US20140303235A1 - Linkage modified gapped oligomeric compounds and uses thereof - Google Patents

Linkage modified gapped oligomeric compounds and uses thereof Download PDF

Info

Publication number
US20140303235A1
US20140303235A1 US14/237,967 US201214237967A US2014303235A1 US 20140303235 A1 US20140303235 A1 US 20140303235A1 US 201214237967 A US201214237967 A US 201214237967A US 2014303235 A1 US2014303235 A1 US 2014303235A1
Authority
US
United States
Prior art keywords
certain embodiments
nucleoside
modified
oligomeric compound
nucleosides
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.)
Abandoned
Application number
US14/237,967
Other languages
English (en)
Inventor
Michael Oestergaard
Thazha P. Prakash
Punit P. Seth
Eric E. Swayze
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
Isis 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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46717938&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20140303235(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Isis Pharmaceuticals Inc filed Critical Isis Pharmaceuticals Inc
Priority to US14/237,967 priority Critical patent/US20140303235A1/en
Publication of US20140303235A1 publication Critical patent/US20140303235A1/en
Assigned to IONIS PHARMACEUTICALS, INC. reassignment IONIS PHARMACEUTICALS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: ISIS PHARMACEUTICALS, INC.
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • 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
    • C12N2310/312Phosphonates
    • 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
    • C12N2310/312Phosphonates
    • C12N2310/3125Methylphosphonates
    • 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
    • C12N2310/314Phosphoramidates
    • 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
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/316Phosphonothioates
    • 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
    • C12N2310/341Gapmers, i.e. of the type ===---===
    • 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
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/34Allele or polymorphism specific uses

Definitions

  • the present invention pertains generally to chemically-modified oligonucleotides for use in research, diagnostics, and/or therapeutics.
  • Antisense compounds have been used to modulate target nucleic acids. Antisense compounds comprising a variety of chemical modifications and motifs have been reported. In certain instances, such compounds are useful as research tools, diagnostic reagents, and as therapeutic agents. In certain instances antisense compounds have been shown to modulate protein expression by binding to a target messenger RNA (mRNA) encoding the protein. In certain instances, such binding of an antisense compound to its target mRNA results in cleavage of the mRNA. Antisense compounds that modulate processing of a pre-mRNA have also been reported. Such antisense compounds alter splicing, interfere with polyadenlyation or prevent formation of the 5′-cap of a pre-mRNA.
  • mRNA target messenger RNA
  • DNA or RNA containing oligonucleotides comprising alkylphosphonate internucleoside linkage backbone have been disclosed (see U.S. Pat. Nos. 5,264,423 and 5,286,717).
  • nitrogen containing backbones include oxime (Sanghvi et al., In Nucleosides and Nucleotides as Antitumor and Antiviral Agents; C. K. Chu and D. C.
  • sulfur-containing backbone modifications such as sulfonamide (McElroy et al., Bioorg. Med. Chem. Lett., 1994, 4, 1071-1076), sulfamoyl (Dewynter et al., Acad. Sci., 1992, 315, 1675-1682), sulfonate (Huang et al., Synlett, 1993, 83-84), sulfide (Wang et al., Chin. Chem.
  • sulfonamide McElroy et al., Bioorg. Med. Chem. Lett., 1994, 4, 1071-1076
  • sulfamoyl Dewynter et al., Acad. Sci., 1992, 315, 1675-1682
  • sulfonate Huang et al., Synlett, 1993, 83-84
  • sulfide Wang et al., Chin. Chem.
  • Oligomeric compounds have been prepared using Click chemistry wherein alkynyl phosphonate intemucleoside linkages on an oligomeric compound attached to a solid support are converted into the 1,2,3-triazolylphosphonate intemucleoside linkages and then cleaved from the solid support (Krishna et al., J. Am. Chem. Soc ., DOI: 101021/ja3026714, published online May 21, 2012).
  • the oligomeric compounds provided herein hybridize to a portion of a target RNA resulting in loss of normal function of the target RNA.
  • the oligomeric compounds disclosed herein provide improved selectivity for a target RNA.
  • the oligomeric compounds provide improved potency for a target RNA.
  • the oligomeric compounds provided herein provide an improvement in the toxicity profile.
  • the oligomeric compounds provided herein provide an improvement in the proinflammatory profile.
  • the oligomeric compounds provide improved potency and selectivity for a target RNA.
  • the oligomeric compounds provide improved potency, selectivity and an improvement in the proinflammatory profile.
  • gapped oligomeric compounds comprising a contiguous sequence of linked monomer subunits having a gap region located between a 5′-region and a 3′-region wherein the 5′ and 3′-regions each, independently, have from 2 to 8 contiguous modified nucleosides wherein essentially each modified nucleoside in the 5′ and 3′-regions are RNA-like and the gap region has from 6 to 14 contiguous monomer subunits selected from ⁇ -D-2′-deoxyribonucleosides and modified nucleosides that are DNA-like and wherein at least one of the internucleoside linking groups in the gap region or linking the gap region and the 5′-region or the 3′-region has Formula I:
  • X is O or S
  • Q is 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, substituted C 2 -C 6 alkynyl, C( ⁇ O)OH or CH 2 C( ⁇ O)OH;
  • each substituted group comprises one or more optionally protected substituent groups independently selected from halogen, OJ 1 , SJ 1 and OC( ⁇ O)J 1 ;
  • each J 1 is, independently, H or C 1 -C 6 alkyl.
  • gapped oligomeric compounds consisting of a contiguous sequence of linked monomer subunits having a gap region located between a 5′-region and a 3′-region wherein the 5′ and 3′-regions each, independently, have from 2 to 8 contiguous modified nucleosides wherein essentially each modified nucleoside in the 5′ and 3′-regions 5′ and 3′-regions are RNA-like and the gap region has from 6 to 14 contiguous monomer subunits selected from ⁇ -D-2′-deoxyribonucleosides and modified nucleosides that are DNA-like and wherein at least one of the internucleoside linking groups in the gap region or linking the gap region and the 5′-region or the 3′-region has Formula I:
  • X is O or S
  • Q is 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, substituted C 2 -C 6 alkynyl, CO( ⁇ O)H or CH 2 C( ⁇ O)OH;
  • each substituted group comprises one or more optionally protected substituent groups independently selected from halogen, OJ 1 , SJ 1 and OC( ⁇ O)J 1 ;
  • each J 1 is, independently, H or C 1 -C 6 alkyl.
  • gapped oligomeric compounds consisting of a contiguous sequence of linked monomer subunits having a gap region located between a 5′-region and a 3′-region wherein the 5′ and 3′-regions each, independently, have from 2 to 8 contiguous modified nucleosides wherein each modified nucleoside in the 5′ and 3′-regions are RNA-like and the gap region has from 6 to 14 contiguous monomer subunits selected from ⁇ -D-2′-deoxyribonucleosides and modified nucleosides that are DNA-like and wherein at least one of the internucleoside linking groups in the gap region or linking the gap region and the 5′-region or the 3′-region has Formula I:
  • X is O or S
  • Q is 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, substituted C 2 -C 6 alkynyl, CO( ⁇ O)H or CH 2 C( ⁇ O)OH;
  • each substituted group comprises one or more optionally protected substituent groups independently selected from halogen, OJ 1 , SJ 1 and OC( ⁇ O)J 1 ;
  • each J 1 is, independently, H or C 1 -C 6 alkyl.
  • gapped oligomeric compounds consisting of a contiguous sequence of linked monomer subunits having a gap region located between a 5′-region and a 3′-region wherein the 5′ and 3′-regions each, independently, have from 2 to 8 contiguous modified nucleosides wherein each modified nucleoside in the 5′ and 3′-regions are RNA-like and the gap region has from 6 to 14 contiguous monomer subunits selected from ⁇ -D-2′-deoxyribonucleosides and wherein at least one of the internucleoside linking groups in the gap region or linking the gap region and the 5′-region or the 3′-region has Formula I:
  • X is O or S
  • Q is 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, substituted C 2 -C 6 alkynyl, C( ⁇ O)OH or CH 2 C( ⁇ O)OH;
  • each substituted group comprises one or more optionally protected substituent groups independently selected from halogen, OJ 1 , SJ 1 and OC( ⁇ O)J 1 ;
  • each J 1 is, independently, H or C 1 -C 6 alkyl.
  • gapped oligomeric compounds are provided having only one internucleoside linking group of Formula I in the gap region. In certain embodiments, gapped oligomeric compounds are provided having only two internucleoside linking groups of Formula I in the gap region. In certain embodiments, gapped oligomeric compounds are provided having only three internucleoside linking groups of Formula I in the gap region. In certain embodiments, the intemucleoside linking groups having Formula I are contiguous. In certain embodiments, gapped oligomeric compounds are provide having at least two intemucleoside linking groups of Formula I in the gap region that are separated by at least one phosphorothioate or phosphodiester internucleoside linking group.
  • gapped oligomeric compounds are provided wherein the internucleoside linking group linking the 5′-region and the gap region has Formula I. In certain embodiments, gapped oligomeric compounds are provided wherein the intemucleoside linking group linking the 5′-region and the gap region and the adjacent internucleoside linkage in the gap region has Formula I. In certain embodiments, the internucleoside linking group linking the 3′-region and the gap region has Formula I. In certain embodiments, gapped oligomeric compounds are provided wherein the internucleoside linking group linking the 3′-region and the gap region and the adjacent internucleoside linkage in the gap region has Formula I.
  • the internucleoside linking group linking the 5′-region and the gap region has Formula I and the internucleoside linking group linking the 3′-region and the gap region has Formula I. In certain embodiments, only one internucleoside linking group of Formula I is located in the gap region.
  • each internucleoside linking group in the 5′ and 3′-regions and each internucleoside linking group in the gap region other than internucleoside linking groups having Formula I is a phosphodiester or a phosphorothioate internucleoside linking group.
  • each internucleoside linking group in the 5′ and 3′-regions and each internucleoside linking group in the gap region other than internucleoside linking groups having Formula I is a phosphorothioate internucleoside linking group.
  • each internucleoside linking group in the 5′ and 3′-regions and each internucleoside linking group in the gap region other than internucleoside linking groups having Formula I is a phosphodiester internucleoside linking group.
  • each monomer subunit comprises a heterocyclic base independently selected from an optionally protected purine, substituted purine, pyrimidine or substituted pyrimidine.
  • each monomer subunit comprises a heterocyclic base independently selected from uracil, thymine, cytosine, 4-N-benzoylcytosine, 5-methylcytosine, 4-N-benzoyl-5-methylcytosine, adenine, 6-N-benzoyladenine, guanine or 2-N-isobutyrylguanine.
  • gapped oligomeric compounds wherein each Q is, independently, selected from C 1 -C 6 alkyl, substituted C 1 -C 6 alkyl, C 2 -C 6 alkenyl, CO( ⁇ O)H and CH 2 C( ⁇ O)OH.
  • each Q is, independently, selected from CH 3 , C( ⁇ O)OH, CH 2 C( ⁇ O)OH, (CH 2 ) 2 OCH 3 , CH ⁇ CH 2 , CH 2 CH ⁇ CH 2 and C ⁇ CH.
  • each Q is CH 2 C( ⁇ O)OH.
  • each Q is CH 3 .
  • each Q is C ⁇ CH.
  • gapped oligomeric compounds are provided wherein each X is O. In certain embodiments, each X is S.
  • gapped oligomeric compounds are provided wherein the chirality of each internucleoside linking group having Formula I is R p . In certain embodiments, the chirality of each internucleoside linking group having Formula I is S p .
  • each modified nucleoside in the 5′ and 3′-regions provides enhanced hybridization affinity for an RNA target as compared to an unmodified nucleoside.
  • each modified nucleoside in the 5′ and 3′-regions comprises a modified sugar moiety.
  • each modified nucleoside in the 5′ and 3′-regions is, independently, a bicyclic nucleoside comprising a bicyclic furanosyl sugar moiety or a modified nucleoside comprising a furanosyl sugar moiety having at least one substituent group.
  • gapped oligomeric compounds wherein the 5′ and 3′-regions comprise one or more 2′-modified nucleosides that each have a 2′-substituent group independently selected from halogen, OCH 3 , OCH 2 F, OCHF 2 , OCF 3 , OCH 2 CH 3 , O(CH 2 ) 2 F, OCH 2 CHF 2 , OCH 2 CF 3 , OCH 2 —CH ⁇ CH 2 , O(CH 2 ) 2 —OCH 3 , O(CH 2 ) 2 —SCH 3 , O(CH 2 ) 2 —OCF 3 , O(CH 2 ) 3 —N(R 3 )(R 4 ), O(CH 2 ) 2 —ON(R 3 )(R 4 ), O(CH 2 ) 2 —O(CH 2 ) 2 —N(R 3 )(R 4 ), OCH 2 C( ⁇ O)—N(R 4 )(R 4 ), OCH 2 C( ⁇ O
  • each 2′-substituent group is independently selected from F, OCH 3 , OCF 3 , OCH 2 CH 3 , OCH 2 CF 3 , OCH 2 —CH ⁇ CH 2 , O(CH 2 ) 2 —OCH 3 , O(CH 2 ) 2 —O(CH 2 ) 2 —N(CH 3 ) 2 , OCH 2 C( ⁇ O)—N(H)CH 3 , OCH 2 C( ⁇ O)—N(H)—(CH 2 ) 2 —N(CH 3 ) 2 and OCH 2 —N(H)—C( ⁇ NH)NH 2 .
  • each 2′-substituent group is independently selected from F, OCH 3 , O(CH 2 ) 2 —OCH 3 and OCH 2 C( ⁇ O)—N(H)CH 3 . In certain embodiments, each 2′-substituent group is O(CH 2 ) 2 —OCH 3 .
  • gapped oligomeric compounds wherein the 5′ and 3′-regions comprise one or more bicyclic nucleosides that each have a bridging group between the 4′ and 2′ carbon atoms of the furanosyl ring independently selected from 4′-(CH 2 )—O-2′, 4′-(CH 2 )—S-2′, 4′-(CH 2 ) 2 —O-2′, 4′-CH(CH 3 )—O-2′, 4′-CH(CH 2 OCH 3 )—O-2′, 4′-C(CH 3 ) 2 —O-2′, 4′-CH 2 —N(OCH 3 )-2′, 4′-CH 2 —O—N(CH 3 )-2′, 4′-CH 2 —NCH 3 —O-2′, 4′-CH 2 —C(H)(CH 3 )-2′ and 4′-CH 2 —C( ⁇ CH 2 )-2′.
  • each of the bridging groups is independently selected from 4′-(CH 2 )—O-2′, 4′-(CH 2 ) 2 —O-2′, 4′-CH(CH 3 )—O-2′, 4′-CH 2 —NCH 3 —O-2′, 4′-CH 2 —C(H)(CH 3 )-2′ and 4′-CH 2 —C( ⁇ CH 2 )-2′.
  • each bridging group is 4′-CH[(S)—(CH 3 )]—O-2′.
  • gapped oligomeric compounds are provided wherein the sugar moieties of each modified nucleoside in the 5′ and 3′-regions are the same. In certain embodiments, gapped oligomeric compounds are provided comprising at least two different types of modified nucleosides in the 5′ and 3′-regions wherein the different types of modified nucleosides have at least different modified sugar moieties. In certain embodiments, the different types of modified nucleosides include bicyclic nucleosides comprising bicyclic furanosyl sugar moieties and modified nucleosides comprising furanosyl sugar moieties having at least one substituent group.
  • the different types of modified nucleosides include 4′-CH[(S)—(CH 3 )]—O-2′ bicyclic nucleosides and 2′-O(CH 2 ) 2 —OCH 3 substituted nucleosides.
  • the 5′ and 3′-regions include only 4′-CH[(S)—(CH 3 )]—O-2′ bicyclic nucleosides and 2′-O(CH 2 ) 2 —OCH 3 substituted nucleosides.
  • gapped oligomeric compounds wherein one or more modified nucleosides in the 5′ and 3′-regions comprise a sugar surrogate.
  • gapped oligomeric compounds are provided wherein each monomer subunit in the gap region is ⁇ -D-2′-deoxyribonucleoside.
  • at least one monomer subunit in the gap region is a modified nucleoside that is DNA-like.
  • each modified nucleoside that is DNA-like is a 2′-(ara)-F modified nucleoside.
  • gapped oligomeric compounds are provided wherein the 5′ and 3′-regions each, independently, have from 2 to 8 monomer subunits. In certain embodiments, the 5′ and 3′-regions each, independently, have from 3 to 6 monomer subunits. In certain embodiments, the gap region has from 6 to 14 monomer subunits. In certain embodiments, the gap region has from 8 to 10 monomer subunits. In certain embodiments, the 5′ and 3′-regions each, independently, have from 3 to 6 monomer subunits and the gap region has from 8 to 14 monomer subunits.
  • the 5′ and 3′-regions each, independently, have from 3 to 6 monomer subunits and the gap region has from 6 to 10 monomer subunits. In certain embodiments, the 5′ and 3′-regions each, independently, have from 3 to 6 monomer subunits and the gap region has from 6 to 8 monomer subunits. In certain embodiments, the 5′ and 3′-regions each, independently, have from 4 to 5 monomer subunits and the gap region has from 7 to 8 monomer subunits.
  • gapped oligomeric compounds wherein at least one modified nucleoside in the 5′ and 3′-regions is other than a 2′-OCH 3 substituted nucleoside or a 2′-O—CH 2 -4′ bridged bicyclic nucleoside.
  • gapped oligomeric compounds are provided that include one 5′-terminal group. In certain embodiments, gapped oligomeric compounds are provided that include one 3′-terminal group. In certain embodiments, gapped oligomeric compounds are provided that include at least one 5′ or 3′-terminal group.
  • the gapped oligomeric compounds provided herein are other than the gapped oligomeric compounds listed below:
  • each internucleoside linkage is a phosphorothioate.
  • Each “ x T” is a 2-thio-thymidine modified nucleoside.
  • a subscript “p” indicates a methyl phosphonate internucleoside linkage (—(P( ⁇ O)(CH 3 ))—).
  • Nucleosides not followed by a subscript are ⁇ -D-2′-deoxyribonucleosides.
  • Nucleosides followed by a subscript “e” are 2′-O-methoxyethyl (MOE) modified nucleosides.
  • Nucleosides followed by a subscript “k” are 6′-(S)—CH 3 (cEt) bicyclic modified nucleosides.
  • Each “ m C” is a 5-methyl cytosine modified nucleoside.
  • the gapped oligomeric compounds provided herein are other than gapped oligomeric compounds complementary to at least a region of a nucleic acid that is a Huntingtin gene transcript. In certain embodiments, the gapped oligomeric compounds provided herein are other than gapped oligomeric compounds complementary to at least a region of a nucleic acid comprising a single-nucleotide polymorphism. In certain embodiments, the gapped oligomeric compounds provided herein are other than gapped oligomeric compounds complementary to at least a region of a nucleic acid comprising a single-nucleotide polymorphism-containing-target nucleic acid of a Huntingtin gene transcript.
  • the gapped oligomeric compounds provided herein are other than gapped oligomeric compounds complementary to at least a region of a nucleic acid comprising a single-nucleotide polymorphism-containing-target nucleic acid of a gene transcript other than Huntingtin.
  • methods of inhibiting gene expression comprising contacting one or more cells, a tissue or an animal with an oligomeric compound as provided herein wherein said oligomeric compound is complementary to a target RNA.
  • the cells are in a human.
  • the target RNA is human mRNA.
  • the target RNA is cleaved thereby inhibiting its function.
  • in vitro methods of inhibiting gene expression comprising contacting one or more cells or a tissue with an oligomeric compound as provided herein.
  • oligomeric compound as provided herein are used in an in vivo method of inhibiting gene expression said method comprising contacting one or more cells, a tissue or an animal with an oligomeric compound as provided herein.
  • oligomeric compounds as provided herein are used in medical therapy.
  • Such gapped oligomeric compounds comprise a contiguous sequence of linked monomer subunits having a gap region located between a 5′-region and a 3′-region wherein the 5′ and 3′-regions each, independently, have from 2 to 8 contiguous modified nucleosides wherein essentially each modified nucleoside in the 5′ and 3′-regions are RNA-like and the gap region has from 6 to 14 contiguous monomer subunits selected from ⁇ -D-2′-deoxyribonucleosides and modified nucleosides that are DNA-like and wherein at least one of the internucleoside linking groups in the gap region or between the gap region and the 5′-region or the 3′-region has Formula I:
  • X is O or S
  • Q is 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, substituted C 2 -C 6 alkynyl, C( ⁇ O)OH or CH 2 CO( ⁇ O)H;
  • each substituted group comprises one or more optionally protected substituent groups independently selected from halogen, OJ 1 , SJ 1 and OC( ⁇ O)J 1 ;
  • each J 1 is, independently, H or C 1 -C 6 alkyl.
  • the gapped oligomeric compounds provided herein have been shown to have improved properties.
  • the activity of an otherwise unmodified gapped oligomeric compound against a target nucleic acid is enhanced by incorporation of at least one internucleoside linking group having Formula I in the gap region.
  • at least one internucleoside linking group having Formula I is located at the gap junction on the 5′ side wherein the internucleoside linkage separates the gap region from the wing region.
  • at least one internucleoside linking group having Formula I is located at the gap junction on the 3′ side.
  • such properties include selectivity, potency, improved toxicity profile and or an improved proinflammatory profile.
  • a gapped oligomeric compound of interest is identified and then a series of identical oligomeric compounds are prepared with a single internucleoside linking group having Formula I walked across the gap region. If there are 8 monomer subunits in the gap then there will be 8 oligomeric compounds prepared having the internucleoside linking group having Formula I located at a different position in each of the oligomeric compounds which are subsequently assayed in one or more assays as illustrated herein to determine the lead from the series.
  • additional internucleoside linking groups having Formula I are incorporated into the gap region of the lead oligomeric compound and assayed in one or more assays as illustrated herein.
  • the lead compound is further functionalized with one or more terminal groups such as for example a conjugate group.
  • a gapped oligomeric compound of interest is identified and then a series of identical oligomeric compounds are prepared with blocks of at least two internucleoside linking group having Formula I walked across the gap region.
  • gapped oligomeric compounds are provided having a reduced proinflammatory response when compared to unmodified gapped oligomeric compounds.
  • a gapped oligomeric compound having alternating methyl thiophosphonate (—P(CH 3 )( ⁇ S)—) internucleoside linkages in the gap reduced the proinflammatory response compared to the an identical oligomeric compound without the modified internucleoside linkages.
  • gapped oligomeric compounds are provided having enhanced potency (IC 50 ) and selectivity when compared to unmodified gapped oligomeric compounds.
  • IC 50 enhanced potency
  • selectivity when compared to unmodified gapped oligomeric compounds.
  • gapped oligomeric compounds are provided having enhanced selectivity with comparable potency (IC 50 ) when compared to unmodified gapped oligomeric compounds.
  • IC 50 potency
  • a gapped oligomeric compound having one or two methyl phosphonate (—P(CH 3 )( ⁇ O)—) or one phosphonoacetate (—P(CH 2 CO 2 ⁇ )( ⁇ O)—) internucleoside linkages in the gap typically showed enhanced selectivity with comparable or slightly lower potency.
  • gapped oligomeric compounds are provided having enhanced selectivity with comparable potency (IC 50 ) when compared to an unmodified gapped oligomeric compound.
  • IC 50 comparable potency
  • a gapped oligomeric compound having one or two methyl phosphonate (—P(CH 3 )( ⁇ O)—) internucleoside linkages in the gap typically showed enhanced selectivity with comparable or slightly lower potency.
  • gapped oligomeric compounds are provided having improved hepatotoxicity profiles when compared to unmodified gapped oligomeric compounds.
  • a gapped oligomeric compound having two methyl phosphonate (—P(CH 3 )( ⁇ O)—) internucleoside linkages at the 5′-end of the gap and linking the gap to the 5′-wing lowered the ALT as compared to the unmodified oligomeric compounds.
  • the activity and organ weights are similar for the SRB-1 assay. The activity is reduced for the modified oligomeric compound in the PTEN assay.
  • the liver is slightly elevated for the modified gapped oligomeric compound whereas for the unmodified oligomeric compound the liver and the spleen weights are both elevated.
  • chemical modification means a chemical difference in a compound when compared to a naturally occurring counterpart.
  • Chemical modifications of oligonucleotides include nucleoside modifications (including sugar moiety modifications and nucleobase modifications) and internucleoside linkage modifications. In reference to an oligonucleotide, chemical modification does not include differences only in nucleobase sequence.
  • furanosyl means a structure comprising a 5-membered ring comprising four carbon atoms and one oxygen atom.
  • naturally occurring sugar moiety means a ribofuranosyl as found in naturally occurring RNA or a 2′-deoxyribofuranosyl as found in naturally occurring DNA.
  • sugar moiety means a naturally occurring sugar moiety or a modified sugar moiety of a nucleoside.
  • modified sugar moiety means a substituted sugar moiety or a sugar surrogate.
  • substituted sugar moiety means a furanosyl that is not a naturally occurring sugar moiety.
  • Substituted sugar moieties include, but are not limited to furanosyls comprising substituents at the 2′-position, the 3′-position, the 5′-position and/or the 4′-position.
  • Certain substituted sugar moieties are bicyclic sugar moieties.
  • 2′-substituted sugar moiety means a furanosyl comprising a substituent at the 2′-position other than H or OH. Unless otherwise indicated, a 2′-substituted sugar moiety is not a bicyclic sugar moiety (i.e., the 2′-substituent of a 2′-substituted sugar moiety does not form a bridge to another atom of the furanosyl ring.
  • MOE means —OCH 2 CH 2 OCH 3 .
  • 2′-F nucleoside refers to a nucleoside comprising a sugar comprising fluorine at the 2′ position. Unless otherwise indicated, the fluorine in a 2′-F nucleoside is in the ribo position (replacing the OH of a natural ribose).
  • 2′-(ara)-F refers to a 2′-F substituted nucleoside, wherein the fluoro group is in the arabino position.
  • sucrose surrogate means a structure that does not comprise a furanosyl ring and that is capable of replacing the naturally occurring sugar moiety of a nucleoside, such that the resulting nucleoside sub-units or monomer subunits are capable of linking together and/or linking to other nucleosides or other monomer subunits to form an oligomeric compound which is capable of hybridizing to a complementary oligomeric compound such as a nucleic acid target.
  • Such structures include rings comprising a different number of atoms than furanosyl (e.g., 4, 6, or 7-membered rings); replacement of the oxygen atom of a furanosyl with a non-oxygen atom (e.g., carbon, sulfur, or nitrogen, wherein replacement of the oxygen atom with sulfur in furanose is generally considered a modified nucleoside as opposed to a sugar surrogate but can be considered both); or both a change in the number of atoms and a replacement of the oxygen.
  • Such structures may also comprise substitutions corresponding to those described for substituted sugar moieties (e.g., 6-membered carbocyclic bicyclic sugar surrogates optionally comprising additional substituents).
  • Sugar surrogates also include more complex sugar replacements (e.g., the non-ring systems of peptide nucleic acid).
  • Sugar surrogates include without limitation morpholinos, cyclohexenyls and cyclohexitols.
  • bicyclic sugar moiety means a modified sugar moiety comprising a 4 to 7 membered ring (including but not limited to a furanosyl) comprising a bridge connecting two atoms of the 4 to 7 membered ring to form a second ring, resulting in a bicyclic structure.
  • the 4 to 7 membered ring is a sugar ring.
  • the 4 to 7 membered ring is a furanosyl.
  • the bridge connects the 2′-carbon and the 4′-carbon of the furanosyl.
  • nucleoside means a compound comprising a nucleobase moiety and a sugar moiety. Nucleosides include, but are not limited to, naturally occurring nucleosides (as found in DNA and RNA) and modified nucleosides. Nucleosides may be linked to a phosphate moiety.
  • nucleotide means a nucleoside further comprising a phosphate linking group.
  • linked nucleosides may or may not be linked by phosphate linkages and thus includes, but is not limited to “linked nucleotides.”
  • linked nucleosides are nucleosides that are connected in a continuous sequence (i.e. no additional nucleosides are present between those that are linked).
  • nucleobase generally refers to the nucleobase of a nucleoside or modified nucleoside.
  • heterocyclic base moiety is broader than the term nucleobase in that it includes any heterocyclic base that can be attached to a sugar to prepare a nucleoside or modified nucleoside.
  • heterocyclic base moieties include but are not limited to naturally occurring nucleobases (adenine, guanine, thymine, cytosine and uracil) and protected forms of unmodified nucleobases (4-N-benzoylcytosine, 6-N-benzoyladenine and 2-N-isobutyrylguanine) as well as modified (5-methyl cytosine) or non-naturally occurring heterocyclic base moieties and synthetic mimetics thereof (such as for example phenoxazines).
  • nucleobases adenine, guanine, thymine, cytosine and uracil
  • protected forms of unmodified nucleobases (4-N-benzoylcytosine, 6-N-benzoyladenine and 2-N-isobutyrylguanine)
  • modified (5-methyl cytosine) or non-naturally occurring heterocyclic base moieties and synthetic mimetics thereof such as for example phenoxazines
  • modified nucleoside refers to a nucleoside comprising a modified heterocyclic base and or a sugar moiety other than ribose and 2′-deoxyribose.
  • a modified nucleoside comprises a modified heterocyclic base moiety.
  • a modified nucleoside comprises a sugar moiety other than ribose and 2′-deoxyribose.
  • a modified nucleoside comprises a modified heterocyclic base moiety and a sugar moiety other than ribose and 2′-deoxyribose.
  • modified nucleoside is intended to include all manner of modified nucleosides that can be incorporated into an oligomeric compound using standard oligomer synthesis protocols. Modified nucleosides include a basic nucleosides but in general a heterocyclic base moiety is included for hybridization to a complementary nucleic acid target.
  • modified nucleosides include a furanose or modified furanose sugar group such as a 4′-S analog (4′-S-modified nucleoside and 4′-S-ribonucleoside refer to replacement of the furanose oxygen atom with S).
  • modified nucleosides include without limitation, substituted nucleosides (such as 2′, 5′, and/or 4′ substituted nucleosides) 4′-S-modified nucleosides, (such as 4′-S-ribonucleosides, 4′-S-2′-deoxyribonucleosides and 4′-S-2′-substituted ribonucleosides), bicyclic modified nucleosides (such as 2′-O—CH(CH 3 )-4′, 2′-O—CH 2 -4′ or 2′-O—(CH 2 ) 2 -4′ bridged furanose analogs) and base modified nucleosides.
  • substituted nucleosides such as 2′, 5′, and/or 4′ substituted nucleosides
  • 4′-S-modified nucleosides such as 4′-S-ribonucleosides, 4′-S-2′-deoxyribonucleosides and 4′-S-2
  • the sugar can be modified with more than one of these modifications listed such as for example a bicyclic modified nucleoside further including a 5′-substitution or a 5′ or 4′ substituted nucleoside further including a 2′ substituent.
  • modified nucleoside also includes combinations of these modifications such as base and sugar modified nucleosides. These modifications are meant to be illustrative and not exhaustive as other modifications are known in the art and are also envisioned as possible modifications for the modified nucleosides described herein.
  • modified nucleosides comprise a sugar surrogate wherein the furanose ring has been replaced with a mono or polycyclic ring system or a non-cyclic sugar surrogate such as that used in peptide nucleic acids.
  • sugar moieties for such modified nucleosides includes without limitation morpholino, hexitol, cyclohexenyl, 2.2.2 and 3.2.1 cyclohexose and open non-cyclic groups.
  • modified nucleosides comprise a non-naturally occurring sugar moiety and a modified heterocyclic base moiety.
  • modified nucleosides include without limitation modified nucleosides wherein the heterocyclic base moiety is replaced with a phenoxazine moiety (for example the 9-(2-aminoethoxy)-1,3-diazaphenoxazine-2-one group, also referred to as a G-clamp which forms four hydrogen bonds when hybridized with a guanosine base) and further replacement of the sugar moiety with a sugar surrogate group such as for example a morpholino, a cyclohexenyl or a bicyclo[3.1.0]hexyl.
  • a sugar surrogate group such as for example a morpholino, a cyclohexenyl or a bicyclo[3.1.0]hexyl.
  • bicyclic nucleoside or “BNA” means a nucleoside comprising a bicyclic sugar moiety.
  • constrained ethyl nucleoside or “cEt” means a nucleoside comprising a bicyclic sugar moiety comprising a 4′-CH(CH 3 )—O-2′ bridge.
  • the cEt comprises a comprising a 4′-CH((S)—CH 3 )—O-2′ bridge.
  • the cEt comprises a comprising a 4′-CH((R)—CH 3 )—O-2′ bridge.
  • locked nucleic acid nucleoside or “LNA” means a nucleoside comprising a bicyclic sugar moiety comprising a 4′-CH 2 —O-2′ bridge.
  • 2′-substituted nucleoside means a ribofuranosyl nucleoside comprising a substituent at the 2′-position other than H or OH. Unless otherwise indicated, a 2′-substituted nucleoside is not a bicyclic nucleoside.
  • 2′-deoxynucleoside means a nucleoside comprising 2′-H(H) furanosyl sugar moiety, as found in naturally occurring deoxyribonucleosides (DNA).
  • a 2′-deoxynucleoside may comprise a modified nucleobase or may comprise an RNA nucleobase (e.g., uracil).
  • RNA-like nucleoside means a modified nucleoside other than a ⁇ -D-ribose nucleoside that provides an A-form (northern) duplex when incorporated into an oligomeric compound and duplexed with a complementary RNA.
  • RNA-like nucleosides are used as replacements for RNA nucleosides in oligomeric compounds to enhance one or more properties such as, for example, nuclease resistance and or hybridization affinity.
  • RNA-like nucleosides include, but are not limited to modified furanosyl nucleosides that adopt a 3′-endo conformational geometry when put into an oligomeric compound.
  • RNA-like nucleosides also include RNA surrogates such as F-HNA.
  • RNA-like nucleosides include but are not limited to modified nucleosides comprising a 2′-substituent group selected from F, O(CH 2 ) 2 OCH 3 (MOE) and OCH 3 .
  • RNA-like nucleosides also include but are not limited to modified nucleosides comprising bicyclic furanosyl sugar moiety comprising a 4′-CH 2 —O-2′, 4′-(CH 2 ) 2 —O-2′, 4′-C(H)[(R)—CH 3 ]-O-2′ or 4′-C(H)[(S)—CH 3 ]-O-2′ bridging group.
  • DNA-like nucleoside means a modified nucleoside other than ⁇ -D-2′-doxyribose nucleoside that provides a B-form (southern) duplex when incorporated into an oligomeric compound and duplexed with a complementary DNA.
  • DNA-like nucleosides provide an intermediate duplex when incorporated into an oligomeric compound and duplexed with a complementary RNA that is between A-form and B-form.
  • DNA-like nucleosides are used as replacements for DNA nucleosides in oligomeric compounds to enhance one or more properties.
  • DNA-like nucleosides include, but are not limited to modified nucleosides that adopt a 2′-endo conformational geometry when put into an oligomeric compound.
  • oligonucleotide means a compound comprising a plurality of linked nucleosides.
  • an oligonucleotide comprises one or more unmodified ribonucleosides (RNA) and/or unmodified deoxyribonucleosides (DNA) and/or one or more modified nucleosides.
  • oligonucleoside means an oligonucleotide in which none of the internucleoside linkages contains a phosphorus atom.
  • oligonucleotides include oligonucleosides.
  • modified oligonucleotide means an oligonucleotide comprising at least one modified nucleoside and/or at least one modified internucleoside linkage.
  • nucleoside linkage means a covalent linkage between adjacent nucleosides in an oligonucleotide.
  • naturally occurring internucleoside linkage means a 3′ to 5′ phosphodiester linkage.
  • modified internucleoside linkage means any internucleoside linkage other than a naturally occurring internucleoside linkage.
  • a monomer subunit is meant to include all manner of monomers that are amenable to oligomer synthesis.
  • a monomer subunit includes at least a sugar moiety having at least two reactive sites that can form linkages to further monomer subunits.
  • all monomer subunits include a heterocyclic base moiety that is hybridizable to a complementary site on a nucleic acid target.
  • Reactive sites on monomer subunits located on the termini of an oligomeric compound can be protected or unprotected (generally OH) or can form an attachment to a terminal group (conjugate or other group).
  • Monomer subunits include, without limitation, nucleosides and modified nucleosides.
  • monomer subunits include nucleosides such as ⁇ -D-ribonucleosides and ⁇ -D-2′-deoxyribnucleosides and modified nucleosides including but not limited to substituted nucleosides (such as 2′, 5′ and bis substituted nucleosides), 4′-S-modified nucleosides (such as 4′-S-ribonucleosides, 4′-S-2′-deoxyribonucleosides and 4′-S-2′-substituted ribonucleosides), bicyclic modified nucleosides (such as bicyclic nucleosides wherein the sugar moiety has a 2′-O—CHR a -4′ bridging group, wherein R a is H, alkyl or substituted alkyl), other modified nucleosides and nucleosides having sugar surrogates.
  • substituted nucleosides such as 2′, 5′ and bis substituted nucleosides
  • conjugate means an atom or group of atoms bound to an oligonucleotide or oligomeric compound.
  • conjugate groups modify one or more properties of the compound to which they are attached, including, but not limited to pharmacodynamic, pharmacokinetic, binding, absorption, cellular distribution, cellular uptake, charge and/or clearance properties.
  • conjugate linking group means any atom or group of atoms used to attach a conjugate to an oligonucleotide or oligomeric compound.
  • antisense compound means a compound comprising or consisting of an oligonucleotide or oligomeric compound wherein at least a portion of which is complementary to a target nucleic acid to which it is capable of hybridizing, resulting in at least one antisense activity.
  • antisense activity means any detectable and/or measurable change attributable to the hybridization of an antisense compound to its target nucleic acid.
  • detecting or “measuring” means that a test or assay for detecting or measuring is performed. Such detection and/or measuring may result in a value of zero. Thus, if a test for detection or measuring results in a finding of no activity (activity of zero), the step of detecting or Measuring the activity has nevertheless been performed.
  • detecttable and/or measurable activity means a statistically significant activity that is not zero.
  • essentially unchanged means little or no change in a particular parameter, particularly relative to another parameter which changes much more.
  • a parameter is essentially unchanged when it changes less than 5%.
  • a parameter is essentially unchanged if it changes less than two-fold while another parameter changes at least ten-fold.
  • an antisense activity is a change in the amount of a target nucleic acid.
  • the amount of a non-target nucleic acid is essentially unchanged if it changes much less than the target nucleic acid does, but the change need not be zero.
  • expression means the process by which a gene ultimately results in a protein.
  • Expression includes, but is not limited to, transcription, post-transcriptional modification (e.g., splicing, polyadenlyation, addition of 5′-cap), and translation.
  • target nucleic acid means a nucleic acid molecule to which an antisense compound hybridizes.
  • mRNA means an RNA molecule that encodes a protein.
  • pre-mRNA means an RNA transcript that has not been fully processed into mRNA. Pre-RNA includes one or more intron.
  • object RNA means an RNA molecule other than a target RNA, the amount, activity, splicing, and/or function of which is modulated, either directly or indirectly, by a target nucleic acid.
  • a target nucleic acid modulates splicing of an object RNA.
  • an antisense compound modulates the amount or activity of the target nucleic acid, resulting in a change in the splicing of an object RNA and ultimately resulting in a change in the activity or function of the object RNA.
  • microRNA means a naturally occurring, small, non-coding RNA that represses gene expression of at least one mRNA.
  • a microRNA represses gene expression by binding to a target site within a 3′ untranslated region of an mRNA.
  • a microRNA has a nucleobase sequence as set forth in miRBase, a database of published microRNA sequences found at http://microrna.sanger.ac.uk/sequences/.
  • a microRNA has a nucleobase sequence as set forth in miRBase version 12.0 released September 2008, which is herein incorporated by reference in its entirety.
  • microRNA mimic means an oligomeric compound having a sequence that is at least partially identical to that of a microRNA.
  • a microRNA mimic comprises the microRNA seed region of a microRNA.
  • a microRNA mimic modulates translation of more than one target nucleic acids.
  • a microRNA mimic is double-stranded.
  • “differentiating nucleobase” means a nucleobase that differs between two nucleic acids.
  • a target region of a target nucleic acid differs by 1-4 nucleobases from a non-target nucleic acid. Each of those differences is referred to as a differentiating nucleobase.
  • a differentiating nucleobase is a single-nucleotide polymorphism.
  • target-selective nucleoside means a nucleoside of an antisense compound that corresponds to a differentiating nucleobase of a target nucleic acid.
  • allelic pair means one of a pair of copies of a gene existing at a particular locus or marker on a specific chromosome, or one member of a pair of nucleobases existing at a particular locus or marker on a specific chromosome, or one member of a pair of nucleobase sequences existing at a particular locus or marker on a specific chromosome.
  • each allelic pair will normally occupy corresponding positions (loci) on a pair of homologous chromosomes, one inherited from the mother and one inherited from the father.
  • the organism or cell is said to be “homozygous” for that allele; if they differ, the organism or cell is said to be “heterozygous” for that allele.
  • Wild-type allele refers to the genotype typically not associated with disease or dysfunction of the gene product.
  • Melt allele refers to the genotype associated with disease or dysfunction of the gene product.
  • allelic variant means a particular identity of an allele, where more than one identity occurs.
  • an allelic variant may refer to either the mutant allele or the wild-type allele.
  • single nucleotide polymorphism or “SNP” means a single nucleotide variation between the genomes of individuals of the same species.
  • a SNP may be a single nucleotide deletion or insertion.
  • SNPs occur relatively frequently in genomes and thus contribute to genetic diversity. The location of a SNP is generally flanked by highly conserved sequences. An individual may be homozygous or heterozygous for an allele at each SNP site.
  • single nucleotide polymorphism site or “SNP site” refers to the nucleotides surrounding a SNP contained in a target nucleic acid to which an antisense compound is targeted.
  • targeting means the association of an antisense compound to a particular target nucleic acid molecule or a particular region of a target nucleic acid molecule.
  • An antisense compound targets a target nucleic acid if it is sufficiently complementary to the target nucleic acid to allow hybridization under physiological conditions.
  • nucleobase complementarity or “complementarity” when in reference to nucleobases means a nucleobase that is capable of base pairing with another nucleobase.
  • adenine (A) is complementary to thymine (T).
  • adenine (A) is complementary to uracil (U).
  • complementary nucleobase means a nucleobase of an antisense compound that is capable of base pairing with a nucleobase of its target nucleic acid.
  • nucleobases at a certain position of an antisense compound are capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid
  • the position of hydrogen bonding between the oligonucleotide and the target nucleic acid is considered to be complementary at that nucleobase pair.
  • Nucleobases comprising certain modifications may maintain the ability to pair with a counterpart nucleobase and thus, are still capable of nucleobase complementarity.
  • non-complementary in reference to nucleobases means a pair of nucleobases that do not form hydrogen bonds with one another.
  • complementary in reference to oligomeric compounds (e.g., linked nucleosides, oligonucleotides, or nucleic acids) means the capacity of such oligomeric compounds or regions thereof to hybridize to another oligomeric compound or region thereof through nucleobase complementarity under stringent conditions.
  • Complementary oligomeric compounds need not have nucleobase complementarity at each nucleoside. Rather, some mismatches are tolerated.
  • complementary oligomeric compounds or regions are complementary at 70% of the nucleobases (70% complementary).
  • complementary oligomeric compounds or regions are 80% complementary.
  • complementary oligomeric compounds or regions are 90% complementary.
  • complementary oligomeric compounds or regions are 95% complementary.
  • complementary oligomeric compounds or regions are 100% complementary.
  • mismatch means a nucleobase of a first oligomeric compound that is not capable of pairing with a nucleobase at a corresponding position of a second oligomeric compound, when the first and second oligomeric compound are aligned.
  • Either or both of the first and second oligomeric compounds may be oligonucleotides.
  • hybridization means the pairing of complementary oligomeric compounds (e.g., an antisense compound and its target nucleic acid). While not limited to a particular mechanism, the most common mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleobases.
  • “specifically hybridizes” means the ability of an oligomeric compound to hybridize to one nucleic acid site with greater affinity than it hybridizes to another nucleic acid site.
  • an antisense oligonucleotide specifically hybridizes to more than one target site.
  • oligonucleotide or portion thereof means that each nucleobase of the oligonucleotide or portion thereof is capable of pairing with a nucleobase of a complementary nucleic acid or contiguous portion thereof.
  • a fully complementary region comprises no mismatches or unhybridized nucleobases in either strand.
  • percent complementarity means the percentage of nucleobases of an oligomeric compound that are complementary to an equal-length portion of a target nucleic acid. Percent complementarity is calculated by dividing the number of nucleobases of the oligomeric compound that are complementary to nucleobases at corresponding positions in the target nucleic acid by the total length of the oligomeric compound.
  • percent identity means the number of nucleobases in a first nucleic acid that are the same type (independent of chemical modification) as nucleobases at corresponding positions in a second nucleic acid, divided by the total number of nucleobases in the first nucleic acid.
  • modulation means a change of amount or quality of a molecule, function, or activity when compared to the amount or quality of a molecule, function, or activity prior to modulation.
  • modulation includes the change, either an increase (stimulation or induction) or a decrease (inhibition or reduction) in gene expression.
  • modulation of expression can include a change in splice site selection of pre-mRNA processing, resulting in a change in the absolute or relative amount of a particular splice-variant compared to the amount in the absence of modulation.
  • modification motif means a pattern of chemical modifications in an oligomeric compound or a region thereof. Motifs may be defined by modifications at certain nucleosides and/or at certain linking groups of an oligomeric compound.
  • nucleoside motif means a pattern of nucleoside modifications in an oligomeric compound or a region thereof.
  • the linkages of such an oligomeric compound may be modified or unmodified.
  • motifs herein describing only nucleosides are intended to be nucleoside motifs. Thus, in such instances, the linkages are not limited.
  • sugar motif means a pattern of sugar modifications in an oligomeric compound or a region thereof.
  • linkage motif means a pattern of linkage modifications in an oligomeric compound or region thereof.
  • the nucleosides of such an oligomeric compound may be modified or unmodified.
  • motifs herein describing only linkages are intended to be linkage motifs. Thus, in such instances, the nucleosides are not limited.
  • nucleobase modification motif means a pattern of modifications to nucleobases along an oligonucleotide. Unless otherwise indicated, a nucleobase modification motif is independent of the nucleobase sequence.
  • sequence motif means a pattern of nucleobases arranged along an oligonucleotide or portion thereof. Unless otherwise indicated, a sequence motif is independent of chemical modifications and thus may have any combination of chemical modifications, including no chemical modifications.
  • nucleoside having a modification of a first type may be an unmodified nucleoside.
  • “differently modified” mean chemical modifications or chemical substituents that are different from one another, including absence of modifications.
  • a MOE nucleoside and an unmodified DNA nucleoside are “differently modified,” even though the DNA nucleoside is unmodified.
  • DNA and RNA are “differently modified,” even though both are naturally-occurring unmodified nucleosides. Nucleosides that are the same but for comprising different nucleobases are not differently modified.
  • nucleoside comprising a 2′-OMe modified sugar and an unmodified adenine nucleobase and a nucleoside comprising a 2′-OMe modified sugar and an unmodified thymine nucleobase are not differently modified.
  • the same type of modifications refers to modifications that are the same as one another, including absence of modifications.
  • two unmodified DNA nucleoside have “the same type of modification,” even though the DNA nucleoside is unmodified.
  • Such nucleosides having the same type modification may comprise different nucleobases.
  • pharmaceutically acceptable carrier or diluent means any substance suitable for use in administering to an animal.
  • a pharmaceutically acceptable carrier or diluent is sterile saline.
  • such sterile saline is pharmaceutical grade saline.
  • substituted nucleoside and “substituent group,” means an atom or group that replaces the atom or group of a named parent compound.
  • a substituent of a modified nucleoside is any atom or group that differs from the atom or group found in a naturally occurring nucleoside (e.g., a modified 2′-substituent is any atom or group at the 2′-position of a nucleoside other than H or OH).
  • Substituent groups can be protected or unprotected.
  • compounds of the present invention have substituents at one or at more than one position of the parent compound. Substituents may also be further substituted with other substituent groups and may be attached directly or via a linking group such as an alkyl or hydrocarbyl group to a parent compound.
  • substituted in reference to a chemical functional group means an atom or group of atoms differs from the atom or a group of atoms normally present in the named functional group.
  • a substituent replaces a hydrogen atom of the functional group (e.g., in certain embodiments, the substituent of a substituted methyl group is an atom or group other than hydrogen which replaces one of the hydrogen atoms of an unsubstituted methyl group).
  • groups amenable for use as substituents include without limitation, halogen, hydroxyl, alkyl, alkenyl, alkynyl, acyl (—C(O)R aa ), carboxyl (—C(O)O—R aa ), aliphatic groups, alicyclic groups, alkoxy, substituted oxy (—O—R aa ), aryl, aralkyl, heterocyclic radical, heteroaryl, heteroarylalkyl, amino (—N(R bb )(R cc )), imino( ⁇ NR bb ), amido (—C(O)N(R bb )(R cc ) or —N(R bb )C(O)R aa ), azido (—N 3 ), nitro (—NO 2 ), cyano (—CN), carbamido (—OC(O)N(R bb )(R cc ) or
  • each R aa , R bb and R cc is, independently, H, an optionally linked chemical functional group or a further substituent group with a preferred list including without limitation, alkyl, alkenyl, alkynyl, aliphatic, alkoxy, acyl, aryl, aralkyl, heteroaryl, alicyclic, heterocyclic and heteroarylalkyl. Selected substituents within the compounds described herein are present to a recursive degree.
  • alkyl means a saturated straight or branched hydrocarbon radical containing up to twenty four carbon atoms.
  • alkyl groups include without limitation, methyl, ethyl, propyl, butyl, isopropyl, n-hexyl, octyl, decyl, dodecyl and the like.
  • Alkyl groups typically include from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms (C 1 -C 12 alkyl) with from 1 to about 6 carbon atoms being more preferred.
  • alkenyl means a straight or branched hydrocarbon chain radical containing up to twenty four carbon atoms and having at least one carbon-carbon double bond.
  • alkenyl groups include without limitation, ethenyl, propenyl, butenyl, dienes such as 1,3-butadiene and the like.
  • Alkenyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred.
  • Alkenyl groups as used herein may optionally include one or more further substituent groups.
  • alkynyl means a straight or branched hydrocarbon radical containing up to twenty four carbon atoms and having at least one carbon-carbon triple bond.
  • alkynyl groups include, without limitation, ethynyl, 1-propynyl, 1-butynyl, and the like.
  • Alkynyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred.
  • Alkynyl groups as used herein may optionally include one or more further substituent groups.
  • acyl means a radical formed by removal of a hydroxyl group from an organic acid and has the general Formula —C(O)—X where X is typically aliphatic, alicyclic or aromatic. Examples include aliphatic carbonyls, aromatic carbonyls, aliphatic sulfonyls, aromatic sulfinyls, aliphatic sulfinyls, aromatic phosphates, aliphatic phosphates and the like. Acyl groups as used herein may optionally include further substituent groups.
  • alicyclic means a cyclic ring system wherein the ring is aliphatic.
  • the ring system can comprise one or more rings wherein at least one ring is aliphatic.
  • Preferred alicyclics include rings having from about 5 to about 9 carbon atoms in the ring.
  • Alicyclic as used herein may optionally include further substituent groups.
  • aliphatic means a straight or branched hydrocarbon radical containing up to twenty four carbon atoms wherein the saturation between any two carbon atoms is a single, double or triple bond.
  • An aliphatic group preferably contains from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms with from 1 to about 6 carbon atoms being more preferred.
  • the straight or branched chain of an aliphatic group may be interrupted with one or more heteroatoms that include nitrogen, oxygen, sulfur and phosphorus.
  • Such aliphatic groups interrupted by heteroatoms include without limitation, polyalkoxys, such as polyalkylene glycols, polyamines, and polyimines. Aliphatic groups as used herein may optionally include further substituent groups.
  • alkoxy means a radical formed between an alkyl group and an oxygen atom wherein the oxygen atom is used to attach the alkoxy group to a parent molecule.
  • alkoxy groups include without limitation, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, neopentoxy, n-hexoxy and the like.
  • Alkoxy groups as used herein may optionally include further substituent groups.
  • aminoalkyl means an amino substituted C 1 -C 12 alkyl radical.
  • the alkyl portion of the radical forms a covalent bond with a parent molecule.
  • the amino group can be located at any position and the aminoalkyl group can be substituted with a further substituent group at the alkyl and/or amino portions.
  • aralkyl and arylalkyl mean an aromatic group that is covalently linked to a C 1 -C 12 alkyl radical.
  • the alkyl radical portion of the resulting aralkyl (or arylalkyl) group forms a covalent bond with a parent molecule. Examples include without limitation, benzyl, phenethyl and the like.
  • Aralkyl groups as used herein may optionally include further substituent groups attached to the alkyl, the aryl or both groups that form the radical group.
  • aryl and “aromatic” mean a mono- or polycyclic carbocyclic ring system radicals having one or more aromatic rings.
  • aryl groups include without limitation, phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like.
  • Preferred aryl ring systems have from about 5 to about 20 carbon atoms in one or more rings.
  • Aryl groups as used herein may optionally include further substituent groups.
  • halo and “halogen,” mean an atom selected from fluorine, chlorine, bromine and iodine.
  • heteroaryl and “heteroaromatic,” mean a radical comprising a mono- or poly-cyclic aromatic ring, ring system or fused ring system wherein at least one of the rings is aromatic and includes one or more heteroatoms. Heteroaryl is also meant to include fused ring systems including systems where one or more of the fused rings contain no heteroatoms. Heteroaryl groups typically include one ring atom selected from sulfur, nitrogen or oxygen.
  • heteroaryl groups include without limitation, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl and the like.
  • Heteroaryl radicals can be attached to a parent molecule directly or through a linking moiety such as an aliphatic group or hetero atom.
  • Heteroaryl groups as used herein may optionally include further substituent groups.
  • oligomeric compound refers to a contiguous sequence of linked monomer subunits. Each linked monomer subunit normally includes a heterocyclic base moiety but monomer subunits also includes those without a heterocyclic base moiety such as a basic monomer subunits. At least some and generally most if not essentially all of the heterocyclic bases in an oligomeric compound are capable of hybridizing to a nucleic acid molecule, normally a preselected RNA target.
  • the term “oligomeric compound” therefore includes oligonucleotides, oligonucleotide analogs and oligonucleosides. It also includes polymers having one or a plurality of nucleosides having sugar surrogate groups.
  • oligomeric compounds comprise a plurality of monomer subunits independently selected from naturally occurring nucleosides, non-naturally occurring nucleosides, modified nucleosides and nucleosides having sugar surrogate groups.
  • oligomeric compounds are single stranded.
  • oligomeric compounds are double stranded comprising a double-stranded duplex.
  • oligomeric compounds comprise one or more conjugate groups and/or terminal groups.
  • the oligomeric compounds as provided herein can be modified by covalent attachment of one or more terminal groups to the 5′ or 3′-terminal groups.
  • a terminal group can also be attached at any other position at one of the terminal ends of the oligomeric compound.
  • the terms “5′-terminal group”, “3′-terminal group”, “terminal group” and combinations thereof are meant to include useful groups known to the art skilled that can be placed on one or both of the terminal ends, including but not limited to the 5′ and 3′-ends of an oligomeric compound respectively, for various purposes such as enabling the tracking of the oligomeric compound (a fluorescent label or other reporter group), improving the pharmacokinetics or pharmacodynamics of the oligomeric compound (such as for example: uptake and/or delivery) or enhancing one or more other desirable properties of the oligomeric compound (a group for improving nuclease stability or binding affinity).
  • 5′ and 3′-terminal groups include without limitation, modified or unmodified nucleosides; two or more linked nucleosides that are independently, modified or unmodified; conjugate groups; capping groups; phosphate moieties; and protecting groups.
  • the present invention provides oligomeric compounds.
  • such oligomeric compounds comprise oligonucleotides optionally comprising one or more conjugate and/or terminal groups.
  • an oligomeric compound consists of an oligonucleotide.
  • oligonucleotides comprise one or more chemical modifications. Such chemical modifications include modifications of one or more nucleoside (including modifications to the sugar moiety and/or the nucleobase) and/or modifications to one or more internucleoside linkage.
  • modified nucleosides comprise a modified sugar moeity, a modified nucleobase, or both a modified sugar moiety and a modified nucleobase.
  • compounds of the invention comprise one or more modified nucleosides comprising a modified sugar moiety.
  • modified nucleosides comprising a modified sugar moiety.
  • Such compounds 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 an oligonucleotide comprising only nucleosides comprising naturally occurring sugar moieties.
  • modified sugar moieties are substituted 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 substituted sugar moieties comprising one or more non-bridging sugar substituents, including but not limited to substituents at the 2′ and/or 5′ positions.
  • sugar substituents suitable for the 2′-position include, but are not limited to: 2′-F, 2′-OCH 3 (“OMe” or “O-methyl”), and 2′-O(CH 2 ) 2 OCH 3 (“MOE”).
  • sugar substituents at the 2′ position are selected from allyl, amino, azido, thio, O-allyl, O—C 1 -C 10 alkyl, O—C 1 -C 10 substituted alkyl; OCF 3 , O(CH 2 ) 2 SCH 3 , O(CH 2 ) 2 —O—N(Rm)(Rn), and O—CH 2 —C( ⁇ O)—N(Rm)(Rn), where each Rm and Rn is, independently, H or substituted or unsubstituted C 1 -C 10 alkyl.
  • sugar substituents at the 5′-position include, but are not limited to: 5′-methyl (R or S); 5′-vinyl, and 5′-methoxy.
  • substituted 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 5′,2′-bis substituted sugar moieties and nucleosides).
  • Nucleosides comprising 2′-substituted sugar moieties are referred to as 2′-substituted nucleosides.
  • a 2′-substituted nucleoside comprises a 2′-substituent group selected from halo, allyl, amino, azido, SH, CN, OCN, CF 3 , OCF 3 , O, S, or N(R m )-alkyl; 0, S, or N(R m )-alkenyl; O, S or 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 —O—N(R m )(R n ) or O—CH 2 —C( ⁇ O)—N(R m
  • These 2′-substituent groups can be further substituted with one or more substituent groups independently selected from hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro (NO 2 ), thiol, thioalkoxy (S-alkyl), halogen, alkyl, aryl, alkenyl and alkynyl.
  • a 2′-substituted nucleoside comprises a 2′-substituent group selected from F, NH 2 , N 3 , OCF 3 , O—CH 3 , O(CH 2 ) 3 NH 2 , CH 2 —CH ⁇ CH 2 , O—CH 2 —CH ⁇ CH 2 , OCH 2 CH 2 OCH 3 , O(CH 2 ) 2 SCH 3 , O—(CH 2 ) 2 —O—N(R m )(R n ), O(CH 2 ) 2 O(CH 2 ) 2 N(CH 3 ) 2 , and N-substituted acetamide (O—CH 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 comprises a sugar moiety comprising a 2′-substituent group selected from F, OCF 3 , O—CH 3 , OCH 2 CH 2 OCH 3 , O(CH 2 ) 2 SCH 3 , O—(CH 2 ) 2 —O—N(CH 3 ) 2 , —O(CH 2 ) 2 O(CH 2 ) 2 N(CH 3 ) 2 , and O—CH 2 —C( ⁇ O)—N(H)CH 3 .
  • a 2′-substituted nucleoside comprises a sugar moiety comprising a 2′-substituent group selected from F, OCF 3 , O—CH 3 , OCH 2 CH 2 OCH 3 , O(CH 2 ) 2 SCH 3 , O—(CH 2 ) 2 —O—N(CH 3 ) 2 , —O(CH 2 ) 2 O(CH 2 ) 2 N(CH 3 ) 2
  • a 2′-substituted nucleoside comprises a sugar moiety comprising a 2′-substituent group selected from F, O—CH 3 , and OCH 2 CH 2 OCH 3 .
  • 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′ sugar substituents include, but are not limited to: —[C(R a )(R b )] n —, —[C(R a R b )] n —O—, —C(R a R b )—N(R)—O— or, —C(R a R b )—O—N(R)—; 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′ (cEt) and 4′-CH(CH 2 OCH 3 )—O-2′, and analogs thereof (see, e.g., U.S.
  • such 4′ to 2′ bridges independently comprise from 1 to 4 linked groups (generally forming a 4 to 6 membered ring with the parent sugar moiety) independently selected from —[C(R a )(R b )] n —, —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 nucleosides include, but are not limited to, (A) ⁇ -L-Methyleneoxy (4′-CH 2 —O-2′) BNA, (B) ⁇ -D-Methyleneoxy (4′-CH 2 —O-2′) BNA (also referred to as locked nucleic acid or LNA), (C) Ethyleneoxy (4′-(CH 2 ) 2 —O-2′) BNA, (D) Aminooxy (4′-CH 2 —O—N(R)-2′) BNA, (E) Oxyamino (4′-CH 2 —N(R)—O-2′) BNA, (F) Methyl(methyleneoxy) (4′-CH(CH 3 )—O-2′) BNA (also referred to as constrained ethyl or cEt), (G) methylene-thio (4′-CH 2 —S
  • Bx is a nucleobase moiety and R is, independently, H, a protecting group, or C 1 -C 12 alkyl.
  • bicyclic sugar moieties and nucleosides incorporating such bicyclic sugar moieties are further defined by isomeric configuration.
  • a nucleoside comprising a 4′-2′ methylene-oxy bridge may be in the ⁇ -L configuration or in the ⁇ -D configuration.
  • ⁇ -L-methyleneoxy (4′-CH 2 —O-2′) bicyclic nucleosides have been incorporated into antisense oligonucleotides that showed antisense activity (Frieden et al., Nucleic Acids Research, 2003, 21, 6365-6372).
  • substituted 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.
  • modified sugar moieties are sugar surrogates.
  • the oxygen atom of the naturally occurring sugar is substituted, e.g., with a sulfer, carbon or nitrogen atom.
  • such modified sugar moiety also comprises bridging and/or non-bridging substituents as described above.
  • certain sugar surogates comprise a 4′-sulfer atom and a substitution at the 2′-position (see, e.g., published U.S. Patent Application US2005/0130923, published on Jun. 16, 2005) and/or the 5′ position.
  • carbocyclic bicyclic nucleosides having a 4′-2′ bridge have been described (see, e.g., Freier et al., Nucleic Acids Research, 1997, 25(22), 4429-4443 and Albaek et al., J. Org. Chem., 2006, 71, 7731-7740).
  • sugar surrogates comprise rings having other than 5-atoms.
  • a sugar surrogate comprises a six-membered tetrahydropyran.
  • 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), and those compounds having Formula VII:
  • Bx is a nucleobase moiety
  • T 3 and T 4 are each, independently, an internucleoside linking group linking the tetrahydropyran nucleoside analog to the antisense compound or one of T 3 and T 4 is an internucleoside linking group linking the tetrahydropyran nucleoside analog to the antisense compound 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.
  • the modified THP nucleosides of Formula VII 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, THP nucleosides of Formula VII are provided wherein one of R 1 and R 2 is F. In certain embodiments, R 1 is fluoro and R 2 is H, R 1 is methoxy and R 2 is H, and R 1 is methoxyethoxy and R 2 is H.
  • the present invention provides oligonucleotides comprising modified nucleosides.
  • modified nucleotides may include modified sugars, modified nucleobases, and/or modified linkages. The specific modifications are selected such that the resulting oligonucleotides possess desirable characteristics.
  • oligonucleotides comprise one or more RNA-like nucleosides. In certain embodiments, oligonucleotides comprise one or more DNA-like nucleosides.
  • the oligomeric compounds provided herein include RNA-like nucleosides that have been modified to influence the sugar conformation to have predominantly 3′-endo conformational geometry.
  • such modified nucleosides include synthetic modifications of the heterocyclic base moiety, the sugar moiety or both to induce a 3′-endo sugar conformation.
  • RNA-like nucleosides are selected from RNA surrogates such as including, but not limited to, F-HNA or cyclohexenyl nucleic acid. RNA-like nucleosides are used to replace and mimic RNA nucleosides in an oligomeric compound so that particular properties of the oligomeric compound can be enhanced.
  • RNA-like nucleosides are used in the 5′ and 3′-regions (wings) of gapped oligomeric compounds to improve stability in the presence of nucleases and also to increase the affinity for nucleic a nucleic acid target.
  • Other properties that can also be enhanced by using RNA-like nucleosides include but aren't limited to modulation of pharmacokinetic properties through modification of protein binding, protein off-rate, absorption and clearance as well as chemical stability and specificity of the oligomeric compound (affinity and specificity for enzymes as well as for complementary sequences); and increasing efficacy of RNA cleavage.
  • RNA-like nucleosides include modified nucleosides comprising one or more 2′, 3′, 4′ and 5′ substituent groups, bicyclic nucleosides and RNA-surrogates.
  • RNA-like nucleosides include, but are not limited to modified nucleosides comprising 2′-ribo-substituent groups selected from: F, OCH 3 , O—C 2 -C 4 alkyl, O—CH 2 CH ⁇ CH 2 , O—(CH 2 ) 2 —O—CH 3 (MOE), O—(CH 2 ) 3 —NH 2 , O—(CH 2 ) 2 —O—N(R 1 ) 2 , O—CH 2 C(O)—N(R 1 ) 2 , O—(CH 2 ) 2 —O—(CH 2 ) 2 —N(R 1 ) 2 , O—(CH 2 ) 3 —NHR 1 and O—CH 2 —N(H)—
  • RNA-like nucleosides also include but are not limited to modified nucleosides having a bicyclic furanosyl sugar moiety (bicyclic nucleosides) comprising a bridging group between the 4′ and 2′-carbon atoms.
  • bicyclic nucleosides bicyclic furanosyl sugar moiety
  • Such bicyclic nucleosides include, but are not limited to bridging groups consisting of from 1 to 3 linked biradical groups selected from O, S, NR a , C(R b )(R c ), C ⁇ O, C(R b ) ⁇ C(R c ) and C[ ⁇ C(R b )(R c )] wherein C(R b ) ⁇ C(R C ) counts as 2 of said biradical groups wherein each R a , R b and R c is, independently, H, C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 2 -C 6 alkenyl or C 2 -C 6 alkynyl.
  • the bridging groups include, but are not limited to 4′-(CH 2 )—O-2′, 4′-(CH 2 )—S-2′, 4′-(CH 2 ) 2 —O-2′, 4′-CH(CH 3 )—O-2′, 4′-CH(CH 2 OCH 3 )—O-2′, 4′-C(CH 3 ) 2 —O-2′, 4′-CH 2 —N(OCH 3 )-2′, 4′-CH 2 —O—N(CH 3 )-2′, 4′-CH 2 —NCH 3 —O-2′, 4′-CH 2 —C(H)(CH 3 )-2′ and 4′-CH 2 —C( ⁇ CH 2 )-2′.
  • the bridging groups include, but are not limited to 4′-CH 2 —O-2′, 4′-(CH 2 ) 2 —O-2′, 4′-C(H)[(R)—CH 3 ]-O-2′ and 4′-C(H)[(S)—CH 3 ]-O-2′.
  • the oligomeric compounds provided herein include DNA-like nucleosides that have been modified to influence the sugar conformation to have predominantly 2′-endo conformational geometry.
  • modified nucleosides can include synthetic modifications of the heterocyclic base moiety, the sugar moiety or both to induce the desired 2′-endo sugar conformation.
  • modified nucleosides are used to mimic RNA nucleosides so that particular properties of an oligomeric compound can be enhanced while maintaining the desirable 2′-endo conformational geometry.
  • nucleosides of the present invention comprise one or more unmodified nucleobases. In certain embodiments, nucleosides of the present invention comprise one or more modified nucleobases (heterocyclic base moieties).
  • a heterocyclic base moiety is any heterocyclic system that contains one or more atoms or groups of atoms capable of hydrogen bonding to a heterocyclic base of a nucleic acid.
  • nucleobase refers to purines, modified purines, pyrimidines and modified pyrimidines.
  • nucleobase refers to unmodified or naturally occurring nucleobases which include, but are not limited to, the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U) and analogs thereof such as 5-methyl cytosine.
  • nucleobase and heterocyclic base moiety also include optional protection for any reactive functional groups such as 4-N-benzoylcytosine, 4-N-benzoyl-5-methylcytosine, 6-N-benzoyladenine or 2-N-isobutyrylguanine.
  • heterocyclic base moieties include without limitation modified nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl (—C ⁇ C—CH 3 ) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines
  • heterocyclic base moieties include without limitation 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).
  • Heterocyclic base moieties 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 heterocyclic base moieties include without limitation those known to the art skilled (see for example: U.S. Pat. No.
  • nucleosides may be linked together using any internucleoside linkage to form oligonucleotides.
  • 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, phosphodiesters (P ⁇ O), phosphotriesters, methylphosphonates, phosphoramidate, and phosphorothioates (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—Si(H) 2 —O—); and N,N′-dimethylhydrazine (—CH 2 —N(CH 3 )—N(CH 3 )—).
  • Modified linkages compared to natural phosphodiester 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 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.
  • oligonucleotides described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), ⁇ or ⁇ such as for sugar anomers, or as (D) or (L) such as for amino acids etc. Included in the antisense compounds provided herein are all such possible isomers, as well as their racemic and optically pure forms.
  • 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′), 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.
  • oligomeric compounds comprise or consist of oligonucleotides.
  • such oligonucleotides comprise one or more chemical modifications.
  • chemically modified oligonucleotides comprise one or more modified sugars.
  • chemically modified oligonucleotides comprise one or more modified nucleobases.
  • chemically modified oligonucleotides comprise one or more modified internucleoside linkages.
  • the chemical modifications (sugar modifications, nucleobase modifications, and/or linkage modifications) define a pattern or motif.
  • the patterns of chemical modifications of sugar moieties, internucleoside linkages, and nucleobases are each independent of one another.
  • an oligonucleotide may be described by its sugar modification motif, internucleoside linkage motif and/or nucleobase modification motif (as used herein, nucleobase modification motif describes the chemical modifications to the nucleobases independent of the sequence of nucleobases).
  • oligonucleotides comprise one or more type of modified sugar moieties and/or naturally occurring sugar moieties arranged along an oligonucleotide or region thereof in a defined pattern or sugar motif.
  • sugar motifs include but are not limited to any of the sugar modifications discussed herein.
  • the oligomeric compounds provided herein comprise a gapmer sugar motif, which comprises two external regions or “wings” and a central or internal region or “gap” (also referred to as 5′-region and 3′-region).
  • the three regions of a gapmer sugar motif (the 5′-wing, the gap, and the 3′-wing) form a contiguous sequence of nucleosides wherein at least some of the sugar moieties of the nucleosides of each of the wings differ from at least some of the sugar moieties of the nucleosides of the gap.
  • the sugar moieties of the nucleosides of each wing that are closest to the gap differ from the sugar moiety of the neighboring gap nucleosides, thus defining the boundary between the wings and the gap.
  • the sugar moieties within the gap are the same as one another.
  • the gap includes one or more nucleoside having a sugar moiety that differs from the sugar moiety of one or more other nucleosides of the gap.
  • the sugar moieties of the two wings are the same as one another (symmetric sugar gapmer).
  • the sugar moieties of the 5′-wing differs from the sugar moieties of the 3′-wing (asymmetric sugar gapmer).
  • the sugar moieties in the two wings are selected from at least two different types that are different from the sugar moieties in the gap and at least one of each are in each wing.
  • the term “gapped oligomeric compound” refers to an oligomeric compound having two external regions or wings and an internal region or gap (also referred to as 5′-region and 3′-region).
  • the three regions form a contiguous sequence of monomer subunits with the sugar moieties of the external regions (wings) being different than the sugar moieties of the internal region (gap).
  • the sugar moieties of each monomer subunit within a particular region is essentially the same.
  • the sugar moieties of each monomer subunit within each wing region is selected independently from 2 different types of modified nucleosides.
  • the sugar moieties of each monomer subunit within each wing region is selected independently from 3 different types of modified nucleosides. In certain embodiments, the sugar moieties of each monomer subunit within each wing region is selected independently from 4 different types of modified nucleosides. In certain embodiments, the sugar moiety of essentially each monomer subunit within the internal region is essentially the same. In certain embodiments, the sugar moiety of each monomer subunit within the internal region is a ⁇ -D-2′-deoxyribonucleoside, a nucleoside that is DNA-like and/or a nucleoside that supports RNaseH when in the gap region.
  • each monomer subunit within a particular region has the same sugar moiety.
  • the gapmer is a symmetric gapmer and when the sugar moiety used in the 5′-external region is different from the sugar moiety used in the 3′-external region, the gapmer is an asymmetric gapmer.
  • the external regions are small (each independently 2, 3, 4, 5 or about 6 monomer subunits) and the monomer subunits comprise non-naturally occurring sugar moieties with the internal region comprising ⁇ -D-2′-deoxyribonucleosides.
  • the external regions each, independently, comprise from 2 to about 8 monomer subunits having non-naturally occurring sugar moieties and the internal region comprises from 6 to 14 unmodified nucleosides.
  • the internal region or the gap generally comprises ⁇ -D-2′-deoxyribonucleosides but can comprise non-naturally occurring sugar moieties.
  • the heterocyclic base and internucleoside linkage is independently variable at each position of a gapped oligomeric compound.
  • a gapped oligomeric compound can further include one or more additional groups including but not limited to capping groups, conjugate groups and other 5′ or 3′-terminal groups.
  • gapped oligomeric compounds comprise an internal region of ⁇ -D-2′-deoxyribonucleosides with a single internucleoside linkage having Formula I. In certain embodiments, gapped oligomeric compounds comprise an internal region of ⁇ -D-2′-deoxyribonucleosides having two internucleoside linkages having Formula I. In certain embodiments, gapped oligomeric compounds comprise an internal region of ⁇ -D-2′-deoxyribonucleosides having three internucleoside linkages having Formula I.
  • the 5′ and 3′-wing regions of gapped oligomeric compounds comprise modified nucleosides wherein all the sugar moieties have the same type of modification such as cEt or MOE.
  • the 5′ and 3′-wing regions of gapped oligomeric compounds comprise two types of modified nucleosides having sugar moieties independently selected from 2′-substituted sugar moieties and furanosyl bicyclic sugar moieties.
  • the 5′ and 3′-wing regions of gapped oligomeric compounds comprise two types of modified nucleosides having sugar moieties independently selected from 2′-MOE substituted sugar moieties and furanosyl bicyclic sugar moieties each having a 4′-CH((S)—CH 3 )—O-2′ bridge.
  • gapped oligomeric compounds are provided that are from about 10 to about 30 monomer subunits in length. In certain embodiments, gapped oligomeric compounds are provided that are from about 12 to about 20 monomer subunits in length. In certain embodiments, gapped oligomeric compounds are provided that are from about 14 to about 20 monomer subunits in length. In certain embodiments, gapped oligomeric compounds are provided that are from about 14 to about 18 monomer subunits in length.
  • oligonucleotides comprise chemical modifications to nucleobases arranged along the oligonucleotide or region thereof in a defined pattern or nucleobases modification motif. In certain embodiments, each nucleobase is modified. In certain embodiments, none of the nucleobases is chemically modified.
  • oligonucleotides comprise a block of modified nucleobases.
  • the block is at the 3′-end of the oligonucleotide.
  • the block is within 3 nucleotides of the 3′-end of the oligonucleotide.
  • the block is at the 5′-end of the oligonucleotide.
  • the block is within 3 nucleotides of the 5′-end of the oligonucleotide.
  • nucleobase modifications are a function of the natural base at a particular position of an oligonucleotide.
  • each purine or each pyrimidine in an oligonucleotide is modified.
  • each adenine is modified.
  • each guanine is modified.
  • each thymine is modified.
  • each cytosine is modified.
  • each uracil is modified.
  • oligonucleotides comprise one or more nucleosides comprising a modified nucleobase.
  • oligonucleotides having a gapmer sugar motif comprise a nucleoside comprising a modified nucleobase.
  • one nucleoside comprising a modified nucleobases is in the gap of an oligonucleotide having a gapmer sugar motif.
  • the sugar is an unmodified 2′ deoxynucleoside.
  • the modified nucleobase is selected from: a 2-thio pyrimidine and a 5-propyne pyrimidine
  • cytosine moieties in an oligonucleotide are 5-methyl cytosine moieties.
  • 5-methyl cytosine is not a “modified nucleobase.” Accordingly, unless otherwise indicated, unmodified nucleobases include both cytosine residues having a 5-methyl and those lacking a 5 methyl. In certain embodiments, the methylation state of all or some cytosine nucleobases is specified.
  • oligomeric compounds comprise nucleosides comprising modified sugar moieties and/or nucleosides comprising modified nucleobases.
  • Such motifs can be described by their sugar motif and their nucleobase motif separately or by their nucleoside motif, which provides positions or patterns of modified nucleosides (whether modified sugar, nucleobase, or both sugar and nucleobase) in an oligonucleotide.
  • the oligomeric compounds comprise or consist of a region having a gapmer nucleoside motif, which comprises two external regions or “wings” and a central or internal region or “gap.”
  • the three regions of a gapmer nucleoside motif (the 5′-wing, the gap, and the 3′-wing) form a contiguous sequence of nucleosides wherein at least some of the sugar moieties and/or nucleobases of the nucleosides of each of the wings differ from at least some of the sugar moieties and/or nucleobase of the nucleosides of the gap.
  • the nucleosides of each wing that are closest to the gap differ from the neighboring gap nucleosides, thus defining the boundary between the wings and the gap.
  • the nucleosides within the gap are the same as one another.
  • the gap includes one or more nucleoside that differs from one or more other nucleosides of the gap.
  • the nucleoside motifs of the two wings are the same as one another (symmetric gapmer).
  • the nucleoside motifs of the 5′-wing differs from the nucleoside motif of the 3′-wing (asymmetric gapmer).
  • the 5′-wing of a gapmer consists of 2 to 8 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 3 to 5 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 4 or 5 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 1 to 4 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 1 to 3 linked nucleosides.
  • the 5′-wing of a gapmer consists of 1 or 2 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 2 to 4 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 2 or 3 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 3 or 4 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 1 nucleoside. In certain embodiments, the 5′-wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 3 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 4 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 5 linked nucleosides.
  • the 5′-wing of a gapmer consists of 2 to 8 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 3 to 5 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 4 or 5 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 2 to 4 linked nucleosides.
  • the 5′-wing of a gapmer consists of 2 to 3 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 3 or 4 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 3 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 4 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 5 linked nucleosides.
  • the 5′-wing of a gapmer consists of 6 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 7 linked nucleosides. In certain embodiments, the 5′-wing of a gapmer consists of 8 linked nucleosides.
  • the 5′-wing of a gapmer comprises at least one bicyclic nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least two bicyclic nucleosides. In certain embodiments, the 5′-wing of a gapmer comprises at least three bicyclic nucleosides. In certain embodiments, the 5′-wing of a gapmer comprises at least four bicyclic nucleosides. In certain embodiments, the 5′-wing of a gapmer comprises at least one constrained ethyl nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least one LNA nucleoside.
  • each nucleoside of the 5′-wing of a gapmer is a bicyclic nucleoside. In certain embodiments, each nucleoside of the 5′-wing of a gapmer is a constrained ethyl nucleoside. In certain embodiments, each nucleoside of the 5′-wing of a gapmer is a LNA nucleoside.
  • the 5′-wing of a gapmer comprises at least one non-bicyclic modified nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least one 2′-substituted nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least one 2′-MOE nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least one 2′-OMe nucleoside. In certain embodiments, each nucleoside of the 5′-wing of a gapmer is a non-bicyclic modified nucleoside.
  • each nucleoside of the 5′-wing of a gapmer is a 2′-substituted nucleoside. In certain embodiments, each nucleoside of the 5′-wing of a gapmer is a 2′-MOE nucleoside. In certain embodiments, each nucleoside of the 5′-wing of a gapmer is a 2′-OMe nucleoside.
  • the 5′-wing of a gapmer comprises at least one bicyclic nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2′-substituted nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2′-MOE nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2′-OMe nucleoside.
  • the 5′-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2′-substituted nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2′-MOE nucleoside. In certain embodiments, the 5′-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2′-OMe nucleoside.
  • the 5′-wing of a gapmer has a nucleoside motif selected from among the following: ABBA; ABB; ABAA; AABAA; AAABAA; AAAABAA; AAAAABAA; AAABAA; AABAA; ABAB; AAABB; AAAAA; ABBC; AA; AAA; AAAA; AAAAB; AAAAAAA; AAAAAAAA; ABBB; AB; ABAB; AAAAB; AABBB; AAAAB; and AABBB, wherein each A is a modified nucleoside of a first type, each B is a modified nucleoside of a second type and each C is a modified nucleoside of a third type.
  • such an oligomeric compound is a gapmer.
  • the 3′-wing of the gapmer may comprise any nucleoside motif.
  • the 3′-wing of a gapmer consists of 2 to 8 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 3 to 5 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 4 or 5 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 1 to 4 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 1 to 3 linked nucleosides.
  • the 3′-wing of a gapmer consists of 1 or 2 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 2 to 4 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 2 or 3 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 3 or 4 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 1 nucleoside. In certain embodiments, the 3′-wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 3 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 4 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 5 linked nucleosides.
  • the 3′-wing of a gapmer consists of 2 to 8 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 3 to 5 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 4 or 5 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 2 to 4 linked nucleosides.
  • the 3′-wing of a gapmer consists of 2 to 3 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 3 or 4 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 3 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 4 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 5 linked nucleosides.
  • the 3′-wing of a gapmer consists of 6 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 7 linked nucleosides. In certain embodiments, the 3′-wing of a gapmer consists of 8 linked nucleosides.
  • the 3′-wing of a gapmer comprises at least one bicyclic nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one constrained ethyl nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one LNA nucleoside. In certain embodiments, each nucleoside of the 3′-wing of a gapmer is a bicyclic nucleoside. In certain embodiments, each nucleoside of the 3′-wing of a gapmer is a constrained ethyl nucleoside. In certain embodiments, each nucleoside of the 3′-wing of a gapmer is a LNA nucleoside.
  • the 3′-wing of a gapmer comprises at least one non-bicyclic modified nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least two non-bicyclic modified nucleosides. In certain embodiments, the 3′-wing of a gapmer comprises at least three non-bicyclic modified nucleosides. In certain embodiments, the 3′-wing of a gapmer comprises at least four non-bicyclic modified nucleosides. In certain embodiments, the 3′-wing of a gapmer comprises at least one 2′-substituted nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one 2′-MOE nucleoside.
  • the 3′-wing of a gapmer comprises at least one 2′-OMe nucleoside.
  • each nucleoside of the 3′-wing of a gapmer is a non-bicyclic modified nucleoside.
  • each nucleoside of the 3′-wing of a gapmer is a 2′-substituted nucleoside.
  • each nucleoside of the 3′-wing of a gapmer is a 2′-MOE nucleoside.
  • each nucleoside of the 3′-wing of a gapmer is a 2′-OMe nucleoside.
  • the 3′-wing of a gapmer comprises at least one bicyclic nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2′-substituted nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2′-MOE nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2′-OMe nucleoside.
  • the 3′-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2′-substituted nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2′-MOE nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2′-OMe nucleoside.
  • the 3′-wing of a gapmer comprises at least one LNA nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one LNA nucleoside and at least one 2′-substituted nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one LNA nucleoside and at least one 2′-MOE nucleoside. In certain embodiments, the 3′-wing of a gapmer comprises at least one LNA nucleoside and at least one 2′-OMe nucleoside.
  • the 3′-wing of a gapmer has a nucleoside motif selected from among the following: ABB; ABAA; AAABAA, AAAAABAA; AABAA; AAAABAA; AAABAA; ABAB; AAAAA; AABBB; AAAAAAAA; AAAAAAA; AAABAA; AAAAB; AAAA; AAA; AA; AB; ABBB; ABAB; AABBB; wherein each A is a modified nucleoside of a first type, each B is a modified nucleoside of a second type.
  • an oligonucleotide comprises any 3′-wing motif provided herein.
  • the 5′-wing of the gapmer may comprise any nucleoside motif.
  • the gap of a gapmer consists of 6 to 20 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 14 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 12 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 or 7 linked nucleosides.
  • the gap of a gapmer consists of 7 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 to 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 or 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 8 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 8 or 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 linked nucleosides.
  • the gap of a gapmer consists of 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 11 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 12 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 13 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 14 linked nucleosides.
  • each nucleoside of the gap of a gapmer is a 2′-deoxynucleoside.
  • the gap comprises one or more modified nucleosides.
  • each nucleoside of the gap of a gapmer is a 2′-deoxynucleoside or is a modified nucleoside that is “DNA-like”.
  • “DNA-like” means that the nucleoside has similar characteristics to DNA, such that a duplex comprising the gapmer and an RNA molecule is capable of activating RNase H.
  • modified nucleosides that are DNA-like are 2′-endo.
  • 2′-(ara)-F have been shown to support RNase H activation, and thus is DNA-like and further has 2′-endo conformation geometry.
  • one or more nucleosides of the gap of a gapmer is not a 2′-deoxynucleoside and is not DNA-like.
  • the gapmer nonetheless supports RNase H activation (e.g., by virtue of the number or placement of the non-DNA nucleosides).
  • the gap comprise a stretch of unmodified 2′-deoxynucleosides interrupted by one or more modified nucleosides, thus resulting in three sub-regions (two stretches of one or more 2′-deoxynucleosides and a stretch of one or more interrupting modified nucleosides).
  • no stretch of unmodified 2′-deoxynucleosides is longer than 5, 6, or 7 nucleosides.
  • such short stretches is achieved by using short gap regions.
  • short stretches are achieved by interrupting a longer gap region.
  • a gapmer comprises a 5′-wing, a gap comprising at least one internucleoside linkage of Formula I, and a 3′ wing, wherein the 5′-wing, gap, and 3′ wing are independently selected from among those discussed above.
  • a gapmer has a 5′-wing, a gap, and a 3′-wing having features selected from among those listed in the following non-limiting table:
  • each A is a modified nucleoside of a first type
  • each B is a modified nucleoside of a second type
  • each D is a ⁇ -D-2′-deoxyribonucleoside or a nucleoside that is DNA-like.
  • Each gap region includes at least one internucleoside linkage of Formula I.
  • each A comprises a modified sugar moiety. In certain embodiments, each A comprises a 2′-substituted sugar moiety. In certain embodiments, each A comprises a 2′-substituted sugar moiety selected from among F, OCH 3 , OCH 2 —C( ⁇ O)—N(H)(CH 3 ) and O(CH 2 ) 2 —OCH 3 . In certain embodiments, each A comprises a bicyclic sugar moiety. In certain embodiments, each A comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2′-thio LNA. In certain embodiments, each A comprises a modified nucleobase. In certain embodiments, each A comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne uridine nucleoside.
  • each B comprises a modified sugar moiety. In certain embodiments, each B comprises a 2′-substituted sugar moiety. In certain embodiments, each B comprises a 2′-substituted sugar moiety selected from among F, OCH 3 , OCH 2 —C( ⁇ O)—N(H)(CH 3 ) and O(CH 2 ) 2 —OCH 3 . In certain embodiments, each B comprises a bicyclic sugar moiety. In certain embodiments, each B comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2′-thio LNA. In certain embodiments, each B comprises a modified nucleobase. In certain embodiments, each B comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne uridine nucleoside.
  • At least one of A or B comprises a bicyclic sugar moiety, and the other comprises a 2′-substituted sugar moiety.
  • one of A or B is an LNA nucleoside and the other of A or B comprises a 2′-substituted sugar moiety.
  • one of A or B is a cEt nucleoside and the other of A or B comprises a 2′-substituted sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2′-substituted sugar moiety.
  • one of A or B is an LNA nucleoside and the other of A or B comprises a 2′-MOE sugar moiety. In certain embodiments, one of A or B is a cEt nucleoside and the other of A or B comprises a 2′-MOE sugar moiety. In certain embodiments, one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2′-MOE sugar moiety. In certain embodiments, one of A or B is an LNA nucleoside and the other of A or B comprises a 2′-F sugar moiety.
  • one of A or B is a cEt nucleoside and the other of A or B comprises a 2′-F sugar moiety. In certain embodiments, one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2′-F sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2′-substituted sugar moiety.
  • A is an LNA nucleoside and B comprises a 2′-substituted sugar moiety.
  • A is a cEt nucleoside and B comprises a 2′-substituted sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2′-substituted sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2′-MOE sugar moiety.
  • A is an LNA nucleoside and B comprises a 2′-MOE sugar moiety.
  • A is a cEt nucleoside and B comprises a 2′-MOE sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2′-MOE sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2′-F sugar moiety.
  • A is an LNA nucleoside and B comprises a 2′-F sugar moiety.
  • A is a cEt nucleoside and B comprises a 2′-F sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2′-F sugar moiety.
  • B comprises a bicyclic sugar moiety, and A comprises a 2′-MOE sugar moiety.
  • B is an LNA nucleoside and A comprises a 2′-MOE sugar moiety.
  • B is a cEt nucleoside and A comprises a 2′-MOE sugar moiety.
  • B is an ⁇ -L-LNA nucleoside and A comprises a 2′-MOE sugar moiety.
  • B comprises a bicyclic sugar moiety, and A comprises a 2′-F sugar moiety.
  • B is an LNA nucleoside and A comprises a 2′-F sugar moiety.
  • B is a cEt nucleoside and A comprises a 2′-F sugar moiety.
  • B is an ⁇ -L-LNA nucleoside and A comprises a 2′-F sugar moiety.
  • each A and B is, independently, a modified nucleoside comprising a bicyclic sugar moiety comprising a 4′-CH(CH 3 )—O-2′ bridge or a modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group.
  • each A and B is, independently, a modified nucleoside comprising a bicyclic sugar moiety comprising a 4′-CH[(S)—(CH 3 )]—O-2′ bridge or a modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group.
  • each A and B is, independently, a modified nucleoside comprising a bicyclic sugar moiety comprising a 4′-CH[(R)—(CH 3 )]—O-2′ bridge or a modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group.
  • at least one modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group and at least one modified nucleoside comprising a 4′-CH(CH 3 )—O-2′ bridge is located in each of the 3′ and 5′ wings.
  • At least one modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group and at least one modified nucleoside comprising a 4′-CH[(S)—(CH 3 )]—O-2′ bridge is located in each of the 3′ and 5′ wings.
  • at least one modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group and at least one modified nucleoside comprising a 4′-CH[(R)—(CH 3 )]—O-2′ bridge is located in each of the 3′ and 5′ wings.
  • oligomeric compounds comprise modified internucleoside linkages arranged along the oligomeric compound or region thereof in a defined pattern or modified internucleoside linkage motif provided that at least one internucleoside linkage has Formula I.
  • internucleoside linkages are arranged in a gapped motif, as described above for nucleoside motif.
  • the internucleoside linkages in each of two wing regions are different from the internucleoside linkages in the gap region.
  • the internucleoside linkages in the wings are phosphodiester and the internucleoside linkages in the gap are phosphorothioate.
  • the nucleoside motif is independently selected, so such oligomeric compounds having a gapped internucleoside linkage motif may or may not have a gapped nucleoside motif and if it does have a gapped nucleoside motif, the wing and gap lengths may or may not be the same.
  • oligomeric compounds comprise a region having an alternating internucleoside linkage motif. In certain embodiments, oligomeric compounds of the present invention comprise a region of uniformly modified internucleoside linkages. In certain such embodiments, the oligomeric compound comprises a region that is uniformly linked by phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide is uniformly linked by phosphorothioate. In certain embodiments, each internucleoside linkage of the oligomeric compound is selected from phosphodiester and phosphorothioate.
  • each internucleoside linkage of the oligomeric compound is selected from phosphodiester and phosphorothioate and at least one internucleoside linkage is phosphorothioate. In certain embodiments, at least one internucleoside linkage of the oligomeric compound is selected from other than phosphodiester and phosphorothioate.
  • the oligomeric compound comprises at least 6 phosphorothioate internucleoside linkages. In certain embodiments, the oligomeric compound comprises at least 8 phosphorothioate internucleoside linkages. In certain embodiments, the oligomeric compound comprises at least 10 phosphorothioate internucleoside linkages. In certain embodiments, the oligomeric compound comprises at least one block of at least 6 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligomeric compound comprises at least one block of at least 8 consecutive phosphorothioate internucleoside linkages.
  • the oligomeric compound comprises at least one block of at least 10 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligomeric compound comprises at least one block of at least one 12 consecutive phosphorothioate internucleoside linkages. In certain such embodiments, at least one such block is located at the 3′ end of the oligomeric compound. In certain such embodiments, at least one such block is located within 3 nucleosides of the 3′ end of the oligomeric compound. In certain embodiments, each internucleoside linkage a phosphorothioate internucleoside linkage.
  • Modification motifs define oligonucleotides by nucleoside motif (sugar motif and nucleobase motif) and linkage motif.
  • nucleoside motif sucrose motif and nucleobase motif
  • linkage motif For example, certain oligonucleotides have the following modification motif:
  • each A is a modified nucleoside comprising a 2′-substituted sugar moiety
  • each D is a ⁇ -D-2′-deoxyribonucleoside or a modified nucleoside having B form conformation geometry
  • each B is a modified nucleoside comprising a bicyclic sugar moiety wherein at least one internucleoside linkage had Formula I.
  • the following non-limiting Table further illustrates certain modification motifs:
  • each A is a modified nucleoside of a first type
  • each B is a modified nucleoside of a second type
  • each D is a ⁇ -D-2′-deoxyribonucleoside or a nucleoside that is DNA-like.
  • Each gap region includes at least one internucleoside linkage of Formula I.
  • each A comprises a modified sugar moiety. In certain embodiments, each A comprises a 2′-substituted sugar moiety. In certain embodiments, each A comprises a 2′-substituted sugar moiety selected from among F, OCH 3 , OCH 2 —C( ⁇ O)—N(H)(CH 3 ) and O(CH 2 ) 2 —OCH 3 . In certain embodiments, each A comprises a bicyclic sugar moiety. In certain embodiments, each A comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2′-thio LNA. In certain embodiments, each A comprises a modified nucleobase. In certain embodiments, each A comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne uridine nucleoside.
  • each B comprises a modified sugar moiety. In certain embodiments, each B comprises a 2′-substituted sugar moiety. In certain embodiments, each B comprises a 2′-substituted sugar moiety selected from among F, OCH 3 , OCH 2 —C( ⁇ O)—N(H)(CH 3 ) and O(CH 2 ) 2 —OCH 3 . In certain embodiments, each B comprises a bicyclic sugar moiety. In certain embodiments, each B comprises a bicyclic sugar moiety selected from among cEt, cMOE, LNA, ⁇ -L-LNA, ENA and 2′-thio LNA. In certain embodiments, each B comprises a modified nucleobase. In certain embodiments, each B comprises a modified nucleobase selected from among 2-thio-thymidine nucleoside and 5-propyne urindine nucleoside.
  • At least one of A or B comprises a bicyclic sugar moiety, and the other comprises a 2′-substituted sugar moiety.
  • one of A or B is an LNA nucleoside and the other of A or B comprises a 2′-substituted sugar moiety.
  • one of A or B is a cEt nucleoside and the other of A or B comprises a 2′-substituted sugar moiety.
  • one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2′-substituted sugar moiety.
  • one of A or B is an LNA nucleoside and the other of A or B comprises a 2′-MOE sugar moiety. In certain embodiments, one of A or B is a cEt nucleoside and the other of A or B comprises a 2′-MOE sugar moiety. In certain embodiments, one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2′-MOE sugar moiety. In certain embodiments, one of A or B is an LNA nucleoside and the other of A or B comprises a 2′-F sugar moiety.
  • one of A or B is a cEt nucleoside and the other of A or B comprises a 2′-F sugar moiety. In certain embodiments, one of A or B is an ⁇ -L-LNA nucleoside and the other of A or B comprises a 2′-F sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2′-substituted sugar moiety.
  • A is an LNA nucleoside and B comprises a 2′-substituted sugar moiety.
  • A is a cEt nucleoside and B comprises a 2′-substituted sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2′-substituted sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2′-MOE sugar moiety.
  • A is an LNA nucleoside and B comprises a 2′-MOE sugar moiety.
  • A is a cEt nucleoside and B comprises a 2′-MOE sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2′-MOE sugar moiety.
  • A comprises a bicyclic sugar moiety, and B comprises a 2′-F sugar moiety.
  • A is an LNA nucleoside and B comprises a 2′-F sugar moiety.
  • A is a cEt nucleoside and B comprises a 2′-F sugar moiety.
  • A is an ⁇ -L-LNA nucleoside and B comprises a 2′-F sugar moiety.
  • B comprises a bicyclic sugar moiety, and A comprises a 2′-MOE sugar moiety.
  • B is an LNA nucleoside and A comprises a 2′-MOE sugar moiety.
  • B is a cEt nucleoside and A comprises a 2′-MOE sugar moiety.
  • B is an ⁇ -L-LNA nucleoside and A comprises a 2′-MOE sugar moiety.
  • B comprises a bicyclic sugar moiety, and A comprises a 2′-F sugar moiety.
  • B is an LNA nucleoside and A comprises a 2′-F sugar moiety.
  • B is a cEt nucleoside and A comprises a 2′-F sugar moiety.
  • B is an ⁇ -L-LNA nucleoside and A comprises a 2′-F sugar moiety.
  • each A and B is, independently, a modified nucleoside comprising a bicyclic sugar moiety comprising a 4′-CH(CH 3 )—O-2′ bridge or a modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group.
  • each A and B is, independently, a modified nucleoside comprising a bicyclic sugar moiety comprising a 4′-CH[(S)—(CH 3 )]—O-2′ bridge or a modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group.
  • each A and B is, independently, a modified nucleoside comprising a bicyclic sugar moiety comprising a 4′-CH[(R)—(CH 3 )]—O-2′ bridge or a modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group.
  • at least one modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group and at least one modified nucleoside comprising a 4′-CH(CH 3 )—O-2′ bridge is located in each of the 3′ and 5′ wings.
  • At least one modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group and at least one modified nucleoside comprising a 4′-CH[(S)—(CH 3 )]—O-2′ bridge is located in each of the 3′ and 5′ wings.
  • at least one modified nucleoside comprising a 2′-OCH 2 CH 2 OCH 3 (MOE) substituent group and at least one modified nucleoside comprising a 4′-CH[(R)—(CH 3 )]—O-2′ bridge is located in each of the 3′ and 5′ wings.
  • the present invention provides oligomeric compounds including oligonucleotides of any of a variety of ranges of lengths.
  • the invention provides oligomeric compounds or oligonucleotides consisting of X to Y linked nucleosides, where X represents the fewest number of nucleosides in the range and Y represents the largest number of nucleosides in the range.
  • X and Y are each independently selected from 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 and 30; provided that X ⁇ Y.
  • the invention provides oligomeric compounds which comprise oligonucleotides consisting of 10 to 11, 10 to 12, 10 to 13, 10 to 14, 10 to 15, 10 to 16, 10 to 17, 10 to 18, 10 to 19, 10 to 20, 10 to 21, 10 to 22, 10 to 23, 10 to 24, 10 to 25, 10 to 26, 10 to 27, 10 to 28, 10 to 29, 10 to 30, 11 to 12, 11 to 13, 11 to 14, 11 to 15, 11 to 16, 11 to 17, 11 to 18, 11 to 19, 11 to 20, 11 to 21, 11 to 22, 11 to 23, 11 to 24, 11 to 25, 11 to 26, 11 to 27, 11 to 28, 11 to 29, 11 to 30, 12 to 13, 12 to 14, 12 to 15, 12 to 16, 12 to 17, 12 to 18, 12 to 19, 12 to 20, 12 to 21, 12 to 22, 12 to 23, 12 to 24, 12 to 25, 12 to 26, 12 to 27, 12 to 28, 12 to 29, 12 to 30, 13 to 14, 13 to 15, 13 to 16, 13 to 17, 13 to 18, 13 to 19, 13 to 20, 13 to 21, 13 to 22, 13 to 23, 13 to 24, 13 to 25, 13 to 26, 13 to 27, 13 to 28, 13 to 29, 13 to 30, 14 to 15, 14 to 15, 14 to 11, to 11,
  • the oligomeric compound or oligonucleotide may, nonetheless further comprise additional other substituents.
  • an oligonucleotide comprising 8-30 nucleosides excludes oligonucleotides having 31 nucleosides, but, unless otherwise indicated, such an oligonucleotide may further comprise, for example one or more conjugates, terminal groups, or other substituents.
  • a gapmer oligonucleotide has any of the above lengths.
  • an oligonucleotide is described by an overall length range and by regions having specified lengths, and where the sum of specified lengths of the regions is less than the upper limit of the overall length range, the oligonucleotide may have additional nucleosides, beyond those of the specified regions, provided that the total number of nucleosides does not exceed the upper limit of the overall length range.
  • oligonucleotides of the present invention are characterized by their modification motif and overall length. In certain embodiments, such parameters are each independent of one another.
  • each internucleoside linkage of an oligonucleotide having a gapmer sugar motif may be modified or unmodified and may or may not follow the gapmer modification pattern of the sugar modifications.
  • the internucleoside linkages within the wing regions of a sugar-gapmer may be the same or different from one another and may be the same or different from the internucleoside linkages of the gap region.
  • sugar-gapmer oligonucleotides may comprise one or more modified nucleobase independent of the gapmer pattern of the sugar modifications.
  • oligonucleotide motifs may be combined to create a variety of oligonucleotides.
  • oligonucleotide or oligomeric compound is silent with respect to one or more parameter, such parameter is not limited.
  • an oligomeric compound described only as having a gapmer sugar motif without further description may have any length, internucleoside linkage motif, and nucleobase modification motif. Unless otherwise indicated, all chemical modifications are independent of nucleobase sequence.
  • oligomeric compounds are modified by attachment of one or more conjugate groups.
  • conjugate groups modify one or more properties of the attached oligomeric compound including but not limited to pharmacodynamics, pharmacokinetics, stability, binding, absorption, cellular distribution, cellular uptake, charge and clearance.
  • Conjugate groups are routinely used in the chemical arts and are linked directly or via an optional conjugate linking moiety or conjugate linking group to a parent compound such as an oligomeric compound, such as an oligonucleotide.
  • Conjugate groups includes without limitation, intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, thioethers, polyethers, cholesterols, thiocholesterols, cholic acid moieties, folate, lipids, phospholipids, biotin, phenazine, phenanthridine, anthraquinone, adamantane, acridine, fluoresceins, rhodamines, coumarins 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), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923-937).
  • a conjugate group comprises an active drug substance, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, (5)-(+)-pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, 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.
  • active drug substance for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, (5)-(+)-pranoprofen, carprofen, dansylsarco
  • conjugate groups are directly attached to oligonucleotides in oligomeric compounds.
  • conjugate groups are attached to oligonucleotides by a conjugate linking group.
  • conjugate linking groups including, but not limited to, bifunctional linking moieties such as those known in the art are amenable to the compounds provided herein.
  • Conjugate linking groups are useful for attachment of conjugate groups, such as chemical stabilizing groups, functional groups, reporter groups and other groups to selective sites in a parent compound such as for example an oligomeric compound.
  • a bifunctional linking moiety comprises a hydrocarbyl moiety having two functional groups.
  • One of the functional groups is selected to bind to a parent molecule or compound of interest and the other is selected to bind essentially any selected group such as chemical functional group or a conjugate group.
  • the conjugate linker comprises a chain structure or an oligomer of repeating units such as ethylene glycol or amino acid units.
  • functional groups that are routinely 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 include amino, hydroxyl, carboxylic acid, thiol, unsaturations (e.g., double or triple bonds), and the like.
  • conjugate linking moieties include 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
  • linking groups include, but are not limited to, substituted 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.
  • Conjugate groups may be attached to either or both ends of an oligonucleotide (terminal conjugate groups) and/or at any internal position.
  • conjugate groups are at the 3′-end of an oligonucleotide of an oligomeric compound. In certain embodiments, conjugate groups are near the 3′-end. In certain embodiments, conjugates are attached at the 3′ end of an oligomeric compound, but before one or more terminal group nucleosides. In certain embodiments, conjugate groups are placed within a terminal group.
  • oligomeric compounds comprise an oligonucleotide.
  • an oligomeric compound comprises an oligonucleotide and one or more conjugate and/or terminal groups.
  • conjugate and/or terminal groups may be added to oligonucleotides having any of the motifs discussed above.
  • an oligomeric compound comprising an oligonucleotide having region of alternating nucleosides may comprise a terminal group.
  • oligomeric compounds provided herein are antisense compounds. Such antisense compounds are capable of hybridizing to a target nucleic acid, resulting in at least one antisense activity. In certain embodiments, antisense compounds specifically hybridize to one or more target nucleic acid.
  • a specifically hybridizing antisense compound has a nucleobase sequence comprising a region having sufficient complementarity to a target nucleic acid to allow hybridization and result in antisense activity and insufficient complementarity to any non-target so as to avoid non-specific hybridization to any non-target nucleic acid sequences under conditions in which specific hybridization is desired (e.g., under physiological conditions for in vivo or therapeutic uses, and under conditions in which assays are performed in the case of in vitro assays).
  • the present invention provides antisense compounds comprising oligonucleotides that are fully complementary to the target nucleic acid over the entire length of the oligonucleotide. In certain embodiments, oligonucleotides are 99% complementary to the target nucleic acid. In certain embodiments, oligonucleotides are 95% complementary to the target nucleic acid. In certain embodiments, such oligonucleotides are 90% complementary to the target nucleic acid.
  • such oligonucleotides are 85% complementary to the target nucleic acid. In certain embodiments, such oligonucleotides are 80% complementary to the target nucleic acid. In certain embodiments, an antisense compound comprises a region that is fully complementary to a target nucleic acid and is at least 80% complementary to the target nucleic acid over the entire length of the oligonucleotide. In certain such embodiments, the region of full complementarity is from 6 to 14 nucleobases in length.
  • hybridization of an antisense compound results in recruitment of a protein that cleaves a target nucleic acid.
  • certain antisense compounds result in RNase H mediated cleavage of target nucleic acid.
  • RNase H is a cellular endonuclease that cleaves the RNA strand of an RNA:DNA duplex.
  • the “DNA” in such an RNA:DNA duplex need not be unmodified DNA.
  • the invention provides antisense compounds that are sufficiently “DNA-like” to elicit RNase H activity.
  • DNA-like antisense compounds include, but are not limited to gapmers having unmodified deoxyfuranose sugar moieties in the nucleosides of the gap and modified sugar moieties in the nucleosides of the wings.
  • Antisense activities may be observed directly or indirectly.
  • observation or detection of an antisense activity involves observation or detection of a change in an amount of a target nucleic acid or protein encoded by such target nucleic acid; a change in the ratio of splice variants of a nucleic acid or protein; and/or a phenotypic change in a cell or animal.
  • compounds comprising oligonucleotides having a gapmer nucleoside motif described herein have desirable properties compared to non-gapmer oligonucleotides or to gapmers having other motifs. In certain circumstances, it is desirable to identify motifs resulting in a favorable combination of potent antisense activity and relatively low toxicity. In certain embodiments, compounds of the present invention have a favorable therapeutic index (measure of potency divided by measure of toxicity).
  • antisense compounds provided herein are selective for a target relative to a non-target nucleic acid.
  • the nucleobase sequences of the target and non-target nucleic acids differ by no more than 4 differentiating nucleobases in the targeted region.
  • the nucleobase sequences of the target and non-target nucleic acids differ by no more than 3 differentiating nucleobases in the targeted region.
  • the nucleobase sequences of the target and non-target nucleic acids differ by no more than 2 differentiating nucleobases in the targeted region.
  • the nucleobase sequences of the target and non-target nucleic acids differ by a single differentiating nucleobase in the targeted region.
  • the target and non-target nucleic acids are transcripts from different genes.
  • the target and non-target nucleic acids are different alleles for the same gene.
  • antisense compounds are achieved, principally, by nucleobase complementarity. For example, if an antisense compound has no mismatches for a target nucleic acid and one or more mismatches for a non-target nucleic acid, some amount of selectivity for the target nucleic acid will result. In certain embodiments, provided herein are antisense compounds with enhanced selectivity (i.e. the ratio of activity for the target to the activity for non-target is greater).
  • a selective nucleoside comprises a particular feature or combination of features (e.g., chemical modification, motif, placement of selective nucleoside, and/or self-complementary region) that increases selectivity of an antisense compound compared to an antisense compound not having that feature or combination of features.
  • a feature or combination of features increases antisense activity for the target.
  • such feature or combination of features decreases activity for the target, but decreases activity for the non-target by a greater amount, thus resulting in an increase in selectivity.
  • a selective antisense compound comprises a modified nucleoside at that same position as a differentiating nucleobase (i.e., the selective nucleoside is modified). That modification may increase the difference in binding affinity of the antisense compound for the target relative to the non-target.
  • the chemical modification may increase the difference in RNAse H activity for the duplex formed by the antisense compound and its target compared to the RNase activity for the duplex formed by the antisense compound and the non-target.
  • the modification may exaggerate a structure that is less compatible for RNase H to bind, cleave and/or release the non-target.
  • Antisense compounds having certain specified motifs have enhanced selectivity, including, but not limited to motifs described above.
  • enhanced selectivity is achieved by oligonucleotides comprising any one or more of:
  • a modification motif comprising a long 5′-wing (longer than 5, 6, or 7 nucleosides);
  • a modification motif comprising a long 3′-wing (longer than 5, 6, or 7 nucleosides);
  • a modification motif comprising a short gap region (shorter than 8, 7, or 6 nucleosides);
  • a modification motif comprising an interrupted gap region (having no uninterrupted stretch of unmodified 2′-deoxynucleosides longer than 7, 6 or 5).
  • selective antisense compounds comprise nucleobase sequence elements.
  • nucleobase sequence elements are independent of modification motifs. Accordingly, oligonucleotides having any of the motifs (modification motifs, nucleoside motifs, sugar motifs, nucleobase modification motifs, and/or linkage motifs) may also comprise one or more of the following nucleobase sequence elements.
  • a target region and a region of a non-target nucleic acid differ by 1-4 differentiating nucleobase.
  • selective antisense compounds have a nucleobase sequence that aligns with the non-target nucleic acid with 1-4 mismatches.
  • a nucleoside of the antisense compound that corresponds to a differentiating nucleobase of the target nucleic acid is referred to herein as a target-selective nucleoside.
  • selective antisense compounds having a gapmer motif align with a non-target nucleic acid, such that a target-selective nucleoside is positioned in the gap.
  • a target-selective nucleoside is the 1 st nucleoside of the gap from the 5′ end. In certain embodiments, a target-selective nucleoside is the 2 nd nucleoside of the gap from the 5′ end. In certain embodiments, a target-selective nucleoside is the 3 rd nucleoside of the gap from the 5′-end. In certain embodiments, a target-selective nucleoside is the 4 th nucleoside of the gap from the 5′-end. In certain embodiments, a target-selective nucleoside is the 5 th nucleoside of the gap from the 5′-end.
  • a target-selective nucleoside is the 6 rd nucleoside of the gap from the 5′-end. In certain embodiments, a target-selective nucleoside is the 8 th nucleoside of the gap from the 3′-end. In certain embodiments, a target-selective nucleoside is the 7 th nucleoside of the gap from the 3′-end. In certain embodiments, a target-selective nucleoside is the 6 th nucleoside of the gap from the 3′-end. In certain embodiments, a target-selective nucleoside is the 5 th nucleoside of the gap from the 3′-end.
  • a target-selective nucleoside is the 4 th nucleoside of the gap from the 3′-end. In certain embodiments, a target-selective nucleoside is the 3 rd nucleoside of the gap from the 3′-end. In certain embodiments, a target-selective nucleoside is the 2 nd nucleoside of the gap from the 3′-end.
  • selective antisense compounds comprise one or more mismatched nucleobases relative to the target nucleic acid.
  • antisense activity against the target is reduced by such mismatch, but activity against the non-target is reduced by a greater amount.
  • selectivity is improved.
  • Any nucleobase other than the differentiating nucleobase is suitable for a mismatch.
  • the mismatch is specifically positioned within the gap of an oligonucleotide having a gapmer motif.
  • a mismatch relative to the target nucleic acid is at positions 1, 2, 3, 4, 5, 6, 7, or 8 from the 5′-end of the gap region.
  • a mismatch relative to the target nucleic acid is at positions 9, 8, 7, 6, 5, 4, 3, 2, 1 of the antisense compounds from the 3′-end of the gap region. In certain embodiments, a mismatch relative to the target nucleic acid is at positions 1, 2, 3, or 4 of the antisense compounds from the 5′-end of the wing region. In certain embodiments, a mismatch relative to the target nucleic acid is at positions 4, 3, 2, or 1 of the antisense compounds from the 3′-end of the wing region.
  • selective antisense compounds comprise a region that is not complementary to the target. In certain embodiments, such region is complementary to another region of the antisense compound. Such regions are referred to herein as self-complementary regions.
  • an antisense compound has a first region at one end that is complementary to a second region at the other end. In certain embodiments, one of the first and second regions is complementary to the target nucleic acid. Unless the target nucleic acid also includes a self-complementary region, the other of the first and second region of the antisense compound will not be complementary to the target nucleic acid.
  • certain antisense compounds have the following nucleobase motif:
  • each of A, B, and C are any nucleobase; A′, B′, and C′ are the complementary bases to A, B, and C, respectively; each X is a nucleobase complementary to the target nucleic acid; and two letters in parentheses (e.g., (X/C′)) indicates that the nucleobase is complementary to the target nucleic acid and to the designated nucleoside within the antisense oligonucleotide.
  • such antisense compounds are expected to form self-structure, which is disrupted upon contact with a target nucleic acid.
  • Contact with a non-target nucleic acid is expected to disrupt the self-structure to a lesser degree, thus increasing selectivity compared to the same antisense compound lacking the self-complementary regions.
  • a single antisense compound may include any one, two, three, or more of self-complementary regions, a mismatch relative to the target nucleic acid, a short nucleoside gap, an interrupted gap, and specific placement of the selective nucleoside.
  • antisense compounds comprise or consist of an oligonucleotide comprising a region that is complementary to a target nucleic acid.
  • the target nucleic acid is an endogenous RNA molecule.
  • the target nucleic acid is a non-coding RNA.
  • the target non-coding RNA is selected from: a long-non-coding RNA, a short non-coding RNA, an intronic RNA molecule, a snoRNA, a scaRNA, a microRNA (including pre-microRNA and mature microRNA), a ribosomal RNA, and promoter directed RNA.
  • the target nucleic acid encodes a protein.
  • the target nucleic acid is selected from: an mRNA and a pre-mRNA, including intronic, exonic and untranslated regions.
  • oligomeric compounds are at least partially complementary to more than one target nucleic acid.
  • antisense compounds of the present invention may mimic microRNAs, which typically bind to multiple targets.
  • the target nucleic acid is a nucleic acid other than a mature mRNA. In certain embodiments, the target nucleic acid is a nucleic acid other than a mature mRNA or a microRNA. In certain embodiments, the target nucleic acid is a non-coding RNA other than a microRNA. In certain embodiments, the target nucleic acid is a non-coding RNA other than a microRNA or an intronic region of a pre-mRNA. In certain embodiments, the target nucleic acid is a long non-coding RNA. In certain embodiments, the target RNA is an mRNA. In certain embodiments, the target nucleic acid is a pre-mRNA.
  • the target region is entirely within an intron. In certain embodiments, the target region spans an intron/exon junction. In certain embodiments, the target region is at least 50% within an intron. In certain embodiments, the target nucleic acid is selected from among non-coding RNA, including exonic regions of pre-mRNA. In certain embodiments, the target nucleic acid is a ribosomal RNA (rRNA). In certain embodiments, the target nucleic acid is a non-coding RNA associated with splicing of other pre-mRNAs. In certain embodiments, the target nucleic acid is a nuclear-retained non-coding RNA.
  • rRNA ribosomal RNA
  • antisense compounds described herein are complementary to a target nucleic acid comprising a single-nucleotide polymorphism.
  • the antisense compound is capable of modulating expression of one allele of the single-nucleotide polymorphism-containing-target nucleic acid to a greater or lesser extent than it modulates another allele.
  • an antisense compound hybridizes to a single-nucleotide polymorphism-containing-target nucleic acid at the single-nucleotide polymorphism site.
  • the target nucleic acid is a Huntingtin gene transcript.
  • the target nucleic acid is a single-nucleotide polymorphism-containing-target nucleic acid of a Huntingtin gene transcript. In certain embodiments, the target nucleic acid is not a Huntingtin gene transcript. In certain embodiments, the target nucleic acid is a single-nucleotide polymorphism-containing-target nucleic acid of a gene transcript other than Huntingtin. In certain embodiments, the target nucleic acid is any nucleic acid other than a Huntingtin gene transcript.
  • Embodiments of the present invention provide methods, compounds, and compositions for selectively inhibiting mRNA and protein expression of an allelic variant of a particular gene or DNA sequence.
  • the allelic variant contains a single nucleotide polymorphism (SNP).
  • SNP single nucleotide polymorphism
  • a SNP is associated with a mutant allele.
  • a mutant SNP is associated with a disease.
  • a mutant SNP is associated with a disease, but is not causative of the disease.
  • mRNA and protein expression of a mutant allele is associated with disease.
  • the expressed gene product of a mutant allele results in aggregation of the mutant proteins causing disease. In certain embodiments, the expressed gene product of a mutant allele results in gain of function causing disease.
  • genes with an autosomal dominant mutation resulting in a toxic gain of function of the protein are the APP gene encoding amyloid precursor protein involved in Alzheimer's disease (Gene, 371: 68, 2006); the PrP gene encoding prion protein involved in Creutzfeldt-Jakob disease and in fatal familial insomnia (Nat. Med. 1997, 3: 1009); GFAP gene encoding glial fibrillary acidic protein involved in Alexander disease (J. Neurosci.
  • alpha-synuclein gene encoding alpha-synuclein protein involved in Parkinson's disease (J. Clin. Invest. 2003, 111: 145); SOD-1 gene encoding the SOD-1 protein involved in amyotrophic lateral sclerosis (Science 1998, 281: 1851); atrophin-1 gene encoding atrophin-1 protein involved in dentato-rubral and pallido-luysian atrophy (DRPA) (Trends Mol. Med. 2001, 7: 479); SCA1 gene encoding ataxin-1 protein involved in spino-cerebellar ataxia-1 (SCA1) (Protein Sci.
  • Ltk gene encoding leukocyte tyrosine kinase protein involved in systemic lupus erythematosus (Hum. Mol. Gen. 2004, 13: 171); PCSK9 gene encoding PCSK9 protein involved in hypercholesterolemia (Hum Mutat. 2009, 30: 520); prolactin receptor gene encoding prolactin receptor protein involved in breast tumors (Proc. Natl. Assoc. Sci. 2008, 105: 4533); CCL5 gene encoding the chemokine CCL5 involved in COPD and asthma (Eur. Respir. J.
  • PTPN22 gene encoding PTPN22 protein involved in Type 1 diabetes, Rheumatoid arthritis, Graves disease, and SLE (Proc. Natl. Assoc. Sci. 2007, 104: 19767); androgen receptor gene encoding the androgen receptor protein involved in spinal and bulbar muscular atrophy or Kennedy's disease (J Steroid Biochem. Mol. Biol. 2008, 108: 245); CHMP4B gene encoding chromatin modifying protein-4B involved in progressive childhood posterior subcapsular cataracts (Am. J. Hum.
  • FXR/NR1H4 gene encoding Farnesoid X receptor protein involved in cholesterol gallstone disease, arthrosclerosis and diabetes (Mol. Endocrinol. 2007, 21: 1769); ABCA1 gene encoding ABCA1 protein involved in cardiovascular disease (Transl. Res. 2007, 149: 205); CaSR gene encoding the calcium sensing receptor protein involved in primary hypercalciuria (Kidney Int. 2007, 71: 1155); alpha-globin gene encoding alpha-globin protein involved in alpha-thallasemia (Science 2006, 312: 1215); httlpr gene encoding HTTLPR protein involved in obsessive compulsive disorder (Am. J.
  • Hum. Genet. 2006, 78: 815 AVP gene encoding arginine vasopressin protein in stress-related disorders such as anxiety disorders and comorbid depression (CNS Neurol. Disord. Drug Targets 2006, 5: 167); GNAS gene encoding G proteins involved in congenital visual defects, hypertension, metabolic syndrome (Trends Pharmacol. Sci. 2006, 27: 260); APAF1 gene encoding APAF1 protein involved in a predisposition to major depression (Mol. Psychiatry 2006, 11: 76); TGF-beta1 gene encoding TGF-beta1 protein involved in breast cancer and prostate cancer (Cancer Epidemiol. Biomarkers Prev.
  • AChR gene encoding acetylcholine receptor involved in congential myasthenic syndrome (Neurology 2004, 62: 1090); P2Y12 gene encoding adenosine diphosphate (ADP) receptor protein involved in risk of peripheral arterial disease (Circulation 2003, 108: 2971); LQT1 gene encoding LQT1 protein involved in atrial fibrillation (Cardiology 2003, 100: 109); RET protooncogene encoding RET protein involved in sporadic pheochromocytoma (J. Clin. Endocrinol. Metab.
  • CA4 gene encoding carbonic anhydrase 4 protein, CRX gene encoding cone-rod homeobox transcription factor protein, FSCN2 gene encoding retinal fascin homolog 2 protein, IMPDH1 gene encoding inosine monophosphate dehydrogenase 1 protein, NR2E3 gene encoding nuclear receptor subfamily 2 group E3 protein, NRL gene encoding neural retina leucine zipper protein, PRPF3 (RP18) gene encoding pre-mRNA splicing factor 3 protein, PRPF8 (RP13) gene encoding pre-mRNA splicing factor 8 protein, PRPF31 (RP 11) gene encoding pre-mRNA splicing factor 31 protein, RDS gene encoding peripherin 2 protein, ROM1 gene encoding rod outer membrane protein 1 protein, RHO gene encoding rhodopsin protein, RP1 gene encoding RP1 protein, RPGR gene encoding retinitis pigmentosa GTP
  • the mutant allele is associated with any disease from the group consisting of Alzheimer's disease, Creutzfeldt-Jakob disease, fatal familial insomnia, Alexander disease, Parkinson's disease, amyotrophic lateral sclerosis, dentato-rubral and pallido-luysian atrophy DRPA, spino-cerebellar ataxia, Torsion dystonia, cardiomyopathy, chronic obstructive pulmonary disease (COPD), liver disease, hepatocellular carcinoma, systemic lupus erythematosus, hypercholesterolemia, breast cancer, asthma, Type 1 diabetes, Rheumatoid arthritis, Graves disease, SLE, spinal and bulbar muscular atrophy, Kennedy's disease, progressive childhood posterior subcapsular cataracts, cholesterol gallstone disease, arthrosclerosis, cardiovascular disease, primary hypercalciuria, alpha-thallasemia, obsessive compulsive disorder, Anxiety, comorbid depression, congenital visual defects, hypertension, metabolic syndrome,
  • any disease
  • an allelic variant of huntingtin is selectively reduced.
  • Nucleotide sequences that encode huntingtin include, without limitation, the following: GENBANK Accession No. NT — 006081.18, truncated from nucleotides 1566000 to 1768000 (replaced by GENBANK Accession No. NT — 006051), incorporated herein as SEQ ID NO: 8, and NM — 002111.6, incorporated herein as SEQ ID NO: 10.
  • Table 3 provides SNPs found in the GM04022, GM04281, GM02171, and GM02173B cell lines. Also provided are the allelic variants found at each SNP position, the genotype for each of the cell lines, and the percentage of HD patients having a particular allelic variant.
  • the two allelic variants for SNP rs6446723 are T and C.
  • the GM04022 cell line is heterozygous TC
  • the GM02171 cell line is homozygous CC
  • the GM02173 cell line is heterozygous TC
  • the GM04281 cell line is homozygous TT.
  • Fifty percent of HD patients have a T at SNP position rs6446723.
  • the present invention provides pharmaceutical compositions comprising one or more antisense compound.
  • such 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.
  • a pharmaceutical composition consists of one or more antisense compound and sterile water.
  • the sterile saline is pharmaceutical grade water.
  • 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 phosphate-buffered saline (PBS). In certain embodiments, the sterile saline is pharmaceutical grade PBS.
  • antisense compounds may be admixed with pharmaceutically acceptable active and/or inert substances for the preparation of pharmaceutical compositions or formulations.
  • Compositions and methods for the formulation of pharmaceutical compositions depend on a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered.
  • compositions comprising antisense compounds encompass any pharmaceutically acceptable salts, esters, or salts of such esters.
  • pharmaceutical compositions comprising antisense compounds comprise one or more oligonucleotide which, upon administration to an animal, including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof.
  • the disclosure is also drawn to pharmaceutically acceptable salts of antisense compounds, prodrugs, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
  • Suitable pharmaceutically acceptable salts include, but are not limited to, sodium and potassium salts.
  • a prodrug can include the incorporation of additional nucleosides at one or both ends of an oligomeric compound which are cleaved by endogenous nucleases within the body, to form the active antisense oligomeric compound.
  • Lipid moieties have been used in nucleic acid therapies in a variety of methods.
  • the nucleic acid is introduced into preformed liposomes or lipoplexes made of mixtures of cationic lipids and neutral lipids.
  • DNA complexes with mono- or poly-cationic lipids are formed without the presence of a neutral lipid.
  • a lipid moiety is selected to increase distribution of a pharmaceutical agent to a particular cell or tissue.
  • a lipid moiety is selected to increase distribution of a pharmaceutical agent to fat tissue.
  • a lipid moiety is selected to increase distribution of a pharmaceutical agent to muscle tissue.
  • compositions provided herein comprise one or more modified oligonucleotides and one or more excipients.
  • excipients are selected from water, salt solutions, alcohol, polyethylene glycols, gelatin, lactose, amylase, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose and polyvinylpyrrolidone.
  • a pharmaceutical composition provided herein comprises a delivery system.
  • delivery systems include, but are not limited to, liposomes and emulsions. Certain delivery systems are useful for preparing certain pharmaceutical compositions including those comprising hydrophobic compounds. In certain embodiments, certain organic solvents such as dimethylsulfoxide are used.
  • a pharmaceutical composition provided herein comprises one or more tissue-specific delivery molecules designed to deliver the one or more pharmaceutical agents of the present invention to specific tissues or cell types.
  • pharmaceutical compositions include liposomes coated with a tissue-specific antibody.
  • a pharmaceutical composition provided herein comprises a co-solvent system.
  • co-solvent systems comprise, for example, benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • co-solvent systems are used for hydrophobic compounds.
  • a non-limiting example of such a co-solvent system is the VPD co-solvent system, which is a solution of absolute ethanol comprising 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 80TM and 65% w/v polyethylene glycol 300.
  • the proportions of such co-solvent systems may be varied considerably without significantly altering their solubility and toxicity characteristics.
  • co-solvent components may be varied: for example, other surfactants may be used instead of Polysorbate 80TM; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
  • a pharmaceutical composition provided herein is prepared for oral administration. In certain embodiments, pharmaceutical compositions are prepared for buccal administration.
  • a pharmaceutical composition is prepared for administration by injection (e.g., intravenous, subcutaneous, intramuscular, etc.).
  • a pharmaceutical composition comprises a carrier and is formulated in aqueous solution, such as water or physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • other ingredients are included (e.g., ingredients that aid in solubility or serve as preservatives).
  • injectable suspensions are prepared using appropriate liquid carriers, suspending agents and the like.
  • compositions for injection are suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • Certain solvents suitable for use in pharmaceutical compositions for injection include, but are not limited to, lipophilic solvents and fatty oils, such as sesame oil, synthetic fatty acid esters, such as ethyl oleate or triglycerides, and liposomes.
  • Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • such suspensions may also contain suitable stabilizers or agents that increase the solubility of the pharmaceutical agents to allow for the preparation of highly concentrated solutions.
  • a pharmaceutical composition is prepared for transmucosal administration.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • a pharmaceutical composition provided herein comprises an oligonucleotide in a therapeutically effective amount.
  • the therapeutically effective amount is sufficient to prevent, alleviate or ameliorate symptoms of a disease or to prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art.
  • one or more modified oligonucleotide provided herein is formulated as a prodrug.
  • a prodrug upon in vivo administration, is chemically converted to the biologically, pharmaceutically or therapeutically more active form of an oligonucleotide.
  • prodrugs are useful because they are easier to administer than the corresponding active form.
  • a prodrug may be more bioavailable (e.g., through oral administration) than is the corresponding active form.
  • a prodrug may have improved solubility compared to the corresponding active form.
  • prodrugs are less water soluble than the corresponding active form.
  • a prodrug is an ester.
  • the ester is metabolically hydrolyzed to carboxylic acid upon administration.
  • the carboxylic acid containing compound is the corresponding active form.
  • a prodrug comprises a short peptide (polyaminoacid) bound to an acid group.
  • the peptide is cleaved upon administration to form the corresponding active form.
  • the present invention provides compositions and methods for reducing the amount or activity of a target nucleic acid in a cell.
  • the cell is in an animal.
  • the animal is a mammal.
  • the animal is a rodent.
  • the animal is a primate.
  • the animal is a non-human primate.
  • the animal is a human.
  • the present invention provides methods of administering a pharmaceutical composition comprising an oligomeric compound of the present invention to an animal.
  • Suitable administration routes include, but are not limited to, oral, rectal, transmucosal, intestinal, enteral, topical, suppository, through inhalation, intrathecal, intracerebroventricular, intraperitoneal, intranasal, intraocular, intratumoral, and parenteral (e.g., intravenous, intramuscular, intramedullary, and subcutaneous).
  • pharmaceutical intrathecals are administered to achieve local rather than systemic exposures.
  • pharmaceutical compositions may be injected directly in the area of desired effect (e.g., into the liver).
  • 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 “AT me CGAUCG,” wherein me C indicates a cytosine base comprising a methyl group at the 5-position.
  • nucleoside phosphoramidites The preparation of nucleoside phosphoramidites is performed following procedures that are illustrated herein and in the art such as but not limited to U.S. Pat. No. 6,426,220 and published PCT WO 02/36743.
  • oligomeric compounds used in accordance with this invention may be conveniently and routinely made through the well-known technique of solid phase synthesis.
  • Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, Calif.). Any other means for such synthesis known in the art may additionally or alternatively be employed. It is well known to use similar techniques to prepare oligonucleotides such as alkylated derivatives and those having phosphorothioate linkages.
  • Oligomeric compounds Unsubstituted and substituted phosphodiester (P ⁇ O) oligomeric compounds, including without limitation, oligonucleotides can be synthesized on an automated DNA synthesizer (Applied Biosystems model 394) using standard phosphoramidite chemistry with oxidation by iodine.
  • phosphorothioate internucleoside linkages are synthesized similar to phosphodiester internucleoside linkages with the following exceptions: thiation is effected by utilizing a 10% w/v solution of 3,H-1,2-benzodithiole-3-one 1,1-dioxide in acetonitrile for the oxidation of the phosphite linkages. The thiation reaction step time is increased to 180 sec and preceded by the normal capping step. After cleavage from the CPG column and deblocking in concentrated ammonium hydroxide at 55° C.
  • the oligomeric compounds are recovered by precipitating with greater than 3 volumes of ethanol from a 1 M NH 4 OAc solution.
  • Phosphinate internucleoside linkages can be prepared as described in U.S. Pat. No. 5,508,270.
  • Alkyl phosphonate internucleoside linkages can be prepared as described in U.S. Pat. No. 4,469,863.
  • 3′-Deoxy-3′-methylene phosphonate internucleoside linkages can be prepared as described in U.S. Pat. Nos. 5,610,289 or 5,625,050.
  • Phosphoramidite internucleoside linkages can be prepared as described in U.S. Pat. No. 5,256,775 or U.S. Pat. No. 5,366,878.
  • Alkylphosphonothioate internucleoside linkages can be prepared as described in published PCT applications PCT/US94/00902 and PCT/US93/06976 (published as WO 94/17093 and WO 94/02499, respectively).
  • 3′-Deoxy-3′-amino phosphoramidate internucleoside linkages can be prepared as described in U.S. Pat. No. 5,476,925.
  • Phosphotriester internucleoside linkages can be prepared as described in U.S. Pat. No. 5,023,243.
  • Borano phosphate internucleoside linkages can be prepared as described in U.S. Pat. Nos. 5,130,302 and 5,177,198.
  • Oligomeric compounds having one or more non-phosphorus containing internucleoside linkages including without limitation methylenemethylimino linked oligonucleosides, also identified as MMI linked oligonucleosides, methylenedimethylhydrazo linked oligonucleosides, also identified as MDH linked oligonucleosides, methylenecarbonylamino linked oligonucleosides, also identified as amide-3 linked oligonucleosides, and methyleneaminocarbonyl linked oligonucleosides, also identified as amide-4 linked oligonucleosides, as well as mixed backbone oligomeric compounds having, for instance, alternating MMI and P ⁇ O or P ⁇ S linkages can be prepared as described in U.S. Pat. Nos. 5,378,825, 5,386,023, 5,489,677, 5,602,240 and 5,610,289.
  • Formacetal and thioformacetal internucleoside linkages can be prepared as described in U.S. Pat. Nos. 5,264,562 and 5,264,564.
  • Ethylene oxide internucleoside linkages can be prepared as described in U.S. Pat. No. 5,223,618.
  • the oligomeric compounds including without limitation oligonucleotides and oligonucleosides, are recovered by precipitation out of 1 M NH 4 OAc with >3 volumes of ethanol. Synthesized oligomeric compounds are analyzed by electrospray mass spectroscopy (molecular weight determination) and by capillary gel electrophoresis. The relative amounts of phosphorothioate and phosphodiester linkages obtained in the synthesis is determined by the ratio of correct molecular weight relative to the ⁇ 16 amu product (+/ ⁇ 32+/ ⁇ 48).
  • oligomeric compounds are purified by HPLC, as described by Chiang et al., J. Biol. Chem. 1991, 266, 18162-18171. Results obtained with HPLC-purified material are generally similar to those obtained with non-HPLC purified material.
  • Oligomeric compounds can be synthesized via solid phase P(III) phosphoramidite chemistry on an automated synthesizer capable of assembling 96 sequences simultaneously in a 96-well format.
  • Phosphodiester internucleoside linkages are afforded by oxidation with aqueous iodine.
  • Phosphorothioate internucleoside linkages are generated by sulfurization utilizing 3,H-1,2 benzodithiole-3-one 1,1 dioxide (Beaucage Reagent) in anhydrous acetonitrile.
  • Standard base-protected beta-cyanoethyl-diiso-propyl phosphoramidites can be purchased from commercial vendors (e.g.
  • Non-standard nucleosides are synthesized as per standard or patented methods and can be functionalized as base protected beta-cyanoethyldiisopropyl phosphoramidites.
  • Oligomeric compounds can be cleaved from support and deprotected with concentrated NH 4 OH at elevated temperature (55-60° C.) for 12-16 hours and the released product then dried in vacuo. The dried product is then re-suspended in sterile water to afford a master plate from which all analytical and test plate samples are then diluted utilizing robotic pipettors.
  • the concentration of oligomeric compounds in each well can be assessed by dilution of samples and UV absorption spectroscopy.
  • the full-length integrity of the individual products can be evaluated by capillary electrophoresis (CE) in either the 96-well format (Beckman P/ACETM MDQ) or, for individually prepared samples, on a commercial CE apparatus (e.g., Beckman P/ACETM 5000, ABI 270). Base and backbone composition is confirmed by mass analysis of the oligomeric compounds utilizing electrospray-mass spectroscopy. All assay test plates are diluted from the master plate using single and multi-channel robotic pipettors. Plates are judged to be acceptable if at least 85% of the oligomeric compounds on the plate are at least 85% full length.
  • oligomeric compounds on target nucleic acid expression is tested in any of a variety of cell types provided that the target nucleic acid is present at measurable levels. This can be routinely determined using, for example, PCR or Northern blot analysis. Cell lines derived from multiple tissues and species can be obtained from American Type Culture Collection (ATCC, Manassas, Va.).
  • the following cell type is provided for illustrative purposes, but other cell types can be routinely used, provided that the target is expressed in the cell type chosen. This can be readily determined by methods routine in the art, for example Northern blot analysis, ribonuclease protection assays or RT-PCR.
  • b.END cells The mouse brain endothelial cell line b.END was obtained from Dr. Werner Risau at the Max Plank Institute (Bad Nauheim, Germany). b.END cells are routinely cultured in DMEM, high glucose (Invitrogen Life Technologies, Carlsbad, Calif.) supplemented with 10% fetal bovine serum (Invitrogen Life Technologies, Carlsbad, Calif.). Cells are routinely passaged by trypsinization and dilution when they reached approximately 90% confluence. Cells are seeded into 96-well plates (Falcon-Primaria #353872, BD Biosciences, Bedford, Mass.) at a density of approximately 3000 cells/well for uses including but not limited to oligomeric compound transfection experiments.
  • oligomeric compounds When cells reached 65-75% confluency, they are treated with one or more oligomeric compounds.
  • the oligomeric compound is mixed with LIPOFECTINTM Invitrogen Life Technologies, Carlsbad, Calif.) in Opti-MEMTM-1 reduced serum medium (Invitrogen Life Technologies, Carlsbad, Calif.) to achieve the desired concentration of the oligomeric compound(s) and a LIPOFECTINTM concentration of 2.5 or 3 ⁇ g/mL per 100 nM oligomeric compound(s).
  • This transfection mixture is incubated at room temperature for approximately 0.5 hours. For cells grown in 96-well plates, wells are washed once with 100 ⁇ L OPTI-MEMTM-1 and then treated with 130 ⁇ L of the transfection mixture.
  • Cells grown in 24-well plates or other standard tissue culture plates are treated similarly, using appropriate volumes of medium and oligomeric compound(s). Cells are treated and data are obtained in duplicate or triplicate. After approximately 4-7 hours of treatment at 37° C., the medium containing the transfection mixture is replaced with fresh culture medium. Cells are harvested 16-24 hours after treatment with oligomeric compound(s).
  • transfection reagents known in the art include, but are not limited to, CYTOFECTINTM, LIPOFECTAMINETM, OLIGOFECTAMINETM, and FUGENETM.
  • Other suitable transfection methods known in the art include, but are not limited to, electroporation.
  • Quantitation of target mRNA levels is accomplished by real-time quantitative PCR using the ABI PRISMTM 7600, 7700, or 7900 Sequence Detection System (PE-Applied Biosystems, Foster City, Calif.) according to manufacturer's instructions.
  • ABI PRISMTM 7600, 7700, or 7900 Sequence Detection System PE-Applied Biosystems, Foster City, Calif.
  • This is a closed-tube, non-gel-based, fluorescence detection system which allows high-throughput quantitation of polymerase chain reaction (PCR) products in real-time.
  • PCR polymerase chain reaction
  • products in real-time quantitative PCR are quantitated as they accumulate. This is accomplished by including in the PCR reaction an oligonucleotide probe that anneals specifically between the forward and reverse PCR primers, and contains two fluorescent dyes.
  • a reporter dye e.g., FAM or JOE, obtained from either PE-Applied Biosystems, Foster City, Calif., Operon Technologies Inc., Alameda, Calif. or Integrated DNA Technologies Inc., Coralville, Iowa
  • a quencher dye e.g., TAMRA, obtained from either PE-Applied Biosystems, Foster City, Calif., Operon Technologies Inc., Alameda, Calif. or Integrated DNA Technologies Inc., Coralville, Iowa
  • TAMRA obtained from either PE-Applied Biosystems, Foster City, Calif., Operon Technologies Inc., Alameda, Calif. or Integrated DNA Technologies Inc., Coralville, Iowa
  • annealing of the probe to the target sequence creates a substrate that can be cleaved by the 5′-exonuclease activity of Taq polymerase.
  • cleavage of the probe by Taq polymerase releases the reporter dye from the remainder of the probe (and hence from the quencher moiety) and a sequence-specific fluorescent signal is generated.
  • additional reporter dye molecules are cleaved from their respective probes, and the fluorescence intensity is monitored at regular intervals by laser optics built into the ABI PRISMTM Sequence Detection System.
  • a series of parallel reactions containing serial dilutions of mRNA from untreated control samples generates a standard curve that is used to quantitate the percent inhibition after antisense oligonucleotide treatment of test samples.
  • primer-probe sets specific to the target gene being measured are evaluated for their ability to be “multiplexed” with a GAPDH amplification reaction.
  • multiplexing both the target gene and the internal standard gene GAPDH are amplified concurrently in a single sample.
  • mRNA isolated from untreated cells is serially diluted. Each dilution is amplified in the presence of primer-probe sets specific for GAPDH only, target gene only (“single-plexing”), or both (multiplexing).
  • standard curves of GAPDH and target mRNA signal as a function of dilution are generated from both the single-plexed and multiplexed samples.
  • the primer-probe set specific for that target is deemed multiplexable.
  • Other methods of PCR are also known in the art.
  • RT and PCR reagents are obtained from Invitrogen Life Technologies (Carlsbad, Calif.).
  • RT real-time PCR is carried out by adding 20 ⁇ L PCR cocktail (2.5 ⁇ PCR buffer minus MgCl 2 , 6.6 mM MgCl 2 , 375 ⁇ M each of dATP, dCTP, dCTP and dGTP, 375 nM each of forward primer and reverse primer, 125 nM of probe, 4 Units RNAse inhibitor, 1.25 Units PLATINUM® Taq, 5 Units MuLV reverse transcriptase, and 2.5 ⁇ ROX dye) to 96-well plates containing 30 ⁇ L total RNA solution (20-200 ng).
  • PCR cocktail 2.5 ⁇ PCR buffer minus MgCl 2 , 6.6 mM MgCl 2 , 375 ⁇ M each of dATP, dCTP, dCTP and dGTP, 375 nM each of forward primer and reverse primer, 125 n
  • the RT reaction is carried out by incubation for 30 minutes at 48° C. Following a 10 minute incubation at 95° C. to activate the PLATINUM® Taq, 40 cycles of a two-step PCR protocol are carried out: 95° C. for 15 seconds (denaturation) followed by 60° C. for 1.5 minutes (annealing/-extension).
  • Gene target quantities obtained by RT, real-time PCR are normalized using either the expression level of GAPDH, a gene whose expression is constant, or by quantifying total RNA using RIBOGREENTM (Molecular Probes, Inc. Eugene, Oreg.).
  • GAPDH expression is quantified by real time RT-PCR, by being run simultaneously with the target, multiplexing, or separately.
  • Total RNA is quantified using RiboGreenTM RNA quantification reagent (Molecular Probes, Inc. Eugene, Oreg.). Methods of RNA quantification by RIBOGREENTM are taught in Jones, L. J., et al, (Analytical Biochemistry, 1998, 265, 368-374).
  • RIBOGREENTM working reagent 170 ⁇ L, of RIBOGREENTM working reagent (RIBOGREENTM reagent diluted 1:350 in 10 mM Tris-HCl, 1 mM EDTA, pH 7.5) is pipetted into a 96-well plate containing 30 ⁇ L purified, cellular RNA.
  • the plate is read in a CytoFluor 4000 (PE Applied Biosystems) with excitation at 485 nm and emission at 530 nm.
  • Antisense modulation of a target expression can be assayed in a variety of ways known in the art.
  • a target mRNA levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or real-time PCR.
  • Real-time quantitative PCR is presently desired.
  • RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA.
  • One method of RNA analysis of the present disclosure is the use of total cellular RNA as described in other examples herein. Methods of RNA isolation are well known in the art.
  • Northern blot analysis is also routine in the art.
  • Real-time quantitative (PCR) can be conveniently accomplished using the commercially available ABI PRISMTM 7600, 7700, or 7900 Sequence Detection System, available from PE-Applied Biosystems, Foster City, Calif. and used according to manufacturer's instructions.
  • Protein levels of a target can be quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), enzyme-linked immunosorbent assay (ELISA) or fluorescence-activated cell sorting (FACS).
  • Antibodies directed to a target can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, Mich.), or can be prepared via conventional monoclonal or polyclonal antibody generation methods well known in the art. Methods for preparation of polyclonal antisera are taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology , Volume 2, pp.
  • Immunoprecipitation methods are standard in the art and can be found at, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology , Volume 2, pp. 10.16.1-10.16.11, John Wiley & Sons, Inc., 1998.
  • Western blot (immunoblot) analysis is standard in the art and can be found at, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology , Volume 2, pp. 10.8.1-10.8.21, John Wiley & Sons, Inc., 1997.
  • Enzyme-linked immunosorbent assays ELISA are standard in the art and can be found at, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology , Volume 2, pp. 11.2.1-11.2.22, John Wiley & Sons, Inc., 1991.
  • the oligomeric compounds are further investigated in one or more phenotypic assays, each having measurable endpoints predictive of efficacy in the treatment of a particular disease state or condition.
  • Phenotypic assays, kits and reagents for their use are well known to those skilled in the art and are herein used to investigate the role and/or association of a target in health and disease.
  • Representative phenotypic assays which can be purchased from any one of several commercial vendors, include those for determining cell viability, cytotoxicity, proliferation or cell survival (Molecular Probes, Eugene, Oreg.; PerkinElmer, Boston, Mass.), protein-based assays including enzymatic assays (Panvera, LLC, Madison, Wis.; BD Biosciences, Franklin Lakes, N.J.; Oncogene Research Products, San Diego, Calif.), cell regulation, signal transduction, inflammation, oxidative processes and apoptosis (Assay Designs Inc., Ann Arbor, Mich.), triglyceride accumulation (Sigma-Aldrich, St.
  • cells determined to be appropriate for a particular phenotypic assay i.e., MCF-7 cells selected for breast cancer studies; adipocytes for obesity studies
  • a target inhibitors identified from the in vitro studies as well as control compounds at optimal concentrations which are determined by the methods described above.
  • treated and untreated cells are analyzed by one or more methods specific for the assay to determine phenotypic outcomes and endpoints.
  • Phenotypic endpoints include changes in cell morphology over time or treatment dose as well as changes in levels of cellular components such as proteins, lipids, nucleic acids, hormones, saccharides or metals. Measurements of cellular status which include pH, stage of the cell cycle, intake or excretion of biological indicators by the cell, are also endpoints of interest.
  • Measurement of the expression of one or more of the genes of the cell after treatment is also used as an indicator of the efficacy or potency of the target inhibitors.
  • Hallmark genes or those genes suspected to be associated with a specific disease state, condition, or phenotype, are measured in both treated and untreated cells.
  • the individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.
  • Poly(A)+ mRNA is isolated according to Miura et al., (Clin. Chem., 1996, 42, 1758-1764). Other methods for poly(A)+ mRNA isolation are routine in the art. Briefly, for cells grown on 96-well plates, growth medium is removed from the cells and each well is washed with 200 ⁇ L cold PBS. 60 ⁇ L lysis buffer (10 mM Tris-HCl, pH 7.6, 1 mM EDTA, 0.5 M NaCl, 0.5% NP-40, 20 mM vanadyl-ribonucleoside complex) is added to each well, the plate is gently agitated and then incubated at room temperature for five minutes.
  • lysis buffer (10 mM Tris-HCl, pH 7.6, 1 mM EDTA, 0.5 M NaCl, 0.5% NP-40, 20 mM vanadyl-ribonucleoside complex
  • 554 of lysate is transferred to Oligo d(T) coated 96-well plates (AGCT Inc., Irvine Calif.). Plates are incubated for 60 minutes at room temperature, washed 3 times with 200 ⁇ L of wash buffer (10 mM Tris-HCl pH 7.6, 1 mM EDTA, 0.3 M NaCl). After the final wash, the plate is blotted on paper towels to remove excess wash buffer and then air-dried for 5 minutes. 60 ⁇ L of elution buffer (5 mM Tris-HCl pH 7.6), preheated to 70° C., is added to each well, the plate is incubated on a 90° C. hot plate for 5 minutes, and the eluate is then transferred to a fresh 96-well plate.
  • wash buffer (10 mM Tris-HCl pH 7.6, 1 mM EDTA, 0.3 M NaCl). After the final wash, the plate is blotted on paper towels to remove excess wash buffer and then air-dried for 5 minutes.
  • Cells grown on 100 mm or other standard plates may be treated similarly, using appropriate volumes of all solutions.
  • Total RNA is isolated using an RNEASY 96TM kit and buffers purchased from Qiagen Inc. (Valencia, Calif.) following the manufacturer's recommended procedures. Briefly, for cells grown on 96-well plates, growth medium is removed from the cells and each well is washed with 200 ⁇ L cold PBS. 150 ⁇ L Buffer RLT is added to each well and the plate vigorously agitated for 20 seconds. 150 ⁇ L of 70% ethanol is then added to each well and the contents mixed by pipetting three times up and down. The samples are then transferred to the RNEASY 96TM well plate attached to a QIAVACTM manifold fitted with a waste collection tray and attached to a vacuum source. Vacuum is applied for 1 minute.
  • Buffer RW1 500 ⁇ L of Buffer RW1 is added to each well of the RNEASY 96TM plate and incubated for 15 minutes and the vacuum is again applied for 1 minute.
  • An additional 500 ⁇ L of Buffer RW1 is added to each well of the RNEASY 96TM plate and the vacuum is applied for 2 minutes.
  • 1 mL of Buffer RPE is then added to each well of the RNEASY 96TM plate and the vacuum applied for a period of 90 seconds.
  • the Buffer RPE wash is then repeated and the vacuum is applied for an additional 3 minutes.
  • the plate is then removed from the QIAVACTM manifold and blotted dry on paper towels.
  • RNA is then eluted by pipetting 140 ⁇ L of RNAse free water into each well, incubating 1 minute, and then applying the vacuum for 3 minutes.
  • the repetitive pipetting and elution steps may be automated using a QIAGEN Bio-Robot 9604 (Qiagen, Inc., Valencia Calif.). Essentially, after lysing of the cells on the culture plate, the plate is transferred to the robot deck where the pipetting, DNase treatment and elution steps are carried out.
  • Probes and primers may be designed to hybridize to a target sequence, using published sequence information.
  • primer-probe set was designed using published sequence information (GENBANKTM accession number U92436.1, SEQ ID NO: 1).
  • FAM-TTGCAGCAATTCACTGTAAAGCTGGAAAGG-TAMRA (SEQ ID NO: 4), where FAM is the fluorescent dye and TAMRA is the quencher dye.
  • Western blot analysis is carried out using standard methods.
  • Cells are harvested 16-20 h after oligonucleotide treatment, washed once with PBS, suspended in Laemmli buffer (100 ⁇ l/well), boiled for 5 minutes and loaded on a 16% SDS-PAGE gel. Gels are run for 1.5 hours at 150 V, and transferred to membrane for western blotting.
  • Appropriate primary antibody directed to a target is used, with a radiolabeled or fluorescently labeled secondary antibody directed against the primary antibody species. Bands are visualized using a PHOSPHORIMAGERTM (Molecular Dynamics, Sunnyvale Calif.).
  • Bx is a heterocyclic base
  • R 1 and R 2 are each independently H, OH, or a 2′-sugar substituent group
  • Compounds 1 and 2 are commercially available from Glen Research.
  • Compounds 3 and 4 are prepared as per the procedures well known in the art as described in the specification herein (see Seth et al., Bioorg. Med. Chem., 2011, 21(4), 1122-1125 , J. Org. Chem., 2010, 75(5), 1569-1581 , Nucleic Acids Symposium Series, 2008, 52(1), 553-554); and also see published PCT International Applications (WO 2011/115818, WO 2010/077578, WO2010/036698, WO2009/143369, WO 2009/006478, and WO 2007/090071), and U.S. Pat. No. 7,569,686).
  • R 3 is —CH 2 CH 3 , —CH ⁇ CH 2 , —CH 2 CH ⁇ CH 2 , —CH 2 CH 2 OCH 3 , —CH 2 CH 2 OPG or —CH 2 CH 2 OCH 2 F;
  • PG is a protecting group
  • Bx is a heterocyclic base
  • Bx is a heterocyclic base
  • R 4 is alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, acyl or substituted acyl; wherein the substituent groups are independently selected from halogen, oxo, hydroxyl, amino, thio, azido, cyano or carboxyl;
  • Compound 15 used in the phosphitylation step serves only to illustrate the compounds described herein and is not intended to be limiting. Additional phosphitylating reagents known in the art as described in the specification herein can also be employed to generate various phosphoramidite analogs of Compound 16 and are used as building blocks for oligonucleotide synthesis.
  • Compounds 1 and 17 are commercially available from Glen Research and Chemexpress. Compounds 18 and 19 were separated by column chromatography. Although one of the methylphosphonate internucleoside linkages in DMT phosphoramidite dimer 20 or 21 is Rp and the other is Sp, the absolute stereochemistry at the phosphorus chiral center (P*) of the dimers has not been determined. The spectral analysis of Compounds 20 and 21 was consistent with the structures.
  • the UnylinkerTM 22 is commercially available.
  • Oligomeric Compound 25 comprising a modified internucleoside linkage e.g. methyl phosphonate, phosphonoacetate or phosphonoformate
  • Oligomeric Compound 25 is prepared using standard procedures in automated DNA/RNA synthesis (see Dupouy et al., Angew. Chem. Int. Ed., 2006, 45, 3623-3627).
  • Phosphoramidite building blocks, Compounds 1, 2, 3, 10 and 13 are prepared as per the procedures illustrated in Examples 13 to 15.
  • reagents and solutions used for the synthesis of oligomeric compounds are purchased from commercial sources.
  • Standard phosphoramidite building blocks and solid support are used for incorporation nucleoside residues which include for example T, A, U, G, C and m C residues.
  • a 0.1 M solution of phosphoramidite in anhydrous acetonitrile was used for ⁇ -D-2′-deoxyribonucleoside, methyl phosphonate and phosphonoacetate.
  • a 0.1 M solution in acetonitrile was used for (R)- or (S)-methyl phosphonate dimeric phosphoramidite.
  • the oligomeric compound was synthesized on VIMAD UnyLinkerTM solid support and the appropriate amounts of solid support were packed in the column for synthesis.
  • Dichloroacetic acid (3%) in DCM was used as detritylating reagent.
  • 4,5-Dicyanoimidazole in the presence of N-methyl-imidazole or 1H-tetrazole in CH 3 CN was used as activator during the coupling step.
  • the synthesis of oligomeric compounds was performed on an ABI394 synthesizer (Applied Biosystems) on a 2 ⁇ mol scale using the procedures set forth below.
  • a solid support preloaded with the UnylinkerTM was loaded into a synthesis column after closing the column bottom outlet and CH 3 CN was added to form a slurry.
  • the swelled support-bound UnylinkerTM was treated with a detritylating reagent containing 3% dichloroacetic acid in DCM to provide the free hydroxyl groups.
  • four to fourteen equivalents of phosphoramidite solutions were delivered with coupling for 6 minutes for unmodified deoxyribonucleoside phosphoramidites and 13 minutes for other modifications. All of the other steps followed standard protocols.
  • Phosphodiester linkages were introduced by oxidation with 10% t-BuOOH solution in CH 3 CN for a contact time of 10 minutes.
  • Phosphorothioate linkages were introduced by sulfurization with PADS (0.2 M) in 1:1 pyridine/CH 3 CN for a contact time of 5 minutes.
  • the cyanoethyl phosphate protecting groups were deprotected using a 1:1 (v/v) mixture of triethylamine and acetonitrile.
  • phosphonoacetate containing oligomeric compound 1.5% DBU solution in CH 3 CN was used.
  • the solid support bound oligomeric compound was washed with acetonitrile and dried under high vacuum.
  • the solid-support bound oligomeric compound was then suspended in ammonia (28-30 wt %) at room temperature for 48 h to remove nucleobase protecting groups and to cleave from the solid support.
  • the unbound oligomeric compound was then filtered and the support was rinsed and filtered with water:ethanol (1:1) followed by water. The filtrate was combined and concentrated to dryness.
  • Each internucleoside linkage is a phosphorothioate (P ⁇ S) except for the internucleoside linkage having a subscript “x”, “y”, “w”, “z” or “q”.
  • Each nucleoside followed by a subscript “x” indicates a methyl phosphonate internucleoside linkage (—P(CH 3 )( ⁇ O)—).
  • Each nucleoside followed by a subscript “y” indicates a phosphonoacetate internucleoside linkage (—P(CH 2 CO 2 ⁇ )( ⁇ O)—).
  • Each nucleoside followed by a subscript “w” indicates a methyl thiophosphonate intemucleoside linkage (—P(CH 3 )( ⁇ S)—).
  • Each nucleoside followed by a subscript “z” indicates an (R)-methyl phosphonate internucleoside linkage (—P—(R)—CH 3 )( ⁇ O)—).
  • Each nucleoside followed by a subscript “q” indicates an (S)-methyl phosphonate intemucleoside linkage (—P—(S)—CH 3 )( ⁇ O)—).
  • Each nucleoside followed by a subscript “d” is a ⁇ -D-2′-deoxyribonucleoside.
  • Each nucleoside followed by a subscript “e” indicates a 2′-O-methoxyethyl (MOE) modified nucleoside.
  • Each nucleoside followed by a subscript “k” indicates a bicyclic nucleoside having a 4′-CH((S)—CH 3 )—O-2′ bridge also referred to as a (S)-cEt modified nucleoside.
  • Each “ m C” is a 5-methyl cytosine modified nucleoside. Nucleosides followed by subscripts “e”, or “k” are further illustrated below.
  • hPBMC Human Peripheral Blood Mononuclear Cells
  • the hPBMC assay was performed using BD Vautainer CPT tube method.
  • a sample of whole blood from volunteered donors with informed consent at US HealthWorks clinic (Faraday & El Camino Real, Carlsbad) was obtained and collected in 4-15 BD Vacutainer CPT 8 ml tubes (VWR Cat.# BD362753).
  • the approximate starting total whole blood volume in the CPT tubes for each donor was recorded using the PBMC assay data sheet.
  • the blood sample was remixed immediately prior to centrifugation by gently inverting tubes 8-10 times.
  • CPT tubes were centrifuged at rt (18-25° C.) in a horizontal (swing-out) rotor for 30 min. at 1500-1800 RCF with brake off (2700 RPM Beckman Allegra 6R).
  • the cells were retrieved from the buffy coat interface (between Ficoll and polymer gel layers); transferred to a sterile 50 ml conical tube and pooled up to 5 CPT tubes/50 ml conical tube/donor.
  • the cells were then washed twice with PBS (Ca ++ , Mg ++ free; GIBCO).
  • the tubes were topped up to 50 ml and mixed by inverting several times.
  • the sample was then centrifuged at 330 ⁇ g for 15 minutes at rt (1215 RPM in Beckman Allegra 6R) and aspirated as much supernatant as possible without disturbing pellet.
  • the cell pellet was dislodged by gently swirling tube and resuspended cells in RPMI+10% FBS+pen/strep ( ⁇ 1 ml/10 ml starting whole blood volume).
  • a 60 ⁇ l sample was pipette into a sample vial (Beckman Coulter) with 600 ⁇ l VersaLyse reagent (Beckman Coulter Cat# A09777) and was gently vortexed for 10-15 sec. The sample was allowed to incubate for 10 min. at rt and being mixed again before counting.
  • the cell suspension was counted on Vicell XR cell viability analyzer (Beckman Coulter) using PBMC cell type (dilution factor of 1:11 was stored with other parameters). The live cell/ml and viability were recorded. The cell suspension was diluted to 1 ⁇ 10 7 live PBMC/ml in RPMI+10% FBS+pen/strep.
  • the cells were plated at 5 ⁇ 10 5 in 50 ⁇ l/well of 96-well tissue culture plate (Falcon Microtest). 50 ⁇ l/well of 2 ⁇ concentration oligos/controls diluted in RPMI+10% FBS+pen/strep. was added according to experiment template (100 ⁇ l/well total). Plates were placed on the shaker and allowed to mix for approx. 1 min. After being incubated for 24 hrs at 37° C.; 5% CO 2 , the plates were centrifuged at 400 ⁇ g for 10 minutes before removing the supernatant for MSD cytokine assay (i.e. human IL-6, IL-10, IL-8 and MCP-1).
  • MSD cytokine assay i.e. human IL-6, IL-10, IL-8 and MCP-1
  • a modified oligonucleotide was designed based on the 3/14/3 MOE gapmer, ISIS 353512. This modified oligonucleotide was created by having alternating methyl thiophosphonate (—P(CH 3 )( ⁇ S)—) internucleoside linkages throughout the gap region. The proinflammatory effect of the modified oligonucleotide targeting hCRP was evaluated in hPBMC assay using the protocol described in Example 20.
  • the hPBMCs were isolated from fresh, volunteered donors and were treated with modified oligonucleotides at 0, 0.0128, 0.064, 0.32, 1.6, 8, 40 and 200 ⁇ M concentrations. After a 24 hr treatment, the cytokine levels were measured.
  • the levels of IL-6 were used as the primary readout and compared to the positive control, oligonucleotide, ISIS 353512 and negative control, ISIS 104838.
  • the results from two donors denoted as “Donor 1” and “Donor 2” are presented below.
  • Each internucleoside linkage is a phosphorothioate (P ⁇ S) except for nucleosides followed by a subscript “w”.
  • Each nucleoside followed by a subscript “w” indicates a methyl thiophosphonate internucleoside linkage (—P(CH 3 )( ⁇ S)—).
  • Each nucleoside followed by a subscript “d” is ⁇ -D-2′-deoxyribonucleoside.
  • Each nucleoside followed by a subscript “e” indicates a 2′-O-methoxyethyl (MOE) modified nucleoside.
  • Each “ m C” is a 5-methyl cytosine modified nucleoside. Nucleosides followed by subscripts “e” are further illustrated below.
  • SNPs Single Nucleotide Polymorphisms (SNPs) in the Huntingtin (HTT) Gene Sequence
  • SNP positions (identified by Hayden et al, WO/2009/135322) associated with the HTT gene were mapped to the HTT genomic sequence, designated herein as SEQ ID NO: 08 (NT — 006081.18 truncated from nucleotides 1566000 to 1768000).
  • NCBI National Center for Biotechnology Information
  • the ‘Reference SNP ID number’ or ‘RS number’ is the number designated to each SNP from the Entrez SNP database at NCBI, incorporated herein by reference.
  • SNP position refers to the nucleotide position of the SNP on SEQ ID NO: 08.
  • Polymorphism indicates the nucleotide variants at that SNP position.
  • Major allele indicates the nucleotide associated with the major allele, or the nucleotide present in a statistically significant proportion of individuals in the human population.
  • ‘Minor allele’ indicates the nucleotide associated with the minor allele, or the nucleotide present in a relatively small proportion of individuals in the human population.
  • SNPs Single Nuclear Polymorphisms
  • SEQ ID NO: 08 SNP Major Minor RS No. position Polymorphism allele allele rs2857936 1963 C/T C T rs12506200 3707 A/G G A rs762855 14449 A/G G A rs3856973 19826 G/A G A rs2285086 28912 G/A A G rs7659144 37974 C/G C G rs16843804 44043 C/T C T rs2024115 44221 G/A A G rs10015979 49095 A/G A G rs7691627 51063 A/G G A rs2798235 54485 G/A G A rs4690072 62160 G/T T G rs6446723 66466 C/T T C rs363081 73280 G/A G A rs363080 73564 T/C C T rs3630
  • a series of modified oligonucleotides were designed based on ISIS 460209 wherein the gap region contains nine ⁇ -D-2′-deoxyribonucleosides.
  • the modified oligonucleotides were designed by introducing a methyl phosphonate internucleoside linkage within the gap region.
  • the oligonucleotides with modified phosphorus containing backbone were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting rs7685686 while leaving the expression of the wild-type (wt) intact.
  • the potency and selectivity of the modified oligonucleotides were evaluated and compared to ISIS 460209.
  • the position on the oligonucleotides opposite to the SNP position, as counted from the 5′-terminus is position 8.
  • Heterozygous fibroblast GM04022 cell line was used for the in vitro assay (from Coriell Institute). Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides. After a treatment period of approximately 24 hours, cells were washed with DPBS buffer and lysed. RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C — 2229297 — 10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 ⁇ L 2 ⁇ PCR buffer, 101 ⁇ L primers (300 ⁇ M from ABI), 1000 ⁇ L water and 40.4 ⁇ L RT MIX. To each well was added 15 ⁇ L of this mixture and 5 ⁇ L of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. The HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN.
  • the half maximal inhibitory concentration (IC 50 ) of each oligonucleotide is presented below and was calculated by plotting the concentrations of oligonucleotides used versus the percent inhibition of HTT mRNA expression achieved at each concentration, and noting the concentration of oligonucleotide at which 50% inhibition of HTT mRNA expression was achieved compared to the control.
  • the IC 50 at which each oligonucleotide inhibits the mutant PITT mRNA expression is denoted as ‘mut IC 50 ’.
  • the IC 50 at which each oligonucleotide inhibits the wild-type HTT mRNA expression is denoted as ‘wt IC 50 ’.
  • Selectivity as expressed in “fold” was calculated by dividing the IC 50 for inhibition of the wild-type HTT versus the IC 50 for inhibiting expression of the mutant HTT mRNA and the results are presented below.
  • Each internucleoside linkage is a phosphorothioate (P ⁇ S) except for the internucleoside linkage having a subscript “x” which indicates a methyl phosphonate internucleoside linkage (—P(CH 3 )( ⁇ O)—).
  • Each nucleoside followed by a subscript “d” is a ⁇ -D-2′-deoxyribonucleoside.
  • Each nucleoside followed by a subscript “e” indicates a 2′-O-methoxyethyl (MOE) modified nucleoside.
  • Each nucleoside followed by a subscript “k” indicates a bicyclic nucleoside having a 4′-CH((S)—CH 3 )—O-2′ bridge also referred to as a (S)-cEt modified nucleoside.
  • Each “ m C” is a 5-methyl cytosine modified nucleoside. Nucleosides followed by subscripts “e” or “k” are further illustrated below.
  • a series of modified oligonucleotides were designed based on ISIS 460209 wherein the gap region contains nine ⁇ -D-2′-deoxyribonucleosides.
  • the modified oligonucleotides were synthesized to include one or more methyl phosphonate or phosphonoacetate internucleoside linkage modifications within the gap region.
  • the oligonucleotides with modified phosphorus containing backbone were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting rs7685686 while leaving the expression of the wild-type (wt) intact.
  • the potency and selectivity of the modified oligonucleotides were evaluated and compared to ISIS 460209.
  • the position on the oligonucleotides opposite to the SNP position, as counted from the 5′-terminus is position 8.
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used (from Coriell Institute).
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.12, 0.37, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C — 2229297 — 10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 ⁇ l 2 ⁇ PCR buffer, 101 ⁇ l primers (300 ⁇ M from ABI), 1000 uL water and 40.4 ⁇ L RT MIX. To each well was added 15 ⁇ L of this mixture and 5 ⁇ L of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. The HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN.
  • Each internucleoside linkage is a phosphorothioate (P ⁇ S) except for the internucleoside linkage having a subscript “x” or “y”.
  • Each nucleoside followed by a subscript “x” indicates a methyl phosphonate internucleoside linkage (—P(CH 3 )( ⁇ O)—).
  • Each nucleoside followed by a subscript “y” indicates a phosphonoacetate internucleoside linkage (—P(CH 2 CO 2 )( ⁇ O)—).
  • Each nucleoside followed by a subscript “d” is a ⁇ -D-2′-deoxyribonucleoside.
  • Each nucleoside followed by a subscript “e” indicates a 2′-O-methoxyethyl (MOE) modified nucleoside.
  • Each nucleoside followed by a subscript “k” indicates a bicyclic nucleoside having a 4′-CH((S)—CH 3 )—O-2′ bridge also referred to as a (S)-cEt modified nucleoside.
  • Each “ m C” is a 5-methyl cytosine modified nucleoside. Nucleosides followed by subscripts “e” or “k” are further illustrated below.
  • a series of modified oligonucleotide was designed based on ISIS 460209 or ISIS 476333, wherein the gap region contains nine ⁇ -D-2′-deoxyribonucleosides.
  • the modified oligonucleotides were designed by introducing methyl phosphonate internucleoside linkage within the gap region at a fixed position and using different wing motifs for 3/9/3 and 4/9/4 gapmer motifs.
  • the modified oligonucleotides were tested for their ability to selectively inhibit mutant (mut) HTT mRNA expression levels targeting rs7685686 while leaving the expression of the wild-type (wt) intact.
  • the potency and selectivity of the modified oligonucleotides were evaluated and compared to ISIS 460209 or ISIS 476333.
  • the position on the oligonucleotides opposite to the SNP position, as counted from the 5′-terminus is position 8 for ISIS 460209 or position 9 for ISIS 476333.
  • the modified oligonucleotides were tested in vitro.
  • Heterozygous fibroblast GM04022 cell line was used (from Coriell Institute).
  • Cultured GM04022 cells at a density of 25,000 cells per well were transfected using electroporation with 0.1, 0.4, 1.1, 3.3 and 10 ⁇ M concentrations of modified oligonucleotides.
  • RNA was extracted using Qiagen RNeasy purification and mRNA levels were measured by quantitative real-time PCR using ABI assay C — 2229297 — 10 which measures at dbSNP rs362303.
  • RT-PCR method in short; A mixture was made using 2020 uL 2 ⁇ PCR buffer, 101 uL primers (300 ⁇ M from ABI), 1000 uL water and 40.4 uL RT MIX. To each well was added 15 uL of this mixture and 5 uL of purified RNA. The mutant and wild-type HTT mRNA levels were measured simultaneously by using two different fluorophores, FAM for mutant allele and VIC for wild-type allele. The HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN.
  • Each internucleoside linkage is a phosphorothioate (P ⁇ S) except for the internucleoside linkage having a subscript “x” which indicates a methyl phosphonate internucleoside linkage (—P(CH 3 )( ⁇ O)—).
  • Each nucleoside followed by a subscript “d” is a ⁇ -D-2′-deoxyribonucleoside.
  • Each nucleoside followed by a subscript “e” indicates a 2′-O-methoxyethyl (MOE) modified nucleoside.
  • Each nucleoside followed by a subscript “k” indicates a bicyclic nucleoside having a 4′-CH((S)—CH 3 )—O-2′ bridge also referred to as a (S)-cEt modified nucleoside.
  • Each “ m C” is a 5-methyl cytosine modified nucleoside. Nucleosides followed by subscripts “e” or “k” are further illustrated below.
  • modified oligonucleotides were designed based on ISIS 482050 and 449093 wherein the gap region contains ten ⁇ -D-2′-deoxyribonucleosides.
  • the modified oligonucleotides were designed by introducing two methyl phosphonate internucleoside linkages at the 5′-end of the gap region with a 3/10/3 motif.
  • the oligonucleotides were evaluated for reduction in PTEN and SRB-1 mRNA expression levels in vivo.
  • the parent gapmers, ISIS 482050 and 449093 were included in the study for comparison.
  • mice Six week old BALB/C mice (purchased from Charles River) were injected subcutaneously twice a week for three weeks at dosage 10 mg/kg or 20 mg/kg with the modified oligonucleotides shown below or with saline control. Each treatment group consisted of 3 animals. The mice were sacrificed 48 hrs following last administration, and organs and plasma were harvested for further analysis.
  • Liver tissues were homogenized and mRNA levels were quantitated using real-time PCR and normalized to RIBOGREEN as described herein. The results below are listed as PTEN or SRB-1 mRNA expression for each treatment group relative to saline-treated control (% UTC). As illustrated, reduction in PTEN or SRB-1 mRNA expression levels was achieved with the oligonucleotides comprising two methyl phosphonate internucleoside linkages at the 5′-end of the gap region, ISIS 582073 and 582074.
  • Plasma chemistry markers such as liver transaminase levels, alanine aminotranferase (ALT) in serum were measured relative to saline injected mice and the results are presented below.
  • Treatment with the oligonucleotides resulted in a reduction in ALT level compared to treatment with the parent gapmer, ISIS 482050 or 449093.
  • the results suggest that introduction of methyl phosphonate internucleoside linkage(s) can be useful for reduction of hepatotoxicity profile of otherwise unmodified parent gapmers.
  • Body weights, as well as liver, kidney and spleen weights were measured at the end of the study. The results below are presented as the average percent of body and organ weights for each treatment group relative to saline-treated control. As illustrated, treatment with ISIS 582073 resulted in a reduction in liver and spleen weights compared to treatment with the parent gapmer, ISIS 482050. The remaining oligonucleotide, ISIS 582074 did not cause any changes in body and organ weights outside the expected range as compared to ISIS 449093.
  • Each internucleoside linkage is a phosphorothioate (P ⁇ S) except for the internucleoside linkage having a subscript “x”.
  • Each nucleoside followed by a subscript “x” indicates a methyl phosphonate internucleoside linkage (—P(CH 3 )( ⁇ O)—).
  • Each nucleoside followed by a subscript “d” is a ⁇ -D-2′-deoxyribonucleoside.
  • Each nucleoside followed by a subscript “k” indicates a bicyclic nucleoside having a 4′-CH((S)—CH 3 )—O-2′ bridge also referred to as a (S)-cEt modified nucleoside.
  • Each “ m C” is a 5-methyl cytosine modified nucleoside. Nucleosides followed by subscript “k” are further illustrated below.
  • modified oligonucleotides were designed in the same manner as the antisense oligonucleotides described in Example 25, wherein two methyl phosphonate internucleoside linkages are introduced at the 5′-end of the gap region.
  • the modified oligonucleotides were designed based on ISIS 464917, 465178, 465984 and 466456 with a 3/10/3 motif.
  • the oligonucleotides were evaluated for reduction in Target-Y mRNA expression levels in vivo.
  • the parent gapmers, ISIS 464917, 465178, 465984 and 466456 were included in the study for comparison.
  • mice Six week old BALB/C mice (purchased from Charles River) were injected subcutaneously twice a week for three weeks at dosage 10 mg/kg or 20 mg/kg with the modified oligonucleotides shown below or with saline control. Each treatment group consisted of 3 animals. The mice were sacrificed 48 hrs following last administration, and organs and plasma were harvested for further analysis.
  • Target-Y mRNA expression for each treatment group relative to saline-treated control (% UTC). As illustrated, reduction in Target-Y mRNA expression levels was achieved with the oligonucleotides comprising two methyl phosphonate internucleoside linkages at the 5′-end of the gap region, ISIS 582071, 582072, 582069 and 582070.
  • Plasma chemistry markers such as liver transaminase levels, alanine aminotranferase (ALT) in serum were measured relative to saline treated mice and the results are presented below.
  • Treatment with the oligonucleotides resulted in a reduction in ALT level compared to treatment with the parent gapmer, ISIS 464917, 465178, 465984 or 466456.
  • the results suggest that introduction of methyl phosphonate internucleoside linkage(s) can be useful for reducing the hepatotoxicity profile of otherwise unmodified parent gapmers.
  • Body weights, as well as liver, kidney and spleen weights were measured at the end of the study. The results below are presented as the average percent of body and organ weights for each treatment group relative to saline-treated control. As illustrated, treatment with ISIS 582070 resulted in a reduction in liver and spleen weights compared to treatment with the parent gapmer, ISIS 466456. An increase in body and organ weights was observed for ISIS 582071 as compared to ISIS 464917. The remaining oligonucleotides, ISIS 582072 and 582069 did not cause any changes in body and organ weights outside the expected range as compared to ISIS 465178 and 465984.
  • Each internucleoside linkage is a phosphorothioate (P ⁇ S) except for the internucleoside linkage having a subscript “x”.
  • Each nucleoside followed by a subscript “x” indicates a methyl phosphonate internucleoside linkage (—P(CH 3 )( ⁇ O)—).
  • Each nucleoside followed by a subscript “d” is a ⁇ -D-2′-deoxyribonucleoside.
  • Each nucleoside followed by a subscript “e” indicates a 2′-O-methoxyethyl (MOE) modified nucleoside.
  • Each nucleoside followed by a subscript “k” indicates a bicyclic nucleoside having a 4′-CH((S)—CH 3 )—O-2′ bridge also referred to as a (S)-cEt modified nucleoside.
  • Each “N” is a modified or naturally occurring nucleobases (A, T, C, G, U, or 5-methyl C). Nucleosides followed by subscripts “e” or “k” are further illustrated below.
US14/237,967 2011-08-11 2012-08-08 Linkage modified gapped oligomeric compounds and uses thereof Abandoned US20140303235A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/237,967 US20140303235A1 (en) 2011-08-11 2012-08-08 Linkage modified gapped oligomeric compounds and uses thereof

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161522659P 2011-08-11 2011-08-11
US201261596723P 2012-02-08 2012-02-08
US201261603196P 2012-02-24 2012-02-24
US14/237,967 US20140303235A1 (en) 2011-08-11 2012-08-08 Linkage modified gapped oligomeric compounds and uses thereof
PCT/US2012/049987 WO2013022966A1 (en) 2011-08-11 2012-08-08 Linkage modified gapped oligomeric compounds and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/049987 A-371-Of-International WO2013022966A1 (en) 2011-08-11 2012-08-08 Linkage modified gapped oligomeric compounds and uses thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/407,037 Continuation US20170130224A1 (en) 2011-08-11 2017-01-16 Linkage modified gapped oligomeric compounds and uses thereof

Publications (1)

Publication Number Publication Date
US20140303235A1 true US20140303235A1 (en) 2014-10-09

Family

ID=46717938

Family Applications (9)

Application Number Title Priority Date Filing Date
US14/238,441 Active US10202599B2 (en) 2011-08-11 2012-08-08 Selective antisense compounds and uses thereof
US14/238,439 Abandoned US20150051389A1 (en) 2011-08-11 2012-08-08 Selective antisense compounds and uses thereof
US14/238,094 Active 2033-06-05 US9752142B2 (en) 2011-08-11 2012-08-08 Gapped oligomeric compounds comprising 5′-modified deoxyribonucleosides in the gap and uses thereof
US14/237,967 Abandoned US20140303235A1 (en) 2011-08-11 2012-08-08 Linkage modified gapped oligomeric compounds and uses thereof
US15/407,037 Abandoned US20170130224A1 (en) 2011-08-11 2017-01-16 Linkage modified gapped oligomeric compounds and uses thereof
US16/222,521 Abandoned US20190338281A1 (en) 2011-08-11 2018-12-17 Selective antisense compounds and uses thereof
US16/946,654 Abandoned US20210147838A1 (en) 2011-08-11 2020-06-30 Selective Antisense Compounds and Uses Thereof
US17/011,395 Active 2032-11-18 US11732261B2 (en) 2011-08-11 2020-09-03 Selective antisense compounds and uses thereof
US17/712,766 Abandoned US20230113863A1 (en) 2011-08-11 2022-04-04 Selective Antisense Compounds and Uses Thereof

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US14/238,441 Active US10202599B2 (en) 2011-08-11 2012-08-08 Selective antisense compounds and uses thereof
US14/238,439 Abandoned US20150051389A1 (en) 2011-08-11 2012-08-08 Selective antisense compounds and uses thereof
US14/238,094 Active 2033-06-05 US9752142B2 (en) 2011-08-11 2012-08-08 Gapped oligomeric compounds comprising 5′-modified deoxyribonucleosides in the gap and uses thereof

Family Applications After (5)

Application Number Title Priority Date Filing Date
US15/407,037 Abandoned US20170130224A1 (en) 2011-08-11 2017-01-16 Linkage modified gapped oligomeric compounds and uses thereof
US16/222,521 Abandoned US20190338281A1 (en) 2011-08-11 2018-12-17 Selective antisense compounds and uses thereof
US16/946,654 Abandoned US20210147838A1 (en) 2011-08-11 2020-06-30 Selective Antisense Compounds and Uses Thereof
US17/011,395 Active 2032-11-18 US11732261B2 (en) 2011-08-11 2020-09-03 Selective antisense compounds and uses thereof
US17/712,766 Abandoned US20230113863A1 (en) 2011-08-11 2022-04-04 Selective Antisense Compounds and Uses Thereof

Country Status (5)

Country Link
US (9) US10202599B2 (da)
EP (7) EP3556859B1 (da)
DK (2) DK2742135T4 (da)
ES (2) ES2651514T3 (da)
WO (4) WO2013022967A1 (da)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9840710B2 (en) 2015-11-18 2017-12-12 Rosalind Franklin University Of Medicine And Science Antisense compounds targeting leucine-rich repeat kinase 2 (LRRK2) for the treatment of parkinsons disease
US10370667B2 (en) 2015-11-18 2019-08-06 Rosalind Franklin University Of Medicine And Science Antisense compounds targeting leucine-rich repeat kinase 2 (LRRK2) for the treatment of parkinsons disease
US10479995B2 (en) 2015-07-22 2019-11-19 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
WO2019245957A1 (en) * 2018-06-18 2019-12-26 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds
US10724035B2 (en) 2016-05-04 2020-07-28 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
CN111868244A (zh) * 2018-03-20 2020-10-30 国立大学法人东京工业大学 降低了毒性的反义寡核苷酸
CN113490742A (zh) * 2019-02-20 2021-10-08 罗氏创新中心哥本哈根有限公司 膦酰基乙酸酯缺口聚物寡核苷酸
US11149264B2 (en) 2018-02-12 2021-10-19 Ionis Pharmaceuticals, Inc. Modified compounds and uses thereof
US11236335B2 (en) 2012-10-12 2022-02-01 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US11261440B2 (en) 2017-02-21 2022-03-01 Osaka University Antisense oligonucleic acid
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
US11732261B2 (en) 2011-08-11 2023-08-22 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof

Families Citing this family (124)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2377327T5 (es) 2006-10-18 2020-04-28 Ionis Pharmaceuticals Inc Compuestos antisentido
JP5645840B2 (ja) 2008-12-02 2014-12-24 株式会社Wave Life Sciences Japan リン原子修飾核酸の合成方法
MX342945B (es) 2009-07-06 2016-10-18 Ontorii Inc * Profármacos de ácido nucleico novedosos y métodos de uso de los mismos.
WO2012039448A1 (ja) 2010-09-24 2012-03-29 株式会社キラルジェン 不斉補助基
EP3067421B1 (en) 2011-02-08 2018-10-10 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
DK2734208T3 (da) 2011-07-19 2017-06-19 Wave Life Sciences Ltd Fremgangsmåder til syntese af funktionaliserede nukleinsyrer
US9914922B2 (en) * 2012-04-20 2018-03-13 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
CN104684923B (zh) 2012-07-13 2018-09-28 株式会社新日本科学 手性核酸佐剂
DK2872147T3 (da) 2012-07-13 2023-02-20 Wave Life Sciences Ltd Fremgangsmåde til fremstilling af kirale oligonukleotider
ES2862073T3 (es) 2012-07-13 2021-10-06 Wave Life Sciences Ltd Grupo auxiliar asimétrico
WO2014059353A2 (en) 2012-10-11 2014-04-17 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleosides and uses thereof
WO2014059341A2 (en) * 2012-10-12 2014-04-17 Isis Pharmaceuticals, Inc. Antisense compounds and uses thereof
EP2951304B1 (en) 2013-02-04 2020-07-08 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US10590412B2 (en) 2013-04-19 2020-03-17 Ionis Pharmaceuticals, Inc. Compositions and methods for modulation nucleic acids through nonsense mediated decay
CN112263682A (zh) * 2013-06-27 2021-01-26 罗氏创新中心哥本哈根有限公司 靶向pcsk9的反义寡聚体和缀合物
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
WO2015108048A1 (ja) 2014-01-15 2015-07-23 株式会社新日本科学 抗腫瘍作用を有するキラル核酸アジュバンド及び抗腫瘍剤
AU2015207773B2 (en) 2014-01-16 2021-06-17 Wave Life Sciences Ltd. Chiral design
EP3137476B1 (en) * 2014-04-28 2019-10-09 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds
WO2016061487A1 (en) * 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
EP3261643B1 (en) 2015-02-26 2020-11-25 Ionis Pharmaceuticals, Inc. Allele specific modulators of p23h rhodopsin
WO2017100726A1 (en) 2015-12-10 2017-06-15 Ptc Therapeutics, Inc. Methods for treatng huntington's disease
JPWO2017111137A1 (ja) * 2015-12-22 2018-10-18 味の素株式会社 オリゴヌクレオチドの製造方法
CN107282130B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 乙烯四聚催化剂组合物及其应用
CN107282120B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯齐聚用催化剂组合物及齐聚方法
CN107282122B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯四聚催化剂组合物及其应用
CN107282132B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯四聚催化剂组合物及应用
CN107282121B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯齐聚用催化剂组合物和齐聚方法
CN107282123B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯齐聚催化剂组合物及其应用
CN107282124B (zh) * 2016-03-31 2019-12-24 中国石油化工股份有限公司 一种乙烯四聚催化剂组合物及四聚方法
MA45496A (fr) 2016-06-17 2019-04-24 Hoffmann La Roche Molécules d'acide nucléique pour la réduction de l'arnm de padd5 ou pad7 pour le traitement d'une infection par l'hépatite b
US11638717B2 (en) 2017-03-29 2023-05-02 Shionogi & Co., Ltd. Complex of nucleic acid medicine and multibranched lipid
EP3634953B1 (en) 2017-06-05 2024-01-03 PTC Therapeutics, Inc. Compounds for treating huntington's disease
CN111182898B (zh) 2017-06-28 2024-04-16 Ptc医疗公司 用于治疗亨廷顿氏病的方法
CA3067592A1 (en) 2017-06-28 2019-01-03 Ptc Therapeutics, Inc. Methods for treating huntington's disease
WO2019006455A1 (en) 2017-06-30 2019-01-03 Solstice Biologics, Ltd. AUXILIARIES OF CHIRAL PHOSPHORAMIDITIS AND METHODS OF USE THEREOF
JP2021502059A (ja) 2017-10-13 2021-01-28 ロシュ イノベーション センター コペンハーゲン エーエス 部分的に立体定義されたオリゴヌクレオチドのサブライブラリーを使用することによる、アンチセンスオリゴヌクレオチドの、改良された立体定義されたホスホロチオエートオリゴヌクレオチド変異体を同定するための方法
IL272566B1 (en) 2017-10-16 2024-03-01 Hoffmann La Roche Nucleic acid molecule to reduce papd5 and papd7 mRNA to treat hepatitis b infection
WO2019077614A1 (en) 2017-10-22 2019-04-25 Lumus Ltd. ENHANCED REALITY DEVICE MOUNTED ON THE HEAD AND USING AN OPTICAL BENCH
WO2019115416A2 (en) 2017-12-11 2019-06-20 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating fndc3b expression
WO2019115417A2 (en) 2017-12-12 2019-06-20 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating rb1 expression
MX2020005872A (es) 2017-12-21 2020-08-13 Hoffmann La Roche Diagnostico complementario para antagonistas de acido ribonucleico (arn) de proteina a1 de alto requerimiento de temperatura a (htra1).
EP4092118A1 (en) 2017-12-22 2022-11-23 Roche Innovation Center Copenhagen A/S Novel thiophosphoramidites
JP7476102B2 (ja) 2017-12-22 2024-04-30 ロシュ イノベーション センター コペンハーゲン エーエス ホスホロジチオアートヌクレオシド間結合を含むオリゴヌクレオチド
AU2018386527A1 (en) 2017-12-22 2020-04-30 Roche Innovation Center Copenhagen A/S Gapmer oligonucleotides comprising a phosphorodithioate internucleoside linkage
CN111699258A (zh) 2018-01-10 2020-09-22 哥本哈根罗氏创新中心 用于调控pias4表达的寡核苷酸
JP2021510295A (ja) 2018-01-12 2021-04-22 ロシュ イノベーション センター コペンハーゲン エーエス Gsk3b発現を調節するためのオリゴヌクレオチド
US20210095276A1 (en) 2018-01-17 2021-04-01 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating erc1 expression
CN111655851A (zh) 2018-01-18 2020-09-11 哥本哈根罗氏创新中心 靶向srebp1的反义寡核苷酸
WO2019145386A1 (en) 2018-01-26 2019-08-01 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating csnk1d expression
JP7281474B2 (ja) 2018-02-09 2023-05-25 ジェネンテック, インコーポレイテッド Tmem106b発現を調節するためのオリゴヌクレオチド
AU2019243048A1 (en) 2018-03-27 2020-10-15 Ptc Therapeutics, Inc. Compounds for treating Huntington's disease
KR20200140853A (ko) 2018-04-05 2020-12-16 에프. 호프만-라 로슈 아게 B형 간염 바이러스 감염을 치료하기 위한 fubp1 억제제의 용도
US20210071247A1 (en) 2018-05-07 2021-03-11 Roche Innovation Center Copenhagen A/S Massively parallel discovery methods for oligonucleotide therapeutics
US20210371860A1 (en) 2018-05-08 2021-12-02 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating myh7 expression
CN112912500A (zh) 2018-06-05 2021-06-04 豪夫迈·罗氏有限公司 用于调节atxn2表达的寡核苷酸
WO2020005873A1 (en) 2018-06-27 2020-01-02 Ptc Therapeutics, Inc. Heterocyclic and heteroaryl compounds for treating huntington's disease
WO2020007700A1 (en) 2018-07-02 2020-01-09 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting spi1
WO2020007772A1 (en) 2018-07-02 2020-01-09 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting gbp-1
WO2020007702A1 (en) 2018-07-02 2020-01-09 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting bcl2l11
EP3818159A1 (en) 2018-07-03 2021-05-12 F. Hoffmann-La Roche AG Oligonucleotides for modulating tau expression
WO2020007826A1 (en) 2018-07-05 2020-01-09 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting mbtps1
WO2020007889A1 (en) 2018-07-05 2020-01-09 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting stat1
WO2020011653A1 (en) 2018-07-09 2020-01-16 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting kynu
WO2020011743A1 (en) 2018-07-09 2020-01-16 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting mafb
WO2020011869A2 (en) 2018-07-11 2020-01-16 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting tlr2
WO2020011744A2 (en) 2018-07-11 2020-01-16 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting cers5
WO2020011745A2 (en) 2018-07-11 2020-01-16 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting cers6
MX2021000404A (es) 2018-07-13 2021-03-25 Hoffmann La Roche Oligonucleotidos para modular la expresion de rtel1.
CN112236439A (zh) 2018-07-31 2021-01-15 罗氏创新中心哥本哈根有限公司 包含三硫代磷酸酯核苷间键的寡核苷酸
TW202020155A (zh) 2018-07-31 2020-06-01 丹麥商羅氏創新中心哥本哈根有限公司 含有三硫代磷酸酯核苷間連結之寡核苷酸
WO2020038976A1 (en) 2018-08-23 2020-02-27 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting usp8
WO2020038973A1 (en) 2018-08-23 2020-02-27 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting sptlc1
WO2020038971A1 (en) 2018-08-23 2020-02-27 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting vcan
EP3620519A1 (en) 2018-09-04 2020-03-11 F. Hoffmann-La Roche AG Use of isolated milk extracellular vesicles for delivering oligonucleotides orally
JP2022512877A (ja) 2018-11-01 2022-02-07 エフ.ホフマン-ラ ロシュ アーゲー Tia1を標的とするアンチセンスオリゴヌクレオチド
WO2020109344A1 (en) 2018-11-29 2020-06-04 F. Hoffmann-La Roche Ag Occular administration device for antisense oligonucleotides
WO2020109343A1 (en) 2018-11-29 2020-06-04 F. Hoffmann-La Roche Ag Combination therapy for treatment of macular degeneration
EP3898975A2 (en) 2018-12-21 2021-10-27 Boehringer Ingelheim International GmbH Antisense oligonucleotides targeting card9
EP3914232A1 (en) 2019-01-25 2021-12-01 F. Hoffmann-La Roche AG Lipid vesicle for oral drug delivery
CA3127483A1 (en) * 2019-01-25 2020-07-30 Nayan Therapeutics, Inc. Nr2e3 expression reducing oligonucleotides, compositions containing the same, and methods of their use
EP3914713A1 (en) * 2019-01-25 2021-12-01 Nayan Therapeutics, Inc. Nrl expression reducing oligonucleotides, compositions containing the same, and methods of their use
CA3126845A1 (en) * 2019-02-01 2020-08-06 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
CA3129646A1 (en) 2019-02-20 2020-08-27 Roche Innovation Center Copenhagen A/S Novel phosphoramidites
CN113474633A (zh) 2019-02-26 2021-10-01 罗氏创新中心哥本哈根有限公司 寡核苷酸配制方法
US11286485B2 (en) 2019-04-04 2022-03-29 Hoffmann-La Roche Inc. Oligonucleotides for modulating ATXN2 expression
SG11202111387YA (en) * 2019-04-25 2021-11-29 Wave Life Sciences Ltd Oligonucleotide compositions and methods of use thereof
EP3980539A1 (en) 2019-06-06 2022-04-13 F. Hoffmann-La Roche AG Antisense oligonucleotides targeting atxn3
WO2021021673A1 (en) * 2019-07-26 2021-02-04 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating gfap
WO2021030778A1 (en) 2019-08-15 2021-02-18 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds and uses thereof
AU2020377204A1 (en) 2019-11-01 2022-06-02 Novartis Ag The use of a splicing modulator for a treatment slowing progression of huntington's disease
WO2021122735A1 (en) 2019-12-19 2021-06-24 F. Hoffmann-La Roche Ag Use of sept9 inhibitors for treating hepatitis b virus infection
JP2023506540A (ja) 2019-12-19 2023-02-16 エフ. ホフマン-ラ ロシュ エージー. B型肝炎ウイルス感染を処置するためのscamp3阻害剤の使用
EP4077671A1 (en) 2019-12-19 2022-10-26 F. Hoffmann-La Roche AG Use of saraf inhibitors for treating hepatitis b virus infection
WO2021122910A1 (en) 2019-12-19 2021-06-24 F. Hoffmann-La Roche Ag Use of sbds inhibitors for treating hepatitis b virus infection
JP2023506547A (ja) 2019-12-19 2023-02-16 エフ. ホフマン-ラ ロシュ エージー. B型肝炎ウイルス感染を処置するためのcops3阻害剤の使用
CN114829603A (zh) 2019-12-20 2022-07-29 豪夫迈·罗氏有限公司 用于抑制scn9a表达的增强寡核苷酸
TW202137987A (zh) 2019-12-24 2021-10-16 瑞士商赫孚孟拉羅股份公司 用於治療hbv之靶向hbv的治療性寡核苷酸及tlr7促效劑之醫藥組合
CN114828852A (zh) 2019-12-24 2022-07-29 豪夫迈·罗氏有限公司 用于治疗hbv的靶向hbv的抗病毒药剂和/或免疫调节剂的药物组合
WO2021152005A1 (en) 2020-01-28 2021-08-05 Universite De Strasbourg Antisense oligonucleotide targeting linc00518 for treating melanoma
WO2021170697A1 (en) 2020-02-28 2021-09-02 F. Hoffmann-La Roche Ag Oligonucleotides for modulating cd73 exon 7 splicing
WO2021231210A1 (en) 2020-05-11 2021-11-18 Genentech, Inc. Complement component c1r inhibitors for treating a neurological disease, and related compositions, systems and methods of using same
JP2023527684A (ja) 2020-05-11 2023-06-30 ジェネンテック, インコーポレイテッド 神経疾患を治療するための補体成分c1s阻害剤、並びにそれを使用する関連組成物、システム及び方法
CN115698290A (zh) 2020-05-11 2023-02-03 基因泰克公司 用于治疗神经系统疾病的补体组分4抑制剂以及使用它们的相关组合物、系统和方法
US20230193269A1 (en) 2020-05-22 2023-06-22 Boehringer Ingelheim International Gmbh Oligonucleotides for splice modulation of card9
CN115702243A (zh) 2020-06-09 2023-02-14 罗氏创新中心哥本哈根有限公司 用于治疗性多核苷酸的鸟苷类似物
AR122731A1 (es) 2020-06-26 2022-10-05 Hoffmann La Roche Oligonucleótidos mejorados para modular la expresión de fubp1
WO2022018155A1 (en) 2020-07-23 2022-01-27 F. Hoffmann-La Roche Ag Lna oligonucleotides for splice modulation of stmn2
JP2023538630A (ja) 2020-08-21 2023-09-08 エフ. ホフマン-ラ ロシュ アーゲー B型肝炎ウイルス感染症を処置するためのa1cf阻害剤の使用
US11447521B2 (en) 2020-11-18 2022-09-20 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
AR124229A1 (es) 2020-12-03 2023-03-01 Hoffmann La Roche Oligonucleótidos antisentido que actúan sobre atxn3
TW202237843A (zh) 2020-12-03 2022-10-01 瑞士商赫孚孟拉羅股份公司 靶向atxn3之反義寡核苷酸
WO2022122613A1 (en) 2020-12-08 2022-06-16 F. Hoffmann-La Roche Ag Novel synthesis of phosphorodithioate oligonucleotides
EP4284513A1 (en) 2021-01-26 2023-12-06 Universite Brest Bretagne Occidentale Novel stim1 splicing variants and uses thereof
TW202246500A (zh) 2021-02-02 2022-12-01 瑞士商赫孚孟拉羅股份公司 用於抑制 rtel1 表現之增強型寡核苷酸
TW202304446A (zh) 2021-03-29 2023-02-01 瑞士商諾華公司 剪接調節子用於減慢杭丁頓氏舞蹈症進展的治療之用途
WO2023052317A1 (en) 2021-09-29 2023-04-06 F. Hoffmann-La Roche Ag Rna editing
WO2023078883A1 (en) 2021-11-03 2023-05-11 F. Hoffmann-La Roche Ag Oligonucleotides for modulating apolipoprotein e4 expression
CA3234478A1 (en) 2021-11-11 2023-05-19 Souphalone LUANGSAY Pharmaceutical combinations for treatment of hbv
WO2023111210A1 (en) 2021-12-17 2023-06-22 F. Hoffmann-La Roche Ag Combination of oligonucleotides for modulating rtel1 and fubp1
WO2023117738A1 (en) 2021-12-20 2023-06-29 F. Hoffmann-La Roche Ag Threose nucleic acid antisense oligonucleotides and methods thereof
WO2023141507A1 (en) 2022-01-20 2023-07-27 Genentech, Inc. Antisense oligonucleotides for modulating tmem106b expression
EP4332221A1 (en) 2022-08-29 2024-03-06 Roche Innovation Center Copenhagen A/S Threose nucleic acid antisense oligonucleotides and methods thereof

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995013834A1 (en) * 1993-11-16 1995-05-26 Genta, Incorporated Chimeric oligonucleoside compounds
US20050142535A1 (en) * 2002-02-01 2005-06-30 Damha Masad J. Ogligonucleotides comprising alternating segments and uses thereof
US20110112170A1 (en) * 2008-04-04 2011-05-12 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleosides and having reduced toxicity
US20110130441A1 (en) * 2008-04-04 2011-06-02 Isis Pharmaceuticals, Inc. Oligomeric compounds having at least one neutrally linked terminal bicyclic nucleosides

Family Cites Families (196)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2699808A (en) 1944-10-06 1955-01-18 Mark W Lowe Apparatus for peeling tomatoes
US2699508A (en) 1951-12-21 1955-01-11 Selectronics Inc Method of mounting and construction of mounting for low frequency piezoelectric crystals
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
JPS5717316A (en) 1980-07-04 1982-01-29 Kawasaki Steel Corp Method for automatic control of screw down of reeler mill
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
FR2567892B1 (fr) 1984-07-19 1989-02-17 Centre Nat Rech Scient Nouveaux oligonucleotides, leur procede de preparation et leurs applications comme mediateurs dans le developpement des effets des interferons
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
FR2575751B1 (fr) 1985-01-08 1987-04-03 Pasteur Institut Nouveaux nucleosides de derives de l'adenosine, leur preparation et leurs applications biologiques
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
ATE113059T1 (de) 1987-06-24 1994-11-15 Florey Howard Inst Nukleosid-derivate.
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5256775A (en) 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
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
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5149797A (en) 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
GB9009980D0 (en) 1990-05-03 1990-06-27 Amersham Int Plc Phosphoramidite derivatives,their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
DE69032425T2 (de) 1990-05-11 1998-11-26 Microprobe Corp Teststreifen zum Eintauchen für Nukleinsäure-Hybridisierungsassays und Verfahren zur kovalenten Immobilisierung von Oligonucleotiden
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
CA2088258C (en) 1990-07-27 2004-09-14 Phillip Dan Cook Nuclease resistant, pyrimidine modified oligonucleotides that detect and modulate gene expression
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
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
US6582908B2 (en) 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
US5965722A (en) 1991-05-21 1999-10-12 Isis Pharmaceuticals, Inc. Antisense inhibition of ras gene with chimeric and alternating oligonucleotides
EP0538194B1 (de) 1991-10-17 1997-06-04 Novartis AG Bicyclische Nukleoside, Oligonukleotide, Verfahren zu deren Herstellung und Zwischenprodukte
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
EP1256589A3 (en) 1991-11-26 2003-09-17 Isis Pharmaceuticals, Inc. Oligomers containing modified pyrimidines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
TW393513B (en) 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
US20010044145A1 (en) 1991-12-24 2001-11-22 Monia Brett P. Methods of using mammalian RNase H and compositions thereof
US5700922A (en) 1991-12-24 1997-12-23 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
FR2687679B1 (fr) 1992-02-05 1994-10-28 Centre Nat Rech Scient Oligothionucleotides.
FR2692265B1 (fr) 1992-05-25 1996-11-08 Centre Nat Rech Scient Composes biologiquement actifs de type phosphotriesters.
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
EP0577558A2 (de) 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclische Nukleoside mit bicyclischen Ringen, Oligonukleotide daraus, Verfahren zu deren Herstellung, deren Verwendung und Zwischenproduckte
WO1994002499A1 (en) 1992-07-27 1994-02-03 Hybridon, Inc. Oligonucleotide alkylphosphonothioates
WO1994014226A1 (en) 1992-12-14 1994-06-23 Honeywell Inc. Motor system with individually controlled redundant windings
ATE138384T1 (de) 1993-01-25 1996-06-15 Hybridon Inc Olionukleotidalkylphosphonate und - phosphonothioate
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
US5446786A (en) 1993-03-03 1995-08-29 Northern Telecom Limited Two-wire telecommunications line detection arrangements
GB9304620D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Compounds
EP0691968B1 (en) 1993-03-30 1997-07-16 Sanofi Acyclic nucleoside analogs and oligonucleotide sequences containing them
EP0691979A1 (en) 1993-03-31 1996-01-17 Sanofi Novel 5'-substituted nucleosides and oligomers produced therefrom
DE4311944A1 (de) 1993-04-10 1994-10-13 Degussa Umhüllte Natriumpercarbonatpartikel, Verfahren zu deren Herstellung und sie enthaltende Wasch-, Reinigungs- und Bleichmittelzusammensetzungen
FR2705099B1 (fr) 1993-05-12 1995-08-04 Centre Nat Rech Scient Oligonucléotides phosphorothioates triesters et procédé de préparation.
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5801154A (en) 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
IL111659A0 (en) * 1993-11-16 1995-01-24 Genta Inc Synthetic oligomers having chirally pure phosphonate internucleosidyl linkages mixed with non-phosphonate internucleosidyl linkages
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
US5446137B1 (en) 1993-12-09 1998-10-06 Behringwerke Ag Oligonucleotides containing 4'-substituted nucleotides
US5595756A (en) 1993-12-22 1997-01-21 Inex Pharmaceuticals Corporation Liposomal compositions for enhanced retention of bioactive agents
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5627053A (en) 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5646269A (en) 1994-04-28 1997-07-08 Gilead Sciences, Inc. Method for oligonucleotide analog synthesis
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
US5597909A (en) 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5792747A (en) 1995-01-24 1998-08-11 The Administrators Of The Tulane Educational Fund Highly potent agonists of growth hormone releasing hormone
US6624293B1 (en) * 1995-08-17 2003-09-23 Hybridon, Inc. Modified protein kinase A-specific oligonucleotides and methods of their use
US20050054600A1 (en) * 1995-08-17 2005-03-10 Hybridon, Inc. Modified protein kinase a-specific oligonucleotides and methods of their use
US6331617B1 (en) * 1996-03-21 2001-12-18 University Of Iowa Research Foundation Positively charged oligonucleotides as regulators of gene expression
US5656408A (en) 1996-04-29 1997-08-12 Xerox Corporation Coated carrier particles
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
WO2005121371A2 (en) 2004-06-03 2005-12-22 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
AU4966997A (en) 1996-11-18 1998-06-10 Takeshi Imanishi Novel nucleotide analogues
JP3756313B2 (ja) 1997-03-07 2006-03-15 武 今西 新規ビシクロヌクレオシド及びオリゴヌクレオチド類縁体
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
CA2303299C (en) 1997-09-12 2016-02-23 Exiqon A/S Oligonucleotide analogues
US20030228597A1 (en) 1998-04-13 2003-12-11 Cowsert Lex M. Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
US6043352A (en) 1998-08-07 2000-03-28 Isis Pharmaceuticals, Inc. 2'-O-Dimethylaminoethyloxyethyl-modified oligonucleotides
NZ513402A (en) 1999-02-12 2003-06-30 Sankyo Co Novel nucleosides and oligonucleotide analogues
US6753422B2 (en) 1999-03-01 2004-06-22 O'brien Thomas G. Odc allelic analysis method for assessing carcinogenic susceptibility
CA2363077A1 (en) 1999-03-01 2000-09-08 Variagenics, Inc. Methods for targeting rna molecules
US7084125B2 (en) 1999-03-18 2006-08-01 Exiqon A/S Xylo-LNA analogues
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
US5998148A (en) 1999-04-08 1999-12-07 Isis Pharmaceuticals Inc. Antisense modulation of microtubule-associated protein 4 expression
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
JP4151751B2 (ja) 1999-07-22 2008-09-17 第一三共株式会社 新規ビシクロヌクレオシド類縁体
US6147200A (en) 1999-08-19 2000-11-14 Isis Pharmaceuticals, Inc. 2'-O-acetamido modified monomers and oligomers
WO2001079283A1 (en) 2000-04-13 2001-10-25 University Of British Columbia Modulating cell survival by modulating huntingtin function
US7135565B2 (en) 2000-07-28 2006-11-14 Agilent Technologies, Inc. Synthesis of polynucleotides using combined oxidation/deprotection chemistry
WO2002027033A1 (en) 2000-09-29 2002-04-04 Isis Pharmaceuticals, Inc. Antisense modulation of mekk4 expression
US6693187B1 (en) 2000-10-17 2004-02-17 Lievre Cornu Llc Phosphinoamidite carboxlates and analogs thereof in the synthesis of oligonucleotides having reduced internucleotide charge
US6426220B1 (en) 2000-10-30 2002-07-30 Isis Pharmaceuticals, Inc. Antisense modulation of calreticulin expression
US20030125241A1 (en) 2001-05-18 2003-07-03 Margit Wissenbach Therapeutic uses of LNA-modified oligonucleotides in infectious diseases
US20050191638A1 (en) 2002-02-20 2005-09-01 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
US20030158403A1 (en) 2001-07-03 2003-08-21 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
EP1499627A2 (en) 2001-07-03 2005-01-26 ISIS Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
US7888324B2 (en) 2001-08-01 2011-02-15 Genzyme Corporation Antisense modulation of apolipoprotein B expression
WO2003013437A2 (en) 2001-08-07 2003-02-20 University Of Delaware Compositions and methods for the prevention and treatment of huntington's disease
US20040096880A1 (en) 2001-08-07 2004-05-20 Kmiec Eric B. Compositions and methods for the treatment of diseases exhibiting protein misassembly and aggregation
DE60337040D1 (de) 2002-02-01 2011-06-16 Life Technologies Corp Oligonukleotid-Zusammensetzungen mit verbesserter Wirksamkeit
US20050096284A1 (en) 2002-02-20 2005-05-05 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
CA2480311C (en) 2002-04-05 2015-01-27 Santaris Pharma A/S Oligomeric compounds for the modulation of hif-1alpha expression
US7569575B2 (en) 2002-05-08 2009-08-04 Santaris Pharma A/S Synthesis of locked nucleic acid derivatives
US20040092465A1 (en) 2002-11-11 2004-05-13 Isis Pharmaceuticals Inc. Modulation of huntingtin interacting protein 1 expression
US20040102398A1 (en) 2002-11-23 2004-05-27 Isis Pharmaceuticals Inc. Modulation of B7H expression
US20080274989A1 (en) 2002-08-05 2008-11-06 University Of Iowa Research Foundation Rna Interference Suppression of Neurodegenerative Diseases and Methods of Use Thereof
US20050255086A1 (en) 2002-08-05 2005-11-17 Davidson Beverly L Nucleic acid silencing of Huntington's Disease gene
US20050042646A1 (en) 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
US20050106731A1 (en) 2002-08-05 2005-05-19 Davidson Beverly L. siRNA-mediated gene silencing with viral vectors
WO2004027030A2 (en) 2002-09-18 2004-04-01 Isis Pharmaceuticals, Inc. Efficient reduction of target rna’s by single- and double-stranded oligomeric compounds
US20040219565A1 (en) 2002-10-21 2004-11-04 Sakari Kauppinen Oligonucleotides useful for detecting and analyzing nucleic acids of interest
WO2004044139A2 (en) 2002-11-05 2004-05-27 Isis Parmaceuticals, Inc. Modified oligonucleotides for use in rna interference
US7696345B2 (en) 2002-11-05 2010-04-13 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
WO2004044181A2 (en) 2002-11-13 2004-05-27 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein b expression
EP2305813A3 (en) 2002-11-14 2012-03-28 Dharmacon, Inc. Fuctional and hyperfunctional sirna
US7687617B2 (en) 2002-11-18 2010-03-30 Santaris Pharma A/S Oligonucleotides with alternating segments of locked and non-locked nucleotides
MXPA05008319A (es) 2003-02-10 2006-02-28 Santaris Pharma As Compuestos oligomericos para la modulacion de la expresion de survivina.
CA2526893C (en) 2003-05-14 2010-10-26 Japan Science And Technology Agency Inhibition of the expression of huntingtin gene
WO2004106356A1 (en) 2003-05-27 2004-12-09 Syddansk Universitet Functionalized nucleotide derivatives
JP4731324B2 (ja) 2003-08-28 2011-07-20 武 今西 N−o結合性架橋構造型新規人工核酸
US20050053981A1 (en) 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
US20050074801A1 (en) 2003-09-09 2005-04-07 Monia Brett P. Chimeric oligomeric compounds comprising alternating regions of northern and southern conformational geometry
US7947658B2 (en) 2003-09-12 2011-05-24 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
JP5379347B2 (ja) 2003-09-18 2013-12-25 アイシス ファーマシューティカルズ, インコーポレーテッド 4’−チオヌクレオシドおよびオリゴマー化合物
TW200519202A (en) 2003-09-18 2005-06-16 Lilly Co Eli Modulation of eIF4E expression
WO2005045032A2 (en) 2003-10-20 2005-05-19 Sima Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF EARLY GROWTH RESPONSE GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
BRPI0418104A (pt) 2003-12-23 2007-04-17 Santaris Pharma As compostos oligoméricos para a modulação de bcl-2
US20050176045A1 (en) 2004-02-06 2005-08-11 Dharmacon, Inc. SNP discriminatory siRNA
GB0407382D0 (en) 2004-03-31 2004-05-05 Univ Cambridge Tech Therapeutic methods and means
WO2005105995A2 (en) 2004-04-14 2005-11-10 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED TREATMENT OF POLYGLUTAMINE (POLYQ) REPEAT EXPANSION DISEASES USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2005116204A1 (ja) 2004-05-11 2005-12-08 Rnai Co., Ltd. Rna干渉を生じさせるポリヌクレオチド、および、これを用いた遺伝子発現抑制方法
US7323308B2 (en) 2004-09-03 2008-01-29 Affymetrix, Inc. Methods of genetic analysis of E. coli
US7919472B2 (en) 2004-09-17 2011-04-05 Isis Pharmaceuticals, Inc. Enhanced antisense oligonucleotides
US20070161590A1 (en) 2005-06-28 2007-07-12 Medtronic, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin
CA2620856C (en) 2005-08-29 2017-11-28 Isis Pharmaceuticals, Inc. Methods for use in modulating mir-122a
EP2338992A3 (en) * 2005-08-29 2011-10-12 Regulus Therapeutics, Inc Antisense compounds having enhanced anti-microRNA activity
US7320965B2 (en) 2005-10-28 2008-01-22 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Huntingtin gene
JP5425474B2 (ja) 2006-01-26 2014-02-26 アイシス ファーマシューティカルズ, インコーポレーテッド ハンチンチン対する、組成物及びその使用
JP5342881B2 (ja) 2006-01-27 2013-11-13 アイシス ファーマシューティカルズ, インコーポレーテッド 6−修飾された二環式核酸類似体
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
US8935416B2 (en) 2006-04-21 2015-01-13 Fortinet, Inc. Method, apparatus, signals and medium for enforcing compliance with a policy on a client computer
DK2015758T3 (da) 2006-05-05 2014-06-23 Isis Pharmaceuticals Inc Forbindelser og fremgangsmåder til modulering af ekspression af apob
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
US7547684B2 (en) 2006-05-11 2009-06-16 Isis Pharmaceuticals, Inc. 5′-modified bicyclic nucleic acid analogs
US9273356B2 (en) 2006-05-24 2016-03-01 Medtronic, Inc. Methods and kits for linking polymorphic sequences to expanded repeat mutations
CA2662704A1 (en) 2006-07-07 2008-01-10 University Of Massachusetts Rna silencing compositions and methods for the treatment of huntington's disease
JP4798093B2 (ja) 2006-08-04 2011-10-19 日産自動車株式会社 流体改質装置及びこれを用いた流体改質方法
PT2049664E (pt) 2006-08-11 2012-01-03 Prosensa Technologies Bv Oligonucleotídeos de cadeia simples complementares dos elementos repetitivos para tratar perturbações genéticas associadas à instabilidade das repetições do adn
ES2377327T5 (es) * 2006-10-18 2020-04-28 Ionis Pharmaceuticals Inc Compuestos antisentido
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
TW200838551A (en) 2006-11-27 2008-10-01 Isis Pharmaceuticals Inc Methods for treating hypercholesterolemia
EP2125852B1 (en) 2007-02-15 2016-04-06 Ionis Pharmaceuticals, Inc. 5'-substituted-2'-f modified nucleosides and oligomeric compounds prepared therefrom
US8278425B2 (en) 2007-05-30 2012-10-02 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
EP2014769B1 (en) 2007-06-18 2010-03-31 Commissariat à l'Energie Atomique Reversible siRNAa-based silencing of mutated and endogenous wild-type huntingtin gene and its application for the treatment of Huntington's disease
AU2008272918B2 (en) 2007-07-05 2012-09-13 Isis Pharmaceuticals, Inc. 6-disubstituted bicyclic nucleic acid analogs
US8088904B2 (en) 2007-08-15 2012-01-03 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
BRPI0820391A2 (pt) 2007-11-09 2015-05-19 Isis Pharmaceutical Inc Composto, composição e métodos de modulação da expressão do fator 7
US8546556B2 (en) 2007-11-21 2013-10-01 Isis Pharmaceuticals, Inc Carbocyclic alpha-L-bicyclic nucleic acid analogs
EP2265627A2 (en) 2008-02-07 2010-12-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexitol nucleic acid analogs
WO2009135322A1 (en) 2008-05-09 2009-11-12 The Universtity Of British Columbia Methods and compositions for the treatment of huntington's disease
US8679750B2 (en) 2008-05-09 2014-03-25 The University Of British Columbia Methods and compositions for the treatment of Huntington'S disease
WO2009143369A2 (en) 2008-05-22 2009-11-26 Isis Pharmaceuticals, Inc. Method of preparing nucleosides and analogs thereof without using chromatography
DK2356129T3 (da) 2008-09-24 2013-05-13 Isis Pharmaceuticals Inc Substituerede alpha-L-bicykliske nukleosider
WO2010048585A2 (en) 2008-10-24 2010-04-29 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
AT507215B1 (de) 2009-01-14 2010-03-15 Boehler Edelstahl Gmbh & Co Kg Verschleissbeständiger werkstoff
WO2011017521A2 (en) 2009-08-06 2011-02-10 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
US9574191B2 (en) 2010-02-03 2017-02-21 The Board Of Regents Of The University Of Texas System Selective inhibition of polyglutamine protein expression
AU2011213562B2 (en) * 2010-02-08 2016-07-07 Ionis Pharmaceuticals, Inc. Selective reduction of allelic variants
CA2789038A1 (en) * 2010-02-08 2011-08-11 Isis Pharmaceuticals, Inc. Selective reduction of allelic variants
US9193752B2 (en) 2010-03-17 2015-11-24 Isis Pharmaceuticals, Inc. 5′-substituted bicyclic nucleosides and oligomeric compounds prepared therefrom
JP6005628B2 (ja) 2010-04-28 2016-10-12 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. 修飾ヌクレオシド、その類似体、およびこれらから調製されるオリゴマー化合物
EP3067421B1 (en) * 2011-02-08 2018-10-10 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
WO2012141960A1 (en) 2011-04-11 2012-10-18 Boston Scientific Neuromodulation Corporation Systems and methods for enhancing paddle lead placement
JP6042871B2 (ja) 2011-04-21 2016-12-14 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. B型肝炎ウイルス(hbv)発現の調節
DK2742135T4 (da) * 2011-08-11 2020-07-13 Ionis Pharmaceuticals Inc Bindingsmodificerede gapped oligomeriske forbindelser og anvendelser deraf
US9778706B2 (en) 2012-02-24 2017-10-03 Blackberry Limited Peekable user interface on a portable electronic device
US9914922B2 (en) 2012-04-20 2018-03-13 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
US9400902B2 (en) 2012-05-22 2016-07-26 Trimble Navigation Limited Multi-modal entity tracking and display
US9984408B1 (en) 2012-05-30 2018-05-29 Amazon Technologies, Inc. Method, medium, and system for live video cooperative shopping
WO2014059353A2 (en) 2012-10-11 2014-04-17 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleosides and uses thereof
DK2906256T3 (da) 2012-10-12 2018-11-19 Ionis Pharmaceuticals Inc Selektive antisense-forbindelser og anvendelser deraf
EP2951304B1 (en) 2013-02-04 2020-07-08 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US9778708B1 (en) 2016-07-18 2017-10-03 Lenovo Enterprise Solutions (Singapore) Pte. Ltd. Dual sided latching retainer for computer modules
AU2019218987A1 (en) * 2018-02-12 2020-07-23 Ionis Pharmaceuticals, Inc. Modified compounds and uses thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995013834A1 (en) * 1993-11-16 1995-05-26 Genta, Incorporated Chimeric oligonucleoside compounds
US20050142535A1 (en) * 2002-02-01 2005-06-30 Damha Masad J. Ogligonucleotides comprising alternating segments and uses thereof
US20110112170A1 (en) * 2008-04-04 2011-05-12 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleosides and having reduced toxicity
US20110130441A1 (en) * 2008-04-04 2011-06-02 Isis Pharmaceuticals, Inc. Oligomeric compounds having at least one neutrally linked terminal bicyclic nucleosides

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Agrawal et al., Site-specific excision from RNA by RNase H and mixed-phosphate-backbone oligodeoxynucleotides, 1990, PNAS, volume 87, pages 1401-1405. *
Clark et al., Clinical use of streptolysin-O to facilitate antisense oligodeoxyribonucleotide delivery for purging autografts in chronic myeloid leukaemia, 1999, Bone Marrow Transplantation, volume 23, pages 1303-1308. *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11732261B2 (en) 2011-08-11 2023-08-22 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US11236335B2 (en) 2012-10-12 2022-02-01 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US10479995B2 (en) 2015-07-22 2019-11-19 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
US11634710B2 (en) 2015-07-22 2023-04-25 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
US10370667B2 (en) 2015-11-18 2019-08-06 Rosalind Franklin University Of Medicine And Science Antisense compounds targeting leucine-rich repeat kinase 2 (LRRK2) for the treatment of parkinsons disease
US10787669B2 (en) 2015-11-18 2020-09-29 Rosalind Franklin University Of Medicine And Science Antisense compounds targeting Leucine-Rich repeat kinase 2(LRRK2) for the treatment of Parkinsons disease
US9840710B2 (en) 2015-11-18 2017-12-12 Rosalind Franklin University Of Medicine And Science Antisense compounds targeting leucine-rich repeat kinase 2 (LRRK2) for the treatment of parkinsons disease
US10724035B2 (en) 2016-05-04 2020-07-28 Wave Life Sciences Ltd. Oligonucleotide compositions and methods thereof
EP4035659A1 (en) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes for delivery of therapeutic agents
US11261440B2 (en) 2017-02-21 2022-03-01 Osaka University Antisense oligonucleic acid
US11149264B2 (en) 2018-02-12 2021-10-19 Ionis Pharmaceuticals, Inc. Modified compounds and uses thereof
US11332733B2 (en) * 2018-02-12 2022-05-17 lonis Pharmaceuticals, Inc. Modified compounds and uses thereof
US20230068063A1 (en) * 2018-02-12 2023-03-02 Ionis Pharmaceuticals, Inc. Modified Compounds and Uses Thereof
CN111868244A (zh) * 2018-03-20 2020-10-30 国立大学法人东京工业大学 降低了毒性的反义寡核苷酸
WO2019245957A1 (en) * 2018-06-18 2019-12-26 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds
CN113490742A (zh) * 2019-02-20 2021-10-08 罗氏创新中心哥本哈根有限公司 膦酰基乙酸酯缺口聚物寡核苷酸

Also Published As

Publication number Publication date
EP2742136B1 (en) 2017-09-27
US20210238591A1 (en) 2021-08-05
EP2742056B1 (en) 2017-01-11
EP3205725B1 (en) 2019-03-27
US20170130224A1 (en) 2017-05-11
EP3556859B1 (en) 2021-04-07
US9752142B2 (en) 2017-09-05
DK2742136T3 (da) 2017-11-20
EP2742056A1 (en) 2014-06-18
US20210147838A1 (en) 2021-05-20
US20140309279A1 (en) 2014-10-16
EP3205725A1 (en) 2017-08-16
US20140323707A1 (en) 2014-10-30
US20150051389A1 (en) 2015-02-19
EP2742135B2 (en) 2020-06-10
EP2742135B1 (en) 2017-05-03
WO2013022984A1 (en) 2013-02-14
EP2742056B2 (en) 2020-06-10
EP4269584A2 (en) 2023-11-01
US20230113863A1 (en) 2023-04-13
ES2635866T3 (es) 2017-10-05
US10202599B2 (en) 2019-02-12
US20190338281A1 (en) 2019-11-07
EP2742135A1 (en) 2014-06-18
US11732261B2 (en) 2023-08-22
EP3556859A1 (en) 2019-10-23
EP3922722B1 (en) 2023-06-28
WO2013022967A1 (en) 2013-02-14
ES2651514T3 (es) 2018-01-26
DK2742135T4 (da) 2020-07-13
EP2742056A4 (en) 2015-03-25
DK2742135T3 (da) 2017-07-10
ES2635866T5 (es) 2021-04-05
EP2742136A1 (en) 2014-06-18
WO2013022966A1 (en) 2013-02-14
EP4269584A3 (en) 2024-03-27
WO2013022990A1 (en) 2013-02-14
EP3922722A1 (en) 2021-12-15

Similar Documents

Publication Publication Date Title
EP2742135B2 (en) Linkage modified gapped oligomeric compounds and uses thereof
US10344275B2 (en) Linkage modified oligomeric compounds
US11236335B2 (en) Selective antisense compounds and uses thereof
US10260069B2 (en) Selective antisense compounds and uses thereof
EP3453761A1 (en) Oligomer-conjugate complexes and their use
US10745756B2 (en) Methods of predicting toxicity
US11180756B2 (en) Morpholino modified oligomeric compounds
US20210254059A1 (en) Linkage modified oligomeric compounds

Legal Events

Date Code Title Description
AS Assignment

Owner name: IONIS PHARMACEUTICALS, INC., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:ISIS PHARMACEUTICALS, INC.;REEL/FRAME:037882/0108

Effective date: 20151218

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION