CA2921518A1 - Compositions and methods - Google Patents

Compositions and methods Download PDF

Info

Publication number
CA2921518A1
CA2921518A1 CA2921518A CA2921518A CA2921518A1 CA 2921518 A1 CA2921518 A1 CA 2921518A1 CA 2921518 A CA2921518 A CA 2921518A CA 2921518 A CA2921518 A CA 2921518A CA 2921518 A1 CA2921518 A1 CA 2921518A1
Authority
CA
Canada
Prior art keywords
compound
modified oligonucleotide
certain embodiments
conjugate
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2921518A
Other languages
French (fr)
Inventor
Thazha P. Prakash
Punit P. Seth
Eric E. Swayze
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ionis Pharmaceuticals Inc
Original Assignee
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=CA2921518(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 CA2921518A1 publication Critical patent/CA2921518A1/en
Abandoned legal-status Critical Current

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

Abstract

Provided herein are oligomeric compounds with conjugate groups. In certain embodiments, the oligomeric compounds are conjugated to N-Acetylgalactosamine.

Description

COMPOSITIONS AND METHODS
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 compounds efficacy.

SUBSTITUTE SHEET (RULE 26) 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 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
2 SUBSTITUTE SHEET (RULE 26) 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 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
3 SUBSTITUTE SHEET (RULE 26) 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 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,
4 SUBSTITUTE SHEET (RULE 26) 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 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:
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:
5 SUBSTITUTE SHEET (RULE 26) E¨F
A¨B¨C¨D
E¨F
where q = 3, the formula is:
E¨F
A¨B¨C¨D¨ E¨F
E¨F
where q = 4, the formula is:
E¨F
7 E¨F
A¨B¨C¨D
E¨F
E¨F
where q = 5, the formula is:
E¨F
¨B¨C¨D ____________________________ E
E¨F
¨F
A
E¨F
E¨F
In certain embodiments, conjugated antisense compounds are provided having the structure:
6 SUBSTITUTE SHEET (RULE 26) Targeling moiety r ASO
HO OH
¨
0=H -OH xr OH
C/,0,7; IN:!"
HO-VCIFIN \-/IF1-'¨( =

- NHAc HO 7 1 I ' _ o___ _ 0 N
b HO_..7.C....\_---C) ,,-----.õ--11----\.711y---- - ------- \O __ H=0 OH

NHAc g 0 crz Linker Cleavable moiety Ligand Tether OH
HO\&70...\ z HN ----, '0 Branching group HO _________________________ 0 NHAc =
In certain embodiments, conjugated antisense compounds are provided having the structure:
Cell targeting moiety HOOH
0 , 0 HO---4\ruNN_____...\ AL
Cleavable moiety AcHN 0 1 0 -, -OH
- -HOOH _ _ , _____ , ,N,21-12 \\I\T
.......Zy-\ , ,,..._,,,,. 11 HO___ `-' -P, 0 1 0 0 _ _ P OvorN
O-_ AcHN __ OH C) 0 Tether _____________________________________________ 1 -0¨P=0 Ligand HO OH 9 y A _ _ ASO

HO OH
NHAc Branching group =
7 SUBSTITUTE SHEET (RULE 26) In certain embodiments, conjugated antisense compounds are provided having the structure:
ASO Cleavable moiety HO¨P.0 0¨cOr N
HO¨P=0 Cell targeting moiety ________________________________________________________________ 0 ________________ (<3 voN
AcHN 0- 0 (()3 HOOH _ ______ Conjugate 0 0 , 0 linker HO `-'\/\V\/N =0 AcHN _ cr. - OH
Tether Ligand HO OH
P, 0-6_ H' N Branching group In certain embodiments, conjugated antisense compounds are provided having the structure:
8 SUBSTITUTE SHEET (RULE 26)
9 ASO
Ligand Tether Cleavable moiety H010 HO OH
Nyio 0 AcHN _ _ 0 (46 NH
HO OH 0HO0N1) __________________________________________________________ 3 4 YHN3( _______________________________________ \O
AcHN 0 Conjugate HO OH
linker 4 1rNN31 AcHN 0 Branching group Cell targeting moiety The present disclosure provides the following non-limiting numbered embodiments:
Embodiment 1. The conjugated antisense compound of any of embodiments 1179 to 1182, wherein the tether has a structure selected from among:

Ylr NC
rssr, or rssf ; wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
Embodiment 2. The conjugated antisense compound of any of embodiments 1179 to 1182, wherein the tether has the structure:

'4 H
isss SUBSTITUTE SHEET (RULE 26) Embodiment 3.
The conjugated antisense compound of any of embodiments 1179 to 1182 or 1688 to 1689, wherein the linker has a structure selected from among:

k)COL
N c5Sc/H \ N^H"'L

2 H "5 OH

0 and 0 5 Embodiment 4. The conjugated antisense compound of any of embodiments 1179 to 1182 or 1688 to 1689, wherein the linker has a structure selected from among:

k)COL
S\HJ-L
N'JI N("r n OH
0 and 0 =
wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
Embodiment 5.
The conjugated antisense compound of any of embodiments 1179 to 1182 or 1688 to 1689, wherein the linker has the structure:

µ)LNO-1 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.
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 SUBSTITUTE SHEET (RULE 26) 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, "2'-F nucleoside" refers to a nucleoside comprising a sugar comprising fluorine at the 2' position. Unless otherwise indicated, the fluorine in a 2'-F nucleoside is in the ribo position (replacing the OH of a natural ribose).
"2'-0-methoxyethyl" (also 2'-MOE and 2'-0(CH2)2-0CH3) refers to an 0-methoxy-ethyl modification of the 2' position of a furosyl ring. A 2'-0-methoxyethyl modified sugar is a modified sugar.
"2'-0-methoxyethyl nucleotide" means a nucleotide comprising a 2'-0-methoxyethyl modified sugar moiety.
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.
"3' target site" refers to the nucleotide of a target nucleic acid which is complementary to the 3'-most nucleotide of a particular antisense compound.
"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.
"5-methylcytosine" means a cytosine modified with a methyl group attached to the 5' position. A 5-methylcytosine is a modified nucleobase.
"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%.

SUBSTITUTE SHEET (RULE 26) "Active pharmaceutical agent" means the substance or substances in a pharmaceutical composition that provide a therapeutic benefit when administered to an individual. For example, in certain embodiments an antisense oligonucleotide targeted to PTP1B is an active pharmaceutical agent.
"Active target region" or "target region" means a region to which one or more active antisense compounds is targeted. "Active antisense compounds" means antisense compounds that reduce target nucleic acid levels or protein levels.
"Adipogenesis" means the development of fat cells from preadipocytes.
"Lipogenesis" means the production or formation of fat, either fatty degeneration or fatty infiltration.
"Adiposity" or "Obesity" refers to the state of being obese or an excessively high amount of body fat or adipose tissue in relation to lean body mass. The amount of body fat includes concern for both the distribution of fat throughout the body and the size and mass of the adipose tissue deposits. Body fat distribution can be estimated by skin-fold measures, waist-to-hip circumference ratios, or techniques such as ultrasound, computed tomography, or magnetic resonance imaging. According to the Center for Disease Control and Prevention, individuals with a body mass index (BMI) of 30 or more are considered obese. The term "Obesity" as used herein includes conditions where there is an increase in body fat beyond the physical requirement as a result of excess accumulation of adipose tissue in the body.
The term "obesity" includes, but is not limited to, the following conditions: adult-onset obesity; alimentary obesity; endogenous or inflammatory obesity; endocrine obesity; familial obesity; hyperinsulinar obesity; hyperplastic-hypertrophic obesity; hypogonadal obesity; hypothyroid obesity; lifelong obesity; morbid obesity and exogenous obesity.
"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.
"Administering" means providing an agent to an animal, and includes, but is not limited to, administering by a medical professional and self-administering.
"Agent" means an active substance that can provide a therapeutic benefit when administered to an animal. "First Agent" means a therapeutic compound provided herein. For example, a first agent can be an antisense oligonucleotide targeting PTP1B. "Second agent" means a second therapeutic compound of the invention (e.g. a second antisense oligonucleotide targeting PTP1B) and/or a non- PTP1B therapeutic compound.
"Amelioration" 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.

SUBSTITUTE SHEET (RULE 26) "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.
"Antisense activity" means any detectable or measurable activity attributable to the hybridization of an antisense compound to its target nucleic acid. In certain embodiments, antisense activity is a decrease in the amount or expression of a target nucleic acid or protein encoded by such target nucleic acid.
"Antisense compound" means an oligomeric compound that is capable of undergoing hybridization to a target nucleic acid through hydrogen bonding.
"Antisense inhibition" means reduction of target nucleic acid levels or target protein levels in the presence of an antisense compound complementary to a target nucleic acid compared to target nucleic acid levels or target protein levels in the absence of the antisense compound.
"Antisense oligonucleotide" means a single-stranded oligonucleotide having a nucleobase sequence that permits hybridization to a corresponding region or segment of a target nucleic acid.
"Bicyclic sugar" means a furosyl ring modified by the bridging of two non-geminal ring atoms. A
bicyclic sugar is a modified sugar.
"Bicyclic nucleic acid" or "BNA" refers to a nucleoside or nucleotide wherein the furanose portion of the nucleoside or nucleotide includes a bridge connecting two carbon atoms on the furanose ring, thereby forming a bicyclic ring system.
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.
"Cap structure" or "terminal cap moiety" means chemical modifications, which have been incorporated at either terminus of an antisense compound.
"Chemically distinct region" refers to a region of an antisense compound that is in some way chemically different than another region of the same antisense compound. For example, a region having 2'-0-methoxyethyl nucleotides is chemically distinct from a region having nucleotides without 2'-0-methoxyethyl modifications.
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.

SUBSTITUTE SHEET (RULE 26) "Chimeric antisense compound" means an antisense compound that has at least two chemically distinct regions.
"Co-administration" means administration of two or more agents to an individual. The two or more agents can be in a single pharmaceutical composition, or can be in separate pharmaceutical compositions.
Each of the two or more agents can be administered through the same or different routes of administration.
Co-administration encompasses parallel or sequential administration.
"Cholesterol" is a sterol molecule found in the cell membranes of all animal tissues. Cholesterol must be transported in an animal's blood plasma by lipoproteins including very low density lipoprotein (VLDL), intermediate density lipoprotein (IDL), low density lipoprotein (LDL), and high density lipoprotein (HDL). "Plasma cholesterol" refers to the sum of all lipoproteins (VDL, IDL, LDL, HDL) esterified and/or non-esterified cholesterol present in the plasma or serum.
"Cholesterol absorption inhibitor" means an agent that inhibits the absorption of exogenous cholesterol obtained from diet.
"Complementarity" means the capacity for pairing between nucleobases of a first nucleic acid and a second nucleic acid.
As used herein, "constrained ethyl nucleoside" or "cEt" means a nucleoside comprising a bicyclic sugar moiety comprising a 4'-CH(CH3)-0-2'bridge.
"Contiguous nucleobases" means nucleobases immediately adjacent to each other.

"Deoxyribonucleotide" means a nucleotide having a hydrogen at the 2' position of the sugar portion of the nucleotide. Deoxyribonucleotides may be modified with any of a variety of substituents.
"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.
"Diabetic dyslipidemia" or "type 2 diabetes with dyslipidemia" means a condition characterized by Type 2 diabetes, reduced HDL-C, elevated triglycerides, and elevated small, dense LDL particles.
"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.

SUBSTITUTE SHEET (RULE 26) "Dyslipidemia" refers to a disorder of lipid and/or lipoprotein metabolism, including lipid and/or lipoprotein overproduction or deficiency. Dyslipidemias may be manifested by elevation of lipids such as cholesterol and triglycerides as well as lipoproteins such as low-density lipoprotein (LDL) cholesterol.
"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 antisense oligonucleotide.
"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.
"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.
"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 target nucleic acid is a second nucleic acid.
As used herein, "furanosyl" means a structure comprising a 5-membered ring comprising four carbon atoms and one oxygen atom.
"Gapmer" means a chimeric antisense compound in which an internal region having a plurality of nucleosides that support RNase H cleavage is positioned between external regions having one or more nucleosides, wherein the nucleosides comprising the internal region are chemically distinct from the nucleoside or nucleosides comprising the external regions. The internal region can be referred to as a "gap segment" and the external regions can be referred to as "wing segments."
"Gap-widened" means a chimeric antisense compound having a gap segment of 12 or more contiguous 2'-deoxyribonucleosides positioned between and immediately adjacent to 5' and 3' wing segments having from one to six nucleosides.
"Glucose" is a monosaccharide used by cells as a source of energy and inflammatory intermediate.
"Plasma glucose" refers to glucose present in the plasma.
SUBSTITUTE SHEET (RULE 26) "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.
"Hybridization" means the annealing of complementary nucleic acid molecules.
In certain embodiments, complementary nucleic acid molecules include an antisense compound and a target nucleic acid.
"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 and triglycerides.
"Hypertriglyceridemia" means a condition characterized by elevated triglyceride levels.
"Identifying" or "selecting an animal with metabolic" means identifying or selecting a subject having been diagnosed with a metabolic disease, or a metabolic disorder; or, identifying or selecting a subject having any symptom of a metabolic disease, including, but not limited to, metabolic syndrome, hyperglycemia, hypertriglyceridemia, hypertension increased insulin resistance, decreased insulin sensitivity, above normal body weight, and/or above normal body fat or any combination thereof. Such identification may be accomplished by any method, including but not limited to, standard clinical tests or assessments, such as measuring serum or circulating (plasma) blood-glucose, measuring serum or circulating (plasma) triglycerides, measuring blood-pressure, measuring body fat, measuring body weight, and the like.
"Immediately adjacent" means there are no intervening elements between the immediately adjacent elements.
"Individual" or "subject" or "animal" means a human or non-human animal selected for treatment or therapy.
"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.
"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.
"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.

SUBSTITUTE SHEET (RULE 26) "Internucleoside linkage" refers to the chemical bond between nucleosides.
"Intravenous administration" means administration into a vein.
"Linked nucleosides" means adjacent nucleosides which are bonded together.
As used herein, "locked nucleic acid nucleoside" or "LNA" means a nucleoside comprising a bicyclic sugar moiety comprising a 4'-CH2-0-2'bridge.
"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 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 statins, fibrates, and MTP inhibitors.
"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, low HDL-C, family history of coronary heart disease, age, and other factors disclosed herein.
"Metabolic disease" or "metabolic disorder" 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 diseases or disorders include, but are not limited to, obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof.
"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).
"Mismatch" or "non-complementary nucleobase" refers to the case when a nucleobase of a first nucleic acid is not capable of pairing with the corresponding nucleobase of a second or target nucleic acid.
"Mixed dyslipidemia" means a condition characterized by elevated cholesterol and elevated triglycerides.
"Modified internucleoside linkage" refers to a substitution or any change from a naturally occurring internucleoside bond (i.e. a phosphodiester internucleoside bond).

SUBSTITUTE SHEET (RULE 26) "Modified nucleobase" refers to any nucleobase other than adenine, cytosine, guanine, thymidine, or uracil. An "unmodified nucleobase" means the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C), and uracil (U).
"Modified nucleoside" means a nucleoside having, independently, a modified sugar moiety or modified nucleobase.
"Modified nucleotide" means a nucleotide having, independently, a modified sugar moiety, modified internucleoside linkage, or modified nucleobase. A "modified nucleoside" means a nucleoside having, independently, a modified sugar moiety or modified nucleobase.
"Modified oligonucleotide" means an oligonucleotide comprising at least one modified nucleotide.
"Modified sugar" refers to a substitution or change from a natural sugar.
As used herein, "modified sugar moiety" means a substituted sugar moiety or a sugar surrogate.
As used herein, "MOE" means -OCH2CH2OCH3.
"Motif' means the pattern of chemically distinct regions in an antisense compound.
"MTP inhibitor" means an agent inhibits the enzyme, microsomal triglyceride transfer protein.
"Naturally occurring internucleoside linkage" means a 3' to 5' phosphodiester linkage.
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.
"Natural sugar moiety" means a sugar found in DNA (2'-H) or RNA (2'-OH).
"Non-alcoholic fatty liver disease" or "NAFLD" means a condition characterized by fatty inflammation of the liver that is not due to excessive alcohol use (for example, alcohol consumption of over 20 g/day). In certain embodiments, NAFLD is related to insulin resistance and the metabolic syndrome.
NAFLD encompasses a disease spectrum ranging from simple triglyceride accumulation in hepatocytes (hepatic steatosis) to hepatic steatosis with inflammation (steatohepatitis), fibrosis, and cirrhosis.
"Nonalcoholic steatohepatitis" (NASH) occurs from progression of NAFLD beyond deposition of triglycerides. A "second hit" capable of inducing necrosis, inflammation, and fibrosis is required for development of NASH. Candidates for the second-hit can be grouped into broad categories: factors causing an increase in oxidative stress and factors promoting expression of proinflammatory cytokines "Nucleic acid" refers to molecules composed of monomeric nucleotides. A
nucleic acid includes ribonucleic acids (RNA), deoxyribonucleic acids (DNA), single-stranded nucleic acids, double-stranded nucleic acids, small interfering ribonucleic acids (siRNA), and microRNAs (miRNA). A nucleic acid can also comprise a combination of these elements in a single molecule.
"Nucleobase" means a heterocyclic moiety capable of pairing with a base of another nucleic acid. 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 SUBSTITUTE SHEET (RULE 26) with a complementary naturally occurring nucleobase of another oligonucleotide or nucleic acid.
Nucleobases may be naturally occurring or may be modified.
"Nucleobase sequence" means the order of contiguous nucleobases independent of any sugar, linkage, or nucleobase modification.
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.
"Nucleoside mimetic" includes those structures used to replace the sugar or the sugar and the base and not necessarily the linkage at one or more positions of an oligomeric compound such as for example nucleoside mimetics having morpholino, cyclohexenyl, cyclohexyl, tetrahydropyranyl, bicyclo or tricyclo sugar mimetics e.g. non furanose sugar units.
"Nucleotide" means a nucleoside having a phosphate group covalently linked to the sugar portion of the nucleoside. 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).
"Nucleotide mimetic" includes those structures used to replace the nucleoside and the linkage at one or more positions of an oligomeric compound such as for example peptide nucleic acids or morpholinos (morpholinos linked by -N(H)-C(=0)-0- or other non-phosphodiester linkage).
"Oligomeric compound" or "oligomer" refers to a polymeric structure comprising two or more sub-structures and capable of hybridizing to a region of a nucleic acid molecule.
In certain embodiments, oligomeric compounds are oligonucleosides. In certain embodiments, oligomeric compounds are oligonucleotides. In certain embodiments, oligomeric compounds are antisense compounds. In certain embodiments, oligomeric compounds are antisense oligonucleotides. In certain embodiments, oligomeric compounds are chimeric oligonucleotides.
"Oligonucleotide" means a polymer of linked nucleosides each of which can be modified or unmodified, independent one from another.
"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, or chronic, or short or intermittent.
"Peptide" means a molecule formed by linking at least two amino acids by amide bonds. Peptide refers to polypeptides and proteins.

SUBSTITUTE SHEET (RULE 26) "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 PTP1B is pharmaceutical agent.
"Pharmaceutical 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 sterile aqueous solution.
"Pharmaceutically acceptable carrier" means a medium or diluent that does not interfere with the structure of the oligonucleotide. Certain, of such carries enable pharmaceutical compositions to be formulated as, for example, tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspension and lozenges for the oral ingestion by a subject. For example, a pharmaceutically acceptable carrier can be a sterile aqueous solution.
"Pharmaceutically acceptable derivative" encompasses pharmaceutically acceptable salts, conjugates, prodrugs or isomers of the compounds described 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 oligonucleotide and do not impart undesired toxicological effects thereto.
"Phosphorothioate linkage" means a linkage between nucleosides where the phosphodiester bond is modified by replacing one of the non-bridging oxygen atoms with a sulfur atom.
A phosphorothioate linkage is a modified internucleoside linkage.
"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.
"Prevent" 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.
"Prodrug" means a therapeutic agent that is prepared in an inactive form that is converted to an active form within the body or cells thereof by the action of endogenous enzymes or other chemicals or conditions.
"Protein tyrosine phosphatase 1B" or "PTP1B" (also known as PTPN1; protein tyrosine phosphatase, non-receptor type 1; PTP-1B; RKPTP) means any nucleic acid or protein of PTP1B.
"PTP1B expression" means the level of mRNA transcribed from the gene encoding PTP1B or the level of protein translated from the mRNA. PTP1B expression can be determined by art known methods such as a Northern or Western blot.
"PTP1B nucleic acid" means any nucleic acid encoding PTP1B. For example, in certain embodiments, a PTP1B nucleic acid includes a DNA sequence encoding PTP1B, a RNA sequence SUBSTITUTE SHEET (RULE 26) transcribed from DNA encoding PTP1B (including genomic DNA comprising introns and exons), and a mRNA sequence encoding PTP1B. "PTP1B mRNA" means a mRNA encoding a PTP1B
protein.
"Side effects" means physiological responses attributable to a treatment other than the desired effects.
In certain embodiments, side effects include injection site reactions, liver function test abnormalities, renal function abnormalities, liver toxicity, renal toxicity, central nervous system abnormalities, myopathies, and malaise. For example, increased aminotransferase levels in serum can indicate liver toxicity or liver function abnormality. For example, increased bilirubin can indicate liver toxicity or liver function abnormality.
"Single-stranded oligonucleotide" means an oligonucleotide which is not hybridized to a complementary strand.
"Specifically hybridizable" refers to an antisense compound having a sufficient degree of complementarity between an antisense oligonucleotide and a target nucleic acid to induce a desired effect, while exhibiting minimal or no effects on non-target nucleic acids under conditions in which specific binding is desired, i.e. under physiological conditions in the case of in vivo assays and therapeutic treatments.
"Statin" means an agent that inhibits the activity of HMG-CoA reductase.
"Subcutaneous administration" means administration just below the skin.
As used herein, "sugar moiety" means a naturally occurring sugar moiety or a modified sugar moiety of a nucleoside.
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 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.

SUBSTITUTE SHEET (RULE 26) "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.
"Target nucleic acid," "target RNA," and "target RNA transcript" all refer to a nucleic acid capable of being targeted by antisense compounds.
"Target segment" means the sequence of nucleotides of a target nucleic acid to which an antisense compound 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.
"Therapeutically effective amount" means an amount of an agent that provides a therapeutic benefit to an individual.
"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.
"Triglyceride" or "TG" means a lipid or neutral fat consisting of glycerol combined with three fatty acid molecules.
"Type 2 diabetes," (also known as "type 2 diabetes mellitus" or "diabetes mellitus, type 2", and formerly called "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.
"Treat" refers to administering a pharmaceutical composition to an animal to effect an alteration or improvement of a disease, disorder, or condition.
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).
"Unmodified nucleotide" means a nucleotide composed of naturally occurring nucleobases, sugar moieties, and internucleoside linkages. In certain embodiments, an unmodified nucleotide is an RNA
nucleotide (i.e. [3-D-ribonuc1eosides) or a DNA nucleotide (i.e. [3-D-deoxyribonuc1eoside).

SUBSTITUTE SHEET (RULE 26)
10 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:

SUBSTITUTE SHEET (RULE 26) vvv Ra Rb=P¨R, Rd vw wherein:
Ra and Rd are each, independently, 0, S, CH2, NH, or N.I1 wherein J1 is C1-C6 alkyl or substituted C1-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-internuoleoside 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-SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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," 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 SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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 affmity 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.

SUBSTITUTE SHEET (RULE 26) 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, SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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)R00), carboxyl (-C(0)0-R00), aliphatic groups, alicyclic groups, alkoxy, substituted oxy (-0-R00), aryl, aralkyl, heterocyclic radical, heteroaryl, heteroarylalkyl, amino (-N(Rbb)(Rõ)), imino(=NRbb), amido (-C(0)N(Rbb)(Rõ) or -N(Rbb)C(0)Raa), azido (-N3), nitro (-NO2), cyano (-CN), carbamido (-0C(0)N(Rbb)(Rõ) or -N(Rbb)C(0)01Z00), ureido (-N(Rbb)C(0)N(Rbb)(Rõ)), thioureido (-N(Rbb)C(S)N(Rbb)-(Rec)), guanidinyl (-N(Rbb)C(=NRbb)N(Rbb)(Rõ)), amidinyl (-C(=NRbb)N(Rbb)(Rce) 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-Ci2a1kyl) with from 1 to about 6 carbon atoms being more preferred.

SUBSTITUTE SHEET (RULE 26) 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.

SUBSTITUTE SHEET (RULE 26) As used herein, "aminoalkyl" means an amino substituted C1-C12alkyl 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 CI-Cu 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.

SUBSTITUTE SHEET (RULE 26) 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-methy1"), 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)2SCH3, 0(CH2)2-0-N(Rm)(Rn), 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-SUBSTITUTE SHEET (RULE 26) 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(R,,)-alkyl; 0, S, or N(Rin)-alkenyl; 0, S or N(R,,)-alkynyl; 0-alkyleny1-0-alkyl, alkynyl, alkaryl, aralkyl, 0-alkaryl, 0-aralkyl, 0(CH2)2SCH3, 0-(CH2)2-0-N(R.)(RJ or 0-CH2-C(=0)-N(Rin)(Rn), where each Rn, 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, O-CH3, 0(CH2)3NH2, CH2-CH=CH2, O-CH2-CH=CH2, OCH2CH2OCH3, 0(CH2)2SCH3, 0-(CH2)2-0-N(Rm)(Rn), 0(CH2)20(CH2)2N(CH3)2, and N-substituted acetamide (0-CH2-C(=0)-N(Rin)(Rii) where each Rin 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)2SCH3, 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(Ra)(Rb)]n-, -[C(Ra)(Rb)b-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).
SUBSTITUTE SHEET (RULE 26) In certain embodiments, such 4' to 2' bridges independently comprise from 1 to 4 linked groups independently selected from -[C(Ra)(Rb)]ii-, -C(Ra)=C(Rb)-, -C(Ra)=N-, -C(=NRa)-, -C(=0)-, -C(=S)-, -0-, -Si(Ra)2-, -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, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C5-C20 aryl, substituted C5-C20 aryl, heterocycle radical, substituted heterocycle radical, heteroaryl, substituted heteroaryl, C5-C7 alicyclic radical, substituted C5-C7 alicyclic radical, halogen, OJI, NJ1J2, SJI, N3, COOJt, acyl (C(=0)-H), substituted acyl, CN, sulfonyl (S(=0)2-J1), or sulfoxyl (S(=0)-J1); 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) [3-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 coN Bx Bx 0 , /
(A) (B) (C) _____________ zO,Bx 5 3¨ OyBx Bx N-W ¨0 H3C
\
(D) (E) (F) SUBSTITUTE SHEET (RULE 26) \ ,0 Bx \ 0 Bx 0 Bx ______________________________ 7/
(H) /-N

(G) CH3 (I) / co?/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., Cum Opinion MoL 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. US2004/0171570, US2007/0287831, and US2008/0039618; 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/US2008/064591, PCT/US2008/066154, 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 [3-D
configuration. Previously, a-L-methyleneoxy (4'-CH2-0-2') bicyclic nucleosides have been incorporated into antisense oligonucleotides that showed antis ens e 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 US2005/0130923, published on June 16, 2005) and/or the 5' position. By way of additional example, carbocyclic bicyclic nucleosides having SUBSTITUTE SHEET (RULE 26) a 4'-2' bridge have been described (see, e.g., Freier et al., Nucleic Acids Research, 1997, 25(22), 4429-4443 and Albaek et al., J. Org. Chem., 2006, 71, 7731-7740).
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., Biochemisay, 2002, 4/, 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:
q2 0 /q3 CI7 _______________________ CI4 q67y\¨Bx /

VI
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, CP/ q5, q6 and q7 are each, independently, H, C1-C6 alkyl, substituted C1-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl, or substituted C2-C6 alkynyl; and SUBSTITUTE SHEET (RULE 26) each of RI and R2 is independently selected from among: hydrogen, halogen, substituted or unsubstituted alkoxy, NJ1J2, S71, N3, OC(=X),11, OC(=X)NJ172, NJ3C(=X)NJ1J2, and CN, wherein X is 0, S or NJI, and each 71, ,12, and 73 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 qt, q2, C13, C14, C15, q6 and q7 is other than H. In certain embodiments, at least one of qi, q2, q3, q4, q5, q6and q7 is methyl. In certain embodiments, THP
nucleosides of Formula VI are provided wherein one of RI and R2 is F. In certain embodiments, RI is fluoro and R2 is H, RI is methoxy and R2 is H, and RI 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 SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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 r3 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 occuning 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 SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) a gapmer consists of 6 to 12 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 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 5 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 10 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 SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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 Len2ths 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, 10 to 28, 10 to 29, 10 to 30, 11 to 12, 11 to 13, 11 to 14, 11 to 15, 11 to 16, 11 to 17, 11 to 18, 11 to 19, 11 to 20, 11 to 21, 11 to 22, 11 to 23, 11 to 24, 11 to 25, 11 to 26, 11 to 27, 11 to 28, 11 to 29, 11 to 30, 12 to 13, 12 to 14, 12 to 15, 12 to 16, 12 to 17, 12 to 18, 12 to 19, 12 to 20, 12 to 21, 12 to 22, 12 to 23, 12 to 24, 12 to 25, 12 to 26, 12 to 27, 12 to 28, 12 to 29, 12 to 30, 13 to 14, 13 to 15, 13 to 16, 13 to 17, 13 to 18, 13 to 19, 13 to 20, 13 to 21, 13 to 22, 13 to 23, 13 to 24, 13 to 25, 13 to 26, 13 to 27, 13 to 28, 13 to 29, 13 to 30, 14 to 15, 14 to 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 SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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-SUBSTITUTE SHEET (RULE 26) 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:

SUBSTITUTE SHEET (RULE 26) 0=P-OH
OBXi 01-0H 0=P-OH
oi yON(Bxi vOyx2 (5\
0=P-OH
oI 04'-OH 04'-OH
oI oI
VO\,13x (C)),Eix2 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:
0=P-OH NH2 oi 0 µN I
N
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 SUBSTITUTE SHEET (RULE 26) 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., SUBSTITUTE SHEET (RULE 26) 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:
NI, --. -EN
0, -NH

0 )0)2. 11 )0-P-OH
N I

H
, ( )n So wv I I 0 , N I

X 0, , r1-0H
, II

' I O
\ N OH =
'LLL)LHO
P I
I I
0 O_ D\ n* 0 õ c=%.,ON, ^4-0'61- r__( /-(- \ OH CN),. 0,1 n , H Hm1,1H,t,,)1 0 ' s" S4 iL
is N S' Mn '0 wv 0,, HHHH H II 0=Nµ,. 0 NN.prT.,,ON, H
n µ in "n in I=
0 ; ,,,,,Ns,S,Hro , /
I

I
0 0 \ 1,.. 0 0 0 0 1 NO ''. 4-01T' OH
...,,,, \ 0 OH 1)1-1\ /¨Pn( 0 CS
S-S n 0 D'Il-C)1 N ; and H
EN,"Hr;rN'9 \.,,N,Hro wherein each n is, independently, from 1 to 20; and p is from 1 to 6.
SUBSTITUTE SHEET (RULE 26) In certain embodiments, a linker has a structure selected from among:
rt-'4 \
154\ 0 (:).
N,,o'µ

, rr\rj 0..

N
0 \
H H Q.
H '\()r.'N 0 ;
n /µ

,L H
\.)N rilN,=11.r\ n NH ,fr.
,-r-rj 0 \ 0 0.
N)0)41 pr-rj \

A
csy,,,,s,sX J(.1,,N.J.,)iN7Ni . 0 NO

H
IyqNN (-C
nOr N
' n H

\

\

prr' CiNI) ir H A
/1\1 \ C)(-1-NA
0 ,r,, \ n H
s: o 0, .
N I
O-P = 0 N 0 I
O-P =0 H OH ;

OH
r."0 fr0 ; and O
o H , SN¨

c"1\1 N
l In HO
wherein each n is, independently, from 1 to 20.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a linker has a structure selected from among:

H
N N

OH
0 H 0 0 HN,,..0 0ck, \,S, j'A-__ s µL(f N csss ; ,-rii s , _-5 H n 7 \
n cssN N Cli ; 0 no H 0 n H
csc N ,1 N A . oLH.,., N
"n ' H ' \ , µ / n =
0 n H
0 0 ' H

n H
H H
Fy.H.N ._cs ; F.)N,,,,r,0Q0/1N csss ; and H H
n wherein n is from 1 to 20.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a linker has a structure selected from among:

,, N .
n n H
, n n H , wv is OH

.
H csss ; , in n , Un "n sIN N -,,,, ; 0 0 H n Is H
, is H n 0 n H , n e FI\11=(.. /I\Iss cir\LHOCOIn "1 ; "5-(0-f-11,;
0 0 o o H
H
Q/4NN, = ccs5NI.H0//1n " ' cs'sN
n 0 0 AOH
(pH
)n ¨1_-...,1,01..r-0 **--H .
in n n "n_ n - n -n _ -n n N
i'kL Hi- and '')L
H n H n =

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

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a linker has a structure selected from among:
, \
q \
0, ),0A
),0;\ 0 N

It)vH
N a, S

\ 0 q O--OH
0,07µ I 0 N
I

H H
= -0-/1' . Nr("7 = 3 0 , -N1,1H
- H

(N I
N
I \ rO-P-01-1 II
." 0 ' \ , )0)L

H H
I
N,v).1\1v(,,k,_, ;

3 I 3 u \ 1,, =
0 ,^-^' 0 0 ,,0 NO' - N-P

Jvuv /-------7-1 6 cs -s 0 I
N
0 , L 7 D\õ.. 0 ,"o . -m-5 -0 o, 0 1 NCI-Oci H, ,). N 12. S,S *-gLO
i N ........$-).-3....*µ*--- NH N )& , H
JVVV

I O, HHHH H 0õ
.2e,p,N.k_vNA,N7N,gi_põ0,,, ; N
NC137(:),1 . r'js H ' 'SS' 1-&
0 ;

/
JVVV
I

õ I 0 (-30 0, 1 __ l( , P
N----/
OH
N ,cos S
0 N)0 \
H
,ss . S-S 0 N 12.vN N,s, ,(,c) ;
c' , H

SUBSTITUTE SHEET (RULE 26) I

,0 O
I\D.4-10110 0 I /----7-1 0 6 cs _ \-P,' 0 0 -s 0 H N
H N
1 =
Nv=I`vkil$1& , and I
o Jvw NO
O
S/( 0 6 cs \ /-----7-1 s-s 0 \ii,..
N
H . 1 IcNN'(9i0 .
H o SUBSTITUTE SHEET (RULE 26) In certain embodiments, a linker has a structure selected from among:

H H
.,1L7. N ..===, N ,2.L . .
H
cr .
H ' 0 , OH

H

0 y N /\/\csss ;
H j. ;
N 0 `z,,L)/\%4/- ;
cssLN /

H
H
/
0(:),N ,,,is .
=7-v).8,,,N 7-/ -/ , H H
.csss .

H
H H
N.,sss ; cs,'-y;s2ryN /1\1 ss ; and 0 0¨' H
H N
csscr N 0// NI =

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a linker has a structure selected from among:

H
H
\.) N NNA
. \ H
, 0 0 ' 0 0 i ;
N

cs N A .
N N , 11--*_,)-------õN,--\ ; csssµ ;

H
H
cccN cX0/\./N csss ;
cscsNH

H
H H N

0 0,1 ; I000,1 ;

H
csss N 0/1\1 ;
,-0¨P-0....p,000Ncsss ;

OH \13 3 LH

0 0,0-110¨ ; ckA--)NNII_Iµ and OH '3 '3 OH0 H ' 6 0 3 HIN " 60H =

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a linker has a structure selected from among:

N),10)\.

and n 0 wherein n is from 1 to 20.
In certain embodiments, a linker has a structure selected from among:
"of; IL07\7.0sc ; and In certain embodiments, a linker has a structure selected from among:
OH
OH

1-04-0 ON-NO rO¨P-0-1 00 and OH "
3 OH OH r-r3 3cr =
In certain embodiments, a linker has a structure selected from among:

f\HJ-L
N c 3 N
0 and 0 6 =
In certain embodiments, the conjugate linker has the structure:
rrri 0, ),(3)L

µ)C) .

In certain embodiments, the conjugate linker has the structure:

µ)C7)LNN("r40¨

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

SUBSTITUTE SHEET (RULE 26) ck)COL

2 H "5 OH

0 and 0 5 In certain embodiments, a linker has a structure selected from among:

A''kA N
Nvi.C)¨P1 0 and 0 wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
iv. Certain Cell-Tamting 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 Branchina 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.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a branching group has a structure selected from among:
¨
0.1.1/4 0 0 \
L
,((jZ
µ
N HO ' 0¨P-0-- µ)L ,t'-, N _\)=.s lri)r ;
NH 0 ( õ n CH3 61-I A-n ; (:)) I
>,.
H 0 ( ln H 0 0,7'hz. n rssYNNA
O
H H ; ;
8 ( )n r õJ 0 n( kr,01r1 H ;
'n m (C)1 iltiC?(µ CH3 n CH3 , I n 01 0 (:)
11/ im ' CH3pallik n i /)\¨NH F5 r 7 n 7 0 ?
.1 ,NH ( )?¨ N H csis ,)n k n .nly ;
) e , FNiThr V¨ N H fsis ,(NH 1---NH n n isss(\i())i1 `zzi. 0 n H
. /NN.7-1NA
H, di r 0 "7.(\)L-NH
V
n ....õ....rri 0 H H i 0 ...õ..... rr" 0 '22t.N NNA' ;and n H H
n H H 0 K =
O(,,/
rosi,6,7)(NH
'22( NH

SUBSTITUTE SHEET (RULE 26) 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:
.., / 7 \
o o \N HO 0-1g-0 .
i n \ ir ' in H l'rnY ;
OH

, n n I CH3 An ' 0) )11.
0 ( in 0 0., _>1. 1 H .. H n n ; vrcr VNr ..<;'µ N N ; )1/4 el .
( ) n H 0 ( )n H
prs4 prfr 0 ( 0 \ /n JUIN m I
N H( C12 S Vz2- 4 n N )ir..),1 CH3 0 . .
, cH3p," n Om NH6 \ n ;and 1 0 ¨
I
,NH (NH /
Jr )n n I
cFcscV.- NH /
H
\ 0)m wherein each n is, independently, from 1 to 20; and m is from 2 to 6.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a branching group has a structure selected from among:

'Llt ,>0 0 rs1.1 H

N H
I
0 /(vw _______________________________________________________________________ C) JVIJV
I ,¨ N
H rsss N H
0 0 o ' \ 0 \ N cr ) ./(0 .
0 , /' N , vv µ 0 H N N H
) y NH
V
H ?
ck N
c'' N
H

H O/

\
H N Ni v NH

N H N H
' \ .,)------H
N H j-L, cs rsss ; and - cr H
H O/ =
O/
\
rsss NH
v NH

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a branching group has a structure selected from among:
vv ( Ai n )nA1 ( fti Ai and %net, 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:

Ai )n A1A
/A1A ) _____________ j() __ A n n n A1 z Al and .5"
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:
iscs csis\
/µ ) )n Ai( in and `11,_)( )n \s3 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:

SUBSTITUTE SHEET (RULE 26) /0¨NH
In certain embodiments, a branching group has a structure selected from among:
o ,vv In certain embodiments, a branching group has a structure selected from among:
\,3 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 SUBSTITUTE SHEET (RULE 26) 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:

,1\1 C)07V\- = 51- "n 0 \

n H n H H
\;/ -()11µ

.

=

;and n H in 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:

µ=,\7.'N'\/ 0/\)117-- ; N Wcss' ; ss\.7\7\,A ;

; cris\/0)11. ; /\/,sss ; and scrcorNsi SUBSTITUTE SHEET (RULE 26) In certain embodiments, a tether has a structure selected from among:
H H
css' N N
"n I

wherein each n is, independently, from 1 to 20.
In certain embodiments, a tether has a structure selected from among:
0 Zi cskR L ,9)11. L.HA
and "LHjjN
mi ' mi mi H7 wherein L is either a phosphorus linking group or a neutral linking group;
Zi 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:
ck,7"\/NN

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

k2F0-11"¨Ot4) and ApA 0-11-01')A
4.1%. mi PH mi im1 H 0 ml wherein Z2 is H or CH3; and each mi 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:

SUBSTITUTE SHEET (RULE 26) "rN
4 H n H
r , 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-carboxyethy1]-2-deoxy- 13-D-g1ucopyranose (13-muramic acid), 2-Deoxy-2-methylamino-L-glucopyranose, 4,6-Dideoxy-4-formamido-2,3-di-0-methyl-D-mannopyranose, 2-Deoxy-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-glucopyranose, Methyl 2,3,4-tri-O-acety1-1-thio-6-0-trityl-a-D-glucopyranoside, 4-Thio-P-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-3-D-ga1actopyranose and a-form: 2-(Acetylamino)-2-deoxy-D-SUBSTITUTE SHEET (RULE 26) galactopyranose. In certain embodiments, both the [3-form: 2-(Acety1amino)-2-deoxy-[3-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 [3-form 2-(Acetylamino)-2-deoxy-D-galactopyranose is the preferred embodiment.

HO 1"/N
H
OH
2-(Acetylamino)-2-deoxy-D-galactopyranose OH
OF(LO
HO-NHAc 2-(Acety1amino)-2-deoxy-I3-D-ga1actopyranose OH
OH

HO
NHAc 2-(Acetylamino)-2-deoxy-a-D-galactopyranose In certain embodiments one or more ligand has a structure selected from among:

SUBSTITUTE SHEET (RULE 2 6 ) O
OH H
OH
HO

H0 --C..L_-i HO¨.7.(....)..) u 0-1HO OH
and R1 R1 0¨.....r.!.:)..\__0-1 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
HO
.....__:)H OH
_....4.)_\/ -0 0 -- 0 \IR ,0 HO .
NHAcNI ; HO mu OH , 0N Hr'i = HO --1 Xf ; HO , \/
HOOH OH
N HO OH HOOH
HO Nws ; 0 0 H HO OH
OH '' OH

HO-4).===\/N\_,/ ; HO---"\--C2---\/ +cirtOv\- ; and r OH
HO
..õ_C_....) HO
0 ________________ HO
HO OH
OH 1?.
-`) .
HO
0\ , In certain embodiments one or more ligand has a structure selected from among:
HOOH

HO N ,,.r NHAc r =

SUBSTITUTE SHEET (RULE 26) In certain embodiments one or more ligand has a structure selected from among:
HOOH
0 n HO-----"\----\/¨ Nos 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

__........70......0 HN N
HO HV\Z f(i \
n n i HO NHAc 0 n \
µ /n H
HO
n n NHAc 0 0 0-rjC in OH
HO HN
HO__,.\...C.:...\,01,,y..,.. n n NHAc 0 =
wherein each n is, independently, from 1 to 20.

SUBSTITUTE SHEET (RULE 26) In certain such embodiments, conjugate groups have the following structure:
HO OH
NHAc 0 HO OH 0¨
HO ..,...
(:).NNN
NHAc 0 0 (:)"
OH
HO
H

HO

NHAc =
In certain such embodiments, conjugate groups have the following structure:
HO H
H H
N._....f0 I
0=P¨OH
_......7Ø....\,...õ,0 N OH
HO NK
111)n 01 n NHAc \\/0),13x µ /n cf:

H n 0¨P=X
HO
n I
NHAc 0 )n HO OH
H

HO 0 9/n n NHAc 0 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.

SUBSTITUTE SHEET (RULE 26) In certain such embodiments, conjugate groups have the following structure:
HO H

__e I
=P-OH

Li OH 0 1.2"\-----NHAc \\) HO OH 0-., 0 ..õ
,0,..
N
H HO 0¨P=X I
NHAc 0 0 OH
0 (:)"
HO OH

HO ON....,..,./
NHAc 0 In certain such embodiments, conjugate groups have the following structure:
HO H
I
_......7Ø....\,....., H H
N.,_...i0 OH 1 0=F)-OH NH2 HO

N)\r) ) e..1., N
% / 3 NHAc \(0),N N.:.--i HO OH
O-.____ ON
-.._., _......\=.C...)....\_____0 µ H H
N,/, Iv N .7=N_____..0--N--.-------'i)r7P.4\ SI
HO
O¨P=0 I
NHAc 00 OH

HO OH
_..s...7Ø.\....õ, O

NHAc 0 In certain such embodiments, conjugate groups have the following structure:
HOOH
s......T.2,\v 0 HO
n 0 113 AcHN OH 1)n HO OH
_.....2...vn 0 0 HO µ-ir\r,-Vcr,,hN ] 1 n VI H_1 I
AcHN OH n0 P ,O) 0 I 0 n HO "n OH
NHAc .

SUBSTITUTE SHEET (RULE 26) In certain such embodiments, conjugate groups have the following structure:
OOH

AcHN 0 0, OH
HO\ ?Fi 0 0 HO---2-\/00'111`013' I
AcHN OH 0 HO OHo HO
NHAc In certain such embodiments, conjugate groups have the following structure:
HOOH

AcHN
OH ))n HOOH (/1\1-_rµNNI12 HO
________________________________________________ 0-P-0(N
n uluµ
AcHN OH OH os, HO-P=0 HO n OH
NHAc SUBSTITUTE SHEET (RULE 26) In certain such embodiments, conjugate groups have the following structure:
HO OH

HO*,..\70 ,K
AcHN
OH
\
vyj\Tr\NNII2 HO OH 0 0-õ. 0 ________________________________________________________ 0- j n 0 N
P--A

AcHN OH Cr 0':
HO-P=0 HO H i(:),, 6 .(21../0,Z'o' a HO
NHAc In certain such embodiments, conjugate groups have the following structure:

HO-1=0 0 (i)--( ,,,,I\I
N--_:---I-s:

I
HO-P=0 O
HO OH On _......(2.\.7 0 HO C)-)\' -1 n 0 1 0 \OH
AcHN OH 11) 0 HO OH n (On O--===="Z\/ 1 ,11, ,/ , H 0..__. 0 r, 0 1 0 , N/0-170 AcHN OH - u OH

,...c.L/ ) 0.,.,,,x....õ , ....
11 J, HO \ in OH
NHAc =

SUBSTITUTE SHEET (RULE 26) In certain such embodiments, conjugate groups have the following structure:

, iNE12 HO-P=0 I 1\1 -rN
0--0,c )õ.N
N-_---/-d HO-P=0 O

\OH
AcHN

(03 HO OH
V
0-.... I ,0 0 HO ___________ -0=71%.-\z -I'- \/W ________ /(:)1=1C1 AcHN OH (:) OH
HO H 9 y P-OH
HO
NHAc .
In certain embodiments, conjugates do not comprise a pyrrolidine.
SUBSTITUTE SHEET (RULE 26) In certain such embodiments, conjugate groups have the following structure:

II _Zi-4N
¨P-O-'ON

HOOH

I
HO-- 077(NN'.(:) 0=P-0-AcHN 0 I

HOOH
HH 0, HO___ ....C2..\/
H \
AcHN 0 0 0' bH
HOOH HN------kj H___/,/ 0 _...z/0____õTr..-N

AcHN .
In certain such embodiments, conjugate groups have the following structure:
HOOH
0 , 0 AcHN 0-HOOH
0 0-- oHO
_......Ø.s\z 0/\ -il, AcHN 0' o, o=P-o-HO H )---J
P- I, .....2.\./0....,z---.0' NHAc .
5 In certain such embodiments, conjugate groups have the following structure:
HO H

AcHN
\N
OH

HO
N \,N---N---ir-N.-0,--= ______________ N2 )6O

HOL%'/) C) 0 0 0' 0 NHAc HNN___Cj OH
HO/
HO
NHAc .

SUBSTITUTE SHEET (RULE 26) In certain such embodiments, conjugate groups have the following structure:

HO_õõ,r,EL-0Nrk AcHN N

/
AcHN 0 HOOH

.....
HO CYTrN

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

HO__Ts.1õ.0-1-r-NC

AcHN X0\ o _.I.C..p...\.,,,OrNJ=c7----0-.,-,,N,IL 0 N^Ho-15¨

z a AcHN 0 HOOH
---CI
__..,!.:)...\,..., HO OTrN 0 AcHN .
In certain such embodiments, conjugate groups have the following structure:
HOOH
H
_.,...?..\zcn=-r---N 0 AcHN

0-rN' N)c HO "4 H FNrHO ri I
H
AcHN
--HO OH

AcHN .

SUBSTITUTE SHEET (RULE 26) In certain such embodiments, conjugate groups have the following structure:
HO OH
H
_.......(2..vo HOr----N 0 AcHN

HO--12--\=/ 4 INI NiWNo¨ 9p / H H 4 - 6¨
AcHN
N
HOOH

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

HO&T.C.1., -------7)LNH
AcHN
OH OH
H0.7.1.....\_0 0 rcH 0 H 0 0,N N ,.......õ--...õ,õ=-=)(N---0--6 ED
N
AcHN 0 0 r .....7.,õ..z..)--NH
HO....;/0 HO
NHAc =
In certain such embodiments, conjugate groups have the following structure:
OH OH

HO&T.C..1., ------)LNH
AcHN
OH OH
HO*, 0 rcH 0 H 0,0 Nvr N---6 (5151 AcHN
0 r HO H
......r(2..\/0 HO
NHAc .

SUBSTITUTE SHEET (RULE 26) In certain such embodiments, conjugate groups have the following structure:
pH
HOOH
HO

AcHN
0=P¨OH
HOOH
HO-1(2-\VorNIR..0 AcHN
0=P¨OH

HOOH
HO
AcHN
In certain such embodiments, conjugate groups have the following structure:
pH
HOOH
HO

AcHN
0=P¨OH

HOOH
HO0'nr 0 AcHN
0=P¨OH
HoOH
HO
ON
AcHN ö.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
0 n AcHN

HO
AcHN
,0 HOOH
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
0 n AcHN

HO
AcHN
,0 HOOH
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"0 n AcHN

HO
AcHN
,0 HOOH
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.
SUBSTITUTE SHEET (RULE 26) In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH

N
Y\ n AcHN N

HO
H
AcHN H
HOOH
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\ n AcHN N

HO
H
AcHN H
HOOH
HO -¨
AcHN
wherein Y and Z are independently selected from a Ci-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.

SUBSTITUTE SHEET (RULE 26) In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH

Y\
AcHN N z -ON
HOOH OH
HO
AcHN H H
N, 0 HOOH y, Ic HO_.1.(2..\0/ 0 AcHN
wherein Y and Z are independently selected from a CI-Cu substituted or unsubstituted alkyl group.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH \
NA(cr HO
H n N
AcHN 0 HOOH
HO___,O;(-);---11 AcHN
HOOH

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:
HOOH
N jyy.,0 HO
H n N
AcHN 0 A
HO
AcHN/1-N1 -160 HOOH c\ 0 HO
AcHN
wherein m is 4, 5, 6, 7, or 8, and n is 1, 2, 3, or 4.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH

HOOH HO
AcHN
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
AcHN
HO ¨--AcHN
OFbH 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 HO
_4,,0AcHN
HO
AcHN
OFbH r- H
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.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
0 , HOOH
F1 0..y AcHN
HO_4,0 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-C11 alkyl group.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
HO
AcHN

O
HO N' AcHN
HOOH
HO
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
HO
AcHN

O HO N' AcHN
HOOH
HO
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.

SUBSTITUTE SHEET (RULE 26) In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HO OH
¨y ¨N
HO
AcHN

HO
AcHN
HOOH
HO
AcHN
wherein Y is selected from a C1-C12 substituted or unsubstituted alkyl group.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
HO n AcHN

nNN

AcHN
HOOH

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
/.pN 0 HO 0n AcHN

HO -AcHN
HOOH
n ENi AcHN
wherein n is 4, 5, 6, 7, or 8.
In certain embodiments, conjugates do not comprise a pyrrolidine.
SUBSTITUTE SHEET (RULE 26) 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¨DiE¨F) 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¨D¨EE¨F) 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.

SUBSTITUTE SHEET (RULE 26) 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¨EE¨F) 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) 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¨D¨EE¨F) wherein SUBSTITUTE SHEET (RULE 26) 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 -D
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
H OH ¨
NH, yy, HO 'N
NHAc 0 Ho r 0 _ 5 0 ___________________________________________________________________ HO NO 8=0 Ligand Tether OH
NHAc 0 0 0 _ 0 Linker Cleavable moiety OH ) \
HO H ,HN
N Branching group HO
NHAc 0 SUBSTITUTE SHEET (RULE 26) In certain embodiments, a conjugated antisense compound has a structure selected from among the following:
Cell targeting moiety HO OH

HO-4\7 A:L
Cleavable moiety AcHN 0 1 0, ¨
¨
OH
_ _ I\I iNE12 HO OH _ , _____ , 00õ 0 N

0 1 001C1-131¨()( _ AcHN OH 1C) 0 Tether y . _____ , ¨ -04=o Ligand HO OH
P-, ASO
_ HO OH
NHAc Branching group In certain embodiments, a conjugated antisense compound has a structure selected from among the following:

SUBSTITUTE SHEET (RULE 26) ASO
Cleavable moiety HO¨P=0 V _CNN
HO¨P=0 Cell targeting moiety ' 0 (s( H OH
AcHN 0 0, (K)3 HOOH 0 _ ______ Conjugate linker HO-4?-1(yilA;(7.W--\./0¨y=0 AcHNcy" - OH
Tether Ligand HO H
0"
HO
NHAc Branching group 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 SUBSTITUTE SHEET (RULE 26) 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;

SUBSTITUTE SHEET (RULE 26) 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; U52011/0097265; 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;
US2008/0281041; US2009/0203135; US2012/0035115; US2012/0095075;
US2012/0101148;
US2012/0128760; US2012/0157509; US2012/0230938; US2013/0109817;
US2013/0121954;
US2013/0178512; US2013/0236968; US2011/0123520; US2003/0077829;
US2008/0108801; and US2009/0203132; 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 SUBSTITUTE SHEET (RULE 26) 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 '-O-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.

SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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 SUBSTITUTE SHEET (RULE 26) 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 release the active oligonucleotide. In certain embodiments, it is not necessary that the entire conjugate group be cleaved from the oligonucleotide. For example, in Example 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 (Table 10a). This conjugated antisense compound demonstrated good potency (Table 10). 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 SUBSTITUTE SHEET (RULE 26) species. In certain embodiments, certain conjugate groups are more amenable to metabolism to the oligonucleotide.
D. Antisense 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 SUBSTITUTE SHEET (RULE 26) 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. Target Nucleic Acids, Regions and Segments In certain embodiments, conjugated antisense compounds target any nucleic acid. In certain embodiments, the target nucleic acid encodes a target protein that is clinically relevant. In such embodiments, modulation of the target nucleic acid results in clinical benefit. Certain target nucleic acids include, but are not limited to, the target nucleic acids illustrated in Table 1.
Table 1: Certain Human Target Nucleic Acids Target GENBANK Accession Number SEQ ID
NO
NM 002827.2 1 PTP1B NT 011362.9 truncated from nucleotides 14178000 to 14256000 NM 002011.3 3 FGFR4 NT 023133.11 truncated from nucleosides 21323018 to 21335213 SUBSTITUTE SHEET (RULE 26) PCT/US2014/036466_ AB209631.1 5 NM 022963.2 6 the complement of GENBANK Accession GCCR No. NT 029289.10 truncated from 7 nucleotides 3818000 to 3980000 NM 000160.3 8 GCGR NW 926918.1 truncated from nucleotides 16865000 to 16885000 NT 027140.6 truncated from nucleotides 1255000 to 1273000 NM 019616.2 11 Factor VII
DB184141.1 12 NW 001104507.1 truncated from nucleotides 691000 to 706000 NM 000128.3 14 NT 022792.17, truncated from 19598000 Factor XI to 19624000 NM 028066.1 16 NW 001118167.1 17 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 5 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 10 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 15 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 SUBSTITUTE SHEET (RULE 26) 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 the nucleic acids listed in Table 1 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 the nucleic acids listed in Table 1 can be conjugated as described herein.
1. Protein tyrosine phosphatase 1B (PTP1B) Protein tyrosine phosphatase 1B (PTP1B) is a member of a family of PTPs (Barford, et al., Science 1994. 263: 1397-1404) and is a cytosolic enzyme (Neel and Tonks, Curr. Opin.
Cell Biol. 1997. 9: 193-204).
PTP1B is expressed ubiquitously including tissues that are key regulators of insulin metabolism such as liver, muscle and fat (Goldstein, Receptor 1993. 3: 1-15), where it is the main PTP
enzyme.
PTP1B is considered to be a negative regulator of insulin signaling. PTP1B
interacts with and dephosphorylates the insulin receptor, thus attenuating and potentially terminating the insulin signaling transduction (Goldstein et al., J. Biol. Chem. 2000. 275: 4383-4389). The physiological role of PTP1B in insulin signaling has been demonstrated in knockout mice models. Mice lacking the PTP1B gene were protected against insulin resistance and obesity (Elchebly et al., Science 1999. 283: 1544-1548). PTP1B-deficient mice had low adiposity, increased basal metabolic rate as well as total energy expenditure and were protected from diet-induced obesity. Insulin-stimulated glucose uptake was elevated in skeletal muscle, whereas adipose tissue was unaffected providing evidence that increased insulin sensitivity in PTP1B-deficient mice was tissue-specific (Klaman et al., Mol. Cell. Biol. 2000. 20:
5479-5489). These mice were phenotypically normal and were also resistant to diet-induced obesity, insulin resistance and had significantly lower triglyceride levels on a high-fat diet. Therefore, inhibition of PTP1B
in patients suffering from Type II
diabetes, metabolic syndrome, diabetic dyslipidemia, or related metabolic diseases would be beneficial.
Antisense inhibition of PTP1B provides a unique advantage over traditional small molecule inhibitors in that antisense inhibitors do not rely on competitive binding of the compound to the protein and inhibit activity directly by reducing the expression of PTP1B. Antisense technology is emerging as an effective SUBSTITUTE SHEET (RULE 26) means for reducing the expression of certain gene products and may therefore prove to be uniquely useful in a number of therapeutic, diagnostic, and research applications for the modulation of PTP1B.
There is a currently a lack of acceptable options for treating metabolic disorders. It is therefore an object herein to provide compounds and methods for the treatment of such diseases and disorder.
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 for the portions of the document discussed herein, as well as in their entirety.
Certain Conjugated Antisense Compounds Targeted to a PTP1B Nucleic Acid In certain embodiments, conjugated antisense compounds are targeted to a PTP1B
nucleic acid having the sequence of GENBANKO Accession No. NM_002827.2, incorporated herein as SEQ ID NO: 1 or GENBANK Accession No. NT 011362.9 truncated from nucleotides 14178000 to 14256000, incorporated herein as SEQ ID NO: 2. In certain such embodiments, a conjugated antisense compound targeted to SEQ ID
NO: 1 is at least 90%, at least 95%, or 100% complementary to SEQ ID NOs: 1 and/or 2.
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: 54. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 54.
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: 55. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 55.
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: 56. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 56.
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: 57. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 57.
Table 2: Antisense Compounds targeted to PTP1B SEQ ID NO: 1 Target SEQ ID
ISIS No Start Sequence (5'-3') Motif NO
Site SUBSTITUTE SHEET (RULE 26) _WO 2014/179629 PCT/US2014/036466_ In certain embodiments, a compound comprises or consists of ISIS 142082 and a conjugate group.
ISIS 142082 is a modified oligonucleotide having the formula: Aes Aes Aes Tes Ges Gds Tds Tds Tds Ads Tds Tds mCds mCds Ads Tes Ges Ges mCes mCe, wherein, A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 404173 and a conjugate group.
ISIS 404173 is a modified oligonucleotide having the formula: Aes Aes Tes Ges Ges Tds Tds Tds Ads Tds Tds mCds mCds Ads Tds Ges Ges mCes mCes Ae, wherein, A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 409826 and a conjugate group.
ISIS 409826 is a modified oligonucleotide having the formula: Ges Ges Tes Tes Tes Ads Tds Tds mCds mCds Ads Tds Gds Gds mCds mCes Aes Tes Tes Ge, wherein, SUBSTITUTE SHEET (RULE 26) A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 446431 and a conjugate group.
ISIS 446431 is a modified oligonucleotide having the formula: Aes Aes Tes Ges Gds Tds Tds Tds Ads Tds Tds mCds mCds Ads Tes Ges Ges mCe, wherein, A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in US
7,563,884 and WO 2007/131237, which is incorporated by reference in its entirety herein, and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 17-96 and 244-389 disclosed in US
7,563,884 and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 886-1552 of SEQ ID Nos WO
2007/131237 and a conjugate group described herein.In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in US 7,563,884 and WO 2007/131237, which is incorporated by reference in its entirety herein, and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs disclosed in US 7,563,884 and WO
2007/131237 and a conjugate group described herein. The nucleobase sequences of all of the aforementioned referenced SEQ ID NOs are incorporated by reference herein.
PTP1B Therapeutic Indications SUBSTITUTE SHEET (RULE 26) In certain embodiments, provided herein are methods of treating an individual comprising administering one or more conjugated pharmaceutical compositions as described herein. In certain embodiments, the individual has metabolic related disease.
As shown in the examples below, conjugated compounds targeted to PTP1B, as described herein, have been shown to reduce the severity of physiological symptoms of metabolic related diseases, including metabolic syndrome, diabetes mellitus, insulin resistance, diabetic dyslipidemia, hypertriglyceridemia, obesity and weight gain. In certain of the experiments, the conjugated compounds reduced blood glucose levels, e.g., the animals continued to experience symptoms, but the symptoms were less severe compared to untreated animals. In other of the experiments, however, the conjugated compounds appear to reduce the symptoms of diabetes; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. In other of the experiments, however, the conjugated compounds appear to inhibit weight gain; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. In other of the experiments, however, the conjugated compounds appear to inhibit hypertriglyceridemia; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. The ability of the conjugated compounds exemplified below to restore function therefore demonstrates that symptoms of the disease may be reversed by treatment with a compound as described herein.
Diabetes mellitus is characterized by numerous physical and physiological symptoms. Any symptom known to one of skill in the art to be associated with Type 2 diabetes can be ameliorated or otherwise modulated as set forth above in the methods described above. In certain embodiments, the symptom is a physical symptom selected from the group consisting of increased glucose levels, increased weight gain, frequent urination, unusual thirst, extreme hunger, extreme fatigue, blurred vision, frequent infections, tingling or numbness at the extremities, dry and itchy skin, weight loss, slow-healing sores, and swollen gums.
In certain embodiments, the symptom is a physiological symptom selected from the group consisting of increased insulin resistance, increased glucose levels, increased fat mass, decreased metabolic rate, decreased glucose clearance, decreased glucose tolerance, decreased insulin sensitivity, decreased hepatic insulin sensitivity, increased adipose tissue size and weight, increased body fat, and increased body weight.
Liu and Chernoff have shown that PTP1B binds to and serves as a substrate for the epidermal growth factor receptor (EGFR) (Liu and Chernoff, Biochem. J., 1997, 327, 139-145).
Furthermore, in A431 human epidermoid carcinoma cells, PT1B was found to be inactivated by the presence of H202 generated by the addition of EGF. These studies indicate that PTP1B can be negatively regulated by the oxidation state of the cell, which is often deregulated during tumorigenesis (Lee et al., J. Biol.
Chem., 1998, 273, 15366-15372).

SUBSTITUTE SHEET (RULE 26) Overexpression of PTP1B has been demonstrated in malignant ovarian cancers and this correlation was accompanied by a concomitant increase in the expression of the associated growth factor receptor (Wiener et al., Am. J. Obstet. GynecoL, 1994, 170, 1177-1183).
PTP1B has been shown to suppress transformation in NIH3T3 cells induced by the neu oncogene (Brown-Shimer et al., Cancer Res., 1992, 52, 478-482), as well as in rat 3Y1 fibroblasts induced by v-srk, v-src, and v-ras (Liu et al., IlloL Cell. Biol., 1998, 18, 250-259) and rat-1 fibroblasts induced by bcr-abl (LaMontagne et al., Proc. Natl. Acad. Sci. U. S. A., 1998, 95, 14094-14099).
It has also been shown that PTP1B promotes differentiation of K562 cells, a chronic myelogenous leukemia cell line, in a similar manner as does an inhibitor of the bcr-abl oncoprotein. These studies describe the possible role of PTP1B in controlling the pathogenesis of chronic myeloid leukemia (LaMontagne et al., Proc. Natl. Acad. Sci. U. S. A., 1998, 95, 14094-14099).
Accordingly, provided herein are methods for ameliorating a symptom associated with hyperproliferative disorders in a subject in need thereof In certain embodiments, the hyperproliferative disorder is cancer. In certain embodiments, provided herein are methods for ameliorating a symptom associated with cancer. In certain embodiments, provided is a method for reducing the rate of onset of a symptom associated with hyperproliferative disorders. In certain embodiments, provided is a method for reducing the rate of onset of a symptom associated with cancer. In certain embodiments, provided is a method for reducing the severity of a symptom associated with hyperproliferative disorders. In certain embodiments, provided is a method for reducing the severity of a symptom associated with cancer. In such embodiments, the methods comprise administering to an individual in need thereof a therapeutically effective amount of a compound targeted to a PTP1B nucleic acid.
In certain embodiments, provided are methods of treating an individual comprising administering one or more conjugated pharmaceutical compositions as described herein. In certain embodiments, the individual has metabolic related disease.
In certain embodiments, administration of a conjugated antisense compound targeted to a PTP1B
nucleic acid results in reduction of PTP1B expression by at least about 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 99%, or a range defined by any two of these values.
In certain embodiments, pharmaceutical compositions comprising a conjugated antisense compound targeted to PTP1B are used for the preparation of a medicament for treating a patient suffering or susceptible to metabolic related disease.
In certain embodiments, the methods described herein include administering a compound comprising a conjugate group and a modified oligonucleotide having a contiguous nucleobases portion as described herein of a sequence recited in SEQ ID NOs: 54-57.

SUBSTITUTE SHEET (RULE 26) It will be understood that any of the compounds described herein can be used in the aforementioned methods and uses. For example, in certain embodiments a conjugated antisense compound targeted to a PTP1B nucleic acid in the aforementioned methods and uses can include, but is not limited to, a conjugated antisense compound targeted to SEQ ID NO: 1 comprising an at least 8 consecutive nucleobase sequence of any of SEQ ID NOs: 54-57; a conjugated antisense compound targeted to SEQ ID
NO: 1 comprising a nucleobase sequence of any of SEQ ID NOs: 54-57; a compound comprising or consisting of ISIS 142082, ISIS 404173, ISIS 409826, or ISIS 446431 and a conjugate group; a compound comprising an antisense oligonucleotide disclosed in US 7,563,884 and WO 2007/131237, which is incorporated by reference in its entirety herein, and a conjugate group; a compound comprising an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 17-96 and 244-389 disclosed in US
7,563,884 and a conjugate group described herein; or a compound comprising an antisense oligonucleotide having a nucleobase sequence of any of¨ SEQ ID NOs 886-1552 disclosed in WO 2007/131237 and a conjugate group described herein.
2. FGFR4 Obesity is considered a long-term metabolic disease. There are several serious medical sequelae related to obesity. There are over 1 billion overweight individuals worldwide with 100 million clinically obese. The increasing health care costs of treating obesity related diseases in the US
alone are estimated at over $100 billion annually. Current methods for treating obesity include behavioral modification, diet, surgery (gastroplasty), administering pharmaceutical agents that block appetite stimulating signals or absorption of nutrients (fat), and administering agents that increase thermogenesis or fat metabolism. Some of these methods have disadvantages in that they rely on patient resolve, are invasive, or have unwanted side effects.
An understanding of the mechanisms by which obesity is regulated may provide important therapeutic information.
Obesity is frequently associated with insulin resistance and together constitutes risk factors for later development of type 2 diabetes and cardiovascular diseases. Insulin resistance occurs well before development of type 2 diabetes, and insulin is overproduced to compensate for the insulin resistance and to maintain normal glucose levels. Type 2 diabetes ensues, as the pancreas can no longer produce enough insulin to maintain normal glucose levels. Early stages of type 2 diabetes are associated with elevated levels of insulin but as the disease progresses the pancreas may fail to produce insulin, resulting in increased blood glucose levels. Diabetes is a significant risk factor for both heart disease and stroke and is the leading cause of blindness and end-stage renal failure.
Diabetes is a disorder characterized by hyperglycemia due to deficient insulin action that may result SUBSTITUTE SHEET (RULE 26) from reduced insulin production or insulin resistance or both. Diabetes mellitus is a polygenic disorder affecting a significant portion of the people in the world. It is divided into two types. In type I diabetes, or insulin-dependent diabetes mellitus (IDDM), patients produce little or no insulin, the hormone that regulates glucose utilization. In type 2 diabetes, or noninsulin-dependent diabetes mellitus (NIDDM), patients often have plasma insulin levels that are the same compared to nondiabetic humans;
however, these patients have developed a resistance to the insulin stimulating effect of glucose and lipid metabolism in the main insulin-sensitive tissues, i.e., muscle, liver and adipose tissues, and the plasma insulin levels are insufficient to overcome the pronounced insulin resistance. Additionally, glucotoxicity, which results from long-term hyperglycemia, induces tissue-dependent insulin resistance (Nawano et al., Ain. J. Physiol. Endocrinol.
Metab., 278, E535-543) exacerbating the disease. Type 2 diabetes accounts for over 90% of all diabetes cases. It is a metabolic disorder characterized by hyperglycemia leading to secondary complications such as neuropathy, nephropathy, retinopathy, hypertriglyceridemia, obesity, and other cardiovascular diseases generally referred to as metabolic syndrome.
Metabolic syndrome is a combination of medical disorders that increase one's risk for cardiovascular disease and diabetes. The symptoms, including high blood pressure, high triglycerides, decreased HDL and obesity, tend to appear together in some individuals.
Metabolic syndrome is known under various other names, such as (metabolic) syndrome X, insulin resistance syndrome or Reaven's syndrome.
Diabetes and obesity (sometimes now collectively referred to as "diabesity") are interrelated in that obesity is known to exacerbate the pathology of diabetes and greater than 60%
of diabetics are obese. Most human obesity is associated with insulin resistance and leptin resistance. In fact, it has been suggested that obesity may have an even greater impact on insulin action than diabetes itself (Sindelka et al., Physiol Res., 51, 85-91). Additionally, several compounds on the market for the treatment of diabetes are known to induce weight gain, a very undesirable side effect to the treatment of this disease.
Therefore, a compound that has the potential to treat both diabetes and obesity would provide a significant improvement over current treatments.
Fibroblast growth factor receptor 4 (also known as FGF receptor-4, TKF;
tyrosine kinase related to fibroblast growth factor receptor; hydroxyaryl-protein kinase; tyrosylprotein kinase; Fgfr4; FGFR-4; FGFR4;
CD334, FGFR4 HUMAN and JTK2) has high affinity for the acidic and/or basic fibroblast growth factors.
(Armstrong et al., Genes Chromosomes Cancer, 4, 94-98).
Although FGFRs generally have been shown to have wide distribution throughout the body, to date, FGFR4 has only been found in a few tissues. Among a wide variety of cells and tissues tested, including human lymphocytes and macrophages, FGFR4 was found to be expressed in the lung and in some tumors of lung origin as well as in malignancies not derived from lung tissues.
(Holtrich et al., Proc. Nat. Acad. Sci.,.

SUBSTITUTE SHEET (RULE 26) 88, 10411-10415). FGFR4 has also been found to be expressed in the liver and in adipose tissues. (Patel et al., JCEM, 90(2), 1226-1232). FGFR4 has also been found to be expressed in certain carcinoma cell lines.
(Bange et al., Cancer Res., 62, 840-847).
Additionally, FGFR4 has been shown to play a role in systemic lipid and glucose homeostasis.
FGFR4-deficient mice on a normal diet exhibited features of metabolic syndrome that include increase mass of insulin resistance, in addition to hypercholesterolemia. FGFR4 deficiency was shown to alleviate high-fat diet-induced fatty liver in a certain obese mouse model, which is also a correlate of metabolic syndrome.
Restoration of FGFR4, specifically in hepatocytes of FGFR4 deficient mice, decrease plasma lipid level and restored the high fat diet-induced fatty liver but failed to restore glucose tolerance and sensitivity to insulin.
(Huang et al., Diabetes, 56, 2501-2510).
Antisense inhibition of FGFR4 provides a unique advantage over traditional small molecule inhibitors in that antisense inhibitors do not rely on competitive binding of the compound to the protein and inhibit activity directly by reducing the expression of FGFR4. A
representative United States patent that teaches FGFR4 antisense inhibitors includes US. Pat. Publication No.
US2010/0292140, of which is herein incorporated by reference in its entirety. Antisense technology is emerging as an effective means for reducing the expression of certain gene products and may therefore prove to be uniquely useful in a number of therapeutic, diagnostic, and research applications for the modulation of FGFR4.
There is a currently a lack of acceptable options for treating metabolic disorders. It is therefore an object herein to provide compounds and methods for the treatment of such diseases and disorder. This invention relates to the discovery of novel, highly potent inhibitors of FGFR4 gene expression.
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 for the portions of the document discussed herein, as well as in their entirety.
Certain Conjugated Antisense Compounds Targeted to a FGFR4 Nucleic Acid In certain embodiments, conjugated antisense compounds are targeted to a FGFR4 nucleic acid having the sequence GENBANK Accession No. NM_002011.3 (incorporated herein as SEQ ID NO: 3), GENBANK Accession No: NT 023133.11 truncated from nucleosides 21323018 to 21335213 (incorporated herein as SEQ ID NO: 4); and GENBANK Accession No. AB209631.1 (incorporated herein as SEQ ID NO:
5); and GENBANK Accession No NM 022963.2 (incorporated herein as SEQ ID NO:
6). In certain such embodiments, a conjugated antisense compound targeted to SEQ ID NOs: 3-6 is at least 90%, at least 95%, or 100% complementary to SEQ ID NOs: 3-6.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
3 comprises an at least 8 consecutive nucleobase sequence of any one of SEQ ID NOs: 58-65. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 3 comprises a nucleobase sequence of any one of SEQ ID NO: 12-19.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
3 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 58. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 3 comprises a nucleobase sequence of SEQ ID
NO: 59.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
3 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 59. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 3 comprises a nucleobase sequence of SEQ ID
NO: 59.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
3 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 60. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 3 comprises a nucleobase sequence of SEQ ID
NO: 60.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
3 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 61. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 3 comprises a nucleobase sequence of SEQ ID
NO: 61.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
3 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 62. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 3 comprises a nucleobase sequence of SEQ ID
NO: 62.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
3 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 63. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 3 comprises a nucleobase sequence of SEQ ID
NO: 63.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
3 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 64. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 3 comprises a nucleobase sequence of SEQ ID
NO: 64.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
3 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 65. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 3 comprises a nucleobase sequence of SEQ ID
NO: 65.
Table 3: Antisense Compounds targeted to FGFR4 SEQ ID NO: 3 ISIS No Target Start Sequence (5'-3') Motif SEQ
ID

SUBSTITUTE SHEET (RULE 26) _________________________________________________________________ _PCT/US2014/036466_ Site NO

In certain embodiments, a compound comprises or consists of ISIS 299005 and a conjugate group.
ISIS 299005 is a modified oligonucleotide having the formula: Ges Ges mCes Aes mCes Ads mCds Tds mCds Ads Gds mCds Ads Gds Gds Aes mCes mCes mCes mCe, wherein A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 463588 and a conjugate group.
ISIS 463588 is a modified oligonucleotide having the formula: Ges mCes Aes mCes Aes mCds Tds mCds Ads Gds mCds Ads Gds Gds Ads mCes mCes mCes mCes mCe, wherein A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, SUBSTITUTE SHEET (RULE 26) d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 463589 and a conjugate group.
ISIS 463589 is a modified oligonucleotide having the formula: Aes Ges Ges mCes Aes mCds Ads mCds Tds mCds Ads Gds mCds Ads Gds Ges Aes mCes mCes mCe, wherein A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 463690 and a conjugate group.
ISIS 463690 is a modified oligonucleotide having the formula: Ges mCes mCes Aes Ges Gds mCds Gds Ads mCds Tds Gds mCds mCds mCds Tes mCes mCes Tes Te, wherein A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 463691 and a conjugate group.
ISIS 463691 is a modified oligonucleotide having the formula: Tes Ges mCes mCes Aes Gds Gds mCds Gds Ads mCds Tds Gds mCds mCds mCes Tes mCes mCes Te, wherein A = an adenine, mC = a 5'-methylcytosine G = a guanine, SUBSTITUTE SHEET (RULE 26) T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 463835 and a conjugate group.
ISIS 463835 is a modified oligonucleotide having the formula: mCes Ges mCes Tes mCes Tds mCds mCds Ads Tds mCds Ads mCds Gds Ads Ges Aes mCes Tes mCe, wherein A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 463837 and a conjugate group.
ISIS 463837 is a modified oligonucleotide having the formula: mCes Aes mCes Ges mCes Tds mCds Tds mCds mCds Ads Tds mCds Ads mCds Ges Aes Ges Aes mCe, wherein A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 464225 and a conjugate group.
ISIS 464225 is a modified oligonucleotide having the formula: mCes Tes Tes mCes mCes Ads Gds mCds Tds Tds mCds Tds mCds Tds Gds Ges Ges mCes Tes mCe, wherein A = an adenine, SUBSTITUTE SHEET (RULE 26) mC = a 5'-methyleytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in WO
2009/046141, which are incorporated by reference in their entireties herein, and a conjugate group. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 21-24, 28, 29, 36, 38, 39, 43, 48, 51, 54-56, 58-60, 64-66, 92-166 disclosed in WO
2009/046141 and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence complementary to a preferred target segment of any of SEQ ID NOs 21-24, 28, 29, 36, 38, 39, 43, 48, 51, 54-56, 58-60, 64-66, 92-166 disclosed in WO
2009/046141 and a conjugate group described herein. The nucleobase sequences of all of the aforementioned referenced SEQ ID NOs are incorporated by reference herein.
FGFR4 Therapeutic Indications In certain embodiments, provided herein are methods of treating an individual comprising administering one or more pharmaceutical compositions as described herein. In certain embodiments, the individual has a metabolic disease.
As shown in the examples below, conjugated compounds targeted to FGFR4, as described herein, have been shown to reduce the severity of physiological symptoms of a metabolic disease, including obesity or adiposity, metabolic syndrome, diabetes mellitus, insulin resistance, diabetic dyslipidemia, and hypertriglyceridemia. In certain of the experiments, the conjugated compounds reduced body weight, e.g., the animals continued to experience symptoms, but the symptoms were less severe compared to untreated animals. In certain of the experiments, the conjugated compounds reduced body fat, e.g., the animals continued to experience symptoms, but the symptoms were less severe compared to untreated animals. In certain of the experiments, the conjugated compounds reduced adipose tissue, e.g., the animals continued to experience symptoms, but the symptoms were less severe compared to untreated animals. In other of the experiments, however, the conjugated compounds appear to reduce the symptoms of obesity; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. In other of the experiments, however, the conjugated compounds appear to SUBSTITUTE SHEET (RULE 26) reduce the symptoms of diabetes; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time.
In other of the experiments, however, the conjugated compounds appear to inhibit weight gain; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time.
In other of the experiments, however, the conjugated compounds appear to reduce glucose levels; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. In other of the experiments, however, the conjugated compounds appear to increase fatty acid oxidation; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. The ability of the conjugated compounds exemplified below to restore function therefore demonstrates that symptoms of the disease may be reversed by treatment with a compound as described herein.
Obesity is characterized by numerous physical and physiological symptoms. Any symptom known to one of skill in the art to be associated with obesity can be ameliorated or otherwise modulated as set forth above in the methods described above. In certain embodiments, the symptom is a physical symptom selected from the group consisting of increased adipose tissue mass or weight, increased weight gain, increased fat pad weight, imbalance with caloric intake and energy expenditure, increase in body fat, increase in body mass, having a body mass index (BMI) of 30 or higher, increase in body frame, increased sweating, sleep apnea, difficulty in sleeping, inability to cope with sudden physical activity, lethargy, back and joint problems, increase in breathlessness, increase in breast region adiposity, increase in abdomen size or fat, extreme hunger, or extreme fatigue.
In certain embodiments, the symptom is a physiological symptom selected from the group consisting of high blood pressure, hypertension, high cholesterol levels, type 2 diabetes, stroke, cardiac insufficiency, heart disease, coronary artery obstruction, breast cancer in women, gastro-oesophageal reflux disease, hip and knee arthrosis, and reduced life expectancy.
In certain embodiments, the physical symptom is excess body weight. In certain embodiments, the symptom is excess fat mass. In certain embodiments, the symptom is a body mass index of 30 or higher. In certain embodiments, the symptom is breathlessness. In certain embodiments, the symptom is increased sweating. In certain embodiments, the symptom is sleep apnea. In certain embodiments, the symptom is difficulty in sleeping. In certain embodiments, the symptom is inability to cope with sudden physical activity.
In certain embodiments, the symptom is lethargy. In certain embodiments, the symptom is back and joint problems.
In certain embodiments, the physiological symptom is high blood pressure. In certain embodiments, the symptom is hypertension. In certain embodiments, the symptom is high cholesterol levels. In certain embodiments, the symptom is type 2 diabetes. In certain embodiments, the symptom is stroke. In certain embodiments, the symptom is cardiac insufficiency. In certain embodiments, the symptom is heart disease. In SUBSTITUTE SHEET (RULE 26) certain embodiments, the symptom is coronary artery obstruction. In certain embodiments, the symptom is breast cancer in women. In certain embodiments, the symptom is gastro-oesophageal reflux disease. In certain embodiments, the symptom is hip and knee arthrosis. In certain embodiments, the symptom is reduced life expectancy.
Diabetes mellitus is characterized by numerous physical and physiological symptoms. Any symptom known to one of skill in the art to be associated with Type 2 diabetes can be ameliorated or otherwise modulated as set forth above in the methods described above. In certain embodiments, the symptom is a physical symptom selected from the group consisting of increased glucose levels, increased weight gain, frequent urination, unusual thirst, extreme hunger, extreme fatigue, blurred vision, frequent infections, tingling or numbness at the extremities, dry and itchy skin, weight loss, slow-healing sores, and swollen gums.
In certain embodiments, the symptom is a physiological symptom selected from the group consisting of increased insulin resistance, increased glucose levels, increased fat mass, decreased metabolic rate, decreased glucose clearance, decreased glucose tolerance, decreased insulin sensitivity, decreased hepatic insulin sensitivity, increased adipose tissue size and weight, increased body fat, and increased body weight.
In certain embodiments, the physical symptom is increased weight gain. In certain embodiments, the symptom is frequent urination. In certain embodiments, the symptom is unusual thirst. In certain embodiments, the symptom is extreme hunger. In certain embodiments, the symptom is extreme fatigue. In certain embodiments, the symptom is blurred vision. In certain embodiments, the symptom is frequent infections. In certain embodiments, the symptom is tingling or numbness at the extremities. In certain embodiments, the symptom is dry and itchy skin. In certain embodiments, the symptom is weight loss. In certain embodiments, the symptom is slow-healing sores. In certain embodiments, the symptom is swollen gums. In certain embodiments, the symptom is increased insulin resistance. In certain embodiments, the symptom is increased fat mass. In certain embodiments, the symptom is decreased metabolic rate. In certain embodiments, the symptom is decreased glucose clearance. In certain embodiments, the symptom is decreased glucose tolerance. In certain embodiments, the symptom is decreased insulin sensitivity. In certain embodiments, the symptom is decreased hepatic insulin sensitivity. In certain embodiments, the symptom is increased adipose tissue size and weight. In certain embodiments, the symptom is increased body fat. In certain embodiments, the symptom is increased body weight.
In certain embodiments, provided are methods of treating an individual comprising administering one or more pharmaceutical compositions as described herein. In certain embodiments, the individual has metabolic related disease.
In certain embodiments, administration of a conjugated antisense compound targeted to a FGFR4 nucleic acid results in reduction of FGFR4 expression by at least about 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 99%, or a range defined by any two of these values.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, pharmaceutical compositions comprising a conjugated antisense compound targeted to FGFR4 are used for the preparation of a medicament for treating a patient suffering or susceptible to a metabolic disease.
In certain embodiments, the methods described herein include administering a conjugated compound comprising a modified oligonucleotide having a contiguous nucleobases portion as described herein of a sequence recited in SEQ ID NO: 58-65.
It will be understood that any of the compounds described herein can be used in the aforementioned methods and uses. For example, in certain embodiments a conjugated antisense compound targeted to a FGFR4 nucleic acid in the aforementioned methods and uses can include, but is not limited to, a conjugated antisense compound targeted to SEQ ID NO: 3 comprising an at least 8 consecutive nucleobase sequence of any one of SEQ ID NOs: 58-65; a conjugated antisense compound targeted to SEQ
ID NO: 3 comprising a nucleobase sequence of any one of SEQ ID NO: 58-65; a compound comprising or consisting of ISIS
299005, ISIS 463588, ISIS 463589, ISIS 463690, ISIS 463691, ISIS 463835, ISIS
463837, or ISIS 464225 and a conjugate group; a compound comprising an antisense oligonucleotide disclosed in WO 2009/046141, which are incorporated by reference in their entireties herein, and a conjugate group; a compound comprising an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs SEQ ID NOs 21-24, 28, 29, 36, 38, 39, 43, 48, 51, 54-56, 58-60, 64-66, 92-166 disclosed in WO
2009/046141 and a conjugate group described herein; or a compound comprising an antisense oligonucleotide having a nucleobase sequence complementary to a preferred target segment of any of SEQ ID NOs 21-24, 28, 29, 36, 38, 39, 43, 48, 51, 54-56, 58-60, 64-66, 92-166 disclosed in WO 2009/046141 and a conjugate group described herein. The nucleobase sequences of all of the aforementioned referenced SEQ ID NOs are incorporated by reference herein.
3. GCCR
Diabetes is a chronic metabolic disorder characterized by impaired insulin secretion and/or action. In type 2 diabetes (T2DM), insulin resistance leads to an inability of insulin to control the activity of gluconeogenic enzymes, and many subjects also exhibit inappropriate levels of circulating glucagon (GC) in the fasting and postprandial state. Glucagon is secreted from the a-cells of the pancreatic islets and regulates glucose homeostasis through modulation of hepatic glucose production (Quesada et al., J. Endocrinol. 2008.
199: 5-19). .
Glucagon exerts its action on target tissues via the activation of glucocorticoid receptor (GCCR). The glucocorticoid receptor is a 62 kDa protein that is a member of the class B G-protein coupled family of receptors (Brubaker et al., Recept. Channels. 2002. 8: 179-88). GCCR
activation leads to signal transduction by G proteins (Gsa and Gq), whereby Gsa activates adenylate cyclase, which causes cAMP production, resulting in an increase in levels of protein kinase A. GCCR signaling in the liver results in increased hepatic SUBSTITUTE SHEET (RULE 26) glucose production by induction of glycogenolysis and gluconeogenesis along with inhibition of glycogenesis (Jiang and Zhang. Am. J. Physiol. Endocrinol. Metab. 2003. 284: E671-E678).
GCCR is also expressed in extrahepatic tissues, which includes heart, intestinal smooth muscle, kidney, brain, and adipose tissue (Hansen et al., Peptides. 1995. 16: 1163-1166).
Development of GCCR inhibitors have been hampered by the unfavorable side effects associated with systemic GCCR inhibition, including activation of the hypothalamic-pituitary adrenal (HPA) axis.
Inhibition of GCCR activity in the brain can lead to an increase in circulating adrenocorticotropic hormone due to feedback regulation and a consequent increase in secretion of adrenal steroids (Philibert et al., Front.
Horm. Res. 1991. 19: 1-17). This, in turn, can produce a myriad of negative chronic steroid-related side-effects. Other studies have demonstrated that specific inactivation of GCCR
resulted in hypoglycemia upon prolonged fasting (Opherk et al., Mol. Endocronol. 2004. 18: 1346-1353).
It has previously been demonstrated in pre-clinical models that administration of GCCR antisense oligonucleotides results in tissue-specific accumulation and reduced GCCR
expression in liver and adipose tissue (PCT Pub. No. W02005/071080, PCT Pub. No. W02007/035759) without affecting GCCR mRNA
levels in the CNS or adrenal glands. Thus, antisense inhibition of GCCR mRNA
expression has be shown to improve hyperglycemia and hyperlipidemia without activating the HPA axis. The present invention provides compositions and methods for modulating GCCR expression. Antisense compounds for modulating expression of GCCR are disclosed in the aforementioned published patent applications. However, there remains a need for additional improved compounds. The compounds and treatment methods described herein provide significant advantages over the treatments options currently available for GCCR related disorders.
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 for the portions of the document discussed herein, as well as in their entirety.
Certain Conjugated Antisense Compounds Targeted to a GCCR Nucleic Acid In certain embodiments, conjugated antisense compounds are targeted to a GCCR
nucleic acid having the sequence the complement of GENBANK Accession No. NT_029289.10 truncated from nucleotides 3818000 to 3980000 (incorporated herein as SEQ ID NO: 7). In certain such embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 is at least 90%, at least 95%, or 100% complementary to SEQ
ID NO: 7.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of any one of SEQ ID NOs: 66-77. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of any one of SEQ ID NO: 66-77.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 66. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 66.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 67. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 67.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 68. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 68.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 69. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 69.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 70. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 70.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 71. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 71.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 72. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 72.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 73. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 73.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 74. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 74.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 75. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 75.

SUBSTITUTE SHEET (RULE 26) In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 76. In certain embodiments, a conjtu,Yatecl antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 76.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
7 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 77. In certain embodiments a conjugated antisense compound targeted to SEQ ID NO: 7 comprises a nucleobase sequence of SEQ ID
NO: 77.
Table 1-54: Antisense Compounds targeted to GCCR SEQ ID NO: 7 Target Start SEQ ID
ISIS No Sequence (5'-3') Motif Site NO

In certain embodiments a comound comprises or consists of ISIS 420470 and. a conjugate group.
ISIS 420470 is a modified oligonucleatide having -the formula: Ges Ges Tes Aes Ges Ads Ads Ads Tds Ads Tds Ads Gds Tds Tds Ges TeS Tes rnCes rnCti.,, wherein A ¨ an adenine, mC ¨ a 5'-meth-v1c-vtosine G ¨ a guanine, T ¨ a thymine, e = a 2'-0-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and $ = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 420476 and a conjugate group.
ISIS 420476 is a modified oligonucleotide haying the formula: Tes Tes mCes Res Tes Gds Tds Gds Tds mCds Tds Gds mCds Ads Tds mCes Res Tes Ges To, wherein A = an adenine, mC = a 5'-methylcytosine G = aguanine,.
T = a thymine, e = a 2'-O-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside 'jaw.
In certain embodiments, a compound comprises or consists of ISIS 426115 and a conjugate group.
ISIS 426115 is a modified oligonucleotide having the formula: Ges mCes Aes Ges mCes mCds Ads Tds Gds Gds Tds Gds Ads Tds mCds Res Ges Ges Aes Ge wherein A ¨ an adenine, mC = a 5'-methylcvtosine G = a guanine,.
T = a thymine, e = a 2'-0-methoxvethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 426130 and a conjugate group.
"ISIS 426130 is a modified oligonucleotide having the formula: Ges rnCes Aes Tes mCes mCds Ads Gds mCds Gds Ads Gds mCds As mCds EnCes Aes Aes Aes Go wherein A = an adenine, mC = a 5'-methvicytosine G = a guanine, T a thymine, e = a 2'-O-methoxvethvl modified nucleoside, d = a 2'-deoxynucleoside. and $ = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 426168 and a conjugate group.
ISIS 426168 is a modified oligonucleotide having the formula: Ges Tes mCes Tes Ges Gds Ads Tds Tds Ads mCds Ads Gds mCds Ads Tes Aes Aes Aes mCe, wherein A = an adenine, inC = a 5'-methvicytosine G = a guanine, T = a thvmine, e = a 2'-O-rnetlioxvethvl modified nucleoside, d = a 2'-deoxynuc1eoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments. a compound comprises or consists of ISIS 426172 and a conjugate group.
ISIS 426172 is a modified oligonucleotide having the formula: mCes mCes Tes Tes Ges Gds Tds mCds Tds Gds Gds Ads Tds Tds Ads mCes Aes Ges mCes Ae. wherein A = an adenine, mC = a 5'-methylcytosine G = a guanine.
T = a thymine.
e == a 2'-0-methoxyethyl modified nucleoside.
d = a 2'-deoxynucleoside. and s = a phosphorothioate internucleoside = In certain embodiments, a compound comprises or consists of ISIS 426183 and a conjugate group.
ISIS 426183 is a modified oligonucleotide having the formula: Aes Ges mCes mCds Ads Tds Gds Gds Ids Gds Ads Tds mCds Ads Gds Gds Ads Ges Ges mCe wherein A = an adenine, mC = a 5'-methvIcvtosine G = a guanine.
T = a thymine.
e = a 2'-0-methoxyethyl modified nucleoside.
d = a 2'-deoxynucleoside. and s = a phosphorothioate internucleoside linkage.
In certain embodiments. a compound comprises or consists of ISIS 426246 and a conjugate group.
ISIS 426246 is a modified oligonucleotide having the formula: Ges Ges Tes mCds Tds Gds Gds Ads Tds Tds Ads mCds Ads Gds mCds Ads TcIs Aes Ass Ae wherein A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-metlioxyethyl modified nucleoside., d = a 2'-deoxynucleoside. and s = a phosphorothioate internucleoside linkage.
In certain embodiments. a compound comprises or consists of ISIS 426261 and a conjugate group. ISIS
426261 is a modified olig.onucleotide haying.the formula: Aes Ges mCds mCds Ads Tds Gds Gds Tds Gds Ads Ids mCds Ads Gds Ges Aes Ges Ges rnCe, wherein A = an adenine, mC = a 5'-methvicvtosine G = a panine, T = a thymine.
e = a 2'-0-methoxyethyl modified nucleoside.
d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments. a compound comprises or consists of ISIS 426262 and a conjugate group. ISIS
426262 is a modified oligonucleotide having the formula: mCes Aes Gds mCds mCds Ads Tds Gds Gds Tds Gds Ads Tds mCds Ads Ges Ges Aes Ges Ge wherein A = an adenine, inC =. a 5'-methylcytosine G = a guanine, T = a thvinine, e = a 2'-O-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 426267 and a conjugate group. ISIS
426267 is a inodified oligonucleotide having the formula: Ges Tes Gds mCds Tds Tds Gds Tds mCds mCds Ads Gds Gds Ads Tds Ges Aes Tes Ges mCe,. wherein A = an adenine, inC ¨ a 5'-methylcytosine G ¨ a guanine, T = a thymine,.
e = a 2'-0-metlioxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a nhosphorothioate internucleoside linkage.
In certain embodiments. a compound comprises or consists of ISIS 426325 and a conhgate group. ISIS
426325 is a modified oligonucleotide having the formula: mCes mCes Tds Tds Gds Gds Tds mCds Tds Gds Gds Ads Tds Tds Ads mCes Aes Ges inCes Ae. wherein A = an adenine, mC = a 5'-methvIcytosine (.ì = a guanine, T = a thymine, e = a 2'-O-rnethoxvethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in WO
2005/071080, WC) 2007/035759, or WO 2007/136988, which are incorporated by reference in their entireties herein, and a conkj.gate group. In certain embodiments,. a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ. 111) NOs 30-216, and 306-310 disclosed in WO 2005/071080 and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 26-113 disclosed in WO 2007/035759 and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 413-485 disclosed in WO 2007/136988 and a collimate group described herein. In certain embodiments a compound comprises an antisense oligonucleotide having a nucleobase sequence complementary to a preferred target segment of any of SEQ ID
NOs 30-216, and 306-310 disclosed in WO 2005/071080, 26-113 disclosed in WO
2007/035759, and 413-485 disclosed in WO 2007/136988, and a conjugate group described herein. The nucleobase sequences of all of the aforementioned referenced SEQ ID NOs are incorporated by reference herein.
GCCR Therapeutic Indications In certain embodiments, provided herein are methods of treating an individual comprising administering one or more pharmaceutical compositions as described herein. In certain embodiments, the individual has metabolic related disease.
As shown in the examples below, conjugated compounds targeted to GCCR, as described herein, have been shown to reduce the severity of physiological symptoms of metabolic related diseases, including metabolic syndrome, diabetes mellitus, insulin resistance, diabetic dyslipidemia, hypertriglyceridemia, obesity and weight gain e.g., the animals continued to experience symptoms, but the symptoms were less severe compared to untreated animals. In certain experiments, the conjugated compounds reduced blood glucose levels,. In other experiments, the conjugated compounds reduce the symptoms of diabetes. In other experiments, the conjugated compounds inhibit weight gain. In other experiments, the conjugated compounds inhibit hypertriglyceridemia. In certain embodiements, the conjugated compounds restore function therefore demonstratingreversal of disease by treatment with a compound as described herein. In certain embodiments, animals treated for a longer period of time experience less severe symptoms than those administered the compounds for a shorter period of time.
Diabetes mellitus is characterized by numerous physical and physiological signs and/or symptoms.
Any symptom known to one of skill in the art to be associated with Type 2 diabetes can be ameliorated or otherwise modulated as set forth above in the methods described above. In certain embodiments, the sign or symptom is a physical symptom such as increased glucose levels, increased weight gain, frequent urination, unusual thirst, extreme hunger, extreme fatigue, blurred vision, frequent infections, tingling or numbness at the extremities, dry and itchy skin, weight loss, slow-healing sores, and swollen gums. In certain embodiments, the sign or symptom is a physiological symptom such as increased insulin resistance, increased glucose levels, increased fat mass, decreased metabolic rate, decreased glucose clearance, decreased glucose tolerance, decreased insulin sensitivity, decreased hepatic insulin sensitivity, increased adipose tissue size and weight, increased body fat, and increased body weight.
In certain embodiments, the physical sign or symptom is increased glucose levels. In certain embodiments, the sign or symptom is weight gain. In certain embodiments, the sign or symptom is frequent urination. In certain embodiments, the sign or symptom is unusual thirst. In certain embodiments, the sign or symptom is extreme hunger. In certain embodiments, the sign or symptom is extreme fatigue. In certain embodiments, the sign or symptom is blurred vision. In certain embodiments, the sign or symptom is frequent infections. In certain embodiments, the sign or symptom is tingling or numbness at the extremities. In certain embodiments, the sign or symptom is dry and itchy skin. In certain embodiments, the sign or symptom is weight loss. In certain embodiments, the sign or symptom is slow-healing sores. In certain embodiments, the sign or symptom is swollen gums. In certain embodiments, the sign or symptom is increased insulin resistance. In certain embodiments, the sign or symptom is increased glucose levels. In certain embodiments, the sign or symptom is increased fat mass. In certain embodiments, the sign or symptom is decreased metabolic rate. In certain embodiments, the sign or symptom is decreased glucose clearance. In certain embodiments, the sign or symptom is decreased glucose tolerance. In certain embodiments, the sign or symptom is decreased insulin sensitivity. In certain embodiments, the sign or symptom is decreased hepatic insulin sensitivity. In certain embodiments, the sign or symptom is increased adipose tissue size and weight.
In certain embodiments, the sign or symptom is increased body fat. In certain embodiments, the sign or symptom is increased body weight.
In certain embodiments, provided are methods of treating an individual comprising administering one or more pharmaceutical compositions as described herein. In certain embodiments, the individual has metabolic related disease.
In certain embodiments, administration of a conjugated antisense compound targeted to a GCCR
nucleic acid results in reduction of GCCR expression by at least about 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 99%, or a range defined by any two of these values.
In certain embodiments, pharmaceutical compositions comprising a conjugated antisense compound targeted to GCCR are used for the preparation of a medicament for treating a patient suffering or susceptible to metabolic related disease.
In certain embodiments, the methods described herein include administering a conjugated compound comprising a modified oligonucleotide having a contiguous nucleobases portion as described herein of a sequence recited in SEQ ID NO: 66-77.
it will be understood that any of the compounds described herein can be used in the aforementioned methods and uses. For example, in certain embodiments a conjugated antisense compound targeted to a GCCR nucleic acid in the aforementioned methods and uses can include,_but is not limited to, a conjugated antisense compound targeted to SEQ ID NO: 7 comprising an at least 8 consecutive nucleobase sequence of any one of SEQ ID NOs: 66-77: a conjugated antisense compound targeted to SEQ
ID NO: 7 comprising a nucleobase sequence of any one of SEQ ID NO: 66-77; a compound comprising or consisting of ISIS
420470, ISIS 420476, ISIS 426115, ISIS 426130, ISIS 426168, ISIS 426172, _ISIS 426183,_ISIS 426246, ISIS
426262, ISIS 426267, or ISIS 426325 and a conjugate group; a compound comprising an antisense oligonucleotide disclosed in VVO 2005/071080, WO 2007/035759, or WO
2007/136988, which are incorporated by reference in their entireties herein, and a conjugate group; a compound comprising an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 26-113 disclosed in Vs,10 2007/035759 and a conjugate group described herein; a compound comprising an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 26-113 disclosed in WO
2007/035759 and a conjugate group. described hereinL. or a compound comprising an antisense oliggnucleotide having a nucleobase sezience of any of SET) ID NOs 413-485 disclosed in \NO 2007/136988 and a conjugate group described herein; The nucleobase sequences of all of the aforementioned referenced SEQ
NOs are incorporated by reference herein.
4. GCGR
Insulin and glucagon are two pancreatic hormones involved in regulating glucose homeostasis and metabolism. Glucagon is secreted from the a-cells of the pancreatic islets and regulates glucose homeostasis through modulation of hepatic glucose production (Quesada et al., J. Endocrinol. 2008. 199: 5-19). The main function of glucagon is to counteract the actions of insulin.
Dysregulation of glucose metabolism may be caused either by defective insulin secretion and/or action, or by impaired postprandial glucagon suppression (Shah et al., Am. J.
Physiol. Endocrinol. Metab.
1999. 277: E283-E290). Inhibition of postprandial glucagon secretion in diabetic subjects has been shown to substantially reduce blood glucose, suggesting that glucagon contributes significantly to the hyperglycemia seen in subjects with type 2 diabetes mellitus (Shah et al., J. Clin.
Endocrinol. Metab. 2000. 85: 4053-4059).
Type 2 diabetes is characterized by impaired insulin secretion and/or action, and many subjects also exhibit inappropriate levels of circulating glucagon in the fasting and postprandial state. An increase in the glucagon/insulin ratio is likely an important determinant of the hyperglycemia seen in type 2 diabetes patients (Baron et al., Diabetes. 1987. 36: 274-283). Lack of suppression of postprandial glucagon secretion in subjects with T2DM also plays an important role in the pathogenesis of postprandial hyperglycemia (Henkel et al., Metabolism. 2005. 54: 1168-1173).
Glucagon exerts its action on target tissues via the activation of its receptor, GCGR. The glucagon receptor is a 62 kDa protein that is a member of the class B G-protein coupled family of receptors (Brubaker et al., Recept. Channels. 2002. 8: 179-88). GCGR activation leads to signal transduction by G proteins (Gsa and Gq), whereby Gsa activates adenylate cyclase, which causes cAMP
production, resulting in an increase in levels of protein kinase A. GCGR signaling in the liver results in increased hepatic glucose production by induction of glycogenolysis and gluconeogenesis along with inhibition of glycogenesis (Jiang and Zhang. Am.
J. Physiol. Endocrinol. Metab. 2003. 284: E671-E678). GCGR is also expressed in extrahepatic tissues, which includes heart, intestinal smooth muscle, kidney, brain, and adipose tissue (Hansen et al., Peptides. 1995.
16: 1163-1166).
Antisense inhibition of GCGR provides a unique advantage over traditional small molecule inhibitors in that antisense inhibitors do not rely on competitive binding of the compound to the protein and inhibit activity directly by reducing the expression of GCGR. A representative United States patent that teaches GCGR antisense inhibitors includes US. Pat. No. 7,750,142, of which is herein incorporated by reference in its entirety. Antisense technology is emerging as an effective means for reducing the expression of certain gene products and may therefore prove to be uniquely useful in a number of therapeutic, diagnostic, and research applications for the modulation of GCGR.
There is a currently a lack of acceptable options for treating metabolic disorders. It is therefore an object herein to provide compounds and methods for the treatment of such diseases and disorder.This invention relates to the discovery of novel, highly potent inhibitors of GCGR
gene expression.
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 for the portions of the document discussed herein, as well as in their entirety.
Certain Conjugated Antisense Compounds Targeted to a GCGR Nucleic Acid In certain embodiments, conjugated antisense compounds are targeted to a GCGR
nucleic acid having the sequence GENBANK Accession No. NM_000160.3 (incorporated herein as SEQ ID NO: 8) or GENBANK Accession No: NW_926918.1 truncated from nucleotides 16865000 to 16885000 (incorporated herein as SEQ ID NO: 9). In certain such embodiments, a conjugated antisense compound targeted to SEQ ID
NOs: 8 and/or 9 is at least 90%, at least 95%, or 100% complementary to SEQ ID
NOs: 8 and/or 9.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
8 and/or 9 comprises an at least 8 consecutive nucleobase sequence of any one of SEQ ID
NOs: 78-83. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: : 8 and/or 9 comprises a nucleobase sequence of any one of SEQ ID NO: 78-83.

In certain emboditnents, a conjugated antisense compo-und targeted to SEQ ID
NO: 8 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 78. In certain embodiments: a conjugated antisense compound -targeted to SEQ ID NO: 8 comprises a nucleobase sequence of SEQ ID
NO: 78.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
9 comprises an at least consecutive nucleobase sequence of SEQ -ID NO: 79. In. certai.n embodim.ents: a conjuptcd antisense cotnpo-u-nd targeted to SEQ ID NO: 9 comprises a nucleobase Sequence of SEQ
ID NO: 79.
In certain embodiments, a conjugated antisense comp_ound tatgetcd to SEQ1D NO:
9 comprises an at least S consecutive nucleobase sequence of SEQ ID NO: 80. In certain embodiments, a conjugated antisense compound targ_eted to SEQ ID NO: 9 comprises a nucleobase sequence of SEQ ID
NO: 80.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
9 comprises an at least S consecutive nucleobase sequence of SEQ ID NO: 81. In certain embodiments, a conjugated antisense comppund targeted to SEQ JD -NO: 9 comprises a nucleobase semence of SEQ ID
NO: 81.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
8 and 9 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 82: in certain embodiments, a conjuoated antisense compound taroeted to SEQ ID -NO: 8 and 9 comprises a nucleobase sequence of SEQ ID
NO: 82.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
9 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 83. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 9 coinprises a nucleobase sequence of SEQ ID
NO: 83:
Table 4-5: Antisense Compounds targeted to GCGR SEQ ID NOs: 8 and 9 Target Target Start Start Site to Site to SEQSEQ ID
ISIS No Sequence (5'-3') Motif SEQ ID ID NO: 9 NO
NO: 8 325568 n/a GCACTTTGTGGTGCCAAGGC 78 n/a 5-10-5 MOE

n/a 5-10-5 MOE

n/a 3-10-4 MOE

459157 n 7341 /a 5-10-6 MOE

In certain embodiments, a compound comprises or consists of ISIS 325568 and a conjugate group.
ISIS 325568 is a modified otigonucleotide having the formula. Ges mCes Ads mCds Tds Tds Tds Gds Tds Gds Gds Tds Gds meds mCds Ads Ads Gds Ges mCe 14:11grch A = an adenine, mC = a 5"-methylcytosine T = a thvmine_, IllgthoxYetilE1_TOSlifig0 juNleoside.
la 2'-deOxyriuedeo5ide .110 lir.11q-ge=
In certain embodiments, a compound comprises or consists of ISIS 398471 and a conjugate group.
ISIS 398471 is a modified ottgonucleotide having the formula: Tes mees mees Aes mCes Ms Gds Gds mCds mCds Ads mOds Ads Gds Gds Tes Ges Ges Ges mCe, wherein A -= an adenine, inC =. a 5'-methylcytosine G = a guanine,.
T = a thvinine, e = a 2'-O-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 448766 and a conjugate group.
ISIS 448766 is a modified oligonucleotide having the formula: Ges triCes Aes Aes Ges Gds mCds Tds mCds Gds Gds Tds Tds Gds Gds Ges mCes Tes Tes mCe wherein A ¨ an adenine, inC ¨ a 5'-methvIcvtosine G ¨ a guanine, T = a thymine,.
e = a 2'-0-methoxvethvl modified nucleoside, d = a 2'-deoxvnucleoside, and s = a nhosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 449884 and a conkgate group.
ISIS 449884 is a modified oligonucleotide having the formula: Ges Ges Tes Tds mCds mCds mCds Gds Ads Gds Gds Tds Gds mCes mCes mCes Ae, wherein A = an adenine, mC = a 5'-methvIcvtosine G = a guanine,.
T = a thymine, e = a 2'-O-rnethoxvethvl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 459014 and a conjugate group.
ISIS 459014 is a modified oligonucleotide having the formula: Ges Ges Ges mCds Ads Ads Tds Gds mCds Ads Gds Tds mCds mCes Tes Ges Ge, wherein A an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-O-methoxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 459157 and a conjugate group.
ISIS 459157 is a modified oligonucleotide haying the formula: Gess Ges Ges Tes Tes mCds mCds mCds Gds Ads Gds Gds Tds Gds mCds raCes mCes Aes Aes Tes Ge, wherein A = an adenine, mC ¨ a 5'-methylcytosine G ¨ a guanine,.
T = a thymine, e = a 2'-0-metlioxyethyl modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in US
7,750,142, US 7,399,853, WO 2007/035771, or WO 2007;134014, which are incorporated by reference in their entireties herein, and a conjugate group. In certain embodiments, a compound comprises an antisense oligonucleotide havinga nucleobase sequence of any of SEQ ID NOs 20-399 disclosed in US 7,750,142 and a coniugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide haying a nucleobase sequence of any of SEQ ID NOs 20-399 disclosed in US 7,399.853 and a conjugate group described herein. In certain embodiments. a compound comprises an antisense oligonucleotide haying a nucleobase sequence of any of SEQ ID NOs 2 disclosed in WO 2007/035771 and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide haying a nucleobase sequence of any of SEO ID NOs: 486-680 disclosed in WO
2007/134014 and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide haying a nucleobase sequence complementary to a preferred target segment of any of SEQ ID NOs 20-399 of US 7,750,142, SE . ID NO: 20-399 of US 7,399,853, SEQ
ID NO 2 of WO

2007/035771, or SEQ ID NOs. 486-680 of WO 2007/134014, and a conjugate group described herein. The nucleobase sequences of all of the aforementioned referenced SEQ ID NOs are incorporated by reference herein, GCGR Therapeutic Indications In certain embodiments, provided herein are methods of treating an individual comprising administering one or more pharmaceutical compositions as described herein. In certain embodiments, the individual has metabolic related disease.
As shown in the examples below, conjugated compounds targeted to GCGR, as described herein, have been shown to reduce the severity of physiological symptoms of metabolic related diseases, including metabolic syndrome, diabetes mellitus, insulin resistance, diabetic dyslipidemia, hypertriglyceridemia, obesity and weight gain. In certain of the experiments, the conjugated compounds reduced blood glucose levels, e.g., the animals continued to experience symptoms, but the symptoms were less severe compared to untreated animals. In other experiments, however, the conjugated compounds appear to reduce the symptoms of diabetes; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. In other experiments, however, the conjugated compounds appear to inhibit weight gain; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. In other experiments, however, the conjugated compounds appear to inhibit hypertriglyceridemia;
e.g., animals treated for a longer period of time experienced less severe signs and/or symptoms than those administered the compounds for a shorter period of time. The ability of the conjugated compounds exemplified below to restore function therefore demonstrates that symptoms of the disease may be reversed by treatment with a compound as described herein.
Diabetes mellitus is characterized by numerous physical and physiological signs and/or symptoms.
Any symptom known to one of skill in the art to be associated with Type 2 diabetes can be ameliorated or otherwise modulated as set forth above in the methods described above. In certain embodiments, the symptom or sign is a physical symptom or sign ssuch as increased glucose levels, increased weight gain, frequent urination, unusual thirst, extreme hunger, extreme fatigue, blurred vision, frequent infections, tingling or numbness at the extremities, dry and itchy skin, weight loss, slow-healing sores, and swollen gums. In certain embodiments, the symptom or sign is a physiological symptom or sign selected from the group consisting of increased insulin resistance, increased glucose levels, increased fat mass, decreased metabolic rate, decreased glucose clearance, decreased glucose tolerance, decreased insulin sensitivity, decreased hepatic insulin sensitivity, increased adipose tissue size and weight, increased body fat, and increased body weight.
In certain embodiments, the physical symptom or sign is increased glucose levels. In certain embodiments, the sign or symptom is weight gain. In certain embodiments, the symptom is frequent urination. In certain embodiments, the symptom is unusual thirst. In certain embodiments, the symptom is extreme hunger. In certain embodiments, the symptom is extreme fatigue. In certain embodiments, the symptom is blurred vision. In certain embodiments, the symptom is frequent infections. In certain embodiments, the symptom is tingling or numbness at the extremities. In certain embodiments, the symptom is dry and itchy skin. In certain embodiments, the sign or symptom is weight loss. In certain embodiments, the symptom is slow-healing sores. In certain embodiments, the symptom is swollen gums. In certain embodiments, the symptom or sign is increased insulin resistance. In certain embodiments, the symptom or sign is increased glucose levels. In certain embodiments, the symptom or sign is increased fat mass. In certain embodiments, the symptom or sign is decreased metabolic rate. In certain embodiments, the symptom o rsign is decreased glucose clearance. In certain embodiments, the symptom or sign is decreased glucose tolerance. In certain embodiments, the symptom or sign is decreased insulin sensitivity. In certain embodiments, the symptom or sign is decreased hepatic insulin sensitivity. In certain embodiments, the symptom or sign is increased adipose tissue size and weight. In certain embodiments, the symptom or sign is increased body fat. In certain embodiments, the sign or symptom is increased body weight.
In certain embodiments, provided are methods of treating an individual comprising administering one or more pharmaceutical compositions as described herein. In certain embodiments, the individual has metabolic related disease.
In certain embodiments, administration of a conjugated antisense compound targeted to a GCGR
nucleic acid results in reduction of GCGR expression by at least about 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 99%, or a range defined by any two of these values.
In certain embodiments, pharmaceutical compositions comprising a conjugated antisense compound targeted to GCGR are used for the preparation of a medicament for treating a patient suffering or susceptible to metabolic related disease.
In certain embodiments, the methods described herein include administering a conjugated compound comprising a modified oligonucleotide having a contiguous nucleobases portion as described herein of a sequence recited in SEQ ID NO: 78-83.
It will be understood that any of the compounds described herein can be used in the aforementioned methods and uses For example, in certain embodiments a conjugated antisense compound targeted to a GCGR nucleic acid in the aforementioned methods and uses can include, but is not limited to, a conjugated antisense compound targeted to SEQ ID NO: 8 and/or 9 comprising an at least 8 consecutive nucleobase sequence of any one of SEQ ID NOs: 78-83a c_millgated antisense compound targeted to SEQ ID NO: 8 and/or 9 comprising a nueleobase sequence of anv one of SEQ 1D NO: 78-83; a compound comprising or consisting of ISIS 325568, ISIS 398471, ISIS 448766, ISIS 449884, ISIS
459014, or ISIS 459157 and a conjugate group; a compound comprising an antisense oligonucleotide disclosed in tY5 7,750,142, US

7,399,853, WO 2007/035771, or WO 2007/134014, which are incorporated by reference in their entireties herein, and a conjugate group: The nucleobase sequences of ail of the aforementioned referenced SEC) ID
NOs are incorpprated by reference herein.
4. 4. Coagulation Factor 7 The circulatory system requires mechanisms that prevent blood loss, as well as those that counteract inappropriate intravascular obstructions. Generally, coagulation comprises a cascade of reactions culminating in the conversion of soluble fibrinogen to an insoluble fibrin gel. The steps of the cascade involve the conversion of an inactive zymogen to an activated enzyme.
The active enzyme then catalyzes the next step in the cascade.
Coagulation Cascade The coagulation cascade may be initiated through two branches, the tissue factor pathway (also "extrinsic pathway"), which is the primary pathway, and the contact activation pathway (also "intrinsic pathway").
The tissue factor pathway is initiated by the cell surface receptor tissue factor (TF, also referred to as factor 111), which is expressed constitutively by extravascular cells (pericytes, cardiomyocytes, smooth muscle cells, and keratinocytes) and expressed by vascular monocytes and endothelial cells upon induction by inflammatory cytokines or endotoxin. (Drake et al., Am J Pathol 1989, 134:1087-1097). TF is the high affinity cellular receptor for coagulation factor Vila, a serine protease. In the absence of TF, Vila has very low catalytic activity, and binding to TF is necessary to render Vila functional through an allosteric mechanism. (Drake et al., Am J Pathol 1989, 134:1087-1097). The TF-VIla complex activates factor X to Xa.
Xa in turn associates with its co-factor factor Va into a prothrombinase complex which in turn activates prothrombin, (also known as factor II or factor 2) to thrombin (also known as factor Ila, or factor 2a).
Thrombin activates platelets, converts fibrinogen to fibrin and promotes fibrin cross-linking by activating factor XIII, thus forming a stable plug at sites where TF is exposed on extravascular cells. In addition, thrombin reinforces the coagulation cascade response by activating factors V
and VIII.
The contact activation pathway is triggered by activation of factor XII to Xlla. Factor Xlla converts XI
to Xla, and Xla converts IX to IXa. IXa associates with its cofactor Villa to convert X to Xa. The two pathways converge at this point as factor Xa associates factor Va to activate prothrombin (factor II) to thrombin (factor 11a).

Inhibition of coagulation At least three mechanisms keep the coagulation cascade in check, namely the action of activated protein C, antithrombin, and tissue factor pathway inhibitor. Activated protein C is a serine protease that degrades cofactors Va and VIIIa. Protein C is activated by thrombin with thrombomodulin, and requires coenzyme Protein S to function. Antithrombin is a serine protease inhibitor (serpin) that inhibits serine proteases: thrombin, Xa, XIIa, XIa and IXa. Tissue factor pathway inhibitor inhibits the action of Xa and the TF-VIIa complex. (Schwartz AL et al., Trends Cardiovasc Med. 1997; 7:234 ¨239.) Disease Thrombosis is the pathological development of blood clots, and an embolism occurs when a blood clot migrates to another part of the body and interferes with organ function.
Thromboembolism may cause conditions such as deep vein thrombosis, pulmonary embolism, myocardial infarction, and stroke.
Significantly, thromboembolism is a major cause of morbidity affecting over 2 million Americans every year.
(Adcock et al. American Journal of Clinical Pathology. 1997;108:434-49). While most cases of thrombosis are due to acquired extrinsic problems, for example, surgery, cancer, immobility, some cases are due to a genetic predisposition, for example, antiphospholipid syndrome and the autosomal dominant condition, Factor V Leiden. (Bertina RM et al. Nature 1994; 369:64-67.) Treatment The most commonly used anticoagulants, warfarin, heparin, and low molecular weight heparin (LMWH) all possess significant drawbacks.
Warfarin is typically used to treat patients suffering from atrial fibrillation. The drug interacts with vitamin K ¨dependent coagulation factors which include factors II, VII, IX and X. Anticoagulant proteins C
and S are also inhibited by warfarin. Drug therapy using warfarin is further complicated by the fact that warfarin interacts with other medications, including drugs used to treat atrial fibrillation, such as amiodarone.
Because therapy with warfarin is difficult to predict, patients must be carefully monitored in order to detect any signs of anomalous bleeding.
Heparin functions by activating antithrombin which inhibits both thrombin and factor X. (Bjork I, Lindahl U. Mol Cell Biochem. 1982 48: 161-182.) Treatment with heparin may cause an immunological reaction that makes platelets aggregate within blood vessels that can lead to thrombosis. This side effect is known as heparin-induced thrombocytopenia (HIT) and requires patient monitoring. Prolonged treatment with heparin may also lead to osteoporosis. LMWH can also inhibit Factor 2, but to a lesser degree than unfractioned heparin (UFH). LMWH has been implicated in the development of HIT.

Thus, current anticoagulant agents lack predictability and specificity and, therefore, require careful patient monitoring to prevent adverse side effects, such as bleeding complications. There are currently no anticoagulants which target only the intrinsic or extrinsic pathway.
Antisense compounds targeting Factor VII have been previously disclosed in WO
2013/119979 and WO 2009/061851, each herein incorporated by reference in its entirety.
Clinical studies are ongoing to assess the effect of antisense compounds targeting Factor VII in patients.
However, there is still a need to provide patients with additional and more potent treatment options.
Certain Conjugated Antisense Compounds Targeted to a Factor VII Nucleic Acid In certain embodiments, conjugated antisense compounds are targeted to a Factor VII nucleic acid having the sequence GENBANK Accession No. NT_027140.6 truncated from nucleotides 1255000 to 1273000, incorporated herein as SEQ ID NO: 10; GENBANK Accession No.
NM_019616.2, incorporated herein as SEQ ID NO: 11; DB184141.1, designated herein as SEQ ID NO: 12; and GENBANK Accession No.
NW_001104507.1 truncated from nucleotides 691000 to 706000, designated herein as SEQ ID NO: 13. In certain such embodiments, a conjugated antisense compound targeted to SEQ ID
NO: 11 is at least 90%, at least 95%, or 100% complementary to SEQ ID NO: 11.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
11 comprises an at least 8 consecutive nucleobase sequence of anv one of SEQ ID NOs: 84-92. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 11 comprises a nucleobase sequence of any one of SEQ ID NO: 84-92.
In certain embodiments, a conjugated antisense compo-und targeted to SEQ ID
NO: 11 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 84. In certain embodiments, a conjugated antisense compound targetgd to SEQ ID NO: 11 comprises a nucleobase sequence of SEQ ID NO: 84.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
11 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 85. ln certain embodiments a conjugated antisense compound targeted to SEQ ID NO: 11 comprises a nucleobase sequence of SEQ ID NO: 85.
In certain embodiments, a conjugated _antisense compound targeted to SEQ1D NO:
11 comprises an al least 8 consecutive nucleobase sequence of SEQ ID NO: 86. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 11 comprises a nucleobase sequence of SEQ ID NO: 86.
In certain embodiments, a conjugated antisense compound targeted to SI-:() ID
NO: 11 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: 11 conzyises a nucleobase sequence of SEQ ID N):87.

In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
11 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 88. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 11 cotnprises a nucleobase sequence of SEQ Ii) NO: 88.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
11 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 89. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 11 comprises a nucleobase sequence of SEQ ID NO: 89.
In certain embodiments, a conjuaated antisense compound targeted to SEQ 11) NO: 11 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 90. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 11 comprises a nucleobase sequence of SEQ ID NO: 90.
Table -1-: Antisense Compounds targeted to Factor VII SEQ ID NO: 11 Target Start ISIS No Sequence (5'-3') Motif SEQ ID NO
Site 15191 eeeeeddddddddddeeeee 15128 kdkdkdddddddddee 1387 eeeeeddddddddddeeeee 15192 kddddddddddkekee 15131 ekkddddddddddkke 2565; 2633;

2667 ACTCCCGGGACACCCA eekddddddddddkke In certain embodiments, a compound comprises or consists of ISIS 420915 and a conjugate group.
ISIS 407935 is a modified oligonucleotide having the formula: Aes Tes Ges mCes Aes Tds Gds Gds Tds Gds Ads Tds Gds mCds Tds Tes mCes Tes Ges Ae, wherein A = adenine T = thymine G = guanine;

mC = 5-methylcytosine; wherein each sugar moiety is indicated according to the following:
k = cEt;
d = 2'-deoxyribose;
e = 2'-M0E; wherein each internucleoside linkage is indicated according to the following:
s = phosphorothioate.
In certain embodiments, a compound comprises or consists of ISIS 304299 and a conjugate group.
ISIS 473589 is a modified oligonucleotide having the formula: Gks mCds Tks Ads Aks Ads mCds Ads Ads mCds mCds Gds mCds mCds Tes Te; wherein, each nucleobase is indicated according to the following:
A = adenine T = thymine G = guanine;
mC = 5-methylcytosine; wherein each sugar moiety is indicated according to the following:
k = cEt;
d = 2'-deoxyribose;
e = 2'-M0E; wherein each internucleoside linkage is indicated according to the following:
s = phosphorothioate.
In certain embodiments, a compound comprises or consists of ISIS 420921 and a conjugate group.
ISIS 490279is a modified oliganucleotide having the formula: mCes mCes mCes Tes mCes mCds Tds Gds Tds Gds mCds mCds Tds Gds Gds Aes Tes Ges mCes Te; wherein, each nucleobase is indicated according to the following:
A = adenine T = thymine G = guanine;

mC = 5-methylcytosine; wherein each sugar moiety is indicated according to the following:
k = cEt;
d = 2'-deoxyribose;
e = 2'-M0E; wherein each internucleoside linkage is indicated according to the following:
s = phosphorothioate.
In certain embodiments, a compound comprises or consists of ISIS 420922 and a conjugate group.
ISIS 529804 is a modified oligonucleotide having the formula: mCks Ads Tds Gds Gds Tds Gds Ads Tds Gds mCds Tks Tes mCks Tes Ge, wherein A = adenine T = thymine G = guanine;
mC = 5-methylcytosine; wherein each sugar moiety is indicated according to the following:
k = cEt;
d = 2'-deoxyribose;
e = 2'-M0E; wherein each internucleoside linkage is indicated according to the following:
s = phosphorothioate.
In certain embodiments, a compound comprises or consists of ISIS 420950 and a conjug,ate group.
ISIS 534796 is a modified oligonticleotide having the formula: Aes Gks Aks Gds mCds Tds Ads Ads Ads mCds Ads Ads mCds mCks Gks mCe, wherein A = adenine T = thymine G = guanine;
mC = 5-methylcytosine; wherein each sugar moiety is indicated according to the following:

k = cEt;
d = 2'-deoxyribose;
e = 2'-M0E; wherein each internucleoside linkage is indicated according to the following:
s = phosphorothioate.
In certain embodiments, a compound comprises or consists of ISIS 420955 and a conjugate group.
ISIS 540162 is a modified oligonucleotide having the fortnula: Ges Aes Aes Tes Ges Tds Tds Tds Tds Ads Tds Tds Gds Tds mCds Tes mCes Tes Ges inCe, wherein A = adenine T = thymine G = guanine;
mC = 5-methylcytosine; wherein each sugar moiety is indicated according to the following:
k = cEt;
d = 2'-deoxyribose;
e = 2'-M0E; wherein each internucleoside linkage is indicated according to the following:
s = phosphorothioate.
In certain embodiments, a compound comprises or consists of ISIS 420957 and a conjugate group.
ISIS 540175 is a modified oligonucleotide having the formula: Ges Ges Aks mCds Ads mCds mCds mCds Ads mCds Gds mCds mCds mCks mCks mCe; wherein, each nucleobase is indicated according to the following:
A = adenine T = thymine G = guanine;
mC = 5-methylcytosine; wherein each sugar moiety is indicated according to the following:
k = cEt;

d = 2'-deoxyribose;
e = 2'-M0E; wherein each internucleoside linkage is indicated according to the following:
s = phosphorothioate.
In certain embodiments, a compound comprises or consists of ISIS 420959 and a conjugate gro-up.
ISIS 540182 is a modified oligonucleotide having the formula: Aes mCes Aks mCds mCds mCds Tds mCds Gds mCds mCds Tds mCds mCks Gks Ge., wherein A = adenine T = thymine G = guanine;
mC = 5-methylcytosine; wherein each sugar moiety is indicated according to the following:
k = cEt;
d = 2'-deoxyribose;
e = 2'-M0E; wherein each internucleoside linkage is indicated according to the following:
s = phosphorothioate.
In certain embodiments, a compound comprises or consists of ISIS 420959 and a conjugate group.
ISIS 540191 is a modified oligonucleotide having the formula: Ges mCes mCks Tds mCds mCds Gds Gds Ads Ads mCds Ads mCds mCks mCks Ae, wherein A = adenine T = thymine G = guanine;
mC = 5-methylcytosine; wherein each sugar moiety is indicated according to the following:
k = cEt;
d = 2'-deoxyribose;
e = 2'-M0E; wherein each internucleoside linkage is indicated according to the following:
s = phosphorothioate.
In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in WO
2013/119979 and WO 2009/061851, which are incorporated by reference in their entireties herein. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 21-659 disclosed in WO 2013/119979 and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 4-159 and 168-611 disclosed in WO 2009/061851 and a conjugate group described herein. The nucleobase sequences of all of the aforementioned referenced SEQ ID NOs are incorporated by reference herein.
Factor VII Therapeutic Indications In certain embodiments, provided herein are compounds and compositions as described herein for use in therapy.
In certain embodiments, provided herein are conjugated antisense compounds and compositions as described herein for use in treating, preventing, or slowing progression of a thromboembolic complication.
In certain embodiments, provided herein are conjugated antisense compounds and compositions as described herein for use in treating, preventing, or slowing progression of a hyperproliferative disorder.
In certain embodiments, provided herein are conjugated antisense compounds and compositions as described herein for use in treating, preventing, or slowing progression of an inflammatory condition.
In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to a Factor VII nucleic acid for modulating the expression of Factor VII in a subject. In certain embodiments, the expression of Factor VII is reduced.
In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to a Factor VII nucleic acid in a pharmaceutical composition for treating a subject. In certain embodiments, the subject has a Factor VII related disease, disorder or condition, or symptom thereof In certain embodiments, the Factor VII related disease, disorder or condition is a thromboembolic complication, a hyperproliferative disorder or an inflammatory condition.
-------- In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to a Factor VII nucleic acid in the preparation of a medicament.

In certain embodiments, the invention provides a conjugated antisense compound targeted to a Factor VII nucleic acid, or a ph.armaceutically acceptable salt thereof for u.se in therapy.
Certain embodiments provide a conjugated antisense compound targeted to a Factor VII nucleic acid for use in the treatment of a Factor VII related disease, disorder or condition, or symptom thereof. In certain embodiments, the Factor VII related disease, disorder or condition is a thromboembolic complication, a hyperproliferative disorder or an inflammatory condition.
In certain embodiments, provided herein are conjugated antisense compounds and compositions as described herein for use in treating, preventing, or slowing progression of a thromboembolic complication.
In certain embodiments, provided herein are conjugated antisense compounds and compositions as described herein for use in treating, preventing, or slowing progression of a hyperproliferative disorder.
In certain embodiments, provided herein are conjugated antisense compounds and compositions as described herein for use in treating, preventing, or slowing progression of an inflammatory condition.
It will. be understood that any of the compounds described herein can be used in the aforementioned methods and uses. For example, in certain embodiments a conjugated antisense compound targeted -to a Factor -VII nucleic acid in the aforementioned methods and uses can include, but is -not limited to, a conjugated antisense compound targeted to SEQ ID NO: 11 comprising an at least 8 consecutive nucleobase sequence of any one of SEQ -ID NOs: 84-92La conkgated antisense compound targeted to SEQ -ID NO: 11 comprising a nucleobase sequence of any one of SEQ ID NO: 84-92; a compound comprising or consisting of ISIS 407935, ISIS 473589, ISIS 490279, ISIS 529804, ISIS 534796, ISIS
540162, ISIS 540175, ISIS 540182, or ISIS 540191 and a conjugate group; a compound comprising an antisense oligonucleotide disclosed in WO
2013/119979 and WO 2009/061851,.. which are incorporated b:y: reference in their entireties herein. and a conjugate group; a cotnpound comprising an antisense olig.onucleotide having a nucleobase sequence of any of SEQ ID NOs 21-659 disclosed in WO 2013/119979 and a conjugate group described herein; an antisense oligon-ucleotide having a nucleobase sequence of any of SEQ ID NOs 4-159 and 168-611 disclosed in WO
2009/061851 and a conjugate group described herein. The nucleobase sequences of all of the aforementioned referenced SE() -ID NOs are incorporated by reference herein.
4. Coagulation Factor 11 The circulatory system requires mechanisms that prevent blood loss, as well as those that counteract inappropriate intravascular obstructions. Generally, coagulation comprises a cascade of reactions culminating in the conversion of soluble fibrinogen to an insoluble fibrin gel. The steps of the cascade involve the conversion of an inactive zymogen to an activated enzyme.
The active enzyme then catalyzes the next step in the cascade.
Coagulation Cascade The coagulation cascade may be initiated through two branches, the tissue factor pathway (also "extrinsic pathway"), which is the primary pathway, and the contact activation pathway (also "intrinsic pathway").
The tissue factor pathway is initiated by the cell surface receptor tissue factor (TF, also referred to as factor 111), which is expressed constitutively by extravascular cells (pericytes, cardiomyocytes, smooth muscle cells, and keratinocytes) and expressed by vascular monocytes and endothelial cells upon induction by inflammatory cytokines or endotoxin. (Drake et al., Am J Pathol 1989, 134:1087-1097). TF is the high affinity cellular receptor for coagulation factor Vila, a serine protease. In the absence of TF, Vila has very low catalytic activity, and binding to TF is necessary to render Vila functional through an allosteric mechanism. (Drake et al., Am J Pathol 1989, 134:1087-1097). The TF-VIla complex activates factor X to Xa.
Xa in turn associates with its co-factor factor Va into a prothrombinase complex which in turn activates prothrombin, (also known as factor II or factor 2) to thrombin (also known as factor Ila, or factor 2a).
Thrombin activates platelets, converts fibrinogen to fibrin and promotes fibrin cross-linking by activating factor XIII, thus forming a stable plug at sites where TF is exposed on extravascular cells. In addition, thrombin reinforces the coagulation cascade response by activating factors V
and VIII.
The contact activation pathway is triggered by activation of factor XII to Xlla. Factor Xlla converts XI
to Xla, and Xla converts IX to IXa. IXa associates with its cofactor Villa to convert X to Xa. The two pathways converge at this point as factor Xa associates factor Va to activate prothrombin (factor II) to thrombin (factor 11a).
Inhibition of coagulation.
At least three mechanisms keep the coagulation cascade in check, namely the action of activated protein C, antithrombin, and tissue factor pathway inhibitor. Activated protein C is a serine protease that degrades cofactors Va and Villa. Protein C is activated by thrombin with thrombomodulin, and requires coenzyme Protein S to function. Antithrombin is a serine protease inhibitor (serpin) that inhibits serine proteases: thrombin, Xa, Xlla, Xla and IXa. Tissue factor pathway inhibitor inhibits the action of Xa and the TF-VIla complex. (Schwartz AL et al., Trends Cardiovasc Med. 1997; 7:234 ¨239.) Disease Thrombosis is the pathological development of blood clots, and an embolism occurs when a blood clot migrates to another part of the body and interferes with organ function.
Thromboembolism may cause conditions such as deep vein thrombosis, pulmonary embolism, myocardial infarction, and stroke.
Significantly, thromboembolism is a major cause of morbidity affecting over 2 million Americans every year.
(Adcock et al. American Journal of Clinical Pathology. 1997;108:434-49). While most cases of thrombosis are due to acquired extrinsic problems, for example, surgery, cancer, immobility, some cases are due to a genetic predisposition, for example, antiphospholipid syndrome and the autosomal dominant condition, Factor V Leiden. (Bertina RM et al. Nature 1994; 369:64-67.) Treatment.
The most commonly used anticoagulants, warfarin, heparin, and low molecular weight heparin (LMWH) all possess significant drawbacks.
Warfarin is typically used to treat patients suffering from atrial fibrillation. The drug interacts with vitamin K ¨dependent coagulation factors which include factors II, VII, IX and X. Anticoagulant proteins C
and S are also inhibited by warfarin. Drug therapy using warfarin is further complicated by the fact that warfarin interacts with other medications, including drugs used to treat atrial fibrillation, such as amiodarone. Because therapy with warfarin is difficult to predict, patients must be carefully monitored in order to detect any signs of anomalous bleeding.
Heparin functions by activating antithrombin which inhibits both thrombin and factor X. (Bjork I, Lindahl U. Mol Cell Biochem. 1982 48: 161-182.) Treatment with heparin may cause an immunological reaction that makes platelets aggregate within blood vessels that can lead to thrombosis. This side effect is known as heparin-induced thrombocytopenia (HIT) and requires patient monitoring. Prolonged treatment with heparin may also lead to osteoporosis. LMWH can also inhibit Factor 2, but to a lesser degree than unfractioned heparin (UFH). LMWH has been implicated in the development of HIT.
Thus, current anticoagulant agents lack predictability and specificity and, therefore, require careful patient monitoring to prevent adverse side effects, such as bleeding complications. There are currently no anticoagulants which target only the intrinsic or extrinsic pathway.
Antisense compounds targeting Factor XI have been previously disclosed in WO
2010/045509 and WO 2010/121074, each herein incorporated by reference in its entirety.
Clinical studies are ongoing to assess the effect of antisense compounds targeting Factor XI in patients.
However, there is still a need to provide patients with additional and more potent treatment options.
Certain Conjugated Antisense Compounds Targeted to a Factor XI Nucleic Acid In certain embodiments, conjugated antisense compounds are targeted to a Factor XI nucleic acid having the sequence GENBANK Accession No. NM_000128.3 (incorporated herein as SEQ ID NO: 14), GENBANK Accession No. NT_022792.17, truncated from 19598000 to 19624000, (incorporated herein as SEQ ID NO: 15), GENBANK Accession No. NM_028066.1 (incorporated herein as SEQ
ID NO: 16), exons 1-15 GENBANK Accession No. NW_001118167.1 (incorporated herein as SEQ ID NO: 17).
In certain such embodiments, a conjugated antisense compound targeted to SEQ ID NO: 2 is at least 90%, at least 95%, or 100% complementary to SEQ ID NO: 2.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
2 comprises an at least 8 consecutive nucleobase sequence of any one of SEQ ID NOs: 12-19. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 2 comprises a nucleobase sequence of any.one of SEQ ID NO: 12-19.
In certain embodiments, a conjugated _antisense compound targeted to SEQ1D NO:
14 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 93. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 14 comprises a nucleobase sequence of SEQ ID NO: 93.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
14 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 94. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 14 conzyises a nucleobase sequence of SEQ ID NO: 94.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
14 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 95. In certain embodiments, a conjugated antisense compound -targeted to SEQ :ID NO: 14 comprises a nucleobase sequence of SEQ ID NO: 95.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
14 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 96. In certain embodiments, a conjugated.
antisense compound targeted to SEQ ID NO: 14 comprises a nucleobase sequence of SEQ ID NO: 96.
In certain embodiments, a conjugated antisense compound -targete(1 -to SEQ ID
NC): 14 comprises an at least 8 consecutive nucleobase sequence of SEQ :ID NO: 97. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 14 comprises a nucleobase sequence of SEQ ID NO: 97.
In certain embodiments, a conjugated antisense compo-und targeted to SEQ ID
NO: 14 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NC): 98. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 14 comprises a nucleobase sequence of SEQ Ii) NO: 98.

In certain embodiments, a conjutc,fated antisense compound targeted to SEQ ID
NO: 14 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 99. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 14 cotnprises a nucleobase sequence of SEQ Ii) NO: 99.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
14 comprises an at least 8 consecutive aucleobase sequence of SEQ ID NO: 100. in certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 14 comprises a nucleobase sequence of SEQ ID NO: 100.
Table ;157: Antisense Compounds targeted to Factor 11 SEQ ID NO: 14-17 Target Start ISIS No Sequence (5'-3') Motif SEQ ID NO
Site In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in WO
2010/045509 and WO 2010/121074, which are incoworated b:y: reference in their entireties herein. and a conjugate group. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 15-270 disclosed in WO 2010/045509 and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide having a nucleobase sequence of auy of SEQ ID NOs 15-270 disclosed in WO 2010/121074 and a conlIgate group described herein, . The nucleobase sequences of all of -the aforetnentioned referenced SEQ ID NOs are incorporated by reference herein.

E. 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 "ATmeCGAUCG," wherein meC 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 DMTOBX DMTO Bx DMTOÇBX
00Me o NC NC mipo2 NC
1 la 2 Bx is a heterocyclic base;
Compounds 1, 1a 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 AcO0Ac Ac0 OAc0 O Ac0 HOCO * 5 TMSOTf, 50 C
l 0 ________________________ AcHN CICH2CH2CI N TMSOTf, DCE
3 (93%) 4 ( 66%) Ac0 OAc AcO0Ac 0 0 H2/Pd 1.r0H
-1C) Me0H ___ Ac0 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 Et0y,, NCr----1 0 0...,, HOõ 0, Et0 CN 9 HCI, Et0H ----NH2 H0,-----NH2 1m- NC--.N.--- =õ..õ----NH2 Is-aq. KOH, Reflux, rt, 0 Et0 0---HO-- 1,4-dioxane, (:) (56%) 8 (40%) NC) 10 d------) 11 õ, Compounds 8 and 9 are commercially available.
Example 4: Preparation of Compound 18 ),....___Et0r4)...____ N 00 io Et0 0 oõ benzylchloroformate, Et0 Et0 --I( Li0H, H20 Dioxane, Na2CO3 )------0-,...---NH2 ________________ )...- H Dioxane __ 0.
(86%) Et 0 0--- Et0 0--(91%) ii 12 NH
' 0 )n _....v 9 H 0 9_ 9 HO
0 0, 9 -0 N1 "-'"--NH2 14 __\/-nr __, H N.....7"\----N 0 HO').----N.--0õ......--N--14"=0 401 _______________ 0 0 0--H
HBTU, DIEA, DMF
0 HO 0-- (69%) 0 +O 15 r,LNN--e 0"--------rj 13 H H ¨
AcO0Ac H
H2N ACOrs\r OrOH

H 0 0, 1 AcHN 0 ---r-N.,0õ.õ. N 0 io HBTU, DIEA, HOBt _____________ 1.. H
0.
95% 0 0' DMF

16 (64%) V\N_____ AcO0Ac Ac0 r OrN1,.õN,0 AcHN 0 AcO0Ac 0 Ac0_....r.Ø.\r Or r0,, N.-J(0 io H
AcHN 0 0 0' AcO0AcHN-----kj Ac0_4r0-------r-N

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.
Med. Chem., 2004, 47, 5798-5808.
Example 5: Preparation of Compound 23 1.

0-------/ ¨b 1. TBDMSCI H N H3C0)tkOH
HBTU, DIEA
N DMF, Imidazode, rt (95 %) TBDMS0 ) DMF, rt (65%) HO--%==== ) _________________________________________________________________ 1.-2. Pd/C, H2, Me0H, rt _ 2.
TEA.3HF, TEA, THF
87% 20 a TBDMS (72%) 19 'OH

1. DMTCI, pyr, rt (75%) OH
OCH _______________________________________ -b1)11q8 3 )1-2. Li0H, Dioxane (97%) 23 Compounds 19 and 21 are commercially available.

Example 6: Preparation of Compound 24 AcO0Ac H H
AcO__....ZrOr N ,..-N,10 AcHN 0 1. H2, Pd/C, Me0H (93%) AcO0Ac 0 H 0, 2. HBTU, DIEA, DMF
(76%) Ac0 ____________________________________________ 0 ,¨ODMT
H .
AcHN 0 0 0' HOJW"N :
N

AcO0Ac H HN----kj Ac0)._\vo---------(N---.7----j AcHN
AcO0Ac H H
__,......(2.\v Ac0 AcHN 0 ODMT
AcO0Ac HH 0õ 0 0 µ
__......(.2..\, Ac0 C)N N7.---N ---n---N.,-0, ___ N "iLqL N
H µ
AcHN 0 0 0' OH
AcO0Ac HH N -----j --_---fr Ac0 0 24 Arl-IN
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..\vOr.N ,..,..N,0 AcHN 0 AcO0Ac ODMT
_....2.._\.r H H 0, 0 0 µ
Ac0 Or NN7N___N y_i_..Ø....___ 1. Succinic anhydride, DMAP, DCE
AcHN 0 0 0' OH 2. DMF, HBTU, EtN(iPr)2, PS-SS
AcO0Ac HN-----H ...../.____/
__,õ,,Zr0----"---rN
Ac0 0 AcHN
AcO0Ac H H
Ac0 Or N,..õ.N ,.(:) AcHN 0 AcO0Ac ODMT

Ac0 N ----1\1"---11-----N---0------ hi --1L('e NQ ,¨NH
AcHN 0 0 0' 04 AcO0Ac H N ------j H___/....__/
Ac0 0------ 0 AcHN
Compound 24 was prepared as per the procedures illustrated in Example 6.

Example 8: Preparation of Compound 26 AcO0Ac H H
Ac0.....72._\,OrNõN,0 AcHN 0 AcO0Ac ODMT
H H 0-, 0 0 Phosphitylation H - µ
AcHN 0 0 0' OH
AcO0AcH HN----j ___/,./
AcOres\,0----_----irN 0 AcHN
AcO0Ac H H
...N ,CD
Ac0 OrN
AcHN 0 AcO0Ac ODMT
/
H H 0, u 0 9 _ Ac0- NN7N---"N O----- NI--Nr H µ
AcHN 0 0 0' HN----- 0 I
NC.õ----Ø-P-N up 0 2 -j AcO0Ac H 0 Ac0o.______N---.7---j 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---7(--)-\r ..(N`.----N`(:) AcHN 0 AcO0Ac ODMT
0 0 µ __ Ac0 ,¨NH
H µ
AcHN 0 0 0 04 HN 1. DCA, DCM
¨Cj AcO0Ac H 0 2. DCI, NMI, ACN
N--,7---/
0----.Thr Phosphoramidite DNA/RNA ' Ac0 building block 1 µautomated synthesizer , AcHN 3. Capping 4. t-BuO0H 0 Bx _ DMTC"c AcO0Ac __..72...\r H H
CN r.N N
,-,C) 0 Ac0 0 0=P-0 AcHN 0 O
AcO0Ac H H 0, -...-C.-c; 0 o Ac0----"'"--N, NNVN----N----if---N.--- 0 õ..õ--- N -11.141"N )\¨NH
H µ
AcHN 0 1. DCA, DCM 0 2. DCI, NMI, ACN
AcO0Ac H HN 0 Phosphoramidite DNA/RNA ' Ac0 0--building block la a..utomated synthesizer , 3. Capping 0 27 4. t-BuO0H
AcHN
DMTO-N(yx 6.. _______________________________________________________ b_/-0 M e op' _0CN
\
0-Nn,Bx AcO0Ac S.-orNHN1H,.0 0 I
Ac0 0=P-0-AcHN 0 O
AcO0Ac __r2s\ro,,,yNN7N.,__N 0 Ac0 ---r..--0-õ....--- N)11--el"N"
)¨NH
H \
AcHN 0 0 0' 04 1. DCA, DCM
AcO0Ac H HN-----µj 0 2. DCI, NIVII, ACN
_...r...:1\r0-----ir N----/----/ Phosphoramidite DNA/RNA
Ac0 building blocks automated synthesize AcHN 3. Capping 4. xanthane hydride or t-BuO0H
5. Et3N/CH3CN (1:1) 6. Aaueous NFL (cleavaue) OH
I s s. OLIGO , I

X=P\-0-0-N0,13x Bx = Heterocyclic base d b-f/-OMe x = 0 or S I
0=P-0-\
0-Na,Bx HOOH

HO-, i-N-'"N`(:) 0=P-0-AcHN 0 I

0 0 I, H \
AcHN 0 0 0' OH
HOOH HN-----Cj C)--w--Ir--N
HO

AcHN
Wherein the protected Ga1NAc3-1 has the structure:

¨P-O-N(oyN õ..,./
1 , N
-HOOH ' !.....:),..\ O H H
f,...õ, 0-I
HO__, N N0 O=P-O-AcHN 0 I

H H 0, 0 0 N.....õ--N __ 0...õ...- N-Iti-eNrsZ
H \
AcHN 0 0 0' OH
HOOH H HN------HO 0Tf.)....\,o--___----(N.-7.---/

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

r-"N"N`O
AcHN 0 I

H H 0, 0 0 ___Tes\,0 N
HO .õ,...õ..-õ,..õ..,...- ...õ7.......õ-N
(:)......õ- N-11148.1LN"
H \
AcHN 0 0 0' OH
HOOHHN-----H 0______/
H 0 0 --------r N

AcH N
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
_____________________________ .., I
0 3. DCA, DCM

-UNL-ODMT 2. DCI, NMI, ACN 0 ...
I 0, 4. DCI, NMI, ACN
30 Phosphoramidite 0-UNL-0-P-Pk-'1N Phosphoramidite 1 building blocks , DNA/RNA DNA/RNA
31 ,automated synthesizer, ,.automated synthesizer, DMT0c (5".Bx 1. Capping (Ac20, NMI, PYr) 2. t-BuO0H 0 3. DCA, DCM NC ' 0-p ... ____________________ 4. DCI, NMI, ACN 0 I
Phosphoramidite 26 COLIGO) ' DNA/RNA ' I
X = 0, or S automated synthesizer 0, I
Bx = Heterocylic base 0-UNL-0-P-0CN
X

AcO0Ac ___..C2 Ac0 s\z H H
AcHN 0 Ac0 OAc OD MT
H H 0, 0 0 Z
01\I N 0 ..........,_ __ N .--J-Lq-L N
Ac0 H \
AcHN 0 0 C) 0 I
HN NCcy P,0,...(or Bx ----Cj Ac0 OAc H , __.......2 0 .\,0----_/\/"----rr N---7------/
NC 0 -P =0 Ac0 0 0 AcHN I
(OLIGO) I
1. Capping (Ac20, NMI, PYr) 0 2. t-BuO0H I
0_u 3. Et3N:CH3CN (1:1 \Iv) NL_ 0 _ p..._0CN
X
4. DCA, DCM
5. NH4, rt (cleavage) 33 HOOH
H H

rN----"N`(:) AcHN 0 HOOH OH
H H 0, 0 0 HO__.,o...,..,.,-...,...õ_-....r.NN.zz.-N__,N.,rN__o.....__õ NA.(411..Nr H 8 \
AcHN 0 0 0' CI) -0--z--p,_OBx H
HOOH HN-- 0 --kj O r z u ______/
0.
HO0---_----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. HON)L0. AcO0Ac Ac0_...\
35 TMSOTf, DCE
N--__H----- 2. H2/Pd, Me0H AcHN 36 Ac0 OAc HBTU, DMF, EtN(iP02 o Ac0 1. H2, Pd/C, Me0H
. ____FN1 Compound 13 AcHN 8 2. HBTU, DIEA, DMF
Ac0 OAc H 0 0 Compound Ac0 .......(..:)...\z0N F

NHAc 0 0 0 OAc ,---) Ac0 0 Ac0.....2...\yoNH
*

AcHN

Ac0 OAc \
Ac0.\,,,) 0 /0DMT ,,ENI =
Phosphitylation _ AcHN 8 0 OAc0\ \¨N
Ac0 OH
Ac0 8 NHAc 0 0 0 OAc )\---) Ac0 NH
AcHN
Ac0 OAc Ac0.\,,,D /ODMT

,,EI
AcHN 8 N 0 -OAc 0\ N
Ac0 H 0 .,:::._ > __ 8 AcO -.'-71. Y---.0 NH 1 P
NHAc 0 0 0 NC (D' N UP 02 OAc )\--j Ac0 Ac01"?....-VC)NH 39 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 E /0DMT I =
_ AcHN 8 0 OAc 0\ -Na H 0 O,._ > __ 8 OH
Ac00.2....\/0....,...visr.,..,,,N\ir.,,-0 NH
Ac0 8 NHAc 0 0 0 1. Succinic anhydride, DMAP, DCE
OAc Ac0 NH 2. DMF, HBTU, EtN(iPr)2, PS-SS
AcOriC2.-VC) AcHN
Ac0 OAc Ac0...r!.:).\õ, /ODMT

_,EN1 AcHN 8 Y 0 _ 0, kig 0 ott OAc Ac0 H
N 0 .,0,._NEi 0---C}-11 AcO*12-V Y---.(:) NHAc 0 0 0 ) OAc \---) Ac0 40 Ac072..-V NH
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 HOOõ )\-0 11 -N

HO 41\ /-0".

Ac0 OAc Ac0,0 AcHN XrX)1\1 0 0-}_ki 1. H2, Pd/C, Me0H
0 ______________________________________________________________ 0-0 2. HBTU, DIEA, DMF
Compound 23 OAc (:) AcOric?....v Ac0 0.,,.,,..--1..r....,...,..NH

AcHN
Ac0 OAc Ac0,\,,,) 0 ODMT
_ N =
AcHN Phosphitylation )0 H

OAc Ac0...70....\, )l-----j..)H
Ac0 0 N

AcHN
Ac0 OAc Ac0...\,) õ.0 ODMT
x_R.......x___H
N =
-AcHN

IN 8 ?
OAc H NC0,PN UP 02 0\\ j Ac0...... 44 C) J.-.NH
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-c) ODMT
H
N
AcHN

H

OAc Ac0 Ac0 .07ZzONH 1. Succinic anhydride, DMAP, DCE

AcHN 2. DMF, HBTU, EtN(iPr)2, PS-SS
Ac0 OAc Ac00 ODMT
H
N4-)N
AcHN

IN) ( 8 C)NElla 0\\ ) OAc )1---- 45 Ac0 Ac007,ZzONH

AcHN
Compound 43 is prepared as per the procedures illustrated in Example 13.
Example 15: Preparation of Compound 47 HO 0 ill DMTO
bl 1. DMTCI, pyr -b1H
_____________________________________ x-2. Pd/C, H2, Me0H
Hd 46 Hd 47 Compound 46 is commercially available.

Example 16: Preparation of Compound 53 HBTU, EtNUP02, DMF 0 _____________________________________ 0.-H3C0 Boc -"----(cIZI F\11Eioc 0 HN v NH
\CBz 50 CBz NH
HN,CBz 0)----.

H300 NCBz 1. TFA Y{4 ;'N NH H 1. Li0H, Me0H
_________________ 0- 0 H _________________________________________ .
2. HBTU, EtNUP02, DMF 2. HBTU, EtNUP02, DMF

HNrCBz Compound 47 HN,CBz CBz OH

DMTO HN-CBz 1. H2, Pd/C
0.77L-I,7,,, ___________________________________________________ =
2. HBTU, EtN(iP02, DMF
HO' N- C ,CBz NH N Compound 17 I" H
0 ' H

HN-CBz OAc OAc_____.\.____ 0 Ac0 0, ,l______.
\ __ 0 NH
NHAc c OAc 0 OAc..____\....___\_ 0 H__õ,),..-----.., 00H
HN------N "
Ac0 __ 0 0 HN--..../Th 7 NHAc 0 r OAc2C)Ac 0 ODMT
7)1 NH 53 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 Ac0......\_....\_ 0 0 ,1______ NHAc OAc OAc_...\_____\ 0 ,.....m. ,,..)L 7"------.=,10H
Ac0 0 N

NHAc 0 r OAcOAc Ac0 NHAc Phosphitylation y OAc OAc O
Ac0 0 ,.[____ NHAc UP 02N, .._ ,_, OAc 0 P `-'\______\
OAc......v...\_CN
0 .._,.....,./0,_}...., 7------..µ%6 0 ,)..
0 N H HN-----N/" 7 N
Ac0 NHAc 0 r OAc' OAc Ac0 0 I NH 54 NHAc Compound 53 is prepared as per the procedures illustrated in Example 16.

Example 18: Preparation of Compound 55 OAc OAc.L_11 Ac0 0 NHAc OAc 0 OAc.L_ 0 Ac0 0 NHAc 0 OAc ODMT
I __ NH 53 Ac0 0 NHAc 1. Succinic anhydride, DMAP, DCE
2. DMF, HBTU, EtN(iPr)2, PS-SS

OAc OAcc_ 0 NHAc OAc O

o Np HN
NHAc 0 ODMT
.7.)1 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, = 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 4-7-'4, 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 4:4, 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 474 Modified ASO targeting ApoC III and SRB-1 SEQ
CalCd Observed ASO Sequence (5 to 3') Target ID
Mass Mass No.
ISIS

AesGesniCesTesTesmCd Td Td Gd Td mCd mCd Ad Gd mCd TesTesTesAesTe ANC 7165 .4 7164.4 20 ISIS AesGesmCesTesTesmCdsTdsTdsGdsTdsmCdsmCdsAdsGdsmCdsTesTesTesAesTeoAdo,-ApoC
9239.5 9237.8 21 647535 GaINAc3-1. 111 ISIS AesGeomCeo Teo TeomCdsTdsTdsGdsTdsmCdsmCdsAdsGdsmCdsTe. Teo TesAesTeoAdo, - ApoC
9142.9 9140.8 21 647536 GaINAc3-1. 111 ISISSRB-TksmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTksmCk 4647.0 4646.4 22 ISIS

TksmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTksmCkoAdo,-GaINAC3-1a 6721.1 6719.4 23 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 "Ga1NAc3-19." 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 -P-;5, 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 Vi5 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 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 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 43,1-6 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 446 Effect of ASO treatment on ApoC III plasma protein levels in human ApoC III
transgenic mice Dose ED50Internucleoside SEQ ID
ASO 3' Conjugate (jlmol/kg) PBS (jnnol/kg) Linkage/Length No.

ISIS 0.08 86 0.73 None PS/20 20 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 21 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 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 247 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 20 304801 2.25 21 6.75 12 0.08 65 ISIS 0.75 9 0.13 Ga1NAc3-1 PS/20 21 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 and ;49. 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.

1 Table 24-8 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 20 304801 2.25 110 6.75 82 0.08 230 ISIS 0.75 82 647535 2.25 86 Ga1NAc3-1 PS/20 21 6.75 99 1 Table 219 Effect of ASO treatment on HDL and LDL cholesterol levels in transgenic mice ASO Dose HDL LDL 3' Internucleoside SEQ
(jlmol/kg) (mg/dL) (mg/dL) Conjugate Linkage/Length ID No.

0.08 17 23 ISIS 0.75 27 12 None PS/20 304801 2.25 50 4 6.75 45 2 0.08 21 21 ISIS 0.75 44 2 GaINAc3-1 PS/20 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 (lg/g) of 1 full-length ISIS 304801 and 647535 was measured and the results are provided in Table -2--.¶0. 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 1 the liver. Indeed, the calculated EC50 (provided in Table 20) 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 ;IMO
PK analysis of ASO treatment in transgenic mice Internucleoside Dose Liver Kidney Liver EC50 3'SEQ
ASO Linkage/Length (jlmol/kg) (m/g) (pg/g) (11,g/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 ;:.'-4a10a. 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 Dal0a 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
6 [3 GalNAc + 3 5-hydroxy-pentanoic acid tether] 9.8 Cleavage Sites () Cleavage site A
i HO 01-I Cleavage site C O=P-OH

Cleavage site D 8 N OH
\ HN (),,,I#54 Njsi HO 0 \ 0 \-----_ \ _____________________________________________________________________ /
NHAc 0 HO OH 0 b ---__ N 1 0 Cleavage site C 0 ¨ Cleavage site 8 H
HO \-0 \ }o ________________________________________ I
N ,--- 0 P=0 NHAc 0 Cleavage site D 0 OH
HO HN

0 \ N
HO-7.1"\-7\ 0 Cleavage site D
NHAc Cleavage site C

() 0=P-OH NH2 Metabolite 1 Metabolite 2ts1_,I.--1,,,,N
L.,,O.,7=N N-, OH

O
0=P-OH NH2 O
H 0rµNx-L, N
OH
1 N.,-J

0 0 d -_ N __ H H
N N
H o __ 17=o 0"--2 Metabolite 3 0=P-OH NH2 H 0z,Nx.-lk, N
H2N --__ _ OH
0---__ 0 N
H H d N N
o _______________________________________________________________ 17=o 0"----Metabolite 4 N

0=P-OH NH2 O Nx-1,..,õ

OH
(3N N,,_]
O___ 0 d N
H
0 _______________________________________________________________ P=0 H 1 Metabolite 5 0=P-OH

O

OH i N
14,..1 _____________________________________________________________________ /
0---___ 0 N _________________________________________________________________ C;
H
H2N N 0 __ l'=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 -P4, 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 -41 1-2-15, 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 :1411 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 20 ISIS 0.3 98 1.9 Ga1NAc3-1 PS/20 21 0.3 103 1.7 Ga1NAc3-1 PS/PO/20 21 1 Table 2-S12 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 20 0.3 98 2.1 Ga1NAc3-1 PS/20 21 0.3 103 1.8 Ga1NAc3-1 PS/PO/20 21 5 1 Table 103 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 20 0.3 100 2.2 Ga1NAc3-1 PS/20 21 ISIS 0.3 95 1.9 Ga1NAc3-1 PS/PO/20 21 1 Table 2!:
Effect of ASO treatment on total cholesterol levels in transgenic mice Dose Internucleoside ASO % PBS 3' Conjugate SEQ ID No.
(mg/kg) Linkage/Length None PS/20 20 0.3 93 GaINAc3-1 PS/20 21 0.3 115 GaINAc3-1 PS/PO/20 21 5 1 Table a815 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 20 0.3 98 86 GaINAc3-1 PS/20 21 0.3 143 89 647536 3 213 33 GaINAc3-1 PS/PO/20 21 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 -3,74, 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 ;s,z16, 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 1416 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
651900 0.7 20 0.3 Ga1NAc3-1 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 ,z-41- 17 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 3-0I 7 Modified ASOs ASO Sequence (5' to 3') Target SEQ ID
No.
ISIS GesmCesTesGesAesTdsTdsAdsGdsAdsGds 104838 AdsGdsAdsGdsGesTesmCesmCesmCe TNFa 24 ISIS TesmCesmCesmCdsAdsTdsrrdsTdsmCdsAdsGds CRP

353512 GdsAdsGdsAdsmCdsmCdsTesGesGe ISIS ikesGesmCesTesTesmCdsTdsTdsGdsTds Ap OC III

304801 mCdsmCdsAdsGdsmCds TesTesTesAesTe ISIS ikesGesmCesTesTesmCdsTdsTdsGdsTds ApoC III

647535 mCdsmCdsAdsGdsmCdsTesTesTesAesTeoAdo,-Ga1NAC3-la ISIS ikesGeomCeoTeoTeomCdsTdsTdsGdsTds ApoC III

616468 mCdsmCdsAdsGasmCdsTeoTeoTesAesTe 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 -1-18,1 Proinflammatory Effect of ASOs targeting ApoC III in hPBMC assay ASO
EC50 Emax E /EC 3' Internucleoside SEQ ID
(IIM) (IIM) max 50 Conjugate Linkage/Length No.

0.01 265.9 26,590 None (high responder) ISIS 304801 0.07 106.55 1,522 None PS/20 20 ISIS 647535 0.12 138 1,150 Ga1NAc3-1 PS/20 21 ISIS 616468 0.32 71.52 224 None PS/PO/20 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 ICso was calculated using the standard methods and the results are presented in Table ;4219. As illustrated, comparable potency was observed in cells treated with ISIS 647535 as compared to the control, ISIS 304801.
Table A2,19 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 20 ISIS
0.31 Ga1NAc3-1 PS/20 21 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 ;4-310, 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 3320 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 20 for 2 wks ISIS
mg/kg/wk 40 None 14 PS/6 PO 20 for 2 wks Example 27: Compound 56 N(iPr)2 DMTO
DMTO
7--........7--...

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 .1:2\
HOõ..-.........õ....-........0Bn 57 0 TMSOTf, DCE _....r......\,0 0 H2/Pd Ac0_. ..\
____________________________________________ Ac0 OBn Me0H
N---------1 AcHN 58 (quant.) ( 71%) CNEtO(N(iPr)2)PC1, Ac0 OAc Ac0 OAc N(iPr)2 ___.....L.).\,0 Ac0 OH Ac0 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 ._,.. OH 1. DMTC1, pyr H
,...0DMT

HO 2. KOH, DMSO 2. Pd/C, H2 __ 0 õ
\ ________ 03 __________ Me0H 3. Phosphitylation Bn0 .---------OH ).- p ----------ODMT
3. HC1, 0 0E-I kop02 ---ODMT
4. NaHCO 3 '--.

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
rj CN
rj ,...0,..-0 1. H
TPDBSOõ.........õ-----..u..---....õ,,OH DMTC1, pyr 2. TBAF 0, .....õ....--ODMT
___________________________________________ ).- P
---0 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 0, 1. DMTC1, pyr HO 0 \.--------0Bn 2. Pd/C, H2 --- 3. Phosphitylation / 63c 63d HO ¨/ DMTO ¨/
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 H2NOTBDMS Ac0 OAc 0 CO2Bn _....1....\i31 ).( Ac0 ,0 OH R _.,..,......\i) Ac0 .roAN OTBDMS
, AcHN 64 HBTU, DIEA AcHN 66 H
R
R = H or CH3 Ac0 OAc 1. TEA.3HF, THF 0 CO2Bn 2. Phosphitylation H I CN
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, 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 IIHTU, D1EA
Ac0 OH ________________ _....rØ4, ).L
OHn N
1.-- Ac0 0 AcHN 64 MT
AcHN 69 H

Ac0 OAc 1. Pd/C, H2 0 _____________________ r .......2..\0., Ac0 ___ , 2. Phosphitylation H I I
AcHN CH3 N(iP02 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 FIN
--..... N......-- I
NC 0 3. CF3CO2Et, Me0H H
N(iPr)2P CN
NC ---"\---- -,,/-0H ____________________ " _________________ F3 C /1\T-----------"" 0 0 NC o 4. TEA.314F, THF 1 5. Phosphitylation 0 HT
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 HO,..õ.....õ....---..õ...Ø....._ -...,,...õ.---..õ.._õ0, DCI, NMI, ACN
1. BnCl, NaH HO
Phosphoramidite 60 DMTO
_____________________________________________________________________________ ).-/..-.,..õ7- HO
-..Ø--- 2. DCA, CH2C12 õ..-....,,7--.Ø---Ac0 OAc NC --.1 __......7.2..\
Ac0 0 0 \------\,---____. p AcHN
NC
1. H2/Pd, Me0H
---__\
Ac0 OAc ' L
0,, O 2. Phosphitylation Ac0 Li ,P,, OBn AcHN (:) NC ----\
\-.... ii Ac0 OAc Ac0 NHAc 78 Ac0 OAc NC -...1 Ac0 0 0 .----\.,---\õ...---N ...1, 0 -0-....
AcHN
NC
Ac0 OAc Ac0 C' __....7.(2...\,/, 1 µ-J/N ,11), /\/, / \ VC'CN

AcHN (:) 1 NC ---\NOT)02 Ac0 OAc 70...\/(:)0'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 Fmoc0 NOP02 HOõ,......_ 1. FmocC1, pyr -,......7".... 1 HO ..,.-=-,..-0-...,......-1 OBn 2. Pd/C, H2 ___________________________________________ 0-7.7.0 .7.cl HO 3. Phosphitylation Fmoc0 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 0u ,....._(:) Ç
ODMT
n7-----l-ODMT
DMT0(5'. rt -,-,2-------/---ODMT
NC '0-P = 0 1. DCA, DCM NC 00 oBx ______________________________________________ .-0 2. DCI, NMI, ACN 0õ
I
OLIGO Phosphoramidite 56 NC 'Th -P =0 , _____________________________________________ , ,. ____________________ ., DNA/RNA 0 I
0 ,autom I
, ated synthesizer , ' I OLIGO
0¨VIMAD_0_p_oCN ,. _______ 31( 0 79b I

VIMAD _0_130CN I¨
X = S- or 0- X
Bx = Heterocylic base 1. Capping (Ac20, NMI, pyr) 80 2. t-BuO0H
3. DCA, DCM
4. DCI, NMI, ACN
AcO0Ac NC ---1 , Phosphoramidite 60 0 , 0 Ac0-----4..u..________N A;
AcHN 0 '0¨

CN
----\
AcO0Ac NC of 0 0-, _.....7.2.\,(-) (Y4(0)-Bx NC-----\j j NC0-P=0 L
Ac0 Ac O 0I
12...\/0Co' i3-0 , OLIGO , Ac0 ,. , -NHAc 01 O¨VIM_013:..0,CN
AD
X
1. Capping (Ac20, NMI, pyr) 81 2. t-BuO0H
3. 20% Et2NH inToluene (v/v) 4. NH4, 55 C, w HO OH

HO
AcH N 0-HO OH
0 0 -,., 0 r-N II I I
HO Li A), 01.-ID(O Bx r AcHN 1C) 0.
O=P-O-OHC=
HO 9 ri P¨

3- 0 , OLIGO , HO , ________ -NHAc 82 wherein Ga1NAc3-2 has the structure:
HOOH

AcHN 0 1 0 NOON
0 0, 0 HO_,ON,00^cOr Bx AcHN 0- 0 (:) 0=1'-0-jj P, I
HO ......r2._v0....._,---,V"---70- 69 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----r.....\ O.......,.....____N ,ig AcHN 0 1 0 NOON
0 0, HO,-, _.......f.2.\, ,-,-,...õ----,..õ----Nco0 ',...-0 AcHN (:) HOOH
P, 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) DMTOcipBxr OS 1. DCA, DCM
NC
O¨P=0 2. DCI, NMI, ACN

Phosphoramidite 79 OLIGO DNA/RNA
,automated synthesizer_ 0¨
X = S- or 0-VIMAD¨O¨PI-0CN
Bx ¨ Heterocyclic base 79b Ac0 OAc NCTh 0¨
AcH N
CN
NC
Ac0 OAc f Ac0--11ZZ 0-i)`030'. 1")-13.¨uc ___ Bx AcHN 0 NC y NC
Ac0 OAc .1\/0(;(13-0 = ________ OLIGO
Ac0 NHAc 0 1. Capping 0¨VIMAD-0 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----,,20 H
N---N----)r-N H 1. H2, Pd/C, Me0H (93%) AcHN
Fi H0 0 0,, A 0 2. BnO0H
83a /----\NN7 N IrN=
Ac0 OAc 0 N

Ac0 0 0 o' HBTU, DIEA, DMF, 76%
________________________________________________________________ i.
NHAc HNVN/NN___Cj 3. H2,Pd/C,Me0H

OAc _7¨/-0 Ac0 OAc Ac0\.,;\...>) i Ac0 Ac0 --...r,C,.0 NHAc H
N---N-----N H
AcHN \....--\,,.N,Iim H H 0 0__ F
83b 0 COCF3 Ac0 F
F Ac0 F NHAc HN
7N/N_Cj ., _________________________ H
Pyridine, DMF
¨
OAc jr N___ 0 83c Ac0(:\...>/) Ac0 OAc Ac0 NHAc Ac0 H 5 0 0 83e 3' N"--N---)r--N H ') I I
AcHN
0 0 F F í.F
OLIGO O-P-0-(CH2)6-NE12 Fi H 1!)1 10, Kv)c 4.
F I
OH
a-0 Borate buffer, DMSO, pH 8.5, rt Ac0 NHAc HN F F7N/NN____Cj H o 83d OAc _7¨/-0 AcO\C\..>) i Ac0 NHAc Ac0 OAc Ac0 - H
Ne---N---)r-N H
AcHN

FIC) H 0 0õ., /1)- OH
5' 3' N-(CH2)6-0-Pl -04 OLIGO ) Ac0 .(:).v0Ac 0 Ac0 NHAc HNVN/NN _____________________ e 83f OAc /¨/-0 Ac0) Ac0 NHAc Aqueous ammonia HO H r HO-----r2.\õ0 H
N'N-----)r-N H
AcHN \----Ny,õ.. 0 H H(:).--- ____ NH N-(CH2)6-0-P-0¨ [
OuG0 j H I I
HO
V 01-1/7....\/0 0 ---/---7-1 N7N-- N- 4-:_e 0 0 83h HN N
NHAc H 0 OH .__/
HO\/0/
HO
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---74:10 H
AcHN H
N'N)7.--N
0 \N
)r 0 0 H 0- )-LA I s N---rr--"N-0...---NH

H
HO
NHAc HNN____-j H
OHr, 0.-7-7 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
H
AcHN N----N---)r-N H
N

H H 0 0.._ -N -1(N-0 .-----N H

H

HO
NHAc N
OH
HO\ H
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 1. DCA
0-UNL-ODMT _____________________________________ 0 r\-..._ 7.--.7.--0Fmoc 2. DCI, NMI, ACN I
300-UNL-0-13,--0CN
Fmoc00--- N(iPr)2 P.Ø------õ,CN 85 DMT000' 3. Capping ODMT r r CN
4. t-BuO0H ----/
..--i OFmoc 0/ /-0Fmoc 1. 2% Piperidine, 2% DBU, 96% DMF 0r 0 kOFmoc _________________ ... I
3. DCI, NMI, ACN 86 O-7 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
Ac0 (._..._ 4. DCI, NMI, ACN
N.NO Phosphoramidite 60 AcHN 0 DNA/RNA
,automated synthesizer, \--\__\____\NC
( 5. Capping Ac0 OAc p O-P
Ac0-......_ NC \
0\ \--\------\19 0 j--CN

AcHN

P=0 NC-\__Q
Ac 13--Ac0 -() _________________ 70......\/0 0 DMTO--N----No-}¨\

I, Ac0 0-UNL-0-13,-0 CN
NHAc 1. t-BuO0H 0 2. DCA
3. Oligo synthesis (DNA/RNA automated synthesizer) 4. Capping 5. Oxidation 6. Et3N:CH3CN (1:1, v/v) Ac0 OAc Ac0 *NO_ AcHN 0 ,0 O-P' / \
Ac00 -o AcHN
\--"\--\----\ P

ci ri 0 ----"\-0 Ac0C)Ac -0\
Ac0 DMT-[ OLIGO )------( I _ ,CN
NHAc 5' 3' 0 0-UNL-0-P-0' (13 HO OH NH4, 55 C
HO*\7_ Y
AcHN 0 O-P' HO ,\

N---\-0 AcHN o\---\___\__\ p , PCO C1-----\--0 HO...._\/C)F1 O-HO OH
HO ---_/0 NHAc 1 OLIGO ) _____ (CM Y
5' 3' Wherein Ga1NAc3-4 has the structure:
HO OH
HON
AcHN 0 HO OH p o-p' HO \
0\--"\--"\-----\ Po- N---\,0 AcHN
/CI

P=0 o 0 _rj HOOH
=P--0 \
_____________ 0 0 OH
NHAc Wherein CM is a cleavable moiety. In certain embodiments, cleavable moiety is:

0=P-OH
\-O-Ncot _UN
0=P-OH
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 HON I

AcHN 0 HO OH \----\--\-------\ p HO

/ \
0\--"\--"\-------\ 90-AcHN 0-0.--P/N0- 0--9-----0 /
õ7---/ P=0 gi)..., jj o 0 \o-O'N\-..,D-----PH
/
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, = 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 3421 ASO comprising a phosphodiester linked GaINAc3-2 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 26 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 ì21a3 ASO comprising a Ga1NAc3-3 conjugate at the 5' position via a hexylamino phosphodiester linkage targeting Malat-1 ISIS,Conjugate Calcd Observed No. Mass Mass Sequence (5 to 3') SEQ ID No.
5'-Ga1NAC3-3,-0,mCesGesGesTesGes 661166 mCdsAdsAdsGdsGdsmCdsTdsTdsAdsGds 5'-Ga1NAc3-3 8992.16 8990.51 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 424.
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 .522, 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 ,I-Pt.22 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 22 0.07 95 0.2 77 651900 0.7 28 0.26 3' Ga1NAc3-1 23 0.07 107 0.2 86 661134 0.7 28 0.25 5' GalNAc3-2 26 Structures for 3' Ga1NAc3-1 and 5' Ga1NAc3-2 were described previously in Examples 9 and 37.
Pharmacokinetics Analysis (PK) The PK of the ASOs from the high dose group (7 mg/kg) was examined and evaluated in the same 15 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 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 20 observed. Unlike its counterpart, additional metabolites similar to those reported previously in Table L4a-10a 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 below.
1C) Table 3e23 Modified ASOs comprising GaINAc3-1 conjugate at the 3' terminus targeting SRB-Chemistry SEQ
ISIS No. Sequence (5' to 3') ID
No.
353382 GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds Full PS no conjugate 28 (parent) mCdsTdsTesmCesmCesTesTe GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds Full PS with 29 mCdsTdsTesmCesmCesTesTeoAdo,-GalNAC3-1 a Ga1NAe3-1 conjugate 655862 GesmCeoTeoTeomCeoAd Gd Td mCd Ad Td Gd Ads Mixed PS/P0 with 29 mCdsTdsTeomCeomCesTesTeoAdo,-GalNAC3-1 a Ga1NAe3-1 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 3424, 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 :.=-7-24 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 353382 Full PS without 10 52.40 10.4 28 (parent) conjugate 30 24.95 0.5 81.22 Full PS with Ga1NAc3-1 1.5 63.51 655861 2.2 conjugate 29 5 24.61 15 14.80 0.5 69.57 1.5 45.78 Mixed PS/P0 with 655862 1.3 29 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 evaluated. The results demonstrated that no elevation in transaminase levels (Table -z-425) 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 ,'4825 Effect of PO/PS linkages on transaminase levels of ASOs comprising Ga1NAc3-1 conjugate at 3' terminus targeting SRB-1 ISIS Dosage ALT AST
Chemistry SEQ
ID No.
No. (mg/kg) (U/L) (U/L) PBS 0 28.5 65 3 50.25 89 353382 Full PS without 10 27.5 79.3 28 (parent) conjugate 30 27.3 97 0.5 28 55.7 1.5 30 78 Full PS with 5 29 63.5 Ga1NAc3-1 15 28.8 67.8 0.5 50 75.5 Mixed PS/P0 with 655862 1.5 21.7 58.5 29 GalNAc3-1 5 29.3 69 Example 45: Preparation of PFP Ester, Compound 110a OAc OAc Pd/C, H2 OAc OAc Et0Ac, Me0H

Ac0 103a; n=1 .........0 0,---_______________________________________________________________________________ ).
103b; n= 7 Ac0 1\----) n ___________________________________ )0- AcHN 104a; n=1 7,-0 104b; n= 7 4 OAc AcONC:Ac AcHN 01\1___(___ OAc OAc OAc OAc n H
Ac00----\.4-^4,-N H2 PFPTFA
__T!.C...:)...n _________________________________________ Ac0 --NHIr\------NO2 AcHN DMF, Pyr AcHN n 105a; n=1 Compound OAc OAc 105b; n= 7 Ac0__T!.C...:)..Ø----,n,HN---(0 AcHN
106a; n=1 106b; n= 7 OAc AcONC:Ac AcHN 0 OAc OAc N
, n H HBTU, DIEA, DMF
Ra-Ni, H2 0 _____________ . Ac01, AcHN c,) rNH _____________________________ ).-.__------N H2 Me0H, Et0Ac 0 0 r OAc OAc Ac0 0.-----HN HO2CLO'Bn , µ

__C...:).. --.0 ` 'n AcHN 99 107a; n=1 107b; n= 7 OAc AcONC:Ac AcHN 0 O
OAc OAc n ...C..:)..\
Ac0 0 AcHN n N hl ___ NH

0 r OAc OAc , \
Ac0 0.----HN"--0 'n AcHN

108a; n=1 108b; n= 7 I3n OAc Ac0 OAc AcHN
Pd/C H2, ONN 0 108a; n=1 Et0Ac,, Me0H OAc OAc 108b; n= 7 NH
NH
AcHN n OAc OAc Ac0 0 AcHN

109a; n=1 HO
109b; n= 7 OAc Ac0 OAc AcHN 0 0 OAcOAc Ac0 NH
AcHN
PFPTFA, DMF, pyr OAc OAc 109a Ac0 0 0 AcHN
O
110a 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
HO OH
0 83e 0 3' 5' 11 AcHN '"'¨O _ I
[ OLIGO )-0-P-0-(CH2)6-NH2 OH OH
110a OH p... HOC) NI-11.______NH
1 Borate buffer, DMSO, pH 8.5, rt AcHN
2 NH3 (aq) , rt 0 r AcHN
NH

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
zofrrs----N 0 "4 AcHN

HO/îO
AcHN
HO OH

HO
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 [tt) and 3 equivalents PFP ester (Compound 110a) dissolved in DMSO (50 [tt) was added.
The PFP ester precipitated upon addition to the ASO solution requiring additional DMSO (600 [tt) 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.
NH2(CH2)6-0AdoGesmCesTesTesmCesAdsGdsTds ISIS 660254 Hexylamine 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GalNAc3-10.-0,AdoGesmCesTesTesmCesAdsGdsTds ISIS 666881 GalNAc3-10 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesrfesrfe 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 BocHN
" NHBoc i N 0 N
% n H
HONc)2 91a; n=1 TFA, DCM
91b, n=2 BocHN 'hNH
n NO2 ________ ).-___________________________________ ).-PFPTFA, DIPEA, DMF

BocHN,(A/HN 0 /n 92a; n=1 92b, n=2 H2N n Fri OAc OAc H N NH
NO2 ; Ac0 TMSOTf, DCM
---I-C-P.1.-0Ac ____________________________________________________________ ).-0 AcHN 3 _..-C-/n 93a; n=1 93b, n=2 94a; m=1 Ac 0 94b, m=2 0 OAc OAc 1-1,(:),Bn V OAc * HO \ /m Ac0 i \
, Ir-,OH
Ac0 ______________________________________ .- AcHN

yO TMSOTf 7; m=1 Pd/C. H2 64, m=2 OAc AcONC:Ac 0 .....õ4......>õ... ..,,.....jt, AcHN 0 \ ,m / n OAcr¨ OAc 0 H
93a (93b) Ra-Ni, H2 _________________ , HBTU, DIPEA, DMF Ac0 AcHN
OAc OAc 0 H
Ac0 NNz 0 --C-P...\ ,,Vr HN
m \ /n 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 011IN
.. 0 -/K.__ H HBTU, DI EA, DMF
OAc OAc 0 ____________________________________________________________ a ....:..\:10(Nr,)^ NH
Ac0 , / m n ir\-----NH2 AcHN H r HO¨ ODMTr OAc OAc 0 Ac0 0,r NN,HN---() ____________________________________________________ N.
AcHN n 0 0 'OH
97a; n=1, m=1 97b; n=1, m=2 97c; n=2, m=1 97d; n=2, m=2 OAc AcONC_)Ac 0 AcHN 0 0 ,)x)( m 1;\(\)7.1 OAc OAc 0 H
0 ODMTr _/(17 b Ac0 (:) .( H
, / m NV<'hNH
n Ir\_.--- N
AcHN H
OAc OAc 0 r ...p..., Ac0 N
0,1".k.7--If NN,4'ir HN--.0 0 "OH
, m AcHN n 98a; n=1, m=1 98b; n=1, m=2 98c; n=2, m=1 98d; n=2, m=2 OAc AcON(:Ac 0 AcH _)N 01')1,1 [\licA, [1_()____ HBTU, DIEA, DMF n n 0 97a, n=1, m=1 OAc OAc 97b, n=1, m=2 v.
Ac0 ..:..:)_\().. N
...----o, 97c, n=2, m=1 0 m H
97d, n=2, m=2 AcHN Bn HO2C0,Bn 01 c OAc 0 r AcOCI)1f, µ H
NN,,,i,yvHN --%
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 o'''.11.)-11 LN 0 Pd(OH)2/C, OAc OAc 0 -(___ 0 0 H2, Et0Ac, PFPTFA, DMF, NAPC)1 AcO.cy'Thy\ANNH
pyr AcHN H
OAc OAc 0 r H
iNN*IviHN--o Ac0 101a, n=1, m=1 AcHN 101b, n=1, m=2 0 101c, n=2, m=1 101d, n=2, m=2 OAc AcONOAc 0 AcHN O'l'h).m N 0 F
OAc_ OAc[1--/(___ 0 = F
F
H
AcHN
OAc OAc 0 r F
H
Ac0--?_\`-''',11 INN..byi HN---0 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 ( OLIGO J-0-P-0-(CH2)6 NH2 i OH
Borate buffer, DMSO, pH 8.5, rt 102d _____________________ >
2. aq. ammonia, rt HO---72--\---0);LIZIFIZI)L--\
AcHN 0 0 OLIGO
HO---""2 _____ \--- -)ANN

AcHN

HO__....2...\011ANI9N(0 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:
HooH 0 0 AcHN 0 0 N)(N 0 El 1 HO
_4,0^ 0 (ciN^er'N H H ' AcHN

HO
_.r.!....\õ: ...,) AcHN =

Example 48: Preparation of Oligonucleotide 119 Comprising GaINAc3-7 AcO0Ac Ac0 OAc _....rØ..\
, NHCBz Ac0 TMSOTf, DCE ______ Ac0-4\r C) Pd(OH)2/C
a 4 (-,NHCBz AcHN H2, Me0H, Et0Ac N -z-_-_-- i HO

4 1 35b 112 HO---in HBTU, DIEA
Ac0 OAc 0 0-1 DMF
NH2 + HO 0_,1 NHCBZ __ _ AcO4vC) AcHN 0 C)L) 105a HO

Ac0 OAc , Ac0-4r H
ON---t....\-' Ac0 OAc AcHN
4r H 0 Ac0 01\1 HCBZ

AcHN

Oiv_ j 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 Ac0-........,:). )^\-) AcHN
Pd/C, H2, Ac0 OAc 0 0 ) Et0Ac, Me0H
______________________ ..-Ac0 AcHN 0 0 Ac0 OAc Ac04,0NH

AcHN
Ac0 OAc H , F
F
F
AcHN -----fl...\' PFPTFA, DMF, Pyr Ac0 OAc )L0 0 0 ..........2..\,Oz(--)NH F
F
Ac0 AcHN 0 0 Ac0 OAc 0j ......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 T
[ OLIGO O¨P-0¨(CH2)6-NH2 I
OH
1. Borate buffer, DMSO, pH 8.5, rt 2. aq. ammonia, rt AcHN N

HO 0(`r N
OLIGO

Z
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:

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

AcHN =
Example 49: Preparation of Oligonucleotide 132 Comprising GaINAc3-5 , Boc HN
,Boc HN,Boc HN
HN,Boc H2NC) Boc 0 Boc ,N
.LOH

Boc,NOH 0 0 HBTU, TEA
Li0H, H20 ,Boc 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 H3N
H2NOH + HO 401 8 = 0 . 0 0 ' 00¨g 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 Boc, Nj.L 0 cF3c00H
123 710- NThr 0 N
HOBt, DIEA, 0 8 CH2Cl2 PyBop, Bop, DMF
r HN,Boc 127 CF3C00- I\TH3 Ac0 OAc i\ij.L N 0 0 Ac0 OH
C)--4,Thr 1-4 AcHN 7 0 ( CF3C00- 0 0 ________________________________________________ - 710-HATU, HOAt, DIEA, DMF
r cF3coo- 0 NH3 128 Ac0 OAc Ac0 N.,......--..õ_,-Nr AcHN
NH
L

Ac0 OAckijL w3-y) 0 HN'Thr N
*Ø...\, Ac0 Oz--i 0 H 0 AcHN 0 /
Ac0 OAc Ac0 n--4-\,-AcHN 0 Ac0 OAc AcO
c 0 AcHN
NH
Pd/C, H2, Me0H H o Ac0 OAc Nj-L
HN NC)1-Ac0 0 AcHN 0 Ac0 OAc o NH
Ac0 Ac0 OAc AcHN 0 130 AcO
( 0 AcHN
NH
PFPTFA, DMF, Pyr HFF
Ac0 OAc HN NC) Ac0 AcHN 0 Ac0 OAc NH
Ac0---&"2-\/ /
AcHN

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

0 83e 3'5 F'1 11 AcHN
OLIGO O¨P-0¨(CH2)6-NH2 NH
OH
1. Borate buffer, DMSO, pH 8.5, rt 131 _______________________________________________ H
2. aq. ammonia, rt HO OH
HN-Thr NH

AcHN 0 HO OH
NH
HO¨&4,(:))/ 0 H 4 N(''')0 OLIGO
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
SOo AcHN
NH
H (PI
HO OH
NH
O
HO
AcHN
HO OH
NH
HO

AcHN
=

Example 50: Preparation of Oligonucleotide 144 Comprising Ga1NAc4-11 DMTO Fmoc 1. TBTU, DIEA DMTO Fmoc Lol ACN, VIMAD Resin Lol pip:DBU:DMF
.2. Ac 20 Capping . 0 0 (2:2:96) b--- )-OH Kaiser: Negetive "610-0 HN-Fmoc DMTO H Fmoc,NOH
LO
H /
0 DMTr-...

Lal b HBTU, DIEA, DMF

135 b NH-Fmoc DMTr ) 1. pip:DBU:DMF 0 / H j 0 1. 2%
hydrazine/DMF
Kaiser: Positive Kaiser: Positive ________________ Ni- ....iN).L(CH2)5' N r ____________________________ 0-2. Dde-Lys(Fmoc)-OH (138) 0 H 2. Fmoc-Lys(Fmoc)-OH
(140) HATU, DIEA, DMF d o HATU, DIEA, DMF
Kaiser: Negative 0 Kaiser: Negative a ,Fmoc W.
HN
) /
HNOH,Fmoc DMTr 0 ti N--11"-(cH2)5- NN Fmoc ....1 HN,Fmoc Ac0 OAc AcOy0 o AcHN NH0 Ac0 OAc AcOO H 0 00 AcHN 0 H
1. pip:DBU:DMF

Kaiser: Positive 2. 7, HATU, DIEA, Ac0 OAc DMF DMTO
Kaiser: Negative i7H NH
AcHN 0 0 Ac0 OAc Ac0or--NH
AcHN 0 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 /
AcHN ---NH
4, \
Ac0 OAc Ac0 0 NI 0 AcHN
DNA syntesizer 0 142 _______________ V.- 11 1 ----A'-)-3-)r__Ny Ac0 OAc 0 ____&Z, C' Ac0 __________________________________________________________________________ , ,CM ) __________________________________________________________________________ s.
ASO ., AcHN
Ac0 OAc Ac0 )7---NH
AcHN 0 HO OH
HO
/
AcHN ----NH

HO OH
HO NI
AcHN 0 H pH
aqueous NH3 0 y H
___________________ ).-( ICMJ ___________________________________________________________________ .
______________________________________________________________________________ , ASO
AcHN ' ___ , AcHN

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 0 HO OH

AcHN N pH

HO OH
oi AcHN O:Y(0 ral HO OH
HO*...\,0 r AcHN
Example 51: Preparation of Oligonucleotide 155 Comprising GaINAc3-6 OH

BrOH 0 0 y ENI1NJL,OH

2M NaOH 00H

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

HO.,....w,N)..,,0 0 Ac0 OAc 35b 4 7,--- AcOo- N A0 40/
TMS-0Tf, 4 A molecular sieves, CH2Cl2, rt H
AcHN

o ,:) kJ' j-L
Ac0 OAc 0 II OH
H2, Pd(OH)2 /C 2 0 147 ....-NH2 ______________________________________________________________ 1.
Et0AdMe0H AcHN 105a HBTU, DIEA, DMF, rt Ac0 OAc 0 H H2, Pd(OH)2 /C, Et0AdMe0H

___________________________________________________________________________ Ac0___ro., ..,....._____O
N
AcHN H

Ac0 OAc AcOoN ..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 E

_____________________________ Ac0 OAcAc0- AcHN 3 H

N 0 =
AcHN 3 H

146 ________________________ Ac0 OAc HBTU, DIEA, DMF
Ac0 3 NHAc Ac0 OAc Ac0 OAcAcOoNH 0 Pd(OH)2/C, H2 AcHN 3 H

Me0H, Et0Ac AcHN 3 H 0 AcOt 1OAc AcO/CIHN

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/ N)N
Ac0 OAc 0 0 AcHN 3 H Z(:_r,) 0 0 0 H ) Bn001-1 AC0-4r()NN----inN N
3 H OBn 83a 0 151 ),,.. AcHN 3 H
'-----_-.:---0 PFP-TFA, DIEA, DMF
Ac0 OAc 0 N)./NH
Ac0 3 H
NHAc Ac0 OAc Ac0(:)NN
Ac0 OAc AcHN 3 H Z,),,C) 0 0 Pd(OH)2/C, H2 ..\.r 0 Ho N N)COH
Ac0 )1N---Tr---N

Me0H, Et0Ac AcHN 3 H 0 ----r.----0 Ac0 OAc 0 ..i..,0....\/0.,_____õ...-õ=(....r, N)./NH
Ac0 NHAc Ac0 OAc Ac0 N)N F
Ac0 OAc AcHN 3 H ,C) F
0 0 .

PFP-TFA, Dl EA
o N) --..,,, }..,,...N---r----NZ,), F
o ____________ ). Ac0 N 3 H F
DMF AcHN 3 H 0 -----r.---0 F
Ac0 OAc 0 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 5', I I HOOH 0 ( OLIGO J-O-PI-0-(CH2)6 NH2 -====r2..\,_--0A NC
HO
OH .4 H
HN\e H
154 1 Borate buffer, DMSO, , HOOH AcHN

H
pH 8.5, rt 2 aq ammonia, rt 0 4 II0 II 5 --, =---=

AcHN
N1'.
HOOH
r 0 HO ON1.---0 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-----\---01`-r'N

AcHN HN

H
N)C'1\11(cyH

HO N
---r-\--"
4 H l''r50 El I
0 o 0 AcHN
ri---µ
HooH , 0 AcHN .
Example 52: Preparation of Oligonucleotide 160 Comprising GaINAc3-9 AcO0Ac 0 AcC??Ac _....7....\\O 0 H0v(0 TMSOTf, 50 C Ac0 10 111#
Ac0---.Z.õ0Ac AcHN CICH2CH2CI, rt, 93% N-----_-_--zq TMSOTf, DCE, 66%

Ac0 OAc Ac0 OAc ........I7 0 4 H2, Pd/C
Ac0 's II" Ac0 '10 Me0H, 95% 10 AcHN 0 AcHN 0 OH
Ac0 OAc HBTU, DMF, EtN(iP02 Phosphitylation __________________________________________________________________________ ).-Ac0 C)t)(7 81%
DMTO '10 AcHN 0 NCR. ODMT
b1H

/
p¨P
Ac0 OAc NUP02 11\R
Ac0 0 'e/r7 '10 AcHN 0 ODMT

Compound 156 was synthesized following the procedure described in the literature (J. Med. Chem.
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

AcHN
0=P¨OH
1 DNA synthesizer HooH
159 _______________ 2 aq NH4OH Ho00 AcHN
0=P¨OH
HOOH
_______________________________________________________ OLIGO
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 (GalNAc3-9a) 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:
..QH
HOOH
HO

AcHN
0=P¨OH
HO OH
HO0orNR.0 AcHN
0=P¨OH
HOPH
HO EMI
AcHN
=

Example 53: Alternate procedure for preparation of Compound 18 (Ga1NAc3-la and Ga1NAc3-3a) Ao H2NNHR H TMSOTf HO N NHR __________ ,...
\) R = H or Cbz OAc 0 O. iogr....
161 I¨ R = H, 162a 0 CbzCI, Et3N 1 R = 162b Ac0 4)O

PFPO
OAc)7--------1 H
Ac0 o 0 NNHR + PFPO0.,..7¨NHCBZ ¨Am-NHAc 0 0 0 0, R = Cbz, 163a K) Pd/C, H2 I¨ PFPO
= H, 163b OAc O. iogr.... 0 0 0,, Ac0 H
NHAc C-) 1:-INN)T.--Th OAc Ac0 0 0_ , JI H
NNH
Ir,./O.,¨NHCBZ
NHAc 0 CI)i 10 OAc OAc HN.N14*--..7 Ac0 ,(...rrk) 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) HO2C"----1 PFPTFA PFP00, 0, DMF, pyr 0 H 0 2 Cr*'..../.õ,/¨

PFPO.,(----/C).õ¨NHCBZ
, 0' 0 OH CI) HO2C.,) PFPO

BocHN N
)1---------\
, BocHNNH2 0 0 1. HCI or TFA
_______________________ BocHNNHIc,.,0....¨NHCBZ ________________________ ,...
DIPEA , 2.
0 0 0 OAc Ogv..... 0 BocHNN)L) Ac0 -0 (0PFF
H
165 NHAc OAc 166 O. iog r.... 0 0 ,, 1. 1,6-hexanediol Ac0 0 H or 1,5-pentane-diol NHAc C-) 1:-INN)T.----....\ TMSOTf + compound 4 OAc 2. TEMPO
?Ac 0 0 0--, 3. PFPTFA, pyr 0 1.r Ac0 0_, JI H
NNH .õ,./0 --.õ..NHCBZ
NHAc 0 C1)1 10 OAc OAc HN.N7-0r, H
Ac0 A0 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 Modified ASO targeting SRB-1 SEQ
ASO Sequence (5 to 3') Motif Conjugate ID No.
ISIS 353382 GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds 5/10/5 none (parent) mCdsTdsTesmCesmCesTesTe GeamCeaTeaTeamCesAd Gd Ta mCd I'd Gd ISIS 655861 sss sssss 5/10/5 Ga1NAc3-1 mCda'r ds'f esmCesmCesT esTeoAdo¨GalNAc3¨la GesmCesTesTesmCesAd Gd mCd Ad I'd Gd Ad ISIS 664078 sss sssss 5/10/5 Ga1NAc3-9 mCdaTdsTesmCesmCesTesTeoAdo¨GalNAc3-9a Ga1NAc3-3a-o'Ado ISIS 661161 GeamCesTesTesmCesAdsGasTasmCdsAdsTasGasAds 5/10/5 Ga1NAc3-3 30 mCdsTdsTesmCesmCesTesTe Ga1NAC3-8a¨o'Ado ISIS 665001 GeamCesTesTesmCesAdsGasTasmCdsAdsTasGasAds 5/10/5 Ga1NAc3-8 30 mCdsTdsTesmCesmCesTesTe 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 4027, 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 4-027 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.

1 Table 4428 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.
1 Table 4-129 Modified ASO targeting SRB-1 SEQ
ASO Sequence (5' to 3') Motif Conjugate ID No.

GesmCesTesTesmCesAdsGasTasmCdsAdsTasGasAds 5/10/5 no conjugate 28 (parent) mCdsTdsTesmCesmCesTesTe GesmCesTesTesmCesAd Gd Id mCd Ad Id Gd Ad sISIS 655861 5/10/5 Ga1NAc3-1 29mCdsTdsTesmCesmCesTesTeoAdo, -Ga1NAC3-1- a GalNAC3-2a-0,AdoGesmCesTesTesmCesAd Gd Td sISIS 664507 5/10/5 GalNAc3-2 30mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe Ga1NAc3-3 am'Ado GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds 5/10/5 Ga1NAc3-3 30 mCdsTdsTesmCesmCesTesTe ISIS 666224 Ga1NAC3-5a-0,AdeGesmCesTesTesmCesAdsGdsTds 5/10/5 Ga1NAc3-5 30 mCdsAdsTdsGdsAdsmCdsTasTesmCesmCesTesTe GalNAc3-6.-0,AdoGesmCesTesTesmCesAdsGasTas ISIS 666961 5/10/5 Ga1NAc3-6 30 mCdaAdaTdaGdaAdamCdaTasTesmCesmCesTesTe GalNAc3-7.-0,AdoGesmCesTesTesmCesAdsGasTas 5/10/5 ISIS 666981 Ga1NAc3-7 30 mCdaAdaTdaGdaAdamCdaTasTesmCesmCesTesTe GalNAc3-10.-0,AdoGesmCesTesTesmCesAdsGasTas 5/10/5 ISIS 666881 Ga1NAc3-10 30 mCdaAdaTdsGdsAdsmCdsTdsrresmCesmCesTesTe 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 43-30, 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 4-330 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 44-31 below.
Table 4-431 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.
1 Table 4532 Modified ASO targeting ApoC III
ASO Sequence (5' to 3') Linkages SEQ ID
No.
ISIS AesGesmCesTesTesmCdsTdsTdsGdsTds PS 20 304801 mCdsmCdsAdsGdsmCdsTeaTeaTesAes're ISIS
ikesGesmCesTesTesmCdsTdsTdsGdsTdsmCdsmCds PS 21 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 4633 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 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 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 4-734, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner. The antisense oligonucleotides comprising the phosphodiester linked Ga1NAc3-1 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.
1 Table 4:734 Modified ASO targeting SRB-1 % Saline SEQ ID
ASO Sequence (5 to 3') Dose mg/kg control No.
Saline 100 0.6 73.45 TIsmCksAdsGdsrrdsmCdsAdsrrd Gd Ad ISIS 440762 s s s 2 59.66 22 mCdsTdsrllsmCk 6 23.50 0.2 62.75 TksmCksAdsGasTasmCdSAdSrrdSGdSAdS 0.6 29.14 mCdsTdsrllsmCkoAdo,-GalNAC3-19 2 8.61 6 5.62 0.2 63.99 TksmCksAdsGdsrfdSniCdSAdSTdSGdSAdS 0.6 33.53 mCdsTdsrllsmCkoAdo,-GalNAC4-11a 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.
1 ALTs, ASTs, total bilirubin and BUN values are shown in Table 4-6-35 below.
1 Table 4835 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 GalNac3-1 (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 .4q,36 Modified ASOs targeting FXI
SEQ ID
ASO Sequence (5' to 3') Linkages No.
ISIS TeaGesGesTesAesAdsTdamCdamCdaAdamCds PS 31 404071 TdsTdsTdsmCdsAesGesAesGesGe ISIS TesGesGesTesAesAdsTasmCdsmCdsAdsmCds PS 32 656172 TdsTdsTdsmCdsAesGesAesGesGeoAdo,-Ga1NAc3-18 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 F41,37 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 Ga1NAc3-1 PS

SIS 0.7 49 I
656173 2 22 Ga1NAc3-1 PO/PS

As illustrated in Table C-s-137, 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 Aa37a, 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).
Tableµ:=4a3.7a 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.

1 Table 5438 Dosage Total Total ISIS No. ALT AST CRE BUN
Conjugate mg/kg Albumin Bilirubin 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.
1 Table 52-39 Modified ASO targeting SRB-1 SEQ
ASO Sequence (5' to 3') Motif Conjugate ID No.
GesdmsCdessTeessTesemsCesAesd eGsd eTd mCd Ad Td Gd Ad s 5/10/5 none 28ISIS 353382m,rrm,mCTT
GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds ISIS 655861 5/10/5 GalNAc3-1 mCdsTdsTesmCesmCesTesTe0AGalNAC3-1 a GesdmsCdesoTeeooTeoemoCeoeAsde sGde oTdd mo,C-d Ad Td G3-d Aad s 5/10/5 Ga1NAc3-1 29ISIS 655862mCTrmCmCTTAGa1NAC1 GalNAc3-3a-0,AdoGesmCesTesTesmCesAdsGds ISIS 661161 5/10/5 GalNAc3-3 30 TdsmCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GalNAC3-8a-o'AdoGesmCesTesTesmCesAdsGds ISIS 665001 5/10/5 GalNAc3-8 TdsmCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds ISIS 664078 5/10/5 GalNAc3-9 mCdsTdsrresmCesmCesTesTe0AGalNAC3-9 a GalNAC3-6a-o'AdoGesmCesTesTesmCesAdsGds ISIS 666961 5/10/5 GalNAc3-6 TdsmCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe Gad1 sNAdsC3d-s2a-ds0,AddsoGedssmCdsesTesesTeessmCeessAde sGed d s 5/10/5 Ga1NAc3-2 30ISIS 664507mCATGAmCTTmCmCTT
Gad1 sNAdsc3d-s10das-0,dAsdoGdsesmdsCeessTesTesesmCesesAesd Ged d s 5/10/5 Ga1NAc3-10 30ISIS 666881mCATGAmCTTmCmCTT
Gad1 sNAdsC3d-s5a-ds0,AddsoGedssmCdsesTesesTeessmCeessAde sGed d s 5/10/5 Ga1NAc3-5 30ISIS 666224mCATGAmCTTmCmCTT
Gad1 sNAdsC3d-s7a-ds0,AddsoGedssmCdsesTesesTeessmCeessAde sGed d s 5/10/5 Ga1NAc3-7 30ISIS 666981mCATGAmCTTmCmCTT

Capital letters indicate the nucleobase for each nucleoside and niC 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 IC50 was calculated using standard methods and the results are presented in Table ,i-340. 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.
Tab1e40 Internucleoside SEQ ID
ASO IC50 (nM) Conjugate linkages No.
ISIS 353382 190a PS none 28 ISIS 655861 lla PS Ga1NAc3-1 29 ISIS 655862 3 PO/PS Ga1NAc3-1 29 ISIS 661161 15a PS Ga1NAc3-3 30 ISIS 665001 20 PS Ga1NAc3-8 30 ISIS 664078 55 PS Ga1NAc3-9 29 ISIS 666961 22a PS Ga1NAc3-6 30 ISIS 664507 30 PS Ga1NAc3-2 30 ISIS 666881 30 PS Ga1NAc3-10 30 ISIS 666224 30a PS Ga1NAc3-5 30 ISIS 666981 40 PS Ga1NAc3-7 30 aAverage of multiple runs.
Example 61: Preparation of oligomeric compound 175 comprising GaINAc3-12 Ac0 0 OAc Bo c , N N H2 H 0 Ac0 Pfp0 Ni____OAc ,)- C) /...C240Ac 91a )0._ Boc N N
HN NAc H H
OAc HN , Ac HOOC
H ) Ac0 OAc CBz N,N \¨COOH
COOH

OAc ________________________________________________________________ H
DC M HN
"Ac HBTU DIEA DMF

Ac0 OAc )r HN ¨Ac HN

}¨N ---/---j 101 Ac0 0 y ENI N 1N \ C) 0 oAc 1.
.......--..,N ).-----0 ,_,,,=\ N "Ac OAc H H H
HN
0 u ----\----A 0 HN Ac0 :)Ac OAc "Ac Ac0 OAc 0 o j21___OAc )1' __ HN Ac HN
Pd(OH)2/C, H2 0 H
}¨N --7-----/
Me0H/Et0Ac _)=._ Ac0 m 0 0 Nz..7.0Ac k '= N N')I---C) OAc ,-, HN H H
HN
r, "Ac----\-----\ 0 HN Ac0 \z_ii)Ac OAc "Ac F

40 0)..L0 F
F
benzyl (perfluorophenyl) glutarate _______________________________ lir DMF
A...c::_)Ac 0 0 OAc Kzõ..õ,,.....,..,r0 HN HN ...Ac }--N--7---/
H Ac0 0 :)Ac O 0 N N \ ?I
0 0 ,\ \NN )0 0 OAc HN
iokc----\------\ 0 HN AcOrkc 0\/0 OAc HN
"Ac Ac0 OAc ,,Ac HN

Pd(OH)2 i C , H2 },.- N--/-----/
172 __________ >
H m 0 0 Ac0 T)Ac Me0H / Et0Ac HO N ¨ \ If \ N
0 0 ,-, OAc H
HN'Ac HN Ac0 1:1:)Ac 0\ /0 OAc 173 HN, Ac Ac0 OAc PFP-TFA
0 0,21.__OAc DI EA DM F
)LVy HN --Ac HN
}
0 H ,¨N --.7----/
F F
H Ac0 0 Ni_ic)Ac F ii 0 NN, ___________ (1:( r- 0 0 N
u F F ' ,\ N OAc H HN
(., 0 'Ac HN Ac0 1:1:)Ac \ /
OAc 174 HN, Ac 83e 3'5') 11 F
( OLIGO O¨P-0¨(CH2)e¨NH2 OH
174 1. Borate buffer, DMSO, pH 8.5, rt ________________________________ >
2. aq. ammonia, rt OH OH
HO.....r.C.,:)..o 0 AcHN
NH
OHOH
\------\¨ENI
HO ,/0 \...-0 AcHN
((),rEi=¨=
OLIGO
HH 6 ..__, ..__..
N

ri 0 H

HOD....\/
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( 1.1 HATU, HOAt AcHN OH _HA
176 H2N- y H DIEA, DMF
O___- 0 V

OAc OAc Ac0--C--)-.\--ONc AcHN NH
OAc OAc H 0 H2, Pd/C
Ac0----!---1.--ON7NNO 401 AcHN
H H
0 y 0 /
OAc OAc HN

Ac0-----\:) .-0 AcHN 0 OAcf- OAc Ac0----\:) AcHN NH
OAc7.- OAc PFPTFA, TEA
Ac00].,Nri\i,).LNOH ________________________________________________ AcHN PO' H H DMF

OAc7.- OAc r 178 HN
Ac0-.)..\--0 AcHN 0 r-Ac OAc AcO
AcHN NH
7.-Ac OAc AcOH
N
AcHN N N F
r-Ac OAc Hr( 179 Ac0 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

AcHNNj-LN-HNo¨ 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
H0 .7..C...:) (jj C)-------WNH
AcHN
OH OH
H0*._ 0 ,cr H 0 H 0 N
Nõ.....õ.õ..-..õ.õ.Thr N___T6 Emi AcHN H 0 L.

0 r NH
HO
HO ...la/O 0..._/....,./...)\--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 NHAc µ h o 0 0 .---IL
"C)/j-hl 0 F
Ac0 / 6 F
NHAc 0 0 0 OAc Ac004,./."-, N N)--) Ac0 % 16H
NHAc OH
83e HE100 H

NI-14, Ac 76NIno 0 ( OLIG0J-0-P-0-(CH2)6-NH 0 0 I HO\(:c) _H
M
_______________________________________________________________________________ -OH Ir.--0 H
187 1. Borate buffer, DMSO, pH 8.5, rt HOZ- ---1----V \ 76 H 6 Z
OLIGO . ., ________________________ r NHAc 0 0 0 2. aq. ammonia,Hc, rt OH
õ( ,N,---) HO Vollik. 0tz '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_...*o'(--r'N
H
AcHN N

HO
/
AcHN 0 HOOH
N--CI
HO-i-j 0 AcHN
Example 64: Preparation of oligomeric compound 197 comprising GaINAc3-15 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
-"1"" =--\ ---Z-----/'"""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 -PH, 0 oi_pi Nr-D
AcHN 0 0 0, HOOH
o HO_...rEs\,) 0 AcHN 0 P, o' op HO OH (x N
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 S-441 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') Conjugate SEQ
No.
ID
No.
353382 none GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe 661161 (-I 1XT A 2 A r.;_ rTT c A A crr aii.,,c3-...,a-0,-40¨esmes es esmses, .-ass=-=ds dsms-,c1s, .clsmscls ds GalNAc3-3 30 m m Tes Ces CesTesTe 671144 GalNAc3-12a-0,AdoGesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTds Ga1NAc3-12 30 m m Tes Ces CesTesTe 670061 GalNAc3-13a-0,AdoGesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTds Ga1NAc3-13 30 m m Tes Ces CesTesTe 671261 GalNAC3-14a-0,AdoGesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTds Ga1NAc3-14 30 m m Tes Ces CesTesTe 671262 GalNAc3-15a-0,AdoGesmCesTesTesmCesAdsGdsrrdsmCdsAdsrrdsGdsAdsmCdsrrds Ga1NAc3-15 30 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 :;-:=42, 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 5S42 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 js-t4-43 below.
Table 5643 Total Dosage ALT BUN
ISIS No. AST (U/L) Bilirubin Conjugate (mg/kg) (U/L) (mg/d1-) (mg/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-
14 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 5744 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
- a ()' ¨ Ga1NAc3-3 a Ad es es es es es ds ds ds ds ds ds m m GdsAds CdsT ds es es Ces CT
dsesTe 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 33 es eo eo eo eo ds ds ds ds ds ds m m GdsAds CdsTdsTeo Ceo CT
dsesTe 671165 Ga1NAc3-13a o - ,A mC T T mC A G T mC A T Ga1NAc3-13 a Ad do 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 f,-845, 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 M.45 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 serum were measured relative to saline injected mice using standard protocols.
Total bilirubin and BUN were 5 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 ',.-e-46_be1ow.
Table FAN6 Dosage ALT AST Total BUN Ga1NAc3 CM
ISIS No. Bilirubin (mg/kg) (U/L) (U/L) (mg/d1-) (mg/d1-) Cluster 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 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 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 Ga1NAc3-3a A, 5 24 49 0.2 29 15 25 87 0.1 25 0.5 30 77 0.2 27 Ga1NAc3-3a Te 1.5 22 55 0.2 30 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 AcHN0 2 N 0 OAc OAc 0 1--l'(____ H 0 ODMTr Ac0 NH N /
ir........--- 1. Succinic anhydride, AcHN DMAP, DCE
OAc OAc 0 r (17 Na 2. DMF, HBTU, DIEA, .
H
Ac0 0....:)..\_, r N....4...),.,HND 0 OH
PS-SS
AcHN 2 2 98d Ac0 OAc H H
____.=.,...1\zOr N 0 Ac0 N
AcHN 0 Ac0 OAc \ 0 0 zODMT
___..z.\/_,'NH'K-----FN1 s / \ /-. 1. DNA Synthesizer Ac0 N N
0,-H )11...-tIL 2. aq. NH3 AcHN 0 0 \ __ C

Ac0 OAc HN
Ac0__...2..\0_,:,).------fr-N 0 AcHN 198 0A.
HO OH
H H
.1 HO0 , ,,r N.,..-, N }D
, __ 0-- , CM ___________________________________________________________ ., i ol go µ¨[
, HO OH AcHN 0 \

, 2 1 AcHN 0 0 ------- OH
HO OH HN

HO ---====="2-= -1--).-------lri 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:

HO_...7,2.0N,.-.H.N....11...õ,.\
4H 2 H [ cm D
/
HOOH
AcHN H 0 0 _-0 0 NN .LIANL......
H
HOC)11A4 HN 2 0 AcHN OH

HO__orf.:)._\01)AN NO

AcHN
Example 68: Preparation of oligomeric compound 200 comprising Ga1NAc3-17 OAc 83e Ac00Ac 0 3' 0 5' I I
0 , , jt AcHN 0" -'' -N""\N 0 F (OLIG0)-0-P-0-(CH2)6-N

H OH
0 H OF la F
0 aA 0 oc . , N H
N.-----jc F 1. Borate buffer, DMSO, pH 8.5, rt Ac0 N 0 AcHNO
OAc OAc HH H

0 F 2. aq. ammonia, rt AcO-r---- 0NHN o AcHN 0 102a HO H
AcHN 0 0 OLIGO) AcHN

HO0')ANN(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:

HO_r2._\,OLNN) AcHN H 0 0 H H

AcHN

AcHN
Example 69: Preparation of oligomeric compound 201 comprising GaINAc3-18 OAc Ac0,0Ac 0 0 83e F 3 5' 0Ac OAc 0 I I
H 0 0 F 16 F í.OLIG0)-0-7-0-(CH2)6-1 r OH
AcHNo_ F 1. Borate buffer, DMSO, pH 8.5, rt Ac OA H c 0 r F ____________________ r HN--(-1 2. aq. ammonia, rt AcHN

102b N) AcHN 0 0 HOOH 0 0 m)c)L
hi0¨ OM ¨ OLIGO
N----[1 ____________________________________________________________________ ..

AcHN

NNO

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 AcHN H 0 0 ,,NyN-NN)CFNI,,, 0¨[cm, ______________________________ i H
FNi 0 AcHN

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
/ (:)(NR.0 AcHN
0=P¨OH

HOOH
HO
AcHN

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

F 0 F EtN(iPr)2, CH3CN F),__1,(,),A

3 N "iii0H
F
kir N....,..,¨..õ¨......A 0 F DMTO 0 -b11-1 Hd AcO0Ac 0 Ac0-2-\,C)opfp AcHN 166 K2CO3/Methanol 3 N "iii0H
________________ ACN

AcO0Ac 0 Phosphitylation _.1.2..\,__NAp..iii0H
Ac0 AcHN
DMTO

Ac0....4,0Ac Ac0 0 p C _______________ 1. DNA synthesizer 1._____C) Nif( 0 õ
: )?... .
2. aq. NH3 AcHN I
209 DMTO (iPr)2N
.pH

74, AcHN I
0=P¨OH
I
p HO
HO _4\z NR., AcHN
I
0=P¨OH
I
p HO_...7....\, irlHJL

0 0 __ avl __ 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:
pH
OH
0 )-L----NR..._.
HO ,3 3 AcHN 0 0 I
0=P¨OH
I
OH p Th AcHN 0 0 I
0=P¨OH
I
OH õ

MN

AcHN 0 0 ell-Example 72: Preparation of oligomeric compound 215 comprising GaINAc3-21 HO
AcO0Ac NH
Ac0--,O AcO0Ac LLOH
AcHN 176 __________________________________________ Ac0--12-\, )LN
OH
BOP, EtN(iPr)2, 1,2-dichloroethane AcHN
OH

ODMT
AcO0Ac DMTCI, Pyridine, O Phosphitylation AcHN
OH

NC
/0¨) AcO0Ac 1. DNA synthesizer 0 N(rPr) 2 ODMT 2. aq. NH3 AcHN

OH
OH
HO
HO

AcHN
0=P¨OH

OH
HO

\ M

AcHN 0 0-=P¨OH

OH
HO
or N
HO
0 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("r'N

AcHN
I
0=P¨OH
I

OH
r--7 HO__....r.0,...\
HO N
0'1`4;
0 ---.
AcHN 0 I
0=P-OH
I

OH
rj 0 oN
HO "3 II
AcHN

Example 73: Preparation of oligomeric compound 221 comprising Ga1NAc3-22 H,N ,OH
F3C .,11 II 0 E11 , II
0 F 0 F H 211 F3C I\ L,N H

OH H
, DIEA ACN
F

DMT-CI F3C .,11 )-LN ,DMTr _______________ ' ________ 1 I I
pyridine 0 H Me0H / H20 H2N N ,:)DMTr Ac0 /0Ac F

H Ac0 218 OH NHAc F
_________________________________ 0 _________________________ 0 OAc Ac01 11 F\J L, ,DMTr 0 0-1 Phosphitylation \/
Ac0 \ 0 H ___________________________________________________________ .-NHAc OAc Ac0 __ C='FI\1)-L,N ,ODMTr \ID\VC) Ac0 ________________ 0 H
NHAc I
NC-0. N(iPr)2 NC P

OF-1\
0\z0 NHAc 1. DNA Synthesizer 0 I ,0 OH -KOH
2. Aq. NH3 0\z0 NHAc OH 0 I ,0 ,P( OH _________________ CO OH
HO ____________________ NHAc 0NIT) ___________________________________________________________ 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:

I=LNzzOH

NHAc o OH 0 1.0 .P:
OH Nz0 OH
\O\ruz.1 NHAc o OH o 1.0 .P:
OF& orNNzzO OH
\/

NHAc (:)( 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 047 Modified ASOs targeting SRB-1 ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
G CTT CAGT CA TGA CTT
353382 es es es es es ds ds ds ds ds ds ds ds ds ds es m m n/a n/a 28 Ces CesTesT
0e GaINAc3-3a- ,Ad G CTT CAGT CAT
661161 0 es es es es es ds ds ds ds ds ds m m Ga1NAc3-3 a Ad GdsAds CdsTdsTes Ces CesTesTe o GaINAc3 -3a - ,G CTT CAGT CAT
666904 es es es es es ds ds ds ds ds ds m m Ga1NAc3-3 a PO

GdsAds CdsTdsTes Ce CesTesTe Ga1NAc3-17a-0,AdoG CT T CAGT CAT
675441 m es es em es m es ds ds ds ds ds ds Ga1NAc3-17a Ad 30 GdsAds CdsTdsTes Ce CesTesTe GaINAC3-18a-0,AdoG CTT CAGT CAT
675442 m es es em es m es ds ds ds ds ds ds Ga1NAc3-18 a Ad 30 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 ,s;4-47 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 i-448, 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 ::3448 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 42 1 Q below.
Table 4249 Dosage ALT AST Total BUN Ga1NAc3 CM
ISIS No.
(mg/kg) (U/L) (U/L) Bilirubin(Ing/d1-) 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 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 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 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 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 -5441, _____ and .j,4--47 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 050 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 62-50 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, 15 particularly when taking 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 HO0..õ...õ.õ--,..,0,---....õ,..0 11 -DP- H000,0Ts Pyr 4, TMSOTf OAc 0 N3 OAc____T..___ OAc N3 224 NHAc Pd(OH)2OAc OAc7..._..\._ ACN
_____________ ).-H2, Et0Ac, Me0H OAc \
NHAc \ F 0¨/K ____________________________________________________________ /3 C¨NO2 OAc OAc H
OAc 0 OAc OAc NHAc H NO2 1) Reduce 0 C)----ON 2) Couple Diacid 0 3) Pd/C
OAc 0 O 4) PFPTFA
NHAc oAcOAc OAc NHAc 228 OAc OAc7.......\. H

OAc F
OAc OAc NHAc H NH F
1C) 0 0 0-----0\/N
OAc ____________ 0 0 0 NHAc oAcOAc OAc F
NH
NHAc 229 83e 3' 5' 11 ( OLIGO )-O-P-0-(CH2)6-NH2 OH
1. Borate buffer, DMSO, pH 8.5, rt 2. aq. ammonia, rt OH H
OH___\.._....\, c!.\C)---07N0 OH
OH H
Hya\11-lir N A.-.)0 in =
____________________________________________________________ OH.1......\vNHAc 0 0 C)=-=-=ON

oligo , ., OH \r0 NHAc 01_11...
...... 0 NH (:;,C) 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
N.,...,0 -.-OH
OH H
0 C)=-=-=0 OH \r.0 NHAc 01_11..___\
(Th NH
0 0`-'s=--0 OH
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.
1 Table=ti'-451 Modified ASOs targeting SRB-1 ISIS, SEQ
Sequences (5' to 3') CM
No. Cluster ID
No.
m m m GalNAc3-3a-0,AdoGes CesTesTes es CAdsGdsTds CdsAdsTds 661161 m m m Ga1NAc3-3a Ad GdsAds CdsTdsTes Ces CesTesTe Ga1NAc3-3 - ,G m m m CTT CAGT CAT
666904 a es es es es es ds ds ds ds ds ds m m m Ga1NAc3-3a PO 28 GdsAds CdsTdsTes Ces CesTesTe Ga1NAC3-10a-0,AdoG m m m CTT CAGT CAT
673502 m es eo eom eo m eo ds ds ds ds ds ds Ga1NAc3-10a Ad 30 GdsAds CdsTdsTeo Ceo CesTesTe m m m GaINAC3-9am'AdoGes CesTesTes es CAdsGdsTds CdsAdsTds 677844 m m m Ga1NAc3-9a Ad GdsAds CdsTdsTes Ces CesTesTe GaINAC3-23a-0,AdoG m m m CTT CAGT CAT
677843 m es es em es m es ds ds ds ds ds ds Ga1NAc3-23a Ad 30 GdsAds CdsTdsTes Ces CesTesTe m m m m m G CTT CAGT CATGA CTT C
655861 es es es es es ds ds ds ds ds ds ds ds ds ds es es m Ga1NAc3-la Ad 29 CesTesTeoAdo,-Ga1NAc3-1a m m m m m G CTT CAGT CATGA CTT C
677841 es es es es mes ds ds ds ds ds ds ds ds ds ds es es Ga1NAc3-19a Ad 29 CesTesTeoAdo,-Ga1NAc3-19a m m m m m G CTT CAGT CAT GA CTT C
677842 es es es es mes ds ds ds ds ds ds ds ds ds ds es es Ga1NAc3-20a Ad 29 CesTesTeoAdo,-Ga1NAc3-209 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 -51 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 52, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner.
Table :3 552 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 Ga1NAc3-10 a 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 677842 1.5 53.56 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 -,(1-53 below.
Table 4453 Total CM
Dosage ALT AST BUN Ga1NAc3 ISIS No. Bilirubin (mg/kg) (U/L) (U/L) (mg/d1-) 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 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 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 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 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 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 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 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 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 054 Modified ASOs targeting AGT
ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID
No.
552668mCAAmm CTGATTTTTGmCmCmC AG
n/a n/a 34 kJesrles e mCesAesmC es es GesAdsT dsT dsT dsT dsT ds GdsmC dsmCdsmCdsAes Ges GalNAc3-la 669509 Ad 35 GesAesTeoAdo'¨Ga1NAc3-18 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 o-;i--54 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 446:5, 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 6855 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 i3-9-56_be1ow.
Table 056 Liver transaminase levels and rat body weights Body CM
Dosage GalNAc3 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 Ga1NAc3-la Ad Example 79: Duration of action in vivo of oligonucleotides targeting APOC-III
comprising a GaINAc3 conjugate The oligonucleotides listed in Table Y-0-57 below were tested in a single dose study for duration of 10 action in mice.
Table 7-457 Modified ASOs targeting APOC-III
ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
AesGesmCesTesTesmCdsTdsTdsGdsTdsmCdsmCdsAdsGdsmCdsTesTes 304801 n/a n/a TesAesTe AesGesmCesTesTesmCdsTdsTdsGdsTdsmCdsmCdsAdsGdsmCdsTesTes 647535 Ga1NAc3-la Ad TesAesTeoAdo'¨GalNAc3¨la GalNAc3-3.-0,AdoAesGesmCesTesTesmCdsTdsrrdsGdsrrdsmCds Ga1NAc3-3a Ad 36 mCdsAdsGdsmCdsTesTes TesAesTe GalNAC3-7,-0'AdoAesGesmCesTesTesmCdsrrdsrrdsGdsrrdsmCds Ga1NAc3-7a Ad 36 mCdsAdsGdsmCdsTesTes TesAesTe GalNAc3-10am'AdoAesGesmCesTesTesmCdsrrdsrrdsGdsTasmCds GalNAc3-10 a Ad mCdsAdsGdsmCdsTesTes TesAesTe GalNAc3-13 am'AdoAesGesmCesTesTesmCdsrrdsrrdsGdsTasmCds 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 Isubcutaneously once with an oligonucleotide listed in Table ---4-57 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.
1 Table 7458 Plasma triglyceride and APOC-III protein levels in transgenic mice Time pointAPOC-III
ISIS Dosage TriglyceridesGalNAc3 CM
(days post- ne protein ( /0 No. (mg/kg) (% baseline) Cluster dose) baseli) 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 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 Table7--59 below were tested in a study for dose-dependent inhibition of Al AT in mice.
Table 7259 Modified ASOs targeting AlAT
ISIS Ga1NAc3 SEQ ID
Sequences (5' to 3') CM
No. Cluster No.
AesmCesmCesmCesAesAdsTdsTdsmCdsAdsGdsAdsAdsGdsGdsAesAes 476366 n/a n/a GesGesAe AesmCesmCesmCesAesAdsTdsTdsmCdsAdsGdsAdsAdsGdsGdsAesAes Ga1NAc3-la Ad 38 GesGesAeoAdo'-Ga1NAc3-18 GalNAc3-3a Ga1NAc3-3a Ad 39 AdsGdsGdsAesAes GesGesAe GalNAC3-7a Ga1NAc3-7a Ad 39 AdsGdsGdsAesAes GesGesAe GalNAc3-10am'AdoAesmCesmCesmCesAesAdsTdsTdsmCdsAdsGds Ga1NAc3-10 a Ad 39 AdsAdsGdsGdsAesAes GesGesAe GalNAc3-13 am'AdoAesmCesmCesmCesAesAdsTdsTdsmCdsAdsGds GalNAc3-13a Ad 39 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 74-59 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 7460, 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 =7460 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 ';'-4-01 below.

Body weight is shown as % relative to baseline. Organ weights are shown as %
of body weight relative to the PBS control group.
1 Table 7461 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 0.6 29 57 40 123 100 103 0.6 26 57 32 117 93 109 0.6 26 42 35 114 100 103 0.6 30 67 38 121 91 100 0.6 36 63 31 118 98 103 Example 81: Duration of action in vivo of oligonucleotides targeting AlAT
comprising a GaINAc3 cluster 1 The oligonucleotides listed in Table :=-2-59 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 Ilisted in Table =24-59 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 ';'-S62 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 -14---63 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).
1 Table 463 Inhibition of SRB-1 expression in vitro ISISGalNAc IC50 SEQ
Sequence (5' to 3') Linkages CM
No. cluster (nM) ID No.
m m m Ges CesTesTes CesAdsGdsTdsCd Ad Id Gd Ad s353382m m m PS n/a n/a 250 28 CdsTdsTes Ces CesTesTe GesmCesTesTesmCesAd Gd I'd mCd Ad I'd Gd Ad GalNAc3 655861 sss sssss PS Ad 40 29 mCdsTdsTesmCesmCesTesTeoAdo,-GalNAC3-1 a -1a m m GalNAc3-3a-0,AdoGes CesTesTes CesAdsGdsTds GalNAc3 661161 m m m m PS Ad 40 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -3a m m GalNAc3-3a-0,AGes CeeTeerree Ce0AdsGd Ta Ga1NAc3 661162 m m m mdo s s PO/PS Ad 8 CdsAdsTdsGdsAds CdsTds Teo Ceo CesTesTe -3a GesmCesTesTesmCesAdsGdsTdaniCasAdaTdaGasAds Ga1NAc3 664078 PS Ad 20 29 mCdsTdsTesmCesmCesTesTeoAdo¨GalNAc3-9a -9a GalNAC3-8a GalNAc3 665001 PS Ad 70 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -8a GalNAC3-5a-0,AdoGesmCesTesTesmCesAdsGdsTds Ga1NAc3 666224 PS Ad 80 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -5a m m m Ges CeoTeoTeo CesAdsGdsTdsCd Ad Td Gd Ad 666841 sssss PO/PS
m m m n/a n/a >250 28 CdsTds Teo Ceo CesTesTe GalNAc3-10am,AdoGesmCesTesTesmCesAdsGdsTds Ga1NAc3 666881 PS Ad 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -10a m m m GalNAc3-3a-,Ges CesTesTes CesAdsGdsTds Cd GalNAc3 666904 m m m0 s r, 5 PO 9 AdsTdsGdsAds CdsTds Tes Ces CesTesTe -3a m m GalNAC3-3a CesTesTes CesAdsGdsTds GalNAc3 666924 m m m m PS Td 15 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -3a GalNAC3-6a GalNAc3 666961 PS Ad 150 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -6a GalNAc3-7a-0,AdoGesmCesTesTesmCesAdsGdsTds Ga1NAc3 666981 PS Ad 20 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -7a m m GalNAC3-13a CesTesTes CesAdsGdsTdsGalNAc3 670061 m m m mdo r, s Ad 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -13a m m Ga1NAc3-3a-0,TdoG C T T C A G T Ga1NAc3 670699 m es eo eo eo eo ds ds ds m m m PO/PS Td 15 CdsAdsTdsGdsAds CdsTdsTeo Ceo CesTesT -3a e m m Ga1NAC3-3am,AeoG CT T CAGT GalNAc3 670700 m es eo eo eo eo ds ds ds m m m PO/PS Ae 30 CdsAdsTdsGdsAds CdsTdsTeo Ceo CesT 3a esT -m m Ga1NAC3-3am,Te0G CT T CAGT GalNAc3 670701 m es eo eo eo eo ds ds ds m m m PO/PS Te 25 CdsAdsTdsGdsAds CdsTdsTeo Ceo CesTesT -3a e 671144 Ga1NAc3-12a-0,AdoGesmCesTesTesmCesAdsGdsTds PS Ga1NAc3 Ad 40 30 m m m m -12a CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe m m Ga1NAc3-139-0,AdoG C T T C A G T GalNAc3 671165 m m es eo eo eo eo ds ds ds m m PO/PS Ad CdsAdsTdsGdsAds CdsTdsTeo Ceo CesT 13a es-m m GalNAc3-14a-0,AdoGes CesTesTes CesAdsGdsTds GalNAc3 671261 m m m m PS Ad >250 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -14a m m GalNAc3-15a-0,AdoGes CesTesTes CesAdsGdsTds GalNAc3 671262 m m m m PS Ad >250 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -15a m m GalNAc3-7a-0,AGes CeoTeoTeo CeoAdsGdsTds GalNAc3 do673501 m m m m PO/PS Ad 30 30 CdsAdsTdsGdsAds CdsTdsTeo Ceo CesTesTe -7a m m GalNAC3-10a-0,AGes CeoTeoTeo CeoAdsGdsTds GalNAc3 do673502 m m m m PO/PS Ad 8 30 CdsAdsTdsGdsAds CdsTds Teo Ceo CesTesTe -10a m m GalNAc3-17a-0,AGes CesTesTes CesAdsGdsTds GalNAc3 do675441 m m m m PS Ad 30 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -17a m m GalNAC3-18a-0,AGes CesTesTes CesAdsGdsTds GalNAc3 do675442 m m m m PS Ad 20 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -18a GeamCeaTeaTeamCesAdaGdsTdsmCdsAdsTdsGdsAds GalNAc3 Ad 40 29 mCdaTdaTeamCeamCeaTearreoAdo¨GalNAc3-19a -19a GeamCeaTeaTeamCesAdaGdsTdsmCdsAdsTdsGdsAds Ga1NAc3 Ad 30 29 mCdaTdaTeamCeamCearrearreoAdo¨GalNAc3-20a -20a m m GalNAc3-23a-0,AGes CesTesTes CesAdsGdsTds GalNAc3 do677843 m m m m PS Ad 40 30 CdsAdsTdsGdsAds 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 GaINAc3 cluster 1 The oligonucleotides listed in Table 7:=2-64 below were tested in a study for dose-dependent inhibition of Factor XI in mice.
1 Table 7:764 Modified oligonucleotides targeting Factor XI
ISISGalNAc SEQ
Sequence (5' to 3') CM
No. cluster ID No.
TeaGeaGeaTesAesAdaTdamCdamCdsAdsmCdsTdsTdsTdsmCdsAesGes 404071 n/a n/a 31 AesGesGe TeaGeoGeorrecAeoAdsTdsmCdsmCdaAdsmCdsTdsTdsTdsmCdsAeoGeo 656173 Ga1NAc3-la Ad 32 AesGesGeoAdo,-GalNAc3-la GalNAc3-3a-0,AdoTesGeoGeoTeoAeoAdsTdsmCdsmCdsAdsmCdsTds 663086 Ga1NAc3-3a Ad 40 TdsTdsmCdsAeoGeoAesGesGe GalNAC3-7a-0,AdoTesGeoGeoTeoAeoAdsTdsmCdsmCdsAdsmCdsTds 678347 Ga1NAc3-7a Ad 40 TdsTdsmCdsAeoGeoAesGesGe GalNAc3-10a-0,AdoTesGeoGeoTeoAeoAdsTdsmCdsmCdsAdsmCds 678348 Ga1NAc3-10a Ad 40 TdsTdsTdsmCdsAeoGeoAesGesGe GalNAc3-13a-0,AdoTesGeoGeoTeoAeoAdsTdsmCdsmCdsAdsmCds Ga1NAc3-13a Ad 40 TdsTdsTdsmCdsAeoGeoAesGesGe 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 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.
1 As illustrated in Table -744,65, 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).
1 Table 7$65 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 6 3 50 63 25 0.15 0.7 33 91 169 25 0.16 2 7 38 55 21 0.16 Ga1NAc3-3a 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 6 1 44 76 19 0.15 678348 0.7 39 43 54 21 0.16 Ga1NAc3-10 a 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 Ga1NAc3-13a 6 2 28 41 20 0.14 Example 84: Duration of action in vivo of oligonucleotides targeting Factor XI
comprising a GaINAc3 Conjugate The oligonucleotides listed in Table 721-64 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 1 Table ";-7-64 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).
1 Table '7466 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 31 656173 6 10 3 Ga1NAc3-la Ad 663086 6 10 2 Ga1NAc3-3a Ad 678347 6 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--63 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 743-63 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 44-67 and =',-1-68, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner.
Table S4167 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 670061 0.1 118 Ga1NAc3-13a Ad 0.3 89 0.1 119 0.3 96 677842 Ga1NAc3-20a Ad 1 Table 8468 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 Q-69 below.
Table ;',?,,,69 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 0.3 29 56 0.15 27 118 655861 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 0.3 42 61 0.15 23 117 661161 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 0.3 49 82 0.16 25 119 666881 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 Ga1NAc3-20a Ad 0.3 31 46 0.17 21 117 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 8,3-70 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 Tables72474.
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 A-71-87 74 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 "BL" indicates baseline, measurements that were taken just prior to the first dose. As illustrated in Tables ____________________________________________________ and 72, 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 :WIO
Oligonucleotides targeting human TTR
GalNAc SEQ
Isis No. Sequence 5 to 3' Linkages CM
cluster ID No.
TesmCesTesTesGesGdsTdsTdsAdsmCdsAdsTdsGdsAdsAds 420915 P S n/a n/a 41 AesTesmCesmCesmCe TesmCesTesTesGesGdsTdsTdsAdsmCdsAdsTdsGdsAdsAds 660261 PS Ga1NAc3-1a Ad 42 AesTesmCesmCesmCeoAdo'-GalNAc3-1, GalNAc3-3a,,,TesmCeorreorreoGeoGasTasTasAdsmCdsAds 682883 PS/P0 Ga1NAc3-3a PO 74 TdsGdsAdsAdsAeorreomCesmCesmCe GalNAc3-7a,,,TesmCeorreorreoGeoGasTdsTasAdsmCdsAds 682884 PS/P0 Ga1NAc3-7a PO 41 TdsGdsAdsAdsAeoTeomCesmCesmCe GalNAc3-10a_0,TesmCeoTeoTeoGeoGdsTdsTdsAdsmCds 682885 PS/PO Ga1NAc3-10a PO 41 AdsTdsGdsAdsAdsAeoTeomCesmCesmCe GalNAc3-13a_0,TesmCeoTeoTeoGeoGdsTdsTdsAdsmCds 682886 PS/P0 Ga1NAc3-13a PO 41 AdsTdsGdsAdsAdsAeoTeomCesmCesmCe TesmCeoTeoTeoGeoGdsTdsTdsAdsmCdsAdsTdsGdsAdsAds 684057 PS/P0 Ga1NAc3-19a Ad 42 AeoTeomCesmCesmCeoAdo,-GalNAc3-19a 1 The legend for Table ,s.,:4--72 can be found in Example 74. The structure of GalNAc3-1 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 GalNAc3-10a was shown in Example 46. The structure of GalNAc3-13a was shown in Example 62. The structure of GalNAc3-19a was shown in Example 70.
1 Table S471 Antisense inhibition of human TTR in vivo Dosage TTR mRNA (% Plasma TTR protein GalNAc cluster CM SEQ
Isis No.
(mg/kg) PBS) (% PBS) ID No.
PBS n/a 100 100 n/a n/a 420915 20 48 65 n/a n/a 41 0.6 113 87 Ga1NAc3-la Ad 42 1 Table &Sin 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) (% PBS) BL Day 3 Day 10 (After sac) cluster No.
PBS n/a 100 100 96 90 114 n/a n/a n/a n/a 41 0.6 60 88 73 63 68 GalNAc3- PO

3a 0.6 56 88 78 63 67 Ga1NAc3- PO

7a 0.6 60 92 77 68 76 GalNAc3- PO

10a 0.6 57 91 70 64 69 682886 2 21 89 50 31 30 Ga1NAc3-6 18 102 41 24 27 a 0.6 53 80 69 56 62 684057 2 21 92 55 34 30 Ga1NAc3-Ad 42 6 11 82 50 18 13 19a Table 8b73 Transaminase levels, body weight changes, and relative organ weights Dos ALT (U/L) AST (U/L) Body Liver Spleen Kidne SEQ
age Isis No. ( BL BL Day Day Day Day Day Day (% (%
(% Y (% ID nig /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 :It?,-.7-74 Transaminase levels, body weight changes, and relative organ weights Dos ALT (U/L) AST (U/L) Body Liver Spleen Kidne SEQ
age Isis No. ( BL BL Day Day Day Day Day Day (% (% (% Y (%
ID nig /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 -.-1,70) were tested in a single dose study for duration of action in mice. ISIS numbers 420915, 682883, and 682885 (see Table 8470) 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 8875 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 660261 13.5 Ga1NAc3-la Ad 42 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 8Q76 Plasma TTR protein levels ISIS Dosage Time point GalNAc3 CM
TTR (% baseline) SEQ ID No.
No. (mg/kg) (days post-dose) Cluster 420915 100 10 48 n/a n/a 682883 10.0 10 38 Ga1NAc3-3a PO 41 682885 10.0 10 34 Ga1NAc3-10 a PO

The results in Tables 88 75 and -89 76 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 1,19-77 were tested for splicing modulation of human survival of motor neuron (SMN) in mice.
Table Q-077 Modified ASOs targeting SMN
ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
AesTesTesmCesAesmCesTesTesTesmCesAesTesAesAesTesGesmCesTesGes 387954 n/a n/a Ge GalNAc3-7 A.
sTesTesmCesAsmCesTesTesTesmCesAsTesAesAes699819 Ga1NAc3-7a PO 43 TesGesmCesTesGesGe GaiNAC3-7a Ga1NAc3-7a PO 43 AeoTeoGeomCeoTesGesGe AesTesTesmCesk,,mCesTesTesTesmCesAesTesAesAesTesGesmCesTesGes Ga1NAc3-la Ad 44 GeoAdo=¨Ga1NAc3-1a 703421 X-ATTmCAmCTTTmCATAATGmCTGG
n/a n/a 43 703422 GalNAc3-7b-X-ATTITAITTTTITATAATGITTGG Ga1NAc3-7b n/a 43 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:
HOOH
HO
AcHN

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 94-78 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 i4-1-78. 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 44-78 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 387954 288 5.00 n/a n/a 699819 32 7.84 Ga1NAc3-7a PO 43 699821 32 7.22 Ga1NAc3-7a PO 43 700000 32 6.91 Ga1NAc3-la Ad 44 703421 32 1.27 n/a n/a 703422 32 4.12 Ga1NAc3-76 n/a 43 Example 89: Antisense inhibition in vivo by oligonucleotides targeting Apolipoprotein A (Apo(a)) comprising a Ga1NAc3 conjugate The oligonucleotides listed in Table below were tested in a study for dose-dependent inhibition of Apo(a) in transgenic mice.
Table 4279 Modified ASOs targeting Apo(a) ISISGalNAc3 SEQ ID
Sequences (5' to 3') CM
No. Cluster No.
es "-esmCes mCdsGdsTdaTdsGdsGdsTdaGdsmC
Tes es ds 494372 n/a n/a 53 TdaTesGesTesTesmCe GalNAc3-7a-0,TesGeomCeoTeoniCeoniCdsGdsTdsTdsGdsGds 681257 Ga1NAc3-7a PO 53 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 1,^-2-79 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 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 0481.
As illustrated in Table i43,80, 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 4481, transaminase levels and body weights were unaffected by the oligonucleotides, indicating that the oligonucleotides were well tolerated.
Table t;g80 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 PBS n/a 100 100 120 119 113 88 121 0.3 75 79 76 89 98 71 94 1 Table 4481 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 GaINAc3 5 cluster 1 Oligonucleotides listed in Table 05 82 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 1 three doses, with an oligonucleotide and dosage listed in Table 143-83 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 1 standard protocols to determine liver human TTR mRNA levels. The results presented in Table '43--83 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 1 Table 04383, 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 q-S82 Oligonucleotides targeting human TTR
GalNAc SEQ
Isis No. Sequence 5' to 3' Linkages CM
cluster ID No.
TesmCesTesTesGesGd Td Td Ad mCd Ad I'd Gd Ad Ad 420915 sss s s ss s s s PS n/a n/a 41 AesTesmCesmCesmCe GalNAc3-3.,,TesmCeoreoreoGeoGdsTdsTdsAdsmCd Ad s682883 PS/PO Ga1NAc3-3a PO 41 TdaGdaAdsAdsAeoTeomCesmCesmCe GalNAc3-3.,,,AdoTesmCeorreoTeoGeoGasTa Ta Ad 666943 s s s PS/PO
GalNAc3-3a Ad 45 mCdaAdaTdaGdaAdaAds AeoTeomCesmCesmCe GalNAC3-7,-0,AdoTesmCeoTeoTeoGeoGdsTd Ta Ad 682887 s s s PS/PO
GalNAc3-7a Ad 45 mCdaAdaTdsGdsAdsAdsAeoTeomCesmCesmCe GalNAc3-10,,,,AdorresmCeorreorreoGeoGdsTd Ta Ad 682888 s s s P S/P 0 GalNAc3-10 a Ad 45 mCdaAdaTdsGdsAdsAdsAeoTeomCesmCesmCe GalNAc3-13._0,AdoTesmCeorreorreoGeoGdsTdsTdsAds GalNAc3-13a Ad 45 mCdaAdaTdsGdsAdsAdsAeoTeomCesmCesmCe 1 The legend for Table 4:c-82 can be found in Example 74. The structure of GalNAc3-3a was shown in Example 39. The structure of GalNAc3-7a was shown in Example 48. The structure of GalNAc3-10a was shown in Example 46. The structure of GalNAc3-13a was shown in Example 62.
1 Table 400 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 Ga1NAc3-3a PO

0.6 74 93 Ga1NAc3-3a Ad 0.6 60 97 Ga1NAc3-7a Ad 0.6 65 92 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 t:-4-84 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 94-84 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 9g-85 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 9g85., 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 -97-84 Oligonucleotides targeting Factor VII
GalNAc SEQ
Isis No. Sequence 5 to 3' Linkages CM
cluster ID No.
AesTesGesmCesAesTdsGdsGdsTdsGdsAdsTdsGdsmCdsTds 407935 PS n/a n/a TesmCesTesGesAe GalNAc3-10._0,AesTesGesmCesAesTdsGdsGdsTdsGds 686892 P S GalNAc3-10a PO 46 ikdsTdsGdsmCdsTds TeamCeaTesGesAe The legend for Table L'7 84 can be found in Example 74. The structure of GalNAc3-10a was shown in Example 46.
Table 9885 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 '4986, 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 4486 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 47 niCesAesGesniCesTesTdsrfdsAdsrfdsrfdsAdsGdsGdsGdsAdsmCes 661180 Ad 1.40 48 AesGesmCesAeo Ado' -GalNAc3-la GalNAC3-3a-o,mCesAesGesmCesTesTdsTasAdsrrdsrrdSAdSGdSGdSGdSAdSmCes 680771 PO 0.70 47 /kesGesmCesAe GalNAc3-7a-o,mCesAesGesmCesTesT dsrr dsAdsrf dsrr dSAdSGdSGdsGd Ad m-a 680772 s s PO 1.70 47 /kesGesmCesAe GalNAc3-10a-o,mCesAesGesmCesTesTdSrfdSAdST dST dSAdSGdSGasGasAdsmCes 680773 PO 2.00 47 /kesGesmCesAe GalNAc3-13a-o,mCesAesGesmCesTesTdSrfdSAdST dST dSAdSGdSGdSGa Ad m-a 680774 s s PO 1.50 47 /kesGesmCesAe GalNA C3-3 a-o' dsrrdSAdSGdSGdsGd Ad m-a 681272 s s PO < 0.46 47 AeoGesmCesAe GalNA C3-3 am,AdomCesAesGesmCesTesT dsr-f dsAdsrf dsrrdsAds GdsGdsGdsAds Ad 681273 1.10 49 mCesAesGesmCesAe mCesAesGesmCesTesrr dST dAdST dST dsAds Gds Gds GdsAdsmC es 683733 Ad 2.50 48 AesGesmCesAeoAdo'-GalNAc3-19a The structure of Ga1NAc3-11 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 -I-40-87 were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.

Table i(-t,87 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') Ga1NAc3 CM
SEQ
No. Cluster ID
No.
449093 TkaTkamCkaAdaGdaTdamCda AdaTda Gds AdsmCdsTasTIsmCksmCk n/a n/a 50 699806 GaINAc3-3a-0,TI,TIsmCksAdsGasTasmCds AdsTds GdsAdsmCds Ga1NAc3-3 a PO
m-TdsTIsksmCk 699807 GaINAc3-7a-0,TI,TIsmCksAdsGasTasmCds AdsTds GdsAdsmCds Ga1NAc3-7a PO
m-TdsTIsksmCk 699809 GaINAc3-7a-o, TiOlsmCksAdsGasTasmCds AdsTds Gds AdsmCds Ga1NAc3-7a PO
m-TdaTeaesmCe 699811 GaINAc3-7 = a-0,TesTesmCesAdsGasTasmCds AdsTas GasAdsmCds Ga1NAc3-7a PO
m TdsTIsksmCk 699813 GaINAc3- 7 = a-o'rrksTasmCksAdsGasTasmCds AdsTas GasAdsmCds Ga1NAc3-7a PO
m-TdsTIsdsmCk 699815 GaINAc3-7a-0,TesTIsmCksAdsGdsTdsmCds AdsTds GdsAdsmCds Ga1NAc3-7a PO
m TdsTIsksmCe 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-5 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 10 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 -1-00-87 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 15 average percent of SRB-1 mRNA levels for each treatment group relative to the saline control group. As illustrated in Table 444-88, 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.
20 Body weights, liver transaminases, total bilirubin, and BUN were also measured, and the average values for each treatment group are shown in Table 0188. Body weight is shown as the average percent body weight relative to the baseline body weight (% BL) measured just prior to the oligonucleotide dose.

1 Table 4188 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 1 111 18 48 0.17 31 449093 3 94 20 43 0.15 26 36 19 50 0.12 29 104 0.1 114 23 58 0.13 26 699806 0.3 59 21 45 0.12 27 1 25 30 61 0.12 30 0.1 121 19 41 0.14 25 699807 0.3 73 23 56 0.13 26 1 24 22 69 0.14 25 0.1 125 23 57 0.14 26 699809 0.3 70 20 49 0.10 25 1 33 34 62 0.17 25 0.1 123 48 77 0.14 24 699811 0.3 94 20 45 0.13 25 1 66 57 104 0.14 24 0.1 95 20 58 0.13 28 699813 0.3 98 22 61 0.17 28 1 49 19 47 0.11 27 0.1 93 30 79 0.17 25 699815 0.3 64 30 61 0.12 26 1 24 18 41 0.14 25 Example 94: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising 2'-sugar 5 modifications and a 5'-GaINAc3 conjugate 1 The oligonucleotides listed in Table 142 89 were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.
1 Table -I 0-289 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') Ga1NAc3 CM
SEQ
No. Cluster ID No.
353382 GesmCesTesTesmCesikasGasTasmCdsAds'fdsGdsAdsmCdsTdsTesmCeamCes n/a n/a TesTe 700989 GmsCinsUmsUmsCmsAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsUmsCmsCms 1-ila n/a UmsUm 666904 Ga1NAc3-3a-0,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 403 90 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 -W90 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 4-04 -91 were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.
Table 14491 Modified ASOs targeting SRB-1 ISIS, SEQ
Sequences (5' to 3') CM
No. Cluster ID No 440762 TkamCkaAdaGdaTdamCdaAdaTdaGdaAdsmCdsTasTksmCk n/a n/a 22 666905 Ga1NAc3-38-0,TIsmCk5Ad5Gd5Td5mCd5Ad5Td5Gd5Ad5mCd5Td5TIsmCk Ga1NAc3-3a 699782 Ga1NAc3-78-0,TIsmCk5Ad5Gd5Td5mCd5Ad5Td5Gd5Ad5mCd5Td5TIsmCk Ga1NAc3-7a 699783 Ga1NAc3-3,-0,TismCisAdsGasTasmCdsAdsTdsGdsAdsmCdsTdsTismCI Ga1NAc3-3a 653621 TiamCisAdsGasTasmCdsAdsTdsGdsAdsmCdsTdsrrlsmCloAdo,-GalNAC3-18 Ga1NAc3-1a Ad 23 439879 TgamCgaAdaGdaTdamCdaAdaTd GdaAdamCdaTdaTgamCg n/a n/a 22 699789 Ga1NAc3-3a-0,TgsmC
gsAdsGdsTdsmCdsAdsTa GdsAdsmCdsTdsTgsmC g Ga1NAc3-3a PO 22 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 V5 92 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 14,:kiK92 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 74-57 targeting ApoC-III and oligonucleotides in Table 106-93 targeting Apo(a) were tested in an ultra-filtration assay in order to assess plasma protein binding.

Table 1413 Modified oligonucleotides targeting Apo(a) ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No TesGesmCesTesmCesmCdsGdsTdsTdsGdsGdsTdsGdsmCdsTdsTesGesTes 494372 n/a n/a 53 TesmCe TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGdsTdsGdsmCdsTdsTeoGeoTes 693401 n/a n/a 53 TesmCe GalNAC3-7a-o'TesGesmCesTesmCesmCdsGdsTdsTdsGdsGdsTdsGdsmCds GalNAc3-7a PO

TdsTesGesTesTesmCe GalNAC3-7a-o'TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGdsTdsGdsmCds GalNAc3-7a PO

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 -7-4-57 and -i-00 93 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 10 and 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 --Ã7----94 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 -94 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 1 The oligonucleotides shown in Table 446-95_comprising a GalNAc conjugate were designed to target TTR.
1 Table -895 Modified oligonucleotides targeting TTR
Ga1NAc3 SEQ ID
ISIS No. Sequences (5' to 3') CM
Cluster No GalNAc3-3a,,,Ado Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds 666941 GalNAc3-3 Ad 45 Ads Tds Gds Ads Ads Aes Tes mCes mCes mCe Tes mCeo Teo Teo Geo Gds Tds Tds Ads mCds Ads Tds Gds Ads Ads 666942 r, r, r, A
GalNAc3-1 Ad 42 -r-leo leo mk-es mk-es mk-eo 1-ido'-3a-3a GalNAc3-3a_0,Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ads Tds GalNAc3-3 Gds Ads Ads Aes Tes mCes mCes mCe GalNAc3-7a_0,Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ads Tds 682877 Ga1NAc3-7 PO 41 Gds Ads Ads Aes Tes mCes mCes mCe Ga1NAC3-10a_0,Tes mCes Tes Tes Ges Gds Tds Tds Ads mCdsAds GalNAc3-10 PO

Tds Gds Ads Ads Aes Tes mCes mCes mCe GalNAC3-13a_0,Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ads 682879 Ga1NAc3-13 PO 41 Tds Gds Ads Ads Aes Tes mCes mCes mCe GalNAC3-7a-0,Ado Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds 682880 Ga1NAc3-7 Ad 45 Ads Tds Gds Ads Ads Aes Tes mCes mCes mCe GalNAC3-10a_0,Ado Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds 682881 Ga1NAc3-10 Ad 45 Ads Tds Gds Ads Ads Aes Tes mCes mCes mCe GalNAC3-13a_0,Ado Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds 682882 Ga1NAc3-13 Ad 45 Ads Tds Gds Ads Ads Aes Tes mCes mCes mCe Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ads Tds Gds Ads Ads 684056 r, r, ,-, A
Ga1NAc3-19 Ad 42 i-les es mk-es mk-es mk-eo 1-ido'-GalNAc3-19a 1 The legend for Table 4--0,5 can be found in Example 74. The structure of GalNAc3-1 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 449-96 and were tested for pro-inflammatory effects in an hPMBC assay as described in Examples 23 and 24. (See Tables 3-017, K.470, 9-J-82, and 1-48----95 for descriptions of the oligonucleotides.) ISIS 353512 is a high responder used as a positive control, and the other oligonucleotides are described in Tables >;370, 91582, and k):95. The results shown in Table *9-96 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 149%
ISIS No. Emax/ECso 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 -I-4-9-97 (see Table 7-6-63 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 counter. Competition experiments utilizing 1251 -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 125I -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 125I 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 -97 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 4-1497 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 :14-1a 98a below were tested in a single dose study for duration of action in mice.
Table 4-1-1,:198a Modified ASOs targeting APO(a) ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID
No.
GalNAc3-7a-0,TeaGeamCeaTeamCeamCdsGasTasTasGasGas 681251 Ga1NAc3-7a PO 53 TdsGdsmCdsTdsTesGes TesTesmCe GalNAC3-7a-o'TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds 681257 Ga1NAc3-7a PO 53 TdsGdsmCdsTdsTeoGeo TesTesmCe 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 -1--14-98b 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 -4-1-lb 98b 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 -,1-44-1498b Apo(a) plasma protein levels Apo(a) at 72 hours Apo(a) at 1 week Apo(a) at 3 weeks ISIS No. Dosage (mg/kg) (% BL) (% BL) (% BL) PBS n/a 116 104 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 6812 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 -1-12 99 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 -1-4-2-99 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 ,L1-2--..99 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 4409 Modified oligonucleotides targeting mouse APOC-III
Dosage APOC-III
ISIS
SEQ
N Sequences (5' to 3') CM (mg/kg) mRNA (%
No.
ID
o.
PBS) mCesAesGesmCesTesTdsrrdsAdsrrdsrrdsAds 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 Ga1NAc3-7a_s,mCesAesGesmCesTesTdsrrdsAdsrrds n/a (PS 2 73 ) 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 that does not comprise a GalNAc conjugate and ISIS 655861 (see Table -,1-423) 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 1-24100. 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 __ 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-WHO
Concentration in whole Concentration in Concentration in non-ISIS Dosage liver (molecules*10^
N 6 hepatocytes parenchymal liver cells o. 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 5 conjugate The oligonucleotides listed in Table 444101 below were tested in a single dose study for duration of action in mice.
Table 14101 Modified ASOs targeting APOC-III
ISIS Sequences (5' to 3') Ga1NAc3 CM
SEQ
No. Cluster ID No.
304801 AesGesmCesTesTesmCdsTdsTdsGdsTdsmCdsmCdsAdsGdsmCdsTesTes n/a n/a TesAesTe 663084 Ga1NAc3-3.-0,AdoAesGeomCeoTeoTeomCdsTdsTdsGdsTdsmCds Ga1NAc3-3a Ad 36 mCdsAdsGdsmCdsTeoTeo rresAesTe 679241 AesGeomCeoTeoTeomCdsTdsTdsGdsTdsmCdsmCdsAdsGdsmCdsTeoTeo Ga1NAc3-19a Ad TesAesTeoAdo'-GalNAc3-19.
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 -1-1-4-101 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 -1-4-5-102 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 8 of example 79 with the results in Table 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.
1 Table 1-1-S102 Plasma triglyceride and APOC-III protein levels in transgenic mice Time point APOC-III
ISIS Dosage Triglycerides Ga1NAc3 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 GalNAc3-Ad 19a Example 104: Synthesis of oligonucleotides comprising a 5'-Ga1NAc2 conjugate HN,Boc HN-Bc)c Boc\.N OH H2No 0 ..
H HBTU, HOBt DIEA, DMF 0 ___________________________________________ )10"- Boc.N EN11.....õ..".õ.õ--,..õ.11..o 0 H TFA

DCM

120 126 85% 231 HN Ed 0 +

AcO-L-:-D-\--0......õko 411) F DIEA
-70.-0 101 AcHN F DMF

OAc/..- OAc 0 OAc OAc ----Z-0, Q
AcHN NH ..24, AcHN NH
1 H2, Pd/C, Me0H
_____________________________________________________________________________ )11.-F
OAc 11.-- OAc 2 PFPTFA , DMF OAc OAc 0 H 0 lk-----4-.0 0\
F F
0r H 0 AcHN AcHN
F

F

0 83e OHOH
3' 5', 11 --0 0 ( OLIGO j-0-P-0-(CH2)6-NH2 __ HO
01H AcHN
1 Borate buffer, DMSO, pH 8.5, rt OHOH
HO /
2 aq ammonia, ...A. N
rt AcHN N

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

AcHN NH
OH OH

AcHN
Example 105: Synthesis of oligonucleotides comprising a Ga1NAc1-25 conjugate 0 83e 3'5') 11 OAcOAc F
OLIGO O¨P-0¨(CH2)6-NH2 AcO01 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 OH O
OAc Ac Ac0 PFPTFA __________________ Ac0 OH
NHAc OH NHAc TEA, Acetonitrile OA OAc tetrazole, 1-Methylimidazole, DMF

Ac0 H
2-cyanoethyltetraisopropyl phosphorodiamidite NHAc (!) LCN
Oligonucleotide OH OH
synthesis HO o 0 _____________ OP-AcHN CM ) OLIGO
N " 0 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

4)01::43 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 446-103 and 1 I 7 104 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 446-103 and 4-4-;1104. 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 4-443-103 and 1-4-7-104.
Table 1-143103 Modified oligonucleotides targeting SRB-1 ISIS,ED50 SEQ
Sequences (5 to 3') GalNAc Cluster No.
(mg/kg) ID No 440762 TkamCkaAdaGdaTdsmCdsAdsr-f dsGdsAdsmCdsTdsTksmCk n/a 4.7 22 GalNAc2-249-0,AdoTkamCksAdsGasTasmCdsAdsrrdsGasAds 686221 Ga1NAc2-24a 0.39 26 mCdsTdsTksmCk GalNAc3-139-0,AdoTIcsmCksAdsGdsTdsmCdsAdsTdsGdsAds 686222 Ga1NAc3-13a 0.41 26 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 -1--1-7-104 Modified oligonucleotides targeting SRB-1 ISIS,ED50 SEQ
Sequences (5 to 3') GalNAc Cluster No.
(mg/kg) ID No 440762 TkamCkaAdaGdaTdsmCdsAdsr-f dsGdsAdsmCdsTdsTksmCk n/a 5 22 GalNAci-259-0,TksmCksAdsGasTasmCdsAdsTasGasAds 708561 Ga1NAc1-25a 0.4 22 mCdaTdaTkamCk See Example 93 for table legend. The structure of GalNAci-25a was shown in Example 105.
The concentrations of the oligonucleotides in Tables 144-103 and 447-104 in liver were also assessed, using procedures described in Example 75. The results shown in Tables 4-44.+-104a and ---1-474--1041-) 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 43/4403 and 444-104 above.
Table 4-1-7-al 04a Liver concentrations of oligonucleotides comprising a Ga1NAc2 or Ga1NAc3 conjugate group Dosage ISIS No. [Antisense oligonucleotide] (m/g) GalNAc cluster CM
(mg/kg) 2 2.1 440762 7 13.1 n/a n/a 20 31.1 0.2 0.9 0.6 2.7 686221 2 12.0 Ga1NAc2-24a Ad 6 26.5 0.2 0.5 0.6 1.6 686222 Ga1NAc3-13a Ad 2 11.6 6 19.8 Table 1-1-7b104b 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 zt. CM Oligo 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 GalNAc1-26 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

HO
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.

AcHN
3' 04 CM Oligo The GalNAci cluster portion (GalNAci-27a) of the conjugate group Ga1NAc1-27 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:
OH

HO

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 48-105 below were tested in a single dose study in mice.
Table -i-1O5 Modified ASOs targeting APO(a) ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
TeaGeamCeaTeamCeamCdaGdaTdaTdsGdsGdsTasGasmCds 494372 n/a n/a 53 TdsTesGesTesTesmCe GaINAc3-7a-0,TeaGeamCesTesmCesmCdsGasTasTasGasGas GalNAc3-7a PO

TdsGdsmCdsTdsTesGes TesTesmCe GaINAc3-3a-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds 681255 Ga1NAc3-3a PO 53 TdsGdsmCdsTdsTeoGeo TesTesmCe GaINAc3-10a-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds 681256 Ga1NAc3-10a PO 53 TdsGdsmCdsTdsTeoGeo TesTesmCe GalNAC3-7a-o'TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds 681257 Ga1NAc3-7a PO 53 TdsGdsmCdsTdsTeoGeo TesTesmCe GaINAc3-13a-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds 681258 GalNAc3-13a PO 53 TdsGdsmCdsTdsTeoGeo TesTesmCe TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds TdsGdsmCdsTdsTeoGeo 681260 Ga1NAc3-19a Ad 52 TesTesmCeoAdo,-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 -44-106 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 106 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 -1--13406 Apo(a) plasma protein levels ISIS No. Dosage (mg/kg) Apo(a) at 1 week PBS n/a 143 Example 109: Synthesis of oligonucleotides comprising a Ga1NAc1-28 or Ga1NAc1-29 conjugate OH 5 __________________________________________________ 3 H CM 0 CM ¨ Oligo =
HO )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 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:
OH
0 .õ0 3 :
AcHN

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.
HO OH

pc HO o N-----.....--N?
AcHN H ____________________ , 3' 5' 242 0¨ CM ¨ Oligo ________________________________________ õ _____ , The GalNAci cluster portion (GalNAci-29a) of the conjugate group Ga1NAc1-29 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:
HO OH .,,OH
HOO
....._\0N --/./.....-N?
AcHN H
0 0¨ Efil Example 110: Synthesis of oligonucleotides comprising a Ga1NAc1-30 conjugate OAc OAc Ac04._ Ac0...r._...\

Ac0-t¨r--.1 -.) Ac0 OOTBDPS
TMSOTf AcHN
yO 243 1. NH3/Me0H ODMTr 2. DMTrCI Ac0...i......
1. TBAF
3. Ac20, pyr 0 Ac0 OOTBDPS 2.
Phosphitilation ________________ p- ____________________________________________ 0.-AcHN

ODMTr Ac0....r.....
1. Couple to 5'-end of ASO

Ac0 00,p,OCE ___________________________________ p-AcHN 1 2. Deprotect and purify ASO using 245 N(iP02 DMT-on purification methods OH
HO
0 5' 3' HO 00õ0,, . =
P Oligo AcHN '-0 Y -,246 Oligonucleotide 246 comprising a Ga1NAc1-30 conjugate group, wherein Y is selected from 0, S, a substituted or unsubstituted CI-CI 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 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
OCE Couple to 5'-end of ASO
2. Phosphitilation ¨OH ¨0-P
N(iPr)2 HO' 247 DMTrO248 Bx 1. Remove DMTr groups DMTrO
2. Couple amidite 245 ¨0õ0 ,X
3. Deprotect and purify ASO using DMTrO
Y 6-01igo DMT-on purification methods OH

HO 00õ0 ,P\
AcHN Y ¨0p\õ0 ____ Oligo 0--F( OH Y

HAcHN
Oligonucleotide 250 comprising a GalNAc2-31 conjugate group, wherein Y is selected from 0, S, a substituted or unsubstituted CI-CI alkyl, amino, substituted amino, azido, alkenyl or alkynyl, is synthesized as shown above. The Ga1NAc2 cluster portion (Ga1NAc2-31a) 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-OH

HO
,P\
AcHN y o-p, y 340 Ho/0 H AcHN

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 ______________________________________________________________________ 0-0, 4. Deprotect and purify ASO using HO DMTrO
,P¨NOP02 DMT-on purification methods OH
HO..____ HO-700p-0õ, , \ 0Y p. 3:
AcHN 0' y õ
¨000P.,0.4 Oligo _________________________________ , ii \

0-F<, OH HO\ ___7--/----/ O Y O

HO NHAc Oligonucleotide 252 comprising a Ga1NAc2-32 conjugate group, wherein Y is selected from 0, S, a substituted or unsubstituted CI-CI 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
HO/______\

HO 00õ0 ,P÷
AcHN 0' y ¨0,p-O0)\..
e 0 Y
0-pl, HO NHAc Example 112: Modified oligonucleotides comprising a GalNAci conjugate The oligonucleotides in Table 120-107 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 440107 GalNAc SEQ
ISIS No. Sequence (5' to 3') CM
cluster ID NO.
711461 Ga1NAc1-258_0,Ado Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds GalNAci-25a Ad 30 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 28 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 28 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 30 Gds Ads mCds Tds Tes mCes mCes Tes Te 711466 Ga1NAci-26a_0,Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds Gds GalNAci-26a PO 28 Ads mCds Tds Tes mces mCes Tes Te 711467 GalNAci-26a_0,Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds GalNAci-26a PO 28 Gds Ads mCds Tds Teo mCeo mCes Tes Te 711468 GalNAci-28a_0,Ado GesmCes Tes Tes mCes Ads Gds Tds mCds Ads Tds GalNAci-28a Ad 30 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 28 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 28 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 28 Tes mCes mCes Tes Te0,_GalNAci-27a 713845 Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-27a PO 28 Teo mCee mCes Tes Te0,_GalNAci-27a 713846 Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-27a Ad 29 Teo mCee 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 28 Tes mCes mCes Tes Te0,_GalNAci-29a 713848 Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-29a PO 28 Teo mCee mCes Tes Te0,_GalNAci-29a 713849 Ges mCes Tes Tes mCes Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-29a Ad 29 Tes mCes mCes Tes Teo Ado,_GalNAci-29a 713850 Ges mCeo Teo Teo mCeo Ads Gds Tds mCds Ads Tds Gds Ads mCds Tds GalNAci-29a Ad 29 Teo mCee mCes Tes Teo Ado,_GalNAci-29a

Claims (219)

CLAIMS:
1. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 1.
2. The compound of claim 1, wherein the nucleobase sequence of the modified oligonucleotide is complementary within nucleobases 3291-3310, 3290-3309, 3287-3306, or 3292-3311 of SEQ ID NO: 1, and wherein said modified oligonucleotide is at least 85%, 90%, 95%, or 100%
complementary to SEQ
ID NO: 1.
3. The compound of claim 1, wherein the modified oligonucleotide consists of 10 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 54, 55, 56, or 57.
4. The compound of claim 3, wherein the modified oligonucleotide has a nucleobase sequence comprising the sequence recited in SEQ ID NOs: 54, 55, 56, or 57.
5. The compound of claim 3, wherein the modified oligonucleotide has a nucleobase sequence consisting of the sequence recited in SEQ ID NOs: 54, 55, 56, or 57.
6. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 1.
7. The compound of claim 1, wherein the nucleobase sequence of the modified oligonucleotide is complementary within nucleobases 3291-3310, 3290-3309, 3287-3306, or 3292-3311 of SEQ ID NO: 1, and wherein said modified oligonucleotide is at least 85%, 90%, 95%, or 100%
complementary to SEQ
ID NO: 1.
8. The compound of claim 1, wherein the modified oligonucleotide consists of 10 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 54, 55, 56, or 57.
9. The compound of claim 3, wherein the modified oligonucleotide has a nucleobase sequence comprising the sequence recited in SEQ ID NOs: 54, 55, 56, or 57.
10. The compound of claim 3, wherein the modified oligonucleotide has a nucleobase sequence consisting of the sequence recited in SEQ ID NOs: 54, 55, 56, or 57.
11. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 3.
12. The compound of claim 11, wherein the nucleobase sequence of the modified oligonucleotide is complementary within nucleobases 192-211, 191-210, 193-212, 369-388, 370-389, 788-807, 790-808, or 2954-2973-of SEQ ID NO: 3, and wherein said modified oligonucleotide is at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 3.
13. The compound of claim 12, wherein the modified oligonucleotide consists of 10 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 58, 59, 60, 61, 62. 63, 64, or 65.
14. The compound of claim 12, wherein the modified oligonucleotide has a nucleobase sequence comprising the sequence recited in SEQ ID NOs: 58, 59, 60, 61, 62. 63, 64, or 65.
15. The compound of claim 12, wherein the modified oligonucleotide has a nucleobase sequence consisting of the sequence recited in SEQ ID NOs: 58, 59, 60, 61, 62. 63, 64, or 65.
16. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 7.
17. The compound of claim 16, wherein the nucleobase sequence of the modified oligonucleotide is complementary to target start sites 57825, 59956, 65940, 63577, 76224, 76229, 65938, 76225, 65938, 65939, 95513, or 76229 of SEQ ID NO: 7, and wherein said modified oligonucleotide is at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 7.
18. The compound of claim 16, wherein the modified oligonucleotide consists of 10 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, or 77.
19. The compound of claim 18, wherein the modified oligonucleotide has a nucleobase sequence comprising the sequence recited in SEQ ID NOs: 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, or 77.
20. The compound of claim 18, wherein the modified oligonucleotide has a nucleobase sequence consisting of the sequence recited in SEQ ID NOs: 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, or 77.
21. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 8.
22. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 9.
23. The compound of claim 21, wherein the nucleobase sequence of the modified oligonucleotide is complementary to target start sites 548 or 227 of SEQ ID NO: 8, and wherein said modified oligonucleotide is at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO:
8.
24. The compound of claim 22, wherein the nucleobase sequence of the modified oligonucleotide is complementary to target start sites 8133, 9804, 7270, 7295, 7319, 7344, 7368, 7392, 7416, 7440, 10718, 7267, 7292, 7316, 7341, 7365, 7389, or 7437 of SEQ ID NO: 9, and wherein said modified oligonucleotide is at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO:
9.
25. The compound of claim 21 or 22, wherein the modified oligonucleotide consists of 10 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 78, 79, 80, 81, 82, or 83.
26. The compound of claim 25, wherein the modified oligonucleotide has a nucleobase sequence comprising the sequence recited in SEQ ID NOs: 78, 79, 80, 81, 82, or 83.
27. The compound of claim 25, wherein the modified oligonucleotide has a nucleobase sequence consisting of the sequence recited in SEQ ID NOs: 78, 79, 80, 81, 82, or 83.
28. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 10.
29. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 11.
30. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 12.
31. The compound of claim 28-30, wherein the modified oligonucleotide consists of 10 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 84, 85, 86, 87, 88, 89, 90, 91, or 92.
32. The compound of claim 28-32, wherein the modified oligonucleotide has a nucleobase sequence comprising the sequence recited in SEQ ID NOs: 84, 85, 86, 87, 88, 89, 90, 91, or 92.
33. The compound of claim 28-32, wherein the modified oligonucleotide has a nucleobase sequence consisting of the sequence recited in SEQ ID NOs: 84, 85, 86, 87, 88, 89, 90, 91, or 92.
34. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 14.
35. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 15.
36. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 16.
37. A compound comprising a modified oligonucleotide and a conjugate group, wherein the modified oligonucleotide consists of 8 to 80 linked nucleosides and has a nucleobase sequence at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 17.
38. The compound of claim 35-37, wherein the modified oligonucleotide consists of 10 to 30 linked nucleosides and has a nucleobase sequence comprising at least 8 contiguous nucleobases of any of the nucleobase sequences of SEQ ID NOs: 84, 85, 86, 87, 88, 89, 90, 91, or 92.
39. The compound of claim 35-37, wherein the modified oligonucleotide has a nucleobase sequence comprising the sequence recited in SEQ ID NOs: 93, 94, 95, 96, 97, 98, 99, 100, or 101.
40. The compound of claim 35-37, wherein the modified oligonucleotide has a nucleobase sequence consisting of the sequence recited in SEQ ID NOs: 93, 94, 95, 96, 97, 98, 99, 100, or 101.
41. The compound of any of the preceding claims, wherein the modified oligonucleotide comprises at least one modified sugar.
42. The compound of claim 41, wherein the modified sugar is a bicyclic sugar.
43. The compound of claim 42, wherein the bicyclic sugar is selected from the group consisting of: 4'-(CH2)-O-2' (LNA); 4'-(CH2)2-O-2' (ENA); and 4'-CH(CH3)-O-2' (cEt).
44. The compound of claim 42, wherein the modified sugar is 2'-O-methoxyethyl.
45. The compound of any of the preceding claims, wherein the modified oligonucleotide comprises at least one modified nucleobase.
46. The compound of claim 45, wherein the modified nucleobase is a 5-methylcytosine.
47. The compound of any of the preceding claims, wherein the modified oligonucleotide consists of 17 linked nucleosides.
48. The compound of any of the preceding claims, wherein the modified oligonucleotide consists of 20 linked nucleosides.
49. The compound of any of the preceding claims, wherein the modified oligonucleotide comprises at least one modified sugar.
50. The compound of claim 49, wherein the modified sugar is a bicyclic sugar.
51. The compound of claim 50, wherein the bicyclic sugar is selected from the group consisting of: 4'-(CH2)-O-2' (LNA); 4'-(CH2)2-O-2' (ENA); and 4'-CH(CH3)-O-2' (cEt).
52. The compound of claim 49, wherein the modified sugar is 2'-O-methoxyethyl.
53. The compound of any of the preceding claims, wherein the modified oligonucleotide comprises at least one modified nucleobase.
54. The compound of claim 53, wherein the modified nucleobase is a 5-methylcytosine.
55. The compound of any of the preceding claims, wherein the compound is single-stranded.
56. The compound of any of the preceding claims, wherein the compound is double-stranded.
57. The compound of any of the preceding claims, wherein the modified oligonucleotide comprises at least one modified internucleoside linkage.
58. The compound of claim 57, wherein the modified internucleoside linkage is a phosphorothioate internucleoside linkage.
59. The compound of claim 57, wherein the modified oligonucleotide comprises at least one phosphodiester internucleoside linkage.
60. The compound of claim 57, wherein the modified oligonucleotide comprises at least 2 phosphodiester internucleoside linkages.
61. The compound of claim 57, wherein the modified oligonucleotide comprises at least 3 phosphodiester internucleoside linkages.
62. The compound of claim 57, wherein the modified oligonucleotide comprises at least 4 phosphodiester internucleoside linkages.
63. The compound of claim 57, wherein the modified oligonucleotide comprises at least 5 phosphodiester internucleoside linkages.
64. The compound of claim 57, wherein the modified oligonucleotide comprises at least 6 phosphodiester internucleoside linkages.
65. The compound of claim 57, wherein the modified oligonucleotide comprises at least 7 phosphodiester internucleoside linkages.
66. The compound of any of the preceding claims, wherein each internucleoside linkage of the modified oligonucleotide is selected from a phosphodiester internucleoside linkage and a phosphorothioate internucleoside linkage.
67. The compound of any of the preceding claim, wherein each internucleoside linkage of the modified oligonucleotide comprises is a phosphorothioate internucleoside linkage.
68. The compound of any preceding claim, wherein the conjugate group is linked to the modified oligonucleotide at the 5' end of the modified oligonucleotide.
69. The compound of any preceding claim, wherein the conjugate group is linked to the modified oligonucleotide at the 3' end of the modified oligonucleotide.
70. The compound of any preceding claim, wherein the conjugate group comprises exactly one ligand.
71. The compound of any preceding claim, wherein the conjugate group comprises exactly two ligands.
72. The compound of any preceding claim, wherein the conjugate group comprises three or more ligands.
73. The compound of any preceding claim, wherein the conjugate group comprises exactly three ligands.
74. The compound of any preceding claim, 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.
75. The compound any preceding claim, wherein each ligand is N-acetyl galactosamine.
76. The compound of any preceding claim, wherein the conjugate group comprises:

77. The compound of any preceding claim, wherein the conjugate group comprises:
78. The compound of any preceding claim, wherein the conjugate group comprises:
79. The compound of any preceding claim, wherein the conjugate group comprises:

80. The compound of any preceding claim, wherein the conjugate group comprises:
81. The compound of any preceding claim, wherein the conjugate group comprises at least one phosphorus linking group or neutral linking group.
82. The compound of any preceding claim, 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.
83. The compound of any preceding claim, 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.
84. The compound of any preceding claim, 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.
85. The compound of any preceding claim, 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.
86. The compound of any preceding claim, wherein the conjugate group is covalently attached to the modified oligonucleotide.
87. The compound of any preceding claim, 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.
88. The compound of any preceding claim, 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.
89. The compound of any preceding claim, 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.
90. The compound of any preceding claim, 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.
91. The compound of any preceding claim, 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.
92. The compound of any preceding claim, 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.
93. The compound of any preceding claim, 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.
94. The compound of any preceding claim, 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.
95. The compound of any preceding claim, 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.
96. The compound of any preceding claim, wherein the conjugate linker has a structure selected from among:
97. The compound of any preceding claim, wherein the conjugate linker has the followingstructure:
98. The compound of any preceding claim, wherein the conjugate linker has a structure selected from among:
99. The compound of any preceding claim, wherein the conjugate linker has a structure selected from among:
100. The compound of any preceding claim, wherein the conjugate linker has a structure selected from among:
101. The compound of any preceding claim, wherein the conjugate linker comprises a pyrrolidine.
102. The compound of any preceding claim, wherein the conjugate linker does not comprise a pyrrolidine.
103. The compound of any preceding claim, wherein the conjugate linker comprises PEG.
104. The compound of any preceding claim, wherein the conjugate linker comprises an amide.
105. The compound of any preceding claim, wherein the conjugate linker comprises at least two amides.
106. The compound of any preceding claim, wherein the conjugate linker does not comprise an amide.
107. The compound of any preceding claim, wherein the conjugate linker comprises a polyamide.
108. The compound of any preceding claim, wherein the conjugate linker comprises an amine.
109. The compound of any preceding claim, wherein the conjugate linker comprises one or more disulfide bonds.
110. The compound of any preceding claim, wherein the conjugate linker comprises a protein binding moiety.
111. The compound of any preceding claim, wherein the protein binding moiety comprises a lipid.
112. The compound of any preceding claim, 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.
113. The compound of any preceding claim, 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.
114. The compound of any preceding claim, 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.
115. The compound of any preceding claim, wherein the conjugate linker has a structure selected from among:

wherein each n is, independently, from 1 to 20.
116.
The compound of any preceding claim, wherein the conjugate linker has a structure selected from among:
117. The compound of any preceding claim, wherein the conjugate linker has a structure selected from among:
wherein n is from 1 to 20.
118. The compound of any preceding claim, wherein the conjugate linker has a structure selected from among:
119. The compound of any preceding claim, wherein the conjugate linker has a structure selected from among:
wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
120. The compound of any preceding claim, wherein the conjugate linker has the following structure:
121. The compound of any preceding claim, 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.
122. The compound of any preceding claim, 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.
123.
The compound of any preceding claim, wherein the branching group has the following structure:
124. The compound of any preceding claim, wherein the branching group has the following structure:
125. The compound of any preceding claim, wherein the branching group has the following structure:
126. The compound of any preceding claim, wherein the branching group has the following structure:
127. The compound of any preceding claim, wherein the branching group comprises an ether.
128. The compound of any preceding claim, wherein the branching group has the following structure:
each n is, independently, from 1 to 20; and m is from 2 to 6.
129. The compound of any preceding claim, wherein the branching group has the following structure:
130. The compound of any preceding claim, wherein the branching group has the following structure:
131. The compound of any preceding claim, 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.
132. The compound of any preceding claim, wherein the branching group comprises:
133. The compound of any preceding claim, 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.
134. The compound of any preceding claim, 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.
135. The compound of any preceding claim, wherein each tether is selected from among:
wherein n is from 1 to 12; and wherein m is from 1 to 12.
136. The compound of any preceding claim, wherein at least one tether comprises ethylene glycol.
137. The compound of any preceding claim, wherein at least one tether comprises an amide.
138. The compound of any preceding claim, wherein at least one tether comprises a polyamide.
139. The compound of any preceding claim, wherein at least one tether comprises an amine.
140. The compound of any preceding claim, wherein at least two tethers are different from one another.
141. The compound of any preceding claim, wherein all of the tethers are the same as one another.
142. The compound of any preceding claim, 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.
143. The compound of any preceding claim, wherein each tether is selected from among:
144. The compound of any preceding claim, wherein each tether has the following structure:
wherein each n is, independently, from 1 to 20.
145. The compound of any preceding claim, wherein each tether has the following structure:
146. The compound of any preceding claim, wherein the tether has a structure selected from among:
wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
147. The compound of any preceding claim, wherein the tether has a structure selected from among:
148. The compound of any preceding claim, wherein the ligand is galactose.
149. The compound of any preceding claim, wherein the ligand is mannose-6-phosphate.
150. The compound of any preceding claim, wherein each ligand is selected from among:
wherein each R1 is selected from OH and NHCOOH.
151. The compound of any preceding claim, wherein each ligand is selected from among:
152. The compound of any preceding claim, wherein each ligand has the following structure:
153. The conjugated antisense compound of any preceding claim, wherein each ligand has the following structure:
154. The compound of any of claims any preceding claim, wherein the conjugate group comprises a cell-targeting moiety.
155. The compound of any preceding claim, wherein the conjugate group comprises a cell-targeting moiety having the following structure:
wherein each n is, independently, from 1 to 20.
156. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
157. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
wherein each n is, independently, from 1 to 20.
158. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
159. The compound of any preceding claim, wherein the cell-targeting moiety comprises:
160. The compound of any preceding claim, wherein the cell-targeting moiety comprises:
161. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
162. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
163. The compound of any preceding claim, wherein the cell-targeting moiety comprises:
164. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
165. The compound of any preceding claim, wherein the cell-targeting moiety comprises:
166. The compound of any preceding claim, wherein the cell-targeting moiety comprises:
167. The compound of any preceding claim, wherein the cell-targeting moiety comprises:
168. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
169. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
170. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
171. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:

172. The compound of any preceding claim, wherein the cell-targeting moiety comprises:
173. The compound of any preceding claim, wherein the cell-targeting moiety comprises:

174. The compound of any preceding claim, wherein the cell-targeting moiety comprises:
175. The compound of any preceding claim, wherein the cell-targeting moiety comprises:

176. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
177. The compound of any preceding claim, wherein the cell-targeting moiety comprises:
178. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:

179. The compound of any preceding claim, 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.
180. The compound of any preceding claim, 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.
181. The compound of any preceding claim, wherein the cell-targeting moiety has the following structure:
wherein each Y is selected from O, S, a substituted or unsubstituted C1-C10 alkyl, amino, substituted amino, azido, alkenyl or alkynyl.
182. The compound of any of any preceding claim, wherein the conjugate group comprises:
183. The compound of any of any preceding claim, wherein the conjugate group comprises:
184. The compound of any of any preceding claim, wherein the conjugate group comprises:
185. The compound of any preceding claim, wherein the conjugate group comprises:
186. The compound of any preceding claim, wherein the conjugate group comprises a cleavable moiety selected from among: a phosphodiester, an amide, or an ester.
187. The compound of any preceding claim, wherein the conjugate group comprises a phosphodiester cleavable moiety.
188. The compound of any preceding claim, 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.
189. The compound of any preceding claim, wherein the conjugate group comprises an amide cleavable moiety.
190. The compound of any preceding claim, wherein the conjugate group comprises an ester cleavable moiety.
191. The compound of any preceding claim, 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.
192. The compound of any preceding claim, 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.
193. The compound of any preceding claim, 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.
194. The compound of any preceding claim, 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.
195. The compound of any preceding claim, 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.
196. The compound of any preceding claim, 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.
197. The compound of any preceding claim, 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.
198. The compound of any preceding claim, 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.
199. The compound of any preceding claim, 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.
200. The compound of any preceding claim, 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.
201. The compound of any preceding claim, 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.
202. The compound of any preceding claim, 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.
203. The compound of any preceding claim, 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.
204. The compound of any preceding claim, 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.
205. The compound of any preceding claim, 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.
206. The compound of any preceding claim, 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.
207. The compound of any preceding claim, 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.
208. The compound of any preceding claim, 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.
209. The compound of any preceding claim, wherein B x is selected from among from adenine, guanine, thymine, uracil, or cytosine, or 5-methyl cytosine.
210. The compound of any preceding claim, wherein B x is adenine.
211. The compound of any preceding claim, wherein B x is thymine.
212. The compound of any preceding claim, wherein Q13 is O(CH2)2-OCH3.
213. The compound of any preceding claim, wherein Q13 is H.
214. A composition comprising the compound of any preceding claim or salt thereof and at least one of a pharmaceutically acceptable carrier or diluent.
215. A prodrug comprising the compound of any preceding claim.
216. A method comprising administering to an animal the compound or composition of any preceding claim.
217. The method of claim 216, wherein the animal is a human.
218. The method of claim 216, comprising co-administering the compound or composition and a second agent.
219. The method of claim 218, wherein the compound or composition and the second agent are administered concomitantly.
CA2921518A 2013-05-01 2014-05-01 Compositions and methods Abandoned CA2921518A1 (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/US2014/036466 WO2014179629A2 (en) 2013-05-01 2014-05-01 Compositions and methods

Publications (1)

Publication Number Publication Date
CA2921518A1 true CA2921518A1 (en) 2014-11-06

Family

ID=51843959

Family Applications (5)

Application Number Title Priority Date Filing Date
CA2921518A Abandoned CA2921518A1 (en) 2013-05-01 2014-05-01 Compositions and methods
CA2921514A Active CA2921514C (en) 2013-05-01 2014-05-01 Compositions and methods for modulating apolipoprotein c-iii expression
CA2921167A Pending CA2921167A1 (en) 2013-05-01 2014-05-01 Compositions and methods for modulating hbv and ttr expression
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

Family Applications After (4)

Application Number Title Priority Date Filing Date
CA2921514A Active CA2921514C (en) 2013-05-01 2014-05-01 Compositions and methods for modulating apolipoprotein c-iii expression
CA2921167A Pending CA2921167A1 (en) 2013-05-01 2014-05-01 Compositions and methods for modulating hbv and ttr expression
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

Country Status (35)

Country Link
US (23) US9714421B2 (en)
EP (11) EP4155403A1 (en)
JP (20) JP6387084B2 (en)
KR (14) KR20230113835A (en)
CN (12) CN105378082B (en)
AU (19) AU2014259750B2 (en)
BR (5) BR112015027319A8 (en)
CA (5) CA2921518A1 (en)
CL (2) CL2015003217A1 (en)
CR (2) CR20190269A (en)
CY (2) CY1121879T1 (en)
DK (4) DK2992009T3 (en)
DO (3) DOP2015000268A (en)
EA (2) EA036584B1 (en)
ES (4) ES2819213T3 (en)
HK (5) HK1221475A1 (en)
HR (2) HRP20190987T1 (en)
HU (2) HUE050394T2 (en)
IL (18) IL284593B2 (en)
LT (2) LT2992009T (en)
ME (1) ME03390B (en)
MX (11) MX2015015220A (en)
MY (2) MY178929A (en)
NZ (6) NZ728517A (en)
PE (2) PE20152002A1 (en)
PH (3) PH12015502493B1 (en)
PL (2) PL2992098T3 (en)
PT (3) PT3524680T (en)
RS (2) RS60796B1 (en)
RU (8) RU2686080C2 (en)
SG (5) SG10201906382QA (en)
SI (2) SI2992098T1 (en)
UA (2) UA121017C2 (en)
WO (5) WO2014179626A2 (en)
ZA (2) ZA201507218B (en)

Families Citing this family (409)

* 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
EP2370451B1 (en) 2008-12-02 2016-11-16 Wave Life Sciences Japan, Inc. Method for the synthesis of phosphorus atom modified nucleic acids
SG177564A1 (en) 2009-07-06 2012-02-28 Ontorii Inc Novel nucleic acid prodrugs and methods of use thereof
JP5868324B2 (en) 2010-09-24 2016-02-24 株式会社Wave Life Sciences Japan Asymmetric auxiliary group
EP2640853B1 (en) 2010-11-17 2018-12-26 Ionis Pharmaceuticals, Inc. Modulation of alpha synuclein expression
RU2014105311A (en) 2011-07-19 2015-08-27 Уэйв Лайф Сайенсес Пте. Лтд. METHODS FOR SYNTHESIS OF FUNCTIONALIZED NUCLEIC ACIDS
DK2751270T3 (en) 2011-08-29 2018-10-29 Ionis Pharmaceuticals Inc OLIGOMER-CONJUGATE COMPLEXES AND THEIR USE
AU2013202595B2 (en) 2012-03-30 2016-04-21 Biogen Ma Inc. Methods for modulating Tau expression for reducing seizure and modifying a neurodegenerative syndrome
EP2839006B1 (en) * 2012-04-20 2018-01-03 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
US9574193B2 (en) 2012-05-17 2017-02-21 Ionis Pharmaceuticals, Inc. Methods and compositions for modulating apolipoprotein (a) expression
US20160002624A1 (en) 2012-05-17 2016-01-07 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions
RU2624028C2 (en) * 2012-05-24 2017-06-30 Ионис Фармасьютикалз, Инк. Methods and compositions for modeling expression of apolipoprotein (a)
SG11201500243WA (en) 2012-07-13 2015-04-29 Shin Nippon Biomedical Lab Ltd Chiral nucleic acid adjuvant
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
SG11201500239VA (en) 2012-07-13 2015-03-30 Wave Life Sciences Japan Asymmetric auxiliary group
US20150291958A1 (en) 2012-11-15 2015-10-15 Roche Innovation Center Copenhagen A/S Anti apob antisense conjugate compounds
DK2951305T3 (en) * 2013-01-30 2018-10-29 Hoffmann La Roche LNA oligonucleotide KULHYDRATKONJUGATER
WO2014127268A2 (en) * 2013-02-14 2014-08-21 Isis Pharmaceuticals, Inc. Modulation of apolipoprotein c-iii (apociii) expression in lipoprotein lipase deficient (lpld) populations
US9309513B2 (en) 2013-05-01 2016-04-12 Regulus Therapeutics Inc. MicroRNA compounds and methods for modulating miR-122
TWI680767B (en) 2013-05-01 2020-01-01 美商雷格勒斯治療公司 Compounds and methods for enhanced cellular uptake
SG10201906382QA (en) 2013-05-01 2019-08-27 Ionis Pharmaceuticals Inc Compositions and methods for modulating hbv and ttr expression
BR112015032432B1 (en) 2013-06-27 2023-02-07 Roche Innovation Center Copenhagen A/S ANTI-SENSE OLIGOMER, ANTI-SENSE OLIGONUCLEOTIDE CONJUGATES, PHARMACEUTICAL COMPOSITION, USE THEREOF FOR THE TREATMENT OF HYPERCHOLESTEROLEMIA OR RELATED DISORDERS, AND IN VITRO METHOD FOR REDUCING THE EXPRESSION LEVELS AND/OR THE ACTIVITY OF PCSK9 IN A CELL
CA2917161C (en) * 2013-07-11 2023-12-12 Alnylam Pharmaceuticals, Inc. Oligonucleotide-ligand conjugates and process for their preparation
EP3022176B8 (en) 2013-07-15 2019-12-25 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
WO2015042447A1 (en) 2013-09-20 2015-03-26 Isis Pharmaceuticals, Inc. Targeted therapeutic nucleosides and their use
CN105793423A (en) * 2013-10-02 2016-07-20 阿尔尼拉姆医药品有限公司 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
WO2015061536A1 (en) * 2013-10-25 2015-04-30 Regulus Therapeutics Inc. Microrna compounds and methods for modulating mir-21 activity
RU2016122168A (en) * 2013-11-14 2017-12-19 Рош Инновейшен Сентер Копенгаген А/С ANTI-SENSE CONJUGATES AIMED AT APOLIPOPROTEIN B
WO2015105083A1 (en) * 2014-01-07 2015-07-16 塩野義製薬株式会社 Double-stranded oligonucleotide containing antisense oligonucleotide and sugar derivative
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
JPWO2015108047A1 (en) 2014-01-15 2017-03-23 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
DK3094728T3 (en) 2014-01-16 2022-05-16 Wave Life Sciences Ltd KIRALT DESIGN
WO2015123264A1 (en) 2014-02-11 2015-08-20 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
RU2019130898A (en) 2014-03-19 2019-11-11 Ионис Фармасьютикалз, Инк. COMPOSITIONS FOR MODULATION OF ATAXIN 2 EXPRESSION
RU2704619C2 (en) 2014-04-01 2019-10-30 Биоген Ма Инк. Compositions for modulating expression of sod-1
AU2015252917B2 (en) 2014-05-01 2019-09-26 Ionis Pharmaceuticals, Inc. 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
SI3137605T1 (en) 2014-05-01 2021-02-26 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating angiopoietin-like 3 expression
SG11201608502TA (en) 2014-05-01 2016-11-29 Ionis Pharmaceuticals Inc Compositions and methods for modulating complement factor b expression
EP3137604B1 (en) 2014-05-01 2020-07-15 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating growth hormone receptor expression
GB201408623D0 (en) 2014-05-15 2014-07-02 Santaris Pharma As Oligomers and oligomer conjugates
EA201692370A1 (en) 2014-05-22 2017-03-31 Элнилэм Фармасьютикалз, Инк. COMPOSITIONS of mRNA ANGIOTENZINOGENA (AGT) AND METHODS OF THEIR USE
GB201410693D0 (en) 2014-06-16 2014-07-30 Univ Southampton Splicing modulation
CN106559995A (en) 2014-08-07 2017-04-05 莱古路斯治疗法股份有限公司 The Microrna of targeting metabolic disorder
KR102620328B1 (en) 2014-10-03 2024-01-02 콜드스프링하버러보러토리 Targeted augmentation of nuclear gene output
CN106795200B (en) * 2014-10-10 2020-06-19 豪夫迈·罗氏有限公司 Galnac phosphoramidites, nucleic acid conjugates thereof, and uses thereof
EP3207138B1 (en) * 2014-10-17 2020-07-15 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
CN107250362B (en) 2014-11-17 2021-10-22 阿尔尼拉姆医药品有限公司 Apolipoprotein C3(APOC3) iRNA compositions and methods of use thereof
JP2018504380A (en) 2014-12-18 2018-02-15 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. REVERSIR ™ compounds
WO2016112132A1 (en) 2015-01-06 2016-07-14 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
WO2016115490A1 (en) 2015-01-16 2016-07-21 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
MX2017012426A (en) 2015-04-03 2018-01-26 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
EP3286310A4 (en) 2015-04-24 2019-01-09 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
MY192997A (en) 2015-07-10 2022-09-20 Ionis Pharmaceuticals Inc Modulators of diacyglycerol acyltransferase 2 (dgat2)
US20180193471A1 (en) * 2015-07-16 2018-07-12 Kyowa Hakko Kirin Co., Ltd. ß2GPI GENE EXPRESSION-SUPPRESSING NUCLEIC ACID CONJUGATE
MA43072A (en) 2015-07-22 2018-05-30 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
CN107709342B (en) 2015-08-06 2021-09-03 豪夫迈·罗氏有限公司 Process for preparing acetylgalactosamine acid derivatives
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
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
WO2017032726A1 (en) 2015-08-24 2017-03-02 Roche Innovation Center Copenhagen A/S Lna-g process
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
CA2999177A1 (en) 2015-09-24 2017-03-30 The Regents Of The University Of 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
WO2017053781A1 (en) 2015-09-25 2017-03-30 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating ataxin 3 expression
US11116843B2 (en) 2015-09-25 2021-09-14 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
EP3353305A4 (en) * 2015-09-25 2019-09-18 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
JP6893505B2 (en) 2015-10-02 2021-06-23 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 Conjugation Method
KR102422625B1 (en) 2015-10-09 2022-07-20 유니버시티 오브 사우스앰톤 Regulation of gene expression and screening of deregulated protein expression
WO2017068087A1 (en) 2015-10-22 2017-04-27 Roche Innovation Center Copenhagen A/S Oligonucleotide detection method
US10955407B2 (en) 2015-10-22 2021-03-23 Roche Innovation Center Copenhagen A/S In vitro toxicity screening assay
PE20181180A1 (en) 2015-11-06 2018-07-20 Ionis Pharmaceuticals Inc MODULATE THE EXPRESSION OF APOLIPOPROTEIN (a)
US20190046555A1 (en) 2015-11-06 2019-02-14 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds for use in therapy
MX2018005733A (en) 2015-11-16 2018-08-09 Hoffmann La Roche GalNAc CLUSTER PHOSPHORAMIDITE.
WO2017096395A1 (en) 2015-12-04 2017-06-08 Ionis Pharmaceuticals, Inc. Methods of treating breast cancer
US11096956B2 (en) 2015-12-14 2021-08-24 Stoke Therapeutics, Inc. Antisense oligomers and uses thereof
SG11201804443UA (en) 2015-12-14 2018-06-28 Cold Spring Harbor Laboratory Antisense oligomers for treatment of autosomal dominant mental retardation-5 and dravet syndrome
AU2017205462A1 (en) 2016-01-05 2018-06-07 Ionis Pharmaceuticals, Inc. Methods for reducing LRRK2 expression
AU2017207341A1 (en) 2016-01-12 2018-08-02 Interleukin Genetics, Inc. Methods for predicting response to treatment
JP6988481B2 (en) 2016-01-26 2022-01-05 日産化学株式会社 Single-stranded oligonucleotide
ES2858090T3 (en) 2016-01-29 2021-09-29 Kyowa Kirin Co Ltd Nucleic acid complex
WO2017132669A1 (en) 2016-01-31 2017-08-03 University Of Massachusetts Branched oligonucleotides
EP3426261A4 (en) 2016-03-07 2020-03-25 Arrowhead Pharmaceuticals, Inc. Targeting ligands for therapeutic compounds
US11136577B2 (en) 2016-03-09 2021-10-05 Ionis Pharmaceuticals, Inc. Methods and compositions for inhibiting PMP22 expression
MY194912A (en) 2016-03-14 2022-12-22 Hoffmann La Roche Oligonucleotides for Reduction of PD-L1 Expression
AU2017234678A1 (en) 2016-03-16 2018-08-16 Ionis Pharmaceuticals, Inc. Methods of modulating KEAP1
WO2017161168A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Modulation of dyrk1b expression
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
US11248019B2 (en) 2016-04-14 2022-02-15 Hoffmann-La Roche Inc. Trityl-mono-GalNAc compounds and their use
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
EP3449002A4 (en) 2016-04-29 2019-09-25 Nanyang Technological University G-quadruplex-containing antisense oligonucleotides
MA45270A (en) 2016-05-04 2017-11-09 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
EP3471781A4 (en) 2016-06-17 2020-05-06 Ionis Pharmaceuticals, Inc. Modulation of gys1 expression
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
EP3472348B1 (en) 2016-06-17 2022-06-29 F. Hoffmann-La Roche AG In vitro nephrotoxicity screening assay
CN109312403B (en) 2016-06-17 2023-06-27 豪夫迈·罗氏有限公司 In vitro nephrotoxicity screening assay
TWI746590B (en) 2016-06-30 2021-11-21 日商協和麒麟股份有限公司 Nucleic acid complex
US11253601B2 (en) 2016-07-11 2022-02-22 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
RS63928B1 (en) 2016-07-15 2023-02-28 Ionis Pharmaceuticals Inc Compounds and methods for modulation of smn2
WO2018013999A1 (en) 2016-07-15 2018-01-18 Am Chemicals Llc Non-nucleosidic solid supports and phosphoramidite building blocks for oligonucleotide synthesis
WO2018031933A2 (en) 2016-08-12 2018-02-15 University Of Massachusetts Conjugated oligonucleotides
AU2017312116A1 (en) * 2016-08-17 2019-03-07 Solstice Biologics, Ltd. Polynucleotide constructs
JP6989521B2 (en) 2016-09-02 2022-01-05 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 Targeting ligand
JOP20190065A1 (en) 2016-09-29 2019-03-28 Ionis Pharmaceuticals Inc Compounds and methods for reducing tau expression
WO2018067900A1 (en) 2016-10-06 2018-04-12 Ionis Pharmaceuticals, Inc. Method of conjugating 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
AU2017356221A1 (en) * 2016-11-11 2019-05-30 Janssen Biopharma, Inc. Oligonucleotide targeting strategy for HBV cccDNA
TWI788312B (en) 2016-11-23 2023-01-01 美商阿尼拉製藥公司 SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP3548005A4 (en) 2016-11-29 2020-06-17 Puretech Health LLC Exosomes for delivery of therapeutic agents
US11033570B2 (en) 2016-12-02 2021-06-15 Cold Spring Harbor Laboratory Modulation of Lnc05 expression
KR101954130B1 (en) * 2016-12-13 2019-03-05 (주)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
US10329620B2 (en) 2017-01-12 2019-06-25 Cardioforecast Ltd. Methods and kits for treating cardiovascular disease
EP3568478A1 (en) 2017-01-13 2019-11-20 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating rel expression
EP3568480A1 (en) 2017-01-13 2019-11-20 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating nfkb2 expression
WO2018130585A1 (en) 2017-01-13 2018-07-19 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating relb expression
WO2018130583A1 (en) 2017-01-13 2018-07-19 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating nfkb1 expression
US20200216845A1 (en) 2017-01-13 2020-07-09 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating rela expression
EP3584319A4 (en) 2017-02-06 2021-04-14 Nissan Chemical Corporation Single-stranded oligonucleotide
US11180756B2 (en) * 2017-03-09 2021-11-23 Ionis Pharmaceuticals Morpholino modified oligomeric compounds
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
NZ757927A (en) 2017-04-11 2023-07-28 Arbutus Biopharma Corp Targeted compositions
AU2018254437A1 (en) 2017-04-18 2019-11-28 Alnylam Pharmaceuticals, Inc. Methods for the treatment of subjects having a hepatitis B virus (HBV) infection
WO2018215049A1 (en) 2017-05-23 2018-11-29 F. Hoffmann-La Roche Ag Process for galnac oligonucleotide conjugates
WO2018216785A1 (en) * 2017-05-26 2018-11-29 国立研究開発法人国立循環器病研究センター Antisense nucleic acid targeting pcsk9
US11603532B2 (en) 2017-06-02 2023-03-14 Wave Life Sciences Ltd. Oligonucleotide compositions and methods of use thereof
WO2018226788A1 (en) * 2017-06-07 2018-12-13 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
BR112020001430A2 (en) 2017-07-26 2020-07-28 Nissan Chemical Corporation 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
TW201909925A (en) * 2017-08-02 2019-03-16 日商協和醱酵麒麟有限公司 Nucleic acid complex
WO2019030313A2 (en) 2017-08-11 2019-02-14 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating ube3c expression
WO2019036613A1 (en) 2017-08-18 2019-02-21 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
PL3673080T3 (en) 2017-08-25 2024-03-11 Stoke Therapeutics, Inc. Antisense oligomers for treatment of conditions and diseases
US10517889B2 (en) 2017-09-08 2019-12-31 Ionis Pharmaceuticals, Inc. Modulators of SMAD7 expression
EP3682007A2 (en) * 2017-09-14 2020-07-22 Janssen BioPharma, Inc. Galnac derivatives
WO2019067872A1 (en) 2017-09-29 2019-04-04 Intellia Therapeutics, Inc. Compositions and methods for ttr gene editing and treating attr amyloidosis
EP3694995A1 (en) 2017-10-13 2020-08-19 Roche Innovation Center Copenhagen A/S Methods for identifying improved stereodefined phosphorothioate oligonucleotide variants of antisense oligonucleotides utilising sub-libraries of partially stereodefined oligonucleotides
TWI816066B (en) 2017-10-16 2023-09-21 瑞士商赫孚孟拉羅股份公司 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
EP3714054A1 (en) 2017-11-20 2020-09-30 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of 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
JP2021504415A (en) 2017-12-01 2021-02-15 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Suzhou Ribo Life Science Co., Ltd. Compositions and complexes containing double-stranded oligonucleotides, double-stranded oligonucleotides, and preparation methods and uses.
EP3718572A4 (en) 2017-12-01 2021-09-08 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, composition and conjugate containing nucleic acid, preparation method and use
JP7365052B2 (en) * 2017-12-01 2023-10-19 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 the nucleic acids, and preparation methods and uses
CN110945131A (en) * 2017-12-01 2020-03-31 苏州瑞博生物技术有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
WO2019105404A1 (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 therefor and use thereof
US20220395526A1 (en) 2017-12-01 2022-12-15 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, composition and conjugate containing same, and preparation method and use
US20200385714A1 (en) 2017-12-11 2020-12-10 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating fndc3b expression
WO2019115417A2 (en) 2017-12-12 2019-06-20 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating rb1 expression
WO2019118916A1 (en) 2017-12-14 2019-06-20 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
AU2018388484A1 (en) 2017-12-18 2020-07-30 Alnylam Pharmaceuticals, Inc. High mobility group box-1 (HMGB1) iRNA compositions and methods of use thereof
WO2019126641A2 (en) 2017-12-21 2019-06-27 Ionis Pharmaceuticals, Inc. Modulation of frataxin expression
BR112020012523A2 (en) 2017-12-21 2020-11-24 F. Hoffmann-La Roche Ag complementary diagnosis for htra1 rna antagonists
EP4092118A1 (en) 2017-12-22 2022-11-23 Roche Innovation Center Copenhagen A/S Novel thiophosphoramidites
WO2019122282A1 (en) 2017-12-22 2019-06-27 Roche Innovation Center Copenhagen A/S Gapmer oligonucleotides comprising a phosphorodithioate internucleoside linkage
EP3728592A1 (en) 2017-12-22 2020-10-28 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorodithioate internucleoside linkage
EP3677588A4 (en) * 2017-12-26 2020-11-25 Guangzhou Ribobio Co., Ltd. Modified oligonucleotides and compound that can be used for synthesizing same
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
AU2018394875B2 (en) * 2017-12-29 2023-08-03 Suzhou Ribo Life Science Co., Ltd. Conjugates and preparation and use thereof
WO2019140102A1 (en) * 2018-01-10 2019-07-18 Translate Bio Ma, Inc. Compositions and methods for facilitating delivery of synthetic nucleic acids to cells
US20210095274A1 (en) 2018-01-10 2021-04-01 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating pias4 expression
CN112424353A (en) 2018-01-12 2021-02-26 罗氏创新中心哥本哈根有限公司 Alpha-synuclein antisense oligonucleotides and uses thereof
KR20200140240A (en) 2018-01-12 2020-12-15 브리스톨-마이어스 스큅 컴퍼니 Antisense oligonucleotides targeting alpha-synuclein and uses thereof
WO2019140231A1 (en) 2018-01-12 2019-07-18 Bristol-Myers Squibb Company Antisense oligonucleotides targeting alpha-synuclein and uses thereof
WO2019137974A1 (en) 2018-01-12 2019-07-18 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating gsk3b expression
MX2020007369A (en) 2018-01-15 2020-10-28 Ionis Pharmaceuticals Inc Modulators of dnm2 expression.
EP3740574A1 (en) 2018-01-17 2020-11-25 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating erc1 expression
US20210095277A1 (en) 2018-01-18 2021-04-01 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting srebp1
WO2019145386A1 (en) 2018-01-26 2019-08-01 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating csnk1d expression
JP7317029B2 (en) 2018-02-12 2023-07-28 アイオーニス ファーマシューティカルズ, インコーポレーテッド Modified compounds and uses thereof
EP3752614A4 (en) 2018-02-14 2021-11-10 Deep Genomics Incorporated Oligonucleotide therapy for wilson disease
SG11202007652UA (en) 2018-02-21 2020-09-29 Bristol Myers Squibb Co Camk2d antisense oligonucleotides and uses thereof
US11732260B2 (en) 2018-03-02 2023-08-22 Ionis Pharmaceuticals, Inc. Compounds and methods for the modulation of amyloid-β precursor protein
TW202000199A (en) 2018-03-02 2020-01-01 美商Ionis製藥公司 Modulators of IRF4 expression
KR102398295B1 (en) 2018-03-09 2022-05-17 다이이찌 산쿄 가부시키가이샤 Glycogen disease type Ia treatment
US11661601B2 (en) 2018-03-22 2023-05-30 Ionis Pharmaceuticals, Inc. Methods for modulating FMR1 expression
CA3097544A1 (en) 2018-04-05 2019-10-10 F. Hoffmann-La Roche Ag Use of fubp1 inhibitors for treating hepatitis b virus infection
SG11202008660TA (en) 2018-04-11 2020-10-29 Ionis Pharmaceuticals Inc Modulators of ezh2 expression
WO2019215067A1 (en) 2018-05-07 2019-11-14 Roche Innovation Center Copenhagen A/S Massively parallel discovery methods for oligonucleotide therapeutics
CR20200604A (en) 2018-05-09 2021-02-09 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn3 expression
BR112020020957B1 (en) 2018-05-09 2022-05-10 Ionis Pharmaceuticals, Inc Oligomeric compounds, population and pharmaceutical composition thereof and their uses
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
US11833168B2 (en) 2018-06-14 2023-12-05 Ionis Pharmaceuticals, Inc. Compounds and methods for increasing STMN2 expression
US11332746B1 (en) 2018-06-27 2022-05-17 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing LRRK2 expression
WO2020007892A1 (en) 2018-07-03 2020-01-09 F. Hoffmann-La Roche Ag Oligonucleotides for modulating tau expression
WO2020007826A1 (en) 2018-07-05 2020-01-09 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides targeting mbtps1
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
CA3106288A1 (en) 2018-07-13 2020-01-16 F.Hoffmann-La Roche Ag Oligonucleotides for modulating rtel1 expression
MX2021000922A (en) 2018-07-25 2021-03-31 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn2 expression.
US20210221837A1 (en) 2018-07-31 2021-07-22 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorotrithioate internucleoside linkage
US20210292358A1 (en) 2018-07-31 2021-09-23 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorotrithioate internucleoside linkage
WO2020033748A1 (en) 2018-08-08 2020-02-13 Arcturus Therapeutics, Inc. Compositions and agents against nonalcoholic steatohepatitis
EP3833763A4 (en) 2018-08-10 2023-07-19 University of Massachusetts Modified oligonucleotides targeting snps
BR112021001613A2 (en) 2018-08-13 2021-05-04 Alnylam Pharmaceuticals, Inc. double-stranded ribonucleic acid agents, cell, pharmaceutical compositions, methods of inhibiting gene expression, inhibiting replication and treating a subject, methods for reducing the level of an antigen and for reducing viral load, and use of an agent dsrna
JP2021533800A (en) 2018-08-21 2021-12-09 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Suzhou Ribo Life Science Co., Ltd. Nucleic acid, drug compositions and complexes containing the nucleic acid, and their use
CN113728102A (en) 2018-08-28 2021-11-30 罗氏创新中心哥本哈根有限公司 Novel antigen engineering using splice-modulating compounds
US20210332367A1 (en) 2018-09-18 2021-10-28 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
TW202023573A (en) 2018-09-19 2020-07-01 美商Ionis製藥公司 Modulators of pnpla3 expression
CN111655297A (en) * 2018-09-30 2020-09-11 苏州瑞博生物技术有限公司 siRNA conjugate and preparation method and application thereof
CN113383076A (en) * 2018-10-18 2021-09-10 莫道克大学 Antisense therapy for PTP 1B-related disorders
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
CN113286885A (en) 2018-11-09 2021-08-20 诺华股份有限公司 Methods of reducing the risk of cardiovascular events 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
BR112021008967A2 (en) 2018-11-15 2021-08-17 Ionis Pharmaceuticals, Inc. irf5 expression modulators
TW202039841A (en) 2018-11-21 2020-11-01 美商Ionis製藥公司 Compounds and methods for reducing prion expression
CN113166760A (en) 2018-11-23 2021-07-23 赛诺菲 Novel RNA compositions and methods for inhibiting ANGPTL8
AU2019390097A1 (en) 2018-11-30 2021-07-15 Kyowa Kirin Co., Ltd. Nucleic acid conjugate
WO2020132521A1 (en) 2018-12-20 2020-06-25 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
WO2020147847A1 (en) * 2019-01-18 2020-07-23 苏州瑞博生物技术有限公司 Nucleic acid, composition and conjugate containing nucleic acid, preparation method, and use
JP2022519532A (en) 2019-01-31 2022-03-24 アイオーニス ファーマシューティカルズ, インコーポレーテッド Modulator of YAP1 expression
CN113490742A (en) 2019-02-20 2021-10-08 罗氏创新中心哥本哈根有限公司 Phosphonoacetate gapmer oligonucleotides
MX2021009949A (en) 2019-02-20 2021-09-21 Roche Innovation Ct Copenhagen As Novel phosphoramidites.
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
WO2020173845A1 (en) 2019-02-26 2020-09-03 Roche Innovation Center Copenhagen A/S Oligonucleotide formulation method
AU2020227824A1 (en) 2019-02-27 2021-08-26 Ionis Pharmaceuticals, Inc. Modulators of MALAT1 expression
US20220145292A1 (en) * 2019-02-28 2022-05-12 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
CA3135794A1 (en) 2019-04-03 2020-10-08 Bristol-Myers Squibb Company Angptl2 antisense oligonucleotides and uses thereof
CN113811613B (en) 2019-05-22 2024-04-05 苏州瑞博生物技术股份有限公司 Nucleic acid, pharmaceutical composition and conjugate, and preparation method and application thereof
US20230193277A1 (en) * 2019-05-22 2023-06-22 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition, conjugate, preparation method, and use
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
US20230313195A1 (en) 2019-05-22 2023-10-05 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition, conjugate, preparation method, and use
EP3978029A4 (en) 2019-05-24 2023-07-19 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition and conjugate, preparation method and use
CN113891939B (en) 2019-05-24 2024-04-02 苏州瑞博生物技术股份有限公司 Nucleic acid, pharmaceutical composition and conjugate, and preparation method and application thereof
US20220235359A1 (en) 2019-05-24 2022-07-28 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition, conjugate, preparation method, and use
CA3141874A1 (en) 2019-05-31 2020-12-03 Aligos Therapeutics, Inc. Modified gapmer oligonucleotides and methods of use
AU2020286382A1 (en) 2019-06-04 2021-11-04 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized TTR locus with a beta-slip mutation and methods of use
AU2020301419A1 (en) 2019-06-25 2022-01-20 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
WO2021022109A1 (en) 2019-08-01 2021-02-04 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
US20220290156A1 (en) 2019-08-27 2022-09-15 Sanofi Compositions and methods for inhibiting pcsk9
CN113891892B (en) 2019-08-29 2024-01-30 苏州瑞博生物技术股份有限公司 Compounds and drug conjugates, methods of preparation and uses thereof
EP4022062A1 (en) 2019-08-30 2022-07-06 Alnylam Pharmaceuticals, Inc. Neurofilament light chain (nfl) as a biomarker for transthyretin amyloidosis polyneuropathy
AU2020340987A1 (en) * 2019-09-03 2022-04-14 Arcturus Therapeutics, Inc. Asialoglycoprotein receptor mediated delivery of therapeutically active conjugates
WO2021049504A1 (en) * 2019-09-10 2021-03-18 第一三共株式会社 Galnac-oligonucleotide conjugate for liver-targeted delivery use, and method for producing same
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
JP2022552249A (en) 2019-10-14 2022-12-15 アストラゼネカ・アクチエボラーグ Modulators of PNPLA3 expression
WO2021076828A1 (en) 2019-10-18 2021-04-22 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
AU2020369515A1 (en) 2019-10-22 2022-04-21 Alnylam Pharmaceuticals, Inc. 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
WO2021096763A1 (en) 2019-11-13 2021-05-20 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
US20230056569A1 (en) 2019-11-22 2023-02-23 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
MX2022006433A (en) 2019-12-13 2022-06-23 Alnylam Pharmaceuticals Inc Human chromosome 9 open reading frame 72 (c9orf72) irna agent compositions and methods of use thereof.
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
EP4077668A1 (en) 2019-12-19 2022-10-26 F. Hoffmann-La Roche AG Use of scamp3 inhibitors for treating hepatitis b virus infection
JP2023506546A (en) 2019-12-19 2023-02-16 エフ. ホフマン-ラ ロシュ エージー. Use of SEPT9 inhibitors to treat hepatitis B virus infection
JP2023506954A (en) 2019-12-19 2023-02-20 エフ. ホフマン-ラ ロシュ エージー. Use of SARAF inhibitors to treat hepatitis B virus infection
WO2021122910A1 (en) 2019-12-19 2021-06-24 F. Hoffmann-La Roche Ag Use of sbds inhibitors for treating hepatitis b virus infection
WO2021122921A1 (en) 2019-12-19 2021-06-24 F. Hoffmann-La Roche Ag Use of cops3 inhibitors for treating hepatitis b virus infection
JP2023509869A (en) 2019-12-24 2023-03-10 エフ. ホフマン-ラ ロシュ アーゲー Methods of treating viral infections with TLR7 agonists
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
WO2021130270A1 (en) 2019-12-24 2021-07-01 F. Hoffmann-La Roche Ag Pharmaceutical combination of antiviral agents targeting hbv and/or an immune modulator for treatment of hbv
JPWO2021153687A1 (en) * 2020-01-30 2021-08-05
CN115427571A (en) 2020-02-10 2022-12-02 阿尔尼拉姆医药品有限公司 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.
TW202140787A (en) 2020-02-28 2021-11-01 美商Ionis製藥公司 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
KR20220152270A (en) 2020-03-06 2022-11-15 알닐람 파마슈티칼스 인코포레이티드 Compositions and methods for inhibiting the expression of transthyretin (TTR)
CA3174725A1 (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
US20230314417A1 (en) 2020-03-23 2023-10-05 Amgen Inc. Monoclonal antibodies to chemically-modified nucleic acids and uses thereof
EP4130266A1 (en) * 2020-03-26 2023-02-08 National Cerebral and Cardiovascular Center Antisense nucleic acid targeting apoc3
WO2021195307A1 (en) 2020-03-26 2021-09-30 Alnylam Pharmaceuticals, Inc. Coronavirus irna compositions and methods of use thereof
US20230295622A1 (en) 2020-04-06 2023-09-21 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing myoc expression
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
EP4133077A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
WO2021206917A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. ANGIOTENSIN-CONVERTING ENZYME 2 (ACE2) 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
BR112022021813A2 (en) 2020-04-27 2023-01-17 Alnylam Pharmaceuticals Inc APOLIPOPROTEIN AND (APOE) IRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
IL297680A (en) 2020-04-30 2022-12-01 Alnylam Pharmaceuticals Inc Complement factor b (cfb) irna compositions and methods of use thereof
EP4143321A2 (en) 2020-05-01 2023-03-08 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating atxn1
WO2021231107A1 (en) 2020-05-11 2021-11-18 Stoke Therapeutics, Inc. Opa1 antisense oligomers for treatment of conditions and diseases
WO2021231685A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
EP4150090A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
EP4150088A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
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)
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)
WO2021231691A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rsi)
EP4150076A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
AR122534A1 (en) 2020-06-03 2022-09-21 Triplet Therapeutics Inc METHODS FOR THE TREATMENT OF NUCLEOTIDE REPEAT EXPANSION DISORDERS ASSOCIATED WITH MSH3 ACTIVITY
EP4162050A1 (en) 2020-06-09 2023-04-12 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
CN115702006A (en) * 2020-06-10 2023-02-14 南京明德新药研发有限公司 Conjugate group and conjugate thereof
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
JP2023532518A (en) 2020-06-29 2023-07-28 アイオーニス ファーマシューティカルズ, インコーポレーテッド Compounds and methods for modulating 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
JP2023540429A (en) 2020-07-10 2023-09-25 アンセルム(アンスティチュート・ナシオナル・ドゥ・ラ・サンテ・エ・ドゥ・ラ・ルシェルシュ・メディカル) Methods and compositions for treating epilepsy
JP2023537943A (en) * 2020-08-13 2023-09-06 アムジェン インコーポレイテッド RNAi constructs and methods for inhibiting MARC1 expression
JP2023538630A (en) 2020-08-21 2023-09-08 エフ. ホフマン-ラ ロシュ アーゲー Use of A1CF inhibitors to treat hepatitis B virus infection
EP4217489A1 (en) 2020-09-24 2023-08-02 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
TW202229552A (en) 2020-10-05 2022-08-01 美商艾拉倫製藥股份有限公司 G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
CN116887842A (en) 2020-10-16 2023-10-13 赛诺菲 Novel RNA compositions and methods for inhibiting ANGPTL3
CN116490195A (en) 2020-10-16 2023-07-25 赛诺菲 RNA compositions and methods for inhibiting lipoprotein (a)
EP4232582A1 (en) 2020-10-23 2023-08-30 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
JP2023549500A (en) 2020-11-13 2023-11-27 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 Coagulation factor V (F5) iRNA composition and method of use thereof
US11447521B2 (en) 2020-11-18 2022-09-20 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
CA3202708A1 (en) 2020-11-23 2022-05-27 Alpha Anomeric Sas Nucleic acid duplexes
EP4259795A1 (en) 2020-12-08 2023-10-18 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
EP4237561A1 (en) 2020-12-23 2023-09-06 Argonaute Rna Limited Treatment of cardiovascular disease
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
WO2022150260A1 (en) 2021-01-05 2022-07-14 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT 9 (C9) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP4175964A1 (en) * 2021-01-30 2023-05-10 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
WO2022162161A1 (en) * 2021-01-30 2022-08-04 E-Therapeutics Plc Conjugated oligonucleotide compounds, methods of making and uses thereof
EP4175965B1 (en) * 2021-01-30 2024-03-13 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
TW202305131A (en) 2021-02-12 2023-02-01 美商艾拉倫製藥股份有限公司 SUPEROXIDE DISMUTASE 1 (SOD1) iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING OR PREVENTING SUPEROXIDE DISMUTASE 1- (SOD1-) ASSOCIATED NEURODEGENERATIVE DISEASES
EP4294449A1 (en) * 2021-02-18 2023-12-27 Oneglobe Holdings Limited Compositions for conjugating oligonucleotides and carbohydrates
CN117222739A (en) 2021-02-25 2023-12-12 阿尔尼拉姆医药品有限公司 Prion protein (PRNP) IRNA compositions and methods of use thereof
WO2022182574A1 (en) 2021-02-26 2022-09-01 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
AU2022231003A1 (en) 2021-03-04 2023-09-14 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
WO2022189861A1 (en) 2021-03-08 2022-09-15 Tollys Carbohydrate conjugates of tlr3 ligands and uses thereof
EP4305168A1 (en) 2021-03-08 2024-01-17 Les Laboratoires Servier Antisense oligonucleotides for inhibiting alpha-synuclein expression
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
JP2024512635A (en) 2021-03-29 2024-03-19 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 Huntingtin (HTT) iRNA agent composition and method 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
KR20240001207A (en) 2021-04-26 2024-01-03 알닐람 파마슈티칼스 인코포레이티드 Transmembrane protease, serine 6 (TMPRSS6) iRNA compositions and methods of using the same
WO2022232343A1 (en) 2021-04-29 2022-11-03 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
WO2022256395A1 (en) 2021-06-02 2022-12-08 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2022256290A2 (en) 2021-06-04 2022-12-08 Alnylam Pharmaceuticals, Inc. HUMAN CHROMOSOME 9 OPEN READING FRAME 72 (C9ORF72) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
AR126070A1 (en) 2021-06-08 2023-09-06 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR TREATING OR PREVENTING STARGARDT DISEASE AND/OR DISORDERS ASSOCIATED WITH RETINOL BORDER PROTEIN 4 (RBP4)
CN117500815A (en) 2021-06-18 2024-02-02 Ionis制药公司 Compounds and methods for reducing IFNAR1 expression
US11702659B2 (en) 2021-06-23 2023-07-18 University Of Massachusetts Optimized anti-FLT1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
IL309503A (en) * 2021-06-24 2024-02-01 Lilly Co Eli Novel therapeutic delivery moieties and uses thereof
WO2022271808A1 (en) * 2021-06-24 2022-12-29 Eli Lilly And Company Novel rna therapeutics 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
CA3225469A1 (en) 2021-06-30 2023-01-05 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
WO2023274395A1 (en) * 2021-07-02 2023-01-05 上海拓界生物医药科技有限公司 Nucleic acid ligand and conjugate thereof, and preparation method therefor and use thereof
CA3225573A1 (en) 2021-07-08 2023-01-12 Nippon Shinyaku Co., Ltd. Precipitation suppressing agent
IL310003A (en) 2021-07-08 2024-03-01 Nippon Shinyaku Co Ltd Nephrotoxicity reducing agent
TW202308664A (en) 2021-07-08 2023-03-01 日商日本新藥股份有限公司 Nephrotoxicity-reducing agent
TW202333748A (en) 2021-07-19 2023-09-01 美商艾拉倫製藥股份有限公司 Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
IL309905A (en) 2021-07-23 2024-03-01 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
IL310244A (en) 2021-08-03 2024-03-01 Alnylam Pharmaceuticals Inc Transthyretin (ttr) 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
CA3228255A1 (en) 2021-08-04 2023-02-09 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing angiotensinogen (agt)
AU2022328347A1 (en) 2021-08-13 2024-02-08 Alnylam Pharmaceuticals, Inc. Factor xii (f12) irna compositions and methods of use thereof
CA3229020A1 (en) 2021-09-14 2023-03-23 Stella PELENGARIS 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)
AU2022348141A1 (en) * 2021-09-18 2024-04-04 Genoval Therapeutics Co., Ltd. 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
AU2022349576A1 (en) * 2021-09-23 2024-02-15 Shanghai Argo Biopharmaceutical Co., Ltd. Multivalent ligand clusters with diamine scaffold for targeted delivery of therapeutic agents
WO2023069603A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
WO2023076451A1 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
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
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
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
WO2023109935A1 (en) * 2021-12-16 2023-06-22 上海拓界生物医药科技有限公司 Dsrna, and preparation method therefor and use thereof
WO2023111210A1 (en) 2021-12-17 2023-06-22 F. Hoffmann-La Roche Ag Combination of oligonucleotides for modulating rtel1 and fubp1
TW202340469A (en) * 2022-01-20 2023-10-16 大陸商上海拓界生物醫藥科技有限公司 Dsrna, its application and preparation method
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
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))
CN116917477B (en) * 2022-01-30 2024-04-09 大睿生物医药科技(上海)有限公司 Targeting ligands containing N-acetylgalactosamine
WO2023156652A1 (en) 2022-02-21 2023-08-24 F. Hoffmann-La Roche Ag 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
US11879125B2 (en) 2022-03-16 2024-01-23 Empirico Inc. 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
CN117568313A (en) * 2024-01-15 2024-02-20 上海贝斯昂科生物科技有限公司 Gene editing composition and use thereof

Family Cites Families (373)

* 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
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of 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
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4668777A (en) 1981-03-27 1987-05-26 University Patents, Inc. Phosphoramidite nucleoside compounds
US4973679A (en) 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
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
US5405938A (en) 1989-12-20 1995-04-11 Anti-Gene Development Group Sequence-specific binding polymers for duplex nucleic acids
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5185444A (en) 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
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
DE3855864T2 (en) 1987-11-30 1997-09-25 Univ Iowa Res Found DNA MOLECULES STABILIZED BY MODIFICATIONS ON THE 3'-TERMINAL PHOSPHODIESTERBINDING, THEIR USE AS NUCLEIC ACID PROBE AND AS A THERAPEUTIC AGENT 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
JPH03503894A (en) 1988-03-25 1991-08-29 ユニバーシィティ オブ バージニア アランミ パテンツ ファウンデイション Oligonucleotide N-alkylphosphoramidate
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
US5721218A (en) 1989-10-23 1998-02-24 Gilead Sciences, Inc. Oligonucleotides with inverted polarity
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
EP0497875B1 (en) 1989-10-24 2000-03-22 Isis Pharmaceuticals, Inc. 2' modified oligonucleotides
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
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5623065A (en) 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US5457191A (en) 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5859221A (en) 1990-01-11 1999-01-12 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US7101993B1 (en) 1990-01-11 2006-09-05 Isis Pharmaceuticals, Inc. Oligonucleotides containing 2′-O-modified purines
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US6005087A (en) 1995-06-06 1999-12-21 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US5587361A (en) 1991-10-15 1996-12-24 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
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
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
US5321131A (en) 1990-03-08 1994-06-14 Hybridon, Inc. Site-specific functionalization of oligodeoxynucleotides for non-radioactive labelling
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
GB9009980D0 (en) 1990-05-03 1990-06-27 Amersham Int Plc Phosphoramidite derivatives,their preparation and the use thereof in the incorporation of reporter groups on synthetic oligonucleotides
ES2116977T3 (en) 1990-05-11 1998-08-01 Microprobe Corp SOLID SUPPORTS FOR NUCLEIC ACID HYBRIDIZATION TESTS AND METHODS TO IMMOBILIZE OLIGONUCLEOTIDES IN A COVALENT WAY.
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
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
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
DE69126530T2 (en) 1990-07-27 1998-02-05 Isis Pharmaceuticals Inc NUCLEASE RESISTANT, PYRIMIDINE MODIFIED OLIGONUCLEOTIDES THAT DETECT AND MODULE GENE EXPRESSION
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
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
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
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
EP0541722B1 (en) 1990-08-03 1995-12-20 Sterling Winthrop Inc. Compounds and methods for inhibiting gene expression
US5177196A (en) 1990-08-16 1993-01-05 Microprobe Corporation Oligo (α-arabinofuranosyl nucleotides) and α-arabinofuranosyl precursors thereof
US5214134A (en) 1990-09-12 1993-05-25 Sterling Winthrop Inc. Process of linking nucleosides with a siloxane bridge
US5561225A (en) 1990-09-19 1996-10-01 Southern Research Institute Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
US5596086A (en) 1990-09-20 1997-01-21 Gilead Sciences, Inc. Modified internucleoside linkages having one nitrogen and two carbon atoms
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
DE59208572D1 (en) 1991-10-17 1997-07-10 Ciba Geigy Ag Bicyclic nucleosides, oligonucleotides, processes for their preparation and intermediates
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
TW393513B (en) 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
ATE226093T1 (en) 1991-11-26 2002-11-15 Isis Pharmaceuticals Inc INCREASED FORMATION OF TRIPLE AND DOUBLE HELICES FROM OLIGOMERS WITH MODIFIED PYRIMIDINES
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl 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
US5700922A (en) 1991-12-24 1997-12-23 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
DK0618925T4 (en) * 1991-12-24 2012-07-09 Isis Pharmaceuticals Inc Antisense oligonucleotides
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
DE69316369D1 (en) 1992-07-27 1998-02-19 Hybridon Inc Oligonukleotid alkylphosphonothiate
EP0673559A1 (en) 1992-12-14 1995-09-27 Honeywell Inc. Motor system with individually controlled redundant windings
DE69400208T2 (en) 1993-01-25 1996-11-28 Hybridon Inc OLIONUCLEOTIDALKYLPHOSPHONATES AND PHOSPHONOTHIOATES
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
GB9304620D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Compounds
GB9304618D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
ATE155467T1 (en) 1993-03-30 1997-08-15 Sanofi Sa ACYCLIC NUCLEOSIDE ANALOGUES AND OLIGONUCLEOTIDE SEQUENCES CONTAINING THEM
JPH08508491A (en) 1993-03-31 1996-09-10 スターリング ウインスロップ インコーポレイティド Oligonucleotides with phosphodiester bonds replaced by amide bonds
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
JP3585238B2 (en) 1993-12-09 2004-11-04 トーマス ジェファーソン ユニバーシティー Compounds and methods for site-directed mutagenesis in eukaryotic cells
US5446137B1 (en) 1993-12-09 1998-10-06 Behringwerke Ag Oligonucleotides containing 4'-substituted nucleotides
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
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
US6172045B1 (en) 1994-12-07 2001-01-09 Neorx Corporation Cluster clearing agents
US6908903B1 (en) 1994-12-07 2005-06-21 Aletheon Pharmaceuticals, Inc. Cluster clearing agents
US5652356A (en) 1995-08-17 1997-07-29 Hybridon, Inc. Inverted chimeric and hybrid oligonucleotides
JP2000501414A (en) 1995-11-22 2000-02-08 ザ・ジョンズ・ホプキンス・ユニバーシティー Ligand enhances cellular uptake of biomolecules
US20030119724A1 (en) 1995-11-22 2003-06-26 Ts`O Paul O.P. 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
WO2005121371A2 (en) 2004-06-03 2005-12-22 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
CN1231675A (en) 1996-09-26 1999-10-13 味之素株式会社 Modified physiologically active proteins and medicinal compositions containing the same
US6770748B2 (en) 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
JP3756313B2 (en) 1997-03-07 2006-03-15 武 今西 Novel bicyclonucleosides and oligonucleotide analogues
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
EP2341057A3 (en) 1997-09-12 2011-11-23 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
JP2004505885A (en) 1998-08-19 2004-02-26 ノース アメリカン ワクチン, インコーポレイテッド Immunogenic β-propionamide linked polysaccharide-protein conjugates useful as vaccines produced using N-acryloylated polysaccharides
PL346645A1 (en) * 1998-08-19 2002-02-25 North American Vaccine IMMUNOGENIC β-PROPIONAMIDO-LINKED POLYSACCHARIDE PROTEIN CONJUGATE USEFUL AS A VACCINE PRODUCED USING AN N-ACRYLOYLATED POLYSACCHARIDE
US6166239A (en) 1998-09-04 2000-12-26 Isis Pharmaceuticals, Inc. Oligonucleotide protecting groups
AU758956B2 (en) 1999-02-12 2003-04-03 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
KR20010112944A (en) 1999-04-21 2001-12-22 이곤 이 버그 Methods and compositions for inhibiting the function of polynucleotide sequences
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
US20080281041A1 (en) 1999-06-07 2008-11-13 Rozema David B Reversibly Masked Polymers
US8137695B2 (en) 2006-08-18 2012-03-20 Arrowhead Madison Inc. Polyconjugates for in vivo delivery of polynucleotides
US8541548B2 (en) 1999-06-07 2013-09-24 Arrowhead Madison Inc. Compounds and methods for reversible modification of biologically active molecules
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
WO2001005825A2 (en) 1999-07-16 2001-01-25 Hyseq, Inc. Nucleic acid sequences encoding putative angiopoietin proteins
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
BR0016257A (en) 1999-12-09 2002-08-20 Method for testing an effect of a substance as a therapeutic or preventive agent for hyperlipidemia. polynucleotide, dna, antibody that specifically recognizes a polypeptide, kit to test a therapeutic or preventive agent for hyperlipidemia, use of a polynucleotide, use of a polypeptide, use of an antibody that specifically recognizes a polypeptide, dna or rna and therapeutic agent for hyperlipidemia.
DE60027203T2 (en) 1999-12-30 2007-01-04 K.U. Leuven Research & Development CYCLOHEXENE NUCLEIC
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
US7179796B2 (en) 2000-01-18 2007-02-20 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
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
US7833992B2 (en) 2001-05-18 2010-11-16 Merck Sharpe & Dohme Conjugates and compositions for cellular delivery
WO2001060860A2 (en) 2000-02-17 2001-08-23 Millennium Predictive Medicine, Inc. Genes differentially expressed in human prostate cancer and their use
US7053199B2 (en) 2000-08-29 2006-05-30 Takeshi Imanishi Nucleoside analogs and oligonucleotide derivatives containing these analogs
US6426220B1 (en) 2000-10-30 2002-07-30 Isis Pharmaceuticals, Inc. Antisense modulation of calreticulin expression
EP1355672A2 (en) 2000-12-01 2003-10-29 Cell Works Inc. Conjugates of glycosylated/galactosylated peptide, bifunctional linker, and nucleotidic monomers/polymers, and related compositions and methods of use
WO2002087541A1 (en) 2001-04-30 2002-11-07 Protiva Biotherapeutics Inc. Lipid-based formulations for gene transfer
WO2002101039A1 (en) 2001-06-08 2002-12-19 Sankyo Company, Limited Method of testing drug for treating or preventing diseases such as hyperlipemia
JP2005504020A (en) 2001-07-03 2005-02-10 アイシス・ファーマシューティカルス・インコーポレーテッド Nuclease resistant chimeric oligonucleotide
US20030175906A1 (en) 2001-07-03 2003-09-18 Muthiah Manoharan Nuclease resistant chimeric oligonucleotides
US20030158403A1 (en) 2001-07-03 2003-08-21 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US7425545B2 (en) * 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. 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
WO2003020739A2 (en) 2001-09-04 2003-03-13 Exiqon A/S Novel lna compositions and uses thereof
US7439043B2 (en) 2001-10-10 2008-10-21 Neose Technologies, Inc. Galactosyl nucleotide sugars
IL161542A0 (en) 2001-11-16 2004-09-27 Genentech Inc Composition comprising and method of using angiopoietin-like protein 3 angplt3
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)
WO2004011624A2 (en) 2002-07-31 2004-02-05 Nucleonics, Inc. Double stranded rna structures and constructs, and methods for generating and using the same
US8729036B2 (en) 2002-08-07 2014-05-20 University Of Massachusetts Compositions for RNA interference and methods of use thereof
AU2003260289A1 (en) 2002-09-11 2004-04-30 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
AU2003290598A1 (en) 2002-11-05 2004-06-03 Isis Pharmaceuticals, Inc. Modified oligonucleotides for use in rna interference
US7696345B2 (en) 2002-11-05 2010-04-13 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
US7511131B2 (en) 2002-11-13 2009-03-31 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
CA2505801A1 (en) 2002-11-13 2004-05-27 Rosanne Crooke Antisense modulation of apolipoprotein b expression
EP2284266B1 (en) 2002-11-14 2013-11-06 Thermo Fisher Scientific Biosciences Inc. siRNA targeting tp53
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
EP2474631B1 (en) 2002-12-20 2014-02-12 Celera Corporation Genetic polymorphisms associated with myocardial infarction, methods of detection and uses thereof
JP4152955B2 (en) 2003-01-09 2008-09-17 ポステック・ファウンデーション 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
WO2004090108A2 (en) * 2003-04-03 2004-10-21 Alnylam Pharmaceuticals Irna conjugates
ATE479752T1 (en) * 2003-03-07 2010-09-15 Alnylam Pharmaceuticals Inc THERAPEUTIC COMPOSITIONS
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
EP2660322A3 (en) 2003-04-17 2013-11-13 Alnylam Pharmaceuticals Inc. Modified iRNA agents
US7723509B2 (en) 2003-04-17 2010-05-25 Alnylam Pharmaceuticals IRNA agents with biocleavable tethers
US7851615B2 (en) 2003-04-17 2010-12-14 Alnylam Pharmaceuticals, Inc. Lipophilic conjugated iRNA agents
US7750142B2 (en) 2003-04-28 2010-07-06 Isis Pharmaceuticals, Inc. Modulation of glucagon receptor expression
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
WO2004101619A1 (en) 2003-05-15 2004-11-25 Shionogi Co., Ltd. Rational design and synthesis of functional glycopeptide
WO2004106356A1 (en) 2003-05-27 2004-12-09 Syddansk Universitet Functionalized nucleotide derivatives
ES2382807T3 (en) 2003-08-28 2012-06-13 Takeshi Imanishi New artificial nucleic acids of the N-O link type with cross-linking
AU2004274021B2 (en) 2003-09-18 2009-08-13 Isis Pharmaceuticals, Inc. 4'-thionucleosides and oligomeric compounds
NZ545134A (en) * 2003-09-18 2009-06-26 Lilly Co Eli Modulation of eIF4E expression
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
JP2007520222A (en) 2004-01-20 2007-07-26 アイシス ファーマシューティカルズ インコーポレイテッド Regulation 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
CN101052717A (en) 2004-05-11 2007-10-10 α基因株式会社 Polynucleotide causing RNA interfere and method of regulating gene expression with the use of the same
RU2380411C2 (en) 2004-07-20 2010-01-27 Дженентек, Инк. Method for inhibition of hepatocyte proliferation, method for inhibition of hepatocyte cell adhesion and method for inhibition of biological activity angptl4 in hepatocytes or hepatocyte precursors
EP2361931B1 (en) 2004-07-20 2017-12-06 Genentech, Inc. Inhibitors of angiopoietin-like 4 protein, combinations, and their use
JP5192234B2 (en) 2004-08-10 2013-05-08 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 Chemically modified oligonucleotide
PL2409713T3 (en) * 2004-08-10 2016-02-29 Kastle Therapeutics Llc Oligonucleotides for use in modulating lipoprotein and cholesterol levels in humans
WO2006031461A2 (en) 2004-09-09 2006-03-23 Isis Pharmaceuticals, Inc. 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
EP1841867A1 (en) 2005-01-24 2007-10-10 Avaris AB COMPLEX CONTAINING SiRNA, ShRNA OR ANTISENSE MOLECULE AND FUNCTIONAL ENTITY, FOR IMPROVED SPECIFICITY AND DELIVERY
EA200800823A1 (en) * 2005-09-15 2008-10-30 Сантарис Фарма А/С COMPOUNDS-ANTAGONISTS OF RNA TO INHIBIT EXPRESSION OF ARO-B100
EA200800868A1 (en) 2005-09-19 2008-10-30 ДЖОНСОН ЭНД ДЖОНСОН ФАРМАСЬЮТИКАЛ РИСЕРЧ ЭНД ДИВЕЛОПМЕНТ, Эл. Эл. Си. MODULATION OF THE GLUCOCORTICOID RECEPTOR EXPRESSION
EP2096170B1 (en) 2005-09-19 2011-08-10 Isis Pharmaceuticals, Inc. Modulation of glucagon receptor expression
US7951934B2 (en) 2006-01-26 2011-05-31 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
JP5342881B2 (en) 2006-01-27 2013-11-13 アイシス ファーマシューティカルズ, インコーポレーテッド 6-modified bicyclic nucleic acid analogues
WO2007134014A2 (en) * 2006-05-05 2007-11-22 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression of gcgr
ES2389737T3 (en) 2006-05-11 2012-10-31 Isis Pharmaceuticals, Inc. 5 'modified bicyclic nucleic acid analogs
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
AU2007281082A1 (en) 2006-08-04 2008-02-07 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
WO2008036825A2 (en) 2006-09-22 2008-03-27 Dharmacon, Inc. Duplex oligonucleotide complexes and methods for gene silencing by rna interference
ES2526295T5 (en) 2006-10-18 2021-05-04 Ionis Pharmaceuticals Inc Antisense compounds
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
JP2010510807A (en) 2006-11-27 2010-04-08 アイシス ファーマシューティカルズ, インコーポレーテッド Methods for treating hypercholesterolemia
MX2009006082A (en) 2006-12-08 2009-08-18 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
EP2125852B1 (en) 2007-02-15 2016-04-06 Ionis Pharmaceuticals, Inc. 5'-substituted-2'-f modified nucleosides and oligomeric compounds prepared therefrom
EP2604283A1 (en) 2007-02-16 2013-06-19 KTB Tumorforschungsgesellschaft mbH Receptor And Antigen Targeted Prodrug
CA2678774A1 (en) 2007-03-01 2008-09-04 Advanced Vision Therapies, Inc. Treatment of diseases characterized by inflammation
EP2126085A2 (en) 2007-03-02 2009-12-02 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
CA2688321A1 (en) 2007-05-30 2008-12-11 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
WO2008154401A2 (en) 2007-06-08 2008-12-18 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
US20090004140A1 (en) 2007-06-26 2009-01-01 Yao-Ling Qiu 4-substituted pyrrolidine as anti-infectives
US8278283B2 (en) 2007-07-05 2012-10-02 Isis Pharmaceuticals, Inc. 6-disubstituted or unsaturated bicyclic nucleic acid analogs
AU2008286771B2 (en) 2007-08-15 2013-08-15 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
AU2008308691B2 (en) * 2007-10-01 2013-11-07 Isis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
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
CA2708173C (en) 2007-12-04 2016-02-02 Alnylam Pharmaceuticals, Inc. Targeting lipids
CA2713379A1 (en) 2008-01-31 2009-11-05 Alnylam Pharmaceuticals, Inc. Optimized methods for delivery of dsrna targeting the pcsk9 gene
EP2265627A2 (en) 2008-02-07 2010-12-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexitol nucleic acid analogs
WO2009142822A2 (en) 2008-03-26 2009-11-26 Alnylam Pharmaceuticals, Inc. 2-f modified rna interference agents
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
US20110237646A1 (en) 2008-08-07 2011-09-29 Isis Pharmaceuticals, Inc. Modulation of transthyretin expression for the treatment of cns related disorders
EP3587434A1 (en) 2008-09-23 2020-01-01 Alnylam Pharmaceuticals Inc. Chemical modifications of monomers and oligonucleotides with click components for conjugation with ligands
US8604192B2 (en) 2008-09-24 2013-12-10 Isis Pharmaceuticals, Inc. Cyclohexenyl nucleic acids analogs
US8501805B2 (en) 2008-09-24 2013-08-06 Isis Pharmaceuticals, Inc. Substituted alpha-L-bicyclic nucleosides
AU2009305636A1 (en) 2008-10-15 2010-04-22 Ionis Pharmaceuticals, Inc. Modulation of Factor 11 expression
NZ592867A (en) * 2008-10-20 2013-05-31 Alnylam Pharmaceuticals Inc 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
WO2010048585A2 (en) 2008-10-24 2010-04-29 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
KR101967417B1 (en) 2008-11-10 2019-04-10 알닐람 파마슈티칼스 인코포레이티드 Novel lipids and compositions for the delivery of therapeutics
CA2750561C (en) 2009-01-26 2017-10-10 Protiva Biotherapeutics, Inc. Compositions and methods for silencing apolipoprotein c-iii expression
AU2010208035B2 (en) 2009-01-29 2016-06-23 Arbutus Biopharma Corporation Improved lipid formulation for the delivery of nucleic acids
EP3424939A1 (en) 2009-03-02 2019-01-09 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
AU2010236286B2 (en) 2009-04-15 2013-06-06 Isis Pharmaceuticals, Inc. Modulation of inflammatory responses by Factor XI
NZ596186A (en) * 2009-05-05 2014-03-28 Alnylam Pharmaceuticals Inc Lipid compositions
US9200276B2 (en) 2009-06-01 2015-12-01 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent RNA interference, compositions and methods of use thereof
KR101766408B1 (en) 2009-06-10 2017-08-10 알닐람 파마슈티칼스 인코포레이티드 Improved lipid formulation
US8273869B2 (en) 2009-06-15 2012-09-25 Alnylam Pharmaceuticals, Inc. Lipid formulated dsRNA targeting the PCSK9 gene
BR112012000421A2 (en) 2009-07-06 2019-09-24 Alnylam Pharmaceuticals Inc compositions and methods for enhancing the production of a biological product.
WO2011005860A2 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. 5' phosphate mimics
WO2011005861A1 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. Oligonucleotide end caps
WO2011008995A1 (en) 2009-07-16 2011-01-20 Isis Pharmaceuticals, Inc. Modulation of factor 7 expression
WO2011017521A2 (en) 2009-08-06 2011-02-10 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
TWI458493B (en) 2009-09-25 2014-11-01 Iner Aec Executive Yuan Novel liver-targeting agents and their synthesis
AU2010306639B2 (en) 2009-10-16 2017-02-16 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
EP2496238A4 (en) * 2009-11-03 2013-10-02 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
WO2011100131A2 (en) 2010-01-28 2011-08-18 Alnylam Pharmacuticals, 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
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
JP2013528665A (en) 2010-03-26 2013-07-11 メルサナ セラピューティックス, インコーポレイテッド Modified polymers for delivery of polynucleotides, methods for their production, and methods of their use
US9102938B2 (en) 2010-04-01 2015-08-11 Alnylam Pharmaceuticals, Inc. 2′ and 5′ modified monomers and oligonucleotides
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
US10913767B2 (en) 2010-04-22 2021-02-09 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
EP2625186B1 (en) 2010-04-28 2016-07-27 Ionis Pharmaceuticals, Inc. 5' modified nucleosides and oligomeric compounds prepared therefrom
KR101869570B1 (en) 2010-04-28 2018-06-20 아이오니스 파마수티컬즈, 인코포레이티드 Modified nucleosides and oligomeric compounds prepared therefrom
WO2011139917A1 (en) * 2010-04-29 2011-11-10 Isis Pharmaceuticals, Inc. Modulation of transthyretin expression
WO2011163121A1 (en) 2010-06-21 2011-12-29 Alnylam Pharmaceuticals, Inc. Multifunctional copolymers for nucleic acid delivery
WO2012037254A1 (en) 2010-09-15 2012-03-22 Alnylam Pharmaceuticals, Inc. MODIFIED iRNA AGENTS
US8603994B2 (en) 2010-11-11 2013-12-10 Valted, Llc 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
KR20130108655A (en) 2010-12-29 2013-10-04 에프. 호프만-라 로슈 아게 Small molecule conjugates for intracellular delivery of nucleic acids
EP3467109A1 (en) 2011-02-08 2019-04-10 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
WO2012135736A2 (en) 2011-04-01 2012-10-04 Isis Pharmaceuticals, Inc. Modulation of signal transducer and activator of transcription 3 (stat3) expression
CN103547588B (en) * 2011-04-13 2016-06-29 Isis制药公司 The antisense that PTP1B expresses regulates
US20120295961A1 (en) 2011-04-21 2012-11-22 Swayze Eric E Modulation of hepatitis b virus (hbv) expression
MX340408B (en) * 2011-04-21 2016-07-07 Ionis Pharmaceuticals Inc Modulation of hepatitis b virus (hbv) expression.
JP6203707B2 (en) * 2011-04-27 2017-09-27 アイオーニス ファーマシューティカルズ, インコーポレーテッドIonis Pharmaceuticals,Inc. Regulation of apolipoprotein CIII (APOCIII) expression
WO2012174154A1 (en) 2011-06-13 2012-12-20 Isis Pharmaceuticals, Inc. Modulation of inflammatory responses by factor vii
EP2721156B1 (en) 2011-06-16 2016-12-21 Ionis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
KR102395085B1 (en) * 2011-06-21 2022-05-09 알닐람 파마슈티칼스 인코포레이티드 Angiopoietin-like 3(angptl3) irna compostions and methods of use thereof
CN112111492A (en) 2011-06-21 2020-12-22 阿尔尼拉姆医药品有限公司 Compositions and methods for inhibiting expression of apolipoprotein C-III (APOC3) gene
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
BR112014004585A2 (en) 2011-08-26 2017-06-13 Arrowhead Res Corp polyvinyl ester polymers for in vivo nucleic acid release
DK2751270T3 (en) 2011-08-29 2018-10-29 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
CN103906838A (en) 2011-10-25 2014-07-02 Isis制药公司 Antisense modulation of GCCR expression
CN104080794B (en) 2011-11-18 2018-03-16 阿尔尼拉姆医药品有限公司 RNAi reagents, composition and its application method for treating transthyretin (TTR) relevant disease
WO2013119979A1 (en) 2012-02-08 2013-08-15 Isis Pharmaceuticals, Inc. Methods and compositions for modulating factor vii expression
WO2013142514A1 (en) 2012-03-19 2013-09-26 Isis 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
WO2013159109A1 (en) 2012-04-20 2013-10-24 Isis Pharmaceuticals, Inc. Modulation of hepatitis b virus (hbv) expression
EP2839006B1 (en) 2012-04-20 2018-01-03 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
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
US20160002624A1 (en) 2012-05-17 2016-01-07 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions
RU2624028C2 (en) 2012-05-24 2017-06-30 Ионис Фармасьютикалз, Инк. Methods and compositions for modeling expression of apolipoprotein (a)
WO2013192233A1 (en) 2012-06-18 2013-12-27 Isis Pharmaceuticals, Inc. Compounds and method for improved cellular uptake of antisense compounds
EP2879718B1 (en) 2012-08-06 2023-06-07 Alnylam Pharmaceuticals, Inc. Processes for the preparation of carbohydrate conjugated rna agents
US9695418B2 (en) 2012-10-11 2017-07-04 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleosides and uses thereof
US20150291958A1 (en) 2012-11-15 2015-10-15 Roche Innovation Center Copenhagen A/S Anti apob antisense conjugate compounds
WO2014118272A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Antimir-122 oligonucleotide carbohydrate conjugates
DK2951305T3 (en) 2013-01-30 2018-10-29 Hoffmann La Roche LNA oligonucleotide KULHYDRATKONJUGATER
SG10201906382QA (en) * 2013-05-01 2019-08-27 Ionis Pharmaceuticals Inc Compositions and methods for modulating hbv and ttr expression
AU2013389474B2 (en) 2013-05-16 2018-11-15 Sumitomo Pharma Co., Ltd. Transplantation adjuvant in cell therapy using neural progenitor cells
CA2916252A1 (en) 2013-06-21 2014-12-24 Isis Pharmaceuticals, Inc. Compositions and methods for modulation of target nucleic acids
BR112015032432B1 (en) 2013-06-27 2023-02-07 Roche Innovation Center Copenhagen A/S ANTI-SENSE OLIGOMER, ANTI-SENSE OLIGONUCLEOTIDE CONJUGATES, PHARMACEUTICAL COMPOSITION, USE THEREOF FOR THE TREATMENT OF HYPERCHOLESTEROLEMIA OR RELATED DISORDERS, AND IN VITRO METHOD FOR REDUCING THE EXPRESSION LEVELS AND/OR THE ACTIVITY OF PCSK9 IN A CELL
KR102236784B1 (en) 2013-07-02 2021-04-05 아이오니스 파마수티컬즈, 인코포레이티드 Modulators of growth hormone receptor
CA2917161C (en) 2013-07-11 2023-12-12 Alnylam Pharmaceuticals, Inc. Oligonucleotide-ligand conjugates and process for their preparation
PE20190354A1 (en) 2013-09-13 2019-03-07 Ionis Pharmaceuticals Inc COMPLEMENT B FACTOR MODULATORS
WO2015042447A1 (en) 2013-09-20 2015-03-26 Isis Pharmaceuticals, Inc. Targeted therapeutic nucleosides and their use
RU2016122168A (en) 2013-11-14 2017-12-19 Рош Инновейшен Сентер Копенгаген А/С ANTI-SENSE CONJUGATES AIMED AT APOLIPOPROTEIN B
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
AU2015252917B2 (en) 2014-05-01 2019-09-26 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating PKK expression
EP3137604B1 (en) 2014-05-01 2020-07-15 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating growth hormone receptor expression
SI3137605T1 (en) 2014-05-01 2021-02-26 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating angiopoietin-like 3 expression
EP3811977A1 (en) 2014-05-01 2021-04-28 Ionis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
SG11201608502TA (en) 2014-05-01 2016-11-29 Ionis Pharmaceuticals Inc Compositions and methods for modulating complement factor b expression
US10570169B2 (en) 2014-05-22 2020-02-25 Ionis 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
PE20181180A1 (en) * 2015-11-06 2018-07-20 Ionis Pharmaceuticals Inc MODULATE THE EXPRESSION OF APOLIPOPROTEIN (a)

Also Published As

Publication number Publication date
RU2699985C2 (en) 2019-09-11
WO2014179629A8 (en) 2016-06-02
CN108064162A (en) 2018-05-22
SG10201801813YA (en) 2018-04-27
PL2992009T3 (en) 2020-11-30
RU2018112167A (en) 2019-03-07
RU2015151200A (en) 2019-01-14
AU2019204784A1 (en) 2019-07-25
CR20150612A (en) 2016-03-03
US9181549B2 (en) 2015-11-10
AU2022202770A1 (en) 2022-05-19
BR112015027319A8 (en) 2018-01-02
SI2992098T1 (en) 2019-06-28
RU2015151200A3 (en) 2019-01-14
RU2015151199A3 (en) 2018-03-27
IL261901A (en) 2018-10-31
IL273312A (en) 2020-04-30
AU2017203436A1 (en) 2017-06-08
CA2921509A1 (en) 2014-11-06
KR102482890B1 (en) 2022-12-30
US9957504B2 (en) 2018-05-01
CN113293163A (en) 2021-08-24
ES2778442T3 (en) 2020-08-10
JP2020007361A (en) 2020-01-16
RU2019110030A (en) 2019-05-06
NZ631537A (en) 2017-05-26
KR102235678B1 (en) 2021-04-05
JP2020039355A (en) 2020-03-19
MY178929A (en) 2020-10-23
ES2819213T3 (en) 2021-04-15
IL283660A (en) 2021-07-29
EP2992009A4 (en) 2016-12-28
RU2015151204A3 (en) 2018-03-27
AU2014259756B2 (en) 2017-02-23
DK2992098T3 (en) 2019-06-17
HK1221475A1 (en) 2017-06-02
RS60796B1 (en) 2020-10-30
MX2015015234A (en) 2016-10-03
MX2015015239A (en) 2016-10-03
BR112015027322A8 (en) 2018-01-02
JP2020074787A (en) 2020-05-21
CN110079524A (en) 2019-08-02
MX2015015220A (en) 2016-01-12
MY198359A (en) 2023-08-28
US20150126720A1 (en) 2015-05-07
IL242126B (en) 2019-01-31
AU2017200365B2 (en) 2018-11-08
BR112015027369A2 (en) 2017-09-26
CN110042098B (en) 2023-02-24
PH12019501191A1 (en) 2021-03-01
SG11201508870VA (en) 2015-11-27
AU2014259755B2 (en) 2018-08-30
US10927372B2 (en) 2021-02-23
RU2018136140A (en) 2018-12-17
US9714421B2 (en) 2017-07-25
IL274064B (en) 2021-06-30
KR20180051678A (en) 2018-05-16
ME03390B (en) 2020-01-20
EP2991656B1 (en) 2019-12-18
JP2024010070A (en) 2024-01-23
BR112015027321A8 (en) 2018-01-02
RU2697152C2 (en) 2019-08-12
CN105377887B (en) 2020-11-03
IL296543A (en) 2022-11-01
EP2992009A1 (en) 2016-03-09
US20140343123A1 (en) 2014-11-20
AU2014259750B2 (en) 2019-02-28
US20150126718A1 (en) 2015-05-07
EA031393B1 (en) 2018-12-28
KR20220108195A (en) 2022-08-02
AU2019204784B2 (en) 2022-01-27
JP2016526874A (en) 2016-09-08
US9932581B2 (en) 2018-04-03
IL284593B2 (en) 2023-02-01
AU2017200950B2 (en) 2019-01-17
EP3524680B1 (en) 2020-11-11
EP2991661A4 (en) 2017-02-15
JP6769866B2 (en) 2020-10-14
MX2020002184A (en) 2020-07-14
CA2921514A1 (en) 2014-11-06
BR112015027319A2 (en) 2017-09-26
AU2014259756A1 (en) 2015-10-22
JP2016523515A (en) 2016-08-12
JP2018183184A (en) 2018-11-22
CN105392488B (en) 2021-04-30
KR20210129257A (en) 2021-10-27
KR20230006933A (en) 2023-01-11
DOP2016000287A (en) 2017-02-15
US20200224198A1 (en) 2020-07-16
US9932580B2 (en) 2018-04-03
JP2020039354A (en) 2020-03-19
KR102212275B1 (en) 2021-02-05
PT2992098T (en) 2019-07-05
CN105392488A (en) 2016-03-09
RU2015151202A3 (en) 2018-03-27
CY1123369T1 (en) 2021-12-31
CL2016002262A1 (en) 2017-06-09
IL270464B (en) 2021-07-29
WO2014179629A3 (en) 2015-01-22
US20190055554A1 (en) 2019-02-21
EP2991656A4 (en) 2017-02-22
WO2014179627A9 (en) 2015-02-26
KR102424855B1 (en) 2022-07-26
AU2014259750A1 (en) 2015-10-22
HK1221404A1 (en) 2017-06-02
IL263843B (en) 2020-03-31
JP2022017514A (en) 2022-01-25
RU2015151203A (en) 2017-06-02
JP6456362B2 (en) 2019-01-23
NZ725538A (en) 2021-02-26
AU2019204784C1 (en) 2022-11-03
PE20161430A1 (en) 2017-01-06
AU2014259755A1 (en) 2015-10-22
MX2015015264A (en) 2016-08-12
EP2991661A1 (en) 2016-03-09
EP2992097A4 (en) 2017-01-04
KR20210037752A (en) 2021-04-06
US20150176007A1 (en) 2015-06-25
MX2019010443A (en) 2019-10-17
US20160076030A1 (en) 2016-03-17
JP6866459B2 (en) 2021-04-28
AU2019203674A1 (en) 2019-06-27
EP3546579A1 (en) 2019-10-02
CA2921162A1 (en) 2014-11-06
KR20210014758A (en) 2021-02-09
US20160076032A1 (en) 2016-03-17
US20160090595A1 (en) 2016-03-31
AU2021204244A1 (en) 2021-07-22
AU2018267625B2 (en) 2020-09-10
BR112015027377A8 (en) 2017-10-03
AU2024200296A1 (en) 2024-02-08
KR20230113835A (en) 2023-08-01
NZ740338A (en) 2022-04-29
EP2992097B1 (en) 2019-11-06
DK3524680T3 (en) 2020-12-14
EP4155403A1 (en) 2023-03-29
MX2021008899A (en) 2021-08-19
CN111593051A (en) 2020-08-28
IL264241B (en) 2020-04-30
IL284593A (en) 2021-08-31
US9145558B2 (en) 2015-09-29
EP2991661B1 (en) 2019-03-13
CN105378085B (en) 2019-02-15
AU2017200365A1 (en) 2017-02-23
CR20190269A (en) 2019-09-13
EP2992009B1 (en) 2020-06-24
IL273184B (en) 2021-07-29
ZA201507216B (en) 2017-08-30
US20150126719A1 (en) 2015-05-07
PH12018501963A1 (en) 2020-07-20
JP6216444B2 (en) 2017-10-18
KR20210151260A (en) 2021-12-13
AU2014259757A1 (en) 2015-10-22
KR102558571B1 (en) 2023-07-21
RU2019124314A (en) 2019-08-21
JP7429103B2 (en) 2024-02-07
WO2014179620A1 (en) 2014-11-06
JP2021107408A (en) 2021-07-29
HUE043697T2 (en) 2019-09-30
HK1221486A1 (en) 2017-06-02
US9127276B2 (en) 2015-09-08
BR112015027377B1 (en) 2023-01-10
PL2992098T3 (en) 2019-09-30
DOP2021000095A (en) 2021-09-15
CN112921036A (en) 2021-06-08
RU2650510C2 (en) 2018-04-16
US20230151365A1 (en) 2023-05-18
US20210130823A1 (en) 2021-05-06
EP2992097A2 (en) 2016-03-09
EP3633039A1 (en) 2020-04-08
UA121017C2 (en) 2020-03-25
KR20160002977A (en) 2016-01-08
US20160090596A1 (en) 2016-03-31
RU2015151204A (en) 2017-06-02
CN105377887A (en) 2016-03-02
WO2014179625A1 (en) 2014-11-06
IL274064A (en) 2020-06-30
MX2019010441A (en) 2019-10-17
AU2014259759B2 (en) 2020-06-18
RU2018136140A3 (en) 2022-04-27
RU2670614C9 (en) 2018-11-23
RU2015151202A (en) 2017-06-06
US20210087566A1 (en) 2021-03-25
RU2015151199A (en) 2017-06-05
AU2020217347A1 (en) 2020-08-27
EA201891479A1 (en) 2018-11-30
AU2014259759A1 (en) 2015-10-22
US20180273952A1 (en) 2018-09-27
AU2020233603A1 (en) 2020-10-01
RU2670614C2 (en) 2018-10-24
IL284593B (en) 2022-10-01
US11299736B1 (en) 2022-04-12
JP7339294B2 (en) 2023-09-05
US10883104B2 (en) 2021-01-05
WO2014179627A2 (en) 2014-11-06
EP3690049A1 (en) 2020-08-05
CA2921514C (en) 2023-10-24
RU2686080C2 (en) 2019-04-24
HRP20190987T1 (en) 2019-09-20
DK2991656T3 (en) 2020-03-23
IL264580B (en) 2020-04-30
KR20200090966A (en) 2020-07-29
JP2018027091A (en) 2018-02-22
UA120287C2 (en) 2019-11-11
AU2019200820B2 (en) 2020-04-30
JP2019056001A (en) 2019-04-11
US20210395734A1 (en) 2021-12-23
NZ728517A (en) 2021-12-24
PE20152002A1 (en) 2016-01-21
PH12015502493A1 (en) 2016-02-22
AU2021204244B2 (en) 2023-10-19
WO2014179627A3 (en) 2015-04-16
US9181550B2 (en) 2015-11-10
IL242132B (en) 2018-10-31
MX2021008901A (en) 2021-08-19
BR112015027377A2 (en) 2017-08-29
KR101857707B1 (en) 2018-05-14
IL264580A (en) 2019-02-28
AU2017203436B2 (en) 2018-10-18
NZ631552A (en) 2017-02-24
AU2020207820A1 (en) 2020-08-06
JP2016522683A (en) 2016-08-04
IL272617A (en) 2020-03-31
CN110042098A (en) 2019-07-23
AU2019203674B2 (en) 2021-03-25
MX2015015263A (en) 2016-12-16
SG10201906382QA (en) 2019-08-27
AU2019202598A1 (en) 2019-05-02
CN105378082B (en) 2020-06-09
HUE050394T2 (en) 2020-11-30
BR112015027369A8 (en) 2018-01-02
NZ753018A (en) 2022-01-28
WO2014179626A3 (en) 2015-02-26
KR20160002976A (en) 2016-01-08
DOP2015000268A (en) 2015-11-30
US10683499B2 (en) 2020-06-16
KR20160002975A (en) 2016-01-08
EP2991656A2 (en) 2016-03-09
CN105378085A (en) 2016-03-02
US20190367914A1 (en) 2019-12-05
CN110066795A (en) 2019-07-30
WO2014179626A2 (en) 2014-11-06
ZA201507218B (en) 2023-09-27
EP2992098A4 (en) 2017-01-11
JP6639629B2 (en) 2020-02-05
JP2020058370A (en) 2020-04-16
KR102138781B1 (en) 2020-07-28
US20220275365A9 (en) 2022-09-01
IL264241A (en) 2019-02-28
HRP20201378T1 (en) 2020-11-27
JP2016526018A (en) 2016-09-01
MX2020004209A (en) 2020-08-13
JP2021020901A (en) 2021-02-18
KR20160002974A (en) 2016-01-08
JP6592486B2 (en) 2019-10-16
EP2992098B1 (en) 2019-03-27
BR112015027321A2 (en) 2017-09-26
ES2885174T3 (en) 2021-12-13
JP7177127B2 (en) 2022-11-22
US11851655B2 (en) 2023-12-26
US20180273953A1 (en) 2018-09-27
EA036584B1 (en) 2020-11-26
CY1121879T1 (en) 2020-10-14
WO2014179629A2 (en) 2014-11-06
IL273205A (en) 2020-04-30
JP2023012548A (en) 2023-01-25
HK1221403A1 (en) 2017-06-02
IL242125B (en) 2019-02-28
JP6995478B2 (en) 2022-01-14
SI2992009T1 (en) 2020-10-30
LT2992009T (en) 2020-11-10
AU2019200820A1 (en) 2019-02-28
ES2730015T3 (en) 2019-11-07
JP6387084B2 (en) 2018-09-05
RS58981B1 (en) 2019-08-30
NZ631512A (en) 2016-10-28
CN105378082A (en) 2016-03-02
IL263843A (en) 2019-01-31
KR20190084138A (en) 2019-07-15
DK2992009T3 (en) 2020-09-14
JP2021074021A (en) 2021-05-20
BR112015027369B1 (en) 2021-06-08
KR20160003723A (en) 2016-01-11
US9163239B2 (en) 2015-10-20
AU2017200365C1 (en) 2019-04-18
IL284000A (en) 2021-07-29
CN114058617A (en) 2022-02-18
IL242124B (en) 2019-02-28
US10844379B2 (en) 2020-11-24
PT3524680T (en) 2021-01-04
PH12015502493B1 (en) 2016-02-22
EA201592093A1 (en) 2016-06-30
US20180044676A1 (en) 2018-02-15
US20160017323A1 (en) 2016-01-21
JP2023113843A (en) 2023-08-16
JP6652602B2 (en) 2020-02-26
AU2014259757B2 (en) 2017-03-02
KR102315836B1 (en) 2021-10-22
EP3524680A1 (en) 2019-08-14
US20210024923A1 (en) 2021-01-28
JP2016522817A (en) 2016-08-04
CA2921167A1 (en) 2014-11-06
IL261901B (en) 2020-05-31
CL2015003217A1 (en) 2016-07-08
CN108064162B (en) 2021-12-03
US20180002693A1 (en) 2018-01-04
HK1221485A1 (en) 2017-06-02
EP3828275A1 (en) 2021-06-02
SG11201508800WA (en) 2015-11-27
NZ712737A (en) 2021-08-27
PT2992009T (en) 2020-09-21
SG10201801507RA (en) 2018-03-28
AU2017200950A1 (en) 2017-03-02
AU2018267625A1 (en) 2018-12-13
EP2992098A2 (en) 2016-03-09
IL273184A (en) 2020-04-30
BR112015027322A2 (en) 2017-09-26
LT2992098T (en) 2019-07-10

Similar Documents

Publication Publication Date Title
US10927372B2 (en) Compositions and methods for modulating apolipoprotein C-III expression
CA2946003A1 (en) Compositions and methods for modulating angiopoietin-like 3 expression
CA2943705A1 (en) Compositions and methods for modulating pkk expression
CA2942570A1 (en) Compositions and methods for modulating growth hormone receptor expression

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20200424

EEER Examination request

Effective date: 20200424

EEER Examination request

Effective date: 20200424

FZDE Discontinued

Effective date: 20231102