WO2016061263A1 - Composés antisens et leurs utilisations - Google Patents

Composés antisens et leurs utilisations Download PDF

Info

Publication number
WO2016061263A1
WO2016061263A1 PCT/US2015/055582 US2015055582W WO2016061263A1 WO 2016061263 A1 WO2016061263 A1 WO 2016061263A1 US 2015055582 W US2015055582 W US 2015055582W WO 2016061263 A1 WO2016061263 A1 WO 2016061263A1
Authority
WO
WIPO (PCT)
Prior art keywords
modified
compound
sugar moiety
nucleosides
oligonucleotide
Prior art date
Application number
PCT/US2015/055582
Other languages
English (en)
Inventor
Frank Rigo
Adrian R. Krainer
Original Assignee
Ionis Pharmaceuticals, Inc.
Cold Spring Harbor Laboratory
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ionis Pharmaceuticals, Inc., Cold Spring Harbor Laboratory filed Critical Ionis Pharmaceuticals, Inc.
Publication of WO2016061263A1 publication Critical patent/WO2016061263A1/fr

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
    • C12N15/1137Non-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 against enzymes
    • 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
    • C12N2310/315Phosphorothioates
    • 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
    • C12N2310/33Chemical 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
    • C12N2310/346Spatial arrangement of the modifications having a combination of backbone and sugar modifications
    • 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
    • C12N2320/31Combination therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/33Alteration of splicing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/01Phosphotransferases with an alcohol group as acceptor (2.7.1)
    • C12Y207/0104Pyruvate kinase (2.7.1.40)

Definitions

  • the pyruvate kinase M (PK-M) gene has 12 exons. Exon 9 and exon 10 are alternatively spliced in a mutually exclusive fashion to give rise to the Ml and M2 isoforms of the PK-M gene. Inclusion of exon 9 and exclusion of exon 10 yields the PK-Ml isoform. Exclusion of exon 9 and inclusion of exon 10 yields the PK-M2 isoform. Exons 9 and 10 each encode a 56 amino acid segment that confers distinctive properties to the respective PK-Ml and PK-M2 isoforms. The PK-M2 isoform is expressed in a broad range of cancer cells, whereas PK-Ml is predominantly expressed in terminally differentiated tissues.
  • 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
  • the present invention provides compounds comprising oligonucleotides.
  • such oligonucleotides are complementary to a pyruvate kinase M (PK-M) transcript.
  • PK-M pyruvate kinase M
  • oligonucleotides are complementary to a target region of the PK-M transcript comprising intron 9 or the junction between intron 9 and exon 10.
  • oligonucleotides are complementary to a target region of the PK-M transcript comprising an intron adjacent to exon 10, for example, intron 9.
  • oligonucleotides are complementary to a target region of the PK-M transcript comprising intron 9.
  • the PK-M transcript comprises an intronic splice silencer for exon 10. In certain embodiments, the PK-M transcript comprises an intronic splice enhancer for exon 9. In certain embodiments, oligonucleotides inhibit inclusion of exon 10. In certain embodiments, oligonucleotides promote skipping of exon 10. In certain embodiments, oligonucleotides promote selection of exon 9. In certain embodiments, oligonucleotides promote skipping of exon 10 and promote inclusion of exon 9. In certain such embodiments, PK-M mRNA with exon 9 mRNA is increased. In certain such embodiments, PK-M mRNA with exon 10 mRNA is decreased. In certain embodiments, the PK-M2 isoform of the PK-M protein is decreased. In certain embodiments, the PK-M1 isoform of the PK-M protein is decreased.
  • contacting a cell with an oligonucleotide described herein results in a decrease in PK-M mRNA with exon 10. In certain embodiments, contacting a cell with an oligonucleotide described herein results in a decrease in the expression of the PK-M2 isoform. In certain such embodiments, contacting a cell with an oligonucleotide described herein results in an increase in the PK-M1 isoform of the PK-M protein and a decrease in the PK-M2 isoform of the PK-M protein.
  • contacting a cell with an oligonucleotide complementary to a target region of the PK-M transcript comprising intron 9 and contacting a cell with a second oligonucleotide complementary to a target region of the PK-M transcript comprising exon 10 results in a decrease in the expression of the PK-M2 isoform
  • PK-M transcript is in a human. In certain embodiments, including, but not limited to any of the below numbered embodiments, the PK-M transcript is in a mouse.
  • splicing of PK-M pre-mRNA is altered by contacting a cell with more than one modified oligonucleotide. In certain embodiments, splicing of PK-M pre- mRNA is altered by contacting a cell with a modified oligonucleotide that targets exon 10 and a second modified oligonucleotide that targets intron 9. For example, in certain embodiments, splicing of PK-M pre-mRNA is altered by contacting a cell both ISIS 549197 and ISIS 461378.
  • splicing of PK-M pre-mRNA is altered by contacting a cell both ISIS 549197 and a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 12 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84
  • splicing of PK-M pre-mRNA is altered by contacting a cell both ISIS 549197 and a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 12 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, and 113.
  • Embodiment 1 A compound comprising a modified oligonucleotide consisting of 8 to 80 linked nucleosides complementary within nucleobases 28726-28743, 28727-28743, 28728-28743, 28729-28743, 28729-
  • Embodiment 2 A compound comprising a modified oligonucleotide consisting of 8 to 80 linked nucleosides complementary within nucleobases 28726-28743, 28727-28743, 28728-28743, 28729-28743, 28729- 28746, 28729-28749, 28729-28753, 28730-28744, 28730-28747, 28730-28754, 28731-28745, 28731- 28748, 28731-28755, 28732-28746, 28732-28749, 28732-28756, 28735-28749, 28738-28752, 28741- 28755, 28744-28758, 28747-28761, 28750-28764, 28753-28767, 28758-28772, 28768-28782, or 28773- 28787 of SEQ ID NO: 1 , wherein said modified oligonucleotide is at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 1.
  • Embodiment 3 A compound comprising a modified oligonucleotide having a nucleobase sequence consisting of any one of SEQ ID NO: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78,
  • Embodiment 4 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 8 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61 , 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 119, 120, 121 , 122, 123,
  • Embodiment 5 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 9 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61 , 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, 85, 119, 120, 121 , 122, 123,
  • Embodiment 6 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 10 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,14, 15, 16, 17, 18, 19, 20,
  • Embodiment 7 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 11 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76,
  • Embodiment 8 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 12 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 119, 120, 121, 122, 123, 124, 125, 126,
  • Embodiment 9 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising the nucleobase sequence of any one of SEQ ID NOs: 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131
  • Embodiment 10 A compound comprising a modified oligonucleotide consisting of 8 to 30 linked nucleosides and having a nucleobase sequence comprising a complementary region, wherein the complementary region comprises at least 8 contiguous nucleobases and is complementary to an equal- length portion of a target region of a P -M transcript.
  • Embodiment 11 The compound of embodiment 10, wherein the target region of the PK-M transcript comprises at least a portion of intron 9 of the PK-M transcript.
  • Embodiment 12 The compound of any of embodiments 10 to 11, wherein the target region of the PK-
  • M transcript comprises at least a portion of exon 10 of the PK-M transcript.
  • Embodiment 13 The compound of embodiment 11 or 12, wherein the complementary region of the modified oligonucleotide is 100% complementary to the target region.
  • Embodiment 14 The compound of any of embodiments 11 to 13, wherein the complementary region of the modified oligonucleotide comprises at least 10 contiguous nucleobases.
  • Embodiment 15 The compound of any of embodiments 11 to 13, wherein the complementary region of the modified oligonucleotide comprises at least 15 contiguous nucleobases.
  • Embodiment 16 The compound of any of embodiments 11 to 13, wherein the complementary region of the modified oligonucleotide comprises at least 18 contiguous nucleobases.
  • Embodiment 17 The compound of any of embodiments 11-16, wherein the nucleobase sequence of the oligonucleotide is at least 80% complementary to an equal-length region of the PK-M transcript, as measured over the entire length of the oligonucleotide.
  • Embodiment 18 The compound of any of embodiments 11-16, wherein the nucleobase sequence of the oligonucleotide is at least 90% complementary to an equal-length region of the PK-M transcript, as measured over the entire length of the oligonucleotide.
  • Embodiment 19 The compound of any of embodiments 11-16, wherein the nucleobase sequence of the oligonucleotide is 100% complementary to an equal-length region of the PK-M transcript, as measured over the entire length of the oligonucleotide.
  • Embodiment 20 The compound of any of embodiments 11-19, wherein the target region is within intron 9 of the PK-M transcript.
  • Embodiment 21 The compound of any of embodiments 1-10, wherein the target region is within nucleobase 28726-28743, 28727-28743, 28728-28743, 28729-28743, 28729-28746, 28729-28749, 28729- 28753, 28730-28744, 28730- 28747 28730- 28754, 28731-28745, 28731-28748, 28731-28755, 28732-28746, 28732-28749, 28732-28756 28735-28749, 28738-28752, 28741-28755, 28744-28758, 28747-28761, 28750-28764, 28753-28767 28758-28772, 28768-28782, 28773-28787 28726-28743, 28727-28743, 28728-28743, 28729-28743 28746, 28729-28749, 28729-28753, 28730-28744,
  • Embodiment 22 The compound of any of embodiments 1-10, wherein the target region is within nucleobase 28726-28743, 28727-28743, 28728-28743, 28729-28743, 28729-28746, 28729-28749, 28729-28753, 28730-28744, 28730-28747, 28730-28754, 28731-28745, 28731-28748, 28731-28755, 28732-28746, 28732-28749, 28732-28756, 28735-28749, 28738-28752, 28741-28755, 28744-28758, 28747-28761, 28750-28764, 28753-28767, 28758-28772, 28768-28782, or 28773-28787 of SEQ ID NO. 1.
  • Embodiment 23 The compound of any of embodiments 1-23, wherein the modified oligonucleotide comprises at least one modified nucleoside.
  • Embodiment 24 The compound of embodiment 23, wherein at least one modified nucleoside comprises a modified sugar moiety.
  • Embodiment 25 The compound of embodiment 24, wherein at least one modified sugar moiety is a 2'-substituted sugar moiety.
  • Embodiment 26 The compound of embodiment 25, wherein the 2'-substitutent of at least one 2'- substituted sugar moiety is selected from among: 2'-OMe, 2'-F, and 2'-MOE.
  • Embodiment 27 The compound of any of embodiments 25-26, wherein the 2'-substiuent of at least one 2 '-substituted sugar moiety is a 2'-MOE.
  • Embodiment 28 The compound of any of embodiments 1-23, wherein at least one modified sugar moiety is a bicyclic sugar moiety.
  • Embodiment 29 The compound of embodiment 28, wherein at least one bicyclic sugar moiety is cEt.
  • Embodiment 30 The compound of embodiment 28, wherein at least one bicyclic sugar moiety is
  • Embodiment 31 The compound of embodiment 25, wherein at least one sugar moiety is a sugar surrogate.
  • Embodiment 32 The compound of embodiment 31, wherein at least one sugar surrogate is a
  • Embodiment 33 The compound of embodiment 31, wherein at least one sugar surrogate is a modified morpholino.
  • Embodiment 34 The compound of any of embodiment 1-33, wherein the modified oligonucleotide comprises at least 5 modified nucleosides, each independently comprising a modified sugar moiety.
  • Embodiment 35 The compound of any of embodiment 1-33, wherein the modified oligonucleotide comprises at least 10 modified nucleosides, each independently comprising a modified sugar moiety.
  • Embodiment 36 The compound of any of embodiment 1-33, wherein the modified oligonucleotide comprises at least 15 modified nucleosides, each independently comprising a modified sugar moiety.
  • Embodiment 37 The compound of any of embodiment 1-33, wherein each nucleoside of the modified oligonucleotide is a modified nucleoside, each independently comprising a modified sugar moiety
  • Embodiment 38 The compound of any of embodiment 1-33, wherein the modified oligonucleotide comprises at least two modified nucleosides comprising modified sugar moieties that are the same as one another.
  • Embodiment 39 The compound of any of embodiments 1-33, wherein the modified oligonucleotide comprises at least two modified nucleosides comprising modified sugar moieties that are different from one another.
  • Embodiment 40 The compound of any of embodiments 1-39, wherein the modified oligonucleotide comprises a modified region of at least 5 contiguous modified nucleosides.
  • Embodiment 41 The compound of any of embodiments 1-39, wherein the modified oligonucleotide comprises a modified region of at least 10 contiguous modified nucleosides.
  • Embodiment 42 The compound of any of embodiments 1-39, wherein the modified oligonucleotide comprises a modified region of at least 15 contiguous modified nucleosides.
  • Embodiment 43 The compound of any of embodiments 1-39, wherein the modified oligonucleotide comprises a modified region of at least 18 contiguous modified nucleosides.
  • Embodiment 44 The compound of any of embodiments37-43, wherein each modified nucleoside of the modified region has a modified sugar moiety independently selected from among: 2'-F, 2'-OMe, 2'- MOE, cEt, LNA, morpholino, and modified morpholino.
  • Embodiment 45 The compound of any of embodiments 40-43, wherein the modified nucleosides of the modified region each comprise the same modification as one another.
  • Embodiment 46 The compound of any of embodiments 40-43, wherein the modified nucleosides of the modified region each comprise the same 2'-substituted sugar moiety.
  • Embodiment 47 The compound of embodiment 46, wherein the 2'-substituted sugar moiety of the modified nucleosides of the region of modified nucleosides is selected from 2'-F, 2'-OMe, and 2'-MOE.
  • Embodiment 48 The compound of embodiment 46, wherein the 2'-substituted sugar moiety of the modified nucleosides of the region of modified nucleosides is 2'-MOE.
  • Embodiment 49 The compound of embodiment 44, wherein the modified nucleosides of the region of modified nucleosides each comprise the same bicyclic sugar moiety.
  • Embodiment 50 The compound of embodiment 49, wherein the bicyclic sugar moiety of the modified nucleosides of the region of modified nucleosides is selected from LNA and cEt.
  • Embodiment 51 The compound of embodiment 44, wherein the sugar surrogate of the modified nucleosides of the region of modified nucleosides is a morpholino.
  • Embodiment 52 The compound of embodiment 44, wherein the sugar surrogate of the modified nucleosides of the region of modified nucleosides is a modified morpholino.
  • Embodiment 53 The compound of any of embodiments 1-35, wherein the modified oligonucleotide comprises no more than 4 contiguous naturally occurring nucleosides.
  • Embodiment 54 The compound of any of embodiments 1-52, wherein each nucleoside of the
  • modified oligonucleotide is a modified nucleoside.
  • Embodiment 55 The compound of embodiment 54 wherein each modified nucleoside comprises a modified sugar moiety.
  • Embodiment 56 The compound of embodiment 55, wherein the modified nucleosides of the modified oligonucleotide comprise the same modification as one another.
  • Embodiment 57 The compound of embodiment 56, wherein the modified nucleosides of the modified oligonucleotide each comprise the same 2'-substituted sugar moiety.
  • Embodiment 58 The compound of embodiment 57, wherein the 2'-substituted sugar moiety of the modified oligonucleotide is selected from 2'-F, 2'-OMe, and 2'-MOE.
  • Embodiment 59 The compound of embodiment 58, wherein the 2'-substituted sugar moiety of the modified oligonucleotide is 2'-MOE.
  • Embodiment 60 The compound of embodiment 56, wherein the modified nucleosides of the modified oligonucleotide each comprise the same bicyclic sugar moiety.
  • Embodiment 61 The compound of embodiment 60, wherein the bicyclic sugar moiety of the modified oligonucleotide is selected from LNA and cEt.
  • Embodiment 62 The compound of embodiment 55, wherein the modified nucleosides of the modified oligonucleotide each comprises a sugar surrogate.
  • Embodiment 63 The compound of embodiment 62, wherein the sugar surrogate of the modified oligonucleotide is a morpholino.
  • Embodiment 64 The compound of embodiment 62, wherein the sugar surrogate of the modified oligonucleotide is a modified morpholino.
  • Embodiment 65 The compound of any of embodiments 1-34 wherein each nucleobase of the
  • modified oligonucleotide is a modified sugar moiety or an unmodified nucleobase.
  • Embodiment 66 The compound of embodiment 65, wherein each unmodified nucleobase is DNA.
  • Embodiment 67 The compound of embodiment 65 or 66, wherein each modified sugar moiety is selected from cEt, LNA, 2'-F, 2'-OMe, and 2'-MOE.
  • Embodiment 68 The compound of embodiment 67, wherein the modified sugar moiety is cEt.
  • Embodiment 69 The compound of embodiment 67, wherein the modified sugar moiety is LNA.
  • Embodiment 70 The compound of embodiment 67, wherein the modified sugar moiety is 2'F.
  • Embodiment 71 The compound of embodiment 67, wherein the modified sugar moiety is 2'-OMe.
  • Embodiment 72 The compound of embodiment 67, wherein the modified sugar moiety is 2'-MOE.
  • Embodiment 73 The compound of any of embodiments 65 to 72, wherein the modified
  • oligonucleotide has a aaddaddaddaddaa motif, wherein each "a” represents a modified sugar moiety and wherein each "d” represents DNA.
  • Embodiment 74 The compound of any of embodiments 1-73, wherein the modified oligonucleotide comprises at least one modified internucleoside linkage.
  • Embodiment 75 The compound of embodiment 74, wherein each internucleoside linkage is a
  • Embodiment 76 The compound of embodiment 74 or 75, comprising at least one phosphorothioate internucleoside linkage.
  • Embodiment 77 The compound of embodiment 75, wherein each internucleoside linkage is a
  • Embodiment 78 The compound of embodiment 77, wherein each internucleoside linkage is a
  • Embodiment 79 The compound of any of embodiments 1-74, wherein each internucleoside linkage is either a phosphodiester internucleoside linkage or a phosphorothioate internucleoside linkage.
  • Embodiment 80 The compound of any of embodiments 1-79 comprising at least one conjugate.
  • Embodiment 81 The compound of any of embodiments 1 -79 consisting of the modified
  • Embodiment 82 The compound of any of embodiments 1-81, wherein the compound modulates splicing of the PK-M transcript.
  • Embodiment 83 The compound of any of embodiments 10-82, having a nucleobase sequence
  • Embodiment 84 A pharmaceutical composition comprising a compound according to any of
  • embodiments 1-83 and a pharmaceutically acceptable carrier or diluent.
  • Embodiment 85 The pharmaceutical composition of embodiment 84, wherein the pharmaceutically acceptable carrier or diluent is sterile saline.
  • Embodiment 86 A method of modulating splicing of a P -M transcript in a cell comprising
  • Embodiment 87 The method of embodiment 86, wherein the cell is in vitro.
  • Embodiment 88 The method of embodiment 86, wherein the cell is in an animal.
  • Embodiment 89 The method of any of embodiments 86-88, wherein inclusion of exon 9 is increased.
  • Embodiment 90 The method of any of embodiments 86-89, wherein exclusion of exon 10 is
  • Embodiment 91 The method of any of embodiments 86-89, wherein inclusion of exon 10 is
  • Embodiment 92 The method of any of embodiments 86-91, wherein PK-M1 mR A expression is increased.
  • Embodiment 93 The method of any of embodiments 86-92, wherein PK-M2 mRNA expression is decreased.
  • Embodiment 94 A method of modulating the expression of PK-M in a cell, comprising contacting the cell with a compound according to any of embodiments 1-85.
  • Embodiment 95 The method of embodiment 94, wherein PK-M1 expression is increased.
  • Embodiment 96 The method of embodiments 94 or 95, wherein PK-M2 expression is decreased.
  • Embodiment 97 The method of embodiment 94, wherein the cell is in vitro.
  • Embodiment 98 The method of embodiment 94, wherein the cell is in an animal.
  • Embodiment 99 A method of inducing apoptosis in a cell, comprising contacting the cell with a compound according to any of embodiments 1-85.
  • Embodiment 100 The method of embodiment 99, wherein the cell is a glial cell.
  • Embodiment 101 The method of embodiment 99 or 100, wherein the cell is in vitro.
  • Embodiment 102 The method of embodiment 99 or 100, wherein the cell is in an animal.
  • Embodiment 103 A method comprising administering the compound according to any of embodiments 1-83 or the pharmaceutical composition of embodiments 84 or 85 to an animal.
  • Embodiment 104 The method of embodiment 103, wherein the administration is
  • Embodiment 105 The method of embodiment 103, wherein the administration is into the central nervous sysem.
  • Embodiment 106 The method of any of embodiments 103-105, wherein the animal has one or more symptoms associated with cancer.
  • Embodiment 107 The method embodiment 106, wherein the cancer is glioblastoma.
  • Embodiment 108 The method of embodiment 107, wherein the administration results in amelioration of at least one symptom of cancer.
  • Embodiment 109 The method of any of embodiments 101-108, wherein the animal is a mouse.
  • Embodiment 110 The method of any of embodiments 101-108, wherein the animal is a human.
  • Embodiment 111 A method of preventing or retarding the growth of a cancerous tumor, comprising administering the compound according to any of embodiments 1-83 or the pharmaceutical composition of embodiments 84 or 85 to an animal in need thereof.
  • Embodiment 112 The method of embodiment 111, wherein the animal is a mouse.
  • Embodiment 113 The method of embodiment 111, wherein the animal is a human.
  • Embodiment 114 The method of embodiment 111 to 113, wherein the cancerous tumor comprises glioblastoma.
  • Embodiment 115 The method of any of embodiments 86 to 102, further comprising contacting a cell with a second modified oligonucleotide, wherein the second modified oligonucleotide is different from the compound according to any of embodiments 1-83.
  • Embodiment 116 The method of any of embodiments 103 to 114, further comprising administering a second modified oligonucleotide, wherein the second modified oligonucleotide is different from the compound according to any of embodiments 1-83.
  • Embodiment 117 The method of embodiment 115 or 116, wherein the second modified
  • oligonucleotide has the nucleobase sequence and motif of ISIS No. 549197.
  • Embodiment 118 Use of the compound according to any of embodiments 1-83 or the pharmaceutical composition of embodiments 84 or 85 for the preparation of a medicament for use in the treatment of cancer.
  • Embodiment 119 Use of the compound according to any of embodiments 1-83 or the pharmaceutical composition of embodiments 84 or 85 for the preparation of a medicament for use in the amelioration of one or more symptoms cancer.
  • Embodiment 120 The use of embodiment 118 or 119, wherein the cancer is glioblastoma.
  • Embodiment 121 Use of the compound according to any of embodiments 1-83 or the pharmaceutical composition of embodiments 84 or 85 for the treatment of cancer.
  • Embodiment 122 Use of the compound according to any of embodiments 1-83 or the pharmaceutical composition of embodiments 84 or 85 for the treatment of glioblastoma.
  • Embodiment 123 The method of any of embodiments 115-117, wherein the first modified
  • oligonucleotide has the nucleobase sequence of Isis No. 461378.
  • Embodiment 124 A compound comprising a modified oligonucleotide consisting of 8 to 80 linked nucleosides complementary within nucleobases 28709-28726, 28710-28727, 28711-28728, 28712- 28729, 28713-28730, 28714-28731, 28715-28732, 28716-28733, 28717-28734, 28718-28735, 28719- 28736, 28720-28737, 28721-28738, 28722-28739, 28723-28740, 28724-28741, 28725-28742 of SEQ ID
  • modified oligonucleotide is at least 85%, 90%>, 95%, or 100%) complementary to SEQ ID NO: 1.
  • Embodiment 125 A compound comprising a modified oligonucleotide having a nucleobase sequence consisting of any one of SEQ ID NO: 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 1 1 1, 112, or 113.
  • Embodiment 126 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 8 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 1 10, 11 1, 1 12, or 113.
  • Embodiment 127 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 9 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 86, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 1 10, 11 1, 1 12, or 113.
  • Embodiment 128 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 10 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 1 10, 11 1, 1 12, or 113.
  • Embodiment 129 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 1 1 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 1 10, 11 1, 1 12, or 113.
  • Embodiment 130 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 12 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, or 113.
  • Embodiment 131 A compound comprising a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising the nucleobase sequence of any one of SEQ ID NOs: 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, or 113.
  • Embodiment 132 The compound of any of embodiments 124-131, wherein the complementary region of the modified oligonucleotide is 90% complementary to the target region.
  • Embodiment 133 The compound of any of embodiments 124-131, wherein the complementary region of the modified oligonucleotide is 100% complementary to the target region.
  • Embodiment 134 The compound of any of embodiments 124-133, wherein the modified
  • oligonucleotide comprises at least one modified nucleoside.
  • Embodiment 135 The compound of embodiment 134, wherein at least one modified nucleoside
  • Embodiment 136 The compound of embodiment 135, wherein at least one modified sugar moiety is a 2'-substituted sugar moiety.
  • Embodiment 137 The compound of embodiment 136, wherein the 2'-substitutent of at least one 2'- substituted sugar moiety is selected from among: 2'-OMe, 2'-F, and 2'-MOE.
  • Embodiment 138 The compound of any of embodiments 136-137, wherein the 2'-substiuent of at least one 2 '-substituted sugar moiety is a 2'-MOE.
  • Embodiment 139 The compound of any of embodiments 124-135, wherein at least one modified sugar moiety is a bicyclic sugar moiety.
  • Embodiment 140 The compound of embodiment 139, wherein at least one bicyclic sugar moiety is cEt.
  • Embodiment 141 The compound of embodiment 139, wherein at least one bicyclic sugar moiety is LNA.
  • Embodiment 142 The compound of embodiment 135, wherein at least one sugar moiety is a sugar surrogate.
  • Embodiment 143 The compound of embodiment 142, wherein at least one sugar surrogate is a
  • Embodiment 144 The compound of embodiment 142, wherein at least one sugar surrogate is a
  • Embodiment 145 The compound of any of embodiments 124-144, wherein the modified
  • oligonucleotide comprises at least 5 modified nucleosides, each independently comprising a modified sugar moiety.
  • Embodiment 146 The compound of any of embodiments 124-144, wherein the modified
  • oligonucleotide comprises at least 10 modified nucleosides, each independently comprising a modified sugar moiety.
  • Embodiment 147 The compound of any of embodiments 124-144, wherein the modified
  • oligonucleotide comprises at least 15 modified nucleosides, each independently comprising a modified sugar moiety.
  • Embodiment 148 The compound of any of embodiment 124-144, wherein each nucleoside of the modified oligonucleotide is a modified nucleoside, each independently comprising a modified sugar moiety
  • Embodiment 149 The compound of any of embodiment 124-144, wherein the modified
  • oligonucleotide comprises at least two modified nucleosides comprising modified sugar moieties that are the same as one another.
  • Embodiment 150 The compound of any of embodiments 124-144, wherein the modified
  • oligonucleotide comprises at least two modified nucleosides comprising modified sugar moieties that are different from one another.
  • Embodiment 151 The compound of any of embodiments 124-150, wherein the modified oligonucleotide comprises a modified region of at least 5 contiguous modified nucleosides.
  • Embodiment 152 The compound of any of embodiments 124-150, wherein the modified
  • oligonucleotide comprises a modified region of at least 10 contiguous modified nucleosides.
  • Embodiment 153 The compound of any of embodiments 124-150, wherein the modified
  • oligonucleotide comprises a modified region of at least 15 contiguous modified nucleosides.
  • Embodiment 154 The compound of any of embodiments 124-150, wherein the modified
  • oligonucleotide comprises a modified region of at least 18 contiguous modified nucleosides.
  • Embodiment 155 The compound of any of embodiments 151-154, wherein each modified nucleoside of the modified region has a modified sugar moiety independently selected from among: 2'-F, 2'-OMe, 2'-MOE, cEt, LNA, morpholino, and modified morpholino.
  • Embodiment 156 The compound of any of embodiments 151-154, wherein the modified nucleosides of the modified region each comprise the same modification as one another.
  • Embodiment 157 The compound of any of embodiments 151-154, wherein the modified nucleosides of the modified region each comprise the same 2'-substituted sugar moiety.
  • Embodiment 158 The compound of embodiment 157, wherein the 2'-substituted sugar moiety of the modified nucleosides of the region of modified nucleosides is selected from 2'-F, 2'-OMe, and 2'-MOE.
  • Embodiment 159 The compound of embodiment 157, wherein the 2'-substituted sugar moiety of the modified nucleosides of the region of modified nucleosides is 2'-MOE.
  • Embodiment 160 The compound of embodiment 155, wherein the modified nucleosides of the region of modified nucleosides each comprise the same bicyclic sugar moiety.
  • Embodiment 161 The compound of embodiment 160, wherein the bicyclic sugar moiety of the
  • modified nucleosides of the region of modified nucleosides is selected from LNA and cEt.
  • Embodiment 162 The compound of embodiment 155, wherein the modified sugar of the modified nucleosides of the region of modified nucleosides is a morpholino.
  • Embodiment 163 The compound of embodiment 155, wherein the modified sugar of the modified nucleosides of the region of modified nucleosides is a modified morpholino.
  • Embodiment 164 The compound of any of embodiments 124-144, wherein the modified
  • oligonucleotide comprises no more than 4 contiguous naturally occurring nucleosides.
  • Embodiment 165 The compound of any of embodiments 124-144, wherein each nucleoside of the modified oligonucleotide is a modified nucleoside.
  • Embodiment 166 The compound of embodiment 165 wherein each modified nucleoside comprises a modified sugar moiety.
  • Embodiment 167 The compound of embodiment 166, wherein the modified nucleosides of the modified oligonucleotide comprise the same modification as one another.
  • Embodiment 168 The compound of embodiment 167, wherein the modified nucleosides of the modified oligonucleotide each comprise the same 2'-substituted sugar moiety.
  • Embodiment 169 The compound of embodiment 168, wherein the 2'-substituted sugar moiety of the modified oligonucleotide is selected from 2'-F, 2'-OMe, and 2'-MOE.
  • Embodiment 170 The compound of embodiment 168, wherein the 2'-substituted sugar moiety of the modified oligonucleotide is 2'-MOE.
  • Embodiment 171 The compound of embodiment 167, wherein the modified nucleosides of the modified oligonucleotide each comprise the same bicyclic sugar moiety.
  • Embodiment 172 The compound of embodiment 171, wherein the bicyclic sugar moiety of the modified oligonucleotide is selected from LNA and cEt.
  • Embodiment 173 The compound of embodiment 167, wherein the modified nucleosides of the modified oligonucleotide each comprises a sugar surrogate.
  • Embodiment 174 The compound of embodiment 173, wherein the sugar surrogate of the modified oligonucleotide is a morpholino.
  • Embodiment 175 The compound of embodiment 173, wherein the sugar surrogate of the modified oligonucleotide is a modified morpholino.
  • Embodiment 176 The compound of any of embodiments 124-144 wherein each sugar moiety of the modified oligonucleotide is a modified sugar moiety or an unmodified sugar moiety.
  • Embodiment 177 The compound of embodiment 176, wherein each unmodified sugar moiety is a 2'- deoxyfuranose sugar moiety.
  • Embodiment 178 The compound of embodiment 176 or 177, wherein each modified sugar moiety is selected from cEt, LNA, 2'-F, 2'-OMe, and 2'-MOE.
  • Embodiment 179 The compound of embodiment 178, wherein the modified sugar moiety is cEt.
  • Embodiment 180 The compound of embodiment 178, wherein the modified sugar moiety is LNA.
  • Embodiment 181 The compound of embodiment 178, wherein the modified sugar moiety is 2'F.
  • Embodiment 182 The compound of embodiment 178, wherein the modified sugar moiety is 2'-OMe.
  • Embodiment 183 The compound of embodiment 178, wherein the modified sugar moiety is 2'-MOE.
  • Embodiment 184 The compound of any of embodiments 177-183, wherein the modified
  • oligonucleotide has a aaddaddaddaddaa motif, wherein each "a” represents a modified sugar moiety and wherein each "d” represents a 2'-deoxyfuranose moiety.
  • Embodiment 185 The compound of any of embodiments 177-183, wherein the modified
  • oligonucleotide has a addaddaddadda motif, wherein each "a” represents a modified sugar moiety and wherein each "d” represents a 2'-deoxyfuranose moiety.
  • Embodiment 186 The compound of any of embodiments 124-144, wherein the modified
  • oligonucleotide has a aeeaeeaeeaeeaeea motif, wherein each "a” represents a bicyclic modified sugar moiety and wherein each "e” represents a 2 '-substituted sugar moiety.
  • Embodiment 187 The compound of any of embodiments 124-144, wherein the modified
  • oligonucleotide has a aaeeaeeaeeaeeeeea motif, wherein each "a” represents a bicyclic modified sugar moiety and wherein each "e” represents a 2 '-substituted sugar moiety.
  • Embodiment 188 The compound of any of embodiments 184-187, wherein each "a” independently represents either a cEt modified sugar moiety or an LNA modified sugar moiety.
  • Embodiment 189 The compound of embodiment 188, wherein each "a” represents a cEt modified sugar moiety.
  • Embodiment 190 The compound of any of embodiments 124-189, wherein the modified
  • oligonucleotide comprises at least one modified internucleoside linkage.
  • Embodiment 191 The compound of embodiment 190, wherein each internucleoside linkage is a
  • Embodiment 192 The compound of embodiment 190 or 191, comprising at least one phosphorothioate internucleoside linkage.
  • Embodiment 193 The compound of embodiment 190, wherein each internucleoside linkage is a
  • each internucleoside linkage comprises the same modification.
  • Embodiment 194 The compound of embodiment 190, wherein each internucleoside linkage is a
  • Embodiment 195 The compound of any of embodiments 124-190, wherein each internucleoside linkage is either a phosphodiester internucleoside linkage or a phosphorothioate internucleoside linkage.
  • Embodiment 196 The compound of any of embodiments 124-195 comprising at least one conjugate.
  • Embodiment 197 The compound of any of embodiments 124-195 consisting of the modified
  • Embodiment 198 The compound of any of embodiments 124-197, wherein the compound modulates splicing of the PK-M transcript.
  • Embodiment 199 A pharmaceutical composition comprising a compound according to any of
  • Embodiment 200 The pharmaceutical composition of embodiment 199, wherein the pharmaceutically acceptable carrier or diluent is sterile saline.
  • Embodiment 201 A method of modulating splicing of a PK-M transcript in a cell comprising
  • Embodiment 202 The method of embodiment 201, wherein the cell is in vitro.
  • Embodiment 203 The method of embodiment 201, wherein the cell is in an animal.
  • Embodiment 204 The method of any of embodiments 201-203, wherein inclusion of exon 9 is
  • Embodiment 205 The method of any of embodiments 201-204, wherein exclusion of exon 10 is
  • Embodiment 206 The method of any of embodiments 201-204, wherein inclusion of exon 10 is
  • Embodiment 207 The method of any of embodiments 201-206, wherein PK-Ml mRNA expression is increased.
  • Embodiment 208 The method of any of embodiments 201-207, wherein PK-M2 mRNA expression is decreased.
  • Embodiment 209 A method of modulating the expression of PK-M in a cell, comprising contacting the cell with a compound according to any of embodiments 124-198.
  • Embodiment 210 The method of embodiment 209, wherein PK-Ml expression is increased.
  • Embodiment 211 The method of embodiments 209 or 210, wherein PK-M2 expression is decreased.
  • Embodiment 212 The method of embodiment 209, wherein the cell is in vitro.
  • Embodiment 213 The method of embodiment 209, wherein the cell is in an animal.
  • Embodiment 214 A method of inducing apoptosis in a cell, comprising contacting the cell with a compound according to any of embodiments 124-198.
  • Embodiment 215 The method of embodiment 214, wherein the cell is a glial cell.
  • Embodiment 216 The method of embodiment 214 or 215, wherein the cell is in vitro.
  • Embodiment 217 The method of embodiment 214 or 215, wherein the cell is in an animal.
  • Embodiment 218 A method comprising administering the compound according to any of embodiments 124-196 or the pharmaceutical composition of embodiments 199 or 200 to an animal.
  • Embodiment 219 The method of embodiment 218, wherein the administration is
  • Embodiment 220 The method of embodiment 218, wherein the administration is into the central nervous sysem.
  • Embodiment 221 The method of any of embodiments 218-220, wherein the animal has one or more symptoms associated with cancer.
  • Embodiment 222 The method embodiment 221, wherein the cancer is glioblastoma.
  • Embodiment 223 The method of embodiment 221, wherein the administration results in amelioration of at least one symptom of cancer.
  • Embodiment 224 The method of any of embodiments 218-223, wherein the animal is a mouse.
  • Embodiment 225 The method of any of embodiments 218-223, wherein the animal is a human.
  • Embodiment 226 A method of preventing or retarding the growth of a cancerous tumor, comprising administering the compound according to any of embodiments 124-198 or the pharmaceutical composition of embodiments 199 or 200 to an animal in need thereof.
  • Embodiment 227 The method of embodiment 226, wherein the animal is a mouse.
  • Embodiment 228 The method of embodiment 226, wherein the animal is a human.
  • Embodiment 229 The method of embodiment 226 to 228, wherein the cancerous tumor comprises glioblastoma.
  • Embodiment 230 The method of any of embodiments 201 to 229, further comprising contacting a cell with a second modified oligonucleotide, wherein the second modified oligonucleotide is different from the compound according to any of embodiments 124-198.
  • Embodiment 231 The method of embodiment 230, wherein the second modified oligonucleotide has the nucleobase sequence and motif of ISIS No. 549197.
  • Embodiment 232 Use of the compound according to any of embodiments 124-198 or the
  • compositions 199 or 200 for the preparation of a medicament for use in the treatment of cancer.
  • Embodiment 233 Use of the compound according to any of embodiments 124-198 or the
  • compositions 199 or 200 for the preparation of a medicament for use in the amelioration of one or more symptoms cancer.
  • Embodiment 234 The use of embodiment 232 or 233, wherein the cancer is glioblastoma.
  • Embodiment 235 Use of the compound according to any of embodiments 124-198 or the
  • Embodiment 236 Use of the compound according to any of embodiments 124-198 or the
  • 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.
  • chemical modification means a chemical difference in a compound when compared to a naturally occurring counterpart.
  • chemical modification does not include differences only in nucleobase sequence.
  • Chemical modifications of oligonucleotides include nucleoside modifications (including sugar moiety modifications and nucleobase modifications) and internucleoside linkage modifications.
  • 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 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, a bicyclic or tricyclic sugar moiety, or a sugar surrogate.
  • substituted sugar moiety means a furanosyl comprising at least one substituent group that differs from that of 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.
  • 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 .
  • 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.
  • sugar surrogate means a structure that does not comprise a furanosyl and that is capable of replacing the naturally occurring sugar moiety of a nucleoside, such that the resulting nucleoside is capable of (1) incorporation into an oligonucleotide and (2) hybridization to a complementary nucleoside.
  • Such structures include rings comprising a different number of atoms than furanosyl (e.g., 4, 6, or 7-membered rings); replacement of the oxygen of a furanosyl with a non-oxygen atom (e.g., carbon, sulfur, or nitrogen); 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 morpholino, modified morpholinos, cyclohexenyls and cyclohexitols.
  • 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 means a group of atoms that can be linked to a sugar moiety to create a nucleoside that is capable of incorporation into an oligonucleotide, and wherein the group of atoms is capable of bonding with a complementary naturally occurring nucleobase of another oligonucleotide or nucleic acid. Nucleobases may be naturally occurring or may be modified.
  • heterocyclic base or “heterocyclic nucleobase” means a nucleobase comprising a heterocyclic structure.
  • unmodified nucleobase or “naturally occurring nucleobase” means the naturally occurring heterocyclic nucleobases of RNA or DNA: the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) (including 5-methyl C), and uracil (U).
  • modified nucleobase means any nucleobase that is not a naturally occurring nucleobase.
  • modified nucleoside means a nucleoside comprising at least one chemical modification compared to naturally occurring RNA or DNA nucleosides. Modified nucleosides comprise a modified sugar moiety and/or a modified nucleobase.
  • 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 )-0-2'bridge.
  • locked nucleic acid nucleoside or "LNA” means a nucleoside comprising a bicyclic sugar moiety comprising a 4'-CH 2 -0-2'bridge.
  • 2 '-substituted nucleoside means a 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 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).
  • 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.
  • oligomeric compound means a polymeric structure comprising two or more substructures.
  • an oligomeric compound comprises an oligonucleotide.
  • an oligomeric compound comprises one or more conjugate groups and/or terminal groups.
  • an oligomeric compound consists of an oligonucleotide.
  • terminal group means one or more atom attached to either, or both, the 3 ' end or the 5' end of an oligonucleotide. In certain embodiments a terminal group is a conjugate group. In certain embodiments, a terminal group comprises one or more terminal group 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 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 measureable 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.
  • transcript means an RNA molecule transcribed from DNA.
  • Transcripts include, but are not limitied to mRNA, pre-mRNA, and partially processed RNA.
  • PK-M transcript means a transcript transcribed from a PK-M gene.
  • a PK-M transcript comprises SEQ ID NO: 1 : the complement of GENBANK Accession No. NT 010194.16 truncated from nucleotides 43281289 to 43314403.
  • PK-M gene means a gene that encodes a pyruvate kinase M protein and any pyruvate kinase M protein isoforms.
  • pyruvate kinase M protein isoforms include pyruvate kinase Ml and pyruvate kinase M2.
  • a pyruvate kinase M gene is represented by GENBANK Accession No. NT 010194.16 truncated from nucleotides 43281289 to
  • a pyruvate kinase M gene is at least 95% identical to GENBANK Accession No. NT 010194.16 truncated from nucleotides 43281289 to 43314403. In certain embodiments, a pyruvate kinase M gene is at least 90% identical to GENBAN Accession No.
  • NT 010194.16 truncated from nucleotides 43281289 to 43314403.
  • PK-M1 means a pyruvate kinase M transcript that includes exon 9 but does not include exon 10.
  • PK-M1 isoform means a pyruvate kinase M protein isoform that includes amino acids encoded from exon 9 but does not include amino acids encoded from exon 10.
  • PK-M2 means a pyruvate kinase M transcript that includes exon 10 but does not include exon 9.
  • PK-M2 isoform means a pyruvate kinase M protein isoform that includes amino acids encoded from exon 10 but does not include amino acids encoded from exon 9.
  • 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. For example, if a nucleobase at a certain position of an antisense compound is capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid, then 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.
  • 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.
  • oligomeric compound specifically hybridizes to more than one target site.
  • 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.
  • 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.
  • 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.
  • telomeres As used herein, “differently modified” mean chemical modifications or chemical substituents that are different from one another, including absence of modifications. Thus, for example, a MOE nucleoside and an unmodified DNA nucleoside are “differently modified,” even though the DNA nucleoside is unmodified. Likewise, 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'- substuent 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).
  • 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 (CrC 1 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, l-methyl-2-buten-l-yl, 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(0)-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 C1-C12 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-C12 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.
  • 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 one or more nucleoside (including modifications to the sugar moiety and/or the nucleobase) and/or modifications to one or more internucleoside linkage.
  • oligomeric compounds of the invention comprise one or more modifed nucleosides comprising a modifed sugar moiety.
  • Such oligomeric 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 oligomeric compounds comprising only nucleosides comprising naturally occurring sugar moieties.
  • modified sugar moieties are substitued sugar moieties.
  • modified sugar moieties are bicyclic or tricyclic sugar moieties.
  • modified sugar moieties are sugar surrogates. Such sugar surogates may comprise one or more substitutions corresponding to those of substituted sugar moieties.
  • modified sugar moieties are substituted sugar moieties comprising one or more substituent, 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'-0(CH 2 ) 2 0CH 3 (“MOE").
  • 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, O- C r C 10 alkoxy; O- C r C 10 substituted alkoxy, SH, CN, OCN, CF 3 , OCF 3 , O-alkyl, S-alkyl, N(R m )-alkyl; O- alkenyl, S- alkenyl, or N(R m )-alkenyl; O- alkynyl, S- alkynyl, N(R m )-alkynyl; O-alkylenyl- O-alkyl, alkynyl, alkaryl, aralkyl, O-alkaryl, O-aralkyl, 0(CH 2
  • These 2'-substituent groups can be further substituted with one or more substituent groups independently selected from hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro (N0 2 ), thiol, thioalkoxy (S-alkyl), halogen, alkyl, aryl, alkenyl and alkynyl.
  • a 2'- substituted nucleoside comprises a sugar moiety comprising a 2'- substituent group selected from F, 0-CH 3 , and OCH 2 CH 2 OCH 3 .
  • Certain modifed 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.
  • Examples of such 4' to 2' sugar substituents include, but are not limited to: -[C(R a )(R b )] n -, -[C(R a )(R b )] n -0-, -C(R a R b )-N(R)-0- or, -C(R a R b )-0-N(R)-; 4'-CH 2 -2', 4'-(CH 2 ) 2 -2 ⁇ 4'-(CH 2 ) 3 -2',.
  • Patent 7,427,672 issued on September 23, 2008
  • 4'-CH 2 - C(H)(CH 3 )-2' see, e.g., Chattopadhyaya, et al, J. Org. Chem.,2009, 74, 118-134
  • x 0, 1, or 2;
  • n 1, 2, 3, or 4;
  • Bicyclic nucleosides include, but are not limited to, (A) a-L-Methyleneoxy (4'-CH 2 -0-2') BNA , (B) ⁇ -D- Methyleneoxy (4'-CH 2 -0-2') BNA (also referred to as locked nucleic acid or LNA) , (C) Ethyleneoxy (4'-
  • Bx is a nucleobase moiety and R is, inde endently, H, a protecting group, or C1-C12 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 a-L configuration or in the ⁇ -D configuration.
  • a-L- methyleneoxy (4'-CH 2 -0-2') 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), ⁇ see, PCT International Application WO 2007/134181, published on 11/22/07, wherein LNA is substituted with, for example, a 5'-methyl or a 5'-vinyl group).
  • bridging sugar substituent e.g., 5 '-substituted and 4 '-2' bridged sugars
  • modified sugar moieties are sugar surrogates.
  • the oxygen atom of the naturally occuring 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 June 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 (UNA), anitol nucleic acid (ANA), manitol nucleic acid (MNA) (see Leumann, CJ. Bioorg. & Med. Chem. (2002) 10:841-854), fluoro HNA (F-HNA), and those compounds having Formula
  • Bx is a nucleobase moiety
  • T3 and T4 are each, independently, an internucleoside linking group linking the tetrahydropyran nucleoside analog to the antisense compound or one of T3 and T 4 is an internucleoside linking group linking the tetrahydropyran nucleoside analog to the antisense compound and the other of T3 and T4 is H, a hydroxyl protecting group, a linked conjugate group, or a 5' or 3'-terminal group;
  • qi, q 2 , q3, q 4 , qs, q6 and q 7 are each, independently, H, Ci-Ce alkyl, substituted C1-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl, or substituted C2-C6 alkynyl; and
  • each of Ri and R2 is independently selected from among: hydrogen, halogen, substituted or unsubstituted alkoxy, NJJ 2 , SJ b N 3 , and CN, wherein X is O, S or NJi, and each Ji, J 2 , and J3 is, independently, H or Ci-Ce alkyl.
  • the modified THP nucleosides of Formula VII are provided wherein qi, q 2 , q3, q 4 , q 5 , q6 and q 7 are each H. In certain embodiments, at least one of qi, q 2 , q3, q 4 , qs, q6 3 ⁇ 4nd q 7 is other than H. In certain embodiments, at least one of qi, q 2 , q3, q 4 , qs, q6 and q 7 is methyl. In certain embodiments, THP nucleosides of Formula VII are provided wherein one of Ri and R 2 is F. In certain embodiments, Ri is fluoro and R 2 is H, Ri is methoxy and R 2 is H, and Ri is methoxyethoxy and R2 is H.
  • sugar surrogates comprise rings having more than 5 atoms and more than one heteroatom.
  • nucleosides comprising morpholino sugar moieties and their use in oligomeric compounds has been reported (see for example: Braasch et al., Biochemistry, 2002, 41, 4503-4510; and U.S. Patents 5,698,685; 5,166,315; 5,185,444; and 5,034,506).
  • morpholino means a sugar llowing structure:
  • morpholinos may be modified, for example by adding or altering various substituent groups from the above morpholino structure.
  • sugar surrogates are refered to herein as "modifed morpholinos.”
  • nucleosides of the present invention comprise one or more unmodified nucleobases. In certain embodiments, nucleosides of the present invention comprise one or more modifed nucleobases.
  • modified nucleobases are selected from: universal bases, hydrophobic bases, promiscuous bases, size-expanded bases, and fluorinated bases as defined herein.
  • nucleobases include tricyclic pyrimidines such as phenoxazine cytidine( [5,4-b][l,4]benzoxazin- 2(3H)-one), phenothiazine cytidine (lH-pyrimido[5,4-b][l,4]benzothiazin-2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g.
  • nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2- pyridone.
  • nucleobases include those disclosed in United States Patent No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, Kroschwitz, J.I., Ed., John Wiley & Sons, 1990, 858-859; those disclosed by Englisch et al, Angewandte Chemie, International Edition, 1991, 30, 613; and those disclosed by Sanghvi, Y.S., Chapter 15, Antisense Research and Applications, Crooke, S.T. and Lebleu, B., Eds., CRC Press, 1993, 273-288.
  • the present invention provides oligomeric compounds comprising linked nucleosides.
  • nucleosides may be linked together using any internucleoside linkage.
  • the two main classes of internucleoside linking groups are defined by the presence or absence of a phosphorus atom.
  • Non-phosphorus containing internucleoside linking groups include, but are not limited to, methylenemethylimino (-CH 2 -N(CH 3 )-0-CH 2 -), thiodiester (-O-C(O)-S-), thionocarbamate (-0- C(0)(NH)-S-); siloxane (-0-Si(H) 2 -0-); and ,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 oligomeric compound.
  • 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), a 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.
  • Further neutral internucleoside linkages include nonionic linkages comprising siloxane (dialkylsiloxane), carboxylate ester, carboxamide, sulfide, sulfonate ester and amides (See for example: Carbohydrate Modifications in Antisense Research; Y.S. Sanghvi and P.D. Cook, Eds., ACS Symposium Series 580; Chapters 3 and 4, 40-65). Further neutral internucleoside linkages include nonionic linkages comprising mixed N, O, S and CH 2 component parts.
  • the present invention provides oligomeric compounds comprising oligonucleotides.
  • such oligonucleotides comprise one or more chemical modification.
  • chemically modified oligonucleotides comprise one or more modified nucleosides.
  • chemically modified oligonucleotides comprise one or more modified nucleosides comprising modified sugars.
  • chemically modified oligonucleotides comprise one or more modified nucleosides comprising one or more modified nucleobases.
  • chemically modified oligonucleotides comprise one or more modified internucleoside linkages.
  • the chemically 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 modification motif.
  • Such motifs may include any of the sugar modifications discussed herein and/or other known sugar modifications.
  • the oligonucleotides comprise or consist of a region having a gapmer sugar modification motif, which comprises two external regions or "wings" and an internal region or "gap.”
  • the three regions of a gapmer 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 modification motifs of the two wings are the same as one another (symmetric gapmer).
  • the sugar modification motifs of the 5'-wing differs from the sugar modification motif of the 3'-wing (asymmetric gapmer).
  • oligonucleotides comprise 2'-MOE modified nucleosides in the wings and 2'-F modified nucleosides in the gap.
  • oligonucleotides are fully modified. In certain such embodiments, oligonucleotides are uniformly modified. In certain embodiments, oligonucleotides are uniform 2'-MOE. In certain embodiments, oligonucleotides are uniform 2'-F. In certain embodiments, oligonucleotides are uniform morpholino. In certain embodiments, oligonucleotides are uniform BNA. In certain embodiments, oligonucleotides are uniform LNA. In certain embodiments, oligonucleotides are uniform cEt.
  • oligonucleotides comprise a uniformly modified region and additional nucleosides that are unmodified or differently modified.
  • the uniformly modified region is at least 5, 10, 15, or 20 nucleosides in length.
  • the uniform region is a 2'- MOE region.
  • the uniform region is a 2'-F region.
  • the uniform region is a morpholino region.
  • the uniform region is a BNA region.
  • the uniform region is a LNA region.
  • the uniform region is a cEt region.
  • the oligonucleotide does not comprise more than 4 contiguous unmodified 2'-deoxynucleosides.
  • antisesense oligonucleotides comprising more than 4 contiguous 2'-deoxynucleosides activate RNase H, resulting in cleavage of the target RNA.
  • such cleavage is avoided by not having more than 4 contiguous 2'-deoxynucleosides, for example, where alteration of splicing and not cleavage of a target RNA is desired.
  • oligonucleotides comprise modified internucleoside linkages arranged along the oligonucleotide or region thereof in a defined pattern or modified internucleoside linkage motif.
  • internucleoside linkages are arranged in a gapped motif, as described above for sugar modification 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 sugar modification motif is independently selected, so such oligonucleotides having a gapped internucleoside linkage motif may or may not have a gapped sugar modification motif and if it does have a gapped sugar motif, the wing and gap lengths may or may not be the same.
  • oligonucleotides comprise a region having an alternating internucleoside linkage motif. In certain embodiments, oligonucleotides of the present invention comprise a region of uniformly modified internucleoside linkages. In certain such embodiments, the oligonucleotide 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 oligonucleotide is selected from phosphodiester and phosphorothioate. In certain
  • each internucleoside linkage of the oligonucleotide is selected from phosphodiester and phosphorothioate and at least one internucleoside linkage is phosphorothioate.
  • the oligonucleotide comprises at least 6 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 8 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 10 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 6 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 8 consecutive phosphorothioate internucleoside linkages.
  • the oligonucleotide comprises at least one block of at least 10 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 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 oligonucleotide. In certain such embodiments, at least one such block is located within 3 nucleosides of the 3 ' end of the oligonucleotide.
  • oligonucleotides comprise chemical modifications to nucleobases arranged along the oligonucleotide or region thereof in a defined pattern or nucleobases modification motif.
  • nucleobase modifications are arranged in a gapped motif.
  • nucleobase modifications are arranged in an alternating motif.
  • each nucleobase is modified.
  • none of the nucleobases is chemically modified.
  • oligonucleotides comprise a block of modified nucleobases. In certain such embodiments, the block is at the 3 '-end of the oligonucleotide. In certain embodiments the block is within 3 nucleotides of the 3'-end of the oligonucleotide. In certain such embodiments, the block is at the 5'-end of the oligonucleotide. In certain embodiments 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.
  • cytosine moieties in an oligonucleotide are 5- methyl cytosine moieties.
  • 5-methyl cytosine is not a "modified nucleobase.”
  • unmodified nucleobases include both cytosine residues having a 5-methyl and those lacking a 5 methyl.
  • the methylation state of all or some cytosine nucleobases is specified.
  • 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 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, and 50; provided that X ⁇ Y.
  • the invention provides oligomeric compounds which comprise oligonucleotides consisting of 8 to 9, 8 to 10, 8 to 11, 8 to 12, 8 to 13, 8 to 14, 8 to 15, 8 to 16, 8 to 17, 8 to 18, 8 to 19, 8 to 20, 8 to 21, 8 to 22, 8 to 23, 8 to 24, 8 to 25, 8 to 26, 8 to 27, 8 to 28, 8 to 29, 8 to 30, 9 to 10, 9 to 11, 9 to 12, 9 to 13, 9 to 14, 9 to 15, 9 to 16, 9 to 17, 9 to 18, 9 to 19, 9 to 20, 9 to 21, 9 to 22, 9 to 23, 9 to 24, 9 to 25, 9 to 26, 9 to 27, 9 to 28, 9 to 29, 9 to 30, 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 15, 11 to 16, 11 to
  • nucleosides of an 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.
  • a gapmer having a 5'-wing region consisting of four nucleotides, a gap consisting of at least six nucleotides, and a 3 '-wing region consisting of three nucleotides cannot have an overall length less than 13 nucleotides.
  • the lower length limit is 13 and that the limit of 10 in "10-20" has no effect in that embodiment.
  • 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.
  • Such additional nucleosides may be 5' of the 5 '-wing and/or 3 ' of the 3 ' wing.
  • oligonucleotides of the present invention are characterized by their sugar motif, internucleoside linkage motif, nucleobase modification motif and overall length. In certain embodiments, such parameters are each independent of one another. Thus, 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. Thus, 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.
  • modified nucleobase independent of the gapmer pattern of the sugar modifications.
  • a description of an 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, 36 1 -3654), a palmityl moiety (Mishra et al., Biochim.
  • a conjugate group comprises an active drug substance, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fen-bufen, ketoprofen, (S)-(+)-pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, 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, (S)-(+)-pranoprofen, carprofen, dansyls
  • 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-l -carboxylate (SMCC) and 6- aminohexanoic acid (AHEX or AHA).
  • ADO 8-amino-3,6- dioxaoctanoic acid
  • SMCC succinimidyl 4-(N-maleimidomethyl) cyclohexane-l -carboxylate
  • AHEX or AHA 6- aminohexanoic acid
  • linking groups include, but are not limited to, substituted Ci- Cio alkyl, substituted or unsubstituted C 2 -Ci 0 alkenyl or substituted or unsubstituted C 2 -Ci 0 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 chemical motifs discussed above.
  • an oligomeric compound comprising an oligonucleotide having region of alternating nucleosides may comprise a terminal group.
  • oligomeric compounds of the present invention 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. In certain embodiments, 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.
  • antisense compounds and antisense oligonucleotides comprise single-strand compounds. In certain embodiments antisense compounds and antisense oligonucleotides comprise double- strand compounds.
  • PK-M2 mediates the Warburg effect.
  • expression of PK-M2 is crucial for tumor cell growth and proliferation.
  • reducing expression of PK-M2 inhibits cancer growth. In certain embodiments, reducing expression of PK-M2 induces apoptosis in a cell. In certain embodiments, the cell is a cancer cell. In certain embodiments, the cell is a tumor cell. In certain imbodiments, the cell is a glioblastoma cell.
  • increasing inclusion of exon 9 of a PK-M transcript inhibits cancer growth. In certain embodiments, increasing exclusion of exon 10 of a PK-M transcript inhibits cancer growth. In certain embodiments, increasing inclusion of exon 9 of a PK-M transcript induces apoptosis in a cell. In certain embodiments, increasing exclusion of exon 10 of a PK-M transcript induces apoptosis in a cell. In certain embodiments, the cell is a cancer cell. In certain embodiments, the cell is a tumor cell. In certain imbodiments, the cell is a glioblastoma cell. In certain embodiments, the downregulation of PK-M2 leads to apoptosis in certain cancer cells. In certain embodiments, the downregulation of PK-M2 leads to apoptosis in certain glioblastoma cell lines.
  • PK-M2 also functions as a co-activator of HIF-1 and/ or ⁇ -catenin. In certain embodiments, reducing expression of PK-M2, as opposed to inhibiting its kinase function, interferes with anti-apoptotic and pro-proliferative functions associated with cancer or tumor cells. In certain embodiments, one or more antisense compounds may be used to target a PK-M2.
  • the administration of a modified oligonucleotide causes a switch in the alternative splicing of the PK-M transcript. In certain embodiments, the administration of a modified oligonucleotide causes increased inclusion of exon 9 mRNA of the PK-M transcript. In certain embodiments, the administration of a modified oligonucleotide causes an increase in the exclusion of exon 10 mRNA of the PK-M transcript. In certain embodiments, the administration of a modified oligonucleotide reduces expression of PK-M2 in a cell. In certain embodiments, the administration of a modified oligonucleotide reduces expression of PK-M2 in a cell and inhibits cancer growth. In certain embodiments, the
  • the cell is a cancer cell.
  • the cell is a tumor cell.
  • the cell is a glioblastoma cell.
  • compounds comprise or consist of a modified 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 pre-mRNA.
  • the target nucleic acid is a PK-M transcript.
  • the target RNA is a PK- M pre-mRNA.
  • a modified oligonucleotide is complementary to a region of PK-M pre- mRNA. In certain embodiments, a modified oligonucleotide is complementary within a region of PK-M pre- mRNA comprising an exon encoding PK-M2. In certain embodiments, a modified oligonucleotide is complementary to a region of PK-M pre-mRNA comprising an intron-exon splice junction. In certain embodiments, a modified oligonucleotide is complementary to a region of P -M pre-mRNA comprising the intron-exon splice junction adjacent to exon 10.
  • a modified oligonucleotide is complementary within a region of PK-M pre- mRNA consisting of exon 10. In certain embodiments, a modified oligonucleotide is complementary within a region of PK-M pre- mRNA consisting of intron 9. In certain embodiments, a modified oligonucleotide is complementary within a region of PK-M pre-mRNA comprising an exonic splicing silencer within exon 10. In certain embodiments, a modified oligonucleotide is complementary within a region of PK-M pre-mRNA comprising an exonic splicing enhancer within intron 9.
  • a modified oligonucleotide is complementary within a region of PK-M pre-mRNA comprising an exonic splicing enhancer within exon 10. In certain embodiments, a modified oligonucleotide is complementary within a region of PK-M pre-mRNA comprising an exonic splicing silencer within exon 9. In certain embodiments, a modified oligonucleotide is complementary within a region of PK-M pre-mRNA comprising an exonic splicing enhancer within exon 9.
  • a modified oligonucleotide comprises a modified oligonucleotide consisting of 8 to 30 linked nucleosides and having a nucleobase sequence comprising a complementary region comprising at least 8 contiguous nucleobases complementary to a target region of equal length of a PK-M transcript.
  • the target region is within nucleobase 28726 and nucleobase 29137 of SEQ ID NO.: 1.
  • the target region is within nucleobase 28726 and nucleobase 28787 of SEQ ID NO. : 1.
  • the target region is within nucleobase 28714 and nucleobase 29257 of SEQ ID NO.: 1.
  • the target region is within nucleobase 29128 and nucleobase 29162 of SEQ ID NO.: 1. In certain embodiments, the target region is within nucleobase 29159 and nucleobase 29193 of SEQ ID NO.: 1. In certain embodiments, the target region is within nucleobase 29164 and nucleobase 29183 of SEQ ID NO.: 1. In certain embodiments, the target region is within nucleobase 28826 and nucleobase 29183 of SEQ ID NO.: 1.
  • a modified oligonucleotide modulates splicing of a pre-mRNA. In certain embodiments, a modified oligonucleotide modulates splicing a PK-M pre-mRNA. In certain embodiments, a modified oligonucleotide increases the amount of PK-M mRNA. In certain embodiments, a modified oligonucleotide increases the inclusion of exon 9 in PK-M mRNA. In certain embodiments, a modified oligonucleotide decreases the inclusion of exon 10 in PK-M mRNA. In certain embodiments, a modified oligonucleotide increases the amount of PK-Ml mRNA. In certain embodiments, a modified oligonucleotide decreases the amount of PK-M2 mRNA.
  • PK-M pre-mRNA it is desirable to alter the splicing of PK-M pre-mRNA to include exon 9 and exclude exon 10.
  • expression of PK-Ml will increase and expression of PK-M2 will decrease.
  • splicing of PK-M pre-mRNA is altered by contacting a cell with more than one modified oligonucleotide. In certain embodiments, splicing of PK-M pre-mRNA is altered by contacting a cell with a modified oligonucleotide that targets ex on 10 and a second modified oligonucleotide that targets intron 9.
  • modified oligonucleotides targeted to exon 10 are described in PCT Publication No. WO 2014/071078, which is hereby incorporated by reference in its entirety. In certain embodiments, modified oligonucleotides that target exon 10 are described in Table A below.
  • modified oligonucleotides in Table A below were designed as uniform oligonucleotides, 15 nucleotides in length, with every nucleoside having a 2'-0-methoxyethyl ribose sugar residue and a fully phosphorothioate backbone. All the cytosine nucleobases are 5-methylcytosines.
  • splicing of PK-M pre-mRNA is altered by contacting a cell with more than one modified oligonucleotide. In certain embodiments, splicing of PK-M pre-mRNA is altered by contacting a cell with a modified oligonucleotide that targets exon 10 and a second modified oligonucleotide that targets intron 9. For example, in certain embodiments, splicing of PK-M pre-mRNA is altered by contacting a cell both ISIS 549197 and ISIS 461378.
  • splicing of PK-M pre-mRNA is altered by contacting a cell both ISIS 549197 and a modified oligonucleotide consisting of 10 to 30 linked nucleosides and having a nucleobase sequence comprising at least 12 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 51 , 52, 53, 54, 55, 56, 57, 58, 59, 60, 61 , 62, 63, 64, 65, 66, 67, 68, 69, 70, 71 , 72, 73, 74, 75, 76, 77, 78, 79, 80, 81,
  • 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.
  • VPD co-solvent system 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.
  • co-solvent systems may be varied considerably without significantly altering their solubility and toxicity characteristics.
  • identity of 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
  • 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).
  • 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 eyes, ears).
  • a pharmaceutical composition is administered to an animal having at least one cancer cell. In certain embodiments, such administration results in apoptosis of at least cancer cell. In certain embodiments, a pharmaceutical composition is administered to an animal having at least one symptom associated with cancer. In certain embodiments, such administration results in amelioration of at least one symptom. In certain embodiments, administration of a pharmaceutical composition to an animal results in a decrease of PK-M2 mRNA in a cell of the animal. In certain embodiments, such administration results in an increase in PK-M1 mRNA. In certain embodiments, such administration results in a decrease in PK-M2 protein and an increase PK-M1 protein.
  • a PK-M1 protein is preferred over a PK-M2 protein.
  • the administration of certain antisense oligonucleotides delays the onset of cancer. In certain embodiments, the administration of certain antisense oligonucleotides slows the proliferation of cancer cells. In certain embodiments, the administration of certain antisense oligonucleotides slows the proliferation of tumor cells. In certain embodiments, the administration of certain antisense oligonucleotides prevents the growth of cancer. In certain embodiments, the administration of certain antisense oligonucleotides prevents the formation of tumors. In certain embodiments, the administration of certain antisense oligonucleotides causes tumor mass to decrease. In certain embodiments, the administration of certain antisense oligonucleotides rescues cellular phenotype.
  • RNA nucleoside comprising a 2' -OH sugar moiety and a thymine base
  • RNA having a modified sugar 2'-OH for the natural 2'-H of DNA
  • RNA having a modified base thymine (methylated uracil) for natural uracil of RNA
  • 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 is 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 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 is 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.
  • 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.
  • Example 1 Effect of antisense oligonucleotides targeting pyruvate kinase M
  • the antisense oligonucleotides (ASOs) in the table below were designed to target intron 9 or the junction between intron 9 and exon 10 of human pyruvate kinase M (P -M).
  • the ASOs comprise uniform
  • NT 010194.16 truncated from nucleotides 43281289 to 43314403 (designated herein as SEQ ID NO: 1), and the start and stop sites of each ASO on that sequence are shown in the table below.
  • HEK-293 cells were transfected with each ASO at a final concentration of 60 nM.
  • HEK-293 cells were obtained from ATCC and grown at a density of 4 x 10 5 cells in 6-cm dishes in DMEM supplemented with 10% (v/v) FBS, penicillin, and streptomycin, at 37°C and 5% CO 2 .
  • Transfections were performed using an ASO: LipofectAMINE2000® ratio of 20 pmoles: 1 ⁇ ,.
  • hPKMF 5'- AGAAACAGCCAAAGGGGACT-3 ' (designated herein as SEQ ID NO: 117)
  • hPKMR 5'- CATTCATGGCAAAGTTCACC-3 ' (designated herein as SEQ ID NO: 118).
  • the products were analyzed on a 5% native polyacrylamide gel, visualized by autoradiography, and quantified on a Typhoon 9410 phosphorimager (GE Healthcare) using Multi Gauge software Version 2.3.
  • the results are presented in Table 1.
  • the % Ml mRNA in endogenous transcripts was calculated using the GC -content-normalized intensities of the top undigested band (Ml), the bottom two digested bands of M2 in the PstI -digested samples, and the double-skipped species (D), if detectable.
  • Ml top undigested band
  • D double-skipped species
  • Each product was quantified as a percentage of the total of Ml, M2, and double-skipped species.
  • %M1 and %M2 are presented in the Table 1 below.
  • the background negative-control levels are ⁇ 4% Ml and 96% M2; " ⁇ " indicates a visually estimated value.
  • Table 1 does not include a column for the double-skipped species (
  • Example 2 Design of antisense oligonucleotides targeting pyruvate kinase M
  • ASOs antisense oligonucleotides
  • the ASOs presented in Table 2 are 15-mer oligonucleotides and comprise uniform 2'-0- methoxyethyl ribose sugar modifications and a uniform phosphorothioate backbone. All the cytosine nucleobases are 5-methylcytosines.
  • the ASOs presented in Table 3 comprise 18-mer oligonucleotides with deoxy and cEt modifications.
  • the Chemistry column denotes the position of the sugar modifications: 'k' is a cEt modification; 'd' is a deoxyribose sugar.
  • the ASOs comprise a uniform phosphorothioate backbone. All the cytosine nucleobases are 5-methylcytosines.
  • the ASOs target intron 9b/exon 10 of SEQ ID NO: 1, and the start and stop sites of each ASO on that sequence are shown in the table below.
  • the ASOs presented in Table 4 are 18-mer oligonucleotides and comprise uniform 2'-0- methoxyethyl ribose sugar modifications and a uniform phosphorothioate backbone. All the cytosine nucleobases are 5-methylcytosines.
  • the ASOs target intron 9b/exon 10 of SEQ ID NO: 1, and the start and stop sites of each ASO on that sequence are shown in the table below.
  • the ASOs presented in Table 5 are 15-mer oligonucleotides and comprise uniform 2'-0- methoxyethyl ribose sugar modifications and a uniform phosphorothioate backbone. All the cytosine nucleobases are 5-methylcytosines.
  • the ASOs target intron 9b/exon 10 of SEQ ID NO: 1, and the start and stop sites of each ASO on that sequence are shown in the table below.
  • the ASOs presented in Table 6 are 15-mer oligonucleotides and comprise uniform 2'-0- methoxyethyl ribose sugar modifications and a uniform phosphorothioate backbone. All the cytosine nucleobases are 5-methylcytosines.
  • the ASOs target the exon 8/intron 9 junction or intron 9 of SEQ ID NO: 1, and the start and stop sites of each ASO on that sequence are shown in the table below.
  • the ASOs presented in Table 7 are oligonucleotides with deoxy ("d"), cEt modifications ("k”), and or 2'-0-methoxyethyl (“e") modifications.
  • the Chemistry column denotes the position of the sugar modifications.
  • the ASOs comprise a uniform phosphorothioate backbone. All the cytosine nucleobases are 5- methylcytosines.
  • the ASOs target intron 9b/exon 10 of SEQ ID NO: 1, and the start and stop sites of each ASO on that sequence are shown in the table below.
  • the ASOs presented in Table 8 are 15-mer oligonucleotides and comprise uniform 2'-0- methoxyethyl ribose sugar modifications and a uniform phosphorothioate backbone. All the cytosine nucleobases are 5-methylcytosines.
  • the ASOs target the exon 10/intron 10 junction or intron 10 of SEQ ID NO: 1, and the start and stop sites of each ASO on that sequence are shown in the table below.
  • Example 3 Effect of antisense oligonucleotides targeting pyruvate kinase M
  • Antisense oligonucleotides described in Example 2 in the table below were tested for their effects on endogenous PK-M transcripts, as described in Example 1. The results are presented in Table 9 below. Table 9 does not include a column for the double-skipped species (D), because it was not detected in this experiment.
  • Antisense oligonucleotides targeting intron 9b/exon 10 of PK-M were tested for their effects on endogenous PK-M transcripts.
  • U87-MG were obtained from ATCC and grown in DMEM supplemented with 10% (v/v) FBS, penicillin, and streptomycin, at 37°C and 5% C0 2 .
  • ASO transfections were conducted as described in Example 1. Splicing of the PK-M transcripts by radioactive RT-PCR was analyzed 48 hrs after transfection, as described in Example 1. The results are presented in Table 10, below.
  • Example 5 Effect combination treatment of antisense oligonucleotides targeting pyruvate kinase M
  • Isis No. 461378 see Table 1
  • Isis No. 549197 see Table A
  • both were transfected into A172 cells at a final, total concentration of 120 nM, and compared to a control ASO that is not complementary to PK-M transcript (ISIS No. 439273).
  • A172 cells were obtained from ATCC and cultured at a density of 4 x 10 5 cells in 6 cm dishes in DMEM media, supplemented with 1% (v/v) FBS, penicillin and streptomycin, at 37°C and 5% C0 2 . Transfections were performed using 7ul of Lipofectamine® RNAiMAX.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Virology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des composés comprenant des oligonucléotides complémentaires à un produit de transcription d'une pyruvate-kinase M. Certains de ces composés sont utiles pour l'hybridation à un produit de transcription d'une pyruvate-kinase M, incluant, sans y être limité, un produit de transcription d'une pyruvate-kinase M dans une cellule. Dans certains modes de réalisation, une telle hybridation aboutit à la modulation de l'épissage du produit de transcription de la pyruvate-kinase M. Dans certains modes de réalisation, ces composés sont utilisés pour traiter un ou plusieurs symptômes associés au cancer.
PCT/US2015/055582 2014-10-14 2015-10-14 Composés antisens et leurs utilisations WO2016061263A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462063892P 2014-10-14 2014-10-14
US62/063,892 2014-10-14

Publications (1)

Publication Number Publication Date
WO2016061263A1 true WO2016061263A1 (fr) 2016-04-21

Family

ID=55747287

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/055582 WO2016061263A1 (fr) 2014-10-14 2015-10-14 Composés antisens et leurs utilisations

Country Status (1)

Country Link
WO (1) WO2016061263A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019138057A1 (fr) * 2018-01-12 2019-07-18 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens d'alpha-synucléine et leurs utilisations
RU2773197C2 (ru) * 2018-01-12 2022-05-31 Рош Инновейшен Сентер Копенгаген А/С Антисмысловые олигонуклеотиды к альфа-синуклеину и их применения
EP4035659A1 (fr) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes destinés à l'administration d'agents thérapeutiques
US20220280545A1 (en) * 2018-09-10 2022-09-08 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating cln3 expression

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040192628A1 (en) * 1999-08-19 2004-09-30 Monia Brett P. Antisense modulation of focal adhesion kinase expression
US20130289092A1 (en) * 2010-06-15 2013-10-31 Cold Spring Harbor Laboratory Compounds and methods for modulating interaction between proteins and target nucleic acids
WO2014071078A1 (fr) * 2012-10-31 2014-05-08 Isis Pharmaceuticals, Inc. Composés antisens et leurs utilisations

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040192628A1 (en) * 1999-08-19 2004-09-30 Monia Brett P. Antisense modulation of focal adhesion kinase expression
US20130289092A1 (en) * 2010-06-15 2013-10-31 Cold Spring Harbor Laboratory Compounds and methods for modulating interaction between proteins and target nucleic acids
WO2014071078A1 (fr) * 2012-10-31 2014-05-08 Isis Pharmaceuticals, Inc. Composés antisens et leurs utilisations

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
WANG ET AL.: "Exon-centric regulation of pyruvate kinase M alternative splicing via mutually exclusive exons", JOURNAL OF MOLECULAR CELL BIOLOGY, vol. 4, 1 November 2011 (2011-11-01), pages 79 - 87, XP055274436 *
WANG ET AL.: "Manipulation of PK -M mutually exclusive alternative splicing by antisense oligonucleotides", OPEN BIOLOGY, vol. 2, 31 October 2012 (2012-10-31), pages 1 - 11, XP055274438 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4035659A1 (fr) 2016-11-29 2022-08-03 PureTech LYT, Inc. Exosomes destinés à l'administration d'agents thérapeutiques
WO2019138057A1 (fr) * 2018-01-12 2019-07-18 Roche Innovation Center Copenhagen A/S Oligonucléotides antisens d'alpha-synucléine et leurs utilisations
RU2773197C2 (ru) * 2018-01-12 2022-05-31 Рош Инновейшен Сентер Копенгаген А/С Антисмысловые олигонуклеотиды к альфа-синуклеину и их применения
US20220280545A1 (en) * 2018-09-10 2022-09-08 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating cln3 expression

Similar Documents

Publication Publication Date Title
EP2839006B1 (fr) Composés oligomères comprenant des nucléotides bicycliques et utilisations de ceux-ci
US9695418B2 (en) Oligomeric compounds comprising bicyclic nucleosides and uses thereof
AU2014236156C1 (en) Compositions and methods for modulating Tau expression
US9518259B2 (en) Compounds and methods for modulating interaction between proteins and target nucleic acids
US10851371B2 (en) Modulation of SMN expression
EP2751269A1 (fr) Procédés et composés utiles dans des pathologies associées à des expansions répétées
WO2012151324A1 (fr) Composés antisens ciblant des gènes associés au syndrome de usher
EP3262174A1 (fr) Composés et procédés pour augmenter l'activité antisens
WO2016154096A1 (fr) Modulation de l'expression de smggds
WO2012027033A1 (fr) Composés et procédés pour la modulation de molécules d'acide nucléique nucléaires et sub-nucléaires cibles dans des cellules et dans des animaux
US11459564B2 (en) Modulation of frataxin expression
WO2013192233A1 (fr) Composés et procédé pour capture cellulaire améliorée de composés antisens
US20190382760A1 (en) Antisense compounds targeting genes associated with fibronectin
EP3750997A1 (fr) Composés antisens et leurs utilisations
WO2015187989A1 (fr) Composés antisens ciblant le récepteur 2 de l'apolipoprotéine e
WO2016061263A1 (fr) Composés antisens et leurs utilisations
WO2019169243A1 (fr) Composés et procédés pour la modulation de la protéine précurseur de l'amyloïde bêta
WO2017218884A1 (fr) Combinaisons pour la modulation de l'expression de smn
WO2014071078A1 (fr) Composés antisens et leurs utilisations
US10287584B2 (en) Compounds and methods for the modulation of COMP
US9487780B2 (en) Antisense compounds targeting genes associated with fibronectin

Legal Events

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

Ref document number: 15850666

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15850666

Country of ref document: EP

Kind code of ref document: A1