WO2018002886A1 - Materials and methods for treatment of spinocerebellar ataxia 3 (sca3) and other related disorders - Google Patents

Materials and methods for treatment of spinocerebellar ataxia 3 (sca3) and other related disorders Download PDF

Info

Publication number
WO2018002886A1
WO2018002886A1 PCT/IB2017/053941 IB2017053941W WO2018002886A1 WO 2018002886 A1 WO2018002886 A1 WO 2018002886A1 IB 2017053941 W IB2017053941 W IB 2017053941W WO 2018002886 A1 WO2018002886 A1 WO 2018002886A1
Authority
WO
WIPO (PCT)
Prior art keywords
atxn3
gene
cell
sequence
dna
Prior art date
Application number
PCT/IB2017/053941
Other languages
French (fr)
Inventor
Ante Sven LUNDBERG
Samarth Kulkarni
Lawrence Klein
Hari Kumar PADMANABHAN
Original Assignee
Crispr Therapeutics Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Crispr Therapeutics Ag filed Critical Crispr Therapeutics Ag
Publication of WO2018002886A1 publication Critical patent/WO2018002886A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications

Definitions

  • the present disclosure relates to the field of gene editing and specifically to the alteration of the Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene.
  • ATXN3 Spinocerebellar Ataxia Type 3 Protein
  • This application contains a Sequence Listing in computer readable form (filename: 170137PCT sequence listing (ATXN3) _(Part 1): 19,889,222 bytes - ASCII text file; created June 26, 2017 and 170137PCT sequence listing (ATXN3) _(Part 2): 7,142,691 bytes - ASCII text file; created June 26, 2017), which is incorporated herein by reference in its entirety and forms part of the disclosure.
  • Genome engineering refers to the strategies and techniques for the targeted, specific modification of the genetic information (genome) of living organisms. Genome engineering is a very active field of research because of the wide range of possible applications, particularly in the areas of human health. For example, genome engineering can be used to alter (e.g., correct or knock-out) a gene carrying a harmful mutation, or to explore the function of a gene. Early technologies developed to insert a transgene into a living cell were often limited by the random nature of the insertion of the new sequence into the genome. Random insertions into the genome may result in disrupting normal regulation of neighboring genes leading to severe unwanted effects. Furthermore, random integration technologies offer little reproducibility, as there is no guarantee that the sequence would be inserted at the same place in two different cells.
  • ZFNs zinc finger nucleases
  • TALENs transcription activator like effector nucleases
  • HEs homing endonucleases
  • MegaTALs enable a specific area of the DNA to be modified, thereby increasing the precision of the alteration compared to early technologies.
  • the resulting therapy may completely remedy certain ATXN3 related indications and/or diseases.
  • cellular, ex vivo and in vivo methods for creating permanent changes to the genome by deleting and/or correcting the trinucleotide repeat expansion or replacing one or more nucleotide bases, or one or more exons and/or introns within or near the Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene, or otherwise introducing insertions, deletions or mutations of at least one nucleotide within or near the ATXN3 gene or other DNA sequences that encode regulatory elements of the ATXN3 gene by genome editing.
  • ATXN3 Spinocerebellar Ataxia Type 3 Protein
  • Such methods can reduce or eliminate the expression or function of aberrant ATXN3 gene products and/or restore the wild-type ATXN3 protein activity, which can be used to treat an ATXN3 related condition or disorder such as Spinocerebellar ataxia 3 (SCA3).
  • SCA3 Spinocerebellar ataxia 3
  • components and compositions, and vectors for performing such methods are provided herein.
  • a method for editing a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell by genome editing comprising: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
  • DNA deoxyribonucleic acid
  • SSBs single-strand breaks
  • DSBs double-strand breaks
  • a method for editing a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell by genome editing comprising: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
  • DNA deoxyribonucleic acid
  • SSBs single-strand breaks
  • DSBs double-strand breaks
  • an ex vivo method for treating a patient having an ATXN3 related condition or disorder comprising: creating a patient specific induced pluripotent stem cell (iPSC); editing within or near a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene or other DNA sequences that encode regulatory elements of the ATXN3 gene of the iPSC; differentiating the genome-edited iPSC into a neuron or glial cell of the Central Nervous System (CNS); and implanting the neuron or glial cell of the Central Nervous System (CNS) into the patient.
  • iPSC patient specific induced pluripotent stem cell
  • ATXN3 Spinocerebellar Ataxia Type 3 Protein
  • the editing step comprises: introducing into the iPSC one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
  • DNA deoxyribonucleic acid
  • the editing step comprises: introducing into the iPSC one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
  • DNA deoxyribonucleic acid
  • an ex vivo method for treating a patient having an ATXN3 related condition or disorder comprising: isolating a mesenchymal stem cell from the patient; editing within or near a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene or other DNA sequences that encode regulatory elements of the ATXN3 gene of the mesenchymal stem cell; differentiating the genome-edited mesenchymal stem cell into a neuron or glial cell of the Central Nervous System (CNS); and implanting the neuron or glial cell of the Central Nervous System (CNS) into the patient.
  • ATXN3 Spinocerebellar Ataxia Type 3 Protein
  • the editing step comprises: introducing into the mesenchymal stem cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
  • DNA deoxyribonucleic acid
  • the editing step comprises: introducing into the mesenchymal stem cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
  • DNA deoxyribonucleic acid
  • an in vivo method for treating a patient with an ATXN3 related disorder comprising: editing the Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell of the patient.
  • ATXN3 Spinocerebellar Ataxia Type 3 Protein
  • the editing step comprises: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
  • DNA deoxyribonucleic acid
  • the editing step comprises: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
  • DNA deoxyribonucleic acid
  • the cell is a cell of the Central Nervous System (CNS).
  • the cell of the Central Nervous System (CNS) is a neuron.
  • the cell of the Central Nervous System (CNS) is a glial cell.
  • DNA endonucleic acid endonuclease is delivered to the cell of the Central Nervous System (CNS) via intraparenchymal, intravenous, intra-arterial, intracerebroventricular, intracisternal, intrathecal, intracranial or intraperitoneal administration routes.
  • CNS Central Nervous System
  • Also provided herein is a method of altering the contiguous genomic sequence of an ATXN3 gene in a cell comprising: contacting the cell with one or more deoxyribonucleic acid (DNA) endonuclease to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs).
  • DNA deoxyribonucleic acid
  • SSBs single-strand breaks
  • DSBs double-strand breaks
  • the alteration of the contiguous genomic sequence occurs in one or more exons of the ATXN3 gene.
  • the alteration of the contiguous genomic sequence occurs in exon 10 of the ATXN3 gene.
  • the one or more deoxyribonucleic acid (DNA) endonuclease is selected from any of those sequences in SEQ ID NOs: 1-620 and variants having at least 90% homology to any of the sequences listed in SEQ ID NOs: 1-620.
  • the one or more deoxyribonucleic acid (DNA) endonuclease is one or more protein or polypeptide. In some aspects, the one or more deoxyribonucleic acid (DNA) endonuclease is one or more polynucleotide encoding the one or more DNA endonuclease. In some aspects, the one or more deoxyribonucleic acid (DNA) endonuclease is one or more ribonucleic acid (RNA) encoding the one or more DNA endonuclease. In some aspects, the one or more ribonucleic acid (RNA) is one or more chemically modified RNA. In some aspects, the one or more ribonucleic acid (RNA) is chemically modified in the coding region. In some aspects, the one or more polynucleotide or one or more ribonucleic acid (RNA) is codon optimized.
  • the methods further comprise: introducing one or more gRNA or one or more sgRNA.
  • the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a sequence within or near the expanded trinucleotide repeat in the ATXN3 gene.
  • the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a DNA sequence within or near the ATXN3 gene.
  • the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a sequence flanking the ATXN3 gene or other sequence that encodes a regulatory element of the ATXN3 gene.
  • the one or more gRNA or one or more sgRNA is chemically modified.
  • the one or more gRNA or one or more sgRNA is pre-complexed with the one or more deoxyribonucleic acid (DNA) endonuclease.
  • the pre- complexing involves a covalent attachment of the one or more gRNA or one or more sgRNA to the one or more deoxyribonucleic acid (DNA) endonuclease.
  • the one or more deoxyribonucleic acid (DNA) endonuclease is formulated in a liposome or lipid nanoparticle. In some aspects, the one or more
  • deoxyribonucleic acid (DNA) endonuclease is formulated in a liposome or a lipid nanoparticle which also comprises the one or more gRNA or one or more sgRNA.
  • the one or more deoxyribonucleic acid (DNA) endonuclease is encoded in an AAV vector particle.
  • the one or more gRNA or one or more sgRNA is encoded in an AAV vector particle.
  • the one or more gRNA or one or more sgRNA is encoded in an AAV vector particle.
  • deoxyribonucleic acid (DNA) endonuclease is encoded in an AAV vector particle which also encodes the one or more gRNA or one or more sgRNA.
  • the AAV vector particle is selected from those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
  • the methods further comprise: introducing into the cell a donor template comprising at least a portion of the wild-type ATXN3 gene.
  • at least a portion of the wild-type ATXN3 gene comprises one or more sequences selected from an ATXN3 exon, an ATXN3 intron, and a sequence comprising an exon:intr on junction of ATXN3.
  • the donor template comprises homologous arms to the genomic locus of the ATXN3 gene.
  • the donor template is either a single or double stranded polynucleotide.
  • the donor template is encoded in an AAV vector particle, where the AAV vector serotype is selected from any of those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
  • deoxyribonucleic acid (DNA) endonuclease is formulated into a lipid nanoparticle, and the one or more gRNA or one or more sgRNA is delivered to the cell ex vivo by electroporation and the donor template is delivered to the cell by an adeno-associated virus (AAV) vector.
  • AAV adeno-associated virus
  • the one or more polynucleotide encoding one or more deoxyribonucleic acid (DNA) endonuclease is formulated into a liposome or lipid nanoparticle which also comprises the one or more gRNA or one or more sgRNA and the donor template.
  • a single-molecule guide RNA comprising: at least a spacer sequence that is an RNA sequence corresponding to any of SEQ ID NOs: 5305-56902.
  • the single-molecule guide RNA further comprises a spacer extension region.
  • the single-molecule guide RNA further comprises a tracrRNA extension region.
  • the single-molecule guide RNA is chemically modified.
  • the single-molecule gudie RNA is pre-complexed with a DNA endonuclease.
  • the DNA endonuclease is a Cas9 or CPfl endonuclease.
  • the Cas9 or Cpfl endonuclease is selected from the group consisting of: S.
  • thermophilus CRISPR 3 Cas9 T. denticola Cas9, L. bacterium ND2006 Cpfl and
  • the Cas9 or Cpfl endonuclease comprises one or more nuclear localization signals (NLSs).
  • NLSs nuclear localization signals
  • at least one NLS is at or within 50 amino acids of the amino-terminus of the Cas9 or Cpfl endonuclease and/or at least one NLS is at or within 50 amino acids of the carboxy-terminus of the Cas9 or Cpfl endonuclease.
  • a non-naturally occurring CRISPR/Cas system comprising a polynucleotide encoding a Cas9 or Cpfl enzyme and at least one single-molecule guide RNA described herein.
  • the polynucleotide encoding a Cas9 or Cpfl endonuclease is selected from a group consisting of: S. pyogenes Cas9, S. aureus Cas9, N. meningitides Cas9, S. thermophilus CRISPR1 Cas9, S. thermophilus CRISPR 3 Cas9, T. denticola Cas9, L. bacterium ND2006 Cpfl and Acidaminococcus sp.
  • the polynucleotide encoding a Cas9 or Cpfl enzyme comprises one or more nuclear localization signals (NLSs).
  • NLSs nuclear localization signals
  • at least one NLS is at or within 50 amino acids of the amino-terminus of the polynucleotide encoding a Cas9 or Cpfl enzyme and/or at least one NLS is at or within 50 amino acids of the carboxy-terminus of the polynucleotide encoding a Cas9 or Cpfl endonuclease.
  • polynucleotide encoding a Cas9 or Cpfl enzyme is codon optimized for expression in a eukaryotic cell.
  • RNA encoding the single-molecule guide RNA described herein is also provided herein.
  • RNA encoding the CRISPR/Cas system described herein is also provided herein.
  • Also provided herein is a DNA encoding the single-molecule guide RNA described herein.
  • a vector comprising a DNA encoding the single-molecule guide RNA and CRISPR/Cas system.
  • the vector is a plasmid.
  • the vector is an AAV vector particle, wherein the AAV vector particle is selected from those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
  • Figures 1 A-B depict the type II CRISPR/Cas system
  • Figure 1 A is a depiction of the type II CRISPR/Cas system including gRNA
  • Figure IB is another depiction of the type II CRISPR/Cas system including sgRNA
  • Figures 2A-D describe the cutting efficiencies of S.pyogenes gRNAs selected via an in-vitro transcribed (IVT) gRNA screen in HEK293T cells; [00042] Figure 2A describes the cutting efficiencies in the range of 12.5 - 36.8% of S.
  • pyogenes gRNAs selected via an in-vitro transcribed (IVT) gRNA screen in HEK293T cells;
  • Figure 2B describes the cutting efficiencies in the range of 8.0 - 12.5% of S. pyogenes gRNAs selected via an in-vitro transcribed (IVT) gRNA screen in HEK293T cells;
  • Figure 2C describes the cutting efficiencies in the range of 3.0 - 7.7% of S. pyogenes gRNAs selected via an in-vitro transcribed (IVT) gRNA screen in HEK293T cells;
  • Figure 2D describes the cutting efficiencies in the range of 0.3 - 2.5% of S. pyogenes gRNAs selected via an in-vitro transcribed (IVT) gRNA screen in HEK293T cells;
  • Figures 3A-B describe the cutting efficiency of S. pyogenes gRNAs in HEK293T cells
  • Figure 3 A describes the cutting efficiency in the range of 8.3 - 36.8% of S. pyogenes gRNAs in HEK293T cells.
  • Figure 3B describes the cutting efficiency in the range of 0.3 - 8.2% of S. pyogenes gRNAs in HEK293T cells.
  • SEQ ID Nos: 1-620 are Cas endonuclease ortholog sequences.
  • SEQ ID NOs: 621-631 do not include sequences.
  • SEQ ID NOs: 632-4715 are microRNA sequences.
  • SEQ ID NOs : 4716-4733 do not include sequences.
  • SEQ ID Nos: 4734-5302 are AAV serotype sequences.
  • SEQ ID NO: 5303 is a ATXN3 nucleotide sequence.
  • SEQ ID NO: 5304 is a gene sequence including 1-5 kilobase pairs upstream and/or downstream of the ATXN3 gene.
  • SEQ ID NOs: 5305 - 5633 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with a T. denticola Cas9 endonuclease.
  • SEQ ID NOs: 5634 - 6353 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with a S. thermophilus Cas9 endonuclease.
  • SEQ ID NOs: 6354 -8658 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with a S. aureus Cas9 endonuclease.
  • SEQ ID NOs: 8659 - 10697 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with a N. meningitides Cas9 endonuclease.
  • SEQ ID NOs: 10698 - 30885 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with a S. pyogenes Cas9 endonuclease.
  • SEQ ID NOs: 30886 - 56902 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with an Acidaminococcus, a Lachnospiraceae, and a Franciscella Novicida Cpf 1 endonuclease.
  • SEQ ID Nos: 56903-56932 do not include sequences.
  • SEQ ID NO: 56933 is a sample guide RNA (gRNA) for a S. pyogenes Cas9 endonuclease.
  • SEQ ID NOs: 56934-56936 show sample sgRNA sequences. Detailed Description
  • alteration refers to any change in the genome of a cell. In the context of treating genetic disorders, alterations may include, but are not limited to, insertion, deletion and correction.
  • insertion refers to an addition of one or more nucleotides in a DNA sequence. Insertions can range from small insertions of a few nucleotides to insertions of large segments such as a cDNA or a gene.
  • deletion refers to a loss or removal of one or more nucleotides in a DNA sequence or a loss or removal of the function of a gene. In some cases, a deletion can include, for example, a loss of a few
  • deletion of a gene refers to the elimination or reduction of the function or expression of a gene or its gene product. This can result from not only a deletion of sequences within or near the gene, but also other events (e.g., insertion, nonsense mutation) that disrupt the expression of the gene.
  • correction refers to a change of one or more nucleotides of a genome in a cell, whether by insertion, deletion or substitution. Such correction may result in a more favorable genotypic or phenotypic outcome, whether in structure or function, to the genomic site which was corrected.
  • a "correction" includes the correction of a mutant or defective sequence to a wild-type sequence which restores structure or function to a gene or its gene product(s). Depending on the nature of the mutation, correction may be achieved via various strategies disclosed herein. In one non-limiting example, a missense mutation may be corrected by replacing the region containing the mutation with its wild-type counterpart. As another example, duplication mutations (e.g., repeat expansions) in a gene may be corrected by removing the extra sequences.
  • alterations may also include a gene knock-in, knock-out or knockdown.
  • knock-in refers to an addition of a DNA sequence, or fragment thereof into a genome.
  • DNA sequences to be knocked-in may include an entire gene or genes, may include regulatory sequences associated with a gene or any portion or fragment of the foregoing.
  • a cDNA encoding the wild-type protein may be inserted into the genome of a cell carrying a mutant gene. Knock-in strategies need not replace the defective gene, in whole or in part.
  • a knock-in strategy may further involve substitution of an existing sequence with the provided sequence, e.g., substitution of a mutant allele with a wild- type copy.
  • the term “knock-out” refers to the elimination of a gene or the expression of a gene.
  • a gene can be knocked out by either a deletion or an addition of a nucleotide sequence that leads to a disruption of the reading frame.
  • a gene may be knocked out by replacing a part of the gene with an irrelevant sequence.
  • knock-down refers to reduction in the expression of a gene or its gene product(s). As a result of a gene knock-down, the protein activity or function may be attenuated or the protein levels may be reduced or eliminated.
  • Genome editing generally refers to the process of modifying the nucleotide sequence of a genome, preferably in a precise or pre-determined manner.
  • methods of genome editing described herein include methods of using site-directed nucleases to cut deoxyribonucleic acid (DNA) at precise target locations in the genome, thereby creating single-strand or double- strand DNA breaks at particular locations within the genome.
  • breaks can be and regularly are repaired by natural, endogenous cellular processes, such as homology-directed repair (HDR) and non-homologous end joining (NHEJ), as reviewed in Cox et al., Nature Medicine 21(2), 121-31 (2015).
  • HDR homology-directed repair
  • NHEJ non-homologous end joining
  • HDR directly joins the DNA ends resulting from a double-strand break, sometimes with the loss or addition of nucleotide sequence, which may disrupt or enhance gene expression.
  • HDR utilizes a homologous sequence, or donor sequence, as a template for inserting a defined DNA sequence at the break point.
  • the homologous sequence can be in the endogenous genome, such as a sister chromatid.
  • the donor can be an exogenous nucleic acid, such as a plasmid, a single-strand oligonucleotide, a double-stranded oligonucleotide, a duplex
  • a third repair mechanism can be microhomology- mediated end joining (MMEJ), also referred to as "Alternative NHEJ,” in which the genetic outcome is similar to NHEJ in that small deletions and insertions can occur at the cleavage site.
  • MMEJ microhomology- mediated end joining
  • MMEJ can make use of homologous sequences of a few base pairs flanking the DNA break site to drive a more favored DNA end joining repair outcome, and recent reports have further elucidated the molecular mechanism of this process; see, e.g., Cho and Greenberg, Nature 518, 174-76 (2015); Kent et a ⁇ ., Nature Structural and Molecular Biology, Adv. Online
  • a step in the genome editing process can be to create one or two DNA breaks, the latter as double-strand breaks or as two single-stranded breaks, in the target locus as near the site of intended mutation. This can be achieved via the use of site-directed polypeptides, as described and illustrated herein.
  • a CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) genomic locus can be found in the genomes of many prokaryotes (e.g., bacteria and archaea). In prokaryotes, the CRISPR locus encodes products that function as a type of immune system to help defend the prokaryotes against foreign invaders, such as virus and phage. There are three stages of CRISPR locus function: integration of new sequences into the CRISPR locus, expression of CRISPR RNA (crRNA), and silencing of foreign invader nucleic acid. Five types of CRISPR systems (e.g., Type I, Type II, Type III, Type U, and Type V) have been identified.
  • a CRISPR locus includes a number of short repeating sequences referred to as "repeats.” When expressed, the repeats can form secondary structures (e.g., hairpins) and/or comprise unstructured single-stranded sequences.
  • the repeats usually occur in clusters and frequently diverge between species.
  • the repeats are regularly interspaced with unique intervening sequences referred to as "spacers," resulting in a repeat-spacer-repeat locus architecture.
  • the spacers are identical to or have high homology with known foreign invader sequences.
  • a spacer-repeat unit encodes a crisprRNA (crRNA), which is processed into a mature form of the spacer-repeat unit.
  • crRNA crisprRNA
  • a crRNA comprises a "seed” or spacer sequence that is involved in targeting a target nucleic acid (in the naturally occurring form in prokaryotes, the spacer sequence targets the foreign invader nucleic acid).
  • a spacer sequence is located at the 5' or 3' end of the crRNA.
  • a CRISPR locus also comprises polynucleotide sequences encoding CRISPR
  • Cas genes encode endonucleases involved in the biogenesis and the interference stages of crRNA function in prokaryotes. Some Cas genes comprise homologous secondary and/or tertiary structures.
  • crRNA biogenesis in a Type II CRISPR system in nature requires a trans-activating CRISPR RNA (tracrRNA).
  • tracrRNA trans-activating CRISPR RNA
  • Figures 1A and IB Non-limiting examples of Type II CRISPR systems are shown in Figures 1A and IB.
  • the tracrRNA can be modified by endogenous RNaselll, and then hybridizes to a crRNA repeat in the pre-crRNA array. Endogenous RNaselll can be recruited to cleave the pre-crRNA. Cleaved crRNAs can be subjected to exoribonuclease trimming to produce the mature crRNA form ⁇ e.g., 5' trimming).
  • the tracrRNA can remain hybridized to the crRNA, and the tracrRNA and the crRNA associate with a site-directed polypeptide ⁇ e.g., Cas9).
  • the crRNA of the crRNA-tracrRNA-Cas9 complex can guide the complex to a target nucleic acid to which the crRNA can hybridize. Hybridization of the crRNA to the target nucleic acid can activate Cas9 for targeted nucleic acid cleavage.
  • the target nucleic acid in a Type II CRISPR system is referred to as a protospacer adjacent motif (PAM).
  • PAM protospacer adjacent motif
  • the PAM is essential to facilitate binding of a site-directed polypeptide ⁇ e.g., Cas9) to the target nucleic acid.
  • Type II systems also referred to as Nmeni or CASS4 are further subdivided into Type II-A (CASS4) and II-B (CASS4a).
  • CASS4a Type II-A
  • WO2013/176772 provides numerous examples and applications of the CRISPR/Cas endonuclease system for site-specific gene editing.
  • Type V CRISPR systems have several important differences from Type II systems.
  • Cpf 1 is a single RNA-guided endonuclease that, in contrast to Type II systems, lacks tracrRNA.
  • Cpf 1 -associated CRISPR arrays can be processed into mature crRNAs without the requirement of an additional trans-activating tracrRNA.
  • the Type V CRISPR array can be processed into short mature crRNAs of 42-44 nucleotides in length, with each mature crRNA beginning with 19 nucleotides of direct repeat followed by 23-25 nucleotides of spacer sequence.
  • mature crRNAs in Type II systems can start with 20-24 nucleotides of spacer sequence followed by about 22 nucleotides of direct repeat.
  • Cpfl can utilize a T- rich protospacer-adjacent motif such that Cpfl-crRNA complexes efficiently cleave target DNA preceded by a short T-rich PAM, which is in contrast to the G-rich PAM following the target DNA for Type II systems.
  • Type V systems cleave at a point that is distant from the PAM
  • Type II systems cleave at a point that is adjacent to the PAM.
  • Cpfl cleaves DNA via a staggered DNA double-stranded break with a 4 or 5 nucleotide 5' overhang.
  • Type II systems cleave via a blunt double-stranded break.
  • Cpfl contains a predicted RuvC-like endonuclease domain, but lacks a second HNH endonuclease domain, which is in contrast to Type II systems.
  • Exemplary CRISPR/Cas polypeptides include the Cas9 polypeptides as published in Fonfara et al, Nucleic Acids Research, 42: 2577-2590 (2014).
  • the CRISPR/Cas gene naming system has undergone extensive rewriting since the Cas genes were discovered.
  • Fonfara et al. also provides PAM sequences for the Cas9 polypeptides from various species (see also Table 1 infra).
  • cellular, ex vivo and in vivo methods for using genome engineering tools to create permanent changes to the genome by: 1) deleting the abnormal repeat expansion within the ATXN3 gene, by inducing two double-stranded DNA breaks at both sides of the expanded region, and replacing with a corrected sequence; 2) deleting the abnormal repeat expansion within the ATXN3 gene, by inducing one double-stranded DNA break proximal to the expanded region, and replacing with a corrected sequence; 3) deleting or mutating the ATXN3 gene by inducing one or more insertions or deletions within or near the ATXN3 gene or other DNA sequences that encode regulatory elements of the ATXN3 gene, or 4) deleting the mutant ATXN3 gene and inserting a wild-type ATXN3 gene, a cDNA or a minigene (comprised of one or more exons and introns or natural or synthetic introns) into the ATXN3 gene locus or a safe harbor locus.
  • Such methods use endonucleases, such as CRISPR-associated (Cas9, Cpfl and the like) nucleases, to permanently edit within or near the genomic locus of the ATXN3 gene or other DNA sequences that encode regulatory elements of the ATXN3 gene.
  • endonucleases such as CRISPR-associated (Cas9, Cpfl and the like) nucleases
  • examples set forth in the present disclosure can help to restore the wild-type sequence of, or otherwise reduce or eliminate the expression of, the mutant ATXN3 gene with as few as a single treatment (rather than deliver potential therapies for the lifetime of the patient).
  • a site-directed polypeptide is a nuclease used in genome editing to cleave DNA.
  • the site-directed polypeptide can be administered to a cell or a patient as either: one or more polypeptides, or one or more mRNAs encoding the polypeptide. Any of the enzymes or orthologs listed in SEQ ID NOs: 1-620, or disclosed herein, may be utilized in the methods herein.
  • the site-directed polypeptide can bind to a guide RNA that, in turn, specifies the site in the target DNA to which the polypeptide is directed.
  • the site-directed polypeptide can be an endonuclease, such as a DNA endonuclease.
  • a site-directed polypeptide can comprise a plurality of nucleic acid-cleaving (i.e., nuclease) domains. Two or more nucleic acid-cleaving domains can be linked together via a linker.
  • the linker can comprise a flexible linker.
  • Linkers can comprise 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, 30, 35, 40 or more amino acids in length.
  • Naturally-occurring wild-type Cas9 enzymes comprise two nuclease domains, a UNH nuclease domain and a RuvC domain.
  • Cas9 refers to both a naturally- occurring and a recombinant Cas9.
  • Cas9 enzymes contemplated herein can comprise a HNH or HNH-like nuclease domain, and/or a RuvC or RuvC-like nuclease domain.
  • HNH or HNH-like domains comprise a McrA-like fold.
  • HNH or HNH-like domains comprises two antiparallel ⁇ -strands and an a-helix.
  • HNH or HNH-like domains comprises a metal binding site (e.g., a divalent cation binding site).
  • HNH or HNH-like domains can cleave one strand of a target nucleic acid (e.g., the complementary strand of the crRNA targeted strand).
  • RuvC or RuvC-like domains comprise an RNaseH or RNaseH-like fold.
  • RuvC/RNaseH domains are involved in a diverse set of nucleic acid-based functions including acting on both RNA and DNA.
  • the RNaseH domain comprises 5 ⁇ -strands surrounded by a plurality of a-helices.
  • RuvC/RNaseH or RuvC/RNaseH-like domains comprise a metal binding site (e.g., a divalent cation binding site).
  • RuvC/RNaseH or RuvC/RNaseH-like domains can cleave one strand of a target nucleic acid (e.g., the non-complementary strand of a double- stranded target DNA).
  • Site-directed polypeptides can introduce double-strand breaks or single-strand breaks in nucleic acids, e.g., genomic DNA.
  • the double-strand break can stimulate a cell's endogenous DNA-repair pathways (e.g., homology-dependent repair (HDR) or NHEJ or alternative nonhomologous end joining (A-NHEJ) or microhomology-mediated end joining (MMEJ)).
  • NHEJ can repair cleaved target nucleic acid without the need for a homologous template. This can sometimes result in small deletions or insertions (indels) in the target nucleic acid at the site of cleavage, and can lead to disruption or alteration of gene expression.
  • HDR can occur when a homologous repair template, or donor, is available.
  • the homologous donor template can comprise sequences that are homologous to sequences flanking the target nucleic acid cleavage site.
  • the sister chromatid can be used by the cell as the repair template.
  • the repair template can be supplied as an exogenous nucleic acid, such as a plasmid, duplex oligonucleotide, single-strand oligonucleotide or viral nucleic acid.
  • an additional nucleic acid sequence such as a transgene
  • modification such as a single or multiple base change or a deletion
  • MMEJ can result in a genetic outcome that is similar to NHEJ in that small deletions and insertions can occur at the cleavage site.
  • MMEJ can make use of homologous sequences of a few base pairs flanking the cleavage site to drive a favored end-joining DNA repair outcome. In some instances, it may be possible to predict likely repair outcomes based on analysis of potential microhomologies in the nuclease target regions.
  • homologous recombination can be used to insert an exogenous polynucleotide sequence into the target nucleic acid cleavage site.
  • An exogenous polynucleotide sequence is termed a "donor polynucleotide" (or donor or donor sequence) herein.
  • the donor polynucleotide, a portion of the donor polynucleotide, a copy of the donor polynucleotide, or a portion of a copy of the donor polynucleotide can be inserted into the target nucleic acid cleavage site.
  • the donor polynucleotide can be an exogenous polynucleotide sequence, i.e., a sequence that does not naturally occur at the target nucleic acid cleavage site.
  • the modifications of the target DNA due to NHEJ and/or HDR can lead to, for example, mutations, deletions, alterations, integrations, gene correction, gene replacement, gene tagging, transgene insertion, nucleotide deletion, gene disruption, translocations and/or gene mutation.
  • the processes of deleting genomic DNA and integrating non-native nucleic acid into genomic DNA are examples of genome editing.
  • the site-directed polypeptide can comprise an amino acid sequence having at least 10%, at least 15%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% amino acid sequence identity to a wild-type exemplary site-directed polypeptide [e.g., Cas9 from S. pyogenes, US2014/0068797 Sequence ID No. 8 or Sapranauskas et al, Nucleic Acids Res, 39(21): 9275-9282 (2011)], and various other site-directed polypeptides.
  • a wild-type exemplary site-directed polypeptide e.g., Cas9 from S. pyogenes, US2014/0068797 Sequence ID No. 8 or Sapranauskas et al, Nucleic Acids Res, 39(21): 9275-9282 (2011)
  • the site-directed polypeptide can comprise at least 70, 75, 80, 85, 90, 95, 97, 99, or 100%) identity to a wild-type site-directed polypeptide ⁇ e.g., Cas9 from S. pyogenes, supra) over 10 contiguous amino acids.
  • the site-directed polypeptide can comprise at most: 70, 75, 80, 85, 90, 95, 97, 99, or 100% identity to a wild-type site-directed polypeptide (e.g., Cas9 from S. pyogenes, supra) over 10 contiguous amino acids.
  • the site-directed polypeptide can comprise at least: 70, 75, 80, 85, 90, 95, 97, 99, or 100% identity to a wild-type site-directed polypeptide (e.g., Cas9 from S.
  • the site-directed polypeptide can comprise at most: 70, 75, 80, 85, 90, 95, 97, 99, or 100%) identity to a wild-type site-directed polypeptide (e.g., Cas9 from S. pyogenes, supra) over 10 contiguous amino acids in a ⁇ nuclease domain of the site-directed polypeptide.
  • the site-directed polypeptide can comprise at least: 70, 75, 80, 85, 90, 95, 97, 99, or 100% identity to a wild-type site-directed polypeptide (e.g., Cas9 from S.
  • the site-directed polypeptide can comprise at most: 70, 75, 80, 85, 90, 95, 97, 99, or 100%) identity to a wild-type site-directed polypeptide (e.g., Cas9 from S. pyogenes, supra) over 10 contiguous amino acids in a RuvC nuclease domain of the site-directed polypeptide.
  • a wild-type site-directed polypeptide e.g., Cas9 from S. pyogenes, supra
  • the site-directed polypeptide can comprise a modified form of a wild-type exemplary site-directed polypeptide.
  • the modified form of the wild- type exemplary site-directed polypeptide can comprise a mutation that reduces the nucleic acid-cleaving activity of the site- directed polypeptide.
  • the modified form of the wild-type exemplary site-directed polypeptide can have less than 90%, less than 80%, less than 70%, less than 60%, less than 50%, less than 40%), less than 30%, less than 20%, less than 10%, less than 5%, or less than 1% of the nucleic acid-cleaving activity of the wild-type exemplary site-directed polypeptide (e.g., Cas9 from S. pyogenes, supra).
  • the modified form of the site-directed polypeptide can have no substantial nucleic acid-cleaving activity.
  • a site-directed polypeptide is a modified form that has no substantial nucleic acid-cleaving activity, it is referred to herein as "enzymatically inactive.”
  • the modified form of the site-directed polypeptide can comprise a mutation such that it can induce a single-strand break (SSB) on a target nucleic acid (e.g., by cutting only one of the sugar-phosphate backbones of a double-strand target nucleic acid).
  • SSB single-strand break
  • the mutation can result in less than 90%, less than 80%>, less than 70%, less than 60%>, less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 5%, or less than 1% of the nucleic acid-cleaving activity in one or more of the plurality of nucleic acid-cleaving domains of the wild-type site directed polypeptide (e.g., Cas9 from S. pyogenes, supra).
  • the wild-type site directed polypeptide e.g., Cas9 from S. pyogenes, supra.
  • the mutation can result in one or more of the plurality of nucleic acid-cleaving domains retaining the ability to cleave the complementary strand of the target nucleic acid, but reducing its ability to cleave the non-complementary strand of the target nucleic acid.
  • the mutation can result in one or more of the plurality of nucleic acid-cleaving domains retaining the ability to cleave the non-complementary strand of the target nucleic acid, but reducing its ability to cleave the complementary strand of the target nucleic acid. For example, residues in the wild-type exemplary S.
  • pyogenes Cas9 polypeptide such as Asp 10, His840, Asn854 and Asn856, are mutated to inactivate one or more of the plurality of nucleic acid-cleaving domains (e.g., nuclease domains).
  • the residues to be mutated can correspond to residues Asp 10, His840, Asn854 and Asn856 in the wild-type exemplary S. pyogenes Cas9 polypeptide (e.g., as determined by sequence and/or structural alignment).
  • Non-limiting examples of mutations include D10A, H840A, N854A or N856A.
  • mutations other than alanine substitutions can be suitable.
  • a D10A mutation can be combined with one or more of H840A, N854A, or N856A mutations to produce a site-directed polypeptide substantially lacking DNA cleavage activity.
  • a H840A mutation can be combined with one or more of D10A, N854A, or N856A mutations to produce a site-directed polypeptide substantially lacking DNA cleavage activity.
  • a N854A mutation can be combined with one or more of H840A, D10A, or N856A mutations to produce a site-directed polypeptide substantially lacking DNA cleavage activity.
  • a N856A mutation can be combined with one or more of H840A, N854A, or D10A mutations to produce a site-directed polypeptide substantially lacking DNA cleavage activity.
  • Site-directed polypeptides that comprise one substantially inactive nuclease domain are referred to as
  • RNA-guided endonucleases for example Cas9
  • Wild type Cas9 is typically guided by a single guide RNA designed to hybridize with a specified -20 nucleotide sequence in the target sequence (such as an endogenous genomic locus).
  • nickase variants of Cas9 each only cut one strand, in order to create a double-strand break it is necessary for a pair of nickases to bind in close proximity and on opposite strands of the target nucleic acid, thereby creating a pair of nicks, which is the equivalent of a double-strand break.
  • nickases can also be used to promote HDR versus NHEJ. HDR can be used to introduce selected changes into target sites in the genome through the use of specific donor sequences that effectively mediate the desired changes.
  • Mutations contemplated can include substitutions, additions, and deletions, or any combination thereof.
  • the mutation converts the mutated amino acid to alanine.
  • the mutation converts the mutated amino acid to another amino acid (e.g., glycine, serine, threonine, cysteine, valine, leucine, isoleucine, methionine, proline, phenylalanine, tyrosine, tryptophan, aspartic acid, glutamic acid, asparagine, glutamine, histidine, lysine, or arginine).
  • the mutation converts the mutated amino acid to a non-natural amino acid (e.g., selenomethionine).
  • the mutation converts the mutated amino acid to amino acid mimics (e.g., phosphomimics).
  • the mutation can be a conservative mutation.
  • the mutation converts the mutated amino acid to amino acids that resemble the size, shape, charge, polarity, conformation, and/or rotamers of the mutated amino acids (e.g., cysteine/serine mutation, lysine/asparagine mutation,
  • the mutation can cause a shift in reading frame and/or the creation of a premature stop codon. Mutations can cause changes to regulatory regions of genes or loci that affect expression of one or more genes.
  • the site-directed polypeptide (e.g., variant, mutated, enzymatically inactive and/or conditionally enzymatically inactive site-directed polypeptide) can target nucleic acid.
  • the site- directed polypeptide (e.g., variant, mutated, enzymatically inactive and/or conditionally enzymatically inactive endoribonuclease) can target DNA.
  • the site-directed polypeptide e.g., variant, mutated, enzymatically inactive and/or conditionally enzymatically inactive
  • endoribonuclease can target RNA.
  • the site-directed polypeptide can comprise one or more non-native sequences (e.g., the site-directed polypeptide is a fusion protein).
  • the site-directed polypeptide can comprise an amino acid sequence comprising at least 15% amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), a nucleic acid binding domain, and two nucleic acid cleaving domains (i.e., a HNH domain and a RuvC domain).
  • a Cas9 from a bacterium e.g., S. pyogenes
  • a nucleic acid binding domain e.g., S. pyogenes
  • two nucleic acid cleaving domains i.e., a HNH domain and a RuvC domain
  • the site-directed polypeptide can comprise an amino acid sequence comprising at least 15% amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), and two nucleic acid cleaving domains (i.e., a HNH domain and a RuvC domain).
  • a Cas9 from a bacterium e.g., S. pyogenes
  • two nucleic acid cleaving domains i.e., a HNH domain and a RuvC domain.
  • the site-directed polypeptide can comprise an amino acid sequence comprising at least 15%) amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), and two nucleic acid cleaving domains, wherein one or both of the nucleic acid cleaving domains comprise at least 50%) amino acid identity to a nuclease domain from Cas9 from a bacterium (e.g., S.
  • the site-directed polypeptide can comprise an amino acid sequence comprising at least 15%) amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), two nucleic acid cleaving domains (i.e., a HNH domain and a RuvC domain), and a non-native sequence (for example, a nuclear localization signal) or a linker linking the site-directed polypeptide to a non- native sequence.
  • a bacterium e.g., S. pyogenes
  • two nucleic acid cleaving domains i.e., a HNH domain and a RuvC domain
  • a non-native sequence for example, a nuclear localization signal
  • the site-directed polypeptide can comprise an amino acid sequence comprising at least 15%o amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), two nucleic acid cleaving domains (i.e., a HNH domain and a RuvC domain), wherein the site-directed polypeptide comprises a mutation in one or both of the nucleic acid cleaving domains that reduces the cleaving activity of the nuclease domains by at least 50%.
  • a bacterium e.g., S. pyogenes
  • two nucleic acid cleaving domains i.e., a HNH domain and a RuvC domain
  • the site-directed polypeptide can comprise an amino acid sequence comprising at least 15%o amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), and two nucleic acid cleaving domains (i.e., a HNH domain and a RuvC domain), wherein one of the nuclease domains comprises mutation of aspartic acid 10, and/or wherein one of the nuclease domains can comprise a mutation of histidine 840, and wherein the mutation reduces the cleaving activity of the nuclease domain(s) by at least 50%.
  • the one or more site-directed polypeptides e.g.
  • DNA endonucleases can comprise two nickases that together effect one double-strand break at a specific locus in the genome, or four nickases that together effect or cause two double-strand breaks at specific loci in the genome.
  • one site-directed polypeptide e.g. DNA endonuclease, can effect or cause one double-strand break at a specific locus in the genome.
  • Non-limiting examples of Cas9 orthologs from other bacterial strains include but are not limited to, Cas proteins identified in Acaryochloris marina MBIC11017; Acetohalobium arabaticum DSM 5501; Acidithiobacillus caldus; Acidithiobacillus ferrooxidans ATCC 23270; Alicyclobacillus acidocaldarius LAAl; Alicyclobacillus acidocaldarius subsp. acidocaldarius DSM 446; Allochromatium vinosum DSM 180; Ammonifex degensii KC4; Anabaena variabilis ATCC 29413; Arthrospira maxima CS-328; Arthrospira platensis sir. Paraca; Arthrospira sp. PCC 8005; Bacillus pseudomycoides DSM 12442; Bacillus selenitireducens MLS10;
  • Cas9 orthologs In addition to Cas9 orthologs, other Cas9 variants such as fusion proteins of inactive dCas9 and effector domains with different functions may be served as a platform for genetic modulation. Any of the foregoing enzymes may be useful in the present disclosure.
  • endonucleases that may be utilized in the present disclosure are given in SEQ ID NOs: 1-620. These proteins may be modified before use or may be encoded in a nucleic acid sequence such as a DNA, RNA or mRNA or within a vector construct such as the plasmids or AAV vectors taught herein. Further, they may be codon optimized.
  • SEQ ID NOs: 1-620 disclose a non-exhaustive listing of endonuclease sequences.
  • the present disclosure provides a genome-targeting nucleic acid that can direct the activities of an associated polypeptide ⁇ e.g., a site-directed polypeptide) to a specific target sequence within a target nucleic acid.
  • the genome-targeting nucleic acid can be an RNA.
  • a genome-targeting RNA is referred to as a "guide RNA" or "gRNA" herein.
  • a guide RNA can comprise at least a spacer sequence that hybridizes to a target nucleic acid sequence of interest, and a CRISPR repeat sequence.
  • the gRNA also comprises a second RNA called the tracrRNA sequence.
  • the CRISPR repeat sequence and tracrRNA sequence hybridize to each other to form a duplex.
  • the crRNA forms a duplex.
  • the duplex can bind a site-directed polypeptide, such that the guide RNA and site-direct polypeptide form a complex.
  • the genome- targeting nucleic acid can provide target specificity to the complex by virtue of its association with the site-directed polypeptide. The genome-targeting nucleic acid thus can direct the activity of the site-directed polypeptide.
  • Exemplary guide RNAs include the spacer sequences in SEQ ID NOs: 5305-56902 of the Sequence Listing.
  • each guide RNA can be designed to include a spacer sequence complementary to its genomic target sequence.
  • each of the spacer sequences in SEQ ID NOs: 5305-56902 of the Sequence Listing can be put into a single RNA chimera or a crRNA (along with a corresponding tracrRNA). See Jinek et al, Science, 337, 816-821 (2012) and Deltcheva et al, Nature, 471, 602-607 (2011).
  • the genome-targeting nucleic acid can be a double-molecule guide RNA.
  • the genome-targeting nucleic acid can be a single-molecule guide RNA.
  • a double-molecule guide RNA can comprise two strands of RNA.
  • the first strand comprises in the 5' to 3' direction, an optional spacer extension sequence, a spacer sequence and a minimum CRISPR repeat sequence.
  • the second strand can comprise a minimum tracrRNA sequence (complementary to the minimum CRISPR repeat sequence), a 3' tracrRNA sequence and an optional tracrRNA extension sequence.
  • a single-molecule guide RNA (sgRNA) in a Type II system can comprise, in the 5' to 3' direction, an optional spacer extension sequence, a spacer sequence, a minimum CRISPR repeat sequence, a single-molecule guide linker, a minimum tracrRNA sequence, a 3 ' tracrRNA sequence and an optional tracrRNA extension sequence.
  • the optional tracrRNA extension can comprise elements that contribute additional functionality (e.g., stability) to the guide RNA.
  • the single-molecule guide linker can link the minimum CRISPR repeat and the minimum tracrRNA sequence to form a hairpin structure.
  • the optional tracrRNA extension can comprise one or more hairpins.
  • the sgRNA can comprise a 20 nucleotide spacer sequence at the 5' end of the sgRNA sequence.
  • the sgRNA can comprise a less than a 20 nucleotide spacer sequence at the 5' end of the sgRNA sequence.
  • the sgRNA can comprise a more than 20 nucleotide spacer sequence at the 5' end of the sgRNA sequence.
  • the sgRNA can comprise a variable length spacer sequence with 17-30 nucleotides at the 5' end of the sgRNA sequence (see Table 1).
  • the sgRNA can comprise no uracil at the 3 'end of the sgRNA sequence, such as in SEQ ID NO: 56935 of Table 1.
  • the sgRNA can comprise one or more uracil at the 3 'end of the sgRNA sequence, such as in SEQ ID NO: 56936 in Table 1.
  • the sgRNA can comprise 1 uracil (U) at the 3 ' end of the sgRNA sequence.
  • the sgRNA can comprise 2 uracil (UU) at the 3 ' end of the sgRNA sequence.
  • the sgRNA can comprise 3 uracil (UUU) at the 3 ' end of the sgRNA sequence.
  • the sgRNA can comprise 4 uracil (UUUU) at the 3 ' end of the sgRNA sequence.
  • the sgRNA can comprise 5 uracil (UUUUU) at the 3 ' end of the sgRNA sequence.
  • the sgRNA can comprise 6 uracil (UUUUUU) at the 3 ' end of the sgRNA sequence.
  • the sgRNA can comprise 7 uracil (ULJLJUUUU) at the 3 ' end of the sgRNA sequence.
  • the sgRNA can comprise 8 uracil (UULJLJUUUU) at the 3 ' end of the sgRNA sequence.
  • modified sgRNAs can comprise one or more 2'-0-methyl phosphorothioate nucleotides.
  • a single-molecule guide RNA (sgRNA) in a Type V system can comprise, in the 5' to 3' direction, a minimum CRISPR repeat sequence and a spacer sequence.
  • guide RNAs used in the CRISPR/Cas9 or CRISPR/Cpfl system can be readily synthesized by chemical means, as illustrated below and described in the art. While chemical synthetic procedures are continually expanding, purifications of such RNAs by procedures such as high performance liquid chromatography (HPLC, which avoids the use of gels such as PAGE) tends to become more challenging as polynucleotide lengths increase significantly beyond a hundred or so nucleotides.
  • HPLC high performance liquid chromatography
  • One approach used for generating RNAs of greater length is to produce two or more molecules that are ligated together.
  • RNAs such as those encoding a Cas9 or Cpf 1 endonuclease
  • RNA modifications can be introduced during or after chemical synthesis and/or enzymatic generation of RNAs, e.g., modifications that enhance stability, reduce the likelihood or degree of innate immune response, and/or enhance other attributes, as described in the art.
  • a spacer extension sequence can modify activity, provide stability and/or provide a location for modifications of a genome- targeting nucleic acid.
  • a spacer extension sequence can modify on- or off-target activity or specificity.
  • a spacer extension sequence can be provided.
  • the spacer extension sequence can have a length of more than 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 220, 240, 260, 280, 300, 320, 340, 360, 380, 400, 1000, 2000, 3000, 4000, 5000, 6000, or 7000 or more nucleotides.
  • the spacer extension sequence can have a length of less than 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 220, 240, 260, 280, 300, 320, 340, 360, 380, 400, 1000, 2000, 3000, 4000, 5000, 6000, 7000 or more nucleotides.
  • the spacer extension sequence can be less than 10 nucleotides in length.
  • the spacer extension sequence can be between 10-30 nucleotides in length.
  • the spacer extension sequence can be between 30-70 nucleotides in length.
  • the spacer extension sequence can comprise another moiety (e.g., a stability control sequence, an endoribonuclease binding sequence, a ribozyme).
  • the moiety can decrease or increase the stability of a nucleic acid targeting nucleic acid.
  • the moiety can be a transcriptional terminator segment (i.e., a transcription termination sequence).
  • the moiety can function in a eukary otic cell.
  • the moiety can function in a prokary otic cell.
  • the moiety can function in both eukaryotic and prokaryotic cells.
  • Non-limiting examples of suitable moieties include: a 5' cap (e.g., a 7-methylguanylate cap (m7 G)), a riboswitch sequence (e.g., to allow for regulated stability and/or regulated accessibility by proteins and protein complexes), a sequence that forms a dsRNA duplex (i.e., a hairpin), a sequence that targets the RNA to a subcellular location (e.g., nucleus, mitochondria, chloroplasts, and the like), a modification or sequence that provides for tracking (e.g., direct conjugation to a fluorescent molecule, conjugation to a moiety that facilitates fluorescent detection, a sequence that allows for fluorescent detection, etc.), and/or a modification or sequence that provides a binding site for proteins (e.g., proteins that act on DNA, including transcriptional activators, transcriptional repressors, DNA methyltransferases, DNA demethylases, histone acetyltransferases, histone deacet
  • the spacer sequence hybridizes to a sequence in a target nucleic acid of interest.
  • the spacer of a genome-targeting nucleic acid can interact with a target nucleic acid in a sequence- specific manner via hybridization (i.e., base pairing).
  • the nucleotide sequence of the spacer can vary depending on the sequence of the target nucleic acid of interest.
  • the spacer sequence can be designed to hybridize to a target nucleic acid that is located 5' of a PAM of the Cas9 enzyme used in the system.
  • the spacer may perfectly match the target sequence or may have mismatches.
  • Each Cas9 enzyme has a particular PAM sequence that it recognizes in a target DNA.
  • S. pyogenes recognizes in a target nucleic acid a PAM that comprises the sequence 5'-NRG-3', where R comprises either A or G, where N is any nucleotide and N is immediately 3' of the target nucleic acid sequence targeted by the spacer sequence.
  • the target nucleic acid sequence can comprise 20 nucleotides.
  • the target nucleic acid can comprise less than 20 nucleotides.
  • the target nucleic acid can comprise more than 20 nucleotides.
  • the target nucleic acid can comprise at least: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides.
  • the target nucleic acid can comprise at most: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides.
  • the target nucleic acid sequence can comprise 20 bases immediately 5' of the first nucleotide of the PAM.
  • the target nucleic acid in a sequence comprising 5'-NNNNNNNNNNNNNNNNNNNNNNNNNRG-3' (SEQ ID NO: 56933), can comprise the sequence that corresponds to the Ns, wherein N is any nucleotide, and the underlined NRG sequence is the S. pyogenes PAM.
  • This target nucleic acid sequence is often referred to as the PAM strand, and the complementary nucleic acid sequence is often referred to the non-PAM strand.
  • the spacer sequence hybridizes to the non-PAM strand of the target nucleic acid ( Figures 1 A and IB).
  • the spacer sequence that hybridizes to the target nucleic acid can have a length of at least about 6 nucleotides (nt).
  • the spacer sequence can be at least about 6 nt, at least about 10 nt, at least about 15 nt, at least about 18 nt, at least about 19 nt, at least about 20 nt, at least about 25 nt, at least about 30 nt, at least about 35 nt or at least about 40 nt, from about 6 nt to about 80 nt, from about 6 nt to about 50 nt, from about 6 nt to about 45 nt, from about 6 nt to about 40 nt, from about 6 nt to about 35 nt, from about 6 nt to about 30 nt, from about 6 nt to about 25 nt, from about 6 nt to about 20 nt, from about 6 nt to about 19 nt, from about 10 nt to about 50 nt, from
  • the spacer sequence can comprise 20 nucleotides. In some examples, the spacer can comprise 19 nucleotides. In some examples, the spacer can comprise 18 nucleotides. In some examples, the spacer can comprise 22 nucleotides.
  • the percent complementarity between the spacer sequence and the target nucleic acid is at least about 30%, at least about 40%, at least about 50%, at least about 60%), at least about 65%>, at least about 70%, at least about 75%, at least about 80%, at least about 85%), at least about 90%, at least about 95%, at least about 97%, at least about 98%, at least about 99%, or 100%.
  • the percent complementarity between the spacer sequence and the target nucleic acid is at most about 30%, at most about 40%, at most about 50%), at most about 60%, at most about 65%, at most about 70%, at most about 75%, at most about 80%), at most about 85%, at most about 90%, at most about 95%, at most about 97%, at most about 98%, at most about 99%, or 100%.
  • the percent complementarity between the spacer sequence and the target nucleic acid is 100% over the six contiguous 5'-most nucleotides of the target sequence of the complementary strand of the target nucleic acid.
  • the percent complementarity between the spacer sequence and the target nucleic acid can be at least 60% over about 20 contiguous nucleotides.
  • the length of the spacer sequence and the target nucleic acid can differ by 1 to 6 nucleotides, which may be thought of as a bulge or bulges.
  • the spacer sequence can be designed or chosen using a computer program.
  • the computer program can use variables, such as predicted melting temperature, secondary structure formation, predicted annealing temperature, sequence identity, genomic context, chromatin accessibility, % GC, frequency of genomic occurrence (e.g., of sequences that are identical or are similar but vary in one or more spots as a result of mismatch, insertion or deletion), methylation status, presence of S Ps, and the like.
  • a minimum CRISPR repeat sequence is a sequence with at least about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%), about 90%, about 95%, or 100% sequence identity to a reference CRISPR repeat sequence (e.g. , crRNA from S. pyogenes).
  • a reference CRISPR repeat sequence e.g. , crRNA from S. pyogenes
  • a minimum CRISPR repeat sequence comprises nucleotides that can hybridize to a minimum tracrRNA sequence in a cell.
  • the minimum CRISPR repeat sequence and a minimum tracrRNA sequence can form a duplex, i.e. a base-paired double-stranded structure. Together, the minimum CRISPR repeat sequence and the minimum tracrRNA sequence can bind to the site-directed polypeptide. At least a part of the minimum CRISPR repeat sequence can hybridize to the minimum tracrRNA sequence.
  • At least a part of the minimum CRISPR repeat sequence can comprise at least about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or 100%) complementary to the minimum tracrRNA sequence. In some aspects, at least a part of the minimum CRISPR repeat sequence comprises at most about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or 100% complementary to the minimum tracrRNA sequence.
  • the minimum CRISPR repeat sequence can have a length from about 7 nucleotides to about 100 nucleotides.
  • the length of the minimum CRISPR repeat sequence is from about 7 nucleotides (nt) to about 50 nt, from about 7 nt to about 40 nt, from about 7 nt to about 30 nt, from about 7 nt to about 25 nt, from about 7 nt to about 20 nt, from about 7 nt to about 15 nt, from about 8 nt to about 40 nt, from about 8 nt to about 30 nt, from about 8 nt to about 25 nt, from about 8 nt to about 20 nt, from about 8 nt to about 15 nt, from about 15 nt to about 100 nt, from about 15 nt to about 80 nt, from about 15 nt to about 50 nt, from about 15 nt to about 40 nt, from about 15 nt to
  • the minimum CRISPR repeat sequence can be at least about 60% identical to a reference minimum CRISPR repeat sequence (e.g., wild-type crRNA from S. pyogenes) over a stretch of at least 6, 7, or 8 contiguous nucleotides.
  • a reference minimum CRISPR repeat sequence e.g., wild-type crRNA from S. pyogenes
  • the minimum CRISPR repeat sequence can be at least about 65% identical, at least about 70% identical, at least about 75% identical, at least about 80% identical, at least about 85% identical, at least about 90% identical, at least about 95% identical, at least about 98% identical, at least about 99% identical or 100% identical to a reference minimum CRISPR repeat sequence over a stretch of at least 6, 7, or 8 contiguous nucleotides.
  • a minimum tracrRNA sequence can be a sequence with at least about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%), about 95%), or 100%> sequence identity to a reference tracrRNA sequence (e.g., wild type tracrRNA from S. pyogenes).
  • a reference tracrRNA sequence e.g., wild type tracrRNA from S. pyogenes.
  • a minimum tracrRNA sequence can comprise nucleotides that hybridize to a minimum CRISPR repeat sequence in a cell.
  • a minimum tracrRNA sequence and a minimum CRISPR repeat sequence form a duplex, i.e. a base-paired double-stranded structure. Together, the minimum tracrRNA sequence and the minimum CRISPR repeat can bind to a site-directed polypeptide. At least a part of the minimum tracrRNA sequence can hybridize to the minimum CRISPR repeat sequence.
  • the minimum tracrRNA sequence can be at least about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%), about 95%, or 100% complementary to the minimum CRISPR repeat sequence.
  • the minimum tracrRNA sequence can have a length from about 7 nucleotides to about 100 nucleotides.
  • the minimum tracrRNA sequence can be from about 7 nucleotides (nt) to about 50 nt, from about 7 nt to about 40 nt, from about 7 nt to about 30 nt, from about 7 nt to about 25 nt, from about 7 nt to about 20 nt, from about 7 nt to about 15 nt, from about 8 nt to about 40 nt, from about 8 nt to about 30 nt, from about 8 nt to about 25 nt, from about 8 nt to about 20 nt, from about 8 nt to about 15 nt, from about 15 nt to about 100 nt, from about 15 nt to about 80 nt, from about 15 nt to about 50 nt, from about 15 nt to about 40 nt, from about 15 nt to about
  • the minimum tracrRNA sequence can be approximately 9 nucleotides in length.
  • the minimum tracrRNA sequence can be approximately 12 nucleotides.
  • the minimum tracrRNA can consist of tracrRNA nt 23-48 described in Jinek et al, supra.
  • the minimum tracrRNA sequence can be at least about 60% identical to a reference minimum tracrRNA ⁇ e.g., wild type, tracrRNA from S. pyogenes) sequence over a stretch of at least 6, 7, or 8 contiguous nucleotides.
  • the minimum tracrRNA sequence can be at least about 65% identical, about 70% identical, about 75% identical, about 80% identical, about 85%) identical, about 90% identical, about 95% identical, about 98% identical, about 99% identical or 100% identical to a reference minimum tracrRNA sequence over a stretch of at least 6, 7, or 8 contiguous nucleotides.
  • the duplex between the minimum CRISPR RNA and the minimum tracrRNA can comprise a double helix.
  • the duplex between the minimum CRISPR RNA and the minimum tracrRNA can comprise at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nucleotides.
  • the duplex between the minimum CRISPR RNA and the minimum tracrRNA can comprise at most about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nucleotides.
  • the duplex can comprise a mismatch (i.e., the two strands of the duplex are not 100% complementary).
  • the duplex can comprise at least about 1, 2, 3, 4, or 5 or mismatches.
  • the duplex can comprise at most about 1, 2, 3, 4, or 5 or mismatches.
  • the duplex can comprise no more than 2 mismatches.
  • a bulge is an unpaired region of nucleotides within the duplex.
  • a bulge can contribute to the binding of the duplex to the site-directed polypeptide.
  • the bulge can comprise, on one side of the duplex, an unpaired 5'-XXXY-3' where X is any purine and Y comprises a nucleotide that can form a wobble pair with a nucleotide on the opposite strand, and an unpaired nucleotide region on the other side of the duplex.
  • the number of unpaired nucleotides on the two sides of the duplex can be different.
  • the bulge can comprise an unpaired purine (e.g., adenine) on the minimum CRISPR repeat strand of the bulge.
  • the bulge can comprise an unpaired 5'-AAGY-3' of the minimum tracrRNA sequence strand of the bulge, where Y comprises a nucleotide that can form a wobble pairing with a nucleotide on the minimum CRISPR repeat strand.
  • a bulge on the minimum CRISPR repeat side of the duplex can comprise at least 1, 2, 3, 4, or 5 or more unpaired nucleotides.
  • a bulge on the minimum CRISPR repeat side of the duplex can comprise at most 1, 2, 3, 4, or 5 or more unpaired nucleotides.
  • a bulge on the minimum CRISPR repeat side of the duplex can comprise 1 unpaired nucleotide.
  • a bulge on the minimum tracrRNA sequence side of the duplex can comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more unpaired nucleotides.
  • a bulge on the minimum tracrRNA sequence side of the duplex can comprise at most 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more unpaired nucleotides.
  • a bulge on a second side of the duplex (e.g., the minimum tracrRNA sequence side of the duplex) can comprise 4 unpaired nucleotides.
  • a bulge can comprise at least one wobble pairing. In some examples, a bulge can comprise at most one wobble pairing. A bulge can comprise at least one purine nucleotide. A bulge can comprise at least 3 purine nucleotides. A bulge sequence can comprise at least 5 purine nucleotides. A bulge sequence can comprise at least one guanine nucleotide. In some examples, a bulge sequence can comprise at least one adenine nucleotide.
  • one or more hairpins can be located 3' to the minimum tracrRNA in the 3' tracrRNA sequence.
  • the hairpin can start at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 or more nucleotides 3' from the last paired nucleotide in the minimum CRISPR repeat and minimum tracrRNA sequence duplex.
  • the hairpin can start at most about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 or more nucleotides 3' of the last paired nucleotide in the minimum CRISPR repeat and minimum tracrRNA sequence duplex.
  • the hairpin can comprise at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 or more consecutive nucleotides.
  • the hairpin can comprise at most about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or more consecutive nucleotides.
  • the hairpin can comprise a CC dinucleotide (i.e., two consecutive cytosine nucleotides).
  • the hairpin can comprise duplexed nucleotides (e.g., nucleotides in a hairpin, hybridized together).
  • a hairpin can comprise a CC dinucleotide that is hybridized to a GG dinucleotide in a hairpin duplex of the 3' tracrRNA sequence.
  • One or more of the hairpins can interact with guide RNA-interacting regions of a site- directed polypeptide.
  • a 3' tracrRNA sequence can comprise a sequence with at least about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%), about 95%), or 100% sequence identity to a reference tracrRNA sequence (e.g., a tracrRNA from S. pyogenes).
  • a reference tracrRNA sequence e.g., a tracrRNA from S. pyogenes.
  • the 3' tracrRNA sequence can have a length from about 6 nucleotides to about 100 nucleotides.
  • the 3' tracrRNA sequence can have a length from about 6 nucleotides (nt) to about 50 nt, from about 6 nt to about 40 nt, from about 6 nt to about 30 nt, from about 6 nt to about 25 nt, from about 6 nt to about 20 nt, from about 6 nt to about 15 nt, from about 8 nt to about 40 nt, from about 8 nt to about 30 nt, from about 8 nt to about 25 nt, from about 8 nt to about 20 nt, from about 8 nt to about 15 nt, from about 15 nt to about 100 nt, from about 15 nt to about 80 nt, from about 15 nt to about 50 nt, from about 15 nt to about 40 nt, from about 15 nt to about
  • the 3' tracrRNA sequence can be at least about 60%> identical to a reference 3' tracrRNA sequence (e.g., wild type 3' tracrRNA sequence from S. pyogenes) over a stretch of at least 6, 7, or 8 contiguous nucleotides.
  • the 3' tracrRNA sequence can be at least about 60%) identical, about 65%> identical, about 70% identical, about 75% identical, about 80% identical, about 85% identical, about 90% identical, about 95% identical, about 98% identical, about 99%) identical, or 100% identical, to a reference 3' tracrRNA sequence (e.g., wild type 3' tracrRNA sequence from S. pyogenes) over a stretch of at least 6, 7, or 8 contiguous nucleotides.
  • a reference 3' tracrRNA sequence e.g., wild type 3' tracrRNA sequence from S. pyogenes
  • the 3' tracrRNA sequence can comprise more than one duplexed region (e.g., hairpin, hybridized region).
  • the 3' tracrRNA sequence can comprise two duplexed regions.
  • the 3' tracrRNA sequence can comprise a stem loop structure.
  • the stem loop structure in the 3' tracrRNA can comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20 or more nucleotides.
  • the stem loop structure in the 3' tracrRNA can comprise at most 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 or more nucleotides.
  • the stem loop structure can comprise a functional moiety.
  • the stem loop structure can comprise an aptamer, a ribozyme, a protein-interacting hairpin, a CRISPR array, an intron, or an exon.
  • the stem loop structure can comprise at least about 1, 2, 3, 4, or 5 or more functional moieties.
  • the stem loop structure can comprise at most about 1, 2, 3, 4, or 5 or more functional moieties.
  • the hairpin in the 3' tracrRNA sequence can comprise a P-domain.
  • the P-domain can comprise a double-stranded region in the hairpin.
  • a tracrRNA extension sequence may be provided whether the tracrRNA is in the context of single-molecule guides or double-molecule guides.
  • the tracrRNA extension sequence can have a length from about 1 nucleotide to about 400 nucleotides.
  • the tracrRNA extension sequence can have a length of more than 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 220, 240, 260, 280, 300, 320, 340, 360, 380, or 400 nucleotides.
  • the tracrRNA extension sequence can have a length from about 20 to about 5000 or more nucleotides.
  • the tracrRNA extension sequence can have a length of more than 1000 nucleotides.
  • the tracrRNA extension sequence can have a length of less than 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 220, 240, 260, 280, 300, 320, 340, 360, 380, 400 or more nucleotides.
  • the tracrRNA extension sequence can have a length of less than 1000 nucleotides.
  • the tracrRNA extension sequence can comprise less than 10 nucleotides in length.
  • the tracrRNA extension sequence can be 10-30 nucleotides in length.
  • the tracrRNA extension sequence can be 30-70 nucleotides in length.
  • the tracrRNA extension sequence can comprise a functional moiety ⁇ e.g., a stability control sequence, ribozyme, endoribonuclease binding sequence).
  • the functional moiety can comprise a transcriptional terminator segment ⁇ i.e., a transcription termination sequence).
  • the functional moiety can have a total length from about 10 nucleotides (nt) to about 100
  • nucleotides from about 10 nt to about 20 nt, from about 20 nt to about 30 nt, from about 30 nt to about 40 nt, from about 40 nt to about 50 nt, from about 50 nt to about 60 nt, from about 60 nt to about 70 nt, from about 70 nt to about 80 nt, from about 80 nt to about 90 nt, or from about 90 nt to about 100 nt, from about 15 nt to about 80 nt, from about 15 nt to about 50 nt, from about 15 nt to about 40 nt, from about 15 nt to about 30 nt, or from about 15 nt to about 25 nt.
  • the functional moiety can function in a eukaryotic cell.
  • the functional moiety can function in a prokaryotic cell.
  • the functional moiety can function in both eukaryotic and prokaryotic cells.
  • Non-limiting examples of suitable tracrRNA extension functional moieties include a 3' poly-adenylated tail, a riboswitch sequence ⁇ e.g., to allow for regulated stability and/or regulated accessibility by proteins and protein complexes), a sequence that forms a dsRNA duplex ⁇ i.e., a hairpin), a sequence that targets the RNA to a subcellular location ⁇ e.g., nucleus, mitochondria, chloroplasts, and the like), a modification or sequence that provides for tracking ⁇ e.g., direct conjugation to a fluorescent molecule, conjugation to a moiety that facilitates fluorescent detection, a sequence that allows for fluorescent detection, etc.), and/or a modification or sequence that provides a binding site for proteins (e.g., proteins that act on DNA, including transcriptional activators, transcriptional repressors, DNA methyltransferases, DNA demethylases, histone acetyltransferases, histone deacety
  • the linker sequence of a single-molecule guide nucleic acid can have a length from about 3 nucleotides to about 100 nucleotides.
  • a simple 4 nucleotide "tetraloop" (-GAAA-) was used, Science, 337(6096):816-821 (2012).
  • An illustrative linker has a length from about 3 nucleotides (nt) to about 90 nt, from about 3 nt to about 80 nt, from about 3 nt to about 70 nt, from about 3 nt to about 60 nt, from about 3 nt to about 50 nt, from about 3 nt to about 40 nt, from about 3 nt to about 30 nt, from about 3 nt to about 20 nt, from about 3 nt to about 10 nt.
  • nt nucleotides
  • the linker can have a length from about 3 nt to about 5 nt, from about 5 nt to about 10 nt, from about 10 nt to about 15 nt, from about 15 nt to about 20 nt, from about 20 nt to about 25 nt, from about 25 nt to about 30 nt, from about 30 nt to about 35 nt, from about 35 nt to about 40 nt, from about 40 nt to about 50 nt, from about 50 nt to about 60 nt, from about 60 nt to about 70 nt, from about 70 nt to about 80 nt, from about 80 nt to about 90 nt, or from about 90 nt to about 100 nt.
  • the linker of a single-molecule guide nucleic acid can be between 4 and 40 nucleotides.
  • the linker can be at least about 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, or 7000 or more nucleotides.
  • the linker can be at most about 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, or 7000 or more nucleotides.
  • Linkers can comprise any of a variety of sequences, although in some examples the linker will not comprise sequences that have extensive regions of homology with other portions of the guide RNA, which might cause intramolecular binding that could interfere with other functional regions of the guide.
  • a simple 4 nucleotide sequence -GAAA- was used, Science, 337(6096):816-821 (2012), but numerous other sequences, including longer sequences can likewise be used.
  • the linker sequence can comprise a functional moiety.
  • the linker sequence can comprise one or more features, including an aptamer, a ribozyme, a protein- interacting hairpin, a protein binding site, a CRISPR array, an intron, or an exon.
  • the linker sequence can comprise at least about 1, 2, 3, 4, or 5 or more functional moieties. In some examples, the linker sequence can comprise at most about 1, 2, 3, 4, or 5 or more functional moieties.
  • polynucleotides introduced into cells can comprise one or more modifications that can be used individually or in combination, for example, to enhance activity, stability or specificity, alter delivery, reduce innate immune responses in host cells, or for other enhancements, as further described herein and known in the art.
  • modified polynucleotides can be used in the CRISPR/Cas9 or CRISPR/Cpfl system, in which case the guide RNAs (either single-molecule guides or double- molecule guides) and/or a DNA or an RNA encoding a Cas9 or Cpfl endonuclease introduced into a cell can be modified, as described and illustrated below.
  • modified polynucleotides can be used in the CRISPR/Cas9 or CRISPR/Cpfl system to edit any one or more genomic loci.
  • modifications of guide RNAs can be used to enhance the formation or stability of the CRISPR/Cas9 or CRISPR/Cpfl genome editing complex comprising guide RNAs, which can be single-molecule guides or double-molecule, and a Cas9 or Cpfl endonuclease.
  • Modifications of guide RNAs can also or alternatively be used to enhance the initiation, stability or kinetics of interactions between the genome editing complex with the target sequence in the genome, which can be used, for example, to enhance on-target activity.
  • Modifications of guide RNAs can also or alternatively be used to enhance specificity, e.g., the relative rates of genome editing at the on-target site as compared to effects at other (off-target) sites.
  • Modifications can also or alternatively be used to increase the stability of a guide RNA, e.g., by increasing its resistance to degradation by ribonucleases (RNases) present in a cell, thereby causing its half-life in the cell to be increased.
  • RNases ribonucleases
  • Modifications enhancing guide RNA half-life can be particularly useful in aspects in which a Cas9 or Cpfl endonuclease is introduced into the cell to be edited via an RNA that needs to be translated in order to generate
  • RNA interference including small-interfering RNAs (siRNAs), as described below and in the art, tend to be associated with reduced half-life of the RNA and/or the elicitation of cytokines or other factors associated with immune responses.
  • RNAs encoding an endonuclease that are introduced into a cell including, without limitation, modifications that enhance the stability of the RNA (such as by increasing its degradation by RNases present in the cell), modifications that enhance translation of the resulting product (i.e. the endonuclease), and/or modifications that decrease the likelihood or degree to which the RNAs introduced into cells elicit innate immune responses.
  • RNAs used in the CRISPR/Cas9 or CRISPR/Cpfl system can be readily synthesized by chemical means, enabling a number of modifications to be readily incorporated, as illustrated below and described in the art. While chemical synthetic procedures are continually expanding, purifications of such RNAs by procedures such as high-performance liquid chromatography (HPLC, which avoids the use of gels such as PAGE) tends to become more challenging as polynucleotide lengths increase significantly beyond a hundred or so nucleotides.
  • HPLC high-performance liquid chromatography
  • One approach that can be used for generating chemically-modified RNAs of greater length is to produce two or more molecules that are ligated together. Much longer RNAs, such as those encoding a Cas9 endonuclease, are more readily generated enzymatically. While fewer types of modifications are available for use in
  • RNAs there are still modifications that can be used to, e.g., enhance stability, reduce the likelihood or degree of innate immune response, and/or enhance other attributes, as described further below and in the art; and new types of modifications are regularly being developed.
  • modifications can comprise one or more nucleotides modified at the 2' position of the sugar, in some aspects a 2'-0-alkyl, 2'-0-alkyl-0- alkyl, or 2'-fluoro-modified nucleotide.
  • RNA modifications can include 2'- fluoro, 2'-amino or 2'-0-methyl modifications on the ribose of pyrimidines, abasic residues, or an inverted base at the 3' end of the RNA.
  • modified oligonucleotides include those comprising modified backbones, for example, phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages.
  • modified backbones for example, phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages.
  • Some oligonucleotides are oligonucleotides with phosphorothioate backbones and those with heteroatom backbones, particularly CH 2 -NH-0-CH 2 ,
  • CH, ⁇ N(CH 3 ) ⁇ 0 ⁇ CH 2 (known as a methylene(methylimino) or MMI backbone), CH 2 -0-N (CH 3 )-CH 2 , CH 2 -N (CH 3 )-N (CH 3 )-CH 2 and O-N (CH 3 )- CH 2 -CH 2 backbones, wherein the native phosphodiester backbone is represented as O- P- O- CH,); amide backbones [see De Mesmaeker et al., Ace. Chem. Res., 28:366-374 (1995)]; morpholino backbone structures (see Summerton and Weller, U.S. Patent No.
  • PNA peptide nucleic acid
  • Phosphorus-containing linkages include, but are not limited to, phosphorothioates, chiral phosphorothioates,
  • phosphorodithioates phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates comprising 3'alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates comprising 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and
  • boranophosphates having normal 3 -5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to 5'-3' or 2'-5' to 5'-2'; see U.S. Patent Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5, 177, 196; 5, 188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321, 131; 5,399,676; 5,405,939; 5,453,496;
  • Morpholino-based oligomeric compounds are described in Braasch and David Corey, Biochemistry, 41(14): 4503-4510 (2002); Genesis, Volume 30, Issue 3, (2001); Heasman, Dev. Biol., 243 : 209-214 (2002); Nasevicius et al., Nat. Genet., 26:216-220 (2000); Lacerra et al, Proc. Natl. Acad. Sci., 97: 9591-9596 (2000); and U.S. Patent No. 5,034,506, issued Jul. 23, 1991.
  • Cyclohexenyl nucleic acid oligonucleotide mimetics are described in Wang et al., J. Am. Chem. Soc, 122: 8595-8602 (2000).
  • Modified oligonucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
  • These comprise those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S, and CH 2 component parts; see U.S. Patent Nos.
  • One or more substituted sugar moieties can also be included, e.g., one of the following at the 2' position: OH, SH, SCH 3 , F, OCN, OCH 3 OCH 3 , OCH 3 0(CH 2 )n CH 3 , 0(CH 2 )n NH 2 , or 0(CH 2 )n CH 3 , where n is from 1 to about 10; CI to CIO lower alkyl, alkoxyalkoxy, substituted lower alkyl, alkaryl or aralkyl; CI; Br; CN; CF 3 ; OCF 3 ; 0-, S-, or N- alkyl; 0-, S-, or N-alkenyl; SOCH 3 ; S0 2 CH 3 ; ON0 2 ; N0 2 ; N 3 ; NH 2 ; heterocycloalkyl;
  • a modification includes 2'- methoxyethoxy (2'-0-CH 2 CH 2 OCH 3 , also known as 2'-0-(2-methoxyethyl)) (Martin et al, Helv. Chim. Acta, 1995, 78, 486).
  • the base units can be maintained for hybridization with an appropriate nucleic acid target compound.
  • an oligomeric compound an oligonucleotide mimetic that has been shown to have excellent hybridization properties
  • PNA peptide nucleic acid
  • the sugar- backbone of an oligonucleotide can be replaced with an amide containing backbone, for example, an aminoethylglycine backbone.
  • the nucleobases can be retained and bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
  • Representative U.S. patents that teach the preparation of PNA compounds comprise, but are not limited to, U.S. Patent Nos. 5,539,082; 5,714,331; and 5,719,262. Further teaching of PNA compounds can be found in Nielsen et al, Science, 254: 1497-1500 (1991).
  • Guide RNAs can also include, additionally or alternatively, nucleobase (often referred to in the art simply as “base”) modifications or substitutions.
  • nucleobases include adenine (A), guanine (G), thymine (T), cytosine (C), and uracil (U).
  • Modified nucleobases include nucleobases found only infrequently or transiently in natural nucleic acids, e.g., hypoxanthine, 6-methyladenine, 5-Me pyrimidines, particularly 5- methylcytosine (also referred to as 5-methyl-2' deoxycytosine and often referred to in the art as 5-Me-C), 5-hydroxymethylcytosine (HMC), glycosyl HMC and gentobiosyl HMC, as well as synthetic nucleobases, e.g., 2-aminoadenine, 2-(methylamino) adenine, 2- (imidazolylalkyl)adenine, 2-(aminoalklyamino) adenine or other heterosubstituted alkyladenines, 2-thiouracil, 2-thiothymine, 5-bromouracil, 5-hydroxymethyluracil, 8-azaguanine, 7- deazaguanine, N6 (6-aminohexyl)
  • Modified nucleobases can comprise other synthetic and natural nucleobases, such as 5 -methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2- aminoadenine, 6-m ethyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5- halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5- uracil (pseudo-uracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8- thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5- brom
  • nucleobases can comprise those disclosed in U.S. Patent No. 3,687,808, those disclosed in 'The Concise Encyclopedia of Polymer Science And Engineering', pages 858- 859, Kroschwitz, J.I., ed. John Wiley & Sons, 1990, those disclosed by Englisch et al,
  • nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the present disclosure. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, comprising 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine.
  • 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2°C (Sanghvi, Y.S., Crooke, S.T. and Lebleu, B., eds, 'Antisense Research and Applications', CRC Press, Boca Raton, 1993, pp. 276-278) and are aspects of base substitutions, even more particularly when combined with 2'-0-methoxyethyl sugar modifications. Modified nucleobases are described in US Patent Nos.
  • modified refers to a non-natural sugar, phosphate, or base that is incorporated into a guide RNA, an endonuclease, or both a guide RNA and an endonuclease. It is not necessary for all positions in a given oligonucleotide to be uniformly modified, and in fact more than one of the aforementioned modifications can be incorporated in a single
  • oligonucleotide or even in a single nucleoside within an oligonucleotide.
  • the guide RNAs and/or mRNA (or DNA) encoding an endonuclease can be chemically linked to one or more moieties or conjugates that enhance the activity, cellular distribution, or cellular uptake of the oligonucleotide.
  • moieties comprise, but are not limited to, lipid moieties such as a cholesterol moiety [Letsinger et al, Proc. Natl. Acad. Sci. USA, 86: 6553-6556 (1989)]; cholic acid [Manoharan et al, Bioorg. Med. Chem.
  • Sugars and other moieties can be used to target proteins and complexes comprising nucleotides, such as cationic polysomes and liposomes, to particular sites.
  • nucleotides such as cationic polysomes and liposomes
  • hepatic cell directed transfer can be mediated via asialoglycoprotein receptors (ASGPRs); see, e.g., Hu, et al, Protein Pept Lett. 21(10): 1025-30 (2014).
  • ASGPRs asialoglycoprotein receptors
  • Other systems known in the art and regularly developed can be used to target biomolecules of use in the present case and/or complexes thereof to particular target cells of interest.
  • targeting moieties or conjugates can include conjugate groups covalently bound to functional groups, such as primary or secondary hydroxyl groups.
  • Conjugate groups of the present disclosure include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers.
  • Typical conjugate groups include cholesterols, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes.
  • Groups that enhance the pharmacodynamic properties include groups that improve uptake, enhance resistance to degradation, and/or strengthen sequence-specific hybridization with the target nucleic acid.
  • Groups that enhance the pharmacokinetic properties include groups that improve uptake, distribution, metabolism or excretion of the compounds of the present disclosure. Representative conjugate groups are disclosed in International Patent Application No. PCT/US92/09196, filed Oct. 23, 1992 (published as WO1993007883), and U.S. Patent No. 6,287,860.
  • Conjugate moieties include, but are not limited to, lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g., hexyl-5-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues, a phospholipid, e.g., di-hexadecyl-rac- glycerol or triethylammonium 1,2-di-O- hexadecyl-rac-glycero-3-H-phosphonate, a polyamine or a polyethylene glycol chain, or adamantane acetic acid, a palmityl moiety, or an octadecylamine or hexylamino-carbonyl-oxy cholesterol moiety.
  • lipid moieties such as a cholesterol moiety, cholic acid, a thioether
  • Longer polynucleotides that are less amenable to chemical synthesis and are typically produced by enzymatic synthesis can also be modified by various means. Such modifications can include, for example, the introduction of certain nucleotide analogs, the incorporation of particular sequences or other moieties at the 5' or 3' ends of molecules, and other modifications.
  • the mRNA encoding Cas9 is approximately 4 kb in length and can be synthesized by in vitro transcription.
  • Modifications to the mRNA can be applied to, e.g., increase its translation or stability (such as by increasing its resistance to degradation with a cell), or to reduce the tendency of the RNA to elicit an innate immune response that is often observed in cells following introduction of exogenous RNAs, particularly longer RNAs such as that encoding Cas9.
  • TriLink Biotech AxoLabs, Bio-Synthesis Inc., Dharmacon and many others.
  • TriLink for example, 5-Methyl-CTP can be used to impart desirable characteristics, such as increased nuclease stability, increased translation or reduced interaction of innate immune receptors with in vitro transcribed RNA.
  • 5-Methylcytidine-5'-Triphosphate 5-Methyl-CTP
  • N6-Methyl-ATP 5-Methyl-ATP
  • Pseudo-UTP and 2-Thio-UTP have also been shown to reduce innate immune stimulation in culture and in vivo while enhancing translation, as illustrated in publications by Kormann et al. and Warren et al. referred to below.
  • iPSCs induced pluripotency stem cells
  • RNA incorporating 5-Methyl-CTP, Pseudo-UTP and an Anti Reverse Cap Analog (ARCA) could be used to effectively evade the cell's antiviral response; see, e.g., Warren et al., supra.
  • polynucleotides described in the art include, for example, the use of polyA tails, the addition of 5' cap analogs (such as m7G(5')ppp(5')G (mCAP)), modifications of 5' or 3' untranslated regions (UTRs), or treatment with phosphatase to remove 5' terminal phosphates - and new approaches are regularly being developed.
  • 5' cap analogs such as m7G(5')ppp(5')G (mCAP)
  • UTRs untranslated regions
  • treatment with phosphatase to remove 5' terminal phosphates - and new approaches are regularly being developed.
  • RNA interference including small-interfering RNAs (siRNAs).
  • siRNAs present particular challenges in vivo because their effects on gene silencing via mRNA interference are generally transient, which can require repeat administration.
  • siRNAs are double-stranded RNAs (dsRNA) and mammalian cells have immune responses that have evolved to detect and neutralize dsRNA, which is often a by-product of viral infection.
  • dsRNA double-stranded RNAs
  • mammalian cells have immune responses that have evolved to detect and neutralize dsRNA, which is often a by-product of viral infection.
  • PKR dsRNA-responsive kinase
  • RIG-I retinoic acid-inducible gene I
  • TLR3, TLR7 and TLR8 Toll-like receptors
  • RNAs can enhance their delivery and/or uptake by cells, including for example, cholesterol, tocopherol and folic acid, lipids, peptides, polymers, linkers and aptamers; see, e.g., the review by Winkler, Ther. Deliv. 4:791- 809 (2013), and references cited therein.
  • a polynucleotide encoding a site-directed polypeptide can be codon-optimized according to methods standard in the art for expression in the cell containing the target DNA of interest. For example, if the intended target nucleic acid is in a human cell, a human codon- optimized polynucleotide encoding Cas9 is contemplated for use for producing the Cas9 polypeptide.
  • a genome-targeting nucleic acid interacts with a site-directed polypeptide ⁇ e.g., a nucleic acid-guided nuclease such as Cas9), thereby forming a complex.
  • the genome-targeting nucleic acid guides the site-directed polypeptide to a target nucleic acid.
  • RNPs Ribonucleoprotein complexes
  • the site-directed polypeptide and genome-targeting nucleic acid can each be administered separately to a cell or a patient.
  • the site-directed polypeptide can be pre-complexed with one or more guide RNAs, or one or more crRNA together with a tracrRNA.
  • the pre-complexed material can then be administered to a cell or a patient.
  • Such pre-complexed material is known as a ribonucleoprotein particle (RNP).
  • the site-directed polypeptide in the RNP can be, for example, a Cas9 endonuclease or a Cpf 1 endonuclease.
  • the site-directed polypeptide can be flanked at the N-terminus, the C-terminus, or both the N- terminus and C-terminus by one or more nuclear localization signals (NLSs).
  • NLSs nuclear localization signals
  • a Cas9 endonuclease can be flanked by two NLSs, one NLS located at the N-terminus and the second NLS located at the C-terminus.
  • the NLS can be any NLS known in the art, such as a SV40 NLS.
  • the weight ratio of genome-targeting nucleic acid to site-directed polypeptide in the RNP can be 1 : 1.
  • the weight ratio of sgRNA to Cas9 endonuclease in the RNP can be 1 : 1.
  • the present disclosure provides a nucleic acid comprising a nucleotide sequence encoding a genome-targeting nucleic acid of the disclosure, a site-directed polypeptide of the disclosure, and/or any nucleic acid or proteinaceous molecule necessary to carry out the aspects of the methods of the disclosure.
  • the nucleic acid encoding a genome-targeting nucleic acid of the disclosure, a site- directed polypeptide of the disclosure, and/or any nucleic acid or proteinaceous molecule necessary to carry out the aspects of the methods of the disclosure can comprise a vector ⁇ e.g., a recombinant expression vector).
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double-stranded DNA loop into which additional nucleic acid segments can be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional nucleic acid segments can be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced ⁇ e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • Other vectors ⁇ e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • vectors can be capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors", or more simply “expression vectors”, which serve equivalent functions.
  • operably linked means that the nucleotide sequence of interest is linked to regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence.
  • regulatory sequence is intended to include, for example, promoters, enhancers and other expression control elements ⁇ e.g., polyadenylation signals). Such regulatory sequences are well known in the art and are described, for example, in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990).
  • Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cells, and those that direct expression of the nucleotide sequence only in certain host cells ⁇ e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the target cell, the level of expression desired, and the like.
  • Expression vectors contemplated include, but are not limited to, viral vectors based on vaccinia virus, poliovirus, adenovirus, adeno-associated virus, SV40, herpes simplex virus, human immunodeficiency virus, retrovirus (e.g., Murine Leukemia Virus, spleen necrosis virus, and vectors derived from retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, a lentivirus, human immunodeficiency virus, myeloproliferative sarcoma virus, and mammary tumor virus) and other recombinant vectors.
  • retrovirus e.g., Murine Leukemia Virus, spleen necrosis virus, and vectors derived from retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, a lentivirus, human immunodeficiency virus, myeloprolif
  • vectors contemplated for eukaryotic target cells include, but are not limited to, the vectors pXTl, pSG5, pSVK3, pBPV, pMSG, and pSVLSV40 (Pharmacia). Other vectors can be used so long as they are compatible with the host cell.
  • a vector can comprise one or more transcription and/or translation control elements.
  • any of a number of suitable transcription and translation control elements including constitutive and inducible promoters, transcription enhancer elements, transcription terminators, etc. can be used in the expression vector.
  • the vector can be a self-inactivating vector that either inactivates the viral sequences or the components of the CRISPR machinery or other elements.
  • Non-limiting examples of suitable eukaryotic promoters include those from cytomegalovirus (CMV) immediate early, herpes simplex virus (HSV) thymidine kinase, early and late SV40, long terminal repeats (LTRs) from retrovirus, human elongation factor-1 promoter (EF1), a hybrid construct comprising the cytomegalovirus (CMV) enhancer fused to the chicken beta-actin promoter (CAG), murine stem cell virus promoter (MSCV), phosphogly cerate kinase- 1 locus promoter (PGK), and mouse metallothionein-I.
  • CMV cytomegalovirus
  • HSV herpes simplex virus
  • LTRs long terminal repeats
  • EF1 human elongation factor-1 promoter
  • CAG chicken beta-actin promoter
  • MSCV murine stem cell virus promoter
  • PGK phosphogly cerate kinase- 1 locus promoter
  • RNA polymerase III promoters including for example U6 and HI
  • descriptions of and parameters for enhancing the use of such promoters are known in art, and additional information and approaches are regularly being described; see, e.g., Ma, H. et al, Molecular Therapy - Nucleic Acids 3, el61 (2014) doi: 10.1038/mtna.2014.12.
  • the expression vector can also contain a ribosome binding site for translation initiation and a transcription terminator.
  • the expression vector can also comprise appropriate sequences for amplifying expression.
  • the expression vector can also include nucleotide sequences encoding non-native tags (e.g., histidine tag, hemagglutinin tag, green fluorescent protein, etc.) that are fused to the site-directed polypeptide, thus resulting in a fusion protein.
  • a promoter can be an inducible promoter (e.g., a heat shock promoter, tetracycline- regulated promoter, steroid-regulated promoter, metal-regulated promoter, estrogen receptor- regulated promoter, etc.).
  • the promoter can be a constitutive promoter (e.g., CMV promoter, UBC promoter).
  • the promoter can be a spatially restricted and/or temporally restricted promoter (e.g., a tissue specific promoter, a cell type specific promoter, etc.).
  • nucleic acid encoding a genome-targeting nucleic acid of the disclosure and/or a site-directed polypeptide can be packaged into or on the surface of delivery vehicles for delivery to cells.
  • Delivery vehicles contemplated include, but are not limited to, nanospheres, liposomes, quantum dots, nanoparticles, polyethylene glycol particles, hydrogels, and micelles.
  • targeting moieties can be used to enhance the preferential interaction of such vehicles with desired cell types or locations.
  • Introduction of the complexes, polypeptides, and nucleic acids of the disclosure into cells can occur by viral or bacteriophage infection, transfection, conjugation, protoplast fusion, lipofection, electroporation, nucleofection, calcium phosphate precipitation, polyethyleneimine (PEI)-mediated transfection, DEAE-dextran mediated transfection, liposome-mediated transfection, particle gun technology, calcium phosphate precipitation, direct micro-injection, nanoparticle-mediated nucleic acid delivery, and the like.
  • PEI polyethyleneimine
  • SCA3 Spinocerebellar ataxia 3
  • SCA3 is caused by abnormal expansion of a trinucleotide CAG repeat in exon 10 of the ATXN3 gene. In most people, the CAG segment is repeated 12 to 43 times. In patients with Spinocerebellar ataxia 3 (SCA3), the number of CAG repeats can be more than 50. People with intermediate numbers of the CAG repeat (about 44-52) may or may not develop the disease. This number can be different from one patient to another.
  • trinucleotide repeat expansion means a series of three bases (for example, CAG) repeated at least twice.
  • the trinucleotide repeat expansion may be located in exon 10 of an ATXN3 nucleic acid.
  • a pathogenic trinucleotide repeat expansion includes at least 50 repeats of CAG in an ATXN3 nucleic acid and is associated with disease.
  • a pathogenic trinucleotide repeat expansion includes at least 51, 52, 53, 54, 55, 60, 65, 70, 75, 80, 85 or more repeats.
  • the repeats are consecutive.
  • the repeats are interrupted by one or more nucleobases.
  • a wild-type trinucleotide repeat expansion includes 43 or fewer repeats of CAG in an ATXN3 nucleic acid. In other examples, a wild-type trinucleotide repeat expansion includes at most 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 25, 20, 15, or 12 repeats. In certain examples, the entire trinucleotide repeat expansion is deleted. In other examples, a portion of the trinucleotide repeat expansion is deleted.
  • a patient specific induced pluripotent stem cell iPSC
  • the chromosomal DNA of these iPS cells can be edited using the materials and methods described herein.
  • the genome-edited iPSCs can be differentiated into cells which are then implanted into the patient.
  • the iPSCs can be differentiated into cells of the central nervous system (e.g., neurons, glial, or retinal cells), cells of the peripheral nervous system or other periphral cell types, including cells of the cardiovascular system (e.g. : cardiac myocytes).
  • a mesenchymal stem cell can be isolated from the patient, which can be isolated from the patient's bone marrow or peripheral blood.
  • the chromosomal DNA of these mesenchymal stem cells can be edited using the materials and methods described herein.
  • the genome-edited mesenchymal stem cells can be differentiated into cells of the central nervous system (e.g., neurons or glial cells).
  • the differentiated cells e.g., cells of the central nervous system (e.g., neurons or glial cells) are implanted into the patient.
  • Nuclease-based therapeutics can have some level of off-target effects.
  • Performing gene editing ex vivo allows one to characterize the edited cell population prior to implantation.
  • the present disclosure includes sequencing the entire genome of the edited cells to ensure that the off-target effects, if any, can be in genomic locations associated with minimal risk to the patient.
  • populations of specific cells, including clonal populations can be isolated prior to implantation.
  • iPSCs are prolific, making it easy to obtain the large number of cells that will be required for a cell-based therapy.
  • iPSCs are an ideal cell type for performing clonal isolations. This allows screening for the correct genomic modification, without risking a decrease in viability.
  • other primary cells such as glial cells, are viable for only a few passages and difficult to clonally expand.
  • SCA3 Spinocerebellar ataxia 3
  • Methods can also include an in vivo based therapy. Chromosomal DNA of the cells in the patient is edited using the materials and methods described herein.
  • the target cell in an in vivo based therapy is a cell of the central nervous system (e.g., a neuron or a glial cell).
  • Additional promoters are inducible, and therefore can be temporally controlled if the nuclease is delivered as a plasmid.
  • the amount of time that delivered RNA and protein remain in the cell can also be adjusted using treatments or domains added to change the half-life.
  • In vivo treatment would eliminate a number of treatment steps, but a lower rate of delivery can require higher rates of editing.
  • In vivo treatment can eliminate problems and losses from ex vivo treatment, engraftment and post-engraftment integration of neurons and glial cells appropriately into existing brain circuits.
  • An advantage of in vivo gene therapy can be the ease of therapeutic production and administration.
  • the same therapeutic approach and therapy will have the potential to be used to treat more than one patient, for example a number of patients who share the same or similar genotype or allele.
  • ex vivo cell therapy typically requires using a patient's own cells, which are isolated, manipulated and returned to the same patient.
  • a cellular method for editing the ATXN3 gene in a cell by genome editing For example, a cell can be isolated from a patient or animal. Then, the chromosomal DNA of the cell can be edited using the materials and methods described herein.
  • the methods provided herein can involve one or a combination of the following: 1) deleting the entire expanded trinucleotide repeat or a portion thereof in or near the ATXN3 gene, by inducing two double- stranded DNA breaks at both sides of the expanded region; 2) deleting the entire expanded trinucleotide repeat or a portion thereof in or near the ATXN3 gene, by inducing one double- stranded DNA break proximal to the expanded region; 3) replacing the deleted expanded trinucleotide repeat sequence with a corrected sequence; 4) deleting the mutant ATXN3 gene and inserting a wild-type ATXN3 gene, a cDNA or a minigene (comprised of one or more exons and introns or natural or synthetic introns) into the ATXN3 gene locus or a safe harbor locus; or 5) correcting a point mutation within or near the ATXN3
  • the dual DSB-induced deletion strategy can involve excising the entire abnormal repeat expansion or a portion thereof in the ATXN3 gene by inducing two or more double stranded breaks at both sides of the repeat region with one or more CRISPR
  • a donor DNA containing the corrected sequence can be provided to restore the wild-type sequence. This approach can require development and optimization of sgRNAs and donor DNA molecules for the ATXN3 gene.
  • the single DSB-induced deletion strategy can involve deleting the entire abnormal repeat expansion or a portion thereof in the ATXN3 gene by inducing one double stranded break at a site proximal to the repeat region with one or more CRISPR endonucleases and a gRNA (e.g., crRNA + tracrRNA, or sgRNA).
  • a donor DNA containing the corrected sequence can be provided to restore the wild-type sequence. This approach can require development and optimization of gRNAs and donor DNA molecules for the ATXN3 gene.
  • replacing the deleted expanded trinucleotide repeat sequence with a corrected sequence can be achieved by delivering into the cell one or more CRISPR
  • gRNAs e.g., crRNA + tracrRNA, or sgRNA
  • sgRNAs targeting upstream or downstream of the expanded trinucleotide repeat sequence
  • donor DNA that contains the desired sequence and homology arms to the flanking regions of the target locus.
  • the whole gene correction strategy can involve deletion of the endogenous, mutated ATXN3 gene and insertion of a wild-type ATXN3 gene, a cDNA or a minigene (comprised of one or more exons and introns or natural or synthetic introns) into the locus of the ATXN3 gene.
  • this can be achieved by delivering into the cell one or more CRISPR endonucleases, a pair of gRNAs (e.g., crRNA + tracrRNA, or sgRNA) targeting upstream and downstream of or in the first and last exon and/or intron of the ATXN3 gene, and a donor DNA that contains the desired sequence and homology arms to the flanking regions of the target locus.
  • gRNAs e.g., crRNA + tracrRNA, or sgRNA
  • this can be achieved by delivering into the cell one or more CRISPR endonucleases, a pair of gRNAs (e.g., crRNA + tracrRNA, or sgRNA) targeting upstream and downstream of or within one or more exon and/or intron of the ATXN3 gene, and a donor DNA that contains the desired sequence and homology arms to the flanking regions of the target locus.
  • gRNAs e.g., crRNA + tracrRNA, or sgRNA
  • a donor DNA that contains the desired sequence and homology arms to the flanking regions of the target locus.
  • the cytogenetic location of the ATXN3 gene is 14q32.12.
  • the wild-type ATXN3 gene, a cDNA or a minigene can be inserted into a safe harbor locus.
  • a "safe harbor locus” refers to a region of the genome where the integrated material can be adequately expressed without perturbing endogenous gene structure or function.
  • the safe harbor loci include but are not limited to AAVSl (PPP1R12C), ALB, Angpt , ApoC3, ASGR2, CCR5, FIX (F9), Gys2, HGD, Lp(a), Pcsk9, Serpinal, TF, and/or TTR.
  • the target sites within the safe harbor loci can be selected from the group consisting of: exon 1-2 of AAVS l (PPP1R12C), exon 1-2 of ALB, exon 1-2 of AngptB, exon 1-2 of ApoC3, exon 1-2 of ASGR2, exon 1-2 of CCR5, exon 1-2 of FIX (F9), exon 1-2 of Gys2, exon 1-2 of HGD, exon 1-2 of Lp(a), exon 1-2 of Pcsk9, exon 1-2 of Serpinal, exon 1-2 of TF, and/or exon 1-2 of TTR.
  • the whole gene correction strategy utilizes a donor DNA template in Homology- Directed Repair (HDR).
  • HDR may be accomplished by making one or more single-stranded breaks (SSBs) or double-stranded breaks (DSBs) at specific sites in the genome by using one or more endonucleases.
  • the donor DNA can be single or double stranded DNA.
  • the donor template can have homologous arms to the 14q32.12 region.
  • the donor template can have homologous arms to a safe harbor locus.
  • the donor template can have homologous arms to an AAVS l safe harbor locus, such as, intron 1 of the PPP1R12C gene.
  • the donor template can have homologous arms to a CCR5 safe harbor locus.
  • correcting a point mutation can involve replacing one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene.
  • Correcting point mutation can involve deleting the sequence containing the mutation by inducing a double stranded break at a site proximal and a site distal to the point mutation with one or more CRISPR endonucleases and a gRNA (e.g., crRNA + tracrRNA, or sgRNA).
  • a donor DNA containing the corrected sequence can be provided to restore the wild-type sequence. This approach can require development and optimization of gRNAs and donor DNA molecule for the ATXN3 gene.
  • Another strategy involves modulating expression, function, or activity of ATXN3 by editing in the regulatory sequence.
  • Cas9 or similar proteins can be used to target effector domains to the same target sites that can be identified for editing, or additional target sites within range of the effector domain.
  • a range of chromatin modifying enzymes, methylases or demethylases can be used to alter expression of the target gene.
  • One possibility is decreasing the expression of the ATXN3 protein if the mutation leads to undesirable activity.
  • a number of types of genomic target sites can be present in addition to the trinucleotide repeat expansion in the coding sequence of the ATXN3 gene.
  • transcription and translation implicates a number of different classes of sites that interact with cellular proteins or nucleotides. Often the DNA binding sites of transcription factors or other proteins can be targeted for mutation or deletion to study the role of the site, though they can also be targeted to change gene expression. Sites can be added through non-homologous end joining NHEJ or direct genome editing by homology directed repair (HDR). Increased use of genome sequencing, RNA expression and genome-wide studies of transcription factor binding have increased our ability to identify how the sites lead to developmental or temporal gene regulation. These control systems can be direct or can involve extensive cooperative regulation that can require the integration of activities from multiple enhancers. Transcription factors typically bind 6-12 bp-long degenerate DNA sequences.
  • binding sites with less degeneracy can provide simpler means of regulation.
  • Artificial transcription factors can be designed to specify longer sequences that have less similar sequences in the genome and have lower potential for off-target cleavage. Any of these types of binding sites can be mutated, deleted or even created to enable changes in gene regulation or expression (Canver, M.C. et al., Nature (2015)).
  • microRNA Another class of gene regulatory regions having these features is microRNA
  • miRNAs are non-coding RNAs that play key roles in post- transcriptional gene regulation. miRNA can regulate the expression of 30% of all mammalian protein-encoding genes. Specific and potent gene silencing by double stranded RNA (RNAi) was discovered, plus additional small non-coding RNA (Canver, M.C. et al., Nature (2015)). The largest class of non-coding RNAs important for gene silencing are miRNAs. In mammals, miRNAs are first transcribed as a long RNA transcript, which can be separate transcriptional units, part of protein introns, or other transcripts. The long transcripts are called primary miRNA (pri-miRNA) that include imperfectly base-paired hairpin structures.
  • pri-miRNA primary miRNA
  • Pre-miRNAs are short stem loops -70 nucleotides in length with a 2-nucleotide 3'- overhang that are exported, into the mature 19-25 nucleotide miRNA:miRNA* duplexes.
  • the miRNA strand with lower base pairing stability (the guide strand) can be loaded onto the RNA- induced silencing complex (RISC).
  • RISC RNA- induced silencing complex
  • the passenger strand (marked with *), can be functional, but is usually degraded.
  • miRNAs can be important in development, differentiation, cell cycle and growth control, and in virtually all biological pathways in mammals and other multicellular organisms. miRNAs can also be involved in cell cycle control, apoptosis and stem cell differentiation, hematopoiesis, hypoxia, muscle development, neurogenesis, insulin secretion, cholesterol metabolism, aging, viral replication and immune responses.
  • a single miRNA can target hundreds of different mRNA transcripts, while an individual miRNA transcript can be targeted by many different miRNAs. More than 28645 microRNAs have been annotated in the latest release of miRBase (v.21). Some miRNAs can be encoded by multiple loci, some of which can be expressed from tandemly co-transcribed clusters. The features allow for complex regulatory networks with multiple pathways and feedback controls. miRNAs can be integral parts of these feedback and regulatory circuits and can help regulate gene expression by keeping protein production within limits (Herranz, H. & Cohen, S.M. Genes Dev 24, 1339-1344 (2010); Posadas, D M. & Carthew, R.W. Curr Opin Genet Dev 27, 1-6 (2014)).
  • miRNA can also be important in a large number of human diseases that are associated with abnormal miRNA expression. This association underscores the importance of the miRNA regulatory pathway. Recent miRNA deletion studies have linked miRNA with regulation of the immune responses (Stern-Ginossar, N. et al., Science 317, 376-381 (2007)).
  • miRNA also has a strong link to cancer and can play a role in different types of cancer. miRNAs have been found to be downregulated in a number of tumors. miRNA can be important in the regulation of key cancer-related pathways, such as cell cycle control and the DNA damage response, and can therefore be used in diagnosis and can be targeted clinically.
  • MicroRNAs can delicately regulate the balance of angiogenesis, such that experiments depleting all microRNAs suppresses tumor angiogenesis (Chen, S. et al., Genes Dev 28, 1054-1067 (2014)).
  • miRNA genes can also be subject to epigenetic changes occurring with cancer. Many miRNA loci can be associated with CpG islands increasing their opportunity for regulation by DNA methylation (Weber, B., Stresemann, C, Brueckner, B. & Lyko, F. Cell Cycle 6, 1001-1005 (2007)). The majority of studies have used treatment with chromatin remodeling drugs to reveal epigenetically silenced miRNAs.
  • miRNA can also activate translation (Posadas, D M. & Carthew, R.W. Curr Opin Genet Dev 27, 1-6 (2014)). Knocking out miRNA sites may lead to decreased expression of the targeted gene, while introducing these sites may increase expression.
  • miRNA could also be inhibited by specific targeting of the special loop region adjacent to the palindromic sequence.
  • Catalytically inactive Cas9 can also be used to inhibit shRNA expression (Zhao, Y. et al., Sci Rep 4, 3943 (2014)).
  • the binding sites can also be targeted and mutated to prevent the silencing by miRNA.
  • any of the microRNA (miRNA) or their binding sites may be incorporated into the compositions of the invention.
  • compositions may have a region such as, but not limited to, a region comprising the sequence of any of the microRNAs listed in SEQ ID NOs: 632-4715, the reverse complement of the microRNAs listed in SEQ ID NOs: 632-4715, or the microRNA anti-seed region of any of the microRNAs listed in SEQ ID NOs: 632-4715.
  • compositions of the present disclosure may comprise one or more microRNA target sequences, microRNA sequences, or microRNA seeds.
  • Such sequences may correspond to any known microRNA such as those taught in U.S. Publication US2005/0261218 and U.S. Publication US2005/0059005.
  • known microRNAs, their sequences and their binding site sequences in the human genome are listed below in SEQ ID NOs: 632- 4715.
  • a microRNA sequence comprises a "seed" sequence, i.e., a sequence in the region of positions 2-8 of the mature microRNA, which sequence has perfect Watson-Crick
  • a microRNA seed may comprise positions 2-8 or 2-7 of the mature microRNA.
  • a microRNA seed may comprise 7 nucleotides (e.g., nucleotides 2-8 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked by an adenine (A) opposed to microRNA position 1.
  • a microRNA seed may comprise 6 nucleotides (e.g., nucleotides 2-7 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked by an adenine (A) opposed to microRNA position 1.
  • miR-122 a microRNA abundant in liver, can inhibit the expression of the sequence delivered if one or multiple target sites of miR-122 are engineered into the
  • polynucleotide encoding that target sequence Introduction of one or multiple binding sites for different microRNA can be engineered to further decrease the longevity, stability, and protein translation hence providing an additional layer of tenability.
  • microRNA site refers to a microRNA target site or a microRNA recognition site, or any nucleotide sequence to which a microRNA binds or associates. It should be understood that “binding” may follow traditional Watson-Crick hybridization rules or may reflect any stable association of the microRNA with the target sequence at or adjacent to the microRNA site.
  • microRNA binding sites can be engineered out of (i.e. removed from) sequences in which they naturally occur in order to increase protein expression in specific tissues.
  • miR-137 binding sites may be removed to improve protein expression in the brain.
  • microRNAs are known to be differentially expressed in immune cells (also called hematopoietic cells), such as antigen presenting cells (APCs) (e.g. dendritic cells and macrophages), macrophages, monocytes, B lymphocytes, T lymphocytes, granulocytes, natural killer cells, etc.
  • APCs antigen presenting cells
  • microRNAs are involved in immunogenicity
  • Immune cells specific microRNAs also regulate many aspects of development, proliferation, differentiation and apoptosis of hematopoietic cells (immune cells).
  • miR-142 and miR-146 are exclusively expressed in the immune cells, particularly abundant in myeloid dendritic cells.
  • Introducing the miR-142 binding site into the 3'-UTR of a polypeptide of the present disclosure can selectively suppress the gene expression in the antigen presenting cells through miR-142 mediated mRNA degradation, limiting antigen presentation in professional APCs (e.g. dendritic cells) and thereby preventing antigen-mediated immune response after gene delivery (see, Annoni A et al., blood, 2009, 114, 5152-5161).
  • microRNAs binding sites that are known to be expressed in immune cells in particular, the antigen presenting cells, can be engineered into the polynucleotides to suppress the expression of the polynucleotide in APCs through microRNA mediated RNA degradation, subduing the antigen-mediated immune response, while the expression of the polynucleotide is maintained in non-immune cells where the immune cell specific microRNAs are not expressed.
  • microRNA expression studies have been conducted, and are described in the art, to profile the differential expression of microRNAs in various cancer cells /tissues and other diseases. Some microRNAs are abnormally over-expressed in certain cancer cells and others are under-expressed. For example, microRNAs are differentially expressed in cancer cells
  • WO2009/070653, US2010/0323357 cutaneous T-cell lymphoma (WO2013/011378); colorectal cancer cells (WO2011/0281756, WO2011/076142); cancer positive lymph nodes
  • Non-limiting examples of microRNA sequences and the targeted tissues and/or cells are disclosed in SEQ ID NOs: 632-4715. Genome engineering strategies
  • the methods of the present disclosure can involve editing one or both of the mutant alleles.
  • Gene editing to modify or correct the ATXN3 gene has the advantage of restoration of correct expression levels or elimination of aberrant gene products and temporal control.
  • a step of the ex vivo methods of the present disclosure can comprise
  • a step of the in vivo methods of the present disclosure involves editing/correcting the cells in a
  • a step in the cellular methods of the present disclosure can comprise editing/correcting the ATXN3 gene in a human cell by genome engineering.
  • SCA3 Spinocerebellar ataxia 3
  • Any CRISPR endonuclease may be used in the methods of the present disclosure, each CRISPR endonuclease having its own associated PAM, which may or may not be disease specific.
  • the trinucleotide repeat expansion may be excised by inducing two double stranded breaks (DSBs) upstream and downstream of the repeat region. Pairs of gRNAs have been used for this type of deletions.
  • the wild-type sequence can be restored by inserting a correct repeat sequence via HDR.
  • the trinucleotide repeat expansion may be deleted after inducing a single DSB near or within the repeat region.
  • Single DSB-induced repeat loss has been reported in several studies including TALEN-cleaved short CAG/CTG repeats engineered in yeast, ZFN- cleaved CAG repeats engineered in human cells, and CRISPR/Cas9-cleaved CTG/CAG repeats engineered in human cells (Richard et al., PLoS ONE (2014), 9(4): e95611; Mittelman et al., Proc Natl Acad Sci USA (2009), 106(24):9607-12; van Agtmaal et al., Mol Ther (2016), http://dx.doi.Org/10.1016/j .ymthe.2016.10.014).
  • a DSB near the repeat region destabilizes the repeat tracts, triggering a contraction (e.g. partial deletion) or, in some cases, a complete deletion of the repeats. Additionally, work by Mittelman et al. suggested that single DSB-induced repeat loss is more effective in longer repeats.
  • the wild-type sequence can be restored by inserting a correct repeat sequence via HDR.
  • expression of the mutant ATXN3 gene may be disrupted or eliminated by introducing random insertions or deletions (indels) that arise due to the imprecise HEJ repair pathway.
  • the target regions may be the coding sequence of the ATXN3 gene (i.e., exons).
  • Inserting or deleting nucleotides into the coding sequence of a gene may cause a "frame shift" where the normal 3-letter codon pattern is disturbed. In this way, gene expression and therefore mutant protein production can be reduced or eliminated.
  • This approach may also be used to target any intron, intron:exon junction, or regulatory DNA element of the ATXN3 gene where sequence alteration may interfere with the expression of the ATXN3 gene. This approach can require development and optimization of sgRNAs for the ATXN3 gene.
  • the entire mutant gene can be deleted and a wild-type gene, a cDNA or a minigene (comprised of one or more exons and introns or natural or synthetic introns) can be knocked into the gene locus or a heterologous location in the genome such as a safe harbor locus. Pairs of nucleases can be used to delete mutated gene regions, and a donor is provided to restore function. In this case two gRNA and one donor sequence would be supplied. A full length cDNA can be knocked into any "safe harbor", but must use a supplied or an endogenous promoter. If this construct is knocked into the ATXN3 gene locus, it will have physiological control, similar to the normal gene.
  • HDR homology directed repair
  • HR homologous recombination
  • Homology directed repair is a cellular mechanism for repairing double-stranded breaks (DSBs). The most common form is homologous recombination. There are additional pathways for HDR, including single-strand annealing and alternative-HDR. Genome
  • Supplied donors for editing by HDR vary markedly but can contain the intended sequence with small or large flanking homology arms to allow annealing to the genomic DNA.
  • the homology regions flanking the introduced genetic changes can be 30 bp or smaller or as large as a multi-kilobase cassette that can contain promoters, cDNAs, and the like.
  • Both single- stranded and double-stranded oligonucleotide donors have been used. These oligonucleotides range in size from less than 100 nt to over many kb, though longer ssDNA can also be generated and used. Double-stranded donors can be used, including PCR amplicons, plasmids, and mini- circles.
  • an AAV vector can be a very effective means of delivery of a donor template, though the packaging limits for individual donors is ⁇ 5kb. Active transcription of the donor increased HDR three-fold, indicating the inclusion of promoter may increase conversion. Conversely, CpG methylation of the donor decreased gene expression and HDR.
  • nickase variants exist that have one or the other nuclease domain inactivated resulting in cutting of only one DNA strand.
  • HDR can be directed from individual Cas nickases or using pairs of nickases that flank the target area.
  • Donors can be single-stranded, nicked, or dsDNA.
  • the donor DNA can be supplied with the nuclease or independently by a variety of different methods, for example by transfection, nano-particle, micro-injection, or viral transduction.
  • a range of tethering options has been proposed to increase the availability of the donors for HDR. Examples include attaching the donor to the nuclease, attaching to DNA binding proteins that bind nearby, or attaching to proteins that are involved in DNA end binding or repair.
  • the repair pathway choice can be guided by a number of culture conditions, such as those that influence cell cycling, or by targeting of DNA repair and associated proteins.
  • a number of culture conditions such as those that influence cell cycling, or by targeting of DNA repair and associated proteins.
  • key NHEJ molecules can be suppressed, such as KU70, KU80 or DNA ligase IV.
  • the ends from a DNA break or ends from different breaks can be joined using the several non-homologous repair pathways in which the DNA ends are joined with little or no base-pairing at the junction.
  • there are similar repair mechanisms such as alt-NHEJ. If there are two breaks, the intervening segment can be deleted or inverted. NHEJ repair pathways can lead to insertions, deletions or mutations at the joints.
  • NHEJ was used to insert a 15-kb inducible gene expression cassette into a defined locus in human cell lines after nuclease cleavage.
  • a 15-kb inducible gene expression cassette into a defined locus in human cell lines after nuclease cleavage.
  • NHEJ may prove effective for ligation in the intron, while the error-free HDR may be better suited in the coding region.
  • the mutation of the ATXN3 gene that causes Spinocerebellar ataxia 3 is a trinucleotide repeat expansion of the three letter string of nucleotides CAG. In healthy individuals, there are few repeats of this trinucleotide, typically about or fewer than 43. In people with the diseases phenotype, the repeat can occur more than 50 times.
  • One or more trinucleotide repeats may be deleted or corrected in order to restore the gene to a wild-type or similar number of trinucleotide repeats. In some aspects, all of the trinucleotide repeats may be deleted. Alternatively, the mutant gene may be knocked out to eliminate toxic gene products.
  • any one or more of the mutations can be repaired in order to restore the ATXN3.
  • an ATXN3 gene or cDNA can be inserted to the locus of the corresponding gene to replace the mutant gene or knocked-in to a safe harbor site, such as AAVS1.
  • the methods can provide one gRNA or a pair of gRNAs that can be used to facilitate
  • deletions can either be single trinucleotide repeat deletions or multi- trinucleotide repeat deletions. While multi-repeat deletions, including complete deletion of the expanded trinucleotide repeat, can reach a larger number of patients, for larger deletions the efficiency of deletion greatly decreases with increased size.
  • the deletions range from 40 to 1,000 base pairs (bp) in size. For example, deletions may range from 40-100; 100- 200; 200-300; 300-400; 400-500; or 500-1,000 base pairs in size.
  • the methods can provide gRNA pairs that make a deletion by cutting the gene twice, one gRNA cutting at the 5' end of the trinucleotide repeats and the other gRNA cutting at the 3' end of the trinucleotide repeats.
  • the cutting can be accomplished by a pair of DNA endonucleases that each makes a D SB in the genome, or by multiple nickases that together make a D SB in the genome.
  • the methods can provide one gRNA to make one double-strand cut around the trinucleotide repeats.
  • the double-strand cut can be made by a single DNA
  • Illustrative modifications within the ATXN3 gene include deletions within or near (proximal) to the trinucleotide repeats referred to above, such as within the region of less than 3 kb, less than 2kb, less than 1 kb, less than 0.5 kb upstream or downstream of the specific mutation.
  • proximal to the trinucleotide repeats referred to above, such as within the region of less than 3 kb, less than 2kb, less than 1 kb, less than 0.5 kb upstream or downstream of the specific mutation.
  • Such variants can include deletions that are larger in the 5' and/or 3' direction than the specific repeat expansion in question, or smaller in either direction. Accordingly, by “near” or “proximal” with respect to specific repeat expansion, it is intended that the SSB or DSB locus associated with a desired deletion boundary (also referred to herein as an endpoint) can be within a region that is less than about 3 kb from the reference locus noted. The SSB or DSB locus can be more proximal and within 2 kb, within 1 kb, within 0.5 kb, or within 0.1 kb.
  • the desired endpoint can be at or "adjacent to" the reference locus, by which it is intended that the endpoint can be within 100 bp, within 50 bp, within 25 bp, or less than about 10 bp to 5 bp from the reference locus.
  • the surrounding splicing signals can be deleted.
  • Splicing donor and acceptors are generally within 100 base pairs of the neighboring intron. Therefore, in some examples, methods can provide all gRNAs that cut approximately +/- 100-3100 bp with respect to each exon/intron junction of interest.
  • gene editing can be confirmed by sequencing or PCR analysis.
  • Shifts in the location of the 5' boundary and/or the 3' boundary relative to particular reference loci can be used to facilitate or enhance particular applications of gene editing, which depend in part on the endonuclease system selected for the editing, as further described and illustrated herein.
  • many endonuclease systems have rules or criteria that can guide the initial selection of potential target sites for cleavage, such as the requirement of a PAM sequence motif in a particular position adjacent to the DNA cleavage sites in the case of CRISPR Type II or Type V endonucleases.
  • the frequency of off-target activity for a particular combination of target sequence and gene editing endonuclease i.e. the frequency of DSBs occurring at sites other than the selected target sequence
  • the frequency of off-target activity for a particular combination of target sequence and gene editing endonuclease can be assessed relative to the frequency of on-target activity.
  • cells that have been correctly edited at the desired locus can have a selective advantage relative to other cells.
  • a selective advantage include the acquisition of attributes such as enhanced rates of replication, persistence, resistance to certain conditions, enhanced rates of successful engraftment or persistence in vivo following
  • cells that have been correctly edited at the desired locus can be positively selected for by one or more screening methods used to identify, sort or otherwise select for cells that have been correctly edited. Both selective advantage and directed selection methods can take advantage of the phenotype associated with the correction.
  • cells can be edited two or more times in order to create a second modification that creates a new phenotype that is used to select or purify the intended population of cells. Such a second modification could be created by adding a second gRNA enabling expression of a selectable or screenable marker.
  • cells can be correctly edited at the desired locus using a DNA fragment that contains the cDNA and also a selectable marker.
  • target sequence selection can also be guided by consideration of off- target frequencies in order to enhance the effectiveness of the application and/or reduce the potential for undesired alterations at sites other than the desired target.
  • off-target activity can be influenced by a number of factors including similarities and dissimilarities between the target site and various off-target sites, as well as the particular endonuclease used.
  • Bioinformatics tools are available that assist in the prediction of off-target activity, and frequently such tools can also be used to identify the most likely sites of off-target activity, which can then be assessed in experimental settings to evaluate relative frequencies of off-target to on-target activity, thereby allowing the selection of sequences that have higher relative on-target activities. Illustrative examples of such techniques are provided herein, and others are known in the art.
  • Another aspect of target sequence selection relates to homologous recombination events. Sequences sharing regions of homology can serve as focal points for homologous recombination events that result in deletion of intervening sequences. Such recombination events occur during the normal course of replication of chromosomes and other DNA sequences, and also at other times when DNA sequences are being synthesized, such as in the case of repairs of double-strand breaks (DSBs), which occur on a regular basis during the normal cell replication cycle but can also be enhanced by the occurrence of various events (such as UV light and other inducers of DNA breakage) or the presence of certain agents (such as various chemical inducers).
  • various events such as UV light and other inducers of DNA breakage
  • certain agents such as various chemical inducers
  • DSBs can be regularly induced and repaired in normal cells.
  • indels small insertions or deletions
  • DSBs can also be specifically induced at particular locations, as in the case of the endonucleases systems described herein, which can be used to cause directed or preferential gene modification events at selected chromosomal locations.
  • the tendency for homologous sequences to be subject to recombination in the context of DNA repair (as well as replication) can be taken advantage of in a number of circumstances, and is the basis for one application of gene editing systems, such as CRISPR, in which homology directed repair is used to insert a sequence of interest, provided through use of a "donor" polynucleotide, into a desired
  • Regions of homology between particular sequences which can be small regions of "microhomology” that can comprise as few as ten base pairs or less, can also be used to bring about desired deletions.
  • a single DSB can be introduced at a site that exhibits microhomology with a nearby sequence.
  • a result that occurs with high frequency is the deletion of the intervening sequence as a result of recombination being facilitated by the DSB and concomitant cellular repair process.
  • target sequences within regions of homology can also give rise to much larger deletions, including gene fusions (when the deletions are in coding regions), which may or may not be desired given the particular circumstances.
  • the examples provided herein further illustrate the selection of various target regions for the creation of DSBs designed to induce deletions or replacements that result in restoration of wild-type or similar levels of trinucleotide repeats in the ATXN3 gene, as well as the selection of specific target sequences within such regions that are designed to minimize off-target events relative to on-target events.
  • the principal targets for gene editing are human cells.
  • the human cells can be somatic cells, which after being modified using the techniques as described, can give rise to differentiated cells, e.g., neurons or progenitor cells.
  • the human cells may be cells from the central nervous system or cells from other affected organs.
  • Stem cells are capable of both proliferation and giving rise to more progenitor cells, these in turn having the ability to generate a large number of mother cells that can in turn give rise to differentiated or differentiable daughter cells.
  • the daughter cells themselves can be induced to proliferate and produce progeny that subsequently differentiate into one or more mature cell types, while also retaining one or more cells with parental developmental potential.
  • stem cell refers then, to a cell with the capacity or potential, under particular circumstances, to differentiate to a more specialized or differentiated phenotype, and which retains the capacity, under certain circumstances, to proliferate without substantially
  • progenitor or stem cell refers to a generalized mother cell whose descendants (progeny) specialize, often in different directions, by differentiation, e.g., by acquiring completely individual characters, as occurs in progressive diversification of embryonic cells and tissues.
  • Cellular differentiation is a complex process typically occurring through many cell divisions.
  • a differentiated cell may derive from a multipotent cell that, itself, is derived from a multipotent cell, and so on. While each of these multipotent cells may be considered stem cells, the range of cell types that each can give rise to may vary considerably.
  • Some differentiated cells also have the capacity to give rise to cells of greater developmental potential. Such capacity may be natural or may be induced artificially upon treatment with various factors.
  • stem cells can also be "multipotent" because they can produce progeny of more than one distinct cell type, but this is not required for "stem-ness.”
  • Self-renewal can be another important aspect of the stem cell.
  • self-renewal can occur by either of two major mechanisms.
  • Stem cells can divide asymmetrically, with one daughter retaining the stem state and the other daughter expressing some distinct other specific function and phenotype.
  • some of the stem cells in a population can divide symmetrically into two stems, thus maintaining some stem cells in the population as a whole, while other cells in the population give rise to differentiated progeny only.
  • stem cells can divide asymmetrically, with one daughter retaining the stem state and the other daughter expressing some distinct other specific function and phenotype.
  • some of the stem cells in a population can divide symmetrically into two stems, thus maintaining some stem cells in the population as a whole, while other cells in the population give rise to differentiated progeny only.
  • stem cells can divide asymmetrically, with one daughter retaining the stem state and the other daughter expressing some distinct other specific function and phenotype.
  • some of the stem cells in a population can divide
  • progenitor cells have a cellular phenotype that is more primitive (i.e., is at an earlier step along a developmental pathway or progression than is a fully differentiated cell). Often, progenitor cells also have significant or very high-proliferative potential. Progenitor cells can give rise to multiple distinct differentiated cell types or to a single differentiated cell type, depending on the developmental pathway and on the environment in which the cells develop and differentiate.
  • differentiated In the context of cell ontogeny, the adjective “differentiated,” or “differentiating” is a relative term.
  • a “differentiated cell” is a cell that has progressed further down the developmental pathway than the cell to which it is being compared.
  • stem cells can differentiate into lineage-restricted precursor cells (such as a neural progenitor cell), which in turn can
  • the genetically engineered human cells described herein can be induced pluripotent stem cells (iPSCs).
  • iPSCs induced pluripotent stem cells
  • An advantage of using iPSCs is that the cells can be derived from the same subject to which the progenitor cells are to be administered. That is, a somatic cell can be obtained from a subject, reprogrammed to an induced pluripotent stem cell, and then re- differentiated into a progenitor cell to be administered to the subject ⁇ e.g., autologous cells). Because the progenitors are essentially derived from an autologous source, the risk of engraftment rejection or allergic response can be reduced compared to the use of cells from another subject or group of subjects.
  • the use of iPSCs negates the need for cells obtained from an embryonic source.
  • the stem cells used in the disclosed methods are not embryonic stem cells.
  • reprogramming refers to a process that alters or reverses the
  • differentiation state of a differentiated cell (e.g., a somatic cell).
  • reprogramming refers to a process of driving the differentiation of a cell backwards to a more undifferentiated or more primitive type of cell.
  • placing many primary cells in culture can lead to some loss of fully differentiated characteristics.
  • simply culturing such cells included in the term differentiated cells does not render these cells non- differentiated cells (e.g., undifferentiated cells) or pluripotent cells.
  • the transition of a differentiated cell to pluripotency requires a reprogramming stimulus beyond the stimuli that lead to partial loss of differentiated character in culture.
  • Reprogrammed cells also have the characteristic of the capacity of extended passaging without loss of growth potential, relative to primary cell parents, which generally have capacity for only a limited number of divisions in culture.
  • the cell to be reprogrammed can be either partially or terminally differentiated prior to reprogramming.
  • Reprogramming can encompass complete reversion of the differentiation state of a differentiated cell (e.g., a somatic cell) to a pluripotent state or a multipotent state.
  • Reprogramming can encompass complete or partial reversion of the differentiation state of a differentiated cell (e.g., a somatic cell) to an undifferentiated cell (e.g., an embryonic-like cell).
  • Reprogramming can result in expression of particular genes by the cells, the expression of which further contributes to reprogramming.
  • reprogramming of a differentiated cell e.g., a somatic cell
  • undifferentiated state e.g., is an undifferentiated cell
  • the resulting cells are referred to as "reprogrammed cells,” or “induced pluripotent stem cells (iPSCs or iPS cells).”
  • Reprogramming can involve alteration, e.g., reversal, of at least some of the heritable patterns of nucleic acid modification (e.g., methylation), chromatin condensation, epigenetic changes, genomic imprinting, etc., that occur during cellular differentiation.
  • Reprogramming is distinct from simply maintaining the existing undifferentiated state of a cell that is already pluripotent or maintaining the existing less than fully differentiated state of a cell that is already a multipotent cell (e.g., a myogenic stem cell).
  • Reprogramming is also distinct from promoting the self-renewal or proliferation of cells that are already pluripotent or multipotent, although the compositions and methods described herein can also be of use for such purposes, in some examples.
  • Mouse somatic cells can be converted to ES cell-like cells with expanded developmental potential by the direct transduction of Oct4, Sox2, Klf4, and c-Myc; see, e.g., Takahashi and Yamanaka, Cell 126(4): 663-76 (2006).
  • iPSCs resemble ES cells, as they restore the pluripotency-associated transcriptional circuitry and much of the epigenetic landscape.
  • mouse iPSCs satisfy all the standard assays for pluripotency: specifically, in vitro differentiation into cell types of the three germ layers, teratoma formation, contribution to chimeras, germline transmission [see, e.g., Maherali and Hochedlinger, Cell Stem Cell. 3(6):595-605 (2008)], and tetraploid complementation.
  • iPSCs can be obtained using similar transduction methods, and the transcription factor trio, OCT4, SOX2, and NANOG, has been established as the core set of transcription factors that govern pluripotency; see, e.g., Budniatzky and Gepstein, Stem Cells Transl Med. 3(4):448-57 (2014); Barrett et al, Stem Cells Trans Med 3 : 1-6 sctm.2014-0121 (2014); Focosi et al., Blood Cancer Journal 4: e211 (2014); and references cited therein.
  • the production of iPSCs can be achieved by the introduction of nucleic acid sequences encoding stem cell-associated genes into an adult, somatic cell, historically using viral vectors.
  • iPSCs can be generated or derived from terminally differentiated somatic cells, as well as from adult stem cells, or somatic stem cells. That is, a non-pluripotent progenitor cell can be rendered pluripotent or multipotent by reprogramming. In such instances, it may not be necessary to include as many reprogramming factors as required to reprogram a terminally differentiated cell.
  • reprogramming can be induced by the non-viral introduction of reprogramming factors, e.g., by introducing the proteins themselves, or by introducing nucleic acids that encode the reprogramming factors, or by introducing messenger RNAs that upon translation produce the reprogramming factors (see e.g., Warren et al., Cell Stem Cell, 7(5):618- 30 (2010).
  • Reprogramming can be achieved by introducing a combination of nucleic acids encoding stem cell-associated genes, including, for example, Oct-4 (also known as Oct-3/4 or Pouf51), Soxl, Sox2, Sox3, Sox 15, Sox 18, NANOG, KM, Klf2, Klf4, Klf5, NR5A2, c-Myc, 1- Myc, n-Myc, Rem2, Tert, and LIN28.
  • Reprogramming using the methods and compositions described herein can further comprise introducing one or more of Oct-3/4, a member of the Sox family, a member of the Klf family, and a member of the Myc family to a somatic cell.
  • the methods and compositions described herein can further comprise introducing one or more of each of Oct-4, Sox2, Nanog, c-MYC and Klf4 for reprogramming.
  • the exact method used for reprogramming is not necessarily critical to the methods and compositions described herein.
  • the reprogramming is not effected by a method that alters the genome.
  • reprogramming can be achieved, e.g., without the use of viral or plasmid vectors.
  • the efficiency of reprogramming i.e., the number of reprogrammed cells derived from a population of starting cells can be enhanced by the addition of various agents, e.g., small molecules, as shown by Shi et al, Cell-Stem Cell 2:525-528 (2008); Huangfu et al, Nature Biotechnology 26(7):795-797 (2008) and Marson et al, Cell-Stem Cell 3 : 132-135 (2008).
  • various agents e.g., small molecules, as shown by Shi et al, Cell-Stem Cell 2:525-528 (2008); Huangfu et al, Nature Biotechnology 26(7):795-797 (2008) and Marson et al, Cell-Stem Cell 3 : 132-135 (2008).
  • agents that enhance the efficiency or rate of induced pluripotent stem cell production can be used in the production of patient-specific or disease- specific iPSCs.
  • agents that enhance reprogramming efficiency include soluble Wnt, Wnt conditioned media, BIX-01294 (a G9a histone methyltransferase), PD0325901 (a MEK inhibitor), DNA methyltransferase inhibitors, histone deacetylase (HDAC) inhibitors, valproic acid, 5'-azacytidine, dexamethasone, suberoylanilide, hydroxamic acid (SAHA), vitamin C, and trichostatin (TSA), among others.
  • reprogramming enhancing agents include:
  • SAHA Suberoylanilide Hydroxamic Acid
  • BML-210 Depudecin (e.g., (-)-Depudecin)
  • HC Toxin Nullscript (4-(l,3-Dioxo-lH,3H- benzo[de]isoquinolin-2-yl)-N-hydroxybutanamide), Phenylbutyrate (e.g., sodium
  • Valproic Acid (VP A) and other short chain fatty acids
  • Scriptaid Suramin Sodium, Trichostatin A (TSA), APHA Compound 8, Apicidin, Sodium Butyrate,
  • pivaloyloxymethyl butyrate Pivanex, AN-9
  • Trapoxin B Chlamydocin
  • Depsipeptide also known as FR901228 or FK2248
  • benzamides e.g., CI-994 (e.g., N-acetyl dinaline) and MS-27- 275)
  • MGCD0103 NVP-LAQ-824
  • CBHA m-carboxycinnaminic acid bishydroxamic acid
  • JNJ16241199 Tubacin
  • 3-Cl-UCHA e.g., 6-(3- chlorophenylureido
  • AOE 2-amino-8-oxo-9, 10-epoxydecanoic acid
  • CHAP31 and CHAP 50 CHAP31 and CHAP 50.
  • reprogramming enhancing agents include, for example, dominant negative forms of the HDACs (e.g., catalytically inactive forms), siRNA inhibitors of the HDACs, and antibodies that specifically bind to the HDACs.
  • HDACs e.g., catalytically inactive forms
  • siRNA inhibitors of the HDACs e.g., siRNA inhibitors of the HDACs
  • antibodies that specifically bind to the HDACs are available, e.g., from BIOMOL International, Fukasawa, Merck Biosciences, Novartis, Gloucester
  • isolated clones can be tested for the expression of a stem cell marker.
  • a stem cell marker can be selected from the non-limiting group including SSEA3, SSEA4, CD9, Nanog, Fbxl5, Ecatl, Esgl, Eras, Gdf3, Fgf4, Cripto, Daxl, Zpf296, Slc2a3, Rexl, Utfl, and Natl.
  • a cell that expresses Oct4 or Nanog is identified as pluripotent.
  • Methods for detecting the expression of such markers can include, for example, RT-PCR and immunological methods that detect the presence of the encoded polypeptides, such as Western blots or flow cytometric analyses. Detection can involve not only RT-PCR, but can also include detection of protein markers. Intracellular markers may be best identified via RT-PCR, or protein detection methods such as immunocytochemistry, while cell surface markers are readily identified, e.g., by immunocytochemi stry .
  • the pluripotent stem cell character of isolated cells can be confirmed by tests evaluating the ability of the iPSCs to differentiate into cells of each of the three germ layers.
  • teratoma formation in nude mice can be used to evaluate the pluripotent character of the isolated clones.
  • the cells can be introduced into nude mice and histology and/or immunohistochemistry can be performed on a tumor arising from the cells.
  • the growth of a tumor comprising cells from all three germ layers, for example, further indicates that the cells are pluripotent stem cells.
  • the genetically engineered human cells described herein are cells of the central nervous system.
  • Neurons which process information
  • glial cells or neuroglia
  • neuroglia which provide mechanical and metabolic support to the nervous system and modulate information processed by neurons
  • Non-limiting examples of neurons include sensory neurons (also referred to as afferent neurons) transfer information from the external environment to the central nervous system, motor neurons (also referred to as efferent neurons) transfer information from the central nervous system to the external environment, and interneurons (also referred to as association neurons) process information in the central nervous system and transfers the information from one neuron to the other within the central nervous system.
  • glial cells include astrocytes, oligodendrocytes, microglia and Schwann cells.
  • CNS progenitor cells can be neural progenitor cells or glial progenitor cells.
  • the typical neuron transmits electrical signals from one cell to another.
  • Neurons contain a cell body, dendrite, axon hillock, axon, nerve endings, neuronal synapases and neuromuscular junctions. Neurons may be named according to shape or the nature of the dendritic tree.
  • Neuroglia differ from neurons in several general ways in that they: do not form synapses, have essentially only one type of process, retain the ability to divide, and are less electrically excitable than neurons.
  • Neuroglia are classified based on size and shape of their nucleus and distinguished from neurons, at the light microscopic level. Neuroglia are divided into two major categories based on size, the macroglia and the microglia.
  • the macroglia are of ectodermal origin and consist of astrocytes, oligodendrocytes and ependymal cells. Microglia cells are probably of mesodermal origin.
  • One step of the ex vivo methods of the present disclosure can involve creating a patient specific iPS cell, patient specific iPS cells, or a patient specific iPS cell line.
  • the creating step can comprise: a) isolating a somatic cell, such as a skin cell or fibroblast, from the patient; and b) introducing a set of pluripotency-associated genes into the somatic cell in order to induce the cell to become a pluripotent stem cell.
  • the set of pluripotency-associated genes can be one or more of the genes selected from the group consisting of OCT4, SOXl, SOX2, SOX3, SOX15, SOX18, NANOG, KLF1, KLF2, KLF4, KLF5, c-MYC, n-MYC, REM2, TERT and LIN28.
  • a biopsy or aspirate is a sample of tissue or fluid taken from the body.
  • biopsies or aspirates There are many different kinds of biopsies or aspirates. Nearly all of them involve using a sharp tool to remove a small amount of tissue. If the biopsy will be on the skin or other sensitive area, numbing medicine can be applied first.
  • a biopsy or aspirate may be performed according to any of the known methods in the art. For example, in a bone marrow aspirate, a large needle is used to enter the pelvis bone to collect bone marrow.
  • Mesenchymal stem cells can be isolated according to any method known in the art, such as from a patient's bone marrow or peripheral blood. For example, marrow aspirate can be collected into a syringe with heparin. Cells can be washed and centrifuged on a Percoll. The cells can be cultured in Dulbecco's modified Eagle's medium (DMEM) (low glucose) containing 10% fetal bovine serum (FBS) (Pittinger MF, Mackay AM, Beck SC et al, Science 1999;
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • genetically modified cell refers to a cell that comprises at least one genetic modification introduced by genome editing ⁇ e.g., using the CRISPR/Cas9 or
  • the genetically modified cell can be genetically modified neural progenitor cell.
  • the genetically modified cell can be a genetically modified cell of the central nervous system (e.g., a neuron or a glial cell).
  • a genetically modified cell comprising an exogenous genome-targeting nucleic acid and/or an exogenous nucleic acid encoding a genome-targeting nucleic acid is contemplated herein.
  • control treated population describes a population of cells that has been treated with identical media, viral induction, nucleic acid sequences, temperature, confluency, flask size, pH, etc., with the exception of the addition of the genome editing components. Any method known in the art can be used to measure restoration of ATXN3 gene or protein expression or activity, for example Western Blot analysis of the ATXN3 protein or real time PCR quantifying ATXN3 mRNA.
  • isolated cell refers to a cell that has been removed from an organism in which it was originally found, or a descendant of such a cell.
  • the cell can be cultured in vitro, e.g., under defined conditions or in the presence of other cells.
  • the cell can be later introduced into a second organism or re-introduced into the organism from which it (or the cell from which it is descended) was isolated.
  • isolated population refers to a population of cells that has been removed and separated from a mixed or heterogeneous population of cells.
  • the isolated population can be a substantially pure population of cells, as compared to the heterogeneous population from which the cells were isolated or enriched.
  • the isolated population can be an isolated population of human progenitor cells, e.g., a substantially pure population of human progenitor cells, as compared to a heterogeneous population of cells comprising human progenitor cells and cells from which the human progenitor cells were derived.
  • the term "substantially enhanced,” with respect to a particular cell population, refers to a population of cells in which the occurrence of a particular type of cell is increased relative to pre-existing or reference levels, by at least 2-fold, at least 3-, at least 4-, at least 5-, at least 6-, at least 7-, at least 8-, at least 9, at least 10-, at least 20-, at least 50-, at least 100-, at least 400-, at least 1000-, at least 5000-, at least 20000-, at least 100000- or more fold depending, e.g., on the desired levels of such cells for ameliorating Spinocerebellar ataxia 3 (SCA3).
  • SCA3 Spinocerebellar ataxia 3
  • substantially enriched with respect to a particular cell population, refers to a population of cells that is at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%), about 70% or more with respect to the cells making up a total cell population.
  • substantially pure with respect to a particular cell population, refers to a population of cells that is at least about 75%, at least about 85%, at least about 90%, or at least about 95%) pure, with respect to the cells making up a total cell population. That is, the terms “substantially pure” or “essentially purified,” with regard to a population of progenitor cells, refers to a population of cells that contain fewer than about 20%, about 15%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1%, or less than 1%), of cells that are not progenitor cells as defined by the terms herein.
  • Another step of the ex vivo methods of the present disclosure can comprise differentiating the genome-edited iPSCs into cells of the central nervous system (CNS) (e.g., neurons or glial cells).
  • the differentiating step may be performed according to any method known in the art.
  • Another step of the ex vivo methods of the present disclosure can comprise differentiating the genome-edited mesenchymal stem cells into cells of the central nervous system (CNS).
  • the differentiating step may be performed according to any method known in the art.
  • Another step of the ex vivo methods of the present disclosure involves implanting the cells of the central nervous system (e.g., neurons or glial cells) into patients.
  • This implanting step may be accomplished using any method of implantation known in the art.
  • the genetically modified neurons may be administered to the patient via intraparenchymal, vascular (e.g., intravenous, intra-arterial), or ventricular (e.g., intracerebroventricular, intracisternal, intrathecal) routes or other routes such as intracranial or intraperitoneal injection.
  • ex vivo methods of administering progenitor cells to a subject contemplated herein involve the use of therapeutic compositions comprising progenitor cells.
  • Therapeutic compositions can contain a physiologically tolerable carrier together with the cell composition, and optionally at least one additional bioactive agent as described herein, dissolved or dispersed therein as an active ingredient.
  • the therapeutically tolerable carrier can contain a physiologically tolerable carrier together with the cell composition, and optionally at least one additional bioactive agent as described herein, dissolved or dispersed therein as an active ingredient.
  • the therapeutically tolerable carrier can contain a physiologically tolerable carrier together with the cell composition, and optionally at least one additional bioactive agent as described herein, dissolved or dispersed therein as an active ingredient.
  • the therapeutic agents as described herein, dissolved or dispersed therein as an active ingredient.
  • composition is not substantially immunogenic when administered to a mammal or human patient for therapeutic purposes, unless so desired.
  • the progenitor cells described herein can be administered as a suspension with a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier to be used in a cell composition will not include buffers, compounds, cryopreservation agents, preservatives, or other agents in amounts that substantially interfere with the viability of the cells to be delivered to the subject.
  • a formulation comprising cells can include e.g., osmotic buffers that permit cell membrane integrity to be maintained, and optionally, nutrients to maintain cell viability or enhance engraftment upon administration.
  • Such formulations and suspensions are known to those of skill in the art and/or can be adapted for use with the progenitor cells, as described herein, using routine experimentation.
  • a cell composition can also be emulsified or presented as a liposome composition, provided that the emulsification procedure does not adversely affect cell viability.
  • the cells and any other active ingredient can be mixed with excipients that are pharmaceutically acceptable and compatible with the active ingredient, and in amounts suitable for use in the therapeutic methods described herein.
  • Additional agents included in a cell composition can include pharmaceutically acceptable salts of the components therein.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide) that are formed with inorganic acids, such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, tartaric, mandelic and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases, such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like.
  • Physiologically tolerable carriers are well known in the art.
  • Exemplary liquid carriers are sterile aqueous solutions that contain no materials in addition to the active ingredients and water, or contain a buffer such as sodium phosphate at physiological pH value, physiological saline or both, such as phosphate-buffered saline.
  • aqueous carriers can contain more than one buffer salt, as well as salts such as sodium and potassium chlorides, dextrose, polyethylene glycol and other solutes.
  • Liquid compositions can also contain liquid phases in addition to and to the exclusion of water. Exemplary of such additional liquid phases are glycerin, vegetable oils such as cottonseed oil, and water-oil emulsions.
  • the amount of an active compound used in the cell compositions that is effective in the treatment of a particular disorder or condition can depend on the nature of the disorder or condition, and can be determined by standard clinical techniques.
  • Guide RNAs of the present disclosure can be formulated with pharmaceutically acceptable excipients such as carriers, solvents, stabilizers, adjuvants, diluents, etc., depending upon the particular mode of administration and dosage form.
  • Guide RNA compositions can be formulated to achieve a physiologically compatible pH, and range from a pH of about 3 to a pH of about 11, about pH 3 to about pH 7, depending on the formulation and route of administration.
  • the pH can be adjusted to a range from about pH 5.0 to about pH 8.
  • the compositions can comprise a therapeutically effective amount of at least one compound as described herein, together with one or more pharmaceutically acceptable excipients.
  • compositions can comprise a combination of the compounds described herein, or can include a second active ingredient useful in the treatment or prevention of bacterial growth (for example and without limitation, anti -bacterial or anti -microbial agents), or can include a combination of reagents of the present disclosure.
  • Suitable excipients include, for example, carrier molecules that include large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles.
  • Other exemplary excipients can include antioxidants (for example and without limitation, ascorbic acid), chelating agents (for example and without limitation, EDTA), carbohydrates (for example and without limitation, dextrin, hydroxyalkylcellulose, and hydroxyalkylmethylcellulose), stearic acid, liquids (for example and without limitation, oils, water, saline, glycerol and ethanol), wetting or emulsifying agents, pH buffering substances, and the like.
  • RNA polynucleotides RNA or DNA
  • endonuclease polynucleotide(s) RNA or DNA
  • viral or non-viral delivery vehicles known in the art.
  • endonuclease polypeptide(s) can be delivered by viral or non-viral delivery vehicles known in the art, such as electroporation or lipid nanoparticles.
  • the DNA endonuclease can be delivered as one or more polypeptides, either alone or pre-complexed with one or more guide RNAs, or one or more crRNA together with a tracrRNA.
  • Polynucleotides can be delivered by non-viral delivery vehicles including, but not limited to, nanoparticles, liposomes, ribonucleoproteins, positively charged peptides, small molecule RNA-conjugates, aptamer-RNA chimeras, and RNA-fusion protein complexes.
  • non-viral delivery vehicles including, but not limited to, nanoparticles, liposomes, ribonucleoproteins, positively charged peptides, small molecule RNA-conjugates, aptamer-RNA chimeras, and RNA-fusion protein complexes.
  • the formulation may be selected from any of those taught, for example, in International Application PCT7US2012/069610.
  • Polynucleotides such as guide RNA, sgRNA, and mRNA encoding an endonuclease, may be delivered to a cell or a patient by a lipid nanoparticle (LNP).
  • LNP lipid nanoparticle
  • a LNP refers to any particle having a diameter of less than 1000 nm, 500 nm, 250 nm, 200 nm, 150 nm, 100 nm, 75 nm, 50 nm, or 25 nm.
  • a nanoparticle may range in size from 1-1000 nm, 1-500 nm, 1-250 nm, 25-200 nm, 25-100 nm, 35-75 nm, or 25-60 nm.
  • LNPs may be made from cationic, anionic, or neutral lipids.
  • Neutral lipids such as the fusogenic phospholipid DOPE or the membrane component cholesterol, may be included in LNPs as 'helper lipids' to enhance transfection activity and nanoparticle stability.
  • Limitations of cationic lipids include low efficacy owing to poor stability and rapid clearance, as well as the generation of inflammatory or anti-inflammatory responses.
  • LNPs may also be comprised of hydrophobic lipids, hydrophilic lipids, or both hydrophobic and hydrophilic lipids.
  • lipids used to produce LNPs are: DOTMA, DOSPA, DOTAP, DMRIE, DC- cholesterol, DOTAP-cholesterol, GAP-DMORIE-DPyPE, and GL67A-DOPE-DMPE- polyethylene glycol (PEG).
  • cationic lipids are: 98N12-5, C 12-200, DLin-KC2- DMA (KC2), DLin-MC3 -DMA (MC3), XTC, MD1, and 7C1.
  • neutral lipids are: DPSC, DPPC, POPC, DOPE, and SM.
  • PEG-modified lipids are: PEG-DMG, PEG- CerC14, and PEG-CerC20.
  • the lipids can be combined in any number of molar ratios to produce a L P.
  • the polynucleotide(s) can be combined with lipid(s) in a wide range of molar ratios to produce a LNP.
  • the site-directed polypeptide and genome-targeting nucleic acid can each be administered separately to a cell or a patient.
  • the site-directed polypeptide can be pre-complexed with one or more guide RNAs, or one or more crRNA together with a tracrRNA.
  • the pre-complexed material can then be administered to a cell or a patient.
  • Such pre-complexed material is known as a ribonucleoprotein particle (RNP).
  • RNA is capable of forming specific interactions with RNA or DNA. While this property is exploited in many biological processes, it also comes with the risk of promiscuous interactions in a nucleic acid-rich cellular environment.
  • One solution to this problem is the formation of ribonucleoprotein particles (RNPs), in which the RNA is pre-complexed with an endonuclease.
  • RNPs ribonucleoprotein particles
  • Another benefit of the RNP is protection of the RNA from degradation.
  • the endonuclease in the RNP can be modified or unmodified.
  • the gRNA, crRNA, tracrRNA, or sgRNA can be modified or unmodified. Numerous modifications are known in the art and can be used.
  • the endonuclease and sgRNA can be generally combined in a 1 : 1 molar ratio.
  • the endonuclease, crRNA and tracrRNA can be generally combined in a 1 : 1 : 1 molar ratio.
  • a wide range of molar ratios can be used to produce a RNP.
  • a recombinant adeno-associated virus (AAV) vector can be used for delivery.
  • rAAV particles in which an AAV genome to be packaged that includes the polynucleotide to be delivered, rep and cap genes, and helper virus functions are provided to a cell are standard in the art. Production of rAAV typically requires that the following components are present within a single cell (denoted herein as a packaging cell): a rAAV genome, AAV rep and cap genes separate from ⁇ i.e., not in) the rAAV genome, and helper virus functions.
  • the AAV rep and cap genes may be from any AAV serotype for which recombinant virus can be derived, and may be from a different AAV serotype than the rAAV genome ITRs, including, but not limited to, AAV serotypes described herein. Production of pseudotyped rAAV is disclosed in, for example, international patent application publication number WO
  • AAV particles packaging polynucleotides encoding compositions of the present disclosure may comprise or be derived from any natural or recombinant AAV serotype.
  • the AAV particles may utilize or be based on a serotype selected from any of the following serotypes, and variants thereof including but not limited to AAV1, AAV10, AAV106.1/hu.37, AAV11, AAV114.3/hu.40, AAV 12, AAV127.2/hu.41, AAV127.5/hu.42, AAV128.1/hu.43, AAV128.3/hu.44, AAV130.4/hu.48, AAV145.1/hu.53, AAV145.5/hu.54, AAV145.6/hu.55, AAV16.12/hu. l l, AAV16.3, AAV16.8/hu. lO,
  • AAVA3.5 AAV A3.7, AAV-b, AAVCl, AAVC2, AAVC5, AAVCh.5, AAVCh.5Rl, AAVcy.2, AAVcy.3, AAVcy.4, AAVcy.5, AAVCy.5Rl, AAVCy.5R2, AAVCy.5R3, AAVCy.5R4, AAVcy.6, AAV-DJ, AAV-DJ8, AAVF3, AAVF5, AAV-h, AAVH-l/hu. l, AAVH2, AAVH- 5/hu.3, AAVH6, AAVhEl .
  • AAVhEr1.14 AAVhErl .16, AAVhErl .18, AAVhER1.23, AAVhErl .35, AAVhErl .36, AAVhErl .5, AAVhErl .7, AAVhErl .8, AAVhEr2.16,
  • the AAV serotype may be, or have, a mutation in the AAV9 sequence as described by N Pulichla et al. (Molecular Therapy 19(6): 1070-1078 (2011)), such as but not limited to, AAV9.9, AAV9.11, AAV9.13, AAV9.16, AAV9.24, AAV9.45, AAV9.47, AAV9.61, AAV9.68, AAV9.84.
  • the AAV serotype may be, or have, a sequence as described in U.S. Patent No. U.S. 6156303, such as, but not limited to, AAV3B (SEQ ID NO: 1 and 10 of US 6156303), AAV6 (SEQ ID NO: 2, 7 and 11 of U.S. 6156303), AAV2 (SEQ ID NO: 3 and 8 of U.S. 6156303), AAV3A (SEQ ID NO: 4 and 9, of U.S. 6156303), or derivatives thereof.
  • AAV3B SEQ ID NO: 1 and 10 of US 6156303
  • AAV6 SEQ ID NO: 2, 7 and 11 of U.S. 6156303
  • AAV2 SEQ ID NO: 3 and 8 of U.S. 6156303
  • AAV3A SEQ ID NO: 4 and 9, of U.S. 6156303
  • the serotype may be AAVDJ or a variant thereof, such as AAVDJ8 (or AAV-DJ8), as described by Grimm et al. (Journal of Virology 82(12): 5887-5911 (2008)).
  • the amino acid sequence of AAVDJ8 may comprise two or more mutations in order to remove the heparin binding domain (HBD).
  • HBD heparin binding domain
  • 7,588,772 may comprise two mutations: (1) R587Q where arginine (R; Arg) at amino acid 587 is changed to glutamine (Q; Gin) and (2) R590T where arginine (R; Arg) at amino acid 590 is changed to threonine (T; Thr).
  • R587Q where arginine (R; Arg) at amino acid 587 is changed to glutamine (Q; Gin)
  • R590T where arginine (R; Arg) at amino acid 590 is changed to threonine (T; Thr.
  • the AAV serotype may be, or have, a sequence as described in International Publication No. WO2015121501, such as, but not limited to, true type AAV
  • WO2015121501 Japanese AAV10 (SEQ ID NO: 9 of WO2015121501), or variants thereof.
  • AAV capsid serotype selection or use may be from a variety of species.
  • the AAV may be an avian AAV (AAAV).
  • the AAAV serotype may be, or have, a sequence as described in U.S. Patent No. U.S. 9238800, such as, but not limited to, AAAV (SEQ ID NO: 1, 2, 4, 6, 8, 10, 12, and 14 of U.S. 9,238,800), or variants thereof.
  • the AAV may be a bovine AAV (BAAV).
  • BAAV serotype may be, or have, a sequence as described in U.S. Patent No. U.S. 9,193,769, such as, but not limited to, BAAV (SEQ ID NO: 1 and 6 of U.S. 9193769), or variants thereof.
  • BAAV serotype may be or have a sequence as described in U.S. Patent No. U.S.7427396, such as, but not limited to, BAAV (SEQ ID NO: 5 and 6 of U.S.7427396), or variants thereof.
  • the AAV may be a caprine AAV.
  • the caprine AAV serotype may be, or have, a sequence as described in U.S. Patent No. U.S.7427396, such as, but not limited to, caprine AAV (SEQ ID NO: 3 of U.S.7427396), or variants thereof.
  • the AAV may be engineered as a hybrid AAV from two or more parental serotypes.
  • the AAV may be AAV2G9 which comprises sequences from AAV2 and AAV9.
  • the AAV2G9 AAV serotype may be, or have, a sequence as described in U.S. Patent Publication No. U.S. 20160017005.
  • the AAV may be a serotype generated by the AAV9 capsid library with mutations in amino acids 390-627 (VPl numbering) as described by Pulichla et al.
  • the serotype and corresponding nucleotide and amino acid substitutions may be, but is not limited to, AAV9.1 (G1594C; D532H), AAV6.2 (T1418A and T1436X; V473D and I479K), AAV9.3 (T1238A; F413Y), AAV9.4 (T1250C and A1617T; F417S), AAV9.5 (A1235G, A1314T, A1642G, C1760T; Q412R, T548A, A587V), AAV9.6 (T1231A; F411I), AAV9.9 (G1203A, G1785T; W595C), AAV9.10 (A1500G, T1676C; M559T), AAV9.11 (A1425T, A1702C, A1769T; T568P, Q590L), AAV9.13 (A1369C, A1720T; N457H
  • the AAV may be a serotype comprising at least one AAV capsid CD8+ T-cell epitope.
  • the serotype may be AAV1, AAV2 or AAV8.
  • the AAV may be a variant, such as PUP. A or PUP.B as described in Deverman. 2016. Nature Biotechnology. 34(2): 204-209.
  • the AAV may be a serotype selected from any of those found in SEQ ID NOs: 4734-5302 and Table 2.
  • the AAV may be encoded by a sequence, fragment or variant as disclosed in SEQ ID NOs: 4734-5302 and Table 2.
  • AAV vector serotypes can be matched to target cell types.
  • the following exemplary cell types can be transduced by the indicated AAV serotypes among others.
  • viral vectors include, but are not limited to, lentivirus, alphavirus, enterovirus, pestivirus, baculovirus, herpesvirus, Epstein Barr virus, papovavirus, poxvirus, vaccinia virus, and herpes simplex virus.
  • Cas9 mRNA, sgRNA targeting one or two sites in ATXN3 gene, and donor DNA can each be separately formulated into lipid nanoparticles, or are all co-formulated into one lipid nanoparticle.
  • Cas9 mRNA can be formulated in a lipid nanoparticle, while sgRNA and donor DNA can be delivered in an AAV vector.
  • the guide RNA can be expressed from the same DNA, or can also be delivered as an RNA.
  • the RNA can be chemically modified to alter or improve its half-life, or decrease the likelihood or degree of immune response.
  • the endonuclease protein can be complexed with the gRNA prior to delivery.
  • Viral vectors allow efficient delivery; split versions of Cas9 and smaller orthologs of Cas9 can be packaged in AAV, as can donors for HDR.
  • a range of non-viral delivery methods also exist that can deliver each of these components, or non-viral and viral methods can be employed in tandem. For example, nano-particles can be used to deliver the protein and guide RNA, while AAV can be used to deliver a donor DNA.
  • cells e.g., progenitor cells
  • a desired site such as a site of injury or repair
  • the cells e.g., progenitor cells, or their differentiated progeny can be administered by any appropriate route that results in delivery to a desired location in the subject where at least a portion of the implanted cells or components of the cells remain viable.
  • the period of viability of the cells after administration to a subject can be as short as a few hours, e.g., twenty-four hours, to a few days, to as long as several years, or even the life time of the patient, i.e., long-term engraftment.
  • an effective amount of neural progenitor cells is administered via a systemic route of administration, such as an intraperitoneal or intravenous route.
  • the terms "individual,” “subject,” “host” and “patient” are used interchangeably herein and refer to any subject for whom diagnosis, treatment or therapy is desired.
  • the subject is a mammal.
  • the subject is a human being.
  • progenitor cells described herein can be any progenitor cells described herein.
  • the prophylactic administration of a progenitor cell population serves to prevent Spinocerebellar ataxia 3 (SCA3).
  • a progenitor cell population being administered according to the methods described herein can comprise allogeneic progenitor cells obtained from one or more donors.
  • Such progenitors may be of any cellular or tissue origin, e.g., liver, muscle, cardiac, brain, etc.
  • Allogeneic refers to a progenitor cell or biological samples comprising progenitor cells obtained from one or more different donors of the same species, where the genes at one or more loci are not identical.
  • a liver progenitor cell population being administered to a subject can be derived from one more unrelated donor subjects, or from one or more non- identical siblings.
  • syngeneic progenitor cell populations can be used, such as those obtained from genetically identical animals, or from identical twins.
  • the progenitor cells can be autologous cells; that is, the progenitor cells are obtained or isolated from a subject and administered to the same subject, i.e., the donor and recipient are the same.
  • the term "effective amount” refers to the amount of a population of progenitor cells or their progeny needed to prevent or alleviate at least one or more signs or symptoms of
  • SCA3 Spinocerebellar ataxia 3
  • SCA3 Spinocerebellar ataxia 3
  • therapeutically effective amount therefore refers to an amount of progenitor cells or a composition comprising progenitor cells that is sufficient to promote a particular effect when administered to a typical subject, such as one who has or is at risk for Spinocerebellar ataxia 3 (SCA3).
  • An effective amount would also include an amount sufficient to prevent or delay the development of a symptom of the disease, alter the course of a symptom of the disease (for example but not limited to, slow the progression of a symptom of the disease), or reverse a symptom of the disease. It is understood that for any given case, an appropriate "effective amount" can be determined by one of ordinary skill in the art using routine experimentation.
  • an effective amount of progenitor cells comprises at least 10 2 progenitor cells, at least 5 X 10 2 progenitor cells, at least 10 3 progenitor cells, at least 5 X 10 3 progenitor cells, at least 10 4 progenitor cells, at least 5 X 10 4 progenitor cells, at least 10 5 progenitor cells, at least 2 X 10 5 progenitor cells, at least 3 X 10 5 progenitor cells, at least 4 X 10 5 progenitor cells, at least 5 X 10 5 progenitor cells, at least 6 X 10 5 progenitor cells, at least 7 X 10 5 progenitor cells, at least 8 X 10 5 progenitor cells, at least 9 X 10 5 progenitor cells, at least 1 X 10 6 progenitor cells, at least 2 X 10 6 progenitor cells, at least 3 X 10 6 progenitor cells, at least 4 X 10 6 progenitor progenitor cells, at least 9 X
  • reduction of the expanded trinucleotide repeats in the ATXN3 gene in cells of patients having Spinocerebellar ataxia 3 (SCA3) can be beneficial for ameliorating one or more symptoms of the disease, for increasing long-term survival, and/or for reducing side effects associated with other treatments.
  • SCA3 Spinocerebellar ataxia 3
  • the presence of progenitors that have wild-type or similar levels of the trinucleotide repeat in the ATXN3gene is beneficial.
  • effective treatment of a subject gives rise to at least about 3%, 5% or 7% transcripts having wild-type or similar levels of trinucleotide repeat relative to total ATXN3 transcripts in the treated subject.
  • transcripts having wild- type or similar levels of trinucleotide repeat will be at least about 10% of total ATXN3 transcripts. In some examples, transcripts having wild-type or similar levels of trinucleotide repeat will be at least about 20% to 30%> of total ATXN3 transcripts. Similarly, the introduction of even relatively limited subpopulations of cells having wild-type levels of trinucleotide repeat in the ATXN3 gene can be beneficial in various patients because in some situations normalized cells will have a selective advantage relative to diseased cells.
  • SCA3 Spinocerebellar ataxia 3
  • about 10%>, about 20%, about 30%>, about 40%, about 50%, about 60%, about 70%), about 80%), about 90% or more of the neural progenitors in patients to whom such cells are administered have wild-type or similar levels of trinucleotide repeat in the ATXN3 gene.
  • administering refers to the delivery of a progenitor cell composition into a subject by a method or route that results in at least partial localization of the cell composition at a desired site.
  • a cell composition can be administered by any appropriate route that results in effective treatment in the subject, i.e. administration results in delivery to a desired location in the subject where at least a portion of the composition delivered, i.e. at least 1 x 10 4 cells are delivered to the desired site for a period of time.
  • the pharmaceutical composition may be administered via a route such as, but not limited to, enteral (into the intestine), gastroenteral, epidural (into the dura matter), oral (by way of the mouth), transdermal, peridural, intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intravenous bolus, intravenous drip, intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavernous injection (into a pathologic cavity
  • intragingival within the gingivae
  • intraileal within the distal portion of the small intestine
  • intralesional within or introduced directly to a localized lesion
  • intraluminal within a lumen of a tube
  • intralymphatic within the lymph
  • intramedullary within the marrow cavity of a bone
  • intrameningeal within the meninges
  • intramyocardial within the myocardium
  • intraocular within the eye
  • intraovarian within the ovary
  • intrapericardial within the pericardium
  • intrapleural within the pleura
  • intraprostatic within the prostate gland
  • intrapulmonary within the lungs or its bronchi
  • intrasinal within the nasal or periorbital sinuses
  • intraspinal within the vertebral column
  • intrasynovial within the synovial cavity of a joint
  • intratendinous within a tendon
  • intratesticular within the test
  • ophthalmic to the external eye
  • oropharyngeal directly to the mouth and pharynx
  • parenteral percutaneous, periarticular, peridural, perineural, periodontal, rectal, respiratory (within the respiratory tract by inhaling orally or nasally for local or systemic effect), retrobulbar (behind the pons or behind the eyeball), intramyocardial (entering the myocardium), soft tissue, subarachnoid, subconjunctival, submucosal, topical, transplacental (through or across the placenta), transtracheal (through the wall of the trachea), transtympanic (across or through the tympanic cavity), ureteral (to the ureter), urethral (to the urethra), vaginal, caudal block, diagnostic, nerve block, biliary perfusion, cardiac perfusion, photo
  • Modes of administration include injection, infusion, instillation, and/or ingestion.
  • injection includes, without limitation, intravenous, intramuscular, intra-arterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal,
  • transtracheal subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal, intracerebro spinal, and intrasternal injection and infusion.
  • the route is intravenous.
  • administration by injection or infusion can be made.
  • the cells can be administered systemically.
  • systemic administration refers to the administration of a population of progenitor cells other than directly into a target site, tissue, or organ, such that it enters, instead, the subject's circulatory system and, thus, is subject to metabolism and other like processes.
  • SCA3 Spinocerebellar ataxia 3
  • a treatment is considered "effective treatment,” if any one or all of the signs or symptoms of, as but one example, levels of trinucleotide repeat in the ATXN3 gene are altered in a beneficial manner (e.g., decreased by at least 10%), or other clinically accepted symptoms or markers of disease are improved or ameliorated.
  • Efficacy can also be measured by failure of an individual to worsen as assessed by hospitalization or need for medical interventions (e.g., progression of the disease is halted or at least slowed). Methods of measuring these indicators are known to those of skill in the art and/or described herein.
  • Treatment includes any treatment of a disease in an individual or an animal (some non-limiting examples include a human, or a mammal) and includes: (1) inhibiting the disease, e.g., arresting, or slowing the progression of symptoms; or (2) relieving the disease, e.g., causing regression of symptoms; and (3) preventing or reducing the likelihood of the development of symptoms.
  • the treatment according to the present disclosure can ameliorate one or more symptoms associated with Spinocerebellar ataxia 3 (SCA3) by reducing the number of trinucleotide repeat in the ATXN3 gene in the individual.
  • SCA3 Spinocerebellar ataxia 3
  • ATXN3 has been associated with diseases and disorders such as, but not limited to, Congenital Abnormality, Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Ataxia, Ataxia Telangiectasia, Cerebellar Ataxia, Cerebellar Diseases, Chorea, Cleft Palate, Cystic Fibrosis, Mental Depression, Depressive disorder, Dystonia, Esophageal Neoplasms, Exotropia, Cardiac Arrest, Huntington Disease, Machado- Joseph Disease, Movement Disorders, Muscular
  • Dystrophy Myotonic Dystrophy, Narcolepsy, Nerve Degeneration, Neuroblastoma, Parkinson Disease, Peripheral Neuropathy, Restless Legs Syndrome, Retinal Degeneration, Retinitis Pigmentosa, Schizophrenia, Shy-Drager Syndrome, Sleep disturbances, Hereditary Spastic Paraplegia, Thromboembolism, Stiff-Person Syndrome, Spinocerebellar Ataxia, Esophageal carcinoma, Polyneuropathy, Effects of heat, Muscle twitch, Extrapyramidal sign, Ataxic, Neurologic Symptoms, Cerebral atrophy, Parkinsonian Disorders, Protein S Deficiency, Cerebellar degeneration, Familial Amyloid Neuropathy Portuguese Type, Spastic syndrome, Vertical Nystagmus, Nystagmus End-Position, Antithrombin III Deficiency, Atrophic,
  • ATXN3 is largely confined to central nervous system and ATXN3 is associated with Spinocerebellar ataxia 3 (SCA3) (also commonly known as Machado-Joseph disease).
  • SCA3 Spinocerebellar ataxia 3
  • SCA3 is caused by expansion of a trinucleotide CAG repeat in the ATXN3 gene. This CAG repeat region is located in exon 10 of the ATXN3 gene and encodes a polyglutamine tract. In most people, the CAG trinucleotide is repeated 12 to 43 times. In individuals with SCA3, the number of CAG repeat is greater than 50.
  • CAG repeat intermediate numbers of the CAG repeat (about 44-52) may or may not develop the disease.
  • the number of repeats in the gene correlates with the age of onset and severity of the disease. People with 75 or fewer repeats tend to develop signs and symptoms of SCA3 in mid-adulthood, while people with around 80 repeats usually have signs and symptoms by their teens.
  • the abnormal repeat expansion leads to the production of an altered version of the ATXN3 protein, which is nonfunctional and aggregates with other proteins in the nucleus, causing toxicity to neuronal cells.
  • SCA3 Spinocerebellar ataxia 3
  • SCA3 is an inherited disease (autosomal dominant) where the body experiences degeneration of cells in the hindbrain.
  • SCA3 affects about 1-2 in 100,000 people worldwide and is more common in Azorean populations.
  • Common symptoms of Spinocerebellar ataxia 3 (SCA3) includes memory deficit, spasticity, difficulty with speech and swallowing, weakness in arms and legs, clumsiness, frequent urination and involuntary eye movements.
  • symptoms may be treated using levodopa therapy to ease stiffness and slowness of movements; antispasmodic drugs, such as baclofen, to reduce spasticity; botulinum toxin to reduce severe spasticity and dystonia; speech therapy to improve speech problems, such as dysarthria and dysphagia; prism glasses to reduce blurred or double vision; and physiotherapy to cope with disability associated with gait problems.
  • antispasmodic drugs such as baclofen
  • botulinum toxin to reduce severe spasticity and dystonia
  • speech therapy to improve speech problems, such as dysarthria and dysphagia
  • prism glasses to reduce blurred or double vision
  • physiotherapy to cope with disability associated with gait problems.
  • the target tissue for the compositions and methods described herein is central nervous system tissue.
  • the gene is Spinocerebellar Ataxia Type 3 Protein (ATXN3) which may also be referred to as Ataxin 3, Machado-Joseph Disease (Spinocerebellar Ataxia 3, Olivopontocerebellar Ataxia 3, Autosomal Dominant, Ataxin 3), Spinocerebellar Ataxia Type 3 Protein, Machado-Joseph Disease Protein 1, SCA3, ATX3, MJDl, Olivopontocerebellar Ataxia, Ataxin 3 Variant Ref, Ataxin 3 Variant An, Ataxin 3 Variant Ao, Ataxin 3 Variant At, Ataxin 3 Variant E, Ataxin 3 Variant H, Ataxin 3 Variant M, Ataxin 3 Variant R, Ataxin 3 Variant Y, EC 3.4.19.12, EC 3.4.22, Josephin, AT3, and JOS.
  • ATXN3 has a cytogenetic location of 14q32
  • ATXN3 The nucleotide sequence of ATXN3 is shown as SEQ ID NO: 5303.
  • NDUFB 1 is the gene upstream of ATXN3 on the reverse strand and TRIP 11 is a gene downstream of and overlapping ATXN3 on the reverse strand.
  • FBLN5 is also a gene downstream of ATXN3, but does not overlap ATXN3.
  • ATXN3 has a NCBI gene ID of 4287, Uniprot ID of P54252 and Ensembl Gene ID of ENSG00000066427.
  • ATXN3 has 2939 SNPs, 117 introns and 80 exons. The exon identifier from Ensembl and the start/stop sites of the introns and exons are shown in Table 3.
  • EX24 ENSE00003543114 92,106,574 - INT24 Intron ENSE00003654296 - 92,080,964 - 92,106,529 ENSE00003723395 92,080,568
  • EX33 ENSE00003734757 92,080,994 - INT33 Intron ENSE00003715808 - 92,077,580 - 92,080,968 ENSE00003713862 92,073,537
  • EX34 ENSE00003635015 92,071,053 - INT34 Intron ENSE00003713862 - 92,073,531 - 92,070,938 ENSE00003746704 92,071,054
  • EX36 ENSE00003497091 92,096,134 - INT36 Intron ENSE00003646843 - 92,096,673 - 92,096,093 ENSE00003608002 92,096,135
  • EX41 ENSE00002023327 92,106,590 - INT41 Intron ENSE00003543114 - 92,106,528 - 92,106,529 ENSE00003499200 92,093,832
  • EX42 ENSE00003677024 92,093,318 - INT42 Intron ENSE00003673714 - 92,106,528 - 92,093,252 ENSE00003497091 92,096,135
  • EX50 ENSE00003561582 92,071,053 - INT50 Intron ENSE00003673714 - 92,106,528 - 92,070,938 ENSE00003616151 92,083,259
  • EX54 ENSE00003677507 92,088,817 - INT54 Intron ENSE00003628190 - 92,088,729 - 92,088,730 ENSE00003529871 92,083,259
  • EX78 ENSE00002048204 92,093,831 - INT78 Intron ENSE00003580006 - 92,082,299 - 92,093,379 ENSE00002498225 92,081,062
  • Table 4 provides information on all of the transcripts for the ATXN3 gene based on the Ensembl database. Provided in Table 4 are the transcript ID from Ensembl and
  • EX46 INT83 ENST0000 ENSPOOOOO Nonsense EX2, EX3, EX4, INT2, INT3, INT4, 0554673.5 452356 mediated EX5, EX24, INT5, INT29, decay EX43, EX44, INT55, INT56,
  • EX50, EX56 INT76, INT77 ENST0000 ENSPOOOOO Nonsense EX2, EX3, EX18, INT2, INT29, 0556274.5 451733 mediated EX24, EX42, INT52, INT53, decay EX44, EX50, INT76, INT77,
  • ATXN3 has 2939 S Ps and the NCBI rs number and/or UniProt VAR number for this ATXN3 gene are rsl0129742, rsl0129909, rsl0131594, rsl0131670, rsl0132620, rsl0133404, rsl0135150, rsl0135173, rsl0135377, rsl0135397, rsl0135695, rsl0137541, rsl0137707, rsl0137727, rsl0138296, rsl0138566, rsl0140469, rsl0140481, rsl0140609, rsl0140666, rsl0140720, rsl0141916, rsl0142041, rsl0143213, rsl0143310, rsl0143456,
  • rs760416170 rs760444067 rs760473424 rs760473560
  • rs760503204, rs760542502 rs760579781,, rs760604342 rs760655980 rs760721207, rs760806353, rs760827214, rs76083403, 1 rs760842332 rs760852869, rs760915098, rs760924707, rs760946854,
  • rs760999964 rs761067047 rs761103772 rs761123692, rs761201018, rs761232988, rs761255070,, rs761263261 rs761293315 rs761316840, rs76132524, rs761352592,
  • rs761352759 rs761471269 rs761487712 rs761551, rs761552, rs761553, rs761590796, rs76164879, ⁇ rs761679000, rs761698296, rs761758856, rs761770661, rs761777507,
  • rs761786252 , rs761867251 rs761883210 rs761953791, rs761959610, rs762007434, rs762049636,, rs762077144 rs762081679 rs762083785, rs762189801, rs762259956, rs762276711,, rs762283961 rs762327715 rs762350146, rs762361430, rs762367488, rs762415878,, rs762420328 rs762428905 rs762429807, rs762458117, rs762497293, rs762526563,, rs762617605 rs762622537 rs762681307, rs762681898, rs762685504, rs762720265,, rs762803649
  • rs765591204 rs765615691 rs765647583, rs765659544, rs765735477, rs765747509, rs765788785,, rs765825926 rs765833506, rs765889218, rs765913054, rs765925436, rs765965653,, rs765985710 rs766001707, rs766144891, rs766148892, rs766197800, rs766237827,, rs766254893 rs766414737, rs766429455, rs766515714, rs766538029, rs766584856,, rs766638246 rs766647421, rs766718545, rs766735261, rs766748486, rs766771475,
  • rs779845888 rs779866271, rs779933798, rs779942460, rs779963548, rs780075943, rs780113339, rs780142385, rs78014519, rs780256409, rs780308101, rs780326779,
  • rs780362590 rs780511182, rs780591975, rs780604497, rs780623755, rs780627255, rs780734968, rs780792601, rs780806094, rs780880202, rs780895696, rs780906123, rs780972744, rs780973227, rs780988220, rs780997768, rs781063243, rs781068463, rs781154422, rs781273115, rs781275143, rs781295503, rs781349710, rs781356865, rs78143585, rs781437447, rs781475859, rs781556964, rs781563953, rs781702814,
  • the guide RNA used in the present disclosure may comprise at least one 20 nucleotide (nt) target nucleic acid sequence listed in Table 5.
  • nt nucleotide
  • Table 5 Provided in Table 5 are the gene symbol and the sequence identifier of the gene (Gene SEQ ID NO), the gene sequence including 1-5 kilobase pairs upstream and/or downstream of the target gene (Extended Gene SEQ ID NO), and the 20 nt target nucleic acid sequence (20 nt Target Sequence SEQ ID NO).
  • the strand for targeting the gene (noted by a (+) strand or (-) strand in the sequence listing)
  • the associated PAM type and the PAM sequence are described for each of the 20 nt target nucleic acid sequences (SEQ ID NO: 5305-56902). It is understood in the art that the spacer sequence, where "T” is "U,” may be an RNA sequence corresponding to the 20 nt sequences listed in Table 5.
  • the guide RNA used in the present disclosure may comprise at least one spacer sequence that, where "T” is "U”, may be an RNA sequence corresponding to a 20 nucleotide (nt) target sequence such as, but not limited to, any of SEQ ID NO: 5305-56902.
  • the guide RNA used in the present disclosure may comprise at least one spacer sequence which, where "T” is "U,” is an RNA sequence corresponding to the 20 nt sequences such as, but not limited to, any of SEQ ID NO: 5305-56902.
  • a guide RNA may comprise a 20 nucleotide (nt) target nucleic acid sequence associated with the PAM type such as, but not limited to, NAAAAC, NNAGAAW, NNGRRT, NNNNGHTT, NRG, or YTN.
  • the 20 nt target nucleic acid sequence for a specific target gene and a specific PAM type may be, where "T" is "U,” the RNA sequence corresponding to any one of the 20 nt nucleic acid sequences in Table 6.
  • a guide RNA may comprise a 20 nucleotide (nt) target nucleic acid sequence associated with the YTN PAM type.
  • the 20 nt target nucleic acid sequence for a specific target gene may comprise a 20 nt core sequence where the 20 nt core sequence, where "T" is "U,” may be the RNA sequence corresponding to SEQ ID NO: 30886-56902.
  • the 20 nt target nucleic acid sequence for a specific target gene may comprise a core sequence where the core sequence, where "T" is "U,” may be a fragment, segment or region of the RNA sequence corresponding to any of SEQ ID NO: 30886-56902.
  • nucleases engineered to target specific sequences there are four major types of nucleases: meganucleases and their derivatives, zinc finger nucleases (ZFNs), transcription activator like effector nucleases (TALENs), and CRISPR-Cas9 nuclease systems.
  • ZFNs zinc finger nucleases
  • TALENs transcription activator like effector nucleases
  • CRISPR-Cas9 nuclease systems The nuclease platforms vary in difficulty of design, targeting density and mode of action, particularly as the specificity of ZFNs and TALENs is through protein-DNA interactions, while RNA-DNA interactions primarily guide Cas9.
  • CRISPR endonucleases such as Cas9
  • Cas9 can be used in the methods of the present disclosure.
  • teachings described herein, such as therapeutic target sites could be applied to other forms of endonucleases, such as ZFNs, TALENs, HEs, or MegaTALs, or using combinations of nucleases.
  • endonucleases such as ZFNs, TALENs, HEs, or MegaTALs, or using combinations of nucleases.
  • Additional binding domains can be fused to the Cas9 protein to increase specificity.
  • the target sites of these constructs would map to the identified gRNA specified site, but would require additional binding motifs, such as for a zinc finger domain.
  • a meganuclease can be fused to a TALE DNA-binding domain.
  • the meganuclease domain can increase specificity and provide the cleavage.
  • inactivated or dead Cas9 dCas9
  • dCas9 inactivated or dead Cas9
  • dCas9 can be fused to a cleavage domain and require the sgRNA/Cas9 target site and adjacent binding site for the fused DNA-binding domain. This likely would require some protein engineering of the dCas9, in addition to the catalytic inactivation, to decrease binding without the additional binding site.
  • Zinc finger nucleases are modular proteins comprised of an engineered zinc finger DNA binding domain linked to the catalytic domain of the type II endonuclease Fokl. Because Fokl functions only as a dimer, a pair of ZFNs must be engineered to bind to cognate target "half-site" sequences on opposite DNA strands and with precise spacing between them to enable the catalytically active Fokl dimer to form. Upon dimerization of the Fokl domain, which itself has no sequence specificity per se, a DNA double-strand break is generated between the ZFN half-sites as the initiating step in genome editing.
  • each ZFN is typically comprised of 3-6 zinc fingers of the abundant Cys2-His2 architecture, with each finger primarily recognizing a triplet of nucleotides on one strand of the target DNA sequence, although cross-strand interaction with a fourth nucleotide also can be important. Alteration of the amino acids of a finger in positions that make key contacts with the DNA alters the sequence specificity of a given finger. Thus, a four-finger zinc finger protein will selectively recognize a 12 bp target sequence, where the target sequence is a composite of the triplet preferences contributed by each finger, although triplet preference can be influenced to varying degrees by neighboring fingers.
  • ZFNs can be readily re-targeted to almost any genomic address simply by modifying individual fingers, although considerable expertise is required to do this well.
  • proteins of 4-6 fingers are used, recognizing 12-18 bp respectively.
  • a pair of ZFNs will typically recognize a combined target sequence of 24-36 bp, not including the typical 5-7 bp spacer between half-sites.
  • the binding sites can be separated further with larger spacers, including 15-17 bp.
  • a target sequence of this length is likely to be unique in the human genome, assuming repetitive sequences or gene homologs are excluded during the design process.
  • the ZFN protein-DNA interactions are not absolute in their specificity so off-target binding and cleavage events do occur, either as a heterodimer between the two ZFNs, or as a homodimer of one or the other of the ZFNs.
  • TALENs represent another format of modular nucleases whereby, as with ZFNs, an engineered DNA binding domain is linked to the Fokl nuclease domain, and a pair of TALENs operate in tandem to achieve targeted DNA cleavage.
  • the major difference from ZFNs is the nature of the DNA binding domain and the associated target DNA sequence recognition properties.
  • the TALEN DNA binding domain derives from TALE proteins, which were originally described in the plant bacterial pathogen Xanthomonas sp.
  • TALEs are comprised of tandem arrays of 33-35 amino acid repeats, with each repeat recognizing a single base pair in the target DNA sequence that is typically up to 20 bp in length, giving a total target sequence length of up to 40 bp.
  • Nucleotide specificity of each repeat is determined by the repeat variable diresidue (RVD), which includes just two amino acids at positions 12 and 13.
  • RVD repeat variable diresidue
  • the bases guanine, adenine, cytosine and thymine are predominantly recognized by the four RVDs: Asn- Asn, Asn-Ile, His-Asp and Asn-Gly, respectively.
  • ZFNs the protein-DNA interactions of TALENs are not absolute in their specificity, and TALENs have also benefitted from the use of obligate heterodimer variants of the Fokl domain to reduce off-target activity.
  • Fokl domains have been created that are deactivated in their catalytic function. If one half of either a TALEN or a ZFN pair contains an inactive Fokl domain, then only single-strand DNA cleavage (nicking) will occur at the target site, rather than a DSB. The outcome is comparable to the use of CRISPR/Cas9 or CRISPR/Cpfl "nickase" mutants in which one of the Cas9 cleavage domains has been deactivated. DNA nicks can be used to drive genome editing by FIDR, but at lower efficiency than with a DSB. The main benefit is that off-target nicks are quickly and accurately repaired, unlike the DSB, which is prone to NHEJ-mediated mis-repair.
  • TALEN-based systems have been described in the art, and modifications thereof are regularly reported; see, e.g., Boch, Science 326(5959): 1509-12 (2009); Mak et al., Science 335(6069):716-9 (2012); and Moscou et al., Science 326(5959): 1501 (2009).
  • the use of TALENs based on the "Golden Gate” platform, or cloning scheme, has been described by multiple groups; see, e.g., Cermak et al., Nucleic Acids Res. 39(12):e82 (2011); Li et al., Nucleic Acids Res.
  • Homing endonucleases are sequence-specific endonucleases that have long recognition sequences (14-44 base pairs) and cleave DNA with high specificity - often at sites unique in the genome.
  • HEs can be used to create a DSB at a target locus as the initial step in genome editing.
  • some natural and engineered HEs cut only a single strand of DNA, thereby functioning as site-specific nickases.
  • the large target sequence of HEs and the specificity that they offer have made them attractive candidates to create site-specific DSBs.
  • the MegaTAL platform and Tev-mTALEN platform use a fusion of TALE DNA binding domains and catalytically active HEs, taking advantage of both the tunable DNA binding and specificity of the TALE, as well as the cleavage sequence specificity of the HE; see, e.g., Boissel et al., NAR 42: 2591-2601 (2014); Kleinstiver et al., G3 4: 1155-65 (2014); and Boissel and Scharenberg, Methods Mol. Biol. 1239: 171-96 (2015).
  • the MegaTev architecture is the fusion of a meganuclease (Mega) with the nuclease domain derived from the GIY-YIG homing endonuclease I-Tevl (Tev).
  • the two active sites are positioned ⁇ 30 bp apart on a DNA substrate and generate two DSBs with non-compatible cohesive ends; see, e.g., Wolfs et al., NAR 42, 8816-29 (2014). It is anticipated that other combinations of existing nuclease-based approaches will evolve and be useful in achieving the targeted genome modifications described herein.
  • the CRISPR genome editing system typically uses a single Cas9 endonuclease to create a DSB.
  • the specificity of targeting is driven by a 20 or 24 nucleotide sequence in the guide RNA that undergoes Watson-Crick base-pairing with the target DNA (plus an additional 2 bases in the adjacent NAG or NGG PAM sequence in the case of Cas9 from S. pyogenes).
  • RNA/DNA interaction is not absolute, with significant promiscuity sometimes tolerated, particularly in the 5' half of the target sequence, effectively reducing the number of bases that drive specificity.
  • One solution to this has been to completely deactivate the Cas9 or Cpfl catalytic function - retaining only the RNA-guided DNA binding function - and instead fusing a Fokl domain to the deactivated Cas9; see, e.g., Tsai et al., Nature Biotech 32: 569-76 (2014); and Guilinger et al., Nature Biotech. 32: 577-82 (2014).
  • Fokl must dimerize to become catalytically active, two guide RNAs are required to tether two Fokl fusions in close proximity to form the dimer and cleave DNA. This essentially doubles the number of bases in the combined target sites, thereby increasing the stringency of targeting by CRISPR-based systems.
  • fusion of the TALE DNA binding domain to a catalytically active FIE, such as I-Tevl takes advantage of both the tunable DNA binding and specificity of the TALE, as well as the cleavage sequence specificity of I-Tevl, with the expectation that off- target cleavage can be further reduced.
  • kits for carrying out the methods described herein.
  • a kit can include one or more of a genome-targeting nucleic acid, a polynucleotide encoding a genome-targeting nucleic acid, a site-directed polypeptide, a polynucleotide encoding a site- directed polypeptide, and/or any nucleic acid or proteinaceous molecule necessary to carry out the aspects of the methods described herein, or any combination thereof.
  • a kit can comprise: (1) a vector comprising a nucleotide sequence encoding a genome-targeting nucleic acid, (2) the site-directed polypeptide or a vector comprising a nucleotide sequence encoding the site-directed polypeptide, and (3) a reagent for reconstitution and/or dilution of the vector(s) and or polypeptide.
  • a kit can comprise: (1) a vector comprising (i) a nucleotide sequence encoding a genome-targeting nucleic acid, and (ii) a nucleotide sequence encoding the site-directed polypeptide; and (2) a reagent for reconstitution and/or dilution of the vector.
  • the kit can comprise a single-molecule guide genome- targeting nucleic acid.
  • the kit can comprise a double-molecule genome- targeting nucleic acid.
  • the kit can comprise two or more double- molecule guides or single-molecule guides.
  • the kits can comprise a vector that encodes the nucleic acid targeting nucleic acid.
  • the kit can further comprise a polynucleotide to be inserted to effect the desired genetic modification.
  • kits can be in separate containers, or combined in a single container.
  • any kit described above can further comprise one or more additional reagents, where such additional reagents are selected from a buffer, a buffer for introducing a polypeptide or polynucleotide into a cell, a wash buffer, a control reagent, a control vector, a control RNA polynucleotide, a reagent for in vitro production of the polypeptide from DNA, adaptors for sequencing and the like.
  • a buffer can be a stabilization buffer, a reconstituting buffer, a diluting buffer, or the like.
  • a kit can also comprise one or more components that can be used to facilitate or enhance the on-target binding or the cleavage of DNA by the endonuclease, or improve the specificity of targeting.
  • a kit can further comprise instructions for using the components of the kit to practice the methods.
  • the instructions for practicing the methods can be recorded on a suitable recording medium.
  • the instructions can be printed on a substrate, such as paper or plastic, etc.
  • the instructions can be present in the kits as a package insert, in the labeling of the container of the kit or components thereof ⁇ i.e., associated with the packaging or subpackaging), etc.
  • the instructions can be present as an electronic storage data file present on a suitable computer readable storage medium, e.g. CD-ROM, diskette, flash drive, etc.
  • the actual instructions are not present in the kit, but means for obtaining the instructions from a remote source ⁇ e.g. via the Internet), can be provided.
  • An example of this case is a kit that comprises a web address where the instructions can be viewed and/or from which the instructions can be downloaded. As with the instructions, this means for obtaining the instructions can be recorded on a suitable substrate.
  • the present disclosure relates in particular to the following non-limiting methods according to the present disclosure:
  • Method 1 provides a method for editing a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell by genome editing comprising: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
  • DNA deoxyribonucleic acid
  • SSBs single-strand breaks
  • DSBs double-strand breaks
  • Method 2 provides a method for editing a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell by genome editing comprising: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
  • DNA deoxyribonucleic acid
  • SSBs single-strand breaks
  • DSBs double-strand breaks
  • Method 3 provides an ex vivo method for treating a patient having an ATXN3 related condition or disorder comprising: creating a patient specific induced pluripotent stem cell (iPSC); editing within or near a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene or other DNA sequences that encode regulatory elements of the ATXN3 gene of the iPSC; differentiating the genome-edited iPSC into a neuron or glial cell of the Central Nervous System (CNS); and implanting the neuron or glial cell of the Central Nervous System (CNS) into the patient.
  • iPSC patient specific induced pluripotent stem cell
  • ATXN3 Spinocerebellar Ataxia Type 3 Protein
  • Method 4 provides the method of Method 3, wherein the editing step comprises: introducing into the iPSC one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
  • DNA deoxyribonucleic acid
  • Method 5 provides the method of Method 3, wherein the editing step comprises: introducing into the iPSC one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
  • DNA deoxyribonucleic acid
  • Method 6 provides an ex vivo method for treating a patient having an ATXN3 related condition or disorder comprising: isolating a mesenchymal stem cell from the patient; editing within or near a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene or other DNA sequences that encode regulatory elements of the ATXN3 gene of the mesenchymal stem cell; differentiating the genome-edited mesenchymal stem cell into a neuron or glial cell of the Central Nervous System (CNS); and implanting the neuron or glial cell of the Central Nervous System (CNS) into the patient.
  • ATXN3 Spinocerebellar Ataxia Type 3 Protein
  • Method 7 provides the method of Method 6, wherein the editing step comprises: introducing into the mesenchymal stem cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
  • DNA deoxyribonucleic acid
  • Method 8 provides the method of Method 6, wherein the editing step comprises: introducing into the mesenchymal stem cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
  • DNA deoxyribonucleic acid
  • SSBs single-strand breaks
  • DSBs double-strand breaks
  • Method 9 provides an in vivo method for treating a patient with an ATXN3 related disorder comprising: editing the Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell of the patient.
  • ATXN3 Spinocerebellar Ataxia Type 3 Protein
  • Method 10 provides the method of Method 9, wherein the editing step comprises: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
  • DNA deoxyribonucleic acid
  • SSBs single-strand breaks
  • DRBs double-strand breaks
  • Method 11 provides the method of Method 9, wherein the editing step comprises: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
  • DNA deoxyribonucleic acid
  • SSBs single-strand breaks
  • DSBs double-strand breaks
  • Method 12 provides the method of any one of Methods 9-11, wherein the cell is a cell of the Central Nervous System (CNS).
  • CNS Central Nervous System
  • Method 13 the present disclosure provides the method of Method 12, wherein the cell of the Central Nervous System (CNS) is a neuron.
  • CNS Central Nervous System
  • Method 14 provides the method of Method 12, wherein the cell of the Central Nervous System (CNS) is a glial cell.
  • CNS Central Nervous System
  • Method 15 provides the method of any one of Methods 12-14, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is delivered to the cell of the Central Nervous System (CNS) via any administration route selected from the group consisting of intraparenchymal, intravenous, intra-arterial,
  • DNA deoxyribonucleic acid
  • CNS Central Nervous System
  • intracerebroventricular, intracisternal, intrathecal, intracranial or intraperitoneal routes are intracerebroventricular, intracisternal, intrathecal, intracranial or intraperitoneal routes.
  • Method 16 provides a method of altering the contiguous genomic sequence of an ATXN3 gene in a cell comprising contacting the cell with one or more deoxyribonucleic acid (DNA) endonuclease to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs).
  • DNA deoxyribonucleic acid
  • SSBs single-strand breaks
  • DSBs double-strand breaks
  • Method 17 the present disclosure provides the method of Method 16, wherein the alteration of the contiguous genomic sequence occurs in one or more exons of the ATXN3 gene.
  • Method 18 provides the method of Method 17, wherein the alteration of the contiguous genomic sequence occurs in exon 10 of the ATXN3 gene.
  • Method 19 provides the method of any one of Methods 1-18, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is selected from any of those sequence in SEQ ID NOs: 1-620 and variants having at least 90% homology to any of those sequences disclosed in SEQ ID NOs: 1-620.
  • DNA deoxyribonucleic acid
  • Method 20 the present disclosure provides the method of Method 19, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is one or more protein or polypeptide.
  • Method 21 the present disclosure provides the method of Method 19, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is one or more polynucleotide encoding the one or more DNA endonuclease.
  • Method 22 provides the method of Method 21, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is one or more ribonucleic acid (RNA) encoding the one or more DNA endonuclease.
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • Method 23 provides the method of Method 22, wherein the one or more ribonucleic acid (RNA) is one or more chemically modified RNA.
  • RNA ribonucleic acid
  • Method 24 provides the method of Method 23, wherein the one or more ribonucleic acid (RNA) is chemically modified in the coding region.
  • RNA ribonucleic acid
  • Method 25 the present disclosure provides the method of any one of Methods 21-24, wherein the one or more polynucleotide or one or more ribonucleic acid (RNA) is codon optimized.
  • Method 26 provides the method of any one of Methods 1-25, wherein the method further comprises: introducing one or more gRNA or one or more sgRNA.
  • Method 27 provides the method of Method 26, wherein the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a sequence within or near the expanded trinucleotide repeat in the ATXN3 gene.
  • Method 28 provides the method of Method 26, wherein the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a DNA sequence within or near the ATXN3 gene.
  • Method 29 provides the method of Method 26, wherein the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a sequence flanking the ATXN3 gene or other sequence that encodes a regulatory element of the ATXN3 gene.
  • Method 30 provides the method of any one of Methods 26-29, wherein the one or more gRNA or one or more sgRNA is chemically modified.
  • Method 31 the present disclosure provides the method of any one of Methods 26-30, wherein the one or more gRNA or one or more sgRNA is pre-complexed with the one or more deoxyribonucleic acid (DNA) endonuclease.
  • Method 32 the present disclosure provides the method of Method 31, wherein the pre-complexing involves a covalent attachment of the one or more gRNA or one or more sgRNA to the one or more deoxyribonucleic acid (DNA) endonuclease.
  • Method 33 the present disclosure provides the method of any one of Methods 19-32, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is formulated in a liposome or lipid nanoparticle.
  • DNA deoxyribonucleic acid
  • Method 34 provides the method of any one of Methods 26-32, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is formulated in a liposome or a lipid nanoparticle which also comprises the one or more gRNA or one or more sgRNA.
  • DNA deoxyribonucleic acid
  • Method 35 provides the method of any one of Methods 19, or 26-29, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is encoded in an AAV vector particle.
  • DNA deoxyribonucleic acid
  • Method 36 provides the method of any of the Methods 26-29, wherein the one or more gRNA or one or more sgRNA is encoded in an AAV vector particle.
  • Method 37 provides the method of any of the Methods 26-29, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is encoded in an AAV vector particle which also encodes the one or more gRNA or one or more sgRNA.
  • DNA deoxyribonucleic acid
  • Method 38 provides the method of any of the Methods 35-37, wherein the AAV vector particle is selected from the group consisting of any of those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
  • Method 39 the present disclosure provides the method of any of Methods 1-38, wherein the method further comprises: introducing into the cell a donor template comprising at least a portion of the wild-type ATXN3 gene.
  • Method 40 provides the method of Method 39, wherein the at least a portion of the wild-type ATXN3 gene comprises one or more sequences selected from the group consisting of: an ATXN3 exon, an ATXN3 intron, and a sequence comprising an exon:intron junction of ATXN3.
  • Method 41 the present disclosure provides the method of any one of Methods 39-40, wherein the donor template comprises homologous arms to the genomic locus of the ATXN3 gene.
  • Method 42 the present disclosure provides the method of any one of Methods 39-41, wherein the donor template is either a single or double stranded
  • Method 43 provides the method of any one of Methods 39-42, wherein the donor template is encoded in an AAV vector particle, where the AAV vector particle is selected from the group consisting of those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
  • Method 44 provides the method of any one of Methods 39-42, wherein the one or more polynucleotide encoding one or more
  • deoxyribonucleic acid (DNA) endonuclease is formulated into a lipid nanoparticle, and the one or more gRNA or one or more sgRNA is delivered to the cell ex vivo by electroporation and the donor template is delivered to the cell by an adeno-associated virus (AAV) vector.
  • AAV adeno-associated virus
  • Method 45 the present disclosure provides the method of any one of Methods 39-42, wherein the one or more polynucleotide encoding one or more
  • deoxyribonucleic acid (DNA) endonuclease is formulated into a liposome or lipid nanoparticle which also comprises the one or more gRNA or one or more sgRNA and the donor template.
  • compositions comprising a single-molecule guide RNA comprising: at least a spacer sequence that is an RNA sequence corresponding to any of SEQ ID NOs: 5305-56902.
  • composition 2 provides the single- molecule guide RNA of Composition 1, wherein the single-molecule guide RNA further comprises a spacer extension region.
  • composition 3 provides the single- molecule guide RNA of Composition 1, wherein the single-molecule guide RNA further comprises a tracrRNA extension region.
  • composition 4 provides the single- molecule guide RNA of Compositions 1-3, wherein the single-molecule guide RNA is chemically modified.
  • composition 5 provides a single- molecule guide RNA of Compositions 1-4 pre-complexed with a DNA endonuclease.
  • Composition 6 the present disclosure provides the composition of Composition 5, wherein the DNA endonuclease is a Cas9 or Cpfl endonuclease.
  • Composition 7 the present disclosure provides the composition of Composition 6, wherein the Cas9 or Cpfl endonuclease is selected from the group consisting of: S. pyogenes Cas9, S. aureus Cas9, N. meningitides Cas9, S. thermophilus CRISPRl Cas9, S. thermophilus CRISPR 3 Cas9, T. denticola Cas9, L. bacterium D2006 Cpfl and Acidaminococcus sp. BV3L6 Cpfl, and variants having at least 90% homology to the endonucleases.
  • composition 8 the present disclosure provides the composition of Composition 7, wherein the Cas9 or Cpfl endonuclease comprises one or more nuclear localization signals (NLSs).
  • NLSs nuclear localization signals
  • composition 9 provides the composition of Composition 8, wherein at least one NLS is at or within 50 amino acids of the amino-terminus of the Cas9 or Cpfl endonucelase and/or at least one NLS is at or within 50 amino acids of the carboxy -terminus of the Cas9 or Cpfl endonucelase.
  • composition 10 provides a DNA encoding the single-molecule guide RNA of any of Compositions 1-4.
  • composition 11 provides a non- naturally occurring CRISPR/Cas system comprising a polynucleotide encoding a Cas9 or Cpfl enzyme and at least one single-molecule guide RNA from Compositions 1-4.
  • composition 12 In another composition, Composition 12, the present disclosure provides the
  • CRISPR/Cas system of Composition 11 wherein the polynucleotide encoding a Cas9 or Cpfl endonuclease is selected from the group consisting of: S. pyogenes Cas9, S. aureus Cas9, N. meningitides Cas9, S. thermophilus CRISPRl Cas9, S. thermophilus CRISPR 3 Cas9, T.
  • denticola Cas9 L. bacterium ND2006 Cpfl and Acidaminococcus sp. BV3L6 Cpfl, and variants having at least 90% homology to the endonucleases.
  • composition 13 In another composition, Composition 13, the present disclosure provides the
  • composition 14 In another composition, Composition 14, the present disclosure provides the
  • composition 15 the present disclosure provides the present disclosure
  • CRISPR/Cas system of Composition 14 wherein the polynucleotide encoding a Cas9 or Cpfl endonuclease is codon optimized for expression in a eukaryotic cell.
  • composition 16 the present disclosure provides a DNA encoding the CRISPR/Cas system of any one of Compositions 11-15.
  • composition 17 the present disclosure provides a vector comprising the DNA of Compositions of 11 or 16.
  • composition 18 provides the vector of Composition 17, wherein the vector is a plasmid.
  • Composition 19 provides the vector of Composition 17, wherein the vector is an AAV vector particle, and the AAV vector serotype is selected from the group consisting of those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
  • compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the aspect.
  • any numerical range recited in this specification describes all sub-ranges of the same numerical precision (i.e., having the same number of specified digits) subsumed within the recited range.
  • a recited range of "1.0 to 10.0” describes all sub-ranges between (and including) the recited minimum value of 1.0 and the recited maximum value of 10.0, such as, for example, "2.4 to 7.6," even if the range of "2.4 to 7.6" is not expressly recited in the text of the specification.
  • the Applicant reserves the right to amend this specification, including the claims, to expressly recite any sub-range of the same numerical precision subsumed within the ranges expressly recited in this specification.

Abstract

The present disclosure provides materials and methods for treating a patient with one or more conditions associated with ATXN3 whether ex vivo or in vivo. In addition, the present disclosure provides materials and methods for editing and/or modulating the expression of ATXN3 gene in a cell by genome editing.

Description

MATERIALS AND METHODS FOR TREATMENT OF SPINOCEREBELLAR ATAXIA
3 (SCA3) AND OTHER RELATED DISORDERS
Field
[0001] The present disclosure relates to the field of gene editing and specifically to the alteration of the Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene.
Related Applications
[0002] This application claims the benefit of U.S. Provisional Application No. 62/355,936 filed June 29, 2016, and U.S. Provisional Application No. 62/461,857 filed February 22, 2017, both of which are incorporated herein in their entirety by reference.
Incorporation by Reference of Sequence Listing
[0003] This application contains a Sequence Listing in computer readable form (filename: 170137PCT sequence listing (ATXN3) _(Part 1): 19,889,222 bytes - ASCII text file; created June 26, 2017 and 170137PCT sequence listing (ATXN3) _(Part 2): 7,142,691 bytes - ASCII text file; created June 26, 2017), which is incorporated herein by reference in its entirety and forms part of the disclosure.
Background
[0004] Genome engineering refers to the strategies and techniques for the targeted, specific modification of the genetic information (genome) of living organisms. Genome engineering is a very active field of research because of the wide range of possible applications, particularly in the areas of human health. For example, genome engineering can be used to alter (e.g., correct or knock-out) a gene carrying a harmful mutation, or to explore the function of a gene. Early technologies developed to insert a transgene into a living cell were often limited by the random nature of the insertion of the new sequence into the genome. Random insertions into the genome may result in disrupting normal regulation of neighboring genes leading to severe unwanted effects. Furthermore, random integration technologies offer little reproducibility, as there is no guarantee that the sequence would be inserted at the same place in two different cells. Recent genome engineering strategies, such as zinc finger nucleases (ZFNs), transcription activator like effector nucleases (TALENs), homing endonucleases (HEs) and MegaTALs, enable a specific area of the DNA to be modified, thereby increasing the precision of the alteration compared to early technologies. These newer platforms offer a much larger degree of reproducibility, but still have their limitations.
[0005] Despite efforts from researchers and medical professionals worldwide who have been trying to address genetic disorders, and despite the promise of genome engineering approaches, there still remains a critical need for developing safe and effective treatments involving ATXN3 related indications.
[0006] By using genome engineering tools to create permanent changes to the genome that can address the ATXN3 related disorders or conditions with as few as a single treatment, the resulting therapy may completely remedy certain ATXN3 related indications and/or diseases.
Summary
[0007] Provided herein are cellular, ex vivo and in vivo methods for creating permanent changes to the genome by deleting and/or correcting the trinucleotide repeat expansion or replacing one or more nucleotide bases, or one or more exons and/or introns within or near the Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene, or otherwise introducing insertions, deletions or mutations of at least one nucleotide within or near the ATXN3 gene or other DNA sequences that encode regulatory elements of the ATXN3 gene by genome editing. Such methods can reduce or eliminate the expression or function of aberrant ATXN3 gene products and/or restore the wild-type ATXN3 protein activity, which can be used to treat an ATXN3 related condition or disorder such as Spinocerebellar ataxia 3 (SCA3). Also provided herein are components and compositions, and vectors for performing such methods.
[0008] Provided herein is a method for editing a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell by genome editing comprising: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
[0009] Also provided herein is a method for editing a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell by genome editing comprising: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
[00010] Also provided herein is an ex vivo method for treating a patient having an ATXN3 related condition or disorder comprising: creating a patient specific induced pluripotent stem cell (iPSC); editing within or near a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene or other DNA sequences that encode regulatory elements of the ATXN3 gene of the iPSC; differentiating the genome-edited iPSC into a neuron or glial cell of the Central Nervous System (CNS); and implanting the neuron or glial cell of the Central Nervous System (CNS) into the patient.
[00011] In some aspects, the editing step comprises: introducing into the iPSC one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
[00012] In some aspects, the editing step comprises: introducing into the iPSC one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
[00013] Also provided herein is an ex vivo method for treating a patient having an ATXN3 related condition or disorder comprising: isolating a mesenchymal stem cell from the patient; editing within or near a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene or other DNA sequences that encode regulatory elements of the ATXN3 gene of the mesenchymal stem cell; differentiating the genome-edited mesenchymal stem cell into a neuron or glial cell of the Central Nervous System (CNS); and implanting the neuron or glial cell of the Central Nervous System (CNS) into the patient.
[00014] In some aspects, the editing step comprises: introducing into the mesenchymal stem cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
[00015] In some aspects, the editing step comprises: introducing into the mesenchymal stem cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
[00016] Also provided herein is an in vivo method for treating a patient with an ATXN3 related disorder comprising: editing the Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell of the patient.
[00017] In some aspects, the editing step comprises: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
[00018] In some aspects, the editing step comprises: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
[00019] In some aspects, the cell is a cell of the Central Nervous System (CNS). In some aspects, the cell of the Central Nervous System (CNS) is a neuron. In some aspects, the cell of the Central Nervous System (CNS) is a glial cell. In some aspects, the one or more
deoxyribonucleic acid (DNA) endonuclease is delivered to the cell of the Central Nervous System (CNS) via intraparenchymal, intravenous, intra-arterial, intracerebroventricular, intracisternal, intrathecal, intracranial or intraperitoneal administration routes.
[00020] Also provided herein is a method of altering the contiguous genomic sequence of an ATXN3 gene in a cell comprising: contacting the cell with one or more deoxyribonucleic acid (DNA) endonuclease to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs). In some aspects, the alteration of the contiguous genomic sequence occurs in one or more exons of the ATXN3 gene. In some aspects, the alteration of the contiguous genomic sequence occurs in exon 10 of the ATXN3 gene.
[00021] In some aspects, the one or more deoxyribonucleic acid (DNA) endonuclease is selected from any of those sequences in SEQ ID NOs: 1-620 and variants having at least 90% homology to any of the sequences listed in SEQ ID NOs: 1-620.
[00022] In some aspects, the one or more deoxyribonucleic acid (DNA) endonuclease is one or more protein or polypeptide. In some aspects, the one or more deoxyribonucleic acid (DNA) endonuclease is one or more polynucleotide encoding the one or more DNA endonuclease. In some aspects, the one or more deoxyribonucleic acid (DNA) endonuclease is one or more ribonucleic acid (RNA) encoding the one or more DNA endonuclease. In some aspects, the one or more ribonucleic acid (RNA) is one or more chemically modified RNA. In some aspects, the one or more ribonucleic acid (RNA) is chemically modified in the coding region. In some aspects, the one or more polynucleotide or one or more ribonucleic acid (RNA) is codon optimized.
[00023] In some aspects, the methods further comprise: introducing one or more gRNA or one or more sgRNA. In some aspects, the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a sequence within or near the expanded trinucleotide repeat in the ATXN3 gene. In some aspects, the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a DNA sequence within or near the ATXN3 gene. In some aspects, the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a sequence flanking the ATXN3 gene or other sequence that encodes a regulatory element of the ATXN3 gene. In some aspects, the one or more gRNA or one or more sgRNA is chemically modified.
[00024] In some aspects, the one or more gRNA or one or more sgRNA is pre-complexed with the one or more deoxyribonucleic acid (DNA) endonuclease. In some aspects, the pre- complexing involves a covalent attachment of the one or more gRNA or one or more sgRNA to the one or more deoxyribonucleic acid (DNA) endonuclease.
[00025] In some aspects, the one or more deoxyribonucleic acid (DNA) endonuclease is formulated in a liposome or lipid nanoparticle. In some aspects, the one or more
deoxyribonucleic acid (DNA) endonuclease is formulated in a liposome or a lipid nanoparticle which also comprises the one or more gRNA or one or more sgRNA.
[00026] In some aspects, the one or more deoxyribonucleic acid (DNA) endonuclease is encoded in an AAV vector particle. In some aspects, the one or more gRNA or one or more sgRNA is encoded in an AAV vector particle. In some aspects, the one or more
deoxyribonucleic acid (DNA) endonuclease is encoded in an AAV vector particle which also encodes the one or more gRNA or one or more sgRNA. In some aspects, the AAV vector particle is selected from those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
[00027] In some aspects, the methods further comprise: introducing into the cell a donor template comprising at least a portion of the wild-type ATXN3 gene. In some aspects, at least a portion of the wild-type ATXN3 gene comprises one or more sequences selected from an ATXN3 exon, an ATXN3 intron, and a sequence comprising an exon:intr on junction of ATXN3. In some aspects, the donor template comprises homologous arms to the genomic locus of the ATXN3 gene. In some aspects, the donor template is either a single or double stranded polynucleotide.
[00028] In some aspects, the donor template is encoded in an AAV vector particle, where the AAV vector serotype is selected from any of those disclosed in SEQ ID NOs: 4734-5302 and Table 2. In some aspects, the one or more polynucleotide encoding one or more
deoxyribonucleic acid (DNA) endonuclease is formulated into a lipid nanoparticle, and the one or more gRNA or one or more sgRNA is delivered to the cell ex vivo by electroporation and the donor template is delivered to the cell by an adeno-associated virus (AAV) vector. In some aspects, the one or more polynucleotide encoding one or more deoxyribonucleic acid (DNA) endonuclease is formulated into a liposome or lipid nanoparticle which also comprises the one or more gRNA or one or more sgRNA and the donor template.
[00029] Also provided herein is a single-molecule guide RNA comprising: at least a spacer sequence that is an RNA sequence corresponding to any of SEQ ID NOs: 5305-56902. In some aspects, the single-molecule guide RNA further comprises a spacer extension region. In some aspects, the single-molecule guide RNA further comprises a tracrRNA extension region. In some aspects, the single-molecule guide RNA is chemically modified.
[00030] In some aspects, the single-molecule gudie RNA is pre-complexed with a DNA endonuclease. In some aspects, the DNA endonuclease is a Cas9 or CPfl endonuclease. In some aspects, the Cas9 or Cpfl endonuclease is selected from the group consisting of: S.
pyogenes Cas9, S. aureus Cas9, N. meningitides Cas9, S. thermophilus CRISPR1 Cas9, S.
thermophilus CRISPR 3 Cas9, T. denticola Cas9, L. bacterium ND2006 Cpfl and
Acidaminococcus sp. BV3L6 Cpfl, and variants having at least 90% homology to these endonucleases. In some aspects, the Cas9 or Cpfl endonuclease comprises one or more nuclear localization signals (NLSs). In some aspects, at least one NLS is at or within 50 amino acids of the amino-terminus of the Cas9 or Cpfl endonuclease and/or at least one NLS is at or within 50 amino acids of the carboxy-terminus of the Cas9 or Cpfl endonuclease.
[00031] Also provided herein is a non-naturally occurring CRISPR/Cas system comprising a polynucleotide encoding a Cas9 or Cpfl enzyme and at least one single-molecule guide RNA described herein. In some aspects, the polynucleotide encoding a Cas9 or Cpfl endonuclease is selected from a group consisting of: S. pyogenes Cas9, S. aureus Cas9, N. meningitides Cas9, S. thermophilus CRISPR1 Cas9, S. thermophilus CRISPR 3 Cas9, T. denticola Cas9, L. bacterium ND2006 Cpfl and Acidaminococcus sp. BV3L6 Cpfl, and variants having at least 90% homology to these endonucleases. In some aspects, the polynucleotide encoding a Cas9 or Cpfl enzyme comprises one or more nuclear localization signals (NLSs). In some aspects, at least one NLS is at or within 50 amino acids of the amino-terminus of the polynucleotide encoding a Cas9 or Cpfl enzyme and/or at least one NLS is at or within 50 amino acids of the carboxy-terminus of the polynucleotide encoding a Cas9 or Cpfl endonuclease. In some aspects, polynucleotide encoding a Cas9 or Cpfl enzyme is codon optimized for expression in a eukaryotic cell.
[00032] Also provided herein is RNA encoding the single-molecule guide RNA described herein.
[00033] Also provided herein is RNA encoding the CRISPR/Cas system described herein.
[00034] Also provided herein is a DNA encoding the single-molecule guide RNA described herein.
[00035] Also provided herein is a DNA encoding the CRISPR/Cas system described herein.
[00036] Also provided herein is a vector comprising a DNA encoding the single-molecule guide RNA and CRISPR/Cas system. In some aspects, the vector is a plasmid. In some aspects, the vector is an AAV vector particle, wherein the AAV vector particle is selected from those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
Brief Description of the Drawings
[00037] Various aspects of materials and methods disclosed and described in this specification can be better understood by reference to the accompanying figures, in which:
[00038] Figures 1 A-B depict the type II CRISPR/Cas system;
[00039] Figure 1 A is a depiction of the type II CRISPR/Cas system including gRNA;
[00040] Figure IB is another depiction of the type II CRISPR/Cas system including sgRNA;
[00041] Figures 2A-D describe the cutting efficiencies of S.pyogenes gRNAs selected via an in-vitro transcribed (IVT) gRNA screen in HEK293T cells; [00042] Figure 2A describes the cutting efficiencies in the range of 12.5 - 36.8% of S.
pyogenes gRNAs selected via an in-vitro transcribed (IVT) gRNA screen in HEK293T cells;
[00043] Figure 2B describes the cutting efficiencies in the range of 8.0 - 12.5% of S. pyogenes gRNAs selected via an in-vitro transcribed (IVT) gRNA screen in HEK293T cells;
[00044] Figure 2C describes the cutting efficiencies in the range of 3.0 - 7.7% of S. pyogenes gRNAs selected via an in-vitro transcribed (IVT) gRNA screen in HEK293T cells;
[00045] Figure 2D describes the cutting efficiencies in the range of 0.3 - 2.5% of S. pyogenes gRNAs selected via an in-vitro transcribed (IVT) gRNA screen in HEK293T cells;
[00046] Figures 3A-B describe the cutting efficiency of S. pyogenes gRNAs in HEK293T cells;
[00047] Figure 3 A describes the cutting efficiency in the range of 8.3 - 36.8% of S. pyogenes gRNAs in HEK293T cells; and
[00048] Figure 3B describes the cutting efficiency in the range of 0.3 - 8.2% of S. pyogenes gRNAs in HEK293T cells.
Brief Description of Sequence Listing
[00049] SEQ ID NOs: 1-620 are Cas endonuclease ortholog sequences.
[00050] SEQ ID NOs: 621-631 do not include sequences.
[00051] SEQ ID NOs: 632-4715 are microRNA sequences.
[00052] SEQ ID NOs : 4716-4733 do not include sequences.
[00053] SEQ ID NOs: 4734-5302 are AAV serotype sequences.
[00054] SEQ ID NO: 5303 is a ATXN3 nucleotide sequence.
[00055] SEQ ID NO: 5304 is a gene sequence including 1-5 kilobase pairs upstream and/or downstream of the ATXN3 gene.
[00056] SEQ ID NOs: 5305 - 5633 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with a T. denticola Cas9 endonuclease.
[00057] SEQ ID NOs: 5634 - 6353 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with a S. thermophilus Cas9 endonuclease.
[00058] SEQ ID NOs: 6354 -8658 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with a S. aureus Cas9 endonuclease. [00059] SEQ ID NOs: 8659 - 10697 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with a N. meningitides Cas9 endonuclease.
[00060] SEQ ID NOs: 10698 - 30885 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with a S. pyogenes Cas9 endonuclease.
[00061] SEQ ID NOs: 30886 - 56902 are 20 bp spacer sequences for targeting within or near a ATXN3 gene or other DNA sequence that encodes a regulatory element of the ATXN3 gene with an Acidaminococcus, a Lachnospiraceae, and a Franciscella Novicida Cpf 1 endonuclease.
[00062] SEQ ID NOs: 56903-56932 do not include sequences.
[00063] SEQ ID NO: 56933 is a sample guide RNA (gRNA) for a S. pyogenes Cas9 endonuclease.
[00064] SEQ ID NOs: 56934-56936 show sample sgRNA sequences. Detailed Description
I. INTRODUCTION
Genome Editing
[00065] The present disclosure provides strategies and techniques for the targeted, specific alteration of the genetic information (genome) of living organisms. As used herein, the term "alteration" or "alteration of genetic information" refers to any change in the genome of a cell. In the context of treating genetic disorders, alterations may include, but are not limited to, insertion, deletion and correction. As used herein, the term "insertion" refers to an addition of one or more nucleotides in a DNA sequence. Insertions can range from small insertions of a few nucleotides to insertions of large segments such as a cDNA or a gene. The term "deletion" refers to a loss or removal of one or more nucleotides in a DNA sequence or a loss or removal of the function of a gene. In some cases, a deletion can include, for example, a loss of a few
nucleotides, an exon, an intron, a gene segment, or the entire sequence of a gene. In some cases, deletion of a gene refers to the elimination or reduction of the function or expression of a gene or its gene product. This can result from not only a deletion of sequences within or near the gene, but also other events (e.g., insertion, nonsense mutation) that disrupt the expression of the gene. The term "correction" as used herein, refers to a change of one or more nucleotides of a genome in a cell, whether by insertion, deletion or substitution. Such correction may result in a more favorable genotypic or phenotypic outcome, whether in structure or function, to the genomic site which was corrected. One non-limiting example of a "correction" includes the correction of a mutant or defective sequence to a wild-type sequence which restores structure or function to a gene or its gene product(s). Depending on the nature of the mutation, correction may be achieved via various strategies disclosed herein. In one non-limiting example, a missense mutation may be corrected by replacing the region containing the mutation with its wild-type counterpart. As another example, duplication mutations (e.g., repeat expansions) in a gene may be corrected by removing the extra sequences.
[00066] In some aspects, alterations may also include a gene knock-in, knock-out or knockdown. As used herein, the term "knock-in" refers to an addition of a DNA sequence, or fragment thereof into a genome. Such DNA sequences to be knocked-in may include an entire gene or genes, may include regulatory sequences associated with a gene or any portion or fragment of the foregoing. For example, a cDNA encoding the wild-type protein may be inserted into the genome of a cell carrying a mutant gene. Knock-in strategies need not replace the defective gene, in whole or in part. In some cases, a knock-in strategy may further involve substitution of an existing sequence with the provided sequence, e.g., substitution of a mutant allele with a wild- type copy. On the other hand, the term "knock-out" refers to the elimination of a gene or the expression of a gene. For example, a gene can be knocked out by either a deletion or an addition of a nucleotide sequence that leads to a disruption of the reading frame. As another example, a gene may be knocked out by replacing a part of the gene with an irrelevant sequence. Finally, the term "knock-down" as used herein refers to reduction in the expression of a gene or its gene product(s). As a result of a gene knock-down, the protein activity or function may be attenuated or the protein levels may be reduced or eliminated.
[00067] Genome editing generally refers to the process of modifying the nucleotide sequence of a genome, preferably in a precise or pre-determined manner. Examples of methods of genome editing described herein include methods of using site-directed nucleases to cut deoxyribonucleic acid (DNA) at precise target locations in the genome, thereby creating single-strand or double- strand DNA breaks at particular locations within the genome. Such breaks can be and regularly are repaired by natural, endogenous cellular processes, such as homology-directed repair (HDR) and non-homologous end joining (NHEJ), as reviewed in Cox et al., Nature Medicine 21(2), 121-31 (2015). These two main DNA repair processes consist of a family of alternative pathways. NHEJ directly joins the DNA ends resulting from a double-strand break, sometimes with the loss or addition of nucleotide sequence, which may disrupt or enhance gene expression. HDR utilizes a homologous sequence, or donor sequence, as a template for inserting a defined DNA sequence at the break point. The homologous sequence can be in the endogenous genome, such as a sister chromatid. Alternatively, the donor can be an exogenous nucleic acid, such as a plasmid, a single-strand oligonucleotide, a double-stranded oligonucleotide, a duplex
oligonucleotide or a virus, that has regions of high homology with the nuclease-cleaved locus, but which can also contain additional sequence or sequence changes including deletions that can be incorporated into the cleaved target locus. A third repair mechanism can be microhomology- mediated end joining (MMEJ), also referred to as "Alternative NHEJ," in which the genetic outcome is similar to NHEJ in that small deletions and insertions can occur at the cleavage site. MMEJ can make use of homologous sequences of a few base pairs flanking the DNA break site to drive a more favored DNA end joining repair outcome, and recent reports have further elucidated the molecular mechanism of this process; see, e.g., Cho and Greenberg, Nature 518, 174-76 (2015); Kent et a\., Nature Structural and Molecular Biology, Adv. Online
doi: 10.1038/nsmb.2961(2015); Mateos-Gomez et al, Nature 518, 254-57 (2015); Ceccaldi et al, Nature 528, 258-62 (2015). In some instances, it may be possible to predict likely repair outcomes based on analysis of potential microhomologies at the site of the DNA break.
[00068] Each of these genome editing mechanisms can be used to create desired genomic alterations. A step in the genome editing process can be to create one or two DNA breaks, the latter as double-strand breaks or as two single-stranded breaks, in the target locus as near the site of intended mutation. This can be achieved via the use of site-directed polypeptides, as described and illustrated herein.
CRISPR Endonuclease System
[00069] A CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) genomic locus can be found in the genomes of many prokaryotes (e.g., bacteria and archaea). In prokaryotes, the CRISPR locus encodes products that function as a type of immune system to help defend the prokaryotes against foreign invaders, such as virus and phage. There are three stages of CRISPR locus function: integration of new sequences into the CRISPR locus, expression of CRISPR RNA (crRNA), and silencing of foreign invader nucleic acid. Five types of CRISPR systems (e.g., Type I, Type II, Type III, Type U, and Type V) have been identified.
[00070] A CRISPR locus includes a number of short repeating sequences referred to as "repeats." When expressed, the repeats can form secondary structures (e.g., hairpins) and/or comprise unstructured single-stranded sequences. The repeats usually occur in clusters and frequently diverge between species. The repeats are regularly interspaced with unique intervening sequences referred to as "spacers," resulting in a repeat-spacer-repeat locus architecture. The spacers are identical to or have high homology with known foreign invader sequences. A spacer-repeat unit encodes a crisprRNA (crRNA), which is processed into a mature form of the spacer-repeat unit. A crRNA comprises a "seed" or spacer sequence that is involved in targeting a target nucleic acid (in the naturally occurring form in prokaryotes, the spacer sequence targets the foreign invader nucleic acid). A spacer sequence is located at the 5' or 3' end of the crRNA.
[00071] A CRISPR locus also comprises polynucleotide sequences encoding CRISPR
Associated (Cas) genes. Cas genes encode endonucleases involved in the biogenesis and the interference stages of crRNA function in prokaryotes. Some Cas genes comprise homologous secondary and/or tertiary structures.
Type II CRISPR Systems
[00072] crRNA biogenesis in a Type II CRISPR system in nature requires a trans-activating CRISPR RNA (tracrRNA). Non-limiting examples of Type II CRISPR systems are shown in Figures 1A and IB. The tracrRNA can be modified by endogenous RNaselll, and then hybridizes to a crRNA repeat in the pre-crRNA array. Endogenous RNaselll can be recruited to cleave the pre-crRNA. Cleaved crRNAs can be subjected to exoribonuclease trimming to produce the mature crRNA form {e.g., 5' trimming). The tracrRNA can remain hybridized to the crRNA, and the tracrRNA and the crRNA associate with a site-directed polypeptide {e.g., Cas9). The crRNA of the crRNA-tracrRNA-Cas9 complex can guide the complex to a target nucleic acid to which the crRNA can hybridize. Hybridization of the crRNA to the target nucleic acid can activate Cas9 for targeted nucleic acid cleavage. The target nucleic acid in a Type II CRISPR system is referred to as a protospacer adjacent motif (PAM). In nature, the PAM is essential to facilitate binding of a site-directed polypeptide {e.g., Cas9) to the target nucleic acid. Type II systems (also referred to as Nmeni or CASS4) are further subdivided into Type II-A (CASS4) and II-B (CASS4a). Jinek et al, Science, 337(6096): 816-821 (2012) showed that the CRISPR/Cas9 system is useful for RNA-programmable genome editing, and international patent application publication number WO2013/176772 provides numerous examples and applications of the CRISPR/Cas endonuclease system for site-specific gene editing.
Type V CRISPR Systems
[00073] Type V CRISPR systems have several important differences from Type II systems. For example, Cpf 1 is a single RNA-guided endonuclease that, in contrast to Type II systems, lacks tracrRNA. In fact, Cpf 1 -associated CRISPR arrays can be processed into mature crRNAs without the requirement of an additional trans-activating tracrRNA. The Type V CRISPR array can be processed into short mature crRNAs of 42-44 nucleotides in length, with each mature crRNA beginning with 19 nucleotides of direct repeat followed by 23-25 nucleotides of spacer sequence. In contrast, mature crRNAs in Type II systems can start with 20-24 nucleotides of spacer sequence followed by about 22 nucleotides of direct repeat. Also, Cpfl can utilize a T- rich protospacer-adjacent motif such that Cpfl-crRNA complexes efficiently cleave target DNA preceded by a short T-rich PAM, which is in contrast to the G-rich PAM following the target DNA for Type II systems. Thus, Type V systems cleave at a point that is distant from the PAM, while Type II systems cleave at a point that is adjacent to the PAM. In addition, in contrast to Type II systems, Cpfl cleaves DNA via a staggered DNA double-stranded break with a 4 or 5 nucleotide 5' overhang. Type II systems cleave via a blunt double-stranded break. Similar to Type II systems, Cpfl contains a predicted RuvC-like endonuclease domain, but lacks a second HNH endonuclease domain, which is in contrast to Type II systems.
Cas Genes/Polypeptides and Protospacer Adjacent Motifs
[00074] Exemplary CRISPR/Cas polypeptides include the Cas9 polypeptides as published in Fonfara et al, Nucleic Acids Research, 42: 2577-2590 (2014). The CRISPR/Cas gene naming system has undergone extensive rewriting since the Cas genes were discovered. Fonfara et al. also provides PAM sequences for the Cas9 polypeptides from various species (see also Table 1 infra). II. COMPOSITIONS AND METHODS OF THE DISCLOSURE
[00075] Provided herein are cellular, ex vivo and in vivo methods for using genome engineering tools to create permanent changes to the genome by: 1) deleting the abnormal repeat expansion within the ATXN3 gene, by inducing two double-stranded DNA breaks at both sides of the expanded region, and replacing with a corrected sequence; 2) deleting the abnormal repeat expansion within the ATXN3 gene, by inducing one double-stranded DNA break proximal to the expanded region, and replacing with a corrected sequence; 3) deleting or mutating the ATXN3 gene by inducing one or more insertions or deletions within or near the ATXN3 gene or other DNA sequences that encode regulatory elements of the ATXN3 gene, or 4) deleting the mutant ATXN3 gene and inserting a wild-type ATXN3 gene, a cDNA or a minigene (comprised of one or more exons and introns or natural or synthetic introns) into the ATXN3 gene locus or a safe harbor locus. Such methods use endonucleases, such as CRISPR-associated (Cas9, Cpfl and the like) nucleases, to permanently edit within or near the genomic locus of the ATXN3 gene or other DNA sequences that encode regulatory elements of the ATXN3 gene. In this way, examples set forth in the present disclosure can help to restore the wild-type sequence of, or otherwise reduce or eliminate the expression of, the mutant ATXN3 gene with as few as a single treatment (rather than deliver potential therapies for the lifetime of the patient).
Site-Directed Polypeptides (endonucleases, enzymes)
[00076] A site-directed polypeptide is a nuclease used in genome editing to cleave DNA. The site-directed polypeptide can be administered to a cell or a patient as either: one or more polypeptides, or one or more mRNAs encoding the polypeptide. Any of the enzymes or orthologs listed in SEQ ID NOs: 1-620, or disclosed herein, may be utilized in the methods herein.
[00077] In the context of a CRISPR/Cas9 or CRISPR/Cpfl system, the site-directed polypeptide can bind to a guide RNA that, in turn, specifies the site in the target DNA to which the polypeptide is directed. In the CRISPR/Cas9 or CRISPR/Cpfl systems disclosed herein, the site-directed polypeptide can be an endonuclease, such as a DNA endonuclease.
[00078] A site-directed polypeptide can comprise a plurality of nucleic acid-cleaving (i.e., nuclease) domains. Two or more nucleic acid-cleaving domains can be linked together via a linker. For example, the linker can comprise a flexible linker. Linkers can comprise 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, 30, 35, 40 or more amino acids in length.
[00079] Naturally-occurring wild-type Cas9 enzymes comprise two nuclease domains, a UNH nuclease domain and a RuvC domain. Herein, the term "Cas9" refers to both a naturally- occurring and a recombinant Cas9. Cas9 enzymes contemplated herein can comprise a HNH or HNH-like nuclease domain, and/or a RuvC or RuvC-like nuclease domain.
[00080] HNH or HNH-like domains comprise a McrA-like fold. HNH or HNH-like domains comprises two antiparallel β-strands and an a-helix. HNH or HNH-like domains comprises a metal binding site (e.g., a divalent cation binding site). HNH or HNH-like domains can cleave one strand of a target nucleic acid (e.g., the complementary strand of the crRNA targeted strand).
[00081] RuvC or RuvC-like domains comprise an RNaseH or RNaseH-like fold.
RuvC/RNaseH domains are involved in a diverse set of nucleic acid-based functions including acting on both RNA and DNA. The RNaseH domain comprises 5 β-strands surrounded by a plurality of a-helices. RuvC/RNaseH or RuvC/RNaseH-like domains comprise a metal binding site (e.g., a divalent cation binding site). RuvC/RNaseH or RuvC/RNaseH-like domains can cleave one strand of a target nucleic acid (e.g., the non-complementary strand of a double- stranded target DNA). [00082] Site-directed polypeptides can introduce double-strand breaks or single-strand breaks in nucleic acids, e.g., genomic DNA. The double-strand break can stimulate a cell's endogenous DNA-repair pathways (e.g., homology-dependent repair (HDR) or NHEJ or alternative nonhomologous end joining (A-NHEJ) or microhomology-mediated end joining (MMEJ)). NHEJ can repair cleaved target nucleic acid without the need for a homologous template. This can sometimes result in small deletions or insertions (indels) in the target nucleic acid at the site of cleavage, and can lead to disruption or alteration of gene expression. HDR can occur when a homologous repair template, or donor, is available. The homologous donor template can comprise sequences that are homologous to sequences flanking the target nucleic acid cleavage site. The sister chromatid can be used by the cell as the repair template. However, for the purposes of genome editing, the repair template can be supplied as an exogenous nucleic acid, such as a plasmid, duplex oligonucleotide, single-strand oligonucleotide or viral nucleic acid. With exogenous donor templates, an additional nucleic acid sequence (such as a transgene) or modification (such as a single or multiple base change or a deletion) can be introduced between the flanking regions of homology so that the additional or altered nucleic acid sequence also becomes incorporated into the target locus. MMEJ can result in a genetic outcome that is similar to NHEJ in that small deletions and insertions can occur at the cleavage site. MMEJ can make use of homologous sequences of a few base pairs flanking the cleavage site to drive a favored end-joining DNA repair outcome. In some instances, it may be possible to predict likely repair outcomes based on analysis of potential microhomologies in the nuclease target regions.
[00083] Thus, in some cases, homologous recombination can be used to insert an exogenous polynucleotide sequence into the target nucleic acid cleavage site. An exogenous polynucleotide sequence is termed a "donor polynucleotide" (or donor or donor sequence) herein. The donor polynucleotide, a portion of the donor polynucleotide, a copy of the donor polynucleotide, or a portion of a copy of the donor polynucleotide can be inserted into the target nucleic acid cleavage site. The donor polynucleotide can be an exogenous polynucleotide sequence, i.e., a sequence that does not naturally occur at the target nucleic acid cleavage site.
[00084] The modifications of the target DNA due to NHEJ and/or HDR can lead to, for example, mutations, deletions, alterations, integrations, gene correction, gene replacement, gene tagging, transgene insertion, nucleotide deletion, gene disruption, translocations and/or gene mutation. The processes of deleting genomic DNA and integrating non-native nucleic acid into genomic DNA are examples of genome editing. [00085] The site-directed polypeptide can comprise an amino acid sequence having at least 10%, at least 15%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% amino acid sequence identity to a wild-type exemplary site-directed polypeptide [e.g., Cas9 from S. pyogenes, US2014/0068797 Sequence ID No. 8 or Sapranauskas et al, Nucleic Acids Res, 39(21): 9275-9282 (2011)], and various other site-directed polypeptides. The site-directed polypeptide can comprise at least 70, 75, 80, 85, 90, 95, 97, 99, or 100%) identity to a wild-type site-directed polypeptide {e.g., Cas9 from S. pyogenes, supra) over 10 contiguous amino acids. The site-directed polypeptide can comprise at most: 70, 75, 80, 85, 90, 95, 97, 99, or 100% identity to a wild-type site-directed polypeptide (e.g., Cas9 from S. pyogenes, supra) over 10 contiguous amino acids. The site-directed polypeptide can comprise at least: 70, 75, 80, 85, 90, 95, 97, 99, or 100% identity to a wild-type site-directed polypeptide (e.g., Cas9 from S.
pyogenes, supra) over 10 contiguous amino acids in a ΗΝΉ nuclease domain of the site-directed polypeptide. The site-directed polypeptide can comprise at most: 70, 75, 80, 85, 90, 95, 97, 99, or 100%) identity to a wild-type site-directed polypeptide (e.g., Cas9 from S. pyogenes, supra) over 10 contiguous amino acids in a ΗΝΉ nuclease domain of the site-directed polypeptide. The site-directed polypeptide can comprise at least: 70, 75, 80, 85, 90, 95, 97, 99, or 100% identity to a wild-type site-directed polypeptide (e.g., Cas9 from S. pyogenes, supra) over 10 contiguous amino acids in a RuvC nuclease domain of the site-directed polypeptide. The site-directed polypeptide can comprise at most: 70, 75, 80, 85, 90, 95, 97, 99, or 100%) identity to a wild-type site-directed polypeptide (e.g., Cas9 from S. pyogenes, supra) over 10 contiguous amino acids in a RuvC nuclease domain of the site-directed polypeptide.
[00086] The site-directed polypeptide can comprise a modified form of a wild-type exemplary site-directed polypeptide. The modified form of the wild- type exemplary site-directed polypeptide can comprise a mutation that reduces the nucleic acid-cleaving activity of the site- directed polypeptide. The modified form of the wild-type exemplary site-directed polypeptide can have less than 90%, less than 80%, less than 70%, less than 60%, less than 50%, less than 40%), less than 30%, less than 20%, less than 10%, less than 5%, or less than 1% of the nucleic acid-cleaving activity of the wild-type exemplary site-directed polypeptide (e.g., Cas9 from S. pyogenes, supra). The modified form of the site-directed polypeptide can have no substantial nucleic acid-cleaving activity. When a site-directed polypeptide is a modified form that has no substantial nucleic acid-cleaving activity, it is referred to herein as "enzymatically inactive." [00087] The modified form of the site-directed polypeptide can comprise a mutation such that it can induce a single-strand break (SSB) on a target nucleic acid (e.g., by cutting only one of the sugar-phosphate backbones of a double-strand target nucleic acid). In some aspects, the mutation can result in less than 90%, less than 80%>, less than 70%, less than 60%>, less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 5%, or less than 1% of the nucleic acid-cleaving activity in one or more of the plurality of nucleic acid-cleaving domains of the wild-type site directed polypeptide (e.g., Cas9 from S. pyogenes, supra). In some aspects, the mutation can result in one or more of the plurality of nucleic acid-cleaving domains retaining the ability to cleave the complementary strand of the target nucleic acid, but reducing its ability to cleave the non-complementary strand of the target nucleic acid. The mutation can result in one or more of the plurality of nucleic acid-cleaving domains retaining the ability to cleave the non-complementary strand of the target nucleic acid, but reducing its ability to cleave the complementary strand of the target nucleic acid. For example, residues in the wild-type exemplary S. pyogenes Cas9 polypeptide, such as Asp 10, His840, Asn854 and Asn856, are mutated to inactivate one or more of the plurality of nucleic acid-cleaving domains (e.g., nuclease domains). The residues to be mutated can correspond to residues Asp 10, His840, Asn854 and Asn856 in the wild-type exemplary S. pyogenes Cas9 polypeptide (e.g., as determined by sequence and/or structural alignment). Non-limiting examples of mutations include D10A, H840A, N854A or N856A. One skilled in the art will recognize that mutations other than alanine substitutions can be suitable.
[00088] In some aspects, a D10A mutation can be combined with one or more of H840A, N854A, or N856A mutations to produce a site-directed polypeptide substantially lacking DNA cleavage activity. A H840A mutation can be combined with one or more of D10A, N854A, or N856A mutations to produce a site-directed polypeptide substantially lacking DNA cleavage activity. A N854A mutation can be combined with one or more of H840A, D10A, or N856A mutations to produce a site-directed polypeptide substantially lacking DNA cleavage activity. A N856A mutation can be combined with one or more of H840A, N854A, or D10A mutations to produce a site-directed polypeptide substantially lacking DNA cleavage activity. Site-directed polypeptides that comprise one substantially inactive nuclease domain are referred to as
"nickases."
[00089] Nickase variants of RNA-guided endonucleases, for example Cas9, can be used to increase the specificity of CRISPR-mediated genome editing. Wild type Cas9 is typically guided by a single guide RNA designed to hybridize with a specified -20 nucleotide sequence in the target sequence (such as an endogenous genomic locus). However, several mismatches can be tolerated between the guide RNA and the target locus, effectively reducing the length of required homology in the target site to, for example, as little as 13 nt of homology, and thereby resulting in elevated potential for binding and double-strand nucleic acid cleavage by the CRISPR/Cas9 complex elsewhere in the target genome - also known as off-target cleavage. Because nickase variants of Cas9 each only cut one strand, in order to create a double-strand break it is necessary for a pair of nickases to bind in close proximity and on opposite strands of the target nucleic acid, thereby creating a pair of nicks, which is the equivalent of a double-strand break. This requires that two separate guide RNAs - one for each nickase - must bind in close proximity and on opposite strands of the target nucleic acid. This requirement essentially doubles the minimum length of homology needed for the double-strand break to occur, thereby reducing the likelihood that a double-strand cleavage event will occur elsewhere in the genome, where the two guide RNA sites - if they exist - are unlikely to be sufficiently close to each other to enable the double-strand break to form. As described in the art, nickases can also be used to promote HDR versus NHEJ. HDR can be used to introduce selected changes into target sites in the genome through the use of specific donor sequences that effectively mediate the desired changes.
[00090] Mutations contemplated can include substitutions, additions, and deletions, or any combination thereof. The mutation converts the mutated amino acid to alanine. The mutation converts the mutated amino acid to another amino acid (e.g., glycine, serine, threonine, cysteine, valine, leucine, isoleucine, methionine, proline, phenylalanine, tyrosine, tryptophan, aspartic acid, glutamic acid, asparagine, glutamine, histidine, lysine, or arginine). The mutation converts the mutated amino acid to a non-natural amino acid (e.g., selenomethionine). The mutation converts the mutated amino acid to amino acid mimics (e.g., phosphomimics). The mutation can be a conservative mutation. For example, the mutation converts the mutated amino acid to amino acids that resemble the size, shape, charge, polarity, conformation, and/or rotamers of the mutated amino acids (e.g., cysteine/serine mutation, lysine/asparagine mutation,
histidine/phenylalanine mutation). The mutation can cause a shift in reading frame and/or the creation of a premature stop codon. Mutations can cause changes to regulatory regions of genes or loci that affect expression of one or more genes.
[00091] The site-directed polypeptide (e.g., variant, mutated, enzymatically inactive and/or conditionally enzymatically inactive site-directed polypeptide) can target nucleic acid. The site- directed polypeptide (e.g., variant, mutated, enzymatically inactive and/or conditionally enzymatically inactive endoribonuclease) can target DNA. The site-directed polypeptide (e.g., variant, mutated, enzymatically inactive and/or conditionally enzymatically inactive
endoribonuclease) can target RNA.
[00092] The site-directed polypeptide can comprise one or more non-native sequences (e.g., the site-directed polypeptide is a fusion protein).
[00093] The site-directed polypeptide can comprise an amino acid sequence comprising at least 15% amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), a nucleic acid binding domain, and two nucleic acid cleaving domains (i.e., a HNH domain and a RuvC domain).
[00094] The site-directed polypeptide can comprise an amino acid sequence comprising at least 15% amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), and two nucleic acid cleaving domains (i.e., a HNH domain and a RuvC domain).
[00095] The site-directed polypeptide can comprise an amino acid sequence comprising at least 15%) amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), and two nucleic acid cleaving domains, wherein one or both of the nucleic acid cleaving domains comprise at least 50%) amino acid identity to a nuclease domain from Cas9 from a bacterium (e.g., S.
pyogenes).
[00096] The site-directed polypeptide can comprise an amino acid sequence comprising at least 15%) amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), two nucleic acid cleaving domains (i.e., a HNH domain and a RuvC domain), and a non-native sequence (for example, a nuclear localization signal) or a linker linking the site-directed polypeptide to a non- native sequence.
[00097] The site-directed polypeptide can comprise an amino acid sequence comprising at least 15%o amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), two nucleic acid cleaving domains (i.e., a HNH domain and a RuvC domain), wherein the site-directed polypeptide comprises a mutation in one or both of the nucleic acid cleaving domains that reduces the cleaving activity of the nuclease domains by at least 50%.
[00098] The site-directed polypeptide can comprise an amino acid sequence comprising at least 15%o amino acid identity to a Cas9 from a bacterium (e.g., S. pyogenes), and two nucleic acid cleaving domains (i.e., a HNH domain and a RuvC domain), wherein one of the nuclease domains comprises mutation of aspartic acid 10, and/or wherein one of the nuclease domains can comprise a mutation of histidine 840, and wherein the mutation reduces the cleaving activity of the nuclease domain(s) by at least 50%. [00099] The one or more site-directed polypeptides, e.g. DNA endonucleases, can comprise two nickases that together effect one double-strand break at a specific locus in the genome, or four nickases that together effect or cause two double-strand breaks at specific loci in the genome. Alternatively, one site-directed polypeptide, e.g. DNA endonuclease, can effect or cause one double-strand break at a specific locus in the genome.
[000100] Non-limiting examples of Cas9 orthologs from other bacterial strains include but are not limited to, Cas proteins identified in Acaryochloris marina MBIC11017; Acetohalobium arabaticum DSM 5501; Acidithiobacillus caldus; Acidithiobacillus ferrooxidans ATCC 23270; Alicyclobacillus acidocaldarius LAAl; Alicyclobacillus acidocaldarius subsp. acidocaldarius DSM 446; Allochromatium vinosum DSM 180; Ammonifex degensii KC4; Anabaena variabilis ATCC 29413; Arthrospira maxima CS-328; Arthrospira platensis sir. Paraca; Arthrospira sp. PCC 8005; Bacillus pseudomycoides DSM 12442; Bacillus selenitireducens MLS10;
Burkholderiales bacterium 1 1 47; Caldicelulosiruptor becscii DSM 6725; Candidatus Desulforudis audaxviator MP104C; Caldicellulosiruptor hydrothermalis 108; Clostridium phage c-st; Clostridium botulinum A3 str. Loch Ma -ee; Clostridium botulinum Ba4 str. 657; Clostridium difficile QCD-63q42; Crocosphaera watsonii WH 8501; Cyanothece sp. ATCC 51142; Cyanothece sp. CCY0110; Cyanothece sp. PCC 7424; Cyanothece sp. PCC 7822;
Exiguobacterium sibiricum 255-15; Finegoldia magna ATCC 29328; Ktedonobacter racemifer DSM 44963; Lactobacillus delbrueckii subsp. bulgaricus PB2003/044-T3-4; Lactobacillus salivarius ATCC 11741; Listeria innocua; Lyngbya sp. PCC 8106; Marinobacter sp. ELB17; Methanohalobium evestigatum Z-7303; Microcystis phage Ma-LMMO 1 ; Microcystis aeruginosa NIES-843 ; Microscilla marina ATCC 23134; Microcoleus chthonoplastes PCC 7420; Neisseria meningitidis; Nitrosococcus halophilus Nc4; Nocardiopsis dassonvillei subsp. dassonvillei DSM 43111; Nodularia spumigena CCY9414; Nostoc sp. PCC 7120; Oscillatoria sp. PCC 6506; Pelotomaculum thermopropionicum SI; Petrotoga mobilis SJ95; Polar omonas
naphthalenivorans CJ2; Polaromonas sp. JS666; Pseudoalter omonas haloplanktis TAC125; Streptomyces pristinaespiralis ATCC 25486; Streptomyces pristinaespiralis ATCC 25486; Streptococcus thermophilus; Streptomyces viridochromogenes DSM 40736; Streptosporangium roseum DSM 43021; Synechococcus sp. PCC 7335; and Thermosipho africanus TCF52B (Chylinski et al., RNA Biol, 2013; 10(5): 726-737).
[000101] In addition to Cas9 orthologs, other Cas9 variants such as fusion proteins of inactive dCas9 and effector domains with different functions may be served as a platform for genetic modulation. Any of the foregoing enzymes may be useful in the present disclosure. [000102] Further examples of endonucleases that may be utilized in the present disclosure are given in SEQ ID NOs: 1-620. These proteins may be modified before use or may be encoded in a nucleic acid sequence such as a DNA, RNA or mRNA or within a vector construct such as the plasmids or AAV vectors taught herein. Further, they may be codon optimized.
[000103] SEQ ID NOs: 1-620 disclose a non-exhaustive listing of endonuclease sequences. Genome-targeting Nucleic Acid
[000104] The present disclosure provides a genome-targeting nucleic acid that can direct the activities of an associated polypeptide {e.g., a site-directed polypeptide) to a specific target sequence within a target nucleic acid. The genome-targeting nucleic acid can be an RNA. A genome-targeting RNA is referred to as a "guide RNA" or "gRNA" herein. A guide RNA can comprise at least a spacer sequence that hybridizes to a target nucleic acid sequence of interest, and a CRISPR repeat sequence. In Type II systems, the gRNA also comprises a second RNA called the tracrRNA sequence. In the Type II guide RNA (gRNA), the CRISPR repeat sequence and tracrRNA sequence hybridize to each other to form a duplex. In the Type V guide RNA (gRNA), the crRNA forms a duplex. In both systems, the duplex can bind a site-directed polypeptide, such that the guide RNA and site-direct polypeptide form a complex. The genome- targeting nucleic acid can provide target specificity to the complex by virtue of its association with the site-directed polypeptide. The genome-targeting nucleic acid thus can direct the activity of the site-directed polypeptide.
[000105] Exemplary guide RNAs include the spacer sequences in SEQ ID NOs: 5305-56902 of the Sequence Listing. As is understood by the person of ordinary skill in the art, each guide RNA can be designed to include a spacer sequence complementary to its genomic target sequence. For example, each of the spacer sequences in SEQ ID NOs: 5305-56902 of the Sequence Listing can be put into a single RNA chimera or a crRNA (along with a corresponding tracrRNA). See Jinek et al, Science, 337, 816-821 (2012) and Deltcheva et al, Nature, 471, 602-607 (2011).
[000106] The genome-targeting nucleic acid can be a double-molecule guide RNA. The genome-targeting nucleic acid can be a single-molecule guide RNA.
[000107] A double-molecule guide RNA can comprise two strands of RNA. The first strand comprises in the 5' to 3' direction, an optional spacer extension sequence, a spacer sequence and a minimum CRISPR repeat sequence. The second strand can comprise a minimum tracrRNA sequence (complementary to the minimum CRISPR repeat sequence), a 3' tracrRNA sequence and an optional tracrRNA extension sequence. [000108] A single-molecule guide RNA (sgRNA) in a Type II system can comprise, in the 5' to 3' direction, an optional spacer extension sequence, a spacer sequence, a minimum CRISPR repeat sequence, a single-molecule guide linker, a minimum tracrRNA sequence, a 3 ' tracrRNA sequence and an optional tracrRNA extension sequence. The optional tracrRNA extension can comprise elements that contribute additional functionality (e.g., stability) to the guide RNA. The single-molecule guide linker can link the minimum CRISPR repeat and the minimum tracrRNA sequence to form a hairpin structure. The optional tracrRNA extension can comprise one or more hairpins.
[000109] The sgRNA can comprise a 20 nucleotide spacer sequence at the 5' end of the sgRNA sequence. The sgRNA can comprise a less than a 20 nucleotide spacer sequence at the 5' end of the sgRNA sequence. The sgRNA can comprise a more than 20 nucleotide spacer sequence at the 5' end of the sgRNA sequence. The sgRNA can comprise a variable length spacer sequence with 17-30 nucleotides at the 5' end of the sgRNA sequence (see Table 1).
[000110] The sgRNA can comprise no uracil at the 3 'end of the sgRNA sequence, such as in SEQ ID NO: 56935 of Table 1. The sgRNA can comprise one or more uracil at the 3 'end of the sgRNA sequence, such as in SEQ ID NO: 56936 in Table 1. For example, the sgRNA can comprise 1 uracil (U) at the 3 ' end of the sgRNA sequence. The sgRNA can comprise 2 uracil (UU) at the 3 ' end of the sgRNA sequence. The sgRNA can comprise 3 uracil (UUU) at the 3 ' end of the sgRNA sequence. The sgRNA can comprise 4 uracil (UUUU) at the 3 ' end of the sgRNA sequence. The sgRNA can comprise 5 uracil (UUUUU) at the 3 ' end of the sgRNA sequence. The sgRNA can comprise 6 uracil (UUUUUU) at the 3 ' end of the sgRNA sequence. The sgRNA can comprise 7 uracil (ULJLJUUUU) at the 3 ' end of the sgRNA sequence. The sgRNA can comprise 8 uracil (UULJLJUUUU) at the 3 ' end of the sgRNA sequence.
[000111] The sgRNA can be unmodified or modified. For example, modified sgRNAs can comprise one or more 2'-0-methyl phosphorothioate nucleotides.
Table 1
SEQ ID NO. sgRNA sequence
56934 nnnnnnnnnnnnnnnnnnnnguuuuagagcuagaaauagcaaguuaaaauaaggcuaguccguuaucaacuuga aaaaguggcaccgagucggugcuuuu 56935 nnnnnnnnnnnnnnnnnnnnguuuuagagcuagaaauagcaaguuaaaauaaggcuaguccguuaucaacuuga aaaaguggcaccgagucggugc
56936 n(i7_30)guuuuagagcuagaaauagcaaguuaaaauaaggcuaguccguuaucaacuugaaaaagu
ggcaccgagucggugcu(i_8)
[000112] A single-molecule guide RNA (sgRNA) in a Type V system can comprise, in the 5' to 3' direction, a minimum CRISPR repeat sequence and a spacer sequence.
[000113] By way of illustration, guide RNAs used in the CRISPR/Cas9 or CRISPR/Cpfl system, or other smaller RNAs can be readily synthesized by chemical means, as illustrated below and described in the art. While chemical synthetic procedures are continually expanding, purifications of such RNAs by procedures such as high performance liquid chromatography (HPLC, which avoids the use of gels such as PAGE) tends to become more challenging as polynucleotide lengths increase significantly beyond a hundred or so nucleotides. One approach used for generating RNAs of greater length is to produce two or more molecules that are ligated together. Much longer RNAs, such as those encoding a Cas9 or Cpf 1 endonuclease, are more readily generated enzymatically. Various types of RNA modifications can be introduced during or after chemical synthesis and/or enzymatic generation of RNAs, e.g., modifications that enhance stability, reduce the likelihood or degree of innate immune response, and/or enhance other attributes, as described in the art.
Spacer Extension Sequence
[000114] In some examples of genome-targeting nucleic acids, a spacer extension sequence can modify activity, provide stability and/or provide a location for modifications of a genome- targeting nucleic acid. A spacer extension sequence can modify on- or off-target activity or specificity. In some examples, a spacer extension sequence can be provided. The spacer extension sequence can have a length of more than 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 220, 240, 260, 280, 300, 320, 340, 360, 380, 400, 1000, 2000, 3000, 4000, 5000, 6000, or 7000 or more nucleotides. The spacer extension sequence can have a length of less than 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 220, 240, 260, 280, 300, 320, 340, 360, 380, 400, 1000, 2000, 3000, 4000, 5000, 6000, 7000 or more nucleotides. The spacer extension sequence can be less than 10 nucleotides in length. The spacer extension sequence can be between 10-30 nucleotides in length. The spacer extension sequence can be between 30-70 nucleotides in length. [000115] The spacer extension sequence can comprise another moiety (e.g., a stability control sequence, an endoribonuclease binding sequence, a ribozyme). The moiety can decrease or increase the stability of a nucleic acid targeting nucleic acid. The moiety can be a transcriptional terminator segment (i.e., a transcription termination sequence). The moiety can function in a eukary otic cell. The moiety can function in a prokary otic cell. The moiety can function in both eukaryotic and prokaryotic cells. Non-limiting examples of suitable moieties include: a 5' cap (e.g., a 7-methylguanylate cap (m7 G)), a riboswitch sequence (e.g., to allow for regulated stability and/or regulated accessibility by proteins and protein complexes), a sequence that forms a dsRNA duplex (i.e., a hairpin), a sequence that targets the RNA to a subcellular location (e.g., nucleus, mitochondria, chloroplasts, and the like), a modification or sequence that provides for tracking (e.g., direct conjugation to a fluorescent molecule, conjugation to a moiety that facilitates fluorescent detection, a sequence that allows for fluorescent detection, etc.), and/or a modification or sequence that provides a binding site for proteins (e.g., proteins that act on DNA, including transcriptional activators, transcriptional repressors, DNA methyltransferases, DNA demethylases, histone acetyltransferases, histone deacetylases, and the like).
Spacer Sequence
[000116] The spacer sequence hybridizes to a sequence in a target nucleic acid of interest. The spacer of a genome-targeting nucleic acid can interact with a target nucleic acid in a sequence- specific manner via hybridization (i.e., base pairing). The nucleotide sequence of the spacer can vary depending on the sequence of the target nucleic acid of interest.
[000117] In a CRISPR/Cas system herein, the spacer sequence can be designed to hybridize to a target nucleic acid that is located 5' of a PAM of the Cas9 enzyme used in the system. The spacer may perfectly match the target sequence or may have mismatches. Each Cas9 enzyme has a particular PAM sequence that it recognizes in a target DNA. For example, S. pyogenes recognizes in a target nucleic acid a PAM that comprises the sequence 5'-NRG-3', where R comprises either A or G, where N is any nucleotide and N is immediately 3' of the target nucleic acid sequence targeted by the spacer sequence.
[000118] The target nucleic acid sequence can comprise 20 nucleotides. The target nucleic acid can comprise less than 20 nucleotides. The target nucleic acid can comprise more than 20 nucleotides. The target nucleic acid can comprise at least: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides. The target nucleic acid can comprise at most: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides. The target nucleic acid sequence can comprise 20 bases immediately 5' of the first nucleotide of the PAM. For example, in a sequence comprising 5'-NNNNNNNNNNNNNNNNNNNNNRG-3' (SEQ ID NO: 56933), the target nucleic acid can comprise the sequence that corresponds to the Ns, wherein N is any nucleotide, and the underlined NRG sequence is the S. pyogenes PAM. This target nucleic acid sequence is often referred to as the PAM strand, and the complementary nucleic acid sequence is often referred to the non-PAM strand. One of skill in the art would recognize that the spacer sequence hybridizes to the non-PAM strand of the target nucleic acid (Figures 1 A and IB).
[000119] The spacer sequence that hybridizes to the target nucleic acid can have a length of at least about 6 nucleotides (nt). The spacer sequence can be at least about 6 nt, at least about 10 nt, at least about 15 nt, at least about 18 nt, at least about 19 nt, at least about 20 nt, at least about 25 nt, at least about 30 nt, at least about 35 nt or at least about 40 nt, from about 6 nt to about 80 nt, from about 6 nt to about 50 nt, from about 6 nt to about 45 nt, from about 6 nt to about 40 nt, from about 6 nt to about 35 nt, from about 6 nt to about 30 nt, from about 6 nt to about 25 nt, from about 6 nt to about 20 nt, from about 6 nt to about 19 nt, from about 10 nt to about 50 nt, from about 10 nt to about 45 nt, from about 10 nt to about 40 nt, from about 10 nt to about 35 nt, from about 10 nt to about 30 nt, from about 10 nt to about 25 nt, from about 10 nt to about 20 nt, from about 10 nt to about 19 nt, from about 19 nt to about 25 nt, from about 19 nt to about 30 nt, from about 19 nt to about 35 nt, from about 19 nt to about 40 nt, from about 19 nt to about 45 nt, from about 19 nt to about 50 nt, from about 19 nt to about 60 nt, from about 20 nt to about 25 nt, from about 20 nt to about 30 nt, from about 20 nt to about 35 nt, from about 20 nt to about 40 nt, from about 20 nt to about 45 nt, from about 20 nt to about 50 nt, or from about 20 nt to about 60 nt. In some examples, the spacer sequence can comprise 20 nucleotides. In some examples, the spacer can comprise 19 nucleotides. In some examples, the spacer can comprise 18 nucleotides. In some examples, the spacer can comprise 22 nucleotides.
[000120] In some examples, the percent complementarity between the spacer sequence and the target nucleic acid is at least about 30%, at least about 40%, at least about 50%, at least about 60%), at least about 65%>, at least about 70%, at least about 75%, at least about 80%, at least about 85%), at least about 90%, at least about 95%, at least about 97%, at least about 98%, at least about 99%, or 100%. In some examples, the percent complementarity between the spacer sequence and the target nucleic acid is at most about 30%, at most about 40%, at most about 50%), at most about 60%, at most about 65%, at most about 70%, at most about 75%, at most about 80%), at most about 85%, at most about 90%, at most about 95%, at most about 97%, at most about 98%, at most about 99%, or 100%. In some examples, the percent complementarity between the spacer sequence and the target nucleic acid is 100% over the six contiguous 5'-most nucleotides of the target sequence of the complementary strand of the target nucleic acid. The percent complementarity between the spacer sequence and the target nucleic acid can be at least 60% over about 20 contiguous nucleotides. The length of the spacer sequence and the target nucleic acid can differ by 1 to 6 nucleotides, which may be thought of as a bulge or bulges.
[000121] The spacer sequence can be designed or chosen using a computer program. The computer program can use variables, such as predicted melting temperature, secondary structure formation, predicted annealing temperature, sequence identity, genomic context, chromatin accessibility, % GC, frequency of genomic occurrence (e.g., of sequences that are identical or are similar but vary in one or more spots as a result of mismatch, insertion or deletion), methylation status, presence of S Ps, and the like.
Minimum CRISPR Repeat Sequence
[000122] In some aspects, a minimum CRISPR repeat sequence is a sequence with at least about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%), about 90%, about 95%, or 100% sequence identity to a reference CRISPR repeat sequence (e.g. , crRNA from S. pyogenes).
[000123] In some aspects, a minimum CRISPR repeat sequence comprises nucleotides that can hybridize to a minimum tracrRNA sequence in a cell. The minimum CRISPR repeat sequence and a minimum tracrRNA sequence can form a duplex, i.e. a base-paired double-stranded structure. Together, the minimum CRISPR repeat sequence and the minimum tracrRNA sequence can bind to the site-directed polypeptide. At least a part of the minimum CRISPR repeat sequence can hybridize to the minimum tracrRNA sequence. At least a part of the minimum CRISPR repeat sequence can comprise at least about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or 100%) complementary to the minimum tracrRNA sequence. In some aspects, at least a part of the minimum CRISPR repeat sequence comprises at most about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or 100% complementary to the minimum tracrRNA sequence.
[000124] The minimum CRISPR repeat sequence can have a length from about 7 nucleotides to about 100 nucleotides. For example, the length of the minimum CRISPR repeat sequence is from about 7 nucleotides (nt) to about 50 nt, from about 7 nt to about 40 nt, from about 7 nt to about 30 nt, from about 7 nt to about 25 nt, from about 7 nt to about 20 nt, from about 7 nt to about 15 nt, from about 8 nt to about 40 nt, from about 8 nt to about 30 nt, from about 8 nt to about 25 nt, from about 8 nt to about 20 nt, from about 8 nt to about 15 nt, from about 15 nt to about 100 nt, from about 15 nt to about 80 nt, from about 15 nt to about 50 nt, from about 15 nt to about 40 nt, from about 15 nt to about 30 nt, or from about 15 nt to about 25 nt. In some aspects, the minimum CRISPR repeat sequence is approximately 9 nucleotides in length. In some aspects, the minimum CRISPR repeat sequence is approximately 12 nucleotides in length.
[000125] The minimum CRISPR repeat sequence can be at least about 60% identical to a reference minimum CRISPR repeat sequence (e.g., wild-type crRNA from S. pyogenes) over a stretch of at least 6, 7, or 8 contiguous nucleotides. For example, the minimum CRISPR repeat sequence can be at least about 65% identical, at least about 70% identical, at least about 75% identical, at least about 80% identical, at least about 85% identical, at least about 90% identical, at least about 95% identical, at least about 98% identical, at least about 99% identical or 100% identical to a reference minimum CRISPR repeat sequence over a stretch of at least 6, 7, or 8 contiguous nucleotides.
Minimum tracrRNA Sequence
[000126] A minimum tracrRNA sequence can be a sequence with at least about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%), about 95%), or 100%> sequence identity to a reference tracrRNA sequence (e.g., wild type tracrRNA from S. pyogenes).
[000127] A minimum tracrRNA sequence can comprise nucleotides that hybridize to a minimum CRISPR repeat sequence in a cell. A minimum tracrRNA sequence and a minimum CRISPR repeat sequence form a duplex, i.e. a base-paired double-stranded structure. Together, the minimum tracrRNA sequence and the minimum CRISPR repeat can bind to a site-directed polypeptide. At least a part of the minimum tracrRNA sequence can hybridize to the minimum CRISPR repeat sequence. The minimum tracrRNA sequence can be at least about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%), about 95%, or 100% complementary to the minimum CRISPR repeat sequence.
[000128] The minimum tracrRNA sequence can have a length from about 7 nucleotides to about 100 nucleotides. For example, the minimum tracrRNA sequence can be from about 7 nucleotides (nt) to about 50 nt, from about 7 nt to about 40 nt, from about 7 nt to about 30 nt, from about 7 nt to about 25 nt, from about 7 nt to about 20 nt, from about 7 nt to about 15 nt, from about 8 nt to about 40 nt, from about 8 nt to about 30 nt, from about 8 nt to about 25 nt, from about 8 nt to about 20 nt, from about 8 nt to about 15 nt, from about 15 nt to about 100 nt, from about 15 nt to about 80 nt, from about 15 nt to about 50 nt, from about 15 nt to about 40 nt, from about 15 nt to about 30 nt or from about 15 nt to about 25 nt long. The minimum tracrRNA sequence can be approximately 9 nucleotides in length. The minimum tracrRNA sequence can be approximately 12 nucleotides. The minimum tracrRNA can consist of tracrRNA nt 23-48 described in Jinek et al, supra.
[000129] The minimum tracrRNA sequence can be at least about 60% identical to a reference minimum tracrRNA {e.g., wild type, tracrRNA from S. pyogenes) sequence over a stretch of at least 6, 7, or 8 contiguous nucleotides. For example, the minimum tracrRNA sequence can be at least about 65% identical, about 70% identical, about 75% identical, about 80% identical, about 85%) identical, about 90% identical, about 95% identical, about 98% identical, about 99% identical or 100% identical to a reference minimum tracrRNA sequence over a stretch of at least 6, 7, or 8 contiguous nucleotides.
[000130] The duplex between the minimum CRISPR RNA and the minimum tracrRNA can comprise a double helix. The duplex between the minimum CRISPR RNA and the minimum tracrRNA can comprise at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nucleotides. The duplex between the minimum CRISPR RNA and the minimum tracrRNA can comprise at most about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more nucleotides.
[000131] The duplex can comprise a mismatch (i.e., the two strands of the duplex are not 100% complementary). The duplex can comprise at least about 1, 2, 3, 4, or 5 or mismatches. The duplex can comprise at most about 1, 2, 3, 4, or 5 or mismatches. The duplex can comprise no more than 2 mismatches.
Bulges
[000132] In some cases, there can be a "bulge" in the duplex between the minimum CRISPR RNA and the minimum tracrRNA. A bulge is an unpaired region of nucleotides within the duplex. A bulge can contribute to the binding of the duplex to the site-directed polypeptide. The bulge can comprise, on one side of the duplex, an unpaired 5'-XXXY-3' where X is any purine and Y comprises a nucleotide that can form a wobble pair with a nucleotide on the opposite strand, and an unpaired nucleotide region on the other side of the duplex. The number of unpaired nucleotides on the two sides of the duplex can be different.
[000133] In one example, the bulge can comprise an unpaired purine (e.g., adenine) on the minimum CRISPR repeat strand of the bulge. In some examples, the bulge can comprise an unpaired 5'-AAGY-3' of the minimum tracrRNA sequence strand of the bulge, where Y comprises a nucleotide that can form a wobble pairing with a nucleotide on the minimum CRISPR repeat strand. [000134] A bulge on the minimum CRISPR repeat side of the duplex can comprise at least 1, 2, 3, 4, or 5 or more unpaired nucleotides. A bulge on the minimum CRISPR repeat side of the duplex can comprise at most 1, 2, 3, 4, or 5 or more unpaired nucleotides. A bulge on the minimum CRISPR repeat side of the duplex can comprise 1 unpaired nucleotide.
[000135] A bulge on the minimum tracrRNA sequence side of the duplex can comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more unpaired nucleotides. A bulge on the minimum tracrRNA sequence side of the duplex can comprise at most 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more unpaired nucleotides. A bulge on a second side of the duplex (e.g., the minimum tracrRNA sequence side of the duplex) can comprise 4 unpaired nucleotides.
[000136] A bulge can comprise at least one wobble pairing. In some examples, a bulge can comprise at most one wobble pairing. A bulge can comprise at least one purine nucleotide. A bulge can comprise at least 3 purine nucleotides. A bulge sequence can comprise at least 5 purine nucleotides. A bulge sequence can comprise at least one guanine nucleotide. In some examples, a bulge sequence can comprise at least one adenine nucleotide.
Hairpins
[000137] In various examples, one or more hairpins can be located 3' to the minimum tracrRNA in the 3' tracrRNA sequence.
[000138] The hairpin can start at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 or more nucleotides 3' from the last paired nucleotide in the minimum CRISPR repeat and minimum tracrRNA sequence duplex. The hairpin can start at most about 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 or more nucleotides 3' of the last paired nucleotide in the minimum CRISPR repeat and minimum tracrRNA sequence duplex.
[000139] The hairpin can comprise at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 or more consecutive nucleotides. The hairpin can comprise at most about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or more consecutive nucleotides.
[000140] The hairpin can comprise a CC dinucleotide (i.e., two consecutive cytosine nucleotides).
[000141] The hairpin can comprise duplexed nucleotides (e.g., nucleotides in a hairpin, hybridized together). For example, a hairpin can comprise a CC dinucleotide that is hybridized to a GG dinucleotide in a hairpin duplex of the 3' tracrRNA sequence.
[000142] One or more of the hairpins can interact with guide RNA-interacting regions of a site- directed polypeptide. [000143] In some examples, there are two or more hairpins, and in other examples there are three or more hairpins.
3 ' tracrRNA sequence
[000144] A 3' tracrRNA sequence can comprise a sequence with at least about 30%, about 40%, about 50%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%), about 95%), or 100% sequence identity to a reference tracrRNA sequence (e.g., a tracrRNA from S. pyogenes).
[000145] The 3' tracrRNA sequence can have a length from about 6 nucleotides to about 100 nucleotides. For example, the 3' tracrRNA sequence can have a length from about 6 nucleotides (nt) to about 50 nt, from about 6 nt to about 40 nt, from about 6 nt to about 30 nt, from about 6 nt to about 25 nt, from about 6 nt to about 20 nt, from about 6 nt to about 15 nt, from about 8 nt to about 40 nt, from about 8 nt to about 30 nt, from about 8 nt to about 25 nt, from about 8 nt to about 20 nt, from about 8 nt to about 15 nt, from about 15 nt to about 100 nt, from about 15 nt to about 80 nt, from about 15 nt to about 50 nt, from about 15 nt to about 40 nt, from about 15 nt to about 30 nt, or from about 15 nt to about 25 nt. The 3' tracrRNA sequence can have a length of approximately 14 nucleotides.
[000146] The 3' tracrRNA sequence can be at least about 60%> identical to a reference 3' tracrRNA sequence (e.g., wild type 3' tracrRNA sequence from S. pyogenes) over a stretch of at least 6, 7, or 8 contiguous nucleotides. For example, the 3' tracrRNA sequence can be at least about 60%) identical, about 65%> identical, about 70% identical, about 75% identical, about 80% identical, about 85% identical, about 90% identical, about 95% identical, about 98% identical, about 99%) identical, or 100% identical, to a reference 3' tracrRNA sequence (e.g., wild type 3' tracrRNA sequence from S. pyogenes) over a stretch of at least 6, 7, or 8 contiguous nucleotides.
[000147] The 3' tracrRNA sequence can comprise more than one duplexed region (e.g., hairpin, hybridized region). The 3' tracrRNA sequence can comprise two duplexed regions.
[000148] The 3' tracrRNA sequence can comprise a stem loop structure. The stem loop structure in the 3' tracrRNA can comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20 or more nucleotides. The stem loop structure in the 3' tracrRNA can comprise at most 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 or more nucleotides. The stem loop structure can comprise a functional moiety. For example, the stem loop structure can comprise an aptamer, a ribozyme, a protein-interacting hairpin, a CRISPR array, an intron, or an exon. The stem loop structure can comprise at least about 1, 2, 3, 4, or 5 or more functional moieties. The stem loop structure can comprise at most about 1, 2, 3, 4, or 5 or more functional moieties. [000149] The hairpin in the 3' tracrRNA sequence can comprise a P-domain. In some examples, the P-domain can comprise a double-stranded region in the hairpin.
tracrRNA Extension Sequence
[000150] A tracrRNA extension sequence may be provided whether the tracrRNA is in the context of single-molecule guides or double-molecule guides. The tracrRNA extension sequence can have a length from about 1 nucleotide to about 400 nucleotides. The tracrRNA extension sequence can have a length of more than 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 220, 240, 260, 280, 300, 320, 340, 360, 380, or 400 nucleotides. The tracrRNA extension sequence can have a length from about 20 to about 5000 or more nucleotides. The tracrRNA extension sequence can have a length of more than 1000 nucleotides. The tracrRNA extension sequence can have a length of less than 1, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 220, 240, 260, 280, 300, 320, 340, 360, 380, 400 or more nucleotides. The tracrRNA extension sequence can have a length of less than 1000 nucleotides. The tracrRNA extension sequence can comprise less than 10 nucleotides in length. The tracrRNA extension sequence can be 10-30 nucleotides in length. The tracrRNA extension sequence can be 30-70 nucleotides in length.
[000151] The tracrRNA extension sequence can comprise a functional moiety {e.g., a stability control sequence, ribozyme, endoribonuclease binding sequence). The functional moiety can comprise a transcriptional terminator segment {i.e., a transcription termination sequence). The functional moiety can have a total length from about 10 nucleotides (nt) to about 100
nucleotides, from about 10 nt to about 20 nt, from about 20 nt to about 30 nt, from about 30 nt to about 40 nt, from about 40 nt to about 50 nt, from about 50 nt to about 60 nt, from about 60 nt to about 70 nt, from about 70 nt to about 80 nt, from about 80 nt to about 90 nt, or from about 90 nt to about 100 nt, from about 15 nt to about 80 nt, from about 15 nt to about 50 nt, from about 15 nt to about 40 nt, from about 15 nt to about 30 nt, or from about 15 nt to about 25 nt. The functional moiety can function in a eukaryotic cell. The functional moiety can function in a prokaryotic cell. The functional moiety can function in both eukaryotic and prokaryotic cells.
[000152] Non-limiting examples of suitable tracrRNA extension functional moieties include a 3' poly-adenylated tail, a riboswitch sequence {e.g., to allow for regulated stability and/or regulated accessibility by proteins and protein complexes), a sequence that forms a dsRNA duplex {i.e., a hairpin), a sequence that targets the RNA to a subcellular location {e.g., nucleus, mitochondria, chloroplasts, and the like), a modification or sequence that provides for tracking {e.g., direct conjugation to a fluorescent molecule, conjugation to a moiety that facilitates fluorescent detection, a sequence that allows for fluorescent detection, etc.), and/or a modification or sequence that provides a binding site for proteins (e.g., proteins that act on DNA, including transcriptional activators, transcriptional repressors, DNA methyltransferases, DNA demethylases, histone acetyltransferases, histone deacetylases, and the like). The tracrRNA extension sequence can comprise a primer binding site or a molecular index (e.g., barcode sequence). The tracrRNA extension sequence can comprise one or more affinity tags.
Single-Molecule Guide Linker Sequence
[000153] The linker sequence of a single-molecule guide nucleic acid can have a length from about 3 nucleotides to about 100 nucleotides. In Jinek et al, supra, for example, a simple 4 nucleotide "tetraloop" (-GAAA-) was used, Science, 337(6096):816-821 (2012). An illustrative linker has a length from about 3 nucleotides (nt) to about 90 nt, from about 3 nt to about 80 nt, from about 3 nt to about 70 nt, from about 3 nt to about 60 nt, from about 3 nt to about 50 nt, from about 3 nt to about 40 nt, from about 3 nt to about 30 nt, from about 3 nt to about 20 nt, from about 3 nt to about 10 nt. For example, the linker can have a length from about 3 nt to about 5 nt, from about 5 nt to about 10 nt, from about 10 nt to about 15 nt, from about 15 nt to about 20 nt, from about 20 nt to about 25 nt, from about 25 nt to about 30 nt, from about 30 nt to about 35 nt, from about 35 nt to about 40 nt, from about 40 nt to about 50 nt, from about 50 nt to about 60 nt, from about 60 nt to about 70 nt, from about 70 nt to about 80 nt, from about 80 nt to about 90 nt, or from about 90 nt to about 100 nt. The linker of a single-molecule guide nucleic acid can be between 4 and 40 nucleotides. The linker can be at least about 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, or 7000 or more nucleotides. The linker can be at most about 100, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, or 7000 or more nucleotides.
[000154] Linkers can comprise any of a variety of sequences, although in some examples the linker will not comprise sequences that have extensive regions of homology with other portions of the guide RNA, which might cause intramolecular binding that could interfere with other functional regions of the guide. In Jinek et al, supra, a simple 4 nucleotide sequence -GAAA- was used, Science, 337(6096):816-821 (2012), but numerous other sequences, including longer sequences can likewise be used.
[000155] The linker sequence can comprise a functional moiety. For example, the linker sequence can comprise one or more features, including an aptamer, a ribozyme, a protein- interacting hairpin, a protein binding site, a CRISPR array, an intron, or an exon. The linker sequence can comprise at least about 1, 2, 3, 4, or 5 or more functional moieties. In some examples, the linker sequence can comprise at most about 1, 2, 3, 4, or 5 or more functional moieties.
Nucleic acid modifications (chemical and structural modifications)
[000156] In some aspects, polynucleotides introduced into cells can comprise one or more modifications that can be used individually or in combination, for example, to enhance activity, stability or specificity, alter delivery, reduce innate immune responses in host cells, or for other enhancements, as further described herein and known in the art.
[000157] In certain examples, modified polynucleotides can be used in the CRISPR/Cas9 or CRISPR/Cpfl system, in which case the guide RNAs (either single-molecule guides or double- molecule guides) and/or a DNA or an RNA encoding a Cas9 or Cpfl endonuclease introduced into a cell can be modified, as described and illustrated below. Such modified polynucleotides can be used in the CRISPR/Cas9 or CRISPR/Cpfl system to edit any one or more genomic loci.
[000158] Using the CRISPR/Cas9 or CRISPR/Cpfl system for purposes of non-limiting illustrations of such uses, modifications of guide RNAs can be used to enhance the formation or stability of the CRISPR/Cas9 or CRISPR/Cpfl genome editing complex comprising guide RNAs, which can be single-molecule guides or double-molecule, and a Cas9 or Cpfl endonuclease. Modifications of guide RNAs can also or alternatively be used to enhance the initiation, stability or kinetics of interactions between the genome editing complex with the target sequence in the genome, which can be used, for example, to enhance on-target activity. Modifications of guide RNAs can also or alternatively be used to enhance specificity, e.g., the relative rates of genome editing at the on-target site as compared to effects at other (off-target) sites.
[000159] Modifications can also or alternatively be used to increase the stability of a guide RNA, e.g., by increasing its resistance to degradation by ribonucleases (RNases) present in a cell, thereby causing its half-life in the cell to be increased. Modifications enhancing guide RNA half-life can be particularly useful in aspects in which a Cas9 or Cpfl endonuclease is introduced into the cell to be edited via an RNA that needs to be translated in order to generate
endonuclease, because increasing the half-life of guide RNAs introduced at the same time as the RNA encoding the endonuclease can be used to increase the time that the guide RNAs and the encoded Cas9 or Cpfl endonuclease co-exist in the cell.
[000160] Modifications can also or alternatively be used to decrease the likelihood or degree to which RNAs introduced into cells elicit innate immune responses. Such responses, which have been well characterized in the context of RNA interference (RNAi), including small-interfering RNAs (siRNAs), as described below and in the art, tend to be associated with reduced half-life of the RNA and/or the elicitation of cytokines or other factors associated with immune responses.
[000161] One or more types of modifications can also be made to RNAs encoding an endonuclease that are introduced into a cell, including, without limitation, modifications that enhance the stability of the RNA (such as by increasing its degradation by RNases present in the cell), modifications that enhance translation of the resulting product (i.e. the endonuclease), and/or modifications that decrease the likelihood or degree to which the RNAs introduced into cells elicit innate immune responses.
[000162] Combinations of modifications, such as the foregoing and others, can likewise be used. In the case of CRISPR/Cas9 or CRISPR/Cpfl, for example, one or more types of modifications can be made to guide RNAs (including those exemplified above), and/or one or more types of modifications can be made to RNAs encoding Cas endonuclease (including those exemplified above).
[000163] By way of illustration, guide RNAs used in the CRISPR/Cas9 or CRISPR/Cpfl system, or other smaller RNAs can be readily synthesized by chemical means, enabling a number of modifications to be readily incorporated, as illustrated below and described in the art. While chemical synthetic procedures are continually expanding, purifications of such RNAs by procedures such as high-performance liquid chromatography (HPLC, which avoids the use of gels such as PAGE) tends to become more challenging as polynucleotide lengths increase significantly beyond a hundred or so nucleotides. One approach that can be used for generating chemically-modified RNAs of greater length is to produce two or more molecules that are ligated together. Much longer RNAs, such as those encoding a Cas9 endonuclease, are more readily generated enzymatically. While fewer types of modifications are available for use in
enzymatically produced RNAs, there are still modifications that can be used to, e.g., enhance stability, reduce the likelihood or degree of innate immune response, and/or enhance other attributes, as described further below and in the art; and new types of modifications are regularly being developed.
[000164] By way of illustration of various types of modifications, especially those used frequently with smaller chemically synthesized RNAs, modifications can comprise one or more nucleotides modified at the 2' position of the sugar, in some aspects a 2'-0-alkyl, 2'-0-alkyl-0- alkyl, or 2'-fluoro-modified nucleotide. In some examples, RNA modifications can include 2'- fluoro, 2'-amino or 2'-0-methyl modifications on the ribose of pyrimidines, abasic residues, or an inverted base at the 3' end of the RNA. Such modifications are routinely incorporated into oligonucleotides and these oligonucleotides have been shown to have a higher Tm (i.e., higher target binding affinity) than 2'-deoxyoligonucleotides against a given target.
[000165] A number of nucleotide and nucleoside modifications have been shown to make the oligonucleotide into which they are incorporated more resistant to nuclease digestion than the native oligonucleotide; these modified oligos survive intact for a longer time than unmodified oligonucleotides. Specific examples of modified oligonucleotides include those comprising modified backbones, for example, phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages. Some oligonucleotides are oligonucleotides with phosphorothioate backbones and those with heteroatom backbones, particularly CH2-NH-0-CH2,
CH,~N(CH3)~0~CH2 (known as a methylene(methylimino) or MMI backbone), CH2-0-N (CH3)-CH2, CH2 -N (CH3)-N (CH3)-CH2 and O-N (CH3)- CH2 -CH2 backbones, wherein the native phosphodiester backbone is represented as O- P- O- CH,); amide backbones [see De Mesmaeker et al., Ace. Chem. Res., 28:366-374 (1995)]; morpholino backbone structures (see Summerton and Weller, U.S. Patent No. 5,034,506); peptide nucleic acid (PNA) backbone (wherein the phosphodiester backbone of the oligonucleotide is replaced with a polyamide backbone, the nucleotides being bound directly or indirectly to the aza nitrogen atoms of the polyamide backbone, see Nielsen et al., Science 1991, 254, 1497). Phosphorus-containing linkages include, but are not limited to, phosphorothioates, chiral phosphorothioates,
phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates comprising 3'alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates comprising 3'-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and
boranophosphates having normal 3 -5' linkages, 2'-5' linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3'-5' to 5'-3' or 2'-5' to 5'-2'; see U.S. Patent Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5, 177, 196; 5, 188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321, 131; 5,399,676; 5,405,939; 5,453,496;
5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253;
5,571,799; 5,587,361; and 5,625,050.
[000166] Morpholino-based oligomeric compounds are described in Braasch and David Corey, Biochemistry, 41(14): 4503-4510 (2002); Genesis, Volume 30, Issue 3, (2001); Heasman, Dev. Biol., 243 : 209-214 (2002); Nasevicius et al., Nat. Genet., 26:216-220 (2000); Lacerra et al, Proc. Natl. Acad. Sci., 97: 9591-9596 (2000); and U.S. Patent No. 5,034,506, issued Jul. 23, 1991.
[000167] Cyclohexenyl nucleic acid oligonucleotide mimetics are described in Wang et al., J. Am. Chem. Soc, 122: 8595-8602 (2000).
[000168] Modified oligonucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These comprise those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S, and CH2 component parts; see U.S. Patent Nos. 5,034,506; 5,166,315; 5, 185,444; 5,214, 134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086;
5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312;
5,633,360; 5,677,437; and 5,677,439.
[000169] One or more substituted sugar moieties can also be included, e.g., one of the following at the 2' position: OH, SH, SCH3, F, OCN, OCH3 OCH3, OCH3 0(CH2)n CH3, 0(CH2)n NH2, or 0(CH2)n CH3, where n is from 1 to about 10; CI to CIO lower alkyl, alkoxyalkoxy, substituted lower alkyl, alkaryl or aralkyl; CI; Br; CN; CF3; OCF3; 0-, S-, or N- alkyl; 0-, S-, or N-alkenyl; SOCH3; S02 CH3; ON02; N02; N3; NH2; heterocycloalkyl;
heterocycloalkaryl; aminoalkylamino; polyalkylamino; substituted silyl; an RNA cleaving group; a reporter group; an intercalator; a group for improving the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties. In some aspects, a modification includes 2'- methoxyethoxy (2'-0-CH2CH2OCH3, also known as 2'-0-(2-methoxyethyl)) (Martin et al, Helv. Chim. Acta, 1995, 78, 486). Other modifications include 2'-methoxy (2'-0-CH3), 2'-propoxy (2'- OCH2 CH2CH3) and 2'-fluoro (2'-F). Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the 3' terminal nucleotide and the 5' position of 5' terminal nucleotide. Oligonucleotides may also have sugar mimetics, such as cyclobutyls in place of the pentofuranosyl group. [000170] In some examples, both a sugar and an internucleoside linkage, i.e., the backbone, of the nucleotide units can be replaced with novel groups. The base units can be maintained for hybridization with an appropriate nucleic acid target compound. One such oligomeric compound, an oligonucleotide mimetic that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA). In PNA compounds, the sugar- backbone of an oligonucleotide can be replaced with an amide containing backbone, for example, an aminoethylglycine backbone. The nucleobases can be retained and bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative U.S. patents that teach the preparation of PNA compounds comprise, but are not limited to, U.S. Patent Nos. 5,539,082; 5,714,331; and 5,719,262. Further teaching of PNA compounds can be found in Nielsen et al, Science, 254: 1497-1500 (1991).
[000171] Guide RNAs can also include, additionally or alternatively, nucleobase (often referred to in the art simply as "base") modifications or substitutions. As used herein, "unmodified" or "natural" nucleobases include adenine (A), guanine (G), thymine (T), cytosine (C), and uracil (U). Modified nucleobases include nucleobases found only infrequently or transiently in natural nucleic acids, e.g., hypoxanthine, 6-methyladenine, 5-Me pyrimidines, particularly 5- methylcytosine (also referred to as 5-methyl-2' deoxycytosine and often referred to in the art as 5-Me-C), 5-hydroxymethylcytosine (HMC), glycosyl HMC and gentobiosyl HMC, as well as synthetic nucleobases, e.g., 2-aminoadenine, 2-(methylamino) adenine, 2- (imidazolylalkyl)adenine, 2-(aminoalklyamino) adenine or other heterosubstituted alkyladenines, 2-thiouracil, 2-thiothymine, 5-bromouracil, 5-hydroxymethyluracil, 8-azaguanine, 7- deazaguanine, N6 (6-aminohexyl)adenine, and 2,6-diaminopurine. Kornberg, A., DNA
Replication, W. H. Freeman & Co., San Francisco, pp 75-77 (1980); Gebeyehu et al., Nucl. Acids Res. 15:4513 (1997). A "universal" base known in the art, e.g., inosine, can also be included. 5-Me-C substitutions have been shown to increase nucleic acid duplex stability by 0.6- 1.2 °C. (Sanghvi, Y. S., in Crooke, S. T. and Lebleu, B., eds., Antisense Research and
Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are aspects of base substitutions.
[000172] Modified nucleobases can comprise other synthetic and natural nucleobases, such as 5 -methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2- aminoadenine, 6-m ethyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5- halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5- uracil (pseudo-uracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8- thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5- bromo, 5-trifluoromethyl and other 5- substituted uracils and cytosines, 7-methylquanine and 7-methyladenine, 8-azaguanine and 8- azaadenine, 7-deazaguanine and 7-deazaadenine, and 3-deazaguanine and 3-deazaadenine.
[000173] Further, nucleobases can comprise those disclosed in U.S. Patent No. 3,687,808, those disclosed in 'The Concise Encyclopedia of Polymer Science And Engineering', pages 858- 859, Kroschwitz, J.I., ed. John Wiley & Sons, 1990, those disclosed by Englisch et al,
Angewandle Chemie, International Edition', 1991, 30, page 613, and those disclosed by Sanghvi, Y. S., Chapter 15, Antisense Research and Applications', pages 289- 302, Crooke, S.T. and Lebleu, B. ea., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the present disclosure. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, comprising 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2°C (Sanghvi, Y.S., Crooke, S.T. and Lebleu, B., eds, 'Antisense Research and Applications', CRC Press, Boca Raton, 1993, pp. 276-278) and are aspects of base substitutions, even more particularly when combined with 2'-0-methoxyethyl sugar modifications. Modified nucleobases are described in US Patent Nos. 3,687,808, as well as 4,845,205; 5,130,302; 5, 134,066; 5, 175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502, 177; 5,525,711; 5,552,540; 5,587,469;
5,596,091; 5,614,617; 5,681,941; 5,750,692; 5,763,588; 5,830,653; 6,005,096; and U.S. Patent Application Publication 2003/0158403.
[000174] Thus, the term "modified" refers to a non-natural sugar, phosphate, or base that is incorporated into a guide RNA, an endonuclease, or both a guide RNA and an endonuclease. It is not necessary for all positions in a given oligonucleotide to be uniformly modified, and in fact more than one of the aforementioned modifications can be incorporated in a single
oligonucleotide, or even in a single nucleoside within an oligonucleotide.
[000175] The guide RNAs and/or mRNA (or DNA) encoding an endonuclease can be chemically linked to one or more moieties or conjugates that enhance the activity, cellular distribution, or cellular uptake of the oligonucleotide. Such moieties comprise, but are not limited to, lipid moieties such as a cholesterol moiety [Letsinger et al, Proc. Natl. Acad. Sci. USA, 86: 6553-6556 (1989)]; cholic acid [Manoharan et al, Bioorg. Med. Chem. Let., 4: 1053- 1060 (1994)]; a thioether, e.g., hexyl-S- tritylthiol [Manoharan et al, Ann. N. Y. Acad. Sci., 660: 306-309 (1992) and Manoharan et al, Bioorg. Med. Chem. Let., 3 : 2765-2770 (1993)]; a thiocholesterol [Oberhauser et al, Nucl. Acids Res., 20: 533-538 (1992)]; an aliphatic chain, e.g., dodecandiol or undecyl residues [Kabanov et a/., FEBS Lett., 259: 327-330 (1990) and Svinarchuk et a/., Biochimie, 75: 49- 54 (1993)]; a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1 ,2-di-O-hexadecyl- rac-glycero-3-H-phosphonate [Manoharan et al., Tetrahedron Lett., 36: 3651-3654 (1995) and Shea et al, Nucl. Acids Res., 18: 3777-3783 (1990)]; a polyamine or a polyethylene glycol chain [Mancharan et al, Nucleosides &
Nucleotides, 14: 969-973 (1995)]; adamantane acetic acid [Manoharan et a/., Tetrahedron Lett., 36: 3651-3654 (1995)]; a palmityl moiety [(Mishra et al., Biochim. Biophys. Acta, 1264: 229- 237 (1995)]; or an octadecylamine or hexylamino-carbonyl-t oxycholesterol moiety [Crooke et al, J. Pharmacol. Exp. Ther., 277: 923-937 (1996)]. See also U.S. Patent Nos. 4,828,979;
4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717; 5,580,731;
5,580,731; 5,591,584; 5,109, 124; 5, 118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439;
5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941;
4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830;
5, 112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873;
5,317,098; 5,371,241; 5,391,723; 5,416,203; 5,451,463; 5,510,475; 5,512,667; 5,514,785;
5,565,552; 5,567,810; 5,574, 142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599, 928 and 5,688,941.
[000176] Sugars and other moieties can be used to target proteins and complexes comprising nucleotides, such as cationic polysomes and liposomes, to particular sites. For example, hepatic cell directed transfer can be mediated via asialoglycoprotein receptors (ASGPRs); see, e.g., Hu, et al, Protein Pept Lett. 21(10): 1025-30 (2014). Other systems known in the art and regularly developed can be used to target biomolecules of use in the present case and/or complexes thereof to particular target cells of interest.
[000177] These targeting moieties or conjugates can include conjugate groups covalently bound to functional groups, such as primary or secondary hydroxyl groups. Conjugate groups of the present disclosure include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers. Typical conjugate groups include cholesterols, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes. Groups that enhance the pharmacodynamic properties, in the context of this disclosure, include groups that improve uptake, enhance resistance to degradation, and/or strengthen sequence-specific hybridization with the target nucleic acid. Groups that enhance the pharmacokinetic properties, in the context of this present disclosure, include groups that improve uptake, distribution, metabolism or excretion of the compounds of the present disclosure. Representative conjugate groups are disclosed in International Patent Application No. PCT/US92/09196, filed Oct. 23, 1992 (published as WO1993007883), and U.S. Patent No. 6,287,860. Conjugate moieties include, but are not limited to, lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g., hexyl-5-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues, a phospholipid, e.g., di-hexadecyl-rac- glycerol or triethylammonium 1,2-di-O- hexadecyl-rac-glycero-3-H-phosphonate, a polyamine or a polyethylene glycol chain, or adamantane acetic acid, a palmityl moiety, or an octadecylamine or hexylamino-carbonyl-oxy cholesterol moiety. See, e.g., U.S. Patent Nos. 4,828,979; 4,948,882; 5,218, 105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,580,731; 5,591,584; 5, 109,124;
5, 118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044;
4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582;
4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022;
5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241; 5,391,723;
5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142;
5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941.
[000178] Longer polynucleotides that are less amenable to chemical synthesis and are typically produced by enzymatic synthesis can also be modified by various means. Such modifications can include, for example, the introduction of certain nucleotide analogs, the incorporation of particular sequences or other moieties at the 5' or 3' ends of molecules, and other modifications. By way of illustration, the mRNA encoding Cas9 is approximately 4 kb in length and can be synthesized by in vitro transcription. Modifications to the mRNA can be applied to, e.g., increase its translation or stability (such as by increasing its resistance to degradation with a cell), or to reduce the tendency of the RNA to elicit an innate immune response that is often observed in cells following introduction of exogenous RNAs, particularly longer RNAs such as that encoding Cas9.
[000179] Numerous such modifications have been described in the art, such as polyA tails, 5' cap analogs {e.g., Anti Reverse Cap Analog (ARC A) or m7G(5')ppp(5')G (mCAP)), modified 5' or 3' untranslated regions (UTRs), use of modified bases (such as Pseudo-UTP, 2-Thio-UTP, 5- Methylcytidine-5'-Triphosphate (5-Methyl-CTP) or N6-Methyl-ATP), or treatment with phosphatase to remove 5' terminal phosphates. These and other modifications are known in the art, and new modifications of RNAs are regularly being developed. [000180] There are numerous commercial suppliers of modified RNAs, including for example, TriLink Biotech, AxoLabs, Bio-Synthesis Inc., Dharmacon and many others. As described by TriLink, for example, 5-Methyl-CTP can be used to impart desirable characteristics, such as increased nuclease stability, increased translation or reduced interaction of innate immune receptors with in vitro transcribed RNA. 5-Methylcytidine-5'-Triphosphate (5-Methyl-CTP), N6-Methyl-ATP, as well as Pseudo-UTP and 2-Thio-UTP, have also been shown to reduce innate immune stimulation in culture and in vivo while enhancing translation, as illustrated in publications by Kormann et al. and Warren et al. referred to below.
[000181] It has been shown that chemically modified mRNA delivered in vivo can be used to achieve improved therapeutic effects; see, e.g., Kormann et al., Nature Biotechnology 29, 154— 157 (2011). Such modifications can be used, for example, to increase the stability of the RNA molecule and/or reduce its immunogenicity. Using chemical modifications such as Pseudo-U, N6-Methyl-A, 2-Thio-U and 5-Methyl-C, it was found that substituting just one quarter of the uridine and cytidine residues with 2-Thio-U and 5-Methyl-C respectively resulted in a significant decrease in toll-like receptor (TLR) mediated recognition of the mRNA in mice. By reducing the activation of the innate immune system, these modifications can be used to effectively increase the stability and longevity of the mRNA in vivo; see, e.g., Kormann et al., supra.
[000182] It has also been shown that repeated administration of synthetic messenger RNAs incorporating modifications designed to bypass innate anti-viral responses can reprogram differentiated human cells to pluripotency. See, e.g., Warren, et al., Cell Stem Cell, 7(5):618-30 (2010). Such modified mRNAs that act as primary reprogramming proteins can be an efficient means of reprogramming multiple human cell types. Such cells are referred to as induced pluripotency stem cells (iPSCs), and it was found that enzymatically synthesized RNA incorporating 5-Methyl-CTP, Pseudo-UTP and an Anti Reverse Cap Analog (ARCA) could be used to effectively evade the cell's antiviral response; see, e.g., Warren et al., supra.
[000183] Other modifications of polynucleotides described in the art include, for example, the use of polyA tails, the addition of 5' cap analogs (such as m7G(5')ppp(5')G (mCAP)), modifications of 5' or 3' untranslated regions (UTRs), or treatment with phosphatase to remove 5' terminal phosphates - and new approaches are regularly being developed.
[000184] A number of compositions and techniques applicable to the generation of modified RNAs for use herein have been developed in connection with the modification of RNA interference (RNAi), including small-interfering RNAs (siRNAs). siRNAs present particular challenges in vivo because their effects on gene silencing via mRNA interference are generally transient, which can require repeat administration. In addition, siRNAs are double-stranded RNAs (dsRNA) and mammalian cells have immune responses that have evolved to detect and neutralize dsRNA, which is often a by-product of viral infection. Thus, there are mammalian enzymes such as PKR (dsRNA-responsive kinase), and potentially retinoic acid-inducible gene I (RIG-I), that can mediate cellular responses to dsRNA, as well as Toll-like receptors (such as TLR3, TLR7 and TLR8) that can trigger the induction of cytokines in response to such molecules; see, e.g., the reviews by Angart et al, Pharmaceuticals (Basel) 6(4): 440-468 (2013); Kanasty et al., Molecular Therapy 20(3): 513-524 (2012); Burnett et al, Biotechnol J.
6(9): 1130-46 (2011); Judge and MacLachlan, Hum Gene Ther 19(2): 111-24 (2008); and references cited therein.
[000185] A large variety of modifications have been developed and applied to enhance RNA stability, reduce innate immune responses, and/or achieve other benefits that can be useful in connection with the introduction of polynucleotides into human cells, as described herein; see, e.g., the reviews by Whitehead KA et al., Annual Review of Chemical and Biomolecular Engineering, 2: 77-96 (2011); Gaglione and Messere, Mini Rev Med Chem, 10(7):578-95
(2010); Chernolovskaya et al, Curr Opin Mol Ther., 12(2): 158-67 (2010); Deleavey et al, Curr Protoc Nucleic Acid Chem Chapter 16:Unit 16.3 (2009); Behlke, Oligonucleotides 18(4):305-19 (2008); Fucini et al, Nucleic Acid Ther 22(3): 205-210 (2012); Bremsen et al, Front Genet 3 : 154 (2012).
[000186] As noted above, there are a number of commercial suppliers of modified RNAs, many of which have specialized in modifications designed to improve the effectiveness of siRNAs. A variety of approaches are offered based on various findings reported in the literature. For example, Dharmacon notes that replacement of a non-bridging oxygen with sulfur
(phosphorothioate, PS) has been extensively used to improve nuclease resistance of siRNAs, as reported by Kole, Nature Reviews Drug Discovery 11 : 125-140 (2012). Modifications of the 2'- position of the ribose have been reported to improve nuclease resistance of the internucleotide phosphate bond while increasing duplex stability (Tm), which has also been shown to provide protection from immune activation. A combination of moderate PS backbone modifications with small, well -tolerated 2'-substitutions (2'-0-Methyl, 2'-Fluoro, 2'-Hydro) have been associated with highly stable siRNAs for applications in vivo, as reported by Soutschek et al. Nature 432: 173-178 (2004); and 2'-0-Methyl modifications have been reported to be effective in improving stability as reported by Volkov, Oligonucleotides 19: 191-202 (2009). With respect to decreasing the induction of innate immune responses, modifying specific sequences with 2'-0- Methyl, 2'-Fluoro, 2'-Hydro have been reported to reduce TLR7/TLR8 interaction while generally preserving silencing activity; see, e.g., Judge et al., Mol. Ther. 13 :494-505 (2006); and Cekaite et al., J. Mol. Biol. 365:90-108 (2007). Additional modifications, such as 2-thiouracil, pseudouracil, 5-methylcytosine, 5-methyluracil, and N6-methyladenosine have also been shown to minimize the immune effects mediated by TLR3, TLR7, and TLR8; see, e.g., Kariko, K. et al, Immunity 23 : 165-175 (2005).
[000187] As is also known in the art, and commercially available, a number of conjugates can be applied to polynucleotides, such as RNAs, for use herein that can enhance their delivery and/or uptake by cells, including for example, cholesterol, tocopherol and folic acid, lipids, peptides, polymers, linkers and aptamers; see, e.g., the review by Winkler, Ther. Deliv. 4:791- 809 (2013), and references cited therein.
Codon- Optimization
[000188] A polynucleotide encoding a site-directed polypeptide can be codon-optimized according to methods standard in the art for expression in the cell containing the target DNA of interest. For example, if the intended target nucleic acid is in a human cell, a human codon- optimized polynucleotide encoding Cas9 is contemplated for use for producing the Cas9 polypeptide.
Complexes of a Genome-targeting Nucleic Acid and a Site-Directed Polypeptide
[000189] A genome-targeting nucleic acid interacts with a site-directed polypeptide {e.g., a nucleic acid-guided nuclease such as Cas9), thereby forming a complex. The genome-targeting nucleic acid guides the site-directed polypeptide to a target nucleic acid.
Ribonucleoprotein complexes (RNPs)
[000190] The site-directed polypeptide and genome-targeting nucleic acid can each be administered separately to a cell or a patient. On the other hand, the site-directed polypeptide can be pre-complexed with one or more guide RNAs, or one or more crRNA together with a tracrRNA. The pre-complexed material can then be administered to a cell or a patient. Such pre-complexed material is known as a ribonucleoprotein particle (RNP). The site-directed polypeptide in the RNP can be, for example, a Cas9 endonuclease or a Cpf 1 endonuclease. The site-directed polypeptide can be flanked at the N-terminus, the C-terminus, or both the N- terminus and C-terminus by one or more nuclear localization signals (NLSs). For example, a Cas9 endonuclease can be flanked by two NLSs, one NLS located at the N-terminus and the second NLS located at the C-terminus. The NLS can be any NLS known in the art, such as a SV40 NLS. The weight ratio of genome-targeting nucleic acid to site-directed polypeptide in the RNP can be 1 : 1. For example, the weight ratio of sgRNA to Cas9 endonuclease in the RNP can be 1 : 1.
Nucleic Acids Encoding System Components
[000191] The present disclosure provides a nucleic acid comprising a nucleotide sequence encoding a genome-targeting nucleic acid of the disclosure, a site-directed polypeptide of the disclosure, and/or any nucleic acid or proteinaceous molecule necessary to carry out the aspects of the methods of the disclosure.
[000192] The nucleic acid encoding a genome-targeting nucleic acid of the disclosure, a site- directed polypeptide of the disclosure, and/or any nucleic acid or proteinaceous molecule necessary to carry out the aspects of the methods of the disclosure can comprise a vector {e.g., a recombinant expression vector).
[000193] The term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double-stranded DNA loop into which additional nucleic acid segments can be ligated. Another type of vector is a viral vector, wherein additional nucleic acid segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced {e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors {e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
[000194] In some examples, vectors can be capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "recombinant expression vectors", or more simply "expression vectors", which serve equivalent functions.
[000195] The term "operably linked" means that the nucleotide sequence of interest is linked to regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence. The term "regulatory sequence" is intended to include, for example, promoters, enhancers and other expression control elements {e.g., polyadenylation signals). Such regulatory sequences are well known in the art and are described, for example, in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cells, and those that direct expression of the nucleotide sequence only in certain host cells {e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the target cell, the level of expression desired, and the like.
[000196] Expression vectors contemplated include, but are not limited to, viral vectors based on vaccinia virus, poliovirus, adenovirus, adeno-associated virus, SV40, herpes simplex virus, human immunodeficiency virus, retrovirus (e.g., Murine Leukemia Virus, spleen necrosis virus, and vectors derived from retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, a lentivirus, human immunodeficiency virus, myeloproliferative sarcoma virus, and mammary tumor virus) and other recombinant vectors. Other vectors contemplated for eukaryotic target cells include, but are not limited to, the vectors pXTl, pSG5, pSVK3, pBPV, pMSG, and pSVLSV40 (Pharmacia). Other vectors can be used so long as they are compatible with the host cell.
[000197] In some examples, a vector can comprise one or more transcription and/or translation control elements. Depending on the host/vector system utilized, any of a number of suitable transcription and translation control elements, including constitutive and inducible promoters, transcription enhancer elements, transcription terminators, etc. can be used in the expression vector. The vector can be a self-inactivating vector that either inactivates the viral sequences or the components of the CRISPR machinery or other elements.
[000198] Non-limiting examples of suitable eukaryotic promoters (i.e., promoters functional in a eukaryotic cell) include those from cytomegalovirus (CMV) immediate early, herpes simplex virus (HSV) thymidine kinase, early and late SV40, long terminal repeats (LTRs) from retrovirus, human elongation factor-1 promoter (EF1), a hybrid construct comprising the cytomegalovirus (CMV) enhancer fused to the chicken beta-actin promoter (CAG), murine stem cell virus promoter (MSCV), phosphogly cerate kinase- 1 locus promoter (PGK), and mouse metallothionein-I.
[000199] For expressing small RNAs, including guide RNAs used in connection with Cas endonuclease, various promoters such as RNA polymerase III promoters, including for example U6 and HI, can be advantageous. Descriptions of and parameters for enhancing the use of such promoters are known in art, and additional information and approaches are regularly being described; see, e.g., Ma, H. et al, Molecular Therapy - Nucleic Acids 3, el61 (2014) doi: 10.1038/mtna.2014.12.
[000200] The expression vector can also contain a ribosome binding site for translation initiation and a transcription terminator. The expression vector can also comprise appropriate sequences for amplifying expression. The expression vector can also include nucleotide sequences encoding non-native tags (e.g., histidine tag, hemagglutinin tag, green fluorescent protein, etc.) that are fused to the site-directed polypeptide, thus resulting in a fusion protein.
[000201] A promoter can be an inducible promoter (e.g., a heat shock promoter, tetracycline- regulated promoter, steroid-regulated promoter, metal-regulated promoter, estrogen receptor- regulated promoter, etc.). The promoter can be a constitutive promoter (e.g., CMV promoter, UBC promoter). In some cases, the promoter can be a spatially restricted and/or temporally restricted promoter (e.g., a tissue specific promoter, a cell type specific promoter, etc.).
[000202] The nucleic acid encoding a genome-targeting nucleic acid of the disclosure and/or a site-directed polypeptide can be packaged into or on the surface of delivery vehicles for delivery to cells. Delivery vehicles contemplated include, but are not limited to, nanospheres, liposomes, quantum dots, nanoparticles, polyethylene glycol particles, hydrogels, and micelles. As described in the art, a variety of targeting moieties can be used to enhance the preferential interaction of such vehicles with desired cell types or locations.
[000203] Introduction of the complexes, polypeptides, and nucleic acids of the disclosure into cells can occur by viral or bacteriophage infection, transfection, conjugation, protoplast fusion, lipofection, electroporation, nucleofection, calcium phosphate precipitation, polyethyleneimine (PEI)-mediated transfection, DEAE-dextran mediated transfection, liposome-mediated transfection, particle gun technology, calcium phosphate precipitation, direct micro-injection, nanoparticle-mediated nucleic acid delivery, and the like.
Therapeutic approach
[000204] Provided herein are methods for treating a patient with Spinocerebellar ataxia 3 (SCA3). Spinocerebellar ataxia 3 (SCA3) is caused by abnormal expansion of a trinucleotide CAG repeat in exon 10 of the ATXN3 gene. In most people, the CAG segment is repeated 12 to 43 times. In patients with Spinocerebellar ataxia 3 (SCA3), the number of CAG repeats can be more than 50. People with intermediate numbers of the CAG repeat (about 44-52) may or may not develop the disease. This number can be different from one patient to another.
[000205] The term "trinucleotide repeat expansion" means a series of three bases (for example, CAG) repeated at least twice. In certain examples, the trinucleotide repeat expansion may be located in exon 10 of an ATXN3 nucleic acid. In certain examples, a pathogenic trinucleotide repeat expansion includes at least 50 repeats of CAG in an ATXN3 nucleic acid and is associated with disease. In other examples, a pathogenic trinucleotide repeat expansion includes at least 51, 52, 53, 54, 55, 60, 65, 70, 75, 80, 85 or more repeats. In certain examples, the repeats are consecutive. In certain examples, the repeats are interrupted by one or more nucleobases. In certain examples, a wild-type trinucleotide repeat expansion includes 43 or fewer repeats of CAG in an ATXN3 nucleic acid. In other examples, a wild-type trinucleotide repeat expansion includes at most 42, 41, 40, 39, 38, 37, 36, 35, 34, 33, 32, 31, 30, 25, 20, 15, or 12 repeats. In certain examples, the entire trinucleotide repeat expansion is deleted. In other examples, a portion of the trinucleotide repeat expansion is deleted.
[000206] Another aspect of such method is an ex vivo cell-based therapy. For example, a patient specific induced pluripotent stem cell (iPSC) can be created. Then, the chromosomal DNA of these iPS cells can be edited using the materials and methods described herein. Next, the genome-edited iPSCs can be differentiated into cells which are then implanted into the patient. In certain aspects, the iPSCs can be differentiated into cells of the central nervous system (e.g., neurons, glial, or retinal cells), cells of the peripheral nervous system or other periphral cell types, including cells of the cardiovascular system (e.g. : cardiac myocytes).
[000207] Yet another aspect of such method is an ex vivo cell-based therapy. For example, a mesenchymal stem cell can be isolated from the patient, which can be isolated from the patient's bone marrow or peripheral blood. Next, the chromosomal DNA of these mesenchymal stem cells can be edited using the materials and methods described herein. Next, the genome-edited mesenchymal stem cells can be differentiated into cells of the central nervous system (e.g., neurons or glial cells). Finally, the differentiated cells, e.g., cells of the central nervous system (e.g., neurons or glial cells) are implanted into the patient.
[000208] One advantage of an ex vivo cell therapy approach is the ability to conduct a comprehensive analysis of the therapeutic prior to administration. Nuclease-based therapeutics can have some level of off-target effects. Performing gene editing ex vivo allows one to characterize the edited cell population prior to implantation. The present disclosure includes sequencing the entire genome of the edited cells to ensure that the off-target effects, if any, can be in genomic locations associated with minimal risk to the patient. Furthermore, populations of specific cells, including clonal populations, can be isolated prior to implantation.
[000209] Another advantage of ex vivo cell therapy relates to genetic modification in iPSCs compared to other primary cell sources. iPSCs are prolific, making it easy to obtain the large number of cells that will be required for a cell-based therapy. Furthermore, iPSCs are an ideal cell type for performing clonal isolations. This allows screening for the correct genomic modification, without risking a decrease in viability. In contrast, other primary cells, such as glial cells, are viable for only a few passages and difficult to clonally expand. Thus, manipulation of iPSCs for the treatment of Spinocerebellar ataxia 3 (SCA3) can be much easier, and can shorten the amount of time needed to make the desired genetic modification.
[000210] Methods can also include an in vivo based therapy. Chromosomal DNA of the cells in the patient is edited using the materials and methods described herein. In some aspects, the target cell in an in vivo based therapy is a cell of the central nervous system (e.g., a neuron or a glial cell).
[000211] Although certain cells present an attractive target for ex vivo treatment and therapy, increased efficacy in delivery may permit direct in vivo delivery to such cells. Ideally the targeting and editing would be directed to the relevant cells. Cleavage in other cells can also be prevented by the use of promoters only active in certain cells and or developmental stages.
Additional promoters are inducible, and therefore can be temporally controlled if the nuclease is delivered as a plasmid. The amount of time that delivered RNA and protein remain in the cell can also be adjusted using treatments or domains added to change the half-life. In vivo treatment would eliminate a number of treatment steps, but a lower rate of delivery can require higher rates of editing. In vivo treatment can eliminate problems and losses from ex vivo treatment, engraftment and post-engraftment integration of neurons and glial cells appropriately into existing brain circuits.
[000212] An advantage of in vivo gene therapy can be the ease of therapeutic production and administration. The same therapeutic approach and therapy will have the potential to be used to treat more than one patient, for example a number of patients who share the same or similar genotype or allele. In contrast, ex vivo cell therapy typically requires using a patient's own cells, which are isolated, manipulated and returned to the same patient.
[000213] Also provided herein is a cellular method for editing the ATXN3 gene in a cell by genome editing. For example, a cell can be isolated from a patient or animal. Then, the chromosomal DNA of the cell can be edited using the materials and methods described herein.
[000214] The methods provided herein, regardless of whether a cellular or ex vivo or in vivo method, can involve one or a combination of the following: 1) deleting the entire expanded trinucleotide repeat or a portion thereof in or near the ATXN3 gene, by inducing two double- stranded DNA breaks at both sides of the expanded region; 2) deleting the entire expanded trinucleotide repeat or a portion thereof in or near the ATXN3 gene, by inducing one double- stranded DNA break proximal to the expanded region; 3) replacing the deleted expanded trinucleotide repeat sequence with a corrected sequence; 4) deleting the mutant ATXN3 gene and inserting a wild-type ATXN3 gene, a cDNA or a minigene (comprised of one or more exons and introns or natural or synthetic introns) into the ATXN3 gene locus or a safe harbor locus; or 5) correcting a point mutation within or near the ATXN3 gene.
[000215] For example, the dual DSB-induced deletion strategy can involve excising the entire abnormal repeat expansion or a portion thereof in the ATXN3 gene by inducing two or more double stranded breaks at both sides of the repeat region with one or more CRISPR
endonucleases and two or more sgRNAs. In certain aspects, a donor DNA containing the corrected sequence can be provided to restore the wild-type sequence. This approach can require development and optimization of sgRNAs and donor DNA molecules for the ATXN3 gene.
[000216] For example, the single DSB-induced deletion strategy can involve deleting the entire abnormal repeat expansion or a portion thereof in the ATXN3 gene by inducing one double stranded break at a site proximal to the repeat region with one or more CRISPR endonucleases and a gRNA (e.g., crRNA + tracrRNA, or sgRNA). In certan aspects, a donor DNA containing the corrected sequence can be provided to restore the wild-type sequence. This approach can require development and optimization of gRNAs and donor DNA molecules for the ATXN3 gene.
[000217] For example, replacing the deleted expanded trinucleotide repeat sequence with a corrected sequence can be achieved by delivering into the cell one or more CRISPR
endonucleases, a pair of gRNAs (e.g., crRNA + tracrRNA, or sgRNA) targeting upstream or downstream of the expanded trinucleotide repeat sequence, and a donor DNA that contains the desired sequence and homology arms to the flanking regions of the target locus. This approach can require development and optimization of sgRNAs for the ATXN3 gene.
[000218] For example, the whole gene correction strategy can involve deletion of the endogenous, mutated ATXN3 gene and insertion of a wild-type ATXN3 gene, a cDNA or a minigene (comprised of one or more exons and introns or natural or synthetic introns) into the locus of the ATXN3 gene. In certain aspects, this can be achieved by delivering into the cell one or more CRISPR endonucleases, a pair of gRNAs (e.g., crRNA + tracrRNA, or sgRNA) targeting upstream and downstream of or in the first and last exon and/or intron of the ATXN3 gene, and a donor DNA that contains the desired sequence and homology arms to the flanking regions of the target locus. In certain aspects, this can be achieved by delivering into the cell one or more CRISPR endonucleases, a pair of gRNAs (e.g., crRNA + tracrRNA, or sgRNA) targeting upstream and downstream of or within one or more exon and/or intron of the ATXN3 gene, and a donor DNA that contains the desired sequence and homology arms to the flanking regions of the target locus. The cytogenetic location of the ATXN3 gene is 14q32.12. Alternatively, the wild-type ATXN3 gene, a cDNA or a minigene (comprised of one or more exons and introns or natural or synthetic introns) can be inserted into a safe harbor locus. A "safe harbor locus" refers to a region of the genome where the integrated material can be adequately expressed without perturbing endogenous gene structure or function. The safe harbor loci include but are not limited to AAVSl (PPP1R12C), ALB, Angpt , ApoC3, ASGR2, CCR5, FIX (F9), Gys2, HGD, Lp(a), Pcsk9, Serpinal, TF, and/or TTR. The target sites within the safe harbor loci can be selected from the group consisting of: exon 1-2 of AAVS l (PPP1R12C), exon 1-2 of ALB, exon 1-2 of AngptB, exon 1-2 of ApoC3, exon 1-2 of ASGR2, exon 1-2 of CCR5, exon 1-2 of FIX (F9), exon 1-2 of Gys2, exon 1-2 of HGD, exon 1-2 of Lp(a), exon 1-2 of Pcsk9, exon 1-2 of Serpinal, exon 1-2 of TF, and/or exon 1-2 of TTR.
[000219] The whole gene correction strategy utilizes a donor DNA template in Homology- Directed Repair (HDR). HDR may be accomplished by making one or more single-stranded breaks (SSBs) or double-stranded breaks (DSBs) at specific sites in the genome by using one or more endonucleases. The donor DNA can be single or double stranded DNA. The donor template can have homologous arms to the 14q32.12 region. The donor template can have homologous arms to a safe harbor locus. For example, the donor template can have homologous arms to an AAVS l safe harbor locus, such as, intron 1 of the PPP1R12C gene. As another example, the donor template can have homologous arms to a CCR5 safe harbor locus.
[000220] For example, correcting a point mutation can involve replacing one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene.
Correcting point mutation can involve deleting the sequence containing the mutation by inducing a double stranded break at a site proximal and a site distal to the point mutation with one or more CRISPR endonucleases and a gRNA (e.g., crRNA + tracrRNA, or sgRNA). In certain aspects, a donor DNA containing the corrected sequence can be provided to restore the wild-type sequence. This approach can require development and optimization of gRNAs and donor DNA molecule for the ATXN3 gene.
[000221] The advantages for the above strategies are similar, including in principle both short and long term beneficial clinical and laboratory effects.
[000222] In addition to the above genome editing strategies, another strategy involves modulating expression, function, or activity of ATXN3 by editing in the regulatory sequence.
[000223] In addition to the editing options listed above, Cas9 or similar proteins can be used to target effector domains to the same target sites that can be identified for editing, or additional target sites within range of the effector domain. A range of chromatin modifying enzymes, methylases or demethylases can be used to alter expression of the target gene. One possibility is decreasing the expression of the ATXN3 protein if the mutation leads to undesirable activity. These types of epigenetic regulation have some advantages, particularly as they are limited in possible off-target effects.
[000224] A number of types of genomic target sites can be present in addition to the trinucleotide repeat expansion in the coding sequence of the ATXN3 gene.
[000225] The regulation of transcription and translation implicates a number of different classes of sites that interact with cellular proteins or nucleotides. Often the DNA binding sites of transcription factors or other proteins can be targeted for mutation or deletion to study the role of the site, though they can also be targeted to change gene expression. Sites can be added through non-homologous end joining NHEJ or direct genome editing by homology directed repair (HDR). Increased use of genome sequencing, RNA expression and genome-wide studies of transcription factor binding have increased our ability to identify how the sites lead to developmental or temporal gene regulation. These control systems can be direct or can involve extensive cooperative regulation that can require the integration of activities from multiple enhancers. Transcription factors typically bind 6-12 bp-long degenerate DNA sequences. The low level of specificity provided by individual sites suggests that complex interactions and rules are involved in binding and the functional outcome. Binding sites with less degeneracy can provide simpler means of regulation. Artificial transcription factors can be designed to specify longer sequences that have less similar sequences in the genome and have lower potential for off-target cleavage. Any of these types of binding sites can be mutated, deleted or even created to enable changes in gene regulation or expression (Canver, M.C. et al., Nature (2015)).
[000226] Another class of gene regulatory regions having these features is microRNA
(miRNA) binding sites. miRNAs are non-coding RNAs that play key roles in post- transcriptional gene regulation. miRNA can regulate the expression of 30% of all mammalian protein-encoding genes. Specific and potent gene silencing by double stranded RNA (RNAi) was discovered, plus additional small non-coding RNA (Canver, M.C. et al., Nature (2015)). The largest class of non-coding RNAs important for gene silencing are miRNAs. In mammals, miRNAs are first transcribed as a long RNA transcript, which can be separate transcriptional units, part of protein introns, or other transcripts. The long transcripts are called primary miRNA (pri-miRNA) that include imperfectly base-paired hairpin structures. These pri-miRNA can be cleaved into one or more shorter precursor miRNAs (pre-miRNAs) by Microprocessor, a protein complex in the nucleus, involving Drosha. [000227] Pre-miRNAs are short stem loops -70 nucleotides in length with a 2-nucleotide 3'- overhang that are exported, into the mature 19-25 nucleotide miRNA:miRNA* duplexes. The miRNA strand with lower base pairing stability (the guide strand) can be loaded onto the RNA- induced silencing complex (RISC). The passenger strand (marked with *), can be functional, but is usually degraded. The mature miRNA tethers RISC to partly complementary sequence motifs in target mRNAs predominantly found within the 3' untranslated regions (UTRs) and induces posttranscriptional gene silencing (Bartel, D.P. Cell 136, 215-233 (2009); Saj, A. & Lai, E.C. Curr Opin Genet Dev 21, 504-510 (2011)).
[000228] miRNAs can be important in development, differentiation, cell cycle and growth control, and in virtually all biological pathways in mammals and other multicellular organisms. miRNAs can also be involved in cell cycle control, apoptosis and stem cell differentiation, hematopoiesis, hypoxia, muscle development, neurogenesis, insulin secretion, cholesterol metabolism, aging, viral replication and immune responses.
[000229] A single miRNA can target hundreds of different mRNA transcripts, while an individual miRNA transcript can be targeted by many different miRNAs. More than 28645 microRNAs have been annotated in the latest release of miRBase (v.21). Some miRNAs can be encoded by multiple loci, some of which can be expressed from tandemly co-transcribed clusters. The features allow for complex regulatory networks with multiple pathways and feedback controls. miRNAs can be integral parts of these feedback and regulatory circuits and can help regulate gene expression by keeping protein production within limits (Herranz, H. & Cohen, S.M. Genes Dev 24, 1339-1344 (2010); Posadas, D M. & Carthew, R.W. Curr Opin Genet Dev 27, 1-6 (2014)).
[000230] miRNA can also be important in a large number of human diseases that are associated with abnormal miRNA expression. This association underscores the importance of the miRNA regulatory pathway. Recent miRNA deletion studies have linked miRNA with regulation of the immune responses (Stern-Ginossar, N. et al., Science 317, 376-381 (2007)).
[000231] miRNA also has a strong link to cancer and can play a role in different types of cancer. miRNAs have been found to be downregulated in a number of tumors. miRNA can be important in the regulation of key cancer-related pathways, such as cell cycle control and the DNA damage response, and can therefore be used in diagnosis and can be targeted clinically.
MicroRNAs can delicately regulate the balance of angiogenesis, such that experiments depleting all microRNAs suppresses tumor angiogenesis (Chen, S. et al., Genes Dev 28, 1054-1067 (2014)). [000232] As has been shown for protein coding genes, miRNA genes can also be subject to epigenetic changes occurring with cancer. Many miRNA loci can be associated with CpG islands increasing their opportunity for regulation by DNA methylation (Weber, B., Stresemann, C, Brueckner, B. & Lyko, F. Cell Cycle 6, 1001-1005 (2007)). The majority of studies have used treatment with chromatin remodeling drugs to reveal epigenetically silenced miRNAs.
[000233] In addition to their role in RNA silencing, miRNA can also activate translation (Posadas, D M. & Carthew, R.W. Curr Opin Genet Dev 27, 1-6 (2014)). Knocking out miRNA sites may lead to decreased expression of the targeted gene, while introducing these sites may increase expression.
[000234] Individual miRNA can be knocked out most effectively by mutating the seed sequence (bases 2-8 of the microRNA), which can be important for binding specificity.
Cleavage in this region, followed by mis-repair by NHEJ can effectively abolish miRNA function by blocking binding to target sites. miRNA could also be inhibited by specific targeting of the special loop region adjacent to the palindromic sequence. Catalytically inactive Cas9 can also be used to inhibit shRNA expression (Zhao, Y. et al., Sci Rep 4, 3943 (2014)). In addition to targeting the miRNA, the binding sites can also be targeted and mutated to prevent the silencing by miRNA.
[000235] According to the present disclosure, any of the microRNA (miRNA) or their binding sites may be incorporated into the compositions of the invention.
[000236] The compositions may have a region such as, but not limited to, a region comprising the sequence of any of the microRNAs listed in SEQ ID NOs: 632-4715, the reverse complement of the microRNAs listed in SEQ ID NOs: 632-4715, or the microRNA anti-seed region of any of the microRNAs listed in SEQ ID NOs: 632-4715.
[000237] The compositions of the present disclosure may comprise one or more microRNA target sequences, microRNA sequences, or microRNA seeds. Such sequences may correspond to any known microRNA such as those taught in U.S. Publication US2005/0261218 and U.S. Publication US2005/0059005. As a non-limiting example, known microRNAs, their sequences and their binding site sequences in the human genome are listed below in SEQ ID NOs: 632- 4715.
[000238] A microRNA sequence comprises a "seed" sequence, i.e., a sequence in the region of positions 2-8 of the mature microRNA, which sequence has perfect Watson-Crick
complementarity to the miRNA target sequence. A microRNA seed may comprise positions 2-8 or 2-7 of the mature microRNA. In some aspects, a microRNA seed may comprise 7 nucleotides (e.g., nucleotides 2-8 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked by an adenine (A) opposed to microRNA position 1. In some aspects, a microRNA seed may comprise 6 nucleotides (e.g., nucleotides 2-7 of the mature microRNA), wherein the seed-complementary site in the corresponding miRNA target is flanked by an adenine (A) opposed to microRNA position 1. See for example, Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP; Mol Cell. 2007 Jul 6;27(1):91-105. The bases of the microRNA seed have complete complementarity with the target sequence.
[000239] Identification of microRNA, microRNA target regions, and their expression patterns and role in biology have been reported (Bonauer et al., Curr Drug Targets 2010 11 : 943 -949; Anand and Cheresh Curr Opin Hematol 2011 18: 171-176; Contreras and Rao Leukemia 2012 26:404-413 (2011 Dec 20. doi: 10.1038/leu.2011.356); Bartel Cell 2009 136:215-233; Landgraf et al, Cell, 2007 129: 1401-1414; Gentner and Naldini, Tissue Antigens. 2012 80:393-403).
[000240] For example, if the composition is not intended to be delivered to the liver but ends up there, then miR-122, a microRNA abundant in liver, can inhibit the expression of the sequence delivered if one or multiple target sites of miR-122 are engineered into the
polynucleotide encoding that target sequence. Introduction of one or multiple binding sites for different microRNA can be engineered to further decrease the longevity, stability, and protein translation hence providing an additional layer of tenability.
[000241] As used herein, the term "microRNA site" refers to a microRNA target site or a microRNA recognition site, or any nucleotide sequence to which a microRNA binds or associates. It should be understood that "binding" may follow traditional Watson-Crick hybridization rules or may reflect any stable association of the microRNA with the target sequence at or adjacent to the microRNA site.
[000242] Conversely, for the purposes of the compositions of the present disclosure, microRNA binding sites can be engineered out of (i.e. removed from) sequences in which they naturally occur in order to increase protein expression in specific tissues. For example, miR-137 binding sites may be removed to improve protein expression in the brain.
[000243] Specifically, microRNAs are known to be differentially expressed in immune cells (also called hematopoietic cells), such as antigen presenting cells (APCs) (e.g. dendritic cells and macrophages), macrophages, monocytes, B lymphocytes, T lymphocytes, granulocytes, natural killer cells, etc. Immune cell specific microRNAs are involved in immunogenicity,
autoimmunity, the immune -response to infection, inflammation, as well as unwanted immune response after gene therapy and tissue/organ transplantation. Immune cells specific microRNAs also regulate many aspects of development, proliferation, differentiation and apoptosis of hematopoietic cells (immune cells). For example, miR-142 and miR-146 are exclusively expressed in the immune cells, particularly abundant in myeloid dendritic cells. Introducing the miR-142 binding site into the 3'-UTR of a polypeptide of the present disclosure can selectively suppress the gene expression in the antigen presenting cells through miR-142 mediated mRNA degradation, limiting antigen presentation in professional APCs (e.g. dendritic cells) and thereby preventing antigen-mediated immune response after gene delivery (see, Annoni A et al., blood, 2009, 114, 5152-5161).
[000244] In one example, microRNAs binding sites that are known to be expressed in immune cells, in particular, the antigen presenting cells, can be engineered into the polynucleotides to suppress the expression of the polynucleotide in APCs through microRNA mediated RNA degradation, subduing the antigen-mediated immune response, while the expression of the polynucleotide is maintained in non-immune cells where the immune cell specific microRNAs are not expressed.
[000245] Many microRNA expression studies have been conducted, and are described in the art, to profile the differential expression of microRNAs in various cancer cells /tissues and other diseases. Some microRNAs are abnormally over-expressed in certain cancer cells and others are under-expressed. For example, microRNAs are differentially expressed in cancer cells
(WO2008/154098, US2013/0059015, US2013/0042333, WO2011/157294); cancer stem cells (US2012/0053224); pancreatic cancers and diseases (US2009/0131348, US2011/0171646, US2010/0286232, US8389210); asthma and inflammation (US8415096); prostate cancer (US2013/0053264); hepatocellular carcinoma (WO2012/151212, US2012/0329672,
WO2008/054828, US8252538); lung cancer cells (WO2011/076143, WO2013/033640,
WO2009/070653, US2010/0323357); cutaneous T-cell lymphoma (WO2013/011378); colorectal cancer cells (WO2011/0281756, WO2011/076142); cancer positive lymph nodes
(WO2009/100430, US2009/0263803); nasopharyngeal carcinoma (EP2112235); chronic obstructive pulmonary disease (US2012/0264626, US2013/0053263); thyroid cancer
(WO2013/066678); ovarian cancer cells ( US2012/0309645, WO2011/095623); breast cancer cells (WO2008/154098, WO2007/081740, US2012/0214699), leukemia and lymphoma
(WO2008/073915, US2009/0092974, US2012/0316081, US2012/0283310, WO2010/018563).
[000246] Non-limiting examples of microRNA sequences and the targeted tissues and/or cells are disclosed in SEQ ID NOs: 632-4715. Genome engineering strategies
[000247] The methods of the present disclosure can involve editing one or both of the mutant alleles. Gene editing to modify or correct the ATXN3 gene has the advantage of restoration of correct expression levels or elimination of aberrant gene products and temporal control.
[000248] A step of the ex vivo methods of the present disclosure can comprise
editing/correcting the patient specific iPSCs or mesenchymal stem cells. Alternatively, a step of the in vivo methods of the present disclosure involves editing/correcting the cells in a
Spinocerebellar ataxia 3 (SCA3) patient using genome engineering. Similarly, a step in the cellular methods of the present disclosure can comprise editing/correcting the ATXN3 gene in a human cell by genome engineering.
[000249] Spinocerebellar ataxia 3 (SCA3) patients exhibit expanded trinucleotide repeats in the ATXN3 gene. Therefore, different patients may use similar editing strategies. Any CRISPR endonuclease may be used in the methods of the present disclosure, each CRISPR endonuclease having its own associated PAM, which may or may not be disease specific.
[000250] In one aspect, the trinucleotide repeat expansion may be excised by inducing two double stranded breaks (DSBs) upstream and downstream of the repeat region. Pairs of gRNAs have been used for this type of deletions. The CRISPR endonucleases, configured with the two gRNAs, induce two DSBs at the desired locations. After cleavage, the two ends, regardless of whether blunt or with overhangs, can be joined by NHEJ, leading to the deletion of the intervening segment. In certain aspects, the wild-type sequence can be restored by inserting a correct repeat sequence via HDR.
[000251] In another aspect, the trinucleotide repeat expansion may be deleted after inducing a single DSB near or within the repeat region. Single DSB-induced repeat loss has been reported in several studies including TALEN-cleaved short CAG/CTG repeats engineered in yeast, ZFN- cleaved CAG repeats engineered in human cells, and CRISPR/Cas9-cleaved CTG/CAG repeats engineered in human cells (Richard et al., PLoS ONE (2014), 9(4): e95611; Mittelman et al., Proc Natl Acad Sci USA (2009), 106(24):9607-12; van Agtmaal et al., Mol Ther (2016), http://dx.doi.Org/10.1016/j .ymthe.2016.10.014). A DSB near the repeat region destabilizes the repeat tracts, triggering a contraction (e.g. partial deletion) or, in some cases, a complete deletion of the repeats. Additionally, work by Mittelman et al. suggested that single DSB-induced repeat loss is more effective in longer repeats. In certain aspects, the wild-type sequence can be restored by inserting a correct repeat sequence via HDR. [000252] Alternatively, expression of the mutant ATXN3 gene may be disrupted or eliminated by introducing random insertions or deletions (indels) that arise due to the imprecise HEJ repair pathway. The target regions may be the coding sequence of the ATXN3 gene (i.e., exons). Inserting or deleting nucleotides into the coding sequence of a gene may cause a "frame shift" where the normal 3-letter codon pattern is disturbed. In this way, gene expression and therefore mutant protein production can be reduced or eliminated. This approach may also be used to target any intron, intron:exon junction, or regulatory DNA element of the ATXN3 gene where sequence alteration may interfere with the expression of the ATXN3 gene. This approach can require development and optimization of sgRNAs for the ATXN3 gene.
[000253] As a further alternative, the entire mutant gene can be deleted and a wild-type gene, a cDNA or a minigene (comprised of one or more exons and introns or natural or synthetic introns) can be knocked into the gene locus or a heterologous location in the genome such as a safe harbor locus. Pairs of nucleases can be used to delete mutated gene regions, and a donor is provided to restore function. In this case two gRNA and one donor sequence would be supplied. A full length cDNA can be knocked into any "safe harbor", but must use a supplied or an endogenous promoter. If this construct is knocked into the ATXN3 gene locus, it will have physiological control, similar to the normal gene.
[000254] Some genome engineering strategies involve correcting the repeat expansion, or inserting a wild-type ATXN3 gene or cDNA or a minigene (comprised of one or more exons and introns or natural or synthetic introns) into the locus of the corresponding gene or a safe harbor locus by homology directed repair (HDR), which is also known as homologous recombination (HR). Homology directed repair can be one strategy for treating patients that have expanded trinucleotides in the ATXN3 gene. These strategies can restore the ATXN3 gene and reverse, treat, and/or mitigate the diseased state.
[000255] Homology directed repair is a cellular mechanism for repairing double-stranded breaks (DSBs). The most common form is homologous recombination. There are additional pathways for HDR, including single-strand annealing and alternative-HDR. Genome
engineering tools allow researchers to manipulate the cellular homologous recombination pathways to create site-specific modifications to the genome. It has been found that cells can repair a double-stranded break using a synthetic donor molecule provided in trans. Therefore, by introducing a double-stranded break near a specific mutation and providing a suitable donor, targeted changes can be made in the genome. Specific cleavage increases the rate of HDR more than 1,000 fold above the rate of 1 in 106 cells receiving a homologous donor alone. The rate of homology directed repair (HDR) at a particular nucleotide is a function of the distance to the cut site, so choosing overlapping or nearest target sites is important. Gene editing offers the advantage over gene addition, as correcting in situ leaves the rest of the genome unperturbed.
[000256] Supplied donors for editing by HDR vary markedly but can contain the intended sequence with small or large flanking homology arms to allow annealing to the genomic DNA. The homology regions flanking the introduced genetic changes can be 30 bp or smaller or as large as a multi-kilobase cassette that can contain promoters, cDNAs, and the like. Both single- stranded and double-stranded oligonucleotide donors have been used. These oligonucleotides range in size from less than 100 nt to over many kb, though longer ssDNA can also be generated and used. Double-stranded donors can be used, including PCR amplicons, plasmids, and mini- circles. In general, it has been found that an AAV vector can be a very effective means of delivery of a donor template, though the packaging limits for individual donors is <5kb. Active transcription of the donor increased HDR three-fold, indicating the inclusion of promoter may increase conversion. Conversely, CpG methylation of the donor decreased gene expression and HDR.
[000257] In addition to wild-type endonucleases, such as Cas9, nickase variants exist that have one or the other nuclease domain inactivated resulting in cutting of only one DNA strand. HDR can be directed from individual Cas nickases or using pairs of nickases that flank the target area. Donors can be single-stranded, nicked, or dsDNA.
[000258] The donor DNA can be supplied with the nuclease or independently by a variety of different methods, for example by transfection, nano-particle, micro-injection, or viral transduction. A range of tethering options has been proposed to increase the availability of the donors for HDR. Examples include attaching the donor to the nuclease, attaching to DNA binding proteins that bind nearby, or attaching to proteins that are involved in DNA end binding or repair.
[000259] The repair pathway choice can be guided by a number of culture conditions, such as those that influence cell cycling, or by targeting of DNA repair and associated proteins. For example, to increase HDR, key NHEJ molecules can be suppressed, such as KU70, KU80 or DNA ligase IV.
[000260] Without a donor present, the ends from a DNA break or ends from different breaks can be joined using the several non-homologous repair pathways in which the DNA ends are joined with little or no base-pairing at the junction. In addition to canonical NHEJ, there are similar repair mechanisms, such as alt-NHEJ. If there are two breaks, the intervening segment can be deleted or inverted. NHEJ repair pathways can lead to insertions, deletions or mutations at the joints.
[000261] NHEJ was used to insert a 15-kb inducible gene expression cassette into a defined locus in human cell lines after nuclease cleavage. (See e.g., Maresca, M., Lin, V.G., Guo, N. & Yang, Y., Genome Res 23, 539-546 (2013); Cristea et al. Biotechnology and Bioengineering, 110 (3):871-80 (2013); Suzuki et al. Nature, 540, 144-149 (2016)).
[000262] In addition to genome editing by NHEJ or HDR, site-specific gene insertions have been conducted that use both the NHEJ pathway and HR. A combination approach may be applicable in certain settings, possibly including intron/exon borders. NHEJ may prove effective for ligation in the intron, while the error-free HDR may be better suited in the coding region.
[000263] The mutation of the ATXN3 gene that causes Spinocerebellar ataxia 3 (SCA3) is a trinucleotide repeat expansion of the three letter string of nucleotides CAG. In healthy individuals, there are few repeats of this trinucleotide, typically about or fewer than 43. In people with the diseases phenotype, the repeat can occur more than 50 times. One or more trinucleotide repeats may be deleted or corrected in order to restore the gene to a wild-type or similar number of trinucleotide repeats. In some aspects, all of the trinucleotide repeats may be deleted. Alternatively, the mutant gene may be knocked out to eliminate toxic gene products. Any one or more of the mutations can be repaired in order to restore the ATXN3. As a further alternative, an ATXN3 gene or cDNA can be inserted to the locus of the corresponding gene to replace the mutant gene or knocked-in to a safe harbor site, such as AAVS1. In some examples, the methods can provide one gRNA or a pair of gRNAs that can be used to facilitate
incorporation of a new sequence from a polynucleotide donor template to knock-in a part of or the entire ATXN3 gene or cDNA.
[000264] Some genome engineering strategies involves repeat deletion. Targeted deletion of trinucleotide repeats is an attractive strategy for treating a large subset of patients with a single therapeutic cocktail. Deletions can either be single trinucleotide repeat deletions or multi- trinucleotide repeat deletions. While multi-repeat deletions, including complete deletion of the expanded trinucleotide repeat, can reach a larger number of patients, for larger deletions the efficiency of deletion greatly decreases with increased size. In some aspects, the deletions range from 40 to 1,000 base pairs (bp) in size. For example, deletions may range from 40-100; 100- 200; 200-300; 300-400; 400-500; or 500-1,000 base pairs in size.
[000265] The methods can provide gRNA pairs that make a deletion by cutting the gene twice, one gRNA cutting at the 5' end of the trinucleotide repeats and the other gRNA cutting at the 3' end of the trinucleotide repeats. The cutting can be accomplished by a pair of DNA endonucleases that each makes a D SB in the genome, or by multiple nickases that together make a D SB in the genome.
[000266] Alternatively, the methods can provide one gRNA to make one double-strand cut around the trinucleotide repeats. The double-strand cut can be made by a single DNA
endonuclease or multiple nickases that together make a D SB in the genome.
[000267] Illustrative modifications within the ATXN3 gene include deletions within or near (proximal) to the trinucleotide repeats referred to above, such as within the region of less than 3 kb, less than 2kb, less than 1 kb, less than 0.5 kb upstream or downstream of the specific mutation. Given the relatively wide variations of trinucleotide repeats in the ATXN3 gene, it will be appreciated that numerous variations of the deletions referenced above (including without limitation larger as well as smaller deletions), would be expected to result in restoration of the ATXN3 gene expression.
[000268] Such variants can include deletions that are larger in the 5' and/or 3' direction than the specific repeat expansion in question, or smaller in either direction. Accordingly, by "near" or "proximal" with respect to specific repeat expansion, it is intended that the SSB or DSB locus associated with a desired deletion boundary (also referred to herein as an endpoint) can be within a region that is less than about 3 kb from the reference locus noted. The SSB or DSB locus can be more proximal and within 2 kb, within 1 kb, within 0.5 kb, or within 0.1 kb. In the case of small deletion, the desired endpoint can be at or "adjacent to" the reference locus, by which it is intended that the endpoint can be within 100 bp, within 50 bp, within 25 bp, or less than about 10 bp to 5 bp from the reference locus.
[000269] In order to ensure that the pre-mRNA is properly processed following deletion, the surrounding splicing signals can be deleted. Splicing donor and acceptors are generally within 100 base pairs of the neighboring intron. Therefore, in some examples, methods can provide all gRNAs that cut approximately +/- 100-3100 bp with respect to each exon/intron junction of interest.
[000270] For any of the genome editing strategies, gene editing can be confirmed by sequencing or PCR analysis.
Target Sequence Selection
[000271] Shifts in the location of the 5' boundary and/or the 3' boundary relative to particular reference loci can be used to facilitate or enhance particular applications of gene editing, which depend in part on the endonuclease system selected for the editing, as further described and illustrated herein.
[000272] In a first non-limiting example of such target sequence selection, many endonuclease systems have rules or criteria that can guide the initial selection of potential target sites for cleavage, such as the requirement of a PAM sequence motif in a particular position adjacent to the DNA cleavage sites in the case of CRISPR Type II or Type V endonucleases.
[000273] In another nonlimiting example of target sequence selection or optimization, the frequency of off-target activity for a particular combination of target sequence and gene editing endonuclease (i.e. the frequency of DSBs occurring at sites other than the selected target sequence) can be assessed relative to the frequency of on-target activity. In some cases, cells that have been correctly edited at the desired locus can have a selective advantage relative to other cells. Illustrative, but nonlimiting, examples of a selective advantage include the acquisition of attributes such as enhanced rates of replication, persistence, resistance to certain conditions, enhanced rates of successful engraftment or persistence in vivo following
introduction into a patient, and other attributes associated with the maintenance or increased numbers or viability of such cells. In other cases, cells that have been correctly edited at the desired locus can be positively selected for by one or more screening methods used to identify, sort or otherwise select for cells that have been correctly edited. Both selective advantage and directed selection methods can take advantage of the phenotype associated with the correction. In some cases, cells can be edited two or more times in order to create a second modification that creates a new phenotype that is used to select or purify the intended population of cells. Such a second modification could be created by adding a second gRNA enabling expression of a selectable or screenable marker. In some cases, cells can be correctly edited at the desired locus using a DNA fragment that contains the cDNA and also a selectable marker.
[000274] Whether any selective advantage is applicable or any directed selection is to be applied in a particular case, target sequence selection can also be guided by consideration of off- target frequencies in order to enhance the effectiveness of the application and/or reduce the potential for undesired alterations at sites other than the desired target. As described further and illustrated herein and in the art, the occurrence of off-target activity can be influenced by a number of factors including similarities and dissimilarities between the target site and various off-target sites, as well as the particular endonuclease used. Bioinformatics tools are available that assist in the prediction of off-target activity, and frequently such tools can also be used to identify the most likely sites of off-target activity, which can then be assessed in experimental settings to evaluate relative frequencies of off-target to on-target activity, thereby allowing the selection of sequences that have higher relative on-target activities. Illustrative examples of such techniques are provided herein, and others are known in the art.
[000275] Another aspect of target sequence selection relates to homologous recombination events. Sequences sharing regions of homology can serve as focal points for homologous recombination events that result in deletion of intervening sequences. Such recombination events occur during the normal course of replication of chromosomes and other DNA sequences, and also at other times when DNA sequences are being synthesized, such as in the case of repairs of double-strand breaks (DSBs), which occur on a regular basis during the normal cell replication cycle but can also be enhanced by the occurrence of various events (such as UV light and other inducers of DNA breakage) or the presence of certain agents (such as various chemical inducers). Many such inducers cause DSBs to occur indiscriminately in the genome, and DSBs can be regularly induced and repaired in normal cells. During repair, the original sequence can be reconstructed with complete fidelity, however, in some cases, small insertions or deletions (referred to as "indels") are introduced at the DSB site.
[000276] DSBs can also be specifically induced at particular locations, as in the case of the endonucleases systems described herein, which can be used to cause directed or preferential gene modification events at selected chromosomal locations. The tendency for homologous sequences to be subject to recombination in the context of DNA repair (as well as replication) can be taken advantage of in a number of circumstances, and is the basis for one application of gene editing systems, such as CRISPR, in which homology directed repair is used to insert a sequence of interest, provided through use of a "donor" polynucleotide, into a desired
chromosomal location.
[000277] Regions of homology between particular sequences, which can be small regions of "microhomology" that can comprise as few as ten base pairs or less, can also be used to bring about desired deletions. For example, a single DSB can be introduced at a site that exhibits microhomology with a nearby sequence. During the normal course of repair of such DSB, a result that occurs with high frequency is the deletion of the intervening sequence as a result of recombination being facilitated by the DSB and concomitant cellular repair process.
[000278] In some circumstances, however, selecting target sequences within regions of homology can also give rise to much larger deletions, including gene fusions (when the deletions are in coding regions), which may or may not be desired given the particular circumstances. [000279] The examples provided herein further illustrate the selection of various target regions for the creation of DSBs designed to induce deletions or replacements that result in restoration of wild-type or similar levels of trinucleotide repeats in the ATXN3 gene, as well as the selection of specific target sequences within such regions that are designed to minimize off-target events relative to on-target events.
Human Cells
[000280] For ameliorating Spinocerebellar ataxia 3 (SCA3) or any disorder associated with ATXN3, as described and illustrated herein, the principal targets for gene editing are human cells. For example, in the ex vivo methods, the human cells can be somatic cells, which after being modified using the techniques as described, can give rise to differentiated cells, e.g., neurons or progenitor cells. For example, in the in vivo methods, the human cells may be cells from the central nervous system or cells from other affected organs.
[000281] By performing gene editing in autologous cells that are derived from and therefore already completely matched with the patient in need, it is possible to generate cells that can be safely re-introduced into the patient, and effectively give rise to a population of cells that will be effective in ameliorating one or more clinical conditions associated with the patient's disease.
[000282] Stem cells are capable of both proliferation and giving rise to more progenitor cells, these in turn having the ability to generate a large number of mother cells that can in turn give rise to differentiated or differentiable daughter cells. The daughter cells themselves can be induced to proliferate and produce progeny that subsequently differentiate into one or more mature cell types, while also retaining one or more cells with parental developmental potential. The term "stem cell" refers then, to a cell with the capacity or potential, under particular circumstances, to differentiate to a more specialized or differentiated phenotype, and which retains the capacity, under certain circumstances, to proliferate without substantially
differentiating. In one aspect, the term progenitor or stem cell refers to a generalized mother cell whose descendants (progeny) specialize, often in different directions, by differentiation, e.g., by acquiring completely individual characters, as occurs in progressive diversification of embryonic cells and tissues. Cellular differentiation is a complex process typically occurring through many cell divisions. A differentiated cell may derive from a multipotent cell that, itself, is derived from a multipotent cell, and so on. While each of these multipotent cells may be considered stem cells, the range of cell types that each can give rise to may vary considerably. Some differentiated cells also have the capacity to give rise to cells of greater developmental potential. Such capacity may be natural or may be induced artificially upon treatment with various factors. In many biological instances, stem cells can also be "multipotent" because they can produce progeny of more than one distinct cell type, but this is not required for "stem-ness."
[000283] Self-renewal can be another important aspect of the stem cell. In theory, self-renewal can occur by either of two major mechanisms. Stem cells can divide asymmetrically, with one daughter retaining the stem state and the other daughter expressing some distinct other specific function and phenotype. Alternatively, some of the stem cells in a population can divide symmetrically into two stems, thus maintaining some stem cells in the population as a whole, while other cells in the population give rise to differentiated progeny only. Generally,
"progenitor cells" have a cellular phenotype that is more primitive (i.e., is at an earlier step along a developmental pathway or progression than is a fully differentiated cell). Often, progenitor cells also have significant or very high-proliferative potential. Progenitor cells can give rise to multiple distinct differentiated cell types or to a single differentiated cell type, depending on the developmental pathway and on the environment in which the cells develop and differentiate.
[000284] In the context of cell ontogeny, the adjective "differentiated," or "differentiating" is a relative term. A "differentiated cell" is a cell that has progressed further down the developmental pathway than the cell to which it is being compared. Thus, stem cells can differentiate into lineage-restricted precursor cells (such as a neural progenitor cell), which in turn can
differentiate into other types of precursor cells further down the pathway (such as a neural precursor), and then to an end-stage differentiated cell, such as a neural, which plays a characteristic role in a certain tissue type, and may or may not retain the capacity to proliferate further.
Induced Pluripotent Stem Cells
[000285] The genetically engineered human cells described herein can be induced pluripotent stem cells (iPSCs). An advantage of using iPSCs is that the cells can be derived from the same subject to which the progenitor cells are to be administered. That is, a somatic cell can be obtained from a subject, reprogrammed to an induced pluripotent stem cell, and then re- differentiated into a progenitor cell to be administered to the subject {e.g., autologous cells). Because the progenitors are essentially derived from an autologous source, the risk of engraftment rejection or allergic response can be reduced compared to the use of cells from another subject or group of subjects. In addition, the use of iPSCs negates the need for cells obtained from an embryonic source. Thus, in one aspect, the stem cells used in the disclosed methods are not embryonic stem cells. [000286] Although differentiation is generally irreversible under physiological contexts, several methods have been recently developed to reprogram somatic cells to iPSCs. Exemplary methods are known to those of skill in the art and are described briefly herein below.
[000287] The term "reprogramming" refers to a process that alters or reverses the
differentiation state of a differentiated cell (e.g., a somatic cell). Stated another way, reprogramming refers to a process of driving the differentiation of a cell backwards to a more undifferentiated or more primitive type of cell. It should be noted that placing many primary cells in culture can lead to some loss of fully differentiated characteristics. Thus, simply culturing such cells included in the term differentiated cells does not render these cells non- differentiated cells (e.g., undifferentiated cells) or pluripotent cells. The transition of a differentiated cell to pluripotency requires a reprogramming stimulus beyond the stimuli that lead to partial loss of differentiated character in culture. Reprogrammed cells also have the characteristic of the capacity of extended passaging without loss of growth potential, relative to primary cell parents, which generally have capacity for only a limited number of divisions in culture.
[000288] The cell to be reprogrammed can be either partially or terminally differentiated prior to reprogramming. Reprogramming can encompass complete reversion of the differentiation state of a differentiated cell (e.g., a somatic cell) to a pluripotent state or a multipotent state. Reprogramming can encompass complete or partial reversion of the differentiation state of a differentiated cell (e.g., a somatic cell) to an undifferentiated cell (e.g., an embryonic-like cell). Reprogramming can result in expression of particular genes by the cells, the expression of which further contributes to reprogramming. In certain examples described herein, reprogramming of a differentiated cell (e.g., a somatic cell) can cause the differentiated cell to assume an
undifferentiated state (e.g., is an undifferentiated cell). The resulting cells are referred to as "reprogrammed cells," or "induced pluripotent stem cells (iPSCs or iPS cells)."
[000289] Reprogramming can involve alteration, e.g., reversal, of at least some of the heritable patterns of nucleic acid modification (e.g., methylation), chromatin condensation, epigenetic changes, genomic imprinting, etc., that occur during cellular differentiation. Reprogramming is distinct from simply maintaining the existing undifferentiated state of a cell that is already pluripotent or maintaining the existing less than fully differentiated state of a cell that is already a multipotent cell (e.g., a myogenic stem cell). Reprogramming is also distinct from promoting the self-renewal or proliferation of cells that are already pluripotent or multipotent, although the compositions and methods described herein can also be of use for such purposes, in some examples.
[000290] Many methods are known in the art that can be used to generate pluripotent stem cells from somatic cells. Any such method that reprograms a somatic cell to the pluripotent phenotype would be appropriate for use in the methods described herein.
[000291] Reprogramming methodologies for generating pluripotent cells using defined combinations of transcription factors have been described. Mouse somatic cells can be converted to ES cell-like cells with expanded developmental potential by the direct transduction of Oct4, Sox2, Klf4, and c-Myc; see, e.g., Takahashi and Yamanaka, Cell 126(4): 663-76 (2006). iPSCs resemble ES cells, as they restore the pluripotency-associated transcriptional circuitry and much of the epigenetic landscape. In addition, mouse iPSCs satisfy all the standard assays for pluripotency: specifically, in vitro differentiation into cell types of the three germ layers, teratoma formation, contribution to chimeras, germline transmission [see, e.g., Maherali and Hochedlinger, Cell Stem Cell. 3(6):595-605 (2008)], and tetraploid complementation.
[000292] Human iPSCs can be obtained using similar transduction methods, and the transcription factor trio, OCT4, SOX2, and NANOG, has been established as the core set of transcription factors that govern pluripotency; see, e.g., Budniatzky and Gepstein, Stem Cells Transl Med. 3(4):448-57 (2014); Barrett et al, Stem Cells Trans Med 3 : 1-6 sctm.2014-0121 (2014); Focosi et al., Blood Cancer Journal 4: e211 (2014); and references cited therein. The production of iPSCs can be achieved by the introduction of nucleic acid sequences encoding stem cell-associated genes into an adult, somatic cell, historically using viral vectors.
[000293] iPSCs can be generated or derived from terminally differentiated somatic cells, as well as from adult stem cells, or somatic stem cells. That is, a non-pluripotent progenitor cell can be rendered pluripotent or multipotent by reprogramming. In such instances, it may not be necessary to include as many reprogramming factors as required to reprogram a terminally differentiated cell. Further, reprogramming can be induced by the non-viral introduction of reprogramming factors, e.g., by introducing the proteins themselves, or by introducing nucleic acids that encode the reprogramming factors, or by introducing messenger RNAs that upon translation produce the reprogramming factors (see e.g., Warren et al., Cell Stem Cell, 7(5):618- 30 (2010). Reprogramming can be achieved by introducing a combination of nucleic acids encoding stem cell-associated genes, including, for example, Oct-4 (also known as Oct-3/4 or Pouf51), Soxl, Sox2, Sox3, Sox 15, Sox 18, NANOG, KM, Klf2, Klf4, Klf5, NR5A2, c-Myc, 1- Myc, n-Myc, Rem2, Tert, and LIN28. Reprogramming using the methods and compositions described herein can further comprise introducing one or more of Oct-3/4, a member of the Sox family, a member of the Klf family, and a member of the Myc family to a somatic cell. The methods and compositions described herein can further comprise introducing one or more of each of Oct-4, Sox2, Nanog, c-MYC and Klf4 for reprogramming. As noted above, the exact method used for reprogramming is not necessarily critical to the methods and compositions described herein. However, where cells differentiated from the reprogrammed cells are to be used in, e.g., human therapy, in one aspect the reprogramming is not effected by a method that alters the genome. Thus, in such examples, reprogramming can be achieved, e.g., without the use of viral or plasmid vectors.
[000294] The efficiency of reprogramming (i.e., the number of reprogrammed cells) derived from a population of starting cells can be enhanced by the addition of various agents, e.g., small molecules, as shown by Shi et al, Cell-Stem Cell 2:525-528 (2008); Huangfu et al, Nature Biotechnology 26(7):795-797 (2008) and Marson et al, Cell-Stem Cell 3 : 132-135 (2008).
Thus, an agent or combination of agents that enhance the efficiency or rate of induced pluripotent stem cell production can be used in the production of patient-specific or disease- specific iPSCs. Some non-limiting examples of agents that enhance reprogramming efficiency include soluble Wnt, Wnt conditioned media, BIX-01294 (a G9a histone methyltransferase), PD0325901 (a MEK inhibitor), DNA methyltransferase inhibitors, histone deacetylase (HDAC) inhibitors, valproic acid, 5'-azacytidine, dexamethasone, suberoylanilide, hydroxamic acid (SAHA), vitamin C, and trichostatin (TSA), among others.
[000295] Other non-limiting examples of reprogramming enhancing agents include:
Suberoylanilide Hydroxamic Acid (SAHA (e.g., MK0683, vorinostat) and other hydroxamic acids), BML-210, Depudecin (e.g., (-)-Depudecin), HC Toxin, Nullscript (4-(l,3-Dioxo-lH,3H- benzo[de]isoquinolin-2-yl)-N-hydroxybutanamide), Phenylbutyrate (e.g., sodium
phenylbutyrate) and Valproic Acid ((VP A) and other short chain fatty acids), Scriptaid, Suramin Sodium, Trichostatin A (TSA), APHA Compound 8, Apicidin, Sodium Butyrate,
pivaloyloxymethyl butyrate (Pivanex, AN-9), Trapoxin B, Chlamydocin, Depsipeptide (also known as FR901228 or FK228), benzamides (e.g., CI-994 (e.g., N-acetyl dinaline) and MS-27- 275), MGCD0103, NVP-LAQ-824, CBHA (m-carboxycinnaminic acid bishydroxamic acid), JNJ16241199, Tubacin, A-161906, proxamide, oxamflatin, 3-Cl-UCHA (e.g., 6-(3- chlorophenylureido) caproic hydroxamic acid), AOE (2-amino-8-oxo-9, 10-epoxydecanoic acid), CHAP31 and CHAP 50. Other reprogramming enhancing agents include, for example, dominant negative forms of the HDACs (e.g., catalytically inactive forms), siRNA inhibitors of the HDACs, and antibodies that specifically bind to the HDACs. Such inhibitors are available, e.g., from BIOMOL International, Fukasawa, Merck Biosciences, Novartis, Gloucester
Pharmaceuticals, Titan Pharmaceuticals, MethylGene, and Sigma Aldrich.
[000296] To confirm the induction of pluripotent stem cells for use with the methods described herein, isolated clones can be tested for the expression of a stem cell marker. Such expression in a cell derived from a somatic cell identifies the cells as induced pluripotent stem cells. Stem cell markers can be selected from the non-limiting group including SSEA3, SSEA4, CD9, Nanog, Fbxl5, Ecatl, Esgl, Eras, Gdf3, Fgf4, Cripto, Daxl, Zpf296, Slc2a3, Rexl, Utfl, and Natl. In one case, for example, a cell that expresses Oct4 or Nanog is identified as pluripotent. Methods for detecting the expression of such markers can include, for example, RT-PCR and immunological methods that detect the presence of the encoded polypeptides, such as Western blots or flow cytometric analyses. Detection can involve not only RT-PCR, but can also include detection of protein markers. Intracellular markers may be best identified via RT-PCR, or protein detection methods such as immunocytochemistry, while cell surface markers are readily identified, e.g., by immunocytochemi stry .
[000297] The pluripotent stem cell character of isolated cells can be confirmed by tests evaluating the ability of the iPSCs to differentiate into cells of each of the three germ layers. As one example, teratoma formation in nude mice can be used to evaluate the pluripotent character of the isolated clones. The cells can be introduced into nude mice and histology and/or immunohistochemistry can be performed on a tumor arising from the cells. The growth of a tumor comprising cells from all three germ layers, for example, further indicates that the cells are pluripotent stem cells.
Cells of the Central Nervous System
[000298] In some aspects, the genetically engineered human cells described herein are cells of the central nervous system. Neurons, which process information, and glial cells (or neuroglia), which provide mechanical and metabolic support to the nervous system and modulate information processed by neurons, are the two main classes of cells of the central nervous system. Non-limiting examples of neurons include sensory neurons (also referred to as afferent neurons) transfer information from the external environment to the central nervous system, motor neurons (also referred to as efferent neurons) transfer information from the central nervous system to the external environment, and interneurons (also referred to as association neurons) process information in the central nervous system and transfers the information from one neuron to the other within the central nervous system. Non-limiting examples of glial cells include astrocytes, oligodendrocytes, microglia and Schwann cells. CNS progenitor cells can be neural progenitor cells or glial progenitor cells.
[000299] The typical neuron transmits electrical signals from one cell to another. Neurons contain a cell body, dendrite, axon hillock, axon, nerve endings, neuronal synapases and neuromuscular junctions. Neurons may be named according to shape or the nature of the dendritic tree.
[000300] Neuroglia differ from neurons in several general ways in that they: do not form synapses, have essentially only one type of process, retain the ability to divide, and are less electrically excitable than neurons. Neuroglia are classified based on size and shape of their nucleus and distinguished from neurons, at the light microscopic level. Neuroglia are divided into two major categories based on size, the macroglia and the microglia. The macroglia are of ectodermal origin and consist of astrocytes, oligodendrocytes and ependymal cells. Microglia cells are probably of mesodermal origin.
Creating patient specific iPSCs
[000301] One step of the ex vivo methods of the present disclosure can involve creating a patient specific iPS cell, patient specific iPS cells, or a patient specific iPS cell line. There are many established methods in the art for creating patient specific iPS cells, as described in Takahashi and Yamanaka 2006; Takahashi, Tanabe et al. 2007. For example, the creating step can comprise: a) isolating a somatic cell, such as a skin cell or fibroblast, from the patient; and b) introducing a set of pluripotency-associated genes into the somatic cell in order to induce the cell to become a pluripotent stem cell. The set of pluripotency-associated genes can be one or more of the genes selected from the group consisting of OCT4, SOXl, SOX2, SOX3, SOX15, SOX18, NANOG, KLF1, KLF2, KLF4, KLF5, c-MYC, n-MYC, REM2, TERT and LIN28.
Performing a biopsy or aspirate of the patient's skin or bone marrow
[000302] A biopsy or aspirate is a sample of tissue or fluid taken from the body. There are many different kinds of biopsies or aspirates. Nearly all of them involve using a sharp tool to remove a small amount of tissue. If the biopsy will be on the skin or other sensitive area, numbing medicine can be applied first. A biopsy or aspirate may be performed according to any of the known methods in the art. For example, in a bone marrow aspirate, a large needle is used to enter the pelvis bone to collect bone marrow.
Isolating a mesenchymal stem cell
[000303] Mesenchymal stem cells can be isolated according to any method known in the art, such as from a patient's bone marrow or peripheral blood. For example, marrow aspirate can be collected into a syringe with heparin. Cells can be washed and centrifuged on a Percoll. The cells can be cultured in Dulbecco's modified Eagle's medium (DMEM) (low glucose) containing 10% fetal bovine serum (FBS) (Pittinger MF, Mackay AM, Beck SC et al, Science 1999;
284: 143-147).
Genetically Modified Cells
[000304] The term "genetically modified cell" refers to a cell that comprises at least one genetic modification introduced by genome editing {e.g., using the CRISPR/Cas9 or
CRISPR/Cpfl system). In some ex vivo examples herein, the genetically modified cell can be genetically modified neural progenitor cell. In some in vivo examples herein, the genetically modified cell can be a genetically modified cell of the central nervous system (e.g., a neuron or a glial cell). A genetically modified cell comprising an exogenous genome-targeting nucleic acid and/or an exogenous nucleic acid encoding a genome-targeting nucleic acid is contemplated herein.
[000305] The term "control treated population" describes a population of cells that has been treated with identical media, viral induction, nucleic acid sequences, temperature, confluency, flask size, pH, etc., with the exception of the addition of the genome editing components. Any method known in the art can be used to measure restoration of ATXN3 gene or protein expression or activity, for example Western Blot analysis of the ATXN3 protein or real time PCR quantifying ATXN3 mRNA.
[000306] The term "isolated cell" refers to a cell that has been removed from an organism in which it was originally found, or a descendant of such a cell. Optionally, the cell can be cultured in vitro, e.g., under defined conditions or in the presence of other cells. Optionally, the cell can be later introduced into a second organism or re-introduced into the organism from which it (or the cell from which it is descended) was isolated.
[000307] The term "isolated population" with respect to an isolated population of cells refers to a population of cells that has been removed and separated from a mixed or heterogeneous population of cells. In some cases, the isolated population can be a substantially pure population of cells, as compared to the heterogeneous population from which the cells were isolated or enriched. In some cases, the isolated population can be an isolated population of human progenitor cells, e.g., a substantially pure population of human progenitor cells, as compared to a heterogeneous population of cells comprising human progenitor cells and cells from which the human progenitor cells were derived. [000308] The term "substantially enhanced," with respect to a particular cell population, refers to a population of cells in which the occurrence of a particular type of cell is increased relative to pre-existing or reference levels, by at least 2-fold, at least 3-, at least 4-, at least 5-, at least 6-, at least 7-, at least 8-, at least 9, at least 10-, at least 20-, at least 50-, at least 100-, at least 400-, at least 1000-, at least 5000-, at least 20000-, at least 100000- or more fold depending, e.g., on the desired levels of such cells for ameliorating Spinocerebellar ataxia 3 (SCA3).
[000309] The term "substantially enriched" with respect to a particular cell population, refers to a population of cells that is at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%), about 70% or more with respect to the cells making up a total cell population.
[000310] The term "substantially pure" with respect to a particular cell population, refers to a population of cells that is at least about 75%, at least about 85%, at least about 90%, or at least about 95%) pure, with respect to the cells making up a total cell population. That is, the terms "substantially pure" or "essentially purified," with regard to a population of progenitor cells, refers to a population of cells that contain fewer than about 20%, about 15%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1%, or less than 1%), of cells that are not progenitor cells as defined by the terms herein.
Differentiation of genome-edited iPSCs into cells of the central nervous system (CNS)
[000311] Another step of the ex vivo methods of the present disclosure can comprise differentiating the genome-edited iPSCs into cells of the central nervous system (CNS) (e.g., neurons or glial cells). The differentiating step may be performed according to any method known in the art.
Differentiation of genome-edited mesenchymal stem cells into cells of the central nervous system (CNS)
[000312] Another step of the ex vivo methods of the present disclosure can comprise differentiating the genome-edited mesenchymal stem cells into cells of the central nervous system (CNS). The differentiating step may be performed according to any method known in the art.
Implanting cells into patients
[000313] Another step of the ex vivo methods of the present disclosure involves implanting the cells of the central nervous system (e.g., neurons or glial cells) into patients. This implanting step may be accomplished using any method of implantation known in the art. For example, the genetically modified neurons may be administered to the patient via intraparenchymal, vascular (e.g., intravenous, intra-arterial), or ventricular (e.g., intracerebroventricular, intracisternal, intrathecal) routes or other routes such as intracranial or intraperitoneal injection.
III. FORMULATIONS AND DELIVERY
Pharmaceutically Acceptable Carriers
[000314] The ex vivo methods of administering progenitor cells to a subject contemplated herein involve the use of therapeutic compositions comprising progenitor cells.
[000315] Therapeutic compositions can contain a physiologically tolerable carrier together with the cell composition, and optionally at least one additional bioactive agent as described herein, dissolved or dispersed therein as an active ingredient. In some cases, the therapeutic
composition is not substantially immunogenic when administered to a mammal or human patient for therapeutic purposes, unless so desired.
[000316] In general, the progenitor cells described herein can be administered as a suspension with a pharmaceutically acceptable carrier. One of skill in the art will recognize that a pharmaceutically acceptable carrier to be used in a cell composition will not include buffers, compounds, cryopreservation agents, preservatives, or other agents in amounts that substantially interfere with the viability of the cells to be delivered to the subject. A formulation comprising cells can include e.g., osmotic buffers that permit cell membrane integrity to be maintained, and optionally, nutrients to maintain cell viability or enhance engraftment upon administration. Such formulations and suspensions are known to those of skill in the art and/or can be adapted for use with the progenitor cells, as described herein, using routine experimentation.
[000317] A cell composition can also be emulsified or presented as a liposome composition, provided that the emulsification procedure does not adversely affect cell viability. The cells and any other active ingredient can be mixed with excipients that are pharmaceutically acceptable and compatible with the active ingredient, and in amounts suitable for use in the therapeutic methods described herein.
[000318] Additional agents included in a cell composition can include pharmaceutically acceptable salts of the components therein. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide) that are formed with inorganic acids, such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, tartaric, mandelic and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases, such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like.
[000319] Physiologically tolerable carriers are well known in the art. Exemplary liquid carriers are sterile aqueous solutions that contain no materials in addition to the active ingredients and water, or contain a buffer such as sodium phosphate at physiological pH value, physiological saline or both, such as phosphate-buffered saline. Still further, aqueous carriers can contain more than one buffer salt, as well as salts such as sodium and potassium chlorides, dextrose, polyethylene glycol and other solutes. Liquid compositions can also contain liquid phases in addition to and to the exclusion of water. Exemplary of such additional liquid phases are glycerin, vegetable oils such as cottonseed oil, and water-oil emulsions. The amount of an active compound used in the cell compositions that is effective in the treatment of a particular disorder or condition can depend on the nature of the disorder or condition, and can be determined by standard clinical techniques.
Guide RNA Formulation
[000320] Guide RNAs of the present disclosure can be formulated with pharmaceutically acceptable excipients such as carriers, solvents, stabilizers, adjuvants, diluents, etc., depending upon the particular mode of administration and dosage form. Guide RNA compositions can be formulated to achieve a physiologically compatible pH, and range from a pH of about 3 to a pH of about 11, about pH 3 to about pH 7, depending on the formulation and route of administration. In some cases, the pH can be adjusted to a range from about pH 5.0 to about pH 8. In some cases, the compositions can comprise a therapeutically effective amount of at least one compound as described herein, together with one or more pharmaceutically acceptable excipients. Optionally, the compositions can comprise a combination of the compounds described herein, or can include a second active ingredient useful in the treatment or prevention of bacterial growth (for example and without limitation, anti -bacterial or anti -microbial agents), or can include a combination of reagents of the present disclosure.
[000321] Suitable excipients include, for example, carrier molecules that include large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Other exemplary excipients can include antioxidants (for example and without limitation, ascorbic acid), chelating agents (for example and without limitation, EDTA), carbohydrates (for example and without limitation, dextrin, hydroxyalkylcellulose, and hydroxyalkylmethylcellulose), stearic acid, liquids (for example and without limitation, oils, water, saline, glycerol and ethanol), wetting or emulsifying agents, pH buffering substances, and the like.
Delivery
[000322] Guide RNA polynucleotides (RNA or DNA) and/or endonuclease polynucleotide(s) (RNA or DNA) can be delivered by viral or non-viral delivery vehicles known in the art.
Alternatively, endonuclease polypeptide(s) can be delivered by viral or non-viral delivery vehicles known in the art, such as electroporation or lipid nanoparticles. In further alternative aspects, the DNA endonuclease can be delivered as one or more polypeptides, either alone or pre-complexed with one or more guide RNAs, or one or more crRNA together with a tracrRNA.
[000323] Polynucleotides can be delivered by non-viral delivery vehicles including, but not limited to, nanoparticles, liposomes, ribonucleoproteins, positively charged peptides, small molecule RNA-conjugates, aptamer-RNA chimeras, and RNA-fusion protein complexes. Some exemplary non-viral delivery vehicles are described in Peer and Lieberman, Gene Therapy, 18: 1127-1133 (2011) (which focuses on non-viral delivery vehicles for siRNA that are also useful for delivery of other polynucleotides).
[000324] For polynucleotides of the present disclosure, the formulation may be selected from any of those taught, for example, in International Application PCT7US2012/069610.
[000325] Polynucleotides, such as guide RNA, sgRNA, and mRNA encoding an endonuclease, may be delivered to a cell or a patient by a lipid nanoparticle (LNP).
[000326] A LNP refers to any particle having a diameter of less than 1000 nm, 500 nm, 250 nm, 200 nm, 150 nm, 100 nm, 75 nm, 50 nm, or 25 nm. Alternatively, a nanoparticle may range in size from 1-1000 nm, 1-500 nm, 1-250 nm, 25-200 nm, 25-100 nm, 35-75 nm, or 25-60 nm.
[000327] LNPs may be made from cationic, anionic, or neutral lipids. Neutral lipids, such as the fusogenic phospholipid DOPE or the membrane component cholesterol, may be included in LNPs as 'helper lipids' to enhance transfection activity and nanoparticle stability. Limitations of cationic lipids include low efficacy owing to poor stability and rapid clearance, as well as the generation of inflammatory or anti-inflammatory responses.
[000328] LNPs may also be comprised of hydrophobic lipids, hydrophilic lipids, or both hydrophobic and hydrophilic lipids.
[000329] Any lipid or combination of lipids that are known in the art can be used to produce a LNP. Examples of lipids used to produce LNPs are: DOTMA, DOSPA, DOTAP, DMRIE, DC- cholesterol, DOTAP-cholesterol, GAP-DMORIE-DPyPE, and GL67A-DOPE-DMPE- polyethylene glycol (PEG). Examples of cationic lipids are: 98N12-5, C 12-200, DLin-KC2- DMA (KC2), DLin-MC3 -DMA (MC3), XTC, MD1, and 7C1. Examples of neutral lipids are: DPSC, DPPC, POPC, DOPE, and SM. Examples of PEG-modified lipids are: PEG-DMG, PEG- CerC14, and PEG-CerC20.
[000330] The lipids can be combined in any number of molar ratios to produce a L P. In addition, the polynucleotide(s) can be combined with lipid(s) in a wide range of molar ratios to produce a LNP.
[000331] As stated previously, the site-directed polypeptide and genome-targeting nucleic acid can each be administered separately to a cell or a patient. On the other hand, the site-directed polypeptide can be pre-complexed with one or more guide RNAs, or one or more crRNA together with a tracrRNA. The pre-complexed material can then be administered to a cell or a patient. Such pre-complexed material is known as a ribonucleoprotein particle (RNP).
[000332] RNA is capable of forming specific interactions with RNA or DNA. While this property is exploited in many biological processes, it also comes with the risk of promiscuous interactions in a nucleic acid-rich cellular environment. One solution to this problem is the formation of ribonucleoprotein particles (RNPs), in which the RNA is pre-complexed with an endonuclease. Another benefit of the RNP is protection of the RNA from degradation.
[000333] The endonuclease in the RNP can be modified or unmodified. Likewise, the gRNA, crRNA, tracrRNA, or sgRNA can be modified or unmodified. Numerous modifications are known in the art and can be used.
[000334] The endonuclease and sgRNA can be generally combined in a 1 : 1 molar ratio.
Alternatively, the endonuclease, crRNA and tracrRNA can be generally combined in a 1 : 1 : 1 molar ratio. However, a wide range of molar ratios can be used to produce a RNP.
1 1 V (adeno associated virus)
[000335] A recombinant adeno-associated virus (AAV) vector can be used for delivery.
Techniques to produce rAAV particles, in which an AAV genome to be packaged that includes the polynucleotide to be delivered, rep and cap genes, and helper virus functions are provided to a cell are standard in the art. Production of rAAV typically requires that the following components are present within a single cell (denoted herein as a packaging cell): a rAAV genome, AAV rep and cap genes separate from {i.e., not in) the rAAV genome, and helper virus functions. The AAV rep and cap genes may be from any AAV serotype for which recombinant virus can be derived, and may be from a different AAV serotype than the rAAV genome ITRs, including, but not limited to, AAV serotypes described herein. Production of pseudotyped rAAV is disclosed in, for example, international patent application publication number WO
01/83692.
AA V Serotypes
[000336] AAV particles packaging polynucleotides encoding compositions of the present disclosure, e.g., endonucleases, donor sequences, or RNA guide molecules, of the present disclosure may comprise or be derived from any natural or recombinant AAV serotype.
According to the present disclosure, the AAV particles may utilize or be based on a serotype selected from any of the following serotypes, and variants thereof including but not limited to AAV1, AAV10, AAV106.1/hu.37, AAV11, AAV114.3/hu.40, AAV 12, AAV127.2/hu.41, AAV127.5/hu.42, AAV128.1/hu.43, AAV128.3/hu.44, AAV130.4/hu.48, AAV145.1/hu.53, AAV145.5/hu.54, AAV145.6/hu.55, AAV16.12/hu. l l, AAV16.3, AAV16.8/hu. lO,
AAV161.10/hu.60, AAV161.6/hu.61, AAVl-7/rh.48, AAVl-8/rh.49, AAV2, AAV2.5T, AAV2- 15/rh.62, AAV223.1, AAV223.2, AAV223.4, AAV223.5, AAV223.6, AAV223.7, AAV2- 3/rh.61, AAV24.1, AAV2-4/rh.50, AAV2-5/rh.51, AAV27.3, AAV29.3/bb. l, AAV29.5/bb.2, AAV2G9, AAV-2-pre-miRNA-101, AAV3, AAV3.1/hu.6, AAV3.1/hu.9, AAV3-1 l/rh.53,
AAV3-3, AAV33.12/hu. l7, AAV33.4/hu. l5, AAV33.8/hu. l6, AAV3-9/rh.52, AAV3a, AAV3b, AAV4, AAV4-19/rh.55, AAV42.12, AAV42-10, AAV42-11, AAV42-12, AAV42-13, AAV42- 15, AAV42-lb, AAV42-2, AAV42-3a, AAV42-3b, AAV42-4, AAV42-5a, AAV42-5b, AAV42- 6b, AAV42-8, AAV42-aa, AAV43-1, AAV43-12, AAV43-20, AAV43-21, AAV43-23, AAV43- 25, AAV43-5, AAV4-4, AAV44.1, AAV44.2, AAV44.5, AAV46.2/hu.28, AAV46.6/hu.29,
AAV4-8/rl l.64, AAV4-8/rh.64, AAV4-9/rh.54, AAV5, AAV52.1/hu.20, AAV52/hu. l9, AAV5- 22/rh.58, AAV5-3/rh.57, AAV54.1/hu.21, AAV54.2/hu.22, AAV54.4R/hu.27, AAV54.5/hu.23, AAV54.7/hu.24, AAV58.2/hu.25, AAV6, AAV6.1, AAV6.1.2, AAV6.2, AAV7, AAV7.2, AAV7.3/hu.7, AAV8, AAV-8b, AAV-8h, AAV9, AAV9.11, AAV9.13, AAV9.16, AAV9.24, AAV9.45, AAV9.47, AAV9.61, AAV9.68, AAV9.84, AAV9.9, AAV A3.3, AAV A3.4,
AAVA3.5, AAV A3.7, AAV-b, AAVCl, AAVC2, AAVC5, AAVCh.5, AAVCh.5Rl, AAVcy.2, AAVcy.3, AAVcy.4, AAVcy.5, AAVCy.5Rl, AAVCy.5R2, AAVCy.5R3, AAVCy.5R4, AAVcy.6, AAV-DJ, AAV-DJ8, AAVF3, AAVF5, AAV-h, AAVH-l/hu. l, AAVH2, AAVH- 5/hu.3, AAVH6, AAVhEl . l, AAVhER1.14, AAVhErl .16, AAVhErl .18, AAVhER1.23, AAVhErl .35, AAVhErl .36, AAVhErl .5, AAVhErl .7, AAVhErl .8, AAVhEr2.16,
AAVhEr2.29, AAVhEr2.30, AAVhEr2.31, AAVhEr2.36, AAVhEr2.4, AAVhEr3.1, AAVhu. l, AAVhu. lO, AAVhu. l l, AAVhu. l l, AAVhu.12, AAVhu.13, AAVhu.14/9, AAVhu.15,
AAVhu.16, AAVhu.17, AAVhu. l 8, AAVhu.19, AAVhu.2, AAVhu.20, AAVhu.21, AAVhu.22, AAVhu.23.2, AAVhu.24, AAVhu.25, AAVhu.27, AAVhu.28, AAVhu.29, AAVhu.29R, AAVhu.3, AAVhu.31, AAVhu.32, AAVhu.34, AAVhu.35, AAVhu.37, AAVhu.39, AAVhu.4, AAVhu.40, AAVhu.41, AAVhu.42, AAVhu.43, AAVhu.44, AAVhu.44Rl, AAVhu.44R2, AAVhu.44R3, AAVhu.45, AAVhu.46, AAVhu.47, AAVhu.48, AAVhu.48Rl, AAVhu.48R2, AAVhu.48R3, AAVhu.49, AAVhu.5, AAVhu.51, AAVhu.52, AAVhu.53, AAVhu.54,
AAVhu.55, AAVhu.56, AAVhu.57, AAVhu.58, AAVhu.6, AAVhu.60, AAVhu.61, AAVhu.63, AAVhu.64, AAVhu.66, AAVhu.67, AAVhu.7, AAVhu.8, AAVhu.9, AAVhu.t 19, AAVLG- 10/rh.40, AAVLG-4/rh.38, AAVLG-9/hu.39, AAVLG-9/hu.39, AAV-LK01, AAV-LK02, AAVLK03, AAV-LK03, AAV-LK04, AAV-LK05, AAV-LK06, AAV-LK07, AAV-LK08, AAV-LK09, AAV-LK10, AAV-LK11, AAV-LK12, AAV-LK13, AAV-LK14, AAV-LK15,
AAV-LK17, AAV-LK18, AAV-LK19, AAVN721-8/rh.43, AAV-PAEC, AAV-PAEC11, AAV- PAEC12, AAV-PAEC2, AAV-PAEC4, AAV-PAEC6, AAV-PAEC7, AAV-PAEC 8, AAVpi. l, AAVpi.2, AAVpi.3, AAVrh. lO, AAVrh.12, AAVrh.13, AAVrh. l3R, AAVrh.14, AAVrh.17, AAVrh.18, AAVrh.19, AAVrh.2, AAVrh.20, AAVrh.21, AAVrh.22, AAVrh.23, AAVrh.24, AAVrh.25, AAVrh.2R, AAVrh.31, AAVrh.32, AAVrh.33, AAVrh.34, AAVrh.35, AAVrh.36, AAVrh.37, AAVrh.37R2, AAVrh.38, AAVrh.39, AAVrh.40, AAVrh.43, AAVrh.44, AAVrh.45, AAVrh.46, AAVrh.47, AAVrh.48, AAVrh.48, AAVrh.48.1, AAVrh.48.1.2, AAVrh.48.2, AAVrh.49, AAVrh.50, AAVrh.51, AAVrh.52, AAVrh.53, AAVrh.54, AAVrh.55, AAVrh.56, AAVrh.57, AAVrh.58, AAVrh.59, AAVrh.60, AAVrh.61, AAVrh.62, AAVrh.64, AAVrh.64Rl, AAVrh.64R2, AAVrh.65, AAVrh.67, AAVrh.68, AAVrh.69, AAVrh.70, AAVrh.72, AAVrh.73, AAVrh.74, AAVrh.8, AAVrh.8R, AAVrh8R, AAVrh8R A586R mutant, AAVrh8R R533A mutant, BAAV, B P61 AAV, B P62 AAV, B P63 AAV, bovine AAV, caprine AAV, Japanese AAV 10, true type AAV (ttAAV), UPENN AAV 10, AAV-LK16, AAAV, AAV Shuffle 100-1, AAV Shuffle 100-2, AAV Shuffle 100-3, AAV Shuffle 100-7, AAV Shuffle 10- 2, AAV Shuffle 10-6, AAV Shuffle 10-8, AAV SM 100-10, AAV SM 100-3, AAV SM 10-1, AAV SM 10-2, and/or AAV SM 10-8.
[000337] In some aspects, the AAV serotype may be, or have, a mutation in the AAV9 sequence as described by N Pulicherla et al. (Molecular Therapy 19(6): 1070-1078 (2011)), such as but not limited to, AAV9.9, AAV9.11, AAV9.13, AAV9.16, AAV9.24, AAV9.45, AAV9.47, AAV9.61, AAV9.68, AAV9.84.
[000338] In some aspects, the AAV serotype may be, or have, a sequence as described in U.S. Patent No. U.S. 6156303, such as, but not limited to, AAV3B (SEQ ID NO: 1 and 10 of US 6156303), AAV6 (SEQ ID NO: 2, 7 and 11 of U.S. 6156303), AAV2 (SEQ ID NO: 3 and 8 of U.S. 6156303), AAV3A (SEQ ID NO: 4 and 9, of U.S. 6156303), or derivatives thereof.
[000339] In some aspects, the serotype may be AAVDJ or a variant thereof, such as AAVDJ8 (or AAV-DJ8), as described by Grimm et al. (Journal of Virology 82(12): 5887-5911 (2008)). The amino acid sequence of AAVDJ8 may comprise two or more mutations in order to remove the heparin binding domain (HBD). As a non-limiting example, the AAV-DJ sequence described as SEQ ID NO: 1 in U.S. Patent No. 7,588,772, may comprise two mutations: (1) R587Q where arginine (R; Arg) at amino acid 587 is changed to glutamine (Q; Gin) and (2) R590T where arginine (R; Arg) at amino acid 590 is changed to threonine (T; Thr). As another non-limiting example, may comprise three mutations: (1) K406R where lysine (K; Lys) at amino acid 406 is changed to arginine (R; Arg), (2) R587Q where arginine (R; Arg) at amino acid 587 is changed to glutamine (Q; Gin) and (3) R590T where arginine (R; Arg) at amino acid 590 is changed to threonine (T; Thr).
[000340] In some aspects, the AAV serotype may be, or have, a sequence as described in International Publication No. WO2015121501, such as, but not limited to, true type AAV
(ttAAV) (SEQ ID NO: 2 of WO2015121501), "UPenn AAV10" (SEQ ID NO: 8 of
WO2015121501), "Japanese AAV10" (SEQ ID NO: 9 of WO2015121501), or variants thereof.
[000341] According to the present disclosure, AAV capsid serotype selection or use may be from a variety of species. In one example, the AAV may be an avian AAV (AAAV). The AAAV serotype may be, or have, a sequence as described in U.S. Patent No. U.S. 9238800, such as, but not limited to, AAAV (SEQ ID NO: 1, 2, 4, 6, 8, 10, 12, and 14 of U.S. 9,238,800), or variants thereof.
[000342] In one example, the AAV may be a bovine AAV (BAAV). The BAAV serotype may be, or have, a sequence as described in U.S. Patent No. U.S. 9,193,769, such as, but not limited to, BAAV (SEQ ID NO: 1 and 6 of U.S. 9193769), or variants thereof. The BAAV serotype may be or have a sequence as described in U.S. Patent No. U.S.7427396, such as, but not limited to, BAAV (SEQ ID NO: 5 and 6 of U.S.7427396), or variants thereof.
[000343] In one example, the AAV may be a caprine AAV. The caprine AAV serotype may be, or have, a sequence as described in U.S. Patent No. U.S.7427396, such as, but not limited to, caprine AAV (SEQ ID NO: 3 of U.S.7427396), or variants thereof.
[000344] In other examples the AAV may be engineered as a hybrid AAV from two or more parental serotypes. In one example, the AAV may be AAV2G9 which comprises sequences from AAV2 and AAV9. The AAV2G9 AAV serotype may be, or have, a sequence as described in U.S. Patent Publication No. U.S. 20160017005.
[000345] In one example, the AAV may be a serotype generated by the AAV9 capsid library with mutations in amino acids 390-627 (VPl numbering) as described by Pulicherla et al.
(Molecular Therapy 19(6): 1070-1078 (2011). The serotype and corresponding nucleotide and amino acid substitutions may be, but is not limited to, AAV9.1 (G1594C; D532H), AAV6.2 (T1418A and T1436X; V473D and I479K), AAV9.3 (T1238A; F413Y), AAV9.4 (T1250C and A1617T; F417S), AAV9.5 (A1235G, A1314T, A1642G, C1760T; Q412R, T548A, A587V), AAV9.6 (T1231A; F411I), AAV9.9 (G1203A, G1785T; W595C), AAV9.10 (A1500G, T1676C; M559T), AAV9.11 (A1425T, A1702C, A1769T; T568P, Q590L), AAV9.13 (A1369C, A1720T; N457H, T574S), AAV9.14 (T1340A, T1362C, T1560C, G1713A; L447H), AAV9.16 (A1775T; Q592L), AAV9.24 (T1507C, T1521G; W503R), AAV9.26 (A1337G, A1769C; Y446C, Q590P), AAV9.33 (A1667C; D556A), AAV9.34 (A1534G, C1794T; N512D), AAV9.35 (A1289T, T1450A, C1494T, A1515T, C1794A, G1816A; Q430L, Y484N, N98K, V606I), AAV9.40 (A1694T, E565V), AAV9.41 (A1348T, T1362C; T450S), AAV9.44 (A1684C, A1701T,
A1737G; N562H, K567N), AAV9.45 (A1492T, C1804T; N498Y, L602F), AAV9.46 (G1441C, T1525C, T1549G; G481R, W509R, L517V), 9.47 (G1241A, G1358A, A1669G, C1745T;
S414N, G453D, K557E, T582I), AAV9.48 (C1445T, A1736T; P482L, Q579L), AAV9.50 (A1638T, C1683T, T1805A; Q546H, L602H), AAV9.53 (G1301A, A1405C, C1664T, G1811T; R134Q, S469R, A555V, G604V), AAV9.54 (C1531A, T1609A; L511I, L537M), AAV9.55
(T1605A; F535L), AAV9.58 (C1475T, C1579A; T492I, H527N), AAV.59 (T1336C; Y446H), AAV9.61 (A1493T; N498I), AAV9.64 (C1531A, A1617T; L511I), AAV9.65 (C1335T, T1530C, C1568A; A523D), AAV9.68 (C1510A; P504T), AAV9.80 (G1441A,;G481R), AAV9.83 (C1402A, A1500T; P468T, E500D), AAV9.87 (T1464C, T1468C; S490P), AAV9.90 (A1196T; Y399F), AAV9.91 (T1316G, A1583T, C1782G, T1806C; L439R, K528I), AAV9.93 (A1273G, A1421G, A1638C, C1712T, G1732A, A1744T, A1832T; S425G, Q474R, Q546H, P571L, G578R, T582S, D611V), AAV9.94 (A1675T; M559L) and AAV9.95 (T1605A; F535L).
[000346] In one example, the AAV may be a serotype comprising at least one AAV capsid CD8+ T-cell epitope. As a non-limiting example, the serotype may be AAV1, AAV2 or AAV8.
[000347] In one example, the AAV may be a variant, such as PUP. A or PUP.B as described in Deverman. 2016. Nature Biotechnology. 34(2): 204-209.
[000348] In one example, the AAV may be a serotype selected from any of those found in SEQ ID NOs: 4734-5302 and Table 2. [000349] In one example, the AAV may be encoded by a sequence, fragment or variant as disclosed in SEQ ID NOs: 4734-5302 and Table 2.
[000350] General principles of rAAV production are reviewed in, for example, Carter, 1992, Current Opinions in Biotechnology, 1533-539; and Muzyczka, 1992, Curr. Topics in Microbial. and Immunol., 158:97-129). Various approaches are described in Ratschin et al., Mol. Cell. Biol. 4:2072 (1984); Hermonat et al, Proc. Natl. Acad. Sci. USA, 81 :6466 (1984); Tratschin et al, Mol . Cell. Biol. 5:3251 (1985); McLaughlin et al, J. Virol., 62: 1963 (1988); and Lebkowski et al, 1988 Mol. Cell. Biol., 7:349 (1988). Samulski et al (1989, J. Virol., 63 :3822-3828); U.S. Patent No. 5,173,414; WO 95/13365 and corresponding U.S. Patent No. 5,658,776; WO
95/13392; WO 96/17947; PCT/US98/18600; WO 97/09441 (PCT/US96/14423); WO 97/08298 (PCT/US96/13872); WO 97/21825 (PCT/US96/20777); WO 97/06243 (PCT/FR96/01064); WO 99/11764; Perrin et al. (1995) Vaccine 13 : 1244-1250; Paul et al. (1993) Human Gene Therapy 4:609-615; Clark et al. (1996) Gene Therapy 3 : 1124-1132; U.S. Patent. No. 5,786,211; U.S. Patent No. 5,871,982; and U.S. Patent. No. 6,258,595.
[000351] AAV vector serotypes can be matched to target cell types. For example, the following exemplary cell types can be transduced by the indicated AAV serotypes among others.
Table 2. Tissue/Cell Types and Serotypes
Figure imgf000081_0001
[000352] In addition to adeno-associated viral vectors, other viral vectors can be used. Such viral vectors include, but are not limited to, lentivirus, alphavirus, enterovirus, pestivirus, baculovirus, herpesvirus, Epstein Barr virus, papovavirus, poxvirus, vaccinia virus, and herpes simplex virus. [000353] In some aspects, Cas9 mRNA, sgRNA targeting one or two sites in ATXN3 gene, and donor DNA can each be separately formulated into lipid nanoparticles, or are all co-formulated into one lipid nanoparticle.
[000354] In some aspects, Cas9 mRNA can be formulated in a lipid nanoparticle, while sgRNA and donor DNA can be delivered in an AAV vector.
[000355] Options are available to deliver the Cas9 nuclease as a DNA plasmid, as mRNA or as a protein. The guide RNA can be expressed from the same DNA, or can also be delivered as an RNA. The RNA can be chemically modified to alter or improve its half-life, or decrease the likelihood or degree of immune response. The endonuclease protein can be complexed with the gRNA prior to delivery. Viral vectors allow efficient delivery; split versions of Cas9 and smaller orthologs of Cas9 can be packaged in AAV, as can donors for HDR. A range of non-viral delivery methods also exist that can deliver each of these components, or non-viral and viral methods can be employed in tandem. For example, nano-particles can be used to deliver the protein and guide RNA, while AAV can be used to deliver a donor DNA.
IV. DOSING AND ADMINISTRATION
[000356] The terms "administering," "introducing" and "transplanting" are used
interchangeably in the context of the placement of cells, e.g., progenitor cells, into a subject, by a method or route that results in at least partial localization of the introduced cells at a desired site, such as a site of injury or repair, such that a desired effect(s) is produced. The cells e.g., progenitor cells, or their differentiated progeny can be administered by any appropriate route that results in delivery to a desired location in the subject where at least a portion of the implanted cells or components of the cells remain viable. The period of viability of the cells after administration to a subject can be as short as a few hours, e.g., twenty-four hours, to a few days, to as long as several years, or even the life time of the patient, i.e., long-term engraftment. For example, in some aspects described herein, an effective amount of neural progenitor cells is administered via a systemic route of administration, such as an intraperitoneal or intravenous route.
[000357] The terms "individual," "subject," "host" and "patient" are used interchangeably herein and refer to any subject for whom diagnosis, treatment or therapy is desired. In some aspects, the subject is a mammal. In some aspects, the subject is a human being.
[000358] When provided prophylactically, progenitor cells described herein can be
administered to a subject in advance of any symptom of Spinocerebellar ataxia 3 (SCA3). Accordingly, the prophylactic administration of a progenitor cell population serves to prevent Spinocerebellar ataxia 3 (SCA3).
[000359] A progenitor cell population being administered according to the methods described herein can comprise allogeneic progenitor cells obtained from one or more donors. Such progenitors may be of any cellular or tissue origin, e.g., liver, muscle, cardiac, brain, etc.
"Allogeneic" refers to a progenitor cell or biological samples comprising progenitor cells obtained from one or more different donors of the same species, where the genes at one or more loci are not identical. For example, a liver progenitor cell population being administered to a subject can be derived from one more unrelated donor subjects, or from one or more non- identical siblings. In some cases, syngeneic progenitor cell populations can be used, such as those obtained from genetically identical animals, or from identical twins. The progenitor cells can be autologous cells; that is, the progenitor cells are obtained or isolated from a subject and administered to the same subject, i.e., the donor and recipient are the same.
[000360] The term "effective amount" refers to the amount of a population of progenitor cells or their progeny needed to prevent or alleviate at least one or more signs or symptoms of
Spinocerebellar ataxia 3 (SCA3), and relates to a sufficient amount of a composition to provide the desired effect, e.g., to treat a subject having Spinocerebellar ataxia 3 (SCA3). The term "therapeutically effective amount" therefore refers to an amount of progenitor cells or a composition comprising progenitor cells that is sufficient to promote a particular effect when administered to a typical subject, such as one who has or is at risk for Spinocerebellar ataxia 3 (SCA3). An effective amount would also include an amount sufficient to prevent or delay the development of a symptom of the disease, alter the course of a symptom of the disease (for example but not limited to, slow the progression of a symptom of the disease), or reverse a symptom of the disease. It is understood that for any given case, an appropriate "effective amount" can be determined by one of ordinary skill in the art using routine experimentation.
[000361] For use in the various aspects described herein, an effective amount of progenitor cells comprises at least 102 progenitor cells, at least 5 X 102 progenitor cells, at least 103 progenitor cells, at least 5 X 103 progenitor cells, at least 104 progenitor cells, at least 5 X 104 progenitor cells, at least 105 progenitor cells, at least 2 X 105 progenitor cells, at least 3 X 105 progenitor cells, at least 4 X 105 progenitor cells, at least 5 X 105 progenitor cells, at least 6 X 105 progenitor cells, at least 7 X 105 progenitor cells, at least 8 X 105 progenitor cells, at least 9 X 105 progenitor cells, at least 1 X 106 progenitor cells, at least 2 X 106 progenitor cells, at least 3 X 106 progenitor cells, at least 4 X 106 progenitor cells, at least 5 X 106 progenitor cells, at least 6 X 106 progenitor cells, at least 7 X 106 progenitor cells, at least 8 X 106 progenitor cells, at least 9 X 106 progenitor cells, or multiples thereof. The progenitor cells can be derived from one or more donors, or can be obtained from an autologous source. In some examples described herein, the progenitor cells can be expanded in culture prior to administration to a subject in need thereof.
[000362] In some aspects, reduction of the expanded trinucleotide repeats in the ATXN3 gene in cells of patients having Spinocerebellar ataxia 3 (SCA3) can be beneficial for ameliorating one or more symptoms of the disease, for increasing long-term survival, and/or for reducing side effects associated with other treatments. Upon administration of such cells to human patients, the presence of progenitors that have wild-type or similar levels of the trinucleotide repeat in the ATXN3gene is beneficial. In some cases, effective treatment of a subject gives rise to at least about 3%, 5% or 7% transcripts having wild-type or similar levels of trinucleotide repeat relative to total ATXN3 transcripts in the treated subject. In some examples, transcripts having wild- type or similar levels of trinucleotide repeat will be at least about 10% of total ATXN3 transcripts. In some examples, transcripts having wild-type or similar levels of trinucleotide repeat will be at least about 20% to 30%> of total ATXN3 transcripts. Similarly, the introduction of even relatively limited subpopulations of cells having wild-type levels of trinucleotide repeat in the ATXN3 gene can be beneficial in various patients because in some situations normalized cells will have a selective advantage relative to diseased cells. However, even modest levels of progenitors with wild-type or similar levels of trinucleotide repeat in the ATXN3 gene can be beneficial for ameliorating one or more aspects of Spinocerebellar ataxia 3 (SCA3) in patients. In some examples, about 10%>, about 20%, about 30%>, about 40%, about 50%, about 60%, about 70%), about 80%), about 90% or more of the neural progenitors in patients to whom such cells are administered have wild-type or similar levels of trinucleotide repeat in the ATXN3 gene.
[000363] "Administered" refers to the delivery of a progenitor cell composition into a subject by a method or route that results in at least partial localization of the cell composition at a desired site. A cell composition can be administered by any appropriate route that results in effective treatment in the subject, i.e. administration results in delivery to a desired location in the subject where at least a portion of the composition delivered, i.e. at least 1 x 104 cells are delivered to the desired site for a period of time.
[000364] In one aspect of the method, the pharmaceutical composition may be administered via a route such as, but not limited to, enteral (into the intestine), gastroenteral, epidural (into the dura matter), oral (by way of the mouth), transdermal, peridural, intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intravenous bolus, intravenous drip, intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavernous injection (into a pathologic cavity) intracavitary (into the base of the penis), intravaginal administration, intrauterine, extra-amniotic administration, transdermal (diffusion through the intact skin for systemic distribution), transmucosal (diffusion through a mucous membrane), transvaginal, insufflation (snorting), sublingual, sublabial, enema, eye drops (onto the conjunctiva), in ear drops, auricular (in or by way of the ear), buccal (directed toward the cheek), conjunctival, cutaneous, dental (to a tooth or teeth), electro-osmosis, endocervical, endosinusial, endotracheal, extracorporeal, hemodialysis, infiltration, interstitial, intra-abdominal, intra- amniotic, intra-articular, intrabiliary, intrabronchial, intrabursal, intracartilaginous (within a cartilage), intracaudal (within the cauda equine), intraci sternal (within the cisterna magna cerebellomedularis), intracorneal (within the cornea), dental intracornal, intracoronary (within the coronary arteries), intracorporus cavernosum (within the dilatable spaces of the corporus cavernosa of the penis), intradiscal (within a disc), intraductal (within a duct of a gland), intraduodenal (within the duodenum), intradural (within or beneath the dura), intraepidermal (to the epidermis), intraesophageal (to the esophagus), intragastric (within the stomach),
intragingival (within the gingivae), intraileal (within the distal portion of the small intestine), intralesional (within or introduced directly to a localized lesion), intraluminal (within a lumen of a tube), intralymphatic (within the lymph), intramedullary (within the marrow cavity of a bone), intrameningeal (within the meninges), intramyocardial (within the myocardium), intraocular (within the eye), intraovarian (within the ovary), intrapericardial (within the pericardium), intrapleural (within the pleura), intraprostatic (within the prostate gland), intrapulmonary (within the lungs or its bronchi), intrasinal (within the nasal or periorbital sinuses), intraspinal (within the vertebral column), intrasynovial (within the synovial cavity of a joint), intratendinous (within a tendon), intratesticular (within the testicle), intrathecal (within the cerebrospinal fluid at any level of the cerebrospinal axis), intrathoracic (within the thorax), intratubular (within the tubules of an organ), intratumor (within a tumor), intratympanic (within the aurus media), intravascular (within a vessel or vessels), intraventricular (within a ventricle), iontophoresis (by means of electric current where ions of soluble salts migrate into the tissues of the body), irrigation (to bathe or flush open wounds or body cavities), laryngeal (directly upon the larynx), nasogastric (through the nose and into the stomach), occlusive dressing technique (topical route
administration, which is then covered by a dressing that occludes the area), ophthalmic (to the external eye), oropharyngeal (directly to the mouth and pharynx), parenteral, percutaneous, periarticular, peridural, perineural, periodontal, rectal, respiratory (within the respiratory tract by inhaling orally or nasally for local or systemic effect), retrobulbar (behind the pons or behind the eyeball), intramyocardial (entering the myocardium), soft tissue, subarachnoid, subconjunctival, submucosal, topical, transplacental (through or across the placenta), transtracheal (through the wall of the trachea), transtympanic (across or through the tympanic cavity), ureteral (to the ureter), urethral (to the urethra), vaginal, caudal block, diagnostic, nerve block, biliary perfusion, cardiac perfusion, photopheresis and spinal.
[000365] Modes of administration include injection, infusion, instillation, and/or ingestion. "Injection" includes, without limitation, intravenous, intramuscular, intra-arterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal,
transtracheal, subcutaneous, subcuticular, intraarticular, sub capsular, subarachnoid, intraspinal, intracerebro spinal, and intrasternal injection and infusion. In some examples, the route is intravenous. For the delivery of cells, administration by injection or infusion can be made.
[000366] The cells can be administered systemically. The phrases "systemic administration," "administered systemically", "peripheral administration" and "administered peripherally" refer to the administration of a population of progenitor cells other than directly into a target site, tissue, or organ, such that it enters, instead, the subject's circulatory system and, thus, is subject to metabolism and other like processes.
[000367] The efficacy of a treatment comprising a composition for the treatment of
Spinocerebellar ataxia 3 (SCA3) can be determined by the skilled clinician. However, a treatment is considered "effective treatment," if any one or all of the signs or symptoms of, as but one example, levels of trinucleotide repeat in the ATXN3 gene are altered in a beneficial manner (e.g., decreased by at least 10%), or other clinically accepted symptoms or markers of disease are improved or ameliorated. Efficacy can also be measured by failure of an individual to worsen as assessed by hospitalization or need for medical interventions (e.g., progression of the disease is halted or at least slowed). Methods of measuring these indicators are known to those of skill in the art and/or described herein. Treatment includes any treatment of a disease in an individual or an animal (some non-limiting examples include a human, or a mammal) and includes: (1) inhibiting the disease, e.g., arresting, or slowing the progression of symptoms; or (2) relieving the disease, e.g., causing regression of symptoms; and (3) preventing or reducing the likelihood of the development of symptoms.
[000368] The treatment according to the present disclosure can ameliorate one or more symptoms associated with Spinocerebellar ataxia 3 (SCA3) by reducing the number of trinucleotide repeat in the ATXN3 gene in the individual.
V. FEATURES AND PROPERTIES OF THE SPINOCEREBELLAR ATAXIA TYPE 3 PROTEIN (ATXN3) GENE
[000369] ATXN3 has been associated with diseases and disorders such as, but not limited to, Congenital Abnormality, Alzheimer's Disease, Amyotrophic Lateral Sclerosis, Ataxia, Ataxia Telangiectasia, Cerebellar Ataxia, Cerebellar Diseases, Chorea, Cleft Palate, Cystic Fibrosis, Mental Depression, Depressive disorder, Dystonia, Esophageal Neoplasms, Exotropia, Cardiac Arrest, Huntington Disease, Machado- Joseph Disease, Movement Disorders, Muscular
Dystrophy, Myotonic Dystrophy, Narcolepsy, Nerve Degeneration, Neuroblastoma, Parkinson Disease, Peripheral Neuropathy, Restless Legs Syndrome, Retinal Degeneration, Retinitis Pigmentosa, Schizophrenia, Shy-Drager Syndrome, Sleep disturbances, Hereditary Spastic Paraplegia, Thromboembolism, Stiff-Person Syndrome, Spinocerebellar Ataxia, Esophageal carcinoma, Polyneuropathy, Effects of heat, Muscle twitch, Extrapyramidal sign, Ataxic, Neurologic Symptoms, Cerebral atrophy, Parkinsonian Disorders, Protein S Deficiency, Cerebellar degeneration, Familial Amyloid Neuropathy Portuguese Type, Spastic syndrome, Vertical Nystagmus, Nystagmus End-Position, Antithrombin III Deficiency, Atrophic,
Complicated hereditary spastic paraplegia, Multiple System Atrophy, Pallidoluysian
degeneration, Dystonia Disorders, Pure Autonomic Failure, Thrombophilia, Protein C
Deficiency, Congenital Myotonic Dystrophy, Motor symptoms, Neuropathy, Neurodegenerative Disorders, Malignant neoplasm of esophagus, Visual disturbance, Activated Protein C
Resistance, Terminal illness, Myokymia, Central neuroblastoma, Dyssomnias, Appendicular Ataxia, Narcolepsy-Cataplexy Syndrome, Machado- Joseph Disease Type I, Machado- Joseph Disease Type II, Machado- Joseph Disease Type III, Dentatorubral-Pallidoluysian Atrophy, Gait Ataxia, Spinocerebellar Ataxia Type 1, Spinocerebellar Ataxia Type 2, Spinocerebellar Ataxia Type 6 (disorder), Spinocerebellar Ataxia Type 7, Muscular Spinobulbar Atrophy, Genomic Instability, Episodic ataxia type 2 (disorder), Bulbo-Spinal Atrophy X-Linked, Fragile X Tremor/ Ataxia Syndrome, Thrombophilia Due to Activated Protein C Resistance (Disorder), Amyotrophic Lateral Sclerosis 1, Neuronal Intranuclear Inclusion Disease, Hereditary Antithrombin Iii Deficiency, and Late-Onset Parkinson Disease. Editing the ATXN3 gene using any of the methods described herein may be used to treat, prevent and/or mitigate the symptoms of the diseases and disorders described herein.
[000370] The activity of ATXN3 is largely confined to central nervous system and ATXN3 is associated with Spinocerebellar ataxia 3 (SCA3) (also commonly known as Machado-Joseph disease). Spinocerebellar ataxia 3 (SCA3) is caused by expansion of a trinucleotide CAG repeat in the ATXN3 gene. This CAG repeat region is located in exon 10 of the ATXN3 gene and encodes a polyglutamine tract. In most people, the CAG trinucleotide is repeated 12 to 43 times. In individuals with SCA3, the number of CAG repeat is greater than 50. People with
intermediate numbers of the CAG repeat (about 44-52) may or may not develop the disease. The number of repeats in the gene correlates with the age of onset and severity of the disease. People with 75 or fewer repeats tend to develop signs and symptoms of SCA3 in mid-adulthood, while people with around 80 repeats usually have signs and symptoms by their teens. The abnormal repeat expansion leads to the production of an altered version of the ATXN3 protein, which is nonfunctional and aggregates with other proteins in the nucleus, causing toxicity to neuronal cells.
[000371] Spinocerebellar ataxia 3 (SCA3) is an inherited disease (autosomal dominant) where the body experiences degeneration of cells in the hindbrain. Spinocerebellar ataxia 3 (SCA3) affects about 1-2 in 100,000 people worldwide and is more common in Azorean populations. Common symptoms of Spinocerebellar ataxia 3 (SCA3) includes memory deficit, spasticity, difficulty with speech and swallowing, weakness in arms and legs, clumsiness, frequent urination and involuntary eye movements. Currently there are no clinical interventions to prevent the onset of the disease symptoms. Yet, symptoms may be treated using levodopa therapy to ease stiffness and slowness of movements; antispasmodic drugs, such as baclofen, to reduce spasticity; botulinum toxin to reduce severe spasticity and dystonia; speech therapy to improve speech problems, such as dysarthria and dysphagia; prism glasses to reduce blurred or double vision; and physiotherapy to cope with disability associated with gait problems.
[000372] In one example, the target tissue for the compositions and methods described herein is central nervous system tissue.
[000373] In one example, the gene is Spinocerebellar Ataxia Type 3 Protein (ATXN3) which may also be referred to as Ataxin 3, Machado-Joseph Disease (Spinocerebellar Ataxia 3, Olivopontocerebellar Ataxia 3, Autosomal Dominant, Ataxin 3), Spinocerebellar Ataxia Type 3 Protein, Machado-Joseph Disease Protein 1, SCA3, ATX3, MJDl, Olivopontocerebellar Ataxia, Ataxin 3 Variant Ref, Ataxin 3 Variant An, Ataxin 3 Variant Ao, Ataxin 3 Variant At, Ataxin 3 Variant E, Ataxin 3 Variant H, Ataxin 3 Variant M, Ataxin 3 Variant R, Ataxin 3 Variant Y, EC 3.4.19.12, EC 3.4.22, Josephin, AT3, and JOS. ATXN3 has a cytogenetic location of 14q32.12 and the genomic coordinate are on Chromosome 14 on the forward strand at position
92,038,652-92,106,621. The nucleotide sequence of ATXN3 is shown as SEQ ID NO: 5303. NDUFB 1 is the gene upstream of ATXN3 on the reverse strand and TRIP 11 is a gene downstream of and overlapping ATXN3 on the reverse strand. FBLN5 is also a gene downstream of ATXN3, but does not overlap ATXN3. ATXN3 has a NCBI gene ID of 4287, Uniprot ID of P54252 and Ensembl Gene ID of ENSG00000066427. ATXN3 has 2939 SNPs, 117 introns and 80 exons. The exon identifier from Ensembl and the start/stop sites of the introns and exons are shown in Table 3.
Table 3. Introns and Exons for ATXN3
Exon Exon ID Start/Stop Intron Intron based on Exon ID Start/Stop No. No.
EX1 ENSE00002073369 92,106,610 - INT1 Intron ENSE00002073369 - 92,106,528 - 92,106,529 ENSE00003646843 92,096,839
EX2 ENSE00003646843 92,096,838 - INT2 Intron ENSE00003646843 - 92,096,673 - 92,096,674 ENSE00003649810 92,096,138
EX3 ENSE00003649810 92,096, 137 - INT3 Intron ENSE00003649810 - 92,096,092 - 92,096,093 ENSE00003499200 92,093,832
EX4 ENSE00003499200 92,093,831 - INT4 Intron ENSE00003499200 - 92,093,745 - 92,093,746 ENSE00003477115 92,093,319
EX5 ENSE00003477115 92,093,318 - INT5 Intron ENSE00003477115 - 92,093,251 - 92,093,252 ENSE00003574977 92,088,818
EX6 ENSE00003574977 92,088,817 - INT6 Intron ENSE00003574977 - 92,088,729 - 92,088,730 ENSE00003530935 92,083,259
EX7 ENSE00003530935 92,083,258 - INT7 Intron ENSE00003530935 - 92,083,125 - 92,083, 126 ENSE00003688920 92,082,467
EX8 ENSE00003688920 92,082,466 - INT8 Intron ENSE00003688920 - 92,082,299 - 92,082,300 ENSE00003654296 92,081,062
EX9 ENSE00003654296 92,081,061 - INT9 Intron ENSE00003654296 - 92,080,964 - 92,080,965 ENSE00003788002 92,071,054
EX10 ENSE00003788002 92,071,053 - INT10 Intron ENSE00003788002 - 92,070,934 - 92,070,935 ENSE00002042510 92,064,415
EX11 ENSE00002042510 92,064,414 - INT11 Intron ENSE00001263799 - 92,106,528 - 92,038,652 ENSE00003499200 92,093,832
EX12 ENSE00001263799 92,106,621 - INT12 Intron ENSE00003788002 - 92,070,934 - 92,106,529 ENSE00001383144 92,064,415
EX13 ENSE00001383144 92,064,414 - INT13 Intron ENSE00002060504 - 92,106,528 - 92,058,552 ENSE00003646843 92,096,839
EX14 ENSE00002060504 92,106,600 - INT14 Intron ENSE00003646843 - 92,096,673 - 92,106,529 ENSE00003499200 92,093,832
EX15 ENSE00002063519 92,064,414 - INT15 Intron ENSE00003788002 - 92,070,934 - 92,064,094 ENSE00002063519 92,064,415
EX16 ENSE00002040834 92,106,582 - INT16 Intron ENSE00002040834 - 92,106,528 - 92,106,529 ENSE00003645711 92,093,319 EX17 ENSE00003645711 92,093,318 - INT17 Intron ENSE00003645711 - 92,093,251 - 92,093,252 ENSE00003628190 92,088,818
EX18 ENSE00003628190 92,088,817 - INT18 Intron ENSE00003628190 - 92,088,729 - 92,088,730 ENSE00003616151 92,083,259
EX19 ENSE00003616151 92,083,258 - INT19 Intron ENSE00003616151 - 92,083,125 - 92,083,126 ENSE00003688920 92,082,467
EX20 ENSE00002025336 92,064,414 - INT20 Intron ENSE00003788002 - 92,070,934 - 92,063,881 ENSE00002025336 92,064,415
EX21 ENSE00003723395 92,080,567 - INT21 IntronENSE00002073369 - 92,106,528 - 92,080,541 ENSE00003649810 92,096,138
EX22 ENSE00002164245 92,106,562 - INT22 IntronENSE00003788002 - 92,070,934 - 92,106,529 ENSE00002063519 92,064,415
EX23 ENSE00002149841 92,071,053 - INT23 Intron ENSE00001263799 - 92,106,528 - 92,070,745 ENSE00003646843 92,096,839
EX24 ENSE00003543114 92,106,574 - INT24 Intron ENSE00003654296 - 92,080,964 - 92,106,529 ENSE00003723395 92,080,568
EX25 ENSE00003723582 92,079,468 - INT25 Intron ENSE00003723395 - 92,080,540 - 92,079,415 ENSE00003788002 92,071,054
EX26 ENSE00001514734 92,071,063 - INT26 Intron ENSE00003649810 - 92,096,092 - 92,070,938 ENSE00003574977 92,088,818
EX27 ENSE00003715808 92,077,602 - INT27 Intron ENSE00002164245 - 92,106,528 - 92,077,581 ENSE00003646843 92,096,839
EX28 ENSE00003713862 92,073,536 - INT28 Intron ENSE00003654296 - 92,080,964 - 92,073,532 ENSE00002149841 92,071,054
EX29 ENSE00003746704 92,071,053 - INT29 Intron ENSE00003543114 - 92,106,528 - 92,071,008 ENSE00003646843 92,096,839
EX30 ENSE00003739428 92,071,005 - INT30 Intron ENSE00003654296 - 92,080,964 - 92,070,976 ENSE00003723582 92,079,469
EX31 ENSE00003608002 92,096,134 - INT31 Intron ENSE00003723582 - 92,079,414 - 92,096,093 ENSE00001514734 92,071,064
EX32 ENSE00002215009 92,081,061 - INT32 Intron ENSE00003654296 - 92,080,964 - 92,080,997 ENSE00003715808 92,077,603
EX33 ENSE00003734757 92,080,994 - INT33 Intron ENSE00003715808 - 92,077,580 - 92,080,968 ENSE00003713862 92,073,537
EX34 ENSE00003635015 92,071,053 - INT34 Intron ENSE00003713862 - 92,073,531 - 92,070,938 ENSE00003746704 92,071,054
EX35 ENSE00003673714 92,106,574 - INT35 Intron ENSE00003746704 - 92,071,007 - 92,106,529 ENSE00003739428 92,071,006
EX36 ENSE00003497091 92,096,134 - INT36 Intron ENSE00003646843 - 92,096,673 - 92,096,093 ENSE00003608002 92,096,135
EX37 ENSE00003538363 92,106,573 - INT37 Intron ENSE00003608002 - 92,096,092 - 92,106,529 ENSE00003499200 92,093,832
EX38 ENSE00002066587 92,081,061 - INT38 Intron ENSE00003688920 - 92,082,299 - 92,081,023 ENSE00002215009 92,081,062
EX39 ENSE00003540947 92,083,235 - INT39 Intron ENSE00002215009 - 92,080,996 - 92,083,126 ENSE00003734757 92,080,995
EX40 ENSE00002180310 92,082,466 - INT40 Intron ENSE00003654296 - 92,080,964 - 92,082,343 ENSE00003635015 92,071,054
EX41 ENSE00002023327 92,106,590 - INT41 Intron ENSE00003543114 - 92,106,528 - 92,106,529 ENSE00003499200 92,093,832
EX42 ENSE00003677024 92,093,318 - INT42 Intron ENSE00003673714 - 92,106,528 - 92,093,252 ENSE00003497091 92,096,135
EX43 ENSE00003529871 92,083,258 - INT43 Intron ENSE00003497091 - 92,096,092 - 92,083,126 ENSE00003477115 92,093,319
EX44 ENSE00003580006 92,082,466 - INT44 Intron ENSE00003543114 - 92,106,528 - 92,082,300 ENSE00003649810 92,096,138 EX45 ENSE00003529130 92,081,061 - INT45 Intron ENSE00003538363 - 92,106,528 - 92,080,965 ENSE00003646843 92,096,839
EX46 ENSE00003549058 92,071,053 - INT46 Intron ENSE00003646843 - 92,096,673 - 92,070,935 ENSE00003574977 92,088,818
EX47 ENSE00002024905 92,064,414 - INT47 Intron ENSE00003688920 - 92,082,299 - 92,062,774 ENSE00002066587 92,081,062
EX48 ENSE00002472395 92,093,850 - INT48 Intron ENSE00003477115 - 92,093,251 - 92,093,746 ENSE00003540947 92,083,236
EX49 ENSE00002437023 92,079,468 - INT49 Intron ENSE00003540947 - 92,083,125 - 92,079,415 ENSE00002180310 92,082,467
EX50 ENSE00003561582 92,071,053 - INT50 Intron ENSE00003673714 - 92,106,528 - 92,070,938 ENSE00003616151 92,083,259
EX51 ENSE00003514170 92,083,503 - INT51 Intron ENSE00002023327 - 92,106,528 - 92,083,417 ENSE00003646843 92,096,839
EX52 ENSE00003690903 92,083,235 - INT52 Intron ENSE00003649810 - 92,096,092 - 92,083,126 ENSE00003677024 92,093,319
EX53 ENSE00003550061 92,083,503 - INT53 Intron ENSE00003677024 - 92,093,251 - 92,083,417 ENSE00003628190 92,088,818
EX54 ENSE00003677507 92,088,817 - INT54 Intron ENSE00003628190 - 92,088,729 - 92,088,730 ENSE00003529871 92,083,259
EX55 ENSE00003517982 92,083,258 - INT55 Intron ENSE00003529871 - 92,083,125 - 92,083,126 ENSE00003580006 92,082,467
EX56 ENSE00003507700 92,083,235 - INT56 Intron ENSE00003580006 - 92,082,299 - 92,083,126 ENSE00003529130 92,081,062
EX57 ENSE00002498225 92,081,061 - INT57 Intron ENSE00003529130 - 92,080,964 - 92,080,953 ENSE00003549058 92,071,054
EX58 ENSE00002492203 92,081,057 - INT58 Intron ENSE00003549058 - 92,070,934 - 92,080,965 ENSE00002024905 92,064,415
EX59 ENSE00002033783 92,106,563 - INT59 Intron ENSE00003649810 - 92,096,092 - 92,106,529 ENSE00002472395 92,093,851
EX60 ENSE00002065708 92,081,061 - INT60 Intron ENSE00002472395 - 92,093,745 - 92,080,988 ENSE00003677024 92,093,319
EX61 ENSE00003563967 92,096,838 - INT61 Intron ENSE00003529130 - 92,080,964 - 92,096,674 ENSE00002437023 92,079,469
EX62 ENSE00003580759 92,096,137 - INT62 Intron ENSE00002437023 - 92,079,414 - 92,096,093 ENSE00003561582 92,071,054
EX63 ENSE00003546230 92,093,831 - INT63 Intron ENSE00003628190 - 92,088,729 - 92,093,746 ENSE00003514170 92,083,504
EX64 ENSE00002595509 92,050,646 - INT64 Intron ENSE00003514170 - 92,083,416 - 92,050,341 ENSE00003529871 92,083,259
EX65 ENSE00002606184 92,049,814 - INT65 Intron ENSE00003574977 - 92,088,729 - 92,049,603 ENSE00003690903 92,083,236
EX66 ENSE00002597953 92,044,958 - INT66 Intron ENSE00003690903 - 92,083,125 - 92,044,806 ENSE00003580006 92,082,467
EX67 ENSE00002614348 92,050,395 - INT67 Intron ENSE00003574977 - 92,088,729 - 92,050,341 ENSE00003550061 92,083,504
EX68 ENSE00002623789 92,048,089 - INT68 Intron ENSE00003550061 - 92,083,416 - 92,047,896 ENSE00003529871 92,083,259
EX69 ENSE00002617700 92,044,958 - INT69 Intron ENSE00003499200 - 92,093,745 - 92,044,775 ENSE00003677507 92,088,818
EX70 ENSE00002077210 92,106,567 - INT70 Intron ENSE00003677507 - 92,088,729 - 92,106,529 ENSE00003529871 92,083,259
EX71 ENSE00002045616 92,090,567 - INT71 Intron ENSE00003529130 - 92,080,964 - 92,090,379 ENSE00003561582 92,071,054
EX72 ENSE00002063456 92,106,591 - INT72 Intron ENSE00003608002 - 92,096,092 - 92,106,529 ENSE00003677024 92,093,319 EX73 ENSE00002054150 92,093,618 - INT73 Intron ENSE00003477115 - 92,093,251 - 92,093,552 ENSE00003517982 92,083,259
EX74 ENSE00002149638 92,093,317 - INT74 Intron ENSE00003517982 - 92,083,125 - 92,093,252 ENSE00003580006 92,082,467
EX75 ENSE00002159684 92,088,817 - INT75 Intron ENSE00003550061 - 92,083,416 - 92,088,768 ENSE00003507700 92,083,236
EX76 ENSE00002146123 92,058,787 - INT76 Intron ENSE00003507700 - 92,083,125 - 92,058,552 ENSE00003580006 92,082,467
EX77 ENSE00003529071 92,106,573 - INT77 Intron ENSE00003628190 - 92,088,729 - 92,106,529 ENSE00003507700 92,083,236
EX78 ENSE00002048204 92,093,831 - INT78 Intron ENSE00003580006 - 92,082,299 - 92,093,379 ENSE00002498225 92,081,062
EX79 ENSE00002023714 92,106,564 - INT79 Intron ENSE00002498225 - 92,080,952 - 92,106,529 ENSE00003561582 92,071,054
EX80 ENSE00002087457 92,096,838 - INT80 Intron ENSE00003688920 - 92,082,299 - 92,096,277 ENSE00002492203 92,081,058
INT81 Intron ENSE00002492203 - 92,080,964 - ENSE00003561582 92,071,054
INT82 Intron ENSE00003649810 - 92,096,092 - ENSE00003517982 92,083,259
INT83 Intron ENSE00003529871 - 92,083,125 - ENSE00003529130 92,081,062
INT84 Intron ENSE00003529871 - 92,083,125 - ENSE00003561582 92,071,054
INT85 Intron ENSE00002033783 - 92,106,528 - ENSE00003646843 92,096,839
INT86 Intron ENSE00003580006 - 92,082,299 - ENSE00002065708 92,081,062
INT87 Intron ENSE00003673714 - 92,106,528 - ENSE00003563967 92,096,839
INT88 Intron ENSE00003563967 - 92,096,673 - ENSE00003580759 92,096,138
INT89 Intron ENSE00003580759 - 92,096,092 - ENSE00003546230 92,093,832
INT90 Intron ENSE00003546230 - 92,093,745 - ENSE00003645711 92,093,319
INT91 Intron ENSE00003580759 - 92,096,092 - ENSE00003628190 92,088,818
INT92 Intron ENSE00003673714 - 92,106,528 - ENSE00003580759 92,096,138
INT93 Intron ENSE00003673714 - 92,106,528 - ENSE00003580759 92,096,138
INT94 Intron ENSE00003580759 - 92,096,092 - ENSE00003645711 92,093,319
INT95 Intron ENSE00003645711 - 92,093,251 - ENSE00003628190 92,088,818
INT96 Intron ENSE00003628190 - 92,088,729 - ENSE00003529871 92,083,259
INT97 Intron ENSE00003529871 - 92,083,125 - ENSE00003580006 92,082,467
INT98 Intron ENSE00003580006 - 92,082,299 - ENSE00002498225 92,081,062
INT99 Intron ENSE00002498225 - 92,080,952 - ENSE00003561582 92,071,054
INT100 Intron ENSE00003673714 - 92,106,528 - ENSE00003645711 92,093,319 ΙΝΤΊ01 Intron ENSE00002595509 - 92,050,340 - ENSE00002606184 92,049,815
INT102 Intron ENSE00002606184 - 92,049,602 - ENSE00002597953 92,044,959
INT103 Intron ENSE00002614348 - 92,050,340 - ENSE00002606184 92,049,815
INT104 Intron ENSE00002606184 - 92,049,602 - ENSE00002623789 92,048,090
INT105 Intron ENSE00002623789 - 92,047,895 - ENSE00002617700 92,044,959
INT106 Intron ENSE00003580759 - 92,096,092 - ENSE00003645711 92,093,319
INT107 Intron ENSE00002077210 - 92,106,528 - ENSE00003563967 92,096,839
INT108 Intron ENSE00003645711 - 92,093,251 - ENSE00002045616 92,090,568
INT109 Intron ENSE00003645711 - 92,093,251 - ENSE00003561582 92,071,054
INT110 Intron ENSE00002063456 - 92,106,528 - ENSE00003563967 92,096,839
INT111 Intron ENSE00003546230 - 92,093,745 - ENSE00002054150 92,093,619
INT112 Intron ENSE00003580759 - 92,096,092 - ENSE00003561582 92,071,054
INT113 Intron ENSE00002149638 - 92,093,251 - ENSE00002159684 92,088,818
INT114 Intron ENSE00002159684 - 92,088,767 - ENSE00002146123 92,058,788
INT115 Intron ENSE00003529071 - 92,106,528 - ENSE00003563967 92,096,839
INT116 Intron ENSE00003580759 - 92,096,092 - ENSE00002048204 92,093,832
INT117 Intron ENSE00002023714 - 92,106,528 - ENSE00002087457 92,096,839
[000374] Table 4 provides information on all of the transcripts for the ATXN3 gene based on the Ensembl database. Provided in Table 4 are the transcript ID from Ensembl and
corresponding NCBI RefSeq ID for the transcript, the translation ID from Ensembl and the corresponding NCBI RefSeq ID for the protein, the biotype of the transcript sequence as classified by Ensembl and the exons and introns in the transcript based on the information in Table 3.
Table 4. Transcript Information for ATXN3
Transcript Transcript Translation Protein Sequence Exon ID from Intron ID from ID NCBI ID NCBI Biotype Table 3 Table 3
RefSeq ID RefSeq
ID
ENST0000 NM 00499 ENSP00000 NP 00498 Protein EXl, EX2, EX3, INT1, INT2, INT3, 0558190.5 3 478320 4 coding EX4, EX5, EX6, INT4, INT5, INT6,
EX7, EX8, EX9, INT7, INT8, INT9, ΕΧΙΟ, ΕΧΠ INT10 ENST0000 - ENSP00000 - Nonsense EX2, EX3, EX18, INT2, INT51,
0359366.9 352324 mediated EX41, EX42, INT52, INT53, decay EX43, EX44, INT54, INT55,
EX45, EX46, INT56, INT57, EX47 INT58
ENST0000 NM 03066 ENSPOOOOO NP 10937 Protein EX1, EX3, EX4, INT3, INT4, INT5,
0340660.10 0 339110 6 coding EX5, EX6, EX7, INT6, INT7, INT8,
EX8, EX9, EX10, INT9, INT15, EX15 INT21
ENST0000 NM 00116 ENSPOOOOO NP 00115 Protein EX8, EX9, EX10, INT8, INT9, INT16,
0502250.5 4780 425322 8252 coding EX16, EX17, INT17, INT18,
EX18, EX19, INT19, INT20 EX20
ENST0000 NM 00112 ENSPOOOOO NP 00112 Protein EX2, EX4, EX5, INT4, INT5, INT6,
0503767.5 7696 426697 1168 coding EX6, EX7, EX8, INT7, INT8, INT9,
EX9, EX10, INT13, INT14, EX14, EX15 INT15
ENST0000 - - - Retained EX79, EX80 INT117
0454964.3 intron
ENSTOOOO - ENSPOOOOO - Nonsense EX2, EX3, EX44, INT2, INT74,
0515746.5 422073 mediated EX55, EX59, INT82, INT85, decay EX60 INT86
ENSTOOOO - ENSPOOOOO - Protein EX2, EX6, EX7, INT6, INT7, INT45,
0506466.5 435571 coding EX8, EX37, INT46, INT47
EX38
ENSTOOOO - - - Retained EX61, EX62, INT88, INT115,
0507965.5 intron EX77, EX78 INT116
ENSTOOOO - - - Processed EX61, EX62, INT88, INT89,
0504047.1 transcript EX63, EX72, ΙΝΤΠΟ, ΙΝΤΙ Π
EX73
ENSTOOOO - - - Processed EX17, EX61, INT88, INT89,
0511362.5 transcript EX62, EX63, INT90, INT107,
EX70, EX71 INT108
ENSTOOOO - - - Processed EX74, EX75, INT113, INT114
0526245.1 transcript EX76
ENSTOOOO - ENSPOOOOO - Protein EX2, EX3, EX4, INT2, INT3, INT4,
0526872.2 474067 coding EX5, EX39, INT48, INT49
EX40
ENSTOOOO - ENSPOOOOO - Protein EX2, EX3, EX4, INT2, INT3, INT4,
0532032.5 437157 coding EX5, EX6, EX7, INT5, INT6, INT7,
EX8, EX9, EX22, INT8, INT27, EX23 INT28
ENSTOOOO - ENSPOOOOO - Protein EX4, EX5, EX6, INT4, INT5, INT6,
0555381.5 451001 coding EX7, EX8, EX9, INT7, INT8, INT9,
EX10, EX24 INT41
ENSTOOOO - ENSPOOOOO - Protein EX5, EX6, EX7, INT5, INT6, INT7,
0554672.5 451417 coding EX8, EX9, EX10, INT8, INT9, INT42,
EX35, EX36 INT43
ENSTOOOO - ENSPOOOOO - Protein EX8, EX9, EX10, INT8, INT9, INT19,
0557311.5 450642 coding EX19, EX35 INT50
ENSTOOOO - ENSPOOOOO - Nonsense EX2, EX3, EX18, INT2, INT29,
0555816.5 451910 mediated EX24, EX42, INT52, INT53, decay EX43, EX45, INT54, INT57,
EX46 INT83 ENST0000 ENSPOOOOO Nonsense EX2, EX3, EX4, INT2, INT3, INT4, 0554673.5 452356 mediated EX5, EX24, INT5, INT29, decay EX43, EX44, INT55, INT56,
EX45, EX53, INT67, INT68 EX6
ENST0000 - - - Processed EX35, EX50, INT87, INT88, 0556644.5 transcript EX61, EX62 INT112
ENST0000 ENSPOOOOO Nonsense EX2, EX3, EX18, INT2, INT29, 0553488.5 452461 mediated EX24, EX42, INT52, INT53, decay EX43, EX44, INT55, INT56,
EX45, EX49, INT61, INT62, EX50, EX51 INT63, INT64
ENST0000 Processed EX17, EX18, INT17, INT54, 0554040.5 transcript EX35, EX43, INT71, INT83,
EX45, EX50, INT92, INT94 EX62
ENST0000 ENSPOOOOO Protein EX2, EX4, EX5, INT4, INT5, INT6, 0554592.5 451385 coding EX6, EX7, EX8, INT7, INT8, INT9,
EX9, EX10, INT29, INT36, EX24, EX31 INT37
ENSTOOOO ENSPOOOOO Nonsense EX2, EX18, INT29, INT36, 0556315.5 451132 mediated EX24, EX31, INT53, INT54, decay EX42, EX43, INT55, INT56,
EX44, EX45, INT57, INT72 EX46
ENSTOOOO ENSPOOOOO Nonsense EX2, EX3, EX4, INT2, INT3, INT4, 0556898.5 451769 mediated EX5, EX6, EX24, INT5, INT29, decay EX44, EX45, INT56, INT57,
EX46, EX52 INT65, INT66
ENSTOOOO ENSPOOOOO Protein EX3, EX6, EX7, INT6, INT7, INT8, 0556220.5 450641 coding EX8, EX9, EX24, INT26, INT40,
EX34 INT44
ENSTOOOO ENSPOOOOO Nonsense EX2, EX3, EX24, INT2, INT29, 0556958.5 452532 mediated EX44, EX45, INT56, INT71, decay EX50, EX55 INT74, INT82
ENSTOOOO ENSPOOOOO Nonsense EX2, EX3, EX18, INT2, INT29, 0556374.5 451399 mediated EX24, EX42, INT53, INT54, decay EX43, EX44, INT55, INT56,
EX45, EX48, INT59, INT60, EX49, EX50 INT61, INT62
ENSTOOOO ENSPOOOOO Protein EX2, EX3, EX6, INT2, INT6, INT7, 0553491.5 451996 coding EX7, EX8, EX9, INT8, INT26,
EX24, EX34 INT29, INT40
ENSTOOOO Processed EX18, EX35, INT54, INT55, 0553686.5 transcript EX43, EX44, INT78, INT79,
EX50, EX57, INT87, INT88, EX61, EX62 INT91
ENSTOOOO Processed EX17, EX18, INT17, INT54, 0554214.5 transcript EX35, EX43, INT55, INT78,
EX44, EX50, INT79, INT92, EX57, EX62 INT94
ENSTOOOO ENSPOOOOO Nonsense EX2, EX3, EX18, INT2, INT29, 0556288.5 450566 mediated EX24, EX42, INT52, INT53, decay EX44, EX45, INT56, INT71,
EX50, EX56 INT76, INT77 ENST0000 ENSPOOOOO Nonsense EX2, EX3, EX18, INT2, INT29, 0556274.5 451733 mediated EX24, EX42, INT52, INT53, decay EX44, EX50, INT76, INT77,
EX56, EX57 INT78, INT79
ENST0000 Processed EX17, EX35, INT87, INT88, 0556339.5 transcript EX50, EX61, INT89, INT90,
EX62, EX63 INT109
ENST0000 Processed EX17, EX18, INT17, INT54, 0555958.5 transcript EX35, EX43, INT55, INT78,
EX44, EX50, INT79, INT89, EX57, EX62, INT90, INT92 EX63
ENST0000 ENSPOOOOO Nonsense EX3, EX4, EX5, INT3, INT4, INT44, 0553570.5 451405 mediated EX24, EX44, INT56, INT71, decay EX45, EX50, INT73, INT74
EX55
ENST0000 ENSPOOOOO Nonsense EX2, EX3, EX4, INT2, INT3, INT4, 0554994.5 451771 mediated EX5, EX24, INT29, INT56, decay EX44, EX45, INT71, INT73,
EX50, EX55 INT74
ENSTOOOO ENSPOOOOO Nonsense EX2, EX3, EX6, INT2, INT26, 0556671.5 451693 mediated EX24, EX44, INT29, INT56, decay EX45, EX50, INT67, INT71,
EX53, EX56 INT75, INT76
ENSTOOOO ENSPOOOOO Nonsense EX2, EX3, EX4, INT2, INT3, INT29, 0554350.5 451103 mediated EX24, EX43, INT55, INT56, decay EX44, EX45, INT69, INT70,
EX50, EX54 INT71
ENSTOOOO Processed EX17, EX18, INT17, INT54, 0553287.5 transcript EX35, EX43, INT55, INT78,
EX44, EX50, INT79, INT100 EX57
ENSTOOOO Processed EX17, EX18, INT17, INT54, 0553309.5 transcript EX35, EX43, INT55, INT78,
EX44, EX50, INT79, INT87, EX57, EX61, INT88, INT89, EX62, EX63 INT90
ENSTOOOO Processed EX17, EX18, INT17, INT54, 0554491.5 transcript EX35, EX43, INT55, INT56,
EX44, EX45, INT61, INT62, EX49, EX50, INT87, INT88, EX61, EX62, INT89, INT90 EX63
ENSTOOOO ENSPOOOOO Nonsense EX2, EX6, EX7, INT6, INT7, INT45, 0557030.5 452139 mediated EX8, EX37, INT46, INT80, decay EX50, EX58 INT81
ENSTOOOO Processed EX18, EX35, INT54, INT55, 0553498.5 transcript EX43, EX44, INT56, INT61,
EX45, EX49, INT62, INT87, EX50, EX61, INT88, INT91 EX62
ENSTOOOO ENSPOOOOO Nonsense EX2, EX3, EX18, INT2, INT29, 0556082.5 450492 mediated EX24, EX42, INT52, INT53, decay EX43, EX50 INT54, INT84 ENST0000 - - - Processed EX65, EX67, INT103, INT104, 0564606.5 transcript EX68, EX69 INT105
ENST0000 Processed EX64, EX65, INT101, INT102 0568290.1 transcript EX66
ENST0000 NM 00112 ENSP00000 NP 00112 Protein EX2, EX3, EX6, INT2, INT6, INT7, 0393287.9 7697, 376965 1169, coding EX7, EX8, EX9, INT8, INT9, INT12,
NM 00116 NP 00115 EX10, EX12, INT23, INT26 4779 8251 EX13
ENST0000 NM 00116 ENSP00000 NP 00115 Protein EX4, EX5, EX6, INT4, INT5, INT6, 0429774.6 4779, 389376 8251, coding EX7, EX8, EX9, INT7, INT8, INT9,
NM 00116 NP 00115 EX10, EX12, INT11, INT12 4781 8253 EX13
ENST0000 ENSP00000 Protein EX2, EX3, EX4, INT2, INT3, INT4, 0545170.5 445618 coding EX5, EX6, EX7, INT5, INT6, INT7,
EX8, EX9, EX10, INT8, INT12, EX12, EX13, INT23, INT24, EX21 INT25
ENSTOOOO ENSP00000 Protein EX2, EX3, EX4, INT2, INT3, INT4, 0617719.4 481998 coding EX5, EX6, EX7, INT5, INT6, INT7,
EX8, EX9, EX24, INT8, INT29, EX25, EX26 INT30, INT31
ENSTOOOO ENSP00000 Protein EX2, EX3, EX4, INT2, INT3, INT4, 0620536.4 484016 coding EX5, EX6, EX7, INT5, INT6, INT7,
EX8, EX9, EX12, INT8, INT23, EX27, EX28, INT32, INT33, EX29, EX30 INT34, INT35
ENSTOOOO ENSP00000 Protein EX2, EX3, EX4, INT2, INT3, INT4, 0621269.4 481390 coding EX5, EX6, EX7, INT5, INT6, INT7,
EX8, EX12, INT23, INT38, EX32, EX33 INT39
[000375] ATXN3 has 2939 S Ps and the NCBI rs number and/or UniProt VAR number for this ATXN3 gene are rsl0129742, rsl0129909, rsl0131594, rsl0131670, rsl0132620, rsl0133404, rsl0135150, rsl0135173, rsl0135377, rsl0135397, rsl0135695, rsl0137541, rsl0137707, rsl0137727, rsl0138296, rsl0138566, rsl0140469, rsl0140481, rsl0140609, rsl0140666, rsl0140720, rsl0141916, rsl0142041, rsl0143213, rsl0143310, rsl0143456, rsl0143581, rsl0145053, rsl0145950, rsl0146087, rsl0146249, rsl0146519, rsl0148817, rsl0150068, rsl0150739, rsl0150904, rsl0151135, rsl0151170, rsl0151930, rsl0151932, rsl0151945, rsl0220696, rsl0467748, rsl0467856, rsl0467857, rsl0467858, rsl0467859, rsl047795, rsl048755 4, rsl055996, rsl0606586, rsl0608041, rsl0672184, rsl072957, rsl072958, rsl0873415, rsl 107115, rsl 11249502, rsl 11272463, rsl 11357714, rsl 11393662, rsl 11459590, rsl 11461096, rsl 11515897, rsl 11579518, rsl 1160038, rsl 1160039, rsl 11669194, rsl 11704070, rsl 11735934, rsl 11754914, rsl 11795258, rsl 118322, rsl 11901666, rsl 11923598, rsl 11950372, rsl 11954089, rsl 11995259, rsl 12033378, rsl 12044110, rsl 12083702, rsl 12091003, rsl 12097797, rsl 12122875, rsl 12149349, rsl 12165995, rsl 12175516, rsl l2188765, rsl l2193853, rsl l2197978, rsl l2205087, rsl l2311135, rsl l2537858, rsl 12548913, rsl 12577035, rsl 12582313, rsl 12586446, rsl 12656324, rsl 12733683, rsl l2737075, rsl l2851203, rsl 13070671, rsl l308589, rsl l3143831, rsl l3221913,
rsl 13255226, rsl 13362282, rsl 1340208, rsl 1340209, rsl 134377, rsl 134378, rsl 13482122, rsl l3534409, rsl l3572439, rsl 13595581, rsl l367316, rsl l3728897, rsl l3752259,
rsl 13777529, rsl 13799332, rsl 13815687, rsl 1388890, rsl 139151 19, rsl 13953465,
rsl 14127501, rsl 1416778, rsl 14495966, rsl 1451253, rsl 14757328, rsl 14977442, rsl 15150898, rsl 15345965, rsl 15790215, rsl 16179168, rsl 1621770, rsl 1628764, rsl 16338117, rsl 16410176, rsl 16420499, rsl 16491853, rsl 16598428, rsl 16816718, rsl 16899751, rsl 16967340, rsl 16994678, rsl 17235977, rsl 17509573, rsl 17601173, rsl 18030094, rsl 1844802, rsl 1846742, rsl 1849632, rsl 1849927, rsl 1849958, rsl 1850101, rsl2147367, rsl2147767, rsl2431688, rsl2433948, rsl2434076, rsl2435687, rsl2436791, rsl2436814, rsl2586471, rsl2586535, rsl2588287, rsl2590497, rsl2878941, rsl2879062, rsl2879090, rsl2889180, rsl2895357, rsl2896583, rsl2896588, rsl2896589, rsl3379287, rsl38030090, rsl38047286, rsl38074617, rsl38111554, rsl38165116, rsl38243333, rsl38320031, rsl38395021, rsl38404351, rsl38429167, rsl38526455, rsl38626759, rsl38631973, rsl38669304, rsl38766948, rsl38868478, rsl38914909, rsl38986677, rsl39050721, rsl391 12984, rsl39131832, rsl39199676, rsl39201400, rsl39337731, rsl39366856, rsl39388399, rsl39505777, rsl39549069, rsl39577236, rsl39703250, rsl40129483, rsl40210106, rsl40285410, rsl40358314, rsl40477113, rsl40562271, rsl40568543, rsl40741824, rsl40763936, rsl40811598, rsl40887946, rsl40908230, rsl40916143, rsl40950495, rsl41210867, rsl41319170, rsl41580377, rsl41672872, rsl41713202, rsl41724936, rsl41777702, rsl41847020, rsl41860581, rsl41863730, rsl41931459, rsl41934300, rsl41991823, rsl41993435, rsl42090748, rsl42148800, rsl42189632, rsl42358420, rsl42371486, rsl42537405, rsl42538026, rsl42622052, rsl42718363, rsl42874189, rsl42951329, rsl42986250, rsl43130472, rsl43159368, rsl43167763, rsl43388799, rsl43402326, rsl43513154, rsl43561346, rsl43617494, rsl43730185, rsl43908400, rsl43923548, rs144045303, rsl44152152, rsl44194306, rsl44235163, rsl44237516, rsl44263831, rsl44310780, rsl44372654, rsl44506566, rsl44550842, rsl44674950, rsl44694865, rs144799309, rsl44833998, rsl44948983, rsl44972902, rsl45019811, rsl45205040, rsl45210459, rsl45219354, rsl45423020, rsl45498216, rsl45536391, rsl45601160, rsl45614854, rsl45755242, rsl45772139, rsl45887922, rsl46144466, rsl46163190, rsl46242536, rsl46283674, rsl46356399, rsl46404413, rsl46425785, rsl46481848,
rsl46634447, rsl46668789, rsl46706522, rsl46832756, rsl47033585, rsl47058331,
rsl47282222, rsl47833264, rsl47932464, rsl47968987, rsl48055766, rsl48181893,
rsl48266518, rsl48366775, rsl48498078, rsl48529923, rsl48585080, rsl48706413,
rsl48796712, rsl48849308, rsl48889666, rsl48949170, rsl48968790, rsl49006887,
rsl49131253, rsl49201025, rsl49310718, rsl49310854, rsl49344198, rsl49379953,
rsl49589054, rsl49656376, rsl49749102, rsl49784856, rsl49821097, rsl49978538,
rsl49978731, rsl50031811, rsl 50065649, rsl50082297, rsl 50278508, rsl50321294,
rsl 50394957, rsl50510850, rsl 50577313, rsl50736485, rsl50738718, rsl50754345,
rsl50823108, rsl51002328, rsl51053179, rsl51107245, rsl51263555, rsl51320956,
rsl6999141, rsl7094810, rsl7127901, rsl7127913, rsl7127915, rsl7127928, rsl7127929, rsl7127939, rsl7127942, rsl7127943, rsl7127945, rsl7734627, rsl7734780, rsl7783552, rsl7807815, rsl7847267, rsl7847274, rsl7847278, rsl7847279, rsl7853195, rsl804683, rsl 80750733, rsl80880452, rsl 80883283, rsl80896627, rsl 80913839, rsl81356152, rsl 81438171, rsl81529854, rsl 81584149, rsl81630218, rsl 81645596, rsl81646028, rsl 81674698, rsl81738620, rsl 81752420, rsl81999063, rsl82003749, rsl82242370, rsl82252142, rsl82317909, rsl82375232, rsl82464102, rsl82487175, rsl82522824, rsl82525095, rsl82587249, rsl82654422, rsl82761342, rsl82777546, rsl82827058, rsl82833108, rsl82945697, rsl82949932, rsl82971628, rsl83056610, rsl83076827, rsl83209452, rsl83215055, rsl834101 17, rsl83516481, rsl83605224, rsl83613344, rsl 83615675, rsl83754939, rsl 83875010, rsl83882162, rsl 83892533, rsl84013158, rsl 84027217, rsl84319601, rsl 84438102, rsl84511990, rsl 84557079, rsl84614940, rsl 84656238, rsl84715319, rsl85153209, rsl85201254, rsl85216841, rsl85271319, rsl85408452, rsl85425916, rsl85548077, rsl86086372, rsl86088509, rsl86104713, rsl86208016, rsl86235175, rsl86244206, rsl86258986, rsl86327684, rsl86345225, rsl86350061, rsl86352724, rsl86369272, rsl86378243, rsl86458642, rsl86461294, rsl86921280, rsl86950202, rsl86962399, rsl87053228, rsl87088090, rsl87133030, rsl 87186734, rsl87390001, rsl 87513377, rsl87682177, rsl 87776046, rsl87901616, rsl 87968063, rsl87973435, rsl 88041208, rsl88165828, rsl 88332533, rsl884774, rsl88626397, rsl 88681362, rsl88685550, rsl88739788, rsl88761840, rsl88811677, rsl88823821, rsl88845799, rsl88893532, rsl88901502, rsl88907529, rsl88945804, rsl89111825, rsl89253199, rsl89397136, rsl89405140, rsl89531839, rsl89650643, rsl89672882, rsl89707249, rsl89713300, rsl89785397, rsl89953842, rsl90049965, rsl90071316, rsl90204338, rsl90227164, rsl90326858, rsl90337187, rsl90465361, rsl90629084, rsl90730589, rsl90824927, rsl91004545, rsl91013089, rsl91145357, rsl91147344, rsl91155540, rsl91168297, rsl91170218, rsl91189424, rsl91291161, rsl91402886, rsl91433400, rsl91436857, rsl91439468, rsl91679404, rsl91893556, rsl91900676, rsl91933456, rsl91951136, rsl91962247, rsl92112040, rsl92116249, rsl92210101, rsl92281900, rsl92345523, rsl92365513, rsl92385527, rsl92441611, rsl92630562, rsl92665869, rsl92725335, rsl92766993, rsl92852260, rsl92985656, rsl93052814, rsl93135154, rsl93285351, rsl956415, rsl956416, rsl956417, rsl99503630, rsl99512590, rsl99526025, rsl99528136, rsl99690211, rsl99790369, rsl997917, rsl997918, rsl997919, rsl997920, rsl99813282, rsl99866952, rsl99876727, rsl99894637, rsl99942851, rs200105341, rs200118135, rs200134701, rs200230139, rs200238931, rs200242111, rs200304679, rs200332234, rs200356462, rs200368717, rs2003721, rs2003722, rs200386468, rs200388434, rs200444985, rs200462149, rs200483057, rs200483759, rs200578869, rs200580896, rs200581319, rs200584159, rs200597744, rs2006047, rs200747001, rs200757726, rs200799469, rs200836653, rs200872467, rs200925789, rs201004528, rs201012097, rs201025371, rs201027114, rs201062643, rs201083534, rs201197090, rs201241311, rs201361883, rs201413769, rs201457134, rs201475233, rs201553525, rs201691672, rs201693494, rs201743985, rs201815350, rs201931150, rs201936460, rs201942080, rs201992162, rs202119840, rs202192680, rs202247493, rs2145127, rs2145128, rs2145129, rs2268007, rs2295175, rs2295176, rs2295177, rs2402090, rs2402103, rs2402104, rs2402105, rs2402106, rs2402107, rs2402108, rs2402109, rs2402110, rs28409802, rs28434101, rs28671628, rs28696879, rs28841105, rs2896195, rs2896196, rs3031725, rs3092822, rs3178662, rs3204346, rs33967699, rs33981553, rs34072011, rs34102054, rs34136360, rs34148613, rs34158455, rs34207682, rs34231755, rs34338098, rs34351382, rs34366039, rs34431920, rs34561748, rs34611489, rs34755730, rs34788226, rs34791653, rs34815272, rs34830555, rs34928160, rs34977286, rs35014837, rs35041243, rs35074352, rs35079962, rs35141835, rs35333165, rs35361481, rs35515547, rs35517863, rs35518817, rs35584810, rs35694255, rs35865969, rs35956988, rs36058127, rs36111025, rs367603239, rs367664867, rs367671984, rs367769116, rs367890627, rs367993031, rs368012807, rs368036940, rs368058740, rs368119597, rs368260154, rs368313233, rs368412477, rs368415165, rs368518223, rs368545828, rs368734521, rs368741975, rs368753991, rs368770562, rs368913861, rs368922228, rs368968720, rs368997837, rs369016842, rs369226337, rs369227029, rs369366170, rs369366675, rs369370330, rs369376709, rs369470265, rs369491189, rs369512418, rs369561103, rs369582326, rs369630957, rs369643233, rs369674541, rs369694569, rs369732127, rs369764366, rs369764546, rs369985856, rs369995156, rs370049538, rs370136800, rs370153151, rs370500473, rs370523862, rs370538321, rs370588550, rs370605537, rs370669483, rs370731812, rs370743817, rs370836455, rs370880141, rs370941249, rs370997724, rs371007546, rs371017302, rs371037244, rs371090121, rs371139662, rs371386339, rs371441035, rs371442064, rs371513086, rs371606868, rs371618456, rs371630976, rs371760164, rs371762976, rs371776548, rs371787135, rs371798010, rs371830105, rs371935073, rs372107686, rs372110002, rs372183020, rs372246575, rs372281452, rs372405510, rs372538978, rs372556545, rs372649153, rs372651076, rs372673909, rs372732054, rs372859033, rs372907618, rs372963663, rs373046214, rs373051426, rs373062241, rs373205243, rs373284826, rs373354514, rs373378578, rs373384259, rs373595567, rs373597908, rs373659686, rs373706702, rs373813370, rs373856745, rs373920734, rs373925635, rs374165861, rs374202884, rs374260891, rs374271659, rs374276626, rs374331774, rs374353802, rs374466872, rs374513078, rs374576695, rs374579221, rs374598990, rs374607093, rs374620100, rs374646934, rs374699412, rs374782928, rs374862126, rs374939668, rs374952426, rs374968114, rs374976997, rs375141956, rs375162806, rs375228860, rs375246638, rs375305746, rs375315995, rs375420935, rs375425370, rs375468007, rs375505876, rs375534522, rs375679519, rs375725623, rs375748430, rs375778279, rs375844803, rs375874329, rs376007653, rs376017626, rs376064779, rs376316878, rs376480119, rs376513911, rs376552690, rs376552813, rs376571187, rs376647564, rs376741760, rs376789129, rs376795114, rs376865350, rs376966269, rs377031212, rs377182886, rs377198910, rs377240305, rs377272847, rs377358074, rs377369716, rs377386978, rs377574592, rs377603133, rs377639670, rs377664561, rs377666680, rs377668917, rs377687859, rs3814834, rs3819779, rs3819780, rs386419521, rs386779982, rs386779983, rs386779986, rs397853158, rs397853212, rs397942802, rs397970086, rs398026196, rs398118522, rs4410011, rs4420459, rs4900095, rs4904833, rs4904834, rs4904835, rs4904836, rs527414821, rs527415987, rs527538369, rs527605104, rs527693624, rs527704689, rs527734891, rs527776399, rs527778752, rs527817146, rs527844598, rs527986629, rs528024685, rs528076299, rs528112066, rs528123223, rs528256797, rs528428826, rs528495057, rs528534459, rs528611560, rs528644988, rs528765879, rs528829474, rs528849723, rs528998151, rs529025874, rs529036679, rs529052030, rs529089703, rs529143309, rs529192554, rs529233605, rs529273872, rs529290085, rs529386181, rs529420712, rs529435200, rs529456177, rs529468483, rs529666926, rs529732410, rs529742601, rs529818301, rs529883696, rs530038772, rs530123411, rs530173072, rs530173230, rs530206594, rs530268223, rs530321101, rs530328167, rs530494230, rs530499241, rs530695347, rs530733613, rs530768711, rs530902537, rs530905428, rs530917270, rs530984812, rs531024510, rs531128594, rs531158651, rs531271792, rs531342378, rs531344370, rs531512266, rs531536759, rs531538876, rs531619809, rs531627273, rs531660116, rs531705648, rs531719156, rs531771761, rs531937444, rs531956768, rs531985176, rs532080357, rs532122634, rs532129302, rs532225438, rs532236578, rs532290325, rs532292373, rs532328336, rs532467898, rs532483134, rs532498095, rs532547403, rs532679042, rs532737742, rs532765285, rs532771040, rs532825289, rs532859750, rs532864068, rs532926609, rs533184429, rs533204251, rs533229850, rs533250394, rs533264801, rs533268889, rs533283411, rs533286136, rs533334971, rs533360926, rs533381185, rs533393923, rs533409972, rs533550346, rs533551674, rs533561522, rs533620624, rs533644996, rs533719629, rs533754030, rs533881446, rs533910617, rs533969980, rs534154599, rs534180475, rs534198061, rs534347269, rs534400789, rs534404823, rs534442386, rs534476352, rs534592736, rs534619015, rs534632654, rs534680546, rs534755253, rs534884019, rs534884559, rs534904175, rs534939832, rs535089903, rs535108713, rs535188218, rs535200709, rs535267443, rs535274801, rs535376586, rs535404667, rs535407026, rs535550478, rs535604824, rs535635553, rs535658858, rs535739678, rs535819907, rs535894743, rs535903212, rs535903273, rs535967073, rs536017064, rs536052200, rs536093584, rs536215415, rs536251415, rs536458345, rs536574503, rs536830767, rs536855091, rs537095615, rs537236516, rs537273938, rs537336114, rs537397429, rs537416040, rs537540803, rs537578893, rs537593516, rs537726421, rs537789812, rs537815029, rs537909855, rs537987995, rs538033752, rs538059127, rs538172199, rs538254237, rs538312555, rs538330559, rs538430022, rs538473764, rs538477284, rs538529483, rs538536526, rs538564457, rs538593472, rs538606816, rs538613356, rs538616103, rs538654106, rs538660492, rs538989117, rs539159813, rs539206751, rs539351859, rs539410331, rs539431582, rs539447346, rs539615462, rs539649672, rs539653266, rs539654605, rs539661232, rs539718709, rs539757716, rs539823167, rs539873020, rs539972120, rs539996979, rs540170798, rs540263466, rs540319960, rs540408025, rs540451699, rs540582368, rs540592864, rs540605576, rs540605599, rs540635345, rs540643404, rs540794174, rs540810112, rs540926560, rs540944019, rs540961318, rs540961626, rs540969159, rs541018001, rs541053091, rs541076192, rs541126375, rs541140524, rs541164672, rs541174896, rs541182858, rs541222335, rs541261309, rs541278875, rs541315490, rs541422032, rs541614407, rs541628028, rs541660577, rs541758473, rs541847763, rs541867968, rs541880574, rs541911807, rs541987773, rs542020280, rs542024827, rs542143798, rs542184279, rs542204082, rs542354616, rs542373809, rs542401270, rs542406498, rs542469912, rs542487157, rs542580529, rs542631421, rs542700748, rs542783414, rs542821056, rs542821912, rs542858618, rs542893885, rs542986852, rs542990076, rs543022880, rs543180973, rs543255653, rs543269362, rs543304516, rs543354203, rs543371608, rs543397236, rs543416541, rs543607269, rs543660755, rs543752947, rs543786776, rs543859927, rs543886934, rs544002893, rs544031301, rs544170451, rs544179293, rs544429100, rs544454211, rs544573012, rs544588049, rs544626652, rs544630122, rs544768913, rs544864800, rs544869630, rs544912832, rs544947224, rs545046382, rs545095173, rs545100447, rs545162568, rs545195105, rs545281918, rs545350906, rs545355002, rs545489675, rs545630452, rs545673644, rs545793946, rs545809463, rs545920908, rs545929028, rs545931595, rs545973052, rs546009964, rs546091180, rs546219427, rs546260267, rs546330910, rs546344527, rs546394320, rs546420283, rs546433513, rs546491366, rs546516438, rs546591975, rs546604466, rs546611585, rs546648423, rs546779978, rs546857485, rs546870758, rs546941052, rs547040313, rs547055277, rs547101726, rs547111329, rs547138475, rs547199233, rs547237619, rs547311096, rs547312155, rs547345432, rs547354686, rs547399238, rs547413151, rs547418279, rs547463247, rs547536133, rs547797764, rs547809200, rs547921194, rs547971490, rs547991708, rs548022637, rs548060993, rs548167341, rs548194496, rs548229374, rs548231381, rs548232356, rs548242072, rs548266802, rs548295561, rs548356150, rs548374312, rs548380834, rs548411078, rs548488269, rs548504120, rs548554248, rs548589805, rs548628650, rs548763388, rs548800762, rs548850926, rs548859340, rs548985761, rs549024988, rs549106315, rs549296263, rs549433428, rs549446149, rs549501128, rs549509990, rs549561009, rs549598045, rs549658135, rs549703643, rs549720329, rs549799213, rs549823402, rs549825768, rs549835945, rs549867277, rs549894981, rs549919052, rs549931896, rs550116664, rs550133771, rs550205548, rs550222516, rs550304745, rs550375382, rs550396213, rs550403883, rs550405473, rs550449337, rs550488125, rs550522547, rs550539467, rs550573299, rs550592525, rs550627847, rs550657682, rs550712214, rs550757499, rs550759966, rs550851689, rs550997833, rs551076709, rs551225394, rs551277425, rs551289688, rs551303066, rs551359376,
rs551522771, rs551666415, rs551819678, rs551829091, rs551936571, rs552042048,
rs5521 15942, rs552206603, rs552284668, rs552321597, rs552474541, rs552474831,
rs552633402, rs552688213, rs552740772, rs552780970, rs552838714, rs552892748,
rs552944466, rs552947804, rs553030192, rs553067160, rs553068814, rs553145415,
rs553176454, rs553180414, rs553217270, rs553243412, rs553303069, rs553332020,
rs553462759, rs553632954, rs553702875, rs553772232, rs553786504, rs553808854, rs553846552, rs553969334, rs554007647, rs554124396, rs554127288, rs554174637, rs554183615, rs554236101, rs554238799, rs554272901, rs554327114, rs554334279, rs554358870, rs554404571, rs554520186, rs554590855, rs554621837, rs554711538, rs554715160, rs554732515, rs554791387, rs554796289, rs554809861, rs554810577, rs554815626, rs554895432, rs554901119, rs554956238, rs554966272, rs554972799, rs555060450, rs555109144, rs555120312, rs555141255, rs555144317, rs555152381, rs55525451 1, rs555353531, rs555385785, rs555401909, rs555522753, rs555535234, rs555548105, rs555559894, rs555565674, rs555598326, rs555614052, rs555621739, rs555631426, rs555644215, rs555646947, rs555688232, rs555745598, rs555785134, rs555841221, rs555869209, rs555894423, rs555906302, rs555974894, rs556002813, rs556234359, rs556241549, rs556355638, rs55636647, rs556422695, rs55644237, rs55656618, rs556566246, rs55656743, rs556596700, rs556690788, rs556693607, rs556725165,
rs556760089, rs55676368, rs55679384, rs55682138, rs556847412, rs556904146, rs55691521, rs55692120, rs55693371, rs55695540, rs55696543, rs556978833, rs556989809, rs55702425, rs55703926, rs55705173, rs557062918, rs557143758, rs557155400, rs557157505, rs557178623, rs55719539, rs557199259, rs55724500, rs557300501, rs55730538, rs55735641, rs557368297, rs55737228, rs557375373, rs557377431, rs55743437, rs557439193, rs557446490, rs55750042, rs557614529, rs557763892, rs557771242, rs557783116, rs55779410, rs557794586, rs55783917, rs557932776, rs55794018, rs55796924, rs55801726, rs558083582, rs55811958, rs558124846, rs558170654, rs558211438, rs558289000, rs55829932, rs55830230, rs558324040, rs55834444, rs558382743, rs558438914, rs55846004, rs55849366, rs558498205, rs55851933, rs55852837, rs55855317, rs558576536, rs55860112, rs55860701, rs558638749, rs558707811, rs55875032, rs55881247, rs558850119, rs55888350, rs558890121, rs558921046, rs55893083, rs55904144, rs55908884, rs55912493, rs55913296, rs55913909, rs55919513, rs55921125, rs55929165, rs559329531, rs559333737, rs55934123, rs55937802, rs559428176, rs559431016, rs55948797, rs559491213, rs55950208, rs55951860, rs559559866, rs55957817, rs559586131, rs55961283, rs55961936, rs559621772, rs559624762, rs559636074, rs55966267, rs55970447, rs55976835, rs559798134, rs559808979, rs55980965, rs55983212, rs559839103, rs559924763, rs55995323, rs559976759, rs559997340, rs56005080, rs560104176, rs560142515, rs560161051, rs56024302, rs56027550, rs56033639, rs560384740, rs56039439, rs560406003, rs560421487, rs560458940, rs56052998, rs56057832, rs56057930, rs560585597, rs560592057, rs560619291, rs560639167, rs56065184, rs560663525, rs56067028, rs560676740, rs560745737, rs560780607, rs560787454, rs560791113, rs56080207, rs560807324, rs560829749, rs56086745, rs56089900, rs56090346, rs560948043, rs560997508, rs56100339, rs56101118, rs561025762, rs56108392, rs56116379, rs561 171480, rs56120704, rs56124506, rs561248414, rs56126302, rs561279560, rs56139005, rs56141463, rs561426690, rs56143352, rs56143414, rs561461022, rs56148011, rs561534277, rs561595983, rs561634929, rs56168906, rs561747168, rs561765335, rs56177134, rs56177322, rs561826679, rs561910395, rs561944356, rs56196346, rs561972494, rs561978307,
rs561982469, rs56199148, rs56199729, rs562006347, rs562049944, rs56208018, rs56212193, rs562165101, rs562182115, rs562187688, rs562189056, rs56220647, rs562218818,
rs562225763, rs56223923, rs5622561 1 1, rs56226118, rs56232098, rs562337291, rs562339414, rs562374393, rs562397414, rs56241 1792, rs562424472, rs562449919, rs562463618, rs562475945, rs56249958, rs56261 1680, rs56262081, rs562671116, rs56268847, rs56273178, rs56277345, rs562774919, rs562787524, rs56280640, rs562809949, rs562932661, rs562933608, rs562941621, rs562971378, rs56297156, rs563015172, rs563065990, rs563090828, rs56311847, rs563148297, rs56315305, rs563156654, rs56315671 1, rs563192094, rs56323605, rs56324055, rs563308766, rs56331048, rs56332270, rs56332503, rs56332990, rs563330768, rs56343705, rs563544827, rs563546390, rs56354799, rs56360110, rs563603966, rs563655446, rs56365709, rs563811378, rs56382532, rs56384416, rs563848209, rs56396823, rs56397248, rs563999311, rs56402719, rs56402956, rs564063187, rs56406349, rs564165980, rs564203964, rs564315315, rs564349535, rs564349806, rs564386402, rs564438404, rs564472295, rs564566793, rs564572743, rs564572941, rs564578175, rs564614805, rs564654862, rs564744388, rs564774169, rs564812406, rs564884511, rs564963926, rs565014021, rs565084148, rs565152112, rs565230800, rs565330272, rs565352190, rs565413341, rs565452568, rs565479771, rs565499310, rs565519868, rs565549046, rs565559987, rs565584720, rs565714983, rs565734430, rs565746295, rs565746446, rs565772024, rs565783047, rs565803496, rs565895221, rs565927216, rs565951567, rs565999797, rs56601 1476, rs566029191, rs566035233, rs566094960, rs566099896, rs566394811, rs566451652, rs566465881, rs566522339, rs566690448, rs566768477, rs566818283, rs566899059, rs566997652, rs567003595, rs567028605, rs567173949, rs567182665, rs567191556, rs567225714, rs567245946, rs56728821, rs567311887, rs567328244, rs567411741, rs567638182, rs567673127, rs567696257, rs567708278, rs567753088, rs567800167, rs567881420, rs568021106, rs568117040, rs568192565, rs568253470, rs568285429, rs568291443, rs568319558, rs568327663, rs568358751, rs568359565, rs568383671, rs568439304, rs568535142, rs568567371, rs568608527, rs568911932, rs568989459, rs568999116, rs569029586, rs569075167, rs569115272, rs569174897, rs569176686, rs569223521, rs569229752, rs569366715, rs569392621, rs569413537, rs569576033, rs569585120, rs569587509, rs569604127, rs569631238, rs569664546, rs569692624, rs569832524, rs569904991, rs569912322, rs569976612, rs570074746, rs570329309, rs570414402, rs570576217, rs570617215, rs570669309, rs570686151, rs570696090, rs570739350, rs570818418, rs570857774, rs57086831, rs570898348, rs570909350, rs570939083, rs570974361, rs571083528, rs571147592, rs571171694, rs571196964, rs571237166, rs571253140, rs571382527, rs571519299, rs571628742, rs571674598, rs571717364, rs571755457, rs571789363, rs571822056, rs571895893, rs571948743, rs571968161, rs571996253, rs572044578, rs572103953, rs572136631, rs572155598, rs572161146, rs572191382, rs572454881, rs572624672, rs572629561, rs572650747, rs572694551, rs572839514, rs572859250, rs572994234, rs573022747, rs573067835, rs573080540, rs573110705, rs573186328, rs573253829, rs573318124, rs573612370, rs573630167, rs573665148, rs573712158, rs573847986, rs574123299, rs574166677, rs574252715, rs574280493, rs574329328, rs574338625, rs574368048, rs574458167, rs574496613, rs574567962, rs574779182, rs574815764, rs574834309, rs574877194, rs574878588, rs574887239, rs574973061, rs575061336, rs575135366, rs575218366, rs575386049, rs575503902, rs575509086, rs575512398, rs575592315, rs575681049, rs575861206, rs57593784, rs576015686, rs576056174, rs576079150, rs576107821, rs576137883, rs576244669, rs576329374, rs576561754, rs576579198, rs576601148, rs576627670, rs576638721, rs576640996, rs576667459, rs576674066, rs576705676, rs576723967, rs576762024, rs576787480, rs576855160, rs576952294, rs577003269, rs577038556, rs577040365, rs577084043, rs577120028, rs577124114, rs577161760, rs577208768, rs577237264, rs577246059, rs577266060, rs577351437, rs577401229, rs577557193, rs577587890, rs577619606, rs577644529, rs577684465, rs577825283, rs577858011, rs577866202, rs577874229, rs577976216, rs578071549, rs578133639, rs578194188, rs578231955, rs578238563, rs5810570, rs5810571, rs5810572, rs5810573, rs5810574, rs5810575, rs58398762, rs58982263, rs59217884, rs59299778, rs59864685, rs60428491, rs60560689, rs60616585, rs61461391, rs61676989, rs61988394, rs61988396, rs61988397, rs61988398, rs6575227, rs6575228, rs66941473, rs67089639, rs709930, rs71123316, rs71301947, rs71301948, rs71301949, rs71412344, rs7142326, rs7142850, rs7143616, rs7143765, rs7143907, rs7144492, rs71461957, rs7146985, rs7148184, rs7148258, rs7149105, rs7149601, rs7149662, rs7149775, rs7150333, rs7150414, rs7150654, rs7151555, rs7152876, rs7153193, rs7153366, rs7153374, rs7153603, rs7153615, rs7153696, rs7154147, rs7154396, rs7154758, rs7154952, rs7155310, rs7155635, rs7155871, rs7157769, rs7158238, rs7158733, rs71952634, rs72471507, rs73323741, rs73323771, rs73325756, rs73325760, rs73327719, rs74071833, rs74071835, rs74071846, rs74071847, rs74071849, rs74071869, rs74071876, rs74071878, rs74071880, rs74244591, rs74357844, rs745364516, rs745392740, rs745440781, rs745474335, rs745477026, rs745521332, rs745527516, rs745625169, rs745628747, rs745655000, rs745718634, rs745719415, rs745821507, rs745853952, rs745862718, rs745890873, rs74589751, rs745959046, rs746020592, rs746062546, rs746101828, rs746109327, rs746133585, rs746142708, rs74619884, rs746205070, rs746216957, rs746232891, rs746294974, rs746454122, rs746532544, rs746622903, rs746644142, rs746678958, rs746711349, rs746720725, rs746773032, rs746789483, rs74679283, rs746867371, rs746995256, rs747056916, rs747075248, rs747089043, rs747156311, rs747262272, rs74731246, rs747360445, rs747415001, rs747424729, rs747462872, rs747490844, rs747520532, rs747549538, rs747557328, rs747578825, rs747586178, rs747666819, rs747666820, rs747720294, rs747754080, rs747774933, rs747804276, rs747811371, rs747857148, rs747923828, rs747933521, rs747941607, rs748016235, rs748042622, rs748079715, rs748088552, rs748105188, rs748168008, rs748191546, rs748209666, rs748281008, rs748324359, rs748394394, rs748421146, rs748481683, rs748502160, rs748571539, rs748604134, rs74873370, rs748855843, rs748857493, rs748879218, rs748959673, rs748990614, rs748996136, rs749077703, rs749214501, rs749214528, rs749230434, rs7492652, rs7493524, rs749363861, rs7493647, rs749388478, rs749523391, rs749561634, rs749575271, rs749645729, rs749731921, rs749794712, rs749904761, rs749906950, rs749952916, rs75002598, rs750040795, rs750048782, rs750086718, rs750174606, rs750298097, rs750301693, rs750309656, rs750329097, rs750375139, rs750414353, rs750449708, rs750538982, rs750559945, rs750579021, rs750631823, rs750672731, rs750717644, rs750733064, rs750747570, rs750759057, rs750833339, rs750845168, rs750893431, rs750938282, rs750955372, rs750977319, rs751059462 rs751064156, rs751097712, rs751145678, rs751182628, rs751203850, rs751303682 rs751329134, rs751433921, rs751439402, rs751466445, rs751499818, rs751501457 rs751510163, rs751558201, rs751647808, rs751648794, rs751649756, rs751746839 rs751821844, rs751841317, rs751874751, rs75188275, rs751908477, rs752062085 rs752111992, rs752168522, rs752183057, rs752316242, rs752379583, rs752449425 rs752468952, rs752481922, rs752526340, rs752537367, rs752540499, rs752547907 rs752589357, rs752600786, rs75271164, rs752749801, rs752783244, rs752841021 rs75293788, rs752940647, rs752981470, rs753004854, rs753009580, rs75308098, rs753344410, rs753369935, rs753394490, rs753515268, rs753538537, rs753544604, rs753559577, rs753584953, rs753586443, rs753645617, rs753669888, rs753799106, rs753855954, rs753916547, rs753922991, rs753957198, rs753978854, rs753984051, rs754007849, rs754084606, rs754121865, rs754143054, rs754185765, rs754237486, rs754349771, rs754352179, rs754361243, rs754449789, rs754470269, rs754509471, rs754556294, rs75456243, rs754590483, rs754605940, rs754658405, rs754727174, rs754760973, rs754789530, rs754813526, rs754842493, rs754873504, rs754953938, rs754993545, rs755048015, rs755097739, rs755104001, rs755214541, rs755241934, rs755302582, rs755356399, rs755427482, rs755478106, rs755481959, rs755699576, rs755717984, rs75573829, rs755755006, rs755774401, rs755842617, rs755849686, rs755879428, rs755897639, rs755952813, rs756053699, rs756057085, rs756112905, rs756119932, rs756154307, rs756178320, rs756180172, rs756200185, rs756235323, rs756246100, rs756268067, rs756295616, rs756404565, rs756415796, rs756448576, rs756452265, rs756462490, rs756464237, rs756473465, rs756505031, rs756515140, rs756529272, rs756680798, rs756696677, rs756710776, rs756765003, rs756818179, rs756854915, rs75690069, rs757033446, rs757192263, rs757230598, rs757256566, rs757257239, rs757269681, rs757273036, rs757280057, rs75734689, rs757359624, rs757422230, rs757469424, rs757477224, rs757489584, rs757526227, rs757575809, rs757599190, rs757629096, rs757666810, rs757688854, rs757874738, rs757901159, rs757950773, rs758100232, rs758278530, rs758286813, rs758310971, rs758381507, rs758405707, rs758426138, rs758472782, rs758482810, rs758562320, rs758734365, rs758952886, rs758956940, rs758961244, rs759023768, rs759040048, rs759040212, rs759055604, rs759067356, rs759127625, rs75924020, rs759247964, rs759252542, rs759252663, rs759318772, rs759332579, rs759366918, rs759393936, rs759456959, rs759500714, rs759642903, rs759693000, rs759738968, rs759748483,, rs759792284 rs759807509, rs759893441, rs759901587, rs759950423, rs759955623,, rs759967732 rs759967924, rs759989319, rs760008124, rs760008689, rs760022885,, rs760060027 rs76012193, rs760128804, rs760232954, rs760253303,
rs760416170,, rs760444067 rs760473424 rs760473560, rs760503204, rs760542502, rs760579781,, rs760604342 rs760655980 rs760721207, rs760806353, rs760827214, rs76083403, 1 rs760842332 rs760852869, rs760915098, rs760924707, rs760946854,
rs760999964,, rs761067047 rs761103772 rs761123692, rs761201018, rs761232988, rs761255070,, rs761263261 rs761293315 rs761316840, rs76132524, rs761352592,
rs761352759,, rs761471269 rs761487712 rs761551, rs761552, rs761553, rs761590796, rs76164879, ι rs761679000, rs761698296, rs761758856, rs761770661, rs761777507,
rs761786252,, rs761867251 rs761883210 rs761953791, rs761959610, rs762007434, rs762049636,, rs762077144 rs762081679 rs762083785, rs762189801, rs762259956, rs762276711,, rs762283961 rs762327715 rs762350146, rs762361430, rs762367488, rs762415878,, rs762420328 rs762428905 rs762429807, rs762458117, rs762497293, rs762526563,, rs762617605 rs762622537 rs762681307, rs762681898, rs762685504, rs762720265,, rs762803649 rs762819478 rs762919869, rs762922388, rs763008544, rs763050363,, rs763078352 rs763100489 rs763185152, rs763210370, rs763221140, rs763317999,, rs763384784 rs763393319 rs763446618, rs763461489, rs763461497, rs763494837,, rs763541221 rs763568106 rs763585539, rs763625200, rs763713753, rs763724702,, rs763751456 rs763802729 rs763927016, rs763929451, rs763940445, rs763954125,, rs763959140 rs763997081 rs764014420, rs764019310, rs764037942, rs764108106,, rs764109174 rs76413672, rs764156853, rs764191833, rs764198570, rs76421660, rs764246485,, rs764384637 rs764428144, rs764436490, rs764463548, rs764500514, rs764509732,, rs764515154 rs764527113, rs764613231, rs764846471, rs764851198, rs764865705,, rs764919608 rs764934054, rs764934309, rs764943652, rs764964194, rs76497846, ι rs765006438 rs76511341, rs765179478, rs765183405, rs765201374, rs765248217, rs765273723,, rs765348620 rs76539413, rs765412392, rs765558781, rs765563243,
rs765591204,, rs765615691 rs765647583, rs765659544, rs765735477, rs765747509, rs765788785,, rs765825926 rs765833506, rs765889218, rs765913054, rs765925436, rs765965653,, rs765985710 rs766001707, rs766144891, rs766148892, rs766197800, rs766237827,, rs766254893 rs766414737, rs766429455, rs766515714, rs766538029, rs766584856,, rs766638246 rs766647421, rs766718545, rs766735261, rs766748486, rs766771475,, rs766775924 rs766803774, rs766826445, rs766858483, rs766951076, rs767110312 rs767119545, rs767169937, rs767292810, rs767295299, rs76736333, rs767384339 rs767439371, rs767440950, rs767469497, rs767472593, rs767487088, rs767492310 rs767543973, rs767546866, rs767691121, rs767773214, rs767876813, rs767904273 rs767933266, rs767957502, rs767986517, rs768003087, rs768019941, rs768023930 rs768123597, rs768134157, rs768161284, rs768169130, rs768171400, rs768341819 rs768351340, rs768416640, rs768509381, rs768588123, rs768626702, rs768632002 rs768777946, rs768859725, rs768864175, rs76891773, rs768919726,
rs768991623 rs769103039, rs769110770, rs769251508, rs769298166, rs769305763, rs769306267 rs769342658, rs769358179, rs76940049, rs769451032, rs769463708,
rs769531021 rs769557211, rs769619585, rs769626047, rs769661532, rs769723876, rs769917266 rs769920949, rs769942768, rs769952981, rs770009219, rs770030225, rs770144688 rs770208148, rs77026122, rs77027532, rs770296185, rs770310984, rs770344520, rs770411294 rs770449937, rs770469290, rs770475357, rs770605522, rs770619918, rs770633953 rs770634865, rs770744771, rs770831500, rs77086230, rs770960334,
rs770983214 rs770993613, rs771012737, rs771030906, rs771083570, rs771098090, rs771119059 rs771135240, rs771162906, rs771181207, rs771187612, rs771252931, rs771418212 rs771430842, rs771434175, rs771460028, rs771470524, rs771525981, rs771549961 rs771553430, rs771599980, rs771687506, rs771749551, rs771793716, rs771829843 rs771865735, rs771915756, rs771969274, rs772002441, rs772056412, rs772116798 rs772130867, rs772184438, rs77219009, rs772206876, rs772207846,
rs772264318 rs772344258, rs772351920, rs772352221, rs772363696, rs772535725, rs772572450 rs772588044, rs772712821, rs772727244, rs772732276, rs772761242, rs772832599 rs772892274, rs772910074, rs773031143, rs77303789, rs773081911,
rs773121195 rs773183853, rs773204962, rs773205912, rs773207107, rs773271553, rs773276931 rs773301187, rs773348467, rs773379500, rs773393343, rs773405950, rs773473590 rs773478445, rs773613201, rs773670890, rs773675927, rs773687956, rs773689935 rs773779655, rs773787977, rs773788085, rs773864210, rs773875930, rs773946774 rs773969792, rs773982221, rs773995813, rs774014555, rs774029356, rs774064667 rs77409924, rs774250246, rs774303481, rs774444716, rs774452961,
rs774505955 rs774520337, rs774529009, rs774532005, rs774687288, rs774695185, rs774699497 rs774718180, rs774747355, rs774914247, rs774939813, rs774964917, rs775029826 rs775080758, rs775201664, rs775298116, rs775305302, rs775314613, rs775378635 rs775431045, rs775494652, rs775498557, rs775653094, rs775657206, rs775680424, rs775690573, rs775738970, rs775780769, rs775830742, rs775844276, rs775850621, rs775851457, rs775879957, rs775929807, rs775931116, rs776010660, rs776051012, rs776061658, rs776076724, rs776116861, rs776222614, rs776239444, rs776252804, rs776507665, rs776508242, rs776597067, rs776600319, rs776626841, rs776628724, rs776707067, rs776716583, rs776828308, rs776899714, rs776908103, rs776918783, rs776934440, rs776950796, rs777008986, rs777061035, rs777068966, rs777075482, rs777100615, rs777117773, rs777178784, rs777178859, rs777206722, rs777278826, rs777319844, rs777323537, rs777361034, rs777457130, rs777496733, rs777522387, rs777541224, rs777548288, rs777573209, rs777583007, rs777648912, rs777729152, rs777844695, rs777859825, rs777912812, rs777952246, rs777954225, rs778112508, rs778143831, rs778200373, rs778221475, rs778227441, rs778230305, rs778249126, rs778309138, rs77840532, rs778421268, rs778421428, rs778504040,
rs778509138, rs778630145, rs778679887, rs778704240, rs778722441, rs778789395, rs778956208, rs779005866, rs779035545, rs779080467, rs779138425, rs779172677, rs779186248, rs779188574, rs779199895, rs779255079, rs779276322, rs779281971, rs779384674, rs779470628, rs779491671, rs779560599, rs779656254, rs779681037, rs77970066, rs779769299, rs779782283, rs779811232, rs779829271, rs779838280,
rs779845888, rs779866271, rs779933798, rs779942460, rs779963548, rs780075943, rs780113339, rs780142385, rs78014519, rs780256409, rs780308101, rs780326779,
rs780362590, rs780511182, rs780591975, rs780604497, rs780623755, rs780627255, rs780734968, rs780792601, rs780806094, rs780880202, rs780895696, rs780906123, rs780972744, rs780973227, rs780988220, rs780997768, rs781063243, rs781068463, rs781154422, rs781273115, rs781275143, rs781295503, rs781349710, rs781356865, rs78143585, rs781437447, rs781475859, rs781556964, rs781563953, rs781702814,
rs781706817, rs781727107, rs78351983, rs78530327, rs78614131, rs78681557, rs78762669, rs78764556, rs78969646, rs79094230, rs79316375, rs79357803, rs79473894, rs79801200, rs79873516, rs8003041, rs8003520, rs8004149, rs8005484, rs8005824, rs8005854, rs8006287, rs8006491, rs8006829, rs8006970, rs8008215, rs8008405, rs8009033, rs8009456, rs8009896, rs8012666, rs8013582, rs8013669, rs8014385, rs8014509, rs8014592, rs8014854, rs80159833, rs80168292, rs8018900, rs8019545, rs8019838, rs8020781, rs8022601, rs8022758, rs80230489, rs872756, rs910369, rs9652396, rs978219, and rs978220.
[000376] In one example, the guide RNA used in the present disclosure may comprise at least one 20 nucleotide (nt) target nucleic acid sequence listed in Table 5. Provided in Table 5 are the gene symbol and the sequence identifier of the gene (Gene SEQ ID NO), the gene sequence including 1-5 kilobase pairs upstream and/or downstream of the target gene (Extended Gene SEQ ID NO), and the 20 nt target nucleic acid sequence (20 nt Target Sequence SEQ ID NO). In the sequence listing the respective target gene, the strand for targeting the gene (noted by a (+) strand or (-) strand in the sequence listing), the associated PAM type and the PAM sequence are described for each of the 20 nt target nucleic acid sequences (SEQ ID NO: 5305-56902). It is understood in the art that the spacer sequence, where "T" is "U," may be an RNA sequence corresponding to the 20 nt sequences listed in Table 5.
Table 5. Nucleic Acid Sequences
Figure imgf000112_0001
[000377] In one example, the guide RNA used in the present disclosure may comprise at least one spacer sequence that, where "T" is "U", may be an RNA sequence corresponding to a 20 nucleotide (nt) target sequence such as, but not limited to, any of SEQ ID NO: 5305-56902.
[000378] In one example, the guide RNA used in the present disclosure may comprise at least one spacer sequence which, where "T" is "U," is an RNA sequence corresponding to the 20 nt sequences such as, but not limited to, any of SEQ ID NO: 5305-56902.
[000379] In one example, a guide RNA may comprise a 20 nucleotide (nt) target nucleic acid sequence associated with the PAM type such as, but not limited to, NAAAAC, NNAGAAW, NNGRRT, NNNNGHTT, NRG, or YTN. As a non-limiting example, the 20 nt target nucleic acid sequence for a specific target gene and a specific PAM type may be, where "T" is "U," the RNA sequence corresponding to any one of the 20 nt nucleic acid sequences in Table 6.
Table 6. Nucleic Acid Sequences by PAM Type
Figure imgf000112_0002
[000380] In one example, a guide RNA may comprise a 20 nucleotide (nt) target nucleic acid sequence associated with the YTN PAM type. As a non-limiting example, the 20 nt target nucleic acid sequence for a specific target gene may comprise a 20 nt core sequence where the 20 nt core sequence, where "T" is "U," may be the RNA sequence corresponding to SEQ ID NO: 30886-56902. As another non-limiting example, the 20 nt target nucleic acid sequence for a specific target gene may comprise a core sequence where the core sequence, where "T" is "U," may be a fragment, segment or region of the RNA sequence corresponding to any of SEQ ID NO: 30886-56902.
VI. OTHER THERAPEUTIC APPROACHES
[000381] Gene editing can be conducted using nucleases engineered to target specific sequences. To date there are four major types of nucleases: meganucleases and their derivatives, zinc finger nucleases (ZFNs), transcription activator like effector nucleases (TALENs), and CRISPR-Cas9 nuclease systems. The nuclease platforms vary in difficulty of design, targeting density and mode of action, particularly as the specificity of ZFNs and TALENs is through protein-DNA interactions, while RNA-DNA interactions primarily guide Cas9.
[000382] CRISPR endonucleases, such as Cas9, can be used in the methods of the present disclosure. However, the teachings described herein, such as therapeutic target sites, could be applied to other forms of endonucleases, such as ZFNs, TALENs, HEs, or MegaTALs, or using combinations of nucleases. However, in order to apply the teachings of the present disclosure to such endonucleases, one would need to, among other things, engineer proteins directed to the specific target sites.
[000383] Additional binding domains can be fused to the Cas9 protein to increase specificity. The target sites of these constructs would map to the identified gRNA specified site, but would require additional binding motifs, such as for a zinc finger domain. In the case of Mega-TAL, a meganuclease can be fused to a TALE DNA-binding domain. The meganuclease domain can increase specificity and provide the cleavage. Similarly, inactivated or dead Cas9 (dCas9) can be fused to a cleavage domain and require the sgRNA/Cas9 target site and adjacent binding site for the fused DNA-binding domain. This likely would require some protein engineering of the dCas9, in addition to the catalytic inactivation, to decrease binding without the additional binding site.
Zinc Finger Nucleases
[000384] Zinc finger nucleases (ZFNs) are modular proteins comprised of an engineered zinc finger DNA binding domain linked to the catalytic domain of the type II endonuclease Fokl. Because Fokl functions only as a dimer, a pair of ZFNs must be engineered to bind to cognate target "half-site" sequences on opposite DNA strands and with precise spacing between them to enable the catalytically active Fokl dimer to form. Upon dimerization of the Fokl domain, which itself has no sequence specificity per se, a DNA double-strand break is generated between the ZFN half-sites as the initiating step in genome editing.
[000385] The DNA binding domain of each ZFN is typically comprised of 3-6 zinc fingers of the abundant Cys2-His2 architecture, with each finger primarily recognizing a triplet of nucleotides on one strand of the target DNA sequence, although cross-strand interaction with a fourth nucleotide also can be important. Alteration of the amino acids of a finger in positions that make key contacts with the DNA alters the sequence specificity of a given finger. Thus, a four-finger zinc finger protein will selectively recognize a 12 bp target sequence, where the target sequence is a composite of the triplet preferences contributed by each finger, although triplet preference can be influenced to varying degrees by neighboring fingers. An important aspect of ZFNs is that they can be readily re-targeted to almost any genomic address simply by modifying individual fingers, although considerable expertise is required to do this well. In most applications of ZFNs, proteins of 4-6 fingers are used, recognizing 12-18 bp respectively.
Hence, a pair of ZFNs will typically recognize a combined target sequence of 24-36 bp, not including the typical 5-7 bp spacer between half-sites. The binding sites can be separated further with larger spacers, including 15-17 bp. A target sequence of this length is likely to be unique in the human genome, assuming repetitive sequences or gene homologs are excluded during the design process. Nevertheless, the ZFN protein-DNA interactions are not absolute in their specificity so off-target binding and cleavage events do occur, either as a heterodimer between the two ZFNs, or as a homodimer of one or the other of the ZFNs. The latter possibility has been effectively eliminated by engineering the dimerization interface of the Fokl domain to create "plus" and "minus" variants, also known as obligate heterodimer variants, which can only dimerize with each other, and not with themselves. Forcing the obligate heterodimer prevents formation of the homodimer. This has greatly enhanced specificity of ZFNs, as well as any other nuclease that adopts these Fokl variants.
[000386] A variety of ZFN-based systems have been described in the art, modifications thereof are regularly reported, and numerous references describe rules and parameters that are used to guide the design of ZFNs; see, e.g., Segal et al., Proc Natl Acad Sci USA 96(6):2758-63 (1999); Dreier B et al., J Mol Biol. 303(4):489-502 (2000); Liu Q et al., J Biol Chem. 277(6):3850-6 (2002); Dreier et al., J Biol Chem 280(42):35588-97 (2005); and Dreier et al., J Biol Chem. 276(31):29466-78 (2001). Transcription Activator-Like Effector Nucleases (TALENs)
[000387] TALENs represent another format of modular nucleases whereby, as with ZFNs, an engineered DNA binding domain is linked to the Fokl nuclease domain, and a pair of TALENs operate in tandem to achieve targeted DNA cleavage. The major difference from ZFNs is the nature of the DNA binding domain and the associated target DNA sequence recognition properties. The TALEN DNA binding domain derives from TALE proteins, which were originally described in the plant bacterial pathogen Xanthomonas sp. TALEs are comprised of tandem arrays of 33-35 amino acid repeats, with each repeat recognizing a single base pair in the target DNA sequence that is typically up to 20 bp in length, giving a total target sequence length of up to 40 bp. Nucleotide specificity of each repeat is determined by the repeat variable diresidue (RVD), which includes just two amino acids at positions 12 and 13. The bases guanine, adenine, cytosine and thymine are predominantly recognized by the four RVDs: Asn- Asn, Asn-Ile, His-Asp and Asn-Gly, respectively. This constitutes a much simpler recognition code than for zinc fingers, and thus represents an advantage over the latter for nuclease design. Nevertheless, as with ZFNs, the protein-DNA interactions of TALENs are not absolute in their specificity, and TALENs have also benefitted from the use of obligate heterodimer variants of the Fokl domain to reduce off-target activity.
[000388] Additional variants of the Fokl domain have been created that are deactivated in their catalytic function. If one half of either a TALEN or a ZFN pair contains an inactive Fokl domain, then only single-strand DNA cleavage (nicking) will occur at the target site, rather than a DSB. The outcome is comparable to the use of CRISPR/Cas9 or CRISPR/Cpfl "nickase" mutants in which one of the Cas9 cleavage domains has been deactivated. DNA nicks can be used to drive genome editing by FIDR, but at lower efficiency than with a DSB. The main benefit is that off-target nicks are quickly and accurately repaired, unlike the DSB, which is prone to NHEJ-mediated mis-repair.
[000389] A variety of TALEN-based systems have been described in the art, and modifications thereof are regularly reported; see, e.g., Boch, Science 326(5959): 1509-12 (2009); Mak et al., Science 335(6069):716-9 (2012); and Moscou et al., Science 326(5959): 1501 (2009). The use of TALENs based on the "Golden Gate" platform, or cloning scheme, has been described by multiple groups; see, e.g., Cermak et al., Nucleic Acids Res. 39(12):e82 (2011); Li et al., Nucleic Acids Res. 39(14):6315-25(2011); Weber et al., PLoS One. 6(2):el6765 (2011); Wang et al., J Genet Genomics 41(6):339-47, Epub 2014 May 17 (2014); and Cermak T et al., Methods Mol Biol. 1239: 133-59 (2015). Homing Endonucleases
[000390] Homing endonucleases (HEs) are sequence-specific endonucleases that have long recognition sequences (14-44 base pairs) and cleave DNA with high specificity - often at sites unique in the genome. There are at least six known families of HEs as classified by their structure, including GIY-YIG, His-Cis box, H-N-H, PD-(D/E)xK, and Vsr-like that are derived from a broad range of hosts, including eukarya, protists, bacteria, archaea, cyanobacteria and phage. As with ZFNs and TALENs, HEs can be used to create a DSB at a target locus as the initial step in genome editing. In addition, some natural and engineered HEs cut only a single strand of DNA, thereby functioning as site-specific nickases. The large target sequence of HEs and the specificity that they offer have made them attractive candidates to create site-specific DSBs.
[000391] A variety of HE-based systems have been described in the art, and modifications thereof are regularly reported; see, e.g., the reviews by Steentoft et al., Glycobiology 24(8):663- 80 (2014); Belfort and Bonocora, Methods Mol Biol. 1123 : 1-26 (2014); Hafez and Hausner, Genome 55(8):553-69 (2012); and references cited therein.
MegaTAL / Tev-mTALEN / MegaTev
[000392] As further examples of hybrid nucleases, the MegaTAL platform and Tev-mTALEN platform use a fusion of TALE DNA binding domains and catalytically active HEs, taking advantage of both the tunable DNA binding and specificity of the TALE, as well as the cleavage sequence specificity of the HE; see, e.g., Boissel et al., NAR 42: 2591-2601 (2014); Kleinstiver et al., G3 4: 1155-65 (2014); and Boissel and Scharenberg, Methods Mol. Biol. 1239: 171-96 (2015).
[000393] In a further variation, the MegaTev architecture is the fusion of a meganuclease (Mega) with the nuclease domain derived from the GIY-YIG homing endonuclease I-Tevl (Tev). The two active sites are positioned ~30 bp apart on a DNA substrate and generate two DSBs with non-compatible cohesive ends; see, e.g., Wolfs et al., NAR 42, 8816-29 (2014). It is anticipated that other combinations of existing nuclease-based approaches will evolve and be useful in achieving the targeted genome modifications described herein.
dCas9-FokI or dCpfl-Fokl and Other Nucleases
[000394] Combining the structural and functional properties of the nuclease platforms described above offers a further approach to genome editing that can potentially overcome some of the inherent deficiencies. As an example, the CRISPR genome editing system typically uses a single Cas9 endonuclease to create a DSB. The specificity of targeting is driven by a 20 or 24 nucleotide sequence in the guide RNA that undergoes Watson-Crick base-pairing with the target DNA (plus an additional 2 bases in the adjacent NAG or NGG PAM sequence in the case of Cas9 from S. pyogenes). Such a sequence is long enough to be unique in the human genome, however, the specificity of the RNA/DNA interaction is not absolute, with significant promiscuity sometimes tolerated, particularly in the 5' half of the target sequence, effectively reducing the number of bases that drive specificity. One solution to this has been to completely deactivate the Cas9 or Cpfl catalytic function - retaining only the RNA-guided DNA binding function - and instead fusing a Fokl domain to the deactivated Cas9; see, e.g., Tsai et al., Nature Biotech 32: 569-76 (2014); and Guilinger et al., Nature Biotech. 32: 577-82 (2014). Because Fokl must dimerize to become catalytically active, two guide RNAs are required to tether two Fokl fusions in close proximity to form the dimer and cleave DNA. This essentially doubles the number of bases in the combined target sites, thereby increasing the stringency of targeting by CRISPR-based systems.
[000395] As further example, fusion of the TALE DNA binding domain to a catalytically active FIE, such as I-Tevl, takes advantage of both the tunable DNA binding and specificity of the TALE, as well as the cleavage sequence specificity of I-Tevl, with the expectation that off- target cleavage can be further reduced.
VII. KITS
[000396] The present disclosure provides kits for carrying out the methods described herein. A kit can include one or more of a genome-targeting nucleic acid, a polynucleotide encoding a genome-targeting nucleic acid, a site-directed polypeptide, a polynucleotide encoding a site- directed polypeptide, and/or any nucleic acid or proteinaceous molecule necessary to carry out the aspects of the methods described herein, or any combination thereof.
[000397] A kit can comprise: (1) a vector comprising a nucleotide sequence encoding a genome-targeting nucleic acid, (2) the site-directed polypeptide or a vector comprising a nucleotide sequence encoding the site-directed polypeptide, and (3) a reagent for reconstitution and/or dilution of the vector(s) and or polypeptide.
[000398] A kit can comprise: (1) a vector comprising (i) a nucleotide sequence encoding a genome-targeting nucleic acid, and (ii) a nucleotide sequence encoding the site-directed polypeptide; and (2) a reagent for reconstitution and/or dilution of the vector.
[000399] In any of the above kits, the kit can comprise a single-molecule guide genome- targeting nucleic acid. In any of the above kits, the kit can comprise a double-molecule genome- targeting nucleic acid. In any of the above kits, the kit can comprise two or more double- molecule guides or single-molecule guides. The kits can comprise a vector that encodes the nucleic acid targeting nucleic acid.
[000400] In any of the above kits, the kit can further comprise a polynucleotide to be inserted to effect the desired genetic modification.
[000401] Components of a kit can be in separate containers, or combined in a single container.
[000402] Any kit described above can further comprise one or more additional reagents, where such additional reagents are selected from a buffer, a buffer for introducing a polypeptide or polynucleotide into a cell, a wash buffer, a control reagent, a control vector, a control RNA polynucleotide, a reagent for in vitro production of the polypeptide from DNA, adaptors for sequencing and the like. A buffer can be a stabilization buffer, a reconstituting buffer, a diluting buffer, or the like. A kit can also comprise one or more components that can be used to facilitate or enhance the on-target binding or the cleavage of DNA by the endonuclease, or improve the specificity of targeting.
[000403] In addition to the above-mentioned components, a kit can further comprise instructions for using the components of the kit to practice the methods. The instructions for practicing the methods can be recorded on a suitable recording medium. For example, the instructions can be printed on a substrate, such as paper or plastic, etc. The instructions can be present in the kits as a package insert, in the labeling of the container of the kit or components thereof {i.e., associated with the packaging or subpackaging), etc. The instructions can be present as an electronic storage data file present on a suitable computer readable storage medium, e.g. CD-ROM, diskette, flash drive, etc. In some instances, the actual instructions are not present in the kit, but means for obtaining the instructions from a remote source {e.g. via the Internet), can be provided. An example of this case is a kit that comprises a web address where the instructions can be viewed and/or from which the instructions can be downloaded. As with the instructions, this means for obtaining the instructions can be recorded on a suitable substrate.
VIII. SPECIFIC METHODS AND COMPOSITIONS OF THE INVENTION
[000404] Accordingly, the present disclosure relates in particular to the following non-limiting methods according to the present disclosure: In a first method, Method 1, the present disclosure provides a method for editing a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell by genome editing comprising: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
[000405] In another method, Method 2, the present disclosure provides a method for editing a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell by genome editing comprising: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
[000406] In another method, Method 3, the present disclosure provides an ex vivo method for treating a patient having an ATXN3 related condition or disorder comprising: creating a patient specific induced pluripotent stem cell (iPSC); editing within or near a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene or other DNA sequences that encode regulatory elements of the ATXN3 gene of the iPSC; differentiating the genome-edited iPSC into a neuron or glial cell of the Central Nervous System (CNS); and implanting the neuron or glial cell of the Central Nervous System (CNS) into the patient.
[000407] In another method, Method 4, the present disclosure provides the method of Method 3, wherein the editing step comprises: introducing into the iPSC one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
[000408] In another method, Method 5, the present disclosure provides the method of Method 3, wherein the editing step comprises: introducing into the iPSC one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
[000409] In another method, Method 6, the present disclosure provides an ex vivo method for treating a patient having an ATXN3 related condition or disorder comprising: isolating a mesenchymal stem cell from the patient; editing within or near a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene or other DNA sequences that encode regulatory elements of the ATXN3 gene of the mesenchymal stem cell; differentiating the genome-edited mesenchymal stem cell into a neuron or glial cell of the Central Nervous System (CNS); and implanting the neuron or glial cell of the Central Nervous System (CNS) into the patient.
[000410] In another method, Method 7, the present disclosure provides the method of Method 6, wherein the editing step comprises: introducing into the mesenchymal stem cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
[000411] In another method, Method 8, the present disclosure provides the method of Method 6, wherein the editing step comprises: introducing into the mesenchymal stem cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
[000412] In another method, Method 9, the present disclosure provides an in vivo method for treating a patient with an ATXN3 related disorder comprising: editing the Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell of the patient.
[000413] In another method, Method 10, the present disclosure provides the method of Method 9, wherein the editing step comprises: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
[000414] In another method, Method 11, the present disclosure provides the method of Method 9, wherein the editing step comprises: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
[000415] In another method, Method 12, the present disclosure provides the method of any one of Methods 9-11, wherein the cell is a cell of the Central Nervous System (CNS).
[000416] In another method, Method 13, the present disclosure provides the method of Method 12, wherein the cell of the Central Nervous System (CNS) is a neuron.
[000417] In another method, Method 14, the present disclosure provides the method of Method 12, wherein the cell of the Central Nervous System (CNS) is a glial cell.
[000418] In another method, Method 15, the present disclosure provides the method of any one of Methods 12-14, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is delivered to the cell of the Central Nervous System (CNS) via any administration route selected from the group consisting of intraparenchymal, intravenous, intra-arterial,
intracerebroventricular, intracisternal, intrathecal, intracranial or intraperitoneal routes.
[000419] In another method, Method 16, the present disclosure provides a method of altering the contiguous genomic sequence of an ATXN3 gene in a cell comprising contacting the cell with one or more deoxyribonucleic acid (DNA) endonuclease to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs).
[000420] In another method, Method 17, the present disclosure provides the method of Method 16, wherein the alteration of the contiguous genomic sequence occurs in one or more exons of the ATXN3 gene.
[000421] In another method, Method 18, the present disclosure provides the method of Method 17, wherein the alteration of the contiguous genomic sequence occurs in exon 10 of the ATXN3 gene.
[000422] In another method, Method 19, the present disclosure provides the method of any one of Methods 1-18, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is selected from any of those sequence in SEQ ID NOs: 1-620 and variants having at least 90% homology to any of those sequences disclosed in SEQ ID NOs: 1-620.
[000423] In another method, Method 20, the present disclosure provides the method of Method 19, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is one or more protein or polypeptide. [000424] In another method, Method 21, the present disclosure provides the method of Method 19, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is one or more polynucleotide encoding the one or more DNA endonuclease.
[000425] In another method, Method 22, the present disclosure provides the method of Method 21, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is one or more ribonucleic acid (RNA) encoding the one or more DNA endonuclease.
[000426] In another method, Method 23, the present disclosure provides the method of Method 22, wherein the one or more ribonucleic acid (RNA) is one or more chemically modified RNA.
[000427] In another method, Method 24, the present disclosure provides the method of Method 23, wherein the one or more ribonucleic acid (RNA) is chemically modified in the coding region.
[000428] In another method, Method 25, the present disclosure provides the method of any one of Methods 21-24, wherein the one or more polynucleotide or one or more ribonucleic acid (RNA) is codon optimized.
[000429] In another method, Method 26, the present disclosure provides the method of any one of Methods 1-25, wherein the method further comprises: introducing one or more gRNA or one or more sgRNA.
[000430] In another method, Method 27, the present disclosure provides the method of Method 26, wherein the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a sequence within or near the expanded trinucleotide repeat in the ATXN3 gene.
[000431] In another method, Method 28, the present disclosure provides the method of Method 26, wherein the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a DNA sequence within or near the ATXN3 gene.
[000432] In another method, Method 29, the present disclosure provides the method of Method 26, wherein the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a sequence flanking the ATXN3 gene or other sequence that encodes a regulatory element of the ATXN3 gene.
[000433] In another method, Method 30, the present disclosure provides the method of any one of Methods 26-29, wherein the one or more gRNA or one or more sgRNA is chemically modified.
[000434] In another method, Method 31, the present disclosure provides the method of any one of Methods 26-30, wherein the one or more gRNA or one or more sgRNA is pre-complexed with the one or more deoxyribonucleic acid (DNA) endonuclease. [000435] In another method, Method 32, the present disclosure provides the method of Method 31, wherein the pre-complexing involves a covalent attachment of the one or more gRNA or one or more sgRNA to the one or more deoxyribonucleic acid (DNA) endonuclease.
[000436] In another method, Method 33, the present disclosure provides the method of any one of Methods 19-32, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is formulated in a liposome or lipid nanoparticle.
[000437] In another method, Method 34, the present disclosure provides the method of any one of Methods 26-32, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is formulated in a liposome or a lipid nanoparticle which also comprises the one or more gRNA or one or more sgRNA.
[000438] In another method, Method 35, the present disclosure provides the method of any one of Methods 19, or 26-29, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is encoded in an AAV vector particle.
[000439] In another method, Method 36, the present disclosure provides the method of any of the Methods 26-29, wherein the one or more gRNA or one or more sgRNA is encoded in an AAV vector particle.
[000440] In another method, Method 37, the present disclosure provides the method of any of the Methods 26-29, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is encoded in an AAV vector particle which also encodes the one or more gRNA or one or more sgRNA.
[000441] In another method, Method 38, the present disclosure provides the method of any of the Methods 35-37, wherein the AAV vector particle is selected from the group consisting of any of those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
[000442] In another method, Method 39, the present disclosure provides the method of any of Methods 1-38, wherein the method further comprises: introducing into the cell a donor template comprising at least a portion of the wild-type ATXN3 gene.
[000443] In another method, Method 40, the present disclosure provides the method of Method 39, wherein the at least a portion of the wild-type ATXN3 gene comprises one or more sequences selected from the group consisting of: an ATXN3 exon, an ATXN3 intron, and a sequence comprising an exon:intron junction of ATXN3.
[000444] In another method, Method 41, the present disclosure provides the method of any one of Methods 39-40, wherein the donor template comprises homologous arms to the genomic locus of the ATXN3 gene. [000445] In another method, Method 42, the present disclosure provides the method of any one of Methods 39-41, wherein the donor template is either a single or double stranded
polynucleotide.
[000446] In another method, Method 43, the present disclosure provides the method of any one of Methods 39-42, wherein the donor template is encoded in an AAV vector particle, where the AAV vector particle is selected from the group consisting of those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
[000447] In another method, Method 44, the present disclosure provides the method of any one of Methods 39-42, wherein the one or more polynucleotide encoding one or more
deoxyribonucleic acid (DNA) endonuclease is formulated into a lipid nanoparticle, and the one or more gRNA or one or more sgRNA is delivered to the cell ex vivo by electroporation and the donor template is delivered to the cell by an adeno-associated virus (AAV) vector.
[000448] In another method, Method 45, the present disclosure provides the method of any one of Methods 39-42, wherein the one or more polynucleotide encoding one or more
deoxyribonucleic acid (DNA) endonuclease is formulated into a liposome or lipid nanoparticle which also comprises the one or more gRNA or one or more sgRNA and the donor template.
[000449] The present disclosure also provides a composition, Composition 1, comprising a single-molecule guide RNA comprising: at least a spacer sequence that is an RNA sequence corresponding to any of SEQ ID NOs: 5305-56902.
[000450] In another composition, Composition 2, the present disclosure provides the single- molecule guide RNA of Composition 1, wherein the single-molecule guide RNA further comprises a spacer extension region.
[000451] In another composition, Composition 3, the present disclosure provides the single- molecule guide RNA of Composition 1, wherein the single-molecule guide RNA further comprises a tracrRNA extension region.
[000452] In another composition, Composition 4, the present disclosure provides the single- molecule guide RNA of Compositions 1-3, wherein the single-molecule guide RNA is chemically modified.
[000453] In another composition, Composition 5, the present disclosure provides a single- molecule guide RNA of Compositions 1-4 pre-complexed with a DNA endonuclease.
[000454] In another composition, Composition 6, the present disclosure provides the composition of Composition 5, wherein the DNA endonuclease is a Cas9 or Cpfl endonuclease. [000455] In another composition, Composition 7, the present disclosure provides the composition of Composition 6, wherein the Cas9 or Cpfl endonuclease is selected from the group consisting of: S. pyogenes Cas9, S. aureus Cas9, N. meningitides Cas9, S. thermophilus CRISPRl Cas9, S. thermophilus CRISPR 3 Cas9, T. denticola Cas9, L. bacterium D2006 Cpfl and Acidaminococcus sp. BV3L6 Cpfl, and variants having at least 90% homology to the endonucleases.
[000456] In another composition, Composition 8, the present disclosure provides the composition of Composition 7, wherein the Cas9 or Cpfl endonuclease comprises one or more nuclear localization signals (NLSs).
[000457] In another composition, Composition 9, the present disclosure provides the composition of Composition 8, wherein at least one NLS is at or within 50 amino acids of the amino-terminus of the Cas9 or Cpfl endonucelase and/or at least one NLS is at or within 50 amino acids of the carboxy -terminus of the Cas9 or Cpfl endonucelase.
[000458] In another compostion, Composition 10, the present disclosure provides a DNA encoding the single-molecule guide RNA of any of Compositions 1-4.
[000459] In another composition, Composition 11, the present disclosure provides a non- naturally occurring CRISPR/Cas system comprising a polynucleotide encoding a Cas9 or Cpfl enzyme and at least one single-molecule guide RNA from Compositions 1-4.
[000460] In another composition, Composition 12, the present disclosure provides the
CRISPR/Cas system of Composition 11, wherein the polynucleotide encoding a Cas9 or Cpfl endonuclease is selected from the group consisting of: S. pyogenes Cas9, S. aureus Cas9, N. meningitides Cas9, S. thermophilus CRISPRl Cas9, S. thermophilus CRISPR 3 Cas9, T.
denticola Cas9, L. bacterium ND2006 Cpfl and Acidaminococcus sp. BV3L6 Cpfl, and variants having at least 90% homology to the endonucleases.
[000461] In another composition, Composition 13, the present disclosure provides the
CRISPR/Cas system of Composition 12, wherein the polynucleotide encoding a Cas9 or Cpfl enzyme comprises one or more nuclear localization signals (NLSs).
[000462] In another composition, Composition 14, the present disclosure provides the
CRISPR/Cas system of Composition 13, wherein at least one NLS is at or within 50 amino acids of the amino-terminus of the polynucleotide encoding a Cas9 or Cpfl endonuclease and/or at least one NLS is at or within 50 amino acids of the carboxy-terminus of the polynucleotide encoding a Cas9 or Cpfl endonuclease. [000463] In another composition, Composition 15, the present disclosure provides the
CRISPR/Cas system of Composition 14, wherein the polynucleotide encoding a Cas9 or Cpfl endonuclease is codon optimized for expression in a eukaryotic cell.
[000464] In another composition, Composition 16, the present disclosure provides a DNA encoding the CRISPR/Cas system of any one of Compositions 11-15.
[000465] In another composition, Composition 17, the present disclosure provides a vector comprising the DNA of Compositions of 11 or 16.
[000466] In another composition, Composition 18, the present disclosure provides the vector of Composition 17, wherein the vector is a plasmid.
[000467] In another composition, Composition 19, the present disclosure provides the vector of Composition 17, wherein the vector is an AAV vector particle, and the AAV vector serotype is selected from the group consisting of those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
IX. DEFINITIONS
[000468] The term "comprising" or "comprises" is used in reference to compositions, methods, and respective component(s) thereof, that are essential to the present disclosure, yet open to the inclusion of unspecified elements, whether essential or not.
[000469] The term "consisting essentially of refers to those elements required for a given aspect. The term permits the presence of additional elements that do not materially affect the basic and novel or functional characteristic(s) of that aspect of the present disclosure.
[000470] The term "consisting of refers to compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the aspect.
[000471] The singular forms "a," "an," and "the" include plural references, unless the context clearly dictates otherwise.
[000472] Any numerical range recited in this specification describes all sub-ranges of the same numerical precision (i.e., having the same number of specified digits) subsumed within the recited range. For example, a recited range of "1.0 to 10.0" describes all sub-ranges between (and including) the recited minimum value of 1.0 and the recited maximum value of 10.0, such as, for example, "2.4 to 7.6," even if the range of "2.4 to 7.6" is not expressly recited in the text of the specification. Accordingly, the Applicant reserves the right to amend this specification, including the claims, to expressly recite any sub-range of the same numerical precision subsumed within the ranges expressly recited in this specification. All such ranges are inherently described in this specification such that amending to expressly recite any such sub-ranges will comply with written description, sufficiency of description, and added matter requirements, including the requirements under 35 U.S.C. § 112(a) and Article 123(2) EPC. Also, unless expressly specified or otherwise required by context, all numerical parameters described in this specification (such as those expressing values, ranges, amounts, percentages, and the like) may be read as if prefaced by the word "about," even if the word "about" does not expressly appear before a number. Additionally, numerical parameters described in this specification should be construed in light of the number of reported significant digits, numerical precision, and by applying ordinary rounding techniques. It is also understood that numerical parameters described in this specification will necessarily possess the inherent variability characteristic of the underlying measurement techniques used to determine the numerical value of the parameter.
[000473] The details of one or more examples of the present disclosure are set forth in the accompanying description below. Although any materials and methods similar or equivalent to those described herein can be used in the practice or testing of the present disclosure, the preferred materials and methods are now described. Other features, objects and advantages of the present disclosure will be apparent from the description. In the description, the singular forms also include the plural unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. In the case of conflict, the present description will control.
[000474] The present disclosure is further illustrated by the following non-limiting examples.
X. EXAMPLES
[000475] The present disclosure will be more fully understood by reference to the following examples, which provide illustrative non-limiting aspects of the invention.
[000476] The examples describe the use of the CRISPR system as an illustrative genome editing technique to create defined therapeutic genomic deletions, insertions, or replacements, termed "genomic modifications" herein, in the ATXN3 gene that lead to permanent deletion or correction of expanded trinucleotide repeats in the ATXN3 gene, knock-out of the ATXN3 gene, or correction of the entire gene or correction of mutations within the gene, that restore the wild- type or similar levels of trinucleotide repeats in the ATXN3 gene and/or eliminate aberrant ATXN3 gene products. [000477] All tested gRNAs can be used for an HDR/correction based editing approach. Single gRNAs can also be used to induce insertions and deletions disrupting the expression of the mutant ATXN3 gene. Selected pairs of gRNAs can be used to make deletions in the mutant ATXN3 gene that disrupts expression.
[000478] Various Cas orthologs are evaluated for cutting. gRNAs are delivered as RNA and expressed from the U6 promoter in plasmids. The corresponding Cas protein is either knocked into the cell line of interest and constitutively expressed, delivered as mRNA, or delivered as protein. The gRNA activity in all formats is evaluated using TIDE analysis in HEK293T cells.
[000479] Introduction of the defined therapeutic modifications described above represents a novel therapeutic strategy for the potential amelioration of Spinocerebellar ataxia 3 (SCA3), and related disorders, as described and illustrated herein.
Example 1 - CRISPR/SpCas9 target sites for the ATXN3 gene
[000480] Regions of the ATXN3 gene were scanned for target sites. Each area was scanned for a protospacer adjacent motif (PAM) having the sequence NRG. gRNA 20 bp spacer sequences corresponding to the PAM were then identified, as shown in SEQ ID NOs: 10698 - 30885 of the Sequence Listing.
Example 2 - CRISPR/SaCas9 target sites for the ATXN3 gene
[000481] Regions of the ATXN3 gene were scanned for target sites. Each area was scanned for a protospacer adjacent motif (PAM) having the sequence NNGRRT. gRNA 20 bp spacer sequences corresponding to the PAM were then identified, as shown in SEQ ID NOs: 6354 - 8658 of the Sequence Listing.
Example 3 - CRISPR/StCas9 target sites for the ATXN3 gene
[000482] Regions of the ATXN3 gene were scanned for target sites. Each area was scanned for a protospacer adjacent motif (PAM) having the sequence NNAGAAW. gRNA 20 bp spacer sequences corresponding to the PAM were then identified, as shown in SEQ ID NOs: 5634 - 6353 of the Sequence Listing.
Example 4 - CRISPR/TdCas9 target sites for the ATXN3 gene
[000483] Regions of the ATXN3 gene were scanned for target sites. Each area was scanned for a protospacer adjacent motif (PAM) having the sequence NAAAAC. gRNA 20 bp spacer sequences corresponding to the PAM were then identified, as shown in SEQ ID NOs: 5305 - 5633 of the Sequence Listing. Example 5 - CRISPR/NmCas9 target sites for the ATXN3 gene
[000484] Regions of the ATXN3 gene were scanned for target sites. Each area was scanned for a protospacer adjacent motif (PAM) having the sequence NNNNGHTT. gRNA 20 bp spacer sequences corresponding to the PAM were then identified, as shown in SEQ ID NOs: 8659 - 10697 of the Sequence Listing.
Example 6 - CRISPR/Cpfl target sites for the ATXN3 gene
[000485] Regions of the ATXN3 gene were scanned for target sites. Each area was scanned for a protospacer adjacent motif (PAM) having the sequence YTN. gRNA 20 bp spacer sequences corresponding to the PAM were then identified, as shown in SEQ ID NOs: 30886 - 56902 of the Sequence Listing.
Example 7 - Bioinformatics analysis of the guide strands
[000486] Candidate guides will then be screened and selected in a single process or multi-step process that involves both theoretical binding and experimentally assessed activity at both on- target and off-target sites. By way of illustration, candidate guides having sequences that match a particular on-target site, such as a site within the ATXN3 gene, with adjacent PAM can be assessed for their potential to cleave at off-target sites having similar sequences, using one or more of a variety of bioinformatics tools available for assessing off-target binding, as described and illustrated in more detail below, in order to assess the likelihood of effects at chromosomal positions other than those intended.
[000487] Candidates predicted to have relatively lower potential for off-target activity can then be assessed experimentally to measure their on-target activity, and then off-target activities at various sites. Preferred guides have sufficiently high on-target activity to achieve desired levels of gene editing at the selected locus, and relatively lower off-target activity to reduce the likelihood of alterations at other chromosomal loci. The ratio of on-target to off-target activity is often referred to as the "specificity" of a guide.
[000488] For initial screening of predicted off-target activities, there are a number of bioinformatics tools known and publicly available that can be used to predict the most likely off- target sites; and since binding to target sites in the CRISPR/Cas9 or CRISPR/Cpfl nuclease system is driven by Watson-Crick base pairing between complementary sequences, the degree of dissimilarity (and therefore reduced potential for off-target binding) is essentially related to primary sequence differences: mismatches and bulges, i.e. bases that are changed to a non- complementary base, and insertions or deletions of bases in the potential off-target site relative to the target site. An exemplary bioinformatics tool called COSMID (CRISPR Off-target Sites with Mismatches, Insertions and Deletions) (available on the web at crispr.bme.gatech.edu) compiles such similarities. Other bioinformatics tools include, but are not limited to
autoCOSMID and CCTop.
[000489] Bioinformatics were used to minimize off-target cleavage in order to reduce the detrimental effects of mutations and chromosomal rearrangements. Studies on CRISPR/Cas9 systems suggested the possibility of off-target activity due to non-specific hybridization of the guide strand to DNA sequences with base pair mismatches and/or bulges, particularly at positions distal from the PAM region. Therefore, it is important to have a bioinformatics tool that can identify potential off-target sites that have insertions and/or deletions between the RNA guide strand and genomic sequences, in addition to base-pair mismatches. Bioinformatics tools based upon the off-target prediction algorithm CCTop were used to search genomes for potential CRISPR off-target sites (CCTop is available on the web at crispr.cos.uni-heidelberg.de/). The output ranked lists of the potential off-target sites based on the number and location of mismatches, allowing more informed choice of target sites, and avoiding the use of sites with more likely off-target cleavage.
[000490] Additional bioinformatics pipelines were employed that weigh the estimated on- and/or off-target activity of gRNA targeting sites in a region. Other features that may be used to predict activity include information about the cell type in question, DNA accessibility, chromatin state, transcription factor binding sites, transcription factor binding data, and other CHIP-seq data. Additional factors were weighed that predict editing efficiency, such as relative positions and directions of pairs of gRNAs, local sequence features and micro-homologies.
[000491] Initial evaluation and screening of CRISPR/Cas9 target sites focused on the Intron 9, ExonlO and Intron 10 regions. gRNAs targeting ExonlO can be used for replacing the mutated endogenous ATXN3 gene and restoring protein expression. gRNAs targeting ExonlO can be used to insert a synthetic exon comprising Exon 10 and Exon 11 coding sequences. The synthetic exon can replace the mutated Exon 10 and restore ATXN3 protein expression. gRNAs targeting Intron 9 can be used to insert a synthetic exon comprising Exon 10 and Exon 11 coding sequences. The synthetic exon inserted into Intron 9 can restore ATXN3 protein expression.
[000492] Guide RNAs can be used to target the expanded repeats. Pairs of gRNAs on either side of the expanded repeat sequence can be used to excise the expanded repeats. gRNAs overlapping with expanded repeat sequence can be used to induce partial or full contraction of the repeats. [000493] In addition, carefully chosen gRNAs together with dCas9 fused to chromatin modifier proteins can be used to alleviate the epigenetic changes that occur at the ATXN3 allele with expanded repeats and restore ATXN3 expression. These gRNAs can also be used to edit one or more nucleotides, exonic sequences and/or intronic sequences.
[000494] Initial bioinformatics analysis identified 1778 guides targeting the Intron 9, ExonlO and Intron 10 regions. Further analysis of predicted off-target sites and evaluation of the gRNA target site within the ATXN3 gene resulted in 192 gRNAs selected for screening. The prioritized list of 192 single gRNAs targeting the ATXN3 gene were tested for cutting efficiencies using spCas9. Data was obtained for 85 of the 192 gRNAs (Table 7).
Table 7
SEQ ID Name Sequence Indel R2 NO.
26186
ATXN3_Int9_Intl 0_T65 TCCTACTAATGAGCTCTACA 36.8 0.9656
15286
ATXN3_Int9_Int 10_T 153 TTAGCTTACATGCCATAAAG 29.7 0.8552
25810
ATXN3_Int9_Intl 0_T208 CTCTGCCCCTCCACTCAAGA 23.7 0.9189
15577
ATXN3_Int9_Intl 0_T44 ACTGCTATAGCAATGATCCT 22.2 0.96
15273
ATXN3_Int9_Int 10_T123 GAATGTCAGGTGGCTACTTT 22 0.9756
15580
ATXN3_Int9_Int 10_T88 AGTTGGGACAATGGGTAATG 20.8 0.98
26157
ATXN3_Int9_Int 10_T 103 GCACCAGGGTCAATTTGGGA 20.3 0.7078
26158
ATXN3_Int9_Int 10_T261 CACCAGGGTCAATTTGGGAA 19 0.7386
15295
ATXN3_Int9_Int 10_T242 ACAGCACAGTTATCAGCACT 18.8 0.9812
26033
ATXN3_Int9_Int 10_T 186 GCTTCACTGCTGTTGCAATG 17.9 0.9726
15275
ATXN3_Int9_Int 10_T260 GATGAAATAATGGGAATGTC 16.6 0.9663
15572
ATXN3_Int9_Intl 0_T60 CCACCACAATTGCCTAGAGA 16.6 0.96
15710
ATXN3_Int9_Int 10 T113 TCTTTGGGAGCTTGGGTCTA 15.3 0.8257
26054
ATXN3_Int9_Int 10_T 128 CAAGTTGGTTTGTGGTACTG 15.2 0.98
15270
ATXN3_Int9_Int 10_T274 GGTGGCTACTTTTGGTGGGA 15 0.9673 26204
ATXN3_Int9_Int 10_T36 CAATCAATCCTCCACCAAGT 14.9 0.9595
26034
ATXN3_Int9_Intl 0_T232 CTTCACTGCTGTTGCAATGG 14.7 0.97
26729
ATXN3_Int9_IntlO_T52 TCCATTCCCCTGATGCTAAC 14.5 0.9274
15422
ATXN3_Int9_Intl 0_T246 CTATAATATTGATGGCACAG 14.4 0.8389
26737
ATXN3_Int9_IntlO_T54 ATGAACTACCACGTCTATTT 13.7 0.9948
26727
ATXN3_Int9_Int 10_T163 TAGGAGTTGATCCAGATTTA 12.9 0.9728
26154
ATXN3_Int9_Intl 0_T70 AATCAGCACCAGGGTCAATT 12.8 0.9859
15755
ATXN3_Int9_Intl 0_T225 TATACCTCCTTCCTACTTGG 12.8 0.9503
15296
ATXN3_Int9_Int 10_T 173 AACAGCACAGTTATCAGCAC 12.5 0.9753
16295
ATXN3_Int9_Intl 0_T29 AAAAAACCTTGCCCCAATAC 12.5 0.9867
26093
ATXN3_Int9_Int 10_T229 GATGTAGTAGAGTTTTGCTT 12.4 0.973
15400
ATXN3_Int9_Int 10_T97 ACATACCTTCTTGAGTGGAG 12.2 0.925
26023
ATXN3_Int9_Int 10_T 144 CCTCCACTTTGGTTGTTCTC 11.9 0.9707
16257
ATXN3_Int9_Int 10_T224 CTTGTAAGTGCTTGCTAAAT 11.9 0.9594
25605
ATXN3_Int9_IntlO_T32 TCACGAATCAAAGTAGTCAC 11.4 0.9919
15268
ATXN3_Int9_Int 10_T197 CTACTTTTGGTGGGATGGTC 11.4 0.9819
25838
ATXN3_Int9_IntlO_Tl 10 CTGTGCCATCAATATTATAG 11 0.8597
26092
ATXN3_Int9_Int 10_T230 TGATGTAGTAGAGTTTTGCT 10.7 0.97
25615
ATXN3_Int9_Int 10_T217 AATATGGAAGTGCTGATCTT 10.6 0.9863
15589
ATXN3_Int9_Int 10_T46 AGTACCACAAACCAACTTGC 9.9 0.99
15271
ATXN3_Int9_Int 10_T74 GTCAGGTGGCTACTTTTGGT 9.7 0.9803
25728
ATXN3_Int9_Int 10_T210 GGCCAATAATTGGGGAATCT 9.7 0.9645
26731
ATXN3_Int9_Intl 0_T21 CCTGATGCTAACAGGCTTAC 9.3 0.9417
15628
ATXN3_Int9_IntlO_T253 ATTAATGTCTTCTCTAGGTG 9.2 0.97
26175
ATXN3_Int9_Intl 0_T47 AGAGAGACAACGAACTACCT 9.1 0.9829 15727
ATXN3_Int9_Int 10_T57 ACCTTGTAGAGCTCATTAGT 9 0.9849
16285
ATXN3_Int9_Int 10_T 124 AAGAATGCCAATTGATCCTT 8.9 0.9909
16262
ATXN3_Int9_Int 10_T 188 GCCTGTTAGCATCAGGGGAA 8.8 0.9741
25702
ATXN3_Int9_Int 10_T 157 AGCATAAACTTATCATCATC 8.3 0.9812
26056
ATXN3_Int9_IntlO_T254 GTTGGTTTGTGGTACTGTGG 8.3 0.99
15582
ATXN3_Int9_IntlO_T87 ACTGCCCCAGTTGGGACAAT 8.2 0.9889
15708
ATXN3_Int9_IntlO_T248 CACCCTTCCCAAATTGACCC 8.2 0.9827
15421
ATXN3_Int9_Intl 0_T240 TATAATATTGATGGCACAGT 8 0.8477
26086
ATXN3_Int9_Int 10_T84 ATGCTTCTGTGAGTTAAGGC 8 0.96
26753
ATXN3_Int9_Intl 0_T226 AGTACATCCAAAGGATCAAT 7.7 0.9905
15571
ATXN3_Int9_IntlO_T51 CACCACAATTGCCTAGAGAT 7.5 0.9617
26747
ATXN3_Int9_Int 10_T45 GGAATTTTGCAGGGTTAATC 7.2 0.9913
25724
ATXN3_Int9_Int 10_T 141 GGTTCAGAAAGGCCAATAAT 6.6 0.9824
26718
ATXN3_Int9_Intl 0_T94 TAGCAAGCACTTACAAGGCT 6.6 0.9755
15415
ATXN3_Int9_IntlO_T233 CACCTCAGTGTCTTCACAGC 6.3 0.8168
25834
ATXN3_Int9_Int 10_T 169 TGAGGTGGAACATGTTCAAG 6.3 0.8445
16275
ATXN3_Int9_IntlO_Tl AGCGCATTCCCAAATAGACG 6.2 0.9952
25822
ATXN3_Int9_Int 10_T227 AGGTGAGATAGCAATTTTAG 5.8 0.8374
15753
ATXN3_Int9_Int 10_T 156 ACCTCCTTCCTACTTGGTGG 5.7 0.9675
26045
ATXN3_Int9_Int 10_T25 TCATTACCCATTGTCCCAAC 5.4 0.99
26733
ATXN3_Int9_Int 10_T 139 GCTTACAGGACTGCCTGAAG 5.4 0.9269
26717
ATXN3_Int9_Int 10_T72 TTAGCAAGCACTTACAAGGC 5.1 0.9424
25716
ATXN3_Int9_Intl 0_T206 CGGTTATCATGTAAATATGT 5 0.9891
15402
ATXN3_Int9_Int 10_T 184 TCACATACCTTCTTGAGTGG 4.6 0.918
15717
ATXN3_Int9_IntlO_T28 AGGTAGTTCGTTGTCTCTCT 4.6 0.9656 26099
ATXN3_Int9_Int 10_T92 ACAGATTTAGAGACTACTGC 4.5 0.9738
16270
ATXN3_Int9_Intl 0_T23 TAGCTGCTCATGGCCGCTTC 4.2 0.9149
26178
ATXN3_Int9_Intl 0_T38 ACTACCTAGGCTAAGTAATC 3.9 0.9839
26762
ATXN3_Int9_Int 10_T152 ATTTATCCAGTATTGGGGCA 3.9 0.9899
16263
ATXN3_Int9_Intl 0_T62 TGTAAGCCTGTTAGCATCAG 3.6 0.9756
15272
ATXN3_Int9_IntlO_Tl 12 TGTCAGGTGGCTACTTTTGG 3.5 0.9793
25853
ATXN3_Int9_Int 10_T 166 TTAAAAAGCAGAAACGGGTC 3.5 0.8964
26736
ATXN3_Int9_Int 10_T7 AATGAACTACCACGTCTATT 3.1 0.9952
16264
ATXN3_Int9_IntlO_Tl 19 CTGTAAGCCTGTTAGCATCA 3 0.9719
26032
ATXN3_Int9_Int 10_T 135 TGCTTCACTGCTGTTGCAAT 2.5 0.9607
15583
ATXN3_Int9_Int 10 T181 CACTGCCCCAGTTGGGACAA 2.2 0.99
26744
ATXN3_Int9_Int 10_T 136 GATTGGTAGAGGAATTTTGC 2.1 0.996
15592
ATXN3_Int9_Int 10_T218 TGAAGGTGCAGCTCTTAGCC 1.8 0.98
25625
ATXN3_Int9_Intl 0_T270 CAAAATAACCTATCATGGCC 1.5 0.9937
16265
ATXN3_Int9_Intl 0_T27 CCTGTAAGCCTGTTAGCATC 1.4 0.9715
25726
ATXN3_Int9_IntlO_T231 TTCAGAAAGGCCAATAATTG 1.3 0.9851
15720
ATXN3_Int9_Int 10_T 174 AATTCCTGATTACTTAGCCT 1 0.9862
15319
ATXN3_Int9_IntlO_T259 ATTGGCCTTTCTGAACCATT 0.7 0.9843
25727
ATXN3_Int9_IntlO_Tl 80 AGGCCAATAATTGGGGAATC 0.5 0.9887
15401
ATXN3_Int9_Int 10_T 129 CACATACCTTCTTGAGTGGA 0.3 0.9227
[000495] Note that the SEQ ID NOs represent the DNA sequence of the genomic target, while the gRNA or sgRNA spacer sequence will be the RNA version of the DNA sequence.
Example 8 - Testing of preferred guides in in in vitro transcribed (TVT) gRNA screen
[000496] To identify a large spectrum of pairs of gRNA s able to edit the cognate DNA target region, an in vitro transcribed (IVT) gRNA screen was conducted. The relevant genomic sequence was submitted for analysis using a gRNA design software. The resulting list of gRNAs was narrowed to a select list of gRNAs as described above based on uniqueness of sequence (only gRNAs without a perfect match somewhere else in the genome were screened) and minimal predicted off targets. This set of gRNAs was in vitro transcribed, and transfected using Lipofectamine MessengerMAX into HEK293T cells that constitutively express Cas9. Cells were harvested 48 hours post transfection, the genomic DNA was isolated, and cutting efficiency was evaluated using TIDE analysis. (Figures 2A-D; Figures 3A-B).
[000497] The gRNA or pairs of gRNA with significant activity can then be followed up in cultured cells to measure correction of the ATXN3 mutation. Off-target events can be followed again. A variety of cells can be transfected and the level of gene correction and possible off- target events measured. These experiments allow optimization of nuclease and donor design and delivery.
Example 9 - Testing of preferred guides in cells for off-target activity
[000498] The gRNAs having the best on-target activity from the IVT screen in the above example are tested for off-target activity using Hybrid capture assays, GUIDE Seq. and whole genome sequencing in addition to other methods.
Example 10 - Testing different approaches for HDR gene editing
[000499] After testing the gRNAs for both on-target activity and off-target activity, repeat expansion correction and whole gene correction strategies will be tested for HDR gene editing.
[000500] For the repeat expansion correction approach, donor DNA template will be provided as a short single-stranded oligonucleotide, a short double-stranded oligonucleotide (PAM sequence intact/PAM sequence mutated), a long single-stranded DNA molecule (PAM sequence intact/PAM sequence mutated) or a long double-stranded DNA molecule (PAM sequence intact/PAM sequence mutated). In addition, the donor DNA template will be delivered by AAV.
[000501] For the whole gene correction approach, a single-stranded or double-stranded DNA having homologous arms to the ATXN3 chromosomal region may include more than 40 nt of the first exon (the first coding exon) of the ATXN3 gene, the complete CDS of the ATXN3 gene and 3' UTR of the ATXN3 gene, and at least 40 nt of the following intron. The single-stranded or double-stranded DNA having homologous arms to the ATXN3 chromosomal region may include more than 80 nt of the first exon of the ATXN3 gene, the complete CDS of the ATXN3 gene and 3' UTR of the ATXN3 gene, and at least 80 nt of the following intron. The single-stranded or double-stranded DNA having homologous arms to the ATXN3 chromosomal region may include more than 100 nt of the first exon of the ATXN3 gene, the complete CDS of the ATXN3 gene and 3' UTR of the ATXN3 gene, and at least 100 nt of the following intron. The single-stranded or double-stranded DNA having homologous arms to the ATXN3 chromosomal region may include more than 150 nt of the first exon of the ATXN3 gene, the complete CDS of the ATXN3 gene and 3' UTR of the ATXN3 gene, and at least 150 nt of the following intron. The single- stranded or double-stranded DNA having homologous arms to the ATXN3 chromosomal region may include more than 300 nt of the first exon of the ATXN3 gene, the complete CDS of the ATXN3 gene and 3' UTR of the ATXN3 gene, and at least 300 nt of the following intron. The single-stranded or double-stranded DNA having homologous arms to the ATXN3 chromosomal region may include more than 400 nt of the first exon of the ATXN3 gene, the complete CDS of the ATXN3 gene and 3 ' UTR of the ATXN3 gene, and at least 400 nt of the following intron.
[000502] Alternatively, the DNA template will be delivered by a recombinant AAV particle such as those taught herein.
[000503] A knock-in of ATXN3 cDNA can be performed into any selected chromosomal location, including the ATXN3 gene locus or in a "safe-harbor" locus, i.e., AAVS1 (PPP1R12C), ALB, Angptl3, ApoC3, ASGR2, CCR5, FIX (F9), Gys2, HGD, Lp(a), Pcsk9, Serpinal, TF, and/or TTR. Assessment of efficiency of FIDR mediated knock-in of cDNA into the first exon can utilize cDNA knock-in into "safe harbor" sites such as: single-stranded or double-stranded DNA having homologous arms to one of the following regions, for example: exon 1-2 of AAVS1 (PPP1R12C), exon 1-2 of ALB, exon 1-2 of Angptl3, exon 1-2 of ApoC3, exon 1-2 of ASGR2, exon 1-2 of CCR5, exon 1-2 of FIX (F9), exon 1-2 of Gys2, exon 1-2 of HGD, exon 1- 2 of Lp(a), exon 1-2 of Pcsk9, exon 1-2 of Serpinal, exon 1-2 of TF, or exon 1-2 of TTR; 5'UTR correspondent to ATXN3 or alternative 5' UTR, complete CDS of ATXN3 and 3' UTR of ATXN3 or modified 3' UTR and at least 80 nt of the first intron, alternatively same DNA template sequence will be delivered by AAV.
Example 11 - Re-assessment of lead CRISPR-Cas9 DNA donor combinations
[000504] After testing the different strategies for gene editing, the lead CRISPR-Cas9/DNA donor combinations will be re-assessed in cells for efficiency of deletion, recombination, and off-target specificity. Cas9 mRNA or RNP will be formulated into lipid nanoparticles for delivery, sgRNAs will be formulated into nanoparticles or delivered as a recombinant AAV particle, and donor DNA will be formulated into nanoparticles or delivered as recombinant AAV particle. Example 12 - In vivo testing in relevant animal model
[000505] After the CRISPR-Cas9/DNA donor combinations have been re-assessed, the lead formulations will be tested in vivo in an animal model.
[000506] Culture in human cells allows direct testing on the human target and the background human genome, as described above.
[000507] Preclinical efficacy and safety evaluations can be observed through engraftment of modified mouse or human neurons in a mouse model. The modified cells can be observed in the months after engraftment. XI. Equivalents and Scope
[000508] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific examples in accordance with the invention described herein. The scope of the present disclosure is not intended to be limited to the above Description, but rather is as set forth in the appended claims.
[000509] Claims or descriptions that include "or" between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The present disclosure includes examples in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The present disclosure includes examples in which more than one, or the entire group members are present in, employed in, or otherwise relevant to a given product or process.
[000510] In addition, it is to be understood that any particular example of the present disclosure that falls within the prior art may be explicitly excluded from any one or more of the claims. Since such examples are deemed to be known to one of ordinary skill in the art, they may be excluded even if the exclusion is not set forth explicitly herein. Any particular example of the compositions of the present disclosure (e.g., any antibiotic, therapeutic or active ingredient; any method of production; any method of use; etc.) can be excluded from any one or more claims, for any reason, whether or not related to the existence of prior art.
[000511] It is to be understood that the words which have been used are words of description rather than limitation, and that changes may be made within the purview of the appended claims without departing from the true scope and spirit of the present disclosure in its broader aspects.
[000512] While the present invention has been described at some length and with some particularity with respect to the several described examples, it is not intended that it should be limited to any such particulars or examples or any particular example, but it is to be construed with references to the appended claims so as to provide the broadest possible interpretation of such claims in view of the prior art and, therefore, to effectively encompass the intended scope of the invention.
Note Regarding Illustrative Examples
[000513] While the present disclosure provides descriptions of various specific aspects for the purpose of illustrating various aspects of the present disclosure and/or its potential applications, it is understood that variations and modifications will occur to those skilled in the art.
Accordingly, the invention or inventions described herein should be understood to be at least as broad as they are claimed, and not as more narrowly defined by particular illustrative aspects provided herein.
[000514] Any patent, publication, or other disclosure material identified herein is incorporated by reference into this specification in its entirety unless otherwise indicated, but only to the extent that the incorporated material does not conflict with existing descriptions, definitions, statements, or other disclosure material expressly set forth in this specification. As such, and to the extent necessary, the express disclosure as set forth in this specification supersedes any conflicting material incorporated by reference. Any material, or portion thereof, that is said to be incorporated by reference into this specification, but which conflicts with existing definitions, statements, or other disclosure material set forth herein, is only incorporated to the extent that no conflict arises between that incorporated material and the existing disclosure material.
Applicants reserve the right to amend this specification to expressly recite any subject matter, or portion thereof, incorporated by reference herein.

Claims

1. A method for editing a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell by genome editing comprising: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
2. A method for editing a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell by genome editing comprising: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
3. An ex vivo method for treating a patient having an ATXN3 related condition or disorder comprising:
(a) creating a patient specific induced pluripotent stem cell (iPSC);
(b) editing within or near a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene or other DNA sequences that encode regulatory elements of the ATXN3 gene of the iPSC;
(c) differentiating the genome-edited iPSC into a neuron or glial cell of the Central Nervous System (CNS); and
(d) implanting said neuron or glial cell of the Central Nervous System (CNS) into the patient.
4. The method of claim 3, wherein the editing step comprises: introducing into the iPSC one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
5. The method of claim 3, wherein the editing step comprises: introducing into the iPSC one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
6. An ex vivo method for treating a patient having an ATXN3 related condition or disorder comprising:
(a) isolating a mesenchymal stem cell from the patient;
(b) editing within or near a Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene or other DNA sequences that encode regulatory elements of the ATXN3 gene of the mesenchymal stem cell;
(c) differentiating the genome-edited mesenchymal stem cell into a neuron or glial cell of the Central Nervous System (CNS); and
(d) implanting the neuron or glial cell of the Central Nervous System (CNS) into the patient.
7. The method of claim 6, wherein the editing step comprises: introducing into the
mesenchymal stem cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
8. The method of claim 6, wherein the editing step comprises: introducing into the
mesenchymal stem cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
9. An in vivo method for treating a patient with an ATXN3 related disorder comprising:
editing the Spinocerebellar Ataxia Type 3 Protein (ATXN3) gene in a cell of the patient.
10. The method of claim 9, wherein the editing step comprises: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene that results in permanent deletion of the expanded trinucleotide repeat or replacement of one or more nucleotide bases, or one or more exons and/or introns within or near the ATXN3 gene, thereby restoring the ATXN3 gene function.
11. The method of claim 9, wherein the editing step comprises: introducing into the cell one or more deoxyribonucleic acid (DNA) endonucleases to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs) within or near the ATXN3 gene or
ATXN3 regulatory elements that results in one or more permanent insertion, deletion or mutation of at least one nucleotide within or near the ATXN3 gene, thereby reducing or eliminating the expression or function of aberrant ATXN3 gene products.
12. The method of any one of claims 9-11, wherein the cell is a cell of the Central Nervous System (CNS).
13. The method of claim 12, wherein the cell of the Central Nervous System (CNS) is a neuron.
14. The method of claim 12, wherein the cell of the Central Nervous System (CNS) is a glial cell.
15. The method of any one of claims 12-14, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is delivered to the cell of the Central Nervous System (CNS) via any administration route selected from the group consisting of: intraparenchymal, intravenous, intra-arterial, intracerebroventricular, intracisternal, intrathecal, intracranial or intraperitoneal routes.
16. A method of altering the contiguous genomic sequence of an ATXN3 gene in a cell
comprising: contacting said cell with one or more deoxyribonucleic acid (DNA) endonuclease to effect one or more single-strand breaks (SSBs) or double-strand breaks (DSBs).
17. The method of claim 16, wherein the alteration of the contiguous genomic sequence occurs in one or more exons of the ATXN3 gene.
18. The method of claim 17, wherein the alteration of the contiguous genomic sequence occurs in exon 10 of the ATXN3 gene.
19. The method of any one of claims 1-18, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is selected from any of those sequences in SEQ ID NOs: 1-620 and variants having at least 90% homology to any of those sequences disclosed in SEQ ID NOs: 1-620.
20. The method of claim 19, wherein the one or more deoxyribonucleic acid (DNA)
endonuclease is one or more protein or polypeptide.
21. The method of claim 19, wherein the one or more deoxyribonucleic acid (DNA)
endonuclease is one or more polynucleotide encoding the one or more DNA
endonuclease.
22. The method of claim 21, wherein the one or more deoxyribonucleic acid (DNA)
endonuclease is one or more ribonucleic acid (RNA) encoding the one or more DNA endonuclease.
23. The method of claim 22, wherein the one or more ribonucleic acid (RNA) is one or more chemically modified RNA.
24. The method of claim 23, wherein the one or more ribonucleic acid (RNA) is chemically modified in the coding region.
25. The method of any one of claim 21-24, wherein the one or more polynucleotide or one or more ribonucleic acid (RNA) is codon optimized.
26. The method of any one of claims 1-25, wherein the method further comprises:
introducing one or more gRNA or one or more sgRNA.
27. The method of claim 26, wherein the one or more gRNA or one or more sgRNA comprises a spacer sequence that is complementary to a sequence within or near the expanded trinucleotide repeat in the ATXN3 gene.
28. The method of claim 26, wherein the one or more gRNA or one or more sgRNA
comprises a spacer sequence that is complementary to a DNA sequence within or near the ATXN3 gene.
29. The method of claim 26, wherein the one or more gRNA or one or more sgRNA
comprises a spacer sequence that is complementary to a sequence flanking the ATXN3 gene or other sequence that encodes a regulatory element of the ATXN3 gene.
30. The method of any one of claims 26-29, wherein said one or more gRNA or one or more sgRNA is chemically modified.
31. The method of any one of claims 26-30, wherein said one or more gRNA or one or more sgRNA is pre-complexed with the one or more deoxyribonucleic acid (DNA)
endonuclease.
32. The method of claim 31, wherein the pre-complexing involves a covalent attachment of said one or more gRNA or one or more sgRNA to the one or more deoxyribonucleic acid (DNA) endonuclease.
33. The method of any one of claims 19-32, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is formulated in a liposome or lipid nanoparticle.
34. The method of any one of claims 26-32, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is formulated in a liposome or a lipid nanoparticle which also comprises the one or more gRNA or one or more sgRNA.
35. The method of any one of claims 19, or 26-29, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is encoded in an AAV vector particle.
36. The method of any of the claims 26-29, wherein the one or more gRNA or one or more sgRNA is encoded in an AAV vector particle.
37. The method of any of the claims 26-29, wherein the one or more deoxyribonucleic acid (DNA) endonuclease is encoded in an AAV vector particle which also encodes the one or more gRNA or one or more sgRNA.
38. The method of any one of claims 35-37, wherein the AAV vector particle is selected from the group consisting of any of those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
39. The method of any of claims 1-38, wherein the method further comprises: introducing into the cell a donor template comprising at least a portion of the wild-type ATXN3 gene.
40. The method of claim 39, wherein the at least a portion of the wild-type ATXN3 gene comprises one or more sequences selected from the group consisting of: an ATXN3 exon, an ATXN3 intron, and a sequence comprising an exon:intron junction of ATXN3.
41. The method of any one of claims 39-40, wherein the donor template comprises
homologous arms to the genomic locus of the ATXN3 gene.
42. The method of any one of claims 39-41, wherein the donor template is either a single or double stranded polynucleotide.
43. The method of any one of claims 39-42, wherein the donor template is encoded in an AAV vector particle, where the AAV vector particle is selected from the group consisting of any of those disclosed in SEQ ID NOs: 4734-5302 and Table 2.
44. The method of any one of claims 39-42, wherein the one or more polynucleotide
encoding one or more deoxyribonucleic acid (DNA) endonuclease is formulated into a lipid nanoparticle, and the one or more gRNA or one or more sgRNA is delivered to the cell ex vivo by electroporation and the donor template is delivered to the cell by an adeno- associated virus (AAV) vector.
45. The method of any one of claims 39-42, wherein the one or more polynucleotide
encoding one or more deoxyribonucleic acid (DNA) endonuclease is formulated into a liposome or lipid nanoparticle which also comprises the one or more gRNA or one or more sgRNA and the donor template.
46. A single-molecule guide RNA comprising: at least a spacer sequence that is an RNA sequence corresponding to any of SEQ ID NOs: 5305-56902.
47. The single-molecule guide RNA of claim 46, wherein the single-molecule guide RNA further comprises a spacer extension region.
48. The single-molecule guide RNA of claim 46, wherein the single-molecule guide RNA further comprises a tracrRNA extension region.
49. The single-molecule guide RNA of any one of claim 46-48, wherein the single-molecule guide RNA is chemically modified.
50. The single-molecule guide RNA of any of claims 46-49 pre-complexed with a DNA
endonuclease.
51. The single-molecule guide RNA of claim 50, wherein the DNA endonuclease is a Cas9 or Cpfl endonuclease.
52. The single-molecule guide RNA of claim 51, wherein the Cas9 or Cpfl endonuclease is selected from the group consisting of: S. pyogenes Cas9, S. aureus Cas9, N. meningitides Cas9, S. thermophilus CRISPR1 Cas9, S. thermophilus CRISPR 3 Cas9, T. denticola Cas9, L. bacterium ND2006 Cpfl and Acidaminococcus sp. BV3L6 Cpfl, and variants having at least 90% homology to said endonucleases.
53. The single-molecule guide RNA of claim 52, wherein the Cas9 or Cpfl endonuclease comprises one or more nuclear localization signals (NLSs).
54. The single-molecule guide RNA of claim 53, wherein at least one NLS is at or within 50 amino acids of the amino-terminus of the Cas9 or Cpfl endonuclease and/or at least one NLS is at or within 50 amino acids of the carboxy-terminus of the Cas9 or Cpfl endonuclease.
55. A DNA encoding the single-molecule guide RNA of any one of claims 46-49.
PCT/IB2017/053941 2016-06-29 2017-06-29 Materials and methods for treatment of spinocerebellar ataxia 3 (sca3) and other related disorders WO2018002886A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662355936P 2016-06-29 2016-06-29
US62/355,936 2016-06-29
US201762461857P 2017-02-22 2017-02-22
US62/461,857 2017-02-22

Publications (1)

Publication Number Publication Date
WO2018002886A1 true WO2018002886A1 (en) 2018-01-04

Family

ID=59501490

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2017/053941 WO2018002886A1 (en) 2016-06-29 2017-06-29 Materials and methods for treatment of spinocerebellar ataxia 3 (sca3) and other related disorders

Country Status (1)

Country Link
WO (1) WO2018002886A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020172559A1 (en) * 2019-02-22 2020-08-27 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing atxn3 expression
CN113302302A (en) * 2019-01-09 2021-08-24 科英布拉大学 Double-stranded RNA and use thereof
CN113383077A (en) * 2018-11-19 2021-09-10 优尼科Ip有限公司 RNAi-induced ataxin-3 reduction for treatment of spinocerebellar ataxia type 3
US11434488B2 (en) 2018-05-09 2022-09-06 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ATXN3 expression
US11583548B2 (en) 2016-11-10 2023-02-21 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ATXN3 expression

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015089354A1 (en) * 2013-12-12 2015-06-18 The Broad Institute Inc. Compositions and methods of use of crispr-cas systems in nucleotide repeat disorders
WO2015173436A1 (en) * 2014-05-16 2015-11-19 Vrije Universiteit Brussel Genetic correction of myotonic dystrophy type 1

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015089354A1 (en) * 2013-12-12 2015-06-18 The Broad Institute Inc. Compositions and methods of use of crispr-cas systems in nucleotide repeat disorders
WO2015173436A1 (en) * 2014-05-16 2015-11-19 Vrije Universiteit Brussel Genetic correction of myotonic dystrophy type 1

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
MELVIN M. EVERS ET AL.: "Ataxin-3 protein modification as a treatment strategy for spinocerebellar ataxia type 3: removal of the CAG containing exon", NEUROBIOLOGY OF DISEASE, vol. 58, 1 October 2013 (2013-10-01), pages 49 - 56, XP055163564, ISSN: 0969-9961, DOI: 10.1016/j.nbd.2013.04.019 *
NAOKI TAJIRI ET AL.: "Autologous stem cell transplant with gene therapy for Friedreich Ataxia", MEDICAL HYPOTHESES, vol. 83, no. 3, 1 September 2014 (2014-09-01), US, pages 296 - 298, XP055408309, ISSN: 0306-9877, DOI: 10.1016/j.mehy.2014.05.022 *
RYAN M. GENGA ET AL.: "Controlling transcription in human pluripotent stem cells using CRISPR-effectors", METHODS, vol. 101, 1 May 2016 (2016-05-01), US, pages 36 - 42, XP055408213, ISSN: 1046-2023, DOI: 10.1016/j.ymeth.2015.10.014 *
XUEBING WU ET AL.: "Target specificity of the CRISPR-Cas9 system", QUANTITATIVE BIOLOGY, vol. 2, no. 2, 1 June 2014 (2014-06-01), pages 59 - 70, XP055176829, ISSN: 2095-4689, DOI: 10.1007/s40484-014-0030-x *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11583548B2 (en) 2016-11-10 2023-02-21 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ATXN3 expression
US20240082291A1 (en) * 2016-11-10 2024-03-14 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing atxn3 expression
US11434488B2 (en) 2018-05-09 2022-09-06 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ATXN3 expression
CN113383077A (en) * 2018-11-19 2021-09-10 优尼科Ip有限公司 RNAi-induced ataxin-3 reduction for treatment of spinocerebellar ataxia type 3
CN113302302A (en) * 2019-01-09 2021-08-24 科英布拉大学 Double-stranded RNA and use thereof
WO2020172559A1 (en) * 2019-02-22 2020-08-27 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing atxn3 expression

Similar Documents

Publication Publication Date Title
US11559588B2 (en) Materials and methods for treatment of Spinocerebellar Ataxia Type 1 (SCA1) and other Spinocerebellar Ataxia Type 1 Protein (ATXN1) gene related conditions or disorders
US11118177B2 (en) Materials and methods for treatment of Usher syndrome type 2A and/or non-syndromic autosomal recessive retinitis pigmentosa (ARRP)
EP3585900B1 (en) Materials and methods for treatment of spinocerebellar ataxia type 2 (sca2) and other spinocerebellar ataxia type 2 protein (atxn2) gene related conditions or disorders
US11407997B2 (en) Materials and methods for treatment of primary hyperoxaluria type 1 (PH1) and other alanine-glyoxylate aminotransferase (AGXT) gene related conditions or disorders
US20230279439A1 (en) Materials and methods for treatment of pain related disorders
US20200095579A1 (en) Materials and methods for treatment of merosin-deficient cogenital muscular dystrophy (mdcmd) and other laminin, alpha 2 (lama2) gene related conditions or disorders
EP3585807A1 (en) Materials and methods for treatment of early onset parkinson&#39;s disease (park1) and other synuclein, alpha (snca) gene related conditions or disorders
EP3585897A1 (en) Materials and methods for treatment of dystrophic epidermolysis bullosa (deb) and other collagen type vii alpha 1 chain (col7a1) gene related conditions or disorders
US20230330270A1 (en) Materials and methods for treatment of friedreich ataxia and other related disorders
US11801313B2 (en) Materials and methods for treatment of pain related disorders
WO2018002886A1 (en) Materials and methods for treatment of spinocerebellar ataxia 3 (sca3) and other related disorders

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17746186

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17746186

Country of ref document: EP

Kind code of ref document: A1