US20230159919A1 - Modified crispr rna and modified single crispr rna and uses thereof - Google Patents

Modified crispr rna and modified single crispr rna and uses thereof Download PDF

Info

Publication number
US20230159919A1
US20230159919A1 US17/850,456 US202217850456A US2023159919A1 US 20230159919 A1 US20230159919 A1 US 20230159919A1 US 202217850456 A US202217850456 A US 202217850456A US 2023159919 A1 US2023159919 A1 US 2023159919A1
Authority
US
United States
Prior art keywords
modified
crrna
compound
recognition portion
certain embodiments
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
US17/850,456
Inventor
Meghdad Rahdar
Thazha P. Prakash
Eric E. Swayze
C. Frank Bennett
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ionis Pharmaceuticals Inc
Original Assignee
Ionis Pharmaceuticals Inc
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 Ionis Pharmaceuticals Inc filed Critical Ionis Pharmaceuticals Inc
Priority to US17/850,456 priority Critical patent/US20230159919A1/en
Publication of US20230159919A1 publication Critical patent/US20230159919A1/en
Pending legal-status Critical Current

Links

Images

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
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. 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
    • 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
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • C12N15/902Stable introduction of foreign DNA into chromosome using homologous recombination
    • C12N15/907Stable introduction of foreign DNA into chromosome using homologous recombination in mammalian cells
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified C
    • C12N2310/33415-Methylcytosine
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/344Position-specific modifications, e.g. on every purine, at the 3'-end
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14311Parvovirus, e.g. minute virus of mice
    • C12N2750/14341Use of virus, viral particle or viral elements as a vector
    • C12N2750/14343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites

Definitions

  • CRISPR Cluster Regulatory Interspaced Short Palindromic Repeats
  • modified crRNA is stabilized at the 5′ end and/or the 3′. In certain embodiments, such stabilized crRNA is resistant to exonuclease and/or endonucleoase digestion. In certain embodiments, modified crRNA have improved affinity for target DNA relative to unmodified crRNA. In certain embodiments, modified crRNA have improved selectivity for target DNA relative to unmodified crRNA. In certain embodiments, modified crRNA have improved affinity for tracrRNA relative to unmodified crRNA. In certain embodiments, modified crRNA have improved cellular uptake relative to unmodified crRNA.
  • modified crRNA is shorter than unmodified crRNA.
  • modified crRNA is 40-50 linked nucleosides in length.
  • modified crRNA is 35-45 linked nucleosides in length.
  • modified crRNA is 30-40 linked nucleosides in length.
  • modified crRNA is 25-35 linked nucleosides in length.
  • modified crRNA is 20-30 linked nucleosides in length.
  • modified crRNA is 25-35 linked nucleosides in length.
  • modified crRNA is 20-30 linked nucleosides in length. In certain such embodiments, such shorter crRNA have improved uptake properties. In certain embodiments, modified crRNA are taken into cells without transfection reagents or electroporation. In certain such embodiments, the cells are in an animal. In certain embodiments, the animal expresses Cas9. In certain embodiments, the animal is previously or concomitantly treated with a means of expressing Cas9. In certain such embodiments, such treatment comprises administration of a vector for delivering Cas9. In certain such embodiments, such vector is a viral vector, for example adeno-associated virus (AAV). In certain such embodiments, the viral vector expresses a S. aureus derived Cas9 that fits into an AAV vector.
  • AAV adeno-associated virus
  • modified scrRNA for use as scrRNA in CRISPR systems.
  • modified scrRNA have improved stability relative to unmodified scrRNA.
  • modified scrRNA is stabilized at the 5′ end and/or the 3′.
  • stabilized scrRNA is resistant to exonuclease and/or endonucleoase digestion.
  • modified scrRNA have improved affinity for scrRNA target DNA relative to unmodified scrRNA.
  • modified scrRNA have improved selectivity for scrRNA target DNA relative to unmodified scrRNA.
  • modified scrRNA have improved affinity for a nuclease relative to unmodified scrRNA.
  • modified scrRNA have improved cellular uptake relative to unmodified scrRNA.
  • modified scrRNA is shorter than unmodified scrRNA.
  • modified scrRNA is 40-50 linked nucleosides in length.
  • modified scrRNA is 35-45 linked nucleosides in length.
  • modified scrRNA is 30-40 linked nucleosides in length.
  • modified scrRNA is 25-35 linked nucleosides in length.
  • modified scrRNA is 20-30 linked nucleosides in length.
  • modified scrRNA is 25-35 linked nucleosides in length.
  • modified scrRNA are taken into cells without transfection reagents or electroporation.
  • the cells are in an animal.
  • the animal expresses a nuclease that is recognized by the scrRNA (e.g., a Cpf1 nuclease).
  • the animal is previously or concomitantly treated with a means of expressing a nuclease that is recognized by the scrRNA (e.g., a Cpf1 nuclease).
  • such treatment comprises administration of a vector for delivering a nuclease that is recognized by the scrRNA (e.g., a Cpf1 nuclease).
  • a vector for delivering a nuclease that is recognized by the scrRNA e.g., a Cpf1 nuclease.
  • such vector is a viral vector, for example adeno-associated virus (AAV).
  • AAV adeno-associated virus
  • the CRISPR system is inhibited after the target gene is edited or the scrRNA target gene is altered.
  • the modified crRNA or modified scrRNA inside a cell is degraded after the target gene or scrRNA target gene has been edited or altered.
  • the nuclease e.g., Cas9 or a Cpf1 nuclease
  • off-target effects of the CRISPR system are decreased relative to a CRISPR system in which all of the components necessary for nuclease activity continue to be expressed indefinitely, e.g. by a viral vector.
  • degradation of the modified crRNA or modified scrRNA is facilitated by hybridization to an oligonucleotide complementary to the crRNA or scrRNA.
  • degradation of the modified crRNA or modified scrRNA is facilitated by nucleases present in the cell.
  • the CRISPR system is inhibited after the target gene is edited via degradation of a tracrRNA inside the cell.
  • degradation of the tracrRNA is facilitated by hybridization to an oligonucleotide complementary to the tracrRNA.
  • degradation of the tracrRNA is facilitated by nucleases present in the cell.
  • the CRISPR system is inhibited after the target gene is edited or the scrRNA target gene is altered via inhibition of the expression of a nuclease (e.g., Cas9 or a Cpf1 nuclease).
  • a nuclease e.g., Cas9 or a Cpf1 nuclease
  • the nuclease gene is edited or altered by a modified crRNA or a modified scrRNA.
  • the nuclease transcript is degraded following hybridization of the nuclease transcript to an oligonucleotide complementary to the nuclease transcript.
  • FIG. 1 is a gel illustrating the extent of gene editing of hLDLR.
  • FIG. 2 is a gel illustrating the extent of gene editing of hVEGFA.
  • FIG. 3 is a gel illustrating the extent of gene editing of hVEGFA using crRNAs, including shortened modified crRNAs.
  • FIGS. 4 a and 4 b are gels that show the effect of truncated scrRNAs comprising a scrRNA target recognition portion that is complementary to DNA (cytosine-5)-methyltransferase 1 (DNMT1) on alteration of the DNMT1 gene.
  • FIGS. 4 a and 4 b show that multiple truncated scrRNAs, including scrRNA containing only 36 nucleosides, altered the DNMT1 gene.
  • FIG. 5 is a gel that shows the extent of activity of truncated tracrRNAs designed and synthesized to edit mouse Proprotein Convertase Subtilisin/Kexin Type 9 (Pcsk9).
  • FIG. 6 is a gel that shows the DNA cutting activity of conjugated and unconjugated modified crRNA targeted to Pcsk9.
  • FIG. 7 is a gel that shows that a modified crRNA disrupted the Pcsk9 gene with similar potency to a sgRNA positive control in hepatocytes ex vivo.
  • 2′-deoxynucleoside means a nucleoside comprising 2′-H(H) furanosyl sugar moiety, as found in naturally occurring deoxyribonucleic acids (DNA).
  • a 2′-deoxynucleoside may comprise a modified nucleobase or may comprise an RNA nucleobase (e.g., uracil).
  • 2′-substituted nucleoside or “2-modified nucleoside” means a nucleoside comprising a 2′-substituted or 2′-modified sugar moiety.
  • 2′-substituted or “2-modified” in reference to a sugar moiety means a furanosyl sugar moiety comprising a 2′-substituent group other than H or OH.
  • 3′-stabilized in reference to a modified oligonucleotide means a modified oligonucleotide comprising a modification or modifications at the 3′-terminus that increase the stability of the oligonucleotide in cells or in an animal relative to a corresponding oligonucleotide that does not comprise the modification or modifications at the 3′-terminus.
  • 5′-stabilized in reference to a modified oligonucleotide means a modified oligonucleotide comprising a modification or modifications at the 5′-terminus that increase the stability of the oligonucleotide in cells or in an animal relative to a corresponding oligonucleotide that does not comprise the modification or modifications at the 5′-terminus.
  • bicyclic nucleoside or “BNA” means a nucleoside comprising a bicyclic sugar moiety.
  • bicyclic sugar or “bicyclic sugar moiety” means a modified sugar moiety comprising two rings, wherein the second ring is formed via a bridge connecting two of the atoms in the first ring thereby forming a bicyclic structure.
  • the first ring of the bicyclic sugar moiety is a furanosyl moiety.
  • the bicyclic sugar moiety does not comprise a furanosyl moiety.
  • Cas9 means a nuclease that recognizes and/or cleaves target DNA when in a complex with crRNA and tracrRNA.
  • Cas9 is derived from S. pyogenes .
  • Cas9 is derived from S. aureus.
  • cell-targeting moiety means a conjugate group or portion of a conjugate group that is capable of binding to a particular cell type or particular cell types.
  • nucleobase sequence of such oligonucleotide or one or more regions thereof matches the nucleobase sequence of another oligonucleotide or nucleic acid or one or more regions thereof when the two nucleobase sequences are aligned in opposing directions.
  • Nucleobase matches or complementary nucleobases, as described herein, are limited to adenine (A) and thymine (T), adenine (A) and uracil (U), cytosine (C) and guanine (G), and 5-methyl cytosine ( m C) and guanine (G) unless otherwise specified.
  • oligonucleotides and/or nucleic acids need not have nucleobase complementarity at each nucleoside. Rather, some mismatches are tolerated.
  • “fully complementary” or “100% complementary” in reference to oligonucleotides means that such oligonucleotides are complementary to another oligonucleotide or nucleic acid at each nucleoside. In such embodiments, mismatches are not tolerated.
  • conjugate group means a group of atoms that is directly or indirectly attached to a parent compound, e.g., an oligonucleotide.
  • conjugate linker means a group of atoms that connects a conjugate group to a parent compound, e.g., an oligonucleotide.
  • oligonucleotide refers to nucleosides, nucleobases, sugar moieties, or internucleoside linkages that are immediately adjacent to each other.
  • contiguous nucleobases means nucleobases that are immediately adjacent to each other
  • crRNA means an oligonucleotide or portion of an oligonucleotide that comprises a DNA recognition portion and a tracrRNA recognition portion.
  • DNA recognition portion is nucleobase sequence that is complementary to a DNA target.
  • tracrRNA recognition portion is a nucleobase sequence that is bound to or is capable of binding to tracrRNA. The tracRNA recognition portion of crRNA may bind to tracrRNA via hybridization or covalent attachment.
  • oligonucleotide in which each sugar moiety is modified.
  • Uniformly modified in reference to an oligonucleotide means a fully modified oligonucleotide in which each at least one modification of each sugar moiety is the same.
  • nucleosides of a uniformly modified oligonucleotide can each have a 2′-MOE modification but different nucleobase modifications, and the internucleoside linkages may be different.
  • gene editing means any process mediated by a Cas9/crRNA/tracrRNA or Cas9/sgRNA complex, including but not limited to gene knock-down, gene knock-out, gene disruption, deletion, insertion, and gene activation.
  • gene alteration means any process mediated by a nuclease/scrRNA containing complex, including but not limited to gene knock-down, gene disruption, deletion, insertion, and gene activation.
  • gRNA comprises both a crRNA and a tracrRNA.
  • the crRNA and tracrRNA of a gRNA are distinct molecules.
  • the crRNA and tracrRNA of a gRNA are portions of one oligonucleotide, wherein the oligonucleotide is referred to as a “sgRNA”.
  • hybridization means the pairing or annealing of complementary oligonucleotides and/or nucleic acids. While not limited to a particular mechanism, the most common mechanism of hybridization involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleobases.
  • “increases”, when used in reference to an effect mediated by a modified oligonucleotide, means that the effect is greater in the presence of the oligonucleotide containing a certain modification than the effect is in the presence of a corresponding oligonucleotide that does not contain the certain modification.
  • internucleoside linkage means a group that forms a covalent linkage between adjacent nucleosides in an oligonucleotide.
  • modified internucleoside linkage means any internucleoside linkage other than a naturally occurring, phosphate internucleoside linkage. Naturally occurring, non-phosphate linkages are referred to herein as modified internucleoside linkages.
  • Phosphorothioate linkage means a linkage between nucleosides wherein the phosphodiester bond of a phosphate linkage is modified by replacing one of the non-bridging oxygen atoms with a sulfur atom.
  • a phosphorothioate linkage is a modified internucleoside linkage.
  • linearly modified sugar or “linearly modified sugar moiety” means a modified sugar moiety that comprises an acyclic or non-bridging modification. Such linear modifications are distinct from bicyclic sugar modifications.
  • linked nucleosides are nucleosides that are connected in a continuous sequence (i.e. no additional nucleosides are present between those that are linked). Linked nucleosides may or may not be linked by internucleoside linkages.
  • mismatch or means a nucleobase of a first oligonucleotide that is not capable of pairing with the corresponding nucleobase of a second oligonucleotide or target nucleic acid when the first and second oligomeric compound are aligned.
  • MOE means methoxyethyl.
  • 2′-MOE means a —OCH 2 CH 2 OCH 3 group at the 2′ position of a furanosyl ring.
  • motif means the pattern of unmodified and/or modified sugar moieties, nucleobases, and/or internucleoside linkages, in an oligonucleotide.
  • nucleobase means a heterocyclic moiety capable of pairing with a second, different nucleobase.
  • nucleobase sequence means the order of contiguous nucleobases independent of any sugar or internucleoside linkage modification.
  • modified nucleobase means a nucleobase other than adenine (A), thymine (T), cytosine (C), uracil (U), and guanine (G), herein defined as the five, unmodified nucleobases.
  • a universal base is a nucleobase that can pair with any one of the five unmodified nucleobases.
  • nucleoside means a compound comprising a nucleobase and a sugar moiety.
  • the nucleobase and sugar moiety are each, independently, unmodified or modified.
  • modified nucleoside means a nucleoside comprising a modified nucleobase and/or a modified sugar moiety. Modified nucleosides include abasic nucleosides.
  • oligonucleotide means a strand of linked nucleosides connected via internucleoside linkages, wherein each nucleoside and internucleoside linkage may be modified or unmodified. Unless otherwise indicated, oligonucleotides consist of 8-50 linked nucleosides.
  • modified oligonucleotide means an oligonucleotide, wherein at least one nucleoside or internucleoside linkage is modified.
  • unmodified oligonucleotide means an oligonucleotide that does not comprise any nucleoside modifications or internucleoside modifications.
  • pharmaceutically acceptable carrier or diluent means any substance suitable for use in administering to an animal. Certain such carriers enable pharmaceutical compositions to be formulated as, for example, tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspension and lozenges for the oral ingestion by a subject.
  • pharmaceutically acceptable salts means physiologically and pharmaceutically acceptable salts of compounds, such as oligomeric compounds, i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto.
  • a pharmaceutical composition means a mixture of substances suitable for administering to a subject.
  • a pharmaceutical composition may comprise an crRNA compound and a sterile aqueous solution.
  • a pharmaceutical composition shows activity in free uptake assay in certain cell lines.
  • phosphorus moiety means a group of atoms comprising a phosphorus atom.
  • a phosphorus moiety comprises a mono-, di-, or tri-phosphate, or phosphorothioate.
  • prodrug means a therapeutic agent in an inactive form that is converted to an active form within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or physiologic conditions.
  • scrRNA or “single crRNA” means an oligonucleotide that comprises a scrRNA target recognition portion and a nuclease recognition portion and does not comprise a tracrRNA recognition portion or a tracrRNA.
  • scrRNAs comprise a self-complementary region.
  • the nuclease recognition portion partially or completely overlaps with the self-complementary region.
  • scrRNA target recognition portion is a portion of an oligonucleotide with a nucleobase sequence that is complementary to a scrRNA DNA target.
  • nuclease recognition portion is a portion of an oligonucleotide that can bind to, associate with, or contribute to the binding to or association with a nuclease that is not a Cas9 nuclease.
  • the nuclease recognition portion of an oligonucleotide binds to or associates with a Cpf1 nuclease.
  • self-complementary in reference to an oligonucleotide means an oligonucleotide that is at least partially complementary to itself.
  • a self-complementary oligonucleotide forms a hairpin when a portion of the self-complementary oligonucleotide hybridizes to itself.
  • sugar moiety means a group of atoms that can link a nucleobase to another group, such as an internucleoside linkage, conjugate group, or terminal group.
  • a sugar moiety is attached to a nucleobase to form a nucleoside.
  • unmodified sugar moiety means a 2′-OH(H) furanosyl moiety, as found in RNA, or a 2′-H(H) moiety, as found in DNA. Unmodified sugar moieties have one hydrogen at each of the 1′, 3′, and 4′ positions, an oxygen at the 3′ position, and two hydrogens at the 5′ position.
  • modified sugar moiety or “modified sugar” means a sugar surrogate or a furanosyl moiety comprising a non-hydrogen substituent in place of at least one hydrogen of an unmodified sugar moiety.
  • a modified sugar moiety is a 2′-substituted sugar moiety.
  • modified sugar moieties include bicyclic sugars and linearly modified sugars.
  • sugar surrogate means a modified sugar moiety having other than a furanosyl moiety that can link a nucleobase to another group, such as an internucleoside linkage, conjugate group, or terminal group.
  • Modified nucleosides comprising sugar surrogates can be incorporated into one or more positions within an oligonucleotide. In certain embodiments, such oligonucleotides are capable of hybridizing to complementary oligomeric compounds or nucleic acids.
  • target nucleic acid means a nucleic acid that a crRNA is designed to affect.
  • scrRNA target nucleic acid scrRNA target DNA
  • scrRNA target gene scrRNA nucleic acid target
  • off-target gene is a gene that a crRNA or a scrRNA is not designed to affect. In certain embodiments, the editing or alteration of an off-target gene is deleterious.
  • terminal group means a chemical group or group of atoms that is covalently linked to a terminus of an oligonucleotide.
  • tracrRNA means an oligonucleotide or portion of an oligonucleotide that can non-covalently bind to a Cas9 protein and that can bind to a crRNA via hybridization or covalent attachment.
  • the present invention provides modified oligonucleotides for use in CRISPR.
  • CRISPR employs CRSPR RNA (crRNA), which hybridizes to target DNA and also hybridizes to trans-activating RNA (tracrRNA), which in turn recruits a nuclease, cas9, which cleaves the target DNA.
  • crRNA CRSPR RNA
  • tracrRNA trans-activating RNA
  • the crRNA in such systems has two functions: (1) recognition and hybridization to the target DNA and (2) recognition and hybridization to the tracrRNA.
  • the crRNA has two portions which correspond to these two functions: a DNA recognition portion and a tracrRNA recognition portion.
  • the present invention provides modified oligonucleotides that may be used in crRNA. Such modified oligonucleotides may have modifications in the DNA recognition portion and/or tracrRNA recognition portion.
  • the tracrRNA recognition portion of the crRNA comprises a portion of the direct repeat sequence from a bacterial species that has a Type II CRISPR system. In certain such embodiments, the tracrRNA recognition portion of the crRNA comprises a sequence selected from the table below. In certain embodiments, the tracrRNA recognition portion of the crRNA comprises the first 12 nucleobases of a sequence selected from the table below. In certain embodiments, the tracrRNA recognition portion of the crRNA comprises the first 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 nucleobases of a sequence selected from the table below. In certain embodiments, the sequence of the tracrRNA recognition portion of the crRNA consists of the first 12 nucleobases of a sequence selected from the table below. In certain embodiments, the sequence of the tracrRNA recognition portion of the crRNA consists of the first 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 nucleobases of a sequence selected from the table below.
  • the crRNA and tracrRNA are joined to one another to form a single molecule referred to as a single guide RNA (sgRNA).
  • sgRNA single guide RNA
  • the present invention provides modified oligonucleotides for use in sgRNA.
  • the present invention provides modified oligonucleotides for use in a CRISPR system that employs scrRNA, which hybridizes to a scrRNA target DNA and participates in recruitment of a nuclease other than Cas9.
  • the nuclease is a Cpf1 nuclease or a variant thereof.
  • the nuclease e.g., the Cpf1 nuclease
  • the scrRNA in such systems has two functions: (1) recognition and hybridization to the scrRNA target DNA and (2) recognition and recruitment of the nuclease.
  • the scrRNA has two portions which correspond to these two functions: a scrRNA target recognition portion and a nuclease recognition portion.
  • the present invention provides modified oligonucleotides that may be used in scrRNA. Such modified oligonucleotides may have modifications in the scrRNA target recognition portion and/or nuclease recognition portion.
  • the nuclease recognition portion is 5′ to the scrRNA target recognition portion. In certain embodiments, the nuclease recognition portion is 3′ to the scrRNA target recognition portion.
  • the nuclease recognition portion of the scrRNA comprises a portion of the direct repeat sequence from a bacterial organism that has a Cpf1 nuclease or a Cpf1 ortholog. In certain such embodiments, the nuclease recognition portion of the scrRNA comprises a sequence selected from the table below. In certain embodiments, the nuclease recognition portion of the scrRNA comprises 12 nucleobases of a sequence selected from the table below. In certain embodiments, the tracrRNA recognition portion of the crRNA comprises 13, 14, 15, 16, 17, 18, or 19 nucleobases of a sequence selected from the table below.
  • the sequence of the nuclease recognition portion of the scrRNA consists of 12 nucleobases of a sequence selected from the table below. In certain embodiments, the sequence of the nuclease recognition portion of the scrRNA consists of 13, 14, 15, 16, 17, 18, 19, 20, or 21 nucleobases of a sequence selected from the table below. In certain embodiments, the nuclease recognition portion of the scrRNA comprises the sequence UCUACU and GUAGAU.
  • modified crRNA comprise a modified oligonucleotide. In certain embodiments, modified crRNA consist of a modified oligonucleotide. Modified oligonucleotides described herein are suitable for use as crRNA.
  • modified oligonucleotides have one or more asymmetric center and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), as a or 13 such as for sugar anomers, or as (D) or (L) such as for amino acids etc.
  • R absolute stereochemistry
  • S a or 13
  • D sugar anomers
  • L such as for amino acids etc.
  • Included in the modified oligonucleotides provided herein are all such possible isomers, including their racemic and optically pure forms, unless specified otherwise. Likewise, all cis- and trans-isomers and tautomeric forms are also included.
  • such modified oligonucleotides may contain any combination of the modified sugar moieties, modified nucleobases, modified internucleoside linkages, motifs, and/or lengths described herein.
  • modified scrRNA comprise a modified oligonucleotide.
  • modified scrRNA consist of a modified oligonucleotide. Modified oligonucleotides described herein are suitable for use as scrRNA.
  • modified oligonucleotides have one or more asymmetric center and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), as a or 13 such as for sugar anomers, or as (D) or (L) such as for amino acids etc.
  • R absolute stereochemistry
  • S a or 13
  • D sugar anomers
  • L such as for amino acids etc.
  • Included in the modified oligonucleotides provided herein are all such possible isomers, including their racemic and optically pure forms, unless specified otherwise. Likewise, all cis- and trans-isomers and tautomeric forms are also included.
  • such modified oligonucleotides may contain any combination of the modified sugar moieties, modified nucleobases, modified internucleoside linkages, motifs, and/or lengths described herein.
  • methods comprising contacting a cell with a compound comprising a modified crRNA are in vitro methods. In certain embodiments, methods comprising contacting a cell with a compound comprising a modified crRNA are ex vivo methods. In certain embodiments, methods comprising contacting a cell with a compound comprising a modified crRNA are in vivo methods.
  • Cas9 variants both naturally occurring and genetically engineered, can be used in the methods of the present invention.
  • Such Cas9 variants include but are not limited to inactive Cas9 mutants that are used in applications that do not require target nucleic acid cleavage, such as gene activation, and truncated Cas9 variants that are suitable for expression in certain vectors, such as AAV vectors.
  • methods comprising contacting a cell with a compound comprising a modified crRNA further comprise contacting the cell with a second compound to inhibit (or turn off) the CRISPR system after the target gene is edited.
  • gene editing methods comprising contacting a cell with a compound comprising a modified crRNA produce fewer and/or less deleterious off-target effects than gene editing methods that use of an unmodified crRNA in place of the modified crRNAs of the invention.
  • methods comprising contacting a cell with a compound comprising a modified scrRNA are in vitro methods. In certain embodiments, methods comprising contacting a cell with a compound comprising a modified scrRNA are ex vivo methods. In certain embodiments, methods comprising contacting a cell with a compound comprising a modified scrRNA are in vivo methods.
  • nuclease variants both naturally occurring and genetically engineered, can be used in the methods of the present invention.
  • Such nuclease variants include but are not limited to inactive nuclease mutants that are used in applications that do not require scrRNA target nucleic acid cleavage, such as gene activation, and truncated nuclease variants that are suitable for expression in certain vectors, such as AAV vectors.
  • methods comprising contacting a cell with a compound comprising a modified scrRNA further comprise contacting the cell with a second compound to inhibit (or turn off) the CRISPR system after the scrRNA target gene is altered.
  • gene altering methods comprising contacting a cell with a compound comprising a modified scrRNA produce fewer and/or less deleterious off-target effects than gene altering methods that use an unmodified scrRNA in place of the modified scrRNAs of the invention.
  • modified nucleosides incorporate modified nucleosides.
  • modified nucleosides without limitation, are suitable for such incorporation into modified oligonucleotides for use as crRNA or scrRNA.
  • modified oligonucleotides comprise at least one modified nucleoside.
  • modified nucleosides comprise a modified sugar moiety or a modified nucleobase or both a modified sugar moiety and a modified nucleobase.
  • modified oligonucleotides such as modified crRNAs or modified scrRNAs, comprise one or more modified nucleosides comprising a modified sugar moiety.
  • modified oligonucleotides comprising one or more sugar-modified nucleosides may have desirable properties, such as enhanced nuclease stability or increased binding affinity with a target nucleic acid relative to oligonucleotides lacking such sugar-modified nucleosides.
  • modified sugar moieties are linearly modified sugar moieties.
  • modified sugar moieties are bicyclic or tricyclic sugar moieties.
  • modified sugar moieties are sugar surrogates. Such sugar surrogates may comprise one or more substitutions corresponding to those of substituted sugar moieties.
  • modified sugar moieties are linearly modified sugar moieties comprising a furanosyl ring with one or more acyclic substituent, including but not limited to substituents at the 2′ and/or 5′ positions.
  • 2′-substituent groups suitable for linearly modified sugar moieties include but are not limited to: 2′-F, 2′-OCH 3 (“OMe” or “O-methyl”), and 2′-O(CH 2 ) 2 OCH 3 (“MOE”).
  • 2′-substituent groups are selected from among: halo, allyl, amino, azido, SH, CN, OCN, CF 3 , OCF 3 , O—C 1 -C 10 alkoxy, O—C 1 -C 10 substituted alkoxy, O—C 1 -C 10 alkyl, O—C 1 -C 10 substituted alkyl, S-alkyl, N(R m )-alkyl, O-alkenyl, S-alkenyl, N(R m )-alkenyl, O-alkynyl, S-alkynyl, N(R m )-alkynyl, O-alkylenyl-O-alkyl, alkynyl, alkaryl, aralkyl, O-alkaryl, O-aralkyl, O(CH 2 ) 2 SCH 3 , O(CH 2 ) 2 ON(R m )(R n ) or
  • 2′-substituent groups can be further substituted with one or more substituent groups independently selected from among: hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro (NO 2 ), thiol, thioalkoxy, thioalkyl, halogen, alkyl, aryl, alkenyl and alkynyl.
  • substituent groups independently selected from among: hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro (NO 2 ), thiol, thioalkoxy, thioalkyl, halogen, alkyl, aryl, alkenyl and alkynyl.
  • substituent groups independently selected from among: hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro (NO 2 ), thiol, thioalkoxy, thioalkyl, halogen, alkyl, aryl, alkeny
  • linearly modified sugars comprise more than one non-bridging sugar substituent, for example, 2′-F-5′-methyl sugar moieties (see, e.g., PCT International Application WO 2008/101157, for additional 2′, 5′-bis substituted sugar moieties and nucleosides).
  • a 2′-substituted nucleoside or 2′-linearly modified nucleoside comprises a sugar moiety comprising a linear 2′-substituent group selected from: F, NH 2 , N 3 , OCF 3 , OCH 3 , O(CH 2 ) 3 NH 2 , CH 2 CH ⁇ CH 2 , OCH 2 CH ⁇ CH 2 , OCH 2 CH 2 OCH 3 , O(CH 2 ) 2 SCH 3 , O(CH 2 ) 2 ON(R m )(R n ), O(CH 2 ) 2 O(CH 2 ) 2 N(CH 3 ) 2 , and N-substituted acetamide (OCH 2 C( ⁇ O)—N(R m )(R n )), where each R m and R n is, independently, H, an amino protecting group, or substituted or unsubstituted C 1 -C 10 alkyl.
  • a 2′-substituted nucleoside or 2′-linearly modified nucleoside comprises a sugar moiety comprising a linear 2′-substituent group selected from: F, OCF 3 , OCH 3 , OCH 2 CH 2 OCH 3 , O(CH 2 ) 2 SCH 3 , O(CH 2 ) 2 ON(CH 3 ) 2 , O(CH 2 ) 2 O(CH 2 ) 2 N(CH 3 ) 2 , and OCH 2 C( ⁇ O)—N(H)CH 3 (“NMA”).
  • NMA OCH 2 C( ⁇ O)—N(H)CH 3
  • a 2′-substituted nucleoside or 2′-linearly modified nucleoside comprises a sugar moiety comprising a linear 2′-substituent group selected from: F, OCH 3 , and OCH 2 CH 2 OCH 3 .
  • Nucleosides comprising modified sugar moieties are referred to by the position(s) of the substitution(s) on the sugar moiety of the nucleoside.
  • nucleosides comprising 2′-substituted or 2-modified sugar moieties are referred to as 2′-substituted nucleosides or 2-modified nucleosides.
  • modified sugar moieties comprise a bridging sugar substituent that forms a second ring resulting in a bicyclic sugar moiety.
  • the bicyclic sugar moiety comprises a bridge between the 4′ and the 2′ furanose ring atoms.
  • 4′ to 2′ bridging sugar substituents include but are not limited to: 4′-CH 2 -2′, 4′-(CH 2 ) 2 -2′, 4′-(CH 2 ) 3 -2′, 4′-CH 2 —O-2′ (“LNA”), 4′-CH 2 —S-2′, 4′-(CH 2 ) 2 —O-2′ (“ENA”), 4′-CH(CH 3 )—O-2′ (referred to as “constrained ethyl” or “cEt” when in the S configuration), 4′-CH 2 —O—CH 2 -2′, 4′-CH 2 —N(R)-2′, 4′-CH(CH 2 OCH 3 )—O-2′ (“constrained MOE” or “cMOE”) and analogs thereof (see, e.g., U.S.
  • such 4′ to 2′ bridges independently comprise from 1 to 4 linked groups independently selected from: —[C(R a )(R b )] n —, —[C(R a )(R b )] n —O—, —C(R a ) ⁇ C(R b )—, —C(R a ) ⁇ N—, —C( ⁇ NR a )—, —C( ⁇ O)—, —C( ⁇ S)—, —O—, —Si(R a ) 2 —, —S( ⁇ O) x —, and —N(R a )—;
  • x 0, 1, or 2;
  • n 1, 2, 3, or 4;
  • each R a and R b is, independently, H, a protecting group, hydroxyl, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl, C 2 -C 12 alkenyl, substituted C 2 -C 12 alkenyl, C 2 -C 12 alkynyl, substituted C 2 -C 12 alkynyl, C 5 -C 20 aryl, substituted C 5 -C 20 aryl, heterocycle radical, substituted heterocycle radical, heteroaryl, substituted heteroaryl, C 5 -C 7 alicyclic radical, substituted C 5 -C 7 alicyclic radical, halogen, OJ 1 , NJ 1 J 2 , SJ 1 , N 3 , COOJ 1 , acyl (C( ⁇ O)—H), substituted acyl, CN, sulfonyl (S( ⁇ O) 2 -J 1 ), or sulfoxyl (S( ⁇ O)-J 1 ); and
  • each J 1 and J 2 is, independently, H, C 1 -C 12 alkyl, substituted C 1 -C 12 alkyl, C 2 -C 12 alkenyl, substituted C 2 -C 12 alkenyl, C 2 -C 12 alkynyl, substituted C 2 -C 12 alkynyl, C 5 -C 20 aryl, substituted C 5 -C 20 aryl, acyl (C( ⁇ O)—H), substituted acyl, a heterocycle radical, a substituted heterocycle radical, C 1 -C 12 aminoalkyl, substituted C 1 -C 12 aminoalkyl, or a protecting group.
  • bicyclic sugar moieties and nucleosides incorporating such bicyclic sugar moieties are further defined by isomeric configuration.
  • an LNA nucleoside (described above) may be in the ⁇ -L configuration or in the ⁇ -D configuration.
  • bicyclic nucleosides include both isomeric configurations.
  • positions of specific bicyclic nucleosides e.g., LNA or cEt
  • they are in the ⁇ -D configuration, unless otherwise specified.
  • modified sugar moieties comprise one or more non-bridging sugar substituent and one or more bridging sugar substituent (e.g., 5′-substituted and 4′-2′ bridged sugars).
  • bridging sugar substituent e.g., 5′-substituted and 4′-2′ bridged sugars.
  • WO 2007/134181 wherein LNA nucleosides are further substituted with, for example, a 5′-methyl or a 5′-vinyl group, and see, e.g., U.S. Pat. Nos. 7,547,684; 7,750,131; 8,030,467; 8,268,980; 7,666,854; and 8,088,746).
  • modified sugar moieties are sugar surrogates.
  • the oxygen atom of the sugar moiety is replaced, e.g., with a sulfur, carbon or nitrogen atom.
  • such modified sugar moieties also comprise bridging and/or non-bridging substituents as described above.
  • certain sugar surrogates comprise a 4′-sulfur atom and a substitution at the 2′-position (see, e.g., US2005/0130923) and/or the 5′ position.
  • sugar surrogates comprise rings having other than 5 atoms.
  • a sugar surrogate comprises a six-membered tetrahydropyran (“THP”).
  • TTP tetrahydropyrans
  • Such tetrahydropyrans may be further modified or substituted.
  • Nucleosides comprising such modified tetrahydropyrans include but are not limited to hexitol nucleic acid (“HNA”), anitol nucleic acid (“ANA”), manitol nucleic acid (“MNA”) (see Leumann, C J. Bioorg . & Med. Chem. 2002, 10, 841-854), fluoro HNA:
  • F-HNA see e.g., U.S. Pat. Nos. 8,088,904; 8,440,803; and 8,796,437, F-HNA can also be referred to as a F-THP or 3′-fluoro tetrahydropyran), and nucleosides comprising additional modified THP compounds having the formula:
  • Bx is a nucleobase moiety
  • T 3 and T 4 are each, independently, an internucleoside linking group linking the modified THP nucleoside to the remainder of an oligonucleotide or one of T 3 and T 4 is an internucleoside linking group linking the modified THP nucleoside to the remainder of an oligonucleotide and the other of T 3 and T 4 is H, a hydroxyl protecting group, a linked conjugate group, or a 5′ or 3′-terminal group;
  • q 1 , q 2 , q 3 , q 4 , q 5 , q 6 and q 7 are each, independently, H, C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, C 2 -C 6 alkenyl, substituted C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, or substituted C 2 -C 6 alkynyl; and
  • each of R 1 and R 2 is independently selected from among: hydrogen, halogen, substituted or unsubstituted alkoxy, NJ 1 J 2 , SJ 1 , N 3 , OC( ⁇ X)J 1 , OC( ⁇ X)NJ 1 J 2 , NJ 3 C( ⁇ X)NJ 1 J 2 , and CN, wherein X is O, S or NJ 1 , and each J 1 , J 2 , and J 3 is, independently, H or C 1 -C 6 alkyl.
  • modified THP nucleosides are provided wherein q 1 , q 2 , q 3 , q 4 , q 5 , q 6 and q 7 are each H. In certain embodiments, at least one of q 1 , q 2 , q 3 , q 4 , q 5 , q 6 and q 7 is other than H. In certain embodiments, at least one of q 1 , q 2 , q 3 , q 4 , q 5 , q 6 and q 7 is methyl. In certain embodiments, modified THP nucleosides are provided wherein one of R 1 and R 2 is F. In certain embodiments, R 1 is F and R 2 is H, in certain embodiments, R 1 is methoxy and R 2 is H, and in certain embodiments, R 1 is methoxyethoxy and R 2 is H.
  • sugar surrogates comprise rings having more than 5 atoms and more than one heteroatom.
  • nucleosides comprising morpholino sugar moieties and their use in oligonucleotides have been reported (see, e.g., Braasch et al., Biochemistry, 2002, 41, 4503-4510 and U.S. Pat. Nos. 5,698,685; 5,166,315; 5,185,444; and 5,034,506).
  • morpholino means a sugar surrogate having the following structure:
  • morpholinos may be modified, for example by adding or altering various substituent groups from the above morpholino structure.
  • sugar surrogates are referred to herein as “modified morpholinos.”
  • sugar surrogates comprise acyclic moieties.
  • nucleosides and oligonucleotides comprising such acyclic sugar surrogates include but are not limited to: peptide nucleic acid (“PNA”), acyclic butyl nucleic acid (see, e.g., Kumar et al., Org. Biomol. Chem., 2013, 11, 5853-5865), and nucleosides and oligonucleotides described in WO2011/133876.
  • modified oligonucleotides such as modified crRNAs or modified scrRNAs, comprise one or more nucleoside comprising an unmodified nucleobase. In certain embodiments, modified oligonucleotides comprise one or more nucleoside comprising a modified nucleobase. In certain embodiments, modified oligonucleotides comprise one or more nucleoside that does not comprise a nucleobase, referred to as an abasic nucleoside.
  • modified nucleobases are selected from: 5-substituted pyrimidines, 6-azapyrimidines, alkyl or alkynyl substituted pyrimidines, alkyl substituted purines, and N-2, N-6 and O-6 substituted purines.
  • modified nucleobases are selected from: 2-aminopropyladenine, 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-N-methylguanine, 6-N-methyladenine, 2-propyladenine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-propynyl (—C ⁇ C—CH 3 ) uracil, 5-propynylcytosine, 6-azouracil, 6-azocytosine, 6-azothymine, 5-ribosyluracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl, 8-aza and other 8-substituted purines, 5-halo, particularly 5-bromo, 5-trifluoromethyl, 5-halouracil, and 5-halocytosine, 7-methylguanine, 7-methyla
  • nucleobases include tricyclic pyrimidines, such as 1,3-diazaphenoxazine-2-one, 1,3-diazaphenothiazine-2-one and 9-(2-aminoethoxy)-1,3-diazaphenoxazine-2-one (G-clamp).
  • Modified nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example, 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone.
  • Further nucleobases include those disclosed in U.S. Pat. No.
  • nucleosides of modified oligonucleotides may be linked together using any internucleoside linkage.
  • the two main classes of internucleoside linking groups are defined by the presence or absence of a phosphorus atom.
  • Representative phosphorus-containing internucleoside linkages include but are not limited to phosphates, which contain a phosphodiester bond (“P ⁇ O”) (also referred to as unmodified or naturally occurring linkages), phosphotriesters, methylphosphonates, phosphoramidates, and phosphorothioates (“P ⁇ S”), and phosphorodithioates (“HS—P ⁇ S”).
  • Non-phosphorus containing internucleoside linking groups include but are not limited to methylenemethylimino (—CH 2 —N(CH 3 )—O—CH 2 —), thiodiester (—O—C( ⁇ O)—S—), thionocarbamate (—O—C( ⁇ O)(NH)—S—); siloxane (—O—SiH 2 —O—); and N,N′-dimethylhydrazine (—CH 2 —N(CH 3 )—N(CH 3 )—).
  • Modified internucleoside linkages compared to naturally occurring phosphate linkages, can be used to alter, typically increase, nuclease resistance of the oligonucleotide.
  • internucleoside linkages having a chiral atom can be prepared as a racemic mixture, or as separate enantiomers.
  • Representative chiral internucleoside linkages include but are not limited to alkylphosphonates and phosphorothioates. Methods of preparation of phosphorous-containing and non-phosphorous-containing internucleoside linkages are well known to those skilled in the art.
  • Neutral internucleoside linkages include, without limitation, phosphotriesters, methylphosphonates, MMI (3′-CH 2 —N(CH 3 )—O-5′), amide-3 (3′-CH 2 —C( ⁇ O)—N(H)-5′), amide-4 (3′-CH 2 —N(H)—C( ⁇ O)-5′), formacetal (3′-O—CH 2 —O-5′), methoxypropyl, and thioformacetal (3′-S—CH 2 —O-5′).
  • Further neutral internucleoside linkages include nonionic linkages comprising siloxane (dialkylsiloxane), carboxylate ester, carboxamide, sulfide, sulfonate ester and amides (See for example: Carbohydrate Modifications in Antisense Research ; Y. S. Sanghvi and P. D. Cook, Eds., ACS Symposium Series 580; Chapters 3 and 4, 40-65). Further neutral internucleoside linkages include nonionic linkages comprising mixed N, O, S and CH 2 component parts.
  • the crRNA has a modification motif selected from the table below.
  • f indicates a 2′-F modified nucleoside
  • r indicates an unmodified 2′-hydroxy sugar containing nucleoside
  • d indicates an unmodified 2′-deoxy sugar containing nucleoside
  • e indicates a 2′-MOE modified nucleoside
  • k indicates a cEt bicyclic sugar containing nucleoside
  • l indicates an LNA bicyclic sugar containing nucleoside.
  • the modifications listed in parentheses are optional modified nucleobases or optional modified internucleoside linkages: “(G-Clamp)” indicates a G-Clamp modified nucleobase that is part of the nucleoside represented by the letter immediately preceding it.
  • (5-Propyne) indicates a 5′-propynyl modified nucleobase that is part of the nucleoside represented by the letter immediately preceding it.
  • (MOP) indicates a mcthoxypropyl modified internucleoside linkage.
  • (MP) indicates a methylphosphonatc internucleoside linkage, and
  • (MMI) indicates an MMI N-methyl internucleoside linkage.
  • crRNAs having a motif with a parenthetical modification listed in the table above include the indicated parenthetical modification.
  • the parenthetical modification of crRNAs having a motif with a parenthetical modification listed in the table above is replaced with a different modified or unmodified nucleobase or internucleoside linkage.
  • the number subscripts in the table above indicate the number of contiguous nucleosides that comprise the identified modification. The lack of a number subscript indicates one nucleoside.
  • the motifs listed in the table “above may be used with any crRNA nucleobase sequence and with any internucleoside linkage motif.
  • all of the nucleobases arc unmodified.
  • at least one nucleobase is a 5-methylcytosine modified nucleobase.
  • the internucleoside linkages are all selected independently from among phosphate and phosphorothioate.
  • one or more internucleoside linkages is a neutral internucleoside linkage.
  • oligonucleotides for use as crRNA or scrRNA further comprise conjugate groups and/or terminal groups.
  • compounds comprising oligonucleotides for use as crRNA or scrRNA further comprise a conjugate group or terminal group.
  • oligonucleotides are covalently attached to one or more conjugate group.
  • conjugate groups modify one or more properties of the attached oligonucleotide, including but not limited to pharmacodynamics, pharmacokinetics, stability, binding, absorption, cellular distribution, cellular uptake, charge and clearance.
  • conjugate groups impart a new property on the attached oligonucleotide, e.g., fluorophores or reporter groups that enable detection of the oligonucleotide.
  • Conjugate groups and/or terminal groups may be added to oligonucleotides having any of the modifications or motifs described above.
  • Conjugate groups include, without limitation, intercalators, reporter molecules, polyamines, polyamides, peptides, carbohydrates, vitamin moieties, polyethylene glycols, thioethers, polyethers, cholesterols, thiocholesterols, cholic acid moieties, folate, lipids, phospholipids, biotin, phenazine, phenanthridine, anthraquinone, adamantane, acridine, fluoresceins, rhodamines, coumarins, fluorophores, and dyes.
  • Certain conjugate groups have been described previously, for example: cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci.
  • Acids Res., 1990, 18, 3777-3783 a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237), an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., H. Pharmacol. Exp.
  • a conjugate group comprises an active drug substance, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, (S)-(+)-pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, fingolimod, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indo-methicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
  • an active drug substance for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, (S)-(+)-pranoprofen, carpro
  • Conjugate groups are attached directly or via an optional conjugate linker to a parent compound, such as a crRNA or scrRNA oligonucleotide.
  • conjugate groups are directly attached to oligonucleotides.
  • conjugate groups are indirectly attached to oligonucleotides via conjugate linkers.
  • the conjugate linker comprises a chain structure, such as a hydrocarbyl chain, or an oligomer of repeating units such as ethylene glycol or amino acid units.
  • conjugate groups comprise a cleavable moiety.
  • conjugate groups are attached to oligonucleotides via a cleavable moiety.
  • conjugate linkers comprise a cleavable moiety.
  • conjugate linkers are attached to oligonucleotides via a cleavable moiety.
  • oligonucleotides comprise a cleavable moiety, wherein the cleavable moiety is a nucleoside is attached to a cleavable internucleoside linkage, such as a phosphate internucleoside linkage.
  • a conjugate group comprises a nucleoside or oligonucleotide, wherein the nucleoside or oligonucleotide of the conjugate group is indirectly attached to a parent oligonucleotide.
  • a conjugate linker comprises one or more groups selected from alkyl, amino, oxo, amide, disulfide, polyethylene glycol, ether, thioether, and hydroxylamino. In certain such embodiments, the conjugate linker comprises groups selected from alkyl, amino, oxo, amide and ether groups. In certain embodiments, the conjugate linker comprises groups selected from alkyl and amide groups. In certain embodiments, the conjugate linker comprises groups selected from alkyl and ether groups. In certain embodiments, the conjugate linker comprises at least one phosphorus moiety. In certain embodiments, the conjugate linker comprises at least one phosphate group. In certain embodiments, the conjugate linker includes at least one neutral linking group.
  • conjugate linkers are bifunctional linking moieties, e.g., those known in the art to be useful for attaching conjugate groups to parent compounds, such as the crRNA oligonucleotides provided herein and the scrRNA oligonucleotides provided herein.
  • a bifunctional linking moiety comprises at least two functional groups. One of the functional groups is selected to bind to a particular site on a parent compound and the other is selected to bind to a conjugate group.
  • Examples of functional groups used in a bifunctional linking moiety include but are not limited to electrophiles for reacting with nucleophilic groups and nucleophiles for reacting with electrophilic groups.
  • bifunctional linking moieties comprise one or more groups selected from amino, hydroxyl, carboxylic acid, thiol, alkyl, alkenyl, and alkynyl.
  • conjugate linkers include but are not limited to pyrrolidine, 8-amino-3,6-dioxaoctanoic acid (ADO), succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC) and 6-aminohexanoic acid (AHEX or AHA).
  • ADO 8-amino-3,6-dioxaoctanoic acid
  • SMCC succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate
  • AHEX or AHA 6-aminohexanoic acid
  • conjugate linkers include but are not limited to substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 2 -C 10 alkenyl or substituted or unsubstituted C 2 -C 10 alkynyl, wherein a nonlimiting list of preferred substituent groups includes hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro, thiol, thioalkoxy, halogen, alkyl, aryl, alkenyl and alkynyl.
  • a cleavable moiety is a cleavable bond. In certain embodiments, a cleavable moiety comprises a cleavable bond. In certain embodiments, a cleavable moiety is a group of atoms comprising at least one cleavable bond. In certain embodiments, a cleavable moiety comprises a group of atoms having one, two, three, four, or more than four cleavable bonds. In certain embodiments, a cleavable moiety is selectively cleaved inside a cell or subcellular compartment, such as a lysosome. In certain embodiments, a cleavable moiety is selectively cleaved by endogenous enzymes, such as nucleases.
  • a cleavable bond is selected from among: an amide, an ester, an ether, one or both esters of a phosphodiester, a phosphate ester, a carbamate, or a disulfide. In certain embodiments, a cleavable bond is one or both of the esters of a phosphodiester. In certain embodiments, a cleavable moiety comprises a phosphate or phosphodiester. In certain embodiments, the cleavable moiety is a phosphate linkage between an oligonucleotide and a conjugate linker or conjugate group.
  • a cleavable moiety is a nucleoside.
  • the unmodified or modified nucleoside comprises an optionally protected heterocyclic base selected from a purine, substituted purine, pyrimidine or substituted pyrimidine.
  • a cleavable moiety is a nucleoside selected from uracil, thymine, cytosine, 4-N-benzoylcytosine, 5-methylcytosine, 4-N-benzoyl-5-methylcytosine, adenine, 6-N-benzoyladenine, guanine and 2-N-isobutyrylguanine.
  • a cleavable moiety is 2′-deoxy nucleoside that is attached to either the 3′ or 5′-terminal nucleoside of an oligonucleotide by a phosphate internucleoside linkage and covalently attached to the conjugate linker or conjugate group by a phosphate or phosphorothioate linkage.
  • the cleavable moiety is 2′-deoxyadenosine.
  • Conjugate groups may be attached to either or both ends of an oligonucleotide and/or at any internal position. In certain embodiments, conjugate groups are attached to the 2′-position of a nucleoside of a modified oligonucleotide. In certain embodiments, conjugate groups that are attached to either or both ends of an oligonucleotide are terminal groups. In certain such embodiments, conjugate groups or terminal groups are attached at the 3′ and/or 5′-end of oligonucleotides. In certain such embodiments, conjugate groups (or terminal groups) are attached at the 3′-end of oligonucleotides. In certain embodiments, conjugate groups are attached near the 3′-end of oligonucleotides. In certain embodiments, conjugate groups (or terminal groups) are attached at the 5′-end of oligonucleotides. In certain embodiments, conjugate groups are attached near the 5′-end of oligonucleotides.
  • terminal groups include but are not limited to conjugate groups, capping groups, phosphate moieties, protecting groups, modified or unmodified nucleosides, and two or more nucleosides that are independently modified or unmodified.
  • a conjugate group is a cell-targeting moiety.
  • a conjugate group, optional conjugate linker, and optional cleavable moiety have the general formula:
  • n is from 1 to about 3, m is 0 when n is 1, m is 1 when n is 2 or greater, j is 1 or 0, and k is 1 or 0.
  • n is 1, j is 1 and k is 0. In certain embodiments, n is 1, j is 0 and k is 1. In certain embodiments, n is 1, j is 1 and k is 1. In certain embodiments, n is 2, j is 1 and k is 0. In certain embodiments, n is 2, j is 0 and k is 1. In certain embodiments, n is 2, j is 1 and k is 1. In certain embodiments, n is 3, j is 1 and k is 0. In certain embodiments, n is 3, j is 0 and k is 1. In certain embodiments, n is 3, j is 1 and k is 1. In certain embodiments, n is 3, j is 1 and k is 1. In certain embodiments, n is 3, j is 1 and k is 1.
  • conjugate groups comprise cell-targeting moieties that have at least one tethered ligand.
  • cell-targeting moieties comprise two tethered ligands covalently attached to a branching group.
  • cell-targeting moieties comprise three tethered ligands covalently attached to a branching group.
  • the cell-targeting moiety comprises a branching group comprising one or more groups selected from alkyl, amino, oxo, amide, disulfide, polyethylene glycol, ether, thioether and hydroxylamino groups.
  • the branching group comprises a branched aliphatic group comprising groups selected from alkyl, amino, oxo, amide, disulfide, polyethylene glycol, ether, thioether and hydroxylamino groups.
  • the branched aliphatic group comprises groups selected from alkyl, amino, oxo, amide and ether groups.
  • the branched aliphatic group comprises groups selected from alkyl, amino and ether groups. In certain such embodiments, the branched aliphatic group comprises groups selected from alkyl and ether groups. In certain embodiments, the branching group comprises a mono or polycyclic ring system.
  • each tether of a cell-targeting moiety comprises one or more groups selected from alkyl, substituted alkyl, ether, thioether, disulfide, amino, oxo, amide, phosphodiester, and polyethylene glycol, in any combination.
  • each tether is a linear aliphatic group comprising one or more groups selected from alkyl, ether, thioether, disulfide, amino, oxo, amide, and polyethylene glycol, in any combination.
  • each tether is a linear aliphatic group comprising one or more groups selected from alkyl, phosphodiester, ether, amino, oxo, and amide, in any combination.
  • each tether is a linear aliphatic group comprising one or more groups selected from alkyl, ether, amino, oxo, and amid, in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, amino, and oxo, in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl and oxo, in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl and phosphodiester, in any combination. In certain embodiments, each tether comprises at least one phosphorus linking group or neutral linking group.
  • each tether comprises a chain from about 6 to about 20 atoms in length. In certain embodiments, each tether comprises a chain from about 10 to about 18 atoms in length. In certain embodiments, each tether comprises about 10 atoms in chain length.
  • each ligand of a cell-targeting moiety has an affinity for at least one type of receptor on a target cell. In certain embodiments, each ligand has an affinity for at least one type of receptor on the surface of a mammalian liver cell. In certain embodiments, each ligand has an affinity for the hepatic asialoglycoprotein receptor (ASGP-R). In certain embodiments, each ligand is a carbohydrate. In certain embodiments, each ligand is, independently selected from galactose, N-acetyl galactoseamine (GalNAc), mannose, glucose, glucosamine and fucose.
  • GalNAc N-acetyl galactoseamine
  • each ligand is N-acetyl galactoseamine (GalNAc).
  • the cell-targeting moiety comprises 3 GalNAc ligands. In certain embodiments, the cell-targeting moiety comprises 2 GalNAc ligands. In certain embodiments, the cell-targeting moiety comprises 1 GalNAc ligand.
  • the present invention provides pharmaceutical compositions comprising one or more crRNA.
  • such pharmaceutical composition comprises a tracrRNA.
  • the pharmaceutical composition comprises a means of expressing Cas9.
  • such means of expressing Cas9 is a plasmid or a viral vector.
  • the pharmaceutical composition comprises a suitable pharmaceutically acceptable diluent or carrier.
  • a pharmaceutical composition comprises a sterile saline solution and one or more antisense compound.
  • such pharmaceutical composition consists of a sterile saline solution and one or more antisense compound.
  • the sterile saline is pharmaceutical grade saline.
  • a pharmaceutical composition comprises one or more antisense compound and sterile water. In certain embodiments, a pharmaceutical composition consists of one antisense compound and sterile water. In certain embodiments, the sterile water is pharmaceutical grade water. In certain embodiments, a pharmaceutical composition comprises one or more antisense compound and phosphate-buffered saline (PBS). In certain embodiments, a pharmaceutical composition consists of one or more antisense compound and sterile PBS. In certain embodiments, the sterile PBS is pharmaceutical grade PBS.
  • PBS phosphate-buffered saline
  • the present invention provides pharmaceutical compositions comprising one or more scrRNA.
  • the pharmaceutical composition comprises a means of expressing a nuclease.
  • such means of expressing the nuclease is a plasmid or a viral vector.
  • the pharmaceutical composition comprises a suitable pharmaceutically acceptable diluent or carrier.
  • a pharmaceutical composition comprises a sterile saline solution and one or more antisense compound.
  • such pharmaceutical composition consists of a sterile saline solution and one or more antisense compound.
  • the sterile saline is pharmaceutical grade saline.
  • a pharmaceutical composition comprises one or more antisense compound and sterile water. In certain embodiments, a pharmaceutical composition consists of one antisense compound and sterile water. In certain embodiments, the sterile water is pharmaceutical grade water. In certain embodiments, a pharmaceutical composition comprises one or more antisense compound and phosphate-buffered saline (PBS). In certain embodiments, a pharmaceutical composition consists of one or more antisense compound and sterile PBS. In certain embodiments, the sterile PBS is pharmaceutical grade PBS.
  • PBS phosphate-buffered saline
  • RNA nucleoside comprising a 2′-OH sugar moiety and a thymine base
  • RNA methylated uracil
  • nucleic acid sequences provided herein are intended to encompass nucleic acids containing any combination of natural or modified RNA and/or DNA, including, but not limited to such nucleic acids having modified nucleobases.
  • an oligomeric compound having the nucleobase sequence “ATCGATCG” encompasses any oligomeric compounds having such nucleobase sequence, whether modified or unmodified, including, but not limited to, such compounds comprising RNA bases, such as those having sequence “AUCGAUCG” and those having some DNA bases and some RNA bases such as “AUCGATCG” and oligomeric compounds having other modified or naturally occurring bases, such as
  • ATmCGAUCG wherein m C indicates a cytosine base comprising a methyl group at the 5-position.
  • oligonucleotide having a particular motif provides reasonable support for additional oligonucleotides having the same or similar motif.
  • motifs of crRNA described herein can also be applied to scrRNAs.
  • motifs of the DNA recognition portions of the crRNAs described herein may be applied to the scrRNA target recognition portions of scrRNAs.
  • motifs of the tracrRNA recognition portions of the crRNAs described herein may be applied to the nuclease recognition portions of scrRNAs.
  • Modified crRNAs comprising a DNA recognition portion that is complementary to hLDLR were designed and synthesized to test their effects on gene editing of the human LDLR locus.
  • HEK 293T cells were transfected with a plasmid expressing Cas9 protein and tracrRNA using Lipofectamine 3000 (Life Technologies).
  • cells were transfected with a plasmid expressing Cas9 protein and a highly active sgRNA as a positive control or no Cas9 (“Cas9 Ctrl”) as a negative control.
  • RNAiMAX Life Technologies
  • neg ctrl no crRNA as a control
  • genomic DNA was isolated from cells and used in a SURVEYOR assay (Integrated DNA Technologies) according to the manufacturer's directions.
  • the PCR primers used to amplify the crRNA target site were forward: 5′-GGAGACCCAAATACAACAAATC-3′ (SEQ ID NO: 1) and reverse: 5′-CTAGACTCCGTCTCAAAGAAG-3′ (SEQ ID NO: 2).
  • crRNA targeting hLDLR Gene disruption normalized SEQ to sgRNA ID Name Sequence (5′ to 3′) (%) NO.
  • crRNA GCGCCTTGCTCCTCGCCGCGG 7 5 GUUUUAGAUCUAUGCUGUUUU G PS crRNA G s C s G s C s CT s T s G s C s T s C s C s T s 33 5 C s G s C s C s C s G s C s G s G s G s G s U s U s U s U s A s G s A s U s C s U s A s U s G s C s U s G s U UU s U s G PS 2′-OMe G ms C ms G m C ms C m T s T s G s C s T s C s 47 5 crRNA C s T s C s G s C
  • Modified crRNAs comprising a DNA recognition portion that is complementary to hVEGFA were designed and synthesized to test their effects on gene editing of the human VEGFA locus.
  • HEK 293T cells were transfected as described in Example 1 using a crRNA described in the table below.
  • the SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the crRNA target site were forward: 5′-TCCAGATGGCACATTGTCAG-3′ (SEQ ID NO: 3) and reverse: 5′-AGGGAGCAGGAAAGTGAGGT-3′ (SEQ ID NO: 4).
  • CelI cleavage the DNA was run on a gel to analyze the extent of gene editing of hVEGFA (see FIG.
  • Example 2 the gel was quantified as described in Example 1.
  • the results for the modified crRNAs were normalized to a positive control sgRNA targeted to hVEGFA to determine the gene disruption percentage shown in the table below. The results indicate that many of the modified crRNAs were active.
  • Modified crRNAs comprising a DNA recognition portion that is complementary to hVEGFA were designed and synthesized to test their effects on gene editing of the human VEGFA locus.
  • HEK 293T cells were transfected as described in Example 1 using a crRNA described in the table below, and the Cas9/tracrRNA load time was 24 hours.
  • the SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the crRNA target site were forward: 5′-TCCAGATGGCACATTGTCAG-3′ (SEQ ID NO: 3) and reverse: 5′-AGGGAGCAGGAAAGTGAGGT-3′ (SEQ ID NO: 4).
  • the DNA was run on a gel to analyze the extent of gene editing of hVEGFA (see FIG. 3 ), and the gel was quantified as described in Example 1.
  • the results for the modified crRNAs were normalized to a positive control sgRNA targeted to hVEGFA to determine the gene disruption percentage shown in the table below. The results indicate that many of the modified crRNAs were active or very active.
  • modified crRNAs comprising a DNA recognition portion that is under 20 nucleosides in length and/or a tracrRNA recognition portion that is under 22 nucleosides in length were designed and synthesized to test their effects on gene editing of the human VEGFA locus.
  • HEK 293T cells were transfected as described in Example 1 using a crRNA described in the table below.
  • the SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the crRNA target site were forward: 5′-TCCAGATGGCACATTGTCAG-3′ (SEQ ID NO: 3) and reverse: 5′-AGGGAGCAGGAAAGTGAGGT-3′ (SEQ ID NO: 4).
  • the DNA was run on a gel to analyze the extent of gene editing of hVEGFA (see FIG. 3 ). The experiment was repeated, and the resulting gel was quantified as described in Example 1.
  • the results for the modified crRNAs were normalized to a positive control sgRNA targeted to hVEGFA to determine the gene disruption percentage shown in the table below. The results indicate that many of the shortened, modified crRNAs were active, including crRNAs that comprise only a 12 nucleoside tracrRNA recognition portion and only a 17 nucleoside DNA recognition portion.
  • Modified crRNAs having the motifs described in the table below can be used for any crRNA nucleobase sequence.
  • the first 17 to 20 nucleosides of each motif represent the DNA recognition portion of the crRNA, and the remaining 12 to 22 nucleosides of each motif represent the tracrRNA recognition portion of the crRNA.
  • the motifs labeled “29-mers” contain 29 linked nucleosides, and the motifs labeled “42-mers” contain 42 linked nucleosides.
  • the motifs described below can also be applied to crRNAs of other lengths, wherein the pattern is extended or shortened as required to fit the oligonucleotide length. The modifications of the motifs are described using the same single letter identifiers used in the subscripts of Tables 1-4 above.
  • the number subscripts indicate the number of contiguous nucleosides that comprise the identified modification. The lack of a number subscript indicates one nucleoside. Additional abbreviations are: “1” indicates an LNA modification, “(MOP)” indicates a methoxypropyl modified internucleoside linkage, “(MP)” indicates a methylphosphonate internucleoside linkage, “(MMI)” indicates an MMI N-methyl internucleoside linkage, “(5-propyne)” indicates a 5-propyne nucleobase modification, and “(G-clamp)” indicates a G-clamp modified nucleobase.
  • Modified crRNAs comprising a DNA recognition portion that is complementary to hVEGFA were designed and synthesized to test their effects on gene editing of the human VEGFA locus.
  • HEK 293T cells were transfected as described in Example 1 using a crRNA described in the table below.
  • the SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the crRNA target site were forward: 5′-TCCAGATGGCACATTGTCAG-3′ (SEQ ID NO: 3) and reverse: 5′-AGGGAGCAGGAAAGTGAGGT-3′ (SEQ ID NO: 4).
  • the DNA was run on a gel to analyze the extent of gene editing of hVEGFA, and the gel was quantified as described in Example 1.
  • the results for the modified crRNAs were normalized to a positive control sgRNA targeted to hVEGFA to determine the gene disruption percentage shown in the table below. The results indicate that many of the modified crRNAs were active and some were even more active than the sgRNA positive control.
  • Isis Numbers 801193 (Example 3), 801381 (Example 4), and 834472 (Example 6) were tested for their effects on gene editing of the human Myc-associated factor X (MAX) locus.
  • MAX Myc-associated factor X
  • the MAX gene carries an 18 out of 20 nucleotide match to a portion of or all of the region of the VEGFA gene targeted by Isis Numbers 801193, 801381, and 834472.
  • HEK 293T cells were transfected as described in Example 1 using Isis No. 801193, 801381, or 834472 as the modified crRNA.
  • the SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the off-target site in the MAX gene were forward: 5′-TACCCGGGCCGTCTGTTAGA-3′ (SEQ ID NO: 17) and reverse: 5′-GAGGGGGAAGTCACCGACAA-3′ (SEQ ID NO: 18). Following Cell cleavage, the DNA was run on a gel to analyze the extent of gene editing of MAX. Quantification was performed as described in Example 1. The results for the modified crRNAs were normalized to a positive control sgRNA targeted to hVEGFA to determine the gene disruption percentage shown in the table below. The results indicate that the modified crRNAs exhibited less off-target effects than the sgRNA control. The on-target effects of the modified crRNAs (see Examples 3, 4, and 6) are shown in the third column below, for comparison.
  • Modified crRNAs comprising a DNA recognition portion that is complementary to human TTR were designed and synthesized to test their effects on gene editing of the hTTR locus.
  • HEK 293T cells were transfected as described in Example 1 using a crRNA described in the table below.
  • the SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the crRNA target site were forward: 5′-GCTGACTAAGCAAAGCTTCCAAATGAC-3′ (SEQ ID NO:41) and reverse: 5′-GATGTCACAGAAACACTCACCGTAG-3′ (SEQ ID NO: 42).
  • the DNA was run on a gel to analyze the extent of gene editing of hTTR, and the gel was quantified as described in Example 1.
  • the results for the modified crRNAs were normalized to a positive control sgRNA targeted to hTTR to determine the gene disruption percentage shown in the table below. The results indicate that many of the modified crRNAs were active and some were even more active than the sgRNA positive control.
  • Truncated scrRNAs comprising a scrRNA target recognition portion that is complementary to DNA (cytosine-5)-methyltransferase 1 (DNMT1) were designed and synthesized to test their effects on alteration of the DNMT1 gene.
  • HEK293T cells were transfected with a plasmid encoding Cpf1 and a double-stranded gblock (IDT, Coralville, Iowa) encoding a scrRNA listed in the table below.
  • the SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the scrRNA site in the DNMT1 gene were forward: 5′-CTGGGACTCAGGCGGGTCAC-3′ (SEQ ID NO: 47) and reverse: 5′-CCTCACACAACAGCTTCATGTCAGC-3′ (SEQ ID NO:). Following Cell cleavage, the DNA was run on a gel to analyze the extent of gene alteration of DNMT1. The results are shown in FIGS. 4 a and 4 b . The results indicate that multiple truncated scrRNAs, including scrRNA containing only 36 nucleosides, altered the target gene.
  • Truncated tracrRNAs were designed and synthesized to test their effects on editing of mouse Proprotein Convertase Subtilisin/Kexin Type 9 (Pcsk9).
  • Pcsk9 DNA To generate Pcsk9 DNA, a portion of the mouse genomic locus encompassing the CRISPR target site was amplified by PCR using primers 5′-CTGAGGCTAGAGGACTGAGC-3′ (SEQ ID NO: 61) and 5′-CAGACGGCTAGATGAGCAGAG-3′ (SEQ ID NO: 62). 30 nM of a modified crRNA, Ion No.
  • 927720 shown in the table below and 30 nM of a tracrRNA shown in the table below and were used to test for Pcsk9 gene disruption in an in vitro biochemical assay. Following cleavage by Cas9, the DNA was run on a gel to analyze the extent of activity. The results are shown in FIG. 5 . The results indicate that the truncated tracrRNAs exhibited activity in vitro.
  • nucleosides that are not underlined represent the tracrRNA recognition portion of the crRNA. All of the nucleosides in the tracrRNAs shown in the table above are unmodified ribonucleosides comprising 2′-hydroxy sugar moieties and phosphate internucleoside linkages.
  • modified crRNAs to activate target genes was tested in a transcriptional activation assay, similar to that described in Konermann et al., Nature 517, 583-588 (2015). Briefly, one MS2 aptamer sequence was inserted at position 58 of tracrRNA.
  • HEK 293 cells were transfected with PBS alone (negative control) or with a plasmids encoding catalytically inactive Cas9 fused to Tetrameric VP16 transcription activator domain (dCas9-VP64), MS2-p65-HSF1 activation helper protein as described in Konermann et al. and the MS2 aptamer containing tracrRNA1.2.
  • Modified crRNA comprising a DNA recognition portion that is complementary to human TTR, listed in the table below, was added in PBS, in the absence of a transfection reagent, at a final concentration of 1 uM. PBS without crRNA was added in the “no RNA” control. After 48 hours, total RNA was isolated, and gene activation was measured using RT-qPCR using forward primer 5′-CTTGCTGGACTGGTATTTGTGTCT-3′(SEQ ID NO: 67), reverse primer 5′-AGAACTTTGACCATCAGAGGACACT-3′ (SEQ ID NO: 68) and probe 5′-CCCTACGGGCACCGGTGAATCC-3′ (SEQ ID NO: 69). The RT-qPCR results were normalized to GAPDH and are presented in the table below as the fold change relative to the negative control, which was set to 1.0. The results show that modified crRNA was taken up by the cells by free uptake and induced target gene activation.
  • Example 12 In Vitro Digestion of Pcsk9 DNA Using Compounds Comprising Modified crRNA
  • Compounds comprising modified crRNAs shown in the tables below comprise a DNA recognition portion that is complementary to mouse Pcsk9.
  • the modified crRNAs shown in Table 12 below are made and tested for their DNA cutting activity and/or gene disruption activity, as described herein.
  • the modified crRNAs shown in Table 13 were synthesized and tested for DNA cutting activity in vitro.
  • Ion No. 927722 comprises a GalNAc conjugate group (“LICA-1”), and the synthesis of Ion No. 927722 is shown below.
  • the DNA cutting assay was carried out as described in Example 10. Ion No. 927720 or 927722 was used with a tracrRNA. An sgRNA was used alone as a positive control. The results are shown in FIG. 6 .
  • the results show that the modified crRNA with no attached conjugate group cut Pcsk9 DNA more potently than the sgRNA positive control in vitro.
  • the modified crRNA attached to the GalNAc conjugate group cut Pcsk9 DNA to an extent approximately equal to that of the sgRNA positive control.
  • Standard phosphoramidites and solid supports were used for incorporation of A, U, G, and C nucleosides.
  • a 0.2 M solution of the amidites in anhydrous acetonitrile was used for the synthesis.
  • a 0.2 M solution of 2′-O-Me A Bz , U, G ibu and C Bz phosphoramidites in anhydrous acetonitrile were used for the incorporation of 2′-O-methyl modified nucleotides.
  • the modified crRNAs (60 ⁇ mol scale) were synthesized using an ⁇ KTAOligopilot synthesizer (GE Healthcare Biosciences) on VIMAD UnyLinkerTM solid support (100 ⁇ mol/g loading) and the appropriate amounts of solid supports were packed in the column for synthesis.
  • Dichloroacetic acid (6%) in toluene was used as detritylating reagent.
  • 4,5-Dicyanoimidazole in the presence of N-methylimidazole in CH 3 CN was used as activator during the coupling step.
  • 0.1 M xanthane hydride solution in 50% pyridine in acetonitrile was used as sulfurizing agent with 3 min contact time.
  • Twelve equivalents of THA-GalNAc phosphoramidite 4 was delivered in 3 portions, each followed by a 12 min coupling wait time. All other steps in the protocol supplied by the manufacturer were used without modification. The coupling efficiencies were more than 97%.
  • solid support was treated with 20% diethylamine in toluene for 45 min to remove cyanoethyl group from phosphorothioate linkages.
  • the solid support was then suspended in aqueous ammonium hydroxide (30 wt. %): ethanol (3:1) and allowed to stir at room temperature for 4 h.
  • aqueous ammonium hydroxide (30 wt. %): ethanol (3:1) and allowed to stir at room temperature for 4 h.
  • the solid support was filtered and the filtrate was concentrated to dryness.
  • the fractions containing full length crRNAs were pooled together was desalted by HPLC on reverse phase column to yield the crRNA in an isolated yield of 10% based on solid-support loading.
  • the oligonucleotides were characterized by ion-pair-HPLC-MS analysis with Agilent 1100 MSD system.
  • Modified crRNA was tested for gene editing of Pcsk9 ex vivo.
  • Hepatocytes from mice that express Cas9 (described in Platt et al., Cell 159, 440-455 (2014)) were cultured in William's media E supplemented with 10% FBS, 4 mM L-Glutamine and 25 mM HEPES.
  • the hepatocytes were transfected with Ion No. 927720 (see Example 12) and a tracrRNA or a sgRNA positive control alone using lipofectamine RNAiMax (Life Technologies, Carlsbad).
  • Pcsk9 gene disruption was measured using the SURVEYOR assay. The results are shown in FIG. 7 .
  • the results indicate that a modified crRNA disrupted the Pcsk9 gene with similar potency to a sgRNA positive control in hepatocytes ex vivo.

Abstract

The present disclosure provides compounds comprising modified oligonucleotides for use in CRISPR. In certain embodiments, such modified oligonucleotides provide improved properties of crRNA. In certain embodiments, such modified oligonucleotides provide improved properties of scrRNA.

Description

    SEQUENCE LISTING
  • The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled CORE0134USC1SEQ_ST25.txt, created May 17, 2022, which is 20 Kb in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.
  • BACKGROUND
  • Use of Cluster Regulatory Interspaced Short Palindromic Repeats (CRISPR) to edit or disable genes has been described. See for example Jinek et al., Science 337: 816-821 (2012); Mali et al. Science 339: 823-826 (2013).
  • SUMMARY
  • Various CRISPR systems have been described. See for example: WO2013/176772; WO2015/006747; Qi et al., Cell 152: 1 173-1 (2013); Gilbert et al., Cell 154: 1-10 (2013) Jinek et al., Science 337: 816-821 (2012); Mali et al. Science 339: 823-826 (2013); Doudna et al., Science 346: 6213 (2014). See also for example: Zetsche et al., Cell 163: 1-13 (2015). The present invention provides modified oligonucleotides for use as crRNA in CRISPR systems. In certain embodiments, such modified crRNA have improved stability relative to unmodified crRNA. In certain embodiments, modified crRNA is stabilized at the 5′ end and/or the 3′. In certain embodiments, such stabilized crRNA is resistant to exonuclease and/or endonucleoase digestion. In certain embodiments, modified crRNA have improved affinity for target DNA relative to unmodified crRNA. In certain embodiments, modified crRNA have improved selectivity for target DNA relative to unmodified crRNA. In certain embodiments, modified crRNA have improved affinity for tracrRNA relative to unmodified crRNA. In certain embodiments, modified crRNA have improved cellular uptake relative to unmodified crRNA.
  • In certain such embodiments, the modifications increase affinity for the target DNA allowing the modified crRNA to be shortened while retaining sufficient affinity to hybridize to target DNA and to tracrRNA. Thus, in certain embodiments, modified crRNA is shorter than unmodified crRNA. In certain embodiments, modified crRNA is 40-50 linked nucleosides in length. In certain embodiments, modified crRNA is 35-45 linked nucleosides in length. In certain embodiments, modified crRNA is 30-40 linked nucleosides in length. In certain embodiments, modified crRNA is 25-35 linked nucleosides in length. In certain embodiments, modified crRNA is 20-30 linked nucleosides in length. In certain embodiments, modified crRNA is 25-35 linked nucleosides in length. In certain embodiments, modified crRNA is 20-30 linked nucleosides in length. In certain such embodiments, such shorter crRNA have improved uptake properties. In certain embodiments, modified crRNA are taken into cells without transfection reagents or electroporation. In certain such embodiments, the cells are in an animal. In certain embodiments, the animal expresses Cas9. In certain embodiments, the animal is previously or concomitantly treated with a means of expressing Cas9. In certain such embodiments, such treatment comprises administration of a vector for delivering Cas9. In certain such embodiments, such vector is a viral vector, for example adeno-associated virus (AAV). In certain such embodiments, the viral vector expresses a S. aureus derived Cas9 that fits into an AAV vector.
  • The present invention also provides modified oligonucleotides for use as scrRNA in CRISPR systems. In certain embodiments, such modified scrRNA have improved stability relative to unmodified scrRNA. In certain embodiments, modified scrRNA is stabilized at the 5′ end and/or the 3′. In certain embodiments, such stabilized scrRNA is resistant to exonuclease and/or endonucleoase digestion. In certain embodiments, modified scrRNA have improved affinity for scrRNA target DNA relative to unmodified scrRNA. In certain embodiments, modified scrRNA have improved selectivity for scrRNA target DNA relative to unmodified scrRNA. In certain embodiments, modified scrRNA have improved affinity for a nuclease relative to unmodified scrRNA. In certain embodiments, modified scrRNA have improved cellular uptake relative to unmodified scrRNA.
  • In certain such embodiments, the modifications increase affinity for the scrRNA target DNA allowing the modified scrRNA to be shortened while retaining sufficient affinity to hybridize to scrRNA target DNA and a nuclease. Thus, in certain embodiments, modified scrRNA is shorter than unmodified scrRNA. In certain embodiments, modified scrRNA is 40-50 linked nucleosides in length. In certain embodiments, modified scrRNA is 35-45 linked nucleosides in length. In certain embodiments, modified scrRNA is 30-40 linked nucleosides in length. In certain embodiments, modified scrRNA is 25-35 linked nucleosides in length. In certain embodiments, modified scrRNA is 20-30 linked nucleosides in length. In certain embodiments, modified scrRNA is 25-35 linked nucleosides in length. In certain such embodiments, such shorter scrRNA have improved uptake properties. In certain embodiments, modified scrRNA are taken into cells without transfection reagents or electroporation. In certain such embodiments, the cells are in an animal. In certain embodiments, the animal expresses a nuclease that is recognized by the scrRNA (e.g., a Cpf1 nuclease). In certain embodiments, the animal is previously or concomitantly treated with a means of expressing a nuclease that is recognized by the scrRNA (e.g., a Cpf1 nuclease). In certain such embodiments, such treatment comprises administration of a vector for delivering a nuclease that is recognized by the scrRNA (e.g., a Cpf1 nuclease). In certain such embodiments, such vector is a viral vector, for example adeno-associated virus (AAV).
  • In certain embodiments, the CRISPR system is inhibited after the target gene is edited or the scrRNA target gene is altered. In certain such embodiments, the modified crRNA or modified scrRNA inside a cell is degraded after the target gene or scrRNA target gene has been edited or altered. In certain such embodiments, the nuclease (e.g., Cas9 or a Cpf1 nuclease) continues to be expressed in the cell but is no longer active because it requires crRNA or scrRNA in order to exhibit nuclease activity. In certain such embodiments, off-target effects of the CRISPR system, such as undesired cleavage of an off-target gene, are decreased relative to a CRISPR system in which all of the components necessary for nuclease activity continue to be expressed indefinitely, e.g. by a viral vector. In certain such embodiments, degradation of the modified crRNA or modified scrRNA is facilitated by hybridization to an oligonucleotide complementary to the crRNA or scrRNA. In certain embodiments, degradation of the modified crRNA or modified scrRNA is facilitated by nucleases present in the cell.
  • In certain embodiments, the CRISPR system is inhibited after the target gene is edited via degradation of a tracrRNA inside the cell. In certain such embodiments, degradation of the tracrRNA is facilitated by hybridization to an oligonucleotide complementary to the tracrRNA. In certain embodiments, degradation of the tracrRNA is facilitated by nucleases present in the cell.
  • In certain embodiments, the CRISPR system is inhibited after the target gene is edited or the scrRNA target gene is altered via inhibition of the expression of a nuclease (e.g., Cas9 or a Cpf1 nuclease). In certain such embodiments, the nuclease gene is edited or altered by a modified crRNA or a modified scrRNA. In certain embodiments, the nuclease transcript is degraded following hybridization of the nuclease transcript to an oligonucleotide complementary to the nuclease transcript.
  • The following non-limiting numbered embodiments are provided.
      • Embodiment 1. A compound comprising a modified crRNA consisting of 20-50 linked nucleosides.
      • Embodiment 2. The compound of embodiment 1, wherein the modified crRNA is 5′-stabilized.
      • Embodiment 3. The compound of embodiment 1 or 2, wherein the modified crRNA is 3′-stabilized.
      • Embodiment 4. The compound of any of embodiments 1-3, wherein the modified crRNA comprises at least one modification that increases affinity of the crRNA for a target DNA.
      • Embodiment 5. The compound of any of embodiments 1-4, wherein the modified crRNA comprises at least one modification that increases affinity of the crRNA for a tracrRNA.
      • Embodiment 6. The compound of any of embodiments 1-5, wherein at least one nucleobase of the modified crRNA is thymine.
      • Embodiment 7. The compound of any of embodiments 1-5, wherein at least one nucleobase of the modified crRNA is a modified nucleobase.
      • Embodiment 8. The compound of embodiment 7, wherein the modified nucleobase is 5-methyl cytosine.
      • Embodiment 9. The compound of any of embodiments 1-8, wherein at least one internucleoside linkage of the modified crRNA is a modified internucleoside linkage.
      • Embodiment 10. The compound of embodiment 9, wherein each internucleoside linkage of the modified crRNA is a modified internucleoside linkage.
      • Embodiment 11. The compound of embodiment 9 or 10, wherein at least one modified internucleoside linkage is a neutral internucleoside linkage.
      • Embodiment 12. The compound of embodiment 11, wherein at least one modified internucleoside linkage comprises a methoxypropyl group.
      • Embodiment 13. The compound of any of embodiments 9-12, wherein at least one modified internucleoside linkage comprises a phosphonoacetate.
      • Embodiment 14. The compound of any of embodiments 9-13, wherein at least one modified internucleoside linkage comprises a methylphosphonate.
      • Embodiment 15. The compound of any of embodiments 9-14, wherein at least one modified internucleoside linkage is a phosphorothioate internucleoside linkage.
      • Embodiment 16. The compound of any of embodiments 9-15, wherein at least two linkages of the modified crRNA are modified internucleoside linkages.
      • Embodiment 17. The compound of embodiment 16, wherein at least two modified linkages of the modified crRNA are the same as one another.
      • Embodiment 18. The compound of embodiment 9-17, wherein the modified crRNA comprises two to five phosphorothioate internucleoside linkages at the 5′-end of the crRNA.
      • Embodiment 19. The compound of embodiment 9-18, wherein the modified crRNA comprises two to five phosphorothioate internucleoside linkages at the 3′-end of the crRNA.
      • Embodiment 20. The compound of embodiment 9, wherein each internucleoside linkage of the crRNA is a phosphorothioate internucleoside linkage.
      • Embodiment 21. The compound of any of embodiments 1-20, wherein the modified crRNA does not comprise a 2′-deoxynucleoside.
      • Embodiment 22. The compound of any of embodiments 1-21, wherein at least one nucleoside of the modified crRNA comprises a modified sugar moiety.
      • Embodiment 23. The compound of embodiment 22, wherein the 5′-terminal nucleoside of the crRNA comprises a modified sugar moiety.
      • Embodiment 24. The compound of embodiment 23, wherein the 5′-terminal nucleoside comprises a non-bicyclic 2′-modified sugar moiety
      • Embodiment 25. The compound of embodiment 23, wherein the 5′-terminal nucleoside comprises a bicyclic sugar moiety.
      • Embodiment 26. The compound of embodiment 23, wherein the 5′-terminal nucleoside comprises a modified sugar moiety selected from among: 2′-O-methyl, 2′-MOE, 2′-F, cEt, and LNA.
      • Embodiment 27. The compound of any of embodiments 22-26, wherein the internucleoside at the 5′-end of the crRNA is a phosphorothioate internucleoside linkage.
      • Embodiment 28. The compound of embodiment 22, wherein the modified crRNA has the formula:

  • 5′-NyzNys-R-3′
        • wherein:
        • each Ny is a nucleoside comprising a sugar moiety independently selected from among an unmodified 2′-deoxy sugar moiety, an unmodified 2′-hydroxy sugar moiety, a 2′-O-methyl modified sugar moiety, a 2′-F modified sugar moiety, and a cEt modified sugar moiety;
        • z is a neutral internucleoside linkage selected from among methoxypropyl phosphonate and methyl phosphonate;
        • s is a phosphorothioate internucleoside linkage; and
        • R is the remaining portion of the crRNA.
      • Embodiment 29. The compound of embodiment 22, wherein the modified crRNA has the formula:

  • 5′-NmsNxs-R-3′
        • wherein:
        • Nm is a nucleoside comprising a 2′-O-methyl modified sugar moiety;
        • Nx is a nucleoside comprising a modified sugar moiety selected from among an unmodified 2′-hydroxy sugar moiety and a 2′-F modified sugar moiety;
        • s is a phosphorothioate internucleoside linkage; and
        • R is the remaining portion of the crRNA.
      • Embodiment 30. The compound of any of embodiments 22-29, wherein the 3′-terminal nucleoside of the crRNA comprises a modified sugar moiety.
      • Embodiment 31. The compound of embodiment 30, wherein the 3′-terminal internucleoside linkage of the crRNA is a phosphorothioate internucleoside linkage.
      • Embodiment 32. The compound of embodiment 31, wherein the modified crRNA has the formula:

  • 5′-A-NrsNr-3′
        • wherein:
        • each Nr is a nucleoside comprising a modified sugar moiety independently selected from among:
        • 2′-O-methyl, 2′-MOE, 2′-F, cEt, and LNA;
        • s is a phosphorothioate internucleoside linkage; and
        • A is the remaining portion of the crRNA.
      • Embodiment 33. The compound of embodiment 30, wherein the modified crRNA has the formula:

  • 5′-A-NrzNr-3′
        • wherein:
        • each Nr is a nucleoside comprising a modified sugar moiety independently selected from among:
        • 2′-O-methyl, 2′-MOE, 2′-F, cEt, and LNA;
        • z is a phosphate internucleoside linkage or a neutral internucleoside linkage selected from among methoxypropyl phosphonate and methyl phosphonate;
        • A is the remaining portion of the crRNA;
        • provided that z is not a phosphate internucleoside linkage if the 3′-terminal Nr comprises a 2′-F sugar moiety.
      • Embodiment 34. The compound of any of embodiments 22-33, wherein the DNA recognition portion of the modified crRNA comprises at least 7 modified nucleosides, wherein the modified nucleosides each comprise a modified sugar moiety.
      • Embodiment 35. The compound of embodiment 34, wherein the seven 5′-terminal nucleosides comprise modified sugar moieties.
      • Embodiment 36. The compound of embodiment 35, wherein the modified sugar moieties of the seven 5′-terminal nucleosides are the same as one another.
      • Embodiment 37. The compound of embodiment 34, wherein the modified sugar moieties of the seven 5′-terminal nucleosides are each independently selected from among 2′-O-methyl and 2′-F.
      • Embodiment 38. The compound of embodiment 37, wherein the modified sugar moieties of the seven 5′-terminal nucleosides alternate between 2′-O-methyl and 2′-F.
      • Embodiment 39. The compound of any of embodiments 1-38, wherein the DNA recognition portion of the crRNA comprises at least one nucleoside comprising an unmodified sugar moiety.
      • Embodiment 40. The compound of any of embodiments 1-39, wherein the tracrRNA recognition portion of the modified crRNA comprises at least 4 modified nucleosides, wherein the modified nucleosides each comprise a modified sugar moiety.
      • Embodiment 41. The compound of embodiment 40, wherein each of the modified sugar moieties of the tracrRNA recognition portion are the same as one another.
      • Embodiment 42. The compound of embodiment 40, wherein each modified sugar moiety of the tracrRNA recognition portion is a cEt.
      • Embodiment 43. The compound of any of embodiments 1-42, wherein the tracrRNA recognition portion of the crRNA comprises at least one nucleoside comprising an unmodified sugar moiety.
      • Embodiment 44. The compound of any of embodiments 1-43, wherein the crRNA consists of 42 linked nucleosides.
      • Embodiment 45. The compound of any of embodiments 1-43, wherein the crRNA consists of 20 to 42 linked nucleosides.
      • Embodiment 46. The compound of embodiment 45, wherein the crRNA consists of 29 to 32 linked nucleosides.
      • Embodiment 47. The compound of embodiment 45, wherein the crRNA consists of 32 linked nucleosides.
      • Embodiment 48. The compound of embodiment 45, wherein the crRNA consists of 29 linked nucleosides.
      • Embodiment 49. The compound of embodiment 45, wherein the crRNA consists of 20-28 linked nucleosides.
      • Embodiment 50. The compound of any of embodiments 1-49, wherein the tracrRNA recognition portion of the crRNA consists of 12 or fewer linked nucleosides.
      • Embodiment 51. The compound of any of embodiments 1-50, wherein the DNA recognition portion of the crRNA consists of 17 or fewer linked nucleosides.
      • Embodiment 52. The compound of any of embodiments 1-50, wherein the tracrRNA recognition portion of the crRNA comprises a modification selected from alkyne or azide.
      • Embodiment 53. The compound of any of embodiments 1-52, wherein the compound consists of the crRNA.
      • Embodiment 54. The compound of any of embodiments 1-52, wherein the compound comprises a conjugate group.
      • Embodiment 55. The compound of embodiment 54, wherein the conjugate group comprises GalNAc.
      • Embodiment 56. The compound of any of embodiments 1-55, wherein the nucleobase sequence of the DNA recognition portion of the crRNA is at least 90% complementary to a target DNA.
      • Embodiment 57. The compound of embodiment 56, wherein the nucleobase sequence of the DNA recognition portion of the crRNA is 100% complementary to a target DNA.
      • Embodiment 58. A method comprising contacting a cell with the compound of any of embodiments 1-57.
      • Embodiment 59. The method of embodiment 58, wherein the cell expresses Cas9.
      • Embodiment 60. A method comprising contacting a cell with the compound of any of embodiments 1-57 and a plasmid that encodes a Cas9 gene.
      • Embodiment 61. A method comprising contacting a cell with the compound of any of embodiments 1-57 and an mRNA that encodes Cas9.
      • Embodiment 62. A method comprising contacting a cell with the compound of any of embodiments 1-57 and a plasmid that encodes a Cas9 gene and a tracrRNA.
      • Embodiment 63. A method comprising contacting a cell with compound of any of embodiments 1-57, a plasmid that encodes a Cas9 gene, and a tracrRNA.
      • Embodiment 64. The method of any of embodiments 58-63, wherein the crRNA consists of 20 to 32 nucleosides.
      • Embodiment 65. The method of any of embodiments 58-64, wherein the crRNA is taken up by the cell in the absence of a transfection reagent.
      • Embodiment 66. A method comprising contacting a cell with the modified crRNA of embodiment 52 and a tracrRNA comprising a modification selected from among: alkyne and azide.
      • Embodiment 67. The method of embodiment 66 comprising contacting the cell with a plasmid that encodes a Cas9 gene.
      • Embodiment 68. The method of embodiment 66, wherein the cell expresses Cas9.
      • Embodiment 69. The method of any of embodiments 58-68, wherein the cell is in an animal.
      • Embodiment 70. A method comprising administering to an animal the modified compound of any of embodiments 1-57.
      • Embodiment 71. The method of embodiment 70, wherein the administration is subcutaneous.
      • Embodiment 72. The method of embodiment 70, wherein the administration is intrathecal.
      • Embodiment 73. The method of any of embodiments 70-72 comprising administering a plasmid that encodes a Cas9 gene.
      • Embodiment 74. The method of any of embodiments 70-72 wherein the animal expresses Cas9.
      • Embodiment 75. The method of any of embodiments 70-72 comprising administering a plasmid that encodes a Cas9 gene and a tracrRNA.
      • Embodiment 76. The method of embodiment 75, wherein the plasmid is delivered to cells within the animal via an adeno-associated virus (AAV).
      • Embodiment 77. The method of embodiment 75, wherein the plasmid is delivered to cells within the animal via a lentivirus.
      • Embodiment 78. The method of any of embodiments 70-77, wherein a target gene is edited.
      • Embodiment 79. The method of embodiment 78, wherein the crRNA is degraded after the target gene is edited.
      • Embodiment 80. The method of embodiment 79, wherein the Cas9 does not exhibit nuclease activity in the absence of the crRNA.
      • Embodiment 81. The compound of embodiment 5, wherein the tracrRNA is unmodified.
      • Embodiment 82. The compound of embodiment 5, wherein the tracrRNA is modified.
      • Embodiment 83. The compound of embodiment 34, wherein the ten 5′-terminal nucleosides comprise modified sugar moieties.
      • Embodiment 84. The compound of embodiment 83, wherein the modified sugar moieties of the ten 5′-terminal nucleosides are the same as one another.
      • Embodiment 85. The compound of embodiment 83, wherein the modified sugar moieties of the ten 5′-terminal nucleosides are each independently selected from among 2′-F and 2′-O-methyl.
      • Embodiment 86. The compound of embodiment 84, wherein the modified sugar moieties of the ten 5′-terminal nucleosides are 2′-F.
      • Embodiment 87. The compound of embodiment 4, wherein the crRNA motif is selected from among the motifs listed in Table A.
      • Embodiment 88. The compound of any of embodiments 40 or 81-87, wherein the at least four modified nucleosides of the tracrRNA recognition portion are the four 3′-terminal nucleosides of the crRNA.
      • Embodiment 89. The compound of embodiment 88, wherein the at least four modified nucleosides of the tracrRNA recognition portion comprise 2′-O-methyl modified sugar moieties.
      • Embodiment 90. The compound of any of embodiments 40 or 81-89, wherein the tracrRNA recognition portion comprises five modified nucleosides.
      • Embodiment 91. The compound of any of embodiments 40 or 81-89, wherein the tracrRNA recognition portion comprises six modified nucleosides.
      • Embodiment 92. The compound of any of embodiments 40 or 81-89, wherein the tracrRNA recognition portion comprises at least seven modified nucleosides.
      • Embodiment 93. The compound of any of embodiments 40, 81-86, or 88-89, wherein the tracrRNA recognition portion comprises nine modified nucleosides.
      • Embodiment 94. The compound of any of embodiments 40 or 81-93, wherein at least one modified sugar moiety of the tracrRNA recognition portion is a bicyclic sugar moiety.
      • Embodiment 95. The compound of embodiment 94, wherein the two 3′-terminal nucleosides of the tracrRNA recognition portion comprise bicyclic sugar moieties.
      • Embodiment 96. The compound of embodiment 95, wherein the tracrRNA recognition portion comprises five bicyclic sugar moieties.
      • Embodiment 97. The compound of embodiment 95, wherein the tracrRNA recognition portion comprises six bicyclic sugar moieties.
      • Embodiment 98. The compound of embodiment 93, wherein the tracrRNA recognition portion comprises nine bicyclic sugar moieties.
      • Embodiment 99. The compound of any of embodiments 94-98, wherein each bicyclic sugar moiety is independently selected from among cEt and LNA.
      • Embodiment 100. The compound of embodiment 99, wherein each bicyclic sugar moiety is cEt.
      • Embodiment 101. The compound of any of embodiments 40 or 88-100, wherein the nucleoside at the 5′-end of the tracrRNA recognition portion of the crRNA comprises a modified sugar moiety.
      • Embodiment 102. The compound of embodiment 101, wherein the nucleoside at the 5′-end of the tracrRNA recognition portion of the crRNA comprises a bicyclic sugar moiety.
      • Embodiment 103. The compound of embodiment 102, wherein the bicyclic sugar moiety is cEt or LNA.
      • Embodiment 104. The compound of embodiment 103, wherein the bicyclic sugar moiety is cEt.
      • Embodiment 105. The compound of any of embodiments 81-104, wherein the DNA recognition portion of the crRNA comprises at least one nucleoside comprising an unmodified sugar moiety.
      • Embodiment 106. The compound of embodiment 88, wherein each of the modified sugar moieties of the tracrRNA recognition portion are the same as one another.
      • Embodiment 107. The compound of any of embodiments 81-106, wherein the tracrRNA recognition portion of the crRNA comprises at least one nucleoside comprising an unmodified sugar moiety.
      • Embodiment 108. The compound of any of embodiments 81-107, wherein the crRNA consists of 42 linked nucleosides.
      • Embodiment 109. The compound of any of embodiments 81-107, wherein the crRNA consists of 20 to 42 linked nucleosides.
      • Embodiment 110. The compound of embodiment 109, wherein the crRNA consists of 29 to 32 linked nucleosides.
      • Embodiment 111. The compound of any of embodiments 81-86 or 88-109, wherein the crRNA consists of 32 linked nucleosides.
      • Embodiment 112. The compound of embodiment 109, wherein the crRNA consists of 29 linked nucleosides.
      • Embodiment 113. The compound of any of embodiments 81-86 or 88-109, wherein the crRNA consists of 20-28 linked nucleosides.
      • Embodiment 114. The compound of any of embodiments 81-113, wherein the tracrRNA recognition portion of the crRNA consists of 12 or fewer linked nucleosides.
      • Embodiment 115. The compound of any of embodiments 81-114, wherein the DNA recognition portion of the crRNA consists of 17 or fewer linked nucleosides.
      • Embodiment 116. The compound of any of embodiments 81-115, wherein the tracrRNA recognition portion of the crRNA comprises a modification selected from alkyne or azide.
      • Embodiment 117. The compound of any of embodiments 81-116, wherein the compound consists of the crRNA.
      • Embodiment 118. The compound of any of embodiments 81-116, wherein the compound comprises a conjugate group.
      • Embodiment 119. The compound of embodiment 118, wherein the conjugate group comprises GalNAc.
      • Embodiment 120. The compound of embodiment 54 or 118, wherein the conjugate group is lipophilic.
      • Embodiment 121. The compound of any of embodiments 81-120, wherein the nucleobase sequence of the DNA recognition portion of the crRNA is at least 90% complementary to a target DNA.
      • Embodiment 122. The compound of embodiment 121, wherein the nucleobase sequence of the DNA recognition portion of the crRNA is 100% complementary to a target DNA.
      • Embodiment 123. A method comprising contacting a cell with the compound of any of embodiments 81-122.
      • Embodiment 124. The method of embodiment 123, wherein the cell expresses Cas9.
      • Embodiment 125. A method comprising contacting a cell with the compound of any of embodiments 81-122 and a plasmid that encodes a Cas9 gene.
      • Embodiment 126. A method comprising contacting a cell with the compound of any of embodiments 81-122 and an mRNA that encodes Cas9.
      • Embodiment 127. A method comprising contacting a cell with the compound of any of embodiments 81-122 and a plasmid that encodes a Cas9 gene and a tracrRNA.
      • Embodiment 128. A method comprising contacting a cell with the compound of any of embodiments 81-122, a plasmid that encodes a Cas9 gene, and a tracrRNA.
      • Embodiment 129. The method of any of embodiments 123-128, wherein the crRNA is taken up by the cell in the absence of a transfection reagent.
      • Embodiment 130. The method of any of embodiments 123-129, wherein the cell is in an animal.
      • Embodiment 131. A method comprising administering to an animal the modified compound of any of embodiments 81-122.
      • Embodiment 132. The method of embodiment 131, wherein the administration is subcutaneous.
      • Embodiment 133. The method of embodiment 131, wherein the administration is intrathecal.
      • Embodiment 134. The method of embodiment 70 or 131, wherein the administration is to the central nervous system.
      • Embodiment 135. The method of any of embodiments 131-134 comprising administering a plasmid that encodes a Cas9 gene.
      • Embodiment 136. The method of any of embodiments 131-134 wherein the animal expresses Cas9.
      • Embodiment 137. The method of any of embodiments 131-134 comprising administering a plasmid that encodes a Cas9 gene and a tracrRNA.
      • Embodiment 138. The method of embodiment 135 or 137, wherein the plasmid is delivered to cells within the animal via an adeno-associated virus (AAV).
      • Embodiment 139. The method of embodiment 135 or 137, wherein the plasmid is delivered to cells within the animal via a lentivirus.
      • Embodiment 140. The method of any of embodiments 131-139, wherein a target gene is edited.
      • Embodiment 141. The method of embodiment 140, wherein the crRNA is degraded after the target gene is edited.
      • Embodiment 142. The method of embodiment 141, wherein the Cas9 does not exhibit nuclease activity in the absence of the crRNA.
      • Embodiment 143. The method of any of embodiments 69-80 or 130-142, wherein the animal is a human.
      • Embodiment 144. A method comprising contacting a cell with the compound of any of embodiments 1-57 or 81-122, editing a target gene, and contacting the cell with a second compound that degrades or inhibits the activity or expression of the crRNA, a tracrRNA, or a Cas9 nuclease.
      • Embodiment 145. The method of embodiment 144, wherein the cell is contacted with the second compound after the target gene has been edited.
      • Embodiment 146. The method of embodiment 144 or 145, wherein the second compound comprises an oligonucleotide that is complementary to the crRNA.
      • Embodiment 147. The method of embodiment 146, wherein the crRNA is degraded.
      • Embodiment 148. The method of embodiment 144 or 145, wherein the second compound comprises an oligonucleotide that is complementary to the tracrRNA.
      • Embodiment 149. The method of embodiment 148, wherein the tracrRNA is degraded.
      • Embodiment 150. The method of embodiment 144 or 145, wherein the second compound comprises a crRNA that targets the Cas9 nuclease gene.
      • Embodiment 151. The method of embodiment 144 or 145, wherein the second compound comprises an oligonucleotide that is complementary to the Cas9 transcript.
      • Embodiment 152. The method of embodiment 150 or 151, wherein the expression of the Cas9 nuclease is inhibited.
      • Embodiment 153. The method of any of embodiments 144-152, wherein the cell is in an animal.
      • Embodiment 154. The method of embodiment 153, wherein the animal is a human.
      • Embodiment 155. The method of embodiment 63 or 128, wherein the tracrRNA is unmodified.
      • Embodiment 156. The method of embodiment 63 or 128, wherein the tracrRNA is modified.
      • Embodiment 157. The method of embodiment 63, 128, or 155-156, wherein both the crRNA and the tracrRNA are taken up by the cell in the absence of a transfection reagent.
      • Embodiment 158. The method of any of embodiments 155-157, wherein the cell is in an animal.
      • Embodiment 159. The method of embodiment 158, wherein the animal is a human.
      • Embodiment 160. A method of genomic loci visualization comprising contacting a genome with a compound of any of embodiments 1-57 or 81-122.
      • Embodiment 161. The method of any of embodiments 58-80 or 123-160, wherein editing of off-target genes is reduced relative to editing of off-target genes when unmodified crRNA or a compound comprising more than 50 nucleosides is used in place of the compound comprising the modified crRNA consisting of 20-50 linked nucleosides.
      • Embodiment 162. A compound comprising a modified scrRNA consisting of 20-50 linked nucleosides.
      • Embodiment 163. The compound of embodiment 162, wherein the modified scrRNA is 5′-stabilized.
      • Embodiment 164. The compound of embodiment 162 or 163, wherein the modified scrRNA is 3′-stabilized.
      • Embodiment 165. The compound of any of embodiments 162-164, wherein the modified scrRNA comprises at least one modification that increases affinity of the scrRNA for a scrRNA target DNA.
      • Embodiment 166. The compound of any of embodiments 161-165, wherein the modified scrRNA comprises at least one modification that increases affinity of the scrRNA for a nuclease
      • Embodiment 167. The compound of embodiment 166, wherein the nuclease is a Cpf1 nuclease.
      • Embodiment 168. The compound of any of embodiments 161-167, wherein at least one nucleobase of the modified scrRNA is thymine.
      • Embodiment 169. The compound of any of embodiments 161-168, wherein at least one nucleobase of the modified scrRNA is a modified nucleobase.
      • Embodiment 170. The compound of embodiment 169, wherein the modified nucleobase is 5-methyl cytosine.
      • Embodiment 171. The compound of any of embodiments 161-170, wherein at least one internucleoside linkage of the modified scrRNA is a modified internucleoside linkage.
      • Embodiment 172. The compound of embodiment 171, wherein each internucleoside linkage of the modified scrRNA is a modified internucleoside linkage.
      • Embodiment 173. The compound of embodiment 171 or 172, wherein at least one modified internucleoside linkage is a neutral internucleoside linkage.
      • Embodiment 174. The compound of embodiment 173, wherein at least one modified internucleoside linkage comprises a methoxypropyl group.
      • Embodiment 175. The compound of any of embodiments 171-174, wherein at least one modified internucleoside linkage comprises a phosphonoacetate.
      • Embodiment 176. The compound of any of embodiments 171-175, wherein at least one modified internucleoside linkage comprises a methylphosphonate.
      • Embodiment 177. The compound of any of embodiments 171-176, wherein at least one modified internucleoside linkage is a phosphorothioate internucleoside linkage.
      • Embodiment 178. The compound of any of embodiments 171-177, wherein at least two linkages of the modified scrRNA are modified internucleoside linkages.
      • Embodiment 179. The compound of embodiment 178, wherein at least two modified linkages of the modified scrRNA are the same as one another.
      • Embodiment 180. The compound of any of embodiments 171-179, wherein the modified scrRNA comprises two to five phosphorothioate internucleoside linkages at the 5′-end of the scrRNA.
      • Embodiment 181. The compound of any of embodiments 171-180, wherein the modified scrRNA comprises two to five phosphorothioate internucleoside linkages at the 3′-end of the scrRNA.
      • Embodiment 182. The compound of embodiment 171, wherein each internucleoside linkage of the scrRNA is a phosphorothioate internucleoside linkage.
      • Embodiment 183. The compound of any of embodiments 161-182, wherein the modified scrRNA does not comprise a 2′-deoxynucleoside.
      • Embodiment 184. The compound of any of embodiments 161-183, wherein at least one nucleoside of the modified scrRNA comprises a modified sugar moiety.
      • Embodiment 185. The compound of embodiment 184, wherein the 5′-terminal nucleoside of the scrRNA comprises a modified sugar moiety.
      • Embodiment 186. The compound of embodiment 185, wherein the 5′-terminal nucleoside comprises a non-bicyclic 2′-modified sugar moiety
      • Embodiment 187. The compound of embodiment 185, wherein the 5′-terminal nucleoside comprises a bicyclic sugar moiety.
      • Embodiment 188. The compound of embodiment 185, wherein the 5′-terminal nucleoside comprises a modified sugar moiety selected from among: 2′-O-methyl, 2′-MOE, 2′-F, cEt, and LNA.
      • Embodiment 189. The compound of any of embodiments 184-188, wherein the internucleoside linkage at the 5′-end of the scrRNA is a phosphorothioate internucleoside linkage.
      • Embodiment 190. The compound of embodiment 184, wherein the modified scrRNA has the formula:

  • 5′-NyzNys-R-3′
        • wherein:
        • each Ny is a nucleoside comprising a sugar moiety independently selected from among an unmodified 2′-deoxy sugar moiety, an unmodified 2′-hydroxy sugar moiety, a 2′-O-methyl modified sugar moiety, a 2′-F modified sugar moiety, and a cEt modified sugar moiety;
        • z is a neutral internucleoside linkage selected from among methoxypropyl phosphonate and methyl phosphonate;
        • s is a phosphorothioate internucleoside linkage; and
        • R is the remaining portion of the scrRNA.
      • Embodiment 191. The compound of embodiment 184, wherein the modified scrRNA has the formula:

  • 5′-NmsNxs-R-3′
        • wherein:
        • Nm is a nucleoside comprising a 2′-O-methyl modified sugar moiety;
        • Nx is a nucleoside comprising a modified sugar moiety selected from among an unmodified 2′-hydroxy sugar moiety and a 2′-F modified sugar moiety;
        • s is a phosphorothioate internucleoside linkage; and
        • R is the remaining portion of the scrRNA.
      • Embodiment 192. The compound of any of embodiments 184-191, wherein the 3′-terminal nucleoside of the scrRNA comprises a modified sugar moiety.
      • Embodiment 193. The compound of embodiment 192, wherein the 3′-terminal internucleoside linkage of the scrRNA is a phosphorothioate internucleoside linkage.
      • Embodiment 194. The compound of embodiment 193, wherein the modified scrRNA has the formula:

  • 5′-A-NrsNr-3′
        • wherein:
        • each Nr is a nucleoside comprising a modified sugar moiety independently selected from among:
        • 2′-O-methyl, 2′-MOE, 2′-F, cEt, and LNA;
        • s is a phosphorothioate internucleoside linkage; and
        • A is the remaining portion of the scrRNA.
      • Embodiment 195. The compound of embodiment 192, wherein the modified scrRNA has the formula:

  • 5′-A-NrzNr-3′
        • wherein:
        • each Nr is a nucleoside comprising a modified sugar moiety independently selected from among:
        • 2′-O-methyl, 2′-MOE, 2′-F, cEt, and LNA;
        • z is a phosphate internucleoside linkage or a neutral internucleoside linkage selected from among methoxypropyl phosphonate and methyl phosphonate;
        • A is the remaining portion of the scrRNA;
        • provided that z is not a phosphate internucleoside linkage if the 3′-terminal Nr comprises a 2′-F sugar moiety.
      • Embodiment 196. The compound of any of embodiments 184-195, wherein the scrRNA target recognition portion of the modified scrRNA comprises at least 7 modified nucleosides, wherein the modified nucleosides each comprise a modified sugar moiety.
      • Embodiment 197. The compound of embodiment 196, wherein the seven 3′-terminal nucleosides comprise modified sugar moieties.
      • Embodiment 198. The compound of embodiment 196, wherein the ten 3′-terminal nucleosides comprise modified sugar moieties.
      • Embodiment 199. The compound of embodiment 197 or 198, wherein the modified sugar moieties of the 3′-terminal nucleosides are the same as one another.
      • Embodiment 200. The compound of embodiment 197 or 198, wherein the modified sugar moieties of the 3′-terminal nucleosides are each independently selected from among 2′-O-methyl and 2′-F.
      • Embodiment 201. The compound of embodiment 200, wherein the modified sugar moieties of the 3′-terminal nucleosides alternate between 2′-O-methyl and 2′-F.
      • Embodiment 202. The compound of any of embodiments 161-201, wherein the scrRNA target recognition portion of the scrRNA comprises at least one nucleoside comprising an unmodified sugar moiety.
      • Embodiment 203. The compound of any of embodiments 161-202, wherein the nuclease recognition portion of the modified scrRNA comprises at least 4 modified nucleosides, wherein the modified nucleosides each comprise a modified sugar moiety.
      • Embodiment 204. The compound of embodiment 203, wherein the four modified nucleosides of the nuclease recognition portion are the four 5′-terminal nucleosides of the scrRNA.
      • Embodiment 205. The compound of embodiment 203 or 204, wherein each of the modified sugar moieties of the nuclease recognition portion is the same as one another.
      • Embodiment 206. The compound of embodiment 205, wherein each modified sugar moiety of the nuclease recognition portion is a cEt or an LNA.
      • Embodiment 207. The compound of any of embodiments 203-205, wherein the at least four modified nucleosides each comprise a 2′-O-methyl modified sugar moiety.
      • Embodiment 208. The compound of any of embodiments 161-207, wherein the nuclease recognition portion of the scrRNA comprises at least one nucleoside comprising an unmodified sugar moiety.
      • Embodiment 209. The compound of any of embodiments 161-208, wherein the nuclease recognition portion comprises five modified nucleosides.
      • Embodiment 210. The compound of any of embodiments 161-208, wherein the nuclease recognition portion comprises six modified nucleosides.
      • Embodiment 211. The compound of any of embodiments 161-208, wherein the nuclease recognition portion comprises at least seven modified nucleosides.
      • Embodiment 212. The compound of any of embodiments 161-208, wherein the nuclease recognition portion comprises nine modified nucleosides.
      • Embodiment 213. The compound of any of embodiments 161-212, wherein at least one modified sugar moiety of the nuclease recognition portion is a bicyclic sugar moiety.
      • Embodiment 214. The compound of embodiment 213, wherein the two 5′-terminal nucleosides of the nuclease recognition portion comprise bicyclic sugar moieties.
      • Embodiment 215. The compound of embodiment 214, wherein the nuclease recognition portion comprises five bicyclic sugar moieties.
      • Embodiment 216. The compound of embodiment 214, wherein the nuclease recognition portion comprises six bicyclic sugar moieties.
      • Embodiment 217. The compound of embodiment 214, wherein the nuclease recognition portion comprises nine bicyclic sugar moieties.
      • Embodiment 218. The compound of any of embodiments 213-217, wherein each bicyclic sugar moiety is independently selected from among cEt and LNA.
      • Embodiment 219. The compound of embodiment 218, wherein each bicyclic sugar moiety is a cEt.
      • Embodiment 220. The compound of any of embodiments 161-219, wherein the scrRNA consists of 42 linked nucleosides.
      • Embodiment 221. The compound of any of embodiments 161-219, wherein the scrRNA consists of 20 to 42 linked nucleosides.
      • Embodiment 222. The compound of embodiment 221, wherein the scrRNA consists of 29 to 32 linked nucleosides.
      • Embodiment 223. The compound of embodiment 221, wherein the scrRNA consists of 32 linked nucleosides.
      • Embodiment 224. The compound of embodiment 221, wherein the scrRNA consists of 29 linked nucleosides.
      • Embodiment 225. The compound of embodiment 221, wherein the scrRNA consists of 20-28 linked nucleosides.
      • Embodiment 226. The compound of any of embodiments 161-225, wherein the nuclease recognition portion of the scrRNA consists of 17 or fewer linked nucleosides.
      • Embodiment 227. The compound of any of embodiments 161-226, wherein the scrRNA target recognition portion of the scrRNA consists of 17 or fewer linked nucleosides.
      • Embodiment 228. The compound of any of embodiments 161-227, wherein the compound consists of the scrRNA.
      • Embodiment 229. The compound of any of embodiments 161-227, wherein the compound comprises a conjugate group.
      • Embodiment 230. The compound of embodiment 229, wherein the conjugate group comprises GalNAc.
      • Embodiment 231. The compound of embodiment 229, wherein the conjugate group comprises a lipophilic group.
      • Embodiment 232. The compound of any of embodiments 161-231, wherein the nucleobase sequence of the scrRNA target recognition portion of the scrRNA is at least 90% complementary to a scrRNA target DNA.
      • Embodiment 233. The compound of embodiment 232, wherein the nucleobase sequence of the scrRNA target recognition portion of the scrRNA is 100% complementary to a scrRNA target DNA.
      • Embodiment 234. The compound of any of embodiments 161-233, wherein the scrRNA comprises a self-complementary region.
      • Embodiment 235. The compound of embodiment 234, wherein the self-complementary region is within the nuclease recognition portion of the scrRNA.
      • Embodiment 236. The compound of embodiment 234 or 235, wherein the self-complementary region can form a hairpin.
      • Embodiment 237. The compound of any of embodiments 234-236, wherein the self-complementary region of the scrRNA comprises at least one modification that increases the stability of the self-complementary region.
      • Embodiment 238. The compound of any of embodiments 234-237, wherein the self-complementary region of the scrRNA comprises at least one modification that increases the hybridization affinity of the self-complementary region.
      • Embodiment 239. A method comprising contacting a cell with the compound of any of embodiments 161-238.
      • Embodiment 240. The method of embodiment 239, wherein the cell expresses a Cpf1 nuclease.
      • Embodiment 241. A method comprising contacting a cell with the compound of any of embodiments 161-238 and a plasmid that encodes a nuclease gene.
      • Embodiment 242. A method comprising contacting a cell with the compound of any of embodiments 161-238 and an mRNA that encodes a nuclease.
      • Embodiment 243. The method of embodiment 241 or 242, wherein the nuclease is a Cpf1 nuclease.
      • Embodiment 244. The method of any of embodiments 239-243, wherein the scrRNA is taken up by the cell in the absence of a transfection reagent.
      • Embodiment 245. The method of any of embodiments 239-244, wherein the cell is in an animal.
      • Embodiment 246. A method comprising administering to an animal the modified compound of any of embodiments 161-238.
      • Embodiment 247. The method of embodiment 246, wherein the administration is subcutaneous.
      • Embodiment 248. The method of embodiment 246, wherein the administration is intrathecal.
      • Embodiment 249. The method of embodiment 246, wherein the administration is to the central nervous system.
      • Embodiment 250. The method of any of embodiments 246-249 comprising administering a plasmid that encodes a nuclease gene.
      • Embodiment 251. The method of any of embodiments 246-249 wherein the animal expresses a nuclease that is recognized by the nuclease recognition portion of the scrRNA.
      • Embodiment 252. The method of any of embodiments 246-249 comprising administering a plasmid that encodes a nuclease gene.
      • Embodiment 253. The method of embodiment 250 or 252, wherein the plasmid is delivered to cells within the animal via an adeno-associated virus (AAV).
      • Embodiment 254. The method of embodiment 250 or 252, wherein the plasmid is delivered to cells within the animal via a lentivirus.
      • Embodiment 255. The method of any of embodiments 250-254, wherein the nuclease is a Cpf1 nuclease.
      • Embodiment 256. The method of any of embodiments 239-255, wherein a scrRNA target gene is altered.
      • Embodiment 257. The method of embodiment 256, wherein the scrRNA is degraded after the scrRNA target gene is altered.
      • Embodiment 258. The method of embodiment 257, wherein the nuclease that is recognized by the nuclease recognition portion of the scrRNA does not exhibit nuclease activity in the absence of the scrRNA.
      • Embodiment 259. The method of any of embodiments 245-258, wherein the animal is a human.
      • Embodiment 260. A method comprising contacting a cell with the compound of any of embodiments 161-238, altering a scrRNA target gene, and contacting the cell with a second compound that degrades or inhibits the activity or expression of the scrRNA or a nuclease.
      • Embodiment 261. The method of embodiment 260, wherein the nuclease is a Cpf1 nuclease.
      • Embodiment 262. The method of embodiment 260 or 261, wherein the cell is contacted with the second compound after the scrRNA target gene has been altered.
      • Embodiment 263. The method of any of embodiments 260-262, wherein the second compound comprises an oligonucleotide that is complementary to the scrRNA.
      • Embodiment 264. The method of embodiment 263, wherein the scrRNA is degraded.
      • Embodiment 265. The method of any of embodiments 260-262, wherein the second compound comprises a scrRNA that targets the nuclease gene.
      • Embodiment 266. The method of any of embodiments 260-262, wherein the second compound comprises an oligonucleotide that is complementary to the nuclease transcript.
      • Embodiment 267. The method of embodiment 265 or 266, wherein the expression of the nuclease is inhibited.
      • Embodiment 268. The method of any of embodiments 260-267, wherein the cell is in an animal.
      • Embodiment 269. The method of embodiment 268, wherein the animal is a human.
      • Embodiment 270. A method of genomic loci visualization comprising contacting a genome with a compound of any of embodiments 161-238.
      • Embodiment 271. The method of any of embodiments 239-269, wherein alteration of off-target genes is reduced relative to alteration of off-target genes when unmodified scrRNA or a compound comprising more than 50 nucleosides is used in place of the compound comprising the modified scrRNA consisting of 20-50 linked nucleosides.
      • Embodiment 272. The compound of any of embodiments 1-57 or 81-122, wherein the sequence of the tracrRNA recognition portion of the crRNA comprises at least 12 contiguous nucleobases of a sequence selected from among SEQ ID Numbers 19, 20, 21, 22, 23, 24, and 25.
      • Embodiment 273. The compound of any of embodiments 1-57 or 81-122, wherein the sequence of the tracrRNA recognition portion of the crRNA comprises the first 12 nucleobases of a sequence selected from among SEQ ID Numbers 19, 20, 21, 22, 23, 24, and 25.
      • Embodiment 274. The compound of any of embodiments 1-57 or 81-122, wherein the sequence of the tracrRNA recognition portion of the crRNA consists of the first 12 nucleobases of a sequence selected from among SEQ ID Numbers 19, 20, 21, 22, 23, 24, and 25.
      • Embodiment 275. The compound of any of embodiments 162-238, wherein the sequence of the nuclease recognition portion of the scrRNA comprises the sequence UCUACU.
      • Embodiment 276. The compound of any of embodiments 162-238, wherein the sequence of the nuclease recognition portion of the scrRNA comprises the sequence GUAGAU.
      • Embodiment 277. The compound of any of embodiments 162-238, wherein the sequence of the nuclease recognition portion of the scrRNA comprises the sequence UCUACU and the sequence GUAGAU.
      • Embodiment 278. The compound of any of embodiments 162-238, wherein the sequence of the nuclease recognition portion of the scrRNA comprises at least 12 nucleobases of a sequence selected from among SEQ ID Numbers 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, and 39.
      • Embodiment 279. The compound of any of embodiments 1-57, 81-86, 88-122, 162-238, or 272-278, wherein the DNA recognition portion comprises 7-9 2′-modified sugar moieties.
      • Embodiment 280. The compound of embodiment 279, wherein the 7-9 2′-modified sugar moieties are 2′-F modified sugar moieties.
      • Embodiment 281. The compound of any of embodiments 279 or 280, wherein the tracrRNA recognition portion or the nuclease recognition portion comprises 5-6 bicyclic sugar moieties.
      • Embodiment 282. The compound of embodiment 281, wherein the 5-6 bicyclic sugar moieties are cEt.
      • Embodiment 283. A pharmaceutical composition comprising the compound of any of embodiments 1-57, 81-122, 162-238, or 272-283.
      • Embodiment 284. The method of any of embodiments 70, 131, or 246, wherein the administration is intravitreal.
      • Embodiment 285. The method of any of embodiments 58-68, 123-129, 144-152, 155-157, 239-244, or 260-267, wherein the cell is a plant cell.
      • Embodiment 286. The method of any of embodiments 58-68, 123-129, 144-152, 155-157, 239-244, or 260-267, wherein the cell is an animal cell.
      • Embodiment 287. The method of any of embodiments 58-68, 123-129, 144-152, 155-157, 239-244, or 260-267, wherein the cell is a T-cell.
      • Embodiment 288. A method of treating a disease in an individual comprising administering the compound of any of embodiments 1-57, 81-122, 162-238, or 272-282, or the composition of embodiment 283 to the individual, thereby treating the disease in the individual.
      • Embodiment 289. Use of the compound of any of embodiments 1-57, 81-122, 162-238, or 272-282 or the composition of embodiment 283 for the treatment of a disease.
      • Embodiment 290. Use of the compound of any of embodiments 1-57, 81-122, 162-238, or 272-282 for preparation of a medicament.
      • Embodiment 291. A method of administering the compound of any of embodiments 1-57, 81-122, 162-238, or 272-282 or the composition of embodiment 283 to an animal, and harvesting an organ from the animal for transplantation into a human.
    BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a gel illustrating the extent of gene editing of hLDLR.
  • FIG. 2 is a gel illustrating the extent of gene editing of hVEGFA.
  • FIG. 3 is a gel illustrating the extent of gene editing of hVEGFA using crRNAs, including shortened modified crRNAs.
  • FIGS. 4 a and 4 b are gels that show the effect of truncated scrRNAs comprising a scrRNA target recognition portion that is complementary to DNA (cytosine-5)-methyltransferase 1 (DNMT1) on alteration of the DNMT1 gene. FIGS. 4 a and 4 b show that multiple truncated scrRNAs, including scrRNA containing only 36 nucleosides, altered the DNMT1 gene.
  • FIG. 5 is a gel that shows the extent of activity of truncated tracrRNAs designed and synthesized to edit mouse Proprotein Convertase Subtilisin/Kexin Type 9 (Pcsk9).
  • FIG. 6 is a gel that shows the DNA cutting activity of conjugated and unconjugated modified crRNA targeted to Pcsk9.
  • FIG. 7 is a gel that shows that a modified crRNA disrupted the Pcsk9 gene with similar potency to a sgRNA positive control in hepatocytes ex vivo.
  • DETAILED DESCRIPTION
  • It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed. Herein, the use of the singular includes the plural unless specifically stated otherwise. As used herein, the use of “or” means “and/or” unless stated otherwise. Furthermore, the use of the term “including” as well as other forms, such as “includes” and “included”, is not limiting. Also, terms such as “element” or “component” encompass both elements and components comprising one unit and elements and components that comprise more than one subunit, unless specifically stated otherwise.
  • The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including, but not limited to, patents, patent applications, articles, books, and treatises, are hereby expressly incorporated by reference in their entirety for any purpose.
  • Definitions
  • Unless otherwise indicated, the following terms have the following meanings:
  • As used herein, “2′-deoxynucleoside” means a nucleoside comprising 2′-H(H) furanosyl sugar moiety, as found in naturally occurring deoxyribonucleic acids (DNA). In certain embodiments, a 2′-deoxynucleoside may comprise a modified nucleobase or may comprise an RNA nucleobase (e.g., uracil).
  • As used herein, “2′-substituted nucleoside” or “2-modified nucleoside” means a nucleoside comprising a 2′-substituted or 2′-modified sugar moiety. As used herein, “2′-substituted” or “2-modified” in reference to a sugar moiety means a furanosyl sugar moiety comprising a 2′-substituent group other than H or OH.
  • As used here, “3′-stabilized” in reference to a modified oligonucleotide means a modified oligonucleotide comprising a modification or modifications at the 3′-terminus that increase the stability of the oligonucleotide in cells or in an animal relative to a corresponding oligonucleotide that does not comprise the modification or modifications at the 3′-terminus.
  • As used here, “5′-stabilized” in reference to a modified oligonucleotide means a modified oligonucleotide comprising a modification or modifications at the 5′-terminus that increase the stability of the oligonucleotide in cells or in an animal relative to a corresponding oligonucleotide that does not comprise the modification or modifications at the 5′-terminus.
  • As used herein, “bicyclic nucleoside” or “BNA” means a nucleoside comprising a bicyclic sugar moiety. As used herein, “bicyclic sugar” or “bicyclic sugar moiety” means a modified sugar moiety comprising two rings, wherein the second ring is formed via a bridge connecting two of the atoms in the first ring thereby forming a bicyclic structure. In certain embodiments, the first ring of the bicyclic sugar moiety is a furanosyl moiety. In certain embodiments, the bicyclic sugar moiety does not comprise a furanosyl moiety.
  • As used herein, “Cas9” means a nuclease that recognizes and/or cleaves target DNA when in a complex with crRNA and tracrRNA. In certain embodiments, Cas9 is derived from S. pyogenes. In certain embodiments, Cas9 is derived from S. aureus.
  • As used herein, “cell-targeting moiety” means a conjugate group or portion of a conjugate group that is capable of binding to a particular cell type or particular cell types.
  • As used herein, “complementary” in reference to an oligonucleotide means the nucleobase sequence of such oligonucleotide or one or more regions thereof matches the nucleobase sequence of another oligonucleotide or nucleic acid or one or more regions thereof when the two nucleobase sequences are aligned in opposing directions. Nucleobase matches or complementary nucleobases, as described herein, are limited to adenine (A) and thymine (T), adenine (A) and uracil (U), cytosine (C) and guanine (G), and 5-methyl cytosine (mC) and guanine (G) unless otherwise specified. Complementary oligonucleotides and/or nucleic acids need not have nucleobase complementarity at each nucleoside. Rather, some mismatches are tolerated. As used herein, “fully complementary” or “100% complementary” in reference to oligonucleotides means that such oligonucleotides are complementary to another oligonucleotide or nucleic acid at each nucleoside. In such embodiments, mismatches are not tolerated.
  • As used herein, “conjugate group” means a group of atoms that is directly or indirectly attached to a parent compound, e.g., an oligonucleotide.
  • As used herein, “conjugate linker” means a group of atoms that connects a conjugate group to a parent compound, e.g., an oligonucleotide.
  • As used herein, “contiguous” in the context of an oligonucleotide refers to nucleosides, nucleobases, sugar moieties, or internucleoside linkages that are immediately adjacent to each other. For example, “contiguous nucleobases” means nucleobases that are immediately adjacent to each other
  • As used herein, “crRNA” means an oligonucleotide or portion of an oligonucleotide that comprises a DNA recognition portion and a tracrRNA recognition portion. As used herein, “DNA recognition portion” is nucleobase sequence that is complementary to a DNA target. As used herein, “tracrRNA recognition portion” is a nucleobase sequence that is bound to or is capable of binding to tracrRNA. The tracRNA recognition portion of crRNA may bind to tracrRNA via hybridization or covalent attachment.
  • As used herein, “fully modified” in reference to an oligonucleotide means a modified oligonucleotide in which each sugar moiety is modified. “Uniformly modified” in reference to an oligonucleotide means a fully modified oligonucleotide in which each at least one modification of each sugar moiety is the same. For example, the nucleosides of a uniformly modified oligonucleotide can each have a 2′-MOE modification but different nucleobase modifications, and the internucleoside linkages may be different.
  • As used herein, “gene editing” means any process mediated by a Cas9/crRNA/tracrRNA or Cas9/sgRNA complex, including but not limited to gene knock-down, gene knock-out, gene disruption, deletion, insertion, and gene activation. As used herein, “gene alteration” means any process mediated by a nuclease/scrRNA containing complex, including but not limited to gene knock-down, gene disruption, deletion, insertion, and gene activation.
  • As used herein, “gRNA” comprises both a crRNA and a tracrRNA. In certain embodiments, the crRNA and tracrRNA of a gRNA are distinct molecules. In certain embodiments, the crRNA and tracrRNA of a gRNA are portions of one oligonucleotide, wherein the oligonucleotide is referred to as a “sgRNA”.
  • As used herein, “hybridization” means the pairing or annealing of complementary oligonucleotides and/or nucleic acids. While not limited to a particular mechanism, the most common mechanism of hybridization involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleobases.
  • As used herein, “increases”, when used in reference to an effect mediated by a modified oligonucleotide, means that the effect is greater in the presence of the oligonucleotide containing a certain modification than the effect is in the presence of a corresponding oligonucleotide that does not contain the certain modification.
  • As used herein, the terms “internucleoside linkage” means a group that forms a covalent linkage between adjacent nucleosides in an oligonucleotide. As used herein “modified internucleoside linkage” means any internucleoside linkage other than a naturally occurring, phosphate internucleoside linkage. Naturally occurring, non-phosphate linkages are referred to herein as modified internucleoside linkages. “Phosphorothioate linkage” means a linkage between nucleosides wherein the phosphodiester bond of a phosphate linkage is modified by replacing one of the non-bridging oxygen atoms with a sulfur atom. A phosphorothioate linkage is a modified internucleoside linkage.
  • As used herein, “linearly modified sugar” or “linearly modified sugar moiety” means a modified sugar moiety that comprises an acyclic or non-bridging modification. Such linear modifications are distinct from bicyclic sugar modifications.
  • As used herein, “linked nucleosides” are nucleosides that are connected in a continuous sequence (i.e. no additional nucleosides are present between those that are linked). Linked nucleosides may or may not be linked by internucleoside linkages.
  • As used herein, “mismatch” or means a nucleobase of a first oligonucleotide that is not capable of pairing with the corresponding nucleobase of a second oligonucleotide or target nucleic acid when the first and second oligomeric compound are aligned.
  • As used herein, “MOE” means methoxyethyl. “2′-MOE” means a —OCH2CH2OCH3 group at the 2′ position of a furanosyl ring.
  • As used herein, “motif” means the pattern of unmodified and/or modified sugar moieties, nucleobases, and/or internucleoside linkages, in an oligonucleotide.
  • As used herein, “naturally occurring” means found in nature.
  • As used herein, “nucleobase” means a heterocyclic moiety capable of pairing with a second, different nucleobase. As used herein, “nucleobase sequence” means the order of contiguous nucleobases independent of any sugar or internucleoside linkage modification. As used herein, “modified nucleobase” means a nucleobase other than adenine (A), thymine (T), cytosine (C), uracil (U), and guanine (G), herein defined as the five, unmodified nucleobases. A universal base is a nucleobase that can pair with any one of the five unmodified nucleobases.
  • As used herein, “nucleoside” means a compound comprising a nucleobase and a sugar moiety. The nucleobase and sugar moiety are each, independently, unmodified or modified. As used herein, “modified nucleoside” means a nucleoside comprising a modified nucleobase and/or a modified sugar moiety. Modified nucleosides include abasic nucleosides.
  • As used herein, “oligonucleotide” means a strand of linked nucleosides connected via internucleoside linkages, wherein each nucleoside and internucleoside linkage may be modified or unmodified. Unless otherwise indicated, oligonucleotides consist of 8-50 linked nucleosides. As used herein, “modified oligonucleotide” means an oligonucleotide, wherein at least one nucleoside or internucleoside linkage is modified. As used herein, “unmodified oligonucleotide” means an oligonucleotide that does not comprise any nucleoside modifications or internucleoside modifications.
  • As used herein, “pharmaceutically acceptable carrier or diluent” means any substance suitable for use in administering to an animal. Certain such carriers enable pharmaceutical compositions to be formulated as, for example, tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspension and lozenges for the oral ingestion by a subject.
  • As used herein “pharmaceutically acceptable salts” means physiologically and pharmaceutically acceptable salts of compounds, such as oligomeric compounds, i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto.
  • As used herein “pharmaceutical composition” means a mixture of substances suitable for administering to a subject. For example, a pharmaceutical composition may comprise an crRNA compound and a sterile aqueous solution. In certain embodiments, a pharmaceutical composition shows activity in free uptake assay in certain cell lines.
  • As used herein, “phosphorus moiety” means a group of atoms comprising a phosphorus atom. In certain embodiments, a phosphorus moiety comprises a mono-, di-, or tri-phosphate, or phosphorothioate.
  • As used herein “prodrug” means a therapeutic agent in an inactive form that is converted to an active form within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or physiologic conditions.
  • As used herein, “scrRNA” or “single crRNA” means an oligonucleotide that comprises a scrRNA target recognition portion and a nuclease recognition portion and does not comprise a tracrRNA recognition portion or a tracrRNA. In certain embodiments, scrRNAs comprise a self-complementary region. In certain such embodiments, the nuclease recognition portion partially or completely overlaps with the self-complementary region. As used herein, “scrRNA target recognition portion” is a portion of an oligonucleotide with a nucleobase sequence that is complementary to a scrRNA DNA target. As used herein, “nuclease recognition portion” is a portion of an oligonucleotide that can bind to, associate with, or contribute to the binding to or association with a nuclease that is not a Cas9 nuclease. In certain embodiments, the nuclease recognition portion of an oligonucleotide binds to or associates with a Cpf1 nuclease.
  • As used herein, “self-complementary” in reference to an oligonucleotide means an oligonucleotide that is at least partially complementary to itself. In certain embodiments, a self-complementary oligonucleotide forms a hairpin when a portion of the self-complementary oligonucleotide hybridizes to itself.
  • As used herein, “sugar moiety” means a group of atoms that can link a nucleobase to another group, such as an internucleoside linkage, conjugate group, or terminal group. In certain embodiments, a sugar moiety is attached to a nucleobase to form a nucleoside. As used herein, “unmodified sugar moiety” means a 2′-OH(H) furanosyl moiety, as found in RNA, or a 2′-H(H) moiety, as found in DNA. Unmodified sugar moieties have one hydrogen at each of the 1′, 3′, and 4′ positions, an oxygen at the 3′ position, and two hydrogens at the 5′ position. As used herein, “modified sugar moiety” or “modified sugar” means a sugar surrogate or a furanosyl moiety comprising a non-hydrogen substituent in place of at least one hydrogen of an unmodified sugar moiety. In certain embodiments, a modified sugar moiety is a 2′-substituted sugar moiety. Such modified sugar moieties include bicyclic sugars and linearly modified sugars.
  • As used herein, “sugar surrogate” means a modified sugar moiety having other than a furanosyl moiety that can link a nucleobase to another group, such as an internucleoside linkage, conjugate group, or terminal group. Modified nucleosides comprising sugar surrogates can be incorporated into one or more positions within an oligonucleotide. In certain embodiments, such oligonucleotides are capable of hybridizing to complementary oligomeric compounds or nucleic acids.
  • As used herein, “target nucleic acid,” “target DNA,” “target gene” and “nucleic acid target” mean a nucleic acid that a crRNA is designed to affect. As used herein, “scrRNA target nucleic acid,” “scrRNA target DNA,” scrRNA target gene” and “scrRNA nucleic acid target” mean a nucleic acid that a scrRNA is designed to affect. An “off-target gene” is a gene that a crRNA or a scrRNA is not designed to affect. In certain embodiments, the editing or alteration of an off-target gene is deleterious.
  • As used herein, “terminal group” means a chemical group or group of atoms that is covalently linked to a terminus of an oligonucleotide.
  • As used herein, “tracrRNA” means an oligonucleotide or portion of an oligonucleotide that can non-covalently bind to a Cas9 protein and that can bind to a crRNA via hybridization or covalent attachment.
  • Certain Oligonucleotides for Use in a CRISPR System
  • I. Certain CRISPR RNA (crRNA)
  • In certain embodiments, the present invention provides modified oligonucleotides for use in CRISPR. Typically, CRISPR employs CRSPR RNA (crRNA), which hybridizes to target DNA and also hybridizes to trans-activating RNA (tracrRNA), which in turn recruits a nuclease, cas9, which cleaves the target DNA. Thus, the crRNA in such systems has two functions: (1) recognition and hybridization to the target DNA and (2) recognition and hybridization to the tracrRNA. Typically, in such systems, the crRNA has two portions which correspond to these two functions: a DNA recognition portion and a tracrRNA recognition portion. The present invention provides modified oligonucleotides that may be used in crRNA. Such modified oligonucleotides may have modifications in the DNA recognition portion and/or tracrRNA recognition portion.
  • In certain embodiments, the tracrRNA recognition portion of the crRNA comprises a portion of the direct repeat sequence from a bacterial species that has a Type II CRISPR system. In certain such embodiments, the tracrRNA recognition portion of the crRNA comprises a sequence selected from the table below. In certain embodiments, the tracrRNA recognition portion of the crRNA comprises the first 12 nucleobases of a sequence selected from the table below. In certain embodiments, the tracrRNA recognition portion of the crRNA comprises the first 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 nucleobases of a sequence selected from the table below. In certain embodiments, the sequence of the tracrRNA recognition portion of the crRNA consists of the first 12 nucleobases of a sequence selected from the table below. In certain embodiments, the sequence of the tracrRNA recognition portion of the crRNA consists of the first 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 nucleobases of a sequence selected from the table below.
  • TABLE B
    Direct repeat sequences used in tracrRNA
    recognition portions of crRNA
    SEQ
    ID
    Species Sequence NO.
    S. pyogenes GUUUUAGAGCU 19
    AUGCUGUUUUG
    S. aureus GUUUUAGUACU 20
    CUGUAAUUUUA
    S. thermophiles GUUUUUGUACU 21
    CUCAAGAUUUA
    S. pasteurianus GUUUUUGUACU 21
    CUCAAGAUUUA
    N. cinerea GUUGUAGCUCC 22
    CAUUCUCAUUU
    C. lari GUUUUAGUCUC 23
    UUUUUAAAUUU
    P. lavamentivoran GCUGCGGAUUG 24
    CGGCCGUCUCU
    C. diptheriae ACUGGGGUUCA 25
    GUUCUCAAAAA
  • In certain instances, the crRNA and tracrRNA are joined to one another to form a single molecule referred to as a single guide RNA (sgRNA). In certain embodiments, the present invention provides modified oligonucleotides for use in sgRNA.
  • II. Certain Single CRISPR RNA (scrRNA)
  • In certain alternative embodiments, the present invention provides modified oligonucleotides for use in a CRISPR system that employs scrRNA, which hybridizes to a scrRNA target DNA and participates in recruitment of a nuclease other than Cas9. In certain such embodiments, the nuclease is a Cpf1 nuclease or a variant thereof. The nuclease (e.g., the Cpf1 nuclease) cleaves the scrRNA target DNA. Thus, the scrRNA in such systems has two functions: (1) recognition and hybridization to the scrRNA target DNA and (2) recognition and recruitment of the nuclease. Typically, in such systems, the scrRNA has two portions which correspond to these two functions: a scrRNA target recognition portion and a nuclease recognition portion. The present invention provides modified oligonucleotides that may be used in scrRNA. Such modified oligonucleotides may have modifications in the scrRNA target recognition portion and/or nuclease recognition portion. In certain embodiments, the nuclease recognition portion is 5′ to the scrRNA target recognition portion. In certain embodiments, the nuclease recognition portion is 3′ to the scrRNA target recognition portion.
  • In certain embodiments, the nuclease recognition portion of the scrRNA comprises a portion of the direct repeat sequence from a bacterial organism that has a Cpf1 nuclease or a Cpf1 ortholog. In certain such embodiments, the nuclease recognition portion of the scrRNA comprises a sequence selected from the table below. In certain embodiments, the nuclease recognition portion of the scrRNA comprises 12 nucleobases of a sequence selected from the table below. In certain embodiments, the tracrRNA recognition portion of the crRNA comprises 13, 14, 15, 16, 17, 18, or 19 nucleobases of a sequence selected from the table below. In certain embodiments, the sequence of the nuclease recognition portion of the scrRNA consists of 12 nucleobases of a sequence selected from the table below. In certain embodiments, the sequence of the nuclease recognition portion of the scrRNA consists of 13, 14, 15, 16, 17, 18, 19, 20, or 21 nucleobases of a sequence selected from the table below. In certain embodiments, the nuclease recognition portion of the scrRNA comprises the sequence UCUACU and GUAGAU.
  • TABLE C
    Direct repeat sequences used in nuclease
    recognition portions of scrRNA
    SEQ
    ID
    Organism Sequence NO.
    Francisella UAAUUUCUACUGUUGUAGAU 26
    novicida
    Lachnospiraceae AGAAAUGCAUGGUUCUCAUGC 27
    bacterium
    MC2017
    Butyrivibrio AAAAUUACCUAGUAAUUAGGU 28
    proteoclasticus
    Peregrinibacteria GGAUUUCUACUUUUGUAGAU
    29
    bacterium
    Parcubacteria AAAUUUCUACUUUUGUAGAU 30
    bacterium
    Smithella GUUUCAAUCCACGCGCCCAC 31
    GCGGGGCGCGAC
    Acidaminococcus UAAUUUCUACUCUUGUAGAU
    32
    Lachnospiraceae GAAUUUCUACUAUUGUAGAU 33
    bacterium
    MA2020
    Candidatus GAAUCUCUACUCUUUGUAGAU 34
    Methanoplasma
    termitum
    Enbacterium UAAUUUCUACUUUGUAGAU 35
    eligens
    Moraxella AAAUUUCUACUGUUUGUAGAU 36
    bovoculi
    Leptospira GAAUUUCUACUUUUGUAGAU 37
    inadai
    Lachnospiraceae UAAUUUCUACUAAGUGUAGAU 38
    bacterium
    ND2006
    Porphyromonas UAAUUUCUACUAUUGUAGAU 39
    crevioricanis
    Prevotella UAAUUUCUACUUCGGUAGAU 40
    disiens
    Porphyromonas UAAUUUCUACUAUUGUAGAU 39
    macacae

    Certain Oligonucleotides for Use as crRNA
  • In certain embodiments, modified crRNA comprise a modified oligonucleotide. In certain embodiments, modified crRNA consist of a modified oligonucleotide. Modified oligonucleotides described herein are suitable for use as crRNA.
  • Certain modified oligonucleotides have one or more asymmetric center and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), as a or 13 such as for sugar anomers, or as (D) or (L) such as for amino acids etc. Included in the modified oligonucleotides provided herein are all such possible isomers, including their racemic and optically pure forms, unless specified otherwise. Likewise, all cis- and trans-isomers and tautomeric forms are also included.
  • In certain embodiments, such modified oligonucleotides may contain any combination of the modified sugar moieties, modified nucleobases, modified internucleoside linkages, motifs, and/or lengths described herein.
  • Certain Oligonucleotides for Use as scrRNA
  • In certain embodiments, modified scrRNA comprise a modified oligonucleotide. In certain embodiments, modified scrRNA consist of a modified oligonucleotide. Modified oligonucleotides described herein are suitable for use as scrRNA.
  • Certain modified oligonucleotides have one or more asymmetric center and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), as a or 13 such as for sugar anomers, or as (D) or (L) such as for amino acids etc. Included in the modified oligonucleotides provided herein are all such possible isomers, including their racemic and optically pure forms, unless specified otherwise. Likewise, all cis- and trans-isomers and tautomeric forms are also included.
  • In certain embodiments, such modified oligonucleotides may contain any combination of the modified sugar moieties, modified nucleobases, modified internucleoside linkages, motifs, and/or lengths described herein.
  • Certain Methods of Use Comprising Modified crRNA
  • In certain embodiments, methods comprising contacting a cell with a compound comprising a modified crRNA are in vitro methods. In certain embodiments, methods comprising contacting a cell with a compound comprising a modified crRNA are ex vivo methods. In certain embodiments, methods comprising contacting a cell with a compound comprising a modified crRNA are in vivo methods.
  • Various Cas9 variants, both naturally occurring and genetically engineered, can be used in the methods of the present invention. Such Cas9 variants include but are not limited to inactive Cas9 mutants that are used in applications that do not require target nucleic acid cleavage, such as gene activation, and truncated Cas9 variants that are suitable for expression in certain vectors, such as AAV vectors.
  • In certain embodiments, methods comprising contacting a cell with a compound comprising a modified crRNA further comprise contacting the cell with a second compound to inhibit (or turn off) the CRISPR system after the target gene is edited.
  • In certain embodiments, gene editing methods comprising contacting a cell with a compound comprising a modified crRNA produce fewer and/or less deleterious off-target effects than gene editing methods that use of an unmodified crRNA in place of the modified crRNAs of the invention.
  • Certain Methods of Use Comprising Modified scrRNA
  • In certain embodiments, methods comprising contacting a cell with a compound comprising a modified scrRNA are in vitro methods. In certain embodiments, methods comprising contacting a cell with a compound comprising a modified scrRNA are ex vivo methods. In certain embodiments, methods comprising contacting a cell with a compound comprising a modified scrRNA are in vivo methods.
  • Various nuclease variants, both naturally occurring and genetically engineered, can be used in the methods of the present invention. Such nuclease variants include but are not limited to inactive nuclease mutants that are used in applications that do not require scrRNA target nucleic acid cleavage, such as gene activation, and truncated nuclease variants that are suitable for expression in certain vectors, such as AAV vectors.
  • In certain embodiments, methods comprising contacting a cell with a compound comprising a modified scrRNA further comprise contacting the cell with a second compound to inhibit (or turn off) the CRISPR system after the scrRNA target gene is altered.
  • In certain embodiments, gene altering methods comprising contacting a cell with a compound comprising a modified scrRNA produce fewer and/or less deleterious off-target effects than gene altering methods that use an unmodified scrRNA in place of the modified scrRNAs of the invention.
  • A. Certain Modified Nucleosides
  • Certain compounds of the present invention incorporate modified nucleosides. Unless otherwise provided, the following modified nucleosides, without limitation, are suitable for such incorporation into modified oligonucleotides for use as crRNA or scrRNA. In certain embodiments, modified oligonucleotides comprise at least one modified nucleoside. Such modified nucleosides comprise a modified sugar moiety or a modified nucleobase or both a modified sugar moiety and a modified nucleobase.
  • 1. Certain Sugar Moieties
  • In certain embodiments, modified oligonucleotides, such as modified crRNAs or modified scrRNAs, comprise one or more modified nucleosides comprising a modified sugar moiety. Such modified oligonucleotides comprising one or more sugar-modified nucleosides may have desirable properties, such as enhanced nuclease stability or increased binding affinity with a target nucleic acid relative to oligonucleotides lacking such sugar-modified nucleosides. In certain embodiments, modified sugar moieties are linearly modified sugar moieties. In certain embodiments, modified sugar moieties are bicyclic or tricyclic sugar moieties. In certain embodiments, modified sugar moieties are sugar surrogates. Such sugar surrogates may comprise one or more substitutions corresponding to those of substituted sugar moieties.
  • In certain embodiments, modified sugar moieties are linearly modified sugar moieties comprising a furanosyl ring with one or more acyclic substituent, including but not limited to substituents at the 2′ and/or 5′ positions. Examples of 2′-substituent groups suitable for linearly modified sugar moieties include but are not limited to: 2′-F, 2′-OCH3 (“OMe” or “O-methyl”), and 2′-O(CH2)2OCH3 (“MOE”). In certain embodiments, 2′-substituent groups are selected from among: halo, allyl, amino, azido, SH, CN, OCN, CF3, OCF3, O—C1-C10 alkoxy, O—C1-C10 substituted alkoxy, O—C1-C10 alkyl, O—C1-C10 substituted alkyl, S-alkyl, N(Rm)-alkyl, O-alkenyl, S-alkenyl, N(Rm)-alkenyl, O-alkynyl, S-alkynyl, N(Rm)-alkynyl, O-alkylenyl-O-alkyl, alkynyl, alkaryl, aralkyl, O-alkaryl, O-aralkyl, O(CH2)2SCH3, O(CH2)2ON(Rm)(Rn) or OCH2C(═O)—N(Rm)(Rn), where each Rm and Rn is, independently, H, an amino protecting group, or substituted or unsubstituted C1-C10 alkyl. Certain embodiments of these 2′-substituent groups can be further substituted with one or more substituent groups independently selected from among: hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro (NO2), thiol, thioalkoxy, thioalkyl, halogen, alkyl, aryl, alkenyl and alkynyl. Examples of 5′-substituent groups suitable for linearly modified sugar moieties include but are not limited to: 5′-methyl (R or S), 5′-vinyl, and 5′-methoxy. In certain embodiments, linearly modified sugars comprise more than one non-bridging sugar substituent, for example, 2′-F-5′-methyl sugar moieties (see, e.g., PCT International Application WO 2008/101157, for additional 2′, 5′-bis substituted sugar moieties and nucleosides).
  • In certain embodiments, a 2′-substituted nucleoside or 2′-linearly modified nucleoside comprises a sugar moiety comprising a linear 2′-substituent group selected from: F, NH2, N3, OCF3, OCH3, O(CH2)3NH2, CH2CH═CH2, OCH2CH═CH2, OCH2CH2OCH3, O(CH2)2SCH3, O(CH2)2ON(Rm)(Rn), O(CH2)2O(CH2)2N(CH3)2, and N-substituted acetamide (OCH2C(═O)—N(Rm)(Rn)), where each Rm and Rn is, independently, H, an amino protecting group, or substituted or unsubstituted C1-C10 alkyl.
  • In certain embodiments, a 2′-substituted nucleoside or 2′-linearly modified nucleoside comprises a sugar moiety comprising a linear 2′-substituent group selected from: F, OCF3, OCH3, OCH2CH2OCH3, O(CH2)2SCH3, O(CH2)2ON(CH3)2, O(CH2)2O(CH2)2N(CH3)2, and OCH2C(═O)—N(H)CH3 (“NMA”).
  • In certain embodiments, a 2′-substituted nucleoside or 2′-linearly modified nucleoside comprises a sugar moiety comprising a linear 2′-substituent group selected from: F, OCH3, and OCH2CH2OCH3.
  • Nucleosides comprising modified sugar moieties, such as linearly modified sugar moieties, are referred to by the position(s) of the substitution(s) on the sugar moiety of the nucleoside. For example, nucleosides comprising 2′-substituted or 2-modified sugar moieties are referred to as 2′-substituted nucleosides or 2-modified nucleosides.
  • Certain modified sugar moieties comprise a bridging sugar substituent that forms a second ring resulting in a bicyclic sugar moiety. In certain such embodiments, the bicyclic sugar moiety comprises a bridge between the 4′ and the 2′ furanose ring atoms. Examples of such 4′ to 2′ bridging sugar substituents include but are not limited to: 4′-CH2-2′, 4′-(CH2)2-2′, 4′-(CH2)3-2′, 4′-CH2—O-2′ (“LNA”), 4′-CH2—S-2′, 4′-(CH2)2—O-2′ (“ENA”), 4′-CH(CH3)—O-2′ (referred to as “constrained ethyl” or “cEt” when in the S configuration), 4′-CH2—O—CH2-2′, 4′-CH2—N(R)-2′, 4′-CH(CH2OCH3)—O-2′ (“constrained MOE” or “cMOE”) and analogs thereof (see, e.g., U.S. Pat. No. 7,399,845), 4′-C(CH3)(CH3)—O-2′ and analogs thereof (see, e.g., WO2009/006478), 4′-CH2—N(OCH3)-2′ and analogs thereof (see, e.g., WO2008/150729), 4′-CH2—O—N(CH3)-2′ (see, e.g., US2004/0171570), 4′-CH2—C(H)(CH3)-2′ (see, e.g., Chattopadhyaya, et al., J. Org. Chem., 2009, 74, 118-134), 4′-CH2—C(═CH2)-2′ and analogs thereof (see, published PCT International Application WO 2008/154401), 4′-C(RaRb)—N(R)—O-2′, 4′-C(RaRb)—O—N(R)-2′, 4′-CH2—O—N(R)-2′, and 4′-CH2—N(R)—O-2′, wherein each R, Ra, and Rb is, independently, H, a protecting group, or Ci-Cu alkyl (see, e.g. U.S. Pat. No. 7,427,672).
  • In certain embodiments, such 4′ to 2′ bridges independently comprise from 1 to 4 linked groups independently selected from: —[C(Ra)(Rb)]n—, —[C(Ra)(Rb)]n—O—, —C(Ra)═C(Rb)—, —C(Ra)═N—, —C(═NRa)—, —C(═O)—, —C(═S)—, —O—, —Si(Ra)2—, —S(═O)x—, and —N(Ra)—;
  • wherein:
  • x is 0, 1, or 2;
  • n is 1, 2, 3, or 4;
  • each Ra and Rb is, independently, H, a protecting group, hydroxyl, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C5-C20 aryl, substituted C5-C20 aryl, heterocycle radical, substituted heterocycle radical, heteroaryl, substituted heteroaryl, C5-C7 alicyclic radical, substituted C5-C7 alicyclic radical, halogen, OJ1, NJ1J2, SJ1, N3, COOJ1, acyl (C(═O)—H), substituted acyl, CN, sulfonyl (S(═O)2-J1), or sulfoxyl (S(═O)-J1); and
  • each J1 and J2 is, independently, H, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C5-C20 aryl, substituted C5-C20 aryl, acyl (C(═O)—H), substituted acyl, a heterocycle radical, a substituted heterocycle radical, C1-C12 aminoalkyl, substituted C1-C12 aminoalkyl, or a protecting group.
  • Additional bicyclic sugar moieties are known in the art, for example: Freier et al., Nucleic Acids Research, 1997, 25(22), 4429-4443, Albaek et al., J. Org. Chem., 2006, 71, 7731-7740, Singh et al., Chem. Commun., 1998, 4, 455-456; Koshkin et al., Tetrahedron, 1998, 54, 3607-3630; Wahlestedt et al., Proc. Natl. Acad. Sci. U.S.A., 2000, 97, 5633-5638; Kumar et al., Bioorg. Med. Chem. Lett., 1998, 8, 2219-2222; Singh et al., J. Org. Chem., 1998, 63, 10035-10039; Srivastava et al., J. Am. Chem. Soc., 20017, 129, 8362-8379; Elayadi et al., Curr. Opinion Invens. Drugs, 2001, 2, 558-561; Braasch et al., Chem. Biol., 2001, 8, 1-7; Orum et al., Curr. Opinion Mol. Ther., 2001, 3, 239-243; U.S. Pat. Nos. 7,053,207, 6,268,490, 6,770,748, 6,794,499, 7,034,133, 6,525,191, 6,670,461, and 7,399,845; WO 2004/106356, WO 1994/14226, WO 2005/021570, and WO 2007/134181; U.S. Patent Publication Nos. US2004/0171570, US2007/0287831, and US2008/0039618; U.S. patent Ser. Nos. 12/129,154, 60/989,574, 61/026,995, 61/026,998, 61/056,564, 61/086,231, 61/097,787, and 61/099,844; and PCT International Applications Nos. PCT/US2008/064591, PCT/US2008/066154, and PCT/US2008/068922.
  • In certain embodiments, bicyclic sugar moieties and nucleosides incorporating such bicyclic sugar moieties are further defined by isomeric configuration. For example, an LNA nucleoside (described above) may be in the α-L configuration or in the β-D configuration.
  • Figure US20230159919A1-20230525-C00001
  • α-L-methyleneoxy (4′-CH2—O-2′) or α-L-LNA bicyclic nucleosides have been incorporated into oligonucleotides (Frieden et al., Nucleic Acids Research, 2003, 21, 6365-6372). Herein, general descriptions of bicyclic nucleosides include both isomeric configurations. When the positions of specific bicyclic nucleosides (e.g., LNA or cEt) are identified in exemplified embodiments herein, they are in the β-D configuration, unless otherwise specified.
  • In certain embodiments, modified sugar moieties comprise one or more non-bridging sugar substituent and one or more bridging sugar substituent (e.g., 5′-substituted and 4′-2′ bridged sugars). (see, e.g., WO 2007/134181, wherein LNA nucleosides are further substituted with, for example, a 5′-methyl or a 5′-vinyl group, and see, e.g., U.S. Pat. Nos. 7,547,684; 7,750,131; 8,030,467; 8,268,980; 7,666,854; and 8,088,746).
  • In certain embodiments, modified sugar moieties are sugar surrogates. In certain such embodiments, the oxygen atom of the sugar moiety is replaced, e.g., with a sulfur, carbon or nitrogen atom. In certain such embodiments, such modified sugar moieties also comprise bridging and/or non-bridging substituents as described above. For example, certain sugar surrogates comprise a 4′-sulfur atom and a substitution at the 2′-position (see, e.g., US2005/0130923) and/or the 5′ position.
  • In certain embodiments, sugar surrogates comprise rings having other than 5 atoms. For example, in certain embodiments, a sugar surrogate comprises a six-membered tetrahydropyran (“THP”). Such tetrahydropyrans may be further modified or substituted. Nucleosides comprising such modified tetrahydropyrans include but are not limited to hexitol nucleic acid (“HNA”), anitol nucleic acid (“ANA”), manitol nucleic acid (“MNA”) (see Leumann, C J. Bioorg. & Med. Chem. 2002, 10, 841-854), fluoro HNA:
  • Figure US20230159919A1-20230525-C00002
  • (“F-HNA”, see e.g., U.S. Pat. Nos. 8,088,904; 8,440,803; and 8,796,437, F-HNA can also be referred to as a F-THP or 3′-fluoro tetrahydropyran), and nucleosides comprising additional modified THP compounds having the formula:
  • Figure US20230159919A1-20230525-C00003
  • wherein, independently, for each of said modified THP nucleoside:
  • Bx is a nucleobase moiety;
  • T3 and T4 are each, independently, an internucleoside linking group linking the modified THP nucleoside to the remainder of an oligonucleotide or one of T3 and T4 is an internucleoside linking group linking the modified THP nucleoside to the remainder of an oligonucleotide and the other of T3 and T4 is H, a hydroxyl protecting group, a linked conjugate group, or a 5′ or 3′-terminal group;
  • q1, q2, q3, q4, q5, q6 and q7 are each, independently, H, C1-C6 alkyl, substituted C1-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl, or substituted C2-C6 alkynyl; and
  • each of R1 and R2 is independently selected from among: hydrogen, halogen, substituted or unsubstituted alkoxy, NJ1J2, SJ1, N3, OC(═X)J1, OC(═X)NJ1J2, NJ3C(═X)NJ1J2, and CN, wherein X is O, S or NJ1, and each J1, J2, and J3 is, independently, H or C1-C6 alkyl.
  • In certain embodiments, modified THP nucleosides are provided wherein q1, q2, q3, q4, q5, q6 and q7 are each H. In certain embodiments, at least one of q1, q2, q3, q4, q5, q6 and q7 is other than H. In certain embodiments, at least one of q1, q2, q3, q4, q5, q6 and q7 is methyl. In certain embodiments, modified THP nucleosides are provided wherein one of R1 and R2 is F. In certain embodiments, R1 is F and R2 is H, in certain embodiments, R1 is methoxy and R2 is H, and in certain embodiments, R1 is methoxyethoxy and R2 is H.
  • In certain embodiments, sugar surrogates comprise rings having more than 5 atoms and more than one heteroatom. For example, nucleosides comprising morpholino sugar moieties and their use in oligonucleotides have been reported (see, e.g., Braasch et al., Biochemistry, 2002, 41, 4503-4510 and U.S. Pat. Nos. 5,698,685; 5,166,315; 5,185,444; and 5,034,506). As used here, the term “morpholino” means a sugar surrogate having the following structure:
  • Figure US20230159919A1-20230525-C00004
  • In certain embodiments, morpholinos may be modified, for example by adding or altering various substituent groups from the above morpholino structure. Such sugar surrogates are referred to herein as “modified morpholinos.”
  • In certain embodiments, sugar surrogates comprise acyclic moieties. Examples of nucleosides and oligonucleotides comprising such acyclic sugar surrogates include but are not limited to: peptide nucleic acid (“PNA”), acyclic butyl nucleic acid (see, e.g., Kumar et al., Org. Biomol. Chem., 2013, 11, 5853-5865), and nucleosides and oligonucleotides described in WO2011/133876.
  • Many other bicyclic and tricyclic sugar and sugar surrogate ring systems are known in the art that can be used in modified nucleosides (see, e.g., Leumann, J. C, Bioorganic & Medicinal Chemistry, 2002, 10, 841-854).
  • 2. Certain Modified Nucleobases
  • In certain embodiments, modified oligonucleotides, such as modified crRNAs or modified scrRNAs, comprise one or more nucleoside comprising an unmodified nucleobase. In certain embodiments, modified oligonucleotides comprise one or more nucleoside comprising a modified nucleobase. In certain embodiments, modified oligonucleotides comprise one or more nucleoside that does not comprise a nucleobase, referred to as an abasic nucleoside.
  • In certain embodiments, modified nucleobases are selected from: 5-substituted pyrimidines, 6-azapyrimidines, alkyl or alkynyl substituted pyrimidines, alkyl substituted purines, and N-2, N-6 and O-6 substituted purines. In certain embodiments, modified nucleobases are selected from: 2-aminopropyladenine, 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-N-methylguanine, 6-N-methyladenine, 2-propyladenine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-propynyl (—C≡C—CH3) uracil, 5-propynylcytosine, 6-azouracil, 6-azocytosine, 6-azothymine, 5-ribosyluracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl, 8-aza and other 8-substituted purines, 5-halo, particularly 5-bromo, 5-trifluoromethyl, 5-halouracil, and 5-halocytosine, 7-methylguanine, 7-methyladenine, 2-F-adenine, 2-aminoadenine, 7-deazaguanine, 7-deazaadenine, 3-deazaguanine, 3-deazaadenine, 6-N-benzoyladenine, 2-N-isobutyrylguanine, 4-N-benzoylcytosine, 4-N-benzoyluracil, 5-methyl 4-N-benzoylcytosine, 5-methyl 4-N-benzoyluracil, universal bases, hydrophobic bases, promiscuous bases, size-expanded bases, and fluorinated bases. Further modified nucleobases include tricyclic pyrimidines, such as 1,3-diazaphenoxazine-2-one, 1,3-diazaphenothiazine-2-one and 9-(2-aminoethoxy)-1,3-diazaphenoxazine-2-one (G-clamp). Modified nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example, 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, Kroschwitz, J. I., Ed., John Wiley & Sons, 1990, 858-859; Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613; Sanghvi, Y. S., Chapter 15, Antisense Research and Applications, Crooke, S. T. and Lebleu, B., Eds., CRC Press, 1993, 273-288; and those disclosed in Chapters 6 and 15, Antisense Drug Technology, Crooke S. T., Ed., CRC Press, 2008, 163-166 and 442-443.
  • Representative United States patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include without limitation, US2003/0158403, U.S. Pat. Nos. 3,687,808; 4,845,205; 5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,434,257; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121; 5,596,091; 5,614,617; 5,645,985; 5,681,941; 5,750,692; 5,763,588; 5,830,653 and 6,005,096.
  • B. Certain Modified Internucleoside Linkages
  • In certain embodiments, nucleosides of modified oligonucleotides, such as modified crRNAs or modified scrRNAs, may be linked together using any internucleoside linkage. The two main classes of internucleoside linking groups are defined by the presence or absence of a phosphorus atom. Representative phosphorus-containing internucleoside linkages include but are not limited to phosphates, which contain a phosphodiester bond (“P═O”) (also referred to as unmodified or naturally occurring linkages), phosphotriesters, methylphosphonates, phosphoramidates, and phosphorothioates (“P═S”), and phosphorodithioates (“HS—P═S”). Representative non-phosphorus containing internucleoside linking groups include but are not limited to methylenemethylimino (—CH2—N(CH3)—O—CH2—), thiodiester (—O—C(═O)—S—), thionocarbamate (—O—C(═O)(NH)—S—); siloxane (—O—SiH2—O—); and N,N′-dimethylhydrazine (—CH2—N(CH3)—N(CH3)—). Modified internucleoside linkages, compared to naturally occurring phosphate linkages, can be used to alter, typically increase, nuclease resistance of the oligonucleotide. In certain embodiments, internucleoside linkages having a chiral atom can be prepared as a racemic mixture, or as separate enantiomers. Representative chiral internucleoside linkages include but are not limited to alkylphosphonates and phosphorothioates. Methods of preparation of phosphorous-containing and non-phosphorous-containing internucleoside linkages are well known to those skilled in the art.
  • Neutral internucleoside linkages include, without limitation, phosphotriesters, methylphosphonates, MMI (3′-CH2—N(CH3)—O-5′), amide-3 (3′-CH2—C(═O)—N(H)-5′), amide-4 (3′-CH2—N(H)—C(═O)-5′), formacetal (3′-O—CH2—O-5′), methoxypropyl, and thioformacetal (3′-S—CH2—O-5′). Further neutral internucleoside linkages include nonionic linkages comprising siloxane (dialkylsiloxane), carboxylate ester, carboxamide, sulfide, sulfonate ester and amides (See for example: Carbohydrate Modifications in Antisense Research; Y. S. Sanghvi and P. D. Cook, Eds., ACS Symposium Series 580; Chapters 3 and 4, 40-65). Further neutral internucleoside linkages include nonionic linkages comprising mixed N, O, S and CH2 component parts.
  • 1. Certain Modification Motifs
  • In certain embodiments, the crRNA has a modification motif selected from the table below.
  • TABLE A
    crRNA modification motifs
    29-mers 42-mers
    f7r6kr3kr3kr3krk2 f10r18kr4kr2kr3k2
    mf6r6kr3kr3kr3krk2 mf9r18kr4kr2kr3k2
    mf6r10k6r2k4 mr27kr4kr2kr3k2
    mr16k6r2k4 m r9f10k6r2kr4kr2kr3k2
    mr6f10k6r2k4 mr9f10I6r2kr4kr2kr3k2
    mf6r10f6r2k4 mr9f16r2kr4kr2kr3k2
    mf6r10I6r2k4 mr32kr2kr3k2
    mr6f10I6r2I4 ef9r18kr4kr2kr3k2
    mf6r10k6r2I4 r(MOP)f9r18kr4kr2kr3k2
    mr16k6r2I4 d(MOP)f9r18kr4kr2kr3k2
    mr6f10k6r2I4 f(MOP)f9r18kr4kr2kr3k2
    mf6r10f6r2I4 r(MP)f9r18kr4kr2kr3k2
    r(MOP)f6r6kr3kr3kr3krk2 d(MP)f9r18kr4kr2kr3k2
    d(MOP)f6r6kr3kr3kr3krk2 f(MP)f9r18kr4kr2kr3k2
    f(MOP)f6r6kr3kr3kr3krk2 r(MMI)f9r18kr4kr2kr3k2
    r(MP)f6r6kr3kr3kr3krk2 d(MMI)f9r18kr4kr2kr3k2
    d(MP)f6r6kr3kr3kr3krk2 f(MMI)f9r18kr4kr2kr3k2
    f(MP)f6r6kr3kr3kr3krk2 mr32kr2k(G-Clamp)r2k2
    r(MOP)f6r10k6r2k4 mr27k3r2kr2kr3k2
    d(MOP)f6r10k6r2k4 mf9r18k3r2kr2kr3k2
    f(MOP)f6r10k6r2k4 Mf9r11(5-Propyne-U)4r3k3r2kr3k2
    r(MP)f6r10k6r2k4 29-mers
    r(MP)f6r10k6r2k4 d(MOP)r6f10k6r2k4
    r(MP)f6r10k6r2k4 f(MOP)r6f10k6r2k4
    r(MOP)r16k6r2k4 r(MP)r6f10k6r2k4
    d(MOP)r16k6r2k4 d(MOP)r6f10k6r2k4
    f(MOP)r16k6r2k4 f(MOP)r6f10k6r2k4
    r(MP)r16k6r2k4 r(MOP)f6r10I6r2k4
    d(MP)r16k6r2k4 d(MOP)f6r10I6r2k4
    f(MP)r16k6r2k4 f(MOP)f6r10I6r2k4
    r(MOP)r6f10k6r2k4 r(MOP)f6r10I6r2k4
    d(MP)f5r10k6r2I4 d(MOP)fer10f6r2I4
    f(MP)f6r10k6r2I4 f(MOP)f6r10f6f2I4
    r(MOP)r16k6r2I4 f7r6kr3kr3kr3k(G-Clamp)k2
    d(MOP)r16k6r2I4 mf6r6kr3kr3kr3k(G-Clamp)rk2
    f(MOP)r16k6r2I4 mf6r10k6r2k(G-Clamp)k2
    r(MP)r16k6r2I4 mr16k6r2k (G-Clamp)k2
    d(MP)r16k6r2I4 mr6f10k6r2k(G-Clamp)k2
    f(MP)r16k6r2I4 mf6r10f6r2k(G-Clamp)k2
    r(MOP)r6f10k6r2I4 mf5r10I6r2k(G-Clamp)k2
    r(MOP)r6f10k6r2I4 mr6f10I6r2k(G-Clamp)k2
    r(MOP)r6f10k6r2I4 mf6r10k6r2I(G-Clamp)I2
    r(MOP)r6f10k6r2I4 mr16k6r2I(G-Clamp)I2
    r(MOP)r6f10k6r2I4 mr6f10k6r2I (G-Clamp)I2
    r(MOP)r6f10k6r2I4 mf6r10f6r2I(G-Clamp)I2
    r(MOP)f6r10f6r2I4 f7r5kr3k(5-Propyne)r3kr3krk2
    d(MOP)f6r10f6r2I4 mf6r6kr3k(5-Propyne)r3kr3krk2
    f(MOP)f6r10f6r2I4 r(MOP)f6r10f6r2I4
    Table A Legend:
    “m” indicates a 2′-O-methyl modified nucleoside.
    “f” indicates a 2′-F modified nucleoside, “r” indicates an unmodified 2′-hydroxy sugar containing nucleoside, “d” indicates an unmodified 2′-deoxy sugar containing nucleoside, “e” indicates a 2′-MOE modified nucleoside, “k” indicates a cEt bicyclic sugar containing nucleoside, and “l” indicates an LNA bicyclic sugar containing nucleoside.
    The modifications listed in parentheses are optional modified nucleobases or optional modified internucleoside linkages: “(G-Clamp)” indicates a G-Clamp modified nucleobase that is part of the nucleoside represented by the letter immediately preceding it.
    “(5-Propyne)” indicates a 5′-propynyl modified nucleobase that is part of the nucleoside represented by the letter immediately preceding it.
    “(MOP)” indicates a mcthoxypropyl modified internucleoside linkage.
    “(MP)” indicates a methylphosphonatc internucleoside linkage, and “(MMI)” indicates an MMI N-methyl internucleoside linkage.
    In certain embodiments, crRNAs having a motif with a parenthetical modification listed in the table above include the indicated parenthetical modification.
    In certain embodiments, the parenthetical modification of crRNAs having a motif with a parenthetical modification listed in the table above is replaced with a different modified or unmodified nucleobase or internucleoside linkage.
    The number subscripts in the table above indicate the number of contiguous nucleosides that comprise the identified modification.
    The lack of a number subscript indicates one nucleoside.
    The motifs listed in the table “above may be used with any crRNA nucleobase sequence and with any internucleoside linkage motif.
    In certain embodiments, all of the nucleobases arc unmodified.
    In certain embodiments, at least one nucleobase is a 5-methylcytosine modified nucleobase.
    In certain embodiments, the internucleoside linkages are all selected independently from among phosphate and phosphorothioate.
    In certain embodiments, one or more internucleoside linkages is a neutral internucleoside linkage.
  • C. Certain Conjugate Groups and Terminal Groups
  • In certain embodiments, oligonucleotides for use as crRNA or scrRNA further comprise conjugate groups and/or terminal groups. In certain embodiments, compounds comprising oligonucleotides for use as crRNA or scrRNA further comprise a conjugate group or terminal group. In certain such embodiments, oligonucleotides are covalently attached to one or more conjugate group. In certain embodiments, conjugate groups modify one or more properties of the attached oligonucleotide, including but not limited to pharmacodynamics, pharmacokinetics, stability, binding, absorption, cellular distribution, cellular uptake, charge and clearance. In certain embodiments, conjugate groups impart a new property on the attached oligonucleotide, e.g., fluorophores or reporter groups that enable detection of the oligonucleotide. Conjugate groups and/or terminal groups may be added to oligonucleotides having any of the modifications or motifs described above.
  • Conjugate groups include, without limitation, intercalators, reporter molecules, polyamines, polyamides, peptides, carbohydrates, vitamin moieties, polyethylene glycols, thioethers, polyethers, cholesterols, thiocholesterols, cholic acid moieties, folate, lipids, phospholipids, biotin, phenazine, phenanthridine, anthraquinone, adamantane, acridine, fluoresceins, rhodamines, coumarins, fluorophores, and dyes. Certain conjugate groups have been described previously, for example: cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Lett., 1994, 4, 1053-1060), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660, 306-309; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3, 2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20, 533-538), an aliphatic chain, e.g., do-decan-diol or undecyl residues (Saison-Behmoaras et al., EMBO J., 1991, 10, 1111-1118; Kabanov et al., FEBS Lett., 1990, 259, 327-330; Svinarchuk et al., Biochimie, 1993, 75, 49-54), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethyl-ammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654; Shea et al., Nucl. Acids Res., 1990, 18, 3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237), an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., H. Pharmacol. Exp. Ther., 1996, 277, 923-937), a tocopherol group (Nishina et al., Molecular Therapy Nucleic Acids, 2015, 4, e220; doi:10.1038/mtna.2014.72 and Nishina et al., Molecular Therapy, 2008, 16, 734-740), or a GalNAc cluster (e.g., WO2014/179620).
  • In certain embodiments, a conjugate group comprises an active drug substance, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, (S)-(+)-pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, fingolimod, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indo-methicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic.
  • Conjugate groups are attached directly or via an optional conjugate linker to a parent compound, such as a crRNA or scrRNA oligonucleotide. In certain embodiments, conjugate groups are directly attached to oligonucleotides. In certain embodiments, conjugate groups are indirectly attached to oligonucleotides via conjugate linkers. In certain embodiments, the conjugate linker comprises a chain structure, such as a hydrocarbyl chain, or an oligomer of repeating units such as ethylene glycol or amino acid units. In certain embodiments, conjugate groups comprise a cleavable moiety. In certain embodiments, conjugate groups are attached to oligonucleotides via a cleavable moiety. In certain embodiments, conjugate linkers comprise a cleavable moiety. In certain such embodiments, conjugate linkers are attached to oligonucleotides via a cleavable moiety. In certain embodiments, oligonucleotides comprise a cleavable moiety, wherein the cleavable moiety is a nucleoside is attached to a cleavable internucleoside linkage, such as a phosphate internucleoside linkage. In certain embodiments, a conjugate group comprises a nucleoside or oligonucleotide, wherein the nucleoside or oligonucleotide of the conjugate group is indirectly attached to a parent oligonucleotide.
  • In certain embodiments, a conjugate linker comprises one or more groups selected from alkyl, amino, oxo, amide, disulfide, polyethylene glycol, ether, thioether, and hydroxylamino. In certain such embodiments, the conjugate linker comprises groups selected from alkyl, amino, oxo, amide and ether groups. In certain embodiments, the conjugate linker comprises groups selected from alkyl and amide groups. In certain embodiments, the conjugate linker comprises groups selected from alkyl and ether groups. In certain embodiments, the conjugate linker comprises at least one phosphorus moiety. In certain embodiments, the conjugate linker comprises at least one phosphate group. In certain embodiments, the conjugate linker includes at least one neutral linking group.
  • In certain embodiments, conjugate linkers, including the conjugate linkers described above, are bifunctional linking moieties, e.g., those known in the art to be useful for attaching conjugate groups to parent compounds, such as the crRNA oligonucleotides provided herein and the scrRNA oligonucleotides provided herein. In general, a bifunctional linking moiety comprises at least two functional groups. One of the functional groups is selected to bind to a particular site on a parent compound and the other is selected to bind to a conjugate group. Examples of functional groups used in a bifunctional linking moiety include but are not limited to electrophiles for reacting with nucleophilic groups and nucleophiles for reacting with electrophilic groups. In certain embodiments, bifunctional linking moieties comprise one or more groups selected from amino, hydroxyl, carboxylic acid, thiol, alkyl, alkenyl, and alkynyl.
  • Examples of conjugate linkers include but are not limited to pyrrolidine, 8-amino-3,6-dioxaoctanoic acid (ADO), succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC) and 6-aminohexanoic acid (AHEX or AHA). Other conjugate linkers include but are not limited to substituted or unsubstituted C1-C10 alkyl, substituted or unsubstituted C2-C10 alkenyl or substituted or unsubstituted C2-C10 alkynyl, wherein a nonlimiting list of preferred substituent groups includes hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro, thiol, thioalkoxy, halogen, alkyl, aryl, alkenyl and alkynyl.
  • In certain embodiments, a cleavable moiety is a cleavable bond. In certain embodiments, a cleavable moiety comprises a cleavable bond. In certain embodiments, a cleavable moiety is a group of atoms comprising at least one cleavable bond. In certain embodiments, a cleavable moiety comprises a group of atoms having one, two, three, four, or more than four cleavable bonds. In certain embodiments, a cleavable moiety is selectively cleaved inside a cell or subcellular compartment, such as a lysosome. In certain embodiments, a cleavable moiety is selectively cleaved by endogenous enzymes, such as nucleases.
  • In certain embodiments, a cleavable bond is selected from among: an amide, an ester, an ether, one or both esters of a phosphodiester, a phosphate ester, a carbamate, or a disulfide. In certain embodiments, a cleavable bond is one or both of the esters of a phosphodiester. In certain embodiments, a cleavable moiety comprises a phosphate or phosphodiester. In certain embodiments, the cleavable moiety is a phosphate linkage between an oligonucleotide and a conjugate linker or conjugate group.
  • In certain embodiments, a cleavable moiety is a nucleoside. In certain such embodiments, the unmodified or modified nucleoside comprises an optionally protected heterocyclic base selected from a purine, substituted purine, pyrimidine or substituted pyrimidine. In certain embodiments, a cleavable moiety is a nucleoside selected from uracil, thymine, cytosine, 4-N-benzoylcytosine, 5-methylcytosine, 4-N-benzoyl-5-methylcytosine, adenine, 6-N-benzoyladenine, guanine and 2-N-isobutyrylguanine. In certain embodiments, a cleavable moiety is 2′-deoxy nucleoside that is attached to either the 3′ or 5′-terminal nucleoside of an oligonucleotide by a phosphate internucleoside linkage and covalently attached to the conjugate linker or conjugate group by a phosphate or phosphorothioate linkage. In certain such embodiments, the cleavable moiety is 2′-deoxyadenosine.
  • Conjugate groups may be attached to either or both ends of an oligonucleotide and/or at any internal position. In certain embodiments, conjugate groups are attached to the 2′-position of a nucleoside of a modified oligonucleotide. In certain embodiments, conjugate groups that are attached to either or both ends of an oligonucleotide are terminal groups. In certain such embodiments, conjugate groups or terminal groups are attached at the 3′ and/or 5′-end of oligonucleotides. In certain such embodiments, conjugate groups (or terminal groups) are attached at the 3′-end of oligonucleotides. In certain embodiments, conjugate groups are attached near the 3′-end of oligonucleotides. In certain embodiments, conjugate groups (or terminal groups) are attached at the 5′-end of oligonucleotides. In certain embodiments, conjugate groups are attached near the 5′-end of oligonucleotides.
  • Examples of terminal groups include but are not limited to conjugate groups, capping groups, phosphate moieties, protecting groups, modified or unmodified nucleosides, and two or more nucleosides that are independently modified or unmodified.
  • In certain embodiments, a conjugate group is a cell-targeting moiety. In certain embodiments, a conjugate group, optional conjugate linker, and optional cleavable moiety have the general formula:
  • Figure US20230159919A1-20230525-C00005
  • wherein n is from 1 to about 3, m is 0 when n is 1, m is 1 when n is 2 or greater, j is 1 or 0, and k is 1 or 0.
  • In certain embodiments, n is 1, j is 1 and k is 0. In certain embodiments, n is 1, j is 0 and k is 1. In certain embodiments, n is 1, j is 1 and k is 1. In certain embodiments, n is 2, j is 1 and k is 0. In certain embodiments, n is 2, j is 0 and k is 1. In certain embodiments, n is 2, j is 1 and k is 1. In certain embodiments, n is 3, j is 1 and k is 0. In certain embodiments, n is 3, j is 0 and k is 1. In certain embodiments, n is 3, j is 1 and k is 1.
  • In certain embodiments, conjugate groups comprise cell-targeting moieties that have at least one tethered ligand. In certain embodiments, cell-targeting moieties comprise two tethered ligands covalently attached to a branching group. In certain embodiments, cell-targeting moieties comprise three tethered ligands covalently attached to a branching group.
  • In certain embodiments, the cell-targeting moiety comprises a branching group comprising one or more groups selected from alkyl, amino, oxo, amide, disulfide, polyethylene glycol, ether, thioether and hydroxylamino groups. In certain embodiments, the branching group comprises a branched aliphatic group comprising groups selected from alkyl, amino, oxo, amide, disulfide, polyethylene glycol, ether, thioether and hydroxylamino groups. In certain such embodiments, the branched aliphatic group comprises groups selected from alkyl, amino, oxo, amide and ether groups. In certain such embodiments, the branched aliphatic group comprises groups selected from alkyl, amino and ether groups. In certain such embodiments, the branched aliphatic group comprises groups selected from alkyl and ether groups. In certain embodiments, the branching group comprises a mono or polycyclic ring system.
  • In certain embodiments, each tether of a cell-targeting moiety comprises one or more groups selected from alkyl, substituted alkyl, ether, thioether, disulfide, amino, oxo, amide, phosphodiester, and polyethylene glycol, in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, ether, thioether, disulfide, amino, oxo, amide, and polyethylene glycol, in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, phosphodiester, ether, amino, oxo, and amide, in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, ether, amino, oxo, and amid, in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, amino, and oxo, in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl and oxo, in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl and phosphodiester, in any combination. In certain embodiments, each tether comprises at least one phosphorus linking group or neutral linking group. In certain embodiments, each tether comprises a chain from about 6 to about 20 atoms in length. In certain embodiments, each tether comprises a chain from about 10 to about 18 atoms in length. In certain embodiments, each tether comprises about 10 atoms in chain length.
  • In certain embodiments, each ligand of a cell-targeting moiety has an affinity for at least one type of receptor on a target cell. In certain embodiments, each ligand has an affinity for at least one type of receptor on the surface of a mammalian liver cell. In certain embodiments, each ligand has an affinity for the hepatic asialoglycoprotein receptor (ASGP-R). In certain embodiments, each ligand is a carbohydrate. In certain embodiments, each ligand is, independently selected from galactose, N-acetyl galactoseamine (GalNAc), mannose, glucose, glucosamine and fucose. In certain embodiments, each ligand is N-acetyl galactoseamine (GalNAc). In certain embodiments, the cell-targeting moiety comprises 3 GalNAc ligands. In certain embodiments, the cell-targeting moiety comprises 2 GalNAc ligands. In certain embodiments, the cell-targeting moiety comprises 1 GalNAc ligand.
  • Certain Pharmaceutical Compositions
  • In certain embodiments, the present invention provides pharmaceutical compositions comprising one or more crRNA. In certain embodiments, such pharmaceutical composition comprises a tracrRNA. In certain embodiments, the pharmaceutical composition comprises a means of expressing Cas9. In certain embodiments, such means of expressing Cas9 is a plasmid or a viral vector. In certain such embodiments, the pharmaceutical composition comprises a suitable pharmaceutically acceptable diluent or carrier. In certain embodiments, a pharmaceutical composition comprises a sterile saline solution and one or more antisense compound. In certain embodiments, such pharmaceutical composition consists of a sterile saline solution and one or more antisense compound. In certain embodiments, the sterile saline is pharmaceutical grade saline. In certain embodiments, a pharmaceutical composition comprises one or more antisense compound and sterile water. In certain embodiments, a pharmaceutical composition consists of one antisense compound and sterile water. In certain embodiments, the sterile water is pharmaceutical grade water. In certain embodiments, a pharmaceutical composition comprises one or more antisense compound and phosphate-buffered saline (PBS). In certain embodiments, a pharmaceutical composition consists of one or more antisense compound and sterile PBS. In certain embodiments, the sterile PBS is pharmaceutical grade PBS.
  • In certain embodiments, the present invention provides pharmaceutical compositions comprising one or more scrRNA. In certain embodiments, the pharmaceutical composition comprises a means of expressing a nuclease. In certain embodiments, such means of expressing the nuclease is a plasmid or a viral vector. In certain such embodiments, the pharmaceutical composition comprises a suitable pharmaceutically acceptable diluent or carrier. In certain embodiments, a pharmaceutical composition comprises a sterile saline solution and one or more antisense compound. In certain embodiments, such pharmaceutical composition consists of a sterile saline solution and one or more antisense compound. In certain embodiments, the sterile saline is pharmaceutical grade saline. In certain embodiments, a pharmaceutical composition comprises one or more antisense compound and sterile water. In certain embodiments, a pharmaceutical composition consists of one antisense compound and sterile water. In certain embodiments, the sterile water is pharmaceutical grade water. In certain embodiments, a pharmaceutical composition comprises one or more antisense compound and phosphate-buffered saline (PBS). In certain embodiments, a pharmaceutical composition consists of one or more antisense compound and sterile PBS. In certain embodiments, the sterile PBS is pharmaceutical grade PBS.
  • NONLIMITING DISCLOSURE AND INCORPORATION BY REFERENCE
  • While certain compounds, compositions and methods described herein have been described with specificity in accordance with certain embodiments, the following examples serve only to illustrate the compounds described herein and are not intended to limit the same. Each of the references, GenBank accession numbers, and the like recited in the present application is incorporated herein by reference in its entirety.
  • Although the sequence listing accompanying this filing identifies each sequence as either “RNA” or “DNA” as required, in reality, those sequences may be modified with any combination of chemical modifications. One of skill in the art will readily appreciate that such designation as “RNA” or “DNA” to describe modified oligonucleotides is, in certain instances, arbitrary. For example, an oligonucleotide comprising a nucleoside comprising a 2′-OH sugar moiety and a thymine base could be described as a DNA having a modified sugar (2′-OH for the natural 2′-H of DNA) or as an RNA having a modified base (thymine (methylated uracil) for natural uracil of RNA).
  • Accordingly, nucleic acid sequences provided herein, including, but not limited to those in the sequence listing, are intended to encompass nucleic acids containing any combination of natural or modified RNA and/or DNA, including, but not limited to such nucleic acids having modified nucleobases. By way of further example and without limitation, an oligomeric compound having the nucleobase sequence “ATCGATCG” encompasses any oligomeric compounds having such nucleobase sequence, whether modified or unmodified, including, but not limited to, such compounds comprising RNA bases, such as those having sequence “AUCGAUCG” and those having some DNA bases and some RNA bases such as “AUCGATCG” and oligomeric compounds having other modified or naturally occurring bases, such as
  • “ATmCGAUCG,” wherein mC indicates a cytosine base comprising a methyl group at the 5-position.
  • EXAMPLES
  • The following examples illustrate certain embodiments of the present invention and are not limiting. Moreover, where specific embodiments are provided, the inventors have contemplated generic application of those specific embodiments. For example, disclosure of an oligonucleotide having a particular motif provides reasonable support for additional oligonucleotides having the same or similar motif. And, for example, where a particular high-affinity modification appears at a particular position, other high-affinity modifications at the same position are considered suitable, unless otherwise indicated. As a further example, the motifs of crRNA described herein can also be applied to scrRNAs. In particular, motifs of the DNA recognition portions of the crRNAs described herein may be applied to the scrRNA target recognition portions of scrRNAs. Similarly, motifs of the tracrRNA recognition portions of the crRNAs described herein may be applied to the nuclease recognition portions of scrRNAs.
  • Example 1: Gene Editing Effects of Modified crRNA on the hLDLR Locus
  • Modified crRNAs comprising a DNA recognition portion that is complementary to hLDLR were designed and synthesized to test their effects on gene editing of the human LDLR locus. HEK 293T cells were transfected with a plasmid expressing Cas9 protein and tracrRNA using Lipofectamine 3000 (Life Technologies). Alternatively, cells were transfected with a plasmid expressing Cas9 protein and a highly active sgRNA as a positive control or no Cas9 (“Cas9 Ctrl”) as a negative control. Six hours later, cells were washed one time with PBS and transfected with a crRNA described in the table below using RNAiMAX (Life Technologies) or with no crRNA as a control (“neg ctrl”). 48 hours following the second transfection, genomic DNA was isolated from cells and used in a SURVEYOR assay (Integrated DNA Technologies) according to the manufacturer's directions. The PCR primers used to amplify the crRNA target site were forward: 5′-GGAGACCCAAATACAACAAATC-3′ (SEQ ID NO: 1) and reverse: 5′-CTAGACTCCGTCTCAAAGAAG-3′ (SEQ ID NO: 2). Following Cell cleavage, the DNA was run on a gel to analyze the extent of gene editing of hLDLR (see FIG. 1 ). Quantification was performed using Image J software, and the indel incidence percentage was calculated using the following formula: indel (%)=100×(1−(1−fraction cut of target gene)0.5), wherein the fraction cut of target gene was determined by dividing the fluorescent signal of the cut target gene fragment(s) by the total fluorescent signal of the cut and intact target gene fragment(s). The indel incidence for each modified crRNA was normalized to the indel incidence of the positive control sgRNA, referred to as the gene disruption percentage. The results, shown in the table below, indicate that the phosphorothioate modified crRNA was more active than the unmodified crRNA, and the phosphorothioate and 2′-O-methyl modified crRNA was even more active than the crRNA that does not comprise sugar modifications.
  • TABLE 1
    crRNA targeting hLDLR
    Gene
    disruption,
    normalized SEQ
    to sgRNA ID
    Name Sequence (5′ to 3′) (%) NO.
    crRNA GCGCCTTGCTCCTCGCCGCGG 7 5
    GUUUUAGAUCUAUGCUGUUUU
    G
    PS crRNA GsCsGsCsCTsTsGsCsTsCsCsTs 33 5
    CsGsCsCsGsCsGsGs GsUsUsUsUs
    AsGsAsUsCsUsAsUsGsCsUsGsU
    UUsUsG
    PS 2′-OMe GmsCmsGmCmsCmTsTsGsCsTsCs 47 5
    crRNA CsTsCsGsCsCsGsCsGs GsGsUs
    UsUsUsAsGsAsUsCsUsAsUsGs
    CsUsGsUmsUmsUmsUmsGm
    Subscripts: “m” indicates a 2′-O-methyl modification, “s” indicates a pbosphorothioate internucleoside linkage. The underlined nucleosides represent the DNA recognition portion of the crRNA, the nucleosides that are not underlined represent the tracrRNA recognition portion of the crRNA.
  • Example 2: Gene Editing Effects of Modified crRNA on the hVEGFA Locus
  • Modified crRNAs comprising a DNA recognition portion that is complementary to hVEGFA were designed and synthesized to test their effects on gene editing of the human VEGFA locus. HEK 293T cells were transfected as described in Example 1 using a crRNA described in the table below. The SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the crRNA target site were forward: 5′-TCCAGATGGCACATTGTCAG-3′ (SEQ ID NO: 3) and reverse: 5′-AGGGAGCAGGAAAGTGAGGT-3′ (SEQ ID NO: 4). Following CelI cleavage, the DNA was run on a gel to analyze the extent of gene editing of hVEGFA (see FIG. 2 ), and the gel was quantified as described in Example 1. The results for the modified crRNAs were normalized to a positive control sgRNA targeted to hVEGFA to determine the gene disruption percentage shown in the table below. The results indicate that many of the modified crRNAs were active.
  • TABLE 2
    crRNA targeting hVEGFA
    Gene
    disruption,
    normalized SEQ
    Isis to ID
    No. Sequence (5′ to 3′) sgRNA (%) NO.
    762453 Gfs Grs Ufs Grs Afs Grs Ufs Grs Afs Grs Ufs <1 6
    Grs Ufs Grs Ufs Grs Cfs Grs Ufs Grs  Grs
    Urs Urs Urs Urs Ars Grs Ars Grs Crs Urs
    Ars Urs Grs Crs Urs Grs Urs Urs Urs Urs
    Gr
    762454 Gfs Gfs Ufs Gfs Afs Gfs Ufs Gfs Afs Gfs Urs 14 6
    Grs Urs Grs Urs Grs Crs Grs Urs Grs  Grs
    Urs Urs Urs Urs Ars Grs Ars Grs Crs Urs
    Ars Urs Grs Crs Urs Grs Urs UTS Urs Urs
    Gr
    762455 Grs Grs Urs Grs Ars Grs Urs Grs Ars Grs Urs 18 6
    Grs Urs Grs Urs Grs Crs Grs Urs Grs  Grs
    Urs Urs Urs Urs Ars Grs Ars Grs Crs Urs
    Afs Urs Gfs Crs Ufs Grs Ufs Urs Uft Urs
    Gf
    762456 Gfs Grs Grs Urs Grs Ars Grs Urs Grs Ars 19 7
    Grs Urs Gra Urs Grs Urs Grs Crs Grs Urs
    Grs
    Grs Urs Urs Urs Urs Ars Grs Ars Grs Crs Urs
    Ars Urs Grs Crs Urs Grs Urs Urs Urs Urs Gr
    762457 Grs Grs Urs Grs Ars Grs Urs Grs Ars Grs Urs 29 6
    Grs Urs Grs Urs Grs Crs Grs Urs Grs  Grs
    Urs Urs Urs Urs Ars Grs Ars Grs Crs Urs Ars
    Urs Grs Crs Urs Grs Urs Urs Urs Urs Gf
    762458 Gfs Grs Grs Urs Grs Ars Grs Urs Grs Ars Grs 18 7
    Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs
    Grs Urs Urs Urs Urs Ars Grs Ars Grs Crs Urs
    Als Urs Grs Crs Urs Grs Urs Urs Urs Urs Gf
    762461 Gms Grs Urs Grs Ams Grs Urs Grs Ams Grs Urs 40 6
    Grs Urs Grs Urs Grs Crs Grs Urs Grs  Grs
    Urs Urs Urs Urs Ars Grs Ars Grs Crs Urs Ars
    Urs Grs Crs Urs Grs Urs Urs Urs Urs Gd
    Subscripts: “m” indicates a 2′-O-methyl modification, “s” indicates a phosphorothioate internucleoside linkage, “f” indicates a 2′-F modification, “r” indicates an unmodified 2′-hydroxy sugar moiety, and “d” indicates an unmodified 2′-deoxy sugar moiety. The underlined nucleosides represent the DNA recognition portion of the crRNA. the nucleosides that arc not underlined represent the tracrRNA recognition portion of the crRNA.
  • Example 3: Gene Editing Effects of Modified crRNA on the hVEGFA Locus
  • Modified crRNAs comprising a DNA recognition portion that is complementary to hVEGFA were designed and synthesized to test their effects on gene editing of the human VEGFA locus. HEK 293T cells were transfected as described in Example 1 using a crRNA described in the table below, and the Cas9/tracrRNA load time was 24 hours. The SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the crRNA target site were forward: 5′-TCCAGATGGCACATTGTCAG-3′ (SEQ ID NO: 3) and reverse: 5′-AGGGAGCAGGAAAGTGAGGT-3′ (SEQ ID NO: 4). Following Cell cleavage, the DNA was run on a gel to analyze the extent of gene editing of hVEGFA (see FIG. 3 ), and the gel was quantified as described in Example 1. The results for the modified crRNAs were normalized to a positive control sgRNA targeted to hVEGFA to determine the gene disruption percentage shown in the table below. The results indicate that many of the modified crRNAs were active or very active.
  • TABLE 3
    crRNA targeting hVEGFA
    Gene
    disruption, SEQ
    Isis normalized to ID
    No. Sequence (5′ to 3′) sgRNA (%) NO.
    801193 Gfs Gfs Ufs Gfs Afs Gfs Ufs Gfs Afs Gfs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Grs Urs 75 8
    Urs Urs Urs Ars Grs Ars Gks Crs Urs Ars Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    801197 Gfs Gfs Ufs Gfs Afs Gfs Ufs Gfs Afs Gfs Urs Grs Urs Gks Urs Gks Crs Grs Urs Grs  Gks Urs <1 8
    Urs Urs Urs Aks Grs Ais Gks Crs Urs Ars Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    801198 Gks Gfs Ufs Gfs Afs Gfs Ufs Gfs Afs Grs Urs Grs Urs Gks Uks Gks Crs Grs Urs  Grs Gks <1 8
    Urs Urs Urs Urs Aks Grs Ars Gks Crs Urs Ars Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    801199 Gms Gfs Ums Gfs Ams Gfs Ums Gfs Ams Gfs Urs Grs Urs Grs Urs Gis Crs Grs Urs Grs  Grs 65 8
    Urs Urs Urs Urs Ars Grs Ars Gks Crs Urs Ars Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    801200 Gms Gfs Ums Gfs Ams Gfs Ums Gfs Ams Gfs Urs Grs Urs Gks Urs Gks Crs Grs Urs Grs  Gks <1 8
    Urs Urs Urs Urs Aks Grs Ars Gks Crs Urs Ars Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    801201 Gks Gfs Ums Gfs Ams Gfs Ums Gfs Ams Gfs Urs Grs Urs Gks Urs Gks Crs Grs Urs Grs  Gks <1 8
    Urs Urs Urs Urs Aks Grs Ars Gks Crs Urs Ars Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    801213 Gms Gfs Ums Gfs Ams Gfs Ums Gfs Ams Gfs Ums Gfs Ums Gfs Ums Gfs Cms Gfs Ums Gfs <1 6
    Gms Ufs Ums Ufs Ums Afs Gms Afs Gms Cfs Ums AfS Ums Gfs Cms Ufs Gms Ufs Ums Ufs
    Ums Gm
    801214 Gms Gfs Umo Gfs Amo Gfs Umo Gfs Amo Gfs Umo Gfs Umo Gfs Umo Gfs Cmo Gfs Umo <1 6
    Gfs Gmo Ufs Umo Ufs Umo Afs Gmo Afs Gmo Cfs Umo Afs Umo Gfs Cmo Ufs Gms Ufs
    Ums
    801216 Gks Gds Tds Gds Aks Gds Tds Gds Ads Gds Tds Gks Tds Gds Tds Gks Cds Gds Tds Gks  Gds <1 9
    Tds Tds Tds Tds Aks Gds Ads Gks Cds Tds Aks Tds Gds Cds Tks Gds Tds Tds Tds Tks Gk
    801219 Gms Gms Ums Gds Ads Gds Ums Gms Ams Gds Tds Gds Ums Gms Ums Gms Cds Gds Tds <1 10
    Gms Gms Ums Ums Tds Tds Ads Gms Ams Gms Cds Tds Ads Ums Gms Cms Tds Gds Tds
    Ums Ums Ums Gm
    801220 Ges Ges Tes Gds Ads Gds Tes Ges Aes Gds Tds Gds Tes Ges Tes Ges Cds Gds Tds Ges  Ges <1 9
    Tes Tes Tds Tds Ads Ges Aes Ges Cds Tds Ads Tes GesmCes Tds Gds Tds Tes Tes Tes Ge
    801222 Gms Gms Ums Gfs Afs Gfs Ums Gms Ams Gfs Ufs Gfs Ums Gms Ums Gms Cfs Gfs Ufs Gms <1 6
    Gms Ums Ums Ufs Ufs Afs Gms Ams Gms Cfs Ufs Afs Ums Gms Cms Ufs Gfs Ufs Ums Ums
    Ums
    801225 Gks Gds Tds Gds Aks Gds Gds Aks Gds Tds Gks Tds Gds Tds Gks Cds Gds Tds Gks  Gds <1 9
    Tds Tds Tks Tds Ads Gds Aks Gks Cds Tds Aks Tds Gds Cds Tks Gds Tds Tds Tks Tks Gk
    Subscripts: “m” indicates a 2′-O-methyl modification, “s” indicates a phosphorothioate interucleoside linkage, “f” indicates a 2′-F modification, “r” indicates an unmodified 2′-hydroxy sugar moiety, “d” indicates an unmodified 2′-deoxy sugar moiety, “e” indicates a 2′-MOE modification, “o” indicates a phosphate intemucleoside linkage, and “k” indicates a cEt modification. Superscript “m” indicates a 5-methyl modification of the nucleobase. The underlined nucleosides represent the DNA recognition portion of the crRNA, the nucleosides that are not underlined represent the tracrRNA recognition portion of the crRNA.
  • VEGFA targeting, modified crRNAs comprising a DNA recognition portion that is under 20 nucleosides in length and/or a tracrRNA recognition portion that is under 22 nucleosides in length were designed and synthesized to test their effects on gene editing of the human VEGFA locus. HEK 293T cells were transfected as described in Example 1 using a crRNA described in the table below. The SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the crRNA target site were forward: 5′-TCCAGATGGCACATTGTCAG-3′ (SEQ ID NO: 3) and reverse: 5′-AGGGAGCAGGAAAGTGAGGT-3′ (SEQ ID NO: 4). Following CelI cleavage, the DNA was run on a gel to analyze the extent of gene editing of hVEGFA (see FIG. 3 ). The experiment was repeated, and the resulting gel was quantified as described in Example 1. The results for the modified crRNAs were normalized to a positive control sgRNA targeted to hVEGFA to determine the gene disruption percentage shown in the table below. The results indicate that many of the shortened, modified crRNAs were active, including crRNAs that comprise only a 12 nucleoside tracrRNA recognition portion and only a 17 nucleoside DNA recognition portion.
  • TABLE 4
    crRNA targeting hVEGFA
    Gene
    disruption,
    normalized SEQ
    Isis to sgRNA ID
    No. Sequence (5′ to 3′) (%) NO.
    801377 Gfs Gfs Ufs Grs Afs Gfs Ufs Gfs Afs 42 11
    Gfs Urs Grs Urs Grs Urs GrsmCks
    Grs Urs Grs  Gks Urs Urs Urs Tks
    Aks Grs Ars Gks Crs Tks Ak
    801379 Gms Gfs Ums Grs Ams Gfs Ums Gfs <1 12
    Ams Grs Ums Grs Ums Gfs Ums Gfs
    Cms Gfs Ums Gfs  Gms Ufs Ums Ufs
    Ums Afs Gms Afs Gms Cfs Ums Am
    801381 Gfs Afs Gfs Ufs Gfs Afs Gfs Urs 42 13
    Grs Urs Grs Urs GrsmCks Grs Urs
    Grs  Gks Urs Urs Urs Tks Ars Grs
    Ars Gks Crs Tks Ak
    801382 Gms Afs Gms Ufs Gms Afs Gms Urs 64 13
    Grs Urs Grs Urs GrsmCks Grs Urs
    Grs Gks Urs Urs Urs Tks Ars Grs
    Ars Gks Cre Tks Ak
    801383 Gms Afs Gms Ufs Gms Afs Gms Ufs <1 14
    Gms Ufs Gms Ufs Gms Cfs Gms Ufs
    Gms  Gfs Ums Ufs Ums Ufs Ams
    Subscripts: “m” indicates a 2′-O-methyl modification, “s” indicates a phosphorothioate interucleoside linkage, “f” indicates a 2′-F modification, “r” indicates an unmodified 2′-hydroxy sugar moiety, “d” indicates an unmodified 2′-deoxy sugar moiety, and “k” indicates a cEt modification. Superscript “m” indicates a 5-methyl modification of the nucleobase. The underlined nucleosides represent the DNA recognition portion of the crRNA, the nucleosides that are not underlined represent the tracrRNA recognition portion of the crRNA.
  • Example 5: crRNA Modification Motifs
  • Modified crRNAs having the motifs described in the table below can be used for any crRNA nucleobase sequence. The first 17 to 20 nucleosides of each motif represent the DNA recognition portion of the crRNA, and the remaining 12 to 22 nucleosides of each motif represent the tracrRNA recognition portion of the crRNA. The motifs labeled “29-mers” contain 29 linked nucleosides, and the motifs labeled “42-mers” contain 42 linked nucleosides. The motifs described below can also be applied to crRNAs of other lengths, wherein the pattern is extended or shortened as required to fit the oligonucleotide length. The modifications of the motifs are described using the same single letter identifiers used in the subscripts of Tables 1-4 above. The number subscripts indicate the number of contiguous nucleosides that comprise the identified modification. The lack of a number subscript indicates one nucleoside. Additional abbreviations are: “1” indicates an LNA modification, “(MOP)” indicates a methoxypropyl modified internucleoside linkage, “(MP)” indicates a methylphosphonate internucleoside linkage, “(MMI)” indicates an MMI N-methyl internucleoside linkage, “(5-propyne)” indicates a 5-propyne nucleobase modification, and “(G-clamp)” indicates a G-clamp modified nucleobase.
  • TABLE 5
    crRNA modification motifs
    29-mers 42-mers
    f7r6kr3kr3kr3krk2 f10r18kr4kr2kr3k2
    mf6r6kr3kr3kr3krk2 mf9r18kr4kr2kr3k2
    mf6r10k6r2k4 mr27kr4kr2kr3k2
    mr16k6r2k4 mr9f10k6r2kr4kr2kr3k2
    mr6f10k6r2k4 mr9f10I6r2kr4kr2kr3k2
    mf6r10f6r2k4 mr9f16r2kr4kr2kr3k2
    mf6r10Ier2k4 mr32kr2kr3k2
    mr6f10I6r2k4 mf9r18kr4kr2kr3k2
    mf6r10k6r2I4 r(MOP)f9r18kr4kr2kr3k2
    mr16k6r2I4 d(MOP)f9r18kr4kr2kr3k2
    mr6f10k6r2I4 f(MOP)f9r18kr4kr2kr3k2
    mf6r10f6r2I4 r(MP)f9r18kr4kr2kr3k2
    r(MOP)f6r6kr3kr3kr3krk2 d(MP)f9r18kr4kr2kr3k2
    d(MOP)f6r6kr3kr3kr3krk2 f(MP)f9r18kr4kr2kr3k2
    f(MOP)frgkrakrakrakrk2 r(MMI)f9r18kr4kr2kr3k2
    r(MP)f6r6kr3kr3kr3krk2 d(MMI)f9r18kr4kr2kr3k2
    d(MP)f6r6kr3kr3kr3krk2 f(MMI)f9r18kr4kr2kr3k2
    f(MP)f6r6kr3kr3kr3krk2 mr32kr2k(G-Clamp)r2k2
    r(MOP)f6r10k6r2k4 mr27k3r2kr2kr3k2
    d(MOP)f6r10k6r2k4 mf9r18k3r2kr2kr3k2
    f(MOP)f6r10k6r2k4 mf9r11(5-Propyne-U)4
    r3k3r2kr2kr3k2
    r(MP)f6r10k6r2k4 29-mers
    r(MP)f6r10k6r2k4 d(MOP)r6f10k6r2k4
    r(MP)f6r10k6r2k4 f(MOP)r6f10k6r2k4
    r(MOP)r16k6r2k4 r(MP)r6f10k6r2k4
    d(MOP)r16k6r2k4 d(MOP)r6f10k6r2k4
    f(MOP)r16k6r2k4 f(MOP)r6f10k6r2k4
    r(MP)r16k6r2k4 r(MOP)f6r10I6r2k4
    d(MP)r16k6r2k4 d(MOP)f6r10I6r2k4
    f(MP)r16k6r2k4 f(MOP)f6r10I6r2k4
    r(MOP)r6f10k6r2k4 r(MOP)f6r10I6r2k4
    d(MP)f6r10k6r2I4 d(MOP)f6r10f6r2I4
    f(MP)fgr10kg+U f(MOP)f6r10f6r2I4
    r(MOP)r16k6r2I4 f7r6kr3kr3kr3k(G-Clamp)k2
    d(MOP)r16k6r2I4 mf6r6kr3kr3kr3k(G-Clamp)rk2
    f(MOP)r16k6r2I4 mf2r10kr2k(G-Clamp)k2
    r(MP)r16k6r2I4 mr16kr2k(G-Clam)k2
    d(MP)r16k6r2I4 mr6f10kr2k(G-Clamp)k2
    f(MP)r16k6r2I4 mf6r10fr2k(G-Clamp)K2
    r(MOP)r6f10k6r2I4 mf6r10I2r2k(G-Clamp)k2
    r(MOP)r6f10k6r2I4 mr6f10Ir2k(G-Clamp)k2
    r(MOP)r6f10k6r2I4 mf6r10ker2I(G-Clamp)I2
    r(MOP)r6f10k6r2I4 mr16k6r2I(G-Clamp)I2
    r(MOP)r6f10k6r2I4 mr6f10kr2I(G-Clamp)I2
    r(MOP)r6f10k6r2I4 mf6r10f6r2I(G-Clamp)I2
    r(MOP)f6r10f6r2I4 f7r6kr3k(5-propyne)r3kr3krk2
    dfMOP)f6r10f6r2I4 mf6r6kr3k(5-Propyne)r3kr3krk2
    f(MOP)f6r10f6r2I4 r(MOP)f6r10f6r2I4
  • Example 6: Gene Editing Effects of Modified crRNA on the hVEGFA Locus
  • Modified crRNAs comprising a DNA recognition portion that is complementary to hVEGFA were designed and synthesized to test their effects on gene editing of the human VEGFA locus. HEK 293T cells were transfected as described in Example 1 using a crRNA described in the table below. The SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the crRNA target site were forward: 5′-TCCAGATGGCACATTGTCAG-3′ (SEQ ID NO: 3) and reverse: 5′-AGGGAGCAGGAAAGTGAGGT-3′ (SEQ ID NO: 4). Following CelI cleavage, the DNA was run on a gel to analyze the extent of gene editing of hVEGFA, and the gel was quantified as described in Example 1. The results for the modified crRNAs were normalized to a positive control sgRNA targeted to hVEGFA to determine the gene disruption percentage shown in the table below. The results indicate that many of the modified crRNAs were active and some were even more active than the sgRNA positive control.
  • TABLE 6
    crRNA targeting hVEGFA
    Gene
    disruption,
    normalized
    Isis to SEQ ID
    No. Sequence (5′ to 3′) sgRNA (%) NO.
    834463 Gms Grs Urs Grs Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Grs 6
    Urs Urs Urs Urs Ars Grs Ars Grs Crs Urs Ars Urs Grs Crs Urs Grs Ums Ums Ums Ums Gm
    834464 Grs Grs Urs Grs Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Crs  63 8
    Urs Urs Urs Urs Ars Grs Ars Gks Crs Urs Ars Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    834465 Gms Grs Urs Grs Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Grs  93 8
    Urs Urs Urs Urs Ars Grs Ars Gks Crs Urs Ars Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    834466 Gfs Gfs Ufs Gfs Afs Gfs Ufs Gfs Afs Gfs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks  61 15
    Urs Urs Urs Tks Ars Grs Ars Gks Crs Tks Aks Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    834467 Grs Grs Urs Grs Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks  57 15
    Urs Urs Urs Tks Ars Grs Ars Gks Crs Tks Aks Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    834468 Gms Grs Urs Grs Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks  38 15
    Urs Urs Urs Tks Ars Grs Ars Gks Crs Tks Aks Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    834469 Gfs Gfs Ufs Gfs Afs Gfs Ufs Gfs Afs Gfs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks  68 11
    Urs Urs Urs Tks Ars Grs Ars Gks Crs Tks Ak
    834470 Grs Grs Urs Grs Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks  75 11
    Urs Urs Urs Tks Ars Grs Ars Gks Crs Tks Ak
    834471 Gms Grs Urs Grs Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks  <1 11
    Urs Urs Urs Tks Ars Grs Ars Gks Crs Tks Ak
    834472 Gfs Afs Gfs Ufs Gfs Afs Gfs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks Urs Urs Urs 107 13
    Tks Ars Grs Ars Gks Crs Tks Ak
    834475 Gfs Afs Gfs Ufs Gfs Afs Gfs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks Urs Urs Urs  <1 16
    Tks Ars Grs Ars Gks Crs Tks Ak
    834476 Gms Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks Urs Urs Urs  71 13
    Tks Ars Grs Ars Gks Crs Tks Ak
    834477 Grs Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks Urs Urs Urs  67 13
    Tks Ars Grs Ars Gks Crs Tks Ak
    834478 Gms Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks Urs Tks Urs  <1 16
    Tks Ars Grs Ars Gks Crs Tks Ak
    834479 Grs Ars Grs Urs Grs Ars Grs Urs Grs Urs Grs Urs Grs Crs Grs Urs Grs  Gks Urs Tks Urs  <1 16
    Tks Ars Grs Ars Gks Crs Tks Ak
    Subscripts: “m” indicates a 2′-O-methyl modification, “s” indicates a phosphorothioate interucleoside linkage, “f” indicates a 2′-F modification, “r” indicates an unmodified 2′-hydroxy sugar moiety, “d” indicates an unmodified 2′-deoxy sugar moiety, and “k” indicates a cEt modification. The underlined nucleosides represent the DNA recognition portion of the crRNA, the nucleosides that are not underlined represent the tracrRNA recognition portion of the crRNA.
  • Example 7: Off-Target Effects of Modified crRNAs
  • In order to test the off-target effects of modified crRNAs, Isis Numbers 801193 (Example 3), 801381 (Example 4), and 834472 (Example 6) were tested for their effects on gene editing of the human Myc-associated factor X (MAX) locus. At chromosome position 14q23, the MAX gene carries an 18 out of 20 nucleotide match to a portion of or all of the region of the VEGFA gene targeted by Isis Numbers 801193, 801381, and 834472. HEK 293T cells were transfected as described in Example 1 using Isis No. 801193, 801381, or 834472 as the modified crRNA. The SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the off-target site in the MAX gene were forward: 5′-TACCCGGGCCGTCTGTTAGA-3′ (SEQ ID NO: 17) and reverse: 5′-GAGGGGGAAGTCACCGACAA-3′ (SEQ ID NO: 18). Following Cell cleavage, the DNA was run on a gel to analyze the extent of gene editing of MAX. Quantification was performed as described in Example 1. The results for the modified crRNAs were normalized to a positive control sgRNA targeted to hVEGFA to determine the gene disruption percentage shown in the table below. The results indicate that the modified crRNAs exhibited less off-target effects than the sgRNA control. The on-target effects of the modified crRNAs (see Examples 3, 4, and 6) are shown in the third column below, for comparison.
  • TABLE 7
    Effect of crRNA targeting VEGFA on off-target MAX
    On-target gene
    Off-target gene disruption, normalized
    disruption, normalized to sgRNA (%, from
    Isis No. to sgRNA (%) above examples)
    801193 28 75
    801381 13 42
    834472 20 107
  • Example 8: Gene Editing Effects of Modified crRNA on hTTR Locus
  • Modified crRNAs comprising a DNA recognition portion that is complementary to human TTR were designed and synthesized to test their effects on gene editing of the hTTR locus. HEK 293T cells were transfected as described in Example 1 using a crRNA described in the table below. The SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the crRNA target site were forward: 5′-GCTGACTAAGCAAAGCTTCCAAATGAC-3′ (SEQ ID NO:41) and reverse: 5′-GATGTCACAGAAACACTCACCGTAG-3′ (SEQ ID NO: 42). Following Cell cleavage, the DNA was run on a gel to analyze the extent of gene editing of hTTR, and the gel was quantified as described in Example 1. The results for the modified crRNAs were normalized to a positive control sgRNA targeted to hTTR to determine the gene disruption percentage shown in the table below. The results indicate that many of the modified crRNAs were active and some were even more active than the sgRNA positive control.
  • TABLE 8
    crRNA targeting hTTR
    Gene
    disruption,
    normalized SEQ
    Name or to sgRNA ID
    Ion No. Sequence (5′ to 3′) (%) NO.
    42RTT Gms Ars Crs Ars Ars Grs Grs Urs Urs Crs Ars Urs 125 43
    MAS Ars Urs Urs Urs Grs Urs Ars Urs  Grs Urs Urs
    Urs Urs Ars Ars Grs Crs Urs Ars Urs Grs Crs
     Urs Grs Ums Ums Ums Ums Gm
    895589 Gfs Afs Cfs Afs Afs Gfs Gfs Ufs Ufs Cfs As 118 44
    Urs Ars Ums Ums Ums Grs Ums Ams Urs  Grs Urs
    Urs Urs Urs Ars Grs Ars Gks Crs Urs Ars Urs
    Gks Crs Urs Gks Urs Urs Urs Tks Gk
    895591 G fs  A fS  C fs  A fS  A fs  G fs  G fs  U fs  U fs  C fsA rs  77 45
    U rs  A rs  U rs  U rs  U rs  G rs  U rs A rs  U rs Gks
    Urs Urs Urs Tks Ars Grs Ars Gks Crs Tks Ak
    895593 Gfs Afs Cfs Afs Afs Gfs Gfs Ufs Ufs Cfs Ars 107 46
    Urs Ars Urs Urs Urs Grs Urs Ars Urs  Gks Urs
    Urs Urs Tks Ars Grs Ars Gks Crs Urs Aks Urs
    Grs Crs Urs Gks Urs Urs Urs Tks Gk
    Subscripts: “m” indicates a 2′-O-methyl modification, “s” indicates a phosphorothioate interucleoside linkage, “f” indicates a 2′-F modification, “r” indicates an unmodified 2′-hydroxy sugar moiety. The underlined nucleosides represent the DNA recognition portion of the crRNA, the nucleosides that are not underlined represent the tracrRNA recognition portion of the crRNA.
  • Example 9: Gene Alteration Effects of Truncated scrRNA
  • Truncated scrRNAs comprising a scrRNA target recognition portion that is complementary to DNA (cytosine-5)-methyltransferase 1 (DNMT1) were designed and synthesized to test their effects on alteration of the DNMT1 gene. HEK293T cells were transfected with a plasmid encoding Cpf1 and a double-stranded gblock (IDT, Coralville, Iowa) encoding a scrRNA listed in the table below. The SURVEYOR assay was performed as described in Example 1, and the PCR primers used to amplify the scrRNA site in the DNMT1 gene were forward: 5′-CTGGGACTCAGGCGGGTCAC-3′ (SEQ ID NO: 47) and reverse: 5′-CCTCACACAACAGCTTCATGTCAGC-3′ (SEQ ID NO:). Following Cell cleavage, the DNA was run on a gel to analyze the extent of gene alteration of DNMT1. The results are shown in FIGS. 4 a and 4 b . The results indicate that multiple truncated scrRNAs, including scrRNA containing only 36 nucleosides, altered the target gene.
  • TABLE 9
    scrRNA targeting DNMT1
    SEQ
    ID
    Name Sequence (5′ to 3′) NO.
    002 TAATTTCTACTCTTG
    TAGATCTGATGGTCC
    ATGTCTGTTACTC 49
    005 TTCTACTCTTGTAGA
    TCTGATGGTCCATGT
    CTGTTACTC 50
    006 TAATTTCTACTCTTG
    TAGATCTGATGGTCC
    ATGTCTGTTA 51
    007 TTCTACTCTTGTAGA
    TCTGATGGTCCATGT
    CTGT 52
    008 TAATTTCTACTCTTG
    TAGATCTGATGGTCC
    ATGTCTGT 53
    009 TTCTACTCTTGTAGA
    TCTGATGGTCCATGT
    CTGTTA
    54
    010 AATTTCTACTCTTGT
    AGATCTGATGGTCCA
    TGTCTGT 55
    011 ATTTCTACTCTTGTA
    GATCTGATGGTCCAT
    GTCTGT 56
    012 TTTCTACTCTTGTAG
    ATCTGATGGTCCATG
    TCTGT 57
    013 AATTTCTACTCTTGT
    AGATCTGATGGTCCA
    TGTCTGTTACTC 58
    014 ATTTCTACTCTTGTA
    GATCTGATGGTCCAT
    GTCTGTTACTC 59
    015 TTTCTACTCTTGTAG
    ATCTGATGGTCCATG
    TCTGTTACTC 60

    All of the nucleosides in the table above are unmodified ribonucleosides comprising 2′-hydroxy sugar moieties and phosphate internucleoside linkages. The underlined nucleosides represent the target recognition portion of the scrRNA, the nucleosides that are not underlined represent the nuclease recognition portion of the scrRNA.
  • Example 10: DNA Cutting Effects of Truncated tracrRNA
  • Truncated tracrRNAs were designed and synthesized to test their effects on editing of mouse Proprotein Convertase Subtilisin/Kexin Type 9 (Pcsk9). To generate Pcsk9 DNA, a portion of the mouse genomic locus encompassing the CRISPR target site was amplified by PCR using primers 5′-CTGAGGCTAGAGGACTGAGC-3′ (SEQ ID NO: 61) and 5′-CAGACGGCTAGATGAGCAGAG-3′ (SEQ ID NO: 62). 30 nM of a modified crRNA, Ion No. 927720, shown in the table below and 30 nM of a tracrRNA shown in the table below and were used to test for Pcsk9 gene disruption in an in vitro biochemical assay. Following cleavage by Cas9, the DNA was run on a gel to analyze the extent of activity. The results are shown in FIG. 5 . The results indicate that the truncated tracrRNAs exhibited activity in vitro.
  • TABLE 10
    Modified crRNA targeting human Pcsk9
    and truncated tracrRNAs
    Ion No. SEQ
    or ID
    Name Sequence (5′ to 3′) NO.
    927720 Ams Crs Crs Grs Crs A r s Grs Crs 63
    Crs Ars Crs Grs Crs Ars Grs Ars
    Grs Crs Ars Grs Grs  Urs Urs
    Urs Urs Ars Grs Ars Grs Crs Urs
    Ars Urs Grs Crs Urs Grs Ums Ums
    Ums Ums Gm
    tracrRNA 1.2 GTTGGAACCATTCAAAACAGCATAGCAAGT 64
    (pos.Ctrl) TAAAATAAGGCTAGTCCGTTATCAACTTGG
    CCAACATGAGGATCACCCATGTCTGCAGGG
    CCAAGTGGCACCGAGTCGGTGCTTT
    tracrRNA
     63 GGAACCATTCAAAACAGCATAGCAAGTTAA 65
    AATAAGGCTAGTCCGTTATCAACTTGAAAA
    AGT
    tracrRNA
     54 CAAAACAGCATAGCAAGTTAAAATAAGGCT 66
    AGTCCGTTATCAACTTGAAAAAGT
    Subscripts: “m” indicates a 2′-O-methyl modification, “s” indicates a phosphorothioate interucleoside linkage, “r” indicates an unmodified 2′-hydroxy sugar moiety, The underlined nucleosides represent the DNA recognition portion of the crRNA. the nucleosides that are not underlined represent the tracrRNA recognition portion of the crRNA. All of the nucleosides in the tracrRNAs shown in the table above are unmodified ribonucleosides comprising 2′-hydroxy sugar moieties and phosphate internucleoside linkages.
  • Example 11: Gene Activation Following Free Uptake of Modified crRNA
  • The ability of modified crRNAs to activate target genes was tested in a transcriptional activation assay, similar to that described in Konermann et al., Nature 517, 583-588 (2015). Briefly, one MS2 aptamer sequence was inserted at position 58 of tracrRNA. HEK 293 cells were transfected with PBS alone (negative control) or with a plasmids encoding catalytically inactive Cas9 fused to Tetrameric VP16 transcription activator domain (dCas9-VP64), MS2-p65-HSF1 activation helper protein as described in Konermann et al. and the MS2 aptamer containing tracrRNA1.2. Modified crRNA comprising a DNA recognition portion that is complementary to human TTR, listed in the table below, was added in PBS, in the absence of a transfection reagent, at a final concentration of 1 uM. PBS without crRNA was added in the “no RNA” control. After 48 hours, total RNA was isolated, and gene activation was measured using RT-qPCR using forward primer 5′-CTTGCTGGACTGGTATTTGTGTCT-3′(SEQ ID NO: 67), reverse primer 5′-AGAACTTTGACCATCAGAGGACACT-3′ (SEQ ID NO: 68) and probe 5′-CCCTACGGGCACCGGTGAATCC-3′ (SEQ ID NO: 69). The RT-qPCR results were normalized to GAPDH and are presented in the table below as the fold change relative to the negative control, which was set to 1.0. The results show that modified crRNA was taken up by the cells by free uptake and induced target gene activation.
  • TABLE 11
    Gene activation following free
    uptake of modified crRNA
    Fold
    change
    (Rel. SEQ
    Sequence to Neg ID
    Name (5′ to 3′) Ctrl) NO.
    Neg Ctrl n/a 1.0
    NoRNA n/a 2.6
    crRNA Gms Ars Crs Ars Ars 10.2 70
    42 Grs Grs Trs Trs Crs
    Ars Trs Ars Trs Trs
    Trs Grs Trs Ars Trs
    Grs Trs Trs Trs Trs
    Ars Grs Ars Grs
    Crs Trs Ars Trs Grs
    Crs Trs Grs
    Tms Tms Tms Tms Gm
    Subscripts: “m” indicates a 2′-O-methyl modification, “s” indicates a phosphorothioate interucleoside linkage, “r” indicates an unmodified 2′-hydroxy sugar moiety. The underlined nucleosides represent the DNA recognition portion of the crRNA.
  • Example 12: In Vitro Digestion of Pcsk9 DNA Using Compounds Comprising Modified crRNA
  • Compounds comprising modified crRNAs shown in the tables below comprise a DNA recognition portion that is complementary to mouse Pcsk9. The modified crRNAs shown in Table 12 below are made and tested for their DNA cutting activity and/or gene disruption activity, as described herein. The modified crRNAs shown in Table 13 were synthesized and tested for DNA cutting activity in vitro. Ion No. 927722 comprises a GalNAc conjugate group (“LICA-1”), and the synthesis of Ion No. 927722 is shown below. The DNA cutting assay was carried out as described in Example 10. Ion No. 927720 or 927722 was used with a tracrRNA. An sgRNA was used alone as a positive control. The results are shown in FIG. 6 . The results show that the modified crRNA with no attached conjugate group cut Pcsk9 DNA more potently than the sgRNA positive control in vitro. The modified crRNA attached to the GalNAc conjugate group cut Pcsk9 DNA to an extent approximately equal to that of the sgRNA positive control.
  • TABLE 12
    Modified crRNA targeting Pcsk9
    Isis or SEQ ID
    Ion No. Sequence (5′ to 3′) NO.
    881061 Afs Cfs Cfs Gfs Cfs Afs Gfs Cfs CfsA fsC rsG rsC rsA rsG rsA rsG rsC rsA rsG rs 71
    Gks Urs Urs Urs Tks Ars Grs Ars Gks Crs Tks Ak
    881063 LICA-1o-Ams Cfs Cfs Gfs Cfs Afs Gfs Cfs Cfs Afs Crs Grs Crs Ars Grs Ars Grs Crs 71
    Ars Grs  Gks Urs Urs Urs Tks Ars Grs Ars Gks Crs Tks Ak
    927719 Ars Crs Crs Grs Crs Ars Grs Crs Crs Ars Crs Grs Crs Ars Grs Ars Grs Crs Ars Grs 63
    Grs Urs Urs Urs Urs Ars Grs Ars Grs Crs Urs Ars Urs Grs Crs Urs Grs Urs Urs Urs Urs Gr
    927723 Afs Cfs Cfs Gfs Cfs Afs Gfs Cfs Cfs Afs Crs Grs Crs Ars Grs Ars Grs Crs Ars Grs 72
    Grs Urs Urs Urs Urs Ars Grs Ars Gks Crs Urs Ars Urs Gks Crs Urs Gks Urs Urs Urs Tks Gk
    927725 LICA-1o-Afs Cfs Cfs Gfs Cfs Afs Gfs Cs Cs Afs Crs Grs Crs Ars Grs Ars Grs Crs Ars 72
    Grs  Grs Urs Urs Urs Urs Ars Grs Ars Gks Crs Urs Ars Urs Gks Crs Urs Gks Urs Urs
    Urs Tks Gk
  • TABLE 13a
    crRNA targeting Pcsk9
    SEQ
    ID
    Ion No. Sequence (5′ to 3′) NO.
    927720 Ams Crs Crs Grs Crs Ars Grs Crs Crs Ars
    Crs Grs Crs Ars Grs Ars Grs Crs Ars Grs 63
    Grs Urs Urs Urs Urs Ars Grs Ars Grs Crs
    Urs Ars Urs Grs Crs Urs Grs Ums Ums Ums
    Ums Gm
    927722 LICA-1o-Ams Crs Crs Grs Crs Ars Grs Crs Crs
    Ars Crs Grs Crs Ars Grs Ars Grs Crs Ars Grs 63
    Grs Urs Urs Urs Urs Ars Grs Ars Grs Crs Urs
    Ars Urs Grs Crs Urs Grs Ums Ums Ums Ums Gm
    Subscripts: “m” indicates a 2′-O-methyl modification, “s” indicates a phosphorothioate interucleoside linkage, “f” indicates a 2′-F modification, “r” indicates an unmodified 2′-hydroxy sugar moiety, The underlined nucleosides represent the DNA recognition portion of the crRNA.
  • Figure US20230159919A1-20230525-C00006
  • Compound 3
  • To a solution of THA-GalNAc3 PFP ester 1 (10 g, 5.3 mmol), TEA (1.47 mL, 10.5 mmol) in dichloromethane (40 mL), 6-amino-1-hexanol in dichloromethane (10 mL) was added dropwise. After stirring at room temperature for 12 h the reaction mixture was concentrated and residue was purified by silica gel column (Biotage Silica Gel Colum Chromatography, 220 g) and eluted with 5-20% MeOH in dichloromethane to yield 3 (9.1 g, 94%). LR MS (ESI) calcd for C84H139O36N8 [M+H]+ m/z=1837.1. found 1837.9.
  • Compound 4
  • To a DMF (25 mL) solution of 3 (8.96 g, 5.0 mmol) and tetrazole (0.273 g, 4.0 mmol) at 0° C., 1-methylimidazole (97 μL, 1 mmol) and phosphitylating reagent (2.3 mL, 7 mmol) were added. The reaction mixture was warmed to room temperature and stirred at the temperature for 12 h. The reaction mixture was extracted with ethyl acetate (100 mL), washed with sat. NaHCO3 (100 mL) and brine (100 mL), dried over Na2SO4. After filtration the ethyl acetate solution was concentrated under reduced pressure. The residue obtained was purified by silica gel column chromatography and eluted first with ethyl acetate, then 50% acetone in ethyl acetate, followed by acetone and 50% acetone in THF to yield 4 (7.5 g, 75%) was obtained as white foam. 31P NMR (121 MHz, CDCl3): δ 147.32; LR MS (ESI) calcd for C93H154O37N10P [M−H] m/z=2035.0. Found 2034.8.
  • Synthesis of Modified crRNAs, Ion Numbers 927720 and 927722
  • Standard phosphoramidites and solid supports were used for incorporation of A, U, G, and C nucleosides. A 0.2 M solution of the amidites in anhydrous acetonitrile was used for the synthesis. A 0.2 M solution of 2′-O-Me ABz, U, Gibu and CBz phosphoramidites in anhydrous acetonitrile were used for the incorporation of 2′-O-methyl modified nucleotides. The modified crRNAs (60 μmol scale) were synthesized using an ÄKTAOligopilot synthesizer (GE Healthcare Biosciences) on VIMAD UnyLinker™ solid support (100 μmol/g loading) and the appropriate amounts of solid supports were packed in the column for synthesis. Dichloroacetic acid (6%) in toluene was used as detritylating reagent. 4,5-Dicyanoimidazole in the presence of N-methylimidazole in CH3CN was used as activator during the coupling step. 0.1 M xanthane hydride solution in 50% pyridine in acetonitrile was used as sulfurizing agent with 3 min contact time. Twelve equivalents of THA-GalNAc phosphoramidite 4 was delivered in 3 portions, each followed by a 12 min coupling wait time. All other steps in the protocol supplied by the manufacturer were used without modification. The coupling efficiencies were more than 97%. After completion of the synthesis, solid support was treated with 20% diethylamine in toluene for 45 min to remove cyanoethyl group from phosphorothioate linkages. The solid support was then suspended in aqueous ammonium hydroxide (30 wt. %): ethanol (3:1) and allowed to stir at room temperature for 4 h. To this 10% (V/V) of methylamine in water (40 wt %) was added and stirring continued at room temperature for 24 h to complete the removal of all protecting groups except TBDMS group at 2′-position. The solid support was filtered and the filtrate was concentrated to dryness. The residue obtained was re-suspended in anhydrous triethylamine trihydrofluoride/triethylamine/1-methyl-2-pyrrolidinone solution (9.75 mL of a solution of 3 mL of triethylamine trihydrofluoride, 2.25 mL triethylamine and 4.5 mL 1-methyl-2-pyrrolidinone, to provide a 1.4 M HF concentration) and heated at 65° C. for 4 h to remove the TBDMS groups at the 2′-position. The reaction was quenched with 1.5 M ammonium bicarbonate (9.95 mL) and diluted with water and purified by HPLC on a strong anion exchange column (GE Healthcare Bioscience, Source 30Q, 30 μm, 2.54×8 cm, A=100 mM ammonium acetate in 30% aqueous CH3CN, B=1.5 M NaBr in A, 0-60% of B in 28 column volume, flow 14 mL min−1). The fractions containing full length crRNAs were pooled together was desalted by HPLC on reverse phase column to yield the crRNA in an isolated yield of 10% based on solid-support loading. The oligonucleotides were characterized by ion-pair-HPLC-MS analysis with Agilent 1100 MSD system.
  • TABLE 13b
    Analytical data of modified crRNAs
    Ion No. Calcd Mass Observed Mass
    927720 14206.1 14205.9
    927722 15725.7 15724.2
  • Example 13: Gene Editing Effects of Modified crRNA on Pcsk9 Ex Vivo
  • Modified crRNA was tested for gene editing of Pcsk9 ex vivo. Hepatocytes from mice that express Cas9 (described in Platt et al., Cell 159, 440-455 (2014)) were cultured in William's media E supplemented with 10% FBS, 4 mM L-Glutamine and 25 mM HEPES. The hepatocytes were transfected with Ion No. 927720 (see Example 12) and a tracrRNA or a sgRNA positive control alone using lipofectamine RNAiMax (Life Technologies, Carlsbad). Pcsk9 gene disruption was measured using the SURVEYOR assay. The results are shown in FIG. 7 . The results indicate that a modified crRNA disrupted the Pcsk9 gene with similar potency to a sgRNA positive control in hepatocytes ex vivo.

Claims (28)

1.-293. (canceled)
294. A modified crRNA consisting of 29 to 42 nucleosides, comprising a DNA recognition portion and tracrRNA recognition portion, wherein the DNA recognition portion is located 5′ of the tracrRNA recognition portion;
the DNA recognition portion comprises at least one modified nucleoside comprising a linearly modified sugar moiety; and
the tracrRNA recognition portion comprises at least one modified nucleoside comprising a bicyclic modified sugar moiety.
295. The modified crRNA of claim 294, wherein at least one linearly modified sugar moiety in the DNA recognition portion of the modified crRNA is selected from a 2′-F and a 2′-OMe modified sugar moiety.
296. The modified crRNA of claim 295, wherein the at least one linearly modified sugar moiety in the DNA recognition portion of the modified crRNA is a 2′-F modified sugar moiety.
297. The modified crRNA of claim 294, wherein the DNA recognition portion of the modified crRNA is at the 5′-terminal of the cRNA.
298. The modified crRNA of claim 294, wherein the DNA recognition portion of the modified crRNA is at the 5′-terminal of the cRNA, and wherein each of the seven 5′-terminal nucleosides comprise linearly modified sugar moieties.
299. The modified crRNA of claim 294, wherein each linearly modified sugar moiety in the DNA recognition portion of the modified crRNA is selected from a 2′-F and a 2′-OMe modified sugar moiety.
300. The modified crRNA of claim 299, wherein the linearly modified sugar moieties of the seven 5′-terminal nucleosides are each 2′-F or 2′-OMe.
301. The modified crRNA of claim 299, wherein the modified sugar moieties of the seven 5′-terminal nucleosides alternate between 2′-F and 2′-OMe.
302. The modified crRNA of claim 294, wherein the DNA recognition portion of the modified cRNA comprises at least one nucleoside comprising an unmodified sugar moiety.
303. The compound of claim 302, wherein the unmodified sugar moiety is a 2′-hydroxy sugar moiety.
304. The modified cRNA of claim 294, wherein the DNA recognition portion of the modified cRNA comprises at least ten consecutive nucleosides each comprising unmodified 2′-hydroxy sugar moieties at the 3′-end of the DNA recognition portion.
305. The modified crRNA of claim 294, wherein the tracrRNA recognition portion of the modified crRNA comprises at least four modified sugar moieties.
306. The modified crRNA of claim 294, wherein the tracrRNA recognition portion of the modified crRNA comprises no more than five modified sugar moieties.
307. The modified crRNA of claim 294, wherein the tracrRNA recognition portion of the modified crRNA comprises exactly five modified sugar moieties.
308. The modified crRNA of claim 294, wherein the nucleoside at the 5′-end of the tracrRNA recognition portion of the crRNA comprises a modified sugar moiety.
309. The modified crRNA of claim 294, wherein each modified sugar moiety within the tracrRNA recognition portion of the modified crRNA is a bicyclic sugar moiety.
310. The modified crRNA of claim 294, wherein each bicyclic sugar moiety is selected from LNA or cEt.
311. The modified crRNA of claim 294, wherein the crRNA consists of 42 linked nucleosides.
312. The modified crRNA of claim 294, wherein the cRNA consists of 29-32 linked nucleosides.
313. The modified crRNA of claim 294, wherein the DNA recognition portion of the crRNA consists of 17 or fewer linked nucleosides.
314. The modified crRNA of claim 294, wherein the tracrRNA recognition portion of the crRNA consists of 12 or fewer linked nucleosides.
315. The modified crRNA of claim 294, wherein the nucleobase sequence of the DNA recognition portion of the cRNA is at least 90%, at least 95%, or 100% complementary to a target DNA.
316. The modified cRNA of claim 294, wherein each internucleoside linkage of the modified cRNA is selected from a phosphodiester internucleoside linkage or a phosphorothioate internucleoside linkage.
317. The modified cRNA of claim 294, wherein each internucleoside linkage of the modified cRNA is a phosphorothioate internucleoside linkage.
318. A method comprising contacting a cell with a modified cRNA of claim 294,
a tracrRNA or a plasmid encoding a tracrRNA, and
a Cas9 protein or a plasmid encoding a Cas9 protein.
319. The method of claim 318, wherein the cell is in an animal.
320. A pharmaceutical composition comprising the modified crRNA of claim 294 and a pharmaceutically acceptable carrier or diluent.
US17/850,456 2015-06-29 2022-06-27 Modified crispr rna and modified single crispr rna and uses thereof Pending US20230159919A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/850,456 US20230159919A1 (en) 2015-06-29 2022-06-27 Modified crispr rna and modified single crispr rna and uses thereof

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201562186363P 2015-06-29 2015-06-29
US201562233281P 2015-09-25 2015-09-25
US201562256649P 2015-11-17 2015-11-17
PCT/US2016/040191 WO2017004261A1 (en) 2015-06-29 2016-06-29 Modified crispr rna and modified single crispr rna and uses thereof
US201715737723A 2017-12-18 2017-12-18
US17/850,456 US20230159919A1 (en) 2015-06-29 2022-06-27 Modified crispr rna and modified single crispr rna and uses thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2016/040191 Continuation WO2017004261A1 (en) 2015-06-29 2016-06-29 Modified crispr rna and modified single crispr rna and uses thereof
US15/737,723 Continuation US11414657B2 (en) 2015-06-29 2016-06-29 Modified CRISPR RNA and modified single CRISPR RNA and uses thereof

Publications (1)

Publication Number Publication Date
US20230159919A1 true US20230159919A1 (en) 2023-05-25

Family

ID=57609084

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/737,723 Active US11414657B2 (en) 2015-06-29 2016-06-29 Modified CRISPR RNA and modified single CRISPR RNA and uses thereof
US17/850,456 Pending US20230159919A1 (en) 2015-06-29 2022-06-27 Modified crispr rna and modified single crispr rna and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/737,723 Active US11414657B2 (en) 2015-06-29 2016-06-29 Modified CRISPR RNA and modified single CRISPR RNA and uses thereof

Country Status (6)

Country Link
US (2) US11414657B2 (en)
EP (1) EP3313989A4 (en)
JP (1) JP2018519811A (en)
AU (1) AU2016285724A1 (en)
CA (1) CA2990699A1 (en)
WO (1) WO2017004261A1 (en)

Families Citing this family (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2616051T3 (en) 2008-12-02 2017-06-09 Wave Life Sciences Japan, Inc. Method for the synthesis of modified nucleic acids in the phosphorus atom
US9744183B2 (en) 2009-07-06 2017-08-29 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
JP5868324B2 (en) 2010-09-24 2016-02-24 株式会社Wave Life Sciences Japan Asymmetric auxiliary group
SG10201700554VA (en) 2011-07-19 2017-03-30 Wave Life Sciences Pte Ltd Methods for the synthesis of functionalized nucleic acids
CA2853829C (en) 2011-07-22 2023-09-26 President And Fellows Of Harvard College Evaluation and improvement of nuclease cleavage specificity
AU2013288048A1 (en) 2012-07-13 2015-01-22 Wave Life Sciences Ltd. Asymmetric auxiliary group
CA2878945A1 (en) 2012-07-13 2014-01-16 Wave Life Sciences Pte. Ltd. Chiral control
US20150044192A1 (en) 2013-08-09 2015-02-12 President And Fellows Of Harvard College Methods for identifying a target site of a cas9 nuclease
US9359599B2 (en) 2013-08-22 2016-06-07 President And Fellows Of Harvard College Engineered transcription activator-like effector (TALE) domains and uses thereof
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9228207B2 (en) 2013-09-06 2016-01-05 President And Fellows Of Harvard College Switchable gRNAs comprising aptamers
US9322037B2 (en) 2013-09-06 2016-04-26 President And Fellows Of Harvard College Cas9-FokI fusion proteins and uses thereof
US11053481B2 (en) 2013-12-12 2021-07-06 President And Fellows Of Harvard College Fusions of Cas9 domains and nucleic acid-editing domains
WO2015108047A1 (en) 2014-01-15 2015-07-23 株式会社新日本科学 Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
JPWO2015108048A1 (en) 2014-01-15 2017-03-23 株式会社新日本科学 Chiral nucleic acid adjuvant and antitumor agent having antitumor activity
KR20220106232A (en) 2014-01-16 2022-07-28 웨이브 라이프 사이언시스 리미티드 Chiral design
HUE049634T2 (en) 2014-02-11 2020-09-28 Univ Colorado Regents Crispr enabled multiplexed genome engineering
WO2016022363A2 (en) 2014-07-30 2016-02-11 President And Fellows Of Harvard College Cas9 proteins including ligand-dependent inteins
CN107532168A (en) 2015-02-23 2018-01-02 克里斯珀医疗股份公司 Treat the material and method of hemoglobinopathy
WO2017004279A2 (en) 2015-06-29 2017-01-05 Massachusetts Institute Of Technology Compositions comprising nucleic acids and methods of using the same
JP2018519811A (en) 2015-06-29 2018-07-26 アイオーニス ファーマシューティカルズ, インコーポレーテッドIonis Pharmaceuticals,Inc. Modified CRISPR RNA and modified single CRISPR RNA and uses thereof
US10167457B2 (en) 2015-10-23 2019-01-01 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2017136794A1 (en) 2016-02-03 2017-08-10 Massachusetts Institute Of Technology Structure-guided chemical modification of guide rna and its applications
US10767175B2 (en) 2016-06-08 2020-09-08 Agilent Technologies, Inc. High specificity genome editing using chemically modified guide RNAs
US10017760B2 (en) 2016-06-24 2018-07-10 Inscripta, Inc. Methods for generating barcoded combinatorial libraries
ES2908919T3 (en) 2016-07-05 2022-05-04 California Inst Of Techn Fractional primer hybridization chain reaction
US11845929B2 (en) 2016-07-08 2023-12-19 Ohio State Innovation Foundation Modified nucleic acids, hybrid guide RNAs, and uses thereof
IL308426A (en) 2016-08-03 2024-01-01 Harvard College Adenosine nucleobase editors and uses thereof
CN109804066A (en) 2016-08-09 2019-05-24 哈佛大学的校长及成员们 Programmable CAS9- recombination enzyme fusion proteins and application thereof
WO2018039438A1 (en) 2016-08-24 2018-03-01 President And Fellows Of Harvard College Incorporation of unnatural amino acids into proteins using base editing
EP3522898A4 (en) * 2016-10-06 2020-05-27 Ionis Pharmaceuticals, Inc. Method of conjugating oligomeric compounds
AU2017342543A1 (en) 2016-10-14 2019-05-02 President And Fellows Of Harvard College AAV delivery of nucleobase editors
WO2018119359A1 (en) 2016-12-23 2018-06-28 President And Fellows Of Harvard College Editing of ccr5 receptor gene to protect against hiv infection
US20190382751A1 (en) * 2016-12-28 2019-12-19 Ionis Pharmaceuticals, Inc. Modified crispr rna and uses thereof
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US11542496B2 (en) 2017-03-10 2023-01-03 President And Fellows Of Harvard College Cytosine to guanine base editor
JP7191388B2 (en) 2017-03-23 2022-12-19 プレジデント アンド フェローズ オブ ハーバード カレッジ Nucleobase editors comprising nucleic acid programmable DNA binding proteins
CN108690845B (en) * 2017-04-10 2021-04-27 中国科学院动物研究所 Genome editing system and method
US11560566B2 (en) 2017-05-12 2023-01-24 President And Fellows Of Harvard College Aptazyme-embedded guide RNAs for use with CRISPR-Cas9 in genome editing and transcriptional activation
CN108977442B (en) * 2017-06-05 2023-01-06 广州市锐博生物科技有限公司 System for DNA editing and application thereof
EP3635113A4 (en) 2017-06-05 2021-03-17 Fred Hutchinson Cancer Research Center Genomic safe harbors for genetic therapies in human stem cells and engineered nanoparticles to provide targeted genetic therapies
EP3638789A4 (en) * 2017-06-12 2021-03-10 California Institute of Technology Conditional guide rnas
US10011849B1 (en) 2017-06-23 2018-07-03 Inscripta, Inc. Nucleic acid-guided nucleases
US9982279B1 (en) 2017-06-23 2018-05-29 Inscripta, Inc. Nucleic acid-guided nucleases
US11866726B2 (en) 2017-07-14 2024-01-09 Editas Medicine, Inc. Systems and methods for targeted integration and genome editing and detection thereof using integrated priming sites
EP3658573A1 (en) 2017-07-28 2020-06-03 President and Fellows of Harvard College Methods and compositions for evolving base editors using phage-assisted continuous evolution (pace)
WO2019027728A1 (en) * 2017-07-31 2019-02-07 Sigma-Aldrich Co. Llc Synthetic guide rna for crispr/cas activator systems
US11319532B2 (en) 2017-08-30 2022-05-03 President And Fellows Of Harvard College High efficiency base editors comprising Gam
EP3697906A1 (en) 2017-10-16 2020-08-26 The Broad Institute, Inc. Uses of adenosine base editors
US20190233816A1 (en) * 2018-01-26 2019-08-01 Massachusetts Institute Of Technology Structure-guided chemical modification of guide rna and its applications
MA51788A (en) 2018-02-05 2020-12-16 Vertex Pharma SUBSTANCES AND METHODS FOR TREATING HEMOGLOBINOPATHIES
US11566236B2 (en) 2018-02-05 2023-01-31 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
WO2019213910A1 (en) * 2018-05-10 2019-11-14 Syngenta Participations Ag Methods and compositions for targeted editing of polynucleotides
KR20210045360A (en) 2018-05-16 2021-04-26 신테고 코포레이션 Methods and systems for guide RNA design and use
US20200255861A1 (en) * 2018-12-17 2020-08-13 The Broad Institute, Inc. Crispr cpf1 direct repeat variants
DE112020001342T5 (en) 2019-03-19 2022-01-13 President and Fellows of Harvard College Methods and compositions for editing nucleotide sequences
JP2023525304A (en) 2020-05-08 2023-06-15 ザ ブロード インスティテュート,インコーポレーテッド Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
KR102493512B1 (en) * 2020-10-08 2023-02-01 주식회사 진코어 An engineered crRNA for CRISPR/Cas12a system
EP4284925A1 (en) 2021-01-26 2023-12-06 California Institute of Technology Allosteric conditional guide rnas for cell-selective regulation of crispr/cas
WO2023091466A1 (en) * 2021-11-16 2023-05-25 Integrated Dna Technologies, Inc. Design of two-part guide rnas for crispra applications

Family Cites Families (140)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2699805A (en) 1951-10-23 1955-01-18 Richter Albert Guiding attachment for hand planes
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4583111A (en) 1983-09-09 1986-04-15 Fairchild Semiconductor Corporation Integrated circuit chip wiring arrangement providing reduced circuit inductance and controlled voltage gradients
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
FR2575751B1 (en) 1985-01-08 1987-04-03 Pasteur Institut NOVEL ADENOSINE DERIVATIVE NUCLEOSIDES, THEIR PREPARATION AND THEIR BIOLOGICAL APPLICATIONS
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5506337A (en) 1985-03-15 1996-04-09 Antivirals Inc. Morpholino-subunit combinatorial library and method
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5166315A (en) 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
ATE113059T1 (en) 1987-06-24 1994-11-15 Florey Howard Inst NUCLEOSIDE DERIVATIVES.
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
EP0544824B1 (en) 1990-07-27 1997-06-11 Isis Pharmaceuticals, Inc. Nuclease resistant, pyrimidine modified oligonucleotides that detect and modulate gene expression
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
EP1256589A3 (en) 1991-11-26 2003-09-17 Isis Pharmaceuticals, Inc. Oligomers containing modified pyrimidines
TW393513B (en) 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
EP0673559A1 (en) 1992-12-14 1995-09-27 Honeywell Inc. Motor system with individually controlled redundant windings
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
US5656408A (en) 1996-04-29 1997-08-12 Xerox Corporation Coated carrier particles
JP3756313B2 (en) 1997-03-07 2006-03-15 武 今西 Novel bicyclonucleosides and oligonucleotide analogues
US6770748B2 (en) 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
US20060172925A1 (en) * 1998-10-26 2006-08-03 Board Of Regents, The University Of Texas System Thio-siRNA aptamers
JP2002535995A (en) 1999-02-03 2002-10-29 ザ チルドレンズ メディカル センター コーポレイション Gene repair involving induction of double-stranded DNA breaks at chromosomal target sites
CA2372085C (en) 1999-05-04 2009-10-27 Exiqon A/S L-ribo-lna analogues
US6525191B1 (en) 1999-05-11 2003-02-25 Kanda S. Ramasamy Conformationally constrained L-nucleosides
US20030158403A1 (en) 2001-07-03 2003-08-21 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
US8604183B2 (en) 2002-11-05 2013-12-10 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
WO2004041889A2 (en) 2002-11-05 2004-05-21 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
WO2004106356A1 (en) 2003-05-27 2004-12-09 Syddansk Universitet Functionalized nucleotide derivatives
JP4731324B2 (en) 2003-08-28 2011-07-20 武 今西 N-O bond cross-linked novel artificial nucleic acid
CA2538252C (en) 2003-09-18 2014-02-25 Isis Pharmaceuticals, Inc. 4'-thionucleosides and oligomeric compounds
US7919277B2 (en) 2004-04-28 2011-04-05 Danisco A/S Detection and typing of bacterial strains
DK2314594T3 (en) 2006-01-27 2014-10-27 Isis Pharmaceuticals Inc 6-modified bicyclic nucleic acid analogues
CA2651453C (en) 2006-05-11 2014-10-14 Isis Pharmaceuticals, Inc. 5'-modified bicyclic nucleic acid analogs
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
JP4798093B2 (en) 2006-08-04 2011-10-19 日産自動車株式会社 Fluid reforming apparatus and fluid reforming method using the same
EP2125852B1 (en) 2007-02-15 2016-04-06 Ionis Pharmaceuticals, Inc. 5'-substituted-2'-f modified nucleosides and oligomeric compounds prepared therefrom
EP2170917B1 (en) 2007-05-30 2012-06-27 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
WO2008154401A2 (en) 2007-06-08 2008-12-18 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
CA2692579C (en) 2007-07-05 2016-05-03 Isis Pharmaceuticals, Inc. 6-disubstituted bicyclic nucleic acid analogs
KR101654007B1 (en) 2007-08-15 2016-09-05 아이오니스 파마수티컬즈, 인코포레이티드 Tetrahydropyran nucleic acid analogs
FR2925918A1 (en) 2007-12-28 2009-07-03 Pasteur Institut Typing or subtyping Salmonella bacteria comprises determining the variable sequence composition of a nucleic acid fragment amplified from the CRISPR1 and/or CRISPR2 locus
US8546553B2 (en) 2008-07-25 2013-10-01 University Of Georgia Research Foundation, Inc. Prokaryotic RNAi-like system and methods of use
PL2367938T3 (en) 2008-12-12 2014-11-28 Dupont Nutrition Biosci Aps Genetic cluster of strains of streptococcus thermophilus having unique rheological properties for dairy fermentation
US10913767B2 (en) 2010-04-22 2021-02-09 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
US9070158B1 (en) 2012-02-24 2015-06-30 Amazon Technologies, Inc. Adaptive information regions displaying content associated with an electronic commerce system
US9637739B2 (en) 2012-03-20 2017-05-02 Vilnius University RNA-directed DNA cleavage by the Cas9-crRNA complex
PE20190844A1 (en) 2012-05-25 2019-06-17 Emmanuelle Charpentier MODULATION OF TRANSCRIPTION WITH ADDRESSING RNA TO GENERIC DNA
US9102936B2 (en) 2012-06-11 2015-08-11 Agilent Technologies, Inc. Method of adaptor-dimer subtraction using a CRISPR CAS6 protein
JP2015527889A (en) 2012-07-25 2015-09-24 ザ ブロード インスティテュート, インコーポレイテッド Inducible DNA binding protein and genomic disruption tools and their applications
KR101656237B1 (en) 2012-10-23 2016-09-12 주식회사 툴젠 Composition for cleaving a target DNA comprising a guide RNA specific for the target DNA and Cas protein-encoding nucleic acid or Cas protein, and use thereof
EP2848690B1 (en) 2012-12-12 2020-08-19 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
US20140242664A1 (en) 2012-12-12 2014-08-28 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions for sequence manipulation
ES2883590T3 (en) 2012-12-12 2021-12-09 Broad Inst Inc Supply, modification and optimization of systems, methods and compositions for sequence manipulation and therapeutic applications
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products
US8993233B2 (en) 2012-12-12 2015-03-31 The Broad Institute Inc. Engineering and optimization of systems, methods and compositions for sequence manipulation with functional domains
EP3252160B1 (en) 2012-12-12 2020-10-28 The Broad Institute, Inc. Crispr-cas component systems, methods and compositions for sequence manipulation
ES2701749T3 (en) 2012-12-12 2019-02-25 Broad Inst Inc Methods, models, systems and apparatus to identify target sequences for Cas enzymes or CRISPR-Cas systems for target sequences and transmit results thereof
US20140310830A1 (en) 2012-12-12 2014-10-16 Feng Zhang CRISPR-Cas Nickase Systems, Methods And Compositions For Sequence Manipulation in Eukaryotes
EP2931899A1 (en) 2012-12-12 2015-10-21 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions, methods, knock out libraries and applications thereof
SG10201801969TA (en) 2012-12-12 2018-04-27 Broad Inst Inc Engineering and Optimization of Improved Systems, Methods and Enzyme Compositions for Sequence Manipulation
SG11201504621RA (en) 2012-12-17 2015-07-30 Harvard College Rna-guided human genome engineering
US11332719B2 (en) 2013-03-15 2022-05-17 The Broad Institute, Inc. Recombinant virus and preparations thereof
JP6980380B2 (en) 2013-03-15 2021-12-15 ザ ジェネラル ホスピタル コーポレイション Increased specificity of RNA-induced genome editing with shortened guide RNA (tru-gRNA)
EP3633039A1 (en) 2013-05-01 2020-04-08 Ionis Pharmaceuticals, Inc. Compositions and methods
DK3309248T3 (en) * 2013-05-29 2021-08-02 Cellectis Method for manipulating T cells for immunotherapy using an RNA-guided CAS nuclease system
US9267135B2 (en) 2013-06-04 2016-02-23 President And Fellows Of Harvard College RNA-guided transcriptional regulation
CA2915837A1 (en) 2013-06-17 2014-12-24 The Broad Institute, Inc. Optimized crispr-cas double nickase systems, methods and compositions for sequence manipulation
DK3011029T3 (en) 2013-06-17 2020-03-16 Broad Inst Inc ADMINISTRATION, MODIFICATION AND OPTIMIZATION OF TANDEM GUIDE SYSTEMS, PROCEDURES AND COMPOSITIONS FOR SEQUENCE MANIPULATION
BR112015031608A2 (en) 2013-06-17 2017-08-22 Massachusetts Inst Technology APPLICATION AND USE OF CRISPR-CAS SYSTEMS, VECTORS AND COMPOSITIONS FOR LIVER TARGETING AND THERAPY
EP3725885A1 (en) 2013-06-17 2020-10-21 The Broad Institute, Inc. Functional genomics using crispr-cas systems, compositions methods, screens and applications thereof
EP3011032B1 (en) 2013-06-17 2019-10-16 The Broad Institute, Inc. Delivery, engineering and optimization of systems, methods and compositions for targeting and modeling diseases and disorders of post mitotic cells
EP3011034B1 (en) 2013-06-17 2019-08-07 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for targeting disorders and diseases using viral components
AU2014287009B2 (en) 2013-07-11 2020-10-29 Modernatx, Inc. Compositions comprising synthetic polynucleotides encoding CRISPR related proteins and synthetic sgRNAs and methods of use
US20150044192A1 (en) 2013-08-09 2015-02-12 President And Fellows Of Harvard College Methods for identifying a target site of a cas9 nuclease
BR112016003561B8 (en) 2013-08-22 2022-11-01 Du Pont METHOD FOR PRODUCING A GENETIC MODIFICATION, METHOD FOR INTRODUCING A POLYNUCLEOTIDE OF INTEREST INTO THE GENOME OF A PLANT, METHOD FOR EDITING A SECOND GENE INTO A PLANT GENOME AND METHOD FOR GENERATING A GLYPHOSATE-RESISTANT CORN PLANT
WO2015048577A2 (en) 2013-09-27 2015-04-02 Editas Medicine, Inc. Crispr-related methods and compositions
WO2015051169A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc. Polynucleotide molecules and uses thereof
WO2015051173A2 (en) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc Polynucleotide molecules and uses thereof
SG11201602503TA (en) 2013-10-03 2016-04-28 Moderna Therapeutics Inc Polynucleotides encoding low density lipoprotein receptor
EP3066201B1 (en) 2013-11-07 2018-03-07 Editas Medicine, Inc. Crispr-related methods and compositions with governing grnas
DE102013223172A1 (en) * 2013-11-14 2015-05-21 Aktiebolaget Skf engine assembly
CA2930877A1 (en) 2013-11-18 2015-05-21 Crispr Therapeutics Ag Crispr-cas system materials and methods
US9074199B1 (en) 2013-11-19 2015-07-07 President And Fellows Of Harvard College Mutant Cas9 proteins
US9546384B2 (en) 2013-12-11 2017-01-17 Regeneron Pharmaceuticals, Inc. Methods and compositions for the targeted modification of a mouse genome
KR20160097327A (en) 2013-12-12 2016-08-17 더 브로드 인스티튜트, 인코퍼레이티드 Crispr-cas systems and methods for altering expression of gene products, structural information and inducible modular cas enzymes
WO2015089364A1 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Crystal structure of a crispr-cas system, and uses thereof
BR112016013213A2 (en) 2013-12-12 2017-12-05 Massachusetts Inst Technology administration, use and therapeutic applications of crisper systems and compositions for targeting disorders and diseases using particle delivery components
CN105899658B (en) 2013-12-12 2020-02-18 布罗德研究所有限公司 Delivery, use and therapeutic applications of CRISPR-CAS systems and compositions for HBV and viral diseases and disorders
WO2015089486A2 (en) 2013-12-12 2015-06-18 The Broad Institute Inc. Systems, methods and compositions for sequence manipulation with optimized functional crispr-cas systems
EP3080259B1 (en) 2013-12-12 2023-02-01 The Broad Institute, Inc. Engineering of systems, methods and optimized guide compositions with new architectures for sequence manipulation
CA2932472A1 (en) 2013-12-12 2015-06-18 Massachusetts Institute Of Technology Compositions and methods of use of crispr-cas systems in nucleotide repeat disorders
EP3653229A1 (en) 2013-12-12 2020-05-20 The Broad Institute, Inc. Delivery, use and therapeutic applications of the crispr-cas systems and compositions for genome editing
US11053481B2 (en) 2013-12-12 2021-07-06 President And Fellows Of Harvard College Fusions of Cas9 domains and nucleic acid-editing domains
US11224603B2 (en) 2013-12-13 2022-01-18 Requis Pharmaceuticals, Inc. Antihistamines in combination with a range of substances for improved health
WO2015134812A1 (en) * 2014-03-05 2015-09-11 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating usher syndrome and retinitis pigmentosa
EP3116997B1 (en) * 2014-03-10 2019-05-15 Editas Medicine, Inc. Crispr/cas-related methods and compositions for treating leber's congenital amaurosis 10 (lca10)
US20150376586A1 (en) 2014-06-25 2015-12-31 Caribou Biosciences, Inc. RNA Modification to Engineer Cas9 Activity
US9778707B1 (en) 2014-08-08 2017-10-03 Performance Contracting, Inc. System for field access of data
ES2780904T3 (en) 2014-08-17 2020-08-27 Broad Inst Inc Genomic editing using Cas9 nickases
WO2016049258A2 (en) 2014-09-25 2016-03-31 The Broad Institute Inc. Functional screening with optimized functional crispr-cas systems
EP3215617A2 (en) 2014-11-07 2017-09-13 Editas Medicine, Inc. Methods for improving crispr/cas-mediated genome-editing
AU2015355546B2 (en) 2014-12-03 2021-10-14 Agilent Technologies, Inc. Guide RNA with chemical modifications
WO2016094867A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Protected guide rnas (pgrnas)
WO2016094872A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Dead guides for crispr transcription factors
WO2016094874A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Escorted and functionalized guides for crispr-cas systems
WO2016094880A1 (en) 2014-12-12 2016-06-16 The Broad Institute Inc. Delivery, use and therapeutic applications of crispr systems and compositions for genome editing as to hematopoietic stem cells (hscs)
JP2017538427A (en) 2014-12-18 2017-12-28 インテグレイテッド ディーエヌエイ テクノロジーズ インコーポレイテッド CRISPR composition and method of use
WO2016100974A1 (en) 2014-12-19 2016-06-23 The Broad Institute Inc. Unbiased identification of double-strand breaks and genomic rearrangement by genome-wide insert capture sequencing
WO2016106236A1 (en) 2014-12-23 2016-06-30 The Broad Institute Inc. Rna-targeting system
WO2016106244A1 (en) 2014-12-24 2016-06-30 The Broad Institute Inc. Crispr having or associated with destabilization domains
HRP20231022T1 (en) 2015-01-28 2023-12-08 Caribou Biosciences, Inc. Crispr hybrid dna/rna polynucleotides and methods of use
BR112017020750A2 (en) 2015-03-27 2018-06-26 Harvard College modified t-cells and methods of producing and using them
GB201506509D0 (en) 2015-04-16 2015-06-03 Univ Wageningen Nuclease-mediated genome editing
WO2016186745A1 (en) 2015-05-15 2016-11-24 Ge Healthcare Dharmacon, Inc. Synthetic single guide rna for cas9-mediated gene editing
EP3666895A1 (en) 2015-06-18 2020-06-17 The Broad Institute, Inc. Novel crispr enzymes and systems
IL284808B (en) 2015-06-18 2022-07-01 Broad Inst Inc Crispr enzyme mutations reducing off-target effects
WO2016205759A1 (en) 2015-06-18 2016-12-22 The Broad Institute Inc. Engineering and optimization of systems, methods, enzymes and guide scaffolds of cas9 orthologs and variants for sequence manipulation
US9790490B2 (en) 2015-06-18 2017-10-17 The Broad Institute Inc. CRISPR enzymes and systems
JP2018519811A (en) 2015-06-29 2018-07-26 アイオーニス ファーマシューティカルズ, インコーポレーテッドIonis Pharmaceuticals,Inc. Modified CRISPR RNA and modified single CRISPR RNA and uses thereof
CN108350454B (en) 2015-09-21 2022-05-10 阿克丘勒斯治疗公司 Allele-selective gene editing and uses thereof
WO2017053729A1 (en) 2015-09-25 2017-03-30 The Board Of Trustees Of The Leland Stanford Junior University Nuclease-mediated genome editing of primary cells and enrichment thereof
US20170145394A1 (en) 2015-11-23 2017-05-25 The Regents Of The University Of California Tracking and manipulating cellular rna via nuclear delivery of crispr/cas9
KR20180133840A (en) 2015-12-04 2018-12-17 노파르티스 아게 Compositions and methods for immuno-oncology
WO2017136794A1 (en) 2016-02-03 2017-08-10 Massachusetts Institute Of Technology Structure-guided chemical modification of guide rna and its applications
US20200172935A1 (en) 2016-04-16 2020-06-04 Ohio State Innovation Foundation Modified cpf1 mrna, modified guide rna, and uses thereof
WO2017190664A1 (en) 2016-05-05 2017-11-09 苏州吉玛基因股份有限公司 Use of chemosynthetic crrna and modified crrna in crispr/cpf1 gene editing systems
CN105907785B (en) 2016-05-05 2020-02-07 苏州吉玛基因股份有限公司 Application of chemically synthesized crRNA in CRISPR/Cpf1 system in gene editing
WO2018098383A1 (en) 2016-11-22 2018-05-31 Integrated Dna Technologies, Inc. Crispr/cpf1 systems and methods

Also Published As

Publication number Publication date
JP2018519811A (en) 2018-07-26
US20180179521A1 (en) 2018-06-28
EP3313989A1 (en) 2018-05-02
CA2990699A1 (en) 2017-01-05
WO2017004261A1 (en) 2017-01-05
US11414657B2 (en) 2022-08-16
AU2016285724A1 (en) 2017-11-16
EP3313989A4 (en) 2018-12-05

Similar Documents

Publication Publication Date Title
US20230159919A1 (en) Modified crispr rna and modified single crispr rna and uses thereof
US11629348B2 (en) Linkage modified oligomeric compounds and uses thereof
EP3011028B1 (en) Compositions and methods for modulation of target nucleic acids
US10370659B2 (en) Compounds and methods for increasing antisense activity
EP3353306A1 (en) Conjugated antisense compounds and their use
WO2017079291A1 (en) Compounds and methods for modulating c90rf72
US9156873B2 (en) Modified 5′ diphosphate nucleosides and oligomeric compounds prepared therefrom
WO2010048585A2 (en) Oligomeric compounds and methods
EP4206213A1 (en) Compounds and methods for modulation of smn2
EP3724206B1 (en) Conjugated antisense compounds and their use
US20220064636A1 (en) Modified oligomeric compounds and uses thereof
US20190382751A1 (en) Modified crispr rna and uses thereof
US11180756B2 (en) Morpholino modified oligomeric compounds
WO2014089146A1 (en) Compositions and methods for in vivo delivery of antisense compounds
WO2021236599A1 (en) Conjugated oligonucleotides and uses thereof
US20210254059A1 (en) Linkage modified oligomeric compounds
US20230105319A1 (en) PRODRUG INCORPORATED sgRNA SYNTHESIS
WO2023235817A1 (en) Linkage modified oligomeric compounds and uses thereof
WO2023023550A1 (en) Linkage modified oligomeric compounds and uses thereof