5712):1114-8. Epub Jan. 13, 2005.
Saha et al., The NIH Somatic Cell Genome Editing program. Nature. Apr. 2021;592(7853):195-204. doi: 10.1038/s41586-021-03191-1. Epub Apr. 7, 2021.
Sakuma et al., MMEJ-assisted gene knock-in using TALENs and CRISPR-Cas9 with the PITCh systems. Nat Protoc. Jan. 2016;11(1):118-33. doi: 10.1038/nprot.2015.140. Epub Dec. 17, 2015.
Sale et al., Y-family DNA polymerases and their role in tolerance of cellular DNA damage. Nat Rev Mol Cell Biol. Feb. 23, 2012;13(3):141-52. doi: 10.1038/nrm3289.
Saleh-Gohari et al., Conservative homologous recombination preferentially repairs DNA double-strand breaks in the S phase of the cell cycle in human cells. Nucleic Acids Res. Jul. 13, 2004;32(12):3683-8. Print 2004.
Samal et al., Cationic polymers and their therapeutic potential. Chem Soc Rev. Nov. 7, 2012;41(21):7147-94. doi: 10.1039/c2cs35094g. Epub Aug. 10, 2012.
Samanta et al., A reverse transcriptase ribozyme. Elife. Sep. 26, 2017;6:e31153. doi: 10.7554/eLife.31153.
Samulski et al., Helper-free stocks of recombinant adeno-associated viruses: normal integration does not require viral gene expression. J Virol. Sep. 1989;63(9):3822-8. doi: 10.1128/JVI.63.9.3822-3828.1989.
San Filippo et al., Mechanism of eukaryotic homologous recombination. Annu Rev Biochem. 2008;77:229-57. doi: 10.1146/annurev.biochem.77.061306.125255.
Sander et al., CRISPR-Cas systems for editing, regulating and targeting genomes. Nat Biotechnol. Apr. 2014;32(4):347-55. doi: 10.1038/nbt.2842. Epub Mar. 2, 2014.
Sander et al., In silico abstraction of zinc finger nuclease cleavage profiles reveals an expanded landscape of off-target sites. Nucleic Acids Res. Oct. 2013;41(19):e181. doi: 10.1093/nar/gkt716. Epub Aug. 14, 2013.
Sander et al., Targeted gene disruption in somatic zebrafish cells using engineered TALENs. Nat Biotechnol. Aug. 5, 2011;29(8):697-8. doi: 10.1038/nbt.1934.
Sang et al., A unique uracil-DNA binding protein of the uracil DNA glycosylase superfamily. Nucleic Acids Res. Sep. 30, 2015;43(17):8452-63. doi: 10.1093/nar/gkv854. Epub Aug. 24, 2015.
Sang, Prospects for transgenesis in the chick. Mech Dev. Sep. 2004;121(9):1179-86.
Sanjana et al., A transcription activator-like effector toolbox for genome engineering. Nat Protoc. Jan. 5, 2012;7(1):171-92. doi: 10.1038/nprot.2011.431.
Sanjurjo-Soriano et al., Genome Editing in Patient iPSCs Corrects the Most Prevalent USH2A Mutations and Reveals Intriguing Mutant mRNA Expression Profiles. Mol Ther Methods Clin Dev. Nov. 27, 2019;17:156-173. doi: 10.1016/j.omtm.2019.11.016.
Santiago et al., Targeted gene knockout in mammalian cells by using engineered zinc-finger nucleases. Proc Natl Acad Sci U S A. Apr. 15, 2008;105(15):5809-14. doi: 10.1073/pnas.0800940105. Epub Mar. 21, 2008.
Santoro et al., Directed evolution of the site specificity of Cre recombinase. Proc Natl Acad Sci U S A. Apr. 2, 2002;99(7):4185-90. Epub Mar. 19, 2002.
Saparbaev et al., Excision of hypoxanthine from DNA containing dIMP residues by the Escherichia coli, yeast, rat, and human alkylpurine DNA glycosylases. Proc Natl Acad Sci U S A. Jun. 21, 1994;91(13):5873-7. doi: 10.1073/pnas.91.13.5873.
Sapranauskas et al., The Streptococcus thermophilus CRISPR/Cas system provides immunity in Escherichia coli. Nucleic Acids Res. Nov. 2011;39(21):9275-82. doi: 10.1093/nar/gkr606. Epub Aug. 3, 2011.
Sapunar et al., Dorsal root ganglion—a potential new therapeutic target for neuropathic pain. J Pain Res. 2012;5:31-8. doi: 10.2147/JPR.S26603. Epub Feb. 16, 2012.
Saraconi et al., The RNA editing enzyme APOBEC1 induces somatic mutations and a compatible mutational signature is present in esophageal adenocarcinomas. Genome Biol. Jul. 31, 2014;15(7):417. doi: 10.1186/s13059-014-0417-z.
Sarkar et al., HIV-1 proviral DNA excision using an evolved recombinase. Science. Jun. 29, 2007;316(5833):1912-5. doi: 10.1126/science.1141453.
Sashital et al., Mechanism of foreign DNA selection in a bacterial adaptive immune system. Mol Cell. Jun. 8, 2012;46(5):606-15. doi: 10.1016/j.molcel.2012.03.020. Epub Apr. 19, 2012.
Sasidharan et al., The selection of acceptable protein mutations. PNAS; Jun. 12, 2007;104(24):10080-5. www.pnas.org/cgi/doi/10.1073.pnas.0703737104.
Satomura et al., Precise genome-wide base editing by the CRISPR Nickase system in yeast. Sci Rep. May 18, 2017;7(1):2095. doi: 10.1038/s41598-017-02013-7.
Saudek et al., A preliminary trial of the programmable implantable medication system for insulin delivery. N Engl J Med. Aug. 31, 1989;321(9):574-9.
Sauer et al., DNA recombination with a heterospecific Cre homolog identified from comparison of the pac-c1 regions of P1-related phages. Nucleic Acids Res. Nov. 18, 2004;32(20):6086-95. doi: 10.1093/nar/gkh941.
Savic et al., Covalent linkage of the DNA repair template to the CRISPR-Cas9 nuclease enhances homology-directed repair. Elife. May 29, 2018;7:e33761. doi: 10.7554/eLife.33761.
Saville et al., A site-specific self-cleavage reaction performed by a novel RNA in Neurospora mitochondria. Cell. May 18, 1990;61(4):685-96. doi: 10.1016/0092-8674(90)90480-3.
Savva et al., The structural basis of specific base-excision repair by uracil-DNA glycosylase. Nature. Feb. 9, 1995;373(6514):487-93. doi: 10.1038/373487a0.
Schaaper et al., Base selection, proofreading, and mismatch repair during DNA replication in Escherichia coli. J Biol Chem. Nov. 15, 1993;268(32):23762-5.
Schaaper et al., Spectra of spontaneous mutations in Escherichia coli strains defective in mismatch correction: the nature of in vivo DNA replication errors. Proc Natl Acad Sci U S A. Sep. 1987;84(17):6220-4.
Schaefer et al., Understanding RNA modifications: the promises and technological bottlenecks of the ‘epitranscriptome’. Open Biol. May 2017;7(5):170077. doi: 10.1098/rsob.170077.
Schechner et al., Multiplexable, locus-specific targeting of long RNAs with CRISPR-Display. Nat Methods. Jul. 2015;12(7):664-70. doi: 10.1038/nmeth.3433. Epub Jun. 1, 2015. Author manuscript entitled CRISPR Display: A modular method for locus-specific targeting of long noncoding RNAs and synthetic RNA devices in vivo.
Schek et al., Definition of the upstream efficiency element of the simian virus 40 late polyadenylation signal by using in vitro analyses. Mol Cell Biol. Dec. 1992;12(12):5386-93. doi: 10.1128/mcb.12.12.5386.
Schenk et al., MPDU1 mutations underlie a novel human congenital disorder of glycosylation, designated type If. J Clin Invest. Dec. 2001; 108(11):1687-95. doi: 10.1172/JCI13419.
Schlacher et al., Double-strand break repair-independent role for BRCA2 in blocking stalled replication fork degradation by MRE11. Cell. May 13, 2011;145(4):529-42. doi: 10.1016/j.cell.2011.03.041. Erratum in: Cell. Jun. 10, 2011;145(6):993.
Schmitz et al., Behavioral abnormalities in prion protein knockout mice and the potential relevance of PrP(C) for the cytoskeleton. Prion. 2014;8(6):381-6. doi: 10.4161/19336896.2014.983746.
Schrank et al., Inactivation of the survival motor neuron gene, a candidate gene for human spinal muscular atrophy, leads to massive cell death in early mouse embryos. Proc Natl Acad Sci USA. Sep. 2, 1997;94(18):9920-5. doi: 10.1073/pnas.94.18.9920.
Schriefer et al., Low pressure DNA shearing: a method for random DNA sequence analysis. Nucleic Acids Res. Dec. 25, 1990;18(24):7455-6.
Schultz et al., Expression and secretion in yeast of a 400-kDa envelope glycoprotein derived from Epstein-Barr virus. Gene. 1987;54(1):113-23. doi: 10.1016/0378-1119(87)90353-2.
Schultz et al., Oligo-2′-fluoro-2′-deoxynucleotide N3′-->P5′ phosphoramidates: synthesis and properties. Nucleic Acids Res. Aug. 1, 1996;24(15):2966-73.
Schwank et al., Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell. Dec. 5, 2013;13(6):653-8. doi:10.1016/j.stem.2013.11.002.
Schwartz et al., Post-translational enzyme activation in an animal via optimized conditional protein splicing. Nat Chem Biol. Jan. 2007;3(1):50-4. Epub Nov. 26, 2006.
Schwarze et al., In vivo protein transduction: delivery of a biologically active protein into the mouse. Science. Sep. 3, 1999;285(5433):1569-72.
Schöller et al., Interactions, localization, and phosphorylation of the m6A generating METTL3-METTL14-WTAP complex. RNA. Apr. 2018;24(4):499-512. doi: 10.1261/rna.064063.117. Epub Jan. 18, 2018.
Sclimenti et al., Directed evolution of a recombinase for improved genomic integration at a native human sequence. Nucleic Acids Res. Dec. 15, 2001;29(24):5044-51.
SCORE Results for Luetticken et al., Complete genome sequence of a Streptococcus dysgalactiae subsp. RT equisimilis strain possessing Lancefield's group A antigen. RL Submitted to the EMBL/GenBank/DDBJ databases. May 2012. 3 pages.
SCORE Results for Okumura et al., Evolutionary paths of streptococcal and staphylococcal superantigens. RL BMC Genomics. 2012;13:404-404. 3 pages.
SCORE Results for Shimomura et al., Complete Genome Sequencing and Analysis of a Lancefield Group G RT Streptococcus dysagalactiae Subsp. Equisimilis Strain Causing Streptococcal RT Toxic Shock Syndrome (STSS). RL BMC Genomics. 2011;12:17-17. 3 pages.
Scott et al., Production of cyclic peptides and proteins in vivo. Proc Natl Acad Sci U S A. Nov. 23, 1999;96(24):13638-43. doi: 10.1073/pnas.96.24.13638.
Sebastían-Martín et al., Transcriptional inaccuracy threshold attenuates differences in RNA-dependent DNA synthesis fidelity between retroviral reverse transcriptases. Sci Rep. Jan. 12, 2018;8(1):627. doi: 10.1038/s41598-017-18974-8.
Seed, An LFA-3 cDNA encodes a phospholipid-linked membrane protein homologous to its receptor CD2. Nature. Oct. 29-Nov. 4, 1987;329(6142):840-2. doi: 10.1038/329840a0.
Sefton et al., Implantable pumps. Crit Rev Biomed Eng. 1987;14(3):201-40.
Segal et al., Toward controlling gene expression at will: selection and design of zinc finger domains recognizing each of the 5′-GNN-3′ DNA target sequences. Proc Natl Acad Sci U S A. Mar. 16, 1999;96(6):2758-63.
Sells et al., Delivery of protein into cells using polycationic liposomes. Biotechniques. Jul. 1995;19(1):72-6, 78.
Semenova et al., Interference by clustered regularly interspaced short palindromic repeat (CRISPR) RNA is governed by a seed sequence. Proc Natl Acad Sci U S A. Jun. 21, 2011;108(25):10098-103. doi: 10.1073/pnas.1104144108. Epub Jun. 6, 2011.
Semple et al., Rational design of cationic lipids for siRNA delivery. Nat Biotechnol. Feb. 2010;28(2):172-6. doi: 10.1038/nbt.1602. Epub Jan. 17, 2010.
Serganov et al., Coenzyme recognition and gene regulation by a flavin mononucleotide riboswitch. Nature. Mar. 12, 2009;458(7235):233-7. doi: 10.1038/nature07642. Epub Jan. 25, 2009.
Serganov et al., Structural basis for discriminative regulation of gene expression by adenine- and guanine-sensing mRNAs. Chem Biol. Dec. 2004; 11(12):1729-41.
Serganov et al., Structural basis for gene regulation by a thiamine pyrophosphate-sensing riboswitch. Nature. Jun. 29, 2006;441(7097):1167-71. Epub May 21, 2006.
Seripa et al., The missing ApoE allele. Ann Hum Genet. Jul. 2007;71(Pt 4):496-500. Epub Jan. 22, 2007.
Serrano-Heras et al., Protein p56 from the Bacillus subtilis phage phi29 inhibits DNA-binding ability of uracil-DNA glycosylase. Nucleic Acids Res. 2007;35(16):5393-401. Epub Aug. 13, 2007.
Setten et al., The current state and future directions of RNAi-based therapeutics. Nat Rev Drug Discov. Jun. 2019; 18(6):421-446. doi: 10.1038/s41573-019-0017-4.
Severinov et al., Expressed protein ligation, a novel method for studying protein-protein interactions in transcription. J Biol Chem. Jun. 26, 1998;273(26):16205-9. doi: 10.1074/jbc.273.26.16205.
Sha et al., Monobodies and other synthetic binding proteins for expanding protein science. Protein Sci. May 2017;26(5):910-924. doi: 10.1002/pro.3148. Epub Mar. 24, 2017.
Shah et al., Inteins: nature's gift to protein chemists. Chem Sci. 2014;5(1):446-461.
Shah et al., Kinetic control of one-pot trans-splicing reactions by using a wild-type and designed split intein. Angew Chem Int Ed Engl. Jul. 11, 2011;50(29):6511-5. doi: 10.1002/anie.201102909. Epub Jun. 8, 2011.
Shah et al., Protospacer recognition motifs: mixed identities and functional diversity. RNA Biol. May 2013;10(5):891-9. doi: 10.4161/rna.23764. Epub Feb. 12, 2013.
Shah et al., Target-specific variants of Flp recombinase mediate genome engineering reactions in mammalian cells. FEBS J. Sep. 2015;282(17):3323-33. doi: 10.1111/febs.13345. Epub Jul. 1, 2015.
Shaikh et al., Chimeras of the Flp and Cre recombinases: tests of the mode of cleavage by Flp and Cre. J Mol Biol. Sep. 8, 2000;302(1):27-48.
Shalem et al., Genome-scale CRISPR-Cas9 knockout screening in human cells. Science. Jan. 3, 2014;343(6166):84-7. doi: 10.1126/science.1247005. Epub Dec. 12, 2013.
Shalem et al., High-throughput functional genomics using CRISPR-Cas9. Nat Rev Genet. May 2015;16(5):299-311. doi: 10.1038/nrg3899. Epub Apr. 9, 2015.
Sharbeen et al., Ectopic restriction of DNA repair reveals that UNG2 excises AID-induced uracils predominantly or exclusively during G1 phase. J Exp Med. May 7, 2012;209(5):965-74. doi: 10.1084/jem.20112379. Epub Apr. 23, 2012.
Sharer et al., The ARF-like 2 (ARL2)-binding protein, BART. Purification, cloning, and initial characterization. J Biol Chem. Sep. 24, 1999;274(39):27553-61. doi: 10.1074/jbc.274.39.27553.
Sharma et al., Efficient introduction of aryl bromide functionality into proteins in vivo. FEBS Lett. Feb. 4, 2000;467(1):37-40.
Sharma et al., Identification of novel methyltransferases, Bmt5 and Bmt6, responsible for the m3U methylations of 25S rRNA in Saccharomyces cerevisiae. Nucleic Acids Res. Mar. 2014;42(5):3246-60. doi: 10.1093/nar/gkt1281. Epub Dec. 11, 2013.
Sharon et al., Functional Genetic Variants Revealed by Massively Parallel Precise Genome Editing. Cell. Oct. 4, 2018;175(2):544-557.e16. doi: 10.1016/j.cell.2018.08.057. Epub Sep. 20, 2018.
Shaw et al., Implications of human genome architecture for rearrangement-based disorders: the genomic basis of disease. Hum Mol Genet. Apr. 1, 2004;13 Spec No. 1:R57-64. doi: 10.1093/hmg/ddh073. Epub Feb. 5, 2004.
Shcherbakova et al., Near-infrared fluorescent proteins for multicolor in vivo imaging. Nat Methods. Aug. 2013;10(8):751-4. doi: 10.1038/nmeth.2521. Epub Jun. 16, 2013.
Shechner et al., Multiplexable, locus-specific targeting of long RNAs with CRISPR-Display. Nat Methods. Jul. 2015;12(7):664-70. doi: 10.1038/nmeth.3433. Epub Jun. 1, 2015.
Shee et al., Engineered proteins detect spontaneous DNA breakage in human and bacterial cells. Elife. Oct. 29, 2013;2:e01222. doi: 10.7554/eLife.01222.
Shen et al., Efficient genome modification by CRISPR-Cas9 nickase with minimal off-target effects. Nat Methods. Apr. 2014; 11(4):399-402. doi: 10.1038/nmeth.2857. Epub Mar. 2, 2014.
Shen et al., Herpes simplex virus 1 (HSV-1) for cancer treatment. Cancer Gene Ther. Nov. 2006;13(11):975-92. doi: 10.1038/sj.cgt.7700946. Epub Apr. 7, 2006.
Shen et al., Predictable and precise template-free CRISPR editing of pathogenic variants. Nature. Nov. 2018;563(7733):646-651. doi: 10.1038/s41586-018-0686-x. Epub Nov. 7, 2018.
Shen, Data processing, Modeling and Analysis scripts for CRISPR-inDelphi. GitHub—maxwshen/indelphi-dataprocessinganalysis at 6b68e3cec73c9358fef6e5f178a935f3c2a4118f. Apr. 10, 2018. Retrieved online via https://github.com/maxwshen/indelphi-sataprocessinganalysis/tree/6b68e3cec73c9358fef6e5f178a935f3c2a4118f Last retrieved on Jul. 26, 2021. 2 pages.
Sheridan, First CRISPR-Cas patent opens race to stake out intellectual property. Nat Biotechnol. 2014;32(7):599-601.
Sheridan, Gene therapy finds its niche. Nat Biotechnol. Feb. 2011;29(2):121-8. doi: 10.1038/nbt.1769.
Sherwood et al., Discovery of directional and nondirectional pioneer transcription factors by modeling DNase profile magnitude and shape. Nat Biotechnol. Feb. 2014;32(2):171-178. doi: 10.1038/nbt.2798. Epub Jan. 19, 2014.
Shi et al., Structural basis for targeted DNA cytosine deamination and mutagenesis by APOBEC3A and APOBEC3B. Nat Struct Mol Biol. Feb. 2017;24(2): 131-139. doi: 10.1038/nsmb.3344. Epub Dec. 19, 2016.
Shi et al., YTHDF3 facilitates translation and decay of N6-methyladenosine-modified RNA. Cell Res. Mar. 2017;27(3):315-328. doi: 10.1038/cr.2017.15. Epub Jan. 20, 2017.
Shimantani et al., Targeted base editing in rice and tomato using a CRISPR-Cas9 cytidine deaminase fusion. Nat Biotechnol. May 2017;35(5):441-443. doi: 10.1038/nbt.3833. Epub Mar. 27, 2017.
Shimojima et al., Spinocerebellar ataxias type 27 derived from a disruption of the fibroblast growth factor 14 gene with mimicking phenotype of paroxysmal non-kinesigenic dyskinesia. Brain Dev. Mar. 2012;34(3):230-3. doi: 10.1016/j.braindev.2011.04.014. Epub May 19, 2011.
Shin et al., CRISPR/Cas9 targeting events cause complex deletions and insertions at 17 sites in the mouse genome. Nat Commun. May 31, 2017;8:15464. doi: 10.1038/ncomms15464.
Shindo et al., A Comparison of Two Single-Stranded DNA Binding Models by Mutational Analysis of APOBEC3G. Biology (Basel). Aug. 2, 2012;1(2):260-76. doi: 10.3390/biology1020260.
Shingledecker et al., Molecular dissection of the Mycobacterium tuberculosis RecA intein: design of a minimal intein and of a trans-splicing system involving two intein fragments. Gene. Jan. 30, 1998;207(2):187-95. doi: 10.1016/s0378-1119(97)00624-0.
Shmakov et al., Discovery and Functional Characterization of Diverse Class 2 CRISPR Cas Systems. Molecular Cell Nov. 2015;60(3):385-97.
Shmakov et al., Diversity and evolution of class 2 CRISPR-Cas systems. Nat Rev Microbiol. Mar. 2017;15(3):169-182. doi: 10.1038/nrmicro.2016.184. Epub Jan. 23, 2017.
Shultz et al., A genome-wide analysis of FRT-like sequences in the human genome. PLoS One. Mar. 23, 2011;6(3):e18077. doi: 10.1371/journal.pone.0018077.
Siebert et al., An improved PCR method for walking in uncloned genomic DNA. Nucleic Acids Res. Mar. 25, 1995;23(6): 1087-8.
Silas et al., Direct CRISPR spacer acquisition from RNA by a natural reverse transcriptase-Cas1 fusion protein. Science. Feb. 26, 2016;351(6276):aad4234. doi: 10.1126/science.aad4234.
Silva et al., Selective disruption of the DNA polymerase III α-β complex by the umuD gene products. Nucleic Acids Res. Jul. 2012;40(12):5511-22. doi: 10.1093/nar/gks229. Epub Mar. 9, 2012.
Simonelli et al., Base excision repair intermediates are mutagenic in mammalian cells. Nucleic Acids Res. Aug. 2, 2005;33(14):4404-11. Print 2005.
Singh et al., Cross-talk between diverse serine integrases. J Mol Biol. Jan. 23, 2014;426(2):318-31. doi: 10.1016/j.jmb.2013.10.013. Epub Oct. 22, 2013.
Singh et al., Real-time observation of DNA recognition and rejection by the RNA-guided endonuclease Cas9. Nat Commun. Sep. 14, 2016;7:12778. doi: 10.1038/ncomms12778.
Singh et al., Real-time observation of DNA target interrogation and product release by the RNA-guided endonuclease CRISPR Cpf1 (Cas12a). Proc Natl Acad Sci U S A. May 22, 2018;115(21):5444-5449. doi: 10.1073/pnas.1718686115. Epub May 7, 2018.
Singh et al., Splicing of a critical exon of human Survival Motor Neuron is regulated by a unique silencer element located in the last intron. Mol Cell Biol. Feb. 2006;26(4):1333-46. doi: 10.1128/MCB.26.4.1333-1346.2006.
Sirk et al., Expanding the zinc-finger recombinase repertoire: directed evolution and mutational analysis of serine recombinase specificity determinants. Nucleic Acids Res. Apr. 2014;42(7):4755-66. doi: 10.1093/nar/gkt1389. Epub Jan. 21, 2014.
Siu et al., Riboregulated toehold-gated gRNA for programmable CRISPR-Cas9 function. Nat Chem Biol. Mar. 2019;15(3):217-220. doi: 10.1038/s41589-018-0186-1. Epub Dec. 10, 2018.
Sivalingam et al., Biosafety assessment of site-directed transgene integration in human umbilical cord-lining cells. Mol Ther. Jul. 2010;18(7):1346-56. doi: 10.1038/mt.2010.61. Epub Apr. 27, 2010.
Sjoblom et al., The consensus coding sequences of human breast and colorectal cancers. Science. Oct. 13, 2006;314(5797):268-74. Epub Sep. 7, 2006.
Skretas et al., Regulation of protein activity with small-molecule-controlled inteins. Protein Sci. Feb. 2005; 14(2):523-32. Epub Jan. 4, 2005.
Slaymaker et al., Rationally engineered Cas9 nucleases with improved specificity. Science. Jan. 1, 2016;351(6268):84-8. doi: 10.1126/science.aad5227. Epub Dec. 1, 2015.
Sledz et al., Structural insights into the molecular mechanism of the m(6)A writer complex. Elife. Sep. 14, 2016;5:e18434. doi: 10.7554/eLife.18434.
Slupphaug et al., A nucleotide-flipping mechanism from the structure of human uracil-DNA glycosylase bound to DNA. Nature. Nov. 7, 1996;384(6604):87-92. doi: 10.1038/384087a0.
Smargon et al., Cas13b Is a Type VI-B CRISPR-Associated RNA-Guided RNase Differentially Regulated by Accessory Proteins Csx27 and Csx28. Mol Cell. Feb. 16, 2017;65(4):618-630.e7. doi: 10.1016/j.molcel.2016.12.023. Epub Jan. 5, 2017.
Smith et al., Diversity in the serine recombinases. Mol Microbiol. Apr. 2002;44(2):299-307. Review.
Smith et al., Expression of a dominant negative retinoic acid receptor ? in Xenopus embryos leads to partial resistance to retinoic acid. Roux Arch Dev Biol. Mar. 1994;203(5):254-265. doi: 10.1007/BF00360521.
Smith et al., Herpesvirus transport to the nervous system and back again. Annu Rev Microbiol. 2012;66:153-76. doi: 10.1146/annurev-micro-092611-150051. Epub Jun. 15, 2012.
Smith et al., Production of human beta interferon in insect cells infected with a baculovirus expression vector. Mol Cell Biol. Dec. 1983;3(12):2156-65. doi: 10.1128/mcb.3.12.2156.
Smith et al., Single-step purification of polypeptides expressed in Escherichia coli as fusions with glutathione S-transferase. Gene. Jul. 15, 1988;67(1):31-40. doi: 10.1016/0378-1119(88)90005-4.
Smith, Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface. Science. Jun. 14, 1985;228(4705):1315-7.
Smith, Phage-encoded Serine Integrases and Other Large Serine Recombinases. Microbiol Spectr. Aug. 2015;3(4). doi: 10.1128/microbiolspec.MDNA3-0059-2014.
Somanathan et al., AAV vectors expressing LDLR gain-of-function variants demonstrate increased efficacy in mouse models of familial hypercholesterolemia. Circ Res. Aug. 29, 2014;115(6):591-9. doi: 10.1161/CIRCRESAHA.115.304008. Epub Jul. 14, 2014.
Sommerfelt et al., Receptor interference groups of 20 retroviruses plating on human cells. Virology. May 1990;176(1):58-69. doi: 10.1016/0042-6822(90)90230-o.
Song et al., Adenine base editing in an adult mouse model of tyrosinaemia. Nat Biomed Eng. Jan. 2020;4(1):125-130. doi: 10.1038/s41551-019-0357-8. Epub Feb. 25, 2019.
Song et al., Delivery of CRISPR/Cas systems for cancer gene therapy and immunotherapy. Adv Drug Deliv Rev. Jan. 2021;168:158-180. doi: 10.1016/j.addr.2020.04.010. Epub May 1, 2020.
Song et al., RS-1 enhances CRISPR/Cas9- and TALEN-mediated knock-in efficiency. Nat Commun. Jan. 28, 2016;7:10548. doi: 10.1038/ncomms10548.
Sorusch et al., Characterization of the ternary Usher syndrome SANS/ush2a/whirlin protein complex. Hum Mol Genet. Mar. 15, 2017;26(6):1157-1172. doi: 10.1093/hmg/ddx027.
Southworth et al., Control of protein splicing by intein fragment reassembly. EMBO J. Feb. 16, 1998;17(4):918-26. doi: 10.1093/emboj/17.4.918.
Southworth et al., Purification of proteins fused to either the amino or carboxy terminus of the Mycobacterium xenopi gyrase A intein. Biotechniques. Jul. 1999;27(1):110-4, 116, 118-20. doi: 10.2144/99271st04.
Spencer et al., A general strategy for producing conditional alleles of Src-like tyrosine kinases. Proc Natl Acad Sci U S A. Oct. 10, 1995;92(21):9805-9. doi: 10.1073/pnas.92.21.9805.
Spencer et al., Controlling signal transduction with synthetic ligands. Science. Nov. 12, 1993;262(5136):1019-24. doi: 10.1126/science.7694365.
Spencer et al., Functional analysis of Fas signaling in vivo using synthetic inducers of dimerization. Curr Biol. Jul. 1, 1996;6(7):839-47. doi: 10.1016/s0960-9822(02)00607-3.
Srivastava et al., An inhibitor of nonhomologous end-joining abrogates double-strand break repair and impedes cancer progression. Cell. Dec. 21, 2012;151(7):1474-87. doi: 10.1016/j.cell.2012.11.054.
Stadtman, Selenocysteine. Annu Rev Biochem. 1996;65:83-100.
Stamos et al., Structure of a Thermostable Group II Intron Reverse Transcriptase with Template-Primer and Its Functional and Evolutionary Implications. Mol Cell. Dec. 7, 2017;68(5):926-939.e4. doi: 10.1016/j.molcel.2017.10.024. Epub Nov. 16, 2017.
Stark et al., ATP hydrolysis by mammalian RAD51 has a key role during homology-directed DNA repair. J Biol Chem. Jun. 7, 2002;277(23):20185-94. doi: 10.1074/jbc.M112132200. Epub Mar. 28, 2002.
Steele et al., The prion protein knockout mouse: a phenotype under challenge. Prion. Apr.-Jun. 2007;1(2):83-93. doi: 10.4161/pri.1.2.4346. Epub Apr. 25, 2007.
Steiner et al., The neurotropic herpes viruses: herpes simplex and varicella-zoster. Lancet Neurol. Nov. 2007;6(11):1015-28. doi: 10.1016/S1474-4422(07)70267-3.
Stella et al., Structure of the Cpf1 endonuclease R-loop complex after target DNA cleavage. Nature. Jun. 22, 2017;546(7659):559-563. doi: 10.1038/nature22398. Epub May 31, 2017.
Stenglein et al., APOBEC3 proteins mediate the clearance of foreign DNA from human cells. Nat Struct Mol Biol. Feb. 2010; 17(2):222-9. doi: 10.1038/nsmb.1744. Epub Jan. 10, 2010.
Stenson et al., The Human Gene Mutation Database: towards a comprehensive repository of inherited mutation data for medical research, genetic diagnosis and next-generation sequencing studies. Hum Genet. Jun. 2017;136(6):665-677. doi: 10.1007/s00439-017-1779-6. Epub Mar. 27, 2017.
Stephens et al., The landscape of cancer genes and mutational processes in breast cancer. Nature Jun. 2012;486:400-404. doi: 10.1038/nature11017.
Sternberg et al., Conformational control of DNA target cleavage by CRISPR-Cas9. Nature. Nov. 5, 2015;527(7576):110-3. doi: 10.1038/nature15544. Epub Oct. 28, 2015.
Sternberg et al., DNA interrogation by the CRISPR RNA-guided endonuclease Cas9. Nature.Mar. 6, 2014;507(7490):62-7. doi: 10.1038/nature13011. Epub Jan. 29, 2014.
Sterne-Weiler et al., Exon identity crisis: disease-causing mutations that disrupt the splicing code. Genome Biol. Jan. 23, 2014;15(1):201. doi: 10.1186/gb4150.
Stevens et al., A promiscuous split intein with expanded protein engineering applications. Proc Natl Acad Sci U S A. Aug. 8, 2017;114(32):8538-8543. doi: 10.1073/pnas.1701083114. Epub Jul. 24, 2017.
Stevens et al., Design of a Split Intein with Exceptional Protein-Splicing Activity. J Am Chem Soc. Feb. 24, 2016;138(7):2162-5. doi: 10.1021/jacs.5b13528. Epub Feb. 8, 2016.
Stockwell et al., Probing the role of homomeric and heteromeric receptor interactions in TGF-beta signaling using small molecule dimerizers. Curr Biol. Jun. 18, 1998;8(13):761-70. doi: 10.1016/s0960-9822(98)70299-4.
Strecker et al., Engineering of CRISPR-Cas12b for human genome editing. Nat Commun. Jan. 22, 2019;10(1):212. doi: 10.1038/s41467-018-08224-4.
Strecker et al., RNA-guided DNA insertion with CRISPR-associated transposases. Science. Jul. 5, 2019;365(6448):48-53. doi: 10.1126/science.aax9181. Epub Jun. 6, 2019.
Strutt et al., RNA-dependent RNA targeting by CRISPR-Cas9. Elife. Jan. 5, 2018;7:e32724. doi: 10.7554/eLife.32724.
Su et al., Human DNA polymerase η has reverse transcriptase activity in cellular environments. J Biol Chem. Apr. 12, 2019;294(15):6073-6081. doi: 10.1074/jbc.RA119.007925. Epub Mar. 6, 2019.
Sudarsan et al., An mRNA structure in bacteria that controls gene expression by binding lysine. Genes Dev. Nov. 1, 2003;17(21):2688-97.
Sudarsan et al., Riboswitches in eubacteria sense the second messenger cyclic di-GMP. Science. Jul. 18, 2008;321(5887):411-3. doi: 10.1126/science.1159519.
Suess et al., A theophylline responsive riboswitch based on helix slipping controls gene expression in vivo. Nucleic Acids Res. Mar. 5, 2004;32(4):1610-4.
Suh et al., Restoration of visual function in adult mice with an inherited retinal disease via adenine base editing. Nat Biomed Eng. Feb. 2021;5(2):169-178. doi: 10.1038/s41551-020-00632-6. Epub Oct. 19, 2020.
Sullenger et al., Ribozyme-mediated repair of defective mRNA by targeted, trans-splicing. Nature. Oct. 13, 1994;371(6498):619-22. doi: 10.1038/371619a0.
Sumner et al., Two breakthrough gene-targeted treatments for spinal muscular atrophy: challenges remain. J Clin Invest. Aug. 1, 2018;128(8):3219-3227. doi: 10.1172/JCI121658. Epub Jul. 9, 2018.
Sun et al., Optimized TAL effector nucleases (TALENs) for use in treatment of sickle cell disease. Mol Biosyst. Apr. 2012;8(4):1255-63. doi: 10.1039/c2mb05461b. Epub Feb. 3, 2012.
Sun et al., The CRISPR/Cas9 system for gene editing and its potential application in pain research. Transl Periop & Pain Med. Aug. 3, 2016;1(3):22-33.
Surun et al., High Efficiency Gene Correction in Hematopoietic Cells by Donor-Template-Free CRISPR/Cas9 Genome Editing. Mol Ther Nucleic Acids. Mar. 2, 2018;10:1-8. doi: 10.1016/j.omtn.2017.11.001. Epub Nov. 10, 2017.
Suzuki et al., Crystal structures reveal an elusive functional domain of pyrrolysyl-tRNA synthetase. Nat Chem Biol. Dec. 2017;13(12):1261-1266. doi: 10.1038/nchembio.2497. Epub Oct. 16, 2017.
Suzuki et al., In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration. Nature. Dec. 1, 2016;540(7631):144-149. doi: 10.1038/nature20565. Epub Nov. 16, 2016.
Suzuki et al., VCre/VloxP and SCre/SloxP: new site-specific recombination systems for genome engineering. Nucleic Acids Res. Apr. 2011;39(8):e49. doi: 10.1093/nar/gkq1280. Epub Feb. 1, 2011.
Swarts et al., Argonaute of the archaeon Pyrococcus furiosus is a DNA-guided nuclease that targets cognate DNA. Nucleic Acids Res. May 26, 2015;43(10):5120-9. doi: 10.1093/nar/gkv415. Epub Apr. 29, 2015.
Swarts et al., DNA-guided DNA interference by a prokaryotic Argonaute. Nature. Mar. 13, 2014;507(7491):258-61. doi: 10.1038/nature12971. Epub Feb. 16, 2014.
Swarts et al., The evolutionary journey of Argonaute proteins. Nat Struct Mol Biol. Sep. 2014;21(9):743-53. doi: 10.1038/nsmb.2879.
Szczepek et al., Structure-based redesign of the dimerization interface reduces the toxicity of zinc-finger nucleases. Nat Biotechnol. Jul. 2007;25(7):786-93. Epub Jul. 1, 2007.
Tabebordbar et al., In vivo gene editing in dystrophic mouse muscle and muscle stem cells. Science. Jan. 22, 2016;351(6271):407-411. doi: 10.1126/science.aad5177. Epub Dec. 31, 2015.
Tagalakis et al., Lack of RNA-DNA oligonucleotide (chimeraplast) mutagenic activity in mouse embryos. Mol Reprod Dev. Jun. 2005;71(2):140-4.
Tahara et al., Potent and Selective Inhibitors of 8-Oxoguanine DNA Glycosylase. J Am Chem Soc. Feb. 14, 2018;140(6):2105-2114. doi: 10.1021/jacs.7b09316. Epub Feb. 5, 2018.
Tajiri et al., Functional cooperation of MutT, MutM and MutY proteins in preventing mutations caused by spontaneous oxidation of guanine nucleotide in Escherichia coli. Mutat Res. May 1995;336(3):257-67. doi: 10.1016/0921-8777(94)00062-b.
Takimoto et al., Stereochemical basis for engineered; pyrrolysyl-tRNA synthetase and the efficient in vivo incorporation of; structurally divergent non-native amino acids. ACS Chem Biol. Jul. 2011; 15;6(7):733-43. doi: 10.1021/cb200057a. Epub May 5, 2011.
Talbot et al., Spinal muscular atrophy. Semin Neurol. Jun. 2001;21(2):189-97. doi: 10.1055/s-2001-15264.
Tambunan et al., Vaccine Design for H5N1 Based on B- and T-cell Epitope Predictions. Bioinform Biol Insights. Apr. 28, 2016;10:27-35. doi: 10.4137/BBI.S38378.
Tan et al., Engineering of high-precision base editors for site-specific single nucleotide replacement. Nat Commun. Jan. 25, 2019;10(1):439. doi: 10.1038/s41467-018-08034-8. Erratum in: Nat Commun. May 1, 2019;10(1):2019.
Tanenbaum et al., A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell. Oct. 23, 2014;159(3):635-46. doi: 10.1016/j.cell.2014.09.039. Epub Oct. 9, 2014.
Tanese et al., Expression of enzymatically active reverse transcriptase in Escherichia coli. Proc Natl Acad Sci U S A. Aug. 1985;82(15):4944-8. doi: 10.1073/pnas.82.15.4944.
Tang et al., Aptazyme-embedded guide RNAs enable ligand-responsive genome editing and transcriptional activation. Nat Commun. Jun. 28, 2017;8:15939. doi: 10.1038/ncomms15939.
Tang et al., Evaluation of Bioinformatic Programmes for the Analysis of Variants within Splice Site Consensus Regions. Adv Bioinformatics. 2016;2016:5614058. doi: 10.1155/2016/5614058. Epub May 24, 2016.
Tang et al., Rewritable multi-event analog recording in bacterial and mammalian cells. Science. Apr. 13, 2018;360(6385):eaap8992. doi: 10.1126/science.aap8992. Epub Feb. 15, 2018.
Tassabehji, Williams-Beuren syndrome: a challenge for genotype-phenotype correlations. Hum Mol Genet. Oct. 15, 2003;12 Spec No. 2:R229-37. doi: 10.1093/hmg/ddg299. Epub Sep. 2, 2003.
Taube et al., Reverse transcriptase of mouse mammary tumour virus: expression in bacteria, purification and biochemical characterization. Biochem J. Feb. 1, 1998;329 ( Pt 3)(Pt 3):579-87. doi: 10.1042/bj3290579. Erratum in: Biochem J Jun. 15, 1998;332(Pt 3):808.
Tebas et al., Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med. Mar. 6, 2014;370(10):901-10. doi: 10.1056/NEJMoa1300662.
Tee et al., Polishing the craft of genetic diversity creation in directed evolution. Biotechnol Adv. Dec. 2013;31(8):1707-21. doi: 10.1016/j.biotechadv.2013.08.021. Epub Sep. 6, 2013.
Telenti et al., The Mycobacterium xenopi GyrA protein splicing element: characterization of a minimal intein. J Bacteriol. Oct. 1997;179(20):6378-82. doi: 10.1128/jb.179.20.6378-6382.1997.
Telesnitsky et al., RNase H domain mutations affect the interaction between Moloney murine leukemia virus reverse transcriptase and its primer-template. Proc Natl Acad Sci U S A. Feb. 15, 1993;90(4):1276-80. doi: 10.1073/pnas.90.4.1276.
Teng et al., Mutational analysis of apolipoprotein B mRNA editing enzyme (APOBEC1). structure-function relationships of RNA editing and dimerization. J Lipid Res. Apr. 1999;40(4):623-35.
Tessarollo et al., Targeted mutation in the neurotrophin-3 gene results in loss of muscle sensory neurons. Proc Natl Acad Sci U S A. Dec. 6, 1994;91(25):11844-8.
Tesson et al., Knockout rats generated by embryo microinjection of TALENs. Nat Biotechnol. Aug. 5, 2011;29(8):695-6. doi: 10.1038/nbt.1940.
Thompson et al., Cellular uptake mechanisms and endosomal trafficking of supercharged proteins. Chem Biol. Jul. 27, 2012;19(7):831-43. doi: 10.1016/j.chembiol.2012.06.014.
Thompson et al., Engineering and identifying supercharged proteins for macromolecule delivery into mammalian cells. Methods Enzymol. 2012;503:293-319. doi: 10.1016/B978-0-12-396962-0.00012-4.
Thompson et al., The Future of Multiplexed Eukaryotic Genome Engineering. ACS Chem Biol. Feb. 16, 2018;13(2):313-325. doi: 10.1021/acschembio.7b00842. Epub Dec. 28, 2017.
Thomson et al., Mutational analysis of loxP sites for efficient Cre-mediated insertion into genomic DNA. Genesis. Jul. 2003;36(3):162-7. doi: 10.1002/gene.10211.
Thorpe et al., Functional correction of episomal mutations with short DNA fragments and RNA-DNA oligonucleotides. J Gene Med. Mar.-Apr. 2002;4(2):195-204.
Thuronyi et al., Continuous evolution of base editors with expanded target compatibility and improved activity. Nat Biotechnol. Sep. 2019;37(9):1070-1079. doi: 10.1038/s41587-019-0193-0. Epub Jul. 22, 2019.
Thyagarajan et al., Creation of engineered human embryonic stem cell lines using phiC31 integrase. Stem Cells. Jan. 2008;26(1):119-26. doi: 10.1634/stemcells.2007-0283. Epub Oct. 25, 2007.
Thyagarajan et al., Mammalian genomes contain active recombinase recognition sites. Gene. Feb. 22, 2000;244(1-2):47-54.
Thyagarajan et al., Site-specific genomic integration in mammalian cells mediated by phage phiC31 integrase. Mol Cell Biol. Jun. 2001;21(12):3926-34.
Tinland et al., The T-DNA-linked VirD2 protein contains two distinct functional nuclear localization signals. Proc Natl Acad Sci U S A. Aug. 15, 1992;89(16):7442-6. doi: 10.1073/pnas.89.16.7442.
Tirumalai et al., Recognition of core-type DNA sites by lambda integrase. J Mol Biol. Jun. 12, 1998;279(3):513-27.
Tom et al., Mechanism whereby proliferating cell nuclear antigen stimulates flap endonuclease 1. J Biol Chem. Apr. 7, 2000;275(14):10498-505. doi: 10.1074/jbc.275.14.10498.
Tone et al., Single-stranded DNA binding protein Gp5 of Bacillus subtilis phage Φ29 is required for viral DNA replication in growth-temperature dependent fashion. Biosci Biotechnol Biochem. 2012;76(12):2351-3. doi: 10.1271/bbb.120587. Epub Dec. 7, 2012.
Toor et al., Crystal structure of a self-spliced group II intron. Science. Apr. 4, 2008;320(5872):77-82. doi: 10.1126/science.1153803.
Toro et al., On the Origin and Evolutionary Relationships of the Reverse Transcriptases Associated With Type III CRISPR-Cas Systems. Front Microbiol. Jun. 15, 2018;9:1317. doi: 10.3389/fmicb.2018.01317.
Toro et al., The Reverse Transcriptases Associated with CRISPR-Cas Systems. Sci Rep. Aug. 2, 2017;7(1):7089. doi: 10.1038/s41598-017-07828-y.
Torres et al., Non-integrative lentivirus drives high-frequency cre-mediated cassette exchange in human cells. PLoS One. 2011;6(5):e19794. doi: 10.1371/journal.pone.0019794. Epub May 23, 2011.
Tourdot et al., A general strategy to enhance immunogenicity of low-affinity HLA-A2. 1-associated peptides: implication in the identification of cryptic tumor epitopes. Eur J Immunol. Dec. 2000;30(12):3411-21.
Townsend et al., Role of HFE in iron metabolism, hereditary haemochromatosis, anaemia of chronic disease, and secondary iron overload. Lancet. Mar. 2, 2002;359(9308):786-90. doi: 10.1016/S0140-6736(02)07885-6.
Tracewell et al., Directed enzyme evolution: climbing fitness peaks one amino acid at a time. Curr Opin Chem Biol. Feb. 2009;13(1):3-9. doi: 10.1016/j.cbpa.2009.01.017. Epub Feb. 25, 2009.
Tratschin et al., A human parvovirus, adeno-associated virus, as a eucaryotic vector: transient expression and encapsidation of the procaryotic gene for chloramphenicol acetyltransferase. Mol Cell Biol. Oct. 1984;4(10):2072-81. doi: 10.1128/mcb.4.10.2072.
Tratschin et al., Adeno-associated virus vector for high-frequency integration, expression, and rescue of genes in mammalian cells. Mol Cell Biol. Nov. 1985;5(11):3251-60. doi: 10.1128/mcb.5.11.3251.
Trausch et al., The structure of a tetrahydrofolate-sensing riboswitch reveals two ligand binding sites in a single aptamer. Structure. Oct. 12, 2011;19(10):1413-23. doi: 10.1016/j.str.2011.06.019. Epub Sep. 8, 2011.
Traxler et al., A genome-editing strategy to treat β-hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition. Nat Med. Sep. 2016;22(9):987-90. doi: 10.1038/nm.4170. Epub Aug. 15, 2016.
Trojan et al., Functional analysis of hMLH1 variants and HNPCC-related mutations using a human expression system. Gastroenterology. Jan. 2002;122(1):211-9. doi: 10.1053/gast.2002.30296.
Trudeau et al., On the Potential Origins of the High Stability of Reconstructed Ancestral Proteins. Mol Biol Evol. Oct. 2016;33(10):2633-41. doi: 10.1093/molbev/msw138. Epub Jul. 12, 2016.
Truong et al., Development of an intein-mediated split-Cas9 system for gene therapy. Nucleic Acids Res. Jul. 27, 2015;43(13):6450-8. doi: 10.1093/nar/gkv601. Epub Jun. 16, 2015 With Supplementary Data.
Tsai et al., CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR-Cas9 nuclease off-targets. Nat Methods. Jun. 2017; 14(6):607-614. doi: 10.1038/nmeth.4278. Epub May 1, 2017.
Tsai et al., Dimeric CRISPR RNA-guided FokI nucleases for highly specific genome editing. Nat Biotechnol. Jun. 2014;32(6):569-76. doi: 10.1038/nbt.2908. Epub Apr. 25, 2014.
Tsai et al., GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat Biotechnol. Feb. 2015;33(2):187-97. doi: 10.1038/nbt.3117. Epub Dec. 16, 2014.
Tsang et al., Specialization of the DNA-cleaving activity of a group I ribozyme through in vitro evolution. J Mol Biol. Sep. 13, 1996;262(1):31-42. doi: 10.1006/jmbi.1996.0496.
Tsutakawa et al., Human flap endonuclease structures, DNA double-base flipping, and a unified understanding of the FEN1 superfamily. Cell. Apr. 15, 2011;145(2): 198-211. doi: 10.1016/j.cell.2011.03.004.
Turan et al., Recombinase-mediated cassette exchange (RMCE)—a rapidly-expanding toolbox for targeted genomic modifications. Gene. Feb. 15, 2013;515(1):1-27. doi: 10.1016/j.gene.2012.11.016. Epub Nov. 29, 2012.
Turan et al., Recombinase-mediated cassette exchange (RMCE): traditional concepts and current challenges. J Mol Biol. Mar. 25, 2011;407(2):193-221. doi: 10.1016/j.jmb.2011.01.004. Epub Jan. 15, 2011.
Turan et al., Site-specific recombinases: from tag-and-target- to tag-and-exchange-based genomic modifications. FASEB J. Dec. 2011;25(12):4088-107. doi: 10.1096/fj.11-186940. Epub Sep. 2, 2011. Review.
Tycko et al., Pairwise library screen systematically interrogates Staphylococcus aureus Cas9 specificity in human cells. bioRxiv. doi: https://doi.org/10.1101/269399 Posted Feb. 22, 2018.
UNIPROT Consortium, UniProt: the universal protein knowledgebase. Nucleic Acids Res. Mar. 16, 2018;46(5):2699. doi: 10.1093/nar/gky092. Erratum for: Nucleic Acids Res. Jan. 4, 2017;45(D1):D158-D169.
UNIPROT Submission; UniProt, Accession No. P01011. Last modified Jun. 11, 2014, version 2. 15 pages.
UNIPROT Submission; UniProt, Accession No. P01011. Last modified Sep. 18, 2013, version 2. 15 pages.
UNIPROT Submission; UniProt, Accession No. P04264. Last modified Jun. 11, 2014, version 6. 15 pages.
UNIPROT Submission; UniProt, Accession No. P04275. Last modified Jul. 9, 2014, version 107. 29 pages.
UniProtein A0A1V6. Dec. 11, 2019.
UNIPROTKB Submission; Accession No. F0NH53. May 3, 2011. 4 pages.
UNIPROTKB Submission; Accession No. F0NN87. May 3, 2011. 4 pages.
UNIPROTKB Submission; Accession No. G3ECR1.2. No Author Listed., Aug. 12, 2020, 8 pages.
UNIPROTKB Submission; Accession No. P04264. No Author Listed., Apr. 7, 2021. 12 pages.
UNIPROTKB Submission; Accession No. P0DOC6. No Author Listed., Oct. 5, 2016. 5 pages.
UNIPROTKB Submission; Accession No. T0D7A2. Oct. 16, 2013. 10 pages.
UNIPROTKB Submission; Accession No. U2UMQ6. No Author Listed., Apr. 7, 2021, 11 pages.
Urasaki et al., Functional dissection of the Tol2 transposable element identified the minimal cis-sequence and a highly repetitive sequence in the subterminal region essential for transposition. Genetics. Oct. 2006;174(2):639-49. doi: 10.1534/genetics.106.060244. Epub Sep. 7, 2006.
Urnov et al., Genome editing with engineered zinc finger nucleases. Nat Rev Genet. Sep. 2010;11(9):636-46. doi: 10.1038/nrg2842.
Urnov et al., Highly efficient endogenous human gene correction using designed zinc-finger nucleases. Nature. Jun. 2, 2005;435(7042):646-51. Epub Apr. 3, 2005.
Usman et al., Exploiting the chemical synthesis of RNA. Trends Biochem Sci. Sep. 1992;17(9):334-9. doi: 10.1016/0968-0004(92)90306-t.
Vagner et al., Efficiency of homologous DNA recombination varies along the Bacillus subtilis chromosome. J Bacteriol. Sep. 1988;170(9):3978-82.
Vakulskas et al., A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human hematopoietic stem and progenitor cells. Nat Med. Aug. 2018;24(8):1216-1224. doi: 10.1038/s41591-018-0137-0. Epub Aug. 6, 2018.
Van Brunt et al., Genetically Encoded Azide Containing Amino Acid in Mammalian Cells Enables Site-Specific Antibody-Drug Conjugates Using Click Cycloaddition Chemistry. Bioconjug Chem. Nov. 18, 2015;26(11):2249-60. doi: 10.1021/acs.bioconjchem.5b00359. Epub Sep. 11, 2015.
Van Brunt et al., Molecular Farming: Transgenic Animals as Bioreactors. Biotechnology (NY). 1988;6(10):1149-1154. doi: 10.1038/nbt1088-1149.
Van Den Oord et al., Pixel Recurrent Neural Networks. Proceedings of the 33rd International Conference on Machine Learning. Journal of Machine Learning Research. Aug. 19, 2016. vol. 48. 11 pages.
Van Duyne et al., Teaching Cre to follow directions. Proc Natl Acad Sci U S A. Jan. 6, 2009;106(1):4-5. doi: 10.1073/pnas.0811624106. Epub Dec. 31, 2008.
Van Overbeek et al., DNA Repair Profiling Reveals Nonrandom Outcomes at Cas9-Mediated Breaks. Mol Cell. Aug. 18, 2016;63(4):633-646. doi: 10.1016/j.molcel.2016.06.037. Epub Aug. 4, 2016.
Van Swieten et al., A mutation in the fibroblast growth factor 14 gene is associated with autosomal dominant cerebellar ataxia [corrected]. Am J Hum Genet. Jan. 2003;72(1):191-9. Epub Dec. 13, 2002.
Van Wijk et al., Identification of 51 novel exons of the Usher syndrome type 2A (USH2A) gene that encode multiple conserved functional domains and that are mutated in patients with Usher syndrome type II. Am J Hum Genet. Apr. 2004;74(4):738-44. doi: 10.1086/383096. Epub Mar. 10, 2004.
Vanamee et al., FokI requires two specific DNA sites for cleavage. J Mol Biol. May 25, 2001;309(1):69-78.
Varga et al., Progressive vascular smooth muscle cell defects in a mouse model of Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci U S A. Feb. 28, 2006;103(9):3250-5. doi: 10.1073/pnas.0600012103. Epub Feb. 21, 2006.
Varshney et al., The regulation and functions of DNA and RNA G-quadruplexes. Nat Rev Mol Cell Biol. Aug. 2020;21(8):459-474. doi: 10.1038/s41580-020-0236-x. Epub Apr. 20, 2020.
Vellore et al., A group II intron-type open reading frame from the thermophile Bacillus (Geobacillus) stearothermophilus encodes a heat-stable reverse transcriptase. Appl Environ Microbiol. Dec. 2004;70(12):7140-7. doi: 10.1128/AEM.70.12.7140-7147.2004.
Venken et al., Genome-wide manipulations of Drosophila melanogaster with transposons, Flp recombinase, and ΦC31 integrase. Methods Mol Biol. 2012;859:203-28. doi: 10.1007/978-1-61779-603-6_12.
Verma, The reverse transcriptase. Biochim Biophys Acta. Mar. 21, 1977;473(1):1-38. doi: 10.1016/0304-419x(77)90005-1.
Vidal et al., Yeast forward and reverse ‘n’-hybrid systems. Nucleic Acids Res. Feb. 15, 1999;27(4):919-29. doi: 10.1093/nar/27.4.919.
Vigne et al., Third-generation adenovectors for gene therapy. Restor Neurol Neurosci. Jan. 1, 1995;8(1):35-6. doi: 10.3233/RNN-1995-81208.
Vik et al., Endonuclease V cleaves at inosines in RNA. Nat Commun. 2013;4:2271. doi: 10.1038/ncomms3271.
Vilenchik et al., Endogenous DNA double-strand breaks: production, fidelity of repair, and induction of cancer. Proc Natl Acad Sci U S A. Oct. 28, 2003;100(22):12871-6. doi: 10.1073/pnas.2135498100. Epub Oct. 17, 2003.
Villiger et al., Treatment of a metabolic liver disease by in vivo genome base editing in adult mice. Nat Med. Oct. 2018;24(10):1519-1525. doi: 10.1038/s41591-018-0209-1. Epub Oct. 8, 2018.
Vitreschak et al., Regulation of the vitamin B12 metabolism and transport in bacteria by a conserved RNA structural element. RNA. Sep. 2003;9(9):1084-97.
Voigt et al., Rational evolutionary design: the theory of in vitro protein evolution. Adv Protein Chem. 2000;55:79-160.
Vriend et al., Nick-initiated homologous recombination: Protecting the genome, one strand at a time. DNA Repair (Amst). Feb. 2017;50:1-13. doi: 10.1016/j.dnarep.2016.12.005. Epub Dec. 29, 2016.
Wacey et al., Disentangling the perturbational effects of amino acid substitutions in the DNA- binding domain of p53. Hum Genet. Jan. 1999; 104(1):15-22.
Wadia et al., Modulation of cellular function by TAT mediated transduction of full length proteins. Curr Protein Pept Sci. Apr. 2003;4(2):97-104.
Wadia et al., Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nat Med. Mar. 2004; 10(3):310-5. Epub Feb. 8, 2004.
Wah et al., Structure of FokI has implications for DNA cleavage. Proc Natl Acad Sci U S A. Sep. 1, 1998;95(18):10564-9.
Wals et al., Unnatural amino acid incorporation in E. coli: current and future applications in the design of therapeutic proteins. Front Chem. Apr. 1, 2014;2:15. doi: 10.3389/fchem.2014.00015. eCollection 2014.
Walton et al., Unconstrained genome targeting with near-PAMless engineered CRISPR-Cas9 variants. Science. Apr. 17, 2020;368(6488):290-296. doi: 10.1126/science.aba8853. Epub Mar. 26, 2020.
Wan et al., Material solutions for delivery of CRISPR/Cas-based genome editing tools: Current status and future outlook. Materials Today. Jun. 2019;26:40-66. doi: 10.1016/j.mattod.2018.12.003.
Wang et al. CRISPR-Cas9 and CRISPR-Assisted Cytidine Deaminase Enable Precise and Efficient Genome Editing in Klebsiella pneumoniae. Appl Environ Microbiol. 2018;84(23):e01834-18. Published Nov. 15, 2018. doi:10.1128/AEM.01834-18.
Wang et al., AID upmutants isolated using a high-throughput screen highlight the immunity/cancer balance limiting DNA deaminase activity. Nat Struct Mol Biol. Jul. 2009;16(7):769-76. doi: 10.1038/nsmb.1623. Epub Jun. 21, 2009.
Wang et al., Continuous directed evolutions of proteins with improved soluble expression. Nature Chemical Biology. Nat Publishing Group. Aug. 20, 2018; 14(10):972-980.
Wang et al., CRISPR-Cas9 Targeting of PCSK9 in Human Hepatocytes In Vivo-Brief Report. Arterioscler Thromb Vasc Biol. May 2016;36(5):783-6. doi: 10.1161/ATVBAHA.116.307227. Epub Mar. 3, 2016.
Wang et al., Efficient delivery of genome-editing proteins using bioreducible lipid nanoparticles. Proc Natl Acad Sci U S A. Feb. 29, 2016. pii: 201520244. [Epub ahead of print].
Wang et al., Enhanced base editing by co-expression of free uracil DNA glycosylase inhibitor. Cell Res. Oct. 2017;27(1):1289-92. doi: 10.1038/cr.2017.111. Epub Aug. 29, 2017.
Wang et al., Evolution of new nonantibody proteins via iterative somatic hypermutation. Proc Natl Acad Sci U S A. Nov. 30, 2004;101(48):16745-9. Epub Nov. 19, 2004.
Wang et al., Expanding the genetic code. Annu Rev Biophys Biomol; Struct. 2006;35:225-49. Review.
Wang et al., Genetic screens in human cells using the CRISPR-Cas9 system. Science. Jan. 3, 2014;343(6166):80-4. doi: 10.1126/science.1246981. Epub Dec. 12, 2013.
Wang et al., Highly efficient CRISPR/HDR-mediated knock-in for mouse embryonic stem cells and zygotes. Biotechniques. 2015:59,201-2;204;206-8.
Wang et al., N(6)-methyladenosine Modulates Messenger RNA Translation Efficiency. Cell. Jun. 4, 2015;161(6):1388-99. doi: 10.1016/j.cell.2015.05.014.
Wang et al., N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. Jan. 2, 2014;505(7481):117-20. doi: 10.1038/nature12730. Epub Nov. 27, 2013.
Wang et al., Nucleation, propagation and cleavage of target RNAs in Ago silencing complexes. Nature. Oct. 8, 2009;461(7265):754-61. doi: 10.1038/nature08434.
Wang et al., One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell. May 9, 2013;153(4):910-8. doi: 10.1016/j.cell.2013.04.025. Epub May 2, 2013.
Wang et al., Optimized paired-sgRNA/Cas9 cloning and expression cassette triggers high- efficiency multiplex genome editing in kiwifruit. Plant Biotechnol J. Aug. 2018;16(8):1424-1433. doi: 10.1111/pbi.12884. Epub Feb. 6, 2018.
Wang et al., Programming cells by multiplex genome engineering and accelerated evolution. Nature. Aug. 13, 2009;460(7257):894-8. Epub Jul. 26, 2009.
Wang et al., Reading RNA methylation codes through methyl-specific binding proteins. RNA Biol. 2014;11(6):669-72. doi: 10.4161/rna.28829. Epub Apr. 24, 2014.
Wang et al., Recombinase technology: applications and possibilities. Plant Cell Rep. Mar. 2011;30(3):267-85. doi: 10.1007/s00299-010-0938-1. Epub Oct. 24, 2010.
Wang et al., Riboswitches that sense S-adenosylhomocysteine and activate genes involved in coenzyme recycling. Mol Cell. Mar. 28, 2008;29(6):691-702. doi: 10.1016/j.molcel.2008.01.012.
Wang et al., Staphylococcus aureus protein SAUGI acts as a uracil-DNA glycosylase inhibitor. Nucleic Acids Res. Jan. 2014;42(2):1354-64. doi: 10.1093/nar/gkt964. Epub Oct. 22, 2013.
Wang et al., Structural basis of N(6)-adenosine methylation by the METTL3-METTL14 complex. Nature. Jun. 23, 2016;534(7608):575-8. doi: 10.1038/nature18298. Epub May 25, 2016.
Wang et al., Targeted gene addition to a predetermined site in the human genome using a ZFN-based nicking enzyme. Genome Res. Jul. 2012;22(7):1316-26. doi: 10.1101/gr.122879.111. Epub Mar. 20, 2012.
Wang et al., Uracil-DNA glycosylase inhibitor gene of bacteriophage PBS2 encodes a binding protein specific for uracil-DNA glycosylase. J Biol Chem. Jan. 15, 1989;264(2):1163-71.
Warren et al., A chimeric Cre recombinase with regulated directionality. Proc Natl Acad Sci U S A. Nov. 25, 2008;105(47):18278-83. doi: 10.1073/pnas.0809949105. Epub Nov. 14, 2008.
Warren et al., Mutations in the amino-terminal domain of lambda-integrase have differential effects on integrative and excisive recombination. Mol Microbiol. Feb. 2005;55(4):1104-12.
Watowich, The erythropoietin receptor: molecular structure and hematopoietic signaling pathways. J Investig Med. Oct. 2011;59(7):1067-72. doi: 10.2310/JIM.0b013e31820fb28c.
Waxman et al., Regulating excitability of peripheral afferents: emerging ion channel targets. Nat Neurosci. Feb. 2014;17(2):153-63. doi: 10.1038/nn.3602. Epub Jan. 28, 2014.
Weber et al., Assembly of designer TAL effectors by Golden Gate cloning. PLoS One. 2011;6(5):e19722. doi:10.1371/journal.pone.0019722. Epub May 19, 2011.
Weill et al., DNA polymerases in adaptive immunity. Nat Rev Immunol. Apr. 2008;8(4):302-12. doi: 10.1038/nri2281. Epub Mar. 14, 2008.
Weinberg et al., New Classes of Self-Cleaving Ribozymes Revealed by Comparative Genomics Analysis. Nat Chem Biol. Aug. 2015;11(8):606-10. doi: 10.1038/nchembio.1846. Epub Jul. 13, 2015.
Weinberg et al., The aptamer core of SAM-IV riboswitches mimics the ligand-binding site of SAM-I riboswitches. RNA. May 2008;14(5):822-8. doi: 10.1261/rna.988608. Epub Mar. 27, 2008.
Weinberger et al., Disease-causing mutations C277R and C277Y modify gating of human CIC-1 chloride channels in myotonia congenita. J Physiol. Aug. 1, 2012;590(Pt 15):3449-64. doi: 0.1113/jphysiol.2012.232785. Epub May 28, 2012.
Weinert et al., Unbiased detection of CRISPR off-targets in vivo using DISCOVER-Seq. Science. Apr. 19, 2019;364(6437):286-289. doi: 10.1126/science.aav9023. Epub Apr. 18, 2019.
Weiss et al., Loss-of-function mutations in sodium channel Nav1.7 cause anosmia. Nature. Apr. 14, 2011;472(7342):186-90. doi: 10.1038/nature09975. Epub Mar. 23, 2011.
Wen et al., Inclusion of a universal tetanus toxoid CD4(+) T cell epitope P2 significantly enhanced the immunogenicity of recombinant rotavirus ΔVP8* subunit parenteral vaccines. Vaccine. Jul. 31, 2014;32(35):4420-4427. doi: 10.1016/j.vaccine.2014.06.060. Epub Jun. 21, 2014.
West et al., Gene expression in adeno-associated virus vectors: the effects of chimeric mRNA structure, helper virus, and adenovirus VA1 RNA. Virology. Sep. 1987;160(1):38-47. doi: 10.1016/0042-6822(87)90041-9.
Wharton et al., A new-specificity mutant of 434 repressor that defines an amino acid-base pair contact. Nature. Apr. 30-May 6, 1987;326(6116):888-91.
Wharton et al., Changing the binding specificity of a repressor by redesigning an alpha- helix. Nature. Aug. 15-21, 1985;316(6029):601-5.
Wheeler et al., The thermostability and specificity of ancient proteins. Curr Opin Struct Biol. Jun. 2016;38:37-43. doi: 10.1016/j.sbi.2016.05.015. Epub Jun. 9, 2016.
Wiedenheft et al., RNA-guided genetic silencing systems in bacteria and archaea. Nature. Feb. 15, 2012;482(7385):331-8. doi: 10.1038/nature10886. Review.
Wienert et al., KLF1 drives the expression of fetal hemoglobin in British HPFH. Blood. Aug. 10, 2017;130(6):803-807. doi: 10.1182/blood-2017-02-767400. Epub Jun. 28, 2017.
Wijesinghe et al., Efficient deamination of 5-methylcytosines in DNA by human APOBEC3A, but not by AID or APOBEC3G. Nucleic Acids Res. Oct. 2012;40(18):9206-17. doi: 10.1093/nar/gks685. Epub Jul. 13, 2012.
Wijnker et al., Managing meiotic recombination in plant breeding. Trends Plant Sci. Dec. 2008;13(12):640-6. doi: 10.1016/j.tplants.2008.09.004. Epub Oct. 22, 2008.
Williams et al., Assessing the accuracy of ancestral protein reconstruction methods. PLoS Comput Biol. Jun. 23, 2006;2(6):e69. doi: 10.1371/journal.pcbi.0020069. Epub Jun. 23, 2006.
Wills et al., Pseudoknot-dependent read-through of retroviral gag termination codons: importance of sequences in the spacer and loop 2. EMBO J. Sep. 1, 1994;13(17):4137-44. doi: 10.1002/j.1460-2075.1994.tb06731.x.
Wilson et al., Assessing annotation transfer for genomics: quantifying the relations between protein sequence, structure and function through traditional and probabilistic scores. J Mol Biol 2000;297:233-49.
Wilson et al., Formation of infectious hybrid virions with gibbon ape leukemia virus and human T-cell leukemia virus retroviral envelope glycoproteins and the gag and pol proteins of Moloney murine leukemia virus. J Virol. May 1989;63(5):2374-8. doi: 10.1128/JVI.63.5.2374-2378.1989.
Wilson et al., In Vitro Selection of Functional Nucleic Acids. Annu Rev Biochem. 1999;68:611-47. doi: 10.1146/annurev.biochem.68.1.611.
Wilson et al., Kinase dynamics. Using ancient protein kinases to unravel a modern cancer drug's mechanism. Science. Feb. 20, 2015;347(6224):882-6. doi: 10.1126/science.aaa1823.
Wilson et al., Programmable m6A modification of cellular RNAs with a Cas13-directed methyltransferase. Nat Biotechnol. Dec. 2020;38(12):1431-1440. doi: 10.1038/s41587-020-0572-6. Epub Jun. 29, 2020.
Winkler et al., An mRNA structure that controls gene expression by binding FMN. Proc Natl Acad Sci U S A. Dec. 10, 2002;99(25):15908-13. Epub Nov. 27, 2002.
Winkler et al., Control of gene expression by a natural metabolite-responsive ribozyme. Nature. Mar. 18, 2004;428(6980):281-6.
Winkler et al., Thiamine derivatives bind messenger RNAs directly to regulate bacterial gene expression. Nature. Oct. 31, 2002;419(6910):952-6. Epub Oct. 16, 2002.
Winoto et al., A novel, inducible and T cell-specific enhancer located at the 3′ end of the T cell receptor alpha locus. Embo J. Mar. 1989;8(3):729-33.
Winter et al., Drug Development. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science. Jun. 19, 2015;348(6241):1376-81. doi:; 10.1126/science.aab1433. Epub May 21, 2015.
Winter et al., Targeted exon skipping with AAV-mediated split adenine base editors. Cell Discov. Aug. 20, 2019;5:41. doi: 10.1038/s41421-019-0109-7.
Wirth et al., Mildly affected patients with spinal muscular atrophy are partially protected by an increased SMN2 copy number. Hum Genet. May 2006;119(4):422-8. doi: 10.1007/s00439-006-0156-7. Epub Mar. 1, 2006.
Wold, Replication protein A: a heterotrimeric, single-stranded DNA-binding protein required for eukaryotic DNA metabolism. Annu Rev Biochem. 1997;66:61-92. doi: 10.1146/annurev.biochem.66.1.61.
Wolf et al., tadA, an essential tRNA-specific adenosine deaminase from Escherichia coli. EMBO J. Jul. 15, 2002;21(14):3841-51.
Wolfe et al., Analysis of zinc fingers optimized via phage display: evaluating the utility of a recognition code. J Mol Biol. Feb. 5, 1999;285(5):1917-34.
Wong et al., A statistical analysis of random mutagenesis methods used for directed protein evolution. J Mol Biol. Jan. 27, 2006;355(4):858-71. Epub Nov. 17, 2005.
Wong et al., The Diversity Challenge in Directed Protein Evolution. Comb Chem High Throughput Screen. May 2006;9(4):271-88.
Woo et al., Gene activation of SMN by selective disruption of IncRNA-mediated recruitment of PRC2 for the treatment of spinal muscular atrophy. Proc Natl Acad Sci U S A. Feb. 21, 2017;114(8):E1509-E1518. doi:10.1073/pnas.1616521114. Epub Feb. 13, 2017.
Wood et al., A genetic system yields self-cleaving inteins for bioseparations. Nat Biotechnol. Sep. 1999;17(9):889-92. doi: 10.1038/12879.
Wood et al., Targeted genome editing across species using ZFNs and TALENs. Science. Jul. 15, 2011;333(6040):307. doi: 10.1126/science.1207773. Epub Jun. 23, 2011.
Woods et al., The phenotype of congenital insensitivity to pain due to the NaV1.9 variant p.L811P. Eur J Hum Genet. May 2015;23(5):561-3. doi: 10.1038/ejhg.2014.166. Epub Aug. 13, 2014.
Wright et al., Continuous in vitro evolution of catalytic function. Science. Apr. 25, 1997;276(5312):614-7.
Wright et al., Rational design of a split-Cas9 enzyme complex. Proc Natl Acad Sci U S A. Mar. 10, 2015;112(10):2984-9. doi: 10.1073/pnas.1501698112. Epub Feb. 23, 2015.
Wu et al., A novel SCN9A mutation responsible for primary erythromelalgia and is resistant to the treatment of sodium channel blockers. PLoS One. 2013;8(1):e55212. doi: 10.1371/journal.pone.0055212. Epub Jan. 31, 2013. 15 pages.
Wu et al., Correction of a genetic disease in mouse via use of CRISPR-Cas9. Cell Stem Cell. Dec. 5, 2013;13(6):659-62. doi: 10.1016/j.stem.2013.10.016.
Wu et al., Genome-wide binding of the CRISPR endonuclease Cas9 in mammalian cells. Nat Biotechnol. Jul. 2014;32(7):670-6. doi: 10.1038/nbt.2889. Epub Apr. 20, 2014.
Wu et al., Human single-stranded DNA binding proteins: guardians of genome stability. Acta Biochim Biophys Sin (Shanghai). Jul. 2016;48(7):671-7. doi: 10.1093/abbs/gmw044. Epub May 23, 2016.
Wu et al., Protein trans-splicing and functional mini-inteins of a cyanobacterial dnaB intein. Biochim Biophys Acta. Sep. 8, 1998;1387(1-2):422-32. doi: 10.1016/s0167-4838(98)00157-5.
Wu et al., Protein trans-splicing by a split intein encoded in a split DnaE gene of Synechocystis sp. PCC6803. Proc Natl Acad Sci U S A. Aug. 4, 1998;95(16):9226-31. doi: 10.1073/pnas.95.16.9226.
Wu et al., Readers, writers and erasers of N6-methylated adenosine modification. Curr Opin Struct Biol. Dec. 2017;47:67-76. doi: 10.1016/j.sbi.2017.05.011. Epub Jun. 16, 2017.
Xiang et al., RNA m6A methylation regulates the ultraviolet-induced DNA damage response. Nature. Mar. 23, 2017;543(7646):573-576. doi: 10.1038/nature21671. Epub Mar. 15, 2017.
Xiao et al., Genetic incorporation of multiple unnatural amino acids into proteins in mammalian cells. Angew Chem Int Ed Engl. Dec. 23, 2013;52(52):14080-3. doi: 10.1002/anie.201308137. Epub Nov. 8, 2013.
Xiao et al., Nuclear m(6)A Reader YTHDC1 Regulates mRNA Splicing. Mol Cell. Feb. 18, 2016;61(4):507-519. doi: 10.1016/j.molcel.2016.01.012. Epub Feb. 11, 2016.
Xie et al., Adjusting the attB site in donor plasmid improves the efficiency of ΦC31 integrase system. DNA Cell Biol. Jul. 2012;31(7):1335-40. doi: 10.1089/dna.2011.1590. Epub Apr. 10, 2012.
Xiong et al., Origin and evolution of retroelements based upon their reverse transcriptase sequences. EMBO J. Oct. 1990;9(10):3353-62.
Xu et al., Accuracy and efficiency define Bxb1 integrase as the best of fifteen candidate serine recombinases for the integration of DNA into the human genome. BMC Biotechnol. Oct. 20, 2013;13:87. doi: 10.1186/1472-6750-13-87.
Xu et al., Chemical ligation of folded recombinant proteins: segmental isotopic labeling of domains for NMR studies. Proc Natl Acad Sci U S A. Jan. 19, 1999;96(2):388-93. doi: 10.1073/pnas.96.2.388.
Xu et al., Multiplex nucleotide editing by high-fidelity Cas9 variants with improved efficiency in rice. BMC Plant Biol. 2019;19(1):511. Published Nov. 21, 2019. doi:10.1186/s12870-019-2131-1. Includes supplementary data and materials.
Xu et al., Protein splicing: an analysis of the branched intermediate and its resolution by succinimide formation. EMBO J. Dec. 1, 1994;13(23):5517-22.
Xu et al., Ptmd: A Database of Human Disease-associated Post-translational Modifications. Genomics Proteomics Bioinformatics. Aug. 2018;16(4):244-251. doi: 10.1016/j.gpb.2018.06.004. Epub Sep. 21, 2018.
Xu et al., Sequence determinants of improved CRISPR sgRNA design. Genome Res. Aug. 2015;25(8): 1147-57. doi: 10.1101/gr.191452.115. Epub Jun. 10, 2015.
Xu et al., Structures of human ALKBH5 demethylase reveal a unique binding mode for specific single-stranded N6-methyladenosine RNA demethylation. J Biol Chem. Jun. 20, 2014;289(25):17299-311. doi: 10.1074/jbc.M114.550350. Epub Apr. 28, 2014.
Xu et al., The mechanism of protein splicing and its modulation by mutation. EMBO J. Oct. 1, 1996;15(19):5146-53.
Yahata et al., Unified, Efficient, and Scalable Synthesis of Halichondrins: Zirconium/Nickel-Mediated One-Pot Ketone Synthesis as the Final Coupling Reaction. Angew Chem Int Ed Engl. Aug. 28, 2017;56(36):10796-10800. doi: 10.1002/anie.201705523. Epub Jul. 28, 2017.
Yamada et al., Crystal Structure of the Minimal Cas9 from Campylobacter jejuni Reveals the Molecular Diversity in the CRISPR-Cas9 Systems. Mol Cell. Mar. 16, 2017;65(6):P1109-1121. /doi.org/10.1016/j.molcel.2017.02.007.
Yamamoto et al., The ons and offs of inducible transgenic technology: a review. Neurobiol Dis. Dec. 2001;8(6):923-32.
Yamamoto et al., Virological and immunological bases for HIV-1 vaccine design. Uirusu 2007;57(2):133-139. https://doi.org/10.2222/jsv.57.133.
Yamane et al., Deep-sequencing identification of the genomic targets of the cytidine deaminase AID and its cofactor RPA in B lymphocytes. Nat Immunol. Jan. 2011;12(1):62-9. doi: 10.1038/ni.1964. Epub Nov. 28, 2010.
Yamano et al., Crystal Structure of Cpf1 in Complex with Guide RNA and Target DNA. Cell. May 5, 2016;165(4):949-62 and Supplemental Info. doi: 10.1016/j.cell.2016.04.003. Epub Apr. 21, 2016.
Yamano et al., Crystal Structure of Cpf1 in Complex with Guide RNA and Target DNA. Cell. May 5, 2016;165(4):949-62. doi: 10.1016/j.cell.2016.04.003. Epub Apr. 21, 2016.
Yamazaki et al., Segmental Isotope Labeling for Protein NMR Using Peptide Splicing. J. Am. Chem. Soc. May 22, 1998; 120(22):5591-2. https://doi.org/10.1021/ja9807760.
Yan et al., Cas13d Is a Compact RNA-Targeting Type VI CRISPR Effector Positively Modulated by a WYL-Domain-Containing Accessory Protein. Mol Cell. Apr. 19, 2018;70(2):327-339.e5. doi: 10.1016/j.molcel.2018.02.028. Epub Mar. 15, 2018.
Yan et al., Functionally diverse type V CRISPR-Cas systems. Science. Jan. 4, 2019;363(6422):88-91. doi: 10.1126/science.aav7271. Epub Dec. 6, 2018.
Yan et al., Highly Efficient A⋅T to G⋅C Base Editing by Cas9n-Guided tRNA Adenosine Deaminase in Rice. Mol Plant. Apr. 2, 2018;11(4):631-634. doi: 10.1016/j.molp.2018.02.008. Epub Feb. 22, 2018.
Yang et al., A Tale of Two Moieties: Rapidly Evolving CRISPR/Cas-Based Genome Editing. Trends Biochem Sci. Oct. 2020;45(10):874-888. doi: 10.1016/j.tibs.2020.06.003. Epub Jun. 30, 2020.
Yang et al., APOBEC: From mutator to editor. J Genet Genomics. Sep. 20, 2017;44(9):423-437. doi: 10.1016/j.jgg.2017.04.009. Epub Aug. 7, 2017.
Yang et al., BRCA2 function in DNA binding and recombination from a BRCA2-DSS1-ssDNA structure. Science. Sep. 13, 2002;297(5588):1837-48. doi: 10.1126/science.297.5588.1837.
Yang et al., Construction of an integration-proficient vector based on the site-specific recombination mechanism of enterococcal temperate phage phiFC1. J Bacteriol. Apr. 2002; 184(7):1859-64. doi: 10.1128/jb.184.7.1859-1864.2002.
Yang et al., Engineering and optimising deaminase fusions for genome editing. Nat Commun. Nov. 2, 2016;7:13330. doi: 10.1038/ncomms13330.
Yang et al., Genome editing with targeted deaminases. BioRxiv. Preprint. First posted online Jul. 28, 2016.
Yang et al., Genome-wide inactivation of porcine endogenous retroviruses (PERVs). Science. Nov. 27, 2015;350(6264):1101-4. doi: 10.1126/science.aad1191. Epub Oct. 11, 2015.
Yang et al., Increasing targeting scope of adenosine base editors in mouse and rat embryos through fusion of TadA deaminase with Cas9 variants. Protein Cell. Sep. 2018;9(9):814-819. doi: 10.1007/s13238-018-0568-x.
Yang et al., Mutations in SCN9A, encoding a sodium channel alpha subunit, in patients with primary erythermalgia. J Med Genet. Mar. 2004;41(3):171-4. doi: 10.1136/jmg.2003.012153.
Yang et al., New CRISPR-Cas systems discovered. Cell Res. Mar. 2017;27(3):313-314. doi: 10.1038/cr.2017.21. Epub Feb. 21, 2017.
Yang et al., One Prime for All Editing. Cell. Dec. 12, 2019;179(7):1448-1450. doi: 10.1016/j.cell.2019.11.030.
Yang et al., One-step generation of mice carrying reporter and conditional alleles by CRISPR/Cas-mediated genome engineering. Cell. Sep. 12, 2013;154(6):1370-9. doi: 10.1016/j.cell.2013.08.022. Epub Aug. 29, 2013.
Yang et al., PAM-dependent Target DNA Recognition and Cleavage by C2C1 CRISPR-Cas endonuclease. Cell Dec. 2016; 167(7):1814-28.
Yang et al., Permanent genetic memory with >1-byte capacity. Nat Methods. Dec. 2014;11(12):1261-6. doi: 10.1038/nmeth.3147. Epub Oct. 26, 2014.
Yang et al., Preparation of RNA-directed DNA polymerase from spleens of Balb-c mice infected with Rauscher leukemia virus. Biochem Biophys Res Commun. Apr. 28, 1972;47(2):505-11. doi: 10.1016/0006-291x(72)90743-7.
Yang et al., Small-molecule control of insulin and PDGF receptor signaling and the role of membrane attachment. Curr Biol. Jan. 1, 1998;8(1):11-8. doi: 10.1016/s0960-9822(98)70015-6.
Yang et al., The BRCA2 homologue Brh2 nucleates RAD51 filament formation at a dsDNA-ssDNA junction. Nature. Feb. 10, 2005;433(7026):653-7. doi: 10.1038/nature03234.
Yang, Development of Human Genome Editing Tools for the Study of Genetic Variations and Gene Therapies. Doctoral Dissertation. Harvard University. 2013. Accessible via nrs.harvard.edu/urn-3:HUL.InstRepos:11181072. 277 pages.
Yang, Nucleases: diversity of structure, function and mechanism. Q Rev Biophys. Feb. 2011;44(1):1-93. doi: 10.1017/S0033583510000181. Epub Sep. 21, 2010.
Yang, PAML 4: phylogenetic analysis by maximum likelihood. Mol Biol Evol. Aug. 2007;24(8): 1586-91. doi: 10.1093/molbev/msm088. Epub May 4, 2007.
Yang, Phylogenetic Analysis by Maximum Likelihood (PAML). //abacus.gene.ucl.ac.uk/software/paml.html Last accessed Apr. 28, 2021.
Yanover et al., Extensive protein and DNA backbone sampling improves structure-based specificity prediction for C2H2 zinc fingers. Nucleic Acids Res. Jun. 2011;39(11):4564-76. doi: 10.1093/nar/gkr048. Epub Feb. 22, 2011.
Yasui et al., Miscoding Properties of 2′-Deoxyinosine, a Nitric Oxide-Derived DNA Adduct, during Translesion Synthesis Catalyzed by Human DNA Polymerases. J Molec Biol. Apr. 4, 2008;377(4):1015-23.
Yasui, Alternative excision repair pathways. Cold Spring Harb Perspect Biol. Jun. 1, 2013;5(6):a012617. doi: 10.1101/cshperspect.a012617.
Yasukawa et al., Characterization of Moloney murine leukaemia virus/avian myeloblastosis virus chimeric reverse transcriptases. J Biochem. Mar. 2009;145(3):315-24. doi: 10.1093/jb/mvn166. Epub Dec. 6, 2008.
Yazaki et al., Hereditary systemic amyloidosis associated with a new apolipoprotein AII stop codon mutation Stop78Arg. Kidney Int. Jul. 2003;64(1):11-6.
Yeh et al., In vivo base editing of post-mitotic sensory cells. Nat Commun. Jun. 5, 2018;9(1):2184. doi: 10.1038/s41467-018-04580-3.
Yeh et al., In vivo base editing restores sensory transduction and transiently improves auditory function in a mouse model of recessive deafness. Sci Transl Med. Jun. 3, 2020;12(546): eaay9101. doi: 10.1126/scitranslmed.aay9101.
Yin et al., Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype. Nat Biotechnol. Jun. 2014;32(6):551-3. doi: 10.1038/nbt.2884. Epub Mar. 30, 2014.
Yokoe et al., Spatial dynamics of GFP-tagged proteins investigated by local fluorescence enhancement. Nat Biotechnol. Oct. 1996; 14(10):1252-6. doi: 10.1038/nbt1096-1252.
Young et al., Beyond the canonical 20 amino acids: expanding the genetic lexicon. J Biol Chem. Apr. 9, 2010;285(15):11039-44. doi: 10.1074/jbc.R109.091306. Epub Feb. 10, 2010.
Yu et al., Circular permutation: a different way to engineer enzyme structure and function. Trends Biotechnol. Jan. 2011;29(1):18-25. doi: 10.1016/j.tibtech.2010.10.004. Epub Nov. 17, 2010.
Yu et al., Cytosine base editors with minimized unguided DNA and RNA off-target events and high on-target activity. Nat Commun. Apr. 28, 2020;11(1):2052. doi: 10.1038/s41467-020-15887-5.
Yu et al., Dynamic control of Rad51 recombinase by self-association and interaction with BRCA2. Mol Cell. Oct. 2003;12(4):1029-41. doi: 10.1016/s1097-2765(03)00394-0.
Yu et al., Liposome-mediated in vivo E1A gene transfer suppressed dissemination of ovarian cancer cells that overexpress HER-2/neu. Oncogene. Oct. 5, 1995;11(7):1383-8.
Yu et al., Progress towards gene therapy for HIV infection. Gene Ther. Jan. 1994;1(1):13-26.
Yu et al., Small molecules enhance CRISPR genome editing in pluripotent stem cells. Cell Stem Cell. Feb. 5, 2015;16(2):142-7. doi: 10.1016/j.stem.2015.01.003.
Yu et al., Synthesis-dependent microhomology-mediated end joining accounts for multiple types of repair junctions. Nucleic Acids Res. Sep. 2010;38(17):5706-17. doi: 10.1093/nar/gkq379. Epub May 11, 2010.
Yuan et al., Laboratory-directed protein evolution. Microbiol Mol Biol Rev. 2005; 69(3):373-92. PMID: 16148303.
Yuan et al., Tetrameric structure of a serine integrase catalytic domain. Structure. Aug. 6, 2008;16(8):1275-86. doi: 10.1016/j.str.2008.04.018.
Yuen et al., Control of transcription factor activity and osteoblast differentiation in mammalian cells using an evolved small-molecule-dependent intein. J Am Chem Soc. Jul. 12, 2006;128(27):8939-46.
Zakas et al., Enhancing the pharmaceutical properties of protein drugs by ancestral sequence reconstruction. Nat Biotechnol. Jan. 2017;35(1):35-37. doi: 10.1038/nbt.3677. Epub Sep. 26, 2016.
Zalatan et al., Engineering complex synthetic transcriptional programs with CRISPR RNA scaffolds. Cell. Jan. 15, 2015;160(1-2):339-50. doi: 10.1016/j.cell.2014.11.052. Epub Dec. 18, 2014.
Zelphati et al., Intracellular delivery of proteins with a new lipid-mediated delivery system. J Biol Chem. Sep. 14, 2001;276(37):35103-10. Epub Jul. 10, 2001.
Zeng et al., Correction of the Marfan Syndrome Pathogenic FBN1 Mutation by Base Editing in Human Cells and Heterozygous Embryos. Mol Ther. Nov. 7, 2018;26(11):2631-2637. doi: 10.1016/j.ymthe.2018.08.007. Epub Aug. 14, 2018.
Zetsche et al., A split-Cas9 architecture for inducible genome editing and transcription modulation. Nat Biotechnol. Feb. 2015;33(2):139-42. doi: 10.1038/nbt.3149.
Zetsche et al., Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell. Oct. 22, 2015;163(3):759-71 and Supplemental Info. doi: 10.1016/j.cell.2015.09.038. Epub Sep. 25, 2015.
Zetsche et al., Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell. Oct. 22, 2015;163(3):759-71. doi: 10.1016/j.cell.2015.09.038. Epub Sep. 25, 2015.
Zettler et al., The naturally split Npu DnaE intein exhibits an extraordinarily high rate in the protein trans-splicing reaction. FEBS Lett. Mar. 4, 2009;583(5):909-14. doi: 10.1016/j.febslet.2009.02.003. Epub Feb. 10, 2009.
Zhang et al., II-Clamp-mediated cysteine conjugation. Nat Chem. Feb. 2016;8(2):120-8. doi: 10.1038/nchem.2413. Epub Dec. 21, 2015.
Zhang et al., A new strategy for the site-specific modification of proteins in vivo. Biochemistry. Jun. 10, 2003;42(22):6735-46.
Zhang et al., Circular intronic long noncoding RNAs. Mol Cell. Sep. 26, 2013;51(6):792-806. doi: 10.1016/j.molcel.2013.08.017. Epub Sep. 12, 2013.
Zhang et al., Comparison of non-canonical PAMs for CRISPR/Cas9-mediated DNA cleavage in human cells. Sci Rep. Jun. 2014;4:5405.
Zhang et al., Conditional gene manipulation: Cre-ating a new biological era. J Zhejiang Univ Sci B. Jul. 2012; 13(7):511-24. doi: 10.1631/jzus.B1200042. Review.
Zhang et al., Copy number variation in human health, disease, and evolution. Annu Rev Genomics Hum Genet. 2009;10:451-81. doi: 10.1146/annurev.genom.9.081307.164217.
Zhang et al., CRISPR/Cas9 for genome editing: progress, implications and challenges. Hum Mol Genet. Sep. 15, 2014;23(R1):R40-6. doi: 10.1093/hmg/ddu125. Epub Mar. 20, 2014.
Zhang et al., Efficient construction of sequence-specific TAL effectors for modulating mammalian transcription. Nat Biotechnol. Feb. 2011;29(2):149-53. doi: 10.1038/nbt.1775. Epub Jan. 19, 2011.
Zhang et al., Efficient precise knockin with a double cut HDR donor after CRISPR/Cas9-mediated double-stranded DNA cleavage. Genome Biol. Feb. 20, 2017;18(1):35. doi: 10.1186/s13059-017-1164-8.
Zhang et al., Global analysis of small RNA and mRNA targets of Hfq. Mol Microbiol. Nov. 2003;50(4):1111-24. doi: 10.1046/j.1365-2958.2003.03734.x.
Zhang et al., Large genomic fragment deletions and insertions in mouse using CRISPR/Cas9. PLoS One. Mar. 24, 2015;10(3):e0120396. doi: 10.1371/journal.pone.0120396. 14 pages.
Zhang et al., Myoediting: Toward Prevention of Muscular Dystrophy by Therapeutic Genome Editing. Physiol Rev. Jul. 1, 2018;98(3):1205-1240. doi: 10.1152/physrev.00046.2017.
Zhang et al., Programmable base editing of zebrafish genome using a modified CRISPR-Cas9 system. Nat Commun. Jul. 25, 2017;8(1):118. doi: 10.1038/s41467-017-00175-6.
Zhang et al., Reversible RNA Modification N1-methyladenosine (m1A) in mRNA and tRNA. Genomics Proteomics Bioinformatics. Jun. 2018;16(3):155-161. doi: 10.1016/j.gpb.2018.03.003. Epub Jun. 14, 2018.
Zhang et al., Ribozymes and Riboswitches: Modulation of RNA Function by Small Molecules. Biochemistry. Nov. 2, 2010;49(43):9123-31. doi: 10.1021/bi1012645.
Zhang et al., Stabilized plasmid-lipid particles for regional gene therapy: formulation and transfection properties. Gene Ther. Aug. 1999;6(8):1438-47.
Zhao et al., An ultraprocessive, accurate reverse transcriptase encoded by a metazoan group II intron. RNA. Feb. 2018;24(2):183-195. doi: 10.1261/rna.063479.117. Epub Nov. 6, 2017.
Zhao et al., Crystal structures of a group II intron maturase reveal a missing link in spliceosome evolution. Nat Struct Mol Biol. Jun. 2016;23(6):558-65. doi: 10.1038/nsmb.3224. Epub May 2, 2016.
Zhao et al., Post-transcriptional gene regulation by mRNA modifications. Nat Rev Mol Cell Biol. Jan. 2017;18(1):31-42. doi: 10.1038/nrm.2016.132. Epub Nov. 3, 2016.
Zheng et al., ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. Jan. 10, 2010;49(1):18-29. doi: 10.1016/j.molcel.2012.10.015. Epub Nov. 21, 2012.
Zheng et al., DNA editing in DNA/RNA hybrids by adenosine deaminases that act on RNA. Nucleic Acids Res. Apr. 7, 2017;45(6):3369-3377. doi: 10.1093/nar/gkx050.
Zheng et al., Highly efficient base editing in bacteria using a Cas9-cytidine deaminase fusion. Commun Biol. Apr. 19, 2018;1:32. doi: 10.1038/s42003-018-0035-5.
Zheng et al., Structural basis for the complete resistance of the human prion protein mutant G127V to prion disease. Sci Rep. Sep. 4, 2018;8(1):13211. doi: 10.1038/s41598-018-31394-6.
Zhong et al., Rational Design of Aptazyme Riboswitches for Efficient Control of Gene Expression in Mammalian Cells. Elife. Nov. 2, 2016;5:e18858. doi: 10.7554/eLife.18858.
Zhou et al., Cas12a variants designed for lower genome-wide off-target effect through stringent PAM recognition. Mol Ther. Jan. 5, 2022;30(1):244-255. doi: 10.1016/j.ymthe.2021.10.010. Epub Oct. 20, 2021.
Zhou et al., Dynamic m(6)A mRNA methylation directs translational control of heat shock response. Nature. Oct. 22, 2015;526(7574):591-4. doi: 10.1038/nature15377. Epub Oct. 12, 2015.
Zhou et al., GISSD: Group I Intron Sequence and Structure Database. Nucleic Acids Res. Jan. 2008;36(Database issue):D31-7. doi: 10.1093/nar/gkm766. Epub Oct. 16, 2007.
Zhou et al., Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature. Jul. 2019;571(7764):275-278. doi: 10.1038/s41586-019-1314-0. Epub Jun. 10, 2019.
Zhou et al., Protective V127 prion variant prevents prion disease by interrupting the formation of dimer and fibril from molecular dynamics simulations. Sci Rep. Feb. 24, 2016;6:21804. doi: 10.1038/srep21804.
Zhou et al., Seamless Genetic Conversion of SMN2 to SMN1 via CRISPR/Cpf1 and Single-Stranded Oligodeoxynucleotides in Spinal Muscular Atrophy Patient-Specific Induced Pluripotent Stem Cells. Hum Gene Ther. Nov. 2018;29(11):1252-1263. doi: 10.1089/hum.2017.255. Epub May 9, 2018.
Zielenski, Genotype and phenotype in cystic fibrosis. Respiration. 2000;67(2):117-33. doi: 10.1159/000029497.
Zimmerly et al., An Unexplored Diversity of Reverse Transcriptases in Bacteria. Microbiol Spectr. Apr. 2015;3(2):MDNA3-0058-2014. doi: 10.1128/microbiolspec.MDNA3-0058-2014.
Zimmerly et al., Group II intron mobility occurs by target DNA-primed reverse transcription. Cell. Aug. 25, 1995;82(4):545-54. doi: 10.1016/0092-8674(95)90027-6.
Zimmermann et al., Molecular interactions and metal binding in the theophylline-binding core of an RNA aptamer. RNA. May 2000;6(5):659-67.
Zolotukhin et al., Production and purification of serotype 1, 2, and 5 recombinant adeno- associated viral vectors. Methods. Oct. 2002;28(2):158-67. doi: 10.1016/s1046-2023(02)00220-7.
Zong et al., Precise base editing in rice, wheat and maize with a Cas9-cytidine deaminase fusion. Nat Biotechnol. May 2017;35(5):438-440. doi: 10.1038/nbt.3811. Epub Feb. 27, 2017.
Zorko et al., Cell-penetrating peptides: mechanism and kinetics of cargo delivery. Adv Drug Deliv Rev. Feb. 28, 2005;57(4):529-45. Epub Jan. 22, 2005.
Zou et al., Gene targeting of a disease-related gene in human induced pluripotent stem and embryonic stem cells. Cell Stem Cell. Jul. 2, 2009;5(1):97-110. doi: 10.1016/j.stem.2009.05.023. Epub Jun. 18, 2009.
Zufferey et al., Woodchuck hepatitis virus posttranscriptional regulatory element enhances expression of transgenes delivered by retroviral vectors. J Virol. Apr. 1999;73(4):2886-92. doi: 10.1128/JVI.73.4.2886-2892.1999.
Zuker et al., Optimal computer folding of large RNA sequences using thermodynamics and auxiliary information. Nucleic Acids Res. Jan. 10, 1981;9(1):133-48. doi: 10.1093/nar/9.1.133.
Zuo et al., Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science. Apr. 19, 2019;364(6437):289-292. doi: 10.1126/science.aav9973. Epub Feb. 28, 2019.
Zuris et al., Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat Biotechnol. 2015;33:73-80.
[No Author Listed], Nav1.7 (SCN9A) (NM_002977) Human Tagged ORF Clone. OriGene. Retrieved from https://www.origene.com/catalog/cdna-clones/expression-plasmids/rc22488/nav17-scn9a-nm_002977-human-tagged-orf-clone on Feb. 13, 2023. 6 pages.
Banskota et al., Engineered virus-like particles for efficient in vivo delivery of therapeutic proteins. Cell. Jan. 20, 2022;185(2):250-265.e16. doi: 10.1016/j.cell.2021.12.021. Epub Jan. 11, 2022.
Basila et al., Minimal 2′-O-methyl phosphorothioate linkage modification pattern of synthetic guide RNAs for increased stability and efficient CRISPR-Cas9 gene editing avoiding cellular toxicity. PLoS One. Nov. 27, 2017;12(11):e0188593. doi: 10.1371/journal.pone.0188593.
Bertsimas et al., Simulated annealing. Statistical Science. Feb. 1993;8(1):10-15. doi: 10.1214/ss/1177011077.
Chen et al., Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell. Dec. 19, 2013;155(7):1479-91. doi: 10.1016/j.cell.2013.12.001. Erratum in: Cell. Jan. 16, 2014;156(1-2):373.
Cheng et al., [Cloning,expression and activity identification of human innate immune protein apolipoprotein B mRNA editing enzyme catalytic subunit 3A(APOBEC3A)]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. Chinese Journal of Cellular and Molecular Immunology, Feb. 2017;33(2):179-84. Chinese.
Fang et al., The Menu of Features that Define Primary MicroRNAs and Enable De Novo Design of MicroRNA Genes. Mol Cell. Oct. 1, 2015;60(1):131-45. doi: 10.1016/j.molcel.2015.08.015. Epub Sep. 24, 2015.
Feng et al., Efficient genome editing in plants using a CRISPR/Cas system. Cell Res. Oct. 2013;23(10):1229-32. doi: 10.1038/cr.2013.114. Epub Aug. 20, 2013.
Geisberg et al., Global analysis of mRNA isoform half-lives reveals stabilizing and destabilizing elements in yeast. Cell. Feb. 13, 2014;156(4):812-24. doi: 10.1016/j.cell.2013.12.026.
Green et al., Characterization of the mechanical unfolding of RNA pseudoknots. J Mol Biol. Jan. 11, 2008;375(2):511-28. doi: 10.1016/j.jmb.2007.05.058. Epub May 26, 2007.
Hamilton et al., Targeted delivery of CRISPR-Cas9 and transgenes enables complex immune cell engineering. Cell Rep. Jun. 1, 2021;35(9):109207 and Suppl Info. doi: 10.1016/j.celrep.2021.109207.
Hänsel-Hertsch et al., DNA G-quadruplexes in the human genome: detection, functions and therapeutic potential. Nat Rev Mol Cell Biol. May 2017;18(5):279-284. doi: 10.1038/nrm.2017.3. Epub Feb. 22, 2017.
Hawley-Nelson et al., Transfection of Cultured Eukaryotic Cells Using Cationic Lipid Reagents. Curr Prot Mol Biol. Jan. 2008;9.4.1-9.4.17. doi: 10.102/0471142727.mb0904s81. 17 pages.
Houck-Loomis et al., An equilibrium-dependent retroviral mRNA switch regulates translational recoding. Nature. Nov. 27, 2011;480(7378):561-4. doi: 10.1038/nature 10657.
Houseley et al., The many pathways of RNA degradation. Cell. Feb. 20, 2009;136(4):763-76. doi: 10.1016/j.cell.2009.01.019.
Humbel et al., Maximizing lentiviral vector gene transfer in the CNS. Gene Ther. Feb. 2021;28(1-2):75-88. doi: 10.1038/s41434-020-0172-6. Epub Jul. 6, 2020. Erratum in: Gene Ther. May 2022;29(5):312.
Ibrahim et al., RNA recognition by 3′-to-5′ exonucleases: the substrate perspective. Biochim Biophys Acta. Apr. 2008;1779(4):256-65. doi: 10.1016/j.bbagrm.2007.11.004. Epub Dec. 3, 2007.
Jost et al., Titrating gene expression using libraries of systematically attenuated CRISPR guide RNAs. Nat Biotechnol. Mar. 2020;38(3):355-364. doi: 10.1038/s41587-019-0387-5. Epub Jan. 13, 2020.
Ku et al., Nucleic Acid Aptamers: An Emerging Tool for Biotechnology and Biomedical Sensing. Sensors (Basel). Jul. 6, 2015;15(7):16281-313. doi: 10.3390/s150716281.
Kuan et al., A systematic evaluation of nucleotide properties for CRISPR sgRNA design. BMC Bioinformatics. Jun. 6, 2017;18(1):297. doi: 10.1186/s12859-017-1697-6.
Kwok et al., G-Quadruplexes: Prediction, Characterization, and Biological Application. Trends Biotechnol. Oct. 2017;35(10):997-1013. doi: 10.1016/j.tibtech.2017.06.012. Epub Jul. 26, 2017.
Longsworth, Expanding the Enzymatic Activity of the Programmable Endonuclease Cas9 in Zebrafish. Thesis. Rice University. Houston, TX. Aug. 2018. 41 pages.
Lyu et al., Virus-Like Particle Mediated CRISPR/Cas9 Delivery for Efficient and Safe Genome Editing. Life (Basel). Dec. 21, 2020;10(12):366. doi: 10.3390/life10120366.
Macfadden et al., Mechanism and structural diversity of exoribonuclease-resistant RNA structures in flaviviral RNAs. Nat Commun. Jan. 9, 2018;9(1):119. doi: 10.1038/s41467-017-02604-y.
Mahoney et al., The Next Immune-Checkpoint Inhibitors: PD-1/PD-L1 Blockade in Melanoma. Clin Ther. Apr. 1, 2015;37(4):764-82. doi: 10.1016/j.clinthera.2015.02.018. Epub Mar. 29, 2015.
Mali et al., CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat Biotechnol. Sep. 2013;31(9):833-8, Supplemental Info. doi: 10.1038/nbt.2675. Epub Aug. 1, 2013.
Mangeot et al., Genome editing in primary cells and in vivo using viral-derived Nanoblades loaded with Cas9-sgRNA ribonucleoproteins. Nat Commun. Jan. 3, 2019;10(1):45. doi: 10.1038/s41467-018-07845-z.
Marcovitz et al., Frustration in protein-DNA binding influences conformational switching and target search kinetics. Proc Natl Acad Sci U S A. Nov. 1, 2011;108(44): 17957-62. doi: 10.1073/pnas.1109594108. Epub Oct. 14, 2011.
Micozzi et al., Human cytidine deaminase: a biochemical characterization of its naturally occurring variants. Int J Biol Macromol. Feb. 2014;63:64-74. doi: 10.1016/j.ijbiomac.2013.10.029. Epub Oct. 29, 2013. Erratum in: Int J Biol Macromol. Feb. 2014;63:262.
Millevoi et al., G-quadruplexes in RNA biology. Wiley Interdiscip Rev RNA. Jul.-Aug. 2012;3(4):495-507. doi: 10.1002/wrna. 1113. Epub Apr. 4, 2012.
Min et al., Deep learning in bioinformatics. Brief Bioinform. Sep. 1, 2017;18(5):851-869. doi: 10.1093/bib/bbw068.
Ousterout et al., Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy. Nat Commun. Feb. 18, 2015;6:6244. doi: 10.1038/ncomms7244.
Pandey et al., Effect of loops and G-quartets on the stability of RNA G-quadruplexes. J Phys Chem B. Jun. 13, 2013;117(23):6896-905. doi: 10.1021/jp401739m. Epub May 29, 2013. Supplementary Information, 21 pages.
Petit et al., Powerful mutators lurking in the genome. Philos Trans R Soc Lond B Biol Sci. Mar. 12, 2009;364(1517):705-15. doi: 10.1098/rstb.2008.0272.
Pijlman et al., A highly structured, nuclease-resistant, noncoding RNA produced by flaviviruses is required for pathogenicity. Cell Host Microbe. Dec. 11, 2008;4(6):579-91. doi: 10.1016/j.chom.2008.10.007.
Piotukh et al., Directed evolution of sortase A mutants with altered substrate selectivity profiles. J Am Chem Soc. Nov. 9, 2011;133(44):17536-9. doi: 10.1021/ja205630g. Epub Oct. 13, 2011.
Podracky et al., Laboratory evolution of a sortase enzyme that modifies amyloid-β protein. Nat Chem Biol. Mar. 2021;17(3):317-325. doi: 10.1038/s41589-020-00706-1. Epub Jan. 11, 2021.
Robert et al., Virus-Like Particles Derived from HIV-1 for Delivery of Nuclear Proteins: Improvement of Production and Activity by Protein Engineering. Mol Biotechnol. Jan. 2017;59(1):9-23. doi: 10.1007/s12033-016-9987-1.
Steckelberg et al., A folded viral noncoding RNA blocks host cell exoribonucleases through a conformationally dynamic RNA structure. Proc Natl Acad Sci U S A. Jun. 19, 2018;115(25):6404-6409. doi: 10.1073/pnas.1802429115. Epub Jun. 4, 2018.
Wu et al., MLV based viral-like-particles for delivery of toxic proteins and nuclear transcription factors. Biomaterials. Sep. 2014;35(29):8416-26. doi: 10.1016/j.biomaterials.2014.06.006. Epub Jul. 3, 2014.
Wu et al., Widespread Influence of 3′-End Structures on Mammalian mRNA Processing and Stability. Cell. May 18, 2017;169(5):905-917.e11. doi: 10.1016/j.cell.2017.04.036.
Yi et al., Engineering of TEV protease variants by yeast ER sequestration screening (YESS) of combinatorial libraries. Proc Natl Acad Sci U S A. Apr. 30, 2013;110(18):7229-34. doi: 10.1073/pnas.1215994110. Epub Apr. 15, 2013.
Zhu et al., Novel Thrombotic Function of a Human SNP in STXBP5 Revealed by CRISPR/Cas9 Gene Editing in Mice. Arterioscler Thromb Vasc Biol. Feb. 2017;37(2):264-270. doi: 10.1161/ATVBAHA.116.308614. Epub Dec. 29, 2016.
[No Author Listed], MutL homolog 1. UniProtKB Acc. No. F1MPG0. May 3, 2011. Accessible at https://rest.uniprot.org/unisave/F1MPG0?format=txt&versions=1. 1 page.
[No Author Listed], RecName: Full=DNA mismatch repair protein Mlh1; AltName: Full=MutL protein homolog 1. UniProtKB/Swiss-Prot Acc. No. P97679.1. May 3, 2023. Accessible at https://www.ncbi.nlm.nih.gov/protein/p97679. 3 pages.
Acharya et al., hMSH2 forms specific mispair-binding complexes with hMSH3 and hMSH6. Proc Natl Acad Sci U S A. Nov. 26, 1996;93(24):13629-34. doi: 10.1073/pnas.93.24.13629.
Barrera-Paez et al., Mitochondrial genome engineering coming-of-age. Trends Genet. Aug. 2022;38(8):869-880. doi: 10.1016/j.tig.2022.04.011. Epub May 19, 2022. Erratum in: Trends Genet. Aug. 26, 2022.
Bosch et al., Precise genome engineering in Drosophila using prime editing. Proc Natl Acad Sci U S A. Jan. 5, 2021;118(1):e2021996118. doi: 10.1073/pnas.2021996118.
Edraki et al., A Compact, High-Accuracy Cas9 with a Dinucleotide PAM for In Vivo Genome Editing. Mol Cell. Feb. 21, 2019;73(4):714-726.e4 and Supplemental Info. doi: 10.1016/j.molcel.2018.12.003. Epub Dec. 20, 2018.
Fang et al., Human strand-specific mismatch repair occurs by a bidirectional mechanism similar to that of the bacterial reaction. J Biol Chem. Jun. 5, 1993;268(16):11838-44.
Fishel et al., The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell. Dec. 3, 1993;75(5):1027-38. doi: 10.1016/0092-8674(93)90546-3. Erratum in: Cell. Apr. 8, 1994;77(1):1 p following 166.
Fu et al., Targeted genome editing in human cells using CRISPR/Cas nucleases and truncated guide RNAs. Methods Enzymol. 2014;546:21-45. doi: 10.1016/B978-0-12-801185-0.00002-7.
Geng et al., In vitro studies of DNA mismatch repair proteins. Anal Biochem. Jun. 15, 2011;413(2):179-84. doi: 10.1016/j.ab.2011.02.017. Epub Feb. 15, 2011.
Genschel et al., Human exonuclease I is required for 5′ and 3′ mismatch repair. J Biol Chem. Apr. 12, 2002;277(15):13302-11. doi: 10.1074/jbc.M111854200. Epub Jan. 24, 2002.
Genschel et al., Isolation of MutSbeta from human cells and comparison of the mismatch repair specificities of MutSbeta and MutSalpha. J Biol Chem. Jul. 31, 1998;273(31):19895-901. doi: 10.1074/jbc.273.31.19895. Erratum in: J Biol Chem Oct. 9, 1998;273(41):27034.
Gueneau et al., Structure of the MutLα C-terminal domain reveals how Mlh1 contributes to Pms1 endonuclease site. Nat Struct Mol Biol. Apr. 2013;20(4):461-8. doi: 10.1038/nsmb.2511. Epub Feb. 24, 2013.
Guerrette et al., The interaction of the human MutL homologues in hereditary nonpolyposis colon cancer. J Biol Chem. Mar. 5, 1999;274(10):6336-41. doi: 10.1074/jbc.274.10.6336.
Gupta et al., Mechanism of mismatch recognition revealed by human MutSβ bound to unpaired DNA loops. Nat Struct Mol Biol. Dec. 18, 2011;19(1):72-8. doi: 10.1038/nsmb.2175.
Hussman et al., Mapping the genetic landscape of DNA double-strand break repair. Cell. Oct. 28, 2021;184(22):5653-5669.e25. doi: 10.1016/j.cell.2021.10.002. Epub Oct. 20, 2021.
Iaccarino et al., hMSH2 and hMSH6 play distinct roles in mismatch binding and contribute differently to the ATPase activity of hMutSalpha. EMBO J. May 1, 1998;17(9):2677-86. doi: 10.1093/emboj/17.9.2677.
Iyer et al., DNA mismatch repair: functions and mechanisms. Chem Rev. Feb. 2006;106(2):302-23. doi: 10.1021/cr0404794.
Kadyrov et al., Endonucleolytic function of MutLalpha in human mismatch repair. Cell. Jul. 28, 2006;126(2):297-308. doi: 10.1016/j.cell.2006.05.039.
Kunkel et al., DNA mismatch repair. Annu Rev Biochem. 2005;74:681-710. doi: 10.1146/annurev.biochem.74.082803.133243.
Lahue et al., DNA mismatch correction in a defined system. Science. Jul. 14, 1989;245(4914):160-4. doi: 10.1126/science.2665076.
Leach et al., Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer. Cell. Dec. 17, 1993;75(6):1215-25. doi: 10.1016/0092-8674(93)90330-s.
Lim et al., Nuclear and mitochondrial DNA editing in human cells with zinc finger deaminases. Nat Commun. Jan. 18, 2022;13(1):366. doi: 10.1038/s41467-022-27962-0.
Lujan et al., Heterogeneous polymerase fidelity and mismatch repair bias genome variation and composition. Genome Res. Nov. 2014;24(11):1751-64. doi: 10.1101/gr.178335.114. Epub Sep. 12, 2014.
Parsons et al., Hypermutability and mismatch repair deficiency in RER+ tumor cells. Cell. Dec. 17, 1993;75(6):1227-36. doi: 10.1016/0092-8674(93)90331-j.
Plotz et al., N-terminus of hMLH1 confers interaction of hMutLalpha and hMutLbeta with hMutSalpha. Nucleic Acids Res. Jun. 15, 2003;31(12):3217-26. doi: 10.1093/nar/gkg420.
Rallapalli et al., The Design and Application of DNA-Editing Enzymes as Base Editors. Annu Rev Biochem. Jun. 20, 2023;92:43-79. doi: 10.1146/annurev-biochem-052521-013938. Epub Apr. 5, 2023.
Räschle et al., Mutations within the hMLH1 and hPMS2 subunits of the human MutLalpha mismatch repair factor affect its ATPase activity, but not its ability to interact with hMutSalpha. J Biol Chem. Jun. 14, 2002;277(24):21810-20. doi: 10.1074/jbc.M108787200. Epub Apr. 10, 2002.
Shcherbakova et al., Mutator phenotypes conferred by MLH1 overexpression and by heterozygosity for mlh1 mutations. Mol Cell Biol. Apr. 1999;19(4):3177-83. doi: 10.1128/MCB.19.4.3177.
Strand et al., Destabilization of tracts of simple repetitive DNA in yeast by mutations affecting DNA mismatch repair. Nature. Sep. 16, 1993;365(6443):274-6. doi: 10.1038/365274a0. Erratum in: Nature Apr. 7, 1994;368(6471);569.
Su et al., Mispair specificity of methyl-directed DNA mismatch correction in vitro. J Biol Chem. May 15, 1988;263(14):6829-35. Erratum in: J Biol Chem Aug. 5, 1988;263(22):11015.
Sugawara et al., Heteroduplex rejection during single-strand annealing requires Sgs1 helicase and mismatch repair proteins Msh2 and Msh6 but not Pms1. Proc Natl Acad Sci U S A. Jun. 22, 2004;101(25):9315-20. doi: 10.1073/pnas.0305749101. Epub Jun. 15, 2004.
Supek et al., Differential DNA mismatch repair underlies mutation rate variation across the human genome. Nature. May 7, 2015;521(7550):81-4. doi: 10.1038/nature14173. Epub Feb. 23, 2015.
Svitashev et al., Targeted Mutagenesis, Precise Gene Editing, and Site-Specific Gene Insertion in Maize Using Cas9 and Guide RNA. Plant Physiol. Oct. 2015; 169(2):931-45. doi: 10.1104/pp.15.00793. Epub Aug. 12, 2015.
Thomas et al., Heteroduplex repair in extracts of human HeLa cells. J Biol Chem. Feb. 25, 1991;266(6):3744-51.
Tomer et al., Contribution of human mlh1 and pms2 ATPase activities to DNA mismatch repair. J Biol Chem. Jun. 14, 2002;277(24):21801-9. doi: 10.1074/jbc.M111342200. Epub Mar. 15, 2002.
Tran et al., Hypermutability of homonucleotide runs in mismatch repair and DNA polymerase proofreading yeast mutants. Mol Cell Biol. May 1997;17(5):2859-65. doi: 10.1128/MCB.17.5.2859.
Umar et al., DNA loop repair by human cell extracts. Science. Nov. 4, 1994;266(5186):814-6. doi: 10.1126/science.7973637.
Warren et al., Structure of the human MutSalpha DNA lesion recognition complex. Mol Cell. May 25, 2007;26(4):579-92. doi: 10.1016/j.molcel.2007.04.018.
Willis et al., Compact zinc finger base editors that edit mitochondrial or nuclear DNA in vitro and in vivo. Nat Commun. Nov. 23, 2022;13(1):7204. doi: 10.1038/s41467-022-34784-7.
Xi et al., C-terminal Loop Mutations Determine Folding and Secretion Properties of PCSK9. Biochem Mol Biol J. 2016;2(3):17. doi: 10.21767/2471-8084.100026. 12 pages.
Zhang et al., Reconstitution of 5′-directed human mismatch repair in a purified system. Cell. Sep. 9, 2005;122(5):693-705. doi: 10.1016/j.cell.2005.06.027.
Related Publications (1)
Number Date Country
20230220374 A1 Jul 2023 US
Provisional Applications (2)
Number Date Country
63116785 Nov 2020 US
63022397 May 2020 US
Continuations (1)
Number Date Country
Parent PCT/US2021/031439 May 2021 US
Child 18053269 US