DIFFERENTIAL KNOCKOUT OF A HETEROZYGOUS ALLELE OF RPE65

Information

  • Patent Application
  • 20220267777
  • Publication Number
    20220267777
  • Date Filed
    July 10, 2020
    3 years ago
  • Date Published
    August 25, 2022
    a year ago
Abstract
RNA molecules comprising a guide sequence portion having 17-25 contiguous nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and compositions, methods, and uses thereof.
Description
REFERENCE TO SEQUENCE LISTING

This application incorporates-by-reference nucleotide sequences which are present in the file named “200710_91034-A-PCT_Sequence_Listing_AWG.txt”, which is 9,361 kilobytes in size, and which was created on Jul. 6, 2020 in the IBM-PC machine format, having an operating system compatibility with MS-Windows, which is contained in the text file filed Jul. 10, 2020 as part of this application.


BACKGROUND OF INVENTION

There are several classes of DNA variation in the human genome, including insertions and deletions, differences in the copy number of repeated sequences, and single nucleotide polymorphisms (SNPs). A SNP is a DNA sequence variation occurring when a single nucleotide (adenine (A), thymine (T), cytosine (C), or guanine (G)) in the genome differs between human subjects or paired chromosomes in an individual. Over the years, the different types of DNA variations have been the focus of the research community either as markers in studies to pinpoint traits or disease causation or as potential causes of genetic disorders.


A genetic disorder is caused by one or more abnormalities in the genome. Genetic disorders may be regarded as either “dominant” or “recessive.” Recessive genetic disorders are those which require two copies (i.e., two alleles) of the abnormal/defective gene to be present. In contrast, a dominant genetic disorder involves a gene or genes which exhibit(s) dominance over a normal (functional/healthy) gene or genes. As such, in dominant genetic disorders only a single copy (i.e., allele) of an abnormal gene is required to cause or contribute to the symptoms of a particular genetic disorder. Such mutations include, for example, gain-of-function mutations in which the altered gene product possesses a new molecular function or a new pattern of gene expression. Other examples include dominant negative mutations, which have a gene product that acts antagonistically to the wild-type allele.


Dominant RPE65 Mutation Related Disorder

Most of the mutations in the retinal pigment epithelium-specific 65 kDa protein gene (RPE65) are recessive. However, an Asp477Gly in Exon 13 was shown to be a dominant RPE65 mutation resulting in retinitis pigmentosa (Sara J. Bowne et al. 2011). Yet another mutation originally characterized to be semidominant in mice (Wright et al. 2013) was identified in humans as well (R44X_rs368088025_G>A).


SUMMARY OF THE INVENTION

Disclosed is an approach for knocking out the expression of a dominant-mutated RPE65 allele by disrupting the dominant-mutated allele or degrading the resulting mRNA.


The present disclosure provides a method for utilizing at least one naturally occurring nucleotide difference or polymorphism (e.g., single nucleotide polymorphism (SNP)) for distinguishing/discriminating between two alleles of a gene, one allele bearing a mutation such that it encodes a mutated protein causing a disease phenotype (“mutated allele”) and a particular sequence in a SNP position (REF/SNP), and the other allele encoding for a functional protein (“functional allele”). In some embodiments, the SNP position is utilized for distinguishing/discriminating between two alleles of a gene bearing one or more disease associated mutations, such as to target one of the alleles bearing both the particular sequence in the SNP position (SNP/REF) and a disease associated mutation. In some embodiments, the disease-associated mutation is targeted. In some embodiments, the method further comprises the step of knocking out expression of the mutated protein and allowing expression of the functional protein.


The present disclosure also provides a method for modifying in a cell a mutant allele of the retinal pigment epithelium-specific 65 kDa protein gene (RPE65) gene having a mutation associated with a dominant RPE65 gene disorder, the method comprising

    • introducing to the cell a composition comprising:
      • a CRISPR nuclease or a sequence encoding the CRISPR nuclease; and
      • a first RNA molecule comprising a guide sequence portion having 17-25 nucleotides or a nucleotide sequence encoding the same,
    • wherein a complex of the CRISPR nuclease and the first RNA molecule affects a double strand break in the mutant allele of the RPE65 gene.


According to embodiments of the present invention, there is provided a first RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID Nos: 1-49516.


According to some embodiments of the present invention, there is provided a composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease.


According to some embodiments of the present invention, there is provided a method for inactivating a mutant RPE65 allele in a cell, the method comprising delivering to the cell a composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease. In some embodiments, the cell is a stem cell. In some embodiments, the stem cell is an autologous pluripotent stem cell or an induced pluripotent stem cell (iPSC). In some embodiments, the stem cell is differentiated into a retinal pigment epithelium cell. In some embodiments, the cell is a retinal pigment epithelium cell. In some embodiments, the delivering to the cell is performed in vitro, ex vivo, or in vivo. In some embodiments, the method is performed ex-vivo and the cell is provided/explanted from an individual patient. In some embodiments, the method further comprises the step of introducing the resulting cell, with the modified/knocked out mutant RPE65 allele, into the individual patient (e.g. autologous transplantation).


According to some embodiments of the present invention, there is provided a method for treating a dominant RPE65 gene disorder, the method comprising delivering to a cell of a subject having a dominant RPE65 gene disorder a composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease.


According to some embodiments of the present invention, there is provided use of a composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease for inactivating a mutant RPE65 allele in a cell, comprising delivering to the cell the composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease.


According to embodiments of the present invention, there is provided a medicament comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease for use in inactivating a mutant RPE65 allele in a cell, wherein the medicament is administered by delivering to the cell the composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease.


According to some embodiments of the present invention, there is provided use of a composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease for treating ameliorating or preventing a dominant RPE65 gene disorder, comprising delivering to a cell of a subject having or at risk of having a dominant RPE65 gene disorder the composition of comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease. In some embodiments, the method is performed ex vivo and the cell is provided/explanted from the subject. In some embodiments, the method further comprises the step of introducing the resulting cell, with the modified/knocked out mutant RPE65 allele, into the subject (e.g. autologous transplantation).


According to some embodiments of the present invention, there is provided a medicament comprising the composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease for use in treating ameliorating or preventing a dominant RPE65 gene disorder, wherein the medicament is administered by delivering to a cell of a subject having or at risk of having a dominant RPE65 gene disorder the composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease.


According to some embodiments of the present invention, there is provided a kit for inactivating a mutant RPE65 allele in a cell, comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516, a CRISPR nuclease, and/or a tracrRNA molecule; and instructions for delivering the RNA molecule; CRISPR nuclease, and/or the tracrRNA to the cell.


According to some embodiments of the present invention, there is provided a kit for treating a dominant RPE65 gene disorder in a subject, comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516, a CRISPR nuclease, and/or a tracrRNA molecule; and instructions for delivering the RNA molecule; CRISPR nuclease, and/or the tracrRNA to a cell of a subject having or at risk of having a dominant RPE65 gene disorder.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A-B: Activity of guides targeting the p.Asp477Gly (c.1430A>G) mutation of RPE65 in patient-derived iPSCs. RNPs complexed with SpCas9 (FIG. 1A) or OMNI-50 (FIG. 1B). The nuclease and specific guide were electroporated into iPSCs to determine their activity. Cells were harvested 72 h post DNA electroporation, genomic was DNA extracted, and the region of the mutation was amplified and analyzed by capillary electrophoreses. The graphs represent the % editing ±STDV of two independent electroporation trials.





DETAILED DESCRIPTION

Unless otherwise defined, all technical and/or scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be necessarily limiting.


It should be understood that the terms “a” and “an” as used above and elsewhere herein refer to “one or more” of the enumerated components. It will be clear to one of ordinary skill in the art that the use of the singular includes the plural unless specifically stated otherwise. Therefore, the terms “a,” “an” and “at least one” are used interchangeably in this application.


For purposes of better understanding the present teachings and in no way limiting the scope of the teachings, unless otherwise indicated, all numbers expressing quantities, percentages or proportions, and other numerical values used in the specification and claims, are to be understood as being modified in all instances by the term “about.” Accordingly, unless indicated to the contrary, the numerical parameters set forth in the following specification and attached claims are approximations that may vary depending upon the desired properties sought to be obtained. At the very least, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques.


Unless otherwise stated, adjectives such as “substantially” and “about” modifying a condition or relationship characteristic of a feature or features of an embodiment of the invention, are understood to mean that the condition or characteristic is defined to within tolerances that are acceptable for operation of the embodiment for an application for which it is intended. Unless otherwise indicated, the word “or” in the specification and claims is considered to be the inclusive “or” rather than the exclusive or, and indicates at least one of, or any combination of items it conjoins.


In the description and claims of the present application, each of the verbs, “comprise,” “include” and “have” and conjugates thereof, are used to indicate that the object or objects of the verb are not necessarily a complete listing of components, elements or parts of the subject or subjects of the verb. Other terms as used herein are meant to be defined by their well-known meanings in the art.


The terms “nucleic acid template” and “donor”, refer to a nucleotide sequence that is inserted or copied into a genome. The nucleic acid template comprises a nucleotide sequence, e.g., of one or more nucleotides, that will be added to or will template a change in the target nucleic acid or may be used to modify the target sequence. A nucleic acid template sequence may be of any length, for example between 2 and 10,000 nucleotides in length, preferably between about 100 and 1,000 nucleotides in length, more preferably between about 200 and 500 nucleotides in length. A nucleic acid template may be a single stranded nucleic acid, a double stranded nucleic acid. In some embodiments, the nucleic acid template comprises a nucleotide sequence, e.g., of one or more nucleotides, that corresponds to wild type sequence of the target nucleic acid, e.g., of the target position. In some embodiments, the nucleic acid template comprises a nucleotide sequence, e.g., of one or more ribonucleotides, that corresponds to wild type sequence of the target nucleic acid, e.g., of the target position. In some embodiments, the nucleic acid template comprises modified nucleotides.


Insertion of an exogenous sequence (also called a “donor sequence,” donor template,” “donor molecule” or “donor”) can also be carried out. For example, a donor sequence can contain a non-homologous sequence flanked by two regions of homology to allow for efficient HDR at the location of interest. Additionally, donor sequences can comprise a vector molecule containing sequences that are not homologous to the region of interest in cellular chromatin. A donor molecule can contain several, discontinuous regions of homology to cellular chromatin. For example, for targeted insertion of sequences not normally present in a region of interest, said sequences can be present in a donor nucleic acid molecule and flanked by regions of homology to sequence in the region of interest. A donor molecule may be any length, for example ranging from several bases e.g. 10-20 bases to multiple kilobases in length.


The donor polynucleotide can be DNA or RNA, single-stranded and/or double-stranded and can be introduced into a cell in linear or circular form. See, e.g., U.S. Patent Publication Nos. 2010/0047805; 2011/0281361; 2011/0207221; and 2019/0330620. If introduced in linear form, the ends of the donor sequence can be protected (e.g., from exonucleolytic degradation) by methods known to those of skill in the art. For example, one or more dideoxynucleotide residues are added to the 3′ terminus of a linear molecule and/or self-complementary oligonucleotides are ligated to one or both ends. See, for example, Chang et al. (1987) and Nehls et al. (1996). Additional methods for protecting exogenous polynucleotides from degradation include, but are not limited to, addition of terminal amino group(s) and the use of modified internucleotide linkages such as, for example, phosphorothioates, phosphoramidates, and O-methyl ribose or deoxyribose residues.


A donor sequence may be an oligonucleotide and be used for targeted alteration of an endogenous sequence. The oligonucleotide may be introduced to the cell on a vector, may be electroporated into the cell, or may be introduced via other methods known in the art. Donor polynucleotides can be introduced as naked nucleic acid, as nucleic acid complexed with an agent such as a liposome or poloxamer, or can be delivered by viruses (e.g., adenovirus, AAV, herpesvirus, retrovirus, lentivirus and integrase defective lentivirus (IDLV)).


As used herein, the term “modified cells” refers to cells in which a double strand break is affected by a complex of an RNA molecule and the CRISPR nuclease as a result of hybridization with the target sequence, i.e. on-target hybridization. The term “modified cells” may further encompass cells in which a repair or correction of a mutation was affected following the double strand break.


This invention provides a modified cell or cells obtained by use of any of the methods described herein. In an embodiment these modified cell or cells are capable of giving rise to progeny cells. In an embodiment these modified cell or cells are capable of giving rise to progeny cells after engraftment. As a non-limiting example, the modified cells may be stem cells, or any cell suitable for an allogenic cell transplant or autologous cell transplant. As a non-limiting example, the modified cell may be a stem cell. In a non-limiting example, the stem cell is an autologous pluripotent stem cell or an induced pluripotent stem cell (iPSC). As another non-limiting example, the stem cell is differentiated into a retinal pigment epithelium cell. In yet another non-limiting example, the modified cell is a retinal pigment epithelium cell.


This invention also provides a composition comprising these modified cells and a pharmaceutically acceptable carrier. Also provided is an in vitro or ex vivo method of preparing this, comprising mixing the cells with the pharmaceutically acceptable carrier.


As used herein, the term “targeting sequence” or “targeting molecule” refers a nucleotide sequence or molecule comprising a nucleotide sequence that is capable of hybridizing to a specific target sequence, e.g., the targeting sequence has a nucleotide sequence which is at least partially complementary to the sequence being targeted along the length of the targeting sequence. The targeting sequence or targeting molecule may be part of an RNA molecule that can form a complex with a CRISPR nuclease with the targeting sequence serving as the targeting portion of the CRISPR complex. When the molecule having the targeting sequence is present contemporaneously with the CRISPR molecule the RNA molecule is capable of targeting the CRISPR nuclease to the specific target sequence. Each possibility represents a separate embodiment. An RNA molecule can be custom designed to target any desired sequence.


The term “targets” as used herein, refers to a targeting sequence or targeting molecule's preferential hybridization to a nucleic acid having a targeted nucleotide sequence. It is understood that the term “targets” encompasses variable hybridization efficiencies, such that there is preferential targeting of the nucleic acid having the targeted nucleotide sequence, but unintentional off-target hybridization in addition to on-target hybridization might also occur. It is understood that where an RNA molecule targets a sequence, a complex of the RNA molecule and a CRISPR nuclease molecule targets the sequence for nuclease activity.


The “guide sequence portion” of an RNA molecule refers to a nucleotide sequence that is capable of hybridizing to a specific target DNA sequence, e.g., the guide sequence portion has a nucleotide sequence which is fully complementary to the DNA sequence being targeted along the length of the guide sequence portion. In some embodiments, the guide sequence portion is 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleotides in length, or approximately 17-25, 17-24, 17-22, 17-21, 18-25, 18-24, 18-23, 18-22, 18-21, 19-25, 19-24, 19-23, 19-22, 19-21, 19-20, 20-22, 18-20, 20-21, 21-22, or 17-20 nucleotides in length. The entire length of the guide sequence portion is fully complementary to the DNA sequence being targeted along the length of the guide sequence portion. The guide sequence portion may be part of an RNA molecule that can form a complex with a CRISPR nuclease with the guide sequence portion serving as the DNA targeting portion of the CRISPR complex. When the DNA molecule having the guide sequence portion is present contemporaneously with the CRISPR molecule the RNA molecule is capable of targeting the CRISPR nuclease to the specific target DNA sequence. Each possibility represents a separate embodiment. An RNA molecule can be custom designed to target any desired sequence.


The term “non-discriminatory” as used herein refers to a guide sequence portion of an RNA molecule that targets a specific DNA sequence that is common both a mutant and functional allele of a gene.


In embodiments of the present invention, an RNA molecule comprises a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516.


The RNA molecule and or the guide sequence portion of the RNA molecule may contain modified nucleotides. Exemplary modifications to nucleotides or polynucleotides may be synthetic and encompass polynucleotides which contain nucleotides comprising bases other than the naturally occurring adenine, cytosine, thymine, uracil, or guanine bases. Modifications to polynucleotides include polynucleotides which contain synthetic, non-naturally occurring nucleosides e.g., locked nucleic acids. Modifications to polynucleotides may be utilized to increase or decrease stability of an RNA. An example of a modified polynucleotide is an mRNA containing 1-methyl pseudo-uridine. For examples of modified polynucleotides and their uses, see U.S. Pat. No. 8,278,036, PCT International Publication No. WO/2015/006747, and Weissman and Kariko (2015), hereby incorporated by reference.


As used herein, “contiguous nucleotides” set forth in a SEQ ID NO refers to nucleotides in a sequence of nucleotides in the order set forth in the SEQ ID NO without any intervening nucleotides.


In embodiments of the present invention, the guide sequence portion may be at least 25 nucleotides in length and contain 20-22 contiguous nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516. In embodiments of the present invention, the guide sequence portion may be less than 22 nucleotides in length. For example, in embodiments of the present invention the guide sequence portion may be 17, 18, 19, 20, or 21 nucleotides in length. In such embodiments the guide sequence portion may consist of 17, 18, 19, 20, or 21 nucleotides, respectively, in the sequence of 17-22 contiguous nucleotides set forth in any one of SEQ ID NOs: 1-49516. For example, a guide sequence portion having 17 nucleotides in the sequence of 17 contiguous nucleotides set forth in SEQ ID NO: 49517 may consist of any one of the following nucleotide sequences (nucleotides excluded from the contiguous sequence are marked in strike-through):











(SEQ ID NO: 49517)



AAAAAAAUGUACUUGGUUCC







17 nucleotide guide sequence 1:



(SEQ ID NO: 49518)




custom-character AAAAUGUACUUGGUUCC








17 nucleotide guide sequence 2:



(SEQ ID NO: 49519)




custom-character AAAAAUGUACUUGGUUcustom-character








17 nucleotide guide sequence 3:



(SEQ ID NO: 49520)




custom-character AAAAAAUGUACUUGGUUcustom-character








17 nucleotide guide sequence 4:



(SEQ ID NO: 49521)



AAAAAAAUGUACUUGGUcustom-character






In embodiments of the present invention, the guide sequence portion may be greater than 20 nucleotides in length. For example, in embodiments of the present invention the guide sequence portion may be 21, 22, 23, 24 or 25 nucleotides in length. In such embodiments the guide sequence portion comprises 17-25 nucleotides containing the sequence of 20, 21 or 22 contiguous nucleotides set forth in any one of SEQ ID NOs: 1-49516 and additional nucleotides fully complimentary to a nucleotide or sequence of nucleotides adjacent to the 3′ end of the target sequence, 5′ end of the target sequence, or both.


In embodiments of the present invention a CRISPR nuclease and an RNA molecule comprising a guide sequence portion form a CRISPR complex that binds to a target DNA sequence to effect cleavage of the target DNA sequence. CRISPR nucleases, e.g. Cpf1, may form a CRISPR complex comprising a CRISPR nuclease and RNA molecule without a further tracrRNA molecule. Alternatively, CRISPR nucleases, e.g. Cas9, may form a CRISPR complex between the CRISPR nuclease, an RNA molecule, and a tracrRNA molecule.


In embodiments of the present invention, the RNA molecule may further comprise the sequence of a tracrRNA molecule. Such embodiments may be designed as a synthetic fusion of the guide portion of the RNA molecule and the trans-activating crRNA (tracrRNA). (See Jinek et al., 2012). Embodiments of the present invention may also form CRISPR complexes utilizing a separate tracrRNA molecule and a separate RNA molecule comprising a guide sequence portion. In such embodiments the tracrRNA molecule may hybridize with the RNA molecule via basepairing and may be advantageous in certain applications of the invention described herein.


The term “tracr mate sequence” refers to a sequence sufficiently complementary to a tracrRNA molecule so as to hybridize to the tracrRNA via basepairing and promote the formation of a CRISPR complex. (See U.S. Pat. No. 8,906,616). In embodiments of the present invention, the RNA molecule may further comprise a portion having a tracr mate sequence.


A “gene,” for the purposes of the present disclosure, includes a DNA region encoding a gene product, as well as all DNA regions which regulate the production of the gene product, whether or not such regulatory sequences are adjacent to coding and/or transcribed sequences. Accordingly, a gene includes, but is not necessarily limited to, promoter sequences, terminators, translational regulatory sequences such as ribosome binding sites and internal ribosome entry sites, enhancers, silencers, insulators, boundary elements, replication origins, matrix attachment sites and locus control regions.


“Eukaryotic” cells include, but are not limited to, fungal cells (such as yeast), plant cells, animal cells, mammalian cells and human cells.


The term “nuclease” as used herein refers to an enzyme capable of cleaving the phosphodiester bonds between the nucleotide subunits of nucleic acid. A nuclease may be isolated or derived from a natural source. The natural source may be any living organism. Alternatively, a nuclease may be a modified or a synthetic protein which retains the phosphodiester bond cleaving activity. Gene modification can be achieved using a nuclease, for example a CRISPR nuclease.


As used herein, “progenitor cell” refers to a lineage cell that is derived from stem cell and retains mitotic capacity and multipotency (e.g., can differentiate or develop into more than one but not all types of mature lineage of cell).


The term “single nucleotide polymorphism (SNP) position”, as used herein, refers to a position in which a single nucleotide DNA sequence variation occurs between members of a species, or between paired chromosomes in an individual. In the case that a SNP position exists at paired chromosomes in an individual, a SNP on one of the chromosomes is a “heterozygous SNP.” The term SNP position refers to the particular nucleic acid position where a specific variation occurs and encompasses both a sequence including the variation from the most frequently occurring base at the particular nucleic acid position (also referred to as “SNP” or alternative “ALT”) and a sequence including the most frequently occurring base at the particular nucleic acid position (also referred to as reference, or “REF”). Accordingly, the sequence of a SNP position may reflect a SNP (i.e. an alternative sequence variant relative to a consensus reference sequence within a population), or the reference sequence itself.


According to embodiments of the present invention, there is provided a method for modifying in a cell a mutant allele of the retinal pigment epithelium-specific 65 kDa protein gene (RPE65) gene having a mutation associated with a dominant RPE65 gene disorder, the method comprising

    • introducing to the cell a composition comprising:
      • at least one CRISPR nuclease or a sequence encoding a CRISPR nuclease; and
      • a first RNA molecule comprising a guide sequence portion having 17-25 nucleotides or a nucleotide sequence encoding the same,
    • wherein a complex of the CRISPR nuclease and the first RNA molecule affects a double strand break in the mutant allele of the RPE65 gene.


In some embodiments, the first RNA molecule targets the CRISPR nuclease to the mutation associated with a dominant RPE65 gene disorder.


In some embodiments, the mutation associated with a dominant RPE65 gene disorder is any one of 1:68431085_T_C and 1:68446825_G_A.


In some embodiments, the guide sequence portion of the first RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 that targets a mutation associated with a dominant RPE65 gene disorder.


In some embodiments, the first RNA molecule targets the CRISPR nuclease to a SNP position of the mutant allele.


In some embodiments, the SNP position is any one of rs60701104, rs9436400, rs868541802, rs3125890, rs75159457, rs1205919238, rs11209300, rs4264030, rs2419988, rs3118415, rs3118416, rs149739986, rs2182315, rs3118418, rs932783, rs12124063, rs77585943, rs1886906, rs3125891, rs11581095, rs12030710, rs1003041423, rs3118419, rs5774935, rs150459448, rs1555845, rs1555846, rs11269074, rs3790469, rs3125894, rs3125895, rs3125896, rs3125897, rs3125898, rs17130688, rs938759267, rs34194247, rs3125900, rs79716012, rs75711879, rs12145904, rs3125902, rs3118420, rs12138573, rs1925955, rs17130691, rs3125904, rs78507000, rs150774295, rs3125905, rs2038900, rs2038901, rs147665807, rs17130694, rs12408546, rs12077372, rs3790472, rs3790473, rs147893529, rs2012235, rs3118423, rs2986125, rs2986124, rs72926973, rs2277874, rs3125906, rs3118426, rs3125907, rs382422, rs3118427, rs3125908, rs12759602, rs2477974, rs3125909, rs3118428, rs12407140, rs72674322, rs72674323, rs3125910, and rs1318744874.


In some embodiments, the guide sequence portion of the first RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 that targets a SNP position of the mutant allele.


In some embodiments, the SNP position is in an exon or intron of the RPE65 mutant allele.


In some embodiments, the SNP position contains a heterozygous SNP.


In some embodiments, the method further comprises introducing to the cell a second RNA molecule comprising a guide sequence portion having 17-25 nucleotides or a nucleotide sequence encoding the same, wherein a complex of the second RNA molecule and a CRISPR nuclease affects a second double strand break in the RPE65 gene.


In some embodiments, the guide sequence portion of the second RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 other than the sequence of the first RNA molecule.


In some embodiments, the second RNA molecule comprises a non-discriminatory guide portion that targets both functional and mutated RPE65 alleles.


In some embodiments, the second RNA molecule comprises a non-discriminatory guide portion that targets any one of Intron 1 of RPE65, Intron 2 of RPE65, a 3′ untranslated region (3′ UTR) of RPE65, and an intergenic region downstream of RPE65.


In some embodiments, the second RNA molecule comprises a non-discriminatory guide portion that targets a sequence that is located within a genomic range selected from any one of 1:68450655-1:68451154, 1:68428322-1:68428821, 1:68437687-1:68438186, 1:68431586-1:68432085, 1:68431377-1:68431469, 1:68431177-1:68431281, 1:68430565-1:68431064, 1:68429928-1:68430427, 1:68448707-1:68449206, 1:68449395-1:68449894, 1:68448124-1:68448623, 1:68446861-1:68447360, 1:68446210-1:68446709, 1:68444884-1:68445383, 1:68444673-1:68444775, 1:68444031-1:68444530, 1:68441001-1:68441500, 1:68440353-1:68440852, 1:68439643-1:68440142, 1:68439324-1:68439560, 1:68439082-1:68439190, and 1:68438317-1:68438941.


In some embodiments, the second RNA molecule comprises a non-discriminatory guide portion that targets a sequence that is located up to 500 base pairs from an exon that is excised by the first and second RNA molecules.


In some embodiments, a portion of an exon is excised from the mutant allele of the RPE65 gene.


In some embodiments, the first RNA molecule targets a SNP position in the 3′ UTR of the mutated allele, and the second RNA molecule comprises a non-discriminatory guide portion that targets downstream of a polyadenylation signal sequence that is common to both a functional allele and the mutant allele of the RPE65 gene.


In some embodiments, the first RNA molecule targets a SNP position downstream of a polyadenylation signal of the mutated allele, and the second RNA molecule comprises a non-discriminatory guide portion that targets a sequence upstream of a polyadenylation signal that is common to both a functional allele and the mutant allele of the RPE65 gene.


In some embodiments, the polyadenylation signal is excised from the mutant allele of the RPE65 gene.


According to embodiments of the present invention, there is provided a modified cell obtained by the method of any one of the embodiments presented herein.


According to embodiments of the present invention, there is provided a first RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516.


According to embodiments of the present invention, there is provided a composition comprising the first RNA molecule and at least one CRISPR nuclease.


In some embodiments, the composition further comprises a second RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides, wherein the second RNA molecule targets a RPE65 allele, and wherein the guide sequence portion of the second RNA molecule is a different sequence from the sequence of the guide sequence portion of the first RNA molecule.


In some embodiments, the guide sequence portion of the second RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 other than the sequence of the first RNA molecule.


According to embodiments of the present invention, there is provided a method for inactivating a mutant RPE65 allele in a cell, the method comprising delivering to the cell the composition of any one of the embodiments presented herein.


According to embodiments of the present invention, there is provided a method for treating a dominant RPE65 gene disorder, the method comprising delivering to a cell of a subject having a dominant RPE65 gene disorder the composition of any one of the embodiments presented herein.


According to embodiments of the present invention, there is provided use of any one of the compositions presented herein for inactivating a mutant RPE65 allele in a cell, comprising delivering to the cell the composition of any one of the embodiments presented herein.


According to embodiments of the present invention, there is provided a medicament comprising the composition of any one of the embodiments presented herein for use in inactivating a mutant RPE65 allele in a cell, wherein the medicament is administered by delivering to the cell the composition of any one of the embodiments presented herein.


According to embodiments of the present invention, there is provided use of the composition of any one of the embodiments presented herein for treating ameliorating or preventing a dominant RPE65 gene disorder, comprising delivering to a cell of a subject having or at risk of having a dominant RPE65 gene disorder the composition of any one of the embodiments presented herein.


According to embodiments of the present invention, there is provided a medicament comprising the composition of any one of the embodiments presented herein for use in treating ameliorating or preventing a dominant RPE65 gene disorder, wherein the medicament is administered by delivering to a cell of a subject having or at risk of having a dominant RPE65 gene disorder the composition of any one of the embodiments presented herein.


According to embodiments of the present invention, there is provided a kit for inactivating a mutant RPE65 allele in a cell, comprising an RNA molecule of any one of the embodiments presented herein, a CRISPR nuclease, and/or a tracrRNA molecule; and instructions for delivering the RNA molecule; CRISPR nuclease, and/or the tracrRNA to the cell.


According to embodiments of the present invention, there is provided a kit for treating a dominant RPE65 gene disorder in a subject, comprising an RNA molecule of any one of the embodiments presented herein, a CRISPR nuclease, and/or a tracrRNA molecule; and instructions for delivering the RNA molecule; CRISPR nuclease, and/or the tracrRNA to a cell of a subject having or at risk of having a dominant RPE65 gene disorder.


According to embodiments of the present invention, there is provided a gene editing composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516. In some embodiments, the RNA molecule further comprises a portion having a sequence which binds to a CRISPR nuclease. In some embodiments, the sequence which binds to a CRISPR nuclease is a tracrRNA sequence.


In some embodiments, the RNA molecule further comprises a portion having a tracr mate sequence.


In some embodiments, the RNA molecule may further comprise one or more linker portions.


According to embodiments of the present invention, an RNA molecule may be up to 300, 290, 280, 270, 260, 250, 240, 230, 220, 210, 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, or 100 nucleotides in length. Each possibility represents a separate embodiment. In embodiments of the present invention, the RNA molecule may be 17 up to 300 nucleotides in length, 100 up to 300 nucleotides in length, 150 up to 300 nucleotides in length, 200 up to 300 nucleotides in length, 100 to 200 nucleotides in length, or 150 up to 250 nucleotides in length. Each possibility represents a separate embodiment.


According to some embodiments of the present invention, the composition further comprises a tracrRNA molecule.


The present disclosure provides a method for utilizing at least one naturally occurring nucleotide difference or polymorphism (e.g., single nucleotide polymorphism (SNP)) for distinguishing/discriminating between two alleles of a gene, one allele bearing a mutation such that it encodes a mutated protein causing a disease phenotype (“mutated allele”) and a particular sequence in a SNP position (SNP/REF), and the other allele encoding for a functional protein (“functional allele”). The method further comprises the step of knocking out expression of the mutated protein and allowing expression of the functional protein. In some embodiments, the method is for treating, ameliorating, or preventing a dominant negative genetic disorder.


According to some embodiments of the present invention, there is provided a method for inactivating a mutant RPE65 allele in a cell, the method comprising delivering to the cell a composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease.


According to some embodiments of the present invention, there is provided a method for treating a dominant RPE65 gene disorder, the method comprising delivering to a cell of a subject having a dominant RPE65 gene disorder a composition comprising an RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 and a CRISPR nuclease.


According to embodiments of the present invention, the composition comprises a second RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516. In some embodiments, the 17-25 nucleotides of the guide sequence portion of the second RNA molecule are in a different sequence from the sequence of the guide sequence portion of the first RNA molecule.


According to embodiments of the present invention, at least one CRISPR nuclease and the RNA molecule or RNA molecules are delivered to the subject and/or cells substantially at the same time or at different times.


In some embodiments, a tracrRNA molecule is delivered to the subject and/or cells substantially at the same time or at different times as the CRISPR nuclease and RNA molecule or RNA molecules.


According to embodiments of the present invention, the first RNA molecule targets a SNP or disease-causing mutation in the exon or promoter of a mutated allele, and the second RNA molecule targets a SNP in an exon of the mutated allele, a SNP in an intron, or a sequence present in both the mutated or functional allele.


According to embodiments of the present invention, the first RNA molecule or the first and the second RNA molecules target a SNP in the promoter region, the start codon, or an untranslated region (UTR) of a mutated allele.


According to embodiments of the present invention, the first RNA molecule or the first and the second RNA molecules targets at least a portion of the promoter and/or the start codon and/or a portion of a UTR of a mutated allele.


According to embodiments of the present invention, the first RNA molecule targets a portion of the promoter, a first SNP in the promoter, or a SNP upstream to the promoter of a mutated allele and the second RNA molecule is targets a second SNP, which is downstream of the first SNP, and is in the promoter, in a UTR, or in an intron or in an exon of a mutated allele.


According to embodiments of the present invention, the first RNA molecule targets a SNP in the promoter, upstream of the promoter, or a UTR of a mutated allele and the second RNA molecule is designed to target a sequence which is present in an intron of both the mutated allele and the functional allele.


According to embodiments of the present invention, the first RNA molecule targets a SNP in an intron of a mutated allele, and wherein the second RNA molecule targets a SNP in an intron of the mutated allele, or a sequence in an intron present in both the mutated and functional allele.


According to embodiments of the present invention, the first RNA molecule targets a sequence upstream of the promotor which is present in both a mutated and functional allele and the second RNA molecule targets a SNP or disease-causing mutation in any location of the gene.


According to embodiments of the present invention, there is provided a method comprising removing an exon containing a disease-causing mutation from a mutated allele, wherein the first RNA molecule or the first and the second RNA molecules target regions flanking an entire exon or a portion of the exon.


According to embodiments of the present invention, there is provided a method comprising removing an exon or a portion thereof from a mutant RPE65 allele, the entire open reading frame of a mutant RPE65 allele, or removing the entire mutant RPE65 allele.


According to embodiments of the present invention, the first RNA molecule targets a SNP or disease-causing mutation in an exon or promoter of a mutated allele, and wherein the second RNA molecule targets a SNP in the same exon of the mutated allele, a SNP in an intron, or a sequence in an intron present in both the mutated and functional allele.


According to embodiments of the present invention, the first RNA molecule or the first and the second RNA molecules target an alternative splicing signal sequence between an exon and an intron of a mutant RPE65 allele.


According to embodiments of the present invention, the second RNA molecule is non-discriminatory targets a sequence present in both a mutated allele and a functional allele.


The compositions and methods of the present disclosure may be utilized for treating, preventing, ameliorating, or slowing progression of an autosomal dominant genetic disorder, such as a dominant RPE65 gene disorder.


In some embodiments, a mutated allele is deactivated by delivering to a cell an RNA molecule which targets a SNP in the promoter region, the start codon, or an untranslated region (UTR) of the mutated allele.


In some embodiments, a mutated allele is inactivated by removing at least a portion of the promoter, and/or removing the start codon, and/or a portion of the UTR, and/or a polyadenylation signal. In such embodiments one RNA molecule may be designed for targeting a first SNP in the promoter or upstream to the promoter and another RNA molecule is designed to target a second SNP, which is downstream of the first SNP, and is in the promoter, in the UTR, in an intron, or in an exon. Alternatively, one RNA molecule may be designed for targeting a SNP in the promoter, upstream of the promoter, or the UTR, and another RNA molecule is designed to target a sequence which is present in an intron of both the mutated allele and the functional allele.


Alternatively, one RNA molecule may be designed for targeting a sequence upstream of the promotor which is present in both the mutated and functional allele and the other guide is designed to target a SNP or disease-causing mutation in any location of the gene e.g., in an exon, intron, UTR, or downstream of the promoter.


In some embodiments, the method of deactivating a mutated allele comprises an exon skipping step comprising removing an exon containing a disease-causing mutation from the mutated allele. Removing an exon containing a disease-causing mutation in the mutated allele requires two RNA molecules which target regions flanking the entire exon or a portion of the exon. Removal of an exon containing the disease-causing mutation may be designed to eliminate the disease-causing action of the protein while allowing for expression of the remaining protein product which retains some or all of the wild-type activity. The entire open reading frame or the entire gene can be excised using two RNA molecules flanking the region desired to be excised.


In some embodiments, the method of deactivating a mutated allele comprises delivering two RNA molecules to a cell, wherein one RNA molecule targets a SNP or disease-causing mutation in an exon or promoter of the mutated allele, and wherein the other RNA molecule targets a SNP in the same of the mutated allele, a SNP in an intron, or a sequence in an intron present in both the mutated or functional allele.


Any one of, or combination of, the above-mentioned strategies for deactivating a mutant allele may be used in the context of the invention.


In embodiments of the present invention, an RNA molecule is used to direct a CRISPR nuclease to an exon or a splice site of a mutated allele in order to create a double-stranded break (DSB), leading to insertion or deletion of nucleotides by inducing an error-prone non-homologous end-joining (NHEJ) mechanism and formation of a frameshift mutation in the mutated allele. The frameshift mutation may result in, for example, inactivation or knockout of the mutated allele by generation of an early stop codon in the mutated allele and to generation of a truncated protein or to nonsense-mediated mRNA decay of the transcript of the mutant allele. In further embodiments, one RNA molecule is used to direct a CRISPR nuclease to a promotor of a mutated allele.


In some embodiments, the method of deactivating a mutated allele further comprises enhancing activity of the functional protein such as by providing a protein/peptide, a nucleic acid encoding a protein/peptide, or a small molecule such as a chemical compound, capable of activating/enhancing activity of the functional protein.


According to some embodiments, the present disclosure provides an RNA sequence (also referred to as an ‘RNA molecule’) which binds to or associates with and/or directs an RNA-guided DNA nuclease e.g., a CRISPR nuclease, to a target sequence comprising at least one nucleotide which differs between a mutated allele and a functional allele (e.g., SNP) of a gene of interest (i.e., a sequence of the mutated allele which is not present in the functional allele).


In some embodiments, the method comprises contacting a mutated allele of a gene of interest with an allele-specific RNA molecule and a CRISPR nuclease e.g., a Cas9 protein, wherein the allele-specific RNA molecule and the CRISPR nuclease associate with a nucleotide sequence of the mutated allele of the gene of interest which differs by at least one nucleotide from a nucleotide sequence of a functional allele of the gene of interest, thereby modifying or knocking-out the mutated allele.


In some embodiments, the allele-specific RNA molecule and a CRISPR nuclease is introduced to a cell encoding the gene of interest. In some embodiments, the cell encoding the gene of interest is in a mammalian subject. In some embodiments, the cell encoding the gene of interest is in a plant.


In some embodiments, the mutated allele is an allele of RPE65 gene. In some embodiments, the RNA molecule targets a SNP which co-exists with or is genetically linked to the mutated sequence associated with a dominant RPE65 gene disorder genetic disorder. In some embodiments, the RNA molecule targets a SNP which is highly prevalent in the population and exists in the mutated allele having the mutated sequence associated with a dominant RPE65 gene disorder genetic disorder and not in the functional allele of an individual subject to be treated. In some embodiments, a disease-causing mutation within a mutated RPE65 allele is targeted.


In some embodiments, the SNP is within an exon of the gene of interest. In such embodiments, a guide sequence portion of an RNA molecule is designed to associate with a sequence of an exon of the gene of interest.


In some embodiments, SNP is within an intron or the exon of the gene of interest. In some embodiments, the SNP is in close proximity to the splice site between an intron and an exon. In some embodiments, the close proximity to a splice site is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides upstream or downstream to the splice site. Each possibility represents a separate embodiment of the present invention. In such embodiments, a guide sequence portion of an RNA molecule may be designed to associate with a sequence of the gene of interest which comprises the splice site.


In some embodiments, the method is utilized for treating a subject having a disease phenotype resulting from the heterozygote RPE65 gene. In such embodiments, the method results in improvement, amelioration or prevention of the disease phenotype.


Embodiments of compositions described herein include at least one CRISPR nuclease, RNA molecule(s), and a tracrRNA molecule, being effective in a subject or cells at the same time. The at least one CRISPR nuclease, RNA molecule(s), and tracrRNA may be delivered substantially at the same time or can be delivered at different times but have effect at the same time. For example, this includes delivering the CRISPR nuclease to the subject or cells before the RNA molecule and/or tracrRNA is substantially extant in the subject or cells.


In some embodiments, the cell is a stem cell. In some embodiments, the stem cell is an autologous pluripotent stem cell or an induced pluripotent stem cell (iPSC). In some embodiments, the stem cell is differentiated into a retinal pigment epithelium cell. In some embodiments, the cell is a retinal pigment epithelium cell.


Dominant Genetic Disorders

One of skill in the art will appreciate that all subjects with any type of heterozygote genetic disorder (e.g., dominant genetic disorder) may be subjected to the methods described herein. In one embodiment, the present invention may be used to target a gene involved in, associated with, or causative of dominant genetic disorders such as, for example, a dominant RPE65 gene disorder. In some embodiments, the dominant genetic disorder is a dominant RPE65 gene disorder. In some embodiments, the target gene is the RPE65 gene. Non-limiting examples of mutations characterized as gain of function mutations associated with a dominant RPE65 gene disorder phenotype include chr:1:68431085(hg398) T to C (c.1430A>G; p.D477G) and chr1:68446825(hg38) G to A (c.130C>T; p.R44X).


RPE65 editing strategies include, but are not limited to, (1) truncation, for example, by targeting a RPE65 mutation or SNP position with one guide RNA molecule to induce a frameshift or nonsense-mediated decay; and (2) allele specific excision using two guide RNA molecules, for example, excision of at least one exon or a portion thereof, knockout of a large portion of the allele or the entire allele, or excision of the polyadenylation signal.


Truncation may be achieved by several approaches. For example, truncation may be achieved by targeting a SNP within a coding exon of a mutant RPE65 allele using a single guide RNA molecule (e.g. a single guide RNA molecule or “sgRNA”). Alternatively, excision may be achieved by targeting the mutant RPE65 allele with two different RNA molecules, with at least one RNA molecule preferably being allele-specific.


In another editing strategy, expression of a mutated RPE65 allele may be inhibited. An example of this strategy includes excising the polyadenylation signal in the 3′UTR region, which leads to an unstable transcript.


CRISPR Nucleases and PAM Recognition

In some embodiments, the sequence specific nuclease is selected from CRISPR nucleases, or a functional variant thereof. In some embodiments, the sequence specific nuclease is an RNA guided DNA nuclease. In such embodiments, the RNA sequence which guides the RNA guided DNA nuclease (e.g., Cpf1) binds to and/or directs the RNA guided DNA nuclease to the sequence comprising at least one nucleotide which differs between a mutated allele and its counterpart functional allele (e.g., SNP). In some embodiments, the CRISPR complex does not further comprise a tracrRNA. In a non-limiting example, in which the RNA guided DNA nuclease is a CRISPR protein, the at least one nucleotide which differs between the dominant mutated allele and the functional allele may be within the PAM site and/or proximal to the PAM site within the region that the RNA molecule is designed to hybridize to. A skilled artisan will appreciate that RNA molecules can be engineered to bind to a target of choice in a genome by commonly known methods in the art.


In embodiments of the present invention, a type II CRISPR system utilizes a mature crRNA:tracrRNA complex directs a CRISPR nuclease, e.g. Cas9, to the target DNA via Watson-Crick base-pairing between the crRNA and the protospacer on the target DNA next to the protospacer adjacent motif (PAM), an additional requirement for target recognition. The CRISPR nuclease then mediates cleavage of target DNA to create a double-stranded break within the protospacer. A skilled artisan will appreciate that each of the engineered RNA molecule of the present invention is further designed such as to associate with a target genomic DNA sequence of interest next to a protospacer adjacent motif (PAM), e.g., a PAM matching the sequence relevant for the type of CRISPR nuclease utilized, such as for a non-limiting example, NGG or NAG, wherein “N” is any nucleobase, for Streptococcus pyogenes Cas9 WT (SpCAS9); NNGRRT for Staphylococcus aureus (SaCas9); NNNVRYM for Jejuni Cas9 WT; NGAN or NGNG for SpCas9-VQR variant; NGCG for SpCas9-VRER variant; NGAG for SpCas9-EQR variant; NNNNGATT for Neisseria meningitidis (NmCas9); or TTV for Cpf1. RNA molecules of the present invention are each designed to form complexes in conjunction with one or more different CRISPR nucleases and designed to target polynucleotide sequences of interest utilizing one or more different PAM sequences respective to the CRISPR nuclease utilized.


In some embodiments, an RNA-guided DNA nuclease e.g., a CRISPR nuclease, may be used to cause a DNA break, either double or single-stranded in nature, at a desired location in the genome of a cell. The most commonly used RNA-guided DNA nucleases are derived from CRISPR systems, however, other RNA-guided DNA nucleases are also contemplated for use in the genome editing compositions and methods described herein. For instance, see U.S. Patent Publication No. 2015/0211023, incorporated herein by reference.


CRISPR systems that may be used in the practice of the invention vary greatly. CRISPR systems can be a type I, a type II, or a type III system. Non-limiting examples of suitable CRISPR proteins include Cas3, Cas4, Cas5, Cas5e (or CasD), Cas6, Cas6e, Cas6f, Cas7, Cas8a1, Cas8a2, Cas8b, Cas8c, Cas9, Casl0, Casl Od, CasF, CasG, CasH, Csy1, Csy2, Csy3, Cse1 (or CasA), Cse2 (or CasB), Cse3 (or CasE), Cse4 (or CasC), Csc1, Csc2, Csa5, Csn2, Csm2, Csm3, Csm4, Csm5, Csm6, Cmr1, Cmr3, Cmr4, Cmr5, Cmr6, Csb1, Csb2, Csb3,Csxl7, Csxl4, Csxl0, Csxl6, CsaX, Csx3, Cszl, Csxl5, Csfl, Csf2, Csf3, Csf4, and Cul966.


In some embodiments, the RNA-guided DNA nuclease is a CRISPR nuclease derived from a type II CRISPR system (e.g., Cas9). The CRISPR nuclease may be derived from Streptococcus pyogenes, Streptococcus thermophilus, Streptococcus sp., Staphylococcus aureus. Neisseria meningitidis, Treponema denticola, Nocardiopsis dassonvillei, Streptomyces pristinaespiralis, Streptomyces viridochromogenes, Streptomyces viridochromogenes, Streptosporangium roseum, Streptosporangium roseum, Alicyclobacillus acidocaldarius, Bacillus pseudomycoides, Bacillus selenitireducens, Exiguobacterium sibiricum, Lactobacillus delbrueckii, Lactobacillus salivarius, Microscilla marina, Burkholderiales bacterium, Polaromonas naphthalenivorans, Polaromonas sp., Crocosphaera watsonii, Cyanothece sp., Microcystis aeruginosa, Synechococcus sp., Acetohalobium arabaticum, Ammonmfex degensii, Caldicelulosiruptor becscii, Candidatus Desulforudis, Clostridium botulinum, Clostridium difjicile, Finegoldia magna, Natranaerobius thermophilus, Pelotomaculumthermopropionicum. Acidithiobacillus caldus, Acidithiobacillus ferrooxidans, Allochromatium vinosum, Marinobacter sp., Nitrosococcus halophilus, Nitrosococcus watsoni, Pseudoalteromonas haloplanktis, Ktedonobacter racemifer, Methanohalobium evestigatum, Anabaena variabilis, Nodularia spumigena, Nostoc sp., Arthrospira maxima, Arthrospira platensis, Arthrospira sp., Lyngbya sp., Microcoleus chthonoplastes, Oscillatoria sp., Petrotoga mobilis, Thermosipho africanus, Acaryochloris marina, or any species which encodes a CRISPR nuclease with a known PAM sequence. CRISPR nucleases encoded by uncultured bacteria may also be used in the context of the invention. (See Burstein et al. Nature, 2017). Variants of CRIPSR proteins having known PAM sequences e.g., SpCas9 D1135E variant, SpCas9 VQR variant, SpCas9 EQR variant, or SpCas9 VRER variant may also be used in the context of the invention.


Thus, an RNA guided DNA nuclease of a CRISPR system, such as a Cas9 protein or modified Cas9 or homolog or ortholog of Cas9, or other RNA guided DNA nucleases belonging to other types of CRISPR systems, such as Cpf1 and its homologs and orthologs, may be used in the compositions of the present invention.


In certain embodiments, the CRIPSR nuclease may be a “functional derivative” of a naturally occurring Cas protein. A “functional derivative” of a native sequence polypeptide is a compound having a qualitative biological property in common with a native sequence polypeptide. “Functional derivatives” include, but are not limited to, fragments of a native sequence and derivatives of a native sequence polypeptide and its fragments, provided that they have a biological activity in common with a corresponding native sequence polypeptide. A biological activity contemplated herein is the ability of the functional derivative to hydrolyze a DNA substrate into fragments. The term “derivative” encompasses both amino acid sequence variants of polypeptide, covalent modifications, and fusions thereof. Suitable derivatives of a Cas polypeptide or a fragment thereof include but are not limited to mutants, fusions, covalent modifications of Cas protein or a fragment thereof. Cas protein, which includes Cas protein or a fragment thereof, as well as derivatives of Cas protein or a fragment thereof, may be obtainable from a cell or synthesized chemically or by a combination of these two procedures. The cell may be a cell that naturally produces Cas protein, or a cell that naturally produces Cas protein and is genetically engineered to produce the endogenous Cas protein at a higher expression level or to produce a Cas protein from an exogenously introduced nucleic acid, which nucleic acid encodes a Cas that is same or different from the endogenous Cas. In some cases, the cell does not naturally produce Cas protein and is genetically engineered to produce a Cas protein.


In some embodiments, the CRISPR nuclease is Cpf1. Cpf1 is a single RNA-guided endonuclease which utilizes a T-rich protospacer-adjacent motif. Cpf1 cleaves DNA via a staggered DNA double-stranded break. Two Cpf1 enzymes from Acidaminococcus and Lachnospiraceae have been shown to carry out efficient genome-editing activity in human cells. (See Zetsche et al., 2015).


Thus, an RNA guided DNA nuclease of a Type II CRISPR System, such as a Cas9 protein or modified Cas9 or homologs, orthologues, or variants of Cas9, or other RNA guided DNA nucleases belonging to other types of CRISPR systems, such as Cpf1 and its homologs, orthologues, or variants, may be used in the present invention.


In some embodiments, the guide molecule comprises one or more chemical modifications which imparts a new or improved property (e.g., improved stability from degradation, improved hybridization energetics, or improved binding properties with an RNA guided DNA nuclease). Suitable chemical modifications include, but are not limited to: modified bases, modified sugar moieties, or modified inter-nucleoside linkages. Non-limiting examples of suitable chemical modifications include: 4-acetylcytidine, 5-(carboxyhydroxymethyl)uridine, 2′-O-methylcytidine, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluridine, dihydrouridine, 2′-O-methylpseudouridine, “beta, D-galactosylqueuosine”, 2′-O-methylguanosine, inosine, N6-isopentenyladenosine, 1-methyladenosine, 1-methylpseudouridine, 1-methylguanosine, 1-methylinosine, “2,2-dimethylguanosine”, 2-methyladenosine, 2-methylguanosine, 3-methylcytidine, 5-methylcytidine, N6-methyladenosine, 7-methylguanosine, 5-methylaminomethyluridine, 5-methoxyaminomethyl-2-thiouridine, “beta, D-mannosylqueuosine”, 5-methoxycarbonylmethyl-2-thiouridine, 5-methoxycarbonylmethyluridine, 5-methoxyuridine, 2-methylthio-N6-isopentenyladenosine, N-((9-beta-D-ribofuranosyl-2-methylthiopurine-6-yl)carbamoyl)threonine, N-((9-beta-D-ribofuranosylpurine-6-yl)N-methylcarbamoyl)threonine, uridine-5-oxyacetic acid-methylester, uridine-5-oxyacetic acid, wybutoxosine, queuosine, 2-thiocytidine, 5-methyl-2-thiouridine, 2-thiouridine, 4-thiouridine, 5-methyluridine, N-((9-beta-D-ribofuranosylpurine-6-yl)-carbamoyl)threonine, 2′-O-methyl-5-methyluridine, 2′-O-methyluridine, wybutosine, “3-(3-amino-3-carboxy-propyl)uridine, (acp3)u”, 2′-0-methyl (M), 3′-phosphorothioate (MS), 3-thioPACE (MSP), pseudouridine, or 1-methyl pseudo-uridine. Each possibility represents a separate embodiment of the present invention.


Guide Sequences which Specifically Target a Mutant Allele


A given gene may contain thousands of SNPs. Utilizing a twenty-five base pair target window for targeting each SNP in a gene would require hundreds of thousands of guide sequences. Any given guide sequence when utilized to target a SNP may result in degradation of the guide sequence, limited activity, no activity, or off-target effects. Accordingly, suitable guide sequences are necessary for targeting a given gene. By the present invention, a novel set of guide sequences have been identified for knocking out expression of a mutated RPE65 protein, inactivating a mutant RPE65 gene allele, and treating a dominant RPE65 gene disorder.


The present disclosure provides guide sequences capable of specifically targeting a mutated allele for inactivation while leaving the functional allele unmodified. The guide sequences of the present invention are designed to, and are most likely to, specifically differentiate between a mutated allele and a functional allele. Of all possible guide sequences which target a mutated allele desired to be inactivated, the specific guide sequences disclosed herein are specifically effective to function with the disclosed embodiments.


Briefly, the guide sequences may have properties as follows: (1) target SNP/insertion/deletion/indel with a high prevalence in the general population, in a specific ethnic population or in a patient population is above 1% and the SNP/insertion/deletion/indel heterozygosity rate in the same population is above 1%; (2) target a location of a SNP/insertion/deletion/indel proximal to a portion of the gene e.g., within 5 k bases of any portion of the gene, for example, a promoter, a UTR, an exon or an intron; and (3) target a mutant allele using an RNA molecule which targets a founder or common pathogenic mutations for the disease/gene. In some embodiments, the prevalence of the SNP/insertion/deletion/indel in the general population, in a specific ethnic population or in a patient population is above 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, or 15% and the SNP/insertion/deletion/indel heterozygosity rate in the same population is above 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, or 15%. Each possibility represents a separate embodiment and may be combined at will.


For each gene, according to SNP/insertion/deletion/indel any one of the following strategies may be used to deactivate the mutated allele: (1) Knockout strategy using one RNA molecule—one RNA molecule is utilized to direct a CRISPR nuclease to a mutated allele and create a double-strand break (DSB) leading to formation of a frameshift mutation in an exon or in a splice site region of the mutated allele; and (2) Excision of at least one coding exon or a complete knockout of a mutant RPE65 allele using two RNA molecules, for example, a first RNA molecule targets a SNP position of an Intron 1 of the mutant RPE65 allele and a second, non-discriminatory RNA molecule targets a sequence in Intron 2 of the RPE65 gene.


Based on the locations of identified SNPs/insertions/deletions/indels for each mutant allele, any one of, or a combination of, the above-mentioned methods to deactivate the mutant allele may be utilized.


In some embodiments of the present invention, an RNA molecule is used to target a pathogenic mutation within a mutant RPE65 allele. In some embodiments of the present invention, an RNA molecule is used to target a SNP position.


Guide sequences of the present invention may: (1) target a heterozygous SNP for the targeted gene; (2) target a heterozygous SNP upstream or downstream of the gene; (3) target a SNP with a prevalence of the SNP/insertion/deletion/indel in the general population, in a specific ethnic population, or in a patient population above 1%; (4) have a guanine-cytosine content of greater than 30% and less than 85%; (5) have no repeat of seven or more guanine, cytosine, uracil, or adenine; and (6) have low or no off-targeting identified by off-target analysis. Guide sequences of the present invention may satisfy any one of the above criteria and are most likely to differentiate between a mutated allele from its corresponding functional allele.


In some embodiments of the present invention, at least one nucleotide which differs between the mutated allele and the functional allele is upstream, downstream or within the sequence of the disease-causing mutation of the gene of interest. The at least one nucleotide which differs between the mutated allele and the functional allele may be within an exon or within an intron of the gene of interest. In some embodiments, the at least one nucleotide which differs between the mutated allele and the functional allele is within an exon of the gene of interest. In some embodiments, the at least one nucleotide which differs between the mutated allele and the functional allele is within an intron or the exon of the gene of interest, in close proximity to the splice site between the intron and the exon e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides upstream or downstream to the splice site.


In some embodiments, the at least one nucleotide is a single nucleotide polymorphism (SNP). In some embodiments, each of the nucleotide variants of the SNP may be expressed in the mutated allele. In some embodiments, the SNP may be a founder or common pathogenic mutation.


Guide sequences may target a SNP which has both (1) a high prevalence in the general population e.g., above 1% in the population; and (2) a high heterozygosity rate in the population, e.g., above 1%. Guide sequences may target a SNP that is globally distributed. A SNP may be a founder or common pathogenic mutation. In some embodiments, the prevalence in the general population is above 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, or 15%. Each possibility represents a separate embodiment. In some embodiments, the heterozygosity rate in the population is above 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, or 15%. Each possibility represents a separate embodiment.


In some embodiments, the at least one nucleotide which differs between the mutated allele and the functional allele is linked to/co-exists with the disease-causing mutation in high prevalence in a population. In such embodiments, “high prevalence” refers to at least 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%. Each possibility represents a separate embodiment of the present invention. In one embodiment, the at least one nucleotide which differs between the mutated allele and the functional allele, is a disease-associated mutation. In some embodiments, the SNP is highly prevalent in the population. In such embodiments, “highly prevalent” refers to at least 10%, 11%, 12%, 13%, 14%, 15%, 20%, 30%, 40%, 50%, 60%, or 70% of a population. Each possibility represents a separate embodiment of the present invention.


Delivery to Cells

The compositions described herein may be delivered to a target cell by any suitable means. Compositions of the present invention may be targeted to any cell which contains and/or expresses a mutated allele, including any mammalian cell, for example a retinal pigment epithelium (RPE) cell. For example, in one embodiment an RNA molecule of the present invention that specifically targets a mutated RPE65 allele is delivered to a target cell and the target cell is a stem cell or a retinal pigment epithelium cell. The delivery to the cell may be performed in-vitro, ex-vivo, or in-vivo. Further, the compositions described herein may comprise any one or more of a DNA molecule, an RNA molecule, a ribonucleoprotein (RNP), a nucleic acid vector, or any combination thereof. In some embodiments, the composition is a naked DNA plasmid. In some embodiments, the composition is a naked RNA. In some embodiments, the composition is an RNP. An RNP composition may be conjugated to a cell-penetrating peptide (CPP), an antibody, a targeting moiety, or any combination thereof.


In some embodiments, the composition is packaged into an adeno-associated virus (AAV). In some embodiments, the composition is packaged into a lentivirus, such as a non-integrating lentivirus or a lentivirus lacking reverse transcription capability. In some embodiments, the composition is packaged into liposomes, extracellular vesicles, or exosomes, which may be pseudotyped with vesicular stomatitis glycoprotein (VSVG) or conjugated to a cell-penetrating peptide, an antibody, a targeting moiety, or any combination thereof.


In preferred embodiments, the composition is delivered in-vivo to retinal pigment epithelium cells within the eye of a subject. The in-vivo delivery to an eye of a subject my occur by subretinal injection, suprachoroidal injection, or injection to the interior chamber of the eye. The injected composition may be packaged in adeno-associated virus (AAV), lentivirus, preferably a non-integrating lentivirus, liposomes, extracellular vesicles, or exosomes. In some embodiments, the injected exosome may be pseudotyped with vesicular stomatitis glycoprotein (VSVG) or conjugated to a cell-penetrating peptide, an antibody, a targeting moiety, or any combination thereof.


In other embodiments, the composition is delivered to a cell ex-vivo. In some embodiments, the cell is a stem cell. In some embodiments, the stem cell is an autologous pluripotent stem cell or an induced pluripotent stem cell (iPSC). In some embodiments, the stem cell is differentiated into a retinal pigment epithelium cell. In some embodiments, the cell is a retinal pigment epithelium cell. The composition may be delivered to the cell by any known ex-vivo delivery method, including but not limited to, electroporation, viral transduction, nanoparticle delivery, liposomes, exosomes etc. Upon ex-vivo delivery of the composition to a cell, the cell may be introduced into the eye of a subject. In one example, the composition is delivered ex-vivo to iPSCs or IPSC-derived retinal pigment epithelium cells expanded into a patch or a tissue that is to be surgically reintroduced to the eye (See Sharma et al. 2019). Additional detailed delivery methods are described throughout this section.


In some embodiments, the RNA molecule comprises a chemical modification. Non-limiting examples of suitable chemical modifications include 2′-O-methyl (M). 2′-O-methyl. 3′phosphorothioate (MS) or 2′-O-methyl, 3′thioPACE (MSP), pseudouridine, and 1-methyl pseudo-uridine. Each possibility represents a separate embodiment of the present invention.


Any suitable viral vector system may be used to deliver nucleic acid compositions e.g., the compositions of the subject invention. Conventional viral and non-viral based gene transfer methods can be used to introduce nucleic acids and target tissues. In certain embodiments, nucleic acids are administered for in vivo or ex vivo gene therapy uses. Non-viral vector delivery systems include naked nucleic acid, and nucleic acid complexed with a delivery vehicle such as a liposome or poloxamer. For a review of gene therapy procedures, see Anderson (1992); Nabel & Felgner (1993); Mitani & Caskey (1993); Dillon (1993); Miller (1992); Van Brunt (1988); Vigne (1995); Kremer & Perricaudet (1995); Haddada et al. (1995); and Yu et al. (1994).


Methods of non-viral delivery of nucleic acids and/or proteins include electroporation, lipofection, microinjection, biolistics, particle gun acceleration, virosomes, liposomes, immunoliposomes, lipid nanoparticles (LNPs), polycation or lipid:nucleic acid conjugates, artificial virions, and agent-enhanced uptake of nucleic acids or can be delivered to plant cells by bacteria or viruses (e.g., Agrobacterium, Rhizobium sp. NGR234, Sinorhizoboiummeliloti, Mesorhizobium loti, tobacco mosaic virus, potato virus X, cauliflower mosaic virus and cassava vein mosaic virus). (See, e.g., Chung et al., 2006). Sonoporation using, e.g., the Sonitron 2000 system (Rich-Mar), can also be used for delivery of nucleic acids. Cationic-lipid mediated delivery of proteins and/or nucleic acids is also contemplated as an in vivo, ex vivo, or in vitro delivery method. (See Zuris et al. (2015); see also Coelho et al. (2013); Judge et al. (2006); and Basha et al. (2011)).


Additional exemplary nucleic acid delivery systems include those provided by Amaxa® Biosystems (Cologne, Germany), Maxcyte, Inc. (Rockville, Md.), BTX Molecular Delivery Systems (Holliston, Mass.) and Copernicus Therapeutics Inc., (see, e.g., U.S. Pat. No. 6,008,336). Lipofection is described in e.g., U.S. Pat. Nos. 5,049,386, 4,946,787; and 4,897,355, and lipofection reagents are sold commercially (e.g., Transfectam™, Lipofectin™ and Lipofectamine™ RNAiMAX). Cationic and neutral lipids that are suitable for efficient receptor-recognition lipofection of polynucleotides include those disclosed in PCT International Publication Nos. WO/1991/017424 and WO/1991/016024. Delivery can be to cells (ex vivo administration) or target tissues (in vivo administration).


The preparation of lipid:nucleic acid complexes, including targeted liposomes such as immunolipid complexes, is well known to one of skill in the art (see, e.g., Crystal, Science (1995); Blaese et al., (1995); Behr et al., (1994); Remy et al. (1994); Gao and Huang (1995); Ahmad and Allen (1992); U.S. Pat. Nos. 4,186,183; 4,217,344; 4,235,871; 4,261,975; 4,485,054; 4,501,728; 4,774,085; 4,837,028; and 4,946,787).


Additional methods of delivery include the use of packaging the nucleic acids to be delivered into EnGeneIC delivery vehicles (EDVs). These EDVs are specifically delivered to target tissues using bispecific antibodies where one arm of the antibody has specificity for the target tissue and the other has specificity for the EDV. The antibody brings the EDVs to the target cell surface and then the EDV is brought into the cell by endocytosis. Once in the cell, the contents are released (See MacDiarmid et al., 2009).


The use of RNA or DNA viral based systems for viral mediated delivery of nucleic acids take advantage of highly evolved processes for targeting a virus to specific cells in the body and trafficking the viral payload to the nucleus. Viral vectors can be administered directly to patients (in vivo) or they can be used to treat cells in vitro and the modified cells are administered to patients (ex vivo). Conventional viral based systems for the delivery of nucleic acids include, but are not limited to, retroviral, lentivirus, adenoviral, adeno-associated, vaccinia and herpes simplex virus vectors for gene transfer.


The tropism of a retrovirus can be altered by incorporating foreign envelope proteins, expanding the potential target population of target cells. Lentiviral vectors are retroviral vectors that are able to transduce or infect non-dividing cells and typically produce high viral titers. Selection of a retroviral gene transfer system depends on the target tissue. Retroviral vectors are comprised of cis-acting long terminal repeats with packaging capacity for up to 6-10 kb of foreign sequence. The minimum cis-acting LTRs are sufficient for replication and packaging of the vectors, which are then used to integrate the therapeutic gene into the target cell to provide permanent transgene expression. Widely used retroviral vectors include those based upon murine leukemia virus (MuLV), gibbon ape leukemia virus (GaLV), Simian Immunodeficiency virus (SIV), human immunodeficiency virus (HIV), and combinations thereof (See, e.g., Buchschacher et al. (1992); Johann et al. (1992); Sommerfelt et al. (1990); Wilson et al. (1989); Miller et al. (1991); PCT International Publication No. WO/1994/026877A1).


At least six viral vector approaches are currently available for gene transfer in clinical trials, which utilize approaches that involve complementation of defective vectors by genes inserted into helper cell lines to generate the transducing agent.


pLASN and MFG-S are examples of retroviral vectors that have been used in clinical trials (See Dunbar et al., 1995; Kohn et al., 1995; Malech et al., 1997). PA317/pLASN was the first therapeutic vector used in a gene therapy trial (Blaese et al., 1995). Transduction efficiencies of 50% or greater have been observed for MFG-S packaged vectors. (Ellem et al., (1997); Dranoff et al., 1997).


Packaging cells are used to form virus particles that are capable of infecting a host cell. Such cells include 293 cells, which package adenovirus, AAV, and Psi-2 cells or PA317 cells, which package retrovirus. Viral vectors used in gene therapy are usually generated by a producer cell line that packages a nucleic acid vector into a viral particle. The vectors typically contain the minimal viral sequences required for packaging and subsequent integration into a host (if applicable), other viral sequences being replaced by an expression cassette encoding the protein to be expressed. The missing viral functions are supplied in trans by the packaging cell line. For example, AAV vectors used in gene therapy typically only possess inverted terminal repeat (ITR) sequences from the AAV genome which are required for packaging and integration into the host genome. Viral DNA is packaged in a cell line, which contains a helper plasmid encoding the other AAV genes, namely rep and cap, but lacking ITR sequences. The cell line is also infected with adenovirus as a helper. The helper virus promotes replication of the AAV vector and expression of AAV genes from the helper plasmid. The helper plasmid is not packaged in significant amounts due to a lack of ITR sequences. Contamination with adenovirus can be reduced by, e.g., heat treatment to which adenovirus is more sensitive than AAV. Additionally, AAV can be produced at clinical scale using baculovirus systems (see U.S. Pat. No. 7,479,554).


In many gene therapy applications, it is desirable that the gene therapy vector be delivered with a high degree of specificity to a particular tissue type. Accordingly, a viral vector can be modified to have specificity for a given cell type by expressing a ligand as a fusion protein with a viral coat protein on the outer surface of the virus. The ligand is chosen to have affinity for a receptor known to be present on the cell type of interest. For example, Han et al. (1995) reported that Moloney murine leukemia virus can be modified to express human heregulin fused to gp70, and the recombinant virus infects certain human breast cancer cells expressing human epidermal growth factor receptor. This principle can be extended to other virus-target cell pairs, in which the target cell expresses a receptor and the virus expresses a fusion protein comprising a ligand for the cell-surface receptor. For example, filamentous phage can be engineered to display antibody fragments (e.g., FAB or Fv) having specific binding affinity for virtually any chosen cellular receptor. Although the above description applies primarily to viral vectors, the same principles can be applied to nonviral vectors. Such vectors can be engineered to contain specific uptake sequences which favor uptake by specific target cells.


Gene therapy vectors can be delivered in vivo by administration to an individual patient, typically by systemic administration (e.g., intravitreal, intravenous, intraperitoneal, intramuscular, subdermal, or intracranial infusion) or topical application, as described below. Alternatively, vectors can be delivered to cells ex vivo, such as cells explanted from an individual patient (e.g., lymphocytes, bone marrow aspirates, tissue biopsy) or universal donor hematopoietic stem cells, followed by reimplantation of the cells into a patient, optionally after selection for cells which have incorporated the vector. A non-limiting exemplary ex vivo approach may involve removal of tissue (e.g., peripheral blood, bone marrow, and spleen) from a patient for culture, nucleic acid transfer to the cultured cells (e.g., hematopoietic stem cells), followed by grafting the cells to a target tissue (e.g., bone marrow, and spleen) of the patient. In some embodiments, the stem cell or hematopoietic stem cell may be further treated with a viability enhancer.


Ex-vivo cell transfection for diagnostics, research, or for gene therapy (e.g., via re-infusion of the transfected cells into the host organism) is well known to those of skill in the art. In a preferred embodiment, cells are isolated from the subject organism, transfected with a nucleic acid composition, and re-infused back into the subject organism (e.g., patient). Various cell types suitable for ex vivo transfection are well known to those of skill in the art (See, e.g., Freshney, “Culture of Animal Cells, A Manual of Basic Technique and Specialized Applications (6th edition, 2010) and the references cited therein for a discussion of how to isolate and culture cells from patients).


Suitable cells include, but are not limited to, eukaryotic cells and/or cell lines. Non-limiting examples of such cells or cell lines generated from such cells include COS, CHO (e.g., CHO—S, CHO-K1, CHO-DG44, CHO-DUXB11, CHO-DUKX, CHOKISV), VERO, MDCK, WI38, V79, B14AF28-G3, BHK, HaK, NSO, SP2/0-Ag14, HeLa, HEK293 (e.g., HEK293-F, HEK293-H, HEK293-T), perC6 cells, any plant cell (differentiated or undifferentiated), as well as insect cells such as Spodopterafugiperda (Sf), or fungal cells such as Saccharomyces, Pichia and Schizosaccharomyces. In certain embodiments, the cell line is a CHO-K1, MDCK or HEK293 cell line. Additionally, primary cells may be isolated and used ex vivo for reintroduction into the subject to be treated following treatment with a guided nuclease system (e.g. CRISPR/Cas). Suitable primary cells include peripheral blood mononuclear cells (PBMC), and other blood cell subsets such as, but not limited to, CD4+ T cells or CD8+ T cells. Suitable cells also include stem cells such as, by way of example, embryonic stem cells, induced pluripotent stem cells, hematopoietic stem cells (CD34+), neuronal stem cells and mesenchymal stem cells.


In one embodiment, stem cells are used in ex vivo procedures for cell transfection and gene therapy. The advantage to using stem cells is that they can be differentiated into other cell types in vitro, or can be introduced into a mammal (such as the donor of the cells) where they will engraft in the bone marrow. Methods for differentiating CD34+ cells in vitro into clinically important immune cell types using cytokines such a GM-CSF, IFN-gamma, and TNF-alpha are known (as a non-limiting example see, Inaba et al., 1992).


Stem cells are isolated for transduction and differentiation using known methods. For example, stem cells are isolated from bone marrow cells by panning the bone marrow cells with antibodies which bind unwanted cells, such as CD4+ and CD8+ (T cells), CD45+ (panB cells), GR-1 (granulocytes), and lad (differentiated antigen presenting cells) (as a non-limiting example, see Inaba et al., 1992). Stem cells that have been modified may also be used in some embodiments.


Vectors (e.g., retroviruses, liposomes, etc.) containing therapeutic nucleic acid compositions can also be administered directly to an organism for transduction of cells in vivo. Administration is by any of the routes normally used for introducing a molecule into ultimate contact with blood or tissue cells including, but not limited to, injection, infusion, topical application (e.g., eye drops and cream) and electroporation. Suitable methods of administering such nucleic acids are available and well known to those of skill in the art, and, although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route. According to some embodiments, the composition is delivered via IV injection.


Vectors suitable for introduction of transgenes into immune cells (e.g., T-cells) include non-integrating lentivirus vectors. See, e.g., U.S. Patent Publication No. 2009/0117617.


Pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions available, as described below (See, e.g., Remington's Pharmaceutical Sciences, 17th ed., 1989).


In accordance with some embodiments, there is provided an RNA molecule which binds to/associates with and/or directs the RNA guided DNA nuclease to a sequence comprising at least one nucleotide which differs between a mutated allele and a functional allele (e.g., SNP) of a gene of interest (i.e., a sequence of the mutated allele which is not present in the functional allele). The sequence may be within the disease associated mutation. The sequence may be upstream or downstream to the disease associated mutation. Any sequence difference between the mutated allele and the functional allele may be targeted by an RNA molecule of the present invention to inactivate the mutant allele, or otherwise disable its dominant disease-causing effects, while preserving the activity of the functional allele.


The disclosed compositions and methods may also be used in the manufacture of a medicament for treating dominant genetic disorders in a patient.


Mechanisms of Action for RPE65 Knockout Methods

Without being bound by any theory or mechanism, the instant invention may be utilized to apply a CRISPR nuclease to process a mutated pathogenic RPE65 allele and not a functional RPE65 allele, such as to prevent expression of the mutated pathogenic allele or to produce a truncated non-pathogenic peptide from the mutated pathogenic allele, in order to prevent or treat a dominant RPE65 gene disorder. A specific guide sequence may be selected from Table 1 based on the targeted SNP position and the type of CRISPR nuclease used (e.g. according to a required PAM sequence).


The RPE65 gene is located in chromosome 1 and encodes the retinal pigment epithelium-specific 65 kDa protein. Editing strategies for RPE65 include (1) truncation strategies requiring only one guide; (2) truncation strategies using two guides; (3) knockout strategies using two guides; and (4) two guide strategies using a first RNA guide specifically targeting a pathogenic mutation in Exon 13 (i.e. one which leads to Asp477Gly) and a second, non-discriminatory RNA guide.


An example of a truncation strategy requiring only one guide RNA molecule includes targeting a pathogenic mutation in order to mediate truncation or nonsense mediated decay (NMD) of an RPE65 mutant allele. As a non-limiting example, a frameshift in a mutated RPE65 allele may be introduced by utilizing one RNA molecule to target a pathogenic mutation in a coding exon of the mutated RPE65 allele in order to mediate a double-strand break, which leads to generation of a frameshift mutation and expression of a truncated protein or nonsense mediated decay (NMD) of its transcripts.


An example of a truncation strategy using two guides includes excision of any one of Exons 5, 6, 9, or 10 by targeting RNA molecules to flanking regions of the exons. One of the two guides must specifically target a mutated RPE65 allele over a functional RPE65 allele, for example, by targeting a SNP position.


Examples of a knockout strategy using two guides include multiple approaches. In one approach, knockout of an RPE65 mutant allele may be achieved by excision of Exon 1 (including the 5′UTR and ORF). Exon 1 may be excised by utilizing SNP positions in Intron 1 or upstream to the promoter region. Only one of the two guides needs to be a discriminatory guide. For example, Exon 1 may be excised by targeting a first RNA molecule to a SNP position in Intron 1 and a second, non-discriminatory RNA molecule targeting a region upstream to the promoter region. Alternatively, Exon 1 may be excised by targeting a first RNA molecule to a SNP position in a region upstream to the promoter region and a second, non-discriminatory RNA molecule targeting Intron 1.


In another approach using two guides, knockout of an RPE65 mutant may be achieved by excision of Exon 2. Exon 2 may be excised by utilizing SNP positions in Intron 1 or Intron 2, however only one of the two guides needs to be a discriminatory guide. Exon 2 excision in this manner generates a peptide of only 23 amino acids.


In yet another approach using two guides, knockout of an RPE65 mutant may be achieved by excision of Exon 14. Exon 14 may be excised by utilizing a SNP position downstream of Exon 14 or in Intron 13. However only one of the two guides needs to be a discriminatory guide. Exon 14 excision in this manner would eliminate the 3′UTR and thereby destabilize the transcript.


Examples of two-guide strategies using a first RNA guide specifically targeting a pathogenic mutation in Exon 13 of RPE65 (i.e. one which leads to Asp477Gly) and a second, non-discriminatory RNA guide include multiple approaches.


For example, in one approach excision from Exon 11 to a pathogenic mutation in Exon 13 is carried out. To facilitate the excision, an allele specific cut is mediated by targeting a first RNA molecule to a pathogenic mutation in Exon 13, and a biallelic cut is mediated by targeting a second, non-discriminatory RNA molecule to Intron 10. Intron 10 is preferably targeted since Intron 11 and Intron 12 are very short and therefore targeting them might cause legions that would be deleterious to a functional RPE65 allele, as well.


In another approach, excision from a pathogenic mutation in Exon 13 to Intron 13 is carried out. To facilitate the excision, an allele specific cut is mediated by targeting a first RNA molecule to a pathogenic mutation in Exon 13, and a biallelic cut is mediated by targeting a second, non-discriminatory RNA molecule to Intron 13. This approach would lead to the elimination of the splice donor at the end of Exon 13. This approach would lead to nonsense-mediated decay (NMD) or to the extension of Exon 13, which would contain stop codons and result in expression of a truncated protein.


In yet another approach, excision from a pathogenic mutation in Exon 13 to the 3′UTR is carried out. To facilitate the excision, an allele specific cut is mediated by targeting a first RNA molecule to a pathogenic mutation in Exon 13, and a biallelic cut is mediated by targeting a second, non-discriminatory RNA downstream to the 3′UTR in Exon 14. An excision performed using this approach would destabilize the transcript of the mutant RPE65 allele.


Examples of RNA Guide Sequences which Specifically Target Mutated Alleles of RPE65 Gene

Although a large number of guide sequences can be designed to target a mutated allele, the nucleotide sequences described in Table 1 identified by SEQ ID NOs: 1-49516 below were specifically selected to effectively implement the methods set forth herein and to effectively discriminate between alleles.


Table 1 shows guide sequences designed for use as described in the embodiments above to associate with different SNPs or pathogenic mutations within a sequence of a mutated RPE65 allele. Each engineered guide molecule is further designed such as to associate with a target genomic DNA sequence of interest that lies next to a protospacer adjacent motif (PAM), e.g., a PAM matching the sequence NGG or NAG, where “N” is any nucleobase. The guide sequences were designed to work in conjunction with one or more different CRISPR nucleases, including, but not limited to, e.g. SpCas9WT (PAM SEQ: NGG), SpCas9.VQR.1 (PAM SEQ: NGAN), SpCas9.VQR2 (PAM SEQ: NGNG), SpCas9.EQR (PAM SEQ: NGAG), SpCas9.VRER (PAM SEQ: NGCG), SaCas9WT (PAM SEQ: NNGRRT), NmCas9WT (PAM SEQ: NNNNGATT), Cpf1 (PAM SEQ: TITV), or JeCas9WT (PAM SEQ: NNNVRYM). RNA molecules of the present invention are each designed to form complexes in conjunction with one or more different CRISPR nucleases and designed to target polynucleotide sequences of interest utilizing one or more different PAM sequences respective to the CRISPR nuclease utilized.









TABLE 1







Guide sequences designed to associate with specific RPE65 gene targets











SEQ ID NOs:
SEQ ID NOs:
SEQ ID NOs:



of 20 base
of 21 base
of 22 base


Target
guides
guides
guides





1:68431085_T_C
1-46
47-94
95-144


1:68446825_G_A
145-190
191-238
239-288


1:68425933_C_CAT
289-302
303-311
312-324


rs60701104_REF





1:68426291_T_C

325-326
327-330


rs9436400_SNP





1:68426379_TA_T
331-347
348-364
365-381


rs868541802_REF





1:68426406_C_T
382-389
390-393
394-399


rs3125890_REF





1:68426406_C_T
400-401




rs3125890_SNP





1:68426632_A_G
402-414
415-432
433-456


rs75159457_REF





1:68426632_A_G
403 405 407
417-418 420
435 437-438


rs75159457_SNP
411 457-488
428-429 489-
441 452-453




523
524-561


1:68427338_GT_G
562-574
575-587
588-602


rs1205919238_REF





1:68427338_GT_G
603

604


rs1205919238_SNP





1:68427665_T_A
605-608
609-612
613-619


rs11209300_REF





1:68427665_T_A
605-608
609-612
613-618 620


rs11209300_SNP





1:68428071_G_A
621-666
667-714
715-764


rs4264030_REF





1:68428071_G_A
627 632 636
673 678 682
721 726 730


rs4264030_SNP
640 648 653
686 695 703
732 735 737



656 662 765-
709 711 800-
753 761 831-



799
830
870


1:68428149_T_C
871-914
915-957
958-1003


rs2419988_REF





1:68428149_T_C
875 885 891-
919 921 935
964 973 979-


rs2419988_SNP
892 894 914
938 1042-1081
980 983 1082-



1004-1041

1123


1:68428613_T_C
1124-1132
1133-1134
1135-1140


rs3118415_REF





1:68428613_T_C
1124-1125 1127
1133 1160-1170
1139 1171-1185


rs3118415_SNP
1130 1141-1159




1:68428861_G_C
1186-1208
1209-1216
1217-1226


rs3118416_REF





1:68428861_G_C
1227-1249
1250-1257
1258-1267


rs3118416_SNP





1:68429047_G_GCT
1268-1279




rs149739986_SNP





1:68429165_C_T
1280-1294
1295-1307
1308-1322


rs2182315_REF





1:68429165_C_T
1281-1282
1295 1301-1302
1309 1315 1320


rs2182315_SNP
1288-1289
1306 1330-1336
1322 1337-1345



1323-1329




1:68429245_T_C
1346-1372
1373-1393
1394-1418


rs3118418_REF





1:68429245_T_C
1358 1361 1364
1382 1385
1408 1415


rs3118418_SNP
1370 1419-1447
1392-1393
1417-1418




1448-1472
1473-1501


1:68430961_G_A
1502-1529
1530-1543
1544-1561


rs932783_REF





1:68430961_G_A
1517 1562-1571
1572-1575
1576-1581


rs932783_SNP





1:68432081_G_A
1582-1600
1601-1619
1620-1638


rs12124063_REF





1:68432081_G_A
1592 1595
1611 1613 1615
1628 1630 1634


rs12124063_SNP
1599-1600
1618 1654-1668
1637 1669-1683



1639-1653




1:68432262_C_G
1684-1729
1730-1777
1778-1827


rs77585943_REF





1:68432262_C_G
1685 1687
1731 1733 1736
1781 1784 1792


rs77585943_SNP
1695-1696 1703
1743 1750 1755
1797 1800 1805



1708 1712 1725
1759 1765
1809 1815



1828-1865
1866-1905
1906-1947


1:68433374_G_A
1948-1993
1994-2041
2042-2091


rs1886906_REF





1:68433374_G_A
1956-1957 1971
2002-2003 2010
2048 2051-2052


rs1886906_SNP
1975 1977 1980
2019 2023 2025
2059 2068 2074



1982 1992
2030 2040
2079 2082



2092-2129
2130-2169
2170-2211


1:68433703_A_G
2212-2248
2249-2275
2276-2308


rs3125891_REF





1:68433703_A_G
2217-2218 2225
2251 2256 2264
2276 2280


rs3125891_SNP
2236 2243 2245
2271 2274-2275
2284-2285 2304



2248 2309-2346
2347-2382
2307-2308





2383-2424


1:68433977_G_A
2425-2470
2471-2518
2519-2568


rs11581095_REF





1:68433977_G_A
2426 2431-2432
2472 2474
2519 2521 2523


rs11581095_SNP
2435 2437-2438
2478-2479 2482
2527 2531 2534



2457 2464
2485 2504-2505
2553 2563



2569-2606
2607-2646
2647-2688


1:68434079_G_A
2689-2734
2735-2781
2782-2829


rs12030710_REF





1:68434079_G_A
2694 2696-2697
2740 2742-2743
2787 2789-2790


rs12030710_SNP
2717 2720
2764 2767
2814 2816-2817



2722-2723 2731
2769-2770
2908-2949



2830-2867
2868-2907



1:68434091_GAT_G
2689 2691 2693
2735 2737 2739
2782 2784 2786


rs1003041423_REF
2696-2700 2702
2741 2743-2747
2788 2790-2794



2708 2712 2714
2749 2755 2759
2796 2802 2806



2716 2718
2761 2763 2765
2808 2810 2812



2722-2726 2728
2769-2773 2775
2815 2817-2821



2730 2733
2777 2780
2823 2825 2828



2950-2954
2955-2958
2959-2961


1:68434556_T_C
2962-3007
3008-3055
3056-3105


rs3118419_REF





1:68434556_T_C
2962-2964
3008 3010
3056 3061 3065


rs3118419_SNP
2970-2971 2981
3016-3017 3032
3081 3084



2986 2991
3035 3038-3039
3087-3088 3101



3106-3143
3144-3183
3184-3225


1:68434669_A_AT
3226-3236




rs5774935_REF





1:68434669_A_AT
3232 3234




rs5774935_SNP





1:68434719_C_CTGTT
3237-3246
3247
3248-3251


rs150459448_REF





1:68434729_G_T
3237-3243
3247
3248-3251


rs1555845_REF
3245-3246




1:68434877_C_T

3252-3253
3254-3260


rs1555846_REF





1:68434877_C_T
3261-3270
3271-3282
3283-3298


rs1555846_SNP





1:68435174_GCTCTTGTTTC-
3299-3344
3345-3392
3393-3442


TTTTTCTGGCTT_G





rs11269074_REF





1:68435749_T_G
3443-3483
3484-3524
3525-3567


rs3790469_REF





1:68435749_T_G
3449 3459 3469
3490 3500 3505
3530 3542 3547


rs3790469_SNP
3474 3568-3605
3515 3606-3644
3558 3645-3685


1:68435988_T_C
3686-3708
3709-3731
3732-3756


rs3125894_REF





1:68435988_T_C
3686 3688 3692
3709 3711 3714
3732 3734 3739


rs3125894_SNP
3698 3757-3787
3722 3788-3808
3747 3809-3831


1:68436052_C_A
3832-3875
3876-3918
3919-3963


rs3125895_REF





1:68436052_C_A
3840 3864 3866
3884 3915 3917
3921 3928 3962


rs3125895_SNP
3871 3873-3874
4002-4041
4042-4083



3964-4001




1:68436153_G_A
4084-4097

4098-4099


rs3125896_REF





1:68436153_G_A
4085 4090




rs3125896_SNP





1:68436172_G_A
4085 4090
4132-4151
4099 4152-4175


rs3125897_REF
4100-4131




1:68436172_G_A
4176-4191
4192-4199
4200-4211


rs3125897_SNP





1:68436228_T_C
4212-4257
4258-4305
4306-4355


rs3125898_REF





1:68436228_T_C
4218-4219 4222
4264-4265 4268
4312 4314 4320


rs3125898_SNP
4227-4228 4243
4271 4274-4275
4323-4324 4330



4245 4256
4290 4293
4340 4343



4356-4393
4394-4433
4434-4475


1:68436309_T_C
4476-4521
4522-4569
4570-4619


rs17130688_REF





1:68436309_T_C
4482 4485 4490
4526 4535 4538
4574 4583 4588


rs17130688_SNP
4493 4495 4505
4541 4543 4553
4590 4592



4510 4520
4558 4568
4607-4608 4618



4620-4657
4658-4697
4698-4739


1:68436444_CTATTTATT_C
4740-4758
4759-4777
4778-4798


rs938759267_REF





1:68436444_CTATT_C
4740-4758
4759-4777
4778-4798


rs938759267_REF





1:68436702_G_A
4799-4831
4832-4866
4867-4903


rs34194247_REF





1:68436715_G_A
4799 4801
4832 4834
4867 4869-4873


rs3125900_REF
4803-4805
4836-4838
4875-4880



4807-4812 4815
4840-4845
4882-4883



4817-4818
4847-4848
4886-4887



4820-4823
4851-4852
4889-4894



4827-4828 4904
4854-4858
4898-4899 4906




4862-4863 4905



1:68436715_G_A
4805 4807-4808
4840-4841 4847
4870 4876 4882


rs3125900_SNP
4827 4907-4927
4862 4928-4952
4898 4953-4981


1:68436720_G_A
4801 4803
4832 4834 4836
4867 4869 4871


rs79716012_REF
4809-4810 4812
4842-4843 4845
4873 4877-4878



4815 4817 4820
4848 4851 4854
4880 4883 4886



4822 4828 4904
4856-4857 4863
4889 4891-4892




4905
4894 4899 4906


1:68436720_G_A
4801 4810 4815
4832 4834 4843
4867 4869 4873


rs79716012_SNP
4828 4982-4992
4848 4993-5007
4878 5008-5026


1:68437256_C_G
5027-5072
5073-5119
5120-5169


rs75711879_REF





1:68437256_C_G
5037 5040 5042
5086 5088 5095
5135 5143


rs75711879_SNP
5049 5051-5052
5097-5098
5145-5146 5151



5064-5065
5111-5112
5160-5161 5165



5170-5207
5208-5247
5248-5289


1:68438259_C_T
5290-5312
5313-5335
5336-5358


rs12145904_REF





1:68438259_C_T
5291 5296 5306
5314 5318-5319
5336 5340-5341


rs12145904_SNP
5308 5359-5377
5331 5378-5396
5343 5397-5415


1:68438583_T_C
5416-5461
5462-5509
5510-5559


rs3125902_REF





1:68438583_T_C
5416 5420 5423
5462 5466 5469
5510 5517 5525


rs3125902_SNP
5429-5431
5477 5479
5527 5532-5533



5436-5437
5483-5485
5541 5549



5560-5597
5598-5637
5638-5679


1:68438672_G_T
5680-5722
5723-5766
5767-5814


rs3118420_REF





1:68438672_G_T
5688 5693 5704
5726 5732 5737
5771 5777 5779


rs3118420_SNP
5710 5718
5748 5754
5794 5796 5802



5815-5849
5850-5882
5883-5919


1:68438830_A_G
5920-5964
5965-5999
6000-6041


rs12138573_REF





1:68438830_A_G
5926 5932 5934
5971 5977 5980
6006 6012 6016


rs12138573_SNP
5937 5939 5942
5982 5992
6018 6023



5944 6042-6079
6080-6119
6033-6034





6120-6161


1:68439675_G_C
6162-6196
6197-6231
6232-6266


rs1925955_REF





1:68439675_G_C
6174 6184-6185
6205 6210 6220
6237 6241 6246


rs1925955_SNP
6191 6267-6297
6226 6298-6328
6256 6329-6359


1:68440298_T_A
6360-6380
6381-6399
6400-6422


rs17130691_REF





1:68440298_T_A
6367 6374 6377
6386 6390 6396
6407 6411 6418


rs17130691_SNP
6380 6423-6439
6399 6440-6454
6422 6455-6473


1:68440407_G_A
6474-6499
6500-6503
6504-6514


rs3125904_REF





1:68440407_G_A
6515-6516

6507


rs3125904_SNP





1:68441530_T_C
6517-6552
6553-6583
6584-6616


rs78507000_REF





1:68441530_T_C
6519 6522 6545
6555 6577
6609 6614


rs78507000_SNP
6617-6651
6652-6686
6687-6723


1:68441814_GA_G
6724-6732




rs150774295_REF





1:68441814_GA_G
6727 6733-6740




rs150774295_SNP





1:68441844_A_C
6741-6758
6759-6775
6776-6794


rs3125905_REF





1:68441844_A_C
6741-6742 6756
6759 6773
6791 6794


rs3125905_SNP
6795-6814
6815-6831
6832-6854


1:68442800_G_A
6855-6900
6901-6948
6949-6998


rs2038900_REF





1:68442800_G_A
6859 6862 6866
6902 6906 6909
6950 6954 6957


rs2038900_SNP
6868-6869 6873
6915-6917 6921
6963 6965 6972



6876 6900
6924 7037-7075
6981 6985



6999-7036

7076-7117


1:68442818_T_C
6856 6859-6860
6901-6903
6949-6951


rs2038901_REF
6863 6866
6906-6907 6910
6954-6955 6958



6868-6869 6874
6913 6916-6917
6961 6964-6965



6877 6887
6922 6925 6935
6970 6973



7118-7153
7154-7189
6980-6981 6984





7190-7225


1:68442818_T_C
6856 6863 6874
6901 6903 6910
6949 6951 6958


rs2038901_SNP
6877 7125 7143
6925 7160 7178
6980 7211 7214



7150-7151
7186 7189
7222 7225



7226-7263
7264-7303
7304-7345


1:68443118_CA_C
7346-7391
7392-7433
7434-7482


rs147665807_REF





1:68443118_CA_C
7353 7364 7374
7408 7410 7420
7440 7442 7451


rs147665807_SNP
7377 7384 7387
7427 7431
7463 7474 7479



7389 7483-7520
7521-7560
7561-7602


1:68443430_C_T
7603-7648
7649-7696
7697-7746


rs17130694_REF





1:68443430_C_T
7604 7607 7615
7650 7653 7669
7701 7717 7720


rs17130694_SNP
7625-7626 7630
7672-7673 7677
7725 7729-7730



7636 7639
7681 7684
7733 7742



7747-7784
7785-7824
7825-7866


1:68443445_A_G
7605 7607-7610
7651 7653-7656
7697 7699


rs12408546_REF
7617 7622-7623
7659 7663 7668
7701-7704 7707



7626 7628-7631
7670 7673
7711 7716 7718



7637 7639 7642
7675-7678 7681
7721 7723-7726



7867-7896
7685 7687 7690
7730 7734 7736




7897-7926
7739 7742





7927-7956


1:68443445_A_G
7605 7623 7628
7651 7659 7670
7697 7699 7707


rs12408546_SNP
7637 7869
7685 7899 7903
7734 7933 7944



7873-7874 7884
7914 7916
7946 7949



7957-7994
7995-8034
8035-8076


1:68443820_G_A
8077-8107
8108-8130
8131-8157


rs12077372_REF





1:68443820_G_A
8079 8084 8088
8110 8114 8117
8133 8140 8144


rs12077372_SNP
8102 8158-8168
8128 8169-8177
8152 8178-8192


1:68445316_C_A
8193-8228
8229-8260
8261-8298


rs3790472_REF





1:68445316_C_A
8195 8197 8202
8229 8232 8234
8261-8262 8265


rs3790472_SNP
8204 8207 8209
8238 8243 8245
8267 8272 8279



8216 8219
8248 8251
8281 8284



8299-8314
8315-8325
8326-8341


1:68445430_A_G
8342-8387
8388-8433
8434-8483


rs3790473_REF





1:68445430_A_G
8351 8353 8358
8391 8398 8405
8435 8438 8445


rs3790473_SNP
8362 8368 8372
8415 8419 8425
8460 8463 8467



8379 8384
8429 8433
8479 8483



8484-8521
8522-8561
8562-8603


1:68446330_T_TGCTA
8604-8648
8649-8693
8694-8740


rs147893529_REF





1:68446330_T_TGCTA
8605 8612 8620
8657 8665 8680
8702 8726 8729


rs147893529_SNP
8638 8741-8771
8683 8772-8804
8737 8805-8839


1:68447072_T_C
8840-8885
8886-8933
8934-8983


rs2012235_REF





1:68447072_T_C
8843 8848 8853
8892 8895 8897
8940 8945 8952


rs2012235_SNP
8859 8862 8872
8901 8907 8910
8956 8959



8877 8879
8925 8927
8974-8975 8977



8984-9021
9022-9061
9062-9103


1:68447254_T_G
9104-9148
9149-9191
9192-9237


rs3118423_REF





1:68447254_T_G
9112 9135-9136
9180 9188
9204 9207 9234


rs3118423_SNP
9144-9145
9190-9191
9236-9237



9147-9148
9276-9315
9316-9357



9238-9275




1:68447776_T_G
9358-9386
9387-9417
9418-9450


rs2986125_REF





1:68447894_C_A
9451-9467
9468-9484
9485-9501


rs2986124_REF





1:68447894_C_A
9456 9462-9463
9478-9479
9496 9499-9501


rs2986124_SNP
9467 9502-9512
9483-9484
9524-9534




9513-9523



1:68448323_C_T
9535-9577
9578-9616
9617-9658


rs72926973_REF





1:68448323_C_T
9550 9554 9562
9589 9593 9607
9629 9633 9648


rs72926973_SNP
9575 9577
9616 9688-9710
9657 9711-9741



9659-9687




1:68448987_A_G
9742-9783
9784-9817
9818-9861


rs2277874_REF





1:68448987_A_G
9746 9750 9752
9786-9787 9790
9822-9823


rs2277874_SNP
9758-9759
9792 9797-9798
9826-9827



9768-9769 9778
9803 9806
9835-9836



9862-9899
9900-9939
9844-9845





9940-9981


1:68449261_A_G
9982-9998
9999-10010
10011-10025


rs3125906_REF





1:68449261_A_G
9983 9985 9990
9999 10001
10011-10012


rs3125906_SNP
9996 10026-
10005 10009
10016 10024



10049
10050-10064
10065-10081


1:68449382_C_A
10082-10126
10127-10168
10169-10216


rs3118426_REF





1:68449382_C_A
10103 10108
10151 10163
10187 10191


rs3118426_SNP
10111 10118
10165 10167
10196 10208



10121 10124-
10249-10275
10214-10215



10125 10217-

10276-10306



10248




1:68449796_A_T
10307-10350
10351-10392
10393-10442


rs3125907_REF





1:68449796_A_T
10308 10313
10351 10353
10393 10395


rs3125907_SNP
10316-10317
10355 10359
10397 10401



10322 10331
10362-10363
10405 10412



10333 10347
10367 10376
10421 10424



10443-10478
10479-10512
10513-10554


1:68450440_G_C
10555-10577
10578-10581
10582-10592


rs382422_REF





1:68450440_G_C
10564 10570
10580-10581
10583 10586


rs382422_SNP
10572-10573
10612-10613
10589-10590



10593-10611

10614-10620


1:68450870_T_C
10621-10661
10662-10702
10703-10745


rs3118427_REF





1:68450870_T_C
10622 10626
10663 10667-
10704-10705


rs3118427_SNP
10641-10642
10668 10682
10709 10724



10746-10783
10784-10822
10823-10863


1:68450986_G_A
10864-10909
10910-10957
10958-11007


rs3125908_REF





1:68450986_G_A
10866 10870
10911 10913
10959-10960


rs3125908_SNP
10874-10875
10917 10921-
10962 10966



10884 10898
10922 10925
10970-10971



10904 10908
10932 10946
10974 10999



11008-11045
11046-11085
11086-11127


1:68451167_G_T
11128-11156
11157-11181
11182-11208


rs12759602_REF





1:68451167_G_T
11132 11151
11169 11176
11195-11196


rs12759602_SNP
11155-11156
11180-11181
11207-11208



11209-11227
11228-11244
11245-11263


1:68451599_A_G
11264-11290
11291-11307
11308-11333


rs2477974_REF





1:68451599_A_G
11266 11272
11293 11297
11312 11314


rs2477974_SNP
11281 11288
11300 11306
11318 11322



11334-11371
11372-11402
11325 11403-





11439


1:68451602_C_A
11264-11265
11291-11292
11308-11311


rs3125909_REF
11267-11271
11294-11296
11313 11315-



11273-11282
11298-11299
11316 11318-



11284-11287
11301-11307
11321 11323-



11289-11290

11333


1:68451602_C_A
11278 11281
11302 11306
11318 11326


rs3125909_SNP
11440-11450
11451
11330 11452-





11453


1:68451671_T_C
11454-11499
11500-11547
11548-11597


rs3118428_REF





1:68451671_T_C
11454-11455
11500 11502
11548 11550


rs3118428_SNP
11460-11461
11507-11508
11556 11562



11475-11476
11523 11533
11572 11583



11486 11497
11544 11547
11594 11597



11598-11635
11636-11675
11676-11717


1:68452065_G_A


11718-11721


rs12407140_REF





1:68452441_G_A
11722-11765
11766-11798
11799-11838


rs72674322_REF





1:68452441_G_A
11735-11736
11774 11778
11809 11813


rs72674322_SNP
11741 11757
11792 11797-
11817 11823



11761 11763-
11798 11863-
11836 11838



11765 11839-
11884
11885-11910



11862




1:68452444_T_C
11722-11731
11766-11772
11799-11805


rs72674323_REF
11733-11735
11775-11777
11807-11808



11737-11746
11779-11784
11810-11812



11748-11757
11786-11792
11814-11821



11759-11763
11794-11797
11824-11831



11911-11916
11917-11921
11833-11838





11922-11926


1:68452444_T_C
11728 11735
11771 11789
11817 11828


rs72674323_SNP
11749 11753
11795 11797
11834 11837-



11761 11913-
11918-11919
11838 11923-



11914 11916
11921 11965-
11924 11926



11927-11964
12004
12005-12046


1:68452650_C_T
12047-12089
12090-12133
12134-12179


rs3125910_REF





1:68452650_C_T
12056-12057
12100 12128
12173 12175


rs3125910_SNP
12073 12085
12130 12132
12177 12179



12087 12180-
12218-12256
12257-12297



12217




1:68452696_CGT_C
12298-12323
12324-12348
12349-12374


rs1318744874_REF





1:68452696_C_CGT
12298-12323
12324-12348
12349-12374


rs1318744874_REF





1:68450655-1:68451154
10621-10634
10662-10676
10703-10718


700-1200 bps Upstream to
10637-10661
10678-10702
10720-10745


Transcriptional Start Site
10864-10909
10910-10957
10958-11007



12375-13051
13052-13753
13754-14477


1:68428322-1:68428821
1127-1128
1133-1134
1135 1137-1139


Intergenic_500 bp Downstream to
14478-14901
14902-15379
15380-15897


Exon 14





1:68437687-1:68438186
15898-16401
16402-16955
16956-17545


Intron 10_500 bp Downstream to





Exon 10





1:68431586-1:68432085
17546-18205
18206-18885
18886-19585


Intron 10_500 bp Upstream to





Exon 11





1:68431377-1:68431469
19586-19681
19682-19785
19786-19893


Intron 11





1:68431177-1:68431281
19894-19965
19966-20055
20056-20167


Intron 12





1:68430565-1:68431064
1502 1505 1510
1530 1532-1535
1544 1546


Intron 13_500 bp Downstream to
1512-1513 1516
1537-1538 1541
1549-1552


Exon 13
1518 1521 1526
1543 20822-
1554-1555



1529 20168-
21518
1558-1561



20821

21519-22246


Intron13_500 bp upstream to
22247-22790
22791-23404
23405-24080


exon14_chr





1:68429928-1:68430427





1:68448707-1:68449206
9742-9748
9784-9816
9818-9861


Intron 1_500 bp Upstream to
9750-9775
24984-25898
25899-26796


Exon 2
9778-9781





24081-24983




1:68449395-1:68449894
10307-10347
10351-10392
10393-10442


Intron 1
26797-27377
27378-28025
28026-28727


1:68448124-1:68448623
9535-9536
9578-9616
9617-9658


Intron 2_500 bp Downstream to
9538-9541
29541-30385
30386-31251


Exon 2
9543-9558





9560-9561





9563-9577





28728-29540




1:68446861-1:68447360
8840-8885
8886-8933
8934-8983


Intron 2_500 bp Upstream to
9105-9109
9150-9154
9193-9197


Exon 3
9111-9128
9156-9173
9199-9237



9130-9135
9175-9191
32583-33248



9137-9144
31917-32582




9146-9147





31252-31916




1:68446210-1:68446709
8604-8648
8649-8693
8694-8740


Intron 3_500 bp Downstream to
33249-33901
33902-34570
34571-35257


Exon 3





1:68444884-1:68445383
8193-8200
8229-8260
8261-8298


Intron 3_500 bp Upstream to
8202-8211
36000-36777
36778-37575


Exon 4
8214-8216





8218-8223 8225





8227-8228





35258-35999




1:68444673-1:68444775
37576-37649
37650-37731
37732-37829


Intron 4





1:68444031-1:68444530
37830-38491
38492-39211
39212-39971


Intron 5_500 bp Downstream to





Exon 5





1:68441001-1:68441500
39972-40665
40666-41395
41396-42151


Intron 5_500 bp Upstream to





Exon 6





1:68440353-1:68440852
42152-42581
6501-6502
6504 6506-6510


Intron 6_500 bp Downstream to

42582-43091
6512 6514


Exon 6


43092-43657


1:68439643-1:68440142
6162-6180
6197-6212
6232-6248


Intron 6_500 bp Upstream to
6182-6189
6214-6224
6250-6251


Exon 7
6191-6192 6194
6226-6227 6229
6253-6262 6264



6196 43658-
6231 44317-
6266 45014-



44316
45013
45738


1:68439324-1:68439560
45739-46010
46011-46300
46301-46608


Intron 7





1:68439082-1:68439190
46609-46634
46635-46662
46663-46692


Intron 8





1:68438317-1:68438941
5416-5461
5462-5509
5510-5559


Intron 9
5680-5722
5723-5766
5767-5814



5920-5922
5965-5988
6000-6010



5924-5930 5932
5990-5999
6012-6015



5936 5938-5950
47602-48545
6017-6029



5952 5955-5958

6031-6041



5961-5964

48546-49516



46693-47601





The indicated locations listed in column 1 of Table 1 are based on gnomAD v3 database and UCSC Genome Browser assembly ID: hg38, Sequencing/Assembly provider ID: Genome Reference Consortium Human GRCh38.p12 (GCA_000001405.27). Assembly date: December 2013 initial release; December 2017 patch release 12.


The SNP details are indicated by the listed SNP ID Nos. (“rs numbers”), which are based on the NCBI 2018 database of Single Nucleotide Polymorphisms (dbSNP)). The indicated DNA mutations are associated with Transcript Consequence NM 000329 as obtained from NCBI RefSeq genes.






Examples are provided below to facilitate a more complete understanding of the invention. The following examples illustrate the exemplary modes of making and practicing the invention. However, the scope of the invention is not limited to specific embodiments disclosed in these Examples, which are for purposes of illustration only.


EXPERIMENTAL DETAILS
Example 1: On-Target Screening Activity

In order to choose optimal RNA guides for editing strategies of an RPE65 Asp477Gly mutation causing autosomal dominant retinitis pigmentosa, three different guides targeting the mutation were screened for high on-target activity in patient derived-iPSCs that harbor the pathogenic mutation. Briefly, 2.5×105 iPSCs were mixed with pre-assembled RNPs composed of either (1) 105 pmole of SpCas9 protein and 120 pmole of 20 bp sgRNA or (2) 105 pmole of OMNI-50 protein and 120 pmole of 22 bp sgRNA. The sgRNAs target the mutated allele and are listed in Table 2. The RNP mix was combined with 100 pmole of electroporation enhancer (IDT-1075916) and electroporated using P3 Primary Cell 4D-nucleofector X Kit S (V4XP-3032, Lonza) by applying the CA-137 program. A fraction of cells were harvested 72 h post-nucleofection, genomic DNA was extracted, the region of the mutation was amplified, and the level of editing was analyzed by performing capillary electrophoreses. Edited amplicons, which contain indels, are distinguished from unedited amplicons according to their size. The graphs in FIG. 1A and FIG. 1B represent the average of % editing f STDV of two independent electroporation trials. According to capillary electrophoreses analysis, both SpCas9 (FIG. 1A) and OMNI-50 (FIG. 1B) guides displayed activity.









TABLE 2







SpCas9 and OMNI-50 sgRNA sequences targeting the


RPE65 mutation p.Asp477Gly (c.1430A > G).









Guide name
Guide sequence
PAM





sg3_spCas
CAUCUUCUUCCAAGGCACCU (SEQ ID NO: 16)
GGG





sg4_spCas
UCAUCUUCUUCCAAGGCACC (SEQ ID NO: 34)
TGG





sg7_spCas
CCCAUCUUUGUUUCUCACCC (SEQ ID NO: 23)
AGG





sg3_OMNI50
AUCAUCUUCUUCCAAGGCACCU (SEQ ID NO:
GGG



103)






sg4_OMNI50
CAUCAUCUUCUUCCAAGGCACC (SEQ ID NO:
TGG



111)






sg7_OMNI50
AACCCAUCUUUGUUUCUCACCC (SEQ ID NO:
AGG



95)









REFERENCES



  • 1. Ahmad and Allen (1992) “Antibody-mediated Specific Binging and Cytotoxicity of Lipsome-entrapped Doxorubicin to Lung Cancer Cells in Vitro”, Cancer Research 52:4817-20.

  • 2. Anderson (1992) “Human gene therapy”, Science 256:808-13.

  • 3. Basha et al. (2011) “Influence of Cationic Lipid Composition on Gene Silencing Properties of Lipid Nanoparticle Formulations of siRNA in Antigen-Presenting Cells”, Mol. Iher. 19(12):2186-200.

  • 4. Behr (1994) Gene transfer with synthetic cationic amphiphiles: Prospects for gene therapy”, Bioconjuage Chem 5:382-89.

  • 5. Blaese (1995) “Vectors in cancer therapy: how will they deliver”, Cancer Gene Ther. 2:291-97.

  • 6. Blaese et al. (1995) “T lympocyte-directed gene therapy for ADA-SCID: initial trial results after 4 years”, Science 270(5235):475-80.

  • 7. Browne et al. (2011) “A dominant mutation in RPE65 identified by whole-exome sequencing causes retinitis pigmentosa with choroidal involvement”, Eur. J. Hum. Genet. 19(10):1074-81.

  • 8. Buchschacher and Panganiban (1992) “Human immunodeficiency virus vectors for inducible expression of foreign genes”, J. Virol. 66:2731-39.

  • 9. Burstein et al. (2017) “New CRISPR-Cas systems from uncultivated microbes”, Nature 542:237-41.

  • 10. Chang and Wilson (1987) “Modification of DNA ends can decrease end-joining relative to homologous recombination in mammalian cells”, Proc. Natl. Acad. Sci. USA 84:4959-4963.

  • 11. Chung et al. (2006) “Agrobacterium is not alone: gene transfer to plants by viruses and other bacteria”, Trends Plant Sci. 11(1):1-4.

  • 12. Coelho et al. (2013) “Safety and efficacy of RNAi therapy for transthyretin amyloidosis” N. Engl. J. Med. 369, 819-829.

  • 13. Crystal (1995) “Transfer of genes to humans: early lessons and obstacles to success”, Science 270(5235):404-10.

  • 14. Dillon (1993) “Regulation gene expression in gene therapy” Trends in Biotechnology 11(5):167-173.

  • 15. Dranoff et al. (1997) “A phase I study of vaccination with autologous, irradiated melanoma cells engineered to secrete human granulocyte macrophage colony stimulating factor”, Hum. Gene Ther. 8(1):111-23.

  • 16. Dunbar et al. (1995) “Retrovirally marked CD34-enriched peripheral blood and bone marrow cells contribute to long-term engraftment after autologous transplantation”, Blood 85:3048-57.

  • 17. Ellem et al. (1997) “A case report: immune responses and clinical course of the first human use of ganulocyte/macrophage-colony-stimulating-factor-transduced autologous melanoma cells for immunotherapy”, Cancer Immunol Immunother 44:10-20.

  • 18. Gao and Huang (1995) “Cationic liposome-mediated gene transfer” Gene Ther. 2(10):710-22.

  • 19. Haddada et al. (1995) “Gene Therapy Using Adenovirus Vectors”, in: The Molecular Repertoire of Adenoviruses III: Biology and Pathogenesis, ed. Doerfler and Bohm, pp. 297-306.

  • 20. Han et al. (1995) “Ligand-directed retro-viral targeting of human breast cancer cells”, Proc Natl Acad Sci U.S.A. 92(21):9747-51.

  • 21. Inaba et al. (1992) “Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor”, J Exp Med. 176(6):1693-702.

  • 22. Jinek et al. (2012) “A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity,” Science 337(6096):816-21.

  • 23. Johan et al. (1992) “GLVR1, a receptor for gibbon ape leukemia virus, is homologous to a phosphate permease of Neurospora crassa and is expressed at high levels in the brain and thymus”, J Virol 66(3):1635-40.

  • 24. Judge et al. (2006) “Design of noninflammatory synthetic siRNA mediating potent gene silencing in vivo”, Mol Ther. 13(3):494-505.

  • 25. Kohn et al. (1995) “Engraftment of gene-modified umbilical cord blood cells in neonates with adnosine deaminase deficiency”, Nature Medicine 1:1017-23.

  • 26. Kremer and Perricaudet (1995) “Adenovirus and adeno-associated virus mediated gene transfer”, Br. Med. Bull. 51(1):31-44.

  • 27. Macdiarmid et al. (2009) “Sequential treatment of drug-resistant tumors with targeted minicells containing siRNA or a cytotoxic drug”, Nat Biotehcnol. 27(7):643-51.

  • 28. Malech et al. (1997) “Prolonged production of NADPH oxidase-corrected granulocyes after gene therapy of chronic granulomatous disease”, PNAS 94(22):12133-38.

  • 29. Miller et al. (1991) “Construction and properties of retrovirus packaging cells based on gibbon ape leukemia virus”, J Virol. 65(5):2220-24.

  • 30. Miller (1992) “Human gene therapy comes of age”, Nature 357:455-60.

  • 31. Mitani and Caskey (1993) “Delivering therapeutic genes—matching approach and application”, Trends in Biotechnology 11(5):162-66.

  • 32. Nabel and Felgner (1993) “Direct gene transfer for immunotherapy and immunization”, Trends in Biotechnology 11(5):211-15.

  • 33. Remy et al. (1994) “Gene Transfer with a Series of Lipphilic DNA-Binding Molecules”, Bioconjugate Chem. 5(6):647-54.

  • 34. Sentmanat et al. (2018) “A Survey of Validation Strategies for CRISPR-Cas9 Editing”, Scientific Reports 8:888, doi:10.1038/s41598-018-19441-8.

  • 35. Sharma et al. (2019) “Clinical-grade stem cell-derived retinal pigment epithelium patch rescues retinal degeneration in rodents and pigs.” Sci. Transl. Med. 11(475):eaat5580.

  • 36. Sommerfelt et al. (1990) “Localization of the receptor gene for type D simian retroviruses on human chromosome 19”, J. Virol. 64(12):6214-20.

  • 37. Van Brunt (1988) “Molecular framing: transgenic animals as bioactors” Biotechnology 6:1149-54.

  • 38. Vigne et al. (1995) “Third-generation adenovectors for gene therapy”, Restorative Neurology and Neuroscience 8(1,2): 35-36.

  • 39. Weissman and Kariko (2015) “mRNA:Fulfilling the promise of gene therapy”, Molecular Therapy (9):1416-7.

  • 40. Wilson et al. (1989) “Formation of infectious hybrid virion with gibbon ape leukemia virus and human T-cell leukemia virus retroviral envelope glycoproteins and the gag and pol proteins of Moloney murine leukemia virus”, J. Virol. 63:2374-78.

  • 41. Wright et al. (2013) “Complementation test of Rpe65 Knockout and tvrm148”, Invest Ophthalmol Vis Sci. 54(7):5111-22.

  • 42. Yu et al. (1994) “Progress towards gene therapy for HIV infection”, Gene Ther. 1(1):13-26.

  • 43. Zetsche et al. (2015) “Cpf1 is a single RNA-guided endonuclease of a class 2 CRIPSR-Cas system” Cell 163(3):759-71.

  • 44. Zuris et al. (2015) “Cationic lipid-mediated delivery of proteins enables efficient protein based genome editing in vitro and in vivo” Nat Biotechnol. 33(1):73-80.


Claims
  • 1. A method for modifying in a cell a mutant allele of the retinal pigment epithelium-specific 65 kDa protein (RPE65) gene having a mutation associated with a dominant RPE65 gene disorder, the method comprising introducing to the cell a composition comprising: at least one CRISPR nuclease or a sequence encoding a CRISPR nuclease; anda first RNA molecule comprising a guide sequence portion having 17-25 nucleotides or a nucleotide sequence encoding the same,wherein a complex of the CRISPR nuclease and the first RNA molecule affects a double strand break in the mutant allele of the RPE65 gene.
  • 2. The method of claim 1, wherein the first RNA molecule targets the CRISPR nuclease to the mutation associated with a dominant RPE65 gene disorder, wherein the mutation associated with a dominant RPE65 gene disorder is any one of 1:68431085T>C and 1:68446825G>A (hg38), and wherein the guide sequence portion of the first RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 that targets a mutation associated with a dominant RPE65 gene disorder.
  • 3. (canceled)
  • 4. (canceled)
  • 5. The method of claim 1, wherein the first RNA molecule targets the CRISPR nuclease to a SNP position of the mutant allele, wherein the SNP position is any one of rs60701104, rs9436400, rs868541802, rs3125890, rs75159457, rs1205919238, rs11209300, rs4264030, rs2419988, rs3118415, rs3118416, rs149739986, rs2182315, rs3118418, rs932783, rs12124063, rs77585943, rs1886906, rs3125891, rs11581095, rs12030710, rs1003041423, rs3118419, rs5774935, rs150459448, rs1555845, rs1555846, rs11269074, rs3790469, rs3125894, rs3125895, rs3125896, rs3125897, rs3125898, rs17130688, rs938759267, rs34194247, rs3125900, rs79716012, rs75711879, rs12145904, rs3125902, rs3118420, rs12138573, rs1925955, rs17130691, rs3125904, rs78507000, rs150774295, rs3125905, rs2038900, rs2038901, rs147665807, rs17130694, rs12408546, rs12077372, rs3790472, rs3790473, rs147893529, rs2012235, rs3118423, rs2986125, rs2986124, rs72926973, rs2277874, rs3125906, rs3118426, rs3125907, rs382422, rs3118427, rs3125908, rs12759602, rs2477974, rs3125909, rs3118428, rs12407140, rs72674322, rs72674323, rs3125910, and rs1318744874, and wherein the guide sequence portion of the first RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 that targets a SNP position of the mutant allele.
  • 6. (canceled)
  • 7. (canceled)
  • 8. The method of claim 5, wherein the SNP position is in an exon or intron of the RPE65 mutant allele.
  • 9. The method of claim 5, wherein the SNP position contains a heterozygous SNP.
  • 10. The method of claim 1, further comprising introducing to the cell a second RNA molecule comprising a guide sequence portion having 17-25 nucleotides or a nucleotide sequence encoding the same, wherein a complex of the second RNA molecule and a CRISPR nuclease affects a second double strand break in the RPE65 gene.
  • 11. The method of claim 10, wherein the guide sequence portion of the second RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 other than the sequence of the first RNA molecule.
  • 12. The method of claim 10, wherein the second RNA molecule comprises a non-discriminatory guide portion that targets both functional and mutated RPE65 alleles.
  • 13. The method of a claim 10, wherein the second RNA molecule comprises a non-discriminatory guide portion that targets any one a region upstream to the RPE65 transcriptional start site, an intron of RPE65, and an intergenic region downstream of RPE65.
  • 14. The method of claim 10, wherein the second RNA molecule comprises a non-discriminatory guide portion that targets a sequence that is located within a genomic range selected from any one of 1:68450655-1:68451154, 1:68428322-1:68428821, 1:68437687-1:68438186, 1:68431586-1:68432085, 1:68431377-1:68431469, 1:68431177-1:68431281, 1:68430565-1:68431064, 1:68429928-1:68430427, 1:68448707-1:68449206, 1:68449395-1:68449894, 1:68448124-1:68448623, 1:68446861-1:68447360, 1:68446210-1:68446709, 1:68444884-1:68445383, 1:68444673-1:68444775, 1:68444031-1:68444530, 1:68441001-1:68441500, 1:68440353-1:68440852, 1:68439643-1:68440142, 1:68439324-1:68439560, 1:68439082-1:68439190, and 1:68438317-1:68438941.
  • 15. The method of claim 10, wherein the second RNA molecule comprises a non-discriminatory guide portion that targets a sequence that is located up to 500 base pairs from an exon that is excised by the first and second RNA molecules.
  • 16. The method of claim 10, wherein an exon or a portion thereof is excised from the mutant allele of the RPE65 gene.
  • 17. A modified cell obtained by the method of claim 1.
  • 18. The method of claim 17, wherein the modified cell is a stem cell or a retinal pigment epithelium cell.
  • 19. A first RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516.
  • 20. A composition comprising the first RNA molecule of claim 19 and at least one CRISPR nuclease.
  • 21. The composition of claim 20, further comprising a second RNA molecule comprising a guide sequence portion having 17-25 contiguous nucleotides, wherein the second RNA molecule targets a RPE65 allele, and wherein the guide sequence portion of the second RNA molecule is a different sequence from the sequence of the guide sequence portion of the first RNA molecule.
  • 22. The composition of claim 21, wherein the guide sequence portion of the second RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-49516 other than the sequence of the first RNA molecule.
  • 23. A method for inactivating a mutant RPE65 allele in a cell, the method comprising delivering to the cell the composition of claim 20.
  • 24. A method for treating a dominant RPE65 gene disorder, the method comprising delivering to a cell of a subject having a dominant RPE65 gene disorder the composition of claim 20.
  • 25-28. (canceled)
Parent Case Info

This application claims the benefit of U.S. Provisional Application No. 62/872,514, filed Jul. 10, 2019, the contents of which are hereby incorporated by reference. Throughout this application, various publications are referenced, including referenced in parenthesis. The disclosures of all publications mentioned in this application in their entireties are hereby incorporated by reference into this application in order to provide additional description of the art to which this invention pertains and of the features in the art which can be employed with this invention.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2020/041569 7/10/2020 WO
Provisional Applications (1)
Number Date Country
62872514 Jul 2019 US