Minimal Virus-Like Particles and Methods of Use Thereof for Delivery of Biomolecules

Abstract
Described herein are minimal virus-like particles (mVLPs), comprising a membrane comprising a phospholipid bilayer with one or more ectodomain-truncated VSV envelope glycoproteins on the external side; and a biomolecule cargo disposed in the core of the mVLP on the inside of the membrane. Preferably, the mVLPs do not comprise a protein from viral gag, pro, pol, or other viral proteins that reside inside of enveloped particles. Also described are methods of use of the mVLPs for delivery of the biomolecule cargo to cells.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on Feb. 22, 2024, is named 29539-0682001_SL.xml and is 195,405 bytes in size.


TECHNICAL FIELD

Described herein are virus-like particles (VLPs) and minimal virus-like particles (mVLPs), comprising a membrane comprising a phospholipid bilayer with one or more ectodomain-truncated VSV envelope glycoproteins on the external side; and a biomolecule cargo disposed in the core of the VLP or mVLP on the inside of the membrane. Preferably, the mVLPs do not comprise any other exogenous virally derived proteins, e.g., proteins from viral gag, pro, or pol, or other viral proteins that reside inside of enveloped particles (unless the cargo comprises the viral protein(s)). Also described are methods of use of the VLPs or mVLPs for delivery of the biomolecule cargo to cells.


BACKGROUND

Delivery of cargo such as proteins, nucleic acids, and/or chemicals into the cytosol of living cells has been a significant hurdle in the development of biological therapeutics.


SUMMARY

Described herein are virus-like particles (VLPs) and minimal virus-like particles (mVLPs) that are capable of packaging and delivering a wide variety of payloads, e.g., biomolecules including nucleic acids (DNA, RNA) or proteins, chemical compounds including small molecules, and/or other molecules, and any combination thereof, into eukaryotic cells. The non-viral mVLP systems described herein have the potential to be simpler, more efficient and safer than conventional, artificially-derived lipid/gold nanoparticles and viral particle-based delivery systems, at least because mVLPs have no virus-derived components except for an ectodomain-truncated envelope glycoprotein, mVLPs can utilize but do not require chemical-based dimerizers, and mVLPs have the ability to package and deliver cargo including, but not limited to, biomolecules including nucleic acids and proteins, e.g., specialty single and/or double-stranded DNA molecules (e.g., plasmid, mini circle, closed-ended linear DNA, AAV DNA, episomes, bacteriophage DNA, homology directed repair templates, etc.), single and/or double-stranded RNA molecules (e.g., single guide RNA, prime editing guide RNA, messenger RNA, transfer RNA, long non-coding RNA, circular RNA, RNA replicon, circular or linear splicing RNA, micro RNA, small interfering RNA, short hairpin RNA, piwi-interacting RNA, toehold switch RNA, RNAs that can be bound by RNA binding proteins, bacteriophage RNA, internal ribosomal entry site containing RNA, etc.), proteins, chemical compounds and/or molecules, and combinations of the above listed cargos (e.g., AAV particles and/or ribonucleoprotein (RNP) complexes comprising RNA and protein, e.g., guide RNA/CRISPR Cas protein complexes). The virus-like particles (VLPs) described herein can comprise ectodomain-truncated envelope glycoproteins. The mVLPs described herein are different from conventional retroviral particles, virus-like particles (VLPs), exosomes and other previously described extracellular vesicles that can be loaded with cargo because of the membrane configuration, ectodomain-truncated envelope glycoprotein, vast diversity of possible cargos that are enabled by novel, innovative loading strategies, the lack of a limiting DNA/RNA length constraint, the lack of proteins derived from any viral gag, pro, or pol, and the mechanism of cellular entry.


Provided herein are truncated glycoproteins/envelope proteins (tENV) comprising: an N-terminal portion comprising a signal sequence, optionally comprising the MKCLLYLAFLFIGVNCK (SEQ ID NO:1) fused to a central portion comprising all or part of a GS domain from at least one vesiculovirus G protein, optionally a VSV-G protein or homolog, ortholog, or paralog thereof, optionally comprising the sequence FEHPHIQDAASQLPDDESLFFGDTGLSKNPIELVEG WFSSWK (SEQ ID NO:2), optionally with a deletion of at least one amino acid (e.g., a truncation from the N terminal end of the central portion), which is fused to a C-terminal portion comprising a transmembrane domain and an intracellular domain, optionally comprising SSIASFFFIIGLIIGLFLVLRVGIHLCIKLKHTKKRQIYTDIE MNRLGK (SEQ ID NO:3). In some embodiments, the central portion comprises a deletion of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 38, 39, 40, 41, or all 42 amino acids of SEQ ID NO:2 amino acids, e.g., at least about 1, 2, 3, 4, 5, 6, 7, 8 or 10 amino acids, up to about 15, 20, 35, 30, 35, 38, 39, 40, 41, or all 42 amino acids, with any range therebetween. In some embodiments, the central portion comprises or consists of FFGDTGLSKNPIELVEGWFSSWK (SEQ ID NO:4) or FEHPHIQDAASQLPDDESLFFGDTGLSKNPIELVEGWFSSWK (SEQ ID NO:2). In some embodiments, the tENV comprises a sequence that is at least 95% identical to a sequence set forth herein, e.g., in Table 1.


Also provided herein are nucleic acids encoding the tENVs, and vectors comprising the nucleic acids, optionally operably linked to a promoter for expression of the tENV, as well as host (e.g., producer) cells comprising the nucleic acids, and optionally expressing the tENV.


Further, provided herein are virus-like particles (VLPs) comprising a tENV described herein. Also provided are minimal virus-like particles (mVLPs), comprising a membrane comprising a phospholipid bilayer and a tENV as described herein; and a cargo disposed in the core of the mVLP, wherein the cargo is optionally fused to a phospholipid bilayer recruitment domain; and, wherein the mVLP does not comprise any exogenous virally derived proteins, e.g., proteins from viral gag, pro, or pol, or other viral proteins that reside inside of enveloped particles (unless the cargo comprises the viral protein(s)). In some embodiments, the VLPs may include endogenous components, e.g., from endogenous retroviral sequences that have integrated into the genome of the cells, e.g., HERVs when particles are produced in human cells. In some embodiments, the VLPs do or do not comprise any human endogenous retroviral (HERV) proteins other than the env, e.g., do not comprise gag, pol, or pro. Exogenous virally-derived gag, pol, or pro refers to any gag, pro, pol, gag-pol, gag-pro-pol, and/or pol protein, or any other protein expressed from gag, pro, or pol, from any virus introduced into the cell.


In some embodiments, the cargo is a therapeutic or diagnostic protein or nucleic acid encoding a therapeutic or diagnostic protein, or a chemical, optionally a small molecule therapeutic or diagnostic. In some embodiments, the cargo is a gene editing or epigenetic modulating reagent. In some embodiments, the gene editing or epigenetic modulating reagent comprises a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof, a nucleic acid encoding a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof, or a ribonucleoprotein complex (RNP) comprising a CRISPR-Cas protein, variant, or fusion thereof and optionally a guide RNA. In some embodiments, the cargo is selected from the proteins listed in Tables 2, 3, 4 & 5, or that is at least 95% identical to a sequence set forth herein, e.g., in Tables 2, 3, 4, and 5. In some embodiments, the cargo comprises a CRISPR-Cas protein, and the mVLP further comprises one or more guide RNAs that bind to and direct the CRISPR-Cas protein to a target nucleic acid sequence. In some embodiments, the cargo comprises a fusion to a phospholipid bilayer recruitment domain, preferably as shown in Table 6, or that is at least 95% identical to a sequence set forth herein in Table 6.


Also provided are methods of delivering a cargo to a target cell, optionally a cell in vivo or in vitro, the method comprising contacting the cell with a VLP or mVLP as described herein comprising the cargo.


Additionally provided herein are method of producing a VLP or an mVLP, optionally comprising a cargo. The methods comprise providing a cell expressing a tENV as described herein and optionally a cargo, optionally wherein the cell does not express exogenous virally derived proteins, e.g., proteins from viral gag, pro, or pol, or other viral proteins that reside inside of enveloped particles (unless the cargo comprises the viral protein(s)); and maintaining the cell under conditions such that the cells produce the VLPs or mVLPs.


In some embodiments, the methods further comprise harvesting and optionally purifying and/or concentrating the produced VLPs or mVLPs. In some embodiments, the cargo is a therapeutic or diagnostic protein or nucleic acid encoding a therapeutic or diagnostic protein, or a small molecule, optionally a therapeutic or diagnostic small molecule. In some embodiments, the cargo is a gene editing or epigenetic modulating reagent. In some embodiments, the gene editing or epigenetic modulating reagent comprises a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof, a nucleic acid encoding a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof, or a ribonucleoprotein complex (RNP) comprising a CRISPR-Cas protein, variant, or fusion thereof and optionally a guide RNA.


In some embodiments, the cargo reagent is selected from the proteins listed in Tables 2, 3, 4 & 5, or that is at least 95% identical to a sequence set forth herein, e.g., in Tables 2, 3, 4, and 5. In some embodiments, the cargo reagent comprises a CRISPR-Cas protein, variant, or fusion thereof and the mVLP further comprises one or more guide RNAs that bind to and direct the CRISPR-based genome editing or modulating protein to a target sequence. In some embodiments, the cargo comprises a fusion to a phospholipid bilayer recruitment domain, preferably as shown in Table 6, or that is at least 95% identical to a sequence set forth herein in Table 6.


Also provided herein are cells expressing a tENV as described herein, and a cargo, optionally wherein the cell does not express an exogenous gag, pro, or pol protein. In some embodiments, the cargo is a therapeutic or diagnostic protein or nucleic acid encoding a therapeutic or diagnostic protein, or a small molecule, optionally a therapeutic or diagnostic small molecule. In some embodiments, the cargo is a gene editing or epigenetic modulating reagent.


In some embodiments, the gene editing or epigenetic modulating reagent comprises a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof, a nucleic acid encoding a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof, or a ribonucleoprotein complex (RNP) comprising a CRISPR-Cas protein, variant, or fusion thereof and optionally a guide RNA. In some embodiments, the cargo reagent is selected from the proteins listed in Tables 2, 3, 4, & 5, or that is at least 95% identical to a sequence set forth herein, e.g., in Tables 2, 3, 4, and 5. In some embodiments, the gene editing or epigenetic modulating reagent comprises a CRISPR-Cas protein, and the mVLP further comprises one or more guide RNAs that bind to and direct the CRISPR-Cas protein to a target sequence. In some embodiments, the cargo comprises a fusion to a phospholipid bilayer recruitment domain, preferably as shown in Table 6, or that is at least 95% identical to a sequence set forth herein in Table 6. In some embodiments, the cells are primary or stable human cell lines. In some embodiments, the cells are Human Embryonic Kidney (HEK) 293 cells or HEK293 T cells.


Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present invention; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.


Other features and advantages of the invention will be apparent from the following detailed description and figures, and from the claims.





DESCRIPTION OF DRAWINGS


FIGS. 1A-B. Diagrams of (A) exemplary mVLP DNA expression constructs that can be transfected into producer cells including cargo components Cas9 and a single guide RNA (sgRNA) and the 421 VSVG truncated ENV protein (wherein VSVG amino acids 421-511 preceded by a signal sequence are present, as described in Table 1) and (B) architecture of an exemplary particle to deliver a Cas9 gene editing protein or RNP complex.



FIG. 2. mVLP-mediated delivery delivery of SpCas9 RNP. K562 cells transduced with mVLPs containing human AKT Pleckstrin homology domain fused to SpCas9 and a guide RNA targeted to VEGF site 3.1. mVLPs were pseudotyped with one of three different ectodomain-truncated versions of VSVG (448-511 (448), 440-511 (440), and 421-511 (421), wherein these aforementioned VSVG residue ranges are preceded by a signal sequence, respectively). Gene modification was measured by targeted amplicon sequencing of the intended VEGF site 3.1 on-target site using NGS.



FIG. 3. Alignment of exemplary tENV sequences. Marked domains:

    • MKCLLYLAFLFIGVNCK—VSVG signal sequence (SEQ ID NO:1)
    • FFGDTGLSKNPIELVEGWFSSWKS—VSVG GS domain (SEQ ID NO:5)
    • FFFIIGLIIGLFLVLRVGIHLCI—VSVG TM domain (SEQ ID NO:6)
    • KLKHTKKRQIYTDIEMNRLGK—VSVG Intracellular domain (SEQ ID NO:7).


      Shown in the figure are SEQ ID NOs:8-23.



FIG. 4. Graphical alignment of VSCG and Cocal glycoproteins with GS-domain, transmembrane domain (Tm), and intracellular domain (ICD) annotated. Shown in the figure are SEQ ID NOs:24-26.



FIG. 5. Exemplary editing efficiencies of eVLPs that have various PH domain-Cas9 fusion cargos packaged together with an sgRNA targeted to the VEGFs3.1 site. HEK293T cells were treated with these eVLPs that had been pseudotyped with VSVG and editing efficiency was determined by amplicon sequencing.

    • PKD: protein kinase D1 (PRKD1)
    • DAPP: dual-adaptor for phosphotyrosine and 3-phosphoinositides-1 (DAPP-1)
    • FAPP: four-phosphate-adaptor protein (FAPP)
    • OSBP: oxysterol-binding protein (OSBP)
    • SWAP70: switch-associated protein 70 (SWAP70)
    • GRP: cytohesin 3 (CYTH3, formerly GRP1)
    • BTK: Bruton's tyrosine kinase (Btk)
    • PHLPP: Pleckstrin Homology Domain Leucine-rich Repeat Protein Phosphatase (PHLPP)
    • AKT: AKT serine/threonine kinase 1 (AKT1)
    • PLC: phospholipase C delta 1 (PLC1)



FIG. 6. Exemplary gene modification efficiencies induced by eVLPs that contained various mutant PH-Cas9/sgRNA (RNF2-targeted) RNP cargos. Primary T cells were treated with eVLPs pseudotyped with either VSVG or VSVG+BaeVTRless and gene modification efficiencies (y-axis) were determined by targeted amplicon sequencing of the RNF2 on-target site in those cells.





DETAILED DESCRIPTION

Therapeutic proteins and nucleic acids hold great promise, but for many of these large biomolecules, delivery into cells is a hurdle to clinical development. Genome editing reagents such as zinc finger nucleases (ZFNs) or RNA-guided, enzymatically active/inactive DNA binding proteins such as Cas9 have undergone rapid advancements in terms of specificity and the types of edits that can be executed, but the hurdle of safe in vivo delivery still remains an important challenge for gene editing and epigenetic editing therapies.


Virus-like particles (VLPs) have been utilized to deliver mRNA and protein cargo into the cytosol of cells.2,3,25-30 VLPs have emerged as an alternative delivery modality to retroviral or lentiviral particles. VLPs can be designed to lack the ability to integrate retroviral DNA, and to package and deliver combinations of protein/RNP/DNA. However, most VLPs, including recently conceived VLPs that deliver genome editing reagents known to date, utilize HIV or other virally-derived gag or gag-pol protein fusions and viral proteases to generate retroviral-like particles.25-27,29,30 Some VLPs containing RNA-guided nucleases (RGNs) also must package and express guide RNAs from a lentiviral DNA transcript,27 and some VLPs require a viral protease in order to form functional particles and release genome editing cargo.25-27,29 Because this viral protease recognizes and cleaves at multiple amino acid motifs, it can cause damage to the protein cargo or potentially to other endogenous proteins in target recipient cells, which could be hazardous or create challenges for therapeutic applications. Most published VLP modalities that deliver genome editing proteins or RNPs to date exhibit low in vitro and in vivo gene modification efficiencies due to low packaging and transduction efficiency.25-27 The complex viral genomes utilized for these VLP components possess multiple reading frames and employ RNA splicing that could result in spurious fusion protein products being delivered.25-27,29,30 The presence of reverse transcriptase, integrase, capsid and a virally-derived envelope protein in these VLPs is not ideal for many therapeutic applications because of immunogenicity and off target concerns. In addition, most retroviral particles, such as lentiviral particles, are pseudotyped with VSVG and nearly all described VLPs that deliver genome editing reagents hitherto possess and rely upon VSVG.2,3,25-30


Lentivirus and standard VLPs commonly require GAG and ENV proteins to drive particle formation via budding off of the plasma membrane of producer cells into the cell culture medium. In addition, the majority of retroviral ENV proteins require post-translational modifications in the form of proteolytic cleavage of the intracellular domain (ICD) of the ENV protein in order to activate the fusogenicity of the ENV protein; this is essential for viral infectivity. Without wishing to be bound by theory, it is believed that the ENV protein alone is responsible for mVLP particle generation and the ability of mVLPs to efficiently deliver cargo into cells. As described herein, the ENV protein on the surface can be truncated and thus lack some or all of the ectodomain (tENV), even including deletion of all or part of the GS domain previously thought to be important (see, e.g., Jeetendra et al., J. Virol. 76(23): 12300-12311 (2002); US 2010/0167377). Surprisingly, as demonstrated herein, even when up to about 90% of the ENV ectodomain was deleted, VLPs comprising the truncated proteins still function to deliver cargo.


Provided herein are virus-like particles (VLPs) and minimal virus-like particles (mVLPs), comprising a membrane comprising a phospholipid bilayer with one or more ectodomain-truncated VSV envelope glycoproteins on the external side; and a biomolecule cargo disposed in the core of the VLP or mVLP on the inside of the membrane. The biomolecule cargo can be fused to a phospholipid bilayer recruitment domain as described herein. Preferably, the mVLPs do not comprise any other exogenous virally derived proteins, e.g., proteins from viral gag, pro, or pol, or other viral proteins that reside inside of enveloped particles, such as int (unless the cargo intentionally comprises the viral protein(s)).


Truncated Envelope Proteins (tENVs)


The truncated envelope proteins (tENVs) described herein include the exemplary sequences in Table 1 that are derived from vesicular stomatitis virus (VSV) and homologs thereof. The wild type sequence for the VSV glycoprotein is:









(SEQ ID NO: 27)








1

MKCLLYLAFL FIGVNCKFTI VFPHNQKGNW KNVPSNYHYC







PSSSDLNWHN DLIGTAIQVK





61
MPKSHKAIQA DGWMCHASKW VTTCDFRWYG PKYITQSIRS






FTPSVEQCKE SIEQTKQGTW





121
LNPGFPPQSC GYATVTDAEA VIVQVTPHHV LVDEYTGEWV






DSQFINGKCS NYICPTVHNS





181
TTWHSDYKVK GLCDSNLISM DITFFSEDGE LSSLGKEGTG






FRSNYFAYET GGKACKMQYC





241
KHWGVRLPSG VWFEMADKDL FAAARFPECP EGSSISAPSQ






TSVDVSLIQD VERILDYSLC





301
QETWSKIRAG LPISPVDLSY LAPKNPGTGP AFTIINGTLK






YFETRYIRVD IAAPILSRMV





361
GMISGTTTER ELWDDWAPYE DVEIGPNGVL RTSSGYKFPL






YMIGHGMLDS DLHLSSKAQV





421

FEHPHIQDAA SQLPDDESLF FGDTGLSKNP IELVEGWESS








WKSSIASFFF IIGLIIGLFL






481

VLRVGIHLCI KLKHTKKRQI YTDIEMNRLG K











VSVG signal sequence


(SEQ ID NO: 1)



MKCLLYLAFLFIGVNCK






VSVG GS domain


(SEQ ID NO: 5)



FFGDTGLSKNPIELVEGWESSWKS






VSVG TM domain


(SEQ ID NO: 6)



FFFIIGLIIGLFLVLRVGIHLCI






VSVG Intracellular domain


(SEQ ID NO: 7)



KLKHTKKRQIYTDIEMNRLGK







Preferably, the tENVs comprise an N-terminal signal sequence. Exemplary signal sequences include the one from the VSV-G protein, e.g., MKCLLYLAFLFIGVNCK (SEQ ID NO:1) and/or any other secretion signal sequence that is derived from VSVG (e.g., MKCLLYLAFLFIGVNC, SEQ ID NO:28) or a homolog thereof, or from a transmembrane protein and/or a synthetic/engineered signal sequence. A number of secretory signal peptide sequences are known in the art, including human signal sequences, examples of which are shown in Table A (Table adapted from novoprolabs.com/support/articles/commonly-used-leader-peptide-sequences-forefficient-secretion-of-a-recombinant-protein-expressed-in-mammalian-cells-201804211337.html).









TABLE A







Exemplary Human Secretory


Signal Peptide Sequences











SEQ


Human Signal

ID


sequence
Sequence
NO:





Oncostatin M
MGVLLTQRTLLSLVLALLFPSMASM
29





IgG2 H
MGWSCIILFLVATATGVHS
30





Secrecon*
MWWRLWWLLLLLLLLWPMVWA
31





IgKVIII
MDMRVPAQLLGLLLLWLRGARC
32





CD33
MPLLLLLPLLWAGALA
33





tPA
MDAMKRGLCCVLLLCGAVFVSPS
34





Chymotrypsinogen
MAFLWLLSCWALLGTTFG
35





trypsinogen-2
MNLLLILTFVAAAVA
36





Interleukin 2
MYRMQLLSCIALSLALVTNS
37


(IL-2)







Albumin (HSA)
MKWVTFISLLFSSAYS
38





insulin
MALWMRLLPLLALLALWGPDPAAA
39





alpha
MPSSVSWGILLLAGLCCLVPVSLA
40


1-antitrypsin





*Barash et al., Biochem Biophys Res Commun. 2002 Jun. 21;294(4):835-42.






In some embodiments, another signal sequence that promotes secretion is used, e.g., as described in Table 5 of U.S. Ser. No. 10/993,967; von Heijne, J Mol Biol. 1985 Jul. 5; 184(1):99-105; Kober et al., Biotechnol. Bioeng. 2013; 110: 1164-1173; Tsuchiya et al., Nucleic Acids Research Supplement No. 3 261-262 (2003).


The signal sequence is fused to a central portion comprising all or part of a GS domain from a VSV-G protein or homolog thereof. In some embodiments, the central portion comprises the sequence FEHPHIQDAASQLPDDESLFFGDTGLSKNPIELVEGWFSSWK (SEQ ID NO:2), optionally with a deletion of at least one amino acid (e.g., a truncation from the N terminal end of the central portion), and a C-terminal comprising an intracellular domain from VSVG or a homolog thereof, e.g., comprising SSIASFFFIIGLIIGLFLVLRVGIHLCIKLKHTKKRQIYTDIEMNRLGK (SEQ ID NO:3). In some embodiments, the central portion comprises a deletion of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 25, 30, 35, 38, 39, 40, 41, or all 42 amino acids of SEQ ID NO:2, and all ranges therebetween (e.g., with the recited numbers as endpoints), e.g., at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 25, 30, 35, 38, 39, 40, 41, or all 42 amino acids of SEQ ID NO:2, with any range therebetween; in some embodiments, the deletion is at the N terminus. In some embodiments, the central portion comprises









(SEQ ID NO: 4)









FFGDTGLSKNPIELVEGWFSSWK



or










(SEQ ID NO: 2)









FEHPHIQDAASQLPDDESLFFGDTGLSKNPIELVEGWFSSWK.







In some embodiments, the sequence is at least 95% identical to a sequence set forth herein.


As an alternative to the VSV G protein, G protein homologs from other viruses can also be used, e.g., Cocal glycoprotein; vesicular stomatitis Alagoas glycoprotein (VSAG); Vesicular stomatitis New Jersey glycoprotein (strain Ogden subtype Concan) (VSNJG); Vesicular stomatitis Indiana virus (strain Orsay) (VSOG); Piry glycoprotein; Maraba glycoprotein; Chandipura glycoprotein; Isfahan glycoprotein; vesicular stomatitis Glasgow glycoprotein (VSGG); Carajas virus glycoprotein (CVG); Radi virus glycoprotein; Jurona glycoprotein; Malpais Spring glycoprotein; Perinet Spring glycoprotein; or Morreton glycoprotein; see, e.g., FIG. 3. The sequences can include one or more deletions in the central GS domain as noted above. In some embodiments, the sequence is at least 95% identical to a sequence set forth herein.


Exemplary tENVs are provided in Table 1.









TABLE 1







Exemplary tENVs











SEQ ID


Source
Sequence
NO:





Vesicular stomatitis
MKCLLYLAFLFIGVNCKFEHPHIQDAASQLPDDE
41.


virus Glycoprotein
SLFFGDTGLSKNPIELVEGWFSSWKSSIASFFFI



(VSVG 421
IGLIIGLFLVLRVGIHLCIKLKHTKKRQIYTDIE



Truncation)
MNRLGK






Vesicular stomatitis
MKCLLYLAFLFIGVNCKFFGDTGLSKNPIELVEG
42.


virus Glycoprotein
WFSSWKSSIASFFFIIGLIIGLFLVLRVGIHLCI



(VSVG 440
KLKHTKKRQIYTDIEMNRLGK



Truncation)







Vesicular stomatitis
MKCLLYLAFLFIGVNCKKNPIELVEGWFSSWKSS
43.


virus Glycoprotein
IASFFFIIGLIIGLFLVLRVGIHLCIKLKHTKKR



(VSVG 448
QIYTDIEMNRLGK



Truncation)







Vesicular stomatitis
MKCLLYLAFLFIGVNCKSWKSSIASFFFIIGLII
44.


virus Glycoprotein
GLFLVLRVGIHLCIKLKHTKKRQIYTDIEMNRLG



(VSVG 460
K



Truncation)







Cocal glycoprotein
MKCLLYLAFLFIGVNCKFEHPHLAEAPKQLPEEE
45.


422 truncation
TLFFGDTGISKNPVELIEGWFSSWKSTVVTFFFA




IGVFILLYVVARIVIAVRYRYQGSNNKRIYNDIE




MSRFRK






Cocal glycoprotein
MKCLLYLAFLFIGVNCKFFGDTGISKNPVELIEG
46.


441 truncation
WFSSWKSTVVTFFFAIGVFILLYVVARIVIAVRY




RYQGSNNKRIYNDIEMSRFRK






Cocal glycoprotein
MKCLLYLAFLFIGVNCKKNPVELIEGWFSSWKST
47.


449 truncation
VVTFFFAIGVFILLYVVARIVIAVRYRYQGSNNK




RIYNDIEMSRFRK






vesicular stomatitis
MKCLLYLAFLFIGVNCKFHHPQIAEAVQKLPDDE
48.


Alagoas
TLFFGDTGISKNPVEVIEGWFSNWRSSVMAIVFA



glycoprotein
ILLLVITVLMVRLCVAFRHFCCQKRHKIYNDLEM



(VSAG) 422
NQLRR



truncation







vesicular stomatitis
MKCLLYLAFLFIGVNCKFFGDTGISKNPVEVIEG
49.


Alagoas
WFSNWRSSVMAIVFAILLLVITVLMVRLCVAFRH



glycoprotein
FCCQKRHKIYNDLEMNQLRR



(VSAG) 441




truncation







vesicular stomatitis
MKCLLYLAFLFIGVNCKKNPVEVIEGWFSNWRSS
50.


Alagoas
VMAIVFAILLLVITVLMVRLCVAFRHFCCQKRHK



glycoprotein
IYNDLEMNQLRR



(VSAG) 449




truncation







Vesicular stomatitis
MKCLLYLAFLFIGVNCKWEHPHIEAAQTFLKKDD
51.


New Jersey
TGEVLYYGDTGVSKNPVELVEGWFSGWRSSLMGV



glycoprotein (strain
LAVIIGFVILMFLIKLIGVLSSLFRPKRRPIYKS



Ogden subtype
DVEMAHFR



Concan) (VSNJG)




425 truncation







Vesicular stomatitis
MKCLLYLAFLFIGVNCKYYGDTGVSKNPVELVEG
52.


New Jersey
WFSGWRSSLMGVLAVIIGFVILMFLIKLIGVLSS



glycoprotein (strain
LFRPKRRPIYKSDVEMAHFR



Ogden subtype




Concan) (VSNJG)




447 truncation







Vesicular stomatitis
MKCLLYLAFLFIGVNCKKNPVELVEGWFSGWRSS
53.


New Jersey
LMGVLAVIIGFVILMFLIKLIGVLSSLFRPKRRP



glycoprotein (strain
IYKSDVEMAHFR



Ogden subtype




Concan) (VSNJG)




455 truncation







Vesicular stomatitis
MKCLLYLAFLFIGVNCKFEHPHIQDAASQLPDDE
54.


Indiana virus (strain
TLFFGDTGLSKNPIEFVEGWFSSWKSSIASFFFI



Orsay) (VSOG) 421
IGLIIGLFLVLRVGIYLCIKLKHTKKRQIYTDIE



truncation
MNRLGK






Vesicular stomatitis
MKCLLYLAFLFIGVNCKFFGDTGLSKNPIEFVEG
55.


Indiana virus (strain
WFSSWKSSIASFFFIIGLIIGLFLVLRVGIYLCI



Orsay) (VSOG) 440
KLKHTKKRQIYTDIEMNRLGK



truncation







Vesicular stomatitis
MKCLLYLAFLFIGVNCKKNPIEFVEGWFSSWKSS
56.


Indiana virus (strain
IASFFFIIGLIIGLFLVLRVGIYLCIKLKHTKKR



Orsay) (VSOG) 448
QIYTDIEMNRLGK



truncation







Piry glycoprotein
MKCLLYLAFLFIGVNCKIDHPQMPDAKSVLPEDE
57.


426 truncation
EIFFGDTGVSKNPIELIQGWFSNWRESVMAIVGI




VLLIVVTFLAIKTVRVLNCLWRPRKKRIVRQEVD




VESRLNHFEMRGFPEYVKR






Piry glycoprotein
MKCLLYLAFLFIGVNCKFFGDTGVSKNPIELIQG
58.


445 truncation
WFSNWRESVMAIVGIVLLIVVTFLAIKTVRVLNC




LWRPRKKRIVRQEVDVESRLNHFEMRGFPEYVKR






Piry glycoprotein
MKCLLYLAFLFIGVNCKKNPIELIQGWFSNWRES
59.


453 truncation
VMAIVGIVLLIVVTFLAIKTVRVLNCLWRPRKKR




IVRQEVDVESRLNHFEMRGFPEYVKR






Maraba glycoprotein
MKCLLYLAFLFIGVNCKFEHPHAKDAASQLPDDE
60.


421 truncation
TLFFGDTGLSKNPVELVEGWFSSWKSTLASFFLI




IGLGVALIFIIRIIVAIRYKYKGRKTQKIYNDVE




MSRLGNK






Maraba glycoprotein
MKCLLYLAFLFIGVNCKFFGDTGLSKNPVELVEG
61.


440 truncation
WFSSWKSTLASFFLIIGLGVALIFIIRIIVAIRY




KYKGRKTQKIYNDVEMSRLGNK






Maraba glycoprotein
MKCLLYLAFLFIGVNCKKNPVELVEGWFSSWKST
62.


448 truncation
LASFFLIIGLGVALIFIIRIIVAIRYKYKGRKTQ




KIYNDVEMSRLGNK






Chandipura
MKCLLYLAFLFIGVNCKLDHPQLPHAQSIADDSE
63.


glycoprotein 430
EIFFGDTGVSKNPVELVTGWFTSWKESLAAGVVL



truncation
ILVVVLIYGVLRCFPVLCTTCRKPKWKKGVERSD




SFEMRIFKPNNMRARV






Chandipura
MKCLLYLAFLFIGVNCKFFGDTGVSKNPVELVTG
64.


glycoprotein 449
WFTSWKESLAAGVVLILVVVLIYGVLRCFPVLCT



truncation
TCRKPKWKKGVERSDSFEMRIFKPNNMRARV






Chandipura
MKCLLYLAFLFIGVNCKKNPVELVTGWFTSWKES
65.


glycoprotein 457
LAAGVVLILVVVLIYGVLRCFPVLCTTCRKPKWK



truncation
KGVERSDSFEMRIFKPNNMRARV






Isfahan glycoprotein
MKCLLYLAFLFIGVNCKVDHPHVPIAQAFVSEGE
66.


429 truncation
EVFFGDTGVSKNPIELISGWFSDWKETAAALGFA




AISVILIIGLMRLLPLLCRRRKQKKVIYKDVELN




SFDPRQAFHR






Isfahan glycoprotein
MKCLLYLAFLFIGVNCKFFGDTGVSKNPIELISG
67.


448 truncation
WFSDWKETAAALGFAAISVILIIGLMRLLPLLCR




RRKQKKVIYKDVELNSFDPRQAFHR






Isfahan glycoprotein
MKCLLYLAFLFIGVNCKKNPIELISGWFSDWKET
68.


456 truncation
AAALGFAAISVILIIGLMRLLPLLCRRRKQKKVI




YKDVELNSFDPRQAFHR






vesicular stomatitis
MKCLLYLAFLFIGVNCKFEHPHIQDAASQLPDDE
69.


Glasgow
ILFFGDTGLSKNPIDFVEGWFSSWKSSIASFFFI



glycoprotein
IGLIIGLFLVLRVGIYLYIKLKHTKKRQIYTDIE



(VSGG) 421
MNRLGR



truncation







vesicular stomatitis
MKCLLYLAFLFIGVNCKFFGDTGLSKNPIDFVEG
70.


Glasgow
WFSSWKSSIASFFFIIGLIIGLFLVLRVGIYLYI



glycoprotein
KLKHTKKRQIYTDIEMNRLGR



(VSGG) 440




truncation







vesicular stomatitis
MKCLLYLAFLFIGVNCKKNPIDFVEGWFSSWKSS
71.


Glasgow
IASFFFIIGLIIGLFLVLRVGIYLYIKLKHTKKR



glycoprotein
QIYTDIEMNRLGR



(VSGG) 448




truncation







Carajas virus
MKCLLYLAFLFIGVNCKIEHPHAKEAQKVVDDSE
72.


glycoprotein (CVG)
VIFFGDTGVSKNPVEVVEGWFSGWRSSLMSIFGI



430 truncation
ILLIVCLVLIVRILIALKYCCVRHKKRTIYKEDL




EMGRIPRRA






Carajas virus
MKCLLYLAFLFIGVNCKFFGDTGVSKNPVEVVEG
73.


glycoprotein (CVG)
WFSGWRSSLMSIFGIILLIVCLVLIVRILIALKY



449 truncation
CCVRHKKRTIYKEDLEMGRIPRRA






Carajas virus
MKCLLYLAFLFIGVNCKKNPVEVVEGWFSGWRSS
74.


glycoprotein (CVG
LMSIFGIILLIVCLVLIVRILIALKYCCVRHKKR



457 truncation)
TIYKEDLEMGRIPRRA






Radi virus
MKCLLYLAFLFIGVNCKIDHPQKAIASVHLNTDE
75.


glycoprotein 427
ELFFGNTGSDSNPVEAVEGWFASWKSAGINMALI



truncation
VLCVLLVLIFLRSLPALIKLIHRYRVSRSRQTDV




ELNSINETARTGSVGPDIIPGAWRVHDSGVRQSQ




FFRNNPRRLGP






Radi virus
MKCLLYLAFLFIGVNCKFFGNTGSDSNPVEAVEG
76.


glycoprotein 446
WFASWKSAGINMALIVLCVLLVLIFLRSLPALIK



truncation
LIHRYRVSRSRQTDVELNSINETARTGSVGPDII




PGAWRVHDSGVRQSQFFRNNPRRLGP






Radi virus
MKCLLYLAFLFIGVNCKSNPVEAVEGWFASWKSA
77.


glycoprotein 454
GINMALIVLCVLLVLIFLRSLPALIKLIHRYRVS



truncation
RSRQTDVELNSINETARTGSVGPDIIPGAWRVHD




SGVRQSQFFRNNPRRLGP






Jurona glycoprotein
MKCLLYLAFLFIGVNCKIDHPQRAHAQAVLGDEE
78.


429 truncation
TLFFGDTGVGKNPVELITGWFSGWKETIMAVVAI




FLLVIVLYGVLRCCPTICVLCKRKSRHRTKDMEM




QYIPNNQRHWR






Jurona glycoprotein
MKCLLYLAFLFIGVNCKFFGDTGVGKNPVELITG
79.


448 truncation
WFSGWKETIMAVVAIFLLVIVLYGVLRCCPTICV




LCKRKSRHRTKDMEMQYIPNNQRHWR






Jurona glycoprotein
MKCLLYLAFLFIGVNCKKNPVELITGWFSGWKET
80.


456 truncation
IMAVVAIFLLVIVLYGVLRCCPTICVLCKRKSRH




RTKDMEMQYIPNNQRHWR






Malpais Spring
MKCLLYLAFLFIGVNCKMDHPHLVHAKKYVSEDD
81.


glycoprotein 428
EIYFGDTGVSHNPVEIFSGWFTNWKEGLMKFSIL



truncation
VLSILIFYVVIRLVMCIPLKCKKERKPRLEFELQ




PREWEYSRA






Malpais Spring
MKCLLYLAFLFIGVNCKYFGDTGVSHNPVEIFSG
82.


glycoprotein 447
WFTNWKEGLMKFSILVLSILIFYVVIRLVMCIPL



truncation
KCKKERKPRLEFELQPREWEYSRA






Malpais Spring
MKCLLYLAFLFIGVNCKHNPVEIFSGWFTNWKEG
83.


glycoprotein 455
LMKFSILVLSILIFYVVIRLVMCIPLKCKKERKP



truncation
RLEFELQPREWEYSRA






Perinet Spring
MKCLLYLAFLFIGVNCKIDHPQIPDASGILPNSE
84.


glycoprotein 429
QVYYGDTGVSKNPIELIEGWFANWKETVMSIVGL



truncation
VLLITIVFTVLKCIGTCRSLRRKRKIEKDIELQE




IGPYQPTTYRPR






Perinet Spring
MKCLLYLAFLFIGVNCKYYGDTGVSKNPIELIEG
85.


glycoprotein 448
WFANWKETVMSIVGLVLLITIVFTVLKCIGTCRS



truncation
LRRKRKIEKDIELQEIGPYQPTTYRPR






Perinet Spring
MKCLLYLAFLFIGVNCKKNPIELIEGWFANWKET
86.


glycoprotein 456
VMSIVGLVLLITIVFTVLKCIGTCRSLRRKRKIE



truncation
KDIELQEIGPYQPTTYRPR






Morreton
MKCLLYLAFLFIGVNCKFEHPHIQDAASQLPDDE
87.


glycoprotein 422
TLFFGDTGLSKNPIELVEGWFSGWKSTIASFFFI



truncation
IGLVIGLYLVLRIGIALCIKCRVQEKRPKIYTDV




EMNRLDR






Morreton
MKCLLYLAFLFIGVNCKFFGDTGLSKNPIELVEG
88.


glycoprotein 441
WFSGWKSTIASFFFIIGLVIGLYLVLRIGIALCI



truncation
KCRVQEKRPKIYTDVEMNRLDR






Morreton
MKCLLYLAFLFIGVNCKKNPIELVEGWFSGWKST
89.


glycoprotein 449
IASFFFIIGLVIGLYLVLRIGIALCIKCRVQEKR



truncation
PKIYTDVEMNRLDR





#, SEQ ID NO:






In some embodiments, the tENV is used in place of an ENV protein in a standard VLP, e.g., those VLPs described in previous publications.29,39,40 In some embodiments, the tENV is the only virally-derived component of eVLPs, e.g., as described in WO 2022/020800 (incorporated herein by reference in its entirety); we refer to these herein as minimal VLPs (mVLPs). In some embodiments, the VLPs or mVLPs can be composed of a mixture of ectosomes and exosomes that can be separated by purification, if desired. In part because of the above mentioned design simplifications and optimizations, mVLPs are particularly suited for delivery of cargo including but not limited to DNA, RNA, protein, or combinations of biomolecules and/or chemicals, such as DNA-encoded or RNP-based genome editing reagents.


Thus described herein are various embodiments of minimal virus-like particles (mVLPs), which provide a platform for the delivery of cargo including nucleic acids and proteins, e.g., genome editing reagents and other biomolecules. The VLPs and mVLPs described herein can deliver a wide variety of cargo including but not limited to DNA only, DNA+RNA+protein, DNA+protein, RNA+protein, or protein only. mVLPs can control the form of the cargo (DNA, protein, and/or RNA).


Phospholipid Bilayer Recruitment Domains

Conventional VLPs that have been engineered to encapsulate and deliver protein-based cargo commonly fuse cargo to the INT or GAG polyprotein.25-27,29,30,39,40 After transient transfection of production plasmid DNA constructs, these protein fusions are translated in the cytosol of conventional VLP production cell lines, the gag matrix is acetylated and recruited to the cell membrane, and the gag fusions are encapsulated (transient transfected DNA is also unintentionally and passively encapsulated) within VLPs as VLPs bud off of the membrane into extracellular space.


In contrast, in some embodiments, proteins can be packaged into the mVLPs by fusing select phospholipid bilayer recruitment domains, preferably human protein-derived phospholipid bilayer recruitment domains to protein-based cargo (e.g., as shown in Table 6).


One such human protein-derived phospholipid bilayer recruitment domain used for this purpose is a human pleckstrin homology (PH) domain. PH domains interact with phosphatidylinositol lipids and proteins within biological membranes, such as PIP2, PIP3, βγ-subunits of GPCRs, and PKC.41,42 Alternatively, the human Arc protein can be fused to protein-based cargo to recruit cargo to the cytosolic side of the phospholipid bilayer.43 These human protein-derived phospholipid bilayer recruitment domains, or variants thereof (e.g., as shown in Table 6) can be fused to the N-terminus or C-terminus of protein-based cargo via polypeptide linkers of variable length regardless of the location or locations of one or more nuclear localization sequence(s) (NLS) within the cargo. Preferably, the linker between protein-based cargo and the phospholipid bilayer recruitment domain is a polypeptide linker 5-20, e.g., 8-12, e.g., 10, amino acids in length primarily composed of glycines and serines. The human protein-derived phospholipid bilayer recruitment domain localizes the cargo to the cytosolic face of the phospholipid bilayer and this protein cargo is packaged within mVLPs that utilize an envelope glycoprotein to trigger budding-off of particles from the producer cell into extracellular space. These human protein-derived domains and proteins can facilitate for localization of cargo to the cytosolic face of the plasma membrane within the mVLP production cells, and they also allow for cargo to localize to the nucleus of mVLP-transduced cells without the utilization of exogenous retroviral gag/pol or chemical and/or light-based dimerization systems. The delivery of Cas9, for example, is significantly more efficiently loaded as cargo into particles with fusion to a phospholipid bilayer recruitment domain compared to without fusion to a phospholipid bilayer recruitment domain.


VLP/mVLP-Mediated Delivery of DNAs, Proteins and RNAs

The VLPs and mVLPs described herein (e.g., comprising tENV proteins) can package and deliver biomolecule cargo. “Cargo” refers to a any payload that can be delivered, including chemicals, e.g., small molecule compounds, and biomolecules, including DNA, RNA, peptide nucleic acid (PNA), RNP, proteins, and combinations thereof, including combinations of DNA and RNP, RNP, combinations of DNA and proteins, or proteins, as well as viruses and portions thereof, e.g., for therapeutic or diagnostic use, or for the applications of genome editing, epigenome modulating, and/or transcriptome modulation. RNA in this context includes, for example, single guide RNA (sgRNA), Clustered Regularly Interspaced Palindromic Repeat (CRISPR) RNA (crRNA), and/or mRNA coding for cargo. Other exemplary nucleic acids can include specialty single and/or double-stranded DNA molecules (e.g., plasmid, mini circle, closed-ended linear DNA, AAV DNA, episomes, bacteriophage DNA, homology directed repair templates, etc.), single and/or double-stranded RNA molecules (e.g., single guide RNA, prime editing guide RNA, crRNA, tracrRNA, messenger RNA, transfer RNA, long non-coding RNA, circular RNA, RNA replicon, circular or linear splicing RNA, micro RNA, small interfering RNA, short hairpin RNA, piwi-interacting RNA, toehold switch RNA, RNAs that can be bound by RNA binding proteins, bacteriophage RNA, or internal ribosomal entry site containing RNA). Combinations of the above cargos (e.g., AAV particles and/or ribonucleoprotein (RNP) complexes comprising RNA and protein, e.g., guide RNA/CRISPR Cas protein complexes) can also be included.


As used herein, “small molecules” refers to small organic or inorganic molecules of molecular weight below about 3,000 Daltons. In general, small molecules useful for the invention have a molecular weight of less than 3,000 Daltons (Da). The small molecules can be, e.g., from at least about 100 Da to about 3,000 Da (e.g., between about 100 to about 3,000 Da, about 100 to about 2500 Da, about 100 to about 2,000 Da, about 100 to about 1,750 Da, about 100 to about 1,500 Da, about 100 to about 1,250 Da, about 100 to about 1,000 Da, about 100 to about 750 Da, about 100 to about 500 Da, about 200 to about 1500, about 500 to about 1000, about 300 to about 1000 Da, or about 100 to about 250 Da).


In some embodiments, the cargo is limited by the diameter of the particles, e.g., which in some embodiments can range from 30 nm to 500 nm.


In some embodiments, the cargo can include a combination of DNA and RNA; for example, the VLPs or mVLPs can be produced via transient transfection of a production cell line. DNA that is transfected into cells will possess size-dependent mobility such that a fraction of the transfected DNA will remain in the cytosol while another fraction of the transfected DNA will localize to the nucleus.44-46 A fraction of the transfected DNA in the nucleus will be expressed components needed to create the VLPs/mVLPs and another fraction in the cytosol/near the plasma membrane will be encapsulated and delivered in VLPs/mVLPs. See, e.g., FIGS. 1-4 of WO 2022/020800.


Cargo developed for applications of genome or gene editing also includes CRISPR-Cas nucleases and fusions and variants thereof, e.g., prime editors, and base editors. Nucleases include ZFNs and Transcription activator-like effector nucleases (TALENs) that comprise a FokI or AcuI nuclease domain; and CRISPR Cas proteins or a functional derivative thereof (e.g., as shown in Table 2) (ZFNs are described, for example, in United States Patent Publications 20030232410; 20050208489; 20050026157; 20050064474; 20060188987; 20060063231; and International Publication WO 07/014275) (TALENs are described, for example, in United States Patent Publication U.S. Pat. No. 9,393,257B2; and International Publication WO2014134412A1) (CRISPR Cas proteins are described, for example, in United States Patent Publications U.S. Pat. No. 8,697,359B1; US20180208976A1; and International Publications WO2014093661A2; WO2017184786A8).34-36 Base editors can include any CRISPR based nuclease orthologs (wt, nickase, or catalytically inactive (CI)), e.g., as shown in Table 2, fused at the N-terminus to a nucleotide deaminase or nucleoside deaminase or a functional derivative thereof (e.g., as shown in Table 3), or comprising a deaminase domain inlaid internally, with or without a fusion at the C-terminus to one or multiple uracil glycosylase inhibitors (UGIs) using polypeptide linkers of variable length (Base editors are described, for example, in United States Patent Publications US20150166982A1; US20180312825A1; U.S. Ser. No. 10/113,163B2; and International Publications WO2015089406A1; WO2018218188A2; WO2017070632A2; WO2018027078A8; WO2018165629A1).37,38 In addition, prime editors are also compatible with mVLP delivery modalities (Prime editors are described, for example, in Anzalone et al., Nature. 2019 December; 576(7785):149-157). Prime editors can be delivered, e.g., as fusions of Cas nickase to a reverse transcriptase or as separate components (see, e.g., Grunewald et al., Nat Biotechnol. 2022 Sep. 26. doi: 10.1038/s41587-022-01473-1; and Liu et al., Nat Biotechnol. 2022 September;40(9):1388-1393).


Cargo designed for the purposes of epigenome modulating includes CRISPR Cas proteins, zinc fingers (ZFs) and TALEs fused to an epigenome/epigenetic modulating agent or combination of epigenome/epigenetic modulating agent or a functional derivative thereof connected together by one or more variable length polypeptide linkers. Exemplary epigenetic modulating agents include CRISPR-Cas proteins (e.g., nickases or catalytically inactive Cas) fused to DNA methylases, histone acetyltransferases, and deacetylases, as well as transcriptional activators or repressors (see, e.g., Tables 2 & 4). Examples include, e.g., transcriptional repressors (e.g., KRAB, ERD, SID, and others, e.g., amino acids 473-530 of the ets2 repressor factor (ERF) repressor domain (ERD), amino acids 1-97 of the KRAB domain of KOX1, or amino acids 1-36 of the Mad mSIN3 interaction domain (SID); see Beerli et al., PNAS USA 95:14628-14633 (1998)) or silencers such as Heterochromatin Protein 1 (HP1, also known as swi6), e.g., HPlu or HP10; proteins or peptides that could recruit long non-coding RNAs (lncRNAs) fused to a fixed RNA binding sequence such as those bound by the MS2 coat protein, endoribonuclease Csy4, or the lambda N protein; enzymes that modify the methylation state of DNA (e.g., DNA methyltransferase (DNMT) or TET proteins); or enzymes that modify histone subunits (e.g., histone acetyltransferases (HAT), histone deacetylases (HDAC), histone methyltransferases (e.g., for methylation of lysine or arginine residues) or histone demethylases (e.g., for demethylation of lysine or arginine residues)) In some embodiments, the sequence of the cargo is at least 95% identical to a sequence set forth herein.


sgRNAs can complex with genome editing reagents during the packaging process to be co-delivered within VLPs/mVLPs as described herein. Also, linear or circular RNAs encoding cargo or edits that are to be installed by a prime editor could be co-packaged with genome editing reagents that are fused to RNA binding proteins, such as MS2, PP7, COM, or TAR hairpin binding protein (TBP) or human SLBP. Cargo designed for the purposes of transcriptome editing includes CRISPR Cas proteins or any functional derivatives thereof (e.g., as shown in Table 5) or CRISPR Cas proteins or any functional derivatives thereof (e.g., as shown in Table 5) fused to nucleotide deaminases or nucleoside deaminases (e.g., as shown in Table 3) by one or more variable length polypeptide linkers.


The cargo can also include any therapeutically or diagnostically useful protein, DNA, RNP, or combination of DNA, protein and/or RNP. See, e.g., WO2014005219; U.S. Ser. No. 10/137,206; US20180339166; U.S. Pat. No. 5,892,020A; EP2134841B1; WO2007020965A1. For example, cargo encoding or composed of nuclease or base editor proteins or RNPs or derivatives thereof can be delivered to retinal cells for the purposes of correcting a splice site defect responsible for Leber Congenital Amaurosis type 10. In the mammalian inner ear, VLP/mVLP delivery of base editing reagents or HDR promoting cargo to sensory cells such as cochlear supporting cells and hair cells for the purposes of editing β-catenin (β-catenin Ser 33 edited to Tyr, Pro, or Cys) in order to better stabilize β-catenin could help reverse hearing loss.


In another application, VLP/mVLP delivery of RNA editing reagents or proteome perturbing reagents could cause a transitory reduction in cellular levels of one or more specific proteins of interest (potentially at a systemic level, in a specific organ or a specific subset of cells, such as a tumor), and this could create a therapeutically actionable window when secondary drug(s) could be administered (this secondary drug is more effective in the absence of the protein of interest or in the presence of lower levels of the protein of interest). For example, mVLP delivery of RNA editing reagents or proteome perturbing reagents could trigger targeted degradation of MAPK and PI3K/AKT proteins and related mRNAs in vemurafenib/dabrafenib-resistant BRAF-driven tumor cells, and this could open a window for the administration of vemurafenib/dabrafenib because BRAF inhibitor resistance is temporarily abolished (resistance mechanisms based in the MAPK/PI3K/AKT pathways are temporarily downregulated by VLP/mVLP cargo). This example is especially pertinent when combined with VLP/mVLPs that are antigen inducible and therefore specific for tumor cells. Alternatively, the transitory reduction in cellular levels of a specific protein of interest may itself have therapeutic benefit.


In some embodiments, VLPs and mVLPs described herein could be used deliver factors, e.g., including the Yamanaka factors Oct3/4, Sox2, Klf4, and c-Myc, to cells such as human or mouse fibroblasts, in order to generate induced pluripotent stem cells or to deliver factors that induce forward differentiation or trans-differentiation into a specific cell-type.


In some embodiments, VLPs and mVLPs described herein could deliver dominant-negative forms of proteins in order to elicit a therapeutic effect.


VLPs and mVLPs described herein that are antigen-specific (e.g., tumor-antigen specific) could be targeted to cancer cells in order to deliver proapoptotic proteins BIM, BID, PUMA, NOXA, BAD, BIK, BAX, BAK and/or HRK in order to trigger apoptosis of cancer cells. Tumor antigens are known in the art.


90% of pancreatic cancer patients present with unresectable disease. Around 30% of patients with unresectable pancreatic tumors will die from local disease progression, so it is desirable to treat locally advanced pancreatic tumors with ablative radiation, but the intestinal tract cannot tolerate high doses of radiation needed to cause tumor ablation. Selective radioprotection of the intestinal tract enables ablative radiation therapy of pancreatic tumors while minimizing damage done to the surrounding gastrointestinal tract. To this end, VLPs and mVLPs described herein could be loaded with dCas9 fused to the transcriptional repressor KRAB and guide RNA targeting EGLN. EGLN inhibition has been shown to significantly reduce gastrointestinal toxicity from ablative radiation treatments because it causes selective radioprotection of the gastrointestinal tract but not the pancreatic tumor.47 Such fusion proteins, VLPs and mVLPs described herein, and methods of making and using the same are provided herein.


Unbound steroid receptors reside in the cytosol. After binding to ligands, these receptors will translocate to the nucleus and initiate transcription of response genes. VLPs and mVLPs described herein could deliver single chain variable fragment (scFv) antibodies to the cytosol of cells that bind to and disrupt cytosolic steroid receptors. For example, the scFv could bind to the glucocorticoid receptor and prevent it from binding dexamethasone, and this would prevent transcription of response genes, such as metallothionein 1E that has been linked to tumorigenesis.48 VLPs and mVLPs described herein can be indicated for treatments that involve targeted disruption of proteins. For example, VLPs and mVLPs described herein can be utilized for targeting and disrupting proteins in the cytosol of cells by delivering antibodies/scFvs to the cytosol of cells. Classically, delivery of antibodies through the plasma membrane to the cytosol of cells has been notoriously difficult and inefficient. This mode of protein inhibition is similar to how a targeted small molecule binds to and disrupts proteins in the cytosol and could be useful for the treatment of a diverse array of diseases.49-51 Such fusion proteins, VLPs and mVLPs described herein, and methods of making and using the same are included herein.


In addition, the targeting of targeted small molecules is limited to proteins of a certain size that contain binding pockets which are relevant to catalytic function or protein-protein interactions. scFvs are not hampered by these limitations because scFvs can be generated that bind to many different moieties of a protein in order to disrupt catalysis and interactions with other proteins. For example, RAS oncoproteins are implicated across a multitude of cancer subtypes, and RAS is one of the most frequently observed oncogenes in cancer. For instance, the International Cancer Genome Consortium found KRAS to be mutated in 95% of their Pancreatic Adenocarcinoma samples. RAS isoforms are known to activate a variety of pathways that are dysregulated in human cancers, like the PI3K and MAPK pathways. Despite the aberrant roles RAS plays in cancer, no efficacious pharmacologic direct or indirect small molecule inhibitors of RAS have been developed and approved for clinical use. One strategy for targeting RAS could be VLPs and mVLPs described herein that can deliver specifically to cancer cells scFvs that bind to and disrupt the function of multiple RAS isoforms.49-51


VLP/mVLP Composition, Production, Purification and Applications

The VLPs and mVLPs described herein (e.g., comprising tENV proteins) can be produced from producer cell lines that are either transiently transfected with at least one plasmid and/or that stably express constructs that have been integrated into the producer cell line genomic DNA. This, in some embodiments, the VLPs and mVLPs described herein can be produced and package protein-based cargo by integrating all production DNA constructs into the genomic DNA of production cell lines. Once cell lines are created, protein delivery VLPs and mVLPs can be produced in a constitutive or inducible fashion.


Some or all of the components for producing VLPs or mVPLs can be transiently expressed. In some embodiments, if a single plasmid is used in the transfection, it should comprise sequences encoding tENV as described herein, cargo (e.g., a therapeutic protein or a gene editing reagent such as a zinc finger, transcription activator-like effector (TALE), and/or CRISPR-based genome editing/modulating protein and/or RNP having a sequence such as those found in Tables 2, 3, 4 & 5; in some embodiments, the sequence is at least 95% identical to a sequence set forth herein), with or without fusion to a phospholipid bilayer recruitment domain (e.g., as shown in Table 6), and a guide RNA, if necessary. Preferably, two to three plasmids are used in the transfection. These two to three plasmids can include the following (any two or more can be combined in a single plasmid):

    • 1. A plasmid comprising sequences encoding cargo, e.g., a therapeutic protein or a genome editing reagent, with or without a fusion to a phospholipid bilayer recruitment domain.
    • 2. A plasmid comprising sequences encoding tENV as described herein.
    • 3. If the genome editing reagent from plasmid 1 requires one or more guide RNAs, a plasmid comprising one or more guide RNAs apposite for the genome editing reagent.


      If it is desired to deliver a type of DNA molecule other than plasmid(s), the above-mentioned transfection can be performed with double-stranded closed-end linear DNA, episome, mini circle, double-stranded oligonucleotide and/or other specialty/modified DNA, RNA, AAV, adenovirus, anellovirus, or peptide nucleic acid (PNA) molecules. Alternatively, the producer cell line can be made to stably express any one or more of the constructs (1 through 3) described in the transfection above.


In some embodiments, the methods can include using cells that have or have not been manipulated to express any exogenous proteins except for a tENV viral envelope protein (as described herein, e.g., as shown in Table 1), and, if desired, a phospholipid bilayer recruitment domain (e.g., as shown in Table 6); in other words, no cargo is expressed. In this embodiment, the “empty” particles that are produced can be loaded with cargo and/or small molecules by utilizing incubation, nucleofection, lipid, polymer, or CaCl2) transfection, sonication, freeze thaw, and/or heat shock of purified particles mixed with cargo. In some embodiments, producer cells do not express any gag, pro, or pol protein. This type of loading allows for cargo to be unmodified by fusions to phospholipid bilayer recruitment domains and represents a significant advancement from previous VLP technology.


The plasmids, or other types of specialty DNA molecules known in the art or described herein, can also preferably include other elements to drive expression or translation of the encoded sequences, e.g., a promoter sequence; an enhancer sequence, e.g., 5′ untranslated region (UTR) or a 3′ UTR; a polyadenylation site; an insulator sequence; or another sequence that increases or controls expression (e.g., an inducible promoter element).


Appropriate producer cell lines can include primary or stable human cell lines refractory to the effects of transfection reagents and fusogenic effects due to virally-derived glycoproteins. Examples of appropriate cell lines include Human Embryonic Kidney (HEK) 293 cells, HEK293 T/17 SF cells kidney-derived Phoenix-AMPHO cells, and placenta-derived BeWo cells. For example, such cells could be selected for their ability to grow as adherent cells, or suspension cells. In some embodiments, the producer cells can be cultured in classical DMEM under serum conditions, serum-free conditions, or exosome-free serum conditions. mVLPs can be produced from cells that have been derived from patients (autologous mVLPs) and other FDA-approved cell lines (allogenic mVLPs) as long as these cells can be transfected with DNA constructs that encode the aforementioned mVLP production components by various techniques known in the art.


In addition, if it is desirable, more than one genome editing reagent can be included in the transfection. The DNA constructs can be designed to overexpress proteins in the producer cell lines. The plasmid backbones, for example, used in the transfection can be familiar to those skilled in the art, such as the pCDNA3 backbone that employs the CMV promoter for RNA polymerase II transcripts or the U6 promoter for RNA polymerase III transcripts. Various techniques known in the art may be employed for introducing nucleic acid molecules into producer cells. Such techniques include chemical-facilitated transfection using compounds such as calcium phosphate, cationic lipids, cationic polymers, liposome-mediated transfection, such as cationic liposome like LIPOFECTAMINE (LIPOFECTAMINE 2000 or 3000 and TransIT-X2), polyethyleneimine, non-chemical methods such as electroporation, particle bombardment, or microinjection.


A human producer cell line that stably expresses the necessary VLP or mVLP components in a constitutive and/or inducible fashion can be used for production of VLPs or mVLPs. For example, mVLPs can be produced from cells that have been derived from patients (autologous mVLPs) and other FDA-approved cell lines (allogenic mVLPs) if these cells have been converted into stable cell lines that express the aforementioned mVLP components.


Also provided herein are the producer cells themselves.


Production of Cargo-Loaded VLPs/mVLPs and Compositions

Preferably the VLPs/mVLPs described herein are harvested from cell culture medium supernatant 36-48 hours post-transfection, or when the VLPs/mVLPs are at the maximum concentration in the medium of the producer cells (the producer cells are expelling particles into the media and at some point in time, the particle concentration in the media will be optimal for harvesting the particles). Supernatant can be purified by any known methods in the art, such as centrifugation, ultracentrifugation, precipitation, ultrafiltration, and/or chromatography. In some embodiments, the supernatant is first filtered, e.g., to remove particles larger than 1 μm, e.g., through 0.45 pore size polyvinylidene fluoride hydrophilic membrane (Millipore Millex-HV) or 0.8 m pore size mixed cellulose esters hydrophilic membrane (Millipore Millex-AA). After filtration, the supernatant can be further purified and concentrated, e.g., using ultracentrifugation, e.g., at a speed of 80,000 to 100,000×g at a temperature between 1° C. and 5° C. for 1 to 2 hours, or at a speed of 8,000 to 15,000 g at a temperature between 1° C. and 5° C. for 10 to 16 hours. After this centrifugation step, the VLPs/mVLPs are concentrated in the form of a centrifugate (pellet), which can be resuspended to a desired concentration, mixed with transduction-enhancing reagents, subjected to a buffer exchange, or used as is. In some embodiments, VLP/mVLP-containing supernatant can be filtered, precipitated, centrifuged and resuspended to a concentrated solution. For example, polyethylene glycol (PEG), e.g., PEG 8000, or antibody-bead conjugates that bind to VLP/mVLP-surface proteins or membrane components can be used to precipitate particles. Purified particles are stable and can be stored at 4° C. for up to a week or −80° C. for years without losing appreciable activity.


Preferably, VLPs/mVLPs are resuspended or undergo buffer exchange so that particles are suspended in an appropriate carrier. In some embodiments, buffer exchange can be performed by ultrafiltration (Sartorius Vivaspin 500 MWCO 100,000). An exemplary appropriate carrier for VLPs/mVLPs to be used for in vitro applications would preferably be a cell culture medium that is suitable for the cells that are to be transduced by VLPs/mVLPs. Transduction-enhancing reagents that can be mixed into the purified and concentrated VLPs/mVLPs solution for in vitro applications include reagents known by those familiar with the art (Miltenyi Biotec Vectofusin-1, Millipore Polybrene, Takara Retronectin, Sigma Protamine Sulfate, and the like). After VLPs/mVLPs in an appropriate carrier are applied to the cells to be transduced, transduction efficiency can be further increased by centrifugation. Preferably, the plate containing VLPs/mVLPs applied to cells can be centrifuged at a speed of 1,150 g at room temperature for 30 minutes. After centrifugation, cells are returned into the appropriate cell culture incubator (humidified incubator at 37° C. with 5% CO2).


An appropriate carrier for VLPs/mVLPs to be administered to a mammal, especially a human, would preferably be a pharmaceutically acceptable composition. A “pharmaceutically acceptable composition” refers to a non-toxic semisolid, liquid, or aerosolized filler, diluent, encapsulating material, colloidal suspension or formulation auxiliary of any type. Preferably, this composition is suitable for injection. These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and and similar solutions or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions. Another appropriate pharmaceutical form would be aerosolized particles for administration by intranasal inhalation or intratracheal intubation.


The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or suspensions. The solution or suspension may comprise additives which are compatible with VLPs/mVLPs and do not prevent VLPs/mVLPs entry into target cells. In all cases, the form must be sterile and must be fluid to the extent that the form can be administered with a syringe. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. An example of an appropriate solution is a buffer, such as phosphate buffered saline.


Methods of formulating suitable pharmaceutical compositions are known in the art, see, e.g., Remington: The Science and Practice of Pharmacy, 21st ed., 2005; and the books in the series Drugs and the Pharmaceutical Sciences: a Series of Textbooks and Monographs (Dekker, NY). For example, solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.


Pharmaceutical compositions suitable for injectable use can include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle, which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying, which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.


Compositions comprising cargo-loaded VLPs/mVLPs as described herein can be included in a container, pack, or dispenser together with instructions for administration.









TABLE 2







Exemplary Potential Cas9 and Cas12a orthologs










DNA-binding Cas
Enzyme class
Nickase mutation
CI mutations





ortholog





SpCas9
Type II-A
D10A
D10A, H840A


SaCas9
Type II-A
D10A
D10A,


CjCas9
Type II-C
D8A
D8A,


NmeCas9
Type II-C
D16A
D16A, H588A


AsCas12a
Type II-C

D908A, E993A


LbCas12a
Type II-C

D832A, E925A





Nickase mutation residues represents a position of the enzyme either known to be required for catalytic activity of the conserved RuvC nuclease domain or predicted to be required for this catalytic activity based on sequence alignment to CjCas9 where structural information is lacking (* indicates which proteins lack sufficient structural information). All positional information refers to the wild-type protein sequences acquired from uniprot.org.













TABLE 3







Exemplary Deaminase domains and their


substrate sequence preferences.








Deaminase
Nucleotide sequence preference





hAID
5′-WRC


rAPOBEC1*
5′-TC ≥ CC ≥ AC > GC


mAPOBEC3
5′-TYC


hAPOBEC3A
5′-TCG


hAPOBEC3B
5′-TCR > TCT


hAPOBEC3C
5′-WYC


hAPOBEC3F
5′-TTC


hAPOBEC3G
5′-CCC


hAPOBEC3H
5′-TTCA ~ TTCT ~ TTCG > ACCCA > TGCA



E. coli TadA

A


hAdar1
A


hAdar2
A





Nucleotide positions that are poorly specified or are permissive of two or more nucleotides are annotated according to IUPAC codes, where W = A or T, R = A or G, and Y = C or T. “h” before the deaminase name indicates Homo sapiens origin. “m” before the deaminase name indicates Mus musculus origin. “r” before the deaminase name indicates Rattus origin.













TABLE 4







Exemplary Epigenetic modulating domains.










Epigenetic modulator
Epigenetic modulation







VP16
transcriptional activation



VP64
transcriptional activation



P65
transcriptional activation



RTA
transcriptional activation



KRAB
transcriptional repression



MeCP2
transcriptional repression



TET1
Methylation



DNMT3A
Methylation

















TABLE 5







Exemplary CRISPR based RNA-guided RNA binding enzymes










RNA-binding Cas ortholog
Enzyme class







LshCas13a
Type-VI



LwaCas13a
Type-VI

















TABLE 6







Exemplary Plasma membrane recruitment domains










Plasma membrane recruitment domain
Substitution(s)







Pleckstrin homology domain of human




phospholipase Cδ1 (hPLCδ1)



Pleckstrin homology domain of human
R40L52



phospholipase Cδ1 (hPLCδ1)



Pleckstrin homology domain of human



AKT1 (hAKT1)



Mutant Pleckstrin homology domain of
E17K53



human AKT1



Pleckstrin homology domain of human 3-



phosphoinositide-dependent protein



kinase 1 (hPDPK1)



Mutant Pleckstrin homology domain of
K14R56



human AKT1



Mutant Pleckstrin homology domain of
K8R57



human AKT1



Mutant Pleckstrin homology domain of
T72A58



human AKT1



Mutant Pleckstrin homology domain of
T92A59



human AKT1



Mutant Pleckstrin homology domain of
R25C52



human AKT1



Mutant Pleckstrin homology domain of
T34D54



human AKT1



Mutant Pleckstrin homology domain of
T34F54



human AKT1



Mutant Pleckstrin homology domain of
T34L54



human AKT1



Mutant Pleckstrin homology domain of
T81Y55



human AKT1



Mutant Pleckstrin homology domain of
K142A, H143A,



human AKT1
R144A60



Mutant Pleckstrin homology domain of
T101C61



human AKT1



Pleckstrin homology domain of Human



Dapp1



Pleckstrin homology domain of Human



GRP1



Pleckstrin homology domain of Human
R284C52



GRP1



Pleckstrin homology domain of Human



OSBP1



Pleckstrin homology domain of Human
R108E52



OSBP1



Pleckstrin homology domain of Human



ARNO (CYTH2)



Pleckstrin homology domain of Human
R279C52



ARNO (CYTH2)



Pleckstrin homology domain of Human



BTK1



Pleckstrin homology domain of Human
R28C52



BTK1



FYVE domain of Human EEA1



FYVE domain of Human EEA1
R1375L52



PX domain of p40phox (NCF4)



PX domain of p40phox (NCF4)
R58L52



Pleckstrin homology domain of Human



FAPP1



Pleckstrin homology domain of Human



CERT



Pleckstrin homology domain of Human



PHLPP1



Pleckstrin homology domain of Human



SWAP70



Pleckstrin homology domain of Human
R223E and



SWAP70
R224E62



Pleckstrin Homology Domain of Human



PKD



Pleckstrin homology domain of Human



MAPKAP1



Pleckstrin homology domain of Human



Son Of Sevenless Homolog 2



Pleckstrin homology domain of Human



Dynamin



Pleckstrin homology domain of Human



BCR



Pleckstrin homology domain of Human



DBS










Exemplary Sequences

In some embodiments, the sequence of a protein or nucleic acid used in a composition or method described herein is at least 80%, 85%, 90%, 95%, 97%, 98%, or 99% identical to a sequence set forth herein. To determine the percent identity of two amino acid sequences, or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). In a preferred embodiment, the length of a reference sequence aligned for comparison purposes is at least 80% of the length of the reference sequence, and in some embodiments is at least 90% or 100%. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid “homology”). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.


The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For example, the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch ((1970) J Mol. Biol. 48:444-453) algorithm which has been incorporated into the GAP program in the GCG software package (available on the world wide web at gcg.com), using the default parameters, e.g., a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.









Prime Editor: spCas9 H840A-MMLV Reverse


Transcriptase (delta RNase H domain):


(SEQ ID NO: 90)


MKRTADGSEFESPKKKRKVDKKYSIGLDIGTNSVGWAVITDEYKVPSKKF





KVLGNTDRHSIKKNLIGALLFDSGETAEATRLKRTARRRYTRRKNRICYL





QEIFSNEMAKVDDSFFHRLEESFLVEEDKKHERHPIFGNIVDEVAYHEKY





PTIYHLRKKLVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNPDNSDVD





KLFIQLVQTYNQLFEENPINASGVDAKAILSARLSKSRRLENLIAQLPGE





KKNGLFGNLIALSLGLTPNFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQI





GDQYADLFLAAKNLSDAILLSDILRVNTEITKAPLSASMIKRYDEHHQDL





TLLKALVRQQLPEKYKEIFFDQSKNGYAGYIDGGASQEEFYKFIKPILEK





MDGTEELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHAILRRQEDFYPF





LKDNREKIEKILTFRIPYYVGPLARGNSRFAWMTRKSEETITPWNFEEVV





DKGASAQSFIERMTNFDKNLPNEKVLPKHSLLYEYFTVYNELTKVKYVTE





GMRKPAFLSGEQKKAIVDLLFKTNRKVTVKQLKEDYFKKIECFDSVEISG





VEDRFNASLGTYHDLLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREMI





EERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKLINGIRDKQSGKTILDF





LKSDGFANRNFMQLIHDDSLTFKEDIQKAQVSGQGDSLHEHIANLAGSPA





IKKGILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQKGQKNSRERMK





RIEEGIKELGSQILKEHPVENTQLQNEKLYLYYLQNGRDMYVDQELDINR





LSDYDVDAIVPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNY





WRQLLNAKLITQRKFDNLTKAERGGLSELDKAGFIKRQLVETRQITKHVA





QILDSRMNTKYDENDKLIREVKVITLKSKLVSDFRKDFQFYKVREINNYH





HAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVYDVRKMIAKSEQEIGKA





TAKYFFYSNIMNFFKTEITLANGEIRKRPLIETNGETGEIVWDKGRDFAT





VRKVLSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIARKKDWDPKKYG





GFDSPTVAYSVLVVAKVEKGKSKKLKSVKELLGITIMERSSFEKNPIDFL





EAKGYKEVKKDLIIKLPKYSLFELENGRKRMLASAGELQKGNELALPSKY





VNFLYLASHYEKLKGSPEDNEQKQLFVEQHKHYLDEIIEQISEFSKRVIL





ADANLDKVLSAYNKHRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTID





RKRYTSTKEVLDATLIHQSITGLYETRIDLSQLGGDSGGSSGGSSGSETP





GTSESATPESSGGSSGGSSTLNIEDEYRLHETSKEPDVSLGSTWLSDFPQ





AWAETGGMGLAVRQAPLIIPLKATSTPVSIKQYPMSQEARLGIKPHIQRL





LDQGILVPCQSPWNTPLLPVKKPGTNDYRPVQDLREVNKRVEDIHPTVPN





PYNLLSGLPPSHQWYTVLDLKDAFFCLRLHPTSQPLFAFEWRDPEMGISG





QLTWTRLPQGFKNSPTLFNEALHRDLADFRIQHPDLILLQYVDDLLLAAT





SELDCQQGTRALLQTLGNLGYRASAKKAQICQKQVKYLGYLLKEGQRWLT





EARKETVMGQPTPKTPRQLREFLGKAGFCRLFIPGFAEMAAPLYPLTKPG





TLFNWGPDQQKAYQEIKQALLTAPALGLPDLTKPFELFVDEKQGYAKGVL





TQKLGPWRRPVAYLSKKLDPVAAGWPPCLRMVAAIAVLTKDAGKLTMGQP





LVILAPHAVEALVKQPPDRWLSNARMTHYQALLLDTDRVQFGPWVALNPA





TLLPLPEEGLQHNCLSGGSKRTADGSEPKKKRKVGS






Rattus norvegicus & synthetic: APOBEC1-XTEN



L8-nspCas9-UGI-SV40 NLS


(SEQ ID NO: 91)


MSSETGPVAVDPTLRRRIEPHEFEVFFDPRELRKETCLLYEINWGGRHSI





WRHTSQNTNKHVEVNFIEKFTTERYFCPNTRCSITWFLSWSPCGECSRAI





TEFLSRYPHVTLFIYIARLYHHADPRNRQGLRDLISSGVTIQIMTEQESG





YCWRNFVNYSPSNEAHWPRYPHLWVRLYVLELYCIILGLPPCLNILRRKQ





PQLTFFTIALQSCHYQRLPPHILWATGLKSGSETPGTSESATPESDKKYS





IGLAIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGALLFDSG





ETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFL





VEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKADLRLIYL





ALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENPINASGV





DAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTPNFKSNF





DLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAILLSDIL





RVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIFFDQSK





NGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLRKQRTFD





NGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLA





RGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDKNLPNEK





VLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVDLLFKTN





RKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKIIKDKDF





LDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQLKRRRY





TGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDDSLTFKE





DIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKWDELVKVMGRHKPE





NIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHPVENTQLQ





NEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFLKDDSIDNKVL





TRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNLTKAERGG





LSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLIREVKVIT





LKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKKYPKLESE





FVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEITLANGEI





RKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEVQTGGFSK





ESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKKL





KSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPKYSLFELE





NGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPEDNEQKQL





FVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDKPIREQAE





NIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQSITGLYE





TRIDLSQLGGDSGGSTNLSDIIEKETGKQLVIQESILMLPEEVEEVIGNK





PESDILVHTAYDESTDENVMLLTSDAPEYKPWALVIQDSNGENKIKMLSG





GSPKKKRKV






Homo sapiens: AID



(SEQ ID NO: 92)


MDSLLMNRRKFLYQFKNVRWAKGRRETYLCYVVKRRDSATSFSLDFGYLR





NKNGCHVELLFLRYISDWDLDPGRCYRVTWFTSWSPCYDCARHVADFLRG





NPNLSLRIFTARLYFCEDRKAEPEGLRRLHRAGVQIAIMTFKDYFYCWNT





FVENHERTFKAWEGLHENSVRLSRQLRRILLPLYEVDDLRDAFRTLGL






Homo sapiens: AIDv solubility variant



lacking N-terminal RNA-binding region


(SEQ ID NO: 93)


LMDPHIFTSNFNNGIGRHKTYLCYEVERLDSATSFSLDFGYLRNKNGCHV





ELLFLRYISDWDLDPGRCYRVTWFTSWSPCYDCARHVADFLRGNPNLSLR





IFTARLYFCEDRKAEPEGLRRLHRAGVQIAIMTFKDYFYCWNTFVENHER





TFKAWEGLHENSVRLSRQLRRILLPLYEVDDLRDAFRTLGL






Homo sapiens: AIDv solubility variant



lacking N-terminal RNA-binding region and


the C-terminal poorly structured region


(SEQ ID NO: 94)


MDPHIFTSNFNNGIGRHKTYLCYEVERLDSATSFSLDFGYLRNKNGCHVE





LLFLRYISDWDLDPGRCYRVTWFTSWSPCYDCARHVADFLRGNPNLSLRI





FTARLYFCEDRKAEPEGLRRLHRAGVQIAIMTFKDYFYCWNTFVENHERT





FKAWEGLHENSVRLSRQLRRILLPL






Rattus norvegicus: APOBEC1



(SEQ ID NO: 95)


MSSETGPVAVDPTLRRRIEPHEFEVFFDPRELRKETCLLYEINWGGRHSI





WRHTSQNTNKHVEVNFIEKFTTERYFCPNTRCSITWFLSWSPCGECSRAI





TEFLSRYPHVTLFIYIARLYHHADPRNRQGLRDLISSGVTIQIMTEQESG





YCWRNFVNYSPSNEAHWPRYPHLWVRLYVLELYCIILGLPPCLNILRRKQ





PQLTFFTIALQSCHYQRLPPHILWATGLK






Mus musculus: APOBEC3



(SEQ ID NO: 96)


MGPFCLGCSHRKCYSPIRNLISQETFKFHFKNLGYAKGRKDTFLCYEVTR





KDCDSPVSLHHGVFKNKDNIHAEICFLYWFHDKVLKVLSPREEFKITWYM





SWSPCFECAEQIVRFLATHHNLSLDIFSSRLYNVQDPETQQNLCRLVQEG





AQVAAMDLYEFKKCWKKFVDNGGRRFRPWKRLLTNFRYQDSKLQEILRRM





DPLSEEEFYSQFYNQRVKHLCYYHRMKPYLCYQLEQFNGQAPLKGCLLSE





KGKQHAEILFLDKIRSMELSQVTITCYLTWSPCPNCAWQLAAFKRDRPDL





ILHIYTSRLYFHWKRPFQKGLCSLWQSGILVDVMDLPQFTDCWTNFVNPK





RPFRPWKGLEIISRRTQRRLRRIKESWGLQDLVNDFGNLQLGPPMSN






Mus musculus: APOBEC3 catalytic domain



(SEQ ID NO: 97)


MGPFCLGCSHRKCYSPIRNLISQETFKFHFKNLGYAKGRKDTFLCYEVTR





KDCDSPVSLHHGVFKNKDNIHAEICFLYWFHDKVLKVLSPREEFKITWYM





SWSPCFECAEQIVRFLATHHNLSLDIFSSRLYNVQDPETQQNLCRLVQEG





AQVAAMDLYEFKKCWKKFVDNGGRRFRPWKRLLTNFRYQDSKLQEILRR






Homo sapiens: APOBEC3A



(SEQ ID NO: 98)


MEASPASGPRHLMDPHIFTSNFNNGIGRHKTYLCYEVERLDNGTSVKMDQ





HRGFLHNQAKNLLCGFYGRHAELRFLDLVPSLQLDPAQIYRVTWFISWSP





CFSWGCAGEVRAFLQENTHVRLRIFAARIYDYDPLYKEALQMLRDAGAQV





SIMTYDEFKHCWDTFVDHQGCPFQPWDGLDEHSQALSGRLRAILQNQGN






Homo sapiens: APOBEC3G



(SEQ ID NO: 99)


MKPHFRNTVERMYRDTFSYNFYNRPILSRRNTVWLCYEVKTKGPSRPPLD





AKIFRGQVYSELKYHPEMRFFHWFSKWRKLHRDQEYEVTWYISWSPCTKC





TRDMATFLAEDPKVTLTIFVARLYYFWDPDYQEALRSLCQKRDGPRATMK





IMNYDEFQHCWSKFVYSQRELFEPWNNLPKYYILLHIMLGEILRHSMDPP





TFTFNFNNEPWVRGRHETYLCYEVERMHNDTWVLLNQRRGFLCNQAPHKH





GFLEGRHAELCFLDVIPFWKLDLDQDYRVTCFTSWSPCFSCAQEMAKFIS





KNKHVSLCIFTARIYDDQGRCQEGLRTLAEAGAKISIMTYSEFKHCWDTF





VDHQGCPFQPWDGLDEHSQDLSGRLRAILQNQEN






Homo sapiens: APOBEC3G catalytic domain



(SEQ ID NO: 100)


PPTFTFNFNNEPWVRGRHETYLCYEVERMHNDTWVLLNQRRGFLCNQAPH





KHGFLEGRHAELCFLDVIPFWKLDLDQDYRVTCFTSWSPCFSCAQEMAKF





ISKNKHVSLCIFTARIYDDQGRCQEGLRTLAEAGAKISIMTYSEFKHCWD





TFVDHQGCPFQPWDGLDEHSQDLSGRLRAILQNQEN






Homo sapiens: APOBEC3H



(SEQ ID NO: 101)


MALLTAETFRLQFNNKRRLRRPYYPRKALLCYQLTPQNGSTPTRGYFENK





KKCHAEICFINEIKSMGLDETQCYQVTCYLTWSPCSSCAWELVDFIKAHD





HLNLGIFASRLYYHWCKPQQKGLRLLCGSQVPVEVMGFPKFADCWENFVD





HEKPLSFNPYKMLEELDKNSRAIKRRLERIKIPGVRAQGRYMDILCDAEV






Homo sapiens: APOBEC3F



(SEQ ID NO: 102)


MKPHFRNTVERMYRDTFSYNFYNRPILSRRNTVWLCYEVKTKGPSRPRLD





AKIFRGQVYSQPEHHAEMCFLSWFCGNQLPAYKCFQITWFVSWTPCPDCV





AKLAEFLAEHPNVTLTISAARLYYYWERDYRRALCRLSQAGARVKIMDDE





EFAYCWENFVYSEGQPFMPWYKFDDNYAFLHRTLKEILRNPMEAMYPHIF





YFHFKNLRKAYGRNESWLCFTMEWKHHSPVSWKRGVFRNQVDPETHCHAE





RCFLSWFCDDILSPNTNYEVTWYTSWSPCPECAGEVAEFLARHSNVNLTI





FTARLYYFWDTDYQEGLRSLSQEGASVEIMGYKDFKYCWENFVYNDDEPF





KPWKGLKYNFLFLDSKLQEILE






Homo sapiens: APOBEC3F catalytic domain



(SEQ ID NO: 103)


KEILRNPMEAMYPHIFYFHFKNLRKAYGRNESWLCFTMEVVKHHSPVSWK





RGVFRNQVDPETHCHAERCFLSWFCDDILSPNTNYEVTWYTSWSPCPECA





GEVAEFLARHSNVNLTIFTARLYYFWDTDYQEGLRSLSQEGASVEIMGYK





DFKYCWENFVYNDDEPFKPWKGLKYNFLFLDSKLQEILE






Escherichia coli: TadA



(SEQ ID NO: 104)


MKRTADGSEFESPKKKRKVSEVEFSHEYWMRHALTLAKRAWDEREVPVGA





VLVHNNRVIGEGWNRPIGRHDPTAHAEIMALRQGGLVMQNYRLIDATLYV





TLEPCVMCAGAMIHSRIGRVVFGARDAKTGAAGSLMDVLHHPGMNHRVEI





TEGILADECAALLSDFFRMRRQEIKAQKKAQSSTDSGGSSGGSSGSETPG





TSESATPESSGGSSGGSSEVEFSHEYWMRHALTLAKRARDEREVPVGAVL





VLNNRVIGEGWNRAIGLHDPTAHAEIMALRQGGLVMQNYRLIDATLYVTF





EPCVMCAGAMIHSRIGRVVFGVRNAKTGAAGSLMDVLHYPGMNHRVEITE





GILADECAALLCYFFRMPRQVFNAQKKAQSSTD






Homo sapiens: Adar1



(SEQ ID NO: 105)


MNPRQGYSLSGYYTHPFQGYEHRQLRYQQPGPGSSPSSFLLKQIEFLKGQ





LPEAPVIGKQTPSLPPSLPGLRPRFPVLLASSTRGRQVDIRGVPRGVHLG





SQGLQRGFQHPSPRGRSLPQRGVDCLSSHFQELSIYQDQEQRILKFLEEL





GEGKATTAHDLSGKLGTPKKEINRVLYSLAKKGKLQKEAGTPPLWKIAVS





TQAWNQHSGVVRPDGHSQGAPNSDPSLEPEDRNSTSVSEDLLEPFIAVSA





QAWNQHSGVVRPDSHSQGSPNSDPGLEPEDSNSTSALEDPLEFLDMAEIK





EKICDYLFNVSDSSALNLAKNIGLTKARDINAVLIDMERQGDVYRQGTTP





PIWHLTDKKRERMQIKRNTNSVPETAPAAIPETKRNAEFLTCNIPTSNAS





NNMVTTEKVENGQEPVIKLENRQEARPEPARLKPPVHYNGPSKAGYVDFE





NGQWATDDIPDDLNSIRAAPGEFRAIMEMPSFYSHGLPRCSPYKKLTECQ





LKNPISGLLEYAQFASQTCEFNMIEQSGPPHEPRFKFQVVINGREFPPAE





AGSKKVAKQDAAMKAMTILLEEAKAKDSGKSEESSHYSTEKESEKTAESQ





TPTPSATSFFSGKSPVTTLLECMHKLGNSCEFRLLSKEGPAHEPKFQYCV





AVGAQTFPSVSAPSKKVAKQMAAEEAMKALHGEATNSMASDNQPEGMISE





SLDNLESMMPNKVRKIGELVRYLNTNPVGGLLEYARSHGFAAEFKLVDQS





GPPHEPKFVYQAKVGGRWFPAVCAHSKKQGKQEAADAALRVLIGENEKAE





RMGFTEVTPVTGASLRRTMLLLSRSPEAQPKTLPLTGSTFHDQIAMLSHR





CFNTLTNSFQPSLLGRKILAAIIMKKDSEDMGVVVSLGTGNRCVKGDSLS





LKGETVNDCHAEIISRRGFIRFLYSELMKYNSQTAKDSIFEPAKGGEKLQ





IKKTVSFHLYISTAPCGDGALFDKSCSDRAMESTESRHYPVFENPKQGKL





RTKVENGEGTIPVESSDIVPTWDGIRLGERLRTMSCSDKILRWNVLGLQG





ALLTHFLQPIYLKSVTLGYLFSQGHLTRAICCRVTRDGSAFEDGLRHPFI





VNHPKVGRVSIYDSKRQSGKTKETSVNWCLADGYDLEILDGTRGTVDGPR





NELSRVSKKNIFLLFKKLCSFRYRRDLLRLSYGEAKKAARDYETAKNYFK





KGLKDMGYGNWISKPQEEKNFYLCPV






Homo sapiens: Adar2



(SEQ ID NO: 106)


MDIEDEENMSSSSTDVKENRNLDNVSPKDGSTPGPGEGSQLSNGGGGGPG





RKRPLEEGSNGHSKYRLKKRRKTPGPVLPKNALMQLNEIKPGLQYTLLSQ





TGPVHAPLFVMSVEVNGQVFEGSGPTKKKAKLHAAEKALRSFVQFPNASE





AHLAMGRTLSVNTDFTSDQADFPDTLFNGFETPDKAEPPFYVGSNGDDSF





SSSGDLSLSASPVPASLAQPPLPVLPPFPPPSGKNPVMILNELRPGLKYD





FLSESGESHAKSFVMSVVVDGQFFEGSGRNKKLAKARAAQSALAAIFNLH





LDQTPSRQPIPSEGLQLHLPQVLADAVSRLVLGKFGDLTDNFSSPHARRK





VLAGVVMTTGTDVKDAKVISVSTGTKCINGEYMSDRGLALNDCHAEIISR





RSLLRFLYTQLELYLNNKDDQKRSIFQKSERGGFRLKENVQFHLYISTSP





CGDARIFSPHEPILEEPADRHPNRKARGQLRTKIESGQGTIPVRSNASIQ





TWDGVLQGERLLTMSCSDKIARWNVVGIQGSLLSIFVEPIYFSSIILGSL





YHGDHLSRAMYQRISNIEDLPPLYTLNKPLLSGISNAEARQPGKAPNFSV





NWTVGDSAIEVINATTGKDELGRASRLCKHALYCRWMRVHGKVPSHLLRS





KITKPNVYHESKLAAKEYQAAKARLFTAFIKAGLGAWVEKPTEQDQFSLT





P






Streptococcus pyogenes: Cas9 Bipartite NLS



(SEQ ID NO: 107)


MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKFKVLGNTDRHSIKKNLIGA





LLFDSGETAEATRLKRTARRRYTRRKNRICYLQEIFSNEMAKVDDSFFHR





LEESFLVEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKKLVDSTDKAD





LRLIYLALAHMIKFRGHFLIEGDLNPDNSDVDKLFIQLVQTYNQLFEENP





INASGVDAKAILSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSLGLTP





NFKSNFDLAEDAKLQLSKDTYDDDLDNLLAQIGDQYADLFLAAKNLSDAI





LLSDILRVNTEITKAPLSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEI





FFDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGTEELLVKLNREDLLR





KQRTFDNGSIPHQIHLGELHAILRRQEDFYPFLKDNREKIEKILTFRIPY





YVGPLARGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSFIERMTNFDK





NLPNEKVLPKHSLLYEYFTVYNELTKVKYVTEGMRKPAFLSGEQKKAIVD





LLFKTNRKVTVKQLKEDYFKKIECFDSVEISGVEDRFNASLGTYHDLLKI





IKDKDFLDNEENEDILEDIVLTLTLFEDREMIEERLKTYAHLFDDKVMKQ





LKRRRYTGWGRLSRKLINGIRDKQSGKTILDFLKSDGFANRNFMQLIHDD





SLTFKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKGILQTVKVVDELVKV





MGRHKPENIVIEMARENQTTQKGQKNSRERMKRIEEGIKELGSQILKEHP





VENTQLQNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHIVPQSFLKDD





SIDNKVLTRSDKNRGKSDNVPSEEVVKKMKNYWRQLLNAKLITQRKFDNL





TKAERGGLSELDKAGFIKRQLVETRQITKHVAQILDSRMNTKYDENDKLI





REVKVITLKSKLVSDFRKDFQFYKVREINNYHHAHDAYLNAVVGTALIKK





YPKLESEFVYGDYKVYDVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEI





TLANGEIRKRPLIETNGETGEIVWDKGRDFATVRKVLSMPQVNIVKKTEV





QTGGFSKESILPKRNSDKLIARKKDWDPKKYGGFDSPTVAYSVLVVAKVE





KGKSKKLKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVKKDLIIKLPK





YSLFELENGRKRMLASAGELQKGNELALPSKYVNFLYLASHYEKLKGSPE





DNEQKQLFVEQHKHYLDEIIEQISEFSKRVILADANLDKVLSAYNKHRDK





PIREQAENIIHLFTLTNLGAPAAFKYFDTTIDRKRYTSTKEVLDATLIHQ





SITGLYETRIDLSQLGGDGSGGGGSGKRTADGSEFEPKKKRKVSSGGDYK





DHDGDYKDHDIDYKDDDDK






Staphylococcus aureus: Cas9



(SEQ ID NO: 108)


MKRNYILGLDIGITSVGYGIIDYETRDVIDAGVRLFKEANVENNEGRRSK





RGARRLKRRRRHRIQRVKKLLFDYNLLTDHSELSGINPYEARVKGLSQKL





SEEEFSAALLHLAKRRGVHNVNEVEEDTGNELSTKEQISRNSKALEEKYV





AELQLERLKKDGEVRGSINRFKTSDYVKEAKQLLKVQKAYHQLDQSFIDT





YIDLLETRRTYYEGPGEGSPFGWKDIKEWYEMLMGHCTYFPEELRSVKYA





YNADLYNALNDLNNLVITRDENEKLEYYEKFQIIENVFKQKKKPTLKQIA





KEILVNEEDIKGYRVTSTGKPEFTNLKVYHDIKDITARKEIIENAELLDQ





IAKILTIYQSSEDIQEELTNLNSELTQEEIEQISNLKGYTGTHNLSLKAI





NLILDELWHTNDNQIAIFNRLKLVPKKVDLSQQKEIPTTLVDDFILSPVV





KRSFIQSIKVINAIIKKYGLPNDIIIELAREKNSKDAQKMINEMQKRNRQ





TNERIEEIIRTTGKENAKYLIEKIKLHDMQEGKCLYSLEAIPLEDLLNNP





FNYEVDHIIPRSVSFDNSFNNKVLVKQEENSKKGNRTPFQYLSSSDSKIS





YETFKKHILNLAKGKGRISKTKKEYLLEERDINRFSVQKDFINRNLVDTR





YATRGLMNLLRSYFRVNNLDVKVKSINGGFTSFLRRKWKFKKERNKGYKH





HAEDALIIANADFIFKEWKKLDKAKKVMENQMFEEKQAESMPEIETEQEY





KEIFITPHQIKHIKDFKDYKYSHRVDKKPNRELINDTLYSTRKDDKGNTL





IVNNLNGLYDKDNDKLKKLINKSPEKLLMYHHDPQTYQKLKLIMEQYGDE





KNPLYKYYEETGNYLTKYSKKDNGPVIKKIKYYGNKLNAHLDITDDYPNS





RNKVVKLSLKPYRFDVYLDNGVYKFVTVKNLDVIKKENYYEVNSKCYEEA





KKLKKISNQAEFIASFYNNDLIKINGELYRVIGVNNDLLNRIEVNMIDIT





YREYLENMNDKRPPRIIKTIASKTQSIKKYSTDILGNLYEVKSKKHPQII





KKG






Campylobacter jejuni: Cas9



(SEQ ID NO: 109)


MARILAFDIGISSIGWAFSENDELKDCGVRIFTKVENPKTGESLALPRRL





ARSARKRLARRKARLNHLKHLIANEFKLNYEDYQSFDESLAKAYKGSLIS





PYELRFRALNELLSKQDFARVILHIAKRRGYDDIKNSDDKEKGAILKAIK





QNEEKLANYQSVGEYLYKEYFQKFKENSKEFTNVRNKKESYERCIAQSFL





KDELKLIFKKQREFGFSFSKKFEEEVLSVAFYKRALKDFSHLVGNCSFFT





DEKRAPKNSPLAFMFVALTRIINLLNNLKNTEGILYTKDDLNALLNEVLK





NGTLTYKQTKKLLGLSDDYEFKGEKGTYFIEFKKYKEFIKALGEHNLSQD





DLNEIAKDITLIKDEIKLKKALAKYDLNQNQIDSLSKLEFKDHLNISFKA





LKLVTPLMLEGKKYDEACNELNLKVAINEDKKDFLPAFNETYYKDEVTNP





VVLRAIKEYRKVLNALLKKYGKVHKINIELAREVGKNHSQRAKIEKEQNE





NYKAKKDAELECEKLGLKINSKNILKLRLFKEQKEFCAYSGEKIKISDLQ





DEKMLEIDHIYPYSRSFDDSYMNKVLVFTKQNQEKLNQTPFEAFGNDSAK





WQKIEVLAKNLPTKKQKRILDKNYKDKEQKNFKDRNLNDTRYIARLVLNY





TKDYLDFLPLSDDENTKLNDTQKGSKVHVEAKSGMLTSALRHTWGFSAKD





RNNHLHHAIDAVIIAYANNSIVKAFSDFKKEQESNSAELYAKKISELDYK





NKRKFFEPFSGFRQKVLDKIDEIFVSKPERKKPSGALHEETFRKEEEFYQ





SYGGKEGVLKALELGKIRKVNGKIVKNGDMFRVDIFKHKKTNKFYAVPIY





TMDFALKVLPNKAVARSKKGEIKDWILMDENYEFCFSLYKDSLILIQTKD





MQEPEFVYYNAFTSSTVSLIVSKHDNKFETLSKNQKILFKNANEKEVIAK





SIGIQNLKVFEKYIVSALGEVTKAEFRQREDFKK






Neisseria meningitidis: Cas9



(SEQ ID NO: 110)


MAAFKPNSINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAE





VPKTGDSLAMARRLARSVRRLTRRRAHRLLRTRRLLKREGVLQAANFDEN





GLIKSLPNTPWQLRAAALDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGET





ADKELGALLKGVAGNAHALQTGDFRTPAELALNKFEKESGHIRNQRSDYS





HTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQRPALSGDA





VQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDT





ERATLMDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEM





KAYHAISRALEKEGLKDKKSPLNLSPELQDEIGTAFSLFKTDEDITGRLK





DRIQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRYDEACAEIYG





DHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPAR





IHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKS





KDILKLRLYEQQHGKCLYSGKEINLGRLNEKGYVEIDHALPFSRTWDDSF





NNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQ





RILLQKFDEDGFKERNLNDTRYVNRFLCQFVADRMRLTGKGKKRVFASNG





QITNLLRGFWGLRKVRAENDRHHALDAWVACSTVAMQQKITRFVRYKEMN





AFDGKTIDKETGEVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEAD





TLEKLRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGQGHMETVKSAK





RLDEGVSVLRVPLTQLKLKDLEKMVNREREPKLYEALKARLEAHKDDPAK





AFAEPFYKYDKAGNRTQQVKAVRVEQVQKTGVWVRNHNGIADNATMVRVD





VFEKGDKYYLVPIYSWQVAKGILPDRAVVQGKDEEDWQLIDDSFNFKFSL





HPNDLVEVITKKARMFGYFASCHRGTGNINIRIHDLDHKIGKNGILEGIG





VKTALSFQKYQIDELGKEIRPCRLKKRPPVR






Acidaminococcus sp. Cas12a



(SEQ ID NO: 111)


MTQFEGFTNLYQVSKTLRFELIPQGKTLKHIQEQGFIEEDKARNDHYKEL





KPIIDRIYKTYADQCLQLVQLDWENLSAAIDSYRKEKTEETRNALIEEQA





TYRNAIHDYFIGRTDNLTDAINKRHAEIYKGLFKAELFNGKVLKQLGTVT





TTEHENALLRSFDKFTTYFSGFYENRKNVFSAEDISTAIPHRIVQDNFPK





FKENCHIFTRLITAVPSLREHFENVKKAIGIFVSTSIEEVFSFPFYNQLL





TQTQIDLYNQLLGGISREAGTEKIKGLNEVLNLAIQKNDETAHIIASLPH





RFIPLFKQILSDRNTLSFILEEFKSDEEVIQSFCKYKTLLRNENVLETAE





ALFNELNSIDLTHIFISHKKLETISSALCDHWDTLRNALYERRISELTGK





ITKSAKEKVQRSLKHEDINLQEIISAAGKELSEAFKQKTSEILSHAHAAL





DQPLPTTLKKQEEKEILKSQLDSLLGLYHLLDWFAVDESNEVDPEFSARL





TGIKLEMEPSLSFYNKARNYATKKPYSVEKFKLNFQMPTLASGWDVNKEK





NNGAILFVKNGLYYLGIMPKQKGRYKALSFEPTEKTSEGFDKMYYDYFPD





AAKMIPKCSTQLKAVTAHFQTHTTPILLSNNFIEPLEITKEIYDLNNPEK





EPKKFQTAYAKKTGDQKGYREALCKWIDFTRDFLSKYTKTTSIDLSSLRP





SSQYKDLGEYYAELNPLLYHISFQRIAEKEIMDAVETGKLYLFQIYNKDF





AKGHHGKPNLHTLYWTGLFSPENLAKTSIKLNGQAELFYRPKSRMKRMAH





RLGEKMLNKKLKDQKTPIPDTLYQELYDYVNHRLSHDLSDEARALLPNVI





TKEVSHEIIKDRRFTSDKFFFHVPITLNYQAANSPSKFNQRVNAYLKEHP





ETPIIGIDRGERNLIYITVIDSTGKILEQRSLNTIQQFDYQKKLDNREKE





RVAARQAWSWVGTIKDLKQGYLSQVIHEIVDLMIHYQAVVVLENLNFGFK





SKRTGIAEKAVYQQFEKMLIDKLNCLVLKDYPAEKVGGVLNPYQLTDQFT





SFAKMGTQSGFLFYVPAPYTSKIDPLTGFVDPFVWKTIKNHESRKHFLEG





FDFLHYDVKTGDFILHFKMNRNLSFQRGLPGFMPAWDIVFEKNETQFDAK





GTPFIAGKRIVPVIENHRFTGRYRDLYPANELIALLEEKGIVFRDGSNIL





PKLLENDDSHAIDTMVALIRSVLQMRNSNAATGEDYINSPVRDLNGVCFD





SRFQNPEWPMDADANGAYHIALKGQLLLNHLKESKDLKLQNGISNQDWLA





YIQELRN






Lachnospiraceae bacterium Cas12a:



(SEQ ID NO: 112)


MSKLEKFTNCYSLSKTLRFKAIPVGKTQENIDNKRLLVEDEKRAEDYKGV





KKLLDRYYLSFINDVLHSIKLKNLNNYISLFRKKTRTEKENKELENLEIN





LRKEIAKAFKGNEGYKSLFKKDIIETILPEFLDDKDEIALVNSFNGFTTA





FTGFFDNRENMFSEEAKSTSIAFRCINENLTRYISNMDIFEKVDAIFDKH





EVQEIKEKILNSDYDVEDFFEGEFFNFVLTQEGIDVYNAIIGGFVTESGE





KIKGLNEYINLYNQKTKQKLPKFKPLYKQVLSDRESLSFYGEGYTSDEEV





LEVFRNTLNKNSEIFSSIKKLEKLFKNFDEYSSAGIFVKNGPAISTISKD





IFGEWNVIRDKWNAEYDDIHLKKKAVVTEKYEDDRRKSFKKIGSFSLEQL





QEYADADLSVVEKLKEIIIQKVDEIYKVYGSSEKLFDADFVLEKSLKKND





AVVAIMKDLLDSVKSFENYIKAFFGEGKETNRDESFYGDFVLAYDILLKV





DHIYDAIRNYVTQKPYSKDKFKLYFQNPQFMGGWDKDKETDYRATILRYG





SKYYLAIMDKKYAKCLQKIDKDDVNGNYEKINYKLLPGPNKMLPKVFFSK





KWMAYYNPSEDIQKIYKNGTFKKGDMFNLNDCHKLIDFFKDSISRYPKWS





NAYDFNFSETEKYKDIAGFYREVEEQGYKVSFESASKKEVDKLVEEGKLY





MFQIYNKDFSDKSHGTPNLHTMYFKLLFDENNHGQIRLSGGAELFMRRAS





LKKEELVVHPANSPIANKNPDNPKKTTTLSYDVYKDKRFSEDQYELHIPI





AINKCPKNIFKINTEVRVLLKHDDNPYVIGIDRGERNLLYIVVVDGKGNI





VEQYSLNEIINNFNGIRIKTDYHSLLDKKEKERFEARQNWTSIENIKELK





AGYISQWVHKICELVEKYDAVIALEDLNSGFKNSRVKVEKQVYQKFEKML





IDKLNYMVDKKSNPCATGGALKGYQITNKFESFKSMSTQNGFIFYIPAWL





TSKIDPSTGFVNLLKTKYTSIADSKKFISSFDRIMYVPEEDLFEFALDYK





NFSRTDADYIKKWKLYSYGNRIRIFRNPKKNNVFDWEEVCLTSAYKELFN





KYGINYQQGDIRALLCEQSDKAFYSSFMALMSLMLQMRNSITGRTDVDFL





ISPVKNSDGIFYDSRNYEAQENAILPKNADANGAYNIARKVLWAIGQFKK





AEDEKLDKVKIAISNKEWLEYAQTSVKH






Leptotrichia shahii Cas13a



(SEQ ID NO: 113)


MGNLFGHKRWYEVRDKKDFKIKRKVKVKRNYDGNKYILNINENNNKEKID





NNKFIRKYINYKKNDNILKEFTRKFHAGNILFKLKGKEGIIRIENNDDFL





ETEEVVLYIEAYGKSEKLKALGITKKKIIDEAIRQGITKDDKKIEIKRQE





NEEEIEIDIRDEYTNKTLNDCSIILRIIENDELETKKSIYEIFKNINMSL





YKIIEKIIENETEKVFENRYYEEHLREKLLKDDKIDVILTNFMEIREKIK





SNLEILGFVKFYLNVGGDKKKSKNKKMLVEKILNINVDLTVEDIADFVIK





ELEFWNITKRIEKVKKVNNEFLEKRRNRTYIKSYVLLDKHEKFKIERENK





KDKIVKFFVENIKNNSIKEKIEKILAEFKIDELIKKLEKELKKGNCDTEI





FGIFKKHYKVNFDSKKFSKKSDEEKELYKIIYRYLKGRIEKILVNEQKVR





LKKMEKIEIEKILNESILSEKILKRVKQYTLEHIMYLGKLRHNDIDMTTV





NTDDFSRLHAKEELDLELITFFASTNMELNKIFSRENINNDENIDFFGGD





REKNYVLDKKILNSKIKIIRDLDFIDNKNNITNNFIRKFTKIGTNERNRI





LHAISKERDLQGTQDDYNKVINIIQNLKISDEEVSKALNLDVVFKDKKNI





ITKINDIKISEENNNDIKYLPSFSKVLPEILNLYRNNPKNEPFDTIETEK





IVLNALIYVNKELYKKLILEDDLEENESKNIFLQELKKTLGNIDEIDENI





IENYYKNAQISASKGNNKAIKKYQKKVIECYIGYLRKNYEELFDFSDFKM





NIQEIKKQIKDINDNKTYERITVKTSDKTIVINDDFEYIISIFALLNSNA





VINKIRNRFFATSVWLNTSEYQNIIDILDEIMQLNTLRNECITENWNLNL





EEFIQKMKEIEKDFDDFKIQTKKEIFNNYYEDIKNNILTEFKDDINGCDV





LEKKLEKIVIFDDETKFEIDKKSNILQDEQRKLSNINKKDLKKKVDQYIK





DKDQEIKSKILCRIIFNSDFLKKYKKEIDNLIEDMESENENKFQEIYYPK





ERKNELYIYKKNLFLNIGNPNFDKIYGLISNDIKMADAKFLFNIDGKNIR





KNKISEIDAILKNLNDKLNGYSKEYKEKYIKKLKENDDFFAKNIQNKNYK





SFEKDYNRVSEYKKIRDLVEFNYLNKIESYLIDINWKLAIQMARFERDMH





YIVNGLRELGIIKLSGYNTGISRAYPKRNGSDGFYTTTAYYKFFDEESYK





KFEKICYGFGIDLSENSEINKPENESIRNYISHFYIVRNPFADYSIAEQI





DRVSNLLSYSTRYNNSTYASVFEVFKKDVNLDYDELKKKFKLIGNNDILE





RLMKPKKVSVLELESYNSDYIKNLIIELLTKIENTNDTL






Leptotrichia wadei Cas13a



(SEQ ID NO: 114)


MKVTKVDGISHKKYIEEGKLVKSTSEENRTSERLSELLSIRLDIYIKNPD





NASEEENRIRRENLKKFFSNKVLHLKDSVLYLKNRKEKNAVQDKNYSEED





ISEYDLKNKNSFSVLKKILLNEDVNSEELEIFRKDVEAKLNKINSLKYSF





EENKANYQKINENNVEKVGGKSKRNIIYDYYRESAKRNDYINNVQEAFDK





LYKKEDIEKLFFLIENSKKHEKYKIREYYHKIIGRKNDKENFAKIIYEEI





QNVNNIKELIEKIPDMSELKKSQVFYKYYLDKEELNDKNIKYAFCHFVEI





EMSQLLKNYVYKRLSNISNDKIKRIFEYQNLKKLIENKLLNKLDTYVRNC





GKYNYYLQVGEIATSDFIARNRQNEAFLRNIIGVSSVAYFSLRNILETEN





ENGITGRMRGKTVKNNKGEEKYVSGEVDKIYNENKQNEVKENLKMFYSYD





FNMDNKNEIEDFFANIDEAISSIRHGIVHFNLELEGKDIFAFKNIAPSEI





SKKMFQNEINEKKLKLKIFKQLNSANVFNYYEKDVIIKYLKNTKFNFVNK





NIPFVPSFTKLYNKIEDLRNTLKFFWSVPKDKEEKDAQIYLLKNIYYGEF





LNKFVKNSKVFFKITNEVIKINKQRNQKTGHYKYQKFENIEKTVPVEYLA





IIQSREMINNQDKEEKNTYIDFIQQIFLKGFIDYLNKNNLKYIESNNNND





NNDIFSKIKIKKDNKEKYDKILKNYEKHNRNKEIPHEINEFVREIKLGKI





LKYTENLNMFYLILKLLNHKELTNLKGSLEKYQSANKEETFSDELELINL





LNLDNNRVTEDFELEANEIGKFLDFNENKIKDRKELKKFDTNKIYFDGEN





IIKHRAFYNIKKYGMLNLLEKIADKAKYKISLKELKEYSNKKNEIEKNYT





MQQNLHRKYARPKKDEKFNDEDYKEYEKAIGNIQKYTHLKNKVEFNELNL





LQGLLLKILHRLVGYTSIWERDLRFRLKGEFPENHYIEEIFNFDNSKNVK





YKSGQIVEKYINFYKELYKDNVEKRSIYSDKKVKKLKQEKKDLYIRNYIA





HFNYIPHAEISLLEVLENLRKLLSYDRKLKNAIMKSIVDILKEYGFVATF





KIGADKKIEIQTLESEKIVHLKNLKKKKLMTDRNSEELCELVKVMFEYKA





LE





Pleckstrin homology domain of Human ARNO:


(SEQ ID NO: 115)


NPDREGWLLKLGGGRVKTWKRRWFILTDNCLYYFEYTTDKEPRGIIPLEN





LSIREVDDPRKPNCFELYIPNNKGQLIKACKTEADGRVVEGNHMVYRISA





PTQEEKDEWIKSIQAAVS





Pleckstrin homology domain of Human ARNO


R279C:


(SEQ ID NO: 116)


NPDREGWLLKLGGGRVKTWKCRWFILTDNCLYYFEYTTDKEPRGIIPLEN





LSIREVDDPRKPNCFELYIPNNKGQLIKACKTEADGRVVEGNHMVYRISA





PTQEEKDEWIKSIQAAVS





FYVE domain of Human EEA1:


(SEQ ID NO: 117)


DNEVQNCMACGKGFSVTVRRHHCRQCGNIFCAECSAKNALTPSSKKPVRV





CDACFNDLQ





FYVE domain of Human EEA1 R1375L:


(SEQ ID NO: 118)


DNEVQNCMACGKGFSVTVRRHHCLQCGNIFCAECSAKNALTPSSKKPVRV





CDACFNDLQ





PX domain of p40phox (NCF4):


(SEQ ID NO: 119)


DVAISANIADIEEKRGFTSHFVFVIEVKTKGGSKYLIYRRYRQFHALQSK





LEERFGPDSKSSALACTLPTLPAKVYVGVKQEIAEMRIPALNAYMKSLLS





LPVWVLMDEDVRIFFYQSPYDS





PX domain of p40phox (NCF4) R58L:


(SEQ ID NO: 120)


DVAISANIADIEEKRGFTSHFVFVIEVKTKGGSKYLIYLRYRQFHALQSK





LEERFGPDSKSSALACTLPTLPAKVYVGVKQEIAEMRIPALNAYMKSLLS





LPVWVLMDEDVRIFFYQSPYDS





Pleckstrin homology domain of



Homo sapiens DAPP1



(SEQ ID NO: 121)


MQTGRTEDDLVPTAPSLGTKEGYLTKQGGLVKTWKTRWFTLHRNELKYFK





DQMSPEPIRILDLTECSAVQFDYSQERVNCFCLVFPFRTFYLCAKTGVEA





DEWIKILRWKLSQIRKQLNQGEGTIR





Pleckstrin homology domain of



Homo sapiens GRP1 (CYTH3)



(SEQ ID NO: 122)


PFKIPEDDGNDLTHTFFNPDREGWLLKLGGRVKTWKRRWFILTDNCLYYF





EYTTDKEPRGIIPLENLSIREVEDPRKPNCFELYNPSHKGQVIKACKTEA





DGRVVEGNHVVYRISAPSPEEKEEWMKSIKASISRDPFYDMLATRKRRIA





NKK





Pleckstrin homology domain of



Homo sapiens GRP1 (CYTH3) R284C:



(SEQ ID NO: 123)


MPFKIPEDDGNDLTHTFFNPDREGWLLKLGGRVKTWKCRWFILTDNCLYY





FEYTTDKEPRGIIPLENLSIREVEDPRKPNCFELYNPSHKGQVIKACKTE





ADGRWVEGNHVVYRISAPSPEEKEEWMKSIKASISRDPFYDMLATRKRRI





ANKK





Pleckstrin homology domain of Human OSBP1


(SEQ ID NO: 124)


MGSGSAREGWLFKWTNYIKGYQRRWFVLSNGLLSYYRSKAEMRHTCRGTI





NLATANITVEDSCNFIISNGGAQTYHLKASSEVERQRWVTALELAKAKAV





K





Pleckstrin homology domain of Human OSBP1


R108E:


(SEQ ID NO: 125)


MGSGSAREGWLFKWTNYIKGYQERWFVLSNGLLSYYRSKAEMRHTCRGTI





NLATANITVEDSCNFIISNGGAQTYHLKASSEVERQRWVTALELAKAKAV





K





Pleckstrin homology domain of Human BTK1


(SEQ ID NO: 126)


MAAVILESIFLKRSQQKKKTSPLNFKKRLFLLTVHKLSYYEYDFERGRRG





SKKGSIDVEKITCVETWPEKNPPPERQIPRRGEESSEMEQISIIERFPYP





FQVVYDEGPLYVFSPTEELRKRWIHQLKNVIRYNSDLVQKYHPCFWIDGQ





YLCCSQTAKNAMGCQILENRNGSLKP





Pleckstrin homology domain of Human BTK1


R28C:


(SEQ ID NO: 127)


MAAVILESIFLKRSQQKKKTSPLNFKKCLFLLTVHKLSYYEYDFERGRRG





SKKGSIDVEKITCVETWPEKNPPPERQIPRRGEESSEMEQISIIERFPYP





FQVVYDEGPLYVFSPTEELRKRWIHQLKNVIRYNSDLVQKYHPCFWIDGQ





YLCCSQTAKNAMGCQILENRNGSLKP





Pleckstrin homology domain of Human FAPP1


(SEQ ID NO: 128)


MEGVLYKWTNYLTGWQPRWFVLDNGILSYYDSQDDVCKGSKGSIKMAVCE





IKVHSADNTRMELIIPGEQHFYMKAVNAAERQRWLVALGSSKACLTDT





Pleckstrin homology domain of Human CERT


(SEQ ID NO: 129)


PVERCGVLSKWTNYIHGWQDRWVVLKNNALSYYKSEDETEYGCRGSICLS





KAVITPHDFDECRFDISVNDSVWYLRAQDPDHRQQWIDAIEQHKT





Pleckstrin homology domain of Human PHLPP1


(SEQ ID NO: 130)


MRIQLSGMYNVRKGKMQLPVNRWTRRQVILCGTCLIVSSVKDSLTGKMHV





LPLIGGKVEEVKKHQHCLAFSSSGPQSQTYYICFDTFTEYLRWLRQVSKV





AS





Pleckstrin homology domain of Human SWAP70


(SEQ ID NO: 131)


MDVLKQGYMMKKGHRRKNWTERWFVLKPNIISYYVSEDLKDKKGDILLDE





NCCVESLPDKDGKKCLFLVKCFDKTFEISASDKKKKQEWIQAIHSTIH





Pleckstrin homology domain of Human SWAP70


R223E, R224E:


(SEQ ID NO: 132)


MDVLKQGYMMKKGHEEKNWTERWFVLKPNIISYYVSEDLKDKKGDILLDE





NCCVESLPDKDGKKCLFLVKCFDKTFEISASDKKKKQEWIQAIHSTIH





Pleckstrin homology domain of Human MAPKAP1


(SEQ ID NO: 133)


MDMLSSHHYKSFKVSMIHRLRFTTDVQLGISGDKVEIDPVTNQKASTKFW





IKQKPISIDSDLLCACDLAEEKSPSHAIFKLTYLSNHDYKHLYFESDAAT





VNEIVLKVNYILES





Pleckstrin Homology Domain of Human PKD


(SEQ ID NO: 134)


MGTVMKEGWMVHYTSKDTLRKRHYWRLDSKCITLFQNDTGSRYYKEIPLS





EILSLEPVKTSALIPNGANPHCFEITTANVVYYVGENVVNPSSPSPNNSV





LTSGVGADVARMWEIAIQHALM





Pleckstrin homology domain of Human Son


Of Sevenless Homolog 2


(SEQ ID NO: 135)


FIMEGPLTRIGAKHERHIFLFDGLMISCKPNHGQTRLPGYSSAEYRLKEK





FVMRKIQICDKEDTCEHKHAFELVSKDENSIIFAAKSAEEKNNWMAALIS





LHYRS





Pleckstrin homology domain of Human Dynamin


(SEQ ID NO: 136)


QGTNLPPSRQIVIRKGWLTISNIGIMKGGSKGYWFVLTAESLSWYKDDEE





KEKKYMLPLDNLKVRDVEKSFMSSKHIFALFNTEQRNVYKDYRFLELACD





SQEDVDS





Pleckstrin homology domain of Human BCR


(SEQ ID NO: 137)


QLLKDSFMVELVEGARKLRHVFLFTDLLLCTKLKKQSGGKTQQYDCKWYI





PLTDLSFQMVDELEAVPNIPLVPDEELDALKIKISQIKNDIQREKRANKG





SKATERLKKKLSEQESLLLLMSPSMAFRVHSRNGKSYTFLISSDYERAEW





RENIREQQK





Pleckstrin homology domain of Human DBS


(SEQ ID NO: 138)


KLLMQGSFSVWTDHKRGHTKVKELARFKPMQRHLFLHEKAVLFCKKREEN





GEGYEKAPSYSYKQSLNMAAVGITENVKGDAKKFEIWYNAREEVYIVQAP





TPEIKAAWVNEIRKVLT





Pleckstrin homology domain of Homo sapiens


phospholipase Cδ1 (hPLCδ1)


(SEQ ID NO: 139)


MDSGRDFLTLHGLQDDEDLQALLKGSQLLKVKSSSWRRERFYKLQEDCKT





IWQESRKVMRTPESQLFSIEDIQEVRMGHRTEGLEKFARDVPEDRCFSIV





FKDQRNTLDLIAPSPADAQHWVLGLHKIIHHSGSMDQRQKLQHWIHSCLR





KADKNKDNKMSFKELQNFLKELNIQ





Pleckstrin homology domain of Homo sapiens


phospholipase Cδ1 (hPLCδ1) R40L:


(SEQ ID NO: 140)


MDSGRDFLTLHGLQDDEDLQALLKGSQLLKVKSTSWRRELFYKLQEDCKT





IWQESRKVMRTPESQLFSIEDIQEVRMGHRTEGLEKFARDVPEDRCFSIV





FKDQRNTLDLIAPSPADAQHWVLGLHKIIHHSGSMDQRQKLQHWIHSCLR





KADKNKDNKMSFKELQNFLK





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT)


(SEQ ID NO: 141)


MSDVAIVKEGWLHKRGEYIKTWRPRYFLLKNDGTFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) E17K:


(SEQ ID NO: 142)


MSDVAIVKEGWLHKRGKYIKTWRPRYFLLKNDGTFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) K14R:


(SEQ ID NO: 143)


MSDVAIVKEGWLHRRGEYIKTWRPRYFLLKNDGTFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) K8R:


(SEQ ID NO: 144)


MSDVAIVREGWLHKRGEYIKTWRPRYFLLKNDGTFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) T72A:


(SEQ ID NO: 145)


MSDVAIVKEGWLHKRGEYIKTWRPRYFLLKNDGTFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNAFIIRCLQWTTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) T92A:


(SEQ ID NO: 146)


MSDVAIVKEGWLHKRGEYIKTWRPRYFLLKNDGTFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVEAPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) R25C:


(SEQ ID NO: 147)


MSDVAIVKEGWLHKRGEYIKTWRPCYFLLKNDGTFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) T34D:


(SEQ ID NO: 148)


MSDVAIVKEGWLHKRGEYIKTWRPRYFLLKNDGDFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) T34F:


(SEQ ID NO: 149)


MSDVAIVKEGWLHKRGEYIKTWRPRYFLLKNDGFFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) T34L:


(SEQ ID NO: 150)


MSDVAIVKEGWLHKRGEYIKTWRPRYFLLKNDGLFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) T81Y:


(SEQ ID NO: 151)


MSDVAIVKEGWLHKRGEYIKTWRPRYFLLKNDGTFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWYTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) K142A, H143A, R144A:


(SEQ ID NO: 152)


MSDVAIVKEGWLHKRGEYIKTWRPRYFLLKNDGTFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWT





TAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPAAAVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


AKT1 (hAKT) T101C:


(SEQ ID NO: 153)


MSDVAIVKEGWLHKRGEYIKTWRPRYFLLKNDGTFIGYKERPQDVDQREA





PLNNFSVAQCQLMKTERPRPNTFIIRCLQWTTVIERTFHVETPEEREEWT





CAIQTVADGLKKQEEEEMDFRSGSPSDNSGAEEMEVSLAKPKHRVTMNEF





EYLKLLGKGTFGKVDPPV





Pleckstrin homology domain of Homo sapiens


PDPK1 (hPDPK1)


(SEQ ID NO: 154)


KMGPVDKRKGLFARRRQLLLTEGPHLYYVDPVNKVLKGEIPWSQELRPEA





KNFKTFFVHTPNRTYYLMDPSGNAHKWCRKIQEVWRQRYQSH





MS2 (RNA Binding protein):


(SEQ ID NO: 155)


MASNFTQFVLVDNGGTGDVTVAPSNFANGIAEWISSNSRSQAYKVTCSVR





QSSAQKRKYTIKVEVPKVATQTVGGVELPVAAWRSYLNMELTIPIFATNS





DCELIVKAMQGLLKDGNPIPSAIAANSGIY





COM (RNA Binding protein):


(SEQ ID NO: 156)


MKSIRCKNONKLLFKADSFDHIEIRCPRCKRHIIMLNACEHPTEKHCGKR





EKITHSDETVRY





PP7 (RNA Binding protein):


(SEQ ID NO: 157)


MAKTIVLAVGEATRTLTEIQSTADRQIFEEKVGPLVGRLRLTASLRQNGA





KTAYRVNLKLDQADVVDASTSVAGELPKVRYTQVWSHDVTIVANSTEASR





KSLYDLTKSLVATSQVEDLVVNLVPLGRSLE





TBP (RNA Binding protein):


(SEQ ID NO: 158)


MAVPETRPNHTIYINNLNSKIKKDELKKSLYAIFSQFGQILDILVPRQRT





PRGQAFVIFKEVSSATNALRSMQGFPFYDKPMRIQYAKTDKRIPAKMKGT





FV





Human SLBP (RNA Binding protein):


(SEQ ID NO: 159)


MADFETDESVLMRRQKQINYGKNTIAYDRYIKEVPRHLRQPGIHPKTPNK





FKKYSRRSWDQQIKLWKVALHFWD





Herpes simplex virus (HSV) type 1:


VP16 Transcription Activation Domain


(SEQ ID NO: 160)


PTDALDDFDLDMLPADALDDFDLDMLPADALDDFDLDM





Herpes simplex virus (HSV) type 1 &


Synthetic: VP64


(SEQ ID NO: 161)


GRADALDDFDLDMLGSDALDDFDLDMLGSDALDDFDLDMLGSDALDDFDL





DML






Homo sapiens: P65



(SEQ ID NO: 162)


SQYLPDTDDRHRIEEKRKRTYETFKSIMKKSPFSGPTDPRPPPRRIAVPS





RSSASVPKPAPQPYPFTSSLSTINYDEFPTMVFPSGQISQASALAPAPPQ





VLPQAPAPAPAPAMVSALAQAPAPVPVLAPGPPQAVAPPAPKPTQAGEGT





LSEALLQLQFDDEDLGALLGNSTDPAVFTDLASVDNSEFQQLLNQGIPVA





PHTTEPMLMEYPEAITRLVTGAQRPPDPAPAPLGAPGLPNGLLSGDEDFS





SIADMDFSALL





Kaposi's Sarcoma-Associated Herpesvirus


Transactivator: RTA


(SEQ ID NO: 163)


RDSREGMFLPKPEAGSAISDVFEGREVCQPKRIRPFHPPGSPWANRPLPA





SLAPTPTGPVHEPVGSLTPAPVPQPLDPAPAVTPEASHLLEDPDEETSQA





VKALREMADTVIPQKEEAAICGQMDLSHPPPRGHLDELTTTLESMTEDLN





LDSPLTPELNEILDTFLNDECLLHAMHISTGLSIFDTSLF






Homo sapiens: KRAB



(SEQ ID NO: 164)


MDAKSLTAWSRTLVTFKDVFVDFTREEWKLLDTAQQIVYRNVMLENYKNL





VSLGYQLTKPDVILRLEKGEEP






Homo sapiens: MeCP2



(SEQ ID NO: 165)


EASVQVKRVLEKSPGKLLVKMPFQASPGGKGEGGGATTSAQVMVIKRPGR





KRKAEADPQAIPKKRGRKPGSVVAAAAAEAKKKAVKESSIRSVQETVLPI





KKRKTRETVSIEVKEVVKPLLVSTLGEKSGKGLKTCKSPGRKSKESSPKG





RSSSASSPPKKEHHHHHHHAESPKAPMPLLPPPPPPEPQSSEDPISPPEP





QDLSSSICKEEKMPRAGSLESDGCPKEPAKTQPMVAAAATTTTTTTTTVA





EKYKHRGEGERKDIVSSSMPRPNREEPVDSRTPVTERVS






Homo sapiens: TET1



(SEQ ID NO: 166)


LPTCSCLDRVIQKDKGPYYTHLGAGPSVAAVREIMENRYGQKGNAIRIEI





VVYTGKEGKSSHGCPIAKWVLRRSSDEEKVLCLVRQRTGHHCPTAVMVVL





IMVWDGIPLPMADRLYTELTENLKSYNGHPTDRRCTLNENRTCTCQGIDP





ETCGASFSFGCSWSMYFNGCKFGRSPSPRRFRIDPSSPLHEKNLEDNLQS





LATRLAPIYKQYAPVAYQNQVEYENVARECRLGSKEGRPFSGVTACLDFC





AHPHRDIHNMNNGSTVVCTLTREDNRSLGVIPQDEQLHVLPLYKLSDTDE





FGSKEGMEAKIKSGAIEVLAPRRKKRTCFTQPVPRSGKKRAAMMTEVLAH





KIRAVEKKPIPRIKRKNNSTTTNNSKPSSLPTLGSNTETVQPEVKSETEP





HFILKSSDNTKTYSLMPSAPHPVKEASPGFSWSPKTASATPAPLKNDATA





SCGFSERSSTPHCTMPSGRLSGANAAAADGPGISQLGEVAPLPTLSAPVM





EPLINSEPSTGVTEPLTPHQPNHQPSFLTSPQDLASSPMEEDEQHSEADE





PPSDEPLSDDPLSPAEEKLPHIDEYWSDSEHIFLDANIGGVAIAPAHGSV





LIECARRELHATTPVEHPNRNHPTRLSLVFYQHKNLNKPQHGFELNKIKF





EAKEAKNKKMKASEQKDQAANEGPEQSSEVNELNQIPSHKALTLTHDNVV





TVSPYALTHVAGPYNHWV






Homo sapiens: DNMT3A



(SEQ ID NO: 167)


MPAMPSSGPGDTSSSAAEREEDRKDGEEQEEPRGKEERQEPSTTARKVGR





PGRKRKHPPVESGDTPKDPAVISKSPSMAQDSGASELLPNGDLEKRSEPQ





PEEGSPAGGQKGGAPAEGEGAAETLPEASRAVENGCCTPKEGRGAPAEAG





KEQKETNIESMKMEGSRGRLRGGLGWESSLRQRPMPRLTFQAGDPYYISK





RKRDEWLARWKREAEKKAKVIAGMNAVEENQGPGESQKVEEASPPAVQQP





TDPASPTVATTPEPVGSDAGDKNATKAGDDEPEYEDGRGFGIGELVWGKL





RGFSWWPGRIVSWWMTGRSRAAEGTRWVMWFGDGKFSVVCVEKLMPLSSF





CSAFHQATYNKQPMYRKAIYEVLQVASSRAGKLFPVCHDSDESDTAKAVE





VQNKPMIEWALGGFQPSGPKGLEPPEEEKNPYKEVYTDMWVEPEAAAYAP





PPPAKKPRKSTAEKPKVKEIIDERTRERLVYEVRQKCRNIEDICISCGSL





NVTLEHPLFVGGMCQNCKNCFLECAYQYDDDGYQSYCTICCGGREVLMCG





NNNCCRCFCVECVDLLVGPGAAQAAIKEDPWNCYMCGHKGTYGLLRRRED





WPSRLQMFFANNHDQEFDPPKVYPPVPAEKRKPIRVLSLFDGIATGLLVL





KDLGIQVDRYIASEVCEDSITVGMVRHQGKIMYVGDVRSVTQKHIQEWGP





FDLVIGGSPCNDLSIVNPARKGLYEGTGRLFFEFYRLLHDARPKEGDDRP





FFWLFENVVAMGVSDKRDISRFLESNPVMIDAKEVSAAHRARYFWGNLPG





MNRPLASTVNDKLELQECLEHGRIAKFSKVRTITTRSNSIKQGKDQHFPV





FMNEKEDILWCTEMERVFGFPVHYTDVSNMSRLARQRLLGRSWSVPVIRH





LFAPLKEYFACV





BaEVTRless


(SEQ ID NO: 168)


MGFTTKIIFLYNLVLVYAGFDDPRKAIELVQKRYGRPCDCSGGQVSEPPS





DRVSQVTCSGKTAYLMPDQRWKCKSIPKDTSPSGPLQECPCNSYQSSVHS





SCYTSYQQCRSGNKTYYTATLLKTQTGGTSDVQVLGSTNKLIQSPCNGIK





GQSICWSTTAPIHVSDGGGPLDTTRIKSVQRKLEEIHKALYPELQYHPLA





IPKVRDNLMVDAQTLNILNATYNLLLMSNTSLVDDCWLCLKLGPPTPLAI





PNFLLSYVTRSSDNISCLIIPPLLVQPMQFSNSSCLFSPSYNSTEEIDLG





HVAFSNCTSITNVTGPICAVNGSVFLCGNNMAYTYLPTNWTGLCVLATLL





PDIDIIPGDEPVPIPAIDHFIYRPKRAIQFIPLLAGLGITAAFTTGATGL





GVSVTQYTKLSNQLISDVQILSSTIQDLQDQVDSLAEVVLQNRRGLDLLT





AEQGGICLALQEKCCFYVNKSGIVRDKIKTLQEELERRRKDLASNPLWTG





LQGLLPYLLPFLGPLLTLLLLLTIGPCIFNRLTAFINDKLNIIHAM






EXAMPLES

The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.


Example 1. Minimal Virus-Like Particles Deliver Gene Editing Cargo to Target Cells
Methods

mVLP particles were produced in HEK293T cells by using polyethylenimine (PEI) to transfect plasmids into these cells. PEI is Polyethylenimine 25 kD linear (Polysciences #23966-2). To make a stock ‘PEI MAX’ solution, Ig of PEI was added to 1 L endotoxin-free dH2O that was previously heated to ˜80° C. and cooled to room temperature. This mixture was neutralized to pH 7.1 by addition of 10N NaOH and filter sterilized with 0.22 m polyethersulfone (PES). PEI MAX was stored at −20° C.


HEK293T cells were split to reach a confluency of 70%-90% at time of transfection and are cultured in 10% FBS DMEM media. Plasmid vector encoding cargo, e.g., one encoding a CMV promoter driving expression of a hPLCS1 PH domain or another PH domain as shown herein fused to codon optimized Cas9 were co-transfected with a plasmids encoding a U6 promoter driving expression of a sgRNA and the VSV-G envelope plasmid. Transfection reactions were assembled in reduced serum media (Opti-MEM; GIBCO #31985-070). For mVLP particle production on 10 cm plates, 7.5 μg PH-Cas9 expressing plasmid, 7.5 μg sgRNA-expression plasmid and 5 μg VSVG expressing plasmid were mixed in 1 mL Opti-MEM, followed by addition of 27.5 μl PEI MAX. After 20-30 min incubation at room temperature, the transfection reactions were dispersed dropwise over the HEK293T cells.


mVLPs were harvested at 48-72 hours post-transfection. To do this, mVLP supernatants were filtered using 0.45 μm PVDF or cellulose acetate or 0.8 μm PES membrane filters and transferred to polypropylene Beckman ultracentrifuge tubes that are used with the SW28 rotor (Beckman Coulter #326823). Each ultracentrifuge tube is filled with mVLP-containing supernatant from three 10 cm plates to reach an approximate final volume of 35-37.5 ml. mVLP supernatant underwent ultracentrifugation at approximately 100,000×g, or 25,000 rpm, at 4° C. for 2 hours. After ultracentrifugation, supernatants were decanted and mVLP pellets resuspended in DMEM 10% FBS media, or other media appropriate for the culturing of recipient cells, such that they are now approximately 1,000 times more concentrated than they were before ultracentrifugation. mVLPs were added dropwise to cells that were seeded in a 24-well plate 24-hours prior to transduction. Polybrene (5-10 μg/mL in cell culture medium; Sigma-Aldrich #TR-1003-G) was supplemented to enhance transduction efficiency, if necessary. Vectofusin-1 (10 μg/mL in cell culture medium, Miltenyi Biotec #130-111-163) was supplemented to enhance transduction efficiency, if necessary. Immediately following the addition of mVLPs, the 24-well plate was centrifuged at 1,150×g for 30 min at room temperature to enhance transduction efficiency, if necessary.


Example 1.1

mVLPs were produced by transient plasmid transfection of HEK293T cells (FIG. 1A). mVLPs were purified and concentrated 100-fold by filtration and PEG precipitation (FIG. 1B) and applied to K562 cells for an incubation period of 48 hours. K562 cells were harvested and genomic DNA was extracted. Targeted amplicon sequencing of extracted genomic DNA was used to quantify frequencies of gene modifications (y-axis) (i.e., gene edits) at the intended VEGFs3.1 on-target site (FIG. 2). We observed high frequencies of gene modification at this intended target sequence in K562 cells, demonstrating that mVLPs are capable of efficiently delivering RNP CRISPR-based cargo into living mammalian cells. FIGS. 3 and 4 are exemplary alignments of tENVs that are described herein.


Example 1.2


FIGS. 5-6 show that different phospholipid bilayer recruitment domains are capable of delivering cargo in previously described eVLPs (WO 2022/020800). eVLPs were produced by transient transfection of HEK293T cells, purified and concentrated 100-fold by filtration and PEG precipitation, and normalized based on Cas9 ELISA prior to transducing HEK293T cells so that the same pmol of Cas9 was applied in each well and comparisons could be made between different PH domains. Percent editing of endogenous VEGF was determined by amplicon sequencing (FIG. 5). These eVLPs were pseudotyped with VSVG. The results showed that different PH domain and mutant PH domain fusions to cargos will result in different delivery efficiencies.


Different mutant PH-Cas9 fusions (and Cas9 lacking a fusion to a PH domain) were packaged in eVLPs (made as described in WO 2022/020800), purified and concentrated 100-fold by PEG precipitation, and normalized by Cas9 ELISA so that 5 pmol of Cas9 was added to 15,000 primary T cells per well. Percent editing of endogenous RNF2 was determined by amplicon sequencing (FIG. 6). These eVLPs were pseudotyped with VSVG or a combination of VSVG and BaEVTRless. The results showed that different PH domain and mutant PH domain fusions to cargos will result in different delivery efficiencies which could be cell type dependent. In addition, different pseudotype combinations can also increase or decrease delivery efficiency.


REFERENCES



  • 1. Parseval, N. et al. Survey of human genes of retroviral origin: identification and transcriptome of the genes with coding capacity for complete envelope proteins. Journal of Virology 77, 10414-10422, (2003).

  • 2. Okimoto, T. et al. VSV-G envelope glycoprotein forms complexes with plasmid DNA and MLV retrovirus-like particles in cell-free conditions and enhances DNA transfection. Molecular Therapy 4, 232-238, (2001).

  • 3. Mangeot, P. et al. Protein transfer into human cells by VSV-G-induced nanovesicles. Molecular Therapy 19, 1656-1666, (2011).

  • 4. Wagner, D. et al. High prevalence of Streptococcus pyogenes Cas9-reactive T cells within the adult human population. Nature Medicine 25, 242-248 (2019)

  • 5. Kim, S. et al. CRISPR RNAs trigger innate immune responses in human cells. Genome Research 28, 1-7 (2018).

  • 6. Charlesworth, C. et al. Identification of preexisting adaptive immunity to Cas9 proteins in humans. Nature Medicine 25, 249-254 (2019)

  • 7. Ferdosi, S. et al. Multifunctional CRISPR-Cas9 with engineered immunosilenced human T cell epitopes. Nature Communications 10, Article number: 1842 (2019).

  • 8. Wang, D. et al. Adenovirus-mediated somatic genome editing of Pten by CRISPR/Cas9 in mouse liver in spite of Cas9-specific immune responses. Human Gene Therapy 26, 432-442 (2015).

  • 9. Devanabanda, M. et al. Immunotoxic effects of gold and silver nanoparticles: Inhibition of mitogen-induced proliferative responses and viability of human and murine lymphocytes in vitro. Journal of Immunotoxicology 13, 1547-6901 (2016).

  • 10. Mout, R. et al. Direct cytosolic delivery of CRISPR/Cas9-ribonucleoprotein for efficient gene editing. ACS Nano 11, 2452-2458 (2017).

  • 11. Yin, H. et al. structure-guided chemical modification of guide RNA enables potent non-viral in vivo genome editing. Nature Biotechnology 35, 1179-1187 (2017).

  • 12. Qiao, J. et al. Cytosolic delivery of CRISPR/Cas9 ribonucleoproteins for genome editing using chitosan-coated red fluorescent protein. Chemical Communications 55, 4707-4710 (2019).

  • 13. Li, L. et al. A rationally designed semiconducting polymer brush for NIR-II imaging guided light-triggered remote control of CRISPR/Cas9 genome editing. Advanced Materials 1901187, 1-9 (2019).

  • 14. Gao, X. et al. Treatment of autosomal dominant hearing loss by in vivo delivery of genome editing agents. Nature 553, 217-221 (2018)

  • 15. Lee, K. et al. Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair. Nature Biomedical Engineering 1, 889-901 (2017).

  • 16. Staahl, B. et al. Efficient genome editing in the mouse brain by local delivery of engineered Cas9 ribonucleoprotein complexes. Nature Biotechnology 35, 431-433 (2017).

  • 17. Zuris, J. et al. Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nature Biotechnology 33, 73-79 (2015).

  • 18. Finn, J. et al. A single administration of CRISPR/Cas9 lipid nanoparticles achieves robust and persistent in vivo genome editing. Cell Reports 22, 2227-2235 (2018).

  • 19. Wang, H. et al. Nonviral gene editing via CRISPR/Cas9 delivery by membrane-disruptive and endosomolytic helical polypeptide. PNAS 115, 4903-4908 (2018).

  • 20. Del'Guidice, T. et al. Membrane permeabilizing amphiphilic peptide delivers recombinant transcription factor and CRISPR-Cas9/Cpf1 ribonucleoproteins in hard-to-modify cells. PLOS ONE 13, e0195558 (2018).

  • 21. Colella, P. et al. Emerging Issues in AAV-Mediated In Vivo Gene Therapy. Molecular Therapy: Methods & Clinical Development 8, 87-104 (2018).

  • 22. Naso, F. et al. Adeno-Associated Virus (AAV) as a Vector for Gene Therapy. BioDrugs 31, 317-334 (2017).

  • 23. Handel, E. et al. Versatile and efficient genome editing in human cells by combining zinc-finger nucleases with adeno-associated viral vectors. Human Gene Therapy 23, 321-329 (2012).

  • 24. Chadwick, A. et al. Reduced Blood Lipid Levels With In Vivo CRISPR-Cas9 Base Editing of ANGPTL3. Circulation 137, 975-977 (2018).

  • 25. Schenkwein, D. et al. Production of HIV-1 Integrase Fusion Protein-Carrying Lentiviral Vectors for Gene Therapy and Protein Transduction. Human Gene Therapy 21, 589-602 (2010).

  • 26. Cai, Y. et al. Targeted genome editing by lentiviral protein transduction of zinc-finger and TAL-effector nucleases. eLife 3, e01911 (2014).

  • 27. Choi, J. et al. Lentivirus pre-packed with Cas9 protein for safer gene editing. Gene Therapy 23, 627-633 (2016).

  • 28. Meyer, C. et al. Pseudotyping exosomes for enhanced protein delivery in mammalian cells. International Journal of Nanomedicine 12, 3153-3170 (2017).

  • 29. Mangeot, P. et al. Genome editing in primary cells and in vivo using viral-derived Nanoblades loaded with Cas9-sgRNA ribonucleoproteins. Nature Communications 10, Article number: 45 (2019).

  • 30. Lu, B. et al. Delivering SaCas9 mRNA by lentivirus-like bionanoparticles for transient expression and efficient genome editing. Nucleic Acids Research 47, e44 (2019).

  • 31. Wang, Q. et al. ARMMs as a versatile platform for intracellular delivery of macromolecules. Nature Communications 9, 1-7 (2018).

  • 32. Lainscek, D. et al. Delivery of an Artificial Transcription Regulator dCas9-VPR by Extracellular Vesicles for Therapeutic Gene Activation. ACS Synthetic Biology 7, 2715-2725 (2018).

  • 33. Fuchs, J. et al. First-in-Human Evaluation of the Safety and Immunogenicity of a Recombinant Vesicular Stomatitis Virus Human Immunodeficiency Virus-1 gag Vaccine (HVTN 090). Open Forum Infectious Diseases 2, 1-9, (2015).

  • 34. Cong, L. et al. Multiplex Genome Engineering Using CRISPR/Cas Systems. Science 339, 819-823, (2013).

  • 35. Ran, F. et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520, 186-191, (2015).

  • 36. Zetsche, B. et al. Cpf1 Is a Single RNA-Guided Endonuclease of a Class 2 CRISPR-Cas System. Cell 163, 759-771, (2015).

  • 37. Komor, A. et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420-424, (2016).

  • 38. Gaudelli, N. et al. Programmable base editing of A·T to G·C in genomic DNA without DNA cleavage. Nature 551, 464-471, (2017).

  • 39. Voelkel, C. et al. Protein transduction from retroviral Gag precursors. Proc Natl Acad Sci USA 107, 7805-7810, (2010).

  • 40. Kaczmarczyk, S. et al. Protein delivery using engineered virus-like particles. Proc Natl Acad Sci USA 108, 16998-17003, (2011).

  • 41. Ebner, M. et al. PI(3,4,5)P3 Engagement Restricts Akt Activity to Cellular Membranes. Mol Cell 65, 416-431, (2017).

  • 42. Urano, E. et al. Substitution of the myristoylation signal of human immunodeficiency virus type 1 Pr55Gag with the phospholipase C-dl pleckstrin homology domain results in infectious pseudovirion production. J. Gen Virology 89, 3144-3149, (2008).

  • 43. Pastuzyn, E. et al. The Neuronal Gene Arc Encodes a Repurposed Retrotransposon Gag Protein that Mediates Intercellular RNA Transfer. Cell 172, 275-288, (2018).

  • 44. Lukacs, G. et al. Size-dependent DNA Mobility in Cytoplasm and Nucleus. Journal of Biological Chemistry 275, 1625-1629, (1999).

  • 45. Kreiss, P. et al. Plasmid DNA size does not affect the physicochemical properties of lipoplexes but modulates gene transfer efficiency. Nucleic Acids Research 27, 3792-3798 (1999).

  • 46. Nafissi, N. et al. DNA Ministrings: Highly Safe and Effective Gene Delivery Vectors. Molecular Therapy-Nucleic Acids 3, e165, (2014).

  • 47. Fujimoto, T. et al. Selective EGLN Inhibition Enables Ablative Radiotherapy and Improves Survival in Unresectable Pancreatic Cancer. Cancer Research 79, 2327-2338 (2019).

  • 48. Tai, S. et al. Differential Expression of Metallothionein 1 and 2 Isoforms in Breast Cancer Lines with Different Invasive Potential: Identification of a Novel Nonsilent Metallothionein-1H Mutant Variant. American Journal of Pathology 163, 2009-2019 (2003).

  • 49. Caussinus, E. et al. Fluorescent fusion protein knockout mediated by anti-GFP nanobody. Nature Structural & Molecular Biology 19, 117-121, (2012).

  • 50. Zhao, W. et al. Quantitatively Predictable Control of Cellular Protein Levels through Proteasomal Degradation. ACS Synthetic Biology 7, 540-552, (2018).

  • 51. Clift, D. et al. A Method for the Acute and Rapid Degradation of Endogenous Proteins. Cell 171, 1692-1706, (2017).

  • 52. Balla, T. & Virnai, T. Visualizing Cellular Phosphoinositide Pools with GFP-Fused Protein-Modules. SCIENCE'S STKE 2002, p. pl3, DOI: 10.1126/stke.2002.125.pl3 (2002).

  • 53. Carpten, J. et al. A transforming mutation in the pleckstrin homology domain of AKT1 in cancer. Nature 448, 439-445, (2007).

  • 54. Virnai, P. et al. Selective cellular effects of overexpressed pleckstrin-homology domains that recognize PtdIns(3,4,5)P3 suggest their interaction with protein binding partners. Journal of Cell Science 118, 4879-4888, (2005).

  • 55. Parikh, C. et al. Disruption of PH-kinase domain interactions leads to oncogenic activation of AKT in human cancers. PNAS 109, 19368-19373, (2012).

  • 56. Jo, H. et al. Small molecule-induced cytosolic activation of protein kinase Akt rescues ischemia-elicited neuronal death. PNAS 109 (26) 10581-10586, (2012).

  • 57. Han, F. et al. The critical role of AMPK in driving Akt activation under stress, tumorigenesis and drug resistance. Nat Commun 9, 4728 (2018). doi.org/10.1038/s41467-018-07188-9

  • 58. Li, X. et al. Autophosphorylation of Akt at Threonine 72 and Serine 246: A POTENTIAL MECHANISM OF REGULATION OF Akt KINASE ACTIVITY*. Journal of Biological Chemistry 281, 13837-13843, (2006).

  • 59. Liao, Y. et al. Peptidyl-prolyl cis/trans isomerase Pin1 is critical for the regulation of PKB/Akt stability and activation phosphorylation. Oncogene 28(26):2436-45, DOI:10.1038/onc.2009.98 (2009).

  • 60. Chu, N. et al. Akt Kinase Activation Mechanisms Revealed Using Protein Semisynthesis. Cell 174(4): 897-907.e14. (2018).

  • 61. Lucid, I. et al. Conformational sampling of membranes by Akt controls its activation and inactivation. Proc Natl Acad Sci USA 115(17): E3940 E3949, (2018).

  • 62. Baranov, M. et al. SWAP70 Organizes the Actin Cytoskeleton and Is Essential for Phagocyiosis. Cell Reports 17, p 1518-1531 (2016)



OTHER EMBODIMENTS

It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims
  • 1. A truncated glycoprotein/envelope protein (tENV) comprising: an N-terminal portion comprising a signal sequence, optionally comprising the MKCLLYLAFLFIGVNCK (SEQ ID NO:1), fused to a central portion comprising all or part of a GS domain from at least one vesiculovirus G protein, optionally a VSV-G protein or homolog, ortholog, or paralog thereof, optionally comprising the sequence FEHPHIQDAASQLPDDESLFFGDTGLSKNPIELVEGWFSSWK (SEQ ID NO:2), optionally with a deletion of at least one amino acid (optionally a truncation from the N terminal end of the central portion), which is fused to a C-terminal portion comprising a transmembrane domain and an intracellular domain, optionally comprising
  • 2. The tENV of claim 1, wherein the central portion comprises a deletion of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 38, 39, 40, 41, or all 42 amino acids of SEQ ID NO:2 amino acids, e.g., at least about 1, 2, 3, 4, 5, 6, 7, 8 or 10 amino acids, up to about 15, 20, 35, 30, 35, 38, 39, 40, 41, or all 42 amino acids, with any range therebetween.
  • 3. The tENV of claim 1, wherein the central portion comprises
  • 4. The tENV of claim 1, comprising a sequence that is at least 95% identical to a sequence set forth herein, e.g., in Table 1.
  • 5. A nucleic acid sequence encoding the tENV of claim 1.
  • 6. A vector comprising a nucleic acid sequence encoding the tENV of claim 1, optionally operably linked to a promoter for expression of the tENV of claim 1.
  • 7. A host cell comprising the nucleic acid sequence encoding the tENV of claim 1, and optionally expressing the tENV of claim 1.
  • 8. A virus-like particle (VLP) comprising the tENV of claim 1.
  • 9. A minimal virus-like particle (mVLP), comprising: a membrane comprising a phospholipid bilayer and the tENV of claim 1; andoptionally, a cargo disposed in the core of the mVLP, wherein the cargo is optionally fused to a phospholipid bilayer recruitment domain; and,wherein the mVLP does not comprise an exogenous gag, pro, or pol protein.
  • 10. The mVLP of claim 9, wherein the cargo is a therapeutic or diagnostic protein or nucleic acid encoding a therapeutic or diagnostic protein, or a chemical, optionally a small molecule therapeutic or diagnostic.
  • 11. The mVLP of claim 9, wherein the cargo is a gene editing or epigenetic modulating reagent.
  • 12. The mVLP of claim 9, wherein the gene editing or epigenetic modulating reagent comprises a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof; a nucleic acid encoding a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof; a guide RNA and/or crRNA; or a ribonucleoprotein complex (RNP) comprising a CRISPR-Cas protein, variant, or fusion thereof and optionally a guide RNA.
  • 13. The mVLP of claim 12, wherein the cargo is selected from the proteins listed in Tables 2, 3, 4 & 5, or that is at least 95% identical to a sequence set forth in Table 2, 3, 4, or 5.
  • 14. The mVLP of claim 12, wherein the cargo comprises a CRISPR-Cas protein, and the mVLP further comprises one or more guide RNAs that bind to and direct the CRISPR-Cas protein to a target nucleic acid sequence.
  • 15. The mVLP of claim 9, wherein the cargo comprises a fusion to a phospholipid bilayer recruitment domain, preferably as shown in Table 6, or that is at least 95% identical to a sequence set forth herein in Table 6.
  • 16. A method of delivering a cargo to a target cell, optionally a cell in vivo or in vitro, the method comprising contacting the cell with the mVLP of claim 9 comprising the cargo.
  • 17. A method of producing a VLP or an mVLP comprising a cargo, the method comprising: providing a cell expressing the tENV of claim 1 and a cargo, optionally wherein the cell does not express an exogenous gag, pro, or pol protein; andmaintaining the cell under conditions such that the cells produce the VLPs or mVLPs.
  • 18. The method of claim 17, further comprising harvesting and optionally purifying and/or concentrating the produced VLPs or mVLPs.
  • 19. The method of claim 17, wherein the cargo is a therapeutic or diagnostic protein or nucleic acid encoding a therapeutic or diagnostic protein, or a small molecule, optionally a therapeutic or diagnostic small molecule.
  • 20. The method of claim 17, wherein the cargo is a gene editing or epigenetic modulating reagent.
  • 21. The method of claim 17, wherein the gene editing or epigenetic modulating reagent comprises a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof; a nucleic acid encoding a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof; a guide RNA and/or crRNA; or a ribonucleoprotein complex (RNP) comprising a CRISPR-Cas protein, variant, or fusion thereof and optionally a guide RNA.
  • 22. The method of claim 21, wherein the cargo reagent is selected from the proteins listed in Tables 2, 3, 4 & 5, or that is at least 95% identical to a sequence set forth in Table 2, 3, 4, or 5.
  • 23. The method of claim 21, wherein the cargo reagent comprises a CRISPR-Cas protein, variant, or fusion thereof and the mVLP further comprises one or more guide RNAs that bind to and direct the CRISPR-based genome editing or modulating protein to a target sequence.
  • 24. The method of claim 17, wherein the cargo comprises a fusion to a phospholipid bilayer recruitment domain, preferably as shown in Table 6, or that is at least 95% identical to a sequence set forth herein in Table 6.
  • 25. A cell expressing the tENV of claim 1, and a cargo, optionally wherein the cell does not express an exogenous gag, pro, or pol protein.
  • 26. The cell of claim 25, wherein the cargo is a therapeutic or diagnostic protein or nucleic acid encoding a therapeutic or diagnostic protein, or a small molecule, optionally a therapeutic or diagnostic small molecule.
  • 27. The cell of claim 25, wherein the cargo is a gene editing or epigenetic modulating reagent.
  • 28. The cell of claim 25, wherein the gene editing or epigenetic modulating reagent comprises a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof; a nucleic acid encoding a zinc finger (ZF), transcription activator-like effector (TALE), and/or CRISPR-Cas protein, variant, or fusion thereof; a guide RNA and/or crRNA; or a ribonucleoprotein complex (RNP) comprising a CRISPR-Cas protein, variant, or fusion thereof and optionally a guide RNA.
  • 29. The cell of claim 28, wherein the cargo reagent is selected from the proteins listed in Tables 2, 3, 4, & 5, or that is at least 95% identical to a sequence set forth in Table 2, 3, 4, or 5.
  • 30. The cell of claim 28, wherein the gene editing or epigenetic modulating reagent comprises a CRISPR-Cas protein, and the mVLP further comprises one or more guide RNAs that bind to and direct the CRISPR-Cas protein to a target sequence.
  • 31. The cell of claim 25, wherein the cargo comprises a fusion to a phospholipid bilayer recruitment domain, preferably as shown in Table 6, or that is at least 95% identical to a sequence set forth herein in Table 6.
  • 32. A primary or stable human cell line comprising cells expressing the tENV of claim 1, and a cargo, optionally wherein the cell does not express an exogenous gag, pro, or pol protein.
  • 33. The cells of claim 32, which are Human Embryonic Kidney (HEK) 293 cells or HEK293 T cells.
CLAIM OF PRIORITY

This application claims the benefit of U.S. Provisional Patent Application Ser. No. 63/425,890, filed on Nov. 16, 2022. The entire contents of the foregoing are hereby incorporated by reference.

FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT

This invention was made with Government support under Grant No. GM118158 awarded by the National Institutes of Health. The Government has certain rights in the invention.

Provisional Applications (1)
Number Date Country
63425890 Nov 2022 US