CRISPR COMPOSITIONS AND METHODS FOR PROMOTING GENE EDITING OF GATA2

Information

  • Patent Application
  • 20220307057
  • Publication Number
    20220307057
  • Date Filed
    April 14, 2020
    4 years ago
  • Date Published
    September 29, 2022
    a year ago
Abstract
RNA molecules comprising a guide sequence portion having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348 and compositions, methods, and uses thereof.
Description

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.


REFERENCE TO SEQUENCE LISTING

This application incorporates-by-reference nucleotide sequences which are present in the file named “200414_90941-A-PCT_Sequence_Listing_AWG.txt”, which is 8,382 kilobytes in size, and which was created on Apr. 14, 2020 in the IBM-PC machine format, having an operating system compatibility with MS-Windows, which is contained in the text file filed Apr. 14, 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 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., an 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. Additional examples include dominant negative mutations, which have a gene product that acts antagonistically to the wild-type allele. Other examples include haploinsufficiency, which may be caused by a loss-of-function mutation in which a reduced dosage of a normal gene product is not sufficient to produce the normal phenotype.


MonoMAC Syndrome

GATA2 is a zinc finger transcription factor essential for embryonic and definitive hematopoiesis as well as lymphatic angiogenesis. GATA2 deficiency is caused by a variety of mutations in the GATA2 gene and can have variable presentation, onset, and outcome. Patients are susceptible to mycobacterial, viral, and fungal infections, and can develop MDS, acute or chronic leukemias, lymphedema and pulmonary alveolar proteinosis. GATA2 deficiency has been referred to as MonoMAC syndrome haploinsufficiency (MonoMAC syndrome), for monocytopenia and Mycobacterium avium complex.


SUMMARY OF THE INVENTION

Disclosed herein is an approach for repairing an allele bearing a disease-associated mutation (“mutant allele”) by utilizing an RNA-guided DNA nuclease to edit, correct, or modify the nucleic acid sequence of the mutant allele such as to express a functional protein. In some embodiments, the approach is for repairing an allele bearing a dominant disease-associated mutation. The method of the enclosed invention may utilize allele-specific or non-discriminatory guide sequences to target and correct a disease-associated mutation.


According to some embodiments, the present disclosure provides a method for differentiating between the mutant allele and the wild type allele of a gene in order to edit only the mutant allele. In some embodiments, the differentiation is based on designing a guide RNA that targets a difference in sequence between the mutant allele and the functional allele. Such difference may be due to a heterozygous SNP, or a heterozygous disease-causing mutation.


According to some embodiments, the method utilizes at least one naturally occurring nucleotide difference or polymorphism (e.g., single nucleotide polymorphism (SNP)) between two alleles of a gene, to target an allele of a gene bearing a mutation causing a disease phenotype and a particular sequence in the SNP position. For example, a mutant allele in a cell can be specifically targeted based on a SNP and/or mutation sequence relative to another functional or wild-type allele in the cell which lacks the SNP and/or mutation. In some embodiments, a particular sequence in the SNP position is utilized for distinguishing or discriminating between two alleles of a gene, with one allele bearing a mutation such that it encodes a mutated protein causing a disease phenotype (“mutant allele”), and the other allele encoding for a functional, normal, or wild type protein (“functional allele”), such as to target the mutant allele bearing both a particular sequence in the SNP position and a disease associated mutation. In some embodiments, the disease-associated mutation is targeted. In some embodiments, the method further comprises the step of allele cleavage by a CRISPR nuclease. The allele cleavage may be in the form of a double-strand break (DSB) or a single-strand break. In some embodiments, the method further comprises the step of correcting the allele such that the corrected allele results in an expression of a functional GATA2 protein. In some embodiments, the correction is performed by homology directed repair (HDR). In some embodiments, the allele is altered such that it no longer possesses dominant negative properties.


According to embodiments of the present invention, there is provided 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: 1-44348.


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: 1-44348 and a CRISPR nuclease. In some embodiments, the composition further comprises a nucleic acid template for homology-directed repair, alteration, or replacement of a target sequence of an allele comprising the disease-associated mutation.


According to some embodiments of the present invention, there is provided a method for repairing or correcting a mutant GATA2 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: 1-44348 and a CRISPR nuclease. In some embodiments, a nucleic acid template is further provided to the cell for homology-directed repair, alteration, or replacement of a target sequence of the mutant GATA2 allele. In some embodiments, an endogenous template is utilized to repair, alter, or replace the target sequence of the mutant GATA2 allele. In some embodiments, non-templated repair alters the GATA2 allele.


According to some embodiments of the present invention, there is provided a method for treating, preventing, or ameliorating a condition in a subject having MonoMAC syndrome, the method comprising delivering to the subject 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: 1-44348 and a CRISPR nuclease. In some embodiments, a nucleic acid template is further provided to the cell for homology-directed repair, alteration, or replacement of a target DNA sequence comprising the pathogenic mutation. In some embodiments, an endogenous template is utilized to repair, alter, or replace the target sequence of the mutant GATA2 allele. In some embodiments, a non-templated repair process alters the GATA2 allele.


In some embodiments, the method is performed ex-vivo and the cell is provided or explanted from an individual subject. In some embodiments, the method further comprises the step of introducing the resulting cell with the corrected, repaired, or modified mutant GATA2 allele, into the individual subject (e.g. autologous transplantation).


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: 1-44348 and a CRISPR nuclease for repairing, correcting, or editing a mutant GATA2 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: 1-44348 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: 1-44348 and a CRISPR nuclease for use in inactivating repairing/correcting/editing a mutant GATA2 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: 1-44348 and a CRISPR nuclease. In some embodiments, the medicament further comprises a nucleic acid template.


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: 1-44348 and a CRISPR nuclease for treating ameliorating or preventing MonoMAC syndrome, comprising delivering to cells of a subject having or at risk of having MonoMAC syndrome, 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: 1-44348 and a CRISPR nuclease.


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: 1-44348 and a CRISPR nuclease for use in treating ameliorating or preventing MonoMAC syndrome, wherein the medicament is administered by delivering to a subject having or at risk of having MonoMAC syndrome 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: 1-44348 and a CRISPR nuclease.


According to some embodiments of the present invention, there is provided a kit for correcting/repairing a mutant GATA2 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: 1-44348, a CRISPR nuclease, and optionally a tracrRNA molecule; and instructions for delivering the RNA molecule; CRISPR nuclease, and/or the tracrRNA to the cell. In some embodiment, the delivery is performed ex-vivo. In some embodiments, the delivery is performed within a subject's body. In some embodiments, the cells are HSC cell originated from the subject.


According to some embodiments of the present invention, there is provided a kit for treating MonoMAC syndrome 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: 1-44348, a CRISPR nuclease, and optionally a tracrRNA molecule; and instructions for delivering the RNA molecule; CRISPR nuclease, and optionally the tracrRNA to a subject having or at risk of having MonoMAC syndrome. According to some embodiments of the present invention, there is provided a kit for treating MonoMAC syndrome 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: 1-44348, a CRISPR nuclease, and optionally a tracrRNA molecule; and instructions for delivering the RNA molecule; CRISPR nuclease, and optionally the tracrRNA to cells of the subject having or at risk of having MonoMAC syndrome. In some embodiments, the cells are HSC cells obtained from the subject and the delivery is ex-vivo.


The present invention also provides a method for repairing, correcting, or modifying a gene bearing a dominant disease-associated mutation. The method utilizes at least one guide sequence for targeting a CRISPR nuclease to a target sequence in a non-coding region of a gene bearing a mutation causing a disease phenotype. In some embodiments, the targeting is to a non-coding region of each of two alleles of the gene, one allele bearing a mutation such that it encodes a mutated protein causing a disease phenotype (“mutant allele”), and the other allele encoding for a functional/normal/wild type protein (“functional allele”). In some embodiments, the target sequence is in a distance of up to 1000, 900, 800, 700, 600, 500, 400, 300, 200 base pairs from the edges of an exon bearing the mutation causing the disease phenotype. Each possibility represents a separate embodiment. In some embodiments, the method further comprises the step of allele cleavage by a CRISPR nuclease. The allele cleavage is selected from the group consisting of a double strand break (DSB) and a single strand break. In some embodiments, the method further comprises the step of correction of the allele such that the corrected allele results in an expression of a functional GATA2 protein. In some embodiments, the correction is performed by homology directed repair (HDR).


The present invention provides for a method for modifying in a cell a mutant allele of the GATA2 gene having a mutation associated with MonoMAC syndrome, the method comprising

    • introducing to the cell a composition comprising:
      • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
      • an 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 RNA molecule affects a double strand break in a non-coding region of the GATA2 gene at a location that is 250 or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome.


The present invention also provides for a modified cell obtained by any of the methods of the present invention.


The present invention provides for a composition comprising the modified cells of the present invention and a pharmaceutically acceptable carrier.


The present invention also provides for an in vitro or ex vivo method of preparing the compositions of the present invention, comprising mixing the cells with the pharmaceutically acceptable carrier.


The present invention provides for a method of preparing in vitro or ex vivo a composition comprising modified cells, the method comprising:

    • a) isolating HSPCs from cells obtained from a subject with a GATA2 gene mutation related to MonoMAC syndrome and/or suffering from MonoMAC syndrome, and obtaining the cell from the subject;
    • b) introducing to the cells of step (a) a composition comprising:
      • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
      • an RNA molecule comprising a guide sequence portion having 17-25 nucleotides,
    •  wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in a non-coding region of the GATA2 gene at a location that is 250 or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome,
      • optionally, introducing to the cells a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele
    •  so as to modify the mutant allele of the GATA2 gene in one or more cells thereby obtaining modified cells; optionally further comprising the step of
    • c) culture expanding the modified cells of step (b),
    • wherein the modified cells are capable of engraftment and giving rise to progeny cells after engraftment.


The present invention provides for use of a composition prepared in vitro by a method comprising:

    • a) isolating HSPCs from cells obtained from a subject with a GATA2 gene mutation related to MonoMAC syndrome and/or suffering from MonoMAC syndrome, and obtaining the cell from the subject;
    • b) introducing to the cells of step (a) a composition comprising:
      • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
      • an RNA molecule comprising a guide sequence portion having 17-25 nucleotides,
    •  wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in a non-coding region of the GATA2 gene at a location that is 250 or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome,
      • optionally, introducing to the cells a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele
    •  so as to modify the mutant allele of the GATA2 gene in one or more cells thereby obtaining modified cells; optionally further comprising an additional step of
    • c) culture expanding the cells of step (b) wherein the modified cells are capable of engraftment and giving rise to progeny cells after engraftment; and
    • d) administering to the subject the cells of step (b) or step (c)
    • for treating the MonoMAC syndrome in the subject.


The present invention provides for a method of treating a subject afflicted with MonoMAC syndrome, comprising administration of a therapeutically effective amount of the modified cells, the compositions, or the compositions prepared by the methods of the present invention.


The present invention provides for a method for treating MonoMAC syndrome in a subject with a GATA2 gene mutation relating to MonoMAC syndrome in need thereof, the method comprising:

    • a) isolating HSPCs from cells obtained from the subject;
    • b) introducing to the cells of step (a) a composition comprising:
      • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
      • an RNA molecule comprising a guide sequence portion having 17-25 nucleotides,
    •  wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in a non-coding region of the GATA2 gene at a location that is 250 or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome,
      • optionally, introducing to the cells a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele
    •  so as to modify the mutant allele of the GATA2 gene in one or more cells thereby obtaining modified cells; optionally further comprising an additional step of
    • c) culture expanding the cells of step (b) wherein the modified cells are capable of engraftment and giving rise to progeny cells after engraftment; and
    • d) administering to the subject the cells of step (b) or step (c)
    • thereby treating the MonoMAC syndrome in the subject.


The present invention provides for a method for treating MonoMAC syndrome in a subject with a GATA2 gene mutation relating to MonoMAC syndrome in need thereof, the method comprising

    • administering to the subject autologous modified cells or progeny of autologous modified cells, wherein the autologous modified cells are modified so as to have a correction of the mutant allele of the GATA2 gene from the mutant phenotype to a non-mutant phenotype,
      • wherein said double strand break results from introduction to the cells of a composition comprising a CRISPR nuclease or sequence encoding the CRISPR nuclease and an RNA molecule wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in a non-coding region of the GATA2 gene at a location that is 250 or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome,
    • thereby treating the MonoMAC syndrome in the subject.


The present invention provides for 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: 1-44348.


The present invention provides for a composition comprising the RNA molecules of the present invention and a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele.


The present invention provides for a method for modifying in a cell a mutant allele of the GATA2 gene, the method comprising delivering to the cell the RNA molecules of the present invention or the compositions of the present invention.


The present invention provides for a method for treating MonoMAC syndrome, the method comprising delivering to a subject having MonoMAC syndrome the RNA molecules of the present invention or the compositions of present invention, or cells modified by the RNA molecules of the present invention or the compositions of the present invention.


The present invention provides for use of the RNA molecules of the present invention, the compositions of the present invention, or the composition prepared by the methods of the present invention, for modifying in a cell a mutant GATA2 allele.


The present invention provides for a medicament comprising the RNA molecules of the present invention, the compositions of the present invention, or the composition prepared by the methods of the present invention for use in inactivating in a cell a mutant GATA2 allele, wherein the medicament is administered by delivering to the cell the RNA molecules of the present invention, the compositions of the present invention, or the composition prepared by the methods of the present invention.


The present invention provides for use of the methods of the present invention, the modified cells of the present invention, the compositions of the present invention, or the compositions prepared by the methods of the present invention, or the RNA molecules of the present invention for treating ameliorating or preventing MonoMAC syndrome in to a subject having or at risk of having MonoMAC syndrome.


The present invention provides for a medicament comprising the RNA molecules of the present invention, the compositions of the present invention, the compositions prepared by the methods of the present invention, or the modified cells of the present invention, for use in treating ameliorating or preventing MonoMAC syndrome, wherein the medicament is administered by delivering to a subject having or at risk of having MonoMAC syndrome the RNA molecules of the present invention, the compositions of the present invention, the compositions prepared by the methods of the present invention, or the modified cells of the present invention.


The present invention provides for a kit for inactivating a mutant GATA2 allele in a cell, comprising an RNA molecule of the present invention, a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease, and/or a tracrRNA molecule or a sequence encoding the tracrRNA; and instructions for delivering the RNA molecule; CRISPR nuclease or a sequence encoding the CRISPR nuclease, and/or the tracrRNA molecule or a sequence encoding the tracrRNA to the cell to modify the mutant GATA2 allele in the cell.


The present invention provides for a kit for treating MonoMAC syndrome in a subject, comprising an RNA molecule of the present invention, a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease, and/or a tracrRNA molecule or a sequence encoding the tracrRNA; and instructions for delivering the RNA molecule; CRISPR nuclease or sequence encoding the CRISPR nuclease, and/or tracrRNA molecule or sequence encoding the tracrRNA to a subject having or at risk of having MonoMAC syndrome so as to treat the MonoMAC syndrome.


The present invention provides for a kit for inactivating a mutant GATA2 allele in a cell, comprising the compositions of the present invention, the compositions prepared by the methods of the present invention, or the modified cells of the present invention, and instructions for delivering the composition to the cell so as to modify the GATA2 gene in the cell.


The present invention provides for a kit for treating MonoMAC syndrome in a subject, comprising the compositions of the present invention, the compositions prepared by the methods of the present invention, or the modified cells of the present invention, and instructions for delivering the compositions of the present inventions, the compositions prepared by the methods of the present invention, or the modified cells of the present invention, to a subject having or at risk of having MonoMAC syndrome so as to treat MonoMAC syndrome.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A-FIG. 1C: Schematic representations of GATA2 editing strategies. FIG. 1A represents an allele specific editing approach based on targeting a nuclease (scissors accompanied by an arrow indicating the target site) to a pathogenic mutation (white star) in an exon (e.g. Exon 3) of a mutant GATA2 allele. FIG. 1B represents an allele specific editing approach based on targeting a nuclease (scissors accompanied by an arrow indicating the target site) to a heterozygous SNP (black star) of a mutant GATA2 allele. FIG. 1C represents a bi-allelic editing approach based on utilizing non-discriminatory guides to target a nuclease (scissors accompanied by an arrow indicating the target site) to both mutant and functional GATA2 alleles at non-coding regions within 250 basepairs of an edge of an exon bearing a pathogenic mutation position.



FIG. 2: Activity screen of guides targeting GATA2 in HeLa cells. An OMNI-50 coding plasmid was co-transfected with each of the guide DNA plasmids. Cells were harvested 72 h post DNA transfection. Genomic DNA was extracted, relevant regions were amplified, and editing level was measured by capillary electrophoresis. The graph represents the average of % editing±STDV of three independent experiments. The guides used in the activity screen are listed in Table 1.













TABLE 1







Guides used in the activity screen of FIG. 2.












Guide
Guide






Short
Long






Name
Name
Region
SNP/mutation
Guide Sequence
PAM





 g1
sgEnhancer 1
Intron 4 of 5
c.1017 + 572_
GGAAACUUCGUGUAUCUGUUUC (SEQ ID NO: 11385)
CGG


 g2
sgEnhancer 2

C > T
GACUCUAAAAACUCGCAGAGUC (SEQ ID NO: 11382)
CGG





 g3
sg152_1
Intron 4 of 5
rs11717152_
GAAGCCUGAUGUUAGAAUCAAC (SEQ ID NO: 29952)
GGG


 g4
sg152_2

T > G
AGAAGCCUGAUGUUAGAAUCAA (SEQ ID NO: 29932)
CGG


 g5
sg152_3


CAAUCCCGUUGAUUCUAACAUC (SEQ ID NO: 29940)
AGG





 g6
sg606_1
Intron 4 of 5
rs1170860_
GGGAGGGCAAGGCAGCGUCAGC (SEQ ID NO: 29699)
AGG


 g7
sg606_2

C > T
GGGCAAGGCAGCGUCAGCAGGC (SEQ ID NO: 29701)
TGG


 g8
sg606_3


GACGCUGCCUUGCCCUCCCAGU (SEQ ID NO: 10598)
CGG





 g9
sg603_1
Intron 5 of 5
rs2713603_
AAGCCCUUCUGGCACUCACUCA (SEQ ID NO: 29547)
GGG


g10
sg603_2

C > T Alt
GAAGCCCUUCUGGCACUCACUC (SEQ ID NO: 29576)
AGG


g11
sg603_3


CUGCCCUGAGUGAGUGCCAGAA (SEQ ID NO: 29562)
GGG


g12
sg603_4


GCUGCCCUGAGUGAGUGCCAGA (SEQ ID NO: 29574)
AUG





g32
sg604_1
Intron 5 of 5
rs2713604_
GGCUAGGACAGGGUCUCGGGCU (SEQ ID NO: 29310)
AGG


g33
sg604_2

T > C (A > G) Alt
GCUAGGACAGGGUCUCGGGCUA (SEQ ID NO: 29308)
GGG


g34
sg604_3


ACAGGGUCUCGGGCUAGGGAAG (SEQ ID NO: 29282)
TGG





g21
sgR396Q_2
Exon 6 of 6
p.R396Q, c.1187G > A
GAAGAAGGAAGGGAUCCAGACU (SEQ ID NO: 6562)
CGG


g24
sgR396Q_3/

p.R398W_c.1192_C > T
GGAAGGGAUCCAGACUCGGAAC (SEQ ID NO: 6427)
CGG



sgR398W_2






g25
sgR398W 3


CUUGGACUUGUUGGACAUCUUC (SEQ ID NO: 6417)
CGG









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.


It is understood that where a numerical range is recited herein, the present invention contemplates each integer between, and including, the upper and lower limits, unless otherwise stated.


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.


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: 1-44348. In some embodiments, the guide sequence portion has 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348. In some embodiments, the aforementioned guides are allele-specific. In some embodiments, the aforementioned guides target a SNP position. In some embodiments, the aforementioned guides target a GATA2 pathogenic mutation. In some embodiments, the aforementioned guides target a non-coding region of both mutant and functional GATA2 alleles.


The term “targets” as used herein, refers to the preferential hybridization of a guide sequence portion of an RNA molecule to a nucleic acid which has 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.


In the context of targeting a DNA sequence that is present in a plurality of cells, it is understood that the targeting encompasses hybridization of the guide sequence portion of the RNA molecule with the sequence in one or more of the cells, and also encompasses hybridization of the RNA molecule with the target sequence in fewer than all of the cells in the plurality of cells. Accordingly, it is understood that where an RNA molecule targets a sequence in a plurality of cells, a complex of the RNA molecule and a CRISPR nuclease is understood to hybridize with the target sequence in one or more of the cells, and also may hybridize with the target sequence in fewer than all of the cells. Accordingly, it is understood that the complex of the RNA molecule and the CRISPR nuclease introduces a double strand break in relation to hybridization with the target sequence in one or more cells and may also introduce a double strand break in relation to hybridization with the target sequence in fewer than all of the cells. 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.


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.


The RNA molecule and or the guide sequence portion of the RNA molecule may contain modified nucleotides. Exemplary modifications to nucleotides/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, (9):1416-7, hereby incorporated by reference.


In embodiments of the present invention, the guide sequence portion may be 25 nucleotides in length and contain 20-22 contiguous nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348. 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: 1-44348. For example, a guide sequence portion having 17 nucleotides in the sequence of 17 contiguous nucleotides set forth in SEQ ID NO: 44349 may consist of any one of the following nucleotide sequences (nucleotides excluded from the contiguous sequence are marked in strike-through):











(SEQ ID NO: 44349)



AAACUGACACCUCAGGGACA







17 nucleotide guide sequence 1:



(SEQ ID NO: 44350)




custom-character CUGACACCUCAGGGACA








17 nucleotide guide sequence 2:



(SEQ ID NO: 44351)




custom-character ACUGACACCUCAGGGACcustom-character








17 nucleotide guide sequence 3:



(SEQ ID NO: 44352)




custom-character AACUGACACCUCAGGGAcustom-character








17 nucleotide guide sequence 4:



(SEQ ID NO: 44353)



AAACUGACACCUCAGGGcustom-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: 1-44348 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 affect 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 comprising a guide sequence portion of the present invention, 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., Science (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 base-pairing 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.


According to embodiments of the present invention, an RNA molecule may be up to 500, 400, 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.


A skilled artisan will appreciate that in each of the embodiments of the present invention, individually, each of the RNA molecules of the present invention are capable of complexing with a nuclease, e.g. a CRISPR nuclease, such as to associate with a target genomic DNA sequence of interest next to a protospacer adjacent motif (PAM). The nuclease then induces cleavage of target DNA to create a double-stranded break within the protospacer. Accordingly, in embodiments of the present invention, the guide sequences and RNA molecules of the present invention may target a location 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or 28 nucleotides upstream or downstream from a PAM site.


Therefore, in embodiments of the present invention, the RNA molecules of the present invention in complex with a nuclease, e.g., a CRISPR nuclease, may affect a double strand break in alleles of a gene 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 upstream or downstream from a target site.


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.


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.


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


As used herein, the term “HSPC” refers to both hematopoietic stem cells and hematopoietic stem progenitor cells. Non-limiting examples of stem cells include a bone marrow cell, a myeloid progenitor cell, a multipotent progenitor cell, a lineage restricted progenitor cell.


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). As used herein “hematopoiesis” or “hemopoiesis” refers to the formation and development of various types of blood cells (e.g., red blood cells, megakaryocytes, myeloid cells (e.g., monocytes, macrophages and neutrophil), and lymphocytes) and other formed elements in the body (e.g., in the bone marrow).


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.


The term “homology-directed repair” or “HDR” refers to a mechanism for repairing DNA damage in cells, for example, during repair of double-stranded and single-stranded breaks in DNA. HDR requires nucleotide sequence homology and uses a “nucleic acid template” (nucleic acid template or donor template used interchangeably herein) to repair the sequence where the double-strand or single strand break occurred (e.g., DNA target sequence). This results in the transfer of genetic information from, for example, the nucleic acid template to the DNA target sequence. HDR may result in alteration of the DNA target sequence (e.g., insertion, deletion, mutation) if the nucleic acid template sequence differs from the DNA target sequence and part or all of the nucleic acid template polynucleotide or oligonucleotide is incorporated into the DNA target sequence. In some embodiments, an entire nucleic acid template polynucleotide, a portion of the nucleic acid template polynucleotide, or a copy of the nucleic acid template is integrated at the site of the DNA target sequence. DNA repair pathways, including but not limited to HDR, play a role in targeted genome modification, which is a powerful tool that can be used to reverse the effect of pathogenic genetic variations and therefore has the potential to provide new therapies for human genetic diseases. Current genome engineering tools, including engineered zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and most recently, RNA-guided DNA endonucleases such as CRISPR/Cas, produce sequence-specific DNA breaks in a genome.


The modification of the genomic sequence occurs at the next step and is the product of the activity cellular DNA repair mechanisms triggered in response to the newly formed DNA break. These mechanisms include, for example: (1) classical non-homologous end-joining (NHEJ) in which the two ends of the break are ligated together in a fast but also inaccurate manner (i.e. frequently resulting in mutation of the DNA at the cleavage site in the form of small insertion or deletions) and (2) homology-directed repair (HDR) in which an intact homologous DNA donor is used to replace the DNA surrounding the cleavage site in an accurate manner. In addition and as discussed above, HDR can also mediate the precise insertion of external or endogenous DNA at the break site.


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 (or any integer value there between or there above), preferably between about 100 and 1,000 nucleotides in length (or any integer there between), 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 embodiment, 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 ribonucleotide 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 embodiment, the nucleic acid template comprises modified ribonucleotides.


Insertion of an exogenous sequence (also called a “donor sequence,” donor template” or “donor”), for example, for correction of a mutant gene or for increased expression of a wild-type gene can also be carried out. It will be readily apparent that the donor sequence is typically not identical to the genomic sequence where it is placed. 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.


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 2019/0330620A1. The donor sequence may be encoded on the same molecule as a guide RNA sequence. See, for example, Anzalone et al. (2019) Nature 576:149-157. 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) Proc. Natl. Acad. Sci. USA 84:4959-4963; Nehls et al. (1996) Science 272:886-889. 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 also be an oligonucleotide and be used for gene correction or 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. The oligonucleotide can be used to ‘correct’ a mutated sequence in an endogenous gene (e.g., the sickle mutation in beta globin), or may be used to insert sequences with a desired purpose into an endogenous locus.


A polynucleotide can be introduced into a cell as part of a vector molecule having additional sequences such as, for example, replication origins, promoters and genes encoding antibiotic resistance. Moreover, 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)).


The donor may be inserted so that its expression is driven by the endogenous promoter at the integration site, namely the promoter that drives expression of the endogenous gene into which the donor is inserted. However, it will be apparent that the donor may comprise a promoter and/or enhancer, for example a constitutive promoter or an inducible or tissue specific promoter.


The donor molecule may be inserted into an endogenous gene such that all, some or none of the endogenous gene is expressed. For example, a transgene as described herein may be inserted into an endogenous locus such that some (N-terminal and/or C-terminal to the transgene) or none of the endogenous sequences are expressed, for example as a fusion with the transgene. In other embodiments, the transgene (e.g., with or without additional coding sequences such as for the endogenous gene) is integrated into any endogenous locus, for example a safe-harbor locus, for example a CCR5 gene, a CXCR4 gene, a PPP1R12c (also known as AAVS1) gene, an albumin gene or a Rosa gene. See, e.g., U.S. Pat. Nos. 7,951,925 and 8,110,379; U.S. Publication Nos. 20080159996; 201000218264; 20100291048; 20120017290; 20110265198; 20130137104; 20130122591; 20130177983 and 20130177960 and U.S. Provisional Application No. 61/823,689).


When endogenous sequences (endogenous or part of the transgene) are expressed with the transgene, the endogenous sequences may be full-length sequences (wild-type or mutant) or partial sequences. Preferably the endogenous sequences are functional. Non-limiting examples of the function of these full length or partial sequences include increasing the serum half-life of the polypeptide expressed by the transgene (e.g., therapeutic gene) and/or acting as a carrier.


Furthermore, although not required for expression, exogenous sequences may also include transcriptional or translational regulatory sequences, for example, promoters, enhancers, insulators, internal ribosome entry sites, sequences encoding 2A peptides and/or polyadenylation signals.


In certain embodiments, the donor molecule comprises a sequence selected from the group consisting of a gene encoding a protein (e.g., a coding sequence encoding a protein that is lacking in the cell or in the individual, or a portion thereof; or an alternate version of a gene encoding a protein, or a portion thereof), a regulatory sequence and/or a sequence that encodes a structural nucleic acid such as a microRNA or siRNA.


One aspect of the present disclosure provides a method for utilizing a guide sequence to target a CRISPR nuclease to affect a DSB to modify an allele of a gene bearing a disease associated mutation (‘mutant allele’). In some embodiments, at least one naturally occurring nucleotide difference or polymorphism (e.g., single nucleotide polymorphism (SNP)) and/or a disease associated mutation is utilized for targeting one of two alleles of a gene which is an allele bearing a mutation causing a disease phenotype (‘mutant allele’) rather than a functional/wildtype allele. In some embodiments, a SNP position is utilized for distinguishing/discriminating between two alleles of a gene, an allele bearing a disease associated mutation and a particular sequence (SNP/REF) in the SNP position, and a functional/wild type allele bearing a different sequence in the SNP position. In some embodiments, a disease associated mutation is utilized for distinguishing/discriminating between two alleles of a gene, an allele bearing the disease associated mutation, and a wildtype mutant allele not bearing the same disease associated mutation or bearing a different disease associated mutation. In other embodiments, a non-coding region of the gene common to both the mutant allele and the functional allele is targeted. In some embodiments, the method is for treating, ameliorating, or preventing a dominant haploinsufficiency genetic disorder. In some embodiments, the method further comprises the step of allele cleavage by a CRISPR nuclease. The allele cleavage is selected from the group consisting of: a double strand break (DSB) and a single strand break. In some embodiments, the allele cleavage is a DSB. In some embodiments, the method further comprises the step of correction of the allele such that the corrected allele results in expression of a functional protein. In some embodiments, the correction is performed by homology directed repair (HDR). In some embodiments, the method further comprises the step of editing/correcting/modifying a sequence of the mutant allele such as to allow expression of a functional protein. In some embodiments, the method further comprises the step of editing/correcting/modifying sequences of the two alleles such as to allow expression of a functional protein.


According to embodiments of the present invention, there is provided a method for modifying in a cell a mutant allele of the GATA2 gene having a mutation associated with MonoMAC syndrome, the method comprising


introducing to the cell a composition comprising:

    • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
    • an 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 RNA molecule affects a double strand break in the mutant allele of the GATA2 gene.


In some embodiments, the RNA molecule targets the CRISPR nuclease to the mutation associated with MonoMAC syndrome.


In some embodiments, the mutation associated with MonoMAC syndrome is any one of 3:128479475_T_C; 3:128479476_A_G; 3:128479846_C_A; 3:128479875_C_T; 3:128480135_T_A; 3:128480322_G_A; 3:128480449_G_A; 3:128480599_C_T; 3:128480829_G_A; 3:128480845_C_T; 3:128480918_C_T; 3:128480935_T_C; 3:128480946_G_A; 3:128481039_T_A; 3:128481047_G_A; 3:128481055_G_T; 3:128481060_C_T; 3:128481071_C_A; 3:128481077_G_T; 3:128481090_G_A; 3:128481092 G_A; 3:128481095_G_A; 3:128481102_G_A; 3:128481115_G_T; 3:128481131_G_A; 3:128481136_G_A; 3:128481142_C_T; 3:128481147_G_A; 3:128481150_C_A; 3:128481162_C_G; 3:128481176_C_G; 3:128481180_A_T; 3:128481188_G_A; 3:128481189_A_G; 3:128481199_C_T; 3:128481214_C_G; 3:128481219_C_T; 3:128481220_G_C; 3:128481224_C_T; 3:128481226_C_A; 3:128481230_G_A; 3:128481230_G_T; 3:128481232_C_A; 3:128481245_T_A; 3:128481262_C_T; 3:128481270_G_A; 3:128481275_C_T; 3:128481276_G_A; 3:128481283_G_C; 3:128481299_A_G; 3:128481302_G_T; 3:128481315_TAACC_T; 3:128481318_C_A; 3:128481819_A_G; 3:128481836_A_AGAGG; 3:128481841_C_T; 3:128481845_A_G; 3:128481849_G_T; 3:128481866_CGTTGGCGTTTCG_C; 3:128481872_C_T; 3:128481877_C_T; 3:128481878_G_A; 3:128481880_C_A; 3:128481881_G_A; 3:128481881_G_C; 3:128481887_A_C; 3:128481897_TGTCG_AAGGC; 3:128481898_G_T; 3:128481901_G_A; 3:128481908_A_G; 3:128481923_T_TGCCGGCTCTTCTGGCG; 3:128481924_G_A; 3:128481926_C_T; 3:128481927_G_A; 3:128481937_G_A; 3:128481937_G_GCGGC; 3:128481938_C_A; 3:128481938_C_T; 3:128481939_GGCCG_AAGGC; 3:128481942_C_T; 3:128481945_C_A; 3:128483288_G_A; 3:128483333_C_T; 3:128483347_C_T; 3:128483375_T_A; 3:128483868_G_A; 3:128483889_G_A; 3:128483905_CT_C; 3:128483906_T_C; 3:128483925_C_CATTGCACAGGT; 3:128483957_C_T; 3:128483973_C_CTGTGGCCCCACAGT; 3:128484010_A_C; 3:128485738_CT; 3:128485741_G_A; 3:128485746_G_A; 3:128485750_C_T; 3:128485758_AG_A; 3:128485766_A_T; 3:128485769_T_C; 3:128485779_T_TC; 3:128485780_C_A; 3:128485783_C_T; 3:128485789_A_C; 3:128485795_C_A; 3:128485798_G_A; 3:128485811_C_T; 3:128485819_T_TAGTCGTGGGC; 3:128485837 G_A; 3:128485837_G_C; 3:128485843_TC; 3:128485850_G_A; 3:128485855 G_A; 3:128485869_G_C; 3:128485871_G_A; 3:128485874_T_C; 3:128485891_A_G; 3:128485892_T_C; 3:128485892_T_G; 3:128485909_C_T; 3:128485910_G_A; 3:128485916_G_T; 3:128485924_C_T; 3:128485929_C_T; 3:128485937_T_G; 3:128485944_C_CGTCAG; 3:128485944_C_T; 3:128485962_C_A; 3:128485966_A_ACGC; 3:128485967_C_G; 3:128485970_CT; 3:128485973_C_T; 3:128485982_C_G; 3:128485998_A_AC; 3:128485999_C_T; 3:128486002_C_A; 3:128486004_C_CGCGGAAGA; 3:128486005_G_A; 3:128486006_C_T; 3:128486023_G_A; 3:128486030_C_T; 3:128486031_C_A; 3:128486038_G_C; 3:128486057_C_T; 3:128486059_T_G; 3:128486066_G_A; 3:128486072_T_G; 3:128486075_G_A; 3:128486103_G_T; 3:128486104_T_A; 3:128486117_G_A; 3:128486129_C_A; 3:128486141_C_T; 3:128486142_G_T; 3:128486143_C_T; 3:128486153_C_T; 3:128486173_G_A; 3:128486179_A_G; 3:128486185_A_G; 3:128486189 G_A; 3:128486201_C_T; 3:128486218_T_C; 3:128486227_G_T; 3:128486240_G_A; 3:128486252_A_AG; 3:128486260_T_C; 3:128486271_C_T; 3:128486284_A_AGG; 3:128486287_G_A; 3:128486288_C_A; 3:128486295_GC_G; 3:128486296_C_T; 3:128486298_CC_AA; 3:128486320_G_T; 3:128486355_T_GC; 3:128486356_C_G; 3:128486365_C_CG; 3:128486805_TC; 3:128486808_G_C; 3:128486826_C_T; 3:128486828_C_A; 3:128486850_G_A; 3:128486856_T_C; 3:128486872_A_C; 3:128486883_1_C; 3:128486890_A_T; 3:128486893_C_T; 3:128486896_C_T; 3:128486911_G_C; 3:128486931_A_G; 3:128486948_G_A; 3:128486956_G_T; 3:128486961_G_GACA; 3:128486967_G_C; 3:128486970_T_A; 3:128486973_T_G; 3:128486982_A_T; 3:128486992_G_A; 3:128487002_C_A; 3:128487004_A_G; 3:128487009_G_T; 3:128487016_CG_GC; 3:128487016_C_T; 3:128487021_G_T; 3:128487036_G_A; 3:128487073_G_C; 3:128492886_G_A; 3:128492959_A_C; 3:128493033_G_T; 3:128493046 G_A; 3:128493068_G_A.


In some embodiments, the guide sequence portion of the RNA molecule comprises a guide sequence of 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348 which targets a mutation associated with MonoMAC syndrome.


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


In some embodiments, the SNP position is in a non-coding region of the GATA2 gene at a location that is 250 or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome.


In some embodiments, the SNP position is located at any one of rs10934857; rs2713604; rs2713603; rs11708606; rs11717152; rs2659689; rs2713602; rs2335052; rs4577488; or rs1573858.


In some embodiments, the guide sequence portion of the RNA molecule comprises a guide sequence having 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348 which targets a sequence in a SNP position of the mutant allele.


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


According to embodiments of the present invention, the RNA molecule targets a particular sequence (SNP/REF) in a SNP position or a disease-causing mutation of a mutant allele.


In some embodiments, the SNP position is in a promoter region, the start codon, an untranslated region (UTR), an intron, an exon, a downstream sequence of a mutant allele. Each possibility represents a separate embodiment. In some embodiments, the SNP position is in a distance of less than 2000 nucleotides, 15000 nucleotides, 1000 nucleotides, 750 nucleotides, 500 nucleotides, 400 nucleotides, 300 nucleotides, 250 nucleotides, 200 nucleotides, 150 nucleotides, or 50 nucleotides from the disease-associated mutation. Each possibility represents a separate embodiment.


In some embodiments, a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in a non-coding region common to both alleles of the GATA2 gene at a location that is 500, 450, 400, 350, 300, 250 or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome. Each possibility represents a separate embodiment.


In some embodiments, the coding region is any one of Exon 2, Exon 3, Exon 4, Exon 5, or Exon 6 of the GATA2 gene.


In some embodiments, the non-coding region is any one of the 3′UTR, Intron 1, Intron 2, Intron 3, Intron 4, or Intron 5 of the GATA2 gene.


In some embodiments, the non-coding region is any one of the 3:128480417-128480717; 3:128487000-128487300; 3:128486500-128486800; 3:128486375-128486675; 3:128485421-128485721; 3:128484028-128484328; 3:128483533-128483833; 3:128481949-128482249; 3:128481516-128481816; or 3:128481320-128481620.


In some embodiments, the guide sequence portion of the RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348 which targets a non-coding region of the GATA2 gene.


In some embodiments, further comprising introduction of a donor template for homology directed repair (HDR), alteration, or replacement of a desired sequence of the GATA2 allele. Accordingly, this method of modifying a GATA2 pathogenic mutation via HDR corrects the dominant haploinsufficiency disorder that manifests as MonoMAC syndrome.


In embodiments of the present invention, the target sequence is in the coding region bearing the mutation associated with MonoMAC syndrome or is the entire coding region bearing the mutation associated with MonoMAC syndrome.


In embodiments of the present invention, the modifying results in correction of the mutant allele of the GATA2 gene from the mutant phenotype to a non-mutant phenotype.


Embodiments of the present invention further comprise obtaining the cell with a GATA2 gene mutation associated with MonoMAC syndrome from a subject with a GATA2 gene mutation related to MonoMAC syndrome and/or suffering from MonoMAC syndrome.


Embodiments of the present invention further comprise first selecting a subject with a GATA2 gene mutation related to MonoMAC syndrome and/or suffering from MonoMAC syndrome.


Embodiments of the present invention further comprise obtaining the cell from the subject by mobilization and/or by apheresis.


Embodiments of the present invention further comprise obtaining the cell from the subject by bone marrow aspiration.


In embodiments of the present invention, the cell is prestimulated prior to introducing the composition to the cell.


Embodiments of the present invention further comprise culture expanding the cell to obtain cells.


In embodiments of the present invention, the cells are cultured with one or more of: stem cell factor (SCF), IL-3, and GM-CSF.


In embodiments of the present invention, the cells are cultured with at least one cytokine.


In embodiments of the present invention, the at least one cytokine is a recombinant human cytokine.


In embodiments of the present invention, the cell is among a plurality of cells, wherein the composition comprising the RNA molecule and/or donor template is introduced into at least the cell as well as other cells among the plurality of cells, and the mutant allele of the GATA2 gene is modified in at least the cell as well as in the other cells among the plurality of cells, thereby obtaining multiple modified cells.


In embodiments of the present invention, introducing the composition comprising the RNA molecule and/or introduction of the donor template comprises electroporation of the cell or cells.


The present invention provides for a modified cell obtained by the methods of the present invention.


The present invention provides for modified cells obtained from culture expanding the modified cell obtained by the methods of the present invention.


In embodiments of the present invention, the modified cell or cells are capable of engraftment.


In embodiments of the present invention, the modified cell or cells are capable of giving rise to progeny cells.


In embodiments of the present invention, the modified cell or cells are capable of giving rise to progeny cells after engraftment.


In embodiments of the present invention, the modified cell or cells are capable of giving rise to progeny cells after an autologous engraftment.


In embodiments of the present invention, the modified cell or cells are capable of giving rise to progeny cells for at least 12 months or at least 24 months after engraftment.


In embodiments of the present invention, the modified cell or cells are hematopoietic stem cells and/or progenitor cells (HSPCs).


In embodiments of the present invention, the modified cell or cells are CD34+ hematopoietic stem cells.


In embodiments of the present invention, the modified cell or cells are bone marrow cells or peripheral mononucleated cells (PMCs).


The present invention provides for a composition comprising the modified cells of the present invention and a pharmaceutically acceptable carrier.


In embodiments of the present invention, an in vitro or ex vivo method of preparing the compositions of the present invention, comprising mixing the cells with the pharmaceutically acceptable carrier.


The present invention provides for a method of preparing in vitro or ex vivo a composition comprising modified cells, the method comprising:

    • a) isolating HSPCs from cells obtained from a subject with a GATA2 gene mutation related to MonoMAC syndrome and/or suffering from MonoMAC syndrome, and obtaining the cell from the subject;
    • b) introducing to the cells of step (a) a composition comprising:
      • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
      • an RNA molecule comprising a guide sequence portion having 17-25 nucleotides,
    •  wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in the mutant allele of the GATA2 gene according to any one of the methods of the invention,
      • optionally, introducing to the cells a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele
    •  so as to modify the mutant allele of the GATA2 gene in one or more cells thereby obtaining modified cells; optionally further comprising the step of
    • c) culture expanding the modified cells of step (b),
    • wherein the modified cells are capable of engraftment and giving rise to progeny cells after engraftment.


The present invention provides for use of a composition prepared in vitro by a method comprising:

    • a) isolating HSPCs from cells obtained from a subject with a GATA2 gene mutation related to MonoMAC syndrome and/or suffering from MonoMAC syndrome, and obtaining the cell from the subject;
    • b) introducing to the cells of step (a) a composition comprising:
      • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
      • an RNA molecule comprising a guide sequence portion having 17-25 nucleotides,
    •  wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in the mutant allele of the GATA2 gene according to any one of the methods of the invention,
      • optionally, introducing to the cells a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele
    •  so as to modify the mutant allele of the GATA2 gene in one or more cells thereby obtaining modified cells; optionally further comprising an additional step of
    • c) culture expanding the cells of step (b) wherein the modified cells are capable of engraftment and giving rise to progeny cells after engraftment; and
    • d) administering to the subject the cells of step (b) or step (c)
    • for treating the MonoMAC syndrome in the subject.


The present invention provides for a method of treating a subject afflicted with MonoMAC syndrome, comprising administration of a therapeutically effective amount of the modified cells of the present invention, the compositions of the present invention, or the composition prepared by the methods of the present invention.


The present invention provides for a method for treating MonoMAC syndrome in a subject with a GATA2 gene mutation relating to MonoMAC syndrome in need thereof, the method comprising:

    • a) isolating HSPCs from cells obtained from the subject;
    • b) introducing to the cells of step (a) a composition comprising:
      • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
      • an RNA molecule comprising a guide sequence portion having 17-25 nucleotides,
    •  wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in the mutant allele of the GATA2 gene according to any one of the methods of the invention,
      • optionally, introducing to the cells a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele
    •  so as to modify the mutant allele of the GATA2 gene in one or more cells thereby obtaining modified cells; optionally further comprising an additional step of
    • c) culture expanding the cells of step (b) wherein the modified cells are capable of engraftment and giving rise to progeny cells after engraftment; and
    • d) administering to the subject the cells of step (b) or step (c)
    • thereby treating the MonoMAC syndrome in the subject.


The present invention provides for a method for treating MonoMAC syndrome in a subject with a GATA2 gene mutation relating to MonoMAC syndrome in need thereof, the method comprising

    • administering to the subject autologous modified cells or progeny of autologous modified cells, wherein the autologous modified cells are modified so as to have a correction of the mutant allele of the GATA2 gene from the mutant phenotype to a non-mutant phenotype,
      • wherein said double strand break results from introduction to the cells of a composition comprising a CRISPR nuclease or sequence encoding the CRISPR nuclease and an RNA molecule wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in the mutant allele of the GATA2 gene according to any one of the methods of the invention,
    • thereby treating the MonoMAC syndrome in the subject.


The present invention provides for 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: 1-44348.


In embodiments of the present invention, the RNA molecule further comprises a portion having a sequence which binds to a CRISPR nuclease.


In embodiments of the present invention, the sequence which binds to a CRISPR nuclease is a tracrRNA sequence.


In embodiments of the present invention, the RNA molecule further comprises one or more linker portions.


In embodiments of the present invention, the RNA molecule further comprises a portion having a tracr mate sequence.


In embodiments of the present invention, the RNA molecule is up to 300 nucleotides in length.


According to embodiments of the present invention, the 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, 30 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.


The present invention provides for a composition comprising the RNA molecules of the present invention and a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele.


In embodiments of the present invention, the composition further comprisesone or more CRISPR nucleases or sequences encoding the one or more CRISPR nucleases, and/or one or more tracrRNA molecules or sequences encoding the one or more tracrRNA molecules.


The present invention provides for a method for modifying in a cell a mutant allele of the GATA2 gene, the method comprising delivering to the cell the RNA molecules of the present invention or the compositions of the present invention.


The present invention provides for a method for treating MonoMAC syndrome, the method comprising delivering to a subject having MonoMAC syndrome the RNA molecules of the present invention or the compositions of the present invention, or cells modified by the RNA molecules of the present invention or the compositions of the present invention.


In embodiments of the present invention, the one or more CRISPR nucleases and/or the tracrRNA and the RNA molecule are delivered to the subject and/or cells substantially at the same time or at different times.


In embodiments of the present invention, the modifying or treating results in a both alleles of the GATA2 gene encoding a functional protein.


The present invention provides for use of the RNA molecules of the present invention, the compositions of the present invention, or the composition prepared by the methods of the present invention, for modifying in a cell a mutant GATA2 allele.


The present invention provides for a medicament comprising the RNA molecules of the present invention, the compositions of the present invention, or the composition prepared by the methods of the present invention for use in inactivating in a cell a mutant GATA2 allele, wherein the medicament is administered by delivering to the cell the RNA molecules of the present invention, the compositions of the present invention, or the composition prepared by the methods of the present invention.


The present invention provides for use of the methods of the present invention, the modified cells of the present invention, the compositions of the present invention, or the compositions prepared by the methods of the present invention, or the RNA molecules of the present invention for treating ameliorating or preventing MonoMAC syndrome in to a subject having or at risk of having MonoMAC syndrome.


The present invention provides for a medicament comprising the RNA molecules of the present invention, the compositions of the present invention, the compositions prepared by the methods of the present invention, or the modified cells of the present invention, for use in treating ameliorating or preventing MonoMAC syndrome, wherein the medicament is administered by delivering to a subject having or at risk of having MonoMAC syndrome the RNA molecules of the present invention, the compositions of the present invention, the compositions prepared by the methods of the present invention, or the modified cells of the present invention.


The present invention provides for a kit for inactivating a mutant GATA2 allele in a cell, comprising an RNA molecule of the present invention, a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease, and/or a tracrRNA molecule or a sequence encoding the tracrRNA; and instructions for delivering the RNA molecule; CRISPR nuclease or a sequence encoding the CRISPR nuclease, and/or the tracrRNA molecule or a sequence encoding the tracrRNA to the cell to modify the mutant GATA2 allele in the cell.


The present invention provides for a kit for treating MonoMAC syndrome in a subject, comprising an RNA molecule of the present invention, a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease, and/or a tracrRNA molecule or a sequence encoding the tracrRNA; and instructions for delivering the RNA molecule; CRISPR nuclease or sequence encoding the CRISPR nuclease, and/or tracrRNA molecule or sequence encoding the tracrRNA to a subject having or at risk of having MonoMAC syndrome so as to treat the MonoMAC syndrome.


The present invention provides for a kit for inactivating a mutant GATA2 allele in a cell, comprising the compositions of the present invention, the compositions prepared by the methods of the present invention, or the modified cells of the present invention, and instructions for delivering the composition to the cell so as to modify the GATA2 gene in the cell.


The present invention provides for a kit for treating MonoMAC syndrome in a subject, comprising the compositions of the present invention, the compositions prepared by the methods of the present invention, or the modified cells of the present invention, and instructions for delivering the compositions of the present inventions, the compositions prepared by the methods of the present invention, or the modified cells of the present invention, to a subject having or at risk of having MonoMAC syndrome so as to treat MonoMAC syndrome.


According to some embodiments of the present invention, there is provided a kit for repairing/correcting/modifying a mutant MonoMAC syndrome allele in a cell(s), 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: 1-44348, a CRISPR nuclease, and/or a tracrRNA molecule; and instructions for delivering the RNA molecule; CRISPR nuclease, and/or the tracrRNA to the cell. In some embodiments, the kit further comprises, a nucleic acid template for homology-directed repair, alteration, or replacement of at least a portion of a target gene.


According to some embodiments of the present invention, there is provided a kit for treating MonoMAC syndrome 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: 1-44348, a CRISPR nuclease, and/or a tracrRNA molecule; and instructions for delivering the RNA molecule; CRISPR nuclease, and/or the tracrRNA to a subject having or at risk of having MonoMAC syndrome. In some embodiments, the kit further comprises, a nucleic acid template for homology-directed repair, alteration, or replacement of at least a portion of a target gene.


The present invention provides for a method for selecting a subject for treatment of MonoMAC syndrome from a pool of subjects diagnosed with MonoMAC syndrome, comprising the steps of:

    • a) obtaining cells from each subject in the pool of subjects;
    • b) screening the cells of each subject for a GATA2 gene mutation related to MonoMAC syndrome and selecting only subjects with a GATA2 gene mutation related to MonoMAC syndrome;
    • c) screening by sequencing the cells of subjects selected in step (b) for heterozygosity at one or more polymorphic sites selected from the group consisting of rs10934857; rs2713604; rs2713603; rs11708606; rs11717152; rs2659689; rs2713602; rs2335052; rs4577488; or rs1573858; and
    • d) selecting for treatment only subjects with cells that are heterozygous at the one or of the more polymorphic sites.


According to some embodiments of the present invention, there is provided a method for treating MonoMAC syndrome, the method comprising delivering to a subject or cell(s) obtained from a subject having MonoMAC syndrome or at risk of MonoMAC syndrome 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: 1-44348 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: 1-44348 and a CRISPR nuclease for use in repairing/correcting/modifying a mutant GATA2 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: 1-44348 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: 1-44348 and a CRISPR nuclease for treating ameliorating or preventing MonoMAC syndrome, comprising delivering to a subject having or at risk of having MonoMAC syndrome 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: 1-44348 and a CRISPR nuclease.


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: 1-44348 and a CRISPR nuclease for use in treating ameliorating or preventing MonoMAC syndrome, wherein the medicament is administered by delivering to a subject having or at risk of having MonoMAC syndrome: 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: 1-44348 and a CRISPR nuclease. In some embodiments, the medicament further comprises, a nucleic acid template for homology-directed repair, alteration, or replacement of at least a portion of a target gene.


The compositions and methods of the present disclosure may be utilized for treating, preventing, ameliorating, or slowing progression of MonoMAC syndrome.


In some embodiments, the method of repairing/correcting a mutant 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.


In embodiments of the present invention, the guide sequence portion of the RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348.


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: 1-44348 and a CRISPR nuclease.


According to embodiments of the present invention, the composition may comprise a nucleic acid template for homology-directed repair, alteration, or replacement of a target DNA sequence comprising the pathogenic mutation (e.g., an allele bearing a disease-associated mutation, or an allele bearing a disease-associated mutation and a particular sequence SNP/REF in the SNP position).


According to some embodiments of the present invention, there is provided a method for repairing/correcting a mutant GATA2 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: 1-44348 and a CRISPR nuclease.


In a non-limiting example, an RNA molecule comprising a guide sequence is utilized to direct a CRISPR nuclease to a mutant allele and create a double-strand break (DSB) and correction/repair of the mutant allele is further performed, such as by utilizing homology directed repair (HDR), which incorporates a homologous strand as a repair template.


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


According to embodiments of the present invention, the tracrRNA is delivered to the subject and/or cells obtained from the subject 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 nucleic acid template is delivered to the subject and/or cells obtained from the subject substantially at the same time or at different times as the CRISPR nuclease and RNA molecule or RNA molecules.


In some embodiments, there is provided a method comprising removing an exon containing a disease-causing mutation from a mutant allele, wherein the RNA molecule or a first and a second RNA molecules target regions flanking an entire exon or a portion of the exon or multiple exons or the entire open reading frame of a gene, or the entire gene. In some embodiments, the method includes exon skipping. In some embodiments, the method further comprises, utilizing a nucleic acid template for homology-directed repair, alteration, or replacement of the entire exon or a portion of the exon or multiple exons or the entire open reading frame of a gene, or the entire gene. Alternatively, repair of the mutant allele sequence is template free.


According to embodiments of the present invention, the method comprises subjecting the mutant allele to error prone non-homologous end joining (NHEJ) mechanism. According to preferred embodiments of the present invention, the method comprises correction or repair of a mutant allele with a template via a homology-directed repair mechanism after a targeted double-strand break in order to remove the haploinsufficiency manifestations of the mutant allele.


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: 1-44348 and a CRISPR nuclease repairing/correcting/modifying a mutant GATA2 allele in a cell, comprising delivering to the cell the 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: 1-44348 and the CRISPR nuclease.


In embodiments of the present invention, the 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: 1-44348.


One aspect of the present invention also provides for a method for modifying in a cell a mutant allele of the GATA2 gene having a mutation associated with MonoMAC syndrome, the method comprising

    • introducing to the cell a composition comprising:
      • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
      • an 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 RNA molecule affects a double strand break in a non-coding region of the GATA2 gene at a location that is 250 or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome.


One aspect of the present invention also provides for a method for modifying in a cell a mutant allele of the GATA2 gene having a mutation associated with MonoMAC syndrome, the method comprising

    • introducing to the cell a composition comprising:
      • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
      • an 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 RNA molecule targets the CRISPR nuclease to the mutation associated with MonoMAC syndrome.


One aspect of the present invention also provides for a method for modifying in a cell a mutant allele of the GATA2 gene having a mutation associated with MonoMAC syndrome, the method comprising

    • introducing to the cell a composition comprising:
      • a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; and
      • an 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 RNA molecule targets the CRISPR nuclease to a SNP position of the mutant allele.


Embodiments of the present invention further comprise introduction of a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele. In some embodiments, the donor template for HDR (e.g., dsDNA or ssODN) is at least 10, 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 1,000, 1,250, 1,500, 2,000, 3,000, or 5,000 base pairs in length, at most 20, 30, 40, 50, 100, 200, 300, 400, 500, 600, 700, 800, 1,000, 1,250, 1,500, 2,000, 3,000, or 5,000 base pairs in length, or between 10 and 5000, 10 and 3000, 10 and 2000, 10 and 1000, 10 and 500, 10 and 400, 10 and 300, 10 and 200, 10 and 100, 10 and 60, 10 and 50, 20 and 5000, 20 and 3000, 20 and 2000, 20 and 1000, 20 and 500, 20 and 400, 20 and 300, 20 and 200, 20 and 100, 20 and 60, 20 and 50, 30 and 5000, 30 and 3000, 30 and 2000, 30 and 1000, 30 and 500, 30 and 400, 30 and 300, 30 and 200, 30 and 100, 30 and 60, or 30 and 50 base pairs in length. Each possibility represents a separate embodiment. In some embodiments, the donor template for HDR overlaps in sequence complementarity with regions upstream and downstream double-strand break site induced by a nuclease programmed by any one of the guides described herein. In some embodiments, the donor template for HDR comprises a sequence that overlaps in sequence complementarity with an entire exon or more.


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


In some embodiments, the method comprises the steps of: contacting a mutant 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 e.g., Cas9 associate with a nucleotide sequence of the mutant allele of the gene of interest which differs by at least one nucleotide from a nucleotide sequence of a second allele of the gene of interest (i.e., a functional/wild type allele), thereby modifying the mutant allele.


Treatment of MonoMAC Syndrome

GATA binding protein 2 (GATA2) is a key transcriptional regulator of hematopoiesis required for the development and maintenance of a healthy stem cell pool. Mutations in GATA2 lead to loss of dendritic cells (DCs), monocytes, B cells and Natural Killer (NK) lymphoid cells (DCML deficiency/MonoMAC). In addition, GATA2 mutations are associated with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). These autosomal dominant mutations cause a reduction, and haploinsufficiency, in the cellular levels of the gene product. Therefore, therapeutic strategies to alleviate such conditions may include HDR-mediated approaches.


Editing strategies include (1) Correcting a pathogenic mutation in a cell by introducing a composition comprising a nuclease and a guide designed to specifically target each mutation, and a donor (e.g. ssODN, dsDNA (PCR product), or AAV) used to correct the mutation; and (2) Replacing the GATA2 exon harboring the pathogenic mutation. GATA2 (NM_032638) contains six exons (See FIG. 1A), five of which, Exons 2-6, encode the GATA2 protein. Pathogenic mutations are distributed along the gene. Accordingly, the second strategy would enable consolidation of all mutations located in a specific exon to a single composition or two compositions.


The second strategy of replacing exons harboring mutations can be accomplished, for example, in the following approaches: (1) Targeting a SNP that is linked to a mutation and located in the vicinity of an edge of an exon. In this approach, a composition comprising a nuclease and a guide are designed to specifically target the reference or the alternative form of the SNP according to which form is linked to the mutation. A long donor (e.g. a ssODN, dsDNA (PCR product) or AAV) will then be used to replace the mutated exon. For this approach, SNPs that are located up to 250 bps from the edge of an exon are prioritized (Table A) because they are close enough to the exon and the targeted mutation to enable efficient HDR.


A second approach to replace exons harboring mutations involves: (2) Non-discriminatory targeting of an intron region, such that the target is located in the vicinity of an edge of an exon. In this approach, a nuclease mediated double-strand break (DSB) will occur in both GATA2 alleles near an edge of an exon but without disrupting regulatory sequences such as splicing elements. A long donor that e.g., a ssODN, dsDNA (PCR product) or AAV will be used to replace the mutated exon. The rationale behind this approach is that DSBs in an intron are repaired either by HDR or NHEJ without disrupting regulatory elements, and thus would not interfere with the expression of the WT GATA2 allele.


In the second strategy of replacing the exon harboring the pathogenic mutation, the sequence of the donor(s) would cover the whole exon and may include synonymous mutations to increase the efficiency of the HDR process and the correction of mutations located at various distances from a DSB.









TABLE A





SNPs for use in GATA2 therapeutic strategies. The table presents the


distance of each SNP relative to the start and end of each exon.


















Exon2
Exon 3















SNP
Position



Start
End
Start
End


Position
GRCh37/hg19
rsID
Ref.
Alt.
128205919
128205646
128205201
128204570





Exon_2 of 6
Chr3: 128205860
rs1573858
G
C
59
−214
−659
−1290


Intron_2 of 5
Chr3: 128205519
rs4577488
G
A
400
127
−318
−949


Exon_3 of 6
Chr3: 128204951
rs2335052
C
T
968
695
250
−381


Intron_3 of 5
Chr3: 128203035
rs2713602
A
G
2884
2611
2166
1535


Intron_3 of 5
Chr3: 128203006
rs2659689
G
T
2913
2640
2195
1564


Intron_4 of 5
Chr3: 128201013
rs11717152
A
C
4906
4633
4188
3557


Intron_4 of 5
Chr3: 128200806
rs11708606
G
A
5113
4840
4395
3764


Intron_5 of 5
Chr3: 128200534
rs2713603
G
A
5385
5112
4667
4036


Intron_5 of 5
Chr3: 128200459
rs2713604
T
C
5460
5187
4742
4111


Exon_6 of 6
Chr3: 128199662
rs10934857
G
A
6257
5984
5539
4908














Exon 4
Exon 5
Exon 6















SNP
Start
End
Start
End
Start
Stop codon



Position
128202848
128202703
128200787
128200662
128200161
128199862







Exon_2 of 6
−3012
−3157
−5073
−5198
−5699
−5998



Intron_2 of 5
−2671
−2816
−4732
−4857
−5358
−5657



Exon_3 of 6
−2103
−2248
−4164
−4289
−4790
−5089



Intron_3 of 5
−187
−332
−2248
−2373
−2874
−3173



Intron_3 of 5
−158
−303
−2219
−2344
−2845
−3144



Intron_4 of 5
1835
1690
−226
−351
−852
−1151



Intron_4 of 5
2042
1897
−19
−144
−645
−944



Intron_5 of 5
2314
2169
253
128
−373
−672



Intron_5 of 5
2389
2244
328
203
−298
−597



Exon_6 of 6
3186
3041
1125
1000
499
200







The gray boxes mark the SNPs relevant to replace each exon via HDR.



Positive values indicate that the SNP is located upstream to the exon and negative values indicate that the SNP is located downstream to the exon.






In embodiments of the present invention, the RNA molecules, compositions, methods, cells, kits, or medicaments are utilized for treating a subject having a disease phenotype resulting from the heterozygote GATA2 gene. In embodiments of the present invention, the disease is MonoMAC syndrome. In such embodiments, the method results in improvement, amelioration or prevention of the disease phenotype. Any one of, or combination of, the strategies for bi-allelelic cut and correction or allele-specific targeting based on a heterozygous SNP or mutation sequence disclosed herein may be used in the context of the invention.


In embodiments of the present invention, the RNA molecules, compositions, methods, cells, kits, or medicaments are utilized for treating a subject having a disease phenotype resulting from the GATA2 gene. In embodiments of the present invention, the disease is MonoMAC syndrome. In such embodiments, the method results in improvement, amelioration or prevention of the disease phenotype.


In embodiments of the present invention, the RNA molecules, compositions, methods, cells, kits, or medicaments of the present invention are utilized in combination with a second therapy for MonoMAC syndrome to treat the subject. In embodiments of the present invention, the RNA molecules, compositions, methods, kits, or medicaments of the present invention are administered prior to administration of the second therapy, during administration of the second therapy, and/or after administration of the second therapy.


In embodiments of the present invention, a cell with a GATA2 gene mutation associated with MonoMAC syndrome may be from a subject with the GATA2 gene mutation and/or afflicted with MonoMAC syndrome. Accordingly, selecting a cell with a GATA2 gene mutation may comprise selecting a subject with the GATA2 gene mutation. In further embodiments of the present invention, selecting a cell may comprise selecting a cell from a subject with the GATA2 gene mutation. In embodiments of the present invention, introducing the compositions of the subject invention to the cell may comprise introducing the compositions of the invention to the cell of a subject afflicted with the GATA2 gene mutation.


Accordingly, embodiments of the present invention encompass the screening of subjects or cells for the GATA2 gene. A person having ordinary skill in the art would readily understand methods of screening for mutations within the GATA2 gene in the art, by way of non-limiting examples, e.g., sequencing-by-synthesis, Sanger sequencing, karyotyping, Fluorescence In situ Hybridization, and/or microarray testing. In embodiments of the present invention, mutations within the GATA2 gene are screened by exon sequencing. In one embodiment of the invention, a subject is screened for heterozygous MonoMAC syndrome mutation or SNPs prior to treatment with a composition that includes guide sequences of the present invention.


In embodiments of the present invention, a subject diagnosed with MonoMAC syndrome is screened by exon sequencing to identify a GATA2 pathogenic mutation in the GATA2 gene.


It is understood that the CRISPR/Cas9 gene editing system enables targeting the nuclease to a target site in a sequence specific manner to address disease-causing mutations. Hematopoietic stem and progenitor cells (HSPCs) have therapeutic potential because of their ability to both self-renew and differentiate (Yu et al., Human Gene Therapy, 2016). Accordingly, embodiments of the present invention apply genome editing to HSPCs.


In embodiments of the present invention, an autologous therapy and utilizes autologous CD34+ hematopoietic stem cells from patients diagnosed with MonoMAC syndrome which are edited with CRISPR/Cas9. In embodiments of the present invention, CD34+ cells are isolated from bone marrow or peripheral blood mononucleated cells (PBMCs) following patient apheresis.


In some embodiments, an allele-specific or bi-allelic targeting RNA molecule and a CRISPR nuclease are 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 a eukaryotic cell. In some embodiments, the cell encoding the gene of interest is a mammalian cell.


In some embodiments, a nucleic acid template is further introduced to the cell encoding the gene of interest for homology-directed repair, alteration, or replacement of a target sequence of the gene of interest to correct/repair the gene of interest such as to express a functional protein.


In some embodiments, the mutant allele desired to be targeted for correction or modification is an allele of the GATA2 gene. In some embodiments, the RNA molecule targets a SNP which co-exists with or is genetically linked to the disease-causing mutation associated with MonoMAC syndrome genetic disorder. In some embodiments, the RNA molecule targets a SNP which is highly prevalent in the population and exists in a mutant allele having the mutated sequence associated with MonoMAC syndrome genetic disorder and not in a second allele (e.g., functional allele) of an individual subject to be treated. In some embodiments, a disease-causing mutation within a mutated GATA2 allele is targeted. In some embodiments, a non-discriminatory guide sequence targets a non-coding region common to both GATA2 alleles up to 250 base pairs from an exon bearing a pathogenic mutation of interest.


In some embodiments, the method is utilized for treating a subject having a disease phenotype resulting from a mutation in the GATA2 gene. In such embodiments, the method results in improvement, amelioration or prevention of the disease phenotype.


Embodiments referred to above refer to a CRISPR nuclease, RNA molecule(s), and optionally tracrRNA being effective in a subject or cells at the same time. The CRISPR, RNA molecule(s), and optionally tracrRNA can 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 tracr RNA is substantially extant in the subject or cells.


In one embodiment, the cell is a stem cell. In one embodiment, the cell is an embryonic stem cell. In some embodiment, the stem cell is a hematopoietic stem/progenitor cell (HSC). As used herein, the term HSC refers to both hematopoietic stem cells and hematopoietic stem progenitor cells. Non-limiting examples of stem cells include bone marrow cells, myeloid progenitor cells, a multipotent progenitor cells, a lineage restricted progenitor cells.


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 a dominant genetic disorders such as, for example MonoMac syndrome. In some embodiments, the target gene is the GATA binding protein 2 (GATA2) gene (Entrez Gene, gene ID No: 2624) which is located on chromosome 3 and encodes a member of the GATA family of zinc-finger transcription factors. GATA2 pathogenic mutations may be corrected or modified by, for example, any one of the following strategies: (1) Utilizing a guide sequence targeting the pathogenic mutation itself to induce a DSB in proximity to the mutation in the mutant allele, and then using HDR for correction; (2) Utilizing a guide sequence targeting a SNP upstream or downstream of the pathogenic mutation, or upstream or downstream of an exon bearing the pathogenic mutation, and then using HDR for correction; or (3) Utilizing a non-discriminatory guide sequence that targets a non-coding region common to both alleles of the gene and is up to 250 base pairs from an exon bearing a pathogenic mutation to induce a DSB in the non-coding region of both alleles, and then using HDR for correction of the mutation and/or correction or replacement of the exon bearing the pathogenic mutation.


CRISPR Nucleases and PAM Recognition

In some embodiments, the sequence specific nuclease is an RNA guided DNA nuclease. In some embodiments, the RNA sequence which guides the RNA guided DNA nuclease binds to and directs the RNA guided DNA nuclease to (1) a sequence comprising at least one nucleotide which differs between a mutant allele and its counterpart functional allele (e.g., SNP or a pathogenic MonoMAC syndrome mutation) or (2) to a non-coding region common to both alleles of a gene bearing the pathogenic mutation (i.e., both the functional allele and the allele bearing the pathogenic mutation). In additional embodiments, the RNA sequence which guides the RNA guided DNA nuclease binds to and directs the RNA guided DNA nuclease to a sequence at a distance of up to 1000 basepairs from an edge of an exon bearing a pathogenic mutation. In some embodiments, the CRISPR complex further comprises a tracrRNA. In some embodiments, the sequence specific nuclease is selected from CRISPR nucleases, or functional variants thereof. In a non-limiting example, in which the RNA guided DNA nuclease is a CRISPR protein, the at least one nucleotide which differs between a target allele (e.g., bearing a pathogenic mutation) and the other allele (e.g., bearing a different or the same pathogenic mutation) 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. In another non-limiting example, 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 TTTV 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.


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. Non-limiting examples of suitable PAM sequences include NGG, NGAN, NGNG, NGAG, NGCG, NNGRRT, NNNNGATT, TTTV, NNNVRYM, NRTAM, NAG, NNYAAT, NRTAM, NRRAR, NGGNR, NRTAH, NGGNG, and NRRWC.


In some embodiments, an RNA-guided DNA nuclease e.g., a CRISPR nuclease, may be used to induce a double-strand DNA break 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, type III or Type V system. Non-limiting examples of suitable CRISPR proteins include Cas3, Cas4, Cas5, Cas5e (or CasD), Cas6, Cas6e, Cas6f, Cas7, Cas8a1, Cas8a2, Cas8b, Cas8c, Cas9, Cas10, Cas12a, Cas12b, Cas12c, Cas12d, Cas1Od, 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, Csx17, Csx14, Csx10, Csx16, CsaX, Csx3, Csz1, Csx15, Csf1, Csf2, Csf3, Csf4, and Cul966 (See Koonin 2017).


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, Ammonifex degensii, Caldicelulosiruptor becscii, Candidatus Desulfbrudis, Clostridium botulinum, Clostridium difficile, 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, Francisella cf. novicida Fx1, Alicyclobacillus acidoterrestris, Oleiphilus sp., Bacterium CG09_39_24, Deltaproteobacteria bacterium, 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., Cell (2015).


In some embodiments, the CRISPR nuclease comprises one or more nuclear localization sequences (NLS), cell penetrating peptide sequences, and/or affinity tags. The NLS may be fused to the CRISPR nuclease or may be an in internal NLS. In an embodiment, the CRISPR nuclease comprises one or more nuclear localization sequences of sufficient strength to drive accumulation of a CRISPR complex comprising the CRISPR nuclease in a detectable amount in the nucleus of a eukaryotic cell.


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 one or more of: 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-yOcarbamoyOthreonine, 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′-O-methyl (M), 3′-phosphorothioate (MS), 3′-thioPACE (MSP), pseudouridine, or 1-methyl pseudo-uridine. Each possibility represents a separate embodiment of the present invention.


Further non-limiting examples of suitable chemical modifications include: m1A (1-methyladenosine); m2A (2-methyladenosine); Am (2′-O-methyladenosine); ms2 m6A (2-methylthio-N6-methyladenosine); i6A (N6-isopentenyladenosine); ms2i6A (2-methylthio-N6isopentenyladeno sine); io6A (N6-(cis-hydroxyisopentenyl)adenosine); ms2io6A (2-methylthio-N6-(cis-hydroxyisopentenyl) adenosine); g6A (N6-glycinylcarbamoyladenosine); t6A (N6-threonylcarbamoyladenosine); ms2t6A (2-methylthio-N6-threonyl carbamoyladenosine); m6t6A (N6-methyl-N6-threonylcarbamoyladenosine); hn6A(N6-hydroxynorvalylcarbamoyladenosine); ms2hn6A (2-methylthio-N6-hydroxynorvalyl carbamoyladenosine); Ar(p) (2′-O-ribosyladenosine (phosphate)); I (inosine); m1I (1-methylinosine); m1Im (1,2′-O-dimethylinosine); m3C (3-methylcytidine); Cm (2′-O-methylcytidine); s2C (2-thiocytidine); ac4C (N4-acetylcytidine); f5C (5-formylcytidine); m5Cm (5,2′-O-dimethylcytidine); ac4Cm (N4-acetyl-2′-O-methylcytidine); k2C (lysidine); m1G (1-methylguanosine); m2G (N2-methylguanosine); m7G (7-methylguanosine); Gm (2′-O-methylguanosine); m22G (N2,N2-dimethylguanosine); m2Gm (N2,2′-O-dimethylguanosine); m22Gm (N2,N2, 2′-O-trimethylguanosine); Gr(p) (2′-O-ribosylguanosine (phosphate)); yW (wybutosine); o2yW (peroxywybutosine); OHyW (hydroxywybutosine); OHyW* (undermodified hydroxywybutosine); imG (wyosine); mimG (methylwyosine); Q (queuosine); oQ (epoxyqueuosine); galQ (galactosyl-queuosine); manQ (mannosyl-queuosine); preQ0 (7-cyano-7-deazaguanosine); preQ1 (7-aminomethyl-7-deazaguanosine); G+ (archaeosine); D (dihydrouridine); m5Um (5,2′-O-dimethyluridine); s4U (4-thiouridine); m5s2U (5-methyl-2-thiouridine); s2Um (2-thio-2′-O-methyluridine); acp3U (3-(3-amino-3-carboxypropyl)uridine); ho5U (5-hydroxyuridine); mo5U (5-methoxyuridine); cmo5U (uridine 5-oxyacetic acid); mcmo5U (uridine 5-oxyacetic acid methyl ester); chm5U (5-(carboxyhydroxymethyl)uridine)); mchm5U (5-(carboxyhydroxymethyl)uridine methyl ester); mcm5U (5-methoxycarbonylmethyluridine); mcm5Um (5-methoxycarbonylmethyl-2′-O-methyluridine); mcm5s2U (5-methoxycarbonylmethyl-2-thiouridine); nm5S2U (5-aminomethyl-2-thiouridine); mnm5U (5-methylaminomethyluridine); mnm5s2U (5-methylaminomethyl-2-thiouridine); mnm5se2U (5-methylaminomethyl-2-selenouridine); ncm5U (5-carbamoylmethyluridine); ncm5Um (5-carbamoylmethyl-2′-O-methyluridine); cmnm5U (5-carboxymethylaminomethyluridine); cmnm5Um (5-carboxymethylaminomethyl-2′-O-methyluridine); cmmm5s2U (5-carboxymethylaminomethyl-2-thiouridine); dimethyladenosine); Im (2′-O-methylinosine); m4C (N4-methylcytidine); m4Cm (N4,2′-O-dimethylcytidine); hm5C (5-hydroxymethylcytidine); m3U (3-methyluridine); cm5U (5-carboxymethyluridine); m6Am (N6,2′-O-dimethyladenosine); m6 2Am (N6,N6,O-2′-trimethyladenosine); m2,7G (N2,7-dimethylguanosine); m2,2,7G (N2,N2,7-trimethylguanosine); m3Um (3,2′-O-dimethyluridine); m5D (5-methyldihydrouridine); f5Cm (5-formyl-2′-O-methylcytidine); m1 Gm (1,2′-O-dimethylguanosine); m1Am (1,2′-O-dimethyladenosine); τm5U (5-taurinomethyluridine); τm5s2U (5-taurinomethyl-2-thiouridine)); imG-14 (4-demethylwyosine); imG2 (isowyosine); or ac6A (N6-acetyladenosine). Each possibility represents a separate embodiment of the present invention. (See e.g. U.S. Pat. No. 9,750,824).


Delivery to Cells

It is understood that in the methods embodied, the RNA molecules and compositions described herein may be delivered to a target cell or subject by any suitable means. The following embodiments provide non-limiting examples of methods of delivery of the RNA molecules and composition of the present invention.


In some embodiments, RNA molecule compositions of the present invention may be targeted to any cell which contains and/or expresses a mutant allele or a dominant negative allele, including any mammalian or plant cell. For example, in one embodiment the RNA molecule specifically targets a mutated GATA2 allele and the target cell is an HSC. The delivery to the cell may be performed in-vitro, ex-vivo, or in-vivo. Further, the nucleic acid compositions described herein may be delivered as one or more of DNA molecules, RNA molecules, Ribonucleoproteins (RNP), nucleic acid vectors, or any combination thereof. In another example, the RNA molecule targets non-coding region common to both alleles of the GATA2 gene and up to 250 base pairs from an exon bearing a pathogenic mutation position of interest. In such a scenario, a double-strand break is induced in the non-coding region of both alleles, and the mutation of interest can be corrected by DNA repair pathway (e.g. HDR).


In some embodiments, the RNA molecule comprises a chemical modification. Non-limiting examples of suitable chemical modifications include 2′-0-methyl (M), 2′-0-methyl, 3′phosphorothioate (MS) or 2′-0-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 RNA molecule 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) Science 256:808-813; Nabel and Feigner (1993) TIBTECH 11:211-217; Mitani and Caskey (1993) TIBTECH 11:162-166; Dillon (1993) TIBTECH 11:167-175; Miller (1992) Nature 357:455-460; Van Brunt (1988) Biotechnology 6(10):1149-1154; Vigne (1995) Restorative Neurology and Neuroscience 8:35-36; Kremer and Perricaudet (1995) British Medical Bulletin 51(1):31-44; Haddada et al. (1995) in Current Topics in Microbiology and Immunology Doerfler and Bohm (eds.); and Yu et al. (1994) Gene Therapy 1:13-26.


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) Trends Plant Sci. 11(1):1-4). 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 or in vitro delivery method. (See Zuris et al. (2015) Nat. Biotechnol. 33(1):73-80; see also Coelho et al. (2013) N. Engl. J. Med. 369, 819-829; Judge et al. (2006) Mol. Ther. 13, 494-505; and Basha et al. (2011) Mol. Ther. 19, 2186-2200).


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 of Felgner et al. (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 (1995) Science 270:404-410; Blaese (1995) Cancer Gene Ther. 2:291-297; Behr et al. (1994) Bioconjugate Chem. 5:382-389; Remy et al. (1994) Bioconjugate Chem. 5:647-654; Gao and Huang (1995) Gene Therapy 2:710-722; Ahmad and Allen (1992) Cancer Res. 52:4817-4820; 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 EnGenelC 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) Nature Biotechnology 27(7):643).


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 and Panganiban (1992) J. Virol. 66:2731-2739; Johann et al. (1992) J. Virol. 66:1635-1640; Sommerfelt et al. (1990) Virol. 176:58-59; Wilson et al. (1989) J. Virol. 63:2374-2378; Miller et al. (1991) J. Virol. 65:2220-2224; PCT/US1994/05700).


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 (Dunbar et al. (1995) Blood 85:3048-305; Kohn et al. (1995) Nat. Med. 1:1017-102; Malech et al. (1997) PNAS 94:22 12133-12138). PA317/pLASN was the first therapeutic vector used in a gene therapy trial. (Blaese et al., Science (1995). Transduction efficiencies of 50% or greater have been observed for MFG-S packaged vectors. (Ellem et al. (1997) Immunol Immunother. 44(1):10-20; Dranoff et al. (1997) Hum. Gene Ther. 1:111-2).


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) Proc. Natl. Acad. Sci. USA 92:9747-9751, 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, usually after selection for cells which have incorporated the vector.


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 et al. (1994) Culture of Animal Cells, A Manual of Basic Technique, 3rd ed, 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, CHOK1SV), 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., J. Exp. Med. 176:1693-1702 (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 Tad (differentiated antigen presenting cells) (as a non-limiting example see Inaba et al. (1992) J. Exp. Med. 176:1693-1702). Stem cells that have been modified may also be used in some embodiments.


Typically, the cells are administered in a pharmaceutical composition comprising at least one pharmaceutically-acceptable carrier. The phrase “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. The phrase “pharmaceutically acceptable carrier” as used herein means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, or solvent encapsulating material.


Any one of the RNA molecule compositions described herein is suitable for genome editing in either mitotic cells or post-mitotic cells or any cell which is not actively dividing, e.g., arrested cells. Examples of post-mitotic cells which may be edited using an RNA molecule composition of the present invention include, but are not limited to, a hepatocyte cell.


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 (e.g., a CRISPR nuclease) to a sequence comprising at least one nucleotide which differs between a mutant allele and a functional allele (e.g., SNP or a disease associated mutation) of a gene of interest (i.e., a sequence of the mutant allele which is not present in the functional allele, a sequence of one of the mutant alleles which is not present in the other mutant allele) or a sequence common to two mutant alleles of a gene of interest. 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 mutant 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. In additional embodiments, there is provided an RNA molecule which binds to/associates with and/or directs the RNA guided DNA nuclease to a sequence in a non-coding region upstream or downstream to an exon baring a disease associated mutation.


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


Guide Sequences which Specifically Target a Mutant Allele


A given gene may contain thousands of SNPs. Utilizing a 24 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 on aspect of the present invention, a novel set of guide sequences have been identified for repairing/correcting/modifying a mutant allele of GATA2 gene to treat/ameliorate/prevent MonoMAC syndrome.


One aspect of the present disclosure provides guide sequences capable of specifically targeting a mutant allele while leaving a second allele (e.g., functional allele) unmodified. The guide sequences of the present invention are designed to, and are most likely to, specifically differentiate between a mutant allele and a functional allele. Of all possible guide sequences which target a mutant allele desired to be edited/corrected/modified/repaired, the specific guide sequences disclosed herein are specifically effective to function with the disclosed embodiments.


The present disclosure also provides guide sequences capable of specifically targeting a mutant allele. Of all possible guide sequences which target the mutant allele desired to be edited/corrected/modified/repaired, 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) targets a SNP/insertion/deletion/indel with a prevalence of heterozygosity in the general population, in a specific ethnic population, or in a patient population above 1% or 10%; (2) targets a location of a SNP/insertion/deletion/indel proximal to a portion of the gene e.g., within 250 base pairs of a mutation or exon of the gene; and (3) targets a mutant allele which by targeting a founder or common pathogenic mutation for the disease/gene. In some embodiments, the SNP/insertion/deletion/indel heterozygosity rate in a population of interest 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.


Guide sequences of the present invention may target a SNP position in a distance of less than 2000 nucleotides, 1000 nucleotides, 500 nucleotides, 400 nucleotides, 300 nucleotides, 250 nucleotides, 200 nucleotides, 100 nucleotides from the disease-associated mutation, or within the disease associated mutation. Each possibility represents a separate embodiment.


Guide sequences of the present invention also may: (1) target a heterozygous sequence in a SNP position for the targeted gene; (2) target a SNP position 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% or 10%; (3) have a guanine-cytosine content of greater than 30% and less than 85%; (4) have no repeats of seven or more thymine/uracil, guanine, cytosine, or adenine residues; (6) have no off-target of zero mismatches in the genome identified by off-target analysis; and (7) preferably target Exons over Introns or be upstream of a SNP rather than downstream of a SNP.


In embodiments of the present invention, the SNP position may be upstream or downstream of the gene. In embodiments of the present invention, the SNP position is within 4,000, 2000, 1000, 500 base pairs upstream or downstream of the gene. Each possibility represents a separate embodiment.


The at least one nucleotide which differs between the mutant allele and the second allele (e.g., functional/wild type allele), may be 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 mutant allele and the second allele, may be within an exon or within an intron of the gene of interest.


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 mutant allele (REF/SNP) and targeted by a guide sequence. In some embodiments, the SNP may be a founder or common pathogenic mutation.


Guide sequences may target a REF/ALT sequence of 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 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 mutant allele and the functional allele is linked to/co-exists with the disease-causing mutation in high prevalence in a population. In one embodiment, the at least one nucleotide which differs between the mutant 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.


Guide sequences of the present invention may satisfy any one of the above criteria and are most likely to differentiate between a mutant allele from its corresponding functional allele.


Strategies for HDR repair of a pathogenic mutation associated with MonoMAC syndrome may involve a guide sequence targeting the pathogenic mutation itself or alternatively a guide sequence targeting a SNP located upstream or downstream to the mutation to mediate a DSB in proximity to the mutation. The strategies may further include a sequence repair/correction step by utilizing a donor/template sequence that (e.g., a single-stranded donor oligonucleotides (ssODN), double-stranded Donor (PCR product), Minicircle or virus (rAAV or Lentivirus)). In some embodiments, a DSB is affected in a non-coding region of both alleles of a gene, and a donor template for HDR overlaps in sequence complementarity with an entire exon in which a pathogenic mutation is located.


In an exemplary strategy, a mutant allele bearing a disease associated mutation, such as a mutation selected at one of the following positions rs387906630, rs387906632, rs387906631, rs387906634, rs376003468, rs387906633, 3:128200118_C_T, and rs387906629, is targeted by guide sequences designed to target the disease associated mutation itself. Non-limiting examples of such guide sequences are indicated in Table 2.


In some embodiments, a mutant allele bearing a mutation is targeted by guide sequences designed to target REF/ALT sequences of SNPs located up to 200, 500, 1000, 1500, 2000 bases from the pathogenic mutation. Each possibility represents a separate embodiment. Non-limiting examples of suitable guide sequences are indicated in Table 2.


In an exemplary strategy, a mutant allele bearing a mutation may be targeted by guide sequences designed to target ALT/REF sequences in SNPs located upstream of the GATA2 gene, downstream of the GATA2 gene, in intron 1, intron 2, intron 3, intron 4, intron 5, exon 1, exon 2, exon 3, exon 4, exon 5, exon 6 of GATA2. Non-limiting examples of such guide sequences are indicated in Table 2.


In an exemplary strategy, a mutant allele bearing a mutation may be targeted by guide sequences designed to target ALT/REF sequences in SNPs located in intron 2 (e.g., r54577488), intron 3 (e.g., rs2713602 and 2659686), intron 4 (e.g., rs12488487, rs55686439, rs11717152, and r511708606), intron 5 (e.g., 2713603 and 2713604), exon 2 (e.g., r51573858), exon 3 (e.g., rs2335052 and r5387906630), exon 4 (e.g., r5387906632), exon 5 (e.g., rs387906631, rs376003468, rs387906633, and r5387906634), exon 6 (e.g., rs387906629, rs3803, rs10934857, 3:128200118_C_T). Non-limiting examples of such guide sequences are indicated in Table 2.


Although a large number of guide sequences can be designed to target a mutant allele, a subset within the sequences identified by SEQ ID NOs: 1-44348 were specifically designed to effectively implement the methods set forth herein and to effectively discriminate between alleles.


A subset of guide sequences for use as described in the embodiments herein were designed to associate with different SNPs or mutations within a sequence of a mutated GATA2 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.VQR.2 (PAM SEQ: NGNG), SpCas9.EQR (PAM SEQ: NGAG), SpCas9.VRER (PAM SEQ: NGCG), SaCas9WT (PAM SEQ: NNGRRT), NmCas9WT (PAM SEQ: NNNNGATT), Cpf1 (PAM SEQ: TTTV), 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.


Guide Sequences which Target a Sequence within a Non-Coding Region in a Proximity to an Exon Bearing a Pathogenic Mutation


The present disclosure also provides guide sequences capable of specifically targeting a CRISPR nuclease to induce a DSB at a location that is 500, 250, or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome desired to be edited/corrected/modified/repaired. The specific guide sequences disclosed herein are specifically effective to function with the disclosed embodiments.


Strategies for HDR repair of a pathogenic mutation associated with MonoMAC syndrome may involve a guide sequence targeting a sequence within a non-coding region of the gene, in a distance of up to 1000, 500, 250, or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome desired to be edited/corrected/modified/repaired. The strategies may further include a sequence repair/correction step by utilizing a donor/template sequence that (e.g., a single-stranded donor oligonucleotides (ssODN), double-stranded Donor (PCR product), Minicircle or virus (rAAV or Lentivirus)). In some embodiments, the donor/template sequence comprises a sequence complimentary to a region extending from the position of the DSB to a pathogenic mutation. In such embodiments, the donor/template sequence further comprises homology arms flanking the site of the DSB and the pathogenic mutation.


In some embodiments, the donor/template sequence comprises a sequence complimentary to a region comprising an entire exon. In some embodiments, the exon containing the pathogenic mutation, or a portion thereof which contains the pathogenic mutation, is corrected or replaced to remove the pathogenic mutation.


In some embodiments, a mutant allele bearing a mutation and functional allele are targeted by guide sequences designed to target a sequences located in a non-coding region up to 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, bases from an edge of a coding region bearing the pathogenic mutation. Each possibility represents a separate embodiment. Non-limiting examples of suitable guide sequences that associate with different sequences in a non-coding region of a GATA2 gene are indicated in Table 2.


In the present invention, a novel set of guide sequences have been identified for repairing/correcting/modifying a mutant allele of GATA2 gene to treat, ameliorate, or prevent MonoMAC syndrome.









TABLE 2







Guide sequences designed for use as described in the embodiments herein. 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).











SEQ ID NOs of
SEQ ID NOs of
SEQ ID NOs of


Target
20 base guides
21 base guides
22 base guides





3:128479475_T_C
 1-45
46-89
 90-136


3:128479476_A_G
1-2, 11, 21, 137-177
50, 54, 178-219
95, 119, 123, 220-264


3:128479846_C_A
265-310
311-358
359-408


3:128479875_C_T
409-454
455-502
503-552


3:128480135_T_A
553-582
583-612
613-644


3:128480322_G_A
645-690
691-738
739-788


3:128480449_G_A
789-834
835-882
883-932


3:128480599_C_T
933-978
979-1026
1027-1076


3:128480829_G_A
1077-1122
1123-1170
1171-1220


3:128480845_C_T
1221-1266
1267-1314
1315-1364


3:128480918_C_T
1365-1410
1411-1448
1449-1494


3:128480935_T_C
1495-1540
1541-1586
1587-1636


3:128480946_G_A
1637-1682
1683-1730
1731-1780


3:128481039_T_A
1781-1826
1827-1872
1873-1921


3:128481047_G_A
1922-1967
1968-2015
2016-2065


3:128481055_G_T
2066-2111
2112-2159
2160-2209


3:128481060_C_T
2210-2255
2256-2303
2304-2353


3:128481071_C_A
2354-2399
2400-2447
2448-2497


3:128481077_G_T
2498-2543
2544-2591
2592-2641


3:128481090_G_A
2642-2687
2688-2735
2736-2785


3:128481092_G_A
2642, 2664, 2671,
2710, 2719, 2724,
2743, 2755, 2769,



2786-2828
2829-2873
2874-2920


3:128481095_G_A
2803, 2921-2965
2845, 2966-3012
2915, 3013-3061


3:128481102_G_A
3062-3107
3108-3155
3156-3205


3:128481115_G_T
3206-3251
3252-3299
3300-3349


3:128481131_G_A
3350-3395
3396-3443
3444-3493


3:128481136_G_A
3494-3539
3540-3587
3588-3637


3:128481142_C_T
3638-3683
3684-3731
3732-3781


3:128481147_G_A
3782-3827
3828-3875
3876-3925


3:128481150_C_A
3802, 3926-3970
3859,3971-4016
3881,4017-4065


3:128481162_C_G
4066-4111
4112-4159
4160-4209


3:128481176_C_G
4210-4255
4256-4303
4304-4353


3:128481180_A_T
4354-4399
4400-4447
4448-4497


3:128481188_G_A
4498-4543
4544-4591
4592-4641


3:128481189_A_G
4512, 4514, 4524,
4570, 4576, 4581,
4614, 4626, 4628,



4534, 4538, 4642-
4585, 4590, 4683-4725
4631, 4640, 4726-4770



4682


3:128481199_C_T
4771-4816
4817-4864
4865-4914


3:128481214_C_G
4915-4960
4961-5008
5009-5058


3:128481219_C_T
5059-5104
5105-5152
5153-5202


3:128481220_G_C
5060, 5069, 5073,
5111, 5116, 5131,
5159, 5173, 5179,



5083, 5085, 5203-
5133, 5147, 5244-5286
5181, 5197, 5287-5331



5243


3:128481224_C_T
5332-5377
5378-5425
5426-5475


3:128481226_C_A
5334, 5338, 5356,
5386, 5394, 5403,
5428, 5435, 5459,



5476-5518
5519-5563
5564-5610


3:128481230_G_A
5611-5656
5657-5704
5705-5754


3:128481230_G_T
5617, 5628, 5632,
5673, 5679, 5689,
5711, 5722, 5746,



5642, 5650, 5652,
5697, 5699, 5703-5704,
5748, 5752-5754, 5835-



5656, 5755-5793
5794-5834
5877


3:128481232_C_A
5617, 5650, 5652,
5673, 5689,5697,
5711,5722, 5752,



5878-5920
5921-5965
5966-6012


3:128481245_T_A
6013-6058
6059-6106
6107-6156


3:128481262_C_T
6157-6202
6203-6250
6251-6300


3:128481270_G_A
6301-6346
6347-6394
6395-6444


3:128481275_C_T
6445-6490
6491-6538
6539-6588


3:128481276_G_A
6445, 6450, 6466,
6491, 6512-6513,6531,
6544, 6551, 6561-6562,



6486, 6490, 6589-
6538, 6630-6672
6581, 6673-6717



6629


3:128481283_G_C
6718-6763
6764-6811
6812-6861


3:128481299_A_G
6862-6907
6908-6955
6956-7005


3:128481302_G_T
6880, 7006-7050
6947, 7051-7097
6976, 7098-7146


3:128481315_TAACC_T
7147-7192
7193-7240
7241-7290


3:128481318_C_A
7147, 7164, 7181,
7206, 7209, 7211,
7256, 7277-7279, 7377-



7189, 7291-7332
7229, 7333-7376
7422


3:128481819_A_G
7423-7464
7465-7506
7507-7550


3:128481836_A_AGAGG
7551-7604
7605-7660
7661-7718


3:128481841_C_T
7552, 7560, 7564,
7607, 7615, 7618,
7662, 7675, 7687,



7575, 7604, 7719-
7630, 7640, 7760-7802
7697, 7700, 7803-7847



7759


3:128481845_A_G
7848-7893
7894-7941
7942-7991


3:128481849_G_T
7992-8037
8038-8085
8086-8135


3:128481866_CGTTGGCGTTTCG_C
7867, 8136-8180
7914, 8181-8227
7963, 8228-8276


3:128481872_C_T
8277-8322
8323-8370
8371-8420


3:128481877_C_T
8137, 8146, 8163,
8182, 8205, 8210,
8235, 8253, 8258,



8421-8463
8464-8508
8509-8555


3:128481878_G_A
8137, 8146, 8158,
8182, 8188, 8205,
8235, 8253, 8258,



8163, 8422, 8431,
8210, 8465, 8488,
8260, 8515, 8535,



8556-8595
8596-8637
8638-8681


3:128481880_C_A
8158, 8163, 8561,
8188, 8205, 8607,
8235, 8260, 8640,



8682-8724
8725-8769
8770-8816


3:128481881_G_A
8158, 8687, 8704,
8188, 8730, 8732,
8260, 8775, 8777,



8711-8712, 8817-
8749, 8756, 8858-8900
8782-8783, 8901-8945



8857


3:128481881_G_C
8158, 8687, 8704,
8188, 8730, 8732,
8260, 8775, 8777,



8711-8712, 8823,
8749, 8756, 8869,
8782-8783, 8929, 8937,



8845, 8946-8984
8886, 8985-9025
9026-9068


3:128481887_A_C
9069-9114
9115-9162
9163-9212


3:128481897_TGTCG_AAGGC
9213-9258
9259-9306
9307-9356


3:128481898_G_T
9228, 9248, 9357-
9295, 9303, 9401-9446
9353, 9356, 9447-9494



9400


3:128481901_G_A
9372,9495-9539
9414, 9540-9586
9451,9587-9635


3:128481908_A_G
9636-9681
9682-9729
9730-9779


3:128481923_T_TGCCGGCTCTTCTGGCG
9780-9857
9858-9935
9936-10017


3:12848I924_G_A
9787, 9791, 9841,
9865, 9870, 9872,
9945, 9950, 9952-9953,



9844, 10018-10059
9919, 10060-10103
10104-10149


3:128481926_C_T
9787, 9791, 9793-
9870, 9872-9873, 9921,
9952-9953, 9968,



9794, 10056, 10150-
10083, 10191-10233
10003, 10145, 10234-



10190

10278


3:128481927_G_A
9791, 9793-9794,
9872-9873, 9888, 9921,
9953, 9959, 9968,



9843, 10154, 10174,
10216, 10228, 10319-
10003, 10258, 10273,



10279-10318
10360
10361-10404


3:128481937_G_A
9780, 9801, 9817,
9866, 9880, 9892,
9938, 9972, 9987,



9827, 9852, 10405-
9905, 9930, 10446-
9992, 10012, 10489-



10445
10488
10533


3:128481937_G_GCGGC
9790, 9801, 9804,
9860, 9880, 9882,
9936, 9940, 9960,



9812, 9817, 9827-
9884, 9892, 9896,
9962, 9964, 9972,



9828, 9832, 9834,
9905-9906, 9910, 9912,
9977, 9987-9988, 9992,



9836, 9849, 9852,
9914, 9930, 9935,
9994, 9996, 10012,



9857, 10534-10564
10565-10585
10586-10614


3:128481938_C_A
9780, 9782, 9801,
9858, 9866, 9892,
9938, 9946, 9962,



9812, 9827, 9852,
9905, 9910, 9930,
9972, 9992, 10012,



10410, 10615-10653
10447, 10654-10694
10495, 10695-10737


3:128481938_C_T
9780, 9782, 9801,
9858, 9866, 9892,
9938, 9946, 9962,



9812, 9827, 9852,
9905, 9910, 9930,
9972, 9992, 10012,



10410, 10738-10776
10447, 10777-10817
10495, 10818-10860


3:128481939_GGCCG_AAGGC
9780, 9782, 9790,
9858, 9860, 9866,
9936, 9938, 9946,



10861-10903
10904-10948
10949-10995


3:128481942_C_T
9832, 10534-10535,
9882, 10565-10566,
9940, 10586, 10589,



10543, 10555,
10568, 10575, 11037-
10598, 10611, 11080-



10996-11036
11079
11124


3:128481945_C_A
9798, 9825, 9850,
9903, 9928, 11072,
10010, 11095, 11212-



11014, 11125-11166
11167-11211
11259


3:128483288_G_A
11260-11305
11306-11353
11354-11403


3:128483333_C_T
11404-11449
11450-11497
11498-11547


3:128483347_C_T
11548-11593
11594-11641
11642-11691


3:128483375_T_A
11692-11703
11704-11713
11714-11727


3:128483868_G_A
11728-11773
11774-11821
11822-11871


3:128483889_G_A
11872-11917
11918-11965
11966-12015


3:128483905_CT_C
12016-12061
12062-12109
12110-12159


3:128483906_T_C
12016, 12021,
12062, 12068, 12070,
12110, 12114, 12116,



12023, 12038,
12085-12086, 12091,
12118, 12133, 12135,



12041, 12043,
12095, 12104, 12106,
12144, 12152, 12156,



12045, 12056,
12197-12235
12236-12276



12058, 12160-12196


3:128483925_C_CATTGCACAGGT
12277-12341
12342-12407
12408-12475


3:128483957_C_T
12331, 12334,
12396-12397, 12400,
12420, 12463-12464,



12476-12515
12516-12550
12551-12589


3:128483973_C_CTGTGGCCCCACAGT
12590-12663
12664-12739
12740-12817


3:128484010_A_C
12818-12863
12739, 12864-12910
12805, 12817, 12911-





12958


3:128485738_C_T
12959-13004
13005-13052
13053-13102


3:128485741_G_A
12976, 13103-13147
13023, 13148-13194
13058, 13195-13243


3:128485746_G_A
13244-13289
13290-13337
13338-13387


3:128485750_C_T
13388-13433
13434-13481
13482-13531


3:128485758_AG_A
12976-12977,
13024-13025, 13030,
13074-13075, 13620-



12982, 13532-13574
13575-13619
13667


3:128485766_A_T
13668-13713
13714-13759
13760-13808


3:128485769_T_C
13705, 13809-13845
13758, 13846-13879
13777, 13880-13920


3:128485779_T_TC
13921-13968
13969-14016
14017-14068


3:128485780_C_A
13922, 13926-
13970, 13974-13976,
14022-14024, 14058-



13928, 13930,
14002, 14008-14009,
14059, 14065-14066,



13956-13957,
14108-14148
14149-14191



14069-14107


3:128485783_C_T
13928, 14192-14236
14009, 14237-14283
14066, 14284-14332


3:128485789_A_C
14333-14369
14370-14386
14387-14408


3:128485795_C_A
14409-14454
14455-14501
14502-14551


3:128485798_G_A
14418, 14552-14596
14456, 14597-14643
14546, 14644-14692


3:128485811_C_T
14693-14738
14739-14786
14787-14836


3:128485819_T_TAGTCGTGGGC
14837-14902
14903-14968
14969-15038


3:128485837_G_A
15039-15084
15085-15132
15133-15182


3:128485837_G_C
15045, 15048,
15094, 15096-15097,
15138, 15145-15146,



15050-15051,
15110, 15115, 15125,
15158, 15165, 15175,



15064, 15069,
15129, 15222-15260
15179, 15261-15303



15083, 15183-15221


3:128485843_T_C
14858, 14883,
14907, 14960, 15347-
14974, 15018, 15025,



14894, 15304-15346
15386
15387-15433


3:128485850_G_A
14845, 14867,
14911, 14934, 14941,
15002, 15009, 15012,



15434-15477
15478-15522
15523-15569


3:128485855_G_A
15570-15615
15616-15663
15664-15713


3:128485869_G_C
15714-15759
15760-15807
15808-15857


3:128485871_G_A
15721, 15745,
15767, 15790, 15802,
15813, 15825, 15852,



15757, 15858-15900
15901-15945
15946-15992


3:128485874_T_C
15872, 15993-16037
15903, 16038-16084
15985, 16085-16133


3:128485891_A_G
16134-16179
16180-16227
16228-16277


3:128485892_T_C
16147, 16153,
16191, 16194, 16200,
16238, 16240, 16243,



16169, 16176,
16218, 16227, 16319-
16262, 16267, 16362-



16179, 16278-16318
16361
16406


3:128485892_T_G
16147, 16153,
16191, 16194, 16200,
16238, 16240, 16243,



16169, 16176,
16218, 16227, 16333,
16262, 16267, 16400,



16179, 16292,
16353, 16446-16486
16406, 16487-16529



16317, 16407-16445


3:128485909_C_T
16530-16575
16576-16623
16624-16673


3:128485910_G_A
16538, 16542-
16578, 16585, 16590,
16625, 16627, 16648,



16543, 16552,
16599, 16602, 16715-
16651, 16670, 16758-



16573, 16674-16714
16757
16802


3:128485916_G_T
16803-16848
16849-16896
16897-16946


3:128485924_C_T
16947-16992
16993-17038
17039-17087


3:128485929_C_T
17088-17133
17134-17181
17182-17231


3:128485937_T_G
17232-17277
17278-17325
17326-17375


3:128485944_C_CGTCAG
17376-17431
17432-17489
17490-17549


3:128485944_C_T
17376, 17387,
17443, 17454, 17469,
17512, 17522, 17528,



17412, 17419,
17476, 17592-17635
17535, 17636-17681



17550-17591


3:128485962_C_A
17682-17727
17728-17775
17776-17825


3:128485966_A_ACGC
17384, 17391,
17440, 17447, 17479,
17505, 17531, 17538,



17416, 17709,
17738, 17762, 17873-
17786, 17807, 17922-



17714, 17826-17872
17921
17972


3:128485967_C_G
17391, 17415,
17456, 17472, 17479,
17514, 17531, 17931,



17422, 17841-
17888-17889, 17893,
17938-17939, 17957,



17842, 17846,
17907, 18012-18052
18053-18096



17858,17973-18011


3:128485970_C_T
17384, 17416,
17440, 17447, 17473,
17498, 17505, 17538,



17835, 17841,
17882, 17887, 17907,
17931, 17937, 17956,



17859, 18097-18137
18138-18179
18180-18223


3:128485973_C_T
18110, 18224-18268
18167, 18269-18315
17532, 18220, 18316-





18363


3:128485982_C_G
18364-18409
18410-18457
18458-18507


3:128485998_A_AC
18508-18555
18556-18605
18606-18657


3:128485999_C_T
18511, 18514,
18562, 18564, 18575,
18614, 18625, 18642,



18516, 18521,
18592, 18596, 18599,
18646, 18651, 18655,



18543, 18547,
18604, 18697-18737
18657, 18738-18780



18550, 18658-18696


3:128486002_C_A
18547, 18781-18825
18575, 18826-18872
18651, 18873-18921


3:128486004_C_CGCGGAAGA
18801, 18814,
18845, 18860, 18868,
18892, 18904, 18909,



18821, 18922-18980
18981-19037
19038-19100


3:128486005_G_A
18814, 18949,
18845, 19010, 19020,
18904, 19071, 19083,



18964, 18979,
19036, 19143-19186
19099, 19187-19232



19101-19142


3:128486006_C_T
18949, 18953,
19010, 19020, 19022,
19071, 19083, 19087,



18964, 18979,
19036, 19160, 19181,
19099, 19211, 19226,



19118, 19124,
19273-19314
19315-19358



19233-19272


3:128486023_G_A
19359-19404
19405-19452
18615, 19453-19501


3:128486030_C_T
18922, 18926,
18981, 19011, 19023,
19064, 19073, 19084,



18955, 19502-19544
19545-19589
19590-19636


3:128486031_C_A
18922, 18926,
18981, 18986, 19011,
19038, 19073, 19084,



18957, 19517,
19565, 19585-19586,
19611, 19629, 19633,



19521, 19541,
19677-19718
19719-19762



19637-19676


3:128486038_G_C
19763-19808
19809-19856
19857-19906


3:128486057_C_T
19907-19952
19953-20000
20001-20050


3:128486059_T_G
19931, 19943,
19996, 19998-19999,
20031, 20043, 20049,



19949, 20051-20093
20094-20138
20139-20185


3:128486066_G_A
20186-20231
20232-20279
20280-20329


3:128486072_T_G
20330-20375
20376-20419
20420-20467


3:128486075_G_A
20341, 20468-20512
20408, 20513-20559
20439, 20560-20608


3:128486103_G_T
20609-20654
20655-20702
20703-20752


3:128486104_T_A
20632-20633,
20666, 20679, 20686,
20705, 20714, 20731,



20635, 20645,
20693, 20700, 20794-
20735, 20750, 20837-



20652, 20753-20793
20836
20881


3:128486117_G_A
20882-20927
20928-20975
20976-21025


3:128486129_C_A
21026-21071
21072-21119
21120-21169


3:128486141_C_T
21170-21214
21215-21256
21257-21302


3:128486142_G_T
21190, 21206-
21250, 21254, 21343-
21294, 21296, 21383-



21207, 21212,
21382
21426



21303-21342


3:128486143_C_T
21190, 21206,
21254, 21363, 21468-
21296, 21425, 21508-



21309, 21427-21467
21507
21551


3:128486153_C_T
21552-21597
21598-21633
21634-21678


3:128486173_G_A
21679-21722
21723-21767
21768-21814


3:128486179_A_G
21815-21860
21861-21908
21909-21958


3:128486185_A_G
21959-22004
22005-22050
22051-22100


3:128486189_G_A
22101-22146
22147-22194
22195-22244


3:128486201_C_T
22245-22290
22291-22338
22339-22388


3:128486218_T_C
22389-22434
22435-22482
22483-22532


3:128486227_G_T
22533-22578
22579-22626
22627-22676


3:128486240_G_A
22677-22722
22723-22770
22771-22820


3:128486252_A_AG
22821-22868
22869-22918
22919-22970


3:128486260_T_C
22971-23012
23013-23051
23052-23095


3:128486271_C_T
23096-23141
23142-23189
23190-23239


3:128486284_A_AGG
23240-23289
23290-23341
23342-23395


3:128486287_G_A
23273, 23279,
23312, 23330, 23336-
23359, 23365, 23389,



23286, 23288-
23337, 23339, 23437-
23393, 23395, 23480-



23289, 23396-23436
23479
23524


3:128486288_C_A
23279, 23288-
23312, 23337, 23339,
23359, 23365, 23389,



23289, 23415,
23452, 23479, 23566-
23494, 23522, 23608-



23432, 23525-23565
23607
23652


3:128486295_GC_G
23244, 23283,
23697-23744
23209, 23745-23793



23653-23696


3:128486296_C_T
23655, 23661,
23700, 23703, 23708,
23748, 23751, 23753,



23674, 23685-
23721,23733-23735,
23758, 23767, 23782-



23687, 23696,
23833-23873
23783, 23874-23916



23794-23832


3:128486298_CC_AA
23661, 23671,
23706, 23708, 23721,
23748, 23753, 23756,



23680, 23685,
23727, 23959-24002
23770, 24003-24048



23917-23958


3:128486320_G_T
24049-24094
24095-24142
24143-24192


3:128486355_T_GC
24193-24233
24234-24264
24265-24300


3:128486356_C_G
24215, 24222-
24236, 24247, 24250,
24265, 24269, 24281,



24224, 24227,
24257-24258,24261-
24284, 24291-24292,



24230-24233,
24264, 24335-24358
24294, 24298-24300,



24301-24334

24359-24386


3:128486365_C_CG
24387-24426
24427-24456
24457-24492


3:128486805_T_C
24493-24534
24535-24556
24557-24590


3:128486808_G_C
24525, 24591-24635
24543, 24636-24682
24579, 24683-24731


3:128486826_C_T
24732-24769
24770-24801
24802-24837


3:128486828_C_A
24732, 24737,
24800, 24873-24902
24833, 24903-24939



24753, 24838-24872


3:128486850_G_A
24940-24984
24985-25029
25030-25076


3:128486856_T_C
25077-25122
25123-25170
25171-25220


3:128486872_A_C
25221-25266
25267-25314
25315-25364


3:128486883_T_C
25365-25410
25411-25458
25459-25508


3:128486890_A_T
25509-25554
25555-25602
25603-25652


3:128486893_C_T
25551, 25653-25697
25574, 25698-25744
25635, 25745-25793


3:128486896_C_T
25694, 25794-25838
25718, 25839-25885
25778, 25886-25934


3:128486911_G_C
25935-25980
25981-26028
26029-26078


3:128486931_A_G
26079-26124
26125-26172
26173-26222


3:128486948_G_A
26223-26268
26269-26316
26317-26366


3:128486956_G_T
26367-26412
26413-26460
26461-26510


3:128486961_G_GACA
26511-26562
26563-26615
26616-26671


3:128486967_G_C
26532, 26672-26716
26580, 26717-26763
26648, 26764-26812


3:128486970_T_A
26701, 26813-26857
26733, 26858-26904
26776, 26905-26953


3:128486973_T_G
26819, 26954-26998
26859, 26999-27045
26912, 27046-27094


3:128486982_A_T
27095-27140
27141-27186
27187-27235


3:128486992_G_A
27236-27281
27282-27329
27330-27379


3:128487002_C_A
27380-27421
27422-27463
27464-27507


3:128487004_A_G
27382, 27508-27525
27428, 27526
27504, 27527-27530


3:128487009_G_T
27531-27576
27577-27613
27614-27657


3:128487016_CG_GC
27658-27691
27692-27715
27716-27743


3:128487016_C_T
27666, 27668-
27707, 27711-27712,
27738-27740, 27817-



27669, 27679-
27784-27816
27855



27680, 27688,



27744-27783


3:128487021_G_T
27856-27900
27901-27936
27937-27978


3:128487036_G_A
27979-28016
28017-28041
28042-28075


3:128487073_G_C
28076-28121
28122-28163
28164-28211


3:128492886_G_A
28212-28257
28258-28305
28306-28355


3:128492959_A_C
28356-28383
28384-28407
28408-28433


3:128493033_G_T
28434-28473
28474-28506
28507-28544


3:128493046_G_A
28545-28582
28583-28616
28617-28653


3:128493068_G_A
28654-28699
28700-28747
28748-28797


3:128480819_G_A
1078, 1085, 1094,
1124, 1131, 1136,
1179, 1184, 1186,


rs10934857 REF
28798-28840
28841-28885
28886-28932


3:128480819_G_A
28803, 28805-
28846, 28848, 28852,
28892, 28898, 28909,


rs10934857 SNP
28806, 28809,
28868-28869, 28872,
28914, 28918, 28929-



28825-28826,
28883, 28972-29012
28930, 29013-29055



28832, 28933-28971


3:128481616_T_C
29056-29101
29102-29149
29150-29199


rs2713604 REF


3:128481616_T_C
29063-29064,
29104, 29110-29111,
29152, 29158-29159,


rs2713604 SNP
29069, 29078,
29125, 29131-29132,
29179, 29182, 29185,



29084, 29086,
29134, 29239-29279
29191, 29280-29322



29100, 29200-29238


3:128481691_G_A
29323-29368
29369-29416
29417-29466


rs2713603 REF


3:128481691_G_A
29324, 29340,
29382, 29387, 29390,
29426, 29431, 29450,


rs2713603 SNP
29343, 29354,
29401, 29404, 29408,
29453, 29457, 29463,



29357, 29361,
29414, 29506-29546
29466, 29547-29589



29367, 29467-29505


3:128481963_G_A
10534, 10539,
10565, 10571, 10575,
9940, 9994, 10593,


rs11708606 REF
11014, 11142,
10577, 11072, 11185,
10598, 10600, 10611,



11153, 11161,
11197, 11208, 29630-
11095, 11231, 11243,



29590-29629
29669
11256, 29670-29709


3:128481963_G_A
10539, 29602,
10571, 10577, 29642,
9994, 10593, 10600,


rs11708606 SNP
29604, 29607,
29644, 29654, 29659,
29693, 29698, 29702,



29614, 29619,
29663, 29749-29789
29709, 29790-29832



29625, 29710-29748


3:128482170_A_C
29833-29878
29879-29926
29927-29976


rsll717152 REF


3:128482170_A_C
29839, 29850-
29882, 29898, 29901,
29927, 29931, 29947,


rs11717152 SNP
29851, 29862,
29910, 29914, 29917,
29950, 29959, 29967,



29866, 29869,
29922, 30016-30056
29972, 30057-30099



29874, 29977-30015


3:128484163_G_T
30100-30145
30146-30193
30194-30243


rs2659689 REF


3:128484163_G_T
30105, 30109,
30147, 30152, 30156,
30194, 30196, 30205,


rs2659689 SNP
30114, 30116,
30161-30162, 30164,
30210-30212,30214,



30120, 30131,
30168, 30283-30323
30324-30366



30140, 30244-30282


3:128484192_A_G
30367-30412
30413-30460
30461-30510


rs2713602 REF


3:128484192_A_G
30385, 30394,
30431, 30444, 30449,
30471, 30498-30499,


rs2713602 SNP
30398, 30404-
30452, 30455, 30458,
30502, 30505, 30508,



30405, 30408,
30460, 30550-30590
30510, 30591-30633



30411, 30511-30549


3:128486108_C_T
20632, 20635,
20679, 20682, 20686,
20727, 20731, 20735,


rs2335052 REF
20638, 20652,
20700, 20817, 20931,
20750, 20874, 20999,



20778, 20885,
20950, 20959, 21084,
21008, 21012, 21143,



20904, 20908,
21094, 21101, 30670-
21150, 21154, 30707-



21038, 21048,
30706
30745



30634-30669


3:128486108_C_T
30634-30636,
30670-30671, 30690,
30707, 30717,30728,


rs2335052 SNP
30652, 30656,
30694-30695, 30699,
30732, 30736, 30738,



30658, 30667,
30704, 30785-30825
30743, 30826-30868



30746-30784


3:128486676_G_A
30869-30914
30915-30962
30963-31012


rs4577488 REF


3:128486676_G_A
30875, 30882,
30918, 30922, 30931,
30966, 30970, 30982,


rs4577488 SNP
30884, 30894,
30934, 30944, 30950,
30990, 30999, 31005-



30896, 30902,
30956, 31052-31092
31006, 31093-31135



30908, 31013-31051


3:128487017_G_C
27511, 27536,
27582, 27586, 27588,
27620, 27625, 27627,


rs1573858 REF
27541, 27564,
27612, 27707, 27712,
27642, 27739-27740,



27575, 27666,
27903, 27905, 27907,
27942, 27944, 27959,



27668-27669,
28030, 28036, 28040,
28061, 28068, 28075,



27679-27680,
31153-31162
31163-31176



27759, 27858,



27861, 27896,



28000, 28002,



28014, 31136-31152


3:128487017_G_C
27666, 27668-
27707,27712, 31155,
27739-27740, 31171,


rs1573858 SNP
27670, 27679-
31203-31221
31222-31244



27680, 31140,



31144, 31177-31202


3′ UTR
796, 801-802, 813,
842, 847, 849, 853,
886, 891, 896, 898,


3:128480417-128480717
818, 820, 822, 935,
861, 866, 870, 986,
903, 911, 916, 1036-



940, 942, 944, 949,
988-989, 991, 996-997,
1038, 1040-1041, 1047,



965, 968, 31245-
1016, 31741-32218
1066, 32219-32705



31740


Intron 1
27511, 27536,
27582, 27586, 27588,
27620, 27625, 27627,


3:128487000-128487300
27541, 27564,
27612, 27903, 27905,
27642, 27942, 27944,



27575, 27666,
27907, 28017, 28030,
27959, 28042, 28048,



27668-27669,
28036, 28040, 28125,
28061, 28068, 28075,



27679, 27759,
28128, 28130, 28138,
28167, 28170, 28172,



27858, 27861,
28151, 28153, 28157,
28175, 28185, 28198,



27865, 27868,
31155-31162, 33186-
28204, 31163, 31166-



27880, 27896,
33617
31169, 31172-31176,



27981, 27986,

33618-34070



27990-27991,



28000, 28002,



28014, 28079,



28085, 28096,



28109, 28111,



28113, 28115,



31136-31138,



31140-31152,



32706-33185


Intron 2
34071-34634
34635-35192
35193-35750


3:128486500-128486800


Intron 2
30869-30914,
30915-30962, 34635-
30963-31012, 35193-


3:128486375-128486675
34071-34072,
34636, 34640, 34642-
35194, 35198, 35200-



34077, 34079-
34644, 34648-34657,
35202, 35207-35216,



34081, 34086-
34659, 34661-34666,
35218, 35220-35225,



34095, 34097,
34669-34674, 34677-
35228-35233, 35236-



34099-34104,
34679, 34684, 34686-
35238, 35243, 35245-



34107-34112,
34689, 34693, 34695-
35248, 35252, 35254-



34115-34117,
34696, 34702, 34705,
35255, 35261, 35264,



34122, 34124-
34708, 34711-34717,
35267, 35270-35276,



34127, 34131,
34720-34721, 34726-
35279-35280, 35285-



34133-34134,
34727, 34729-34731,
35286, 35288-35290,



34140, 34143,
34733, 34735-34740,
35292, 35294-35299,



34146, 34149-
34742, 34746, 34750-
35301, 35308-35309,



34155, 34158-
34751, 34754-34756,
35312, 35314-35315,



34159, 34164-
34758, 34761-34763,
35317, 35320-35322,



34165, 34167-
34765-34766, 34768-
35324-35325, 35327-



34169, 34171,
34769, 34771-34774,
35328, 35330-35333,



34173-34178,
34778-34779, 34782-
35337-35338, 35341-



34180, 34184,
34788, 34792, 34794-
35347, 35351, 35353-



34188-34189,
34796, 34798, 34801-
35355, 35357, 35360-



34192, 34194-
34803, 34805-34808,
35362, 35364-35367,



34195, 34197,
34811, 34813-34814,
35370, 35372-35373,



34200-34202,
34816-34817, 34819-
35375-35376, 35378-



34204-34205,
34821, 34823, 34825-
35380, 35382, 35384-



34207-34208,
34827, 34830, 34832,
35386, 35389, 35391,



34210-34213,
34834, 34838-34842,
35393, 35397-35401,



34217-34218,
34844-34845, 34848,
35403-35404, 35407,



34221-34227,
34850, 34854-34859,
35409, 35412-35417,



34231, 34233-
34861, 34863-34864,
35419, 35421-35422,



34235, 34237,
34866, 34868, 34871,
35424, 35426, 35429,



34240-34242,
34875, 34877, 34880,
35433, 35435, 35438,



34244-34247,
34882, 34884, 34886,
35440, 35442, 35444,



34250, 34252-
34888, 34891-34892,
35446, 35449-35450,



34253, 34255-
34894, 34897, 34899-
35452, 35455, 35457-



34256, 34258-
34906, 34910-34911,
35464, 35468-35469,



34260, 34262,
34915-34921, 34923-
35473-35479, 35481-



34264-34266,
34924, 34926-34928,
35482, 35484-35486,



34269, 34271,
34931, 34936, 34938,
35489, 35494, 35496,



34273, 34277-
34942-34944, 34952-
35500-35502, 35510-



34281, 34283-
34953, 34955, 34958,
35511, 35513, 35516,



34284, 34287,
34960-34961, 34963,
35518-35519, 35521,



34289, 34293-
34965, 34967, 34969-
35523, 35525, 35527-



34298, 34300,
34970, 34975-34977,
35528, 35533-35535,



34302-34303,
34979, 34981-34987,
35537, 35539-35545,



34305, 34307,
34989-34991, 34993,
35547-35549, 35551,



34310, 34314,
34998, 35001-35004,
35556, 35559-35562,



34316, 34319,
35007-35008, 35010-
35565-35566, 35568-



34321, 34323,
35011, 35013, 35017,
35569, 35571, 35575,



34326, 34328,
35021, 35025, 35028-
35579, 35583, 35586-



34331-34332,
35030, 35032-35035,
35588, 35590-35593,



34334, 34337,
35038-35041, 35044-
35596-35599, 35602-



34339-34346,
35046, 35048-35049,
35604, 35606-35607,



34350-34351,
35051, 35054, 35056-
35609, 35612, 35614-



34355-34361,
35057, 35059-35060,
35615, 35617-35618,



34363-34364,
35062, 35064-35065,
35620, 35622-35623,



34366-34368,
35067-35069, 35072-
35625-35627, 35630-



34371, 34376,
35075, 35077, 35079-
35633, 35635, 35637-



34378, 34382-
35083, 35086, 35090-
35641, 35644, 35648-



34384, 34392-
35091, 35093, 35095,
35649, 35651, 35653,



34393, 34395,
35097-35098, 35102,
35655-35656, 35660,



34398, 34400-
35104-35105, 35107,
35662-35663, 35665,



34401, 34403,
35109, 35112-35116,
35667, 35670-35674,



34405, 34407,
35118, 35121, 35124,
35676, 35679, 35682,



34409-34410,
35127, 35130, 35132,
35685, 35688, 35690,



34415-34417,
35135, 35138-35139,
35693, 35696-35697,



34419, 34421-
35141, 35144-35147,
35699, 35702-35705,



34427, 34429-
35149-35150, 35152,
35707-35708, 35710,



34431, 34433,
35155, 35157-35158,
35713, 35715-35716,



34438, 34441-
35160, 35162-35164,
35718, 35720-35722,



34444, 34447-
35166-35167, 35169,
35724-35725, 35729-



34448, 34450-
35171-35177, 35179,
35735, 35737, 35742,



34451, 34453,
35184, 35186-35187,
35744-35745, 35747,



34457, 34461,
35189, 35927-36080
36081-36240



34465, 34468-



34470, 34472-



34475, 34478-



34481, 34484-



34486, 34488-



34489, 34491,



34494, 34496-



34497, 34499-



34500, 34502,



34504-34505,



34507-34509,



34512-34515,



34517, 34519-



34523, 34525-



34526, 34530-



34531, 34533,



34536, 34538-



34539, 34543,



34545-34546,



34548, 34550,



34553-34558,



34560, 34563,



34566, 34569,



34572, 34574,



34577, 34580-



34581, 34583,



34586-34589,



34591-34592,



34594, 34597,



34599-34600,



34602, 34604-



34606, 34608-



34609, 34611,



34613-34619,



34621, 34626,



34628-34629,



34631, 35751-35926


Intron 3
30100-30145,
30146-30193, 30413-
30194-30243, 30461-


3:128485421-128485721
30367-30412,
30460, 36695-37130
30510, 37131-37564



36241-36694


Intron 3
37565-38118
38119-38662
38663-39208


3:128484028-128484328


Intron 4
11153, 29590,
11208, 29630, 29632-
11231, 29670, 29672-


3:128483533-128483833
29592-29601,
29641, 29643-29646,
29681, 29683-29685,



29603-29606,
29648-29669, 29879-
29687-29709, 29927-



29608-29629,
29926, 39659-40078
29976, 40079-40512



29833-29878,



39209-39658


Intron 4
40513-41026
41027-41508
41509-42002


3:128481949-128482249


Intron 5
7147, 7160, 7164,
7206, 7211, 7227,
7255-7256, 7277, 7279,


3:128481516-128481816
7181, 29062-29063,
7229, 29104, 29110,
29152, 29158, 29169,



29069, 29074,
29116, 29121, 29129,
29177, 29180, 29184,



29082, 29084,
29132, 29136, 29146,
29191, 29196, 43047-



29088, 29098,
42527-43046
43562



42003-42526


Intron 5
29056-29101,
29102-29149, 29369-
29150-29199, 29417-


3:128481320-128481620
29323-29368,
29416, 42527, 42538,
29466, 43057, 43063,



42003-42004,
42544, 42556-42559,
43075-43078, 43083,



42010, 42016,
42564, 42566, 42568,
43085, 43087, 43095-



42022, 42034-
42576-42577, 42591,
43096, 43110, 43113,



42037, 42042,
42594, 42603-42604,
43122-43123, 43125,



42044, 42046,
42606, 42615-42616,
43134-43135, 43150,



42054-42055,
42631, 42636-42638,
43155-43157, 43159,



42069, 42072,
42640, 42647-42648,
43166-43167, 43176,



42081-42082,
42657, 42659, 42666-
43178, 43185-43186,



42084, 42093-
42667, 42670, 42674-
43189, 43193-43194,



42094, 42109,
42675, 42681-42682,
43200-43201, 43214-



42114-42116,
42695-42697, 42701,
43216, 43220, 43222-



42118, 42125-
42703-42704, 42710,
43223, 43229, 43231-



42126, 42135,
42712-42713, 42715,
43232, 43234, 43236,



42137, 42144-
42717, 42724, 42727,
43243, 43246, 43249,



42145, 42148-
42730, 42736, 42738,
43255, 43257, 43264,



42149, 42153-
42745, 42749, 42754,
43268, 43273-43275,



42154, 42160-
42756-42757, 42760,
43278, 43280-43281,



42161, 42174-
42762-42763, 42765,
43283, 43287, 43289,



42176, 42180,
42769, 42771, 42774,
43292, 43296-43297,



42182-42183,
42778-42779, 42785,
43303, 43310, 43317,



42189, 42191-
42792, 42799, 42803,
43321, 43325, 43336,



42192, 42194,
42807, 42818, 42821,
43339, 43341-43342,



42196, 42203,
42823-42825, 42828,
43345, 43350-43351,



42206, 42209,
42833-42834, 42842,
43359, 43362, 43366-



42215, 42217,
42845, 42849-42850,
43367, 43369, 43376,



42224, 42228,
42852, 42859, 42866,
43383, 43386, 43392,



42233, 42235-
42869, 42875, 42877,
43394, 43400, 43402,



42236, 42239,
42883, 42885, 42887,
43404, 43408, 43412,



42241-42242,
42891, 42895, 42899,
43416, 43418-43419,



42244, 42248,
42901-42902, 42917,
43434, 43437, 43447,



42250, 42253,
42920, 42930, 42934,
43453, 43458-43459,



42257-42258,
42936, 42941-42942,
43461, 43465, 43475,



42264, 42271,
42944-42945, 42949,
43483, 43491-43493,



42278, 42282,
42959, 42967, 42975-
43499, 43504-43506,



42286, 42297,
42977, 42983, 42988-
43509, 43511-43512,



42300, 42302-
42990, 42993, 42995-
43517, 43521-43522,



42304, 42307,
42996, 43001, 43005-
43528, 43534, 43539-



42312-42313,
43006, 43012, 43018,
43540, 43542, 43551,



42321, 42324,
43023-43024, 43026,
43555, 43557, 43559-



42328-42329,
43035, 43039, 43041,
43560, 43562, 44091-



42331, 42338,
43043-43044, 43046,
44348



42345, 42348,
43831-44090



42354, 42356,



42362, 42364,



42366, 42370,



42374, 42378,



42380-42381,



42396, 42399,



42409, 42413,



42415, 42420-



42421, 42423-



42424, 42428,



42438, 42446,



42454, 42456-



42457, 42463,



42468-42470,



42473, 42475-



42476, 42481,



42485-42486,



42491-42492,



42498, 42503-



42504, 42506,



42515, 42519,



42521, 42523-



42524, 42526,



43563-43830









The SNP ID NOs indicated in Table 2 are based on NCBI's 2018 database of Single Nucleotide Polymorphisms (dbSNP). For variants/mutations with no available rs number, variant/mutation characteristics are indicated based on gnomAD v3 database and UCSC Genome Browser assembly ID: hg38, sequencing/assembly provider ID: Genome reference consortium Human GRCh38.p12(GCA_00001405.27) Assembly date December 2013 initial release December 2017 patch release 12. The introns, exons, and other transcript regions indicated in Table 2 correspond to the GATA2 transcript consequence NM_032638.


For the foregoing embodiments, each embodiment disclosed herein is contemplated as being applicable to each of the other disclosed embodiments. For example, it is understood that any of the RNA molecules or compositions of the present invention may be utilized in any of the methods of the present invention.


EXPERIMENTAL DETAILS
Example 1: GATA2 Correction Analysis

Guide sequences comprising 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348 are screened for high on target activity. On target activity is determined by DNA capillary electrophoresis analysis.


According to DNA capillary electrophoresis analysis, guide sequences comprising 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348 are tested for ability to correct the GATA2 gene.


Example 2: GATA2 Guide Activity Screen

To choose optimal guides for HDR strategies targeting three SNPs bounding Exon 5 and three mutations targeting Exon 6 or Intron 4 of GATA2 gene, 18 distinct DNA guides were screened for high on-target activity in HeLa cells (See Table 2 for guide sequences).


According to GATA2 genotyping, of HeLa cells were found to be homozygous for the reference form of the SNPs rs11717152 and rs11708606, therefore these regions were tested with guides targeting the reference sequence. Furthermore, HeLa cells are homozygous to the alternative form of SNPs rs2713603 and rs2713604, and were tested using guides targeting the alternative sequence (Table B). HeLa cells do not harbor the mutations p.R396Q, p.R398W or c.1017+572_C>T in GATA2, however, mutations are located in the PAM sequence of the listed guides, thus the activity of the guides can be tested in this cell model.









TABLE B





GATA2 genotyping in HeLa cells


















rs11717152
rs11708606
rs2713603
rs2713604


A > C (T > G)
G > A (C > T)
G > A (C > T)
T > C (A > G)


AA
GG
AA
CC









For assessing the activity of the guides, an OMNI-50 nuclease coding plasmid (64 ng) was co-transfected with each of the guide DNA plasmids (20 ng) in 96 well plate format using jetOPTIMUS reagent (Polyplus). Cells were harvested 72 h post transfection. Genomic DNA was extracted and the genomic regions flanking the SNPs and the mutations were amplified by PCR and the level of editing was measured by capillary electrophoresis. The graph in FIG. 2 represents the average of % editing±STDV of three independent experiments. The results indicate that the activity of the guides is variable.


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

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

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

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

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

  • 12. 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.

  • 13. 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.

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

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

  • 16. 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.

  • 17. 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.

  • 18. 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.

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

  • 20. 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.

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

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

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

  • 24. 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.

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

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

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

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

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

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

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

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

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

  • 34. 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.

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

  • 36. Yu et al. (2016) “Gene editing of human hematopoietic stem and progenitor cells: Promise and potential hurdles”, Hum Gene Ther. 27(10):729-740.

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

  • 38. 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 GATA2 gene having a mutation associated with MonoMAC syndrome, the method comprising introducing to the cell a composition comprising: a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; andan RNA molecule comprising a guide sequence portion having 17-25 nucleotides or a nucleotide sequence encoding the same,
  • 2. The method of claim 1, wherein the RNA molecule targets the CRISPR nuclease to the mutation associated with MonoMAC syndrome, wherein the mutation associated with MonoMAC syndrome is selected from the group consisting of 3:128479475_T_C; 3:128479476_A_G; 3:128479846_C_A; 3:128479875_C_T; 3:128480135_T_A; 3:128480322_G_A; 3:128480449_G_A; 3:128480599_C_T; 3:128480829_G_A; 3:128480845_C_T; 3:128480918_C_T; 3:128480935_T_C; 3:128480946_G_A; 3:128481039_T_A; 3:128481047_G_A; 3:128481055_G_T; 3:128481060_C_T; 3:128481071_C_A; 3:128481077_G_T; 3:128481090_G_A; 3:128481092_G_A; 3:128481095_G_A; 3:128481102_G_A; 3:128481115_G_T; 3:128481131_G_A; 3:128481136_G_A; 3:128481142_C_T; 3:128481147_G_A; 3:128481150_C_A; 3:128481162_C_G; 3:128481176_C_G; 3:128481180_A_T; 3:128481188_G_A; 3:128481189_A_G; 3:128481199_C_T; 3:128481214_C_G; 3:128481219_C_T; 3:128481220_G_C; 3:128481224_C_T; 3:128481226_C_A; 3:128481230_G_A; 3:128481230_G_T; 3:128481232_C_A; 3:128481245_T_A; 3:128481262_C_T; 3:128481270_G_A; 3:128481275_C_T; 3:128481276_G_A; 3:128481283_G_C; 3:128481299_A_G; 3:128481302_G_T; 3:128481315_T_AACC_T; 3:128481318_C_A; 3:128481819_A_G; 3:128481836_A_AGAGG; 3:128481841_C_T; 3:128481845_A_G; 3:128481849_G_T; 3:128481866_CGTTGGCGTTTCG_C; 3:128481872_C_T; 3:128481877_C_T; 3:128481878_G_A; 3:128481880_C_A; 3:128481881_G_A; 3:128481881_G_C; 3:128481887_A_C; 3:128481897_TGTCG_AAGGC; 3:128481898_G_T; 3:128481901_G_A; 3:128481908_A_G; 3:128481923_T_TGCCGGCTCTTCTGGCG; 3:128481924_G_A; 3:128481926_C_T; 3:128481927_G_A; 3:128481937_G_A; 3:128481937_G_GCGGC; 3:128481938_C_A; 3:128481938_C_T; 3:128481939_GGCCG_AAGGC; 3:128481942_C_T; 3:128481945_C_A; 3:128483288_G_A; 3:128483333_C_T; 3:128483347_C_T; 3:128483375_T_A; 3:128483868_G_A; 3:128483889_G_A; 3:128483905_CT_C; 3:128483906_T_C; 3:128483925_C_CATTGCACAGGT; 3:128483957_C_T; 3:128483973_C_CTGTGGCCCCACAGT; 3:128484010_A_C; 3:128485738_C_T; 3:128485741_G_A; 3:128485746_G_A; 3:128485750_C_T; 3:128485758_AG_A; 3:128485766_A_T; 3:128485769_T_C; 3:128485779_T_TC; 3:128485780_C_A; 3:128485783_C_T; 3:128485789_A_C; 3:128485795_C_A; 3:128485798_G_A; 3:128485811_C_T; 3:128485819 T_TAGTCGTGGGC; 3:128485837_G_A; 3:128485837_G_C; 3:128485843_T_C; 3:128485850_G_A; 3:128485855_G_A; 3:128485869_G_C; 3:128485871_G_A; 3:128485874_T_C; 3:128485891_A_G; 3:128485892_T_C; 3:128485892_T_G; 3:128485909_C_T; 3:128485910_G_A; 3:128485916_G_T; 3:128485924_C_T; 3:128485929_C_T; 3:128485937_T_G; 3:128485944_C_CGTCAG; 3:128485944_C_T; 3:128485962_C_A; 3:128485966_A_ACGC; 3:128485967_C_G; 3:128485970_C_T; 3:128485973_C_T; 3:128485982_C_G; 3:128485998_A_AC; 3:128485999_C_T; 3:128486002_C_A; 3:128486004_C_CGCGGAAGA; 3:128486005_G_A; 3:128486006_C_T; 3:128486023_G_A; 3:128486030_C_T; 3:128486031_C_A; 3:128486038_G_C; 3:128486057_C_T; 3:128486059_T_G; 3:128486066_G_A; 3:128486072_T_G; 3:128486075_G_A; 3:128486103_G_T; 3:128486104_T_A; 3:128486117_G_A; 3:128486129_C_A; 3:128486141_C_T; 3:128486142_G_T; 3:128486143_C_T; 3:128486153_C_T; 3:128486173_G_A; 3:128486179_A_G; 3:128486185_A_G; 3:128486189_G_A; 3:128486201_C_T; 3:128486218_T_C; 3:128486227_G_T; 3:128486240_G_A; 3:128486252_A_AG; 3:128486260_T_C; 3:128486271_C_T; 3:128486284_A_AGG; 3:128486287_G_A; 3:128486288_C_A; 3:128486295_GC_G; 3:128486296_C_T; 3:128486298_CC_AA; 3:128486320_G_T; 3:128486355_T_GC; 3:128486356_C_G; 3:128486365_C_CG; 3:128486805_T_C; 3:128486808_G_C; 3:128486826_C_T; 3:128486828_C_A; 3:128486850_G_A; 3:128486856_T_C; 3:128486872_A_C; 3:128486883_T_C; 3:128486890_A_T; 3:128486893_C_T; 3:128486896_C_T; 3:128486911_G_C; 3:128486931_A_G; 3:128486948_G_A; 3:128486956_G_T; 3:128486961_G_GACA; 3:128486967_G_C; 3:128486970_T_A; 3:128486973_T_G; 3:128486982_A_T; 3:128486992_G_A; 3:128487002_C_A; 3:128487004_A_G; 3:128487009_G_T; 3:128487016_CG_GC; 3:128487016_C_T; 3:128487021_G_T; 3:128487036_G_A; 3:128487073_G_C; 3:128492886_G_A; 3:128492959_A_C; 3:128493033_G_T; 3:128493046_G_A; 3:128493068_G_A,wherein the guide sequence portion of the RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348 which targets a mutation associated with MonoMAC syndrome.
  • 3-4. (canceled)
  • 5. The method of claim 1, wherein the RNA molecule targets the CRISPR nuclease to a SNP position of the mutant allele, wherein the SNP position is located at any one of rs10934857; rs2713604; rs2713603; rs11708606; rs11717152; rs2659689; rs2713602; rs2335052; rs4577488; or rs1573858, andwherein the guide sequence portion of the RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348 which targets a SNP position of the mutant allele.
  • 6. The method of claim 5, wherein the SNP position is in a non-coding region of the GATA2 gene at a location that is 250 or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome.
  • 7-8. (canceled)
  • 9. The method of claim 5, wherein the SNP position contains a heterozygous SNP.
  • 10. The method of claim 1, wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in a non-coding region of the GATA2 gene at a location that is 250 or fewer nucleotides from the first or the last nucleotide of a coding region of the GATA2 gene containing the mutation associated with MonoMAC syndrome, wherein the coding region is any one of Exon 2, Exon 3, Exon 4, Exon 5, or Exon 6 of the GATA2 gene, andwherein the non-coding region is any one of the 3′UTR, Intron 1, Intron 2, Intron 3, Intron 4, or Intron 5 of the GATA2 gene.
  • 11-12. (canceled)
  • 13. The method of claim 10, wherein the non-coding region is any one of the 3:128480417-128480717; 3:128487000-128487300; 3:128486500-128486800; 3:128486375-128486675; 3:128485421-128485721; 3:128484028-128484328; 3:128483533-128483833; 3:128481949-128482249; 3:128481516-128481816; or 3:128481320-128481620.
  • 14. The method of claim 13, wherein the guide sequence portion of the RNA molecule comprises 17-25 contiguous nucleotides containing nucleotides in the sequence set forth in any one of SEQ ID NOs: 1-44348 which targets a non-coding region of the GATA2 gene.
  • 15. The method of claim 1, further comprising introducing to the cell a donor template for homology directed repair (HDR), alteration, or replacement of a sequence of the GATA2 allele, wherein the modifying results in correction of the mutant allele of the GATA2 gene from the mutant phenotype to a non-mutant phenotype.
  • 16. (canceled)
  • 17. The method of claim 1, comprising first obtaining a cell with a GATA2 gene mutation associated with MonoMAC syndrome from a subject with a GATA2 gene mutation related to MonoMAC syndrome and/or suffering from MonoMAC syndrome.
  • 18. (canceled)
  • 19. A composition comprising a modified cell obtained by the method of claim 1.
  • 20. The composition of claim 19, further comprising a pharmaceutically acceptable carrier.
  • 21. (canceled)
  • 22. An in vitro or ex vivo method of preparing a composition comprising a modified cell, the method comprising: a) isolating HSPCs from cells obtained from a subject with a GATA2 gene mutation related to MonoMAC syndrome and/or suffering from MonoMAC syndrome, and obtaining the cell from the subject;b) introducing to the cells of step (a) a composition comprising: a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; andan RNA molecule comprising a guide sequence portion having 17-25 nucleotides, wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in the mutant allele of the GATA2 gene according to the method of claim 1, optionally, introducing to the cells a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele so as to modify the mutant allele of the GATA2 gene in one or more cells thereby obtaining modified cells; optionally further comprising the step ofc) culture expanding the modified cells of step (b), wherein the modified cells are capable of engraftment and giving rise to progeny cells after engraftment.
  • 23. (canceled)
  • 24. A method of treating a subject afflicted with MonoMAC syndrome, comprising administration of a therapeutically effective amount of the modified cells of the composition of claim 19.
  • 25. A method for treating MonoMAC syndrome in a subject with a GATA2 gene mutation relating to MonoMAC syndrome in need thereof, the method comprising: a) isolating HSPCs from cells obtained from the subject;b) introducing to the cells of step (a) a composition comprising: a CRISPR nuclease or a nucleotide sequence encoding the CRISPR nuclease; andan RNA molecule comprising a guide sequence portion having 17-25 nucleotides, wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in the mutant allele of the GATA2 gene according to the method of claim 1, optionally, introducing to the cells a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele so as to modify the mutant allele of the GATA2 gene in one or more cells thereby obtaining modified cells; optionally further comprising an additional step ofc) culture expanding the cells of step (b) wherein the modified cells are capable of engraftment and giving rise to progeny cells after engraftment; andd) administering to the subject the cells of step (b) or step (c)thereby treating the MonoMAC syndrome in the subject.
  • 26. A method for treating MonoMAC syndrome in a subject with a GATA2 gene mutation relating to MonoMAC syndrome in need thereof, the method comprising administering to the subject autologous modified cells or progeny of autologous modified cells, wherein the autologous modified cells are modified so as to have a correction of the mutant allele of the GATA2 gene from the mutant phenotype to a non-mutant phenotype, wherein said double strand break results from introduction to the cells of a composition comprising a CRISPR nuclease or sequence encoding the CRISPR nuclease and an RNA molecule wherein a complex of the CRISPR nuclease and the RNA molecule affects a double strand break in the mutant allele of the GATA2 gene according to the method of claim 1,thereby treating the MonoMAC syndrome in the subject.
  • 27. A composition comprising an RNA molecule, the 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: 1-44348.
  • 28. The composition of claim 27, further comprising a donor template for homology directed repair (HDR), alteration, or replacement of a target sequence of the GATA2 allele and one or more CRISPR nucleases or polynucleotide molecules encoding the one or more CRISPR nucleases.
  • 29. (canceled)
  • 30. A method for modifying in a cell a mutant allele of the GATA2 gene, the method comprising delivering to the cell the composition of claim 27.
  • 31. A method for treating MonoMAC syndrome, the method comprising delivering to a subject having MonoMAC syndrome the composition of claim 27.
  • 32-34. (canceled)
Parent Case Info

This application claims the benefit of U.S. Provisional Application Nos. 62/863,129, filed Jun. 18, 2019 and 62/834,230, filed Apr. 15, 2019, the contents of which are hereby incorporated by reference.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2020/028161 4/14/2020 WO
Provisional Applications (2)
Number Date Country
62863129 Jun 2019 US
62834230 Apr 2019 US