WO2007027775A2 - Methods for use in modulating mir-122a - Google Patents

Methods for use in modulating mir-122a Download PDF

Info

Publication number
WO2007027775A2
WO2007027775A2 PCT/US2006/033866 US2006033866W WO2007027775A2 WO 2007027775 A2 WO2007027775 A2 WO 2007027775A2 US 2006033866 W US2006033866 W US 2006033866W WO 2007027775 A2 WO2007027775 A2 WO 2007027775A2
Authority
WO
WIPO (PCT)
Prior art keywords
mir
antisense compound
antisense
nucleic acid
nucleosides
Prior art date
Application number
PCT/US2006/033866
Other languages
French (fr)
Other versions
WO2007027775A3 (en
Inventor
Christine Esau
Sanjay Bhanot
Original Assignee
Isis Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=37809465&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2007027775(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Isis Pharmaceuticals, Inc. filed Critical Isis Pharmaceuticals, Inc.
Priority to AT06813949T priority Critical patent/ATE494372T1/en
Priority to AU2006284855A priority patent/AU2006284855B2/en
Priority to CA2620856A priority patent/CA2620856C/en
Priority to JP2008529232A priority patent/JP5523705B2/en
Priority to EP06813949.2A priority patent/EP1931782B2/en
Priority to DE602006019455T priority patent/DE602006019455D1/en
Publication of WO2007027775A2 publication Critical patent/WO2007027775A2/en
Publication of WO2007027775A3 publication Critical patent/WO2007027775A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • C12N2310/111Antisense spanning the whole gene, or a large part of it
    • 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
    • C12N2310/113Antisense targeting other non-coding nucleic acids, e.g. antagomirs
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified C
    • C12N2310/33415-Methylcytosine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/341Gapmers, i.e. of the type ===---===
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/345Spatial arrangement of the modifications having at least two different backbone 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
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate

Definitions

  • the present invention provides methods for the modulation of small non-coding RNAs, in particular miR- 122a.
  • Methods are provided for the treatment of cardiovascular or metabolic diseases characterized by elevated serum total cholesterol, elevated serum LDL-cholesterol, or elevated serum triglycerides, through the administration of an antisense compound which inhibits the levels or activity of miR- 122a.
  • Further provided are methods for reducing hepatic steatosis or liver tissue triglyceride accumulation through the administration of an antisense compound which inhibits the levels or activity of miR-122a.
  • Such methods employ antisense compounds which hybridize with or sterically interfere with nucleic acid molecules comprising or encoding miR- 122a.
  • antisense compounds may include one or more modifications thereon, which may improve the activity, stability, or nuclease resistance of the antisense compound. These modified antisense compounds are used as single compounds or in compositions, including pharmaceutical compositions, to inhibit miR- 122a.
  • MicroRNAs are small (approximately 21-24 nucleotides in length, these are also known as "mature” miRNA), non-coding RNA molecules encoded in the genomes of plants and animals. These highly conserved, endogenously expressed RNAs are believed to regulate the expression of genes by binding to the 3'-untranslated regions (3'-UTR) of specific mRNAs. MiRNAs may act as key regulators of cellular processes such as cell proliferation, cell death (apoptosis), metabolism, and cell differentiation. On a larger scale, miRNA expression has been implicated in early development, brain development, disease progression (such as cancers and viral infections).
  • miRNAs appear to originate from long endogenous primary miRNA transcripts (also known as pri-miRNAs, pri-mirs, pri-miRs or pri-pre- miRNAs) that are often hundreds of nucleotides in length (Lee, et al., EMBO J., 2002, 21(17), 4663- 4670).
  • miRNAs In mammals, only a few miRNAs have been assigned any function, although they are predicted to regulate a large percentage of genes, with estimates based on bioinformatic target prediction ranging as high as 30% (Lewis et al., 2005). Based on the early studies in invertebrates, miRNAs are expected to have similar roles in developmental regulation and cell differentiation in mammals, and roles for miRNAs in cardiogenesis (Zhao et al., 2005) and lymphocyte development (Chen et al., 2004) have been demonstrated.
  • miRNA genes are often found in genomic regions linked to cancer (Calin et al., 2004; McManus, 2003) and a study correlating miRNA expression profiles with developmental lineage and differentiation state of tumors (Lu et al., 2005).
  • a potential role for miRNAs in metabolic pathways has been suggested by studies implicating miRNAs in regulation of adipocyte differentiation (Esau et al., 2004) and glucose-stimulated insulin secretion from pancreatic islet cells (Poy et al., 2004).
  • miR-122a is expressed in the developing liver (Chang et al., 2004) and at high levels in the adult liver, where it makes up 70% of all miRNA (Chang et al., 2004; LagosQuintana et al., 2002). It is one of many tissue-specific microRNAs thought to be important for establishing patterns of gene expression which may be responsible for maintaining the differentiated state of a tissue (Lagos-Quintana et al., 2002; Lim et al., 2005). miR-122a was also reported to enhance replication of HCV through a novel mechanism which is not yet understood, making it also a potential therapeutic target for HCV infection (Jopling et al., 2005).
  • the present invention therefore provides antisense compounds and methods useful for modulating miR- 122a, to achieve clinically desirable changes in cholesterol and lipid profiles in animals.
  • the present invention provides methods of lowering a serum indicator of cardiovascular disease risk, which include elevated serum total cholesterol elevated serum LDL-cholesterol, and elevated serum triglyceride levels, comprising selecting an animal having an elevate serum indicator of cardiovascular disease, and administering to the animal an antisense compound targeted to miR-122a.
  • the present invention provides methods of lowering serum cholesterol in an animal comprising selecting an animal having elevated serum cholesterol levels, and then administering to the animal a therapeutically effective amount of an antisense compound essentially complementary to a miR-122a nucleic acid.
  • the serum cholesterol levels may be total serum cholesterol levels or serum LDL- cholesterol levels.
  • the present invention further provides a method of lowering triglyceride levels in an animal comprising selecting an animal having elevated triglyceride levels, and then administering to the animal a therapeutically effective amount of an antisense compound essentially complementary to a miR- 122a nucleic acid.
  • the triglyceride levels may be serum triglyceride levels or liver tissue triglyceride levels.
  • a method of lowering serum lipoproteins in an animal comprising selecting an animal having elevated serum lipoproteins and administering to the animal a therapeutically effective amount of an antisense compound essentially complementary to a miR-122a nucleic acid.
  • the serum lipoprotein may be apolipoprotein B-IOO.
  • the present invention provides methods of reducing hepatic steatosis in an animal comprising selecting an animal having hepatic steatosis and administering to the animal a therapeutically effective amount of an antisense compound essentially complementary to a miR-122a nucleic acid.
  • the hepatic steatosis may be steatohepatitis or non-alcoholic steatohepatitis.
  • the present invention provides methods of modulating a metabolic pathway in an animal, comprising administering to an animal a therapeutically effective amount of an antisense compound essentially complementary to a miR- 122a nucleic acid.
  • the metabolic pathway is selected from lipogenesis, fatty acid oxidation, fatty acid synthesis rate, or sterol synthesis.
  • the methods provided reduce lipogenesis, reduce fatty acid synthesis rate, reduce sterol synthesis, or increase fatty acid oxidation.
  • a method of improving hepatic function in an animal comprising contacting an animal with a therapeutically effective amount of an antisense compound essentially complementary to a miR-122a nucleic acid.
  • the improvement in hepatic function is measured by measuring the levels of plasma transaminases, wherein a decrease in plasma transaminases indicate an improvement in hepatic function.
  • the animal is treated through the administration of a therapeutically effective amount of an antisense compound essentially complementary to a miR- 122a nucleic acid.
  • the antisense compound is essentially fully complementary to a miR-122a target nucleic acid.
  • the antisense compound is fully complementary to a miR-122a target nucleic acid.
  • the antisense compound comprises at plurality of sugar modified nucleosides.
  • the plurality of sugar modified nucleosides may, in some embodiments comprise 2 distinct sugar modifications.
  • the sugar modified nucleosides may further comprise at least one bicyclic sugar modification.
  • each nucleoside of the plurality of sugar modified nucleosides may comprise a 2'-MOE sugar modification.
  • the antisense compound may comprise at least one phosphorothioate internucleoside linkage.
  • the antisense compound may further comrise a least one 5-methylcytosine.
  • An antisense compound comprising the nucleobase sequence SEQ ID NO: 1 may be used in any of the aforementioned methods.
  • an antisense compound consisting of the nucleobase sequence of SEQ ID NO: 2. may be used in any of the aforementioned methods.
  • the antisense compound may comprise ISIS 327895 or ISIS 387574.
  • the present invention provides the use of an antisense compound essentially fully complementary to a miR-122 nucleic acid for the preparation of a medicament for lowering a serum indicator of cardiovascular disease risk wherein the serum indicator of cardiovascular disease risk is selected from elevated serum cholesterol levels, elevated serum triglyceride levels, or elevated lipoprotein levels.
  • the elevated serum cholesterol may be elevated LDL-cholesterol, or elevated serum total cholesterol.
  • the elevated serum lipoprotein may be elevated serum ApoB-100.
  • the present invention further provides the use of an antisense compound essentially fully complementary to a miR-122a nucleic acid for the preparation of a medicament for reducing hepatic steatosis.
  • the hepatic steatosis is steatohepatitis or non-alcoholic steatohepatitis.
  • the present invention additionally provides the use of an antisense compound essentially fully complementary to a miR-122a nucleic acid in the preparation of a medicament to treat a disease or disorder selected from diabetes, obesity, hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, hyperfattyacidemia, nonalcoholic fatty liver disease, non alcoholic steatohepatitis, or metabolic syndrome.
  • a disease or disorder selected from diabetes, obesity, hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, hyperfattyacidemia, nonalcoholic fatty liver disease, non alcoholic steatohepatitis, or metabolic syndrome.
  • the antisense compound used in the preparation of the medicament is 21 to 23 nucleosides in length. Further, the antisense compound may be fully complementary to a miR-122a nucleic acid. Additionally, the antisense compound may comprise a plurality of 2 '-sugar modified nucleosides. The antisense compound may further comprise at least one bicyclic sugar modified nucleoside. Additionally, the antisense compound comprises the nucleobase sequence of SEQ ID NO: 1. Alternatively, the antisense compound consists of the nucleobase sequence of SEQ ID NO: 2. The antisense compound used in the preparation of the medicament may be uniformly comprised of 2'-MOE sugar modified nucleosides. The antisense compound may further comprise at least W 2
  • the antisense compound may additionally comprise at least one 5-methylcytosine.
  • the present invention provides antisense compounds that hybridize to and inhibit the levels, activity or expression of a miR-122a target nucleic acid, i.e. the antisense compounds are targeted to a miR-122a nucleic acid.
  • the antisense compounds are chemically modified to enhance stability and affinity for a miR-122a nucleic acid.
  • the antisense compounds comprise a plurality of sugar modified nucleosides.
  • the plurality of sugar modified nucleosides comprises at least 2 distinct sugar modified nucleosides.
  • the plurality of sugar modified nucleosides comprises at least one bicyclic sugar modified nucleoside.
  • the antisense compounds are uniformly modified such that each nucleoside bears a 2'-MOE modification.
  • the antisense compound comprises the nucleobase sequence CAAACACCATTGTCACACTCCA (SEQ ID NO: 1).
  • the antisense compound has the nucleobase sequence ACAAACACCATTGTCACACTCCA (SEQ ID NO: 2).
  • the antisense compound is ISIS 327895 or ISIS 387574.
  • the present invention provides methods of administering antisense compounds targeted to a miR-122a nucleic acid to an animal to lower serum cholesterol, serum LDL-cholesterol, or serum triglycerides.
  • the present invention also provides methods of reducing hepatic steatosis, liver triglyceride levels and liver weights. Additionally provided are methods of improving liver function, which is evaluated, for example, by decreases in serum transaminases. Also provided are methods of modulating the expression of a miR-122a mRNA target, such as ALDO A.
  • the antisense compounds of the invention are also used for the modulation of metabolic pathways, for example, to lower sterol synthesis, to reduce lipogenesis, reduce fatty acid synthesis rate, reduce sterol synthesis, or increase fatty acid oxidation.
  • the present invention provides methods for the treatment of diseases or conditions characterized by elevated serum cholesterol levels, elevated serum triglyercide levels, or elevated liver triglyceride levels. These diseases or conditions may be further characterized by compromised liver function as measured by increases in plasma transaminases.
  • the methods provided herein are useful for the treatment of diseases or conditions including hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, or hyperfattyacidemia.
  • the methods are useful for the treatment of hepatic steatosis or non-alcholic fatty liver disease.
  • the steatosis is steatohepatitis.
  • the steatotis is NASH.
  • a further embodiment includes the use of the methods provided for the treatment of metabolic sydrome.
  • the methods comprise the administration of antisense compounds targeted to miR-122a to an animal, particularly a human, having or susceptible to a disease or condition including, but not limited to, hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, hyperfattyacidemia, steatohepatitis, non-alcoholic steatohepatitis, non-alcholic fatty liver disease, or metabolic syndrome.
  • the disease or condition is a cardiovascular disease or a metabolic disease.
  • the disease or condition is treated or ameliorated through the administration to the human of a therapeutically effective amount of an antisense compound of the invention.
  • the onset of the disease or condition is delayed or prevented through the administration to the human of a prophylactically effective amount of an antisense compound of the invention.
  • Embodiments described herein additionally relate to the use of an antisense compound targeted to a miR- 122a nucleic acid in the preparation of a medicament for the treatment of a disease or condition including hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, or hyperfattyacidemia.
  • the medicament is used for the treatment of hepatic steatosis, which may be steatohepatitis or NASH, or non-alcoholic fatty liver disease.
  • the medicament is used for the treatment of metabolic syndrome.
  • the medicament is used for the improvement of liver function.
  • the medicament may, in some embodiments, be administered parenterally, which includes intravenous administration and subcutaneous administration.
  • Antisense compounds of the invention modulate the levels, activity, or expression of a miR- 122a nucleic acid by hybridizing to a miR-122a nucleic acid ⁇ i.e., antisense compounds target the miR- 122a nucleic acids) and thereby interfering with its endogenous function.
  • the hybridization of an antisense compound targeting a miR-122a nucleic acid may trigger the degradation, cleavage, and/or sequestration of the miR- 122a nucleic acid, each of which interferes with the activity of the miR- 122a nucleic acid.
  • the hybridization of an antisense compound targeting miR- 122a may also interfere with the activity of miR-122a by steric occlusion.
  • the inhibition of miR-122a levels, activity or expression that results from an antisense compound hybridizing to a miR- 122a nucleic acid is referred to as "antisense inhibition.”
  • modulation of a miR- 122a nucleic acid means either an increase (stimulation) or a decrease (inhibition) in the level, activity, or expression of a miR- 122a nucleic acid.
  • stimulation As microRNAs negatively regulate protein-coding nucleic acids, inhibition of a microRNA generally results in stimulation of expression of one or more protein-coding nucleic acids regulated by the microRNA.
  • Inhibition of miR-122a is a preferred form of modulation of a miR-122a nucleic acid.
  • Stimulation is a preferred form of modulation of mRNAs regulated by miR-122a, such as ALDO A.
  • the level, activity or expression of a miR- 122a nucleic acid is inhibited to a degree that results in a phenotypic change, such as lowered serum cholesterol or reduced hepatic steatosis.
  • miR-122a nucleic acid level indicates the abundance of a miR-122a nucleic acid in a sample, such as an animal cell or tissue.
  • miR- 122a level may also indicate the relative abundance of a miR-122a nucleic acid in an experimental sample (e.g., tissue from an animal treated with an antisense compound targeted to miR-122a) as compared to a control sample (e.g., tissue from an untreated animal).
  • miR-122a activity refers to the regulation of a protein-coding nucleic acid by miR-122a.
  • miR-122a expression refers to the process by which mature miR-122a is derived from a DNA sequence that codes for miR-122a, which includes several steps, for example, transcription, Drosha processing, and Dicer processing. miR-122a expression may be regulated at a single step of this process, or at multiple steps.
  • small non-coding RNA is used to encompass, without limitation, a polynucleotide molecule ranging from about 17 to about 450 nucleosides in length, which can be endogenously transcribed or produced exogenously (chemically or synthetically), but is not translated into a protein.
  • primary miRNAs also known as pri-pre-miRNAs, pri-miRs and pri- miRNAs
  • pri-pre-miRNAs also known as pri-pre-miRNAs, pri-miRs and pri- miRNAs
  • the primary miRNA is processed by Drosha to yield a pre-miRNA (also known as pre-mirs and foldback miRNA precursors), which ranges from around 50 nucleosides to around 110 nucleosides in length.
  • the pre-miRNA is in turn processed by Dicer to yield a miRNA (also known as microRNAs, Mirs, miRs, mirs, and mature miRNAs), which ranges from 19 to 24 nucleosides in length.
  • Small non-coding RNAs may include isolated single-, double-, or multiple-stranded molecules, any of which may include regions of intrastrand nucleobase complementarity, said regions capable of folding and forming a molecule with fully or partially double-stranded or multiple-stranded character based on regions of perfect or imperfect complementarity.
  • a "miR-122a nucleic acid” includes pri-miR-122a, pre-miR-122a, and miR- 122a.
  • pri-miR-122a is a primary miRNA
  • pre-miR-122a is a pre- miRNA
  • miR-122a is a mature miRNA.
  • mature miR-122a and “miR-122a” may be used interchangably herein.
  • miR-122a or a precursor thereof encompasses miR-122a, pre-miR-122a, pri- miR-122a, or a primary RNA transcript from which miR-122a or its precursors are derived.
  • miR-122a target nucleic acids include ⁇ ri-miR-122a, pre-miR-122a, and miR-
  • miR-122a i.e. mature miR-122a
  • miR-122a/b Tuschl
  • miR-122a/b Tuschl
  • miR-122a/b Tuschl
  • antisense compounds of the invention may target either miR-122a or miR-122a/b Tuschl.
  • miR-122a seed sequence refers to nucleosides 2 through 8 from the 5'-end of the miR-122a sequence.
  • the miR-122a seed sequence is 5'-GGAGUGU-3'
  • antisense compounds known as "miR-122a mimics", wherein the antisense compounds include one or more modifications or structural elements or motifs that render the compound capable of mimicking or replacing miR-122a or a precursor thereof.
  • antisense compounds targeting miR-122a can be harnessed by those of skill in the art for therapeutic uses. Numerous clinical trials testing antisense compounds to other targets in a variety of therapeutic areas are presently underway.
  • a "human in need of treatment” or “subject in need of treatment” includes humans or subjects diagnosed as having or being susceptible to a disease or condition that can be treated, ameliorated, or prevented by the administration of an antisense compound targeted to a miR-122a nucleic acid.
  • diseases or conditions include, but are not limited to, hypercholesterolemia, hypertriglyceridemia, hyperfattyacidemia, or hyperlipidemia, nonalcoholic fatty liver disease, hepatic steatosis (including non- alchoholic steatohepatitis and steatohepatitis), and metabolic syndrome.
  • Subjects in need of treatment may include subjects diagnosed with HCV who also have one or more of the diseases or conditions mentioned herein, such as, for example, hypercholesterolemia or hepatic steatosis. Diagnosis of any of the aforementioned diseases or conditions routinely occurs in a clinical setting, for example, by a physician. Humans or subjects in need of treatment include those who are identified by a medical professional, such as a physician, to have risk factors for cardiovascular disease and/or risk determinants of metabolic syndrome.
  • a "therapeutically effective amount” is an amount of an antisense compound targeted to a miR-122a nucleic acid that, when administered to a human having one or more of the aforementioned diseases or conditions, results in a clinically desirable outcome.
  • Such clinically desirable outcomes may include, without limitation, lowered serum total cholesterol, lowered serum LDL- cholesterol, lowered serum triglycerides, lowered liver triglycerides, reduced steatosis, improved liver function, or increased fatty acid oxidation.
  • a “prophylactically effective amount” is an amount of an antisense compound targeted to a miR-122a nucleic acid that, when administered to a human, prevents or reduces the susceptibility of the human to one or more of the aforementioned diseases or conditions. Prevention or reduction of the susceptibility of the human to the aforementioned diseases or conditions may be accomplished by preventing elevated serum total cholesterol, elevated serum LDL-cholesterol, elevated serum triglycerides, elevated liver triglycerides, hepatic steatosis, aberrant liver function, or reductions in fatty acid oxidation.
  • prevention means to delay or forestall onset or development of a condition or disease for a period of time, preferably for weeks, months or years.
  • aboration means a lessening of the severity of a condition or a disease, as evidenced by an improvement of one or more of the surrogate indicators disclosed herein. The improvement of such indicators may be determined by subjective or objective measures which are known to those skilled in the art.
  • treatment means to administer a composition of the invention to effect an alteration or improvement of the disease or condition.
  • Prevention, amelioration, and/or treatment may require administration of multiple doses of an antisense compound targeted to a miR-122a nucleic acid to alter the course of the condition or disease.
  • a single antisense compound may be used in a single individual for the purposes of achieving any combination of prevention, amelioration, or treatment of a condition or disease, and such combinations may be pursued concurrently or sequentially.
  • Antisense compounds of the present invention may, in some instances, be administered with other treatments.
  • a suitable method of administration is parenteral administration, which includes, for example, intravenous administration, subcutaneous administration, and intraperitoneal administration.
  • the present invention provides the use of an antisense compound targeted to a miR-122a nucleic acid for the preparation of a medicament that is administered parenterally, for example, intravenously or subcutaneously.
  • the term "serum marker of cardiovascular disease risk” includes risk factors recognized by medical professionals, including those set forth by the National Cholesterol Education Program (NCP), such as elevated serum cholesterol, elevated serum triglycerides, or elevated serum lipoprotein. Serum cholesterol further comprises serum total cholesterol and serum LDL-cholesterol. Serum lipoproteins further comprise serum ApoB-100 protein. In a clinical setting, serum markers of cardiovascular disease risk are measured to determine the need for treatment or prevention of a disease or condition described herein.
  • the guidelines for lipid lowering therapy were established in 2001 by Adult Treatment Panel III (ATP m) of the NCEP, and udpated in 2004 (Grundy et al., Circulation, 2004, 110, 227-239).
  • the guidelines include determining LDL-cholesterol, total cholesterol, and HDL-cholesterol levels (i.e., determining lipoprotein levels).
  • LDL- cholesterol levels of 130-159 mg/dL, 160-189 mg/dL, and greater than or equal to 190 mg/dL are considered borderline high, high, and very high, respectively.
  • Total cholesterol levels of 200-239 and greater than or equal to 240 are considered borderline high and high, respectively.
  • HDL-cholesterol levels of less than 40 are considered low.
  • Serum triglyceride levels of 150-199, 200-499, and greater than or equal to 500 are considered borderline high, high, and very high, respectively.
  • the serum cholesterol level (e.g. LDL-cholesterol, total cholesterol) and/or serum triglyceride level at which treatment is initiated depends upon the presence of clinical atherosclerotic disease that confers a high risk for coronary heart disease, such as clinical coronary heart disease, symptomatic carotid artery disease, peripheral arterial disease, and or abdominal aortic aneurysm, as well as additional risk factors, such as cigarette smoking, hypertension, low HDL-cholesterol, family history of coronary heart disease, and age.
  • a subject' response to treatment is used by a physician to determine the amount and duration of treatment.
  • the NCEP ATP III has also established criteria for the diagnosis of metabolic syndrome when three or more of five risk determinants are present.
  • “Metabolic syndrome” is defined as a clustering of lipid and non-lipid cardiovascular risk factors of metabolic origin. It is closely linked to the generalized metabolic disorder known as insulin resistance.
  • the five risk determinants are abdominal obesity defined as waist circumference of greater than 102 cm for men or greater than 88 cm for women, triglyceride levels greater than or equal to 150 mg/dL, HDL cholesterol levels of less than 40 mg/dL for men and less than 50 mg/dL for women, blood pressure greater than or equal to 130/85 mm Hg and fasting glucose levels greater than or equal to 110 mg/dL. These determinants can be readily measured in clinical practice (JAMA, 2001, 285, 2486-2497).
  • a physician may modify cardiovascular risk determination for individual patients, in cases where more or less aggressive therapy is needed.
  • One of skill will also understand the scope of the invention includes the practice of the methods herein as applied to any altered guidelines provided by the NCEP, or other entities that establish guidelines for physician's used in treating any of the diseases or conditions listed here, for determining cardiovascular disease risk and diagnosing metabolic syndrome.
  • the embodiments herein provide the use of an antisense compound targeted to a miR-122a nucleic acid in the preparation of a medicament for lowering a serum indicator of cardiovascular disease risk, wherein the serum indicator is selected from elevated serum total cholesterol, elevated serum LDL- cholesterol, or elevated serum triglycerides.
  • surrogate indicators are used to evaluate phenotypic changes that result from antisense inhibition of miR- 122a.
  • Surrogate indicators are often found in the serum, or alternatively in the plasma, of an animal. Methods of obtaining serum or plasma samples for analysis and methods of preparation of the serum samples to allow for analysis are well known to those skilled in the art. With regard to measurements of lipoproteins, cholesterol, and triglyceride, the terms "serum” and "plasma” are herein used interchangeably.
  • surrogate indicators include serum indicators of cardiovascular disease risk, such as serum LDL-cholesterol levels, and serum total cholesterol levels, and serum triglyceride levels. Serum indicators further include serum transaminase levels.
  • serum LDL-cholesterol levels refer to the amounts of cholesterol that are carried in serum LDL particles, and are typically measured as mg/dL or nmol/L.
  • serum total cholesterol levels also expressed as mg/dL, refers to the sum of all different types of serum cholesterol, including, among others, LDL-cholesterol and HDL-cholesterol.
  • Preferred alterations in surrogate indicators of miR-122a inhibition include lowered serum
  • LDL-cholesterol levels lowered serum triglyceride levels, lowered lipoprotein levels.
  • “Serum lipoproteins” include without limitation apolipoprotein B (ApoB-100), low-density lipoprotein (LDL), and very low-density lipoprotein (VLDL).
  • Methods for the detection of hepatic steatosis are well known in the art, and include magnetic resonance imaging, computed tomography, and ultrasonagraphy. Methods used for the detection of hepatic steatosis may also be used to monitor the prevention, amelioration and/or treatment of the diseases and disorders disclosed herein.
  • the term "improved hepatic function" or “improved liver function” refers to the improvement in the regular functions performed by the liver of an animal.
  • a test for liver function involves the measurement of serum transaminases, which are additional surrogate indicators, wherein a reduction in serum transaminases such as alanine aminotransferase or aspartamine aminotransferase indicates an improvement in liver function.
  • “Serum transaminase levels” refer to the abundance of transaminase in the serum (typically expressed as units /dL), and include “serum alanine aminotransferase levels” and “serum aspartamine aminotransferase levels.” Increases in these enzymes are frequently associated with inflammation of the liver or liver cell death and, in some cases, can be caused by a condition such as hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, or hyperlipidemia. Reductions in serum transaminases are clinically desirable and are useful for the prevention, amelioration and/or treatment of the diseases and disorders disclosed herein.
  • nonalcoholic fatty liver disease encompasses a disease spectrum ranging from simple triglyceride accumulation in hepatocytes (hepatic steatosis) to hepatic steatosis with inflammation (steatohepatitis), fibrosis, and cirrhosis.
  • NASH nonalcoholic steatohepatitis
  • a second physiological insult capable of inducing necrosis, inflammation, and fibrosis is required for development of NASH.
  • Candidates for the second insult can be grouped into broad categories: factors causing an increase in oxidative stress and factors promoting expression of proinflammatory cytokines.
  • liver triglycerides lead to increased oxidative stress in hepatocytes of animals and humans, indicating a potential cause-and-effect relationship between hepatic triglyceride accumulation, oxidative stress, and the progression of hepatic steatosis to NASH (Browning and Horton, J. Clin. Invest., 2004, 114, 147- 152).
  • Hypertriglyceridemia and hyperfattyacidemia can cause triglyceride accumulation in peripheral tissues (Shimamura et al., Biochem. Biophys. Res. Commun., 2004, 322, 1080-1085).
  • additional clinically desirable outcomes include reductions in hepatic steatosis, steatohepatisis, fibrosis or cirrhosis.
  • the present invention provides the use of an antisense compound targeted to a miR-122a nucleic acid in the preparation of a medicament for lowering serum cholesterol, serum triglycerides or serum lipoproteins in an animal having HCV.
  • antisense compounds and compositions of the present invention can additionally be utilized for research and drug discovery.
  • antisense compounds of the present invention are used to interfere with the normal function of miR-122a target nucleic acids.
  • Expression patterns within cells or tissues treated with one or more antisense compounds or compositions of the invention are compared to control cells or tissues not treated with the antisense compounds or compositions and the patterns produced are analyzed for differential levels of nucleic acid expression as they pertain, for example, to disease association, signaling pathway, cellular localization, expression level, size, structure or function of the genes examined. These analyses can be performed on stimulated or unstimulated cells and in the presence or absence of other compounds that affect expression patterns.
  • antisense compounds targeted to miR-122a were used to elucidate the metabolic pathways that are affected by miR-122a regulation.
  • antisense compounds of the present invention are used to elucidate relationships that exist between miR-122a, or a precursor thereof and a disease state, phenotype, or condition. These methods include detecting or modulating a miR-122a nucleic acid comprising contacting a sample, tissue, cell, or organism with the antisense compounds and compositions of the present invention, measuring the levels of the target miR-122a nucleic acid and/or the levels of downstream gene products including rnRNA or proteins encoded thereby, and further evaluating phenotypic changes at some time after treatment.
  • antisense compounds targeted to miR-122a were used to effect phenotypic changes such as lowered serum cholesterol, lowered liver triglycerides, and reduced hepatic steatosis.
  • oligomeric compound(s) refers to polymeric structures which are capable of hybridizing to at least a region of an RNA molecule.
  • an oligomeric compound is "antisense" to a target nucleic acid when, written in the 5' to 3' direction, it comprises the reverse complement of the corresponding region of the target nucleic acid.
  • antisense compounds include, without limitation, oligonucleotides
  • an antisense compound comprises a backbone of linked monomelic subunits (sugar moieties) where each linked monomelic subunit is directly or indirectly attached to a heterocyclic base moiety.
  • Modifications to antisense compounds encompass substitutions or changes to internucleoside linkages, sugar moieties, or heterocyclic base moieties, such as those described below.
  • the term "modification" includes substitution and/or any change from a starting or natural nucleoside or nucleotide.
  • Modified antisense compounds are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target, increased stability in the presence of nucleases, or increased inhibitory activity.
  • Antisense compounds are often defined in the art to comprise the further limitation of, once hybridized to a target, being able to induce or trigger a reduction in target gene expression or target gene activity. La one embodiment, the antisense compounds trigger a reduction in the levels, activity or expression of a miR- 122a nucleic acid target.
  • Antisense compounds are routinely prepared linearly but can be joined or otherwise prepared to be circular and may also include branching. Separate antisense compounds can hybridize to form double stranded compounds that can be blunt-ended or may include overhangs on one or both termini.
  • the antisense compounds in accordance with this invention comprise from 15 to 30 nucleosides in length, i.e., from 15 to 30 linked nucleosides.
  • 15 to 30 linked nucleosides One of skill in the art will appreciate that this embodies antisense compounds of 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleosides in length.
  • the antisense compounds of the invention are 17 to 25 nucleosides in length, as exemplified herein.
  • the antisense compounds of the invention are 19, 20, 21, 22 or 23 nucleosides in length.
  • An antisense compound and the target nucleic acid are "fully complementary” to each other when each nucleobase of the antisense compound is complementary to an equal number of nuclebases in the target nucleic acid.
  • an antisense compound 23 nucleosides in length targeted to miR-122a is fully complementary to miR-122a when each of the 23 nucleobases in the antisense compound is complementary to miR-122a.
  • the antisense compound and the target nucleic acid are "essentially fully complementary” to each other when the degree of precise base pairing permits stable and specific binding between the antisense compound and a target nucleic acid, so that the antisense compound inhibits the level, activity or expression of the target nucleic acid.
  • Antisense compounds having one or two non- complementary nucleobases with respect to a miR-122a target nucleic acid are considered essentially fully complementary. “Sufficiently complementary” may be used in place of “essentially fully complementary”.
  • “hybridization” means the pairing of nucleobases of an antisense compound with corresponding nucleobases in a target nucleic acid.
  • the mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between corresponding nucleobases.
  • adenine and thymine are complementary nucleobases that pair through the formation of hydrogen bonds. Both natural and modified nucleobases are capable of participating in hydrogen bonding. Hybridization can occur under varying circumstances.
  • sequence of the antisense compound need not be fully complementary to that of its target nucleic acid to be active in inhibiting the activity of the target nucleic acid.
  • an antisense compound may hybridize over one or more segments such that intervening or adjacent segments are not involved in the hybridization (e.g., a bulge, a loop structure or a hairpin structure).
  • there are "non-complementary" positions, also known as “mismatches", between the antisense compound and the target nucleic acid and such non-complementary positions may be tolerated between an antisense compound and the target nucleic acid provided that the antisense compound remains specifically hybridizable to the target nucleic acid.
  • non-complementary nucleobase means a nucleobase of an antisense compound that is unable to undergo precise base pairing with a nucleobase at a corresponding position in a target nucleic acid.
  • the terms “non- complementary” and “mismatch” are interchangable. Up to 3 non-complementary nucleobases are often tolerated in an antisense compound without causing a significant decrease in the ability of the antisense compound to modulate the activity, level or function of a target nucleic acid.
  • the antisense compound contains no more than 2, or no more than 1 , non-complementary nucleobases with respect to a miR-122a target nucleic acid.
  • an antisense compound 23 nucleosides in length, 22 of which are able to undergo precise base pairing with nucleobases in corresponding positions in a target nucleic acid, and one of which is not able to undergo such base pairing is considered to have one non-complementary nucleobase.
  • the location of such a non-complementary nucleobase at the 5 ' end or 3' end of the antisense compound is preferred, however, the non-complementary nucleobase may be at any position in the antisense compound.
  • non-complementary nucleobases are preferred outside of the region of the antisense compound which is complementary to the seed sequence of miR-122a. When two or more non-complementary nucleobases are present, they may be contiguous (i.e. linked) or non-contiguous.
  • the antisense compounds comprise at least 90% sequence complementarity to a miR-122a target nucleic acid. In further embodiments of the invention, the antisense compounds comprise at least 95% sequence complementarity to a miR-122a target nucleic acid. For example, an antisense compound in which 22 of 23 nucleobases of the antisense compound are complementary (i.e. one nucleobase is non-complementary) to a miR-122a nucleic acid would represent 95.6 % complementarity. Likewise, an antisense compound in which 21 of the 23 nucleobases are complementary (i.e.
  • % complementarity Percent complementarity of an antisense compound with a region of a target nucleic acid can be determined routinely by those having ordinary skill in the art, and may be accomplished using BLAST programs (basic local alignment search tools) and PowerBLAST programs known in the art (Altschul et al., J. MoL Biol, 1990, 215, 403-410; Zhang and Madden, Genome Res., 1997, 7, 649-656).
  • stringent hybridization conditions or “stringent conditions” refers to conditions under which an antisense compound of the invention will hybridize to its target sequence, but to a minimal number of other sequences.
  • Stringent conditions are sequence-dependent and will vary with different circumstances and in the context of this invention; "stringent conditions” under which antisense compounds hybridize to a target sequence are determined by the nature and composition of the antisense compounds and the assays in which they are being investigated.
  • Antisense compounds may have a defined percent identity to a SEQ ID NO, or an antisense compound having a specific ISIS number.
  • antisense compound need not have an identical sequence to those described herein to function similarly to the antisense compounds described herein. Shortened or truncated versions of antisense compounds taught herein have one, two or more nucleosides deleted, and fall within the scope of the invention.
  • the deleted nucleosides may be adjacent to each other, for example, in an antisense compound having two nucleosides truncated from the 5' end (5' truncation), or alternatively from the 3' end (3' truncation), of the antisense compound.
  • the delelted nucleosides may be dispersed through out the antisense, for example, in an antisense compound having one nucleoside deleted from the 5' end and one nucleoside deleted from the 3' end.
  • antisense compounds taught herein i.e. antisense compounds having one or more additional nucleosides relative to an antisense compound disclosed herein.
  • the added nucleosides may be adjacent to each other, for example, in an antisense compound having two nucleosides added to the 5' end (5' addition), or alternatively to the 3' end (3' addition), of the antisense compound.
  • the added nucleosides may be dispersed throughout the antisense compound, for example, in an antisense compound having one nucleoside added to the 5' end and one nucleoside added to the 3' end.
  • Percent identity of an antisense compound is calculated according to the number of nucleobases that are identical to the SEQ ID NO or antisense compound to which it is being compared. Such calculations are well within the ability of those skilled in the art. For example, a 21 nucleoside antisense compound having the same nucleobase sequence as nucleosides 2-21 of a 23 nucleoside antisense compound is 91.3% identical to the 23 nucleoside antisense compound. Alternatively, a 24 nucleoside antisense compound that differs in sequence only by the addition of one nucleoside relative to a 23 nucleoside antisense compound may have 100% identity over 23 nucleosides, but 95.8 % identity overall.
  • Targeting an antisense compound to a particular nucleic acid molecule, including a miR-122a nucleic acid, in the context of this invention, can be a multistep process.
  • the process usually begins with the identification of a target nucleic acid whose levels, expression or function is to be modulated.
  • the target nucleic acid is a miR-122a target nucleic acid.
  • the targeting process usually also includes determination of at least one target segment within a miR-122a target nucleic acid for the interaction to occur such that the desired effect, e.g., modulation of levels, activity, or expression, will result.
  • a target segment means a sequence of a miR- 122a nucleic acid to which one or more antisense compounds are complementary. Multiple antisense compounds complementary to a given target segment may or may not have overlapping sequences.
  • target site is defined as a sequence of a miR-122a nucleic acid to which one nucleobase sequence is complementary.
  • the nucleobase sequence of SEQ ID NO: 8 is complementary to target site 27 to 49 of SEQ ID NO: 5
  • the nucleobase sequence of SEQ ID NO: 2 is complementary to target site 29 to 51 of SEQ NO: 5. Nucleosides 27 to 51 of SEQ ID NO: 5 thus represent a target segment of SEQ ID NO: 5.
  • a target segment and target site will be represented by the same nucleobase sequence.
  • Target sites and target segments may also be found in an miRNA gene from which a pri-miR- 122a is derived, which may be found as a solitary transcript, or it may be found within a 5' untranslated region (5'UTR), within in an intron, or within a 3' untranslated region (3 TJTR) of a gene.
  • 5'UTR 5' untranslated region
  • 3 TJTR 3' untranslated region
  • Antisense compounds of the invention may be also be described as complementary to a portion of a target site.
  • a "portion" is defined as at least 18 contiguous nucleosides of a target site. In other embodiments, a portion is 19 or 20 contiguous nucleosides of a target site. In preferred embodiments, a portion is 21 or 22 contiguous nucleosides of a target site.
  • antisense compounds are designed to be sufficiently complementary to the target segments or target sites, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired effect.
  • the desired effect may include, but is not limited to modulation of the levels, expression or activity of the a miR-122a target nucleic acid. Desired effects further include phenotypic changes.
  • the antisense compounds of the invention may be in the form of single-stranded, double- stranded, circular or hairpin antisense compounds and may contain structural elements such as internal or terminal bulges or loops. Once introduced to a system, the antisense compounds of the invention may elicit or inhibit the action of one or more enzymes or proteins to effect modulation of the levels, W
  • a further non-limiting example involves the enzymes of the RISC complex.
  • Antisense compounds or compositions of the invention may be used to induce potent and specific modulation of gene function through interactions with or mimicry of small non-coding RNAs that are processed by the RISC complex.
  • These compounds include single-stranded antisense compounds that bind in a RISC complex, double-stranded antisense/sense pairs of antisense compounds, or single- stranded antisense compounds that include both an antisense portion and a sense portion.
  • Oligonucleotide Synthesis Antisense compounds and phosphoramidites are made by methods well known to those skilled in the art. Oligomerization of modified and unmodified nucleosides is performed according to literature procedures for DNA like compounds (Protocols for Oligonucleotides and Analogs, Ed. Agrawal (1993),
  • RNA oligomers can be synthesized by methods disclosed herein or purchased from various RNA synthesis companies such as for example
  • the antisense compounds used in accordance with this invention may be conveniently and routinely made through the well-known technique of solid phase synthesis.
  • Equipment for such synthesis is sold by several vendors including, for example, Applied
  • a 96-well plate format is particularly useful for the synthesis, isolation and analysis of oligonucleotides.
  • Antisense Compound Modifications Any of the antisense compounds taught herein may contain modifications which confer desirable pharmacokinetic and/or pharmacodynamic properties to the antisense compound including, but are not limited to, improved binding affinity, stability, charge, localization or uptake. Further, modified synthetic antisense compounds of the present invention may be designed to mimic endogenous small non- coding RNAs.
  • nucleoside is a base-sugar combination.
  • the base (also known as nucleobase) portion of the nucleoside is normally a heterocyclic base moiety.
  • the two most common classes of such heterocyclic bases are purines and pyrimidines.
  • Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a pentofuranosyl sugar, the phosphate group can be linked to the 2', 3' or 5' hydroxyl moiety of the sugar.
  • the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound.
  • the respective ends of this linear polymeric structure can be joined to form a circular structure by hybridization or by formation of a covalent bond.
  • linear compounds may have internal base (i.e. nucleobase) complementarity and may therefore fold in a manner as to produce a fully or partially double-stranded structure.
  • the phosphate groups are commonly referred to as forming the internucleoside linkages of the oligonucleotide.
  • the normal internucleoside linkage of RNA and DNA is a 3' to 5' phosphodiester linkage.
  • oligonucleotide refers generally to an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA), and may be used to refer to unmodified oligonucleotides or oligonucleotide analogs.
  • unmodified oligonucleotide refers generally to oligonucleotides composed of naturally occuring nucleobases, sugars, and covalent internucleoside linkages.
  • oligonucleotide analog refers to oligonucleotides that have one or more non-naturally occurring nucleobases, sugars, and/or internucleoside linkages.
  • non-naturally occurring oligonucleotides are often selected over naturally occurring forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for other oligonucleotides or nucleic acid targets, increased stability in the presence of nucleases, or increased inhibitory activity.
  • Modified Internucleoside Linkages such as, for example, enhanced cellular uptake, enhanced affinity for other oligonucleotides or nucleic acid targets, increased stability in the presence of nucleases, or increased inhibitory activity.
  • antisense compounds useful in this invention include oligonucleotides containing modified, i.e. non-naturally occurring, internucleoside linkages.
  • modified, i.e. non-naturally occurring, internucleoside linkages are often selected over naturally occurring forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for other oligonucleotides or nucleic acid targets and increased stability in the presence of nucleases.
  • Antisense compounds of the invention can have one or more modified internucleoside linkages.
  • oligonucleotides having modified internucleoside linkages include internucleoside linkages that retain a phosphorus atom and internucleoside linkages that do not have a phosphorus atom.
  • modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be "oligonucleosides”.
  • a suitable phosphorus-containing modified internucleoside linkage is the phosphorothioate internucleoside linkage.
  • Additional modified internucleoside linkages containing a phosphorus atom therein include, for example, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates, 5'- alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3 '-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3'-5' linkages, 2'-5' linlced analogs of these, and those having inverted polarity wherein one or more internu
  • Oligonucleotides having inverted polarity comprise a single 3' to 3' linkage at the 3'-most internucleoside linkage i.e. a single inverted nucleoside residue which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof).
  • Various salts, mixed salts and free acid forms are also included.
  • the MMI type internucleoside linkages are disclosed in the above referenced U.S. patent 5,489,677.
  • Amide internucleoside linkages are disclosed in the above referenced U.S. patent 5,602,240.
  • Modified internucleoside linkages that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages.
  • morpholino linkages formed in part from the sugar portion of a nucleoside
  • siloxane backbones sulfide, sulfoxide and sulfone backbones
  • formacetyl and thioformacetyl backbones methylene formacetyl and thioformacetyl backbones
  • riboacetyl backbones alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH 2 component parts.
  • U.S. patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S.: 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; 5,792,608; 5,646,269 and 5,677,439, each of which is herein incorporated by reference.
  • Antisense compounds can also include an "oligonucleotide mimetic", which refers to oligonucleotides in which only the furanose ring or both the furanose ring and the internucleoside linkage are replaced with novel groups. Replacement of only the furanose ring is also referred to in the art as being a sugar surrogate. The heterocyclic base moiety or a modified heterocyclic base moiety is maintained for hybridization with an appropriate target nucleic acid. Oligonucleotide mimetics can include antisense compounds containing peptide nucleic acid (PNA) modifications.
  • PNA peptide nucleic acid
  • PNA antisense compounds the sugar-backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone.
  • the nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.
  • Representative U.S. patents that teach the preparation of PNA antisense compounds include, but are not limited to, U.S.: 5,539,082; 5,714,331; and 5,719,262, each of which is herein incorporated by reference. Teaching of PNA antisense compounds can be found in Nielsen et al, Science, 1991, 254, 1497-1500. PNA has been modified to incorporate numerous modifications since the basic PNA structure was first prepared.
  • oligonucleotide mimetic Another class of oligonucleotide mimetic that has been studied is based on linked morpholino units (morpholino nucleic acid) having heterocyclic bases attached to the morpholino ring.
  • a number of linking groups have been reported that link the morpholino monomelic units in a morpholino nucleic acid.
  • Morpholino-based antisense compounds are disclosed in U.S. Patent 5,034,506, issued July 23, 1991.
  • the morpholino class of antisense compounds have been prepared having a variety of different linking groups joining the monomelic subunits.
  • CeNA cyclohexenyl nucleic acids
  • the furanose ring normally present in an DNA/RNA molecule is replaced with a cyclohenyl ring.
  • CeNA DMT protected phosphoramidite monomers have been prepared and used for antisense compound synthesis following classical phosphoramidite chemistry.
  • Fully modified CeNA antisense compounds and oligonucleotides having specific positions modified with CeNA have been prepared and studied (see Wang et al., J. Am. Chem. Soc, 2000, 122, 8595-8602). Modified Sugar Moieties
  • Antisense compounds of the invention may also contain one or more modified or substituted sugar moieties.
  • the base moieties (natural, modified or a combination thereof) are maintained for hybridization with an appropriate nucleic acid target.
  • Sugar modifications may impart nuclease stability, binding affinity or some other beneficial biological property to the antisense compounds.
  • Representative modified sugars include carbocyclic or acyclic sugars, sugars having substituent groups at one or more of their T, 3' or 4' positions, sugars having substituents in place of one or more hydrogen atoms of the sugar, and sugars having a linkage between any two other atoms in the sugar.
  • Antisense compounds of particular use in the instant invention may comprise a sugar substituent group selected from: OH; halo; O-, S-, or N- allcyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-0-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C 1 to C 10 alkyl or C 2 to Ci 0 alkenyl and alkynyl.
  • Some oligonucleotides comprise a sugar substituent group selected from: Ci to C ]0 lower alkyl, substituted lower allcyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH 3 , OCN, F, Cl, Br, CN, CF 3 , OCF 3 , SOCH 3 , SO 2 CH 3 , ONO 2 , NO 2 , N 3 , NH 2 , heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties.
  • a sugar substituent group selected from: Ci to C ]0
  • Oligonucleotides having the 2'-0-methoxyethyl substituent also have been shown to be antisense inhibitors of gene expression with promising features for in vivo use (Martin, P., HeIv. Chim. Acta, 1995, 78, 486-504; Altmann et al., Chimia, 1996, 50, 168-176; Altmann et al., Biochem. Soc. Trans., 1996, 24, 630-637; and Altmann et al., Nucleosides Nucleotides, 1997, 16, 917- 926). Relative to DNA, the oligonucleotides having the 2'-MOE modification displayed improved RNA affinity and higher nuclease resistance.
  • Antisense compounds having one or more 2'-MOE modifications are capable of inhibiting miRNA activity in vitro and in vivo (Esau et al., J. Biol. Chem., 2004, 279, 52361-52365; U.S. Application Publication No. 2005/0261218).
  • Additional modifications include 2'-dimethylaminooxyethoxy, i.e., a O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2'-DMAOE, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-O-dimethyl- amino-ethoxy-ethyl or 2'-DMAEOE), i.e., 2'-O-CH 2 -O-CH 2 -N(CH 3 ) 2 .
  • 2'-dimethylaminooxyethoxy i.e., a O(CH 2 ) 2 ON(CH 3 ) 2 group, also known as 2'-DMAOE
  • 2'-dimethylaminoethoxyethoxy also known in the art as 2'-O-dimethyl- amino-ethoxy-ethyl or 2'-DMAEOE
  • 2'-Sugar substituent groups may be in the arabino (up) position or ribo (down) position. Similar modifications may also be made at other positions on the antisense compound, particularly the 3' position of the sugar on the 3' terminal nucleoside or in T- 5' linked oligonucleotides and the 5' position of 5' terminal nucleoside.
  • Antisense compounds may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar.
  • Representative U.S. patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S.: 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785;
  • dimethylaminoethyloxyethyl substituent groups are disclosed in International Patent Application PCT/US99/17895, entitled ' ⁇ '-O-Dimethylaminoethyloxyethyl-Oligomeric compounds", filed August 6, 1999, hereby incorporated by reference in its entirety.
  • An additional sugar modification includes a bicyclic sugar moiety, which has a 2', 4' bridge that forces the sugar ring into a locked 3'-endo conformational geometry.
  • Bicyclic modifications imparts to an antisense compound greatly increased affinity for a nucleic acid target.
  • nucleosides having bicyclic sugar modifications can act cooperatively with DNA and RNA in chimeric antisense compounds to enhance the affinity of a chimeric antisense compound for a nucleic acid target.
  • Bicyclic sugar moieties can be represented by the formula 4'-(CH2)n-X-2', where X can be, for example, O or S.
  • LNATM is a bicyclic sugar moiety having a 4'-CH 2 -O-2' bridge (i.e.
  • X is O and n is 1).
  • the alpha-L nucleoside has also been reported wherein the linkage is above the ring and the heterocyclic base is in the alpha rather than the beta-conformation (see U.S. Patent Application Publication No.: Application 2003/0087230).
  • the xylo analog has also been prepared (see U.S. Patent Application Publication No.: 2003/0082807).
  • Another bicyclic sugar moiety is ENATM, where in the aforementioned formula X is O and n is 2.
  • the term 'locked nucleic acid' can also be used to describe any bicyclic sugar moiety that has a "locked” conformation.
  • LNATM LNATM monomers adenine, cytosine, guanine, 5-methyl- cytosine, thymine and uracil, along with their oligomerization, and nucleic acid recognition properties have been described (Koshkin et al., Tetrahedron, 1998, 54, 3607-3630). LNAs and preparation thereof are also described in WO 98/39352 and WO 99/14226. Analogs of LNATM, such as phosphorothioate-LNA and 2'-thio-LNAs (i.e. 2'-S-CH2-4'), have also been prepared (Kumar et al., Bioorg. Med. Chem.
  • oligonucleotide mimetics have been prepared to include bicyclic and tricyclic nucleoside analogs (see Steffens et al., HeIv. Chim. Acta, 1997, 80, 2426-2439; Steffens et al., J. Am. Chem. Soc, 1999, 121, 3249-3255; and Renneberg et al., J. Am. Chem. Soc, 2002, 124, 5993-6002).
  • These modified nucleoside analogs have been oligomerized using the phosphoramidite approach and the resulting antisense compounds containing tricyclic nucleoside analogs have shown increased thermal stabilities (Tm' s) when hybridized to DNA, RNA and itself.
  • Antisense compounds containing bicyclic nucleoside analogs have shown thermal stabilities approaching that of DNA duplexes.
  • oligonucleotide mimetic is referred to as phosphonomonoester nucleic acid and incorporates a phosphorus group in the backbone. This class of oligonucleotide mimetic is reported to have useful physical and biological and pharmacological properties in the areas of inhibiting gene expression. Nucleobase Modifications
  • Antisense compounds of the invention may also contain one or more nucleobase (often referred to in the art simply as "base”) modifications or substitutions which are structurally distinguishable from, yet functionally interchangeable with, naturally occurring or synthetic unmodified nucleobases. Such nucleobase modifications may impart nuclease stability, binding affinity or some other beneficial biological property to the antisense compounds.
  • nucleobase modifications include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U).
  • Modified nucleobases also referred to herein as heterocyclic base moieties include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2- propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl (-C ⁇ C-CH.
  • uracil and cytosine and other alkynyl derivatives of pyrimidine bases 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8- amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7- methyladenine, 2-F-adenine, 2-amino-adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7- deazaadenine and 3-deazaguanine and 3-deazaadenine.
  • Heterocyclic base moieties 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.
  • Some nucleobases include those disclosed in U.S. Patent No. 3,687,808, those disclosed in
  • nucleobases are particularly useful for increasing the binding affinity of the antisense compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including
  • antisense compounds are prepared having polycyclic heterocyclic compounds in place of one or more heterocyclic base moieties.
  • a number of tricyclic heterocyclic compounds have been previously reported. These compounds are routinely used in antisense applications to increase the binding properties of the modified strand to a target strand. The most studied modifications are targeted to guanosines, and have been termed G-clamps or cytidine analogs.
  • Tricyclic heterocyclic compounds and methods of using them that are amenable to the present invention are disclosed in U.S. Patent 6,028,183, and U.S. Patent 6,007,992, the contents of both are incorporated herein in their entirety.
  • Modified polycyclic heterocyclic compounds useful as heterocyclic bases are disclosed in but not limited to, the above noted U.S. 3,687,808, as well as U.S.: 4,845,205; 5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,434,257; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; 5,645,985; 5,646,269; 5,750,692; 5,830,653; 5,763,588; 6,005,096; and 5,681,941, and U.S.
  • nucleobase substitutions including 5-methylcytosinse substitutions, are particularly useful for increasing the binding affinity of the oligonucleotides of the invention.
  • 5- methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2 0 C (Sanghvi, Y.S., Crooke, S.T. and Lebleu, B., eds., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are presently preferred base substitutions, even more particularly when combined with 2'-O-methoxyethyl sugar modifications.
  • One substitution that can be appended to the antisense compounds of the invention involves the linkage of one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the resulting antisense compounds.
  • such modified antisense compounds are prepared by covalently attaching conjugate groups to functional groups such as hydroxyl or amino groups.
  • Conjugate groups of the invention include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of the antisense compounds.
  • Typical conjugate groups include cholesterol moieties and lipid moieties.
  • Additional conjugate groups include carbohydrates, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes.
  • Groups that enhance the pharmacodynamic properties include groups that impart to the antisense compound properties such as improved uptake, enhanced resistance to degradation, and/or enhance hybridization with RNA.
  • Groups that enhance the pharmacokinetic properties include groups impart to the antisense compound properties such as improved uptake, distribution, metabolism or excretion.
  • the antisense compounds of the invention can have one or more conjugated moieties to facilitate the active or passive transport, localization, or compartmentalization of the antisense compound.
  • Cellular localization includes, but is not limited to, localization to within the nucleus, the nucleolus, or the cytoplasm.
  • Compartmentalization includes, but is not limited to, any directed movement of the antisense of the invention to a cellular compartment including the nucleus, nucleolus, mitochondrion, or imbedding into a cellular membrane.
  • the antisense compounds of the invention comprise one or more conjugate moieties which facilitate posttranscriptional modification.
  • Antisense compounds used in the compositions of the present invention can also be modified to have one or more stabilizing groups that are generally attached to one or both termini of antisense compounds to enhance properties such as, for example, nuclease stability. Included in stabilizing groups are cap structures. By “cap structure” or “terminal cap moiety” is meant chemical modifications, which have been incorporated at either terminus of an antisense compound (see for example Wincott et al., WO 97/26270, incorporated by reference herein). These terminal modifications protect the antisense compounds having terminal nucleic acid molecules from exonuclease degradation, and can help in delivery and/or localization within a cell.
  • the cap can be present at the 5'-terminus (5'-cap), or at the 3'- terminus (3 '-cap), or can be present on both termini.
  • the cap may be present at either or both termini of either strand.
  • Cap structures are well known in the art and include, for example, inverted deoxy abasic caps.
  • 3' and 5 '-stabilizing groups that can be used to cap one or both ends of an antisense compound to impart nuclease stability include those disclosed in WO 03/004602 published on January 16, 2003.
  • Antisense compounds of this invention may have the chemically modified subunits arranged in patterns enhance the inhibitory activity of the antisense compounds. These patterns are described herein as "motifs", and include uniformly modified, positionally modified, gapmer or gapped, alternating, hemimer, and blockmer.
  • the term "uniform motif is meant to include antisense compounds wherein each nucleoside bears the same type of sugar moiety, which may be a naturally occuring sugar or a modified sugar.
  • "uniformly modified motif,” “uniform modifications,” and “uniformly modified” are meant to include antisense compounds wherein each nucleoside bears the same type of sugar modification.
  • Suitable sugar moieties include, but are not limited to 2'-O(CH 2 ) 2 ⁇ CH 3 [2'- MOE], 2'-OCH 3 [2'-O-methyl], LNA and ENATM.
  • an antisense compound may be uniformly modified such that each sugar modification is a 2'-MOE sugar modification.
  • an antisense compound may be uniformly modified such that each sugar modification is a 2'-O-methyl sugar modification.
  • the term "gapped motif or "gapmer” is meant to include an antisense compound having an internal region (also referred to as a “gap” or “gap segment”) positioned between two external regions (also referred to as "wing” or “wing segment”).
  • the regions are differentiated by the types of sugar moieties comprising each distinct region.
  • sugar moieties that are used to differentiate the regions of a gapmer include ⁇ -D-ribonucleosides, ⁇ -D- deoxyribonucleosides, 2'-modified nucleosides (such 2 '-modified nucleosides may include 2'-MOE, and 2'-O-CH 3 , among others), and bicyclic sugar modified nucleosides (such bicyclic sugar modified nucleosides may include LNATM or ENATM, among others).
  • each distinct region has a uniform motif sugar moieties within a region are uniform. Gapped motifs or gapmers are further defined as being either "symmetric" or "asymmetric".
  • a gapmer wherein the nucleosides of the first wing have the same sugar modifications as the nucleosides of the second wing is termed a symmetric gapped antisense compound.
  • Symmetric gapmers can have, for example, an internal region comprising a first sugar moiety, and external regions each comprising a second sugar moiety, wherein at least one sugar moiety is a modified sugar moiety.
  • Gapmers as used in the present invention include wings that independently have from 1 to 7 nucleosides. The present invention therefore includes gapmers wherein each wing independently comprises 1, 2, 3, 4, 5, 6 or 7 nucleosides. The number of nucleosides in each wing can be the same or different.
  • the internal or gap region comprises from 17 to 21 nucleosides, which is understood to include 17, 18, 19, 20, or 21 nucleosides.
  • positionally modified motif is meant to include a sequence of ⁇ -D-ribonucleosides, ⁇ -D-deoxyribonucleosides, or sugar-modified nucleosides wherein the sequence is interrupted by two or more regions comprising from 1 to about 8 sugar modified nucleosides wherein internal regions are generally from 1 to about 4 nucleosides. In other words, regions having a particular sugar moiety are separated by regions having different sugar moieties.
  • Regions comprised of sugar modified nucleosides have the same sugar modification or vary such that one region has a different sugar modification than another region.
  • Positionally modified motifs are not determined by the nucleobase sequence or the location or types of internucleoside linkages.
  • the term positionally modified motif includes many different specific substitution patterns. A number of these substitution patterns have been prepared and tested in compositions.
  • the present invention includes antisense compounds having a positionally modified motif characterized by regions of 2'-sugar modified nucleosides which are separated by regions of a second, distinct 2'-sugar modified nucleosides or bicyclic modified sugar nucleosides.
  • Preferred 2'-sugar moieties include 2'-O-methyl and 2'-MOE.
  • Preferred bicyclic sugar moieties include LNA and ENA.
  • the regions of 2 '-sugar modified moieties may be 2 to 8 nucleosides in length, and the regions of bicyclic modified sugar moieties may be 1 or 2 nucleosides in length.
  • antisense compounds of the invention are characterized by regions of two 2'-sugar modified nucleosides separated by regions of one bicyclic modified nucleoside, such that, beginning at the 5'-terminus, the antisense compounds have a 2'-sugar modified nucleoside at every first and second position, and a bicyclic modified nucleoside at every third position.
  • Such a motif is described by the formula 5'-(A-A-B)n(-A)nn ⁇ 3', wherein A is a first sugar moiety, B is a second sugar moiety, n is 6 to 7 and nn is 0 to 2.
  • A is 2'-MOE and B is LNA.
  • A is 2'-O-methyl and B is LNA.
  • a 2'-sugar modified nucleoside is incorporated in place of a bicyclic modified nucleoside. For example, if n is 7, and nn is zero, a 2'-sugar modified nucleoside would be utilized at the 3 '-terminal position in place of a bicyclic modified nucleoside.
  • Positionally modified motifs having less regular patterns are also included in the present invention.
  • the majority of the 2 '-sugar modified regions may be 2 nucleosides in length, and a minority of 2'-sugar modified regions may be 1 nucleoside in length.
  • the majority of the bicyclic modified regions may be 1 nucleoside in length, and a minority of the bicyclic modified regions may be 2 nucleosides in length.
  • One non-limiting example of such an antisense compound includes a positionally modified motif as described in the preceding paragraph, having one 2 '-sugar modified region one nucleoside in length, and one bicyclic modified region two nucleosides in length.
  • alternating motif is meant to include a contiguous sequence of two distinct alternating nucleosides for essentially the entire length of the compound.
  • the pattern of alternation can be described by the formula: 5'-A(B-A)n(-B)nn-3' where A and B are nucleosides differentiated by having at least different sugar moieties, nn is 0 or 1 and n is from about 7 to about 11.
  • a and B are nucleosides differentiated by having at least different sugar moieties
  • nn is 0 or 1
  • n is from about 7 to about 11.
  • This length range is not meant to be limiting as longer and shorter antisense compounds are also amenable to the present invention.
  • This formula also allows for even and odd lengths for alternating antisense compounds wherein the 5'- and 3 '-terminal nucleosides comprise the same (odd) or different (even) sugar moieties.
  • the "A" and “B” nucleosides comprising alternating antisense compounds of the present invention are differentiated from each other by having at least different sugar moieties.
  • Each of the A and B nucleosides has a modified sugar moiety selected from ⁇ -D-ribonucleosides, ⁇ -D- deoxyribonucleosides, 2'-modified nucleosides (such 2 '-modified nucleosides may include 2'-MOE, and 2'-O-CH3, among others), and bicyclic sugar modified nucleosides (such bicyclic sugar modified nucleosides may include LNA or ENA, among others).
  • the alternating motif is independent from the nucleobase sequence and the internucleoside linkages.
  • the internucleoside linkage can vary at each position or at particular selected positions or can be uniform or alternating throughout the antisense compound.
  • hemimer motif is meant to include a sequence of nucleosides that have uniform sugar moieties (identical sugars, modified or unmodified) and wherein one of the 5'-end or the 3'-end has a sequence of from 2 to 12 nucleosides that are sugar modified nucleosides that are different from the other nucleosides in the hemimer modified antisense compound.
  • An example of a typical hemimer is an antisense compound comprising a sequence of ⁇ -D-ribonucleosides or ⁇ -D- deoxyribonucleosides at one terminus and a sequence of sugar modified nucleosides at the other terminus.
  • One hemimer motif includes a sequence of ⁇ -D-ribonucleosides or ⁇ -D-deoxyribonucleosides at one terminus, followed or preceded by a sequence of 2 to 12 sugar modified nucleosides at the other terminus.
  • Another hemimer motif includes a sequence of ⁇ -D-ribonucleosides or ⁇ -D-deoxyribonucleosides at one terminus, followed or preceded by a sequence of 2 to 6 sugar modified nucleosides located at the other terminus, with from 2 to 4 sugar modified nucleosides being suitable as well.
  • the hemimer antisense compound comprises a region of 2'-MOE modified nuculeosides and a region of ⁇ -D-deoxyribonucleosides.
  • the ⁇ -D-deoxyribonucleosides comprise less than 13 contiguous nucleosides within the antisense compound.
  • blockmer motif is meant to include a sequence of nucleosides that have uniform sugars (identical sugars, modified or unmodified) that is internally interrupted by a block of sugar modified nucleosides that are uniformly modified and wherein the modification is different from the other nucleosides.
  • antisense compounds having a blockmer motif comprise a sequence of ⁇ -D-ribonucleosides or ⁇ -D-deoxyribonucleosides having one internal block of from 2 to 6, or from 2 to 4 sugar modified nucleosides.
  • the internal block region can be at any position within the antisense compound as long as it is not at one of the termini which would then make it a hemimer.
  • the base sequence and internucleoside linkages can vary at any position within a blockmer motif.
  • Antisense compounds having motifs selected from uniform, positionally modified, alternating, gapped, hemimer or blockmer may further comprise internucleoside linkage modifications or nucleobase modifications, such as those described herein.
  • "Chimeric antisense compounds” or “chimeras,” in the context of this invention, are antisense compounds that at least 2 chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide or nucleoside in the case of a nucleic acid based antisense compound. Accordingly, antisense compounds having a motif selected from positionally modified, gapmer, alternating, hemimer, or blockmer are considered chimeric antisense compounds.
  • Chimeric antisense compounds typically contain at least one region modified so as to confer increased resistance to nuclease degradation, increased cellular uptake, increased binding affinity for the target nucleic acid, and/or increased inhibitory activity.
  • an antisense compound may be designed to comprise a region that serves as a substrate for the cellular endonuclease KNase H, which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H by an antisense compound having a cleavage region, therefore, results in cleavage of the RNA target, thereby enhancing the efficiency of the antisense compound.
  • an antisense compound may be designed to comprise a region that imparts to the antisense compound increased affinity or activity such that antisense inhibition is achieved through a mechanism such as steric occlusion, rather than enzymatic cleavage.
  • Chemically modified nucleosides may also be employed to increase the binding affinity of a shortened or truncated antisense compound for its target nucleic acid. Consequently, comparable results can often be obtained with shorter antisense compounds that have such chemically modified nucleosides.
  • antisense compounds described herein having motifs selected from uniform, positionally modified, alternating, gapmer, hemimer, or blockmer, may further comprise intemucleoside linkage modifications and/or nucleobase modifications, such as those described herein.
  • Nucleosides both native and modified, have a certain conformational geometry which affects their hybridization and affinity properties.
  • the terms used to describe the conformational geometry of homoduplex nucleic acids are "A Form” for RNA and "B Form” for DNA.
  • RNA:RNA duplexes are more stable and have higher melting temperatures (Tm's) than DNA:DNA duplexes (Sanger et al., Principles of Nucleic Acid Structure, 1984, Springer- Verlag; New York, NY.; Lesnik et al., Biochemistry, 1995, 34, 10807-10815; Conte et al., Nucleic Acids Res., 1997, 25, 2627-2634).
  • RNA duplex The increased stability of RNA has been attributed to several structural features, most notably the improved base stacking interactions that result from an A-form geometry (Searle et al., Nucleic Acids Res., 1993, 21, 2051-2056).
  • the presence of the 2' hydroxyl in RNA biases the sugar toward a C3' endo pucker, i.e., also designated as Northern pucker, which causes the duplex to favor the A-form geometry.
  • the 2' hydroxyl groups of RNA can form a network of water mediated hydrogen bonds that help stabilize the RNA duplex (EgIi et al., Biochemistry, 1996, 35, 8489-8494).
  • deoxy nucleic acids prefer a C2 1 endo sugar pucker, i.e., also known as Southern pucker, which is thought to impart a less stable B-form geometry (Sanger, W. (1984) Principles of Nucleic Acid Structure, Springer- Verlag, New York, NY).
  • B-form geometry is inclusive of both C2'-endo pucker and O4'-endo pucker. This is consistent withBerger, et. al., Nucleic Acids Research, 1998, 26, 2473-2480, who pointed out that in considering the furanose conformations which give rise to B-form duplexes consideration should also be given to a O4'-endo pucker contribution.
  • DNA:RNA hybrid duplexes are usually less stable than pure RNA:RNA duplexes, and depending on their sequence may be either more or less stable than DNA:DNA duplexes (Searle et al., Nucleic Acids Res., 1993, 21, 2051-2056).
  • the structure of a hybrid duplex is intermediate between A- and B-form geometries, which may result in poor stacking interactions (Lane et al., Eur. J. Biochem., 1993, 215, 297-306; Fedoroff et al., J. MoI. Biol., 1993, 233, 509-523; Gonzalez et al., Biochemistry, 1995, 34, 4969-4982; Horton et al., J. MoI.
  • the stability of the duplex formed between a target RNA and a synthetic sequence is central to therapies such as, but not limited to, antisense mechanisms, including RNase H-mediated and RNA interference mechanisms, as these mechanisms involved the hybridization of a synthetic sequence strand to an RNA target strand or steric hindrance.
  • therapies such as, but not limited to, antisense mechanisms, including RNase H-mediated and RNA interference mechanisms, as these mechanisms involved the hybridization of a synthetic sequence strand to an RNA target strand or steric hindrance.
  • One routinely used method of modifying the sugar puckering is the substitution of the sugar at the 2'-position with a substituent group that influences the sugar geometry.
  • the influence on ring conformation is dependent on the nature of the substituent at the 2'-position.
  • a number of different substituents have been studied to determine their sugar puckering effect.
  • duplex stability can be enhanced by incorporating modified sugar moieties which exhibit A-form geometry.
  • modified sugar moieties which exhibit A-form geometry.
  • One of skill in the art can determine whether a given substituted sugar will have A-form geometry.
  • antisense compounds include nucleosides synthetically modified to induce a 3'-endo sugar conformation.
  • a nucleoside can incorporate synthetic modifications of the heterocyclic base, the sugar moiety or both to induce a desired 3'-endo sugar conformation.
  • These modified nucleosides are used to mimic RNA-like nucleosides so that particular properties of an antisense compound can be enhanced while maintaining the desirable 3'-endo conformational geometry.
  • RNA type duplex A form helix, predominantly 3'-endo
  • RNA interference e.g. trigger
  • Properties that are enhanced by using more stable 3'-endo nucleosides include, but are not limited to, modulation of one or more of the following: protein binding; protein off- rate; absorption and clearance; modulation of nuclease stability as well as chemical stability; modulation of the binding affinity and specificity of the antisense compound (affinity and specificity for enzymes as well as for complementary sequences); increased efficacy of RNA cleavage; and increase steric occulsion.
  • the present invention provides antisense compounds designed to act as triggers of RNAi having one or more nucleosides modified in such a way as to favor a C3'-endo type conformation.
  • modified nucleosides and the antisense compounds containing them can be estimated by various methods such as molecular dynamics calculations, nuclear magnetic resonance spectroscopy and CD measurements. Hence, modifications predicted to induce RNA-like conformations (A-form duplex geometry in an antisense compound context), are useful in the antisense compounds of the present invention.
  • the synthesis of modified nucleosides amenable to the present invention are known in the art (see for example, Chemistry of Nucleosides and Nucleotides VoI 1-3, ed. Leroy B. Townsend, 1988, Plenum Press.)
  • the present invention is directed to antisense compounds that are designed to have enhanced properties compared to native RNA.
  • One method to design optimized or enhanced antisense compounds involves each nucleoside of the selected sequence being scrutinized for possible enhancing modifications.
  • One modification would be the replacement of one or more RNA nucleosides with nucleosides that have the same 3'-endo conformational geometry.
  • Such modifications can enhance chemical and nuclease stability relative to native RNA while at the same time being much cheaper and easier to synthesize and/or incorporate into an oligonucleotide.
  • the sequence can be further divided into regions and the nucleosides of each region evaluated for enhancing modifications that can be the result of a chimeric configuration.
  • the antisense compounds of the present invention may include at least one 5 '-modified phosphate group on a single strand or on at least one 5'-position of a double-stranded sequence or sequences.
  • Other modifications considered are internucleoside linkages, conjugate groups, substitute sugars or bases, substitution of one or more nucleosides with nucleoside mimetics and any other modification that can enhance the desired property of the antisense compound.
  • alkyl means C 1 -C] 2 , C 1 -Cs, or Ci-C ⁇ , straight or (where possible) branched chain aliphatic hydrocarbyl.
  • heteroalkyl means C 1 -Cj 2 , C]-C 8 , or C]-C 6 , straight or (where possible) branched chain aliphatic hydrocarbyl containing at least one, or about 1 to about 3 hetero atoms in the chain, including the terminal portion of the chain.
  • Suitable heteroatoms include N, O and S.
  • cycloalkyl means C 3 -C] 2 , C 3 -C 8 , or C 3 -C 6 , aliphatic hydrocarbyl ring.
  • alkenyl means C 2 -C 12 , C 2 -C 8 , or C 2 -C 6 alkenyl, which may be straight or (where possible) branched hydrocarbyl moiety, which contains at least one carbon-carbon double bond.
  • alkynyl means C 2 -C] 2 , C 2 -C 8 , or C 2 -C 6 alkynyl, which may be straight or (where possible) branched hydrocarbyl moiety, which contains at least one carbon-carbon triple bond.
  • heterocycloalkyl means a ring moiety containing at least three ring members, at least one of which is carbon, and of which 1, 2 or three ring members are other than carbon.
  • the number of carbon atoms can vary from 1 to about 12, from 1 to about 6, and the total number of ring members varies from three to about 15, or from about 3 to about 8.
  • Suitable ring heteroatoms are N, O and S.
  • Suitable heterocycloalkyl groups include, but are not limited to, morpholino, thiomorpholino, piperidinyl, piperazinyl, homopiperidinyl, homopiperazinyl, homomorpholino, homothiomorpholino, pyrrolodinyl, tetrahydrooxazolyl, tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydroisoxazolyl, tetrahydropyrrazolyl, furanyl, pyranyl, and tetrahydroisothiazolyl.
  • aryl means any hydrocarbon ring structure containing at least one aryl ring. Suitable aryl rings have about 6 to about 20 ring carbons. Especially suitable aryl rings include phenyl, napthyl, anthracenyl, and phenanthrenyl.
  • hetaryl means a ring moiety containing at least one fully unsaturated ring, the ring consisting of carbon and non-carbon atoms.
  • the ring system can contain about 1 to about 4 rings.
  • the number of carbon atoms can vary from 1 to about 12, from 1 to about 6, and the total number of ring members varies from three to about 15, or from about 3 to about 8.
  • Suitable ring heteroatoms are N, O and S.
  • Suitable hetaryl moieties include, but are not limited to, pyrazolyl, thiophenyl, pyridyl, imidazolyl, tetrazolyl, pyridyl, pyrimidinyl, purinyl, quinazolinyl, quinoxalinyl, benzimidazolyl, benzothiophenyl, etc.
  • an electron withdrawing group is a group, such as the cyano or isocyanato group that draws electronic charge away from the carbon to which it is attached.
  • Other electron withdrawing groups of note include those whose electronegativities exceed that of carbon, for example halogen, nitro, or phenyl substituted in the ortho- or para-position with one or more cyano, isothiocyanato, nitro or halo groups.
  • halogen and halo have their ordinary meanings. Suitable halo (halogen) substituents are F, Cl, Br, and I.
  • halogens F, Cl, Br, I
  • alkyl alkenyl, and alkynyl moieties
  • NO 2 NH 3
  • acid moieties e.g. -CO 2 H, -OSO 3 H 2 , etc.
  • heterocycloalkyl moieties hetaryl moieties, aryl moieties, etc.
  • Phosphate protecting groups include those described in US Patents No. US 5,760,209, US
  • Screening Antisense Compounds Screening methods for the identification of effective modulators of miR-122a target nucleic acids are also comprehended by the instant invention and comprise the steps of contacting a miR-122a target nucleic acid, or portion thereof, with one or more candidate modulators, and selecting for one or more candidate modulators which inhibit the levels, expression or function of a miR-122a target nucleic acid.
  • the candidate modulator can be an antisense compound targeted to a pri- miRNA, or any portion thereof, including the mature miRNA, the Drosha recognition region, the Drosha cleavage region, the stem of the hairpin, or the loop of the hairpin.
  • Candidate modulators further include small molecule compounds that bind to structured regions of miR-122a target nucleic acids, such as structured regions within pri-miR-122a. Once it is shown that the candidate modulator or modulators are capable of modulating, preferably decreasing, the levels, expression or function of a miR-122a target nucleic acid, the modulator may then be employed in further investigative studies, or for use as a target validation, research, diagnostic, or therapeutic agent in accordance with the present invention.
  • compositions and formulations that include the antisense compounds and compositions of the invention.
  • Compositions and methods for the formulation of pharmaceutical compositions are dependent upon a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered. Such considerations are well understood by those skilled in the art.
  • the antisense compounds and compositions of the invention can be utilized in pharmaceutical compositions by adding an effective amount of the compound or composition to a suitable pharmaceutically acceptable diluent or carrier. Use of the antisense compounds and methods of the invention may also be useful prophylactically.
  • the antisense compounds and compositions of the invention encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal, including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to prodrugs and pharmaceutically acceptable salts of the antisense compounds of the invention, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
  • prodrug indicates a therapeutic agent that is prepared in an inactive form that is converted to an active form (i.e., drug) within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or conditions. This can include the incorporation of additional nucleosides at one or both ends of an antisense compound which are cleaved by endogenous nucleases within the body, to form the active antisense compound.
  • pharmaceutically acceptable salts refers to physiologically and pharmaceutically acceptable salts of the antisense compounds and compositions of the invention: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto. Suitable examples include, but are not limited to, sodium and postassium salts.
  • an antisense compound can be administered to a subject via an oral route of administration.
  • the subject may be a mammal, such as a mouse, a rat, a dog, a guinea pig, a non- human primate, or a human.
  • the subject may be in need of modulation of the level or expression a miR-122a nucleic acid, or in need of modulation of surrogate indicators as desribed here.
  • compositions for administration to a subject will comprise modified oligonucleotides having one or more modifications, as described herein.
  • Illustrative cell types include, but are not limited to: T-24 cells, A549 cells, normal human mammary epithelial cells (HMECs), MCF7 cells, T47D cells, BJ cells, B16-F10 cells, human vascular endothelial cells (HUVECs), human neonatal dermal fibroblast (NHDF) cells, human embryonic keratinocytes (HEK), 293T cells , HepG2 , human preadipocytes, human differentiated adipocytes (preapidocytes differentiated according to methods known in the art), NT2 cells (also known as NTERA-2 cl.Dl), and HeLa cells. Treatment of cells with antisense compounds
  • One reagent commonly used to introduce antisense compounds into cultured cells includes the cationic lipid transfection reagent LIPOFECTIN® (Invitrogen, Carlsbad, CA).
  • Antisense compounds are mixed with LIPOFECTIN® in OPTI-MEM® 1 (Invitrogen, Carlsbad, CA) to achieve the desired final concentration of antisense compound and a LIPOFECTIN® concentration that typically ranges 2 to 12 ⁇ g/mL per 100 nM antisense compound.
  • Another reagent used to introduce antisense compounds into cultured cells includes LIPOFECTAMINE® (Invitrogen, Carlsbad, CA). Antisense compound is mixed with
  • LIPOFECTAMINE® in OPTI-MEM® 1 reduced serum medium (Invitrogen, Carlsbad, CA) to achieve the desired concentration of antisense compound and a LIPOFECTAMINE® concentration that typically ranges 2 to 12 ⁇ g/mL per 100 nM antisense compound.
  • RNA or protein levels of target nucleic acids are measured by methods known in the art and described herein.
  • the target nucleic acid is a miRNA
  • the RNA or protein level of a protein-coding RNA regulated by a miRNA may be measured to evaluate the effects of antisense compounds targeted to a miRNA.
  • the data are presented as the average of the replicate treatments.
  • the concentration of antisense used varies from cell line to cell line. Methods to determine the optimal antisense concentration for a particular cell line are well known in the art. Antisense compounds are typically used at concentrations ranging from 1 nM to 300 nM.
  • RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA. Methods of RNA isolation are well known in the art. RNA is prepared using methods well known in the art, for example, using the TRIZOL® Reagent (Invitrogen, Carlsbad, CA) according to the manufacturer's recommended protocols.
  • Target nucleic acid levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or quantitaive real-time PCR.
  • RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA. Methods of RNA isolation are well known in the art. Northern blot analysis is also routine in the art. Quantitative real-time PCR can be conveniently accomplished using the commercially available ABI PRISM® 7600, 7700, or 7900 Sequence Detection System, available from PE-Applied Biosystems, Foster City, CA and used according to manufacturer's instructions.
  • Additional examples of methods of gene expression analysis known in the art include DNA arrays or microarrays (Brazma and ViIo, FEBS Lett., 2000, 480, 17-24; Celis, et al, FEBS Lett., 2000, 480, 2-16), SAGE (serial analysis of gene expression)(Madden, et al., Drug Discov. Today, 2000, 5, 415- 425), READS (restriction enzyme amplification of digested cDNAs) (Prashar and Weissman, Methods Enzymol., 1999, 303, 258-72), TOGA (total gene expression analysis) (Sutcliffe, et al., Proc. Natl. Acad. Sci. U. S.
  • Quantitative Real-Time PCR Analysis of Target RNA Levels Quantitation of target RNA levels is accomplished by quantitative real-time PCR using the ABI
  • PRISM® 7600, 7700, or 7900 Sequence Detection System PE-Applied Biosystems, Foster City, CA
  • Methods of quantitative real-time PCR are well known in the art.
  • RNA Prior to real-time PCR, the isolated RNA is subjected to a reverse transcriptase (RT) reaction, which produces complementary DNA (cDNA) that is then used as the substrate for the real-time PCR amplification.
  • RT reverse transcriptase
  • cDNA complementary DNA
  • RT and real-time PCR reagents are obtained from Invitrogen (Carlsbad, CA). RT, real-time-PCR reactions are carried out by methods well known to those skilled in the art.
  • Gene (or RNA) target quantities obtained by real time PCR are normalized using either the expression level of a gene whose expression is constant, such as GAPDH, or by quantifying total RNA using RIBOGREEN® (Molecular Probes, Inc. Eugene, OR). GAPDH expression is quantified by real time PCR, by being run simultaneously with the target, multiplexing, or separately. Total RNA is quantified using RIBOGREEN® RNA quantification reagent (Molecular Probes, Inc. Eugene, OR).
  • RNA quantification by RIBOGREEN® are taught in Jones, LJ., et al, (Analytical Biochemistry, 1998, 265, 368-374).
  • a CYTOFLUOR® 4000 instrument PE Applied Biosystems
  • Probes and primers are designed to hybridize to the target sequence, which includes a protein- coding RNA that is regulated by a miR-122a, such as ALDO A..
  • Methods for designing real-time PCR probes and primers are well known in the art, and may include the use of software such as PRIMER EXPRESS® Software (Applied Biosystems, Foster City, CA).
  • Northern blot analysis is performed according to routine procedures known in the art. Higher percentage acrylamide gels, for example, 10 to 15% acrylamide urea gels, are generally used to resolve miRNA. Fifteen to twenty micrograms of total RNA is fractionated by electrophoresis through 10% acrylamide urea gels using a TBE buffer system (Invitrogen). RNA is transferred from the gel to
  • HYB0NDTM-N+ nylon membranes (Amersham Pharmacia Biotech, Piscataway, NJ) by electroblotting in an Xcell SURELOCKTM Minicell (Invitrogen, Carlsbad, CA). Membranes are fixed by UV cross-linking using a STRATALINKER® UV Crosslinker 2400 (Stratagene, Inc, La Jolla, CA) and then probed using RAPID HYBTM buffer solution (Amersham) using manufacturer's recommendations for oligonucleotide probes.
  • a target specific DNA oligonucleotide probe with the sequence is used to detect the RNA of interest.
  • Probes used to detect miRNAs are synthesized by commercial vendors such as IDT (Coralville, IA). The probe is 5' end-labeled with T4 polynucleotide kinase with (gamma- 32 P) ATP (Promega, Madison, WI).
  • T4 polynucleotide kinase with (gamma- 32 P) ATP gamma- 32 P) ATP (Promega, Madison, WI).
  • T4 polynucleotide kinase with (gamma- 32 P) ATP gamma- 32 P) ATP (Promega, Madison, WI).
  • T4 polynucleotide kinase with (gamma- 32 P) ATP gamma- 32 P) ATP (Promega, Madison, WI).
  • U6 RNA is used to normalize for variations in loading and
  • Protein levels of a downstream target modulated or regulated by miR-122a can be evaluated or quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), enzyme-linked immunosorbent assay (ELISA), quantitative protein assays, protein activity assays (for example, caspase activity assays), immunohistochemistry, immunocytochemistry or fluorescence-activated cell sorting (FACS).
  • Antibodies directed to a target can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, MI), or can be prepared via conventional monoclonal or polyclonal antibody generation methods well known in the art.
  • antisense compounds of the invention are tested in various animal models of disease, including normal, lean mice, ob/ob mice, db/db mice, and diet-induced obese mice, and mouse and rat models of diet induced obesity.
  • Leptin is a hormone produced by fat cells that regulates appetite. Deficiencies in this hormone in both humans and non-human animals leads to obesity, ob/ob mice have a mutation in the leptin gene which results in obesity and hyperglycemia. As such, these mice are a useful model for the investigation of obesity and diabetes and treatments designed to treat these conditions, ob/ob mice have higher circulating levels of insulin and are less hyperglycemic than db/db mice, which harbor a mutation in the leptin receptor, db/db mice have a mutation in the leptin receptor gene which results in obesity and hyperglycemia.
  • mice are a useful model for the investigation of obesity and diabetes and treatments designed to treat these conditions
  • db/db mice which have lower circulating levels of insulin and are more hyperglycemic than ob/ob mice which harbor a mutation in the leptin gene, are often used as a rodent model of type 2 diabetes.
  • C57B1/6 mice are maintained on a standard rodent diet for use as control (lean) animals. Additionally, the C57BL/6 mouse strain is reported to be susceptible to hyperlipidemia-induced atherosclerotic plaque formation. Consequently, when these mice are fed a high-fat diet, they develop diet-induced obesity. Accordingly these mice are a useful model for the investigation of obesity and treatments designed to treat these conditions.
  • the Levin Model is a polygenic model of rats selectively bred to develop diet-induced obesity (DIO) associated with impaired glucose tolerance, dyslipidemra and insulin resistance when fed a high-fat diet (Levin, et al., Am. J. Physiol, 1997, 273, R725-30). This model is useful in investigating the antisense compounds of the present invention for their ability to affect obesity and related complications, such as impaired glucose tolerance, dyslipidemia and insulin resistance.
  • DIO diet-induced obesity
  • sequence of pri-miR-122a was used in certain embodiments of the invention, for example, in the design of antisense compounds targeting a miR-122a nucleic acid.
  • sequence of the human pri- miR-122a was extracted from the sequence having GENBANK® accession number NT_033907.3 is incorpated herein as SEQ ID NO: 5
  • Mature miRNAs found within the pri-miR-122a sequence were used in certain embodiments of the present invention, for example, in the design of antisense compounds targeting a miR-122a nucleic acid.
  • Antisense compounds can also be designed to mimic miR-122a while incorporating certain chemical modifications that alter one or more properties of the mimic, thereby creating a construct with superior properties over the endogenous miRNA.
  • Mature miR-122a sequences are shown in Table 1. Mature miR-122a sequences from pri-miR- 122a precursors have been proposed by several groups; consequently, two mature miR-122a sequences are disclosed in Table 1. The sequences are written in the 5' to 3' direction and are represented in the RNA form. It is understood that a person having ordinary skill in the art would be able to convert the sequence of the targets to a DNA form by simply replacing the uracil (U) with thymidine (T) in the sequence.
  • U uracil
  • T thymidine
  • antisense compounds were designed and synthesized to target various sites within miR-122a nucleic acids.
  • the antisense compounds are analyzed for their effect on miRNA, pre-miRNA or pri-miRNA levels by Northern blotting or quantitative realtime PCR as described, supra, or they can be used in other assays to investigate the role of miR-122a or miR-122a downstream targets.
  • the target sites to which the following antisense compounds are complementary include mature miR-122a, as well as target sites of pri-miR-122a.
  • an antisense compound having the sequence of SEQ ID NO: 12 was designed to target the stem loop of the pri-miR-122a structure.
  • nucleobase sequences and target sites of the antisense compounds of the invention are shown in Table 2.
  • the target site is the sequence of SEQ ID NO: 5 to which the antisense compound is complementary.
  • nucleobase sequences in Table 2 are shown having the DNA nucleobases "T”, one of skill in the art understand that in RNA sequences, "T” is replaced with "U”.
  • nucleobase sequence of the uniform RNA antisense compound ISIS 342970 "T” is replaced with "U”.
  • Table 4 lists antisense compounds having one of the sequences shown in Table 2 and one of the motifs desribed herein.
  • “Sugar” indicates the sugar type(s) found in the antisense compound;
  • “Backbone” indicates the type of internucleoside linkages;
  • “Motif indicates the particular motif of the antisense compound. Motifs are described according to the number of nucleosides in each distinct region. For example, the positionally modified motif "5-1-5-1-4-1-6", having 2'-MOE and LNA sugars has a region of 5 2'-MOE modified nucleosides, followed by a region of one LNA modified nucleoside, and so forth. The type of sugar, internucleoside linkage or base modification is noted using the tRNA modification-like abbreviation system. Sequence, internucleoside linkage, nucleobase, and sugar types are all shown and indicated as follows:
  • ISIS 343160 is an oligomeric compound representing a chemically modified miR-122a mimic. It is also understood that the human miR-122a mimic compound may also mimic other rniRNAs from mammals, such as those from rodent species, for example. It is also understood that these mimics can serve as the basis for several variations of nucleic acid oligomeric compounds, including compounds with chemical modifications such as uniform or chimeric 2'-MOE oligomeric compounds, as well as LNAs and PNAs; such oligomeric compounds are also within the scope of the invention.
  • Example 4 Effects of antisense compounds targeting miR-122a in phenotypic assays
  • antisense oligonucleotides ISIS 328372, ISIS 328373, and ISIS 327895 on miR- 122a were tested in several phenotypic assays, including an apoptosis assay, an adipocyte differentiation assay, and an insulin signaling assay.
  • the experimental details of these assays, and the results obtained, are described in detail in U.S. Application Publication No. 2005/0261218, which is hereby incorporated by reference in its entirety.
  • ISIS 328372 and ISIS 328373 did not induce apoptosis in the apoptosis assay.
  • ISIS 327895 reduced levels of markers of adipocyte differentiation, and decreased mRNA expression of follistatin and PEPCKc in the insulin signaling assay.
  • Mouse primary hepatocytes were isolated from Balb/c mice using methods described in the art (Neufeld, 1997, Methods MoI. Biol., 75:145-151.). Cells were seeded into 96-well plates in culture medium (Williams' Medium E supplemented with 10% fetal bovine serum, 10 nM HEPES, and penicillin/streptomycin) and cultured overnight before transfection. Transfection of oligonucleotide in LIPOFECTIN® (Invitrogen, Carlsbad, CA) was performed in triplicate, according to the manufacturer's instructions.
  • RNA samples were treated with either ISIS 327895 or the control oligonucleotide ISIS 342683 at concentrations of 15 nM, 44 nM, or 133 nM. Cells were lysed 24 hours after transfection and total RNA was harvested using using QIAGEN® RNEASY® 96 columns on a BIOROBOT® 3000 (Qiagen, Valencia, CA). The mRNA levels of miR-122a target genes predicted by the TargetScan algorithm (Lewis et al,
  • the miR-122a duplex RNA that was used as a miR-122a mimetic was purchased from Dharmacon (Lafayette, CO).
  • the miR-122a duplex contained: UGGAGUGUGACAAUGGUGUUUGU (SEQ ID NO: 3) and its complement AAACACCAUUGUCACACUCCAUA (SEQ ID NO: 25).
  • the 5' mismatch miR-122a duplex comprised: UGCACAGAGACAAUGGUGUUUGU (SEQ ID NO: 26) and its complement AAACACCAUUGUCUCUGUGCAUA (SEQ ID NO: 27).
  • the 3' mismatch miR- 122a duplex comprise UGGAGUGUGACAUUGCAGUAGUG (SEQ ID NO: 28) and its complement AUACUGCAAUGUCACACUCCAUA (SEQ ID NO: 29).
  • AML12 cells do not have high expression levels of miR-122a as are observed in primary hepatocytes (see Figure 6).
  • ISIS 342683 is CCTTCCCTGAAGGTTCCTCCTT (SEQ ID NO: 30); this oligomeric compound is uniformly composed of 2'-MOE nucleosides, has a phosphorothioate backbone, and all cytosines are 5-methylcytosines.
  • ISIS 327895 and ISIS 365219 were effective in inhibiting miR-122a, as evidenced by the increased expression of GYSl.
  • ISIS 386653 and ISIS 386654 increased GYSl mRNA levels to a lesser extent.
  • the gapmer ISIS 345363 did not markedly increase GYSl mRNA levels.
  • ISIS 386655, ISIS 386656, ISIS 386657, ISIS 386658, ISIS 386659, or ISIS 386660 at doses of 15 nM, 44 nM, 133 nM, or 400 nM. While truncations resulted in slightly less stimulation of GYSl mRNA levels, some truncated antisense compound were able to stimulate GYSl mRNA expression. Antisense compounds in which 2, 4, or 6 nucleosides were truncated from the 5' end, in this assay, were able to stimulate GYSl mRNA expression, relative to control samples.
  • An antisense compound in which 2 nucleosides were truncated from the 3' end was able to stimulate GYSl mRNA expression, relative to control samples.
  • truncations of 4 or 6 nucleosides from the 3' end significantly reduced the ability of the antisense compounds to stimulate GYSl mRNA expression.
  • mouse primary hepatocytes were treated with oligonucleotides bearing mismatches to miR-122a (ISIS 386661, ISIS 386662, ISIS 386663, ISIS 386664, ISIS 386665, and ISIS 386666). Increasing the number of mismatches appears to decrease the ability of the antisense oligonucleotides to modulate GYSl mRNA levels.
  • Embodiments of the present invention include methods of validating predicted miR-122a targets. Also provided are methods of modulating miR-122a targets with an antisense compound which targets miR-122a.
  • the antisense compound has the nucleobase sequence of SEQ ID NO: 2.
  • the antisense compound is uniformly comprised of 2'-MOE sugar modifications.
  • the antisense compound is ISIS 327895. Further provided are methods of assaying for inhibition of miR-122a comprising monitoring expression of a miR-122a target gene.
  • Example 7 Modulation of miR-122a activity in vivo with miRNA antisense—Normal mice W 2
  • mice Male C57BL/6 mice were obtained from The Jackson Laboratory. The mice were separated into treatment groups and were treated intraperitoneally with doses of ISIS 327895 ranging from 12.5 to 75 mg/kg or with 75 mg/kg of the control oligonucleotide ISIS 342683 twice weekly for four weeks. Mice treated with saline alone served as further controls. The mice appeared healthy and normal at the end of treatment with plasma AST and ALT levels in the normal range and no loss of body weight or reduced food intake. Histological analysis of liver sections revealed no apparent change in morphology. (Histological analysis of liver was carried out via routine procedures known in the art. Briefly, liver was fixed in 10% buffered formalin and embedded in paraffin wax. 4-mm sections were cut and mounted on glass slides.
  • the levels of the five miR-122a target mRNAs identified in vitro were evaluated in liver tissue using Taqman real-time PCR.
  • Four of the five mRNAs (GYSl, ALDOA, P4HA1, and CCNGl) were increased in the. mice treated with ISIS 327895, while no target mRNA changes were observed in mice treated with the control oligonucleotide, demonstrating specific inhibition of miR-122a in the liver.
  • P4HA1 displayed maximal upregulation at the lowest dose of ISIS 327895 tested.
  • An embodiment of the present invention is a method of modulating miR-122a targets in an animal comprising administering an antisense oligonucleotide targeting miR-122a.
  • the targets are modulated in the liver.
  • said antisense oligonucleotide is ISIS 327895.
  • An embodiment of the present invention is a method of reducing miR-122a levels in the liver of an animal comprising administering an antisense oligonucleotide targeting miR-122a.
  • said antisense oligonucleotide is ISIS 327895.
  • Plasma levels of total cholesterol, triglycerides and glucose were also monitored using methods known in the art (for example, via Olympus AU400e automated clinical chemistry analyzer, Melville,
  • Embodiments of the present invention include methods of reducing serum cholesterol, glucose, or triglycerides in an animal comprising administering an antisense compound targete to miR-122a.
  • the antisense compound has the nucleobase sequence of SEQ ID NO: 2.
  • the antisense compound is uniformly comprised of 2'-MOE sugar modifications.
  • the antisense compound is ISIS 327895.
  • Example 8 Effects of miR-122a antisense oligonucleotide treatment in a diet-induced model of obesity: 25 mg/week study
  • Mice were fed ad libitum. Starting insulin levels were approximately 15ng/dL and body composition was approximately 20% fat.
  • Mice were dosed subcutaneously at 12.5 mg/kg twice weekly (i.e. 25 mg/wk), for a total of 11 total doses. Each treatment group consisted of 5 mice.
  • Treatment groups were as follows: saline; ISIS 327895, antisense to miR-122a; ISIS 342683, a scrambled control oligonucleotide.
  • the sequence of ISIS 342683 is CCTTCCCTGAAGGTTCCTCCTT (SEQ ID NO: 30); this oligomeric compound is uniformly composed of 2'-MOE nucleosides, has a phosphorotliioate backbone, and all cytosines are 5-methylcytosines.
  • Body composition was determined by MRI at the start of the study and following 3.5 and 5 weeks of treatment. Serum chemistries were analyzed following 2, 3, 4, 5, and 6 weeks of treatment.
  • Insulin was measured via routine ELISA in the fed state following 3 weeks of treatment, and in the fasted state following 5 weeks of treatment.
  • An oral glucose tolerance test was performed following 5 weeks of treatment (via routine procedures: glucose was administered, blood samples were collected and analyzed on a clinical analyzer).
  • Serum lipoprotein and cholesterol profiling was performed as described (Kieft et al., 1991) using a Beckman System Gold 126 HPLC system, a 507e refi ⁇ dgerated autosampler, a 126 photodiode array detector (Beckman Instruments) and a Superose 6 HR 10/30 column (Pfizer). HDL, LDL, and VLDL fractions were measured at a wavelength of 505 nm and valiated with a cholesterol calibration kit (Sigma, St. Louis, MO). For each experiment, a three-point standard curve was performed in triplicate to determine the absolute concentration of each lipoprotein fraction. Lipoprotein analysis performed in this manner showed that the decrease in total cholesterol reflected a reduction in both low-density lipoprotein and high-density lipoprotein fractions.
  • Glycogen synthase 1 is a miR-122a target and its expression is modulated by antisense inhibition of miR-122a. As glycogen synthase 1 expression was elevated in livers of mice treated with ISIS 327895, glycogen content in livers was evaluated. A significant increase in glycogen levels in the livers of the ISIS 327895-treated mice was observed, suggesting that miR-122a may also regulate glycogen storage through its repression of the muscle glycogen synthase (GYSl) mRNA.
  • GYSl muscle glycogen synthase
  • Table 9 Mean serum chemistry levels in 25 mg/wk DIO Study.
  • GLUC glucose
  • ALT alanine aminotransferase
  • AST aspartate aminotransferase
  • chol total cholesterol
  • trigs triglycerides
  • HDL HDL-cholesterol
  • LDL LDL-cholesterol
  • NEFA non-esterif ⁇ ed fatty acids
  • miR-122a target genes whose expression is upregulated following antisense inhibition of miR- 122a in vivo included GYS 1 , ALDO A, CCNGl , and P4HA1. mRNA levels were measured by real-time PCR using procedures described herein and commonly known in the art. Upregulation of these genes in mice treated with ISIS 327895 confirms inhibition of miR-122a activity.
  • liver steatosis showed substantial reduction of liver steatosis with ISIS 327895 treatment. Liver triglyceride levels were also reduced with antisense inhibition of miR-122a. Liver weights were also decreased. A trend toward reduction in plasma transaminase levels in ISIS
  • Embodiments of the present invention include methods of improving insulin sensitivity, methods of reducing cholesterol, methods of reducing LDL cholesterol, methods of reducing FFA, methods of reducing ApoB-100 levels, and methods of reducing triglycerides in the serum of an animal by administering an antisense compound targeted to miR-122a.
  • improvement in insulin sensitivity is measured as a reduction in circulating insulin levels.
  • the antisense compound has the nucleobase sequence of SEQ ID NO: 2.
  • the antisense compound is uniformly comprised of 2'-MOE sugar modifications.
  • the antiense compound is ISIS 327895.
  • liver gene expression following antisense inhibition of miR- 122a were evaluated by microarray analysis.
  • Liver KNA was isolated from normal mice treated with 50 mg/kg of ISIS 327895 twice weekly and from animals treated with saline alone, and microarray analysis was performed.
  • GE CODELINKTM Mouse Whole Genome Bioarrays were used according to the manufacturer's protocols. Analysis of modulated genes by Gene Ontology category revealed that many genes involved in regulation of lipid and carbohydrate metabolism were affected in the ISIS 327895 treated mice. Key eixzymes in the fatty acid synthesis pathway, cholesterol biosynthesis pathway, acetyl CoA transport pathway, and glycolysis pathway were inhibited.
  • Phosphomevalonate kinase (PMVK) mRNA downregulation was statistically significant.
  • Several other genes related to cholesterol metabolism were also reduced, including HMGCR and PMVK.
  • genes indicative of hepatocyte de-differentiation for example, HNF-lalpha, HNF-I beta, HNF-4, albumin, cytokeratin 19, and alpha-fetoprotein, did not exhibit altered expression following antisense inhibition of miR-122a.
  • the microarray data is deposited in a publicly accessible database Gene
  • another embodiment of the present invention is a method of identifying miR-122a targets.
  • Another embodiment of the present invention is a method of modulating the expression of any of the 108 targets identified in the preceding table comprising modulating the miR-122a.
  • miR-122a is modulated by an antisense compound.
  • the antisense compound has the nucleobase sequence of SEQ ID NO: 2.
  • the antisense compound is uniformly comprised of 2'-MOE sugar modifications.
  • the antiense compound is ISIS 327895.
  • Example 10 Modulation of lipid metabolism following antisense inhibition of miR-122a
  • Fatty acid oxidation is an ATP-producing catabolic pathway.
  • the central metabolic sensor AMPK is a key regulatory enzyme that acts to promote ATP-generating pathways such as fatty acid oxidation and inhibitis energy storage pathways such as fatty acid synthesis.
  • hepatocytes Fatty acid oxidation was measured in hepatocytes isolated from livers of ISIS 327895-treated mice.
  • Primary hepatocytes were isolated from lean C57B1/6 mice treated with saline, ISIS 327895, or ISIS 342683, at 25 mg/kg, twice per week, for a total of 5 doses.
  • Hepatocytes were plated in 25 cm 2 culture flasks, at a density of 1 X 10 6 cells per flask. Prior to plating of the cells, the flasks were treated with 2 ml of 0.1 mg/ml collagen in phosphate-buffered saline for 10 minutes at room temperature.
  • Each liver yielded 4 flasks, 3 of which were used for fatty acid oxidation experiments and one of which was reserved for protein measurements.
  • To each flask of hepatocytes was added 3 ml of William E culture medium, supplemented with 1.0 nM insulin and 10% fetal bovine serum (medium and supplements available from, for example, Invitrogen Life Technologies, Carlsbad, CA). Following overnight culture, the cells were washed twice with phosphate-buffered saline and were then cultured in 2.0 ml of low- glucose DMEM containing 0.25% bovine serum albumin and 0.25 uCi 14 C-oleaic acid.
  • the flask was filled with 5% CO 2 /95%0 2 and capped with a septa rubber stop, to which was attached a 0.5 ml tube with a filter paper.
  • a 0.5 ml tube with a filter paper Three hours lated, 0.4 ml of 70% HCIO 4 was injected into the culture medium and 0.05 ml of 25% NaOH was immediated injected onto the filter paper to collect the CO 2 .
  • the filter paper was removed and placed in a scintillation vial. The 0.5 ml tube that contained the filter paper was washed twice with water and the water was transferred into the scintillation vial.
  • Embodiments of the present invention include methods of lowering hepatic fatty acid synthesis and sterol synthesis and methods of increasing hepatic fatty acid oxidation in an animal comprising administering to said animal an antisense compound targeted to miR-122a.
  • the antisense compound has the nucleobase sequence of SEQ ID NO: 2.
  • the antisense compound is uniformly comprised of 2'-MOE sugar modifications.
  • the antiense compound is ISIS 327895.
  • Example 11 Effects of miR-122a antisense oligonucleotide treatment in ob/ob mice
  • mice Eight week old ob/ob mice were given an intraperitoneal injection of ISIS 327895 or a control oligonucleotide dissolved in saline at a dose of 25 mg/kg twice a week for 4.5 weeks. Blood was collected and analyzed using routine clinical analyzer instruments (for example, an Olympus AU400e automated clinical chemistry analyzer, Melville, NY). Treatment with ISIS 327895 caused significant decrease in plasma cholesterol levels (about 168 mg/dL) as compared to treatment with control oligonucleotide (about 237 mg/dL) or saline-treatment alone (about 241 mg/dL).
  • Antisense inhibition of miR-122a also reduced hepatic triglyceride levels (about 89 mg/g) as compared to treatment with control oligonucleotide (about 161 mg/g) or saline-treatment alone (about 151 mg/g). Liver triglyceride levels are measured using routine methods described in the art (Desai, et al. 2001, Diabetes, 50: 2287-2295. The reduction in steatosis was reflected as a decrease in plasma ALT and AST levels in the group treated with ISIS 327895.
  • embodiments of the present invention include methods of decreasing hepatic triglycerides in an animal, methods of decreasing serum cholesterol in an animal, and methods of improving hepatic function in an animal comprising administering an antisense oligonucleotide which inhibits miR-122a function.
  • the present invention also contemplates methods of ameliorating or preventing hyperlipidemia or hepatic steatosis in an animal comprising administering an antisense compound targeted to miR-122a.
  • the antisense compound has the nucleobase sequence of SEQ ID NO: 2.
  • the antisense compound is uniformly comprised of 2'-MOE sugar modifications.
  • the antiense compound is ISIS 327895.
  • miR-122a plays a role in regulating cholesterol and lipid metabolism. The effects are independent of leptin action, and there are no overt side effects caused by reduction of miR-122a.
  • Another embodiment of the present invention is a method of regulating cholesterol and lipid metabolism by inhibiting miR-122a.
  • Example 12 Antisense inhibition of miR-122a activity in vivo using enhanced antisense compounds
  • the antisense compounds of the invention modulate the activity or function of the small non- coding RNAs to which they are targeted.
  • antisense compounds targeted to miR-122a are illustrated; however, the modifications in the antisense compounds of the invention are not limited to those antisense compounds that modulate miR-122a.
  • mice Male C57BL/6 mice were obtained from The Jackson Laboratory. Mice were treated with antisense compounds targeting miR-122a, or received saline as a control treatment.
  • One of the antisense compounds tested included ISIS 327895, a 2'-MOE uniformly modified compound fully complementary to miR-122a. Also tested in vivo was ISIS 387574, a positionally modified antisense compound having the motif (A-A-B)7(-A-A)l where A is 2'-MOE and B is LNA. ISIS 387574 is fully complementary to miR-122a. Mice were administered 25 mg/kg doses of antisense compound intraperitoneally, for a total of 6 doses. Following the end of the treatment period, RNA was isolated from liver and the levels of a miR- 122a target mRNA, ALDOA, were measured using Taqman real-time PCR.
  • Serum total cholesterol levels were also monitored using methods known in the art (for example, via Olympus AU400e automated clinical chemistry analyzer, Melville, NY). Reductions in total cholesterol were observed in mice treated with either the 2'-MOE uniformly modified compound or the positionally modified 2 ' -MOE/LNA compound.
  • Additional measurements performed on serum samples include measurements of LDL- cholesterol, triglycerides, and serum tranaminases. Additional analyses that are performed in such in vivo studies included histological analysis of liver sections, to evaluate changes in morphology. Histological analysis of liver is carried out via routine procedures known in the art. Briefly, liver is fixed in 10% buffered formalin and embedded in paraffin wax. 4-mm sections are cut and mounted on glass slides. After dehydration, the sections are stained with hematoxylin and eosin. Morphological analysis may also include evaluation of hepatic steatosis, using oil Red O staining procedures known in the art.
  • Embodiments of the present invention include methods of reducing serum total cholesterol in an animal comprising administering an antisense compound targeted to a miR-122a nucleic acid.
  • the antisense compound has the nucleobase sequence of SEQ ID NO: 2.
  • the antisense compound is uniformly comprised of 2'-MOE sugar modifications.
  • the antisense compound has a positionally modified motif, 5'-(A-A-B)n(-A)nn- 3', where A is 2'-MOE, B is LNA, n is 7, and nn is 2.
  • the antiense compound is ISIS 327895.
  • the antisense compound is ISIS 387574. Additionally, either of these antisense compounds is used to achieve one or more additional phenotypic changes, such as lowered serum LDL-cholesterol, lowered serum triglycerides, or reduced hepatic steatosis.
  • antisense compounds targeted to miR-122a and having truncations were tested in vivo. Truncation of a single nucleoside from the 5' end of the antisense compound yielded increases in ALDO A mRNA levels comparable to those of the full-length antisense compound, ISIS 327895. Truncations of two nucleosides from the 5' end, 5 nucleosides from the 3' end, or 1 nucleoside from the 3' end resulted in slightly elevated ALDO A mRNA levels. Truncation of 3 nucleosides from the 3' end resulted in no increase in ALDO A mRNA levels.
  • a further embodiment of the invention includes practicing any of the methods described herein using an antisense compound targeted to miR-122a, wherein the antisense compound has no more than 2 truncations from the 5' end or no more than 1 truncation from the 3' end, relative to the nucleobase sequence of SEQ ID NO: 2.

Abstract

Methods are provided for the treatment of cardiovascular or metabolic diseases characterized by elevated serum total cholesterol, elevated serum LDL-cholesterol, or elevated serum triglycerides, through the administration of an oligomeric compound which modulates the levels or activity of miR-122a. Further provided are methods for reducing hepatic steatosis or liver tissue triglyceride accumulation through the administration of an oligomeric compound which modulates the levels or activity of miR-122a. Such methods employ oligomeric compounds which hybridize with or sterically interfere with nucleic acid molecules comprising or encoding miR-122a. Such oligomeric compounds may include one or more modifications thereon, which may improve the activity, stability, or nuclease resistance of the oligomeric compound. These modified oligomeric compounds are used as single compounds or in compositions, including pharmaceutical compositions, to modulate or mimic the targeted nucleic acid comprising or encoding miR-122a.

Description

METHODS FOR USE IN MODULATING MIR-122a
Cross-Reference To Related Applications
This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Application Serial No. 60/712,211 filed August 29, 2005, U.S. Provisional Application Serial No. 60/731,377 filed October 28, 2005, and U.S. Provisional Application Serial No. 60/771,592, filed February 7, 2006, each of which is incorporated herein by reference in its entirety.
This application is related to US Application Serial No. 10/909,125 filed July 30, 2004 which claims priority to U.S. provisional applications Serial No. 60/492,056 filed July 31, 2003, Serial No. 60/516,303 filed October 31, 2003, Serial No. 60/531,596 filed December 19, 2003, and Serial No. 60/562,417 filed April 14, 2004, each of which is incorporated herein by reference in its entirety.
Field of the Invention
The present invention provides methods for the modulation of small non-coding RNAs, in particular miR- 122a. Methods are provided for the treatment of cardiovascular or metabolic diseases characterized by elevated serum total cholesterol, elevated serum LDL-cholesterol, or elevated serum triglycerides, through the administration of an antisense compound which inhibits the levels or activity of miR- 122a. Further provided are methods for reducing hepatic steatosis or liver tissue triglyceride accumulation through the administration of an antisense compound which inhibits the levels or activity of miR-122a. Such methods employ antisense compounds which hybridize with or sterically interfere with nucleic acid molecules comprising or encoding miR- 122a. Such antisense compounds may include one or more modifications thereon, which may improve the activity, stability, or nuclease resistance of the antisense compound. These modified antisense compounds are used as single compounds or in compositions, including pharmaceutical compositions, to inhibit miR- 122a.
Background of the Invention
MicroRNAs (miRNAs) are small (approximately 21-24 nucleotides in length, these are also known as "mature" miRNA), non-coding RNA molecules encoded in the genomes of plants and animals. These highly conserved, endogenously expressed RNAs are believed to regulate the expression of genes by binding to the 3'-untranslated regions (3'-UTR) of specific mRNAs. MiRNAs may act as key regulators of cellular processes such as cell proliferation, cell death (apoptosis), metabolism, and cell differentiation. On a larger scale, miRNA expression has been implicated in early development, brain development, disease progression (such as cancers and viral infections). There is speculation that in higher eukaryotes, the role of miRNAs in regulating gene expression could be as important as that of transcription factors. More than 200 different miRNAs have been identified in plants and animals (Ambros et al., Curr. Biol., 2003, 13, 807-818). Mature miRNAs appear to originate from long endogenous primary miRNA transcripts (also known as pri-miRNAs, pri-mirs, pri-miRs or pri-pre- miRNAs) that are often hundreds of nucleotides in length (Lee, et al., EMBO J., 2002, 21(17), 4663- 4670).
In mammals, only a few miRNAs have been assigned any function, although they are predicted to regulate a large percentage of genes, with estimates based on bioinformatic target prediction ranging as high as 30% (Lewis et al., 2005). Based on the early studies in invertebrates, miRNAs are expected to have similar roles in developmental regulation and cell differentiation in mammals, and roles for miRNAs in cardiogenesis (Zhao et al., 2005) and lymphocyte development (Chen et al., 2004) have been demonstrated. Several studies have also found a strong connection between miRNA and human cancer, including a report that miRNA genes are often found in genomic regions linked to cancer (Calin et al., 2004; McManus, 2003) and a study correlating miRNA expression profiles with developmental lineage and differentiation state of tumors (Lu et al., 2005). A potential role for miRNAs in metabolic pathways has been suggested by studies implicating miRNAs in regulation of adipocyte differentiation (Esau et al., 2004) and glucose-stimulated insulin secretion from pancreatic islet cells (Poy et al., 2004). miR-122a is expressed in the developing liver (Chang et al., 2004) and at high levels in the adult liver, where it makes up 70% of all miRNA (Chang et al., 2004; LagosQuintana et al., 2002). It is one of many tissue-specific microRNAs thought to be important for establishing patterns of gene expression which may be responsible for maintaining the differentiated state of a tissue (Lagos-Quintana et al., 2002; Lim et al., 2005). miR-122a was also reported to enhance replication of HCV through a novel mechanism which is not yet understood, making it also a potential therapeutic target for HCV infection (Jopling et al., 2005).
It is described herein that the modulation of miR-122a is an attractive approach for diseases and conditions characterized by elevated serum cholesterol, elevated serum triglycerides, or hepatic steatosis. The present invention therefore provides antisense compounds and methods useful for modulating miR- 122a, to achieve clinically desirable changes in cholesterol and lipid profiles in animals.
Summary of the Invention
The present invention provides methods of lowering a serum indicator of cardiovascular disease risk, which include elevated serum total cholesterol elevated serum LDL-cholesterol, and elevated serum triglyceride levels, comprising selecting an animal having an elevate serum indicator of cardiovascular disease, and administering to the animal an antisense compound targeted to miR-122a. The present invention provides methods of lowering serum cholesterol in an animal comprising selecting an animal having elevated serum cholesterol levels, and then administering to the animal a therapeutically effective amount of an antisense compound essentially complementary to a miR-122a nucleic acid. The serum cholesterol levels may be total serum cholesterol levels or serum LDL- cholesterol levels.
The present invention further provides a method of lowering triglyceride levels in an animal comprising selecting an animal having elevated triglyceride levels, and then administering to the animal a therapeutically effective amount of an antisense compound essentially complementary to a miR- 122a nucleic acid. The triglyceride levels may be serum triglyceride levels or liver tissue triglyceride levels. Further provided is a method of lowering serum lipoproteins in an animal comprising selecting an animal having elevated serum lipoproteins and administering to the animal a therapeutically effective amount of an antisense compound essentially complementary to a miR-122a nucleic acid. The serum lipoprotein may be apolipoprotein B-IOO.
The present invention provides methods of reducing hepatic steatosis in an animal comprising selecting an animal having hepatic steatosis and administering to the animal a therapeutically effective amount of an antisense compound essentially complementary to a miR-122a nucleic acid. The hepatic steatosis may be steatohepatitis or non-alcoholic steatohepatitis.
Additionally provides are methods of reducing triglyceride accumulation in the liver of an animal comprising administering to the animal a therapeutically effective amount of an antisense compound essentially complementary to a miR- 122a nucleic acid.
The present invention provides methods of modulating a metabolic pathway in an animal, comprising administering to an animal a therapeutically effective amount of an antisense compound essentially complementary to a miR- 122a nucleic acid. The metabolic pathway is selected from lipogenesis, fatty acid oxidation, fatty acid synthesis rate, or sterol synthesis. The methods provided reduce lipogenesis, reduce fatty acid synthesis rate, reduce sterol synthesis, or increase fatty acid oxidation.
Further provided is a method of improving hepatic function in an animal comprising contacting an animal with a therapeutically effective amount of an antisense compound essentially complementary to a miR-122a nucleic acid. The improvement in hepatic function is measured by measuring the levels of plasma transaminases, wherein a decrease in plasma transaminases indicate an improvement in hepatic function.
Also provided are methods of treating a cardiovacular or metabolic disease or disorder selected from diabetes, obesity, hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, hyperfattyacidemia, nonalcoholic fatty liver disease, non alcoholic steatohepatitis, or metabolic syndrome. The animal is treated through the administration of a therapeutically effective amount of an antisense compound essentially complementary to a miR- 122a nucleic acid. W 2
In any of the aforementioned methods, the antisense compound is essentially fully complementary to a miR-122a target nucleic acid. Alternatively, the antisense compound is fully complementary to a miR-122a target nucleic acid. The antisense compound comprises at plurality of sugar modified nucleosides. The plurality of sugar modified nucleosides may, in some embodiments comprise 2 distinct sugar modifications. In certain embodiments the sugar modified nucleosides may further comprise at least one bicyclic sugar modification. In some embodiments, each nucleoside of the plurality of sugar modified nucleosides may comprise a 2'-MOE sugar modification. The antisense compound may comprise at least one phosphorothioate internucleoside linkage. The antisense compound may further comrise a least one 5-methylcytosine. An antisense compound comprising the nucleobase sequence SEQ ID NO: 1 may be used in any of the aforementioned methods. Further, an antisense compound consisting of the nucleobase sequence of SEQ ID NO: 2. may be used in any of the aforementioned methods. The antisense compound may comprise ISIS 327895 or ISIS 387574.
The present invention provides the use of an antisense compound essentially fully complementary to a miR-122 nucleic acid for the preparation of a medicament for lowering a serum indicator of cardiovascular disease risk wherein the serum indicator of cardiovascular disease risk is selected from elevated serum cholesterol levels, elevated serum triglyceride levels, or elevated lipoprotein levels. The elevated serum cholesterol may be elevated LDL-cholesterol, or elevated serum total cholesterol. The elevated serum lipoprotein may be elevated serum ApoB-100. The present invention further provides the use of an antisense compound essentially fully complementary to a miR-122a nucleic acid for the preparation of a medicament for reducing hepatic steatosis. The hepatic steatosis is steatohepatitis or non-alcoholic steatohepatitis.
The present invention additionally provides the use of an antisense compound essentially fully complementary to a miR-122a nucleic acid in the preparation of a medicament to treat a disease or disorder selected from diabetes, obesity, hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, hyperfattyacidemia, nonalcoholic fatty liver disease, non alcoholic steatohepatitis, or metabolic syndrome.
In any of the aforementioned uses, the antisense compound used in the preparation of the medicament is 21 to 23 nucleosides in length. Further, the antisense compound may be fully complementary to a miR-122a nucleic acid. Additionally, the antisense compound may comprise a plurality of 2 '-sugar modified nucleosides. The antisense compound may further comprise at least one bicyclic sugar modified nucleoside. Additionally, the antisense compound comprises the nucleobase sequence of SEQ ID NO: 1. Alternatively, the antisense compound consists of the nucleobase sequence of SEQ ID NO: 2. The antisense compound used in the preparation of the medicament may be uniformly comprised of 2'-MOE sugar modified nucleosides. The antisense compound may further comprise at least W 2
one phosphorothioate intemucleoside linkage. The antisense compound may additionally comprise at least one 5-methylcytosine.
Detailed Description of the Invention It has been found that antisense inhibition of miR-122a resulted in clinically desirable improvements in cholesterol and lipid profiles in animal models of hyperlipidemia and obesity. Accordingly, the present invention provides antisense compounds that hybridize to and inhibit the levels, activity or expression of a miR-122a target nucleic acid, i.e. the antisense compounds are targeted to a miR-122a nucleic acid. The antisense compounds are chemically modified to enhance stability and affinity for a miR-122a nucleic acid. In one embodiment, the antisense compounds comprise a plurality of sugar modified nucleosides. In another embodiment, the plurality of sugar modified nucleosides comprises at least 2 distinct sugar modified nucleosides. Ih some embodiment, the plurality of sugar modified nucleosides comprises at least one bicyclic sugar modified nucleoside. In an additional embodiment, the antisense compounds are uniformly modified such that each nucleoside bears a 2'-MOE modification. In some embodiments, the antisense compound comprises the nucleobase sequence CAAACACCATTGTCACACTCCA (SEQ ID NO: 1). m preferred embodiments, the antisense compound has the nucleobase sequence ACAAACACCATTGTCACACTCCA (SEQ ID NO: 2). In further embodiments, the antisense compound is ISIS 327895 or ISIS 387574.
The present invention provides methods of administering antisense compounds targeted to a miR-122a nucleic acid to an animal to lower serum cholesterol, serum LDL-cholesterol, or serum triglycerides. The present invention also provides methods of reducing hepatic steatosis, liver triglyceride levels and liver weights. Additionally provided are methods of improving liver function, which is evaluated, for example, by decreases in serum transaminases. Also provided are methods of modulating the expression of a miR-122a mRNA target, such as ALDO A. The antisense compounds of the invention are also used for the modulation of metabolic pathways, for example, to lower sterol synthesis, to reduce lipogenesis, reduce fatty acid synthesis rate, reduce sterol synthesis, or increase fatty acid oxidation.
The present invention provides methods for the treatment of diseases or conditions characterized by elevated serum cholesterol levels, elevated serum triglyercide levels, or elevated liver triglyceride levels. These diseases or conditions may be further characterized by compromised liver function as measured by increases in plasma transaminases.
In one embodiment, the methods provided herein are useful for the treatment of diseases or conditions including hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, or hyperfattyacidemia. In a further embodiment, the methods are useful for the treatment of hepatic steatosis or non-alcholic fatty liver disease. In some embodiments, the steatosis is steatohepatitis. In further embodiments, the steatotis is NASH. A further embodiment includes the use of the methods provided for the treatment of metabolic sydrome. In one embodiment, the methods comprise the administration of antisense compounds targeted to miR-122a to an animal, particularly a human, having or susceptible to a disease or condition including, but not limited to, hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, hyperfattyacidemia, steatohepatitis, non-alcoholic steatohepatitis, non-alcholic fatty liver disease, or metabolic syndrome. In another embodiment, the disease or condition is a cardiovascular disease or a metabolic disease. In one aspect, the disease or condition is treated or ameliorated through the administration to the human of a therapeutically effective amount of an antisense compound of the invention. Alternatively, the onset of the disease or condition is delayed or prevented through the administration to the human of a prophylactically effective amount of an antisense compound of the invention.
Embodiments described herein additionally relate to the use of an antisense compound targeted to a miR- 122a nucleic acid in the preparation of a medicament for the treatment of a disease or condition including hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, or hyperfattyacidemia. In further embodiments, the medicament is used for the treatment of hepatic steatosis, which may be steatohepatitis or NASH, or non-alcoholic fatty liver disease. In an additional embodiment, the medicament is used for the treatment of metabolic syndrome. In a further embodiment, the medicament is used for the improvement of liver function. The medicament may, in some embodiments, be administered parenterally, which includes intravenous administration and subcutaneous administration.
Antisense compounds of the invention modulate the levels, activity, or expression of a miR- 122a nucleic acid by hybridizing to a miR-122a nucleic acid {i.e., antisense compounds target the miR- 122a nucleic acids) and thereby interfering with its endogenous function. The hybridization of an antisense compound targeting a miR-122a nucleic acid may trigger the degradation, cleavage, and/or sequestration of the miR- 122a nucleic acid, each of which interferes with the activity of the miR- 122a nucleic acid. The hybridization of an antisense compound targeting miR- 122a may also interfere with the activity of miR-122a by steric occlusion. The inhibition of miR-122a levels, activity or expression that results from an antisense compound hybridizing to a miR- 122a nucleic acid is referred to as "antisense inhibition."
In the context of the present invention, "modulation of a miR- 122a nucleic acid" means either an increase (stimulation) or a decrease (inhibition) in the level, activity, or expression of a miR- 122a nucleic acid. As microRNAs negatively regulate protein-coding nucleic acids, inhibition of a microRNA generally results in stimulation of expression of one or more protein-coding nucleic acids regulated by the microRNA. Inhibition of miR-122a is a preferred form of modulation of a miR-122a nucleic acid. Stimulation is a preferred form of modulation of mRNAs regulated by miR-122a, such as ALDO A.
In one embodiment, the level, activity or expression of a miR- 122a nucleic acid is inhibited to a degree that results in a phenotypic change, such as lowered serum cholesterol or reduced hepatic steatosis. "miR-122a nucleic acid level" or "miR-122a level" indicates the abundance of a miR-122a nucleic acid in a sample, such as an animal cell or tissue. "miR- 122a level" may also indicate the relative abundance of a miR-122a nucleic acid in an experimental sample (e.g., tissue from an animal treated with an antisense compound targeted to miR-122a) as compared to a control sample (e.g., tissue from an untreated animal). "miR-122a activity" refers to the regulation of a protein-coding nucleic acid by miR-122a. "miR-122a expression" refers to the process by which mature miR-122a is derived from a DNA sequence that codes for miR-122a, which includes several steps, for example, transcription, Drosha processing, and Dicer processing. miR-122a expression may be regulated at a single step of this process, or at multiple steps. As used herein, the term "small non-coding RNA" is used to encompass, without limitation, a polynucleotide molecule ranging from about 17 to about 450 nucleosides in length, which can be endogenously transcribed or produced exogenously (chemically or synthetically), but is not translated into a protein. As is known in the art, primary miRNAs (also known as pri-pre-miRNAs, pri-miRs and pri- miRNAs) range from around 70 nucleosides to about 450 nucleosides in length and often take the form of a hairpin structure. The primary miRNA is processed by Drosha to yield a pre-miRNA (also known as pre-mirs and foldback miRNA precursors), which ranges from around 50 nucleosides to around 110 nucleosides in length. The pre-miRNA is in turn processed by Dicer to yield a miRNA (also known as microRNAs, Mirs, miRs, mirs, and mature miRNAs), which ranges from 19 to 24 nucleosides in length. Small non-coding RNAs may include isolated single-, double-, or multiple-stranded molecules, any of which may include regions of intrastrand nucleobase complementarity, said regions capable of folding and forming a molecule with fully or partially double-stranded or multiple-stranded character based on regions of perfect or imperfect complementarity.
As used herein, a "miR-122a nucleic acid" includes pri-miR-122a, pre-miR-122a, and miR- 122a. In the context of the present invention, pri-miR-122a is a primary miRNA, pre-miR-122a is a pre- miRNA, and miR-122a is a mature miRNA. "mature miR-122a" and "miR-122a" may be used interchangably herein.
As used herein, "miR-122a or a precursor thereof encompasses miR-122a, pre-miR-122a, pri- miR-122a, or a primary RNA transcript from which miR-122a or its precursors are derived. As used herein, "miR-122a target nucleic acids" include ρri-miR-122a, pre-miR-122a, and miR-
122a.
In the context of the present invention, miR-122a (i.e. mature miR-122a) has the nucleobase sequence 5'-UGGAGUGUGACAAUGGUGUUUGU-S' (SEQ ID NO: 3). An alternative miR-122a, "miR-122a/b (Tuschl)", has been proposed in the art. miR-122a/b (Tuschl) lacks the 3 '-most nucleoside relative to miR-122a and thus has the nucleobase sequence 5'-UGGAGUGUGACAAUGGUGUUUG-3' (SEQ ID NO: 4). One having ordinary skill in the art would understand that antisense compounds of the invention may target either miR-122a or miR-122a/b Tuschl.
As used herein, the term "miR-122a seed sequence" refers to nucleosides 2 through 8 from the 5'-end of the miR-122a sequence. In the context of the present invention, the miR-122a seed sequence is 5'-GGAGUGU-3' The present invention also provides antisense compounds known as "miR-122a mimics", wherein the antisense compounds include one or more modifications or structural elements or motifs that render the compound capable of mimicking or replacing miR-122a or a precursor thereof.
Therapeutics
The specificity and sensitivity of antisense compounds targeting miR-122a, and compositions thereof, can be harnessed by those of skill in the art for therapeutic uses. Numerous clinical trials testing antisense compounds to other targets in a variety of therapeutic areas are presently underway.
A "human in need of treatment" or "subject in need of treatment" includes humans or subjects diagnosed as having or being susceptible to a disease or condition that can be treated, ameliorated, or prevented by the administration of an antisense compound targeted to a miR-122a nucleic acid. Such diseases or conditions include, but are not limited to, hypercholesterolemia, hypertriglyceridemia, hyperfattyacidemia, or hyperlipidemia, nonalcoholic fatty liver disease, hepatic steatosis (including non- alchoholic steatohepatitis and steatohepatitis), and metabolic syndrome. Subjects in need of treatment may include subjects diagnosed with HCV who also have one or more of the diseases or conditions mentioned herein, such as, for example, hypercholesterolemia or hepatic steatosis. Diagnosis of any of the aforementioned diseases or conditions routinely occurs in a clinical setting, for example, by a physician. Humans or subjects in need of treatment include those who are identified by a medical professional, such as a physician, to have risk factors for cardiovascular disease and/or risk determinants of metabolic syndrome.
As used herein, a "therapeutically effective amount" is an amount of an antisense compound targeted to a miR-122a nucleic acid that, when administered to a human having one or more of the aforementioned diseases or conditions, results in a clinically desirable outcome. Such clinically desirable outcomes may include, without limitation, lowered serum total cholesterol, lowered serum LDL- cholesterol, lowered serum triglycerides, lowered liver triglycerides, reduced steatosis, improved liver function, or increased fatty acid oxidation.
As used herein, a "prophylactically effective amount" is an amount of an antisense compound targeted to a miR-122a nucleic acid that, when administered to a human, prevents or reduces the susceptibility of the human to one or more of the aforementioned diseases or conditions. Prevention or reduction of the susceptibility of the human to the aforementioned diseases or conditions may be accomplished by preventing elevated serum total cholesterol, elevated serum LDL-cholesterol, elevated serum triglycerides, elevated liver triglycerides, hepatic steatosis, aberrant liver function, or reductions in fatty acid oxidation.
As used herein, the term "prevention" means to delay or forestall onset or development of a condition or disease for a period of time, preferably for weeks, months or years. As used herein, the term "amelioration" means a lessening of the severity of a condition or a disease, as evidenced by an improvement of one or more of the surrogate indicators disclosed herein. The improvement of such indicators may be determined by subjective or objective measures which are known to those skilled in the art. As used herein, "treatment" means to administer a composition of the invention to effect an alteration or improvement of the disease or condition. Prevention, amelioration, and/or treatment may require administration of multiple doses of an antisense compound targeted to a miR-122a nucleic acid to alter the course of the condition or disease. Moreover, a single antisense compound may be used in a single individual for the purposes of achieving any combination of prevention, amelioration, or treatment of a condition or disease, and such combinations may be pursued concurrently or sequentially. Antisense compounds of the present invention may, in some instances, be administered with other treatments. A suitable method of administration is parenteral administration, which includes, for example, intravenous administration, subcutaneous administration, and intraperitoneal administration. The present invention provides the use of an antisense compound targeted to a miR-122a nucleic acid for the preparation of a medicament that is administered parenterally, for example, intravenously or subcutaneously. As used herein, the term "serum marker of cardiovascular disease risk" includes risk factors recognized by medical professionals, including those set forth by the National Cholesterol Education Program (NCP), such as elevated serum cholesterol, elevated serum triglycerides, or elevated serum lipoprotein. Serum cholesterol further comprises serum total cholesterol and serum LDL-cholesterol. Serum lipoproteins further comprise serum ApoB-100 protein. In a clinical setting, serum markers of cardiovascular disease risk are measured to determine the need for treatment or prevention of a disease or condition described herein. The guidelines for lipid lowering therapy were established in 2001 by Adult Treatment Panel III (ATP m) of the NCEP, and udpated in 2004 (Grundy et al., Circulation, 2004, 110, 227-239). The guidelines include determining LDL-cholesterol, total cholesterol, and HDL-cholesterol levels (i.e., determining lipoprotein levels). According to the most recently established guidelines, LDL- cholesterol levels of 130-159 mg/dL, 160-189 mg/dL, and greater than or equal to 190 mg/dL are considered borderline high, high, and very high, respectively. Total cholesterol levels of 200-239 and greater than or equal to 240 are considered borderline high and high, respectively. HDL-cholesterol levels of less than 40 are considered low. Serum triglyceride levels of 150-199, 200-499, and greater than or equal to 500 are considered borderline high, high, and very high, respectively. The serum cholesterol level (e.g. LDL-cholesterol, total cholesterol) and/or serum triglyceride level at which treatment is initiated depends upon the presence of clinical atherosclerotic disease that confers a high risk for coronary heart disease, such as clinical coronary heart disease, symptomatic carotid artery disease, peripheral arterial disease, and or abdominal aortic aneurysm, as well as additional risk factors, such as cigarette smoking, hypertension, low HDL-cholesterol, family history of coronary heart disease, and age. A subject' response to treatment is used by a physician to determine the amount and duration of treatment. The NCEP ATP III has also established criteria for the diagnosis of metabolic syndrome when three or more of five risk determinants are present. "Metabolic syndrome" is defined as a clustering of lipid and non-lipid cardiovascular risk factors of metabolic origin. It is closely linked to the generalized metabolic disorder known as insulin resistance. The five risk determinants are abdominal obesity defined as waist circumference of greater than 102 cm for men or greater than 88 cm for women, triglyceride levels greater than or equal to 150 mg/dL, HDL cholesterol levels of less than 40 mg/dL for men and less than 50 mg/dL for women, blood pressure greater than or equal to 130/85 mm Hg and fasting glucose levels greater than or equal to 110 mg/dL. These determinants can be readily measured in clinical practice (JAMA, 2001, 285, 2486-2497).
One of skill in the art will readily appreciate that a physician may modify cardiovascular risk determination for individual patients, in cases where more or less aggressive therapy is needed. One of skill will also understand the scope of the invention includes the practice of the methods herein as applied to any altered guidelines provided by the NCEP, or other entities that establish guidelines for physician's used in treating any of the diseases or conditions listed here, for determining cardiovascular disease risk and diagnosing metabolic syndrome. The embodiments herein provide the use of an antisense compound targeted to a miR-122a nucleic acid in the preparation of a medicament for lowering a serum indicator of cardiovascular disease risk, wherein the serum indicator is selected from elevated serum total cholesterol, elevated serum LDL- cholesterol, or elevated serum triglycerides.
The difficulty in directly measuring miR-122a levels, activity or expression following the administration of antisense compounds targeted to a miR-122a nucleic acid necessitates the use of surrogate indicators to assess the effects of administration of antisense compounds of the invention. Surrogate indicators are used to evaluate phenotypic changes that result from antisense inhibition of miR- 122a. Surrogate indicators are often found in the serum, or alternatively in the plasma, of an animal. Methods of obtaining serum or plasma samples for analysis and methods of preparation of the serum samples to allow for analysis are well known to those skilled in the art. With regard to measurements of lipoproteins, cholesterol, and triglyceride, the terms "serum" and "plasma" are herein used interchangeably.
In the context of the present invention, "surrogate indicators" include serum indicators of cardiovascular disease risk, such as serum LDL-cholesterol levels, and serum total cholesterol levels, and serum triglyceride levels. Serum indicators further include serum transaminase levels. In the context of the present invention, "serum LDL-cholesterol levels" refer to the amounts of cholesterol that are carried in serum LDL particles, and are typically measured as mg/dL or nmol/L. "Serum total cholesterol levels", also expressed as mg/dL, refers to the sum of all different types of serum cholesterol, including, among others, LDL-cholesterol and HDL-cholesterol. Preferred alterations in surrogate indicators of miR-122a inhibition include lowered serum
LDL-cholesterol levels, lowered serum triglyceride levels, lowered lipoprotein levels. These aforementioned alterations are clinically desirable and are useful for the prevention, amelioration and/or treatment of the diseases and disorders disclosed herein.
"Serum lipoproteins" include without limitation apolipoprotein B (ApoB-100), low-density lipoprotein (LDL), and very low-density lipoprotein (VLDL). Methods for the detection of hepatic steatosis are well known in the art, and include magnetic resonance imaging, computed tomography, and ultrasonagraphy. Methods used for the detection of hepatic steatosis may also be used to monitor the prevention, amelioration and/or treatment of the diseases and disorders disclosed herein.
In the context of the present invention, the term "improved hepatic function" or "improved liver function" refers to the improvement in the regular functions performed by the liver of an animal. One non-limiting example of a test for liver function involves the measurement of serum transaminases, which are additional surrogate indicators, wherein a reduction in serum transaminases such as alanine aminotransferase or aspartamine aminotransferase indicates an improvement in liver function. "Serum transaminase levels" refer to the abundance of transaminase in the serum (typically expressed as units /dL), and include "serum alanine aminotransferase levels" and "serum aspartamine aminotransferase levels." Increases in these enzymes are frequently associated with inflammation of the liver or liver cell death and, in some cases, can be caused by a condition such as hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, or hyperlipidemia. Reductions in serum transaminases are clinically desirable and are useful for the prevention, amelioration and/or treatment of the diseases and disorders disclosed herein. The term "nonalcoholic fatty liver disease" (NAFLD) encompasses a disease spectrum ranging from simple triglyceride accumulation in hepatocytes (hepatic steatosis) to hepatic steatosis with inflammation (steatohepatitis), fibrosis, and cirrhosis. Nonalcoholic steatohepatitis (NASH) occurs from progression of NAFLD beyond deposition of triglycerides. A second physiological insult capable of inducing necrosis, inflammation, and fibrosis is required for development of NASH. Candidates for the second insult can be grouped into broad categories: factors causing an increase in oxidative stress and factors promoting expression of proinflammatory cytokines. It has been suggested that increased liver triglycerides lead to increased oxidative stress in hepatocytes of animals and humans, indicating a potential cause-and-effect relationship between hepatic triglyceride accumulation, oxidative stress, and the progression of hepatic steatosis to NASH (Browning and Horton, J. Clin. Invest., 2004, 114, 147- 152). Hypertriglyceridemia and hyperfattyacidemia can cause triglyceride accumulation in peripheral tissues (Shimamura et al., Biochem. Biophys. Res. Commun., 2004, 322, 1080-1085). In the context of the present invention, additional clinically desirable outcomes include reductions in hepatic steatosis, steatohepatisis, fibrosis or cirrhosis.
Diseases or conditions described herein, such as elevated serum LDL-cholesterol or hepatic steatosis, may occur in humans diagnosed with HCV infection. Accordingly, the methods provided herein may also be used in subjects diagnosed with HCV infection. Additionally, the present invention provides the use of an antisense compound targeted to a miR-122a nucleic acid in the preparation of a medicament for lowering serum cholesterol, serum triglycerides or serum lipoproteins in an animal having HCV.
Drug Discovery The antisense compounds and compositions of the present invention can additionally be utilized for research and drug discovery.
For use in research, antisense compounds of the present invention are used to interfere with the normal function of miR-122a target nucleic acids. Expression patterns within cells or tissues treated with one or more antisense compounds or compositions of the invention are compared to control cells or tissues not treated with the antisense compounds or compositions and the patterns produced are analyzed for differential levels of nucleic acid expression as they pertain, for example, to disease association, signaling pathway, cellular localization, expression level, size, structure or function of the genes examined. These analyses can be performed on stimulated or unstimulated cells and in the presence or absence of other compounds that affect expression patterns. By way of example, antisense compounds targeted to miR-122a were used to elucidate the metabolic pathways that are affected by miR-122a regulation.
For use in drug discovery, antisense compounds of the present invention are used to elucidate relationships that exist between miR-122a, or a precursor thereof and a disease state, phenotype, or condition. These methods include detecting or modulating a miR-122a nucleic acid comprising contacting a sample, tissue, cell, or organism with the antisense compounds and compositions of the present invention, measuring the levels of the target miR-122a nucleic acid and/or the levels of downstream gene products including rnRNA or proteins encoded thereby, and further evaluating phenotypic changes at some time after treatment. These methods can also be performed in parallel or in combination with other experiments to determine the function of unknown genes for the process of target validation or to determine the validity of a particular miR-122a target nucleic acid as a target for treatment or prevention of a disease. By way of example, as described herein, antisense compounds targeted to miR-122a were used to effect phenotypic changes such as lowered serum cholesterol, lowered liver triglycerides, and reduced hepatic steatosis.
Antisense Compounds
In the context of the present invention, the term "oligomeric compound(s)" refers to polymeric structures which are capable of hybridizing to at least a region of an RNA molecule. Generally, an oligomeric compound is "antisense" to a target nucleic acid when, written in the 5' to 3' direction, it comprises the reverse complement of the corresponding region of the target nucleic acid. Such oligomeric compounds are known as "antisense compounds", which include, without limitation, oligonucleotides
{i.e. antisense oligonucleotides), oligonucleosides, oligonucleotide analogs, oligonucleotide mimetics and combinations of these. In general, an antisense compound comprises a backbone of linked monomelic subunits (sugar moieties) where each linked monomelic subunit is directly or indirectly attached to a heterocyclic base moiety. Modifications to antisense compounds encompass substitutions or changes to internucleoside linkages, sugar moieties, or heterocyclic base moieties, such as those described below. As used herein, the term "modification" includes substitution and/or any change from a starting or natural nucleoside or nucleotide. Modified antisense compounds are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target, increased stability in the presence of nucleases, or increased inhibitory activity. Antisense compounds are often defined in the art to comprise the further limitation of, once hybridized to a target, being able to induce or trigger a reduction in target gene expression or target gene activity. La one embodiment, the antisense compounds trigger a reduction in the levels, activity or expression of a miR- 122a nucleic acid target.
Antisense compounds are routinely prepared linearly but can be joined or otherwise prepared to be circular and may also include branching. Separate antisense compounds can hybridize to form double stranded compounds that can be blunt-ended or may include overhangs on one or both termini.
The antisense compounds in accordance with this invention comprise from 15 to 30 nucleosides in length, i.e., from 15 to 30 linked nucleosides. One of skill in the art will appreciate that this embodies antisense compounds of 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleosides in length. In one embodiment, the antisense compounds of the invention are 17 to 25 nucleosides in length, as exemplified herein.
In preferred embodiments, the antisense compounds of the invention are 19, 20, 21, 22 or 23 nucleosides in length.
As used herein, the term "about" means ± 5% of the variable thereafter. "Complementary," as used herein, refers to the capacity for hybridization between nucleobases.
An antisense compound and the target nucleic acid are "fully complementary" to each other when each nucleobase of the antisense compound is complementary to an equal number of nuclebases in the target nucleic acid. For example, an antisense compound 23 nucleosides in length targeted to miR-122a is fully complementary to miR-122a when each of the 23 nucleobases in the antisense compound is complementary to miR-122a. The antisense compound and the target nucleic acid are "essentially fully complementary" to each other when the degree of precise base pairing permits stable and specific binding between the antisense compound and a target nucleic acid, so that the antisense compound inhibits the level, activity or expression of the target nucleic acid. Antisense compounds having one or two non- complementary nucleobases with respect to a miR-122a target nucleic acid are considered essentially fully complementary. "Sufficiently complementary" may be used in place of "essentially fully complementary". In the context of this invention, "hybridization" means the pairing of nucleobases of an antisense compound with corresponding nucleobases in a target nucleic acid. In the context of the present invention, the mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between corresponding nucleobases. For example, adenine and thymine are complementary nucleobases that pair through the formation of hydrogen bonds. Both natural and modified nucleobases are capable of participating in hydrogen bonding. Hybridization can occur under varying circumstances.
It is understood in the art that the sequence of the antisense compound need not be fully complementary to that of its target nucleic acid to be active in inhibiting the activity of the target nucleic acid. Moreover, an antisense compound may hybridize over one or more segments such that intervening or adjacent segments are not involved in the hybridization (e.g., a bulge, a loop structure or a hairpin structure). In some embodiments there are "non-complementary" positions, also known as "mismatches", between the antisense compound and the target nucleic acid, and such non-complementary positions may be tolerated between an antisense compound and the target nucleic acid provided that the antisense compound remains specifically hybridizable to the target nucleic acid. A "non-complementary nucleobase" means a nucleobase of an antisense compound that is unable to undergo precise base pairing with a nucleobase at a corresponding position in a target nucleic acid. As used herein, the terms "non- complementary" and "mismatch" are interchangable. Up to 3 non-complementary nucleobases are often tolerated in an antisense compound without causing a significant decrease in the ability of the antisense compound to modulate the activity, level or function of a target nucleic acid. In preferred embodiments, the antisense compound contains no more than 2, or no more than 1 , non-complementary nucleobases with respect to a miR-122a target nucleic acid. For example, an antisense compound 23 nucleosides in length, 22 of which are able to undergo precise base pairing with nucleobases in corresponding positions in a target nucleic acid, and one of which is not able to undergo such base pairing, is considered to have one non-complementary nucleobase. The location of such a non-complementary nucleobase at the 5 ' end or 3' end of the antisense compound is preferred, however, the non-complementary nucleobase may be at any position in the antisense compound. In some embodiments, non-complementary nucleobases are preferred outside of the region of the antisense compound which is complementary to the seed sequence of miR-122a. When two or more non-complementary nucleobases are present, they may be contiguous (i.e. linked) or non-contiguous.
In other embodiments of the invention, the antisense compounds comprise at least 90% sequence complementarity to a miR-122a target nucleic acid. In further embodiments of the invention, the antisense compounds comprise at least 95% sequence complementarity to a miR-122a target nucleic acid. For example, an antisense compound in which 22 of 23 nucleobases of the antisense compound are complementary (i.e. one nucleobase is non-complementary) to a miR-122a nucleic acid would represent 95.6 % complementarity. Likewise, an antisense compound in which 21 of the 23 nucleobases are complementary (i.e. two nucleobases are non-complementary) to a miR-122a nucleic acid would represent 91.3 % complementarity. Percent complementarity of an antisense compound with a region of a target nucleic acid can be determined routinely by those having ordinary skill in the art, and may be accomplished using BLAST programs (basic local alignment search tools) and PowerBLAST programs known in the art (Altschul et al., J. MoL Biol, 1990, 215, 403-410; Zhang and Madden, Genome Res., 1997, 7, 649-656).
Li the present invention the phrase "stringent hybridization conditions" or "stringent conditions" refers to conditions under which an antisense compound of the invention will hybridize to its target sequence, but to a minimal number of other sequences. Stringent conditions are sequence-dependent and will vary with different circumstances and in the context of this invention; "stringent conditions" under which antisense compounds hybridize to a target sequence are determined by the nature and composition of the antisense compounds and the assays in which they are being investigated. One having ordinary skill in the art will understand variability in the experimental protocols and be able to determine when conditions are optimal for stringent hybridization with minimal non-specific hybridization events. Antisense compounds may have a defined percent identity to a SEQ ID NO, or an antisense compound having a specific ISIS number. This identity may be over the entire length of the antisense compound, or over less than the entire length of the antisense compound. It is understood by those skilled in the art that an antisense compound need not have an identical sequence to those described herein to function similarly to the antisense compounds described herein. Shortened or truncated versions of antisense compounds taught herein have one, two or more nucleosides deleted, and fall within the scope of the invention. When an antisense compound has two or more deleted nucleosides, the deleted nucleosides may be adjacent to each other, for example, in an antisense compound having two nucleosides truncated from the 5' end (5' truncation), or alternatively from the 3' end (3' truncation), of the antisense compound. Alternatively, the delelted nucleosides may be dispersed through out the antisense, for example, in an antisense compound having one nucleoside deleted from the 5' end and one nucleoside deleted from the 3' end.
Also falling within the scope of the invention are lengthened versions of antisense compounds taught herein, i.e. antisense compounds having one or more additional nucleosides relative to an antisense compound disclosed herein. When two are more additional nucleosides are present, the added nucleosides may be adjacent to each other, for example, in an antisense compound having two nucleosides added to the 5' end (5' addition), or alternatively to the 3' end (3' addition), of the antisense compound. Alternatively, the added nucleosides may be dispersed throughout the antisense compound, for example, in an antisense compound having one nucleoside added to the 5' end and one nucleoside added to the 3' end. Percent identity of an antisense compound is calculated according to the number of nucleobases that are identical to the SEQ ID NO or antisense compound to which it is being compared. Such calculations are well within the ability of those skilled in the art. For example, a 21 nucleoside antisense compound having the same nucleobase sequence as nucleosides 2-21 of a 23 nucleoside antisense compound is 91.3% identical to the 23 nucleoside antisense compound. Alternatively, a 24 nucleoside antisense compound that differs in sequence only by the addition of one nucleoside relative to a 23 nucleoside antisense compound may have 100% identity over 23 nucleosides, but 95.8 % identity overall. "Targeting" an antisense compound to a particular nucleic acid molecule, including a miR-122a nucleic acid, in the context of this invention, can be a multistep process. The process usually begins with the identification of a target nucleic acid whose levels, expression or function is to be modulated. In the context of the present invention, the target nucleic acid is a miR-122a target nucleic acid. The targeting process usually also includes determination of at least one target segment within a miR-122a target nucleic acid for the interaction to occur such that the desired effect, e.g., modulation of levels, activity, or expression, will result. As used herein, a "target segment" means a sequence of a miR- 122a nucleic acid to which one or more antisense compounds are complementary. Multiple antisense compounds complementary to a given target segment may or may not have overlapping sequences. Within the context of the present invention, the term "target site" is defined as a sequence of a miR-122a nucleic acid to which one nucleobase sequence is complementary. For example, the nucleobase sequence of SEQ ID NO: 8 is complementary to target site 27 to 49 of SEQ ID NO: 5, and the nucleobase sequence of SEQ ID NO: 2 is complementary to target site 29 to 51 of SEQ NO: 5. Nucleosides 27 to 51 of SEQ ID NO: 5 thus represent a target segment of SEQ ID NO: 5. In some embodiments, a target segment and target site will be represented by the same nucleobase sequence.
Target sites and target segments may also be found in an miRNA gene from which a pri-miR- 122a is derived, which may be found as a solitary transcript, or it may be found within a 5' untranslated region (5'UTR), within in an intron, or within a 3' untranslated region (3 TJTR) of a gene.
Antisense compounds of the invention may be also be described as complementary to a portion of a target site. A "portion" is defined as at least 18 contiguous nucleosides of a target site. In other embodiments, a portion is 19 or 20 contiguous nucleosides of a target site. In preferred embodiments, a portion is 21 or 22 contiguous nucleosides of a target site.
Once one or more target segments or target sites have been identified, antisense compounds are designed to be sufficiently complementary to the target segments or target sites, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired effect. The desired effect may include, but is not limited to modulation of the levels, expression or activity of the a miR-122a target nucleic acid. Desired effects further include phenotypic changes.
The antisense compounds of the invention may be in the form of single-stranded, double- stranded, circular or hairpin antisense compounds and may contain structural elements such as internal or terminal bulges or loops. Once introduced to a system, the antisense compounds of the invention may elicit or inhibit the action of one or more enzymes or proteins to effect modulation of the levels, W
expression or function of the target nucleic acid. One non-limiting example of such a protein is the Drosha RNase HI enzyme. A further non-limiting example involves the enzymes of the RISC complex.
Antisense compounds or compositions of the invention may be used to induce potent and specific modulation of gene function through interactions with or mimicry of small non-coding RNAs that are processed by the RISC complex. These compounds include single-stranded antisense compounds that bind in a RISC complex, double-stranded antisense/sense pairs of antisense compounds, or single- stranded antisense compounds that include both an antisense portion and a sense portion.
Oligonucleotide Synthesis Antisense compounds and phosphoramidites are made by methods well known to those skilled in the art. Oligomerization of modified and unmodified nucleosides is performed according to literature procedures for DNA like compounds (Protocols for Oligonucleotides and Analogs, Ed. Agrawal (1993),
Humana Press) and/or RNA like compounds (Scaringe, Methods (2001), 23, 206-217. Gait et al.,
Applications of Chemically synthesized RNA in RNA:Protein Interactions, Ed. Smith (1998), 1-36. Gallo et al., Tetrahedron (2001), 57, 5707-5713) synthesis as appropriate. RNA oligomers can be synthesized by methods disclosed herein or purchased from various RNA synthesis companies such as for example
Dharmacon Research Inc., (Lafayette, CO).
Irrespective of the particular protocol used, the antisense compounds used in accordance with this invention may be conveniently and routinely made through the well-known technique of solid phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied
Biosystems (Foster City, CA). Any other means for such synthesis known in the art may additionally or alternatively be employed.
Methods of isolation and analysis of oligonucleotides are well known in the art. A 96-well plate format is particularly useful for the synthesis, isolation and analysis of oligonucleotides.
RNA Synthesis
Methods of RNA synthesis are well known in the art (Scaringe, S. A. Ph.D. Thesis, University of Colorado, 1996; Scaringe, S. A., et al., J. Am. Chem. Soc, 1998, 120, 11820-11821; Matteucci, M. D. and Carathers, M. H. J. Am. Chem. Soc, 1981, 103, 3185-3191; Beaucage, S. L. and Caruthers, M. H. Tetrahedron Lett, 1981, 22, 1859-1862; Dahl, B. J., et al., Acta Chem. Scand,. 1990, 44, 639-641 ;
Reddy, M. P., et al., Tetrahedrom Lett, 1994, 25, 4311-4314; Wincott, F. et al., Nucleic Acids Res., 1995,
23, 2677-2684; Griffin, B. E., et al., Tetrahedron, 1967, 23, 2301-2313; Griffin, B. E., et al., Tetrahedron,
1967, 23, 2315-2331).
Antisense Compound Modifications Any of the antisense compounds taught herein may contain modifications which confer desirable pharmacokinetic and/or pharmacodynamic properties to the antisense compound including, but are not limited to, improved binding affinity, stability, charge, localization or uptake. Further, modified synthetic antisense compounds of the present invention may be designed to mimic endogenous small non- coding RNAs.
As is known in the art, a nucleoside is a base-sugar combination. The base (also known as nucleobase) portion of the nucleoside is normally a heterocyclic base moiety. The two most common classes of such heterocyclic bases are purines and pyrimidines. Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a pentofuranosyl sugar, the phosphate group can be linked to the 2', 3' or 5' hydroxyl moiety of the sugar. In forming oligonucleotides, the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound. The respective ends of this linear polymeric structure can be joined to form a circular structure by hybridization or by formation of a covalent bond. In addition, linear compounds may have internal base (i.e. nucleobase) complementarity and may therefore fold in a manner as to produce a fully or partially double-stranded structure. Within the oligonucleotide structure, the phosphate groups are commonly referred to as forming the internucleoside linkages of the oligonucleotide. The normal internucleoside linkage of RNA and DNA is a 3' to 5' phosphodiester linkage.
In the context of this invention, the term "oligonucleotide" refers generally to an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA), and may be used to refer to unmodified oligonucleotides or oligonucleotide analogs. The term "unmodified oligonucleotide" refers generally to oligonucleotides composed of naturally occuring nucleobases, sugars, and covalent internucleoside linkages. The term "oligonucleotide analog" refers to oligonucleotides that have one or more non-naturally occurring nucleobases, sugars, and/or internucleoside linkages. Such non-naturally occurring oligonucleotides are often selected over naturally occurring forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for other oligonucleotides or nucleic acid targets, increased stability in the presence of nucleases, or increased inhibitory activity. Modified Internucleoside Linkages
Specific examples of antisense compounds useful in this invention include oligonucleotides containing modified, i.e. non-naturally occurring, internucleoside linkages. Such non-naturally internucleoside linkages are often selected over naturally occurring forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for other oligonucleotides or nucleic acid targets and increased stability in the presence of nucleases.
Antisense compounds of the invention can have one or more modified internucleoside linkages. As defined in this specification, oligonucleotides having modified internucleoside linkages include internucleoside linkages that retain a phosphorus atom and internucleoside linkages that do not have a phosphorus atom. For the purposes of this specification, and as sometimes referenced in the art, modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be "oligonucleosides".
A suitable phosphorus-containing modified internucleoside linkage is the phosphorothioate internucleoside linkage. Additional modified internucleoside linkages containing a phosphorus atom therein include, for example, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3'-alkylene phosphonates, 5'- alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3 '-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3'-5' linkages, 2'-5' linlced analogs of these, and those having inverted polarity wherein one or more internucleoside linkages is a 3' to 3', 5' to 5' or 2' to 2' linkage. Oligonucleotides having inverted polarity comprise a single 3' to 3' linkage at the 3'-most internucleoside linkage i.e. a single inverted nucleoside residue which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof). Various salts, mixed salts and free acid forms are also included.
Representative U.S. patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S.: 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,194,599; 5,565,555; 5,527,899; 5,721,218; 5,672,697 and 5,625,050, each of which is herein incorporated by reference.
In other embodiments of the invention, antisense compounds have one or more phosphorothioate and/or heteroatom internucleoside linkages, in particular -CH2-NH-O-CH2-, -CH2- N(CH3)-O-CH2- (known as a methylene (methylimino) or MMI backbone), -CH2-O-N(CHa)-CH2-, -CH2- N(CH3)-N(CH3)-CH2- and -O-N(CH3)-CH2-CH2- (wherein the native phosphodiester internucleoside linkage is represented as -O-P(=O)(OH)-O-CH2-). The MMI type internucleoside linkages are disclosed in the above referenced U.S. patent 5,489,677. Amide internucleoside linkages are disclosed in the above referenced U.S. patent 5,602,240.
Modified internucleoside linkages that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; riboacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts.
Representative U.S. patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S.: 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; 5,792,608; 5,646,269 and 5,677,439, each of which is herein incorporated by reference.
Antisense compounds can also include an "oligonucleotide mimetic", which refers to oligonucleotides in which only the furanose ring or both the furanose ring and the internucleoside linkage are replaced with novel groups. Replacement of only the furanose ring is also referred to in the art as being a sugar surrogate. The heterocyclic base moiety or a modified heterocyclic base moiety is maintained for hybridization with an appropriate target nucleic acid. Oligonucleotide mimetics can include antisense compounds containing peptide nucleic acid (PNA) modifications. In PNA antisense compounds, the sugar-backbone of an oligonucleotide is replaced with an amide containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative U.S. patents that teach the preparation of PNA antisense compounds include, but are not limited to, U.S.: 5,539,082; 5,714,331; and 5,719,262, each of which is herein incorporated by reference. Teaching of PNA antisense compounds can be found in Nielsen et al, Science, 1991, 254, 1497-1500. PNA has been modified to incorporate numerous modifications since the basic PNA structure was first prepared. Another class of oligonucleotide mimetic that has been studied is based on linked morpholino units (morpholino nucleic acid) having heterocyclic bases attached to the morpholino ring. A number of linking groups have been reported that link the morpholino monomelic units in a morpholino nucleic acid. Morpholino-based antisense compounds are disclosed in U.S. Patent 5,034,506, issued July 23, 1991. The morpholino class of antisense compounds have been prepared having a variety of different linking groups joining the monomelic subunits.
Another class of oligonucleotide mimetic is referred to as cyclohexenyl nucleic acids (CeNA). The furanose ring normally present in an DNA/RNA molecule is replaced with a cyclohenyl ring. CeNA DMT protected phosphoramidite monomers have been prepared and used for antisense compound synthesis following classical phosphoramidite chemistry. Fully modified CeNA antisense compounds and oligonucleotides having specific positions modified with CeNA have been prepared and studied (see Wang et al., J. Am. Chem. Soc, 2000, 122, 8595-8602). Modified Sugar Moieties
Antisense compounds of the invention may also contain one or more modified or substituted sugar moieties. The base moieties (natural, modified or a combination thereof) are maintained for hybridization with an appropriate nucleic acid target. Sugar modifications may impart nuclease stability, binding affinity or some other beneficial biological property to the antisense compounds. Representative modified sugars include carbocyclic or acyclic sugars, sugars having substituent groups at one or more of their T, 3' or 4' positions, sugars having substituents in place of one or more hydrogen atoms of the sugar, and sugars having a linkage between any two other atoms in the sugar. Antisense compounds of particular use in the instant invention may comprise a sugar substituent group selected from: OH; halo; O-, S-, or N- allcyl; O-, S-, or N-alkenyl; O-, S- or N-alkynyl; or O-alkyl-0-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C1 to C10 alkyl or C2 to Ci0 alkenyl and alkynyl. Particularly suitable are O((CH2)nO)mCH3, O(CH2)nOCH3, O(CH2)nNH2, O(CH2)nCH3, O(CH2)nONH2, and O(CH2)nON((CH2)nCH3)2, where n and m are from zero to about 10. Some oligonucleotides comprise a sugar substituent group selected from: Ci to C]0 lower alkyl, substituted lower allcyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, F, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties. One modification that imparts increased nuclease resitance and a very high binding affinity to nucleosides is the 2'-methoxyethoxy (2'-MOE or 2'-OCH2CH2OCH3) side chain (Baker et al., J. Biol. Chem., 1997, 272, 11944-12000). One of the immediate advantages of the 2'-MOE substitution is the improvement in binding affinity, which is greater than many similar 2' modifications such as (9-methyl, 0-propyl, and O-aminopropyl. Oligonucleotides having the 2'-0-methoxyethyl substituent also have been shown to be antisense inhibitors of gene expression with promising features for in vivo use (Martin, P., HeIv. Chim. Acta, 1995, 78, 486-504; Altmann et al., Chimia, 1996, 50, 168-176; Altmann et al., Biochem. Soc. Trans., 1996, 24, 630-637; and Altmann et al., Nucleosides Nucleotides, 1997, 16, 917- 926). Relative to DNA, the oligonucleotides having the 2'-MOE modification displayed improved RNA affinity and higher nuclease resistance. Antisense compounds having one or more 2'-MOE modifications are capable of inhibiting miRNA activity in vitro and in vivo (Esau et al., J. Biol. Chem., 2004, 279, 52361-52365; U.S. Application Publication No. 2005/0261218).
Additional modifications include 2'-dimethylaminooxyethoxy, i.e., a O(CH2)2ON(CH3)2 group, also known as 2'-DMAOE, and 2'-dimethylaminoethoxyethoxy (also known in the art as 2'-O-dimethyl- amino-ethoxy-ethyl or 2'-DMAEOE), i.e., 2'-O-CH2-O-CH2-N(CH3)2. Other sugar substituent groups include methoxy (-O-CH3), aminopropoxy (-OCH2CH2CH2NH2), allyl (-CH2-CH=CH2), and -O-allyl (-0-CH2-CH=CH2). 2'-Sugar substituent groups may be in the arabino (up) position or ribo (down) position. Similar modifications may also be made at other positions on the antisense compound, particularly the 3' position of the sugar on the 3' terminal nucleoside or in T- 5' linked oligonucleotides and the 5' position of 5' terminal nucleoside. Antisense compounds may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative U.S. patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S.: 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785;
5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265;
5,658,873; 5,670,633; 5,792,747; and 5,700,920, each of which is herein incorporated by reference in its entirety. Representative substituents groups are disclosed in U.S. Patent No. 6,172,209 entitled "Capped
2'-Oxyethoxy Oligonucleotides," hereby incorporated by reference in its entirety.
Representative cyclic substituent groups are disclosed in U.S. Patent No. 6,271,358 entitled
"RNA Targeted 2'-Oligomeric compounds that are Confoπnationally Preorganized," hereby incorporated by reference in its entirety. Representative guanidino substituent groups are disclosed in U.S. Patent 6,593,466 entitled
"Functionalized Oligomers," hereby incorporated by reference in its entirety.
Representative acetamido substituent groups are disclosed in U.S. Patent 6,147,200 which is hereby incorporated by reference in its entirety.
Representative dimethylaminoethyloxyethyl substituent groups are disclosed in International Patent Application PCT/US99/17895, entitled '^'-O-Dimethylaminoethyloxyethyl-Oligomeric compounds", filed August 6, 1999, hereby incorporated by reference in its entirety.
An additional sugar modification includes a bicyclic sugar moiety, which has a 2', 4' bridge that forces the sugar ring into a locked 3'-endo conformational geometry. Bicyclic modifications imparts to an antisense compound greatly increased affinity for a nucleic acid target. Furthermore, nucleosides having bicyclic sugar modifications can act cooperatively with DNA and RNA in chimeric antisense compounds to enhance the affinity of a chimeric antisense compound for a nucleic acid target. Bicyclic sugar moieties can be represented by the formula 4'-(CH2)n-X-2', where X can be, for example, O or S. LNA™ is a bicyclic sugar moiety having a 4'-CH2-O-2' bridge (i.e. X is O and n is 1). The alpha-L nucleoside has also been reported wherein the linkage is above the ring and the heterocyclic base is in the alpha rather than the beta-conformation (see U.S. Patent Application Publication No.: Application 2003/0087230).
The xylo analog has also been prepared (see U.S. Patent Application Publication No.: 2003/0082807).
Another bicyclic sugar moiety is ENA™, where in the aforementioned formula X is O and n is 2. The term 'locked nucleic acid' can also be used to describe any bicyclic sugar moiety that has a "locked" conformation. Antisense compounds incorporating LNA™ and ENA™ analogs display very high duplex thermal stabilities with complementary DNA and RNA (Tm = +3 to +10 C), stability towards 3'- exonucleolytic degradation and good solubility properties.
The synthesis and preparation of the LNA™ monomers adenine, cytosine, guanine, 5-methyl- cytosine, thymine and uracil, along with their oligomerization, and nucleic acid recognition properties have been described (Koshkin et al., Tetrahedron, 1998, 54, 3607-3630). LNAs and preparation thereof are also described in WO 98/39352 and WO 99/14226. Analogs of LNA™, such as phosphorothioate-LNA and 2'-thio-LNAs (i.e. 2'-S-CH2-4'), have also been prepared (Kumar et al., Bioorg. Med. Chem. Lett., 1998, 8, 2219-2222). Preparation of locked nucleoside analogs containing oligodeoxyribonucleotide duplexes as substrates for nucleic acid polymerases has also been described (Wengel et al., PCT International Application WO 98-DK393 19980914). Furthermore, synthesis of 2'-amino-LNA, a novel conformationally restricted high-affinity oligonucleotide analog with a handle has been described in the art (Singh et al., J. Org. Chem., 1998, 63, 10035-10039). In addition, 2'-Amino- and 2'-methylamino-LNA's have been prepared and the thermal stability of their duplexes with complementary RNA and DNA strands has been previously reported.
Some oligonucleotide mimetics have been prepared to include bicyclic and tricyclic nucleoside analogs (see Steffens et al., HeIv. Chim. Acta, 1997, 80, 2426-2439; Steffens et al., J. Am. Chem. Soc, 1999, 121, 3249-3255; and Renneberg et al., J. Am. Chem. Soc, 2002, 124, 5993-6002). These modified nucleoside analogs have been oligomerized using the phosphoramidite approach and the resulting antisense compounds containing tricyclic nucleoside analogs have shown increased thermal stabilities (Tm' s) when hybridized to DNA, RNA and itself. Antisense compounds containing bicyclic nucleoside analogs have shown thermal stabilities approaching that of DNA duplexes.
Another class of oligonucleotide mimetic is referred to as phosphonomonoester nucleic acid and incorporates a phosphorus group in the backbone. This class of oligonucleotide mimetic is reported to have useful physical and biological and pharmacological properties in the areas of inhibiting gene expression. Nucleobase Modifications
Antisense compounds of the invention may also contain one or more nucleobase (often referred to in the art simply as "base") modifications or substitutions which are structurally distinguishable from, yet functionally interchangeable with, naturally occurring or synthetic unmodified nucleobases. Such nucleobase modifications may impart nuclease stability, binding affinity or some other beneficial biological property to the antisense compounds. As used herein, "unmodified" or "natural" nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleobases also referred to herein as heterocyclic base moieties include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2- propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl (-C≡C-CH.3) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8- amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7- methyladenine, 2-F-adenine, 2-amino-adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7- deazaadenine and 3-deazaguanine and 3-deazaadenine.
Heterocyclic base moieties 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. Some nucleobases include those disclosed in U.S. Patent No. 3,687,808, those disclosed in
The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J.I., ed.
John Wiley & Sons, 1990, those disclosed by Englisch et al., Angewandte Chemie, International Edition,
1991, 30, 613, and those disclosed by Sanghvi, Y.S., Chapter 15, Antisense Research and Applications, pages 289-302, Crooke, S.T. and Lebleu, B. , ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the antisense compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including
2 aminopropyladenine, 5-propynyluracil and 5-propynylcytosine.
In one aspect of the present invention antisense compounds are prepared having polycyclic heterocyclic compounds in place of one or more heterocyclic base moieties. A number of tricyclic heterocyclic compounds have been previously reported. These compounds are routinely used in antisense applications to increase the binding properties of the modified strand to a target strand. The most studied modifications are targeted to guanosines, and have been termed G-clamps or cytidine analogs.
Representative cytosine analogs that make 3 hydrogen bonds with a guanosine in a second strand include l,3-diazaphenoxazine-2-one (Rio= O, Rn - Rw= H) (Kurchavov, et al., Nucleosides and Nucleotides, 1997, 16, 1837-1846), l,3-diazaphenothiazine-2-one (R10= S, Rn - Ri4= H), (Lin, K.-Y.;
Jones, R. J.; Matteucci, M. J. Am. Chem. Soc. 1995, 117, 3873-3874) and 6,7,8,9-tetrafluoro-l,3- diazaphenoxazine-2-one (Ri0 = O, Rn - Rj4 = F) (Wang, J.; Lin, K.-Y., Matteucci, M. Tetrahedron Lett.
1998, 39, 8385-8388). When incorporated into oligonucleotides, these base modifications were shown to hybridize with complementary guanine and the latter was also shown to hybridize with adenine and to enhance helical thermal stability by extended stacking interactions (also see U.S. Patent Application
Publication 20030207804 and U.S. Patent Application Publication 20030175906, both of which are incorporated herein by reference in their entirety).
Tricyclic heterocyclic compounds and methods of using them that are amenable to the present invention are disclosed in U.S. Patent 6,028,183, and U.S. Patent 6,007,992, the contents of both are incorporated herein in their entirety.
The enhanced binding affinity of the phenoxazine derivatives together with their sequence specificity makes them valuable nucleobase analogs for the development of more potent antisense-based drugs. In fact, promising data have been derived from in vitro experiments demonstrating that heptanucleotides containing phenoxazine substitutions can activate RNaseH, enhance cellular uptake and exhibit an increased antisense activity (Lin, K-Y; Matteucci, M. J. Am. Chem. Soc. 1998, 120, 8531-
8532). The activity enhancement was even more pronounced in case of G-clamp, as a single substitution was shown to significantly improve the in vitro potency of a 20 nucleoside 2'-deoxyphosphorothioate oligonucleotides (Flanagan, W. M.; Wolf, J.J.; Olson, P.; Grant, D.; Lin, K.-Y.; Wagner, R. W.; Matteucci, M. Proc. Natl. Acad. Sci. USA, 1999, 96, 3513-3518).
Modified polycyclic heterocyclic compounds useful as heterocyclic bases are disclosed in but not limited to, the above noted U.S. 3,687,808, as well as U.S.: 4,845,205; 5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,434,257; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; 5,645,985; 5,646,269; 5,750,692; 5,830,653; 5,763,588; 6,005,096; and 5,681,941, and U.S. Patent Application Publication 20030158403, each of which is incorporated herein by reference in its entirety. Certain nucleobase substitutions, including 5-methylcytosinse substitutions, are particularly useful for increasing the binding affinity of the oligonucleotides of the invention. For example, 5- methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.20C (Sanghvi, Y.S., Crooke, S.T. and Lebleu, B., eds., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are presently preferred base substitutions, even more particularly when combined with 2'-O-methoxyethyl sugar modifications. Conjugated Antisense Compounds
One substitution that can be appended to the antisense compounds of the invention involves the linkage of one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the resulting antisense compounds. In one embodiment such modified antisense compounds are prepared by covalently attaching conjugate groups to functional groups such as hydroxyl or amino groups. Conjugate groups of the invention include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of the antisense compounds. Typical conjugate groups include cholesterol moieties and lipid moieties. Additional conjugate groups include carbohydrates, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes. Groups that enhance the pharmacodynamic properties, in the context of this invention, include groups that impart to the antisense compound properties such as improved uptake, enhanced resistance to degradation, and/or enhance hybridization with RNA. Groups that enhance the pharmacokinetic properties, in the context of this invention, include groups impart to the antisense compound properties such as improved uptake, distribution, metabolism or excretion.
Representative conjugate groups are disclosed in International Patent Application PCT/US92/09196, filed October 23, 1992 the entire disclosure of which is incorporated herein by reference.
Furthermore, the antisense compounds of the invention can have one or more conjugated moieties to facilitate the active or passive transport, localization, or compartmentalization of the antisense compound. Cellular localization includes, but is not limited to, localization to within the nucleus, the nucleolus, or the cytoplasm. Compartmentalization includes, but is not limited to, any directed movement of the antisense of the invention to a cellular compartment including the nucleus, nucleolus, mitochondrion, or imbedding into a cellular membrane. Furthermore, the antisense compounds of the invention comprise one or more conjugate moieties which facilitate posttranscriptional modification.
Antisense compounds used in the compositions of the present invention can also be modified to have one or more stabilizing groups that are generally attached to one or both termini of antisense compounds to enhance properties such as, for example, nuclease stability. Included in stabilizing groups are cap structures. By "cap structure" or "terminal cap moiety" is meant chemical modifications, which have been incorporated at either terminus of an antisense compound (see for example Wincott et al., WO 97/26270, incorporated by reference herein). These terminal modifications protect the antisense compounds having terminal nucleic acid molecules from exonuclease degradation, and can help in delivery and/or localization within a cell. The cap can be present at the 5'-terminus (5'-cap), or at the 3'- terminus (3 '-cap), or can be present on both termini. For double-stranded antisense compounds, the cap may be present at either or both termini of either strand. Cap structures are well known in the art and include, for example, inverted deoxy abasic caps. Further 3' and 5 '-stabilizing groups that can be used to cap one or both ends of an antisense compound to impart nuclease stability include those disclosed in WO 03/004602 published on January 16, 2003.
Antisense Compound Motifs
Antisense compounds of this invention may have the chemically modified subunits arranged in patterns enhance the inhibitory activity of the antisense compounds. These patterns are described herein as "motifs", and include uniformly modified, positionally modified, gapmer or gapped, alternating, hemimer, and blockmer.
As used in the present invention the term "uniform motif is meant to include antisense compounds wherein each nucleoside bears the same type of sugar moiety, which may be a naturally occuring sugar or a modified sugar. Further, "uniformly modified motif," "uniform modifications," and "uniformly modified" are meant to include antisense compounds wherein each nucleoside bears the same type of sugar modification. Suitable sugar moieties include, but are not limited to 2'-O(CH2)2θCH3 [2'- MOE], 2'-OCH3 [2'-O-methyl], LNA and ENA™. For example, an antisense compound may be uniformly modified such that each sugar modification is a 2'-MOE sugar modification. Alternatively, an antisense compound may be uniformly modified such that each sugar modification is a 2'-O-methyl sugar modification.
As used in the present invention the term "gapped motif or "gapmer" is meant to include an antisense compound having an internal region (also referred to as a "gap" or "gap segment") positioned between two external regions (also referred to as "wing" or "wing segment"). The regions are differentiated by the types of sugar moieties comprising each distinct region. The types of sugar moieties that are used to differentiate the regions of a gapmer include β-D-ribonucleosides, β-D- deoxyribonucleosides, 2'-modified nucleosides (such 2 '-modified nucleosides may include 2'-MOE, and 2'-O-CH3, among others), and bicyclic sugar modified nucleosides (such bicyclic sugar modified nucleosides may include LNA™ or ENA™, among others). In general, each distinct region has a uniform motif sugar moieties within a region are uniform. Gapped motifs or gapmers are further defined as being either "symmetric" or "asymmetric". A gapmer wherein the nucleosides of the first wing have the same sugar modifications as the nucleosides of the second wing is termed a symmetric gapped antisense compound. Symmetric gapmers can have, for example, an internal region comprising a first sugar moiety, and external regions each comprising a second sugar moiety, wherein at least one sugar moiety is a modified sugar moiety. Gapmers as used in the present invention include wings that independently have from 1 to 7 nucleosides. The present invention therefore includes gapmers wherein each wing independently comprises 1, 2, 3, 4, 5, 6 or 7 nucleosides. The number of nucleosides in each wing can be the same or different. In one embodiment, the internal or gap region comprises from 17 to 21 nucleosides, which is understood to include 17, 18, 19, 20, or 21 nucleosides. As used in the present invention the term "positionally modified motif is meant to include a sequence of β-D-ribonucleosides, β-D-deoxyribonucleosides, or sugar-modified nucleosides wherein the sequence is interrupted by two or more regions comprising from 1 to about 8 sugar modified nucleosides wherein internal regions are generally from 1 to about 4 nucleosides. In other words, regions having a particular sugar moiety are separated by regions having different sugar moieties. Regions comprised of sugar modified nucleosides have the same sugar modification or vary such that one region has a different sugar modification than another region. Positionally modified motifs are not determined by the nucleobase sequence or the location or types of internucleoside linkages. The term positionally modified motif includes many different specific substitution patterns. A number of these substitution patterns have been prepared and tested in compositions. The present invention includes antisense compounds having a positionally modified motif characterized by regions of 2'-sugar modified nucleosides which are separated by regions of a second, distinct 2'-sugar modified nucleosides or bicyclic modified sugar nucleosides. Preferred 2'-sugar moieties include 2'-O-methyl and 2'-MOE. Preferred bicyclic sugar moieties include LNA and ENA. The regions of 2 '-sugar modified moieties may be 2 to 8 nucleosides in length, and the regions of bicyclic modified sugar moieties may be 1 or 2 nucleosides in length.
In one embodiment, antisense compounds of the invention are characterized by regions of two 2'-sugar modified nucleosides separated by regions of one bicyclic modified nucleoside, such that, beginning at the 5'-terminus, the antisense compounds have a 2'-sugar modified nucleoside at every first and second position, and a bicyclic modified nucleoside at every third position. Such a motif is described by the formula 5'-(A-A-B)n(-A)nn~3', wherein A is a first sugar moiety, B is a second sugar moiety, n is 6 to 7 and nn is 0 to 2. In preferred embodiments, A is 2'-MOE and B is LNA. In further embodiments, A is 2'-O-methyl and B is LNA. In some embodiments, when such a motif would yield a bicyclic nucleoside at the 3 '-terminus of the antisense compound (e.g, in an antisense compound 21 nucleosides in length), a 2'-sugar modified nucleoside is incorporated in place of a bicyclic modified nucleoside. For example, if n is 7, and nn is zero, a 2'-sugar modified nucleoside would be utilized at the 3 '-terminal position in place of a bicyclic modified nucleoside.
Positionally modified motifs having less regular patterns are also included in the present invention. For example, the majority of the 2 '-sugar modified regions may be 2 nucleosides in length, and a minority of 2'-sugar modified regions may be 1 nucleoside in length. Likewise, the majority of the bicyclic modified regions may be 1 nucleoside in length, and a minority of the bicyclic modified regions may be 2 nucleosides in length. One non-limiting example of such an antisense compound includes a positionally modified motif as described in the preceding paragraph, having one 2 '-sugar modified region one nucleoside in length, and one bicyclic modified region two nucleosides in length.
As used in the present invention the term "alternating motif is meant to include a contiguous sequence of two distinct alternating nucleosides for essentially the entire length of the compound. The pattern of alternation can be described by the formula: 5'-A(B-A)n(-B)nn-3' where A and B are nucleosides differentiated by having at least different sugar moieties, nn is 0 or 1 and n is from about 7 to about 11. This permits antisense compounds from 17 to 24 nucleosides in length. This length range is not meant to be limiting as longer and shorter antisense compounds are also amenable to the present invention. This formula also allows for even and odd lengths for alternating antisense compounds wherein the 5'- and 3 '-terminal nucleosides comprise the same (odd) or different (even) sugar moieties.
The "A" and "B" nucleosides comprising alternating antisense compounds of the present invention are differentiated from each other by having at least different sugar moieties. Each of the A and B nucleosides has a modified sugar moiety selected from β-D-ribonucleosides, β-D- deoxyribonucleosides, 2'-modified nucleosides (such 2 '-modified nucleosides may include 2'-MOE, and 2'-O-CH3, among others), and bicyclic sugar modified nucleosides (such bicyclic sugar modified nucleosides may include LNA or ENA, among others). The alternating motif is independent from the nucleobase sequence and the internucleoside linkages. The internucleoside linkage can vary at each position or at particular selected positions or can be uniform or alternating throughout the antisense compound. As used in the present invention the term "hemimer motif is meant to include a sequence of nucleosides that have uniform sugar moieties (identical sugars, modified or unmodified) and wherein one of the 5'-end or the 3'-end has a sequence of from 2 to 12 nucleosides that are sugar modified nucleosides that are different from the other nucleosides in the hemimer modified antisense compound. An example of a typical hemimer is an antisense compound comprising a sequence of β-D-ribonucleosides or β-D- deoxyribonucleosides at one terminus and a sequence of sugar modified nucleosides at the other terminus. One hemimer motif includes a sequence of β-D-ribonucleosides or β-D-deoxyribonucleosides at one terminus, followed or preceded by a sequence of 2 to 12 sugar modified nucleosides at the other terminus. Another hemimer motif includes a sequence of β-D-ribonucleosides or β-D-deoxyribonucleosides at one terminus, followed or preceded by a sequence of 2 to 6 sugar modified nucleosides located at the other terminus, with from 2 to 4 sugar modified nucleosides being suitable as well. In a preferred embodiment of the invention, the hemimer antisense compound comprises a region of 2'-MOE modified nuculeosides and a region of β-D-deoxyribonucleosides. In one embodiment, the β-D-deoxyribonucleosides comprise less than 13 contiguous nucleosides within the antisense compound.
As used in the present invention the term "blockmer motif is meant to include a sequence of nucleosides that have uniform sugars (identical sugars, modified or unmodified) that is internally interrupted by a block of sugar modified nucleosides that are uniformly modified and wherein the modification is different from the other nucleosides. More generally, antisense compounds having a blockmer motif comprise a sequence of β-D-ribonucleosides or β-D-deoxyribonucleosides having one internal block of from 2 to 6, or from 2 to 4 sugar modified nucleosides. The internal block region can be at any position within the antisense compound as long as it is not at one of the termini which would then make it a hemimer. The base sequence and internucleoside linkages can vary at any position within a blockmer motif.
Antisense compounds having motifs selected from uniform, positionally modified, alternating, gapped, hemimer or blockmer may further comprise internucleoside linkage modifications or nucleobase modifications, such as those described herein. "Chimeric antisense compounds" or "chimeras," in the context of this invention, are antisense compounds that at least 2 chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide or nucleoside in the case of a nucleic acid based antisense compound. Accordingly, antisense compounds having a motif selected from positionally modified, gapmer, alternating, hemimer, or blockmer are considered chimeric antisense compounds. Chimeric antisense compounds typically contain at least one region modified so as to confer increased resistance to nuclease degradation, increased cellular uptake, increased binding affinity for the target nucleic acid, and/or increased inhibitory activity. By way of example, an antisense compound may be designed to comprise a region that serves as a substrate for the cellular endonuclease KNase H, which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H by an antisense compound having a cleavage region, therefore, results in cleavage of the RNA target, thereby enhancing the efficiency of the antisense compound. Alternatively, an antisense compound may be designed to comprise a region that imparts to the antisense compound increased affinity or activity such that antisense inhibition is achieved through a mechanism such as steric occlusion, rather than enzymatic cleavage. Chemically modified nucleosides may also be employed to increase the binding affinity of a shortened or truncated antisense compound for its target nucleic acid. Consequently, comparable results can often be obtained with shorter antisense compounds that have such chemically modified nucleosides. Any of the antisense compounds described herein, having motifs selected from uniform, positionally modified, alternating, gapmer, hemimer, or blockmer, may further comprise intemucleoside linkage modifications and/or nucleobase modifications, such as those described herein.
Nucleosides, both native and modified, have a certain conformational geometry which affects their hybridization and affinity properties. The terms used to describe the conformational geometry of homoduplex nucleic acids are "A Form" for RNA and "B Form" for DNA. In general, RNA:RNA duplexes are more stable and have higher melting temperatures (Tm's) than DNA:DNA duplexes (Sanger et al., Principles of Nucleic Acid Structure, 1984, Springer- Verlag; New York, NY.; Lesnik et al., Biochemistry, 1995, 34, 10807-10815; Conte et al., Nucleic Acids Res., 1997, 25, 2627-2634). The increased stability of RNA has been attributed to several structural features, most notably the improved base stacking interactions that result from an A-form geometry (Searle et al., Nucleic Acids Res., 1993, 21, 2051-2056). The presence of the 2' hydroxyl in RNA biases the sugar toward a C3' endo pucker, i.e., also designated as Northern pucker, which causes the duplex to favor the A-form geometry. In addition, the 2' hydroxyl groups of RNA can form a network of water mediated hydrogen bonds that help stabilize the RNA duplex (EgIi et al., Biochemistry, 1996, 35, 8489-8494). On the other hand, deoxy nucleic acids prefer a C21 endo sugar pucker, i.e., also known as Southern pucker, which is thought to impart a less stable B-form geometry (Sanger, W. (1984) Principles of Nucleic Acid Structure, Springer- Verlag, New York, NY). As used herein, B-form geometry is inclusive of both C2'-endo pucker and O4'-endo pucker. This is consistent withBerger, et. al., Nucleic Acids Research, 1998, 26, 2473-2480, who pointed out that in considering the furanose conformations which give rise to B-form duplexes consideration should also be given to a O4'-endo pucker contribution.
DNA:RNA hybrid duplexes, however, are usually less stable than pure RNA:RNA duplexes, and depending on their sequence may be either more or less stable than DNA:DNA duplexes (Searle et al., Nucleic Acids Res., 1993, 21, 2051-2056). The structure of a hybrid duplex is intermediate between A- and B-form geometries, which may result in poor stacking interactions (Lane et al., Eur. J. Biochem., 1993, 215, 297-306; Fedoroff et al., J. MoI. Biol., 1993, 233, 509-523; Gonzalez et al., Biochemistry, 1995, 34, 4969-4982; Horton et al., J. MoI. Biol., 1996, 264, 521-533). The stability of the duplex formed between a target RNA and a synthetic sequence is central to therapies such as, but not limited to, antisense mechanisms, including RNase H-mediated and RNA interference mechanisms, as these mechanisms involved the hybridization of a synthetic sequence strand to an RNA target strand or steric hindrance.
One routinely used method of modifying the sugar puckering is the substitution of the sugar at the 2'-position with a substituent group that influences the sugar geometry. The influence on ring conformation is dependent on the nature of the substituent at the 2'-position. A number of different substituents have been studied to determine their sugar puckering effect.
In some embodiments of this invention, duplex stability can be enhanced by incorporating modified sugar moieties which exhibit A-form geometry. One of skill in the art can determine whether a given substituted sugar will have A-form geometry.
In one aspect of the present invention antisense compounds include nucleosides synthetically modified to induce a 3'-endo sugar conformation. A nucleoside can incorporate synthetic modifications of the heterocyclic base, the sugar moiety or both to induce a desired 3'-endo sugar conformation. These modified nucleosides are used to mimic RNA-like nucleosides so that particular properties of an antisense compound can be enhanced while maintaining the desirable 3'-endo conformational geometry. There is an apparent preference for an RNA type duplex (A form helix, predominantly 3'-endo) as a requirement (e.g. trigger) of RNA interference. Properties that are enhanced by using more stable 3'-endo nucleosides include, but are not limited to, modulation of one or more of the following: protein binding; protein off- rate; absorption and clearance; modulation of nuclease stability as well as chemical stability; modulation of the binding affinity and specificity of the antisense compound (affinity and specificity for enzymes as well as for complementary sequences); increased efficacy of RNA cleavage; and increase steric occulsion. The present invention provides antisense compounds designed to act as triggers of RNAi having one or more nucleosides modified in such a way as to favor a C3'-endo type conformation.
The conformation of modified nucleosides and the antisense compounds containing them can be estimated by various methods such as molecular dynamics calculations, nuclear magnetic resonance spectroscopy and CD measurements. Hence, modifications predicted to induce RNA-like conformations (A-form duplex geometry in an antisense compound context), are useful in the antisense compounds of the present invention. The synthesis of modified nucleosides amenable to the present invention are known in the art (see for example, Chemistry of Nucleosides and Nucleotides VoI 1-3, ed. Leroy B. Townsend, 1988, Plenum Press.)
In one aspect, the present invention is directed to antisense compounds that are designed to have enhanced properties compared to native RNA. One method to design optimized or enhanced antisense compounds involves each nucleoside of the selected sequence being scrutinized for possible enhancing modifications. One modification would be the replacement of one or more RNA nucleosides with nucleosides that have the same 3'-endo conformational geometry. Such modifications can enhance chemical and nuclease stability relative to native RNA while at the same time being much cheaper and easier to synthesize and/or incorporate into an oligonucleotide. The sequence can be further divided into regions and the nucleosides of each region evaluated for enhancing modifications that can be the result of a chimeric configuration. Consideration is also given to the 5' and 3'-termini as there are often advantageous modifications that can be made to one or more of the terminal nucleosides. The antisense compounds of the present invention may include at least one 5 '-modified phosphate group on a single strand or on at least one 5'-position of a double-stranded sequence or sequences. Other modifications considered are internucleoside linkages, conjugate groups, substitute sugars or bases, substitution of one or more nucleosides with nucleoside mimetics and any other modification that can enhance the desired property of the antisense compound.
Unless otherwise defined herein, alkyl means C1-C]2, C1-Cs, or Ci-Cβ, straight or (where possible) branched chain aliphatic hydrocarbyl.
Unless otherwise defined herein, heteroalkyl means C1-Cj2, C]-C8, or C]-C6, straight or (where possible) branched chain aliphatic hydrocarbyl containing at least one, or about 1 to about 3 hetero atoms in the chain, including the terminal portion of the chain. Suitable heteroatoms include N, O and S.
Unless otherwise defined herein, cycloalkyl means C3-C]2, C3-C8, or C3-C6, aliphatic hydrocarbyl ring.
Unless otherwise defined herein, alkenyl means C2-C12, C2-C8, or C2-C6 alkenyl, which may be straight or (where possible) branched hydrocarbyl moiety, which contains at least one carbon-carbon double bond.
Unless otherwise defined herein, alkynyl means C2-C]2, C2-C8, or C2-C6 alkynyl, which may be straight or (where possible) branched hydrocarbyl moiety, which contains at least one carbon-carbon triple bond. Unless otherwise defined herein, heterocycloalkyl means a ring moiety containing at least three ring members, at least one of which is carbon, and of which 1, 2 or three ring members are other than carbon. The number of carbon atoms can vary from 1 to about 12, from 1 to about 6, and the total number of ring members varies from three to about 15, or from about 3 to about 8. Suitable ring heteroatoms are N, O and S. Suitable heterocycloalkyl groups include, but are not limited to, morpholino, thiomorpholino, piperidinyl, piperazinyl, homopiperidinyl, homopiperazinyl, homomorpholino, homothiomorpholino, pyrrolodinyl, tetrahydrooxazolyl, tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydroisoxazolyl, tetrahydropyrrazolyl, furanyl, pyranyl, and tetrahydroisothiazolyl.
Unless otherwise defined herein, aryl means any hydrocarbon ring structure containing at least one aryl ring. Suitable aryl rings have about 6 to about 20 ring carbons. Especially suitable aryl rings include phenyl, napthyl, anthracenyl, and phenanthrenyl.
Unless otherwise defined herein, hetaryl means a ring moiety containing at least one fully unsaturated ring, the ring consisting of carbon and non-carbon atoms. The ring system can contain about 1 to about 4 rings. The number of carbon atoms can vary from 1 to about 12, from 1 to about 6, and the total number of ring members varies from three to about 15, or from about 3 to about 8. Suitable ring heteroatoms are N, O and S. Suitable hetaryl moieties include, but are not limited to, pyrazolyl, thiophenyl, pyridyl, imidazolyl, tetrazolyl, pyridyl, pyrimidinyl, purinyl, quinazolinyl, quinoxalinyl, benzimidazolyl, benzothiophenyl, etc.
Unless otherwise defined herein, where a moiety is defined as a compound moiety, such as hetarylalkyl (hetaryl and alkyl), aralkyl (aryl and alkyl), etc., each of the sub-moieties is as defined herein. Unless otherwise defined herein, an electron withdrawing group is a group, such as the cyano or isocyanato group that draws electronic charge away from the carbon to which it is attached. Other electron withdrawing groups of note include those whose electronegativities exceed that of carbon, for example halogen, nitro, or phenyl substituted in the ortho- or para-position with one or more cyano, isothiocyanato, nitro or halo groups.
Unless otherwise defined herein, the terms halogen and halo have their ordinary meanings. Suitable halo (halogen) substituents are F, Cl, Br, and I.
The aforementioned optional substituents are, unless otherwise herein defined, suitable substituents depending upon desired properties. Included are halogens (F, Cl, Br, I), alkyl, alkenyl, and alkynyl moieties, NO2, NH3 (substituted and unsubstituted), acid moieties (e.g. -CO2H, -OSO3H2, etc.), heterocycloalkyl moieties, hetaryl moieties, aryl moieties, etc. Phosphate protecting groups include those described in US Patents No. US 5,760,209, US
5,614,621, US 6,051,699, US 6,020,475, US 6,326,478, US 6,169,177, US 6,121,437, US 6,465,628 each of which is expressly incorporated herein by reference in its entirety.
Screening Antisense Compounds Screening methods for the identification of effective modulators of miR-122a target nucleic acids are also comprehended by the instant invention and comprise the steps of contacting a miR-122a target nucleic acid, or portion thereof, with one or more candidate modulators, and selecting for one or more candidate modulators which inhibit the levels, expression or function of a miR-122a target nucleic acid. As described herein, the candidate modulator can be an antisense compound targeted to a pri- miRNA, or any portion thereof, including the mature miRNA, the Drosha recognition region, the Drosha cleavage region, the stem of the hairpin, or the loop of the hairpin. Candidate modulators further include small molecule compounds that bind to structured regions of miR-122a target nucleic acids, such as structured regions within pri-miR-122a. Once it is shown that the candidate modulator or modulators are capable of modulating, preferably decreasing, the levels, expression or function of a miR-122a target nucleic acid, the modulator may then be employed in further investigative studies, or for use as a target validation, research, diagnostic, or therapeutic agent in accordance with the present invention.
Compositions and Methods for Formulating Pharmaceutical Compositions
The present invention also include pharmaceutical compositions and formulations that include the antisense compounds and compositions of the invention. Compositions and methods for the formulation of pharmaceutical compositions are dependent upon a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered. Such considerations are well understood by those skilled in the art.
The antisense compounds and compositions of the invention can be utilized in pharmaceutical compositions by adding an effective amount of the compound or composition to a suitable pharmaceutically acceptable diluent or carrier. Use of the antisense compounds and methods of the invention may also be useful prophylactically.
The antisense compounds and compositions of the invention encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal, including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to prodrugs and pharmaceutically acceptable salts of the antisense compounds of the invention, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
The term "prodrug" indicates a therapeutic agent that is prepared in an inactive form that is converted to an active form (i.e., drug) within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or conditions. This can include the incorporation of additional nucleosides at one or both ends of an antisense compound which are cleaved by endogenous nucleases within the body, to form the active antisense compound.
The term "pharmaceutically acceptable salts" refers to physiologically and pharmaceutically acceptable salts of the antisense compounds and compositions of the invention: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto. Suitable examples include, but are not limited to, sodium and postassium salts.
In some embodiments, an antisense compound can be administered to a subject via an oral route of administration. The subject may be a mammal, such as a mouse, a rat, a dog, a guinea pig, a non- human primate, or a human. In certain embodiments, the subject may be in need of modulation of the level or expression a miR-122a nucleic acid, or in need of modulation of surrogate indicators as desribed here. In some embodiments, compositions for administration to a subject will comprise modified oligonucleotides having one or more modifications, as described herein.
Cell culture and antisense compound treatment The effects of antisense compounds the level, activity or expression of miR-122a target nucleic acids, protein-coding RNAs regulated by miR-122a, can be tested in vitro in a variety of cell types. Cell types used for such analyses are available from commerical vendors (e.g. American Type Culture Collection, Manassus, VA; Zen-Bio, Inc., Research Triangle Park, NC; Clonetics Corporation, Walkersville, MD) and cells are cultured according to the vendor's instructions using commercially available reagents (e.g. Invitrogen Life Technologies, Carlsbad, CA). Illustrative cell types include, but are not limited to: T-24 cells, A549 cells, normal human mammary epithelial cells (HMECs), MCF7 cells, T47D cells, BJ cells, B16-F10 cells, human vascular endothelial cells (HUVECs), human neonatal dermal fibroblast (NHDF) cells, human embryonic keratinocytes (HEK), 293T cells , HepG2 , human preadipocytes, human differentiated adipocytes (preapidocytes differentiated according to methods known in the art), NT2 cells (also known as NTERA-2 cl.Dl), and HeLa cells. Treatment of cells with antisense compounds
In general, when cells reach approximately 60-80% confluency, they are treated with antisense compounds of the invention.
One reagent commonly used to introduce antisense compounds into cultured cells includes the cationic lipid transfection reagent LIPOFECTIN® (Invitrogen, Carlsbad, CA). Antisense compounds are mixed with LIPOFECTIN® in OPTI-MEM® 1 (Invitrogen, Carlsbad, CA) to achieve the desired final concentration of antisense compound and a LIPOFECTIN® concentration that typically ranges 2 to 12 μg/mL per 100 nM antisense compound.
Another reagent used to introduce antisense compounds into cultured cells includes LIPOFECTAMINE® (Invitrogen, Carlsbad, CA). Antisense compound is mixed with
LIPOFECTAMINE® in OPTI-MEM® 1 reduced serum medium (Invitrogen, Carlsbad, CA) to achieve the desired concentration of antisense compound and a LIPOFECTAMINE® concentration that typically ranges 2 to 12 μg/mL per 100 nM antisense compound.
Cells are treated with antisense compounds by routine methods well known to those skilled in the art. Cells are typically harvested 16-24 hours after antisense compound treatment, at which time RNA or protein levels of target nucleic acids are measured by methods known in the art and described herein. When the target nucleic acid is a miRNA, the RNA or protein level of a protein-coding RNA regulated by a miRNA may be measured to evaluate the effects of antisense compounds targeted to a miRNA. In general, when treatments are performed in multiple replicates, the data are presented as the average of the replicate treatments.
The concentration of antisense used varies from cell line to cell line. Methods to determine the optimal antisense concentration for a particular cell line are well known in the art. Antisense compounds are typically used at concentrations ranging from 1 nM to 300 nM.
RNA Isolation
RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA. Methods of RNA isolation are well known in the art. RNA is prepared using methods well known in the art, for example, using the TRIZOL® Reagent (Invitrogen, Carlsbad, CA) according to the manufacturer's recommended protocols.
Analysis of inhibition of target levels or expression
Inhibition of levels or expression of a miR- 122a nucleic acid, or modulation of a protein-coding RNA target of miR-122a (e.g. ALDO A), can be assayed in a variety of ways known in the art. For example, target nucleic acid levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or quantitaive real-time PCR. RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA. Methods of RNA isolation are well known in the art. Northern blot analysis is also routine in the art. Quantitative real-time PCR can be conveniently accomplished using the commercially available ABI PRISM® 7600, 7700, or 7900 Sequence Detection System, available from PE-Applied Biosystems, Foster City, CA and used according to manufacturer's instructions.
Additional examples of methods of gene expression analysis known in the art include DNA arrays or microarrays (Brazma and ViIo, FEBS Lett., 2000, 480, 17-24; Celis, et al, FEBS Lett., 2000, 480, 2-16), SAGE (serial analysis of gene expression)(Madden, et al., Drug Discov. Today, 2000, 5, 415- 425), READS (restriction enzyme amplification of digested cDNAs) (Prashar and Weissman, Methods Enzymol., 1999, 303, 258-72), TOGA (total gene expression analysis) (Sutcliffe, et al., Proc. Natl. Acad. Sci. U. S. A., 2000, 97, 1976-81), protein arrays and proteomics (Celis, et al., FEBS Lett., 2000, 480, 2- 16; Jungblut, et al., Electrophoresis, 1999, 20, 2100-10), expressed sequence tag (EST) sequencing (Celis, et al., FEBS Lett., 2000, 480, 2-16; Larsson, et al., J. Biotechnol., 2000, 80, 143-57), subtractive RNA fingerprinting (SuRF) (Fuchs, et al., Anal. Biochem., 2000, 286, 91-98; Larson, et al., Cytometry, 2000, 41, 203-208), subtractive cloning, differential display (DD) (Jurecic and Belmont, Curr. Opin. Microbiol., 2000, 3, 316-21), comparative genomic hybridization (Carulli, et al., J. Cell Biochem. Suppl., 1998, 31, 286-96), FISH (fluorescent in situ hybridization) techniques (Going and Gusterson, Eur. J. Cancer, 1999, 35, 1895-904), and mass spectrometry methods (To, Comb. Chem. High Throughput Screen, 2000, 3, 235-41).
Quantitative Real-Time PCR Analysis of Target RNA Levels Quantitation of target RNA levels is accomplished by quantitative real-time PCR using the ABI
PRISM® 7600, 7700, or 7900 Sequence Detection System (PE-Applied Biosystems, Foster City, CA) according to manufacturer's instructions. Methods of quantitative real-time PCR are well known in the art.
Prior to real-time PCR, the isolated RNA is subjected to a reverse transcriptase (RT) reaction, which produces complementary DNA (cDNA) that is then used as the substrate for the real-time PCR amplification. The RT and real-time PCR reactions are performed sequentially in the same sample well.
RT and real-time PCR reagents are obtained from Invitrogen (Carlsbad, CA). RT, real-time-PCR reactions are carried out by methods well known to those skilled in the art.
Gene (or RNA) target quantities obtained by real time PCR are normalized using either the expression level of a gene whose expression is constant, such as GAPDH, or by quantifying total RNA using RIBOGREEN® (Molecular Probes, Inc. Eugene, OR). GAPDH expression is quantified by real time PCR, by being run simultaneously with the target, multiplexing, or separately. Total RNA is quantified using RIBOGREEN® RNA quantification reagent (Molecular Probes, Inc. Eugene, OR).
Methods of RNA quantification by RIBOGREEN® are taught in Jones, LJ., et al, (Analytical Biochemistry, 1998, 265, 368-374). A CYTOFLUOR® 4000 instrument (PE Applied Biosystems) is used to measure RIBOGREEN® fluorescence. Probes and primers are designed to hybridize to the target sequence, which includes a protein- coding RNA that is regulated by a miR-122a, such as ALDO A.. Methods for designing real-time PCR probes and primers are well known in the art, and may include the use of software such as PRIMER EXPRESS® Software (Applied Biosystems, Foster City, CA).
Northern blot analysis of target RNA levels
Northern blot analysis is performed according to routine procedures known in the art. Higher percentage acrylamide gels, for example, 10 to 15% acrylamide urea gels, are generally used to resolve miRNA. Fifteen to twenty micrograms of total RNA is fractionated by electrophoresis through 10% acrylamide urea gels using a TBE buffer system (Invitrogen). RNA is transferred from the gel to
HYB0ND™-N+ nylon membranes (Amersham Pharmacia Biotech, Piscataway, NJ) by electroblotting in an Xcell SURELOCK™ Minicell (Invitrogen, Carlsbad, CA). Membranes are fixed by UV cross-linking using a STRATALINKER® UV Crosslinker 2400 (Stratagene, Inc, La Jolla, CA) and then probed using RAPID HYB™ buffer solution (Amersham) using manufacturer's recommendations for oligonucleotide probes.
A target specific DNA oligonucleotide probe with the sequence is used to detect the RNA of interest. Probes used to detect miRNAs are synthesized by commercial vendors such as IDT (Coralville, IA). The probe is 5' end-labeled with T4 polynucleotide kinase with (gamma-32P) ATP (Promega, Madison, WI). To normalize for variations in loading and transfer efficiency membranes are stripped and re-probed for an RNA whose level is constant, such as GAPDH. For higher percentage acrylamide gels used to resolve miRNA, U6 RNA is used to normalize for variations in loading and transfer efficiency . U6 RNA. Hybridized membranes are visualized and quantitated using a STORM® 860 PHOSPHORIMAGER® System and IMAGEQUANT® Software V3.3 (Molecular Dynamics, Sunnyvale, CA).
Analysis of Protein Levels
Protein levels of a downstream target modulated or regulated by miR-122a, such as ALDO A, can be evaluated or quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), enzyme-linked immunosorbent assay (ELISA), quantitative protein assays, protein activity assays (for example, caspase activity assays), immunohistochemistry, immunocytochemistry or fluorescence-activated cell sorting (FACS). Antibodies directed to a target can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, MI), or can be prepared via conventional monoclonal or polyclonal antibody generation methods well known in the art.
In vivo studies In accordance with the present invention, antisense compounds of the invention are tested in various animal models of disease, including normal, lean mice, ob/ob mice, db/db mice, and diet-induced obese mice, and mouse and rat models of diet induced obesity.
Leptin is a hormone produced by fat cells that regulates appetite. Deficiencies in this hormone in both humans and non-human animals leads to obesity, ob/ob mice have a mutation in the leptin gene which results in obesity and hyperglycemia. As such, these mice are a useful model for the investigation of obesity and diabetes and treatments designed to treat these conditions, ob/ob mice have higher circulating levels of insulin and are less hyperglycemic than db/db mice, which harbor a mutation in the leptin receptor, db/db mice have a mutation in the leptin receptor gene which results in obesity and hyperglycemia. As such, these mice are a useful model for the investigation of obesity and diabetes and treatments designed to treat these conditions, db/db mice, which have lower circulating levels of insulin and are more hyperglycemic than ob/ob mice which harbor a mutation in the leptin gene, are often used as a rodent model of type 2 diabetes.
C57B1/6 mice are maintained on a standard rodent diet for use as control (lean) animals. Additionally, the C57BL/6 mouse strain is reported to be susceptible to hyperlipidemia-induced atherosclerotic plaque formation. Consequently, when these mice are fed a high-fat diet, they develop diet-induced obesity. Accordingly these mice are a useful model for the investigation of obesity and treatments designed to treat these conditions.
The Levin Model is a polygenic model of rats selectively bred to develop diet-induced obesity (DIO) associated with impaired glucose tolerance, dyslipidemra and insulin resistance when fed a high-fat diet (Levin, et al., Am. J. Physiol, 1997, 273, R725-30). This model is useful in investigating the antisense compounds of the present invention for their ability to affect obesity and related complications, such as impaired glucose tolerance, dyslipidemia and insulin resistance.
In order that the invention disclosed herein may be more efficiently understood, examples are provided below. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting the invention in any manner. Throughout these examples, molecular cloning reactions, and other standard recombinant DNA techniques, were carried out according to methods described in Maniatis et al., Molecular Cloning - A Laboratory Manual, 2nd ed., Cold Spring Harbor Press (1989), using commercially available reagents, except where otherwise noted.
Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference (including, but not limited to, journal articles, U.S. and non-U.S. patents, patent application publications, international patent application publications, GENBANK® accession numbers, and the like) cited in the present application is incorporated herein by reference in its entirety. EXAMPLES Example 1: pri-miRNA sequences
The sequence of pri-miR-122a was used in certain embodiments of the invention, for example, in the design of antisense compounds targeting a miR-122a nucleic acid. The sequence of the human pri- miR-122a was extracted from the sequence having GENBANK® accession number NT_033907.3 is incorpated herein as SEQ ID NO: 5
(UGGCUACAGAGUUUCCUUAGCAGAGCUGUGGAGUGUGACAAUGGUGUUUGUGUCUAAAC
UAUCAAACGCCAUUAUCACACUAAAUAGCUACUGCUAGGCAAUCCUUCCCU). The sequence of the mouse pri-miR-122a is incorporated herein as SEQ DD NO: 6
(UUCCUUAGCAGAGCUGUGGAGUGUGACAAUGGUGUUUGUGUCCAAACCAUCAAACGCCA
UUAUCACACUAAAUAGCUACUG). The sequence of the rat pri-miR-122a is incorporated herein as
SEQ ID NO: 7
(UCCUUAGCAGAGCUCUGGAGUGUGACAAUGGUGUUUGUGUCCAAAACAUCAAACGCCAU CAUCACACUAAACAGCUACUG). The sequences are written in the 5' to 3' direction and are represented in the DNA form. It is understood that a person having ordinary skill in the art would be able to convert the sequence of the targets to their RNA form by simply replacing the thymidine (T) with uracil (U) in the sequence.
Example 2: mature miRNA sequences
Mature miRNAs found within the pri-miR-122a sequence were used in certain embodiments of the present invention, for example, in the design of antisense compounds targeting a miR-122a nucleic acid. Antisense compounds can also be designed to mimic miR-122a while incorporating certain chemical modifications that alter one or more properties of the mimic, thereby creating a construct with superior properties over the endogenous miRNA.
Mature miR-122a sequences are shown in Table 1. Mature miR-122a sequences from pri-miR- 122a precursors have been proposed by several groups; consequently, two mature miR-122a sequences are disclosed in Table 1. The sequences are written in the 5' to 3' direction and are represented in the RNA form. It is understood that a person having ordinary skill in the art would be able to convert the sequence of the targets to a DNA form by simply replacing the uracil (U) with thymidine (T) in the sequence.
Table 1
Mature miR-122a found within pri-miR-122a
Figure imgf000040_0001
Figure imgf000041_0001
Example 3: Antisense compounds targeting miR-122a
Ia accordance with the present invention, a series of antisense compounds was designed and synthesized to target various sites within miR-122a nucleic acids. The antisense compounds are analyzed for their effect on miRNA, pre-miRNA or pri-miRNA levels by Northern blotting or quantitative realtime PCR as described, supra, or they can be used in other assays to investigate the role of miR-122a or miR-122a downstream targets.
The target sites to which the following antisense compounds are complementary include mature miR-122a, as well as target sites of pri-miR-122a. For example, an antisense compound having the sequence of SEQ ID NO: 12 was designed to target the stem loop of the pri-miR-122a structure.
The nucleobase sequences and target sites of the antisense compounds of the invention are shown in Table 2. In Table 2, the target site is the sequence of SEQ ID NO: 5 to which the antisense compound is complementary. While the nucleobase sequences in Table 2 are shown having the DNA nucleobases "T", one of skill in the art understand that in RNA sequences, "T" is replaced with "U". For example, in the nucleobase sequence of the uniform RNA antisense compound ISIS 342970, "T" is replaced with "U".
Table 2: Sequences of antisense compounds targeting pri-miR-122a or miR-122a
Figure imgf000041_0002
Table 4 lists antisense compounds having one of the sequences shown in Table 2 and one of the motifs desribed herein. In Table 4, "Sugar" indicates the sugar type(s) found in the antisense compound; "Backbone" indicates the type of internucleoside linkages; "Motif indicates the particular motif of the antisense compound. Motifs are described according to the number of nucleosides in each distinct region. For example, the positionally modified motif "5-1-5-1-4-1-6", having 2'-MOE and LNA sugars has a region of 5 2'-MOE modified nucleosides, followed by a region of one LNA modified nucleoside, and so forth. The type of sugar, internucleoside linkage or base modification is noted using the tRNA modification-like abbreviation system. Sequence, internucleoside linkage, nucleobase, and sugar types are all shown and indicated as follows:
• Bases in the sequence are indicated with capital letters
• Base modifications are shown before the base residue with a superscripted small letter code (a number can indicate position), and are optional
• Sugar type is shown after the base residue with a subscripted small letter code
• Internucleoside linkage type is shown after the sugar (3 '-side) with a subscripted small letter code
Sugar, nucleobase, and internucleoside abbreviations are shown in Table 3.
Table 3
Figure imgf000042_0001
Table 4: Antisense Compounds Targeting a miR-122a nucleic acid
Figure imgf000042_0002
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Table 5: Truncated antisense compounds targeting miR-122a: uniformly modified with 2'-MOE and phosphorothioate linkages
Figure imgf000045_0002
Table 6: Mismatched antisense compounds targeting miR-122a: uniformly modified with 2'-MOE and phosphorothioate linkages
Figure imgf000045_0003
ISIS 343160 is an oligomeric compound representing a chemically modified miR-122a mimic. It is also understood that the human miR-122a mimic compound may also mimic other rniRNAs from mammals, such as those from rodent species, for example. It is also understood that these mimics can serve as the basis for several variations of nucleic acid oligomeric compounds, including compounds with chemical modifications such as uniform or chimeric 2'-MOE oligomeric compounds, as well as LNAs and PNAs; such oligomeric compounds are also within the scope of the invention.
Example 4: Effects of antisense compounds targeting miR-122a in phenotypic assays
The effects of antisense oligonucleotides ISIS 328372, ISIS 328373, and ISIS 327895 on miR- 122a were tested in several phenotypic assays, including an apoptosis assay, an adipocyte differentiation assay, and an insulin signaling assay. The experimental details of these assays, and the results obtained, are described in detail in U.S. Application Publication No. 2005/0261218, which is hereby incorporated by reference in its entirety. As described therein, ISIS 328372 and ISIS 328373 did not induce apoptosis in the apoptosis assay. ISIS 327895 reduced levels of markers of adipocyte differentiation, and decreased mRNA expression of follistatin and PEPCKc in the insulin signaling assay.
Example 5: Modulation of miR-122a activity in vitro with antisense compounds
To monitor inhibition of miR-122a with antisense oligonucleotides, regulation of several computationally predicted miR-122a targets was assayed in vitro.
Mouse primary hepatocytes were isolated from Balb/c mice using methods described in the art (Neufeld, 1997, Methods MoI. Biol., 75:145-151.). Cells were seeded into 96-well plates in culture medium (Williams' Medium E supplemented with 10% fetal bovine serum, 10 nM HEPES, and penicillin/streptomycin) and cultured overnight before transfection. Transfection of oligonucleotide in LIPOFECTIN® (Invitrogen, Carlsbad, CA) was performed in triplicate, according to the manufacturer's instructions. Cells were treated with either ISIS 327895 or the control oligonucleotide ISIS 342683 at concentrations of 15 nM, 44 nM, or 133 nM. Cells were lysed 24 hours after transfection and total RNA was harvested using using QIAGEN® RNEASY® 96 columns on a BIOROBOT® 3000 (Qiagen, Valencia, CA). The mRNA levels of miR-122a target genes predicted by the TargetScan algorithm (Lewis et al,
2005, Cell, 120: 15-20; Lewis et al., 2003, Cell, 115:787-798) were measured using primer probe sets designed to hybridize to each particular predicted target gene and routine TAQMAN® real-time PCR methods. The predicted target mRNAs examined are shown in Table 7. Also shown in Table 7 are GENBANK® accession numbers associated with the mouse and human target mRNAs. Table 7. miR-122a target genes predicted by the TargetScan algorithm
Figure imgf000046_0001
Figure imgf000047_0001
Five of the targets (GYSl, SLC7A1, ALDOA, CCNGl, P4HA1) were upregulated 1.5 to 3 fold after treatment with ISIS 327895, while none were affected by treatment with the control oligonucleotide. Only the predicted MINKl (referred to as mMINK in Figures) target was unaffected after treatment with ISIS 327895.
In a reciprocal experiment, treatment of the mouse liver carcinoma AMLl 2 cell line with a miR-122a mimetic caused inhibition of GYSl, SLC7A1/CAT-1, and ALDOA, as assayed by real-time PCR. The miR-122a duplex RNA that was used as a miR-122a mimetic was purchased from Dharmacon (Lafayette, CO). The miR-122a duplex contained: UGGAGUGUGACAAUGGUGUUUGU (SEQ ID NO: 3) and its complement AAACACCAUUGUCACACUCCAUA (SEQ ID NO: 25). The 5' mismatch miR-122a duplex comprised: UGCACAGAGACAAUGGUGUUUGU (SEQ ID NO: 26) and its complement AAACACCAUUGUCUCUGUGCAUA (SEQ ID NO: 27). The 3' mismatch miR- 122a duplex comprise UGGAGUGUGACAUUGCAGUAGUG (SEQ ID NO: 28) and its complement AUACUGCAAUGUCACACUCCAUA (SEQ ID NO: 29). AML12 cells do not have high expression levels of miR-122a as are observed in primary hepatocytes (see Figure 6). Transfection of the miR-122a mimic led to a decrease in mRNA levels for the same 5 targets (GYSl, SLC7A1, ALDOA, CCNGl, P4HA1) that were upregulated following antisense inhibition of miR-122a. Interestingly, the miR-122a mimetic did not modulate expression of MINK or NPEPPS, showing that in vitro antisense and miR mimetic effects on miR- 122a target genes correlate.
Example 6: Modulation of miR-122a activity in vitro with miRNA antisense
To monitor inhibition of miR- 122a with antisense oligonucleotides bearing varied modifications, regulation of the predicted miR-122a target GYSl was assayed in mouse primary hepatocytes, using real-time PCR methods as described herein. Cells were treated with ISIS 327895, ISIS 365219, ISIS 386653, ISIS 386654, ISIS 345363, or the control oligonucleotide ISIS 342683 at doses of 15 nM, 44 nM, 133 nM, or 400 nM. The sequence of ISIS 342683 is CCTTCCCTGAAGGTTCCTCCTT (SEQ ID NO: 30); this oligomeric compound is uniformly composed of 2'-MOE nucleosides, has a phosphorothioate backbone, and all cytosines are 5-methylcytosines. ISIS 327895 and ISIS 365219 were effective in inhibiting miR-122a, as evidenced by the increased expression of GYSl. ISIS 386653 and ISIS 386654 increased GYSl mRNA levels to a lesser extent. The gapmer ISIS 345363 did not markedly increase GYSl mRNA levels.
Similar experiments suggested that of six predicted miR-122a target genes tested (GYSl, SLC7A1, ALDO A, CCNGl, MINK, P4HA1), mMINK is not modulated by antisense oligonucleotides targeting miR-122a. However, mCAT-1 does appear to be modulated by targeting miR-122a. To examine the effects of oligonucleotides of varying lengths on miR-122a inhibition, regulation of the predicted miR-122a target GYSl was assayed in mouse primary hepatocytes, using methods as described herein. Cells were treated with ISIS 386655, ISIS 386656, ISIS 386657, ISIS 386658, ISIS 386659, or ISIS 386660 at doses of 15 nM, 44 nM, 133 nM, or 400 nM. While truncations resulted in slightly less stimulation of GYSl mRNA levels, some truncated antisense compound were able to stimulate GYSl mRNA expression. Antisense compounds in which 2, 4, or 6 nucleosides were truncated from the 5' end, in this assay, were able to stimulate GYSl mRNA expression, relative to control samples. An antisense compound in which 2 nucleosides were truncated from the 3' end was able to stimulate GYSl mRNA expression, relative to control samples. However, truncations of 4 or 6 nucleosides from the 3' end significantly reduced the ability of the antisense compounds to stimulate GYSl mRNA expression.
To examine the effects of mismatches on the ability of oligonucleotides targeted to miR-122a to inhibit its effects on the predicted target GYSl, mouse primary hepatocytes were treated with oligonucleotides bearing mismatches to miR-122a (ISIS 386661, ISIS 386662, ISIS 386663, ISIS 386664, ISIS 386665, and ISIS 386666). Increasing the number of mismatches appears to decrease the ability of the antisense oligonucleotides to modulate GYSl mRNA levels. One mismatch was tolerated; however, 2 mismatches resulted in only a slight increase in GYSl mRNA levels, whereas 3 to 6 mismatches completely ablated miR-122a antisense oligonucleotide activity.
Embodiments of the present invention include methods of validating predicted miR-122a targets. Also provided are methods of modulating miR-122a targets with an antisense compound which targets miR-122a. In one embodiment, the antisense compound has the nucleobase sequence of SEQ ID NO: 2. In another embodiment, the antisense compound is uniformly comprised of 2'-MOE sugar modifications. In another embodiment, the antisense compound is ISIS 327895. Further provided are methods of assaying for inhibition of miR-122a comprising monitoring expression of a miR-122a target gene.
Example 7: Modulation of miR-122a activity in vivo with miRNA antisense—Normal mice W 2
Male C57BL/6 mice were obtained from The Jackson Laboratory. The mice were separated into treatment groups and were treated intraperitoneally with doses of ISIS 327895 ranging from 12.5 to 75 mg/kg or with 75 mg/kg of the control oligonucleotide ISIS 342683 twice weekly for four weeks. Mice treated with saline alone served as further controls. The mice appeared healthy and normal at the end of treatment with plasma AST and ALT levels in the normal range and no loss of body weight or reduced food intake. Histological analysis of liver sections revealed no apparent change in morphology. (Histological analysis of liver was carried out via routine procedures known in the art. Briefly, liver was fixed in 10% buffered formalin and embedded in paraffin wax. 4-mm sections were cut and mounted on glass slides. After dehydration, the sections were stained with hematoxylin and eosin.) The levels of the five miR-122a target mRNAs identified in vitro were evaluated in liver tissue using Taqman real-time PCR. Four of the five mRNAs (GYSl, ALDOA, P4HA1, and CCNGl) were increased in the. mice treated with ISIS 327895, while no target mRNA changes were observed in mice treated with the control oligonucleotide, demonstrating specific inhibition of miR-122a in the liver. P4HA1 displayed maximal upregulation at the lowest dose of ISIS 327895 tested. An embodiment of the present invention is a method of modulating miR-122a targets in an animal comprising administering an antisense oligonucleotide targeting miR-122a. In one embodiment, the targets are modulated in the liver. In one embodiment, said antisense oligonucleotide is ISIS 327895.
Northern blotting for miR-122a in liver RNA from the treated mice revelaed a three-fold reduction in miR-122a levels after treatment with the lowest dose (12.5 mg/kg) of ISIS 327895 and a redcution of greater than 10-fold at the highest (75 mg/kg) dose, but the control oligonucleotide had no effect on miR-122a levels in the liver. An embodiment of the present invention is a method of reducing miR-122a levels in the liver of an animal comprising administering an antisense oligonucleotide targeting miR-122a. In one embodiment, said antisense oligonucleotide is ISIS 327895.
Plasma levels of total cholesterol, triglycerides and glucose were also monitored using methods known in the art (for example, via Olympus AU400e automated clinical chemistry analyzer, Melville,
NY). After four weeks of treatment, inhibition of miR-122a with ISIS 327895 did not significantly affect plasma glucose levels, however, significant reductions in total cholesterol and triglycerides were observed at all doses tested as compared to saline or control-oligonucleotide treated mice.
Embodiments of the present invention include methods of reducing serum cholesterol, glucose, or triglycerides in an animal comprising administering an antisense compound targete to miR-122a. In one embodiment, the antisense compound has the nucleobase sequence of SEQ ID NO: 2. In another embodiment, the antisense compound is uniformly comprised of 2'-MOE sugar modifications. In another embodiment, the antisense compound is ISIS 327895.
Example 8: Effects of miR-122a antisense oligonucleotide treatment in a diet-induced model of obesity: 25 mg/week study Male, C57W/6 mice, 6-7 weeks old, were fed a high fat diet (60% kcal from fat) for 19 weeks prior to beginning treatment with antisense oligonucleotides. Mice were fed ad libitum. Starting insulin levels were approximately 15ng/dL and body composition was approximately 20% fat. Mice were dosed subcutaneously at 12.5 mg/kg twice weekly (i.e. 25 mg/wk), for a total of 11 total doses. Each treatment group consisted of 5 mice.
Treatment groups were as follows: saline; ISIS 327895, antisense to miR-122a; ISIS 342683, a scrambled control oligonucleotide. The sequence of ISIS 342683 is CCTTCCCTGAAGGTTCCTCCTT (SEQ ID NO: 30); this oligomeric compound is uniformly composed of 2'-MOE nucleosides, has a phosphorotliioate backbone, and all cytosines are 5-methylcytosines. Body composition was determined by MRI at the start of the study and following 3.5 and 5 weeks of treatment. Serum chemistries were analyzed following 2, 3, 4, 5, and 6 weeks of treatment. Insulin was measured via routine ELISA in the fed state following 3 weeks of treatment, and in the fasted state following 5 weeks of treatment. An oral glucose tolerance test was performed following 5 weeks of treatment (via routine procedures: glucose was administered, blood samples were collected and analyzed on a clinical analyzer).
Serum lipoprotein and cholesterol profiling was performed as described (Kieft et al., 1991) using a Beckman System Gold 126 HPLC system, a 507e refiϊdgerated autosampler, a 126 photodiode array detector (Beckman Instruments) and a Superose 6 HR 10/30 column (Pfizer). HDL, LDL, and VLDL fractions were measured at a wavelength of 505 nm and valiated with a cholesterol calibration kit (Sigma, St. Louis, MO). For each experiment, a three-point standard curve was performed in triplicate to determine the absolute concentration of each lipoprotein fraction. Lipoprotein analysis performed in this manner showed that the decrease in total cholesterol reflected a reduction in both low-density lipoprotein and high-density lipoprotein fractions.
Glycogen synthase 1 is a miR-122a target and its expression is modulated by antisense inhibition of miR-122a. As glycogen synthase 1 expression was elevated in livers of mice treated with ISIS 327895, glycogen content in livers was evaluated. A significant increase in glycogen levels in the livers of the ISIS 327895-treated mice was observed, suggesting that miR-122a may also regulate glycogen storage through its repression of the muscle glycogen synthase (GYSl) mRNA.
Mean insulin levels are shown in Table 8. Serum chemistry analysis is shown in Table 9.
Table 8: Mean insulin levels in 25 mg/wk DIO Study
Figure imgf000050_0001
Table 9: Mean serum chemistry levels in 25 mg/wk DIO Study.
GLUC = glucose; ALT = alanine aminotransferase; AST = aspartate aminotransferase; chol = total cholesterol; trigs = triglycerides; HDL = HDL-cholesterol; LDL = LDL-cholesterol; NEFA = non-esterifϊed fatty acids
Figure imgf000051_0001
miR-122a target genes whose expression is upregulated following antisense inhibition of miR- 122a in vivo included GYS 1 , ALDO A, CCNGl , and P4HA1. mRNA levels were measured by real-time PCR using procedures described herein and commonly known in the art. Upregulation of these genes in mice treated with ISIS 327895 confirms inhibition of miR-122a activity.
Histological analysis of the liver showed substantial reduction of liver steatosis with ISIS 327895 treatment. Liver triglyceride levels were also reduced with antisense inhibition of miR-122a. Liver weights were also decreased. A trend toward reduction in plasma transaminase levels in ISIS
327895-treated mice indicates an improvement in hepatic function. These changes also correlated with a reduction in mRNA levels of key enzymes FASN, ACC2, and SCDl. A slight lowering of food consumption was observed in the mice treated with ISIS 327895. Cholesterol was lowered by approximately 35% following 2 weeks of treatment and by about 40% at the end of the study. Following 4 weeks of treatment, HDL was decreased by 50% and LDL was decreased by 28%. Consistent with the observed cholesterol reduction, levels of ApoB-100 protein in the plasma of ISIS 327895-treated mice were reduced compared to saline-treated mice, as measured by Western blotting. No significant changes in glucose, serum transaminases, body composition or food consumption were observed. Slight changes in FFA and serum triglycerides were observed. Changes were observed in insulin levels as shown in Table 8. Treatment with ISIS 327895 slightly increased liver glycogen levels. Embodiments of the present invention include methods of improving insulin sensitivity, methods of reducing cholesterol, methods of reducing LDL cholesterol, methods of reducing FFA, methods of reducing ApoB-100 levels, and methods of reducing triglycerides in the serum of an animal by administering an antisense compound targeted to miR-122a. In a further embodiment, improvement in insulin sensitivity is measured as a reduction in circulating insulin levels. In one embodiment, the antisense compound has the nucleobase sequence of SEQ ID NO: 2. In another embodiment, the antisense compound is uniformly comprised of 2'-MOE sugar modifications. In a further embodiment, the antiense compound is ISIS 327895.
Example 9: Gene Expression Changes Following antisense inhibition of miR-122a
In one embodiment, changes in liver gene expression following antisense inhibition of miR- 122a were evaluated by microarray analysis. Liver KNA was isolated from normal mice treated with 50 mg/kg of ISIS 327895 twice weekly and from animals treated with saline alone, and microarray analysis was performed. GE CODELINK™ Mouse Whole Genome Bioarrays were used according to the manufacturer's protocols. Analysis of modulated genes by Gene Ontology category revealed that many genes involved in regulation of lipid and carbohydrate metabolism were affected in the ISIS 327895 treated mice. Key eixzymes in the fatty acid synthesis pathway, cholesterol biosynthesis pathway, acetyl CoA transport pathway, and glycolysis pathway were inhibited. Phosphomevalonate kinase (PMVK) mRNA downregulation was statistically significant. Several other genes related to cholesterol metabolism were also reduced, including HMGCR and PMVK. Several key genes known to regulate fatty acid synthesis and oxidation, including ACCl, ACC2, ACLY, SCDl and FASN, were significantly downregulated in the microarray experiment, and the downregulation of these 5 genes was confirmed by real-time PCR.
Many of the upregulated genes show enriched expression in brain, fertilized eggs, and placenta. Genes indicative of hepatocyte de-differentiation, for example, HNF-lalpha, HNF-I beta, HNF-4, albumin, cytokeratin 19, and alpha-fetoprotein, did not exhibit altered expression following antisense inhibition of miR-122a. The microarray data is deposited in a publicly accessible database Gene
Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/geoΛ with series accession GSE3603 and are herein incorporated by reference.
Although the microarray experiment was designed primarily to uncover the downstream effects of miR-122a inhibition after chronic treatment, rather than identify direct miR-122a target genes, 108 significantly upregulated genes with a strict 7 or 8 nucleoside seed match in their 3' UTR were identified. These may be direct miR-122a target genes. Most of these mRNAs were modestly increased by only 1.5- 3-fold. These genes are identified in the following table (Table 10) by their GENBANK® accession numbers and the associated gene symbol. Also indicated is whether the target includes a 7 or 8 nucleoside seed. Table 10: Target gene expression profile following antisense inhibition of miR-122a in vivo
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Therefore, another embodiment of the present invention is a method of identifying miR-122a targets. Another embodiment of the present invention is a method of modulating the expression of any of the 108 targets identified in the preceding table comprising modulating the miR-122a. In a particular embodiment, miR-122a is modulated by an antisense compound. In one embodiment, the antisense compound has the nucleobase sequence of SEQ ID NO: 2. In another embodiment, the antisense compound is uniformly comprised of 2'-MOE sugar modifications. In a further embodiment, the antiense compound is ISIS 327895.
Example 10: Modulation of lipid metabolism following antisense inhibition of miR-122a
Fatty acid oxidation is an ATP-producing catabolic pathway. The central metabolic sensor AMPK is a key regulatory enzyme that acts to promote ATP-generating pathways such as fatty acid oxidation and inhibitis energy storage pathways such as fatty acid synthesis.
Fatty acid oxidation was measured in hepatocytes isolated from livers of ISIS 327895-treated mice. Primary hepatocytes were isolated from lean C57B1/6 mice treated with saline, ISIS 327895, or ISIS 342683, at 25 mg/kg, twice per week, for a total of 5 doses. Hepatocytes were plated in 25 cm2 culture flasks, at a density of 1 X 106 cells per flask. Prior to plating of the cells, the flasks were treated with 2 ml of 0.1 mg/ml collagen in phosphate-buffered saline for 10 minutes at room temperature. Each liver yielded 4 flasks, 3 of which were used for fatty acid oxidation experiments and one of which was reserved for protein measurements. To each flask of hepatocytes was added 3 ml of William E culture medium, supplemented with 1.0 nM insulin and 10% fetal bovine serum (medium and supplements available from, for example, Invitrogen Life Technologies, Carlsbad, CA). Following overnight culture, the cells were washed twice with phosphate-buffered saline and were then cultured in 2.0 ml of low- glucose DMEM containing 0.25% bovine serum albumin and 0.25 uCi 14C-oleaic acid. The flask was filled with 5% CO2/95%02 and capped with a septa rubber stop, to which was attached a 0.5 ml tube with a filter paper. Three hours lated, 0.4 ml of 70% HCIO4 was injected into the culture medium and 0.05 ml of 25% NaOH was immediated injected onto the filter paper to collect the CO2. After 3-4 hours of CO2 collection, the filter paper was removed and placed in a scintillation vial. The 0.5 ml tube that contained the filter paper was washed twice with water and the water was transferred into the scintillation vial. 1.2 ml of the culture medium was collected from each flask and centrifuged, and the supernatant was transferred to an additional scintillation tube. The radioactivity in the CO2 collection and the culture medium supernatant was determined and used to calculate the fatty acid oxidation rate. Fatty acid oxidation rates were normalized by protein levels in each flask.
The results are shown in Figure 5. Treatment with ISIS 327895 significantly increased fatty acid oxidation, relative to both saline and control oligonucleotide-treated samples. AMPK levels were also examined by western blotting of whole liver extracts of mice treated with ISIS 327895 and compared to saline or control oligomeric compound-treated mice. Total AMPKalphal protein level was unchanged in the ISIS 327895-treated mice, but the levels of phosphorylated AMPKalphal were increased more than 2.5 times relative to saline-treated mice. In accordance with the observed reduction in FASN, ACCl and ACC2 mRNA after miR-122a inhibition, the fatty acid synthesis rate was reduced almost 2-fold in hepatocytes from mice treated with ISIS 327895 relative to hepatocytes from saline-treated mice. The rate of sterol synthesis was also significantly reduced. Sterol synthesis and fatty acid synthesis rates were determined by measuring the incorporation of [14C] acetate into sterols and fatty acids, respectively, using methods described in the art (Jiang, et al, 2005 J. Clin. Invest. 115:1030-1038).
Embodiments of the present invention include methods of lowering hepatic fatty acid synthesis and sterol synthesis and methods of increasing hepatic fatty acid oxidation in an animal comprising administering to said animal an antisense compound targeted to miR-122a. In one embodiment, the antisense compound has the nucleobase sequence of SEQ ID NO: 2. In another embodiment, the antisense compound is uniformly comprised of 2'-MOE sugar modifications. In a further embodiment, the antiense compound is ISIS 327895.
Example 11: Effects of miR-122a antisense oligonucleotide treatment in ob/ob mice
Eight week old ob/ob mice were given an intraperitoneal injection of ISIS 327895 or a control oligonucleotide dissolved in saline at a dose of 25 mg/kg twice a week for 4.5 weeks. Blood was collected and analyzed using routine clinical analyzer instruments (for example, an Olympus AU400e automated clinical chemistry analyzer, Melville, NY). Treatment with ISIS 327895 caused significant decrease in plasma cholesterol levels (about 168 mg/dL) as compared to treatment with control oligonucleotide (about 237 mg/dL) or saline-treatment alone (about 241 mg/dL).
Antisense inhibition of miR-122a also reduced hepatic triglyceride levels (about 89 mg/g) as compared to treatment with control oligonucleotide (about 161 mg/g) or saline-treatment alone (about 151 mg/g). Liver triglyceride levels are measured using routine methods described in the art (Desai, et al. 2001, Diabetes, 50: 2287-2295. The reduction in steatosis was reflected as a decrease in plasma ALT and AST levels in the group treated with ISIS 327895. Thus, other embodiments of the present invention include methods of decreasing hepatic triglycerides in an animal, methods of decreasing serum cholesterol in an animal, and methods of improving hepatic function in an animal comprising administering an antisense oligonucleotide which inhibits miR-122a function. The present invention also contemplates methods of ameliorating or preventing hyperlipidemia or hepatic steatosis in an animal comprising administering an antisense compound targeted to miR-122a. In one embodiment, the antisense compound has the nucleobase sequence of SEQ ID NO: 2. In another embodiment, the antisense compound is uniformly comprised of 2'-MOE sugar modifications. In a further embodiment, the antiense compound is ISIS 327895.
No change in plasma glucose or glucose tolerance was observed, and no overt toxicities, change in food intake or body weight were observed across treatment groups. These results demonstrate that miR-122a plays a role in regulating cholesterol and lipid metabolism. The effects are independent of leptin action, and there are no overt side effects caused by reduction of miR-122a.
Another embodiment of the present invention is a method of regulating cholesterol and lipid metabolism by inhibiting miR-122a.
Example 12: Antisense inhibition of miR-122a activity in vivo using enhanced antisense compounds The antisense compounds of the invention modulate the activity or function of the small non- coding RNAs to which they are targeted. In this example, antisense compounds targeted to miR-122a are illustrated; however, the modifications in the antisense compounds of the invention are not limited to those antisense compounds that modulate miR-122a.
Male C57BL/6 mice were obtained from The Jackson Laboratory. Mice were treated with antisense compounds targeting miR-122a, or received saline as a control treatment.
One of the antisense compounds tested included ISIS 327895, a 2'-MOE uniformly modified compound fully complementary to miR-122a. Also tested in vivo was ISIS 387574, a positionally modified antisense compound having the motif (A-A-B)7(-A-A)l where A is 2'-MOE and B is LNA. ISIS 387574 is fully complementary to miR-122a. Mice were administered 25 mg/kg doses of antisense compound intraperitoneally, for a total of 6 doses. Following the end of the treatment period, RNA was isolated from liver and the levels of a miR- 122a target mRNA, ALDOA, were measured using Taqman real-time PCR. Relative to saline-treated animals, treatment with the 2'-MOE uniformly modified compound resulted in ALDO A mRNA levels approximately 4 times those in saline-treated animals. Treatment with the positionally modified compound resulted in ALDO A mRNA levels approximately 5 times those observed in saline-treated animals. Thus, incorporation of a bicyclic sugar moiety into an otherwise uniformly modified background enhanced the ability of an antisense compound to inhibit miR-122a activity in vivo.
Serum total cholesterol levels were also monitored using methods known in the art (for example, via Olympus AU400e automated clinical chemistry analyzer, Melville, NY). Reductions in total cholesterol were observed in mice treated with either the 2'-MOE uniformly modified compound or the positionally modified 2 ' -MOE/LNA compound.
Additional measurements performed on serum samples include measurements of LDL- cholesterol, triglycerides, and serum tranaminases. Additional analyses that are performed in such in vivo studies included histological analysis of liver sections, to evaluate changes in morphology. Histological analysis of liver is carried out via routine procedures known in the art. Briefly, liver is fixed in 10% buffered formalin and embedded in paraffin wax. 4-mm sections are cut and mounted on glass slides. After dehydration, the sections are stained with hematoxylin and eosin. Morphological analysis may also include evaluation of hepatic steatosis, using oil Red O staining procedures known in the art.
Embodiments of the present invention include methods of reducing serum total cholesterol in an animal comprising administering an antisense compound targeted to a miR-122a nucleic acid. In one embodiment, the antisense compound has the nucleobase sequence of SEQ ID NO: 2. In another embodiment, the antisense compound is uniformly comprised of 2'-MOE sugar modifications. In additional embodiments, the antisense compound has a positionally modified motif, 5'-(A-A-B)n(-A)nn- 3', where A is 2'-MOE, B is LNA, n is 7, and nn is 2. In a further embodiment, the antiense compound is ISIS 327895. In another embodiment, the antisense compound is ISIS 387574. Additionally, either of these antisense compounds is used to achieve one or more additional phenotypic changes, such as lowered serum LDL-cholesterol, lowered serum triglycerides, or reduced hepatic steatosis.
In a similar study, antisense compounds targeted to miR-122a and having truncations were tested in vivo. Truncation of a single nucleoside from the 5' end of the antisense compound yielded increases in ALDO A mRNA levels comparable to those of the full-length antisense compound, ISIS 327895. Truncations of two nucleosides from the 5' end, 5 nucleosides from the 3' end, or 1 nucleoside from the 3' end resulted in slightly elevated ALDO A mRNA levels. Truncation of 3 nucleosides from the 3' end resulted in no increase in ALDO A mRNA levels. Accordinly, a further embodiment of the invention includes practicing any of the methods described herein using an antisense compound targeted to miR-122a, wherein the antisense compound has no more than 2 truncations from the 5' end or no more than 1 truncation from the 3' end, relative to the nucleobase sequence of SEQ ID NO: 2.

Claims

What is claimed is:
1. Use of an antisense compound essentially fully complementary to a miR-122 nucleic acid for the preparation of a medicament for lowering a serum indicator of cardiovascular disease risk wherein said serum indicator of cardiovascular disease risk is selected from elevated serum cholesterol levels, elevated serum triglyceride levels, or elevated lipoprotein levels.
2. The use of claim 1 , wherein the elevated serum cholesterol is elevated LDL-cholesterol.
3. The use of claim 1 , wherein the elevated serum cholesterol is elevated serum total cholesterol.
4. The use of claim 1 , wherein the elevated serum lipoprotein is elevated serum ApoB-100 protein.
5. Use of an antisense compound essentially fully complementary to a miR-122a nucleic acid in the preparation of a medicament for reducing hepatic steatosis.
6. The use of claim 5 wherein the hepatic steatosis is steatohepatitis or non-alcoholic steatohepatitis.
7. The use of an antisense compound essentially fully complementary to a miR-122a nucleic acid in the preparation of a medicament to treat a disease or disorder selected from diabetes, obesity, hyperlipidemia, hypercholesterolemia, hypertriglyceridemia, hyperfattyacidemia, nonalcoholic fatty liver disease, non alcoholic steatohepatitis, or metabolic syndrome.
8. The use of any of claims 1 to 7, wherein the antisense compound is 21 to 23 nucleosides in length.
9. The use of claim 8, wherein the antisense compound is fully complementary to a miR-122a target nucleic acid.
10. The use of claims 8 or 9, wherein the antisense compound comprises a plurality of 2'-sugar modified nucleosides.
11. The use of any of claims 8 to 10, wherein the antisense compound further comprises at least one bicyclic sugar modified nucleoside.
12. The use of any of claims 8 to 11, wherein the antisense compound comprises the nucleobase sequence of SEQ ID NO: 1.
13. The use of any of claims 8 to 12, wherein the antisense compound consists of the nucleobase sequence of SEQ TD NO: 2.
14. The use of any of claims 8 to 13, wherein the antisense compound is uniformly comprised of 2 ' -
MOE sugar modified nucleosides.
15. The use of any of claims 8 to 14, wherein the antisense compound comprises at least one phosphorothioate internucleoside linkage.
16. The use of any of claims 8 to 15, wherein the antisense compound comprises at least one 5- methylcytosine.
PCT/US2006/033866 2005-08-29 2006-08-29 Methods for use in modulating mir-122a WO2007027775A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
AT06813949T ATE494372T1 (en) 2005-08-29 2006-08-29 METHOD FOR MIR-122A MODULATION
AU2006284855A AU2006284855B2 (en) 2005-08-29 2006-08-29 Methods for use in modulating miR-122a
CA2620856A CA2620856C (en) 2005-08-29 2006-08-29 Methods for use in modulating mir-122a
JP2008529232A JP5523705B2 (en) 2005-08-29 2006-08-29 Method of using to modulate MIR-122A
EP06813949.2A EP1931782B2 (en) 2005-08-29 2006-08-29 Methods for use in modulating mir-122a
DE602006019455T DE602006019455D1 (en) 2005-08-29 2006-08-29 PROCESS FOR MIR-122A MODULATION

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US71221105P 2005-08-29 2005-08-29
US60/712,211 2005-08-29
US73137705P 2005-10-28 2005-10-28
US60/731,377 2005-10-28
US77159206P 2006-02-07 2006-02-07
US60/771,592 2006-02-07

Publications (2)

Publication Number Publication Date
WO2007027775A2 true WO2007027775A2 (en) 2007-03-08
WO2007027775A3 WO2007027775A3 (en) 2007-08-02

Family

ID=37809465

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/033866 WO2007027775A2 (en) 2005-08-29 2006-08-29 Methods for use in modulating mir-122a

Country Status (8)

Country Link
US (2) US8969314B2 (en)
EP (2) EP1931782B2 (en)
JP (1) JP5523705B2 (en)
AT (1) ATE494372T1 (en)
AU (1) AU2006284855B2 (en)
CA (1) CA2620856C (en)
DE (1) DE602006019455D1 (en)
WO (1) WO2007027775A2 (en)

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008049085A1 (en) * 2006-10-18 2008-04-24 Isis Pharmaceuticals, Inc. Antisense compounds
WO2009043354A2 (en) * 2007-10-04 2009-04-09 Santaris Pharma A/S Combination treatment for the treatment of hepatitis c virus infection
WO2009109665A1 (en) * 2008-03-07 2009-09-11 Santaris Pharma A/S Pharmaceutical compositions for treatment of microrna related diseases
WO2011017697A1 (en) 2009-08-07 2011-02-10 New York University Compositions and methods for treating inflammatory disorders
EP2338992A3 (en) * 2005-08-29 2011-10-12 Regulus Therapeutics, Inc Antisense compounds having enhanced anti-microRNA activity
WO2011133901A2 (en) 2010-04-23 2011-10-27 University Of Massachusetts Aav-based treatment of cholesterol-related disorders
WO2012027704A1 (en) 2010-08-27 2012-03-01 New York University Mir-33 inhibitors and uses thereof
US8163708B2 (en) 2006-04-03 2012-04-24 Santaris Pharma A/S Pharmaceutical composition comprising anti-mirna antisense oligonucleotide
WO2012145374A1 (en) * 2011-04-19 2012-10-26 Regulus Therapeutics Inc. TARGETING miR-378 FAMILY MEMBERS FOR THE TREATMENT OF METABOLIC DISORDERS
WO2012149557A1 (en) 2011-04-28 2012-11-01 New York University miR-33 INHIBITORS AND USES THEREOF TO DECREASE INFLAMMATION
WO2012175733A1 (en) 2011-06-23 2012-12-27 Santaris Pharma A/S Hcv combination therapy
WO2013000855A1 (en) 2011-06-30 2013-01-03 Santaris Pharma A/S Hcv combination therapy
WO2013000856A1 (en) 2011-06-30 2013-01-03 Santaris Pharma A/S Hcv combination therapy
WO2013068348A1 (en) 2011-11-07 2013-05-16 Santaris Pharma A/S Lna oligomers for improvement in hepatic function
WO2013068347A1 (en) 2011-11-07 2013-05-16 Santaris Pharma A/S Prognostic method for checking efficacy of micro rna-122 inhibitors in hcv+ patients
US8492357B2 (en) 2008-08-01 2013-07-23 Santaris Pharma A/S Micro-RNA mediated modulation of colony stimulating factors
US8546350B2 (en) 2003-07-31 2013-10-01 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US8598143B2 (en) 2003-11-26 2013-12-03 University Of Massachusetts Sequence-specific inhibition of small RNA function
US8729250B2 (en) 2006-04-03 2014-05-20 Joacim Elmén Antisense oligonucleotides for inhibition of microRNA-21
WO2014076195A1 (en) 2012-11-15 2014-05-22 Santaris Pharma A/S Oligonucleotide conjugates
WO2014118272A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Antimir-122 oligonucleotide carbohydrate conjugates
WO2014118267A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Lna oligonucleotide carbohydrate conjugates
US8815826B2 (en) 2010-07-23 2014-08-26 Regulus Therapeutics, Inc. Targeting microRNAs for the treatment of fibrosis
US8859519B2 (en) 2010-08-25 2014-10-14 The General Hospital Corporation Methods targeting miR-33 microRNAs for regulating lipid metabolism
WO2014179446A3 (en) * 2013-05-01 2015-02-19 Regulus Therapeutics Inc. Microrna compounds and methods for modulating mir-122
US8969314B2 (en) 2003-07-31 2015-03-03 Regulus Therapeutics, Inc. Methods for use in modulating miR-122a
AU2012216487B2 (en) * 2006-04-03 2015-05-14 Roche Innovation Center Copenhagen A/S Pharmaceutical composition comprising anti-miRNA antisense oligonucleotides
US9034837B2 (en) 2009-04-24 2015-05-19 Roche Innovation Center Copenhagen A/S Pharmaceutical compositions for treatment of HCV patients that are poor-responders to interferon
WO2015175539A1 (en) 2014-05-12 2015-11-19 The Johns Hopkins University Engineering synthetic brain penetrating gene vectors
WO2015175545A1 (en) 2014-05-12 2015-11-19 The Johns Hopkins University Highly stable biodegradable gene vector platforms for overcoming biological barriers
US9217155B2 (en) 2008-05-28 2015-12-22 University Of Massachusetts Isolation of novel AAV'S and uses thereof
US9226976B2 (en) 2011-04-21 2016-01-05 University Of Massachusetts RAAV-based compositions and methods for treating alpha-1 anti-trypsin deficiencies
WO2016042561A2 (en) 2014-09-21 2016-03-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Downregulating mir-132 for the treatment of lipid related disorders
US9394333B2 (en) 2008-12-02 2016-07-19 Wave Life Sciences Japan Method for the synthesis of phosphorus atom modified nucleic acids
WO2017021963A1 (en) 2015-08-03 2017-02-09 Biokine Therapeutics Ltd. Cxcr4 binding agents for treatment of diseases
US9598458B2 (en) 2012-07-13 2017-03-21 Wave Life Sciences Japan, Inc. Asymmetric auxiliary group
US9605019B2 (en) 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US9617547B2 (en) 2012-07-13 2017-04-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
US9644207B2 (en) 2013-03-14 2017-05-09 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating Tau expression
US9683235B2 (en) 2013-07-19 2017-06-20 Ionis Pharmaceuticals, Inc. Compositions for modulating Tau expression
US9695418B2 (en) 2012-10-11 2017-07-04 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleosides and uses thereof
US9744183B2 (en) 2009-07-06 2017-08-29 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US9914922B2 (en) 2012-04-20 2018-03-13 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
US10017764B2 (en) 2011-02-08 2018-07-10 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
US10072251B2 (en) 2014-02-19 2018-09-11 University Of Massachusetts Recombinant AAVS having useful transcytosis properties
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design
US10273474B2 (en) 2012-03-30 2019-04-30 Washington University Methods for modulating Tau expression for reducing seizure and modifying a neurodegenerative syndrome
US10280418B2 (en) 2014-03-18 2019-05-07 Univeristy Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10370432B2 (en) 2014-10-03 2019-08-06 University Of Massachusetts Heterologous targeting peptide grafted AAVS
US10407680B2 (en) 2016-09-29 2019-09-10 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing Tau expression
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
US10457940B2 (en) 2016-09-22 2019-10-29 University Of Massachusetts AAV treatment of Huntington's disease
US10480011B2 (en) 2014-10-21 2019-11-19 University Of Massachusetts Recombinant AAV variants and uses thereof
WO2020038968A1 (en) 2018-08-23 2020-02-27 Roche Innovation Center Copenhagen A/S Microrna-134 biomarker
US10584321B2 (en) 2015-02-13 2020-03-10 University Of Massachusetts Compositions and methods for transient delivery of nucleases
WO2020261227A1 (en) 2019-06-26 2020-12-30 Biorchestra Co., Ltd. Micellar nanoparticles and uses thereof
US10975391B2 (en) 2014-04-25 2021-04-13 University Of Massachusetts Recombinant AAV vectors useful for reducing immunity against transgene products
US11046955B2 (en) 2015-04-24 2021-06-29 University Of Massachusetts Modified AAV constructs and uses thereof
US11060088B2 (en) 2016-02-12 2021-07-13 University Of Massachusetts Anti-angiogenic miRNA therapeutics for inhibiting corneal neovascularization
WO2022174113A1 (en) 2021-02-12 2022-08-18 Merand Pharmaceuticals, Inc. Agents, compositions, and methods for the treatment of hypoxia and ischemia-related disorders
US11578107B2 (en) 2016-12-22 2023-02-14 Ohio State Innovation Foundation Compositions and methods for reprogramming somatic cells into induced vasculogenic cells
US11578340B2 (en) 2016-10-13 2023-02-14 University Of Massachusetts AAV capsid designs
US11732261B2 (en) 2011-08-11 2023-08-22 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
US11739330B2 (en) 2017-09-22 2023-08-29 University Of Massachusetts SOD1 dual expression vectors and uses thereof
US11834474B2 (en) 2009-05-28 2023-12-05 University Of Massachusetts AAV's and uses thereof
US11859179B2 (en) 2017-05-09 2024-01-02 University Of Massachusetts Methods of treating amyotrophic lateral sclerosis (ALS)

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070213292A1 (en) * 2005-08-10 2007-09-13 The Rockefeller University Chemically modified oligonucleotides for use in modulating micro RNA and uses thereof
EP2489742A1 (en) * 2006-10-09 2012-08-22 Julius-Maximilians-Universität Würzburg MicroRNA (miRNA) for the diagnosis and treatment of heart diseases
US20110046206A1 (en) * 2007-06-22 2011-02-24 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
EP2723865B1 (en) 2011-06-21 2019-03-27 Alnylam Pharmaceuticals, Inc. METHODS FOR DETERMINING ACTIVITY OF RNAi IN A SUBJECT
US10202643B2 (en) 2011-10-31 2019-02-12 University Of Utah Research Foundation Genetic alterations in glioma
US10260089B2 (en) 2012-10-29 2019-04-16 The Research Foundation Of The State University Of New York Compositions and methods for recognition of RNA using triple helical peptide nucleic acids
KR101570269B1 (en) 2013-06-19 2015-11-19 가톨릭대학교 산학협력단 Biomaker miR-451 for distinguishing non-alcoholic steatohepatitis
CN106536736B (en) * 2014-07-31 2021-02-19 新加坡科技研究局 Modified anti-MIR-138 oligonucleotides
JP7441002B2 (en) * 2015-10-14 2024-02-29 バイオ-パス ホールディングス, インコーポレイテッド p-ethoxy nucleic acids for liposome formulations
ES2928654T3 (en) 2016-09-16 2022-11-21 Bio Path Holdings Inc Combination therapy with liposomal antisense oligonucleotides
EP3548005A4 (en) 2016-11-29 2020-06-17 Puretech Health LLC Exosomes for delivery of therapeutic agents
BR112019021771A2 (en) 2017-04-19 2020-05-05 Bio Path Holdings Inc p-ethoxy nucleic acids for stat3 inhibition
CN109722435B (en) * 2018-04-08 2021-03-16 中国药科大学 Islet long-chain non-coding RNA1810019D21Rik and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003029459A2 (en) * 2001-09-28 2003-04-10 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Micro-rna molecules
WO2005013901A2 (en) * 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas

Family Cites Families (134)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US4476301A (en) 1982-04-29 1984-10-09 Centre National De La Recherche Scientifique Oligonucleotides, a process for preparing the same and their application as mediators of the action of interferon
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
US5550111A (en) 1984-07-11 1996-08-27 Temple University-Of The Commonwealth System Of Higher Education Dual action 2',5'-oligoadenylate antiviral derivatives and uses thereof
FR2567892B1 (en) 1984-07-19 1989-02-17 Centre Nat Rech Scient NOVEL OLIGONUCLEOTIDES, THEIR PREPARATION PROCESS AND THEIR APPLICATIONS AS MEDIATORS IN DEVELOPING THE EFFECTS OF INTERFERONS
US5367066A (en) 1984-10-16 1994-11-22 Chiron Corporation Oligonucleotides with selectably cleavable and/or abasic sites
FR2575751B1 (en) 1985-01-08 1987-04-03 Pasteur Institut NOVEL ADENOSINE DERIVATIVE NUCLEOSIDES, THEIR PREPARATION AND THEIR BIOLOGICAL APPLICATIONS
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5166315A (en) 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5405938A (en) 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
AU598946B2 (en) 1987-06-24 1990-07-05 Howard Florey Institute Of Experimental Physiology And Medicine Nucleoside derivatives
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
WO1989009221A1 (en) 1988-03-25 1989-10-05 University Of Virginia Alumni Patents Foundation Oligonucleotide n-alkylphosphoramidates
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5194599A (en) 1988-09-23 1993-03-16 Gilead Sciences, Inc. Hydrogen phosphonodithioate compositions
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
US5721218A (en) 1989-10-23 1998-02-24 Gilead Sciences, Inc. Oligonucleotides with inverted polarity
WO1991006556A1 (en) 1989-10-24 1991-05-16 Gilead Sciences, Inc. 2' modified oligonucleotides
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5321131A (en) 1990-03-08 1994-06-14 Hybridon, Inc. Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
GB9009980D0 (en) 1990-05-03 1990-06-27 Amersham Int Plc Phosphoramidite derivatives,their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
ES2116977T3 (en) 1990-05-11 1998-08-01 Microprobe Corp SOLID SUPPORTS FOR NUCLEIC ACID HYBRIDIZATION TESTS AND METHODS TO IMMOBILIZE OLIGONUCLEOTIDES IN A COVALENT WAY.
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
WO1992002258A1 (en) 1990-07-27 1992-02-20 Isis Pharmaceuticals, Inc. Nuclease resistant, pyrimidine modified oligonucleotides that detect and modulate gene expression
EP0541722B1 (en) 1990-08-03 1995-12-20 Sterling Winthrop Inc. Compounds and methods for inhibiting gene expression
US5177196A (en) 1990-08-16 1993-01-05 Microprobe Corporation Oligo (α-arabinofuranosyl nucleotides) and α-arabinofuranosyl precursors thereof
US5214134A (en) 1990-09-12 1993-05-25 Sterling Winthrop Inc. Process of linking nucleosides with a siloxane bridge
US5561225A (en) 1990-09-19 1996-10-01 Southern Research Institute Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
WO1992005186A1 (en) 1990-09-20 1992-04-02 Gilead Sciences Modified internucleoside linkages
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
US5672697A (en) 1991-02-08 1997-09-30 Gilead Sciences, Inc. Nucleoside 5'-methylene phosphonates
US5539082A (en) 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5719262A (en) 1993-11-22 1998-02-17 Buchardt, Deceased; Ole Peptide nucleic acids having amino acid side chains
US5714331A (en) 1991-05-24 1998-02-03 Buchardt, Deceased; Ole Peptide nucleic acids having enhanced binding affinity, sequence specificity and solubility
US5571799A (en) 1991-08-12 1996-11-05 Basco, Ltd. (2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response
EP0538194B1 (en) 1991-10-17 1997-06-04 Novartis AG Bicyclic nucleosides, oligonucleotides, their method of preparation and intermediates therein
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
WO1993010820A1 (en) 1991-11-26 1993-06-10 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
TW393513B (en) 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5792608A (en) 1991-12-12 1998-08-11 Gilead Sciences, Inc. Nuclease stable and binding competent oligomers and methods for their use
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
FR2687679B1 (en) 1992-02-05 1994-10-28 Centre Nat Rech Scient OLIGOTHIONUCLEOTIDES.
US5633360A (en) 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
EP0577558A2 (en) 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclic nucleosides having bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
GB9304618D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
EP0691968B1 (en) 1993-03-30 1997-07-16 Sanofi Acyclic nucleoside analogs and oligonucleotide sequences containing them
ATE160572T1 (en) 1993-03-31 1997-12-15 Sanofi Sa OLIGONUCLEOTIDES WITH AMIDE CHAINS THAT USE PHOSPHOESTER CHAINS
DE4311944A1 (en) 1993-04-10 1994-10-13 Degussa Coated sodium percarbonate particles, process for their preparation and detergent, cleaning and bleaching compositions containing them
US5614621A (en) 1993-07-29 1997-03-25 Isis Pharmaceuticals, Inc. Process for preparing oligonucleotides using silyl-containing diamino phosphorous reagents
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
US5446137B1 (en) 1993-12-09 1998-10-06 Behringwerke Ag Oligonucleotides containing 4'-substituted nucleotides
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5627053A (en) 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5646269A (en) 1994-04-28 1997-07-08 Gilead Sciences, Inc. Method for oligonucleotide analog synthesis
US5525711A (en) 1994-05-18 1996-06-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pteridine nucleotide analogs as fluorescent DNA probes
US5597909A (en) 1994-08-25 1997-01-28 Chiron Corporation Polynucleotide reagents containing modified deoxyribose moieties, and associated methods of synthesis and use
US5792747A (en) 1995-01-24 1998-08-11 The Administrators Of The Tulane Educational Fund Highly potent agonists of growth hormone releasing hormone
US5705621A (en) 1995-11-17 1998-01-06 Isis Pharmaceuticals, Inc. Oligomeric phosphite, phosphodiester, Phosphorothioate and phosphorodithioate compounds and intermediates for preparing same
AU1430097A (en) 1996-01-16 1997-08-11 Ribozyme Pharmaceuticals, Inc. Synthesis of methoxy nucleosides and enzymatic nucleic acid molecules
US6172209B1 (en) 1997-02-14 2001-01-09 Isis Pharmaceuticals Inc. Aminooxy-modified oligonucleotides and methods for making same
US5760209A (en) 1997-03-03 1998-06-02 Isis Pharmaceuticals, Inc. Protecting group for synthesizing oligonucleotide analogs
JP3756313B2 (en) 1997-03-07 2006-03-15 武 今西 Novel bicyclonucleosides and oligonucleotide analogues
DE69829760T3 (en) 1997-09-12 2016-04-14 Exiqon A/S BI- AND TRI-CYCLIC-NUCLEOSIDE, NUCLEOTIDE AND OLIGONUCLEOTIDE ANALOG
US6028183A (en) 1997-11-07 2000-02-22 Gilead Sciences, Inc. Pyrimidine derivatives and oligonucleotides containing same
US6007992A (en) 1997-11-10 1999-12-28 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US6020475A (en) 1998-02-10 2000-02-01 Isis Pharmeuticals, Inc. Process for the synthesis of oligomeric compounds
US6326478B1 (en) 1998-07-08 2001-12-04 Isis Pharmaceuticals, Inc. Process for the synthesis of oligomeric compounds
US6271358B1 (en) 1998-07-27 2001-08-07 Isis Pharmaceuticals, Inc. RNA targeted 2′-modified oligonucleotides that are conformationally preorganized
US6169177B1 (en) 1998-11-06 2001-01-02 Isis Pharmaceuticals, Inc. Processes for the synthesis of oligomeric compounds
US6465628B1 (en) 1999-02-04 2002-10-15 Isis Pharmaceuticals, Inc. Process for the synthesis of oligomeric compounds
US6121437A (en) 1999-03-16 2000-09-19 Isis Pharmaceuticals, Inc. Phosphate and thiophosphate protecting groups
US7084125B2 (en) 1999-03-18 2006-08-01 Exiqon A/S Xylo-LNA analogues
DK1178999T3 (en) 1999-05-04 2007-08-06 Santaris Pharma As L-ribo-LNA analogues
US6593466B1 (en) 1999-07-07 2003-07-15 Isis Pharmaceuticals, Inc. Guanidinium functionalized nucleotides and precursors thereof
US6147200A (en) 1999-08-19 2000-11-14 Isis Pharmaceuticals, Inc. 2'-O-acetamido modified monomers and oligomers
JP2003511016A (en) 1999-10-04 2003-03-25 エクシコン エ/エス Design of High Affinity RNase H to Replenish Oligonucleotides
WO2002028880A2 (en) * 2000-10-06 2002-04-11 Aeson Therapeutics Inc. Compounds useful for treating hypertriglyceridemia
WO2002097134A2 (en) 2001-05-25 2002-12-05 Isis Pharmaceuticals, Inc. Modified peptide nucleic acid
US20030175906A1 (en) 2001-07-03 2003-09-18 Muthiah Manoharan Nuclease resistant chimeric oligonucleotides
WO2003004602A2 (en) 2001-07-03 2003-01-16 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
US20030158403A1 (en) 2001-07-03 2003-08-21 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
US7034751B2 (en) 2002-05-20 2006-04-25 Raytheon Company Reflective and transmissive mode monolithic millimeter wave array system and in-line amplifier using same
US7511131B2 (en) * 2002-11-13 2009-03-31 Genzyme Corporation Antisense modulation of apolipoprotein B expression
US8124582B2 (en) * 2002-12-06 2012-02-28 Fibrogen, Inc. Treatment of diabetes
US7825235B2 (en) * 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
CA2538843A1 (en) * 2003-09-12 2005-03-24 Vertex Pharmaceuticals Incorporated Animal model for protease activity and liver damage
EP1747023B2 (en) 2004-05-04 2016-03-09 The Board of Trustees of The Leland Stanford Junior University Methods and compositions for reducing hcv viral genome amounts in a target cell
AU2005272816B2 (en) * 2004-08-10 2011-08-11 Alnylam Pharmaceuticals, Inc. Chemically modified oligonucleotides
DK1786472T3 (en) * 2004-08-10 2013-04-15 Genzyme Corp Antisense modulation of apolipoprotein B expression
EP2397563A3 (en) * 2004-09-17 2012-07-18 Isis Pharmaceuticals, Inc. Enhanced antisense oligonucleotides
US20060185027A1 (en) * 2004-12-23 2006-08-17 David Bartel Systems and methods for identifying miRNA targets and for altering miRNA and target expression
US20060265771A1 (en) * 2005-05-17 2006-11-23 Lewis David L Monitoring microrna expression and function
US20070213292A1 (en) * 2005-08-10 2007-09-13 The Rockefeller University Chemically modified oligonucleotides for use in modulating micro RNA and uses thereof
CA2618995A1 (en) * 2005-08-10 2007-02-22 The Rockefeller University Antagomirs for use in inhibiting mir-122
JP5523705B2 (en) 2005-08-29 2014-06-18 レグルス・セラピューティクス・インコーポレイテッド Method of using to modulate MIR-122A
EP2338992A3 (en) 2005-08-29 2011-10-12 Regulus Therapeutics, Inc Antisense compounds having enhanced anti-microRNA activity
EA015570B1 (en) 2006-04-03 2011-10-31 Сантарис Фарма А/С Pharmaceutical composition
WO2013080784A1 (en) 2011-11-30 2013-06-06 シャープ株式会社 Memory circuit, drive method for same, nonvolatile storage device using same, and liquid crystal display device
KR101595393B1 (en) 2014-05-22 2016-02-18 엘지전자 주식회사 Information providing system and method thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003029459A2 (en) * 2001-09-28 2003-04-10 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Micro-rna molecules
WO2005013901A2 (en) * 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ESAU CHRISTINE ET AL: "miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting" CELL METABOLISM, vol. 3, no. 2, February 2006 (2006-02), pages 87-98 URL, XP002424535 ISSN: 1550-4131 *

Cited By (165)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9994842B2 (en) 2003-07-31 2018-06-12 Regulus Therapeutics Inc. Methods for use in modulating miR-122A
US9447413B2 (en) 2003-07-31 2016-09-20 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US8946179B2 (en) 2003-07-31 2015-02-03 Regulus Therapeutics, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9447412B2 (en) 2003-07-31 2016-09-20 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAS
US8859521B2 (en) 2003-07-31 2014-10-14 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
EP2530157B1 (en) 2003-07-31 2016-09-28 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of miRNAs
US10093926B2 (en) 2003-07-31 2018-10-09 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US10584336B2 (en) 2003-07-31 2020-03-10 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9139832B2 (en) 2003-07-31 2015-09-22 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US8969314B2 (en) 2003-07-31 2015-03-03 Regulus Therapeutics, Inc. Methods for use in modulating miR-122a
US8697663B2 (en) 2003-07-31 2014-04-15 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9663787B2 (en) 2003-07-31 2017-05-30 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US10072265B2 (en) 2003-07-31 2018-09-11 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9267138B2 (en) 2003-07-31 2016-02-23 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9528108B2 (en) 2003-07-31 2016-12-27 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAS
US8809294B2 (en) 2003-07-31 2014-08-19 Regulus Therapeutics Inc. Method of inhibiting miR-33 using a modified oligonucleotide
US8765701B2 (en) 2003-07-31 2014-07-01 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9752146B2 (en) 2003-07-31 2017-09-05 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US8546350B2 (en) 2003-07-31 2013-10-01 Regulus Therapeutics Inc. Oligomeric compounds and compositions for use in modulation of small non-coding RNAs
US9334497B2 (en) 2003-11-26 2016-05-10 University Of Massachusetts Sequence-specific inhibition of small RNA function
US11359196B2 (en) 2003-11-26 2022-06-14 University Of Massachusetts Sequence-specific inhibition of small RNA function
US8685946B2 (en) 2003-11-26 2014-04-01 Universiy of Massachusetts Sequence-specific inhibition of small RNA function
US8598143B2 (en) 2003-11-26 2013-12-03 University Of Massachusetts Sequence-specific inhibition of small RNA function
EP1931780B1 (en) * 2005-08-29 2016-01-06 Regulus Therapeutics Inc. Antisense compounds having enhanced anti-microrna activity
EP2338992A3 (en) * 2005-08-29 2011-10-12 Regulus Therapeutics, Inc Antisense compounds having enhanced anti-microRNA activity
EP3502255A1 (en) * 2006-04-03 2019-06-26 Roche Innovation Center Copenhagen A/S Pharmaceutical composition
US8729250B2 (en) 2006-04-03 2014-05-20 Joacim Elmén Antisense oligonucleotides for inhibition of microRNA-21
AU2012216487B2 (en) * 2006-04-03 2015-05-14 Roche Innovation Center Copenhagen A/S Pharmaceutical composition comprising anti-miRNA antisense oligonucleotides
US8163708B2 (en) 2006-04-03 2012-04-24 Santaris Pharma A/S Pharmaceutical composition comprising anti-mirna antisense oligonucleotide
JP2010507579A (en) * 2006-10-18 2010-03-11 アイシス ファーマシューティカルズ, インコーポレーテッド Antisense compound
EP2410054A1 (en) * 2006-10-18 2012-01-25 Isis Pharmaceuticals, Inc. Antisense compounds
AU2007310989B2 (en) * 2006-10-18 2014-05-29 Isis Pharmaceuticals, Inc. Antisense compounds
EP2410053A1 (en) * 2006-10-18 2012-01-25 Isis Pharmaceuticals, Inc. Antisense compounds
AU2014203243A1 (en) * 2006-10-18 2014-07-10 Ionis Pharmaceuticals, Inc. Antisense compounds
JP2014221817A (en) * 2006-10-18 2014-11-27 アイシス ファーマシューティカルズ, インコーポレーテッド Antisense compounds
US9550988B2 (en) 2006-10-18 2017-01-24 Ionis Pharmaceuticals, Inc. Antisense compounds
AU2014203243B2 (en) * 2006-10-18 2016-06-30 Ionis Pharmaceuticals, Inc. Antisense compounds
WO2008049085A1 (en) * 2006-10-18 2008-04-24 Isis Pharmaceuticals, Inc. Antisense compounds
US10450564B2 (en) 2007-10-04 2019-10-22 Roche Innovation Center Copenhagen A/S Micromirs
WO2009043354A2 (en) * 2007-10-04 2009-04-09 Santaris Pharma A/S Combination treatment for the treatment of hepatitis c virus infection
US8440637B2 (en) 2007-10-04 2013-05-14 Santaris Pharma A/S Combination treatment for the treatment of hepatitis C virus infection
US8906871B2 (en) 2007-10-04 2014-12-09 Santaris Pharma A/S MicromiRs
WO2009043354A3 (en) * 2007-10-04 2009-08-20 Santaris Pharma As Combination treatment for the treatment of hepatitis c virus infection
US8288356B2 (en) 2007-10-04 2012-10-16 Santaris Pharma A/S MicroRNAs
US8361980B2 (en) 2008-03-07 2013-01-29 Santaris Pharma A/S Pharmaceutical compositions for treatment of microRNA related diseases
AU2009221064B2 (en) * 2008-03-07 2014-12-11 Roche Innovation Center Copenhagen A/S Pharmaceutical compositions for treatment of microRNA related diseases
US8404659B2 (en) 2008-03-07 2013-03-26 Santaris Pharma A/S Pharmaceutical compositions for treatment of MicroRNA related diseases
WO2009109665A1 (en) * 2008-03-07 2009-09-11 Santaris Pharma A/S Pharmaceutical compositions for treatment of microrna related diseases
US9217155B2 (en) 2008-05-28 2015-12-22 University Of Massachusetts Isolation of novel AAV'S and uses thereof
US10905776B2 (en) 2008-05-28 2021-02-02 University Of Massachusetts Isolation of novel AAV's and uses thereof
US11826434B2 (en) 2008-05-28 2023-11-28 University Of Massachusetts Isolation of novel AAV's and uses thereof
US10300146B2 (en) 2008-05-28 2019-05-28 University Of Massachusetts Isolation of novel AAV's and uses thereof
US9596835B2 (en) 2008-05-28 2017-03-21 University Of Massachusetts Isolation of novel AAV's and uses thereof
US10166297B2 (en) 2008-05-28 2019-01-01 University Of Massachusetts Isolation of novel AAV's and uses thereof
US8492357B2 (en) 2008-08-01 2013-07-23 Santaris Pharma A/S Micro-RNA mediated modulation of colony stimulating factors
US10329318B2 (en) 2008-12-02 2019-06-25 Wave Life Sciences Ltd. Method for the synthesis of phosphorus atom modified nucleic acids
US9695211B2 (en) 2008-12-02 2017-07-04 Wave Life Sciences Japan, Inc. Method for the synthesis of phosphorus atom modified nucleic acids
US9394333B2 (en) 2008-12-02 2016-07-19 Wave Life Sciences Japan Method for the synthesis of phosphorus atom modified nucleic acids
US9034837B2 (en) 2009-04-24 2015-05-19 Roche Innovation Center Copenhagen A/S Pharmaceutical compositions for treatment of HCV patients that are poor-responders to interferon
US11834474B2 (en) 2009-05-28 2023-12-05 University Of Massachusetts AAV's and uses thereof
US9744183B2 (en) 2009-07-06 2017-08-29 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US10307434B2 (en) 2009-07-06 2019-06-04 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
WO2011017697A1 (en) 2009-08-07 2011-02-10 New York University Compositions and methods for treating inflammatory disorders
US11421230B2 (en) 2010-04-23 2022-08-23 University Of Massachusetts AAV-based treatment of cholesterol-related disorders
US10202600B2 (en) 2010-04-23 2019-02-12 University Of Massachusetts AAV-based treatment of cholesterol-related disorders
US9272053B2 (en) 2010-04-23 2016-03-01 University Of Massachusetts AAV-based treatment of cholesterol-related disorders
EP3444346A1 (en) * 2010-04-23 2019-02-20 University of Massachusetts Aav-based treatment of cholesterol-related disorders
US10731158B2 (en) 2010-04-23 2020-08-04 University Of Massachusetts AAV-based treatment of cholesterol-related disorders
WO2011133901A2 (en) 2010-04-23 2011-10-27 University Of Massachusetts Aav-based treatment of cholesterol-related disorders
EP2561075A2 (en) * 2010-04-23 2013-02-27 University of Massachusetts Aav-based treatment of cholesterol-related disorders
EP2561075A4 (en) * 2010-04-23 2015-04-15 Univ Massachusetts Aav-based treatment of cholesterol-related disorders
US9181548B2 (en) 2010-07-23 2015-11-10 Regulus Therapeutics Inc. Targeting micrornas for the treatment of fibrosis
US9556435B2 (en) 2010-07-23 2017-01-31 Regulus Therapeutics Inc. Targeting microRNAs for the treatment of fibrosis
US9970007B2 (en) 2010-07-23 2018-05-15 Regulus Therapeutics Inc. Targeting microRNAs for the treatment of fibrosis
US8815826B2 (en) 2010-07-23 2014-08-26 Regulus Therapeutics, Inc. Targeting microRNAs for the treatment of fibrosis
US8859519B2 (en) 2010-08-25 2014-10-14 The General Hospital Corporation Methods targeting miR-33 microRNAs for regulating lipid metabolism
WO2012027704A1 (en) 2010-08-27 2012-03-01 New York University Mir-33 inhibitors and uses thereof
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
US10017764B2 (en) 2011-02-08 2018-07-10 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
WO2012145374A1 (en) * 2011-04-19 2012-10-26 Regulus Therapeutics Inc. TARGETING miR-378 FAMILY MEMBERS FOR THE TREATMENT OF METABOLIC DISORDERS
US10597656B2 (en) 2011-04-21 2020-03-24 University Of Massachusetts RAAV-based compositions and methods
US10077452B2 (en) 2011-04-21 2018-09-18 University Of Massachusetts rAAV-based compositions and methods
US11920133B2 (en) 2011-04-21 2024-03-05 University Of Massachusetts RAAV-based compositions and methods
US9885057B2 (en) 2011-04-21 2018-02-06 University Of Massachusetts RAAV-based compositions and methods for treating alpha-1 anti-trypsin deficiencies
US11254939B2 (en) 2011-04-21 2022-02-22 University Of Massachusetts RAAV-based compositions and methods
US9226976B2 (en) 2011-04-21 2016-01-05 University Of Massachusetts RAAV-based compositions and methods for treating alpha-1 anti-trypsin deficiencies
WO2012149557A1 (en) 2011-04-28 2012-11-01 New York University miR-33 INHIBITORS AND USES THEREOF TO DECREASE INFLAMMATION
US9241950B2 (en) 2011-04-28 2016-01-26 New York University MiR-33 inhibitors and uses thereof to decrease inflammation
WO2012175733A1 (en) 2011-06-23 2012-12-27 Santaris Pharma A/S Hcv combination therapy
WO2013000855A1 (en) 2011-06-30 2013-01-03 Santaris Pharma A/S Hcv combination therapy
WO2013000856A1 (en) 2011-06-30 2013-01-03 Santaris Pharma A/S Hcv combination therapy
US9605019B2 (en) 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US10280192B2 (en) 2011-07-19 2019-05-07 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US11732261B2 (en) 2011-08-11 2023-08-22 Ionis Pharmaceuticals, Inc. Selective antisense compounds and uses thereof
WO2013068347A1 (en) 2011-11-07 2013-05-16 Santaris Pharma A/S Prognostic method for checking efficacy of micro rna-122 inhibitors in hcv+ patients
WO2013068348A1 (en) 2011-11-07 2013-05-16 Santaris Pharma A/S Lna oligomers for improvement in hepatic function
US10273474B2 (en) 2012-03-30 2019-04-30 Washington University Methods for modulating Tau expression for reducing seizure and modifying a neurodegenerative syndrome
US11781135B2 (en) 2012-03-30 2023-10-10 Washington University Methods for treating Alzheimer's disease
US9914922B2 (en) 2012-04-20 2018-03-13 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
US11566245B2 (en) 2012-04-20 2023-01-31 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
US10590413B2 (en) 2012-07-13 2020-03-17 Wave Life Sciences Ltd. Chiral control
US9598458B2 (en) 2012-07-13 2017-03-21 Wave Life Sciences Japan, Inc. Asymmetric auxiliary group
US10167309B2 (en) 2012-07-13 2019-01-01 Wave Life Sciences Ltd. Asymmetric auxiliary group
US9617547B2 (en) 2012-07-13 2017-04-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
US9695418B2 (en) 2012-10-11 2017-07-04 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleosides and uses thereof
US11155816B2 (en) 2012-11-15 2021-10-26 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugates
EP3406718A1 (en) 2012-11-15 2018-11-28 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugates
US10077443B2 (en) 2012-11-15 2018-09-18 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugates
WO2014076195A1 (en) 2012-11-15 2014-05-22 Santaris Pharma A/S Oligonucleotide conjugates
WO2014118272A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Antimir-122 oligonucleotide carbohydrate conjugates
WO2014118267A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Lna oligonucleotide carbohydrate conjugates
US11155815B2 (en) 2013-03-14 2021-10-26 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating Tau expression
US9644207B2 (en) 2013-03-14 2017-05-09 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating Tau expression
KR102214740B1 (en) 2013-05-01 2021-02-10 레굴루스 테라퓨틱스 인크 Microrna compounds and methods for modulating mir-122
EA034731B1 (en) * 2013-05-01 2020-03-13 Регьюлэс Терапьютикс Инк. MICRORNA COMPOUNDS AND METHODS FOR MODULATING miR-122 ACTIVITY
US9157083B2 (en) 2013-05-01 2015-10-13 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-122
US9309513B2 (en) 2013-05-01 2016-04-12 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-122
US9574194B2 (en) 2013-05-01 2017-02-21 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-122
KR20160002862A (en) * 2013-05-01 2016-01-08 레굴루스 테라퓨틱스 인크 Microrna compounds and methods for modulating mir-122
WO2014179446A3 (en) * 2013-05-01 2015-02-19 Regulus Therapeutics Inc. Microrna compounds and methods for modulating mir-122
US10150967B2 (en) 2013-05-01 2018-12-11 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-122
US10793856B2 (en) 2013-07-19 2020-10-06 Biogen Ma Inc. Compositions for modulating Tau expression
US11591595B2 (en) 2013-07-19 2023-02-28 Biogen Ma Inc. Compositions for modulating Tau expression
US9683235B2 (en) 2013-07-19 2017-06-20 Ionis Pharmaceuticals, Inc. Compositions for modulating Tau expression
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design
US10894949B2 (en) 2014-02-19 2021-01-19 University Of Massachusetts Recombinant AAVS having useful transcytosis properties
US10072251B2 (en) 2014-02-19 2018-09-11 University Of Massachusetts Recombinant AAVS having useful transcytosis properties
US10954518B2 (en) 2014-03-18 2021-03-23 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10851375B2 (en) 2014-03-18 2020-12-01 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10711274B2 (en) 2014-03-18 2020-07-14 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10280418B2 (en) 2014-03-18 2019-05-07 Univeristy Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US11760999B2 (en) 2014-03-18 2023-09-19 University Of Massachusetts RAAV-based compositions and methods for treating amyotrophic lateral sclerosis
US10975391B2 (en) 2014-04-25 2021-04-13 University Of Massachusetts Recombinant AAV vectors useful for reducing immunity against transgene products
US9937270B2 (en) 2014-05-12 2018-04-10 The John Hopkins University Engineering synthethic brain penetrating gene vectors
WO2015175545A1 (en) 2014-05-12 2015-11-19 The Johns Hopkins University Highly stable biodegradable gene vector platforms for overcoming biological barriers
US10695442B2 (en) 2014-05-12 2020-06-30 The Johns Hopkins University Engineering synthetic brain penetrating gene vectors
WO2015175539A1 (en) 2014-05-12 2015-11-19 The Johns Hopkins University Engineering synthetic brain penetrating gene vectors
WO2016042561A2 (en) 2014-09-21 2016-03-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Downregulating mir-132 for the treatment of lipid related disorders
US10370432B2 (en) 2014-10-03 2019-08-06 University Of Massachusetts Heterologous targeting peptide grafted AAVS
US11014976B2 (en) 2014-10-03 2021-05-25 University Of Massachusetts Heterologous targeting peptide grafted AAVS
US10480011B2 (en) 2014-10-21 2019-11-19 University Of Massachusetts Recombinant AAV variants and uses thereof
US11542525B2 (en) 2014-10-21 2023-01-03 University Of Massachusetts Recombinant AAV variants and uses thereof
US10584321B2 (en) 2015-02-13 2020-03-10 University Of Massachusetts Compositions and methods for transient delivery of nucleases
US11920168B2 (en) 2015-02-13 2024-03-05 University Of Massachusetts Compositions and methods for transient delivery of nucleases
US11046955B2 (en) 2015-04-24 2021-06-29 University Of Massachusetts Modified AAV constructs and uses thereof
WO2017021963A1 (en) 2015-08-03 2017-02-09 Biokine Therapeutics Ltd. Cxcr4 binding agents for treatment of diseases
US11851657B2 (en) 2016-02-12 2023-12-26 University Of Massachusetts Anti-angiogenic miRNA therapeutics for inhibiting corneal neovascularization
US11060088B2 (en) 2016-02-12 2021-07-13 University Of Massachusetts Anti-angiogenic miRNA therapeutics for inhibiting corneal neovascularization
US11046957B2 (en) 2016-09-22 2021-06-29 University Of Massachusetts AAV treatment of Huntington's disease
US11773392B2 (en) 2016-09-22 2023-10-03 University Of Massachusetts AAV treatment of Huntington's disease
US10457940B2 (en) 2016-09-22 2019-10-29 University Of Massachusetts AAV treatment of Huntington's disease
US11053498B2 (en) 2016-09-29 2021-07-06 Biogen Ma Inc. Compounds and methods for reducing Tau expression
US10407680B2 (en) 2016-09-29 2019-09-10 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing Tau expression
US11578340B2 (en) 2016-10-13 2023-02-14 University Of Massachusetts AAV capsid designs
US11578107B2 (en) 2016-12-22 2023-02-14 Ohio State Innovation Foundation Compositions and methods for reprogramming somatic cells into induced vasculogenic cells
US11859179B2 (en) 2017-05-09 2024-01-02 University Of Massachusetts Methods of treating amyotrophic lateral sclerosis (ALS)
US11739330B2 (en) 2017-09-22 2023-08-29 University Of Massachusetts SOD1 dual expression vectors and uses thereof
WO2020038968A1 (en) 2018-08-23 2020-02-27 Roche Innovation Center Copenhagen A/S Microrna-134 biomarker
US11839624B2 (en) 2019-06-26 2023-12-12 Biorchestra Co., Ltd. Micellar nanoparticles and uses thereof
WO2020261227A1 (en) 2019-06-26 2020-12-30 Biorchestra Co., Ltd. Micellar nanoparticles and uses thereof
WO2022174113A1 (en) 2021-02-12 2022-08-18 Merand Pharmaceuticals, Inc. Agents, compositions, and methods for the treatment of hypoxia and ischemia-related disorders

Also Published As

Publication number Publication date
ATE494372T1 (en) 2011-01-15
EP1931782A2 (en) 2008-06-18
DE602006019455D1 (en) 2011-02-17
EP1931782B1 (en) 2011-01-05
CA2620856A1 (en) 2007-03-08
US9994842B2 (en) 2018-06-12
EP2338991B1 (en) 2017-01-18
US8969314B2 (en) 2015-03-03
EP1931782B2 (en) 2016-04-20
EP2338991A2 (en) 2011-06-29
WO2007027775A3 (en) 2007-08-02
JP2009506124A (en) 2009-02-12
US20150232841A1 (en) 2015-08-20
US20070049547A1 (en) 2007-03-01
CA2620856C (en) 2017-11-28
JP5523705B2 (en) 2014-06-18
EP2338991A3 (en) 2011-09-07
AU2006284855B2 (en) 2011-10-13
AU2006284855A1 (en) 2007-03-08

Similar Documents

Publication Publication Date Title
US9994842B2 (en) Methods for use in modulating miR-122A
EP1931780B1 (en) Antisense compounds having enhanced anti-microrna activity
US9598693B2 (en) Oligomeric compounds and compositions for the use in modulation of micrornas
US8466120B2 (en) Oligomeric compounds and compositions for use in modulation of pri-miRNAs
EP3950006A1 (en) Compound, method and pharmaceutical composition for dux4 expression adjustment
WO2007137301A2 (en) Modulation of chrebp expression
JP2022055361A (en) Pharmaceutical compositions for modulating dux4 expression
AU2011254085B2 (en) Methods for use in modulating miR-122a

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 2620856

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2008529232

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2006284855

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006813949

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2006284855

Country of ref document: AU

Date of ref document: 20060829

Kind code of ref document: A

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

Ref document number: 06813949

Country of ref document: EP

Kind code of ref document: A2