CA2921514A1 - Compositions and methods for modulating apolipoprotein c-iii expression - Google Patents

Compositions and methods for modulating apolipoprotein c-iii expression Download PDF

Info

Publication number
CA2921514A1
CA2921514A1 CA2921514A CA2921514A CA2921514A1 CA 2921514 A1 CA2921514 A1 CA 2921514A1 CA 2921514 A CA2921514 A CA 2921514A CA 2921514 A CA2921514 A CA 2921514A CA 2921514 A1 CA2921514 A1 CA 2921514A1
Authority
CA
Canada
Prior art keywords
compound
certain embodiments
modified oligonucleotide
group
conjugate
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA2921514A
Other languages
French (fr)
Other versions
CA2921514C (en
Inventor
Thazha P. Prakash
Punit P. Seth
Eric E. Swayze
Mark J. Graham
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ionis Pharmaceuticals Inc
Original Assignee
Ionis Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=51843959&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2921514(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Ionis Pharmaceuticals Inc filed Critical Ionis Pharmaceuticals Inc
Publication of CA2921514A1 publication Critical patent/CA2921514A1/en
Application granted granted Critical
Publication of CA2921514C publication Critical patent/CA2921514C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • A61K31/7125Nucleic acids or oligonucleotides having modified internucleoside linkage, i.e. other than 3'-5' phosphodiesters
    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • 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
    • A61K47/56Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal 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 macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/03Phosphoric monoester hydrolases (3.1.3)
    • C12Y301/03048Protein-tyrosine-phosphatase (3.1.3.48)
    • 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
    • C12N2310/113Antisense 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
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • 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
    • C12N2310/17Immunomodulatory nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • 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
    • C12N2310/3222'-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
    • 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
    • C12N2310/346Spatial 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3511Conjugate intercalating or cleaving agent
    • 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
    • C12N2310/3513Protein; Peptide
    • 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
    • C12N2310/3515Lipophilic moiety, e.g. cholesterol
    • 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
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • C12N2310/3525MOE, methoxyethoxy
    • 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
    • C12N2310/353Nature of the modification linked to the nucleic acid via an atom other than carbon
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Neurosurgery (AREA)
  • Virology (AREA)
  • Neurology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Saccharide Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Polysaccharides And Polysaccharide Derivatives (AREA)
  • Peptides Or Proteins (AREA)
  • Steroid Compounds (AREA)
  • Liquid Crystal Substances (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Provided herein are oligomeric compounds with conjugate groups targeting apoplipoprotein C-III (ApoClll). In certain embodiments, the ApoClll targeting oligomeric compounds are conjugated to N-Acetylgalactosamine. Also disclosed herein are conjugated oligomeric compounds targeting ApoClll for use in decreasing ApoClll to treat, prevent, or ameliorate diseases, disorders or conditions related to ApoClll. Certain diseases, disorders or conditions related to ApoClll include inflammatory, cardiovascular and/or metabolic diseases, disorders or conditions. The conjugated oligomeric compounds disclosed herein can be used to treat such diseases, disorders or conditions in an individual in need thereof.

Description

COMPOSITIONS AND METHODS FOR MODULATING APOLIPOPROTEIN C-III
EXPRESSION
SEQUENCE LISTING
The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled BIOL0249WOSEQ_ST25.txt, created on May 1, 2014, which is 68 Kb in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.
BACKGROUND OF THE INVENTION
The principle behind antisense technology is that an antisense compound hybridizes to a target nucleic acid and modulates the amount, activity, and/or function of the target nucleic acid. For example in certain instances, antisense compounds result in altered transcription or translation of a target. Such modulation of expression can be achieved by, for example, target mRNA
degradation or occupancy-based inhibition. An example of modulation of RNA target function by degradation is RNase H-based degradation of the target RNA upon hybridization with a DNA-like antisense compound.
Another example of modulation of gene expression by target degradation is RNA interference (RNAi). RNAi refers to antisense-mediated gene silencing through a mechanism that utilizes the RNA-induced siliencing complex (RISC). An additional example of modulation of RNA target function is by an occupancy-based mechanism such as is employed naturally by microRNA. MicroRNAs are small non-coding RNAs that regulate the expression of protein-coding RNAs. The binding of an antisense compound to a microRNA prevents that microRNA from binding to its messenger RNA targets, and thus interferes with the function of the microRNA. MicroRNA mimics can enhance native microRNA function. Certain antisense compounds alter splicing of pre-mRNA.
Regardless of the specific mechanism, sequence-specificity makes antisense compounds attractive as tools for target validation and gene functionalization, as well as therapeutics to selectively modulate the expression of genes involved in the pathogenesis of diseases.
Antisense technology is an effective means for modulating the expression of one or more specific gene products and can therefore prove to be uniquely useful in a number of therapeutic, diagnostic, and research applications. Chemically modified nucleosides may be incorporated into antisense compounds to enhance one or more properties, such as nuclease resistance, pharmacokinetics or affinity for a target nucleic acid. In 1998, the antisense compound, Vitravene0 (fomivirsen; developed by Isis Pharmaceuticals Inc., Carlsbad, CA) was the first antisense drug to achieve marketing clearance from the U.S. Food and Drug Administration (FDA), and is currently a treatment of cytomegalovirus (CMV)-induced retinitis in AIDS
patients.

New chemical modifications have improved the potency and efficacy of antisense compounds, uncovering the potential for oral delivery as well as enhancing subcutaneous administration, decreasing potential for side effects, and leading to improvements in patient convenience. Chemical modifications increasing potency of antisense compounds allow administration of lower doses, which reduces the potential for toxicity, as well as decreasing overall cost of therapy. Modifications increasing the resistance to degradation result in slower clearance from the body, allowing for less frequent dosing. Different types of chemical modifications can be combined in one compound to further optimize the compound's efficacy.
Apolipoprotein C-III (also called APOC3, APOC-III, ApoCIII, and APO C-III) is a constituent of HDL and of triglyceride (TG)-rich lipoproteins. Elevated ApoCIII levels are associated with elevated TG
levels and diseases such as cardiovascular disease, metabolic syndrome, obesity and diabetes (Chan et al., Int J Clin Pract, 2008, 62:799-809; Onat et at., Atherosclerosis, 2003, 168:81-89;
Mendivil et al., Circulation, 2011, 124:2065-2072; Mauger et al., J. Lipid Res, 2006. 47: 1212-1218; Chan et al., Clin. Chem, 2002. 278-283; Ooi et al., Clin. Sci, 2008. 114: 611-624; Davidsson et al., J. Lipid Res. 2005. 46: 1999-2006; Sacks et al., Circulation, 2000. 102: 1886-1892; Lee et al., Arterioscler Thromb Vasc Biol, 2003. 23: 853-858).
ApoCIII slows clearance of TG-rich lipoproteins by inhibiting lipolysis through inhibition of lipoprotein lipase (LPL) and through interfering with lipoprotein binding to cell-surface glycosaminoglycan matrix (Shachter, Curr. Opin. Lipidol, 2001, 12, 297-304).
Antisense technology is emerging as an effective means for reducing the expression of certain gene products and may prove to be uniquely useful in a number of therapeutic, diagnostic, and research applications for the modulation of ApoCIII. Antisense compounds targeting ApoCIII and associated methods for inhibiting ApoCIII have been previously disclosed (see e.g., U.S. Patent 7,598,227, U.S. Patent 7,750,141, PCT publication WO 2004/093783, PCT publication WO 2012/149495 and PCT/U514/016546, all incorporated-by-reference herein). An antisense compound targeting ApoCIII, ISIS-APOCIIIR,õ has been tested in a Phase I and II clinical trials. However, no antisense compounds targeting ApoCIII have been approved for commercial use, accordingly, there is still a need to provide patients with additional and more potent treatment options.
SUMMARY OF THE INVENTION
In certain embodiments, the present disclosure provides conjugated antisense compounds. In certain embodiments, the present disclosure provides conjugated antisense compounds comprising an antisense oligonucleotide complementary to a nucleic acid transcript. In certain embodiments, the present disclosure provides methods comprising contacting a cell with a conjugated antisense compound comprising an antisense oligonucleotide complementary to a nucleic acid transcript. In certain embodiments, the present
2 disclosure provides methods comprising contacting a cell with a conjugated antisense compound comprising an antisense oligonucleotide and reducing the amount or activity of a nucleic acid transcript in a cell.
The asialoglycoprotein receptor (ASGP-R) has been described previously. See e.g., Park et al., PNAS vol. 102, No. 47, pp 17125-17129 (2005). Such receptors are expressed on liver cells, particularly hepatocytes. Further, it has been shown that compounds comprising clusters of three N-acetylgalactosamine (GalNAc) ligands are capable of binding to the ASGP-R, resulting in uptake of the compound into the cell. See e.g., Khorev et al., Bioorganic and Medicinal Chemistry, 16, 9, pp 5216-5231 (May 2008). Accordingly, conjugates comprising such GalNAc clusters have been used to facilitate uptake of certain compounds into liver cells, specifically hepatocytes. For example it has been shown that certain GalNAc-containing conjugates increase activity of duplex siRNA compounds in liver cells in vivo. In such instances, the GalNAc-containing conjugate is typically attached to the sense strand of the siRNA duplex.
Since the sense strand is discarded before the antisense strand ultimately hybridizes with the target nucleic acid, there is little concern that the conjugate will interfere with activity.
Typically, the conjugate is attached to the 3' end of the sense strand of the siRNA. See e.g., U.S. Patent 8,106,022. Certain conjugate groups described herein are more active and/or easier to synthesize than conjugate groups previously described.
In certain embodiments of the present invention, conjugates are attached to single-stranded antisense compounds, including, but not limited to RNase H based antisense compounds and antisense compounds that alter splicing of a pre-mRNA target nucleic acid. In such embodiments, the conjugate should remain attached to the antisense compound long enough to provide benefit (improved uptake into cells) but then should either be cleaved, or otherwise not interfere with the subsequent steps necessary for activity, such as hybridization to a target nucleic acid and interaction with RNase H or enzymes associated with splicing or splice modulation. This balance of properties is more important in the setting of single-stranded antisense compounds than in siRNA compounds, where the conjugate may simply be attached to the sense strand.
Disclosed herein are conjugated single-stranded antisense compounds having improved potency in liver cells in vivo compared with the same antisense compound lacking the conjugate. Given the required balance of properties for these compounds such improved potency is surprising.
In certain embodiments, conjugate groups herein comprise a cleavable moiety.
As noted, without wishing to be bound by mechanism, it is logical that the conjugate should remain on the compound long enough to provide enhancement in uptake, but after that, it is desirable for some portion or, ideally, all of the conjugate to be cleaved, releasing the parent compound (e.g., antisense compound) in its most active form. In certain embodiments, the cleavable moiety is a cleavable nucleoside. Such embodiments take advantage of endogenous nucleases in the cell by attaching the rest of the conjugate (the cluster) to the antisense oligonucleotide through a nucleoside via one or more cleavable bonds, such as those of a phosphodiester linkage. In certain embodiments, the cluster is bound to the cleavable nucleoside through a phosphodiester linkage. In certain embodiments, the cleavable nucleoside is attached to the antisense oligonucleotide (antisense compound) by a phosphodiester linkage. In certain embodiments, the conjugate group may
3 comprise two or three cleavable nucleosides. In such embodiments, such cleavable nucleosides are linked to one another, to the antisense compound and/or to the cluster via cleavable bonds (such as those of a phosphodiester linkage). Certain conjugates herein do not comprise a cleavable nucleoside and instead comprise a cleavable bond. It is shown that that sufficient cleavage of the conjugate from the oligonucleotide is provided by at least one bond that is vulnerable to cleavage in the cell (a cleavable bond).
In certain embodiments, conjugated antisense compounds are prodrugs. Such prodrugs are administered to an animal and are ultimately metabolized to a more active form. For example, conjugated antisense compounds are cleaved to remove all or part of the conjugate resulting in the active (or more active) form of the antisense compound lacking all or some of the conjugate.
In certain embodiments, conjugates are attached at the 5' end of an oligonucleotide. Certain such 5'-conjugates are cleaved more efficiently than counterparts having a similar conjugate group attached at the 3' end. In certain embodiments, improved activity may correlate with improved cleavage. In certain embodiments, oligonucleotides comprising a conjugate at the 5' end have greater efficacy than oligonucleotides comprising a conjugate at the 3' end (see, for example, Examples 56, 81, 83, and 84).
Further, 5'-attachment allows simpler oligonucleotide synthesis. Typically, oligonucleotides are synthesized on a solid support in the 3' to 5' direction. To make a 3'-conjugated oligonucleotide, typically one attaches a pre-conjugated 3' nucleoside to the solid support and then builds the oligonucleotide as usual. However, attaching that conjugated nucleoside to the solid support adds complication to the synthesis. Further, using that approach, the conjugate is then present throughout the synthesis of the oligonucleotide and can become degraded during subsequent steps or may limit the sorts of reactions and reagents that can be used. Using the structures and techniques described herein for 5'-conjugated oligonucleotides, one can synthesize the oligonucleotide using standard automated techniques and introduce the conjugate with the final (5'-most) nucleoside or after the oligonucleotide has been cleaved from the solid support.
In view of the art and the present disclosure, one of ordinary skill can easily make any of the conjugates and conjugated oligonucleotides herein. Moreover, synthesis of certain such conjugates and conjugated oligonucleotides disclosed herein is easier and/or requires few steps, and is therefore less expensive than that of conjugates previously disclosed, providing advantages in manufacturing. For example, the synthesis of certain conjugate groups consists of fewer synthetic steps, resulting in increased yield, relative to conjugate groups previously described. Conjugate groups such as Ga1NAc3-10 in Example 46 and Ga1NAc3-7 in Example 48 are much simpler than previously described conjugates such as those described in U.S. 8,106,022 or U.S. 7,262,177 that require assembly of more chemical intermediates. Accordingly, these and other conjugates described herein have advantages over previously described compounds for use with any oligonucleotide, including single-stranded oligonucleotides and either strand of double-stranded oligonucleotides (e.g., siRNA).
Similarly, disclosed herein are conjugate groups having only one or two GalNAc ligands. As shown, such conjugates groups improve activity of antisense compounds. Such compounds are much easier to
4 prepare than conjugates comprising three GalNAc ligands. Conjugate groups comprising one or two GalNAc ligands may be attached to any antisense compounds, including single-stranded oligonucleotides and either strand of double-stranded oligonucleotides (e.g., siRNA).
In certain embodiments, the conjugates herein do not substantially alter certain measures of tolerability. For example, it is shown herein that conjugated antisense compounds are not more immunogenic than unconjugated parent compounds. Since potency is improved, embodiments in which tolerability remains the same (or indeed even if tolerability worsens only slightly compared to the gains in potency) have improved properties for therapy.
In certain embodiments, conjugation allows one to alter antisense compounds in ways that have less attractive consequences in the absence of conjugation. For example, in certain embodiments, replacing one or more phosphorothioate linkages of a fully phosphorothioate antisense compound with phosphodiester linkages results in improvement in some measures of tolerability. For example, in certain instances, such antisense compounds having one or more phosphodiester are less immunogenic than the same compound in which each linkage is a phosphorothioate. However, in certain instances, as shown in Example 26, that same replacement of one or more phosphorothioate linkages with phosphodiester linkages also results in reduced cellular uptake and/or loss in potency. In certain embodiments, conjugated antisense compounds described herein tolerate such change in linkages with little or no loss in uptake and potency when compared to the conjugated full-phosphorothioate counterpart. In fact, in certain embodiments, for example, in Examples 44, 57, 59, and 86, oligonucleotides comprising a conjugate and at least one phosphodiester internucleoside linkage actually exhibit increased potency in vivo even relative to a full phosphorothioate counterpart also comprising the same conjugate. Moreover, since conjugation results in substantial increases in uptake/potency a small loss in that substantial gain may be acceptable to achieve improved tolerability.
Accordingly, in certain embodiments, conjugated antisense compounds comprise at least one phosphodiester linkage.
In certain embodiments, conjugation of antisense compounds herein results in increased delivery, uptake and activity in hepatocytes. Thus, more compound is delivered to liver tissue. However, in certain embodiments, that increased delivery alone does not explain the entire increase in activity. In certain such embodiments, more compound enters hepatocytes. In certain embodiments, even that increased hepatocyte uptake does not explain the entire increase in activity. In such embodiments, productive uptake of the conjugated compound is increased. For example, as shown in Example 102, certain embodiments of GalNAc-containing conjugates increase enrichment of antisense oligonucleotides in hepatocytes versus non-parenchymal cells. This enrichment is beneficial for oligonucleotides that target genes that are expressed in hepatocytes.
In certain embodiments, conjugated antisense compounds herein result in reduced kidney exposure.
For example, as shown in Example 20, the concentrations of antisense oligonucleotides comprising certain embodiments of GalNAc-containing conjugates are lower in the kidney than that of antisense
5
6 oligonucleotides lacking a GalNAc-containing conjugate.
This has several beneficial therapeutic implications. For therapeutic indications where activity in the kidney is not sought, exposure to kidney risks kidney toxicity without corresponding benefit. Moreover, high concentration in kidney typically results in loss of compound to the urine resulting in faster clearance. Accordingly for non-kidney targets, kidney accumulation is undesired.
In certain embodiments, the present disclosure provides conjugated antisense compounds represented by the formula:
A¨B¨C¨DiE¨) q wherein A is the antisense oligonucleotide;
B is the cleavable moiety C is the conjugate linker D is the branching group each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In the above diagram and in similar diagrams herein, the branching group "D"
branches as many times as is necessary to accommodate the number of (E-F) groups as indicated by "q". Thus, where q = 1, the formula is:
A ¨B¨C¨D¨E¨F
where q = 2, the formula is:
E¨F
A ¨B¨C¨D/
\ E¨F
where q = 3, the formula is:
E¨F
E¨F
A ¨B¨C¨/D¨ E¨F
\

where q = 4, the formula is:
E¨F
E¨F
A ¨B¨C¨D
E¨F
E¨F
where q = 5, the formula is:
E¨F
E¨F
A ¨B ¨C ___________________________ E¨F
E¨F
E¨F
In certain embodiments, conjugated antisense compounds are provided having the structure:
TargeUng moiety ASO
HO OH
HO
OH ¨ 0=P-OH

ON N
NHAc _ N _____________________________________________________________ NO N oP=0 HO OH

NHAc0 0 _ 0 Linker Cleavable moiety Ligand Tether , I , ¨
OH
HO HN

Branching group NHAc
7 In certain embodiments, conjugated antisense compounds are provided having the structure:
Cell targeting moiety HO OH

HO--4)õ\0______ ,iL
Cleavable moiety AcHN 0 1 0¨

OH
HO OH _ ______________________ K,I\T__(NE12 0 0-, 0 0 \ 4 liN
___.rØ....\yr) 1 1 N
HO ,-,-,./N -P., ,o, ---õ,_,--C1 i'-(Y.4%

0 1 o 6-AcHN OH - (:) 0 Tether, -04=0 Ligand HO OH 9, y P- ASO

HO OH
NHAc Branching group
8 In certain embodiments, conjugated antisense compounds are provided having the structure:
ASO
Cleavable moiety HO¨P=0 ____________________________________________________________________ 0 CN¨rµl\T

HO¨P=0 Cell targeting moiety OH
o-AcHN 0 0 (03 HOOH _ ________ Conjugate linker 0-131 =0 AcHN _ o' - OH
Tether Ligand 0 HO y OH A
P, HO
NHAc Branching group In certain embodiments, conjugated antisense compounds are provided having the structure:
9 ASO
I
Ligand _ 0 _ Tether Cleavable moiety HO-=O
HO OH _ Hi -HO---12.-\, 4 Nlro ¨
_ -- 0 AcHN 0 \ (6 NH

.._.....rØ...\, H H )3 HO C)N
4 Ir(-)NI _______________________________________ N ______ AcHN 0 - 0 -Conjugate HO-"'/
_4 linker , H

AcHN 0 _ _ Branching group Cell targeting moiety The present disclosure provides the following non-limiting embodiments:
In embodiments having more than one of a particular variable (e.g., more than one "m" or "n"), unless otherwise indicated, each such particular variable is selected independently. Thus, for a structure having more than one n, each n is selected independently, so they may or may not be the same as one another.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides. In certain embodiments, the modified oligonucleotide with the conjugate group consists of 20 linked nucleosides.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and comprises a nucleobase sequence complementary to an equal length portion of nucleobases 3533 to 3552 of SEQ ID NO: 3, wherein the nucleobase sequence of the modified oligonucleotide is at least 80%
complementary to SEQ ID NO: 3.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and comprises a nucleobase sequence complementary to an equal length portion of nucleobases 3514 to 3558 of SEQ ID NO: 3, wherein the nucleobase sequence of the modified oligonucleotide is at least 80%
complementary to SEQ ID NO: 3.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and has a nucleobase sequence of any of the nucleobase sequences of SEQ ID NOs: 19-96, 209-221. In certain embodiments, the conjugated modified oligonucleotide has a nucleobase sequence comprising at least 8 contiguous nucleobases of any one of the nucleobase sequences of SEQ ID NOs:
19-96, 209-221. In certain embodiments, the compound consists of any one of SEQ ID NOs: 19-96, 209-221 and a conjugate group.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and has a nucleobase sequence of SEQ ID NO: 87. In certain embodiments, the modified oligonucleotide with the conjugate group has a nucleobase sequence comprising at least 8 contiguous nucleobases of the nucleobase sequence of SEQ ID NO: 87. In certain embodiments, the compound consists of SEQ ID NO: 87 and a conjugate group.
In certain embodiments, the present disclosure provides conjugated antisense compounds represented by the following structure. In certain embodiments, the antisense compound comprises the modified oligonucleotide ISIS 304801 with a 5'-X, wherein X is a conjugate group comprising GalNAc. In certain embodiments, the antisense compound consists of the modified oligonucleotide ISIS 304801 with a 5'-X, wherein X is a conjugate group comprising GalNAc.

N2();NI 0 NH2 )i I
0 N N 111....r At-IL
);_Dy 0 N"....0 co_y co_l 0 S-P=0 N
o\ N N NH2 9 S 0 0 I 1)51.1 0 -P=0 NXII....NH 0 S-P=0 0 C:, N al---V24/

0 0) NH2 0 e 9 I 0 0) S-P.0 e 1 Atit S-P=0 ....111..NH 0 0 al e 1 S-P=0 O
eLIH
6o c_o_VN 0 0 C)) 0 0 NH2 cliN 0 9S-P=0 S -P=0 l I ....iit'NH
8 I C) oI, leo 0) s-=o eL.=NH
¨Yo c/
_ON 0 O\NH2 N 0 I C)/) 0 e 1 co4/o o S-P=0 al N
0) NH2 (S)c_c31 0 0 0 NH2 S1=0 I
0 CI) 0) NH2 e 1 S -P=0NIA 0 N N
S-P=0 O I "
N N, cly o O A)), \
\
cc3/
0) o o 0 51.1 21.) S-P=0 9 O ilLNH
S-PO=0 N0 6\c_oiN N NH2 c_O_I

OH 0õ) e e 1 o S-P=0 S-P=0 O ___________ O __________________________________________ In certain embodiments, the present disclosure provides conjugated antisense compounds represented by the following structure. In certain embodiments, the antisense compound comprises the conjugated modified oligonucleotide ISIS 678354. In certain embodiments, the antisense compound consists of the conjugated modified oligonucleotide ISIS 678354.

90 NI---)"---N
HO OH 0 ,y0-F' =0 I ,) Al FINIcr, 0 N N NO
HO--.72-- -.---Tri 1-- 0 1_0_i _ -- __ \_0_)/
---,ir NH ' CV 0 HO OH 0 o (:) 1 NH2 S-P=0 S-p -o Oz_3 <iqNljt:N --'-'1-HNH2 e 9 'IN
- I ,...
4 H OS/,N0 -iNH OV

HO 0--- HO OH o O 0--- 9 i NH2 Nj----1,,N
e 1 S-F'=0 I

4 H ''CLHI 0 N N
W

NH
0 )_0_/0 o 9 0 9 ol S-op=0i 0 llINZ
----1_01 N NH2 , o es4-;)=O Aftt o o,i o o NO
, 1[I'NH
0 Ic_0_)/ 0 , 0 o 9 iLLyH
NH2 S-F'=0 NN) -eS-(1-,)=0 o,) NO

S-P=0 1[1-NH
P
iS-F'=0 8 tt' NH NO
(:)_)/ o,, o o (:)-) o 0 o SOi =

S-P =0 A)LI y H
o --'0 N--'0 '-_Oj 0 CDS-CP=0 (11\11-11'ON---11FINH2 0'-CD

o 9 Nx-L,N

)_5 0 0 o_=

S-P=0 'IlLYFI 0P
0-0"---0 S-F'=0 2.-i 0 o o s-1,=0 OH 0,, J
0 ____________________________________________________________ In certain embodiments, the present disclosure provides conjugated antisense compounds represented by the following structure. In certain embodiments, the antisense compound comprises the conjugated modified oligonucleotide ISIS 678357. In certain embodiments, the antisense compound consists of the conjugated modified oligonucleotide ISIS 678357.

HO OH 0 ,O-F'=0 ., I ;1' irl'' N
_....72.. HN-`< cr, 0N N
0----rN-11--- N--0 1i---,ir NH NO N 0 HO OH 0 N 0 a -9 (:)') NH2 sl' = /(:) (INI:oN':111-FIN H2 -F'=0 '1 r )11 o 4 H --1_5/N NO
---,ir OV

HO OHa 9 o 0--,_.--j NH2 __...72...\ ,-.) 1 S-F'=0 I _j O-F' =0 0 N W-4 H A)'N_k ---,ir NH 0 W

a 9 O 9 O==) 0 S0 0'NH----1_0_1N N NH2 0-P=0 i 0)(Ly _o NH2 o c's1:2 ''t =0 , O 0,) o 0 N 0 (z..., - 0-p =0 IW:r lc_0_1 ., O===/:_o a 9 ilt-yhi NH2 S-F'=0 N-0-9p =0 o,1 N 0 (y ,z) ----) 0 O-P =0 1[1 NH
a 9 Ars-1,zo 8 NH

0)2_/

0 0)0 S-F' =0 '1)1'1 "
a 9 c) o-F1,=o ill'i-1 ll (D)__/N--ND

, _CDj 0 0 OP 1,µ1/11,1,H

S-P N N NH2 =0 ' Nf,k1 S-F' =0 I

N N
)_5 0 o 5P0 YCIF1. a 9 0¨] () /N 0 5P0 'IH

o 9 o S-p =0 OH CO
0 __________________________________________________________________ In certain embodiments, the present disclosure provides conjugated antisense compounds represented by the following structure. In certain embodiments, the antisense compound comprises a modified oligonucleotide with the nucleobase sequence of SEQ ID NO: 87 with a 5'-GalNAc with variability in the sugar mods of the wings. In certain embodiments, the antisense compound consists of a modified oligonucleotide with the nucleobase sequence of SEQ ID NO: 87 with a 5'-GalNAc with variability in the sugar mods of the wings.

, `-'NI----j-=--NL R5N
HO OH 0 00-F'=0 I ,) HN'<' cs, 0 N N
---.Ir NH CV R21 HO OH 0 o o Ri 0 0 0 R51., S-P =0 (---i- N
4 H O NI-IL' NH
() : N N NH2 (/ I S-F' =0 I ,I,L

,Ir NH OZ

HO OH 12 e 0 N
0 R S-F' NH2 f---- N
__....72._v =0 j HO 0-1-n N 0 4 H Z -IR =0 R5rJ.;,,,.. 0 N
N

W
R2 cN 0 0 e 0 (!`111LL'õr RI
0 S-F' =0 Z-P =0 R5,L011,õ 0 O
- - - iN N NH2 0 o R3 R5--C.,N
O R" 0 NO
z_=0 R5I-ILNH
O I j olri2¨/
NO

0. o R3 R5' NH

0 R3 R5,,i,,, S-P =0 ", ----Z-P =0 µõ i 'N 0 ------o L. IR

(D 47 ,/ R3 0 R ' 47¨r, 0 Z-P =0 R51-ji NH
0 0 R- R5I.,,LL, I I
S-F' =0 1 X 0 No R47¨T
Z-I(;=0 R5I---[1'NH
0 0 IR-, R51,11, NH

O N'-'0 0 , _CLy 0 P N

S-P0 l'IL- NH
= I Z-F' =0 I
0.0,N N NH2 N N

R5 o'IR), s-=o OP RVL
SP =O
NH
O_ /N O
I

S-P =0 ) R, O ____________________________________________________________ Wherein either R1 is ¨OCH2CH2OCH3 (M0E)and R2 is H; or R1 and R2 together form a bridge, wherein R1 is ¨0- and R2 is ¨CH2-, -CH(CH3)-, or -CH2CH2-, and R1 and R2 are directly connected such that the resulting bridge is selected from: -0-CH2-, -0-CH(CH3)-, and ¨0-CH2CH2-;
And for each pair of R3 and R4 on the same ring, independently for each ring:
either R3 is selected from H and -OCH2CH2OCH3 and R4 is H; or R3 and R4 together form a bridge, wherein R3 is ¨0-, and R4 is ¨
CH2-, -CH(CH3)-, or -CH2CH2-and R3 and R4 are directly connected such that the resulting bridge is selected from: -0-CH2-, -0-CH(CH3)-, and ¨0-CH2CH2-;
And R5 is selected from H and ¨CH3;

And Z is selected from S- and 0-.
The present disclosure provides the following non-limiting numbered embodiments:
DETAILED DESCRIPTION
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the disclosure. Herein, the use of the singular includes the plural unless specifically stated otherwise. As used herein, the use of "or" means "and/or" unless stated otherwise. Furthermore, the use of the term "including"
as well as other forms, such as "includes" and "included", is not limiting. Also, terms such as "element" or "component" encompass both elements and components comprising one unit and elements and components that comprise more than one subunit, unless specifically stated otherwise.
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including, but not limited to, patents, patent applications, articles, books, and treatises, are hereby expressly incorporated by reference in their entirety for any purpose.
A. Definitions Unless specific definitions are provided, the nomenclature used in connection with, and the procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques may be used for chemical synthesis, and chemical analysis. Certain such techniques and procedures may be found for example in "Carbohydrate Modifications in Antisense Research" Edited by Sangvi and Cook, American Chemical Society , Washington D.C., 1994;
"Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, Pa., 21st edition, 2005; and "Antisense Drug Technology, Principles, Strategies, and Applications" Edited by Stanley T. Crooke, CRC Press, Boca Raton, Florida; and Sambrook et al., "Molecular Cloning, A laboratory Manual," 2nd Edition, Cold Spring Harbor Laboratory Press, 1989, which are hereby incorporated by reference for any purpose. Where permitted, all patents, applications, published applications and other publications and other data referred to throughout in the disclosure are incorporated by reference herein in their entirety.
Unless otherwise indicated, the following terms have the following meanings:
As used herein, "nucleoside" means a compound comprising a nucleobase moiety and a sugar moiety. Nucleosides include, but are not limited to, naturally occurring nucleosides (as found in DNA and RNA) and modified nucleosides. Nucleosides may be linked to a phosphate moiety.
As used herein, "chemical modification" means a chemical difference in a compound when compared to a naturally occurring counterpart. Chemical modifications of oligonucleotides include nucleoside modifications (including sugar moiety modifications and nucleobase modifications) and internucleoside linkage modifications. In reference to an oligonucleotide, chemical modification does not include differences only in nucleobase sequence.
As used herein, "furanosyl" means a structure comprising a 5-membered ring comprising four carbon atoms and one oxygen atom.
As used herein, "naturally occurring sugar moiety" means a ribofuranosyl as found in naturally occurring RNA or a deoxyribofuranosyl as found in naturally occurring DNA.
As used herein, "sugar moiety" means a naturally occurring sugar moiety or a modified sugar moiety of a nucleoside.
As used herein, "modified sugar moiety" means a substituted sugar moiety or a sugar surrogate.
As used herein, "substituted sugar moiety" means a furanosyl that is not a naturally occurring sugar moiety. Substituted sugar moieties include, but are not limited to furanosyls comprising substituents at the 2'-position, the 3'-position, the 5'-position and/or the 4'-position. Certain substituted sugar moieties are bicyclic sugar moieties.
As used herein, "2'-substituted sugar moiety" means a furanosyl comprising a substituent at the 2'-position other than H or OH. Unless otherwise indicated, a 2'-substituted sugar moiety is not a bicyclic sugar moiety (i.e., the 2'-substituent of a 2'-substituted sugar moiety does not form a bridge to another atom of the furanosyl ring.
As used herein, "MOE" means -OCH2CH2OCH3.
As used herein, "2'-F nucleoside" refers to a nucleoside comprising a sugar comprising fluorine at the 2' position. Unless otherwise indicated, the fluorine in a 2'-F nucleoside is in the ribo position (replacing the OH of a natural ribose).
As used herein the term "sugar surrogate" means a structure that does not comprise a furanosyl and that is capable of replacing the naturally occurring sugar moiety of a nucleoside, such that the resulting nucleoside sub-units are capable of linking together and/or linking to other nucleosides to form an oligomeric compound which is capable of hybridizing to a complementary oligomeric compound. Such structures include rings comprising a different number of atoms than furanosyl (e.g., 4, 6, or 7-membered rings);
replacement of the oxygen of a furanosyl with a non-oxygen atom (e.g., carbon, sulfur, or nitrogen); or both a change in the number of atoms and a replacement of the oxygen. Such structures may also comprise substitutions corresponding to those described for substituted sugar moieties (e.g., 6-membered carbocyclic bicyclic sugar surrogates optionally comprising additional substituents).
Sugar surrogates also include more complex sugar replacements (e.g., the non-ring systems of peptide nucleic acid). Sugar surrogates include without limitation morpholinos, cyclohexenyls and cyclohexitols.
As used herein, "bicyclic sugar moiety" means a modified sugar moiety comprising a 4 to 7 membered ring (including but not limited to a furanosyl) comprising a bridge connecting two atoms of the 4 to 7 membered ring to form a second ring, resulting in a bicyclic structure.
In certain embodiments, the 4 to 7 membered ring is a sugar ring. In certain embodiments the 4 to 7 membered ring is a furanosyl. In certain such embodiments, the bridge connects the 2'-carbon and the 4'-carbon of the furanosyl.
As used herein, "nucleotide" means a nucleoside further comprising a phosphate linking group. As used herein, "linked nucleosides" may or may not be linked by phosphate linkages and thus includes, but is not limited to "linked nucleotides." As used herein, "linked nucleosides" are nucleosides that are connected in a continuous sequence (i.e. no additional nucleosides are present between those that are linked).
As used herein, "nucleic acid" refers to molecules composed of monomeric nucleotides. A nucleic acid includes ribonucleic acids (RNA), deoxyribonucleic acids (DNA), single-stranded nucleic acids (ssDNA), double-stranded nucleic acids (dsDNA), small interfering ribonucleic acids (siRNA), and microRNAs (miRNA). A nucleic acid may also comprise any combination of these elements in a single molecule.
As used herein, "nucleotide" means a nucleoside further comprising a phosphate linking group. As used herein, "linked nucleosides" may or may not be linked by phosphate linkages and thus includes, but is not limited to "linked nucleotides." As used herein, "linked nucleosides" are nucleosides that are connected in a continuous sequence (i.e. no additional nucleosides are present between those that are linked).
As used herein, "nucleobase" means a group of atoms that can be linked to a sugar moiety to create a nucleoside that is capable of incorporation into an oligonucleotide, and wherein the group of atoms is capable of bonding with a complementary naturally occurring nucleobase of another oligonucleotide or nucleic acid.
Nucleobases may be naturally occurring or may be modified. As used herein, "nucleobase sequence" means the order of contiguous nucleobases independent of any sugar, linkage, or nucleobase modification.
As used herein the terms, "unmodified nucleobase" or "naturally occurring nucleobase" means the naturally occurring heterocyclic nucleobases of RNA or DNA: the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) (including 5-methyl C), and uracil (U).
As used herein, "modified nucleobase" means any nucleobase that is not a naturally occurring nucleobase.
As used herein, "modified nucleoside" means a nucleoside comprising at least one chemical modification compared to naturally occurring RNA or DNA nucleosides. Modified nucleosides comprise a modified sugar moiety and/or a modified nucleobase.
As used herein, "bicyclic nucleoside" or "BNA" means a nucleoside comprising a bicyclic sugar moiety.
As used herein, "constrained ethyl nucleoside" or "cEt" means a nucleoside comprising a bicyclic sugar moiety comprising a 4'-CH(CH3)-0-2'bridge.
As used herein, "locked nucleic acid nucleoside" or "LNA" means a nucleoside comprising a bicyclic sugar moiety comprising a 4'-CH2-0-2'bridge.
As used herein, "2'-substituted nucleoside" means a nucleoside comprising a substituent at the 2'-position other than H or OH. Unless otherwise indicated, a 2'-substituted nucleoside is not a bicyclic nucleoside.
As used herein, "deoxynucleoside" means a nucleoside comprising 2'-H furanosyl sugar moiety, as found in naturally occurring deoxyribonucleosides (DNA). In certain embodiments, a 2'-deoxynucleoside may comprise a modified nucleobase or may comprise an RNA nucleobase (e.g., uracil).
As used herein, "oligonucleotide" means a compound comprising a plurality of linked nucleosides.
In certain embodiments, an oligonucleotide comprises one or more unmodified ribonucleosides (RNA) and/or unmodified deoxyribonucleosides (DNA) and/or one or more modified nucleosides.
As used herein "oligonucleoside" means an oligonucleotide in which none of the internucleoside linkages contains a phosphorus atom. As used herein, oligonucleotides include oligonucleosides.
As used herein, "modified oligonucleotide" means an oligonucleotide comprising at least one modified nucleoside and/or at least one modified internucleoside linkage.
As used herein, "linkage" or "linking group" means a group of atoms that link together two or more other groups of atoms.
As used herein "internucleoside linkage" means a covalent linkage between adjacent nucleosides in an oligonucleotide.
As used herein "naturally occurring internucleoside linkage" means a 3' to 5' phosphodiester linkage.
As used herein, "modified internucleoside linkage" means any internucleoside linkage other than a naturally occurring internucleoside linkage.
As used herein, "terminal internucleoside linkage" means the linkage between the last two nucleosides of an oligonucleotide or defined region thereof As used herein, "phosphorus linking group" means a linking group comprising a phosphorus atom.
Phosphorus linking groups include without limitation groups having the formula:
vw Ra Rb=P¨R, Rd wherein:
Ra and Rd are each, independently, 0, S, CH2, NH, or NJI wherein J1 is C1-C6 alkyl or substituted CI-C6 alkyl;
Rb is 0 or S;
Re is OH, SH, C1-C6 alkyl, substituted C1-C6 alkyl, C1-C6 alkoxy, substituted C1-C6 alkoxy, amino or substituted amino; and Ji iS Rb iS 0 or S.
Phosphorus linking groups include without limitation, phosphodiester, phosphorothioate, phosphorodithioate, phosphonate, phosphoramidate, phosphorothioamidate, thionoalkylphosphonate, phosphotriesters, thionoalkylphosphotriester and boranophosphate.
As used herein, "internucleoside phosphorus linking group" means a phosphorus linking group that directly links two nucleosides.
As used herein, "non-internucleoside phosphorus linking group" means a phosphorus linking group that does not directly link two nucleosides. In certain embodiments, a non-internucleoside phosphorus linking group links a nucleoside to a group other than a nucleoside. In certain embodiments, a non-internucleoside phosphorus linking group links two groups, neither of which is a nucleoside.
As used herein, "neutral linking group" means a linking group that is not charged. Neutral linking groups include without limitation phosphotriesters, methylphosphonates, MMI (-CH2-N(CH3)-0-), amide-3 (-CH2-C(=0)-N(H)-), amide-4 (-CH2-N(H)-C(=0)-), formacetal (-0-CH2-0-), and thioformacetal (-S-CH2-0-).
Further neutral linking groups include nonionic linkages comprising siloxane (dialkylsiloxane), carboxylate ester, carboxamide, sulfide, sulfonate ester and amides (See for example:
Carbohydrate Modifications in Antisense Research; Y.S. Sanghvi and P.D. Cook Eds. ACS Symposium Series 580;
Chapters 3 and 4, (pp.
40-65)). Further neutral linking groups include nonionic linkages comprising mixed N, 0, S and CH2 component parts.
As used herein, "internucleoside neutral linking group" means a neutral linking group that directly links two nucleosides.
As used herein, "non-internucleoside neutral linking group" means a neutral linking group that does not directly link two nucleosides. In certain embodiments, a non-internucleoside neutral linking group links a nucleoside to a group other than a nucleoside. In certain embodiments, a non-internucleoside neutral linking group links two groups, neither of which is a nucleoside.
As used herein, "oligomeric compound" means a polymeric structure comprising two or more sub-structures. In certain embodiments, an oligomeric compound comprises an oligonucleotide. In certain embodiments, an oligomeric compound comprises one or more conjugate groups and/or terminal groups. In certain embodiments, an oligomeric compound consists of an oligonucleotide.
Oligomeric compounds also include naturally occurring nucleic acids. In certain embodiments, an oligomeric compound comprises a backbone of one or more linked monomeric subunits where each linked monomeric subunit is directly or indirectly attached to a heterocyclic base moiety. In certain embodiments, oligomeric compounds may also include monomeric subunits that are not linked to a heterocyclic base moiety, thereby providing abasic sites.
In certain embodiments, the linkages joining the monomeric subunits, the sugar moieties or surrogates and the heterocyclic base moieties can be independently modified. In certain embodiments, the linkage-sugar unit, which may or may not include a heterocyclic base, may be substituted with a mimetic such as the monomers in peptide nucleic acids.
As used herein, "terminal group" means one or more atom attached to either, or both, the 3' end or the 5' end of an oligonucleotide. In certain embodiments a terminal group is a conjugate group. In certain embodiments, a terminal group comprises one or more terminal group nucleosides.

As used herein, "conjugate" or "conjugate group" means an atom or group of atoms bound to an oligonucleotide or oligomeric compound. In general, conjugate groups modify one or more properties of the compound to which they are attached, including, but not limited to pharmacodynamic, pharmacokinetic, binding, absorption, cellular distribution, cellular uptake, charge and/or clearance properties.
As used herein, "conjugate linker" or "linker" in the context of a conjugate group means a portion of a conjugate group comprising any atom or group of atoms and which covalently link (1) an oligonucleotide to another portion of the conjugate group or (2) two or more portions of the conjugate group.
Conjugate groups are shown herein as radicals, providing a bond for forming covalent attachment to an oligomeric compound such as an antisense oligonucleotide. In certain embodiments, the point of attachment on the oligomeric compound is the 3'-oxygen atom of the 3'-hydroxyl group of the 3' terminal nucleoside of the oligomeric compound. In certain embodiments the point of attachment on the oligomeric compound is the 5'-oxygen atom of the 5'-hydroxyl group of the 5' terminal nucleoside of the oligomeric compound. In certain embodiments, the bond for forming attachment to the oligomeric compound is a cleavable bond. In certain such embodiments, such cleavable bond constitutes all or part of a cleavable moiety.
In certain embodiments, conjugate groups comprise a cleavable moiety (e.g., a cleavable bond or cleavable nucleoside) and a carbohydrate cluster portion, such as a GalNAc cluster portion. Such carbohydrate cluster portion comprises: a targeting moiety and, optionally, a conjugate linker. In certain embodiments, the carbohydrate cluster portion is identified by the number and identity of the ligand. For example, in certain embodiments, the carbohydrate cluster portion comprises 3 GalNAc groups and is designated "Ga1NAc3". In certain embodiments, the carbohydrate cluster portion comprises 4 GalNAc groups and is designated "Ga1NAc4". Specific carbohydrate cluster portions (having specific tether, branching and conjugate linker groups) are described herein and designated by Roman numeral followed by subscript "a". Accordingly "GalNac3-1,7 refers to a specific carbohydrate cluster portion of a conjugate group having 3 GalNac groups and specifically identified tether, branching and linking groups. Such carbohydrate cluster fragment is attached to an oligomeric compound via a cleavable moiety, such as a cleavable bond or cleavable nucleoside.
As used herein, "cleavable moiety" means a bond or group that is capable of being split under physiological conditions. In certain embodiments, a cleavable moiety is cleaved inside a cell or sub-cellular compartments, such as a lysosome. In certain embodiments, a cleavable moiety is cleaved by endogenous enzymes, such as nucleases. In certain embodiments, a cleavable moiety comprises a group of atoms having one, two, three, four, or more than four cleavable bonds.
As used herein, "cleavable bond" means any chemical bond capable of being split. In certain embodiments, a cleavable bond is selected from among: an amide, a polyamide, an ester, an ether, one or both esters of a phosphodiester, a phosphate ester, a carbamate, a di-sulfide, or a peptide.

As used herein, "carbohydrate cluster" means a compound having one or more carbohydrate residues attached to a scaffold or linker group. (see, e.g., Maier et al., "Synthesis of Antisense Oligonucleotides Conjugated to a Multivalent Carbohydrate Cluster for Cellular Targeting,"
Bioconjugate Chemistry, 2003, (14): 18-29, which is incorporated herein by reference in its entirety, or Rensen et al., "Design and Synthesis of Novel N-Acetylgalactosamine-Terminated Glycolipids for Targeting of Lipoproteins to the Hepatic Asiaglycoprotein Receptor," J. Med. Chem. 2004, (47): 5798-5808, for examples of carbohydrate conjugate clusters).
As used herein, "modified carbohydrate" means any carbohydrate having one or more chemical modifications relative to naturally occurring carbohydrates.
As used herein, "carbohydrate derivative" means any compound which may be synthesized using a carbohydrate as a starting material or intermediate.
As used herein, "carbohydrate" means a naturally occurring carbohydrate, a modified carbohydrate, or a carbohydrate derivative.
As used herein "protecting group" means any compound or protecting group known to those having skill in the art. Non-limiting examples of protecting groups may be found in "Protective Groups in Organic Chemistry", T. W. Greene, P. G. M. Wuts, ISBN 0-471-62301-6, John Wiley &
Sons, Inc, New York, which is incorporated herein by reference in its entirety.
As used herein, "single-stranded" means an oligomeric compound that is not hybridized to its complement and which lacks sufficient self-complementarity to form a stable self-duplex.
As used herein, "double stranded" means a pair of oligomeric compounds that are hybridized to one another or a single self-complementary oligomeric compound that forms a hairpin structure. In certain embodiments, a double-stranded oligomeric compound comprises a first and a second oligomeric compound.
As used herein, "antisense compound" means a compound comprising or consisting of an oligonucleotide at least a portion of which is complementary to a target nucleic acid to which it is capable of hybridizing, resulting in at least one antisense activity.
As used herein, "antisense activity" means any detectable and/or measurable change attributable to the hybridization of an antisense compound to its target nucleic acid. In certain embodiments, antisense activity includes modulation of the amount or activity of a target nucleic acid transcript (e.g. mRNA). In certain embodiments, antisense activity includes modulation of the splicing of pre-mRNA.
As used herein, "RNase H based antisense compound" means an antisense compound wherein at least some of the antisense activity of the antisense compound is attributable to hybridization of the antisense compound to a target nucleic acid and subsequent cleavage of the target nucleic acid by RNase H.
As used herein, "RISC based antisense compound" means an antisense compound wherein at least some of the antisense activity of the antisense compound is attributable to the RNA Induced Silencing Complex (RISC).
As used herein, "detecting" or "measuring" means that a test or assay for detecting or measuring is performed. Such detection and/or measuring may result in a value of zero.
Thus, if a test for detection or measuring results in a finding of no activity (activity of zero), the step of detecting or measuring the activity has nevertheless been performed.
As used herein, "detectable and/or measureable activity" means a statistically significant activity that is not zero.
As used herein, "essentially unchanged" means little or no change in a particular parameter, particularly relative to another parameter which changes much more. In certain embodiments, a parameter is essentially unchanged when it changes less than 5%. In certain embodiments, a parameter is essentially unchanged if it changes less than two-fold while another parameter changes at least ten-fold. For example, in certain embodiments, an antisense activity is a change in the amount of a target nucleic acid. In certain such embodiments, the amount of a non-target nucleic acid is essentially unchanged if it changes much less than the target nucleic acid does, but the change need not be zero.
As used herein, "expression" means the process by which a gene ultimately results in a protein.
Expression includes, but is not limited to, transcription, post-transcriptional modification (e.g., splicing, polyadenlyation, addition of 5'-cap), and translation.
As used herein, "target nucleic acid" means a nucleic acid molecule to which an antisense compound is intended to hybridize to result in a desired antisense activity. Antisense oligonucleotides have sufficient complementarity to their target nucleic acids to allow hybridization under physiological conditions.
As used herein, "nucleobase complementarity" or "complementarity" when in reference to nucleobases means a nucleobase that is capable of base pairing with another nucleobase. For example, in DNA, adenine (A) is complementary to thymine (T). For example, in RNA, adenine (A) is complementary to uracil (U). In certain embodiments, complementary nucleobase means a nucleobase of an antisense compound that is capable of base pairing with a nucleobase of its target nucleic acid. For example, if a nucleobase at a certain position of an antisense compound is capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid, then the position of hydrogen bonding between the oligonucleotide and the target nucleic acid is considered to be complementary at that nucleobase pair.
Nucleobases comprising certain modifications may maintain the ability to pair with a counterpart nucleobase and thus, are still capable of nucleobase complementarity.
As used herein, "non-complementary" in reference to nucleobases means a pair of nucleobases that do not form hydrogen bonds with one another.
As used herein, "complementary" in reference to oligomeric compounds (e.g., linked nucleosides, oligonucleotides, or nucleic acids) means the capacity of such oligomeric compounds or regions thereof to hybridize to another oligomeric compound or region thereof through nucleobase complementarity.
Complementary oligomeric compounds need not have nucleobase complementarity at each nucleoside.
Rather, some mismatches are tolerated. In certain embodiments, complementary oligomeric compounds or regions are complementary at 70% of the nucleobases (70% complementary). In certain embodiments, complementary oligomeric compounds or regions are 80% complementary. In certain embodiments, complementary oligomeric compounds or regions are 90% complementary. In certain embodiments, complementary oligomeric compounds or regions are 95% complementary. In certain embodiments, complementary oligomeric compounds or regions are 100% complementary.
As used herein, "mismatch" means a nucleobase of a first oligomeric compound that is not capable of pairing with a nucleobase at a corresponding position of a second oligomeric compound, when the first and second oligomeric compound are aligned. Either or both of the first and second oligomeric compounds may be oligonucleotides.
As used herein, "hybridization" means the pairing of complementary oligomeric compounds (e.g., an antisense compound and its target nucleic acid). While not limited to a particular mechanism, the most common mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleobases.
As used herein, "specifically hybridizes" means the ability of an oligomeric compound to hybridize to one nucleic acid site with greater affinity than it hybridizes to another nucleic acid site.
As used herein, "fully complementary" in reference to an oligonucleotide or portion thereof means that each nucleobase of the oligonucleotide or portion thereof is capable of pairing with a nucleobase of a complementary nucleic acid or contiguous portion thereof Thus, a fully complementary region comprises no mismatches or unhybridized nucleobases in either strand.
As used herein, "percent complementarity" means the percentage of nucleobases of an oligomeric compound that are complementary to an equal-length portion of a target nucleic acid. Percent complementarity is calculated by dividing the number of nucleobases of the oligomeric compound that are complementary to nucleobases at corresponding positions in the target nucleic acid by the total length of the oligomeric compound.
As used herein, "percent identity" means the number of nucleobases in a first nucleic acid that are the same type (independent of chemical modification) as nucleobases at corresponding positions in a second nucleic acid, divided by the total number of nucleobases in the first nucleic acid.
As used herein, "modulation" means a change of amount or quality of a molecule, function, or activity when compared to the amount or quality of a molecule, function, or activity prior to modulation. For example, modulation includes the change, either an increase (stimulation or induction) or a decrease (inhibition or reduction) in gene expression. As a further example, modulation of expression can include a change in splice site selection of pre-mRNA processing, resulting in a change in the absolute or relative amount of a particular splice-variant compared to the amount in the absence of modulation.
As used herein, "chemical motif' means a pattern of chemical modifications in an oligonucleotide or a region thereof Motifs may be defined by modifications at certain nucleosides and/or at certain linking groups of an oligonucleotide.

As used herein, "nucleoside motif' means a pattern of nucleoside modifications in an oligonucleotide or a region thereof The linkages of such an oligonucleotide may be modified or unmodified. Unless otherwise indicated, motifs herein describing only nucleosides are intended to be nucleoside motifs. Thus, in such instances, the linkages are not limited.
As used herein, "sugar motif' means a pattern of sugar modifications in an oligonucleotide or a region thereof As used herein, "linkage motif' means a pattern of linkage modifications in an oligonucleotide or region thereof The nucleosides of such an oligonucleotide may be modified or unmodified. Unless otherwise indicated, motifs herein describing only linkages are intended to be linkage motifs. Thus, in such instances, the nucleosides are not limited.
As used herein, "nucleobase modification motif' means a pattern of modifications to nucleobases along an oligonucleotide. Unless otherwise indicated, a nucleobase modification motif is independent of the nucleobase sequence.
As used herein, "sequence motif' means a pattern of nucleobases arranged along an oligonucleotide or portion thereof Unless otherwise indicated, a sequence motif is independent of chemical modifications and thus may have any combination of chemical modifications, including no chemical modifications.
As used herein, "type of modification" in reference to a nucleoside or a nucleoside of a "type" means the chemical modification of a nucleoside and includes modified and unmodified nucleosides. Accordingly, unless otherwise indicated, a "nucleoside having a modification of a first type" may be an unmodified nucleoside.
As used herein, "differently modified" mean chemical modifications or chemical substituents that are different from one another, including absence of modifications. Thus, for example, a MOE nucleoside and an unmodified DNA nucleoside are "differently modified," even though the DNA
nucleoside is unmodified.
Likewise, DNA and RNA are "differently modified," even though both are naturally-occurring unmodified nucleosides. Nucleosides that are the same but for comprising different nucleobases are not differently modified. For example, a nucleoside comprising a 2'-0Me modified sugar and an unmodified adenine nucleobase and a nucleoside comprising a 2'-0Me modified sugar and an unmodified thymine nucleobase are not differently modified.
As used herein, "the same type of modifications" refers to modifications that are the same as one another, including absence of modifications. Thus, for example, two unmodified DNA nucleosides have "the same type of modification," even though the DNA nucleoside is unmodified.
Such nucleosides having the same type modification may comprise different nucleobases.
As used herein, "separate regions" means portions of an oligonucleotide wherein the chemical modifications or the motif of chemical modifications of any neighboring portions include at least one difference to allow the separate regions to be distinguished from one another.
As used herein, "pharmaceutically acceptable carrier or diluent" means any substance suitable for use in administering to an animal. In certain embodiments, a pharmaceutically acceptable carrier or diluent is sterile saline. In certain embodiments, such sterile saline is pharmaceutical grade saline.
As used herein the term "metabolic disorder" means a disease or condition principally characterized by dysregulation of metabolism ¨ the complex set of chemical reactions associated with breakdown of food to produce energy.
As used herein, the term "cardiovascular disorder" means a disease or condition principally characterized by impaired function of the heart or blood vessels.
As used herein the term "mono or polycyclic ring system" is meant to include all ring systems selected from single or polycyclic radical ring systems wherein the rings are fused or linked and is meant to be inclusive of single and mixed ring systems individually selected from aliphatic, alicyclic, aryl, heteroaryl, aralkyl, arylalkyl, heterocyclic, heteroaryl, heteroaromatic and heteroarylalkyl. Such mono and poly cyclic structures can contain rings that each have the same level of saturation or each, independently, have varying degrees of saturation including fully saturated, partially saturated or fully unsaturated. Each ring can comprise ring atoms selected from C, N, 0 and S to give rise to heterocyclic rings as well as rings comprising only C ring atoms which can be present in a mixed motif such as for example benzimidazole wherein one ring has only carbon ring atoms and the fused ring has two nitrogen atoms. The mono or polycyclic ring system can be further substituted with substituent groups such as for example phthalimide which has two =0 groups attached to one of the rings. Mono or polycyclic ring systems can be attached to parent molecules using various strategies such as directly through a ring atom, fused through multiple ring atoms, through a substituent group or through a bifunctional linking moiety.
As used herein, "prodrug" means an inactive or less active form of a compound which, when administered to a subject, is metabolized to form the active, or more active, compound (e.g., drug).
As used herein, "substituent" and "substituent group," means an atom or group that replaces the atom or group of a named parent compound. For example a substituent of a modified nucleoside is any atom or group that differs from the atom or group found in a naturally occurring nucleoside (e.g., a modified 2'-substuent is any atom or group at the 2'-position of a nucleoside other than H
or OH). Substituent groups can be protected or unprotected. In certain embodiments, compounds of the present disclosure have substituents at one or at more than one position of the parent compound. Substituents may also be further substituted with other substituent groups and may be attached directly or via a linking group such as an alkyl or hydrocarbyl group to a parent compound.
Likewise, as used herein, "substituent" in reference to a chemical functional group means an atom or group of atoms that differs from the atom or a group of atoms normally present in the named functional group. In certain embodiments, a substituent replaces a hydrogen atom of the functional group (e.g., in certain embodiments, the substituent of a substituted methyl group is an atom or group other than hydrogen which replaces one of the hydrogen atoms of an unsubstituted methyl group).
Unless otherwise indicated, groups amenable for use as substituents include without limitation, halogen, hydroxyl, alkyl, alkenyl, alkynyl, acyl (-C(0)Raa), carboxyl (-C(0)0-Raa), aliphatic groups, alicyclic groups, alkoxy, substituted oxy (-O-Raa), aryl, aralkyl, heterocyclic radical, heteroaryl, heteroarylalkyl, amino (-N(Rbb)(Ree)), imino(=NRbb), amido (-C(0)N(Rbb)(Ree) or -N(Rbb)C(0)Ra.), azido (-N3), nitro (-NO2), cyano (-CN), carbamido (-0C(0)N(Rbb)(Ree) or -N(Rbb)C(0)0Raa), ureido (-N(Rbb)C(0)N(Rbb)(Rõ)), thioureido (-N(Rbb)C(S)N(Rbb)-(Rõ)), guanidinyl (-N(Rbb)C(=NRbb)N(Rbb)(Ree)), amidinyl (-C(=NRbb)N(Rbb)(Ree) or -N(Rbb)C(=NRbb)(Raa.)), thiol (-SRbb), sulfinyl (-S(0)Rbb), sulfonyl (-S(0)2Rbb) and sulfonamidyl (-S(0)2N(Rbb)(Ree) or -N(Rbb)S-(0)2Rbb). Wherein each Raa, Rbb and Ree is, independently, H, an optionally linked chemical functional group or a further substituent group with a preferred list including without limitation, alkyl, alkenyl, alkynyl, aliphatic, alkoxy, acyl, aryl, aralkyl, heteroaryl, alicyclic, heterocyclic and heteroarylalkyl. Selected substituents within the compounds described herein are present to a recursive degree.
As used herein, "alkyl," as used herein, means a saturated straight or branched hydrocarbon radical containing up to twenty four carbon atoms. Examples of alkyl groups include without limitation, methyl, ethyl, propyl, butyl, isopropyl, n-hexyl, octyl, decyl, dodecyl and the like.
Alkyl groups typically include from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms (Ci-C12alkyl) with from 1 to about 6 carbon atoms being more preferred.
As used herein, "alkenyl," means a straight or branched hydrocarbon chain radical containing up to twenty four carbon atoms and having at least one carbon-carbon double bond.
Examples of alkenyl groups include without limitation, ethenyl, propenyl, butenyl, 1-methy1-2-buten-1-yl, dienes such as 1,3-butadiene and the like. Alkenyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred. Alkenyl groups as used herein may optionally include one or more further substituent groups.
As used herein, "alkynyl," means a straight or branched hydrocarbon radical containing up to twenty four carbon atoms and having at least one carbon-carbon triple bond. Examples of alkynyl groups include, without limitation, ethynyl, 1-propynyl, 1-butynyl, and the like. Alkynyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred. Alkynyl groups as used herein may optionally include one or more further substituent groups.
As used herein, "acyl," means a radical formed by removal of a hydroxyl group from an organic acid and has the general Formula -C(0)-X where X is typically aliphatic, alicyclic or aromatic. Examples include aliphatic carbonyls, aromatic carbonyls, aliphatic sulfonyls, aromatic sulfinyls, aliphatic sulfinyls, aromatic phosphates, aliphatic phosphates and the like. Acyl groups as used herein may optionally include further substituent groups.
As used herein, "alicyclic" means a cyclic ring system wherein the ring is aliphatic. The ring system can comprise one or more rings wherein at least one ring is aliphatic.
Preferred alicyclics include rings having from about 5 to about 9 carbon atoms in the ring. Alicyclic as used herein may optionally include further substituent groups.

As used herein, "aliphatic" means a straight or branched hydrocarbon radical containing up to twenty four carbon atoms wherein the saturation between any two carbon atoms is a single, double or triple bond.
An aliphatic group preferably contains from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms with from 1 to about 6 carbon atoms being more preferred. The straight or branched chain of an aliphatic group may be interrupted with one or more heteroatoms that include nitrogen, oxygen, sulfur and phosphorus. Such aliphatic groups interrupted by heteroatoms include without limitation, polyalkoxys, such as polyalkylene glycols, polyamines, and polyimines. Aliphatic groups as used herein may optionally include further substituent groups.
As used herein, "alkoxy" means a radical formed between an alkyl group and an oxygen atom wherein the oxygen atom is used to attach the alkoxy group to a parent molecule. Examples of alkoxy groups include without limitation, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, neopentoxy, n-hexoxy and the like. Alkoxy groups as used herein may optionally include further substituent groups.
As used herein, "aminoalkyl" means an amino substituted C1-C12 alkyl radical.
The alkyl portion of the radical forms a covalent bond with a parent molecule. The amino group can be located at any position and the aminoalkyl group can be substituted with a further substituent group at the alkyl and/or amino portions.
As used herein, "aralkyl" and "arylalkyl" mean an aromatic group that is covalently linked to a C1-C12 alkyl radical. The alkyl radical portion of the resulting aralkyl (or arylalkyl) group forms a covalent bond with a parent molecule. Examples include without limitation, benzyl, phenethyl and the like. Aralkyl groups as used herein may optionally include further substituent groups attached to the alkyl, the aryl or both groups that form the radical group.
As used herein, "aryl" and "aromatic" mean a mono- or polycyclic carbocyclic ring system radicals having one or more aromatic rings. Examples of aryl groups include without limitation, phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like. Preferred aryl ring systems have from about 5 to about 20 carbon atoms in one or more rings. Aryl groups as used herein may optionally include further substituent groups.
As used herein, "halo" and "halogen," mean an atom selected from fluorine, chlorine, bromine and iodine.
As used herein, "heteroaryl," and "heteroaromatic," mean a radical comprising a mono- or poly-cyclic aromatic ring, ring system or fused ring system wherein at least one of the rings is aromatic and includes one or more heteroatoms. Heteroaryl is also meant to include fused ring systems including systems where one or more of the fused rings contain no heteroatoms. Heteroaryl groups typically include one ring atom selected from sulfur, nitrogen or oxygen. Examples of heteroaryl groups include without limitation, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl and the like. Heteroaryl radicals can be attached to a parent molecule directly or through a linking moiety such as an aliphatic group or hetero atom. Heteroaryl groups as used herein may optionally include further substituent groups.
As used herein, "conjugate compound" means any atoms, group of atoms, or group of linked atoms suitable for use as a conjugate group. In certain embodiments, conjugate compounds may possess or impart one or more properties, including, but not limited to pharmacodynamic, pharmacokinetic, binding, absorption, cellular distribution, cellular uptake, charge and/or clearance properties.
As used herein, unless otherwise indicated or modified, the term "double-stranded" refers to two separate oligomeric compounds that are hybridized to one another. Such double stranded compounds may have one or more or non-hybridizing nucleosides at one or both ends of one or both strands (overhangs) and/or one or more internal non-hybridizing nucleosides (mismatches) provided there is sufficient complementarity to maintain hybridization under physiologically relevant conditions.
As used herein, "5' target site" refers to the nucleotide of a target nucleic acid which is complementary to the 5'-most nucleotide of a particular antisense compound.
As used herein, "about" means within 10% of a value. For example, if it is stated, "a marker may be increased by about 50%", it is implied that the marker may be increased between 45%-55%.
As used herein, "administered concomitantly" refers to the co-administration of two agents in any manner in which the pharmacological effects of both are manifest in the patient at the same time.
Concomitant administration does not require that both agents be administered in a single pharmaceutical composition, in the same dosage form, or by the same route of administration.
The effects of both agents need not manifest themselves at the same time. The effects need only be overlapping for a period of time and need not be coextensive.
As used herein, "administering" or "administration" means providing a pharmaceutical agent to an individual, and includes, but is not limited to, administering by a medical professional and self-administering.
Administration of a pharmaceutical agent to an individual can be continuous, chronic, short or intermittent.
Administration can parenteral or non-parenteral.
As used herein, "agent" means an active substance that can provide a therapeutic benefit when administered to an animal. "First agent" means a therapeutic compound of the invention. For example, a first agent can be an antisense oligonucleotide targeting apoCIII. "Second agent"
means a second therapeutic compound of the invention (e.g. a second antisense oligonucleotide targeting apoCIII) and/or a non-apoCIII
therapeutic compound.
As used herein, "amelioration" or "ameliorate" or "ameliorating" refers to a lessening of at least one indicator, sign, or symptom of an associated disease, disorder, or condition.
The severity of indicators can be determined by subjective or objective measures, which are known to those skilled in the art.

As used herein, "animal" refers to a human or non-human animal, including, but not limited to, mice, rats, rabbits, dogs, cats, pigs, and non-human primates, including, but not limited to, monkeys and chimpanzees.
As used herein, "ApoCIII", "Apolipoprotein C-III" or "ApoC3" means any nucleic acid or protein sequence encoding ApoCIII. For example, in certain embodiments, an ApoCIII
includes a DNA sequence encoding ApoCIII, a RNA sequence transcribed from DNA encoding ApoCIII
(including genomic DNA
comprising introns and exons), a mRNA sequence encoding ApoCIII, or a peptide sequence encoding ApoCIII.
As used herein, "ApoCIII nucleic acid" means any nucleic acid encoding ApoCIII. For example, in certain embodiments, an ApoCIII nucleic acid includes a DNA sequence encoding ApoCIII, a RNA sequence transcribed from DNA encoding ApoCIII (including genomic DNA comprising introns and exons), and a mRNA sequence encoding ApoCIII.
As used herein, "ApoCIII specific inhibitor" refers to any agent capable of specifically inhibiting the expression of ApoCIII mRNA and/or the expression or activity of ApoCIII
protein at the molecular level.
For example, ApoCIII specific inhibitors include nucleic acids (including antisense compounds), peptides, antibodies, small molecules, and other agents capable of inhibiting the expression of ApoCIII mRNA and/or ApoCIII protein. In certain embodiments, the nucleic acid is an antisense compound. In certain embodiments, the antisense compound is a an oligonucleotide targeting ApoCIII.
In certain embodiments, the oligonucleotide targeting ApoCIII is a modified oligonucleotide targeting ApoCIII. In certain embodiments, the oligonucleotide targeting ApoCIII is a modified oligonucleotide targeting ApoCIII with a conjugate group. In certain embodiments, the oligonucleotide targeting ApoCIII has a sequence as shown in SEQ ID
NOs:19-96, 209-221 or another sequence (for example, such as those disclosed in PCT Publication WO
2004/093783 or PCT Publication WO 2012/149495, all incorporated-by-reference herein). In certain embodiments, by specifically modulating ApoCIII mRNA level and/or ApoCIII
protein expression, ApoCIII
specific inhibitors may affect components of the lipogenic or glucogenic pathway. Similarly, in certain embodiments, ApoCIII specific inhibitors may affect other molecular processes in an animal.
As used herein, "ApoCIII mRNA" means a mRNA encoding an ApoCIII protein.
As used herein, "ApoCIII protein" means any protein sequence encoding ApoCIII.
As used herein, "atherosclerosis" means a hardening of the arteries affecting large and medium-sized arteries and is characterized by the presence of fatty deposits. The fatty deposits are called "atheromas" or "plaques," which consist mainly of cholesterol and other fats, calcium and scar tissue, and damage the lining of arteries.
As used herein, "coronary heart disease (CHD)" means a narrowing of the small blood vessels that supply blood and oxygen to the heart, which is often a result of atherosclerosis.
As used herein, "diabetes mellitus" or "diabetes" is a syndrome characterized by disordered metabolism and abnormally high blood sugar (hyperglycemia) resulting from insufficient levels of insulin or reduced insulin sensitivity. The characteristic symptoms are excessive urine production (polyuria) due to high blood glucose levels, excessive thirst and increased fluid intake (polydipsia) attempting to compensate for increased urination, blurred vision due to high blood glucose effects on the eye's optics, unexplained weight loss, and lethargy.
As used herein, "diabetic dyslipidemia" or "type 2 diabetes with dyslipidemia"
means a condition characterized by Type 2 diabetes, reduced HDL-C, elevated triglycerides (TG), and elevated small, dense LDL particles.
As used herein, "diluent" means an ingredient in a composition that lacks pharmacological activity, but is pharmaceutically necessary or desirable. For example, the diluent in an injected composition can be a liquid, e.g. saline solution.
As used herein, "dyslipidemia" refers to a disorder of lipid and/or lipoprotein metabolism, including lipid and/or lipoprotein overproduction or deficiency. Dyslipidemias can be manifested by elevation of lipids such as chylomicron, cholesterol and triglycerides as well as lipoproteins such as low-density lipoprotein (LDL) cholesterol.
As used herein, "dosage unit" means a form in which a pharmaceutical agent is provided, e.g. pill, tablet, or other dosage unit known in the art. In certain embodiments, a dosage unit is a vial containing lyophilized antisense oligonucleotide. In certain embodiments, a dosage unit is a vial containing reconstituted antis ens e oligonucleotide.
As used herein, "dose" means a specified quantity of a pharmaceutical agent provided in a single administration, or in a specified time period. In certain embodiments, a dose can be administered in one, two, or more boluses, tablets, or injections. For example, in certain embodiments where subcutaneous administration is desired, the desired dose requires a volume not easily accommodated by a single injection, therefore, two or more injections can be used to achieve the desired dose. In certain embodiments, the pharmaceutical agent is administered by infusion over an extended period of time or continuously. Doses can be stated as the amount of pharmaceutical agent per hour, day, week, or month.
Doses can also be stated as mg/kg or g/kg.
As used herein, "effective amount" or "therapeutically effective amount" means the amount of active pharmaceutical agent sufficient to effectuate a desired physiological outcome in an individual in need of the agent. The effective amount can vary among individuals depending on the health and physical condition of the individual to be treated, the taxonomic group of the individuals to be treated, the formulation of the composition, assessment of the individual's medical condition, and other relevant factors.
As used herein, "Fredrickson Type I" is also known as "Lipoprotein lipase deficiency", "LPLD", "Familial Chylomicronemia Syndrome" or "FCS" and exists in several forms: Type la (also known as Buerger-Gruestz syndrome) is a lipoprotein lipase deficiency commonly due to a deficiency of LPL or altered ApoC-II; Type Ib (also known as familial apoprotein CII deficiency) is a condition caused by lack of lipoprotein lipase activator apoprotein C-II; and Type Ic is a chylomicronemia due to circulating inhibitor of lipoprotein lipase. Type I is a rare disorder that usually presents in childhood. It is characterized by severe elevations in chylomicrons and extremely elevated TG levels (always reaching well above 1000 mg/dL and not infrequently rising as high as 10,000 mg/dL or more) with episodes of abdominal pain, recurrent acute pancreatitis, eruptive cutaneous xanthomata, and hepatosplenomegaly. Patients rarely develop atherosclerosis, perhaps because their plasma lipoprotein particles are too large to enter into the arterial intima (Nordestgaard et al., J Lipid Res, 1988, 29:1491-1500; Nordestgaard et al., Arteriosclerosis, 1988, 8:421-428). Type I is usually caused by mutations of either the LPL gene, or of the gene's cofactor ApoC-II, resulting in the inability of affected individuals to produce sufficient functionally active LPL. Patients are either homozygous for such mutations or compound heterozygous. Fredrickson Type I can also be due to mutations in the GPIHBP1, AP0A5, LMF1 or other genes leading to dysfunctional LPL. Brunzell, In: Pagon RA, Adam MP, Bird TD, Dolan CR, Fong CT, Stephens K, editors. GeneReviewsTM
[Internet]. Seattle (WA):
University of Washington, Seattle; 1993-2013.1999 Oct 12 [updated 2011 Dec 15]. Further, Fredrickson Type I, in some instances, can be due to the presence of LPL inhibitors (e.g., anti-LPL antibodies) in an individual causing dysfunctional LPL. The prevalence of Fredrickson Type I is approximately 1 in 1,000,000 in the general population and much higher in South Africa and Eastern Quebec as a result of a founder effect.
Patients respond minimally, or not at all, to TG-lowering drugs (Tremblay et al., J Clin Lipidol, 2011, 5:37-44; Brisson et al., Pharmacogenet Genom, 2010, 20:742-747) and hence restriction of dietary fat to 20 grams/day or less is used to manage the symptoms of this rare disorder.
As used herein, "fully complementary" or "100% complementary" means each nucleobase of a nucleobase sequence of a first nucleic acid has a complementary nucleobase in a second nucleobase sequence of a second nucleic acid. In certain embodiments, a first nucleic acid is an antisense compound and a second nucleic acid is a target nucleic acid.
As used herein, "glucose" is a monosaccharide used by cells as a source of energy and inflammatory intermediate. "Plasma glucose" refers to glucose present in the plasma.
As used herein, "high density lipoprotein-C" or "HDL-C" means cholesterol associated with high density lipoprotein particles. Concentration of HDL-C in serum (or plasma) is typically quantified in mg/dL or nmon. "Serum HDL-C" and "plasma HDL-C" mean HDL-C in serum and plasma, respectively.
As used herein, "HMG-CoA reductase inhibitor" means an agent that acts through the inhibition of the enzyme HMG-CoA reductase, such as atorvastatin, rosuvastatin, fluvastatin, lovastatin, pravastatin, and simvastatin.
As used herein, "hypercholesterolemia" means a condition characterized by elevated cholesterol or circulating (plasma) cholesterol, LDL-cholesterol and VLDL-cholesterol, as per the guidelines of the Expert Panel Report of the National Cholesterol Educational Program (NCEP) of Detection, Evaluation of Treatment of high cholesterol in adults (see, Arch. Int. Med. (1988) 148, 36-39).
As used herein, "hyperlipidemia" or "hyperlipemia" is a condition characterized by elevated serum lipids or circulating (plasma) lipids. This condition manifests an abnormally high concentration of fats. The lipid fractions in the circulating blood are cholesterol, low density lipoproteins, very low density lipoproteins, chylomicrons and triglycerides. The Fredrickson classification of hyperlipidemias is based on the pattern of TG and cholesterol-rich lipoprotein particles, as measured by electrophoresis or ultracentrifugation and is commonly used to characterize primary causes of hyperlipidemias such as hypertriglyceridemia (Fredrickson and Lee, Circulation, 1965, 31:321-327; Fredrickson et al., New Eng J Med, 1967, 276 (1): 34-42).
As used herein, "hypertriglyceridemia" means a condition characterized by elevated triglyceride levels. Hypertriglyceridemia is the consequence of increased production and/or reduced or delayed catabolism of triglyceride (TG)-rich lipoproteins: VLDL and, to a lesser extent, chylomicrons (CM). Its etiology includes primary (i.e. genetic causes) and secondary (other underlying causes such as diabetes, metabolic syndrome/insulin resistance, obesity, physical inactivity, cigarette smoking, excess alcohol and a diet very high in carbohydrates) factors or, most often, a combination of both (Yuan et al. CMAJ, 2007, 176:1113-1120). Hypertriglyceridemia is a common clinical trait associated with an increased risk of cardiometabolic disease (Hegele et al. 2009, Hum Mol Genet, 18: 4189-4194;
Hegele and PoIlex 2009, Mol Cell Biochem, 326: 35-43) as well as of occurrence of acute pancreatitis in the most severe forms (Toskes 1990, Gastroenterol Clin North Am, 19: 783-791; Gaudet et al. 2010, Atherosclerosis Supplements, 11: 55-60; Catapano et al. 2011, Atherosclerosis, 217S: S 1 -S44; Tremblay et al. 2011, J Clin Lipidol, 5: 37-44). Examples of cardiometabolic disease include, but are not limited to, diabetes, metabolic syndrome/insulin resistance, and genetic disorders such as familial chylomicronemia syndrome (FCS), familial combined hyperlipidemia and familial hypertriglyceridemia. Borderline high TG levels (150-199 mg/dL) are commonly found in the general population and are a common component of the metabolic syndrome/insulin resistance states. The same is true for high TG levels (200-499 mg/dL) except that as plasma TG levels increase, underlying genetic factors play an increasingly important etiologic role. Very high TG levels (,..-500 mg/dL) are most often associated with elevated CM levels as well, and are accompanied by increasing risk for acute pancreatitis. The risk of pancreatitis is considered clinically significant if TG levels exceed 880 mg/dL (>10 mmol) and the European Atherosclerosis Society/European Society of Cardiology (EAS/ESC) 2011 guidelines state that actions to prevent acute pancreatitis are mandatory (Catapano et al.
2011, Atherosclerosis, 217S: S1-S44). According to the EAS/ESC 2011 guidelines, hypertriglyceridemia is the cause of approximately 10% of all cases of pancreatitis, and development of pancreatitis can occur at TG
levels between 440-880 mg/dL. Based on evidence from clinical studies demonstrating that elevated TG
levels are an independent risk factor for atherosclerotic CVD, the guidelines from both the National Cholesterol Education Program Adult Treatment Panel III (NCEP 2002, Circulation, 106: 3143-421) and the American Diabetes Association (ADA 2008, Diabetes Care, 31: S12-S54.) recommend a target TG level of less than 150 mg/dL to reduce cardiovascular risk.
As used herein, "identifying" or "selecting an animal with metabolic or cardiovascular disease"
means identifying or selecting a subject prone to or having been diagnosed with a metabolic disease, a cardiovascular disease, or a metabolic syndrome; or, identifying or selecting a subject having any symptom of a metabolic disease, cardiovascular disease, or metabolic syndrome including, but not limited to, hypercholesterolemia, hyperglycemia, hyperlipidemia, hypertriglyceridemia, hypertension increased insulin resistance, decreased insulin sensitivity, above normal body weight, and/or above normal body fat content or any combination thereof Such identification can be accomplished by any method, including but not limited to, standard clinical tests or assessments, such as measuring serum or circulating (plasma) cholesterol, measuring serum or circulating (plasma) blood-glucose, measuring serum or circulating (plasma) triglycerides, measuring blood-pressure, measuring body fat content, measuring body weight, and the like.
As used herein, "improved cardiovascular outcome" means a reduction in the occurrence of adverse cardiovascular events, or the risk thereof Examples of adverse cardiovascular events include, without limitation, death, reinfarction, stroke, cardiogenic shock, pulmonary edema, cardiac arrest, and atrial dysrhythmia.
As used herein, "immediately adjacent" means there are no intervening elements between the immediately adjacent elements, for example, between regions, segments, nucleotides and/or nucleosides.
As used herein, "increasing HDL" or "raising HDL" means increasing the level of HDL in an animal after administration of at least one compound of the invention, compared to the HDL level in an animal not administered any compound.
As used herein, "individual" or "subject" or "animal" means a human or non-human animal selected for treatment or therapy.
As used herein, "individual in need thereof' refers to a human or non-human animal selected for treatment or therapy that is in need of such treatment or therapy.
As used herein, "induce", "inhibit", "potentiate", "elevate", "increase", "decrease", "reduce" or the like denote quantitative differences between two states. For example, "an amount effective to inhibit the activity or expression of apoCIII" means that the level of activity or expression of apoCIII in a treated sample will differ from the level of apoCIII activity or expression in an untreated sample. Such terms are applied to, for example, levels of expression, and levels of activity.
As used herein, "inflammatory condition" refers to a disease, disease state, syndrome, or other condition resulting in inflammation. For example, rheumatoid arthritis and liver fibrosis are inflammatory conditions. Other examples of inflammatory conditions include sepsis, myocardial ischemia/reperfusion injury, adult respiratory distress syndrome, nephritis, graft rejection, inflammatory bowel disease, multiple sclerosis, arteriosclerosis, atherosclerosis and vasculitis.
As used herein, "inhibiting the expression or activity" refers to a reduction or blockade of the expression or activity of a RNA or protein and does not necessarily indicate a total elimination of expression or activity.
As used herein, "insulin resistance" is defined as the condition in which normal amounts of insulin are inadequate to produce a normal insulin response from fat, muscle and liver cells. Insulin resistance in fat cells results in hydrolysis of stored triglycerides, which elevates free fatty acids in the blood plasma. Insulin resistance in muscle reduces glucose uptake whereas insulin resistance in liver reduces glucose storage, with both effects serving to elevate blood glucose. High plasma levels of insulin and glucose due to insulin resistance often leads to metabolic syndrome and type 2 diabetes.
As used herein, "insulin sensitivity" is a measure of how effectively an individual processes glucose.
An individual having high insulin sensitivity effectively processes glucose whereas an individual with low insulin sensitivity does not effectively process glucose.
As used herein, "lipid-lowering" means a reduction in one or more lipids (e.g., LDL, VLDL) in a subject. "Lipid-raising" means an increase in a lipid (e.g., HDL) in a subject. Lipid-lowering or lipid-raising can occur with one or more doses over time.
As used herein, "lipid-lowering therapy" or "lipid lowering agent" means a therapeutic regimen provided to a subject to reduce one or more lipids in a subject. In certain embodiments, a lipid-lowering therapy is provided to reduce one or more of apo(a), apoCIII, CETP, apoB, total cholesterol, LDL-C, VLDL-C, IDL-C, non-HDL-C, triglycerides, small dense LDL particles, and Lp(a) in a subject. Examples of lipid-lowering therapy include, but are not limited to, apoB inhibitors, statins, fibrates and MTP inhibitors.
As used herein, "lipoprotein", such as VLDL, LDL and HDL, refers to a group of proteins found in the serum, plasma and lymph and are important for lipid transport. The chemical composition of each lipoprotein differs, for example, in that the HDL has a higher proportion of protein versus lipid, whereas the VLDL has a lower proportion of protein versus lipid.
As used herein, "Lipoprotein Lipase" or "LPL" refers to an enzyme that hydrolyzes TGs found in lipoproteins, such as CM or VLDL, into free fatty acids and monoacylglycerols.
LPL requires apo C-II as a cofactor to function in hydrolyzing TGs. LPL is mainly produced in skeletal muscle, fat tissue, and heart muscle. Hydrolysis and removal of TG from CM and VLDL normally protects against excessive postprandial rise in CM mass and TG.
As used herein, "Lipoprotein lipase deficient", "lipoprotein lipase deficiency", "LPL deficiency" or "LPLD" is also known as "Fredrickson's Type I dyslipidemia", "chylomicronemia", "Familial Chylomicronemia Syndrome" or "FCS". Although subjects with LPLD generally lack LPL or LPL activity necessary for effective breakdown of fatty acids such as TGs, these subjects may still have a minimal LPL
activity or express a minimal level of LPL. In some instances, a LPLD subject may express LPL or have LPL
activity up to about, or no more than, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2% or 1% activity. In other instances, the LPLD
subject has no measurable LPL
or LPL activity. One embodiment of LPLD encompasses subjects with "hyperlipoproteinemia type Ia" (also known as "Fredrickson's Type Ia") and refers to the inability of the subjects to produce sufficient functional lipoprotein lipase enzymes necessary for effective breakdown of fatty acids such as TGs. The inability to breakdown TGs leads to hypertriglyceridemia in the subject and, often more than 12 hours after meals, hyperTG and chylomicronemia are still present and visible as lipemia. Type Ia is commonly caused by one or more mutations in the LPL gene. As disclosed herein, LPLD also encompasses subjects that have dysfunctional lipoprotein lipase such as those subjects with "hyperlipoproteinemia type Ib" (also known as "Fredrickson's Type Ib") and "hyperlipoproteinemia type Ic" (also known as "Fredrickson's Type Ic"). Type Ib is caused by lack of lipoprotein lipase activator apoprotein C-II. Type Ic is due to a circulating inhibitor of lipoprotein lipase. As with Type la, Type lb/lc subjects suffer from an inability to breakdown TGs leading to hypertriglyceridemia and hyperTG and chylomicronemia are still present and visible as lipemia often more than 12 hours after meals. In certain embodiments, LPLD is associated with at least one mutation in the LPL
gene such as P207L, G188L or D9N or other mutations that affect LPL (Brunzell, In: Pagon RA, Adam MP, Bird TD, Dolan CR, Fong CT, Stephens K, editors. GeneReviewsTM [Internet].
Seattle (WA): University of Washington, Seattle; 1993-2013.1999 Oct 12 [updated 2011 Dec 15]).
As used herein, "low density lipoprotein-cholesterol (LDL-C)" means cholesterol carried in low density lipoprotein particles. Concentration of LDL-C in serum (or plasma) is typically quantified in mg/dL
or nmon. "Serum LDL-C" and "plasma LDL-C" mean LDL-C in the serum and plasma, respectively.
As used herein, "major risk factors" refers to factors that contribute to a high risk for a particular disease or condition. In certain embodiments, major risk factors for coronary heart disease include, without limitation, cigarette smoking, hypertension, high LDL, low HDL-C, family history of coronary heart disease, age, and other factors disclosed herein.
As used herein, "metabolic disorder" or "metabolic disease" refers to a condition characterized by an alteration or disturbance in metabolic function. "Metabolic" and "metabolism"
are terms well known in the art and generally include the whole range of biochemical processes that occur within a living organism.
Metabolic disorders include, but are not limited to, hyperglycemia, prediabetes, diabetes (type 1 and type 2), obesity, insulin resistance, metabolic syndrome and dyslipidemia due to type 2 diabetes.
As used herein, "metabolic syndrome" means a condition characterized by a clustering of lipid and non-lipid cardiovascular risk factors of metabolic origin. In certain embodiments, metabolic syndrome is identified by the presence of any 3 of the following factors: waist circumference of greater than 102 cm in men or greater than 88 cm in women; serum triglyceride of at least 150 mg/dL;
HDL-C less than 40 mg/dL in men or less than 50 mg/dL in women; blood pressure of at least 130/85 mmHg;
and fasting glucose of at least 110 mg/dL. These determinants can be readily measured in clinical practice (JAMA, 2001, 285: 2486-2497).
"Parenteral administration" means administration through injection or infusion. Parenteral administration includes subcutaneous administration, intravenous administration, intramuscular administration, intraarterial administration, intraperitoneal administration, or intracranial administration, e.g.
intrathecal or intracerebroventricular administration. Administration can be continuous, chronic, short or intermittent.
As used herein, "peptide" means a molecule formed by linking at least two amino acids by amide bonds. Peptide refers to polypeptides and proteins.

As used herein, "pharmaceutical agent" means a substance that provides a therapeutic benefit when administered to an individual. For example, in certain embodiments, an antisense oligonucleotide targeted to apoCIII is a pharmaceutical agent.
As used herein, "pharmaceutical composition" or "composition" means a mixture of substances suitable for administering to an individual. For example, a pharmaceutical composition can comprise one or more active agents and a pharmaceutical carrier e.g., a sterile aqueous solution.
As used herein, "pharmaceutically acceptable derivative" encompasses derivatives of the compounds described herein such as solvates, hydrates, esters, prodrugs, polymorphs, isomers, isotopically labelled variants, pharmaceutically acceptable salts and other derivatives known in the art.
As used herein, "pharmaceutically acceptable salts" means physiologically and pharmaceutically acceptable salts of antisense compounds, i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto. The term "pharmaceutically acceptable salt" or "salt" includes a salt prepared from pharmaceutically acceptable non-toxic acids or bases, including inorganic or organic acids and bases. "Pharmaceutically acceptable salts" of the compounds described herein may be prepared by methods well-known in the art. For a review of pharmaceutically acceptable salts, see Stahl and Wermuth, Handbook of Pharmaceutical Salts: Properties, Selection and Use (Wiley-VCH, Weinheim, Germany, 2002). Sodium salts of antisense oligonucleotides are useful and are well accepted for therapeutic administration to humans. Accordingly, in one embodiment the compounds described herein are in the form of a sodium salt.
As used herein, "portion" means a defined number of contiguous (i.e. linked) nucleobases of a nucleic acid. In certain embodiments, a portion is a defined number of contiguous nucleobases of a target nucleic acid. In certain embodiments, a portion is a defined number of contiguous nucleobases of an antisense compound.
As used herein, "prevent"or "preventing" refers to delaying or forestalling the onset or development of a disease, disorder, or condition for a period of time from minutes to indefinitely. Prevent also means reducing risk of developing a disease, disorder, or condition.
As used herein, "raise" means to increase in amount. For example, to raise plasma HDL levels means to increase the amount of HDL in the plasma.
As used herein, "reduce" means to bring down to a smaller extent, size, amount, or number. For example, to reduce plasma triglyceride levels means to bring down the amount of triglyceride in the plasma.
As used herein, "region" or "target region" is defined as a portion of the target nucleic acid having at least one identifiable structure, function, or characteristic. For example, a target region may encompass a 3' UTR, a 5' UTR, an exon, an intron, an exon/intron junction, a coding region, a translation initiation region, translation termination region, or other defined nucleic acid region. The structurally defined regions for apoCIII can be obtained by accession number from sequence databases such as NCBI and such information is incorporated herein by reference. In certain embodiments, a target region may encompass the sequence from a 5' target site of one target segment within the target region to a 3' target site of another target segment within the target region.
As used herein, "second agent" or "second therapeutic agent" means an agent that can be used in combination with a "first agent". A second therapeutic agent can include, but is not limited to, antisense oligonucleotides targeting apoCIII. A second agent can also include anti-apoCIII antibodies, apoCIII peptide inhibitors, cholesterol lowering agents, lipid lowering agents, glucose lowering agents and anti-inflammatory agents.
As used herein, "segments" are defined as smaller, sub-portions of regions within a nucleic acid. For example, a "target segment" means the sequence of nucleotides of a target nucleic acid to which one or more antisense compounds is targeted. "5' target site" refers to the 5'-most nucleotide of a target segment. "3' target site" refers to the 3'-most nucleotide of a target segment.
Alternatively, a "start site" can refer to the 5'-most nucleotide of a target segment and a "stop site" refers to the 3'-most nucleotide of a target segment. A
target segment can also begin at the "start site" of one sequence and end at the "stop site" of another sequence.
As used herein, "statin" means an agent that inhibits the activity of HMG-CoA
reductase.
As used herein, "subcutaneous administration" means administration just below the skin.
As used herein, "subject" means a human or non-human animal selected for treatment or therapy.
As used herein, "symptom of cardiovascular disease or disorder" means a phenomenon that arises from and accompanies the cardiovascular disease or disorder and serves as an indication of it. For example, angina; chest pain; shortness of breath; palpitations; weakness; dizziness;
nausea; sweating; tachycardia;
bradycardia; arrhythmia; atrial fibrillation; swelling in the lower extremities; cyanosis; fatigue; fainting;
numbness of the face; numbness of the limbs; claudication or cramping of muscles; bloating of the abdomen;
or fever are symptoms of cardiovascular disease or disorder.
As used herein, "targeting" or "targeted" means the process of design and selection of an antisense compound that will specifically hybridize to a target nucleic acid and induce a desired effect.
As used herein, "therapeutically effective amount" means an amount of a pharmaceutical agent that provides a therapeutic benefit to an individual.
As used herein, "therapeutic lifestyle change" means dietary and lifestyle changes intended to lower fat/adipose tissue mass and/or cholesterol. Such change can reduce the risk of developing heart disease, and may includes recommendations for dietary intake of total daily calories, total fat, saturated fat, polyunsaturated fat, monounsaturated fat, carbohydrate, protein, cholesterol, insoluble fiber, as well as recommendations for physical activity.
As used herein, "treat" or "treating" refers to administering a compound described herein to effect an alteration or improvement of a disease, disorder, or condition.
As used herein, "triglyceride" or "TG" means a lipid or neutral fat consisting of glycerol combined with three fatty acid molecules.

As used herein, "type 2 diabetes," (also known as "type 2 diabetes mellitus", "diabetes mellitus, type 2", "non-insulin-dependent diabetes", "NIDDM", "obesity related diabetes", or "adult-onset diabetes") is a metabolic disorder that is primarily characterized by insulin resistance, relative insulin deficiency, and hyperglycemia.
Certain Embodiments Certain embodiments provide a compounds and methods for decreasing ApoCIII
mRNA and protein expression. In certain embodiments, the compound is an ApoCIII specific inhibitor for treating, preventing, or ameliorating an ApoCIII associated disease. In certain embodiments, the compound is an antisense oligonucleotide targeting ApoCIII. In certain embodiments, the compound is an modified oligonucleotide targeting ApoCIII and a conjugate group.
In certain embodiments, a compound comprises a siRNA or antisense oligonucleotide targeted to Apolipoprotein C-III (ApoC-III) known in the art and a conjugate group described herein. Examples of antisense oligonucleotides targeted to ApoC-III suitable for conjugation include but are not limited to those disclosed in US Patent Application Publication No. US 2013/0317085, which is incorporated by reference in its entirety herein. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 19-96 and 209-221 disclosed in US
2013/0317085 and a conjugate group described herein. The nucleobase sequences of all of the aforementioned referenced SEQ ID
NOs are incorporated by reference herein.
In certain embodiments, the modified oligonucleotide with the conjugate group has a nucleobase sequence comprising at least 8 contiguous nucleobases of a sequence selected from any sequence disclosed in U.S. Patent 7,598,227, U.S. Patent 7,750,141, PCT Publication WO 2004/093783 or PCT Publication WO
2012/149495, all incorporated-by-reference herein. In certain embodiments, the modified oligonucleotide has a sequence selected from any sequence disclosed in U.S. Patent 7,598,227, U.S.
Patent 7,750,141, PCT
Publication WO 2004/093783 or PCT Publication WO 2012/149495, all incorporated-by-reference herein.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides. In certain embodiments, the modified oligonucleotide with the conjugate group consists of 15 to 30, 18 to 24, 19 to 22, 13 to 25, 14 to 25, 15 to 25 linked nucleosides. In certain embodiments, the modified oligonucleotide with the conjugate group comprises at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29 or 30 linked nucleosides. In certain embodiments, the modified oligonucleotide with the conjugate group consists of 20 linked nucleosides.
Certain embodiments provide a compound comprising a modified oligonucleotide with a conjugate group targeting ApoCIII and has a sequence complementary to any of the sequences set forth in GENBANK

Accession No. NM 000040.1 (incorporated herein as SEQ ID NO: 1), GENBANK
Accession No.
NT 033899.8 truncated from nucleotides 20262640 to 20266603 (incorporated herein as SEQ ID NO: 2), and/or GenBank Accession No. NT_035088.1 truncated from nucleotides 6238608 to 6242565 (incorporated herein as SEQ ID NO: 3). In certain embodiments, the modified oligonucleotide is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or at least 100% complementary to any of SEQ ID NOs: 1-3. In certain embodiments, the compound comprises a modified oligonucleotide targeting ApoCIII and a conjugate group, wherein the modified oligonucleotide comprises at least 8, at least 9, at least
10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, or 20 contiguous nucleobases complementary to an equal length portion of any of SEQ ID NOs: 1-3. In certain embodiments, the compound comprises a modified oligonucleotide targeting an ApoCIII segment and a conjugate group, wherein the modified oligonucleotide comprises at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, or 20 contiguous nucleobases complementary to an equal length portion of any of the target segments shown in Tables 121 and 124. In the tables, the "Start Site" refers to the 5'-most nucleotide of a target segment and "Stop Site" refers to the 3'-most nucleotide of a target segment. A target segment can range from the start site to the stop site of each sequence listed in the tables. Alternatively, the target segment can range from the start site of one sequence and end at the stop site of another sequence. For example, as shown in the tables, a target segment can range from 3533 to 3552, the start site to the stop site of SEQ ID NO: 87.
In another example, as shown in the tables, a target segment can range from 3514 to 3558, the start site of SEQ ID NO: 83 to the stop site of SEQ ID NO: 88. In certain embodiments, the antisense compound comprises at least 8 nucleobases of the sequence of SEQ ID NO: 87. In certain embodiments, the antisense compound comprises the sequence of SEQ ID NO: 87. In certain embodiments, the antisense compound consists of the sequence of SEQ ID NO:
87. In certain embodiments, the antisense compound is ISIS 304801.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the nucleobase sequence of the modified oligonucleotide is at least 80%, at least 85%, at least 90%, at least 95%, or 100% complementary to any of SEQ ID
NOs: 1-3. Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII and a conjugate group, wherein the nucleobase sequence of the modified oligonucleotide is at least 80%, at least 85%, at least 90%, at least 95%, or 100% complementary to any of the target segments disclosed herein.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and comprises a nucleobase sequence comprising a portion of at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, or 20 contiguous nucleobases complementary to an equal length portion of nucleobases 3533 to 3552 of SEQ ID NO: 3, wherein the nucleobase sequence of the modified oligonucleotide is at least 80% complementary to SEQ ID
NO: 3.

Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and comprises a nucleobase sequence comprising at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29 or 30 contiguous nucleobases complementary to an equal length portion of nucleobases 3514 to 3558 of SEQ ID
NO: 3, wherein the nucleobase sequence of the modified oligonucleotide is at least 80%
complementary to SEQ ID NO: 3.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8, at least 9, at least 10, at least
11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, or 20 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 19-96, 209-221. In certain embodiments, the conjugated modified oligonucleotide has a nucleobase sequence comprising at least 8 contiguous nucleobases of any one of the nucleobase sequences of SEQ ID NOs: 19-96, 209-221. In certain embodiments, the compound consists of any one of SEQ ID NOs: 19-96, 209-221 and a conjugate group.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, or 20 contiguous nucleobases of the nucleobase sequence of SEQ ID NO: 87. In certain embodiments, the modified oligonucleotide with the conjugate group has a nucleobase sequence comprising at least 8 contiguous nucleobases of the nucleobase sequence of SEQ
ID NO: 87. In certain embodiments, the compound consists of SEQ ID NO: 87 and a conjugate group.
In certain embodiments, the present disclosure provides conjugated antisense compounds represented by the following structure. In certain embodiments, the antisense compound comprises a modified oligonucleotide with ISIS 304801 with a 5'-X, wherein X is a conjugate group comprising GalNAc. In certain embodiments, the antisense compound consists of a modified oligonucleotide with ISIS 304801 with a 5'-X, wherein X is a conjugate group comprising GalNAc.

N2();NI o NH2 )i I
0 N N ilLyld At-IL
);_Dy o W.....0 0. a, (:)) o ccLy co_l 0 S-P=0 N 0 o I 1)5.1o S-P=0 NXII....NH o 'Irli'NH
o S-P=0 0 N al---V24/
N o---0 0) NH2 o e NH o 0,) S-P.0 e 1 o ATNo S-P=0 ....111..
al e 1 S-P=0 O
eLIH
6o c_o_VN 0 0 C)) 0 0 NH2 cliN 0 S-P=0 S -P=0 9 I ....iit'NH
o oI, lel 8 I o' 0,) O o S-P=0 eL.=NH
¨
c/
_ON 0 9 O\NH2 N 0 oC)/o 0 I C)/) 0 e 1 co4/o es-=o S-P=0 al o,) (S)c_c31 0 0 o NH2 S1=0 I
0 CI) 0) NH2 e 1 S -P=0NIA o N N
S-P=0 O I "
N N, cly o O A)), \
\
cc3/
0) 0 e?
o o o o 21.)51.1 S-P=0 ilLNH
S-PO=0 N0 6\c_oiN N NH2 c_O_I

O
OH 0õ) e 1 o e S-P=0 S-P=0 O ___________ O __________________________________________ In certain embodiments, the present disclosure provides conjugated antisense compounds represented by the following structure. In certain embodiments, the antisense compound comprises the conjugated modified oligonucleotide ISIS 678354. In certain embodiments, the antisense compound consists of the conjugated modified oligonucleotide ISIS 678354.

, NH2 'C) `T
NI---) HO OH 0 ,y0-F' =0 <1N
FINIcr, 0 N 1 N NO
Hjj--- 0 _0_i _ -- __ ---,ir NH ' CV 0 HO OH 0 0 (:)--) S-P =0 S-p -o Oz_3 <iqNljt:N --'-'1-HNH2 e 9 'IN
- I ,...
4 H OS/,N0 -iNH OV

HO OH o O 0-----i NH2 j---j==-N
e 1 -F'0 = I

HO 0----r-N 0 4 H ''CLHI o N N
W

NH
0 )_0_/0 o 9 0 9 ol S-P =00 0 illINZ
----1_01 N NH2 , 0 es4-;)=O Aftt o c:0 o o NO
, 1[I'NH
0 Ic_0_)/ 0 , 0 o 9 ill--11H
NH2 S-F'=0 N--0 O (:) S-(1-,)=0 ,) 1[1'NH

S-F'=0 itt NH 0 NO
(:)_)/ 0,, 0 0 (j) o 0 o e S-P =0 S-P =0 A)Ll IIH
0 N o --'0 N--'0 '-_Oj 0 CDS-CP=0 (11\11-11'ON---11FINH2 0'-CD

o 9 Nx-L,N

)_5 0 0 o_=

S-P =0 'IlL 0P
0-__N----o S-F'=0 2.-i 0 o o s-1, =0 OH 0,) 0 ____________________________________________________________ In certain embodiments, the present disclosure provides conjugated antisense compounds represented by the following structure. In certain embodiments, the antisense compound comprises the conjugated modified oligonucleotide ISIS 678357. In certain embodiments, the antisense compound consists of the conjugated modified oligonucleotide ISIS 678357.

9e NH2 NI-----HO OH 0 õO-F'=0 n, 1 ;1' AT'L' N
Ni< 0 N N
NO
HO 4 H o Hc ---,ir NH No csi 0 HO OH 0 0 a 9 (:)--) NH2 s-o=(:) (INIII:N'; ji-FIN H2 0 9 S-F'=0 A )11 HO 0---1-nN
4 H --i_o_l 0 ---,ir NH OZ

HO OH

---'N --- NH2 Nf----..
__...72...\ (.=) 1 a 9 S-=O
I fil ' O-F' =0 HO 0-- 0 o 0_--j ALIµl )_ 0 N N"---O
---,ir NH 0 __ W

0 9 0-..õõ-/ 0 S0 Arr ----10_)/N N NH2 0-P=0 O)_o_y o NH2 o c's1:2=o '1-,t O o,) o o N 0 (z..., - 0 -F1' =0 1[I'NH
0 0 lc_0_1 0 N--, 0 9 0,1 NH2 S-F'=0 NO
0-p =0 0 --__(y ,z) ij'''' N
o o o O-P=O
1[1',ZH

s-1,zo itt'XiN 0 _CLIN 0 0 0 0)0 a 9c) o-FI, =0 AlkNH
S-=O
NH
--,(:) N
0'0 (N bi-HN H2 0' e_P

e 9 sp N----)'--- N
S-F' =0 I

N N
)_5 0 o 9CO 0 S-P =0 '1)1'1 X 0 9 o---N 0 S-F'=0 itt:Z

o 9 o s-1, =0 OH 0-,_,-J
0 __________________________________________________________________ In certain embodiments, the present disclosure provides conjugated antisense compounds represented by the following structure. In certain embodiments, the antisense compound comprises a modified oligonucleotide with the nucleobase sequence of SEQ ID NO: 87 with a 5'-GalNAc with variability in the sugar mods of the wings. In certain embodiments, the antisense compound consists of a modified oligonucleotide with the nucleobase sequence of SEQ ID NO: 87 with a 5'-GalNAc with variability in the sugar mods of the wings.

, `-'NI----j-=--NL R5N
HO OH 0 00-F'=0 I ,) HN'<' cs, 0 N N
---.Ir NH CV R21 HO OH 0 o o Ri 0 0 0 R51., S-P =0 (---i- N
4 H O NI-IL' NH
() : N N NH2 (/ I S-F' =0 I ,I,L

,Ir NH OZ

HO OH 12 e 0 N
0 R S-F' NH2 f---- N
__....72._v =0 j HO 0-1-n N 0 4 H Z -IR =0 R5rJ.;,,,.. 0 N
N

W
R2 cN 0 0 e 0 (!`111LL'õr RI
0 S-F' =0 Z-P =0 R5,L011,õ 0 O
- - - iN N NH2 0 o R3 R5--C.,N
O R" 0 NO
z_=0 R5I-ILNH
O I j olri2¨/
NO

0. o R3 R5' NH

0 R3 R5,,i,,, S-P =0 ", ----Z-P =0 µõ i 'N 0 ------o L. IR

(D 47 ,/ R3 0 R ' 47¨r, 0 Z-P =0 R51-ji NH
0 0 R- R5I.,,LL, I I
S-F' =0 1 X 0 No R47¨T
Z-I(;=0 R5I---[1'NH
0 0 IR-, R51,11, NH

O N'-'0 0 , _CLy 0 P N

S-P0 l'IL- NH
= I Z-F' =0 I
0.0,N N NH2 N N

R5 o'IR), s-=o OP RVL
SP =O
NH
O_ /N O
I

S-P =0 ) R, O ____________________________________________________________ Wherein either R1 is ¨OCH2CH2OCH3 (M0E)and R2 is H; or R1 and R2 together form a bridge, wherein R1 is ¨0- and R2 is ¨CH2-, -CH(CH3)-, or -CH2CH2-, and R1 and R2 are directly connected such that the resulting bridge is selected from: -0-CH2-, -0-CH(CH3)-, and ¨0-CH2CH2-;
And for each pair of R3 and R4 on the same ring, independently for each ring:
either R3 is selected from H and -OCH2CH2OCH3 and R4 is H; or R3 and R4 together form a bridge, wherein R3 is ¨0-, and R4 is ¨
CH2-, -CH(CH3)-, or -CH2CH2-and R3 and R4 are directly connected such that the resulting bridge is selected from: -0-CH2-, -0-CH(CH3)-, and ¨0-CH2CH2-;
And R5 is selected from H and ¨CH3;

And Z is selected from S- and 0-.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide is single-stranded.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein at least one internucleoside linkage is a modified internucleoside linkage. In certain embodiments, the modified internucleoside linkage is a phosphorothioate internucleoside linkage. In certain embodiments, at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9 or at least 10 internucleoside linkages of said modified oligonucleotide are phosphorothioate internucleoside linkages. In certain embodiments, each internucleoside linkage is a phosphorothioate internucleoside linkage. In certain embodiments, the modified oligonucleotide comprises at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9 or at least 10 phosphodiester internucleoside linkages. In certain embodiments, each internucleoside linkage of the modified oligonucleotide is selected from a phosphodiester internucleoside linkage and a phosphorothioate internucleoside linkage.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein at least one nucleoside comprises a modified nucleobase. In certain embodiments, the modified nucleobase is a 5-methylcytosine.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide comprises at least one modified sugar. In certain embodiments, the modified sugar is a bicyclic sugar. In certain embodiments, the modified sugar comprises a 2'-0-methoxyethyl, a constrained ethyl, a 3'-fluoro-HNA or a 4'- (CH2)-0-2' bridge, wherein n is 1 or 2.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and comprises: (a) a gap segment consisting of linked deoxynucleosides; (b) a 5' wing segment consisting of linked nucleosides; (c) a 3' wing segment consisting of linked nucleosides;
and wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment and wherein each nucleoside of each wing segment comprises a modified sugar.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 20 linked nucleosides and comprises: (a) a gap segment consisting of ten linked deoxynucleosides; (b) a 5' wing segment consisting of five linked nucleosides; (c) a 3' wing segment consisting of five linked nucleosides; and wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment, wherein each nucleoside of each wing segment comprises a 2'-0-methoxyethyl sugar, wherein at least one internucleoside linkage is a phosphorothioate linkage and wherein each cytosine residue is a 5-methylcytosine.

Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 20 linked nucleosides and has a nucleobase sequence comprising at least 8 contiguous nucleobases of any of SEQ
ID NOs: 19-96, 209-221, wherein the modified oligonucleotide comprises: (a) a gap segment consisting of ten linked deoxynucleosides; (b) a 5' wing segment consisting of five linked nucleosides;
(c) a 3' wing segment consisting of five linked nucleosides; and wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment, wherein each nucleoside of each wing segment comprises a 2'-0-methoxyethyl sugar, wherein at least one internucleoside linkage is a phosphorothioate linkage and wherein each cytosine residue is a 5-methylcytosine.
Certain embodiments provide a compound comprising a modified oligonucleotide targeting ApoCIII
and a conjugate group, wherein the modified oligonucleotide consists of 20 linked nucleosides and has a nucleobase sequence comprising at least 8 contiguous nucleobases of SEQ ID NO:
87, wherein the modified oligonucleotide comprises: (a) a gap segment consisting of ten linked deoxynucleosides; (b) a 5' wing segment consisting of five linked nucleosides; (c) a 3' wing segment consisting of five linked nucleosides;
and wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment, wherein each nucleoside of each wing segment comprises a 2'-0-methoxyethyl sugar, wherein at least one internucleoside linkage is a phosphorothioate linkage and wherein each cytosine residue is a 5-methylcytosine.
Certain embodiments provide a modified oligonucleotide targeting ApoCIII and a conjugate group, wherein the modified oligonucleotide consists of 20 linked nucleosides with the nucleobase sequence of SEQ
ID NO: 87, wherein the modified oligonucleotide comprises: (a) a gap segment consisting of ten linked deoxynucleosides; (b) a 5' wing segment consisting of five linked nucleosides;
(c) a 3' wing segment consisting of five linked nucleosides; and wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment, wherein each nucleoside of each wing segment comprises a 2'4)-methoxyethyl sugar, wherein at least one internucleoside linkage is a phosphorothioate linkage and wherein each cytosine residue is a 5-methylcytosine.
In certain embodiments, the conjugate group is linked to the modified oligonucleotide at the 5' end of the modified oligonucleotide. In certain embodiments, the conjugate group is linked to the modified oligonucleotide at the 3' end of the modified oligonucleotide.
In certain embodiments, the conjugate group comprises exactly one ligand. In certain embodiments, the conjugate group comprises one or more ligands. In certain embodiments, the conjugate group comprises exactly two ligands. In certain embodiments, the conjugate group comprises two or more ligands. In certain embodiments, the conjugate group comprises three or more ligands. In certain embodiments, the conjugate group comprises exactly three ligands. In certain embodiments, each ligand is selected from among: a polysaccharide, modified polysaccharide, mannose, galactose, a mannose derivative, a galactose derivative, D-mannopyranose, L-Mannopyranose, D-Arabinose, L-Galactose, D-xylofuranose, L-xylofuranose, D-glucose, L-glucose, D-Galactose, L-Galactose, a-D-Mannofuranose, 13-D -Manno furanos e, a-D-Mannopyranose, 13-D -Mannopyranos e, a-D-Glucopyranose, [3-D -Glue opyranos e, a-D-Glucofuranose, [3-D-Glucofuranose, a-D-fructofuranose, a-D-fructopyranose, a-D-Galactopyranose, 13 -D-Galactopyranose, a-D-Galactofuranos e, 13 -D-Galactofuranose, gluc o s amine, sialic acid, a-D-galactos amine, N-Acetylgalactosamine, 2-Amino-3 - 0 -[(R)- 1 -carb oxyethyl] -2- deoxy- [3-D-g1ucopyranose, 2-D eoxy-2 -methylamino-L-gluc opyranos e, 4,6-Dideoxy-4-formamido-2,3 -di- 0-methyl-D-mannopyranose, 2-D eoxy-2 -sulfoamino-D-glucopyranose, N-Glycoloyl-a-neuraminic acid, 5-thio-13-D-g1ucopyranose, methyl 2,3,4-tri-O-acety1-1-thio-6-0-trityl-a-D-glucopyranoside, 4-Thio-13-D-ga1actopyranose, ethyl 3,4,6,7-tetra-0-acety1-2-deoxy-1,5-dithio-a-D-g/uco-heptopyranoside, 2,5-Anhydro-D-allononitrile, ribose, D-ribose, D-4-thioribose, L-ribose, L-4-thioribose. In certain embodiments, each ligand is N-acetyl galactosamine.
In certain embodiments, the conjugate group comprises:
HO H
Th¨N
AcHN H
0 nN
1r o 0 H H 8& ).1A
___/----7-1N"----i---"N y----- 0 -........-- N H N-(CF-12)6-0¨

H
./1:11v0 0 0 0-HO
NHAc HNrNHN--e0 OH
0¨/¨/¨%
HO\c>/`) HO
"\

In certain embodiments, the conjugate group comprises:

_r12.\/c(TcH)c HO
AcHN N

Z
AcHN 0 HOOH

AcHN

In certain embodiments, the conjugate group comprises:
HOOH
H

HO
--\
AcHN

AcHN H
--(HOOH
_...f.(2._\./011 0 HO "4 AcHN
In certain embodiments, the conjugate group comprises:
OH OH

HO....r.:....\__O

AcHN
OH OH
HO E
.T. ...\._ ) 0 ,crH 0 H oss 0 N e AcHN INI 0 INI 0 HO H
.....12..\/0 HO
NHAc In certain embodiments, the conjugate group comprises:

pH
HOOH
HOON

AcHN
0=P¨OH
HO OH
HOOiNIR., AcHN
0=P¨OH
HOOH
HO0rNr-Zo AcHN
In certain embodiments, the conjugate group comprises at least one phosphorus linking group or neutral linking group.
In certain embodiments, the conjugate group comprises a structure selected from among:
OH OH

FO¨P-0,Ak00,p,O,ss N

=
0 and OH
ccss cs&fr),11 m HN 0 \
"m wherein n is from 1 to 12; and wherein m is from 1 to 12.
In certain embodiments, the conjugate group has a tether having a structure selected from among:

0 Zi "sH-1¨H)2L. and cs5sNI-SeL
mi mi mi H ml wherein L is either a phosphorus linking group or a neutral linking group;
Z1 is C(=0)0-R2;
Z2 is H, C1-C6 alkyl or substituted C1-C6 alkY;
R2 is H, C1-C6 alkyl or substituted C1-C6 alky; and each m1 is, independently, from 0 to 20 wherein at least one m1 is greater than 0 for each tether.
In certain embodiments, the conjugate group has a tether having a structure selected from among:
0 COOH OH 2z.
/H-04-0 _____________ / and , m1 I mi m N mi OH ¨1 H 0 wherein Z2 is H or CH3; and each m1 is, independently, from 0 to 20 wherein at least one m1 is greater than 0 for each tether.
In certain embodiments, the conjugate group has tether having a structure selected from among:
cs&fr)1Fi m HN CSS5N 0 OH
m =
wherein n is from 1 to 12; and wherein m is from 1 to 12.
In certain embodiments, the conjugate group is covalently attached to the modified oligonucleotide.
In certain embodiments, the compound has a structure represented by the formula:

A ¨B¨C¨D¨EE ¨F) q wherein A is the modified oligonucleotide;
B is the cleavable moiety C is the conjugate linker D is the branching group each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, the compound has a structure represented by the formula:
A ¨(¨B ) ( C ) ( D ) ( E¨F) n2 ni n3 q wherein:
A is the modified oligonucleotide;
B is the cleavable moiety C is the conjugate linker D is the branching group each E is a tether;
each F is a ligand;
each n is independently 0 or 1; and q is an integer between 1 and 5.
In certain embodiments, the compound has a structure represented by the formula:
A¨B¨CiE¨F) q wherein A is the modified oligonucleotide;

B is the cleavable moiety;
C is the conjugate linker;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, the compound has a structure represented by the formula:
A¨C¨D¨(¨E¨F) q wherein A is the modified oligonucleotide;
C is the conjugate linker;
D is the branching group;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, the compound has a structure represented by the formula:
A¨C¨EE¨F) q wherein A is the modified oligonucleotide;
C is the conjugate linker;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, the compound has a structure represented by the formula:
A¨B¨D¨EE¨F) q wherein A is the modified oligonucleotide;
B is the cleavable moiety;
D is the branching group;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, the compound has a structure represented by the formula:
A¨B¨(E¨F) q wherein A is the modified oligonucleotide;
B is the cleavable moiety;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, the compound has a structure represented by the formula:
A ¨D ¨(¨E ¨F) q wherein A is the modified oligonucleotide;
D is the branching group;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.

In certain embodiments, the conjugate linker has a structure selected from among:

H
H H
n H , OH

ni\111 ; S-nSNS-1 .' ;
N
sk N H , , 0 n H n i µ
- - - hi N
----.1---)i--yi W-...N--- = '1_ ..,)11,...õ).=.. ./.....N = 1 µ 1 FN1 =

H H H
H N
slv ir N 00/'HNn NK0Q0/'H n ; ,-(ose;
n n n n H
H Q . , H
rrNH 1-C) n 0 0 _pH
iv0H
\/ 1n L ¨ .
"n_ n "n n n "n -n _ n _ -n csscri)LNI")-1-,5.7c and 1/A-OL

wherein each L is, independently, a phosphorus linking group or a neutral linking group; and each n is, independently, from 1 to 20.

In certain embodiments, the conjugate linker has a structure selected from among:

H H
µ). N 1AN =yµ. )NA .
H H ' 0 , H
' 0 HN 0 .
issLN ENI,s ; 0 Is-H

Oss _ A .
N.1");=rNISiiN , Ç-__ H H

H
H
csssN cX0/\/N sYs ; H H

H
"s H
`sssfN PC)//N csss sssLOF ; s&00,0s ; ssC000,sss ;

H
"s-rHThr N 0//N s's ;
e 0+0,1,r00,frrOoss ;

OH "3 3 H

1-0¨P-0 0 0 04-0-1 ; cs'sr('OL3 N6''2L
and csssWLNYOI¨O-1 H " 60H =

In certain embodiments, the conjugate linker has the followingstructure:
prrj O NO
6 0 .
In certain embodiments, the conjugate linker has a structure selected from among:
Os' ; sss'e.\/.\/ ; and In certain embodiments, the conjugate linker has a structure selected from among:
/OH
/OH
and OH "3 3 OH OH '3 "3 c' =
In certain embodiments, the conjugate linker has a structure selected from among:

O¨P ¨0 csss ccW HN 0 6 and ccW N K5µ

In certain embodiments, the conjugate linker comprises a pyrrolidine. In certain embodiments, the conjugate linker does not comprise a pyrrolidine.
In certain embodiments, the conjugate linker comprises PEG.

In certain embodiments, the conjugate linker comprises an amide. In certain embodiments, the conjugate linker comprises at least two amides. In certain embodiments, the conjugate linker does not comprise an amide. In certain embodiments, the conjugate linker comprises a polyamide.
In certain embodiments, the conjugate linker comprises an amine.
In certain embodiments, the conjugate linker comprises one or more disulfide bonds.
In certain embodiments, the conjugate linker comprises a protein binding moiety. In certain embodiments, the protein binding moiety comprises a lipid. In certain embodiments, the protein binding moiety is selected from among: cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-0(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, 03-(oleoyl)lithocholic acid, 03-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine), a vitamin (e.g., folate, vitamin A, vitamin E, biotin, pyridoxal), a peptide, a carbohydrate (e.g., monosaccharide, disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, polysaccharide), an endosomolytic component, a steroid (e.g., uvaol, hecigenin, diosgenin), a terpene (e.g., triterpene, e.g., sarsasapogenin, friedelin, epifriedelanol derivatized lithocholic acid), or a cationic lipid. In certain embodiments, the protein binding moiety is selected from among: a C16 to C22 long chain saturated or unsaturated fatty acid, cholesterol, cholic acid, vitamin E, adamantane or 1-pentafluoropropyl.

In certain embodiments, the conjugate linker has a structure selected from among:
H H 1¨NH
µ
N.j,,I.ri N I

N
I
css, I
H
, H
( ) n ,s I I

N
I
rO-P-01-1 , II
II
1 (. 0 -P-0 0 C

N I 1¨NH
I OH =
\ OH;
p N ; `11,<LHO

I I
0 04, (:) ,0 1 0t c)..6-0;1:1)/ OH
N IC)/

Th\1 n 0 IC)c"
H
H N . n , csc'= ' rrrrS'S*-r n 0 H

I
0, N
C)/
HHHH H
N;
n ,,,I,N .,.S.Hri=Lo , 0 ;:p.,,,,OF
I

I

0 0 1 ___ '/K \ j\.
,OH

10-6-0P" CSOH
\ ________________________________________________ n\ /1"'N ...%- PA/
S-S n 0 N
\ õõ H
N , ) and H N 'I'')N' IWILO
v,)r, H

wherein each n is, independently, is from 1 to 20; and p is from 1 to 6.

In certain embodiments, the conjugate linker has a structure selected from among:
\ S
q.
),O)L 0 N 0 H ,22.)LN,is,Sic) .

444' \
q.
)0)'L rrs4 0 N \
H 1,,y1N./Hrr H C).
N
n 0 N)0)a2"

I H
µ)e', N N JVII .
n rrJ4 O -1-\ 0 0,.
N o \
q H
csYPSX'IN )1(-0 ;
0;'aL

H N
IsylILN (:),ION 10 =
n H

µ
C) C0 C)\
IN j.L(-r hl A--rs\4 C0 IN rN)''2-. rr q n H
\
s b 0, N I
0 ¨P =0 N 0 I
O¨P=0 H 01-I ;

fl µ,N
µ0 , and O
O
H , css'N N
1- )n H

HO
wherein each n is, independently, from 1 to 20.

In certain embodiments, the conjugate linker has a structure selected from among:
, \
_r-rrj O.
\

NO
N
0 )aaL
10)'L
H

rrs4 1¨NH
\ A 0 cl, N II
0¨P¨OH
0 N)0;2''' I I

H H
V N ,..,...õ..11., õ..-...,ErIT,,NõH.,..k.,0 =
¨NH HO¨/

Z N )0)'L e, N I
I \ rO¨P¨OH
II
0 ' J6rs\j ' 0 1¨NH
A
))2.

H H

0 ¨I- 0Cy (:) põ0 OH
-0' 1 7 1-s 0 H N
0 vN (), \ 1 , - = D..
/OH
N 0 6\ ,,,,. ) 0 c ¨
S
S' l'-gLO
H

I O.
0, HHHH H

N'C),s0 NNN,--).,,õ0...,./ ; N)C) 0¨) H css' ; ,-rc SS O
' ;
.,,r N0 I

\ i... (.... õO
P OH
N ) Ot ,05 1 N----/
H )10 2 ¨ 0 e ; and H
i H

In certain embodiments, the conjugate linker has a structure selected from among:

J-Pri J-rsj \
\ 0 Q.
0)42- N0)'''"

µ)0 and µn wherein n is from 1 to 20.
In certain embodiments, the conjugate linker has a structure selected from among:

II µ
csss v,,y 0 ¨ Fi' ¨ 0 ¨ 6IWNK

0 and 0 In certain embodiments, the conjugate linker has a structure selected from among:

II
skeOLNHA
0¨P-0¨

"Wilv)in OHn H n 0 and 0 =
, wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
In certain embodiments, the conjugate linker has the following structure:

H .
In certain embodiments, the branching group has one of the following structures:
I I I

1_ 1 ) A1-1 Nn A¨

/ 1 /AA n . J/1 )1 A1 ( In 1¨ A A and 1¨ r1 Aj/1 )( l'n sµsts 5µ51.0 srs4 wherein each A1 is independently, 0, S, C=0 or NH; and each n is, independently, from 1 to 20.

In certain embodiments, the branching group has one of the following structures:
isfr scss and \ n n Ai \ss >0 s, wherein each A1 is independently, 0, S, C=0 or NH; and each n is, independently, from 1 to 20.
In certain embodiments, the branching group has the following structure:
\
0' / .
In certain embodiments, the branching group has the following structure:
\
0-.....
A
/0.____.-1 0' / .
In certain embodiments, the branching group has the following structure:
\---...
A
H

In certain embodiments, the branching group has the following structure:
vcc ______________________ )2' \s" .
In certain embodiments, the branching group comprises an ether.
In certain embodiments, the branching group has the following structure:
0 0 I 0 0 C)\ 0 NH
µ)N(-=r\- . HO 0 P 0 __ '2,,_) -(1\1-HL =
'=
NH 0 n OH
CH3 /m (:)) H 0 ( ln H 0 , rrss'NN;a2L ;
40 =
, r-rrr 0 (1.1 'n ,flflt rn NH 02i.\o n CH3 .,.E13 ( \ n i O / im ' cOsN
H ,_, \- ' 0 (,NH / .
CH3pgdpk, n irn (PRI
, and O
7 o ____ NH ( ____ , NH rs, cssL(NIL
H 0/m each n is, independently, from 1 to 20; and m is from 2 to 6.

In certain embodiments, the branching group has the following structure:

0, ill.

, 0 -,-rr''. \

<) H
= 0 0 7,¨NH rr's I

alit, I )¨ NH
NH
0 (:)'\ 0 IA . `'22.)(N-Nss c' .
./<

=
0 7 0....õ,..,, 7 cs5LN \" ; and '??-r NH
r"
H
.1lAPJ 0 In certain embodiments, the branching group has the following structure:
\
/
/0õ,......,-0 ,,, o¨

/
' ..,..,.,,, In certain embodiments, the branching group comprises:
o '7'LLNH

NH `':,1-----NH ,KN1"¨rr'ENIANA
H H
,sss,( 1,L O(/
N ON"."--..
H

0 vNH
, , , o 0 `24.. N H
0 rr'H 0 H H
n H H
O(/ 0 (( µ..... NH
,or o =
, wherein each j is an integer from 1 to 3; and wherein each n is an integer from 1 to 20.
In certain embodiments, the branching group comprises:
µ o H N
) \---",.....----\ )1"----NH

/
H j NH
N _P H I 1 ik N
N..,...õ......,"
H H i( 0 \
O/

HNJcc H . NH , 0 y',../¨',)L"----NH
NH
.......)....."

Y .11C--....'.--.....")1... N
N H IL N ---r .), 0 , H

ros,...r. NH
NH
, or o =
In certain embodiments, each tether is selected from among:

'&(;)-L1He'L and NI¨H)22"
m1 m1 m1 H m 1 wherein L is selected from a phosphorus linking group and a neutral linking group;
Z1 is C(=0)0-R2;
Z2 is H, C1-C6 alkyl or substituted C1-C6 alkY;
R2 is H, C1-C6 alkyl or substituted C1-C6 alky; and each m1 is, independently, from 0 to 20 wherein at least one m1 is greater than 0 for each tether.
In certain embodiments, each tether is selected from among:
_cp ;IL 0 COOH OH 2z.
/H /-04-0 ') and , m2 z I M2 m, N I I M2 OH ... H

wherein Z2 is H or CH3; and each m2 is, independently, from 0 to 20 wherein at least one m2 is greater than 0 for each tether.
In certain embodiments, each tether is selected from among:
OH
CI'fr)1Fd m HN CSSS)rN 0 m =
wherein n is from 1 to 12; and wherein m is from 1 to 12.
In certain embodiments, at least one tether comprises ethylene glycol.
In certain embodiments, at least one tether comprises an amide. In certain embodiments, at least one tether comprises a polyamide.

In certain embodiments, at least one tether comprises an amine.
In certain embodiments, at least two tethers are different from one another.
In certain embodiments, all of the tethers are the same as one another.
In certain embodiments, each tether is selected from among:

NF)c)(:)sr'lt . H ,= s 0 ;
Is3jµ Ck)r-1-1 n =
n H H
N n ;
.
; -k=\ rrss) \
' - - H
;
'fl H in II k in cs' n =

1¨N 0 0 o 0 1(iNgss' = 1.11 ;and lyN
n H n wherein each n is, independently, from 1 to 20; and each p is from 1 to about 6.
In certain embodiments, each tether is selected from among:

=
s, =
N 0 ; N

and -,sooss In certain embodiments, each tether has the following structure:

H H
N N
"n in n wherein each n is, independently, from 1 to 20.
In certain embodiments, each tether has the following structure:
N N

In certain embodiments, the tether has a structure selected from among:

Y(rN,c Y(rN,c 4 H n H
, or ; wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
In certain embodiments, the tether has a structure selected from among:

Y(rNC

,rsrr In certain embodiments, the ligand is galactose.
In certain embodiments, the ligand is mannose-6-phosphate.
In certain embodiments, each ligand is selected from among:

O
OH H
OH

and HO -i OH

wherein each R1 is selected from OH and NHCOOH.
In certain embodiments, each ligand is selected from among:
HOOH OH HO HO
OH OH
o HO
,rr . Hp() -0 ; HO HO -0 ------7-----\, - \rrs- ; HO is =
, NHAc r OH '' , 11 \ss44 HOOH OH
N
HO OH OH HOOH
HO is ; 0 0 H HO OH
OH
OH rrrr , HO---4--)---\/)+710'' ;
and OH OH
HO
OH
HO
_(.\,)1-%

HO
0 __________________ HO OH
OH
HO -C) HO

0 \sss' In certain embodiments, each ligand has the following structure:
HOOF!
HO _.\, \ , NHAc 1.- =

In certain embodiments, each ligand has the following structure:
HOOH

Nris NHAc =
In certain embodiments, the conjugate group comprises a cell-targeting moiety.
In certain embodiments, the conjugate group comprises a cell-targeting moiety having the following structure:
HOOH

n 0 , AcHN OH ))n HOOH
"n I .
AcHN OH n 0 0 n HO n OH
lo NHAc wherein each n is, independently, from 1 to 20.
In certain embodiments, the cell-targeting moiety has the following structure:

HO OH

H
AcHN 0 1 (i) OH ----HO OH

AcHN OH 0 0 y HO H
/0....õ7----/-------0-131µ..-0 HO OH
NHAc .
In certain embodiments, the cell-targeting moiety has the following structure:
HO OH

HO
)n HO OH NHAc H H ,t_ 1 \ in H 1 HO 01,...),....,N,Icr..^x7.N0,.......1 n n NHAc 0 0 0'"C in )n OH
HO HN----o H
__...,...70.....\70 N
)(n HO n NHAc 0 =
wherein each n is, independently, from 1 to 20.
In certain embodiments, the cell-targeting moiety has the following structure:

HO OH
H0*=\D

HO OH NHAc 0 HO
NHAc 0 OH
HO HN
NH
HO

NHAc =
In certain embodiments, the cell-targeting moiety comprises:
HO OH
HO
AcHN
NH
O
HO OH H
csss.
HN'ThrN
HOo AcHN 0 H? /OH
NH
HO
AcHN
=
In certain embodiments, the cell-targeting moiety comprises:

HN
AcHN
HOOH
HOCZ/C)1NN

AcHN NI'µ

HOOH

AcHN .
In certain embodiments, the cell-targeting moiety has the following structure:

_...1.2..\/0"6-r N).0 HO ` /4 H
N
AcHN 0 N--11,õ,------0,-.N>l, HO "4 H
Z
AcHN 0 H
HO OH

AcHN .
In certain embodiments, the cell-targeting moiety has the following structure:

(:)--r,iLil, r --h, l )-----\
HO
AcHN 0 µ

HO_....rf.:?...\/0N H

AcHN

HO_......r2._\/0-1ANIN

AcHN .
In certain embodiments, the cell-targeting moiety comprises:

pH
HOOH

AcHN
0=P¨OH
HOOH

AcHN
0=P¨OH

HOOH

AcHN =
In certain embodiments, the cell-targeting moiety has the following structure:

HOOH
HO "4 AcHN

/\, A
HO "4 H
AcHN
HOOH
HO
AcHN
=
In certain embodiments, the cell-targeting moiety comprises:

HO OH
AcHN ..>/---NH

HO OH
HO \r_H 0 N ic,scs AcHN

HO OH
HO
AcHN 0 1\0 HO OH

AcHN
0 .

In certain embodiments, the cell-targeting moiety comprises:
OH OH
HO*,0 0 AcHN N--NNANH
01-bH \--------\__H
N
HO :) \-_,---0 AcHN
N---.../"----N
H H
H
if N \(:) j\¨NH

HO, HO
NHAc .
In certain embodiments, the cell-targeting moiety comprises:
OH OH
HO....r:Z__0______)(t, NH
AcHN
OH OH
AcHN H 0 0 r HO
NHAc =
In certain embodiments, the cell-targeting moiety has the following structure:

HO
/O
"io H
N
AcHN 0 .-11......---0.....NA
HO
Z
AcHN 0 H0a0H
--CI
HO"r2-=\(rio HN
NHAc .

In certain embodiments, the cell-targeting moiety has the following structure:

NOON
0 oi_pi AcHN 0 o 0, HOOH cy, HO--7*--=\, iN
AcHN 0 9, jj P-HO.,(..:). 1-1 ocN
HO-" O
NHAc =
In certain embodiments, the cell-targeting moiety has the following structure:

HO--2--\---- N1NA----\

AcHN H

HO Tri4L'il 2 0 H
AcHN

_.4:2\vcg))ANI¨rN(c) AcHN .
In certain embodiments, the cell-targeting moiety has the following structure:

AcHN
H
HOOH 0 N µ.
HO--72--\11 0 AcHN

H0\7C) AcHN =
In certain embodiments, the cell-targeting moiety has the following structure:

0 k AcHN

µH
_CZ/0^()?N' AcHN

N N(0 AcHN
In certain embodiments, the cell-targeting moiety comprises:
pH
HO OH
HO

AcHN
0=P¨OH
HO OH
AcHN
0=P¨OH
HO OH
HO
AcHN
In certain embodiments, the cell-targeting moiety comprises:

g H
HO OH
3 NR.
HO

AcHN
0=P¨OH

AcHN
0=P¨OH
HO OH H
HO
AcHN
In certain embodiments, the cell-targeting moiety comprises:
HOOH r\OH
0 µ---0 AcHN
0=P¨OH

HO OH r---1 AcHN
0=P¨OH

HO OH
N
HO01`4Thr3 AcHN
In certain embodiments, the cell-targeting moiety comprises:

HO OH H 0 i¨/OH
N,(-)1---N___I
HO 0 \--0 AcHN I
0=P-OH
I

HO OH H 0 r---/
.1.,(2.svOrN)1--N----\
HO_ 0 L-0 AcHN I
0=P-OH
I

HO OH H 0 r--/
_...f,2..\/OrN'f*)1-3 NI
HO 0 "------0-/
AcHN .
In certain embodiments, the cell-targeting moiety has the following structure:
OH H
01-1.1........ N 0 OH
OH
OH____\...._...\vNHAc H NH
N cs.rc OH
OH 0 .,--_---- 0 NHAc OH
OH o s-' ....T.......
n..---,,cr-....õ.,..NH

NHAc .
In certain embodiments, the cell-targeting moiety comprises:
01 H r OH

HO ----:)..\- 0 AcHN NH
01 H r OH

HO ----)-1--0 N N y AcHN

=
In certain embodiments, the cell-targeting moiety has the following structure:

OH OH
AcHN =
In certain embodiments, the cell-targeting moiety comprises:
OH
HO...7.......

HO 00,p,0 AcHN o"\
P\

0¨K
FICkcHN =
, wherein each Y is selected from 0, S, a substituted or unsubstituted C1-C10 alkyl, amino, substituted amino, azido, alkenyl or alkynyl.
In certain embodiments, the conjugate group comprises:
H? eH
0õY
AcHN 0 Y ¨(:)õ0 .%1"
P\ 0 rsss 0 e Y
0¨K
OH _x¨r-j 8 Y
FICkcHN =
, wherein each Y is selected from 0, S, a substituted or unsubstituted C1-C10 alkyl, amino, substituted amino, azido, alkenyl or alkynyl.
In certain embodiments, the cell-targeting moiety has the following structure:

OH
HO

HO
AcHN
0 Y =
wherein each Y is selected from 0, S, a substituted or unsubstituted C1-C10 alkyl, amino, substituted amino, azido, alkenyl or alkynyl.
In certain embodiments, the conjugate group comprises:
OH

HO
AcHN
OH
In certain embodiments, the conjugate group comprises:
OH

0 ..µµO
HO H
AcHN
T In certain embodiments, the conjugate group comprises:
OH
HO

HO 0 0õ,...____,õõ_õõ)c AcHN

OH
In certain embodiments, the conjugate group comprises:
OH

HO /\./\./Ic NrN?
AcHN

In certain embodiments, the conjugate group comprises a cleavable moiety selected from among: a phosphodiester, an amide, or an ester.
In certain embodiments, the conjugate group comprises a phosphodiester cleavable moiety.
In certain embodiments, the conjugate group does not comprise a cleavable moiety, and wherein the conjugate group comprises a phosphorothioate linkage between the conjugate group and the oligonucleotide.
In certain embodiments, the conjugate group comprises an amide cleavable moiety.
In certain embodiments, the conjugate group comprises an ester cleavable moiety.
In certain embodiments, the compound has the following structure:
HOOF!

HO ___________________ -7.....\.0 n 0 1 0 AcHN
OH ) HOOH 1n 0 0 Bx II u 0 HO¨P=0 Q13 (-- )n 1 HO "n OH A
NHAc wherein each n is, independently, from 1 to 20;
Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.

In certain embodiments, the compound has the following structure:
HO OH
0 n 0 HO-4...\vki.,---õ,--N...,,N
..,K
AcHN 0 10 OH ---HO OH 0 0, 0 IIec)"4 0 Bx OH ==
AcHN OH (:) 0' Q13 HO-P=0 HO H 9 y P, I
A
1.2...\/0 OH
HO
NHAc wherein each n is, independently, from 1 to 20;
Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.

In certain embodiments, the compound has the following structure:
A
I
HO¨P=0 I
0 ________________________________________________________________ ,c0.Bx ='s Q13 I
HO¨P=0 I

HO OH (411 0 , 0 'n AcHN OH 1) 0 \
Z
HO OH n (On HO--="2...\VC1/ ,P, " 0 0 , I
n 0 1 0----;¨e ______________________________________ z0¨P=0 AcHN OH I _____ OH

HO......o....\/ H jc_o_c )n HO OH
NHAc wherein each n is, independently, from 1 to 20;
Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide;
Z is H or a linked solid support; and Bx is a heterocyclic base moiety.

In certain embodiments, the compound has the following structure:
A
I
HO¨P=0 I
0 ____________________________________________________________ -.,c OzoBx I
HO¨P=0 O
(3 AcHN

()3 0 0-, I
ii HOO p O¨P=0 W 1 (i)07 I
AcHN OH (:) OH

HO H
P-OH
NHAc wherein each n is, independently, from 1 to 20;
Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide;
Z is H or a linked solid support; and Bx is a heterocyclic base moiety.

In certain embodiments, the compound has the following structure:
OH OH
HO.r.C.L0 AcHN N-----N---NH
OH
N
HO:)µ___ \-:_----0 0\_......\___ 0 AcHN )-- /C) N----,./.."--N
N
H H 11 'El\II-WC) H 6 HO-P=0 if N \(:) 0 -0Bx H
cis Q13 I

H0/(7:)...v HO-P=0 I
HO A
NHAC
wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide;and Bx is a heterocyclic base moiety.
In certain embodiments, the compound has the following structure:
OH OH

H0.7.2.__ -"---LNH
AcHN
OH OH
H0*.... 0 ,crEi 0 H , 0 0,........---,õ..".....)t, N ......õ..õ...õ.õõThrN-,---y6 NNN
AcHN 11 0 il 0 HO-P=0 I
0 r 0-cOrBx C' Q13 I
HO HO-P=0 NHAc I
A , wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide;and Bx is a heterocyclic base moiety.

In certain embodiments, the compound has the following structure:

HO "ioH
AcHN N

__..T.C2..\cN,,.-NN)-( N n 0 HO "io I-1 Z H H 4 N
HO-P=0 AcHN 0 I
HOOH
--C1 0OrBx CZ" Q13 NHAc io HO-P=0 I
A, wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
In certain embodiments, the compound has the following structure:

HOOH
0 oi_pi AcHN 0 0 0, HOOH r(3,-0111yo' (31\
o o' HO-P=0 HO---7*--\, rN I
Bx AcHN 0 HO g CI::..:)...v1-1 HO-P=0 (51 s Q13 I

NHAc wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.

In certain embodiments, the compound has the following structure:
A
I
HO-P=0 I
0--....,c0rBx HO OH 0 0 cis' Q13 I
HO-P=0 HO--1.24 HN ')2 I-IN \
/
AcHN
HO OH 0 N .(-N-,,N,1-LNL......
HO--72-\/ 11 2 0 H
AcH N OH

HO
0N^Ã,N(0 AcHN
wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
In certain embodiments, the compound has the following structure:

AcHN H 0 0 HOoN H H 4 I
3H 0 HO-P=0 AcHN

( I
0¨(0z.Bx I
AcHN HO-P=0 I
A
wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.

In certain embodiments, the compound has the following structure:

AcHN 0 0 11'40\
4 H HO-P=0 AcHN
HOOH 0¨cOrBx HO 4 H (1 Q13 AcHN HO-P=0 A
wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
In certain embodiments, the compound has the following structure:
pH
HOOH
HO-Z/C)Nr?._ AcHN
0=P-OH
HOOH
HO

AcHN
0=P-OH A
g HO-P=0 HOOH 0I¨.,(Oz.Bx AcHN I
P=0 OH
wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.

In certain embodiments, the compound has the following structure:
pH
HOOH 0 =
HO

AcHN
0=P-OH

HO--re-\/ 113I, N

AcHN
0=P-OH
A
HOOH H 0 _____ HO-p=0 HO-4)--\/ 1Thr 0 3 0 N N 0¨(0z.13x AcHN Cs' Q13 I
P=0 OH
wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.

In certain embodiments, the compound has the following structure:
HooH ("OH
H 0 --12--\/
N
3 0 \---P
AcHN
0=P-OH
"DI
HO OH
H 0 --"1"(:-)./ 1rx /3 0 N
AcHN
0=P-OH A
HO-p=0 HO OH 0 Bx HO01,1Thr N
3 0 0 d" Q13 AcHN I
P=0 OH
wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
In certain embodiments, the compound has the following structure:
OH

AcHN
0=P-OH

HON

AcHN
0=P-OH A
0 HO-P=0 0¨(0z.Bx HO-112-\/ TnrN

AcHN
P=0 OH
wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
In certain embodiments, the conjugate group comprises:
HO OH

AcHN N--N--)/--Nx--\NH
0y----1 o 0 H H 0 0, ).1).L 9 0 Bx N---n----N---0.--- ¨
-NH N-(CH2)6-0p-0 HO H H
(\i)..\/0 OH ==

HO-' ) HO-P=0 NHAc Al HN

OH
HO
HO\C\)'>/
NHAc wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
In certain embodiments, the conjugate group comprises:

HO "4 H
AcHN NON o HO_....72.....\/0--'fri. --1\r("rO 0 H H
4,(ClyBx AcHN 0 OH

== ______________________________________________________________ ( HO OH
--C HO-P=0 I
crifi. 11 0 AcHN
wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
In certain embodiments, the conjugate group comprises:

HOOH
AcHN

HO-79--\/ (-CNN)LNO-P-0 4 H 4 (i)tHx AcHN OH0 _______ (Q13 HOOH
HO-P=0 A
HO "4 AcHN
wherein Q13 is H or 0(CH2)2-0CH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
In certain embodiments, Bx is selected from among from adenine, guanine, thymine, uracil, or cytosine, or 5-methyl cytosine. In certain embodiments, Bx is adenine. In certain embodiments, Bx is thymine.
In certain embodiments, Q13 is 0(CH2)2-0CH3. In certain embodiments, Q13 is H.
Certain embodiments of the invention provide a prodrug comprising the compositions or compounds disclosed herein.
In certain embodiments, the compound is in a salt form. In further embodiments, the compound further comprises of a pharmaceutically acceptable carrier or diluent. In certain embodiments, the compound comprises a modified oligonucleotide targeting ApoCIII and a conjugate group, or a salt thereof, and a pharmaceutically acceptable carrier or diluent.
Certain embodiments provide compositions and methods comprising administering to an animal a conjugated antisense compound or composition disclosed herein. In certain embodiments, administering the conjugated antisense compound prevents, treats, ameliorates, or slows progression of a cardiovascular, metabolic and/or inflammatory disease.
Certain embodiments provide compositions and methods for use in therapy to treat an ApoCIII
related disease, disorder or condition. In certain embodiments, the ApoCIII
levels are elevated in an animal.
In certain embodiments, the composition is a compound comprising an ApoCIII
specific inhibitor. In certain embodiments, the ApoCIII specific inhibitor is a nucleic acid. In certain embodiments, the nucleic acid is an antisense compound. In certain embodiments, the antisense compound is a modified oligonucleotide targeting ApoCIII. In certain embodiments, the antisense compound is a modified oligonucleotide targeting ApoCIII
and a conjugate group. In certain embodiments, the modified oligonucleotide targeting ApoCIII with the conjugate group, is used in treating, preventing, slowing progression, ameliorating an inflammatory, cardiovascular and/or metabolic disease, disorder or condition. In certain embodiments, the compositions and methods for therapy include administering an ApoCIII specific inhibitor to an individual in need thereof Certain embodiments provide conjugated antisense compounds and compositions and methods for reducing ApoCIII levels. In certain embodiments, ApoCIII levels are reduced in the liver, adipose tissue, heart, skeletal muscle or small intestine.
In certain embodiments, reducing ApoCIII levels in a tissue, organ or subject increases HDL levels.
In certain embodiments, the HDL levels are increased by at least 90%, by at least 80%, by at least 70%, by at least 60%, by at least 50%, by at least 45%, at least 40%, by at least 35%, by at least 30%, by at least 25%, by at least 20%, by at least 15%, by at least 10% or by at least 5% from the baseline HDL level.
In certain embodiments, reducing ApoCIII levels in a tissue, organ or subject reduces TG levels. In certain embodiments, the subject has a triglyceride level > 100 mg/dL, > 200 mg/dL, > 300 mg/dL, > 400 mg/dL, > 440 mg/dL, > 500 mg/dL, > 600 mg/dL, > 700 mg/dL, > 800 mg/dL, > 880 mg/dL, > 900 mg/dL, >
1000 mg/dL, > 1100 mg/dL, > 1200 mg/dL, > 1300 mg/dL, > 1400 mg/dL, > 1500 mg/dL, > 1600 mg/dL, >
1700 mg/dL, > 1800 mg/dL, > 1900 mg/dL, > 2000 mg/dL.
In certain embodiments, the TG levels (postprandial or fasting) are decreased by at least 90%, by at least 80%, by at least 70%, by at least 60%, by at least 50%, by at least 45%, at least 40%, by at least 35%, by at least 30%, by at least 25%, by at least 20%, by at least 15%, by at least 10%, by at least 5% or by at least 1% from the baseline TG level. In certain embodiments, the TG (postprandial or fasting) level is decreased to <1900mg/dL, <1800mg/dL, <1700mg/dL, <1600mg/dL, <1500mg/dL, <1400mg/dL, <1300mg/dL, <1200mg/dL, <1100mg/dL, <1000mg/dL, <900mg/dL, <800mg/dL, <750mg/dL, <700mg/dL, <650mg/dL, <600mg/dL, <550mg/dL, <500mg/dL, <450mg/dL, <400mg/dL, <350mg/dL, <300mg/dL, <250mg/dL, <200mg/dL, <150mg/dL or <100mg/dL.
In certain embodiments, reducing ApoCIII levels in a tissue, organ or subject improves the ratio of LDL to HDL or the ratio of TG to HDL.
In certain embodiments, reducing ApoCIII levels in a tissue, organ or subject improves insulin sensitivity.
In certain embodiments, reducing ApoCIII levels in a tissue, organ or subject increases chylomicron clearance.
Certain embodiments provide compositions and methods to reduce ApoCIII mRNA or protein expression in an animal comprising administering to the animal a conjugated antisense compound or composition disclosed herein to reduce ApoCIII mRNA or protein expression in the animal.
Certain embodiments provide conjugated antisense compounds and compositions and methods for preventing, treating, delaying, slowing the progression and/or ameliorating ApoCIII related diseases, disorders, and conditions in a subject in need thereof In certain embodiments, such diseases, disorders, and conditions include inflammatory, cardiovascular and/or metabolic diseases, disorders, and conditions. Certain such cardiovascular diseases, disorders or conditions include, but are not limited to, chylomicronemia, hypertriglyceridemia, aortic stenosis, aneurysm (e.g., abdominal aortic aneurysm), angina, arrhythmia, atherosclerosis, cerebrovascular disease, coronary artery disease, coronary heart disease, dyslipidemia, hypercholesterolemia, hyperlipidemia, hypertension, myocardial infarction, peripheral vascular disease (e.g., peripheral artery disease, peripheral artery occlusive disease), Fredrickson Type I dyslipidemia, FCS, LPL
deficiency, retinal vascular occlusion, or stroke. Certain such metabolic diseases, disorders or conditions include, but are not limited to, hyperglycemia, prediabetes, diabetes (type I
and type II), obesity, insulin resistance, metabolic syndrome and diabetic dyslipidemia. Certain such inflammatory diseases, disorders or conditions include, but are not limited to, pancreatitis, aortic stenosis, coronary artey disease (CAD), Alzheimer's Disease and thromb oemb olic diseases, disorder or conditions.
Certain thromb oemb olic diseases, disorders or conditions include, but are not limited to, stroke, thrombosis (e.g., venous thromboembolism), myocardial infarction and peripheral vascular disease. Certain embodiments provide conjugated antisense compounds and compositions and methods for preventing, treating, delaying, slowing the progression and/or ameliorating hypertriglyceridemia. Certain embodiments provide conjugated antisense compounds and compositions and methods for preventing, treating, delaying, slowing the progression and/or ameliorating chylomicronemia. Certain embodiments provide conjugated antisense compounds and compositions and methods for preventing, treating, delaying, slowing the progression and/or ameliorating pancreatitis.
Certain embodiments provide a method of reducing at least one symptom of a cardiovascular disease, disorder or condition. In certain embodiments, the symptoms include, but are not limited to, angina, chest pain, shortness of breath, palpitations, weakness, dizziness, nausea, sweating, tachycardia, bradycardia, arrhythmia, atrial fibrillation, swelling in the lower extremities, cyanosis, fatigue, fainting, numbness of the face, numbness of the limbs, claudication or cramping of muscles, bloating of the abdomen, and fever. In certain embodiments, symptoms of a metabolic disease, disorder or condition include, but are not limited to, frequent urination, unusual thirst, extreme hunger, unusual weight loss, extreme fatigue, iIrritability, frequent infections, blurred vision, cuts/bruises that are slow to heal, tingling/numbness in the hands/feet and recurring skin, gum, or bladder infections. Certain embodiments provide a method of reducing at least one symptom of hypertriglyceridemia. Certain embodiments provide a method of reducing at least one symptom of chylomicronemia. Certain embodiments provide a method of reducing at least one symptom of pancreatitis.
In certain embodiments, the modulation of ApoCIII expression occurs in a cell, tissue or organ. In certain embodiments, the modulations occur in a cell, tissue or organ in an animal. In certain embodiments, the modulation is a reduction in ApoCIII mRNA level. In certain embodiments, the modulation is a reduction in ApoCIII protein level. In certain embodiments, both ApoCIII mRNA and protein levels are reduced. Such reduction may occur in a time-dependent or in a dose-dependent manner.
In certain embodiments, the subject or animal is human.
In certain embodiments, the compound is parenterally administered. In further embodiments, the parenteral administration is subcutaneous.

In certain embodiments, the conjugated antisense compound or composition is co-administered with a second agent or therapy. In certain embodiments, the conjugated antisense compound or composition and the second agent are administered concomitantly.
In certain embodiments, the second agent is a glucose-lowering agent. In certain embodiments, the second agent is a LDL, TG or cholesterol lowering agent. In certain embodiments, the second agent is an anti-inflammatory agent. In certain embodiments, the second agent is an Alzheimer Disease drug. In certain embodiments, the second agent can be, but is not limited to, a non-steroidal anti-inflammatory drug (NSAID
e.g., aspirin), niacin (e.g., Niaspan), nicotinic acid, an apoB inhibitor (e.g., Mipomersen), a CETP inhibitor (e.g., Anacetrapib), an apo(a) inhibitor, a thyroid hormone analog (e.g., Eprotirome), a HMG-CoA reductase inhibitor (e.g., a statin), a fibrate (e.g., Gemfibrozil) and an microsomal triglyceride transfer protein inhibitor (e.g., Lomitapide). Agents or therapies can be co-administered or administered concomitantly. Agents or therapies can be sequentially or subsequently administered.
Certain embodiments provide use of the compositions and conjugated antisense compounds described herein targeted to ApoCIII for decreasing ApoCIII levels in an animal. Certain embodiments provide use of a compound targeted to ApoCIII for decreasing ApoCIII levels in an animal. Certain embodiments provide use of a compound targeted to ApoCIII for increasing HDL
levels in an animal. Certain embodiments provide use of a compound targeted to ApoCIII for increasing HDL
chylomicron clearance in an animal. Certain embodiments provide use of a compounds targeted to ApoCIII
for the treatment, prevention, or amelioration of a disease, disorder, or condition associated with ApoCIII. Certain embodiments provide use of a compound targeted to ApoCIII for the treatment, prevention, or amelioration of a hypertriglyceridemia. Certain embodiments provide use of a compound targeted to ApoCIII for the treatment, prevention, or amelioration of a chylomicronemia (e.g., FCS and/or LPLD). Certain embodiments provide use of a compound targeted to ApoCIII for the treatment, prevention, or amelioration of a pancreatitis.
Certain embodiments provide use of the compositions and conjugated antisense compounds described herein targeted to ApoCIII in the preparation of a medicament for decreasing ApoCIII levels in an animal. Certain embodiments provide use of the compositions and compounds for the preparation of a medicament for the treatment, prevention, or amelioration of a disease, disorder, or condition associated with ApoCIII.
Certain embodiments provide the use of the compositions and conjugated antisense compounds as described herein in the manufacture of a medicament for treating, ameliorating, delaying or preventing one or more of a disease related to ApoCIII.
Certain embodiments provide a kit for treating, preventing, or ameliorating a disease, disorder or condition as described herein wherein the kit comprises: (i) an ApoCIII
specific inhibitor as described herein;
and optionally (ii) a second agent or therapy as described herein.

A kit of the present invention can further include instructions for using the kit to treat, prevent, or ameliorate a disease, disorder or condition as described herein by combination therapy as described herein.
B. Certain Compounds In certain embodiments, the invention provides conjugated antisense compounds comprising antisense oligonucleoitdes and a conjugate.
a. Certain Antisense Oligonucleotides In certain embodiments, the invention provides antisense oligonucleotides.
Such antisense oligonucleotides comprise linked nucleosides, each nucleoside comprising a sugar moiety and a nucleobase.
The structure of such antisense oligonucleotides may be considered in terms of chemical features (e.g., modifications and patterns of modifications) and nucleobase sequence (e.g., sequence of antisense oligonucleotide, idenity and sequence of target nucleic acid).
i. Certain Chemistry Features In certain embodiments, antisense oligonucleotide comprise one or more modification. In certain such embodiments, antisense oligonucleotides comprise one or more modified nucleosides and/or modified internucleoside linkages. In certain embodiments, modified nucleosides comprise a modifed sugar moirty and/or modifed nucleobase.
1. Certain Sugar Moieties In certain embodiments, compounds of the disclosure comprise one or more modifed nucleosides comprising a modifed sugar moiety. Such compounds comprising one or more sugar-modified nucleosides may have desirable properties, such as enhanced nuclease stability or increased binding affinity with a target nucleic acid relative to an oligonucleotide comprising only nucleosides comprising naturally occurring sugar moieties. In certain embodiments, modified sugar moieties are substitued sugar moieties. In certain embodiments, modified sugar moieties are sugar surrogates. Such sugar surrogates may comprise one or more substitutions corresponding to those of substituted sugar moieties.
In certain embodiments, modified sugar moieties are substituted sugar moieties comprising one or more non-bridging sugar substituent, including but not limited to substituents at the 2' and/or 5' positions.
Examples of sugar substituents suitable for the 2'-position, include, but are not limited to: 2'-F, 2'-OCH3 ("OMe" or "0-methyl"), and 2'-0(CH2)20CH3("MOE"). In certain embodiments, sugar substituents at the 2' position is selected from allyl, amino, azido, thio, 0-allyl, 0-C1-C10 alkyl, 0-C1-C10 substituted alkyl; OCF3, 0(CH2)25CH3, 0(CH2)2-0-N(Rm)(R4 and 0-CH2-C(=0)-N(Rm)(Rn), where each Rm and Rn is, independently, H or substituted or unsubstituted C1-C10 alkyl. Examples of sugar substituents at the 5'-position, include, but are not limited to:, 5'-methyl (R or S); 5'-vinyl, and 5'-methoxy. In certain embodiments, substituted sugars comprise more than one non-bridging sugar substituent, for example, 2'-F-5'-methyl sugar moieties (see,e.g., PCT International Application WO
2008/101157, for additional 5', 2'-bis substituted sugar moieties and nucleosides).
Nucleosides comprising 2'-substituted sugar moieties are referred to as 2'-substituted nucleosides. In certain embodiments, a 2'- substituted nucleoside comprises a 2'-substituent group selected from halo, allyl, amino, azido, SH, CN, OCN, CF3, OCF3, 0, S, or N(Rm)-alkyl; 0, S, or N(Rm)-alkenyl; 0, S or N(Rm)-alkynyl; 0-alkyleny1-0-alkyl, alkynyl, alkaryl, aralkyl, 0-alkaryl, 0-aralkyl, 0(CH2)2SCH3, 0-(CH2)2-0-N(Rm)(Rii) or 0-CH2-C(=0)-N(Rm)(Rii), where each Rm and Rn is, independently, H, an amino protecting group or substituted or unsubstituted C1-C10 alkyl. These 2'-substituent groups can be further substituted with one or more substituent groups independently selected from hydroxyl, amino, alkoxy, carboxy, benzyl, phenyl, nitro (NO2), thiol, thioalkoxy (S-alkyl), halogen, alkyl, aryl, alkenyl and alkynyl.
In certain embodiments, a 2'- substituted nucleoside comprises a 2'-substituent group selected from F, NH2, N3, OCF3, 0-CH3, 0(CH2)3NH2, CH2-CH=CH2, 0-CH2-CH=CH2, OCH2CH2OCH3, 0(CH2)2SCH3, 0-(CH2)2-0-N(Rm)(R.), 0(CH2)20(CH2)2N(CH3)2, and N-substituted acetamide (0-CH2-C(=0)-N(Rm)(RO
where each Rm and Rn is, independently, H, an amino protecting group or substituted or unsubstituted C1-C10 alkyl.
In certain embodiments, a 2'- substituted nucleoside comprises a sugar moiety comprising a 2'-substituent group selected from F, OCF3, 0-CH3, OCH2CH2OCH3, 0(CH2)25CH3, 0-(CH2)2-0-N(CH3)2, -0(CH2)20(CH2)2N(CH3)2, and 0-CH2-C(=0)-N(H)CH3.
In certain embodiments, a 2'- substituted nucleoside comprises a sugar moiety comprising a 2'-substituent group selected from F, 0-CH3, and OCH2CH2OCH3.
Certain modifed sugar moieties comprise a bridging sugar substituent that forms a second ring resulting in a bicyclic sugar moiety. In certain such embodiments, the bicyclic sugar moiety comprises a bridge between the 4' and the 2' furanose ring atoms. Examples of such 4' to 2' sugar substituents, include, but are not limited to: -[C(RO(Rb)in-, -[C(RO(Rb)].-0-, -C(RaRb)-N(R)-0- or, -C(RaRb)-0-N(R)-; 4'-CH2-2', 4'-(CH2)2-2', 4'-(CH2)3-2',. 4'-(CH2)-0-2' (LNA); 4'-(CH2)-S-2'; 4'-(CH2)2-0-2' (ENA); 4'-CH(CH3)-0-2' (cEt) and 4'-CH(CH2OCH3)-0-2',and analogs thereof (see, e.g., U.S. Patent 7,399,845, issued on July 15, 2008); 4'-C(CH3)(CH3)-0-2'and analogs thereof, (see, e.g., W02009/006478, published January 8, 2009); 4'-CH2-N(OCH3)-2' and analogs thereof (see, e.g., W02008/150729, published December 11, 2008); 4'-CH2-0-N(CH3)-2' (see, e.g., U52004/0171570, published September 2, 2004); 4'-CH2-0-N(R)-2', and 4'-CH2-N(R)-0-2'-, wherein each R is, independently, H, a protecting group, or C1-C12 alkyl; 4'-CH2-N(R)-0-2', wherein R
is H, C1-C12 alkyl, or a protecting group (see, U.S. Patent 7,427,672, issued on September 23, 2008); 4'-CH2-C(H)(CH3)-2' (see, e.g., Chattopadhyaya, et al., J. Org. Chem.,2009, 74, 118-134); and 4'-CH2-C(=CH2)-2' and analogs thereof (see, published PCT International Application WO
2008/154401, published on December 8, 2008).

In certain embodiments, such 4' to 2' bridges independently comprise from 1 to 4 linked groups independently selected from -[C(Ra)(Rb)]a-, -C(Ra)=C(Rb)-, -C(Ra)=N-, -C(=NRa)-, -C(=0)-, -C(=S)-, -0-, -Si(Ra)27, -S(=0)õ-, and -N(Ra)-;
wherein:
x is 0, 1, or 2;
n is 1, 2, 3, or 4;
each Ra and Rb is, independently, H, a protecting group, hydroxyl, CI-Cu.
alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C5-C20 aryl, substituted C5-C20 aryl, heterocycle radical, substituted heterocycle radical, heteroaryl, substituted heteroaryl, C5-C7 alicyclic radical, substituted C5-C7 alicyclic radical, halogen, OJI, NJ1J2, SJI, N3, COOJI, acyl (C(=0)-H), substituted acyl, CN, sulfonyl (S(=0)2-Ji), or sulfoxyl (S(=0)-Ji); and each J1 andJ2 is, independently, H, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C5-C20 aryl, substituted C5-C20 aryl, acyl (C(=0)-H), substituted acyl, a heterocycle radical, a substituted heterocycle radical, C1-C12 aminoalkyl, substituted C1-C12 aminoalkyl, or a protecting group.
Nucleosides comprising bicyclic sugar moieties are referred to as bicyclic nucleosides or BNAs.
Bicyclic nucleosides include, but are not limited to, (A) a-L-Methyleneoxy (4'-CH2-0-2') BNA, (B) 13-D-Methyleneoxy (4'-CH2-0-2') BNA (also referred to as locked nucleic acid or LNA) , (C) Ethyleneoxy (4'-(CH2)2-0-2') BNA, (D) Aminooxy (4'-CH2-0-N(R)-2') BNA, (E) Oxyamino (4'-CH2-N(R)-0-2') BNA, (F) Methyl(methyleneoxy) (4'-CH(CH3)-0-2') BNA (also referred to as constrained ethyl or cEt), (G) methylene-thio (4' -CH2-S-2') BNA, (H) methylene-amino (4'-CH2-N(R)-2') BNA, (I) methyl carbocyclic (4'-CH2-CH(CH3)-2') BNA, and (J) propylene carbocyclic (4'-(CH2)3-2') BNA as depicted below.
_______________________________ 0 Bx 0 Bx Bx u ¨0 ¨ ¨0 (A) (B) (C) ______________ 0 Bx 0 Bx 0 H3C _________________ Bx (D) (E) (F) Bx 0 y Bx 0 Bx (G) R . n3 (1) 0, Bx (J) wherein Bx is a nucleobase moiety and R is, independently, H, a protecting group, or C1-C12 alkyl.
Additional bicyclic sugar moieties are known in the art, for example: Singh et al., Chem. Commun., 1998, 4, 455-456; Koshkin et al., Tetrahedron, 1998, 54, 3607-3630; Wahlestedt et al., Proc. Natl. Acad. Sci.
U. S. A., 2000, 97, 5633-5638; Kumar et al., Bioorg. Med. Chem. Lett., 1998, 8, 2219-2222; Singh et al., J.
Org. Chem., 1998, 63, 10035-10039; Srivastava et al., J. Am. Chem. Soc., 129(26) 8362-8379 (Jul. 4, 2007);
Elayadi et al., Curr. Opinion Invens. Drugs, 2001, 2, 558-561; Braasch et al., Chem. Biol., 2001, 8, 1-7;
Orum et al., Curr. Opinion Mot. Ther., 2001, 3, 239-243; U.S. Patent Nos.
7,053,207, 6,268,490, 6,770,748, 6,794,499, 7,034,133, 6,525,191, 6,670,461, and 7,399,845; WO 2004/106356, WO
1994/14226, WO
2005/021570, and WO 2007/134181; U.S. Patent Publication Nos. U52004/0171570, U52007/0287831, and U52008/0039618; U.S. Patent Serial Nos. 12/129,154, 60/989,574, 61/026,995, 61/026,998, 61/056,564, 61/086,231, 61/097,787, and 61/099,844; and PCT International Applications Nos. PCT/U52008/064591, PCT/U52008/066154, and PCT/U52008/068922.
In certain embodiments, bicyclic sugar moieties and nucleosides incorporating such bicyclic sugar moieties are further defined by isomeric configuration. For example, a nucleoside comprising a 4'-2' methylene-oxy bridge, may be in the a-L configuration or in the P-D
configuration. Previously, a-L-methyleneoxy (4'-CH2-0-2') bicyclic nucleosides have been incorporated into antisense oligonucleotides that showed antisense activity (Frieden et al., Nucleic Acids Research, 2003, 21, 6365-6372).
In certain embodiments, substituted sugar moieties comprise one or more non-bridging sugar substituent and one or more bridging sugar substituent (e.g., 5'-substituted and 4'-2' bridged sugars). (see, PCT International Application WO 2007/134181, published on 11/22/07, wherein LNA is substituted with, for example, a 5'-methyl or a 5'-vinyl group).
In certain embodiments, modified sugar moieties are sugar surrogates. In certain such embodiments, the oxygen atom of the naturally occuring sugar is substituted, e.g., with a sulfer, carbon or nitrogen atom. In certain such embodiments, such modified sugar moiety also comprises bridging and/or non-bridging substituents as described above. For example, certain sugar surrogates comprise a 4'-sulfer atom and a substitution at the 2'-position (see,e.g., published U.S. Patent Application U52005/0130923, published on June 16, 2005) and/or the 5' position. By way of additional example, carbocyclic bicyclic nucleosides having a 4'-2' bridge have been described (see, e.g., Freier et al., Nucleic Acids Research, 1997, 25(22), 4429-4443 and Albaek et al., J. Org. Chem., 2006, 7/, 7731-7740).
In certain embodiments, sugar surrogates comprise rings having other than 5-atoms. For example, in certain embodiments, a sugar surrogate comprises a morphlino. Morpholino compounds and their use in oligomeric compounds has been reported in numerous patents and published articles (see for example:
Braasch et al., Biochemistry, 2002, 41, 4503-4510; and U.S. Patents 5,698,685;
5,166,315; 5,185,444; and 5,034,506). As used here, the term "morpholino" means a sugar surrogate having the following structure:
In certain embodiments, morpholinos may be modified, for example by adding or altering various substituent groups from the above morpholino structure. Such sugar surrogates are refered to herein as "modifed morpholinos."
For another example, in certain embodiments, a sugar surrogate comprises a six-membered tetrahydropyran. Such tetrahydropyrans may be further modified or substituted.
Nucleosides comprising such modified tetrahydropyrans include, but are not limited to, hexitol nucleic acid (HNA), anitol nucleic acid (ANA), manitol nucleic acid (MNA) (see Leumann, CJ. Bioorg. & Med. Chem.
(2002) 10:841-854), fluoro HNA (F-HNA), and those compounds having Formula VI:
C11 Cl2 C16 Bx /o R1 R2CI5 vT
wherein independently for each of said at least one tetrahydropyran nucleoside analog of Formula VI:
Bx is a nucleobase moiety;
T3 and T4 are each, independently, an internucleoside linking group linking the tetrahydropyran nucleoside analog to the antisense compound or one of T3 and T4 is an internucleoside linking group linking the tetrahydropyran nucleoside analog to the antisense compound and the other of T3 and T4 is H, a hydroxyl protecting group, a linked conjugate group, or a 5' or 3'-terminal group;
qi, q2, q3, q4, q5, q6 and (17 are each, independently, H, Ci-C6 alkyl, substituted Ci-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl, or substituted C2-C6 alkynyl; and each of R1 and R2 is independently selected from among: hydrogen, halogen, substituted or unsubstituted alkoxy, NJ1J2, SJI, N3, OC(=X)Ji, OC(=X)NJ1J2, NJ3C(=X)NJ1J2, and CN, wherein X is 0, S or NJI, and each J1, J2, and J3 is, independently, H or C1-C6 alkyl.
In certain embodiments, the modified THP nucleosides of Formula VI are provided wherein qi, q2, q3, q4, q5, q6 and q7 are each H. In certain embodiments, at least one of qi, q2, q3, q4, q5, q6 and (47 is other than H. In certain embodiments, at least one of qi, q2, q3, q4, q5, q6 and q7 is methyl. In certain embodiments, THP
nucleosides of Formula VI are provided wherein one of R1 and R2 is F. In certain embodiments, R1 is fluoro and R2 is H, R1 is methoxy and R2 is H, and R1 is methoxyethoxy and R2 is H.
Many other bicyclo and tricyclo sugar surrogate ring systems are also known in the art that can be used to modify nucleosides for incorporation into antisense compounds (see, e.g., review article: Leumann, J.
C, Bioorganic & Medicinal Chemistry, 2002, 10, 841-854).
Combinations of modifications are also provided without limitation, such as 2'-F-5'-methyl substituted nucleosides (see PCT International Application WO 2008/101157 Published on 8/21/08 for other disclosed 5', 2'-bis substituted nucleosides) and replacement of the ribosyl ring oxygen atom with S and further substitution at the 2'-position (see published U.S. Patent Application US2005-0130923, published on June 16, 2005) or alternatively 5'-substitution of a bicyclic nucleic acid (see PCT International Application WO 2007/134181, published on 11/22/07 wherein a 4'-CH2-0-2' bicyclic nucleoside is further substituted at the 5' position with a 5'-methyl or a 5'-vinyl group). The synthesis and preparation of carbocyclic bicyclic nucleosides along with their oligomerization and biochemical studies have also been described (see, e.g., Srivastava et al., J. Am. Chem. Soc. 2007, 129(26), 8362-8379).
In certain embodiments, the present disclosure provides oligonucleotides comprising modified nucleosides.
Those modified nucleotides may include modified sugars, modified nucleobases, and/or modified linkages.
The specific modifications are selected such that the resulting oligonucleotides possess desireable characteristics. In certain embodmiments, oligonucleotides comprise one or more RNA-like nucleosides. In certain embodiments, oligonucleotides comprise one or more DNA-like nucleotides.
2. Certain Nucleobase Modifications In certain embodiments, nucleosides of the present disclosure comprise one or more unmodified nucleobases. In certain embodiments, nucleosides of the present disclosure comprise one or more modifed nucleobases.
In certain embodiments, modified nucleobases are selected from: universal bases, hydrophobic bases, promiscuous bases, size-expanded bases, and fluorinated bases as defined herein. 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil; 5-propynylcytosine; 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 CH3) 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, 3-deazaguanine and 3-deazaadenine, universal bases, hydrophobic bases, promiscuous bases, size-expanded bases, and fluorinated bases as defined herein. Further modified nucleobases include tricyclic pyrimidines such as phenoxazine cytidine( [5,4-b][1,4]benzoxazin-2(3H)-one), phenothiazine cytidine (1H-pyrimido[5,4-b][1,4]benzothiazin-2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g. 9-(2-aminoethoxy)-H-pyrimido[5,4-b][1,4]benzoxazin-2(3H)-one), carbazole cytidine (2H-pyrimido[4,5-b]indo1-2-one), pyridoindole cytidine (H-pyrido[3',2':4,5]pyrrolo[2,3-d]pyrimidin-2-one). Modified nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Further nucleobases include those disclosed in United States Patent No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, Kroschwitz, J.I., Ed., John Wiley &
Sons, 1990, 858-859; those disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613; and those disclosed by Sanghvi, Y.S., Chapter 15, Antisense Research and Applications, Crooke, S.T.
and Lebleu, B., Eds., CRC Press, 1993, 273-288.
Representative United States patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include without limitation, U.S. 3,687,808; 4,845,205;
5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255;
5,484,908; 5,502,177;
5,525,711; 5,552,540; 5,587,469; 5,594,121; 5,596,091; 5,614,617; 5,645,985;
5,681,941; 5,750,692;
5,763,588; 5,830,653 and 6,005,096, certain of which are commonly owned with the instant application, and each of which is herein incorporated by reference in its entirety.
3. Certain Internucleoside Linkages In certain embodiments, the present disclosure provides oligonucleotides comprising linked nucleosides. In such embodiments, nucleosides may be linked together using any internucleoside linkage.
The two main classes of internucleoside linking groups are defined by the presence or absence of a phosphorus atom. Representative phosphorus containing internucleoside linkages include, but are not limited to, phosphodiesters (PO), phosphotriesters, methylphosphonates, phosphoramidate, and phosphorothioates (PS). Representative non-phosphorus containing internucleoside linking groups include, but are not limited to, methylenemethylimino (-CH2-N(CH3)-0-CH2-), thiodiester (-0-C(0)-S-), thionocarbamate (-0-C(0)(NH)-S-); siloxane (-0-Si(H)2-0-); and N,N'-dimethylhydrazine (-CH2-N(CH3)-N(CH3)-). Modified linkages, compared to natural phosphodiester linkages, can be used to alter, typically increase, nuclease resistance of the oligonucleotide. In certain embodiments, internucleoside linkages having a chiral atom can be prepared as a racemic mixture, or as separate enantiomers. Representative chiral linkages include, but are not limited to, alkylphosphonates and phosphorothioates. Methods of preparation of phosphorous-containing and non-phosphorous-containing internucleoside linkages are well known to those skilled in the art.

The oligonucleotides described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), a or 13 such as for sugar anomers, or as (D) or (L) such as for amino acids etc.
Included in the antisense compounds provided herein are all such possible isomers, as well as their racemic and optically pure forms.
Neutral internucleoside linkages include without limitation, phosphotriesters, methylphosphonates, MMI (3'-CH2-N(CH3)-0-5'), amide-3 (3'-CH2-C(=0)-N(H)-5'), amide-4 (3'-CH2-N(H)-C(=0)-5'), formacetal (3'-0-CH2-0-5'), and thioformacetal (3'-S-CH2-0-5'). Further neutral internucleoside linkages include nonionic linkages comprising siloxane (dialkylsiloxane), carboxylate ester, carboxamide, sulfide, sulfonate ester and amides (See for example: Carbohydrate Modifications in Antisense Research; Y.S. Sanghvi and P.D. Cook, Eds., ACS Symposium Series 580; Chapters 3 and 4, 40-65). Further neutral internucleoside linkages include nonionic linkages comprising mixed N, 0, S and CH2 component parts.
4. Certain Motifs In certain embodiments, antisense oligonucleotides comprise one or more modified nucleoside (e.g., nucleoside comprising a modified sugar and/or modified nucleobase) and/or one or more modified internucleoside linkage. The pattern of such modifications on an oligonucleotide is referred to herein as a motif In certain embodiments, sugar, nucleobase, and linkage motifs are independent of one another.
a. Certain sugar motifs In certain embodiments, oligonucleotides comprise one or more type of modified sugar moieties and/or naturally occurring sugar moieties arranged along an oligonucleotide or region thereof in a defined pattern or sugar modification motif Such motifs may include any of the sugar modifications discussed herein and/or other known sugar modifications.
In certain embodiments, the oligonucleotides comprise or consist of a region having a gapmer sugar motif, which comprises two external regions or "wings" and a central or internal region or "gap." The three regions of a gapmer sugar motif (the 5'-wing, the gap, and the 3'-wing) form a contiguous sequence of nucleosides wherein at least some of the sugar moieties of the nucleosides of each of the wings differ from at least some of the sugar moieties of the nucleosides of the gap. Specifically, at least the sugar moieties of the nucleosides of each wing that are closest to the gap (the 3'-most nucleoside of the 5'-wing and the 5'-most nucleoside of the 3'-wing) differ from the sugar moiety of the neighboring gap nucleosides, thus defining the boundary between the wings and the gap. In certain embodiments, the sugar moieties within the gap are the same as one another. In certain embodiments, the gap includes one or more nucleoside having a sugar moiety that differs from the sugar moiety of one or more other nucleosides of the gap. In certain embodiments, the sugar motifs of the two wings are the same as one another (symmetric sugar gapmer). In certain embodiments, the sugar motifs of the 5'-wing differs from the sugar motif of the 3'-wing (asymmetric sugar gapmer).
i. Certain 5'-wings In certain embodiments, the 5'- wing of a gapmer consists of 1 to 8 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 to 7 linked nucleosides.
In certain embodiments, the 5'-wing of a gapmer consists of 1 to 6 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 to 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 3 to 5 linked nucleosides.
In certain embodiments, the 5'- wing of a gapmer consists of 4 or 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 to 4 linked nucleosides.
In certain embodiments, the 5'-wing of a gapmer consists of 1 to 3 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 or 2 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 2 to 4 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 2 or 3 linked nucleosides.
In certain embodiments, the 5'- wing of a gapmer consists of 3 or 4 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 nucleoside. In certain embodiments, the 5'- wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 3 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 4 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 6 linked nucleosides.
In certain embodiments, the 5'- wing of a gapmer comprises at least one bicyclic nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least two bicyclic nucleosides. In certain embodiments, the 5'- wing of a gapmer comprises at least three bicyclic nucleosides. In certain embodiments, the 5'- wing of a gapmer comprises at least four bicyclic nucleosides. In certain embodiments, the 5'- wing of a gapmer comprises at least one constrained ethyl nucleoside.
In certain embodiments, the 5'-wing of a gapmer comprises at least one LNA nucleoside. In certain embodiments, each nucleoside of the 5'-wing of a gapmer is a bicyclic nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a constrained ethyl nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a LNA nucleoside.
In certain embodiments, the 5'- wing of a gapmer comprises at least one non-bicyclic modified nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-substituted nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-MOE nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-0Me nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a non-bicyclic modified nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-substituted nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-MOE
nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-0Me nucleoside.
In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-deoxynucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-deoxynucleoside. In a certain embodiments, the 5'- wing of a gapmer comprises at least one ribonucleoside.
In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a ribonucleoside. In certain embodiments, one, more than one, or each of the nucleosides of the 5'- wing is an RNA-like nucleoside.
In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-MOE
nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-0Me nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-deoxynucleoside.
In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-MOE nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-0Me nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-deoxynucleoside.
ii. Certain 3'-wings In certain embodiments, the 3'- wing of a gapmer consists of 1 to 8 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 to 7 linked nucleosides.
In certain embodiments, the 3'-wing of a gapmer consists of 1 to 6 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 to 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 3 to 5 linked nucleosides.
In certain embodiments, the 3'- wing of a gapmer consists of 4 or 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 to 4 linked nucleosides.
In certain embodiments, the 3'-wing of a gapmer consists of 1 to 3 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 or 2 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 2 to 4 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 2 or 3 linked nucleosides.
In certain embodiments, the 3'- wing of a gapmer consists of 3 or 4 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 nucleoside. In certain embodiments, the 3'- wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 31inked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 4 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 6 linked nucleosides.
In certain embodiments, the 3'- wing of a gapmer comprises at least one bicyclic nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one constrained ethyl nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one LNA nucleoside.
In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a bicyclic nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a constrained ethyl nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a LNA nucleoside.
In certain embodiments, the 3'- wing of a gapmer comprises at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least two non-bicyclic modified nucleosides. In certain embodiments, the 3'- wing of a gapmer comprises at least three non-bicyclic modified nucleosides. In certain embodiments, the 3'- wing of a gapmer comprises at least four non-bicyclic modified nucleosides. In certain embodiments, the 3'- wing of a gapmer comprises at least one 2'-substituted nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one 2'-MOE nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one 2'-0Me nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a non-bicyclic modified nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-substituted nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-MOE
nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-0Me nucleoside.
In certain embodiments, the 3'- wing of a gapmer comprises at least one 2'-deoxynucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-deoxynucleoside. In a certain embodiments, the 3'- wing of a gapmer comprises at least one ribonucleoside.
In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a ribonucleoside. In certain embodiments, one, more than one, or each of the nucleosides of the 5'- wing is an RNA-like nucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-MOE
nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-0Me nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-MOE nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-0Me nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA
nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one 2'-MOE
nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one 2'-0Me nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA
nucleoside and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one non-bicyclic modified nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one non-bicyclic modified nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside, at least one non-bicyclic modified nucleoside, and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one 2'-substituted nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one 2'-substituted nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA
nucleoside, at least one 2'-substituted nucleoside, and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one 2'-MOE nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one 2'-MOE nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA
nucleoside, at least one 2'-MOE nucleoside, and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one 2'-0Me nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one 2'-0Me nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA
nucleoside, at least one 2'-0Me nucleoside, and at least one 2'-deoxynucleoside.
iii. Certain Central Regions (gaps) In certain embodiments, the gap of a gapmer consists of 6 to 20 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 15 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 12 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 or 7 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 to 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 or 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 8 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 8 or 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 11 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 12 linked nucleosides.
In certain embodiments, each nucleoside of the gap of a gapmer is a 2'-deoxynucleoside. In certain embodiments, the gap comprises one or more modified nucleosides. In certain embodiments, each nucleoside of the gap of a gapmer is a 2'-deoxynucleoside or is a modified nucleoside that is "DNA-like." In such embodiments, "DNA-like" means that the nucleoside has similar characteristics to DNA, such that a duplex comprising the gapmer and an RNA molecule is capable of activating RNase H.
For example, under certain conditions, 2'-(ara)-F have been shown to support RNase H activation, and thus is DNA-like. In certain embodiments, one or more nucleosides of the gap of a gapmer is not a 2'-deoxynucleoside and is not DNA-like. In certain such embodiments, the gapmer nonetheless supports RNase H
activation (e.g., by virtue of the number or placement of the non-DNA nucleosides).
In certain embodiments, gaps comprise a stretch of unmodified 2'-deoxynucleoside interrupted by one or more modified nucleosides, thus resulting in three sub-regions (two stretches of one or more 2'-deoxynucleosides and a stretch of one or more interrupting modified nucleosides). In certain embodiments, no stretch of unmodified 2'-deoxynucleosides is longer than 5, 6, or 7 nucleosides. In certain embodiments, such short stretches is achieved by using short gap regions. In certain embodiments, short stretches are achieved by interrupting a longer gap region.
In certain embodiments, the gap comprises one or more modified nucleosides. In certain embodiments, the gap comprises one or more modified nucleosides selected from among cEt, FHNA, LNA, and 2-thio-thymidine. In certain embodiments, the gap comprises one modified nucleoside. In certain embodiments, the gap comprises a 5'-substituted sugar moiety selected from among 5'-Me, and 5'-(R)-Me.
In certain embodiments, the gap comprises two modified nucleosides. In certain embodiments, the gap comprises three modified nucleosides. In certain embodiments, the gap comprises four modified nucleosides.
In certain embodiments, the gap comprises two or more modified nucleosides and each modified nucleoside is the same. In certain embodiments, the gap comprises two or more modified nucleosides and each modified nucleoside is different.
In certain embodiments, the gap comprises one or more modified linkages. In certain embodiments, the gap comprises one or more methyl phosphonate linkages. In certain embodiments the gap comprises two or more modified linkages. In certain embodiments, the gap comprises one or more modified linkages and one or more modified nucleosides. In certain embodiments, the gap comprises one modified linkage and one modified nucleoside. In certain embodiments, the gap comprises two modified linkages and two or more modified nucleosides.

b. Certain Internucleoside Linkage Motifs In certain embodiments, oligonucleotides comprise modified internucleoside linkages arranged along the oligonucleotide or region thereof in a defined pattern or modified internucleoside linkage motif In certain embodiments, oligonucleotides comprise a region having an alternating internucleoside linkage motif In certain embodiments, oligonucleotides of the present disclosure comprise a region of uniformly modified internucleoside linkages. In certain such embodiments, the oligonucleotide comprises a region that is uniformly linked by phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide is uniformly linked by phosphorothioate internucleoside linkages.
In certain embodiments, each internucleoside linkage of the oligonucleotide is selected from phosphodiester and phosphorothioate. In certain embodiments, each internucleoside linkage of the oligonucleotide is selected from phosphodiester and phosphorothioate and at least one internucleoside linkage is phosphorothioate.
In certain embodiments, the oligonucleotide comprises at least 6 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 7 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 8 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 9 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 10 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 11 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 12 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 13 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 14 phosphorothioate internucleoside linkages.
In certain embodiments, the oligonucleotide comprises at least one block of at least 6 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 7 consecutive phosphorothioate internucleoside linkages.
In certain embodiments, the oligonucleotide comprises at least one block of at least 8 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 9 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 10 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least block of at least one 12 consecutive phosphorothioate internucleoside linkages. In certain such embodiments, at least one such block is located at the 3' end of the oligonucleotide.
In certain such embodiments, at least one such block is located within 3 nucleosides of the 3' end of the oligonucleotide.In certain embodiments, the oligonucleotide comprises less than 15 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 14 phosphoro-thioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 13 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than
12 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 11 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 10 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 9 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 8 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 7 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 6 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 5 phosphorothioate internucleoside linkages.
c. Certain Nucleobase Modification Motifs In certain embodiments, oligonucleotides comprise chemical modifications to nucleobases arranged along the oligonucleotide or region thereof in a defined pattern or nucleobases modification motif In certain such embodiments, nucleobase modifications are arranged in a gapped motif In certain embodiments, nucleobase modifications are arranged in an alternating motif In certain embodiments, each nucleobase is modified. In certain embodiments, none of the nucleobases is chemically modified.
In certain embodiments, oligonucleotides comprise a block of modified nucleobases. In certain such embodiments, the block is at the 3'-end of the oligonucleotide. In certain embodiments the block is within 3 nucleotides of the 3'-end of the oligonucleotide. In certain such embodiments, the block is at the 5'-end of the oligonucleotide. In certain embodiments the block is within 3 nucleotides of the 5'-end of the oligonucleotide.
In certain embodiments, nucleobase modifications are a function of the natural base at a particular position of an oligonucleotide. For example, in certain embodiments each purine or each pyrimidine in an oligonucleotide is modified. In certain embodiments, each adenine is modified.
In certain embodiments, each guanine is modified. In certain embodiments, each thymine is modified. In certain embodiments, each cytosine is modified. In certain embodiments, each uracil is modified.
In certain embodiments, some, all, or none of the cytosine moieties in an oligonucleotide are 5-methyl cytosine moieties. Herein, 5-methyl cytosine is not a "modified nucleobase." Accordingly, unless otherwise indicated, unmodified nucleobases include both cytosine residues having a 5-methyl and those lacking a 5 methyl. In certain embodiments, the methylation state of all or some cytosine nucleobases is specified.
In certain embodiments, chemical modifications to nucleobases comprise attachment of certain conjugate groups to nucleobases. In certain embodiments, each purine or each pyrimidine in an oligonucleotide may be optionally modified to comprise a conjugate group.
d. Certain Overall Lengths In certain embodiments, the present disclosure provides oligonucleotides of any of a variety of ranges of lengths. In certain embodiments, oligonucleotides consist of X to Y linked nucleosides, where X
represents the fewest number of nucleosides in the range and Y represents the largest number of nucleosides in the range. In certain such embodiments, X and Y are each independently selected from 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, and 50; provided that X<Y. For example, in certain embodiments, the oligonucleotide may consist of 8 to 9, 8 to 10, 8 to 11, 8 to 12, 8 to 13, 8 to 14, 8 to 15, 8 to 16, 8 to 17, 8 to 18, 8 to 19, 8 to 20, 8 to 21, 8 to 22, 8 to 23, 8 to 24, 8 to 25, 8 to 26, 8 to 27, 8 to 28, 8 to 29, 8 to 30, 9 to 10, 9 to 11, 9 to 12, 9 to 13, 9 to 14, 9 to 15, 9 to 16, 9 to 17, 9 to 18, 9 to 19, 9 to 20, 9 to 21, 9 to 22, 9 to 23, 9 to 24, 9 to 25, 9 to 26, 9 to 27, 9 to 28, 9 to 29, 9 to 30, 10 to 11, 10 to 12, 10 to 13, 10 to 14, 10 to 15, 10 to 16, 10 to 17, 10 to 18, 10 to 19, 10 to 20, 10 to 21, 10 to 22, 10 to 23, 10 to 24, 10 to 25, 10 to 26, 10 to 27, to 28, 10 to 29, 10 to 30, 11 to 12, 11 to 13, 11 to 14, 11 to 15, 11 to 16, 11 to 17, 11 to 18, 11 to 19, 11 to 20, 11 to 21, 11 to 22, 11 to 23, 11 to 24, 11 to 25, 11 to 26, 11 to 27, 11 to 28, 11 to 29, 11 to 30, 12 to 13, 12 to 14, 12 to 15, 12 to 16, 12 to 17, 12 to 18, 12 to 19, 12 to 20, 12 to 21, 12 to 22, 12 to 23, 12 to 24, 12 to 25, 12 to 26, 12 to 27, 12 to 28, 12 to 29, 12 to 30, 13 to 14, 13 to 15, 13 to 16, 13 to 17, 13 to 18, 13 to 19,
13 to 20, 13 to 21, 13 to 22, 13 to 23, 13 to 24, 13 to 25, 13 to 26, 13 to 27, 13 to 28, 13 to 29, 13 to 30, 14 to 15, 14 to 16, 14 to 17, 14 to 18, 14 to 19, 14 to 20, 14 to 21, 14 to 22, 14 to 23, 14 to 24, 14 to 25, 14 to 26,
14 to 27, 14 to 28, 14 to 29, 14 to 30, 15 to 16, 15 to 17, 15 to 18, 15 to 19, 15 to 20, 15 to 21, 15 to 22, 15 to 23, 15 to 24, 15 to 25, 15 to 26, 15 to 27, 15 to 28, 15 to 29, 15 to 30, 16 to 17, 16 to 18, 16 to 19, 16 to 20, 16 to 21, 16 to 22, 16 to 23, 16 to 24, 16 to 25, 16 to 26, 16 to 27, 16 to 28, 16 to 29, 16 to 30, 17 to 18, 17 to 19, 17 to 20, 17 to 21, 17 to 22, 17 to 23, 17 to 24, 17 to 25, 17 to 26, 17 to 27, 17 to 28, 17 to 29, 17 to 30, 18 to 19, 18 to 20, 18 to 21, 18 to 22, 18 to 23, 18 to 24, 18 to 25, 18 to 26, 18 to 27, 18 to 28, 18 to 29, 18 to 30, 19 to 20, 19 to 21, 19 to 22, 19 to 23, 19 to 24, 19 to 25, 19 to 26, 19 to 29, 19 to 28, 19 to 29, 19 to 30, 20 to 21, 20 to 22, 20 to 23, 20 to 24, 20 to 25, 20 to 26, 20 to 27, 20 to 28, 20 to 29, 20 to 30, 21 to 22, 21 to 23, 21 to 24, 21 to 25, 21 to 26, 21 to 27, 21 to 28, 21 to 29, 21 to 30, 22 to 23, 22 to 24, 22 to 25, 22 to 26, 22 to 27, 22 to 28, 22 to 29, 22 to 30, 23 to 24, 23 to 25, 23 to 26, 23 to 27, 23 to 28, 23 to 29, 23 to 30, 24 to 25, 24 to 26, 24 to 27, 24 to 28, 24 to 29, 24 to 30, 25 to 26, 25 to 27, 25 to 28, 25 to 29, 25 to 30, 26 to 27, 26 to 28, 26 to 29, 26 to 30, 27 to 28, 27 to 29, 27 to 30, 28 to 29, 28 to 30, or 29 to 30 linked nucleosides. In embodiments where the number of nucleosides of an oligonucleotide of a compound is limited, whether to a range or to a specific number, the compound may, nonetheless further comprise additional other substituents.
For example, an oligonucleotide comprising 8-30 nucleosides excludes oligonucleotides having 31 nucleosides, but, unless otherwise indicated, such an oligonucleotide may further comprise, for example one or more conjugate groups, terminal groups, or other substituents.
Further, where an oligonucleotide is described by an overall length range and by regions having specified lengths, and where the sum of specified lengths of the regions is less than the upper limit of the overall length range, the oligonucleotide may have additional nucleosides, beyond those of the specified regions, provided that the total number of nucleosides does not exceed the upper limit of the overall length range.
5. Certain Antisense Oligonucleotide Chemistry Motifs In certain embodiments, the chemical structural features of antisense oligonucleotides are characterized by their sugar motif, internucleoside linkage motif, nucleobase modification motif and overall length. In certain embodiments, such parameters are each independent of one another. Thus, each internucleoside linkage of an oligonucleotide having a gapmer sugar motif may be modified or unmodified and may or may not follow the gapmer modification pattern of the sugar modifications. Thus, the internucleoside linkages within the wing regions of a sugar-gapmer may be the same or different from one another and may be the same or different from the internucleoside linkages of the gap region. Likewise, such sugar-gapmer oligonucleotides may comprise one or more modified nucleobase independent of the gapmer pattern of the sugar modifications. One of skill in the art will appreciate that such motifs may be combined to create a variety of oligonucleotides.
In certain embodiments, the selection of internucleoside linkage and nucleoside modification are not independent of one another.
i. Certain Sequences and Targets In certain embodiments, the invention provides antisense oligonucleotides having a sequence complementary to a target nucleic acid. Such antisense compounds are capable of hybridizing to a target nucleic acid, resulting in at least one antisense activity. In certain embodiments, antisense compounds specifically hybridize to one or more target nucleic acid. In certain embodiments, a specifically hybridizing antisense compound has a nucleobase sequence comprising a region having sufficient complementarity to a target nucleic acid to allow hybridization and result in antisense activity and insufficient complementarity to any non-target so as to avoid or reduce non-specific hybridization to non-target nucleic acid sequences under conditions in which specific hybridization is desired (e.g., under physiological conditions for in vivo or therapeutic uses, and under conditions in which assays are performed in the case of in vitro assays). In certain embodiments, oligonucleotides are selective between a target and non-target, even though both target and non-target comprise the target sequence. In such embodiments, selectivity may result from relative accessibility of the target region of one nucleic acid molecule compared to the other.
In certain embodiments, the present disclosure provides antisense compounds comprising oligonucleotides that are fully complementary to the target nucleic acid over the entire length of the oligonucleotide. In certain embodiments, oligonucleotides are 99%
complementary to the target nucleic acid.
In certain embodiments, oligonucleotides are 95% complementary to the target nucleic acid. In certain embodiments, such oligonucleotides are 90% complementary to the target nucleic acid.
In certain embodiments, such oligonucleotides are 85% complementary to the target nucleic acid. In certain embodiments, such oligonucleotides are 80% complementary to the target nucleic acid. In certain embodiments, an antisense compound comprises a region that is fully complementary to a target nucleic acid and is at least 80% complementary to the target nucleic acid over the entire length of the oligonucleotide. In certain such embodiments, the region of full complementarity is from 6 to 14 nucleobases in length.

In certain embodiments, oligonucleotides comprise a hybridizing region and a terminal region. In certain such embodiments, the hybridizing region consists of 12-30 linked nucleosides and is fully complementary to the target nucleic acid. In certain embodiments, the hybridizing region includes one mismatch relative to the target nucleic acid. In certain embodiments, the hybridizing region includes two mismatches relative to the target nucleic acid. In certain embodiments, the hybridizing region includes three mismatches relative to the target nucleic acid. In certain embodiments, the terminal region consists of 1-4 terminal nucleosides. In certain embodiments, the terminal nucleosides are at the 3' end. In certain embodiments, one or more of the terminal nucleosides are not complementary to the target nucleic acid.
Antisense mechanisms include any mechanism involving the hybridization of an oligonucleotide with target nucleic acid, wherein the hybridization results in a biological effect.
In certain embodiments, such hybridization results in either target nucleic acid degradation or occupancy with concomitant inhibition or stimulation of the cellular machinery involving, for example, translation, transcription, or splicing of the target nucleic acid.
One type of antisense mechanism involving degradation of target RNA is RNase H
mediated antisense. RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. It is known in the art that single-stranded antisense compounds which are "DNA-like"
elicit RNase H activity in mammalian cells. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of DNA-like oligonucleotide-mediated inhibition of gene expression.
In certain embodiments, a conjugate group comprises a cleavable moiety. In certain embodiments, a conjugate group comprises one or more cleavable bond. In certain embodiments, a conjugate group comprises a linker. In certain embodiments, a linker comprises a protein binding moiety. In certain embodiments, a conjugate group comprises a cell-targeting moiety (also referred to as a cell-targeting group).
In certain embodiments a cell-targeting moiety comprises a branching group. In certain embodiments, a cell-targeting moiety comprises one or more tethers. In certain embodiments, a cell-targeting moiety comprises a carbohydrate or carbohydrate cluster.
ii. Certain Cleavable Moieties In certain embodiments, a cleavable moiety is a cleavable bond. In certain embodiments, a cleavable moiety comprises a cleavable bond. In certain embodiments, the conjugate group comprises a cleavable moiety. In certain such embodiments, the cleavable moiety attaches to the antisense oligonucleotide. In certain such embodiments, the cleavable moiety attaches directly to the cell-targeting moiety. In certain such embodiments, the cleavable moiety attaches to the conjugate linker. In certain embodiments, the cleavable moiety comprises a phosphate or phosphodiester. In certain embodiments, the cleavable moiety is a cleavable nucleoside or nucleoside analog. In certain embodiments, the nucleoside or nucleoside analog comprises an optionally protected heterocyclic base selected from a purine, substituted purine, pyrimidine or substituted pyrimidine. In certain embodiments, the cleavable moiety is a nucleoside comprising an optionally protected heterocyclic base selected from uracil, thymine, cytosine, 4-N-benzoylcytosine, 5-methylcytosine, 4-N-benzoy1-5-methylcytosine, adenine, 6-N-benzoyladenine, guanine and 2-N-isobutyrylguanine. In certain embodiments, the cleavable moiety is 2'-deoxy nucleoside that is attached to the 3' position of the antisense oligonucleotide by a phosphodiester linkage and is attached to the linker by a phosphodiester or phosphorothioate linkage. In certain embodiments, the cleavable moiety is 2'-deoxy adenosine that is attached to the 3' position of the antisense oligonucleotide by a phosphodiester linkage and is attached to the linker by a phosphodiester or phosphorothioate linkage. In certain embodiments, the cleavable moiety is 2'-deoxy adenosine that is attached to the 3' position of the antisense oligonucleotide by a phosphodiester linkage and is attached to the linker by a phosphodiester linkage.
In certain embodiments, the cleavable moiety is attached to the 3' position of the antisense oligonucleotide. In certain embodiments, the cleavable moiety is attached to the 5' position of the antisense oligonucleotide. In certain embodiments, the cleavable moiety is attached to a 2' position of the antisense oligonucleotide. In certain embodiments, the cleavable moiety is attached to the antisense oligonucleotide by a phosphodiester linkage. In certain embodiments, the cleavable moiety is attached to the linker by either a phosphodiester or a phosphorothioate linkage. In certain embodiments, the cleavable moiety is attached to the linker by a phosphodiester linkage. In certain embodiments, the conjugate group does not include a cleavable moiety.
In certain embodiments, the cleavable moiety is cleaved after the complex has been administered to an animal only after being internalized by a targeted cell. Inside the cell the cleavable moiety is cleaved thereby releasing the active antisense oligonucleotide. While not wanting to be bound by theory it is believed that the cleavable moiety is cleaved by one or more nucleases within the cell.
In certain embodiments, the one or more nucleases cleave the phosphodiester linkage between the cleavable moiety and the linker. In certain embodiments, the cleavable moiety has a structure selected from among the following:
0=P-OH
LcoNtBxi 0=P-OH 0=P-OH
\coRy cf\
0=P-OH
0=p-OH 0=p-OH

c0),13x2 c0),13x3 , and 0=P-OH 04'-OH 0=P-OH

wherein each of Bx, Bxi, Bx2, and Bx3 is independently a heterocyclic base moiety. In certain embodiments, the cleavable moiety has a structure selected from among the following:
I
0=P-OH NH2 O N..1õ.--Li \c0),N NI-) (1 0=P-OH

iii. Certain Linkers In certain embodiments, the conjugate groups comprise a linker. In certain such embodiments, the linker is covalently bound to the cleavable moiety. In certain such embodiments, the linker is covalently bound to the antisense oligonucleotide. In certain embodiments, the linker is covalently bound to a cell-targeting moiety. In certain embodiments, the linker further comprises a covalent attachment to a solid support. In certain embodiments, the linker further comprises a covalent attachment to a protein binding moiety. In certain embodiments, the linker further comprises a covalent attachment to a solid support and further comprises a covalent attachment to a protein binding moiety. In certain embodiments, the linker includes multiple positions for attachment of tethered ligands. In certain embodiments, the linker includes multiple positions for attachment of tethered ligands and is not attached to a branching group. In certain embodiments, the linker further comprises one or more cleavable bond. In certain embodiments, the conjugate group does not include a linker.
In certain embodiments, the linker includes at least a linear group comprising groups selected from alkyl, amide, disulfide, polyethylene glycol, ether, thioether (-S-) and hydroxylamino (-0-N(H)-) groups. In certain embodiments, the linear group comprises groups selected from alkyl, amide and ether groups. In certain embodiments, the linear group comprises groups selected from alkyl and ether groups. In certain embodiments, the linear group comprises at least one phosphorus linking group.
In certain embodiments, the linear group comprises at least one phosphodiester group. In certain embodiments, the linear group includes at least one neutral linking group. In certain embodiments, the linear group is covalently attached to the cell-targeting moiety and the cleavable moiety. In certain embodiments, the linear group is covalently attached to the cell-targeting moiety and the antisense oligonucleotide. In certain embodiments, the linear group is covalently attached to the cell-targeting moiety, the cleavable moiety and a solid support. In certain embodiments, the linear group is covalently attached to the cell-targeting moiety, the cleavable moiety, a solid support and a protein binding moiety. In certain embodiments, the linear group includes one or more cleavable bond.
In certain embodiments, the linker includes the linear group covalently attached to a scaffold group.
In certain embodiments, the scaffold includes a branched aliphatic group comprising groups selected from alkyl, amide, disulfide, polyethylene glycol, ether, thioether and hydroxylamino groups. In certain embodiments, the scaffold includes a branched aliphatic group comprising groups selected from alkyl, amide and ether groups. In certain embodiments, the scaffold includes at least one mono or polycyclic ring system.
In certain embodiments, the scaffold includes at least two mono or polycyclic ring systems. In certain embodiments, the linear group is covalently attached to the scaffold group and the scaffold group is covalently attached to the cleavable moiety and the linker. In certain embodiments, the linear group is covalently attached to the scaffold group and the scaffold group is covalently attached to the cleavable moiety, the linker and a solid support. In certain embodiments, the linear group is covalently attached to the scaffold group and the scaffold group is covalently attached to the cleavable moiety, the linker and a protein binding moiety. In certain embodiments, the linear group is covalently attached to the scaffold group and the scaffold group is covalently attached to the cleavable moiety, the linker, a protein binding moiety and a solid support. In certain embodiments, the scaffold group includes one or more cleavable bond.
In certain embodiments, the linker includes a protein binding moiety. In certain embodiments, the protein binding moiety is a lipid such as for example including but not limited to cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-0(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, 03-(oleoyl)lithocholic acid, 03-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine), a vitamin (e.g., folate, vitamin A, vitamin E, biotin, pyridoxal), a peptide, a carbohydrate (e.g., monosaccharide, disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, polysaccharide), an endosomolytic component, a steroid (e.g., uvaol, hecigenin, diosgenin), a terpene (e.g., triterpene, e.g., sarsasapogenin, friedelin, epifriedelanol derivatized lithocholic acid), or a cationic lipid. In certain embodiments, the protein binding moiety is a C16 to C22 long chain saturated or unsaturated fatty acid, cholesterol, cholic acid, vitamin E, adamantane or 1-pentafluoropropyl.
In certain embodiments, a linker has a structure selected from among:

H H ¨NH
0,µ 0 0 )c)A )0¨P¨OH
N I

H .
1.---NO , vN,HriL0 , H ( ri_OA
( )n C)? N I
0,µ r011-0H , , X \

1¨NH
\ I .
\ OHiNI)OH
P I
I I
0 0, I , ' \. ON , 0 /OH
1D CN3C),/
H N . n , H 1 , rrcs,s,wLi 0 , "1\1N'HN
H n I
0,,.

HHHH H
v N,p,riN,(,),nN,v,ikl. N
')An 1\0.sµ , H .
0 , 0¨) /
I

\ ,,,, ON ,0 1 __ \ 10.-6- (0 /

OH ks )n \ /¨(-/N
11"1\D=di-O-r S¨S n 0 N
¨S7 741 6 N ; and skiliNH.(õ),0 H
v N,9riLo wherein each n is, independently, from 1 to 20; and p is from 1 to 6.

In certain embodiments, a linker has a structure selected from among:
, \
-rscj q q.
õ0)''-õ0 0 N

\
q.
N O
)\õA rrri 0 \
H H 0, N )0'-µ
l'ild 0 0 µ)NrCil\lrEd)n n 0 , \ 0 q.
N O
\
ckr,(S-!,,VA,Jr =

' n H
c'ssLI\IC) =11\10 =
n H

µ
(:) 'N.
0 (:) prrj ((iNC j=L(') hl A
\ (J:rNA.
5J-r' n H
q a \
9.
N I

I
O-P=0 H OH ;

,,zrNo \(--41-0 ;and H ya:iN ___________________________ H
0 , c'N N S,L>N¨i l-,n H

#
HO
wherein each n is, independently, from 1 to 20.

In certain embodiments, a linker has a structure selected from among:

H
,122H H
N \
n r%

HN,..0 H cso,S, 0 µ)/nN-in S ;
H
N.q. , c'CN 0 n H 0 n "sN N -H A
H n H ; i.õ;.....--1-i.........1-----õN_--N ;
0 n H
0 0 ' H
/2 1-N.HOO/H n f ; / H H
Q z,&
n O 0 n H
H H
; and n c3 n H
H N
"sr-iN'HO/N n n n wherein n is from 1 to 20.

In certain embodiments, a linker has a structure selected from among:

H
H H
n YI'l)nci ;

OH

W ;
i& N H , N n 0 ' n H n 11\in N 'Hµ ,1--N \ I
\ 1 EN-I =
;

H H
H
N csss sos N
n Kii 0 CrHn 1 ; '3tc)-C\IY ' ciNH"c) 0/Hn ;
n n H
H H
siN.H0Q0/'H'n ' N . cs N
n f===1---:':0 0 \ -/ n s' n 0 0 AOH
( I ()pH
- n -)n -I¨ ¨ ' .
"n_ n -n _ -n _ -n "rcYHriANH-1-, and csrYOL

wherein each L is, independently, a phosphorus linking group or a neutral linking group; and each n is, independently, from 1 to 20.
In certain embodiments, a linker has a structure selected from among:

pi's\
pi's\ 0 :
(:), )0)µ' )0A 0 N

µ)N s,So , µ) 0 II
0¨P¨OH

)A I I

H H
µ,...ecN -.-N(,,)...,N...,.....õ(=---..,, = 0 - H

I )0A I 0 I
N
I \N rO¨P¨OH
II
, (:). 1¨NH
)0A

N ),,k = I
\ 1, 0 0 vv."' 0 ,0 0-.6_ -s 0 I N H
0 v, N f.,gLo 01,, 0 1 CN). 0,5ss H

I

I '1 HHHH H
CNI) ,0c N,{4N,k,rN,I.NiNip,,O, ;
Q.Ne.
= c5ss S
H ' S' 1")&
0 , 0 ,\:õ.N0 I

0 0 ,0 0, I
\ 7------.7---IND'I-0/ OH
, 0\
H
0 Q 'C), . S¨S 0 N
H

I

6 cs H N
JVVIJ
N 3 0 \,,...
HN
izi H )& ; =
,s N

and I
O
o \ ".. o> o ,o I
i /K 0.4%- ' o 1 \ 7.-----/-----\C o CS
s-s o \,,...
N
H
lKi .
H

In certain embodiments, a linker has a structure selected from among:

H H
µ., N N
. ,222L N .1.r-, N A . `z22..) EN-I =IrWL -s cs' .
0 0 ' 0 ,222.). H

0 N ;
`sCN EI\11,5s ;
r 0 A .
css''=
hi '')( N 'HC' hi , H

H
cey H
and H
H
css' N 0//N "" =

In certain embodiments, a linker has a structure selected from among:

.
0 0 ' H 0 H ' 0 , H

H j:? \
)N \)cs5 cssLN N 4 ; 0 e '.
er H
\ .

\.,--5----(___)------_ sk/(2r;2'z - ;

H
H
H H

H
HH
sssN rcj ; ''N OQC)//N

IsOS ; s&00,05 ; s&0 00ss ;

H N ,s II
0¨P-0 -,4,A,000 / ;

OH "3 "3 XH

II
F 0 ¨P-0....ro oo¨A-0¨ ; coy--kAm and OH "3 3 OH 3 .,, H

ssLN H-C)¨C)¨C)-3 H 60H ' In certain embodiments, a linker has a structure selected from among:
J-rs' NOA

and µn wherein n is from 1 to 20.
In certain embodiments, a linker has a structure selected from among:
'of; sK0e.\/.1 ; and In certain embodiments, a linker has a structure selected from among:
/OH
/OH
and OH "3 3 OH OH '3 "3 c' =
In certain embodiments, a linker has a structure selected from among:

csss l\e'k)L
3 NO-1:1'-0¨ 3 NK

0 and 0 =
In certain embodiments, the conjugate linker has the structure:
-rfj\j O
q.
)0)2-µ0 In certain embodiments, the conjugate linker has the structure:

H =
In certain embodiments, a linker has a structure selected from among:

OH =
0 and 0 In certain embodiments, a linker has a structure selected from among:

f\A-OL
NvH- sscs.Ln n OH
0 and 0 =
5 wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
iv. Certain Cell-Targeting Moieties In certain embodiments, conjugate groups comprise cell-targeting moieties.
Certain such cell-targeting moieties increase cellular uptake of antisense compounds. In certain embodiments, cell-targeting moieties comprise a branching group, one or more tether, and one or more ligand. In certain embodiments, cell-targeting moieties comprise a branching group, one or more tether, one or more ligand and one or more cleavable bond.
1. Certain Branching Groups In certain embodiments, the conjugate groups comprise a targeting moiety comprising a branching group and at least two tethered ligands. In certain embodiments, the branching group attaches the conjugate linker. In certain embodiments, the branching group attaches the cleavable moiety. In certain embodiments, the branching group attaches the antisense oligonucleotide. In certain embodiments, the branching group is covalently attached to the linker and each of the tethered ligands. In certain embodiments, the branching group comprises a branched aliphatic group comprising groups selected from alkyl, amide, disulfide, polyethylene glycol, ether, thioether and hydroxylamino groups. In certain embodiments, the branching group comprises groups selected from alkyl, amide and ether groups. In certain embodiments, the branching group comprises groups selected from alkyl and ether groups. In certain embodiments, the branching group comprises a mono or polycyclic ring system. In certain embodiments, the branching group comprises one or more cleavable bond. In certain embodiments, the conjugate group does not include a branching group.
In certain embodiments, a branching group has a structure selected from among:

JIM, 0 /'Z71 \

O
\
NH
; HO
NH 0 n CH3 H im I
JVIN
0 ( in 0 H)L h n n v N N.{Ne-N,(-----W
rsssYr>\ N N
0 , ( )n H 8 ( )n H =
0 (0/
.r5Jj rs'r C/s, n m itCY1L
0: I-13 n CH3 ( n n 0 01 \ 0 ,/<
\ CH3.4 ois im ' " n ( NH rscr \ l'ri rd 1 0\ __ .(:) NH NH rsss )n ( ln ; 0 )0 41, ' , V¨NH /
H

NH
C)II
css5 )ri \
Zn H 0 I .

, "s'N
H

0 (< H
(H , 0 v NH
\ = n NH
\ n NH
)n 0 Zi 0 H
\ n N
;and H

=
H H (< (<
p5cA-)r NH
v NH

wherein each n is, independently, from 1 to 20;
j is from 1 to 3; and m is from 2 to 6.

In certain embodiments, a branching group has a structure selected from among:

.217--0 0 0 I \ 0 C)\ 0 \ N \ ; HO 0¨FLO '2,?_) -1\1,'õ?L
=

'n .I'YX =
H
NH 0 n OHn I CH3 /m ' 10) e '117._ H 0 ( I, n H 0 n n N
\ N N e-'N
el =
, (LH 0 n H
pria .4J't 0 ( L.1, /n OWL/ J1Ilflr m NH (02, 1 ''''Orµ . )n CH3 n CH3 n Nk, , ,,,' im ' s.õ(,N

; 0 rj.
( t-NH
CH3 pmoiik n 0 m ed 0 ;and o I
O
I =,, , cl) ¨
NH ( NH 0-)n H
0 m wherein each n is, independently, from 1 to 20; and m is from 2 to 6.
In certain embodiments, a branching group has a structure selected from among:

0, i'LL

N,\/\;\ ; N'WN A

H
H . 0 NH 0 , NH /
I

JuW
0 ____________________________________________________________________ ./<
,V1J1J g.
l / ___ N H
,s' NH
0 0 Ct\ 0 A
µ N , ) /KO .
0 ; . /' , ,V1J1J H

\. 0 HN `22z)-1----NH
) \/\/\)1"---NH

H ?css\N i\ij-., , ; , ' siN N ssss ;
H csssN µ H

H O/

HN
v NH

NH NH
0 i_i 0 H Nj-L, j rsjs ; and H
H O/ ' O/
cissNH
,NH

In certain embodiments, a branching group has a structure selected from among:
\ I
Al_ -L, A A1 /
Ali .
FAV in ' In Xi- Ai / and wherein each A1 is independently, 0, S, C=0 or NH; and each n is, independently, from 1 to 20.
In certain embodiments, a branching group has a structure selected from among:
Juuv (1)n A1-1 Nn A1-1 ¨A ()in je) __ A *1 - A
/-µ1 and n, n /-µ1 Srr) wherein each A1 is independently, 0, S, C=0 or NH; and each n is, independently, from 1 to 20.
In certain embodiments, a branching group has a structure selected from among:
psis )n n and 'tz.,_)( irn n n \s"
wherein A1 is 0, S, C=0 or NH; and each n is, independently, from 1 to 20.
In certain embodiments, a branching group has a structure selected from among:
0, =
In certain embodiments, a branching group has a structure selected from among:

In certain embodiments, a branching group has a structure selected from among:
\ss s.
2. Certain Tethers In certain embodiments, conjugate groups comprise one or more tethers covalently attached to the branching group. In certain embodiments, conjugate groups comprise one or more tethers covalently attached to the linking group. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, ether, thioether, disulfide, amide and polyethylene glycol groups in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, substituted alkyl, ether, thioether, disulfide, amide, phosphodiester and polyethylene glycol groups in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, ether and amide groups in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, substituted alkyl, phosphodiester, ether and amide groups in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl and phosphodiester in any combination.
In certain embodiments, each tether comprises at least one phosphorus linking group or neutral linking group.
In certain embodiments, the tether includes one or more cleavable bond. In certain embodiments, the tether is attached to the branching group through either an amide or an ether group. In certain embodiments, the tether is attached to the branching group through a phosphodiester group. In certain embodiments, the tether is attached to the branching group through a phosphorus linking group or neutral linking group. In certain embodiments, the tether is attached to the branching group through an ether group.
In certain embodiments, the tether is attached to the ligand through either an amide or an ether group. In certain embodiments, the tether is attached to the ligand through an ether group. In certain embodiments, the tether is attached to the ligand through either an amide or an ether group. In certain embodiments, the tether is attached to the ligand through an ether group.
In certain embodiments, each tether comprises from about 8 to about 20 atoms in chain length between the ligand and the branching group. In certain embodiments, each tether group comprises from about 10 to about 18 atoms in chain length between the ligand and the branching group. In certain embodiments, each tether group comprises about 13 atoms in chain length.
In certain embodiments, a tether has a structure selected from among:
\,azzCLI\r61 oc),c) Frilt = ,z27_ 1\1.:11 = ; 1 n =
µ
N
''n 'fl 'n H n H H
N
N n ;\
n Il n n ;

o H
; rrrr.(, 00 N n N
.j(.41y1 /n H \ \ in \ ''n 'n jr).L N
n N ; ;and wherein each n is, independently, from 1 to 20; and each p is from 1 to about 6.
In certain embodiments, a tether has a structure selected from among:

N ; N

; ; ; and In certain embodiments, a tether has a structure selected from among:
H H
N N
\
0 no wherein each n is, independently, from 1 to 20.

In certain embodiments, a tether has a structure selected from among:
0 Zi c&efl-feL and `YLNI-Sek mi mi mi H m 1 wherein L is either a phosphorus linking group or a neutral linking group;
Z1 is C(=0)0-R2;
Z2 is H, C1-C6 alkyl or substituted C1-C6 alkY;
R2 is H, C1-C6 alkyl or substituted C1-C6 alky; and each m1 is, independently, from 0 to 20 wherein at least one m1 is greater than 0 for each tether.
In certain embodiments, a tether has a structure selected from among:

In certain embodiments, a tether has a structure selected from among:
0 411. 0 COOH OH
+n and and csis 04-0¨WA
411, 6H m Nr wherein Z2 is H or CH3; and each m1 is, independently, from 0 to 20 wherein at least one m1 is greater than 0 for each tether.
In certain embodiments, a tether has a structure selected from among:

4 H n H
, or ; wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
In certain embodiments, a tether comprises a phosphorus linking group. In certain embodiments, a tether does not comprise any amide bonds. In certain embodiments, a tether comprises a phosphorus linking group and does not comprise any amide bonds.

3. Certain Ligands In certain embodiments, the present disclosure provides ligands wherein each ligand is covalently attached to a tether. In certain embodiments, each ligand is selected to have an affinity for at least one type of receptor on a target cell. In certain embodiments, ligands are selected that have an affinity for at least one type of receptor on the surface of a mammalian liver cell. In certain embodiments, ligands are selected that have an affinity for the hepatic asialoglycoprotein receptor (ASGP-R). In certain embodiments, each ligand is a carbohydrate. In certain embodiments, each ligand is, independently selected from galactose, N-acetyl galactoseamine, mannose, glucose, glucosamone and fucose. In certain embodiments, each ligand is N-acetyl galactoseamine (GalNAc). In certain embodiments, the targeting moiety comprises 2 to 6 ligands. In certain embodiments, the targeting moiety comprises 3 ligands. In certain embodiments, the targeting moiety comprises 3 N-acetyl galactoseamine ligands.
In certain embodiments, the ligand is a carbohydrate, carbohydrate derivative, modified carbohydrate, multivalent carbohydrate cluster, polysaccharide, modified polysaccharide, or polysaccharide derivative. In certain embodiments, the ligand is an amino sugar or a thio sugar. For example, amino sugars may be selected from any number of compounds known in the art, for example glucosamine, sialic acid, a-D-galactosamine, N-Acetylgalactosamine, 2-acetamido-2-deoxy-D-galactopyranose (GalNAc), 2-Amino-3-0-[(R)-1- carb oxyethyl] -2- deoxy-13-D- gluc opyranos e (13-muramic acid), 2-D eoxy-2-methylamino-L-gluc opyranos e, 4,6-Dideoxy-4-formamido-2,3 -di- 0-methyl-D-mannopyranose, 2-D eoxy-2 -sulfoamino-D-glucopyranose and N-sulfo-D-glucosamine, and N-Glycoloyl-a-neuraminic acid.
For example, thio sugars may be selected from the group consisting of 5-Thio-13-D-g1ucopyranose, Methyl 2,3,4-tri-O-acety1-1-thio-6-0-trity1-a-D-g1ucopyranoside, 4-Thio-13-D-ga1actopyranose, and ethyl 3,4,6,7-tetra-0-acety1-2-deoxy-1,5-dithio-a-D-g/uco-heptopyranoside.
In certain embodiments, "GalNac" or "Gal-NAc" refers to 2-(Acetylamino)-2-deoxy-D-galactopyranose, commonly referred to in the literature as N-acetyl galactosamine. In certain embodiments, "N-acetyl galactosamine" refers to 2-(Acetylamino)-2-deoxy-D-galactopyranose.
In certain embodiments, "GalNac" or "Gal-NAc" refers to 2-(Acetylamino)-2-deoxy-D-galactopyranose. In certain embodiments, "GalNac" or "Gal-NAc" refers to 2-(Acetylamino)-2-deoxy-D-galactopyranose, which includes both the [3-form: 2-(Acety1amino)-2-deoxy-13-D-ga1actopyranose and a-form: 2-(Acetylamino)-2-deoxy-D-galactopyranose. In certain embodiments, both the 13-form: 2-(Acety1amino)-2-deoxy-13-D-ga1actopyranose and a-form: 2-(Acetylamino)-2-deoxy-D-galactopyranose may be used interchangeably. Accordingly, in structures in which one form is depicted, these structures are intended to include the other form as well. For example, where the structure for an a-form: 2-(Acetylamino)-2-deoxy-D-galactopyranose is shown, this structure is intended to include the other form as well. In certain embodiments, In certain preferred embodiments, the 13-form 2-(Acetylamino)-2-deoxy-D-galactopyranose is the preferred embodiment.

.41%ko%%,04. jsri-OH

HO ..11/4/iN
H
OH
2-(Acetylamino)-2-deoxy-D-galactopyranose OH
OH

HO 0¨

NHAc 2-(Acety1amino)-2-deoxy-13-D-ga1actopyranose OH
OH

HO
NHAc Osss5 2-(Acetylamino)-2-deoxy-a-D-galactopyranose In certain embodiments one or more ligand has a structure selected from among:
OH
OH
OH
7..(_.:..) HO0 HO¨

...õ.r(2.\.__. HO OH

Ri and Ri Ri wherein each R1 is selected from OH and NHCOOH.

In certain embodiments one or more ligand has a structure selected from among:
HOOH OH HO HO
OHOH

HO-VOX ,ss ; HO x_ss . 1-1S?0 oNsse ;
HO =
, NHAc r OH e , 11 \rrr' HOOH OH

HO----/-:::7\0 OH
HO-------\--------\,NN,rss , HOOH
. _....\....:)._\, HN/[7)0 OHil OH

\?..
HO HO, Nrer , HO (:) ; and OH OH
OH
HO
_ (..\11-%

HO
0 _________________ HO OH
OH
HO
F-K,-\._ A

I .
HO
0\

In certain embodiments one or more ligand has a structure selected from among:
HOOH
_\12 HO r N, -NHAc r .
In certain embodiments one or more ligand has a structure selected from among:
HOOH
0 (-1 HO-=-"\------=\/¨X.rjs NHAc r =

i. Certain Conjugates In certain embodiments, conjugate groups comprise the structural features above. In certain such embodiments, conjugate groups have the following structure:
HO OH

HO
O) )n NHAc 0 HO OH
0 H H \
N 0,t_ in H
HO¨I
n n NHAc 0 )n OH
HO

_......7:\zo,H,-......
n HO n NHAc 0 =
wherein each n is, independently, from 1 to 20.
In certain such embodiments, conjugate groups have the following structure:
HO OH
_......70......\..õ., H 0 HO
NHAc 0 N¨I

HO0...,...õ..............,.....õ---NN,7"\\ZN --........,-"N. -.......s.........
NHAc 0 0 CD"
OH
HO
H
¨

HO

NHAc =
In certain such embodiments, conjugate groups have the following structure:

HO H

0=P-OH

N N OH
O
n --------ci \
NHAc n 01...r....,.........ANKINN(,...).õ-0 0/Bx N
HO n ----H n P=-I
NHAc 0 :4) 0 0 n )11 OH
1-1(2:;i::\--------H HN-j:

HO 0/n n NHAc O
wherein each n is, independently, from 1 to 20;
Z is H or a linked solid support;
Q is an antisense compound;
X is 0 or S; and Bx is a heterocyclic base moiety.
In certain such embodiments, conjugate groups have the following structure:
HO H

0=P-OH
OH "---..7N NN--------ti O
HO
NHAc \,cTONyBx HO
0 ¨P=X
I
NHAc 0 0 ,/ 0 OH

OH
1-1(:::;i::\--------H HN-j:

HO ON'"----.../
NHAc In certain such embodiments, conjugate groups have the following structure:
HO H

0=P-OH

N N

NJN
3 -----ti I
NHAc Lco),N N,..) -......_ .
n.,õ\ 6=
O¨P=0 I
NHAc 0 0 OH

HO H
HN
HO -----olr3N-..,&))/ 0 NHAc o In certain such embodiments, conjugate groups have the following structure:
HOOH
HOO,W\ ,IC, n 0 1 0 AcH N ()n HO OH OH) H ___,T.9....\,0 9 0 O IH--1.1;`03¨hk ] I
AcH N OH n 0 ,...1.2.....v0.4../y...,0,1"1,0,(J )n HO "n OH
N HAc .
In certain such embodiments, conjugate groups have the following structure:

HO OH

HO----riC2.\.,,...N AL
AcHN 0 1 0 OH ---] I

AcHN OH 0 HOOH 9 y P-..

NHAc =
In certain such embodiments, conjugate groups have the following structure:
HOOH
HOki ,-, 0 n-k , AcHN
OH)n NH2 HOOH N_______µ
0 0-, 0 / N
HN¨cm.,:j AcHN OH OH 0s, HOOH 0 (:) )n 6 HO¨P=0 OH
NHAc .
In certain such embodiments, conjugate groups have the following structure:

HO OH
0 , 0 HOK
AcHN o 1 0-___.
OH
HO OH,J\T NH2 0 0,_ 0 HO___,,,..r..C2,\,0 , AcHN OH (:) OH u ,.,.==
HO-P=0 HO OH 9 y P, O

(;,- 6X
HO
NHAc =
In certain such embodiments, conjugate groups have the following structure:
I ,,I\I NH2 HO¨P=0 I V _ZrµN
0 __ ,(OrN
N-_-:---/
U
I
HO-P=0 O
HO OH (11 0 , 0 ' 'n 0 I 0 \OH
AcHN OH 1) 0 HO OH n (on 0 0-...,. 0 HO C)\ ,V'" I
'n 0 I 0--1,z0-iF'=0 AcHN OH
(:) OH
HO H 01 1 t \
iy 0 ,Põ,k- ) 1 o n HO '- in OH
NHAc =
In certain such embodiments, conjugate groups have the following structure:

HO-P=0 ________________________________________________________________ 0 C/N¨rµN, HO¨P=0 \OH
AcHN 0 0, (03 HOOH

HO õ
0 0" I
AcHN OH O OH
HO OH 9 y P-OH
HO
NHAc In certain embodiments, conjugates do not comprise a pyrrolidine.
In certain such embodiments, conjugate groups have the following structure:

HOOH

(N`=-"N(:) 0=P-0-AcHN 0 0, I, 3 HO_,0 N"
H
AcHN 0 0O¨

HN
bH
HOOH

HO(:)-W---rN

AcHN
=
In certain such embodiments, conjugate groups have the following structure:

HOOH
-........õ...--...õ_,-\ ,..p AcHN
0- ---.
HOOH
_....72..\;-, 0 0, 9 AcHN 0' d NHAc In certain such embodiments, conjugate groups have the following structure:
HO OH

AcHN '---)r-N...--\.:Ni H H 00.
HO OHo N-(CH2)6-0-p¨

H I I
HO/r-V 0 0 0- 0 NHAc HNIrNHN--e0 OH /¨/-0 H0)\,>/
:) HO
NHAc .
In certain such embodiments, conjugate groups have the following structure:

HO--+E)-\CYTCH
AcHN N

__,.."õEj.....-Or N.A...,.....------0....N..k.,......--...,.....)k, AcHN 07 HO OH
HOOrN"C-10 AcHN .
In certain such embodiments, conjugate groups have the following structure:

AcHN N0 4 H--LL------ --------.'HN...IL.---------)(HN ¨ol:')¨
Z
AcHN 0 HOOH
HO-AcHN .
In certain such embodiments, conjugate groups have the following structure:
HOOH H
AcHN

HO "4 H N,i 0 El H H
AcHN
HOOH
N-ro H0.4._\/) 01¨H
AcHN .
In certain such embodiments, conjugate groups have the following structure:
HOOH H
_.....r..?...vorN

''0'.../
AcHN

H HIN "4 g-1 AcHN
HOOH
N--( HO__.....r.L.v) Or 0TCH
AcHN .
In certain such embodiments, conjugate groups have the following structure:

OH OH

AcHN
OH OH
0 ,crH H 0 NYLI\IrN-6 EMI
AcHN 0 0 HOOH
0 r NHAc In certain such embodiments, conjugate groups have the following structure:
OH OH

AcHN
OH OH
AcHNHOOH

0 r NHAc In certain such embodiments, conjugate groups have the following structure:
pH
HO OH

AcHN
0=P-OH
HO OH
HO
AcHN
0=P-OH
HO OH
HO EMI
AcHN
In certain such embodiments, conjugate groups have the following structure:

pH
HOOH

AcHN
0=P¨OH
HOOH

AcHN
0=P¨OH
HOOH

AcHN 6 .
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
HOS'0 n AcHN \O
HOOH
HO
AcHN

HOOH X/
HO
AcHN
wherein X is a substituted or unsubstituted tether of six to eleven consecutively bonded atoms.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
HOS'0 n AcHN \O
HOOH
HO
AcHN

HOOH X/
HO
AcHN
wherein X is a substituted or unsubstituted tether of ten consecutively bonded atoms.

In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
HO---C)X
AcHN \

H
AcHN Z
JO
/
HOOH X
_.....72.\.,,,,o/
HO
AcHN
wherein X is a substituted or unsubstituted tether of four to eleven consecutively bonded atoms and wherein the tether comprises exactly one amide bond.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
_...7.2..\ N

HO YN A
z, AcHN N -uN

HO H
AcHN H H
,Nõ.1(Z----OZ
HO OH Y

HO
AcHN
wherein Y and Z are independently selected from a C1-C12 substituted or unsubstituted alkyl, alkenyl, or alkynyl group, or a group comprising an ether, a ketone, an amide, an ester, a carbamate, an amine, a piperidine, a phosphate, a phosphodiester, a phosphorothioate, a triazole, a pyrrolidine, a disulfide, or a thioether.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:

HOOH

Y, AcHN N z¨uN

HO
AcHN H H
HO OH
HO_ (:) AcHN
wherein Y and Z are independently selected from a C1-C12 substituted or unsubstituted alkyl group, or a group comprising exactly one ether or exactly two ethers, an amide, an amine, a piperidine, a phosphate, a phosphodiester, or a phosphorothioate.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH

Y, AcHN N z¨uN

HO
AcHN H H
HO OH

HO
AcHN
wherein Y and Z are independently selected from a C i-C12 substituted or unsubstituted alkyl group.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:

NAm:o, HO
AcHN 0 HO OH ON A
)1-e)/n HO
AcHN n 0 HO_ 0 AcHN
wherein m and n are independently selected from 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:

NA(1,0 HO
AcHN 0 HOOH
j+-N)11-i1C)FiNA
HO 0 m H
H_4,1Z
AcHN NO

HOOF Ic( HO
AcHN
wherein m is 4, 5, 6, 7, or 8, and n is 1, 2, 3, or 4.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
HOOH
FIC)C) o AcHN
HO
AcHN
OH0H r H
AcHN
wherein X is a substituted or unsubstituted tether of four to thirteen consecutively bonded atoms, and wherein X does not comprise an ether group.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
HOOH
FIC)C) o AcHN
HO
AcHN
OH0H r H
AcHN
wherein X is a substituted or unsubstituted tether of eight consecutively bonded atoms, and wherein X does not comprise an ether group.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:

HOOH
HOOH
AcHN
__,72..\___ 1/4---0 __________________ HO X-----\_k_ `x N
AcHN
OH0H r H
_.--X

AcHN
wherein X is a substituted or unsubstituted tether of four to thirteen consecutively bonded atoms, and wherein the tether comprises exactly one amide bond, and wherein X does not comprise an ether group.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
HOOH
__,72..\,,0AcHN 1/4---HO
N
AcHN
OH0H r H

AcHN
wherein X is a substituted or unsubstituted tether of four to thirteen consecutively bonded atoms and wherein the tether consists of an amide bond and a substituted or unsubstituted C2-Ci1 alkyl group.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH H
HO_Zzo¨y---N......f0 AcHN

_......2.\/0----YN)C\ µ.
HO H N
H
AcHN
HOOH Ni---( HO_.=.72..\/0¨y------i_l 0 AcHN
wherein Y is selected from a C1-C12 substituted or unsubstituted alkyl, alkenyl, or alkynyl group, or a group comprising an ether, a ketone, an amide, an ester, a carbamate, an amine, a piperidine, a phosphate, a phosphodiester, a phosphorothioate, a triazole, a pyrrolidine, a disulfide, or a thioether.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:

HOOH
H
HO_,.r.?...v0¨y¨N.....,,e AcHN

__.....2.\/0---YN)C\ µ.
HO H N
H
AcHN
HOOH
HO__...4/0¨Y--1-1 O
AcHN
wherein Y is selected from a C1-C12 substituted or unsubstituted alkyl group, or a group comprising an ether, an amine, a piperidine, a phosphate, a phosphodiester, or a phosphorothioate.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
H
HO_,.r.?...v0¨y--N....,e AcHN

HO H N
H
AcHN
HOOH m--( HO__...4/0¨Y--1-1 O
AcHN
wherein Y is selected from a C i-C12 substituted or unsubstituted alkyl group.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HO OH H
HO \ irl-AcHN

HO -H
H
AcHN
I \ ....r.-HOOH
HO H
AcHN
Wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:

HOOH H
AcHN

HO
_ vcy ..T2..-N µ.
/nH Nr H
AcHN
HO
__,....f..2..vo 0 H
AcHN
wherein n is 4, 5, 6, 7, or 8.
b. Certain conjugated antisense compounds In certain embodiments, the conjugates are bound to a nucleoside of the antisense oligonucleotide at the 2', 3', of 5' position of the nucleoside. In certain embodiments, a conjugated antisense compound has the following structure:
A¨B¨C¨D¨EE¨F) q wherein A is the antisense oligonucleotide;
B is the cleavable moiety C is the conjugate linker D is the branching group each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, a conjugated antisense compound has the following structure:
A¨C¨DiE¨F) q wherein A is the antisense oligonucleotide;
C is the conjugate linker D is the branching group each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain such embodiments, the conjugate linker comprises at least one cleavable bond.
In certain such embodiments, the branching group comprises at least one cleavable bond.
In certain embodiments each tether comprises at least one cleavable bond.
In certain embodiments, the conjugates are bound to a nucleoside of the antisense oligonucleotide at the 2', 3', of 5' position of the nucleoside.
In certain embodiments, a conjugated antisense compound has the following structure:
A¨B¨CiE¨F) q wherein A is the antisense oligonucleotide;
B is the cleavable moiety C is the conjugate linker each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, the conjugates are bound to a nucleoside of the antisense oligonucleotide at the 2', 3', of 5' position of the nucleoside. In certain embodiments, a conjugated antisense compound has the following structure:
A ¨CiE¨F) q wherein A is the antisense oligonucleotide;
C is the conjugate linker each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, a conjugated antisense compound has the following structure:
A¨B¨DiE¨F) q wherein A is the antisense oligonucleotide;
B is the cleavable moiety D is the branching group each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, a conjugated antisense compound has the following structure:
A ¨DiE¨F) q wherein A is the antisense oligonucleotide;
D is the branching group each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain such embodiments, the conjugate linker comprises at least one cleavable bond.
In certain embodiments each tether comprises at least one cleavable bond.
In certain embodiments, a conjugated antisense compound has a structure selected from among the following:

Targeting moiety ASO
HO OH
_ O=P¨OH NH2 OH
HN
\,,.0,7=N -N--,J
HO
NHAc 0 1 I ' _ 0-2_ HO N
N ci 0 y,,,_ :0 -,i N_ 0 0 __ -c, OH
NHAc 0 0 ,--_ 0 Linker Cleavable moiety Ligand Tether , I i ¨

_s......7C.3....\ ,70 /1 Branching group NHAc =
In certain embodiments, a conjugated antisense compound has a structure selected from among the following:
Cell targeting moiety - Cleavable moiety -AcHN
OH
_ , __ , lz____<
HO OH
0 0-._ 0 zr ) HO '-' 0:1-13-0,0rN

---iN
_ AcHN _ _ OH _0"--- 0 Tether _________________________________________________ , -0-1"=0 Ligand A

-HO OH 1, P- ASO
HO OH
NHAc Branching group =
In certain embodiments, a conjugated antisense compound has a structure selected from among the following:

ASO Cleavable moiety ,2412 HO¨P=0 1\1 I 4 n\T
d I
HO¨P=0 Cell targeting moiety I
' 0 ' AcHN 0- 0 , (()3 HO OH _ i _____ Conjugate 0 0-, 0 I linker u 0-u AcHN _ 0--- - OH
______ 1 I
Tether , Ligand 0 y H A
HO H fL.
\.i(2..\/00-6_ 0 O
NHAc Branching group In certain embodiments, the conjugated antisense compound has the following structure:

NI---)"---N
HO OH 0 ,y0-F' =0 I ,) Al 0 FINP c, 0 N N NO
HO -==="7`Cµ'-'tn N't 1_0_i _ ---\ _0_)/
---,ir NH N csi 0 HO OH 0 O N 0 e 0 o) NH2 e 0 S-P =0 Oct;ljt:N---FINH2 S-P =0 Ai -,iiNH OZ

0 0,...--i NH2 N N
---j=,--e S10 S-F'=0 j l N
HO 0---r-N1 0 4 H Al 0 N

)NHW

0 )_0_/0 e 0 oe) 0 S-F'=0 S-P =0i 0 o1 llINZ
----1_01 N NH2 N ) 0 0_/ , o es-P=O Aftt O o=)o o1 es-F1' =0 1[I'NH
lc_0_ ,, 0 0 ill'NH
O 0 0,) NH2 S-F'=0 N'-'0 S-F'=0A 0 1-(-- N ).-0-;
____________________________________________________________________ 0 NO
_CL)/ 0 0 O e s-p-o 1[1 NH
P
1 S-F'=0 8 4' NH NO
(:)_)/ 0,, 0 0 (:) ) 0 e o 5=O S-P =0 4-NH
S-F'=0 ''''CLLOIH 1 0N-^-0 N
CDS-CID)=0 (1111-11'oN":11-FINH2 0' (DI

NI)", N
0 S-P =0 1 N N
)_5 0 0 o_=

S-P =0 IF1 ,/ e 0 -= ,N 0 S-F'=0 2.-i o 5-F=O
OH 0,_,-i _____________________________________________________________________________ .
Representative United States patents, United States patent application publications, and international patent application publications that teach the preparation of certain of the above noted conjugates, conjugated antisense compounds, tethers, linkers, branching groups, ligands, cleavable moieties as well as other modifications include without limitation, US 5,994,517, US 6,300,319, US
6,660,720, US 6,906,182, US
7,262,177, US 7,491,805, US 8,106,022, US 7,723,509, US 2006/0148740, US
2011/0123520, WO
2013/033230 and WO 2012/037254, each of which is incorporated by reference herein in its entirety.
Representative publications that teach the preparation of certain of the above noted conjugates, conjugated antisense compounds, tethers, linkers, branching groups, ligands, cleavable moieties as well as other modifications include without limitation, BIESSEN et al., "The Cholesterol Derivative of a Triantennary Galactoside with High Affinity for the Hepatic Asialoglycoprotein Receptor: a Potent Cholesterol Lowering Agent" J. Med. Chem. (1995) 38:1846-1852, BIESSEN et al., "Synthesis of Cluster Galactosides with High Affinity for the Hepatic Asialoglycoprotein Receptor"
J. Med. Chem. (1995) 38:1538-1546, LEE et al., "New and more efficient multivalent glyco-ligands for asialoglycoprotein receptor of mammalian hepatocytes" Bioorganic & Medicinal Chemistry (2011) 19:2494-2500, RENSEN et al., "Determination of the Upper Size Limit for Uptake and Processing of Ligands by the Asialoglycoprotein Receptor on Hepatocytes in Vitro and in Vivo" J. Biol. Chem. (2001) 276(40):37577-37584, RENSEN et al., "Design and Synthesis of Novel N-Acetylgalactosamine-Terminated Glycolipids for Targeting of Lipoproteins to the Hepatic Asialoglycoprotein Receptor" J. Med. Chem. (2004) 47:5798-5808, SLIEDREGT
et al., "Design and Synthesis of Novel Amphiphilic Dendritic Galactosides for Selective Targeting of Liposomes to the Hepatic Asialoglycoprotein Receptor" J. Med. Chem. (1999) 42:609-618, and Valentijn et al., "Solid-phase synthesis of lysine-based cluster galactosides with high affinity for the Asialoglycoprotein Receptor" Tetrahedron, 1997, 53(2), 759-770, each of which is incorporated by reference herein in its entirety.
In certain embodiments, conjugated antisense compounds comprise an RNase H
based oligonucleotide (such as a gapmer) or a splice modulating oligonucleotide (such as a fully modified oligonucleotide) and any conjugate group comprising at least one, two, or three GalNAc groups. In certain embodiments a conjugated antisense compound comprises any conjugate group found in any of the following references: Lee, Carbohydr Res, 1978, 67, 509-514; Connolly et al., J Biol Chem, 1982, 257, 939-945; Pavia et al., Int J Pep Protein Res, 1983, 22, 539-548; Lee et al., Biochem, 1984, 23, 4255-4261; Lee et al., Glycoconjugate J, 1987, 4, 317-328; Toyokuni et al., Tetrahedron Lett, 1990, 31, 2673-2676; Biessen et al., J
Med Chem, 1995, 38, 1538-1546; Valentijn et al., Tetrahedron, 1997, 53, 759-770; Kim et al., Tetrahedron Lett, 1997, 38, 3487-3490; Lee et al., Bioconjug Chem, 1997, 8, 762-765; Kato et al., Glycobiol, 2001, 11, 821-829; Rensen et al., J Biol Chem, 2001, 276, 37577-37584; Lee et al., Methods Enzymol, 2003, 362, 38-43; Westerlind et al., Glycoconj J, 2004, 21, 227-241; Lee et al., Bioorg Med Chem Lett, 2006, 16(19), 5132-5135; Maierhofer et al., Bioorg Med Chem, 2007, 15, 7661-7676; Khorev et al., Bioorg Med Chem, 2008, 16, 5216-5231; Lee et al., Bioorg Med Chem, 2011, 19, 2494-2500; Kornilova et al., Analyt Biochem, 2012, 425, 43-46; Pujol et al., Angew Chemie Int Ed Engl, 2012, 51, 7445-7448; Biessen et al., J Med Chem, 1995, 38, 1846-1852; Sliedregt et al., J Med Chem, 1999, 42, 609-618; Rensen et al., J
Med Chem, 2004, 47, 5798-5808; Rensen et al., Arterioscler Thromb Vasc Biol, 2006, 26, 169-175; van Rossenberg et al., Gene Ther, 2004, 11, 457-464; Sato et al., J Am Chem Soc, 2004, 126, 14013-14022; Lee et al., J Org Chem, 2012, 77, 7564-7571; Biessen et al., FASEB J, 2000, 14, 1784-1792; Rajur et al., Bioconjug Chem, 1997, 8, 935-940;
Duff et al., Methods Enzymol, 2000, 313, 297-321; Maier et al., Bioconjug Chem, 2003, 14, 18-29;
Jayaprakash et al., Org Lett, 2010, 12, 5410-5413; Manoharan, Antisense Nucleic Acid Drug Dev, 2002, 12, 103-128; Merwin et al., Bioconjug Chem, 1994, 5, 612-620; Tomiya et al., Bioorg Med Chem, 2013, 21, 5275-5281; International applications W01998/013381; W02011/038356;
W01997/046098;
W02008/098788; W02004/101619; W02012/037254; W02011/120053;
W02011/100131;
W02011/163121; W02012/177947; W02013/033230; W02013/075035;
W02012/083185;
W02012/083046; W02009/082607; W02009/134487; W02010/144740; W02010/148013;
W01997/020563; W02010/088537; W02002/043771; W02010/129709; W02012/068187;
W02009/126933; W02004/024757; W02010/054406; W02012/089352; W02012/089602;
W02013/166121; W02013/165816; U.S. Patents 4,751,219; 8,552,163; 6,908,903;
7,262,177; 5,994,517;
6,300,319; 8,106,022; 7,491,805; 7,491,805; 7,582,744; 8,137,695; 6,383,812;
6,525,031; 6,660,720;
7,723,509; 8,541,548; 8,344,125; 8,313,772; 8,349,308; 8,450,467; 8,501,930;
8,158,601; 7,262,177;
6,906,182; 6,620,916; 8,435,491; 8,404,862; 7,851,615; Published U.S. Patent Application Publications US2011/0097264; US2011/0097265; U52013/0004427; U52005/0164235;
U52006/0148740;
U52008/0281044; U52010/0240730; US2003/0119724; U52006/0183886;
U52008/0206869;
US2011/0269814; U52009/0286973; US2011/0207799; U52012/0136042;
U52012/0165393;
U52008/0281041; U52009/0203135; U52012/0035115; U52012/0095075;
U52012/0101148;
U52012/0128760; US2012/0157509; U52012/0230938; U52013/0109817;
U52013/0121954;
U52013/0178512; U52013/0236968; U52011/0123520; U52003/0077829;
U52008/0108801; and U52009/0203132; each of which is incorporated by reference in its entirety.
C. Certain Uses and Features In certain embodiments, conjugated antisense compounds exhibit potent target RNA reduction in vivo. In certain embodiments, unconjugated antisense compounds accumulate in the kidney. In certain embodiments, conjugated antisense compounds accumulate in the liver. In certain embodiments, conjugated antisense compounds are well tolerated. Such properties render conjugated antisense compounds particularly useful for inhibition of many target RNAs, including, but not limited to those involved in metabolic, cardiovascular and other diseases, disorders or conditions. Thus, provided herein are methods of treating such diseases, disorders or conditions by contacting liver tissues with the conjugated antisense compounds targeted to RNAs associated with such diseases, disorders or conditions. Thus, also provided are methods for ameliorating any of a variety of metabolic, cardiovascular and other diseases, disorders or conditions with the conjugated antisense compounds of the present invention.
In certain embodiments, conjugated antisense compounds are more potent than unconjugated counterpart at a particular tissue concentration. Without wishing to be bound by any theory or mechanism, in certain embodiemtns, the conjugate may allow the conjugated antisense compound to enter the cell more efficiently or to enter the cell more productively. For example, in certain embodiments conjugated antisense compounds may exhibit greater target reduction as compared to its unconjugated counterpart wherein both the conjugated antisense compound and its unconjugated counterpart are present in the tissue at the same concentrations. For example, in certain embodiments conjugated antisense compounds may exhibit greater target reduction as compared to its unconjugated counterpart wherein both the conjugated antisense compound and its unconjugated counterpart are present in the liver at the same concentrations.
Productive and non-productive uptake of oligonucleotides has beed discussed previously (See e.g.
Geary, R. S., E. Wancewicz, et al. (2009). "Effect of Dose and Plasma Concentration on Liver Uptake and Pharmacologic Activity of a 2'-Methoxyethyl Modified Chimeric Antisense Oligonucleotide Targeting PTEN." Biochem. Pharmacol. 78(3): 284-91; & Koller, E., T. M. Vincent, et al.
(2011). "Mechanisms of single-stranded phosphorothioate modified antisense oligonucleotide accumulation in hepatocytes." Nucleic Acids Res. 39(11): 4795-807). Conjugate groups described herein may improve productive uptake.
In certain embodiments, the conjugate groups described herein may further improve potency by increasing the affinity of the conjugated antisense compound for a particular type of cell or tissue. In certain embodiments, the conjugate groups described herein may further improve potency by increasing recognition of the conjugated antisense compound by one or more cell-surface receptors. .
In certain embodiments, the conjugate groups described herein may further improve potency by facilitating endocytosis of the conjugated antisense compound.
In certain embodiments, the cleavable moiety may further improve potency by allowing the conjugate to be cleaved from the antisense oligonucleotide after the conjugated antisense compound has entered the cell. Accordingly, in certain embodiments, conjugated antisense compounds can be administed at doses lower than would be necessary for unconjugated antisense oligonucleotides.
Phosphorothioate linkages have been incorporated into antisense oligonucleotides previously. Such phosphorothioate linkages are resistant to nucleases and so improve stability of the oligonucleotide. Further, phosphorothioate linkages also bind certain proteins, which results in accumulation of antisense oligonucleotide in the liver. Oligonucleotides with fewer phosphorothioate linkages accumulate less in the liver and more in the kidney (see, for example, Geary, R., "Pharmacokinetic Properties of 2'4)-(2-Methoxyethyl)-Modified Oligonucleotide Analogs in Rats," Journal of Pharmacology and Experimental Therapeutics, Vol. 296, No. 3, 890-897; & Pharmacological Properties of 2 '-0-Methoxyethyl Modified Oligonucleotides in Antisense a Drug Technology, Chapter 10, Crooke, S.T., ed., 2008) In certain embodiments, oligonucleotides with fewer phosphorothioate internculeoside linkages and more phosphodiester internucleoside linkages accumulate less in the liver and more in the kidney. When treating diseases in the liver, this is undesibable for several reasons (1) less drug is getting to the site of desired action (liver); (2) drug is escaping into the urine; and (3) the kidney is exposed to relatively high concentration of drug which can result in toxicities in the kidney. Thus, for liver diseases, phosphorothioate linkages provide important benefits.

In certain embodiments, however, administration of oligonucleotides uniformly linked by phosphoro-thioate internucleoside linkages induces one or more proinflammatory reactions. (see for example: J Lab Clin Med. 1996 Sep;128(3):329-38. "Amplification of antibody production by phosphorothioate oligodeoxynucleotides". Branda et al.; and see also for example: Toxicologic Properties in Antisense a Drug Technology, Chapter 12, pages 342-351, Crooke, S.T., ed., 2008). In certain embodiments, administration of oligonucleotides wherein most of the internucleoside linkages comprise phosphorothioate internucleoside linkages induces one or more proinflammatory reactions.
In certain embodiments, the degree of proinflammatory effect may depend on several variables (e.g.
backbone modification, off-target effects, nucleobase modifications, and/or nucleoside modifications) see for example: Toxicologic Properties in Antisense a Drug Technology, Chapter 12, pages 342-351, Crooke, S.T., ed., 2008). In certain embodiments, the degree of proinflammatory effect may be mitigated by adjusting one or more variables. For example the degree of proinflammatory effect of a given oligonucleotide may be mitigated by replacing any number of phosphorothioate internucleoside linkages with phosphodiester internucleoside linkages and thereby reducing the total number of phosphorothioate internucleoside linkages.
In certain embodiments, it would be desirable to reduce the number of phosphorothioate linkages, if doing so could be done without losing stability and without shifting the distribution from liver to kidney. For example, in certain embodiments, the number of phosphorothioate linkages may be reduced by replacing phosphorothioate linkages with phosphodiester linkages. In such an embodiment, the antisense compound having fewer phosphorothioate linkages and more phosphodiester linkages may induce less proinflammatory reactions or no proinflammatory reaction. Although the the antisense compound having fewer phosphoro-thioate linkages and more phosphodiester linkages may induce fewer proinflammatory reactions, the antisense compound having fewer phosphorothioate linkages and more phosphodiester linkages may not accumulate in the liver and may be less efficacious at the same or similar dose as compared to an antisense compound having more phosphorothioate linkages. In certain embodiments, it is therefore desirable to design an antisense compound that has a plurality of phosphodiester bonds and a plurality of phosphorothioate bonds but which also possesses stability and good distribution to the liver.
In certain embodiments, conjugated antisense compounds accumulate more in the liver and less in the kidney than unconjugated counterparts, even when some of the phosporothioate linkages are replaced with less proinflammatory phosphodiester internucleoside linkages. In certain embodiments, conjugated antisense compounds accumulate more in the liver and are not excreted as much in the urine compared to its unonjugated counterparts, even when some of the phosporothioate linkages are replaced with less proinflammatory phosphodiester internucleoside linkages. In certain embodiments, the use of a conjugate allows one to design more potent and better tolerated antisense drugs. Indeed, in certain emobidments, conjugated antisense compounds have larger therapeutic indexes than unconjugated counterparts. This allows the conjugated antisense compound to be administered at a higher absolute dose, because there is less risk of proinflammatory response and less risk of kidney toxicity. This higher dose, allows one to dose less frequently, since the clearance (metabolism) is expected to be similar.
Further, because the compound is more potent, as described above, one can allow the concentration to go lower before the next dose without losing therapeutic activity, allowing for even longer periods between dosing.
In certain embodiments, the inclusion of some phosphorothioate linkages remains desirable. For example, the terminal linkages are vulnerable to exonucleoases and so in certain embodiments, those linkages are phosphorothioate or other modified linkage. Internucleoside linkages linking two deoxynucleosides are vulnerable to endonucleases and so in certain embodiments those those linkages are phosphorothioate or other modified linkage. Internucleoside linkages between a modified nucleoside and a deoxynucleoside where the deoxynucleoside is on the 5' side of the linkage deoxynucleosides are vulnerable to endonucleases and so in certain embodiments those those linkages are phosphorothioate or other modified linkage.
Internucleoside linkages between two modified nucleosides of certain types and between a deoxynucleoside and a modified nucleoside of certain typ where the modified nucleoside is at the 5' side of the linkage are sufficiently resistant to nuclease digestion, that the linkage can be phosphodiester.
In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 16 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 15 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 14 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 13 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 12 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 11 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 10 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 9 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 8 phosphorthioate linkages.
In certain embodiments, antisense compounds comprsing one or more conjugae group described herein has increased activity and/or potency and/or tolerability compared to a parent antisense compound lacking such one or more conjugate group. Accordingly, in certain embodiments, attachment of such conjugate groups to an oligonucleotide is desirable. Such conjugate groups may be attached at the 5'-, and/or 3'- end of an oligonucleotide. In certain instances, attachment at the 5'-end is synthetically desireable.
Typically, oligonucleietides are synthesized by attachment of the 3' terminal nucleoside to a solid support and sequential coupling of nucleosides from 3' to 5' using techniques that are well known in the art.
Accordingly if a conjugate group is desred at the 3'-terminus, one may (1) attach the conjugate group to the 3'-terminal nucleoside and attach that conjugated nucleoside to the solid support for subsequent preparation of the oligonucleotide or (2) attach the conjugate group to the 3'-terminal nucleoside of a completed oligonucleotide after synthesis. Niether of these approaches is very efficient and thus both are costly. In particular, attachment of the conjugated nucleoside to the solid support, while demonstrated in the Examples herein, is an inefficient process. In certain embodiments, attaching a conjugate group to the 5'-terminal nucleoside is synthetically easier than attachment at the 3'-end. One may attach a non-conjugated 3' terminal nucleoside to the solid support and prepare the oligonucleotide using standard and well characterized reastions. One then needs only to attach a 5'nucleoside having a conjugate group at the final coupling step.
In certain embodiments, this is more efficient than attaching a conjugated nucleoside directly to the solid support as is typically done to prepare a 3'-conjugated oligonucleotide. The Examples herein demonstrate attachment at the 5'-end. In addition, certain conjugate groups have synthetic advantages. For Example, certain conjugate groups comprising phosphorus linkage groups are synthetically simpler and more efficiently prepared than other conjugate groups, including conjugate groups reported previously (e.g., WO/2012/037254).
In certain embodiments, conjugated antisense compounds are administered to a subject. In such embodiments, antisense compounds comprsing one or more conjugae group described herein has increased activity and/or potency and/or tolerability compared to a parent antisense compound lacking such one or more conjugate group. Without being bound by mechanism, it is believed that the conjugate group helps with distribution, delivery, and/or uptake into a target cell or tissue. In certain embodiments, once inside the target cell or tissue, it is desirable that all or part of the conjugate group to be cleaved to releas the active oligonucleitde. In certain embodiments, it is not necessary that the entire conjugate group be cleaved from the oligonucleotide. For example, in emort*20 a conjugated oligonucleotide was administered to mice and a number of different chemical species, each comprising a different portion of the conjugate group remaining on the oligonucleotide, were detected (Wit 0234). Thisconjugated antisense compound demonstrated good potency (Db1013). Thus, in certain embodiments, such metabolite profile of multiple partial cleavage of the conjugate group does not interfere with activity/potency. Nevertheless, in certain embodiments it is desirable that a prodrug (conjugated oligonucleotide) yield a single active compound. In certain instances, if multiple forms of the active compound are found, it may be necessary to determine relative amounts and activities for each one. In certain embodiments where regulatory review is required (e.g., USFDA or counterpart) it is desirable to have a single (or predominantly single) active species. In certain such embodiments, it is desirable that such single active species be the antisense oligonucleotide lacking any portion of the conjugate group. In certain embodiments, conjugate groups at the 5'-end are more likely to result in complete metabolism of the conjugate group. Without being bound by mechanism it may be that endogenous enzymes responsible for metabolism at the 5' end (e.g., 5' nucleases) are more active/efficient than the 3' counterparts.
In certain embodiments, the specific conjugate groups are more amenable to metabolism to a single active species. In certain embodiments, certain conjugate groups are more amenable to metabolism to the oligonucleotide.
D. Antis ens e In certain embodiments, oligomeric compounds of the present invention are antisense compounds.
In such embodiments, the oligomeric compound is complementary to a target nucleic acid. In certain embodiments, a target nucleic acid is an RNA. In certain embodiments, a target nucleic acid is a non-coding RNA. In certain embodiments, a target nucleic acid encodes a protein. In certain embodiments, a target nucleic acid is selected from a mRNA, a pre-mRNA, a microRNA, a non-coding RNA, including small non-coding RNA, and a promoter-directed RNA. In certain embodiments, oligomeric compounds are at least partially complementary to more than one target nucleic acid. For example, oligomeric compounds of the present invention may be microRNA mimics, which typically bind to multiple targets.
In certain embodiments, antisense compounds comprise a portion having a nucleobase sequence at least 70% complementary to the nucleobase sequence of a target nucleic acid.
In certain embodiments, antisense compounds comprise a portion having a nucleobase sequence at least 80% complementary to the nucleobase sequence of a target nucleic acid. In certain embodiments, antisense compounds comprise a portion having a nucleobase sequence at least 90% complementary to the nucleobase sequence of a target nucleic acid. In certain embodiments, antisense compounds comprise a portion having a nucleobase sequence at least 95% complementary to the nucleobase sequence of a target nucleic acid. In certain embodiments, antisense compounds comprise a portion having a nucleobase sequence at least 98%
complementary to the nucleobase sequence of a target nucleic acid. In certain embodiments, antisense compounds comprise a portion having a nucleobase sequence that is 100%
complementary to the nucleobase sequence of a target nucleic acid. In certain embodiments, antisense compounds are at least 70%, 80%, 90%, 95%, 98%, or 100% complementary to the nucleobase sequence of a target nucleic acid over the entire length of the antisense compound.
Antisense mechanisms include any mechanism involving the hybridization of an oligomeric compound with target nucleic acid, wherein the hybridization results in a biological effect. In certain embodiments, such hybridization results in either target nucleic acid degradation or occupancy with concomitant inhibition or stimulation of the cellular machinery involving, for example, translation, transcription, or polyadenylation of the target nucleic acid or of a nucleic acid with which the target nucleic acid may otherwise interact.
One type of antisense mechanism involving degradation of target RNA is RNase H
mediated antisense. RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. It is known in the art that single-stranded antisense compounds which are "DNA-like"
elicit RNase H activity in mammalian cells. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of DNA-like oligonucleotide-mediated inhibition of gene expression.
Antisense mechanisms also include, without limitation RNAi mechanisms, which utilize the RISC
pathway. Such RNAi mechanisms include, without limitation siRNA, ssRNA and microRNA mechanisms.
Such mechanisms include creation of a microRNA mimic and/or an anti-microRNA.
Antisense mechanisms also include, without limitation, mechanisms that hybridize or mimic non-coding RNA other than microRNA or mRNA. Such non-coding RNA includes, but is not limited to promoter-directed RNA and short and long RNA that effects transcription or translation of one or more nucleic acids.
In certain embodiments, oligonucleotides comprising conjugates described herein are RNAi compounds. In certain embodiments, oligomeric oligonucleotides comprising conjugates described herein are ssRNA compounds. In certain embodiments, oligonucleotides comprising conjugates described herein are paired with a second oligomeric compound to form an siRNA. In certain such embodiments, the second oligomeric compound also comprises a conjugate. In certain embodiments, the second oligomeric compound is any modified or unmodified nucleic acid. In certain embodiments, the oligonucleotides comprising conjugates described herein is the antisense strand in an siRNA compound. In certain embodiments, the oligonucleotides comprising conjugates described herein is the sense strand in an siRNA compound. In embodiments in which the conjugated oligomeric compound is double-stranded siRnA, the conjugate may be on the sense strand, the antisense strand or both the sense strand and the antisense strand.
D. Apolipoprotein C-III (apoCIII) In certain embodiments, conjugated antisense compounds target any ApoCIII
nucleic acid. In certain embodiments, the target nucleic acid encodes an ApoCIII target protein that is clinically relevant. In such embodiments, modulation of the target nucleic acid results in clinical benefit.
The targeting process usually includes determination of at least one target region, segment, or site within the target nucleic acid for the antisense interaction to occur such that the desired effect will result.
In certain embodiments, a target region is a structurally defined region of the nucleic acid. For example, in certain such embodiments, a target region may encompass a 3' UTR, a 5' UTR, an exon, an intron, a coding region, a translation initiation region, translation termination region, or other defined nucleic acid region or target segment.
In certain embodiments, a target segment is at least about an 8-nucleobase portion of a target region to which a conjugated antisense compound is targeted. Target segments can include DNA or RNA sequences that comprise at least 8 consecutive nucleobases from the 5'-terminus of one of the target segments (the remaining nucleobases being a consecutive stretch of the same DNA or RNA
beginning immediately upstream of the 5'-terminus of the target segment and continuing until the DNA
or RNA comprises about 8 to about 30 nucleobases). Target segments are also represented by DNA or RNA
sequences that comprise at least 8 consecutive nucleobases from the 3'-terminus of one of the target segments (the remaining nucleobases being a consecutive stretch of the same DNA or RNA beginning immediately downstream of the 3'-terminus of the target segment and continuing until the DNA or RNA
comprises about 8 to about 30 nucleobases). Target segments can also be represented by DNA or RNA sequences that comprise at least 8 consecutive nucleobases from an internal portion of the sequence of a target segment, and may extend in either or both directions until the conjugated antisense compound comprises about 8 to about 30 nucleobases.
In certain embodiments, antisense compounds targeted to an ApoCIII nucleic acid can be modified as described herein. In certain embodiments, the antisense compounds can have a modified sugar moiety, an unmodified sugar moiety or a mixture of modified and unmodified sugar moieties as described herein. In certain embodiments, the antisense compounds can have a modified internucleoside linkage, an unmodified internucleoside linkage or a mixture of modified and unmodified internucleoside linkages as described herein. In certain embodiments, the antisense compounds can have a modified nucleobase, an unmodified nucleobase or a mixture of modified and unmodified nucleobases as described herein. In certain embodiments, the antisense compounds can have a motif as described herein.
In certain embodiments, antisense compounds targeted to ApoCIII nucleic acids can be conjugated as described herein.
ApoCIII is a constituent of HDL and of triglyceride (TG)-rich lipoproteins.
Elevated ApoCIII levels are associated with elevated TG levels and diseases such as cardiovascular disease, metabolic syndrome, obesity and diabetes. Elevated TG levels are associated with pancreatitis.
ApoCIII slows clearance of TG-rich lipoproteins by inhibiting lipolysis through inhibition of lipoprotein lipase (LPL) and through interfering with lipoprotein binding to cell-surface glycosaminoglycan matrix. Antisense compounds targeting ApoCIII have been previously disclosed in W02004/093783 and W02012/149495, each herein incorporated by reference in its entirety.
Certain Conjugated Antisense Compounds Targeted to an ApoCIII Nucleic Acid In certain embodiments, conjugated antisense compounds are targeted to an ApoCIII nucleic acid having the sequence of any of GENBANKO Accession No. NM_000040.1 (incorporated herein as SEQ ID
NO: 1); GENBANK Accession No. NT 033899.8 truncated from nucleotides 20262640 to 20266603 (incorporated herein as SEQ ID NO: 2); and GenBank Accession No. NT_035088.1 truncated from nucleotides 6238608 to 6242565 (incorporated herein as SEQ ID NO: 3). In certain such embodiments, a conjugated antisense compound is at least 90%, at least 95%, or 100%
complementary to any of SEQ ID
NO s : 1-3.

In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
1 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 87. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 87.
Table A: Antisense Compounds targeted to ApoCIII SEQ ID NO: 1 Target Start SEQ ID
ISIS No Sequence (5'-3') Motif Site NO
304801 508 AGCTTCTTGTCCAGCTTTAT eeeeeddddddddddeeeee 87 647535 508 AGCTTCTTGTCCAGCTTTAT eeeeeddddddddddeeeeeod 87 616468 508 AGCTTCTTGTCCAGCTTTAT eeeeeddddddddddeeeee 87 647536 508 AGCTTCTTGTCCAGCTTTAT eeoeoeoeoddddddddddeoe87 oeeeod ApoCIII Therapeutic Indications In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to an ApoCIII nucleic acid for modulating the expression of ApoCIII
in a subject. In certain embodiments, the expression of ApoCIII is reduced.
In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to an ApoCIII nucleic acid in a pharmaceutical composition for treating a subject. In certain embodiments, the subject has a cardiovascular and/or metabolic disease, disorder or condition. In certain embodiments, the subject has hypertriglyceridemia, non-familial hypertriglyceridemia, familial hypertriglyceridemia, heterozygous familial hypertriglyceridemia, homozygous familial hypertriglyceridemia, mixed dyslipidemia, atherosclerosis, a risk of developing atherosclerosis, coronary heart disease, a history of coronary heart disease, early onset coronary heart disease, one or more risk factors for coronary heart disease, type II diabetes, type II diabetes with dyslipidemia, dyslipidemia, hyperlipidemia, hypercholesterolemia, hyperfattyacidemia, hepatic steatosis, non-alcoholic steatohepatitis, pancreatitis and/or non-alcoholic fatty liver disease.
In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to an ApoCIII nucleic acid in the preparation of a medicament.
E. Certain Nucleic Acid GaINAc Conjugates In certain embodiments, conjugated antisense compounds comprise antisense compounds having the nucleobase sequence and modifications of the antisense compounds in the Table below attached to a GalNAc conjugate. All internucleoside linkages are phosphorothioate internucleoside linkages unless otherwise indicated. A subscript "1" indicates an LNA bicyclic nucleoside. A
subscript "d" indicates a 2'-deoxy nucleoside. A subscript "e" indicates a 2'-MOE modified nucleoside. A
"V" indicates a 2-amino-2'-deoxyadenosine.
Table B
Sequence Motif Chemistry Internucleoside SEQ
5' to 3' Linkages ID
NO.
TIGIGICdAdAdGaCdAdTdCdCaTIGITIAd 3-9-3-1 LNA/deoxy phosphorothioate 222 CITICIAIAdTdCdCdAdTaGaGaCIAIGICa 4-8-3-1 LNA/deoxy phosphorothioate 223 AICICIAdAdGdTdTaTdCaTaTaCciAIGICI
3-10-3 LNA/deoxy phosphorothioate 224 GICIAdTdTaGaGaTdAdTaTICA
2-8-3 LNA/deoxy phosphorothioate 225 TITICIAIGICdAdTdTdGaGaTdAdTaTdCAGITIGI 5-10-5 LNA/deoxy phosphorothioate 226 CIAIGICdAdTdTaGaGaTdAdTaTICAGd 3-10-3 LNA/deoxy phosphorothioate 227 CIAIGICdAdTaTaGaGaTdAdTaTICA
3-9-3 LNA/deoxy phosphorothioate 228 AIGICIAdTdTdGaGaTdAdTaTICA
3-8-3 LNA/deoxy phosphorothioate 229 GICIAdTdTaGaGaTdAdTaTICI
2-8-2 LNA/deoxy phosphorothioate 230 CGGCATGTCTATTTTGTA
phosphorothioate 231 GGCTAAATCGCTCCACCAAG
phosphorothioate 232 CTCTAGCGTCTTAAAGCCGA
phosphorothioate 233 GCTGCATGATCTCCTTGGCG
phosphorothioate 234 ACGTTGAGGGGCATCGTCGC Morpholino GGGTCTGCVGCGGGVTGGT
phosphorothioate 236 GTTVCTVCTTCCVCCTGCCTG
phosphorothioate 237 TATCCGGAGGGCTCGCCATGCTGCT
phosphorothioate 238 TeC,C,C,G,C,CTGTGACATeGeCeAeTeTe 6-8-6 MOE/deoxy CeA,G,C,AGCAGAGTCTTCATeCeAeTe 4-13-4 MOE/deoxy GeGeG,A,CdGdCdGaGaCaGaCdTdCaGaGaTeCeA,T, 4-12-4 MOE/deoxy C,C,A,C,A,AdGdCaTaGaTdCdCdAdGaTeCeTeAeA, 5-10-5 MOE/deoxy C,C,G,CdAdGdCdCdAdTdGdCdGeCeTeCeTeTeGeGe 3-9-8 MOE/deoxy F. Certain Pharmaceutical Compositions In certain embodiments, the present disclosure provides pharmaceutical compositions comprising one or more antisense compound. In certain embodiments, such pharmaceutical composition comprises a suitable pharmaceutically acceptable diluent or carrier. In certain embodiments, a pharmaceutical composition comprises a sterile saline solution and one or more antisense compound. In certain embodiments, such pharmaceutical composition consists of a sterile saline solution and one or more antisense compound. In certain embodiments, the sterile saline is pharmaceutical grade saline. In certain embodiments, a pharmaceutical composition comprises one or more antisense compound and sterile water. In certain embodiments, a pharmaceutical composition consists of one or more antisense compound and sterile water.
In certain embodiments, the sterile saline is pharmaceutical grade water. In certain embodiments, a pharmaceutical composition comprises one or more antisense compound and phosphate-buffered saline (PBS). In certain embodiments, a pharmaceutical composition consists of one or more antisense compound and sterile phosphate-buffered saline (PBS). In certain embodiments, the sterile saline is pharmaceutical grade PBS.
In certain embodiments, antisense compounds may be admixed with pharmaceutically acceptable active and/or inert substances for the preparation of pharmaceutical compositions or formulations.
Compositions and methods for the formulation of pharmaceutical compositions depend on a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered.
Pharmaceutical compositions comprising antisense compounds encompass any pharmaceutically acceptable salts, esters, or salts of such esters. In certain embodiments, pharmaceutical compositions comprising antisense compounds comprise one or more oligonucleotide which, upon administration to an animal, including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof Accordingly, for example, the disclosure is also drawn to pharmaceutically acceptable salts of antisense compounds, prodrugs, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents. Suitable pharmaceutically acceptable salts include, but are not limited to, sodium and potassium salts.
A prodrug can include the incorporation of additional nucleosides at one or both ends of an oligonucleotide which are cleaved by endogenous nucleases within the body, to form the active antisense oligonucleotide.
Lipid moieties have been used in nucleic acid therapies in a variety of methods. In certain such methods, the nucleic acid is introduced into preformed liposomes or lipoplexes made of mixtures of cationic lipids and neutral lipids. In certain methods, DNA complexes with mono- or poly-cationic lipids are formed without the presence of a neutral lipid. In certain embodiments, a lipid moiety is selected to increase distribution of a pharmaceutical agent to a particular cell or tissue. In certain embodiments, a lipid moiety is selected to increase distribution of a pharmaceutical agent to fat tissue. In certain embodiments, a lipid moiety is selected to increase distribution of a pharmaceutical agent to muscle tissue.
In certain embodiments, pharmaceutical compositions provided herein comprise one or more modified oligonucleotides and one or more excipients. In certain such embodiments, excipients are selected from water, salt solutions, alcohol, polyethylene glycols, gelatin, lactose, amylase, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose and polyvinylpyrrolidone.
In certain embodiments, a pharmaceutical composition provided herein comprises a delivery system.
Examples of delivery systems include, but are not limited to, liposomes and emulsions. Certain delivery systems are useful for preparing certain pharmaceutical compositions including those comprising hydrophobic compounds. In certain embodiments, certain organic solvents such as dimethylsulfoxide are used.
In certain embodiments, a pharmaceutical composition provided herein comprises one or more tissue-specific delivery molecules designed to deliver the one or more pharmaceutical agents of the present disclosure to specific tissues or cell types. For example, in certain embodiments, pharmaceutical compositions include liposomes coated with a tissue-specific antibody.
In certain embodiments, a pharmaceutical composition provided herein comprises a co-solvent system. Certain of such co-solvent systems comprise, for example, benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. In certain embodiments, such co-solvent systems are used for hydrophobic compounds. A non-limiting example of such a co-solvent system is the VPD co-solvent system, which is a solution of absolute ethanol comprising 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant Polysorbate 8OTM and 65% w/v polyethylene glycol 300. The proportions of such co-solvent systems may be varied considerably without significantly altering their solubility and toxicity characteristics.
Furthermore, the identity of co-solvent components may be varied: for example, other surfactants may be used instead of Polysorbate 8OTM; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g., polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
In certain embodiments, a pharmaceutical composition provided herein is prepared for oral administration. In certain embodiments, pharmaceutical compositions are prepared for buccal administration.
In certain embodiments, a pharmaceutical composition is prepared for administration by injection (e.g., intravenous, subcutaneous, intramuscular, etc.). In certain of such embodiments, a pharmaceutical composition comprises a carrier and is formulated in aqueous solution, such as water or physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. In certain embodiments, other ingredients are included (e.g., ingredients that aid in solubility or serve as preservatives).
In certain embodiments, injectable suspensions are prepared using appropriate liquid carriers, suspending agents and the like. Certain pharmaceutical compositions for injection are presented in unit dosage form, e.g., in ampoules or in multi-dose containers. Certain pharmaceutical compositions for injection are suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Certain solvents suitable for use in pharmaceutical compositions for injection include, but are not limited to, lipophilic solvents and fatty oils, such as sesame oil, synthetic fatty acid esters, such as ethyl oleate or triglycerides, and liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, such suspensions may also contain suitable stabilizers or agents that increase the solubility of the pharmaceutical agents to allow for the preparation of highly concentrated solutions.
In certain embodiments, a pharmaceutical composition is prepared for transmucosal administration.
In certain of such embodiments penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
In certain embodiments, a pharmaceutical composition provided herein comprises an oligonucleotide in a therapeutically effective amount. In certain embodiments, the therapeutically effective amount is sufficient to prevent, alleviate or ameliorate symptoms of a disease or to prolong the survival of the subject being treated. Determination of a therapeutically effective amount is well within the capability of those skilled in the art.
In certain embodiments, one or more modified oligonucleotide provided herein is formulated as a prodrug. In certain embodiments, upon in vivo administration, a prodrug is chemically converted to the biologically, pharmaceutically or therapeutically more active form of an oligonucleotide. In certain embodiments, prodrugs are useful because they are easier to administer than the corresponding active form.
For example, in certain instances, a prodrug may be more bioavailable (e.g., through oral administration) than is the corresponding active form. In certain instances, a prodrug may have improved solubility compared to the corresponding active form. In certain embodiments, prodrugs are less water soluble than the corresponding active form. In certain instances, such prodrugs possess superior transmittal across cell membranes, where water solubility is detrimental to mobility. In certain embodiments, a prodrug is an ester.
In certain such embodiments, the ester is metabolically hydrolyzed to carboxylic acid upon administration. In certain instances the carboxylic acid containing compound is the corresponding active form. In certain embodiments, a prodrug comprises a short peptide (polyaminoacid) bound to an acid group. In certain of such embodiments, the peptide is cleaved upon administration to form the corresponding active form.
In certain embodiments, the present disclosure provides compositions and methods for reducing the amount or activity of a target nucleic acid in a cell. In certain embodiments, the cell is in an animal. In certain embodiments, the animal is a mammal. In certain embodiments, the animal is a rodent. In certain embodiments, the animal is a primate. In certain embodiments, the animal is a non-human primate. In certain embodiments, the animal is a human.
In certain embodiments, the present disclosure provides methods of administering a pharmaceutical composition comprising an oligonucleotide of the present disclosure to an animal. Suitable administration routes include, but are not limited to, oral, rectal, transmucosal, intestinal, enteral, topical, suppository, through inhalation, intrathecal, intracerebroventricular, intraperitoneal, intranasal, intraocular, intratumoral, and parenteral (e.g., intravenous, intramuscular, intramedullary, and subcutaneous). In certain embodiments, pharmaceutical intrathecals are administered to achieve local rather than systemic exposures. For example, pharmaceutical compositions may be injected directly in the area of desired effect (e.g., into the liver).
Nonlimiting disclosure and incorporation by reference While certain compounds, compositions and methods described herein have been described with specificity in accordance with certain embodiments, the following examples serve only to illustrate the compounds described herein and are not intended to limit the same. Each of the references, GenBank accession numbers, and the like recited in the present application is incorporated herein by reference in its entirety.
Although the sequence listing accompanying this filing identifies each sequence as either "RNA" or "DNA" as required, in reality, those sequences may be modified with any combination of chemical modifications. One of skill in the art will readily appreciate that such designation as "RNA" or "DNA" to describe modified oligonucleotides is, in certain instances, arbitrary. For example, an oligonucleotide comprising a nucleoside comprising a 2'-OH sugar moiety and a thymine base could be described as a DNA
having a modified sugar (2'-OH for the natural 2'-H of DNA) or as an RNA
having a modified base (thymine (methylated uracil) for natural uracil of RNA).
Accordingly, nucleic acid sequences provided herein, including, but not limited to those in the sequence listing, are intended to encompass nucleic acids containing any combination of natural or modified RNA and/or DNA, including, but not limited to such nucleic acids having modified nucleobases. By way of further example and without limitation, an oligonucleotide having the nucleobase sequence "ATCGATCG"
encompasses any oligonucleotides having such nucleobase sequence, whether modified or unmodified, including, but not limited to, such compounds comprising RNA bases, such as those having sequence "AUCGAUCG" and those having some DNA bases and some RNA bases such as "AUCGATCG" and oligonucleotides having other modified bases, such as "AT'CGAUCG," wherein IneC indicates a cytosine base comprising a methyl group at the 5-position.
EXAMPLES
The following examples illustrate certain embodiments of the present disclosure and are not limiting.
Moreover, where specific embodiments are provided, the inventors have contemplated generic application of those specific embodiments. For example, disclosure of an oligonucleotide having a particular motif provides reasonable support for additional oligonucleotides having the same or similar motif And, for example, where a particular high-affinity modification appears at a particular position, other high-affinity modifications at the same position are considered suitable, unless otherwise indicated.
Example 1: General Method for the Preparation of Phosphoramidites, Compounds 1, la and 2 Bx Bx DMT0( r DMTO _______________________ DMT0/41*--c OMe H3C"¨c NCO A"- N(iPr)2 NC012),N(iPr)2 NC013,N(iPr)2 1 la 2 Bx is a heterocyclic base;
Compounds 1, la and 2 were prepared as per the procedures well known in the art as described in the specification herein (see Seth et al., Bioorg. Med. Chem., 2011, 21(4), 1122-1125, J. Org. Chem., 2010, 75(5), 1569-1581, Nucleic Acids Symposium Series, 2008, 52(1), 553-554); and also see published PCT

International Applications (WO 2011/115818, WO 2010/077578, W02010/036698, W02009/143369, WO
2009/006478, and WO 2007/090071), and US patent 7,569,686).
Example 2: Preparation of Compound 7 Ac0 OAc Ac0 OAc 40 m 0 Ac0 OAc _______________________________________________ HO-'O oil& 5 Ac0 ___....Z.\., TMSOTf, 50 C
11"- )..-CICH2CH2CI N------____----.. TMSOTf, DCE
AcHN
3 (93%) ( 66%) Ac0 OAc Ac0 OAc ___....2..\., H2/Pd 0 _________________________________________ 0 __ Ac0 Ac0 0,...,............---,,,, Me0H
AcHN 0 AcHN 0 (95%) Compounds 3 (2-acetamido-1,3,4,6-tetra-O-acety1-2-deoxy-13-Dga1actopyranose or galactosamine pentaacetate) is commercially available. Compound 5 was prepared according to published procedures (Weber et al., J. Med. Chem., 1991, 34, 2692).
Example 3: Preparation of Compound 11 NCr-----1 Z CL
HO-..õ Et00 .....õ--_,CN 9 HCI, Et0H
HON H2 ).- NC---N----(:).------ ---- __ NH2 0.-aq KOH, Reflux, rt' 0 Et0 0"
HO" 1,4-dioxane, 0' (56%) (40%) NC-..,I 10 d"---) 11 õ
Compounds 8 and 9 are commercially available.

Example 4: Preparation of Compound 18 ),.....,Et01.no 0......õ_o__ NI 0 io Et0 0 0_, benzylchloroformate, Et0 Et0 Li0H, H20 Dioxane, Na2CO3 )------0,õ....---NH2 _______________ a H Dioxane p.
O Et0 0-- (86%) 0 Et0 0--(91%) >ro,___114NH
HO ' õõ..1O
0 9, 9,-0------1\1NH2 14 _)-,1r --J
H N..-N.----Ny=-=,0õ,..õ_N 0 HOµii-----N--0õ,.......--N--,0 =101 HBTU, DIEA, DMF
0 HO 0-- (69%) 0 --)ThLNN---e (D-------) 13 H H -r, AcO0Ac H

0 wiroH

H 0 0, ?, AcHN 0 ----r..,-0.-----N---N0 io HBTU, DIEA, HOBt ______________ ).- H
r 95% 0 0' DMF
16 (64%) V\N_____kj AcO0Ac _....r.Ø.\r H H
r,1\1.õ..N.r,01 Ac0 O
AcHN 0 AcO0Ac 0 _....r.Ø.\r H H 0, Ac0 0 õõ.....-õir-Ns.õ.õ......N,r.N._0-_,-- N--1(0 io H
AcHN 0 0 0' AcO0Ac HN-----kj H___z___/
Ac0 0 AcHN 18 Compound 11 was prepared as per the procedures illustrated in Example 3.
Compound 14 is commercially available. Compound 17 was prepared using similar procedures reported by Rensen et al., J.
5 Med. Chem., 2004, 47, 5798-5808.

Example 5: Preparation of Compound 23 1. TBDMSCI 1. H
N) H3C0)tkOH 21 HBTU, DIEA
/ -b TBDMS0 N DMF, Imidazode, rt (95 %) DMF, rt (65%) HO----. ) _______________________ "...
2. Pd/C, H2, Me0H, rt - 2.
TEA.3HF, TEA, THF
87% 20 0- TBDMS (72%) -OH

HO).H 0 0 1. DMTCI, pyr, rt (75%) OH
OCH ______________ -b1jgt' 3 )m-2. Li0H, Dioxane (97%) 23 z 22 6H
OH
Compounds 19 and 21 are commercially available.
Example 6: Preparation of Compound 24 AcO0Ac H H
Ac0_...\,' 0 Wy AcHN 0 1.
H2, Pd/C, Me0H (93%) AcO0Ac ' 2. HBTU, DIEA, DMF (76%) H z-AcHN 0 0 (:) HO'LRIN =
c 23 AcO0Ac H HN----4-1 Ac00-----r N----/----/

AcHN
AcO0Ac H H
Ac0(2..\,ONN,0 AcHN 0 AcO0Ac ODMT
0 0 µ

H µ
AcHN 0 0 (:) OH
AcO0Ac H HN-----kj Ac0o----------( N---/-----/

Ar.HNI

Compounds 18 and 23 were prepared as per the procedures illustrated in Examples 4 and 5.
Example 7: Preparation of Compound 25 AcO0Ac H H
Ac0.72..\r0r,N.,..N,0 AcHN 0 AcO0Ac ODMT
0 0 ,/_ 1. Succinic anhydride, DMAP, DCE
AcHN 0 0 0' OH
2. DMF, HBTU, EtN(iPr)2, PS-SS
AcO0Ac H HN-----kj __õ,..2..\.r0---- N
N--ir Ac0 0 AcHN
AcO0Ac H H
Ac0 Or N N,.(:) AcHN 0 AcO0Ac ODMT
e _..r.2.._\.v H H 0, 0 0 Ac0 OrNIN 0 ___________________________________ NH
AcHN

,¨NH
H
AcHN 0 0 0' 04 AcO0Ac H N -----kj H___/....__/
Ac0 0----- 0 AcHN
5 Compound 24 was prepared as per the procedures illustrated in Example 6.

Example 8: Preparation of Compound 26 AcO0Ac H H
AcO___Z,0,..N.õN,D
ODMT
AcHN 0 AcO0Ac H H 0-, 0 0 OH
Phosphitylation H 8 \
AcHN 0 0 0' HN------k-j AcO0Ac H 0 N---.7.--/
_....7.2..\,C)-----/\/""---r Ac0 0 AcHN
AcO0Ac __......r2..\, H H
0 N ,,õ-N,.0 Ac0 AcHN 0 AcO0Ac ODMT
H H 0, 0it µ? _ N-1--18-'N--H µ
AcHN 0 0 0' 0 I
NC.,...,..--Ø-P-N up 0 2 AcO0Ac H HN------kj Ac0 _._...!...Ds\ro._._....._.----rN----7------/

Compound 24 is prepared as per the procedures illustrated in Example 6.

Example 9: General preparation of conjugated ASOs comprising Ga1NAc3-1 at the 3' terminus, Compound 29 AcO0Ac H H
Ac0 0---72-\r .rN ,--N -c) AcHN 0 ODMT
AcO0Ac e ./, Ac0----\, C)NNVNNyN,0 eLH8LNIµ )\¨NH
AcHN 0 0 0 04 HN 1. DCA, DCM
¨C1 AcO0Ac H 0 2. DCI, NMI, ACN
_,..(..:.....\,) ....Ø______,....,....õ----ir--N---.7"---/
Phosphoramidite DNA/RNA
Ac0 building block 1 a.utomated synthesizer AcHN 3. Capping 4. t-BuO0H DMT0,\o a,Bx _ AcO0Ac _.....r..c.1\ror NH ,,,, NH ,0 0 Ac0 0=P-0 AcHN 0 O
AcO0Ac Ac0-1=*--\, NNV\I\Iy,0- hl )L NI\
)\¨NH
AcHN 0 0 0' 04 _____.J 1. DCA, DCM 0 2. DCI, NMI, ACN
HN
AcO0Ac H 0 Phosphoramidite DNA/RNA ' Ac0 _..r.C.......,0___........õ-----..w--N building block la a.utomated synthesizer , 3. Capping 0 27 4. t-BuO0H
AcHN
, D M TO -N(0),Bx (5== b_/-0Me 0.p1_0CN
\
0-N(043x AcO0Ac ,\ __ /
_......2.\r H H
N -,N ,C) 0 I
Ac0 0 O=-0-AcHN 0 O
AcO0Ac H H 0, 0 0 ./, _\_&z,0N 0 LN NH
,Ny.,.._............, N.---kq, 0 Ac0 H µ
AcHN 0 0 0' 04 N 1. DCA, DCM
H¨C1 AcO0Ac H 0 2. DCI, NMI, ACN
Ac0 _....72..s\r0-----/\/---ir N--/-----/ Phosphoramidite DNA/RNA
building blocks a.utomated synthesize AcHN 3. Capping 4. xanthane hydride or t-BuO0H
5. Et3N/CH3CN (1:1) 6. Aaueous NH, (cleavaue) OH
I
, OLIGO , X=P\-0-0¨NO,Bx Bx = Heterocyclic base 6- b_/-0Me X=OorS l O=P-0-\
0¨Nro,ipx HOOH .\ __ i H H a HO0r-N,,,N,D 1 0=P-0 HOOH -AcHN 0 O
H H 0, 0 0 ./, ,,.....õ,õ.õ.õ ...,7--.........õN 0õ.-- NACtil'NrµZ
H \
AcHN 0 0 0' OH
HOOH HN-----kj HO0----__ri-N-1---/----/

AcHN
Wherein the protected Ga1NAc3-1 has the structure:

1¨F1)-0¨NoN

HOOH

1,.. N N
õ,0 1 HO 0 0=P-0-AcHN 0 1 HOOH
H H0, 0 0 õ,..,õ¨,,.,õõõ,e,..- N.7-,..,,,N 0......_, N"111-e'N--sZ
H \
AcHN 0 0 0' OH
HOOH HN-----kj H00--_--rN

AcHN
The Ga1NAc3 cluster portion of the conjugate group GalNAc3-1 (Ga1NAc3-1a) can be combined with any cleavable moiety to provide a variety of conjugate groups. Wherein Ga1NAc3-1a has the formula:

HOOH
H H
HO__,c,.......õ.õ....õ..¨.1(N,..õ---...õ-N.,...,0 AcHN 0 I

HO .õ,_,.......õ.-..,õ-- N.,...--........_,N 0õ-- NARIIVNZ
H µ
AcHN 0 0 0 OH
HOOHHN-----k-j H 0 ./......./
HO__....,r2....\,0-W---rN

AcHN
The solid support bound protected Ga1NAc3-1, Compound 25, was prepared as per the procedures illustrated in Example 7. Oligomeric Compound 29 comprising Ga1NAc3-1 at the 3' terminus was prepared using standard procedures in automated DNA/RNA synthesis (see Dupouy et al., Angew. Chem. Int. Ed., 2006, 45, 3623-3627). Phosphoramidite building blocks, Compounds 1 and la were prepared as per the procedures illustrated in Example 1. The phosphoramidites illustrated are meant to be representative and not intended to be limiting as other phosphoramidite building blocks can be used to prepare oligomeric compounds having a predetermined sequence and composition. The order and quantity of phosphoramidites added to the solid support can be adjusted to prepare gapped oligomeric compounds as described herein.
Such gapped oligomeric compounds can have predetermined composition and base sequence as dictated by any given target.

Example 10: General preparation conjugated ASOs comprising GaINAc3-1 at the 5' terminus, Compound 34 ODMT 1. Capping (Ac20, NMI, PYr) I
1. DCA, DCM (OLIGO) 2. PADS or t-BuO0H
_____________________________ . 3. DCA, DCM I
(a-UNL-ODMT 2. DCI, NMI, ACN 0 ..
I 0, 4. DCI, NMI, ACN
30 Phosphoramidite 1N Phosphoramidite 1 building blocks , DNA/RNA DNA/RNA
31 ,automated synthesizer, ,.automated synthesizer , DMT0(5"Bx 1. Capping (Ac20, NMI, PYr) 2. t-BuO0H 0 3. DCA, DCM NC 0_1;
... ____________________ 4. DCI, NMI, ACN 0 I
Phosphoramidite 26 (.OLIGO) ' DNA/RNA ' I
X = 0, or S automated synthesizer 0 , I
Bx = Heterocylic base 0-UNL-01-AcO0Ac ___...?
Ac0 ...\z H H
AcHN 0 Ac0 OAc 0 D MT
H H 0, 0 0 __T.C.)...\,0 Ac0 NN (:),....,-- _____ NARILN
H \
AcHN 0 0 0' 0 I
NC,0,1=',00y,Bx H N-----Cj AcO0Ac H

___......2 0 .\.z0 õ,-- N----7------/
NCO-P=0 Ac0 0 0 AcHN I
(OLIGO) I
1. Capping (Ac20, NMI, PYr) 0 2. t-BuO0H I
3. Et3N:CH3CN (1:1 v/v) 0-UNL-0-P-0CN
X
4. DCA, DCM
5. NH4, rt (cleavage) 33 HOOH
H H

HO--41, r'N"N'.
AcHN 0 HOOH OH
H H 0, 0 0 ..
HO_...7!.1,o-õ,.....--.,....¨...õ---N.N..õ--",..,õ...-N 0.,.....- N-11.1-38-11'N
H \
AcHN 0 0 0' CI) -0-/--R_u,,.%,c Or Bx 1-1_.y....... j 0 0.
HO_.....r2.\,0-_____---ir N
-04=0 o 6 AcHN 34 I
(OLIGO) I
OT T
The UnylinkerTM 30 is commercially available. Oligomeric Compound 34 comprising a Ga1NAc3-1 cluster at the 5' terminus is prepared using standard procedures in automated DNA/RNA synthesis (see Dupouy et al., Angew. Chem. Int. Ed., 2006, 45, 3623-3627). Phosphoramidite building blocks, Compounds 1 and la were prepared as per the procedures illustrated in Example 1. The phosphoramidites illustrated are meant to be representative and not intended to be limiting as other phosphoramidite building blocks can be used to prepare an oligomeric compound having a predetermined sequence and composition. The order and quantity of phosphoramidites added to the solid support can be adjusted to prepare gapped oligomeric compounds as described herein. Such gapped oligomeric compounds can have predetermined composition and base sequence as dictated by any given target.
Example 11: Preparation of Compound 39 AcO0Ac )-1. HO N 0 AcO0Ac Ac0___\ .
35 TMSOTf, DCE Ac0 C), NH2 ______________________________________________ ).- 8 N--__H----- 2. H2/Pd, Me0H AcHN 36 Ac0 OAc Ac0 1. H2, Pd/C, Me0H
HBTU, DMF, EtN(iP02 ____________________ 0.- o\----NO__Nõ,k1 ________________________ 0 __ Compound 13 AcHN 8 2. HBTU, DIEA, DMF
OAc Ac0 H 0 0 Compound 23 Ac0 8 0 NHAc 0 0 0 OAc ,----) Ac0 0 Ac0.....zyoNH
411t AcHN

Ac0 OAc \
Ac00 /0DMT ---=4...N__FNi = Phosphitylation _ AcHN 8 0 OAc 0\ ¨Na H 0 .,.0 8 AcO
OHm.I.,/0.........Nyõ.0 NH
Ac0 8 NHAc 0 0 0 OAc )\--) Ac0 380...izz NH
Ac0 C)K7 AcHN
Ac0 OAc Ac0.7.2.\õ, /ODMT

__FNi AcHN 8 0 -OAc F 0 0 0\ N
Ac0 Ni .,,.} > 8 ACOTZ\Z 0 NH 1 P
NHAc 0 0 0 NC (D' N UP 02 OAc AcOoric?....v Ac0 C)e AcHN
Compounds 4, 13 and 23 were prepared as per the procedures illustrated in Examples 2, 4, and 5.
Compound 35 is prepared using similar procedures published in Rouchaud et al., Eur. J. Org. Chem., 2011, 12, 2346-2353.

Example 12: Preparation of Compound 40 Ac0 OAc Ac00 /0DMT
FNi AcHN 8 OAc ON
)¨Na Ac0 H 0 0 > __ 8 OH
Ac0 ....2...\,/07,,i,,),.rNy.,- ONH

NHAc 0 0 0 1. Succinic anhydride, DMAP, DCE
OAc Ac0....Trzr NH 2. DMF, HBTU, EtN(iPr)2, PS-SS
Ac0 C)-,1; 38 AcHN
Ac0 OAc Ac0 0 ..... ODMT
/
__FNI

AcHN 8 0 )¨N 0 0 Ac0 OAc H 0 0 8 ---{"j"--N y.,..-0 NH 0 Ac0:) =Z 8 NHAc 0 0 0 OAc AcOrio....\, )\---) 40 Ac0 NH
,8 AcHN
Compound 38 is prepared as per the procedures illustrated in Example 11.
Example 13: Preparation of Compound 44 AcO0Ac HBTU, DMF, EtN(iP02 Ac0 AcHN 36 H01.0õ )\-0 *
¨N

HO 41\ /-0".

Ac0 OAc Ac0,0 AcHN X-rX)N1 O 0-....kil 1. H2, Pd/C, Me0H
0 ______________________________________________________________ 0-0 2. HBTU, DIEA, DMF
0)\__) Compound 23 OAc 0 Ac0.7Ø...\/
fk Ac0 AcHN
Ac0 OAc ¨ N ODMT
Ac0 u -.7.4":)=-\--'x,R.......x___H
-, AcHN 8 0 0 - Phosphitylation x-}N)8 NaOH
H

0\\ ) 43 OAc AcO
Ac0rao.s\zo y-----NH

AcHN
Ac0 OAc Ac0 ¨u N ODMT
=
AcHN
0 0 , 01-Na OAc I N 8 ?
H NC0,PN(iPr)2 0).\_) AcO4, 44 ONH
Ac0 AcHN
Compounds 23 and 36 are prepared as per the procedures illustrated in Examples 5 and 11.
Compound 41 is prepared using similar procedures published in WO 2009082607.

Example 14: Preparation of Compound 45 Ac0 OAc Ac0 - N ODMT
AcHN
IN _ OH
H

0\\ ) 43 OAc Ac0 Ac07Ø...\,0 y-----/
NH 1. Succinic anhydride, DMAP, DCE

_______________________________________________________________________________ 0.
AcHN 2. DMF, HBTU, EtN(iPr)2, PS-SS
Ac0 OAc Ac00N__(...)(____N__N ODMT
H
AcHN 8 0 -IN)\

0\\ i OAc )1---- 45 Ac0 Ac0 ...,7,2..\70NH
'8 AcHN
Compound 43 is prepared as per the procedures illustrated in Example 13.
Example 15: Preparation of Compound 47 H0)10,-0 * DMTO
1. DMTCI, pyr sb1H
_____________________________________ ..
d= 46 2. Pd/C, H2, Me0H :
H
H d 47 Compound 46 is commercially available.

Example 16: Preparation of Compound 53 HBTU, EtNUP02, DMF 0 = H3C0--N
v Boc HN''Boc NH
\CBz 50 CBz NH
HN,CBz 0)----.

H3COyJ NCBz 1. TFA N NH H 1. Li0H, Me0H
_________________ 0- 0 H __________________________________________ .
2. HBTU, EtNUP02, DMF 2. HBTU, EtNUP02, DMF

HNrCBz Compound 47 HN,CBz 0 --\--/¨NicICBz OH

DMTO HN-CBz 1. H2, Pd/C
0.,z,,, 2. HBTU, EtN(iP02 , DMF
HO'' eN--0--r"N- C ,CBz NH N Compound 17 "H
0 ' H

HN-CBz OAcOAc Ac0 _____\....___\_ 0 0 ,L_____ NHAc OAc 0 OAc..____\.......\_ 0 II),,N7-------...,00H
Ac0 0 ,)..

HN--...1}...,(L.
.-------NHAc OAc 0 OAc' O

Ac0------"\-----\-0 NHAc Compounds 48 and 49 are commercially available. Compounds 17 and 47 are prepared as per the procedures illustrated in Examples 4 and 15.

Example 17: Preparation of Compound 54 OAc OAc......\,.....\._ 0 Ac0 0 ,.[_____ NHAc OAc 0 OAc.....v...\._ 0 ,.....it,),.., r-----.=,10H
N
Ac0 NHAc 0 r OAc 0 ODMT
OAT( 0 0 v)1 NH 53 Ac0----\------\--0 NHAc Phosphitylation y OAc OAc....7.....\_ 0 Ac0 0 ,.[____ NHAc \
(iPr)2N, p ¨0 OAc ----- 0 , \----\
OAc_...\,..___\_ CN
0 1_,.....,./i, 7------_,,%0 Ac0 0 0 HN HN------N/ 7 1\1_______ NHAc 0 l OAcOAc 0 v)1 Ac0 0 NHAc Compound 53 is prepared as per the procedures illustrated in Example 16.

Example 18: Preparation of Compound 55 OAc Ac0 0 NHAc OAc 0 Ac0 0 NHAc 0 OAc ODMT
I _____________________________________________ NH 53 NHAc 1. Succinic anhydride, DMAP, DCE
2. DMF, HBTU, EtN(iPr)2, PS-SS
OAc Ac0 0 NHAc OAc Ac0 0 NHAc 0 OAc ODMT
I _____________________________________ NH 55 Ac0 0 NHAc Compound 53 is prepared as per the procedures illustrated in Example 16.
Example 19: General method for the preparation of conjugated ASOs comprising GaINAc3-1 at the 3' position via solid phase techniques (preparation of ISIS 647535, 647536 and 651900) Unless otherwise stated, all reagents and solutions used for the synthesis of oligomeric compounds are purchased from commercial sources. Standard phosphoramidite building blocks and solid support are used for incorporation nucleoside residues which include for example T, A, G, and InC residues. A 0.1 M
solution of phosphoramidite in anhydrous acetonitrile was used for 13-D-2'-deoxyribonucleoside and 2'-MOE.
The ASO syntheses were performed on ABI 394 synthesizer (1-2 [mot scale) or on GE Healthcare Bioscience AKTA oligopilot synthesizer (40-200 [mot scale) by the phosphoramidite coupling method on an Ga1NAc3-1 loaded VIMAD solid support (110 i.tmol/g, Guzaev et al., 2003) packed in the column. For the coupling step, the phosphoramidites were delivered 4 fold excess over the loading on the solid support and phosphoramidite condensation was carried out for 10 min. All other steps followed standard protocols supplied by the manufacturer. A solution of 6% dichloroacetic acid in toluene was used for removing dimethoxytrityl (DMT) group from 5'-hydroxyl group of the nucleotide. 4,5-Dicyanoimidazole (0.7 M) in anhydrous CH3CN was used as activator during coupling step. Phosphorothioate linkages were introduced by sulfurization with 0.1 M solution of xanthane hydride in 1:1 pyridine/CH3CN
for a contact time of 3 minutes.
A solution of 20% tert-butylhydroperoxide in CH3CN containing 6% water was used as an oxidizing agent to provide phosphodiester internucleoside linkages with a contact time of 12 minutes.
After the desired sequence was assembled, the cyanoethyl phosphate protecting groups were deprotected using a 1:1 (v/v) mixture of triethylamine and acetonitrile with a contact time of 45 minutes. The solid-support bound ASOs were suspended in aqueous ammonia (28-30 wt %) and heated at 55 C for 6 h.
The unbound ASOs were then filtered and the ammonia was boiled off The residue was purified by high pressure liquid chromatography on a strong anion exchange column (GE
Healthcare Bioscience, Source 30Q, 30 [tin, 2.54 x 8 cm, A = 100 mM ammonium acetate in 30% aqueous CH3CN, B
= 1.5 M NaBr in A, 0-40% of B in 60 min, flow 14 mL min-1, k = 260 nm). The residue was desalted by HPLC on a reverse phase column to yield the desired ASOs in an isolated yield of 15-30% based on the initial loading on the solid support. The ASOs were characterized by ion-pair-HPLC coupled MS analysis with Agilent 1100 MSD
system.
Antisense oligonucleotides not comprising a conjugate were synthesized using standard oligonucleotide synthesis procedures well known in the art.
Using these methods, three separate antisense compounds targeting ApoC III
were prepared. As summarized in Table 17, below, each of the three antisense compounds targeting ApoC III had the same nucleobase sequence; ISIS 304801 is a 5-10-5 MOE gapmer having all phosphorothioate linkages; ISIS
647535 is the same as ISIS 304801, except that it had a Ga1NAc3-1 conjugated at its 3'end; and ISIS 647536 is the same as ISIS 647535 except that certain internucleoside linkages of that compound are phosphodiester linkages. As further summarized in Table 17, two separate antisense compounds targeting SRB-1 were synthesized. ISIS 440762 was a 2-10-2 cEt gapmer with all phosphorothioate internucleoside linkages; ISIS
651900 is the same as ISIS 440762, except that it included a Ga1NAc3-1 at its 3'-end.
Table 17 Modified ASO targeting ApoC III and SRB-1 SEQ
CalCd Observed ASO Sequence (5 to 3') Target ID
Mass Mass No.
ISIS

1 ss ApoC
AesGesniCesTesTesmCd Td Td Gd Td mCd mCd Ad Gd mCd TesTesTesAesTe 7 1 65 .4 7164.4 244 ISIS AesGesnaCesTesTesmCdsTdsTdsGds TdsmCdsmCdsAdsGdsmCds Tes Tes TesAes TeoAdo, - ApoC
9239.5 9237.8 245 647535 Ga1NAc3-1. III
ISIS AesGeonaCee Tee TeemCds TdsTdsGds TdsmCdsmCdsAdsGdsmCds Teo Teo TesAes TeoAdo, - ApoC
9142.9 9140.8 245 647536 Ga1NAc3-1. III

ISISSRB-TksmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTksmCk 4647.0 4646.4 246 ISISSRB-TksmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTksmCkoAdo,-GaINAC3-1a 6721.1 6719.4 247 Subscripts: "e" indicates 2'-MOE modified nucleoside; "d" indicates [3-D-2'-deoxyribonuc1eoside; "k"
indicates 6'-(S)-CH3 bicyclic nucleoside (e.g. cEt); "s" indicates phosphorothioate internucleoside linkages (PS); "o" indicates phosphodiester internucleoside linkages (PO); and "o¨
indicates -0-P(=0)(OH)-.
Superscript "m" indicates 5-methylcytosines. "Ga1NAc3-1" indicates a conjugate group having the structure shown previously in Example 9. Note that Ga1NAc3-1 comprises a cleavable adenosine which links the ASO
to remainder of the conjugate, which is designated "Ga1NAc3-1.." This nomenclature is used in the above table to show the full nucleobase sequence, including the adenosine, which is part of the conjugate. Thus, in the above table, the sequences could also be listed as ending with "Ga1NAc3-1"
with the "Ado" omitted. This convention of using the subscript "a" to indicate the portion of a conjugate group lacking a cleavable nucleoside or cleavable moiety is used throughout these Examples. This portion of a conjugate group lacking the cleavable moiety is referred to herein as a "cluster" or "conjugate cluster" or "Ga1NAc3 cluster." In certain instances it is convenient to describe a conjugate group by separately providing its cluster and its cleavable moiety.
Example 20: Dose-dependent antisense inhibition of human ApoC III in huApoC
III transgenic mice ISIS 304801 and ISIS 647535, each targeting human ApoC III and described above, were separately tested and evaluated in a dose-dependent study for their ability to inhibit human ApoC III in human ApoC III
transgenic mice.
Treatment Human ApoCIII transgenic mice were maintained on a 12-hour light/dark cycle and fed ad libitum Teklad lab chow. Animals were acclimated for at least 7 days in the research facility before initiation of the experiment. ASOs were prepared in PBS and sterilized by filtering through a 0.2 micron filter. ASOs were dissolved in 0.9% PBS for injection.
Human ApoC III transgenic mice were injected intraperitoneally once a week for two weeks with ISIS 304801 or 647535 at 0.08, 0.25. 0.75, 2.25 or 6.75 [tmol/kg or with PBS
as a control. Each treatment group consisted of 4 animals. Forty-eight hours after the administration of the last dose, blood was drawn from each mouse and the mice were sacrificed and tissues were collected.
ApoC III mRNA Analysis ApoC III mRNA levels in the mice's livers were determined using real-time PCR
and RIBOGREENO RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. ApoC III mRNA levels were determined relative to total RNA (using Ribogreen), prior to normalization to PBS-treated control. The results below are presented as the average percent of ApoC III

mRNA levels for each treatment group, normalized to PBS-treated control and are denoted as "% PBS". The half maximal effective dosage (ED50) of each ASO is also presented in Table 18, below.
As illustrated, both antisense compounds reduced ApoC III RNA relative to the PBS control.
Further, the antisense compound conjugated to Ga1NAc3-1 (ISIS 647535) was substantially more potent than the antisense compound lacking the Ga1NAc3-1 conjugate (ISIS 304801).
Table 18 Effect of ASO treatment on ApoC III mRNA levels in human ApoC III transgenic mice Dose % ED50Internucleoside SEQ ID
ASO 3' Conjugate Olmol/kg) PBS Onnol/kg) linkage/Length No.

0.08 95 ISIS 0.75 42 0.77 None PS/20 244 304801 2.25 32 6.75 19 0.08 50 SIS 0.75 15 I
647535 2.25 17 0.074 Ga1NAc3-1 PS/20 245 6.75 8 ApoC III Protein Analysis (Turbidometric Assay) Plasma ApoC III protein analysis was determined using procedures reported by Graham et al, Circulation Research, published online before print March 29, 2013.
Approximately 100 1 of plasma isolated from mice was analyzed without dilution using an Olympus Clinical Analyzer and a commercially available turbidometric ApoC III assay (Kamiya, Cat# KAI-006, Kamiya Biomedical, Seattle, WA). The assay protocol was performed as described by the vendor.
As shown in the Table 19 below, both antisense compounds reduced ApoC III
protein relative to the PBS control. Further, the antisense compound conjugated to Ga1NAc3-1 (ISIS
647535) was substantially more potent than the antisense compound lacking the Ga1NAc3-1 conjugate (ISIS
304801).
Table 19 Effect of ASO treatment on ApoC III plasma protein levels in human ApoC III
transgenic mice Dose ED50Internucleoside SEQ ID
ASO 3' Conjugate (jnnol/kg) PBS (jnnol/kg) Linkage/Length No.

ISIS 0.08 86 0.73 None PS/20 244 304801 0.75 51 2.25 23 6.75 13 0.08 72 ISIS 0.75 14 647535 2.25 12 0.19 Ga1NAc3-1 PS/20 245 6.75 11 Plasma triglycerides and cholesterol were extracted by the method of Bligh and Dyer (Bligh, E.G.
and Dyer, W.J. Can. J. Biochem. Physiol. 37: 911-917, 1959)(Bligh, E and Dyer, W, Can J Biochem Physiol, 37, 911-917, 1959)(Bligh, E and Dyer, W, Can J Biochem Physiol, 37, 911-917, 1959) and measured by using a Beckmann Coulter clinical analyzer and commercially available reagents.
The triglyceride levels were measured relative to PBS injected mice and are denoted as "%
PBS". Results are presented in Table 20. As illustrated, both antisense compounds lowered triglyceride levels. Further, the antisense compound conjugated to Ga1NAc3-1 (ISIS 647535) was substantially more potent than the antisense compound lacking the Ga1NAc3-1 conjugate (ISIS
304801).
Table 20 Effect of ASO treatment on triglyceride levels in transgenic mice ASO Dose % ED50 3' Internucleoside SEQ ID
(jlmol/kg) PBS (jlmol/kg) Conjugate Linkage/Length No.

0.08 87 ISIS 0.75 46 0.63 None PS/20 244 304801 2.25 21 6.75 12 0.08 65 ISIS 0.75 9 0.13 Ga1NAc3-1 PS/20 245 647535 2.25 8 6.75 9 Plasma samples were analyzed by HPLC to determine the amount of total cholesterol and of different fractions of cholesterol (HDL and LDL). Results are presented in Tables 21 and 22. As illustrated, both antisense compounds lowered total cholesterol levels; both lowered LDL; and both raised HDL. Further, the antisense compound conjugated to Ga1NAc3-1 (ISIS 647535) was substantially more potent than the antisense compound lacking the Ga1NAc3-1 conjugate (ISIS 304801). An increase in HDL and a decrease in LDL levels is a cardiovascular beneficial effect of antisense inhibition of ApoC III.

Table 21 Effect of ASO treatment on total cholesterol levels in transgenic mice ASO Dose Total Cholesterol 3' Internucleoside SEQ
(jlmol/kg) (mg/dL) Conjugate Linkage/Length ID
No.

0.08 226 ISIS 0.75 164 None PS/20 244 304801 2.25 110 6.75 82 0.08 230 ISIS 0.75 82 647535 2.25 86 Ga1NAc3-1 PS/20 245 6.75 99 Table 22 Effect of ASO treatment on HDL and LDL cholesterol levels in transgenic mice ASO Dose HDL LDL 3' Internucleoside SEQ
(iamol/kg) (mg/dL) (mg/dL) Conjugate Linkage/Length ID
No.

0.08 17 23 ISIS 0.75 27 12 None PS/20 244 304801 2.25 50 4 6.75 45 2 0.08 21 21 ISIS 0.75 44 2 Ga1NAc3-1 PS/20 245 647535 2.25 50 2 6.75 58 2 Pharmacokinetics Analysis (PK) The PK of the ASOs was also evaluated. Liver and kidney samples were minced and extracted using standard protocols. Samples were analyzed on MSD1 utilizing IP-HPLC-MS. The tissue level ( g/g) of full-length ISIS 304801 and 647535 was measured and the results are provided in Table 23. As illustrated, liver concentrations of total full-length antisense compounds were similar for the two antisense compounds.
Thus, even though the Ga1NAc3-1 -conjugated antisense compound is more active in the liver (as demonstrated by the RNA and protein data above), it is not present at substantially higher concentration in the liver. Indeed, the calculated EC50 (provided in Table 23) confirms that the observed increase in potency of the conjugated compound cannot be entirely attributed to increased accumulation. This result suggests that the conjugate improved potency by a mechanism other than liver accumulation alone, possibly by improving the productive uptake of the antisense compound into cells.
The results also show that the concentration of Ga1NAc3-1 conjugated antisense compound in the kidney is lower than that of antisense compound lacking the GalNAc conjugate.
This has several beneficial therapeutic implications. For therapeutic indications where activity in the kidney is not sought, exposure to kidney risks kidney toxicity without corresponding benefit. Moreover, high concentration in kidney typically results in loss of compound to the urine resulting in faster clearance.
Accordingly, for non-kidney targets, kidney accumulation is undesired. These data suggest that Ga1NAc3-1 conjugation reduces kidney accumulation.
Table 23 PK analysis of ASO treatment in transgenic mice Internucleoside Dose Liver Kidney Liver EC50 3'SEQ
ASO Linkage/Length (jnnol/kg) (m/g) (pg/g) (pg/g) Conjugate ID
No.
0.1 5.2 2.1 ISIS 0.8 62.8 119.6 53 None PS/20 304801 2.3 142.3 191.5 6.8 202.3 337.7 0.1 3.8 0.7 ISIS 0.8 72.7 34.3 3.8 Ga1NAc3-1 PS/20 647535 2.3 106.8 111.4 6.8 237.2 179.3 Metabolites of ISIS 647535 were also identified and their masses were confirmed by high resolution mass spectrometry analysis. The cleavage sites and structures of the observed metabolites are shown below.
The relative % of full length ASO was calculated using standard procedures and the results are presented in Table 23a. The major metabolite of ISIS 647535 was full-length ASO lacking the entire conjugate (i.e. ISIS
304801), which results from cleavage at cleavage site A, shown below. Further, additional metabolites resulting from other cleavage sites were also observed. These results suggest that introducing other cleabable bonds such as esters, peptides, disulfides, phosphoramidates or acyl-hydrazones between the Ga1NAc3-1 sugar and the ASO, which can be cleaved by enzymes inside the cell, or which may cleave in the reductive environment of the cytosol, or which are labile to the acidic pH inside endosomes and lyzosomes, can also be useful.
Table 23a Observed full length metabolites of ISIS 647535 Metabolite ASO
Cleavage site Relative %
1 ISIS 304801 A 36.1 2 ISIS 304801 + dA B 10.5 3 ISIS 647535 minus [3 GalNAc] C 16.1 ISIS 647535 minus 4 D 17.6 [3 GalNAc + 1 5-hydroxy-pentanoic acid tether]
ISIS 647535 minus D 9.9 [2 GalNAc + 2 5-hydroxy-pentanoic acid tether]
ISIS 647535 minus D
69.8 [3 GalNAc + 3 5-hydroxy-pentanoic acid tether]

Cleavage Sites () Cleavage site A
I
HO OH Cleavage site C 0=P-OH

Cleavage site D O

Nx1...õ,..
HO \-0 \_HN N
-----(_ OH ONN i HO OH NHAc 0 0 0 d 0 Cleavage site C
H H
¨ Cleavage site B
I
HO \C3 \ N N 0---hl 0 __ P=0 NHAc Cleavage site D 0 0 OH

OH
HO HN

\ 0 \ N
Cleavage site D
"Ac Cleavage site C 0 0=P-OH NH2 ASO 304801 (ID
Metabolite 1 Metabolite 2 N_I-4,,,.N
,C),,N N,----J
OH
HO' 0=P¨OH NH2 O

OH (/
i N
N...-,J

H H
N ___________________________________________________________ NH __ 0 P=0 0¨' Metabolite 3 H

0=P¨OH NH2 OH

H H
N N
0 _______________________________________________________________ 17-0 Metabolite 4 N

0=P¨OH NH2 O
islx-IN., N

OH
<N N-----1 0--___ 0 0 N
H
H2N N 0 __ 0 ..-----Metabolite 5 HN

0=P¨OH NH2 O

OH
N,--1 ______________________________________________________________________ /

N ___________________________________________________________ H2N l' 11 0 =0 Metabolite 6 HN

Example 21: Antisense inhibition of human ApoC III in human ApoC III
transgenic mice in single administration study ISIS 304801, 647535 and 647536 each targeting human ApoC III and described in Table 17, were further evaluated in a single administration study for their ability to inhibit human ApoC III in human ApoC
III transgenic mice.
Treatment Human ApoCIII transgenic mice were maintained on a 12-hour light/dark cycle and fed ad libitum Teklad lab chow. Animals were acclimated for at least 7 days in the research facility before initiation of the experiment. ASOs were prepared in PBS and sterilized by filtering through a 0.2 micron filter. ASOs were dissolved in 0.9% PBS for injection.
Human ApoC III transgenic mice were injected intraperitoneally once at the dosage shown below with ISIS 304801, 647535 or 647536 (described above) or with PBS treated control. The treatment group consisted of 3 animals and the control group consisted of 4 animals. Prior to the treatment as well as after the last dose, blood was drawn from each mouse and plasma samples were analyzed.
The mice were sacrificed 72 hours following the last administration.
Samples were collected and analyzed to determine the ApoC III mRNA and protein levels in the liver; plasma triglycerides; and cholesterol, including HDL and LDL fractions were assessed as described above (Example 20). Data from those analyses are presented in Tables 24-28, below. Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in serum were measured relative to saline injected mice using standard protocols. The ALT and AST
levels showed that the antisense compounds were well tolerated at all administered doses.
These results show improvement in potency for antisense compounds comprising a Ga1NAc3-1 conjugate at the 3' terminus (ISIS 647535 and 647536) compared to the antisense compound lacking a Ga1NAc3-1 conjugate (ISIS 304801). Further, ISIS 647536, which comprises a Ga1NAc3-1 conjugate and some phosphodiester linkages was as potent as ISIS 647535, which comprises the same conjugate and all internucleoside linkages within the ASO are phosphorothioate.
Table 24 Effect of ASO treatment on ApoC III mRNA levels in human ApoC III transgenic mice A Dose ED50 3' Internucleoside SEQ
ID
SO % PBS
(mg/kg) (mg/kg) Conjugate linkage/Length No.

13.2 None PS/20 244 ISIS 0.3 98 1.9 Ga1NAc3-1 PS/20 245 0.3 103 1.7 Ga1NAc3-1 PS/PO/20 245 Table 25 Effect of ASO treatment on ApoC III plasma protein levels in human ApoC III
transgenic mice Dose ED50 3' Internucleoside SEQ ID
ASO % PBS
(mg/kg) (mg/kg) Conjugate Linkage/Length No.

1 104 23.2 None PS/20 244 0.3 98 2.1 Ga1NAc3-1 PS/20 245 0.3 103 1.8 Ga1NAc3-1 PS/PO/20 245 5 Table 26 Effect of ASO treatment on triglyceride levels in transgenic mice Dose ED50 Internucleoside SEQ ID
ASO % PBS 3' Conjugate (mg/kg) (mg/kg) Linkage/Length No.

29.1 None PS/20 244 0.3 100 2.2 Ga1NAc3-1 PS/20 245 ISIS 0.3 95 1.9 Ga1NAc3-1 PS/PO/20 245 Table 27 Effect of ASO treatment on total cholesterol levels in transgenic mice Dose Internucleoside ASO % PBS 3' ConjugateSEQ ID No.
(mg/kg) Linkage/Length None PS/20 244 0.3 93 Ga1NAc3-1 PS/20 245 0.3 115 Ga1NAc3-1 PS/PO/20 245 5 Table 28 Effect of ASO treatment on HDL and LDL cholesterol levels in transgenic mice Dose HDL LDL 3' Internucleoside SEQ
ID
ASO
(mg/kg) % PBS % PBS Conjugate Linkage/Length No.

None PS/20 244 0.3 98 86 Ga1NAc3-1 PS/20 245 0.3 143 89 Ga1NAc3-1 PS/PO/20 245 These results confirm that the Ga1NAc3-1 conjugate improves potency of an antisense compound.
The results also show equal potency of a Ga1NAc3-1 conjugated antisense compounds where the antisense oligonucleotides have mixed linkages (ISIS 647536 which has six phosphodiester linkages) and a full phosphorothioate version of the same antisense compound (ISIS 647535).
Phosphorothioate linkages provide several properties to antisense compounds.
For example, they resist nuclease digestion and they bind proteins resulting in accumulation of compound in the liver, rather than in the kidney/urine. These are desirable properties, particularly when treating an indication in the liver.
However, phosphorothioate linkages have also been associated with an inflammatory response. Accordingly, reducing the number of phosphorothioate linkages in a compound is expected to reduce the risk of inflammation, but also lower concentration of the compound in liver, increase concentration in the kidney and urine, decrease stability in the presence of nucleases, and lower overall potency. The present results show that a Ga1NAc3-1 conjugated antisense compound where certain phosphorothioate linkages have been replaced with phosphodiester linkages is as potent against a target in the liver as a counterpart having full phosphorothioate linkages. Such compounds are expected to be less proinflammatory (See Example 24 describing an experiment showing reduction of PS results in reduced inflammatory effect).
Example 22: Effect of GaINAc3-1 conjugated modified ASO targeting SRB-1 in vivo ISIS 440762 and 651900, each targeting SRB-1 and described in Table 17, were evaluated in a dose-dependent study for their ability to inhibit SRB-1 in Balb/c mice.
Treatment Six week old male Balb/c mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once at the dosage shown below with ISIS 440762, 651900 or with PBS treated control. Each treatment group consisted of 4 animals. The mice were sacrificed 48 hours following the final administration to determine the SRB-1 mRNA levels in liver using real-time PCR and RIBOGREENO
RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols.
SRB-1 mRNA levels were determined relative to total RNA (using Ribogreen), prior to normalization to PBS-treated control. The results below are presented as the average percent of SRB-1 mRNA levels for each treatment group, normalized to PBS-treated control and is denoted as "% PBS".
As illustrated in Table 29, both antisense compounds lowered SRB-1 mRNA
levels. Further, the antisense compound comprising the Ga1NAc3-1 conjugate (ISIS 651900) was substantially more potent than the antisense compound lacking the Ga1NAc3-1 conjugate (ISIS 440762). These results demonstrate that the potency benefit of Ga1NAc3-1 conjugates are observed using antisense oligonucleotides complementary to a different target and having different chemically modified nucleosides, in this instance modified nucleosides comprise constrained ethyl sugar moieties (a bicyclic sugar moiety).

Table 29 Effect of ASO treatment on SRB-1 mRNA levels in Balb/c mice Internucleosid Dose Liver ED50SEQ ID
ASO 3' Conjugate (mg/kg) % PBS (mg/kg) linkage/Lengt No.

0.7 85 440762 7 12 2.2 None PS/14 0.07 98 0.2 63 ISIS
00 0.7 20 0.3 GaINAc3-1 PS/14 Example 23: Human Peripheral Blood Mononuclear Cells (hPBMC) Assay Protocol The hPBMC assay was performed using BD Vautainer CPT tube method. A sample of whole blood from volunteered donors with informed consent at US HealthWorks clinic (Faraday & El Camino Real, Carlsbad) was obtained and collected in 4-15 BD Vacutainer CPT 8 ml tubes (VWR
Cat.# BD362753). The approximate starting total whole blood volume in the CPT tubes for each donor was recorded using the PBMC assay data sheet.
The blood sample was remixed immediately prior to centrifugation by gently inverting tubes 8-10 times. CPT tubes were centrifuged at rt (18-25 C) in a horizontal (swing-out) rotor for 30 min. at 1500-1800 RCF with brake off (2700 RPM Beckman Allegra 6R). The cells were retrieved from the buffy coat interface (between Ficoll and polymer gel layers); transferred to a sterile 50 ml conical tube and pooled up to 5 CPT
tubes/50 ml conical tube/donor. The cells were then washed twice with PBS (Ca, Mg ++ free; GIBCO). The tubes were topped up to 50 ml and mixed by inverting several times. The sample was then centrifuged at 330 x g for 15 minutes at rt (1215 RPM in Beckman Allegra 6R) and aspirated as much supernatant as possible without disturbing pellet. The cell pellet was dislodged by gently swirling tube and resuspended cells in RPMI+10% FBS+pen/strep (-1 ml / 10 ml starting whole blood volume). A 60 1 sample was pipette into a sample vial (Beckman Coulter) with 600 1 VersaLyse reagent (Beckman Coulter Cat# A09777) and was gently vortexed for 10-15 sec. The sample was allowed to incubate for 10 min.
at rt and being mixed again before counting. The cell suspension was counted on Vicell XR cell viability analyzer (Beckman Coulter) using PBMC cell type (dilution factor of 1:11 was stored with other parameters). The live cell/m1 and viability were recorded. The cell suspension was diluted to 1 x 107 live PBMC/ml in RPMI+ 10%
FBS+pen/strep.

The cells were plated at 5 x 105 in 50 [tl/well of 96-well tissue culture plate (Falcon Microtest). 50 [Wwell of 2x concentration oligos/controls diluted in RPMI+10% FBS+pen/strep.
was added according to experiment template (100 [Ll/well total). Plates were placed on the shaker and allowed to mix for approx. 1 min. After being incubated for 24 hrs at 37 C; 5% CO2, the plates were centrifuged at 400 x g for 10 minutes before removing the supernatant for MSD cytokine assay (i.e. human IL-6, IL-10, IL-8 and MCP-1).
Example 24: Evaluation of Proinflammatory Effects in hPBMC Assay for GaINAc3-1 conjugated ASOs The antisense oligonucleotides (ASOs) listed in Table 30 were evaluated for proinflammatory effect in hPBMC assay using the protocol described in Example 23. ISIS 353512 is an internal standard known to be a high responder for IL-6 release in the assay. The hPBMCs were isolated from fresh, volunteered donors and were treated with ASOs at 0, 0.0128, 0.064, 0.32, 1.6, 8, 40 and 200 [LM
concentrations. After a 24 hr treatment, the cytokine levels were measured.
The levels of IL-6 were used as the primary readout. The EC50 and Emax was calculated using standard procedures. Results are expressed as the average ratio of Emax/EC50 from two donors and is denoted as "Emax/EC50." The lower ratio indicates a relative decrease in the proinflammatory response and the higher ratio indicates a relative increase in the proinflammatory response.
With regard to the test compounds, the least proinflammatory compound was the PS/P0 linked ASO
(ISIS 616468). The Ga1NAc3-1 conjugated ASO, ISIS 647535 was slightly less proinflammatory than its non-conjugated counterpart ISIS 304801. These results indicate that incorporation of some PO linkages reduces proinflammatory reaction and addition of a Ga1NAc3-1 conjugate does not make a compound more proinflammatory and may reduce proinflammatory response. Accordingly, one would expect that an antisense compound comprising both mixed PS/P0 linkages and a Ga1NAc3-1 conjugate would produce lower proinflammatory responses relative to full PS linked antisense compound with or without a Ga1NAc3-1 conjugate. These results show that Ga1NAc3_1 conjugated antisense compounds, particularly those having reduced PS content are less proinflammatory.
Together, these results suggest that a Ga1NAc3-1 conjugated compound, particularly one with reduced PS content, can be administered at a higher dose than a counterpart full PS antisense compound lacking a Ga1NAc3-1 conjugate. Since half-life is not expected to be substantially different for these compounds, such higher administration would result in less frequent dosing.
Indeed such administration could be even less frequent, because the Ga1NAc3-1 conjugated compounds are more potent (See Examples 20-22) and re-dosing is necessary once the concentration of a compound has dropped below a desired level, where such desired level is based on potency.

Table 30 Modified ASOs ASO Sequence (5' to 3') Target SEQ ID
No.
ISIS GesmCesTesGesAesTdsTdsAdsGdsAdsGds 104838 AdsGdsAdsGdsGesTesmCesmCesmCe TNFa 248 ISIS TesmCesmCesmCdsAdsrfdsrrdsTdsmCdsAdsGds CRP 249 353512 GdsAdsGdsAdsmCdsmCdsTesGesGe ISIS ikesGesmCesTesTesmCdsTdsTdsGdsTds ANC III 244 304801 mCdsmCdsAdsGdsmCds TesTesTesAesTe ISIS ikesGesmCesTesTesmCdsTdsTdsGdsTds ApoC III 245 647535 mCdsmCdsAdsGdsmCdsTesTesTesAesTeoAdo,-GaINAC3-la ISIS ikesGeomCeoTeoTeomCdsTdsTdsGdsTds ApoC III 244 616468 mCdsmCdsAdsGdsmCdsTeoTeoTesAesTe Subscripts: "e" indicates 2'-MOE modified nucleoside; "d" indicates 13-D-2'-deoxyribonucleoside; "k" indicates 6'-(S)-CH3 bicyclic nucleoside (e.g. cEt);
"s" indicates phosphorothioate internucleoside linkages (PS); "o" indicates phosphodiester internucleoside linkages (PO); and "o¨ indicates -0-P(=0)(OH)-. Superscript "m" indicates 5-methylcytosines. "Ado¨Ga1NAc3-1a"
indicates a conjugate having the structure Ga1NAc3-1 shown in Example 9 attached to the 3'-end of the antisense oligonucleotide, as indicated.
Table 31 Proinflammatory Effect of ASOs targeting ApoC III in hPBMC assay ASO
EC50 Emax E /EC 3' Internucleoside SEQ ID
(11M) (11M) max 5 0 Conjugate Linkage/Length No.

0.01 265.9 26,590 None PS/20 (high responder) ISIS 304801 0.07 106.55 1,522 None PS/20 ISIS 647535 0.12 138 1,150 Ga1NAc3-1 PS/20 ISIS 616468 0.32 71.52 224 None PS/PO/20 Example 25: Effect of GaINAc3-1 conjugated modified ASO targeting human ApoC
III in vitro ISIS 304801 and 647535 described above were tested in vitro. Primary hepatocyte cells from transgenic mice at a density of 25,000 cells per well were treated with 0.03,0.08, 0.24, 0.74, 2.22, 6.67 and 20 [LM concentrations of modified oligonucleotides. After a treatment period of approximately 16 hours, RNA

was isolated from the cells and mRNA levels were measured by quantitative real-time PCR and the hApoC
III mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN.
The IC50 was calculated using the standard methods and the results are presented in Table 32. As illustrated, comparable potency was observed in cells treated with ISIS 647535 as compared to the control, ISIS 304801.
Table 32 Modified ASO targeting human ApoC III in primary hepatocytes Internucleoside SEQ
ASO ICso (111\4) 3 Conjugate linkage/Length ID No.
ISIS
0.44 None PS/20 244 ISIS
0.31 Ga1NAc3-1 PS/20 245 In this experiment, the large potency benefits of Ga1NAc3-1 conjugation that are observed in vivo were not observed in vitro. Subsequent free uptake experiments in primary hepatocytes in vitro did show increased potency of oligonucleotides comprising various GalNAc conjugates relative to oligonucleotides that lacking the GalNAc conjugate.(see Examples 60, 82, and 92) Example 26: Effect of PO/PS linkages on ApoC III ASO Activity Human ApoC III transgenic mice were injected intraperitoneally once at 25 mg/kg of ISIS 304801, or ISIS 616468 (both described above) or with PBS treated control once per week for two weeks. The treatment group consisted of 3 animals and the control group consisted of 4 animals. Prior to the treatment as well as after the last dose, blood was drawn from each mouse and plasma samples were analyzed. The mice were sacrificed 72 hours following the last administration.
Samples were collected and analyzed to determine the ApoC III protein levels in the liver as described above (Example 20). Data from those analyses are presented in Table 33, below.
These results show reduction in potency for antisense compounds with PO/PS
(ISIS 616468) in the wings relative to full PS (ISIS 304801).
Table 33 Effect of ASO treatment on ApoC III protein levels in human ApoC III
transgenic mice PBS
ASO
Dose 3' Internucleoside SEQ ID

(mg/kg) Conjugate linkage/Length No.

ISIS
mg/kg/wk 24 None Full PS 244 for 2 wks ISIS
mg/kg/wk 40 None 14 PS/6 PO 244 for 2 wks Example 27: Compound 56 N(iPr)2 DMTO
o oCN

,---......,.......-----.0 Compound 56 is commercially available from Glen Research or may be prepared according to published procedures reported by Shchepinov et al., Nucleic Acids Research, 1997, 25(22), 4447-4454.
Example 28: Preparation of Compound 60 Ac0 OAc Ac0 OAc Ac0 _....7(.2..\ HO,...-..........,-....,...-.......õ..0Bn 57 __......r...\,0 0 H2/Pd ________________________________________ Ac0 OBn Me0H
TMSOTf, DCE
1\1"--1 AcHN 58 (quant.) ( 71%) CNEtO(N(iPr)2)PC1, Ac0 OAc Ac0 OAc N(iPr)2 ED1P __..........\,0 0 ,I
___...2..\,0 Ac0 Ac0 OH CH202 10 AcHN 59 (80%) AcHN 60 Compound 4 was prepared as per the procedures illustrated in Example 2.
Compound 57 is commercially available. Compound 60 was confirmed by structural analysis.
Compound 57 is meant to be representative and not intended to be limiting as other monoprotected substituted or unsubstituted alkyl diols including but not limited to those presented in the specification herein
15 can be used to prepare phosphoramidites having a predetermined composition.
Example 29: Preparation of Compound 63 CN
1. BnC1 .õ..OH 1. DMTC1, pyr H
,..0DMT

HO 2. KOH, DMSO , Bno 2. Pd/C, H2 __ 0õ
\ ________ cCH3 ____________________ .----------OH . p ---------ODMT
3. HC1, Me0H 3. Phosphitylation 0 kop02 ---ODMT
4. NaHCO 3 '--.0E-I

Compounds 61 and 62 are prepared using procedures similar to those reported by Tober et al., Eur. J.
Org. Chem., 2013, 3, 566-577; and Jiang et al., Tetrahedron, 2007, 63(19), 3982-3988.
Alternatively, Compound 63 is prepared using procedures similar to those reported in scientific and patent literature by Kim et al., Synlett, 2003, 12, 1838-1840; and Kim et al., published PCT International Application, WO 2004063208.
Example 30: Preparation of Compound 63b OH ODMT
CN

1. DMTC1, pyr 2. TBAF
3. Phosphitylation I
N(iPr)2 63a OH 63b ODMT
Compound 63a is prepared using procedures similar to those reported by Hanessian et al., Canadian Journal of Chemistry, 1996, 74(9), 1731-1737.
Example 31: Preparation of Compound 63d HO _\ DMT0¨\
0, N(iPr)2 0õ 1. DMTC1, pyr HOOOOB 2. Pd/C, H2 3. Phosphitylation 63c 63d HO¨/ DMT0¨/
Compound 63c is prepared using procedures similar to those reported by Chen et al., Chinese Chemical Letters, 1998, 9(5), 451-453.
Example 32: Preparation of Compound 67 CO2Bn Ac0 OAc 0 ,OTBDMS Ac0 OAc H2N 0 CO2Bn Ac0 OH ________________ Ac0 /0).c OTBDMS
AcHN 64 HBTU, DIEA AcHN 66 R = H or CH3 1. TEA.3HF Ac0 OAc, THF 0 CO2Bn 2. Phosphitylation H I
AcHN R N(iPr)2 Compound 64 was prepared as per the procedures illustrated in Example 2.
Compound 65 is prepared using procedures similar to those reported by Or et al., published PCT International Application, 20 WO 2009003009. The protecting groups used for Compound 65 are meant to be representative and not intended to be limiting as other protecting groups including but not limited to those presented in the specification herein can be used.
Example 33: Preparation of Compound 70 OBn Ac0 OAc H2N 68 0 CH3 Ac0 OAc ___.....2..\ro ).( IIHTU, DA Ac0 OH _______________________________ N
___......Ø..\r ).- Ac0 o OBn AcHN 64 MT
AcHN 69 H

Ac0 OAc 1. Pd/C, H2 0 __....Ø..\ r 0 - Ac0 2. Phosphitylation H I I
AcHN CH3 N(iPr)2 Compound 64 was prepared as per the procedures illustrated in Example 2.
Compound 68 is commercially available. The protecting group used for Compound 68 is meant to be representative and not intended to be limiting as other protecting groups including but not limited to those presented in the specification herein can be used.
Example 34: Preparation of Compound 75a 1. TBDMSC1, pyr 2. Pd/C, H2 HN
N(iPr)2 ------,....-- 0 ----_ 31 NC 3. CF3CO2Et, Me0H H
NC ---"\-- -,./-Th)H ____________________ x. F3C /1\I
NC o 4. TEA.314F, THF 1 5. Phosphitylation 75 OCF 3 75a Compound 75 is prepared according to published procedures reported by Shchepinov et al., Nucleic Acids Research, 1997, 25(22), 4447-4454.

Example 35: Preparation of Compound 79 DMTO 0 HO,..õ.....7".õ,,..0,.
-...,,...õ..---...õ.0,õ DCI, NMI, ACN
1. BnCl, NaH
DMTO.,..0 /------OH ____________________ ).- HO ..-----\----C'OBn Phosphoramidite 60 _____________________________________________________________________________ x-0 2. DCA, CH2C12 õ..-..,......õ,---...0,--DMTO HO

Ac0 OAc NC m Ac0 0 0 P
AcHN
NC
1. H2/Pd, Me0H
--_\
Ac0 OAc ' L
0,õ O 2. Phosphitylation Ac0 ,0=11), ,/\/', / \.---. Bn AcHN (:) NC ----\
Ac0 OAc Ac0 NHAc 78 Ac0 OAc NC -...1 Ac0 0 0 AcHN
--_\
Ac0 OAc NC
LO 0,, v(I)ci\T

AcHN (:) 1 NC ---\NOT)02 Ac0 OAc 1.Ø..._\/0 ID'PO
Ac0 NHAc Compound 76 was prepared according to published procedures reported by Shchepinov et al., Nucleic Acids Research, 1997, 25(22), 4447-4454.
Example 36: Preparation of Compound 79a HO
Fmoc0 -.......7"..õ-:h.00..., 1. FmocC1, pyr -,......7",..õ- NOP021 HO0-....,....õ1 OBn 2. Pd/C, H2 Fmoc0-0 ,P, CN

_________________________________________ ,.-7-..,...7----......-1 .õ--...,....
HO 0 3. Phosphitylation Fmoc0 ,----...0 77 79a Compound 77 is prepared as per the procedures illustrated in Example 35.
Example 37: General method for the preparation of conjugated oligomeric compound 82 comprising a phosphodiester linked Ga1NAc3-2 conjugate at 5' terminus via solid support (Method I) Bx ,....._0 ( --/ 7---/7ODMT
n---,--ODMT
DMT0((i)r - -----Z--ODMT
0 u NC0-P=0 1. DCA, DCM NC 00 oBx 0 2. DCI, NMI, ACN 0 I
OLIGO Phosphoramidite 56 NC0-P=0 , ______________________________________________ , s. ____________________ ., DNA/RNA 0 I
0 ,.autom , I
ated synthesizer . , ' I OLIGO
0¨VIMAD_0_p_oCN ' __ 1 31( 0 79b I
_0_130CN
0¨VIMAD
X = S- or 0- X
Bx = Heterocylic base 1. Capping (Ac20, NMI, pyr) 80 2. t-BuO0H
3. DCA, DCM
4. DCI, NMI, ACN
Ac00Ac NC ---.\
Phosphoramidite 60 0 , 0 Ac0--*......\k-)..õ--N.______\ ,k AcHN 0 0¨

CN
-----\
AcO0Ac NC of 0 0-, Bx AcHN 0' 0 NC -----\ y NC 'Th -P =0 Ac0 Ac ---0 0I
....12...\/0õ,...z----õZ"-----70' i3-0 ' , OLIGO
Ac0 ' __ 1 1 .' NHAc 0 I
0¨VIMAD
X
1. Capping (Ac20, NMI, pyr) 81 2. t-BuO0H
3. 20% Et2NH inToluene (v/v) 4. NH4, 55 C, HO OH
0 , HO----4u 0 ....--N...,.......õ--\ Ag AcHN 0 1 0 HO OH 0, 0 r, 0 ON.4.Bx AcHN 0 0' O.
0=P-0-P
y , (5 HOOH

, OLIGO
HO, _________________________________________________________________ , NHAc 82 wherein Ga1NAc3-2 has the structure:
HOOH

-.....----......õ..¨..õ_õ...-\ A....
AcHN 0 0-0¨
HOOH
__....72..\/-, 0 0, 9 HO %-, P
, õ..-..,..õ....-... -....-0 0 ,,0 0,- -P-Oc0rBx AcHN (:) ,P- I
1.2_\/0C1 60 HO
NHAc The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-2 (Ga1NAc3-2a) can be combined with any cleavable moiety to provide a variety of conjugate groups. Wherein Ga1NAc3-2a has the formula:
HOOH
HO.*.2..\.0 0 AcHN 0 I 'D¨

O-HOOH
0 0, _.....72..\;-%
o , o o-AcHN (:) HO OH y P, HO
NHAc The VIMAD-bound oligomeric compound 79b was prepared using standard procedures for automated DNA/RNA synthesis (see Dupouy et al., Angew. Chem. Int. Ed, 2006, 45, 3623-3627). The phosphoramidite Compounds 56 and 60 were prepared as per the procedures illustrated in Examples 27 and 28, respectively. The phosphoramidites illustrated are meant to be representative and not intended to be limiting as other phosphoramidite building blocks including but not limited those presented in the specification herein can be used to prepare an oligomeric compound having a phosphodiester linked conjugate group at the 5' terminus. The order and quantity of phosphoramidites added to the solid support can be adjusted to prepare the oligomeric compounds as described herein having any predetermined sequence and composition.

Example 38: Alternative method for the preparation of oligomeric compound 82 comprising a phosphodiester linked Ga1NAc3-2 conjugate at 5' terminus (Method II) DMT0 )' Bx CY 1. DCA, DCM
NC 1 ___________________________ ..-O¨P=0 2. DCI, NMI, ACN

1 Phosphoramidite 79 OLIGO DNA/RNA
. ., I µautomated synthesizer _ I X= S- or 0-CI¨VIMAD¨O¨P-0CN
K Bx ¨ Heterocyclic base 79b Ac0 OAc NC m 0 0¨

AcH N
Ac0 OAc CN
NC ---\___ I
0 0,, 0 Ac0 ___,.Ø....\z, jrN ,i), (31_1_030).õõBx AcHN ICI O.
NC---\.... y NC 1 O¨P=0 Ac0 OAc I
OLIGO
Ac0 ,. ________ I -N HAc 0 I
1. Capping Ci¨VIMAD-0¨P-0CN
K
2. t-BuO0H
3. Et3N:CH3CN (1:1 v/v) 83 4. NH4, 55 C
Oligomeric Compound 82 The VIMAD-bound oligomeric compound 79b was prepared using standard procedures for automated DNA/RNA synthesis (see Dupouy et al., Angew. Chem. Int. Ed, 2006, 45, 3623-3627). The Ga1NAc3-2 cluster phosphoramidite, Compound 79 was prepared as per the procedures illustrated in Example 35. This alternative method allows a one-step installation of the phosphodiester linked Ga1NAc3-2 conjugate to the oligomeric compound at the final step of the synthesis. The phosphoramidites illustrated are meant to be representative and not intended to be limiting, as other phosphoramidite building blocks including but not limited to those presented in the specification herein can be used to prepare oligomeric compounds having a phosphodiester conjugate at the 5' terminus. The order and quantity of phosphoramidites added to the solid support can be adjusted to prepare the oligomeric compounds as described herein having any predetermined sequence and composition.

Example 39: General method for the preparation of oligomeric compound 83h comprising a Ga1NAc3-3 Conjugate at the 5' Terminus (Ga1NAc3-1 modified for 5' end attachment) via Solid Support Ac0 OAc Ac0--0 H
N---N----)r-N H 1 H2, Pd/C, Me0H (93%) AcHN \....--\,,.N
0 0 y-y0H
H H 0 (:)õ

83a .\?....\oAc 0....y......./-1 N----N--Ny----0..._.--N 0 go BnO0 0 Ac0 H
, DIEA, DMF, 76%
Ac0 /
HBTU ____________________________________________________________ i.-NHAc HNN ____,3.H2,Pd/C,Me0H
õ
/¨/-0 OAc H ,-/
Ac0 ........r.00Ac Ac0e Ac0 Ac0 0 H
NHAc N---N---)T--N H
AcHN \---NN

OH

0 N OAc NN.õ-N
83b F
F Ac0 F NHAc HN N VN/N ____ ,_, 83c - __________________________ Pyridine, DMF
OAc 01-1--H k-/

Ac0 _.....r.2.0Ac Ac0 AcC/
NHAc Ac0 0 H 3'0 5'1 II 83e N--N----)r--N H
F ( OLIGO -0-P-0-(CH2)6-AcHN n 0 0 F j (:)_õ, )L7)IN441, OH
N F
a-0 Borate buffer, DMSO, pH 8.5, rt Ac0,0 Ac0 NHAc HN N F F
OAc j¨r1/40 k-/ ,-"N.----N ____õe ,_, H 83d Ac0\&\.>1 Ac0 NHAc Ac0 OAc Ac00 H
N"--------)T--N H
AcHN \,---\,,.N..........õ, H H 8 L ,L,) __ I __ N N-(CH2)6-0-P-0¨( OLIGO ) Ac0,0 H I I
Ac0 NHAc HN N VN/N ____ 83f OAc H k-/
,_, _7¨/-0 \&\....
Ac0 Ac0 NHAc Aqueous ammonia Th¨ N H
0 )rm H H0....- _____ NH N-(CH2)6-0-P-0¨ [
OLIGO ) HO OH

0,... H 11 HO
V./7....\/ H

0 0 ____e 83h HN N
NHAc H
/

OH j 0 1-11--) NHAc Compound 18 was prepared as per the procedures illustrated in Example 4.
Compounds 83a and 83b are commercially available. Oligomeric Compound 83e comprising a phosphodiester linked hexylamine was prepared using standard oligonucleotide synthesis procedures. Treatment of the protected oligomeric compound with aqueous ammonia provided the 5'-Ga1NAc3-3 conjugated oligomeric compound (83h).
Wherein Ga1NAc3-3 has the structure:
HO OH
HO-----ro H
Ni.--N
AcHN H
0 ni\I
)r o 0 OH
H H)L7).-L I
N---rf----N-0.----NH

HO H H
HO
NHAc _4) HI\IN OH
HO
NHAc .
The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-3 (Ga1NAc3-3a) can be combined with any cleavable moiety to provide a variety of conjugate groups. Wherein Ga1NAc3-3a has the formula:

HO OH

AcHN N---N---)---N H
O
H H 0 0...
7X---N-ii-N--0..---NH N¨(CH2)6-0¨

H
0 0"
HO
NHAc HN N
OH
HO\
HO
NHAc .

Example 40: General method for the preparation of oligomeric compound 89 comprising a phosphodiester linked Ga1NAc3-4 conjugate at the 3' terminus via solid support (0,/--/ODMT
1. DCA
0-UNL-ODMT _______________________________ 0 07---õ,---OFmoc 2. DCI, NMI, ACN I

Fmoc0 õ..-0--... N(iPr)2 P.Ø------õ,CN 85 DMT0,---õ00' 3. Capping ODMT (CN
4. t-BuO0H r0--7---/
..--i r OFmoc 0/ /-0Fmoc 1. 2% Piperidine, r\-- 70I0 __ Fo/
2% DBU, 96% DMF 0 0 OFmoc 3. DCI, NMI, ACN 86 Phosphoramidite 79a 0-UNL-0-13,-0CN

' DNA/RNA ' 1. Capping ,automated synthesizer 2. t-BuO0H, 3. 2% Piperidine, Ac0 OAc 2% DBU, 96% DMF
AcONi 4. DCI, NMI, ACN
o Phosphoramidite 60 , _____________________________________________________________ , AcHN 0 DNA/RNA
,automated synthesizer, \--\__\____\NC
( 5. Capping Ac0 OAc p r O-P
Ac0-.....,A) _ NC \
0\--\--\------\1,9 0 j--CN

AcHN
P=0 NC-\__Q
Ac z__/0-13--Ac0 -() _________________ \..(:...)...\/0 DMTO"N-----No_}---\

I , zCN
Ac0 0-UNL-0-13,-NHAc 1. t-BuO0H o 2. DCA
3. Oligo synthesis (DNA/RNA automated synthesizer) 4. Capping 5. Oxidation 6. Et3N:CH3CN (1:1, v/v) Ac0 OAc Ac0*\01_ AcHN 0 AcO 0-p' Lo -0' O
'------\-o AcHN

P=0 IP------\--0 Ac0\1*/ Ac\0_ -0\ /0--\----No_F-N
Ac0 DMT-[ OLIGO }------Pt o_uNL_O-1t-cr _ ,CN
NHAc 5' 3' HO OH NH4, 55 C
H0*\0L
AcHN 0 0-p' HOo 0-N----\--0 AcHN
\---\--\---\P- 89 (?
r--' 0 =,--0 HO..._\/ 1-1\O-j-----\OH
õ,--...../-HO
NHAc I OLIGO j __ (CM __ e 0 5' 3' Wherein Ga1NAc3-4 has the structure:

HON
AcHN 0 HO OH \---\----\---\ p HO / \
0\--\--\------\ 9 0- o AcHN
r.----0 I
o-p. 1 07----/ P=0 \---0 HO(2..\/0 H 0" \
\\. 0-03)---\ce NHAc 1¨(CMY
Wherein CM is a cleavable moiety. In certain embodiments, cleavable moiety is:

0=P-OH ,J\I NH2 I \ -4 \'N
O-NafrN N.,......j CI
I
0=P-OH
I
The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-4 (Ga1NAc3-4a) can be combined with any cleavable moiety to provide a variety of conjugate groups. Wherein Ga1NAc3-4a has the formula:

HO oFi HO*N__ AcHN 0 HO OH \---\---\---\ p 0-Pi HO / \c, 0\--\--\------\ 90-AcHN 0-u ,-, P=0 u-o I
()0 HO H 0' \o-LC) , Pn)-------\OH
.1.1.,, -NHAc The protected Unylinker functionalized solid support Compound 30 is commercially available.
Compound 84 is prepared using procedures similar to those reported in the literature (see Shchepinov et al., Nucleic Acids Research, 1997, 25(22), 4447-4454; Shchepinov et al., Nucleic Acids Research, 1999, 27, 3035-3041; and Hornet et al., Nucleic Acids Research, 1997, 25, 4842-4849).
The phosphoramidite building blocks, Compounds 60 and 79a are prepared as per the procedures illustrated in Examples 28 and 36. The phosphoramidites illustrated are meant to be representative and not intended to be limiting as other phosphoramidite building blocks can be used to prepare an oligomeric compound having a phosphodiester linked conjugate at the 3' terminus with a predetermined sequence and composition. The order and quantity of phosphoramidites added to the solid support can be adjusted to prepare the oligomeric compounds as described herein having any predetermined sequence and composition.
Example 41: General method for the preparation of ASOs comprising a phosphodiester linked Ga1NAc3-2 (see Example 37, Bx is adenine) conjugate at the 5' position via solid phase techniques (preparation of ISIS 661134) Unless otherwise stated, all reagents and solutions used for the synthesis of oligomeric compounds are purchased from commercial sources. Standard phosphoramidite building blocks and solid support are used for incorporation nucleoside residues which include for example T, A, G, and mC residues.
Phosphoramidite compounds 56 and 60 were used to synthesize the phosphodiester linked Ga1NAc3-2 conjugate at the 5' terminus. A 0.1 M solution of phosphoramidite in anhydrous acetonitrile was used for 13-D-2'-deoxyribonucleoside and 2'-M0E.
The ASO syntheses were performed on ABI 394 synthesizer (1-2 [mot scale) or on GE Healthcare Bioscience AKTA oligopilot synthesizer (40-200 [mot scale) by the phosphoramidite coupling method on VIMAD solid support (110 [tmol/g, Guzaev et al., 2003) packed in the column.
For the coupling step, the phosphoramidites were delivered at a 4 fold excess over the initial loading of the solid support and phosphoramidite coupling was carried out for 10 min. All other steps followed standard protocols supplied by the manufacturer. A solution of 6% dichloroacetic acid in toluene was used for removing the dimethoxytrityl (DMT) groups from 5'-hydroxyl groups of the nucleotide. 4,5-Dicyanoimidazole (0.7 M) in anhydrous CH3CN was used as activator during the coupling step.
Phosphorothioate linkages were introduced by sulfurization with 0.1 M solution of xanthane hydride in 1:1 pyridine/CH3CN for a contact time of 3 minutes. A solution of 20% tert-butylhydroperoxide in CH3CN containing 6%
water was used as an oxidizing agent to provide phosphodiester internucleoside linkages with a contact time of 12 minutes.
After the desired sequence was assembled, the cyanoethyl phosphate protecting groups were deprotected using a 20% diethylamine in toluene (v/v) with a contact time of 45 minutes. The solid-support bound ASOs were suspended in aqueous ammonia (28-30 wt %) and heated at 55 C
for 6 h.
The unbound ASOs were then filtered and the ammonia was boiled off The residue was purified by high pressure liquid chromatography on a strong anion exchange column (GE
Healthcare Bioscience, Source 30Q, 30 [tin, 2.54 x 8 cm, A = 100 mM ammonium acetate in 30% aqueous CH3CN, B =
1.5 M NaBr in A, 0-40%
of B in 60 min, flow 14 mL min-1, k = 260 nm). The residue was desalted by HPLC on a reverse phase column to yield the desired ASOs in an isolated yield of 15-30% based on the initial loading on the solid support. The ASOs were characterized by ion-pair-HPLC coupled MS analysis with Agilent 1100 MSD
system.
Table 34 ASO comprising a phosphodiester linked Ga1NAc3-2 conjugate at the 5' position targeting SRB-1 Observed SEQ
ID
ISIS No. Sequence (5 to 3') CalCd Mass Mass No.
GalNAC3-2a-otAdoTksmCksAdsGdsTdsmCdsAdsrrds 661134 6482.2 6481.6 250 Gds AdsmCdsTdsTksmCk Subscripts: "e" indicates 2'-MOE modified nucleoside;
"d" indicates 13-D-2' -deoxyribonucleoside; "k" indicates 6'-(S)-CH3 bicyclic nucleoside (e.g. cEt);
"s" indicates phosphorothioate internucleoside linkages (PS); "o" indicates phosphodiester internucleoside linkages (PO); and "o¨ indicates -0-P(=0)(OH)-. Superscript "m" indicates 5-methylcytosines. The structure of Ga1NAc3-2a is shown in Example 37.

Example 42: General method for the preparation of ASOs comprising a Ga1NAc3-3 conjugate at the 5' position via solid phase techniques (preparation of ISIS 661166) The synthesis for ISIS 661166 was performed using similar procedures as illustrated in Examples 39 and 41.
ISIS 661166 is a 5-10-5 MOE gapmer, wherein the 5' position comprises a Ga1NAc3-3 conjugate.
The ASO was characterized by ion-pair-HPLC coupled MS analysis with Agilent 1100 MSD system.
Table 34a ASO comprising a Ga1NAc3-3 conjugate at the 5' position via a hexylamino phosphodiester linkage targeting Malat-1 ISIS, Calcd Observed No.
Sequence (5' to 3') Mass Mass SEQ ID No.
5'-Ga1NAC3-3,-0,mCesGesGesTesGes 661166 mCdsAdsAdsGdsGdsmCdsTdsTdsAdsGds 5'-Ga1NAc3-3 8992.16 8990.51 251 GesAesAes TesTe Subscripts: "e" indicates 2'-MOE modified nucleoside; "d" indicates [3-D-2'-deoxyribonuc1eoside;
"s" indicates phosphorothioate internucleoside linkages (PS); "o" indicates phosphodiester internucleoside linkages (PO); and "o" indicates -0-P(=0)(OH)-. Superscript "m" indicates 5-methylcytosines. The structure of "5'-GalNAc3-3a" is shown in Example 39.
Example 43: Dose-dependent study of phosphodiester linked Ga1NAc3-2 (see examples 37 and 41, Bx is adenine) at the 5' terminus targeting SRB-1 in vivo ISIS 661134 (see Example 41) comprising a phosphodiester linked Ga1NAc3-2 conjugate at the 5' terminus was tested in a dose-dependent study for antisense inhibition of SRB-1 in mice. Unconjugated ISIS
440762 and 651900 (Ga1NAc3-1 conjugate at 3' terminus, see Example 9) were included in the study for comparison and are described previously in Table 17.
Treatment Six week old male Balb/c mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once at the dosage shown below with ISIS 440762, 651900, 661134 or with PBS
treated control. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration to determine the liver SRB-1 mRNA levels using real-time PCR and RIBOGREENO
RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols.
SRB-1 mRNA levels were determined relative to total RNA (using Ribogreen), prior to normalization to PBS-treated control. The results below are presented as the average percent of SRB-1 mRNA levels for each treatment group, normalized to PBS-treated control and is denoted as "% PBS". The ED50s were measured using similar methods as described previously and are presented below.
As illustrated in Table 35, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner. Indeed, the antisense oligonucleotides comprising the phosphodiester linked Ga1NAc3-2 conjugate at the 5' terminus (ISIS 661134) or the Ga1NAc3-1 conjugate linked at the 3' terminus (ISIS 651900) showed substantial improvement in potency compared to the unconjugated antisense oligonucleotide (ISIS 440762). Further, ISIS 661134, which comprises the phosphodiester linked Ga1NAc3-2 conjugate at the 5' terminus was equipotent compared to ISIS 651900, which comprises the Ga1NAc3-1 conjugate at the 3' terminus.
Table 35 ASOs containing Ga1NAc3-1 or Ga1NAc3-2 targeting SRB-1 ISIS Dosage SRB-1 mRNA ED50 Conjugate SE Q ID No.
No. (mg/kg) levels (% PBS) (mg/kg) 0.2 116 0.7 91 440762 2 69 2.58 No conjugate 250 0.07 95 0.2 77 651900 0.7 28 0.26 3' Ga1NAc3-1 247 0.07 107 0.2 86 661134 0.7 28 0.25 5' GalNAc3-2 250 Structures for 3' Ga1NAc3-1 and 5' Ga1NAc3-2 were described previously in Examples 9 and 37.
15 Pharmacokinetics Analysis (PK) The PK of the ASOs from the high dose group (7 mg/kg) was examined and evaluated in the same manner as illustrated in Example 20. Liver sample was minced and extracted using standard protocols. The full length metabolites of 661134 (5' Ga1NAc3-2) and ISIS 651900 (3' Ga1NAc3-1) were identified and their masses were confirmed by high resolution mass spectrometry analysis. The results showed that the major 20 metabolite detected for the ASO comprising a phosphodiester linked Ga1NAc3-2 conjugate at the 5' terminus (ISIS 661134) was ISIS 440762 (data not shown). No additional metabolites, at a detectable level, were observed. Unlike its counterpart, additional metabolites similar to those reported previously in Table 23a were observed for the ASO having the Ga1NAc3-1 conjugate at the 3' terminus (ISIS 651900). These results suggest that having the phosphodiester linked GalNAc3-1 or Ga1NAc3-2 conjugate may improve the PK
profile of ASOs without compromising their potency.
Example 44: Effect of PO/PS linkages on antisense inhibition of ASOs comprising GaINAc3-1 conjugate (see Example 9) at the 3' terminus targeting SRB-1 ISIS 655861 and 655862 comprising a Ga1NAc3-1 conjugate at the 3' terminus each targeting SRB-1 were tested in a single administration study for their ability to inhibit SRB-1 in mice. The parent unconjugated compound, ISIS 353382 was included in the study for comparison.
The ASOs are 5-10-5 MOE gapmers, wherein the gap region comprises ten 2'-deoxyribonucleosides and each wing region comprises five 2'-MOE modified nucleosides. The ASOs were prepared using similar methods as illustrated previously in Example 19 and are described Table 36, below.
Table 36 Modified ASOs comprising GaINAc3-1 conjugate at the 3' terminus targeting SRB-Chemistry SEQ
ISIS No. Sequence (5' to 3') ID
No.
353382 GesmCesTesTesmCesAdsGasTasmCdsAdsTasGasAds Full PS no conjugate 252 (parent) mCdsTdsTesmCesmCesTesTe GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds Full PS with 253 mCdsTdsTesmCesmCesTesTeoAdo,-Ga1NAC3-1 a Ga1NAc3-1 conjugate 655862 GesmCeoTeoTeomCeoAdsGdsTdsmCdsAdsTdsGdsAds Mixed PS/P0 with 253 mCdsTdsTeomCeomCesTesTeoAdo,-G alNAC3-1 a Ga1NAc3-1 conjugate Subscripts: "e" indicates 2'-MOE modified nucleoside; "d" indicates [3-D-2'-deoxyribonuc1eoside;
"s" indicates phosphorothioate internucleoside linkages (PS); "o" indicates phosphodiester internucleoside linkages (PO); and "o" indicates -0-P(=0)(OH)-. Superscript "m" indicates 5-methylcytosines. The structure of "GalNAc3-1" is shown in Example 9.
Treatment Six week old male Balb/c mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once at the dosage shown below with ISIS 353382, 655861, 655862 or with PBS
treated control. Each treatment group consisted of 4 animals. Prior to the treatment as well as after the last dose, blood was drawn from each mouse and plasma samples were analyzed. The mice were sacrificed 72 hours following the final administration to determine the liver SRB-1 mRNA levels using real-time PCR
and RIBOGREENO RNA
quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. SRB-1 mRNA
levels were determined relative to total RNA (using Ribogreen), prior to normalization to PBS-treated control. The results below are presented as the average percent of SRB-1 mRNA
levels for each treatment group, normalized to PBS-treated control and is denoted as "% PBS". The ED50s were measured using similar methods as described previously and are reported below.

As illustrated in Table 37, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner compared to PBS treated control. Indeed, the antisense oligonucleotides comprising the Ga1NAc3-1 conjugate at the 3' terminus (ISIS 655861 and 655862) showed substantial improvement in potency comparing to the unconjugated antisense oligonucleotide (ISIS 353382). Further, ISIS 655862 with mixed PS/P0 linkages showed an improvement in potency relative to full PS (ISIS
655861).
Table 37 Effect of PO/PS linkages on antisense inhibition of ASOs comprising Ga1NAc3-1 conjugate at 3' terminus targeting SRB-1 ISIS Dosage SRB-1 mRNA ED50 Chemistry SEQ ID No.
No. (mg/kg) levels (% PBS) (mg/kg) 3 76.65 10 52.40 10.4 Full PS without conjugate 252 (parent) 30 24.95 0.5 81.22 1.5 63.51 Full PS with GalNAc3-1 655861 5 24.61 2.2 conjugate 253 14.80 0.5 69.57 1.5 45.78 Mixed PS/P0 with 655862 1.3 253 5 19.70 Ga1NAc3-1 conjugate 15 12.90 Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in serum were measured relative to saline injected mice using standard protocols.
Organ weights were also 15 evaluated. The results demonstrated that no elevation in transaminase levels (Table 38) or organ weights (data not shown) were observed in mice treated with ASOs compared to PBS
control. Further, the ASO with mixed PS/P0 linkages (ISIS 655862) showed similar transaminase levels compared to full PS (ISIS 655861).
Table 38 Effect of PO/PS linkages on transaminase levels of ASOs comprising Ga1NAc3-1 conjugate at 3' terminus targeting SRB-1 ISIS Dosage ALT AST
No. (mg/kg) (U/L) (U/L) Chemistry SEQ ID No.
PBS 0 28.5 65 3 50.25 89 353382 Full PS without 10 27.5 79.3 252 (parent) conjugate 27.3 97 655861 0.5 28 55.7 Full PS with 253 1.5 30 78 Ga1NAc3-1 29 63.5 28.8 67.8 0.5 50 75.5 1.5 21.7 58.5 Mixed PS/P0 with 5 29.3 69 Ga1NAc3-1 Example 45: Preparation of PFP Ester, Compound 110a , \
HO N3 Pd/C, H2 " n OAc OAc OAc OAc Et0Ac, Me0H
103a; n=1 .....Ø..\õ,0 ..----,......N3 ___________________________________________________________________________ 1.
Ac0* 103b; n= 7 Ac0 ...n 0. AcHN
N 104a; n=1 104b; n= 7 4 OAc AcONC:Ac AcHN 0 OAc OAc OAc OAc n N
H
__T!.C...D...\
Ac0--C--)._\ -----NH2 PFPTFA
__________________________________ ).- Ac0 0 n NHIr.______NO2 AcHN DMF, Pyr AcHN n 105a; n=1 Compound 90 0 r OAc OAc 105b; n= 7 __T!.C...D...\0.----HN---.0 Ac0 n AcHN
106a; n=1 106b; n= 7 OAc AcONC:Ac AcHN 0 N
OAc OAc i n H
Ra-Ni, H2 Ac0 0 __C...:)..\ HBTU, DIEA, DMF
__________ . _________________________________________________________ )..-rNHIr\------NH2 Me0H, Et0Ac AcHN

OAc OAcBn __C...:).Ø..----j HN4 Ac0 n AcHN 99 107a; n=1 107b; n= 7 OAc AcONC:Ac AcHN 0 rt0 OAc OAc n ...C..:)..\.
Ac0 0 AcHN n NHIr.______ NH

0 r OAc OAc , \
Ac0 ......)...\õ,0,---HN ---.0 'n AcHN
108a; n=1 0 108b; n= 7 1 Bn OAc Ac0 OAc AcHN
Pd/C H2, r 108a; n=1 Et0Ac,, Me0H ¨Ac OAc 108b; n= 7 NH
AcHN \ n O

OAc OAc 0 0.----HNZ0 Ac0 'n AcHN
109a; n=1 HO
109b; n= 7 OAc Ac0 OAc AcHN
rAc OAc AcHN NH

PFPTFA, DMF, pyr OAc OAc 109a Ac0 AcHN
O
110a O F
F F
F F
Compound 4 (9.5g, 28.8 mmoles) was treated with compound 103a or 103b (38 mmoles), individually, and TMSOTf (0.5 eq.) and molecular sieves in dichloromethane (200 mL), and stirred for 16 hours at room temperature. At that time, the organic layer was filtered thru celite, then washed with sodium bicarbonate, water and brine. The organic layer was then separated and dried over sodium sulfate, filtered and reduced under reduced pressure. The resultant oil was purified by silica gel chromatography (2%-->10%
methanadichloromethane) to give compounds 104a and 104b in >80% yield. LCMS
and proton NMR was consistent with the structure.
Compounds 104a and 104b were treated to the same conditions as for compounds 100a-d (Example 47), to give compounds 105a and 105b in >90% yield. LCMS and proton NMR was consistent with the structure.
Compounds 105a and 105b were treated, individually, with compound 90 under the same conditions as for compounds 901a-d, to give compounds 106a (80%) and 106b (20%). LCMS and proton NMR was consistent with the structure.

Compounds 106a and 106b were treated to the same conditions as for compounds 96a-d (Example 47), to give 107a (60%) and 107b (20%). LCMS and proton NMR was consistent with the structure.
Compounds 107a and 107b were treated to the same conditions as for compounds 97a-d (Example 47), to give compounds 108a and 108b in 40-60% yield. LCMS and proton NMR was consistent with the structure.
Compounds 108a (60%) and 108b (40%) were treated to the same conditions as for compounds 100a-d (Example 47), to give compounds 109a and 109b in >80% yields. LCMS and proton NMR was consistent with the structure.
Compound 109a was treated to the same conditions as for compounds 101a-d (Example 47), to give Compound 110a in 30-60% yield. LCMS and proton NMR was consistent with the structure. Alternatively, Compound 110b can be prepared in a similar manner starting with Compound 109b.
Example 46: General Procedure for Conjugation with PFP Esters (Oligonucleotide 111); Preparation of ISIS 666881 (Ga1NAc3-10) A 5'-hexylamino modified oligonucleotide was synthesized and purified using standard solid-phase oligonucleotide procedures. The 5'-hexylamino modified oligonucleotide was dissolved in 0.1 M sodium tetraborate, pH 8.5 (200 [LI-) and 3 equivalents of a selected PFP esterified Ga1NAc3 cluster dissolved in DMSO (50 [LI-) was added. If the PFP ester precipitated upon addition to the ASO solution DMSO was added until all PFP ester was in solution. The reaction was complete after about 16 h of mixing at room temperature. The resulting solution was diluted with water to 12 mL and then spun down at 3000 rpm in a spin filter with a mass cut off of 3000 Da. This process was repeated twice to remove small molecule impurities. The solution was then lyophilized to dryness and redissolved in concentrated aqueous ammonia and mixed at room temperature for 2.5 h followed by concentration in vacuo to remove most of the ammonia.
The conjugated oligonucleotide was purified and desalted by RP-HPLC and lyophilized to provide the Ga1NAc3 conjugated oligonucleotide.

OH
HONC:H

3' 83e 11 AcHN 0 0 [ OLIGO j-O-P-0-(CH2)6-NH2 011-1 r OH
110a OH
HO NH 1 Borate buffer, DMSO, pH 8.5, rt AcHN NH
2 NH3 (aq), rt 0 0 OH OH
HO

AcHN
(OLIG0)¨(21\10-0 Oligonucleotide 111 is conjugated with Ga1NAc3-10. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-10 (Ga1NAc3-10a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)- as shown in the oligonucleotide (ISIS 666881) synthesized with Ga1NAc3-10 below. The structure of Ga1NAc3-10 (Ga1NAc3-10a-CM-) is shown below:
HO OH
HO
_.7.2..v0(--r"-N 0 "4 AcHN

AcHN
HO OH
-HN
AcHN
Following this general procedure ISIS 666881 was prepared. 5'-hexylamino modified oligonucleotide, ISIS 660254, was synthesized and purified using standard solid-phase oligonucleotide procedures. ISIS 660254 (40 mg, 5.2 [mot) was dissolved in 0.1 M sodium tetraborate, pH 8.5 (200 I.LL) and 3 equivalents PFP ester (Compound 110a) dissolved in DMSO (50 I.LL) was added.
The PFP ester precipitated upon addition to the ASO solution requiring additional DMSO (600 I.LL) to fully dissolve the PFP
ester. The reaction was complete after 16 h of mixing at room temperature. The solution was diluted with water to 12 mL total volume and spun down at 3000 rpm in a spin filter with a mass cut off of 3000 Da. This process was repeated twice to remove small molecule impurities. The solution was lyophilized to dryness and redissolved in concentrated aqueous ammonia with mixing at room temperature for 2.5 h followed by concentration in vacuo to remove most of the ammonia. The conjugated oligonucleotide was purified and desalted by RP-HPLC and lyophilized to give ISIS 666881 in 90% yield by weight (42 mg, 4.7 [mot).
Ga1NAc3-10 conjugated oligonucleotide SEQ
ASO Sequence (5' to 3') 5' group ID No.
NHACH2)6-0AdoGesmCesTesTesmCesAdsGdsTds ISIS 660254 Hexylamine 254 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GalNAc3-10.-0,AdoGesmCesTesTesmCesAdsGdsTds ISIS 666881 GalNAc3-10 254 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe Capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine.
Subscripts: "e" indicates a 2'-MOE modified nucleoside; "d" indicates a [3-D-2'-deoxyribonuc1eoside; "s"
indicates a phosphorothioate internucleoside linkage (PS); "o" indicates a phosphodiester internucleoside linkage (PO); and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.

Example 47: Preparation of Oligonucleotide 102 Comprising GaINAc3-8 HO¨
H2N- NHBoc BocHNN 0 n H
91a; n=1 HO...- NO2 91b, n=2 BocHNNH
n NO2 TFA, DCM
_________________________________________________________________________ )1-________________________________ ).-PFPTFA, DIPEA, DMF 0 HO 0 BocHN ,(:i/HN 0 /n 92a; n=1 92b, n=2 H2N1)nN-1(..._ /n H
¨I "--- OAc OAc H2N-- NH NO2 ; TMSOTf, DCM
Ac0-0Ac ______________________________________________________________ ).-0 AcHN 3 \ Z
H2N(HN 0 /n 93a; n=1 93b, n=2 94a; m=1 VAc 0OAc 94b, m=2 0 OAc OAc ,<,0,Bn Ac0 Ac0 i \
HO \ /m 0,1r.k7).i0H
__________________________________ .- AcHN m yO TMSOTf 7; m=1 Pd/C. H2 64, m=2 OAc AcONC:Ac 0 AcHN
OAc OAc 0 H
93a (93b) Ra-Ni, H2 _____________ .- NH ii.
HBTU, DIPEA, DMF Ac0 / m H NO2 _________ AcHN
OAc OAc 0 H
AcO_I_C__:)__\0(NNIN 0 AcHN 0 96a; n=1, m=1 96b; n=1, m=2 96c; n=2, m=1 96d: n=2. m=2 OAc AcONC:Ac 0 AcHN 0 m ric n n N HBTU, DIEA, DMF
OAc OAc _..:)..\c),)).L /ThNH ______________ Ac0 N
\ / m n ir\--- NH2 %
AcHN H
OAc OAc 0 r 0 ODMTr HO--/KIL
.:....:)..\.,, Ac0 1/4_/), 1/-17--iNN,,(1/HN"-. N.
AcHN
97a; n=1, m=1 97b; n=1, m=2 23 97c; n=2, m=1 97d; n=2, m=2 OAc AcONC:Ac 0 AcHN 0 OAcr- OAc 0 H
ODMTr Ac0--/:--)..\)Th)Ti (N' \-)n NH H 0 AcHN H Ir\--- N
OAc OAc N 0 r )7 Nb.,, H
.:...:,..\/, ,' \
Ac0 1/4-J jµ 1--LI,NN,µ,NzHN----0 0 OH
AcHN % 'no 98a; n=1, m=1 98b; n=1, m=2 98c; n=2, m=1 98d; n=2, m=2 OAc AcON(: 0 AcH -)Ac N C), [1:\ [1__(___ HBTU, DIEA, DMF k 7 n 0 97a, n=1, m=1 OAc OAc 97b, n=1, m=2 a Ac0- CY Nr'PhNH ...----0, 97c, n=2, m=1 0 -;----1,--) 97d, n=2, m=2 AcHN Bn HO2C0,Bn OAc OAc 0 r AcO-LEINN,(1F111-0 99 m AcHN n 100a, n=1, m=1 100b, n=1, m=2 100c, n=2, m=1 OAc 100d, n=2, m=2 AcON(:)Ac 0 AcHN 0N 0 Pd(OH)2/C, OAc OAc 0 . 0 0 H2, Et0Ac, PFPTFA, DMF, nripn1-1 , AcO.AC__\I
pyr _ AcHN
OAc OAc 0 r H
HN ---o 101a, n=1, m=1 AcHN m 0 n 101b, n=1, m=2 101c, n=2, m=1 101d, n=2, m=2 OAc Ac0(:)Ac 0 AcHN0 F F
OAc OAc n ir\----[1-----Co \ i m F
AcHN H
OAc OAc 0 r F
H
.........\0 /
Ac0 C)) 1N \,(11(Hr\l'o 102a, n=1, m=1 AcHN 102b, n=1, m=2 102c, n=2, m=1 102d, n=2, m=2 The triacid 90 (4 g, 14.43 mmol) was dissolved in DMF (120 mL) and N,N-Diisopropylethylamine (12.35 mL, 72 mmoles). Pentafluorophenyl trifluoroacetate (8.9 mL, 52 mmoles) was added dropwise, under argon, and the reaction was allowed to stir at room temperature for 30 minutes. Boc-diamine 91a or 91b (68.87 mmol) was added, along with N,N-Diisopropylethylamine (12.35 mL, 72 mmoles), and the reaction was allowed to stir at room temperature for 16 hours. At that time, the DMF
was reduced by >75% under reduced pressure, and then the mixture was dissolved in dichloromethane. The organic layer was washed with sodium bicarbonate, water and brine. The organic layer was then separated and dried over sodium sulfate, filtered and reduced to an oil under reduced pressure. The resultant oil was purified by silica gel chromatography (2%-->10% methanol/dichloromethane) to give compounds 92a and 92b in an approximate 80% yield. LCMS and proton NMR were consistent with the structure.
Compound 92a or 92b (6.7 mmoles) was treated with 20 mL of dichloromethane and 20 mL of trifluoroacetic acid at room temperature for 16 hours. The resultant solution was evaporated and then dissolved in methanol and treated with DOWEX-OH resin for 30 minutes. The resultant solution was filtered and reduced to an oil under reduced pressure to give 85-90% yield of compounds 93a and 93b.
Compounds 7 or 64 (9.6 mmoles) were treated with HBTU (3.7g, 9.6 mmoles) and N,N-Diisopropylethylamine (5 mL) in DMF (20 mL) for 15 minutes. To this was added either compounds 93a or 93b (3 mmoles), and allowed to stir at room temperature for 16 hours. At that time, the DMF was reduced by >75% under reduced pressure, and then the mixture was dissolved in dichloromethane. The organic layer was washed with sodium bicarbonate, water and brine. The organic layer was then separated and dried over sodium sulfate, filtered and reduced to an oil under reduced pressure. The resultant oil was purified by silica gel chromatography (5%-->20% methanol/dichloromethane) to give compounds 96a-d in 20-40% yield.
LCMS and proton NMR was consistent with the structure.
Compounds 96a-d (0.75 mmoles), individually, were hydrogenated over Raney Nickel for 3 hours in Ethanol (75 mL). At that time, the catalyst was removed by filtration thru celite, and the ethanol removed under reduced pressure to give compounds 97a-d in 80-90% yield. LCMS and proton NMR were consistent with the structure.
Compound 23 (0.32g, 0.53 mmoles) was treated with HBTU (0.2g, 0.53 mmoles) and N,N-Diisopropylethylamine (0.19 mL, 1.14 mmoles) in DMF (30mL) for 15 minutes. To this was added compounds 97a-d (0.38 mmoles), individually, and allowed to stir at room temperature for 16 hours. At that time, the DMF was reduced by >75% under reduced pressure, and then the mixture was dissolved in dichloromethane. The organic layer was washed with sodium bicarbonate, water and brine. The organic layer was then separated and dried over sodium sulfate, filtered and reduced to an oil under reduced pressure.
The resultant oil was purified by silica gel chromatography (2%-->20%
methanol/dichloromethane) to give compounds 98a-d in 30-40% yield. LCMS and proton NMR was consistent with the structure.
Compound 99 (0.17g, 0.76 mmoles) was treated with HBTU (0.29 g, 0.76 mmoles) and N,N-Diisopropylethylamine (0.35 mL, 2.0 mmoles) in DMF (50mL) for 15 minutes. To this was added compounds 97a-d (0.51 mmoles), individually, and allowed to stir at room temperature for 16 hours. At that time, the DMF was reduced by >75% under reduced pressure, and then the mixture was dissolved in dichloromethane. The organic layer was washed with sodium bicarbonate, water and brine. The organic layer was then separated and dried over sodium sulfate, filtered and reduced to an oil under reduced pressure.
The resultant oil was purified by silica gel chromatography (5%-->20%
methanol/ dichloromethane) to give compounds 100a-d in 40-60% yield. LCMS and proton NMR was consistent with the structure.
Compounds 100a-d (0.16 mmoles), individually, were hydrogenated over 10%
Pd(OH)2/C for 3 hours in methanoVethyl acetate (1:1, 50 mL). At that time, the catalyst was removed by filtration thru celite, and the organics removed under reduced pressure to give compounds 101a-d in 80-90% yield. LCMS and proton NMR was consistent with the structure.
Compounds 101a-d (0.15 mmoles), individually, were dissolved in DMF (15 mL) and pyridine (0.016 mL, 0.2 mmoles). Pentafluorophenyl trifluoroacetate (0.034 mL, 0.2 mmoles) was added dropwise, under argon, and the reaction was allowed to stir at room temperature for 30 minutes. At that time, the DMF
was reduced by >75% under reduced pressure, and then the mixture was dissolved in dichloromethane. The organic layer was washed with sodium bicarbonate, water and brine. The organic layer was then separated and dried over sodium sulfate, filtered and reduced to an oil under reduced pressure. The resultant oil was purified by silica gel chromatography (2%-->5% methanadichloromethane) to give compounds 102a-d in an approximate 80% yield. LCMS and proton NMR were consistent with the structure.
83e 3'5', 11 ( ou t Go O-P-0-(CH2)6NH2 I
OH
Borate buffer, DMSO, pH 8.5, rt 102d 2. aq. ammonia, rt HO--f'2-\---0hll'rhl)L-\
AcHN 0 0 HOOH 0 0 _ 0¨ CM ______________________________________________________________________ OLIGO
AcHN

AcHN
Oligomeric Compound 102, comprising a Ga1NAc3-8 conjugate group, was prepared using the general procedures illustrated in Example 46. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-8 (Ga1NAc3-8a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In a preferred embodiment, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-.
The structure of Ga1NAc3-8 (Ga1NAc3-8a-CM-) is shown below:

HO
Of AcHN 0 0 H 0 ,011) N t=-rN H H 4 AcHN

HO
_....7.2..\,01.1)'LN,I.N

AcHN =

Example 48: Preparation of Oligonucleotide 119 Comprising GaINAc3-7 AcO0Ac Ac0 OAc ONHCBz Ac0-....7C2\0 TMSOTf, DCE Ac0-.......\r Pd(OH)2/C
,f(-.NHCBz AcHN H2, Me0H, Et0Ac N"-------1 HO 3 4 1 35b 112 HO---in HBTU, DIEA
Ac0 OAc 0 0-i_ DMF
.4rONH2 + HO NHCBZ
Ac0 AcHN 0 y) 105a HO

Ac0 OAc H , Ac0-4,r N"--f2....1j Ac0 OAc AcHN
Ac0 Of-)N
0,4-NHCBZ

AcHN

Ac0 OAc Ac0 4rONH

AcHN

Ac0 OAc H , Ac0.....).,..\,0N----ts.;

AcHN
Ac0 OAc Pd/C, H2,2 0 AcHN 0 0 Ac0 OAc .......2...\õONH
Ac0 AcHN

Ac0 OAc H , Ac047 N------t;

HBTU, DIEA, DMF AcHN 0 0 Ac0 OAc _________________________ ).-Ac0 4 )..n....بNH
0 AcHN 0 0 HOO
Ac0 OAc Ac0 ........2...\õONH

83a AcHN

Compound 112 was synthesized following the procedure described in the literature (J. Med. Chem.
2004, 47, 5798-5808).
Compound 112 (5 g, 8.6 mmol) was dissolved in 1:1 methanol/ethyl acetate (22 mL/22 mL).
Palladium hydroxide on carbon (0.5 g) was added. The reaction mixture was stirred at room temperature under hydrogen for 12 h. The reaction mixture was filtered through a pad of celite and washed the pad with 1:1 methanol/ethyl acetate. The filtrate and the washings were combined and concentrated to dryness to yield Compound 105a (quantitative). The structure was confirmed by LCMS.
Compound 113 (1.25 g, 2.7 mmol), HBTU (3.2 g, 8.4 mmol) and DIEA (2.8 mL, 16.2 mmol) were dissolved in anhydrous DMF (17 mL) and the reaction mixture was stirred at room temperature for 5 min. To this a solution of Compound 105a (3.77 g, 8.4 mmol) in anhydrous DMF (20 mL) was added. The reaction was stirred at room temperature for 6 h. Solvent was removed under reduced pressure to get an oil. The residue was dissolved in CH2C12 (100 mL) and washed with aqueous saturated NaHCO3 solution (100 mL) and brine (100 mL). The organic phase was separated, dried (Na2SO4), filtered and evaporated. The residue was purified by silica gel column chromatography and eluted with 10 to 20 %
Me0H in dichloromethane to yield Compound 114 (1.45 g, 30%). The structure was confirmed by LCMS and 1H
NMR analysis.
Compound 114 (1.43 g, 0.8 mmol) was dissolved in 1:1 methanol/ethyl acetate (4 mL/4 mL).
Palladium on carbon (wet, 0.14 g) was added. The reaction mixture was flushed with hydrogen and stirred at room temperature under hydrogen for 12 h. The reaction mixture was filtered through a pad of celite. The celite pad was washed with methanol/ethyl acetate (1:1). The filtrate and the washings were combined together and evaporated under reduced pressure to yield Compound 115 (quantitative). The structure was confirmed by LCMS and 1H NMR analysis.
Compound 83a (0.17 g, 0.75 mmol), HBTU (0.31 g, 0.83 mmol) and DIEA (0.26 mL, 1.5 mmol) were dissolved in anhydrous DMF (5 mL) and the reaction mixture was stirred at room temperature for 5 min. To this a solution of Compound 115 (1.22 g, 0.75 mmol) in anhydrous DMF
was added and the reaction was stirred at room temperature for 6 h. The solvent was removed under reduced pressure and the residue was dissolved in CH2C12. The organic layer was washed aqueous saturated NaHCO3 solution and brine and dried over anhydrous Na2SO4 and filtered. The organic layer was concentrated to dryness and the residue obtained was purified by silica gel column chromatography and eluted with 3 to 15 % Me0H in dichloromethane to yield Compound 116 (0.84 g, 61%). The structure was confirmed by LC MS and 1H
NMR analysis.
Ac0 OAc H , Ac0Ø24,C)N -----t....\' AcHN
Pd/C, H2, Ac0 OAc 0 0 Et0Ac, Me0H
.....?....\,0(--)NH
,...

AcHN 0 0 Ac0 OAc ......2..\,ONH
Ac0 AcHN
Ac0 OAc F
F

AcHN
PFPTFA, DMF, Pyr Ac0 OAc )LLc) 0 0 ........2...\,ONH
Ac0 F
F
4 )..nõ..Ø.....¨NH
AcHN 0 0 Ac0 OAc ") .......2...\,,ONH 118 Ac0 AcHN

Compound 116 (0.74 g, 0.4 mmol) was dissolved in 1:1 methanol/ethyl acetate (5 mL/5 mL).
Palladium on carbon (wet, 0.074 g) was added. The reaction mixture was flushed with hydrogen and stirred at room temperature under hydrogen for 12 h. The reaction mixture was filtered through a pad of celite. The celite pad was washed with methanol/ethyl acetate (1:1). The filtrate and the washings were combined together and evaporated under reduced pressure to yield compound 117 (0.73 g, 98%). The structure was confirmed by LCMS and 1H NMR analysis.
Compound 117 (0.63 g, 0.36 mmol) was dissolved in anhydrous DMF (3 mL). To this solution N,N-Diisopropylethylamine (70 [LL, 0.4 mmol) and pentafluorophenyl trifluoroacetate (72 [tt, 0.42 mmol) were added. The reaction mixture was stirred at room temperature for 12 h and poured into a aqueous saturated NaHCO3 solution. The mixture was extracted with dichloromethane, washed with brine and dried over anhydrous Na2SO4. The dichloromethane solution was concentrated to dryness and purified with silica gel column chromatography and eluted with 5 to 10 % Me0H in dichloromethane to yield compound 118 (0.51 g, 79%). The structure was confirmed by LCMS and 1H and 1H and 19F NMR.
83e 3'5'1 11 j [ OLIGO ¨0¨P-0¨(CH2)6-NH2 OH
1. Borate buffer, DMSO, pH 8.5, rt 118 ____ vi __________________ 2. aq. ammonia, rt AcHN N

OLIGO

AcHN 0/
HO OH

AcHN
Oligomeric Compound 119, comprising a Ga1NAc3-7 conjugate group, was prepared using the general procedures illustrated in Example 46. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-7 (Ga1NAc3-7a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-.
The structure of Ga1NAc3-7 (Ga1NAc3-7a-CM-) is shown below:

HOOH
AcHN No 4 1-1)---C31-1-IN''IL(HN
AcHN OZ
HOOH

AcHN =
Example 49: Preparation of Oligonucleotide 132 Comprising Ga1NAc3-5 HNBoc HN,Boc Boc HN,Boc HN, H2NC) Boo, E)-L
N N1-1 o Boo,NN)-L,OH

Boo, N 0 0 HBTU, TEA
Li0H, H20 ___________________________________________________________ 10' ,Boo Me0H, THF
DMF HN HN,Boc 78% 123 Compound 120 (14.01 g, 40 mmol) and HBTU (14.06 g, 37 mmol) were dissolved in anhydrous DMF (80 mL). Triethylamine (11.2 mL, 80.35 mmol) was added and stirred for 5 min. The reaction mixture was cooled in an ice bath and a solution of compound 121 (10 g, mmol) in anhydrous DMF (20 mL) was added. Additional triethylamine (4.5 mL, 32.28 mmol) was added and the reaction mixture was stirred for 18 h under an argon atmosphere. The reaction was monitored by TLC (ethyl acetate:hexane; 1:1; Rf = 0.47).
The solvent was removed under reduced pressure. The residue was taken up in Et0Ac (300 mL) and washed with 1M NaHSO4 ( 3 x 150 mL), aqueous saturated NaHCO3 solution (3 x 150 mL) and brine (2 x 100 mL).
Organic layer was dried with Na2SO4. Drying agent was removed by filtration and organic layer was concentrated by rotary evaporation. Crude mixture was purified by silica gel column chromatography and eluted by using 35 ¨ 50% Et0Ac in hexane to yield a compound 122 (15.50 g, 78.13%). The structure was confirmed by LCMS and 1H NMR analysis. Mass m/z 589.3 [M + H] .
A solution of LiOH (92.15 mmol) in water (20 mL) and THF (10 mL) was added to a cooled solution of Compound 122 (7.75 g,13.16 mmol) dissolved in methanol (15 mL). The reaction mixture was stirred at room temperature for 45 min. and monitored by TLC (Et0Ac:hexane; 1:1). The reaction mixture was concentrated to half the volume under reduced pressure. The remaining solution was cooled an ice bath and neutralized by adding concentrated HC1. The reaction mixture was diluted, extracted with Et0Ac (120 mL) and washed with brine (100 mL). An emulsion formed and cleared upon standing overnight. The organic layer was separated dried (Na2SO4), filtered and evaporated to yield Compound 123 (8.42 g). Residual salt is the likely cause of excess mass. LCMS is consistent with structure. Product was used without any further purification. M.W.cal:574.36; M.W.fd:575.3 [M + H] .

. ii 0 S¨OH = H20 H 3 N --...f.,,,,,-"-----,-1, H2N-L,OH + HO 401 0 ' 0 0 0 0 0¨g II
Toluene, Reflux li 996%
Compound 126 was synthesized following the procedure described in the literature (J. Am. Chem.
Soc. 2011, /33, 958-963).

HN,Boc 126 BocN
...."...p,/\,,e, 0 CF3COOH
123 Vo- Thr 0 _____________________________________________________________________ OP-H H "3 8 CH2Cl2 HOBt, DIEA, 0 PyBop, Bop, DMF
r HN,Boc 127 CF3C00- I\TH3 Ac0 OAc H AcO0r Nj=L 0 0 H3Nr N AcHN 7 0 CF3C00- 0 ________________________________________________ - 7..-HATU, HOAt, DIEA, DMF
r cF3coo- 0 NH3 128 Ac0 OAc Ac0 N.,.....---..Nr0 AcHN
NH

Ac0 OAc i<)L w3-(c) I.
HN-----y N
H
Ac0 Oz--i 0 0 AcHN 0 /
Ac0 OAc Ac0--4r, H
-\,-AcHN 0 Ac0 OAc ( 0 Ac0----tor.s\v 0 AcHN
NH

Pd/C, H2, Me0H
129 _______________________ )...-Ac0 OAc INI j-L
HN NC)F
Ac0µ.......õ......õ¨........õ. 0 AcHN 0 /
Ac0 OAc 2..\/-, NH
Ac0 Ac0 OAc AcHN 0 130 ----C ______________________ 0 Ac0 ______________________ .7......\v 0 AcHN
NH
PFPTFA, DMF, Pyr F
HN NC) F
AcHN 0 / F
Ac0 OAc F
(2.\;-) NH
Ac0 AcHN 0 Compound 123 (7.419 g, 12.91 mmol), HOBt (3.49 g, 25.82 mmol) and compound 126 (6.33 g,
16.14 mmol) were dissolved in and DMF (40 mL) and the resulting reaction mixture was cooled in an ice bath. To this N,N-Diisopropylethylamine (4.42 mL, 25.82 mmol), PyBop (8.7 g, 16.7 mmol) followed by Bop coupling reagent (1.17 g, 2.66 mmol) were added under an argon atmosphere.
The ice bath was removed and the solution was allowed to warm to room temperature. The reaction was completed after 1 h as determined by TLC (DCM:MeOH:AA; 89:10:1). The reaction mixture was concentrated under reduced pressure. The residue was dissolved in Et0Ac (200 mL) and washed with 1 M
NaHSO4 (3x100 mL), aqueous saturated NaHCO3 (3x100 mL) and brine (2x100 mL). The organic phase separated dried (Na2SO4), filtered and concentrated. The residue was purified by silica gel column chromatography with a gradient of 50% hexanes/EtOAC to 100% Et0Ac to yield Compound 127 (9.4 g) as a white foam.
LCMS and 1H NMR

were consistent with structure. Mass m/z 778.4 [M + H] +.
Trifluoroacetic acid (12 mL) was added to a solution of compound 127 (1.57 g, 2.02 mmol) in dichloromethane (12 mL) and stirred at room temperature for 1 h. The reaction mixture was co-evaporated with toluene (30 mL) under reduced pressure to dryness. The residue obtained was co-evaporated twice with acetonitrile (30 mL) and toluene (40 mL) to yield Compound 128 (1.67 g) as trifluoro acetate salt and used for next step without further purification. LCMS and 1H NMR were consistent with structure. Mass m/z 478.2 [M + H] +.
Compound 7 (0.43 g, 0.963 mmol), HATU (0.35 g, 0.91 mmol), and HOAt (0.035 g, 0.26 mmol) were combined together and dried for 4 h over P205 under reduced pressure in a round bottom flask and then dissolved in anhydrous DMF (1 mL) and stirred for 5 min. To this a solution of compound 128 (0.20 g, 0.26 mmol) in anhydrous DMF (0.2 mL) and N,N-Diisopropylethylamine (0.2 mL) was added. The reaction mixture was stirred at room temperature under an argon atmosphere. The reaction was complete after 30 min as determined by LCMS and TLC (7% Me0H/DCM). The reaction mixture was concentrated under reduced pressure. The residue was dissolved in DCM (30 mL) and washed with 1 M NaHSO4 (3x20 mL), aqueous saturated NaHCO3 (3 x 20 mL) and brine (3x20 mL). The organic phase was separated, dried over Na2504, filtered and concentrated. The residue was purified by silica gel column chromatography using 5-15%
Me0H in dichloromethane to yield Compound 129 (96.6 mg). LC MS and 1H NMR are consistent with structure. Mass m/z 883.4 [M + 2H] .
Compound 129 (0.09 g, 0.051 mmol) was dissolved in methanol (5 mL) in 20 mL
scintillation vial.
To this was added a small amount of 10% Pd/C (0.015 mg) and the reaction vessel was flushed with H2 gas.
The reaction mixture was stirred at room temperature under H2 atmosphere for 18 h. The reaction mixture was filtered through a pad of Celite and the Celite pad was washed with methanol. The filtrate washings were pooled together and concentrated under reduced pressure to yield Compound 130 (0.08 g). LCMS and 1H NMR were consistent with structure. The product was used without further purification. Mass m/z 838.3 [M + 2H] .
To a 10 mL pointed round bottom flask were added compound 130 (75.8 mg, 0.046 mmol), 0.37 M
pyridine/DMF (200 [LI-) and a stir bar. To this solution was added 0.7 M
pentafluorophenyl trifluoroacetate/DMF (100 [LL) drop wise with stirring. The reaction was completed after 1 h as determined by LC MS. The solvent was removed under reduced pressure and the residue was dissolved in CHC13 (¨ 10 mL). The organic layer was partitioned against NaHSO4 (1 M, 10 mL) , aqueous saturated NaHCO3 (10 mL) and brine (10 mL) three times each. The organic phase separated and dried over Na2504, filtered and concentrated to yield Compound 131 (77.7 mg). LCMS is consistent with structure. Used without further purification. Mass m/z 921.3 [M + 2H] .

HO OH
3' 5'1 II AcHN
( OLIGO r0-P-0-(CH2)6-NH2 NH
I
OH
1. Borate buffer, DMSO, pH 8.5, rt 131 ______________________ ).-2. aq. ammonia, rt HO OH N ).L,NH
HN-Thr HO Oz=----. 0 ..., AcHN 0 /
HO OH
NH

AcHN 0 Oligomeric Compound 132, comprising a Ga1NAc3-5 conjugate group, was prepared using the general procedures illustrated in Example 46. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-5 (Ga1NAc3-5a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(01-1)-Ad-P(=0)(OH)-.
The structure of Ga1NAc3-5 (Ga1NAc3-5a-CM-) is shown below:
HO OH
HO Or0 AcHN
NH

HO OH
HN'ThzNANH

HO
AcHN 0 /
HO OH
0 I\1("r0¨(CMH

AcHN 0 H
=

Example 50: Preparation of Oligonucleotide 144 Comprising Ga1NAc4-11 DMTO Fmoc 1. TBTU, DIEA DMTO Fmoc La ACN, VIMAD Resin Lol pip:DBU:DMF
________________________________ 70 0,--. 0 2. Ac 20 Capping . 0 0 (2:2:96) 3 __________ -I-4 -OH Kaiser: Negetive b10-0 HN-Fmoc DMTO 6 Fmoc,NOH
L. 11 H /
0 DMTN....

L. al b HBTU, DIEA, DMF
. 0 0 135 b NH-Fmoc DMTr ) ib 1. pip:DBU:DMF 0 / H 0 1. 2% hydrazine/DMF
Kaiser: Positive ....iN)L(CH2)5' N y'Nfj Kaiser: Positive ________________ ) ________________________________________________________ ).--2. Dde-Lys(Fmoc)-OH (138) 0 H 2. Fmoc-Lys(Fmoc)-OH
(140) HATU, DIEA, DMF d 0 HATU, DIEA, DMF
Kaiser: Negative 0 Kaiser: Negative ,Fmoc HN
) /
HNOniFmoc DMTr O
O

(cH
N --11--2)5.- -11-----"---Fmoc ....1 Y'hi d HN,Fmoc Ac0 OAc AcO___&Z,0 AcHN 41W, Mr' Ac0 OAc Ac0 N o AcHN
,0 1. pip:DBU:DMF 0 H
0 1.(1\1 Kaiser: Positive HN?
_______________________ 70-2. 7, HATU, DIEA, Ac0 OAc 0 DMF DMTO
Kaiser: Negative Ac0 0 NH
AcHN 7/0 Ac0 OAc Ac0 )=/--NH
AcHN

Synthesis of Compound 134. To a Merrifield flask was added aminomethyl VIMAD
resin (2.5 g, 450 [tmol/g) that was washed with acetonitrile, dimethylformamide, dichloromethane and acetonitrile. The resin was swelled in acetonitrile (4 mL). Compound 133 was pre-activated in a 100 mL round bottom flask by adding 20 (1.0 mmol, 0.747 g), TBTU (1.0 mmol, 0.321 g), acetonitrile (5 mL) and DIEA (3.0 mmol, 0.5 mL). This solution was allowed to stir for 5 min and was then added to the Merrifield flask with shaking.
The suspension was allowed to shake for 3 h. The reaction mixture was drained and the resin was washed with acetonitrile, DMF and DCM. New resin loading was quantitated by measuring the absorbance of the DMT cation at 500 nm (extinction coefficient = 76000) in DCM and determined to be 238 [tmol/g. The resin was capped by suspending in an acetic anhydride solution for ten minutes three times.
The solid support bound compound 141 was synthesized using iterative Fmoc-based solid phase peptide synthesis methods. A small amount of solid support was withdrawn and suspended in aqueous ammonia (28-30 wt%) for 6 h. The cleaved compound was analyzed by LC-MS and the observed mass was consistent with structure. Mass m/z 1063.8 [M + 2I-1] .
The solid support bound compound 142 was synthesized using solid phase peptide synthesis methods.

Ac0 OAc AcO___&Z,0 AcHN -NH

Ac0 OAc (400-Ac0 N Ni AcHN 0 H p DNA syntesizer 0 H _Ny 142 _______________ ).--Ac0 OAc 0 __&...2.\,0 H NH I
Ac0 \ , __ , ( CM J ___________________________________________________________________ ASO
AcHN . -Ac0 OAc Ac0 )i---NH
AcHN 0 HO OH

/----NH
AcHN 0 HO OH
HO NI
AcHN
aqueous NH3 0 H l(F\J------Af\-)-3¨)7_Ny ___________________ 1.-HO
[CM) )T¨NNH __________________________________________________________ , , ____ , _______________________________________________________________________________ ASO
AcHN -HO OH 0 o AcHN 0 The solid support bound compound 143 was synthesized using standard solid phase synthesis on a DNA synthesizer.
The solid support bound compound 143 was suspended in aqueous ammonia (28-30 wt%) and heated at 55 C for 16 h. The solution was cooled and the solid support was filtered.
The filtrate was concentrated and the residue dissolved in water and purified by HPLC on a strong anion exchange column. The fractions containing full length compound 144 were pooled together and desalted. The resulting Ga1NAc4-11 conjugated oligomeric compound was analyzed by LC-MS and the observed mass was consistent with structure.
The Ga1NAc4 cluster portion of the conjugate group Ga1NAc4-11 (Ga1NAc4-11a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-.
The structure of GalNAc4-11 (Ga1NAc4-11a-CM) is shown below:
HO OH
AcHN

HO OH
HO
AcHN 0 H

NH
HO
AcHN 0 0 HO OH
HO
AcHN

Example 51: Preparation of Oligonucleotide 155 Comprising GaINAc3-6 OH

S) OyN.NH2 0 OH _______________________________________________ = 0 NI Nj-LOH

2M NaOH 0 Compound 146 was synthesized as described in the literature (Analytical Biochemistry 1995, 229, 54-60).

HO ..õ....-...õ....-.....--., 1 HI \ I Ao 0 Ac0 OAc 35b ____&; 0 ...Ø., 4 ____________________________________ )1,- Ac0 N Ä0 401 TMS-0Tf, 4 A molecular sieves, CH2Cl2, it H
AcHN

Ac0 OAc 0 ,c)1,)-LOH
II
H2, Pd(OH)2 /C ____.......C..:)..\, 0 147 Et0Ac/Me0H AcHN 105a HBTU, DIEA, DMF, rt Ac0 OAc 2 0 H H2, Pd(OH)2 /C, Et0Ac/Me0H
N}..,...N
AcHN H

Ac0 OAc Ac00-----./N_NH2 AcHN H

Compound 4 (15 g, 45.55 mmol) and compound 35b (14.3 grams, 57 mmol) were dissolved in CH2C12 (200 m1). Activated molecular sieves (4 A. 2 g, powdered) were added, and the reaction was allowed to stir for 30 minutes under nitrogen atmosphere. TMS-0Tf was added (4.1 ml, 22.77 mmol) and the reaction was allowed to stir at room temp overnight. Upon completion, the reaction was quenched by pouring into solution of saturated aqueous NaHCO3 (500 ml) and crushed ice (¨
150 g). The organic layer was separated, washed with brine, dried over MgSO4, filtered, and was concentrated to an orange oil under reduced pressure. The crude material was purified by silica gel column chromatography and eluted with 2-10 % Me0H in CH2C12to yield Compound 112 (16.53 g, 63 %). LCMS and 1H NMR were consistent with the expected compound.
Compound 112 (4.27 g, 7.35 mmol) was dissolved in 1:1 Me0H/Et0Ac (40 m1). The reaction mixture was purged by bubbling a stream of argon through the solution for 15 minutes. Pearlman's catalyst (palladium hydroxide on carbon, 400 mg) was added, and hydrogen gas was bubbled through the solution for 30 minutes. Upon completion (TLC 10% Me0H in CH2C12, and LCMS), the catalyst was removed by filtration through a pad of celite. The filtrate was concentrated by rotary evaporation, and was dried briefly under high vacuum to yield Compound 105a (3.28 g). LCMS and 1H NMR were consistent with desired product.
Compound 147 (2.31 g, 11 mmol) was dissolved in anhydrous DMF (100 mL). N,N-Diisopropylethylamine (DIEA, 3.9 mL, 22 mmol) was added, followed by HBTU (4 g, 10.5 mmol). The reaction mixture was allowed to stir for ¨ 15 minutes under nitrogen. To this a solution of compound 105a (3.3 g, 7.4 mmol) in dry DMF was added and stirred for 2 h under nitrogen atmosphere. The reaction was diluted with Et0Ac and washed with saturated aqueous NaHCO3 and brine. The organics phase was separated, dried (MgSO4), filtered, and concentrated to an orange syrup. The crude material was purified by column chromatography 2-5 % Me0H in CH2C12 to yield Compound 148 (3.44 g, 73 %). LCMS and 1H
NMR were consistent with the expected product.
Compound 148 (3.3 g, 5.2 mmol) was dissolved in 1:1 Me0H/Et0Ac (75 m1). The reaction mixture was purged by bubbling a stream of argon through the solution for 15 minutes.
Pearlman's catalyst (palladium hydroxide on carbon) was added (350 mg). Hydrogen gas was bubbled through the solution for 30 minutes. Upon completion (TLC 10% Me0H in DCM, and LCMS), the catalyst was removed by filtration through a pad of celite. The filtrate was concentrated by rotary evaporation, and was dried briefly under high vacuum to yield Compound 149 (2.6 g). LCMS was consistent with desired product. The residue was dissolved in dry DMF (10 ml) was used immediately in the next step.
Ac0 OAc Ac0 OAcAcOoN)1 0 0 AcHN 3 H

AcO,oN)1----../N-1( )1-----N H
AcHN 3 H 0 146 ________________ )...- Ac0 OAc 0 HBTU, DIEA, DMF )i.........NH
...C.......\:) zo.........7¨...1..õ1----, N
Ac0 3 H
NHAc Ac0 OAc Ac0 011OAcAcOoN1 Pd(OH)2/C, H2 AcHN 3 H
Me0H, Et0Ac AcHN 3 H 0 AcO\ OAc 0 AcO 0...........74----.., )1------/NH
N
="\"'"-\/ 3 H
NHAc Compound 146 (0.68 g, 1.73 mmol) was dissolved in dry DMF (20 m1). To this DIEA (450 [tt, 2.6 mmol, 1.5 eq.) and HBTU (1.96 g, 0.5.2 mmol) were added. The reaction mixture was allowed to stir for 15 minutes at room temperature under nitrogen. A solution of compound 149 (2.6 g) in anhydrous DMF (10 mL) was added. The pH of the reaction was adjusted to pH = 9-10 by addition of DIEA (if necessary). The reaction was allowed to stir at room temperature under nitrogen for 2 h. Upon completion the reaction was diluted with Et0Ac (100 mL), and washed with aqueous saturated aqueous NaHCO3, followed by brine. The organic phase was separated, dried over MgSO4, filtered, and concentrated. The residue was purified by silica gel column chromatography and eluted with 2-10 % Me0H in CH2C12to yield Compound 150 (0.62 g, 20 %). LCMS and 1H NMR were consistent with the desired product.
Compound 150 (0.62 g) was dissolved in 1:1 Me0H/ Et0Ac (5 L). The reaction mixture was purged by bubbling a stream of argon through the solution for 15 minutes. Pearlman's catalyst (palladium hydroxide on carbon) was added (60 mg). Hydrogen gas was bubbled through the solution for 30 minutes. Upon completion (TLC 10% Me0H in DCM, and LCMS), the catalyst was removed by filtration (syringe-tip Teflon filter, 0.45 [Lin). The filtrate was concentrated by rotary evaporation, and was dried briefly under high vacuum to yield Compound 151 (0.57 g). The LCMS was consistent with the desired product. The product was dissolved in 4 mL dry DMF and was used immediately in the next step.

Ac0 OAc Ac0---4/ ) Ac0 OAcNN \r0 0 0 AcHN 3 H 0 0 0 H )) Bn0)01-1 Ac0--4r(DN)N-----inN N
3 H OBn 83a 0 151 _____________ 7 AcHN 3 H
----,-..---0 PFP-TFA, DIEA, DMF
Ac0 OAc 0 N NH
Ac0 3 H
NHAc Ac0 OAc Ac0--4.-\/ N)N
Ac OAc AcHN 3 H
_....(...)..\r0 Pd(OH)2/C, H2 0 ______________ v.- Ac0 Me0H, Et0Ac AcHN 3 H 0 ----1-.---0 Ac0 OAc 0 4/0.,.....,..õ--._ N)./NH
Ac0 NHAc Ac0 OAc ___...1- ..2..\
Ac0 )1....._____O Ed F
\(,,rN
.0 F
Ac_4r0 OAc AcHN 3 H 0 0 PFP-TFA, Dl EA 0 ..,., N }....,... N\ r N "c) v.- Ac0 3 H F
DMF AcHN 3 H 0 ------,-:---0 F
Ac0 OAc 0 )1______NH
N
Ac0 NHAc Compound 83a (0.11 g, 0.33 mmol) was dissolved in anhydrous DMF (5 mL) and N,N-Diisopropylethylamine (75 I.LL, 1 mmol) and PFP-TFA (90 I.LL, 0.76 mmol) were added. The reaction mixture turned magenta upon contact, and gradually turned orange over the next 30 minutes. Progress of reaction was monitored by TLC and LCMS. Upon completion (formation of the PFP
ester), a solution of compound 151 (0.57 g, 0.33 mmol) in DMF was added. The pH of the reaction was adjusted to pH = 9-10 by addition of N,N-Diisopropylethylamine (if necessary). The reaction mixture was stirred under nitrogen for ¨
30 min. Upon completion, the majority of the solvent was removed under reduced pressure. The residue was diluted with CH2C12 and washed with aqueous saturated NaHCO3, followed by brine. The organic phase separated, dried over MgSO4, filtered, and concentrated to an orange syrup.
The residue was purified by silica gel column chromatography (2-10 % Me0H in CH2C12) to yield Compound 152 (0.35 g, 55 %). LCMS
and 1H NMR were consistent with the desired product.
Compound 152 (0.35 g, 0.182 mmol) was dissolved in 1:1 Me0H/Et0Ac (10 mL). The reaction mixture was purged by bubbling a stream of argon thru the solution for 15 minutes. Pearlman's catalyst (palladium hydroxide on carbon) was added (35 mg). Hydrogen gas was bubbled thru the solution for 30 minutes. Upon completion (TLC 10% Me0H in DCM, and LCMS), the catalyst was removed by filtration (syringe-tip Teflon filter, 0.45 [tin). The filtrate was concentrated by rotary evaporation, and was dried briefly under high vacuum to yield Compound 153 (0.33 g, quantitative). The LCMS was consistent with desired product.
Compound 153 (0.33 g, 0.18 mmol) was dissolved in anhydrous DMF (5 mL) with stirring under nitrogen. To this N,N-Diisopropylethylamine (65 [tt, 0.37 mmol) and PFP-TFA
(35 [tt, 0.28 mmol) were added. The reaction mixture was stirred under nitrogen for ¨ 30 min. The reaction mixture turned magenta upon contact, and gradually turned orange. The pH of the reaction mixture was maintained at pH = 9-10 by adding more N,-Diisopropylethylamine. The progress of the reaction was monitored by TLC and LCMS.
Upon completion, the majority of the solvent was removed under reduced pressure. The residue was diluted with CH2C12 (50 mL), and washed with saturated aqueous NaHCO3, followed by brine. The organic layer was dried over Mg504, filtered, and concentrated to an orange syrup. The residue was purified by column chromatography and eluted with 2-10 % Me0H in CH2C12 to yield Compound 154 (0.29 g, 79 %). LCMS
and 1H NMR were consistent with the desired product.
83e 3 6, I I HOOH o [ OLIGO J-O-P1-0-(CH2)6 NH2 __...7.2...\....._0õ..-.44,-.., NA, 0 H HO "4 H
AcHN HN
\OH

1 Borate buffer, DMSO, HOOH

H
pH 8.5, rt ' 2 aq AcHN
ammonia, rt 0 0 0 NI'µ

AcHN
Oligomeric Compound 155, comprising a Ga1NAc3-6 conjugate group, was prepared using the general procedures illustrated in Example 46. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-6 (Ga1NAc3-6a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-.
The structure of Ga1NAc3-6 (Ga1NAc3-6a-CM-) is shown below:

HoOH 0 HO --.1*--\-----01")N

AcHN HN

H H
HO--1"--\---Of'rN)'NN(õ).NrN.H,5-0 ml AcHN
ri---µ
HooH - 0 AcHN .
Example 52: Preparation of Oligonucleotide 160 Comprising GaINAc3-9 AcO0Ac 0 Ac0 rc HOvj'-'--0 , 50 C Ac0 10 IP
Ac0-. TMSOTf, .....Z_OAc _______________________________________________________________ AcHN CICH2CH2CI, rt, 93% N-------___----_ TMSOTf, DCE, 66%

Ac0 OAc Ac0 OAc H2, Pd/C 0 (Th AC0-1(....)..\7 'Ho 4 _______________________ Me0H, 95% \ 10 AcHN 0 AcHN 0 OH
., Ac0 OAc HBTU, DMF, EtN(iP02 .....::..:)..\.,Ac0 0 71\ Phosphitylation s' 4,¨IR
DMTO "10 81%
AcHN 0 ODMT
--b1H

Hd 47 NC

/
p¨P
Ac0 OAc NUP02 .....Z\vo :IR
Ac0 'e/r7 '10 AcHN 0 ODMT

Compound 156 was synthesized following the procedure described in the literature (J. Med. Chem.
5 2004, 47, 5798-5808).
Compound 156, (18.60 g, 29.28 mmol) was dissolved in methanol (200 mL).
Palladium on carbon (6.15 g, 10 wt%, loading (dry basis), matrix carbon powder, wet) was added.
The reaction mixture was stirred at room temperature under hydrogen for 18 h. The reaction mixture was filtered through a pad of celite and the celite pad was washed thoroughly with methanol. The combined filtrate was washed and concentrated to dryness. The residue was purified by silica gel column chromatography and eluted with 5-10 % methanol in dichloromethane to yield Compound 157 (14.26 g, 89%). Mass m/z 544.1 [M-H].
Compound 157 (5 g, 9.17 mmol) was dissolved in anhydrous DMF (30 mL). HBTU
(3.65 g, 9.61 mmol) and N,N-Diisopropylethylamine (13.73 mL, 78.81 mmol) were added and the reaction mixture was stirred at room temperature for 5 minutes. To this a solution of compound 47 (2.96 g, 7.04 mmol) was added.
The reaction was stirred at room temperature for 8 h. The reaction mixture was poured into a saturated NaHCO3 aqueous solution. The mixture was extracted with ethyl acetate and the organic layer was washed with brine and dried (Na2SO4), filtered and evaporated. The residue obtained was purified by silica gel column chromatography and eluted with 50% ethyl acetate in hexane to yield compound 158 (8.25g, 73.3%).
The structure was confirmed by MS and 1H NMR analysis.
Compound 158 (7.2 g, 7.61 mmol) was dried over P205 under reduced pressure.
The dried compound was dissolved in anhydrous DMF (50 mL). To this 1H-tetrazole (0.43 g, 6.09 mmol) and N-methylimidazole (0.3 mL, 3.81 mmol) and 2-cyanoethyl-N,N,/VV\P-tetraisopropyl phosphorodiamidite (3.65 mL, 11.50 mmol) were added. The reaction mixture was stirred t under an argon atmosphere for 4 h. The reaction mixture was diluted with ethyl acetate (200 mL). The reaction mixture was washed with saturated NaHCO3 and brine. The organic phase was separated, dried (Na2SO4), filtered and evaporated. The residue was purified by silica gel column chromatography and eluted with 50-90 % ethyl acetate in hexane to yield Compound 159 (7.82 g, 80.5%). The structure was confirmed by LCMS and 31P NMR
analysis.
pH
HOOH
HO 9Nr AcHN
0=P¨OH
1. DNA synthesizer HOOH
159 _______________ 2. aq. NH4OH 0 0 AcHN
0=P¨OH
HOOH
HOO NZ .
0 _____________________________________________________ .1[74.) AcHN

Oligomeric Compound 160, comprising a Ga1NAc3-9 conjugate group, was prepared using standard oligonucleotide synthesis procedures. Three units of compound 159 were coupled to the solid support, followed by nucleotide phosphoramidites. Treatment of the protected oligomeric compound with aqueous ammonia yielded compound 160. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-9 (Ga1NAc3-9a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-. The structure of Ga1NAc3-9 (Ga1NAc3-9a-CM) is shown below:
pH
HOOH
HO

AcHN 1 0=P¨OH

HoOH
HO0orNIR...0 AcHN 1 0=P¨OH

HO OH
HO
AcHN
=

Example 53: Alternate procedure for preparation of Compound 18 (Ga1NAc3-la and Ga1NAc3-3a) Ao H2NNHR H TMSOTf HONNHR ____________________________________________________________ ,..
\) R = H or Cbz 0:g OAc 0 7........\
161 I¨ R = H 162a 0 CbzCI, Et3N 1, R . L
CDZ , 162b Ac0 N
4 "_-0 PFPO
OAc )r-Th H
0 ..õ.7 ¨limo-Ac0 0,,NNHR + PFP0,1r-,r 0 NHCBZ
NHAc 0 0 0 or Cbz, 163a K) Pd/C R = Cb , H2 I¨ PFPO
1¨,- R = H, 163b OAc OAc O

0 0, Ac0 H
, NHAc 12-INN
OAc 0.pg_v____ 0 0 0 ___________________ H H NHCBZ
Ac0 N NI.r---....r -----../
1"").-1 ,r NHAc 0 (:\) IL' OAc OAc HN.r.........õõr=-õN/"----7 Ac0 NHAc Lactone 161 was reacted with diamino propane (3-5 eq) or Mono-Boc protected diamino propane (1 eq) to provide alcohol 162a or 162b. When unprotected propanediamine was used for the above reaction, the excess diamine was removed by evaporation under high vacuum and the free amino group in 162a was protected using CbzCl to provide 162b as a white solid after purification by column chromatography.
Alcohol 162b was further reacted with compound 4 in the presence of TMSOTf to provide 163a which was converted to 163b by removal of the Cbz group using catalytic hydrogenation.
The pentafluorophenyl (PFP) ester 164 was prepared by reacting triacid 113 (see Example 48) with PFPTFA
(3.5 eq) and pyridine (3.5 eq) in DMF (0.1 to 0.5 M). The triester 164 was directly reacted with the amine 163b (3-4 eq) and DIPEA (3-4 eq) to provide Compound 18. The above method greatly facilitates purification of intermediates and minimizes the formation of byproducts which are formed using the procedure described in Example 4.

Example 54: Alternate procedure for preparation of Compound 18 (Ga1NAc3-la and Ga1NAc3-3a) HO2C7Th PFPTFA PFP0 0, 0, DMF, pyr 0 ,-........70....,v __________ NHCBZ ___________ ,. PFP0-....(....,0-........
NHCBZ
r Or 0 Ott 0 HO2C) PFPO

BocHN N

BocHN NH2 H 1. HCI or TFA
____________________ ,-DIPEA BocHN N y---,r ---..V __ NHCBZ _____________ ,..
,r 2.

0.. ingc.7....... 0 0 o BocHNN)/ Ac0 -4.LI OPFF
H
165 NHAc OAc 166 0.. ing_v_____. 0 0 0, ,,,I.,L 1. 1,6-hexanediol Ac0 H or 1,5-pentane-diol NHAc '-) IIINN),r-----,\ TMSOTf + compound 4 OAc 2. TEMPO
0.. ing 0 0 0, 3. PFPTFA, pyr H H
N
M4 r NHAc o 0 0 OAc K
HNN) OAc Ac0 NHAc The triPFP ester 164 was prepared from acid 113 using the procedure outlined in example 53 above and reacted with mono-Boc protected diamine to provide 165 in essentially quantitative yield. The Boc groups were removed with hydrochloric acid or trifluoroacetic acid to provide the triamine which was reacted with the PFP activated acid 166 in the presence of a suitable base such as DIPEA to provide Compound 18.
The PFP protected Gal-NAc acid 166 was prepared from the corresponding acid by treatment with PFPTFA (1-1.2 eq) and pyridine (1-1.2 eq) in DMF. The precursor acid in turn was prepared from the corresponding alcohol by oxidation using TEMPO (0.2 eq) and BAIB in acetonitrile and water. The precursor alcohol was prepared from sugar intermediate 4 by reaction with 1,6-hexanediol (or 1,5-pentanediol or other diol for other n values) (2-4 eq) and TMSOTf using conditions described previously in example 47.

Example 55: Dose-dependent study of oligonucleotides comprising either a 3' or 5'-conjugate group (comparison of Ga1NAc3-1, 3, 8 and 9) targeting SRB-1 in vivo The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice. Unconjugated ISIS 353382 was included as a standard. Each of the various Ga1NAc3 conjugate groups was attached at either the 3' or 5' terminus of the respective oligonucleotide by a phosphodiester linked 2'-deoxyadenosine nucleoside (cleavable moiety).
Table 39 Modified ASO targeting SRB-1 SEQ
ASO Sequence (5 to 3') Motif Conjugate ID No.
ISIS 353382 GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds 5/10/5 none (parent) mCdaTdaTeamCeamCeaTeaTe G mC T T mC Ad Gd IT,' Ad I'd Gd ISIS 655861 es eseses essss sssss 5/10/5 Ga1NAc3-1 mCda'r ds'f esmCesmCesTesTeoAdo¨GalNAc3¨la Ges es es es es s s mC T T mC Ad Gd s IT,'s s Ad I'ds Gds s ISIS 664078 5/10/5 Ga1NAc3-9 mC m daTdsTesC m esCesTesTeoAdo¨es-9a Ga1NAc3-3a-o'Ado ISIS 661161 GeamCeaTeaTeamCesAdaGasTasmCdsAdsTasGasAds 5/10/5 Ga1NAc3-3 254 mCdaTdaTeamCesmCesTesTe Ga1NAC3-8a¨o'Ado ISIS 665001 GeamCeaTeaTeamCesAdaGdaTasmCdsAdsTasGasAds 5/10/5 Ga1NAc3-8 254 mCdaTdaTeamCesmCesTesTe Capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine.
Subscripts: "e" indicates a 2'-MOE modified nucleoside; "d" indicates a [3-D-2'-deoxyribonuc1eoside; "s"
indicates a phosphorothioate internucleoside linkage (PS); "o" indicates a phosphodiester internucleoside linkage (PO); and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.
The structure of Ga1NAc3-1a was shown previously in Example 9. The structure of Ga1NAc3-9 was shown previously in Example 52. The structure of Ga1NAc3-3 was shown previously in Example 39. The structure of Ga1NAc3-8 was shown previously in Example 47.
Treatment Six week old male Balb/c mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once at the dosage shown below with ISIS 353382, 655861, 664078, 661161, 665001 or with saline. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration to determine the liver SRB-1 mRNA levels using real-time PCR and RIBOGREENO
RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols.
The results below are presented as the average percent of SRB-1 mRNA levels for each treatment group, normalized to the saline control.

As illustrated in Table 40, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner. Indeed, the antisense oligonucleotides comprising the phosphodiester linked Ga1NAc3-1 and Ga1NAc3-9 conjugates at the 3' terminus (ISIS 655861 and ISIS
664078) and the Ga1NAc3-3 and Ga1NAc3-8 conjugates linked at the 5' terminus (ISIS 661161 and ISIS
665001) showed substantial improvement in potency compared to the unconjugated antisense oligonucleotide (ISIS 353382).
Furthermore, ISIS 664078, comprising a Ga1NAc3-9 conjugate at the 3' terminus was essentially equipotent compared to ISIS 655861, which comprises a Ga1NAc3-1 conjugate at the 3' terminus. The 5' conjugated antisense oligonucleotides, ISIS 661161 and ISIS 665001, comprising a Ga1NAc3-3 or Ga1NAc3-9, respectively, had increased potency compared to the 3' conjugated antisense oligonucleotides (ISIS 655861 and ISIS 664078).
Table 40 ASOs containing Ga1NAc3-1, 3, 8 or 9 targeting SRB-1 Dosage SRB-1 mRNA
ISIS No.Conj ug ate (mg/kg) (`)/0 Saline) Saline n/a 100 353382 10 68 none 0.5 98 1.5 76 655861 GalNac3 -1 (3') 0.5 88 1.5 85 664078 GalNac3-9 (3') 0.5 92 1.5 59 661161 GalNac3-3 (5') 0.5 100 1.5 73 665001 GalNac3-8 (5') Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in 15 serum were measured relative to saline injected mice using standard protocols. Total bilirubin and BUN were also evaluated. The change in body weights was evaluated with no significant change from the saline group.
ALTs, ASTs, total bilirubin and BUN values are shown in the table below.

Table 41 Dosage Total ISIS No. ALT AST BUN Conjugate mg/kg Bilirubin Saline 24 59 0.1 37.52 3 21 66 0.2 34.65 353382 10 22 54 0.2 34.2 none 30 22 49 0.2 33.72 0.5 25 62 0.2 30.65 1.5 23 48 0.2 30.97 655861 GalNac3-1 (3') 28 49 0.1 32.92 40 97 0.1 31.62 0.5 40 74 0.1 35.3 1.5 47 104 0.1 32.75 664078 GalNac3-9 (3') 5 20 43 0.1 30.62 15 38 92 0.1 26.2 0.5 101 162 0.1 34.17 1.5 g 42 100 0.1 33.37 661161 GalNac3-3 (5') 5 g 23 99 0.1 34.97 15 53 83 0.1 34.8 0.5 28 54 0.1 31.32 1.5 42 75 0.1 32.32 665001 GalNac3-8 (5') 5 24 42 0.1 31.85 15 32 67 0.1 31.
Example 56: Dose-dependent study of oligonucleotides comprising either a 3' or 5'-conjugate group 5 (comparison of Ga1NAc3-1, 2, 3, 5, 6, 7 and 10) targeting SRB-1 in vivo The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice. Unconjugated ISIS 353382 was included as a standard. Each of the various Ga1NAc3 conjugate groups was attached at the 5' terminus of the respective oligonucleotide by a phosphodiester linked 2'-deoxyadenosine nucleoside (cleavable moiety) except for ISIS 655861 which had the Ga1NAc3 conjugate 10 group attached at the 3' terminus.
Table 42 Modified ASO targeting SRB-1 SEQ
ASO Sequence (5' to 3') Motif Conjugate ID No.
ISIS 3 53 382 GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds 5/10/5 no conjugate 252 (parent) mCdsTdsTesmCesmCesTesTe G mC T T mC Ad Gd Td mCd Ad Td Gd Ad ISIS 655861 es eseses es s s s s s s s s 5/10/5 Ga1NAc3-1 mCdsTdsTesmCesmCesTesTeoAdo,-GaliSTAC3-1a GalINTAC3-2a-0,AdoGesmCesTesTesmCesAdsGdsrrds 5/10/5 ISIS 664507 Ga1NAc3-2 254 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe Ga1NAc3-3a-o'Ado GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds 5/10/5 Ga1NAc3-3 254 mCdsTdsTesmCesmCesTesTe ISIS 666224 Ga1NAC3-5a-0,AdoGesmCesTesTesmCesAdsGdsTds 5/10/5 Ga1NAc3-5 254 mCdsAdsTdsGdsAdsmCdsTasTesmCesmCesTesTe GalNAc3-6.-0,AdoGesmCesTesTesmCesAdsGasTas ISIS 666961 5/10/5 Ga1NAc3-6 254 mCdaAdaTdaGdaAdamCdaTasTesmCesmCesTesTe GalNAc3-7.-0,AdoGesmCesTesTesmCesAdsGasTas 5/10/5 ISIS 666981 Ga1NAc3-7 254 mCdaAdaTdaGdaAdamCdaTasTesmCesmCesTesTe GalNAc3-10.-0,AdoGesmCesTesTesmCesAdsGasTas 5/10/5 ISIS 666881 Ga1NAc3-10 254 mCdaAdaTdaGdaAdamCdaTasTesmCesmCesTesTe Capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine.
Subscripts: "e" indicates a 2'-MOE modified nucleoside; "d" indicates a [3-D-2'-deoxyribonuc1eoside; "s"
indicates a phosphorothioate internucleoside linkage (PS); "o" indicates a phosphodiester internucleoside linkage (PO); and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.
The structure of Ga1NAc3-1a was shown previously in Example 9. The structure of Ga1NAc3-2a was shown previously in Example 37. The structure of Ga1NAc3-3a was shown previously in Example 39. The structure of Ga1NAc3-5a was shown previously in Example 49. The structure of Ga1NAc3-6a was shown previously in Example 51. The structure of Ga1NAc3-7a was shown previously in Example 48. The structure of GalNAc3-10a was shown previously in Example 46.
Treatment Six week old male Balb/c mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once at the dosage shown below with ISIS 353382, 655861, 664507, 661161, 666224, 666961, 666981, 666881 or with saline. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration to determine the liver SRB-1 mRNA levels using real-time PCR and RIBOGREENO RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. The results below are presented as the average percent of SRB-1 mRNA levels for each treatment group, normalized to the saline control.
As illustrated in Table 43, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner.
Indeed, the conjugated antisense oligonucleotides showed substantial improvement in potency compared to the unconjugated antisense oligonucleotide (ISIS 353382). The 5' conjugated antisense oligonucleotides showed a slight increase in potency compared to the 3' conjugated antisense oligonucleotide.
Table 43 Dosage SRB-1 mRNA
ISIS No.Conjugate (mg/kg) (% Saline) Saline n/a 100.0 3 96.0 353382 10 73.1 none 36.1 655861 0.5 99.4 GalNac3-1 (3') 1.5 81.2 33.9 15.2 0.5 102.0 1.5 73.2 664507 GalNac3-2 (5') 5 31.3 15 10.8 0.5 90.7 1.5 67.6 661161 GalNac3-3 (5') 5 24.3 15 11.5 0.5 96.1 1.5 61.6 666224 GalNac3-5 (5') 5 25.6 15 11.7 0.5 85.5 1.5 56.3 666961 Ga1NAc3-6 (5') 5 34.2 15 13.1 0.5 84.7 1.5 59.9 666981 Ga1NAc3-7 (5') 5 24.9 15 8.5 0.5 100.0 1.5 65.8 666881 Ga1NAc3-10 (5') 5 26.0 15 13.0 Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in serum were measured relative to saline injected mice using standard protocols.
Total bilirubin and BUN were also evaluated. The change in body weights was evaluated with no significant change from the saline group.
5 ALTs, ASTs, total bilirubin and BUN values are shown in Table 44 below.
Table 44 Dosage Total ISIS No. ALT ASTBUN Conjugate mg/kg Bilirubin Saline 26 57 0.2 27 3 25 92 0.2 27 353382 10 23 40 0.2 25 none 30 29 54 0.1 28 0.5 25 71 0.2 34 1.5 28 60 0.2 26 655861 GalNac3-1 (3') 5 26 63 0.2 28 15 25 61 0.2 28 0.5 25 62 0.2 25 1.5 24 49 0.2 26 664507 GalNac3-2 (5') 5 21 50 0.2 26 15 59 84 0.1 22 0.5 20 42 0.2 29 1.5 g 37 74 0.2 25 661161 GalNac3-3 (5') g 28 61 0.2 29 21 41 0.2 25 0.5 34 48 0.2 21 1.5 23 46 0.2 26 666224 GalNac3-5 (5') 5 24 47 0.2 23 15 32 49 0.1 26 0.5 17 63 0.2 26 1.5 23 68 0.2 26 666961 Ga1NAc3-6 (5') 5 25 66 0.2 26 15 29 107 0.2 28 0.5 24 48 0.2 26 1.5 30 55 0.2 24 666981 Ga1NAc3-7 (5') 5 46 74 0.1 24 15 29 58 0.1 26 0.5 20 65 0.2 27 1.5 23 59 0.2 24 666881 Ga1NAc3-10 (5') 5 45 70 0.2 26 15 21 57 0.2 24 Example 57: Duration of action study of oligonucleotides comprising a 3'-conjugate group targeting ApoC III in vivo Mice were injected once with the doses indicated below and monitored over the course of 42 days for ApoC-III and plasma triglycerides (Plasma TG) levels. The study was performed using 3 transgenic mice that express human APOC-III in each group.
Table 45 Modified ASO targeting ApoC III
ASO Sequence (5' to 3') Linkages SEQ ID
No.
ISIS AesGesmCesTesTesmCdsTdsTdsGdsTds PS 244 304801 mCdsmCdsAdsGdsmCdsTeaTeaTesAes're ISIS
ikesGesmCesTesTesmCdsTdsTdsGdsTdsmCdsmCds PS 245 647535 AdsGdsmCdsTesTesTesAesTeoAdo,-GaINAC3-la ISIS AesGeomCeoTeoTeomCdsTdsTdsGdsTdsmCdsmCds PO/PS

647536 AdaGdamCdaTeoTeorresAesTeoAdo-GalNAc3-la Capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine.
Subscripts: "e" indicates a 2'-MOE modified nucleoside; "d" indicates a [3-D-2'-deoxyribonuc1eoside; "s"
indicates a phosphorothioate internucleoside linkage (PS); "o" indicates a phosphodiester internucleoside linkage (PO); and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.
The structure of Ga1NAc3-11 was shown previously in Example 9.

Table 46 ApoC III mRNA ( /0 Saline on Day 1) and Plasma TG Levels ( /0 Saline on Day 1) ASO Dose Target Day 3 Day 7 Day 14 Day 35 Day 42 Saline 0 mg/kg ApoC-III 98 100 100 95 116 ISIS 304801 30 mg/kg ApoC-III 28 30 41 65 74 ISIS 647535 10 mg/kg ApoC-III 16 19 25 74 94 ISIS 647536 10 mg/kg ApoC-III 18 16 17 35 51 Saline 0 mg/kg Plasma TG 121 130 123 105 109 ISIS 304801 30 mg/kg Plasma TG 34 37 50 69 69 ISIS 647535 10 mg/kg Plasma TG 18 14 24 18 71 ISIS 647536 10 mg/kg Plasma TG 21 19 15 32 35 As can be seen in the table above the duration of action increased with addition of the 3'-conjugate group compared to the unconjugated oligonucleotide. There was a further increase in the duration of action for the conjugated mixed PO/PS oligonucleotide 647536 as compared to the conjugated full PS
oligonucleotide 647535.
Example 58: Dose-dependent study of oligonucleotides comprising a 3'-conjugate group (comparison of Ga1NAc3-1 and Ga1NAc4-11) targeting SRB-1 in vivo The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice. Unconjugated ISIS 440762 was included as an unconjugated standard. Each of the conjugate groups were attached at the 3' terminus of the respective oligonucleotide by a phosphodiester linked 2'-deoxyadenosine nucleoside cleavable moiety.
The structure of Ga1NAc3-la was shown previously in Example 9. The structure of Ga1NAc3-11a was shown previously in Example 50.
Treatment Six week old male Balb/c mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once at the dosage shown below with ISIS 440762, 651900, 663748 or with saline. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration to determine the liver SRB-1 mRNA levels using real-time PCR and RIBOGREENO RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. The results below are presented as the average percent of SRB-1 mRNA levels for each treatment group, normalized to the saline control.
As illustrated in Table 47, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner. The antisense oligonucleotides comprising the phosphodiester linked GalNAc3-1 and Ga1NAc4-11 conjugates at the 3' terminus (ISIS 651900 and ISIS 663748) showed substantial improvement in potency compared to the unconjugated antisense oligonucleotide (ISIS 440762). The two conjugated oligonucleotides, GalNAc3-1 and Ga1NAc4-11, were equipotent.
Table 47 Modified ASO targeting SRB-1 % Saline SEQ ID
ASO Sequence (5 to 3') Dose mg/kg control No.
Saline 100 0.6 73.45 TIsmCksAdsGdsTasmCdsAdsrrdsGdsAds 2 59.66 ISIS 440762 mCdsTdsrIlsmCk 6 23.50 0.2 62.75 TksmCksAdsGdsrrdsmCdsAdsrrdsGdsAds 0.6 29.14 mCdsTdsTIsmCkoAdo-GallsTAC3-1. 2 8.61 6 5.62 0.2 63.99 TksmCksAdsGdsrrdsmCdsAdsrrdsGdsAds 0.6 33.53 mCdsTdsTIsmCkoAdo-GallsTAC4-11. 2 7.58 6 5.52 Capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine.
Subscripts: "e" indicates a 2'-MOE modified nucleoside; "k" indicates 6'-(S)-CH3 bicyclic nucleoside; "d"
indicates a 13-D-2'-deoxyribonuc1eoside; "s" indicates a phosphorothioate internucleoside linkage (PS); "o"
indicates a phosphodiester internucleoside linkage (PO); and "o- indicates -0-P(=0)(OH)-. Conjugate groups are in bold.
Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in serum were measured relative to saline injected mice using standard protocols.
Total bilirubin and BUN were also evaluated. The change in body weights was evaluated with no significant change from the saline group.
ALTs, ASTs, total bilirubin and BUN values are shown in Table 48 below.
Table 48 Dosage Total ISIS No. ALT AST BUN Conjugate mg/kg Bilirubin Saline 30 76 0.2 40 0.60 32 70 0.1 35 440762 2 26 57 0.1 35 none 6 31 48 0.1 39 0.2 32 115 0.2 39 0.6 33 61 0.1 35 651900 Ga1Nac3-1 (3') 2 30 50 0.1 37 6 34 52 0.1 36 0.2 28 56 0.2 36 663748 0.6 34 60 0.1 35 GalNac4-11 (3') 2 44 62 0.1 36 6 38 71 0.1 33 Example 59: Effects of GaINAc3-1 conjugated ASOs targeting FXI in vivo The oligonucleotides listed below were tested in a multiple dose study for antisense inhibition of FXI
in mice. ISIS 404071 was included as an unconjugated standard. Each of the conjugate groups was attached at the 3' terminus of the respective oligonucleotide by a phosphodiester linked 2'-deoxyadenosine nucleoside cleavable moiety.
Table 49 Modified ASOs targeting FXI
SEQ ID
ASO Sequence (5' to 3') Linkages No.
ISIS TeaGesGesTesAesAdsTdamCdamCdaAdamCds 404071 TdsTdsTdsmCdsAesGesAesGesGe ISIS TesGesGesTesAesAdsTasmCdsmCdsAdsmCds PS 256 656172 TdsTdsTdsmCdsAesGesAesGesGeoAdo,-Ga1NAc3-1a ISIS TesGeoGeorreoAeoAdsTdsmCdsmCdsAdsmCds 656173 TdsTdsTdsmCdsAeoGeoAesGesGeoAdo,-Ga1NAC3-1a Capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine.
Subscripts: "e" indicates a 2'-MOE modified nucleoside; "d" indicates a [3-D-2'-deoxyribonuc1eoside; "s"
indicates a phosphorothioate internucleoside linkage (PS); "o" indicates a phosphodiester internucleoside linkage (PO); and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.
The structure of Ga1NAc3-1a was shown previously in Example 9.
Treatment Six week old male Balb/c mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously twice a week for 3 weeks at the dosage shown below with ISIS 404071, 656172, 656173 or with PBS treated control. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration to determine the liver FXI mRNA levels using real-time PCR and RIBOGREENO RNA
quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. Plasma FXI
protein levels were also measured using ELISA. FXI mRNA levels were determined relative to total RNA
(using RIBOGREENO), prior to normalization to PBS-treated control. The results below are presented as the average percent of FXI mRNA levels for each treatment group. The data was normalized to PBS-treated control and is denoted as "% PBS". The ED50s were measured using similar methods as described previously and are presented below.
Table 50 Factor XI mRNA (% Saline) Dose ASO % Control Conjugate Linkages mg/kg Saline 100 none 404071 10 40 none PS

SIS 0.7 74 I
656172 2 33 GaINAc3-1 PS

SIS 0.7 49 I
656173 2 22 GaINAc3-1 PO/PS

As illustrated in Table 50, treatment with antisense oligonucleotides lowered FXI mRNA levels in a dose-dependent manner.
The oligonucleotides comprising a 3'-Ga1NAc3-1 conjugate group showed substantial improvement in potency compared to the unconjugated antisense oligonucleotide (ISIS 404071).
Between the two conjugated oligonucleotides an improvement in potency was further provided by substituting some of the PS linkages with PO (ISIS 656173).
As illustrated in Table 50a, treatment with antisense oligonucleotides lowered FXI protein levels in a dose-dependent manner.
The oligonucleotides comprising a 3'-Ga1NAc3-1 conjugate group showed substantial improvement in potency compared to the unconjugated antisense oligonucleotide (ISIS 404071).
Between the two conjugated oligonucleotides an improvement in potency was further provided by substituting some of the PS linkages with PO (ISIS 656173).
Table 50a Factor XI protein (% Saline) Dose Protein (%
ASO Conjugate Linkages mg/kg Control) Saline 100 none 404071 10 32 none PS

0.7 ISIS
656172 2 23 Ga1NAc3-1 PS

0.7 ISIS
656173 2 6 Ga1NAc3-1 PO/PS

Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in serum were measured relative to saline injected mice using standard protocols.
Total bilirubin, total albumin, CRE and BUN were also evaluated. The change in body weights was evaluated with no significant change from the saline group. ALTs, ASTs, total bilirubin and BUN values are shown in the table below.

Table 51 Dosage Total Total ISIS No. ALT AST
Albumin Bilirubin CRE BUN Conjugate mg/kg Saline 71.8 84.0 3.1 0.2 0.2 22.9 3 152.8 176.0 3.1 0.3 0.2 23.0 404071 10 73.3 121.5 3.0 0.2 0.2 21.4 none 30 82.5 92.3 3.0 0.2 0.2 23.0 0.7 62.5 111.5 3.1 0.2 0.2 23.8 656172 2 33.0 51.8 2.9 0.2 0.2 22.0 GalNac3-1 (3') 6 65.0 71.5 3.2 0.2 0.2 23.9 0.7 54.8 90.5 3.0 0.2 0.2 24.9 656173 2 85.8 71.5 3.2 0.2 0.2 21.0 GalNac3-1 (3') 6 114.0 101.8 3.3 0.2 0.2 22.7 Example 60: Effects of conjugated ASOs targeting SRB-1 in vitro The oligonucleotides listed below were tested in a multiple dose study for antisense inhibition of SRB-1 in primary mouse hepatocytes. ISIS 353382 was included as an unconjugated standard. Each of the conjugate groups were attached at the 3' or 5' terminus of the respective oligonucleotide by a phosphodiester linked 2'-deoxyadenosine nucleoside cleavable moiety.
Table 52 Modified ASO targeting SRB-1 SEQ
ASO Sequence (5' to 3') Motif Conjugate ID No.
G mC T T mC Ad Gd Td mCd Ad Td Gd Ad ISIS 353382 es eseses es s s s s s s s s 5/10/5 none mCdsTdsTesmCesmCesTesTe G mC T T mC Ad Gd Td mCd Ad Td Gd Ad ISIS 655861 es eseses es s s s s s s s s 5/10/5 GalNAc3-1 253 mCdsTdsTesmCesmCesTesTeoAdo,-Ga1NAC3-1 a G mC T T mC Ad Gd I'd mCd Ad I'd Gd Ad ISIS 655862 es eoeoeo eo s s s s s s s s 5/10/5 GalNAc3-1 253 mCdsTdsTeomCeomCesTesTeoAdo,-G aINAC3-1 a GaINAc3-3a-o'AdoGesmCesTesTesmCesAdsGds ISIS 661161 5/10/5 GaINAc3-3 TdsmCdsAdsTdsGdsAdsmCdsrrdsrresmCesniCesTesTe GalNAC3-8a-o'AdoGesmCesTesTesmCesAdsGds ISIS 665001 5/10/5 Ga1NAc3-8 254 TdsmCdsAdsTdsGdsAdsmCdsrrdsrresmCesniCesTesTe G mC T T mC Ad Gd Td mCd Ad Td Gd Ad ISIS 664078 es eseses es s s s s s s s s 5/10/5 GalNAc3-9 253 niCdsTdsTesmCesmCesTesTeoAdo,-Ga1NAC3-9a GaINAc3-6a-0,AdoGesmCesTesTesmCesAdsGas ISIS 666961 5/10/5 Ga1NAc3-6 TdsmCdsAdsTdsGdsAdsmCdsrrdsrresmCesmCesTesTe GalNAC3-2a-o'AdoGesmCesTesTesmCesAdsGdsrrds 5/10/5 ISIS 664507 Ga1NAc3-2 254 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GaINAc3-10a-0,AdoGesmCesTesTesmCesAdsGasTas 5/10/5 ISIS 666881 Ga1NAc3-10 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GalNAC3-5a-o'AdoGesmCesTesTesmCesAdsGdsrrds 5/10/5 ISIS 666224 Ga1NAc3-5 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GalNAC3-7a-o'AdoGesmCesTesTesmCesAdsGdsrrds 5/10/5 ISIS 666981 Ga1NAc3-7 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe Capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine.
Subscripts: "e" indicates a 2'-MOE modified nucleoside; "d" indicates a [3-D-2'-deoxyribonuc1eoside; "s"
indicates a phosphorothioate internucleoside linkage (PS); "o" indicates a phosphodiester internucleoside linkage (PO); and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.
The structure of Ga1NAc3-la was shown previously in Example 9. The structure of Ga1NAc3-3a was shown previously in Example 39. The structure of Ga1NAc3-8a was shown previously in Example 47. The structure of Ga1NAc3-9a was shown previously in Example 52. The structure of Ga1NAc3-6a was shown previously in Example 51. The structure of GalNAc3-2a was shown previously in Example 37. The structure of Ga1NAc3-10a was shown previously in Example 46. The structure of Ga1NAc3-5a was shown previously in Example 49. The structure of Ga1NAc3-7a was shown previously in Example 48.
Treatment The oligonucleotides listed above were tested in vitro in primary mouse hepatocyte cells plated at a density of 25,000 cells per well and treated with 0.03, 0.08, 0.24, 0.74, 2.22, 6.67 or 20 nM modified oligonucleotide. After a treatment period of approximately 16 hours, RNA was isolated from the cells and mRNA levels were measured by quantitative real-time PCR and the SRB-1 mRNA
levels were adjusted according to total RNA content, as measured by RIBOGREENO.
The 1050 was calculated using standard methods and the results are presented in Table 53. The results show that, under free uptake conditions in which no reagents or electroporation techniques are used to artificially promote entry of the oligonucleotides into cells, the oligonucleotides comprising a GalNAc conjugate were significantly more potent in hepatocytes than the parent oligonucleotide (ISIS 353382) that does not comprise a GalNAc conjugate.
Table 53 Internucleoside SEQ ID
ASO IC50 (nM) Conjugate linkages No.
ISIS 353382 190' PS none 252 ISIS 655861 1 la PS Ga1NAc3-1 253 ISIS 655862 3 PO/PS Ga1NAc3-1 253 ISIS 661161 15' PS Ga1NAc3-3 254 ISIS 665001 20 PS Ga1NAc3-8 254 ISIS 664078 55 PS Ga1NAc3-9 253 ISIS 666961 22' PS Ga1NAc3-6 254 ISIS 664507 30 PS Ga1NAc3-2 254 ISIS 666881 30 PS Ga1NAc3-10 254 ISIS 666224 30' PS Ga1NAc3-5 254 ISIS 666981 40 PS Ga1NAc3-7 254 'Average of multiple runs.

Example 61: Preparation of oligomeric compound 175 comprising GaINAc3-12 Ac0 0 OAc Bo c , H 0 Ac0 pfp(:),C)- ---T---c 91a Bo 0Ac c , HN...
______________________________________ 0 _____ N N 0 H
HN H
OAc 166 HN , Ac HOOC
H
N) N
0 Ac0 OAc CBz' \¨COOH

DC M HN
"Ac HBTU DIEA DMF

A.,10)Ac 0 0 OAc HN HN -Ac }_--N--7----j H Ac0 0 _____F)Ac O ON ..---õ,,..õ..----..._N \ v 0 ,,,,, \N--'N ).------C) OAc k-' HN H H
HN
"Ac----\---A 0 HN \ Azcl OAc OAc "Ac Ac..:10Ac 0 0 OAc Pd(OH)2/C, H2 0 H HN
HN Ac Me0H/Et0Ac }_--N --.7-----/
___________ 0-0 0 Ac0 Nz..7.0Ac H2N......õ..---,..õ.,....N \ 1t, ,\ N N
OAc HN H H
HN
"Ac----\-----\ 0 HN \ Azcl OAc OAc "Ac OF
benzyl (perfluorophenyl) glutarate DMF
Ac0 OAc HN HN,Ac Ac0 ON 0 OAc N
OAc H
HN
0 0 NNAc HN Ac0 OAc OAc HN, Ac Ac0 OAc K0 0,..2-0Ac ,,Ac HN

Pd(OH)2 i C , H2 },.- N--/-----/
172 __________ >
H m 0 0 Ac0 )Ac T
Me0H / Et0Ac HO N
0 0 ,-,\ \ N N
OAc H
HN'Ac HN Ac0 1:1:)Ac 0\ /0 OAc 173 HN, Ac Ac0 OAc PFP-TFA
0 0,21.__OAc DIEA DMF
HN)LVy HN --Ac OH
},¨N --.7----/
F F
HH Ac0 OAc 0 Ni_i F ii 0 N,,,N, ;:).

N
F F ' 0 ,_, u ,\ N
OAc HN--1-1 0 HN'Ac HN Ac0 1:1:)Ac \ /0 OAc 174 HN, Ac 83e 3'5' 11 J
L OLIGO -0-P-0-(CH2)6¨NH2 I
OH
174 1. Borate buffer, DMSO, pH 8.5, rt ________________________________ r 2. aq. ammonia, rt OH OH
HO...r(2,\

oN---\----N__Ic AcHN
NH

\------\--ENI
HO..)\...,o \--0 AcHN
___________________________________________________________________ OLIGO

N

if 0 j--NH

H0µ.....v HO
NHAc Compound 169 is commercially available. Compound 172 was prepared by addition of benzyl (perfluorophenyl) glutarate to compound 171. The benzyl (perfluorophenyl) glutarate was prepared by adding PFP-TFA and DIEA to 5-(benzyloxy)-5-oxopentanoic acid in DMF. Oligomeric compound 175, comprising a Ga1NAc3-12 conjugate group, was prepared from compound 174 using the general procedures illustrated in Example 46. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-12 (Ga1NAc3-12a) can be combined with any cleavable moiety to provide a variety of conjugate groups.
In a certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-. The structure of GalNAc3-12 (Ga1NAc3-12a-CM-) is shown below:

OH OH
HO*,o 0 AcHN N-------NANH
01-0H \------\__H
HOo\__N__\_ N N
AcHN /0 H H ri __ (,.,.....,r ENi0 En N
ri 0 0 _r_ri\--NH

HO,(7)....\./
HO
NHAc Example 62: Preparation of oligomeric compound 180 comprising Ga1NAc3-13 OAc OAc 0 \ , 0 Ac0----P__\_-0(OH + 1.1 HATU, HOAt AcHN
176 H2N- y N \V\V\v(:) H DIEA, DMF
O___- 0 V

OAc OAc Ac0--C--)-.\--ONc AcHN NH
OAc OAc H 0 H2, Pd/C
Ac0--C--:-1.--0)1N7NNO 401 AcHN
H H
0 y 0 /
OAc OAc HN

Ac0-----\:) .-0 AcHN 0 OAc7.- OAc Ac0--(2.\._-0),c AcHN NH
OAcf.- OAc O.

PFPTFA, TEA
Ac0---)-.\--01.,Nri\ij=Li\ jr0H _______________________ AcHN
H H DMF

OAcf- OAc r 178 HN
Ac0---.\-) -0 AcHN 0 f.-Ac OAc AcO
AcHN NH
r-Ac OAc j=L .ro F
AcHN LNri\j rAc OAc AcHN
83e 3'5'1 I I
j OLIGO -0-P-0-(CH2)6-NH2 OH
1. Borate buffer, DMSO, pH 8.5, rt 2. aq. ammonia, rt r-H OH
HOO

AcHN NH
r..-H OH

AcHN )NHJN-IHNt")0¨ cm ¨ OLIGO

011-1 r OH 180 HOO
HN
AcHN 0 Compound 176 was prepared using the general procedure shown in Example 2.
Oligomeric compound 180, comprising a Ga1NAc3-13 conjugate group, was prepared from compound 177 using the general procedures illustrated in Example 49. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-13 (Ga1NAc3-130 can be combined with any cleavable moiety to provide a variety of conjugate groups. In a certainembodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-. The structure of Ga1NAc3-13 (Ga1NAc3-13a-CM-) is shown below:
OH OH

NH
AcHN
OH OH
0 ,cr HO H 0 0-..........."..,,,Ai\I-6 AcHN N NNr H 0 L'H 0 o( H
HO....12._\/ H 0 HO
NHAc Example 63: Preparation of oligomeric compound 188 comprising Ga1NAc3-14 H OAc HOIn HON)-NH2 HO 6 N-N Ac01r) 0 0 6 H 0 OTh Ac0 HO 0.¨NHCBz 181 HO)'61\11 H¨NHCBz N \ 0 4 r 0 0 HBTU, DIEA 0 0 HOHO.( OAc OAc Ac0\_( _ AcO\ ( H H
N
Ac0 111110N-6NICI Ac0 Villikk. -0N-6 lin OAc NHAc 0 0 OAc NHAc 0 0 Ac0 , H AcO\ ( _ oNE11..r 0N-N 0NHCBz Pd/C, H2 Ac0 '6 Ac0 .Ø..4-N
H21111./ ' / 6 NHAc NHAc 0 0 0 OAc OAc HN4 Ac0 ,( __ 16 0 Ac00{)\---) 0 Ac0 6H
Ac0 NHAc NHAc 183 OAc Ac0 H
N
Ac0 ON-6 In 0 H01..i0 el OAc NHAc 0 0 H 1. Pd/C, H2 2. PFP.TFA, pyr, Ac0 -N--6-N1c.õ-0,4-NI DMF

HBTU, NHAc 0 OAc DIEA, Ac000N)\---) 4.
DMF
Ac0 6H
NHAc OAc Ac0 H F
Ac0 ON-6N1n o F 0 F
\.,:,/0Ac Ac0 h NHAc µ o o 0 .---IL
0-1 N y(:)/}h 0 F
Ac0 / 6 F
NHAc 0 0 0 OAc Ac004,./."-, N N)--) Ac0 % 16H
NHAc 83e HOOH, ( III

3'-re ( OLIG0J-0-P-0-(CH2)6-NH2 0 0 OH HON NHAc H 0 LT.
OH cyN-N1r,0,1¨N CM

187 1. Borate buffer, DMSO, pH 8.5, rt HO 6 H 6 OLIGO
NHAc 0 0 0 2. aq. ammonia,HO
rt OH
H0\111/ /6H 188 NHAc Compounds 181 and 185 are commercially available. Oligomeric compound 188, comprising a Ga1NAc3-14 conjugate group, was prepared from compound 187 using the general procedures illustrated in Example 46.
The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-14 (Ga1NAc3-14a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-. The structure of GalNAc3-14 (Ga1NAc3-14a-CM-) is shown below:

HO N
io H
AcHN

0 Ea H H
AcHN 0 HOOH
HO
io H
AcHN
Example 64: Preparation of oligomeric compound 197 comprising Ga1NAc3-15 Ac0 OA OTBS OTBS
AcO__..r!..:)..\0/---,/---1OH
)\ Ac0 OA

Ac0_..i.Ø...vo._z=-----.../'-""
AcHN N 0 H

HBTU, DIEA AcHN

7 N NH3/Me0H OTBS
___________ ..-0 _________________________________________________________ 1 HO Bz20, DMAP
-112--\ ----/-----/'""1 AcHN

OH
OTBS
Bz0 Bz Bz0 OBz_* 0 0 Et3N.HF Bz0 ___T.O.s\z 0 Bz0 0 AcHN
AcHN 193 -----( (0-F,\,'r Phosphitylation Bz0 Bz __________ i.
___..Ø....\.,0õ./-----.../N 5 Bz0 1\i AcHN

DMTO
N(iPr)2 DMTO /
DMTO
O
DMT0/\7---0 5' 3' 195 CM j Oligo =
DMT0/\7----0 SS, DNA synthesizer 196 OH
,---OH
HO NI_ 1. 194, DNA synthesizer AcHN

2. Aq NH3 55 C, 18 h OH o 0-17N/7) __________________________________________________________________ Oligo NHAc 0 0¨P¨OH

OH
H0/0\...>/
HO NHAc Compound 189 is commercially available. Compound 195 was prepared using the general procedure shown in Example 31. Oligomeric compound 197, comprising a Ga1NAc3-15 conjugate group, was prepared from compounds 194 and 195 using standard oligonucleotide synthesis procedures. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-15 (Ga1NAc3-15a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-.
The structure of GalNAc3-15 (Ga1NAc3-15a-CM-) is shown below:

HOOH
AcHN 0 0, HOOH
og HO
AcHN 0 1) HO H
HO
NHAc Example 65: Dose-dependent study of oligonucleotides comprising a 5'-conjugate group (comparison of Ga1NAc3-3, 12, 13, 14, and 15) targeting SRB-1 in vivo The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice. Unconjugated ISIS 353382 was included as a standard. Each of the Ga1NAc3 conjugate groups was attached at the 5' terminus of the respective oligonucleotide by a phosphodiester linked 2'-deoxyadenosine nucleoside (cleavable moiety).
Table 54 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') Conjugate SEQ
No.
ID
No.
353382 none GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe 661161 17 1XT A 2 A r.;_ rTT cA, T A A
aii.,,c3-,a-0,-40¨esm¨es es esms,s, .-ass=-=ds dsms-,ds, .clsnisds ds Ga1NAc3-3 254 m m Tes Ces CesTesTe 671144 GalNAc3-12a-0,AdoGesmCesTesTesmCesAdsGdsrrdsmCdsAdsrrdsGdsAdsmCdsrrds Ga1NAc3-12 254 m m Tes Ces CesTesTe 670061 GalNAc3-13a-0,AdoGesmCesTesTesmCesAdsGdsrrdsmCdsAdsrrdsGdsAdsmCdsrrds Ga1NAc3-13 254 m m Tes Ces CesTesTe 671261 GalNAC3-14a-0,AdoGesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTds Ga1NAc3-14 254 m m Tes Ces CesTesTe 671262 GalNAc3-15a-0,AdoGesmCesTesTesmCesAdsGdsrrdsmCdsAdsrrdsGdsAdsmCdsrrds Ga1NAc3-15 254 m m Tes Ces CesTesTe Capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine. Subscripts:
"e" indicates a 2'-MOE modified nucleoside; "d" indicates a [3-D-2'-deoxyribonuc1eoside; "s" indicates a phosphorothioate internucleoside linkage (PS); "o" indicates a phosphodiester internucleoside linkage (PO);
and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.

The structure of Ga1NAc3-3a was shown previously in Example 39. The structure of Ga1NAc3-12a was shown previously in Example 61. The structure of Ga1NAc3-13a was shown previously in Example 62.
The structure of Ga1NAc3-14a was shown previously in Example 63. The structure of Ga1NAc3-15a was shown previously in Example 64.
Treatment Six to eight week old C57b16 mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once or twice at the dosage shown below with ISIS 353382, 661161, 671144, 670061, 671261, 671262, or with saline. Mice that were dosed twice received the second dose three days after the first dose. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration to determine the liver SRB-1 mRNA levels using real-time PCR and RIBOGREENO
RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. The results below are presented as the average percent of SRB-1 mRNA levels for each treatment group, normalized to the saline control.
As illustrated in Table 55, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner. No significant differences in target knockdown were observed between animals that received a single dose and animals that received two doses (see ISIS
353382 dosages 30 and 2 x 15 mg/kg; and ISIS 661161 dosages 5 and 2 x 2.5 mg/kg). The antisense oligonucleotides comprising the phosphodiester linked Ga1NAc3-3, 12, 13, 14, and 15 conjugates showed substantial improvement in potency compared to the unconjugated antisense oligonucleotide (ISIS 335382).
Table 55 SRB-1 mRNA (% Saline) ISIS No. Dosage (mg/kg) SRB-1 mRNA (% ED50 (mg/kg) Conjugate Saline) Saline n/a 100.0 n/a n/a 3 85.0 10 69.2 353382 30 34.2 22.4 none 2 x 15 36.0 0.5 87.4 1.5 59.0 661161 5 25.6 2.2 Ga1NAc3-3 2 x 2.5 27.5 15 17.4 0.5 101.2 1.5 76.
671144 3.4 Ga1NAc3-12 5 32.0 15 17.6 0.5 94.8 670061 1.5 57.8 2.1 Ga1NAc3-13 5 20.7 15 13.3 0.5 110.7 1.5 81.9 671261 4.1 Ga1NAc3-14 39.8 15 14.1 0.5 109.4 1.5 99.5 671262 9.8 Ga1NAc3-15 5 69.2 15 36.1 Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in serum were measured relative to saline injected mice using standard protocols.
Total bilirubin and BUN were also evaluated. The changes in body weights were evaluated with no significant differences from the saline 5 group (data not shown). ALTs, ASTs, total bilirubin and BUN values are shown in Table 56 below.
Table 56 Total Dosage ALT BUN
ISIS No. AST (U/L) Bilirubin Conjugate (mg/kg) (U/L) (Ing/d1-) (Ing/d1-) Saline n/a 28 60 0.1 39 n/a 3 30 77 0.2 36 10 25 78 0.2 36 353382 none 30 28 62 0.2 35 2 x 15 22 59 0.2 33 0.5 39 72 0.2 34 1.5 26 50 0.2 33 661161 5 41 80 0.2 32 Ga1NAc3-3 2 x 2.5 24 72 0.2 28 15 32 69 0.2 36 0.5 25 39 0.2 34 1.5 26 55 0.2 28 671144 Ga1NAc3-5 48 82 0.2 34 15 23 46 0.2 32 0.5 27 53 0.2 33 1.5 24 45 0.2 35 670061 Ga1NAc3-5 23 58 0.1 34 15 24 72 0.1 31 0.5 69 99 0.1 33 1.5 34 62 0.1 33 671261 Ga1NAc3-5 43 73 0.1 32 15 32 53 0.2 30 0.5 24 51 0.2 29 1.5 32 62 0.1 31 671262 Ga1NAc3-5 30 76 0.2 32 15 31 64 0.1 32 Example 66: Effect of various cleavable moieties on antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising a 5'-GaINAc3 cluster The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice. Each of the Ga1NAc3 conjugate groups was attached at the 5' terminus of the respective oligonucleotide by a phosphodiester linked nucleoside (cleavable moiety (CM)).
Table 57 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') Ga1NAc3 CM SEQ
No. Cluster ID
No.
661161 Ga1NAc1-3 - Ad mC T T mC A G T mC A T Ga1NAc3-3 a Ad 254 - a ()' ¨ es es es es es ds ds ds ds ds ds m m GdsAds CdsT ds es es Ces CesTesTe 670699 Ga1NAc3-3a-0 -0 ,Td mC T T mC A G T mC A T Ga1NAc3-3 a Td es eo eo eo eo ds ds ds ds ds ds m m GdsAds CdsTdsTeo Ceo CT
dsesTe 670700 Ga1NAc1-3 - A G mC T T mC A G T mC A T
- a ()' a Ga1NAc3-3 a Ae es eo eo eo eo ds ds ds ds ds ds m m GdsAds CdsTdsTeo Ceo CesT es 670701 Ga1NAc3-3a - ,T mC T T mC A G T mC A T
a Ga1NAc3-3 a Te 257 es eo eo eo eo ds ds ds ds ds ds m m GdsAds CdsTdsTeo Ceo CT
dsesTe 671165 Ga1NAc3-13 - mC T T mC A G T m o,A
a doC A T Ga1NAc3-13 a Ad es eo eo eo eo ds ds ds ds ds ds m m GdsAds CdsTdsTeo Ceo CesT es Capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine. Subscripts:
"e" indicates a 2'-MOE modified nucleoside; "d" indicates a [3-D-2'-deoxyribonuc1eoside; "s" indicates a phosphorothioate internucleoside linkage (PS); "o" indicates a phosphodiester internucleoside linkage (PO);
and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.
The structure of Ga1NAc3-3a was shown previously in Example 39. The structure of Ga1NAc3-13a was shown previously in Example 62.
Treatment Six to eight week old C57b16 mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once at the dosage shown below with ISIS 661161, 670699, 670700, 670701, 671165, or with saline. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration to determine the liver SRB-1 mRNA levels using real-time PCR
and RIBOGREENO RNA
quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. The results below are presented as the average percent of SRB-1 mRNA levels for each treatment group, normalized to the saline control.
As illustrated in Table 58, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner. The antisense oligonucleotides comprising various cleavable moieties all showed similar potencies.

Table 58 SRB-1 mRNA (% Saline) ISIS No. Dosage (mg/kg) SRB-1 mRNA Ga1NAc3 CM
(% Saline) Cluster Saline n/a 100.0 n/a n/a 0.5 87.8 1.5 61.3 661161 Ga1NAc3-3a Ad 33.8 14.0 0.5 89.4 1.5 59.4 670699 Ga1NAc3-3a Td 5 31.3 15 17.1 0.5 79.0 1.5 63.3 670700 Ga1NAc3-3a Ae 5 32.8 15 17.9 0.5 79.1 1.5 59.2 670701 Ga1NAc3-3a I', 5 35.8 15 17.7 0.5 76.4 1.5 43.2 671165 Ga1NAc3-13a Ad 5 22.6 15 10.0 Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in 5 serum were measured relative to saline injected mice using standard protocols. Total bilirubin and BUN were also evaluated. The changes in body weights were evaluated with no significant differences from the saline group (data not shown). ALTs, ASTs, total bilirubin and BUN values are shown in Table 56 below.
Table 59 Total CM
Dosage ALT AST . BUN Ga1NAc3 ISIS No. Bilirubm (mg/kg) (U/L) (U/L) (mg/dL) Cluster (mg/d1-) Saline n/a 24 64 0.2 31 n/a n/a 0.5 25 64 0.2 31 1.5 24 50 0.2 32 661161 Ga1NAc3-3a Ad 5 26 55 0.2 28 15 27 52 0.2 31 0.5 42 83 0.2 31 1.5 33 58 0.2 32 670699 Ga1NAc3-3a Td 5 26 70 0.2 29 15 25 67 0.2 29 0.5 40 74 0.2 27 1.5 23 62 0.2 27 670700 Ga1NAc3-3a A, 5 24 49 0.2 29 15 25 87 0.1 25 0.5 30 77 0.2 27 1.5 22 55 0.2 30 670701 Ga1NAc3-3a Te 81 101 0.2 25 31 82 0.2 24 0.5 44 84 0.2 26 1.5 47 71 0.1 24 671165 Ga1NAc3-13a Ad 5 33 91 0.2 26 15 33 56 0.2 29 Example 67: Preparation of oligomeric compound 199 comprising GaINAc3-16 OAc AcONC:Ac 0 AcHN 0(1\1 HN-- CC

OAc OAc 0 H
AcOo<t\)( ,NH H 0 zODMTr \ 2 \ i 2 --N 1. Succinic anhydride, AcHN DMAP, DCE
OAc OAc 0 r ) 7 r---i .-.r!:..:)...\,-, ir,c2," ENIN,hy HN ---0 / N 2. DMF, HBTU, DIEA, PS-SS
Ac0 1/4-',1 0 OH
AcHN 0 98d Ac0 OAc ___..!..D..\, H H
N ,A-- N 0 Ac0 , f 2 AcHN 0 ODMT
Ac0 OAc H H
1. DNA Synthesizer Ac0__ Ni--2 H \ 2. aq. NH3 AcHN 0 0 0 Ac0 OAc 0 ri,_:_,,r/
_L,)-v----õ---- 2 0 Ac0 0 AcHN 198 t HO OH
H H
...:0 HO__ _______________________________________________________________________ , HO OH AcHN 0 CM }Holigo . ___________________________________________________________________________ ' \ 0 0 Z
H :
HO 0...........õN.Nhi H
----y------____---NL
AcHN
HO OH HN

HO
__ r 0..,..,)-v------i, , , 2 AcHN

Oligomeric compound 199, comprising a Ga1NAc3-16 conjugate group, is prepared using the general procedures illustrated in Examples 7 and 9. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-16 (Ga1NAc3-16a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-.The structure of Ga1NAc3-16 (Ga1NAc3-16a-CM-) is shown below:

4 H 2 H ( cm ) /
AcHN H 0 0 _-0 HOOH 0 N J-.L F
NR1-10--72.-1)4LH (*(NEIN
AcHN OH

HO__..7.2...\,,0 .---AcHN
Example 68: Preparation of oligomeric compound 200 comprising GaINAc3-17 OAc 0 0 83e Ac00Ac 3' 5, II
0 , A
AcHN 0" ' ' .N1"N 0 F (OLIG0J-0-P-0-(CH2)6-0 H 0 F fa F OH
Oa: i_cLA0c . N H
1. Borate buffer, DMSO, pH 8.5, rt Ac0 N 0 F ________________ AcHN OAc OAc H H H v.-0 F 2. aq. ammonia, rt Ac0 0 N
7----.......- HN n -AcHN 0 102a HO__.7.2..\01-)LENIN) H
AcHN 0 0 N. 0¨TiV1 ___________________________________________________________ CF_IG:) AcHN

( HO_...r..2Ø---...(1.-11.N.-...õ..--,,,N 0 AcHN

Oligomeric compound 200, comprising a Ga1NAc3-17 conjugate group, was prepared using the general procedures illustrated in Example 46. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-17 (Ga1NAc3-17a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-. The structure of Ga1NAc3-17 (Ga1NAc3-17a-CM-) is shown below:

_,Ii!.....\,D ...Ø--TjAN.--..,.....--.,N..11...........\

AcHN H 0 0 H H
AcHN

AcHN
Example 69: Preparation of oligomeric compound 201 comprising GaINAc3-18 OAc Ac0 00Ac 0 83e AcHN ON"...\NNI_? F 3' 5' II
H 0 0 F 16 F ( OLIG0)-0-7-0-(CH2)6-NH2 OAcc__ OAc F OH
AcHNOAc OAc /2 H H 1=Borate buffer, DMSO, pH 8.5, rt H 0 r F . 0-AcO-r----C) 071_,)ThrNHN-0 2. aq. ammonia, rt AcHN 2 0 102b _..1.2..\,..õ
HO 01/4$11\j) H
AcHN 0 0 ,---..k.A4 .,...._ " " 0¨ CM __________________________________________________________________ OLIGO
HO---).-\--- 11N.--------'7 H H
H
AcHN ( ___7,2..\,,..
HO 0$1N
.4 H
AcHN 201 Oligomeric compound 201, comprising a Ga1NAc3-18 conjugate group, was prepared using the general procedures illustrated in Example 46. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-18 (Ga1NAc3-18a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-. The structure of Ga1NAc3-18 (Ga1NAc3-18a-CM-) is shown below:
HOOH o 0 HO\
H
AcHN H 0 0 .(/)kFNIO¨(cvi, ______________________________________ 4 AcHN

HO-- 0,--h.--..õ..õ---,N NO
e. H
AcHN

Example 70: Preparation of oligomeric compound 204 comprising GaINAc3-19 AcO0Ac AcO0Ac HBTU, DMF, DIEA
Ac0 OH __________________ Ac0-712-\, N
AcHN DMTO AcHN

DMTO
HC5: 47 AcO0Ac Phosphitylation Ac0 1C
N ...,10 NC 1. DNA
synthesizer AcHN
2. aq. NH3 203 DMTO (iPr)2N
.gH
HO OH
HO
j 0 0 AcHN
0=P¨OH

HO OH
HO

AcHN
0=P¨OH
HO OH
HO
0 cm __ OLIGO
AcHN

Oligomeric compound 204, comprising a Ga1NAc3-19 conjugate group, was prepared from compound 64 using the general procedures illustrated in Example 52. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-19 (Ga1NAc3-19a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-. The structure of Ga1NAc3-19 (Ga1NAc3-19a-CM-) is shown below:

pH
HooH
HO

AcHN
0=P¨OH
HoOH
HOAorN0 AcHN
0=P¨OH
11) HOOH
HO
AcHN

Example 71: Preparation of oligomeric compound 210 comprising GaINAc3-20 F

F 0 F EtN(iPr)2, CH3CN Fr.õ.1,,),)..L

FF H ___________________________________ )x 3 N -iii0H
F)KriN DMTO 0 -b11-1 HO
AcO0Ac 0 Ac0C)Aopfp K2CO3/Methanol H2N,A-i,A AcHN 166 3 N =.iii0H
ACN

Ac0_...zrOAc 01..,....Nr(:)N
0 Phosphitylation ..iii0H _____ 3.--Ac0 AcHN
DMTO

AcO0Ac 1. DNA synthesizer NO
______________________________________________________________ ..-_....r.2...\,..____ A...iio\p,..0)NC
Ac0 0 p 2. aq. NH3 AcHN I
209 DMTO (iPr)2N
pH
HO OH

__....r....\, 0 02,y\------AcHN I
0=P¨OH
I
p OH 0 ..
HO__T.......\, AcHN
I
0=P¨OH
I
p OH 0 .='' H0_4, 111("iJL

0 0 __ Cm __ OLIGO
AcHN 210 Compound 205 was prepared by adding PFP-TFA and DIEA to 6-(2,2,2-trifluoroacetamido)hexanoic acid in acetonitrile ,which was prepared by adding triflic anhydride to 6-aminohexanoic acid. The reaction mixture was heated to 80 C, then lowered to rt. Oligomeric compound 210, comprising a Ga1NAc3-20 conjugate group, was prepared from compound 208 using the general procedures illustrated in Example 52. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-20 (Ga1NAc3-20a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-. The structure of Ga1NAc3-20 (Ga1NAc3-20a-CM-) is shown below:

AcHN 0 0 0=P ¨OH

OH

AcHN 0 0 0=---P ¨OH
OH
,0 HO '3 3 AcHN 0 0 gril Example 72: Preparation of oligomeric compound 215 comprising GaINAc3-21 HO----L AcO0Ac 0 OH
NH
H O AcO0Ac Ac0--4.,kOH
AcHN 176 _______________________________________ ).- 0 Ac0 11---rti-j BOP, EtN(iPr)2, 1,2-dichloroethane AcHN OH

ODMT
AcO0Ac 0 DMICI, Pyridine,rt Phosphitylation ______________ )1.- Ac0---12--\,(:).----Ifl AcHN
OH

NC
0¨) /
0P\ 1. DNA synthesizer AcO0Ac 0N(iPr)2 ___________________________ .
2. aq. NH3 Ac0---, AcHN
----I¨ODMT

OH
OH
rj HO___72..\
HO N--.____\._____.

AcHN
I
0=P¨OH
I

OH
r----/
HO
HO___4.
0 (:) I N
"3 I
O
AcHN
I
0-=P¨OH
I

OH
nj HO*,,,,(2..\
0' HO N
0 -----10 ____ cm OLIGO
AcHN

Compound 211 is commercially available. Oligomeric compound 215, comprising a Ga1NAc3-21 conjugate group, was prepared from compound 213 using the general procedures illustrated in Example 52. The Ga1NAc3 cluster portion of the conjugate group GalNAc3-21 (GalNAc3-21 a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-. The structure of GalNAc3-21 (Ga1NAc3-21a-CM-) is shown below:
OH
OH
nj HO 0`)CrN-------1_, AcHN
I
0=P¨OH
I

OH
r---i HO___070.
O N

AcHN a I
0=P¨OH
I

OH
FI
0 --------0 El AcHN

Example 73: Preparation of oligomeric compound 221 comprising Ga1NAc3-22 H ,N OH H

).L,NxOH
II

DIEA ACN
F

H
DMT-CI F3CN Nx0DMTr _______________ , II
pyridine 0 H Me0H / H20 H2N N x0DMTr Ac0 /0Ac F

NHAc F F
F

OAc Ac011 )-.NI ODMTr L C0\70-1 Phosphitylation Ac0/\ 0 H ___________________________________________________________ .
NHAc OAc AcO
Ac0 \ EN11.LN x0DMTr \/
H
NHAc 220 NC c), PI NN(i p 02 01H(,0 NHAc 1. DNA Synthesizer 0 I ,0 OH , OHCO2. Aq. NH3 P(OH\V

NHAc OH 0 1,0 ,P( OHCo OH
HO
\V 0 NHAc 0.(NTo _____________________________________________________ F:)ligc Compound 220 was prepared from compound 219 using diisopropylammonium tetrazolide. Oligomeric compound 221, comprising a Ga1NAc3-21 conjugate group, is prepared from compound 220 using the general procedure illustrated in Example 52. The Ga1NAc3 cluster portion of the conjugate group Ga1NAc3-22 (Ga1NAc3-22a) can be combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, the cleavable moiety is -P(=0)(OH)-Ad-P(=0)(OH)-. The structure of Ga1NAc3-22 (Ga1NAc3-22a-CM-) is shown below:

OF-&_\vor OH

NHAc o OH o 1C) .P.
OF& o.rNNO OH
\/

NHAc o OH o 1.0 01-&_\vorN OH

NHAc C)( __ Example 74: Effect of various cleavable moieties on antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising a 5'-GaINAc3 conjugate The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice. Each of the Ga1NAc3 conjugate groups was attached at the 5' terminus of the respective oligonucleotide.

Table 60 Modified ASOs targeting SRB-1 ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
m m m m G CTT CAGT CA
TGA CTT
353382 m es m es es es es ds ds ds ds ds dsdsds dsdses rila n/a 252 Ces CesTesTe GaINAC3-3am m m m'Ad G CTT CAGT CAT
m 661161 m 0 es es es es es ds ds ds ds ds ds m Ga1NAc3-3 a Ad GdsAds CdsTdsTes Ces CesTesTe o GalNAc3-3a - ,G m m m CTT CAGT CAT
666904 mm es mes es es es ds ds ds ds ds ds Ga1NAc3-3 a PO 252 GdsAds CdsTdsTes Ces CesTesTe GalNAc3-17a-0,AdoG m m m CT T CAGT CAT
675441 m m m es es es es es ds ds ds ds ds ds Ga1NAc3-17 a Ad 254 GdsAds CdsTdsTes Ces CesTesTe GaINAC3-18a-0,AdoG m m m CTT CAGT CAT
675442 m m m es es es es es ds ds ds ds ds ds Ga1NAc3-18 a Ad 254 GdsAds CdsTdsTes Ces CesTesTe In all tables, capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine.
Subscripts: "e" indicates a 2'-MOE modified nucleoside; "d" indicates a [3-D-2'-deoxyribonuc1eoside; "s"
indicates a phosphorothioate internucleoside linkage (PS); "o" indicates a phosphodiester internucleoside linkage (PO); and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.
The structure of Ga1NAc3-3a was shown previously in Example 39. The structure of Ga1NAc3-17a was shown previously in Example 68, and the structure of GalNAc3-18a was shown in Example 69.
Treatment Six to eight week old C57BL/6 mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once at the dosage shown below with an oligonucleotide listed in Table 60 or with saline.
Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration to determine the SRB-1 mRNA levels using real-time PCR and RIBOGREENO RNA
quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. The results below are presented as the average percent of SRB-1 mRNA levels for each treatment group, normalized to the saline control.
As illustrated in Table 61, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner. The antisense oligonucleotides comprising a GalNAc conjugate showed similar potencies and were significantly more potent than the parent oligonucleotide lacking a GalNAc conjugate.
Table 61 SRB-1 mRNA (% Saline) ISIS No. Dosage (mg/kg) SRB-1 mRNA Ga1NAc3 CM
(% Saline) Cluster Saline n/a 100.0 n/a n/a 3 79.38 353382 10 68.67 n/a n/a 30 40.70 0.5 79.18 1.5 75.96 661161 Ga1NAc3-3a Ad 30.53 12.52 0.5 91.30 1.5 57.88 666904 Ga1NAc3-3a PO
5 21.22 15 16.49 0.5 76.71 1.5 63.63 675441 Ga1NAc3-17a Ad 5 29.57 15 13.49 0.5 95.03 1.5 60.06 675442 Ga1NAc3-18a Ad 5 31.04 15 19.40 Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in serum were measured relative to saline injected mice using standard protocols.
Total bilirubin and BUN were also evaluated. The change in body weights was evaluated with no significant change from the saline group 5 (data not shown). ALTs, ASTs, total bilirubin and BUN values are shown in Table 62 below.
Table 62 Total CM
Dosage ALT AST. BUN Ga1NAc3 ISIS No. Bilirubm (mg/kg) (U/L) (U/L) (mg/dL) Cluster (mg/d1-) Saline n/a 26 59 0.16 42 n/a n/a 3 23 58 0.18 39 353382 10 28 58 0.16 43 n/a n/a 30 20 48 0.12 34 0.5 30 47 0.13 35 1.5 23 53 0.14 37 661161 Ga1NAc3-3a Ad 5 26 48 0.15 39 15 32 57 0.15 42 0.5 24 73 0.13 36 1.5 21 48 0.12 32 666904 Ga1NAc3-3a PO
5 19 49 0.14 33 15 20 52 0.15 26 0.5 42 148 0.21 36 1.5 60 95 0.16 34 675441 Ga1NAc3-17a Ad 5 27 75 0.14 37 15 24 61 0.14 36 0.5 26 65 0.15 37 1.5 25 64 0.15 43 675442 Ga1NAc3-18a Ad 5 27 69 0.15 37 15 30 84 0.14 37 Example 75: Pharmacokinetic analysis of oligonucleotides comprising a 5'-conjugate group The PK of the ASOs in Tables 54, 57 and 60 above was evaluated using liver samples that were obtained following the treatment procedures described in Examples 65, 66, and 74. The liver samples were minced and extracted using standard protocols and analyzed by IP-HPLC-MS
alongside an internal standard.
The combined tissue level (Kg/g) of all metabolites was measured by integrating the appropriate UV peaks, and the tissue level of the full-length ASO missing the conjugate ("parent,"
which is Isis No. 353382 in this case) was measured using the appropriate extracted ion chromatograms (EIC).
Table 63 PK Analysis in Liver ISIS No. Dosage Total Tissue Level Parent ASO Tissue Ga1NAc3 CM
(mg/kg) by UV (Kg/g) Level by EIC (pg/g) Cluster 353382 3 8.9 8.6 10 22.4 21.0 n/a n/a 30 54.2 44.2 661161 5 32.4 20.7 Ga1NAc3-3a Ad 63.2 44.1 671144 5 20.5 19.2 Ga1NAc3-12a Ad 15 48.6 41.5 670061 5 31.6 28.0 Ga1NAc3-13a Ad 15 67.6 55.5 671261 5 19.8 16.8 Ga1NAc3-14a Ad 15 64.7 49.1 671262 5 18.5 7.4 Ga1NAc3-15a Ad 15 52.3 24.2 670699 5 16.4 10.4 Ga1NAc3-3a Td 15 31.5 22.5 670700 5 19.3 10.9 Ga1NAc3-3a A, 15 38.1 20.0 670701 5 21.8 8.8 Ga1NAc3-3a Te 15 35.2 16.1 671165 5 27.1 26.5 Ga1NAc3-13a Ad 15 48.3 44.3 666904 5 30.8 24.0 Ga1NAc3-3a PO
15 52.6 37.6 675441 5 25.4 19.0 Ga1NAc3-17a Ad 15 54.2 42.1 675442 5 22.2 20.7 Ga1NAc3-18a Ad 15 39.6 29.0 The results in Table 63 above show that there were greater liver tissue levels of the oligonucleotides comprising a Ga1NAc3 conjugate group than of the parent oligonucleotide that does not comprise a Ga1NAc3 conjugate group (ISIS 353382) 72 hours following oligonucleotide administration, particularly when taking 15 into consideration the differences in dosing between the oligonucleotides with and without a Ga1NAc3 conjugate group. Furthermore, by 72 hours, 40-98% of each oligonucleotide comprising a Ga1NAc3 conjugate group was metabolized to the parent compound, indicating that the Ga1NAc3 conjugate groups were cleaved from the oligonucleotides.
Example 76: Preparation of oligomeric compound 230 comprising GaINAc3-23 L, ToSCI NaN3 HO,---..õ.Ø.õ..õ,...--..,00 11 -DP- H0000Ts Pyr 4, TMSOTf OAc 0 N3 OAc____T..___ OAc 224 NHAc Pd(OH)2 OAcC)Ac ACN
_____________ ).-________________________________________________________________________ lr H2, Et0Ac, Me0H OAc 0 0 H2 7 F F \
NHAc \ F 0¨/K )/3 C¨ NO2 OAcOAc H
OAc 0 OAc OAc NHAc H NO2 1) Reduce 0 C)----ON 2) Couple Diacid 0 3) Pd/C
OAc 0 0 4) PFPTFA
NHAc OAc OAc ,NH
(:)C) OAc NHAc 228 OAc OAc____T..___ H

OAc F
OAc Ac NHAc H NH F
1C) 0 0 0-----0\/N
OAc 0 0 0 -0 NHAc oAcOAc ._---OAc F
NH
NHAc 229 83e 3' 5' ( OLIGO )¨O¨P-0¨(CH2)6-NI-12 I
OH
1. Borate buffer, DMSO, pH 8.5, rt ____________________________ "..-2. aq. ammonia, rt OH H

OH._.\---__frOC).--" 7 OH H
H 1.(11-11i N A.-.)0 sm ____________________________________________________________ OHNHAc -i , 0 C)=-=-=ON

oligo OH 0 0 0 , ______ ., NHAc OF___7......
0...... -.NH
0 (:) 0 OH
NHAc 230 Compound 222 is commercially available. 44.48 ml (0.33 mol) of compound 222 was treated with tosyl chloride (25.39 g, 0.13 mol) in pyridine (500mL) for 16 hours. The reaction was then evaporated to an oil, dissolved in Et0Ac and washed with water, sat. NaHCO3, brine, and dried over Na2SO4. The ethyl acetate was concentrated to dryness and purified by column chromatography, eluted with Et0Ac/hexanes (1:1) followed by 10% methanol in CH2C12 to give compound 223 as a colorless oil. LCMS and NMR were consistent with the structure. 10 g (32.86 mmol) of 1-Tosyltriethylene glycol (compound 223) was treated with sodium azide (10.68 g, 164.28 mmol) in DMSO (100mL) at room temperature for 17 hours. The reaction mixture was then poured onto water, and extracted with Et0Ac. The organic layer was washed with water three times and dried over Na2504. The organic layer was concentrated to dryness to give 5.3g of compound 224 (92%). LCMS and NMR were consistent with the structure. 1-Azidotriethylene glycol (compound 224, 5.53 g, 23.69 mmol) and compound 4 (6 g, 18.22 mmol) were treated with 4A molecular sieves (5g), and TMSOTf (1.65 ml, 9.11 mmol) in dichloromethane (100mL) under an inert atmosphere.
After 14 hours, the reaction was filtered to remove the sieves, and the organic layer was washed with sat.
NaHCO3, water, brine, and dried over Na2504. The organic layer was concentrated to dryness and purified by column chromatography, eluted with a gradient of 2 to 4% methanol in dichloromethane to give compound 225. LCMS and NMR were consistent with the structure. Compound 225 (11.9 g, 23.59 mmol) was hydrogenated in Et0Ac/Methanol (4:1, 250mL) over Pearlman's catalyst.
After 8 hours, the catalyst was removed by filtration and the solvents removed to dryness to give compound 226. LCMS and NMR were consistent with the structure.
In order to generate compound 227, a solution of nitromethanetrispropionic acid (4.17 g, 15.04 mmol) and Hunig's base (10.3 ml, 60.17 mmol) in DMF (100mL) were treated dropwise with pentaflourotrifluoro acetate (9.05 ml, 52.65 mmol). After 30 minutes, the reaction was poured onto ice water and extracted with Et0Ac. The organic layer was washed with water, brine, and dried over Na2SO4. The organic layer was concentrated to dryness and then recrystallized from heptane to give compound 227 as a white solid. LCMS and NMR were consistent with the structure. Compound 227 (1.5 g, 1.93 mmol) and compound 226 (3.7 g, 7.74 mmol) were stirred at room temperature in acetonitrile (15 mL) for 2 hours. The reaction was then evaporated to dryness and purified by column chromatography, eluting with a gradient of 2 tol 0% methanol in dichloromethane to give compound 228. LCMS and NMR were consistent with the structure. Compound 228 (1.7 g, 1.02 mmol) was treated with Raney Nickel (about 2g wet) in ethanol (100mL) in an atmosphere of hydrogen. After 12 hours, the catalyst was removed by filtration and the organic layer was evaporated to a solid that was used directly in the next step. LCMS and NMR were consistent with the structure. This solid (0.87 g, 0.53 mmol) was treated with benzylglutaric acid (0.18 g, 0.8 mmol), HBTU (0.3 g, 0.8 mmol) and DIEA (273.7 [tl, 1.6 mmol) in DMF (5mL).
After 16 hours, the DMF
was removed under reduced pressure at 65 C to an oil, and the oil was dissolved in dichloromethane. The organic layer was washed with sat. NaHCO3, brine, and dried over Na2504. After evaporation of the organic layer, the compound was purified by column chromatography and eluted with a gradient of 2 to 20%
methanol in dichloromethane to give the coupled product. LCMS and NMR were consistent with the structure. The benzyl ester was deprotected with Pearlman's catalyst under a hydrogen atmosphere for 1 hour. The catalyst was them removed by filtration and the solvents removed to dryness to give the acid.
LCMS and NMR were consistent with the structure. The acid (486 mg, 0.27 mmol) was dissolved in dry DMF (3 mL). Pyridine (53.61 [tl, 0.66 mmol) was added and the reaction was purged with argon.
Pentaflourotriflouro acetate (46.39 [tl, 0.4 mmol) was slowly added to the reaction mixture. The color of the reaction changed from pale yellow to burgundy, and gave off a light smoke which was blown away with a stream of argon. The reaction was allowed to stir at room temperature for one hour (completion of reaction was confirmed by LCMS). The solvent was removed under reduced pressure (rotovap) at 70 C. The residue was diluted with DCM and washed with 1N NaHSO4, brine, saturated sodium bicarbonate and brine again. The organics were dried over Na2504, filtered, and were concentrated to dryness to give 225 mg of compound 229 as a brittle yellow foam. LCMS and NMR were consistent with the structure.
Oligomeric compound 230, comprising a Ga1NAc3-23 conjugate group, was prepared from compound 229 using the general procedure illustrated in Example 46. The Ga1NAc3 cluster portion of the Ga1NAc3-23 conjugate group (Ga1NAc3-23a) can be combined with any cleavable moiety to provide a variety of conjugate groups. The structure of GalNAc3-23 (Ga1NAc3-23a-CM) is shown below:

OH
01-1.v..... H

0 c0 N7 OH
OH H
01-1....\vNHAcH NH N (,-.)C) 0 (:) ""----ONI-r Ir 4 In OH
+

OH 0\r0 NHAc Oi__ ...... 0 OH NH

NHAc Example 77: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising a Ga1NAc3 conjugate The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.
Table 64 Modified ASOs targeting SRB-1 ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID
No.
m m m GalNAc3-3a-0,AdoG C T T C A G T C A T
661161 m m mes es es es es ds ds ds ds ds ds Ga1NAc3-3a Ad 254 GdsAds CdsTdsTes Ces CesTesTe m m m GalNAc1-3 - ,G CTT CAGT CAT
666904 m - a es es es es es ds ds ds ds ds ds m m Ga1NAc3-3a PO 252 GdsAds CdsTdsTes Ces CesTesTe m m m GaiNAC3-10a-0,AdoG CTT CAGT CAT
673502 m m m es eo eo eo eo ds ds ds ds ds ds Ga1NAc3-10a Ad 254 GdsAds CdsTdsTeo Ceo CesTesTe m m m GaINAC3-9am'AdoG CTT CAGT CAT
677844 m m mes es es es es ds ds ds ds ds ds Ga1NAc3-9a Ad 254 GdsAds CdsTdsTes Ces CesTesTe m m m GaINAC3-23a-0,AdoG CTT CAGT CAT
677843 m m m es es es es es ds ds ds ds ds ds Ga1NAc3-23a Ad 254 GdsAds CdsTdsTes Ces CesTesTe m m m m m G CTT CAGT CATGA CTT C
655861 m es es es es es ds ds ds ds ds ds ds ds ds ds es es Ga1NAc3-la Ad 253 CesTesTeoAdo,-GalNAc3-la m m m m m G CTT CAGT CATGA CTT C
677841 m es es es es es ds ds ds ds ds ds ds ds ds ds es es Ga1NAc3-19a Ad 253 CesTesTeoAdo,-GalNAc3-19a m m m m m G CTT CAGT CAT GA CTT C
677842 m es es es es es ds ds ds ds ds ds ds ds ds ds es es Ga1NAc3-20a Ad 253 CesTesTeoAdo,-GalNAc3-20a The structure of Ga1NAc3-1a was shown previously in Example 9, Ga1NAc3-3a was shown in Example 39, Ga1NAc3-9a was shown in Example 52, Ga1NAc3-10a was shown in Example 46, Ga1NAc3-19a was shown in Example 70, Ga1NAc3-20a was shown in Example 71, and Ga1NAc3-23a was shown in Example 76.

Treatment Six to eight week old C57BL/6 mice (Jackson Laboratory, Bar Harbor, ME) were each injected subcutaneously once at a dosage shown below with an oligonucleotide listed in Table 64 or with saline. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration to determine the SRB-1 mRNA levels using real-time PCR and RIBOGREENO RNA
quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. The results below are presented as the average percent of SRB-1 mRNA levels for each treatment group, normalized to the saline control.
As illustrated in Table 65, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner.
Table 65 SRB-1 mRNA (% Saline) ISIS No. Dosage (mg/kg) SRB-1 mRNA Ga1NAc3 CM
(% Saline) Cluster Saline n/a 100.0 n/a n/a 0.5 89.18 1. 77.02 661161 Ga1NAc3-3a Ad 5 29.10 12.64 0.5 93.11 1. 55.85 666904 Ga1NAc3-3a PO
5 21.29 15 13.43 0.5 77.75 1. 41.05 673502 Ga1NAc3-10a Ad 5 19.27 15 14.41 0.5 87.65 1. 93.04 677844 Ga1NAc3-9a Ad 5 40.77 15 16.95 0.5 102.28 1. 70.51 677843 Ga1NAc3-23a Ad 5 30.68 15 13.26 0.5 79.72 1. 55.48 655861 Ga1NAc3-la Ad 5 26.99 15 17.58 0.5 67.43 1. 45.13 677841 Ga1NAc3-19a Ad 5 27.02 15 12.41 0.5 64.13 1. 53.56 677842 Ga1NAc3-20a Ad 5 20.47 15 10.23 Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in serum were also measured using standard protocols. Total bilirubin and BUN
were also evaluated. Changes in body weights were evaluated, with no significant change from the saline group (data not shown). ALTs, ASTs, total bilirubin and BUN values are shown in Table 66 below.
Table 66 Total CM
Dosage ALT AST. BUN Ga1NAc3 ISIS No. Bilirubm (mg/kg) (U/L) (U/L) (mg/dL) Cluster (mg/d1-) Saline n/a 21 45 0.13 34 n/a n/a 0.5 28 51 0.14 39 1.5 23 42 0.13 39 661161 Ga1NAc3-3a Ad 5 22 59 0.13 37 21 56 0.15 35 0.5 24 56 0.14 37 1.5 26 68 0.15 35 666904 Ga1NAc3-3a PO
5 23 77 0.14 34 15 24 60 0.13 35 0.5 24 59 0.16 34 1.5 20 46 0.17 32 673502 Ga1NAc3-10 a Ad 5 24 45 0.12 31 15 24 47 0.13 34 0.5 25 61 0.14 37 1.5 23 64 0.17 33 677844 Ga1NAc3-9a Ad 5 25 58 0.13 35 15 22 65 0.14 34 0.5 53 53 0.13 35 1.5 25 54 0.13 34 677843 Ga1NAc3-23a Ad 5 21 60 0.15 34 15 22 43 0.12 38 0.5 21 48 0.15 33 1.5 28 54 0.12 35 655861 Ga1NAc3-la Ad 5 22 60 0.13 36 15 21 55 0.17 30 0.5 32 54 0.13 34 1.5 24 56 0.14 34 677841 Ga1NAc3-19a Ad 5 23 92 0.18 31 15 24 58 0.15 31 0.5 23 61 0.15 35 1.5 24 57 0.14 34 677842 Ga1NAc3-20a Ad 5 41 62 0.15 35 15 24 37 0.14 32 Example 78: Antisense inhibition in vivo by oligonucleotides targeting Angiotensinogen comprising a Ga1NAc3 conjugate 10 The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of Angiotensinogen (AGT) in normotensive Sprague Dawley rats.

Table 67 Modified ASOs targeting AGT
ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID
No.
mCesAesmCesTesGesikdsTdsTdsTasTasTdsGdsmCdsmCdsmCdsAesGes 552668 A n/a n/a 258 e mCesAesmCesTesGesAdsTdsTdsTdsTdsTdsGdsmCdsmCdsmCdsAesGes GalNAc3-la 669509 Ad 259 GesAesTeoAdo'¨Ga1NAc3-1a The structure of Ga1NAc3-1a was shown previously in Example 9.
Treatment Six week old, male Sprague Dawley rats were each injected subcutaneously once per week at a dosage shown below, for a total of three doses, with an oligonucleotide listed in Table 67 or with PBS. Each treatment group consisted of 4 animals. The rats were sacrificed 72 hours following the final dose. AGT liver mRNA levels were measured using real-time PCR and RIBOGREENO RNA
quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. AGT
plasma protein levels were measured using the Total Angiotensinogen ELISA (Catalog # JP27412, IBL
International, Toronto, ON) with plasma diluted 1:20,000. The results below are presented as the average percent of AGT mRNA levels in liver or AGT protein levels in plasma for each treatment group, normalized to the PBS control.
As illustrated in Table 68, treatment with antisense oligonucleotides lowered AGT liver mRNA and plasma protein levels in a dose-dependent manner, and the oligonucleotide comprising a GalNAc conjugate was significantly more potent than the parent oligonucleotide lacking a GalNAc conjugate.
Table 68 AGT liver mRNA and plasma protein levels ISIS Dosage (mg/kg) AGT liver AGT plasma Ga1NAc3 Cluster CM
No. mRNA (% PBS) protein (% PBS) PBS n/a 100 100 n/a n/a 552668 n/a n/a 0.3 95 70 669509 Ga1NAc3-la Ad Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in plasma and body weights were also measured at time of sacrifice using standard protocols. The results are shown in Table 69 below.
Table 69 Liver transaminase levels and rat body weights Body CM
Dosage Ga1NAc3 ISIS No. ALT (U/L) AST (U/L) Weight (%
(mg/kg) Cluster of baseline) PBS n/a 51 81 186 n/a n/a 552668 n/a n/a 0.3 53 90 190 669509 Ga1NAc3-la Ad Example 79: Duration of action in vivo of oligonucleotides targeting APOC-III
comprising a GaINAc3 conjugate The oligonucleotides listed in Table 70 below were tested in a single dose study for duration of action 10 in mice.
Table 70 Modified ASOs targeting APOC-III
ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
A G InC T T mCd Td I'd Gd Ta InCd mCd Ad Gd mCd T T
304801 es es es es es sss ss s s s s s es es rila n/a 244 TesAesTe Ae GesmCesTesTesmCd Td Id Gd Ta nCd mCd Ad Gd mCd TesT
es647535 Ga1NAc3-la Ad 245 TesAesTeoAdw-GalNAc3-la GalNAC3-3a-o'AdoAesGesmCesTesTesmCdsTdsTdsGdsTdsmCds 663083 Ga1NAc3-3a Ad 260 mCdsAdsGdsmCdsTesTes TesAesTe GalNAC3-7,-0'AdoAesGesmCesTesTesmCdsrrdsrrdsGdsrrdsmCds 674449 Ga1NAc3-7a Ad 260 mCdsAdsGdsmCdsTesTes TesAesTe GalNAc3-10am'AdoAesGesmCesTesTesniCdsrrdsrrdsGdsrrdsmCds GalNAc3-10 a Ad mCdsAdsGdsmCdsTesTes TesAesTe GalNAc3-13 am'AdoAesGesmCesTesTesmCdsrrdsrrdsGdsrrdsmCds GalNAc3-13a Ad mCdsAdsGdsmCdsTesTes TesAesTe The structure of Ga1NAc3-1 a was shown previously in Example 9, Ga1NAc3-3a was shown in Example 39, Ga1NAc3-7a was shown in Example 48, Ga1NAc3-10a was shown in Example 46, and Ga1NAc3-13a was shown in Example 62.

Treatment Six to eight week old transgenic mice that express human APOC-III were each injected subcutaneously once with an oligonucleotide listed in Table 70 or with PBS.
Each treatment group consisted of 3 animals. Blood was drawn before dosing to determine baseline and at 72 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, and 6 weeks following the dose. Plasma triglyceride and APOC-III protein levels were measured as described in Example 20. The results below are presented as the average percent of plasma triglyceride and APOC-III levels for each treatment group, normalized to baseline levels, showing that the oligonucleotides comprising a GalNAc conjugate group exhibited a longer duration of action than the parent oligonucleotide without a conjugate group (ISIS 304801) even though the dosage of the parent was three times the dosage of the oligonucleotides comprising a GalNAc conjugate group.
Table 71 Plasma triglyceride and APOC-III protein levels in transgenic mice Time point .APOC-III
ISIS Dosage TriglyceridesGalNAc3 CM
(days post- protein (%
No. (mg/kg) (% baseline) Cluster dose) baseline) PBS n/a 21 107 107 n/a n/a 304801 30 21 50 50 n/a n/a 647535 10 21 41 41 Ga1NAc3-la Ad 663083 10 21 28 28 Ga1NAc3-3a Ad 674449 10 3 29 26 Ga1NAc3-7a Ad 674450 10 21 44 44 Ga1NAc3-10 a Ad 674451 10 21 48 48 Ga1NAc3-13a Ad Example 80: Antisense inhibition in vivo by oligonucleotides targeting Alpha-1 Antitrypsin (AlAT) comprising a Ga1NAc3 Conjugate The oligonucleotides listed in Table 72 below were tested in a study for dose-dependent inhibition of Al AT in mice.
Table 72 Modified ASOs targeting AlAT
ISISGalNAc3 SEQ ID
Sequences (5' to 3') CM
No. Cluster No.
mCmC ITesAesAd Td Td mCd Ad Gd Ad Ad Gd Gd AesA
esAes es *_es rila n/a 261 GesGesAe 656326AmCmC
mC¨AAdTd¨TdmCdAdGdAdAdGdGdAA Ga1NAc3-la Ad 262 Uesresieoido'Ga1NAc31a GalNAc3-3a-o'AdoAesmCesmCesmCesAesAdsTdsTdsmCdsAdsGdsAds GalNAc3-3a Ad 263 AdsGdsGdsAesAes GesGesAe GalNAC3-7a¨o'AdoAesmCesmCesmCesAesAdsTdsTdsmCdsAdsGdsAds GalNAc3-7a Ad 263 AdsGdsGdsAesAes GesGesAe GalNAc3-10am'AdoAesmCesmCesmCesAesAdsTdsTdsmCdsAdsGds GalNAc3-10a Ad 263 AdsAdsGdsGdsAesAes GesGesAe GalNAc3-13 a-o'AdoAesmCesmCesmCesAesAdsTdsTdsmCdsAdsGds GalNAc3-13a Ad 263 AdsAdsGdsGdsAesAes GesGesAe The structure of Ga1NAc3-1 a was shown previously in Example 9, Ga1NAc3-3a was shown in Example 39, Ga1NAc3-7a was shown in Example 48, Ga1NAc3-10a was shown in Example 46, and Ga1NAc3-13a was shown in Example 62.

Treatment Six week old, male C57BL/6 mice (Jackson Laboratory, Bar Harbor, ME) were each injected subcutaneously once per week at a dosage shown below, for a total of three doses, with an oligonucleotide listed in Table 72 or with PBS. Each treatment group consisted of 4 animals.
The mice were sacrificed 72 hours following the final administration. Al AT liver mRNA levels were determined using real-time PCR and RIBOGREENO RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. AlAT plasma protein levels were determined using the Mouse Alpha 1-Antitrypsin ELISA
(catalog # 41-A1AMS-E01, Alpco, Salem, NH). The results below are presented as the average percent of Al AT liver mRNA and plasma protein levels for each treatment group, normalized to the PBS control.
As illustrated in Table 73, treatment with antisense oligonucleotides lowered Al AT liver mRNA and Al AT plasma protein levels in a dose-dependent manner. The oligonucleotides comprising a GalNAc conjugate were significantly more potent than the parent (ISIS 476366).
Table 73 AlAT liver mRNA and plasma protein levels ISIS Dosage (mg/kg) Al AT liver Al AT plasma Ga1NAc3 Cluster CM
No. mRNA (% PBS) protein (% PBS) PBS n/a 100 100 n/a n/a 15 73 61 n/a n/a 0.6 99 90 656326 Ga1NAc3-la Ad 0.6 105 90 6 16 20 Ga1NAc3-3a Ad 0.6 90 79 678382 Ga1NAc3-7a Ad 0.6 94 84 678383 Ga1NAc3-10 a Ad 0.6 106 91 678384 Ga1NAc3-13a Ad Liver transaminase and BUN levels in plasma were measured at time of sacrifice using standard protocols. Body weights and organ weights were also measured. The results are shown in Table 74 below.

Body weight is shown as % relative to baseline. Organ weights are shown as %
of body weight relative to the PBS control group.
Table 74 Body Liver Kidney Spleen ISIS Dosage ALT AST BUN
weight (% weight (Rel weight (Rel weight (Rel No. (mg/kg) (U/L) (U/L) (mg/dL) baseline) % BW) % BW) % BW) PBS n/a 25 51 37 119 100 100 100 0.6 29 57 40 123 100 103 119 0.6 26 57 32 117 93 109 110 0.6 26 42 35 114 100 103 103 0.6 30 67 38 121 91 100 123 0.6 36 63 31 118 98 103 98 Example 81: Duration of action in vivo of oligonucleotides targeting AlAT
comprising a GaINAc3 cluster The oligonucleotides listed in Table 72 were tested in a single dose study for duration of action in mice.
Treatment Six week old, male C57BL/6 mice were each injected subcutaneously once with an oligonucleotide listed in Table 72 or with PBS. Each treatment group consisted of 4 animals.
Blood was drawn the day before dosing to determine baseline and at 5, 12, 19, and 25 days following the dose. Plasma Al AT protein levels were measured via ELISA (see Example 80). The results below are presented as the average percent of plasma Al AT protein levels for each treatment group, normalized to baseline levels. The results show that the oligonucleotides comprising a GalNAc conjugate were more potent and had longer duration of action than the parent lacking a GalNAc conjugate (ISIS 476366). Furthermore, the oligonucleotides comprising a 5'-GalNAc conjugate (ISIS 678381, 678382, 678383, and 678384) were generally even more potent with even longer duration of action than the oligonucleotide comprising a 3'-GalNAc conjugate (ISIS 656326).
Table 75 Plasma AlAT protein levels in mice ISIS Dosage Time point Al AT (% Ga1NAc3 CM
No. (mg/kg) (days post- baseline) Cluster dose) PBS n/a n/a n/a 476366 100 n/a n/a 656326 18 Ga1NAc3-la Ad 678381 18 Ga1NAc3-3a Ad 678382 18 Ga1NAc3-7a Ad 678383 18 Ga1NAc3-10 a Ad 678384 18 Ga1NAc3-13a Ad Example 82: Antisense inhibition in vitro by oligonucleotides targeting SRB-1 comprising a GaINAc3 conjugate Primary mouse liver hepatocytes were seeded in 96 well plates at 15,000 cells/well 2 hours prior to treatment. The oligonucleotides listed in Table 76 were added at 2, 10, 50, or 250 nM in Williams E medium and cells were incubated overnight at 37 C in 5% CO2. Cells were lysed 16 hours following oligonucleotide addition, and total RNA was purified using RNease 3000 BioRobot (Qiagen). SRB-1 mRNA levels were determined using real-time PCR and RIBOGREENO RNA quantification reagent (Molecular Probes, Inc.
Eugene, OR) according to standard protocols. IC50 values were determined using Prism 4 software (GraphPad). The results show that oligonucleotides comprising a variety of different GalNAc conjugate groups and a variety of different cleavable moieties are significantly more potent in an in vitro free uptake experiment than the parent oligonucleotides lacking a GalNAc conjugate group (ISIS 353382 and 666841).
Table 76 Inhibition of SRB-1 expression in vitro ISISGalNAc IC50 SEQ
Sequence (5' to 3') Linkages CM
No. cluster (nM) ID
No.
m m m s Goa CesTesTes CesAdsGdsTds CdsAdsTds Gd Ads 353382 m m m Ps n/a n/a 250 252 CdsTdsTes Ces CesTesTe GoaniCeaTeaTeamCesAd Gd Ta mCd Ad I'd Gd Ad GalNAc3 655861 sss sssss Ps Ad 40 253 mCdaTdaT eamCesmCesT esTeoAdo,-GalNAC3-1 a -1a m m GalNAC3-3a-0,AdoGes CesTesTes CesAdsGds 'fas GalNAc3 661161 m m m m Ps Ad 40 254 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -3a m m GalNAc3-3a-,AdoGes CeoTeoTeo CeoAdsGdsTds Ga1NAc3 Ad 8 254 PO/PS
661162 m m m m0 CdsAdsTdsGdsAds CdsTds Teo Ceo CesTesTe -3a GoaniCeaTeaTeamCesAdsGdsTdsmCdsAdsTdsGdsAds Ga1NAc3 Ad 20 253 mCdaTdaTeamCesmCesTesTeoAdo¨GalNAc3-9a -9a GalNAC3-8a-0,AdoGesmCesTesTesmCesAd Gd Td GalNAc3 665001 s s s Ps Ad 70 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -8a GalNAC3-5a-0,AdoGesmCesTesTesmCesAdsGdsTds Ga1NAc3 Ad 80 254 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -5a m m m Goa CeoTeoTeo CesAdsGdsTds CdsAdsTdsGdsAds 666841 m m m PO/PS n/a n/a >250 252 CdsTds Teo Ceo CesTesTe GalNAc3-10a-0,AdoGesmCesTesTesmCesAdsGdsTds Ga1NAc3 Ad 30 254 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -10a m m m GalNAC3-3-0,Ges CesTesTes CesAdsGdsTds Cd GalNAc3 666904 m m ma s Ps PO 9 AdsTdsGdsAds CdaTds Tes Ces CesTesTe -3a m m s GalNAC3-3a-0,TdoGes CesTesTes CesAds Gd 'fas GalNAc3 666924 m m m m Ps Td 15 257 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -3a GalNAC3-6a Gd rra GalNAc3 666961 s s s Ps Ad 150 254 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -6a GalNAC3-7a Ga1NAc3 Ad 20 254 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -7a m m GalNAC3-13a-,AdoGes CesTesTes CesAdsGd 'fa GalNAc3 670061 m m m m0 s s Ps Ad 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -13a m m Ga1NAC3-3a-0,TdoG CTT CAGT Ga1NAc3 670699 m m es eo eo eo eo ds ds ds m m PO/PS Td 15 257 -3a CdsAdsTds GdsAds CdsTdsTeo Ceo CesTesTe m m Ga1NAC3-3a-0,AeoG CTT CAGT GalNAc3 Ae 30 670700 m m es eo eo eo eo ds ds ds m m PO/PS
-3a CdsAdsTds GdsAds CdsTdsTeo Ceo CesTesT
m m Ga1NAC3-3a-0,Te0G CTT CAGT GalNAc3 T
670701 m m es eo eo eo eo ds ds ds m m PO/PS e 25 257 -3a CdsAdsTds GdsAds CdsTdsTeo Ceo CesTesTe m m GalNAC3-12a-,AdoGes CesTesTes CesAdsGd 'fa GalNAc3 671144 m m m m0 s s Ps Ad 40 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -12a m m Ga1NAc3-139-0,AdoG CTT CAGT Ga1NAc3 Ad 8 254 671165 m m es eo eo eo eo ds ds ds m m PO/PS
CdsAdsTdsGdsAds CdsTdsTeo Ceo CesT 13a es-m m GalNAc3-14a-0,AdoGes CesTesTes CesAdsGdsTds GalNAc3 671261 m m m m PS
Ad >250 254 CdaAdaTdaGdaAda CdsTds Tes Ces CesTesTe -14a m m GalNAc3-15a-0,AdoGes CesTesTes CesAdsGdsTds GalNAc3 671262 m m m m PS
Ad >250 254 CdaAdaTdaGdaAda CdsTds Tes Ces CesTesTe -15a m m GalNAc3-7a-0,AoGes CeoTeoTeo CeoAdsGdsTds GalNAc3 d673501 m m m m PO/PS Ad 30 254 CdaAdaTdaGdaAda CdsTdsTeo Ceo CesTesTe -7a m m GalNAC3-10a-,AdoGes CeoTeoTeo CeoAdsGdsTds GalNAc3 0673502 m m m m PO/PS Ad 8 254 CdaAdaTdaGdaAda CdsTds Teo Ceo CesTesTe -10a m m GalNAc3-17a-,AdoGes CesTesTes CesAdsGdsTds GalNAc3 0675441 m m m m PS Ad 30 254 CdaAdaTdaGdaAda CdsTds Tes Ces CesTesTe -17a m m GalNAc3-18a-,AdoGes CesTesTes CesAdsGdsTds GalNAc3 0675442 m m m m PS Ad 20 254 CdaAdaTdaGdaAda CdsTds Tes Ces CesTesTe -18a GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds GalNAc3 Ad 40 253 mCdsTdsTesmCesmCesTesTeoAdo,-GalNAC3-19a -19a GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds GalNAc3 Ad 30 253 mCdsTdsTesmCesmCesTesTeoAdo,-GalNAC3-20a -20a m m GalNAC3-23a-,AdoGes CesTesTes CesAdsGdsTds GalNAc3 0677843 m m m m PS Ad 40 254 CdaAdaTdaGdaAda CdsTds Tes Ces CesTesTe -23a The structure of Ga1NAc3-1 a was shown previously in Example 9, Ga1NAc3-3a was shown in Example 39, Ga1NAc3-5a was shown in Example 49, Ga1NAc3-6a was shown in Example 51, Ga1NAc3-7a was shown in Example 48, Ga1NAc3-8a was shown in Example 47, Ga1NAc3-9a was shown in Example 52, Ga1NAc3-10a was shown in Example 46, Ga1NAc3-12a was shown in Example 61, Ga1NAc3-13a was shown in Example 62, Ga1NAc3-14a was shown in Example 63, Ga1NAc3-15a was shown in Example 64, Ga1NAc3-17a was shown in Example 68, Ga1NAc3-18a was shown in Example 69, Ga1NAc3-19a was shown in Example 70, Ga1NAc3-20a was shown in Example 71, and Ga1NAc3-23a was shown in Example 76.
Example 83: Antisense inhibition in vivo by oligonucleotides targeting Factor XI comprising a Ga1NAc3 cluster The oligonucleotides listed in Table 77 below were tested in a study for dose-dependent inhibition of Factor XI in mice.
Table 77 Modified oligonucleotides targeting Factor XI
ISIS GalNAc SEQ
Sequence (5' to 3') CM
No. cluster ID No.
TesGesGesTesAesAdsTdsmCdsmCdsAdsmCdsTdsTdsTdsmCdsAesGes 404071 A f-, f-, n/a n/a -/-lesUesk-le TesGeoGeoTecAeoAdsTdsmCdsmCdsAdsmCdsTdsTdsTdsmCdsAeoGeo 656173 Ga1NAc3-la Ad 256 AesGesGeoAdo,-GalNAc3-1 a 663086 Ga1NAc3-3a-0,AdoTesGeoGeoTeoAeoAdsTdsmCdsmCdsAdsmCdsTds Ga1NAc3-3a Ad TasTasmCdsAeoGeoAesGesGe GalNAc3-7a-0,AdoTesGeoGeoTeoAeoAdsTdsmCdsmCdsAdsmCdsTds GalNAc3-7a Ad TdsTdsmCdsAeoGeoAesGesGe GalNAc3-10a-0,AdoTesGeoGeoTeoAeoAdsTdsmCdsmCdsAdsmCds 678348 Ga1NAc3-10a Ad 264 TdsTdsTdsmCdsAeoGeoAesGesGe GalNAc3-13a-0,AdoTesGeoGeoTeoAeoAdsTdsmCdsmCdsAdsmCds 678349 Ga1NAc3-13a Ad 264 TdsTdsTdsmCdsAeoGeoAesGesGe The structure of Ga1NAc3-la was shown previously in Example 9, Ga1NAc3-3a was shown in Example 39, Ga1NAc3-7a was shown in Example 48, Ga1NAc3-10a was shown in Example 46, and Ga1NAc3-13a was shown in Example 62.
Treatment Six to eight week old mice were each injected subcutaneously once per week at a dosage shown below, for a total of three doses, with an oligonucleotide listed below or with PBS. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final dose. Factor XI liver mRNA
levels were measured using real-time PCR and normalized to cyclophilin according to standard protocols.
Liver transaminases, BUN, and bilirubin were also measured. The results below are presented as the average percent for each treatment group, normalized to the PBS control.
As illustrated in Table 78, treatment with antisense oligonucleotides lowered Factor XI liver mRNA
in a dose-dependent manner. The results show that the oligonucleotides comprising a GalNAc conjugate were more potent than the parent lacking a GalNAc conjugate (ISIS 404071).
Furthermore, the oligonucleotides comprising a 5'-GalNAc conjugate (ISIS 663086, 678347, 678348, and 678349) were even more potent than the oligonucleotide comprising a 3'-GalNAc conjugate (ISIS 656173).
Table 78 Factor XI liver mRNA, liver transaminase, BUN, and bilirubin levels ISIS Dosage Factor XI ALT AST BUN Bilirubin Ga1NAc3 SEQ
No. (mg/kg) mRNA (% PBS) (U/L) (U/L) (mg/dL) (mg/dL) Cluster ID No.
PBS n/a 100 63 70 21 0.18 n/a n/a 3 65 41 58 21 0.15 10 33 49 53 23 0.15 n/a 30 17 43 57 22 0.14 0.7 43 90 89 21 0.16 656173 2 9 36 58 26 0.17 Ga1NAc3-la 256 6 3 50 63 25 0.15 0.7 33 91 169 25 0.16 2 7 38 55 21 0.16 Ga1NAc3-3a 264 6 1 34 40 23 0.14 0.7 35 28 49 20 0.14 678347 2 10 180 149 21 0.18 Ga1NAc3-7a 264 6 1 44 76 19 0.15 0. 39 43 54 21 0.16 Ga1NAc3-10 a 264 2 5 38 55 22 0.17 6 2 25 38 20 0.14 0.7 34 39 46 20 0.16 678349 2 8 43 63 21 0.14 Ga1NAc3-13 a 6 2 28 41 20 0.14 Example 84: Duration of action in vivo of oligonucleotides targeting Factor XI
comprising a GaINAc3 Conj ug ate The oligonucleotides listed in Table 77 were tested in a single dose study for duration of action in mice.
Treatment Six to eight week old mice were each injected subcutaneously once with an oligonucleotide listed in Table 77 or with PBS. Each treatment group consisted of 4 animals. Blood was drawn by tail bleeds the day before dosing to determine baseline and at 3, 10, and 17 days following the dose. Plasma Factor XI protein levels were measured by ELISA using Factor XI capture and biotinylated detection antibodies from R & D
Systems, Minneapolis, MN (catalog # AF2460 and # BAF2460, respectively) and the OptEIA Reagent Set B
(Catalog # 550534, BD Biosciences, San Jose, CA). The results below are presented as the average percent of plasma Factor XI protein levels for each treatment group, normalized to baseline levels. The results show that the oligonucleotides comprising a GalNAc conjugate were more potent with longer duration of action than the parent lacking a GalNAc conjugate (ISIS 404071). Furthermore, the oligonucleotides comprising a 5'-GalNAc conjugate (ISIS 663086, 678347, 678348, and 678349) were even more potent with an even longer duration of action than the oligonucleotide comprising a 3'-GalNAc conjugate (ISIS 656173).
Table 79 Plasma Factor XI protein levels in mice ISIS Dosage Time point (days Factor XI (% CM
SEQ ID
GalNAc3 Cluster No. (mg/kg) post-dose) baseline) No.

PBS n/a 10 56 n/a n/a n/a
17 100 404071 30 10 47 n/a n/a 656173 6 10 3 Ga1NAc3-la Ad 663086 6 10 2 Ga1NAc3-3 a Ad 678347 6 10 1 Ga1NAc3-7a Ad 678348 6 10 1 Ga1NAc3-10 a Ad 678349 6 10 1 Ga1NAc3-13a Ad Example 85: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising a GaINAc3 Conjugate Oligonucleotides listed in Table 76 were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.
Treatment Six to eight week old C57BL/6 mice were each injected subcutaneously once per week at a dosage shown below, for a total of three doses, with an oligonucleotide listed in Table 76 or with saline. Each treatment group consisted of 4 animals. The mice were sacrificed 48 hours following the final administration to determine the SRB-1 mRNA levels using real-time PCR and RIBOGREENO RNA
quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. The results below are presented as the average percent of liver SRB-1 mRNA levels for each treatment group, normalized to the saline control.
As illustrated in Tables 80 and 81, treatment with antisense oligonucleotides lowered SRB-1 mRNA
levels in a dose-dependent manner.
Table 80 SRB-1 mRNA in liver ISIS No. Dosage (mg/kg) SRB-1 mRNA (% Ga1NAc3 Cluster CM
Saline) Saline n/a 100 n/a n/a 0.1 94 0.3 119 655861 Ga1NAc3-la Ad 0.1 120 0.3 107 661161 Ga1NAc3-3a Ad 0.1 107 0.3 107 666881 Ga1NAc3-10 a Ad 0.1 120 0. 103 666981 Ga1NAc3-7a Ad 0.
670061 Ga1NAc3-13a Ad 0.3 89 0.1 119 0.3 96 677842 Ga1NAc3-20a Ad Table 81 SRB-1 mRNA in liver ISIS No. Dosage (mg/kg) SRB-1 mRNA (% Ga1NAc3 Cluster CM
Saline) 0.1 107 0.3 95 661161 Ga1NAc3-3a Ad 0.1 110 0.3 88 677841 Ga1NAc3-19a Ad Liver transaminase levels, total bilirubin, BUN, and body weights were also measured using standard protocols. Average values for each treatment group are shown in Table 82 below.
Table 82 ISIS Dosage ALT AST Bilirubin BUN Body Weight Ga1NAc3 CM
No. (mg/kg) (U/L) (U/L) (mg/dL) (mg/dL) (% baseline) Cluster Saline n/a 19 39 0.17 26 118 n/a n/a 0.1 25 47 0.17 27 114 655861 0.3 29 56 0.15 27 118 Ga1NAc3-la Ad 1 20 32 0.14 24 112 3 27 54 0.14 24 115 0.1 35 83 0.13 24 113 42 61 0.15 23 117 661161 0.3 Ga1NAc3-3a Ad 1 34 60 0.18 22 116 3 29 52 0.13 25 117 0.1 30 51 0.15 23 118 666881 0.3 49 82 0.16 25 119 Ga1NAc3-10 a Ad 1 23 45 0.14 24 117 3 20 38 0.15 21 112 0.1 21 41 0.14 22 113 0.3 29 49 0.16 24 112 666981 Ga1NAc3-7a Ad 1 19 34 0.15 22 111 3 77 78 0.18 25 115 0.1 20 63 0.18 24 111 0.3 20 57 0.15 21 115 670061 Ga1NAc3-13a Ad 1 20 35 0.14 20 115 3 27 42 0.12 20 116 0.1 20 38 0.17 24 114 677842 0.3 31 46 0.17 21 117 Ga1NAc3-20a Ad 1 22 34 0.15 21 119 3 41 57 0.14 23 118 Example 86: Antisense inhibition in vivo by oligonucleotides targeting TTR
comprising a Ga1NAc3 cluster Oligonucleotides listed in Table 83 below were tested in a dose-dependent study for antisense inhibition of human transthyretin (TTR) in transgenic mice that express the human TTR gene.
Treatment Eight week old TTR transgenic mice were each injected subcutaneously once per week for three weeks, for a total of three doses, with an oligonucleotide and dosage listed in the tables below or with PBS.
Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration. Tail bleeds were performed at various time points throughout the experiment, and plasma TTR protein, ALT, and AST levels were measured and reported in Tables 85-87.
After the animals were sacrificed, plasma ALT, AST, and human TTR levels were measured, as were body weights, organ weights, and liver human TTR mRNA levels. TTR protein levels were measured using a clinical analyzer (AU480, Beckman Coulter, CA). Real-time PCR and RIBOGREENO RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) were used according to standard protocols to determine liver human TTR mRNA levels.
The results presented in Tables 84-87 are the average values for each treatment group. The mRNA levels are the average values relative to the average for the PBS group. Plasma protein levels are the average values relative to the average value for the PBS group at baseline. Body weights are the average percent weight change from baseline until sacrifice for each individual treatment group.
Organ weights shown are normalized to the animal's body weight, and the average normalized organ weight for each treatment group is then presented relative to the average normalized organ weight for the PBS
group.
In Tables 84-87, "BL" indicates baseline, measurements that were taken just prior to the first dose.
As illustrated in Tables 84 and 85, treatment with antisense oligonucleotides lowered TTR expression levels in a dose-dependent manner. The oligonucleotides comprising a GalNAc conjugate were more potent than the parent lacking a GalNAc conjugate (ISIS 420915). Furthermore, the oligonucleotides comprising a GalNAc conjugate and mixed PS/P0 internucleoside linkages were even more potent than the oligonucleotide comprising a GalNAc conjugate and full PS linkages.
Table 83 Oligonucleotides targeting human TTR
GalNAc SEQ
Isis No. Sequence 5 to 3' Linkages CM
cluster ID No.
T mC T I' G GdsTasTdsAdsmCdsAdsTdsGdsAdsAds 420915 es es es es es PS n/a n/a AesTesmCesmCesmCe TesmCesTesTesGesGdsTdsTdsAdsmCdsAdsTdsGdsAdsAds 660261 PS GalNAc3-1 a Ad 266 AesTesmCesmCesmCeoAdo'-GalNAc3-1, 682883 Ga1NAc3-3a_0,TesmCeoTeoTeoGeoGdsTdsrrdsAdsmCdsAds PS/PO
Ga1NAc3-3 a PO 265 TdsGasAdsAdsAeoTeomCesmCesmCe GalNAc3-7a0,TesmCeoreoreoGeoGasTasTasAdsmCd Ad s682884 PS/P0 Ga1NAc3-7a PO 265 TdsGdsAdsAdsAeoTeomCesmCesmCe GalNAc3-10a_0,TesmCeoTeoTeoGeoGdsTdsTdsAdsmCd 682885 s PS/P0 Ga1NAc3-10a PO 265 AdsTdsGdsAdsAdsAeoTeomCesmCesmCe GalNAc3-13a_0,TesmCeoTeoTeoGeoGdsTdsTdsAdsmCd 682886 s PS/P0 Ga1NAc3-13a PO 265 AdsTdsGdsAdsAdsAeoTeomCesmCesmCe TesmC7iTeoTeoGfGdsTdsTdsAdsmCdsAdsTdsGdsAdsAds 684057 PS/P0 Ga1NAc3-19a Ad 266 AeoTeo Ces Ces CeoAdo'-GalNAc3-19a The legend for Table 85 can be found in Example 74. The structure of Ga1NAc3-1 was shown in Example 9.
The structure of Ga1NAc3-3a was shown in Example 39. The structure of Ga1NAc3-7a was shown in Example 48. The structure of Ga1NAc3-10a was shown in Example 46. The structure of Ga1NAc3-13a was shown in Example 62. The structure of GalNAc3-19a was shown in Example 70.
Table 84 Antisense inhibition of human TTR in vivo Dosage TTR mRNA (% Plasma TTR protein SEQ
Isis No. GalNAc cluster CM
(mg/kg) PBS) (% PBS) ID No.
PBS n/a 100 100 n/a n/a 420915 20 48 65 n/a n/a 265 0.6 113 87 Ga1NAc3-la Ad 266 Table 85 Antisense inhibition of human TTR in vivo TTR Plasma TTR protein (% PBS at BL) SEQ
Dosage GalNAc Isis No. mRNA Day 17 CM ID
(mg/kg) BL Day 3 Day 10 cluster (% PBS) (After sac) No.
PBS n/a 100 100 96 90 114 n/a n/a 420915 20 43 102 66 61 58 n/a n/a 265 0.6 60 88 73 63 68 GalNAc3- PO

3a 0.6 56 88 78 63 67 GalNAc3- PO

7a 0.6 60 92 77 68 76 GalNAc3- PO

10a 682886 0.6 57 91 70 64 69 Ga1NAc3- PO 265 12 121 1 89 1 50 1 31 1 30 1 13a 1 1 0.6 53 80 69 56 62 684057 2 21 92 55 34 30 Ga1NAc3-Ad 266 6 11 82 50 18 13 19a Table 86 Transaminase levels, body weight changes, and relative organ weights Dos ALT (U/L) AST (U/L) Body Liver Spleen Kidne SEQ
age Isis No. (mg BL BL Day Day Day Day Day Day (%
(% (% Y (% ID
/kg) 3 10 17 3 10 17 BL) PBS) PBS) PBS) No.
PBS n/a 33 34 33 24 58 62 67 52 105 100 100 100 n/a 0.6 33 38 28 26 70 71 63 59 111 96 99 92 Table 87 Transaminase levels, body weight changes, and relative organ weights Dos ALT (U/L) AST (U/L) Body Liver Spleen Kidne SEQ
age Isis No. (mg BL Day Day Day BL Day Day Day (% (% (% y ID
/kg) 3 10 17 3 10 17 BL) PBS) PBS) PBS) No.
PBS n/a 32 34 37 41 62 78 76 77 104 100 100 100 n/a 0.6 32 35 38 40 53 81 74 76 104 101 112 95 0.6 33 32 35 34 70 74 75 67 101 100 130 99 0.6 39 26 37 35 63 63 77 59 100 109 109 112 0.6 30 40 34 36 58 87 54 61 104 99 120 101 0.6 35 26 33 39 56 51 51 69 104 99 110 102 Example 87: Duration of action in vivo by single closes of oligonucleotides targeting TTR comprising a Ga1NAc3 cluster ISIS numbers 420915 and 660261 (see Table 83) were tested in a single dose study for duration of action in mice. ISIS numbers 420915, 682883, and 682885 (see Table 83) were also tested in a single dose study for duration of action in mice.
Treatment Eight week old, male transgenic mice that express human TTR were each injected subcutaneously once with 100 mg/kg ISIS No. 420915 or 13.5 mg/kg ISIS No. 660261. Each treatment group consisted of 4 animals. Tail bleeds were performed before dosing to determine baseline and at days 3, 7, 10, 17, 24, and 39 following the dose. Plasma TTR protein levels were measured as described in Example 86. The results below are presented as the average percent of plasma TTR levels for each treatment group, normalized to baseline levels.
Table 88 Plasma TTR protein levels ISIS Dosage Time pointGalNAc3 CM
TTR (% baseline) SEQ
ID No.
No. (mg/kg) (days post-dose) Cluster 420915 100 n/a n/a 265 660261 13.5 Ga1NAc3-la Ad 266 Treatment Female transgenic mice that express human TTR were each injected subcutaneously once with 100 mg/kg ISIS No. 420915, 10.0 mg/kg ISIS No. 682883, or 10.0 mg/kg 682885. Each treatment group consisted of 4 animals. Tail bleeds were performed before dosing to determine baseline and at days 3, 7, 10, 17, 24, and 39 following the dose. Plasma TTR protein levels were measured as described in Example 86.
The results below are presented as the average percent of plasma TTR levels for each treatment group, normalized to baseline levels.

Table 89 Plasma TTR protein levels ISIS Dosage Time pointGalNAc3 CM
TTR (% baseline) SEQ
ID No.
No. (mg/kg) (days post-dose) Cluster 420915 100 10 48 n/a n/a 265 682883 10.0 10 38 Ga1NAc3-3a PO 265 682885 10.0 10 34 Ga1NAc3-10 a PO 265 The results in Tables 88 and 89 show that the oligonucleotides comprising a GalNAc conjugate are more potent with a longer duration of action than the parent oligonucleotide lacking a conjugate (ISIS 420915).
Example 88: Splicing modulation in vivo by oligonucleotides targeting SMN
comprising a GaINAc3 conjugate The oligonucleotides listed in Table 90 were tested for splicing modulation of human survival of motor neuron (SMN) in mice.
Table 90 Modified ASOs targeting SMN
ISIS, SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
ATTmCAmCTTTmCATAATGmCTG
387954 es es es es es es es es es es es es es es es es es es es n/a n/a 267 Ge GalNAc3-7 A- - T T C A- C T T T C A- T A-699819 Ga1NAc3-7a PO 267 TesGesmCesTesGesGe 699821Ga1NAc3-7a-0'A- - TeoTeomCeoA- CeoTeoTeoTeomC eoA- TeoA, Ga1NAc3-7a PO 267 AeoTeoGeomCeoTesGesGe 700000 eseses e AesT T mC Ae mC sTesTesTesmCesAesTesAesAesTesGesmCesTesGes s Ga1NAc3-1a Ad 268 GeoAdo.-Ga1NAc3-1a 703421 X-ATTmCAmCTTTmCATAATGmCTGG n/a n/a 703422 Ga1NAc3-7b-X-ATTmCAmCTTTmCATAATGmCTGG Ga1NAc3-7b n/a The structure of Ga1NAc3-7a was shown previously in Example 48. "X" indicates a 5' primary amine generated by Gene Tools (Philomath, OR), and Ga1NAc3-7b indicates the structure of Ga1NAc3-7a lacking the ¨NH-C6-0 portion of the linker as shown below:

HO----"(2-\----azt HN'-iC
AcHN N

H , AcHN 0/
HOOH

AcHN .
ISIS numbers 703421 and 703422 are morphlino oligonucleotides, wherein each nucleotide of the two oligonucleotides is a morpholino nucleotide.
Treatment Six week old transgenic mice that express human SMN were injected subcutaneously once with an oligonucleotide listed in Table 91 or with saline. Each treatment group consisted of 2 males and 2 females.
The mice were sacrificed 3 days following the dose to determine the liver human SMN mRNA levels both with and without exon 7 using real-time PCR according to standard protocols.
Total RNA was measured using Ribogreen reagent. The SMN mRNA levels were normalized to total mRNA, and further normalized to the averages for the saline treatment group. The resulting average ratios of SMN mRNA including exon 7 to SMN mRNA missing exon 7 are shown in Table 91. The results show that fully modified oligonucleotides that modulate splicing and comprise a GalNAc conjugate are significantly more potent in altering splicing in the liver than the parent oligonucleotides lacking a GlaNAc conjugate.
Furthermore, this trend is maintained for multiple modification chemistries, including 2'-MOE and morpholino modified oligonucleotides.
Table 91 Effect of oligonucleotides targeting human SMN in vivo ISIS Ga1NAc3 CM SEQ
Dose (mg/kg) +Exon 7 / -Exon 7 No. Cluster ID
No.
Saline n/a 1.00 n/a n/a n/a 387954 32 1.65 n/a n/a 267 387954 288 5.00 n/a n/a 267 699819 32 7.84 Ga1NAc3-7a PO 267 699821 32 7.22 Ga1NAc3-7a PO 267 700000 32 6.91 Ga1NAc3-la Ad 268 703421 32 1.27 n/a n/a 267 703422 32 4.12 Ga1NAc3-76 n/a 267 Example 89: Antisense inhibition in vivo by oligonucleotides targeting Apolipoprotein A (Apo(a)) comprising a Ga1NAc3 conjugate The oligonucleotides listed in Table 92 below were tested in a study for dose-dependent inhibition of Apo(a) in transgenic mice.
Table 92 Modified ASOs targeting Apo(a) ISISGalNAc3 SEQ ID
Sequences (5' to 3') CM
No. Cluster No.
es "-esmCes mCdsGdsTasTdsGdsGdsTasGasmC
Tes es ds 494372 n/a n/a 277 TasTesGesTesTesmCe GalNAc3-7a-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds 68 1257 Ga1NAc3-7a PO 277 TdsGdsmCds TdsTeoGeoTesTesmCe The structure of GalNAc3-7a was shown in Example 48.
Treatment Eight week old, female C57BL/6 mice (Jackson Laboratory, Bar Harbor, ME) were each injected subcutaneously once per week at a dosage shown below, for a total of six doses, with an oligonucleotide listed in Table 92 or with PBS. Each treatment group consisted of 3-4 animals.
Tail bleeds were performed the day before the first dose and weekly following each dose to determine plasma Apo(a) protein levels. The mice were sacrificed two days following the final administration. Apo(a) liver mRNA levels were determined using real-time PCR and RIBOGREENO RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) according to standard protocols. Apo(a) plasma protein levels were determined using ELISA, and liver transaminase levels were determined. The mRNA and plasma protein results in Table 93 are presented as the treatment group average percent relative to the PBS treated group. Plasma protein levels were further normalized to the baseline (BL) value for the PBS group. Average absolute transaminase levels and body weights (% relative to baseline averages) are reported in Table 94.
As illustrated in Table 93, treatment with the oligonucleotides lowered Apo(a) liver mRNA and plasma protein levels in a dose-dependent manner. Furthermore, the oligonucleotide comprising the GalNAc conjugate was significantly more potent with a longer duration of action than the parent oligonucleotide lacking a GalNAc conjugate. As illustrated in Table 94, transaminase levels and body weights were unaffected by the oligonucleotides, indicating that the oligonucleotides were well tolerated.
Table 93 Apo(a) liver mRNA and plasma protein levels ISIS Dosage Apo(a) mRNA Apo(a) plasma protein (% PBS) No. (mg/kg) (% PBS) BL Week 1 Week 2 Week 3 Week 4 Week 5 Week 6 PBS n/a 100 100 120 119 113 88 0.3 75 79 76 89 98 71 94 Table 94 ISIS No. Dosage (mg/kg) ALT (U/L) AST (U/L) Body weight (%
baseline) PBS n/a 37 54 103 0.3 30 80 104 Example 90: Antisense inhibition in vivo by oligonucleotides targeting TTR
comprising a Ga1NAc3 5 cluster Oligonucleotides listed in Table 95 below were tested in a dose-dependent study for antisense inhibition of human transthyretin (TTR) in transgenic mice that express the human TTR gene.
Treatment 10 TTR transgenic mice were each injected subcutaneously once per week for three weeks, for a total of three doses, with an oligonucleotide and dosage listed in Table 96 or with PBS. Each treatment group consisted of 4 animals. Prior to the first dose, a tail bleed was performed to determine plasma TTR protein levels at baseline (BL). The mice were sacrificed 72 hours following the final administration. TTR protein levels were measured using a clinical analyzer (AU480, Beckman Coulter, CA).
Real-time PCR and RIBOGREENO RNA quantification reagent (Molecular Probes, Inc. Eugene, OR) were used according to standard protocols to determine liver human TTR mRNA levels. The results presented in Table 96 are the average values for each treatment group. The mRNA levels are the average values relative to the average for the PBS group. Plasma protein levels are the average values relative to the average value for the PBS group at baseline. "BL" indicates baseline, measurements that were taken just prior to the first dose. As illustrated in Table 96, treatment with antisense oligonucleotides lowered TTR expression levels in a dose-dependent manner. The oligonucleotides comprising a GalNAc conjugate were more potent than the parent lacking a GalNAc conjugate (ISIS 420915), and oligonucleotides comprising a phosphodiester or deoxyadenosine cleavable moiety showed significant improvements in potency compared to the parent lacking a conjugate (see ISIS numbers 682883 and 666943 vs 420915 and see Examples 86 and 87).

Table 95 Oligonucleotides targeting human TTR
GalNAc SEQ
Isis No. Sequence 5' to 3' Linkages CM
cluster ID No.
T mC T T G Gd Td Td Ad mCd Ad I'd Gd Ad Ad 420915 es eseses es sss s ssssss Ps n/a n/a 265 AesTesmCesmCesmCe GalNAc3-3.,,TesmCeoreoreoGeoGdsrdsrdsAdsmCd Ad s682883 PS/P0 Ga1NAc3-3a PO 265 TdsGdsAdsAdsAeoTommm *_es *_es e GalNAC3-3a-0,AdorresmCeorreoTeoGeoGdsTdsTdsAds 666943 PS/P0 GalNAc3-3a Ad 269 mCdsAdsTdsGdsAdsAds AeoTeomCesmCesmCe GalNAC3-7a-0,AdorresmCeorreoTeoGeoGdsTdsTdsAds 682887 PS/P0 GalNAc3-7a Ad 269 mCdsAdsTdsGdsAdsAdsAeoTeomCesmCesmCe GalNAc3-10a_0,AdorresmCeorreorreoGeoGdsrrdsTasAd 682888 s PS/PO GalNAc3-10 a Ad 269 mCdsAdsTdsGdsAdsAdsAeoTeomCesmCesmCe GalNAc3-13a_0,AdoTesmCeorreorreoGeoGdsrrdsTasAd 682889 s PS/P0 GalNAc3-13a Ad 269 mCdsAdsTdsGdsAdsAdsAeoTeomCesmCesmCe The legend for Table 95 can be found in Example 74. The structure of Ga1NAc3-3a was shown in Example 39. The structure of Ga1NAc3-7a was shown in Example 48. The structure of Ga1NAc3-10a was shown in Example 46. The structure of GalNAc3-13a was shown in Example 62.
Table 96 Antisense inhibition of human TTR in vivo Isis No. Dosage (mg/kg) TTR mRNA (% PBS) TTR protein (% BL) GalNAc cluster CM
PBS n/a 100 124 n/a n/a 420915 20 71 86 n/a n/a 0.6 61 73 682883 2 23 36 Ga1NAc3-3a PO

0.6 74 93 666943 2 33 57 Ga1NAc3-3a Ad 0.6 60 97 682887 2 36 49 Ga1NAc3-7a Ad 0.6 65 92 682888 2 32 46 Ga1NAc3-10 a Ad 0.6 72 74 Ga1NAc3-13a Ad Example 91: Antisense inhibition in vivo by oligonucleotides targeting Factor VII comprising a Ga1NAc3 conjugate in non-human primates Oligonucleotides listed in Table 97 below were tested in a non-terminal, dose escalation study for antisense inhibition of Factor VII in monkeys.
Treatment Non-naïve monkeys were each injected subcutaneously on days 0, 15, and 29 with escalating doses of an oligonucleotide listed in Table 97 or with PBS. Each treatment group consisted of 4 males and 1 female. Prior to the first dose and at various time points thereafter, blood draws were performed to determine plasma Factor VII protein levels. Factor VII protein levels were measured by ELISA. The results presented in Table 98 are the average values for each treatment group relative to the average value for the PBS group at baseline (BL), the measurements taken just prior to the first dose. As illustrated in Table 98, treatment with antisense oligonucleotides lowered Factor VII expression levels in a dose-dependent manner, and the oligonucleotide comprising the GalNAc conjugate was significantly more potent in monkeys compared to the oligonucleotide lacking a GalNAc conjugate.
Table 97 Oligonucleotides targeting Factor VII
GalNAc SEQ
Isis No. Sequence 5 to 3' Linkages CM
cluster ID No.
A T G mC A Td Gd Gd Td Gd Ad Td Gd mCd Td 407935 eseses esesssssssss ss Ps n/a n/a TesmCesTesGesAe GalNAc3-10.-0,AesTesGesmCesAesTdsGdsGdsrrdsGds 686892 PS GalNAc3-10a PO 270 ikdsTdsGdsmCdsTds TesmCesTesGesAe The legend for Table 97 can be found in Example 74. The structure of Ga1NAc3-10a was shown in Example 46.
Table 98 Factor VII plasma protein levels ISIS No. Day Dose (mg/kg) Factor VII
(% BL) 0 n/a 100 22 n/a 92 36 n/a 46 43 n/a 43 22 n/a 29 36 n/a 15 43 n/a 11 Example 92: Antisense inhibition in primary hepatocytes by antisense oligonucleotides targeting Apo-CIII comprising a Ga1NAc3 conjugate Primary mouse hepatocytes were seeded in 96-well plates at 15,000 cells per well, and the oligonucleotides listed in Table 99, targeting mouse ApoC-III, were added at 0.46, 1.37, 4.12, or 12.35, 37.04, 111.11, or 333.33 nM or 1.00 [LM. After incubation with the oligonucleotides for 24 hours, the cells were lysed and total RNA was purified using RNeasy (Qiagen). ApoC-III mRNA
levels were determined using real-time PCR and RIBOGREENO RNA quantification reagent (Molecular Probes, Inc.) according to standard protocols. IC50 values were determined using Prism 4 software (GraphPad). The results show that regardless of whether the cleavable moiety was a phosphodiester or a phosphodiester-linked deoxyadensoine, the oligonucleotides comprising a GalNAc conjugate were significantly more potent than the parent oligonucleotide lacking a conjugate.
Table 99 Inhibition of mouse APOC-III expression in mouse primary hepatocytes ISIS,IC50 SEQ
Sequence (5 to 3') CM
No. (nM) ID No.
440670 mCesAesGesmCesTesTdsTdsAdsTdsTdsAdsGdsGdsGdsAdsmCesAesGesmCesAe n/a 13.20 271 mC A G MC rf rfd rfd Ad rfd rfd Ad Gd Gd Gd Ad mC
661180 ese;ies eses ss sss s s s s s es Ad 1.40 272 AesGes CesAeo Ado'-GalNAc3-la GallsTAc3-3.,mCesAesGesmCesTesTasTasAdsrfasTdsAdsGdsGdsGdsAdsmC
680771 es PO 0.70 271 /kesGesmCesAe GallsTAc3-7.,mCesAesGesmCesTesTdsrrdsAdsrrdsrrdsAdsGdsGdsGdsAdsmC
680772 es PO 1.70 271 /kesGesmCesAe GallsTAc3-10.,lliCesAesGesmCesrf esrf dsrrdsAdsrrdsrrdsAdsGdsGdsGdsAdsniC
680773 es PO 2.00 271 /kesGesmCesAe GallsTAc3-13.,mCesAesGesmCesrresTdsrrdsAdsrrdsrrdsAdsGdsGdsGdsAdsniC
680774 es PO 1.50 271 /kesGesmCesAe GallsTAc3-3.,InCesAeoGeomCeoTeoTdsTdsAdsrfasrfdsAdsGdsGdsGdsAdsmCe PO < 0.46 271 AeoGesmCesAe GalNAC3-3a-0,AdomCesAesGesmCesTesTdsTdsAdsrrdsrrdSAdSGdSGdSGdAds A 1. 10 681273 A r, A
d mues.L%esu esmk,si-ve 683733m m-Arfrf AGA mCeses es Ceses d d d d dd d d d d C
es Ad 2.50 272 AesGesmCesAeoAdo' GalNAc3-19a The structure of Ga1NAc3-1 a was shown previously in Example 9, Ga1NAc3-31 was shown in Example 39, Ga1NAc3-71 was shown in Example 48, Ga1NAc3-101 was shown in Example 46, Ga1NAc3-131 was shown in Example 62, and Ga1NAc3-191 was shown in Example 70.
Example 93: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising mixed wings and a 5'-Ga1NAc3 conjugate The oligonucleotides listed in Table 100 were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.

Table 100 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') Ga1NAc3 CM SEQ
No. Cluster ID
No.
449093 TkaTkamCkaAdaGdaTdamCds Ads 'r ds Gds AdsmCdsTasrilsmCksmCk n/a n/a 274 699806 Ga1NAc3-3a-0,TI,TIsmCksAdsGasTdsmCds AdsTds GdsAdsmCds Ga1NAc3-3 a PO

m TdsTississmCk 699807 Ga1NAc3- 7a-0,TkaTkamCksAdsGdaTdsmCds AdsTds GdsAdsmCds Ga1NAc3-7 a PO

m TdsTississmCk 699809 Ga1NAc3-7a-o, rri.srrIsmCksAdsGdsrrdsmCds AdsTds Gds AdsmCds Ga1NAc3-7a PO

m-TdaTeaesmCe 699811 Ga1NAc3-7a-0,TeaTeamCesAdsGdarrdsmCds AdsTds GdsAdsmCds Ga1NAc3-7a PO

m TdsTississmCk 699813 Ga1NAc3-7a-o'TksTdsmCksAdsGdsTdsmCds AdsTds GdsAdsmCds Ga1NAc3-7a PO

m-TdsTissdsmCk 699815 Ga1NAc3-7a-0,TesTIsmCksAdsGdsTdsmCds AdsTds GdsAdsmCds Ga1NAc3-7 a PO

m TdsTississmCe The structure of Ga1NAc3-3a was shown previously in Example 39, and the structure of Ga1NAc3-7a was shown previously in Example 48. Subscripts: "e" indicates 2'-MOE modified nucleoside; "d" indicates [3-D-2'-deoxyribonucleoside; "k" indicates 6'-(S)-CH3 bicyclic nucleoside (cEt);
"s" indicates phosphorothioate internucleoside linkages (PS); "o" indicates phosphodiester internucleoside linkages (PO). Supersript "m"
indicates 5-methylcytosines.
Treatment Six to eight week old C57BL/6 mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once at the dosage shown below with an oligonucleotide listed in Table 100 or with saline.
Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration. Liver SRB-1 mRNA levels were measured using real-time PCR. SRB-1 mRNA levels were normalized to cyclophilin mRNA levels according to standard protocols. The results are presented as the average percent of SRB-1 mRNA levels for each treatment group relative to the saline control group. As illustrated in Table 101, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner, and the gapmer oligonucleotides comprising a GalNAc conjugate and having wings that were either full cEt or mixed sugar modifications were significantly more potent than the parent oligonucleotide lacking a conjugate and comprising full cEt modified wings.
Body weights, liver transaminases, total bilirubin, and BUN were also measured, and the average values for each treatment group are shown in Table 101. Body weight is shown as the average percent body weight relative to the baseline body weight (% BL) measured just prior to the oligonucleotide dose.

Table 101 SRB-1 mRNA, ALT, AST, BUN, and total bilirubin levels and body weights ISIS Dosage SRB-1 mRNA ALT ASTBil BUN Body weight No. (mg/kg) (% PBS) (U/L) (U/L) (% BL) PBS n/a 100 31 84 0.15 28 102 1 111 18 48 0.17 31 104 449093 3 94 20 43 0.15 26 103 36 19 50 0.12 29 104 0.1 114 23 58 0.13 26 107 699806 0.3 59 21 45 0.12 27 108 1 25 30 61 0.12 30 104 0.1 121 19 41 0.14 25 100 699807 0.3 73 23 56 0.13 26 105 1 24 22 69 0.14 25 102 0.1 125 23 57 0.14 26 104 699809 0.3 70 20 49 0.10 25 105 1 33 34 62 0.17 25 107 0.1 123 48 77 0.14 24 106 699811 0.3 94 20 45 0.13 25 101 1 66 57 104 0.14 24 107 0.1 95 20 58 0.13 28 104 699813 0.3 98 22 61 0.17 28 105 1 49 19 47 0.11 27 106 0.1 93 30 79 0.17 25 105 699815 0.3 64 30 61 0.12 26 105 1 24 18 41 0.14 25 106 Example 94: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising 2'-sugar 5 modifications and a 5'-GaINAc3 conjugate The oligonucleotides listed in Table 102 were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.
Table 102 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') Ga1NAc3 CM SEQ
No. Cluster ID No.
353382 GesmCesTesTesmCesikasGasTasmCdsAds'r dsGdsAdsmCdsTdsTesmCesmCes n/a n/a TesTe 700989 GmsCmsUmsUmsCmsAdsGasTasmCdsAdsTdsGdsAdsmCdsTdsUmsCmsCms 1" n/a UmsUm 666904 Ga1NAc3-3a-o, GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds Ga1NAc3-3a PO

mCdsTdsTesmCesmCesTesTe 700991 Ga1NAc3-7.70,GmsCinsUinsUmsCinsAdsGdsTdsmCdsAdsTdsGds Ga1NAc3-7a PO

AdsmCdsTdsUmsCmsCmsUmsUm 10 Subscript "m" indicates a 2'-0-methyl modified nucleoside. See Example 74 for complete table legend. The structure of Ga1NAc3-31 was shown previously in Example 39, and the structure of Ga1NAc3-7a was shown previously in Example 48.

Treatment The study was completed using the protocol described in Example 93. Results are shown in Table 103 below and show that both the 2'-MOE and 2'-0Me modified oligonucleotides comprising a GalNAc conjugate were significantly more potent than the respective parent oligonucleotides lacking a conjugate. The results of the body weights, liver transaminases, total bilirubin, and BUN
measurements indicated that the compounds were all well tolerated.
Table 103 SRB-1 mRNA
ISIS No. Dosage (mg/kg) SRB-1 mRNA (% PBS) PBS n/a 100 Example 95: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising bicyclic nucleosides and a 5'-Ga1NAc3 conjugate The oligonucleotides listed in Table 104 were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.
Table 104 Modified ASOs targeting SRB-1 ISIS, SEQ
Sequences (5' to 3') CM
No. Cluster ID No 440762 IlsmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTIsmCk n/a n/a 666905 GaINAc3-3a-0,TIsmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTIsmCk Ga1NAc3-3a 699782 Ga1NAc3-7a-0,TIsmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTIsmCk Ga1NAc3-7a 699783 Ga1NAc3-39-0,T1smCisAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTlsmC1 Ga1NAc3-3a 653621 TiamCisAdsGasTasmCdsAdsTdsGdsAdsmCdsTdsrrlsmCloAdo,-GalNAC3-19 Ga1NAc3-1a Ad 247 439879 T,InC,AdaGdaTdamCdaAdaTa GdsAdsmCdsTdsT,mCg n/a n/a 699789 Ga1NAc3-39-0,TgsmC gsAdsGdsTdsmC dsAdsTa GdsAdsmCdsTdsT,mC g Ga1NAc3-3a PO 246 Subscript "g" indicates a fluoro-HNA nucleoside, subscript "1" indicates a locked nucleoside comprising a 2'-0-CH2-4' bridge. See the Example 74 table legend for other abbreviations. The structure of Ga1NAc3-1a was shown previously in Example 9, the structure of Ga1NAc3-3a was shown previously in Example 39, and the structure of Ga1NAc3-7a was shown previously in Example 48.
Treatment The study was completed using the protocol described in Example 93. Results are shown in Table 105 below and show that oligonucleotides comprising a GalNAc conjugate and various bicyclic nucleoside modifications were significantly more potent than the parent oligonucleotide lacking a conjugate and comprising bicyclic nucleoside modifications. Furthermore, the oligonucleotide comprising a GalNAc conjugate and fluoro-HNA modifications was significantly more potent than the parent lacking a conjugate and comprising fluoro-HNA modifications. The results of the body weights, liver transaminases, total bilirubin, and BUN measurements indicated that the compounds were all well tolerated.
Table 105 SRB-1 mRNA, ALT, AST, BUN, and total bilirubin levels and body weights ISIS No. Dosage (mg/kg) SRB-1 mRNA (% PBS) PBS n/a 100 0.1 105 666905 0.3 56 0.1 93 699782 0.3 63 0.1 105 699783 0.3 53 0.1 109 653621 0.3 82 0.1 82 699789 0.3 69 Example 96: Plasma protein binding of antisense oligonucleotides comprising a GaINAc3 conjugate group Oligonucleotides listed in Table 70 targeting ApoC-III and oligonucleotides in Table 106 targeting Apo(a) were tested in an ultra-filtration assay in order to assess plasma protein binding.

Table 106 Modified oligonucleotides targeting Apo(a) ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No T G mCesTesmCesmCdsGdsTasTasGasGasTasGasmCdsTasTesGesTes 494372 mesmc,es n/a n/a 1 es µ=-e TesGeomCeoTeomCeomCdsGdsTasTdsGdsGdsTdsGdsmCdsTasTeoGeorres 693401 n/a n/a TesmCe GalNAc3-7a-0,TesGesmCesTesmCesmCdsGdsTdsTdsGdsGdsTdsGdsmCds GalNAc3-7a PO 277 TdsTesGesTesTesmCe GalNAC3-7a-o'TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGdsTdsGdsmCds GalNAc3-7a PO 277 TdsTeoGeoTesTesmCe See the Example 74 for table legend. The structure of Ga1NAc3-7a was shown previously in Example 48.
Ultrafree-MC ultrafiltration units (30,000 NMWL, low-binding regenerated cellulose membrane, Millipore, Bedford, MA) were pre-conditioned with 300 [tt of 0.5% Tween 80 and centrifuged at 2000 g for minutes, then with 300 L of a 300 [tg/mL solution of a control oligonucleotide in H20 and centrifuged at 2000 g for 16 minutes. In order to assess non-specific binding to the filters of each test oligonucleotide from Tables 70 and 106 to be used in the studies, 300 [tt of a 250 ng/mL solution of oligonucleotide in H20 at pH
7.4 was placed in the pre-conditioned filters and centrifuged at 2000 g for 16 minutes. The unfiltered and 10 filtered samples were analyzed by an ELISA assay to determine the oligonucleotide concentrations. Three replicates were used to obtain an average concentration for each sample. The average concentration of the filtered sample relative to the unfiltered sample is used to determine the percent of oligonucleotide that is recovered through the filter in the absence of plasma (% recovery).
Frozen whole plasma samples collected in K3-EDTA from normal, drug-free human volunteers, cynomolgus monkeys, and CD-1 mice, were purchased from Bioreclamation LLC
(Westbury, NY). The test oligonucleotides were added to 1.2 mL aliquots of plasma at two concentrations (5 and 150 [tg/mL). An aliquot (300 [tt) of each spiked plasma sample was placed in a pre-conditioned filter unit and incubated at 37 C for 30 minutes, immediately followed by centrifugation at 2000 g for 16 minutes. Aliquots of filtered and unfiltered spiked plasma samples were analyzed by an ELISA to determine the oligonucleotide concentration in each sample. Three replicates per concentration were used to determine the average percentage of bound and unbound oligonucleotide in each sample. The average concentration of the filtered sample relative to the concentration of the unfiltered sample is used to determine the percent of oligonucleotide in the plasma that is not bound to plasma proteins (%
unbound). The final unbound oligonucleotide values are corrected for non-specific binding by dividing the % unbound by the % recovery for each oligonucleotide. The final % bound oligonucleotide values are determined by subtracting the final %
unbound values from 100. The results are shown in Table 107 for the two concentrations of oligonucleotide tested (5 and 150 [tg/mL) in each species of plasma. The results show that GalNAc conjugate groups do not have a significant impact on plasma protein binding. Furthermore, oligonucleotides with full PS

internucleoside linkages and mixed PO/PS linkages both bind plasma proteins, and those with full PS
linkages bind plasma proteins to a somewhat greater extent than those with mixed PO/PS linkages.
Table 107 Percent of modified oligonucleotide bound to plasma proteins ISIS Human plasma Monkey plasma Mouse plasma No. 5 ug/mL 150 ug/mL 5 ug/mL 150 ug/mL 5 ug/mL 150 ug/mL
304801 99.2 98.0 99.8 99.5 98.1 97.2 663083 97.8 90.9 99.3 99.3 96.5 93.0 674450 96.2 97.0 98.6 94.4 94.6 89.3 494372 94.1 89.3 98.9 97.5 97.2 93.6 693401 93.6 89.9 96.7 92.0 94.6 90.2 681251 95.4 93.9 99.1 98.2 97.8 96.1 681257 93.4 90.5 97.6 93.7 95.6 92.7 Example 97: Modified oligonucleotides targeting TTR comprising a GaINAc3 conjugate group The oligonucleotides shown in Table 108 comprising a GalNAc conjugate were designed to target TTR.
Table 108 Modified oligonucleotides targeting TTR
Ga1NAc3 SEQ ID
ISIS No. Sequences (5' to 3') CM
Cluster No Ga1NAc3-3a_0,Ado Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds GalNAc3-3 Ad Ads Tds Gds Ads Ads Aes Tes mCes mCes mCe Tes mCeemTee mTeo Gmeo Gds Tds Tds Ads mCds Ads Tds Gds Ads Ads 666942 GalNAc3-1 Ad 266 Aeo Teo Ces Ces Ceo Ado'-GalNAc3-3a Ga1NAc3-3a_0,Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ads Tds GalNAc3-3 682876 , A A Aes Tes ,-, ,-, ,-, uds /Ads ltds i-les mCes mk-es nik-,e Ga1NAc3-7a_0,Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ads Tds GalNAc3-7 682877 , A A Aes Tes ,-., r, ,-.õ PO

uds /Ads ltds i-les mCes mk-es nik-,e GalNAC3-10a_0,Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ad 682878 s Ga1NAc3-10 PO 265 Tds Gds Ads Ads Aes Tes mCes mCes mCe GalNAc3-13a_0,Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ad 682879 s Ga1NAc3-13 PO 265 Tds Gds Ads Ads Aes Tes mCes mCes mCe Ga1NAC3-7a-0,Ado Tes mCes Tes Tes Ges Gds Tds Tds Ads mCdsGalNAc3-7 Ad Ads Tds Gds Ads Ads Aes Tes mCes mCes mCe Ga1NAC3-10a_0,Ado Tes mCes Tes Tes Ges Gds Tds Tds Ads mCdsGalNAc3-10 Ad Ads Tds Gds Ads Ads Aes Tes mCes mCes mCe Ga1NAC3-13a_0,Ado Tes mCes Tes Tes Ges Gds Tds Tds Ads mCdsGalNAc3-13 Ad Ads Tds Gds Ads Ads Aes Tes mCes mCes mCe Tes mCes Tes mTes Ges Gds Tds Tds Ads mCds Ads Tds Gds Ads Ads 684056 Ga1NAc3-19 Ad 266 Aes Tes mCes Ces Ceo Ado'-GalNAc3-19a The legend for Table 108 can be found in Example 74. The structure of Ga1NAc3-1 was shown in Example 9.
The structure of Ga1NAc3-3a was shown in Example 39. The structure of Ga1NAc3-7a was shown in Example 48. The structure of Ga1NAc3-10a was shown in Example 46. The structure of Ga1NAc3-13a was shown in Example 62. The structure of GalNAc3-19a was shown in Example 70.

Example 98: Evaluation of pro-inflammatory effects of oligonucleotides comprising a GaINAc conjugate in hPMBC assay The oligonucleotides listed in Table 109 and were tested for pro-inflammatory effects in an hPMBC
assay as described in Examples 23 and 24. (See Tables 30, 83, 95, and 108 for descriptions of the oligonucleotides.) ISIS 353512 is a high responder used as a positive control, and the other oligonucleotides are described in Tables 83, 95, and 108. The results shown in Table 109 were obtained using blood from one volunteer donor. The results show that the oligonucleotides comprising mixed PO/PS internucleoside linkages produced significantly lower pro-inflammatory responses compared to the same oligonucleotides having full PS linkages. Furthermore, the GalNAc conjugate group did not have a significant effect in this assay.
Table 109 ISIS No. Einax/EC50 Ga1NAc3 cluster Linkages CM
353512 3630 n/a PS n/a 420915 802 n/a PS n/a 682881 1311 Ga1NAc3-10 PS Ad 682888 0.26 Ga1NAc3-10 PO/PS Ad 684057 1.03 Ga1NAc3-19 PO/PS Ad Example 99: Binding affinities of oligonucleotides comprising a GaINAc conjugate for the asialoglycoprotein receptor The binding affinities of the oligonucleotides listed in Table 110 (see Table 76 for descriptions of the oligonucleotides) for the asialoglycoprotein receptor were tested in a competitive receptor binding assay. The competitor ligand, al-acid glycoprotein (AGP), was incubated in 50 mM sodium acetate buffer (pH 5) with 1 U neuraminidase-agarose for 16 hours at 37 C, and > 90% desialylation was confirmed by either sialic acid assay or size exclusion chromatography (SEC). Iodine monochloride was used to iodinate the AGP according to the procedure by Atsma et al. (see J Lipid Res. 1991 Jan; 32(1):173-81.) In this method, desialylated al -acid glycoprotein (de-AGP) was added to 10 mM iodine chloride, Na1251, and 1 M
glycine in 0.25 M NaOH.
After incubation for 10 minutes at room temperature, 1251 -labeled de-AGP was separated from free 1251 by concentrating the mixture twice utilizing a 3 KDMWCO spin column. The protein was tested for labeling efficiency and purity on a HPLC system equipped with an Agilent SEC-3 column (7.8x300mm) and a 13-RAM 1251 counter. Competition experiments utilizing -labeled de-AGP and various GalNAc-cluster containing ASOs were performed as follows. Human HepG2 cells (106 cells/m1) were plated on 6-well plates in 2 ml of appropriate growth media. MEM media supplemented with 10% fetal bovine serum (FBS), 2 mM
L-Glutamine and 10mM HEPES was used. Cells were incubated 16-20 hours @ 37 C
with 5% and 10% CO2 respectively. Cells were washed with media without FBS prior to the experiment. Cells were incubated for 30 min @37 C with lml competition mix containing appropriate growth media with 2%
FBS, 10-8 M 1251 _ labeled de-AGP and GalNAc-cluster containing ASOs at concentrations ranging from 10-11 to 10-5 M. Non-specific binding was determined in the presence of 10-2 M GalNAc sugar. Cells were washed twice with media without FBS to remove unbound 1251 -labeled de-AGP and competitor GalNAc ASO. Cells were lysed using Qiagen's RLT buffer containing 1% 13-mercaptoethanol. Lysates were transferred to round bottom assay tubes after a brief 10 min freeze/thaw cycle and assayed on a y-counter.
Non-specific binding was subtracted before dividing 1251 protein counts by the value of the lowest GalNAc-ASO concentration counts.
The inhibition curves were fitted according to a single site competition binding equation using a nonlinear regression algorithm to calculate the binding affinities (KD's).
The results in Table 110 were obtained from experiments performed on five different days. Results for oligonucleotides marked with superscript "a" are the average of experiments run on two different days.
The results show that the oligonucleotides comprising a GalNAc conjugate group on the 5'-end bound the asialoglycoprotein receptor on human HepG2 cells with 1.5 to 16-fold greater affinity than the oligonucleotides comprising a GalNAc conjugate group on the 3'-end.
Table 110 Asialoglycoprotein receptor binding assay results Oligonucleotide end to ISIS No. GalNAc conjugate which GalNAc conjugate KD (nM) is attached 661161' Ga1NAc3-3 5' 3.7 666881' Ga1NAc3-10 5' 7.6 666981 Ga1NAc3-7 5' 6.0 670061 Ga1NAc3-13 5' 7.4 655861' Ga1NAc3-1 3' 11.6 677841' Ga1NAc3-19 3' 60.8 Example 100: Antisense inhibition in vivo by oligonucleotides comprising a GaINAc conjugate group targeting Apo(a) in vivo The oligonucleotides listed in Table 111a below were tested in a single dose study for duration of action in mice.
Table 111a Modified ASOs targeting APO(a) ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID
No.
GalNAc3-7a-0,TesGesmCesTesmCesmCdsGasTasTasGasGas Ga1NAc3-7a PO 277 TdsGdsmCdsTdsTesGes TesTesmCe 681257 Ga1NAC3-7a-o'TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds Ga1NAc3-7a PO 277 m-TdsGasdsTasTe0GeeTesTesmCe The structure of GalNAc3-7a was shown in Example 48.
Treatment Female transgenic mice that express human Apo(a) were each injected subcutaneously once per week, for a total of 6 doses, with an oligonucleotide and dosage listed in Table 111b or with PBS. Each treatment group consisted of 3 animals. Blood was drawn the day before dosing to determine baseline levels of Apo(a) protein in plasma and at 72 hours, 1 week, and 2 weeks following the first dose. Additional blood draws will occur at 3 weeks, 4 weeks, 5 weeks, and 6 weeks following the first dose. Plasma Apo(a) protein levels were measured using an ELISA. The results in Table 111b are presented as the average percent of plasma Apo(a) protein levels for each treatment group, normalized to baseline levels (% BL), The results show that the oligonucleotides comprising a GalNAc conjugate group exhibited potent reduction in Apo(a) expression. This potent effect was observed for the oligonucleotide that comprises full PS internucleoside linkages and the oligonucleotide that comprises mixed PO and PS linkages.
Table 111b Apo(a) plasma protein levels ISIS N Apo(a) at 72 hours Apo(a) at 1 week Apo(a) at 3 weeks o. Dosage (mg/kg) (% BL) (% BL) (% BL) PBS n/a 116 104 107 0.3 97 108 93 681251 1.0 85 77 57 3.0 54 49 11 10.0 23 15 4 0.3 114 138 104 681257 1.0 91 98 54 3.0 69 40 6 10.0 30 21 4 Example 101: Antisense inhibition by oligonucleotides comprising a GaINAc cluster linked via a stable moiety The oligonucleotides listed in Table 112 were tested for inhibition of mouse APOC-III expression in vivo. C57B1/6 mice were each injected subcutaneously once with an oligonucleotide listed in Table 112 or with PBS. Each treatment group consisted of 4 animals. Each mouse treated with ISIS 440670 received a dose of 2, 6, 20, or 60 mg/kg. Each mouse treated with ISIS 680772 or 696847 received 0.6, 2, 6, or 20 mg/kg. The GalNAc conjugate group of ISIS 696847 is linked via a stable moiety, a phosphorothioate linkage instead of a readily cleavable phosphodiester containing linkage. The animals were sacrificed 72 hours after the dose. Liver APOC-III mRNA levels were measured using real-time PCR. APOC-III mRNA
levels were normalized to cyclophilin mRNA levels according to standard protocols. The results are presented in Table 112 as the average percent of APOC-III mRNA levels for each treatment group relative to the saline control group. The results show that the oligonucleotides comprising a GalNAc conjugate group were significantly more potent than the oligonucleotide lacking a conjugate group. Furthermore, the oligonucleotide comprising a GalNAc conjugate group linked to the oligonucleotide via a cleavable moiety (ISIS 680772) was even more potent than the oligonucleotide comprising a GalNAc conjugate group linked to the oligonucleotide via a stable moiety (ISIS 696847).
Table 112 Modified oligonucleotides targeting mouse APOC-III
Dosage APOC-III
ISIS
SEQ
N Sequences (5' to 3') CM (mg/kg) mRNA (%
No.
ID
o.
PBS) mCesAesGesmCesTes'r dsTdsAdsTdsTdsAds 6 86 440670 n/a GdsGdsGdsAdsmCes AesGes mCesAe 20 59 0.6 79 GalNAc3-7._0,mCesAesGesmCesTesTdsrrdsAds 2 58 TdsTdsAdsGds GdsGdsAdsmCes /kesGesmCesAe 6 31 0.6 83 Ga1NAc3-7,_s,mCesAesGesmCesTesTdsrrdsAdsrrds n/a (PS 2 7 ) TdsAdsGdsGdsGdsAdsmCesikesGesmCesAe 6 40 The structure of GalNAc3-7a was shown in Example 48.
Example 102: Distribution in liver of antisense oligonucleotides comprising a GaINAc conjugate The liver distribution of ISIS 353382 (see Table 36) that does not comprise a GalNAc conjugate and ISIS 655861 (see Table 36) that does comprise a GalNAc conjugate was evaluated. Male balb/c mice were subcutaneously injected once with ISIS 353382 or 655861 at a dosage listed in Table 113. Each treatment group consisted of 3 animals except for the 18 mg/kg group for ISIS 655861, which consisted of 2 animals.
The animals were sacrificed 48 hours following the dose to determine the liver distribution of the oligonucleotides. In order to measure the number of antisense oligonucleotide molecules per cell, a Ruthenium (II) tris-bipyridine tag (MSD TAG, Meso Scale Discovery) was conjugated to an oligonucleotide probe used to detect the antisense oligonucleotides. The results presented in Table 113 are the average concentrations of oligonucleotide for each treatment group in units of millions of oligonucleotide molecules per cell. The results show that at equivalent doses, the oligonucleotide comprising a GalNAc conjugate was present at higher concentrations in the total liver and in hepatocytes than the oligonucleotide that does not comprise a GalNAc conjugate. Furthermore, the oligonucleotide comprising a GalNAc conjugate was present at lower concentrations in non-parenchymal liver cells than the oligonucleotide that does not comprise a GalNAc conjugate. And while the concentrations of ISIS 655861 in hepatocytes and non-parenchymal liver cells were similar per cell, the liver is approximately 80% hepatocytes by volume. Thus, the majority of the ISIS 655861 oligonucleotide that was present in the liver was found in hepatocytes, whereas the majority of the ISIS 353382 oligonucleotide that was present in the liver was found in non-parenchymal liver cells.
Table 113 Concentration in whole Concentration in Concentration in non-ISIS Dosage liver (molecules*10^6 hepatocytes parenchymal liver cells No. (mg/kg) per cell) (molecules*10^6 per cell) (molecules*10^6 per cell) 3 9.7 1.2 37.2 17.3 4.5 34.0 23.6 6.6 65.6 29.1 11.7 80.0 60 73.4 14.8 98.0 90 89.6 18.5 119.9 0.5 2.6 2.9 3.2 1 6.2 7.0 8.8 655861 3 19.1 25.1 28.5 6 44.1 48.7 55.0
18 76.6 82.3 77.1 Example 103: Duration of action in vivo of oligonucleotides targeting APOC-III
comprising a GaINAc3 conjugate The oligonucleotides listed in Table 114 below were tested in a single dose study for duration of 10 action in mice.
Table 114 Modified ASOs targeting APOC-III
ISIS Sequences (5' to 3') Ga1NAc3 CM SEQ
No. Cluster ID No.
304801 AesGesmCesTesTesmCdsTdsTdsGdsTdsmCdsmCdsAdsGdsmCdsTesTes n/a TesAesTe 663084 Ga1NAc3-3.-0,AdoAesGeomCeoTeoTeomCdsTdsTdsGdsTdsmCds Ga1NAc3-3a Ad mCdsAdsGdsmCdsTeoTeo TesAesTe 679241 AesGeomCeoTeoTeomCdsTdsTdsGdsTdsmCdsmCdsAdsGdsmCdsTeoTeo Ga1NAc3-19a Ad TesAesTeoAdo'-GaINAc3-19.
The structure of GalNAc3-3a was shown in Example 39, and Ga1NAc3-19a was shown in Example 70.
Treatment Female transgenic mice that express human APOC-III were each injected subcutaneously once with an oligonucleotide listed in Table 114 or with PBS. Each treatment group consisted of 3 animals. Blood was drawn before dosing to determine baseline and at 3, 7, 14, 21, 28, 35, and 42 days following the dose. Plasma triglyceride and APOC-III protein levels were measured as described in Example 20. The results in Table 115 are presented as the average percent of plasma triglyceride and APOC-III
levels for each treatment group, normalized to baseline levels. A comparison of the results in Table 71 of example 79 with the results in Table 115 below show that oligonucleotides comprising a mixture of phosphodiester and phosphorothioate internucleoside linkages exhibited increased duration of action than equivalent oligonucleotides comprising only phosphorothioate internucleoside linkages.
Table 115 Plasma triglyceride and APOC-III protein levels in transgenic mice Time pointAPOC-III
ISIS Dosage TriglyceridesGa1NAc3 CM
(days post- protein (%
No. (mg/kg) (% baseline)Cluster dose) baseline) PBS n/a 21 69 92 n/a n/a 304801 30 21 67 81 n/a n/a 663084 10 21 23 29 Ga1NAc3-3a Ad -679241 10 21 36 34 Ga1NAc3Ad
19 28 48 34 a Example 104: Synthesis of oligonucleotides comprising a 5'-Ga1NAc2 conjugate HN,Boc HN-B(Dc Boc.N OH H2No 0 ____________________ H HBTU, HOBt DIEA, DMF N'''' Boc.Nj.o 0 H TFA
¨1/0-DCM

120 126 85% 231 \r 101 +

Ac0E--:1-0.,,,,,,....,õ-.,,,,,,,ko 010 F DIEA
0 AcHN F DMF

OAc/.-- OAc 0 OAc OAc ----Z-0, Cd AcHN NH .õ.^......,...õ..,4, AcHN NH
1 H2, Pd/C, Me0H
____________________________________________ lb-F
OAc 1 f- OAc 2 PFPTFA DMF OAc OAc , F 0 F
0 0 OAc L H 0 Ac0-1:--D-1-0..õõ,----.....AN NH õ,_.....õ,-...,,Ao 0 AcHN AcHN
F

F

0 83e OHOH
3' 5, II 1--0 0 ( OLIGO )-0-P-0-(CH2)6-NH2 __ HO
I AcHN NH
OH
1 Borate buffer, DMSO, pH 8.5, rt OHOH
HO / --1-0....õ,õ--,..õ..11...N N E11.....õ-,....¨õIl.
2 aq ammonia, rt AcHN N.----W-0.47q¨FIG7 Compound 120 is commercially available, and the synthesis of compound 126 is described in Example 49. Compound 120 (1 g, 2.89 mmol), HBTU (0.39 g, 2.89 mmol), and HOBt (1.64 g, 4.33 mmol) were dissolved in DMF (10 mL. and N,N-diisopropylethylamine (1.75 mL, 10.1 mmol) were added. After about 5 min, aminohexanoic acid benzyl ester (1.36 g, 3.46 mmol) was added to the reaction. After 3h, the reaction mixture was poured into 100 mL of 1 M NaHSO4 and extracted with 2 x 50 mL ethyl acetate.
Organic layers were combined and washed with 3 x 40 mL sat NaHCO3 and 2 x brine, dried with Na2SO4, filtered and concentrated. The product was purified by silica gel column chromatography (DCM:EA:Hex , 1:1:1) to yield compound 231. LCMS and NMR were consistent with the structure.
Compounds 231 (1.34 g, 2.438 mmol) was dissolved in dichloromethane (10 mL) and trifluoracetic acid (10 mL) was added. After stirring at room temperature for 2h, the reaction mixture was concentrated under reduced pressure and co-evaporated with toluene ( 3 x 10 mL). The residue was dried under reduced pressure to yield compound 232 as the trifuloracetate salt. The synthesis of compound 166 is described in Example 54. Compound 166 (3.39 g, 5.40 mmol) was dissolved in DMF (3 mL). A solution of compound 232 (1.3 g, 2.25 mmol) was dissolved in DMF (3 mL) and N,N-diisopropylethylamine (1.55 mL) was added. The reaction was stirred at room temperature for 30 minutes, then poured into water (80 mL) and the aqueous layer was extracted with Et0Ac (2x100 mL). The organic phase was separated and washed with sat. aqueous NaHCO3 (3 x 80 mL), 1 M NaHSO4 (3 x 80 mL) and brine (2 x 80 mL), then dried (Na2SO4), filtered, and concentrated. The residue was purified by silica gel column chromatography to yield compound 233. LCMS
and NMR were consistent with the structure. Compound 233 (0.59 g, 0.48 mmol) was dissolved in methanol (2.2 mL) and ethyl acetate (2.2 mL). Palladium on carbon (10 wt% Pd/C, wet, 0.07 g) was added, and the reaction mixture was stirred under hydrogen atmosphere for 3 h. The reaction mixture was filtered through a pad of Celite and concentrated to yield the carboxylic acid. The carboxylic acid (1.32 g, 1.15 mmol, cluster free acid) was dissolved in DMF (3.2 mL). To this N,N-diisopropylehtylamine (0.3 mL, 1.73 mmol) and PFPTFA (0.30 mL, 1.73 mmol) were added. After 30 min stirring at room temperature the reaction mixture was poured into water (40 mL) and extracted with Et0Ac (2 x 50 mL). A standard work-up was completed as described above to yield compound 234. LCMS and NMR were consistent with the structure.
Oligonucleotide 235 was prepared using the general procedure described in Example 46. The Ga1NAc2 cluster portion (Ga1NAc2-24a) of the conjugate group Ga1NAc2-24 can be combined with any cleavable moiety present on the oligonucleotide to provide a variety of conjugate groups. The structure of Ga1NAc2-24 (Ga1NAc2-24a-CM) is shown below:
r_H OH

HO
AcHN NH
rH OH

rENI En AcHN N

Example 105: Synthesis of oligonucleotides comprising a Ga1NAc1-25 conjugate 0 83e 3'5') 11 OAc OAc F
OLIGO O¨P-0¨(CH2)6-NFI2 AcO01 F= F

1. Borate buffer, DMSO, pH 8.5, rt AcHN
166 2. aq. ammonia, rt OH OH

cm OLIGO
AcHN H 6 The synthesis of compound 166 is described in Example 54. Oligonucleotide 236 was prepared using the general procedure described in Example 46.
Alternatively, oligonucleotide 236 was synthesized using the scheme shown below, and compound 238 was used to form the oligonucleotide 236 using procedures described in Example 10.

OA OAc H2N" OA OH OAc Ac0 -F PFPTFA ______________ Ac0 OH
NHAc OH NHAc TEA, Acetonitrile OA OAc tetrazole, 1-Methylimidazole, DMF

__________________________________________ Ac0 O 11 2-cyanoethyltetraisopropyl phosphorodiamidite NHAc (!) LCN
Oligonucleotide OH OH
synthesis HO o 0 _____________ >a-_______________________________________________________ OLIGO
AcHN H 6 ¨

The GalNAci cluster portion (GalNAci-25,) of the conjugate group GalNAc1-25 can be combined with any cleavable moiety present on the oligonucleotide to provide a variety of conjugate groups. The structure of Ga1NAc1-25 (GalNAci-25a-CM) is shown below:
OH OH

go AcHN H 6 Example 106: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising a 5'-Ga1NAc2or a 5'-Ga1NAc3 conjugate Oligonucleotides listed in Tables 116 and 117 were tested in dose-dependent studies for antisense inhibition of SRB-1 in mice.
Treatment Six to week old, male C57BL/6 mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once with 2, 7, or 20 mg/kg of ISIS No. 440762; or with 0.2, 0.6, 2, 6, or 20 mg/kg of ISIS
No. 686221, 686222, or 708561; or with saline. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final administration. Liver SRB-1 mRNA
levels were measured using real-time PCR. SRB-1 mRNA levels were normalized to cyclophilin mRNA levels according to standard protocols. The antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner, and the ED50 results are presented in Tables 116 and 117. Although previous studies showed that trivalent GalNAc-conjugated oligonucleotides were significantly more potent than divalent GalNAc-conjugated oligonucleotides, which were in turn significantly more potent than monovalent GalNAc conjugated oligonucleotides (see, e.g., Khorev et al., Bioorg. & Med. Chem., Vol. 16, 5216-5231 (2008)), treatment with antisense oligonucleotides comprising monovalent, divalent, and trivalent GalNAc clusters lowered SRB-1 mRNA levels with similar potencies as shown in Tables 116 and 117.
Table 116 Modified oligonucleotides targeting SRB-1 ISIS,ED50 SEQ
Sequences (5 to 3') GalNAc Cluster No. (mg/kg) ID No 440762 TIsmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTi.smCk n/a 4.7 GalNAc2-24a-0,AdoTkamCksAdsGasTasmCdsAdsTasGasAds 686221 Ga1NAc2-24a 0.39 mCdsTdsTksmCk GalNAc3-13a-0,AdoTksmCksAdsGdsTdsmCdsAdsTdsGdsAds 686222 Ga1NAc3-13a 0.41 mCdsTdsTksmCk See Example 93 for table legend. The structure of Ga1NAc3-13a was shown in Example 62, and the structure of GalNAc2-24a was shown in Example 104.
Table 117 Modified oligonucleotides targeting SRB-1 ISIS,ED50 SEQ
Sequences (5 to 3') GalNAc Cluster No. (mg/kg) ID No 440762 TkamCkaAdaGdaTdsmCdsAds'r dsGdsAdsmCdsTasTi.smCk n/a 5 GalNAci-25a-0,TksmCksAdsGasTasmCdsAdsTasGasAds 708561 Ga1NAc1-25a 0.4 mCm daTdaTkaCk See Example 93 for table legend. The structure of GalNAci-25a was shown in Example 105.
The concentrations of the oligonucleotides in Tables 116 and 117 in liver were also assessed, using procedures described in Example 75. The results shown in Tables 117a and 117b below are the average total antisense oligonucleotide tissues levels for each treatment group, as measured by UV in units of [tg oligonucleotide per gram of liver tissue. The results show that the oligonucleotides comprising a GalNAc conjugate group accumulated in the liver at significantly higher levels than the same dose of the oligonucleotide lacking a GalNAc conjugate group. Furthermore, the antisense oligonucleotides comprising one, two, or three GalNAc ligands in their respective conjugate groups all accumulated in the liver at similar levels. This result is surprising in view of the Khorev et al. literature reference cited above and is consistent with the activity data shown in Tables 116 and 117 above.
Table 117a Liver concentrations of oligonucleotides comprising a Ga1NAc2 or Ga1NAc3 conjugate group Dosage ISIS No. (g/k [Antisense oligonucleotide] (m/g) GalNAc cluster CM
mg) 2 2.1 440762 7 13.1 n/a n/a
20 31.1 0.2 0.9 686221 0.6 2.7 Ga1NAc2-24a Ad 2 12.0 6 26.5 0.2 0.5 0.6 1.6 .6 686222 11 Ga1NAc3-13a Ad 6 19.8 Table 117b Liver concentrations of oligonucleotides comprising a GalNAci conjugate group Dosage ISIS No. [Antisense oligonucleotide] (pg/g) GalNAc cluster CM
(mg/kg) 2 2.3 440762 7 8.9 n/a n/a 20 23.7 0.2 0.4 0.6 1.1 708561 2 5.9 Ga1NAc1-25a PO
6 23.7 20 53.9 Example 107: Synthesis of oligonucleotides comprising a Ga1NAc1-26 or Ga1NAc1-27 conjugate HO
OH /[. CM Olga HO
AcHN

OH
Oligonucleotide 239 is synthesized via coupling of compound 47 (see Example 15) to acid 64 (see Example 32) using HBTU and DIEA in DMF. The resulting amide containing compound is phosphitylated, then added to the 5'-end of an oligonucleotide using procedures described in Example 10. The GalNAci cluster portion (GalNAci-26a) of the conjugate group Ga1NAc1-26 can be combined with any cleavable moiety present on the oligonucleotide to provide a variety of conjugate groups. The structure of Ga1NAci-26 (GalNAci-26a-CM) is shown below:
HO OH

AcHN
OH

In order to add the GalNAci conjugate group to the 3'-end of an oligonucleotide, the amide formed from the reaction of compounds 47 and 64 is added to a solid support using procedures described in Example 7. The oligonucleotide synthesis is then completed using procedures described in Example 9 in order to form oligonucleotide 240.
HO H
HO
AcHN
___________________________________________ 3' 5' 04 , _____ CM Oligo The GalNAci cluster portion (GalNAci-27,) of the conjugate group GalNAc1-27 can be combined with any cleavable moiety present on the oligonucleotide to provide a variety of conjugate groups. The structure of Ga1NAc1-27 (GalNAci-27a-CM) is shown below:

AcHN
0 El Example 108: Antisense inhibition in vivo by oligonucleotides comprising a GaINAc conjugate group targeting Apo(a) in vivo The oligonucleotides listed in Table 118 below were tested in a single dose study in mice.
Table 118 Modified ASOs targeting APO(a) ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
TesGesmCesTesmCesmCd Gd Ta Ta Gd Gd Ta Gd ssssssss s 494372 n/a n/a 277 TasTesGesTesTesmCe GalNAc3-7a-0,TesGesmCesTesmCesmCdsGasTasTasGasGas 681251 Ga1NAc3-7a PO 277 TdsGasmCdsTasTesGes TesTesmCe GalNAc3-3a-0,TesGeomCeoTeomCeomCdsGasTasTasGasGas 681255 Ga1NAc3-3a PO 277 TdsGasmCdsTasTe0Gee TesTesmCe GalNAc3-10a-0,TesGeomCeoTeomCeomCdsGdsTdsTasGasGas 681256 Ga1NAc3-10a PO 277 Td5Ga5mCdsTasTe0Gee TesTesmCe GalNAc3-7a-0,TesGeemCeeTeemCeemCdsGasTasTasGasGas 681257 Ga1NAc3-7a PO 277 TdsGasmCdsTasTe0Gee TesTesmCe GalNAc3-13a-0,TesGeomCeoTeomCeomCdsGdsTdsTasGasGas 681258 Ga1NAc3-13a PO 277 TdsGasmCdsTasTe0Gee TesTesmCe 681260 TesGeomCeoTeomCeomCdsGdsTasTasGasGas TasGasmCdsTdsrreoGeo Ga1NAc3-19a Ad 276 TõTealliCeoAdo¨Ga1NAc3-19 The structure of GalNAc3-7a was shown in Example 48.
Treatment Male transgenic mice that express human Apo(a) were each injected subcutaneously once with an oligonucleotide and dosage listed in Table 119 or with PBS. Each treatment group consisted of 4 animals.
Blood was drawn the day before dosing to determine baseline levels of Apo(a) protein in plasma and at 1 week following the first dose. Additional blood draws will occur weekly for approximately 8 weeks. Plasma Apo(a) protein levels were measured using an ELISA. The results in Table 119 are presented as the average percent of plasma Apo(a) protein levels for each treatment group, normalized to baseline levels (% BL), The results show that the antisense oligonucleotides reduced Apo(a) protein expression. Furthermore, the oligonucleotides comprising a GalNAc conjugate group exhibited even more potent reduction in Apo(a) expression than the oligonucleotide that does not comprise a conjugate group.
Table 119 Apo(a) plasma protein levels Apo(a) at ISIS No. Dosage (mg/kg) 1 week PBS n/a 143 Example 109: Synthesis of oligonucleotides comprising a Ga1NAc1-28 or Ga1NAc1-29 conjugate OH ____________________________________________ , 5 3' HOµ ss0------"
0 = CM ¨ Oligo HO C))C, AcHN

Oligonucleotide 241 is synthesized using procedures similar to those described in Example 71 to form the phosphoramidite intermediate, followed by procedures described in Example 10 to synthesize the oligonucleotide. The GalNAci cluster portion (GalNAci-28a) of the conjugate group Ga1NAc1-28 can be OH
ss0 _______________________________________ 0 = 359CM

AcHN

combined with any cleavable moiety present on the oligonucleotide to provide a variety of conjugate groups.
The structure of Ga1NAc1-28 (GalNAci-28a-CM) is shown below:
In order to add the GalNAci conjugate group to the 3'-end of an oligonucleotide, procedures similar to those described in Example 71 are used to form the hydroxyl intermediate, which is then added to the solid support using procedures described in Example 7. The oligonucleotide synthesis is then completed using procedures described in Example 9 in order to form oligonucleotide 242.
OH
HOµ 0 .õOH
HOO....,__,..........õ.....),, N-----...,----.....õ--,..õ--N?
AcHN H 3' 5' , ____________________________________________ .
242 0 0¨ CM ¨Oligo .. ___________________________________________ õ ., The GalNAci cluster portion (GalNAci-29a) of the conjugate group Gall\lAc1-29 can be combined with any cleavable moiety present on the oligonucleotide to provide a variety of conjugate groups. The structure of Ga1NAc1-29 (GalNAci-29a-CM) is shown below:
OH
HO () .õOH
HO r9..c)c N ----....-N?
AcHN H

Example 110: Synthesis of oligonucleotides comprising a Ga1NAc1-30 conjugate OAc OAc AIL Ac0....7____.
HOWOTBDPS Ac0 OOTBD

Ac0 t¨i---) ____________________ , PS
TMSOTf AcHN
y0 243 1. NH3/Me0H ODMTr 2. DMTrCI Ac0....r......
1. TBAF
3. Ac20, pyr 0 Ac0 OOTBDPS 2.
Phosphitilation ________________ , _____________________________________________ ).-AcHN 244 ODMTr Ac0....r......
1. Couple to 5'-end of ASO

Ac0 00õOCE _____________________________________________________ P ,..
AcHN 1 245 N(iPr)2 2. Deprotect and purify ASO using DMT-on purification methods HO\
3' ago AcHN

Oligonucleotide 246 comprising a Ga1NAc1-30 conjugate group, wherein Y is selected from 0, S, a substituted or unsubstituted C1-C10 alkyl, amino, substituted amino, azido, alkenyl or alkynyl, is synthesized as shown above. The GalNAci cluster portion (GalNAci-30a) of the conjugate group Ga1NAc1-30 can be 5 combined with any cleavable moiety to provide a variety of conjugate groups. In certain embodiments, Y is part of the cleavable moiety. In certain embodiments, Y is part of a stable moiety, and the cleavable moiety is present on the oligonucleotide. The structure of GalNAci-30a is shown below:
OH
HO\

HO
AcHN
Example 111: Synthesis of oligonucleotides comprising a Ga1NAc2-31 or Ga1NAc2-32 conjugate HO 1. DMTrCI DMTrO Couple to 5'-end of ASO
2. Phosphitilati OCE on ¨0-P
¨OH _______________________________ N(iPr)2 HODMTrO

Bx 1. Remove DMTr groups DMTrO 2. Couple amidite 245 ¨0õO ''IX
3. Deprotect and purify ASO using o DMTrO 0 Y 0-Oligo DMT-on purification methods OH

HO
,P\
AcHN 0' y-0õO _______ op\ Oligo OHY
Hov.(rLN/0 HCAcHN
Oligonucleotide 250 comprising a GalNAc2-31 conjugate group, wherein Y is selected from 0, S, a OH

HO
AcHN o'\' 0-p, OH

HO\,(72._\./0 H AcHN

substituted or unsubstituted C1-C10 alkyl, amino, substituted amino, azido, alkenyl or alkynyl, is synthesized as shown above. The Ga1NAc2 cluster portion (Ga1NAc2-3 1 a) of the conjugate group GalNAc2-31 can be combined with any cleavable moiety to provide a variety of conjugate groups.
In certain embodiments, the Y-containing group directly adjacent to the 5'-end of the oligonucleotide is part of the cleavable moiety. In certain embodiments, the Y-containing group directly adjacent to the 5'-end of the oligonucleotide is part of a stable moiety, and the cleavable moiety is present on the oligonucleotide. The structure of Ga1NAc2-31a is shown below:

The synthesis of an oligonucleotide comprising a Ga1NAc2-32 conjugate is shown below.
1. DMTrCI
2. Ally! Br 3. 0s04, Na104 1. Couple to 5'-end of ASO
HO 4. NaBH4 DMTrO 2. Remove DMTr groups 5. Phosphitilation 3. Couple amidite 245 ¨OH ______________________ 0, 4. Deprotect and purify ASO using HO DMTrO
,P¨NOP02 DMT-on purification methods OH

HO
\ 0õy 5' AcHN
¨0õO ;p, Oligo ,P\ 0 0 __ Y
OH d HO NHAc Oligonucleotide 252 comprising a Ga1NAc2-32 conjugate group, wherein Y is selected from 0, S, a substituted or unsubstituted C1-C10 alkyl, amino, substituted amino, azido, alkenyl or alkynyl, is synthesized as shown above. The Ga1NAc2 cluster portion (Ga1NAc2-32a) of the conjugate group Ga1NAc2-32 can be combined with any cleavable moiety to provide a variety of conjugate groups.
In certain embodiments, the Y-containing group directly adjacent to the 5'-end of the oligonucleotide is part of the cleavable moiety. In certain embodiments, the Y-containing group directly adjacent to the 5'-end of the oligonucleotide is part of a stable moiety, and the cleavable moiety is present on the oligonucleotide. The structure of Ga1NAc2-32a is shown below:
OH
________ 0 HO
ID
AcHN /\
Y

e 0 Y
OH O' Y

HO NHAc Example 112: Modified oligonucleotides comprising a GalNAci conjugate The oligonucleotides in Table 120 targeting SRB-1 were synthesized with a GalNAci conjugate group in order to further test the potency of oligonucleotides comprising conjugate groups that contain one GalNAc ligand.

Table 120 GalNAc SEQ
ISIS No. Sequence (5' to 3') CM
cluster ID NO.
711461 Ga1NAc1-25a_0,Ado Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds GalNAci-25a Ad 254 Gds Ads mCds Tds Tes mCes mCes Tes Te 711462 Ga1NAci-25a_0,Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds Gds GalNAci-25a PO 252 Ads mCds Tds Tes mCes mCes Tes Te 711463 Ga1NAci-25a_0,Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds GalNAci-25a PO 252 Gds Ads mCds Tds Teo mCeo mCes Tes Te 711465 Ga1NAci-26a_0,Ado Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds GalNAci-26a Ad 254 Gds Ads mCds Tds Tes mCes mCes Tes Te 711466 GalNAci-26a_0,Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds Gds GalNAci-26a PO 252 Ads mCds Tds Tes mCes mCes Tes Te 711467 Ga1NAci-26a_0,Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds GalNAci-26a PO 252 Gds Ads mCds Tds Teo mCeo mCes Tes Te 711468 Ga1NAci-28a_0,Ado Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds GalNAci-28a Ad 254 Gds Ads mCds Tds Tes mCes mCes Tes Te 711469 GalNAci-28a_0,Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds Gds GalNAci-28a PO 252 Ads mCds Tds Tes mCes mCes Tes Te 711470 GalNAci-28a-0,Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds GalNAci-28a PO 252 Gds Ads mCds Tds Teo mCeo mCes Tes Te 713844 Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-27a PO 252 Tes mCes mCes Tes Tew_GalNAci-27a 713845 Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-27a PO 252 Teo mCeo mCes Tes Tew_GalNAci-27a 713846 Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-27a Ad 253 Teo mCeo mCes Tes Teo Ado,_GalNAci-27a 713847 Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-29a PO 252 Tes mCes mCes Tes Te0,_GalNAel-29a 713848 Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-29a PO 252 Teo mCeo mCes Tes Te0,_GalNAel-29a 713849 Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-29a Ad 253 Tes mCes mCes Tes Teo Ado,_GalNAei-29a 713850 Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-29a Ad 253 Teo mCeo mCes Tes Teo Ado,_GalNAei-29a Example 113: Design and screening of duplexed antisense compounds targeting apolipoprotein C-III
In accordance with the present invention, a series of nucleic acid duplexes comprising the antisense compounds of the present invention and their complements are designed to target apolipoprotein C-III. The nucleobase sequence of the antisense strand of the duplex comprises at least a portion of an oligonucleotide in Table 121. The ends of the strands may be modified by the addition of one or more natural or modified nucleobases to form an overhang. The sense strand of the dsRNA is then designed and synthesized as the complement of the antisense strand and may also contain modifications or additions to either terminus. For example, in one embodiment, both strands of the dsRNA duplex would be complementary over the central nucleobases, each having overhangs at one or both termini.
For example, a duplex comprising an antisense strand having the sequence CGAGAGGCGGACGGGACCG (SEQ ID NO: 12) and having a two-nucleobase overhang of deoxythymidine(dT) would have the following structure (Antisense SEQ ID NO:
13, Complement SEQ ID
NO: 14):
cgagaggcggacgggaccgTT Antisense Strand (SEQ ID
NO: 13) TTgctctccgcctgccctggc Complement (SEQ ID NO: 14) In another embodiment, a duplex comprising an antisense strand having the same sequence CGAGAGGCGGACGGGACCG (SEQ ID NO: 12) may be prepared with blunt ends (no single stranded overhang) as shown (Antisense SEQ ID NO: 15, Complement SEQ ID NO: 16):
cgagaggcggacgggaccg Antisense Strand (SEQ ID
NO: 15) gctctccgcctgccctggc Complement (SEQ ID NO: 16) RNA strands of the duplex can be synthesized by methods disclosed herein or purchased from Dharmacon Research Inc., (Lafayette, CO). Once synthesized, the complementary strands are annealed. The single strands are aliquoted and diluted to a concentration of 50 [LM. Once diluted, 30 [LI., of each strand is combined with 15 L of a 5X solution of annealing buffer. The final concentration of said buffer is 100 mM
potassium acetate, 30 mM HEPES-KOH pH 7.4, and 2mM magnesium acetate. The final volume is 75 [LL.
This solution is incubated for 1 minute at 90 C and then centrifuged for 15 seconds. The tube is allowed to sit for 1 hour at 37 C at which time the dsRNA duplexes are used in experimentation. The final concentration of the dsRNA duplex is 20 [LM. This solution can be stored frozen (-20 C) and freeze-thawed up to 5 times.
Once prepared, the duplexed antisense compounds are evaluated for their ability to modulate apolipoprotein C-III expression.
When cells reached 80% confluency, they are treated with duplexed antisense compounds of the invention. For cells grown in 96-well plates, wells are washed once with 200 [LI., OPTI-MEM-1 TM reduced-serum medium (Gibco BRL) and then treated with 130 [LI., of OPTI-MEM-1 TM
medium containing 12 [tg/mL
LIPOFECTINTm reagent (Gibco BRL) and the desired duplex antisense compound at a final concentration of 200 nM. After 5 hours of treatment, the medium is replaced with fesh medium.
Cells are harvested 16 hours after treatment, at which time RNA is isolated and target reduction measured by RT-PCR.
Example 114:
Antisense inhibition of human apolipoprotein C-III expression by chimeric phosphorothioate oligonucleotides haying 2'-MOE wings and a deoxy gap In accordance with the present invention, a series of antisense compounds was designed to target different regions of the human apolipoprotein C-III RNA, using published sequences (nucleotides 6238608 to 6242565 of GenBank accession number NT_035088.1, representing a genomic sequence, incorporated herein as SEQ ID NO: 3, and GenBank accession number NM_000040.1, incorporated herein as SEQ ID NO: 1).
The compounds are shown in Table 121. "Target site" indicates the first (5'-most) nucleotide number on the particular target sequence to which the compound binds. All compounds in Table 121 are chimeric oligonucleotides ("gapmers") 20 nucleotides in length, composed of a central "gap" region consisting of ten 2'-deoxynucleotides, which is flanked on both sides (5' and 3' directions) by five-nucleotide "wings". The wings are composed of 2 '-0-(2-methoxyethyl) nucleotides, also known as (2'-M0E)nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P=S) throughout the oligonucleotide. All cytidine residues are 5-methylcytidines. The compounds were analyzed for their effect on human apolipoprotein C-III
mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from three experiments in which HepG2 cells were treated with the antisense oligonucleotides of the present invention. The positive control for each datapoint is identified in the table by sequence ID number. If present, "N.D." indicates "no data".
Table 121 - Inhibition of human apolipoprotein C-III mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap TARGET

ISIS # REGION SEQ ID SEQUENCE
SITE INHIB ID NO SEQ ID
NO
NO
167824 5'UTR 3 414 ctggagcagctgcctctagg 79 19 1 167835 Coding 3 1292 ccctgcatgaagctgagaag 60 20 1 167837 Coding 1 141 gtgcttcatgtaaccctgca 88 21 1 167846 Coding 3 1369 tggcctgctgggccacctgg 66 22 1 167848 Coding 3 3278 tgctccagtagtctttcagg 81 23 1 167851 Coding 3 3326 tgacctcagggtccaaatcc 41 24 1 304739 5'UTR 3 401 ctctagggatgaactgagca 62 25 1 304740 5'UTR 3 408 cagctgcctctagggatgaa 44 26 1 304741 5'UTR 1 17 ttcctggagcagctgcctct 57 27 1 304742 5'UTR 1 24 acctctgttcctggagcagc 78 28 1 304743 Start Codon 1 29 atggcacctctgttcctgga 78 29 1 304744 Start Codon 3 1065 gggctgcatggcacctctgt 73 30 1 304745 Coding 3 1086 ggcaacaacaaggagtaccc 90 31 1 304746 Coding 3 1090 ggagggcaacaacaaggagt 80 32 1 304747 Coding 1 87 agctcgggcagaggccagga 49 33 1 304748 Coding 1 92 tctgaagctcgggcagaggc 72 34 1 304749 Coding 1 97 cggcctctgaagctcgggca 11 35 1 304750 Coding 3 1267 catcctcggcctctgaagct 49 36 1 304751 Coding 3 1273 gggaggcatcctcggcctct 65 37 1 304752 Coding 3 1278 gagaagggaggcatcctcgg 82 38 1 304753 Coding 3 1281 gctgagaagggaggcatcct 75 39 1 304754 Coding 3 1289 tgcatgaagctgagaaggga 74 40 1 304755 Coding 1 143 gcgtgcttcatgtaaccctg 95 41 1 304756 Coding 3 1313 ttggtggcgtgcttcatgta 92 42 1 304757 Coding 3 1328 gcatccttggcggtcttggt 98 43 1 304758 Coding 3 1334 ctcagtgcatccttggcggt 97 44 1 304759 Coding 3 1336 tgctcagtgcatccttggcg 93 45 1 304760 Coding 3 1347 ctcctgcacgctgctcagtg 65 46 1 304761 Coding 3 1349 gactcctgcacgctgctcag 77 47 1 304762 Coding 3 1358 gccacctgggactcctgcac 89 48 1 304763 Coding 1 210 gcccctggcctgctgggcca 71 49 1 304764 Coding 1 211 agcccctggcctgctgggcc 62 50 1 304765 Coding 3 3253 gaagccatcggtcacccagc 71 51 1 304766 Coding 3 3255 ctgaagccatcggtcaccca 85 52 1 304767 Coding 3 3265 tttcagggaactgaagccat 73 53 1 304768 Coding 3 3273 cagtagtctttcagggaact 40 54 1 304769 Coding 3 3283 aacggtgctccagtagtctt 66 55 1 304770 Coding 3 3287 ccttaacggtgctccagtag 88 56 1 304771 Coding 3 3295 gaacttgtccttaacggtgc 59 57 1 304772 Coding 3 3301 ctcagagaacttgtccttaa 88 58 1 304773 Coding 3 3305 agaactcagagaacttgtcc 75 59 1 304774 Coding 3 3310 atcccagaactcagagaact 0 60 1 304775 Coding 3 3320 cagggtccaaatcccagaac 70 61 1 304776 Coding 3 3332 ttggtctgacctcagggtcc 90 62 1 304777 Coding 3 3333 gttggtctgacctcagggtc 84 63 1 304778 Coding 3 3339 gctgaagttggtctgacctc 81 64 1 304779 Coding 3 3347 cagccacggctgaagttggt 75 65 1 304780 Stop Codon 3 3351 caggcagccacggctgaagt 83 66 1 304781 Stop Codon 3 3361 attgaggtctcaggcagcca 79 67 1 304782 3'UTR 3 3385 tggataggcaggtggacttg 64 68 1 304783 3'UTR 1 369 ctcgcaggatggataggcag 76 69 1 304784 3'UTR 1 374 aggagctcgcaggatggata 58 70 1 304785 3'UTR 1 380 gacccaaggagctcgcagga 73 71 1 304786 3'UTR 1 385 tgcaggacccaaggagctcg 92 72 1 304787 3'UTR 3 3417 tggagattgcaggacccaag 88 73 1 304788 3'UTR 3 3422 agccctggagattgcaggac 69 74 1 304789 3'UTR 3 3425 ggcagccctggagattgcag 76 75 1 304790 3'UTR 3 3445 ccttttaagcaacctacagg 65 76 1 304791 3'UTR 3 3450 ctgtcccttttaagcaacct 53 77 1 304792 3'UTR 3 3456 agaatactgtcccttttaag 72 78 1 304793 3'UTR 3 3461 cactgagaatactgtccctt 67 79 1 304794 3'UTR 3 3469 taggagagcactgagaatac 59 80 1 304795 3'UTR 3 3472 gggtaggagagcactgagaa 74 81 1 304796 3'UTR 3 3509 aggccagcatgcctggaggg 63 82 1 304797 3'UTR 3 3514 ttgggaggccagcatgcctg 55 83 1 304798 3'UTR 3 3521 agctttattgggaggccagc 90 84 1 304799 3'UTR 3 3526 tgtccagctttattgggagg 85 85 1 304800 3'UTR 3 3528 cttgtccagctttattggga 94 86 1 304801 3'UTR 3 3533 agcttcttgtccagctttat 74 87 1 304802 3'UTR 3 3539 catagcagcttcttgtccag 73 88 1 exon:intron 304803 . . 3 416 acctggagcagctgcctcta 87 89 1 junction exon:intron 304804 junction 3 424 agggcattacctggagcagc 68 90 1 intron:exon 304805 . . 3 1053 acctctgttcctgcaaggaa 74 91 1 junction exon:intron 304806 . . 3 1121 aagtgcttacgggcagaggc 78 92 1 junction exon:intron 304807 . . 3 1380 gcgggtgtacctggcctgct 52 93 1 junction 304808 intron 3 2337 aaccctgttgtgaactgcac 59 94 1 304809 intron 3 2405 agtgagcaataccgcctgag 80 95 1 304810 intron 3 2542 cgggcttgaattaggtcagg 56 96 1 As shown in the table above, SEQ ID NOs 19, 20, 21, 22, 23, 25, 27, 28, 29, 30, 31, 32, 33, 34, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 55, 56, 57, 58, 59, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95 and 96 demonstrated at least 45% inhibition of human apolipoprotein C-III
expression in this assay and are therefore preferred. More preferred are SEQ ID NOs 75, 86 and 85. The target regions to which these preferred sequences are complementary are herein referred to as "preferred target segments" and are therefore preferred for targeting by compounds of the present invention. These preferred target segments are shown in a Table 123 below. The sequences represent the reverse complement of the preferred antisense compounds shown in the table above. "Target site" indicates the first (5'-most) nucleotide number on the particular target nucleic acid to which the oligonucleotide binds. Also shown in Table 123 is the species in which each of the preferred target segments was found.
Example 115: Antisense inhibition of mouse apolipoprotein C-III
expression by chimeric phosphorothioate oligonucleotides haying 2'-MOE wings and a deoxy gap.
In accordance with the present invention, a second series of antisense compounds was designed to target different regions of the mouse apolipoprotein C-III RNA, using published sequences (GenBank accession number L04150.1, incorporated herein as SEQ ID NO: 11). The compounds are shown in Table 2.
"Target site" indicates the first (5'-most) nucleotide number on the particular target nucleic acid to which the compound binds. All compounds in Table 2 are chimeric oligonucleotides ("gapmers") 20 nucleotides in length, composed of a central "gap" region consisting of ten 2'-deoxynucleotides, which is flanked on both sides (5' and 3' directions) by five-nucleotide "wings".
The wings are composed of 2 '-0-(2-methoxyethyl)nucleotides, also known as (2'-M0E)nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P=S) throughout the oligonucleotide. All cytidine residues are 5-methylcytidines. The compounds were analyzed for their effect on mouse apolipoprotein C-III mRNA
levels by quantitative real-time PCR as described in other examples herein. Data are averages from three experiments in which mouse primary hepatocyte cells were treated with the antisense oligonucleotides of the present invention. If present, "N.D." indicates "no data".
Table 122 - Inhibition of mouse apolipoprotein C-III mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap ISIS # REGION TARGET TARGET SEQUENCE % SEQ
SEQ ID NO SITE INHIB ID NO
167858 5'UTR 11 1 tagggataaaactgagcagg 47 97 167859 5'UTR 11 21 ctggagtagctagctgcttc 30 98 167860 start codon 11 41 gctgcatggcacctacgtac 80 99 167861 coding 11 62 ccacagtgaggagcgtccgg 86 100 167862 coding 11 88 ggcagatgccaggagagcca 55 101 167863 coding 11 104 ctacctcttcagctcgggca 56 167864 coding 11 121 cagcagcaaggatccctcta 83 167865 coding 11 131 gcacagagcccagcagcaag 49 104 167867 coding 11 215 ccctggccaccgcagctata 67 167868 coding 11 239 atctgaagtgattgtccatc 11 167869 coding 11 254 agtagcctttcaggaatctg 57 167870 coding 11 274 cttgtcagtaaacttgctcc 89 167871 coding 11 286 gaagccggtgaacttgtcag 55 109 167872 coding 11 294 gaatcccagaagccggtgaa 29 110 167873 coding 11 299 ggttagaatcccagaagccg 55 111 167874 coding 11 319 tggagttggttggtcctcag 79 167875 stop codon 11 334 tcacgactcaatagctggag 77 167877 3'UTR 11 421 cccttaaagcaaccttcagg 71 167878 3'UTR 11 441 agacatgagaacatactttc 81 167879 3'UTR 11 471 catgtttaggtgagatctag 87 167880 3'UTR 11 496 tcttatccagctttattagg 98 As shown in the table above, SEQ ID NOs 97, 99, 100, 101, 102, 103, 104, 105, 107, 108, 109, 111, 112, 113, 114, 115, 116 and 117 demonstrated at least 45% inhibition of mouse apolipoprotein C-III
expression in this experiment and are therefore preferred. More preferred are SEQ ID NOs 117, 116, and 100. The target regions to which these preferred sequences are complementary are herein referred to as "preferred target segments" and are therefore preferred for targeting by compounds of the present invention.
These preferred target segments are shown in the table below. The sequences represent the reverse complement of the preferred antisense compounds shown in the table above.
These sequences are shown to contain thymine (T) but one of skill in the art will appreciate that thymine (T) is generally replaced by uracil (U) in RNA sequences. "Target site" indicates the first (5'-most) nucleotide number on the particular target nucleic acid to which the oligonucleotide binds. Also shown in Table 3 is the species in which each of the preferred target segments was found.
Table 123 - Sequence and position of preferred target segments identified in apolipoprotein C-III.
SITE TARGE TARGE SEQUENCE REV COMP ACTIVE IN SEQ ID
ID T SEQ T SITE OF SEQ ID NO
ID NO
82975 3 414 cctagaggcagctgctccag 19 H. sapiens 118 82980 3 1292 cttctcagcttcatgcaggg 20 H. sapiens 119 82981 1 141 tgcagggttacatgaagcac 21 H. sapiens 120 82985 3 1369 ccaggtggcccagcaggcca 22 H. sapiens 121 82987 3 3278 cctgaaagactactggagca 23 H. sapiens 122 220510 3 401 tgctcagttcatccctagag 25 H. sapiens 123 220512 1 17 agaggcagctgctccaggaa 27 H. sapiens 124 220513 1 24 gctgctccaggaacagaggt 28 H. sapiens 125 220514 1 29 tccaggaacagaggtgccat 29 H. sapiens 126 220515 3 1065 acagaggtgccatgcagccc 30 H. sapiens 127 220516 3 1086 gggtactccttgttgttgcc 31 H. sapiens 128 220517 3 1090 actccttgttgttgccctcc 32 H. sapiens 129 220518 1 87 tcctggcctctgcccgagct 33 H. sapiens 130 220519 1 92 gcctctgcccgagcttcaga 34 H. sapiens 131 220521 3 1267 agcttcagaggccgaggatg 36 H. sapiens 132 220522 3 1273 agaggccgaggatgcctccc 37 H. sapiens 133 220523 3 1278 ccgaggatgcctcccttctc 38 H. sapiens 134 220524 3 1281 aggatgcctcccttctcagc 39 H. sapiens 135 220525 3 1289 tcccttctcagcttcatgca 40 H. sapiens 136 220526 1 143 cagggttacatgaagcacgc 41 H. sapiens 137 220527 3 1313 tacatgaagcacgccaccaa 42 H. sapiens 138 220528 3 1328 accaagaccgccaaggatgc 43 H. sapiens 139 220529 3 1334 accgccaaggatgcactgag 44 H. sapiens 140 220530 3 1336 cgccaaggatgcactgagca 45 H. sapiens 141 220531 3 1347 cactgagcagcgtgcaggag 46 H. sapiens 142 220532 3 1349 ctgagcagcgtgcaggagtc 47 H. sapiens 143 220533 3 1358 gtgcaggagtcccaggtggc 48 H. sapiens 144 220534 1 210 tggcccagcaggccaggggc 49 H. sapiens 145 220535 1 211 ggcccagcaggccaggggct 50 H. sapiens 146 220536 3 3253 gctgggtgaccgatggcttc 51 H. sapiens 147 220537 3 3255 tgggtgaccgatggcttcag 52 H. sapiens 148 220538 3 3265 atggcttcagttccctgaaa 53 H. sapiens 149 220540 3 3283 aagactactggagcaccgtt 55 H. sapiens 150 220541 3 3287 ctactggagcaccgttaagg 56 H. sapiens 151 220542 3 3295 gcaccgttaaggacaagttc 57 H. sapiens 152 220543 3 3301 ttaaggacaagttctctgag 58 H. sapiens 153 220544 3 3305 ggacaagttctctgagttct 59 H. sapiens 154 220546 3 3320 gttctgggatttggaccctg 61 H. sapiens 155 220547 3 3332 ggaccctgaggtcagaccaa 62 H. sapiens 156 220548 3 3333 gaccctgaggtcagaccaac 63 H. sapiens 157 220549 3 3339 gaggtcagaccaacttcagc 64 H. sapiens 158 220550 3 3347 accaacttcagccgtggctg 65 H. sapiens 159 220551 3 3351 acttcagccgtggctgcctg 66 H. sapiens 160 220552 3 3361 tggctgcctgagacctcaat 67 H. sapiens 161 220553 3 3385 caagtccacctgcctatcca 68 H. sapiens 162 220554 1 369 ctgcctatccatcctgcgag 69 H. sapiens 163 220555 1 374 tatccatcctgcgagctcct 70 H. sapiens 164 220556 1 380 tcctgcgagctccttgggtc 71 H. sapiens 165 220557 1 385 cgagctccttgggtcctgca 72 H. sapiens 166 220558 3 3417 cttgggtcctgcaatctcca 73 H. sapiens 167 220559 3 3422 gtcctgcaatctccagggct 74 H. sapiens 168 220560 3 3425 ctgcaatctccagggctgcc 75 H. sapiens 169 220561 3 3445 cctgtaggttgcttaaaagg 76 H. sapiens 170 220562 3 3450 aggttgcttaaaagggacag 77 H. sapiens 171 220563 3 3456 cttaaaagggacagtattct 78 H. sapiens 172 220564 3 3461 aagggacagtattctcagtg 79 H. sapiens 173 220565 3 3469 gtattctcagtgctctccta 80 H. sapiens 174 220566 3 3472 ttctcagtgctctcctaccc 81 H. sapiens 175 220567 3 3509 ccctccaggcatgctggcct 82 H. sapiens 176 220568 3 3514 caggcatgctggcctcccaa 83 H. sapiens 177 220569 3 3521 gctggcctcccaataaagct 84 H. sapiens 178 220570 3 3526 cctcccaataaagctggaca 85 H. sapiens 179 220571 3 3528 tcccaataaagctggacaag 86 H. sapiens 180 220572 3 3533 ataaagctggacaagaagct 87 H. sapiens 181 220573 3 3539 ctggacaagaagctgctatg 88 H. sapiens 182 220574 3 416 tagaggcagctgctccaggt 89 H. sapiens 183 220575 3 424 gctgctccaggtaatgccct 90 H. sapiens 184 220576 3 1053 ttccttgcaggaacagaggt 91 H. sapiens 185 220577 3 1121 gcctctgcccgtaagcactt 92 H. sapiens 186 220578 3 1380 agcaggccaggtacacccgc 93 H. sapiens 187 220579 3 2337 gtgcagttcacaacagggtt 94 H. sapiens 188 220580 3 2405 ctcaggcggtattgctcact 95 H. sapiens 189 220581 3 2542 cctgacctaattcaagcccg 96 H. sapiens 190 82997 11 1 cctgctcagttttatcccta 97 M. musculus 191 82999 11 41 gtacgtaggtgccatgcagc 99 M. musculus 192 83000 11 62 ccggacgctcctcactgtgg 100 M. musculus 193 83001 11 88 tggctctcctggcatctgcc 101 M. musculus 194 83002 11 104 tgcccgagctgaagaggtag 102 M. musculus 195 83003 11 121 tagagggatccttgctgctg 103 M. musculus 196 83004 11 131 cttgctgctgggctctgtgc 104 M. musculus 197 83006 11 215 tatagctgcggtggccaggg 105 M. musculus 198 83008 11 254 cagattcctgaaaggctact 107 M. musculus 199 83009 11 274 ggagcaagtttactgacaag 108 M. musculus 200 83010 11 286 ctgacaagttcaccggcttc 109 M. musculus 201 83012 11 299 cggcttctgggattctaacc 111 M. musculus 202 83013 11 319 ctgaggaccaaccaactcca 112 M. musculus 203 83014 11 334 ctccagctattgagtcgtga 113 M. musculus 204 83016 11 421 cctgaaggttgctttaaggg 114 M. musculus 83017 11 441 gaaagtatgttctcatgtct 115 M. musculus 83018 11 471 ctagatctcacctaaacatg 116 M. musculus 83019 11 496 cctaataaagctggataaga 117 M. musculus As these "preferred target segments" have been found by experimentation to be open to, and accessible for, hybridization with the antisense compounds of the present invention, one of skill in the art will recognize or be able to ascertain, using no more than routine experimentation, further embodiments of the invention that encompass other compounds that specifically hybridize to these preferred target segments and consequently inhibit the expression of apolipoprotein C-III.
According to the present invention, antisense compounds include antisense oligomeric compounds, antisense oligonucleotides, ribozymes, external guide sequence (EGS) oligonucleotides, alternate splicers, primers, probes, and other short oligomeric compounds that hybridize to at least a portion of the target nucleic acid.
Example 116:
Antisense inhibition of human apolipoprotein C-III expression by chimeric phosphorothioate oligonucleotides haying 2'-MOE wings and a deoxy gap ¨
additional antisense compounds In accordance with the present invention, an additional series of antisense compounds was designed to target different regions of the human apolipoprotein C-III RNA, using published sequences (nucleotides 6238608 to 6242565 of the sequence with GenBank accession number NT_035088.1, representing a genomic sequence, incorporated herein as SEQ ID NO: 3, and GenBank accession number NM_000040.1, incorporated herein as SEQ ID NO: 1). The compounds are shown in Table 124.
"Target site" indicates the first (5'-most) nucleotide number on the particular target sequence to which the compound binds. All compounds in Table 124 are chimeric oligonucleotides ("gapmers") 20 nucleotides in length, composed of a central "gap" region consisting of ten 2'-deoxynucleotides, which is flanked on both sides (5' and 3' directions) by five-nucleotide "wings". The wings are composed of 2 '-0-(2-methoxyethyl)nucleotides , also known as (2'-M0E) nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P=S) throughout the oligonucleotide. All cytidine residues are 5-methylcytidines.
The compounds were analyzed for their effect on human apolipoprotein C-III mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from three experiments in which HepG2 cells were treated with the antisense oligonucleotides of the present invention. If present, "N.D."
indicates "no data".
Table 124 - Inhibition of human apolipoprotein C-III mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap ISIS # TARGET TARGET SEQUENCE % INHIB SEQ ID
SEQ ID SITE NO
NO
167826 3 1063 gctgcatggcacctctgttc 0 209 167828 3 1110 ggcagaggccaggagcgcca 0 210 167830 1 91 ctgaagctcgggcagaggcc 9 211 167832 1 101 tcctcggcctctgaagctcg 0 212 167840 3 1315 tcttggtggcgtgcttcatg 0 213 167842 3 1335 gctcagtgcatccttggcgg 38 214 167844 3 1345 cctgcacgctgctcagtgca 28 215 167847 3 3256 actgaagccatcggtcaccc 0 216 167850 3 3306 cagaactcagagaacttgtc 0 217 167852 3 3336 gaagttggtctgacctcagg 0 218 167853 3 3420 ccctggagattgcaggaccc 0 219 167854 3 3426 gggcagccctggagattgca 22 220 167855 3 3446 cccttttaagcaacctacag 27 221 Example 117: Antisense inhibition of human apolipoprotein C-III
expression by chimeric phosphorothioate oligonucleotides haying 2'-MOE wings and a deoxy gap: dose-response study in HepG2 cells In accordance with the present invention, a subset of the antisense oligonucleotides from Examples and 17 was further investigated in a dose-response study. Treatment doses of ISIS 167842 (SEQ ID NO:
214), ISIS 167844 (SEQ ID NO: 215), ISIS 167846 (SEQ ID NO: 22), ISIS 167837 (SEQ ID NO: 21), ISIS
304789 (SEQ ID NO: 75), ISIS 304799 (SEQ ID NO: 85), and ISIS 304800 (SEQ ID:
86) were 50, 150 and 15 300 nM. The compounds were analyzed for their effect on human apolipoprotein C-III mRNA levels in HepG2 cells by quantitative real-time PCR as described in other examples herein. Data are averages from two experiments and are shown in the table below.

Table 125 - Inhibition of human apolipoprotein C-III mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap Dose of oligonucleotide SEQ ID 50 nm 150 nM 300 nM
ISIS # NO
Percent Inhibition These data demonstrate that the expression of apolipoprotein C-III is inhibited in a dose-dependent manner upon treatment of cells with antisense compounds targeting apolipoprotein C-III. These compounds were further analyzed in Hep3B cells for their ability to reduce mRNA levels in Hep3B cells and it was determined that ISIS 167842 and 167837 inhibited apolipoprotein C-III
expression in a dose dependent manner in this cell line as well.
Example 118: Antisense inhibition of apolipoprotein C-III in Cynomolgus monkey primary hepatocytes In a further embodiment, antisense compounds targeted to human apolipoprotein C-III were tested for their effects on apolipoprotein C-III expression in primary Cynomolgus monkey hepatocytes. Pre-plated primary Cynomolgus monkey hepatocytes were purchased from InVitro Technologies (Baltimore, MD).
Cells were cultured in high-glucose DMEM (Invitrogen Life Technologies, Carlsbad, CA) supplemented with 10% fetal bovine serum (Invitrogen Life Technologies, Carlsbad, CA), 100 units/mL and 100 [tg/mL
streptomycin (Invitrogen Life Technologies, Carlsbad, CA).
Cells at a density of 80,000 cells per well in a 24-well plate were treated with 10, 50, 150 and 300 nM of ISIS 304789 (SEQ ID NO: 75), ISIS 304799 (SEQ ID NO: 85) or ISIS 304800 (SEQ ID NO: 86).
ISIS 113529 (CTCTTACTGTGCTGTGGACA, SEQ ID NO: 17) served as a control oligonucleotide. ISIS
113529 is a chimeric oligonucleotide ("gapmer") 20 nucleotides in length, composed of a central "gap"
region consisting of ten 2'-deoxynucleotides, which is flanked on both sides (5' and 3' directions) by five-nucleotide "wings". The wings are composed of 2 '-0-(2-methoxyethyl)nucleotides , also known as (2'-MOE)nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P=S) throughout the oligonucleotide. All cytidine residues are 5-methylcytidines.
Following 24 hours of treatment with antisense oligonucleotides, apolipoprotein C-III mRNA was measured by real-time PCR as described by other examples herein, using the primers and probe designed to the human apolipoprotein C-III sequence (forward primer:
TCAGCTTCATGCAGGGTTACAT (SEQ ID
NO: 5) reverse primer: ACGCTGCTCAGTGCATCCT (SEQ ID NO: 6) and the PCR probe was: FAM-AAGCACGCCACCAAGACCGCC-TAMRA (SEQ ID NO: 7)) to measure Cynomolgous monkey apolipoprotein C-III mRNA. Primers and probe designed to human GAPDH (forward primer:
GAAGGTGAAGGTCGGAGTC (SEQ ID NO: 8) reverse primer: GAAGATGGTGATGGGATTTC (SEQ
ID NO: 9) and the PCR probe was: 5' JOE-CAAGCTTCCCGTTCTCAGCC- TAMRA 3' (SEQ ID
NO: 10)) were used to measure Cynomolgous monkey GAPDH mRNA expression, for the purpose of normalizing gene target quantities obtained by real-time PCR. Untreated cells served as the control to which data were normalized. Data are the average of three experiments and are presented in the table below.
Table 126 - Antisense inhibition of apolipoprotein C-III in Cynomolgus monkey primary hepatocytes Dose of Oligonucleotide ISIS # SEQ ID10 nM 50 nM 150 nM 300 nM
NO % Inhibition 113529 222 N.D. N.D. 0 0 Example 119: Cynomolgus monkey apolipoprotein C-III sequence In a further embodiment, a portion of the Cynomolgus monkey apolipoprotein C-III gene was sequenced. Positions 8 to 476 of the human apolipoprotein C-III mRNA sequence (incorporated in its entirety herein as SEQ ID NO: 1) contain the target segment to which ISIS
304789 hybridizes. The corresponding region of Cynomolgus monkey apolipoprotein C-III mRNA was sequenced. RNA was isolated and purified from primary Cynomolgus monkey hepatocytes (InVitro Technologies, Gaithersburg, MD) and was subjected to a reverse transcriptase reaction (kit from Invitrogen Life Technologies, Carlsbad, CA). The resultant cDNA was the substrate for 40 rounds of PCR amplification, using 5' and 3' primers designed to positions 8 and 476, respectively, of the human apolipoprotein C-III mRNA (Amplitaq PCR kit, Invitrogen Life Technologies, Carlsbad, CA). Following gel purification of the resultant 468 bp fragment, the forward and reverse sequencing reactions of each product were performed by Retrogen (San Diego, CA).
This Cynomolgus monkey sequence is incorporated herein as SEQ ID NO: 223 and is 92% identical to positions 8 to 476 of the human apolipoprotein C-III mRNA.

Example 120: Chimeric phosphorothioate oligonucleotide having 2'-MOE wings and a deoxy gap, targeted to Cynomolgus monkey apolipoprotein C-III
In a further embodiment, the sequence of Cynomolgus monkey apolipoprotein C-III incorporated herein as SEQ ID NO: 223 was used to design an antisense oligonucleotide having 100% complementarity to Cynomolgus apolipoprotein C-III mRNA. ISIS 340340 (GGCAGCCCTGGAGGCTGCAG;
incorporated herein as SEQ ID NO: 18) targets nucleotide 397 of SEQ ID NO: 223, within a region corresponding to the 3' UTR of the human apolipoprotein C-III to which ISIS 304789 hybridizes. ISIS
340340 is a chimeric oligonucleotide ("gapmer") 20 nucleotide in length composed of a central "gap"
region consisting of 2'deoxynucleotides, which is flanked on both sides (5' and 3' directions) by 5 nucleotide "wings". The wings are composed of 2'methoxyethyl (2'-M0E) nucleotides. Internucleoside (backbone) linkages are phosphorothioate (P=S) throughout the nucleotide. All cytidine residues are 5-methyl cytidines.

Claims (242)

CLAIMS:
1. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and comprises a nucleobase sequence comprising a portion of at least 8 contiguous nucleobases complementary to an equal length portion of nucleobases 3533 to 3552 of SEQ ID NO: 3, wherein the nucleobase sequence of the modified oligonucleotide is at least 80%
complementary to SEQ ID NO: 3.
2. The compound of claim 1, wherein the modified oligonucleotide comprises a nucleobase sequence comprising a portion of at least 10, at least 12, at least 14, at least 16, at least 18, at least 19, or at least 20 contiguous nucleobases complementary to an equal length portion of SEQ ID NO:
3.
3. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and comprises a nucleobase sequence comprising a portion of at least 8, at least 10, at least 12, at least 14, at least 15, or at least 16 contiguous nucleobases complementary to an equal length portion of nucleobases 3514 to 3558 of SEQ ID
NO: 1, wherein the nucleobase sequence of the modified oligonucleotide is at least 80%
complementary to SEQ ID NO: 3.
4. The compound of any preceding claim, wherein the nucleobase sequence of the modified oligonucleotide is at least 85%, at least 90%, at least 95%, or 100%
complementary to SEQ ID NO: 3.
5. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8, least 9, least 10, least 11, at least 12, least 13, at least 14, at least 15, at least 16, least 17, least 18, least 19, or 20 contiguous nucleobases of the nucleobase sequence of SEQ ID NO: 87.
6. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8, least 9, least 10, least 11, at least 12, least 13, at least 14, at least 15, at least 16, least 17, least 18, least 19, or 20 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NO: 19-96, 209-221.
7. The compound of any preceding claim, wherein the modified oligonucleotide is single-stranded.
8. The compound of any of claims 1-6, wherein the modified oligonucleotide is double stranded.
9. The compound of any preceding claim, wherein the modified oligonucleotide comprises at least one modified internucleoside linkage.
10. The compound of claim 9, wherein the modified internucleoside linkage is a phosphorothioate internucleoside linkage.
11. The compound of claim 10, wherein the modified oligonucleotide comprises at least one phosphodiester internucleoside linkage.
12. The compound of claim 10, wherein the modified oligonucleotide comprises at least 2 phosphodiester internucleoside linkages.
13. The compound of claim 10, wherein the modified oligonucleotide comprises at least 3 phosphodiester internucleoside linkages.
14. The compound of claim 10, wherein the modified oligonucleotide comprises at least 4 phosphodiester internucleoside linkages.
15. The compound of claim 10, wherein the modified oligonucleotide comprises at least 5 phosphodiester internucleoside linkages.
16. The compound of claim 10, wherein the modified oligonucleotide comprises at least 6 phosphodiester internucleoside linkages.
17. The compound of claim 10, wherein the modified oligonucleotide comprises at least 7 phosphodiester internucleoside linkages.
18. The compound of any of claims 11 to 17, wherein each internucleoside linkage of the modified oligonucleotide is selected from a phosphodiester internucleoside linkage and a phosphorothioate internucleoside linkage.
19. The compound of any of claims 1 to 8, wherein each internucleoside linkage of the modified oligonucleotide comprises is a phosphorothioate internucleoside linkage.
20. A compound consisting of ISIS 304801 and a conjugate group.
21. The compound of any preceding claim, wherein the modified oligonucleotide comprises at least one modified sugar.
22. The compound of claim 21, wherein at least one modified sugar is a bicyclic sugar.
23. The compound of claim 21, wherein at least one modified sugar comprises a 2'-O-methoxyethyl, a constrained ethyl, a 3'-fluoro-HNA or a 4'- (CH2),-O-2' bridge, wherein n is 1 or 2.
24. The compound of any preceding claim, wherein at least one nucleoside comprises a modified nucleobase.
25. The compound of claim 24, wherein the modified nucleobase is a 5-methylcytosine.
26. The compound of any preceding claim, wherein the modified oligonucleotide consists of 12 to 30 linked nucleosides and comprises:
a gap segment consisting of linked deoxynucleosides;
a 5' wing segment consisting of linked nucleosides;
a 3' wing segment consisting of linked nucleosides;
wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment and wherein each nucleoside of each wing segment comprises a modified sugar.
27. The compound of any preceding claim, wherein the modified oligonucleotide consists of 15 to 30, 18 to 24, 19 to 22, 13 to 25, 14 to 25, 15 to 25, 16 or 20 linked nucleosides.
28. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 20 linked nucleosides and has a nucleobase sequence comprising at least 8 contiguous nucleobases complementary to an equal length portion of any of SEQ
ID NO: 87, wherein the modified oligonucleotide comprises:
a gap segment consisting of ten linked deoxynucleosides;
a 5' wing segment consisting of five linked nucleosides;
a 3' wing segment consisting of five linked nucleosides;
wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment, wherein each nucleoside of each wing segment comprises a 2'-O-methoxyethyl sugar, wherein each internucleoside linkage is a phosphorothioate linkage and wherein each cytosine residue is a 5-methylcytosine.
29. The compound of any of claims 1 to 28, wherein the conjugate group is linked to the modified oligonucleotide at the 5' end of the modified oligonucleotide.
30. The compound of any of claims 1 to 28, wherein the conjugate group is linked to the modified oligonucleotide at the 3' end of the modified oligonucleotide.
31. The compound of any of claims 1-30, wherein the conjugate group comprises exactly one ligand.
32. The compound of any of claims 1-30, wherein the conjugate group comprises exactly two ligands.
33. The compound of any of claims 1-30, wherein the conjugate group comprises three or more ligands.
34. The compound of any of claims 1-30, wherein the conjugate group comprises exactly three ligands.
35. The compound of any of claims 31-34, wherein each ligand is selected from among: a polysaccharide, modified polysaccharide, mannose, galactose, a mannose derivative, a galactose derivative, D-mannopyranose, L-Mannopyranose, D-Arabinose, L-Galactose, D-xylofuranose, L-xylofuranose, D-glucose, L-glucose, D-Galactose, L-Galactose, .alpha.-D-Mannofuranose, .beta.-D-Mannofuranose, .alpha.-D-Mannopyranose, .beta.-D-Mannopyranose, .alpha.-D-Glucopyranose, .beta.-D-Glucopyranose, .alpha.-D-Glucofuranose, .beta.-D-Glucofuranose, .alpha.-D-fructofuranose, .alpha.-D-fructopyranose, .alpha.-D-Galactopyranose, .beta. -D-Galactopyranose, .alpha.-D-Galactofuranose, .beta. -D-Galactofuranose, glucosamine, sialic acid, .alpha.-D-galactosamine, N-Acetylgalactosamine, 2-Amino-3-O-[(R)-1-carboxyethyl]-2-deoxy-.beta.-D-glucopyranose, 2-Deoxy-2-methylamino-L-glucopyranose, 4,6-Dideoxy-4-formamido-2,3-di-O-methyl-D-mannopyranose, 2-Deoxy-2-sulfoamino-D-glucopyranose, N-Glycoloyl-.alpha.-neuraminic acid, 5-thio-.beta.-D-glucopyranose, methyl 2,3,4-tri-O-acetyl-1-thio-6-O-trityl-.alpha.-D-glucopyranoside, 4-Thio-.beta.-D-galactopyranose, ethyl 3,4,6,7-tetra-O-acetyl-2-deoxy-1,5-dithio-.alpha.-D-gluco-heptopyranoside, 2,5-Anhydro-D-allononitrile, ribose, D-ribose, D-4-thioribose, L-ribose, L-4-thioribose.
36. The compound of claim 35, wherein each ligand is N-acetyl galactosamine.
37. The compound of any of claims 1 to 30, wherein the conjugate group comprises:

38. The compound of any of claims 1 to 30, wherein the conjugate group comprises:
39. The compound of any of claims 1 to 30, wherein the conjugate group comprises:
40. The compound of any of claims 1 to 30, wherein the conjugate group comprises:

41. The compound of any of claims 1 to 30, wherein the conjugate group comprises:
42. The compound of any of claims 30 to 36, wherein the conjugate group comprises at least one phosphorus linking group or neutral linking group.
43. The compound of any of claims 1 to 42, wherein the conjugate group comprises a structure selected from among:

wherein n is from 1 to 12; and wherein m is from 1 to 12.
44. The compound of any of claims 1 to 42, wherein the conjugate group has a tether having a structure selected from among:
wherein L is either a phosphorus linking group or a neutral linking group;
Z1 is C(=O)O-R2;
Z2 is H, C1-C6 alkyl or substituted C1-C6 alky;
R2 is H, C1-C6 alkyl or substituted C1-C6 alky; and each m1 is, independently, from 0 to 20 wherein at least one m1 is greater than 0 for each tether.
45. The compound of claim 44, wherein conjugate group has a tether having a structure selected from among:
wherein Z2 is H or CH3; and each m1 is, independently, from 0 to 20 wherein at least one m1 is greater than 0 for each tether.
46. The compound of any of claims 30 to 36, wherein the conjugate group has tether having a structure selected from among:
wherein n is from 1 to 12; and wherein m is from 1 to 12.
47. The compound of any of claims 1 to 46, wherein the conjugate group is covalently attached to the modified oligonucleotide.
48. The compound of any of claims 1 to 47, wherein the compound has a structure represented by the formula:
wherein A is the modified oligonucleotide;
B is the cleavable moiety C is the conjugate linker D is the branching group each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
49. The compound of any of claims 1 to 47, wherein the compound has a structure represented by the formula:
wherein:
A is the modified oligonucleotide;
B is the cleavable moiety C is the conjugate linker D is the branching group each E is a tether;
each F is a ligand;
each n is independently 0 or 1; and q is an integer between 1 and 5.
50. The compound of any of claims 1 to 47, wherein the compound has a structure represented by the formula:
wherein A is the modified oligonucleotide;
B is the cleavable moiety;
C is the conjugate linker;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
51. The compound of any of claims 1 to 47, wherein the compound has a structure represented by the formula:
wherein A is the modified oligonucleotide;
C is the conjugate linker;
D is the branching group;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
52. The compound of any of claims 1 to 47, wherein the compound has a structure represented by the formula:
wherein A is the modified oligonucleotide;
C is the conjugate linker;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
53. The compound of any of claims 1 to 47, wherein the compound has a structure represented by the formula:
wherein A is the modified oligonucleotide;
B is the cleavable moiety;
D is the branching group;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
54. The compound of any of claims 1 to 47, wherein the compound has a structure represented by the formula:
wherein A is the modified oligonucleotide;
B is the cleavable moiety;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
55. The compound of any of claims 1 to 47, wherein the compound has a structure represented by the formula:
wherein A is the modified oligonucleotide;
D is the branching group;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
56. The compound of any of claims 29 to 55, wherein the conjugate linker has a structure selected from among:

wherein each L is, independently, a phosphorus linking group or a neutral linking group; and each n is, independently, from 1 to 20.
57. The compound of any of claims 29 to 55, wherein the conjugate linker has a structure selected from among:

58. The compound of any of claims 29 to 55, wherein the conjugate linker has the followingstructure:
59. The compound of any of claims 29 to 55, wherein the conjugate linker has a structure selected from among:
60. The compound of any of claims 29 to 55, wherein the conjugate linker has a structure selected from among:
61. The compound of any of claims 29 to 55, wherein the conjugate linker has a structure selected from among:
62. The compound of any of claims 29 to 61, wherein the conjugate linker comprises a pyrrolidine.
63. The compound of any of claims 29 to 61, wherein the conjugate linker does not comprise a pyrrolidine.
64. The compound of any of claims 29 to 63, wherein the conjugate linker comprises PEG.
65. The compound of any of claims 29 to 64, wherein the conjugate linker comprises an amide.
66. The compound of any of claims 29 to 64, wherein the conjugate linker comprises at least two amides.
67. The compound of any of claims 29 to 64, wherein the conjugate linker does not comprise an amide.
68. The compound of any of claims 29 to 67, wherein the conjugate linker comprises a polyamide.
69. The compound of any of claims 29 to 68, wherein the conjugate linker comprises an amine.
70. The compound of any of claims 29to 69, wherein the conjugate linker comprises one or more disulfide bonds.
71. The compound of any of claims 29 to 70, wherein the conjugate linker comprises a protein binding moiety.
72. The compound of claim 71, wherein the protein binding moiety comprises a lipid.
73. The compound of claim 71, wherein the protein binding moiety is selected from among: cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, O3-(oleoyl)lithocholic acid, O3-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine), a vitamin (e.g., folate, vitamin A, vitamin E, biotin, pyridoxal), a peptide, a carbohydrate (e.g., monosaccharide, disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, polysaccharide), an endosomolytic component, a steroid (e.g., uvaol, hecigenin, diosgenin), a terpene (e.g., triterpene, e.g., sarsasapogenin, friedelin, epifriedelanol derivatized lithocholic acid), or a cationic lipid.
74. The compound of claim 71, wherein the protein binding moiety is selected from among: a C16 to C22 long chain saturated or unsaturated fatty acid, cholesterol, cholic acid, vitamin E, adamantane or 1-pentafluoropropyl.
75. The compound of any of claims 48 to 74, wherein the conjugate linker has a structure selected from among:
wherein each n is, independently, is from 1 to 20; and p is from 1 to 6.
76. The compound of any of claims 29 to 75, wherein the conjugate linker has a structure selected from among:

wherein each n is, independently, from 1 to 20.
77. The compound of any of claims 29 to 75, wherein the conjugate linker has a structure selected from among:
78. The compound of any of claims 29 to 75, wherein the conjugate linker has a structure selected from among:
wherein n is from 1 to 20.
79. The compound of any of claims 29 to 75, wherein the conjugate linker has a structure selected from among:
80. The compound of any of claims 29 to 75, wherein the conjugate linker has a structure selected from among:
wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
81. The compound of any of claims 29 to 75, wherein the conjugate linker has the following structure:
82. The compound of any of claims 29 to 81, wherein the branching group has one of the following structures:

wherein each A1 is independently, O, S, C=O or NH; and each n is, independently, from 1 to 20.
83. The compound of any of claims 29 to 81, wherein the branching group has one of the following structures:
wherein each A1 is independently, O, S, C=O or NH; and each n is, independently, from 1 to 20.
84. The compound of any of claims 29 to 81, wherein the branching group has the following structure:
85. The compound of any of claims 29 to 81, wherein the branching group has the following structure:
86. The compound of any of claims 29 to 81, wherein the branching group has the following structure:
87. The compound of any of claims 29 to 81, wherein the branching group has the following structure:
88. The compound of any of claims 29 to 81, wherein the branching group comprises an ether.
89. The compound of any of claims 29 to 81, wherein the branching group has the following structure:
each n is, independently, from 1 to 20; and m is from 2 to 6.
90. The compound of any of claims 29 to 81, wherein the branching group has the following structure:
91. The compound of any of claims 29 to 81, wherein the branching group has the following structure:
92. The compound of any of claims 29 to 81, wherein the branching group comprises:
wherein each j is an integer from 1 to 3; and wherein each n is an integer from 1 to 20.
93. The compound of any of claims 29 to 81, wherein the branching group comprises:
94. The compound of any of claims 29 to 93, wherein each tether is selected from among:
wherein L is selected from a phosphorus linking group and a neutral linking group;
Z1 is C(=O)O-R2;
Z2 is H, C1-C6 alkyl or substituted C1-C6 alky;
R2 is H, C1-C6 alkyl or substituted C1-C6 alky; and each m1 is, independently, from 0 to 20 wherein at least one m1 is greater than 0 for each tether.
95. The compound of any of claims 29 to 93, wherein each tether is selected from among:
wherein Z2 is H or CH3; and each m2 is, independently, from 0 to 20 wherein at least one m2 is greater than 0 for each tether.
96. The compound of any of claims 29 to 93, wherein each tether is selected from among:
wherein n is from 1 to 12; and wherein m is from 1 to 12.
97. The compound of any of claims 29 to 93, wherein at least one tether comprises ethylene glycol.
98. The compound of any of claims 29 to 93 or 95, wherein at least one tether comprises an amide.
99. The compound of any of claims 29 to 93 or 95, wherein at least one tether comprises a polyamide.
100. The compound of any of claims 29 to 93 or 95, wherein at least one tether comprises an amine.
101. The compound of any of claims 29 to 93 or 95, wherein at least two tethers are different from one another.
102. The compound of any of claims 29 to 93 or 95, wherein all of the tethers are the same as one another.
103. The compound of any of claims 29 to 93, wherein each tether is selected from among:
wherein each n is, independently, from 1 to 20; and each p is from 1 to about 6.
104. The compound of any of claims 29 to 93, wherein each tether is selected from among:
105. The compound of any of claims 29 to 93, wherein each tether has the following structure:

wherein each n is, independently, from 1 to 20.
106. The compound of any of claims 29 to 93, wherein each tether has the following structure:
107. The compound of any of claims 29 to 93, wherein the tether has a structure selected from among:
wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
108. The compound of any of claims 29 to 93, wherein the tether has a structure selected from among:
109. The compound of any of claims 29 to 108, wherein the ligand is galactose.
110. The compound of any of claims 29 to 108, wherein the ligand is mannose-6-phosphate.
111. The compound of any of claims 29 to 108, wherein each ligand is selected from among:
wherein each R1 is selected from OH and NHCOOH.
112. The compound of any of claims 29 to 108, wherein each ligand is selected from among:
113. The compound of any of claims 29 to 108, wherein each ligand has the following structure:
114. The conjugated antisense compound of any of claims 29 to 108, wherein each ligand has the following structure:
115. The compound of any of claims 1 to 30 or 29 to 81, wherein the conjugate group comprises a cell-targeting moiety.
116. The compound of claim 115, wherein the cell-targeting moiety comprises:
wherein each n is, independently, from 1 to 20.
117. The compound of any of claims 115, wherein the cell-targeting moiety comprises:
118. The compound of claim 115, wherein the cell-targeting moiety comprises:
wherein each n is, independently, from 1 to 20.
119. The compound of claim 115, wherein the cell-targeting moiety comprises:
120. The compound of claim 115, the cell-targeting moiety comprises:
121. The compound of claim 115, wherein the cell-targeting moiety comprises:
122. The compound of claim 115, wherein the cell-targeting moiety comprises:
123. The compound of claim 115, wherein the cell-targeting moiety comprises:
124. The compound of claim 115, wherein the cell-targeting moiety comprises:
125. The compound of claim 115, wherein the cell-targeting moiety comprises:
126. The compound of claim 115, wherein the cell-targeting moiety comprises:
127. The compound of claim 115, wherein the cell-targeting moiety comprises:
128. The compound of claim 115, wherein the cell-targeting moiety comprises:
129. The compound of claim 115, wherein the cell-targeting moiety comprises:
130. The compound of claim 115, wherein the cell-targeting moiety comprises:
131. The compound of claim 115, wherein the cell-targeting moiety comprises:
132. The compound of claim 115, wherein the cell-targeting moiety comprises:
133. The compound of claim 115, wherein the cell-targeting moiety comprises:

134. The compound of claim 115, wherein the cell-targeting moiety comprises:
135. The compound of claim 115, wherein the cell-targeting moiety comprises:

136. The compound of claim 115, wherein the cell-targeting moiety comprises:
137. The compound of claim 115, wherein the cell-targeting moiety comprises:

138. The compound of claim 115, wherein the cell-targeting moiety comprises:
139. The compound of claim 115, wherein the cell-targeting moiety comprises:
140. The compound of claim 115, wherein the cell-targeting moiety comprises:

141. The compound of claim 115, wherein the cell-targeting moiety comprises:
wherein each Y is selected from O, S, a substituted or unsubstituted C1-C10 alkyl, amino, substituted amino, azido, alkenyl or alkynyl.
142. The compound of claim 115, wherein the conjugate group comprises:
wherein each Y is selected from O, S, a substituted or unsubstituted C1-C10 alkyl, amino, substituted amino, azido, alkenyl or alkynyl.
143. The compound of claim 115, wherein the cell-targeting moiety comprises:
wherein each Y is selected from O, S, a substituted or unsubstituted C1-C10 alkyl, amino, substituted amino, azido, alkenyl or alkynyl.
144. The compound of any of claims 1 to 30, wherein the conjugate group comprises:
145. The compound of any of claims 1 to 30, wherein the conjugate group comprises:
146. The compound of any of claims 1 to 30, wherein the conjugate group comprises:
147. The compound of claim 117, wherein the conjugate group comprises:
148. The compound of any of claims 1 to 147, wherein the conjugate group comprises a cleavable moiety selected from among: a phosphodiester, an amide, or an ester.
149. The compound of any of claims 1 to 147, wherein the conjugate group comprises a phosphodiester cleavable moiety.
150. The compound of any of claims 1 to 147, wherein the conjugate group does not comprise a cleavable moiety, and wherein the conjugate group comprises a phosphorothioate linkage between the conjugate group and the oligonucleotide.
151. The compound of any of claims 1 to 151, wherein the conjugate group comprises an amide cleavable moiety.
152. The compound of any of claims 1 to 151, wherein the conjugate group comprises an ester cleavable moiety.
153. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein each n is, independently, from 1 to 20;
Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
154. The compound of any of claims 1 to 30, wherein the compound has the following structure:

wherein each n is, independently, from 1 to 20;
Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
155. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein each n is, independently, from 1 to 20;
Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide;
Z is H or a linked solid support; and Bx is a heterocyclic base moiety.
156. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein each n is, independently, from 1 to 20;
Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide;
Z is H or a linked solid support; and Bx is a heterocyclic base moiety.
157. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide;and Bx is a heterocyclic base moiety.
158. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide;and Bx is a heterocyclic base moiety.
159. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
160. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
161. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
162. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
163. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
164. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
165. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
166. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
167. The compound of any of claims 1 to 30, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
168. The compound of any of claims 1 to 30, wherein the conjugate group comprises:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
169. The compound of any of claims 1 to 30, wherein the conjugate group comprises:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
170. The compound of any of claims 1 to 30, wherein the conjugate group comprises:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
171. The compound of any of claims 152 to 169, wherein B x is selected from among from adenine, guanine, thymine, uracil, or cytosine, or 5-methyl cytosine.
172. The compound of any of claims 155 to 171, wherein B x is adenine.
173. The compound of any of claims 155 to 171, wherein B x is thymine.
174. The compound of any of claims 155 to 171, wherein Q13 is O(CH2)2-OCH3.
175. The compound of any of claims 155 to 171, wherein Q13 is H.
176. A compound having the formula:
177. A compound having the formula:

178. A compound having the formula
179. 4. Markush Wherein either R1 is ¨OCH2CH2OCH3 (MOE)and R2 is H; or R1 and R2 together form a bridge, wherein R1 is ¨O- and R2 is ¨CH2-, -CH(CH3)-, or -CH2CH2-, and R1 and R2 are directly connected such that the resulting bridge is selected from: -O-CH2-, -O-CH(CH3)-, and ¨O-CH2CH2-;
And for each pair of R3 and R4 on the same ring, independently for each ring:
either R3 is selected from H
and -OCH2CH2OCH3 and R4 is H; or R3 and R4 together form a bridge, wherein R3 is ¨O-, and R4 is ¨CH2-, -CH(CH3)-, or -CH2CH2-and R3 and R4 are directly connected such that the resulting bridge is selected from: -O-CH2-, -O-CH(CH3)-, and ¨O-CH2CH2-;

And R5 is selected from H and ¨CH3;
And Z is selected from S- and O-.
180. A composition comprising the compound of any of claims 1-179 or salt thereof and at least one of a pharmaceutically acceptable carrier or diluent.
181. A prodrug comprising the compound of any of claims 1 to 179.
182. A method comprising administering to an animal the compound or composition of any of claims 1-179.
183. The method of claim 182, wherein the animal is a human.
184. The method of claim 182, for use in treating, preventing, or slowing progression of a disease related to elevated ApoCIII.
185. The method of claim 182, wherein administering the compound prevents, treats, ameliorates, or slows progression of a cardiovascular, metabolic and/or inflammatory disease, disorder or conditin in the animal.
186. The method of claim 182, comprising co-administering the compound or composition and a second agent.
187. The method of claim 186, wherein the compound or composition and the second agent are administered concomitantly.
188. The method of claim 182, wherein the administering is parenteral.
189. The method of claim 182, wherein the administering is subcutaneous.
190. The method of claim 182, for use in reducing ApoCIII mRNA or protein expression in an animal.
191. The method of claim 182, for use in reducing triglyceride level in the animal.
192. The method of claim 182, for use in increasing HDL levels in the animal.
193. The method of claim 184, for use in increasing chylomicron clearance in the animal, whereby the increase in chylomicron clearance treats, prevents, delays or ameliorates pancreatitis in the animal.
194. The method of claim 184, wherein the animal has, or is at risk of having, any one or more of hypertriglyceridemia, Fredrickson Type I dyslipidemia, FCS, LPLD, pancreatitis, diabetes, insulin insensitivity.
195. A compound having the formula:
Wherein n is 1, 2, 3, 4, 5, or 6; and wherein R' is selected from H and a protecting group.
196. A compound having the formula:

Wherein R comprises a reactive ester, an anhydride, a carbamimidic anhydride, an acyl halide, an acyl azide, or a phosphonium ion; and wherein n is 1, 2, 3, 4, 5, or 6; and wherein R' is selected from H and a protecting group.
197. The compound of claim 196, wherein R is a reactive ester.
198. The compound of claim 197, wherein R is wherein R2 is F or SO3-.
199. The compound of claim 197, wherein R is wherein R2 is H or SO3-.
200. The compound of claim 197, wherein R comprises wherein R3 is N or CH and R4 is H or Cl.
201. The compound of claim 197, wherein R comprises wherein R3 is N or CH.
202. The compound of claim 196, wherein R comprises
203. The compound of claim 196, wherein R is
204. The compound of claim 196, wherein R is
205. The compound of claim 196, wherein R is
206. A compound having the formula:
Wherein n is 1, 2, 3, 4, 5, or 6; and wherein R' is selected from H and a protecting group.
207. A compound having the formula:
Wherein R comprises a reactive ester, an anhydride, a carbamimidic anhydride, an acyl halide, an acyl azide, or a phosphonium ion; and wherein n is 1, 2, 3, 4, 5, or 6; and wherein R' is selected from H and a protecting group.
208. The compound of claim 207, wherein R is a reactive ester.
209. The compound of claim 208, wherein R is wherein R2 is F or SO3-.
210. The compound of claim 208, wherein R is wherein R2 is H or SO3-.
211. The compound of claim 208, wherein R comprises wherein R3 is N or CH and R4 is H or Cl.
212. The compound of claim 208, wherein R comprises wherein R3 is N or CH.
213. The compound of claim 207, wherein R comprises
214. The compound of claim 207, wherein R is
215. The compound of claim 207, wherein R is
216. The compound of claim 207, wherein R is
217. A compound having the formula:

Wherein n is 1, 2, 3, 4, 5, or 6; and wherein R' is selected from H and a protecting group.
218. A compound having the formula:
Wherein R comprises a reactive ester, an anhydride, a carbamimidic anhydride, an acyl halide, an acyl azide, or a phosphonium ion; and wherein n is 1, 2, 3, 4, 5 or 6; and wherein R' is selected from H and a protecting group.
219. The compound of claim 218, wherein R is a reactive ester.
220. The compound of claim 219, wherein R is wherein R2 is F or SO3-.
221. The compound of claim 219, wherein R is wherein R2 is H or SO3-.
222. The compound of claim 219, wherein R comprises wherein R3 is N or CH and R4 is H or Cl.
223. The compound of claim 219, wherein R comprises wherein R3 is N or CH.
224. The compound of claim 219, wherein R comprises
225. The compound of claim 218, wherein R is
226. The compound of claim 218, wherein R is
227. The compound of claim 218, wherein R is
228. A compound having the formula:
Wherein n is 1, 2, 3, 4, 5, or 6; and wherein R' is selected from H and a protecting group.
229. A compound having the formula:

Wherein R comprises a reactive ester, an anhydride, a carbamimidic anhydride, an acyl halide, an acyl azide, or a phosphonium ion and wherein n is 1, 2, 3, 4, 5 or 6; and wherein R' is selected from H and a protecting group.
230. The compound of claim 229, wherein R is a reactive ester.
231. The compound of claim 230, wherein R is wherein R2 is F or SO3-.
232. The compound of claim 230, wherein R is wherein R2 is H or SO3-.
233. The compound of claim 230, wherein R comprises wherein R3 is N or CH and R4 is H or Cl.
234. The compound of claim 230, wherein R comprises wherein R3 is N or CH.
235. The compound of claim 229, wherein R comprises
236. The compound of claim 229, wherein R is
237. The compound of claim 229, wherein R is
238. The compound of claim 229, wherein R is
239. The compound of any of claims 206-238, wherein R' is H.
240. The compound of any of claims 206-238, wherein R' is acetyl (¨C(O)CH3).
241. The compound of any of claims 206-238, wherein R' is benzoyl (¨C(O)C6H5).
242. A method comprising conjugation of the compound of any of claims 206-238 to an oligonucleotide, wherein the oligonucleotide comprises an amino group at the 5'-end.
CA2921514A 2013-05-01 2014-05-01 Compositions and methods for modulating apolipoprotein c-iii expression Active CA2921514C (en)

Applications Claiming Priority (15)

Application Number Priority Date Filing Date Title
US201361818442P 2013-05-01 2013-05-01
US61/818,442 2013-05-01
US201361823826P 2013-05-15 2013-05-15
US61/823,826 2013-05-15
US201361843887P 2013-07-08 2013-07-08
US61/843,887 2013-07-08
US201361871673P 2013-08-29 2013-08-29
US61/871,673 2013-08-29
US201361880790P 2013-09-20 2013-09-20
US61/880,790 2013-09-20
US201461976991P 2014-04-08 2014-04-08
US61/976,991 2014-04-08
US201461986867P 2014-04-30 2014-04-30
US61/986,867 2014-04-30
PCT/US2014/036462 WO2014179626A2 (en) 2013-05-01 2014-05-01 Compositions and methods for modulating apolipoprotein c-iii expression

Publications (2)

Publication Number Publication Date
CA2921514A1 true CA2921514A1 (en) 2014-11-06
CA2921514C CA2921514C (en) 2023-10-24

Family

ID=51843959

Family Applications (5)

Application Number Title Priority Date Filing Date
CA2921162A Pending CA2921162A1 (en) 2013-05-01 2014-05-01 Conjugated antisense compounds and their use
CA2921509A Pending CA2921509A1 (en) 2013-05-01 2014-05-01 Compositions and methods for modulating apolipoprotein (a) expression
CA2921167A Pending CA2921167A1 (en) 2013-05-01 2014-05-01 Compositions and methods for modulating hbv and ttr expression
CA2921514A Active CA2921514C (en) 2013-05-01 2014-05-01 Compositions and methods for modulating apolipoprotein c-iii expression
CA2921518A Abandoned CA2921518A1 (en) 2013-05-01 2014-05-01 Compositions and methods

Family Applications Before (3)

Application Number Title Priority Date Filing Date
CA2921162A Pending CA2921162A1 (en) 2013-05-01 2014-05-01 Conjugated antisense compounds and their use
CA2921509A Pending CA2921509A1 (en) 2013-05-01 2014-05-01 Compositions and methods for modulating apolipoprotein (a) expression
CA2921167A Pending CA2921167A1 (en) 2013-05-01 2014-05-01 Compositions and methods for modulating hbv and ttr expression

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA2921518A Abandoned CA2921518A1 (en) 2013-05-01 2014-05-01 Compositions and methods

Country Status (35)

Country Link
US (23) US9127276B2 (en)
EP (11) EP2991661B1 (en)
JP (20) JP6216444B2 (en)
KR (16) KR20210014758A (en)
CN (12) CN110066795A (en)
AU (19) AU2014259757B2 (en)
BR (5) BR112015027369B1 (en)
CA (5) CA2921162A1 (en)
CL (2) CL2015003217A1 (en)
CR (2) CR20190269A (en)
CY (2) CY1121879T1 (en)
DK (4) DK2992098T3 (en)
DO (3) DOP2015000268A (en)
EA (2) EA031393B1 (en)
ES (4) ES2885174T3 (en)
HK (5) HK1221485A1 (en)
HR (2) HRP20190987T1 (en)
HU (2) HUE050394T2 (en)
IL (18) IL296543A (en)
LT (2) LT2992009T (en)
ME (1) ME03390B (en)
MX (11) MX2015015264A (en)
MY (2) MY198359A (en)
NZ (6) NZ631537A (en)
PE (2) PE20152002A1 (en)
PH (3) PH12015502493B1 (en)
PL (2) PL2992009T3 (en)
PT (3) PT3524680T (en)
RS (2) RS60796B1 (en)
RU (8) RU2019124314A (en)
SG (5) SG11201508800WA (en)
SI (2) SI2992098T1 (en)
UA (2) UA121017C2 (en)
WO (5) WO2014179620A1 (en)
ZA (2) ZA201507218B (en)

Families Citing this family (410)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2009305636A1 (en) 2008-10-15 2010-04-22 Ionis Pharmaceuticals, Inc. Modulation of Factor 11 expression
KR101881596B1 (en) 2008-12-02 2018-07-24 웨이브 라이프 사이언시스 재팬 인코포레이티드 Method for the synthesis of phosphorous atom modified nucleic acids
IN2012DN00720A (en) 2009-07-06 2015-06-19 Ontorii Inc
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
EP2640853B1 (en) 2010-11-17 2018-12-26 Ionis Pharmaceuticals, Inc. Modulation of alpha synuclein expression
BR112014001244A2 (en) 2011-07-19 2017-02-21 Wave Life Sciences Pte Ltd methods for the synthesis of functionalized nucleic acids
EP3640332A1 (en) * 2011-08-29 2020-04-22 Ionis Pharmaceuticals, Inc. Oligomer-conjugate complexes and their use
EP2831231A1 (en) 2012-03-30 2015-02-04 Isis Pharmaceuticals, Inc. Compositions and methods for modulating tau expression for reducing seizure and modifying a neurodegenerative syndrome
WO2013159108A2 (en) * 2012-04-20 2013-10-24 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
WO2013173789A2 (en) 2012-05-17 2013-11-21 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions
US9574193B2 (en) 2012-05-17 2017-02-21 Ionis Pharmaceuticals, Inc. Methods and compositions for modulating apolipoprotein (a) expression
KR20170142997A (en) * 2012-05-24 2017-12-28 아이오니스 파마수티컬즈, 인코포레이티드 Methods and compositions for modulating apolipoprotein (a) expression
BR112015000784A8 (en) 2012-07-13 2018-04-03 Wave Life Sciences Japan ASYMMETRICAL AUXILIARY GROUP
EP2873674B1 (en) 2012-07-13 2020-05-06 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
EP2872147B1 (en) 2012-07-13 2022-12-21 Wave Life Sciences Ltd. Method for making chiral oligonucleotides
WO2014076195A1 (en) * 2012-11-15 2014-05-22 Santaris Pharma A/S Oligonucleotide conjugates
RU2649367C2 (en) * 2013-01-30 2018-04-02 Ф. Хоффманн-Ля Рош Аг Lna oligonucleotide carbohydrate conjugates
US9593333B2 (en) * 2013-02-14 2017-03-14 Ionis Pharmaceuticals, Inc. Modulation of apolipoprotein C-III (ApoCIII) expression in lipoprotein lipase deficient (LPLD) populations
MX371093B (en) 2013-05-01 2020-01-16 Regulus Therapeutics Inc Microrna compounds and methods for modulating mir-122.
EA031393B1 (en) * 2013-05-01 2018-12-28 Ионис Фармасьютикалз, Инк. Compositions and methods for modulating hbv and ttr expression
WO2014179445A1 (en) 2013-05-01 2014-11-06 Regulus Therapeutics Inc. Compounds and methods for enhanced cellular uptake
EA201592215A1 (en) 2013-06-27 2016-05-31 Рош Инновейшен Сентер Копенгаген А/С ANTISMINAL OLIGOMERS AND THEIR CONJUGATES AIMED AT PROPROTEIN CONVERTASY SUBTILYSINE / KEXIN TYPE 9 (PCSK9)
DK3019200T3 (en) * 2013-07-11 2022-06-20 Alnylam Pharmaceuticals Inc OLIGONUCLEOTIDE-LIGAND CONJUGATES AND METHOD OF PREPARATION
US10077236B2 (en) 2013-07-15 2018-09-18 The Regents Of The University Of California Azacyclic constrained analogs of FTY720
TWI657819B (en) 2013-07-19 2019-05-01 美商Ionis製藥公司 Compositions for modulating tau expression
US9943604B2 (en) 2013-09-20 2018-04-17 Ionis Pharmaceuticals, Inc. Targeted therapeutic nucleosides and their use
EP3052626A1 (en) * 2013-10-02 2016-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
US11162096B2 (en) 2013-10-14 2021-11-02 Ionis Pharmaceuticals, Inc Methods for modulating expression of C9ORF72 antisense transcript
US9994846B2 (en) * 2013-10-25 2018-06-12 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-21 activity
CA2928349A1 (en) * 2013-11-14 2015-05-21 Roche Innovation Center Copenhagen A/S Apob antisense conjugate compounds
JP6482475B2 (en) * 2014-01-07 2019-03-13 レナセラピューティクス株式会社 Double stranded oligonucleotides comprising antisense oligonucleotides and sugar derivatives
WO2015108047A1 (en) 2014-01-15 2015-07-23 株式会社新日本科学 Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
JPWO2015108046A1 (en) 2014-01-15 2017-03-23 株式会社新日本科学 Chiral nucleic acid adjuvant and antiallergic agent having antiallergic action
JPWO2015108048A1 (en) 2014-01-15 2017-03-23 株式会社新日本科学 Chiral nucleic acid adjuvant and antitumor agent having antitumor activity
KR20230152178A (en) 2014-01-16 2023-11-02 웨이브 라이프 사이언시스 리미티드 Chiral design
EP3960860A3 (en) 2014-02-11 2022-06-08 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
US10006027B2 (en) 2014-03-19 2018-06-26 Ionis Pharmaceuticals, Inc. Methods for modulating Ataxin 2 expression
AU2015231130C1 (en) 2014-03-19 2021-12-23 Ionis Pharmaceuticals, Inc. Compositions for modulating Ataxin 2 expression
AU2015240761B2 (en) 2014-04-01 2019-09-12 Biogen Ma Inc. Compositions for modulating SOD-1 expression
ES2812099T3 (en) 2014-05-01 2021-03-16 Ionis Pharmaceuticals Inc Compositions and methods for modulating growth hormone receptor expression
CA2946003A1 (en) 2014-05-01 2015-11-05 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating angiopoietin-like 3 expression
BR112016022593B1 (en) 2014-05-01 2022-04-26 Ionis Pharmaceuticals, Inc Oligomeric compounds, compositions comprising them, and uses thereof
EP3811977A1 (en) * 2014-05-01 2021-04-28 Ionis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
RU2703411C2 (en) 2014-05-01 2019-10-16 Ионис Фармасьютикалз, Инк. Compositions and methods for modulating pkk expression
GB201408623D0 (en) * 2014-05-15 2014-07-02 Santaris Pharma As Oligomers and oligomer conjugates
AU2015264038B2 (en) 2014-05-22 2021-02-11 Alnylam Pharmaceuticals, Inc. Angiotensinogen (AGT) iRNA compositions and methods of use thereof
GB201410693D0 (en) 2014-06-16 2014-07-30 Univ Southampton Splicing modulation
MA40463A (en) 2014-08-07 2017-06-14 Regulus Therapeutics Inc Targeting micrornas for metabolic disorders
JP6867945B2 (en) 2014-10-03 2021-05-12 コールド スプリング ハーバー ラボラトリー Targeted enhancement of nuclear gene output
SG11201702877TA (en) * 2014-10-10 2017-05-30 Hoffmann La Roche Galnac phosphoramidites, nucleic acid conjugates thereof and their use
WO2016061487A1 (en) * 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
AU2015350120B2 (en) * 2014-11-17 2021-05-27 Alnylam Pharmaceuticals, Inc. Apolipoprotein C3 (APOC3) iRNA compositions and methods of use thereof
WO2016100716A1 (en) 2014-12-18 2016-06-23 Vasant Jadhav Reversirtm compounds
US10793855B2 (en) 2015-01-06 2020-10-06 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of C9ORF72 antisense transcript
US10538763B2 (en) 2015-01-16 2020-01-21 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of DUX4
CA2976445A1 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2016128583A2 (en) * 2015-02-15 2016-08-18 Ribo Task Aps Acyl-amino-lna and/or hydrocarbyl-amino-lna oligonucleotides
US11129844B2 (en) 2015-03-03 2021-09-28 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating MECP2 expression
BR112017015864A2 (en) * 2015-04-03 2018-07-31 Ionis Pharmaceuticals, Inc. compounds and methods for modulating tmprss6 expression
WO2016164746A1 (en) 2015-04-08 2016-10-13 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
WO2016167780A1 (en) 2015-04-16 2016-10-20 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
US11249071B2 (en) 2015-04-24 2022-02-15 California Institute Of Technology Reactivation of x chromosome genes
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
EP3313989A4 (en) 2015-06-29 2018-12-05 Ionis Pharmaceuticals, Inc. Modified crispr rna and modified single crispr rna and uses thereof
US10494632B2 (en) 2015-07-10 2019-12-03 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (IGFALS) compositions and methods of use thereof
ES2917181T3 (en) 2015-07-10 2022-07-07 Ionis Pharmaceuticals Inc Diacylglycerol acyltransferase 2 (DGAT2) modulators
EP3323893A4 (en) * 2015-07-16 2019-03-27 Kyowa Hakko Kirin Co., Ltd. Beta2gpi gene expression inhibiting nucleic acid complex
MA43072A (en) 2015-07-22 2018-05-30 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
PL3331892T3 (en) 2015-08-06 2019-11-29 Hoffmann La Roche Processes for the preparation of galnac acid derivatives
CN105111118B (en) * 2015-08-14 2017-04-12 天津小新医药科技有限公司 L-menthol P2Y12 receptor antagonists, preparation method thereof and use thereof
CN105111119B (en) * 2015-08-14 2017-04-12 天津小新医药科技有限公司 Halogenobenzene L-menthol P2Y12 receptor antagonists and application thereof
KR20180043819A (en) 2015-08-24 2018-04-30 로슈 이노베이션 센터 코펜하겐 에이/에스 LNA-G method
CN114525280A (en) 2015-09-02 2022-05-24 阿尔尼拉姆医药品有限公司 iRNA compositions of programmed cell death 1 ligand 1(PD-L1) and methods of use thereof
RU2018113709A (en) 2015-09-24 2019-10-30 Айонис Фармасьютикалз, Инк. KRAS EXPRESSION MODULATORS
MX2018003685A (en) 2015-09-24 2018-08-15 Univ California Synthetic sphingolipid-like molecules, drugs, methods of their synthesis and methods of treatment.
US20180273948A1 (en) * 2015-09-25 2018-09-27 Tarveda Therapeutics, Inc. RNAi CONJUGATES, PARTICLES AND FORMULATIONS THEREOF
US10533175B2 (en) 2015-09-25 2020-01-14 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating Ataxin 3 expression
US20210052631A1 (en) * 2015-09-25 2021-02-25 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
EP3353306A4 (en) 2015-09-25 2019-06-05 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
JOP20210043A1 (en) * 2015-10-01 2017-06-16 Arrowhead Pharmaceuticals Inc Compositions and Methods for Inhibiting Gene Expression of LPA
US10577388B2 (en) 2015-10-02 2020-03-03 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugation process
SG11201802870RA (en) 2015-10-09 2018-05-30 Univ Southampton Modulation of gene expression and screening for deregulated protein expression
WO2017068087A1 (en) 2015-10-22 2017-04-27 Roche Innovation Center Copenhagen A/S Oligonucleotide detection method
WO2017067970A1 (en) 2015-10-22 2017-04-27 Roche Innovation Center Copenhagen A/S In vitro toxicity screening assay
US20190046555A1 (en) 2015-11-06 2019-02-14 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds for use in therapy
WO2017079739A1 (en) 2015-11-06 2017-05-11 Ionis Pharmaceuticals, Inc. MODULATING APOLIPOPROTEIN (a) EXPRESSION
WO2017084987A1 (en) * 2015-11-16 2017-05-26 F. Hoffmann-La Roche Ag GalNAc CLUSTER PHOSPHORAMIDITE
US11058709B1 (en) 2015-12-04 2021-07-13 Ionis Pharmaceuticals, Inc. Methods of treating breast cancer
EP3933041B1 (en) 2015-12-14 2024-01-31 Cold Spring Harbor Laboratory Antisense oligomers for treatment of autosomal dominant retardation
US11096956B2 (en) 2015-12-14 2021-08-24 Stoke Therapeutics, Inc. Antisense oligomers and uses thereof
AU2017205462A1 (en) 2016-01-05 2018-06-07 Ionis Pharmaceuticals, Inc. Methods for reducing LRRK2 expression
WO2017123696A1 (en) 2016-01-12 2017-07-20 Interleukin Genetics, Inc. Methods for predicting response to treatment
AU2017211461B2 (en) 2016-01-26 2023-01-12 Nissan Chemical Corporation Single-stranded oligonucleotide
WO2017131236A1 (en) * 2016-01-29 2017-08-03 協和発酵キリン株式会社 Nucleic acid complex
EP3408391A4 (en) 2016-01-31 2019-08-28 University of Massachusetts Branched oligonucleotides
JP6983797B2 (en) 2016-03-07 2021-12-17 アローヘッド ファーマシューティカルズ インコーポレイテッド Targeted ligand for therapeutic compounds
US11136577B2 (en) 2016-03-09 2021-10-05 Ionis Pharmaceuticals, Inc. Methods and compositions for inhibiting PMP22 expression
HUE053172T2 (en) 2016-03-14 2021-06-28 Hoffmann La Roche Oligonucleotides for reduction of pd-l1 expression
US10577607B2 (en) 2016-03-16 2020-03-03 Ionis Pharmaceuticals, Inc. Modulation of DYRK1B expression
CA3013799A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Methods of modulating keap1
EP3228326A1 (en) * 2016-04-05 2017-10-11 Silence Therapeutics GmbH Nucleic acid linked to a trivalent glycoconjugate
MA45478A (en) 2016-04-11 2019-02-20 Arbutus Biopharma Corp TARGETED NUCLEIC ACID CONJUGATE COMPOSITIONS
ES2933435T3 (en) 2016-04-13 2023-02-08 Ionis Pharmaceuticals Inc Methods to reduce the expression of C9ORF72
KR102468177B1 (en) 2016-04-14 2022-11-16 에프. 호프만-라 로슈 아게 Trityl-mono-GalNAc Compounds and Uses Thereof
MA45295A (en) 2016-04-19 2019-02-27 Alnylam Pharmaceuticals Inc HIGH DENSITY LIPOPROTEIN BINDING PROTEIN (HDLBP / VIGILINE) RNA COMPOSITION AND METHODS FOR USING THEM
SG11201809002RA (en) 2016-04-29 2018-11-29 Univ Nanyang Tech G-quadruplex-containing antisense oligonucleotides
MA45270A (en) 2016-05-04 2017-11-09 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
US11236339B2 (en) 2016-06-17 2022-02-01 Ionis Pharmaceuticals, Inc. Modulation of GYS1 expression
US11105794B2 (en) 2016-06-17 2021-08-31 Hoffmann-La Roche Inc. In vitro nephrotoxicity screening assay
WO2017216325A1 (en) 2016-06-17 2017-12-21 F. Hoffmann-La Roche Ag In vitro nephrotoxicity screening assay
MA45496A (en) 2016-06-17 2019-04-24 Hoffmann La Roche NUCLEIC ACID MOLECULES FOR PADD5 OR PAD7 MRNA REDUCTION FOR TREATMENT OF HEPATITIS B INFECTION
CN109415401B (en) * 2016-06-30 2023-02-03 协和麒麟株式会社 Nucleic acid complexes
AU2017296195A1 (en) 2016-07-11 2019-01-24 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
PL3484524T3 (en) 2016-07-15 2023-03-20 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of smn2
IL291323B2 (en) 2016-07-15 2023-09-01 Am Chemicals Llc Non-nucleosidic solid supports and phosphoramidite building blocks for oligonucleotide synthesis
CA3033368A1 (en) 2016-08-12 2018-02-15 University Of Massachusetts Conjugated oligonucleotides
KR20190058477A (en) * 2016-08-17 2019-05-29 솔스티스 바이올로직스, 리미티드 Polynucleotide construct
CN116942841A (en) 2016-09-02 2023-10-27 箭头药业股份有限公司 Targeting ligands
JOP20190065A1 (en) 2016-09-29 2019-03-28 Ionis Pharmaceuticals Inc Compounds and methods for reducing tau expression
KR20190065341A (en) 2016-10-06 2019-06-11 아이오니스 파마수티컬즈, 인코포레이티드 Method of joining oligomeric compounds
WO2018081661A1 (en) 2016-10-27 2018-05-03 California Institute Of Technology Hdac inhibitor compositions for reactivation of the x chromosome
JOP20190104A1 (en) 2016-11-10 2019-05-07 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn3 expression
WO2018089914A1 (en) * 2016-11-11 2018-05-17 Alios Biopharma, Inc. Oligonucleotide targeting strategy for hbv cccdna
TW202313978A (en) 2016-11-23 2023-04-01 美商阿尼拉製藥公司 Serpina1 irna compositions and methods of use thereof
AU2017368050A1 (en) 2016-11-29 2019-06-20 Puretech Lyt, Inc. Exosomes for delivery of therapeutic agents
EP3548620A4 (en) 2016-12-02 2020-07-22 Cold Spring Harbor Laboratory Modulation of lnc05 expression
WO2018110980A1 (en) * 2016-12-13 2018-06-21 주식회사 에이엠사이언스 Pharmaceutical composition for preventing or treating hepatitis b
KR20230166146A (en) 2016-12-16 2023-12-06 알닐람 파마슈티칼스 인코포레이티드 Methods for treating or preventing ttr-associated diseases using transthyretin(ttr) irna compositions
CN108239644B (en) * 2016-12-23 2021-05-28 苏州瑞博生物技术股份有限公司 Small interfering nucleic acid, pharmaceutical composition and application thereof
US10337070B2 (en) 2017-01-12 2019-07-02 Cardioforecast Ltd. Methods and kits for treating cardiovascular disease
US20190345496A1 (en) 2017-01-13 2019-11-14 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating relb expression
EP3568478A1 (en) 2017-01-13 2019-11-20 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating rel expression
WO2018130581A1 (en) 2017-01-13 2018-07-19 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating rela expression
US20190367920A1 (en) 2017-01-13 2019-12-05 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating nfkb1 expression
EP3568480A1 (en) 2017-01-13 2019-11-20 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating nfkb2 expression
WO2018143475A1 (en) 2017-02-06 2018-08-09 日産化学工業株式会社 Single-stranded oligonucleotide
WO2018165564A1 (en) * 2017-03-09 2018-09-13 Ionis Pharmaceuticals, Inc. Morpholino modified oligomeric compounds
EP3594346A4 (en) 2017-03-10 2020-12-16 National Center For Child Health And Development Antisense oligonucleotide and composition for prevention or treatment of glycogen storage disease type ia
JOP20190215A1 (en) * 2017-03-24 2019-09-19 Ionis Pharmaceuticals Inc Modulators of pcsk9 expression
EP3385272A1 (en) * 2017-04-05 2018-10-10 Silence Therapeutics GmbH Further novel oligonucleotide-ligand conjugates
SG11201909433XA (en) 2017-04-11 2019-11-28 Arbutus Biopharma Corp Targeted compositions
KR20200015895A (en) 2017-04-18 2020-02-13 알닐람 파마슈티칼스 인코포레이티드 How to treat a subject infected with hepatitis B virus (HBV)
WO2018215049A1 (en) 2017-05-23 2018-11-29 F. Hoffmann-La Roche Ag Process for galnac oligonucleotide conjugates
EP3633036A4 (en) * 2017-05-26 2021-03-17 National Cerebral and Cardiovascular Center Antisense nucleic acid targeting pcsk9
US11603532B2 (en) 2017-06-02 2023-03-14 Wave Life Sciences Ltd. Oligonucleotide compositions and methods of use thereof
EP3599844A4 (en) * 2017-06-07 2021-01-13 University of Massachusetts Anti-adam33 oligonucleotides and related methods
WO2019014530A1 (en) 2017-07-13 2019-01-17 Alnylam Pharmaceuticals Inc. Lactate dehydrogenase a (ldha) irna compositions and methods of use thereof
TW201920671A (en) 2017-07-26 2019-06-01 日商日產化學股份有限公司 Single-stranded oligonucleotide
TW201920668A (en) * 2017-08-02 2019-06-01 日商協和醱酵麒麟有限公司 Nucleic acid complex
WO2019027015A1 (en) * 2017-08-02 2019-02-07 協和発酵キリン株式会社 Nucleic acid complex
WO2019030313A2 (en) 2017-08-11 2019-02-14 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating ube3c expression
US11197884B2 (en) 2017-08-18 2021-12-14 Ionis Pharmaceuticals, Inc. Modulation of the notch signaling pathway for treatment of respiratory disorders
WO2019038228A1 (en) 2017-08-22 2019-02-28 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating tom1 expression
SG10202108375XA (en) 2017-08-25 2021-09-29 Stoke Therapeutics Inc Antisense oligomers for treatment of conditions and diseases
WO2019051173A1 (en) 2017-09-08 2019-03-14 Ionis Pharmaceuticals, Inc. Modulators of smad7 expression
AU2018332216A1 (en) * 2017-09-14 2020-03-26 Janssen Biopharma, Inc. Galnac derivatives
AU2018338787A1 (en) 2017-09-29 2020-04-16 Intellia Therapeutics, Inc. Compositions and methods for TTR gene editing and treating ATTR amyloidosis
WO2019073018A1 (en) 2017-10-13 2019-04-18 Roche Innovation Center Copenhagen A/S Methods for identifying improved stereodefined phosphorothioate oligonucleotide variants of antisense oligonucleotides utilising sub-libraries of partially stereodefined oligonucleotides
SG11202003461TA (en) 2017-10-16 2020-05-28 Hoffmann La Roche NUCLEIC ACID MOLECULE FOR REDUCTION OF PAPD5 AND PAPD7 mRNA FOR TREATING HEPATITIS B INFECTION
WO2019089922A1 (en) 2017-11-01 2019-05-09 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
TWI809004B (en) 2017-11-09 2023-07-21 美商Ionis製藥公司 Compounds and methods for reducing snca expression
US20200385719A1 (en) 2017-11-16 2020-12-10 Alnylam Pharmaceuticals, Inc. Kisspeptin 1 (kiss1) irna compositions and methods of use thereof
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
WO2019105403A1 (en) * 2017-12-01 2019-06-06 苏州瑞博生物技术有限公司 Nucleic acid, composition and conjugate containing the same, preparation method and use thereof
AU2018374219C1 (en) * 2017-12-01 2023-05-11 Suzhou Ribo Life Science Co., Ltd. Double-stranded oligonucleotide, composition and conjugate comprising double-stranded oligonucleotide, preparation method therefor and use thereof
JP7261494B2 (en) 2017-12-01 2023-04-20 スーチョウ リボ ライフ サイエンス カンパニー、リミテッド Nucleic acids, compositions and complexes containing said nucleic acids, preparation methods and uses
WO2019105419A1 (en) 2017-12-01 2019-06-06 苏州瑞博生物技术有限公司 Nucleic acid, composition and conjugate containing same, preparation method, and use
WO2019105435A1 (en) 2017-12-01 2019-06-06 苏州瑞博生物技术有限公司 Nucleic acid, composition and conjugate containing nucleic acid, preparation method and use
WO2019105437A1 (en) 2017-12-01 2019-06-06 苏州瑞博生物技术有限公司 Nucleic acid, composition and conjugate containing same, and preparation method and use
CN111050807A (en) * 2017-12-01 2020-04-21 苏州瑞博生物技术有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
CN111344408A (en) 2017-12-11 2020-06-26 哥本哈根罗氏创新中心 Oligonucleotides for modulating FNDC3B expression
WO2019115417A2 (en) 2017-12-12 2019-06-20 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating rb1 expression
EP3724206B1 (en) 2017-12-14 2023-06-28 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
JP2021508491A (en) 2017-12-18 2021-03-11 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. High Mobility Group Box-1 (HMGB1) iRNA Composition and How to Use It
BR112020012523A2 (en) 2017-12-21 2020-11-24 F. Hoffmann-La Roche Ag complementary diagnosis for htra1 rna antagonists
WO2019126641A2 (en) 2017-12-21 2019-06-27 Ionis Pharmaceuticals, Inc. Modulation of frataxin expression
KR20200104302A (en) 2017-12-22 2020-09-03 로슈 이노베이션 센터 코펜하겐 에이/에스 New thiophosphoramidite
EP4074724A1 (en) 2017-12-22 2022-10-19 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorodithioate internucleoside linkage
US20200332289A1 (en) 2017-12-22 2020-10-22 Roche Innovation Center Copenhagen A/S Gapmer oligonucleotides comprising a phosphorodithioate internucleoside linkage
CN109957566B (en) * 2017-12-26 2023-08-25 广州市锐博生物科技有限公司 Modified oligonucleotides and compounds useful for synthesizing modified oligonucleotides
US11485752B2 (en) * 2017-12-26 2022-11-01 Guangzhou Ribobio Co., Ltd. Modified oligonucleotides and compound that can be used for synthesizing same
AU2018394875B2 (en) * 2017-12-29 2023-08-03 Suzhou Ribo Life Science Co., Ltd. Conjugates and preparation and use thereof
EP3737758A1 (en) 2018-01-10 2020-11-18 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating pias4 expression
WO2019140102A1 (en) * 2018-01-10 2019-07-18 Translate Bio Ma, Inc. Compositions and methods for facilitating delivery of synthetic nucleic acids to cells
CR20200301A (en) 2018-01-12 2020-10-26 Roche Innovation Ct Copenhagen As Alpha-synuclein antisense oligonucleotides and uses thereof
EP3737761A1 (en) 2018-01-12 2020-11-18 Bristol-Myers Squibb Company Antisense oligonucleotides targeting alpha-synuclein and uses thereof
JP7455746B2 (en) 2018-01-12 2024-03-26 ブリストル-マイヤーズ スクイブ カンパニー Antisense oligonucleotides targeting alpha-synuclein and their uses
JP2021510295A (en) 2018-01-12 2021-04-22 ロシュ イノベーション センター コペンハーゲン エーエス Oligonucleotides for regulating GSK3B expression
CN111902537A (en) 2018-01-15 2020-11-06 Ionis制药公司 Modulators of DNM2 expression
WO2019141656A1 (en) 2018-01-17 2019-07-25 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating erc1 expression
JP2021511029A (en) 2018-01-18 2021-05-06 ロシュ イノベーション センター コペンハーゲン エーエス Antisense oligonucleotide targeting SREBP1
WO2019145386A1 (en) 2018-01-26 2019-08-01 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating csnk1d expression
US11332733B2 (en) 2018-02-12 2022-05-17 lonis Pharmaceuticals, Inc. Modified compounds and uses thereof
CN112041439A (en) 2018-02-14 2020-12-04 深度基因组学公司 Oligonucleotide therapy for wilson's disease
JP2021513861A (en) 2018-02-21 2021-06-03 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company CAMK2D antisense oligonucleotide and its use
BR112020016001A2 (en) 2018-03-02 2020-12-15 Ionis Pharmaceuticals, Inc. IRF4 EXPRESSION MODULATORS
WO2019169243A1 (en) 2018-03-02 2019-09-06 Ionis Pharmaceuticals, Inc. Compounds and methods for the modulation of amyloid-beta precursor protein
CN115976028A (en) 2018-03-09 2023-04-18 第一三共株式会社 Remedies for glycogen disease type Ia
WO2019183440A1 (en) 2018-03-22 2019-09-26 Ionis Pharmaceuticals, Inc. Methods for modulating fmr1 expression
SG11202009680XA (en) 2018-04-05 2020-10-29 Hoffmann La Roche Use of fubp1 inhibitors for treating hepatitis b virus infection
CN112041440A (en) 2018-04-11 2020-12-04 Ionis制药公司 Modulators of EZH2 expression
EP3790989A1 (en) 2018-05-07 2021-03-17 Roche Innovation Center Copenhagen A/S Massively parallel discovery methods for oligonucleotide therapeutics
SG11202010215TA (en) 2018-05-09 2020-11-27 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn3 expression
CU20200082A7 (en) 2018-05-09 2021-06-08 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS FOR REDUCING THE EXPRESSION OF FXI
TW202016304A (en) 2018-05-14 2020-05-01 美商阿尼拉製藥公司 Angiotensinogen (agt) irna compositions and methods of use thereof
CA3103429A1 (en) 2018-06-14 2019-12-19 Don W. Cleveland Compounds and methods for increasing stmn2 expression
WO2020006267A1 (en) 2018-06-27 2020-01-02 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing lrrk2 expression
PE20220013A1 (en) 2018-07-03 2022-01-11 Hoffmann La Roche OLIGONUCLEOTIDES TO MODULATE THE EXPRESSION OF TAU
WO2020007826A1 (en) 2018-07-05 2020-01-09 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting mbtps1
WO2020011745A2 (en) 2018-07-11 2020-01-16 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting cers6
WO2020011744A2 (en) 2018-07-11 2020-01-16 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting cers5
CN112534055A (en) 2018-07-13 2021-03-19 豪夫迈·罗氏有限公司 Oligonucleotides for modulating expression of RTEL1
AR115847A1 (en) 2018-07-25 2021-03-03 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS TO REDUCE THE EXPRESSION OF ATXN2
MX2021001019A (en) 2018-07-31 2021-04-19 Roche Innovation Ct Copenhagen As Oligonucleotides comprising a phosphorotrithioate internucleoside linkage.
KR20210041537A (en) 2018-07-31 2021-04-15 로슈 이노베이션 센터 코펜하겐 에이/에스 Oligonucleotides Containing Linkages Between Phosphorotrithioate Nucleosides
WO2020033748A1 (en) 2018-08-08 2020-02-13 Arcturus Therapeutics, Inc. Compositions and agents against nonalcoholic steatohepatitis
SG11202101288TA (en) 2018-08-10 2021-03-30 Univ Massachusetts Modified oligonucleotides targeting snps
SG11202100715WA (en) 2018-08-13 2021-02-25 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) dsRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
WO2020038377A1 (en) 2018-08-21 2020-02-27 苏州瑞博生物技术有限公司 Nucleic acid, pharmaceutical composition and conjugate containing nucleic acid, and use thereof
US20220160870A1 (en) 2018-08-28 2022-05-26 Roche Innovation Center Copenhagen A/S Neoantigen engineering using splice modulating compounds
JP2022500003A (en) 2018-09-18 2022-01-04 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Ketohexokinase (KHK) iRNA composition and its usage
TW202023573A (en) 2018-09-19 2020-07-01 美商Ionis製藥公司 Modulators of pnpla3 expression
EP3862024A4 (en) * 2018-09-30 2022-08-17 Suzhou Ribo Life Science Co., Ltd. Sirna conjugate, preparation method therefor and use thereof
EP3867382A4 (en) * 2018-10-18 2022-09-07 Murdoch University Antisense therapy for ptp1b related conditions
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
AU2019377275A1 (en) 2018-11-09 2021-03-25 Novartis Ag Method for reducing the risk of a cardiovascular event with conjugated antisense compounds targeting apo(a)
TW202028222A (en) 2018-11-14 2020-08-01 美商Ionis製藥公司 Modulators of foxp3 expression
AU2019380940A1 (en) 2018-11-15 2021-06-03 Ionis Pharmaceuticals, Inc. Modulators of IRF5 expression
CA3117694A1 (en) 2018-11-21 2020-05-28 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing prion expression
US20220025367A1 (en) 2018-11-23 2022-01-27 Sanofi Novel rna compositions and methods for inhibiting angptl8
AU2019390097A1 (en) 2018-11-30 2021-07-15 Kyowa Kirin Co., Ltd. Nucleic acid conjugate
AU2019406186A1 (en) 2018-12-20 2021-07-15 Praxis Precision Medicines, Inc. Compositions and methods for the treatment of KCNT1 related disorders
KR20210110839A (en) * 2018-12-28 2021-09-09 쑤저우 리보 라이프 사이언스 컴퍼니, 리미티드 Nucleic acids, compositions and conjugates containing nucleic acids, methods for their preparation and uses
GB201821269D0 (en) 2018-12-28 2019-02-13 Nippon Shinyaku Co Ltd Myostatin signal inhibitor
CN111377985B (en) * 2018-12-29 2023-11-10 苏州瑞博生物技术股份有限公司 Compounds and conjugates, methods of making and uses thereof
CN113330117A (en) * 2019-01-18 2021-08-31 苏州瑞博生物技术股份有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
KR20210122809A (en) 2019-01-31 2021-10-12 아이오니스 파마수티컬즈, 인코포레이티드 Regulators of YAP1 expression
MX2021009949A (en) 2019-02-20 2021-09-21 Roche Innovation Ct Copenhagen As Novel phosphoramidites.
TW202102516A (en) 2019-02-20 2021-01-16 丹麥商羅氏創新中心哥本哈根有限公司 Phosphonoacetate gapmer oligonucleotides
CN109799330B (en) * 2019-02-22 2021-03-02 华中科技大学同济医学院附属同济医院 Application of neuraminic acid and neuraminidase inhibitor in chronic heart failure
JP2022522430A (en) 2019-02-26 2022-04-19 ロシュ イノベーション センター コペンハーゲン エーエス Oligonucleotide formulation method
BR112021015323A2 (en) 2019-02-27 2021-10-05 Ionis Pharmaceuticals, Inc. MALAT1 EXPRESSION MODULATORS
WO2020172755A1 (en) * 2019-02-28 2020-09-03 Deep Genomics Incorporated Ligand clusters and methods of their use and preparation
WO2020205463A1 (en) 2019-03-29 2020-10-08 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating ube3a-ats
US20220177894A1 (en) 2019-04-02 2022-06-09 Proqr Therapeutics Ii B.V. Antisense oligonucleotides for immunotherapy
EA202192733A1 (en) 2019-04-03 2022-03-14 Бристол-Маерс Сквибб Компани ANGPTL2 ANTISENSE OLIGONUCLEOTIDES AND THEIR APPLICATIONS
JP2022533419A (en) 2019-05-22 2022-07-22 スーチョウ リボ ライフ サイエンス カンパニー、リミテッド Nucleic acids, drug compositions and conjugates and methods of preparation and uses
EP3974532A4 (en) * 2019-05-22 2024-01-24 Suzhou Ribo Life Science Co Ltd Nucleic acid, pharmaceutical composition, conjugate, preparation method, and use
KR20220011656A (en) 2019-05-22 2022-01-28 쑤저우 리보 라이프 사이언스 컴퍼니, 리미티드 Nucleic acids, pharmaceutical compositions, conjugates, methods of preparation, and uses
JP2022533421A (en) 2019-05-22 2022-07-22 スーチョウ リボ ライフ サイエンス カンパニー、リミテッド Nucleic acids, drug compositions and conjugates and methods of preparation and uses
US20220315929A1 (en) 2019-05-24 2022-10-06 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition and conjugate, preparation method therefor and use thereof
CN113795582A (en) 2019-05-24 2021-12-14 苏州瑞博生物技术股份有限公司 Nucleic acid, pharmaceutical composition and conjugate, preparation method and application
US20220186221A1 (en) 2019-05-24 2022-06-16 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition and conjugate, preparation method and use
WO2020243292A1 (en) 2019-05-28 2020-12-03 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing fus expression
TW202113079A (en) 2019-05-31 2021-04-01 美商艾利格斯醫療公司 Modified gapmer oligonucleotides and methods of use
JP2022534867A (en) 2019-06-04 2022-08-04 リジェネロン・ファーマシューティカルズ・インコーポレイテッド Non-human animals containing humanized TTR loci with beta slip mutations and methods of use
MX2021015758A (en) 2019-06-25 2022-04-01 Amgen Inc Purification methods for carbohydrate-linked oligonucleotides.
EP3956450A4 (en) 2019-07-26 2022-11-16 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating gfap
EP4007812A1 (en) 2019-08-01 2022-06-08 Alnylam Pharmaceuticals, Inc. Serpin family f member 2 (serpinf2) irna compositions and methods of use thereof
WO2021022108A2 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. CARBOXYPEPTIDASE B2 (CPB2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021030522A1 (en) 2019-08-13 2021-02-18 Alnylam Pharmaceuticals, Inc. SMALL RIBOSOMAL PROTEIN SUBUNIT 25 (RPS25) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
EP4013767A4 (en) * 2019-08-15 2023-10-25 Ionis Pharmaceuticals, Inc. Linkage modified oligomeric compounds and uses thereof
EP4022061A1 (en) 2019-08-27 2022-07-06 Sanofi Compositions and methods for inhibiting pcsk9
US20230076803A1 (en) 2019-08-29 2023-03-09 Suzhou Ribo Life Science Co., Ltd. Compound and drug conjugate, and preparation method and use thereof
EP4022062A1 (en) 2019-08-30 2022-07-06 Alnylam Pharmaceuticals, Inc. Neurofilament light chain (nfl) as a biomarker for transthyretin amyloidosis polyneuropathy
US20210060168A1 (en) * 2019-09-03 2021-03-04 Arcturus Therapeutics, Inc. Asialoglycoprotein receptor mediated delivery of therapeutically active conjugates
AU2020345317A1 (en) 2019-09-10 2022-04-21 Daiichi Sankyo Company, Limited CONJUGATE OF GalNAc-OLIGONUCLEOTIDE FOR DELIVERY TO LIVER AND MANUFACTURING METHOD THEREOF
CN114423430A (en) 2019-09-20 2022-04-29 豪夫迈·罗氏有限公司 Methods of treating HBV infection using allosteric modulators of core protein
CN114728017A (en) 2019-10-14 2022-07-08 阿斯利康(瑞典)有限公司 Modulators of PNPLA3 expression
EP4045652A1 (en) 2019-10-18 2022-08-24 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
TW202134435A (en) 2019-10-22 2021-09-16 美商阿尼拉製藥公司 Complement component c3 irna compositions and methods of use thereof
TW202132567A (en) 2019-11-01 2021-09-01 美商阿尼拉製藥公司 Huntingtin (htt) irna agent compositions and methods of use thereof
MX2022005692A (en) 2019-11-13 2022-06-08 Alnylam Pharmaceuticals Inc Methods and compositions for treating an angiotensinogen- (agt-) associated disorder.
EP4061945A1 (en) 2019-11-22 2022-09-28 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
CN112876534B (en) * 2019-11-29 2024-02-09 苏州瑞博生物技术股份有限公司 Liver targeting compounds and conjugates
KR20220115995A (en) 2019-12-13 2022-08-19 알닐람 파마슈티칼스 인코포레이티드 Human chromosome 9 open reading frame 72 (C9orf72) iRNA preparation compositions and methods of using the same
WO2021126734A1 (en) 2019-12-16 2021-06-24 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
CN111041025B (en) 2019-12-17 2021-06-18 深圳市瑞吉生物科技有限公司 mRNA targeting molecule based on combination of N-acetylgalactosamine polypeptide and preparation method thereof
EP4077667A1 (en) 2019-12-19 2022-10-26 F. Hoffmann-La Roche AG Use of sept9 inhibitors for treating hepatitis b virus infection
EP4077669A1 (en) 2019-12-19 2022-10-26 F. Hoffmann-La Roche AG Use of sbds inhibitors for treating hepatitis b virus infection
WO2021122993A1 (en) 2019-12-19 2021-06-24 F. Hoffmann-La Roche Ag Use of saraf inhibitors for treating hepatitis b virus infection
CN115516091A (en) 2019-12-19 2022-12-23 豪夫迈·罗氏有限公司 Use of COPS3 inhibitors for the treatment of hepatitis b virus infection
CN114829599A (en) 2019-12-19 2022-07-29 豪夫迈·罗氏有限公司 Use of SCAMP3 inhibitors for treating hepatitis b virus infection
US20230044958A1 (en) 2019-12-24 2023-02-09 Hoffmann-La Roche Inc. Method of treating virus infection using a tlr7 agonist
JP2023509872A (en) 2019-12-24 2023-03-10 エフ. ホフマン-ラ ロシュ アーゲー Pharmaceutical combinations of antiviral agents targeting HBV and/or immunomodulatory agents for the treatment of HBV
EP4081639A1 (en) 2019-12-24 2022-11-02 F. Hoffmann-La Roche AG Pharmaceutical combination of a therapeutic oligonucleotide targeting hbv and a tlr7 agonist for treatment of hbv
US20230151360A1 (en) * 2020-01-30 2023-05-18 Eisai R&D Management Co., Ltd. Nucleic acid complex and pharmaceutical composition containing same
IL295445A (en) 2020-02-10 2022-10-01 Alnylam Pharmaceuticals Inc Compositions and methods for silencing vegf-a expression
MX2022010052A (en) 2020-02-18 2022-09-05 Alnylam Pharmaceuticals Inc Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof.
CR20220485A (en) 2020-02-28 2022-11-10 Ionis Pharmaceuticals Inc Compounds and methods for modulating smn2
EP4114947A1 (en) 2020-03-05 2023-01-11 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
WO2021178778A1 (en) 2020-03-06 2021-09-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin (ttr)
WO2021178736A1 (en) 2020-03-06 2021-09-10 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP4121534A1 (en) 2020-03-18 2023-01-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
MX2022011801A (en) 2020-03-23 2022-10-07 Amgen Inc Monoclonal antibodies to chemically-modified nucleic acids and uses thereof.
WO2021193965A1 (en) * 2020-03-26 2021-09-30 国立研究開発法人国立循環器病研究センター Antisense nucleic acid targeting apoc3
WO2021195307A1 (en) 2020-03-26 2021-09-30 Alnylam Pharmaceuticals, Inc. Coronavirus irna compositions and methods of use thereof
CA3179411A1 (en) 2020-04-06 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing myoc expression
WO2021206917A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. ANGIOTENSIN-CONVERTING ENZYME 2 (ACE2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
CN116134135A (en) 2020-04-07 2023-05-16 阿尔尼拉姆医药品有限公司 Compositions and methods for silencing SCN9A expression
WO2021206922A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
CN111575279A (en) * 2020-04-27 2020-08-25 江苏为真生物医药技术股份有限公司 Method for capturing extrahepatic vesicle or circulating tumor cell by using ASGPR (adenosine triphosphate) small molecule ligand specificity
CA3181400A1 (en) 2020-04-27 2021-11-04 Alnylam Pharmaceuticals, Inc. Apolipoprotein e (apoe) irna agent compositions and methods of use thereof
WO2021222549A1 (en) 2020-04-30 2021-11-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
TW202206596A (en) 2020-05-01 2022-02-16 美商Ionis製藥公司 Compounds and methods for modulating atxn1
JP2023525799A (en) 2020-05-11 2023-06-19 ストーク セラピューティクス,インク. OPA1 antisense oligomers for treatment of conditions and diseases
EP4150077A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
EP4150089A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rs1)
WO2021231673A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
EP4150078A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
WO2021231675A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
EP4150087A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
WO2021231680A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
WO2021231692A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
AR122534A1 (en) 2020-06-03 2022-09-21 Triplet Therapeutics Inc METHODS FOR THE TREATMENT OF NUCLEOTIDE REPEAT EXPANSION DISORDERS ASSOCIATED WITH MSH3 ACTIVITY
WO2021252557A1 (en) 2020-06-09 2021-12-16 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
WO2021249484A1 (en) * 2020-06-10 2021-12-16 南京明德新药研发有限公司 Conjugate group and conjugate
BR112022024420A2 (en) 2020-06-18 2023-01-17 Alnylam Pharmaceuticals Inc XANTHINE DEHYDROGENASE (XDH) IRNA COMPOSITIONS AND METHODS OF USE THEREOF
AR122731A1 (en) 2020-06-26 2022-10-05 Hoffmann La Roche IMPROVED OLIGONUCLEOTIDES TO MODULATE FUBP1 EXPRESSION
BR112022024206A2 (en) 2020-06-29 2023-01-03 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS TO MODULATE PLP1
CN111744019B (en) 2020-07-01 2023-08-04 深圳瑞吉生物科技有限公司 Mannose-based mRNA targeted delivery system and application thereof
IL299771A (en) 2020-07-10 2023-03-01 Inst Nat Sante Rech Med Methods and compositions for treating epilepsy
EP4196584A2 (en) * 2020-08-13 2023-06-21 Amgen Inc. Rnai constructs and methods for inhibiting marc1 expression
WO2022038211A2 (en) 2020-08-21 2022-02-24 F. Hoffmann-La Roche Ag Use of a1cf inhibitors for treating hepatitis b virus infection
WO2022066847A1 (en) 2020-09-24 2022-03-31 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
EP4225917A1 (en) 2020-10-05 2023-08-16 Alnylam Pharmaceuticals, Inc. G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
JP2023546103A (en) 2020-10-16 2023-11-01 サノフイ Novel RNA compositions and methods for inhibiting ANGPTL3
US20240035029A1 (en) 2020-10-16 2024-02-01 Sanofi Rna compositions and methods for inhibiting lipoprotein(a)
WO2022087329A1 (en) 2020-10-23 2022-04-28 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
IL302709A (en) 2020-11-13 2023-07-01 Alnylam Pharmaceuticals Inc COAGULATION FACTOR V (F5) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US11447521B2 (en) 2020-11-18 2022-09-20 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
WO2022106695A1 (en) 2020-11-23 2022-05-27 Alpha Anomeric Sas Nucleic acid duplexes
WO2022125490A1 (en) 2020-12-08 2022-06-16 Alnylam Pharmaceuticals, Inc. Coagulation factor x (f10) irna compositions and methods of use thereof
GB2603454A (en) 2020-12-09 2022-08-10 Ucl Business Ltd Novel therapeutics for the treatment of neurodegenerative disorders
JP2024500035A (en) 2020-12-23 2024-01-04 アルゴノート アールエヌエー リミテッド Treatment of cardiovascular diseases
WO2022143531A1 (en) 2020-12-29 2022-07-07 苏州瑞博生物技术股份有限公司 Nucleic acid, pharmaceutical composition and sirna conjugate containing the nucleic acid, preparation method therefor, and use thereof
EP4274896A1 (en) 2021-01-05 2023-11-15 Alnylam Pharmaceuticals, Inc. Complement component 9 (c9) irna compositions and methods of use thereof
WO2022162161A1 (en) * 2021-01-30 2022-08-04 E-Therapeutics Plc Conjugated oligonucleotide compounds, methods of making and uses thereof
WO2022162157A1 (en) * 2021-01-30 2022-08-04 E-Therapeutics Plc Conjugated oligonucleotide compounds, methods of making and uses thereof
CA3204317A1 (en) * 2021-01-30 2022-08-04 Ahmad Ali MORTAZAVI Conjugated oligonucleotide compounds, methods of making and uses thereof
AU2022214281A1 (en) * 2021-01-30 2023-07-27 E-Therapeutics Plc Conjugated oligonucleotide compounds, methods of making and uses thereof
TW202246500A (en) 2021-02-02 2022-12-01 瑞士商赫孚孟拉羅股份公司 Enhanced oligonucleotides for inhibiting rtel1 expression
JP2024508714A (en) 2021-02-12 2024-02-28 アルナイラム ファーマシューティカルズ, インコーポレイテッド Superoxide dismutase 1 (SOD1) iRNA composition and method for using the same for treating or preventing superoxide dismutase 1- (SOD1-) related neurodegenerative diseases
JP2024508780A (en) * 2021-02-18 2024-02-28 ワングローブ ホールディングス リミテッド Compositions for conjugating oligonucleotides and carbohydrates
JP2024509783A (en) 2021-02-25 2024-03-05 アルナイラム ファーマシューティカルズ, インコーポレイテッド Prion protein (PRNP) IRNA compositions and methods of use thereof
KR20230150844A (en) 2021-02-26 2023-10-31 알닐람 파마슈티칼스 인코포레이티드 Ketohexokinase (KHK) iRNA compositions and methods of using the same
CA3212128A1 (en) 2021-03-04 2022-09-09 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
WO2022189363A1 (en) 2021-03-08 2022-09-15 Les Laboratoires Servier Antisense oligonucleotides for inhibiting alpha-synuclein expression
WO2022189861A1 (en) 2021-03-08 2022-09-15 Tollys Carbohydrate conjugates of tlr3 ligands and uses thereof
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
WO2022212231A2 (en) 2021-03-29 2022-10-06 Alnylam Pharmaceuticals, Inc. Huntingtin (htt) irna agent compositions and methods of use thereof
WO2022212153A1 (en) 2021-04-01 2022-10-06 Alnylam Pharmaceuticals, Inc. Proline dehydrogenase 2 (prodh2) irna compositions and methods of use thereof
IL307926A (en) 2021-04-26 2023-12-01 Alnylam Pharmaceuticals Inc Transmembrane protease, serine 6 (tmprss6) irna compositions and methods of use thereof
EP4330396A1 (en) 2021-04-29 2024-03-06 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
WO2022232650A1 (en) * 2021-04-30 2022-11-03 Ionis Pharmaceuticals, Inc. Methods for reducing agt expression
WO2022245583A1 (en) 2021-05-18 2022-11-24 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
EP4341405A1 (en) 2021-05-20 2024-03-27 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
EP4347823A1 (en) 2021-06-02 2024-04-10 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
CN117561334A (en) 2021-06-04 2024-02-13 阿尔尼拉姆医药品有限公司 Human chromosome 9 open reading frame 72 (C9 ORF 72) iRNA pharmaceutical compositions and methods of use thereof
WO2022260939A2 (en) 2021-06-08 2022-12-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating or preventing stargardt's disease and/or retinal binding protein 4 (rbp4)-associated disorders
CN117500815A (en) 2021-06-18 2024-02-02 Ionis制药公司 Compounds and methods for reducing IFNAR1 expression
CA3174095A1 (en) 2021-06-23 2022-12-29 Vignesh Narayan HARIHARAN Optimized anti-flt1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
WO2022271808A1 (en) * 2021-06-24 2022-12-29 Eli Lilly And Company Novel rna therapeutics and uses thereof
WO2022271806A1 (en) * 2021-06-24 2022-12-29 Eli Lilly And Company Novel therapeutic delivery moieties and uses thereof
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
KR20240026203A (en) 2021-06-30 2024-02-27 알닐람 파마슈티칼스 인코포레이티드 Methods and compositions for treating angiotensinogen (AGT)-related disorders
CN117377765A (en) * 2021-07-02 2024-01-09 上海拓界生物医药科技有限公司 Nucleic acid ligand and conjugate thereof, preparation method and application thereof
JPWO2023282345A1 (en) 2021-07-08 2023-01-12
TW202317147A (en) 2021-07-08 2023-05-01 日商日本新藥股份有限公司 Precipitation inhibitor
TW202308664A (en) 2021-07-08 2023-03-01 日商日本新藥股份有限公司 Nephrotoxicity-reducing agent
WO2023003805A1 (en) 2021-07-19 2023-01-26 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
WO2023003995A1 (en) 2021-07-23 2023-01-26 Alnylam Pharmaceuticals, Inc. Beta-catenin (ctnnb1) irna compositions and methods of use thereof
WO2023009687A1 (en) 2021-07-29 2023-02-02 Alnylam Pharmaceuticals, Inc. 3-hydroxy-3-methylglutaryl-coa reductase (hmgcr) irna compositions and methods of use thereof
WO2023015223A2 (en) * 2021-08-03 2023-02-09 Verve Therapeutics, Inc. Compositions and methods for targeted rna delivery
WO2023014677A1 (en) 2021-08-03 2023-02-09 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof
KR20240042016A (en) 2021-08-04 2024-04-01 알닐람 파마슈티칼스 인코포레이티드 iRNA compositions and methods for silencing angiotensinogen (AGT)
IL310407A (en) 2021-08-13 2024-03-01 Alnylam Pharmaceuticals Inc Factor xii (f12) irna compositions and methods of use thereof
WO2023041508A2 (en) 2021-09-14 2023-03-23 Argonaute RNA Limited Treatment of cardiovascular disease
WO2023044370A2 (en) 2021-09-17 2023-03-23 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing complement component 3 (c3)
CA3232191A1 (en) * 2021-09-18 2023-03-23 Chong Li Lpa inhibitor and use thereof
CA3232420A1 (en) 2021-09-20 2023-03-23 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
TW202327658A (en) * 2021-09-23 2023-07-16 大陸商上海舶望製藥有限公司 Multivalent ligand clusters with diamine scaffold for targeted delivery of therapeutic agent
CA3234835A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
TW202334418A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Huntingtin (htt) irna agent compositions and methods of use thereof
CA3234636A1 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
WO2023083906A2 (en) 2021-11-11 2023-05-19 F. Hoffmann-La Roche Ag Pharmaceutical combinations for treatment of hbv
WO2023102188A1 (en) * 2021-12-03 2023-06-08 Quralis Corporation Gapmer antisense oligonucleotides with modified backbone chemistries
GB202117758D0 (en) 2021-12-09 2022-01-26 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
TW202340466A (en) * 2021-12-16 2023-10-16 大陸商上海拓界生物醫藥科技有限公司 Dsrna, preparation method and application thereof
TW202340465A (en) * 2021-12-16 2023-10-16 大陸商上海拓界生物醫藥科技有限公司 Lpa-targeting sirna and conjugates
WO2023111210A1 (en) 2021-12-17 2023-06-22 F. Hoffmann-La Roche Ag Combination of oligonucleotides for modulating rtel1 and fubp1
WO2023138659A1 (en) * 2022-01-20 2023-07-27 上海拓界生物医药科技有限公司 Dsrna, use thereof and preparation method therefor
TW202345865A (en) * 2022-01-24 2023-12-01 大陸商上海舶望製藥有限公司 Composition and method for inhibiting expression of protein LPA(Apo(a))
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
CN117756866A (en) * 2022-01-30 2024-03-26 大睿生物医药科技(上海)有限公司 Targeting ligands containing N-acetylgalactosamine
TW202346587A (en) 2022-02-21 2023-12-01 瑞士商赫孚孟拉羅股份公司 Antisense oligonucleotide
CN114703184A (en) * 2022-03-11 2022-07-05 厦门甘宝利生物医药有限公司 LPA inhibitor and use thereof
TW202400787A (en) 2022-03-16 2024-01-01 美商安彼瑞可股份有限公司 Galnac compositions for improving sirna bioavailability
CN115028670B (en) * 2022-06-24 2023-07-28 四川大学华西医院 Preparation method of N-acetyl-D-galactosamine trimer precursor
WO2024001172A1 (en) * 2022-06-27 2024-01-04 Ractigen Therapeutics Oligonucleotide modulators activating complement factor h expression
WO2024013360A1 (en) 2022-07-15 2024-01-18 Proqr Therapeutics Ii B.V. Chemically modified oligonucleotides for adar-mediated rna editing
WO2024013361A1 (en) 2022-07-15 2024-01-18 Proqr Therapeutics Ii B.V. Oligonucleotides for adar-mediated rna editing and use thereof
CN116814621A (en) * 2022-08-05 2023-09-29 厦门甘宝利生物医药有限公司 RNA inhibitor for inhibiting APOC3 gene expression and application thereof
WO2024039776A2 (en) 2022-08-18 2024-02-22 Alnylam Pharmaceuticals, Inc. Universal non-targeting sirna compositions and methods of use thereof
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof
CN116925160B (en) * 2023-09-15 2023-12-08 天津全和诚科技有限责任公司 GalNAc sugar-containing ring intermediate and preparation method thereof

Family Cites Families (374)

* 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
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US5132418A (en) 1980-02-29 1992-07-21 University Patents, Inc. Process for preparing 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
US4973679A (en) 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
US4668777A (en) 1981-03-27 1987-05-26 University Patents, Inc. Phosphoramidite nucleoside compounds
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
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
DE3329892A1 (en) 1983-08-18 1985-03-07 Köster, Hubert, Prof. Dr., 2000 Hamburg METHOD FOR PRODUCING OLIGONUCLEOTIDES
US5118800A (en) 1983-12-20 1992-06-02 California Institute Of Technology Oligonucleotides possessing a primary amino group in the terminal nucleotide
USRE34036E (en) 1984-06-06 1992-08-18 National Research Development Corporation Data transmission using a transparent tone-in band system
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
US4751219A (en) 1985-02-05 1988-06-14 Nederlandse Centrale Organisatie Voor Toegepast-Natuur-Wetenschappelijk Onderzoek Synthetic glycolipides, a process for the preparation thereof and several uses for these synthetic glycolipides
US5506337A (en) 1985-03-15 1996-04-09 Antivirals Inc. Morpholino-subunit combinatorial library and method
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5405938A (en) 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5166315A (en) 1989-12-20 1992-11-24 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
DE3788914T2 (en) 1986-09-08 1994-08-25 Ajinomoto Kk Compounds for cleaving RNA at a specific position, oligomers used in the preparation of these compounds and starting materials for the synthesis of these oligomers.
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
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
GB8712540D0 (en) * 1987-05-28 1987-07-01 Ucb Sa Expression of human proapolipoprotein a-i
DE3851889T2 (en) 1987-06-24 1995-04-13 Florey Howard Inst 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
ATE151467T1 (en) 1987-11-30 1997-04-15 Univ Iowa Res Found DNA MOLECULES STABILIZED BY MODIFICATIONS TO THE 3'-TERMINAL PHOSPHODIESTER BOND, THEIR USE AS NUCLEIC ACID PROBE AND AS THERAPEUTIC AGENTS FOR INHIBITING THE EXPRESSION OF SPECIFIC TARGET GENES
US5403711A (en) 1987-11-30 1995-04-04 University Of Iowa Research Foundation Nucleic acid hybridization and amplification method for detection of specific sequences in which a complementary labeled nucleic acid probe is cleaved
EP0406309A4 (en) 1988-03-25 1992-08-19 The 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
US5256775A (en) 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
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
EP0942000B1 (en) 1989-10-24 2004-06-23 Isis Pharmaceuticals, Inc. 2'-Modified oligonucleotides
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US6005087A (en) 1995-06-06 1999-12-21 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US5623065A (en) 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US7101993B1 (en) 1990-01-11 2006-09-05 Isis Pharmaceuticals, Inc. Oligonucleotides containing 2′-O-modified purines
US5457191A (en) 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5859221A (en) 1990-01-11 1999-01-12 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US5220007A (en) 1990-02-15 1993-06-15 The Worcester Foundation For Experimental Biology Method of site-specific alteration of RNA and production of encoded polypeptides
US5149797A (en) 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
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
DK0455905T3 (en) 1990-05-11 1998-12-07 Microprobe Corp Dipsticks for nucleic acid hybridization assays and method for covalent immobilization of oligonucleotides
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
ATE154246T1 (en) 1990-07-27 1997-06-15 Isis Pharmaceuticals Inc NUCLEASE RESISTANT PYRIMIDINE MODIFIED OLIGONUCLEOTIDES THAT DETECTE AND MODULATE GENE EXPRESSION
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
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
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
WO1992002534A2 (en) 1990-08-03 1992-02-20 Sterling Drug, 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
US6582908B2 (en) 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
US5948903A (en) 1991-01-11 1999-09-07 Isis Pharmaceuticals, Inc. Synthesis of 3-deazapurines
US5672697A (en) 1991-02-08 1997-09-30 Gilead Sciences, Inc. Nucleoside 5'-methylene phosphonates
US7015315B1 (en) 1991-12-24 2006-03-21 Isis Pharmaceuticals, Inc. Gapped oligonucleotides
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
DE637965T1 (en) 1991-11-26 1995-12-14 Gilead Sciences Inc INCREASED FORMATION OF TRIPLE AND DOUBLE HELICOS FROM OLIGOMERS WITH 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
EP1695979B1 (en) * 1991-12-24 2011-07-06 Isis Pharmaceuticals, Inc. Gapped modified oligonucleotides
US5700922A (en) 1991-12-24 1997-12-23 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
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
US5652355A (en) 1992-07-23 1997-07-29 Worcester Foundation For Experimental Biology Hybrid oligonucleotide phosphorothioates
JPH08502723A (en) 1992-07-27 1996-03-26 ハイブライドン インコーポレイテッド Oligonucleotide alkylphosphonothioate
JPH08504559A (en) 1992-12-14 1996-05-14 ハネウエル・インコーポレーテッド Motor system with individually controlled redundant windings
CN1121721A (en) 1993-01-25 1996-05-01 海布里顿公司 Oligonucleotido alky phosphate and alky substituted phosphate
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
GB9304620D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Compounds
EP0691968B1 (en) 1993-03-30 1997-07-16 Sanofi Acyclic nucleoside analogs and oligonucleotide sequences containing them
EP0691977B1 (en) 1993-03-31 1997-11-26 Sanofi Oligonucleotides with amide linkages replacing phosphodiester linkages
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5801154A (en) 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
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
DE69425903T2 (en) 1993-12-09 2001-02-15 Thomas Jefferson University Ph CONNECTIONS AND METHOD FOR LOCATION-SPECIFIC MUTATION IN EUKARYOTIC CELLS
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5728518A (en) 1994-01-12 1998-03-17 The Immune Response Corporation Antiviral poly-and oligonucleotides
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
US5599706A (en) 1994-09-23 1997-02-04 Stinchcomb; Dan T. Ribozymes targeted to apo(a) mRNA
US5681940A (en) 1994-11-02 1997-10-28 Icn Pharmaceuticals Sugar modified nucleosides and oligonucleotides
US6908903B1 (en) 1994-12-07 2005-06-21 Aletheon Pharmaceuticals, Inc. Cluster clearing agents
US6172045B1 (en) 1994-12-07 2001-01-09 Neorx Corporation Cluster clearing agents
US5652356A (en) 1995-08-17 1997-07-29 Hybridon, Inc. Inverted chimeric and hybrid oligonucleotides
US20030119724A1 (en) 1995-11-22 2003-06-26 Ts`O Paul O.P. Ligands to enhance cellular uptake of biomolecules
US5994517A (en) 1995-11-22 1999-11-30 Paul O. P. Ts'o Ligands to enhance cellular uptake of biomolecules
US5998203A (en) 1996-04-16 1999-12-07 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids containing 5'-and/or 3'-cap structures
US6620916B1 (en) 1996-09-26 2003-09-16 Ajinomoto Co., Inc. Modified physiologically active proteins and medicinal compositions containing the same
JP3756313B2 (en) 1997-03-07 2006-03-15 武 今西 Novel bicyclonucleosides and oligonucleotide analogues
US6770748B2 (en) 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
USRE44779E1 (en) 1997-03-07 2014-02-25 Santaris Pharma A/S Bicyclonucleoside and oligonucleotide analogues
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
CA2303299C (en) 1997-09-12 2016-02-23 Exiqon A/S Oligonucleotide analogues
US7572582B2 (en) 1997-09-12 2009-08-11 Exiqon A/S Oligonucleotide analogues
US20040171564A1 (en) 1997-11-20 2004-09-02 Honkanen Richard E. Antisense oligonucleotide modulation of human serine/threonine protein phosphatase gene expression
US20030228597A1 (en) 1998-04-13 2003-12-11 Cowsert Lex M. Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
US6300319B1 (en) 1998-06-16 2001-10-09 Isis Pharmaceuticals, Inc. Targeted oligonucleotide conjugates
US6043352A (en) 1998-08-07 2000-03-28 Isis Pharmaceuticals, Inc. 2'-O-Dimethylaminoethyloxyethyl-modified oligonucleotides
RU2249463C2 (en) * 1998-08-19 2005-04-10 Бакстер Хелфкэа С.А. Immunogenic conjugate of beta-propionamide-bound polysaccharide with protein applied as vaccine
DE69941574D1 (en) 1998-08-19 2009-12-03 Baxter Healthcare Sa IMMUNOGENOUS BETA-PROPIONAMIDO-BONDED POLYSACCHARID PROTEIN CONJUGATE IS SUITABLE FOR VACCINATION AND MANUFACTURED BY USING N-ACRYLOYLATED POLYSACCHARIDE
US6166239A (en) 1998-09-04 2000-12-26 Isis Pharmaceuticals, Inc. Oligonucleotide protecting groups
EP1152009B2 (en) 1999-02-12 2017-09-06 Daiichi Sankyo Company, Limited Novel nucleosides and oligonucleotide analogues
US20030170249A1 (en) 1999-02-19 2003-09-11 Hakomori Sen-Itiroh Vaccines directed to cancer-associated carbohydrate antigens
US7084125B2 (en) 1999-03-18 2006-08-01 Exiqon A/S Xylo-LNA analogues
US7098192B2 (en) * 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
US6159694A (en) 1999-04-08 2000-12-12 Isis Pharmaceuticals Inc. Antisense modulation of stat3 expression
JP2002542263A (en) 1999-04-21 2002-12-10 ワイス Methods and compositions for inhibiting the function of a polynucleotide sequence
ATE356824T1 (en) 1999-05-04 2007-04-15 Santaris Pharma As L-RIBO-LNA ANALOGUE
US6525191B1 (en) 1999-05-11 2003-02-25 Kanda S. Ramasamy Conformationally constrained L-nucleosides
US6383812B1 (en) 1999-05-28 2002-05-07 Academia Sinica Anti liver disease drug R-YEEE and method of synthesizing branched galactose-terminal glycoproteins
US8541548B2 (en) 1999-06-07 2013-09-24 Arrowhead Madison Inc. Compounds and methods for reversible modification of biologically active molecules
US20080281041A1 (en) 1999-06-07 2008-11-13 Rozema David B Reversibly Masked Polymers
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
CA2379152A1 (en) 1999-07-16 2001-01-25 Hyseq, Inc. Novel angiopoietin materials and methods
JP4151751B2 (en) 1999-07-22 2008-09-17 第一三共株式会社 New bicyclonucleoside analogues
DE19935303A1 (en) * 1999-07-28 2001-02-08 Aventis Pharma Gmbh Oligonucleotides to inhibit the expression of human eg5
US20020082227A1 (en) 1999-09-30 2002-06-27 Scott Henry Use of oligonucleotides for inhibition of complement activation
US20050112118A1 (en) 1999-12-02 2005-05-26 Myriad Genetics, Incorporated Compositions and methods for treating inflammatory disorders
RU2002115295A (en) 1999-12-09 2004-01-10 Санкио Компани, Лимитед (Jp) A method of testing a therapeutic or prophylactic agent for hyperlipidemia
JP2003519231A (en) 1999-12-30 2003-06-17 カー・ユ・ルーベン・リサーチ・アンド・ディベロップメント Cyclohexene nucleic acid
US7179796B2 (en) 2000-01-18 2007-02-20 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US6261840B1 (en) 2000-01-18 2001-07-17 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US6602857B1 (en) 2000-01-18 2003-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US20020055479A1 (en) 2000-01-18 2002-05-09 Cowsert Lex M. Antisense modulation of PTP1B expression
US7833992B2 (en) 2001-05-18 2010-11-16 Merck Sharpe & Dohme Conjugates and compositions for cellular delivery
US7491805B2 (en) 2001-05-18 2009-02-17 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US8202979B2 (en) 2002-02-20 2012-06-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid
WO2001060860A2 (en) 2000-02-17 2001-08-23 Millennium Predictive Medicine, Inc. Genes differentially expressed in human prostate cancer and their use
EP1314734A1 (en) 2000-08-29 2003-05-28 Takeshi Imanishi Novel nucleoside analogs and oligonucleotide derivatives containing these analogs
US6426220B1 (en) 2000-10-30 2002-07-30 Isis Pharmaceuticals, Inc. Antisense modulation of calreticulin expression
CN100406065C (en) 2000-12-01 2008-07-30 细胞工厂治疗公司 Conjugates of glycosylated/galactosylated peptide, bifunctional linker and nucleotidic monomers/polymers, and related compositions and method of use
US20030077829A1 (en) 2001-04-30 2003-04-24 Protiva Biotherapeutics Inc.. Lipid-based formulations
CA2450022A1 (en) 2001-06-08 2002-12-19 Sankyo Company, Limited Method of testing drug for treating or preventing diseases such as hyperlipemia
CA2452458A1 (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
US20030175906A1 (en) 2001-07-03 2003-09-18 Muthiah Manoharan Nuclease resistant chimeric oligonucleotides
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US7227014B2 (en) * 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
US7259150B2 (en) 2001-08-07 2007-08-21 Isis Pharmaceuticals, Inc. Modulation of apolipoprotein (a) expression
WO2003014397A1 (en) 2001-08-09 2003-02-20 Biomedlab Corporation Probe for detection of enteric virus detection kit and method for enteric virus with the same
EP1446412B1 (en) 2001-09-04 2012-03-07 Exiqon A/S Novel lna compositions and uses thereof
US7439043B2 (en) 2001-10-10 2008-10-21 Neose Technologies, Inc. Galactosyl nucleotide sugars
EP1451578B1 (en) 2001-11-16 2013-08-21 Genentech, Inc. Use of angptl3 antagonists for the treatment of liver diseases
US20100240730A1 (en) 2002-02-20 2010-09-23 Merck Sharp And Dohme Corp. RNA Interference Mediated Inhibition of Gene Expression Using Chemically Modified Short Interfering Nucleic Acid (siNA)
AU2003274906A1 (en) 2002-07-31 2004-02-16 Nucleonics, Inc. Double stranded rna structures and constructs, and methods for generating and using the same
AU2003261449A1 (en) 2002-08-07 2004-02-25 Compositions for rna interference and methods of use thereof
EP1543019A2 (en) 2002-09-11 2005-06-22 Santaris Pharma A/S Modified pna molecules
US20060035344A1 (en) 2002-10-18 2006-02-16 Pachuk Catherine J Double-stranded rna structures and constructs, and methods for generating and using the same
EP1578765A4 (en) 2002-11-05 2008-04-23 Isis Pharmaceuticals Inc Sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
AU2003291753B2 (en) 2002-11-05 2010-07-08 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
US7511131B2 (en) 2002-11-13 2009-03-31 Genzyme Corporation Antisense modulation of apolipoprotein B expression
EP1569695B1 (en) 2002-11-13 2013-05-15 Genzyme Corporation Antisense modulation of apolipoprotein b expression
US20060009410A1 (en) 2002-11-13 2006-01-12 Crooke Rosanne M Effects of apolipoprotein B inhibition on gene expression profiles in animals
JP2006507841A (en) 2002-11-14 2006-03-09 ダーマコン, インコーポレイテッド Functional and ultrafunctional siRNA
JP5479663B2 (en) 2002-12-20 2014-04-23 セレラ コーポレーション Genetic polymorphism associated with myocardial infarction, detection method and use thereof
US6673661B1 (en) 2002-12-20 2004-01-06 Taiwan Semiconductor Manufacturing Co., Ltd. Self-aligned method for forming dual gate thin film transistor (TFT) device
WO2004063208A1 (en) 2003-01-09 2004-07-29 Postech Foundation New phosphoramidite compounds
WO2004072046A2 (en) 2003-02-12 2004-08-26 Carex S.A. Quinoline derivatives and their use for modulation of lxr activity
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
AU2004220556B2 (en) * 2003-03-07 2009-05-07 Alnylam Pharmaceuticals, Inc. Therapeutic compositions
EP1608735A4 (en) * 2003-04-03 2008-11-05 Alnylam Pharmaceuticals Irna conjugates
US7598227B2 (en) * 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
US7851615B2 (en) 2003-04-17 2010-12-14 Alnylam Pharmaceuticals, Inc. Lipophilic conjugated iRNA agents
US7723509B2 (en) 2003-04-17 2010-05-25 Alnylam Pharmaceuticals IRNA agents with biocleavable tethers
EP2664672A1 (en) 2003-04-17 2013-11-20 Alnylam Pharmaceuticals Inc. Modified iRNA agents
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
US7750142B2 (en) 2003-04-28 2010-07-06 Isis Pharmaceuticals, Inc. Modulation of glucagon receptor expression
JPWO2004101619A1 (en) 2003-05-15 2006-10-26 塩野義製薬株式会社 Rational design and synthesis of functional glycopeptides
WO2004106356A1 (en) 2003-05-27 2004-12-09 Syddansk Universitet Functionalized nucleotide derivatives
US7427672B2 (en) 2003-08-28 2008-09-23 Takeshi Imanishi Artificial nucleic acids of n-o bond crosslinkage type
EP2182063A3 (en) 2003-09-18 2012-07-18 Isis Pharmaceuticals, Inc. Modulation of EIF4E expression
AU2004274021B2 (en) 2003-09-18 2009-08-13 Isis Pharmaceuticals, Inc. 4'-thionucleosides and oligomeric compounds
US7959919B2 (en) 2003-11-19 2011-06-14 Novelmed Therapeutics, Inc. Method of inhibiting factor B-mediated complement activation
WO2005065686A1 (en) 2004-01-07 2005-07-21 Adipogen Pharmaceuticals Pty Limited Differentiation modulating agents and uses therefor
EP1711606A2 (en) 2004-01-20 2006-10-18 Isis Pharmaceuticals, Inc. Modulation of glucocorticoid receptor expression
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
WO2005097155A1 (en) 2004-04-08 2005-10-20 Takara Bio Inc. Neurite elongation inducing agent
CA2566286A1 (en) 2004-05-11 2005-12-08 Rnai Co., Ltd. Polynucleotide causing rna interfere and method of regulating gene expression with the use of the same
AU2005252662B2 (en) 2004-06-03 2011-08-18 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
DK1781698T3 (en) 2004-07-20 2016-10-03 Genentech Inc COMPOSITIONS AND METHODS FOR THE USE OF Angiopoietin-like-4-PROTEIN
EP1771474B1 (en) 2004-07-20 2010-01-27 Genentech, Inc. Inhibitors of angiopoietin-like 4 protein, combinations, and their use
DK2997980T3 (en) 2004-08-10 2018-03-26 Kastle Therapeutics Llc PROCEDURES FOR MODULATING LIPROPROTEIN AND CHOLESTEROL LEVELS
EP2990410A1 (en) 2004-08-10 2016-03-02 Alnylam Pharmaceuticals Inc. Chemically modified oligonucleotides
US20090203132A1 (en) 2004-09-09 2009-08-13 Swayze Eric E Pyrrolidinyl groups for attaching conjugates to oligomeric compounds
US20090036355A1 (en) 2004-10-13 2009-02-05 Sanjay Bhanot Antisense Modulation of PTP1B Expression
EP1812569A2 (en) 2004-11-08 2007-08-01 K.U. Leuven Research and Development Modified nucleosides for rna interference
US20060148740A1 (en) 2005-01-05 2006-07-06 Prosensa B.V. Mannose-6-phosphate receptor mediated gene transfer into muscle cells
WO2006078217A1 (en) 2005-01-24 2006-07-27 Avaris Ab COMPLEX CONTAINING SiRNA, ShRNA OR ANTISENSE MOLECULE AND FUNCTIONAL ENTITY, FOR IMPROVED SPECIFICITY AND DELIVERY
WO2007031081A2 (en) * 2005-09-15 2007-03-22 Santaris Pharma A/S RNA ANTAGONIST COMPOUNDS FOR THE INHIBITION OF APO-Bl00 EXPRESSION
EP1937813A2 (en) 2005-09-19 2008-07-02 Isis Pharmaceuticals, Inc. Modulation of glucagon receptor expression
US20070066557A1 (en) 2005-09-19 2007-03-22 Monia Brett P Modulation of glucocorticoid receptor expression
PT2161038E (en) 2006-01-26 2014-03-10 Isis Pharmaceuticals Inc Compositions and their uses directed to huntingtin
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
EP2314594B1 (en) 2006-01-27 2014-07-23 Isis Pharmaceuticals, Inc. 6-modified bicyclic nucleic acid analogs
US20090326041A1 (en) * 2006-05-05 2009-12-31 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression of sglt2
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
DK2066684T3 (en) 2006-05-11 2012-10-22 Isis Pharmaceuticals Inc 5'-Modified Bicyclic Nucleic Acid Analogs
EP2057284A4 (en) 2006-08-04 2011-06-29 Isis Pharmaceuticals Inc Compositions and methods for the modulation of jnk proteins
US8658211B2 (en) 2006-08-18 2014-02-25 Arrowhead Madison Inc. Polyconjugates for in vivo delivery of polynucleotides
AU2007285782B2 (en) 2006-08-18 2010-06-24 Arrowhead Research Corporation Polyconjugates for in vivo delivery of polynucleotides
CA2663601C (en) 2006-09-22 2014-11-25 Dharmacon, Inc. Duplex oligonucleotide complexes and methods for gene silencing by rna interference
AU2007310989B2 (en) 2006-10-18 2014-05-29 Isis Pharmaceuticals, Inc. Antisense compounds
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
CA2670563A1 (en) 2006-11-27 2008-06-05 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
PT2121751T (en) 2006-12-08 2017-04-18 Lexicon Pharmaceuticals Inc Monoclonal antibodies against angptl3
CA2839162A1 (en) * 2006-12-20 2008-06-26 Xoma Technology Ltd. Methods for the treatment of il-1-.beta. related diseases
US20100190837A1 (en) 2007-02-15 2010-07-29 Isis Pharmaceuticals, Inc. 5'-Substituted-2-F' Modified Nucleosides and Oligomeric Compounds Prepared Therefrom
EP2129402A2 (en) 2007-02-16 2009-12-09 KTB Tumorforschungsgesellschaft mbH Receptor and antigen targeted prodrug
EP2134173A4 (en) 2007-03-01 2010-11-10 Wellstat Ophthalmics Corp Treatment of diseases characterized by inflammation
CA2679244A1 (en) 2007-03-02 2009-03-05 Mdrna, Inc. Nucleic acid compounds for inhibiting myc gene expression and uses thereof
US8877917B2 (en) 2007-04-23 2014-11-04 Alnylam Pharmaceuticals, Inc. Glycoconjugates of RNA interference agents
AU2008260277C1 (en) 2007-05-30 2014-04-17 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
DK2173760T4 (en) 2007-06-08 2016-02-08 Isis Pharmaceuticals Inc Carbocyclic bicyclic nukleinsyreanaloge
WO2009003009A1 (en) 2007-06-26 2008-12-31 Enanta Pharmaceuticals, Inc. Substituted pyrrolidine as anti-infectives
ES2376507T5 (en) 2007-07-05 2015-08-31 Isis Pharmaceuticals, Inc. 6-disubstituted bicyclic nucleic acid analogs
EP2188298B1 (en) 2007-08-15 2013-09-18 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
BRPI0817527A2 (en) * 2007-10-01 2017-05-02 Isis Pharmaceuticals Inc antisense modulation of human fibroblast growth factor receptor expression 4
RU2010119775A (en) 2007-11-09 2011-12-27 Айсис Фармасьютикалс,Инк. (Us) MODULATION OF EXPRESSION OF FACTOR 7
WO2009067647A1 (en) 2007-11-21 2009-05-28 Isis Pharmaceuticals, Inc. Carbocyclic alpha-l-bicyclic nucleic acid analogs
CA2910760C (en) 2007-12-04 2019-07-09 Muthiah Manoharan Targeting lipids
SG10201402793PA (en) * 2007-12-27 2014-10-30 Stelic Inst Of Regenerative Medicine Stelic Inst & Co Sugar chain-related gene and use thereof
AU2009241591A1 (en) 2008-01-31 2009-11-05 Alnylam Pharmaceuticals, Inc. Optimized methods for delivery of DSRNA targeting the PCSK9 gene
WO2009100320A2 (en) 2008-02-07 2009-08-13 Isis Pharmaceuticals, Inc. Bicyclic cyclohexitol nucleic acid analogs
US20110130440A1 (en) 2008-03-26 2011-06-02 Alnylam Pharmaceuticals, Inc. Non-natural ribonucleotides, and methods of use thereof
CA2721183C (en) 2008-04-11 2019-07-16 Alnylam Pharmaceuticals, Inc. Site-specific delivery of nucleic acids by combining targeting ligands with endosomolytic components
WO2009143369A2 (en) 2008-05-22 2009-11-26 Isis Pharmaceuticals, Inc. Method of preparing nucleosides and analogs thereof without using chromatography
WO2009148605A2 (en) 2008-06-04 2009-12-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
EP2733222A1 (en) 2008-07-09 2014-05-21 Celera Corporation Genetic polymorphisms associated with cardiovascular diseases, methods of detection and uses thereof
WO2010017509A1 (en) 2008-08-07 2010-02-11 Isis Pharmaceuticals, Inc. Modulation of transthyretin expression for the treatment of cns related disorders
JP2012513953A (en) 2008-09-23 2012-06-21 アルニラム ファーマスーティカルズ インコーポレイテッド Chemical modification of monomers and oligonucleotides using cycloaddition
US8501805B2 (en) 2008-09-24 2013-08-06 Isis Pharmaceuticals, Inc. Substituted alpha-L-bicyclic nucleosides
EP2361256B1 (en) 2008-09-24 2013-04-10 Isis Pharmaceuticals, Inc. Cyclohexenyl nucleic acid analogs
AU2009305636A1 (en) 2008-10-15 2010-04-22 Ionis Pharmaceuticals, Inc. Modulation of Factor 11 expression
EA029762B1 (en) 2008-10-20 2018-05-31 Элнилэм Фармасьютикалз, Инк. Compositions and methods for inhibiting expression of transthyretin
AU2009308217B2 (en) 2008-10-24 2016-01-21 Ionis Pharmaceuticals, Inc. 5' and 2' bis-substituted nucleosides and oligomeric compounds prepared therefrom
EP2447274B1 (en) 2008-10-24 2017-10-04 Ionis Pharmaceuticals, Inc. Oligomeric compounds and methods
KR20220150995A (en) 2008-11-10 2022-11-11 알닐람 파마슈티칼스 인코포레이티드 Novel lipids and compositions for the delivery of therapeutics
US9023820B2 (en) 2009-01-26 2015-05-05 Protiva Biotherapeutics, Inc. Compositions and methods for silencing apolipoprotein C-III expression
CA2751342C (en) 2009-01-29 2019-05-07 Alnylam Pharmaceuticals, Inc. Lipid formulations comprising cationic lipid and a targeting lipid comprising n-acetyl galactosamine for delivery of nucleic acid
US8975389B2 (en) 2009-03-02 2015-03-10 Alnylam Pharmaceuticals, Inc. Nucleic acid chemical modifications
FR2943060B1 (en) 2009-03-13 2013-01-04 Commissariat Energie Atomique METAL ION CHELATING AGENTS, PROCESSES FOR THEIR PREPARATION AND THEIR APPLICATIONS
CN102458480A (en) 2009-04-15 2012-05-16 Isis制药公司 Modulation of inflammatory responses by factor xi
SG10201402054UA (en) 2009-05-05 2014-09-26 Muthiah Manoharan Lipid compositions
CN102575252B (en) 2009-06-01 2016-04-20 光环生物干扰疗法公司 For polynucleotide, the compoistion and method of use of multivalence RNA interference
CN104873464B (en) * 2009-06-10 2018-06-22 阿布特斯生物制药公司 Improved lipid formulations
CN104651408A (en) 2009-06-15 2015-05-27 阿尔尼拉姆医药品有限公司 Lipid formulated siRNA targeted to PCSK9 gene
BR112012000421A2 (en) 2009-07-06 2019-09-24 Alnylam Pharmaceuticals Inc compositions and methods for enhancing the production of a biological product.
US9512164B2 (en) 2009-07-07 2016-12-06 Alnylam Pharmaceuticals, Inc. Oligonucleotide end caps
US8927513B2 (en) 2009-07-07 2015-01-06 Alnylam Pharmaceuticals, Inc. 5′ phosphate mimics
WO2011008995A1 (en) 2009-07-16 2011-01-20 Isis Pharmaceuticals, Inc. Modulation of factor 7 expression
EP2462153B1 (en) 2009-08-06 2015-07-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
US8552163B2 (en) 2009-09-25 2013-10-08 Johns Hopkins University Liver-targeting agents and their synthesis
US8598334B2 (en) 2009-10-16 2013-12-03 Glaxo Group Limited HBV antisense inhibitors
TWI391144B (en) 2009-10-26 2013-04-01 Iner Aec Executive Yuan A quantification method for remaining liver function with a novel liver receptor imaging agent
TWI388338B (en) 2009-10-26 2013-03-11 Iner Aec Executive Yuan Method of radiolabelling multivalent glycoside for using as hepatic receptor imaging agent
US9101643B2 (en) * 2009-11-03 2015-08-11 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of transthyretin (TTR)
WO2011072290A2 (en) 2009-12-11 2011-06-16 The Regents Of The University Of Michigan Targeted dendrimer-drug conjugates
US8653047B2 (en) 2010-01-08 2014-02-18 Isis Pharmaceuticals, Inc. Modulation of angiopoietin-like 3 expression
US9198972B2 (en) 2010-01-28 2015-12-01 Alnylam Pharmaceuticals, Inc. Monomers and oligonucleotides comprising cycloaddition adduct(s)
EA024534B1 (en) * 2010-02-24 2016-09-30 Эрроухэд Рисерч Корпорейшн CONJUGATE DELIVERY SYSTEM AND COMPOSITION FOR TARGETED DELIVERY OF SMALL INTERFERING RNA (siRNA) AND PROCESS FOR PREPARING THE COMPOSITION
US9193752B2 (en) 2010-03-17 2015-11-24 Isis Pharmaceuticals, Inc. 5′-substituted bicyclic nucleosides and oligomeric compounds prepared therefrom
WO2011120053A1 (en) 2010-03-26 2011-09-29 Mersana Therapeutics, Inc. Modified polymers for delivery of polynucleotides, method of manufacture, and methods of use thereof
US20130109817A1 (en) 2010-03-26 2013-05-02 Mersana Therapeutics, Inc. Modified Polymers for Delivery of Polynucleotides, Method of Manufacture, and Methods of Use Thereof
WO2011123621A2 (en) 2010-04-01 2011-10-06 Alnylam Pharmaceuticals Inc. 2' and 5' modified monomers and oligonucleotides
US10913767B2 (en) 2010-04-22 2021-02-09 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
US9725479B2 (en) 2010-04-22 2017-08-08 Ionis Pharmaceuticals, Inc. 5′-end derivatives
CN103154014B (en) 2010-04-28 2015-03-25 Isis制药公司 Modified nucleosides, modified nucleosides-like and oligomeric compounds prepared therefrom
EP2625186B1 (en) 2010-04-28 2016-07-27 Ionis Pharmaceuticals, Inc. 5' modified nucleosides and oligomeric compounds prepared therefrom
ES2625689T3 (en) 2010-04-29 2017-07-20 Ionis Pharmaceuticals, Inc. Modulation of transthyretin expression
US20130236968A1 (en) 2010-06-21 2013-09-12 Alnylam Pharmaceuticals, Inc. Multifunctional copolymers for nucleic acid delivery
EP2616543A1 (en) * 2010-09-15 2013-07-24 Alnylam Pharmaceuticals, Inc. MODIFIED iRNA AGENTS
WO2012067970A2 (en) 2010-11-11 2012-05-24 Ted M Dawson Transcriptional repression leading to parkinson's disease
US8987377B2 (en) 2010-11-19 2015-03-24 Alnylam Pharmaceuticals, Inc. Poly(amide) polymers for the delivery of oligonucleotides
US8501930B2 (en) 2010-12-17 2013-08-06 Arrowhead Madison Inc. Peptide-based in vivo siRNA delivery system
SG189474A1 (en) 2010-12-17 2013-05-31 Arrowhead Res Corp Galactose cluster-pharmacokinetic modulator targeting moiety for sirna
WO2012089352A1 (en) 2010-12-29 2012-07-05 F. Hoffmann-La Roche Ag Small molecule conjugates for intracellular delivery of nucleic acids
US10017764B2 (en) 2011-02-08 2018-07-10 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
ES2709495T3 (en) 2011-04-01 2019-04-16 Ionis Pharmaceuticals Inc Modulation of signal transducer expression and transcription activator 3 (stat3)
CN105886506A (en) * 2011-04-13 2016-08-24 Isis制药公司 Antisense modulation of PTP1B expression
EP2699583A4 (en) 2011-04-21 2015-04-15 Isis Pharmaceuticals Inc Modulation of hepatitis b virus (hbv) expression
US20120295961A1 (en) 2011-04-21 2012-11-22 Swayze Eric E Modulation of hepatitis b virus (hbv) expression
US9157082B2 (en) 2011-04-27 2015-10-13 Isis Pharmaceuticals, Inc. Modulation of apolipoprotein CIII (ApoCIII) expression
WO2012174154A1 (en) 2011-06-13 2012-12-20 Isis Pharmaceuticals, Inc. Modulation of inflammatory responses by factor vii
CA2839437A1 (en) 2011-06-16 2012-12-20 Isis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
CN103890000B (en) 2011-06-21 2017-09-01 阿尔尼拉姆医药品有限公司 (ANGPTL3) the iRNA compositions of angiopoietin-like 3 and its application method
RU2631805C2 (en) * 2011-06-21 2017-09-26 Элнилэм Фармасьютикалз, Инк. Compositions and methods for apolipoprotein c-iii (apoc3) gene expression inhibition
WO2012177639A2 (en) 2011-06-22 2012-12-27 Alnylam Pharmaceuticals, Inc. Bioprocessing and bioproduction using avian cell lines
US20130017250A1 (en) * 2011-07-15 2013-01-17 The Trustees Of Columbia University In The City Of New York Methods For High Density Lipoprotein Cholesterol Regulation
KR20140051357A (en) 2011-08-26 2014-04-30 애로우헤드 리서치 코오포레이션 Poly(vinyl ester) polymers for in vivo nucleic acid delivery
EP3640332A1 (en) 2011-08-29 2020-04-22 Ionis Pharmaceuticals, Inc. Oligomer-conjugate complexes and their use
EP3401401B1 (en) 2011-09-20 2020-04-15 Ionis Pharmaceuticals, Inc. Antisense modulation of gcgr expression
RU2014119787A (en) 2011-10-25 2015-12-10 Айсис Фармасьютикалс, Инк. GCCR ANTI-SENSE REGULATION
KR102385013B1 (en) 2011-11-18 2022-04-12 알닐람 파마슈티칼스 인코포레이티드 RNAi AGENTS, COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING TRANSTHYRETIN (TTR) ASSOCIATED DISEASES
AU2013216852A1 (en) 2012-02-08 2014-08-21 Isis Pharmaceuticals, Inc. Methods and compositions for modulating Factor VII expression
US9340784B2 (en) 2012-03-19 2016-05-17 Ionis Pharmaceuticals, Inc. Methods and compositions for modulating alpha-1-antitrypsin expression
WO2013142571A2 (en) 2012-03-20 2013-09-26 Cornell University Assays for the identification of compounds that modulate lipid homeostasis
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
WO2013159108A2 (en) 2012-04-20 2013-10-24 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
WO2013159109A1 (en) 2012-04-20 2013-10-24 Isis Pharmaceuticals, Inc. Modulation of hepatitis b virus (hbv) expression
TWI595885B (en) 2012-05-02 2017-08-21 喜納製藥公司 Novel tetragalnac containing conjugates and methods for delivery of oligonucleotides
US20150299696A1 (en) 2012-05-02 2015-10-22 Sirna Therapeutics, Inc. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
US9574193B2 (en) 2012-05-17 2017-02-21 Ionis Pharmaceuticals, Inc. Methods and compositions for modulating apolipoprotein (a) expression
WO2013173789A2 (en) 2012-05-17 2013-11-21 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions
KR20170142997A (en) 2012-05-24 2017-12-28 아이오니스 파마수티컬즈, 인코포레이티드 Methods and compositions for modulating apolipoprotein (a) expression
US20150322428A1 (en) 2012-06-18 2015-11-12 Isis Pharmaceuticals, Inc. Compounds and methods for improved cellular uptake of antisense compounds
EP4253395A3 (en) 2012-08-06 2023-11-29 Alnylam Pharmaceuticals, Inc. Processes for the preparation of carbohydrate conjugated rna agents
EP2906699A4 (en) 2012-10-11 2016-06-08 Ionis Pharmaceuticals Inc Oligomeric compounds comprising bicyclic nucleosides and uses thereof
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
RU2649367C2 (en) 2013-01-30 2018-04-02 Ф. Хоффманн-Ля Рош Аг Lna oligonucleotide carbohydrate conjugates
EA031393B1 (en) 2013-05-01 2018-12-28 Ионис Фармасьютикалз, Инк. Compositions and methods for modulating hbv and ttr expression
CA2912345C (en) 2013-05-16 2020-10-06 Sumitomo Dainippon Pharma Co., Ltd. Transplantation adjuvant in cell therapy using neural progenitor cells
US20160122761A1 (en) 2013-06-21 2016-05-05 Isis Pharmaceuticals, Inc. Compositions and methods for modulation of target nucleic acids
EA201592215A1 (en) 2013-06-27 2016-05-31 Рош Инновейшен Сентер Копенгаген А/С ANTISMINAL OLIGOMERS AND THEIR CONJUGATES AIMED AT PROPROTEIN CONVERTASY SUBTILYSINE / KEXIN TYPE 9 (PCSK9)
WO2015002971A2 (en) 2013-07-02 2015-01-08 Isis Pharmaceuticals, Inc. Modulators of growth hormone receptor
DK3019200T3 (en) 2013-07-11 2022-06-20 Alnylam Pharmaceuticals Inc OLIGONUCLEOTIDE-LIGAND CONJUGATES AND METHOD OF PREPARATION
EP3043827B1 (en) 2013-09-13 2019-07-03 Ionis Pharmaceuticals, Inc. Modulators of complement factor b
US9943604B2 (en) 2013-09-20 2018-04-17 Ionis Pharmaceuticals, Inc. Targeted therapeutic nucleosides and their use
CA2928349A1 (en) 2013-11-14 2015-05-21 Roche Innovation Center Copenhagen A/S Apob antisense conjugate compounds
EP3077511A4 (en) 2013-12-06 2017-07-05 Dicerna Pharmaceuticals Inc. Methods and compositions for the specific inhibition of transthyretin (ttr) by double-stranded rna
ES2812099T3 (en) 2014-05-01 2021-03-16 Ionis Pharmaceuticals Inc Compositions and methods for modulating growth hormone receptor expression
CA2946003A1 (en) 2014-05-01 2015-11-05 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating angiopoietin-like 3 expression
RU2703411C2 (en) 2014-05-01 2019-10-16 Ионис Фармасьютикалз, Инк. Compositions and methods for modulating pkk expression
EP3811977A1 (en) 2014-05-01 2021-04-28 Ionis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
BR112016022593B1 (en) 2014-05-01 2022-04-26 Ionis Pharmaceuticals, Inc Oligomeric compounds, compositions comprising them, and uses thereof
WO2015179693A1 (en) 2014-05-22 2015-11-26 Isis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
US20170145424A1 (en) 2014-06-06 2017-05-25 Ionis Pharmaceuticals, Inc. Compositions and methods for enhanced intestinal absorption of conjugated oligomeric compounds
US20190046555A1 (en) * 2015-11-06 2019-02-14 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds for use in therapy
WO2017079739A1 (en) * 2015-11-06 2017-05-11 Ionis Pharmaceuticals, Inc. MODULATING APOLIPOPROTEIN (a) EXPRESSION

Also Published As

Publication number Publication date
EP3524680B1 (en) 2020-11-11
AU2017203436A1 (en) 2017-06-08
KR102235678B1 (en) 2021-04-05
BR112015027369B1 (en) 2021-06-08
AU2017203436B2 (en) 2018-10-18
EP2992009A4 (en) 2016-12-28
CN105378085B (en) 2019-02-15
EP2992009A1 (en) 2016-03-09
IL284593B2 (en) 2023-02-01
PH12018501963A1 (en) 2020-07-20
CL2015003217A1 (en) 2016-07-08
CN108064162B (en) 2021-12-03
BR112015027369A8 (en) 2018-01-02
HRP20201378T1 (en) 2020-11-27
BR112015027321A2 (en) 2017-09-26
AU2014259757B2 (en) 2017-03-02
AU2019202598A1 (en) 2019-05-02
KR20210151260A (en) 2021-12-13
US9181550B2 (en) 2015-11-10
AU2020233603A1 (en) 2020-10-01
US20200224198A1 (en) 2020-07-16
AU2014259750A1 (en) 2015-10-22
IL264580B (en) 2020-04-30
ZA201507218B (en) 2023-09-27
SG10201801507RA (en) 2018-03-28
CN105377887B (en) 2020-11-03
HRP20190987T1 (en) 2019-09-20
AU2020217347A1 (en) 2020-08-27
EA201891479A1 (en) 2018-11-30
CA2921518A1 (en) 2014-11-06
AU2014259750B2 (en) 2019-02-28
US20150176007A1 (en) 2015-06-25
KR102424855B1 (en) 2022-07-26
RU2015151200A3 (en) 2019-01-14
KR20180051678A (en) 2018-05-16
ME03390B (en) 2020-01-20
NZ712737A (en) 2021-08-27
PT2992009T (en) 2020-09-21
BR112015027319A2 (en) 2017-09-26
US9957504B2 (en) 2018-05-01
PL2992098T3 (en) 2019-09-30
RU2015151202A3 (en) 2018-03-27
RU2670614C9 (en) 2018-11-23
AU2019200820A1 (en) 2019-02-28
KR20210037752A (en) 2021-04-06
BR112015027322A8 (en) 2018-01-02
KR102138781B1 (en) 2020-07-28
US20220275365A9 (en) 2022-09-01
IL284000A (en) 2021-07-29
US9932580B2 (en) 2018-04-03
AU2022202770A1 (en) 2022-05-19
CN105378082A (en) 2016-03-02
SG10201801813YA (en) 2018-04-27
IL264241B (en) 2020-04-30
CN114058617A (en) 2022-02-18
RU2015151204A (en) 2017-06-02
AU2021204244A1 (en) 2021-07-22
AU2021204244B2 (en) 2023-10-19
US20210024923A1 (en) 2021-01-28
IL261901B (en) 2020-05-31
US20210087566A1 (en) 2021-03-25
IL273184B (en) 2021-07-29
AU2019200820B2 (en) 2020-04-30
JP2021020901A (en) 2021-02-18
EP2991656A2 (en) 2016-03-09
JP7177127B2 (en) 2022-11-22
JP2018183184A (en) 2018-11-22
MX2019010443A (en) 2019-10-17
JP2020074787A (en) 2020-05-21
WO2014179626A2 (en) 2014-11-06
US10883104B2 (en) 2021-01-05
CR20190269A (en) 2019-09-13
ZA201507216B (en) 2017-08-30
RU2019124314A (en) 2019-08-21
KR20220108195A (en) 2022-08-02
US20140343123A1 (en) 2014-11-20
AU2014259755B2 (en) 2018-08-30
AU2019204784A1 (en) 2019-07-25
BR112015027377A2 (en) 2017-08-29
MX2015015263A (en) 2016-12-16
SG10201906382QA (en) 2019-08-27
PH12015502493A1 (en) 2016-02-22
DOP2016000287A (en) 2017-02-15
EP3524680A1 (en) 2019-08-14
IL264241A (en) 2019-02-28
KR20190084138A (en) 2019-07-15
CN110066795A (en) 2019-07-30
US11299736B1 (en) 2022-04-12
ES2819213T3 (en) 2021-04-15
AU2014259759B2 (en) 2020-06-18
WO2014179625A1 (en) 2014-11-06
JP2024010070A (en) 2024-01-23
JP2020039355A (en) 2020-03-19
CN112921036A (en) 2021-06-08
EP2992098A4 (en) 2017-01-11
UA120287C2 (en) 2019-11-11
HK1221404A1 (en) 2017-06-02
HK1221403A1 (en) 2017-06-02
IL242125B (en) 2019-02-28
BR112015027319A8 (en) 2018-01-02
AU2014259759A1 (en) 2015-10-22
NZ728517A (en) 2021-12-24
LT2992009T (en) 2020-11-10
PT3524680T (en) 2021-01-04
DK2992009T3 (en) 2020-09-14
US9145558B2 (en) 2015-09-29
CN110079524A (en) 2019-08-02
EP2992097A2 (en) 2016-03-09
MX2020002184A (en) 2020-07-14
CN105378085A (en) 2016-03-02
IL296543A (en) 2022-11-01
MX2015015220A (en) 2016-01-12
MY198359A (en) 2023-08-28
DOP2015000268A (en) 2015-11-30
SI2992009T1 (en) 2020-10-30
MX2015015234A (en) 2016-10-03
US9163239B2 (en) 2015-10-20
IL270464B (en) 2021-07-29
KR20160002974A (en) 2016-01-08
WO2014179627A3 (en) 2015-04-16
JP6387084B2 (en) 2018-09-05
RS58981B1 (en) 2019-08-30
RU2015151199A (en) 2017-06-05
JP2023012548A (en) 2023-01-25
US20160090596A1 (en) 2016-03-31
HUE050394T2 (en) 2020-11-30
WO2014179629A2 (en) 2014-11-06
CA2921167A1 (en) 2014-11-06
EA031393B1 (en) 2018-12-28
KR20160003723A (en) 2016-01-11
AU2019204784C1 (en) 2022-11-03
EP3633039A1 (en) 2020-04-08
NZ631537A (en) 2017-05-26
JP2022017514A (en) 2022-01-25
JP2020007361A (en) 2020-01-16
KR20160002977A (en) 2016-01-08
CL2016002262A1 (en) 2017-06-09
EP2991661A1 (en) 2016-03-09
EP2992098A2 (en) 2016-03-09
IL263843B (en) 2020-03-31
WO2014179620A1 (en) 2014-11-06
IL261901A (en) 2018-10-31
JP6592486B2 (en) 2019-10-16
KR101857707B1 (en) 2018-05-14
EP2991661A4 (en) 2017-02-15
US20180002693A1 (en) 2018-01-04
US20190367914A1 (en) 2019-12-05
ES2778442T3 (en) 2020-08-10
KR20240042220A (en) 2024-04-01
EP2991661B1 (en) 2019-03-13
ES2730015T3 (en) 2019-11-07
CA2921162A1 (en) 2014-11-06
KR102482890B1 (en) 2022-12-30
JP7339294B2 (en) 2023-09-05
WO2014179629A3 (en) 2015-01-22
RU2015151203A (en) 2017-06-02
US10683499B2 (en) 2020-06-16
IL272617A (en) 2020-03-31
IL274064B (en) 2021-06-30
EP2991656B1 (en) 2019-12-18
MX2015015239A (en) 2016-10-03
JP7429103B2 (en) 2024-02-07
RU2650510C2 (en) 2018-04-16
US20230151365A1 (en) 2023-05-18
AU2014259755A1 (en) 2015-10-22
AU2019203674A1 (en) 2019-06-27
AU2024200296A1 (en) 2024-02-08
US20150126720A1 (en) 2015-05-07
KR20230113835A (en) 2023-08-01
JP2016522683A (en) 2016-08-04
EP3828275A1 (en) 2021-06-02
US9714421B2 (en) 2017-07-25
BR112015027377B1 (en) 2023-01-10
HK1221485A1 (en) 2017-06-02
CY1121879T1 (en) 2020-10-14
SG11201508870VA (en) 2015-11-27
MX2015015264A (en) 2016-08-12
PH12019501191A1 (en) 2021-03-01
JP6995478B2 (en) 2022-01-14
KR20160002975A (en) 2016-01-08
IL273184A (en) 2020-04-30
BR112015027321A8 (en) 2018-01-02
CA2921514C (en) 2023-10-24
CN105392488A (en) 2016-03-09
BR112015027377A8 (en) 2017-10-03
US20180273953A1 (en) 2018-09-27
IL274064A (en) 2020-06-30
PE20161430A1 (en) 2017-01-06
UA121017C2 (en) 2020-03-25
RU2018136140A (en) 2018-12-17
AU2020207820A1 (en) 2020-08-06
AU2014259756B2 (en) 2017-02-23
DK2992098T3 (en) 2019-06-17
US20180273952A1 (en) 2018-09-27
IL242126B (en) 2019-01-31
AU2017200950A1 (en) 2017-03-02
AU2018267625A1 (en) 2018-12-13
JP2021107408A (en) 2021-07-29
EP2991656A4 (en) 2017-02-22
LT2992098T (en) 2019-07-10
IL284593A (en) 2021-08-31
US10927372B2 (en) 2021-02-23
RU2015151202A (en) 2017-06-06
CY1123369T1 (en) 2021-12-31
US9127276B2 (en) 2015-09-08
KR102212275B1 (en) 2021-02-05
US10844379B2 (en) 2020-11-24
KR102315836B1 (en) 2021-10-22
NZ753018A (en) 2022-01-28
JP2021074021A (en) 2021-05-20
JP6456362B2 (en) 2019-01-23
US9181549B2 (en) 2015-11-10
MY178929A (en) 2020-10-23
IL242132B (en) 2018-10-31
NZ740338A (en) 2022-04-29
RU2019110030A (en) 2019-05-06
RS60796B1 (en) 2020-10-30
RU2699985C2 (en) 2019-09-11
US9932581B2 (en) 2018-04-03
RU2697152C2 (en) 2019-08-12
US20180044676A1 (en) 2018-02-15
AU2017200365B2 (en) 2018-11-08
WO2014179627A9 (en) 2015-02-26
US20160017323A1 (en) 2016-01-21
JP2023113843A (en) 2023-08-16
US11851655B2 (en) 2023-12-26
KR102558571B1 (en) 2023-07-21
ES2885174T3 (en) 2021-12-13
PH12015502493B1 (en) 2016-02-22
AU2017200365A1 (en) 2017-02-23
DK3524680T3 (en) 2020-12-14
BR112015027369A2 (en) 2017-09-26
RU2686080C2 (en) 2019-04-24
AU2018267625B2 (en) 2020-09-10
EA201592093A1 (en) 2016-06-30
US20210130823A1 (en) 2021-05-06
MX2021008899A (en) 2021-08-19
EP4155403A1 (en) 2023-03-29
RU2015151200A (en) 2019-01-14
RU2018136140A3 (en) 2022-04-27
JP6769866B2 (en) 2020-10-14
IL284593B (en) 2022-10-01
KR102651423B1 (en) 2024-03-27
JP6652602B2 (en) 2020-02-26
US20160076030A1 (en) 2016-03-17
DK2991656T3 (en) 2020-03-23
HK1221475A1 (en) 2017-06-02
JP2020039354A (en) 2020-03-19
JP6866459B2 (en) 2021-04-28
WO2014179629A8 (en) 2016-06-02
EA036584B1 (en) 2020-11-26
EP2992009B1 (en) 2020-06-24
JP6639629B2 (en) 2020-02-05
NZ631552A (en) 2017-02-24
PE20152002A1 (en) 2016-01-21
HUE043697T2 (en) 2019-09-30
WO2014179627A2 (en) 2014-11-06
RU2015151204A3 (en) 2018-03-27
CR20150612A (en) 2016-03-03
AU2017200950B2 (en) 2019-01-17
RU2015151199A3 (en) 2018-03-27
IL263843A (en) 2019-01-31
DOP2021000095A (en) 2021-09-15
MX2021008901A (en) 2021-08-19
US20210395734A1 (en) 2021-12-23
EP2992098B1 (en) 2019-03-27
US20150126718A1 (en) 2015-05-07
MX2019010441A (en) 2019-10-17
JP2016522817A (en) 2016-08-04
SI2992098T1 (en) 2019-06-28
KR20160002976A (en) 2016-01-08
CN113293163A (en) 2021-08-24
CA2921509A1 (en) 2014-11-06
EP3546579A1 (en) 2019-10-02
EP3690049A1 (en) 2020-08-05
CN105378082B (en) 2020-06-09
AU2014259756A1 (en) 2015-10-22
RU2670614C2 (en) 2018-10-24
US20190055554A1 (en) 2019-02-21
PL2992009T3 (en) 2020-11-30
RU2018112167A (en) 2019-03-07
AU2019203674B2 (en) 2021-03-25
JP2020058370A (en) 2020-04-16
NZ631512A (en) 2016-10-28
US20160076032A1 (en) 2016-03-17
JP2016526018A (en) 2016-09-01
JP2018027091A (en) 2018-02-22
IL273205A (en) 2020-04-30
AU2019204784B2 (en) 2022-01-27
AU2014259757A1 (en) 2015-10-22
MX2020004209A (en) 2020-08-13
AU2017200365C1 (en) 2019-04-18
CN105377887A (en) 2016-03-02
JP2016523515A (en) 2016-08-12
US20160090595A1 (en) 2016-03-31
PT2992098T (en) 2019-07-05
BR112015027322A2 (en) 2017-09-26
IL283660A (en) 2021-07-29
KR20230006933A (en) 2023-01-11
KR20200090966A (en) 2020-07-29
EP2992097A4 (en) 2017-01-04
CN110042098A (en) 2019-07-23
IL242124B (en) 2019-02-28
HK1221486A1 (en) 2017-06-02
JP2016526874A (en) 2016-09-08
EP2992097B1 (en) 2019-11-06
WO2014179626A3 (en) 2015-02-26
JP6216444B2 (en) 2017-10-18
CN108064162A (en) 2018-05-22
US20150126719A1 (en) 2015-05-07
KR20210129257A (en) 2021-10-27
JP2019056001A (en) 2019-04-11
CN105392488B (en) 2021-04-30
NZ725538A (en) 2021-02-26
KR20210014758A (en) 2021-02-09
CN110042098B (en) 2023-02-24
SG11201508800WA (en) 2015-11-27
IL264580A (en) 2019-02-28
IL273312A (en) 2020-04-30
CN111593051A (en) 2020-08-28

Similar Documents

Publication Publication Date Title
AU2017200365C1 (en) Compositions and methods for modulating apolipoprotein c-iii expression
CA2946003A1 (en) Compositions and methods for modulating angiopoietin-like 3 expression
BR122022020181B1 (en) OLIGOMERIC COMPOUNDS WITH CONJUGATE GROUPS, COMPOSITIONS COMPRISING SAID COMPOUNDS AND USES THEREOF
NZ725538B2 (en) Compositions and methods for modulating apolipoprotein (a) expression

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20200430

EEER Examination request

Effective date: 20200430

EEER Examination request

Effective date: 20200430

EEER Examination request

Effective date: 20200430

EEER Examination request

Effective date: 20200430

EEER Examination request

Effective date: 20200430

EEER Examination request

Effective date: 20200430