CA2921167A1 - Compositions and methods for modulating hbv and ttr expression - Google Patents

Compositions and methods for modulating hbv and ttr expression Download PDF

Info

Publication number
CA2921167A1
CA2921167A1 CA2921167A CA2921167A CA2921167A1 CA 2921167 A1 CA2921167 A1 CA 2921167A1 CA 2921167 A CA2921167 A CA 2921167A CA 2921167 A CA2921167 A CA 2921167A CA 2921167 A1 CA2921167 A1 CA 2921167A1
Authority
CA
Canada
Prior art keywords
compound
conjugate
modified oligonucleotide
certain embodiments
following structure
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.)
Pending
Application number
CA2921167A
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=CA2921167(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 CA2921167A1 publication Critical patent/CA2921167A1/en
Pending 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. 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 of transthyretin (TTR).

Description

COMPOSITIONS AND METHODS FOR MODULATING HBV AND TTR EXPRESSION
SEQUENCE LISTING
The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled BIOL0248WOSEQ_ST25.txt, created on May 1, 2014, which is 16 Kb in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.
BACKGROUND OF THE INVENTION
The principle behind antisense technology is that an antisense compound hybridizes to a target nucleic acid and modulates the amount, activity, and/or function of the target nucleic acid. For example in certain instances, antisense compounds result in altered transcription or translation of a target. Such modulation of expression can be achieved by, for example, target mRNA
degradation or occupancy-based inhibition. An example of modulation of RNA target function by degradation is RNase H-based degradation of the target RNA upon hybridization with a DNA-like antisense compound.
Another example of modulation of gene expression by target degradation is RNA interference (RNAi). RNAi refers to antisense-mediated gene silencing through a mechanism that utilizes the RNA-induced siliencing complex (RISC). An additional example of modulation of RNA target function is by an occupancy-based mechanism such as is employed naturally by microRNA. MicroRNAs are small non-coding RNAs that regulate the expression of protein-coding RNAs. The binding of an antisense compound to a microRNA prevents that microRNA from binding to its messenger RNA targets, and thus interferes with the function of the microRNA. MicroRNA mimics can enhance native microRNA function. Certain antisense compounds alter splicing of pre-mRNA.
Regardless of the specific mechanism, sequence-specificity makes antisense compounds attractive as tools for target validation and gene functionalization, as well as therapeutics to selectively modulate the expression of genes involved in the pathogenesis of diseases.
Antisense technology is an effective means for modulating the expression of one or more specific gene products and can therefore prove to be uniquely useful in a number of therapeutic, diagnostic, and research applications. Chemically modified nucleosides may be incorporated into antisense compounds to enhance one or more properties, such as nuclease resistance, pharmacokinetics or affinity for a target nucleic acid. In 1998, the antisense compound, Vitravene0 (fomivirsen; developed by Isis Pharmaceuticals Inc., Carlsbad, CA) was the first antisense drug to achieve marketing clearance from the U.S. Food and Drug Administration (FDA), and is currently a treatment of cytomegalovirus (CMV)-induced retinitis in AIDS
patients.

New chemical modifications have improved the potency and efficacy of antisense compounds, uncovering the potential for oral delivery as well as enhancing subcutaneous administration, decreasing potential for side effects, and leading to improvements in patient convenience. Chemical modifications increasing potency of antisense compounds allow administration of lower doses, which reduces the potential for toxicity, as well as decreasing overall cost of therapy. Modifications increasing the resistance to degradation result in slower clearance from the body, allowing for less frequent dosing. Different types of chemical modifications can be combined in one compound to further optimize the compound's efficacy.
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
2 modulation. This balance of properties is more important in the setting of single-stranded antisense compounds than in siRNA compounds, where the conjugate may simply be attached to the sense strand.
Disclosed herein are conjugated single-stranded antisense compounds having improved potency in liver cells in vivo compared with the same antisense compound lacking the conjugate. Given the required balance of properties for these compounds such improved potency is surprising.
In certain embodiments, conjugate groups herein comprise a cleavable moiety.
As noted, without wishing to be bound by mechanism, it is logical that the conjugate should remain on the compound long enough to provide enhancement in uptake, but after that, it is desirable for some portion or, ideally, all of the conjugate to be cleaved, releasing the parent compound (e.g., antisense compound) in its most active form. In certain embodiments, the cleavable moiety is a cleavable nucleoside. Such embodiments take advantage of endogenous nucleases in the cell by attaching the rest of the conjugate (the cluster) to the antisense oligonucleotide through a nucleoside via one or more cleavable bonds, such as those of a phosphodiester linkage. In certain embodiments, the cluster is bound to the cleavable nucleoside through a phosphodiester linkage. In certain embodiments, the cleavable nucleoside is attached to the antisense oligonucleotide (antisense compound) by a phosphodiester linkage. In certain embodiments, the conjugate group may 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
3 oligonucleotide using standard automated techniques and introduce the conjugate with the final (5'-most) nucleoside or after the oligonucleotide has been cleaved from the solid support.
In view of the art and the present disclosure, one of ordinary skill can easily make any of the conjugates and conjugated oligonucleotides herein. Moreover, synthesis of certain such conjugates and conjugated oligonucleotides disclosed herein is easier and/or requires few steps, and is therefore less expensive than that of conjugates previously disclosed, providing advantages in manufacturing. For example, the synthesis of certain conjugate groups consists of fewer synthetic steps, resulting in increased yield, relative to conjugate groups previously described. Conjugate groups such as GalNAc3-10 in Example 46 and GalNAc3-7 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
4 uptake/potency a small loss in that substantial gain may be acceptable to achieve improved tolerability.
Accordingly, in certain embodiments, conjugated antisense compounds comprise at least one phosphodiester linkage.
In certain embodiments, conjugation of antisense compounds herein results in increased delivery, uptake and activity in hepatocytes. Thus, more compound is delivered to liver tissue. However, in certain embodiments, that increased delivery alone does not explain the entire increase in activity. In certain such embodiments, more compound enters hepatocytes. In certain embodiments, even that increased hepatocyte uptake does not explain the entire increase in activity. In such embodiments, productive uptake of the conjugated compound is increased. For example, as shown in Example 102, certain embodiments of GalNAc-containing conjugates increase enrichment of antisense oligonucleotides in hepatocytes versus non-parenchymal cells. This enrichment is beneficial for oligonucleotides that target genes that are expressed in hepatocytes.
In certain embodiments, conjugated antisense compounds herein result in reduced kidney exposure.
For example, as shown in Example 20, the concentrations of antisense oligonucleotides comprising certain embodiments of GalNAc-containing conjugates are lower in the kidney than that of antisense 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:
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
E¨F
A¨B¨C¨D
E¨F
E¨F
where q = 5, the formula is:

E¨F
/ E¨F
_________________________________ E¨F
A¨B¨C D
N E¨F
E¨F
In certain embodiments, conjugated antisense compounds are provided having the structure:
Targeting moiety ASO
HO OH
¨ 0=P-OH NH, O htli-J,õN
HO---'.
- NHAc ' I 1 _.7........\___O 0 H H 0' N N H=0 NHA TO
H 0 __ OH

c 0 0 0 Linker - Ligand Tether HO HN---- \ Cleavable moiety OH

_..\..Ø....\70 N
Branching group HO
NHAc 0 In certain embodiments, conjugated antisense compounds are provided having the structure:
Cell targeting moiety HO OH
0 , 0 HO--4,k-)..,..-_____N
P.
Cleavable moiety AcHN 0 1 0--_ ¨ ¨
OH

HO H _ 1 ____ 1 0-..õ - 0 .....r0,....\ze-N II II
HO \õ.-.N -P. --.(y --..,_,,,--0-1,3¨ r N----/
0 1 0 _ 0- ss _ AcHN OH - 0' 0 Tether. _ -041=0 Ligand _ HO H y P, ASO
HO
1"

OH
NHAc Branching group In certain embodiments, conjugated antisense compounds are provided having the structure:
ASO
Cleavable moiety HO¨P=0 0 (N¨rµ/N
HO¨P=0 Cell targeting moiety HO OH

HO-4.\u oOH
AcHN 0-(0 3 HOOH _ _______ 0 0 Conjugate linker AcHN - 0 OH
Tether HO
Ligand y AO
H


HO
NHAc Branching group In certain embodiments, conjugated antisense compounds are provided having the structure:

ASO
Ligand 0 Tether Cleavable moiety HO¨=O
HOOH
Hyio AcHN 0 _ -NH
HO OH
01) HO 0N1r(^)2(y 3 AcHN 0 0 Conjugate HOOH linker AcHN 0 Branching group Cell targeting moiety 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 incorporated by reference in their entirety for any purpose.
A. Definitions Unless specific definitions are provided, the nomenclature used in connection with, and the procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques may be used for chemical synthesis, and chemical analysis. Certain such techniques and procedures may be found for example in "Carbohydrate Modifications in Antisense Research" Edited by Sangvi and Cook, American Chemical Society , Washington D.C., 1994;
"Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, Pa., 21st edition, 2005; and "Antisense Drug Technology, Principles, Strategies, and Applications" Edited by Stanley T. Crooke, CRC Press, Boca Raton, Florida; and Sambrook et al., "Molecular Cloning, A laboratory Manual," 2nd Edition, Cold Spring Harbor Laboratory Press, 1989, which are hereby incorporated by reference for any purpose. Where permitted, all patents, applications, published applications and other publications and other data referred to throughout in the disclosure are incorporated by reference herein in their entirety.
Unless otherwise indicated, the following terms have the following meanings:
As used herein, "nucleoside" means a compound comprising a nucleobase moiety and a sugar moiety. Nucleosides include, but are not limited to, naturally occurring nucleosides (as found in DNA and RNA) and modified nucleosides. Nucleosides may be linked to a phosphate moiety.
As used herein, "chemical modification" means a chemical difference in a compound when compared to a naturally occurring counterpart. Chemical modifications of oligonucleotides include nucleoside modifications (including sugar moiety modifications and nucleobase modifications) and internucleoside linkage modifications. In reference to an oligonucleotide, chemical modification does not include differences only in nucleobase sequence.
As used herein, "furanosyl" means a structure comprising a 5-membered ring comprising four carbon atoms and one oxygen atom.
As used herein, "naturally occurring sugar moiety" means a ribofuranosyl as found in naturally occurring RNA or a deoxyribofuranosyl as found in naturally occurring DNA.
As used herein, "sugar moiety" means a naturally occurring sugar moiety or a modified sugar moiety of a nucleoside.
As used herein, "modified sugar moiety" means a substituted sugar moiety or a sugar surrogate.
As used herein, "substituted sugar moiety" means a furanosyl that is not a naturally occurring sugar moiety. Substituted sugar moieties include, but are not limited to furanosyls comprising substituents at the 2'-position, the 3'-position, the 5'-position and/or the 4'-position. Certain substituted sugar moieties are bicyclic sugar moieties.
As used herein, "2'-substituted sugar moiety" means a furanosyl comprising a substituent at the 2'-position other than H or OH. Unless otherwise indicated, a 2'-substituted sugar moiety is not a bicyclic sugar moiety (i.e., the 2'-substituent of a 2'-substituted sugar moiety does not form a bridge to another atom of the furanosyl ring.
As used herein, "MOE" means -OCH2CH2OCH3.
As used herein, "2'-F nucleoside" refers to a nucleoside comprising a sugar comprising fluorine at the 2' position. Unless otherwise indicated, the fluorine in a 2'-F nucleoside is in the ribo position (replacing the OH of a natural ribose).
As used herein the term "sugar surrogate" means a structure that does not comprise a furanosyl and that is capable of replacing the naturally occurring sugar moiety of a nucleoside, such that the resulting nucleoside sub-units are capable of linking together and/or linking to other nucleosides to form an oligomeric compound which is capable of hybridizing to a complementary oligomeric compound. Such structures include rings comprising a different number of atoms than furanosyl (e.g., 4, 6, or 7-membered rings);
replacement of the oxygen of a furanosyl with a non-oxygen atom (e.g., carbon, sulfur, or nitrogen); or both a change in the number of atoms and a replacement of the oxygen. Such structures may also comprise substitutions corresponding to those described for substituted sugar moieties (e.g., 6-membered carbocyclic bicyclic sugar surrogates optionally comprising additional substituents).
Sugar surrogates also include more complex sugar replacements (e.g., the non-ring systems of peptide nucleic acid). Sugar surrogates include without limitation morpholinos, cyclohexenyls and cyclohexitols.
As used herein, "bicyclic sugar moiety" means a modified sugar moiety comprising a 4 to 7 membered ring (including but not limited to a furanosyl) comprising a bridge connecting two atoms of the 4 to 7 membered ring to form a second ring, resulting in a bicyclic structure.
In certain embodiments, the 4 to 7 membered ring is a sugar ring. In certain embodiments the 4 to 7 membered ring is a furanosyl. In certain such embodiments, the bridge connects the 2'-carbon and the 4'-carbon of the furanosyl.
As used herein, "nucleotide" means a nucleoside further comprising a phosphate linking group. As used herein, "linked nucleosides" may or may not be linked by phosphate linkages and thus includes, but is not limited to "linked nucleotides." As used herein, "linked nucleosides" are nucleosides that are connected in a continuous sequence (i.e. no additional nucleosides are present between those that are linked).
As used herein, "nucleobase" means a group of atoms that can be linked to a sugar moiety to create a nucleoside that is capable of incorporation into an oligonucleotide, and wherein the group of atoms is capable of bonding with a complementary naturally occurring nucleobase of another oligonucleotide or nucleic acid.
Nucleobases may be naturally occurring or may be modified.
As used herein the terms, "unmodified nucleobase" or "naturally occurring nucleobase" means the naturally occurring heterocyclic nucleobases of RNA or DNA: the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) (including 5-methyl C), and uracil (U).
As used herein, "modified nucleobase" means any nucleobase that is not a naturally occurring nucleobase.

As used herein, "modified nucleoside" means a nucleoside comprising at least one chemical modification compared to naturally occurring RNA or DNA nucleosides. Modified nucleosides comprise a modified sugar moiety and/or a modified nucleobase.
As used herein, "bicyclic nucleoside" or "BNA" means a nucleoside comprising a bicyclic sugar moiety.
As used herein, "constrained ethyl nucleoside" or "cEt" means a nucleoside comprising a bicyclic sugar moiety comprising a 4' -CH(CH3)-0-2'bridge.
As used herein, "locked nucleic acid nucleoside" or "LNA" means a nucleoside comprising a bicyclic sugar moiety comprising a 4' -CH2-0-2'bridge.
As used herein, "2'-substituted nucleoside" means a nucleoside comprising a substituent at the 2'-position other than H or OH. Unless otherwise indicated, a 2'-substituted nucleoside is not a bicyclic nucleoside.
As used herein, "deoxynucleoside" means a nucleoside comprising 2'-H furanosyl sugar moiety, as found in naturally occurring deoxyribonucleosides (DNA). In certain embodiments, a 2'-deoxynucleoside may comprise a modified nucleobase or may comprise an RNA nucleobase (e.g., uracil).
As used herein, "oligonucleotide" means a compound comprising a plurality of linked nucleosides.
In certain embodiments, an oligonucleotide comprises one or more unmodified ribonucleosides (RNA) and/or unmodified deoxyribonucleosides (DNA) and/or one or more modified nucleosides.
As used herein "oligonucleoside" means an oligonucleotide in which none of the internucleoside linkages contains a phosphorus atom. As used herein, oligonucleotides include oligonucleosides.
As used herein, "modified oligonucleotide" means an oligonucleotide comprising at least one modified nucleoside and/or at least one modified internucleoside linkage.
As used herein, "linkage" or "linking group" means a group of atoms that link together two or more other groups of atoms.
As used herein "internucleoside linkage" means a covalent linkage between adjacent nucleosides in an oligonucleotide.
As used herein "naturally occurring internucleoside linkage" means a 3' to 5' phosphodiester linkage.
As used herein, "modified internucleoside linkage" means any internucleoside linkage other than a naturally occurring internucleoside linkage.
As used herein, "terminal internucleoside linkage" means the linkage between the last two nucleosides of an oligonucleotide or defined region thereof.
As used herein, "phosphorus linking group" means a linking group comprising a phosphorus atom.
Phosphorus linking groups include without limitation groups having the formula:

JVW
Rb=P¨R, Rd JVVV
wherein:
Ra and Rd are each, independently, 0, S, CH2, NH, or NJI 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 J1 is Rb is 0 or S.
Phosphorus linking groups include without limitation, phosphodiester, phosphorothioate, phosphorodithioate, phosphonate, phosphoramidate, phosphorothioamidate, thionoalkylphosphonate, phosphotriesters, thionoalkylphosphotriester and boranophosphate.
As used herein, "internucleoside phosphorus linking group" means a phosphorus linking group that directly links two nucleosides.
As used herein, "non-internucleoside phosphorus linking group" means a phosphorus linking group that does not directly link two nucleosides. In certain embodiments, a non-internucleoside phosphorus linking group links a nucleoside to a group other than a nucleoside. In certain embodiments, a non-internucleoside phosphorus linking group links two groups, neither of which is a nucleoside.
As used herein, "neutral linking group" means a linking group that is not charged. Neutral linking groups include without limitation phosphotriesters, methylphosphonates, MMI (-CH2-N(CH3)-0-), amide-3 (-CH2-C(=0)-N(H)-), amide-4 (-CH2-N(H)-C(=0)-), formacetal (-0-CH2-0-), and thioformacetal (-S-CH2-0-).
Further neutral linking groups include nonionic linkages comprising siloxane (dialkylsiloxane), carboxylate ester, carboxamide, sulfide, sulfonate ester and amides (See for example:
Carbohydrate Modifications in Antisense Research; Y.S. Sanghvi and P.D. Cook Eds. ACS Symposium Series 580;
Chapters 3 and 4, (pp.
40-65)). Further neutral linking groups include nonionic linkages comprising mixed N, 0, S and CH2 component parts.
As used herein, "internucleoside neutral linking group" means a neutral linking group that directly links two nucleosides.
As used herein, "non-internucleoside neutral linking group" means a neutral linking group that does not directly link two nucleosides. In certain embodiments, a non-internucleoside neutral linking group links a nucleoside to a group other than a nucleoside. In certain embodiments, a non-internucleoside neutral linking group links two groups, neither of which is a nucleoside.
As used herein, "oligomeric compound" means a polymeric structure comprising two or more sub-structures. In certain embodiments, an oligomeric compound comprises an oligonucleotide. In certain embodiments, an oligomeric compound comprises one or more conjugate groups and/or terminal groups. In certain embodiments, an oligomeric compound consists of an oligonucleotide.
Oligomeric compounds also include naturally occurring nucleic acids. In certain embodiments, an oligomeric compound comprises a backbone of one or more linked monomeric subunits where each linked monomeric subunit is directly or indirectly attached to a heterocyclic base moiety. In certain embodiments, oligomeric compounds may also include monomeric subunits that are not linked to a heterocyclic base moiety, thereby providing abasic sites.
In certain embodiments, the linkages joining the monomeric subunits, the sugar moieties or surrogates and the heterocyclic base moieties can be independently modified. In certain embodiments, the linkage-sugar unit, which may or may not include a heterocyclic base, may be substituted with a mimetic such as the monomers in peptide nucleic acids.
As used herein, "terminal group" means one or more atom attached to either, or both, the 3' end or the 5' end of an oligonucleotide. In certain embodiments a terminal group is a conjugate group. In certain embodiments, a terminal group comprises one or more terminal group nucleosides.
As used herein, "conjugate" or "conjugate group" means an atom or group of atoms bound to an oligonucleotide or oligomeric compound. In general, conjugate groups modify one or more properties of the compound to which they are attached, including, but not limited to pharmacodynamic, pharmacokinetic, binding, absorption, cellular distribution, cellular uptake, charge and/or clearance properties.
As used herein, "conjugate linker" or "linker" in the context of a conjugate group means a portion of a conjugate group comprising any atom or group of atoms and which covalently link (1) an oligonucleotide to another portion of the conjugate group or (2) two or more portions of the conjugate group.
Conjugate groups are shown herein as radicals, providing a bond for forming covalent attachment to an oligomeric compound such as an antisense oligonucleotide. In certain embodiments, the point of attachment on the oligomeric compound is the 3'-oxygen atom of the 3'-hydroxyl group of the 3' terminal nucleoside of the oligomeric compound. In certain embodiments the point of attachment on the oligomeric compound is the 5'-oxygen atom of the 5'-hydroxyl group of the 5' terminal nucleoside of the oligomeric compound. In certain embodiments, the bond for forming attachment to the oligomeric compound is a cleavable bond. In certain such embodiments, such cleavable bond constitutes all or part of a cleavable moiety.
In certain embodiments, conjugate groups comprise a cleavable moiety (e.g., a cleavable bond or cleavable nucleoside) and a carbohydrate cluster portion, such as a GalNAc cluster portion. Such carbohydrate cluster portion comprises: a targeting moiety and, optionally, a conjugate linker. In certain embodiments, the carbohydrate cluster portion is identified by the number and identity of the ligand. For example, in certain embodiments, the carbohydrate cluster portion comprises 3 GalNAc groups and is designated "GalNAc3". In certain embodiments, the carbohydrate cluster portion comprises 4 GalNAc groups and is designated "GalNAc4". 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 "GalNac3-1a" 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,"1 Med. Chem. 2004, (47): 5798-5808, for examples of carbohydrate conjugate clusters).
As used herein, "carbohydrate derivative" means any compound which may be synthesized using a carbohydrate as a starting material or intermediate.
As used herein, "carbohydrate" means a naturally occurring carbohydrate, a modified carbohydrate, or a carbohydrate derivative.
As used herein "protecting group" means any compound or protecting group known to those having skill in the art. Non-limiting examples of protecting groups may be found in "Protective Groups in Organic Chemistry", T. W. Greene, P. G. M. Wuts, ISBN 0-471-62301-6, John Wiley &
Sons, Inc, New York, which is incorporated herein by reference in its entirety.
As used herein, "single-stranded" means an oligomeric compound that is not hybridized to its complement and which lacks sufficient self-complementarity to form a stable self-duplex.
As used herein, "double stranded" means a pair of oligomeric compounds that are hybridized to one another or a single self-complementary oligomeric compound that forms a hairpin structure. In certain embodiments, a double-stranded oligomeric compound comprises a first and a second oligomeric compound.
As used herein, "antisense compound" means a compound comprising or consisting of an oligonucleotide at least a portion of which is complementary to a target nucleic acid to which it is capable of hybridizing, resulting in at least one antisense activity.

As used herein, "antisense activity" means any detectable and/or measurable change attributable to the hybridization of an antisense compound to its target nucleic acid. In certain embodiments, antisense activity includes modulation of the amount or activity of a target nucleic acid transcript (e.g. mRNA). In certain embodiments, antisense activity includes modulation of the splicing of pre-mRNA.
As used herein, "RNase H based antisense compound" means an antisense compound wherein at least some of the antisense activity of the antisense compound is attributable to hybridization of the antisense compound to a target nucleic acid and subsequent cleavage of the target nucleic acid by RNase H.
As used herein, "RISC based antisense compound" means an antisense compound wherein at least some of the antisense activity of the antisense compound is attributable to the RNA Induced Silencing Complex (RISC).
As used herein, "detecting" or "measuring" means that a test or assay for detecting or measuring is performed. Such detection and/or measuring may result in a value of zero.
Thus, if a test for detection or measuring results in a finding of no activity (activity of zero), the step of detecting or measuring the activity has nevertheless been performed.
As used herein, "detectable and/or measureable activity" means a statistically significant activity that is not zero.
As used herein, "essentially unchanged" means little or no change in a particular parameter, particularly relative to another parameter which changes much more. In certain embodiments, a parameter is essentially unchanged when it changes less than 5%. In certain embodiments, a parameter is essentially unchanged if it changes less than two-fold while another parameter changes at least ten-fold. For example, in certain embodiments, an antisense activity is a change in the amount of a target nucleic acid. In certain such embodiments, the amount of a non-target nucleic acid is essentially unchanged if it changes much less than the target nucleic acid does, but the change need not be zero.
As used herein, "expression" means the process by which a gene ultimately results in a protein.
Expression includes, but is not limited to, transcription, post-transcriptional modification (e.g., splicing, polyadenlyation, addition of 5 '-cap), and translation.
As used herein, "target nucleic acid" means a nucleic acid molecule to which an antisense compound is intended to hybridize to result in a desired antisense activity. Antisense oligonucleotides have sufficient complementarity to their target nucleic acids to allow hybridization under physiological conditions.
As used herein, "nucleobase complementarity" or "complementarity" when in reference to nucleobases means a nucleobase that is capable of base pairing with another nucleobase. For example, in DNA, adenine (A) is complementary to thymine (T). For example, in RNA, adenine (A) is complementary to uracil (U). In certain embodiments, complementary nucleobase means a nucleobase of an antisense compound that is capable of base pairing with a nucleobase of its target nucleic acid. For example, if a nucleobase at a certain position of an antisense compound is capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid, then the position of hydrogen bonding between the oligonucleotide and the target nucleic acid is considered to be complementary at that nucleobase pair.
Nucleobases comprising certain modifications may maintain the ability to pair with a counterpart nucleobase and thus, are still capable of nucleobase complementarity.
As used herein, "non-complementary" in reference to nucleobases means a pair of nucleobases that do not form hydrogen bonds with one another.
As used herein, "complementary" in reference to oligomeric compounds (e.g., linked nucleosides, oligonucleotides, or nucleic acids) means the capacity of such oligomeric compounds or regions thereof to hybridize to another oligomeric compound or region thereof through nucleobase complementarity.
Complementary oligomeric compounds need not have nucleobase complementarity at each nucleoside.
Rather, some mismatches are tolerated. In certain embodiments, complementary oligomeric compounds or regions are complementary at 70% of the nucleobases (70% complementary). In certain embodiments, complementary oligomeric compounds or regions are 80% complementary. In certain embodiments, complementary oligomeric compounds or regions are 90% complementary. In certain embodiments, complementary oligomeric compounds or regions are 95% complementary. In certain embodiments, complementary oligomeric compounds or regions are 100% complementary.
As used herein, "mismatch" means a nucleobase of a first oligomeric compound that is not capable of pairing with a nucleobase at a corresponding position of a second oligomeric compound, when the first and second oligomeric compound are aligned. Either or both of the first and second oligomeric compounds may be oligonucleotides.
As used herein, "hybridization" means the pairing of complementary oligomeric compounds (e.g., an antisense compound and its target nucleic acid). While not limited to a particular mechanism, the most common mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleobases.
As used herein, "specifically hybridizes" means the ability of an oligomeric compound to hybridize to one nucleic acid site with greater affinity than it hybridizes to another nucleic acid site.
As used herein, "fully complementary" in reference to an oligonucleotide or portion thereof means that each nucleobase of the oligonucleotide or portion thereof is capable of pairing with a nucleobase of a complementary nucleic acid or contiguous portion thereof. Thus, a fully complementary region comprises no mismatches or unhybridized nucleobases in either strand.
As used herein, "percent complementarity" means the percentage of nucleobases of an oligomeric compound that are complementary to an equal-length portion of a target nucleic acid. Percent complementarity is calculated by dividing the number of nucleobases of the oligomeric compound that are complementary to nucleobases at corresponding positions in the target nucleic acid by the total length of the oligomeric compound.

As used herein, "percent identity" means the number of nucleobases in a first nucleic acid that are the same type (independent of chemical modification) as nucleobases at corresponding positions in a second nucleic acid, divided by the total number of nucleobases in the first nucleic acid.
As used herein, "modulation" means a change of amount or quality of a molecule, function, or activity when compared to the amount or quality of a molecule, function, or activity prior to modulation. For example, modulation includes the change, either an increase (stimulation or induction) or a decrease (inhibition or reduction) in gene expression. As a further example, modulation of expression can include a change in splice site selection of pre-mRNA processing, resulting in a change in the absolute or relative amount of a particular splice-variant compared to the amount in the absence of modulation.
As used herein, "chemical motif" means a pattern of chemical modifications in an oligonucleotide or a region thereof. Motifs may be defined by modifications at certain nucleosides and/or at certain linking groups of an oligonucleotide.
As used herein, "nucleoside motif" means a pattern of nucleoside modifications in an oligonucleotide or a region thereof. The linkages of such an oligonucleotide may be modified or unmodified. Unless otherwise indicated, motifs herein describing only nucleosides are intended to be nucleoside motifs. Thus, in such instances, the linkages are not limited.
As used herein, "sugar motif" means a pattern of sugar modifications in an oligonucleotide or a region thereof.
As used herein, "linkage motif" means a pattern of linkage modifications in an oligonucleotide or region thereof. The nucleosides of such an oligonucleotide may be modified or unmodified. Unless otherwise indicated, motifs herein describing only linkages are intended to be linkage motifs. Thus, in such instances, the nucleosides are not limited.
As used herein, "nucleobase modification motif" means a pattern of modifications to nucleobases along an oligonucleotide. Unless otherwise indicated, a nucleobase modification motif is independent of the nucleobase sequence.
As used herein, "sequence motif" means a pattern of nucleobases arranged along an oligonucleotide or portion thereof. Unless otherwise indicated, a sequence motif is independent of chemical modifications and thus may have any combination of chemical modifications, including no chemical modifications.
As used herein, "type of modification" in reference to a nucleoside or a nucleoside of a "type" means the chemical modification of a nucleoside and includes modified and unmodified nucleosides. Accordingly, unless otherwise indicated, a "nucleoside having a modification of a first type" may be an unmodified nucleoside.
As used herein, "differently modified" mean chemical modifications or chemical substituents that are different from one another, including absence of modifications. Thus, for example, a MOE nucleoside and an unmodified DNA nucleoside are "differently modified," even though the DNA
nucleoside is unmodified.
Likewise, DNA and RNA are "differently modified," even though both are naturally-occurring unmodified nucleosides. Nucleosides that are the same but for comprising different nucleobases are not differently modified. For example, a nucleoside comprising a 2'-0Me modified sugar and an unmodified adenine nucleobase and a nucleoside comprising a 2'-0Me modified sugar and an unmodified thymine nucleobase are not differently modified.
As used herein, "the same type of modifications" refers to modifications that are the same as one another, including absence of modifications. Thus, for example, two unmodified DNA nucleosides have "the same type of modification," even though the DNA nucleoside is unmodified.
Such nucleosides having the same type modification may comprise different nucleobases.
As used herein, "separate regions" means portions of an oligonucleotide wherein the chemical modifications or the motif of chemical modifications of any neighboring portions include at least one difference to allow the separate regions to be distinguished from one another.
As used herein, "pharmaceutically acceptable carrier or diluent" means any substance suitable for use in administering to an animal. In certain embodiments, a pharmaceutically acceptable carrier or diluent is sterile saline. In certain embodiments, such sterile saline is pharmaceutical grade saline.
As used herein the term "metabolic disorder" means a disease or condition principally characterized by dysregulation of metabolism ¨ the complex set of chemical reactions associated with breakdown of food to produce energy.
As used herein, the term "cardiovascular disorder" means a disease or condition principally characterized by impaired function of the heart or blood vessels.
As used herein the term "mono or polycyclic ring system" is meant to include all ring systems selected from single or polycyclic radical ring systems wherein the rings are fused or linked and is meant to be inclusive of single and mixed ring systems individually selected from aliphatic, alicyclic, aryl, heteroaryl, aralkyl, arylalkyl, heterocyclic, heteroaryl, heteroaromatic and heteroarylalkyl. Such mono and poly cyclic structures can contain rings that each have the same level of saturation or each, independently, have varying degrees of saturation including fully saturated, partially saturated or fully unsaturated. Each ring can comprise ring atoms selected from C, N, 0 and S to give rise to heterocyclic rings as well as rings comprising only C ring atoms which can be present in a mixed motif such as for example benzimidazole wherein one ring has only carbon ring atoms and the fused ring has two nitrogen atoms. The mono or polycyclic ring system can be further substituted with substituent groups such as for example phthalimide which has two =0 groups attached to one of the rings. Mono or polycyclic ring systems can be attached to parent molecules using various strategies such as directly through a ring atom, fused through multiple ring atoms, through a substituent group or through a bifunctional linking moiety.
As used herein, "prodrug" means an inactive or less active form of a compound which, when administered to a subject, is metabolized to form the active, or more active, compound (e.g., drug).
As used herein, "substituent" and "substituent group," means an atom or group that replaces the atom or group of a named parent compound. For example a substituent of a modified nucleoside is any atom or group that differs from the atom or group found in a naturally occurring nucleoside (e.g., a modified 2'-substuent is any atom or group at the 2'-position of a nucleoside other than H
or OH). Substituent groups can be protected or unprotected. In certain embodiments, compounds of the present disclosure have substituents at one or at more than one position of the parent compound. Substituents may also be further substituted with other substituent groups and may be attached directly or via a linking group such as an alkyl or hydrocarbyl group to a parent compound.
Likewise, as used herein, "substituent" in reference to a chemical functional group means an atom or group of atoms that differs from the atom or a group of atoms normally present in the named functional group. In certain embodiments, a substituent replaces a hydrogen atom of the functional group (e.g., in certain embodiments, the substituent of a substituted methyl group is an atom or group other than hydrogen which replaces one of the hydrogen atoms of an unsubstituted methyl group).
Unless otherwise indicated, groups amenable for use as substituents include without limitation, halogen, hydroxyl, alkyl, alkenyl, alkynyl, acyl (-C(0)Raa), carboxyl (-C(0)0-Raa), aliphatic groups, alicyclic groups, alkoxy, substituted oxy (-O-R.), aryl, aralkyl, heterocyclic radical, heteroaryl, heteroarylalkyl, amino (-N(Rbb)(Ree)), imino(=NRbb), amido (-C(0)N(Rbb)(Ree) or -N(Rbb)C(0)Ra.), azido (-N3), nitro (-NO2), cyano (-CN), carbamido (-0C(0)N(Rbb)(Ree) or -N(Rbb)C(0)0Raa), ureido (-N(Rbb)C(0)N(Rbb)(Ree)), thioureido (-N(Rbb)C(S)N(Rbb)-(Rõ)), guanidinyl (-N(Rbb)C(=NRON(Rbb)(Ree)), amidinyl (-C(=NRON(Rbb)(Ree) or -N(Rbb)C(=NRbb)(R.)), 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 (C1-C12 alkyl) with from 1 to about 6 carbon atoms being more preferred.
As used herein, "alkenyl," means a straight or branched hydrocarbon chain radical containing up to twenty four carbon atoms and having at least one carbon-carbon double bond.
Examples of alkenyl groups include without limitation, ethenyl, propenyl, butenyl, 1-methy1-2-buten-1-yl, dienes such as 1,3-butadiene and the like. Alkenyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred. Alkenyl groups as used herein may optionally include one or more further substituent groups.
As used herein, "alkynyl," means a straight or branched hydrocarbon radical containing up to twenty four carbon atoms and having at least one carbon-carbon triple bond. Examples of alkynyl groups include, without limitation, ethynyl, 1-propynyl, 1-butynyl, and the like. Alkynyl groups typically include from 2 to about 24 carbon atoms, more typically from 2 to about 12 carbon atoms with from 2 to about 6 carbon atoms being more preferred. Alkynyl groups as used herein may optionally include one or more further substituent groups.
As used herein, "acyl," means a radical formed by removal of a hydroxyl group from an organic acid and has the general Formula -C(0)-X where X is typically aliphatic, alicyclic or aromatic. Examples include aliphatic carbonyls, aromatic carbonyls, aliphatic sulfonyls, aromatic sulfinyls, aliphatic sulfinyls, aromatic phosphates, aliphatic phosphates and the like. Acyl groups as used herein may optionally include further substituent groups.
As used herein, "alicyclic" means a cyclic ring system wherein the ring is aliphatic. The ring system can comprise one or more rings wherein at least one ring is aliphatic.
Preferred alicyclics include rings having from about 5 to about 9 carbon atoms in the ring. Alicyclic as used herein may optionally include further substituent groups.
As used herein, "aliphatic" means a straight or branched hydrocarbon radical containing up to twenty four carbon atoms wherein the saturation between any two carbon atoms is a single, double or triple bond.
An aliphatic group preferably contains from 1 to about 24 carbon atoms, more typically from 1 to about 12 carbon atoms with from 1 to about 6 carbon atoms being more preferred. The straight or branched chain of an aliphatic group may be interrupted with one or more heteroatoms that include nitrogen, oxygen, sulfur and phosphorus. Such aliphatic groups interrupted by heteroatoms include without limitation, polyalkoxys, such as polyalkylene glycols, polyamines, and polyimines. Aliphatic groups as used herein may optionally include further substituent groups.
As used herein, "alkoxy" means a radical formed between an alkyl group and an oxygen atom wherein the oxygen atom is used to attach the alkoxy group to a parent molecule. Examples of alkoxy groups include without limitation, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, neopentoxy, n-hexoxy and the like. Alkoxy groups as used herein may optionally include further substituent groups.
As used herein, "aminoalkyl" means an amino substituted C1-C12 alkyl radical.
The alkyl portion of the radical forms a covalent bond with a parent molecule. The amino group can be located at any position and the aminoalkyl group can be substituted with a further substituent group at the alkyl and/or amino portions.
As used herein, "aralkyl" and "arylalkyl" mean an aromatic group that is covalently linked to a C1-C12 alkyl radical. The alkyl radical portion of the resulting aralkyl (or arylalkyl) group forms a covalent bond with a parent molecule. Examples include without limitation, benzyl, phenethyl and the like. Aralkyl groups as used herein may optionally include further substituent groups attached to the alkyl, the aryl or both groups that form the radical group.
As used herein, "aryl" and "aromatic" mean a mono- or polycyclic carbocyclic ring system radicals having one or more aromatic rings. Examples of aryl groups include without limitation, phenyl, naphthyl, tetrahydronaphthyl, indanyl, idenyl and the like. Preferred aryl ring systems have from about 5 to about 20 carbon atoms in one or more rings. Aryl groups as used herein may optionally include further substituent groups.
As used herein, "halo" and "halogen," mean an atom selected from fluorine, chlorine, bromine and iodine.
As used herein, "heteroaryl," and "heteroaromatic," mean a radical comprising a mono- or poly-cyclic aromatic ring, ring system or fused ring system wherein at least one of the rings is aromatic and includes one or more heteroatoms. Heteroaryl is also meant to include fused ring systems including systems where one or more of the fused rings contain no heteroatoms. Heteroaryl groups typically include one ring atom selected from sulfur, nitrogen or oxygen. Examples of heteroaryl groups include without limitation, pyridinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, thiophenyl, furanyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzooxazolyl, quinoxalinyl and the like. Heteroaryl radicals can be attached to a parent molecule directly or through a linking moiety such as an aliphatic group or hetero atom. Heteroaryl groups as used herein may optionally include further substituent groups.
As used herein, "conjugate compound" means any atoms, group of atoms, or group of linked atoms suitable for use as a conjugate group. In certain embodiments, conjugate compounds may possess or impart one or more properties, including, but not limited to pharmacodynamic, pharmacokinetic, binding, absorption, cellular distribution, cellular uptake, charge and/or clearance properties.
As used herein, unless otherwise indicated or modified, the term "double-stranded" refers to two separate oligomeric compounds that are hybridized to one another. Such double stranded compounds may have one or more or non-hybridizing nucleosides at one or both ends of one or both strands (overhangs) and/or one or more internal non-hybridizing nucleosides (mismatches) provided there is sufficient complementarity to maintain hybridization under physiologically relevant conditions.
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 Su2ar Moieties In certain embodiments, compounds of the disclosure comprise one or more modifed nucleosides comprising a modifed sugar moiety. Such compounds comprising one or more sugar-modified nucleosides may have desirable properties, such as enhanced nuclease stability or increased binding affinity with a target nucleic acid relative to an oligonucleotide comprising only nucleosides comprising naturally occurring sugar moieties. In certain embodiments, modified sugar moieties are substitued sugar moieties. In certain embodiments, modified sugar moieties are sugar surrogates. Such sugar surrogates may comprise one or more substitutions corresponding to those of substituted sugar moieties.
In certain embodiments, modified sugar moieties are substituted sugar moieties comprising one or more non-bridging sugar substituent, including but not limited to substituents at the 2' and/or 5' positions.
Examples of sugar substituents suitable for the 2'-position, include, but are not limited to: 2'-F, 2'-OCH3 ("OMe" or "0-methyl"), and 2'-0(CH2)20CH3 ("MOE"). In certain embodiments, sugar substituents at the 2' position is selected from allyl, amino, azido, thio, 0-allyl, 0-C1-C10 alkyl, 0-C1-C10 substituted alkyl; OCF3, 0(CH2)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 CI-CH, alkyl. Examples of sugar substituents at the 5'-position, include, but are not limited to:, 5'-methyl (R or S); 5'-vinyl, and
5'-methoxy. In certain embodiments, substituted sugars comprise more than one non-bridging sugar substituent, for example, 2'-F-5'-methyl sugar moieties (see,e.g., PCT International Application WO
2008/101157, for additional 5', 2'-bis substituted sugar moieties and nucleosides).
Nucleosides comprising 2'-substituted sugar moieties are referred to as 2'-substituted nucleosides. In certain embodiments, a 2'- substituted nucleoside comprises a 2'-substituent group selected from halo, allyl, amino, azido, SH, CN, OCN, CF3, OCF3, 0, S, or N(Rm)-alkyl; 0, S, or N(Rm)-alkenyl; 0, S or N(Rm)-alkynyl; 0-alkyleny1-0-alkyl, alkynyl, alkaryl, aralkyl, 0-alkaryl, 0-aralkyl, 0(CH2)25CH3, 0-(CH2)2-0-N(Rm)(Rn) or 0-CH2-C(=0)-N(Rm)(Rõ), where each Rm and Rii is, independently, H, an amino protecting group or substituted or unsubstituted Ci-Clo 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)25CH3, 0-(CH2)2-0-N(Rm)(Rn), 0(CH2)20(CH2)2N(CH3)2, and N-substituted acetamide (0-CH2-C(=0)-N(Rm)(Rn) where each Rm and Rn is, independently, H, an amino protecting group or substituted or unsubstituted C1-Clo 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(RO(Rb)]n-, -[C(Ra)(Rb)]n-0-, -C(RaRb)-N(R)-0- or, -C(RaRb)-0-N(R)-; 4'-CH2-2', 4'-(CH2)2-2', 4'-(CH2)-0-2' (LNA); 4'-(CH2)-S-2'; 4'-(CH2)2-0-2' (ENA);
4'-CH(CH3)-0-2' (cEt) and 4'-CH(CH2OCH3)-0-2',and analogs thereof (see, e.g., U.S. Patent 7,399,845, issued on July 15, 2008); 4'-C(CH3)(CH3)-0-2'and analogs thereof, (see, e.g., W02009/006478, published January 8, 2009); 4'-CH2-N(OCH3)-2' and analogs thereof (see, e.g., W02008/150729, published December 11, 2008); 4'-CH2-0-N(CH3)-2' (see, e.g., U52004/0171570, published September 2, 2004); 4'-CH2-0-N(R)-2', and 4'-CH2-N(R)-0-2'-, wherein each R is, independently, H, a protecting group, or C1-C12 alkyl; 4'-CH2-N(R)-0-2', wherein R
is H, C1-C12 alkyl, or a protecting group (see, U.S. Patent 7,427,672, issued on September 23, 2008); 4'-CH2-C(H)(CH3)-2' (see, e.g., Chattopadhyaya, et al., J Org. Chem.,2009, 74, 118-134); and 4'-CH2-C(=CH2)-2' and analogs thereof (see, published PCT International Application WO
2008/154401, published on December 8, 2008).
In certain embodiments, such 4' to 2' bridges independently comprise from 1 to 4 linked groups independently selected from -[C(RO(Rb)]n-, -C(RO=C(Rb)-, -C(Ra)=N-, -C(=NRO-, -C(=0)-, -C(=S)-, -0-, -Si(Ra)2-, -S(=0)x-, and -N(RO-;
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-C7alicyclic radical, halogen, OJI, NJ1J2, SJI, N3, COOJI, acyl (C(=0)-H), substituted acyl, CN, sulfonyl (S(=0)2-J1), or sulfoxyl (S(=0)-Ji); and each J1 and J2 is, independently, H, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C5-C20 aryl, substituted C5-C20 aryl, acyl (C(=0)-H), substituted acyl, a heterocycle radical, a substituted heterocycle radical, C1-C12 aminoalkyl, substituted C1-C12 aminoalkyl, or a protecting group.

Nucleosides comprising bicyclic sugar moieties are referred to as bicyclic nucleosides or BNAs.
Bicyclic nucleosides include, but are not limited to, (A) a-L-Methyleneoxy (4'-CH2-0-2') BNA , (B) 13-D-Methyleneoxy (4'-CH2-0-2') BNA (also referred to as locked nucleic acid or LNA) , (C) Ethyleneoxy (4'-(CH2)2-0-2') BNA, (D) Aminooxy (4'-CH2-0-N(R)-2') BNA, (E) Oxyamino (4'-CH2-N(R)-0-2') BNA, (F) Methyl(methyleneoxy) (4'-CH(CH3)-0-2') BNA (also referred to as constrained ethyl or cEt), (G) methylene-thio (4'-CH2-S-2') BNA, (H) methylene-amino (4'-CH2-N(R)-2') BNA, (I) methyl carbocyclic (4'-CH2-CH(CH3)-2') BNA, and (J) propylene carbocyclic (4'-(CH2)3-2') BNA as depicted below.

nx -.--.

--t-- -0 _0 (A) (B) (C) _________ ()yBx 1 / OyBx H
7 ____()yBx 0¨ ¨N R-N- ¨0 3C 1,1/4:,,/, (D) R (E) (F) ____________________________________________ (07/Bx 07/Bx ...,zBx --S
(G) \R
(1) ____________________________ 0/Bx (J) wherein Bx is a nucleobase moiety and R is, independently, H, a protecting group, or CI-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.,1 Am. Chem. Soc., 129(26) 8362-8379 (Jul. 4, 2007);
Elayadi et al., Curr. Opinion Invens. Drugs, 2001, 2, 558-561; Braasch et al., Chem. Biol., 2001, 8, 1-7;
Orum et al., Curr. Opinion 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. U52004/0171570, U52007/0287831, and U52008/0039618; U.S. Patent Serial Nos. 12/129,154, 60/989,574, 61/026,995, 61/026,998, 61/056,564,
7 PCT/US2014/036463 61/086,231, 61/097,787, and 61/099,844; and PCT International Applications Nos. PCT/US2008/064591, PCT/US2008/066154, and PCT/US2008/068922.
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 13-D
configuration. Previously, a-L-methyleneoxy (4'-CH2-0-2') bicyclic nucleosides have been incorporated into antisense oligonucleotides that showed antisense activity (Frieden et al., Nucleic Acids Research, 2003, 21, 6365-6372).
In certain embodiments, substituted sugar moieties comprise one or more non-bridging sugar substituent and one or more bridging sugar substituent (e.g., 5'-substituted and 4'-2' bridged sugars). (see, PCT International Application WO 2007/134181, published on 11/22/07, wherein LNA is substituted with, for example, a 5'-methyl or a 5'-vinyl group).
In certain embodiments, modified sugar moieties are sugar surrogates. In certain such embodiments, the oxygen atom of the naturally occuring sugar is substituted, e.g., with a sulfer, carbon or nitrogen atom. In certain such embodiments, such modified sugar moiety also comprises bridging and/or non-bridging substituents as described above. For example, certain sugar surrogates comprise a 4'-sulfer atom and a substitution at the 2'-position (see,e.g., published U.S. Patent Application US2005/0130923, published on June 16, 2005) and/or the 5' position. By way of additional example, carbocyclic bicyclic nucleosides having a 4'-2' bridge have been described (see, e.g., Freier et al., Nucleic Acids Research, 1997, 25(22), 4429-4443 and Albaek et al., 1 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., Biochemistry, 2002, 41, 4503-4510; and U.S. Patents 5,698,685;
5,166,315; 5,185,444; and 5,034,506). As used here, the term "morpholino" means a sugar surrogate having the following structure:
1¨ 0¨

In certain embodiments, morpholinos may be modified, for example by adding or altering various substituent groups from the above morpholino structure. Such sugar surrogates are refered to herein as "modifed morpholinos."
For another example, in certain embodiments, a sugar surrogate comprises a six-membered tetrahydropyran. Such tetrahydropyrans may be further modified or substituted.
Nucleosides comprising such modified tetrahydropyrans include, but are not limited to, hexitol nucleic acid (HNA), anitol nucleic acid (ANA), manitol nucleic acid (MNA) (see Leumann, CJ. Bioorg. & Med. Chem.
(2002) 10:841-854), fluoro HNA (F-HNA), and those compounds having Formula VI:
c11 q2 CI6 Bx % R1 R2 CI5 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, q4, q5, q6 and q7 are each, independently, H, C1-C6 alkyl, substituted Ci-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl, or substituted C2-C6 alkynyl; and each of R1 and R2 is independently selected from among: hydrogen, halogen, substituted or unsubstituted alkoxy, NJ1J2, SJI, N3, OC(=X)Ji, OC(=X)NJ1J2, NJ3C(=X)NJ1J2, and CN, wherein X is 0, S or NJI, and each J1, J2, and J3 is, independently, H or C1-C6 alkyl.
In certain embodiments, the modified THP nucleosides of Formula VI are provided wherein qi, q2, q3, q4, q5, q6and q7 are each H. In certain embodiments, at least one of q1, q2, q3, q4, q5, q6and q7 is other than H. In certain embodiments, at least one of ql, q2, q3, q4, q5, q6and q7 is methyl. In certain embodiments, THP
nucleosides of Formula VI are provided wherein one of R1 and R2 is F. In certain embodiments, R1 is fluoro and R2 is H, R1 is methoxy and R2 is H, and R1 is methoxyethoxy and R2 is H.
Many other bicyclo and tricyclo sugar surrogate ring systems are also known in the art that can be used to modify nucleosides for incorporation into antisense compounds (see, e.g., review article: Leumann, J.
C, Bioorganic & Medicinal Chemistry, 2002, 10, 841-854).
Combinations of modifications are also provided without limitation, such as 2'-F-5'-methyl substituted nucleosides (see PCT International Application WO 2008/101157 Published on 8/21/08 for other disclosed 5', 2'-bis substituted nucleosides) and replacement of the ribosyl ring oxygen atom with S and further substitution at the 2'-position (see published U.S. Patent Application US2005-0130923, published on June 16, 2005) or alternatively 5'-substitution of a bicyclic nucleic acid (see PCT International Application WO 2007/134181, published on 11/22/07 wherein a 4'-CH2-0-2' bicyclic nucleoside is further substituted at the 5' position with a 5'-methyl or a 5'-vinyl group). The synthesis and preparation of carbocyclic bicyclic nucleosides along with their oligomerization and biochemical studies have also been described (see, e.g., Srivastava et al.,1 Am. Chem. Soc. 2007, 129(26), 8362-8379).
In certain embodiments, the present disclosure provides oligonucleotides comprising modified nucleosides.
Those modified nucleotides may include modified sugars, modified nucleobases, and/or modified linkages.
The specific modifications are selected such that the resulting oligonucleotides possess desireable characteristics. In certain embodmiments, oligonucleotides comprise one or more RNA-like nucleosides. In certain embodiments, oligonucleotides comprise one or more DNA-like nucleotides.
2. Certain Nucleobase Modifications In certain embodiments, nucleosides of the present disclosure comprise one or more unmodified nucleobases. In certain embodiments, nucleosides of the present disclosure comprise one or more modifed nucleobases.
In certain embodiments, modified nucleobases are selected from: universal bases, hydrophobic bases, promiscuous bases, size-expanded bases, and fluorinated bases as defined herein. 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil; 5-propynylcytosine; 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl CH3) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 2-F-adenine, 2-amino-adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine, 3-deazaguanine and 3-deazaadenine, universal bases, hydrophobic bases, promiscuous bases, size-expanded bases, and fluorinated bases as defined herein. Further modified nucleobases include tricyclic pyrimidines such as phenoxazine cytidine( [5,4-b][1,4]benzoxazin-2(3H)-one), phenothiazine cytidine (1H-pyrimido[5,4-b][1,4]benzothiazin-2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g. 9-(2-aminoethoxy)-H-pyrimido[5,4-b][1,4]benzoxazin-2(3H)-one), carbazole cytidine (2H-pyrimido[4,5-b]indo1-2-one), pyridoindole cytidine (H-pyrido[3',2':4,5]pyrrolo[2,3-d]pyrimidin-2-one). Modified nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Further nucleobases include those disclosed in United States Patent No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, Kroschwitz, J.I., Ed., John Wiley &
Sons, 1990, 858-859; those disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613; and those disclosed by Sanghvi, Y.S., Chapter 15, Antisense Research and Applications, Crooke, S.T.
and Lebleu, B., Eds., CRC Press, 1993, 273-288.
Representative United States patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include without limitation, U.S. 3,687,808; 4,845,205;

5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255;
5,484,908; 5,502,177;
5,525,711; 5,552,540; 5,587,469; 5,594,121; 5,596,091; 5,614,617; 5,645,985;
5,681,941; 5,750,692;
5,763,588; 5,830,653 and 6,005,096, certain of which are commonly owned with the instant application, and each of which is herein incorporated by reference in its entirety.
3. Certain Internucleoside Linkages In certain embodiments, the present disclosure provides oligonucleotides comprising linked nucleosides. In such embodiments, nucleosides may be linked together using any internucleoside linkage.
The two main classes of internucleoside linking groups are defined by the presence or absence of a phosphorus atom. Representative phosphorus containing internucleoside linkages include, but are not limited to, phosphodiesters (PO), phosphotriesters, methylphosphonates, phosphoramidate, and phosphorothioates (PS). Representative non-phosphorus containing internucleoside linking groups include, but are not limited to, methylenemethylimino (-CH2-N(CH3)-0-CH2-), thiodiester (-0-C(0)-S-), thionocarbamate (-0-C(0)(NH)-S-); siloxane (-0-Si(H)2-0-); and N,N'-dimethylhydrazine (-CH2-N(CH3)-N(CH3)-). Modified linkages, compared to natural phosphodiester linkages, can be used to alter, typically increase, nuclease resistance of the oligonucleotide. In certain embodiments, internucleoside linkages having a chiral atom can be prepared as a racemic mixture, or as separate enantiomers. Representative chiral linkages include, but are not limited to, alkylphosphonates and phosphorothioates. Methods of preparation of phosphorous-containing and non-phosphorous-containing internucleoside linkages are well known to those skilled in the art.
The oligonucleotides described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), a or [3 such as for sugar anomers, or as (D) or (L) such as for amino acids etc.
Included in the antisense compounds provided herein are all such possible isomers, as well as their racemic and optically pure forms.
Neutral internucleoside linkages include without limitation, phosphotriesters, methylphosphonates, MMI (3'-CH2-N(CH3)-0-5'), amide-3 (3'-CH2-C(=0)-N(H)-5'), amide-4 (3'-CH2-N(H)-C(=0)-5'), formacetal (3'-0-CH2-0-5'), and thioformacetal (3'-S-CH2-0-5'). Further neutral internucleoside linkages include nonionic linkages comprising siloxane (dialkylsiloxane), carboxylate ester, carboxamide, sulfide, sulfonate ester and amides (See for example: Carbohydrate Modifications in Antisense Research; Y. S. Sanghvi and P.D. Cook, Eds., ACS Symposium Series 580; Chapters 3 and 4, 40-65). Further neutral internucleoside linkages include nonionic linkages comprising mixed N, 0, S and CH2 component parts.
4. Certain Motifs In certain embodiments, antisense oligonucleotides comprise one or more modified nucleoside (e.g., nucleoside comprising a modified sugar and/or modified nucleobase) and/or one or more modified internucleoside linkage. The pattern of such modifications on an oligonucleotide is referred to herein as a motif. In certain embodiments, sugar, nucleobase, and linkage motifs are independent of one another.
a. Certain sugar motifs In certain embodiments, oligonucleotides comprise one or more type of modified sugar moieties and/or naturally occurring sugar moieties arranged along an oligonucleotide or region thereof in a defined pattern or sugar modification motif. Such motifs may include any of the sugar modifications discussed herein and/or other known sugar modifications.
In certain embodiments, the oligonucleotides comprise or consist of a region having a gapmer sugar motif, which comprises two external regions or "wings" and a central or internal region or "gap." The three regions of a gapmer sugar motif (the 5'-wing, the gap, and the 3'-wing) form a contiguous sequence of nucleosides wherein at least some of the sugar moieties of the nucleosides of each of the wings differ from at least some of the sugar moieties of the nucleosides of the gap. Specifically, at least the sugar moieties of the nucleosides of each wing that are closest to the gap (the 3'-most nucleoside of the 5'-wing and the 5'-most nucleoside of the 3'-wing) differ from the sugar moiety of the neighboring gap nucleosides, thus defining the boundary between the wings and the gap. In certain embodiments, the sugar moieties within the gap are the same as one another. In certain embodiments, the gap includes one or more nucleoside having a sugar moiety that differs from the sugar moiety of one or more other nucleosides of the gap. In certain embodiments, the sugar motifs of the two wings are the same as one another (symmetric sugar gapmer). In certain embodiments, the sugar motifs of the 5'-wing differs from the sugar motif of the 3'-wing (asymmetric sugar gapmer).
i. Certain 5'-wings In certain embodiments, the 5'- wing of a gapmer consists of 1 to 8 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 to 7 linked nucleosides.
In certain embodiments, the 5'-wing of a gapmer consists of 1 to 6 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 to 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 3 to 5 linked nucleosides.
In certain embodiments, the 5'- wing of a gapmer consists of 4 or 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 to 4 linked nucleosides.
In certain embodiments, the 5'-wing of a gapmer consists of 1 to 3 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 or 2 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 2 to 4 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 2 or 3 linked nucleosides.
In certain embodiments, the 5'- wing of a gapmer consists of 3 or 4 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 1 nucleoside. In certain embodiments, the 5'- wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 3 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 4 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 5 linked nucleosides. In certain embodiments, the 5'- wing of a gapmer consists of 6 linked nucleosides.
In certain embodiments, the 5'- wing of a gapmer comprises at least one bicyclic nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least two bicyclic nucleosides. In certain embodiments, the 5'- wing of a gapmer comprises at least three bicyclic nucleosides. In certain embodiments, the 5'- wing of a gapmer comprises at least four bicyclic nucleosides. In certain embodiments, the 5'- wing of a gapmer comprises at least one constrained ethyl nucleoside.
In certain embodiments, the 5'-wing of a gapmer comprises at least one LNA nucleoside. In certain embodiments, each nucleoside of the 5'-wing of a gapmer is a bicyclic nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a constrained ethyl nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a LNA nucleoside.
In certain embodiments, the 5'- wing of a gapmer comprises at least one non-bicyclic modified nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-substituted nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-MOE nucleoside. In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-0Me nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a non-bicyclic modified nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-substituted nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-MOE
nucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-0Me nucleoside.
In certain embodiments, the 5'- wing of a gapmer comprises at least one 2'-deoxynucleoside. In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a 2'-deoxynucleoside. In a certain embodiments, the 5'- wing of a gapmer comprises at least one ribonucleoside.
In certain embodiments, each nucleoside of the 5'- wing of a gapmer is a ribonucleoside. In certain embodiments, one, more than one, or each of the nucleosides of the 5'- wing is an RNA-like nucleoside.
In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-MOE
nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-0Me nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-deoxynucleoside.
In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-MOE nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-0Me nucleoside. In certain embodiments, the 5'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-deoxynucleoside.
ii. Certain 3'-wings In certain embodiments, the 3'- wing of a gapmer consists of 1 to 8 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 to 7 linked nucleosides.
In certain embodiments, the 3'-wing of a gapmer consists of 1 to 6 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 to 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 2 to 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 3 to 5 linked nucleosides.
In certain embodiments, the 3'- wing of a gapmer consists of 4 or 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 to 4 linked nucleosides.
In certain embodiments, the 3'-wing of a gapmer consists of 1 to 3 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 or 2 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 2 to 4 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 2 or 3 linked nucleosides.
In certain embodiments, the 3'- wing of a gapmer consists of 3 or 4 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 1 nucleoside. In certain embodiments, the 3'- wing of a gapmer consists of 2 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 31inked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 4 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 5 linked nucleosides. In certain embodiments, the 3'- wing of a gapmer consists of 6 linked nucleosides.
In certain embodiments, the 3'- wing of a gapmer comprises at least one bicyclic nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one constrained ethyl nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one LNA nucleoside.
In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a bicyclic nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a constrained ethyl nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a LNA nucleoside.
In certain embodiments, the 3'- wing of a gapmer comprises at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least two non-bicyclic modified nucleosides. In certain embodiments, the 3'- wing of a gapmer comprises at least three non-bicyclic modified nucleosides. In certain embodiments, the 3'- wing of a gapmer comprises at least four non-bicyclic modified nucleosides. In certain embodiments, the 3'- wing of a gapmer comprises at least one 2'-substituted nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one 2'-MOE nucleoside. In certain embodiments, the 3'- wing of a gapmer comprises at least one 2'-0Me nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a non-bicyclic modified nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-substituted nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-MOE
nucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-0Me nucleoside.
In certain embodiments, the 3'- wing of a gapmer comprises at least one 2'-deoxynucleoside. In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a 2'-deoxynucleoside. In a certain embodiments, the 3'- wing of a gapmer comprises at least one ribonucleoside.
In certain embodiments, each nucleoside of the 3'- wing of a gapmer is a ribonucleoside. In certain embodiments, one, more than one, or each of the nucleosides of the 5'- wing is an RNA-like nucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-MOE
nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-0Me nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-MOE nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-0Me nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA
nucleoside and at least one non-bicyclic modified nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one 2'-substituted nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one 2'-MOE
nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside and at least one 2'-0Me nucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA
nucleoside and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one non-bicyclic modified nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one non-bicyclic modified nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA nucleoside, at least one non-bicyclic modified nucleoside, and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one 2'-substituted nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one 2'-substituted nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA
nucleoside, at least one 2'-substituted nucleoside, and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one 2'-MOE nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one 2'-MOE nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA
nucleoside, at least one 2'-MOE nucleoside, and at least one 2'-deoxynucleoside.
In certain embodiments, the 3'-wing of a gapmer comprises at least one bicyclic nucleoside, at least one 2'-0Me nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one constrained ethyl nucleoside, at least one 2'-0Me nucleoside, and at least one 2'-deoxynucleoside. In certain embodiments, the 3'-wing of a gapmer comprises at least one LNA
nucleoside, at least one 2'-0Me nucleoside, and at least one 2'-deoxynucleoside.
iii. Certain Central Regions (gaps) In certain embodiments, the gap of a gapmer consists of 6 to 20 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 15 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 12 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 to 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 or 7 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 to 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 or 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 8 to 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 8 or 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 6 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 7 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 8 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 9 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 10 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 11 linked nucleosides. In certain embodiments, the gap of a gapmer consists of 12 linked nucleosides.
In certain embodiments, each nucleoside of the gap of a gapmer is a 2'-deoxynucleoside. In certain embodiments, the gap comprises one or more modified nucleosides. In certain embodiments, each nucleoside of the gap of a gapmer is a 2'-deoxynucleoside or is a modified nucleoside that is "DNA-like." In such embodiments, "DNA-like" means that the nucleoside has similar characteristics to DNA, such that a duplex comprising the gapmer and an RNA molecule is capable of activating RNase H.
For example, under certain conditions, 2'-(ara)-F have been shown to support RNase H activation, and thus is DNA-like. In certain embodiments, one or more nucleosides of the gap of a gapmer is not a 2'-deoxynucleoside and is not DNA-like. In certain such embodiments, the gapmer nonetheless supports RNase H
activation (e.g., by virtue of the number or placement of the non-DNA nucleosides).
In certain embodiments, gaps comprise a stretch of unmodified 2'-deoxynucleoside interrupted by one or more modified nucleosides, thus resulting in three sub-regions (two stretches of one or more 2'-deoxynucleosides and a stretch of one or more interrupting modified nucleosides). In certain embodiments, no stretch of unmodified 2'-deoxynucleosides is longer than 5, 6, or 7 nucleosides. In certain embodiments, such short stretches is achieved by using short gap regions. In certain embodiments, short stretches are achieved by interrupting a longer gap region.
In certain embodiments, the gap comprises one or more modified nucleosides. In certain embodiments, the gap comprises one or more modified nucleosides selected from among cEt, FHNA, LNA, and 2-thio-thymidine. In certain embodiments, the gap comprises one modified nucleoside. In certain embodiments, the gap comprises a 5'-substituted sugar moiety selected from among 5'-Me, and 5'-(R)-Me.
In certain embodiments, the gap comprises two modified nucleosides. In certain embodiments, the gap comprises three modified nucleosides. In certain embodiments, the gap comprises four modified nucleosides.
In certain embodiments, the gap comprises two or more modified nucleosides and each modified nucleoside is the same. In certain embodiments, the gap comprises two or more modified nucleosides and each modified nucleoside is different.
In certain embodiments, the gap comprises one or more modified linkages. In certain embodiments, the gap comprises one or more methyl phosphonate linkages. In certain embodiments the gap comprises two or more modified linkages. In certain embodiments, the gap comprises one or more modified linkages and one or more modified nucleosides. In certain embodiments, the gap comprises one modified linkage and one modified nucleoside. In certain embodiments, the gap comprises two modified linkages and two or more modified nucleosides.
b. Certain Internucleoside Linkage Motifs In certain embodiments, oligonucleotides comprise modified internucleoside linkages arranged along the oligonucleotide or region thereof in a defined pattern or modified internucleoside linkage motif. In certain embodiments, oligonucleotides comprise a region having an alternating internucleoside linkage motif.
In certain embodiments, oligonucleotides of the present disclosure comprise a region of uniformly modified internucleoside linkages. In certain such embodiments, the oligonucleotide comprises a region that is uniformly linked by phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide is uniformly linked by phosphorothioate internucleoside linkages.
In certain embodiments, each internucleoside linkage of the oligonucleotide is selected from phosphodiester and phosphorothioate. In certain embodiments, each internucleoside linkage of the oligonucleotide is selected from phosphodiester and phosphorothioate and at least one internucleoside linkage is phosphorothioate.
In certain embodiments, the oligonucleotide comprises at least 6 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 7 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 8 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 9 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 10 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 11 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 12 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 13 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least 14 phosphorothioate internucleoside linkages.
In certain embodiments, the oligonucleotide comprises at least one block of at least 6 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 7 consecutive phosphorothioate internucleoside linkages.
In certain embodiments, the oligonucleotide comprises at least one block of at least 8 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 9 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least one block of at least 10 consecutive phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises at least block of at least one 12 consecutive phosphorothioate internucleoside linkages. In certain such embodiments, at least one such block is located at the 3' end of the oligonucleotide.
In certain such embodiments, at least one such block is located within 3 nucleosides of the 3' end of the oligonucleotide.In certain embodiments, the oligonucleotide comprises less than 15 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 14 phosphoro-thioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 13 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 12 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 11 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 10 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 9 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 8 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 7 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 6 phosphorothioate internucleoside linkages. In certain embodiments, the oligonucleotide comprises less than 5 phosphorothioate internucleoside linkages.
c. Certain Nucleobase Modification Motifs In certain embodiments, oligonucleotides comprise chemical modifications to nucleobases arranged along the oligonucleotide or region thereof in a defined pattern or nucleobases modification motif. In certain such embodiments, nucleobase modifications are arranged in a gapped motif. In certain embodiments, nucleobase modifications are arranged in an alternating motif. In certain embodiments, each nucleobase is modified. In certain embodiments, none of the nucleobases is chemically modified.
In certain embodiments, oligonucleotides comprise a block of modified nucleobases. In certain such embodiments, the block is at the 3'-end of the oligonucleotide. In certain embodiments the block is within 3 nucleotides of the 3'-end of the oligonucleotide. In certain such embodiments, the block is at the 5'-end of the oligonucleotide. In certain embodiments the block is within 3 nucleotides of the 5'-end of the oligonucleotide.
In certain embodiments, nucleobase modifications are a function of the natural base at a particular position of an oligonucleotide. For example, in certain embodiments each purine or each pyrimidine in an oligonucleotide is modified. In certain embodiments, each adenine is modified.
In certain embodiments, each guanine is modified. In certain embodiments, each thymine is modified. In certain embodiments, each cytosine is modified. In certain embodiments, each uracil is modified.
In certain embodiments, some, all, or none of the cytosine moieties in an oligonucleotide are 5-methyl cytosine moieties. Herein, 5-methyl cytosine is not a "modified nucleobase." Accordingly, unless otherwise indicated, unmodified nucleobases include both cytosine residues having a 5-methyl and those lacking a 5 methyl. In certain embodiments, the methylation state of all or some cytosine nucleobases is specified.
In certain embodiments, chemical modifications to nucleobases comprise attachment of certain conjugate groups to nucleobases. In certain embodiments, each purine or each pyrimidine in an oligonucleotide may be optionally modified to comprise a conjugate group.
d. Certain Overall Lengths In certain embodiments, the present disclosure provides oligonucleotides of any of a variety of ranges of lengths. In certain embodiments, oligonucleotides consist of X to Y linked nucleosides, where X
represents the fewest number of nucleosides in the range and Y represents the largest number of nucleosides in the range. In certain such embodiments, X and Y are each independently selected from 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, and 50; provided that X<Y. For example, in certain embodiments, the oligonucleotide may consist of 8 to 9, 8 to 10, 8 to 11, 8 to 12, 8 to 13, 8 to 14, 8 to 15, 8 to 16, 8 to 17, 8 to 18, 8 to 19, 8 to 20, 8 to 21, 8 to 22, 8 to 23, 8 to 24, 8 to 25, 8 to 26, 8 to 27, 8 to 28, 8 to 29, 8 to 30, 9 to 10, 9 to 11, 9 to 12, 9 to 13, 9 to 14, 9 to 15, 9 to 16, 9 to 17, 9 to 18, 9 to 19, 9 to 20, 9 to 21, 9 to 22, 9 to 23, 9 to 24, 9 to 25, 9 to 26, 9 to 27, 9 to 28, 9 to 29, 9 to 30, 10 to 11, 10 to 12, 10 to 13, 10 to 14, 10 to 15, 10 to 16, 10 to 17, 10 to 18, 10 to 19, 10 to 20, 10 to 21, 10 to 22, 10 to 23, 10 to 24, 10 to 25, 10 to 26, 10 to 27, to 28, 10 to 29, 10 to 30, 11 to 12, 11 to 13, 11 to 14, 11 to 15, 11 to 16, 11 to 17, 11 to 18, 11 to 19, 11 to 20, 11 to 21, 11 to 22, 11 to 23, 11 to 24, 11 to 25, 11 to 26, 11 to 27, 11 to 28, 11 to 29, 11 to 30, 12 to 13, 12 to 14, 12 to 15, 12 to 16, 12 to 17, 12 to 18, 12 to 19, 12 to 20, 12 to 21, 12 to 22, 12 to 23, 12 to 24, 12 to 25, 12 to 26, 12 to 27, 12 to 28, 12 to 29, 12 to 30, 13 to 14, 13 to 15, 13 to 16, 13 to 17, 13 to 18, 13 to 19, 13 to 20, 13 to 21, 13 to 22, 13 to 23, 13 to 24, 13 to 25, 13 to 26, 13 to 27, 13 to 28, 13 to 29, 13 to 30, 14 to 15, 14 to 16, 14 to 17, 14 to 18, 14 to 19, 14 to 20, 14 to 21, 14 to 22, 14 to 23, 14 to 24, 14 to 25, 14 to 26, 14 to 27, 14 to 28, 14 to 29, 14 to 30, 15 to 16, 15 to 17, 15 to 18, 15 to 19, 15 to 20, 15 to 21, 15 to 22, 15 to 23, 15 to 24, 15 to 25, 15 to 26, 15 to 27, 15 to 28, 15 to 29, 15 to 30, 16 to 17, 16 to 18, 16 to 19, 16 to 20, 16 to 21, 16 to 22, 16 to 23, 16 to 24, 16 to 25, 16 to 26, 16 to 27, 16 to 28, 16 to 29, 16 to 30, 17 to 18, 17 to 19, 17 to 20, 17 to 21, 17 to 22, 17 to 23, 17 to 24, 17 to 25, 17 to 26, 17 to 27, 17 to 28, 17 to 29, 17 to 30, 18 to 19, 18 to 20, 18 to 21, 18 to 22, 18 to 23, 18 to 24, 18 to 25, 18 to 26, 18 to 27, 18 to 28, 18 to 29, 18 to 30, 19 to 20, 19 to 21, 19 to 22, 19 to 23, 19 to 24, 19 to 25, 19 to 26, 19 to 29, 19 to 28, 19 to 29, 19 to 30, 20 to 21, 20 to 22, 20 to 23, 20 to 24, 20 to 25, 20 to 26, 20 to 27, 20 to 28, 20 to 29, 20 to 30, 21 to 22, 21 to 23, 21 to 24, 21 to 25, 21 to 26, 21 to 27, 21 to 28, 21 to 29, 21 to 30, 22 to 23, 22 to 24, 22 to 25, 22 to 26, 22 to 27, 22 to 28, 22 to 29, 22 to 30, 23 to 24, 23 to 25, 23 to 26, 23 to 27, 23 to 28, 23 to 29, 23 to 30, 24 to 25, 24 to 26, 24 to 27, 24 to 28, 24 to 29, 24 to 30, 25 to 26, 25 to 27, 25 to 28, 25 to 29, 25 to 30, 26 to 27, 26 to 28, 26 to 29, 26 to 30, 27 to 28, 27 to 29, 27 to 30, 28 to 29, 28 to 30, or 29 to 30 linked nucleosides. In embodiments where the number of nucleosides of an oligonucleotide of a compound is limited, whether to a range or to a specific number, the compound may, nonetheless further comprise additional other substituents.
For example, an oligonucleotide comprising 8-30 nucleosides excludes oligonucleotides having 31 nucleosides, but, unless otherwise indicated, such an oligonucleotide may further comprise, for example one or more conjugate groups, terminal groups, or other substituents.
Further, where an oligonucleotide is described by an overall length range and by regions having specified lengths, and where the sum of specified lengths of the regions is less than the upper limit of the overall length range, the oligonucleotide may have additional nucleosides, beyond those of the specified regions, provided that the total number of nucleosides does not exceed the upper limit of the overall length range.
5. Certain Antisense Oligonucleotide Chemistry Motifs In certain embodiments, the chemical structural features of antisense oligonucleotides are characterized by their sugar motif, internucleoside linkage motif, nucleobase modification motif and overall length. In certain embodiments, such parameters are each independent of one another. Thus, each internucleoside linkage of an oligonucleotide having a gapmer sugar motif may be modified or unmodified and may or may not follow the gapmer modification pattern of the sugar modifications. Thus, the internucleoside linkages within the wing regions of a sugar-gapmer may be the same or different from one another and may be the same or different from the internucleoside linkages of the gap region. Likewise, such sugar-gapmer oligonucleotides may comprise one or more modified nucleobase independent of the gapmer pattern of the sugar modifications. One of skill in the art will appreciate that such motifs may be combined to create a variety of oligonucleotides.
In certain embodiments, the selection of internucleoside linkage and nucleoside modification are not independent of one another.
i. Certain Sequences and Targets In certain embodiments, the invention provides antisense oligonucleotides having a sequence complementary to a target nucleic acid. Such antisense compounds are capable of hybridizing to a target nucleic acid, resulting in at least one antisense activity. In certain embodiments, antisense compounds specifically hybridize to one or more target nucleic acid. In certain embodiments, a specifically hybridizing antisense compound has a nucleobase sequence comprising a region having sufficient complementarity to a target nucleic acid to allow hybridization and result in antisense activity and insufficient complementarity to any non-target so as to avoid or reduce non-specific hybridization to non-target nucleic acid sequences under conditions in which specific hybridization is desired (e.g., under physiological conditions for in vivo or therapeutic uses, and under conditions in which assays are performed in the case of in vitro assays). In certain embodiments, oligonucleotides are selective between a target and non-target, even though both target and non-target comprise the target sequence. In such embodiments, selectivity may result from relative accessibility of the target region of one nucleic acid molecule compared to the other.
In certain embodiments, the present disclosure provides antisense compounds comprising oligonucleotides that are fully complementary to the target nucleic acid over the entire length of the oligonucleotide. In certain embodiments, oligonucleotides are 99%
complementary to the target nucleic acid.
In certain embodiments, oligonucleotides are 95% complementary to the target nucleic acid. In certain embodiments, such oligonucleotides are 90% complementary to the target nucleic acid.
In certain embodiments, such oligonucleotides are 85% complementary to the target nucleic acid. In certain embodiments, such oligonucleotides are 80% complementary to the target nucleic acid. In certain embodiments, an antisense compound comprises a region that is fully complementary to a target nucleic acid and is at least 80% complementary to the target nucleic acid over the entire length of the oligonucleotide. In certain such embodiments, the region of full complementarity is from 6 to 14 nucleobases in length.
In certain embodiments, oligonucleotides comprise a hybridizing region and a terminal region. In certain such embodiments, the hybridizing region consists of 12-30 linked nucleosides and is fully complementary to the target nucleic acid. In certain embodiments, the hybridizing region includes one mismatch relative to the target nucleic acid. In certain embodiments, the hybridizing region includes two mismatches relative to the target nucleic acid. In certain embodiments, the hybridizing region includes three mismatches relative to the target nucleic acid. In certain embodiments, the terminal region consists of 1-4 terminal nucleosides. In certain embodiments, the terminal nucleosides are at the 3' end. In certain embodiments, one or more of the terminal nucleosides are not complementary to the target nucleic acid.
Antisense mechanisms include any mechanism involving the hybridization of an oligonucleotide with target nucleic acid, wherein the hybridization results in a biological effect.
In certain embodiments, such hybridization results in either target nucleic acid degradation or occupancy with concomitant inhibition or stimulation of the cellular machinery involving, for example, translation, transcription, or splicing of the target nucleic acid.
One type of antisense mechanism involving degradation of target RNA is RNase H
mediated antisense. RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. It is known in the art that single-stranded antisense compounds which are "DNA-like"
elicit RNase H activity in manmialian cells. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of DNA-like oligonucleotide-mediated inhibition of gene expression.
In certain embodiments, a conjugate group comprises a cleavable moiety. In certain embodiments, a conjugate group comprises one or more cleavable bond. In certain embodiments, a conjugate group comprises a linker. In certain embodiments, a linker comprises a protein binding moiety. In certain embodiments, a conjugate group comprises a cell-targeting moiety (also referred to as a cell-targeting group).
In certain embodiments a cell-targeting moiety comprises a branching group. In certain embodiments, a cell-targeting moiety comprises one or more tethers. In certain embodiments, a cell-targeting moiety comprises a carbohydrate or carbohydrate cluster.
ii. Certain Cleavable Moieties In certain embodiments, a cleavable moiety is a cleavable bond. In certain embodiments, a cleavable moiety comprises a cleavable bond. In certain embodiments, the conjugate group comprises a cleavable moiety. In certain such embodiments, the cleavable moiety attaches to the antisense oligonucleotide. In certain such embodiments, the cleavable moiety attaches directly to the cell-targeting moiety. In certain such embodiments, the cleavable moiety attaches to the conjugate linker. In certain embodiments, the cleavable moiety comprises a phosphate or phosphodiester. In certain embodiments, the cleavable moiety is a cleavable nucleoside or nucleoside analog. In certain embodiments, the nucleoside or nucleoside analog comprises an optionally protected heterocyclic base selected from a purine, substituted purine, pyrimidine or substituted pyrimidine. In certain embodiments, the cleavable moiety is a nucleoside comprising an optionally protected heterocyclic base selected from uracil, thymine, cytosine, 4-N-benzoylcytosine, 5-methylcytosine, 4-N-benzoy1-5-methylcytosine, adenine, 6-N-benzoyladenine, guanine and 2-N-isobutyrylguanine. In certain embodiments, the cleavable moiety is 2'-deoxy nucleoside that is attached to the 3' position of the antisense oligonucleotide by a phosphodiester linkage and is attached to the linker by a phosphodiester or phosphorothioate linkage. In certain embodiments, the cleavable moiety is 2'-deoxy adenosine that is attached to the 3' position of the antisense oligonucleotide by a phosphodiester linkage and is attached to the linker by a phosphodiester or phosphorothioate linkage. In certain embodiments, the cleavable moiety is 2'-deoxy adenosine that is attached to the 3' position of the antisense oligonucleotide by a phosphodiester linkage and is attached to the linker by a phosphodiester linkage.
In certain embodiments, the cleavable moiety is attached to the 3' position of the antisense oligonucleotide. In certain embodiments, the cleavable moiety is attached to the 5' position of the antisense oligonucleotide. In certain embodiments, the cleavable moiety is attached to a 2' position of the antisense oligonucleotide. In certain embodiments, the cleavable moiety is attached to the antisense oligonucleotide by a phosphodiester linkage. In certain embodiments, the cleavable moiety is attached to the linker by either a phosphodiester or a phosphorothioate linkage. In certain embodiments, the cleavable moiety is attached to the linker by a phosphodiester linkage. In certain embodiments, the conjugate group does not include a cleavable moiety.

In certain embodiments, the cleavable moiety is cleaved after the complex has been administered to an animal only after being internalized by a targeted cell. Inside the cell the cleavable moiety is cleaved thereby releasing the active antisense oligonucleotide. While not wanting to be bound by theory it is believed that the cleavable moiety is cleaved by one or more nucleases within the cell.
In certain embodiments, the one or more nucleases cleave the phosphodiester linkage between the cleavable moiety and the linker. In certain embodiments, the cleavable moiety has a structure selected from among the following:
0=P-OH
:ON43)(1 0=P-OH 0=P-OH
(1) NoB)(2 0=P-OH
0=P-OH
o 0=1-0H

L(..ON,Bx c())/13)(2 /0..11(3 ; and , z 0=P-OH 0=P-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 aN
0=P-OH
iii. Certain Linkers In certain embodiments, the conjugate groups comprise a linker. In certain such embodiments, the linker is covalently bound to the cleavable moiety. In certain such embodiments, the linker is covalently bound to the antisense oligonucleotide. In certain embodiments, the linker is covalently bound to a cell-targeting moiety. In certain embodiments, the linker further comprises a covalent attachment to a solid support. In certain embodiments, the linker further comprises a covalent attachment to a protein binding moiety. In certain embodiments, the linker further comprises a covalent attachment to a solid support and further comprises a covalent attachment to a protein binding moiety. In certain embodiments, the linker includes multiple positions for attachment of tethered ligands. In certain embodiments, the linker includes multiple positions for attachment of tethered ligands and is not attached to a branching group. In certain embodiments, the linker further comprises one or more cleavable bond. In certain embodiments, the conjugate group does not include a linker.
In certain embodiments, the linker includes at least a linear group comprising groups selected from alkyl, amide, disulfide, polyethylene glycol, ether, thioether (-S-) and hydroxylamino (-0-N(H)-) groups. In certain embodiments, the linear group comprises groups selected from alkyl, amide and ether groups. In certain embodiments, the linear group comprises groups selected from alkyl and ether groups. In certain embodiments, the linear group comprises at least one phosphorus linking group.
In certain embodiments, the linear group comprises at least one phosphodiester group. In certain embodiments, the linear group includes at least one neutral linking group. In certain embodiments, the linear group is covalently attached to the cell-targeting moiety and the cleavable moiety. In certain embodiments, the linear group is covalently attached to the cell-targeting moiety and the antisense oligonucleotide. In certain embodiments, the linear group is covalently attached to the cell-targeting moiety, the cleavable moiety and a solid support. In certain embodiments, the linear group is covalently attached to the cell-targeting moiety, the cleavable moiety, a solid support and a protein binding moiety. In certain embodiments, the linear group includes one or more cleavable bond.
In certain embodiments, the linker includes the linear group covalently attached to a scaffold group.
In certain embodiments, the scaffold includes a branched aliphatic group comprising groups selected from alkyl, amide, disulfide, polyethylene glycol, ether, thioether and hydroxylamino groups. In certain embodiments, the scaffold includes a branched aliphatic group comprising groups selected from alkyl, amide and ether groups. In certain embodiments, the scaffold includes at least one mono or polycyclic ring system.
In certain embodiments, the scaffold includes at least two mono or polycyclic ring systems. In certain embodiments, the linear group is covalently attached to the scaffold group and the scaffold group is covalently attached to the cleavable moiety and the linker. In certain embodiments, the linear group is covalently attached to the scaffold group and the scaffold group is covalently attached to the cleavable moiety, the linker and a solid support. In certain embodiments, the linear group is covalently attached to the scaffold group and the scaffold group is covalently attached to the cleavable moiety, the linker and a protein binding moiety. In certain embodiments, the linear group is covalently attached to the scaffold group and the scaffold group is covalently attached to the cleavable moiety, the linker, a protein binding moiety and a solid support. In certain embodiments, the scaffold group includes one or more cleavable bond.

In certain embodiments, the linker includes a protein binding moiety. In certain embodiments, the protein binding moiety is a lipid such as for example including but not limited to cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-0(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, 03-(oleoyl)lithocholic acid, 03-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine), a vitamin (e.g., folate, vitamin A, vitamin E, biotin, pyridoxal), a peptide, a carbohydrate (e.g., monosaccharide, disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, polysaccharide), an endosomolytic component, a steroid (e.g., uvaol, hecigenin, diosgenin), a terpene (e.g., triterpene, e.g., sarsasapogenin, friedelin, epifriedelanol derivatized lithocholic acid), or a cationic lipid. In certain embodiments, the protein binding moiety is a C16 to C22 long chain saturated or unsaturated fatty acid, cholesterol, cholic acid, vitamin E, adamantane or 1-pentafluoropropyl.
In certain embodiments, a linker has a structure selected from among:

\ b 0 ,, N I
N CNI-)0,scs H -- N r'HiL0 vN.wLi 0 ' , H ( rL0-1 ( o) n A Jvvv l I
's N

0, JO-P-OH ,
8 P
N I 0 QN.,sss 1-NH
\ Ivv OH
\ , N OH;
41.,,) 0 JWV
N' I
I I
0 0, \I, - NOp00 I
0, N n 0 N Y 41/4z,)1(S-S'WILO
N .
H izi t,,nrH,(,,)Li 0n ck N ' 6sC S'S'WLI 0 I
O,, HHHH H
,222(N,p,nN.m,N ,e1 .N 1, - Ni4r.fp,,,051 H .
s,_vLi 0 0JvW

JvVV
/
I

0 0 ,0 \ 1, - 1 0... 00N, p,,0 1 ,P, 0 CS H )n \ /-(-C
\i,..=

1-1-(jC10 \1., N ; and S-S
.= n N 0 N
ri H ,wLi 0 "s N
H
N,HrIL0 wherein each n is, independently, from 1 to 20; and p is from 1 to 6.

In certain embodiments, a linker has a structure selected from among:
srri \
Prr\j 0 00.
,A 0 N)0 N O )N' sJ.Pi \
o.
o N)0A- Jsrrj \
H q.
7(ni rH 0 ;
n i H N
n )A

I H
n JJJ'' 0 -I- n \ 0 0..
)0A , \
N Os = 0 NO
r ti 'S n H
"s)-N =-(C0-1N 0 ;
n H n e \.
\
0 e sJ=rj ((il \I )(r N A
n H
q. xrri \ n H

N I
0 -P =0 N 0 I
0 -P=0 H OH ; 0 OH
and s,w_l H
c&N N
n HO
wherein each n is, independently, from 1 to 20.

In certain embodiments, a linker has a structure selected from among:
µAHHO 0 0 0 0 H
H H

0 µ)L(1,1 csssQ,S,ski,i .
/ ; " n n , N n 0 n 0 H n / µ
Th\li .rNS-1 =
N7 ; CS5SyV rryµ
0 n H
0 0 ' H
N H
css'r NH H..00/`H n n / H H
N Q H
YInNcsss ; cli0/'W"sss ; and O o 0 H
H N
css.'i NICO/C/).ncsss n wherein n is from 1 to 20.

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

H
\.)LH H
eyNIrmNy'L . ,zzNA `,2a.)L(L-rssc .

0; .
AN H
" n n n 0 H n C k 1 H 0 cs Ni,.,ri rii A ; t171,_______ H
N-----N ; isssi \ . csss-N ;
H 0 n H r' y ,ss n c"

t\-11 csss NH,H.-0/`H n csss; cs'i .r NH -H^0C0/1'eHn isss ; citIOI \ 1 ;
n n n n H H kil H
cirvri.ri NI.H0Q0/`Hn sss , If n n Tr Kii 0 0/4n csss ;
N . csssi J, -N
n 0 0 AOH OH
"n_ n -n _ -n _ -n cssW(1\lii- csss and csssreOL

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

strj\
:Pr\j Q
Q C)10)zzL

0 N ,z2L).L.r H

'Prj\j 0.
C)0)2L N
I I

H H
- H

/N I
N
I JVW\ rO-P-OH
n 4S<0 .
' q 1-N'H
C)0)22L

)" Ell ir N NH
"() ; I

0 -^j" 0 \u'.
(">o N OH

0 CS1¨S 0 H N
I JVVV

N,Hg c L0 ) \i...
104 C) OH

i N
'HgLO ;
HJv I 0, 0 0, HHHH H
i4_ Np 3 "4 C-N--) o,ssc = r5( 0 H ' SS' 0 ;

/
JVW
I
IJVW
0, I 0 \ 1 ...
0..=0, ,0 0, OH
S¨/L\

cl /C 0 CS
H
1 0 CN --)(:),05 = S-S 0 N
vN s,S,H5Lo ;
sk ;
47,)L40 N NH
'HgLO
H

1 \I... 0 ,0 0 NO-.4-\I... 0 ' 0 1 /C
0-4 o- =13 o 1 N¨s o \,,,"<, ) N
H N
JVVI/

H H N µ222( Vi& 0, , H 0 CI\ 31 C)csss ''1/4.)SSACgLO ;
and Jvvv I
1-1L\ 0.44-0--P
1 ' 0 S¨S 0 N
H
'sss N -r N $')gLO .

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

H
H
)c N
v.1.1.,,,N,,Ti.õ..,-...õ.....,........1c---...y....µ. y...........-...
H H ' 0 H

H ID vitõ,..õ. N ,r.õ,),õ,;sss ;

rr csss A..
'''N-====-'===(--"c)rN1-)S'N , ,i1c...--11---(õr"--õki,---6\ ; i \ ;

0 4 il H
H
csssyr-8( N cX:(\/N',50 ; i H H
Ncsss .
N'000// ' H H H ; and cOsN cso ; cOsN CP0/./N csss H
H
css-r-r N'-0//N csss =

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

H
H
,,z2.) N 1)=( N . ,zza.A. i \j hi A ;
H 0 ' 0 0 ' 0 H
H ji y 1 1.,%.õ... N ..1c.,jty ; 0 HN

,ssL N rr' H
. 0 csrVrgyµ ;

H
H
NI -'()CµN
o o o 0 H
H H
csss N cos ;
csscr N '.0Q0//N

ssCOss ; 100,1 ; 5sC000,sss ;
H s 9 0 ¨F1)-0 -õ,000 csss ;
OH "3 3 H

1-0¨P-0 ...,p,0 00¨P-0-1 ; ily^OL3 NK6µ and OH "3 3 OH H

CI? 5 cW(NlfrYOI¨O¨
H 6 OH =

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

µ).Hri0 and µ)ri wherein n is from 1 to 20.
In certain embodiments, a linker has a structure selected from among:
of; ssC00/\/\/ ; and ssCooe\/\csss .
In certain embodiments, a linker has a structure selected from among:
/OH

and OH "3 "3 OH OH "3 3 =
In certain embodiments, a linker has a structure selected from among:

csssWL N `1A.

0 and 0 In certain embodiments, the conjugate linker has the structure:
.rrr\i NO

6 0 .
In certain embodiments, the conjugate linker has the structure:

`ziL)C.)LN(`-r0-1 In certain embodiments, a linker has a structure selected from among:

csscrhYLN)r 0 and 0 In certain embodiments, a linker has a structure selected from among:

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

Jvvv o \
HO ' 0-1L0 __ 1 µ)L I \rd`= N -.)jisss =
NH 0 n (13oH / n ' julv CH3 /m ; 01) JvW
>1_ H 0 ( ln H 0 0 )11t- 1 n J`rls' ePrjj. 0(0 n ( ,v1\1zANNLNictil cssj. N (`=)n N .22'.
)n H 8 ( )n H =
, n .. LJ
= =
" el' % *
Jvw Ill ilt0\;222- CH3 n CH3 :

i n ic> /m; 0 0 CH3PIIIIk n (t N H oss t,md ?
T o ) I
o ¨
NH ( ¨NH, csss Jr )n \ n )o L
, 0 vuv , (NH
csssl(N)) ,,zz. n 0 n H
H ' 0 , css'N \ ; css'N
H Nj=LNA
H ;
H 11 0( i '722. 10,!)-1--NH
0 n 0 0 n H 0 µfr)L N j=L NA
\
n H H rfi)LN N j=N A ; and 0 di v NH

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

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

,( ( 0 \ 0 0 0 II
`'2z.(N n-rµ = HO ' 0 ¨P ¨0 ___ 1 .
NH 0 n OH n e I CH3 im ' 0 n /4/1..
H 0 ( In H 0 0 \z.
n n 'sssr(1/4L N N )22- ;
Hn H 8 Hn H i n H ;
.r=P'4 rr's 0 n(0 =
n m %NW
I
NH (C):11 4m CH3 V( \ n ' c- i ,N
0 .
, CH

\ rfss m H NH rsss CH3 f.911, n ( / n rid 0 ;and I 0 __ e NH (,NH rssf n \ n nO 0 I
V-- NH rr H
/m wherein each n is, independently, from 1 to 20; and m is from 2 to 6.
In certain embodiments, a branching group has a structure selected from among:

0 4t.

/ .k.
µ)HrAN , H

vvu vv avvy I ,¨
N11-1 \cSrr NH
0 0 CI)) 0 14 . A jj-L
\ N N csss 0 ; 01) ; ci,N \ = V-- N H / ' , JVVV H
vvv 0 \ 0 HN NH
) `zzL)LNH
H jj H 0 css\ N
css'N N Jcsss ;
N Thr css' ;
H ck N ' H

H O/

H N rsss v NH

µ)1---NH NH

H
and 'LA FNi 11 NHAcsss O/
vOs NH .r NH
v In certain embodiments, a branching group has a structure selected from among:
\ I
Ai, 'i-L, A1 /
,A1 --- A1 t - 1 1K, in ' in Ki¨ A1 / and 1,,, 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:
JVVV
Ai Ai Ai 1¨/ )n(f1 A A1-1 j, (fn i A 1_ )A n n 1 A1 and 1¨
frµ A1 (n i sss' 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:
n and Ai \53 wherein A1 is 0, S, C=0 or NH; and each n is, independently, from 1 to 20.
In certain embodiments, a branching group has a structure selected from among:
0-, C) In certain embodiments, a branching group has a structure selected from among:

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

(s's 41z( 2. Certain Tethers In certain embodiments, conjugate groups comprise one or more tethers covalently attached to the branching group. In certain embodiments, conjugate groups comprise one or more tethers covalently attached to the linking group. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, ether, thioether, disulfide, amide and polyethylene glycol groups in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, substituted alkyl, ether, thioether, disulfide, amide, phosphodiester and polyethylene glycol groups in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, ether and amide groups in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl, substituted alkyl, phosphodiester, ether and amide groups in any combination. In certain embodiments, each tether is a linear aliphatic group comprising one or more groups selected from alkyl and phosphodiester in any combination.
In certain embodiments, each tether comprises at least one phosphorus linking group or neutral linking group.
In certain embodiments, the tether includes one or more cleavable bond. In certain embodiments, the tether is attached to the branching group through either an amide or an ether group. In certain embodiments, the tether is attached to the branching group through a phosphodiester group. In certain embodiments, the tether is attached to the branching group through a phosphorus linking group or neutral linking group. In certain embodiments, the tether is attached to the branching group through an ether group.
In certain embodiments, the tether is attached to the ligand through either an amide or an ether group. In certain embodiments, the tether is attached to the ligand through an ether group. In certain embodiments, the tether is attached to the ligand through either an amide or an ether group. In certain embodiments, the tether is attached to the ligand through an ether group.
In certain embodiments, each tether comprises from about 8 to about 20 atoms in chain length between the ligand and the branching group. In certain embodiments, each tether group comprises from about 10 to about 18 atoms in chain length between the ligand and the branching group. In certain embodiments, each tether group comprises about 13 atoms in chain length.
In certain embodiments, a tether has a structure selected from among:

=:r11Yµ.
0 , n H n H H
p " n H ' 'n Hi _NN
¨N
¨ la- ,ssi N N =
H Thr A9- N9- ;
n n n n n 1¨N 0 0 'n N ; ;and if H'n wherein each n is, independently, from 1 to 20; and each p is from 1 to about 6.
In certain embodiments, a tether has a structure selected from among:

N 0()>\ ; N csss ;

and s'ssossss In certain embodiments, a tether has a structure selected from among:
H H
csss N .(,)r N
\ in n H "n wherein each n is, independently, from 1 to 20.

In certain embodiments, a tether has a structure selected from among:
0 Zi and `sssHjLNI¨H)22-mi mi mi H m 1 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:
css5rN

In certain embodiments, a tether has a structure selected from among:
0 I IX. 0 COOH OH
and cskpJ-L )(0¨P-0 I 4-4)L
mi 01H mi m N

wherein Z2 is H or CH3; and each m1 is, independently, from 0 to 20 wherein at least one m1 is greater than 0 for each tether.
In certain embodiments, a tether has a structure selected from among:

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

3. Certain Li2ands 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-glucopyranose (13-muramic acid), 2-Deoxy-2-methylamino-L-glucopyranose, 4,6-Dideoxy-4-formamido-2,3-di-O-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-acetyl-1-thio-6-0-trityl-a-D-glucopyranoside, 4-Thio-13-D-galactopyranose, and ethyl 3,4,6,7-tetra-0-acetyl-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 13-form: 2-(Acetylamino)-2-deoxy-13-D-galactopyranose and a-form: 2-(Acetylamino)-2-deoxy-D-galactopyranose. In certain embodiments, both the 13-form: 2-(Acetylamino)-2-deoxy-13-D-galactopyranose and a-form: 2-(Acetylamino)-2-deoxy-D-galactopyranose may be used interchangeably. Accordingly, in structures in which one form is depicted, these structures are intended to include the other form as well. For example, where the structure for an a-form: 2-(Acetylamino)-2-deoxy-D-galactopyranose is shown, this structure is intended to include the other form as well. In certain embodiments, In certain preferred embodiments, the 13-form 2-(Acetylamino)-2-deoxy-D-galactopyranose is the preferred embodiment.

..4k440..,,OH

Holy1/41/1H\1 e OH
2-(Acetylamino)-2-deoxy-D-galactopyranose OH
OH

HO 0¨
NHAc 2-(Acetylamino)-2-deoxy-p-D-galactopyranose OH
OH

HO
NHAc Oci 2-(Acetylamino)-2-deoxy-a-D-galactopyranose In certain embodiments one or more ligand has a structure selected from among:
O
OH H
OH
HO C. 0 _____________ HO*
HO¨ µ---T¨C)----..r.?._\__O-1 HO OH
and Ri Ri Ri wherein each R1 is selected from OH and NHCOOH.

In certain embodiments one or more ligand has a structure selected from among:
HOOH OH HO HO
OH .._\:)1......-i ..\
0 HO Ho 0 0 HO --¨"\------\, ¨n ,s.r ; Nojs . H HO Ns, ; HO -, NHAc r OH

\s"s3 HOOH OH
N . HO"--1--7\ OH HOOH
HO No.ss , 0 OH C)r 0 HO OH
OH
HO HO Ncsrs ; HOOO)\ , and OH
HO
_....\:A...,- ..\i HO
0 ____________ HO OH

HO
0\ , 0 is' In certain embodiments one or more ligand has a structure selected from among:
HOOH
_.0_\r HO N, NHAc r =
In certain embodiments one or more ligand has a structure selected from among:
HOOH
0 n HO-----"\------\,¨N,rr NHAc r =

i. Certain Coniu2ates In certain embodiments, conjugate groups comprise the structural features above. In certain such embodiments, conjugate groups have the following structure:
HO OH

HN
HO
In NHAc 0 HO OH
01.. 1 In H
HO
n n n NHAc OH
HO HN

N
__,.\... ,....\70 r,cir n HO
NHAc 0 =
wherein each n is, independently, from 1 to 20.
In certain such embodiments, conjugate groups have the following structure:
HO OH
HO
NHAc 0 HO H 0.-HO -, ¨I
\/\/\ I\IN/N N y ----N
NHAc 0 0 o/
OH
HO
H
HN
_sl..o....\.zor 4 HO

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

HO H

O=P¨OH OH

NHAc 0 Bx 0 )n HO H 0 ___________ \C )?
N
HO (D.w.,11-0 N IY7 H n 0¨P=X
NHAc 0 n ) OH
0 0 n )n HO H
N....+K 0 HO 0 n NHAc wherein each n is, independently, from 1 to 20;
Z is H or a linked solid support;
Q is an antisense compound;
X is 0 or S; and Bx is a heterocyclic base moiety.
In certain such embodiments, conjugate groups have the following structure:
HO H
0=P¨OH
OH

HO
NHAc \())/Bx NO :
0f---N H H
N.______H
HOil -----N 0--P==X
I
NHAc 0 0 OH

HO OH
--1...1\------ H HN¨j:

N=----,/
HO O
NHAc In certain such embodiments, conjugate groups have the following structure:
HO H
H

=P
N N

HO's\----- N(...), 3 ------ti NHAc--1:-. Y

HO OH 0-...,..... 0 N
0,,y3--,...... .(c.,N,,,,,-...N........Ø.............----N

HO H O¨P=0 I
NHAc 0 / 0 OH

HO OH
_\,..Ø......\______ HN
../ O
HO
(3 NHAc In certain such embodiments, conjugate groups have the following structure:
HOOF!
HO-40 , 0 ...\-)1)\n-k AcHN OH ()n HO OH

HO---1"2-\, -FL
n 010 __________________________________ I
no' AcHN OH
0' H 0 \
.131,0..... in O "n OH
NHAc .
In certain such embodiments, conjugate groups have the following structure:

HOOH

HO-4) AcHN 0 1 0, OH
HOOH
0 0, ii HO---(2*--\, 0-Pi`oo I
AcHN OH (31 HO OH y P, l 0 HO OH
NHAc .
In certain such embodiments, conjugate groups have the following structure:
HOOH
0 r, P
HO,-/Vr\ -K
n 0 1 , AcHN 0 ()n HOOH OH
_..r.12._\,0 0 O 0 H il / N
O 'W\ -il, ,.(0),..N¨(N_____i n 0 I 0---Hio----,0 AcHN OH OH cis, HOOH 0 (31 An H04=0 'Hrri-611) HO
NHAc .
In certain such embodiments, conjugate groups have the following structure:

NOON
AcHN 0 0, OH

0 0, 0 HOrs2._\zo AcHN OH 0' OH Os' HO-P=0 HO H OH
P, HO
NHAc In certain such embodiments, conjugate groups have the following structure:

HO-P=0 OON
HO-P=0 HO OH On HO oW\
n 010 \OH
AcHN OH 1) 0 HO OH fl (On 1C) 0 HO C)1 n 0 1 0 n e N701=0 AcHN OH
OH
0' HO n OH
NHAc In certain such embodiments, conjugate groups have the following structure:

HO¨P=0 0 ,_1\1 0---= ),õN 'Ns---/
a HO¨P=0 O

H OH
....K
--\OH
AcHN

HO OH (03 0 (31 I
________________________________________________ .,(:)-1)=(i) AcHN OH 0' OH

HO H II
P, OH
HO
NHAc .
In certain embodiments, conjugates do not comprise a pyrrolidine.
In certain such embodiments, conjugate groups have the following structure:

s 9 _Zi4N
¨1=1)-0¨NfkiN Nrj 0- '\ __ 1 HOOH

0=P-0 HOOH -AcHN 0 O
H H (:) 0 0 --C=C), 0 H \
AcHN 0 0 0' OH
H 00 HHN---kj H.......y 0 H0_,1 0--___--rN

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

HOOH
HO*4,0N-----N-----N....----N -119, 0 , 0 AcHN 0- --HOOH
0 0, 0 o-11-O

AcHN 0 6 '
9 I
o=f)-o-HO H P, NHAc In certain such embodiments, conjugate groups have the following structure:
HO OH

AcHN N---N----)r-N H
"\,N
0 )1-----1 0 0 OH

N"----N----N---r\---0,......---NH

HO--V/7-1 0 0 (Do 0 NHAc FINN H --e0 OH /--/¨%
HO) ,\õ>/
) HO
NHAc .
In certain such embodiments, conjugate groups have the following structure:

U4 _.,..r.?...\0(- N) AcHN N
O
HOOH 0 N o 0 HO ON

AcHN o7 HOOH
HO__....r.C.)...\CY1rN"---10 AcHN .
In certain such embodiments, conjugate groups have the following structure:

AcHN N

HO OrN

AcHN 0 HOOH

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

HO "4 H Ell 1 AcHN
NOON
AcHN .
In certain such embodiments, conjugate groups have the following structure:
NOON H
AcHN

H H
AcHN 0 NOON
N--(0 AcHN .
In certain such embodiments, conjugate groups have the following structure:

OH OH
HO
AcHN
OH OH
H0*., 0 CirH 0 H 0 AcHN H 0H 0 o( HO j¨NH
HO
NHAc In certain such embodiments, conjugate groups have the following structure:
OH OH

HOTELI
AcHN
OH OH
AcHN H 0 0 0 0 r H0 OH j¨NH
HO
NHAc In certain such embodiments, conjugate groups have the following structure:
gH
HOOH

AcHN
0=P¨OH
HOOH

AcHN
0=P¨OH
HOOH
¨[cm, AcHN
In certain such embodiments, conjugate groups have the following structure:

pH
HOOH

AcHN
0=P¨OH
HOOH
HO....7.2..\00(NR.' 0 AcHN
0=P¨OH
HOOH
HO
AcHN ó.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
HO.0 n AcHN

y HO
AcHN /07 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
HO-0 n AcHN

y HO
AcHN ,OZ
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
AcHN
HOOH N
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.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
N

HO A
AcHN N z-uN

HO
H
AcHN H
I\1 HOOH
/

AcHN
wherein Y and Z are independently selected from a C 1-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 NYN
AcHN N n HO
H H
AcHNzZ
HOOH

AcHN
wherein Y and Z are independently selected from a C1-c12 substituted or unsubstituted alkyl group, or a group comprising exactly one ether or exactly two ethers, an amide, an amine, a piperidine, a phosphate, a phosphodiester, or a phosphorothioate.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH

YN
AcHN N n HO
H H
AcHNzZ
HOOH
HOoe0 0 AcHN
wherein Y and Z are independently 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:

HO

AcHN 0 HOOH
n HO

AcHN
HOOHsjK, 0 HO
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:

AcHN 0 HOOH
HO 1114) AcHNHOOH n 0 HO
AcHN
wherein m is 4, 5, 6, 7, or 8, and n is 1, 2, 3, or 4.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH

o AcHN
HO --AcHN
OH0H r H
HO _______ 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 HO
o AcHN
H0-'-AcHN
OH0H r H
HO _______ 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 -'--1\1 AcHN
OH0H 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.
In certain embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH

HOOH
HO 0 ___ AcHN
OH0H r H
H0.7 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 __ 1*---V
AcHN

O
N )C
HO
AcHN
HOOH
HO
AcHN
wherein Y is selected from a Cl-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
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.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
HO
AcHN

O
N A
HO N
AcHN
HOOH
HO
AcHN
wherein Y is selected from a Ci-C12 substituted or unsubstituted alkyl group.
In certain such embodiments, the cell-targeting moiety of the conjugate group has the following structure:
HOOH
./0.pN 0 HO n AcHN

HO -H
AcHN
HOOH
HO /0")C7I 0 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

HO n AcHN

zcye)-NNA
HO nH
AcHN
HOOH , HO
AcHN
wherein n is 4, 5, 6, 7, or 8.
b. Certain coniu2ated 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:
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.
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(E¨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-F')¨F) wherein A is the antisense oligonucleotide;
B is the cleavable moiety D is the branching group each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
In certain embodiments, a conjugated antisense compound has the following structure:
A¨D¨(¨E¨F) 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
¨ OH
O¨OH NH2 Nx1,,s, :
LOoN NvAl N_HAc 0 M
[ HO ==_µ,....._.\__.,, 0 C'ND....\
0 H H e HO , 0....,,,,,_,...1_,N [0 ] ________________________________________________________________ P=0 OH

NHAc g 0 _ 0 Linker Cleavable moiety Ligand Tether i I 1 ¨
¨
OH
HO HN.--( \
H
N..,2,../
.......7Ø.....\70ir Branching group NHAc =
In certain embodiments, a conjugated antisense compound has a structure selected from among the following:
Cell targeting moiety HOOH
_ Cleavable moiety ¨
AcHN
OH

HO H _ _ 1 _____ i Nz.4 0 0 --.õ -O
__...r0,.....\yr) II
¨I
HO ----.,-/N I
.........,___-0-P,-0^(0N

- 0" s, N¨

_ AcHN __ OH - 0 0 Tether Ligand _______________________________________________ i "0-113=0 , I
HO H 9 y -.. _ _ ,12....\/0,,,./--____Z------cy 1 0 HO P ASO
OH
NHAc Branching group =
In certain embodiments, a conjugated antisense compound has a structure selected from among the following:

ASO
Cleavable moiety HO¨P=0 o N

HO¨P=0 Cell targeting moiety ' 0 HO OH

0 \OH
AcHN 0-(03 HOOH
_ _____________________________________________________ Conjugate linker HO
0' 1;i1;i3; \/O-1), =O
0"
AcHN _ _ OH
Tether Ligand HO I-1 (;), 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 Cholesterol Lowering Agent" J. Med. Chem. (1995) 38:1846-1852, BIESSEN et al., "Synthesis of Cluster Galactosides with High Affinity for the Hepatic Asialoglycoprotein Receptor"
J. Med. Chem. (1995) 38:1538-1546, LEE et al., "New and more efficient multivalent glyco-ligands for asialoglycoprotein receptor of mammalian hepatocytes" Bioorganic & Medicinal Chemistry (2011) 19:2494-2500, RENSEN et al., "Determination of the Upper Size Limit for Uptake and Processing of Ligands by the Asialoglycoprotein Receptor on Hepatocytes in Vitro and in Vivo" J. Biol. Chem. (2001) 276(40):37577-37584, RENSEN et al., "Design and Synthesis of Novel N-Acetylgalactosamine-Terminated Glycolipids for Targeting of Lipoproteins to the Hepatic Asialoglycoprotein Receptor" J. Med. Chem. (2004) 47:5798-5808, SLIEDREGT
et al., "Design and Synthesis of Novel Amphiphilic Dendritic Galactosides for Selective Targeting of Liposomes to the Hepatic Asialoglycoprotein Receptor" J. Med. Chem. (1999) 42:609-618, and Valentijn et al., "Solid-phase synthesis of lysine-based cluster galactosides with high affinity for the Asialoglycoprotein Receptor" Tetrahedron, 1997, 53(2), 759-770, each of which is incorporated by reference herein in its entirety.
In certain embodiments, conjugated antisense compounds comprise an RNase H
based oligonucleotide (such as a gapmer) or a splice modulating oligonucleotide (such as a fully modified oligonucleotide) and any conjugate group comprising at least one, two, or three GalNAc groups. In certain embodiments a conjugated antisense compound comprises any conjugate group found in any of the following references: Lee, Carbohydr Res, 1978, 67, 509-514; Connolly et al., J Biol Chem, 1982, 257, 939-945; Pavia et al., Int J Pep Protein Res, 1983, 22, 539-548; Lee et al., Biochem, 1984, 23, 4255-4261; Lee et al., Glycoconjugate J, 1987, 4, 317-328; Toyokuni et al., Tetrahedron Lett, 1990, 31, 2673-2676; Biessen et al., J
Med Chem, 1995, 38, 1538-1546; Valentijn et al., Tetrahedron, 1997, 53, 759-770; Kim et al., Tetrahedron Lett, 1997, 38, 3487-3490; Lee et al., Bioconjug Chem, 1997, 8, 762-765; Kato et al., Glycobiol, 2001, 11, 821-829; Rensen et al., J Biol Chem, 2001, 276, 37577-37584; Lee et al., Methods Enzymol, 2003, 362, 38-43; Westerlind et al., Glycoconj J, 2004, 21, 227-241; Lee et al., Bioorg Med Chem Lett, 2006, 16(19), 5132-5135; Maierhofer et al., Bioorg Med Chem, 2007, 15, 7661-7676; Khorev et al., Bioorg Med Chem, 2008, 16, 5216-5231; Lee et al., Bioorg Med Chem, 2011, 19, 2494-2500; Kornilova et al., Analyt Biochem, 2012, 425, 43-46; Pujol et al., Angew Chemie Int Ed Engl, 2012, 51, 7445-7448; Biessen et al., J Med Chem, 1995, 38, 1846-1852; Sliedregt et al., J Med Chem, 1999, 42, 609-618; Rensen et al., J
Med Chem, 2004, 47, 5798-5808; Rensen et al., Arterioscler Thromb Vasc Biol, 2006, 26, 169-175; van Rossenberg et al., Gene Ther, 2004, 11, 457-464; Sato et al., J Am Chem Soc, 2004, 126, 14013-14022; Lee et al., J Org Chem, 2012, 77, 7564-7571; Biessen et al., FASEB J, 2000, 14, 1784-1792; Rajur et al., Bioconjug Chem, 1997, 8, 935-940;
Duff et al., Methods Enzymol, 2000, 313, 297-321; Maier et al., Bioconjug Chem, 2003, 14, 18-29;
Jayaprakash et al., Org Lett, 2010, 12, 5410-5413; Manoharan, Antisense Nucleic Acid Drug Dev, 2002, 12, 103-128; Merwin et al., Bioconjug Chem, 1994, 5, 612-620; Tomiya et al., Bioorg Med Chem, 2013, 21, 5275-5281; International applications W01998/013381; W02011/038356;
W01997/046098;

W02008/098788; W02004/101619; W02012/037254; W02011/120053; W02011/100131;
W02011/163121; W02012/177947; W02013/033230; W02013/075035; W02012/083185;
W02012/083046; W02009/082607; W02009/134487; W02010/144740; W02010/148013;
W01997/020563; W02010/088537; W02002/043771; W02010/129709; W02012/068187;
W02009/126933; W02004/024757; W02010/054406; W02012/089352; W02012/089602;
W02013/166121; W02013/165816; U.S. Patents 4,751,219; 8,552,163; 6,908,903;
7,262,177; 5,994,517;
6,300,319; 8,106,022; 7,491,805; 7,491,805; 7,582,744; 8,137,695; 6,383,812;
6,525,031; 6,660,720;
7,723,509; 8,541,548; 8,344,125; 8,313,772; 8,349,308; 8,450,467; 8,501,930;
8,158,601; 7,262,177;
6,906,182; 6,620,916; 8,435,491; 8,404,862; 7,851,615; Published U.S. Patent Application Publications US2011/0097264; US2011/0097265; U52013/0004427; US2005/0164235;
U52006/0148740;
U52008/0281044; US2010/0240730; U52003/0119724; US2006/0183886;
U52008/0206869;
US2011/0269814; U52009/0286973; US2011/0207799; U52012/0136042;
U52012/0165393;
U52008/0281041; U52009/0203135; US2012/0035115; U52012/0095075;
US2012/0101148;
U52012/0128760; U52012/0157509; U52012/0230938; U52013/0109817;
U52013/0121954;
U52013/0178512; U52013/0236968; US2011/0123520; U52003/0077829;
U52008/0108801; and U52009/0203132; each of which is incorporated by reference in its entirety.
C. Certain Uses and Features In certain embodiments, conjugated antisense compounds exhibit potent target RNA reduction in vivo. In certain embodiments, unconjugated antisense compounds accumulate in the kidney. In certain embodiments, conjugated antisense compounds accumulate in the liver. In certain embodiments, conjugated antisense compounds are well tolerated. Such properties render conjugated antisense compounds particularly useful for inhibition of many target RNAs, including, but not limited to those involved in metabolic, cardiovascular and other diseases, disorders or conditions. Thus, provided herein are methods of treating such diseases, disorders or conditions by contacting liver tissues with the conjugated antisense compounds targeted to RNAs associated with such diseases, disorders or conditions. Thus, also provided are methods for ameliorating any of a variety of metabolic, cardiovascular and other diseases, disorders or conditions with the conjugated antisense compounds of the present invention.
In certain embodiments, conjugated antisense compounds are more potent than unconjugated counterpart at a particular tissue concentration. Without wishing to be bound by any theory or mechanism, in certain embodiemtns, the conjugate may allow the conjugated antisense compound to enter the cell more efficiently or to enter the cell more productively. For example, in certain embodiments conjugated antisense compounds may exhibit greater target reduction as compared to its unconjugated counterpart wherein both the conjugated antisense compound and its unconjugated counterpart are present in the tissue at the same concentrations. For example, in certain embodiments conjugated antisense compounds may exhibit greater target reduction as compared to its unconjugated counterpart wherein both the conjugated antisense compound and its unconjugated counterpart are present in the liver at the same concentrations.
Productive and non-productive uptake of oligonucleotides has beed discussed previously (See e.g.
Geary, R. S., E. Wancewicz, et al. (2009). "Effect of Dose and Plasma Concentration on Liver Uptake and Pharmacologic Activity of a 2'-Methoxyethyl Modified Chimeric Antisense Oligonucleotide Targeting PTEN." Biochem. Pharmacol. 78(3): 284-91; & Koller, E., T. M. Vincent, et al.
(2011). "Mechanisms of single-stranded phosphorothioate modified antisense oligonucleotide accumulation in hepatocytes." Nucleic Acids Res. 39(11): 4795-807). Conjugate groups described herein may improve productive uptake.
In certain embodiments, the conjugate groups described herein may further improve potency by increasing the affinity of the conjugated antisense compound for a particular type of cell or tissue. In certain embodiments, the conjugate groups described herein may further improve potency by increasing recognition of the conjugated antisense compound by one or more cell-surface receptors. .
In certain embodiments, the conjugate groups described herein may further improve potency by facilitating endocytosis of the conjugated antisense compound.
In certain embodiments, the cleavable moiety may further improve potency by allowing the conjugate to be cleaved from the antisense oligonucleotide after the conjugated antisense compound has entered the cell. Accordingly, in certain embodiments, conjugated antisense compounds can be administed at doses lower than would be necessary for unconjugated antisense oligonucleotides.
Phosphorothioate linkages have been incorporated into antisense oligonucleotides previously. Such phosphorothioate linkages are resistant to nucleases and so improve stability of the oligonucleotide. Further, phosphorothioate linkages also bind certain proteins, which results in accumulation of antisense oligonucleotide in the liver. Oligonucleotides with fewer phosphorothioate linkages accumulate less in the liver and more in the kidney (see, for example, Geary, R., "Pharmacokinetic Properties of 2'-0-(2-Methoxyethyl)-Modified Oligonucleotide Analogs in Rats," Journal of Pharmacology and Experimental Therapeutics, Vol. 296, No. 3, 890-897; & Pharmacological Properties of 2 '-0-Methoxyethyl Modified Oligonucleotides in Antisense a Drug Technology, Chapter 10, Crooke, S.T., ed., 2008) In certain embodiments, oligonucleotides with fewer phosphorothioate internculeoside linkages and more phosphodiester internucleoside linkages accumulate less in the liver and more in the kidney. When treating diseases in the liver, this is undesibable for several reasons (1) less drug is getting to the site of desired action (liver); (2) drug is escaping into the urine; and (3) the kidney is exposed to relatively high concentration of drug which can result in toxicities in the kidney. Thus, for liver diseases, phosphorothioate linkages provide important benefits.

In certain embodiments, however, administration of oligonucleotides uniformly linked by phosphoro-thioate internucleoside linkages induces one or more proinflammatory reactions. (see for example: J Lab Clin Med. 1996 Sep;128(3):329-38. "Amplification of antibody production by phosphorothioate oligodeoxynucleotides". Branda et al.; and see also for example: Toxicologic Properties in Antisense a Drug Technology, Chapter 12, pages 342-351, Crooke, S.T., ed., 2008). In certain embodiments, administration of oligonucleotides wherein most of the internucleoside linkages comprise phosphorothioate internucleoside linkages induces one or more proinflammatory reactions.
In certain embodiments, the degree of proinflammatory effect may depend on several variables (e.g.
backbone modification, off-target effects, nucleobase modifications, and/or nucleoside modifications) see for example: Toxicologic Properties in Antisense a Drug Technology, Chapter 12, pages 342-351, Crooke, S.T., ed., 2008). In certain embodiments, the degree of proinflammatory effect may be mitigated by adjusting one or more variables. For example the degree of proinflammatory effect of a given oligonucleotide may be mitigated by replacing any number of phosphorothioate internucleoside linkages with phosphodiester internucleoside linkages and thereby reducing the total number of phosphorothioate internucleoside linkages.
In certain embodiments, it would be desirable to reduce the number of phosphorothioate linkages, if doing so could be done without losing stability and without shifting the distribution from liver to kidney. For example, in certain embodiments, the number of phosphorothioate linkages may be reduced by replacing phosphorothioate linkages with phosphodiester linkages. In such an embodiment, the antisense compound having fewer phosphorothioate linkages and more phosphodiester linkages may induce less proinflammatory reactions or no proinflammatory reaction. Although the the antisense compound having fewer phosphoro-thioate linkages and more phosphodiester linkages may induce fewer proinflammatory reactions, the antisense compound having fewer phosphorothioate linkages and more phosphodiester linkages may not accumulate in the liver and may be less efficacious at the same or similar dose as compared to an antisense compound having more phosphorothioate linkages. In certain embodiments, it is therefore desirable to design an antisense compound that has a plurality of phosphodiester bonds and a plurality of phosphorothioate bonds but which also possesses stability and good distribution to the liver.
In certain embodiments, conjugated antisense compounds accumulate more in the liver and less in the kidney than unconjugated counterparts, even when some of the phosporothioate linkages are replaced with less proinflammatory phosphodiester internucleoside linkages. In certain embodiments, conjugated antisense compounds accumulate more in the liver and are not excreted as much in the urine compared to its unonjugated counterparts, even when some of the phosporothioate linkages are replaced with less proinflammatory phosphodiester internucleoside linkages. In certain embodiments, the use of a conjugate allows one to design more potent and better tolerated antisense drugs. Indeed, in certain emobidments, conjugated antisense compounds have larger therapeutic indexes than unconjugated counterparts. This allows the conjugated antisense compound to be administered at a higher absolute dose, because there is less risk of proinflammatory response and less risk of kidney toxicity. This higher dose, allows one to dose less frequently, since the clearance (metabolism) is expected to be similar.
Further, because the compound is more potent, as described above, one can allow the concentration to go lower before the next dose without losing therapeutic activity, allowing for even longer periods between dosing.
In certain embodiments, the inclusion of some phosphorothioate linkages remains desirable. For example, the terminal linkages are vulnerable to exonucleoases and so in certain embodiments, those linkages are phosphorothioate or other modified linkage. Internucleoside linkages linking two deoxynucleosides are vulnerable to endonucleases and so in certain embodiments those those linkages are phosphorothioate or other modified linkage. Internucleoside linkages between a modified nucleoside and a deoxynucleoside where the deoxynucleoside is on the 5' side of the linkage deoxynucleosides are vulnerable to endonucleases and so in certain embodiments those those linkages are phosphorothioate or other modified linkage.
Internucleoside linkages between two modified nucleosides of certain types and between a deoxynucleoside and a modified nucleoside of certain typ where the modified nucleoside is at the 5' side of the linkage are sufficiently resistant to nuclease digestion, that the linkage can be phosphodiester.
In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 16 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 15 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 14 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 13 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 12 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 11 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 10 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 9 phosphorthioate linkages. In certain embodiments, the antisense oligonucleotide of a conjugated antisense compound comprises fewer than 8 phosphorthioate linkages.
In certain embodiments, antisense compounds comprsing one or more conjugae group described herein has increased activity and/or potency and/or tolerability compared to a parent antisense compound lacking such one or more conjugate group. Accordingly, in certain embodiments, attachment of such conjugate groups to an oligonucleotide is desirable. Such conjugate groups may be attached at the 5'-, and/or 3'- end of an oligonucleotide. In certain instances, attachment at the 5'-end is synthetically desireable.
Typically, oligonucleietides are synthesized by attachment of the 3' terminal nucleoside to a solid support and sequential coupling of nucleosides from 3' to 5' using techniques that are well known in the art.
Accordingly if a conjugate group is desred at the 3'-terminus, one may (1) attach the conjugate group to the 3'-terminal nucleoside and attach that conjugated nucleoside to the solid support for subsequent preparation of the oligonucleotide or (2) attach the conjugate group to the 3'-terminal nucleoside of a completed oligonucleotide after synthesis. Niether of these approaches is very efficient and thus both are costly. In particular, attachment of the conjugated nucleoside to the solid support, while demonstrated in the Examples herein, is an inefficient process. In certain embodiments, attaching a conjugate group to the 5'-terminal nucleoside is synthetically easier than attachment at the 3'-end. One may attach a non-conjugated 3' terminal nucleoside to the solid support and prepare the oligonucleotide using standard and well characterized reastions. One then needs only to attach a 5'nucleoside having a conjugate group at the final coupling step.
In certain embodiments, this is more efficient than attaching a conjugated nucleoside directly to the solid support as is typically done to prepare a 3'-conjugated oligonucleotide. The Examples herein demonstrate attachment at the 5'-end. In addition, certain conjugate groups have synthetic advantages. For Example, certain conjugate groups comprising phosphorus linkage groups are synthetically simpler and more efficiently prepared than other conjugate groups, including conjugate groups reported previously (e.g., WO/2012/037254).
In certain embodiments, conjugated antisense compounds are administered to a subject. In such embodiments, antisense compounds comprsing one or more conjugae group described herein has increased activity and/or potency and/or tolerability compared to a parent antisense compound lacking such one or more conjugate group. Without being bound by mechanism, it is believed that the conjugate group helps with distribution, delivery, and/or uptake into a target cell or tissue. In certain embodiments, once inside the target cell or tissue, it is desirable that all or part of the conjugate group to be cleaved to releas the active oligonucleitde. In certain embodiments, it is not necessary that the entire conjugate group be cleaved from the oligonucleotide. For example, in 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). Thisconjugated 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 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 manmialian 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 Target Nucleic Acids Target Species GENBANK Accession Number SEQ ID
NO
HBV Human U95551.1 1 Transthyretin (TTR) Human NM 000371.3 2 The targeting process usually includes determination of at least one target region, segment, or site within the target nucleic acid for the antisense interaction to occur such that the desired effect will result.

In certain embodiments, a target region is a structurally defined region of the nucleic acid. For example, in certain such embodiments, a target region may encompass a 3' UTR, a 5' UTR, an exon, an intron, a coding region, a translation initiation region, translation termination region, or other defined nucleic acid region or target segment.
In certain embodiments, a target segment is at least about an 8-nucleobase portion of a target region to which a conjugated antisense compound is targeted. Target segments can include DNA or RNA sequences that comprise at least 8 consecutive nucleobases from the 5'-terminus of one of the target segments (the remaining nucleobases being a consecutive stretch of the same DNA or RNA
beginning immediately upstream of the 5'-terminus of the target segment and continuing until the DNA
or RNA comprises about 8 to about 30 nucleobases). Target segments are also represented by DNA or RNA
sequences that comprise at least 8 consecutive nucleobases from the 3'-terminus of one of the target segments (the remaining nucleobases being a consecutive stretch of the same DNA or RNA beginning immediately downstream of the 3'-terminus of the target segment and continuing until the DNA or RNA
comprises about 8 to about 30 nucleobases). Target segments can also be represented by DNA or RNA sequences that comprise at least 8 consecutive nucleobases from an internal portion of the sequence of a target segment, and may extend in either or both directions until the conjugated antisense compound comprises about 8 to about 30 nucleobases.
In certain embodiments, antisense compounds targeted to 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 intemucleoside 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. Hepatitis B (HBV) Hepatitis B is a viral disease transmitted parenterally by contaminated material such as blood and blood products, contaminated needles, sexually and vertically from infected or carrier mothers to their offspring. It is estimated by the World Health Organization that more than 2 billion people have been infected worldwide, with about 4 million acute cases per year, 1 million deaths per year, and 350-400 million chronic carriers (World Health Organization: Geographic Prevalence of Hepatitis B
Prevalence, 2004.
http://www.who.int/vaccines-surveillance/graphics/htmls/hepbprev.htm).

The virus, HBV, is a double-stranded hepatotropic virus which infects only humans and non-human primates. Viral replication takes place predominantly in the liver and, to a lesser extent, in the kidneys, pancreas, bone marrow and spleen (Hepatitis B virus biology. Microbiol Mol Biol Rev. 64: 2000; 51-68.).
Viral and immune markers are detectable in blood and characteristic antigen-antibody patterns evolve over time. The first detectable viral marker is HBsAg, followed by hepatitis B e antigen (HBeAg) and HBV DNA.
Titers may be high during the incubation period, but HBV DNA and HBeAg levels begin to fall at the onset of illness and may be undetectable at the time of peak clinical illness (Hepatitis B virus infection¨natural history and clinical consequences. N Engl J Med.. 350: 2004; 1118-1129). HBeAg is a viral marker detectable in blood and correlates with active viral replication, and therefore high viral load and infectivity (Hepatitis B e antigen¨the dangerous end game of hepatitis B. N Engl J Med.
347: 2002; 208-210). The presence of anti-HBsAb and anti-HBcAb (IgG) indicates recovery and immunity in a previously infected individual.
Currently the recommended therapies for chronic HBV infection by the American Association for the Study of Liver Diseases (AASLD) and the European Association for the Study of the Liver (EASL) include interferon alpha (INFa), pegylated interferon alpha-2a (Peg-IFN2a), entecavir, and tenofovir. The nucleoside and nucleobase therapies, entecavir and tenofovir, are successful at reducing viral load, but the rates of HBeAg seroconversion and HBsAg loss are even lower than those obtained using IFNa therapy. Other similar therapies, including lamivudine (3TC), telbivudine (LdT), and adefovir are also used, but for nucleoside/nucleobase therapies in general, the emergence of resistance limits therapeutic efficacy.
Thus, there is a need in the art to discover and develop new anti-viral therapies. Additionally, there is a need for new anti-HBV therapies capable of increasing HBeAg and HBsAg seroconversion rates. Recent clinical research has found a correlation between seroconversion and reductions in HBeAg (Fried et al (2008) Hepatology 47:428) and reductions in HBsAg (Moucari et al (2009) Hepatology 49:1151). Reductions in antigen levels may have allowed immunological control of HBV infection because high levels of antigens are thought to induce immunological tolerance. Current nucleoside therapies for HBV are capable of dramatic reductions in serum levels of HBV but have little impact on HBeAg and HBsAg levels.
Antisense compounds targeting HBV have been previously disclosed in W02011/047312, W02012/145674, and W02012/145697, each herein incorporated by reference in its entirety. Clinical studies are planned to assess the effect of antisense compounds targeting HBV 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 HB V Nucleic Acid In certain embodiments, conjugated antisense compounds are targeted to a HBV
nucleic acid having the sequence of GENBANKO Accession No. U95551.1, incorporated herein as SEQ ID
NO: 1. 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 NO: 1.
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: 3. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 3.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
1 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 4. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 4.
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: 5. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 5.
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: 6. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 6.
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: 7. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 7.
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: 8. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 8.
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: 9. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 9.
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: 10. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 10.
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: 11. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 1 comprises a nucleobase sequence of SEQ ID
NO: 11.

Table 2: Antisense Compounds targeted to HBV SEQ ID NO: 1 Target SEQ ID
ISIS No Start Sequence (5'-3') Motif NO
Site 505358 1583 GCAGAGGTGAAGCGAAGTGC eeeeeddddddddddeeeee 3 eeeeeddddddddddeeeee 4 510100 411 GGCATAGCAGCAGGATG eeeddddddddddeeee 5 eeeeeeddddddddddeeee 6 552024 1577 GTGAAGCGAAGTGCACACGG eeeeeeddddddddddeeee 7 eeeeeeddddddddddeeee 8 552859 1583 AGGTGAAGCGAAGTGC ekkddddddddddkke 9 552925 1264 TCCGCAGTATGGATCG ekddddddddddkeke 10 577119 1780 AATTTATGCCTACAGCCT kdkdkddddddddeeeee 11 In certain embodiments, a compound comprises or consists of ISIS 505358 and a conjugate group.
ISIS 505358 is a modified oligonucleotide haying the formula: Ges mCes Aes Ges Aes Gds Gds Tds Gds Ads Ads Gds mCds Gds Ads Aes Ges Tes 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 or consists of ISIS 509934 and a conjugate group.
ISIS 509934 is a modified oligonucleotide haying the formula: mCes mCes Aes Aes Tes Tds Tds Ads Tds Gds mCds mCds Tds Ads mCds Aes Ges mCes mCes 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 510100 and a conjugate group.
ISIS 510100 is a modified oligonucleotide having the formula: Ges Ges mCes Ads Tds Ads Gds mCds Ads Gds mCds Ads Gds Ges Aes Tes Ge, 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 552023 and a conjugate group.
ISIS 552023 is a modified oligonucleotide having the formula: Aes Ges Ges Aes Ges Tes Tds mCds mCds Gds mCds Ads Gds Tds Ads Tds Ges Ges Aes 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 552024 and a conjugate group.
ISIS 552024 is a modified oligonucleotide having the formula: Ges Tes Ges Aes Aes Ges mCds Gds Ads Ads Gds Tds Gds mCds Ads mCds Aes mCes Ges Ge, 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 552032 and a conjugate group.
ISIS 552032 is a modified oligonucleotide having the formula: Ges Tes Ges mCes Aes Ges Ads Gds Gds Tds Gds Ads Ads Gds mCds Gds Aes Aes Ges 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 552859 and a conjugate group.
ISIS 552859 is a modified oligonucleotide having the formula: Aes Gks Gks Tds Gds Ads Ads Gds mCds Gds Ads Ads Gds Tks Gks mCe, wherein, A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, k = a cEt modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.

In certain embodiments, a compound comprises or consists of ISIS 552925 and a conjugate group.
ISIS 552925 is a modified oligonucleotide having the formula: Tes mCks mCds Gds mCds Ads Gds Tds Ads Tds Gds Gds Aks Tes mCks Ge, wherein, A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, k = a cEt modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound comprises or consists of ISIS 577119 and a conjugate group.
ISIS 577119 is a modified oligonucleotide having the formula: Aks Ads Tks Tds Tks Ads Tds Gds mCds mCds Tds Ads mCds Aes Ges mCes mCes Te, wherein, A = an adenine, mC = a 5'-methylcytosine G = a guanine, T = a thymine, e = a 2'-0-methoxyethyl modified nucleoside, k = a cEt modified nucleoside, d = a 2'-deoxynucleoside, and s = a phosphorothioate internucleoside linkage.
In certain embodiments, a compound having the following chemical structure comprises or consists of ISIS 505358 with a 5'-X, wherein X is a conjugate group as described herein:

N
)i Xj(11 0 NH2 ( N1AN
o ) (:) NO N N
O 0, J NH2 S-P=0 NH2 I

S-P=0 N G 0 N1AN
/N 0 1 hi S-P=0 I _j N 1\r NI-12 e 0 (2I) NH2 NH2 0 - -1 7/) , I) e 1 0 S-P=0 NIA N 0 , N1/LN 0 0 I ) S-P=0 S-P=0 (5/N N
N I yLNH
e,, I *I, CL)/ N NH2 0 ' S-P=0 N e S-P=0 N1A
0õ) oI hi I I A

N N' NH2 \s`v_ I C)J
e S-P=0 O
\ A-it'yH
N--.0 c04/
e =o " ey(NH
S-P=0 N1AN
i I I *I, 0)c_oj" e N NH2 0õ) 0 e , N
0 0 NH2 S_=0 1 1-11'NH
O ()-1),) 0 e 1 exit-NH S- k N N NH2 P=0 N 0 1 S-P=0 I
(L/
6, \ 1 0 c(5/
CcLVN N NH2 0 0õ) o o e Y N S-P=0 O 1 S-p=0 1-1-NH
e NH
I
ILN
S-P=0 N 1 0 I

o<yl'NH c) ccjN N NH2 \ 1 CcLy c_OjN N NH2 e e 1 0 S-P=0 i S-P=0 0 ____________ i 0 ______________________________ In certain embodiments, a compound comprises or consists of ISIS 712408 having the following chemical structure:

,0 NH2 Y N
HO OH f_y: Nx),N.N
HO_...,r2..\, H1\14<\ c, 0 N N NH2 1 0---ty-NA-- N N
4 I-1 0 --v_io .....rNH N 0-' ______________________________________________ Ic_5/
HO OH 0 N. Io e 91 (!)...) NH2 o s-ol'=-.5/ N NH2 e 9 _..T.2.\õ, ..11.,_,õ0.,_õ.--NH S-P1=0 N (õN/111:411H
HO 0 -**--1-ri. 11 0 I
Ni.
0" ) /N 0 NH
--k-1 0--- NH2 HO OH NH 0 .
0 0õ) 2 N
_.,...r2...\.., 0 1 S-p=0 1 0 __.L.
HO 01-1:Thil 0 S-p=0 NiA,..N 0 N

0 e 9 N/ANH

(.7) 0- N)O S-p=0 . 0 S-P=0 :r 0 e 9 NIAN.N
0 S-p=0 1 9 c),) NH2 0 N
5_0 NI-A.,,N N

N CLy N N

0 e 9 NIAN.N

0 9 o,..1 S-p=0 N N
N 0...., /
0i 0 S-F,'=0 0 <'' :LAXNH2 -...w ....=
N N
S2OP' N NNI--11:- NIEINH2 e 9 N I
S1=0 <,:el:rjH
O.,,_,,,, p (D
0 0 o) 0 e 99 .
5-P=0 """NH
S-p=0 AA.YH

ON N'.-..-0 ci/,,y e9 N111 e 9 Ni 0 S-p=0 1-1 -0 N N NH2 S-p=0 1jj'NH

''''.': H/ l NH2 i_Oi/

e 9 Ni-k-N ....) s-F.'=o I 9 OW N S-p=0 iik'N

.."
9 0"--S-P=0 OH 0,) 0 ______________________________________________________ In certain embodiments, a compound comprises or consists of ISIS 695324 having the following chemical structure:

Y N
HO OH y(,,,,NH Nx-LN
H1\14<\ c, 0 2 I
N N
N N
4 H 0 'V_10 IcLy ....rNH
HO OH 0 N. Io 0 91 (!)...) NH2 o s-o'''=-..5/ N NH2 NH
e 9 S-P1=0 N <,NIIINII1H
HO 0---/n1 Ni.
OV /1\1 0 --k-1 0"-- NH2 HO OH NH 0 .
0 0,) 2 0 N
_.......4., 0 ' S-=0 HO 01-r:Thil 0 0-1=0 NI-A,. N 0 - N 0 N

W
0 e 9 ,NNH
I
(.7) 0- ON) S-1=0 . 0 P= 0 111:11H

0\WN N NH2 NH2 o' S-p=0 N1AN

o c),) NH2 0 N
90-p=o Ni1.-.N N
, I
N cLy N N

(Y e 9 Nx-LN

0 9 o,..1 S-1=0 N N
N 0...,0i/ 0 Ot=0 0 <,, litr ,,wNH2 ..--N N
C)- 0P' =w NNI.11: NIEINH2 e 9 Nxt 1 S1=0 <, rim o...,._)/1\1 NO 0 ()) 0 e 9o .
O-P=0 'NH
S-=0 AA.YH
0 i(L1 ON N--..-0 NO
(: 0 cLci/(y e9 N 0 S-1=0 ' x-u...x e 9 NINH
A
0 N N NH2 S-p=0 I

NH2 i_Oi/ c) 9NIA-N .....) NH2 S-1=0 I 9 0---5/N N S-1'=0 1LN

Cr-S+0 OH 0,..-I
0 ______________________________________________________ In certain embodiments, a compound comprises or consists of SEQ ID NO: 3, 5'-GalNAc, and chemical modifications as represented by the following chemical structure:

O
ne NH2 Y N
HO OH fX
HO--**T2--VOrN
4 H'It' 0 HN4<\
HO H O o 2 1 N N
NH
NO N NH
N I 14211õ R 1 0 0 c) S-P=0 R5 NH2 S-p=0 2e1:Y1:1 HO rN

-...liNH OV O
_0_yN 0 0 HO OH R NH2 9 9 R5 , y Ri __....rf....\., S-p=0 'ell Z-P=0 NIA.,N
o N 0 ,1rNH Oli:4/N N

R2 e 9 NYLNH
I
9 Ri os-F.,=o Z-=0 0 p, 1t IV
eN le (Xi 0 y R3 NH2 s-=o Na*N

z-7=o N3c),,N 6 I
1W_o4/
N N

)c_04/ R3 R4 0 o NIA,N
I
y R3 S-p=0 o--ic N N
Z-p=0 N:CI(NH
I
o 1447r 9 o (DR'c ? R3 o Z- P=0 I <N2I'll:r S-p=0 NIANH
Olco_;/ 9 224/N N NH2 N N NH

O

S-F'= R1, Z-P=0 R5,(11.
NH
, NH 0 I
0 I ,.

S-P=O
o 9 N2L)I'NH
I Z-1=0 <N2eX

)c2j 0 N.),,, S-p=o <2IN

R5,NH..2 avrIL)/N N S-p=0 1 I
ok.c.//N 0 S+0 OH R' O ______________________________________________________ wherein either R1 is ¨OCH2CH2OCH3 (M0E)and R2 is H; or R1 and R2 together form a bridge, wherein R1 is ¨0- and R2 is ¨CH2-, -CH(CH3)-, or -CH2CH2-, and R1 and R2 are directly connected such that the resulting bridge is selected from: -0-CH2-, -0-CH(CH3)-, and ¨0-CH2CH2-;
and for each pair of R3 and R4 on the same ring, independently for each ring:
either R3 is selected from H and -OCH2CH2OCH3 and R4 is H; or R3 and R4 together form a bridge, wherein R3 is ¨0-, and R4 is ¨CH2-, -CH(CH3)-, or -CH2CH2-and R3 and R4 are directly connected such that the resulting bridge is selected from: -0-CH2-, -0-CH(CH3)-, and ¨0-CH2CH2-;
and R5 is selected from H and ¨CH3;

and Z is selected from S- and 0-.
In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in WO
2012/145697, 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 5-310, 321-802, 804-1272, 1288-1350, 1364-1372, 1375, 1376, and 1379 disclosed in WO 2012/145697 and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in WO 2011/ 047312, 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 14-22 disclosed in WO
2011/ 047312 and a conjugate group described herein. In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in WO 2012/145674, 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 18-35 disclosed in WO 2012/145674.
In certain embodiments, a compound comprises a double-stranded oligonucleotide disclosed in WO
2013/159109, which is incorporated by reference in its entirety herein, and a conjugate group described herein. In certain embodiments, a compound comprises a double-stranded oligonucleotide in which one strand has a nucleobase sequence of any of SEQ ID NOs 30-125 disclosed in WO
2013/159109. The nucleobase sequences of all of the aforementioned referenced SEQ ID NOs are incorporated by reference herein.
HB V Therapeutic Indications In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to a HBV nucleic acid for modulating the expression of HBV in a subject. In certain embodiments, the expression of HBV is reduced.
In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to a HBV nucleic acid in a pharmaceutical composition for treating a subject. In certain embodiments, the subject has a HBV-related condition. In certain embodiments, the HBV-related condition includes, but is not limited to, chronic HBV infection, inflammation, fibrosis, cirrhosis, liver cancer, serum hepatitis, jaundice, liver cancer, liver inflammation, liver fibrosis, liver cirrhosis, liver failure, diffuse hepatocellular inflammatory disease, hemophagocytic syndrome, serum hepatitis, and HBV viremia. In certain embodiments, the HBV-related condition may have symptoms which may include any or all of the following: flu-like illness, weakness, aches, headache, fever, loss of appetite, diarrhea, jaundice, nausea and vomiting, pain over the liver area of the body, clay- or grey-colored stool, itching all over, and dark-colored urine, when coupled with a positive test for presence of a hepatitis B virus, a hepatitis B viral antigen, or a positive test for the presence of an antibody specific for a hepatitis B viral antigen. In certain embodiments, the subject is at risk for an HBV-related condition. This includes subjects having one or more risk factors for developing an HBV-related condition, including sexual exposure to an individual infected with Hepatitis B
virus, living in the same house as an individual with a lifelong hepatitis B
virus infection, exposure to human blood infected with the hepatitis B virus, injection of illicit drugs, being a person who has hemophilia, and visiting an area where hepatitis B is common. In certain embodiments, the subject has been identified as in need of treatment for an HBV-related condition.
Certain embodiments provide a method of reducing HBV DNA and/or HBV antigen levels in a animal infected with HBV comprising administering to the animal a conjugated antisense compound targeted to a HBV nucleic acid. In certain embodiments, the antigen is HBsAG or HBeAG.
In certain embodiments, the amount of HBV antigen may be sufficiently reduced to result in seroconversion.
In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to a HBV nucleic acid in the preparation of a medicament.
In certain embodiments, the invention provides a conjugated antisense compound targeted to a HBV
nucleic acid, or a pharmaceutically acceptable salt thereof, for use in therapy.
Certain embodiments provide a conjugated antisense compound targeted to a HBV
nucleic acid for use in the treatment of a HBV-related condition. The HBV-related condition includes, but is not limited to, chronic HBV infection, inflammation, fibrosis, cirrhosis, liver cancer, serum hepatitis, jaundice, liver cancer, liver inflammation, liver fibrosis, liver cirrhosis, liver failure, diffuse hepatocellular inflammatory disease, hemophagocytic syndrome, serum hepatitis, and HBV viremia.
Certain embodiments provide a conjugated antisense compound targeted to a HBV
nucleic acid for use in reducing HBV DNA and/or HBV antigen levels in a animal infected with HBV comprising administering to the animal a conjugated antisense compound targeted to a HBV
nucleic acid. In certain embodiments, the antigen is HBsAG or HBeAG. In certain embodiments, the amount of HBV antigen may be sufficiently reduced to result in seroconversion.
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 HBV
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: 3-11; a conjugated antisense compound targeted to SEQ ID NO: 1 comprising a nucleobase sequence of any of SEQ ID NOs: 3-11; a compound comprising or consisting of ISIS 505358, ISIS 509934, ISIS 510100, ISIS 552023, ISIS 552024, ISIS 552032, ISIS 552859, ISIS 552925, or ISIS 577119 and a conjugate group; a compound comprising an antisense oligonucleotide disclosed in WO 2012/145697, 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 5-310, 321-802, 804-1272, 1288-1350, 1364-1372, 1375, 1376, and 1379 disclosed in WO 2012/145697 and a conjugate group described herein; a compound comprising an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID
NOs 14-22 disclosed in WO 2011/ 047312 and a conjugate group described herein;
a compound comprising an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 18-35 disclosed in WO
2012/145674; or a compound comprising a double-stranded oligonucleotide in which one strand has a nucleobase sequence of any of SEQ ID NOs 30-125 disclosed in WO 2013/159109.
2. Transthyretin (TTR) TTR (also known as prealbumin, hyperthytoxinemia, dysprealbuminemic, thyroxine; senile systemic amyloidosis, amyloid polyneuropathy, amyloidosis I, PALB;
dystransthyretinemic, HST2651; TBPA;
dysprealbuminemic euthyroidal hyperthyroxinemia) is a serum/plasma and cerebrospinal fluid protein responsible for the transport of thyroxine and retinol (Sakaki et al, Mol Biol Med. 1989, 6:161-8).
Structurally, TTR is a homotetramer; point mutations and misfolding of the protein leads to deposition of amyloid fibrils and is associated with disorders, such as senile systemic amyloidosis (SSA), familial amyloid polyneuropathy (FAP), and familial amyloid cardiopathy (FAC).
TTR is synthesized primarily by the liver and the choroid plexus of the brain and, to a lesser degree, by the retina in humans (Palha, Clin Chem Lab Med, 2002, 40, 1292-1300).
Transthyretin that is synthesized in the liver is secreted into the blood, whereas transthyretin originating in the choroid plexus is destined for the CSF. In the choroid plexus, transthyretin synthesis represents about 20%
of total local protein synthesis and as much as 25% of the total CSF protein (Dickson et al., J Biol Chem, 1986, 261, 3475-3478).
With the availability of genetic and immunohistochemical diagnostic tests, patients with TTR
amyloidosis have been found in many nations worldwide. Recent studies indicate that TTR amyloidosis is not a rare endemic disease as previously thought, and may affect as much as 25% of the elderly population (Tanskanen et al, Ann Med. 2008;40(3):232-9).
At the biochemical level, TTR was identified as the major protein component in the amyloid deposits of FAP patients (Costa et al, Proc. Natl. Acad. Sci. USA 1978, 75:4499-4503) and later, a substitution of methionine for valine at position 30 of the protein was found to be the most common molecular defect causing the disease (Saraiva et al, i Clin. Invest. 1984, 74: 104-119). In FAP, widespread systemic extracellular deposition of TTR aggregates and amyloid fibrils occurs throughout the connective tissue, particularly in the peripheral nervous system (Sousa and Saraiva, Prog.
Neurobiol. 2003, 71: 385-400).
Following TTR deposition, axonal degeneration occurs, starting in the unmyelinated and myelinated fibers of low diameter, and ultimately leading to neuronal loss at ganglionic sites.
Antisense compounds targeting TTR have been previously disclosed in US2005/0244869, W02010/017509, and W02011/139917, each herein incorporated by reference in its entirety. An antisense oligonucleobase targeting TTR, ISIS-TTR, is currently in Phase 2/3 clinical trials to study its effectiveness in treating subjects with Familial Amyloid Polyneuropathy. However, there is still a need to provide patients with additional and more potent treatment options.
Certain Conjugated Antisense Compounds Targeted to a TTR Nucleic Acid In certain embodiments, conjugated antisense compounds are targeted to a TTR
nucleic acid having the sequence of GENBANKO Accession No. NM_000371.3, incorporated herein as SEQ
ID NO: 2. 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 SEQ ID NO:
2 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 12. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 2 comprises a nucleobase sequence of SEQ ID
NO: 12.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
2 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 13. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 2 comprises a nucleobase sequence of SEQ ID
NO: 13.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
2 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 14. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 2 comprises a nucleobase sequence of SEQ ID
NO: 14.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
2 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 15. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 2 comprises a nucleobase sequence of SEQ ID
NO: 15.

In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
16 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: 16 comprises a nucleobase sequence of SEQ ID NO: 78.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
2 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 17. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 2 comprises a nucleobase sequence of SEQ ID
NO: 17.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
2 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 18. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 2 comprises a nucleobase sequence of SEQ ID
NO: 18.
In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO:
2 comprises an at least 8 consecutive nucleobase sequence of SEQ ID NO: 19. In certain embodiments, a conjugated antisense compound targeted to SEQ ID NO: 2 comprises a nucleobase sequence of SEQ ID
NO: 19.
Table 3: Antisense Compounds targeted to TTR SEQ ID NO: 2 Target Start ISIS No Sequence (5'-3') Motif SEQ ID NO
Site 420915 508 TCTTGGTTACATGAAATCCC eeeeeddddddddddeeeee 12 304299 507 CTTGGTTACATGAAATCCCA eeeeeddddddddddeeeee 13 420921 515 GGAATACTCTTGGTTACATG eeeeeddddddddddeeeee 14 420922 516 TGGAATACTCTTGGTTACAT eeeeeddddddddddeeeee 15 420950 580 TTTTATTGTCTCTGCCTGGA eeeeeddddddddddeeeee 16 420955 585 GAATGTTTTATTGTCTCTGC eeeeeddddddddddeeeee 17 420957 587 AGGAATGTTTTATTGTCTCT eeeeeddddddddddeeeee 18 420959 589 ACAGGAATGTTTTATTGTCT eeeeeddddddddddeeeee 19 In certain embodiments, a compound comprises or consists of ISIS 420915 and a conjugate group.
ISIS 420915 is a modified oligonucleotide having the formula: Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ads Tds Gds Ads Ads Aes Tes 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 304299 and a conjugate group.
ISIS 304299 is a modified oligonucleotide having the formula: mCes Tes Tes Ges Ges Tds Tds Ads mCds Ads Tds Gds Ads Ads Ads Tes mCes 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 420921 and a conjugate group.
ISIS 420921 is a modified oligonucleotide having the formula: Ges Ges Aes Aes Tes Ads mCds Tds mCds Tds Tds Gds Gds Tds Tds Aes mCes Aes Tes Ge, 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 420922 and a conjugate group.
ISIS 420922 is a modified oligonucleotide having the formula: Tes Ges Ges Aes Aes Tds Ads mCds Tds mCds Tds Tds Gds Gds Tds Tes Aes mCes Aes 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 420950 and a conjugate group.
ISIS 420950 is a modified oligonucleotide having the formula: Tes Tes Tes Tes Aes Tds Tds Gds Tds mCds Tds mCds Tds Gds mCds mCes Tes Ges Ges 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 420955 and a conjugate group.
ISIS 420955 is a modified oligonucleotide having the formula: Ges Aes Aes Tes Ges Tds Tds Tds Tds Ads Tds Tds Gds Tds mCds Tes mCes Tes 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 or consists of ISIS 420957 and a conjugate group.
ISIS 420957 is a modified oligonucleotide having the formula: Aes Ges Ges Aes Aes Tds Gds Tds Tds Tds Tds Ads Tds Tds Gds Tes mCes Tes mCes 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 420959 and a conjugate group.
ISIS 420959 is a modified oligonucleotide having the formula: Aes mCes Aes Ges Ges Ads Ads Tds Gds Tds Tds Tds Tds Ads Tds Tes Ges Tes mCes 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 having the following chemical structure comprises or consists of ISIS
420915 with a 5'-X, wherein X is a conjugate group as described herein:

'llu:NZ 0 NH
= N 0 "Irui!NZ N-.1,,N
1 ,J
...._ N 0 N N

O y 0,......) NH2 (c/ cf-eM/X
S-P.0 N.õ....../, NH2 0 Ni 0 S-P=0 N2eN
.:. e 9 I I S -P=0 N N

I N#j ....-=

O ON,) 0 NH2 6a'' S-p=0 \ )N
111-1 0 ' N o '''' e I -S -P=0 C\L/N 0 I
...-- 0,,...Nb y 0,) 0 ' 0 S -P=0 NI...),":-N
I 'IN X i 0õ) 1 ,J

O\ O\ N
e , ....." 0 0 tNb 0 0 \
0,) S-P
e I e , NH
S-p=0 =0 N
-f-fe/O
...--I X-jj***X 0N'0 NwN N NH2 N
0) NH2 SI:f-/ e , , S-P=0 "N

6,,õ. NO
O 9 oõ) o e i N
S-POL;yfll'N:ZINH2 S-P=0 N
6, 1-"1-1-1 N, 0"---NH2 0,,) O 0 e 9 N2e.:-N e , S -P=0 O , s-F.,=o N I ,j S-P=0 (5 1 -IritX N
= N, N 0 0 ...--e , 0P=0 e , S-, S-P=0 0 ___________ I
0 _______________________________ In certain embodiments, a compound comprises or consists of ISIS 682877 having the following chemical structure:

HO OH 0 04)=0 '11)(Nr NIAN
HO,OrN) HNRC, 0 N'-'0 N I
N

iff2,/
.1.NH
NO I --Y¨Y 0"--HO OH 0 N oeyoõ) NH2 S-P=0)1 s c) s(*N
S-0 I'll 0,.._227/N--'0 NN

HO OHO a Y N
' S-1=0 .(11)1:y1 HO :1 O C)N"-4-jo S-1=0 4 H --e-Nr 0 NN NH2 W
.1iNH

o <NJJ
, c, 0õ) 0 S-1=0 I
N N

A)1211:1 Nssii.:22e/N 0 NH2 .....- a Y
o sl,=0 I
O o,) o )N N
eS-P=0 N

....)224/..-Nõ... N NH2 e Y N1).....,N
8 y oõ) o sl,o 1 N N
S-1=0 <,,,N, 1-111;:r 0.......yi(Ly0 0,.., ...-, -P=0 a c) 1 'ILI:r s-1,=0 NH o N ...-0,...., Ni-j.0 0,) N

O 8 , S-P=0 'IAN

N. NO
(cL'l0_04/
...-NH2 0,) NH2 a Y
sl,=0 NIAN
Y
sl,o o ,v,_/N
o NH2 , e?
IN 0õ) NH2 S-1=0 1 ,..L e Y
O-W 0 S-1=0 '`C-LN
0..../IN,....L0 0 y o' s-1;,=o OH 0õ) 0 _______________________________________________________ In certain embodiments, a compound comprises or consists of ISIS 682884 having the following chemical structure:

, 0 NH2 HO OH 0 04=0 'Ne(Nr NI-LN
HO,0(-rN
4 1-ril'' 0 HNRC,,:, 0 0 N'-'0 I
N N
,...NH
NO 0"--HO OH 0 N 0 8 9 ("") "2 0 HO__.,..,rõ2..\,' ,0--trN )__, 4 H 0O.¨NHPS- =0 I
0 _NT*N
I S-F0 '11)11 0-..._227/N--'0 ....IrNH
OV 1_0?/N 0 HO OH 0 0,) 0 e 9 N
--\-L:Ar 0 O''-l-rN"-4-jo 0-1=0 HO 4 H -,e-Nr 0 NN NH2 W
.1iNH

o9 NI-LN
e 9 oõ) o 5-1;,=0 I
N N

A)1211:1 Nsii.:22e/N 0 NH2 .....- e 9 o sl,=o I
9 o,) o (:),3//N N
80-P=0 N
1 <õ, 1A:

......)224/,N Nr NH2 0,....1 e? N1).....,N
8 9 oõ) o sl,=o I
N N
0I'=0 O
111:_r 0...vi2i,,,/, 0,.., ...-':LI0 0 0 e0-1=0 e 9 'ILI:r 5-1,=o A-ANH 0 Nylx/N 0 0-......, 0,) NH2 e 98 , O-P=0 --TLN
S-=0 -Nel'NH I _L

N. NO

NH2 0,) sl,=o NI-LN

0 .
S-F,,=0 eN, 0 N N 0 N.--.0 I

V1-2-Y0 .....-N 0õ) NH2 sl.=0 I .õ.L e c?
O-W 0 S-1=0 o' s-1;,=o OH
0õ) 0 _______ In certain embodiments, a compound comprises or consists of SEQ ID NO: 12, 5'-GalNAc, and chemical modifications as represented by the following chemical structure:

O
98 R5'NH NH2 HO OH I 1 NIA,N
_.,..,rf..õ,) 0 --m--N HN Rcõ, 0 --.'N 0 I

,1rNH No 1421¨f 0 HO OH 0 0 N 0 e Y Ri NH2 Y_ ,NH
...11.,_õ0,,,--N H S-P=0 R5 o R3 ,,,L
HO_ oI ' N S-p ( -0 1 ..,..4,0--trN

NH ON-sb ....Ir OV )c4/1 0 -1c_O_V
2? Ri HO OH e Y N
__.....rØ.\" S-p=O <, :LAX
HO 01-rN 0 4 H Z-C'=C) RV(NH N.b/N N NH2 NH N
0 R(' k_04/- -..' NH2 o Y NIA,N
y Fl 0 S-1=0 I
Z-P=0 RV'NH 0 . 0.,:r4N N
O: 1 _04/NO NH2 e (i) R-R 0 SI,=0 NIA.,N
y R3 N 6 I
N
Z-p=0 2(11:r N NH2 0.õ.sk_iN N NH2 o Y N2e,N
I
N N
ZR-4?0=,0C) N
R3 ciN1151'ZI s1-0 N H2 0-... 0 R4 c0 Y R5,()( _ e Y R-, Ferk Z-p=0 Sp0 1 WI
N,L-= 1 NH
o'Fic(L,N-.0 olt_04/

Y N
eY R3 RVLNH Z-P=0 R3,N
S -p=0 I t NO
NH2 c5/
ON Nrs'0 0 I.
e Y N1)...,õN R1 R5....
S=0 I y 1 1 N
0 N N Z-p=0 I

o? R3 ,AN
R2 Ri NH2 e C? IR3, 0-vitz/N- '0 5-1=0 1 y ok_04/---0 o Y
OH RI
0 _______________________________________________________ wherein either R1 is ¨OCH2CH2OCH3 (M0E)and R2 is H; or R1 and R2 together form a bridge, wherein R1 is ¨0- and R2 is ¨CH2-, -CH(CH3)-, or -CH2CH2-, and R1 and R2 are directly connected such that the resulting bridge is selected from: -0-CH2-, -0-CH(CH3)-, and ¨0-CH2CH2-;
and for each pair of R3 and R4 on the same ring, independently for each ring:
either R3 is selected from H and -OCH2CH2OCH3 and R4 is H; or R3 and R4 together form a bridge, wherein R3 is ¨0-, and R4 is ¨CH2-, -CH(CH3)-, or -CH2CH2-and R3 and R4 are directly connected such that the resulting bridge is selected from: -0-CH2-, -0-CH(CH3)-, and ¨0-CH2CH2-;
and R5 is selected from H and ¨CH3;

and Z is selected from 5- and 0-.
In certain embodiments, a compound comprises an antisense oligonucleotide disclosed in WO
2011/139917 or US 8,101,743, 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 8-160, 170-177 disclosed in WO 2011/139917 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 12-89 disclosed in US 8,101,743 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 90-133 disclosed in US 8,101,743 and a conjugate group described herein. The nucleobase sequences of all of the aforementioned referenced SEQ ID NOs are incorporated by reference herein.
TTR Therapeutic Indications In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to a TTR nucleic acid for modulating the expression of TTR in a subject. In certain embodiments, the expression of TTR is reduced.
In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to a TTR nucleic acid in a pharmaceutical composition for treating a subject. In certain embodiments, the subject has a transthyretin related disease, disorder or condition, or symptom thereof. In certain embodiments, the transthyretin related disease, disorder or condition is transthyretin amyloidosis.
"Transthyretin-related amyloidosis" or "transthyretin amyloidosis" or "Transthyretin amyloid disease", as used herein, is any pathology or disease associated with dysfunction or dysregulation of transthyretin that result in formation of transthyretin-containing amyloid fibrils. Transthyretin amyloidosis includes, but is not limited to, hereditary TTR amyloidosis, leptomeningeal amyloidosis, familial amyloid polyneuropathy (FAP), familial amyloid cardiomyopathy, familial oculoleptomeningeal amyloidosis, senile cardiac amyloidosis, or senile systemic amyloidosis.
In certain embodiments, the invention provides methods for using a conjugated antisense compound targeted to a TTR nucleic acid in the preparation of a medicament.
In certain embodiments, the invention provides a conjugated antisense compound targeted to a TTR
nucleic acid, or a pharmaceutically acceptable salt thereof, for use in therapy.
Certain embodiments provide a conjugated antisense compound targeted to a TTR
nucleic acid for use in the treatment of a transthyretin related disease, disorder or condition, or symptom thereof. In certain embodiments, the transthyretin related disease, disorder or condition is transthyretin amyloidosis.

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 TTR
nucleic acid in the aforementioned methods and uses can include, but is not limited to, a conjugated antisense compound targeted to SEQ ID NO: 2 comprising an at least 8 consecutive nucleobase sequence of any one of SEQ ID NOs: 12-19; a conjugated antisense compound targeted to SEQ ID NO: 2 comprising a nucleobase sequence of any one of SEQ ID NO: 12-19; a compound comprising or consisting of ISIS 420915, ISIS
304299, ISIS 420921, ISIS 420922, ISIS 420950, ISIS 420955, ISIS 420957, or ISIS 420959 and a conjugate group; a compound comprising an antisense oligonucleotide disclosed in WO
2011/139917 or US 8,101,743, 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 8-160, 170-177 disclosed in WO 2011/139917 and a conjugate group described herein; an antisense oligonucleotide having a nucleobase sequence of any of SEQ ID NOs 12-89 disclosed in US 8,101,743 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 90-133 disclosed in US 8,101,743 and a conjugate group described herein. The nucleobase sequences of all of the aforementioned 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.
Certain compounds, compositions, and methods herein are described as "comprising exactly" or "comprises exactly" a particular number of a particular element or feature.
Such descriptions are used to indicate that while the compound, composition, or method may comprise additional other elements, the number of the particular element or feature is the identified number. For example, "a conjugate comprising exactly one GalNAc" is a conjugate that contains one and only one GalNAc, though it may contain other elements in addition to the one GalNAc.
Although the sequence listing accompanying this filing identifies each sequence as either "RNA" or "DNA" as required, in reality, those sequences may be modified with any combination of chemical modifications. One of skill in the art will readily appreciate that such designation as "RNA" or "DNA" to describe modified oligonucleotides is, in certain instances, arbitrary. For example, an oligonucleotide comprising a nucleoside comprising a 2'-OH sugar moiety and a thymine base could be described as a DNA
having a modified sugar (2'-OH for the natural 2'-H of DNA) or as an RNA
having a modified base (thymine (methylated uracil) for natural uracil of RNA).
Accordingly, nucleic acid sequences provided herein, including, but not limited to those in the sequence listing, are intended to encompass nucleic acids containing any combination of natural or modified RNA and/or DNA, including, but not limited to such nucleic acids having modified nucleobases. By way of further example and without limitation, an oligonucleotide having the nucleobase sequence "ATCGATCG"
encompasses any oligonucleotides having such nucleobase sequence, whether modified or unmodified, including, but not limited to, such compounds comprising RNA bases, such as those having sequence "AUCGAUCG" and those having some DNA bases and some RNA bases such as "AUCGATCG" and oligonucleotides having other modified bases, such as "AT'CGAUCG," wherein 'C
indicates a cytosine base comprising a methyl group at the 5-position.
EXAMPLES
The following examples illustrate certain embodiments of the present disclosure and are not limiting.
Moreover, where specific embodiments are provided, the inventors have contemplated generic application of those specific embodiments. For example, disclosure of an oligonucleotide having a particular motif provides reasonable support for additional oligonucleotides having the same or similar motif. And, for example, where a particular high-affinity modification appears at a particular position, other high-affinity modifications at the same position are considered suitable, unless otherwise indicated.
Example 1: General Method for the Preparation of Phosphoramidites, Compounds 1, la and 2 BX
DMT0BX /***--c DMT0BX DMTO/46---c M e Ys NCP-N(iPr)2 NCP-N(iPr)2 NC0N(iPr)2 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 AcO0Ac )._ Ac0 TMSOTf, 50 ___....70...\0 0 HOCO * 5 AcO___2.....\ C
OAc ____________________________________________________________ )...
CICH2CH2CI N ---z..-1 TMSOTf, DCE
AcHN
3 (93%) 4 ( 66%) I
AcO0Ac AcO0Ac H2/Pd ..-.0H
Ac0 Ac0 Me0H
AcHN 0 AcHN 0 (95%) Compounds 3 (2-acetamido-1,3,4,6-tetra-0-acety1-2-deoxy-p-Dgalactopyranose 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 Et0,y,¨,1 NC7---1 0 0.,_ -..õ (:) Et0 HO.
CN 9 HCI, Et0H
HO.,õ---NH2)10.-NC---"\---0.....õ----NH2 aq. KOH, Reflux, rt, 0 Et0 0--HO" 1,4-dioxane, 0' (56%) 8 (40%) NC-I 10 d-----) 11 z Compounds 8 and 9 are commercially available.

Example 4: Preparation of Compound 18 Et0 Et0 )n 0 0 0õ. benzylchloroformate, 0 0, Dioxane, Na2CO3 Et0 ki--i( Li0H, H20 y-N,-0-........----11 0 _______________________________ 1. H Dioxane (86%) O Et0 0' Et0 0' (91%) ) ii o 12 >\.-01 H
II N7Nõ..-N
HO y.õ1 o , N 0 0,0......õ.._N
0 0, I 0 H H 9 ---------NNH2 14 --)---1-N --=HO H 0 sr.........._0...õ..._N--4,0 õI
HBTU, DIEA, DMF
" 0 (69%) 15 13 +ON N'7N H H - ¨ e 0 n AcO0Ac H
H2N\----\Ny..) Ac0 o .r -4-\r 0 w.i0H

H 0 ID, 1 AcHN 0 CF3COOH H2NN7N___N
----if---\--0...--N 0 401 HBTU, DIEA, HOBt ________ P H 0-95 % 0 0' DMF
H2N 16 (64%) V-N.N_____kj AcO0Ac _....TZrorN N
Ac0 AcHN 0 AcO0Ac 0 H H 0, N)(0 io H
AcHN 0 0 0' AcO0AcHN¨"kj Ac00----"---(1-\-11--/¨"/

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., 1 Med. Chem., 2004, 47, 5798-5808.

Example 5: Preparation of Compound 23 1.

1. TBDMSCI H
HBTU, DIEA
b TBDMSO N
N DMF, Imidazode, rt (95 %) DMF, rt (65%) HO---. ) _______________________ 0.
2. Pd/C, H2, Me0H, rt _ 2. TEA.3HF, TEA, THF )P
87% 20 0- TBDMS (72%) ''OH

"-b 1. DMTCI, pyr, rt (75%) --bi)OH
OCH 1). 3 ________________ ).
2. LION, Dioxane (97%) 23 OH
Compounds 19 and 21 are commercially available.
Example 6: Preparation of Compound 24 AcO0Ac H H
AGO_....r2,r0rN,--N,0 AcHN 0 1 H2, Pd/C, Me0H (93%) AcO0Ac 0 2. HBTU, DIEA, DMF (76%) () 0 0 ,--ODMT
H .
AcHN 0 0 (:) HO)N ' c 23 AcO0Ac HN---kj Ac00,...-r-kl---.7.---/

AcHN
AcO0Ac H H
Ac0_....,r(2..\v0rN.-1\1::) AcHN 0 ODMT
AcO0Ac H H0, 0 0 µ
_________________________________________ N--1.LqLN"
H \
AcHN 0 0 AcO0Ac H HN---kj Ac0 Ar-1N

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_04, 0i,õNN,C) AcHN 0 AcO0Ac ODMT

N 1. Succinic anhydride, DMAP, DCE
Ac0--72-\ror NN----N----n-----i---0-----. 11--1.LNQ ____________________ .
AcHN 0 0 0' OH 2. DMF, HBTU, EtN(iPr)2, PS-SS
AcO0Ac HN------µj H._./,/ 0 __01.2s\r____Ir--N
Ac0 o 0 24 AcHN
AcO0Ac H H
AcHN 0 ODMT
AcO0Ac 0 0 .,/.
0 p Ac0--72-\r yNNVN----N y-N.-- 0 AcHN 0 0 0' 04 AcO0Ac HN--j o---N
Ac0 0 25 AcHN
Compound 24 was prepared as per the procedures illustrated in Example 6.

Example 8: Preparation of Compound 26 AcO0Ac H H
AcOOr.N .,--N ,C) AcHN 0 AcO0Ac ODMT
H H 0, Ac0---'72-\'oNNV.N.--N-----rr-N.,-0-õ--- ri "1. NI\ /.
Phosphitylation AcHN 0 0 0' OH
AcO0Ac HN-----"J

_.....r.C.I..\raw---fr-I-N-1----7-----j Ac0 0 24 AcHN
AcO0Ac H H
Ac0OrN,..,,N,0 AcHN 0 0 DMT
AcO0Ac H H 0, Ac0--72--\vorNN7N--N----rN.,-0,,-- __________ ri--1q AcHN 0 0 0' 0 I
-NC C)' P N(iP02 AcO0Ac H HN----kj O
Ac0 __...!....\:),0õTi--N---/-----j Compound 24 is prepared as per the procedures illustrated in Example 6.

Example 9: General preparation of conjugated ASOs comprising GaINAc3-1 at the 3' terminus, Compound 29 AcO0Ac H H
Ac0 AcHN 0 ODMT
AcO0Ac tt H H 0, Ac0--4-\r =)-(NNVX-.--N----ii---\--0-õ,-- 11--IL(--eq ,¨NH
AcHN 0 0 0' 1. DCA, DCM
HN--j AcO0Ac H 0 2. DCI, NMI, ACN _____ Ac0_....,12..\r0----N"--7-----/ Phosphoramidite ' DNA/RNA ' 0 building block 1 sautomated synthesizer 25 ___________________________ , AcHN 3. Capping 4. t-BuO0HDMTO" 0' Bx y \( ) AcO0Ac _....r?...\r H H a OrN,,...N,0 1 /../CN
AGO 0=P1-0 AcHN 0 0 AcO0Ac 0 0 õ!
e Ac0-r ()-(NNVXN 0 y-N.-- ----- 1-(-- NH
AcHN 0 0 0' 04 i. DCA, DCM 0 . DCI NMI ACN
AcO0Ac H HN 0 Phosphoramidite ' DNA/RNA
' N---/---/
Ac0 C) building block la ,automated synthesizer, --../\/"----r 3. Capping 0 27 4. t-BuO0H
AcHN
DMTO-N(0),Bx (5, _____________________________________________________ b_/-0Me 0¨\(),13x AcO0Ac _.....r?..\., H H 0 rl\l,,...N ,D 1 Ac0 O 0=P-0-AcHN 0 o1 AcO0Ac H H 0, 0 0.,!
0 p AcO- 0 (NN N 0 ----R----N-- ------ ____________________________ ENIN,Q ,¨NH
AcHN 0 0 0' 04 1. DCA, DCM
AcO0Ac H HN-----kj 0 2. DCI, NMI, ACN
N---/---j Ac0 __....72.\/(:)----/\/---Tr Phosphoramidite ' DNA/RNA
0 28 building blocks automated synthesize AcHN 3. Capping 4. xanthane hydride or t-BuO0H
5. Et3N/CH3CN (1:1) 6. Aaueous NH, (cleavaael OH
I , s OLIGO

X=P\-0-0¨Ncyx ______________________________________________________ /
Bx = Heterocyclic base d b ¨OMe-f X=OorS I
0=P-0-\
0¨N(OBx HOOH
HO.__...r?..\., H H
OrN,.õ-N,0 a 0=P-0-HOOH AcHN 0 O
H H 0, 0 0 ..
HOoNNN.-----N-----ii----"\---0¨_--- N"ILHN"
H \
AcHN 0 0 0' OH
HOOH H HN-----kj N--.7---j __...72..\r(:)---...rr AcHN
Wherein the protected Ga1NAc3-1 has the structure:

s 9 0_Z/--4 ¨1=1)-0¨=\,,)N N
HOOH

H0.12..0i--N,-N,0 I
0=P-0 HOOH -AcHN 0 O
H H 0, HOT.,?...\v0rNNVN---N-----Ti----\--0,..---- N"I'LkNr H
AcHN 0 0 0' OH
HOOH H HN-----kj HO0---/Thri\l----7----/

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

HOOH

AcHN 0 HOOH
N-J.L(4LN
AcHN 0 0 OH
HOOH

AcHN
The solid support bound protected Ga1NAc3-1, Compound 25, was prepared as per the procedures illustrated in Example 7. Oligomeric Compound 29 comprising GaINAc3-1 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 Ga1NAc3-1 at the 5' terminus, Compound 34 ODMT 1. Capping (Ac20, NMI, pyr) I
1. DCA, DCM (OLIGO) 2. PADS or t-BuO0H
_____________________________ . I O¨UNL¨ODMT 2. DCI, NMI, ACN ___ 0 3. DCA, DCM ..-I 4. DCI, NMI, ACN
30 Phosphoramidite p..-0CN
Phosphoramidite 1 building blocks 01¨UNL-0¨
DNA/RNA

DNA/RNA NA/RNA
3 I ,automated synthesizer, ,automated synthesizer, DMT0(5"Bx 1. Capping (Ac20, NMI, pyr) 2. t-BuO0H 0' 3. DCA, DCM NC,. 1 O¨P
, ___________________________________________ 4. DCI, NMI, ACN (SI
Phosphoramidite 26 (OLIGO) DNA/RNA I
X = 0, or S µautomated synthesizer 0, I
Bx ¨ Heterocylic base 0¨UNL-0¨P-0CN

Ac0 OAc Ac0___ ..
.....s\r H H
AcHN 0 Ac0 OAc ODMT
N
NN_,N...ir..-N_ _________________________________ N---*(4'LN
Ac0 0 H 8 \
AcHN 0 0 C) 0 I
NC icy P.cy..,(0Bx Ac0 OAc FIN----C1 ___.....2..\r(:)----..Tr EN-I NC 1 Ac0 O¨P=0 .6 AcHN I
(OLIGO) I
1. Capping (Ac20, NMI, pyr) 0 2. t-BuO0H I
_ 3. Et3N:CH3CN (1:1 v/v) 0¨UNL-0¨p0CN
4. DCA, DCM K
5. NH4, rt (cleavage) 33 HOOH
H H
N,,,-NO
AcHN 0 HOOH OH
HOv0________ ____________________________ N.--ILHN, H \
AcHN 0 0 0' ?
O.õBx HOOH H HN----kj AcHN 34 I
(OLIGO) I
()H
The UnylinkerTm 30 is commercially available. Oligomeric Compound 34 comprising a Ga1NAe3-1 cluster at the 5' terminus is prepared using standard procedures in automated DNA/RNA synthesis (see Dupouy et al., Angew. Chem. Int. Ed., 2006, 45, 3623-3627). Phosphoramidite building blocks, Compounds 1 and la were prepared as per the procedures illustrated in Example 1. The phosphoramidites illustrated are meant to be representative and not intended to be limiting as other phosphoramidite building blocks can be used to prepare an oligomeric compound having a predetermined sequence and composition. The order and quantity of phosphoramidites added to the solid support can be adjusted to prepare gapped oligomeric compounds as described herein. Such gapped oligomeric compounds can have predetermined composition and base sequence as dictated by any given target.
Example 11: Preparation of Compound 39 AcO0Ac 1. HO p N 0 )L AcO0Ac H 4.0 Ac0 35 TMSOTf, DCE
.....r.(2.\r0 Ac0 NH2 ________________________________________ ).- 8 NI-- ---:...I 2. H2/Pd, Me0H AcHN 36 Ac0 OAc Ac0....i 1:)\ 1. H2, Pd/C, Me0H
HBTU, DMF, EtN(/P02 _______________ ).- oNNWNEd __________________________________ .-Compound 13 AcHN 8 2. HBTU, DIEA, DMF
Ac0 OAc H 0 0 Compound 23 Ac0 0 N (:) 0 .--Ed.o 0'114 '7-.'K..''' y"--NHAc 0 0 0 C) OAc Ac0 )\---) Ac01**=-\r AcHN

Ac0 OAc Ac0.7.2..\_,0 F /0DMTNi _ Phosphitylation AcHN OAc 8 0 % = i.-Ac0 H 0 0 , OH
AcO*2/ N YCH¨N H
NHAc 0 0 0 OAc )\--) Ac0 Ac0/12-\,C)NH
AcHN
Ac0 OAc Ac0 0 / ODMT
_ 0Ac AcHN 8 0 Ac0 H 0 0 AcOrCL\;) N (:),]¨NH 1 NC0'13N(iPr)2 NHAc 0 0\ CI) OAc )\------Ac0 Ac0=01"2-\,oNH 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. 1 Org. Chem., 2011, 12, 2346-2353.

Example 12: Preparation of Compound 40 Ac0 OAc Ac00 /0DMT
AcHN 8 0 O
H 0 0 _________________________________________ N
Ac0 Ac 0,..õ....../...isr,,...,.N 0 NH OH
Ac0 8 NHAc 0 0\ 10 1. Succinic anhydride, DMAP, DCE
OAc Ac0 38 2. DMF, HBTU, EtN(iPr)2, PS-SS
Ac0742-\,C)NH
AcHN
Ac0 OAc Ac0 ODMT
AcHN oF1\1 /
8 0 =
_ 0 "

OAc 0 )¨N
Ac0 H 0 C4 08 0----C)---11 AcOlill1/0 NHAc 0 0 0 OAc )\--) Ac0 Ac01112-\roNH
AcHN
Compound 38 is prepared as per the procedures illustrated in Example 11.
Example 13: Preparation of Compound 44 AcO0Ac HBTU, DMF, EtN(P02 _......T.C?_\,ONH2 ____________________________________ ).
Ac0 AcHN 36 HOO, )Lo 11 ¨N

HO\ /-0-- 41 Ac0 OAc Ac0---i lUO
N
AcHN C-N)11 0 0Fil 1. H2, Pd/C, Me0H
0 0 2. HBTU, DIEA, DMF
0)\___ j Compound 23 OAc 0 Ac0 .....r.I,0õ,,...õ---.H......õ...õ.NH
'ft Ac0 AcHN
Ac0 OAc Ac0 ODMT
H
=
AcHN 8 _ Phosphitylation j.

()IN 8 OH
H
0\\ ) 43 OAc /---' Ac0 Ac0 ,..r2....\roNH

AcHN
Ac0 OAc Ac0 C) ODMT
H
=
AcHN 8 -}N 8 H NC0,1D.N(iP02 0)\___ j Ac0.72..\,(31Ac 44 ONH
Ac0 AcHN
Compounds 23 and 36 are prepared as per the procedures illustrated in Examples 5 and 11.
Compound 41 is prepared using similar procedures published in WO 2009082607.

Example 14: Preparation of Compound 45 Ac0 OAc H
AcOu¨N4-)L,N,N ODMT
=
AcHN

IN OH
H
0\\ 3 43 OAc )L---/
Ac0 fliii?...\"0,... j...4......., NH 1. Succinic anhydride, DMAP, DCE
Ac0 8 _________________________________________________________________________ ).-AcHN 2. DMF, HBTU, EtN(iPr)2, PS-SS
Ac0 OAc H
AcOu-'\4')N ODMT
_ AcHN 8 0 ' 0 O)( 8 N ft 0)r C\1\ 3 OAc Ac0 Ac0.7.2..\/0 NH

AcHN
Compound 43 is prepared as per the procedures illustrated in Example 13.
Example 15: Preparation of Compound 47 HOb00 . DMTO
1. DMICI, pyr -sbIH
________________________________ ,.-2. Pd/C, H2, Me0H .-z 46 Hd 47 Hd Compound 46 is commercially available.

Example 16: Preparation of Compound 53 HBTU, EtN(iPr)2, DMF 0 YNNI-12 ____________________________ ).-H3CO-fril NBoc Boc HN

NH
0 \CBz 50 CBzõNH
HN,CBz 0 ), CBz N 1. Li0H, Me0H
H3C011N"....
1. TFA¨NH H
_________________ .- 0 ' H ______________________________________________ , 2. HBTU, EtN(iPr)2, DMF 2. HBTU, EtN(iPr)2, DMF

HN,CBz Compound 47 0)---\ ____ r-NICICBz HN,CBz OH

DMTO HN-CBz 0 ' 1. H2, Pd/C
0 ____________________________________________________________ *
CBz 2. HBTU, EtN(iPr)2, DMF
HO" \ N NH
N Compound 17 I'eN ______ r(' Fi FiN-CBz OAc OAc.L. 0 Ac0 NHAc OAc 0 OAc c. 11.,...,AliL.NT----..µµµOH

Ac0 0 HN
NHAc 0 0 --.----OAc ODMT
OAcL__ ---.7--(}"--(L
0 I __ 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____\....,\__ 0 Ac0 0 ).1_____ NHAc Ac0 OAc 0 OAc....\......\_ 0 i----...,%%0H

NHAc OAc --------/-----0\--tL.0 OAT( ODMT
I
AcO-V0 7) NH 53 ----\----\--0 NHAc Phosphitylation -OAc OAc.....\....\___ 0 Ac0 0 )1_..._ NHAc (iPr)2N,p-0 OAc Ac0 OAc.,...\..._\__ r---....,,,t0 CN

NHAc 0 HN HN ---"-A 1-7'' N

-------OAc OAT( -..-/--C}r ODMT

Ac0.---\-----\--0 NHAc Compound 53 is prepared as per the procedures illustrated in Example 16.

Example 18: Preparation of Compound 55 OAc Ac0 0 NHAc OAc Ac0 0 0 H N H N -Np N HAc 0 OAc ODMT

I _______________________________________ NH 53 N HAc 1. Succinic anhydride, DMAP, DCE
2. DMF, HBTU, EtN(iPr)2, PS-SS
OAc OA.

Ac0 0 NH
N HAc OAc O

Ac0 0 NHAc 0 OAKOAc ODMT
AcOO 0 I _______________________________ NH 55 NHAc Compound 53 is prepared as per the procedures illustrated in Example 16.
Example 19: General method for the preparation of conjugated ASOs comprising Ga1NAc3-1 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 mol scale) or on GE Healthcare Bioscience AKTA oligopilot synthesizer (40-200 mol scale) by the phosphoramidite coupling method on an GaINAc3-11oaded VIMAD solid support (110 molig, 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 um, 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, 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 GaINAc3-1 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, 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 GaINAc3-1 at its 3'-end.
Table 4 Modified ASO targeting ApoC III and SRB-1 SEQ
CalCd Observed ASO Sequence (5 to 3') Target ID
Mass Mass No.
ISISApoC
s s s s s s s AesGesmCesTesTesmCd Td Td Gd TasinCd InCd AdsGd InCds TesTesTesAesTe 7165.4 7164.4 20 ISIS AesGesinCesTesTesmCdsTdsTdsGdsTdsmCdsinCdsAdsGdsinCasTesTesTesAesTe.Ado,-ApoC
9239.5 9237.8 21 647535 Ga1NAc3-la ISIS
AesGeolliCeorreorreoinCdsTdsTdsGdsTdsinCdsinCdsAdsGdsinCdsTeerreeTesAesTeoAdo,-ApoC
9142.9 9140.8 21 647536 Ga1NAc3-la ISIS
TksmCksAdsGdsTasmCdsAdsT&GdsAcismCdsTasTksraCk 4647.0 4646.4 22 ISIS
TksmCksAdsGdsTasmCdsAdsT&GdsAdsmCdsTasTksmCk.Ado,-GaINAC3-la 6721.1 6719.4 23 Subscripts: "e" indicates 2'-MOE modified nucleoside; "d" indicates f3-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. "GaINAc3-1" 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 "Ga1NAc3-1a." 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 "GalNAc3 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 umol/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 HI 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 5, below.
As illustrated, both antisense compounds reduced ApoC III RNA relative to the PBS control.
Further, the antisense compound conjugated to Ga1NAe3-1 (ISIS 647535) was substantially more potent than the antisense compound lacking the Ga1NAe3-1 conjugate (ISIS 304801).
Table 5 Effect of ASO treatment on ApoC III mRNA levels in human ApoC III transgenic mice Dose ED50Internucleoside SEQ ID
ASO 3' Conjugate (.tmo1ikg) PBS (imolikg) linkage/Length No.

0.08 95 ISIS 0.75 42 0.77 None PS/20 20 304801 2.25 32 6.75 19 0.08 50 0.75 15 ISIS
647535 2.25 17 0.074 Ga1NAe3-1 PS/20 21 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 [t1 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 6 below, both antisense compounds reduced ApoC III
protein relative to the PBS control. Further, the antisense compound conjugated to Ga1NAe3-1 (ISIS
647535) was substantially more potent than the antisense compound lacking the Ga1NAe3-1 conjugate (ISIS
304801).
Table 6 Effect of ASO treatment on ApoC III plasma protein levels in human ApoC III
transgenic mice Dose ED50Internucleoside SEQ ID
ASO 3' Conjugate (.tmo1ikg) PBS (imolikg) 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 0.19 Ga1NAc3-1 PS/20 21 647535 2.25 12 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 7. 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 GaINAc3-1 conjugate (ISIS
304801).
Table 7 Effect of ASO treatment on triglyceride levels in transgenic mice ASO Dose ED50 3' Internucleoside SEQ ID
(imolikg) PBS (imolikg) 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 647535 2.25 8 0.13 Ga1NAc3-1 PS/20 21 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 8 and 9. As illustrated, both antisense compounds lowered total cholesterol levels; both lowered LDL; and both raised HDL. Further, the antisense compound conjugated to GaINAc3-1 (ISIS 647535) was substantially more potent than the antisense compound lacking the GaINAc3-1 conjugate (ISIS 304801). An increase in HDL and a decrease in LDL levels is a cardiovascular beneficial effect of antisense inhibition of ApoC III.

Table 8 Effect of ASO treatment on total cholesterol levels in transgenic mice ASO Dose Total Cholesterol 3' Internucleoside SEQ
(umol/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 Ga1NAe3-1 PS/20 21 6.75 99 Table 9 Effect of ASO treatment on HDL and LDL cholesterol levels in transgenic mice ASO Dose HDL LDL 3' Internucleoside SEQ
(umol/kg) (mg/dL) (mg/dL) Conjugate Linkage/Length ID No.

0.08 17 23 ISIS 0.75 27 12 None PS/20 32 304801 2.25 50 4 6.75 45 2 0.08 21 21 ISIS 0.75 44 2 Ga1NAe3-1 PS/20 111 647535 2.25 50 2 6.75 58 2 Pharmacokinetics Analysis (PK) The PK of the ASOs was also evaluated. Liver and kidney samples were minced and extracted using standard protocols. Samples were analyzed on MSD1 utilizing IP-HPLC-MS. The tissue level ( g/g) of full-length ISIS 304801 and 647535 was measured and the results are provided in Table 10. As illustrated, liver concentrations of total full-length antisense compounds were similar for the two antisense compounds.
Thus, even though the Ga1NAe3-1 -conjugated antisense compound is more active in the liver (as demonstrated by the RNA and protein data above), it is not present at substantially higher concentration in the liver. Indeed, the calculated EC50 (provided in Table 10) 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 GaINAc3-1 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 10 PK analysis of ASO treatment in transgenic mice Internucleoside Dose Liver Kidney Liver EC50 3'SEQ
ASO Linkage/Length ([1,Mo1Ikg) (m/g) (m/g) 040 Conjugate ID No.
0.1 5.2 2.1 ISIS 0.8 62.8 119.6 53 None PS/20 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 21 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 10a. 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 GaINAc3-1 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 10a 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 I

Cleavage site A ¨I

HO OH Cleavage site C 0.i Cleavage site D 0 /NIx--Li N
H 0 OH <
NHAc 0 d HO OH o...0 -.___..
Cleavage site B
I C\lea_voage site C 0 P=0 HO
NHAc Cleavage site D 0 0 0_.
OH
HO HN

_,...7Ø...\x0w HO Ir\ Cleavage site D
NHAc Cleavage site C 0 I
0=P¨OH NH2 i Metabolite 1 Metabolite 2 OH
\ __ /
H

Is 304801 0.1?-0H NH, H 0 ex-L.N
HO,....,......nr,HNN.---ti OH
_________________________________________________________________ /

H H ____________________________________ I
P=0 OH

Metabolite 3 HO ENI.,..,7 ------./ 0 0=P-OH NH, 6 Nf, H,NI OHN...vniõ...,..N.--.ti 0 d R
H H I
______________________________________________________________ P=0 I

Metabolite 4 HO

0=P-OH NH, HO OH
H,NIN._,,,,,,,,,,,N
d NR
H I
H,NIN.7/\.,,.....,N0 N
H 0 __ P=0 I

Metabolite 5 HN-----HO

0=-OH NH, H 0 exL, N
OH
H2Nõ7\õN........t Lco N el )' 0 d 0_,_.
C2.,\
H _______________________________________________________________ I
P=0 Metabolite 6 HN -----H,N1.........7 \Z./

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 4, 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 11-15, 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 GaINAc3-1 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 11 Effect of ASO treatment on ApoC III mRNA levels in human ApoC III transgenic mice PBS S
ASO
Dose ED50 3' Internucleoside SEQ ID
cY0 (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
10 21 Table 12 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 Table 13 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 Table 14 Effect of ASO treatment on total cholesterol levels in transgenic mice DoseInternucleoside ASO % PBS 3' ConjugateSEQ ID No.
(mg/kg) Linkage/Length None PS/20 20 0.3 93 Ga1NAc3-1 PS/20 21 0.3 115 Ga1NAc3-1 PS/PO/20 21 Table 15 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 Ga1NAe3-1 PS/20 21 0.3 143 89 Ga1NAe3-1 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 4, 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 16, both antisense compounds lowered SRB-1 mRNA
levels. Further, the antisense compound comprising the GaINAc3-1 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 GaINAc3-1 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 16 Effect of ASO treatment on SRB-1 mRNA levels in Balb/c mice Internucleosid ASO
Dose Liver ED50 e SEQ ID
3' j (mg/kg) % PBS (mg/kg) Conugate linkage/Lengt No.

0.7 85 440762 7 12 2.2 None PS/14 22 0.07 98 ISIS 0.2 63 651900 0.7 20 0.3 Ga1NAc3-1 PS/14 23 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+penistrep (-1 ml / 10 ml starting whole blood volume). A 60 [El sample was pipette into a sample vial (Beckman Coulter) with 600 IA VersaLyse reagent (Beckman Coulter Cat# A09777) and was gently vortexed for 10-15 sec. The sample was allowed to incubate for 10 min.
at rt and being mixed again before counting. The cell suspension was counted on Vicell XR cell viability analyzer (Beckman Coulter) using PBMC cell type (dilution factor of 1:11 was stored with other parameters). The live cell/ml and viability were recorded. The cell suspension was diluted to 1 x 107 live PBMC/ml in RPMI+ 10%
FB S+pen/strep.

The cells were plated at 5 x 105 in 50 u1/well of 96-well tissue culture plate (Falcon Microtest). 50 u1/well of 2x concentration oligos/controls diluted in RPMI+10% FBS+penistrep.
was added according to experiment template (100 u1/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 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 uM
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 GaINAc3-1 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 Ga1NAc31 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 GaINAc3-1 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 GaINAc3-1 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 17 Modified ASOs ASO Sequence (5' to 3') Target SEQ ID
No.
ISIS GesmCesTesGesAesTdsTdsAdsGdsAdsGds TNFa 24 104838 AdsGasAdsGasGesTesmCesmCesmCe ISIS TesmCesmCesmCdsAdsTdsTdsTdsmCdsAdsGds CRP 25 353512 GasAdsGasAdsmCdsmCds esGesGe ISIS AesGesmCesTesTesmCdsrrdsrrdsGds-rds Apoc III 20 304801 mCdsmCdsAdsGdsmCds TesTesTesAesTe ISIS AesGesmCesTes rr esmC ds T dsTdsGdsT ds 647535 mCdsmCdsAdsGdsmCdsTesTesTesAesTeoAdo,-GalNAC3-la ApoC III 21 ISIS AesGeomCeoTeoTeomCdsTdsTdsGdsTds ApoC III 20 616468 mCdsmCdsAdsGdsmCdsTeoTeoTesAesTe Subscripts: "e" indicates 2' -MOE modified nucleoside;
"d" indicates fl-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 18 Proinflammatory Effect of ASOs targeting ApoC III in hPBMC assay ASO
EC50 E. E /EC3' Internucleoside SEQ ID
max 011\4) 01M)50 Conjugate Linkage/Length No.

0.01 265.9 26,590 None PS/20 25 (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 ILEM concentrations of modified oligonucleotides. After a treatment period of approximately 16 hours, RNA

was isolated from the cells and mRNA levels were measured by quantitative real-time PCR and the hApoC
III mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN.
The IC50 was calculated using the standard methods and the results are presented in Table 19. As illustrated, comparable potency was observed in cells treated with ISIS 647535 as compared to the control, ISIS 304801.
Table 19 Modified ASO targeting human ApoC III in primary hepatocytes Internucleoside SEQ
ASO IC50 (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 20, 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 20 Effect of ASO treatment on ApoC III protein levels in human ApoC III
transgenic mice PBS S
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 ,IL CN
DMTO....0 0 0 Compound 56 is commercially available from Glen Research or may be prepared according to published procedures reported by Shchepinov et al., Nucleic Acids Research, 1997, 25(22), 4447-4454.
Example 28: Preparation of Compound 60 Ac0 OAc Ac0 OAc ._...70...\0 H2/Pd Ac0 OBn Me0H
TMSOTf, DCE
NJ AcHN 58 (quant.) ( 71%) CNEtO(N(iPr)2)PC1, Ac0 OAc Ac0 OAc N(Pr)2 ___...2..\,n ____________ v.
Ac0--"*"2--\r 0130//
Ac0 '' ====OH CH2C12 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 can be used to prepare phosphoramidites having a predetermined composition.
Example 29: Preparation of Compound 63 CN
1. BnC1 ,,,OH 1. DMTC1, pyr HO 2. KOH, DMSO
_____________________________ Bn0 ). 014 2. Pd/C, H2 ,.. 0 PõOODMT
\
3. HC1, Me0H 3.
Phosphitylation 1 4. NaHCO3 61 N(iPr)2 Compounds 61 and 62 are prepared using procedures similar to those reported by Tober et al., Eur.1 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
ri CN
rj 1. DMTC1, pyr H
0....---...,...õ..OH
2. TBAF
______________________________________ _ P
3. Phosphitylation 1 \--\ O
N(iPr)2 63a OH 63b ODMT
Compound 63a is prepared using procedures similar to those reported by Hanessian et al., Canadian Journal of Chemistly, 1996, 74(9), 1731-1737.
Example 31: Preparation of Compound 63d HO ¨ \ DMT0¨\
\
O. N(iPr)2 0, 1. DMTC1, pyr 1 HO 0 \/-------00Bn 2. Pd/C, H2 /O /
3. Phosphitylation O.--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 H2N IrOTBDMS Ac0 OAc 0 CO2Bn Ac0 0 OH R ______________________________ Ac0__......(2..\.,0).LN OTBDMS
AcHN 64 HBTU, DlEA AcHN 66 H
R
R = H or CH3 1. TEA.3HF Ac0 OAc, THE 0 CO2Bn ________________ ..-Ac0 P
2. Phosphitylation H I
AcHN R N(iPr)2 Compound 64 was prepared as per the procedures illustrated in Example 2.
Compound 65 is prepared using procedures similar to those reported by Or et al., published PCT International Application, 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 (i)Bn Ac0 OAc H2N r CH3 Ac0 OAc Ac0 0 ___...1.2...\,0 OH _____________________________________ ....).L
H BTU, DIEA
___...7(...)..\,0 .LN rOBn ).- Ac0 DM F H µ..., AcHN 64 AcHN 69 k_, H3 Ac0 OAc 1. Pd/C. H2 0 ________________ w (Ø.:l 0 Ac0___ , N 0,p,0 CN
2. Phosphitylation H I I
AcHN CH3 N(iPr)2 Compound 64 was prepared as per the procedures illustrated in Example 2.
Compound 68 is commercially available. The protecting group used for Compound 68 is meant to be representative and not intended to be limiting as other protecting groups including but not limited to those presented in the specification herein can be used.
Example 34: Preparation of Compound 75a O
1. TBDMSC1, pyr YCF3 2. Pd/C, H2 HN N(iPr)2 ...._ NC 3. CF3CO2Et, Me0H H
NC OH _________________ "- F3C/I\I
NC 0 4. TEA.3HF, THF [I 0 o HN
5. Phosphitylation 0 CF3 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,.....,..---0I..õ.., -"--() DCI, NMI, ACN
1. BnCI, NaH
DMT00-.,_=,.- i OH ___________ . HO 0 OBn Phosphoramidite 60 õ,..,s.õ...7.----- cr.) 2. DCA, CH2Cl2 0 DMTO HO

Ac0 OAc NC.., LO

AcHN
NC --.1 1. H2/Pd, Me0H
_____________________________________________________________________ .-Ac0 OAc LO 0, 2. Phosphitylation 0 , 1 AcHN IC) NC---\___ fol Ac0 OAc t Ac0 NHAc 78 Ac0 OAc NC...
Ac0 o\-----Nõ..----\ ), 0 c;1 AcHN
NC --...\
Ac0 OAc LO 0, ......7.2...\,0 i 13'p7()CN
AcHN 1C) 1 NC¨k N(iP02 \--(?
Ac0 OAc 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)....3,,, N(iP02 HO-..........7\....::aõ, 1. FmocC1, pyr 1 HO.,,,,=\_=,0 OBn 2. Pd/C, H2 Fmoc00 ,13 CN

____________________________________ 0.

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) ( ,...._00DMT
(..1-----7"-ODMT
Bx DMTOC'r rt -i-------/---ODMT

0 NC,0,i),0,(0),Bx N C '0¨ P =0 1. DCA, DCM
6 2. DCI, NMI, ACN __ .-0'.
, I , Phosphoramidite 56 NC '.0--P=0 OLIGO , _____________ .
, _________________ , DNA/RNA 6 IDl ,automated synthesizer, I , .
I OLIGO
C)¨VIMAD-0¨p_oCN .
O , X
79b I
0¨VIMAD_o_p_oCN
X = S- or 0- X
Bx = Heterocylic base 1. Capping (Ac20, NMI, pyr) 80 2. t-BuO0H
3. DCA, DCM
-....\ 4.. DCI, NMI, ACN
Ac0 OAc NC
, Phosphoramidite 60 Ac00,,,,______\ A;
AcHN
rCN
Ac0 OAc NC---1 ---'0 .C. 0) Bx AcHN C) 0' NC----\ i j NC 0¨P =0 \----0 6 Ac0 Ac , I
OLIGO , Ac0 /1 0 , ________ oI , NHAc I
0¨VIMAD-0-13_0CN
X
1. Capping (Ac20, NMI, pyr) 81 2. t-BuO0H
3. 20% Et2NH inToluene (v/v) 4. NH4, 55 C, V

HO OH
u HO---r.C.2...\--s 0 ,K
0 i 0 AcHN
HO OH
0 0, 0 _.1..2...\,(Th II ii r)NeõBx HO `-'./\/\./N .1). o __ ,0-1), -kJ \ /

AcHN 0' 0' 0=P-0-HO H 9 y O

lic2_voco- 6, , OLIGO .
HO ' ________ , NHAc 82 wherein GalNAc3-2 has the structure:
HO OH
_IV
AcHN 0- --HO OH
0 0, 0 _.....1.2.\, --,(y--,0-1h0'4=c(5''Bx AcHN 0 o ' 0=1-0-HO H y HO" 1 P, ,....12..\.0' 60 NHAc The GalNAc3 cluster portion of the conjugate group GalNAc3-2 (GalNAc3-2a) can be combined with any cleavable moiety to provide a variety of conjugate groups. Wherein GalNAc3-2a has the formula:
HO OH
HO--4)..0 2 -.....----,...,, o , 0 AcHN 0- ---HOOH
0 0, 0 , 0 AcHN 0' HO OH 9 y .....12...\/0...._z=--,7"-----0 69 HO
NHAc The VIMAD-bound oligomeric compound 79b was prepared using standard procedures for automated DNA/RNA synthesis (see Dupouy et al., Angew. Chem. Int. Ed., 2006, 45, 3623-3627). The phosphoramidite Compounds 56 and 60 were prepared as per the procedures illustrated in Examples 27 and 28, respectively. The phosphoramidites illustrated are meant to be representative and not intended to be limiting as other phosphoramidite building blocks including but not limited those presented in the specification herein can be used to prepare an oligomeric compound having a phosphodiester linked conjugate group at the 5' terminus. The order and quantity of phosphoramidites added to the solid support can be adjusted to prepare the oligomeric compounds as described herein having any predetermined sequence and composition.

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

I Phosphoramidite 79 , OLIGO -sDNA/RNA \
\ ____________________ i I automated synthesizer, 0¨

VIMAD¨O¨P0C N X = S" or 0" ¨
K Bx = Heterocyclic base 79b Ac0 OAc NC-...\
p ......r.....\.7õ L.o Ac0 u ,K
AcHN 0 oi ---\
OxAc0 OAc NC 0 0....., O-F)-0(Or Bx AcHN 0' 0' NC---\..... i 6i NC 1 O¨P=0 Ac0 Ac 0 I
..2...\/oo' 13-0 OLIGO
Ac0 = _________ NHAc O
I
p_..
1. Capping 0¨VIMAD-0-'c 2. t-BuO0H i 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 GalNAc3-2 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 GalNAc3-2 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 Ac00 H
N"----N----)r-N H 1 H2, Pd/C, Me0H
(93%) AcHN \...---Ny_,..1 H H 0 O.
Äo 2 Bn0 OH
0...õ..--N 0 40 83a OAc 0 0 Ac0 H
0 0 o-- HBTU, DIEA, DMF, 76%
Ac0 __________________________________________________________ i.-NHAc HNN___C-j 3. H2,Pd/C,Me0H

OAc 0 Ac0 OAc Ac0 Aci:/ Ac0--...r.2.0 H
NHAc N"---N-----)r-N H
AcHN

H H 8 L ,L,)c F
110z-INNVN---N--11.--N--0,- __________________________________ NH
83b o o cr"
F
Ac0 F NHAc HN N____ .VN.7N,Cj ... ______________________________________________ 83c Pyridine, DMF H 0 /--OAc Ac0 /
Ac0 OAc Ac0 NHAc 0 83e Ac0--. N

H 3' 5', I I
N----N-----)r--N H
AcHN
0 o F F ( OLIGO j-0-P-0-(CH2)6-NH2 ill 7.N.,...-NH 0 0 o_.... fa OH
F

Ac0 OAc NH 0 Borate buffer, DMSO, pH
8.5, rt Ac0/ 0 F F
NHAc HNVN./\ N____e H 0 83d OAc _/--/-0 Ac00,\.>/
Ac0 NHAc Ac0 OAc Ac0.-.../%2.0 H
N"--N----)r--N H
AcHN \---\N ,,.,_ OH

N N N-(CI-12)6-0-P-0¨( OLIGO ) AcO OAc H I I
Ac0 0 0 0-- 0 NHAc HNN __ e 83f /-OAc ___/
Oe Ac0\
Ac0 NHAc Aqueous ammonia HO OH
HO----122.0 H
H
AcHN
0 \---\Ny..õ..1 0 0- /,-.....__v-...N
HN7N......4___.-0.-- NH N-(CH2)6-0 -P-0- ( OLIGO ) H07....\/ H

HO 0--7----/-"IN

0 0 _____e 83h HN N
NHAc H 0 /
OH _1 0 HO \i/07/
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'-GalNAc3-3 conjugated oligomeric compound (83h).
Wherein GalNAc3-3 has the structure:
HO OH
H 0 - --= r ..C.3.. 0 H
AcHN N--N--)r-N H
ni\I
0 y----1 o o OH
NN---N---N'--rN--O,.....----NH

H I I
HO
NHAc HNr-NN_____Ci OH __/--/-%
HO
NHAc .
The GalNAc3 cluster portion of the conjugate group GalNAc3-3 (GalNAc3-3a) can be combined with any cleavable moiety to provide a variety of conjugate groups. Wherein GalNAc3-3a has the formula:

HO OH
HO--¨r.2\0 H
AcHN
\N
0 ).1-----1 0 0 H H 0 0- )LA
NN----N---11---N--0.----NH N-(CH2)6-0-1 HO
NHAc HN N
H
OH
HO
HO
NHAc .

Example 40: General method for the preparation of oligomeric compound 89 comprising a phosphodiester linked Ga1NAc3-4 conjugate at the 3' terminus via solid support (0--/---/ODMT

1. DCA UNL 0 0 -ODMT ______________________________________ . rt_ ----.7.---0Fmoe 2. DC1, NMI, ACN I
30 N(iP02.
Fmoc00, I O

DMTOõ...0ThA

3. CappingDO MT rCN
4. t-BuO0H (0,/---/
0-j r OFmoc F; /-0Fmoc 1. 2% Piperidine, /
2% DBU, 96% DMF 0 0 0 __ OFmoc _________________ ..- I
3. DCI, NMI, ACN C4-UNL-0-P-0CN 86 0-/
Phosphoramidite 79a O
, ________________ , DNA/RNA 1. Capping ,automated synthesizer 2. t-BuO0H, 3. 2% Piperidine, Ac0 OAc 2% DBU, 96% DMF
Ac0 4. DCI, NMI, ACN
*\0 Phosphoramidite 60 , AcHNo\--\_\__ DNA/RNA
,automated synthesizer, __ ( 5. Capping Ac0 OAc 0 r Ac0 ______ NC \
0 _1- CN
AcHN o\-----N___N_______\

\--0 N---"\--0 0-i)¨
Ac%,.....?.....\/ Ac os....../--...
DMTO--N--"N .}----\

I ,CN
Ac0 0-UNL-0-P-NHAc 1. t-BuO0H
O
2. DCA
3. Oligo synthesis (DNA/RNA automated synthesizer) 4. Capping 5. Oxidation 6. Et3N:CH3CN (1:1, v/v) Ac0 OAc Ac0 *\OL
AcHN 0 Ac0 OAc -Ap Ac0 0-P
/\

----\...-0 AcHN o\-----\---\----\ P

13-0 -----\--0 Ac0 coCI 0=\, 0- -0\ /0"-N----NoF\0 Ac0 DMT-( OLIGO }-----"P\\ r-% I zzCN

NHAc 5' 3' 0 W-HO OH NH4, 55 C
HO*\01_ Y
AcHN 0 HO OH
\----\---\--A P
o-p / \

=----"\-0 AcHN \---\___N__\ p 0 )=-=.. /
0-13N P1=0 N---\--0 HO H 0'1)\--_____ \.(:2 0_\./-õ//-- 0-)------\OH
HO
NHAc ( OLIGO ) __ (CM Y
5' 3' Wherein GalNAc3-4 has the structure:

HO*\0L
AcHN 0 HO OH
\-\-\-\
HO ________________________ O-P\

0- o AcHN
P=0 0"

HO\c2..\/ H

NHAc 1¨(CMY
Wherein CM is a cleavable moiety. In certain embodiments, cleavable moiety is:

0=P-OH N NH2 \ -4 1\1 0-NcOt cif 0=P-OH
The GalNAc3 cluster portion of the conjugate group GalNAc3-4 (GalNAc3-4a) can be combined with any cleavable moiety to provide a variety of conjugate groups. Wherein GalNAc3-4a has the formula:

HO*\0L
AcHN 0 HO OH p HO

AcHN
P=0 HO
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 GalNAc3-2 conjugate at the 5' terminus. A 0.1 M solution of phosphoramidite in anhydrous acetonitrile was used for [3-D-2'-deoxyribonucleoside and 2' -MOE.
The ASO syntheses were performed on ABI 394 synthesizer (1-2 umol scale) or on GE Healthcare Bioscience AKTA oligopilot synthesizer (40-200 umol scale) by the phosphoramidite coupling method on VIMAD solid support (110 umol/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 um, 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 21 ASO comprising a phosphodiester linked Ga1NAc3-2 conjugate at the 5' position targeting SRB-1 Observed SEQ ID
ISIS No. Sequence (5 to 3') CalCd Mass Mass No.
GallNAc3-2a-0,Ado T ksmCksAdsGds T as mC asAdsT as 661134 6482.2 6481.6 26 Gds AdsmCdsTdsTIcsmCk Subscripts: "e" indicates 2' -MOE modified nucleoside;
"d" indicates 0-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 GalNAc3-2a 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 GalNAc3-3 conjugate.
The ASO was characterized by ion-pair-HPLC coupled MS analysis with Agilent 1100 MSD system.
Table 21a ASO comprising a Ga1NAc3-3 conjugate at the 5' position via a hexylamino phosphodiester linkage targeting Malat-1 ISIS, Calcd Observed No.
Sequence (5' to 3') Mass Mass SEQ ID No.
5' -Ga1NAc3-3._0,mCesGesGesTesGes 661166 mCdsAdsAdsGdsGdsmCdsTdsTdsAdsGds 5'-Ga1NAc3-3 8992.16 8990.51 27 GesAesAes TesTe Subscripts: "e" indicates 2'-MOE modified nucleoside; "d" indicates f3-D-2'-deoxyribonucleoside;
"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 GalNAc3-2 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 (GalNAc3-1 conjugate at 3' terminus, see Example 9) were included in the study for comparison and are described previously in Table 4.
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 22, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner. Indeed, the antisense oligonucleotides comprising the phosphodiester linked GalNAc3-2 conjugate at the 5' terminus (ISIS 661134) or the GalNAc3-1 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 GalNAc3-2 conjugate at the 5' terminus was equipotent compared to ISIS 651900, which comprises the GalNAc3-1 conjugate at the 3' terminus.
Table 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' Ga1NAc3-2 26 Structures for 3' GalNAc3-1 and 5' GalNAc3-2 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 manner as illustrated in Example 20. Liver sample was minced and extracted using standard protocols. The full length metabolites of 661134 (5' GalNAc3-2) and ISIS 651900 (3' GalNAc3-1) 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 GalNAc3-2 conjugate at the 5' terminus (ISIS 661134) was ISIS 440762 (data not shown). No additional metabolites, at a detectable level, were observed. Unlike its counterpart, additional metabolites similar to those reported previously in Table 10a were observed for the ASO having the GalNAc3-1 conjugate at the 3' terminus (ISIS 651900). These results suggest that having the phosphodiester linked GalNAc3-1 or GalNAc3-2 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 Ga1NAc3-1 conjugate (see Example 9) at the 3' terminus targeting SRB-1 ISIS 655861 and 655862 comprising a GalNAc3-1 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 23, below.
Table 23 Modified ASOs comprising Ga1NAc3-1 conjugate at the 3' terminus targeting SRB-Chemistry SEQ
ISIS No. Sequence (5' to 3') ID
No.
353382 GesmCesTesTesmCesAdsGasTasmCdsAdsTasGdsAds Full PS no conjugate 28 (parent) mCdsTdsTesmCesmCesTesTe G mC T T mC Ad GdsTdsmCdsAdsTdsGdsAds Full PS with 29 655861 m es eses esess CdsTdsTesm Cesm CesTesTeAdo¨GalNAc3-1. Ga1NAc3-1 conjugate G mC Teo Teo mCeo AdsGdsTasmCdsAdsTdsGdsAds Mixed PS/P0 with 29 655862 m es eo CdsTdsTeomCeomCesTesTeoAdo-Ga1NAc3-1. Ga1NAc3-1 conjugate Subscripts: "e" indicates 2'-MOE modified nucleoside; "d" indicates f3-D-2'-deoxyribonucleoside;
"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 24, 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 GalNAc3-1 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 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 52.40 10.4 Full PS without conjugate 28 (parent) 30 24.95 0.5 81.22 Full PS with Ga1NAc3-1 1.5 63.51 655861 5 24.61 2.2 conjugate 29 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 25) 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 25 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 HON3 Pd/C, H2 " n OAc OAc I N Et0Ac, Me0H
103a; n=1 Ac0* OAc OAc 0 Ir.----(-/n N3 103b; n= 7 Ac0 ________________________________ 0.- AcHN
N 104a; n=1 7.__-0 104b; n= 7 4 OAc AcONC:Ac AcHN 0 N........õ<2,....õ 0 OAc OAc OAc OAc N
k / n H
PFPTFA
(---)..\.o NH
AcHN DMF, Pyr AcHN n Ac0 105a; n=1 Compound 90 OAc OAc 105b; n= 7 c)HN 0 n AcHN
106a; n=1 106b; n= 7 OAc AcON:::Ac AcHN ON, 0 OAc OAc n N
H
Ra-Ni, H2 :,:.\:1 HBTU, DIEA, DMF
NH
Me0H, Et0Ac AcHN NH2 OAc OAc HO2CO'Bn r \
....:,:).Ø---\jr,,HN 0 2 Ac0 n AcHN 99 107a; n=1 107b; n= 7 OAc AcON:::Ac OAc OAc n N
H
AcO__:)._\ONH
AcHN _______________________________ NH
(0 OAc OAc Ac0..s..).Ø----.J,HN 0 AcHN
108a; n=1 o0 108b; n= 7 1 Bn OAc AcOAc AcHN 0 Pd/CF12, OAc 108a; n=1 Et0A, c, M0eH OAc n 108b; n= 7 Ac0 NH
AcHNjIIJr-NH

OAc OAc Ac0 "n AcHN
109a; n=1 HO
109b; n= 7 OAc AcONC:Ac AcHN 0 0 r¨OAc OAc AcHN
PFPTFA, DMF, pyr OAc OAc 109a Ac0 AcHN
O
110a 0 F
F F
F F
Compound 4 (9.5g, 28.8 mmoles) was treated with compound 103a or 103b (38 mmoles), individually, and TMSOTf (0.5 eq.) and molecular sieves in dichloromethane (200 mL), and stirred for 16 hours at room temperature. At that time, the organic layer was filtered thru celite, then washed with sodium bicarbonate, water and brine. The organic layer was then separated and dried over sodium sulfate, filtered and reduced under reduced pressure. The resultant oil was purified by silica gel chromatography (2%-->10%
methanol/dichloromethane) 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 !IL) and 3 equivalents of a selected PFP esterified GalNAc3 cluster dissolved in DMSO (50 !IL) 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 GalNAc3 conjugated oligonucleotide.
OH
HONC: H
0 83e 0 3' 5' 11 AcHN C) OLIGO YO¨P-0¨(CH2)6-NH2 OH OH
110a OH HO NH
1. Borate buffer, DMSO, pH 8.5, rt AcHN NH
2. NH3 (aq), rt 0 OH OH

AcHN

Oligonucleotide 111 is conjugated with GalNAc3-10. The GalNAc3 cluster portion of the conjugate group GalNAc3-10 (GalNAc3-10a) 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 GalNAc3-10 below. The structure of GalNAc3-10 (GalNAc3-10a-CM-) is shown below:
NOON

HO "4 AcHN

AcHN
NOON
¨HN
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 umol) was dissolved in 0.1 M sodium tetraborate, pH 8.5 (200 !IL) and 3 equivalents PFP ester (Compound 110a) dissolved in DMSO (50 !IL) was added.
The PFP ester precipitated upon addition to the ASO solution requiring additional DMSO (600 !IL) 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 umol).
Ga1NAc3-10 conjugated oligonucleotide SEQ
ASO Sequence (5' to 3') 5' group ID No.
NHACH2)6-0AdoGesmCesTesTesmCesAdsGdsTds ISIS 660254 Hexylamine 30 mCdsAdsTdsGdsAdsmCdsTasTesmCesmCesTesTe GatNAc3-10.-0,AdoGesmCesTesTesmCesAdsGdsTds ISIS 666881 GalNAc3-10 30 mCdsAdsTds GdsAdsmCdsTasTesmCesmCesTesTe 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 f3-D-2'-deoxyribonucleoside; "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¨c____ H2N (`NHBoc0 BocHNN¨

n H
91a; n=1 HOy¨NO2 BocHNNH TFA, DCM
91b, n=2 n 1..-------- 2 NO
_______________________________ ]..-0 r 0 PFPTFA, DIPEA, DMF
HO
'.0 BocH N ,e.:/ H N "Co /n 92a; n=1 92b, n=2 H2Nill ,..,/ N ,...._ OAcr- OAc I-12N-- -?;.--in -NH NO2 ;= TMSOTf, DCM
Ac0--:-:)..\--0Ac J...
0 AcHN 3 H2N (..:irHN 0 in 93a; n=1 93b, n=2 94a; m=1 94b, m=2 0 OAc OAc OAc OAc ..,,=)-0,Bn Ac0 * HO \ 'mAc0 04-µlin rOH
_________________________________ ... AcHN

yO TMSOTf 7; m=1 Pd/C. H2 64, m=2 OAc AcONC:Ac 0 0 i N
AcHN 0 ri V7 n N
OAc OAc 0 H
93a (93b) Ra-Ni, H2 _____________ ).-HBTU, DIPEA, DMF , m H n AcHN
OAcr_ OAc 0 r H
Ac0---S-).\.0P. N HN '0 AcHN ' ' Ill 96a; n=1, m=1 96b; n=1, m=2 96c; n=2, m=1 96d: n=2. m=2 OAc AcONOAc 0 0 I %
AcHN 0 NTh HBTU, DIEA, DMF
OAc OAc 0 H __________________________ J.
Ac0---_\:NV n NH.1(\...------NH2 AcHN H 0 ODMTr OAc OAc 0 r HO--/(17 Nb H
AcO04.µ N HN0 m A n 0 '''0H
cHN 0 97a; n=1, m=1 97b; n=1, m=2 97c; n=2, m=1 97d; n=2, m=2 OAc AcONC:Ac 0 0 , AcHN 0),, (N"--\(\> 0 H N
i n OAc OAc 0 H
H :) 0 ODMTr '))'')LI\IV'RH (f) N
Ac0 N
k M H
AcHN ) OAc OAc 0 7 H Nb N 0 0 '''OH
AcHN 0 98a; n=1, m=1 98b; n=1, m=2 98c; n=2, m=1 98d; n=2, m=2 OAc Ac0fAc 0 AcHN 11 (Z) \ 11..___()_ HBTU, DIEA, DMF \ 7 n N
97a, n=1, m=1 OAc OAc 0 H 0 0 97b, n=1, m=2 97c, n=2, m=1 0 Ac0 0....c OAcµ 0 1 m 0, 97d, n LO-= HO2C
2, m=2 AcHN H Bn Bn r Ac0 0 i NH (,-,LHN---0 m AcHN

100a, n=1, m=1 100b, n=1, m=2 100c, n=2, m=1 OAc 100d; n=2, m=2 Ac0 \_(:)Ac 0 AcHN O'H N 0 OAc Pd(OH)2/C, Hi rrj(___ 0 0 1 r- OAc 0 H2, Et0Ac, PFPTFA, DMF, me0H
pyr AcHN
OAc ,-OAc 0 r H
.L.E.r?
Ac0 __________________ 04ylli f NN.4...y.HN---0 101a, n=1, m=1 AcHN n 101b, n=1, m=2 0 101c, n=2, m=1 101d; n=2, m=2 OAc Ac0 \C:Ac 0 AcHN ON \(c) F
H n N-(....
OAc r-OAc 0 H 0 0 F 0 F
NVH ii NFIlor7N------1o AGO ---t-1-CrLim F
H H
AcHN F
OAc OAc H
Ac0---P.\11 f 102a, n=1, m=1 AcHN 0 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% methanolidichloromethane) 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% methanolklichloromethane) 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 methanol/ethyl 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% methanol/dichloromethane) to give compounds 102a-d in an approximate 80% yield. LCMS and proton NMR were consistent with the structure.
0 83e 3'5', 11 OLIGO J-0-P-0-(CH2)6 NH2 OH
Borate buffer, DMSO, pH 8.5, rt 102d 2. aq. ammonia, rt AcHN 0 0 4 0¨ CM ____________________________________________________________ OLIGO
HO "4 H 2 H
AcHN

AcHN
Oligomeric Compound 102, comprising a GalNAc3-8 conjugate group, was prepared using the general procedures illustrated in Example 46. The GalNAc3 cluster portion of the conjugate group GalNAc3-8 (GalNAc3-8a) 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 GalNAc3-8 (GalNAc3-8a-CM-) is shown below:

AcHN 0 0 N)LN'-'H,t 0 OM
HO_....,r2..\/0(-)AN-rN----(NH H =

AcHN

AcHN =

Example 48: Preparation of Oligonucleotide 119 Comprising GaINAc3-7 AcO0Ac Ac0 OAc __....i.f\c) TMSOTf, DCE N
Ac0 ________________________ Ac04, C)\,H HCBz Pd(OH)2/C
a 4 ..-v(--4NHCBz AcHN H2, Me0H, Et0Ac N----J HO

4 1 35b 112 HO¨.(----1 HBTU, DIEA
Ac0 OAc 0 0 DMF
0 HO 0 NHCBZ _________ .-Ac0-4r,-,n NH2 +

AcHN 0 (:)L j 105a HO

Ac0 OAc H
Ac0-4.,CN"--t..;

Ac0 OAc AcHN
4r H 0 Ac0 ON
0,,..¨NHCBZ

AcHN 0 0 Ac0 OAc 0)\__ j Ac0.4.,ONH

AcHN

Ac0 OAc H

AcHN
Ac0 OAc Pd/C, H2, Ac0 AcHN 0 Ac0 OAc NH

AcHN

Ac0 OAc H

HBTU, DIEA, DMF AcHN 0 0 Ac0 OAc Ac04, )L.-)LOBn AcHN 0 0 HOr0 Ac0 OAc Ac0 83a AcHN

Compound 112 was synthesized following the procedure described in the literature (J. Med. Chem.
2004, 47, 5798-5808).
Compound 112 (5 g, 8.6 mmol) was dissolved in 1:1 methanol/ethyl acetate (22 mL/22 mL).
Palladium hydroxide on carbon (0.5 g) was added. The reaction mixture was stirred at room temperature under hydrogen for 12 h. The reaction mixture was filtered through a pad of celite and washed the pad with 1:1 methanol/ethyl acetate. The filtrate and the washings were combined and concentrated to dryness to yield Compound 105a (quantitative). The structure was confirmed by LCMS.
Compound 113 (1.25 g, 2.7 mmol), HBTU (3.2 g, 8.4 mmol) and DIEA (2.8 mL, 16.2 mmol) were dissolved in anhydrous DMF (17 mL) and the reaction mixture was stirred at room temperature for 5 min. To this a solution of Compound 105a (3.77 g, 8.4 mmol) in anhydrous DMF (20 mL) was added. The reaction was stirred at room temperature for 6 h. Solvent was removed under reduced pressure to get an oil. The residue was dissolved in CH2C12 (100 mL) and washed with aqueous saturated NaHCO3 solution (100 mL) and brine (100 mL). The organic phase was separated, dried (Na2SO4), filtered and evaporated. The residue was purified by silica gel column chromatography and eluted with 10 to 20 %
Me0H in dichloromethane to yield Compound 114 (1.45 g, 30%). The structure was confirmed by LCMS and 1H
NMR analysis.
Compound 114 (1.43 g, 0.8 mmol) was dissolved in 1:1 methanol/ethyl acetate (4 mL/4 mL).
Palladium on carbon (wet, 0.14 g) was added. The reaction mixture was flushed with hydrogen and stirred at room temperature under hydrogen for 12 h. The reaction mixture was filtered through a pad of celite. The celite pad was washed with methanol/ethyl acetate (1:1). The filtrate and the washings were combined together and evaporated under reduced pressure to yield Compound 115 (quantitative). The structure was confirmed by LCMS and 1H NMR analysis.
Compound 83a (0.17 g, 0.75 mmol), HBTU (0.31 g, 0.83 mmol) and DIEA (0.26 mL, 1.5 mmol) were dissolved in anhydrous DMF (5 mL) and the reaction mixture was stirred at room temperature for 5 min. To this a solution of Compound 115 (1.22 g, 0.75 mmol) in anhydrous DMF
was added and the reaction was stirred at room temperature for 6 h. The solvent was removed under reduced pressure and the residue was dissolved in CH2C12. The organic layer was washed aqueous saturated NaHCO3 solution and brine and dried over anhydrous Na2SO4 and filtered. The organic layer was concentrated to dryness and the residue obtained was purified by silica gel column chromatography and eluted with 3 to 15 % Me0H in dichloromethane to yield Compound 116 (0.84 g, 61%). The structure was confirmed by LC MS and 1H
NMR analysis.
Ac0 OAc H õ
.._.\

AcHN
Pd/C, H2, Ac0 OAc 0 0 116 Et0Ac, Me0H
AcHN 0 0 Ac0 OAc Ac0 ......1:..2....\,0 NH

AcHN
Ac0 OAc H
j õ, F

AcHN
PFPTFA, DMF, Pyr Ac0 OAc )CI 0 . F0 F

AcHN 0 0 Cy j Ac0 OAc Ac0 ........C2...\,0 NH 118 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 1I-INMR analysis.
Compound 117 (0.63 g, 0.36 mmol) was dissolved in anhydrous DMF (3 mL). To this solution N,N-Diisopropylethylamine (70 [EL, 0.4 mmol) and pentafluorophenyl trifluoroacetate (72 [EL, 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 1I-1 and 1I-1 and 19F NMR.
83e 3' 5') 11 F
OLIGO O¨P-0¨(CH2)6-NH2 OH
1. Borate buffer, DMSO, pH 8.5, rt 2. aq. ammonia, rt HO--1Z\01r AcHN No HO 0(r N
OLIGO

AcHN
HO OH

AcHN
Oligomeric Compound 119, comprising a GalNAc3-7 conjugate group, was prepared using the general procedures illustrated in Example 46. The GalNAc3 cluster portion of the conjugate group GalNAc3-7 (GalNAc3-7a) 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 GalNAc3-7 (GalNAc3-7a-CM-) is shown below:

ON) AcHN No HO ON

AcHN OZ
HOOH

AcHN =
Example 49: Preparation of Oligonucleotide 132 Comprising Ga1NAc3-5 HN,Boc HN,Boc ,Boc HN,Boc HN

H2Nro Boc,NThr N o 0 H Boc,NThr N')(OH
Boo,NrOH 0 H

H 0 HBTU, TEA
r Li0H, H20 ______________________ "a- _31._ r ,Boo Me0H, THF
DMF HN HN,Boc 78% 123 Compound 120 (14.01 g, 40 mmol) and HBTU (14.06 g, 37 mmol) were dissolved in anhydrous DMF (80 mL). Triethylamine (11.2 mL, 80.35 mmol) was added and stirred for 5 min. The reaction mixture was cooled in an ice bath and a solution of compound 121 (10 g, mmol) in anhydrous DMF (20 mL) was added. Additional triethylamine (4.5 mL, 32.28 mmol) was added and the reaction mixture was stirred for 18 h under an argon atmosphere. The reaction was monitored by TLC (ethyl acetate:hexane; 1:1; Rf = 0.47).
The solvent was removed under reduced pressure. The residue was taken up in Et0Ac (300 mL) and washed with 1M NaHSO4 ( 3 x 150 mL), aqueous saturated NaHCO3 solution (3 x 150 mL) and brine (2 x 100 mL).
Organic layer was dried with Na2SO4. Drying agent was removed by filtration and organic layer was concentrated by rotary evaporation. Crude mixture was purified by silica gel column chromatography and eluted by using 35 ¨ 50% Et0Ac in hexane to yield a compound 122 (15.50 g, 78.13%). The structure was confirmed by LCMS and 1H NMR analysis. Mass m/z 589.3 [M + H]+.
A solution of LiOH (92.15 mmol) in water (20 mL) and THF (10 mL) was added to a cooled solution of Compound 122 (7.75 g,13.16 mmol) dissolved in methanol (15 mL). The reaction mixture was stirred at room temperature for 45 min. and monitored by TLC (Et0Ac:hexane; 1:1). The reaction mixture was concentrated to half the volume under reduced pressure. The remaining solution was cooled an ice bath and neutralized by adding concentrated HC1. The reaction mixture was diluted, extracted with Et0Ac (120 mL) and washed with brine (100 mL). An emulsion formed and cleared upon standing overnight. The organic layer was separated dried (Na2SO4), filtered and evaporated to yield Compound 123 (8.42 g). Residual salt is the likely cause of excess mass. LCMS is consistent with structure. Product was used without any further purification. M.W.cal:574.36; M.W.fd:575.3 [M + H]+.

=S¨OH = H20 H3NL
H2N=OH + HO
0 ' 0 0 (10 __________________________________________________ e 0¨g 411 Toluene, Reflux 8 99.6%
Compound 126 was synthesized following the procedure described in the literature (1 Am. Chem.
Soc. 2011, 133, 958-963).

HN,Boc 126 BocN
, N j=L 0 0 710.- -r N-r ___________________________________________________________________ O.-H H -3 (I) CH2Cl2 HOBt, DIEA, 0 PyBop, Bop, DMF
r HN,Boc 127 Ac0 OAc Ac0-4-\V

H3N-r HJLN=AcHN 7 0 CF3C00- 0 0 Firc) _____________________________________________ VP-HATU, HOAt, DIEA, DMF
r cF3coo- NH3 128 Ac0 OAc .._..7Ø.
Ac0 Orc) AcHN
NH

Ac0 OAc HN'Thr N JL f,y3-ro 0 N
Ac0 __&.(..:).\, H
OZ-i 0 0 AcHN 0 /
Ac0 OAc Ac0 n ---4-\,-AcHN 0 129 Ac0 OAc O
AcHN
NH
H
Pd/C, H2, Me0H 0 Ac0 OAc Ac0 0 0 AcHN 0 Ac0 OAc NH
Ac0.-r C),r Ac0 OAc AcHN 0 130 Ac00 AcHN
NH
PFPTFA, DMF, Pyr Ac0 OAc Ac0 0 0 AcHN 0 Ac0 OAc NH
AcHN 0 Compound 123 (7.419 g, 12.91 mmol), HOBt (3.49 g, 25.82 mmol) and compound 126 (6.33 g, 16.14 mmol) were dissolved in and DMF (40 mL) and the resulting reaction mixture was cooled in an ice bath. To this N,N-Diisopropylethylamine (4.42 mL, 25.82 mmol), PyBop (8.7 g, 16.7 mmol) followed by Bop coupling reagent (1.17 g, 2.66 mmol) were added under an argon atmosphere.
The ice bath was removed and the solution was allowed to warm to room temperature. The reaction was completed after 1 h as determined by TLC (DCM:MeOH:AA; 89:10:1). The reaction mixture was concentrated under reduced pressure. The residue was dissolved in Et0Ac (200 mL) and washed with 1 M
NaHSO4 (3x100 mL), aqueous saturated NaHCO3 (3x100 mL) and brine (2x100 mL). The organic phase separated dried (Na2SO4), filtered and concentrated. The residue was purified by silica gel column chromatography with a gradient of 50% hexanes/EtOAC to 100% Et0Ac to yield Compound 127 (9.4 g) as a white foam.
LCMS and 1H NMR

were consistent with structure. Mass m/z 778.4 [M + H] -P.
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] -P.
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 [EL) and a stir bar. To this solution was added 0.7 M
pentafluorophenyl trifluoroacetate/DMF (100 [EL) 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

N,...--..õ,...
3' 5', I I 83e HO-&0 0 ( OLIGO J-0-P-0-(CH2)6-1\IH2 AcHN NH
I
OH
1. Borate buffer, DMSO, pH 8.5, rt 131 _________________ ).-2. aq. ammonia, rt HO OH
HN-Thri\kANH
__&....Z, HOOZ---i 0 ...
AcHN 0 /
HO pH
NH
0 NO¨(a/7)¨ 01:)=) AcHN 0 Oligomeric Compound 132, comprising a GalNAc3-5 conjugate group, was prepared using the general procedures illustrated in Example 46. The GalNAc3 cluster portion of the conjugate group GalNAc3-(GalNAc3-5a) 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 GalNAc3-5 (GalNAc3-5a-CM-) is shown below:
HO OH

AcHN
NH

HO OH
HN'ThrNj'LNH
.....rØ....\ro 0 ...
HO
AcHN 0 /
HO OH
0 n NH
HO--4-\v0 N0¨(CM)-1 AcHN 0 =

Example 50: Preparation of Oligonucleotide 144 Comprising Ga1NAc4-11 DMTO rloc 1. TBTU, DIEA DMTO Fmoc Lai ACN, VIMAD Resin pip:DBU:DMF
_______________________________ 0.--L ______________________________________________________________ 0.--. 0 0 2. Ac 20 Capping . a (2:2:96) 0 0 "0-1( )OH Kaiser: Negetive -O
=
\.õ....;

HN-Fmoc DMTO o Fmoc,NOH
Lll 0 DMTr--0 Lol o HBTU, DIEA, DMF
. 0 0 135 "0 NH-Fmoc DMTr ) 1. pip:DBU:DMF 0 / H j 0 1. 2% hydrazine/DMF
Kaiser: Positive Kaiser: Positive H ______ a..

OP ...1.1\1)L(CH2)5'N'fiN....._ O.--2. Dde-Lys(Fmoc)-OH (138) 0 2. Fmoc-Lys(Fmoc)-OH
(140) HATU, DIEA, DMF .:
0 0 HATU, DIEA, DMF
Kaiser: Negative 0 Kaiser: Negative ,Fmoc HN
) /
HNOri,Fmoc DMTr N)L0 0 11 (CH2)5'FIN)-'1\LFmoc ....1 H
. 0 d __Oicla HN,Fmoc Ac0 OAc AcHN
=

Ac0 OAc Ac0-4, H 0 00 AcHN0 1. pip:DBU:DMF

141 Kaiser: Positive HN?
2. 7, HATU, DIEA, Ac0 OAc 0 DMF DMTO
Kaiser: Negative NH
Ac0_.4...\,AcHN
Ac0 OAc AcHN 0 Synthesis of Compound 134. To a Merrifield flask was added aminomethyl VIMAD
resin (2.5 g, 450 umol/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 umolig. 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 + 2H]+.
The solid support bound compound 142 was synthesized using solid phase peptide synthesis methods.

Ac0 OAc AcO____&Z,0 AcHN ---"NH =

\ . .
Ac0 OAc Ac0 0 N NI 0 AcHN 0 H p DNA syntesizer 0 N------AC-)-3-N?
H
142 __________ ).
Ac0 OAc o H NH l (CM) AcO__c, Orrr\ON , , ASO
, AcHN ' Ac0 OAc AcO_,0 143 AcHN 0 HO OH
NH
AcHN 0 HO OH

N NI
AcHN 0 H pH
aqueous NH3 0 H
______________ ).
HO OH o)0 0 0 1\r/OH NH I
HO
( CM ) _____________________________________________________________ ( ASO j AcHN
HO OH
HO___&..1E)...\,0 Th--NH
AcHN 0 The solid support bound compound 143 was synthesized using standard solid phase synthesis on a DNA synthesizer.
The solid support bound compound 143 was suspended in aqueous ammonia (28-30 wt%) and heated at 55 C for 16 h. The solution was cooled and the solid support was filtered.
The filtrate was concentrated and the residue dissolved in water and purified by HPLC on a strong anion exchange column. The fractions containing full length compound 144 were pooled together and desalted. The resulting GalNAc4-11 conjugated oligomeric compound was analyzed by LC-MS and the observed mass was consistent with structure.
The GalNAc4 cluster portion of the conjugate group GalNAc4-11 (GalNAc4-1 1 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 GalNAc4-11 (GalNAc4-11a-CM) is shown below:
HO OH
HO-4-\ro N H
AcHN 0 HO OH

AcHN HN N pH

HO OH

HO NH
ooN fal AcHN
HO OH /
HOo r r¨NH
AcHN

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

OykiNH2 Br)-LOH 0 0 y NIOH

2M NaOHOOH

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

HONA00 Ac0 OAc 35b 4 ___________________________ > Ac0 -.....,..õõ.....õ...õ,...--õ, ..
jt, N 0 .
TMS-0Tf, 4 A molecular sieves, CH2Cl2, rt H
AcHN

0 (Di\ij=OH
Ac0 OAc II
H2, Pd(OH)2 /C
n 0 147 -............,,,,,,_õ,-....õ...õ....-..õ
Et0Ac/Me0H AcHN 105a HBTU, DIEA, DMF, rt Ac0 OAc lik H2, Pd(OH)2 /C, Et0Ac/Me0H
_______________________________________________________________________ )1..
,,,,,,,,,,,..õ--....,....õ........õ
N)1---No AcHN H

Ac0 OAc Ac0 -, N
............w )1õ.............õ-N H2 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 1I-1 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 11-1 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 ..--0 0 0 Ac0- N ).Fr - \irvNyLo Ac0 OAc AcHN 3 H

AcHN 3 H 0 146 ___________ )... Ac0 OAc 0 HBTU, DIEA, DMF ).........NH
_.1..!.:).....vo........."--.1..r.õ
N
Ac0 3 H
NHAc Ac0 OAc Ac0 _____________________________ N N \r07 Pd(OH)2/C, H2 Ac0 OAc AcHN 3 H
_____________ ii. 0 H
..4,0 Me0H, Et0Ac Ac0 N
AcHN
Ac0 OAc 0 NH
0 0........"-.1..r....
N).---NHAc Compound 146 (0.68 g, 1.73 mmol) was dissolved in dry DMF (20 m1). To this DIEA (450 !IL, 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 um). 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 Ac00 NN
Ac0 OAc 0 0 AcHN 3 H

BnO)L0 N1) H Ac0-4\r NN---lrN 3 H OBn 83a 0 151 _________ N.- AcHN 3 H ----.....--:-_0 PFP-TFA, DIEA, DMF
Ac0 OAc 0 NH
...1.1/0.........7,14-.....
N)1-----Ac0 3 H
NHAc Ac0 OAc Ac0-4-\/o H
H'N)N
Ac0 OAc AcHN 3 H O 0 0 Pd(OH)2/C, H2 Ac0 0 \N N C
_________ ).- \/(Jr/N-----1(N 3 H OH
Me0H, Et0Ac AcHN 3 H 0 -_-:---0 Ac0 OAc 0 NH
N)1.----Ac0 NHAc Ac0 OAc Ac0---4..
H'N)NF
Ac0 OAc AcHN 3 H HN--- \r(1, 0 0 F
PFP-TFA, Dl EA 0 N)/\)co .
_______ N... Ac0 '1----F1\1"---1(---N 3 H F
DMF AcHN 3 H 0 -----,-.---0 F
Ac0 OAc 0 NH
Ac0 NHAc Compound 83a (0.11 g, 0.33 mmol) was dissolved in anhydrous DMF (5 mL) and N,N-Diisopropylethylamine (75 uL, 1 mmol) and PFP-TFA (90 uL, 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 1I-INMR 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 [tm). 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 [EL, 0.37 mmol) and PFP-TFA
(35 [EL, 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 CH2C12to yield Compound 154 (0.29 g, 79 %). LCMS
and 1I-INMR were consistent with the desired product.
83e 3' 5', I I HO OH
OLIGO J-O-P-0-(OH2)6 NH2 AcHN HN
1. Borate buffer, DMSO, HOOH0 pH 8.5, rt cm HO O AcHN 4 )C1\1)(N(õ).1\11(N.µõ,y-i 0_¨
¨1oLiGo N

2. aq. ammonia, rt 0 0 0 HOOH
r 0 HO 1\1"--0 AcHN
Oligomeric Compound 155, comprising a GalNAc3-6 conjugate group, was prepared using the general procedures illustrated in Example 46. The GalNAc3 cluster portion of the conjugate group GalNAc3-6 (GalNAc3-6a) 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 GalNAc3-6 (GalNAc3-6a-CM-) is shown below:

Ho_k4-\ONA

AcHN HN

0 y ,INI4NINI9,50 Eg !

AcHN
ENI-----\<

N
AcHN .
Example 52: Preparation of Oligonucleotide 160 Comprising GaINAc3-9 AcO0Ac 0 Ac0 Ac 0 Ac0 0A TMSOTf, 50 C Ac0 10 c ----"T"\O
AcHN CICH2CH2CI, rt, 93% N ---:....-1 TMSOTf, DCE, 66%

Ac0 OAc Ac0 OAc 40 .......2.\,0 o H2, Pd/C
Ac0 1.... 4 t);C Ac0 ,00H
:r Me0H, 95%
AcHN 0 AcHN 0 PH
.-s Ac0 OAc HBTU, DMF, EtN(P02 Ac0 04Nr1R Phosphitylation ...r.?....\, v.-J."
DMTO F10 81%
AcHN 0 ODMT
--b1H

--Hd 47 NC

/
SO¨P
.-s \
Ac0 OAc N(iPr)2 Ac04,0NR
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-HI.
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,/VVV'-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 HO 0 0 AcHN
0=P¨OH
HOOH
HO._õ1.2..\0(NR___0.
0 ¨( CM __ OLIGO
AcHN

Oligomeric Compound 160, comprising a GalNAc3-9 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 GalNAc3 cluster portion of the conjugate group GalNAc3-9 (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 GalNAc3-9 (GalNAc3-9a-CM) is shown below:
pH
HO OH
HO

AcHN
0P¨OH
HO OH
AcHN
0=P¨OH
NOON
AcHN

Example 53: Alternate procedure for preparation of Compound 18 (Ga1NAc3-la and Ga1NAc3-3a) H2NNHR H TMSOTf HO N NHR __________ R = H or Cbz OAc 0 Pgr.....\1 161 RR = z1621a62b 0 CbzCI, Et3N Ac0 N õ
4 r 1/4-) PFPO
OAc Ac0 o 0 NNHR + PFP00NHCBZ
NHAc 0 0 011 Cr FR = Cbz, 163a 316b3a Pd/C, H2 PHD() OAc OAc o
11 Ac0 0 0_ s HNN
NHAc OAc Ac 0 0, AcOO ____________ N
0,,7--NHCBZ

NHAc 0 Oil 10 OAc OAc HNN

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

Example 54: Alternate procedure for preparation of Compound 18 (Ga1NAc3-la and Ga1NAc3-3a) HO2C7Th PFPTFA PFP0 0--. DMF, pyr 0 0 PFP0...ir\r "===-7¨NHCBZ
CY 0 On ICY
H02C.,) PFPO

BocHNN
BocHNN H2 H 0 0,. 1. HCI or TFA
________________________________ '- BocHNN
..NHCBZ ___________________________________________________________ 1 DIPEA , 2.
0 Oiµ 10 OAc 0:gv..... 0 "---7-...----..õ...---. 0 0 BocHN N Ac0 ---1-1 OPFF
H
165 NHAc OAc 166 OPg.
A . v..... 0 c0 0 0_ A
H 1. 1,6-hexanediol "I'l NHNN _ or 1,5-pentane-diol NHAc TMSOTf + compound 4 OAc )rI 2. TEMPO
0:gi........ 0 0..._ 0 , 3. PFPTFA, pyr 0 0_ JI N NH
Ac0 NHAc 0 1:?\ 10 OAc OAc HN.-----....õ------,N11/4---/-0 r, H
Ac0 NHAc The triPFP ester 164 was prepared from acid 113 using the procedure outlined in example 53 above and reacted with mono-Boc protected diamine to provide 165 in essentially quantitative yield. The Boc groups were removed with hydrochloric acid or trifluoroacetic acid to provide the triamine which was reacted with the PFP activated acid 166 in the presence of a suitable base such as DIPEA to provide Compound 18.
The PFP protected Gal-NAc acid 166 was prepared from the corresponding acid by treatment with PFPTFA (1-1.2 eq) and pyridine (1-1.2 eq) in DMF. The precursor acid in turn was prepared from the corresponding alcohol by oxidation using TEMPO (0.2 eq) and BAIB in acetonitrile and water. The precursor alcohol was prepared from sugar intermediate 4 by reaction with 1,6-hexanediol (or 1,5-pentanediol or other diol for other n values) (2-4 eq) and TMSOTf using conditions described previously in example 47.

Example 55: Dose-dependent study of oligonucleotides comprising either a 3' or 5'-conjugate group (comparison of GaINAc3-1, 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 GalNAc3 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 26 Modified ASO targeting SRB-1 SEQ
ASO Sequence (5 to 3') Motif Conjugate ID No.
ISIS 353382 GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds 5/10/5 none 28 (parent) mCdsTasTesmCesmCesTesTe Ges mCesTesTesmCesAd GdsTd mC-d Ad Td GdsAdsISIS 655861 mCTjmCmCANA
5/10/5 Ga1NAc3-1 29 ddesesesTesTedo,Galc3-1.
Ges mC T T mC Ad GdsTd mCd Ad Td GdsAds m ISIS 664078 es es es es s s ss s 5/10/5 Ga1NAc3-C ,r ,rm esCm esCesTesTeoAdo,-es-9.
Ga1NAc3-3.-0,Ado ISIS 661161 GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds 5/10/5 Ga1NAc3-mCdsTdsTesmCesmCesrresrre Ga1NAC3-8.-o'Ado ISIS 665001 GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds 5/10/5 Ga1NAc3-mCdsTasTesmCes mCesTesTe 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 f3-D-2'-deoxyribonucleoside; "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 GalNAc3-1a. was shown previously in Example 9. The structure of GalNAc3-9 was shown previously in Example 52. The structure of GalNAc3-3 was shown previously in Example 39. The structure of GalNAc3-8 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 27, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner. Indeed, the antisense oligonucleotides comprising the phosphodiester linked GalNAc3-1 and GalNAc3-9 conjugates at the 3' terminus (ISIS 655861 and ISIS
664078) and the GalNAc3-3 and GalNAc3-8 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 GalNAc3-9 conjugate at the 3' terminus was essentially equipotent compared to ISIS 655861, which comprises a GalNAc3-1 conjugate at the 3' terminus. The 5' conjugated antisense oligonucleotides, ISIS 661161 and ISIS 665001, comprising a GalNAc3-3 or GalNAc3-9, respectively, had increased potency compared to the 3' conjugated antisense oligonucleotides (ISIS 655861 and ISIS 664078).
Table 27 ASOs containing Ga1NAc3-1, 3, 8 or 9 targeting SRB-1 Dosage SRB-1 mRNA
ISIS No.Conj ugate (mg/kg) (% Saline) Saline 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 serum were measured relative to saline injected mice using standard protocols.
Total bilirubin and BUN were also evaluated. The change in body weights was evaluated with no significant change from the saline group.
ALTs, ASTs, total bilirubin and BUN values are shown in the table below.

Table 28 Dosage Total ISIS No. ALT AST
Bilirubin BUN Conjugate mg/kg 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 (comparison of GaINAc3-1, 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 GalNAc3 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 GalNAc3 conjugate group attached at the 3' terminus.
Table 29 Modified ASO targeting SRB-1 SEQ
ASO Sequence (5' to 3') Motif Conjugate ID No.
ISIS 353382 GesmCesTesTesmCesAdsGasTasmCdsAdsTdsGdsAds 5/10/5 no conjugate 28 (parent) mCcisTdsTesmCesmCesTesTe G mC T T mC Ad GdsTd mCd Ad Td GdsAds ISIS 655861 es es es es es s s s s s 5/10/5 GalNAc3-1 29 mCdsTdsTesmCesmCesTesTeoAdo,-Ga1NAC3-1a GalNAC3-2a-0,AdoGesmCesTõTesmCesAdsGdsTds ISIS 664507 5/10/5 Ga1NAc3-2 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe Ga1NAC3-3a-o'Ado GesmCesTesTesmCesAdsGasTasmCdsAdsTdsGdsAds 5/10/5 Ga1NAc3-3 30 mCdsTdsTesmCesmCesTesTe ISIS 666224 Ga1NAc3-5a-0,AdoGesmCesTesTesmCesAdsGdsTds 5/10/5 Ga1NAc3-5 30 mCdsAdsTdsGdsAdsmCdsTdsTõmCesmCesTesTe GatNAc3-6.-0,AdoGesincõTõTõmCõAdsGdsTds ISIS 666961 5/10/5 Ga1NAc3-6 30 mCdsAdsTdsGdsAdsmCdsTdsrresmCesmCesTesTe GalNAc3-7.-0,AdoGesinCesTõTesinCesAdsGdsTds ISIS 666981 5/10/5 Ga1NAc3-7 30 mCdsAdsTdsGdsAdsmCdsrrdsrresmCesmCesTesTe GalNAc3-10.-0,AdoGesinCesTesTesinCesAdsGdsTas 5/10/5 ISIS 666881 Ga1NAc3-10 30 mCdsAdsTdsGdsAdsmCdsrr dsrresmCesmCesTesTe 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 f3-D-2'-deoxyribonucleoside; "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 GalNAc3-la was shown previously in Example 9. The structure of GalNAc3-2a was shown previously in Example 37. The structure of GalNAc3-3a was shown previously in Example 39. The structure of GalNAc3-5a was shown previously in Example 49. The structure of GalNAc3-6a was shown previously in Example 51. The structure of GalNAc3-7a 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 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 30 Dosage SRB-1 mRNA
ISIS No.Conjugate (mg/kg) (% Saline) Saline 100.0 3 96.0 353382 10 73.1 none 30 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.
ALTs, ASTs, total bilirubin and BUN values are shown in Table 31 below.
Table 31 Dosage Total ISIS No. ALT AST BUN 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 g 1. 37 74 0.2 25 661161 GalNac3-3 (5') 5 g 28 61 0.2 29 21 41 0.2 25 0.5 34 48 0.2 21 1.5 23 46 0.2 26 666224 GalNac3-5 (5') 5 24 47 0.2 23 15 32 49 0.1 26 0.5 17 63 0.2 26 1.5 23 68 0.2 26 666961 Ga1NAc3-6 (5') 5 25 66 0.2 26 15 29 107 0.2 28 0.5 24 48 0.2 26 1.5 30 55 0.2 24 666981 Ga1NAc3-7 (5') 5 46 74 0.1 24 15 29 58 0.1 26 0.5 20 65 0.2 27 1.5 23 59 0.2 24 666881 Ga1NAc3-10 (5') 5 45 70 0.2 26 15 21 57 0.2 24 Example 57: Duration of action study of oligonucleotides comprising a 3'-conjugate group targeting ApoC III in vivo Mice were injected once with the doses indicated below and monitored over the course of 42 days for ApoC-III and plasma triglycerides (Plasma TG) levels. The study was performed using 3 transgenic mice that express human APOC-III in each group.
Table 32 Modified ASO targeting ApoC III
ASO Sequence (5' to 3') Linkages SEQ ID
No.
ISIS AesGesmCesTesTesmCdsTdsTdsGdsTds PS 20 304801 mCdsmCdsAdsGdsmCdsTesTesTesAesTe Isis AesGesinCesTesTesmCdsTasTasGdsTdsmCdsmCds PS 21 647535 AdsGdsmCdsTesTesTesAesTeAdo-Ga1NAc3-1.
ISIS ikesGeomCeorrejeomCcisrrdsrrdsGdsTcismCcismCcIs PO/PS

647536 AdsGdsmCdsTeoTeoTesAesTeoAdo,-Ga1NAC34.
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 f3-D-2'-deoxyribonucleoside; "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 GalNAc3-la was shown previously in Example 9.

Table 33 ApoC III mRNA (% Saline on Day 1) and Plasma TG Levels (% Saline on Day 1) ASO Dose Target Day 3 Day 7 Day 14 Day 35 Day 42 Saline 0 mg/kg ApoC-III 98 100 100 95 116 ISIS 304801 30 mg/kg ApoC-III 28 30 41 65 74 ISIS 647535 10 mg/kg ApoC-III 16 19 25 74 94 ISIS 647536 10 mg/kg ApoC-III 18 16 17 35 51 Saline 0 mg/kg Plasma TG 121 130 123 105 109 ISIS 304801 30 mg/kg Plasma TG 34 37 50 69 69 ISIS 647535 10 mg/kg Plasma TG 18 14 24 18 71 ISIS 647536 10 mg/kg Plasma TG 21 19 15 32 35 As can be seen in the table above the duration of action increased with addition of the 3'-conjugate group compared to the unconjugated oligonucleotide. There was a further increase in the duration of action for the conjugated mixed PO/PS oligonucleotide 647536 as compared to the conjugated full PS
oligonucleotide 647535.
Example 58: Dose-dependent study of oligonucleotides comprising a 3'-conjugate group (comparison of Ga1NAc3-1 and Ga1NAc4-11) targeting SRB-1 in vivo The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice. Unconjugated ISIS 440762 was included as an unconjugated standard. Each of the conjugate groups were attached at the 3' terminus of the respective oligonucleotide by a phosphodiester linked 2'-deoxyadenosine nucleoside cleavable moiety.
The structure of GalNAc3-1a was shown previously in Example 9. The structure of GalNAc3-11a 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 34, treatment with antisense oligonucleotides lowered SRB-1 mRNA levels in a dose-dependent manner. The antisense oligonucleotides comprising the phosphodiester linked GalNAc3-1 and GalNAc4-11 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 GalNAc4-11, were equipotent.
Table 34 Modified ASO targeting SRB-1 % Saline SEQ ID
ASO Sequence (5 to 3') Dose mg/kg control No.
Saline 100 0.6 73.45 'fksmCksAdsGdsTdsmCdsAdsTdsGdsAds 2 ISIS 440762 mr, ..,-, ..,-, r, 59.66 t.,,ds ids I ksmk-,1( 6 23.50 0.2 62.75 TksmCksAdsGdsTdsmCdsi6idsrrdsGdsAds 0.6 29.14 mCdsTdsTksmCkoAGalNAC3-1. 2 8.61 6 5.62 0.2 63.99 TicsmCksAdsGdsTdsmCdsAdsTdsGdsAds 0.6 33.53 ISIS 663748 mCdsTdsTicsmCkoAdo,-GalNAC4-11, 2 7.58 6 5.52 Capital letters indicate the nucleobase for each nucleoside and mC indicates a 5-methyl cytosine.
Subscripts: "e" indicates a 2'-MOE modified nucleoside; "k" indicates 6'-(S)-CH3 bicyclic nucleoside; "d"
indicates a 13-D-2'-deoxyribonucleoside; "s" indicates a phosphorothioate internucleoside linkage (PS); "o"
indicates a phosphodiester internucleoside linkage (PO); and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.
Liver transaminase levels, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), in serum were measured relative to saline injected mice using standard protocols.
Total bilirubin and BUN were also evaluated. The change in body weights was evaluated with no significant change from the saline group.
ALTs, ASTs, total bilirubin and BUN values are shown in Table 35 below.
Table 35 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 Ga1NAc3-1 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 36 Modified ASOs targeting FXI
SEQ ID
ASO Sequence (5' to 3') Linkages No.
ISIS T es Ges GesTesAesAdsTdsmCdsmCdsAdsmCds 404071 TdsTdsTdsmCdsAesGesAesGesGe ISIS T es Ges GesTesAesAdsTdsmCdsmCdsAdsmCds 656172 TdsTdsTdsmCdsAesGesAesGesGeoAdo,-GalNAc3-1.
ISIS TesGeoGeoTe.AeoAdsTdsmC dsmCdsAdsmCds PO/PS 32 656173 TdsTdsTdsmCdsAeoGeoAesGesGeoAdo,-GalNAC3-1.
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 f3-D-2'-deoxyribonucleoside; "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 GalNAc3-la 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 37 Factor XI mRNA (% Saline) Dose ASO % Control Conjugate Linkages mg/kg Saline 100 none 404071 10 40 none PS

ISIS 0.7 74 656172 2 33 Ga1NAc3-1 PS

ISIS 0.7 49 656173 2 22 Ga1NAc3-1 Po/PS

As illustrated in Table 37, treatment with antisense oligonucleotides lowered FXI mRNA levels in a dose-dependent manner. The oligonucleotides comprising a 3'-GalNAc3-1 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 37a, treatment with antisense oligonucleotides lowered FXI protein levels in a dose-dependent manner. The oligonucleotides comprising a 3'-GalNAc3-1 conjugate group showed substantial improvement in potency compared to the unconjugated antisense oligonucleotide (ISIS 404071).
Between the two conjugated oligonucleotides an improvement in potency was further provided by substituting some of the PS linkages with PO (ISIS 656173).
Table 37a 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

ISIS .7 656173 2 6 Ga1NAc3-1 PO/PS

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

Table 38 ISIS No. Dosage ALT AST Total Total 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.
Table 39 Modified ASO targeting SRB-1 SEQ
ASO Sequence (5' to 3') Motif Conjugate ID No.
G mC T T mC Ad GdsTd mCd Ad Td GdsAds ISIS 353382 es es es es es s s s s s 5/10/5 none mCdsTdsTesmCesmCesTesTe G mC T T mC Ad GdsTd mCd Ad Td GdsAds ISIS 655861 es es es es es s s s s s 5/10/5 GalNAc3-1 29 mCdsTdsTesmCesmCesTesTeoAdo,-GalNACrla G mC T T mC AdsGd TdsmCd Ad TdsGdsAds ISIS 655862 es eo eo eo eo s s s 5/10/5 GalNAc3-1 29 mCdsTdsTeomCeomCesTesTeoAdo-Ga1NAc3-1a GalNAc3-3a_0,AdoGesmCesTesTesmCesAdsGds ISIS 661161 5/10/5 GalNAc3-3 30 TdsmCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GalNAc3-8a_0,AdoGesmCesTesTesmCesAdsGds ISIS 665001 5/10/5 Ga1NAc3-8 30 TdsmCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe G mC T T mC Ad GdsTd mCd Ad Td GdsAds ISIS 664078 es es es es es s s s s s 5/10/5 GalNAc3-9 29 mCdsTdsTesmCesmCesTesTeGAdo,-Ga1NAC3-9a GalNAC3-6a-0,AdoGesmCesTõTesmCesAdsGds ISIS 666961 5/10/5 Ga1NAc3-6 30 TdsmCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GalNAC3-2a-0,AdoGesmCesTõTesmCesAdsGdsTds ISIS 664507 5/10/5 GalNAc3-2 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GalNAC3-10a-0,AdoGesmCesTesTesmCesAdsGdsTas 5/10/5 ISIS 666881 Ga1NAc3-10 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GalNAC3-5a-0,AdoGesmCesTõTesmCesAdsGdsTds ISIS 666224 5/10/5 GalNAc3-5 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe GalNAC3-7a-0,AdoGesmCesTõTesmCesAdsGdsTds ISIS 666981 5/10/5 GalNAc3-7 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe Capital letters indicate the nucleobase for each nucleoside and 'V indicates a 5-methyl cytosine.
Subscripts: "e" indicates a 2'-MOE modified nucleoside; "d" indicates a f3-D-2'-deoxyribonucleoside; "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 GalNAc3-la was shown previously in Example 9. The structure of GalNAc3-3a was shown previously in Example 39. The structure of GalNAc3-8a was shown previously in Example 47. The structure of GalNAc3-9a was shown previously in Example 52. The structure of GalNAc3-6a was shown previously in Example 51. The structure of GalNAc3-2a was shown previously in Example 37. The structure of GalNAc3-10a was shown previously in Example 46. The structure of GalNAc3-5a was shown previously in Example 49. The structure of GalNAc3-7a 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 IC50was calculated using standard methods and the results are presented in Table 40. The results show that, under free uptake conditions in which no reagents or electroporation techniques are used to artificially promote entry of the oligonucleotides into cells, the oligonucleotides comprising a GalNAc conjugate were significantly more potent in hepatocytes than the parent oligonucleotide (ISIS 353382) that does not comprise a GalNAc conjugate.
Table 40 Internucleoside SEQ ID
ASO IC50 (nM) Conjugate linkages No.
ISIS 353382 190a PS none 28 ISIS 655861 11 a PS Ga1NAc3-1 29 ISIS 655862 3 PO/PS Ga1NAc3-1 29 ISIS 661161 15' PS Ga1NAc3-3 30 ISIS 665001 20 PS Ga1NAc3-8 30 ISIS 664078 55 PS Ga1NAc3-9 29 ISIS 666961 22' PS Ga1NAc3-6 30 ISIS 664507 30 PS Ga1NAc3-2 30 ISIS 666881 30 PS GaINAc3-10 30 ISIS 666224 30a PS Ga1NAc3-5 30 ISIS 666981 40 PS Ga1NAc3-7 30 'Average of multiple runs.
Example 61: Preparation of oligomeric compound 175 comprising GaINAc3-12 Ac0 OAc Boc , Ac0 H 0 Nzl...
Pfp0)01.C2-1--.Ac 91a )p.._ B OAc oc.N N --Jc.,õ.õõN__ 0 0 HN 'Ac H H OAc HN 'Ac166 HOOC
H >
N
CBz, N \¨ COO H
Ac0 0 Nz...7.0Ac COOH

_,..TFA
____________________________________________________________________ i.-OAc H
DC M HN iokc HBTU DIEA DMF

Ac0 OAc K0 Oji---0Ac Vr HN --Ac HN

}¨N---.7.--/
y 0 Ac0 0 IRII N 0 Ni..1..)Ac \ li ,,,--.........õ,..-^.., ,-, N N OAc Li HN H
HN
0 H iokc ----\-Th 0 HN :7:4Ac OAc HN, -Ac Ac0 OAc K0 o ji___OAc V7 HN,Ac HN
Pd(OH)2/C, H2 0 H
},-N---.7.--/
Me0H/Et0Ac _Jo..
Ac0 Ni..1..)Ac lk N N).'--....-----C) \
Li HN H H
HN
r, iokc ----\-Th 0 HN :7:4Ac OAc HN, -Ac F

F
benzyl (perfluorophenyl) glutarate ______________________________ )0' DMF
Pk:: :).Ac 0 0 OAc Ky......,7-.õ...,0 HN HN....Ac H
Ki 0 0 Ac0 Nz.1.:)Ac 0 1.r.r N
.....--....---õ,..õ.." \ k 0 0 N N).-'--C) OAc O HN H H
HN, Ac ---\-Th 0 HN O.
Azcl OAc OAc HN
'Ac A.,c(j:_)Ac 0 0 OAc HN H N 0 ...Ac H
Pd(OH)2 / C, H2 N
Hml 0 0 AGO
z.1..
Me0H / Et0Ac N N O c2 :)Ac L' H0 N
\
0 0 ,.,\
.OAc HN H H .
HN \Ac---\----\ 0 HN \/____Ac0 OAc OAc 'Ac ici.0:)Ac .P-TFA

0 OAc A DM F
HN H N
0-,Ac H
\--N--.7.---/
F F
HAc0 AAc F ilk 0 N,./N\ 11\ 0 F F 0 OAc HN , -Ac HN \_Ac0 OAc OAc "Ac 83e 3'5'.) 11 J
OLIGO ¨0¨P-0¨(CH2)6¨NH2 OH
174 1. Borate buffer, DMSO, pH 8.5, rt 2. aq. ammonia, rt OH OH
HO

AcHN
NH

HO
0 \--0 AcHN
0 =¨=

0 rf 0 0 j\--NH

OH
H0,000C\2....v HO
NHAc Compound 169 is commercially available. Compound 172 was prepared by addition of benzyl (perfluorophenyl) glutarate to compound 171. The benzyl (perfluorophenyl) glutarate was prepared by adding PFP-TFA and DIEA to 5-(benzyloxy)-5-oxopentanoic acid in DMF. Oligomeric compound 175, comprising a GalNAc3-12 conjugate group, was prepared from compound 174 using the general procedures illustrated in Example 46. The GalNAc3 cluster portion of the conjugate group GalNAc3-12 (GalNAc3-12a) 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 (GalNAc3-12a-CM-) is shown below:

OH OH
H0o 0 AcHN N--NANH
ONDH \------\___H
/
H0o\_\_____\_x AcHN 0 -----/----N N
H H hi 'FNI--er El if NO 0 HO,(\:2_v HO
NHAc Example 62: Preparation of oligomeric compound 180 comprising Ga1NAc3-13 OAcr- OAc 0 \ 0 Ac0---.)\--0 OH + * HATU, HOAt AcHN N j\-11N) ___________________________ IP-176 H2N y HN---.......-------''''.7-Y DIEA, DMF

r OAcr- OAc Ac0---)..\--0)c AcHN NH
OAc OAc Nj-L H2, Pd/C
AcA0cHN 0).L
N Thr N 77).rC) lei H H

/
OAcr- OAc HN
Ac0---.3-\--0 177o AcHN
OAcr- OAc Ac00 AcHN NH
OAc OAc OH
Ac0,\¨OCIL rilVij PFPTFA, TEA
Nr AcHN N _)=,_ H H DMF

OAc OAc r 178 HN
Ac0-...\_:) -0o AcHN

rOAc OAc AcOO 0 AcHN NH
rAc 0Ac 1.4 0 ACOi\IJL r(:) F
AcHN NThr F F
r-Ac OAc AcO
HN
AcHN 0 83e 3'5:1 11 r OLIGO 0-P-0-(CH2)6-NH2 OH
1. Borate buffer, DMSO, pH 8.5, rt 2. aq. ammonia, rt r.-H OH

AcHN NH
OH rOH

/LNI-IN(:)¨ cm ¨ OLIGO) AcHN
Fi 0 0 011-1 r OH 180 HN
AcHN
Compound 176 was prepared using the general procedure shown in Example 2.
Oligomeric compound 180, comprising a GalNAc3-13 conjugate group, was prepared from compound 177 using the general procedures illustrated in Example 49. The GalNAc3 cluster portion of the conjugate group GalNAc3-13 (GalNAc3-13.) 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 GalNAc3-13 (GalNAc3-13a-CM-) is shown below:
OH OH

HO*_ "---NH
AcHN
OH OH
H0*._0 ,crFi 0 H o 0,..,LN N?LNri"-r6 AcHN ki o H 0 0 r HO ...\ õH 0......../...,_,./ j¨NH
HO
NHAc Example 63: Preparation of oligomeric compound 188 comprising Ga1NAc3-14 H OAc Ac0 HOIn HO-(-)' NH2 ' 6 0 0 6 0 0 Ac0 H
HO 0NHCBz 181 ... HON'6N).õ-0 N HCBz N , 0 4 r 0 0 HBTU, D I EA 0 0 Hak). N
HO

OAc OAc AcO\ ( Ac0 H H
Ac0 11100N-6Ny"---\ Ac0 ON-6N l'n OAc NHAc 0 0 OAc NHAc 0 0 Ac0.1õ, Pd/C, 2 Ac0 H
ON H
..4--NHCBz 6- N .r..,,,ON H2 Ac0 0 µ 76 -I" Ac0 0 0NHAc 0 0 0 NHAc /
Ac0 OAc OAc H N4 Ac006H
Ac001 __________ )6 0 Ac0 NHAc NHAc 183 OAc AcO\ ( H
N
HO 0 Ac0 11110N-6 11"----1 0 1. Pd/C, H2 r0 OAc NHAc H 0 0 2. PFP.TFA, pyr, 0 AcO/0 NH

______________ .- Ac0 / 6 HBTU, NHAc OAc 0 DIEA, Ac0 Ac0 V
NHAc OAc AcO\ ( H F
Ac0 11111-0-N-6N 0 F 0 F
/OAc Ac0 NHAc 0 0 0 H
Ac0 / 6 F
NHAc 00 OAc (3)---) Ac0 Ac0 0 OTli NHAc 83e HOOH

E

i OLIG0)-0-P-0-(CH2)6¨NH2 0 0 I HVH?okc H 0 187 1. Borate buffer, DMSO, pH 8.5, rt HO H 6 CM __ OLIGO
___________________ x- NHAc 0 0 0 2. aq. ammonia, rt HO OH\ AN)"\---) HO IPlak µ-"6H 188 NHAc Compounds 181 and 185 are commercially available. Oligomeric compound 188, comprising a GalNAc3-14 conjugate group, was prepared from compound 187 using the general procedures illustrated in Example 46.
The GalNAc3 cluster portion of the conjugate group GalNAc3-14 (GalNAc3-14a) 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 (GalNAc3-14a-CM-) is shown below:

io H
AcHN N

Z
AcHN 0 HOOH
N"--i HO- c H
AcHN
Example 64: Preparation of oligomeric compound 197 comprising GaINAc3-15 Ac0 OAc OTBS OTBS
OH
)\ A
Ac0-- c0 OAc r2-\----- "--Z--Z--\C

AcHN

Ac0_.....r.C...),..vo..._7----..../¨"I

H

HBTU, DIEA AcHN
__________________________________ .

7 N NH3/Me0HOTBS
__________ . Bz20, DMAP
HO OH
___72...Ø___/---_./..--1 AcHN

OH
OTBS
Bz0 OBz Bz0 OBzNO
NO Et3N.HF
0_,,z----__Z---1 Bz0 0 0 _____________________________________ .-Bz0 0 AcHN
AcHN 193 ----Phosphitylation Bz0 OBz _________ ..-.72...\,0....../----..N 5 Bz0 0 NC
AcHN

DMTO
N---\__.--0, N(iPr)2 MTO /
\ DMTO
/
0--\
DMTOV"----/-----0 N-----\_--0, \---CN 5' 3' DMTO
195 N-----N.....-0-----....õ0---FT) ( Oligo /
DMTOV"----------0 _____________ .-SS, DNA synthesizer 196 OH
<11 HO

1. 194, DNA synthesizer AcHN

\¨\--)--Na¨ '-P
2. Aq NH3 55 C, 18 h I

0¨___ N---"\--0, HO...F(..).H 8 ..\
r. PI ¨C) \---N.,.....0-_---,../ "--Frv7) ( Oligo j HO Or N OH /

NHAc 0 0¨P¨OH
\ \
i 0 N

OH j HO....V
HO NHAc Compound 189 is commercially available. Compound 195 was prepared using the general procedure shown in Example 31. Oligomeric compound 197, comprising a GalNAc3-15 conjugate group, was prepared from compounds 194 and 195 using standard oligonucleotide synthesis procedures. The GalNAc3 cluster portion of the conjugate group GalNAc3-15 (GalNAc3-150) 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 (GalNAc3-15a-CM-) is shown below:

NrY) AcHN 0 0 0, HOOH
AcHN 0 9 y 0 op HO H

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 GalNAc3 conjugate groups was attached at the 5' terminus of the respective oligonucleotide by a phosphodiester linked 2'-deoxyadenosine nucleoside (cleavable moiety).
Table 41 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') Conjugate SEQ
No. ID
No.
m 353382 GesCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds mCdsTdsTesmCesmCesTesTe none 28 661161 Ga1NAc3-3.-0,AdoGesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTds GalNAc3-3 30 m m Tes Ces CesTesTe 671144 GalNAc3-12.-0,AdoGesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTds GalNAc3-12 30 m m Tes Ces CesTesTe 670061 GalNAc3-13.-0,AdoGesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTds GalNAc3-13 30 m m Tes Ces CesTesTe 671261 GalNAc3-14.-0,AdoGesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTds GalNAc3-14 30 m m Tes Ces CesTesTe 671262 GalNAc3-15.-0,AdoGesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTds GalNAc3-15 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 f3-D-2'-deoxyribonucleoside; "s" indicates a phosphorothioate intemucleoside linkage (PS); "o" indicates a phosphodiester intemucleoside linkage (PO);
and "o" indicates -0-P(=0)(OH)-. Conjugate groups are in bold.

The structure of GalNAc3-3a was shown previously in Example 39. The structure of GalNAc3-12a was shown previously in Example 61. The structure of GalNAc3-13a was shown previously in Example 62.
The structure of GalNAc3-14a was shown previously in Example 63. The structure of GalNAc3-15a 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 GalNAc3-3, 12, 13, 14, and 15 conjugates showed substantial improvement in potency compared to the unconjugated antisense oligonucleotide (ISIS 335382).
Table 42 SRB-1 mRNA (% Saline) ISIS No. Dosage (mg/kg) SRB-1 mRNA (% ED50 (mg/kg) Conjugate Saline) Saline nia 100.0 nia nia 3 85.0 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 GalNAc3-3 2 x 2.5 27.5 17.4 0.5 101.2 1. 76.1 671144 3.4 GalNAc3-12 5 32.0 15 17.6 0.5 94.8 670061 1.5 57.8 2.1 GalNAc3-13 5 20.7 15 13.3 0.5 110.7 1.5 81.9 671261 4.1 GalNAc3-14 39.8 15 14.1 0.5 109.4 1.5 99.5 671262 9.8 GalNAc3-15 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 group (data not shown). ALTs, ASTs, total bilirubin and BUN values are shown in Table 43 below.
Table 43 Total Dosage ALT BUN
ISIS No. AST (U/L) Bilirubin --Conjugate (mg/kg) (U/L) (mg/dL) (mg/dL) Saline nia 28 60 0.1 39 nia 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 GalNAc3-3 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 GalNAc3-12 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 GalNAc3-13 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 GalNAc3-14 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 GalNAc3-15 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 GalNAc3 conjugate groups was attached at the 5' terminus of the respective oligonucleotide by a phosphodiester linked nucleoside (cleavable moiety (CM)).
Table 44 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') GalNAc3 CM SEQ
No. Cluster ID No.
661161 Ga1NAc3-3 - Ad G mC T T mC A G T mC A T
¨ a ¨ GalNAc3-3a Ad 30 es es es es es ds ds ds ds ds ds m m Gds Ads C dsTdsT es C es CTes esTe 670699 Ga1NAc3-3 - Td G mC T T mC A G T mC A T
¨ a ¨ GalNAc3-3a Td 33 es eo eo eo eo ds ds ds ds ds ds m m Gds Ads C dsTdsTeo Ceo C esT esTe 670700 Ga1NAc3-3 - A G mC T T mC A G T mC A T
¨ a e GalNAc3-3a Ae 30 es eo eo eo eo ds ds ds ds ds ds m m Gds Ads C dsTdsTeo Ceo C esT es 670701 Ga1NAc3-3a - ,T G mC T T mC A G T mC A T
e GalNAc3-3a Te 33 es eo eo eo eo ds ds ds ds ds ds m m Gds Ads C dsTdsTeo Ceo C esT esTe 671165 Ga1NAc1-13 - ¨ G mC T T mC A G T mC A T
¨ a ds ds ds ds ds GalNAc3-13a Ad 30 es eo eo eo eo ds m m Gds Ads C dsTdsTeo Ceo C esT 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 f3-D-2'-deoxyribonucleoside; "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 GalNAc3-3a was shown previously in Example 39. The structure of GalNAc3-13a 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 45, 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 45 SRB-1 mRNA (% Saline) ISIS No. Dosage (mg/kg) SRB-1 mRNA GalNAc3 CM
(% Saline) Cluster Saline n/a 100.0 n/a n/a 0.5 87.8 1.5 61.3 661161 GalNAc3-3a Ad 33.8 14.0 0.5 89.4 1.5 59.4 670699 GalNAc3-3a Td 5 31.3 15 17.1 0.5 79.0 1.5 63.3 670700 GalNAc3-3a A, 5 32.8 15 17.9 0.5 79.1 1.5 59.2 670701 GalNAc3-3a 1', 5 35.8 15 17.7 0.5 76.4 1.5 43.2 671165 GalNAc3-13a 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 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 46 below.
Table 46 Total CM
Dosage ALT AST. . BUN GalNAc3 ISIS No. Bihrubm (mg/kg) (U/L) (U/L) (mg/dL) (mg/dL) 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 661161 GalNAc3-3a Ad 5 26 55 0.2 28 15 27 52 0.2 31 0.5 42 83 0.2 31 1.5 33 58 0.2 32 670699 GalNAc3-3a Td 5 26 70 0.2 29 15 25 67 0.2 29 0.5 40 74 0.2 27 1.5 23 62 0.2 27 670700 GalNAc3-3a Ae 5 24 49 0.2 29 15 25 87 0.1 25 0.5 30 77 0.2 27 670701 GalNAc3-3a 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 GalNAc3-13a Ad 5 33 91 0.2 26 15 33 56 0.2 29 Example 67: Preparation of oligomeric compound 199 comprising GaINAc3-16 OAc AcOC:Ac 0 AcHN 0 õ 2 0 H N(1\'YN
OAcr- OAc 0 H
HrTh<C)DMTr O 1. Succinic anhydride, H
AcHN DMAP, DCE
OAc OAc 0 7 __ N 2. DMF, HBTU, DIEA, Ac0õ i' H
._,(NN.,,i).HN 0 0 OH PS-SS
% 2 2 AcHN 0 98d Ac0 OAc ___...L\
Ac0 .)...., H H
2 \ ) 2 AcHN 0 ODMT
Ac0 OAc 0 H H 0 s/
.__.....C.)...\.,0,HwNsm,____N /. 1. DNA
Synthesizer Ac0 N N ON-2 H )11....tit' \ ________________________________________________________ 2. aq. NH3 AcHN 0 0 0 Ac0 OAc HN

Ac0 0 HN
AcHN 198 HO OH
H H
HO 01_,., NN,c0 CM oligo , , HO OH AcHN
'¨( H
HO
\ _______________________________________________________ AcHN 0 0 OH
HO OH HN--NNO
AcHN

Oligomeric compound 199, comprising a GalNAc3-16 conjugate group, is prepared using the general procedures illustrated in Examples 7 and 9. The GalNAc3 cluster portion of the conjugate group GalNAc3-16 (GalNAc3-16a) 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 GalNAc3-16 (GalNAc3-16a-CM-) is shown below:

HO\
4 H 2 H i cm )-1 AcHN H 0 0 ,--0 NR
AcHN OH

HOr..,N(.0 AcHN
Example 68: Preparation of oligomeric compound 200 comprising GaINAc3-17 OAc 83e Ac0 ' ,0Ac 0 0 5' II

AcHN 0 N---.D F ( OLIGOYO-P-0-(CH2)6-NFI2 H
OAcr-OAc On 0 1. Borate buffer, DMSO, pH 8.5, rt AcHNOAc OAcH 0 r F 2. aq. ammonia, rt Ac0 0N H N0 AcHN 0 102a HO\
H
AcHN 0 0 HOOH 0 0 r,i))-L
, N0¨ CM _____________________________________________________________ OLIGO, H . ______________________________________________________________ , HO-4\[\1FiN-1'Fl AcHN

(.
H
AcHN

Oligomeric compound 200, comprising a GalNAc3-17 conjugate group, was prepared using the general procedures illustrated in Example 46. The GalNAc3 cluster portion of the conjugate group GalNAc3-17 (GalNAc3-17a) 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 GalNAc3-17 (GalNAc3-17a-CM-) is shown below:

AcHN H 0 0 N)Nri'. Ezi H

AcHN

( HO__.72._\(1).L3 NON
H H
AcHN
Example 69: Preparation of oligomeric compound 201 comprising GaINAc3-18 OAc Ac00Ac 0 83e AcHN 0"--***--Y2'..'"'"ANN_40 F
H 0 0 F 16 F ( OLIG0)-0-7-0-(CH2)6-NH2 OAcc- OAc F OH
F
AcHNOI
OAc OAc H H 1. Borate buffer, DMSO, pH 8.5, rt H 0 r ______________________________________________________________ 1.-AcOr-----(:) (:)7,i.,)ThrNHNI"N) 2. aq. ammonia, rt AcHN 2 0 102b HOOr N
i H
AcHN 0 0 9 _¨,N)LN ^ ¨
"4 0¨ CM ¨ OLIGO
HO Ot-etHN'./ H H
H
AcHN

( N

AcHN 201 Oligomeric compound 201, comprising a GalNAc3-18 conjugate group, was prepared using the general procedures illustrated in Example 46. The GalNAc3 cluster portion of the conjugate group GalNAc3-18 (GalNAc3-18a) 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 GalNAc3-18 (GalNAc3-18a-CM-) is shown below:

AcHN H 0 0 0._(cm ) _________________________________________ i _.2......001õ)).LNNH H

AcHN
( _.72..\cy-11)-LNN 0 AcHN

Example 70: Preparation of oligomeric compound 204 comprising GaINAc3-19 AcO0Ac AcO0Ac HBTU, DMF, DIEA
___________________________________________ AcO¨r(jAN
Ac0---T2-\" C0H ...110H
AcHN DMTO AcHN

DMTO

HO
AcO0Ac 0 O Phosphitylation N NC 1. DNA synthesizer AcHN
203 DMTO 2. aq. NH3 (iPr)2N
OH
HO OH

AcHN
0=P¨OH
HO OH

AcHN
0=P¨OH
HO OH
0 0 __ Cm __ OLIGO
AcHN , __ , Oligomeric compound 204, comprising a GalNAc3-19 conjugate group, was prepared from compound 64 using the general procedures illustrated in Example 52. The GalNAc3 cluster portion of the conjugate group GalNAc3-19 (GalNAc3-19a) 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 GalNAc3-19 (GalNAc3-19a-CM-) is shown below:

pH
HooH
HO

AcHN
0=P¨OH
NOON
HO__....7.E)..\000(NR_. 0 AcHN
0=P¨OH
HOOH
HO 0 0 ____ ¨{cm AcHN

Example 71: Preparation of oligomeric compound 210 comprising GaINAc3-20 F 0 2, 3 FF.r..õ1,.(,),)c _______________________________________________________ F 3 N ..iii0H
F...kiiN,Ao DMTO 0 F EtN(iPr) CHCN
F

H 206 DiviTo Hd AcO0Ac 0 0 Ac0 Aopfp K2CO3/Methanol H2NA-i,A AcHN 166 ACN

AcO0Ac 0Phosphitylation __,...T.C.).\,0)........ rYcp...õOH Jo.
Ac0 NH
AcHN
DMTO

AcO0Ac 1. DNA synthesizer ___....2..\,0J____, r(--pp...,10 NC , Ac0 NH \ .,..-0) P 2. aq. NH3 AcHN I
209 DMTO (iPr)2N
pH

F1_0 4., HO o(..-y\-----\1("i.)----NR._ AcHN I
0=P¨OH
I

HO......r.....\z IRII

AcHN I
O=P¨OH
I

OH 0 .' F10....7......\, NR___ 0 0 __ Cm __ OLIGO
AcHN 210 . , '-Compound 205 was prepared by adding PFP-TFA and DIEA to 6-(2,2,2-trifluoroacetamido)hexanoic acid in acetonitrile ,which was prepared by adding triflic anhydride to 6-aminohexanoic acid. The reaction mixture was heated to 80 C, then lowered to rt. Oligomeric compound 210, comprising a GalNAc3-20 conjugate group, was prepared from compound 208 using the general procedures illustrated in Example 52. The GalNAc3 cluster portion of the conjugate group GalNAc3-20 (GalNAc3-20a) 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 GalNAc3-20 (GalNAc3-20a-CM-) is shown below:

AcHN 0 0 0=P ¨0 H
OH

AcHN 0 0 0=--P¨OH
OH

HO cNIZ

AcHN 0 0 El Example 72: Preparation of oligomeric compound 215 comprising Ga1NAc3-21 HO----L AcO0Ac 0 OH
NH
Ac0--"(*.
_________________________________________ Ac0 :)-\/ )LOH
AcHN 176 * AcO0Ac 0 -0=72-\,)-----11 j BOP, EtN(iPr)2, 1,2-dichloroethane AcHN OH

ODMT
AcO0Ac 0 DMTCI, Pyridine,rt Phosphitylation ______________ )... Ac0---\, 11----( AcHN
OH

NC
0---) /
0P\ 1. DNA synthesizer AcO0Ac 0 N(iP02 ________________ .-2. aq. NH3 Ac0 ------11 j AcHN

OH
OH
ni H0*.c..
HO 0--(,-rN--------1_____ AcHN I
0=P¨OH
I

OH
r---j H0*
HO .,..._ 0 c) /N
"3 II

AcHN 0 I
0-=-P¨OH
I

OH
r----/
HO*.z.

HO erN ________ 0 _________________________________ cm __ OLIGO
AcHN

Compound 211 is commercially available. Oligomeric compound 215, comprising a GalNAc3-21 conjugate group, was prepared from compound 213 using the general procedures illustrated in Example 52. The GalNAc3 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 (GalNAc3-21a-CM-) is shown below:
OH
OH
02'Or HO

AcHN
0=P¨OH

OH

"3 H

AcHN
O=P¨OH

OH
0');IrN
HO

AcHN

Example 73: Preparation of oligomeric compound 221 comprising Ga1NAc3-22 H,N .0H H
F3C EN1 .)"L
I I o F3CII N N OH

H
OH
____________________________________________ ..-F DIEA ACN

H
DMT-CI F3C NNODMTr ____________________________ .-__________ ..
II
pyridine 0 H Me0H / H20 H2N N ODMTr Ac0 ,,OAc F
H AcOO___\70-1 0 F

218 OH NHAc F
0,-OAc Ac0EI\11 [ bD\z0.r N ODMTr Phosphitylation Ac0 _______________ 0 H _________________________________________________________ .
NHAc OAc H
Ac0b N
Ac0 N ODMTr H
NHAc I
220 NC (j-P.N opo2 \/

H
NHAc 1. DNA Synthesizer 0 I , 0 ____________ 0.- OH H 11 P( OH 0-r N 0 2. Aq. NH3 N OH
HO

NHAc H

, P( OF-&)_\0 FN11)-LN 0 OH

H
NHAc 221 [i)ig) Compound 220 was prepared from compound 219 using diisopropylammonium tetrazolide. Oligomeric compound 221, comprising a GalNAc3-21 conjugate group, is prepared from compound 220 using the general procedure illustrated in Example 52. The GalNAc3 cluster portion of the conjugate group GalNAc3-22 (GalNAc3-22a) 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 GalNAc3-22 (GalNAc3-22a-CM-) is shown below:

H
NHAc OH

H
NHAc OH H 0 1,0 P' OF-&70,---wir.N(:) H
N

NHAc 0( ___________________________________________ 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 GalNAc3 conjugate groups was attached at the 5' terminus of the respective oligonucleotide.

Table 47 Modified ASOs targeting SRB-1 ISISGalNAc 3 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 Ces CesTesTe Ga1NAc3-3a-0,AdoG es C es T esT es C es AdsGdsTds CdsAdsTds 661161 m m GalNAc3-3a Ad 30 GA CTTCdsTdsT es C es CTes esTe GalNAc1-3 - ,G CT T CA GT CAT
666904 - a es es es es es ds ds ds ds ds ds m m GalNAc3-3a PO 28 GdsAds CdsTdsT es C es C es es es Te Ga1NAc3-17a-0,AdoG CTT CA G T CAT
675441 m es es 711 es m es ds ds ds ds ds ds GalNAc3-17a Ad 30 GA CTTCdsTdsT es C es CTes esTe Ga. MAC 3-1 8 am Ado G CTT CAGT CAT
675442 m es es 711 es m es ds ds ds ds ds ds GalNAc3-18a Ad 30 GdsAds CdsTdsT es C es C es es es Te 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 f3-D-2'-deoxyribonucleoside; "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 GalNAc3-3a was shown previously in Example 39. The structure of GalNAc3-17a 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 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 48, 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 48 SRB-1 mRNA (% Saline) ISIS No. Dosage (mg/kg) SRB -1 mRNA GalNAc3 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 GalNAc3-3a Ad 30.53
12.52 0.5 91.30 1.5 57.88 666904 GalNAc3-3a PO
5 21.22 15 16.49 0.5 76.71 1.5 63.63 675441 GalNAc3-17a Ad 5 29.57 15 13.49 0.5 95.03 1.5 60.06 675442 GalNAc3-18a 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 (data not shown). ALTs, ASTs, total bilirubin and BUN values are shown in Table 49 below.
Table 49 Dosage ALT AST Total . . BUN GalNAc3 CM
ISIS No. Bihrubm (mg/kg) (U/L) (U/L) (mg/dL) (mg/dL) Cluster Saline n/a 26 59 0.16 42 n/a n/a 3 23 58 0.18 39 353382 10 28 58 0.16 43 n/a n/a 30 20 48 0.12 34 0.5 30 47 0.13 35 1.5 23 53 0.14 37 661161 GalNAc3-3a Ad 5 26 48 0.15 39 15 32 57 0.15 42 0.5 24 73 0.13 36 1.5 21 48 0.12 32 666904 GalNAc3-3a PO
5 19 49 0.14 33 15 20 52 0.15 26 0.5 42 148 0.21 36 1.5 60 95 0.16 34 675441 GalNAc3-17a Ad 5 27 75 0.14 37 15 24 61 0.14 36 0.5 26 65 0.15 37 1.5 25 64 0.15 43 675442 GalNAc3-18a 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 41, 44 and 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 (m/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 50 PK Analysis in Liver ISIS No. Dosage Total Tissue Level Parent ASO Tissue GalNAc3 CM
(mg/kg) by UV (m/g) Level by EIC (m/g) Cluster 353382 3 8.9 8.6 22.4 21.0 n/a n/a 30 54.2 44.2 661161 5 32.4 20.7 GalNAc3-3a Ad 63.2 44.1 671144 5 20.5 19.2 GalNAc3-12a Ad 15 48.6 41.5 670061 5 31.6 28.0 GalNAc3-13a Ad 15 67.6 55.5 671261 5 19.8 16.8 GalNAc3-14a Ad 15 64.7 49.1 671262 5 18.5 7.4 GalNAc3-15a Ad 15 52.3 24.2 670699 5 16.4 10.4 GalNAc3-3a Td 15 31.5 22.5 670700 5 19.3 10.9 GalNAc3-3a Ae 15 38.1 20.0 670701 5 21.8 8.8 GalNAc3-3a Te 15 35.2 16.1 671165 5 27.1 26.5 GalNAc3-13a Ad 15 48.3 44.3 666904 5 30.8 24.0 GalNAc3-3a PO
15 52.6 37.6 675441 5 25.4 19.0 GalNAc3-17a Ad 15 54.2 42.1 675442 5 22.2 20.7 GalNAc3-18a Ad 15 39.6 29.0 The results in Table 50 above show that there were greater liver tissue levels of the oligonucleotides comprising a GalNAc3 conjugate group than of the parent oligonucleotide that does not comprise a GalNAc3 conjugate group (ISIS 353382) 72 hours following oligonucleotide administration, particularly when taking into consideration the differences in dosing between the oligonucleotides with and without a GalNAc3 conjugate group. Furthermore, by 72 hours, 40-98% of each oligonucleotide comprising a GalNAc3 conjugate group was metabolized to the parent compound, indicating that the GalNAc3 conjugate groups were cleaved from the oligonucleotides.
Example 76: Preparation of oligomeric compound 230 comprising GaINAc3-23 õ ToSCI NaN3 HOC)(y-OTs Pyr 4, TMSOTf OAc 0 N3 OAci..,\

OAc N3 224 NHAc Pd(OH)2OAc OAcT...,\.,, ACN
C)e\ NH2 _____________________________________________________________ lo H2, Et0Ac, Me0H OAc (:) NHAc 7 F F
\
226 F * F

\ F 0¨ /

¨NO

OAc H
OAci.......
(:) OAc OAc OAc NHAc Hir.. \102 1) Reduce 0 0C)---0 N 2) Couple Diacid OAc 3) Pd/C

NHAc oAcOAc 1 4) PFPTFA
0 0 0 0 NH ________________ v.
----OAc NHAc 228 OAc OAc____ H
N ,C) (:) OAc F
Hira\11-10 F
OAc OAc NHAc OAc 0 0 F F

OAc 0 NHAc OAc OAc .:1.____ 0._\c)0 NH

NHAc 229 83e 3' 5' 11 OLIGO .)-0-P-0-(CH2)6-NH2 OH
1. Borate buffer, DMSO, pH 8.5, rt 2. aq. ammonia, rt OH

OH
OH
H N
0 4 (7N/7) oligo OH
NHAc OH
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 D1EA (273.7 t1, 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 t1, 0.66 mmol) was added and the reaction was purged with argon.
Pentaflourotriflouro acetate (46.39 t1, 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 GalNAc3-23 conjugate group, was prepared from compound 229 using the general procedure illustrated in Example 46. The GalNAc3 cluster portion of the GalNAc3-23 conjugate group (GalNAc3-23a) can be combined with any cleavable moiety to provide a variety of conjugate groups. The structure of GalNAc3-23 (GalNAc3-23a-CM) is shown below:

OH

OH N,0 OH H
H i.r.VF11.r.r N (.-.)0f, _______________________________________________ 01-14...\/NHAc N 4 ( CM H-O

OH \r0 NHAc 01.-OH c;, `-' n............õ--,0õ.--...,..0õ,NH

NHAc Example 77: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising a Ga1NAc3 conjugate The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.
Table 51 Modified ASOs targeting SRB-1 ISIS GalNAc 3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
m m m Ga1NAc3-3a-0,AdoG C T T C A G T C A T
es es es es es ds ds ds ds ds ds 661161 m m m GalNAc3-3a Ad 30 GA CTTC dsTdsT es C es CesTesTe m m m Ga1NAc3-3a - ,G CTT CAGT CAT
666904 es es es es es ds ds ds ds ds ds m m m GalNAc3-3a PO 28 GdsAds CdsTdsT es C es CesTesTe m m m Ga1NAC340a-0,AdoG C T T C A G T C A T
es eo eo eo eo ds ds ds ds ds ds 673502 m n )1 m GalNAc 3-10a Ad 30 GA CTTCdsTdsT. Ceo CesTesTe m m m Ga1NAC3-9 a-0' Ado G CTT CAGT CAT
es es es es es ds ds ds ds ds ds 677844 m m m GalNAc3-9a Ad 30 GA CTTC dsTdsT es C es CesTesTe m m m Ga1NAC3-2 3 am, AdoG CTT CAGT CAT
es es es es es ds ds ds ds ds ds 677843 m m m GalNAc3-23a Ad 30 GA CTTC dsTdsT es C es CTes esTe m m m m m G CTT CAGT CATGA CTT C
655861 es es es es nes ds ds ds ds ds ds ds ds ds ds es es GalNAc 3-la Ad 29 Ces TesTeoAdo,-GalNAc3-1 a 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 GalNAc 3-19a Ad 29 Ces Tes TeoAdo¨GalNAc3-19a 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 GalNAc3-20a Ad 29 Ces Tes TeoAdo¨GalNAc3-20a The structure of GalNAc3-1a was shown previously in Example 9, GalNAc3-3a was shown in Example 39, GalNAc3-9a was shown in Example 52, GalNAc3-10a was shown in Example 46, GalNAc3-19a was shown in Example 70, GalNAc3-20a was shown in Example 71, and GalNAc3-23a 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 52 SRB-1 mRNA (% Saline) ISIS No. Dosage (mg/kg) SRB-1 mRNA GalNAc3 CM
(% Saline) Cluster Saline n/a 100.0 n/a n/a 0.5 89.18 1. 77.02 661161 GalNAc3-3a Ad 5 29.10 12.64 0.5 93.11 1. 55.85 666904 GalNAc3-3a PO
5 21.29 15 13.43 0.5 77.75 1. 41.05 673502 GalNAc3-10a Ad 5 19.27 15 14.41 0.5 87.65 1. 93.04 677844 GalNAc3-9a Ad 5 40.77 15 16.95 0.5 102.28 1. 70.51 677843 GalNAc3-23a Ad 5 30.68 15 13.26 0.5 79.72 1. 55.48 655861 GalNAc3-la Ad 5 26.99 15 17.58 0.5 67.43 1. 45.13 677841 GalNAc3-19a Ad 5 27.02 15 12.41 0.5 64.13 1. 53.56 677842 GalNAc3-20a 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 53 below.
Table 53 Total CM
Dosage ALT AST BUN GalNAc3 ISIS No. Bilirubin (mg/kg) (U/L) (U/L) (mg/dL) (mg/dL) Cluster Saline nia 21 45 0.13 34 nia nia 0.5 28 51 0.14 39 1.5 23 42 0.13 39 661161 GalNAc3-3a Ad 22 59 0.13 37 21 56 0.15 35 0.5 24 56 0.14 37 1.5 26 68 0.15 35 666904 GalNAc3-3a PO
5 23 77 0.14 34 15 24 60 0.13 35 0.5 24 59 0.16 34 1.5 20 46 0.17 32 673502 GalNAc 3-10a Ad 5 24 45 0.12 31 15 24 47 0.13 34 0.5 25 61 0.14 37 1.5 23 64 0.17 33 677844 GalNAc3-9a Ad 5 25 58 0.13 35 15 22 65 0.14 34 0.5 53 53 0.13 35 1.5 25 54 0.13 34 677843 GalNAc3-23a Ad 5 21 60 0.15 34 15 22 43 0.12 38 0.5 21 48 0.15 33 1.5 28 54 0.12 35 655861 GalNAc 3-la Ad 5 22 60 0.13 36 15 21 55 0.17 30 0.5 32 54 0.13 34 1.5 24 56 0.14 34 677841 GalNAc 3-19a Ad 5 23 92 0.18 31 15 24 58 0.15 31 0.5 23 61 0.15 35 1.5 24 57 0.14 34 677842 GalNAc3-20a 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 The oligonucleotides listed below were tested in a dose-dependent study for antisense inhibition of Angiotensinogen (AGT) in normotensive Sprague Dawley rats.

Table 54 Modified ASOs targeting AGT
ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
m A GA GC AG
552668 Ceses esTesesd TdsTa Td Td Tdsa d Cds Cdsõ
es Iva n/a 34 GesAesTe mC A mC T G Ad TdsTd Td Td TdsGd mCd mCdsmCdsAesG' GalNAc3-1a 669509 es es es es es s s s s s s Ad GesAesTeoAdo¨Ga1NAc3-1.
The structure of GalNAc3-la 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 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 55, 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 55 AGT liver mRNA and plasma protein levels ISIS Dosage (mg/kg) AGT liver AGT plasma GalNAc3 Cluster CM
No. mRNA (% PBS) protein (% PBS) PBS n/a 100 100 nia nia 552668 nia n/a 0.3 95 70 669509 GalNAc3- 1 a 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 56 below.
Table 56 Liver transaminase levels and rat body weights Body CM
ISIS No. Dosage (mg/kg) ALT (U/L) AST (U/L) Weight (% GalNAc3 Cluster of baseline) PBS nia 51 81 186 nia nia 552668 nia nia 0.3 53 90 190 669509 GalNAc 3-1 a Ad Example 79: Duration of action in vivo of oligonucleotides targeting APOC-III
comprising a GaINAc3 conjugate The oligonucleotides listed in Table 57 below were tested in a single dose study for duration of action in mice.
Table 57 Modified ASOs targeting APOC-III
ISIS, SEQ
Sequences (5' to 3') CM
No. Cluster ID No.
IkesGesmCesTesTesmCd Td Td Gd Td mcd mcdsAdsGdsmcdsTõTõ
304801 sssss s nia n/a T õA esTe AesGesmCesTesTesmCd Td Td Gd Ta mCdsAdsGdsmCdsTõTõ GalNAc3-la Ad 647535 sssss s TesAõTe.Ado¨GaiNAc3-1.
GalNAc3-3.-0,AdoAesGesmCesTesTesmCdsTdsTdsGdsTdsmCds 663083 GalNAc3-3a Ad 36 mCdsAdsGdsmCdsTesrres rresAesrre GatNAc3-7.-0,AdoAesGesmcesTesTesmCdsTdsTdsGdsTdsmCds GalNAc3-7a Ad 36 mCdaAdaGdamCdsTesTes TõAesTe GaiNAc3-10.-0,AdoAesGesmCesTesTesmCdsTdsTdsGdsTasmCds GalNAc3-10a Ad 36 mCdaAdaGdamCdsTesTes TõAesTe GalNAc3-13.-0,AdoAesGesmCesTesTesmCdsTdsTdsGdsTdsmCds GalNAc3-13a Ad 36 mCdsAdsGdsmCdsrresTes TesAesTe The structure of GalNAc3-la was shown previously in Example 9, GalNAc3-3a was shown in Example 39, GalNAc3-7a was shown in Example 48, GalNAc3-10a was shown in Example 46, and GalNAc3-13a was shown in Example 62.

Treatment Six to eight week old transgenic mice that express human APOC-III were each injected subcutaneously once with an oligonucleotide listed in Table 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.
Table 58 Plasma triglyceride and APOC-III protein levels in transgenic mice Time pointAPOC-III
ISIS Dosage Triglycerides GalNAc3 CM
(days post- protein (%
No. (mg/kg) (% baseline) Cluster dose) baseline) PBS n/a 21 107 107 n/a n/a 304801 30 21 50 50 n/a n/a 647535 10 21 41 41 GalNAc3-la Ad 663083 10 21 28 28 GalNAc3-3a Ad 674449 10 GalNAc3-7a Ad 674450 10 21 44 44 GalNAc 3-10a Ad 674451 10 21 48 48 GalNAc3-13a Ad Example 80: Antisense inhibition in vivo by oligonucleotides targeting Alpha-1 Antitrypsin (AlAT) comprising a GaINAc3 Conjugate The oligonucleotides listed in Table 59 below were tested in a study for dose-dependent inhibition of AlAT in mice.
Table 59 Modified ASOs targeting AlAT
ISISGalNAc3 SEQ ID
Sequences (5' to 3') CM
No. Cluster No.
Aes 'Ces nCes nCesAesAdsTdsTdsmCdsAdsGdsAdsAdsGdsGdsAeaAes476366 ilia ilia GesGesAe AesmCesmCesmCesAesAdsTdsTdsmCdsAdsGdsAdsAdsGdsGdsAesAes GalNAc3-la Ad 38 GesGesAeoAdo,-Ga1NAc34.
GalNAc3-3.-0,AdoAesmCesmCesmCesAesAdsTdsTdsmCdsAdsGdsAds GalNAc3-3a Ad 39 AdsGdsGdsAesAes GesGesAe GalNAC3-7a-o'AdoAesmCesmCesmCesAesAdsTdsTdsmCdsAdsGdsAds GalNAc3-7a Ad 39 AdsGdsGdsAesAes GesGesAe GaiNAe3-10a-0,AdoAesmCesmCesmCesAesAdsTdsTdsmCdsAdsGds GalNAc3-1 Oa Ad 39 AdsAdsGdsGdsAesAes GesGesAe GalNAc3-13 a-0,AdoAesmCesmCesmCesAesAdsTdsTdsmCdsAdsGds GalNAc3-13a Ad 39 AdsAdsGdsGdsAesAes GesGesAe The structure of GalNAc3-la was shown previously in Example 9, GalNAc3-3a was shown in Example 39, GalNAc3-7a was shown in Example 48, GalNAc3-10a was shown in Example 46, and GalNAc3-13a 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 59 or with PBS. Each treatment group consisted of 4 animals.
The mice were sacrificed 72 hours following the final administration. AlAT 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 AlAT liver mRNA and plasma protein levels for each treatment group, normalized to the PBS control.
As illustrated in Table 60, treatment with antisense oligonucleotides lowered AlAT liver mRNA and AlAT plasma protein levels in a dose-dependent manner. The oligonucleotides comprising a GalNAc conjugate were significantly more potent than the parent (ISIS 476366).
Table 60 AlAT liver mRNA and plasma protein levels ISIS Dosage (mg/kg) AlAT liver AlAT plasma GalNAc3 Cluster CM
No. mRNA (% PBS) protein (% PBS) PBS nia 100 100 nia nia 73 61 n/a n/a 0.6 99 90 656326 GalNAc3-la Ad 0.6 105 90 6 16 20 GalNAc3-3a Ad 0.6 90 79 678382 GalNAc3-7a Ad 0.6 94 84 678383 GalNAc3-10a Ad 0.6 106 91 678384 GalNAc3-13a 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 61 below.

Body weight is shown as % relative to baseline. Organ weights are shown as %
of body weight relative to the PBS control group.
Table 61 Body Liver Kidney Spleen ISIS Dosage ALT AST BUN
weight (% weight (Rel weight (Rel weight (Rel No. (mg/kg) (U/L) (U/L) (mg/dL) baseline) % BW) % BW) %
BW) PBS n/a 25 51 37 119 100 100 100 0.6 29 57 40 123 100 103 119 0.6 26 57 32 117 93 109 110 0.6 26 42 35 114 100 103 103 0.6 30 67 38 121 91 100 123 0.6 36 63 31 118 98 103 98 Example 81: Duration of action in vivo of oligonucleotides targeting AlAT
comprising a GaINAc3 cluster The oligonucleotides listed in Table 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 listed in Table 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 AlAT protein levels were measured via ELISA (see Example 80). The results below are presented as the average percent of plasma AlAT 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 62 Plasma AlAT protein levels in mice ISIS Dosage Time point AlAT (% GalNAc 3 CM
No. (mg/kg) (days post- baseline) Cluster dose) PBS n/a nia nia 476366 100 nia nia 656326 18 GalNAc3-la Ad 678381 18 GalNAc3-3a Ad 678382 18 GalNAc3-7a Ad 678383 18 GalNAc 3-10a Ad 678384 18 GalNAc 3-13a 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 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).
Table 63 Inhibition of SRB-1 expression in vitro ISIS GalNAc IC50 SEQ
Sequence (5' to 3') Linkages CM
No. cluster (nM) ID No.
m m m Ges CesTesTes CesAdsGdsTds CdsAdsTasGasAds 353382 m m m PS n/a n/a 250 28 CdsTdsTes Ces CesTesTe 655861 GesmCesTesTesmCesAdsGasTasmCdsAdsTasGasAds GalNAc3 PS Ad 40 29 mCdsTdsTesmCesmCesTesTeoAGa1NAC3-1a -1a m m GalNAC3-3a-0,AdoGes CesTesTes CesAdsGdsTds GalNAc3 661161 m m m m PS Ad 40 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -3a m m GalNAC3-3a-0,AGes CeoTeoTeo CeoAdsGdsTds GalNAc3 do661162 m m m m PO/PS Ad 8 30 CdsAdsTdsGdsAds CdsTds Teo Ceo CesTesTe -3a 664078 GesmCesTesTesmCesAdsGasTasmCdsAdsTasGasAds GalNAc3 PS Ad 20 29 mCdsTdsTesmCesmCesTesTeoAGa1NAC3-9a -9a GalNAC3-8a-0,AdoGesmCesTesTesmCesAdsGdsTds GalNAc3 665001 PS Ad 70 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -8a GalNAe3-5a-0,AdoGesmCesTesTesmCesAdsGdsTds GalNAc3 666224 PS Ad 80 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -5a m m m G, CeoTeoTeo CesAdsGdsTds CdsAdsTdsGdsAds m m m PO/PS n/a n/a >250 28 CdsTds Teo Ceo CesTesTe GalNAe3-10a-0,AdoGesmCesTesTesmCesAdsGa I'd GalNAc3 666881 s s Ps Ad 30 30 mCdsAdsTdsGdsAdsmCdsTasTesmCesmCesTesTe -10a m m m GalNAe3-3a-0,Ges CesTesTes CesAdsGdsTds Cds GalNAc3 666904 m m m PS PO 9 28 AdsTdsGdsAds CdsTds Tes Ces CesTesTe -3a m m GalNAC3-3a-0,TdoGes CesTesTes CesAdsGdsTds GalNAc3 666924 m m m m PS Td 15 33 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -3a GalNAC3-6a-0,AdoGesmCesTesTesmCesAdsGdsTds GalNAc3 666961 PS Ad 150 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -6a GalNAC3-7a-0,AdoGesmCesTesTesmCesAdsGdsTds GalNAc3 666981 PS Ad 20 30 mCdsAdsTdsGdsAdsmCdsTdsTesmCesmCesTesTe -7a m m GalNAC3-13a-0,AGes CesTesTes CesAdsGd Ta GalNAc3 670061 m m m mdo s s Ps Ad 30 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -13a m m Ga1NAe3-3a-0,TdoG CTT CAGT GalNAc3 ds 670699 m es eo eo eo eo ds ds m m m PO/PS Td 15 33 CdsAdsTdsGdsAds CdsTdsTeo Ceo CesTesT -3a e m m Ga1NAe3-3a-0,AeoG CTT CAGT GalNAc ds 3 670700 m es eo eo eo eo ds ds m m m PO/PS Ae 30 30 CdsAdsTdsGdsAds CdsTdsTeo Ceo CesT 3a esT -m m Ga1NAe3-3a-0,Te0G CTT CAGT GalNAc ds 3 670701 m es eo eo eo eo ds ds m m m PO/PS Te 25 33 CdsAdsTdsGdsAds CdsTdsTeo Ceo CesTesT -3a e m m GalNAC3-12a-0,AdoGes CesTesTes CesAdsGds I'ds GalNAc3 671144 m m m m Ps Ad 40 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -12a m m Ga1NAc3-13.-0,AdoG CTT CAGT 7 GalNAc3 A
6'1165 m m es eo eo eo eo ds ds ds /
m m POPS d 8 30 CdsAdsTdsGdsAds CdsTdsTeo Ceo CesTesT -13a m m GalNAc3-14.-0,AdoGes CesTesTes CesAdsGasTas GalNAc3 671261 m m m m PS Ad >250 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -14a m m GalNAc3-15.-0,AdoGes CesTesTes CesAdsGdsTds GalNAc3 671262 m m m m PS Ad >250 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -15a m m GalNAc3-7.-0,AdoGes CeoTeoTeo CeoAdsGdsTds GalNAc3 673501 m m m m PO/PS Ad 30 CdsAdsTdsGdsAds CdsTdsTeo Ceo CesTesTe -7a m m GalNAc3-10.-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,AdoGes CesTesTes CesAdsGdsTds GalNAc3 675441 m m m m PS Ad 30 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -17a m m GalNAc3-18a-0,AdoGes CesTesTes CesAdsGdsTds GalNAc3 675442 m m m m PS Ad 20 CdaAdaTdaGdsAds Ca:1'as Tes Ces CesTesTe -18a 677841 GesmCesTesTesmCesAdsGasTasmCdsAdsTasGasAds GalNAc3 mCdsTdsTesmCesmCesTesTeoAdo,-GalNAc3-19a -19a Ad GesmCesTesTesmCesAdsGds-es-rdsGdsAds GalNAc3 677842 PS Ad mCdsTdsTesmCesmCesTesTeoAdo,-GalNAc3-20a -20a m m GalNAc3-23a-0,AdoGes CesTesTes CesAdsGdsTds GalNAc3 677843 m m m m PS Ad 40 CdsAdsTdsGdsAds CdsTds Tes Ces CesTesTe -23a The structure of GalNAc3-la was shown previously in Example 9, GalNAc3-3a was shown in Example 39, GalNAc3-5a was shown in Example 49, GalNAc3-6a was shown in Example 51, GalNAc3-7a was shown in Example 48, GalNAc3-8a was shown in Example 47, GalNAc3-9a was shown in Example 52, GalNAc3-10a was shown in Example 46, GalNAc3-12a was shown in Example 61, GalNAc3-13a was shown in Example 62, GalNAc3-14a was shown in Example 63, GalNAc3-15a was shown in Example 64, GalNAc3-17a was shown in Example 68, GalNAc3-18a was shown in Example 69, GalNAc3-19a was shown in Example 70, GalNAc3-20a was shown in Example 71, and GalNAc3-23a was shown in Example 76.
Example 83: Antisense inhibition in vivo by oligonucleotides targeting Factor XI comprising a GaINAc3 Cluster The oligonucleotides listed in Table 64 below were tested in a study for dose-dependent inhibition of Factor XI in mice.
Table 64 Modified oligonucleotides targeting Factor XI
ISIS,GalNAc SEQ
Sequence (5 to 3') CM
No. cluster ID No.
TesGesGesTesAesAdsTdsmCdsmCdsAdsmCdsTdsTdsTasmCdsAesGes 4041:Y71 nia nia 31 A,GesGe TeaGe0GeeTe0Ae0AdsTasmCdsmCdaAdsmCdaTasTasTdamCdsAeoGeo GalNAc3-la Ad 32 AesGesGeoAdo¨Ga1NAc3-la 663086 Ga1NAc3-3a-0,AdoTesGeoGeoTeoAeoAdsTdsmCdsmCdsAdsmCdsTds GalNAc3-3a Ad TdsTdsmCdsAeoGeoAesGesGe GalNAC3-7.-0,AdoTesGeoGeoTe.AeoAdsTdsmCdsmCdsAdsmCdsTds GalNAc3-7a Ad 40 678347 TdsTdsmCdsAeoGeoAesGesGe GalNAC3-10.-0,AdoTesGeoGeoTe0AeoAdsTdsmCdsmCdsAdsmCds 678348 GalNAc3-10a Ad 40 TdsTdsTdsmCdsAeoGeoA.GesGe GalNAc3-13.-0,AdoTesGeoGeoTe0AeoAdsTdsmCdsmCdsAdsmCds 678349 GalNAc3-13a Ad 40 TdsTdsTdsmCdsAeoGeoA.GesGe The structure of GalNAc3-la was shown previously in Example 9, GalNAc3-3a was shown in Example 39, GalNAc3-7a was shown in Example 48, GalNAc3-10a was shown in Example 46, and GalNAc3-13a was shown in Example 62.
Treatment Six to eight week old mice were each injected subcutaneously once per week at a dosage shown below, for a total of three doses, with an oligonucleotide listed below or with PBS. Each treatment group consisted of 4 animals. The mice were sacrificed 72 hours following the final dose. Factor XI liver mRNA
levels were measured using real-time PCR and normalized to cyclophilin according to standard protocols.
Liver transaminases, BUN, and bilirubin were also measured. The results below are presented as the average percent for each treatment group, normalized to the PBS control.
As illustrated in Table 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).
Table 65 Factor XI liver mRNA, liver transaminase, BUN, and bilirubin levels ISIS Dosage Factor XI ALT AST BUN Bilirubin GalNAc3 SEQ
No. (mg/kg) mRNA (% PBS) (U/L) (U/L) (mg/dL) (mg/dL) Cluster ID No.
PBS nia 100 63 70 21 0.18 nia nia 3 65 41 58 21 0.15 33 49 53 23 0.15 nia 31 30 17 43 57 22 0.14 0.7 43 90 89 21 0.16 656173 2 9 36 58 26 0.17 GalNAc 3-la 32 6 3 50 63 25 0.15 0.7 33 91 169 25 0.16 2 7 38 55 21 0.16 GalNAc3-3a 40 6 1 34 40 23 0.14 0.7 35 28 49 20 0.14 678347 2 10 180 149 21 0.18 GalNAc3-7a 40 6 1 44 76 19 0.15 0. 39 43 54 21 0.16 678348 GalNAc 3-10a 40 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 GalNAc 3-13a 40 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 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 Table 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).
Table 66 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 404071 30 10 47 n/a n/a 31 656173 6 10 3 GalNAc3-la Ad 32 663086 6 10 2 GalNAc3-3 a Ad 40 678347 6 10 1 GalNAc3-7a Ad 40 678348 6 10 1 GalNAc 3-10a Ad 678349 6 10 1 GalNAc 3-13a Ad Example 85: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising a Ga1NAc3 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 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 67 and 68, treatment with antisense oligonucleotides lowered SRB-1 mRNA
levels in a dose-dependent manner.
Table 67 SRB-1 mRNA in liver ISIS No. Dosage (mg/kg) SRB-1 mRNA (% GalNAc 3 Cluster CM
Saline) Saline nia 100 nia nia 0.1 94 0.3 119 655861 GalNAc 3-la Ad 0.1 120 0. 107 661161 GalNAc3-3a Ad 0.1 107 0. 107 666881 GalNAc 3-10a Ad 0.1 120 0. 103 666981 GalNAc3-7a Ad 0.1 670061 118 GalNAc 3-13a Ad 0.3 89 0.1 119 0.3 96 677842 GalNAc3-20a Ad Table 68 SRB-1 mRNA in liver ISIS No. Dosage (mg/kg) SRB-1 mRNA (% GalNAc3 Cluster CM
Saline) 0.1 107 0.3 95 661161 GalNAc3-3a Ad 0.1 110 0.3 88 677841 GalNAc3-19a 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 69 below.
Table 69 ISIS Dosage ALT AST Bilirubin BUN Body Weight GalNAc3 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 GalNAc3-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 GalNAc3-3a 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 GalNAc3-10a 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 GalNAc3-7a 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 GalNAc3-13a Ad 1 20 35 0.14 20 115 3 27 42 0.12 20 116 0.1 20 38 0.17 24 114 677842 0.3 31 46 0.17 21 117 GalNAc3-20a Ad 1 22 34 0.15 21 119 3 41 57 0.14 23 118 Example 86: Antisense inhibition in vivo by oligonucleotides targeting TTR
comprising a Ga1NAc3 cluster Oligonucleotides listed in Table 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 Tables 72-74.
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 71-74 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 71-74, "BL" indicates baseline, measurements that were taken just prior to the first dose.
As illustrated in Tables 71 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 70 Oligonucleotides targeting human TTR
GalNAc SEQ
Isis No. Sequence 5 to 3' Linkages CM
cluster ID
No.
TesmCesTesTesGesGdsTdsTdsAdsmCdsAdsTasGasAdsAds 420915 PS n/a n/a 41 AesTõmCesmCesmCe TesmCesTesTesGesGdsTdsTdsAdsmCdsAdsTdsGdsAdsAds 660261 PS GalNAc3-1a Ad 42 AesTesmCesmCesmCeoAdo¨GalNAc3-1.
682883 Ga1NAc3-3._0,TesmCeorreoTeoGeoGdsTdsTdsAdsmCdsAds PS/PO GalNAc3-3a PO

Tds GdsAdsAdsAeoTeomCesmCesmCe GalNAc3-7._0,TesmCeoTeoTeoGeoGasTdsTdsAdsmCdsAds 682884 PS/P0 GalNAc3-7a PO 41 Tds GdsAdsAdsAeoTeomCesmCesmCe GalNAc3-10._0,TesmCeoTeoTe. GeoGdsTdsTdsAdsmCds 682885 PS/PO GalNAc3-1 Oa PO 41 AdsTdsGdsAdsAdsAeoTeomCesmCesmCe GalNAc3-13._0,TesmCeoTeoTe. GeoGdsTdsTdsAdsmCds 682886 PS/P0 GalNAc3-13a PO 41 AdsTdsGdsAdsAdsAeoTeomCesmCesmCe TesmCeoTeoTeo Geo GdsTdsTdsAdsmCdsAdsTds GdsAdsAds 684057 PS/P0 GalNAc3-19a Ad 42 AeoTeomCesmCesmCeoAdo¨GalNAc3-19.
The legend for Table 72 can be found in Example 74. The structure of GalNAc3-1 was shown in Example 9.
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. The structure of GalNAc3-19a. was shown in Example 70.
Table 71 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 nia 100 100 nia nia 420915 20 48 65 nia nia 41 0.6 113 87 660261 GalNAc 3-la Ad 42 Table 72 Antisense inhibition of human TTR in vivo TTR Plasma TTR protein (% PBS
at BL) SEQ
Isis No. Dosage GalNAc mRNA Day 17 CM ID
((mg/kg)\ (% PBS) BL Day 3 Day 10 (After sac) cluster No.
PBS nia 100 100 96 90 114 nia nia 420915 20 43 102 66 61 58 nia nia 41 0.6 60 88 73 63 68 GalNAc3- PO

3a 0.6 56 88 78 63 67 GalNAc3- PO

7a 0.6 60 92 77 68 76 GalNAc3- PO

10a 682886 0.6 57 91 70 64 69 GalNAc3- PO 41 2 21 89 50 31 30 13a 0.6 53 80 69 56 62 -684057 2 21 92 55 34 30 GalNAc3Ad 42 6 11 82 50 18 13 19a Table 73 Transaminase levels, body weight changes, and relative organ weights Dos ALT (U/L) AST (U/L) Body Liver Spleen Kidne SEQ
age Isis No. (mg BL Day Day Day BL Day Day Day (% (% ID
/kg) 3 10 17 3 10 17 BL) PBS) PBS) PBS) No.
PBS ilia 33 34 33 24 58 62 67 52 105 100 100 100 ilia 0.6 33 38 28 26 70 71 63 59 111 96 99 92 Table 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. (mg Day Day Day Day Day Day (% y (%
ID
/kg) BL BL

10 17 BL) PBS) PBS) PBS) No.
PBS ilia 32 34 37 41 62 78 76 77 104 100 100 100 ilia 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 doses of oligonucleotides targeting TTR comprising a Ga1NAc3 cluster ISIS numbers 420915 and 660261 (see Table 70) were tested in a single dose study for duration of action in mice. ISIS numbers 420915, 682883, and 682885 (see Table 70) 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 75 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 41 660261 13.5 GalNAc 3-1 a 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 76 Plasma TTR protein levels ISIS Dosage Time pointGalNAc3 CM
TTR (% baseline) SEQ ID No.
No. (mg/kg) (days post-dose) Cluster 420915 100 10 48 n/a n/a 41 682883 10.0 10 38 GalNAc3-3a PO

682885 10.0 10 34 GalNAc 3-10a PO 41 The results in Tables 75 and 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 77 were tested for splicing modulation of human survival of motor neuron (SMN) in mice.
Table 77 Modified ASOs targeting SMN
ISISGalNAc 3 SEQ
Sequences (5' to 3') CM
No. Cluster ID
No.
ATTmCAmCTTTITATAATGITTG
387954 es es es es es es es es es es es es es es es es es es es Ge n/a n/a 43 699819 Ga1NAc3-7a-0,AesTesTesmCesAesmCesTesTesTesmCesAesTesAesA' GalNAc3-7a PO

TesGesmCesTeSGeSGe 699821 Ga1NAc3-7a-0,AesTeoTeomCeoAeomCeoTe0T.TeomCe0A.Te0A, GalNAc3-7a PO 43 A,,TeõGeõmCe.TõGesGe mC mC sõmCesAesTõAõAõTesGõmCesTõ
700000 A' ' ' esA ' ' T G' GalNAc 3-la Ad 44 GeoAdo¨Ga1NAc3-1.
703421 X-ATTmCAmCTTTmCATAATGmCTGG n/a n/a 43 703422 Ga1NAc3-7b-X-ATTmCAmCTTTmCATAATGmCTGG GalNAc3-7b n/a 43 The structure of GalNAc3-7a was shown previously in Example 48. "X" indicates a 5' primary amine generated by Gene Tools (Philomath, OR), and GalNAc3-7b indicates the structure of GalNAc3-7a lacking the ¨NH-C6-0 portion of the linker as shown below:

02,,cE1).
AcHN

HO-12-\--- 1r4 HN
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 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 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 78 Effect of oligonucleotides targeting human SMN in vivo ISIS GalNAc 3 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 43 387954 288 5.00 n/a n/a 43 699819 32 7.84 GalNAc3-7a PO 43 699821 32 7.22 GalNAc3-7a PO 43 700000 32 6.91 GalNAc 3-1 a Ad 44 703421 32 1.27 n/a n/a 43 703422 32 4.12 GalNAc3-7b n/a 43 Example 89: Antisense inhibition in vivo by oligonucleotides targeting Apolipoprotein A (Apo(a)) comprising a GaINAc3 conjugate The oligonucleotides listed in Table 79 below were tested in a study for dose-dependent inhibition of Apo(a) in transgenic mice.
Table 79 Modified ASOs targeting Apo(a) ISISGalNAc3 SEQ ID
Sequences (5' to 3') CM
No. Cluster No.
TesGesmCesTesmCesmCdsGdsTdsTdsGdsGdsTdsGdsmCds 494372 n/a n/a 53 TdsTõGesTesTesmCe GalNAc3-7.-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds 68 125 7 GalNAc3-7a 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 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 80 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 81.
As illustrated in Table 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 81, transaminase levels and body weights were unaffected by the oligonucleotides, indicating that the oligonucleotides were well tolerated.
Table 80 Apo(a) liver mRNA and plasma protein levels ISIS Dosage Apo(a) mRNA Apo(a) plasma protein (% PBS) No. (mg/kg) (% PBS) BL Week 1 Week 2 Week 3 Week 4 Week 5 Week 6 PBS n/a 100 100 120 119 113 88 121 97 0.3 75 79 76 89 98 71 94 78 Table 81 ISIS No. Dosage (mg/kg) ALT (U/L) AST (U/L) Body weight (% baseline) PBS nia 37 54 103 0.3 30 80 104 Example 90: Antisense inhibition in vivo by oligonucleotides targeting TTR
comprising a GaINAc3 cluster Oligonucleotides listed in Table 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 TTR transgenic mice were each injected subcutaneously once per week for three weeks, for a total of three doses, with an oligonucleotide and dosage listed in Table 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 standard protocols to determine liver human TTR mRNA levels. The results presented in Table 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 Table 83, 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 82 Oligonucleotides targeting human TTR
GalNAc SEQ
Isis No. Sequence 5' to 3' Linkages CM
cluster ID
No.
TesmCesTesTesGesGdsTd TdsAdsmC dsAd Td Gd Ad Ad s420915 PS nia n/a AesTesmCesmCesCe GalNAc3-3._0,TesinCeoTeoTeoGeoGdsTdsTdsAdsmCdsAds 682883 PS/P0 GalNAc3-3a PO 41 TdsGdsAdsAdsAeoTeomCesmCesmCe GalNAC3-3,-0,AdoTesmCeoTeoTeoGeoGdsTdsTdsAd 666943 s PS/P0 GalNAc3-3a Ad 45 mCdsAdsTdsGdsAdsAds AeoTeomCesmCesmCe GalNAC3-7a-0,AdoTesmCeoTeoTeoGeoGdsTdsTasAd 682887 s PS/P0 GalNAc3-7a Ad 45 mCdsAdsTdsGdsAdsAdsAeoTeoinCesinCesinCe GalNAC3-10a,,AdoTesmCeoTeoTeoGeoGdsTdsTasAd 682888 s PS/PO GalNAc3-1 Oa Ad 45 mCdsAdsTdsGdsAdsAdsAeoTeoinCesinCesinCe GalNAC3-13a_0,AdoTesmCeoTeoTeoGeoGdsTdsTasAd 682889 s PS/P0 GalNAc3-13a Ad 45 mCdsAdsTdsGdsAdsAdsAeoTeomCesmCesmCe The legend for Table 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.
Table 83 Antisense inhibition of human TTR in vivo Isis No. Dosage (mg/kg) TTR mRNA (% PBS) TTR protein (% BL) GalNAc cluster CM
PBS nia 100 124 nia nia 420915 20 71 86 nia n/a 0.6 61 73 682883 2 23 36 GalNAc3-3a PO

0.6 74 93 666943 2 33 57 GalNAc3-3a Ad 0.6 60 97 682887 2 36 49 GalNAc3-7a Ad 0.6 65 92 682888 2 32 46 GalNAc3-1 Oa Ad 0.6 72 74 682889 2 38 45 GalNAc3-13a Ad Example 91: Antisense inhibition in vivo by oligonucleotides targeting Factor VII comprising a Ga1NAc3 conjugate in non-human primates Oligonucleotides listed in Table 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 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 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 85, 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 84 Oligonucleotides targeting Factor VII
GalNAc SEQ
Isis No. Sequence 5 to 3' Linkages CM
cluster ID
No.
AesTesGesmCesAesTasGasGasTasGasAdsTdsGdsmCdsTds 407935 PS n/a n/a 46 TesmCesTesGesAe GalNAC3-10a_o'AesTesGesmCesAesTdsGdsGdsTdsGds 686892 PS GalNAc3-10a PO 46 Ado- ds GdsmC dsT ds esmCes TesGesAe The legend for Table 84 can be found in Example 74. The structure of GalNAc3-10a. was shown in Example 46.
Table 85 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 86, 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 M. 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 86 Inhibition of mouse APOC-III expression in mouse primary hepatocytes ISISÚIC50 SEQ
Sequence (5 to 3') CM
No. (nM) ID No.
440670 mCesAesGesmCesTesTdsTdsAdsTdsTdsAdsGdsGdsGdsAdsmCesAesGesmCesAe ilia
13.20 47 mCesAesGesmCesTesTdsTdsAdsTdsTdsAdsGdsGdsGdsAdsmCes 661180 Ad 1.40 48 AeaGeamCesAeo Ado, -GalNAc3-1.
GalNAC3-3a-o,mCesAesGesmCesTesTdsTdsAdsTdsTdsAdsGdsGdsGdsAdsmC
680771 es PO 0.70 47 AeaGesmCesIke GalNAC3-7a-o,mCesAesGesmCes T es ds dsAdsT ds dsA ds Gds Gds ds Ads mC
680772 es PO 1.70 47 AeaGesmCesAe GalNAc3-10a-o,mCesAesGesmCesTesTdsTdsAdsTdsTdsAdsGdsGdsGdsAdsmC
680773 es PO 2.00 47 AeaGesmCesAe GalNAc3-13a-o,mCesAesGesmCesTejdsTdsAdsTdsTdsAdsGdsGdsGdsAdsmC
680774 es PO 1.50 47 AeaGesmCesAe GalNAc3-3a-c,,mCesAeoGeomCejejdsTdsAdsTdsTdsAdsGdsGdsGdsAdsmC
681272 ee PO < 0.46 47 AeoGesmCesAe GalNAC3-3 am'AdomCesAesGesmCes TesTdsTdsAdsTdsTdsAdsGds GdsGdsAds Ad 1.10 49 es mCesAesGmCesAe mCesAesGesmCesTesTdsTdsAdsTdsTdsAdsGdsGdsGdsAdsmCes 683733 Ad 2.50 48 AesGesmCesAeoAdo,-GalNAc3-19a The structure of GalNAc3-la was shown previously in Example 9, GalNAc3-3a was shown in Example 39, GalNAc3-7a was shown in Example 48, GalNAc3-10a was shown in Example 46, GalNAc3-13a was shown in Example 62, and GalNAc3-19a was shown in Example 70.
Example 93: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising mixed wings and a 5'-GaINAc3 conjugate The oligonucleotides listed in Table 87 were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.

Table 87 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') GalNAc 3 CM SEQ
No. Cluster ID No.
449093 TksrfksmCksAdsGdsTdsmCds AdsTds Gds AdsmCdsTdsTksmCksmCk n/a n/a 699806 GaINAc3-3a-0,TksTksmCksAdsGdsTdsmCds Ads T ds GdsAdsmC ds GalNAc3-3a PO
TdsTks mCks mCk 699807 GaINAc3-7a-o,TksTksmCksAdsGdsTdsmCds AdsTds GdsAdsmCds GalNAc3-7a PO
sTks mCks mCk 699809 GaINAc3-7a-o, TksTksmCksAdsGdsTasmCds Ads Tds Gds AdsmC ds GalNAc3-7a PO
T dsT es m C es mC e 699811 GaINAc3-7a-0,TesTesmCesAdsGdsTdsmCds AdsTds GdsAdsmCds GalNAc3-7a PO
TdsTksmCksmCk 699813 GaINAc3-7a-o,TksTdsmCksAdsGdsTdsmCds Ads T ds GdsAdsmC ds GalNAc3-7a PO
sTks mCds mCk 699815 GaINAc3-7a-o,TesTksmCksAdsGdsTdsmCds Ads T ds GdsAdsmCds GalNAc3-7a PO
TdsTksmCksmCe The structure of GalNAc3-3a was shown previously in Example 39, and the structure of GalNAc3-7a was shown previously in Example 48. Subscripts: "e" indicates 2'-MOE modified nucleoside; "d" indicates f3-D-2'-deoxyribonucleoside; "k" indicates 6'-(S)-CH3 bicyclic nucleoside (cEt);
"s" indicates phosphorothioate internucleoside linkages (PS); "o" indicates phosphodiester internucleoside linkages (PO). Supersript "m"
indicates 5-methylcytosines.
Treatment Six to eight week old C57BL/6 mice (Jackson Laboratory, Bar Harbor, ME) were injected subcutaneously once at the dosage shown below with an oligonucleotide listed in Table 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 average percent of SRB-1 mRNA levels for each treatment group relative to the saline control group. As illustrated in Table 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.
Body weights, liver transaminases, total bilirubin, and BUN were also measured, and the average values for each treatment group are shown in Table 88. Body weight is shown as the average percent body weight relative to the baseline body weight (% BL) measured just prior to the oligonucleotide dose.

Table 88 SRB-1 mRNA, ALT, AST, BUN, and total bilirubin levels and body weights ISIS Dosage SRB-1 mRNA ALT AST Bil BUN Body weight No. (mg/kg) (% PBS) (U/L) (U/L) (% BL) PBS n/a 100 31 84 0.15 28 102 1 111 18 48 0.17 31 104 449093 3 94 20 43 0.15 26 103 36 19 50 0.12 29 104 0.1 114 23 58 0.13 26 107 699806 0.3 59 21 45 0.12 27 108 1 25 30 61 0.12 30 104 0.1 121 19 41 0.14 25 100 699807 0.3 73 23 56 0.13 26 105 1 24 22 69 0.14 25 102 0.1 125 23 57 0.14 26 104 699809 0.3 70 20 49 0.10 25 105 1 33 34 62 0.17 25 107 0.1 123 48 77 0.14 24 106 699811 0.3 94 20 45 0.13 25 101 1 66 57 104 0.14 24 107 0.1 95 20 58 0.13 28 104 699813 0.3 98 22 61 0.17 28 105 1 49 19 47 0.11 27 106 0.1 93 30 79 0.17 25 105 699815 0.3 64 30 61 0.12 26 105 1 24 18 41 0.14 25 106 Example 94: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising 2'-sugar modifications and a 5'-GaINAc3 conjugate The oligonucleotides listed in Table 89 were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.
Table 89 Modified ASOs targeting SRB-1 ISIS Sequences (5' to 3') GalNAc 3 CM SEQ
No. Cluster ID No.
353382 GesmCesTesTesmCesAdsGdsTdsmCdsAdsTds GdsAdsmC dsTdsTesmCesmCes n/a n/a TesTe 700989 Gins CmsUmsUmsCmsAdsGdsTdsmC dsAdsTdsGdsAdsmCds TdsUms Cms Cms n/a n/a UmsUm 666904 Ga1NAc3-3 am, GesmCesTesTesmCesAdsGdsTdsmCdsAdsTdsGdsAds GalNAc3-3a PO

mCdsTdsTesmCesmCesTesTe 700991 Ga1NAc3-7a-0,GmsCinsUinsUinsCinsAds GdsTdsmCdsAdsTdsGds GalNAc3-7a PO

AdsmCdsTdsUmsCmsCmsUmsUm Subscript "m" indicates a 2'-0-methyl modified nucleoside. See Example 74 for complete table legend. The structure of GalNAc3-3a was shown previously in Example 39, and the structure of GalNAc3-7a was shown previously in Example 48.

Treatment The study was completed using the protocol described in Example 93. Results are shown in Table 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 90 SRB-1 mRNA
ISIS No. Dosage (mg/kg) SRB-1 mRNA (% PBS) PBS nia 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 91 were tested in a dose-dependent study for antisense inhibition of SRB-1 in mice.
Table 91 Modified ASOs targeting SRB-1 ISIS, SEQ
Sequences (5' to 3') CM
No. Cluster ID
No 440762 TksmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTksmCk nia n/a 666905 GalNAe3-3.-0,TksmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTksmCk GalNAc3-3a 699782 GalNAe3-7.-0,TksmCksAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTksmCk GalNAc3-7a 699783 Ga1NAe3-3.-0,T]smClsAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsT]smCI GalNAc3-3a 653621 TiamCisAdsGdsTdsmCdsAdsTdsGdsAdsmCdsTdsTlsmCloAdo,-Ga1NAC3-1a GalNAc3-1a Ad 23 439879 TgsmCgsAdsGdsTdsmCdsAdsTa GdsAdsmCdsTdsTgsmCg nia n/a 699789 Ga1NAe3-3a-0,TgsmCgsAdsGdsTdsmCdsAdsrra GdsAdsmCdsTdsTgsmCg GalNAc3-3a 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 GalNAc3-1 a was shown previously in Example 9, the structure of GalNAc3-3a was shown previously in Example 39, and the structure of GalNAc3-7a was shown previously in Example 48.
Treatment The study was completed using the protocol described in Example 93. Results are shown in Table 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 92 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 57 targeting ApoC-III and oligonucleotides in Table 93 targeting Apo(a) were tested in an ultra-filtration assay in order to assess plasma protein binding.

Table 93 Modified oligonucleotides targeting Apo(a) ISISGalNAc3 SEQ
Sequences (5' to 3') CM
No. Cluster ID No TesGesmCesTesmCesmCdsGdsTdsTdsGdsGdsTdsGdsmCdsTdsTesGesTes 494372 n/a n/a TesmCe TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGdsTdsGdsmCdsTdsTeoGeoTes 693401 n/a n/a TesmCe GalNAc3-7.-0,TesGesmCesTesmCesmCdsGdsTdsTdsGdsGdsTdsGdsmCds GalNAc3-7a. PO 53 TdsTesGesTesTesmCe GalNAc3-7.-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGdsTdsGdsmCds GalNAc3-7a. PO

TdsTeoGeoTesTesmCe See the Example 74 for table legend. The structure of GalNAc3-7a 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 mt of 0.5% Tween 80 and centrifuged at 2000 g for minutes, then with 300 L of a 300 ug/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 57 and 93 to be used in the studies, 300 L of a 250 ng/mL solution of oligonucleotide in H20 at pH
7.4 was placed in the pre-conditioned filters and centrifuged at 2000 g for 16 minutes. The unfiltered and 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 ug/mL). An aliquot (300 L) 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 94 for the two concentrations of oligonucleotide tested (5 and 150 ug/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 lag/mL 150 i.tg/mL 5 i.tg/mL 150 i.tg/mL
5 i.tg/mL 150 i.tg/mL
304801 99.2 98.0 99.8 99.5 98.1 97.2 663083 97.8 90.9 99.3 99.3 96.5 93.0 674450 96.2 97.0 98.6 94.4 94.6 89.3 494372 94.1 89.3 98.9 97.5 97.2 93.6 693401 93.6 89.9 96.7 92.0 94.6 90.2 681251 95.4 93.9 99.1 98.2 97.8 96.1 681257 93.4 90.5 97.6 93.7 95.6 92.7 Example 97: Modified oligonucleotides targeting TTR comprising a GaINAc3 conjugate group The oligonucleotides shown in Table 95 comprising a GalNAc conjugate were designed to target TTR.
Table 95 Modified oligonucleotides targeting TTR
GalNAc3 SEQ ID
ISIS No. Sequences (5' to 3') CM
Cluster No GalNAc3-3._0,Ado Tes mCes Tes Tes Ges Gds Tds Td. Ads mCds 666941 GalNAc3-3 Ad 45 Ads Td. Gds Ads Ads Aes Tes mCes mCes mCe T mC T T G Gds Td. Td Ad mCd Ads Td Gd Ads Ad 666942 es eoeoeomeom ms s s ss s GalNAc3-1 Ad 42 Aeo Teo Ces Ces Ceo Ado-Ga1NAc3-3.
GalNAc3-3._0,Tes mCes Tes Tes Gõ Gds Td. Td. Ads mCds Ads Tds GalNAc3-3 PO 41 Gds Ads Ads Aes Tes mCes mCes mCe GalNAc3-7._0,Tes mCes Tes Tes Gõ Gds Tds Td. Ads mCds Ads Tds 682877 GalNAc3-7 PO 41 Gds Ads Ads Aes Tes mCes mCes mCe GalNAc3-10._0,Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ads GalNAc3-10 Td. Gds Ads Ads Aes Tes mCes mCes mCe GalNAc3-13._0,Tes mCes Tes Tes Ges Gds Tds Tds Ads mCds Ads GalNAc3-13 Td. Gds Ads Ads Aes Tes mCes mCes mCe GalNAC3-7a_0,Ado Tes mCes Tes Tes Ges Gds Td. Td. Ads mCds GalNAc3-7 Ad 45 Ads Td. Gds Ads Ads Aes Tes mCes mCes mCe GalNAC3-10a-o'Ado Tes mCes Tes Tes Ges Gds Tds Td. Ads mCds GalNAc3-10 Ad 45 Ads Td. Gds Ads Ads Aes Tes mCes mCes mCe GalNAC3-13a-o'Ado Tes mCes Tes Tes Ges Gds Tds Td. Ads mCds 682882 GalNAc3-13 Ad 45 Ads Td. Gds Ads Ads Aes Tes mCes mCes mCe T T T G Gd Td Td Ads mCds Ads Tds Gds Ads Ads GalNAc3-19 Ad 42 684056 es es es es es s s s Aes Tõ mCes mCes mCeo Ado-Ga1NAc3-19a The legend for Table 95 can be found in Example 74. The structure of GalNAc3-1 was shown in Example 9.
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. 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 96 and were tested for pro-inflammatory effects in an hPMBC
assay as described in Examples 23 and 24. (See Tables 17, 70, 82, and 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 70, 82, and 95. The results shown in Table 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 96 ISIS No. Emax/EC50 GalNAc3 cluster Linkages CM
353512 3630 nia PS nia 420915 802 nia PS nia 682881 1311 GalNAc3- 10 PS Ad 682888 0.26 GalNAc3- 10 PO/PS Ad 684057 1.03 GalNAc3-19 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 97 (see Table 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, Na125I, 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 -labeled de-AGP and various GalNAc-cluster containing ASOs were performed as follows. Human HepG2 cells (106 cells/me 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 g37 C with lml competition mix containing appropriate growth media with 2%
FBS, 10-8 M 1251 _ labeled de-AGP and GalNAc-cluster containing ASOs at concentrations ranging from 10-11 to 10-5 M. Non-specific binding was determined in the presence of 10-2 M GalNAc sugar. Cells were washed twice with media without FBS to remove unbound 1251 -labeled de-AGP and competitor GalNAc ASO. Cells were lysed using Qiagen's RLT buffer containing 1% 13-mercaptoethanol. Lysates were transferred to round bottom assay tubes after a brief 10 min freeze/thaw cycle and assayed on a y-counter.
Non-specific binding was subtracted before dividing 1251 protein counts by the value of the lowest GalNAc-ASO concentration counts.
The inhibition curves were fitted according to a single site competition binding equation using a nonlinear regression algorithm to calculate the binding affinities (KD's).
The results in Table 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 97 Asialoglycoprotein receptor binding assay results Oligonucleotide end to ISIS No. GalNAc conjugate which GalNAc conjugate KD (nM) is attached 661161a GalNAc3-3 5' 3.7 666881a GalNAc3-10 5' 7.6 666981 GalNAc3-7 5' 6.0 670061 GalNAc3-13 5' 7.4 655861a GalNAc3-1 3' 11.6 677841a GalNAc3-19 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 98a below were tested in a single dose study for duration of action in mice.
Table 98a Modified ASOs targeting APO(a) ISIS,GalNAc3 SEQ
Sequences (5 to 3') CM
No. Cluster ID No.
GalNAc3-7.-0,TesGesmCesTesmCesmCdsGdsTdsTdsGdsGds GalNAc3-7a PO 53 TdsGdsmCdsTdsTesGes TesTesmCe GalNAc3-7.-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds 681257 GalNAc3-7a PO 53 TdsGdsmCdsTdsTeoGe. 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 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 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 98b Apo(a) plasma protein levels ISIS N D ( mg/k Apo(a) at 72 hours Apo(a) at 1 week Apo(a) at 3 weeks o. osage g) (% BL) (% BL) (% BL) 0.3 97 108 93 1.0 85 77 57 3.0 54 49 11 10.0 23 15 4 0.3 114 138 104 681257 1.0 91 98 54 3.0 69 40 6 10.0 30 21 4 Example 101: Antisense inhibition by oligonucleotides comprising a GaINAc cluster linked via a stable moiety The oligonucleotides listed in Table 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 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 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 99 Modified oligonucleotides targeting mouse APOC-III
Dosage APOC-III
ISIS SE
N Q
Sequences (5' to 3') CM (mg/kg) mRNA (%
No.
ID
o.
PBS) inCesikesGesmCesTesTdsTdsAdsTdsrrdsAds 6 86 440670 n/a 47 GdsGdsGdsAdsmCes AesGes mCesAe 20 59 0.6 79 GalNAC3-7.-0'mCesAesGesmCesTesTdsTdsAds 2 58 TdsTdsAdsGds GdsGdsAdsmCes AesGesmCesAe 6 31 0.6 83 GaINAc3-7._s,mCesAesGesmCesTesTdsTdsAdsTds 2 73 696847 n/a (PS) 47 TdsAdsGdsGdsGdsAdsmCes AesGesmCesAe 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 23) that does not comprise a GalNAc conjugate and ISIS 655861 (see Table 23) 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 100. 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 100 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 100 Concentration in whole Concentration in Concentration in non-ISIS Dosage liver (molecules*10^
N mg/kg) 6 hepatocytes parenchymal liver cells o. ( 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 353382 20 23.6 6.6 65.6 30 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 Ga1NAc3 conjugate The oligonucleotides listed in Table 101 below were tested in a single dose study for duration of action in mice.
Table 101 Modified ASOs targeting APOC-III
ISIS Sequences (5' to 3') GalNAc3 CM
SEQ
No. Cluster ID
No.
304801 AesGesmCesTesTesmCdsTdsTdsGdsTdsmCdsmCdsAdsGdsmCdsTesTes nia 20 TesAesTe 663084 Ga1NAc3-3.-0,AdoAesGeomCeoTeoTeomCdsTdsTds GdsTdsmCds GalNAc3-3a Ad 36 m A GmCTrrTATCdsdsdsdseoe. esese 679241 AesGeomCeoTeoTeomC dsTdsTds GdsTdsmC dsmC dsAds GdsmCds Teo Teo GalNAc3-19a Ad 21 TesAõTeoAdo-Ga1NAc3-19.
The structure of GalNAc3-3a was shown in Example 39, and GalNAc3-19a. 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 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 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 58 of example 79 with the results in Table 102 below show that oligonucleotides comprising a mixture of phosphodiester and phosphorothioate internucleoside linkages exhibited increased duration of action than equivalent oligonucleotides comprising only phosphorothioate intemucleoside linkages.
Table 102 Plasma triglyceride and APOC-III protein levels in transgenic mice Time point APOC-III
ISIS Dosage (days post- protein (% Triglycerides GalNAc3 CM
(% baseline) Cluster No. (mg/kg) dose) baseline)
14 91 103 PBS nia 21 69 92 nia n/a 304801 30 21 67 81 n/a n/a 663084 10 21 23 29 GalNAc3-3a Ad GalNAc3-679241 10 21 36 34 Ad 19a Example 104: Synthesis of oligonucleotides comprising a 5'-Ga1NAc2 conjugate HN..Boc HNI-B c Boc.N _____________________________ OH H2N.,......"._.1..o 0 1.'llir H HBTU, HOBt DIEA, DMF A"- Boc.N
DCM

120 126 85% 231 I'llY

01 /...AC OAC

Ac0P-\--0...õ.õ...-',-,,,Ao 110 F DIEA
_),...
0 * AcHN F DMF

OAcr.- OAc 0 OAcOAc AcO.D..\-o...,-.-......õ11.,.
AcHN NH Ac0-0.õõ,..^.õ--..._,..k., 1 H2, Pd/C, Me0H AcHN NH
OAcr- OAc 1 2. PFPTFA, DMF OAcOAc F 40 F
AcHN AcHN F

0 83e OHOH
3' 5', 11 -.--Z--0 9 r , 1 OLIGO O-P-0-(CH2)6-NFI2 HO
1 AcHN NH
OH
1. Borate buffer, DMSO, pH 8.5, rt OH OH
________________ o-...p.\..._ 0\
2. aq. ammonia, rFi 0 HO o ..õ---...õ..,õõ.1. N 0GO
rt AcHN

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 GalNAc2 cluster portion (GalNAc2-24a) of the conjugate group GalNAc2-24 can be combined with any cleavable moiety present on the oligonucleotide to provide a variety of conjugate groups. The structure of GalNAc2-24 (GalNAc2-24a-CM) is shown below:
Ho HO
AcHN NH
H r OH

AcHN

Example 105: Synthesis of oligonucleotides comprising a Ga1NAc1-25 conjugate 0 83e 3'5'1 11 OAc OA
OLIGO j-0¨P-0¨(CH2)6-NH2 c 1 Ac0 F = F *., 0 OH
1. Borate buffer, DMSO, pH 8.5, rt AcHN
166 2 aq. ammonia, rt OH OH
A 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.

OAc H2NSOH OAAC

Ac0 ________________ PFPTFA ______________ Ac0 H
NHAc -NHAc OH TEA, Acetonitrile OA OAc tetrazole, 1-Methylimidazole, DMF

____________________________ Ac0 0 2-cyanoethyltetraisopropyl phosphorodiamidite NHAcN-N\)LN'r 'P-NT
d) LCN
Oligonucleotide OH OH
synthesis 0 ________ VW- [70 ________________________________________________ OLIGO
N " 0 AcHN H 6 The GalNAci cluster portion (GalNAci-250 of the conjugate group GalNAc 1-25 can be combined with any cleavable moiety present on the oligonucleotide to provide a variety of conjugate groups. The structure of GalNAc1-25 (GalNAci-25a-CM) is shown below:
OH OH

No gill 1 AcHN H 6 Example 106: Antisense inhibition in vivo by oligonucleotides targeting SRB-1 comprising a 5%
GalNAc2 or a 5'-GaINAc3 conjugate Oligonucleotides listed in Tables 103 and 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 103 and 104. 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 103 and 104.
Table 103 Modified oligonucleotides targeting SRB-1 Sequences (5' to 3') GalNAc Cluster No.
(mg/kg) ID No 440762 TmCksAds GdsTdsmCdsAdsTds GdsAdsmCdsTdsTks m ks Ck nia 4.7 22 GalNAc2-24.-0,AdoTksmCksAdsGdsTdsmCdsAdsTdsGdsAds 686221 GalNAc2-24a 0.39 26 mC dsTdsTksmCk GalNAC3-13 a-0,AdoTksmCksAdsGdsTdsmCdsAdsTdsGdsAds 686222 GalNAc 3-13a 0.41 26 mCdsTdsTksmCk See Example 93 for table legend. The structure of GalNAc3-13a was shown in Example 62, and the structure of GalNAc2-24a was shown in Example 104.
Table 104 Modified oligonucleotides targeting SRB-1 ISIS,ED5o SEQ
Sequences (5 to 3') GalNAc Cluster No.
(mg/kg) ID No 440762 TksmCksAds Gds-rdsmCdsAdsTds GdsAdsmCdsTdjksmCk 5 22 GalNAci-25a-0,TicsmCksAdsGasTasmCdsAdsTasGasAds 708561 GalNAci-25a 0.4 22 mC dsTdsTksmCk See Example 93 for table legend. The structure of GalNAci-25a was shown in Example 105.
The concentrations of the oligonucleotides in Tables 103 and 104 in liver were also assessed, using procedures described in Example 75. The results shown in Tables 104a and 104b below are the average total antisense oligonucleotide tissues levels for each treatment group, as measured by UV in units of lag 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 103 and 104 above.
Table 104a Liver concentrations of oligonucleotides comprising a Ga1NAc2 or Ga1NAc3 conjugate group Dosage ISIS No. [Antisense oligonucleotide] (Kg/g) GalNAc cluster CM
(mg/kg) 2 2.1 440762 7 13.1 nia nia 20 31.1 0.2 0.9 2.7 686221 0.6 GalNAc2-24a Ad 2 12.0 6 26.5 0.2 0.5 0.6 1.6 686222 GalNAc3-13a Ad 2 11.6 6 19.8 Table 104b Liver concentrations of oligonucleotides comprising a GalNAci conjugate group Dosage ISIS No. [Antisense oligonucleotide] ( g/g) GalNAc cluster CM
(mg/kg) 2 2.3 440762 7 8.9 nia nia 20 23.7 0.2 0.4 0.6 1.1 708561 2 5.9 GalNAci-25a PO
6 23.7 20 53.9 Example 107: Synthesis of oligonucleotides comprising a Ga1NAc1-26 or Ga1NAc1-27 conjugate A 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 GalNAci-26 can be combined with any cleavable moiety present on the oligonucleotide to provide a variety of conjugate groups. The structure of GalNAci-26 (GalNAci-26a-CM) is shown below:
HO OH

HOO/\/)\
AcHN
OH
In order to add the GalNAci conjugate group to the 3'-end of an oligonucleotide, the amide formed from the reaction of compounds 47 and 64 is added to a solid support using procedures described in Example 7. The oligonucleotide synthesis is then completed using procedures described in Example 9 in order to form oligonucleotide 240.
HO OH
HO
AcHN
240 _________________________ , 3' 5' 0¨L CM __________________________ Oligo _____________________________ = _____ The GalNAci cluster portion (GalNAci-27a) of the conjugate group GalNAc1-27 can be combined with any cleavable moiety present on the oligonucleotide to provide a variety of conjugate groups. The structure of GalNAc1-27 (GalNAci-27a-CM) is shown below:
HO OH
N OH
HO
AcHN
Example 108: Antisense inhibition in vivo by oligonucleotides comprising a GaINAc conjugate group targeting Apo(a) in vivo The oligonucleotides listed in Table 105 below were tested in a single dose study in mice.
Table 105 Modified ASOs targeting APO(a) ISIS GalNAc 3 SEQ
Sequences (5' to 3') CM
No. Cluster ID
No.
TesGesmCesTesmCesmCdsGdsTdsTdsGdsGdsTdsGdsmCds 494372 n/a n/a 53 TdsTesGesTesTesmCe GalNAc3-7.-0,TesGesmCesTesmCesmCdsGdsTdsTdsGdsGds GalNAc3-7a PO 53 TdsGdsmCdsTdsTesGes TesTesmCe GalNAc3-3.-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds GalNAc3-3a PO 53 TdsGdsmCdsTdsTeoGeo TesTesmCe GalNAc3-10.-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds 681256 GalNAc 3-10a PO 53 TdsGdsmCdsTdsTeoGeo TesTesmCe GalNAc3-7.-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds GalNAc3-7a PO 53 TdsGdsmCdsTdsTeoGeo TesTesmCe GaINAc3-13.-0,TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds GalNAc3-13a PO 53 TdsGdsmCdsTdsTeoGeo TesTesmCe TesGeomCeoTeomCeomCdsGdsTdsTdsGdsGds TdsGdsmCdsTdsTeoGeo 681260 GalNAc 3-19a Ad 52 TesTesmCeoAdo,-Ga1NAC3-19 The structure of GalNAc3-7a was shown in Example 48.

Treafitlent transgenic mice that eApress human Apo(a) were each injected subcutaneously .ome xvith Oligomicleotide and dosage. listed in Table 106 or with PBS.. Each treatment group. consisted of 4 animals.
Blood was drawn the (by before dosing to determine baseline levelt 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 wae measured using tin ELIS.A. The results in. 'Fable 106 are presented as the average perc=ent of plasma Apoto protein levels =fer CEWh treatment eroup, normalized to baseline levels (% BL), The =results show that the antisense oligonucleotides =reducal Apo(a) protein expression. Furthermore, tho oligonueleotides comprising a GalN.Ae conjugate group exhibited even mom:
potent reduction in Apota) =impression than the oligonucleotide that does ni-it comprise a conjugate group.
Table .106 Apo(a) plasma protein levels =Apo(a) at I week ISIS No. DoSage (itiglg) ---1.0 24 681258 1.0 22 6:81260 10 26 Example 109: Synthesis of oligonocleotides comprising a GaINAtil-28 or GaINAi)-29 conjugate HO ___________________________________________ s 34 fv` C 1 (go HO
AcHN

Oligortncleatide 241 is:.iynthesized ping procedures similar to thOse described in EXa.rople 71 to form the phosphoramidite intetmediate, followed by procedures describod in Example. 10 to synthesize the oligonneleotide.h.. GaINAe einster portion (Oa IN!kc i-2144,1 of the conjugate group GaIN,Acr28 ean combined with any cleavable maitfy present on the oligonueleotide to provide a variety of conjugate groups.
The structure of GaINAer28 (CialN,1/41-28,-CM.) is Shown below:
Mg 0 s - cm ALHN
'0H

RECTIFIED SHEET (RULE 91) ill Order tO add the UNA() conjugate group to the V-erld t.lf afil oligonucleotide, procedures similar to those. described in Example 71 are wed to form the hydroxyl intermaiiate, which is then added to.lbe solid support using prboedures described ia EXample 7, Tho oligonucicaide synthesis .is the completed mil%
procedoes described in Example 9 in order ttt, lam ()Ligon= tootide 242.
DH
HO <' 0 ,cni 242 (") '0-1 CM j [
'.BlEgo ............................................... ;
The GaINAel cluster portion KialNAc1-290 of the conjugate group GaINAci-29 can be combined with any cleavable moiety present COL the oligonueleotidc to provide a variety of conjugate groups. The structure of Ga1NAer29 (GaNAcr29õ-CM) is shown below:
PH
HD/
...,-AdiN

Example 110: 'Synthesis of olig;onucleotides comprising a GaINAc1-30 conjugate Ac0 (10Ac OA
Ac9 K, HO"--N'F--"---OTBDPS ---___--0 Ac0 N-7--) TMSOTf __ b. Ac0113DPS
AcHN

4 t 1 1 , NR5iMe0H ODMTr 2, DMIrCi AcOL 1. TBAF
3 A(.10,pyr 2. Phosphititation ' .............. . Ac0--,-.7----- 0 OTBDPS _______ .
AcHN 244 Ac0 /0DMTr ....-.-,0 1, Couple to 5"-end of ASO
Ac0 AcHN 1 2. Deorotect and purify ASO using LMT-on purification methods OH
HO /
; \
\--.\--0 5' 3' Oligo 1 AchIN ............................... , RECTIFIED SHEET (RULE 91) Oligomeleoti& 246 comprng a GaINAc;,-30 conjugate group, wherein Y is:
selected 'from S, wbstituted or unsubstituted ;,-Clo alkyl amino, substituted.amiao,ar alkenyir k' 'iv is 5ynthesized :as stiOwn above: Th iNAci diger portion (GaINAcl-30õ)ot the conjugate group GaINAer:30 can. be combined with any cleavable moiety to provide a Variety of conjugate groups.
In certain embodiments, Y :is part of the cleavable 111014. In certain embodimentfi,õ Y is part of a stable moiety, and the cleavable moiety is present on the oligonueleOtide The str tur fGa1Nikel-3% is shown below:
OH
¨0 HO
AcHN
Example 111: Synthesis of oligonueltotides comprising a GaINAc2-31 or GaINAer32 conjugate HO,, 1. DMTra DMIr0..õ
oCE Couple to 5'-end of ASO
2. Phosphitilation ----OH .
N(ij2 DWITrO" Pr Bx 1. Remove DIVIrr groups DNITrO, 0õ1 2. Couple arnicke 245 3. Deprotect and purify ASO using OKITr0-. DIVIT-on purification methods OH
HO /
AcHN o Y ¨0õ0õt5s R aigo I
QH .7-1 6 Y
HQ
HCAc114 250 Ofigonucleotide 250 comprising a CitiNAcj,31 conjugate group, wherein Y. is selected from 0, g, a SUbstituted or MISubstituted alkyl, :amino, substituted amino, azido, alkenyt or alkynyi, synthesized.
as shown above. The GaINAe,,f cluster portion (CAIN Aer3 lõ) (tithe= conjugate group GaINA.c:1-31 can be combined with any eleavable moiety to provide a variety uf conjul...,:ate groups. In certain embodiments, the Y-containing group directly adjacent to the 5'-end of the oligonucleotide is part of the cleavable moiety, hi certain embodiments, the Y-contai ning group directly adjacent tio the 5":-cnii (If the oligonueleoti4 is part a a stable moiety, and the cleavable moiety is present on the oligonucleotide. The structure of GaINAe2-31, is =shown beic,nv:

RECTIFIED SHEET (RULE 91) OH
HD ( HO

AcHN Y ¨0 0¨Ft UH 0 y .1\1"
H%311N
The synthesis of an eligenucieotide comprising a CraINAc:2;-32 conjugate is Oewn beim.
1. DIVITrCI
2. Ally! Br 3. 0s04, Nal04 1. Couple to 5"-end of ASO
HO,4 1\laBH4 DMTr0'. 2. Remove DMTr !groups 5. Phosphitilation 3. Couple amidite 245 -----¨¨--DT. Deprotect and purify AS0 ming ,µP¨NOP02 DMI-on purification methods HQ /H

AcHN
O, oligo Ky.') 0 Y

HQ.

NHA.
Oligonucleotide=252: comprising a GitiNA02-32 conjugate group, wherein Y. is selected from 0, S, substituted or utisubsthuted CC w alkyl, amino, substituted amino, azido, alkenyl oralkynyl,ìs synthmized as shown above, The GaINAc, cluster portion ON .:2) of the conjugate: group GaINA32 can he 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-containirig group diredly adjacent to the 5k)rid of-the oligonneleotide is pan of a gable moiety, and the cleavable moiety is pment on the oligonuelemicie. The structure of GaINAcz-32,ìs showri below:

RECTIFIED SHEET (RULE 91) HO
OH
AcHN
0#. 15NY
9Hy d µ\
\--/.
t HO¨ kHAc Example 112: Moded oligonucleotides comprising a GaINAci conjugate The oligonucleotides in Table .107 targeting SRB- I were synthesized with a GaINAci coniupte group in order to further test the potency of oligonucleotides comprising conjugate groups that contain one GalNAc ligand.
Table 107 SEQ
Islos Sequence (5' to 3') GANA C.M
.1) NO.
cluster 1 711461 -GaINAcr25õ,=1do Gss mCss Tõ meõ, Ad, Gds Ta, Ad, Id, GaINAct-25õ Ad 0,16 Ads MC:6 rive, mcv, T,õ
711462 GaINAe1-25.-G. "t. Ads G4s r A Adg Tth.
Gd* GaINAcr25õ PO 28 I
Ads T& Tõ "C.'õõ me. T. T.
711163 GaINAer25õ.,,,O. "Xis.. T., Tee mcõ,A. Gd, 'fat cs'C.41 A4s -fez:
GaINAcr25, PO 28 G,t, Ad, T. T. '(. L Tõ
711465 GaINAci.26,Aah utts, Tv, Tv, inCes Gds T&
A& id, GaINAC1.-26, Ad 30 Gds Ad$ Id, Its- eitet: terts, Tts Te 711466 GaINAci-26..As 'TIN To, T. atõ A& G4, T, A Gõ
GaINAcr26õ PO 28 Ad, "IC& Tds Tõ me. nr. Te 71.1467 GaINAc1269.A. iRC T. T. me. A. Gas T(N ;0C.14, A4 To, GaINAcr269 Gds Ads n'Cdsi Ico Tr.i. Tea L
711468 GaINAe1-28=Ad,, Gõ "tõT A, Gd, T.
'Ca, Att, Td, CiaINAc1-2L Ad 30 G. Ads 1A74T.Tõ, mcõ T Te 711;469 GaINAc1-28 n'eõÅ Gd, Td, IT,1õA. Td,G GaINAcr28, Ad, Tõ "C. "Cõ,, 711470 TftI Ads- Gds. Ids Tik A4,4 Td,, GaINAc f -28, PO 28 Cids Ads InCes c. o.Ts 713844 Ges T, T, Ads Cid* Tds mCds Ads Ids Git,AdsattiNT
GaINAcr27, PO 28 Tõ, rnc mT5 Tee.GaINAt1-273, 713845 G,õ To T., To me. Ad; Co& Tdi n1Cds Ad, Td, Cht., A& me d.,T
GaINAcr27, PO 28 TU3c 'fT GaINAe,-27 uS QS -=
713846 Ciõ T. To T. 'T.,* Ad. Gds Tds rir4 A..4 Ad, nre,$ Td, GaINAcr27, A4 29 nse. T. T. Adu,..GaINAel-275 713847 C r mCes Tas Cds Ads fdsCi, Ads mCa$'r,. GaINAcr295 PO 28 RECTIFIED SHEET (RULE 91) 713848 Gõ Iõ Ad, Gd, MCI'X n'CdAt 6a1NAel,29õ PO

T,4,,CaINAgt,29, 71384-9 Go 'Co. Tõ G'4, CI& Amci Tjzz GaINAct-29õ
Tõ 'C6 atc, Ado,GaINAer29, 713850 'X.:co Teo Teo 'Teo 4t(iA, (d: -A6 wCtiR GalNAel-293 Tõ, C.o To To Ade,GIIINAcr-29õ

Example 113: Modified ofigonueleotides eomprising a GaINAe conjugate gyoup targeting Hepatits B
Virus (HBV) The oligontieleotides ligted in Table 108 below were designed to target REV.
In certain embodiments; the eteaVabW rnoidy phosphodiestcr linkage, Table 108 Sequences (5' to 3') SEQ ID No.
Gal.N.Ae3-3-G:sõ't G NAG sst 3 GaINAe3-3-GtsTokberwkoCV:idl d,;(36A,IsAd(3d;nCdAsAtt,..A,,i0,õ17,,i6o 7-( 'A ,,G A., A G mC NGTG 3 _ , , õ tõ m (1 -As c!$.4- - = (?:.:

GaI
3 NAcr G.'''CxA,,,,Gt,,A,,GAidA'LtG:.t,A,-],sA,R(ja;uCitS;,-JA4kA,A13:,,,Tcsa:C,A;aINAC3't9 3 GAINAz3-25.-C-i;1Q,A,AN.,,G,j-lj.,J,G,4A A i:(1&'''CJ%Cl 3 GaINAc3-25-6,,,A;AoCe0A,,*(i&GdsTo.s-G,gektAisCid;''CdSidsAd.si'W-i,:,:;1`,,sG,,suVe 3 1.0 RECTIFIED SHEET (RULE 91)

Claims (390)

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: 2 encoding transthyretin (TTR).
2. The compound of claim 1, wherein the nucleobase sequence of the modified oligonucleotide is complementary within nucleobases 507-608 of SEQ ID NO: 2, and wherein said modified oligonucleotide is at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO:
2.
3. The compound of claim 1, wherein the nucleobase sequence of the modified oligonucleotide is complementary within nucleobases 507-526, 508-527, 515-534, 516-535, 580-599, 585-604, 587-606, or 589-608 of SEQ ID NO: 2, and wherein said modified oligonucleotide is at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 2.
4. 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: 12, 13, 14, 15, 16, 17, 18, or 19.
5. The compound of claim 4, wherein the modified oligonucleotide has a nucleobase sequence comprising the sequence recited in SEQ ID NOs: 12, 13, 14, 15, 16, 17, 18, or 19.
6. The compound of claim 4, wherein the modified oligonucleotide has a nucleobase sequence consisting of the sequence recited in SEQ ID NOs: 12, 13, 14, 15, 16, 17, 18, or 19.
7. The compound of any of claims 1 to 6, wherein the modified oligonucleotide consists of 20 linked nucleosides.
8. The compound of any of claims 1 to 7, wherein the modified oligonucleotide comprises at least one modified sugar.
9. The compound of claim 8, wherein the modified sugar is a bicyclic sugar.
10. The compound of claim 9, 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).
11. The compound of claim 8, wherein the modified sugar is 2'-O-methoxyethyl.
12. The compound of any of claims 1 to 11, wherein the modified oligonucleotide comprises at least one modified nucleobase.
13. The compound of claim 12, wherein the modified nucleobase is a 5-methylcytosine.
14. The compound of any of claims 1 to 13, comprising a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence consisting of the sequence recited in SEQ ID NOs: 12, 13, 14, 15, 16, 17, 18, or 19, wherein the modified oligonucleotide comprises a gap segment consisting of ten linked deoxynucleosides;
a 5' wing segment consisting of five linked nucleosides; and a 3' wing segment consisting of five linked nucleosides;
wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment;
wherein each nucleoside of the 5' wing segment comprises a 2'-O-methoxyethyl sugar; wherein each nucleoside of the 3' wing segment comprises a 2'-O-methoxyethyl sugar; wherein each internucleoside linkage is a phosphorothioate linkage; and wherein each cytosine is a 5-methylcytosine.
15. The compound of any of claims 1 to 14, wherein the compound is single-stranded.
16. The compound of any of claims 1 to 14, wherein the compound is double-stranded.
17. The compound of any of claims 1 to 16, wherein the modified oligonucleotide comprises at least one modified internucleoside linkage.
18. The compound of claim 17, wherein the modified internucleoside linkage is a phosphorothioate internucleoside linkage.
19. The compound of claim 18, wherein the modified oligonucleotide comprises at least one phosphodiester internucleoside linkage.
20. The compound of claim 18, wherein the modified oligonucleotide comprises at least 2 phosphodiester internucleoside linkages.
21. The compound of claim 18, wherein the modified oligonucleotide comprises at least 3 phosphodiester internucleoside linkages.
22. The compound of claim 18, wherein the modified oligonucleotide comprises at least 4 phosphodiester internucleoside linkages.
23. The compound of claim 18, wherein the modified oligonucleotide comprises at least 5 phosphodiester internucleoside linkages.
24. The compound of claim 18, wherein the modified oligonucleotide comprises at least 6 phosphodiester internucleoside linkages.
25. The compound of claim 18, wherein the modified oligonucleotide comprises at least 7 phosphodiester internucleoside linkages.
26. The compound of any of claims 19 to 25, wherein each internucleoside linkage of the modified oligonucleotide is selected from a phosphodiester internucleoside linkage and a phosphorothioate internucleoside linkage.
27. The compound of claim 18, wherein each internucleoside linkage of the modified oligonucleotide comprises is a phosphorothioate internucleoside linkage.
28. A compound consisting of ISIS 304299 and a conjugate group.
29. A compound consisting of ISIS 420915 and a conjugate group.
30. A compound consisting of ISIS 420921 and a conjugate group.
31. A compound consisting of ISIS 420922 and a conjugate group.
32. A compound consisting of ISIS 420950 and a conjugate group.
33. A compound consisting of ISIS 420955 and a conjugate group.
34. A compound consisting of ISIS 420957 and a conjugate group.
35. A compound consisting of ISIS 420959 and a conjugate group.
36. The compound of any of claims 1 to 35, wherein the conjugate group is linked to the modified oligonucleotide at the 5' end of the modified oligonucleotide.
37. The compound of any of claims 1 to 35, wherein the conjugate group is linked to the modified oligonucleotide at the 3' end of the modified oligonucleotide.
38. The compound of any of claims 1-37, wherein the conjugate group comprises exactly one ligand.
39. The compound of any of claims 1-37, wherein the conjugate group comprises exactly two ligands.
40. The compound of any of claims 1-37, wherein the conjugate group comprises three or more ligands.
41. The compound of any of claims 1-37, wherein the conjugate group comprises exactly three ligands.
42. The compound of any of claims 38-41, 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.
43. The compound of claim 42, wherein each ligand is N-acetyl galactosamine.
44. The compound of any of claims 1 to 37, wherein the conjugate group comprises:

45. The compound of any of claims 1 to 37, wherein the conjugate group comprises:
46. The compound of any of claims 1 to 37, wherein the conjugate group comprises:
47. The compound of any of claims 1 to 37, wherein the conjugate group comprises:

48. The compound of any of claims 1 to 37, wherein the conjugate group comprises:
49. The compound of any of claims 1 to 48, wherein the conjugate group comprises at least one phosphorus linking group or neutral linking group.
50. The compound of any of claims 1 to 49, 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.
51. The compound of any of claims 1 to 49, 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.
52. The compound of claim any of claims 1 to 51, 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.
53. The compound of any of claims 1 to 51, 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.
54. The compound of any of claims 1 to 53, wherein the conjugate group is covalently attached to the modified oligonucleotide.
55. The compound of any of claims 1 to 54, 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.
56. The compound of any of claims 1 to 54, 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.
57. The compound of any of claims 1 to 54, 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.
58. The compound of any of claims 1 to 54, 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.
59. The compound of any of claims 1 to 54, wherein the compound has a structure represented by the formula:
A ¨C¨( E¨F)q wherein A is the modified oligonucleotide;
C is the conjugate linker;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
60. The compound of any of claims 1 to 54, wherein the compound has a structure represented by the formula:
A¨B¨D¨(E¨F)q wherein A is the modified oligonucleotide;
B is the cleavable moiety;
D is the branching group;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
61. The compound of any of claims 1 to 54, wherein the compound has a structure represented by the formula:
A ¨B¨( E¨F)q wherein A is the modified oligonucleotide;
B is the cleavable moiety;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
62. The compound of any of claims 1 to 54, wherein the compound has a structure represented by the formula:
A¨D¨(¨E¨F)q wherein A is the modified oligonucleotide;
D is the branching group;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
63. The compound of any of claims 1 to 62, 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.
64. The compound of any of claims 1 to 62, wherein the conjugate linker has a structure selected from among:

65. The compound of any of claims 1 to 62, wherein the conjugate linker has the followingstructure:

66. The compound of any of claims 1 to 62, wherein the conjugate linker has a structure selected from among:
67. The compound of any of claims 1 to 62, wherein the conjugate linker has a structure selected from among:
68. The compound of any of claims 1 to 63, wherein the conjugate linker has a structure selected from among:
69. The compound of any of claims 1 to 63, wherein the conjugate linker comprises a pyrrolidine.
70. The compound of any of claims 1 to 64, wherein the conjugate linker does not comprise a pyrrolidine.
71. The compound of any of claims 1 to 63 or 69 to 70, wherein the conjugate linker comprises PEG.
72. The compound of any of claims 1 to 63 or 69 to 71, wherein the conjugate linker comprises an amide.
73. The compound of any of claims 1 to 63 or 69 to 72, wherein the conjugate linker comprises at least two amides.
74. The compound of any of claims 1 to 63 or 71, wherein the conjugate linker does not comprise an amide.
75. The compound of any of claims 1 to 63 or 69 to 73, wherein the conjugate linker comprises a polyamide.
76. The compound of any of claims 1 to 63 or 69 to 75, wherein the conjugate linker comprises an amine.
77. The compound of any of claims 1 to 63 or 69 to 76, wherein the conjugate linker comprises one or more disulfide bonds.
78. The compound of any of claims 1 to 63 or 69 to 77, wherein the conjugate linker comprises a protein binding moiety.
79. The compound of claim 78, wherein the protein binding moiety comprises a lipid.
80. The compound of claim 78, 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.
81. The compound of claim 78, 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.
82. The compound of any of claims 1 to 63, 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.
83. The compound of any of claims 1 to 63, wherein the conjugate linker has a structure selected from among:

wherein each n is, independently, from 1 to 20.
84. The compound of any of claims 1 to 63, wherein the conjugate linker has a structure selected from among:
85. The compound of any of claims 1 to 63, wherein the conjugate linker has a structure selected from among:
wherein n is from 1 to 20.
86. The compound of any of claims 1 to 63, wherein the conjugate linker has a structure selected from among:
87. The compound of any of claims 1 to 63, wherein the conjugate linker has a structure selected from among:
wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
88. The compound of any of claims 1 to 63, wherein the conjugate linker has the following structure:
89. The compound of any of claims 1 to 88, 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.
90. The compound of any of claims 1 to 88, 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.
91. The compound of any of claims 1 to 88, wherein the branching group has the following structure:
92. The compound of any of claims 1 to 88, wherein the branching group has the following structure:
93. The compound of any of claims 1 to 88, wherein the branching group has the following structure:

94. The compound of any of claims 1 to 88, wherein the branching group has the following structure:
95. The compound of any of claims 1 to 88, wherein the branching group comprises an ether.
96. The compound of any of claims 1 to 88, wherein the branching group has the following structure:
each n is, independently, from 1 to 20; and m is from 2 to 6.
97. The compound of any of claims 1 to 88, wherein the branching group has the following structure:
98. The compound of any of claims 1 to 88, wherein the branching group has the following structure:
99. The compound of any of claims 1 to 88, 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.
100. The compound of any of claims 1 to 88, wherein the branching group comprises:
101. The compound of any of claims 1 to 100, wherein each tether is selected from among:
wherein L is selected from a phosphorus linking group and a neutral linking group;
Z1 is C(=C)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.
102. The compound of any of claims 1 to 100, 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.
103. The compound of any of claims 1 to 100, wherein each tether is selected from among:
wherein n is from 1 to 12; and wherein m is from 1 to 12.
104. The compound of any of claims 1 to 100, wherein at least one tether comprises ethylene glycol.
105. The compound of any of claims 1 to 100 or 102, wherein at least one tether comprises an amide.
106. The compound of any of claims 1 to 100 or 102, wherein at least one tether comprises a polyamide.
107. The compound of any of claims 1 to 100 or 102, wherein at least one tether comprises an amine.
108. The compound of any of claims 1 to 100 or 102 to 107, wherein at least two tethers are different from one another.
109. The compound of any of claims 1 to 100 or 102 to 107, wherein all of the tethers are the same as one another.
110. The compound of any of claims 1 to 100, 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.
111. The compound of any of claims 1 to 100, wherein each tether is selected from among:
112. The compound of any of claims 1 to 100, wherein each tether has the following structure:

wherein each n is, independently, from 1 to 20.
113. The compound of any of claims 1 to 100, wherein each tether has the following structure:
114. The compound of any of claims 1 to 100, wherein the tether has a structure selected from among:
; wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
115. The compound of any of claims 1 to 100, wherein the tether has a structure selected from among:
116. The compound of any of claims 1 to 115, wherein the ligand is galactose.
117. The compound of any of claims 1 to 115, wherein the ligand is mannose-6-phosphate.
118. The compound of any of claims 1 to 115, wherein each ligand is selected from among:
wherein each R1 is selected from OH and NHCOOH.
119. The compound of any of claims 1 to 115, wherein each ligand is selected from among:
120. The compound of any of claims 1 to 115, wherein each ligand has the following structure:
121. The conjugated antisense compound of any of claims 1 to 115, wherein each ligand has the following structure:
122. The compound of any of claims 1 to 121, wherein the conjugate group comprises a cell-targeting moiety.
123. The compound of claim 122, wherein the conjugate group comprises a cell-targeting moiety having the following structure:
wherein each n is, independently, from 1 to 20.
124. The compound of any of claim 122, wherein the cell-targeting moiety has the following structure:
125. The compound of claim 122, wherein the cell-targeting moiety has the following structure:
wherein each n is, independently, from 1 to 20.
126. The compound of claim 122, wherein the cell-targeting moiety has the following structure:
127. The compound of claim 122, wherein the cell-targeting moiety comprises:
128. The compound of claim 122, wherein the cell-targeting moiety comprises:
129. The compound of claim 122, wherein the cell-targeting moiety has the following structure:
130. The compound of claim 122, wherein the cell-targeting moiety has the following structure:
131. The compound of claim 122, wherein the cell-targeting moiety comprises:
132. The compound of claim 122, wherein the cell-targeting moiety has the following structure:
133. The compound of claim 122, wherein the cell-targeting moiety comprises:
134. The compound of claim 122, wherein the cell-targeting moiety comprises:
135. The compound of claim 122, wherein the cell-targeting moiety comprises:
136. The compound of claim 122, wherein the cell-targeting moiety has the following structure:
The compound of claim 122, wherein the cell-targeting moiety has the following structure:
137. The compound of claim 122, wherein the cell-targeting moiety has the following structure:
138. The compound of claim 122, wherein the cell-targeting moiety has the following structure:
139. The compound of claim 122, wherein the cell-targeting moiety has the following structure:

140. The compound of claim 122, wherein the cell-targeting moiety comprises:
141. The compound of claim 122, wherein the cell-targeting moiety comprises:

142. The compound of claim 122, wherein the cell-targeting moiety comprises:
143. The compound of claim 122, wherein the cell-targeting moiety comprises:

144. The compound of claim 122, wherein the cell-targeting moiety has the following structure:
145. The compound of claim 122, wherein the cell-targeting moiety comprises:
146. The compound of claim 122, wherein the cell-targeting moiety has the following structure:

147. The compound of claim 122, 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.
148. The compound of any of claims 1 to 121, 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.
149. The compound of claim 122, 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.
150. The compound of any of claims 1 to 149, wherein the conjugate group comprises:
151. The compound of any of claims 1 to 149, wherein the conjugate group comprises:
152. The compound of any of claims 1 to 149, wherein the conjugate group comprises:
153. The compound of any of claims 1 to 149, wherein the conjugate group comprises:
154. The compound of any of claims 1 to 153, wherein the conjugate group comprises a cleavable moiety selected from among: a phosphodiester, an amide, a deoxynucleoside, or an ester.
155. The compound of any of claims 1 to 154, wherein the conjugate group comprises a phosphodiester cleavable moiety.
156. The compound of any of claims 1 to 152, 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.
157. The compound of any of claims 1 to 156, wherein the conjugate group comprises an amide cleavable moiety.
158. The compound of any of claims 1 to 156, wherein the conjugate group comprises an ester cleavable moiety.
159. The compound of any of claims 1 to 158, 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.
160. The compound of any of claims 1 to 158, 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.
161. The compound of any of claims 1 to 158, 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.
162. The compound of any of claims 1 to 158, 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.
163. The compound of any of claims 1 to 158, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide;and Bx is a heterocyclic base moiety.
164. The compound of any of claims 1 to 158, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide;and Bx is a heterocyclic base moiety.
165. The compound of any of claims 1 to 158, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
166. The compound of any of claims 1 to 158, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
167. The compound of any of claims 1 to 158, wherein the compound has the following structure:
wherein Q13 is H or O(CH2)2-OCH3;
A is the modified oligonucleotide; and Bx is a heterocyclic base moiety.
168. The compound of any of claims 1 to 158, 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.
169. The compound of any of claims 1 to 158, 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.
170. The compound of any of claims 1 to 158, 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.
171. The compound of any of claims 1 to 158, 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.
172. The compound of any of claims 1 to 158, 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.
173. The compound of any of claims 1 to 158, 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.
174. The compound of any of claims 1 to 158, 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.
175. The compound of any of claims 1 to 158, 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.
176. The compound of any of claims 1 to 158, 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.
177. The compound of any of claims 159 to 176, wherein Bx is selected from among from adenine, guanine, thymine, uracil, or cytosine, or 5-methyl cytosine.
178. The compound of any of claims 159 to 177, wherein Bx is adenine.
179. The compound of any of claims 159 to 177, wherein Bx is thymine.
180. The compound of any of claims 159 to 176, wherein Q13 is O(CH2)2-OCH3.
181. The compound of any of claims 159 to 176, wherein Q13 is H.
182. A composition comprising the compound of any of claims 1-181 or salt thereof and at least one of a pharmaceutically acceptable carrier or diluent.
183. A prodrug comprising the compound of any of claims 1 to 181.
184. A method comprising administering to an animal the compound of any of claims 1-181, the composition of claim 182, or the prodrug of claim 183.
185. The method of claim 184, wherein the animal is a human.
186. The method of claim 184 or 185, wherein administering the compound prevents, treats, ameliorates, or slows progression of transthyretin amyloidosis.
187. The method of any of claims 184 to 186, comprising co-administering the compound or composition and a second agent.
188. The method of claim 187, wherein the compound or composition and the second agent are administered concomitantly.
189. The method of any of claims 184 to 188, wherein the administering is to the choroid plexus.
190. A method of reducing transthyretin mRNA or protein expression in an animal comprising administering to the animal the compound of any of claims 1-181, the composition of claim 182, or the prodrug of claim 183, thereby reducing transthyretin mRNA or protein expression in the animal.
191. The method of claim 190, wherein the animal is a human.
192. The method of claim 190 or 191, wherein reducing transthyretin mRNA or protein expression prevents, treats, ameliorates, or slows progression of transthyretin amyloidosis.
193. The method of any of claims 190 to 192, comprising co-administering the compound or composition and a second agent.
194. The method of claim 193, wherein the compound or composition and the second agent are administered concomitantly.
195. The method of any of claims 190 to 194, wherein the compound or composition is administered to the choroid plexus.
196. A method of treating transthyretin amyloidosis in a subject comprising administering to the subject a therapeutically effective amount of the compound of any of claims 1-181, the composition of claim 182, or the prodrug of claim 183.
197. The method of claim 196, wherein administering the compound or composition reduces at least one symptom associated with transthyretin amyloidosis selected from the group consisting of restlessness, lack of coordination, nystagmus, spastic paraparesis, lack of muscle coordination, impaired vision, insomnia, unusual sensations, myoclonus, blindness, loss of speech, Carpal tunnel syndrome, seizures, subarachnoid hemorrhages, stroke and bleeding in the brain, hydrocephalus, ataxia, and spastic paralysis, coma, sensory neuropathy, parathesia, hypesthesia, motor neuropathy, autonomic neuropathy, orthostatic hypotension, cyclic constipation, cyclic diarrhea, nausea, vomiting, reduced sweating, impotence, delayed gastric emptying, urinary retention, urinary incontinence, progressive cardiopathy, fatigue, shortness of breath, weight loss, lack of appetite, numbness, tingling, weakness, enlarged tongue, nephrotic syndrome, congestive heart failure, dyspnea on exertion, peripheral edema, arrhythmias, palpitations, light-headedness, syncope, postural hypotension, peripheral nerve problems, sensory motor impairment, lower limb neuropathy, upper limb neuropathy, hyperalgesia, altered temperature sensation, lower extremity weakness, cachexia, peripheral edema, hepatomegaly, purpura, diastolic dysfunction, premature ventricular contractions, cranial neuropathy, diminished deep tendon reflexes, amyloid deposits in the corpus vitreum, vitreous opacity, dry eyes, glaucoma, scalloped appearance in the pupils, and swelling of the feet due to water retention.
198. The method of claim 196 or 197, comprising co-administering the compound or composition and a second agent.
199. The method of claim 198, wherein the compound or composition and the second agent are administered concomitantly.
200. The method of any of claims 196 to 199, wherein the compound or composition is administered to the choroid plexus.
201. The method of any of claims 196 to 200, wherein the subject is a human.
202. 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 encoding hepatitis B virus (HBV).
203. The compound of claim 202, wherein the nucleobase sequence of the modified oligonucleotide is complementary within nucleobases 1583-1602, 1780-1799, 411-427, 1266-1285, 1577-1596, 1585-1604, 1583-1598, 1264-1279, or 1780-1797 of SEQ ID NO: 1, and wherein said modified oligonucleotide is at least 85%, 90%, 95%, or 100% complementary to SEQ ID NO: 1.
204. The compound of claim 202, 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: 3, 4, 5, 6, 7, 8, 9, 10, or 11.
205. The compound of claim 204, wherein the modified oligonucleotide has a nucleobase sequence comprising the sequence recited in SEQ ID NOs: 3, 4, 5, 6, 7, 8, 9, 10, or 11.
206. The compound of claim 204, wherein the modified oligonucleotide has a nucleobase sequence consisting of the sequence recited in SEQ ID NOs: 3, 4, 5, 6, 7, 8, 9, 10, or 11.
207. The compound of any of claims 202 to 206 wherein the modified oligonucleotide comprises a gap segment consisting of linked deoxynucleosides;
a 5' wing segment consisting of linked nucleosides; and a 3' wing segment consisting of linked nucleosides;
wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment;
wherein each nucleoside of the 5' wing segment comprises a 2'-O-methoxyethyl sugar; wherein each nucleoside of the 3' wing segment comprises a 2'-O-methoxyethyl sugar; wherein each internucleoside linkage is a phosphorothioate linkage; and wherein each cytosine is a 5-methylcytosine.
208. The compound of any of claims 202 to 207 wherein the modified oligonucleotide comprises a gap segment consisting of linked deoxynucleosides;
a 5' wing segment consisting of linked nucleosides; and a 3' wing segment consisting of linked nucleosides;
wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment;
wherein each nucleoside of the 5' wing segment comprises a 2'-O-methoxyethyl sugar or constrained ethyl sugar; wherein each nucleoside of the 3' wing segment comprises a 2'-O-methoxyethyl sugar or constrained ethyl sugar; wherein each internucleoside linkage is a phosphorothioate linkage; and wherein each cytosine is a 5-methylcytosine.
209. The compound of any of claims 202 to 208, wherein the compound is single-stranded.
210. The compound of any of claims 202 to 208, wherein the compound is double-stranded.
211. The compound of any of claims 202 to 210, wherein the modified oligonucleotide comprises at least one modified internucleoside linkage.
212. The compound of claim 211, wherein the modified internucleoside linkage is a phosphorothioate internucleoside linkage.
213. The compound of claim 212, wherein the modified oligonucleotide comprises at least one phosphodiester internucleoside linkage.
214. The compound of claim 212, wherein the modified oligonucleotide comprises at least 2 phosphodiester internucleoside linkages.
215. The compound of claim 212, wherein the modified oligonucleotide comprises at least 3 phosphodiester internucleoside linkages.
216. The compound of claim 212, wherein the modified oligonucleotide comprises at least 4 phosphodiester internucleoside linkages.
217. The compound of claim 212, wherein the modified oligonucleotide comprises at least 5 phosphodiester internucleoside linkages.
218. The compound of claim 212, wherein the modified oligonucleotide comprises at least 6 phosphodiester internucleoside linkages.
219. The compound of claim 212, wherein the modified oligonucleotide comprises at least 7 phosphodiester internucleoside linkages.
220. The compound of any of claims 213 to 219, wherein each internucleoside linkage of the modified oligonucleotide is selected from a phosphodiester internucleoside linkage and a phosphorothioate internucleoside linkage.
221. The compound of claim 212, wherein each internucleoside linkage of the modified oligonucleotide comprises is a phosphorothioate internucleoside linkage.
222. A compound consisting of ISIS 505358 and a conjugate group.
223. A compound consisting of ISIS 509934 and a conjugate group.
224. A compound consisting of ISIS 510100 and a conjugate group.
225. A compound consisting of ISIS 552023 and a conjugate group.
226. A compound consisting of ISIS 552024 and a conjugate group.
227. A compound consisting of ISIS 552032 and a conjugate group.
228. A compound consisting of ISIS 552859 and a conjugate group.
229. A compound consisting of ISIS 552925 and a conjugate group.
230. A compound consisting of ISIS 577119 and a conjugate group.
231. The compound of any of claims 202 to 230, wherein the conjugate group is linked to the modified oligonucleotide at the 5' end of the modified oligonucleotide.
232. The compound of any of claims 202 to 230, wherein the conjugate group is linked to the modified oligonucleotide at the 3' end of the modified oligonucleotide.
233. The compound of any of claims 202-232, wherein the conjugate group comprises exactly one ligand.
234. The compound of any of claims 202-232, wherein the conjugate group comprises exactly two ligands.
235. The compound of any of claims 202-232, wherein the conjugate group comprises three or more ligands.
236. The compound of any of claims 202-232, wherein the conjugate group comprises exactly three ligands.
237. The compound of any of claims 202-236, 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.
238. The compound of claim 237, wherein each ligand is N-acetyl galactosamine.
239. The compound of any of claims 202 to 238, wherein the conjugate group comprises:

240. The compound of any of claims 202 to 238, wherein the conjugate group comprises:
241. The compound of any of claims 202 to 238, wherein the conjugate group comprises:
242. The compound of any of claims 202 to 238, wherein the conjugate group comprises:

243. The compound of any of claims 202 to 238, wherein the conjugate group comprises:
244. The compound of any of claims 202 to 243, wherein the conjugate group comprises at least one phosphorus linking group or neutral linking group.
245. The compound of any of claims 202 to 244, 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.
246. The compound of any of claims 202 to 245, 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.
247. The compound of any of claims 202 to 246, 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.
248. The compound of any of claims 202 to 247, 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.
249. The compound of any of claims 202 to 248, wherein the conjugate group is covalently attached to the modified oligonucleotide.
250. The compound of any of claims 202 to 249, wherein the compound has a structure represented by the formula:
A-B-C-D~E-F)q wherein A is the modified oligonucleotide;
B is the cleavable moiety C is the conjugate linker D is the branching group each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
251. The compound of any of claims 202 to 249, 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.
252. The compound of any of claims 202 to 249, wherein the compound has a structure represented by the formula:
A-B-C~E-F)q wherein A is the modified oligonucleotide;
B is the cleavable moiety;
C is the conjugate linker;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
253. The compound of any of claims 202 to 249, wherein the compound has a structure represented by the formula:
A-C-D~E-F)q wherein A is the modified oligonucleotide;
C is the conjugate linker;
D is the branching group;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
254. The compound of any of claims 202 to 249, wherein the compound has a structure represented by the formula:
A ¨C¨( E¨F)q wherein A is the modified oligonucleotide;
C is the conjugate linker;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
255. The compound of any of claims 202 to 249, wherein the compound has a structure represented by the formula:
A¨B¨D¨(E¨F)q wherein A is the modified oligonucleotide;
B is the cleavable moiety;
D is the branching group;
each E is a tether;
each F is a ligand; and q is an integer between 1 and 5.
256. The compound of any of claims 202 to 249, 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.
257. The compound of any of claims 202 to 249, 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.
258. The compound of any of claims 202 to 257, 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.
259. The compound of any of claims 202 to 257, wherein the conjugate linker has a structure selected from among:

260.
The compound of any of claims 202 to 257, wherein the conjugate linker has the followingstructure:

261. The compound of any of claims 202 to 257, wherein the conjugate linker has a structure selected from among:
262. The compound of any of claims 202 to 257, wherein the conjugate linker has a structure selected from among:
263. The compound of any of claims 202 to 257, wherein the conjugate linker has a structure selected from among:
264. The compound of any of claims 202 to 263, wherein the conjugate linker comprises a pyrrolidine.
265. The compound of any of claims 202 to 263, wherein the conjugate linker does not comprise a pyrrolidine.
266. The compound of any of claims 202 to 265, wherein the conjugate linker comprises PEG.
267. The compound of any of claims 202 to 266, wherein the conjugate linker comprises an amide.
268. The compound of any of claims 202 to 266, wherein the conjugate linker comprises at least two amides.
269. The compound of any of claims 202 to 266, wherein the conjugate linker does not comprise an amide.
270. The compound of any of claims 202 to 269, wherein the conjugate linker comprises a polyamide.
271. The compound of any of claims 202 to 270, wherein the conjugate linker comprises an amine.
272. The compound of any of claims 202 to 271, wherein the conjugate linker comprises one or more disulfide bonds.
273. The compound of any of claims 202 to 272, wherein the conjugate linker comprises a protein binding moiety.
274. The compound of claim 273, wherein the protein binding moiety comprises a lipid.
275. The compound of claim 273, 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.
276. The compound of claim 273, 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.
277. The compound of any of claims 202 to 276, 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.
278. The compound of any of claims 202 to 277, wherein the conjugate linker has a structure selected from among:

wherein each n is, independently, from 1 to 20.
279. The compound of any of claims 202 to 277, wherein the conjugate linker has a structure selected from among:

280. The compound of any of claims 202 to 277, wherein the conjugate linker has a structure selected from among:
wherein n is from 1 to 20.
281. The compound of any of claims 202 to 277, wherein the conjugate linker has a structure selected from among:
282. The compound of any of claims 202 to 277, wherein the conjugate linker has a structure selected from among:
wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
283. The compound of any of claims 202 to 277, wherein the conjugate linker has the following structure:
284. The compound of any of claims 202 to 283, 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.
285. The compound of any of claims 202 to 283, wherein the branching group has one of the following structures:
wherein each A1 is independently, 0, S, C=O or NH; and each n is, independently, from 1 to 20.
286. The compound of any of claims 202 to 283, wherein the branching group has the following structure:
287. The compound of any of claims 202 to 283, wherein the branching group has the following structure:
288. The compound of any of claims 202 to 283, wherein the branching group has the following structure:
289. The compound of any of claims 202 to 283, wherein the branching group has the following structure:
290. The compound of any of claims 202 to 283, wherein the branching group comprises an ether.
291.
The compound of any of claims 202 to 283, wherein the branching group has the following structure:
each n is, independently, from 1 to 20; and m is from 2 to 6.
292. The compound of any of claims 202 to 283, wherein the branching group has the following structure:
293. The compound of any of claims 202 to 283, wherein the branching group has the following structure:
294. The compound of any of claims 202 to 283, 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.
295. The compound of any of claims 202 to 283, wherein the branching group comprises:
296. The compound of any of claims 202 to 295, 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.
297. The compound of any of claims 202 to 295, 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.
298. The compound of any of claims 202 to 295, wherein each tether is selected from among:
wherein n is from 1 to 12; and wherein m is from 1 to 12.
299. The compound of any of claims 202 to 295, wherein at least one tether comprises ethylene glycol.
300. The compound of any of claims 202 to 295 or 297, wherein at least one tether comprises an amide.
301. The compound of any of claims 202 to 295 or 297, wherein at least one tether comprises a polyamide.
302. The compound of any of claims 202 to 295 or 297, wherein at least one tether comprises an amine.
303. The compound of any of claims 202 to 295 or 297, wherein at least two tethers are different from one another.
304. The compound of any of claims 202 to 295 or 297, wherein all of the tethers are the same as one another.
305. The compound of any of claims 202 to 304, 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.
306. The compound of any of claims 202 to 304, wherein each tether is selected from among:
307. The compound of any of claims 202 to 304, wherein each tether has the following structure:

wherein each n is, independently, from 1 to 20.
308. The compound of any of claims 202 to 304, wherein each tether has the following structure:
309. The compound of any of claims 202 to 304, wherein the tether has a structure selected from among:
; wherein each n is independently, 0, 1, 2, 3, 4, 5, 6, or 7.
310. The compound of any of claims 202 to 304, wherein the tether has a structure selected from among:
311. The compound of any of claims 202 to 310, wherein the ligand is galactose.
312. The compound of any of claims 202 to 310, wherein the ligand is mannose-6-phosphate.
313. The compound of any of claims 202 to 310, wherein each ligand is selected from among:
wherein each R1 is selected from OH and NHCOOH.
314. The compound of any of claims 202 to 310, wherein each ligand is selected from among:
315. The compound of any of claims 202 to 310, wherein each ligand has the following structure:
316. The conjugated antisense compound of any of claims 202 to 310, wherein each ligand has the following structure:
317. The compound of any of claims 202 to 317, wherein the conjugate group comprises a cell-targeting moiety.
318. The compound of claim 317, wherein the conjugate group comprises a cell-targeting moiety having the following structure:
wherein each n is, independently, from 1 to 20.
319. The compound of claim 317, wherein the cell-targeting moiety has the following structure:
320. The compound of claim 317, wherein the cell-targeting moiety has the following structure:
wherein each n is, independently, from 1 to 20.
321. The compound of claim 317, wherein the cell-targeting moiety has the following structure:
322. The compound of claim 317, wherein the cell-targeting moiety comprises:
323. The compound of claim 317, wherein the cell-targeting moiety comprises:
324. The compound of claim 317, wherein the cell-targeting moiety has the following structure:
325. The compound of claim 317, wherein the cell-targeting moiety has the following structure:
326. The compound of claim 37, wherein the cell-targeting moiety comprises:
327. The compound of claim 317, wherein the cell-targeting moiety has the following structure:
328. The compound of claim 317, wherein the cell-targeting moiety comprises:
329. The compound of claim 317, wherein the cell-targeting moiety comprises:
330. The compound of claim 317, wherein the cell-targeting moiety comprises:
331. The compound of claim 317, wherein the cell-targeting moiety has the following structure:
The compound of claim 317, wherein the cell-targeting moiety has the following structure:
332. The compound of claim 317, wherein the cell-targeting moiety has the following structure:
333. The compound of claim 317, wherein the cell-targeting moiety has the following structure:
334. The compound of claim 317, wherein the cell-targeting moiety has the following structure:

335. The compound of claim 317, wherein the cell-targeting moiety comprises:
336. The compound of claim 317, wherein the cell-targeting moiety comprises:

337. The compound of claim 317, wherein the cell-targeting moiety comprises:
338. The compound of claim 317, wherein the cell-targeting moiety comprises:

339. The compound of claim 317, wherein the cell-targeting moiety has the following structure:
340. The compound of claim 317, wherein the cell-targeting moiety comprises:
341. The compound of claim 317, wherein the cell-targeting moiety has the following structure:

342. The compound of claim 317, 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.
343. The compound of any of claims 202 to 317, 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.
344. The compound of claim 317, 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.
345. The compound of any of claims 202 to 317, wherein the conjugate group comprises:
346. The compound of any of claims 202 to 317, wherein the conjugate group comprises:
347. The compound of any of claims 202 to 317, wherein the conjugate group comprises:
348. The compound of any of claims 202-317, wherein the conjugate group comprises:
349. The compound of any of claims 202 to 348, wherein the conjugate group comprises a cleavable moiety selected from among: a phosphodiester, an amide, a deoxynucleoside, or an ester.
350. The compound of any of claims 202 to 348, wherein the conjugate group comprises a phosphodiester cleavable moiety.
351. The compound of any of claims 202 to 348, 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.
352. The compound of any of claims 202 to 351, wherein the conjugate group comprises an amide cleavable moiety.
353. The compound of any of claims 202 to 351, wherein the conjugate group comprises an ester cleavable moiety.
354. The compound of any of claims 202 to 353, 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.
355. The compound of any of claims 202 to 353, 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.
356. The compound of any of claims 202 to 353, 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.
357. The compound of any of claims 202 to 353, 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.
358. The compound of any of claims 202 to 353, 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.
359. The compound of any of claims 202 to 353, 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.
360. The compound of any of claims 202 to 353, 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.
361. The compound of any of claims 202 to 353, 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.
362. The compound of any of claims 202 to 353, 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.
363. The compound of any of claims 202 to 353, 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.
364. The compound of any of claims 202 to 353, 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.
365. The compound of any of claims 202 to 353, 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.
366. The compound of any of claims 202 to 353, 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.
367. The compound of any of claims 202 to 353, 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.
368. The compound of any of claims 202 to 353, 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.
369. The compound of any of claims 202 to 353, 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.
370. The compound of any of claims 202 to 353, 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.
371. The compound of any of claims 202 to 353, 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.
372. The compound of any of claims 354 to 371, wherein B x is selected from among from adenine, guanine, thymine, uracil, or cytosine, or 5-methyl cytosine.
373. The compound of any of claims 354 to 372, wherein B x is adenine.
374. The compound of any of claims 354 to 372, wherein B x is thymine.
375. The compound of any of claims 354 to 371, wherein Q13 is O(CH2)2-OCH3.
376. The compound of any of claims 354 to 371, wherein Q13 is H.
377. A composition comprising the compound of any of claims 202 to 376 or salt thereof and at least one of a pharmaceutically acceptable carrier or diluent.
378. A prodrug comprising the compound of any of claims 202 to 376.
379. A method of treating a HBV-related disease, disorder or condition in a subject comprising administering the compound of any of claims 202 to 376, the composition of claim 377, or the prodrug of claim 378 to the subject, wherein the disease, disorder or condition is jaundice, liver inflammation, liver fibrosis, inflammation, liver cirrhosis, liver failure, liver cancer, diffuse hepatocellular inflammatory disease, hemophagocytic syndrome, serum hepatitis, HBV viremia, or liver disease-related transplantation.
380. A method of reducing HBV antigen levels in a subject infected with HBV
comprising administering the compound of any of claims 202 to 376, the composition of claim 377, or the prodrug of claim 378 to the subject, thereby reducing HBV antigen levels in the subject.
381. The method of claim 380, wherein the HBV antigen is HBsAG.
382. The method of claim 380, wherein the HBV antigen is HBeAG.
383. A compound comprising the following structure:

wherein X is a conjugate group comprising GalNAc.
384. A compound comprising the following structure:

385. A compound comprising the following structure:

386. A compound comprising the following structure:

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

and Z is selected from S- and O-.
387. A compound comprising the following structure:
wherein X is a conjugate group comprising GalNAc.
388. A compound comprising the following structure:

389. A compound comprising the following structure:

390. A compound comprising the following structure:

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

and Z is selected from S- and O-.
CA2921167A 2013-05-01 2014-05-01 Compositions and methods for modulating hbv and ttr expression Pending CA2921167A1 (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/US2014/036463 WO2014179627A2 (en) 2013-05-01 2014-05-01 Compositions and methods for modulating hbv and ttr expression

Publications (1)

Publication Number Publication Date
CA2921167A1 true CA2921167A1 (en) 2014-11-06

Family

ID=51843959

Family Applications (5)

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

Family Applications Before (4)

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

Country Status (35)

Country Link
US (23) US9127276B2 (en)
EP (11) EP4155403A1 (en)
JP (20) JP6456362B2 (en)
KR (16) KR102138781B1 (en)
CN (12) CN105392488B (en)
AU (19) AU2014259755B2 (en)
BR (5) BR112015027322A8 (en)
CA (5) CA2921509A1 (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) ES2730015T3 (en)
HK (5) HK1221485A1 (en)
HR (2) HRP20190987T1 (en)
HU (2) HUE043697T2 (en)
IL (18) IL296543A (en)
LT (2) LT2992009T (en)
ME (1) ME03390B (en)
MX (11) MX2015015263A (en)
MY (2) MY198359A (en)
NZ (6) NZ631512A (en)
PE (2) PE20152002A1 (en)
PH (3) PH12015502493B1 (en)
PL (2) PL2992009T3 (en)
PT (3) PT2992009T (en)
RS (2) RS60796B1 (en)
RU (8) RU2699985C2 (en)
SG (5) SG10201906382QA (en)
SI (2) SI2992009T1 (en)
UA (2) UA121017C2 (en)
WO (5) WO2014179625A1 (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
KR101979134B1 (en) 2008-10-15 2019-05-15 아이오니스 파마수티컬즈, 인코포레이티드 Modulation of factor 11 expression
ES2616051T3 (en) 2008-12-02 2017-06-09 Wave Life Sciences Japan, Inc. Method for the synthesis of modified nucleic acids in the phosphorus atom
AU2010270714B2 (en) 2009-07-06 2015-08-13 Wave Life Sciences Ltd. Novel nucleic acid prodrugs and methods use thereof
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
CA2817960C (en) 2010-11-17 2020-06-09 Isis Pharmaceuticals, Inc. Modulation of alpha synuclein expression
EP2734208B1 (en) 2011-07-19 2017-03-01 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
DK2751270T3 (en) 2011-08-29 2018-10-29 Ionis Pharmaceuticals Inc OLIGOMER-CONJUGATE COMPLEXES AND THEIR USE
JP6492003B2 (en) 2012-03-30 2019-03-27 ワシントン・ユニバーシティWashington University Methods of modulating tau expression to reduce stroke and to modify neurodegenerative syndrome
EP3336189A1 (en) * 2012-04-20 2018-06-20 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
US20160002624A1 (en) 2012-05-17 2016-01-07 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions
US9574193B2 (en) 2012-05-17 2017-02-21 Ionis Pharmaceuticals, Inc. Methods and compositions for modulating apolipoprotein (a) expression
NZ631071A (en) * 2012-05-24 2017-02-24 Ionis Pharmaceuticals Inc Methods and compositions for modulating apolipoprotein(a) expression
MX2015000577A (en) 2012-07-13 2015-08-14 Wave Life Sciences Pte Ltd Chiral control.
CN107011400B (en) 2012-07-13 2021-05-25 波涛生命科学有限公司 Asymmetric auxiliary group
JP6246121B2 (en) 2012-07-13 2017-12-13 株式会社新日本科学 Chiral nucleic acid adjuvant
EP2920304B1 (en) 2012-11-15 2019-03-06 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugates
DK2951305T3 (en) * 2013-01-30 2018-10-29 Hoffmann La Roche LNA oligonucleotide KULHYDRATKONJUGATER
AU2014216137B2 (en) 2013-02-14 2018-05-10 Ionis 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
JP6649248B2 (en) 2013-05-01 2020-02-19 レグルス セラピューティクス インコーポレイテッド Compounds and methods for enhancing cellular uptake
NZ631512A (en) 2013-05-01 2016-10-28 Ionis Pharmaceuticals Inc Compositions and methods for modulating apolipoprotein (a) expression
MY183049A (en) 2013-06-27 2021-02-09 Roche Innovation Ct Copenhagen As Antisense oligomers and conjugates targeting pcsk9
WO2015006740A2 (en) * 2013-07-11 2015-01-15 Alnylam Pharmaceuticals, Inc. Oligonucleotide-ligand conjugates and process for their preparation
JP6617702B2 (en) 2013-07-15 2019-12-11 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア FTY720 azacyclic constraint analog
TWI657819B (en) 2013-07-19 2019-05-01 美商Ionis製藥公司 Compositions for modulating tau expression
US9943604B2 (en) 2013-09-20 2018-04-17 Ionis Pharmaceuticals, Inc. Targeted therapeutic nucleosides and their use
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
EP3060664B1 (en) 2013-10-25 2021-07-07 Sanofi Microrna compounds and methods for modulating mir-21 activity
US20160289677A1 (en) * 2013-11-14 2016-10-06 Roche Innovation Center Copenhagen A/S APOB Antisense Conjugate Compounds
JP6482475B2 (en) * 2014-01-07 2019-03-13 レナセラピューティクス株式会社 Double stranded oligonucleotides comprising antisense oligonucleotides and sugar derivatives
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
KR102423317B1 (en) 2014-01-16 2022-07-22 웨이브 라이프 사이언시스 리미티드 Chiral design
EA201691587A1 (en) 2014-02-11 2017-01-30 Элнилэм Фармасьютикалз, Инк. COMPOSITIONS BASED ON iRNA FOR KETOGEXOKINASE (KHK) AND METHODS OF THEIR APPLICATION
CN116970607A (en) 2014-03-19 2023-10-31 Ionis制药公司 Compositions for modulating ataxin 2 expression
WO2015143245A1 (en) 2014-03-19 2015-09-24 Isis Pharmaceuticals, Inc. Methods for modulating ataxin 2 expression
EP3757214B1 (en) 2014-04-01 2022-06-15 Biogen MA Inc. Compositions for modulating sod-1 expression
RU2703411C2 (en) 2014-05-01 2019-10-16 Ионис Фармасьютикалз, Инк. Compositions and methods for modulating pkk expression
KR20200102553A (en) 2014-05-01 2020-08-31 아이오니스 파마수티컬즈, 인코포레이티드 Compositions and methods for modulating growth hormone receptor expression
EP3608406B1 (en) * 2014-05-01 2023-02-15 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating complement factor b expression
EP3137115B1 (en) * 2014-05-01 2020-10-14 Ionis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
PT3137605T (en) 2014-05-01 2020-12-18 Ionis Pharmaceuticals Inc Compositions and methods for modulating angiopoietin-like 3 expression
GB201408623D0 (en) * 2014-05-15 2014-07-02 Santaris Pharma As Oligomers and oligomer conjugates
SG11201609376SA (en) 2014-05-22 2016-12-29 Alnylam Pharmaceuticals Inc Angiotensinogen (agt) irna compositions and methods of use thereof
GB201410693D0 (en) 2014-06-16 2014-07-30 Univ Southampton Splicing modulation
AU2015301057A1 (en) 2014-08-07 2017-01-19 Regulus Therapeutics Inc. Targeting micrornas for metabolic disorders
AU2015327836B2 (en) 2014-10-03 2021-07-01 Cold Spring Harbor Laboratory Targeted augmentation of nuclear gene output
US20170327524A1 (en) 2014-10-10 2017-11-16 Hoffmann-La Roche, Inc. Galnac phosphoramidites, nucleic acid conjugates thereof and their use
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
WO2016081444A1 (en) 2014-11-17 2016-05-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
AU2015364508A1 (en) 2014-12-18 2017-07-06 Alnylam Pharmaceuticals, Inc. ReversirTM compounds
US10793855B2 (en) 2015-01-06 2020-10-06 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
US10731154B2 (en) 2015-02-15 2020-08-04 Arcturus Therapeutics, Inc. 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
EP3277817A4 (en) * 2015-04-03 2018-12-05 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating tmprss6 expression
US10745702B2 (en) 2015-04-08 2020-08-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the LECT2 gene
WO2016167780A1 (en) 2015-04-16 2016-10-20 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
US11249071B2 (en) 2015-04-24 2022-02-15 California Institute Of Technology Reactivation of x chromosome genes
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
CA2990699A1 (en) 2015-06-29 2017-01-05 Ionis Pharmaceuticals, Inc. Modified crispr rna and modified single crispr rna and uses thereof
US10494632B2 (en) 2015-07-10 2019-12-03 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (IGFALS) compositions and methods of use thereof
ES2917181T3 (en) 2015-07-10 2022-07-07 Ionis Pharmaceuticals Inc Diacylglycerol acyltransferase 2 (DGAT2) modulators
WO2017010575A1 (en) * 2015-07-16 2017-01-19 協和発酵キリン株式会社 β2GPI GENE EXPRESSION INHIBITING NUCLEIC ACID COMPLEX
MA43072A (en) 2015-07-22 2018-05-30 Wave Life Sciences Ltd COMPOSITIONS OF OLIGONUCLEOTIDES AND RELATED PROCESSES
JP6940477B2 (en) 2015-08-06 2021-09-29 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Preparation method of GalNAc acid derivative
CN105111118B (en) * 2015-08-14 2017-04-12 天津小新医药科技有限公司 L-menthol P2Y12 receptor antagonists, preparation method thereof and use thereof
CN105111119B (en) * 2015-08-14 2017-04-12 天津小新医药科技有限公司 Halogenobenzene L-menthol P2Y12 receptor antagonists and application thereof
WO2017032726A1 (en) 2015-08-24 2017-03-02 Roche Innovation Center Copenhagen A/S Lna-g process
EP3344769B1 (en) 2015-09-02 2024-04-17 Alnylam Pharmaceuticals, Inc. Programmed cell death 1 ligand 1 (pd-l1) irna compositions and methods of use thereof
EP3353328A4 (en) 2015-09-24 2019-06-12 Ionis Pharmaceuticals, Inc. Modulators of kras expression
MX2018003685A (en) 2015-09-24 2018-08-15 Univ California Synthetic sphingolipid-like molecules, drugs, methods of their synthesis and methods of treatment.
PT3353303T (en) 2015-09-25 2023-10-10 Academisch Ziekenhuis Leiden Compositions and methods for modulating ataxin 3 expression
US20180273948A1 (en) * 2015-09-25 2018-09-27 Tarveda Therapeutics, Inc. RNAi CONJUGATES, PARTICLES AND FORMULATIONS THEREOF
US11116843B2 (en) 2015-09-25 2021-09-14 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
US20210052631A1 (en) * 2015-09-25 2021-02-25 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
EP3355932B1 (en) 2015-10-02 2023-04-12 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugation process
WO2017060731A1 (en) 2015-10-09 2017-04-13 University Of Southampton Modulation of gene expression and screening for deregulated protein expression
WO2017068087A1 (en) 2015-10-22 2017-04-27 Roche Innovation Center Copenhagen A/S Oligonucleotide detection method
US10955407B2 (en) 2015-10-22 2021-03-23 Roche Innovation Center Copenhagen A/S In vitro toxicity screening assay
BR122023026882A2 (en) * 2015-11-06 2024-01-23 Ionis Pharmaceuticals, Inc USE OF AN OLIGOMERIC COMPOUND
WO2017079745A1 (en) 2015-11-06 2017-05-11 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds for use in therapy
BR112018003910B1 (en) * 2015-11-16 2022-03-22 F. Hoffmann-La Roche Ag Galnac phosphoramidite derivative, process for the preparation of a galnac phosphoramidite derivative of formula i and use of the galnac phosphoramidite derivatives of formula i
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
EP3933041B1 (en) 2015-12-14 2024-01-31 Cold Spring Harbor Laboratory Antisense oligomers for treatment of autosomal dominant retardation
CA3006599A1 (en) 2016-01-05 2017-07-13 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
AU2017211461B2 (en) 2016-01-26 2023-01-12 Nissan Chemical Corporation Single-stranded oligonucleotide
WO2017131236A1 (en) 2016-01-29 2017-08-03 協和発酵キリン株式会社 Nucleic acid complex
JP2019503394A (en) * 2016-01-31 2019-02-07 ユニバーシティ・オブ・マサチューセッツUniversity Of Massachusetts Branched oligonucleotide
KR20230048446A (en) 2016-03-07 2023-04-11 애로우헤드 파마슈티컬스 인코포레이티드 Targeting ligands for therapeutic compounds
AU2017229778A1 (en) 2016-03-09 2018-08-16 Ionis Pharmaceuticals, Inc. Methods and compositions for inhibiting PMP22 expression
ES2857702T3 (en) 2016-03-14 2021-09-29 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
AU2017248637A1 (en) 2016-04-13 2018-09-27 Ionis Pharmaceuticals, Inc. Methods for reducing C9ORF72 expression
JP6985288B2 (en) 2016-04-14 2021-12-22 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Trityl-mono-GalNAc compounds and their uses
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
WO2017188898A1 (en) 2016-04-29 2017-11-02 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
WO2017219017A1 (en) 2016-06-17 2017-12-21 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
CN109328236B (en) 2016-06-17 2022-10-25 豪夫迈·罗氏有限公司 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
JP7022687B2 (en) * 2016-06-30 2022-02-18 協和キリン株式会社 Nucleic acid complex
CA3030701A1 (en) 2016-07-11 2018-01-18 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
PL3484524T3 (en) 2016-07-15 2023-03-20 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of smn2
WO2018013999A1 (en) 2016-07-15 2018-01-18 Am Chemicals Llc Non-nucleosidic solid supports and phosphoramidite building blocks for oligonucleotide synthesis
EP3496758A4 (en) 2016-08-12 2020-11-11 University of Massachusetts Conjugated oligonucleotides
KR20190058477A (en) * 2016-08-17 2019-05-29 솔스티스 바이올로직스, 리미티드 Polynucleotide construct
IL300869A (en) * 2016-09-02 2023-04-01 Arrowhead Pharmaceuticals Inc Targeting ligands, compositions comprising same and uses thereof
JOP20190065A1 (en) 2016-09-29 2019-03-28 Ionis Pharmaceuticals Inc Compounds and methods for reducing tau expression
US11400161B2 (en) 2016-10-06 2022-08-02 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
US20180179542A1 (en) * 2016-11-11 2018-06-28 Alios Biopharma, Inc. OLIGONUCLEOTIDE TARGETING STRATEGY FOR 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
WO2018102397A1 (en) 2016-11-29 2018-06-07 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
WO2018110980A1 (en) * 2016-12-13 2018-06-21 주식회사 에이엠사이언스 Pharmaceutical composition for preventing or treating hepatitis b
WO2018112320A1 (en) 2016-12-16 2018-06-21 Alnylam Pharmaceuticals, Inc. 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
EP3568480A1 (en) 2017-01-13 2019-11-20 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating nfkb2 expression
US20200216845A1 (en) 2017-01-13 2020-07-09 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating rela expression
WO2018130585A1 (en) 2017-01-13 2018-07-19 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating relb expression
US20190338286A1 (en) 2017-01-13 2019-11-07 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating rel expression
WO2018130583A1 (en) 2017-01-13 2018-07-19 Roche Innovation Center Copenhagen A/S Antisense oligonucleotides for modulating nfkb1 expression
US11572558B2 (en) 2017-02-06 2023-02-07 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
WO2018191278A2 (en) 2017-04-11 2018-10-18 Arbutus Biopharma Corporation Targeted compositions
BR112019021852A2 (en) 2017-04-18 2020-06-02 Alnylam Pharmaceuticals, Inc. RNAI AGENT AND A VACCINE AGAINST HBV, USE OR METHOD AND KIT FOR TREATMENT
WO2018215049A1 (en) 2017-05-23 2018-11-29 F. Hoffmann-La Roche Ag Process for galnac oligonucleotide conjugates
EP3633036A4 (en) * 2017-05-26 2021-03-17 National Cerebral and Cardiovascular Center Antisense nucleic acid targeting pcsk9
CN111050806A (en) 2017-06-02 2020-04-21 波涛生命科学有限公司 Oligonucleotide compositions and methods of use thereof
EP3599844A4 (en) * 2017-06-07 2021-01-13 University of Massachusetts Anti-adam33 oligonucleotides and related methods
EP3652317A1 (en) 2017-07-13 2020-05-20 Alnylam Pharmaceuticals, Inc. Lactate dehydrogenase a (ldha) irna compositions and methods of use thereof
EP3660153A4 (en) 2017-07-26 2021-04-21 Nissan Chemical Corporation Single-stranded oligonucleotide
WO2019027015A1 (en) * 2017-08-02 2019-02-07 協和発酵キリン株式会社 Nucleic acid complex
WO2019027009A1 (en) * 2017-08-02 2019-02-07 協和発酵キリン株式会社 Nucleic acid complex
WO2019030313A2 (en) 2017-08-11 2019-02-14 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating ube3c expression
CA3071033A1 (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
SI3673080T1 (en) 2017-08-25 2024-03-29 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
SG11202002149XA (en) * 2017-09-14 2020-04-29 Janssen Biopharma Inc Galnac derivatives
CN111417728A (en) 2017-09-29 2020-07-14 因特利亚治疗公司 Compositions and methods for TTR gene editing and treatment of ATTR amyloidosis
JP2021502059A (en) 2017-10-13 2021-01-28 ロシュ イノベーション センター コペンハーゲン エーエス A method for identifying improved three-dimensionally defined phosphorothioate oligonucleotide variants of antisense oligonucleotides by using a sub-library of partially three-dimensionally defined oligonucleotides.
MA52151A (en) 2017-10-16 2020-05-06 Hoffmann La Roche NUCLEIC ACID MOLECULE FOR PAPD5 AND PAPD7 mRNA REDUCTION FOR TREATMENT OF HEPATITIS B INFECTION
CA3078971A1 (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
WO2019099610A1 (en) 2017-11-16 2019-05-23 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
EP3719128A4 (en) 2017-12-01 2021-10-27 Suzhou Ribo Life Science Co., Ltd. Double-stranded oligonucleotide, composition and conjugate comprising double-stranded oligonucleotide, preparation method therefor and use thereof
CN110944675A (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
WO2019105403A1 (en) * 2017-12-01 2019-06-06 苏州瑞博生物技术有限公司 Nucleic acid, composition and conjugate containing the same, preparation method and use thereof
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
US11660347B2 (en) 2017-12-01 2023-05-30 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, composition and conjugate containing same, preparation method, and use thereof
EP3719125A4 (en) * 2017-12-01 2021-09-08 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, composition and conjugate containing same, and preparation method and use
WO2019115416A2 (en) 2017-12-11 2019-06-20 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating fndc3b expression
WO2019115417A2 (en) 2017-12-12 2019-06-20 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating rb1 expression
WO2019118916A1 (en) 2017-12-14 2019-06-20 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
CA3086343A1 (en) 2017-12-18 2019-06-27 Alnylam Pharmaceuticals, Inc. High mobility group box-1 (hmgb1) irna compositions and methods of use thereof
WO2019121838A1 (en) 2017-12-21 2019-06-27 F. Hoffmann-La Roche Ag Companion diagnostic for htra1 rna antagonists
WO2019126641A2 (en) 2017-12-21 2019-06-27 Ionis Pharmaceuticals, Inc. Modulation of frataxin expression
EP3728590A1 (en) 2017-12-22 2020-10-28 Roche Innovation Center Copenhagen A/S Novel thiophosphoramidites
AU2018386524A1 (en) 2017-12-22 2020-07-02 Roche Innovation Center Copenhagen A/S Oligonucleotides comprising a phosphorodithioate internucleoside linkage
BR112020010090A2 (en) 2017-12-22 2020-11-03 Roche Innovation Center Copenhagen A/S gapmer oligonucleotide, pharmaceutically acceptable salt thereof, conjugate, pharmaceutical composition and use thereof
US11485752B2 (en) * 2017-12-26 2022-11-01 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
EP3732185A4 (en) 2017-12-29 2021-11-10 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
EP3737758A1 (en) 2018-01-10 2020-11-18 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating pias4 expression
EP3737759A1 (en) 2018-01-12 2020-11-18 Roche Innovation Center Copenhagen A/S Alpha-synuclein antisense oligonucleotides and uses thereof
EP3737761A1 (en) 2018-01-12 2020-11-18 Bristol-Myers Squibb Company Antisense oligonucleotides targeting alpha-synuclein and uses thereof
SG11202006526YA (en) 2018-01-12 2020-08-28 Bristol Myers Squibb Co Antisense oligonucleotides targeting alpha-synuclein and uses thereof
US20210095275A1 (en) 2018-01-12 2021-04-01 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating gsk3b expression
CN111902537A (en) 2018-01-15 2020-11-06 Ionis制药公司 Modulators of DNM2 expression
US20210095276A1 (en) 2018-01-17 2021-04-01 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating erc1 expression
WO2019141723A1 (en) 2018-01-18 2019-07-25 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
EP3752612A4 (en) 2018-02-12 2021-11-10 Ionis Pharmaceuticals, Inc. Modified compounds and uses thereof
CA3090517A1 (en) 2018-02-14 2019-08-22 Deep Genomics Incorporated Oligonucleotide therapy for wilson disease
KR20200140805A (en) 2018-02-21 2020-12-16 브리스톨-마이어스 스큅 컴퍼니 CAMK2D antisense oligonucleotide and uses thereof
WO2019169243A1 (en) 2018-03-02 2019-09-06 Ionis Pharmaceuticals, Inc. Compounds and methods for the modulation of amyloid-beta precursor protein
TW202000199A (en) 2018-03-02 2020-01-01 美商Ionis製藥公司 Modulators of IRF4 expression
WO2019172286A1 (en) * 2018-03-09 2019-09-12 第一三共株式会社 Therapeutic agent for glycogen storage disease type ia
US11661601B2 (en) 2018-03-22 2023-05-30 Ionis Pharmaceuticals, Inc. Methods for modulating FMR1 expression
AU2019247645A1 (en) 2018-04-05 2020-10-15 Centre Leon Berard Use of FUBP1 inhibitors for treating hepatitis B virus infection
US11365416B2 (en) 2018-04-11 2022-06-21 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
BR112020021253A2 (en) 2018-05-09 2021-02-02 Ionis Pharmaceuticals, Inc. compounds and methods for reducing the expression of atxn3
EP3799604A4 (en) 2018-05-09 2022-09-07 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing fxi expression
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
EP3807411A4 (en) 2018-06-14 2022-08-03 Ionis Pharmaceuticals, Inc. Compounds and methods for increasing stmn2 expression
SG11202011864XA (en) 2018-06-27 2020-12-30 Ionis Pharmaceuticals Inc Compounds and methods for reducing lrrk2 expression
SG11202012759XA (en) 2018-07-03 2021-01-28 Hoffmann La Roche 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
SG11202100077PA (en) 2018-07-25 2021-02-25 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn2 expression
MX2020013270A (en) 2018-07-31 2021-02-18 Roche Innovation Ct Copenhagen As Oligonucleotides comprising a phosphorotrithioate internucleoside linkage.
JP7470097B2 (en) 2018-07-31 2024-04-17 ロシュ イノベーション センター コペンハーゲン エーエス Oligonucleotides containing phosphorotrithioate internucleoside linkages
WO2020033748A1 (en) 2018-08-08 2020-02-13 Arcturus Therapeutics, Inc. Compositions and agents against nonalcoholic steatohepatitis
US11827882B2 (en) 2018-08-10 2023-11-28 University Of Massachusetts Modified oligonucleotides targeting SNPs
AU2019321375A1 (en) 2018-08-13 2021-03-11 Alnylam Pharmaceuticals, Inc. Hepatitis B virus (HBV) dsRNA agent compositions and methods of use thereof
US11918600B2 (en) 2018-08-21 2024-03-05 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition and conjugate containing nucleic acid, and use thereof
WO2020043750A1 (en) 2018-08-28 2020-03-05 Roche Innovation Center Copenhagen A/S Neoantigen 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
JP2022512911A (en) 2018-11-09 2022-02-07 ノバルティス アーゲー Methods of reducing the risk of cardiovascular events using conjugated antisense compounds that target apo (a)
TW202028222A (en) 2018-11-14 2020-08-01 美商Ionis製藥公司 Modulators of foxp3 expression
JOP20210108A1 (en) 2018-11-15 2023-01-30 Ionis Pharmaceuticals Inc Modulators of irf5 expression
TW202039841A (en) 2018-11-21 2020-11-01 美商Ionis製藥公司 Compounds and methods for reducing prion expression
WO2020104649A2 (en) 2018-11-23 2020-05-28 Sanofi Novel rna compositions and methods for inhibiting angptl8
AU2019390097A1 (en) 2018-11-30 2021-07-15 Kyowa Kirin Co., Ltd. Nucleic acid conjugate
JP2022515744A (en) 2018-12-20 2022-02-22 プラクシス プレシジョン メディシンズ, インコーポレイテッド Compositions and Methods for the Treatment of KCNT1-Related Disorders
GB201821269D0 (en) 2018-12-28 2019-02-13 Nippon Shinyaku Co Ltd Myostatin signal inhibitor
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
CN111377985B (en) * 2018-12-29 2023-11-10 苏州瑞博生物技术股份有限公司 Compounds and conjugates, methods of making and uses thereof
CN113330117A (en) * 2019-01-18 2021-08-31 苏州瑞博生物技术股份有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
CA3128093A1 (en) 2019-01-31 2020-08-06 Ionis Pharmaceuticals, Inc. Modulators of yap1 expression
WO2020169695A1 (en) 2019-02-20 2020-08-27 Roche Innovation Center Copenhagen A/S 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
JP2022522430A (en) 2019-02-26 2022-04-19 ロシュ イノベーション センター コペンハーゲン エーエス Oligonucleotide formulation method
TW202045724A (en) 2019-02-27 2020-12-16 美商Ionis製藥公司 Modulators of malat1 expression
EP3931323A4 (en) * 2019-02-28 2023-04-05 Deep Genomics Incorporated Ligand clusters and methods of their use and preparation
CN117431244A (en) 2019-03-29 2024-01-23 Ionis制药公司 Compounds and methods for modulating UBE3A-ATS
US20220177894A1 (en) 2019-04-02 2022-06-09 Proqr Therapeutics Ii B.V. Antisense oligonucleotides for immunotherapy
CN113906139A (en) 2019-04-03 2022-01-07 百时美施贵宝公司 ANGPTL2 antisense oligonucleotides and uses thereof
TW202111119A (en) 2019-05-22 2021-03-16 大陸商蘇州瑞博生物技術股份有限公司 Nucleotide, pharmaceutical composition, conjugate and preparation method and use thereof which can effectively treat and/or prevent abnormal uric acid metabolism or diseases or physiological conditions caused by abnormal uric acid metabolism
CA3139195A1 (en) 2019-05-22 2020-11-26 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition, conjugate, preparation method, and use
JP2022533841A (en) 2019-05-22 2022-07-26 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
WO2020233680A1 (en) 2019-05-22 2020-11-26 苏州瑞博生物技术股份有限公司 Nucleic acid, pharmaceutical composition, conjugate, preparation method, and use
US20220186221A1 (en) 2019-05-24 2022-06-16 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
JP2022534702A (en) 2019-05-24 2022-08-03 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
TW202111123A (en) 2019-05-28 2021-03-16 美商Ionis製藥公司 Compounds and methods for reducing fus expression
KR20220024192A (en) 2019-05-31 2022-03-03 알리고스 테라퓨틱스 인코포레이티드 Modified gapmer oligonucleotides and methods of use
CA3137761A1 (en) 2019-06-04 2020-12-10 Regeneron Pharmaceuticals, Inc. Non-human animals comprising a humanized ttr locus with a beta-slip mutation and methods of use
EP3990465A1 (en) 2019-06-25 2022-05-04 Amgen Inc. Purification methods for carbohydrate-linked oligonucleotides
US11786546B2 (en) 2019-07-26 2023-10-17 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
CN114555621A (en) * 2019-08-15 2022-05-27 Ionis制药公司 Bond-modified oligomeric compounds and uses thereof
US20220290156A1 (en) 2019-08-27 2022-09-15 Sanofi Compositions and methods for inhibiting pcsk9
TW202122093A (en) 2019-08-29 2021-06-16 大陸商蘇州瑞博生物技術股份有限公司 Compound and drug conjugate, and preparation method and use thereof
WO2021041884A1 (en) 2019-08-30 2021-03-04 Alnylam Pharmaceuticals, Inc. Neurofilament light chain (nfl) as a biomarker for transthyretin amyloidosis polyneuropathy
KR20220058578A (en) 2019-09-03 2022-05-09 아크투루스 쎄라퓨틱스, 인크. 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
EP4031140A1 (en) 2019-09-20 2022-07-27 F. Hoffmann-La Roche AG Method of treating hbv infection using a core protein allosteric modulator
CN114728017A (en) 2019-10-14 2022-07-08 阿斯利康(瑞典)有限公司 Modulators of PNPLA3 expression
EP4045652A1 (en) 2019-10-18 2022-08-24 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
KR20220084399A (en) 2019-10-22 2022-06-21 알닐람 파마슈티칼스 인코포레이티드 Complement component C3 iRNA compositions and methods of use thereof
AR120341A1 (en) 2019-11-01 2022-02-09 Alnylam Pharmaceuticals Inc COMPOSITIONS OF RNAi AGENTS AGAINST HUNTINGTINE (HTT) AND THEIR METHODS OF USE
PE20230179A1 (en) 2019-11-13 2023-02-01 Alnylam Pharmaceuticals Inc METHODS AND COMPOSITIONS FOR THE TREATMENT OF A DISORDER ASSOCIATED WITH ANGIOTENSINOGEN (AGT)
EP4061945A1 (en) 2019-11-22 2022-09-28 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
CN112876534B (en) * 2019-11-29 2024-02-09 苏州瑞博生物技术股份有限公司 Liver targeting compounds and conjugates
WO2021119226A1 (en) 2019-12-13 2021-06-17 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
WO2021122910A1 (en) 2019-12-19 2021-06-24 F. Hoffmann-La Roche Ag Use of sbds inhibitors for treating hepatitis b virus infection
EP4077671A1 (en) 2019-12-19 2022-10-26 F. Hoffmann-La Roche AG Use of saraf inhibitors for treating hepatitis b virus infection
WO2021122735A1 (en) 2019-12-19 2021-06-24 F. Hoffmann-La Roche Ag Use of sept9 inhibitors for treating hepatitis b virus infection
EP4077670A1 (en) 2019-12-19 2022-10-26 F. Hoffmann-La Roche AG Use of cops3 inhibitors for treating hepatitis b virus infection
EP4077668A1 (en) 2019-12-19 2022-10-26 F. Hoffmann-La Roche AG Use of scamp3 inhibitors for treating hepatitis b virus infection
CA3163646A1 (en) 2019-12-24 2021-07-01 F. Hoffman-La Roche Ag Pharmaceutical combination of antiviral agents targeting hbv and/or an immune modulator for treatment of hbv
CA3163490A1 (en) 2019-12-24 2021-07-01 F. Hoffman-La Roche Ag Pharmaceutical combination of a therapeutic oligonucleotide targeting hbv and a tlr7 agonist for treatment of hbv
MX2022007911A (en) 2019-12-24 2022-07-21 Hoffmann La Roche Method of treating virus infection using a tlr7 agonist.
JPWO2021153687A1 (en) * 2020-01-30 2021-08-05
WO2021163066A1 (en) 2020-02-10 2021-08-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
AU2021224778A1 (en) 2020-02-18 2022-09-29 Alnylam Pharmaceuticals, Inc. Apolipoprotein C3 (APOC3) iRNA compositions and methods of use thereof
US20220064638A1 (en) 2020-02-28 2022-03-03 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating smn2
WO2021178607A1 (en) 2020-03-05 2021-09-10 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
JP2023516095A (en) 2020-03-06 2023-04-17 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 Ketohexokinase (KHK) iRNA compositions and methods of use thereof
JP2023516748A (en) 2020-03-06 2023-04-20 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 Compositions and methods for inhibiting expression of transthyretin (TTR)
WO2021188611A1 (en) 2020-03-18 2021-09-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
JP2023519215A (en) 2020-03-23 2023-05-10 アムジエン・インコーポレーテツド Monoclonal antibody against chemically modified nucleic acid and its use
WO2021193965A1 (en) * 2020-03-26 2021-09-30 国立研究開発法人国立循環器病研究センター Antisense nucleic acid targeting apoc3
TW202204615A (en) 2020-03-26 2022-02-01 美商阿尼拉製藥公司 Coronavirus irna compositions and methods of use thereof
EP4133078A1 (en) 2020-04-06 2023-02-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing myoc expression
WO2021206922A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
TW202204617A (en) 2020-04-07 2022-02-01 美商艾爾妮蘭製藥公司 Compositions and methods for silencing scn9a expression
EP4133076A1 (en) 2020-04-07 2023-02-15 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
EP4143319A1 (en) 2020-04-27 2023-03-08 Alnylam Pharmaceuticals, Inc. Apolipoprotein e (apoe) irna agent compositions and methods of use thereof
BR112022021136A2 (en) 2020-04-30 2022-11-29 Alnylam Pharmaceuticals Inc COMPLEMENT FACTOR B IRNA COMPOSITIONS (CFB) AND METHODS OF USE THEREOF
CA3181546A1 (en) 2020-05-01 2021-11-04 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating atxn1
JP2023525799A (en) 2020-05-11 2023-06-19 ストーク セラピューティクス,インク. OPA1 antisense oligomers for treatment of conditions and diseases
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)
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)
WO2021231692A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
WO2021231679A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
WO2021231698A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
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)
WO2021231675A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
US11408000B2 (en) 2020-06-03 2022-08-09 Triplet Therapeutics, Inc. Oligonucleotides 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
CA3184289A1 (en) 2020-06-18 2021-12-23 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
EP4179091A1 (en) 2020-07-10 2023-05-17 Institut National De La Sante Et De La Recherche Medicale - Inserm Methods and compositions for treating epilepsy
AU2021326521A1 (en) * 2020-08-13 2023-03-09 Amgen Inc. RNAi constructs and methods for inhibiting MARC1 expression
WO2022038211A2 (en) 2020-08-21 2022-02-24 F. Hoffmann-La Roche Ag Use of a1cf inhibitors for treating hepatitis b virus infection
EP4217489A1 (en) 2020-09-24 2023-08-02 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
JP2023544413A (en) 2020-10-05 2023-10-23 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 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
US20240035029A1 (en) 2020-10-16 2024-02-01 Sanofi 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
AU2021380809A1 (en) 2020-11-13 2023-06-22 Alnylam Pharmaceuticals, Inc. COAGULATION FACTOR V (F5) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP4136092A4 (en) 2020-11-18 2023-10-11 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
WO2022106695A1 (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
EP4274896A1 (en) 2021-01-05 2023-11-15 Alnylam Pharmaceuticals, Inc. Complement component 9 (c9) irna compositions and methods of use thereof
WO2022162161A1 (en) * 2021-01-30 2022-08-04 E-Therapeutics Plc Conjugated oligonucleotide compounds, methods of making and uses thereof
WO2022162153A1 (en) * 2021-01-30 2022-08-04 E-Therapeutics Plc Conjugated oligonucleotide compounds, methods of making and uses thereof
WO2022162157A1 (en) * 2021-01-30 2022-08-04 E-Therapeutics Plc Conjugated oligonucleotide compounds, methods of making and uses thereof
WO2022162154A1 (en) * 2021-01-30 2022-08-04 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
CA3210763A1 (en) 2021-02-12 2022-08-18 Alnylam Pharmaceuticals, Inc. 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
EP4298220A1 (en) 2021-02-25 2024-01-03 Alnylam Pharmaceuticals, Inc. Prion protein (prnp) irna compositions and methods of use thereof
TW202302847A (en) 2021-02-26 2023-01-16 美商艾拉倫製藥股份有限公司 Ketohexokinase (khk) irna compositions and methods of use thereof
KR20230150843A (en) 2021-03-04 2023-10-31 알닐람 파마슈티칼스 인코포레이티드 Angiopoietin-like 3 (ANGPTL3) iRNA compositions and methods of using the same
WO2022189861A1 (en) 2021-03-08 2022-09-15 Tollys Carbohydrate conjugates of tlr3 ligands and uses thereof
WO2022189363A1 (en) 2021-03-08 2022-09-15 Les Laboratoires Servier Antisense oligonucleotides for inhibiting alpha-synuclein expression
EP4305169A1 (en) 2021-03-12 2024-01-17 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
KR20230162024A (en) 2021-03-29 2023-11-28 알닐람 파마슈티칼스 인코포레이티드 Huntingtin (HTT) iRNA preparation composition and method of use thereof
EP4314293A1 (en) 2021-04-01 2024-02-07 Alnylam Pharmaceuticals, Inc. Proline dehydrogenase 2 (prodh2) irna compositions and methods of use thereof
WO2022231999A1 (en) 2021-04-26 2022-11-03 Alnylam Pharmaceuticals, Inc. Transmembrane protease, serine 6 (tmprss6) irna compositions and methods of use thereof
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
AR126000A1 (en) 2021-06-04 2023-08-30 Alnylam Pharmaceuticals Inc ARNI AGENTS OF OPEN READING FRAME 72 OF HUMAN CHROMOSOME 9 (C9ORF72), COMPOSITIONS AND METHODS OF USE THEREOF
EP4351541A2 (en) 2021-06-08 2024-04-17 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating or preventing stargardt's disease and/or retinal binding protein 4 (rbp4)-associated disorders
CA3223192A1 (en) 2021-06-18 2022-12-22 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ifnar1 expression
BR112023026862A2 (en) 2021-06-23 2024-03-05 Beth Israel Deaconess Medical Ct Inc ANTI-FLT1 OLIGONUCLEOTIDE COMPOUNDS OPTIMIZED FOR THE TREATMENT OF PRE-ECLAMPSIA AND OTHER ANGIOGENIC DISORDERS
IL309411A (en) * 2021-06-24 2024-02-01 Lilly Co Eli Novel rna therapeutics and uses thereof
CN117858948A (en) * 2021-06-24 2024-04-09 伊莱利利公司 Novel therapeutic agent delivery moieties and uses thereof
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
CA3225469A1 (en) 2021-06-30 2023-01-05 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
KR20240028502A (en) * 2021-07-02 2024-03-05 투오지에 바이오텍 (상하이) 컴퍼니 리미티드 Nucleic acid ligands and conjugates thereof, preparation methods therefor, and uses thereof
JPWO2023282344A1 (en) 2021-07-08 2023-01-12
WO2023282345A1 (en) 2021-07-08 2023-01-12 日本新薬株式会社 Nephrotoxicity reducing agent
KR20240035503A (en) 2021-07-08 2024-03-15 니뽄 신야쿠 가부시키가이샤 precipitation inhibitor
WO2023003805A1 (en) 2021-07-19 2023-01-26 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
KR20240037293A (en) 2021-07-23 2024-03-21 알닐람 파마슈티칼스 인코포레이티드 Beta-catenin (CTNNB1) iRNA composition and methods of use thereof
WO2023009687A1 (en) 2021-07-29 2023-02-02 Alnylam Pharmaceuticals, Inc. 3-hydroxy-3-methylglutaryl-coa reductase (hmgcr) irna compositions and methods of use thereof
WO2023015223A2 (en) * 2021-08-03 2023-02-09 Verve Therapeutics, Inc. Compositions and methods for targeted rna delivery
AU2022323090A1 (en) 2021-08-03 2024-02-01 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof
WO2023014765A1 (en) 2021-08-04 2023-02-09 Alnylam Pharmaceuticals, Inc. iRNA COMPOSITIONS AND METHODS FOR SILENCING ANGIOTENSINOGEN (AGT)
TW202334413A (en) 2021-08-13 2023-09-01 美商艾拉倫製藥股份有限公司 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)
WO2023041079A1 (en) * 2021-09-18 2023-03-23 上海金中锘美生物医药科技有限公司 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
WO2023045995A1 (en) * 2021-09-23 2023-03-30 Shanghai Argo Biopharmaceutical Co., Ltd. Multivalent Ligand Clusters with Diamine Scaffold for Targeted Delivery of Therapeutic Agents
CA3234835A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
WO2023076450A2 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
TW202333749A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Complement factor b (cfb) irna compositions and methods of use thereof
WO2023083906A2 (en) 2021-11-11 2023-05-19 F. Hoffmann-La Roche Ag Pharmaceutical combinations for treatment of hbv
WO2023102188A1 (en) * 2021-12-03 2023-06-08 Quralis Corporation Gapmer antisense oligonucleotides with modified backbone chemistries
GB202117758D0 (en) 2021-12-09 2022-01-26 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
TW202333751A (en) * 2021-12-16 2023-09-01 大陸商上海拓界生物醫藥科技有限公司 Dsrna, a method for preparing thereof and use thereof
WO2023109940A1 (en) * 2021-12-16 2023-06-22 上海拓界生物医药科技有限公司 Lpa-targeting sirna and conjugate
WO2023111210A1 (en) 2021-12-17 2023-06-22 F. Hoffmann-La Roche Ag Combination of oligonucleotides for modulating rtel1 and fubp1
WO2023138659A1 (en) * 2022-01-20 2023-07-27 上海拓界生物医药科技有限公司 Dsrna, use thereof and preparation method therefor
TW202345865A (en) * 2022-01-24 2023-12-01 大陸商上海舶望製藥有限公司 Composition and method for inhibiting expression of protein LPA(Apo(a))
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
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

Family Cites Families (374)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3687808A (en) 1969-08-14 1972-08-29 Univ Leland Stanford Junior Synthetic polynucleotides
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US5132418A (en) 1980-02-29 1992-07-21 University Patents, Inc. Process for preparing polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 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
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
US5506337A (en) 1985-03-15 1996-04-09 Antivirals Inc. Morpholino-subunit combinatorial library and method
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
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
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.
US5276019A (en) 1987-03-25 1994-01-04 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
GB8712540D0 (en) * 1987-05-28 1987-07-01 Ucb Sa Expression of human proapolipoprotein a-i
AU598946B2 (en) 1987-06-24 1990-07-05 Howard Florey Institute Of Experimental Physiology And Medicine Nucleoside derivatives
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
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
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
EP0406309A4 (en) 1988-03-25 1992-08-19 The University Of Virginia Alumni Patents Foundation Oligonucleotide n-alkylphosphoramidates
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5216141A (en) 1988-06-06 1993-06-01 Benner Steven A Oligonucleotide analogs containing sulfur linkages
US5175273A (en) 1988-07-01 1992-12-29 Genentech, Inc. Nucleic acid intercalating agents
US5194599A (en) 1988-09-23 1993-03-16 Gilead Sciences, Inc. Hydrogen phosphonodithioate compositions
US5256775A (en) 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US5134066A (en) 1989-08-29 1992-07-28 Monsanto Company Improved probes using nucleosides containing 3-dezauracil analogs
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
US5399676A (en) 1989-10-23 1995-03-21 Gilead Sciences Oligonucleotides with inverted polarity
US5721218A (en) 1989-10-23 1998-02-24 Gilead Sciences, Inc. Oligonucleotides with inverted polarity
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
DK0497875T3 (en) 1989-10-24 2000-07-03 Gilead Sciences Inc 2'-modified oligonucleotides
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
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5587470A (en) 1990-01-11 1996-12-24 Isis Pharmaceuticals, Inc. 3-deazapurines
US5457191A (en) 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5859221A (en) 1990-01-11 1999-01-12 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
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
US5623065A (en) 1990-08-13 1997-04-22 Isis Pharmaceuticals, Inc. Gapped 2' modified oligonucleotides
US7101993B1 (en) 1990-01-11 2006-09-05 Isis Pharmaceuticals, Inc. Oligonucleotides containing 2′-O-modified purines
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
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
DE69032425T2 (en) 1990-05-11 1998-11-26 Microprobe Corp Immersion test strips for nucleic acid hybridization assays and methods for covalently immobilizing oligonucleotides
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5618704A (en) 1990-07-27 1997-04-08 Isis Pharmacueticals, Inc. Backbone-modified oligonucleotide analogs and preparation thereof through radical coupling
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
US5608046A (en) 1990-07-27 1997-03-04 Isis Pharmaceuticals, Inc. Conjugated 4'-desmethyl nucleoside analog compounds
US5677437A (en) 1990-07-27 1997-10-14 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5541307A (en) 1990-07-27 1996-07-30 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs and solid phase synthesis thereof
WO1992002258A1 (en) 1990-07-27 1992-02-20 Isis Pharmaceuticals, Inc. Nuclease resistant, pyrimidine modified oligonucleotides that detect and modulate gene expression
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
DK0541722T3 (en) 1990-08-03 1996-04-22 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
CA2092002A1 (en) 1990-09-20 1992-03-21 Mark Matteucci Modified internucleoside linkages
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
US6582908B2 (en) 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
US5948903A (en) 1991-01-11 1999-09-07 Isis Pharmaceuticals, Inc. Synthesis of 3-deazapurines
US5672697A (en) 1991-02-08 1997-09-30 Gilead Sciences, Inc. Nucleoside 5'-methylene phosphonates
US7015315B1 (en) 1991-12-24 2006-03-21 Isis Pharmaceuticals, Inc. Gapped oligonucleotides
US5571799A (en) 1991-08-12 1996-11-05 Basco, Ltd. (2'-5') oligoadenylate analogues useful as inhibitors of host-v5.-graft response
ES2103918T3 (en) 1991-10-17 1997-10-01 Ciba Geigy Ag BICYCLE NUCLEOSIDES, OLIGONUCLEOTIDES, PROCEDURE FOR THEIR OBTAINING AND INTERMEDIATE PRODUCTS.
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
AU3222793A (en) 1991-11-26 1993-06-28 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
TW393513B (en) 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
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
CA2126691C (en) * 1991-12-24 2003-05-06 Phillip Dan Cook Gapped 2' modified oligonucleotides
US5700922A (en) 1991-12-24 1997-12-23 Isis Pharmaceuticals, Inc. PNA-DNA-PNA chimeric macromolecules
FR2687679B1 (en) 1992-02-05 1994-10-28 Centre Nat Rech Scient OLIGOTHIONUCLEOTIDES.
US5633360A (en) 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
US5434257A (en) 1992-06-01 1995-07-18 Gilead Sciences, Inc. Binding compentent oligomers containing unsaturated 3',5' and 2',5' linkages
EP0577558A2 (en) 1992-07-01 1994-01-05 Ciba-Geigy Ag Carbocyclic nucleosides having bicyclic rings, oligonucleotides therefrom, process for their preparation, their use and intermediates
US5652355A (en) 1992-07-23 1997-07-29 Worcester Foundation For Experimental Biology Hybrid oligonucleotide phosphorothioates
RU95104940A (en) 1992-07-27 1997-01-10 Хайбрайдон Method of incorporation of alkylphosphonothioate or arylphosphonothioate internucleotide linkage in oligonucleotide, method of oligonucleotide synthesis, method of gene expression inhibition, treatment method
WO1994014226A1 (en) 1992-12-14 1994-06-23 Honeywell Inc. Motor system with individually controlled redundant windings
KR960701077A (en) 1993-01-25 1996-02-24 다알렌 반스톤 Oligonucleotide Alkylphosphonate and Alkylphosphonothioate (ALIGLPHOSPHONATES AND ALKYLPHOSPHONOTHIOATES)
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
GB9304618D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Chemical compounds
GB9304620D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Compounds
ES2107205T3 (en) 1993-03-30 1997-11-16 Sanofi Sa ANALOGS OF ACICLIC NUCLEOSIDES AND OLIGONUCLEOTIDE SEQUENCES THAT CONTAIN THEM.
CA2159629A1 (en) 1993-03-31 1994-10-13 Sanofi Oligonucleotides with amide linkages replacing phosphodiester linkages
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
US5801154A (en) 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
US5457187A (en) 1993-12-08 1995-10-10 Board Of Regents University Of Nebraska Oligonucleotides containing 5-fluorouracil
ES2149962T3 (en) 1993-12-09 2000-11-16 Univ Jefferson COMPOUNDS AND METHODS FOR MUTATIONS DIRECTED TO THE SITE IN EUCARYTIC 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
EP0862439A4 (en) 1995-11-22 2001-01-10 O Paul O P Ts Ligands to enhance 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
EP0950665A4 (en) 1996-09-26 2004-05-06 Ajinomoto Kk Modified physiologically active proteins and medicinal compositions containing the same
US6770748B2 (en) 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
USRE44779E1 (en) 1997-03-07 2014-02-25 Santaris Pharma A/S Bicyclonucleoside and oligonucleotide analogues
JP3756313B2 (en) 1997-03-07 2006-03-15 武 今西 Novel bicyclonucleosides 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
DE69941574D1 (en) 1998-08-19 2009-12-03 Baxter Healthcare Sa IMMUNOGENOUS BETA-PROPIONAMIDO-BONDED POLYSACCHARID PROTEIN CONJUGATE IS SUITABLE FOR VACCINATION AND MANUFACTURED BY USING N-ACRYLOYLATED POLYSACCHARIDE
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
US6159694A (en) 1999-04-08 2000-12-12 Isis Pharmaceuticals Inc. Antisense modulation of stat3 expression
US7098192B2 (en) * 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
IL145778A0 (en) 1999-04-21 2002-07-25 American Home Prod Methods and compositions for inhibiting the function of polynucleotide sequences
EP1178999B1 (en) 1999-05-04 2007-03-14 Santaris Pharma A/S L-ribo-lna analogues
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
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
AU6605900A (en) 1999-07-16 2001-02-05 Hyseq, Inc. Novel angiopoietin materials and methods
JP4151751B2 (en) 1999-07-22 2008-09-17 第一三共株式会社 New bicyclonucleoside analogues
DE19935303A1 (en) * 1999-07-28 2001-02-08 Aventis Pharma Gmbh Oligonucleotides to inhibit the expression of human eg5
US20020082227A1 (en) 1999-09-30 2002-06-27 Scott Henry Use of oligonucleotides for inhibition of complement activation
US20050112118A1 (en) 1999-12-02 2005-05-26 Myriad Genetics, Incorporated Compositions and methods for treating inflammatory disorders
IL149849A0 (en) 1999-12-09 2002-11-10 Sankyo Co A method and kit for testing an agent for treatment of hyperlipemia
DE60027203T2 (en) 1999-12-30 2007-01-04 K.U. Leuven Research & Development CYCLOHEXENE NUCLEIC
US6602857B1 (en) 2000-01-18 2003-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US7179796B2 (en) 2000-01-18 2007-02-20 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US6261840B1 (en) 2000-01-18 2001-07-17 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
US20020055479A1 (en) 2000-01-18 2002-05-09 Cowsert Lex M. Antisense modulation of PTP1B expression
US7491805B2 (en) 2001-05-18 2009-02-17 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US7833992B2 (en) 2001-05-18 2010-11-16 Merck Sharpe & Dohme 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
AU2001241541A1 (en) 2000-02-17 2001-08-27 Millennium Predictive Medicine, Inc. Novel genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of human prostate cancer
EP1314734A1 (en) 2000-08-29 2003-05-28 Takeshi Imanishi Novel nucleoside analogs and oligonucleotide derivatives containing these analogs
US6426220B1 (en) 2000-10-30 2002-07-30 Isis Pharmaceuticals, Inc. Antisense modulation of calreticulin expression
AU2002217980A1 (en) 2000-12-01 2002-06-11 Cell Works Inc. Conjugates of glycosylated/galactosylated peptide
WO2002087541A1 (en) 2001-04-30 2002-11-07 Protiva Biotherapeutics Inc. Lipid-based formulations for gene transfer
IL159153A0 (en) 2001-06-08 2004-06-01 Sankyo Co Methods for testing drugs for treating or preventing diseases such as hyperlipidemia
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
CA2452458A1 (en) 2001-07-03 2003-01-16 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US7259150B2 (en) 2001-08-07 2007-08-21 Isis Pharmaceuticals, Inc. Modulation of apolipoprotein (a) expression
US7227014B2 (en) * 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense 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
CA2459347C (en) 2001-09-04 2012-10-09 Exiqon A/S Locked nucleic acid (lna) compositions and uses thereof
US7439043B2 (en) 2001-10-10 2008-10-21 Neose Technologies, Inc. Galactosyl nucleotide sugars
JP5105696B2 (en) 2001-11-16 2012-12-26 ジェネンテック, インコーポレイテッド Angiopoietin-like protein 3 Angptl3-containing composition and method of use thereof
US20100240730A1 (en) 2002-02-20 2010-09-23 Merck Sharp And Dohme Corp. RNA Interference Mediated Inhibition of Gene Expression Using Chemically Modified Short Interfering Nucleic Acid (siNA)
AU2003274906A1 (en) 2002-07-31 2004-02-16 Nucleonics, Inc. Double stranded rna structures and constructs, and methods for generating and using the same
WO2004014933A1 (en) 2002-08-07 2004-02-19 University Of Massachusetts Compositions for rna interference and methods of use thereof
WO2004024757A2 (en) 2002-09-11 2004-03-25 Santaris Pharma A/S Modified pna molecules
AU2003284323A1 (en) 2002-10-18 2004-05-04 Alnylam Pharmaceuticals Inc Double-stranded rna structures and constructs, and methods for generating and using the same
WO2004041889A2 (en) 2002-11-05 2004-05-21 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
WO2004044136A2 (en) 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2’-modified nucleosides for use in gene modulation
US7511131B2 (en) 2002-11-13 2009-03-31 Genzyme Corporation Antisense modulation of apolipoprotein B expression
EP2336318B1 (en) 2002-11-13 2013-04-24 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
EP2305812A3 (en) 2002-11-14 2012-06-06 Dharmacon, Inc. Fuctional and hyperfunctional sirna
EP2474630B1 (en) 2002-12-20 2016-04-27 Celera Corporation Genetic polymorphisms associated with myocardial infarction, methods of detection and uses thereof
US6673661B1 (en) 2002-12-20 2004-01-06 Taiwan Semiconductor Manufacturing Co., Ltd. Self-aligned method for forming dual gate thin film transistor (TFT) device
WO2004063208A1 (en) 2003-01-09 2004-07-29 Postech Foundation New phosphoramidite compounds
WO2004072046A2 (en) 2003-02-12 2004-08-26 Carex S.A. Quinoline derivatives and their use for modulation of lxr activity
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
EP1605978B1 (en) * 2003-03-07 2010-09-01 Alnylam Pharmaceuticals Inc. Therapeutic compositions
EP1608735A4 (en) * 2003-04-03 2008-11-05 Alnylam Pharmaceuticals Irna conjugates
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
EP1620544B1 (en) 2003-04-17 2018-09-19 Alnylam Pharmaceuticals Inc. MODIFIED iRNA AGENTS
US7851615B2 (en) 2003-04-17 2010-12-14 Alnylam Pharmaceuticals, Inc. Lipophilic conjugated iRNA agents
US7723509B2 (en) 2003-04-17 2010-05-25 Alnylam Pharmaceuticals IRNA agents with biocleavable tethers
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
US7750142B2 (en) 2003-04-28 2010-07-06 Isis Pharmaceuticals, Inc. Modulation of glucagon receptor expression
JPWO2004101619A1 (en) 2003-05-15 2006-10-26 塩野義製薬株式会社 Rational design and synthesis of functional glycopeptides
WO2004106356A1 (en) 2003-05-27 2004-12-09 Syddansk Universitet Functionalized nucleotide derivatives
ES2382807T3 (en) 2003-08-28 2012-06-13 Takeshi Imanishi New artificial nucleic acids of the N-O link type with cross-linking
JP5379347B2 (en) 2003-09-18 2013-12-25 アイシス ファーマシューティカルズ, インコーポレーテッド 4'-thionucleosides and oligomeric compounds
TW200519202A (en) 2003-09-18 2005-06-16 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
WO2005071080A2 (en) 2004-01-20 2005-08-04 Isis Pharmaceuticals, Inc. Modulation of glucocorticoid receptor expression
US20050244869A1 (en) 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
WO2005097155A1 (en) 2004-04-08 2005-10-20 Takara Bio Inc. Neurite elongation inducing agent
AU2005248147A1 (en) 2004-05-11 2005-12-08 Alphagen Co., Ltd. Polynucleotides for causing RNA interference and method for inhibiting gene expression using the same
JP2008501693A (en) 2004-06-03 2008-01-24 アイシス ファーマシューティカルズ、インク. Double-stranded composition with individually regulated strands for use in gene regulation
BRPI0513477A (en) 2004-07-20 2008-05-06 Genentech Inc Angiotensin-like protein 4 compositions and processes
NZ552956A (en) 2004-07-20 2010-03-26 Genentech Inc Inhibitors of angiopoietin-like 4 protein (ANGPTL4), combinations, and their use for treating cancer
HUE026284T2 (en) * 2004-08-10 2016-06-28 Genzyme Corp Oligonucleotides for use in modulating lipoprotein and cholesterol levels in humans
US7582744B2 (en) 2004-08-10 2009-09-01 Alnylam Pharmaceuticals, Inc. Chemically modified oligonucleotides
WO2006031461A2 (en) 2004-09-09 2006-03-23 Isis Pharmaceuticals, Inc. Pyrrolidinyl groups for attaching conjugates to oligomeric compounds
US20060089325A1 (en) 2004-10-13 2006-04-27 Sanjay Bhanot Antisense modulation of PTP1B expression
US20080261905A1 (en) 2004-11-08 2008-10-23 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
US20080206869A1 (en) 2005-01-24 2008-08-28 Avaris Ab Nucleic Acid Complex
WO2007031081A2 (en) * 2005-09-15 2007-03-22 Santaris Pharma A/S RNA ANTAGONIST COMPOUNDS FOR THE INHIBITION OF APO-Bl00 EXPRESSION
ES2369328T3 (en) 2005-09-19 2011-11-29 Isis Pharmaceuticals, Inc. MODULATION OF GLUCAGON RECEIVER EXPRESSION.
KR20080065617A (en) 2005-09-19 2008-07-14 존슨 앤드 존슨 파머슈티컬 리서치 앤드 디벨로프먼트 엘엘씨 Modulation of glucocorticoid receptor expression
JP5425474B2 (en) 2006-01-26 2014-02-26 アイシス ファーマシューティカルズ, インコーポレーテッド Composition and use thereof for huntingtin
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
CA2640171C (en) 2006-01-27 2014-10-28 Isis Pharmaceuticals, Inc. 6-modified bicyclic nucleic acid analogs
AU2007258117B2 (en) 2006-05-05 2013-05-30 Isis Pharmaceuticals, Inc. Compounds and methods for modulating gene expression
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
US8101585B2 (en) 2006-08-04 2012-01-24 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
CA2660842C (en) 2006-08-18 2012-03-13 F. Hoffmann-La Roche Ag 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
US9550988B2 (en) 2006-10-18 2017-01-24 Ionis Pharmaceuticals, Inc. Antisense compounds
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
EP2455471A3 (en) 2006-11-27 2012-09-12 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
JP5484063B2 (en) 2006-12-08 2014-05-07 レキシコン ファーマシューティカルズ インコーポレーテッド Monoclonal antibody against ANGPTL3
CN101616690B (en) * 2006-12-20 2015-11-25 爱克索马美国有限责任公司 Be used for the treatment of the method for IL-1 ss related diseases
EP2125852B1 (en) 2007-02-15 2016-04-06 Ionis Pharmaceuticals, Inc. 5'-substituted-2'-f modified nucleosides and oligomeric compounds prepared therefrom
US9216228B2 (en) 2007-02-16 2015-12-22 KTB Tumorforschungsgesellschaft MBM Receptor and antigen targeted prodrug
AU2008221287A1 (en) 2007-03-01 2008-09-04 Wellstat Immunotherapeutics, Llc Treatment of diseases characterized by inflammation
US20100055782A1 (en) 2007-03-02 2010-03-04 Mdrna, Inc. Nucleic acid compounds for inhibiting myc gene expression and uses thereof
AU2008242583B2 (en) 2007-04-23 2013-10-10 Alnylam Pharmaceuticals, Inc. Glycoconjugates of RNA interference agents
WO2008150729A2 (en) 2007-05-30 2008-12-11 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
EP2173760B2 (en) 2007-06-08 2015-11-04 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
WO2009003009A1 (en) 2007-06-26 2008-12-31 Enanta Pharmaceuticals, Inc. Substituted pyrrolidine as anti-infectives
US8278283B2 (en) 2007-07-05 2012-10-02 Isis Pharmaceuticals, Inc. 6-disubstituted or unsaturated bicyclic nucleic acid analogs
KR101654007B1 (en) 2007-08-15 2016-09-05 아이오니스 파마수티컬즈, 인코포레이티드 Tetrahydropyran nucleic acid analogs
BRPI0817527A2 (en) * 2007-10-01 2017-05-02 Isis Pharmaceuticals Inc antisense modulation of human fibroblast growth factor receptor expression 4
US9029337B2 (en) 2007-11-09 2015-05-12 Isis Pharmaceuticals, Inc. Modulation of factor 7 expression
WO2009067647A1 (en) 2007-11-21 2009-05-28 Isis Pharmaceuticals, Inc. Carbocyclic alpha-l-bicyclic nucleic acid analogs
AU2008333811B2 (en) 2007-12-04 2014-05-01 Alnylam Pharmaceuticals, Inc. Carbohydrate conjugates as delivery agents for oligonucleotides
EP3673910B1 (en) 2007-12-27 2023-11-22 Stelic Institute & Co., Inc. Sugar chain-related gene and use thereof
AU2009241591A1 (en) 2008-01-31 2009-11-05 Alnylam Pharmaceuticals, Inc. Optimized methods for delivery of DSRNA targeting the PCSK9 gene
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
JP5788312B2 (en) 2008-04-11 2015-09-30 アルニラム ファーマスーティカルズ インコーポレイテッドAlnylam Pharmaceuticals, Inc. Site-specific delivery of nucleic acids by combining targeting ligands with endosomal degradable components
WO2009143369A2 (en) 2008-05-22 2009-11-26 Isis Pharmaceuticals, Inc. Method of preparing nucleosides and analogs thereof without using chromatography
WO2009148605A2 (en) 2008-06-04 2009-12-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
EP2733222A1 (en) 2008-07-09 2014-05-21 Celera Corporation Genetic polymorphisms associated with cardiovascular diseases, methods of detection and uses thereof
WO2010017509A1 (en) 2008-08-07 2010-02-11 Isis Pharmaceuticals, Inc. Modulation of transthyretin expression for the treatment of cns related disorders
WO2010039548A2 (en) 2008-09-23 2010-04-08 Alnylam Pharmaceuticals, Inc. Chemical modifications of monomers and oligonucleotides with cycloaddition
EP2356129B1 (en) 2008-09-24 2013-04-03 Isis Pharmaceuticals, Inc. Substituted alpha-l-bicyclic nucleosides
US8604192B2 (en) 2008-09-24 2013-12-10 Isis Pharmaceuticals, Inc. Cyclohexenyl nucleic acids analogs
KR101979134B1 (en) 2008-10-15 2019-05-15 아이오니스 파마수티컬즈, 인코포레이티드 Modulation of factor 11 expression
SI2937418T1 (en) * 2008-10-20 2018-02-28 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
US8987435B2 (en) 2008-10-24 2015-03-24 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
US8883752B2 (en) 2008-10-24 2014-11-11 Isis Pharmaceuticals, Inc. 5′ and 2′ BIS-substituted nucleosides and oligomeric compounds prepared therefrom
KR20220150995A (en) 2008-11-10 2022-11-11 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
EP3243504A1 (en) 2009-01-29 2017-11-15 Arbutus Biopharma Corporation Improved lipid formulation
WO2010101951A1 (en) 2009-03-02 2010-09-10 Alnylam Pharmaceuticals, Inc. Nucleic acid chemical modifications
FR2943060B1 (en) 2009-03-13 2013-01-04 Commissariat Energie Atomique METAL ION CHELATING AGENTS, PROCESSES FOR THEIR PREPARATION AND THEIR APPLICATIONS
AU2010236286B2 (en) 2009-04-15 2013-06-06 Isis Pharmaceuticals, Inc. Modulation of inflammatory responses by Factor XI
CA3042927C (en) 2009-05-05 2022-05-17 Arbutus Biopharma Corporation Lipid compositions for the delivery of therapeutic agents
US9200276B2 (en) 2009-06-01 2015-12-01 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent RNA interference, compositions and methods of use thereof
KR102205886B1 (en) 2009-06-10 2021-01-21 알닐람 파마슈티칼스 인코포레이티드 Improved lipid formulation
KR20120050429A (en) 2009-06-15 2012-05-18 알닐람 파마슈티칼스 인코포레이티드 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.
US9512164B2 (en) 2009-07-07 2016-12-06 Alnylam Pharmaceuticals, Inc. Oligonucleotide end caps
WO2011005860A2 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. 5' phosphate mimics
WO2011008995A1 (en) 2009-07-16 2011-01-20 Isis Pharmaceuticals, Inc. Modulation of factor 7 expression
EP2462153B1 (en) 2009-08-06 2015-07-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
WO2011038356A2 (en) 2009-09-25 2011-03-31 Johns Hopkins University Novel liver-targeting agents and their synthesis
BR112012008865A2 (en) 2009-10-16 2019-09-24 Glaxo Group Ltd antisense hbv inhibitors.
TWI391144B (en) 2009-10-26 2013-04-01 Iner Aec Executive Yuan A quantification method for remaining liver function with a novel liver receptor imaging agent
TWI388338B (en) 2009-10-26 2013-03-11 Iner Aec Executive Yuan Method of radiolabelling multivalent glycoside for using as hepatic receptor imaging agent
US9101643B2 (en) * 2009-11-03 2015-08-11 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of transthyretin (TTR)
WO2011072290A2 (en) 2009-12-11 2011-06-16 The Regents Of The University Of Michigan Targeted dendrimer-drug conjugates
WO2011085271A2 (en) 2010-01-08 2011-07-14 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)
AP3284A (en) 2010-02-24 2015-05-31 Arrowhead Res Corp Compositions for targeted delivery of SIRNA
US9193752B2 (en) 2010-03-17 2015-11-24 Isis Pharmaceuticals, Inc. 5′-substituted bicyclic nucleosides and oligomeric compounds prepared therefrom
CA2794307A1 (en) 2010-03-26 2011-09-29 Mersana Therapeutics, Inc. Modified polymers for delivery of polynucleotides, method of manufacture, and methods of use thereof
US20130109817A1 (en) 2010-03-26 2013-05-02 Mersana Therapeutics, Inc. Modified Polymers for Delivery of Polynucleotides, Method of Manufacture, and Methods of Use Thereof
WO2011123621A2 (en) 2010-04-01 2011-10-06 Alnylam Pharmaceuticals Inc. 2' and 5' modified monomers and oligonucleotides
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
WO2011139702A2 (en) 2010-04-28 2011-11-10 Isis Pharmaceuticals, Inc. Modified nucleosides and oligomeric compounds prepared therefrom
BR112012027547B1 (en) 2010-04-29 2022-06-14 Ionis Pharmaceuticals, Inc SINGLE STRIP MODIFIED OLIGONUCLEOTIDE, COMPOSITION, AND ITS USES TO TREAT TRANSTHIRRETIN AYLOIDOSIS, REDUCE ITS SYMPTOMS, AND TO REDUCE TRANSTHIRRETIN MRNA OR PROTEIN EXPRESSION
WO2011163121A1 (en) 2010-06-21 2011-12-29 Alnylam Pharmaceuticals, Inc. Multifunctional copolymers for nucleic acid delivery
CA2812046A1 (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
RU2013117288A (en) 2010-12-17 2015-01-27 Эрроухэд Рисерч Корпорейшн CONTAINING GALACTOSY CLUSTER A TARGETING GROUP FOR miRNA MODULATING FORMKOKINETIC PROPERTIES
US8501930B2 (en) 2010-12-17 2013-08-06 Arrowhead Madison Inc. Peptide-based in vivo siRNA delivery system
RU2582235C2 (en) 2010-12-29 2016-04-20 Ф.Хоффманн-Ля Рош Аг Low-molecular 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
CA2831915C (en) 2011-04-01 2021-01-12 Isis Pharmaceuticals, Inc. Modulation of signal transducer and activator of transcription 3 (stat3) expression
JP5951752B2 (en) * 2011-04-13 2016-07-13 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. Antisense regulation of PTP1B expression
WO2012145674A1 (en) 2011-04-21 2012-10-26 Isis Pharmaceuticals, Inc. Modulation of hepatitis b virus (hbv) expression
UA118951C2 (en) * 2011-04-21 2019-04-10 Айоніс Фармасьютікалз, Інк. Modulation of hepatitis b virus (hbv) expression
WO2012149495A1 (en) 2011-04-27 2012-11-01 Isis Pharmaceuticals, Inc. Modulation of apolipoprotein ciii (apociii) expression
WO2012174154A1 (en) 2011-06-13 2012-12-20 Isis Pharmaceuticals, Inc. Modulation of inflammatory responses by factor vii
EP2721156B1 (en) 2011-06-16 2016-12-21 Ionis Pharmaceuticals, Inc. Antisense modulation of fibroblast growth factor receptor 4 expression
AU2012272970A1 (en) 2011-06-21 2014-02-06 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (ANGPTL3) iRNA compositions and methods of use thereof
RU2631805C2 (en) 2011-06-21 2017-09-26 Элнилэм Фармасьютикалз, Инк. Compositions and methods for apolipoprotein c-iii (apoc3) gene expression inhibition
WO2012177639A2 (en) 2011-06-22 2012-12-27 Alnylam Pharmaceuticals, Inc. Bioprocessing and bioproduction using avian cell lines
US20130017250A1 (en) * 2011-07-15 2013-01-17 The Trustees Of Columbia University In The City Of New York Methods For High Density Lipoprotein Cholesterol Regulation
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
BR112014006587B1 (en) * 2011-09-20 2021-07-27 Ionis Pharmaceuticals, Inc COMPOUND COMPRISING OLIGONUCLEOTIDE THAT MODULATES GCGR EXPRESSION, COMPOSITION, AS WELL AS ITS USE IN THE TREATMENT OR PREVENTION OF DIABETES
US8901098B2 (en) 2011-10-25 2014-12-02 Isis Pharmaceuticals, Inc. Antisense modulation of GCCR expression
EP3301177B1 (en) 2011-11-18 2020-03-25 Alnylam Pharmaceuticals, Inc. Rnai agents, compositions and methods of use thereof for treating transthyretin (ttr) associated diseases
AU2013216852A1 (en) 2012-02-08 2014-08-21 Isis Pharmaceuticals, Inc. Methods and compositions for modulating Factor VII expression
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
EP3336189A1 (en) 2012-04-20 2018-06-20 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleotides and uses thereof
WO2013159109A1 (en) 2012-04-20 2013-10-24 Isis Pharmaceuticals, Inc. Modulation of hepatitis b virus (hbv) expression
EP3919620A1 (en) 2012-05-02 2021-12-08 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) compositions
AR090906A1 (en) 2012-05-02 2014-12-17 Merck Sharp & Dohme CONJUGATES CONTAINING TETRAGALNAC AND PROCEDURES FOR THE ADMINISTRATION OF OLIGONUCLEOTIDES
US20160002624A1 (en) 2012-05-17 2016-01-07 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions
US9574193B2 (en) 2012-05-17 2017-02-21 Ionis Pharmaceuticals, Inc. Methods and compositions for modulating apolipoprotein (a) expression
NZ631071A (en) 2012-05-24 2017-02-24 Ionis Pharmaceuticals Inc Methods and compositions for modulating apolipoprotein(a) expression
US20150322428A1 (en) 2012-06-18 2015-11-12 Isis Pharmaceuticals, Inc. Compounds and methods for improved cellular uptake of antisense compounds
WO2014025805A1 (en) 2012-08-06 2014-02-13 Alnylam Pharmaceuticals, Inc. Carbohydrate conjugated rna agents and process for their preparation
US9695418B2 (en) 2012-10-11 2017-07-04 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleosides and uses thereof
EP2920304B1 (en) 2012-11-15 2019-03-06 Roche Innovation Center Copenhagen A/S Oligonucleotide conjugates
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
NZ631512A (en) * 2013-05-01 2016-10-28 Ionis Pharmaceuticals Inc Compositions and methods for modulating apolipoprotein (a) expression
KR20200085922A (en) 2013-05-16 2020-07-15 다이니뽄 스미토모 세이야쿠 가부시키가이샤 Transplantation adjuvant in cell therapy using neural progenitor cells
US20160122761A1 (en) 2013-06-21 2016-05-05 Isis Pharmaceuticals, Inc. Compositions and methods for modulation of target nucleic acids
MY183049A (en) 2013-06-27 2021-02-09 Roche Innovation Ct Copenhagen As Antisense oligomers and conjugates targeting pcsk9
CN105452461B (en) 2013-07-02 2021-04-13 Ionis制药公司 Modulators of growth hormone receptors
WO2015006740A2 (en) 2013-07-11 2015-01-15 Alnylam Pharmaceuticals, Inc. Oligonucleotide-ligand conjugates and process for their preparation
DK3043827T3 (en) 2013-09-13 2019-08-26 Ionis Pharmaceuticals Inc MODULATORS OF COMPLEMENT FACTOR B
US9943604B2 (en) 2013-09-20 2018-04-17 Ionis Pharmaceuticals, Inc. Targeted therapeutic nucleosides and their use
US20160289677A1 (en) 2013-11-14 2016-10-06 Roche Innovation Center Copenhagen A/S APOB Antisense Conjugate Compounds
WO2015085158A1 (en) 2013-12-06 2015-06-11 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of transthyretin (ttr) by double-stranded rna
EP3608406B1 (en) 2014-05-01 2023-02-15 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating complement factor b expression
PT3137605T (en) 2014-05-01 2020-12-18 Ionis Pharmaceuticals Inc Compositions and methods for modulating angiopoietin-like 3 expression
KR20200102553A (en) 2014-05-01 2020-08-31 아이오니스 파마수티컬즈, 인코포레이티드 Compositions and methods for modulating growth hormone receptor expression
EP3137115B1 (en) 2014-05-01 2020-10-14 Ionis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
RU2703411C2 (en) 2014-05-01 2019-10-16 Ионис Фармасьютикалз, Инк. Compositions and methods for modulating pkk expression
WO2015179693A1 (en) 2014-05-22 2015-11-26 Isis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
WO2015188194A1 (en) 2014-06-06 2015-12-10 Isis Pharmaceuticals, Inc. Compositions and methods for enhanced intestinal absorption of conjugated oligomeric compounds
BR122023026882A2 (en) * 2015-11-06 2024-01-23 Ionis Pharmaceuticals, Inc USE OF AN OLIGOMERIC COMPOUND
WO2017079745A1 (en) * 2015-11-06 2017-05-11 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds for use in therapy

Also Published As

Publication number Publication date
IL284593B (en) 2022-10-01
IL264241A (en) 2019-02-28
DK2991656T3 (en) 2020-03-23
US9181549B2 (en) 2015-11-10
KR102315836B1 (en) 2021-10-22
AU2014259750A1 (en) 2015-10-22
LT2992009T (en) 2020-11-10
HK1221404A1 (en) 2017-06-02
AU2019203674B2 (en) 2021-03-25
EP2992097A2 (en) 2016-03-09
EP3633039A1 (en) 2020-04-08
SI2992009T1 (en) 2020-10-30
CN112921036A (en) 2021-06-08
IL261901A (en) 2018-10-31
CN105377887B (en) 2020-11-03
US9932580B2 (en) 2018-04-03
US9145558B2 (en) 2015-09-29
CN114058617A (en) 2022-02-18
AU2014259757A1 (en) 2015-10-22
CR20150612A (en) 2016-03-03
WO2014179629A8 (en) 2016-06-02
US20180273953A1 (en) 2018-09-27
KR102212275B1 (en) 2021-02-05
US20190367914A1 (en) 2019-12-05
RU2015151204A (en) 2017-06-02
EP3524680A1 (en) 2019-08-14
SG10201801813YA (en) 2018-04-27
AU2017200365B2 (en) 2018-11-08
NZ725538A (en) 2021-02-26
ES2730015T3 (en) 2019-11-07
JP2018183184A (en) 2018-11-22
EP3524680B1 (en) 2020-11-11
US20180002693A1 (en) 2018-01-04
MX2015015264A (en) 2016-08-12
MX2021008901A (en) 2021-08-19
JP6866459B2 (en) 2021-04-28
US20160017323A1 (en) 2016-01-21
CN105392488B (en) 2021-04-30
KR20230113835A (en) 2023-08-01
SG11201508800WA (en) 2015-11-27
HK1221403A1 (en) 2017-06-02
BR112015027369A8 (en) 2018-01-02
RU2019110030A (en) 2019-05-06
AU2020233603A1 (en) 2020-10-01
RU2015151202A (en) 2017-06-06
AU2017200950B2 (en) 2019-01-17
EP3690049A1 (en) 2020-08-05
US20190055554A1 (en) 2019-02-21
MX2015015263A (en) 2016-12-16
PT2992098T (en) 2019-07-05
AU2021204244A1 (en) 2021-07-22
RU2697152C2 (en) 2019-08-12
EP2991656A4 (en) 2017-02-22
MX2020002184A (en) 2020-07-14
RU2670614C9 (en) 2018-11-23
US10683499B2 (en) 2020-06-16
IL264580A (en) 2019-02-28
BR112015027377A2 (en) 2017-08-29
RU2018136140A (en) 2018-12-17
JP2016522817A (en) 2016-08-04
JP2016526018A (en) 2016-09-01
IL284000A (en) 2021-07-29
ZA201507218B (en) 2023-09-27
HUE050394T2 (en) 2020-11-30
HRP20201378T1 (en) 2020-11-27
KR20210014758A (en) 2021-02-09
KR102558571B1 (en) 2023-07-21
RU2015151202A3 (en) 2018-03-27
JP7177127B2 (en) 2022-11-22
JP2016523515A (en) 2016-08-12
KR20160003723A (en) 2016-01-11
MX2019010443A (en) 2019-10-17
JP2018027091A (en) 2018-02-22
CY1123369T1 (en) 2021-12-31
MX2015015234A (en) 2016-10-03
US20210087566A1 (en) 2021-03-25
ES2819213T3 (en) 2021-04-15
US9181550B2 (en) 2015-11-10
JP2021074021A (en) 2021-05-20
AU2014259759B2 (en) 2020-06-18
AU2014259757B2 (en) 2017-03-02
US20150126718A1 (en) 2015-05-07
CA2921514A1 (en) 2014-11-06
NZ740338A (en) 2022-04-29
JP2020039354A (en) 2020-03-19
US20180044676A1 (en) 2018-02-15
UA121017C2 (en) 2020-03-25
PH12019501191A1 (en) 2021-03-01
JP7429103B2 (en) 2024-02-07
AU2017200950A1 (en) 2017-03-02
AU2018267625A1 (en) 2018-12-13
MX2015015220A (en) 2016-01-12
MY198359A (en) 2023-08-28
IL242124B (en) 2019-02-28
ES2885174T3 (en) 2021-12-13
CN108064162A (en) 2018-05-22
IL284593B2 (en) 2023-02-01
ES2778442T3 (en) 2020-08-10
WO2014179626A3 (en) 2015-02-26
KR20200090966A (en) 2020-07-29
JP2023012548A (en) 2023-01-25
CN105378082B (en) 2020-06-09
DOP2021000095A (en) 2021-09-15
RU2015151200A (en) 2019-01-14
JP2023113843A (en) 2023-08-16
DOP2015000268A (en) 2015-11-30
KR102138781B1 (en) 2020-07-28
US20220275365A9 (en) 2022-09-01
EA201592093A1 (en) 2016-06-30
US9127276B2 (en) 2015-09-08
PH12015502493A1 (en) 2016-02-22
DK2992009T3 (en) 2020-09-14
KR20180051678A (en) 2018-05-16
ME03390B (en) 2020-01-20
US20210024923A1 (en) 2021-01-28
PE20161430A1 (en) 2017-01-06
IL263843A (en) 2019-01-31
RU2015151203A (en) 2017-06-02
KR20160002977A (en) 2016-01-08
RU2019124314A (en) 2019-08-21
AU2014259755B2 (en) 2018-08-30
KR20220108195A (en) 2022-08-02
JP2020039355A (en) 2020-03-19
EP2992097A4 (en) 2017-01-04
IL274064B (en) 2021-06-30
EA036584B1 (en) 2020-11-26
RU2015151199A3 (en) 2018-03-27
HK1221486A1 (en) 2017-06-02
US20160090595A1 (en) 2016-03-31
CN105378085B (en) 2019-02-15
MX2020004209A (en) 2020-08-13
EP2991661B1 (en) 2019-03-13
RU2686080C2 (en) 2019-04-24
BR112015027377B1 (en) 2023-01-10
EA031393B1 (en) 2018-12-28
IL274064A (en) 2020-06-30
IL284593A (en) 2021-08-31
AU2020217347A1 (en) 2020-08-27
WO2014179620A1 (en) 2014-11-06
RU2670614C2 (en) 2018-10-24
KR20190084138A (en) 2019-07-15
AU2019202598A1 (en) 2019-05-02
JP6995478B2 (en) 2022-01-14
AU2019200820B2 (en) 2020-04-30
PT3524680T (en) 2021-01-04
JP2022017514A (en) 2022-01-25
IL273205A (en) 2020-04-30
KR20160002976A (en) 2016-01-08
WO2014179625A1 (en) 2014-11-06
EP2992009A4 (en) 2016-12-28
CL2016002262A1 (en) 2017-06-09
AU2020207820A1 (en) 2020-08-06
JP2020074787A (en) 2020-05-21
US20150126720A1 (en) 2015-05-07
CA2921518A1 (en) 2014-11-06
RU2018136140A3 (en) 2022-04-27
JP6216444B2 (en) 2017-10-18
BR112015027319A8 (en) 2018-01-02
AU2017200365A1 (en) 2017-02-23
NZ631537A (en) 2017-05-26
CN110042098A (en) 2019-07-23
KR20210151260A (en) 2021-12-13
ZA201507216B (en) 2017-08-30
NZ753018A (en) 2022-01-28
SG10201906382QA (en) 2019-08-27
KR102651423B1 (en) 2024-03-27
US20210395734A1 (en) 2021-12-23
RU2015151199A (en) 2017-06-05
JP2021020901A (en) 2021-02-18
IL272617A (en) 2020-03-31
US9163239B2 (en) 2015-10-20
WO2014179627A2 (en) 2014-11-06
HUE043697T2 (en) 2019-09-30
KR102424855B1 (en) 2022-07-26
WO2014179626A2 (en) 2014-11-06
US9932581B2 (en) 2018-04-03
EP2992009B1 (en) 2020-06-24
MX2019010441A (en) 2019-10-17
JP2016526874A (en) 2016-09-08
AU2021204244B2 (en) 2023-10-19
WO2014179629A3 (en) 2015-01-22
WO2014179629A2 (en) 2014-11-06
EA201891479A1 (en) 2018-11-30
IL242125B (en) 2019-02-28
EP2991656A2 (en) 2016-03-09
JP6639629B2 (en) 2020-02-05
CN110042098B (en) 2023-02-24
US20150126719A1 (en) 2015-05-07
CN105377887A (en) 2016-03-02
JP2020058370A (en) 2020-04-16
AU2014259756B2 (en) 2017-02-23
DK2992098T3 (en) 2019-06-17
KR102482890B1 (en) 2022-12-30
WO2014179627A9 (en) 2015-02-26
AU2017200365C1 (en) 2019-04-18
US9957504B2 (en) 2018-05-01
HK1221475A1 (en) 2017-06-02
SI2992098T1 (en) 2019-06-28
BR112015027319A2 (en) 2017-09-26
BR112015027369B1 (en) 2021-06-08
EP2992009A1 (en) 2016-03-09
US20160076032A1 (en) 2016-03-17
CN111593051A (en) 2020-08-28
IL273312A (en) 2020-04-30
CA2921514C (en) 2023-10-24
RU2015151204A3 (en) 2018-03-27
DK3524680T3 (en) 2020-12-14
KR20160002975A (en) 2016-01-08
US20230151365A1 (en) 2023-05-18
IL296543A (en) 2022-11-01
US10883104B2 (en) 2021-01-05
KR101857707B1 (en) 2018-05-14
US11851655B2 (en) 2023-12-26
EP2991661A4 (en) 2017-02-15
IL242132B (en) 2018-10-31
IL242126B (en) 2019-01-31
CN110079524A (en) 2019-08-02
IL263843B (en) 2020-03-31
US10844379B2 (en) 2020-11-24
JP2019056001A (en) 2019-04-11
BR112015027321A2 (en) 2017-09-26
CN110066795A (en) 2019-07-30
CA2921162A1 (en) 2014-11-06
NZ712737A (en) 2021-08-27
PT2992009T (en) 2020-09-21
CN105378082A (en) 2016-03-02
US20140343123A1 (en) 2014-11-20
JP7339294B2 (en) 2023-09-05
UA120287C2 (en) 2019-11-11
CY1121879T1 (en) 2020-10-14
IL264241B (en) 2020-04-30
BR112015027321A8 (en) 2018-01-02
US20200224198A1 (en) 2020-07-16
AU2019203674A1 (en) 2019-06-27
EP3546579A1 (en) 2019-10-02
IL264580B (en) 2020-04-30
US11299736B1 (en) 2022-04-12
JP6387084B2 (en) 2018-09-05
RS58981B1 (en) 2019-08-30
BR112015027377A8 (en) 2017-10-03
EP2992098A2 (en) 2016-03-09
KR20210129257A (en) 2021-10-27
JP2021107408A (en) 2021-07-29
EP2991656B1 (en) 2019-12-18
CN105392488A (en) 2016-03-09
PE20152002A1 (en) 2016-01-21
EP2991661A1 (en) 2016-03-09
NZ728517A (en) 2021-12-24
BR112015027322A2 (en) 2017-09-26
IL283660A (en) 2021-07-29
IL270464B (en) 2021-07-29
RU2015151200A3 (en) 2019-01-14
AU2019204784A1 (en) 2019-07-25
US20160090596A1 (en) 2016-03-31
EP4155403A1 (en) 2023-03-29
AU2019200820A1 (en) 2019-02-28
JP2024010070A (en) 2024-01-23
AU2019204784B2 (en) 2022-01-27
EP2992098A4 (en) 2017-01-11
EP2992098B1 (en) 2019-03-27
WO2014179627A3 (en) 2015-04-16
KR20230006933A (en) 2023-01-11
EP2992097B1 (en) 2019-11-06
IL261901B (en) 2020-05-31
LT2992098T (en) 2019-07-10
US20150176007A1 (en) 2015-06-25
JP6456362B2 (en) 2019-01-23
AU2017203436A1 (en) 2017-06-08
RU2699985C2 (en) 2019-09-11
NZ631552A (en) 2017-02-24
KR20160002974A (en) 2016-01-08
AU2014259759A1 (en) 2015-10-22
CL2015003217A1 (en) 2016-07-08
JP2020007361A (en) 2020-01-16
CN105378085A (en) 2016-03-02
US9714421B2 (en) 2017-07-25
AU2014259756A1 (en) 2015-10-22
JP6769866B2 (en) 2020-10-14
HK1221485A1 (en) 2017-06-02
KR20240042220A (en) 2024-04-01
KR102235678B1 (en) 2021-04-05
MX2021008899A (en) 2021-08-19
AU2014259755A1 (en) 2015-10-22
US20210130823A1 (en) 2021-05-06
US20160076030A1 (en) 2016-03-17
AU2014259750B2 (en) 2019-02-28
JP2016522683A (en) 2016-08-04
RS60796B1 (en) 2020-10-30
PL2992098T3 (en) 2019-09-30
SG10201801507RA (en) 2018-03-28
JP6592486B2 (en) 2019-10-16
US20180273952A1 (en) 2018-09-27
RU2650510C2 (en) 2018-04-16
HRP20190987T1 (en) 2019-09-20
NZ631512A (en) 2016-10-28
KR20210037752A (en) 2021-04-06
EP3828275A1 (en) 2021-06-02
AU2019204784C1 (en) 2022-11-03
IL273184B (en) 2021-07-29
PL2992009T3 (en) 2020-11-30
RU2018112167A (en) 2019-03-07
US10927372B2 (en) 2021-02-23
DOP2016000287A (en) 2017-02-15
AU2018267625B2 (en) 2020-09-10
PH12015502493B1 (en) 2016-02-22
BR112015027369A2 (en) 2017-09-26
AU2017203436B2 (en) 2018-10-18
BR112015027322A8 (en) 2018-01-02
AU2024200296A1 (en) 2024-02-08
JP6652602B2 (en) 2020-02-26
CR20190269A (en) 2019-09-13
CA2921509A1 (en) 2014-11-06
MX2015015239A (en) 2016-10-03
CN113293163A (en) 2021-08-24
IL273184A (en) 2020-04-30
SG11201508870VA (en) 2015-11-27
CN108064162B (en) 2021-12-03
MY178929A (en) 2020-10-23
PH12018501963A1 (en) 2020-07-20
AU2022202770A1 (en) 2022-05-19

Similar Documents

Publication Publication Date Title
AU2019204784C1 (en) Compositions and methods for modulating hbv and ttr expression
NZ728517B2 (en) Compositions and methods for modulating ttr expression
BR122018009831B1 (en) COMPOUNDS COMPRISING A MODIFIED OLIGONUCLEOTIDE AND A GROUP OF CONJUGATE, A COMPOSITION COMPRISING SUCH COMPOUNDS AND USES THEREOF IN THE TREATMENT OF TRANSTHYRETIN AYLOIDOSIS
EA046363B1 (en) OLIGOMERIC COMPOUNDS FOR MODULATING TTR EXPRESSION

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20200430

FZDE Discontinued

Effective date: 20220907

FZDE Discontinued

Effective date: 20220907

FZDC Discontinued application reinstated

Effective date: 20230310

FZDC Discontinued application reinstated

Effective date: 20230310

FZDC Discontinued application reinstated

Effective date: 20230310

FZDC Discontinued application reinstated

Effective date: 20230310

FZDC Discontinued application reinstated

Effective date: 20230310

FZDC Discontinued application reinstated

Effective date: 20230310