WO1996010030A1 - Oligomers having heteroatom-linked conjugate groups - Google Patents

Oligomers having heteroatom-linked conjugate groups Download PDF

Info

Publication number
WO1996010030A1
WO1996010030A1 PCT/US1995/013038 US9513038W WO9610030A1 WO 1996010030 A1 WO1996010030 A1 WO 1996010030A1 US 9513038 W US9513038 W US 9513038W WO 9610030 A1 WO9610030 A1 WO 9610030A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
molecule
group
aromatic
alkyl
Prior art date
Application number
PCT/US1995/013038
Other languages
French (fr)
Inventor
Phillip Dan Cook
Kelly Teng
Original Assignee
Isis Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Isis Pharmaceuticals, Inc. filed Critical Isis Pharmaceuticals, Inc.
Priority to US08/809,239 priority Critical patent/US5998603A/en
Priority to AU38923/95A priority patent/AU3892395A/en
Publication of WO1996010030A1 publication Critical patent/WO1996010030A1/en

Links

Classifications

    • 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
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic System
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/18Compounds having one or more C—Si linkages as well as one or more C—O—Si linkages
    • C07F7/1804Compounds having Si-O-C linkages
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals

Definitions

  • This invention is directed to oligomeric compounds that are functionalized to include covalently bound groups.
  • the oligomeric compounds include a tetrahydrofuran moiety that is functionalized with a pendant conjugate group.
  • Oligonucleotides and their analogs have been developed and used in molecular biology in certain procedures as probes, primers, linkers, adapters, and gene fragments. Modifications to oligonucleotides used in these procedures include labeling with non-isotopic labels, e.g. fluorescein, biotin, digoxigenin, alkaline phosphatase, or other reporter molecules. Other modifications have been made to the ribose phosphate backbone to increase the nuclease stability of the resulting analog.
  • non-isotopic labels e.g. fluorescein, biotin, digoxigenin, alkaline phosphatase, or other reporter molecules.
  • Other modifications have been made to the ribose phosphate backbone to increase the nuclease stability of the resulting analog.
  • modifications include use of methyl and other alkyl phosphonates, phosphorothioates, phosphorodithioate, phosphoamidate and phosphotriester linkages, and 2'-O-methyl ribose sugar units. Further modifications include modification made to modulate uptake and cellular distribution. Phosphorothioate oligonucleotides are presently being used in human clinical trials for various disease states, including use as antiviral agents. In view of the success of these oligonucleotides for both diagnostic and therapeutic uses, there exists an ongoing demand for improved oligonucleotide analogs.
  • Oligonucleotides and like molecules can interact with native DNA and RNA in several ways. One of these is duplex formation between an oligonucleotide and a single stranded nucleic acid. A further method is via triplex formation between an oligonucleotide and double stranded DNA to form a triplex structure.
  • oligomeric compounds Naturally occurring or synthetic oligonucleotides, together with hybrid species having both synthetic and natural components, can collectively be referenced as "oligomeric compounds.” Because of their properties, these oligomeric compounds are known to be useful in a number of different areas. They can be used as probes in cloning, blotting procedures, and in applications such as fluorescence in situ hybridization (FISH). Also, since local triplex formation inhibits gene transcription, such oligomeric compounds can be used to inhibit gene transcription. Labeled oligomers can also be used to directly map DNA molecules, such as by tagging an oligomer with a fluorescent label and effecting hybridization to complementary sequences in duplex DNA.
  • FISH fluorescence in situ hybridization
  • Oligomers can also be used as identification tags in combinatorial chemical libraries as is disclosed in patent publication WO 94/08051 and Ohlmeyer et al., Proc. Natl. Acad. Sci. USA, 1993, 90, 10922-10926.
  • the present invention provides compounds useful in diagnostic assays and as research reagents, as well as methods and intermediates for the preparation thereof.
  • R L is azido or a group of formula: R C - [Y] e -Z- in which
  • Z is O, S or HN
  • Y is a bivalent linker
  • e is 0 or 1;
  • R C is alkyl, alkenyl, alkynyl, O-alkyl, O-alkenyl, O- alkynyl, aryl, alkaryl, phosphinyl, a polyamine, a polyether, polyglycol, a steroid molecule, a reporter molecule, an aromatic or nonaromatic ring system, an aromatic lipophilic molecule, a non-aromatic lipophilic molecule, a reporter enzyme, a peptide, a protein, a water soluble vitamin, a lipid soluble vitamin, a carbohydrate, a terpene molecule, a phospholipid, an intercalator, a cell receptor binding molecule, a crosslinking agent, or a porphyrin;
  • B x is a nucleobase
  • X is H, OH, O-alkyl, O-alkoxyalkyl, O-alkylamino or
  • Q is O, S, CH 2 , CHF or CF 2 ;
  • R D is H, hydroxyl, an activated phosphorous group, a nucleoside, an activated nucleotide, a nucleotide, an oligonucleotide, an oligonucleoside or a protected derivative thereof.
  • L is a group of formula: R 7 -[S-(CH 2 ) q ] u -[HN-(CH 2 ) n ] t -[O-(CH 2 ) m ] v -Z- in which
  • Z is O, S or HN
  • t, u and v are each independently integers from 0 to
  • n, n and q are each independently integers from 1 to
  • R 7 is R C , H or a protecting group
  • X, B x , R C and R D are defined as above.
  • t and u are 0, or u and v are zero, or t and v are zero.
  • t and u are 0 and m is 2.
  • t and v are 0 and q is 2
  • u and v are 0 and n is 2.
  • u is 0 and each of t and v are 1, and m and n are each 2.
  • R C , B x , X, Q, and R D are defined as above.
  • R C is a polyether or a polyamine.
  • R C is a steroid molecule, preferably cholic acid, deoxycholic acid, dehydrocholic acid, cortisone, digoxigenin, testosterone, cholesterol or 3- trimethylaminomethylhydrazido cortisone.
  • R C is a water soluble vitamin, preferably thiamine, riboflavin, nicotinic acid, pyridoxal phosphate, pyridoxine, pyridoxamine, deoxy- pyridoxine, pantothenic acid, biotin, folic acid, 5'- deoxyadenosylcobalamin, inositol, choline or ascorbic acid.
  • R C is a lipid soluble vitamin, preferably a retinal, a retinol, retinoic acid, ⁇ - carotene, vitamin D, cholecalciferol, a tocopherol, or a phytol.
  • R C is a protein, preferably a phosphodiesterase, a peroxidase, a phosphatase or a nuclease.
  • R C is a reporter molecule, preferably a chromaphore, a fluorophore or a radiolabel-containing moiety.
  • Preferred fluorophores include fluorescein, chrysine, anthracene and perylene.
  • X is H, OH, F, O-alkyl having from one to six carbons, O-alkylamino having from one to six carbons or O-alkoxyalkyl having from one to six carbons.
  • X is H or OH mimicking natural deoxyribo and ribo sugar analogs.
  • X is F, O-alkyl having from one to six carbons, alkylamino having from one to six carbons or O-alkoxyalkyl having from one to six carbons such that the compounds are homologous to certain nucleotide species that have either greater nuclease resistance or high binding affinity.
  • R D is H or OH; or R D is an activated phosphorous group, or R D is an oligonucleotide.
  • the present invention also provides novel synthons useful for the preparation of monomeric and polymeric conjugate compounds.
  • these synthons have the structure: wherein J is a leaving group and Q, R D , B x and X are as defined above.
  • J is OH, SH, NH 2 , trifluoromethylsulfonyl, methylsulfonyl, halogen, O- trichloroacetimidate, acyloxy, dialkyl phosphite, 2,4,6- trichlorophenyl, p-toluenesulfonyl, 4-dimethylaminoazo- benzenesulfonyl or 5-dimethylaminonaphthalenesulfonyl .
  • Preferred methods comprise the steps of:
  • R E is an activated phosphorous group
  • W is H or a hydroxyl protecting group
  • k is an integer from 0 to 50
  • E is a phosphorous linking species including phosphodiester, phosphotriester, phosphorothioate, phosphorodithioate, alkyl phosphonates especially methyl phosphonates and phosphoramidates phosphorous linking groups
  • Q, R L , B x and X are as defined above.
  • the present invention also provides methods for forming a 5'-desmethyl conjugated monomer having structure:
  • J is OH, SH, NH 2 , trifluoromethylsulfonyl, methylsulfonyl, halogen, O- trichloroacetimidate, acyloxy, dialkyl phosphite, 2,4,6- trichlorophenyl, p-toluenesulfonyl, 4-dimethylaminoazo- benzenesulfonyl or 5-dimethylaminonaphthalenesulfonyl; more preferably trifluoromethylsulfonyl, methylsulfonyl, halogen or acyloxy.
  • nucleoside refers to a unit composed of a heterocyclic base and a sugar, generally a pentose sugar.
  • the heterocyclic base typically is guanine, adenine, cytosine, thymine or uracil, and the sugar is normally deoxyribose, i.e., erythro-pento- furanosyl, or ribose, i.e., ribopentofuranosyl.
  • Synthetic sugars also are known, including arabino, xylo or lyxo pentofuranosyl sugars and hexose sugars. Further synthetic sugar are the 4'-deoxy-4'-thio sugars (both 2'-deoxy and ribo varieties) that have been described by Secrist et al., J. Med. Chem. 1991, 34, 2361-2366 and patent application PCT/FR93/00115, respectively, (whereby Q in the above structures is S).
  • nucleoside or other nucleic acid species shall be understood to refer to naturally occurring deoxyribose and ribose sugars and also to sugar species which can replace the sugar moieties of naturally occurring nucleic acids.
  • nucleobase or heterocyclic base portion of a nucleoside or other nucleic acid species shall be understood to refer to naturally-occurring nucleobases, modified derivatives thereof, or to synthetic nucleobases. Synthetic nucleobases are those which can replace one or more of the naturally occurring base moieties of wild type nucleic acids.
  • inter-sugar linkages shall be taken to include moieties that join the sugar portions of nucleosides or nucleotides.
  • nucleotide refers to a nucleoside having a phosphate group esterified to one of its 2', 3' or 5' sugar hydroxyl groups.
  • the phosphate group normally is a monophosphate, diphosphate or triphosphate.
  • oligonucleotide refers to a plurality of monophosphate nucleotide units that are typically formed from naturally occurring bases and pentofuranosyl sugars joined by native phosphodiester bonds in a specific sequence. A homo-oligonucleotide is formed from nucleotide units having the same heterocyclic base, e.g. polyadenosine.
  • oligonucleotide analog has been used in various published patent application specifications and other literature to refer to molecular species that are similar to oligonucleotides, but that have non-naturally occurring portions.
  • oligonucleotide analog has been used to denote structures having altered inter-sugar linkages including phosphorothioate, phosphorodithioate, methyl phosphonate, phosphotriester or phosphoramidate inter-nucleoside linkages in place of phosphodiester inter- nucleoside linkages; purine and pyrimidine heterocyclic bases other than guanine, adenine, cytosine, thymine or uracil and sugars having other than the ⁇ pentof ⁇ ranosyl configuration or sugars having substituent groups at their 2' position or substitutions for one or more of the hydrogen atoms.
  • modified oligonucleotide also has been used in the literature to denote such structures.
  • oligonucleotide mimic has also been used to refer to macromolecular moieties that function similarly to or “mimic” the function of oligonucleotides but have non- naturally occurring inter-sugar linkages. Oligonucleotide mimics thus can have natural or altered or non-naturally occurring sugar moieties and natural or altered or non- naturally occurring base moieties in combination with non- naturally occurring dephospho linkages. Certain dephospho linkages have been reviewed by Uhlmann, E. and Peyman, A., "Oligonucleotide Analogs Containing Dephospho Internucleo- side Linkages" in Methods in Molecular Biology, Chapter 16, Oligonucleotide Synthetic Protocols, S. Agrawal, Ed., The Humana Press, Inc., Totowa, NJ, 1993.
  • oligomers is intended to encompass oligonucleotides, oligonucleotide analogs, oligonucleosides or oligonucleotide-mimicking macromolecules.
  • oligomers refers to nucleosides or nucleoside analogs that are joined together via either natural phosphodiester bonds or via other linkages.
  • the nucleoside compounds of the invention lack the 5'-methylene group present in conventional pentofuranosyl nucleosides.
  • a heteroatom occupies the position normally occupied, by the missing 5'-methylene group.
  • these compounds can be considered as 4'-desmethyl pentofuranosyl nucleosides.
  • the lack of a 5'-methylene carbon on the "sugar portion" of the nucleosides of the invention require their designation as tetrahydrofuranyl moieties.
  • established hierarchical or priority nomenclature rules are followed.
  • Compounds of the invention include 4'-desmethyl nucleoside monomers modified to bear conjugate groups on their 4'-carbons (i.e., 4'-desmethyl conjugate monomers) or oligomers which contain such modified nucleosides (4'- desmethyl conjugate oligomers).
  • 4'-desmethyl nucleoside monomers modified to bear conjugate groups on their 4'-carbons
  • oligomers which contain such modified nucleosides (4'- desmethyl conjugate oligomers.
  • the "E" group can be any of the commonly use oligonucleotide phosphorous linkage including phosphodiester, phosphotriester, phosphorothioate, phosphorodithioate, alkyl phosphonate and phosphoamidate linkages.
  • the pendant conjugate group can be a group of formula: R C -[S-(CH 2 ) q ] u -[HN-(CH 2 ) n ] t -[O-(CH 2 ) m ] v -Z- wherein Z is O, NH or S, preferably O or NH, t, u and v are each independently integers from 0 to 200; m, n and q are each independently integers from 1 to 4; and R C is alkyl, alkenyl, alkynyl, aryl, alkaryl, phosphinyl, O-alkyl, O- alkenyl, O-alkynyl, a polyglycol, a polyamine, a polyether, a steroid molecule, an aromatic or nonaromatic ring system, a reporter molecule, an aromatic lipophilic molecule, a nonaromatic lipophilic molecule, a reporter enzyme, a peptide, a protein
  • oligomers of the invention at least one 4'- desmethyl nucleoside unit incorporated therein has been derivatized to bear a pendant conjugate group as described above.
  • the oligomeric compound is useful, for example, in a diagnostic or other preparation that includes a nucleic acid binding agent to assist in identification of the oligomeric compound, to aid in transfer of the compound across cellular membranes, or to impart other properties.
  • a diagnostic or nucleic acid binding agent is formed from an oligomeric compound of the invention wherein the oligomeric compound includes monomeric units bearing natural or non-natural occurring bases in a sequence that is complementary to and will specifically hybridize with a region of an R ⁇ A or D ⁇ A of interest.
  • a functionalized oligomeric compound according to the invention can be characterized as a substituent-bearing (e.g., steroid- bearing) oligomeric compound.
  • oligomeric compounds will have at least one pendant group attached thereto to modulate their activity.
  • the compounds of the inventions have various uses including being useful as research reagents and in diagnostic assays.
  • diagnostic assay compounds of the inventions are used to isolate the effects of one cellular adhesion molecule from those of a further cellular adhesion molecule in a protein expression assay.
  • ICAM-1 intercellular adhesion molecule-1
  • the oligomeric compounds bearing pendant conjugate groups of the present invention can be used to bind to various other target molecules.
  • Target molecules of the present invention can include any of a variety of biologically significant molecules.
  • Other such target molecules can be nucleic acids, carbohydrates, glycoproteins or other proteins.
  • the functionalized oligomeric compounds of the invention bind with complementary strands of RNA or DNA.
  • the oligomeric compound and the RNA or DNA strand can be considered to be complementary strands which are "duplexed" in a manner analogous to native, double-stranded DNA.
  • the individual strands are positioned in such a manner with respect to one another so as to allow Watson-Crick type, Hoogsteen type or another type hybridization of the heterocyclic bases of one strand to the heterocyclic bases of the opposing strand.
  • binding to nucleic acids can be practiced against nucleic acids from a variety of sources including organisms ranging from unicellular prokaryotes and eukaryotes to multicellular eukaryotes.
  • the nucleic acid from any organism that utilizes DNA-RNA transcription or RNA-protein translation as a fundamental part of its hereditary, metabolic or cellular control is susceptible to such binding. Seemingly diverse organisms such as bacteria, yeast, virus, protozoa, algae, all plant and all higher animal forms, including warm-blooded animals, are sources of such nucleic acid.
  • each of the cells of multicellular eukaryotes includes both DNA-RNA transcription and RNA-protein translation as an integral part of their cellular activity
  • nucleic acid binding for various purposes, e.g. diagnostics, can also be practiced on such cellular populations.
  • organelles e.g., mitochondria and chloroplasts
  • single cells, cellular populations or organelles can also be included within the definition of organisms that are capable of being treated with nucleic acid binders.
  • the target molecule is a protein such as an immunoglobulin, receptor, receptor binding ligand, antigen or enzyme and more specifically can be a phospholipase, tumor necrosis factor, endotoxin, interleukin, plasminogen activator, protein kinase, cell adhesion molecule, lipoxygenase, hydrolase or transacylase.
  • the target molecules can be important regions of the human immunodeficiency virus, Candida, herpes viruses, papillomaviruses, cytomegalovirus, rhinoviruses, hepatitis viruses, or influenza viruses.
  • the target molecules can be regions of an oncogene.
  • the target molecule is ras 47-mer stem loop RNA, the TAR element of human immunodeficiency virus or the gag-pol stem loop of human immunodeficiency virus (HIV).
  • targets can induce cellular activity.
  • a target can induce interferon.
  • Binding also can be practiced against transcription factors.
  • the transcription factor or other target molecule need not be purified. It can be present, for example, in a whole cell, in a humoral fluid, in a crude cell lysate, in serum or in other humoral or cellular extracts.
  • purified transcription factor or a purified form of another target molecule is also useful in some aspects of the invention.
  • synthetically prepared transcription factors or other target molecules can be useful.
  • a transcription factor or other target molecule also can be modified, such as by biotinylation or radiolabeling.
  • a synthetically prepared transcription factor can incorporate one or more biotin molecules during synthesis or can be modified post- synthesis.
  • Transcription factors are DNA- or RNA-binding proteins that regulate the expression of genes. HIV tat and c-rel are examples of transcription factors which regulate the expression of genes. Also encompassed by the term are DNA and RNA binding proteins which are not strictly considered transcription factors, but which are known to be involved in cell proliferation. These transcription factors include c-myc, fos, and jun. Methods of the present invention are particularly suitable for use with transcription factors as target molecules since transcription factors generally occur in very small cellular quantities.
  • nucleic acid binding can be used to treat objects (glasswares, petri dishes, instruments or the like) to sterilize, sanitize or disinfect such objects that may harbor an organism, e.g., bacterial, protozoan, viral, fungal or other infectious or non- infectious agent on the object.
  • an organism e.g., bacterial, protozoan, viral, fungal or other infectious or non- infectious agent on the object.
  • the compounds of the invention will kill, abrogate, inhibit, curtail or other wise control, eradicate or render harmless the cellular growth or gene expression of such organisms on the object.
  • Such nucleic acid binding can also be used to render such organisms incapable or otherwise unable to reproduce themselves.
  • nucleic acid binding properties of the compounds of the invention can be used to form duplex structures with an unknown oligomer species for identification of that species by gel analysis including slab and capillary gel electrophoresis.
  • the compounds of the invention can be used as tags in identification of other compounds in combinatorial libraries.
  • Preferred nucleobase units for oligomeric compounds of the invention include naturally occurring or synthetic purine or pyrimidine heterocyclic bases, including but not limited to adenine, guanine, cytosine, thymine, uracil, 5- methylcytosine, hypoxanthine or 2-aminoadenine.
  • Other such heterocyclic bases include 2-methylpurine, 2,6-diaminopurine, 6-mercaptopurine, 2,6-dimercaptopurine, 2-amino-6- mercaptopurine, 5-methylcytosine, 4-amino-2-mercaptopyrimidine, 2,4-dimercaptopyrimidine and 5-fluorocytosine.
  • Representative heterocyclic bases are disclosed in U.S. Patent No. 3,687,808 (Merigan, et al.), which is incorporated herein by reference.
  • alkyl groups of the invention include but are not limited to C 1 -C 12 straight and branched chained alkyls such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl, isopropyl, 2 -butyl, isobutyl, 2-methylbutyl, isopentyl, 2-methylpentyl, 3-methylpentyl, 2-ethylhexyl and 2-propylpentyl.
  • Alkenyl groups include but are not limited to unsaturated moieties derived from the above alkyl groups including but not limited to vinyl, allyl and crotyl.
  • Alkynyl groups include unsaturated moieties having at least one triple bond that are derived from the above alkyl groups including but are not limited to ethynyl and propargyl.
  • Alkanoyl groups according to the invention are alkyl, alkenyl or alkynyl groups attached through a carbonyl group.
  • aryl is intended to denote monocyclic and polycyclic aromatic groups including, for example, phenyl, naphthyl, xylyl, pyrrole, and furyl groups.
  • aryl groups e.g., imidazo groups
  • preferred aryl groups have 6 to about 14 carbon atoms, more preferably 6 to about 10 carbon atoms.
  • Aralkyl and alkaryl groups according to the invention each include alkyl and aryl portions. Aralkyl groups are attached through their alkyl portions, and alkaryl groups are attached through their aryl portions. Benzyl groups provide one example of an aralkyl group, and p-tolyl provides an example of an alkaryl group.
  • Alkylamino and aminoalkyl groups according to the invention each include amino and alkyl portions. Alkylamino groups are attached through their amino portions, and aminoalkyl groups are attached through their alkyl portions. Methylamino groups provide one example of an alkylamino group, a ⁇ -aminobutyl group is one example of an aminoalkyl group.
  • a ring system is defined to include two or more single rings that join together to form an extended or condensed ring.
  • ring systems include extended aromatic systems, alicyclic systems, araalicyclic systems, and bicyclic systems. Examples include aromatic, alicyclic and mixed aromatic- alicyclic (araalicyclic) multiple ring systems, bicyclic systems, non-aromatic multiple ring systems such as adamantane, decalin, and norbornane.
  • Ring systems also include, but are not limited to, naphthalene, tetrahydronaphthalene (tetralin), anthracene, phenanthrene, fluorene, pyrene, coronene, azulene, cluorene, benzonaphthene, benzo [8] annulene, pentalene, heptalane, octalene, indene, isoindene biphenyl, biphenylene and triphenylene condensed rings.
  • alkyl, alkaryl, aralkyl and aryl are intended to denote both substituted (e.g., halogenated and hydroxylated) and unsubstituted moieties.
  • Halogens include fluorine, chlorine and bromine.
  • Suitable heterocyclic groups include but are not limited to imidazole, tetrazoie, triazole, pyrrolidine, piperidine, piperazine and morpholine.
  • Heterocycloalkyl groups are cyclic alkyl groups containing a heretoatom.
  • Heterocycloalkaryl groups are aryl heterocycles bearing at least one alkyl substituent.
  • Amines include amines of all of the above alkyl, alkenyl and aryl groups including primary and secondary amines and "masked amines" such as phthalimide. Amines are also meant to include polyalkylamino compounds and aminoalkylamines such as aminopropylamine and further heterocycloalkylamines such as imidazol-1, 2 or 4 -yl-propylamine.
  • Substituent groups for the above as well as for other moieties listed below include but are not limited to other alkyl, haloalkyl, alkenyl, alkynyl, alkoxy, thioalkoxy, haloalkoxy and aryl groups as well as halogen, hydroxyl, amino, azido, carboxy, cyano, nitro, mercapto, sulfides, sulfones, sulfoxides, keto, carboxy, nitrates, nitrites, nitroso, nitrile, trifluoromethyl, O-alkyl, S-alkyl, NH- alkyl, amino, silyl, amides, ester, ethers, carbonates, carbamates, ureas, imidazoles, intercalators, conjugates, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligonucleotides, and groups that
  • substituent groups also include rhodamines, coumarins, acridones, pyrenes, stilbenes, oxazolopyridocarbazoles, anthraquinones, phenanthridines, phenazines, azidobenzenes, psoralens, porphyrins and cholesterols.
  • rhodamines coumarins, acridones, pyrenes, stilbenes, oxazolopyridocarbazoles, anthraquinones, phenanthridines, phenazines, azidobenzenes, psoralens, porphyrins and cholesterols.
  • CF 3 is CF 3 .
  • polyether means a linear or branched alkyl chain periodically interrupted by oxygen atoms.
  • a polyethers is polyethylene glycol.
  • polyglycol as used herein, includes compounds such as glycerol, and derivative groups such as 2,3-dialkoxypropyl groups.
  • polyamine as used herein includes the nitrogen analogs of such structures, and the term polythioether includes the sulfur analogs of such structures.
  • Carbohydrates according to the invention are inclusive of pentose, hexose and higher sugars, and the polymeric species thereof.
  • Representative carbohydrates include glucose and galactose and their derivatives including, as for example, glycals, glycal epoxides and glycosides.
  • Terpenes are known in the art as oligomers of isoprene, particularly the dipentenes, pinenes, and myrcenes. Included within the definition of terpene molecules are terpene derivatives such as camphor and menthol.
  • phospholipid as used herein includes those compounds which upon hydrolysis yield phosphoric acid, an alcohol and one or more fatty acids.
  • Representative examples of phospholipids include lecithin, cephalin and sphingomyelin.
  • groups that enhance the pharmacodynamic properties include groups that improve oligonucleotide uptake, enhance oligonucleotide resistance to degradation, and/or strengthen sequence-specific hybridization with RNA.
  • groups that enhance the pharmaco- kinetic properties include groups that improve oligonucleotide uptake, distribution, metabolism or excretion.
  • reporter molecule and “reporter enzyme” are inclusive of those molecules or enzymes that have physical or chemical properties that allow them to be identified in gels, fluids, whole cellular systems, broken cellular systems and the like utilizing physical properties such as spectroscopy, radioactivity, colorimetric assays, fluorescence, and specific binding.
  • reporter molecules particularly useful as reporter molecules are fluorophores, chromaphores and radiolabel-containing moieties. Fluorophores are molecules detectable by fluorescence spectroscopy. Examples of preferred fluorophores are fluorescein and rhodamine dyes and acridines.
  • Chromaphores are molecules capable of detection by visible or ultraviolet (UV-VIS) absorbance spectroscopy.
  • UV-VIS ultraviolet
  • chromaphores polynuclear aromatics such as anthracene, perylene, pyrene, rhodamine and chrysene.
  • Radiolabel-containing moieties are molecules incorporating at least one radioactive atom, such as 3 H or 14 C, enabling detection thereby.
  • Reporter enzymes may be detected directly or via their enzymatic products by any of the methods mentioned above. Particularly useful as reporter enzymes are alkaline phosphatase and horseradish peroxidase.
  • Steroid molecules according to the invention include those chemical compounds that contain a perhydro-1, 2- cyclopentanophenanthrene ring system.
  • Particularly useful as steroid molecules are the bile acids including cholic acid, deoxycholic acid and dehydrocholic acid; steroids including cortisone, digoxigenin, testosterone and cholesterol and cationic steroids such as cortisone having a trimethylaminomethyl hydrazide group attached via a double bond at the 3 -position of the cortisone ring (3- trimethylaminomethylhydrazido cortisone).
  • Proteins and peptides are utilized in their usual sense as polymers of amino acids. Normally peptides comprise such polymers that contain a smaller number of amino acids per unit molecule than do the proteins. Particularly useful as peptides and proteins are sequence- specific peptides and proteins including phosphodiesterases, peroxidases, phosphatases and nucleases. Such peptides and proteins include, but are not limited to, SV40 peptide, RNase A, RNase H and Staphylococcal nuclease.
  • Lipophilic molecules include naturally-occurring and synthetic aromatic and non-aromatic moieties such as fatty acids, esters and alcohols, other lipid molecules, cage structures such as adamantane and buckminsterfullerenes, and aromatic hydrocarbons such as benzene, perylene, phenanthrene, anthracene, naphthalene, pyrene, chrysene, and naphthacene.
  • Particularly useful as lipophilic molecules are alicyclic hydrocarbons, saturated and unsaturated fatty acids, waxes, terpenes and polyalicyclic hydrocarbons including adamantane and buckminsterfullerenes.
  • Alkylators according to the invention are moieties that can effect attachment of electrophilic groups to targeted molecular structures. Representative alkylators are disclosed by Meyer, et al., J. Am. Chem. Soc. 1989, 111, 8517.
  • Intercalators are polycyclic aromatic moieties that can insert between adjacent base pairs without affecting normal Watson-Crick base pairing, and include hybrid intercalator/ligands such as the photonuclease/intercalator ligand 6-[[[9-[[6-(4-nitrobenzamido)hexyl]amino] acridin-4- yl]carbonyl]amino] hexanoylpentafluorophenyl ester.
  • This compound has two noteworthy features: an acridine moiety that is an intercalator and a p-nitrobenzamido group that is a photonuclease.
  • Other representative intercalators are disclosed by Manoharan, M., Antisense Research and Applications, Crooke and Lebleu, eds., CRC Press, Boca Raton, 1993.
  • Cell receptor binding molecules according to the invention are vitamins and carbohydrate moieties for which specific receptors exist within a cell.
  • Representative cell receptor binding molecules are disclosed by Application Serial No. PCT/US92/09196, filed October 23, 1992, the contents of which are incorporated herein by reference.
  • Crosslinking agents are moieties that can effect intrastrand or interstrand covalent binding of RNA and/or DNA, and include photo-crosslinking agents.
  • photocrosslinking agents include aryl azides such as N- hydroxysuccinimidyl-4-azidobenzoate (HSAB) and N- succinimidyl-6(-4'-azido-2'-nitrophenylamino) hexanoate
  • SANPAH Aryl azides conjugated to oligomers will effect crosslinking with nucleic acids and proteins upon irradiation.
  • Other representative crosslinking agents are disclosed in International Patent Application Serial No. PCT/US93/02059, filed March 5, 1993, which is incorporated herein by reference.
  • crown amines are disclosed by Studer, et al., Helv. Chim. Acta 1986, 69, 2081 and Smith-Jones, et al., Bioconjugate Chem. 1991, 2, 415.
  • Vitamins according to the invention generally can be classified as water soluble or lipid soluble.
  • Water soluble vitamins include thiamine, riboflavin, nicotinic acid or niacin, the vitamin B 6 pyridoxal group,, pantothenic acid, biotin, folic acid, the B 12 cobalamin coenzymes, inositol, choline and ascorbic acid.
  • Lipid soluble vitamins include the vitamin A family, vitamin D, the vitamin E tocopherol family and vitamin K (and phytols).
  • the vitamin A family including retinoic acid and retinol, are absorbed and transported to target tissues through their interaction with specific proteins such as cytosol retinol-binding protein type II (CRBP-II), Retinol-binding protein (RBP), and cellular retinol-binding protein (CRBP). These proteins, which have been found in various parts of the human body, have molecular weights of approximately 15 kD. They have specific interactions with compounds of the vitamin A family, especially, retinoic acid and retinol.
  • CRBP-II cytosol retinol-binding protein type II
  • RBP Retinol-binding protein
  • CRBP cellular retinol-binding protein
  • the vitamin A family of compounds can be attached to oligomers of the invention via acid or alcohol functionalities found in the various family members. For example, conjugation of an N-hydroxysuccinimide ester of an acid moiety of retinoic acid to an amine function of a pendant group of the oligomer results in linkage of the vitamin A compound to the oligomer, via an amide bond.
  • standard esterification chemistries may be used to attach the acid moiety of the retinoic acid group to a 4'-oxygen of a compound of the invention, or to a hydroxyl function of a pendent group thereof.
  • ⁇ -Tocopherol (vitamin E) and other tocopherols can be similarly conjugated to oligomers also to enhance uptake due to their lipophilic character.
  • the lipophilic vitamin, vitamin D, and its ergosterol precursors can be conjugated to oligomers through their hydroxyl groups by first activating the hydroxyl groups by forming, for example, hemisuccinate esters. Conjugation then is effected, as for instance, to an aminolinker pendant from the oligomer, or through other suitable functional groups described herein.
  • vitamins that can be conjugated to oligomers through hydroxyl groups on the vitamins include thiamine, riboflavin, pyridoxine, pyridoxamine, pyridoxal, deoxypyridoxine.
  • Lipid soluble vitamin K's and related quinone-containing compounds can be conjugated via carbonyl groups on the quinone ring.
  • the phytol moiety of vitamin K may also serve to enhance binding of the oligomers to cells.
  • Pyridoxal phosphate is the coenzyme form of Vitamin B 6 .
  • Vitamin B 6 is also known as pyridoxine.
  • Pyridoxal has specific B 6 -binding proteins. The role of these proteins in pyridoxal transport has been studied by Zhang and McCormick, Proc. Natl. Acad. Sci. USA, 1991 88, 10407.
  • Zhang and McCormick showed that a series of N-(4'-pyridoxyl) amines, in which several synthetic amines were conjugated at the 4'- position of pyridoxal, were able to enter cells by a process facilitated by the B 6 transporter.
  • Zhang and McCormick also demonstrated the release of these synthetic amines within the cell.
  • pyridoxal family members include pyridoxine, pyridoxamine, pyridoxal phosphate, and pyridoxic acid.
  • Pyridoxic acid, niacin, pantothenic acid, biotin, folic acid and ascorbic acid can be conjugated to oligomers using N-hydroxysuccinimide esters as described above for retinoic acid.
  • RNA cleavers include o- phenanthroline/Cu complexes and Ru (bipyridine) 3 2+ complexes.
  • the Ru (bipyridine) 3 2+ complexes interact with nucleic acids and cleave nucleic acids photochemically.
  • Metal chelators include EDTA, DTPA, and o-phenanthroline.
  • Alkylators include compounds such as iodoacetamide.
  • Porphyrins include porphine, its substituted forms, and metal complexes.
  • Pyrenes include pyrene and other pyrene-based carboxylic acids that can be conjugated using protocols similar to those specified above.
  • protecting groups are known in the art for protecting the several functional groups of the compounds of the invention during synthesis.
  • Protecting groups are known per se as chemical functional groups that can be selectively appended to and removed from functionalities, such as hydroxyl groups and carboxyl groups. These groups are present in a chemical compound to render such functionality inert to chemical reaction conditions to which the compound is exposed.
  • Protecting groups useful in the context of the present invention include but are not limited to hydroxyl protecting groups such as t-butyldiphenylsilyl, t-butyldimethylsilyl, and dimethoxytrityl groups, thiol protecting groups such as S- trityl, S-p-methoxybenzylthioether, S-p-nitrobenzyl- thioether, and S-t-butylthioether.
  • hydroxyl protecting groups such as t-butyldiphenylsilyl, t-butyldimethylsilyl, and dimethoxytrityl groups
  • thiol protecting groups such as S- trityl, S-p-methoxybenzylthioether, S-p-nitrobenzyl- thioether, and S-t-butylthioether.
  • the present invention is directed to nucleoside monomers that bear at least one conjugate group at the 4'-position.
  • the conjugate-bearing nucleoside is the 5'-terminal residue oligomer.
  • the conjugate group is a polyamine, and in other preferred embodiments the conjugate group is a polyether. In further preferred embodiments the conjugate group is a polythioether.
  • the conjugate groups of the preceding paragraph can further serve as bivalent linkers for other conjugate groups.
  • a hydroxyl, amino or thiol terminated polyether, polyamine or polythioether is used a linker and the terminal group is then reacted with a suitable functional group on a conjugate molecules to link the conjugate molecule to the polyether, polyamine or polythioether.
  • Suitable functional groups for reacting with hydroxyl, amine or thiol groups include a gamut of groups known in the organic chemical arts including, but not limited to, acid chlorides, anhydrides, cyclic anhydrides, alkyl halides, organometallics, chloroformates, isocynates and the like.
  • terminal groups can be utilized on the linker including, but not limited to, hydrazines, aldehydes, acids, hydroxylamines, semicarbazides, thiosemicarbazides, hydrazones, hydrazides and the like.
  • Protected forms of such compounds can also be used, as for example, protected aldehydes including, C- formyl, o-methylaminobenzenethio, aryl substituted imidazolidino moieties and the like.
  • protected thiol include trityl thiol that can be deblocked allowing a disulfide bonds to be formed between the linker and the a sulfur bearing conjugate group to join the conjugate group to the linker.
  • Various nitrogen linkages can be used to join the conjugate groups including hydrazones, oximes, hydrazide-hydrazone, semicarbazone and semithiocarbazones.
  • polyamine species including polyamines, as used herein refers to species that have a plurality of nitrogen atoms thereon.
  • Polyamines include primary amines, hydrazines, semicarbazines, thiosemicarbazines and similar nitrogenous species.
  • Such species can be symmetrical species such as polyamine containing polymers or they can be unsymmetrical wherein the amine functionalities of the polyamine are separated in space by different moieties.
  • other atomic species such as nitrogen and sulfur may also be incorporated into the polyamine species.
  • at least one nitrogen atom of the polyamine has a free electron pair.
  • Preferred as polyamine species are species that range in length from about 3 to about 20 units. More preferably species having at least one nitrogen atom and are of the general formula H 2 N[(CH 2 ) n -(NH) y ] t - wherein n is an integer between 2 and 8 and t is an integer between 1 and 10. These species can be linear or cyclic. Cyclic amines would include crown amines and mixed crown amines/crown ethers.
  • suitable nitrogen-containing compounds suitable for the formation of polyamine species include C 1 -C 20 straight chain alkylamine, C 1 -C 20 straight chain substituted alkylamine, C 2 -C 50 branched chain alkylamine, C 2 -C 50 branched chain substituted.
  • the conjugate groups of the invention may be attached to the 4'-position of the nucleosidic sugar via a linker.
  • Further linking groups include ⁇ -aminoalkoxy moieties and ⁇ -aminoalkylamino moieties for linking a conjugate group to a 4'-hydroxyl group.
  • Many other linking groups are known including many that are commercially available, including heterobifunctional and homobifunctional linking moieties available from the Pierce Co. (Rockford, Il).
  • a further preferred group of linking groups include those groups used as the internucleoside linkages between 4'-desmethyl nucleosides of the parent application, application serial number 039,834, filed March 30, 1993, the entire contents of which are herein expressed incorporated by reference.
  • a linking moiety located on a 4-desmethyl nucleoside compound of the invention may be reacted with an active ester derivative of a conjugation moiety of the invention (e.g., cholic acid).
  • an active ester derivative of a conjugation moiety of the invention e.g., cholic acid
  • Such linking moieties are particularly useful in extending polyalkylamine moieties extending from the oligomeric compounds of the invention.
  • Active ester derivatives are well known to those skilled in the art. Representative active esters include N-hydroxysuccinimide esters, tetrafluorophenolic esters, pentafluorophenolic esters and pentachlorophenolic esters.
  • cholic acid For cholic acid, the reaction of a pendant amino group and the active ester produces an oligomer in which cholic acid is attached to the N-terminal position through a linking group.
  • Cholic acid or other such pendent groups can be attached to the carboxy terminal of the oligomeric compound by conversion to the N-hydroxysuccinimide ester thereof, and then by further reaction with the oligomeric compound of interest.
  • Conjugate molecules of the invention can also be directly connected to the 4'-desmethyl nucleosides compounds of the invention without use of linkers.
  • a conjugate molecule bearing a hydroxyl group, a thiol group, or an amino group is reacted with a 4'-desmethyl nucleoside compound of the invention that has a leaving group at the 4' position.
  • the OH, SH or NH 2 moiety of the conjugate groups displaces the leaving group to directly join the conjugate to the 4'-desmethyl nucleoside compound of the invention.
  • the oligomeric compounds of the invention are synthesized utilizing at least two different methods, both of which can use art recognized solid state synthesis or solution phase synthesis.
  • the "4-desmethyl" nucleoside that is to be functionalized with a conjugate group can be attached to a growing oligomeric compound and then so functionalized.
  • the 4'-desmethyl nucleoside monomer can be first functionalized and then attached to the remainder of the oligomeric compound.
  • a conjugate group of a compound of the invention can be linked to a 4'-desmethyl nucleoside monomer or oligomer through displacement of a leaving group from the 4'-position (as numbered using the sugar ring numbering of a pentofuranosyl nucleoside) of a nucleoside, or the 4'- terminal position (as numbered using the sugar ring numbering of a pentofuranosyl nucleoside) of an oligonucleoside.
  • the 4'-desmethyl compound has structure:
  • leaving groups are functional moieties which are designed to be easily displaced.
  • Representative leaving groups include alkyl and arylsulfonyls including p-toluenesulfonyl (tosyl), 4- dimethylaminoazobenzenesulfonyl (dabsyl), 5-dimethylaminonaphthalenesulfonyl (dansyl), trifluoromethylsulfonyl (triflate), methylsulfonyl (mesyl); halogens; o- trichloroacetimidates; 2,4,6-trichlorophenyl; dialkyl phosphite and acyloxy leaving groups including acetoxy, benzoyloxy, p-methoxybenzoyloxy and p-methylbenzoyloxy and other known leaving groups.
  • Acyloxy leaving groups (-OR E where R E is C(O)-) are preferred, particularly OC(O)CH 3
  • R D can be H, hydroxyl, a nucleoside, an activated nucleotide, a nucleotide, an oligonucleotide or an oligonucleoside or a protected derivative thereof.
  • B x is a naturally occurring or non-naturally occurring heterocyclic base as described supra, and also includes such bases bearing protecting groups.
  • the leaving group can be attached above or below the plane of the sugar ring, i.e., cis or trans to the heretocyclic base.
  • the leaving group may therefore be displaced by either a bimolecular or unimolecular mechanism, dependent upon the specific leaving group and reaction conditions employed.
  • the leaving group is trans to the hetrocyclic base, and is displaced via a bimolecular nucleophilic displacement.
  • R D can be an activated phosphorous group.
  • activated phosphorous groups are groups which are used to facilitate the creation of internucleosidic bonds in nucleic acid synthetic regimes.
  • activated phosphorous groups include phosphate groups having P v such as, for example, activated phosphate esters, and phosphite groups having P III such as phosphoramidites or H-phosphonates.
  • An activated nucleotide according to the invention is a nucleoside which bears an activated phosphorous group.
  • a compound of the above structure is incorporated as the terminal nucleoside unit of a preformed oligomeric structure.
  • oligomeric structures can be any of the various natural or synthetic oligonucleotides or oligonuleotide mimics as known in the art.
  • a nucleoside analog is attached to a solid support in any conventional fashion. It is customary, and preferred, to employ a linker to a solid support such as a polymeric carrier at the 3' position.
  • An oligonucleotide or oligonucleoside is then synthesized as per any of the various known solid state technologies.
  • a final 4'-desmethyl nucleotide is then added to the growing oligomeric compound.
  • the 4'-desmethyl nucleotide is substituted in the 4'-position with leaving group J as described above.
  • the leaving group is then displaced with a linker or directly with a conjugate group of interest. If a linker is utilized the conjugate group is then attached to the linker, either while still on the solid support or post removal from the solid support.
  • the 4'-desmethyl nucleotide is first reacted with a linker or directly with a conjugate group.
  • the modified nucleotide is then attached to the growing oligomeric compound on the solid support.
  • the 4'-desmethyl nucleoside (or nucleotide) is prepared with any base or base analog, B x , and either a pentofuranosyl moiety, where Q is oxygen, a cyclopentane moiety or fluorinated analogue thereof where Q is CH 2 , CHF or CF 2 or a 4'-deoxy-4'-thio moiety, where Q is S.
  • the nucleoside is a ribonucleoside having a 2'-hydroxyl functionality. Such a functionality can be any of various 2'-groups known in the art. If the 2' functionality is OH
  • the 2'-hydroxyl groups can be protected as necessary by means well known to persons of ordinary skill in the art. The only requirement for this group and for its protection is that the same be designed so as to not interfere with substantive reactions in accordance with this invention.
  • the 2'-hydroxyl group will be replaced with other functional groups as for example 2'-alkoxy groups, 2'- alkoxy groups that are substituted with other groups including imidazole and other heterocyclic and amino groups, 2'-halogen, particularly fluoro.
  • Particularly useful 2'-O- substituent groups include 2'-aminopropoxy, 2'- imidazoylbutoxy, 2'-methoxy, 2'-ethoxy, 2'-propoxy, 2'- methoxyethoxy and 2'-deoxy-2'-fluoro.
  • the silyl ether 4 (96.0 g, 136.4 mmol) in THF (600 mL) was treated with hydrogen fluoride-pyridine (70% HF in pyridine, 30 mL) at 0 °C for 4 h under a N 2 atmosphere.
  • the resultant mixture was diluted with AcOEt (600 mL) and washed with H 2 O (2 x 300 mL).
  • the organic layer was dried (MgSO 4 ) and concentrated at reduced pressure.
  • the residue was purified by silica gel column chromatography (CH 2 Cl 2 /AcOEt 10:1) to give 61.6 g (97%) of 5 as a white solid.
  • the resultant mixture was stirred at -23 °C for 15 minutes and then placed in the freezer (-15 °C) for 16 hours.
  • the reaction was quenched by pouring into 1 L of a 4 : 1 mixture of AcOEt/H 2 O containing 20 mL of Et 3 N.
  • the organic layer was washed with H 2 O (300 mL), dried (MgSO 4 ), and concentrated at reduced pressure.
  • the residue was purified by SiO 2 column chromatography (hexanes/AcOEt 1:1) and gave 12.4 g (84.8 %) of 14 as very hydroscopic white foam.
  • Oligonucleotides are prepared utilizing normal protocol for solid state DNA synthesis, (see Oligonucleotide synthesis, a practice approach, Ed. M.J. Gait, IRL Press 1984, Oxford University Press, New York). DMT protected phosphoramidite nucleotides are added to the growing oligonucleotide structure on a solid state support in the normal manner. Coupling efficiencies are typically greater than 96% for each step of the synthesis with the overall coupling efficiency greater than 91% for the oligomer. The resulting oligomers are characterized by both gel chromatography and by HPLC using standard protocols.
  • An oligonucleotide of the desired sequence is prepared as per the preceding Example 17 in the normal 3' to 5' direction.
  • the last nucleotide of the sequence is preformed to include the desired 4' -desmethyl structure bearing a conjugate group, e.g. compounds 12 and 17, attached via linker groups and compound 18, directly attached, thereon as per the above Examples 10, 15 and 17.
  • This last nucleotide unit, as a 3' -phosphoamidite is coupled to the remainder of the oligonucleotide sequence utilizing normal protocol for solid state DNA synthesis, as above. As with the remainder of the sequences, coupling efficiencies are generally greater than 96% for this step of the synthesis.
  • the sequences was synthesizer up to and including the penultimate nucleotide unit.
  • the conjugate group bearing, 4-desmethyl, terminal nucleotide, e.g. compound 12, was then used as the ultimate nucleotide unit.
  • the oligonucleotide was removed from the solid support and purified by HPLC in the standard manner.
  • adhesion molecules are known that are associated with certain adverse events in biological systems. The study of these molecules can be compounded by overlapping activities. Since the intercellular adhesion molecule, ICAM-1, and the vascular adhesion molecule, VCAM- 1, both are capable of expressing certain similar properties, e.g. cell adhesion or clumping, segregation of certain of their properties, one from the other, would assist in the evaluation of the properties of a singular adhesion molecule. This cellular assay isolates cytokine induced protein induction of ICAM-1 from that of VCAM-1.
  • the assay differentiates specific protein inhibition of ICAM-1 expression from VCAM-1 expression via use of a probe oligonucleotide having a sequence complementary to the ICAM- 1 messenger RNA. Inhibition of the induction of ICAM-1 messenger R ⁇ A is effected by treatment of the cells with the probe oligonucleotide in presence of a cationic liposome formulation (DOTMA/DOPE) . Cells showing positive adhesion molecule expression in response to cytokine treatment are identified by increases in basal levels of the adhesion molecule protein after treatment with a combination of human TNF-or and murine I ⁇ F-T using a fluorescence activated cell sorter to identify the cells stained with fluorescent antibodies to either the ICAM-1 or VCAM-1 expressed proteins.
  • DOTMA/DOPE cationic liposome formulation
  • the oligonucleotide utilized as the probe was an oligonucleotide of the invention of the sequence T * GC ATC CCC CAG GCC ACC AT, SEQ ID NO. 1, complementary to the 3' untranslated region of murine ICAM-1 messenger RNA. It includes a conjugation PEG molecule (compound 12 above) as the 5' terminal T nucleotide (nucleotide T * in the above sequence). This oligonucleotide is then used to differentiate ICAM-1 expression from VCAM-1 expression in the test protocol. In cell lines from other species, the base sequence of conjugated oligonucleotide of the invention used in the test is synthesized to be of a sequence complementary to a portion of the ICAM-1 messenger RNA for the species of interest.
  • the cells were treated with nanomolar concentrations of the conjugated oligonucleotide, SEQ ID NO. 1, in the presence of 15 ⁇ g/ml DOTMA/DOPE liposome (Lipofectin) for 4 hours in a serum free media (Opti-MEM serum-free medium, GIBCO, Grand Island, NY) in the manner described by Chiang et al., J. Biol. Chem., 1991, 266, 18162-18171 or Bennett, et al., J.
  • the cells were stained with ICAM-1- PE fluorescent antibodies (anti- ICAM-1 mAB, PharMingen, San Diego, CA) and VCAM-1-FITC fluorescent antibodies (anti- VCAM-1 mAB, PharMingen, San Diego, CA) and read on a fluorescence activated cell sorter.
  • ICAM-1- PE fluorescent antibodies anti- ICAM-1 mAB, PharMingen, San Diego, CA
  • VCAM-1-FITC fluorescent antibodies anti- VCAM-1 mAB, PharMingen, San Diego, CA
  • Concurrent treatment of the same bEND.3 cell line absent the conjugated oligonucleotide served as a positive control. Both ICAM-1 and VCAM-1 induction were observed in the positive control and normalized to 100%.
  • the conjugated oligonucleotide was used at 0.1, 0.3 and 0.5 uM and the levels of both of induced expression ICAM-1 and VCAM-1 were compared to this positive control. Inhibition of VCAM-1 expression was observed to be 90.08%, 103.82% and 96.33% of control for the 0.1, 0.3 and 0.5 uM concentrations, respectively whereas inhibition of ICAM-1 expression was 3.77%, 4.40% and 8.74% of control for the 0.1, 0.3 and 0.5 uM concentrations, respectively.

Abstract

Tetrahydrofuranyl compounds are provided that are functionalized to include pendant conjugate groups, and which are useful in diagnostic assays and as research reagents. Novel intermediates for the synthesis of the compounds are also provided.

Description

OLIGOMERS HAVING HETEROATOM-LINKED CONJUGATE GROUPS
CROSS REFERENCE TO RELATED APPLICATIONS
This application is a continuation-in-part of U.S. Serial No. 314,877, filed September 29, 1994, which is a continuation-in-part of U.S. Serial No. 039,846, filed March 30, 1993, which is a continuation-in-part of U.S. Serial No. 903,160, filed June 24, 1992, and PCT Application No. US 92/04294, filed May 21, 1992, which both are continuations- in-part of U.S. Serial No. 703,619, filed May 21, 1991, which is a continuation-in-part of U.S. Serial No. 566,836, filed August 13, 1990, and of U.S. Serial No. 558,663, filed July 27, 1990. This application is also a continuation-in- part of PCT Application No. US 91/05713, filed August 12, 1991, which is a continuation-in-part of U.S. Serial No. 566,836. Each of the foregoing patent applications are assigned to the assignee of this application and are incorporated by reference herein.
FIELD OF THE INVENTION
This invention is directed to oligomeric compounds that are functionalized to include covalently bound groups.
Specifically, the oligomeric compounds include a tetrahydrofuran moiety that is functionalized with a pendant conjugate group.
BACKGROUND OF THE INVENTION
Oligonucleotides and their analogs have been developed and used in molecular biology in certain procedures as probes, primers, linkers, adapters, and gene fragments. Modifications to oligonucleotides used in these procedures include labeling with non-isotopic labels, e.g. fluorescein, biotin, digoxigenin, alkaline phosphatase, or other reporter molecules. Other modifications have been made to the ribose phosphate backbone to increase the nuclease stability of the resulting analog. These modifications include use of methyl and other alkyl phosphonates, phosphorothioates, phosphorodithioate, phosphoamidate and phosphotriester linkages, and 2'-O-methyl ribose sugar units. Further modifications include modification made to modulate uptake and cellular distribution. Phosphorothioate oligonucleotides are presently being used in human clinical trials for various disease states, including use as antiviral agents. In view of the success of these oligonucleotides for both diagnostic and therapeutic uses, there exists an ongoing demand for improved oligonucleotide analogs.
Oligonucleotides and like molecules can interact with native DNA and RNA in several ways. One of these is duplex formation between an oligonucleotide and a single stranded nucleic acid. A further method is via triplex formation between an oligonucleotide and double stranded DNA to form a triplex structure.
Naturally occurring or synthetic oligonucleotides, together with hybrid species having both synthetic and natural components, can collectively be referenced as "oligomeric compounds." Because of their properties, these oligomeric compounds are known to be useful in a number of different areas. They can be used as probes in cloning, blotting procedures, and in applications such as fluorescence in situ hybridization (FISH). Also, since local triplex formation inhibits gene transcription, such oligomeric compounds can be used to inhibit gene transcription. Labeled oligomers can also be used to directly map DNA molecules, such as by tagging an oligomer with a fluorescent label and effecting hybridization to complementary sequences in duplex DNA. Oligomers can also be used as identification tags in combinatorial chemical libraries as is disclosed in patent publication WO 94/08051 and Ohlmeyer et al., Proc. Natl. Acad. Sci. USA, 1993, 90, 10922-10926.
Considerable research is being directed to the application of oligonucleotides and oligonucleotide analogs that bind complernentary DNA and RNA strands for use as diagnostics, research reagents and potential therapeutics.
For most uses, it is desirable to append to oligomeric compounds groups that modulate or otherwise influence their activity or their membrane or cellular transport. One method of increasing such transport is by the attachment of a pendant lipophilic group. United States application serial number 117,363, filed Sept. 3, 1993, entitled "Amine- Derivatized Nucleosides and Oligonucleosides", describes several alkylamino functionalities and their use in the attachment of such pendant groups to oligonucleosides. Additionally, United States application serial number 07/943,516, filed Sept. 11, 1992, entitled "Novel Amines and Methods of Making and Using the Same" and corresponding to published PCT application WO 94/06815 describe other novel amine-containing compounds, and their incorporation into oligonucleotides for, inter alia, the purposes of enhancing cellular uptake, increasing lipophilicity, causing greater cellular retention and increasing the distribution of the compound within the cell. United States application serial number 08/116,801, filed Sept. 3, 1993, entitled "Thiol- Derivatized Nucleosides and Oligonucleosides", describes nucleosides and oligonucleosides derivatized to include a thioalkyl functionality, through which pendant groups are attached.
Although each of the above-noted patent applications describe useful compounds, there remains a need in the art for additional stable compounds that bind complementary DNA and RNA. There further remains a need in the art for additional methods of attaching pendant groups to oligomeric compounds to further enhance or modulate their binding, cellular uptake, or other activity.
OBJECTS OF THE INVENTION
It is an object of the invention to provide novel nucleosides for use in attaching pendant conjugate groups to oligomeric compounds.
It is a further object of the invention to provide oligomeric compounds that have pendant intercalators, nucleic acid cleaving agents, cell surface phospholipids, diagnostic agents, fluorescent agents and other conjugate groups.
It is yet another object of the invention to provide improvements in research and diagnostic methods and materials for assaying bodily states in animals, especially disease states.
It is a further object of the present invention to provide methods of producing these novel compounds.
These and other objects will become apparent from the following description and accompanying claims.
SUMMARY OF THE INVENTION
The present invention provides compounds useful in diagnostic assays and as research reagents, as well as methods and intermediates for the preparation thereof.
In one aspect of the invention there are provided compounds of structure:
Figure imgf000006_0001
wherein:
RL is azido or a group of formula: RC- [Y]e-Z- in which
Z is O, S or HN;
Y is a bivalent linker;
e is 0 or 1;
and
RC is alkyl, alkenyl, alkynyl, O-alkyl, O-alkenyl, O- alkynyl, aryl, alkaryl, phosphinyl, a polyamine, a polyether, polyglycol, a steroid molecule, a reporter molecule, an aromatic or nonaromatic ring system, an aromatic lipophilic molecule, a non-aromatic lipophilic molecule, a reporter enzyme, a peptide, a protein, a water soluble vitamin, a lipid soluble vitamin, a carbohydrate, a terpene molecule, a phospholipid, an intercalator, a cell receptor binding molecule, a crosslinking agent, or a porphyrin;
Bx is a nucleobase;
X is H, OH, O-alkyl, O-alkoxyalkyl, O-alkylamino or
F;
Q is O, S, CH2, CHF or CF2; and
RD is H, hydroxyl, an activated phosphorous group, a nucleoside, an activated nucleotide, a nucleotide, an oligonucleotide, an oligonucleoside or a protected derivative thereof.
Compounds are also provided according to the invention having structure:
Figure imgf000007_0001
wherein:
L is a group of formula: R7-[S-(CH2)q]u-[HN-(CH2)n]t-[O-(CH2)m]v-Z- in which
Z is O, S or HN;
t, u and v are each independently integers from 0 to
200;
m, n and q are each independently integers from 1 to
4; and
R7 is RC, H or a protecting group; and
X, Bx, RC and RD are defined as above.
In some preferred embodiments t and u are 0, or u and v are zero, or t and v are zero. In other preferred embodiments t and u are 0 and m is 2. In other preferred embodiments t and v are 0 and q is 2 , or u and v are 0 and n is 2. In even other preferred embodiments u is 0 and each of t and v are 1, and m and n are each 2.
Also provided according to the invention are compounds of structure:
Figure imgf000008_0001
wherein:
RC, Bx, X, Q, and RD are defined as above.
In the above structures, in some preferred compounds, RC is a polyether or a polyamine. In other preferred embodiments, RC is a steroid molecule, preferably cholic acid, deoxycholic acid, dehydrocholic acid, cortisone, digoxigenin, testosterone, cholesterol or 3- trimethylaminomethylhydrazido cortisone.
In some preferred embodiments RC is a water soluble vitamin, preferably thiamine, riboflavin, nicotinic acid, pyridoxal phosphate, pyridoxine, pyridoxamine, deoxy- pyridoxine, pantothenic acid, biotin, folic acid, 5'- deoxyadenosylcobalamin, inositol, choline or ascorbic acid.
In other preferred embodiments RC is a lipid soluble vitamin, preferably a retinal, a retinol, retinoic acid, β- carotene, vitamin D, cholecalciferol, a tocopherol, or a phytol.
In some preferred embodiments RC is a protein, preferably a phosphodiesterase, a peroxidase, a phosphatase or a nuclease. In other preferred embodiments RC is a reporter molecule, preferably a chromaphore, a fluorophore or a radiolabel-containing moiety. Preferred fluorophores include fluorescein, chrysine, anthracene and perylene.
In some preferred embodiments, X is H, OH, F, O-alkyl having from one to six carbons, O-alkylamino having from one to six carbons or O-alkoxyalkyl having from one to six carbons. In certain more preferred embodiments X is H or OH mimicking natural deoxyribo and ribo sugar analogs. In other more preferred embodiments X is F, O-alkyl having from one to six carbons, alkylamino having from one to six carbons or O-alkoxyalkyl having from one to six carbons such that the compounds are homologous to certain nucleotide species that have either greater nuclease resistance or high binding affinity.
In other preferred embodiments, RD is H or OH; or RD is an activated phosphorous group, or RD is an oligonucleotide.
The present invention also provides novel synthons useful for the preparation of monomeric and polymeric conjugate compounds. In some embodiments these synthons have the structure:
Figure imgf000010_0001
wherein J is a leaving group and Q, RD, Bx and X are as defined above. Preferably, J is OH, SH, NH2, trifluoromethylsulfonyl, methylsulfonyl, halogen, O- trichloroacetimidate, acyloxy, dialkyl phosphite, 2,4,6- trichlorophenyl, p-toluenesulfonyl, 4-dimethylaminoazo- benzenesulfonyl or 5-dimethylaminonaphthalenesulfonyl .
Also provided are methods for forming a 5'-desmethyl conjugate oligomer having structure:
Figure imgf000010_0002
Preferred methods comprise the steps of:
(a) providing a first synthon having structure
Figure imgf000011_0001
and
(b) contacting the first synthon with a second synthon having structure:
Figure imgf000011_0002
for a time and under reaction conditions sufficient to form the conjugated oligomer; wherein RE is an activated phosphorous group, W is H or a hydroxyl protecting group, k is an integer from 0 to 50, E is a phosphorous linking species including phosphodiester, phosphotriester, phosphorothioate, phosphorodithioate, alkyl phosphonates especially methyl phosphonates and phosphoramidates phosphorous linking groups, and Q, RL, Bx and X are as defined above.
The present invention also provides methods for forming a 5'-desmethyl conjugated monomer having structure:
Figure imgf000012_0001
In certain embodiments these methods comprise the steps of providing a synthon having structure:
Figure imgf000012_0002
and contacting this synthon with an activated conjugating group. This contacting should be for a time and under reaction conditions sufficient to form the conjugated oligomer. Preferably, J is OH, SH, NH2, trifluoromethylsulfonyl, methylsulfonyl, halogen, O- trichloroacetimidate, acyloxy, dialkyl phosphite, 2,4,6- trichlorophenyl, p-toluenesulfonyl, 4-dimethylaminoazo- benzenesulfonyl or 5-dimethylaminonaphthalenesulfonyl; more preferably trifluoromethylsulfonyl, methylsulfonyl, halogen or acyloxy.
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
The present invention provides novel nucleosides for incorporation into nucleic acids. In one aspect, the invention provides nucleosides bearing pendant conjugate groups. The term "nucleoside" refers to a unit composed of a heterocyclic base and a sugar, generally a pentose sugar. In naturally occurring nucleosides, the heterocyclic base typically is guanine, adenine, cytosine, thymine or uracil, and the sugar is normally deoxyribose, i.e., erythro-pento- furanosyl, or ribose, i.e., ribopentofuranosyl. Synthetic sugars also are known, including arabino, xylo or lyxo pentofuranosyl sugars and hexose sugars. Further synthetic sugar are the 4'-deoxy-4'-thio sugars (both 2'-deoxy and ribo varieties) that have been described by Secrist et al., J. Med. Chem. 1991, 34, 2361-2366 and patent application PCT/FR93/00115, respectively, (whereby Q in the above structures is S). Throughout this specification, reference to the sugar portion of a nucleoside or other nucleic acid species shall be understood to refer to naturally occurring deoxyribose and ribose sugars and also to sugar species which can replace the sugar moieties of naturally occurring nucleic acids.
Reference to the nucleobase or heterocyclic base portion of a nucleoside or other nucleic acid species shall be understood to refer to naturally-occurring nucleobases, modified derivatives thereof, or to synthetic nucleobases. Synthetic nucleobases are those which can replace one or more of the naturally occurring base moieties of wild type nucleic acids.
Reference to inter-sugar linkages shall be taken to include moieties that join the sugar portions of nucleosides or nucleotides.
The term "nucleotide" refers to a nucleoside having a phosphate group esterified to one of its 2', 3' or 5' sugar hydroxyl groups. The phosphate group normally is a monophosphate, diphosphate or triphosphate.
The term "oligonucleotide" refers to a plurality of monophosphate nucleotide units that are typically formed from naturally occurring bases and pentofuranosyl sugars joined by native phosphodiester bonds in a specific sequence. A homo-oligonucleotide is formed from nucleotide units having the same heterocyclic base, e.g. polyadenosine. The term "oligonucleotide analog" has been used in various published patent application specifications and other literature to refer to molecular species that are similar to oligonucleotides, but that have non-naturally occurring portions. This term has been used to identify oligonucleotide- like molecules that have altered sugar moieties, altered base moieties or altered inter-sugar linkages. Thus, the term oligonucleotide analog has been used to denote structures having altered inter-sugar linkages including phosphorothioate, phosphorodithioate, methyl phosphonate, phosphotriester or phosphoramidate inter-nucleoside linkages in place of phosphodiester inter- nucleoside linkages; purine and pyrimidine heterocyclic bases other than guanine, adenine, cytosine, thymine or uracil and sugars having other than the β pentofβranosyl configuration or sugars having substituent groups at their 2' position or substitutions for one or more of the hydrogen atoms. The term "modified oligonucleotide" also has been used in the literature to denote such structures.
The term "oligonucleotide mimic" has also been used to refer to macromolecular moieties that function similarly to or "mimic" the function of oligonucleotides but have non- naturally occurring inter-sugar linkages. Oligonucleotide mimics thus can have natural or altered or non-naturally occurring sugar moieties and natural or altered or non- naturally occurring base moieties in combination with non- naturally occurring dephospho linkages. Certain dephospho linkages have been reviewed by Uhlmann, E. and Peyman, A., "Oligonucleotide Analogs Containing Dephospho Internucleo- side Linkages" in Methods in Molecular Biology, Chapter 16, Oligonucleotide Synthetic Protocols, S. Agrawal, Ed., The Humana Press, Inc., Totowa, NJ, 1993.
The term "oligomers" is intended to encompass oligonucleotides, oligonucleotide analogs, oligonucleosides or oligonucleotide-mimicking macromolecules. Thus, "oligomers" refers to nucleosides or nucleoside analogs that are joined together via either natural phosphodiester bonds or via other linkages.
In certain embodiments the nucleoside compounds of the invention lack the 5'-methylene group present in conventional pentofuranosyl nucleosides. In certain preferred embodiments a heteroatom occupies the position normally occupied, by the missing 5'-methylene group. In one sense, these compounds can be considered as 4'-desmethyl pentofuranosyl nucleosides. However, in accordance with IUPAC rules, the lack of a 5'-methylene carbon on the "sugar portion" of the nucleosides of the invention require their designation as tetrahydrofuranyl moieties. Thus, in naming these compounds according to IUPAC rules, as for example in the identification of the structural positions of the compound, established hierarchical or priority nomenclature rules are followed. Accordingly, in embodiments wherein a heteroatom has replaced the 5'-methylene group, that heteroatom and the pendant conjugate group attached thereto takes priority over the heterocyclic base of the nucleoside. In such structures, the tetrahydrofuranyl ring is numbered counterclockwise and the position occupied by the heteroatom (in what would be the 5' position of a conventional nucleoside) is identified as the 2-position. However, in identifying certain of the protons in the NMR spectra, conventional pentofuranosyl nucleoside numbering has been used (except where otherwise noted) for the tetrahydrofuranyl nucleosides.
For the purpose of this specification and the claims appended hereto, when an oligomeric structure of the invention is being considered, the ends of this structure are referenced in the same manner as for conventional oligonucleotides. Thus, they are identified either as a 3' end or a 5' end. In other instances where analogy to conventional pentofuranosyl nucleosides is made, strict IUPAC naming rules are deviated from and the numbering system of the conventional pentofuranosyl nucleosides is maintained. In these instances it is convenient to consider certain tetrahydrofuranyl compounds more as 4'-desmethyl pentofuranosyl compounds and thus attachment is noted as being at the 4' position.
Compounds of the invention include 4'-desmethyl nucleoside monomers modified to bear conjugate groups on their 4'-carbons (i.e., 4'-desmethyl conjugate monomers) or oligomers which contain such modified nucleosides (4'- desmethyl conjugate oligomers). Some oligomeric embodiments have the structure:
Figure imgf000016_0001
wherein a pendant conjugate group RL is attached to the 4'- position of a nucleoside of the invention. In this structure the "E" group can be any of the commonly use oligonucleotide phosphorous linkage including phosphodiester, phosphotriester, phosphorothioate, phosphorodithioate, alkyl phosphonate and phosphoamidate linkages. The pendant conjugate group can be a group of formula: RC-[S-(CH2)q]u-[HN-(CH2)n]t-[O-(CH2)m]v-Z- wherein Z is O, NH or S, preferably O or NH, t, u and v are each independently integers from 0 to 200; m, n and q are each independently integers from 1 to 4; and RC is alkyl, alkenyl, alkynyl, aryl, alkaryl, phosphinyl, O-alkyl, O- alkenyl, O-alkynyl, a polyglycol, a polyamine, a polyether, a steroid molecule, an aromatic or nonaromatic ring system, a reporter molecule, an aromatic lipophilic molecule, a nonaromatic lipophilic molecule, a reporter enzyme, a peptide, a protein, a water soluble vitamin, a lipid soluble vitamin, a carbohydrate, a terpene molecule, a phospholipid, an intercalator, a. cell receptor binding molecule, a crosslinking agent, an RNA cleaving complex, a metal chelator, an alkylator or a porphyrin.
In oligomers of the invention at least one 4'- desmethyl nucleoside unit incorporated therein has been derivatized to bear a pendant conjugate group as described above. When so derivatized, the oligomeric compound is useful, for example, in a diagnostic or other preparation that includes a nucleic acid binding agent to assist in identification of the oligomeric compound, to aid in transfer of the compound across cellular membranes, or to impart other properties. Such a diagnostic or nucleic acid binding agent is formed from an oligomeric compound of the invention wherein the oligomeric compound includes monomeric units bearing natural or non-natural occurring bases in a sequence that is complementary to and will specifically hybridize with a region of an RΝA or DΝA of interest.
For the purpose of identification, a functionalized oligomeric compound according to the invention can be characterized as a substituent-bearing (e.g., steroid- bearing) oligomeric compound. Such oligomeric compounds will have at least one pendant group attached thereto to modulate their activity.
The compounds of the inventions have various uses including being useful as research reagents and in diagnostic assays. In one particular diagnostic assay, compounds of the inventions are used to isolate the effects of one cellular adhesion molecule from those of a further cellular adhesion molecule in a protein expression assay. In this assay, the effect of induced expression of intercellular adhesion molecule-1 (ICAM-1) were diminished while those of VCAM-1 were maintained. This allows for analysis of the VCAM-1 protein expression without concurrent expression and interference of ICAM-1 protein.
In general, the oligomeric compounds bearing pendant conjugate groups of the present invention can be used to bind to various other target molecules. Target molecules of the present invention can include any of a variety of biologically significant molecules. Other such target molecules can be nucleic acids, carbohydrates, glycoproteins or other proteins.
In binding to nucleic acids, the functionalized oligomeric compounds of the invention bind with complementary strands of RNA or DNA. After binding, the oligomeric compound and the RNA or DNA strand can be considered to be complementary strands which are "duplexed" in a manner analogous to native, double-stranded DNA. In such complementary strands, the individual strands are positioned in such a manner with respect to one another so as to allow Watson-Crick type, Hoogsteen type or another type hybridization of the heterocyclic bases of one strand to the heterocyclic bases of the opposing strand.
Binding to nucleic acids can be practiced against nucleic acids from a variety of sources including organisms ranging from unicellular prokaryotes and eukaryotes to multicellular eukaryotes. The nucleic acid from any organism that utilizes DNA-RNA transcription or RNA-protein translation as a fundamental part of its hereditary, metabolic or cellular control is susceptible to such binding. Seemingly diverse organisms such as bacteria, yeast, virus, protozoa, algae, all plant and all higher animal forms, including warm-blooded animals, are sources of such nucleic acid. Further, since each of the cells of multicellular eukaryotes includes both DNA-RNA transcription and RNA-protein translation as an integral part of their cellular activity, nucleic acid binding for various purposes, e.g. diagnostics, can also be practiced on such cellular populations. Furthermore, many of the organelles, e.g., mitochondria and chloroplasts, of eukaryotic cells include transcription and translation mechanisms. Thus, single cells, cellular populations or organelles can also be included within the definition of organisms that are capable of being treated with nucleic acid binders. In some preferred embodiments of the present invention, the target molecule is a protein such as an immunoglobulin, receptor, receptor binding ligand, antigen or enzyme and more specifically can be a phospholipase, tumor necrosis factor, endotoxin, interleukin, plasminogen activator, protein kinase, cell adhesion molecule, lipoxygenase, hydrolase or transacylase. In other embodiments of the invention the target molecules can be important regions of the human immunodeficiency virus, Candida, herpes viruses, papillomaviruses, cytomegalovirus, rhinoviruses, hepatitis viruses, or influenza viruses. In further embodiments, the target molecules can be regions of an oncogene. In still further embodiments, the target molecule is ras 47-mer stem loop RNA, the TAR element of human immunodeficiency virus or the gag-pol stem loop of human immunodeficiency virus (HIV). Still other targets can induce cellular activity. For example, a target can induce interferon.
Binding also can be practiced against transcription factors. In binding to transcription factors or other target molecules, the transcription factor or other target molecule need not be purified. It can be present, for example, in a whole cell, in a humoral fluid, in a crude cell lysate, in serum or in other humoral or cellular extracts. Of course, purified transcription factor or a purified form of another target molecule is also useful in some aspects of the invention.
In still other embodiments of the present invention, synthetically prepared transcription factors or other target molecules can be useful. A transcription factor or other target molecule also can be modified, such as by biotinylation or radiolabeling. For example, a synthetically prepared transcription factor can incorporate one or more biotin molecules during synthesis or can be modified post- synthesis.
Transcription factors, as the term is used herein, are DNA- or RNA-binding proteins that regulate the expression of genes. HIV tat and c-rel are examples of transcription factors which regulate the expression of genes. Also encompassed by the term are DNA and RNA binding proteins which are not strictly considered transcription factors, but which are known to be involved in cell proliferation. These transcription factors include c-myc, fos, and jun. Methods of the present invention are particularly suitable for use with transcription factors as target molecules since transcription factors generally occur in very small cellular quantities.
In still other embodiments of the invention, nucleic acid binding can be used to treat objects (glasswares, petri dishes, instruments or the like) to sterilize, sanitize or disinfect such objects that may harbor an organism, e.g., bacterial, protozoan, viral, fungal or other infectious or non- infectious agent on the object. In binding to the nucleic acid of the agent, the compounds of the invention will kill, abrogate, inhibit, curtail or other wise control, eradicate or render harmless the cellular growth or gene expression of such organisms on the object. Such nucleic acid binding can also be used to render such organisms incapable or otherwise unable to reproduce themselves.
In still further embodiments of the inventions, the nucleic acid binding properties of the compounds of the invention can be used to form duplex structures with an unknown oligomer species for identification of that species by gel analysis including slab and capillary gel electrophoresis. In still further embodiments of the inventions, the compounds of the invention can be used as tags in identification of other compounds in combinatorial libraries.
Preferred nucleobase units for oligomeric compounds of the invention include naturally occurring or synthetic purine or pyrimidine heterocyclic bases, including but not limited to adenine, guanine, cytosine, thymine, uracil, 5- methylcytosine, hypoxanthine or 2-aminoadenine. Other such heterocyclic bases include 2-methylpurine, 2,6-diaminopurine, 6-mercaptopurine, 2,6-dimercaptopurine, 2-amino-6- mercaptopurine, 5-methylcytosine, 4-amino-2-mercaptopyrimidine, 2,4-dimercaptopyrimidine and 5-fluorocytosine. Representative heterocyclic bases are disclosed in U.S. Patent No. 3,687,808 (Merigan, et al.), which is incorporated herein by reference.
In preferred embodiments, the oligomeric compounds of the invention include from about 2 to about 50 nucleoside subunits (i.e., k = about 1 to about 49).
As used in this specification, alkyl groups of the invention include but are not limited to C1-C12 straight and branched chained alkyls such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl, isopropyl, 2 -butyl, isobutyl, 2-methylbutyl, isopentyl, 2-methylpentyl, 3-methylpentyl, 2-ethylhexyl and 2-propylpentyl. Alkenyl groups include but are not limited to unsaturated moieties derived from the above alkyl groups including but not limited to vinyl, allyl and crotyl. Alkynyl groups include unsaturated moieties having at least one triple bond that are derived from the above alkyl groups including but are not limited to ethynyl and propargyl.
Alkanoyl groups according to the invention are alkyl, alkenyl or alkynyl groups attached through a carbonyl group.
The term aryl is intended to denote monocyclic and polycyclic aromatic groups including, for example, phenyl, naphthyl, xylyl, pyrrole, and furyl groups. Although aryl groups (e.g., imidazo groups) can include as few as 3 carbon atoms, preferred aryl groups have 6 to about 14 carbon atoms, more preferably 6 to about 10 carbon atoms. Aralkyl and alkaryl groups according to the invention each include alkyl and aryl portions. Aralkyl groups are attached through their alkyl portions, and alkaryl groups are attached through their aryl portions. Benzyl groups provide one example of an aralkyl group, and p-tolyl provides an example of an alkaryl group.
Alkylamino and aminoalkyl groups according to the invention each include amino and alkyl portions. Alkylamino groups are attached through their amino portions, and aminoalkyl groups are attached through their alkyl portions. Methylamino groups provide one example of an alkylamino group, a β-aminobutyl group is one example of an aminoalkyl group.
For the purposes of this invention a ring system is defined to include two or more single rings that join together to form an extended or condensed ring. Such ring systems include extended aromatic systems, alicyclic systems, araalicyclic systems, and bicyclic systems. Examples include aromatic, alicyclic and mixed aromatic- alicyclic (araalicyclic) multiple ring systems, bicyclic systems, non-aromatic multiple ring systems such as adamantane, decalin, and norbornane. Ring systems also include, but are not limited to, naphthalene, tetrahydronaphthalene (tetralin), anthracene, phenanthrene, fluorene, pyrene, coronene, azulene, cluorene, benzonaphthene, benzo [8] annulene, pentalene, heptalane, octalene, indene, isoindene biphenyl, biphenylene and triphenylene condensed rings.
The terms alkyl, alkaryl, aralkyl and aryl are intended to denote both substituted (e.g., halogenated and hydroxylated) and unsubstituted moieties.
Halogens include fluorine, chlorine and bromine. Suitable heterocyclic groups include but are not limited to imidazole, tetrazoie, triazole, pyrrolidine, piperidine, piperazine and morpholine. Heterocycloalkyl groups are cyclic alkyl groups containing a heretoatom. Heterocycloalkaryl groups are aryl heterocycles bearing at least one alkyl substituent.
Amines include amines of all of the above alkyl, alkenyl and aryl groups including primary and secondary amines and "masked amines" such as phthalimide. Amines are also meant to include polyalkylamino compounds and aminoalkylamines such as aminopropylamine and further heterocycloalkylamines such as imidazol-1, 2 or 4 -yl-propylamine.
Substituent groups for the above as well as for other moieties listed below include but are not limited to other alkyl, haloalkyl, alkenyl, alkynyl, alkoxy, thioalkoxy, haloalkoxy and aryl groups as well as halogen, hydroxyl, amino, azido, carboxy, cyano, nitro, mercapto, sulfides, sulfones, sulfoxides, keto, carboxy, nitrates, nitrites, nitroso, nitrile, trifluoromethyl, O-alkyl, S-alkyl, NH- alkyl, amino, silyl, amides, ester, ethers, carbonates, carbamates, ureas, imidazoles, intercalators, conjugates, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligonucleotides, and groups that enhance the pharmacokinetic properties of oligonucleotides. Other suitable substituent groups also include rhodamines, coumarins, acridones, pyrenes, stilbenes, oxazolopyridocarbazoles, anthraquinones, phenanthridines, phenazines, azidobenzenes, psoralens, porphyrins and cholesterols. One particularly preferred group is CF3.
As used in the present invention, the term polyether means a linear or branched alkyl chain periodically interrupted by oxygen atoms. One preferred example of a polyethers is polyethylene glycol. The term polyglycol, as used herein, includes compounds such as glycerol, and derivative groups such as 2,3-dialkoxypropyl groups. The term polyamine as used herein includes the nitrogen analogs of such structures, and the term polythioether includes the sulfur analogs of such structures.
Carbohydrates according to the invention are inclusive of pentose, hexose and higher sugars, and the polymeric species thereof. Representative carbohydrates include glucose and galactose and their derivatives including, as for example, glycals, glycal epoxides and glycosides.
Terpenes are known in the art as oligomers of isoprene, particularly the dipentenes, pinenes, and myrcenes. Included within the definition of terpene molecules are terpene derivatives such as camphor and menthol.
The term phospholipid as used herein includes those compounds which upon hydrolysis yield phosphoric acid, an alcohol and one or more fatty acids. Representative examples of phospholipids include lecithin, cephalin and sphingomyelin.
As used in the present invention, groups that enhance the pharmacodynamic properties include groups that improve oligonucleotide uptake, enhance oligonucleotide resistance to degradation, and/or strengthen sequence-specific hybridization with RNA. Groups that enhance the pharmaco- kinetic properties, in the context of this invention, include groups that improve oligonucleotide uptake, distribution, metabolism or excretion.
For the purposes of this invention, the terms "reporter molecule" and "reporter enzyme" are inclusive of those molecules or enzymes that have physical or chemical properties that allow them to be identified in gels, fluids, whole cellular systems, broken cellular systems and the like utilizing physical properties such as spectroscopy, radioactivity, colorimetric assays, fluorescence, and specific binding. Particularly useful as reporter molecules are fluorophores, chromaphores and radiolabel-containing moieties. Fluorophores are molecules detectable by fluorescence spectroscopy. Examples of preferred fluorophores are fluorescein and rhodamine dyes and acridines. There are numerous commercial available fluorophores including "Texas Red" and other like fluoresceins and rhodamines available for Molecular Probes, Eugene, OR. Chromaphores are molecules capable of detection by visible or ultraviolet (UV-VIS) absorbance spectroscopy. Examples of chromaphores are polynuclear aromatics such as anthracene, perylene, pyrene, rhodamine and chrysene. Radiolabel-containing moieties, as used herein, are molecules incorporating at least one radioactive atom, such as 3H or 14C, enabling detection thereby. Reporter enzymes may be detected directly or via their enzymatic products by any of the methods mentioned above. Particularly useful as reporter enzymes are alkaline phosphatase and horseradish peroxidase.
Steroid molecules according to the invention include those chemical compounds that contain a perhydro-1, 2- cyclopentanophenanthrene ring system. Particularly useful as steroid molecules are the bile acids including cholic acid, deoxycholic acid and dehydrocholic acid; steroids including cortisone, digoxigenin, testosterone and cholesterol and cationic steroids such as cortisone having a trimethylaminomethyl hydrazide group attached via a double bond at the 3 -position of the cortisone ring (3- trimethylaminomethylhydrazido cortisone).
Proteins and peptides are utilized in their usual sense as polymers of amino acids. Normally peptides comprise such polymers that contain a smaller number of amino acids per unit molecule than do the proteins. Particularly useful as peptides and proteins are sequence- specific peptides and proteins including phosphodiesterases, peroxidases, phosphatases and nucleases. Such peptides and proteins include, but are not limited to, SV40 peptide, RNase A, RNase H and Staphylococcal nuclease.
Lipophilic molecules include naturally-occurring and synthetic aromatic and non-aromatic moieties such as fatty acids, esters and alcohols, other lipid molecules, cage structures such as adamantane and buckminsterfullerenes, and aromatic hydrocarbons such as benzene, perylene, phenanthrene, anthracene, naphthalene, pyrene, chrysene, and naphthacene. Particularly useful as lipophilic molecules are alicyclic hydrocarbons, saturated and unsaturated fatty acids, waxes, terpenes and polyalicyclic hydrocarbons including adamantane and buckminsterfullerenes.
Alkylators according to the invention are moieties that can effect attachment of electrophilic groups to targeted molecular structures. Representative alkylators are disclosed by Meyer, et al., J. Am. Chem. Soc. 1989, 111, 8517.
Intercalators are polycyclic aromatic moieties that can insert between adjacent base pairs without affecting normal Watson-Crick base pairing, and include hybrid intercalator/ligands such as the photonuclease/intercalator ligand 6-[[[9-[[6-(4-nitrobenzamido)hexyl]amino] acridin-4- yl]carbonyl]amino] hexanoylpentafluorophenyl ester. This compound has two noteworthy features: an acridine moiety that is an intercalator and a p-nitrobenzamido group that is a photonuclease. Other representative intercalators are disclosed by Manoharan, M., Antisense Research and Applications, Crooke and Lebleu, eds., CRC Press, Boca Raton, 1993.
Cell receptor binding molecules according to the invention are vitamins and carbohydrate moieties for which specific receptors exist within a cell. Representative cell receptor binding molecules are disclosed by Application Serial No. PCT/US92/09196, filed October 23, 1992, the contents of which are incorporated herein by reference.
Crosslinking agents are moieties that can effect intrastrand or interstrand covalent binding of RNA and/or DNA, and include photo-crosslinking agents. Examples of photocrosslinking agents include aryl azides such as N- hydroxysuccinimidyl-4-azidobenzoate (HSAB) and N- succinimidyl-6(-4'-azido-2'-nitrophenylamino) hexanoate
(SANPAH). Aryl azides conjugated to oligomers will effect crosslinking with nucleic acids and proteins upon irradiation. Other representative crosslinking agents are disclosed in International Patent Application Serial No. PCT/US93/02059, filed March 5, 1993, which is incorporated herein by reference.
Useful crown amines are disclosed by Studer, et al., Helv. Chim. Acta 1986, 69, 2081 and Smith-Jones, et al., Bioconjugate Chem. 1991, 2, 415.
Vitamins according to the invention generally can be classified as water soluble or lipid soluble. Water soluble vitamins include thiamine, riboflavin, nicotinic acid or niacin, the vitamin B6 pyridoxal group,, pantothenic acid, biotin, folic acid, the B12 cobalamin coenzymes, inositol, choline and ascorbic acid. Lipid soluble vitamins include the vitamin A family, vitamin D, the vitamin E tocopherol family and vitamin K (and phytols). The vitamin A family, including retinoic acid and retinol, are absorbed and transported to target tissues through their interaction with specific proteins such as cytosol retinol-binding protein type II (CRBP-II), Retinol-binding protein (RBP), and cellular retinol-binding protein (CRBP). These proteins, which have been found in various parts of the human body, have molecular weights of approximately 15 kD. They have specific interactions with compounds of the vitamin A family, especially, retinoic acid and retinol.
The vitamin A family of compounds can be attached to oligomers of the invention via acid or alcohol functionalities found in the various family members. For example, conjugation of an N-hydroxysuccinimide ester of an acid moiety of retinoic acid to an amine function of a pendant group of the oligomer results in linkage of the vitamin A compound to the oligomer, via an amide bond. In similar fashion, standard esterification chemistries may be used to attach the acid moiety of the retinoic acid group to a 4'-oxygen of a compound of the invention, or to a hydroxyl function of a pendent group thereof.
α-Tocopherol (vitamin E) and other tocopherols (beta through zeta) can be similarly conjugated to oligomers also to enhance uptake due to their lipophilic character. The lipophilic vitamin, vitamin D, and its ergosterol precursors can be conjugated to oligomers through their hydroxyl groups by first activating the hydroxyl groups by forming, for example, hemisuccinate esters. Conjugation then is effected, as for instance, to an aminolinker pendant from the oligomer, or through other suitable functional groups described herein. Other vitamins that can be conjugated to oligomers through hydroxyl groups on the vitamins include thiamine, riboflavin, pyridoxine, pyridoxamine, pyridoxal, deoxypyridoxine. Lipid soluble vitamin K's and related quinone-containing compounds can be conjugated via carbonyl groups on the quinone ring. The phytol moiety of vitamin K may also serve to enhance binding of the oligomers to cells.
Pyridoxal phosphate is the coenzyme form of Vitamin B6. Vitamin B6 is also known as pyridoxine. Pyridoxal has specific B6-binding proteins. The role of these proteins in pyridoxal transport has been studied by Zhang and McCormick, Proc. Natl. Acad. Sci. USA, 1991 88, 10407. Zhang and McCormick showed that a series of N-(4'-pyridoxyl) amines, in which several synthetic amines were conjugated at the 4'- position of pyridoxal, were able to enter cells by a process facilitated by the B6 transporter. Zhang and McCormick also demonstrated the release of these synthetic amines within the cell. Other pyridoxal family members include pyridoxine, pyridoxamine, pyridoxal phosphate, and pyridoxic acid. Pyridoxic acid, niacin, pantothenic acid, biotin, folic acid and ascorbic acid can be conjugated to oligomers using N-hydroxysuccinimide esters as described above for retinoic acid.
Other groups for modifying properties of oligomers include RNA cleaving complexes, pyrenes, metal chelators, porphyrins, alkylators, hybrid intercalator/ligands and photo-crosslinking agents. RNA cleavers include o- phenanthroline/Cu complexes and Ru (bipyridine)3 2+ complexes. The Ru (bipyridine)3 2+ complexes interact with nucleic acids and cleave nucleic acids photochemically. Metal chelators include EDTA, DTPA, and o-phenanthroline. Alkylators include compounds such as iodoacetamide. Porphyrins include porphine, its substituted forms, and metal complexes. Pyrenes include pyrene and other pyrene-based carboxylic acids that can be conjugated using protocols similar to those specified above.
Numerous suitable protecting groups are known in the art for protecting the several functional groups of the compounds of the invention during synthesis. Protecting groups are known per se as chemical functional groups that can be selectively appended to and removed from functionalities, such as hydroxyl groups and carboxyl groups. These groups are present in a chemical compound to render such functionality inert to chemical reaction conditions to which the compound is exposed. Protecting groups useful in the context of the present invention include but are not limited to hydroxyl protecting groups such as t-butyldiphenylsilyl, t-butyldimethylsilyl, and dimethoxytrityl groups, thiol protecting groups such as S- trityl, S-p-methoxybenzylthioether, S-p-nitrobenzyl- thioether, and S-t-butylthioether. (see, e.g., Veber and Hirschmann, et al., J. Org. Chem. 1977, 42, 3286 and Atherton, et al., The Peptides, Gross and Meienhofer, Eds, Academic Press; New York, 1983; Vol. 9 pp. 1- 38). Other representative protecting groups suitable for practice in the invention may be found in Greene, T.W. and Wuts, P.G.M., "Protective Groups in Organic Synthesis" 2d. Ed., Wiley & Sons, 1991.
In one aspect, the present invention is directed to nucleoside monomers that bear at least one conjugate group at the 4'-position. In oligomeric embodiments, the conjugate-bearing nucleoside is the 5'-terminal residue oligomer. In some preferred embodiments the conjugate group is a polyamine, and in other preferred embodiments the conjugate group is a polyether. In further preferred embodiments the conjugate group is a polythioether.
The conjugate groups of the preceding paragraph can further serve as bivalent linkers for other conjugate groups. Thus for instances, a hydroxyl, amino or thiol terminated polyether, polyamine or polythioether is used a linker and the terminal group is then reacted with a suitable functional group on a conjugate molecules to link the conjugate molecule to the polyether, polyamine or polythioether. Suitable functional groups for reacting with hydroxyl, amine or thiol groups include a gamut of groups known in the organic chemical arts including, but not limited to, acid chlorides, anhydrides, cyclic anhydrides, alkyl halides, organometallics, chloroformates, isocynates and the like. In a like manner other terminal groups can be utilized on the linker including, but not limited to, hydrazines, aldehydes, acids, hydroxylamines, semicarbazides, thiosemicarbazides, hydrazones, hydrazides and the like. Protected forms of such compounds can also be used, as for example, protected aldehydes including, C- formyl, o-methylaminobenzenethio, aryl substituted imidazolidino moieties and the like. In a like manner, protected thiol include trityl thiol that can be deblocked allowing a disulfide bonds to be formed between the linker and the a sulfur bearing conjugate group to join the conjugate group to the linker. Various nitrogen linkages can be used to join the conjugate groups including hydrazones, oximes, hydrazide-hydrazone, semicarbazone and semithiocarbazones.
The phrase polyamine species, including polyamines, as used herein refers to species that have a plurality of nitrogen atoms thereon. Polyamines include primary amines, hydrazines, semicarbazines, thiosemicarbazines and similar nitrogenous species. Such species can be symmetrical species such as polyamine containing polymers or they can be unsymmetrical wherein the amine functionalities of the polyamine are separated in space by different moieties. In addition to carbon atoms other atomic species such as nitrogen and sulfur may also be incorporated into the polyamine species. In some preferred embodiments of the invention, at least one nitrogen atom of the polyamine has a free electron pair.
Preferred as polyamine species are species that range in length from about 3 to about 20 units. More preferably species having at least one nitrogen atom and are of the general formula H2N[(CH2)n-(NH)y]t- wherein n is an integer between 2 and 8 and t is an integer between 1 and 10. These species can be linear or cyclic. Cyclic amines would include crown amines and mixed crown amines/crown ethers.
Other suitable nitrogen-containing compounds suitable for the formation of polyamine species include C1-C20 straight chain alkylamine, C1-C20 straight chain substituted alkylamine, C2-C50 branched chain alkylamine, C2-C50 branched chain substituted. alkylamine, C3-C50 cyclic alkylamine, C3-C50 cyclic substituted alkylamine, C2-C20 straight chain alkenylamine, C2-C20 straight chain substituted alkenylamine, C3-C50 branched chain alkenylamine, C3-C50 branched chain substituted alkenylamine, C3-C50 cyclic alkenylamine, C3-C50 cyclic substituted alkenylamine, C2-C20 straight chain alkynylamine, C2-C20 straight chain substituted alkynylamine, C3-C50 branched chain alkynylamine, C3-C50 branched chain substituted alkynylamine, C3-C50 cyclic alkynylamine, C3-C50 cyclic substituted alkynylamine, C1-C20 straight chain alkylhydrazine, C1-C50 straight chain substituted alkylhydrazine, C2-C50 branched chain alkylhydrazine, C2-C50 branched chain substituted alkylhydrazine, C3-C50 cyclic hydrazoalkane, C3-C50 cyclic substituted hydrazoalkane, C2-C20 straight chain alkenylhydrazine, C2-C20 straight chain substituted alkenylhydrazine, C3-C50 branched chain alkenylhydrazine, C3-C50 branched chain substituted alkenylhydrazine, C3-C50 cyclic hydrazoalkene, C3-C50 cyclic substituted hydrazoalkene, C2-C20 straight chain alkynylhydrazine, C2-C20 straight chain substituted alkynylhydrazine, C3-C50 branched chain alkynylhydrazine, C3-C50 branched chain substituted alkynylhydrazine, C2-C30 cyclic hydrazoalkyne, C3-C50 cyclic substituted hydrazoalkyne, C1-C20 straight chain alkylhydroxyamine, C1-C20 straight chain substituted alkylhydroxyamine, C2-C50 branched chain alkylhydroxyamine, C2-C50 branched chain substituted alkylhydroxyamine, C3-C50 cyclic oxyalkylamine, C3-C50 cyclic substituted oxyalkylamine, C2-C20 straight chain alkenylhydroxyamine, C2-C20 straight chain substituted alkenylhydroxyamine, C3-C50 branched chain alkenylhydroxyamine, C3-C50 branched chain substituted alkenylhydroxyamine, C3-C50 cyclic oxyalkenylamine, C3-C50 cyclic substituted oxyalkenylamine, C2-C20 straight chain alkynylhydroxyamine, C2-C20 straight chain substituted alkynylhydroxyamine, C3-C50 branched chain alkynylhydroxyamine, C3-C50 branched chain substituted alkynylhydroxyamine, C3-C50 cyclic oxyalkynylamine, C3-C50 cyclic substituted oxyalkynylamine, C1-C20 straight chain alkylsemicarbazide, C1-C20 straight chain substituted alkylsemicarbazide, C2-C50 branched chain alkylsemicarbazide, C2-C50 branched chain substituted alkylsemicarbazide, C3-C50 cyclic alkylsemicarbazide, C3-C50 cyclic substituted alkylsemicarbazide, C2-C20 straight chain alkenylsemicarbazide, C2-C20 straight chain substituted alkenylsemicarbazide, C3-C50 branched chain alkenylsemicarbazide, C3-C50 branched chain substituted alkenylsemicarbazide, C3-C50 cyclic alkenylsemicarbazide, C3- C50 cyclic substituted alkenylsemicarbazide, C2-C20 straight chain alkynylsemicarbazide, C2-C20 straight chain substituted alkynylsemicarbazide, C3-C50 branched chain alkynylsemicarbazide, C3-C50 branched chain substituted alkynylsemicarbazide, C3-C50 cyclic alkynylsemicarbazide, C3- C50 cyclic substituted alkynylsemicarbazide, C1-C20 straight chain alkylthiosemicarbazide, C1-C20 straight chain substituted alkylthiosemicarbazide, C2-C50 branched chain alkylthiosemicarbazide, C2-C50 branched chain substituted alkylthiosemicarbazide, C3-C50 cyclic alkylthiosemicarbazide, C3-C50 cyclic substituted alkylthiosemicarbazide, C2-C20 straight chain alkenylthiosemicarbazide, C2-C20 straight chain substituted alkenylthiosemicarbazide, C3-C50 branched chain alkenylthiosemicarbazide, C3-C50 branched chain substituted alkenylthiosemicarbazide, C3-C50 cyclic alkenylthiosemicarbazide, C3-C50 cyclic substituted alkenylthiosemicarbazide, C2-C20 straight chain alkynylthiosemicarbazide, C2-C20 straight chain substituted alkynylthiosemicarbazide, C3-C50 branched chain alkynylthiosemicarbazide, C3-C50 branched chain substituted alkynylthiosemicarbazide, C3-C50 cyclic alkynylthiosemicarbazide, C3-C50 cyclic substituted alkynylthiosemicarbazide, C1-C20 straight chain alkylhydrazone, C1-C20 straight chain substituted alkylhydrazone, C2-C50 branched chain alkylhydrazone, C2-C50 branched chain substituted alkylhydrazone, C3-C50 cyclic hydrazoalkane, C3-C50 cyclic substituted hydrazoalkane, C2-C20 straight chain alkenylhydrazone, C2-C20 straight chain substituted alkenylhydrazone, C3-C50 branched chain alkenylhydrazone, C3-C50 branched chain substituted alkenylhydrazone, C3-C50 cyclic hydrazoalkene, C3-C50 cyclic substituted hydrazoalkene, C2-C20 straight chain alkynylhydrazone, C2-C20 straight chain substituted alkynylhydrazone, C3-C50 branched chain alkynylhydrazone, C3- C50 branched chain substituted alkynylhydrazone, C3-C50 cyclic hydrazoalkyne, C3-C50 cyclic substituted hydrazoalkyne, C1-C20 straight chain alkylhydrazide, C1-C20 straight chain substituted alkylhydrazide, C3-C5o branched chain alkylhydrazide, C3-C50 branched chain substituted alkylhydrazide, C3-C50 cyclic alkylhydrazide, C3-C50 cyclic substituted alkylhydrazide, C2-C2Q straight chain alkenylhydrazide, C2-C20 straight chain substituted alkenylhydrazide, C3-C50 branched chain alkenylhydrazide, C3- C50 branched chain substituted alkenylhydrazide, C3-C50 cyclic alkenylhydrazide, C3-C50 cyclic substituted alkenylhydrazide, C2-C20 straight chain alkynylhydrazide, C2- C20 straight chain substituted alkynylhydrazide, C3-C50 branched chain alkynylhydrazide, C3-C50 branched chain substituted alkynylhydrazide, C3-C50 cyclic alkynylhydrazide and C3-C50 cyclic substituted alkynylhydrazide.
As noted above, the conjugate groups of the invention may be attached to the 4'-position of the nucleosidic sugar via a linker. Further linking groups include Ω-aminoalkoxy moieties and Ω-aminoalkylamino moieties for linking a conjugate group to a 4'-hydroxyl group. Many other linking groups are known including many that are commercially available, including heterobifunctional and homobifunctional linking moieties available from the Pierce Co. (Rockford, Il).
A further preferred group of linking groups include those groups used as the internucleoside linkages between 4'-desmethyl nucleosides of the parent application, application serial number 039,834, filed March 30, 1993, the entire contents of which are herein expressed incorporated by reference.
In one embodiment for attaching a conjugate molecule to a 4-desmethyl nucleoside, a linking moiety located on a 4-desmethyl nucleoside compound of the invention may be reacted with an active ester derivative of a conjugation moiety of the invention (e.g., cholic acid). Such linking moieties are particularly useful in extending polyalkylamine moieties extending from the oligomeric compounds of the invention. Active ester derivatives are well known to those skilled in the art. Representative active esters include N-hydroxysuccinimide esters, tetrafluorophenolic esters, pentafluorophenolic esters and pentachlorophenolic esters. For cholic acid, the reaction of a pendant amino group and the active ester produces an oligomer in which cholic acid is attached to the N-terminal position through a linking group. Cholic acid or other such pendent groups can be attached to the carboxy terminal of the oligomeric compound by conversion to the N-hydroxysuccinimide ester thereof, and then by further reaction with the oligomeric compound of interest.
Conjugate molecules of the invention can also be directly connected to the 4'-desmethyl nucleosides compounds of the invention without use of linkers. As for instance a conjugate molecule bearing a hydroxyl group, a thiol group, or an amino group is reacted with a 4'-desmethyl nucleoside compound of the invention that has a leaving group at the 4' position. The OH, SH or NH2 moiety of the conjugate groups displaces the leaving group to directly join the conjugate to the 4'-desmethyl nucleoside compound of the invention.
The oligomeric compounds of the invention are synthesized utilizing at least two different methods, both of which can use art recognized solid state synthesis or solution phase synthesis. In a first process, the "4-desmethyl" nucleoside that is to be functionalized with a conjugate group can be attached to a growing oligomeric compound and then so functionalized. In a second process the 4'-desmethyl nucleoside monomer can be first functionalized and then attached to the remainder of the oligomeric compound.
A conjugate group of a compound of the invention can be linked to a 4'-desmethyl nucleoside monomer or oligomer through displacement of a leaving group from the 4'-position (as numbered using the sugar ring numbering of a pentofuranosyl nucleoside) of a nucleoside, or the 4'- terminal position (as numbered using the sugar ring numbering of a pentofuranosyl nucleoside) of an oligonucleoside. In preferred embodiments, the 4'-desmethyl compound has structure:
Figure imgf000035_0001
where J is a leaving group. Leaving groups are functional moieties which are designed to be easily displaced. Representative leaving groups include alkyl and arylsulfonyls including p-toluenesulfonyl (tosyl), 4- dimethylaminoazobenzenesulfonyl (dabsyl), 5-dimethylaminonaphthalenesulfonyl (dansyl), trifluoromethylsulfonyl (triflate), methylsulfonyl (mesyl); halogens; o- trichloroacetimidates; 2,4,6-trichlorophenyl; dialkyl phosphite and acyloxy leaving groups including acetoxy, benzoyloxy, p-methoxybenzoyloxy and p-methylbenzoyloxy and other known leaving groups. Acyloxy leaving groups (-ORE where RE is C(O)-) are preferred, particularly OC(O)CH3.
RD can be H, hydroxyl, a nucleoside, an activated nucleotide, a nucleotide, an oligonucleotide or an oligonucleoside or a protected derivative thereof.
Bx is a naturally occurring or non-naturally occurring heterocyclic base as described supra, and also includes such bases bearing protecting groups.
The leaving group can be attached above or below the plane of the sugar ring, i.e., cis or trans to the heretocyclic base. The leaving group may therefore be displaced by either a bimolecular or unimolecular mechanism, dependent upon the specific leaving group and reaction conditions employed. In preferred embodiments the leaving group is trans to the hetrocyclic base, and is displaced via a bimolecular nucleophilic displacement.
In preferred methods of the invention compounds having the above noted structure can be incorporated into a wide variety of oligomeric compounds by standard chemistries. Thus, RD can be an activated phosphorous group. In the context of the invention activated phosphorous groups are groups which are used to facilitate the creation of internucleosidic bonds in nucleic acid synthetic regimes. Thus, activated phosphorous groups include phosphate groups having Pv such as, for example, activated phosphate esters, and phosphite groups having PIII such as phosphoramidites or H-phosphonates. An activated nucleotide according to the invention is a nucleoside which bears an activated phosphorous group.
In preferred processes of the invention a compound of the above structure is incorporated as the terminal nucleoside unit of a preformed oligomeric structure. Such oligomeric structures can be any of the various natural or synthetic oligonucleotides or oligonuleotide mimics as known in the art.
In accordance with the present invention, methods which are amenable to automated synthetic schemes, especially solid-state support synthetic schemes, are preferred. While a number of methodologies can be employed, one preferred methodology follows. A nucleoside analog is attached to a solid support in any conventional fashion. It is customary, and preferred, to employ a linker to a solid support such as a polymeric carrier at the 3' position. An oligonucleotide or oligonucleoside is then synthesized as per any of the various known solid state technologies. A final 4'-desmethyl nucleotide is then added to the growing oligomeric compound.
In one process of the invention, the 4'-desmethyl nucleotide is substituted in the 4'-position with leaving group J as described above. The leaving group is then displaced with a linker or directly with a conjugate group of interest. If a linker is utilized the conjugate group is then attached to the linker, either while still on the solid support or post removal from the solid support.
Alternately the 4'-desmethyl nucleotide is first reacted with a linker or directly with a conjugate group. The modified nucleotide is then attached to the growing oligomeric compound on the solid support.
The 4'-desmethyl nucleoside (or nucleotide) is prepared with any base or base analog, Bx, and either a pentofuranosyl moiety, where Q is oxygen, a cyclopentane moiety or fluorinated analogue thereof where Q is CH2, CHF or CF2 or a 4'-deoxy-4'-thio moiety, where Q is S. In certain preferred embodiments of the invention, the nucleoside is a ribonucleoside having a 2'-hydroxyl functionality. Such a functionality can be any of various 2'-groups known in the art. If the 2' functionality is OH
(a normal ribonucleotide), the 2'-hydroxyl groups can be protected as necessary by means well known to persons of ordinary skill in the art. The only requirement for this group and for its protection is that the same be designed so as to not interfere with substantive reactions in accordance with this invention. In other preferred embodiments, the 2'-hydroxyl group will be replaced with other functional groups as for example 2'-alkoxy groups, 2'- alkoxy groups that are substituted with other groups including imidazole and other heterocyclic and amino groups, 2'-halogen, particularly fluoro. Particularly useful 2'-O- substituent groups include 2'-aminopropoxy, 2'- imidazoylbutoxy, 2'-methoxy, 2'-ethoxy, 2'-propoxy, 2'- methoxyethoxy and 2'-deoxy-2'-fluoro.
Additional objects, advantages, and novel features of this invention will become apparent to those skilled in the art upon examination of the following examples thereof, which are not intended to be limiting.
EXAMPLE 1
5'-O-tert-Butyldiphenylsilylthymidine, 2.
A stirring solution of thymidine (50.0 g, 207 mmol) and DMAP (10 mg, 8.2 x 10-2 mmol) in 400 mL of pyridine was treated with TBDPSCl (43.4 g, 248 mmol) at 25°C for 16 h. The solvent was removed under reduced pressure and the residue was diluted with 1 L of AcOEt. The mixture was washed with 5% aqueous HCl (2 x 100 mL) and H2O (100 mL). The organic layer was dried (MgSO4) and concentrated under reduced pressure. The product was purified by silica gel chromatography (CH2Cl2/MeOH 20:1) to give 87.3 g (88%) of 2 as a white solid. An analytical sample was crystallized from diethylether. mp 164-166 °C (170-171 °C per Matulic- Adamic, J. Chem. Soc., Chem. Comm. 1985, 21, 1535)
Rf (CH2Cl2/MeOH 10:1) 0.31. 1H-NMR (CDCl3): 1.08 (s, 9H, C-
Me3), 1.61 (s, 3H, 5-Me), 2.18 (ddd, 1H, J=13.8, 8.5, 6.0
Hz, 2'Hβ), 2.19 (br s, 1H, D2O exchangeable, 3'-OH), 2.44
(ddd, 1H, J=13.8, 5.6, 2.1 Hz, 2'-Hα), 3.25 (br s, 1H, 5'- OH, D2O exchangeable), 3.85 (dd, 1H, J=11.5, 2.5 Hz, 5'-
CHH), 3.98 (dd, 1H J=11.5, 2.3 Hz, 5'-CHH), 4.06 (dd, 1H,
J=2.5, 2.3 Hz, 4'H), 4.55 (dd, 1HJ=6.0, 5.6 Hz, 3'-H), 6.43
(dd, 1H, J=8.5, 5.6 Hz, l'-H), 7.26-7.51 (m, 6H, aromatic-
H), 7.64-7.68 (m, 5H, 6-H and aromatic-H), 9.57 (s, 1H, NH, D2O exchangeable). EXAMPLE 2
N3-Benzyloxymethyl-5'-O-tert-butyldiphenylsilylthymidine, 3.
To a stirred solution of 2 (117.0 g, 243.8 mmol) and Hunig's base (diisopropylethylamine, 63.0 g, 487.5 mmol) in CH2Cl2 (400 mL) at 23 °C was added a solution of benzyl chloromethyl ether (40.7 g, 260.0 mmol) over a 15 min. period. The resultant mixture was maintained at 23 °C and stirred for 14 h. Ether (1 L) was added to the mixture and the ethereal solution was washed with 10% aqueous HCl (2 x 100 mL) and H2O (200 mL). The organic layer was dried
(MgSO4) and concentrated under reduced pressure. The residue was purified by silica gel column chromatography
(CH2Cl2/AcOEt 40:1 then 10:1) to yield 128.9 g (88%) of 3 as a white solid. Rf (CH2Cl2/AcOEt 10:1) 0.31. 1H-NMR (CDCl3) : 1.10 (s, 9H, C-Me3), 1.65 (s, 3H, 5-Me), 2.16 (m, 1H, 2'-
Hβ), 2.41 (m, 1H, 2'-Hβ), 2.53 (br s, 1H 3'-OH), 3.84 (d,
1H, J=8.8 Hz, 5'-CHH), 3.98 (d, 1H, J=8.8 Hz, 5'-CHH), 4.01
(s, 1H, 4'-H), 6.64 (br s, 1H, 3'-H), 4.70 (s, 2H, OCH2Ph),
5.50 (s, 2H, NCH2O), 6.41 (dd, 1H, J=8 .0, 5.8 Hz, 1'-H), 7.20-7.50 (m, 13H, aromatic-H), 7.65-7.69 (m, 3H, 6-H and aromatic-H). 13C-NMR (CDCl3) : 12.86 (-5-CH3), 19.45 (+, C-
Me3), 27.09 (-, C-Me3), 41.16 (+, 2'-C), 64.25 (+, 5'-C),
70.70 (+, O-C-Ph), 71.97 (-, 4'-C), 72.29 (+, N-C-O), 85.51
(-, 3'-C), 87.20 (-, 1'-C), 110.47 (+, 5-C), 127.72, 128.08, 128.36 (-, aromatic-C), 130.25 (-, 6-C), 132.42, 133.00 (+, aromatic-C) 134.39, 135.37, 135.62 (-, aromatic-C) , 137.95 (+, aromatic-C), 151.01 (+, 2-C), 163.68 (+, 4-C).
EXAMPLE 3
N3-Benzyloxymethyl-3' -O-benzoyl-5' -O-tert- butyldiphenylsilyl-thymidirie, 4.
A stirred solution of 3 (128.0 g, 213.3 mmol) in a 4:1 mixture of CH2Cl2/Et3N (500 mL) was treated with (48.4 g, 40 mL, 344.6 mmol) of BzCl at 23 °C for 8 h. The resultant precipitate was removed by filtration. The filtrate was concentrated under reduced pressure to leave the crude product as a brownish syrup. Purification of the syrup by silica gel column chromatography (hexanes/AcOEt 10:1 then 1:1) gave 130.7 g (87%) of 4 as a white solid. Rf (Hexanes/AcOEt 1:1) 0.82 1H-NMR (CDCl3) : 1.40 (s, 9H, C- Me3), 1.60 (s, 3H, 5-Me), 2.37 (ddd, 1H, J=13.8, 9.3, 7.2 Hz, 2'-Hβ), 2.62 (dd, 1H, J=13.8, 4.3 Hz, 2'-Hβ), 4.09 (m, 2H, 5'-H), 4.26 (m, 1H, 4'-H), 4.74 (s, 2H, 0-CH2-Ph), 5.54 (s, 2H, N-CH2-O), 5.71 (d, 1H, J=7.2 Hz, 3' -H), 6.57 (dd, 1H, J=9.3, 4.3 Hz, 1'-H), 7.24-7.74 (m, 13H, aromatic-H), 8.05-8.15 (m, 3H, 6 -H and aromatic-H). 13C-NMR (CDCl3) : 12.82 (-, 5-Me), 19.54 ( + , C-Me3), 27.16 (C-Me3), 38.28 ( + , 2'-C), 64.41 (+, 5'-C), 70.80 (+, O-C-Ph), 72.29 (+, N-C-O), 75.60 (-, 4'-C), 85.28 (-, 1'-C and 3'-C), 110.95 ( +, 5-C), 127.72, 128.23, 128.37, 128.50, 128.63 (-, aromatic-C), 129.43 (+, aromatic-C), 129.84, 130.22 (-, aromatic-C), 132.14, 133.07 (+, aromatic-C), 133.60 (-, 6-C), 133.94, 135.32, 135.65 (-, aromatic-C), 138.15 (+, aromatic-C), 151.19 (+, 2-C), 163.50 (+, 4-C), 166.11 (+, benzoyl C=O).
EXAMPLE 4
N3-Benzyloxymethyl-3'-O-benzoylthymidine, 5.
The silyl ether 4 (96.0 g, 136.4 mmol) in THF (600 mL) was treated with hydrogen fluoride-pyridine (70% HF in pyridine, 30 mL) at 0 °C for 4 h under a N2 atmosphere. The resultant mixture was diluted with AcOEt (600 mL) and washed with H2O (2 x 300 mL). The organic layer was dried (MgSO4) and concentrated at reduced pressure. The residue was purified by silica gel column chromatography (CH2Cl2/AcOEt 10:1) to give 61.6 g (97%) of 5 as a white solid. Rf (CH2Cl2/AcOEt 10:1) 0.29. 1H-NMR (CDCl3+D2O) : 1.95 (s, 3H, 5-Me), 2.53 (m, 2H, 2'-H), 4.00 (m, 2H, 5'-H), 4.25 (m, 1H, 4'-H), 4.71 (s, 2H, O-CH2-Ph), 5.51 (s, 2H, N-CH2-O), 5.60 (m, 1H 3'-H), 6.36 (dd, 1H, J=7.6, 6.6 Hz, 1'-H), 7.25-7.66 (m, 9H, 6-H and aromatic-H), 8.05 (d, 2H, J=7.1 Hz, aromatic-H). 13C-NMR (CDCl3) : 13.29 (-, 5-Me), 37.82 (+, 2'- C), 62.54 (+, 5'-C), 70.73 (+, O-C-Ph), 72.25 (+, N-C-O), 75.82 (-, 4'-C), 85.43 (-, 3'-C), 86.13 (-, 1'-C), 110.41 (+, 5-C), 127.65, 128.36, 128.59 (-, aromatic-C), 129.34 (+, aromatic-C), 129.69 (-, 6-C), 133.60, 135.40 (-, aromatic- C), 137.87 (+, aromatic-C), 151.18 (+, 2-C), 163.65 (+, 4- C), 166.11 (+, benzoyl C=O).
EXAMPLE 5
tert-Butyl N3-Benzoxyιαethyl-3'-O-benzoylthymidine-5'- carboxylate, 6 .
The reaction was performed as described by Corey and Samuelsson, J. Org. Chem. 1984, 49, 4735, for the oxidation of 5' -OH of the uridine derivative to its corresponding 5'- tert-butyl carboxylate. Chromium (VI) oxide (31.4 g, 314.2 mmol) in CH2Cl2 (480 mL) was cooled to 0 °C and then pyridine (49.7 g, 638.4 mmol) in DMF (120 mL) was added dropwise to the reaction mixture (caution: extremely exothermic) over a period of 1 h. The mixture was stirred at 0 °C for 30 min. Alcohol 5 (36.6 g, 78.5 mmol) in CH2Cl2/DMF (4:1 v/v, 100 mL) was added followed by acetic anhydride (64.1 g, 628.4 mmol) and t-BuOH (116.4 g, 1.57 mmol). The resultant mixture was warmed to 23 °C and stirred for 18 h. Ethanol (20 mL) was added to the reaction and the mixture was stirred for additional 15 min. The reaction mixture was poured into AcOEt (400 mL) and the insoluble material was filtrated through a Buchner funnel padded with 200 g of silica gel and 50 g of MgSO4. The solid left in the funnel was rinsed with AcOEt (4 x 100 mL). The combined filtrate and rinses were concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexanes/AcOEt 3:1) to give 25.6 g (61%) of 5 as a white solid. An analytical sample (about 400 mg) was crystallized from ether/hexanes afforded white, needle-like crystals. mp 80-82°C. Rf (hexanes/AcOEt 3:1) 0.23. 1H-NMR(CDCl3) : 1.55 (s, 9H, C-Me3), 2.21 (ddd, 1H, J=14.3, 9.1, 5.0 Hz, 2'-Hβ), 2.61 (dd, 1H, J=14.3, 5.2 Hz, 2'-Hα), 4.63 (s, 1H, 4'-H), 4.71 (s, 2H, O-CH2-Ph), 5.51 (s, 2H, N-CH2-O), 5.65 (d, 1H, J=5.0 Hz, 3'-H), 6.61 (dd, 1H, J=9.1, 5.2 Hz, 1'-H), 7.24-7.63 (m, 8H, aromatic-H), 8.07 (d, 2H, J=7.1 Hz, aromatic-H), 8.09 (s, 1H, 6-H). 13C- NMR(CDCl3) : 13.40 (-, 5-Me), 27.92 (-, C-Me3), 36.65 (+ , 2'-C), 70.58 (+, O-C-Ph), 72.09 (+, N-C-O), 76.66 (-, 4'C), 82.55 (-, 3'-C), 83.36 (+, C-Me3), 86.88 (-, 1'-C), 110.61
(+, 5-C), 127.55, 127.24 128.23, 128.53 (-, aromatic-C), 128.99 (+, aromatic-C), 129.78 (-, 6-C), 133.71, 134.72 (-, aromatic-C), 138.06 (+, aromatic-C), 151.14 (+, 2-C), 163.28
(+, 4-C), 165.26 (+, benzoyl C=O), 169.40 (+, 5'-C).
EXAMPLE 6
N3-Benzoxylmethyl-3'-O-benzoylthymidine-5'-carboxylicacid, 7.
A solution of 6 (22.0 g, 41.0 mmol) in CF3COOH (100 mL) was stirred at 23 °C for 2 h. Toluene (200 Ml) was then added and the mixture was concentrated under reduced pressure. The coevaporation of toluene was repeated twice to ensure complete removal of the CF3COOH. The resultant light yellow powder was purified by silica gel column chromatography (CH2Cl2/AcOEt 8:1) to afford 19.3 g (87%) of 7 as a white powder. Rf (CHCl3/MeOH 4:1) 0.39, 1H- NMR(CDCl3+D2O) : 1.95 (s, 3H, 5-Me), 2.27 (ddd, 1H, J=14.3, 9.1, 4.9 Hz, 2'-Hβ), 2.68 (dd, 1H, J=14.3, 5.2 Hz, 2'Hα), 4.71 (s, 2H, O-CH2-Ph), 4.79 (s, 1H, 4'-H), 5.52 (s, 2H, N- CH2-O), 5.76 (d, 1H, J=4.9 Hz, 3'-H), 6.55 (dd, 1H, J=9.1, 5.2 Hz, 1'-H), 7.24-7.60 (m, 8H, aromatic-H), 7.97 (s, 1H, 6-H), 8.06 (d, 2H, J=7.1 Hz, aromatic-H). 13C-NMR (CDCl3) : 13.42 (+, 5-Me), 36.68 (+, 2'-C), 70.83 (+, O-C-Ph), 73.38 (+, N-C-O), 76.93 (-, 4'-C), 82.01 (-, 3'-C), 87.58 (-, 1'- C) , 110.80 (+, 5-C), 127.72, 127.86, 128.39, 128.70 (-, aromatic-C), 128.71 (+, aromatic-C), 129.90 (-, 6-C), 133.95, 135.63 (-, aromatic-C), 128.53 (+, aromatic-C), 11.20 (+, 2-C), 163.94 (+, 4-C), 165.61 (+, benzoyl C=O), 171.93 (+, 5' -C).
EXAMPLE 7
(2'S, 4'S, 5'R)-1-[5'-Acetoxy-4'-benzoyloxy-tetrahydrofuran- 2' -yl] -N3-benzoxymethylthymine, 8.
A stirred solution of 7 (10.6 g, 22.0 mmol) in DMF (75 mL) was treated with Pb(OAc)4 (11.8 g, 26.5 mmol) at 23 °C for 2 h under darkness. The mixture was diluted with AcOEt (250 mL) and the resultant suspension was filtrated through a Celite pad (50 g). The solid was rinsed several times with AcOEt. The combined filtrate and rinses were concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexanes/AcOEt 1:1) to give 6.5 g (60%) of a 3:7 α/β (as determined by the 1H-NMR 2'-H ratio) anomeric mixture of 8 as a light yellow syrup. An aliquot of the anomeric mixture (~0.2 g) was separated on a silica gel column (hexanes/AcOEt 8:1 → 2:1 gradient) to afford 53 mg of 8α and 121 mg of 8β, both as white foams.
8α.: Rf (hexanes/AcOEt, 1:1) 0.53. 1H -NMR (CDCl3) : 1.95 (d, 3H, J = 1.1 Hz, 5-CH3), 2.05 (s, 3H, acetoxy-CH3), 2.52-2.75
(m, 2H, 3'-H), 4.70 (s, 2H, OCH2Ph), 5.49 (s, 2H, NCH2O),
5.60-5.65 (m, 1H, 4'-H), 6.40 (dd, 1H, J = 7.9, 3.8 Hz, 2'-
H), 6.72 (d, 1H, J = 4.0 Hz, 5'-H), 7.06 (d, 1H, J = 1.1 Hz,
6-H), 7.22-7.60 (m, 8H, aromatic-H), 8.02 (d, 2H, J = 7.6 Hz, aromatic-H). 13C-NMR (CDCl3) : 13.04 (-, 5-CH3), 20.73 (- , acetoxy-CH3), 33.81 (+, 3'-C), 70.45 (+ , O-C-Ph), 71.08 (- , 4'-C), 72.11 (+, N-C-O), 85.47 (-, 2'-C), 94.10 (-, 5'-C), 110.90 (+, 5-C), 127.47, 128.11, 128.45 (-, aromatic-C), 128.64 (+, aromatic-C), 129.54 (-, 6-C), 133.55, 133.92 (-, aromatic-C), 137.75 (+, aromatic-C), 150.55 (+, 2-C), 163.01
(+, 4-C), 165.43 (+, benzoyl C=O), 169.18 (+, acetoxy C=O) .
Anal. Calcd. for C26H26N2O8 : C, 63.16; H, 5.26; N, 5.67.
Found: C, 63.12; H, 5.38; N, 5.45.
8ß: Rf (hexanes/AcOEt 1:1) 0.47. 1H-NMR (CDCl3) : 1.95 (s, 3H, 5-CH3), 2.18 (s, 3H, acetoxy-CH3), 2.33 (ddd, 1H, J = 14.9, 8.2, 4.9 Hz, 3'-Hβ), 2.75 (dd, 1H, J = 14.9, 6.2 Hz, 3'-Hα), 4.70 (s, 2H, OCH2Ph), 5.50 (s, 2H, NCH2O), 5.52 (d, 1H, J = 4.9 Hz, 4'-H), 6.42 (s, 1H, 5'-H), 6.73 (dd, 1H, J = 8.2, 6.2 Hz, 2'-H), 7.22-7.63 (m, 9H, 6-H and aromatic-H), 8.04 (d, 2H, J = 7.5 Hz, aromatic-H). 13C-NMR (CDCl3) : 13.40 (-, 5-CH3), 20.82 (-, acetoxy-CH3), 34.95 (+ , 3'-C), 70.51 (+, O-C-Ph), 72.07 (+, N-C-O), 76.64 (-, 4'-C), 87.15 (-, 2'-C), 98.61 (-, 5'-C), 111.03 (+, 5-C), 127.45, 128.11, 128.45 (-, aromatic-C), 129.68 (-, 6-C), 133.02, 133.69 (-, aromatic- C), 137.77 (+, aromatic-C), 150.91 (+, 2-C), 162.84 (+, 4- C), 165.12 (+, benzoyl C=O), 169.13 (+, acetoxy C=O). Anal. Calcd for C26H26N208 H2O: C, 60.94; H, 5.47; N, 5.47. Found: C, 60.98; H, 5.18; N, 5.30.
EXAMPLE 8
(2'R,4'S,5'S)-(4'-Benzoxy-5'-O-polyethyleneglycoltetrahydro- furan-2'-yl)-N3-(benzyloxymethyl)thymine (9)
A solution of 3.7 g (7.5 mmol) of acetate 8, 25-35 mmol of PEG alcohol, and 4 mL (3.0 g, 30 mmol) of Et3Ν in 150 mL of CH2Cl2 was added 14 mL (16.2 g, 75 mmol) of TMSOTf at -23 °C under Ar atmosphere. The reaction mixture was stirred at -23 °C for 2 h, then placed in the freezer (-15°C) for 16-24 h. The resultant mixture was poured into a 500 mL bilayer mixture of AcOEt/H2O (9:1) containing 15 mL of Et3N. The organic layer was dried (MgSO4) and concentrated at reduced pressure. The residue was purified by Si02 column chromatography to yield 9.
9a: yield 84.9% (colorless syrup). Rf (hexanes/AcOEt 1:1) 0.22. 1H NMR (CDCl3+D2O) δ 1.95 (s, 3H, 5-CH3), 2.34 (ddd, J = 14.3, 8.1, 5.1 Hz, 3' -Hα), 2.62 (dd, 1H, J = 14.3, 6.5 Hz, 3'-Hß), 3.60-3.95 (m, 4H), 4.70 (s, 2H, OCH2Ph), 5.25 (s, 1H, 5'-H), 5.47 (d, J = 5.1 Hz, 4'-H), 5.50 (s, 2H, NCH2O), 6.80 (dd, 1H, J = 8.1, 6.5 Hz, 2'-H), 7.18-7.65 (m, 9H, 6-H and aromatic-H), 8.04-8.07 (m, 2H, aromatic-H). 13C NMR (CDCl3+D2O) δ 13.16, 35.00 (3'-C), 61.41, 70.33, 70.76, 72.29, 77.67 (4'-C), 86.53 (2'-C), 106.61 (5'-C), 111.56 (5-C), 127.66, 128.06, 128.33 (6-C), 128.97, 129.84, 133.80, 134.38, 137.99, 151.41 (2-C), 163.35 (4-C), 165.76 (benzoyl C=O).
9b: yield 84.3% (colorless syrup). Rf (hexanes/AcOEt 1:1) 0.18. 1H NMR (CDCl3) δ 1.95 (s, 3H, 5-CH3), 2.34 (ddd, 1H, J = 14.7, 8.1, 4.9 Hz, 3'-Hα), 2,62 (dd, 1H, J = 14.7, 6.3 Hz, 3'-Hδ), 3.36 (s, 3H, CH3O), 3.54-3.98 (m, 12 H), 4.72 (S, 2H, OCH2Ph), 5.24 (s, 1H, 5'-H), 5.49 (d, 1H, J = 4.9 Hz, 4'-H), 5.52 (s, 2H, NCH2O), 6.80 (dd, 1H, J = 8.1, 6.3 Hz, 2'-H), 7.26-7.65 (m, 9H, 6-H and aromatic-H), 8.02- 8.06 (m, 2H, aromatic-H). 13C NMR (CDCl3) δ 13.29, 35.06 (3'- C), 59.04 (CH30), 68.00, 70.14, 70.61, 71.91, 72.19, 77.51 (4'-C), 86.38 (2'-C), 106.43 (5'-C), 111.43 (5-C), 127.65, 128.31, 128.60 (6-C), 129.07, 129.81, 133.70, 134.45, 138.04, 151.41 (2-C), 163.28 (4-C), 165.51 (benzoyl C=O) .
9c: 76.5% yield (white foam). Rf (hexanes/AcOEt 1:1) 0.05. 1H NMR (CDCl3) δ 1.98 (s, 3H, 5-CH3), 2.37 (ddd, 1H, J = 14.7, 8.2, 4.9 Hz, 3' -Ha), 2,62 (dd, 1H, J = 14.7, 6.5
Hz, 3'-Hβ), 2.78 (br s, 1H, OH, D2O exchangable), 3.57-3.99
(m, 16 H), 4.72 (s, 2H, OCH2Ph), 5.25 (s, 1H, 5'-H), 5.50
(d, 1H, J = 4.9 Hz, 4'-H), 5.52 (s, 2H, NCH2O), 6.80 (dd,
1H, J = 8.2, 6.5 Hz, 2'-H), 7.26-7.65 (m, 9H, 6-H and aromatic-H), 8.02-8.06 (m, 2H, aromatic-H). 13C NMR (CDCl3) δ 13.27, 35.02 (3'-C), 61.70, 68.02, 70.15, 70.28, 70.54, 70.73, 72.26, 72.61, 77.61 (4'-C), 86.47 (2'-C), 106.55 (5'- C), 111.40 (5-C), 127.66, 128.31, 128.60 (6-C), 129.12, 129.84, 133.71, 134.48, 138.04, 151.45 (2-C), 163.35 (4-C), 165.61 (benzoyl C=O).
9d: 75.9% yield (yellowish syrup). Rf (hexanes/AcOEt 1:1) 0.10. 1H NMR (CDCl3) δ 1.96 (s, 3H, 5-CH3), 2.34 (ddd, 1H, J = 14.7, 7.9, 4.6 Hz, 3'-Hα), 2.37 (br s, 1H, OH, D2O exchangable), 2,62 (dd, 1H, J = 14.7, 6.8 Hz, 3'-Hβ), 3.44- 4.01 (m, 24 H), 4.72 (s, 2H, OCH2Ph), 5.25 (s, 1H, 5'-H), 5.48 (d, 1H, J = 4.6 Hz, 4'-H), 5.52 (s, 2H, NCH2O), 6.79 (dd, 1H, J = 7.9, 6.8 Hz, 2'-H), 7.25-7.65 (m, 9H, 6-H and aromatic-H), 7.98-8.06 (m, 2H, aromatic-H). 13C NMR (CDCl3) δ 13.03, 34.85 (3'-C), 61.28, 67.83, 70.12, 70.33, 71.38, 71.96, 72.47, 77.44 (4'-C), 86.22 (2'-C), 106.27 (5'-C), 111.08 (5-C), 127.39, 128.08, 128.44 (6-C), 128.94, 129.60, 133.54, 134.38, 137.92, 151.17 (2-C), 163.08 (4-C), 165.29 (benzoyl C=O).
EXAMPLE 9
(2'R,4'S,5'S)-(4'-Hydroxy-5'-O-polyethyleneglycoltetrahydro furan-2'-yl)thymine (10) A suspension of 4.5 mmol of 9 and 2-3 g of Pd(OH)2/C in 40 mL of 3:1 MeOH/acetone was stirred at 23 °C under H2 atmosphere for 16 h. The suspended material was filtrated through a pad of Celite in a Bϋchner funnel and the solid in the funnel was rinsed several times with acetone. The combined filtrate and rinses were concentrated at reduced pressure to give a light yellow syrup. The syrup was redissolved in 25 mL of MeOH and 0.3 g (7.5 mmol) of NaOH was added. The mixture was stirred at 23 °C for 6 h and concentated at reduced pressure. The residue was purifed by Si02 column chromatography to yield 10.
10a: yield 88.3% (white solid), mp 210 °C (dec.). Rf (CHCl3/MeOH 9:1) 0.22. 1H NMR (D2O) δ 1.84 (s, 3H, 5-CH3), 2.34 (m, 2H, 3'-H), 3.57-3.83 (m, 4H), 4.42 (dd, J = 3.7, 2.1 Hz, 4' -H), 5.08 (s, 1H, 5' -H), 6.56 (t, 1H, J = 7.1 Hz, 2'-H), 7.56 (s, 1H, 6-H). 13C NMR (DMSO-d6) δ 12.07, 36.94 (3'-C), 59.98, 69.33, 74.11 (4'-C), 84.99 (2'-C), 108.50 (5'-C), 110.43 (5-C), 135.96 (6-C), 150.71 (2-C), 163.67 (4- C) . Anal Calcd for C11H16N2O60.7CH3OH : C, 47 . 69 ; H, 6 . 38 ; N, 9 . 51 . Found : C, 47 . 70 ; H, 6 . 01 ; N, 9.52.
10b: yield 90.6% (white solid). mp 54-56 °C. Rf (CHCl3/MeOH 9:1) 0.41. 1H NMR (CDCl3) δ 1.93 (s, 3H, 5-CH3), 2.11 (ddd, 1H, J - 14.2, 7.9, 4.9 Hz, 3' -Hα), 2.33 (br s, 1H, 4' -OH), 2,41 (dd, 1H, J = 14.2, 6.5 Hz, 3' -Hβ), 3.38 (s, 3H, CH3O), 3.54-3.94 (m, 12 H), 4.37 (d, 1H, J= 4.9 Hz, 4'- H), 5.14 (s, 1H, 5'-H), 6.68 (dd, 1H, J = 7.9, 6.5 Hz, 2'- H), 7.45 (s, 1H, 6-H), 9.25 (s, 1H, NH). 13C NMR (CDCl3) δ 12.50, 37.42 (3'-C), 58.84 (CH30), 67.52, 70.25, 70.40, 71.71, 75.01 (4'-C), 85.90 (2'-C), 109.23 (5'-C), 111.60 (5- C), 136.29 (6-C), 151.10 (2-C), 164.40 (4-C). Anal Calcd for C16H26N2OB2H2O: C, 46.83; H, 7.32; N, 6.83. Found: C, 46.94; H, 7.50; N, 6.74.
10c: yield 85.9% (white foam). Rf (CHCl3/MeOH 9:1) 0.36. 1H NMR (CDCl3) δ 1.92 (s, 3H, 5-CH3), 2.09 (ddd, 1H, J = 14.2, 7.8, 4.9 Hz, 3'-Hα), 2,41 (dd, 1H, J = 14.2, 6.6 Hz, 3'-Hβ), 3.58-3.90 (m, 16 H), 4.36 (d, 1H, J = 4.9 Hz, 4'-H), 5.20 (s, 1H, 5'-H), 6.63 (dd, 1H, J = 7.8, 6.6 Hz, 2'-H), 7.46 (s, 1H, 6-H), 8.87 (s, 1H, NH). 13C NMR (CDCl3) δ 12.53, 37.35 (3'-C), 61.26, 67.47, 70.00, 70.35, 72.51, 74.99, 77.71 (4'-C), 85.90 (2'-C), 109.27 (5'-C), 111.47 (5- C), 136.33 (6-C), 151.05 (2-C), 164.45 (4-C). Anal Calcd for C17H28N2O9·0.5 H2O: C, 49.39; H, 7.02; N, 6.78. Found: C, 49.58; H, 7.12; N, 6.66.
10d: yield. 87.3% (white wax). Rf (CHCl3/MeOH 10:1) 0.40. 1H NMR (CDCl3) δ 1.92 (s, 3H, 5-CH3), 2.08 (ddd, 1H, J = 13.8, 7.7, 4.8 Hz, 3'-Hα), 2,39 (dd, 1H, J = 13.8, 6.5 Hz, 3'-Hβ), 2.81 (s, 1H, 4' -OH), 3.32-3.92 (m, 24 H), 4.01
(s, 1H, OH), 4.37 (d, 1H, J - 4.8 Hz, 4'-H), 5.16 (s, 1H,
5'-H), 6.66 (dd, 1H, J = 7.8, 6.5 Hz, 2'-H), 7.45 (s, 1H,
6-H), 8.90 (s, 1H, NH). 13C NMR (CDCl3) δ 12.49, 37.50 (3'-
C), 61.40, 67.53, 70.14, 70.41, 72.53, 75.01 (4'-C), 85.92 (2'-C), 109.27 (5'-C), 111.50 (5-C), 136.20 (6-C), 151.00
(2-C), 164.25 (4-C). Anal Calcd for C21H36N2On·0.75 H2O: C, 49.85; H, 7.42; N, 5.54. Found: C, 49.79; H, 7.50; N, 5.60.
EXAMPLE 10
(2'R,4'S,5'S)-(4'-(2-Cyanoethyl-N,N-diisopropylaminophosphor ityl)-5'-0-(dimethoxytrityloxytetraethyleneglycol)tetrahydro furan-2'-yl)thymine (12)
A solution of 1.3 g (3.2 mmol) of 10c, 1.0 x 10-2 g (8.2 x 10-2 mmol) of DMAP, and 1.0 g (10.0 mmol) of Et3Ν in 30 mL of pyridine was treated with 1.6 g (4.8 mmol) of DMTrCl at 23 °C for 14 h. The solvent was removed at reduced pressure and the residue was purified by Si02 column chromatography (CHCl3/MeOH 20:1 then 10:1) gave 1.9 g (83.6%) of 11 as a yellow foam. Rf (CHCl3/MeOH 20:1) 0.23. 1H NMR (CDCl3) δ 1.92 (s, 3H, 5-CH3), 2.04 (ddd, 1H, J = 14.2, 7.6, 4.8 Hz, 3'-Hα), 2,32 (dd, 1H, J = 14.2, 7.0 Hz, 3'-Hß), 3.23 (t, 2H, J = 5.0 Hz), 3.29 (br s, 1H, 4'-OH), 3.59-3.89 (m, 14 H), 3.79 (s, 6H, 2 x OCH3), 4.31 (d, 1H, J = 4.8 Hz, 4'-H), 5.10 (s, 1H, 5'-H), 6.62 (dd, 1H, J = 7.6, 7.0 Hz, 2'-H), 6.82 (d, 4H, J= 8.9 Hz), 7.20-7.48 (m, 10H, 6-H and aromatic-H), 8.38 (s, 1H, NH). 13C NMR (CDCl3) δ 12.69, 37.58 (3'-C), 55.26, 63.15, 67.74, 70.73, 75.24 (4'- C), 86.01 (2'-C), 86.08, 109.49 (5'-C), 111.80 (5-C), 113.12, 126.74, 127.68, 127.83, 128.24, 130.13, 136.31 (6- C), 145.13, 151.24 (2-C), 158.41, 164.44 (4-C).
A solution of 1.8 g (2.6 mmol) of 11 and 3 x 10-2 g (1.8 x 10-1 mmol) of N, N-diisopropylammonium tetrazolide in 50 mL of CH2Cl2 was treated with 1.1 g (3.6 mmol) of 2- cyanoethoxy-N, N-diisopropylphosphoramidite at 23 °C for 16 h. The mixture was then dilutd with 100 mL CH2Cl2 and washed with sat. aqueous ΝaHCO3 (30 mL) and brine (30 mL). The organic layer was dried (MgSO4) and concentrated at reduced pressure. The residue was purified by SiO2 column chromatography (0.5% Et3N in AcOEt) gave 1.9 g (77.9%) of 12 as a slightly yellow foam. 31P NMR (CDCl3) δ 149.33 and 149.53 ppm. EXAMPLE 11
2-(2-Phthalimidoethoxy)ethanol (13)
A suspension of 30.0 g (202.5 mmol) of phthalic anhydride, 20 mL (21.0 g, 199.4 mmol) of 2- (2-aminoethoxy)- ethanol in 400 mL of benzene was added 1.0 x 10-2 g (5.3 x 10-5 mmol) of TsOHH2O. The reaction flask was connected to a Dean-Stark condenser and then heated to reflux for 14 h. The solvent was removed at reduced pressure and the residue was purified by SiO2 column chromatography (hexanes/AcOEt 1:1 then CHCl3/MeOH 20:1) gave 43.7 g (93.1%) of 13 as a light yellow syrup. This syrup slowly crystallized on standing at room temperature overnight as needle-like crystals, mp 66-66.5 °C. Rf (hexanes/AcOEt 1:1) 0.17. 1H NMR (CDCl3) d 2.41 (t, 1H, J = 6.2 Hz, OH), 3.57-3.93 (m, 8 H), 7.69-7.87 (m, 4H). 13C NMR (CDCl3) d 37.52, 61.62, 68.24, 72.24, 123.36, 131.97, 134.00, 168.38 (C=O) . Anal Calcd for C12H13NO4 : C, 61.28; H, 5.53; N, 5.96. Found: C, 61.34; H, 5.62; N, 5.94.
EXAMPLE 12A
(2'R,4'S,5'S)-4'-Benzoxy-5'-O-((2-(2-phthalimidoethoxy)- ethyl)-tetrahydrofuran-2'-yl)-N3-(benzoxymethyl)thymine (14) A solution of 10.8 g (22.0 mmol) of 8, 10.0 g (42.6 mmol) of 13, and 5.7 g (8.0 mL, 57.0 mmol) of Et3N in 200 mL of CH2CL2 was chilled to -23 °C. To this solution, 16.9 g (14.6 mL, 76.0 mmol) of TMSOTf was added in one portion. The resultant mixture was stirred at -23 °C for 15 minutes and then placed in the freezer (-15 °C) for 16 hours. The reaction was quenched by pouring into 1 L of a 4 : 1 mixture of AcOEt/H2O containing 20 mL of Et3N. The organic layer was washed with H2O (300 mL), dried (MgSO4), and concentrated at reduced pressure. The residue was purified by SiO2 column chromatography (hexanes/AcOEt 1:1) and gave 12.4 g (84.8 %) of 14 as very hydroscopic white foam. Rf (hexanes/AcOEt 1:1) 0.22. 1H NMR (CDCl3) δ 1.89 (s, 3H, 5-CH3), 2.30 (ddd, 1H, J = 13.2, 8.3, 4.6 Hz, 3' -HJ, 2.57 (dd, 1H, J = 13.2, 6.5 Hz, 3'-Hβ), 3.63-4.01 (m, 8 H), 4.71 (s, 2H, OCH2Ph), 5.19
(s, 1H, 5'-H), 5.36 (d, 1H, J = 4.6 Hz, 4'-H), 5.48 (s, 2H,
NCH2O), 6.72 (dd, 1H, J = 8.3, 6.5 Hz, 2'-H), 7.28-8.01 (m,
13H, 6-H and aromatic-H), 8.02 (dd, 2H, J= 7.8, 1.5 Hz, aromatic-H). 13C NMR (CDCl3) δ 13.01 (5-CH3), 34.87 (3'-C), 36.91, 67.64, 67.78, 69.34, 70.62, 72.05, 73.60 (4'-C), 86.30 (2'-C), 106.16 (5'-C), 111.13 (5-C), 123.08, 127.51, 127.76, 128.21, 128.50, 129.20, 129.70, 131.92, 133.54 (6- C), 134.00, 134.41, 138.08, 151.26 (2-C), 163.07 (4-C), 165.29 (phthalimido C=O) 168.02 (benzoxy C=O). Anal Calcd for C36H35N3O10 0.7 CHCl 3 : C, 58.51; H, 4.74; N, 5.58. Found: C, 58.78; H, 4.74; N, 5.60.
EXAMPLE 12B
(2'R,4'S,5'S)-4'-Benzoxy-5'-O-((S)-(2,3-ditetradecanyloxy)- propoxy)-tetrahydrofuran-2'-yl)-N3-(benzoxymethyl)thymine (12f)
(S)-1-0-(4-Methoxybenzyl)-2,3-O-isopropylidene-glycerol (12b)
A suspension of (S) -2-2-dimethyl-l, 3-dioxolane-4- methanol (5.0 g, 37.8 mmol) and NaH (1.4 g, 58.3 mmol) in DMF (100 mL) was treated with 4-methoxybenzyl chloride (7.7 g, 49.2 mmol) at 0ºC for 4 hours. The resultant mixture was diluted with ether (300 mL) then washed with H2O (100 mL).
After standard work-up and purification procedure, the title compound was obtained (8.7 g, 91.1%) as a cololess liquid. bp 152-155 ºC/1.1 torr. Rf (hexanes/AcOEt 3:1) 0.44. 1H NMR
(CDCl3) δ 1.36 (s, 3H), 1.42 (s, 3H), 3.43 (dd, 1H, J = 9.7,
5.7 Hz, 1-CHH), 3.53 (dd, 1H, J = 9.7, 5.6 Hz, 1-CHH), 3.72
(dd, 1H, J = 8.2, 6.3 Hz, 3-CHH), 3.80 (s, 3H, OCH3), 4.05
(dd, 1H, J = 8.2, 6.3 Hz, 3-CHH), 4.29 (m, 1H, 2-H), 4.50 (ABq, 2H, J = 11.7 Hz), 6.86-6.90 (m, 2H), 7.26-7.28 (m,
2H). 13C NMR (CDCI3) δ 25.45, 26.83, 55.20, 66.93, 70.82 (1-
C), 73.16 (3-C), 74.80 (2-C), 109.34, 113,82, 129.40,
130.09, 159.32.
(R)-1-(4-Methoxybenzyloxy)-2,3-dihydroxypropane (12c)
A solution of 12b (8.5 g, 33.7 mmol) in THF (100 mL) was treated with 10% aqueous HCl (10 mL) at 0 ºC for 4 hours. The reaction was quenched by adding Et3N (20 mL) at 0 ºC. After standard work-up and purification procedure, 12c was obtained (7.0 g, 97.9%) as a slightly yellow syrup. Rf (CHCl3/MeOH 10:1) 0.12. 1H NMR (CDCl3) δ 2.45 (br s, 2H, OH), 2.51-3.74 (m, 4H), 3.53 (dd, 1H, J = 9.7, 5.6 Hz, 1- CHH), 3.72 (dd, 1H, J = 8.2, 6.3 Hz, 3-CHH), 3.81 (s, 3H, OCH3), 3.85-3.90 (m, 1H) 4.29 (m, 1H, 2-H), 4.87 (s, 2H), 6.86-6.91 (m, 2H), 7.24-7.28 (m, 2H). 13C NMR (CDCl3) δ 55.30, 64.05, 70.87 (2-C), 71.40 (1-C), 73.19 (3-C), 113,89, 129.53, 129.87, 159.36.
(R)-1-(4-Me4hoxybenzyloxy)-2,3-(ditetradecanyl-1-oxy)propane (12d)
A suspension of 12c (7.0 g, 33.0 mmol) and NaH (3.2 g, 132.0 mmol) in DMF (100 mL) was treated with 1- bromotetradecane (27.5 g, 99.0 mmol) at 23 ºC for 14 hours. The mixture was diluted with AcOEt (500 mL) and washed with H2O (100 mL). After standard work-up and purification procedure folloewd by distillation, 12d was obtained (16.1 g, 80.7%) as a semi-solid wax. bp 244-248 ºC/1.5 torr. Rf (hexanes/AcOEt 40:1) 0.22. 1H NMR (CDCl3) δ 0.88 (t, 6H. J = 6.7 Hz), 1.26 (m, 48H) 1.51-1.60 (m, 4H), 3.42 (t, 1H, J = 6.7 Hz, 3-CHH), 3.47-3.56 (m, 8H), 3.81 (s, 3H, OCH3), 4.48 (S, 2H), 6.85-6.89 (m, 2H), 7.24-7.28 (m, 2H). 13C NMR (CDCl3) δ 14.17, 22.76, 26.19, 27.74, 30.17, 32.00, 55.18, 69.99, 70.60, 70.85, 71.67 (1-C), 73.04 (3-C), 77.99 (2-C), 113,72, 129.21, 130.56, 159.17.
(S)-(2,3-Ditetradecanyloxy)propan-1-ol (12e)
A solution of 12d (14.0 g, 23.2 mmol) in CH2Cl2 (180 mL) was added to bromodimethylborane (4.0 g, 32.8 mmol) at - 78 ºC and the solution was warmed up to 0º C and stirred for 5 minutes. The resultant mixture was poured into a solution of 4:1 ether/H2O (500 mL). After standard work-up and purification procedure, 12e was obtained (11.1 g, 98.9%) as a white solid. Rf (hexanes/AcOEt 9:1) 0.10. 1H NMR (CDCI3) δ 0.88 (t, 6H. J = 6.7 Hz), 1.26 (m, 48H) 1.51-1.60 (m, 4H), 2.19 (t, 1H, J = 6.6 Hz, OH), 3.43 (t, 1H, J = 6.7 Hz, 3-CHH), 3.47-3.78 (m, 8H). 13C NMR (CDCl3) δ 14.13, 22.72, 26.13, 29.41, 29.52, 29.70, 30.11, 31.96, 63.00 (1- C), 70.42, 70.88, 71.84 (3-C), 78.41 (2-C).
(2'R,4'S,5'S)-4'-Benzoxy-5'-O-((S)-(2,3-ditetradecanyloxy)- propoxy)-tetrahydrofuran-2'-yl)-N3-(benzoxymethyl)thymine (12f)
The title compound was prepared according to the procedure of Example 12A. The yield was 93% (light yellow syrup) for the final step. Rf (hexanes/AcOEt 9:1) 0.26. 1H
NMR (CDCI3) δ 0.88 (t, 6H, J = 6.7 Hz), 1.15-1.34 (m, 48H)
1.51-1.60 (m, 4H), 1.98 (s, 3H, 5-CH3), 2.29 (ddd, 1H, J =
14.5, 8.3, 4.3 Hz, 3'-Ha), 2.62 (dd, 1H, J = 14.5, 6.2 Hz, 3'-Hb), 3.41-3.86 (m, 9H), 4.73 (s, 2H), 5.25 (s, 1H, 5'-H),
5.46 (d, 1H, J = 4.3 Hz, 4'-H), 5.52 (s, 2H), 6.78 (dd, 1H,
J = 8.3, 6.3 Hz, 2'-H), 7.25-7.65 (m, 9H), 8.02-8.07 (m,
2H). 13C NMR (CDCI3) δ 13.21, 14.18, 22.75, 26.17, 29.42,
29.72, 30.10, 31.97, 35.17 (3'-C), 68.72, 70.18, 70.67, 70.93, 71.85, 72.25, 77.52 (4'-C), 86.52 (2'-C), 106.36 (5'- C), 111.53 (5-C), 127.64, 128.30, 128.57, 129.31, 129.85, 133.66, 134.33 (6-C), 138.10, 151.39 (2-C), 163.30 (4-C), 165.52 (benzoyl C=O). Anal Calcd for C55H86N2O9 : C, 71.90; H, 9.37; N, 3.05. Found: C, 71.61; H, 9.24; N, 2.95. EXAMPLE 12C
(2'R,4'S,5'S)-4'-Benzoxy-5'-O-(diethoxyphosphinylmethoxy)- N3-(benzoxymethyl)thymine (12g)
The title compound was prepared using diethoxyphosphinylhydroxymethyl (prepared according to the procedure of Phillion, D.P., et.al., Tetrahedron Lett., 1986, 21, 1477) according to the procedure of Example 12A. The yield was 82% (colorless syrup). Rf (CH2Cl2/AcOEt 3:1) 0.24. 1H ΝMR (CDCl3) δ 1.33 (t, 3H, J = 7.0 Hz), 1.39 (t, 3H, J = 7.0 Hz), 2.03 (s, 3H, 5-CH3), 2.36 (ddd, 1H, J = 14.8, 8.4, 4.9 Hz, 3'-Ha), 2.63 (dd, 1H, J = 14.8, 6.3 Hz, 3'-Hb), 3.84 (dd, H, 2Jgem = 13.3, 2JP-H = 10.0 Hz, P-CHH-O), 4.02 (dd, H, 2Jgem = 13.3, 2JP-H = 10.0 Hz, P-CHH-O), 4.18 (qd, 2H, 3JH-H = 7.0 Hz, 3JP-H = 0.8 HZ, CH3CH2-O-P), 4.26 (qd, 2H, 3JH-H = 7.0 Hz, 3JP-H = 0.8 Hz, CH3CH2-O-P), 4.71 (s, 2H), 5.23 (s, 1H, 5'-H), 5.52 (s, 2H), 5.53 (d, 1H, J = 4.9 Hz, 4'-H), 6.86 (dd, 1H, J = 8.4, 6.3 Hz, 2'-H), 7.27-7.66 (m,
9H), 8.02-8.06 (m, 2H). 13C ΝMR (CDCl3) δ 13.15, 16.72 (d, 3JP-C = 5.5 Hz, O-C-C), 34.67 (3'-C), 61.51 (d, 1JP-C = 135.5 Hz, P-C-0), 63.28 (d, 2JP-C = 28.9 Hz, O-C-C), 70.93, 72.40, 77.41 (4'-C), 86.71 (2'-C), 107.02 (d, 3JP-C = 12.8 Hz, 5'- C), 112.29 (5-C), 127.84, 128.42, 128.85, 129.09, 129.97, 133.91, 138.21, 151.59 (2-C), 163.34 (4-C), 165.63 (benzoyl C=O). 31P ΝMR (CDCl3) δ 20.45. Anal Calcd for C29H35Ν2O10P- 0.3 CHCl 3: C, 55.06; H, 5.53; N, 4.39; P, 4.86. Found: C, 54.95; H, 5.58; N, 4.28; P, 4.65.
EXAMPLE 12D
(2'R,4'S,5'S) -4'-Benzoxy-5'-O- (benzyloxy) -N3- (benzoxymethyl)thymine (12h)
The title compound was prepared using benzyl alcohol according to the procedure of Example 12A. The yield was 90%.
EXAMPLE 12E
(2'R,4'S,5'S)-4'-Benzoxy-5'-O-(cyclohexyloxy)-N3- (benzoxymethyl)thymine (121)
The title compound was prepared using cyclohexanol according to the .procedure of Example 12A. The yield was
93% (white solid), mp 141-3 ºC. Rf (CH2Cl2/AcOEt 1:1) 0.58.
1H NMR (CDCl3) δ 1.26-1.60 (m, 6H), 1.69-2.04 (m, 4H), 1.96
(s, 3H, 5-CH3), 2.31 (ddd, 1H, J = 14.7, 7.9, 4.8 Hz, 3'- Ha), 2.63 (dd, 1H, J = 14.7, 6.5 Hz, 3'-Hb), 3.73 (m, 1H), 4.72 (s, 2H), 5.36 (s, 1H, 5'-H), 5.38 (d, 1H, J = 4.8 Hz, 4'-H), 5.52 (s, 2H), 6.75 (dd, 1H, J = 7.9, 6.5 Hz, 2'-H), 7.22-7.65 (m, 9H), 8.02-8.06 (m, 2H). 13C NMR (CDCl3) δ 13.25, 23.74, 23.96, 25.53, 31.83, 33.29. 35.29 (3'-C), 70.69, 72.26, 77.21, 78.26 (4'-C), 86.31 (2'-C), 104.58
(5'-C), 110.88 (5-C), 127.65, 128.31, 128.53, 129.20,
129.84, 133.66, 134.81 (6-C), 138.11, 150.41 (2-C), 163.33
(4-C), 165.74 (benzoyl C=O). Anal Calcd for C30H34N2O7 0.5 H2O: C, 66.30; H, 6.45; N, 5.16. Found: C, 66.58; H, 6.44; N, 5.09.
EXAMPLE 12F
(2'R,4'S,5'S)-4'-Benzoxy-5'-0-(1-adamantyloxy)-N3-(benzoxymethyl)thymine (12j)
The title compound was prepared using 1-adamantyl alcohol according to the procedure of Example 12A. The yield was 86% (white solid). mp 98-100 ºC.
Rf (hexanes/AcOEt 1:1) 0.60. 1H NMR (CDCl3) δ 1.50-1.95 (m,
12H), 1.98 (s, 3H, 5-CH3), 2.05-2.25 (m, 3H), 2.30 (ddd, 1H,
J = 14.5, 7.9, 4.5 Hz, 3'-Ha), 2.63 (dd, 1H, J = 14.5, 6.5 Hz, 3'-Hb), 4.73 (s, 2H), 5.27 (d, 1H, J = 4.5 Hz, 4"-H),
5.52 (s, 2H), 5.61 (s, 1H, 5'-H), 6.71 (dd, 1H, J = 7.9, 6.5
Hz, 2'-H), 7.20-7.65 (m, 8H), 7.78 (s, 1H, 6-H), 8.08 (d,
2H, J = 7.6 Hz). 13C NMR (CDCl3) δ 13.37, 30.58, 35.42 (3'-
C), 42.66, 45.33, 70.65, 72.20, 75.96, 79.14 (4'-C), 86.31 (2'-C), 99.48 (5'-C), 110.54 (5-C), 127.61, 128.28, 129.25, 129.83, 133.61, 135.36 (6-C), 138.14, 151.37 (2-C), 163.36 (4-C), 165.77 (benzoyl C=O). Anal Calcd for C34H38N2O7 0.4 CHCl 3: C, 65.13; H, 6.06; N, 4.42. Found: C, 64.97; H, 6.34; N, 4.30. EXAMPLE 12G
(2'R,4'S,5'S)-4'-Benzoxy-5'-O-(2-bromoethoxy)-N3- (benzoxymethyl)thymine (12k)
The title compound was prepared using 2-bromoethanol according to the procedure of Example 12A. The yield was 91% (white solid), mp 84-6 ºC. Rf (hexanes/AcOEt 3:1) 0.32. 1H NMR (CDCl3) δ 1.99 (s, 3H, 5-CH3), 2.35 (ddd, 1H, J = 14.8, 8.4, 4.9 Hz, 3'-Ha), 2.63 (dd, 1H, J = 14.8, 6.4 Hz, 3'-Hb), 3.57 (t, 2H, J = 5.5 Hz, BrCH2CH2O), 3.99 (dt, 1H, J = 16.4, 5.5 Hz, BrCH2CHHO), 4.10 (dt, 1H, J = 16.4, 5.5 Hz, BrCH2CHHO), 4.72 (s, 2H), 5.25 (s, 1H, 5'-H), 5.49 (d, 1H,
J = 4.9 Hz, 4'-H), 5.52 (s, 2H), 6.82 (dd, 1H, J = 8.4, 6.4
Hz, 2'-H), 7.27-7.66 (m, 9H), 8.02-8.06 (m, 2H). 13C NMR
(CDCl3) δ 13.42 (5-CH3), 30.26 (Br-C-C-O), 34.73 (3'-C),
68.87 (Br-C-C-O), 70.76, 72.28, 77.48 (4'-C), 86.46 (2'-C), 106.40 (5'-C), 111.63 (5-C), 127.68, 128.34 (6-C), 128.66, 128.96, 129.86, 133.83, 134.94, 138.05, 151.42 (2-C), 163.24 (4-C), 165.65 (benzoyl C=O). Anal Calcd for C26H27N2O7Br : C, 55.82; H, 4.83; N, 5.01; Br, 14.30. Found: C, 55.78; H, 4.85; N, 4.76, Br, 14.43. EXAMPLE 12H
(2'R,4'S,5'S)-4'-Benzoxy-5'-O-(2-propenyl-1-oxy)-N3- (benzoxymethyl)thymine (121)
The title compound was prepared using allyl alcohol according to the procedure of Example 12A. The yield was 84% (white solid), mp 112-3 ºC. Rf (hexanes/AcOEt 1:1) 0.60. 1H NMR (CDCl3) δ 1.93 (s, 3H, 5-CH3), 2.33 (ddd, 1 H, J = 14.5, 6.8, 4.7 Hz, 3'-Ha), 2.64 (dd, 1H, J = 14.5, 8.1 Hz, 3'-Hb), 4.17 (dd, 1H, J = 12.6, 4.1, 1.0 Hz, CHH-CH=CH2), 4.29 (ddd, 1H, J = 12.6, 5.8, 1.0 Hz, CHH-CH=CH2), 4.72 (s, 2H), 5.24 (s, 1H, 5'-H), 5.30 (ddd, 1H, J = 10.2, 1.3, 1.0 Hz, CH2-CH=CHHtrans), 5.30 (ddd, 1H, J = 17.3, 1.5, 1.0 Hz,
CH2-CH=CHHCiS) , 5.47 (d, J = 4.7 Hz, 4'-H), 5.51 (s, 2H),
5.97 (dddd, 1H, J = 17.3, 10.2, 5.8, 4.1 Hz, CH2-CH=CH2),
6.82 (dd, 1H, J = 8.1, 6.8 Hz, 2'-H), 7.18-7.65 (m, 9H), 8.03-8.06 (m, 2H). 13C NMR (CDCl3) δ 13.32 (5-CH3), 35.18
(3'-C), 69.10 (C=C-C), 70.76, 72.29, 77.59 (4'-C), 86.41
(2'-C), 105.52 (5'-C), 111.43 (5-C), 118.29 (C=C-C), 127.62,
128.31 (6-C), 128.60, 129.12, 129.84, 132.94 (C=C-C),
133.70, 134.21, 138.10, 151.43 (2-C), 163.22 (4-C), 165.60 (benzoyl C=O). Anal Calcd for C28H2BN2O7: C, 68.29; H, 5.69;
N, 5.69. Found: C, 68.76; H, 5.63; N, 5.60.
EXAMPLE 121
(2'R,4'S,5'S) -3-Benzoxymethyl-(4'-benzoxy-5' - azidotetrahydrofuran-2'-yl)thymidine
The title compound was prepared using TMSN3 according to the procedure of Example 12A. The yield was 78%. Rf(hexanes/AcOEt 1:1) 0.36. 1H NMR (CDCl3) δ 1.50-1.95 (m, 12H), 1.98 (s, 3H, 5-CH3), 2.27 (ddd, 1H, J = 14.8, 8.7, 4.9 Hz, 3'-Ha), 2.66 (dd, 1H, J = 14.8, 6.0 Hz, 3' -Hb), 4.70 (s, 2H), 5.37 (d, 1H, J = 4.9 Hz, 4'-H), 5.51 (s, 2H), 5.60 (s,
1H, 5'-H), 6.74 (dd, 1H, J = 8.7, 6.0 Hz, 2'-H), 7.20-7.65
(m, 8H), 7.78 (s, 1H, 6-H), 8.08 (d, 2H, J = 7.6 Hz). 13C
NMR (CDCl3) δ 13.47, 34.71 (3'-C), 70.80, 72.33, 77.60 (4'-
C), 87.14 (2'-C), 95.07 (5'-C), 111.74 (5-C), 127.61, 127.76, 127.78, 128.31, 128.69, 129.88, 133.29, 133.94 (6- C), 138.04, 151.23 (2-C), 163.04 (4-C), 165.51 (benzoyl C=O). Anal Calcd for C24H23N5O6· 0.45 CHCl 3 : C, 55.28; H, 4.42; N, 13.19. Found: C, 55.38; H, 4.39; N, 13.00.
EXAMPLE 13
(2'R,4'S,5'S)-4'-Hydroxy-5'-O-((2-(2-aminoethoxy)ethyl)-tetr ahydrofuran-2'-y1)thymine (15)
A suspension of 12.4 g (18.5 mmol) of 14 and 7.6 g of 10% Pd(OH)2/C in 100 mL of1 : 1 mixture of acetone/MeOH was stirred under an H2 atmosphere for 16 hours. The suspended material was filtered through a pad of Celite and the filterate was concentrated at reduced pressure to give 7.8 g (76.7%) of de-BOM product as a white form. The de-BOM product was re-dissolved in 100 mL MeOH and 1.54 g (48.0 mmol) of H2NNH2 was added. The mixture was heated at reflux for 16 hours and then concentrated at reduced pressure. The resultant crude, free amine product ( 8 g) was redissolved in 100 mL MeOH, and 4.0 g (100.0 mmol) of NaOH was then added. The mixture was stirred at 45 °C for 4 hours, and the solvent was then removed at reduced pressure. The residue was purified by SiO2 column chromatography (2% Et3N in
CHCl3/MeOH 1:1) to give 2.2 g (37.7% from 14) of 15 as a white solid. Rf (2% Et3N in CHCl3/MeOH 1:1) 0.21. 1H NMR
(CD3OD) δ 1.93 (s, 3H, 5-CH3), 2.27 (m, 2H, 3'-H), 2.82 (m, 2H), 3.58-3.93 (m, 6H), 4.30 (d, 1H, J= 4.5 Hz, 4'-H), 5.05 (s, 1H, 5'-H), 6.62 (t, 1H, J = 8.1 Hz, 2'-H), 7.58 (s, 1H, 6-H). 13C NMR (CD3OD) δ 12.80 (5-CH3), 38.33 (3'-C), 51.23, 68.77, 70.00, 71.31, 76.18 (4'-C), 87.29 (2'-C), 110.77 (5'- C), 112.37 (5-C), 137.84 (6-C), 152.75 (2-C), 166.31 (4-C). EXAMPLE 14
(2'R,4'S,5'S)-(4'-Hydroxy-5'-O-((2-aminoethoxy)ethyl)-tetrah ydrofuran-2'-yl)thymine, cholesterylforamide (16)
A stirred solution of 2.1 g (6.8 mmol) of 15 in 20 mL of 1:1 CH2Cl2/pyridine was added a solution of 3.3 g (7.5 mmol) of cholesteryl chloroformate in 20 mL CH2Cl2 at 0 °C over 45 min period. The resultant mixture was stirred for additional 3 hours at 0 °C and then diluted with 200 mL CH2Cl2, washed with sat. aqueous NaHCO3 (50 mL) and brine (50 mL). The organic layer was dried (MgSO4) and concentrated at reduced pressure. The residue was purified by SiO2 column chromatography (CH2Cl2/Ac0Et 9:1 then AcOEt) to give 2.2 g (45.4%) of 16 as a white solid. Rf (AcOEt) 0.44. 1H NMR (DMSO-d6, assignment for some characteristic proton resonances) : δ 4.93 (s, 1H, 5'-H), 5.33 (d, 1H, J = 3.8 Hz, 4'-OH), 5.45 (d, 1H, J = 3.8 Hz, 4'-H), 6.48 (t, 1H, J = 8.2 Hz, 2'-H), 8.37 (s, 1H, 6-H), 11.38 (s, 1H, NH). 13CC NMR (DMSO-d6) d 11.65, 12.18, 18.55, 18.98, 22.37, 22.61, 23.38, 23.89, 27.43, 27.83, 31.42, 34.72, 34.87, 35.30, 35,78, 36.10, 41.89, (3'-C), 47.38, 49.56, 55.74, 56.20, 66.82, 69.15, 73.01, 73.88, 74.11 (4'-C), 79.20, 85.09 (2'- C), 108.61 (5'-C), 110.34 (5-C), 139.73 (6-C), 150.67 (2-C), 154.99 (formamide C=O), 163.55 (4-C).
EXAMPLE 15
(2'R,4'S,5'S)-(4'-O-(2-Cyanoethoxy-N,N-diisopropylamino-phos phorityl)-5'-O-((2-aminoethoxy)ethyl)-tetrahydrofuran-2'-yl) thymine, cholesterylforamide (17)
A solution of 1.7 g (2.3 mmol) of 16 and 1.4 g (1.9 mL, 11.0 mmol) of i-Pr2NEt in 40 mL CH2Cl2 was treated with 1.1 g (1.0 mL, 4.6 mmol) of 2-cyanoethyl N, N-diisopropyl- chlorophosphoramidite at room temperature for 24 hours. The resultant mixture was diluted with 100 mL CH2Cl2 and washed with sat. aqueous ΝaHCO3 (30 mL) and brine (30 mL). The organic layer was dried (MgSO4) and concentrated at reduced pressure. The residue was purified by SiO2 column chromatography (0.5% Et3N in hexanes/AcOEt 1:1 then AcOEt) to give 0.36 g (16.6%) of 17 as a white foam. 31P NMR (CDCl3) δ 149.23, 149.43 ppm.
A summary of the synthesis of compounds 8 through 17 are presented on the following pages.
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
EXAMPLE 16
N3-Benzoxymethyl-(4'-O-benzoyl-5'-O-adamantyl-tetrahydro- furan-2'-yl)thymine (18)
To a solution of 8 (3.5 g, 7.1 mmol) and 1-adamantanol (2.2 g, 14.2 mmol) in CH2Cl2 (100 ml) was added TMSOTf (2.7 ml, 3.2 g, 14.2 mmol) at -23 °C in one portion via a syringe. The resultant mixture was stirred at -23 °C for 0.5 hr and then placed in a freezer (-15 °C) for 18 hr. The reaction mixture was then poured into a 40:10:1 bilayer solution of AcOEt/H2O/Et3N (500 ml), the organic layer dried over MgSO4 and concentrated at reduced pressure. The residue was purified by SiO2 column chromatography (hexanes/AcOEt 4:1 then 2:1) to give 3.6 g (86.7%) of the title compound as a white solid. Rf (hexanes/AcOEt 1:1) 0.60. 1H NMR (CDCl3) δ 1.50-1.95 (m, 12 H, adamantyl-H), 1.98 (s, 3H, 5-CH3), 2.05-2.25 (M, 4H, adamantyl-H), 2.3 (ddd, 1H, J = 14.5, 7.9, 4.5 Hz, 3' -HJ, 2.63 (DD, 1H, J = 14.5, 6.5 Hz, 2'-Hß), 4.73 (s, 2H, 0CH2Ph) , 5.27 (d, 1H, J = 4.5 Hz, 4'-H), 5.52 (s, 2H, NCH2O), 5.61 (s, 1H, 5'-H), 6.71 (dd, 1H, J = 7.9, 6.5Hz, 2'-H), 7.2-7.65 (m, 8H, aromatic-H), 7.78 (s, 1H, 6-H), 8.08 (d, 2H, J = 7.6 Hz, aromatic-H). 13C-NMR (CDCl3) δ 13.37, 30.58, 35.42 (3'-C), 42.66, 45.33, 70.65, 72.20, 75.96, 79.14 (4'-C) 86.31 (2'- C), 99.48 (5'-C), 110.54 (5-C), 127.61, 128.28, 129.25, 129.83, 133.61, 135.36 (6-C), 138.14, 151.37 (2-C), 163.36 (4-C), 165.77 (benzoyl C=O).
EXAMPLE 17
Preparation of Oligonucleotides
Oligonucleotides are prepared utilizing normal protocol for solid state DNA synthesis, (see Oligonucleotide synthesis, a practice approach, Ed. M.J. Gait, IRL Press 1984, Oxford University Press, New York). DMT protected phosphoramidite nucleotides are added to the growing oligonucleotide structure on a solid state support in the normal manner. Coupling efficiencies are typically greater than 96% for each step of the synthesis with the overall coupling efficiency greater than 91% for the oligomer. The resulting oligomers are characterized by both gel chromatography and by HPLC using standard protocols.
EXAMPLE 18
Preparation of Oligonucleotides Containing 5' Terminal Nucleotides Having 4'-Desmethyl Structure Bearing A Conjugation Molecule
An oligonucleotide of the desired sequence is prepared as per the preceding Example 17 in the normal 3' to 5' direction. The last nucleotide of the sequence is preformed to include the desired 4' -desmethyl structure bearing a conjugate group, e.g. compounds 12 and 17, attached via linker groups and compound 18, directly attached, thereon as per the above Examples 10, 15 and 17. This last nucleotide unit, as a 3' -phosphoamidite, is coupled to the remainder of the oligonucleotide sequence utilizing normal protocol for solid state DNA synthesis, as above. As with the remainder of the sequences, coupling efficiencies are generally greater than 96% for this step of the synthesis.
A typical synthesis was that of an oligonucleotide of the sequence:
T*GC ATC CCC CAG GCC ACC AT, SEQ ID NO. 1, where T* is the nucleotide of compound 12, compound 17 or compound 18 above. The sequences was synthesizer up to and including the penultimate nucleotide unit. The conjugate group bearing, 4-desmethyl, terminal nucleotide, e.g. compound 12, was then used as the ultimate nucleotide unit. The oligonucleotide was removed from the solid support and purified by HPLC in the standard manner. EXAMPLE 18
Assay for Separating Expression of Intercellular Adhesion Molecule From Vascular Adhesion Molecules Using Selective Protein Inhibition
Various adhesion molecules are known that are associated with certain adverse events in biological systems. The study of these molecules can be compounded by overlapping activities. Since the intercellular adhesion molecule, ICAM-1, and the vascular adhesion molecule, VCAM- 1, both are capable of expressing certain similar properties, e.g. cell adhesion or clumping, segregation of certain of their properties, one from the other, would assist in the evaluation of the properties of a singular adhesion molecule. This cellular assay isolates cytokine induced protein induction of ICAM-1 from that of VCAM-1. The assay differentiates specific protein inhibition of ICAM-1 expression from VCAM-1 expression via use of a probe oligonucleotide having a sequence complementary to the ICAM- 1 messenger RNA. Inhibition of the induction of ICAM-1 messenger RΝA is effected by treatment of the cells with the probe oligonucleotide in presence of a cationic liposome formulation (DOTMA/DOPE) . Cells showing positive adhesion molecule expression in response to cytokine treatment are identified by increases in basal levels of the adhesion molecule protein after treatment with a combination of human TNF-or and murine IΝF-T using a fluorescence activated cell sorter to identify the cells stained with fluorescent antibodies to either the ICAM-1 or VCAM-1 expressed proteins.
The oligonucleotide utilized as the probe was an oligonucleotide of the invention of the sequence T*GC ATC CCC CAG GCC ACC AT, SEQ ID NO. 1, complementary to the 3' untranslated region of murine ICAM-1 messenger RNA. It includes a conjugation PEG molecule (compound 12 above) as the 5' terminal T nucleotide (nucleotide T* in the above sequence). This oligonucleotide is then used to differentiate ICAM-1 expression from VCAM-1 expression in the test protocol. In cell lines from other species, the base sequence of conjugated oligonucleotide of the invention used in the test is synthesized to be of a sequence complementary to a portion of the ICAM-1 messenger RNA for the species of interest.
A murine endothelioma cell line designated bEND.3, supplied by Dr. Werner Risau, Max-Planck-Institute, Planegg- Martinsreid, Germany, is tested for adhesion molecule expression as follows. The cells were treated with nanomolar concentrations of the conjugated oligonucleotide, SEQ ID NO. 1, in the presence of 15 μg/ml DOTMA/DOPE liposome (Lipofectin) for 4 hours in a serum free media (Opti-MEM serum-free medium, GIBCO, Grand Island, NY) in the manner described by Chiang et al., J. Biol. Chem., 1991, 266, 18162-18171 or Bennett, et al., J. Immunol, 1994, 152, 3530-3540. The media was aspirate and adhesion molecule expression was induced with human TNF-α (5 ng/ml, R&D Systems, Minneapolis, MN) and murine IFN-τ (1000 μ/ml, Genzyme, Cambridge, MA) overnight in DMEM high glucose with 10% fetal bovine serum (Hyclone, Logan, UT). The cells were trypsinized and washed. The cells were stained with ICAM-1- PE fluorescent antibodies (anti- ICAM-1 mAB, PharMingen, San Diego, CA) and VCAM-1-FITC fluorescent antibodies (anti- VCAM-1 mAB, PharMingen, San Diego, CA) and read on a fluorescence activated cell sorter.
Concurrent treatment of the same bEND.3 cell line absent the conjugated oligonucleotide served as a positive control. Both ICAM-1 and VCAM-1 induction were observed in the positive control and normalized to 100%. The conjugated oligonucleotide was used at 0.1, 0.3 and 0.5 uM and the levels of both of induced expression ICAM-1 and VCAM-1 were compared to this positive control. Inhibition of VCAM-1 expression was observed to be 90.08%, 103.82% and 96.33% of control for the 0.1, 0.3 and 0.5 uM concentrations, respectively whereas inhibition of ICAM-1 expression was 3.77%, 4.40% and 8.74% of control for the 0.1, 0.3 and 0.5 uM concentrations, respectively.
As these results show, in the murine bEND.3 cell line both ICAM-1 and VCAM-1 expression were induced in the positive control. However, in the presence of the ICAM-1 specific messenger RNA complementary oligonucleotide of the invention, VCAM-1 protein expression was maintained but ICAM-1 protein expression was inhibited. Thus the expression of VCAM-1 is disconnect from that of ICAM-1 such that VCAM-1 expression can be examined independent of ICAM-1 expression.

Claims

WHAT IS CLAIMED IS:
1. A compound of structure:
Figure imgf000066_0001
wherein:
RL is azido or a group
RC-[Y]e-Z- wherein:
Z is 0, S or HN;
Y is a bivalent linker;
e is 0 or 1;
RC is alkyl, alkenyl, alkynyl, aryl, alkaryl, aralkyl, phosphinyl, a polyglycol, a polyamine, a polyether, a polythioether, an aromati or nonaromatic ring system, a steroid molecule, a reporter molecule, an aromatic lipophilic molecule, a non-aromatic lipophilic molecule, a reporter enzyme, a peptide, a protein, a water soluble vitamin, a lipid soluble vitamin, a carbohydrate, a terpene molecule, a phospholipid, an intercalator, a cell receptor binding molecule, a crosslinking agent, or a porphyrin;
Bx is a nucleobase;
X is H, OH, O-alkyl, O-alkoxylalkyl, O-alkylamino, or F;
Q is O, S, CH2, CHF or CF2; and
RD is H, hydroxyl, an activated phosphorous group, a nucleoside, an activated nucleotide, a nucleotide, an oligonucleotide, an oligonucleoside or a protected derivative thereof.
2. A compound having structure:
Figure imgf000067_0001
wherein:
L is a group of formula:
R7-[S-(CH2)q]u-[HN-(CH2)n]t-[O-(CH2)m]v-Z- wherein:
t, u and v are each independently integers from 0 to
200,
m, n and q are each independently integers from 1 to
Z is S, O or HN;
R7 is RC, H, or a protecting group;
Bx is a nucleobase;
X is H, OH, O-alkyl, O-alkoxylalkyl, O-alkylamino, or Q is O;
RD is H, hydroxyl, an activated phosphorous group, a nucleoside, an activated nucleotide, a nucleotide, an oligonucleotide, an oligonucleoside or a protected protected derivative thereof;
RC is alkyl, alkenyl, alkynyl, aryl, Alkaryl, aralkyl, phosphinyl, a polyglycol, a polyamine, a polyether, a polythioether, an aromatic or nonaromatic ring system, a steroid molecule, a reporter molecule, an aromatic lipophilic molecule, a non-aromatic lipophilic molecule, a reporter enzyme, a peptide, a protein, a water soluble vitamin, a lipid soluble vitamin, a carbohydrate, a terpene molecule, a phospholipid, an intercalator, a cell receptor binding molecule, a crosslinking agent, or a porphyrin; and provided that t, u and v are not all simultaneously 0.
3. A compound of claim 2 wherein t and u are 0.
4. A compound of claim 2 wherein t and u are 0, and m is 2.
5. A comound of claim 2 wherein u and v are 0.
6. A compound of claim 2 wherein u and v are 0 and n is 2.
7. A compound of claim 2 wherein X is H or OH.
8. A compound of claim 2 wherein RD is H or OH.
9. A compound of claim 2 wherein RD is an activated phosphorous group.
10. A compound of claim 2 wherein RD is an oligonucleotide.
11. A comound of claim 2 wherein t and v are 0.
12. A compound of claim 2 wherein t and v are 0 and q is 2.
13. A compound of claim 2 wherein Z is O or N.
14. A compound of claim 2 wherein RC is a steroid molecule.
15. A compound of claim 14 wherein the steroid molecule is cholic acid, deoxycholic acid, dehydrocholic acid, cortisone, digoxigenin, testosterone, cholesterol or 3 -trimethylaminomethylhydrazido cortisone.
16. A compound of claim 2 wherein RC is a water soluble vitamin.
17. A compound of claim 16 wherein the water soluble vitamin is thiamine, riboflavin, nicotinic acid, pyridoxal phosphate, pyridoxine, pyridoxamine, deoxypyridoxine, pantothenic acid, biotin, folic acid, 5' -deoxyadenosyl- cobalamin, inositol, choline or ascorbic acid.
18. A compound of claim 2 wherein RC is a lipid soluble vitamin.
19. A compound of claim 18 wherein the lipid soluble vitamin is a retinal, a retinol, retinoic acid, β-carotene, vitamin D, cholecalciferol, a tocopherol, or a phytol.
20. A compound of claim 2 wherein RC is a protein.
21. A compound of claim 20 wherein the protein is a phosphodiesterase, a peroxidase, a phosphatase or a nuclease.
22. A compound of claim 2 wherein RC is a reporter molecule.
23. A compound of claim 22 wherein the reporter molecule is a chromaphore, a fluorophore or a radiolabel- containing moiety.
24. A compound of claim 23 wherein the fluorophore is fluorescein, chrysine, anthracene, perylene, pyrene, rhodamine.
25. A compound of claim 2 wherein X is F, O-alkyl having from one to six carbons, O-alkoxyalkyl having from 2 to 6 carbons, or O-alkylamino having from one to six carbons.
26. A compound of claim 2 wherein RD is H or OH.
27. A compound having structure:
Figure imgf000070_0001
wherein:
RC is O-alkyl, O-alkenyl, O-alkynyl, O-aryl, O- alkaryl, O-aralkyl, a polyglycol, an aromatic or nonaromatic ring system, a polyamine, a polyether, a steroid molecule, a reporter molecule, an aromatic lipophilic molecule, a nonaromatic lipophilic molecule, a reporter enzyme, a peptide, a protein, a water soluble vitamin, a lipid soluble vitamin, a carbohydrate, a terpene molecule, a phospholipid, an intercalator, a cell receptor binding molecule, a crosslinking agent, or a porphyrin;
Bx is a nucleobase;
X is H, OH, O-alkyl, O-alkoxylalkyl, O-alkylamino, or
F;
Q is O, S, CH2, CHF or CF2; and
RD is H, hydroxyl, an activated phosphorous group, a nucleoside, an activated nucleotide, a nucleotide, an oligonucleotide, an oligonucleoside or a protected protected derivative thereof.
28. A compound of claim 27 wherein RC is a polyether, a polyamine or a non-aromatic lipophilic molecule.
29. A compound of structure;
Figure imgf000071_0001
wherein:
J is a leaving group;
Bx is a nucleobase;
X is H, OH, O-alkyl, O-alkoxylalkyl, O-alkylamino, or F;
Q is 0, S, CH2, CHF or CF2; and
RD is H, hydroxyl, an activated phosphorous group, a nucleoside, an activated nucleotide, a nucleotide, an oligonucleotide, an oligonucleoside or a protected derivative thereof.
30. A compound of claim 29 wherein J is OH, SH, NH2, trifluoromethylsulfonyl, methylsulfonyl, halogen, O- trichloroacetimidate, acyloxy, dialkyl phosphite, 2,4,6- trichlorophenyl, p-toluenesulfonyl, 4-dimethylaminoazo- benzenesulfonyl or 5-dimethylaminonaphthalenesulfonyl.
31. A method for forming a 5' -desmethyl conjugate oligomer having structure:
Figure imgf000072_0001
comprising the steps of:
(a) providing a first synthon having structure
Figure imgf000072_0002
and
(b) contacting the first synthon with a second synthon having structure:
Figure imgf000072_0003
the contacting being for a time and under reaction conditions sufficient to form the conjugated oligomer; wherein:
RL is azido or a group of formula:
RC-[Y]e-Z- Z is O, S or HN;
Y is a bivalent linker;
e is 0 or 1;
RC is alkyl,. alkenyl, alkynyl, aryl, alkaryl, aralkyl, phosphinyl, a polyglycol, a polyamine, a polyether, a polythioether, an aromati or nonaromatic ring system, a steroid molecule, a reporter molecule, an aromatic lipophilic molecule, a non-aromatic lipophilic molecule, a reporter enzyme, a peptide, a protein, a water soluble vitamin, a lipid soluble vitamin, a carbohydrate, a terpene molecule, a phospholipid, an intercalator, a cell receptor binding molecule, a crosslinking agent, or a porphyrin;
E is a phosphodiester, phosphorothioate, phosphorodithioate, phosphamidate, alkyl phosphonate or phosphotriester phosphorous linking group;
RE is an activated phosphorous group;
W is H, a hydroxyl protecting group or a solid state support;
k is an integer from 0 to 50;
Bx is a nucleobase;
X is H, OH, O-alkyl, O-alkoxylalkyl, O-alkylamino, or F;
Q is O, S, CH2, CHF or CF2; and
RD is H, hydroxyl, an activated phosphorous group, a nucleoside, an activated nucleotide, a nucleotide, an oligonucleotide, an oligonucleoside or a protected derivative thereof.
32. A method for forming a 5'-desmethyl conjugated monomer having structure:
Figure imgf000074_0001
comprising the steps of:
(a) providing a synthon having structure
Figure imgf000074_0002
and
(b) contacting this synthon with an activated conjugating group;
the contacting being for a time and under reaction conditions sufficient to form the conjugated monomer;
wherein:
RL is a group of formula:
RC-[Y]e-Z-
Z is O, S or HN;
Y is a bivalent linker;
e is 0 or 1;
RC is alkyl, alkenyl, alkynyl, aryl, alkaryl, aralkyl, phosphinyl, a polyglycol, a polyamine, a polyether, a polythioether, an aromati or nonaromatic ring system, a steroid molecule, a reporter molecule, an aromatic lipophilic molecule, a non-aromatic lipophilic molecule, a reporter enzyme, a peptide, a protein, a water soluble vitamin, a lipid soluble vitamin, a carbohydrate, a terpene molecule, a phospholipid, an intercalator, a cell receptor binding molecule, a crosslinking agent, or a porphyrin;
J is a leaving group;
Bx is a nucleobase;
X is H, OH, O-alkyl, O-alkoxylalkyl, O-alkylamino, or F;
Q is O, S, CH2, CHF or CF2; and
RD is H, hydroxyl, an activated phosphorous group, a nucleoside, an activated nucleotide, a nucleotide, an oligonucleotide, an oligonucleoside or a protected derivative thereof.
33. The method of claim 32 wherein J is OH, SH, NH2, trifluoromethylsulfonyl, methylsulfonyl, halogen, O- trichloroacetimidate, acyloxy, dialkyl phosphite, 2,4,6- trichlorophenyl, p-toluenesulfonyl, 4-dimethylaminoazo- benzenesulfonyl or 5-dimethylaminonaphthalenesulfonyl.
34. The method of claim 32 wherein J is trifluoromethylsulfonyl, methylsulfonyl, halogen or acyloxy.
PCT/US1995/013038 1994-09-29 1995-09-29 Oligomers having heteroatom-linked conjugate groups WO1996010030A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US08/809,239 US5998603A (en) 1994-09-29 1995-09-29 4'-desmethyl nucleoside analogs, and oligomers thereof
AU38923/95A AU3892395A (en) 1994-09-29 1995-09-29 Oligomers having heteroatom-linked conjugate groups

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/314,877 1994-09-29
US08/314,877 US5608046A (en) 1990-07-27 1994-09-29 Conjugated 4'-desmethyl nucleoside analog compounds

Publications (1)

Publication Number Publication Date
WO1996010030A1 true WO1996010030A1 (en) 1996-04-04

Family

ID=23221863

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/013038 WO1996010030A1 (en) 1994-09-29 1995-09-29 Oligomers having heteroatom-linked conjugate groups

Country Status (3)

Country Link
US (1) US5608046A (en)
AU (1) AU3892395A (en)
WO (1) WO1996010030A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001010882A1 (en) * 1999-08-10 2001-02-15 Adani, Alexander Depot forms of modified nucleoside 5'-hydrogenphosphates as inhibitors of reproduction of human immunodeficiency virus and human hepatitis b virus
WO2001034622A1 (en) * 1999-11-08 2001-05-17 Origenix Technologies, Inc. Combinatorial library synthesis and pharmaceutically active compounds produced thereby
US6245750B1 (en) * 1998-01-23 2001-06-12 Newbiotics, Inc. Enzyme catalyzed therapeutic agents
US6495553B1 (en) 1997-08-08 2002-12-17 Newbiotics, Inc. Methods and compositions for overcoming resistance to biologic and chemotherapy
US6683061B1 (en) 1999-07-22 2004-01-27 Newbiotics, Inc. Enzyme catalyzed therapeutic activation
US7138388B2 (en) 2001-01-19 2006-11-21 Celmed Oncology (Usa), Inc. Methods to treat autoimmune and inflammatory conditions
US7256179B2 (en) 2001-05-16 2007-08-14 Migenix, Inc. Nucleic acid-based compounds and methods of use thereof
US7462605B2 (en) 1998-01-23 2008-12-09 Celmed Oncology (Usa), Inc. Phosphoramidate compounds and methods of use

Families Citing this family (962)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6753423B1 (en) 1990-01-11 2004-06-22 Isis Pharmaceuticals, Inc. Compositions and methods for enhanced biostability and altered biodistribution of oligonucleotides in mammals
US20040142899A1 (en) * 1990-01-11 2004-07-22 Isis Pharmaceuticals, Inc. Compositions and methods for enhanced biostability and altered biodistribution of oligonucleotides in mammals
US5688941A (en) * 1990-07-27 1997-11-18 Isis Pharmaceuticals, Inc. Methods of making conjugated 4' desmethyl nucleoside analog compounds
US5998603A (en) 1994-09-29 1999-12-07 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analogs, and oligomers thereof
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
US8153602B1 (en) 1991-11-19 2012-04-10 Isis Pharmaceuticals, Inc. Composition and methods for the pulmonary delivery of nucleic acids
US6001982A (en) * 1993-07-29 1999-12-14 Isis Pharmaceuticals, Inc. Synthesis of oligonucleotides
CA2170869C (en) 1993-09-03 1999-09-14 Phillip Dan Cook Amine-derivatized nucleosides and oligonucleosides
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
US6025335A (en) * 1995-09-21 2000-02-15 Lipitek International, Inc. L-Nucleoside Dimer Compounds and therapeutic uses
EP0863910A1 (en) 1995-10-19 1998-09-16 NeXstar Pharmaceuticals, Inc. Method for solution phase synthesis of oligonucleotides
US20030044941A1 (en) 1996-06-06 2003-03-06 Crooke Stanley T. Human RNase III and compositions and uses thereof
US20040266706A1 (en) * 2002-11-05 2004-12-30 Muthiah Manoharan Cross-linked oligomeric compounds and their use in gene modulation
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US20040171031A1 (en) * 1996-06-06 2004-09-02 Baker Brenda F. Sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US20050119470A1 (en) * 1996-06-06 2005-06-02 Muthiah Manoharan Conjugated oligomeric compounds and their use in gene modulation
US7812149B2 (en) * 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US20040203024A1 (en) * 1996-06-06 2004-10-14 Baker Brenda F. Modified oligonucleotides for use in RNA interference
US20050053976A1 (en) * 1996-06-06 2005-03-10 Baker Brenda F. Chimeric oligomeric compounds and their use in gene modulation
US20040147022A1 (en) * 1996-06-06 2004-07-29 Baker Brenda F. 2'-methoxy substituted oligomeric compounds and compositions for use in gene modulations
US6315978B1 (en) 1996-08-27 2001-11-13 University Of Utah, Research Foundation Bioconjugates and delivery of bioactive agents
US5908845A (en) * 1996-10-30 1999-06-01 Segev; David Polyether nucleic acids
EP1012331B1 (en) 1997-07-01 2006-03-29 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
US5877309A (en) 1997-08-13 1999-03-02 Isis Pharmaceuticals, Inc. Antisense oligonucleotides against JNK
US20070149472A1 (en) * 1997-08-13 2007-06-28 Mckay Robert Antisense oligonucleotide compositions and methods for the modulation of jnk proteins
US6809193B2 (en) 1997-08-13 2004-10-26 Isis Pharmaceuticals, Inc. Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US6133246A (en) * 1997-08-13 2000-10-17 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
US7321828B2 (en) * 1998-04-13 2008-01-22 Isis Pharmaceuticals, Inc. System of components for preparing oligonucleotides
US20040186071A1 (en) * 1998-04-13 2004-09-23 Bennett C. Frank Antisense modulation of CD40 expression
CA2329252A1 (en) * 1998-05-21 1999-11-25 Isis Pharmaceuticals Inc. Compositions and methods for topical delivery of oligonucleotides
JP2003524586A (en) * 1998-05-21 2003-08-19 アイシス・ファーマシューティカルス・インコーポレーテッド Compositions and methods for parenteral administration of oligonucleotides
US6300319B1 (en) 1998-06-16 2001-10-09 Isis Pharmaceuticals, Inc. Targeted oligonucleotide conjugates
US6124099A (en) * 1998-06-22 2000-09-26 The University Of Vermont And State Agricultural College Method for placing a photo-cross-linking agent at specific internal sites within the sequence of synthetic strands of ribonucleic acids
US6867294B1 (en) * 1998-07-14 2005-03-15 Isis Pharmaceuticals, Inc. Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
US6242589B1 (en) 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
US6225293B1 (en) 1998-09-02 2001-05-01 Isis Pharmaceuticals, Inc. Methods and compounds for tracking the biodistribution of macromolecule-carrier combinations
US6077709A (en) 1998-09-29 2000-06-20 Isis Pharmaceuticals Inc. Antisense modulation of Survivin expression
IL142094A0 (en) 1998-09-29 2002-03-10 Gamida Cell Ltd Methods of controlling proliferation and differentiation of stem and progenitor cells
US6492111B1 (en) * 1998-11-25 2002-12-10 Isis Pharmaceuticals, Inc. In situ binary synthesis of biologically effective molecules
US6300320B1 (en) 1999-01-05 2001-10-09 Isis Pharmaceuticals, Inc. Modulation of c-jun using inhibitors of protein kinase C
US6127124A (en) 1999-01-20 2000-10-03 Isis Pharmaceuticals, Inc. Fluorescence based nuclease assay
US7098192B2 (en) 1999-04-08 2006-08-29 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of STAT3 expression
US7534605B2 (en) * 1999-06-08 2009-05-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem CD44 polypeptides, polynucleotides encoding same, antibodies directed thereagainst and method of using same for diagnosing and treating inflammatory diseases
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
US6593466B1 (en) 1999-07-07 2003-07-15 Isis Pharmaceuticals, Inc. Guanidinium functionalized nucleotides and precursors thereof
US6114518A (en) * 1999-09-30 2000-09-05 Becton, Dickinson And Company Synthesis and use of labelled phosphoramidite compositions
US7668658B2 (en) * 1999-10-13 2010-02-23 Sequenom, Inc. Methods for generating databases and databases for identifying polymorphic genetic markers
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
US6693033B2 (en) 2000-02-10 2004-02-17 Motorola, Inc. Method of removing an amorphous oxide from a monocrystalline surface
US6392257B1 (en) * 2000-02-10 2002-05-21 Motorola Inc. Semiconductor structure, semiconductor device, communicating device, integrated circuit, and process for fabricating the same
US20030176385A1 (en) * 2000-02-15 2003-09-18 Jingfang Ju Antisense modulation of protein expression
EP2202241A3 (en) * 2000-04-13 2010-07-28 Thomas N. Wight Therapeutic compounds and methods for modulating V3, a versican isoform
US6680172B1 (en) 2000-05-16 2004-01-20 Regents Of The University Of Michigan Treatments and markers for cancers of the central nervous system
US6656700B2 (en) * 2000-05-26 2003-12-02 Amersham Plc Isoforms of human pregnancy-associated protein-E
US6686188B2 (en) * 2000-05-26 2004-02-03 Amersham Plc Polynucleotide encoding a human myosin-like polypeptide expressed predominantly in heart and muscle
US20060166227A1 (en) * 2000-06-20 2006-07-27 Stephen Kingsmore Protein expression profiling
US6323009B1 (en) * 2000-06-28 2001-11-27 Molecular Staging, Inc. Multiply-primed amplification of nucleic acid sequences
AU2001264987A1 (en) * 2000-06-30 2002-01-14 Motorola, Inc., A Corporation Of The State Of Delware Hybrid semiconductor structure and device
US6958214B2 (en) 2000-07-10 2005-10-25 Sequenom, Inc. Polymorphic kinase anchor proteins and nucleic acids encoding the same
US6555946B1 (en) 2000-07-24 2003-04-29 Motorola, Inc. Acoustic wave device and process for forming the same
US8568766B2 (en) * 2000-08-24 2013-10-29 Gattadahalli M. Anantharamaiah Peptides and peptide mimetics to treat pathologies associated with eye disease
EP1191097A1 (en) * 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induction of exon skipping in eukaryotic cells
US6638838B1 (en) 2000-10-02 2003-10-28 Motorola, Inc. Semiconductor structure including a partially annealed layer and method of forming the same
US20020123474A1 (en) * 2000-10-04 2002-09-05 Shannon Mark E. Human GTP-Rho binding protein2
EP2336166A1 (en) 2000-10-12 2011-06-22 University Of Rochester Compositions that inhibit proliferation of cancer cells
US7767802B2 (en) 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US6673646B2 (en) 2001-02-28 2004-01-06 Motorola, Inc. Growth of compound semiconductor structures on patterned oxide films and process for fabricating same
US6573051B2 (en) * 2001-03-09 2003-06-03 Molecular Staging, Inc. Open circle probes with intramolecular stem structures
ES2328796T3 (en) 2001-03-14 2009-11-18 Myriad Genetics, Inc. TSG101-GAG INTERACTION AND USE OF THE SAME.
CA2446619C (en) 2001-05-11 2011-04-26 Elan Corporation, Plc Permeation enhancers
DK2000545T3 (en) 2001-06-20 2011-11-28 Genentech Inc Compositions and methods for diagnosis and treatment of lung tumor
US7803915B2 (en) * 2001-06-20 2010-09-28 Genentech, Inc. Antibody compositions for the diagnosis and treatment of tumor
US20050107595A1 (en) * 2001-06-20 2005-05-19 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
CA2451643C (en) 2001-06-21 2012-11-13 Isis Pharmaceuticals, Inc. Antisense modulation of superoxide dismutase 1, soluble expression
US6709989B2 (en) 2001-06-21 2004-03-23 Motorola, Inc. Method for fabricating a semiconductor structure including a metal oxide interface with silicon
US20030010992A1 (en) * 2001-07-16 2003-01-16 Motorola, Inc. Semiconductor structure and method for implementing cross-point switch functionality
US6646293B2 (en) 2001-07-18 2003-11-11 Motorola, Inc. Structure for fabricating high electron mobility transistors utilizing the formation of complaint substrates
US6693298B2 (en) 2001-07-20 2004-02-17 Motorola, Inc. Structure and method for fabricating epitaxial semiconductor on insulator (SOI) structures and devices utilizing the formation of a compliant substrate for materials used to form same
US6855992B2 (en) * 2001-07-24 2005-02-15 Motorola Inc. Structure and method for fabricating configurable transistor devices utilizing the formation of a compliant substrate for materials used to form the same
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US6667196B2 (en) 2001-07-25 2003-12-23 Motorola, Inc. Method for real-time monitoring and controlling perovskite oxide film growth and semiconductor structure formed using the method
US6964950B2 (en) 2001-07-25 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of C-reactive protein expression
US20030096772A1 (en) 2001-07-30 2003-05-22 Crooke Rosanne M. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
US7407943B2 (en) 2001-08-01 2008-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein B expression
US6639249B2 (en) 2001-08-06 2003-10-28 Motorola, Inc. Structure and method for fabrication for a solid-state lighting device
US6589856B2 (en) 2001-08-06 2003-07-08 Motorola, Inc. Method and apparatus for controlling anti-phase domains in semiconductor structures and devices
US20040096880A1 (en) * 2001-08-07 2004-05-20 Kmiec Eric B. Compositions and methods for the treatment of diseases exhibiting protein misassembly and aggregation
WO2003013437A2 (en) * 2001-08-07 2003-02-20 University Of Delaware Compositions and methods for the prevention and treatment of huntington's disease
US7227014B2 (en) 2001-08-07 2007-06-05 Isis Pharmaceuticals, Inc. Antisense modulation of apolipoprotein (a) expression
US6673667B2 (en) 2001-08-15 2004-01-06 Motorola, Inc. Method for manufacturing a substantially integral monolithic apparatus including a plurality of semiconductor materials
US20030036217A1 (en) * 2001-08-16 2003-02-20 Motorola, Inc. Microcavity semiconductor laser coupled to a waveguide
CA2460120A1 (en) 2001-09-18 2003-03-27 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
ATE516364T1 (en) 2001-10-09 2011-07-15 Isis Pharmaceuticals Inc ANTISENSE MODULATION OF EXPRESSION OF THE INSULIN-LIKE GROWTH FACTOR BINDING PROTEY S 5
US6750019B2 (en) 2001-10-09 2004-06-15 Isis Pharmaceuticals, Inc. Antisense modulation of insulin-like growth factor binding protein 5 expression
US20030071327A1 (en) * 2001-10-17 2003-04-17 Motorola, Inc. Method and apparatus utilizing monocrystalline insulator
US20030170678A1 (en) * 2001-10-25 2003-09-11 Neurogenetics, Inc. Genetic markers for Alzheimer's disease and methods using the same
AU2002364945A1 (en) * 2001-10-25 2003-07-09 Neurogenetics, Inc. Genes and polymorphisms on chromosome 10 associated with alzheimer's disease and other neurodegenerative diseases
US20030224380A1 (en) * 2001-10-25 2003-12-04 The General Hospital Corporation Genes and polymorphisms on chromosome 10 associated with Alzheimer's disease and other neurodegenerative diseases
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
JP2005525095A (en) 2002-01-02 2005-08-25 ジェネンテック・インコーポレーテッド Compositions and methods for tumor diagnosis and treatment
IL152904A0 (en) * 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
WO2003062404A1 (en) * 2002-01-25 2003-07-31 Gamida-Cell Ltd. Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
US7553619B2 (en) * 2002-02-08 2009-06-30 Qiagen Gmbh Detection method using dissociated rolling circle amplification
US20030180712A1 (en) 2002-03-20 2003-09-25 Biostratum Ab Inhibition of the beta3 subunit of L-type Ca2+ channels
US7169916B2 (en) * 2002-04-01 2007-01-30 Isis Pharmaceuticals, Inc. Chloral-free DCA in oligonucleotide synthesis
AU2003230874A1 (en) 2002-04-16 2003-11-03 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
CA2484676A1 (en) * 2002-05-03 2003-11-13 Sequenom, Inc. Kinase anchor protein muteins, peptides thereof, and related methods
US6916717B2 (en) * 2002-05-03 2005-07-12 Motorola, Inc. Method for growing a monocrystalline oxide layer and for fabricating a semiconductor device on a monocrystalline substrate
US7199107B2 (en) 2002-05-23 2007-04-03 Isis Pharmaceuticals, Inc. Antisense modulation of kinesin-like 1 expression
WO2003106617A2 (en) * 2002-06-12 2003-12-24 Tel Aviv Medical Center Research Development Fund Oligonucleotides antibodies and kits including same for treating prostate cancer and determining predisposition thereto
AU2003276131A1 (en) * 2002-06-18 2003-12-31 Epigenesis Pharmaceuticals, Inc. A dry powder oligonucleotide formulation, preparation and its uses
US20040019000A1 (en) * 2002-07-19 2004-01-29 Muthiah Manoharan Polyalkyleneamine-containing oligomers
WO2004013160A2 (en) 2002-08-05 2004-02-12 University Of Rochester Protein transducing domain/deaminase chimeric proteins, related compounds, and uses thereof
US6878805B2 (en) * 2002-08-16 2005-04-12 Isis Pharmaceuticals, Inc. Peptide-conjugated oligomeric compounds
BR0314236A (en) 2002-09-13 2005-08-09 Replicor Inc Oligonucleotide formulation, pharmaceutical composition, kit, antiviral compound, preparation of oligonucleotide and methods for selection of an antiviral oligonucleotide for use as an antiviral agent, for prophylaxis or treatment of a viral infection in a patient, for prophylactic treatment of cancer caused by oncoviruses. for identifying a compound that alters the binding of an oligonucleotide to at least one viral component, for purifying oligonucleotide binding to at least one viral component and for enriching oligonucleotides from an oligonucleotide cluster
EP1546170A4 (en) * 2002-09-20 2007-08-29 Univ Yale Riboswitches, methods for their use, and compositions for use with riboswitches
EP2272958A1 (en) 2002-09-26 2011-01-12 ISIS Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
US20040070312A1 (en) * 2002-10-10 2004-04-15 Motorola, Inc. Integrated circuit and process for fabricating the same
AU2003304203A1 (en) * 2002-10-29 2005-01-04 Pharmacia Corporation Differentially expressed genes involved in cancer, the polypeptides encoded thereby, and methods of using the same
US9150606B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
US7696345B2 (en) 2002-11-05 2010-04-13 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
CA2504929C (en) * 2002-11-05 2014-07-22 Charles Allerson Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
US9150605B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
WO2004044138A2 (en) 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
JP4986109B2 (en) 2002-11-13 2012-07-25 ジェンザイム・コーポレーション Antisense regulation of apolipoprotein B expression
DK1569695T3 (en) 2002-11-13 2013-08-05 Genzyme Corp ANTISENSE MODULATION OF APOLIPOPROTEIN-B EXPRESSION
US20060009378A1 (en) * 2002-11-14 2006-01-12 Itshak Golan Novel galectin sequences and compositions and methods utilizing same for treating or diagnosing arthritis and other chronic inflammatory diseases
AU2003298650B2 (en) 2002-11-15 2010-03-11 Musc Foundation For Research Development Complement receptor 2 targeted complement modulators
CA2506127C (en) 2002-11-15 2013-07-09 Morphotek, Inc. Methods of generating high-production of antibodies from hybridomas created by in vitro immunization
EP1624753B1 (en) 2002-11-21 2012-01-25 The University of Utah Research Foundation Purinergic modulation of smell
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
US20040121338A1 (en) * 2002-12-19 2004-06-24 Alsmadi Osama A. Real-time detection of rolling circle amplification products
AU2003299694A1 (en) 2002-12-20 2004-07-22 Qiagen Gmbh Nucleic acid amplification
US9487823B2 (en) * 2002-12-20 2016-11-08 Qiagen Gmbh Nucleic acid amplification
US7020374B2 (en) * 2003-02-03 2006-03-28 Freescale Semiconductor, Inc. Optical waveguide structure and method for fabricating the same
US6965128B2 (en) * 2003-02-03 2005-11-15 Freescale Semiconductor, Inc. Structure and method for fabricating semiconductor microresonator devices
DK1597366T3 (en) 2003-02-11 2013-02-25 Antisense Therapeutics Ltd Modulation of expression of insulin-like growth factor receptor I
US7002006B2 (en) * 2003-02-12 2006-02-21 Isis Pharmaceuticals, Inc. Protection of nucleosides
US20040164315A1 (en) * 2003-02-25 2004-08-26 Motorola, Inc. Structure and device including a tunneling piezoelectric switch and method of forming same
CA2517074A1 (en) 2003-02-27 2004-09-10 Yeda Research And Development Co., Ltd. Nucleic acid molecules, polypeptides, antibodies and compositions containing same useful for treating and detecting influenza virus infection
US7803781B2 (en) 2003-02-28 2010-09-28 Isis Pharmaceuticals, Inc. Modulation of growth hormone receptor expression and insulin-like growth factor expression
US20070141570A1 (en) * 2003-03-07 2007-06-21 Sequenom, Inc. Association of polymorphic kinase anchor proteins with cardiac phenotypes and related methods
US20040185559A1 (en) 2003-03-21 2004-09-23 Isis Pharmaceuticals Inc. Modulation of diacylglycerol acyltransferase 1 expression
CA2524255C (en) 2003-03-21 2014-02-11 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
US8043834B2 (en) 2003-03-31 2011-10-25 Qiagen Gmbh Universal reagents for rolling circle amplification and methods of use
US20040198640A1 (en) * 2003-04-02 2004-10-07 Dharmacon, Inc. Stabilized polynucleotides for use in RNA interference
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
US7399853B2 (en) 2003-04-28 2008-07-15 Isis Pharmaceuticals Modulation of glucagon receptor expression
US7276599B2 (en) * 2003-06-02 2007-10-02 Isis Pharmaceuticals, Inc. Oligonucleotide synthesis with alternative solvents
AU2004253455B2 (en) 2003-06-03 2011-03-03 Eli Lilly And Company Modulation of survivin expression
GB2417727B (en) 2003-06-13 2008-01-16 Alnylam Europe Ag Double-stranded ribonucleic acid with increased effectiveness in an organism
US20060241072A1 (en) * 2003-06-20 2006-10-26 Isis Pharmaceuticals, Inc. Oligomeric compounds for use in gene modulation
EP1644475A4 (en) * 2003-06-20 2009-06-03 Isis Pharmaceuticals Inc Double stranded compositions comprising a 3'-endo modified strand for use in gene modulation
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
US20050053981A1 (en) * 2003-09-09 2005-03-10 Swayze Eric E. Gapped oligomeric compounds having linked bicyclic sugar moieties at the termini
US20070123480A1 (en) * 2003-09-11 2007-05-31 Replicor Inc. Oligonucleotides targeting prion diseases
SG146682A1 (en) 2003-09-18 2008-10-30 Isis Pharmaceuticals Inc Modulation of eif4e expression
EP1677822B1 (en) * 2003-09-18 2014-04-23 Isis Pharmaceuticals, Inc. 4'-thionucleosides and oligomeric compounds
US7662929B2 (en) 2003-10-10 2010-02-16 Alchemia Oncology Pty Limited Antibody that specifically binds hyaluronan synthase
US20050191653A1 (en) 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
CA2747871C (en) 2003-11-17 2018-04-10 Genentech, Inc. Compositions and methods for the treatment of tumor of hematopoietic origin
WO2006054296A2 (en) 2004-11-17 2006-05-26 Spectrum Dynamics Llc Methods of detecting prostate cancer
WO2005071080A2 (en) 2004-01-20 2005-08-04 Isis Pharmaceuticals, Inc. Modulation of glucocorticoid receptor expression
US8778900B2 (en) * 2004-01-22 2014-07-15 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP1 expression
US7468431B2 (en) * 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
US7842459B2 (en) 2004-01-27 2010-11-30 Compugen Ltd. Nucleotide and amino acid sequences, and assays and methods of use thereof for diagnosis
US20090280567A1 (en) * 2004-02-06 2009-11-12 Dharmacon, Inc. Stabilized sirnas as transfection controls and silencing reagents
US20070269889A1 (en) * 2004-02-06 2007-11-22 Dharmacon, Inc. Stabilized siRNAs as transfection controls and silencing reagents
US8569474B2 (en) * 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
EP2700720A3 (en) 2004-03-15 2015-01-28 Isis Pharmaceuticals, Inc. Compositions and methods for optimizing cleavage of RNA by RNASE H
KR101147147B1 (en) * 2004-04-01 2012-05-25 머크 샤프 앤드 돔 코포레이션 Modified polynucleotides for reducing off-target effects in rna interference
AU2005230684B2 (en) 2004-04-05 2011-10-06 Alnylam Pharmaceuticals, Inc. Process and reagents for oligonucleotide synthesis and purification
US20050244869A1 (en) * 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
US20050260755A1 (en) * 2004-04-06 2005-11-24 Isis Pharmaceuticals, Inc. Sequential delivery of oligomeric compounds
CA2562151C (en) 2004-04-30 2016-09-06 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a c5-modified pyrimidine
EP3225633B1 (en) 2004-05-21 2020-03-25 The UAB Research Foundation Variable lymphocyte receptors, related polypeptides and nucleic acids, and uses thereof
JP2008501335A (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. Chimeric gapped oligomer composition
US8394947B2 (en) * 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
AU2005252663B2 (en) * 2004-06-03 2011-07-07 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
WO2006023880A2 (en) * 2004-08-23 2006-03-02 Isis Pharmaceuticals, Inc. Compounds and methods for the characterization of oligonucleotides
US7884086B2 (en) * 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
WO2006030442A2 (en) * 2004-09-16 2006-03-23 Gamida-Cell Ltd. Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
US20060063147A1 (en) * 2004-09-21 2006-03-23 Chernov Boris K Omega-amino-PEG-phosphoramidites and conjugates thereof
DK1809303T3 (en) 2004-09-23 2019-06-11 Arc Medical Devices Inc PHARMACEUTICAL COMPOSITION AND METHODS IN RELATION TO FIBROSE ADHESION OR INFLAMMATORIC DISEASE USING LOW SULPHATE FUCAN
WO2006050999A2 (en) * 2004-11-15 2006-05-18 Obe Therapy Biotechnology S.A.S Methods of reducing body fat
US7923207B2 (en) 2004-11-22 2011-04-12 Dharmacon, Inc. Apparatus and system having dry gene silencing pools
US7935811B2 (en) * 2004-11-22 2011-05-03 Dharmacon, Inc. Apparatus and system having dry gene silencing compositions
US7923206B2 (en) * 2004-11-22 2011-04-12 Dharmacon, Inc. Method of determining a cellular response to a biological agent
CA2597673C (en) 2005-02-11 2014-07-08 Harold Varmus Methods and compositions for detecting a drug resistant egfr mutant
RU2007137489A (en) 2005-03-10 2009-04-20 Дженентек, Инк. (Us) METHODS AND COMPOSITIONS FOR MODULATION OF VESSEL INTEGRITY
US7476733B2 (en) * 2005-03-25 2009-01-13 The United States Of America As Represented By The Department Of Health And Human Services Development of a real-time PCR assay for detection of pneumococcal DNA and diagnosis of pneumococccal disease
US20060223777A1 (en) * 2005-03-29 2006-10-05 Dharmacon, Inc. Highly functional short hairpin RNA
US8309303B2 (en) * 2005-04-01 2012-11-13 Qiagen Gmbh Reverse transcription and amplification of RNA with simultaneous degradation of DNA
CA2605512A1 (en) * 2005-04-22 2006-10-26 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the binding of sr proteins and by interfering with secondary rna structure.
EP2298829B1 (en) 2005-05-31 2017-09-20 École Polytechnique Fédérale de Lausanne (EPFL) Triblock copolymers for cytoplasmic delivery of gene-based drugs
EP1904111A4 (en) 2005-06-03 2009-08-19 Univ Johns Hopkins Compositions and methods for decreasing microrna expression for the treatment of neoplasia
US8252756B2 (en) 2005-06-14 2012-08-28 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
WO2007007317A1 (en) 2005-07-07 2007-01-18 Yissum Research Development Company Of The Hebrew University Of Jerusalem Nucleic acid agents for downregulating h19, and methods of using same
US8067571B2 (en) 2005-07-13 2011-11-29 Avi Biopharma, Inc. Antibacterial antisense oligonucleotide and method
US7776532B2 (en) 2005-08-11 2010-08-17 Synthetic Genomics, Inc. Method for in vitro recombination
US20100015604A1 (en) 2005-08-17 2010-01-21 Evriklia Lianidou Composition and method for determination of ck19 expression
EP2338991B1 (en) 2005-08-29 2017-01-18 Regulus Therapeutics Inc. Methods for use in modulating MIR-122a
EP1762627A1 (en) 2005-09-09 2007-03-14 Qiagen GmbH Method for the activation of a nucleic acid for performing a polymerase reaction
IL172297A (en) 2005-10-03 2016-03-31 Compugen Ltd Soluble vegfr-1 variants for diagnosis of preeclamsia
US8080534B2 (en) 2005-10-14 2011-12-20 Phigenix, Inc Targeting PAX2 for the treatment of breast cancer
EP2392645A1 (en) 2005-10-14 2011-12-07 MUSC Foundation For Research Development Targeting PAX2 for the induction of DEFB1-mediated tumor immunity and cancer therapy
CA2627025A1 (en) 2005-10-28 2007-05-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of huntingtin gene
WO2007056331A2 (en) 2005-11-09 2007-05-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor v leiden mutant gene
CA2630602A1 (en) 2005-11-21 2007-05-31 Isis Pharmaceuticals, Inc. Modulation of eif4e-bp2 expression
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
US8313901B2 (en) * 2005-12-21 2012-11-20 Yale University Methods and compositions related to the modulation of riboswitches
EP1976567B1 (en) 2005-12-28 2020-05-13 The Scripps Research Institute Natural antisense and non-coding rna transcripts as drug targets
EP2216339A1 (en) 2006-01-16 2010-08-11 Compugen Ltd. Novel nucleotide and amino acid sequences, and methods of use thereof for diagnosis
DE602007009487D1 (en) * 2006-01-27 2010-11-11 Isis Pharmaceutical Inc 6-MODIFIED BICYCLIC NUCLEIC ACID ANALOGUE
US8129515B2 (en) 2006-01-27 2012-03-06 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for the use in modulation of microRNAs
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
AU2007233109B2 (en) 2006-03-31 2010-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Eg5 gene
WO2007123391A1 (en) * 2006-04-20 2007-11-01 Academisch Ziekenhuis Leiden Therapeutic intervention in a genetic disease in an individual by modifying expression of an aberrantly expressed gene.
CN101437943A (en) * 2006-05-03 2009-05-20 波罗的科技发展有限公司 Antisense agents combining strongly bound base - modified oligonucleotide and artificial nuclease
EP2015758B1 (en) 2006-05-05 2014-04-02 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression apob
DE102006020885A1 (en) * 2006-05-05 2007-11-08 Qiagen Gmbh Inserting a tag sequence into a nucleic acid comprises using an anchor oligonucleotide comprising a hybridizing anchor sequence and a nonhybridizing tag-template sequence
US7666854B2 (en) * 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
PL2194128T3 (en) 2006-05-11 2012-12-31 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of the PCSK9 gene
CN101490074B (en) * 2006-05-11 2013-06-26 Isis制药公司 5'-modified bicyclic nucleic acid analogs
EP1857548A1 (en) * 2006-05-19 2007-11-21 Academisch Ziekenhuis Leiden Means and method for inducing exon-skipping
BRPI0712034A2 (en) 2006-05-19 2012-01-10 Alnylam Pharmaceuticals Inc aha rnai modulation and therapeutic uses thereof
EP2192200B1 (en) 2006-05-22 2012-10-24 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of IKK-B gene
WO2007137301A2 (en) * 2006-05-23 2007-11-29 Isis Pharmaceuticals, Inc. Modulation of chrebp expression
WO2008097328A2 (en) * 2006-06-23 2008-08-14 Northwestern University Asymmetric functionalized nanoparticles and methods of use
WO2008011473A2 (en) 2006-07-19 2008-01-24 Isis Pharmaceuticals, Inc. Compositions and their uses directed to hbxip
EP2049664B1 (en) 2006-08-11 2011-09-14 Prosensa Technologies B.V. Single stranded oligonucleotides complementary to repetitive elements for treating DNA repeat instability associated genetic disorders
JP2010502752A (en) * 2006-09-11 2010-01-28 イェール ユニバーシティー Lysine riboswitch, structure-based compound design using lysine riboswitch, and methods and compositions for use with lysine riboswitch
WO2008036933A2 (en) 2006-09-21 2008-03-27 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the hamp gene
EP2064223B1 (en) * 2006-09-22 2013-04-24 Dharmacon, Inc. Duplex oligonucleotide complexes and methods for gene silencing by RNA interference
CA2927045A1 (en) 2006-10-03 2008-04-10 Muthiah Manoharan Lipid containing formulations
US20100166743A1 (en) 2006-10-06 2010-07-01 University Of Utah Research Foundation Method of detecting ocular diseases and pathologic conditions and treatment of same
US8999317B2 (en) 2006-11-01 2015-04-07 University Of Rochester Methods and compositions related to the structure and function of APOBEC3G
CA2672297A1 (en) * 2006-12-11 2008-06-19 University Of Utah Research Foundation Compositions and methods for treating pathologic angiogenesis and vascular permeability
EP2097448A4 (en) 2006-12-22 2010-07-21 Univ Utah Res Found Method of detecting ocular diseases and pathologic conditions and treatment of same
US20100086526A1 (en) * 2007-01-16 2010-04-08 Abraham Hochberg Nucleic acid constructs and methods for specific silencing of h19
US20100196403A1 (en) * 2007-01-29 2010-08-05 Jacob Hochman Antibody conjugates for circumventing multi-drug resistance
AU2008212820B2 (en) * 2007-02-09 2014-01-30 Northwestern University Particles for detecting intracellular targets
EP2129803A4 (en) * 2007-02-27 2010-11-03 Univ Northwestern Molecule attachment to nanoparticles
EP2471925A1 (en) 2007-03-22 2012-07-04 Yale University Methods and compositions related to riboswitches that control alternative splicing
PE20090064A1 (en) 2007-03-26 2009-03-02 Novartis Ag DOUBLE-CHAIN RIBONUCLEIC ACID TO INHIBIT THE EXPRESSION OF THE HUMAN E6AP GENE AND THE PHARMACEUTICAL COMPOSITION THAT INCLUDES IT
JP5350360B2 (en) 2007-03-29 2013-11-27 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for inhibiting the expression of genes from Ebola
CA2690281A1 (en) 2007-05-11 2008-11-20 The Johns Hopkins University Biomarkers for melanoma
MX2009012647A (en) * 2007-05-29 2009-12-14 Univ Yale Methods and compositions related to riboswitches that control alternative splicing and rna processing.
KR20100017905A (en) 2007-05-29 2010-02-16 예일 유니버시티 Riboswitches and methods and compositions for use of and with riboswitches
CA2688321A1 (en) 2007-05-30 2008-12-11 Isis Pharmaceuticals, Inc. N-substituted-aminomethylene bridged bicyclic nucleic acid analogs
EP2160464B1 (en) * 2007-05-30 2014-05-21 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
US7807372B2 (en) * 2007-06-04 2010-10-05 Northwestern University Screening sequence selectivity of oligonucleotide-binding molecules using nanoparticle based colorimetric assay
EP2173760B2 (en) 2007-06-08 2015-11-04 Isis Pharmaceuticals, Inc. Carbocyclic bicyclic nucleic acid analogs
CA2692579C (en) * 2007-07-05 2016-05-03 Isis Pharmaceuticals, Inc. 6-disubstituted bicyclic nucleic acid analogs
EP2319926B1 (en) 2007-07-05 2016-08-31 Arrowhead Research Corporation DSRNA for treating viral infection
WO2009008725A2 (en) * 2007-07-12 2009-01-15 Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs, tissues or tumor cells
JP5706157B2 (en) * 2007-07-12 2015-04-22 プロセンサ テクノロジーズ ビー.ブイ.Prosensa Technologies B.V. Molecules for targeting compounds to various selected organs or tissues
CN101821277B (en) * 2007-08-15 2014-05-07 Isis制药公司 Tetrahydropyran nucleic acid analogs
US9422363B2 (en) * 2007-08-28 2016-08-23 Uab Research Foundation Synthetic apolipoprotein E mimicking polypeptides and methods of use
CA2697957A1 (en) 2007-08-28 2009-03-12 Uab Research Foundation Synthetic apolipoprotein e mimicking polypeptides and methods of use
LT2769729T (en) 2007-09-04 2019-05-10 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
US8445217B2 (en) 2007-09-20 2013-05-21 Vanderbilt University Free solution measurement of molecular interactions by backscattering interferometry
US7951785B2 (en) * 2007-09-21 2011-05-31 California Institute Of Technology NFIA in glial fate determination, glioma therapy and astrocytoma treatment
JP2010539978A (en) 2007-10-02 2010-12-24 アムジェン インコーポレイテッド Increased erythropoietin using nucleic acids that can hybridize to micro-RNA and its precursors
WO2009045536A2 (en) * 2007-10-05 2009-04-09 The University Of North Carolina At Chapel Hill Receptor targeted oligonucleotides
USRE48468E1 (en) 2007-10-26 2021-03-16 Biomarin Technologies B.V. Means and methods for counteracting muscle disorders
JP5600064B2 (en) 2007-10-26 2014-10-01 アカデミシュ ジーケンハウス ライデン Means and methods for offsetting myopathy
JP2011502502A (en) * 2007-11-05 2011-01-27 バルティック テクロノジー デヴェロプメント,リミテッド Use of oligonucleotides containing modified bases in nucleic acid hybridization
US8097712B2 (en) 2007-11-07 2012-01-17 Beelogics Inc. Compositions for conferring tolerance to viral disease in social insects, and the use thereof
USRE47320E1 (en) 2007-11-20 2019-03-26 Ionis Pharmaceuticals, Inc. Modulation of CD40 expression
US8546556B2 (en) * 2007-11-21 2013-10-01 Isis Pharmaceuticals, Inc Carbocyclic alpha-L-bicyclic nucleic acid analogs
EP2848688A1 (en) 2007-12-10 2015-03-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor VII gene
US7845686B2 (en) * 2007-12-17 2010-12-07 S & B Technical Products, Inc. Restrained pipe joining system for plastic pipe
EP3100718B1 (en) 2008-01-02 2019-11-27 Arbutus Biopharma Corporation Improved compositions and methods for the delivery of nucleic acids
US8530640B2 (en) * 2008-02-07 2013-09-10 Isis Pharmaceuticals, Inc. Bicyclic cyclohexitol nucleic acid analogs
AU2009210872A1 (en) * 2008-02-08 2009-08-13 Prosensa Holding Bv Methods and means for treating DNA repeat instability associated genetic disorders
US8188060B2 (en) 2008-02-11 2012-05-29 Dharmacon, Inc. Duplex oligonucleotides with enhanced functionality in gene regulation
KR101397407B1 (en) 2008-03-05 2014-06-19 알닐람 파마슈티칼스 인코포레이티드 Compositions and methods for inhibiting expression of Eg5 and VEGF genes
US8426378B2 (en) 2008-03-21 2013-04-23 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising tricyclic nucelosides and methods for their use
EP2105145A1 (en) * 2008-03-27 2009-09-30 ETH Zürich Method for muscle-specific delivery lipid-conjugated oligonucleotides
WO2009124238A1 (en) * 2008-04-04 2009-10-08 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising neutrally linked terminal bicyclic nucleosides
WO2009124295A2 (en) * 2008-04-04 2009-10-08 Isis Pharmaceuticals, Inc. Oligomeric compounds comprising bicyclic nucleosides and having reduced toxicity
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
DK2982753T3 (en) 2008-04-18 2018-09-03 Baxter Int Microsphere-based composition to prevent and / or reverse newly started autoimmune diabetes
US8324366B2 (en) 2008-04-29 2012-12-04 Alnylam Pharmaceuticals, Inc. Compositions and methods for delivering RNAI using lipoproteins
CA2722668A1 (en) * 2008-04-29 2009-11-05 Wyeth Llc Methods for treating inflammation
EP2119783A1 (en) 2008-05-14 2009-11-18 Prosensa Technologies B.V. Method for efficient exon (44) skipping in Duchenne Muscular Dystrophy and associated means
US8082730B2 (en) * 2008-05-20 2011-12-27 Caterpillar Inc. Engine system having particulate reduction device and method
KR20110017005A (en) * 2008-07-15 2011-02-18 에프. 호프만-라 로슈 아게 Compositions and methods for inhibiting expression of tgf-beta receptor genes
EP2323667A4 (en) * 2008-08-07 2012-07-25 Isis Pharmaceuticals Inc Modulation of transthyretin expression for the treatment of cns related disorders
JP5420668B2 (en) 2008-08-25 2014-02-19 エクスカリアード・ファーマシューティカルズ,インコーポレイテッド Antisense oligonucleotides for connective tissue growth factor and uses thereof
EP2331690B1 (en) 2008-09-02 2016-01-13 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of mutant egfr gene
US8962580B2 (en) 2008-09-23 2015-02-24 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
WO2010036696A1 (en) * 2008-09-24 2010-04-01 Isis Pharmaceuticals, Inc. Cyclohexenyl nucleic acid analogs
EP3109321B1 (en) 2008-09-25 2019-05-01 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of serum amyloid a gene
CA2740000C (en) 2008-10-09 2017-12-12 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010045509A2 (en) 2008-10-15 2010-04-22 Isis Pharmaceuticals, Inc. Modulation of factor 11 expression
EP3848461A1 (en) 2008-10-20 2021-07-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
CN102264374B (en) 2008-10-24 2015-01-07 Isis制药公司 5' and 2' bis-substituted nucleosides and oligomeric compounds prepared therefrom
US8987435B2 (en) 2008-10-24 2015-03-24 Isis Pharmaceuticals, Inc. Oligomeric compounds and methods
AU2009313201B2 (en) 2008-11-10 2016-06-16 Arbutus Biopharma Corporation Novel lipids and compositions for the delivery of therapeutics
RU2011124146A (en) * 2008-11-17 2012-12-27 Ф.Хоффманн-Ля Рош Аг COMPOSITIONS AND METHODS OF SUPPRESSING EXPRESSION OF FACTOR VII GENES
MX2011005429A (en) 2008-11-24 2011-06-21 Univ Northwestern Polyvalent rna-nanoparticle compositions.
WO2010061393A1 (en) 2008-11-30 2010-06-03 Compugen Ltd. He4 variant nucleotide and amino acid sequences, and methods of use thereof
US9394333B2 (en) 2008-12-02 2016-07-19 Wave Life Sciences Japan Method for the synthesis of phosphorus atom modified nucleic acids
WO2010065662A2 (en) 2008-12-04 2010-06-10 Curna, Inc. Treatment of sirtuin 1 (sirt1) related diseases by inhibition of natural antisense transcript to sirtuin 1
RU2746478C2 (en) 2008-12-04 2021-04-14 КьюРНА, Инк. Treatment of tumors of diseases related to the genom-suppressor by therapy of natural transcript inhibition in anti-significant orientation regarding this gene
KR101829469B1 (en) 2008-12-04 2018-03-30 큐알엔에이, 인크. Treatment of erythropoietin (epo) related diseases by inhibition of natural antisense transcript to epo
EP2633854B1 (en) 2008-12-05 2015-09-16 Yeda Research And Development Co. Ltd. miRNA-9 or miRNA-9* for use in treating ALS
JP5855462B2 (en) 2008-12-10 2016-02-09 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. DsRNA compositions targeting GNAQ and methods for inhibiting expression
CA2746508A1 (en) 2008-12-17 2010-07-15 Avi Biopharma, Inc. Antisense compositions and methods for modulating contact hypersensitivity or contact dermatitis
CA2749536A1 (en) * 2009-01-08 2010-07-15 Northwestern University Inhibition of bacterial protein production by polyvalent oligonucleotide modified nanoparticle conjugates
US20100233270A1 (en) 2009-01-08 2010-09-16 Northwestern University Delivery of Oligonucleotide-Functionalized Nanoparticles
KR101546673B1 (en) * 2009-01-15 2015-08-25 삼성전자주식회사 Toner for electrophotographic and process for preparing the same
WO2010088286A1 (en) * 2009-01-28 2010-08-05 Smartcells, Inc. Synthetic conjugates and uses thereof
WO2010088261A1 (en) * 2009-01-28 2010-08-05 Smartcells, Inc. Binding-site modified lectins and uses thereof
CA3036963A1 (en) 2009-01-29 2010-08-05 Arbutus Biopharma Corporation Lipid formulations comprising cationic lipid and a targeting lipid comprising n-acetyl galactosamine for delivery of nucleic acid
CN102307996A (en) * 2009-02-03 2012-01-04 弗·哈夫曼-拉罗切有限公司 Compositions and methods for inhibiting expression of PTP1B genes
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US8536320B2 (en) 2009-02-06 2013-09-17 Isis Pharmaceuticals, Inc. Tetrahydropyran nucleic acid analogs
US20120021515A1 (en) 2009-02-06 2012-01-26 Swayze Eric E Oligomeric compounds and methods
ES2658626T3 (en) 2009-02-12 2018-03-12 Curna, Inc. Treatment of diseases related to glial cell-derived neurotrophic factor (GDNF) by inhibition of natural antisense transcript to GDNF
KR101682735B1 (en) 2009-02-12 2016-12-06 큐알엔에이, 인크. Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
US20120041051A1 (en) 2009-02-26 2012-02-16 Kevin Fitzgerald Compositions And Methods For Inhibiting Expression Of MIG-12 Gene
EP2403863B1 (en) 2009-03-02 2013-08-28 Alnylam Pharmaceuticals Inc. Nucleic acid chemical modifications
JP6250263B2 (en) 2009-03-04 2017-12-20 クルナ・インコーポレーテッド Treatment of SIRT1-related diseases by suppression of natural antisense transcripts against sirtuin 1 (SIRT1)
EP2406376A1 (en) 2009-03-12 2012-01-18 Alnylam Pharmaceuticals, Inc. LIPID FORMULATED COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF Eg5 AND VEGF GENES
ES2656290T3 (en) 2009-03-16 2018-02-26 Curna, Inc. Treatment of diseases related to nuclear factor (derived from erythroid 2) similar to 2 (NRF2) by inhibition of natural antisense transcript to NRF2
EP2408920B1 (en) 2009-03-17 2017-03-08 CuRNA, Inc. Treatment of delta-like 1 homolog (dlk1) related diseases by inhibition of natural antisense transcript to dlk1
KR20120022938A (en) 2009-04-15 2012-03-12 노오쓰웨스턴 유니버시티 Delivery of oligonucleotide-functionalized nanoparticles
EP3524275A1 (en) 2009-04-22 2019-08-14 Massachusetts Institute Of Technology Innate immune supression enables repeated delivery of long rna molecules
CA2759899A1 (en) 2009-04-24 2010-10-28 Prosensa Technologies B.V. Oligonucleotide comprising an inosine for treating dmd
EP2424987B1 (en) 2009-05-01 2017-11-15 CuRNA, Inc. Treatment of hemoglobin (hbf/hbg) related diseases by inhibition of natural antisense transcript to hbf/hbg
CA2760706C (en) 2009-05-05 2019-08-06 Alnylam Pharmaceuticals, Inc. Methods of delivering oligonucleotides to immune cells
KR20220150411A (en) 2009-05-05 2022-11-10 알닐람 파마슈티칼스 인코포레이티드 Lipid compositions
JP5883782B2 (en) 2009-05-06 2016-03-15 クルナ・インコーポレーテッド Treatment of lipid transport metabolism gene-related diseases by suppression of natural antisense transcripts on lipid transport metabolism genes
WO2010129746A2 (en) 2009-05-06 2010-11-11 Curna, Inc. Treatment of tristetraproline (ttp) related diseases by inhibition of natural antisense transcript to ttp
WO2010132665A1 (en) 2009-05-15 2010-11-18 Yale University Gemm riboswitches, structure-based compound design with gemm riboswitches, and methods and compositions for use of and with gemm riboswitches
KR20120069610A (en) * 2009-05-15 2012-06-28 에프. 호프만-라 로슈 아게 Compositions and methods for inhibiting expression of glucocorticoid receptor (gcr) genes
DK2432881T3 (en) 2009-05-18 2018-02-26 Curna Inc TREATMENT OF REPROGRAMMING FACTOR-RELATED DISEASES BY INHIBITING NATURAL ANTISENSE TRANSCRIPTS TO A REPROGRAMMING FACTOR
KR101703695B1 (en) 2009-05-22 2017-02-08 큐알엔에이, 인크. Treatment of transcription factor e3 (tfe3) and insulin receptor substrate 2 (irs2) related diseases by inhibition of natural antisense transcript to tfe3
CN103221541B (en) 2009-05-28 2017-03-01 库尔纳公司 Antiviral gene relevant disease is treated by the natural antisense transcript suppressing antiviral gene
PL3431076T3 (en) 2009-06-10 2022-01-31 Arbutus Biopharma Corporation Improved lipid formulation
WO2010148050A2 (en) 2009-06-16 2010-12-23 Curna, Inc. Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene
JP6128846B2 (en) 2009-06-16 2017-05-17 クルナ・インコーポレーテッド Treatment of PON1 gene-related diseases by suppression of natural antisense transcripts against paraoxonase (PON1)
LT3449926T (en) 2009-06-17 2020-01-27 Biogen Ma Inc. Compositions and methods for modulation of smn2 splicing in a subject
US8859515B2 (en) 2009-06-24 2014-10-14 Curna, Inc. Treatment of tumor necrosis factor receptor 2 (TNFR2) related diseases by inhibition of natural antisense transcript to TNFR2
EP2446037B1 (en) 2009-06-26 2016-04-20 CuRNA, Inc. Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
WO2011005761A1 (en) 2009-07-06 2011-01-13 Ontorii, Inc Novel nucleic acid prodrugs and methods use thereof
WO2011005860A2 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. 5' phosphate mimics
WO2011005861A1 (en) 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. Oligonucleotide end caps
CA2769665A1 (en) 2009-08-05 2011-02-10 Opko Curna, Llc Treatment of insulin gene (ins) related diseases by inhibition of natural antisense transcript to an insulin gene (ins)
EP2462153B1 (en) 2009-08-06 2015-07-29 Isis Pharmaceuticals, Inc. Bicyclic cyclohexose nucleic acid analogs
US9029338B2 (en) 2009-08-14 2015-05-12 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of a gene from the ebola virus
US20120157324A1 (en) 2009-08-17 2012-06-21 Yale University Methylation biomarkers and methods of use
CN102482671B (en) 2009-08-25 2017-12-01 库尔纳公司 IQGAP relevant diseases are treated by suppressing the natural antisense transcript of ' gtpase activating protein containing IQ die bodys ' (IQGAP)
CA2772715C (en) 2009-09-02 2019-03-26 Genentech, Inc. Mutant smoothened and methods of using the same
CA2777448C (en) 2009-10-14 2023-02-28 Ilan Sela Compositions for controlling varroa mites in bees
US8962584B2 (en) 2009-10-14 2015-02-24 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Compositions for controlling Varroa mites in bees
WO2011050194A1 (en) 2009-10-22 2011-04-28 Genentech, Inc. Methods and compositions for modulating hepsin activation of macrophage-stimulating protein
US20110129832A1 (en) * 2009-10-27 2011-06-02 Swift Biosciences, Inc. Polynucleotide Primers and Probes
CA2779099C (en) 2009-10-30 2021-08-10 Northwestern University Templated nanoconjugates
WO2011056215A1 (en) 2009-11-03 2011-05-12 Landers James P Versatile, visible method for detecting polymeric analytes
WO2011054939A2 (en) 2009-11-09 2011-05-12 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of kif10 genes
WO2011058555A1 (en) 2009-11-12 2011-05-19 Yeda Research And Development Co. Ltd. A method of editing dna in a cell and constructs capable of same
JP5991922B2 (en) 2009-11-13 2016-09-14 サレプタ セラピューティクス, インコーポレイテッド Antisense antiviral compounds and methods for treating influenza virus infection
US8916362B2 (en) 2009-11-23 2014-12-23 Swift Biosciences, Inc. Devices comprising a polynucleotide to extend single stranded target molecules
TWI507524B (en) 2009-11-30 2015-11-11 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
EP3296398A1 (en) 2009-12-07 2018-03-21 Arbutus Biopharma Corporation Compositions for nucleic acid delivery
WO2011084455A2 (en) 2009-12-16 2011-07-14 Opko Curna, Llc. Treatment of membrane bound transcription factor peptidase, site 1 (mbtps1) related diseases by inhibition of natural antisense transcript to mbtps1
CA2784568A1 (en) 2009-12-18 2011-06-23 Martin A. Maier Lipid particles for delivery of nucleic acids
US20110152349A1 (en) * 2009-12-18 2011-06-23 Anke Geick Compositions and methods for inhibiting expression of il-18 genes
KR101891352B1 (en) 2009-12-23 2018-08-24 큐알엔에이, 인크. Treatment of hepatocyte growth factor (hgf) related diseases by inhibition of natural antisense transcript to hgf
RU2619185C2 (en) 2009-12-23 2017-05-12 Курна, Инк. Treatment of diseases associated with uncoupling proteins 2 (ucp2), by inhibiting of natural antisense transcript to ucp2
CA2785451C (en) 2009-12-24 2019-01-22 Prosensa Technologies B.V. Molecule for treating an inflammatory disorder
JP5982288B2 (en) 2009-12-29 2016-08-31 カッパーアールエヌエー,インコーポレイテッド Treatment of tumor protein 63-related diseases by inhibition of natural antisense transcripts against tumor protein 63 (p63)
EP2519633B1 (en) 2009-12-29 2017-10-25 CuRNA, Inc. Treatment of nuclear respiratory factor 1 (nrf1) related diseases by inhibition of natural antisense transcript to nrf1
NO2521784T3 (en) 2010-01-04 2018-05-05
EP2521785B1 (en) 2010-01-06 2022-03-09 CuRNA, Inc. Inhibition of natural antisense transcript to a pancreatic developmental gene for use in a treatment of pancreatic developmental gene related diseases
US8779118B2 (en) 2010-01-11 2014-07-15 Isis Pharmaceuticals, Inc. Base modified bicyclic nucleosides and oligomeric compounds prepared therefrom
ES2664866T3 (en) 2010-01-11 2018-04-23 Curna, Inc. Treatment of diseases related to sex hormone binding globulin (shbg) by inhibition of the natural antisense transcript to shbg
WO2011088076A2 (en) 2010-01-12 2011-07-21 Yale University Structured rna motifs and compounds and methods for their use
CA2786568A1 (en) 2010-01-25 2011-07-28 Curna, Inc. Treatment of rnase h1 related diseases by inhibition of natural antisense transcript to rnase h1
WO2011100131A2 (en) 2010-01-28 2011-08-18 Alnylam Pharmacuticals, Inc. Monomers and oligonucleotides comprising cycloaddition adduct(s)
WO2011094580A2 (en) 2010-01-28 2011-08-04 Alnylam Pharmaceuticals, Inc. Chelated copper for use in the preparation of conjugated oligonucleotides
WO2011097407A1 (en) 2010-02-04 2011-08-11 Ico Therapeutics Inc. Dosing regimens for treating and preventing ocular disorders using c-raf antisense
TW201129365A (en) * 2010-02-05 2011-09-01 Hoffmann La Roche Compositions and methods for inhibiting expression of IKK2 genes
JP5976548B2 (en) 2010-02-22 2016-08-23 カッパーアールエヌエー,インコーポレイテッド Treatment of pyrroline-5-carboxylate reductase 1 (PYCR1) related diseases by inhibition of natural antisense transcripts against PYCR1
WO2011105900A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-alpha (c8-alpha) and uses thereof
SG183335A1 (en) 2010-02-23 2012-09-27 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
WO2011105902A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 8-beta (c8-beta) and uses thereof
WO2011105901A2 (en) 2010-02-23 2011-09-01 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Antagonists of complement component 9 (c9) and uses thereof
WO2011112516A1 (en) 2010-03-08 2011-09-15 Ico Therapeutics Inc. Treating and preventing hepatitis c virus infection using c-raf kinase antisense oligonucleotides
AU2011224570B2 (en) 2010-03-08 2014-08-14 Monsanto Technology Llc Polynucleotide molecules for gene regulation in plants
US9068185B2 (en) 2010-03-12 2015-06-30 Sarepta Therapeutics, Inc. Antisense modulation of nuclear hormone receptors
US20130101512A1 (en) 2010-03-12 2013-04-25 Chad A. Mirkin Crosslinked polynucleotide structure
WO2011112732A2 (en) 2010-03-12 2011-09-15 The Brigham And Women's Hospital, Inc. Methods of treating vascular inflammatory disorders
US9193752B2 (en) 2010-03-17 2015-11-24 Isis Pharmaceuticals, Inc. 5′-substituted bicyclic nucleosides and oligomeric compounds prepared therefrom
US8889350B2 (en) 2010-03-26 2014-11-18 Swift Biosciences, Inc. Methods and compositions for isolating polynucleotides
WO2011123468A1 (en) 2010-03-29 2011-10-06 Alnylam Pharmaceuticals, Inc. Sirna therapy for transthyretin (ttr) related ocular amyloidosis
US9102938B2 (en) 2010-04-01 2015-08-11 Alnylam Pharmaceuticals, Inc. 2′ and 5′ modified monomers and oligonucleotides
WO2011127337A2 (en) 2010-04-09 2011-10-13 Opko Curna Llc Treatment of fibroblast growth factor 21 (fgf21) related diseases by inhibition of natural antisense transcript to fgf21
WO2011133695A2 (en) 2010-04-20 2011-10-27 Swift Biosciences, Inc. Materials and methods for nucleic acid fractionation by solid phase entrapment and enzyme-mediated detachment
WO2011133876A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
WO2011133868A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Conformationally restricted dinucleotide monomers and oligonucleotides
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
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
US9156873B2 (en) 2010-04-28 2015-10-13 Isis Pharmaceuticals, Inc. Modified 5′ diphosphate nucleosides and oligomeric compounds prepared therefrom
WO2011139911A2 (en) 2010-04-29 2011-11-10 Isis Pharmaceuticals, Inc. Lipid formulated single stranded rna
SI2563920T1 (en) 2010-04-29 2017-05-31 Ionis Pharmaceuticals, Inc. Modulation of transthyretin expression
SG185027A1 (en) 2010-05-03 2012-11-29 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
WO2011139387A1 (en) 2010-05-03 2011-11-10 Opko Curna, Llc Treatment of sirtuin (sirt) related diseases by inhibition of natural antisense transcript to a sirtuin (sirt)
CA3090304A1 (en) 2010-05-13 2011-11-17 Sarepta Therapeutics, Inc. Antisense modulation of interleukins 17 and 23 signaling
TWI586356B (en) 2010-05-14 2017-06-11 可娜公司 Treatment of par4 related diseases by inhibition of natural antisense transcript to par4
RU2585229C2 (en) 2010-05-26 2016-05-27 Курна, Инк. Treatment of diseases associated with atonal homolog 1 (aton1) by inhibiting natural antisense transcript of gene aton1
US20130203045A1 (en) 2010-05-26 2013-08-08 University Of Virginia Patent Foundation Method for detecting nucleic acids based on aggregate formation
CA2799501C (en) 2010-05-28 2022-02-15 Sarepta Therapeutics, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
CA3102008A1 (en) 2010-06-02 2011-12-08 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
WO2011156278A1 (en) 2010-06-07 2011-12-15 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
US8846637B2 (en) 2010-06-08 2014-09-30 Isis Pharmaceuticals, Inc. Substituted 2′-amino and 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
US9638632B2 (en) 2010-06-11 2017-05-02 Vanderbilt University Multiplexed interferometric detection system and method
WO2011163121A1 (en) 2010-06-21 2011-12-29 Alnylam Pharmaceuticals, Inc. Multifunctional copolymers for nucleic acid delivery
WO2011163466A1 (en) 2010-06-23 2011-12-29 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Regulation of skin pigmentation by neuregulin-1 (nrg-1)
US20130209463A1 (en) 2010-06-30 2013-08-15 Compugen Ltd. Polypeptides and uses thereof as a drug for treatment of multiple sclerosis, rheumatoid arthritis and other autoimmune disorders
US20130323269A1 (en) 2010-07-30 2013-12-05 Muthiah Manoharan Methods and compositions for delivery of active agents
US20130202652A1 (en) 2010-07-30 2013-08-08 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
WO2012021554A1 (en) 2010-08-09 2012-02-16 Yale University Cyclic di-gmp-ii riboswitches, motifs, and compounds, and methods for their use
US10017763B2 (en) 2010-09-03 2018-07-10 Sarepta Therapeutics, Inc. dsRNA molecules comprising oligonucleotide analogs having modified intersubunit linkages and/or terminal groups
US20130210901A1 (en) 2010-09-20 2013-08-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method of treating neurodegenerative diseases
WO2012039448A1 (en) 2010-09-24 2012-03-29 株式会社キラルジェン Asymmetric auxiliary group
SG10201508118WA (en) 2010-09-30 2015-11-27 Agency Science Tech & Res Methods and reagents for detection and treatment of esophageal metaplasia
JP5974012B2 (en) 2010-10-05 2016-08-23 ジェネンテック, インコーポレイテッド Mutant smoothened and method of using the same
ES2640755T3 (en) 2010-10-06 2017-11-06 Curna, Inc. Treatment of diseases related to Sialidase 4 (neu4) by inhibition of the natural antisense transcript to the neu4 gene
EP2625292B1 (en) 2010-10-07 2018-12-05 The General Hospital Corporation Biomarkers of cancer
WO2012052872A2 (en) 2010-10-17 2012-04-26 Yeda Research And Development Co. Ltd. Methods and compositions for the treatment of insulin-associated medical conditions
EP2851426B1 (en) 2010-10-18 2018-08-22 Arrowhead Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of RRM2 genes
WO2012054723A2 (en) 2010-10-22 2012-04-26 Opko Curna Llc Treatment of alpha-l-iduronidase (idua) related diseases by inhibition of natural antisense transcript to idua
RU2611195C2 (en) 2010-10-27 2017-02-21 Курна, Инк. Treatment of interferon-related developmental regulator 1 (ifrd1) associated diseases by inhibition of natural antisense transcript to ifrd1
CN110123830A (en) 2010-11-09 2019-08-16 阿尔尼拉姆医药品有限公司 Composition and method for inhibiting the lipid of the expression of Eg5 and VEGF gene to prepare
AU2011325956B2 (en) 2010-11-12 2016-07-14 The General Hospital Corporation Polycomb-associated non-coding RNAs
CA3077910A1 (en) 2010-11-17 2012-05-24 Ionis Pharmaceuticals, Inc. Modulation of alpha synuclein expression
CA2818824A1 (en) 2010-11-23 2012-05-31 Joseph Collard Treatment of nanog related diseases by inhibition of natural antisense transcript to nanog
US9150926B2 (en) 2010-12-06 2015-10-06 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Diagnosis and treatment of adrenocortical tumors using human microRNA-483
EP2649182A4 (en) 2010-12-10 2015-05-06 Alnylam Pharmaceuticals Inc Compositions and methods for increasing erythropoietin (epo) production
WO2012079046A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
AU2012207606B2 (en) 2011-01-11 2017-02-23 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
EP2663323B1 (en) 2011-01-14 2017-08-16 The General Hospital Corporation Methods targeting mir-128 for regulating cholesterol/lipid metabolism
KR101697396B1 (en) 2011-02-02 2017-01-17 엑스칼리아드 파마슈티컬즈, 인코포레이티드 Method of treating keloids or hypertrophic scars using antisense compounds targeting connective tissue growth factor (ctgf)
EP2670404B1 (en) 2011-02-02 2018-08-29 The Trustees of Princeton University Sirtuin modulators as virus production modulators
US20140044644A1 (en) 2011-02-21 2014-02-13 University Of Zurich Ankyrin g and modulators thereof for the treatment of neurodegenerative disorders
US9562853B2 (en) 2011-02-22 2017-02-07 Vanderbilt University Nonaqueous backscattering interferometric methods
SG193923A1 (en) 2011-03-29 2013-11-29 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of tmprss6 gene
WO2012138453A1 (en) 2011-04-03 2012-10-11 The General Hospital Corporation Efficient protein expression in vivo using modified rna (mod-rna)
EP2697256A1 (en) 2011-04-15 2014-02-19 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
WO2012149154A1 (en) 2011-04-26 2012-11-01 Swift Biosciences, Inc. Polynucleotide primers and probes
WO2012151268A1 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system for high throughput optical and label free detection of analytes
WO2012151289A2 (en) 2011-05-02 2012-11-08 University Of Virginia Patent Foundation Method and system to detect aggregate formation on a substrate
WO2012170347A1 (en) 2011-06-09 2012-12-13 Isis Pharmaceuticals, Inc. Bicyclic nucleosides and oligomeric compounds prepared therefrom
ES2653247T3 (en) 2011-06-09 2018-02-06 Curna, Inc. Treatment of frataxin-related diseases (FXN) by inhibiting the natural antisense transcript to the FXN gene
EP3388068A1 (en) 2011-06-21 2018-10-17 Alnylam Pharmaceuticals, Inc. Composition and methods for inhibition of expression of protein c (proc) genes
CN103890000B (en) 2011-06-21 2017-09-01 阿尔尼拉姆医药品有限公司 (ANGPTL3) the iRNA compositions of angiopoietin-like 3 and its application method
CA3191066A1 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes
EP4134433A1 (en) 2011-06-23 2023-02-15 Alnylam Pharmaceuticals, Inc. Serpina1 sirnas: compositions of matter and methods of treatment
TW202244278A (en) 2011-06-30 2022-11-16 美商艾羅海德製藥公司 Compositions and methods for inhibiting gene expression of hepatitis b virus
CA2836855C (en) 2011-06-30 2020-07-14 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
US20140227293A1 (en) 2011-06-30 2014-08-14 Trustees Of Boston University Method for controlling tumor growth, angiogenesis and metastasis using immunoglobulin containing and proline rich receptor-1 (igpr-1)
RU2014105311A (en) 2011-07-19 2015-08-27 Уэйв Лайф Сайенсес Пте. Лтд. METHODS FOR SYNTHESIS OF FUNCTIONALIZED NUCLEIC ACIDS
WO2013019857A2 (en) 2011-08-01 2013-02-07 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
BR112014002737B1 (en) 2011-08-04 2022-08-02 Yeda Research And Development Co. Ltd. MIR-135
US10806146B2 (en) 2011-09-13 2020-10-20 Monsanto Technology Llc Methods and compositions for weed control
CA2848695A1 (en) 2011-09-13 2013-03-21 Monsanto Technology Llc Methods and composition for weed control comprising inhibiting ppg oxidase
EP3382027A3 (en) 2011-09-13 2018-10-31 Monsanto Technology LLC Methods and compositions for weed control
US9840715B1 (en) 2011-09-13 2017-12-12 Monsanto Technology Llc Methods and compositions for delaying senescence and improving disease tolerance and yield in plants
US10829828B2 (en) 2011-09-13 2020-11-10 Monsanto Technology Llc Methods and compositions for weed control
US10760086B2 (en) 2011-09-13 2020-09-01 Monsanto Technology Llc Methods and compositions for weed control
EP2755466A4 (en) 2011-09-13 2015-04-15 Monsanto Technology Llc Methods and compositions for weed control
CN103957697B (en) 2011-09-13 2017-10-24 孟山都技术公司 Method and composition for Weeds distribution
EP2755692B1 (en) 2011-09-14 2020-11-25 Northwestern University Nanoconjugates able to cross the blood-brain barrier
US9920326B1 (en) 2011-09-14 2018-03-20 Monsanto Technology Llc Methods and compositions for increasing invertase activity in plants
AU2012308320C1 (en) 2011-09-14 2018-08-23 Translate Bio Ma, Inc. Multimeric oligonucleotide compounds
US9580713B2 (en) 2011-09-17 2017-02-28 Yale University Fluoride-responsive riboswitches, fluoride transporters, and methods of use
EP2760477B1 (en) 2011-09-27 2018-08-08 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
WO2013055865A1 (en) 2011-10-11 2013-04-18 The Brigham And Women's Hospital, Inc. Micrornas in neurodegenerative disorders
AU2012322618A1 (en) 2011-10-14 2014-05-29 Genentech, Inc. Anti-HtrA1 antibodies and methods of use
US9145559B2 (en) 2011-10-27 2015-09-29 Yeda Research And Development Co. Ltd. Methods of treating cancer
EP2780299A4 (en) 2011-11-18 2015-04-22 Sarepta Therapeutics Inc Functionally-modified oligonucleotides and subunits thereof
CA2895077A1 (en) 2011-12-12 2013-06-20 Beverly Packard In vivo delivery of oligonucleotides
WO2013090648A1 (en) 2011-12-16 2013-06-20 modeRNA Therapeutics Modified nucleoside, nucleotide, and nucleic acid compositions
CA2860731C (en) 2012-01-10 2023-02-28 M. Mahmood Hussain Method of treating hyperlipidemia and atherosclerosis with mir-30c
WO2013112053A1 (en) 2012-01-27 2013-08-01 Prosensa Technologies B.V. Rna modulating oligonucleotides with improved characteristics for the treatment of duchenne and becker muscular dystrophy
JP2015512616A (en) 2012-02-01 2015-04-30 コンピュゲン エルティーディー. C1ORF32 antibody and its use for the treatment of cancer
WO2013121426A1 (en) 2012-02-13 2013-08-22 Gamida-Cell Ltd. Culturing of mesenchymal stem cells
JP2015509366A (en) 2012-02-22 2015-03-30 ブレインステム バイオテック リミテッド Generation of neural stem cells and motor neurons
US10034902B2 (en) 2012-02-22 2018-07-31 Exostem Biotec Ltd. MicroRNAs for the generation of astrocytes
CA2866625C (en) 2012-03-13 2020-12-08 Swift Biosciences, Inc. Methods and compositions for size-controlled homopolymer tailing of substrate polynucleotides by a nucleic acid polymerase
EP2639238A1 (en) 2012-03-15 2013-09-18 Universität Bern Tricyclic nucleosides and oligomeric compounds prepared therefrom
CA2867262C (en) 2012-03-15 2021-03-16 Curna, Inc. Treatment of brain derived neurotrophic factor (bdnf) related diseases by inhibition of natural antisense transcript to bdnf
CN108949772A (en) 2012-04-02 2018-12-07 现代泰克斯公司 For generating the modification polynucleotides of biological agent relevant to human diseases and protein
AU2013243948A1 (en) 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with human disease
WO2013154799A1 (en) 2012-04-09 2013-10-17 Isis Pharmaceuticals, Inc. Tricyclic nucleosides and oligomeric compounds prepared therefrom
WO2013154798A1 (en) 2012-04-09 2013-10-17 Isis Pharmaceuticals, Inc. Tricyclic nucleic acid analogs
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
WO2013159091A2 (en) 2012-04-20 2013-10-24 Aptamir Therapeutics, Inc. Mirna modulators of thermogenesis
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
WO2013163628A2 (en) 2012-04-27 2013-10-31 Duke University Genetic correction of mutated genes
US9273949B2 (en) 2012-05-11 2016-03-01 Vanderbilt University Backscattering interferometric methods
EP2850186B1 (en) 2012-05-16 2018-12-19 Translate Bio MA, Inc. Compositions and methods for modulating smn gene family expression
EP2850189B8 (en) 2012-05-16 2019-01-23 Translate Bio MA, Inc. Compositions and methods for modulating gene expression
AU2013264742B2 (en) 2012-05-24 2018-10-04 A.B. Seeds Ltd. Compositions and methods for silencing gene expression
WO2013184209A1 (en) 2012-06-04 2013-12-12 Ludwig Institute For Cancer Research Ltd. Mif for use in methods of treating subjects with a neurodegenerative disorder
CN104684893B (en) 2012-07-13 2016-10-26 日本波涛生命科学公司 Asymmetric auxiliary group
US20140038182A1 (en) 2012-07-17 2014-02-06 Dna Logix, Inc. Cooperative primers, probes, and applications thereof
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
CA2880833A1 (en) 2012-08-03 2014-02-06 Aptamir Therapeutics, Inc. Cell-specific delivery of mirna modulators for the treatment of obesity and related disorders
WO2014028739A1 (en) 2012-08-15 2014-02-20 Isis Pharmaceuticals, Inc. Method of preparing oligomeric compounds using modified capping protocols
CN104781271B (en) 2012-08-20 2018-07-06 加利福尼亚大学董事会 The polynucleotides of group with bio-reversible
US9029335B2 (en) 2012-10-16 2015-05-12 Isis Pharmaceuticals, Inc. Substituted 2′-thio-bicyclic nucleosides and oligomeric compounds prepared therefrom
WO2014062989A2 (en) 2012-10-18 2014-04-24 Monsanto Technology Llc Methods and compositions for plant pest control
CA2890207A1 (en) 2012-11-05 2014-05-08 Foundation Medicine, Inc. Novel ntrk1 fusion molecules and uses thereof
EP2920307B1 (en) 2012-11-15 2018-05-02 Roche Innovation Center Copenhagen A/S Anti apob antisense conjugate compounds
ES2921623T3 (en) 2012-11-26 2022-08-30 Modernatx Inc terminally modified RNA
BR112015015976A2 (en) 2013-01-01 2018-04-10 A. B. Seeds Ltd. method for introducing dsrna into seeds to modulate gene expression
US10683505B2 (en) 2013-01-01 2020-06-16 Monsanto Technology Llc Methods of introducing dsRNA to plant seeds for modulating gene expression
WO2014113089A2 (en) 2013-01-17 2014-07-24 Moderna Therapeutics, Inc. Signal-sensor polynucleotides for the alteration of cellular phenotypes
CA2898326C (en) 2013-01-18 2022-05-17 Foundation Medicine, Inc. Methods of treating cholangiocarcinoma
US10000767B2 (en) 2013-01-28 2018-06-19 Monsanto Technology Llc Methods and compositions for plant pest control
RU2649367C2 (en) 2013-01-30 2018-04-02 Ф. Хоффманн-Ля Рош Аг Lna oligonucleotide carbohydrate conjugates
WO2014118272A1 (en) 2013-01-30 2014-08-07 Santaris Pharma A/S Antimir-122 oligonucleotide carbohydrate conjugates
US9701708B2 (en) 2013-01-31 2017-07-11 Ionis Pharmaceuticals, Inc. Method of preparing oligomeric compounds using modified coupling protocols
US20150366890A1 (en) 2013-02-25 2015-12-24 Trustees Of Boston University Compositions and methods for treating fungal infections
MX2015012334A (en) 2013-03-13 2016-02-05 Monsanto Technology Llc Methods and compositions for weed control.
WO2014164761A1 (en) 2013-03-13 2014-10-09 Monsanto Technology Llc Methods and compositions for weed control
US20160024181A1 (en) 2013-03-13 2016-01-28 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
EP2971010B1 (en) 2013-03-14 2020-06-10 ModernaTX, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
EP2970970B1 (en) 2013-03-14 2018-12-12 Andes Biotechnologies Global, Inc. Antisense oligonucleotides for treatment of cancer stem cells
US20140283211A1 (en) 2013-03-14 2014-09-18 Monsanto Technology Llc Methods and Compositions for Plant Pest Control
PE20160046A1 (en) 2013-03-14 2016-02-14 Alnylam Pharmaceuticals Inc COMPOSITION OF RNAi AGAINST THE C5 COMPONENT OF THE COMPLEMENT AND METHODS FOR ITS USE
CN105283466A (en) 2013-03-14 2016-01-27 安第斯生物技术股份有限公司 Methods for detecting and treating multiple myeloma
US10568328B2 (en) 2013-03-15 2020-02-25 Monsanto Technology Llc Methods and compositions for weed control
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
EP3708184A1 (en) 2013-03-27 2020-09-16 The General Hospital Corporation Methods and agents for treating alzheimer s disease
EP4286517A3 (en) 2013-04-04 2024-03-13 President and Fellows of Harvard College Therapeutic uses of genome editing with crispr/cas systems
WO2014172698A1 (en) 2013-04-19 2014-10-23 Isis Pharmaceuticals, Inc. Compositions and methods for modulation nucleic acids through nonsense mediated decay
BR112015027377B1 (en) 2013-05-01 2023-01-10 Ionis Pharmaceuticals, Inc OLIGOMERIC COMPOUNDS WITH CONJUGATE GROUPS, COMPOSITIONS COMPRISING THE SAID COMPOUNDS AND USES THEREOF
SG11201510565TA (en) 2013-05-22 2016-01-28 Alnylam Pharmaceuticals Inc Tmprss6 irna compositions and methods of use thereof
SG10201913872XA (en) 2013-05-22 2020-03-30 Alnylam Pharmaceuticals Inc SERPINA1 iRNA COMPOSITIONS AND METHODS OF USE THEREOF
US20160113911A1 (en) 2013-06-06 2016-04-28 The General Hospital Corporation Methods and compositions for the treatment of cancer
EP3007704B1 (en) 2013-06-13 2021-01-06 Antisense Therapeutics Ltd Combination therapy for acromegaly
PL3013959T3 (en) 2013-06-27 2020-05-18 Roche Innovation Center Copenhagen A/S Antisense oligomers and conjugates targeting pcsk9
HUE056760T2 (en) 2013-07-11 2022-03-28 Modernatx Inc Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
US9850496B2 (en) 2013-07-19 2017-12-26 Monsanto Technology Llc Compositions and methods for controlling Leptinotarsa
EP3030663B1 (en) 2013-07-19 2019-09-04 Monsanto Technology LLC Compositions and methods for controlling leptinotarsa
US10894963B2 (en) 2013-07-25 2021-01-19 Exicure, Inc. Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
RS59991B1 (en) 2013-08-08 2020-04-30 Scripps Research Inst A method for the site-specific enzymatic labelling of nucleic acids in vitro by incorporation of unnatural nucleotides
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
AU2014315287A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
EP3052626A1 (en) 2013-10-02 2016-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
EA201690675A1 (en) 2013-10-03 2016-08-31 Модерна Терапьютикс, Инк. POLYNUCLEOTES ENCODING THE RECEPTOR OF LOW DENSITY LIPOPROTEINS
WO2015051366A2 (en) 2013-10-04 2015-04-09 Novartis Ag Novel formats for organic compounds for use in rna interference
CA2925129C (en) 2013-10-04 2023-04-04 Novartis Ag 3' end caps for rnai agents for use in rna interference
TW202310853A (en) 2013-10-04 2023-03-16 美國西奈山伊坎醫學院 Compositions and methods for inhibiting expression of the alas1 gene
US10519446B2 (en) 2013-10-04 2019-12-31 Novartis Ag Organic compounds to treat hepatitis B virus
EP3055426B1 (en) 2013-10-09 2019-06-19 The United States of America as represented by The Secretary Department of Health and Human Services Detection of hepatitis delta virus (hdv) for the diagnosis and treatment of sjögren's syndrome and lymphoma
US11162096B2 (en) 2013-10-14 2021-11-02 Ionis Pharmaceuticals, Inc Methods for modulating expression of C9ORF72 antisense transcript
US9758546B2 (en) 2013-10-21 2017-09-12 Ionis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
CA2928779A1 (en) 2013-10-21 2015-04-30 The General Hospital Corporation Methods relating to circulating tumor cell clusters and the treatment of cancer
EP3066200A1 (en) 2013-11-04 2016-09-14 Monsanto Technology LLC Compositions and methods for controlling arthropod parasite and pest infestations
US10301622B2 (en) 2013-11-04 2019-05-28 Northwestern University Quantification and spatio-temporal tracking of a target using a spherical nucleic acid (SNA)
SI3071696T1 (en) 2013-11-22 2019-11-29 Mina Therapeutics Ltd C/ebp alpha short activating rna compositions and methods of use
CN112107693B (en) 2013-12-03 2023-05-26 西北大学 Liposome particles, method for preparing said liposome particles and use thereof
CA2844640A1 (en) 2013-12-06 2015-06-06 The University Of British Columbia Method for treatment of castration-resistant prostate cancer
US10385388B2 (en) 2013-12-06 2019-08-20 Swift Biosciences, Inc. Cleavable competitor polynucleotides
UA119253C2 (en) 2013-12-10 2019-05-27 Біолоджикс, Інк. Compositions and methods for virus control in varroa mite and bees
CA2931090A1 (en) 2013-12-12 2015-06-18 Alnylam Pharmaceuticals, Inc. Complement component irna compositions and methods of use thereof
AU2014364520B2 (en) 2013-12-20 2020-01-02 The General Hospital Corporation Methods and assays relating to circulating tumor cells
UA121462C2 (en) 2014-01-15 2020-06-10 Монсанто Текнолоджі Елелсі Methods and compositions for weed control using epsps polynucleotides
ES2917473T3 (en) 2014-01-16 2022-07-08 Wave Life Sciences Ltd chiral design
AU2015214264B2 (en) 2014-02-04 2018-12-20 Curis, Inc. Mutant Smoothened and methods of using the same
CA2936158C (en) 2014-02-05 2023-06-13 Yeda Research And Development Co. Ltd. Use of mir-135 or precursor thereof for the treatment and diagnosis of a bipolar disease
WO2015123264A1 (en) 2014-02-11 2015-08-20 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
US10036019B2 (en) 2014-03-17 2018-07-31 Ionis Pharmaceuticals, Inc. Bicyclic carbocyclic nucleosides and oligomeric compounds prepared therefrom
US10006027B2 (en) 2014-03-19 2018-06-26 Ionis Pharmaceuticals, Inc. Methods for modulating Ataxin 2 expression
MX2016012123A (en) 2014-03-19 2017-01-19 Ionis Pharmaceuticals Inc Compositions for modulating ataxin 2 expression.
ES2926869T3 (en) 2014-04-01 2022-10-31 Biogen Ma Inc Compositions to modulate the expression of SOD-1
US11091770B2 (en) 2014-04-01 2021-08-17 Monsanto Technology Llc Compositions and methods for controlling insect pests
TWI638047B (en) 2014-04-09 2018-10-11 史基普研究協會 Import of unnatural or modified nucleoside triphosphates into cells via nucleic acid triphosphate transporters
ES2932304T3 (en) 2014-04-17 2023-01-17 Biogen Ma Inc Compositions and methods for modulation of SMN2 splicing in a subject
US10221416B2 (en) 2014-04-24 2019-03-05 Ionis Pharmaceuticals, Inc. Oligomeric compounds comprising alpha-beta-constrained nucleic acid
EP3137115B1 (en) 2014-05-01 2020-10-14 Ionis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
CA2943894A1 (en) 2014-05-01 2015-11-05 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating complement factor b expression
WO2015171918A2 (en) 2014-05-07 2015-11-12 Louisiana State University And Agricultural And Mechanical College Compositions and uses for treatment thereof
TW201607559A (en) 2014-05-12 2016-03-01 阿尼拉製藥公司 Methods and compositions for treating a SERPINC1-associated disorder
GB201408623D0 (en) 2014-05-15 2014-07-02 Santaris Pharma As Oligomers and oligomer conjugates
EA201692370A1 (en) 2014-05-22 2017-03-31 Элнилэм Фармасьютикалз, Инк. COMPOSITIONS of mRNA ANGIOTENZINOGENA (AGT) AND METHODS OF THEIR USE
US10456445B2 (en) 2014-06-02 2019-10-29 Children's Medical Center Corporation Methods and compositions for immunomodulation
WO2015190922A1 (en) 2014-06-10 2015-12-17 Erasmus University Medical Center Rotterdam Antisense oligonucleotides useful in treatment of pompe disease
AU2015280252A1 (en) 2014-06-23 2017-01-12 Monsanto Technology Llc Compositions and methods for regulating gene expression via RNA interference
US11807857B2 (en) 2014-06-25 2023-11-07 Monsanto Technology Llc Methods and compositions for delivering nucleic acids to plant cells and regulating gene expression
WO2015200697A1 (en) 2014-06-25 2015-12-30 The General Hospital Corporation Targeting human satellite ii (hsatii)
AU2015291151B2 (en) 2014-07-15 2019-10-10 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Isolated polypeptides of CD44 and uses thereof
JP2017524357A (en) 2014-07-16 2017-08-31 モデルナティエックス インコーポレイテッドModernaTX,Inc. Chimeric polynucleotide
US9951327B1 (en) 2014-07-17 2018-04-24 Integrated Dna Technologies, Inc. Efficient and rapid method for assembling and cloning double-stranded DNA fragments
EP3171895A1 (en) 2014-07-23 2017-05-31 Modernatx, Inc. Modified polynucleotides for the production of intrabodies
CA2955842A1 (en) 2014-07-29 2016-02-04 Monsanto Technology Llc Compositions and methods for controlling insect pests
MX2017001432A (en) 2014-07-31 2017-05-09 Uab Res Found Apoe mimetic peptides and higher potency to clear plasma cholesterol.
CA2958431A1 (en) 2014-08-19 2016-02-25 Northwestern University Protein/oligonucleotide core-shell nanoparticle therapeutics
WO2016030899A1 (en) 2014-08-28 2016-03-03 Yeda Research And Development Co. Ltd. Methods of treating amyotrophic lateral scleroses
WO2016033424A1 (en) 2014-08-29 2016-03-03 Genzyme Corporation Methods for the prevention and treatment of major adverse cardiovascular events using compounds that modulate apolipoprotein b
JP2017526367A (en) 2014-08-29 2017-09-14 チルドレンズ メディカル センター コーポレーション Methods and compositions for the treatment of cancer
ES2928500T3 (en) 2014-08-29 2022-11-18 Alnylam Pharmaceuticals Inc Patisiran for use in the treatment of transthyretin-mediated amyloidosis
WO2016040589A1 (en) 2014-09-12 2016-03-17 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
EP3194581A4 (en) 2014-09-15 2018-04-25 Children's Medical Center Corporation Methods and compositions to increase somatic cell nuclear transfer (scnt) efficiency by removing histone h3-lysine trimethylation
US10556020B2 (en) 2014-09-26 2020-02-11 University Of Massachusetts RNA-modulating agents
JOP20200115A1 (en) 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
WO2016061487A1 (en) 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
WO2016069694A2 (en) 2014-10-30 2016-05-06 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
CN113846101A (en) 2014-11-17 2021-12-28 阿尔尼拉姆医药品有限公司 Apolipoprotein C3(APOC3) iRNA compositions and methods of use thereof
CN107106493A (en) 2014-11-21 2017-08-29 西北大学 The sequence-specific cellular uptake of spherical nucleic acid nano particle conjugate
AU2015360794B2 (en) 2014-12-08 2021-07-08 The Board Of Regents Of The University Of Texas System Lipocationic polymers and uses thereof
CA2969464A1 (en) 2014-12-12 2016-06-16 Tod M. Woolf Compositions and methods for editing nucleic acids in cells utilizing oligonucleotides
WO2016100716A1 (en) 2014-12-18 2016-06-23 Vasant Jadhav Reversirtm compounds
US9688707B2 (en) 2014-12-30 2017-06-27 Ionis Pharmaceuticals, Inc. Bicyclic morpholino compounds and oligomeric compounds prepared therefrom
WO2016112132A1 (en) 2015-01-06 2016-07-14 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of c9orf72 antisense transcript
US10538763B2 (en) 2015-01-16 2020-01-21 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of DUX4
PL3256589T3 (en) 2015-01-22 2022-02-21 Monsanto Technology Llc Compositions and methods for controlling leptinotarsa
JP2018506715A (en) 2015-01-23 2018-03-08 ヴァンダービルト ユニバーシティー Robust interferometer and method of use
US10676726B2 (en) 2015-02-09 2020-06-09 Duke University Compositions and methods for epigenome editing
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
US10450342B2 (en) 2015-02-23 2019-10-22 Ionis Pharmaceuticals, Inc. Method for solution phase detritylation of oligomeric compounds
WO2016135559A2 (en) 2015-02-23 2016-09-01 Crispr Therapeutics Ag Materials and methods for treatment of human genetic diseases including hemoglobinopathies
US11129844B2 (en) 2015-03-03 2021-09-28 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating MECP2 expression
EP3268475B1 (en) 2015-03-11 2020-10-21 Yissum Research and Development Company of the Hebrew University of Jerusalem Ltd. Decoy oligonucleotides for the treatment of diseases
CA2981068C (en) 2015-03-26 2021-12-14 Women & Infants Hospital Of Rhode Island Therapy for malignant disease comprising the inhibition of human epididymal secretory protein e4 and immune checkpoint inhibitors
EP4218771A1 (en) 2015-03-27 2023-08-02 Yeda Research and Development Co. Ltd Methods of treating motor neuron diseases
BR112017020750A2 (en) 2015-03-27 2018-06-26 Harvard College modified t-cells and methods of producing and using them
US10961532B2 (en) 2015-04-07 2021-03-30 The General Hospital Corporation Methods for reactivating genes on the inactive X chromosome
US10745702B2 (en) 2015-04-08 2020-08-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the LECT2 gene
US10407678B2 (en) 2015-04-16 2019-09-10 Ionis Pharmaceuticals, Inc. Compositions for modulating expression of C9ORF72 antisense transcript
EP3291677A4 (en) 2015-05-04 2019-02-13 Monsanto Technology LLC Compositions and methods for controlling arthropod parasite and pest infestations
WO2016181393A1 (en) 2015-05-11 2016-11-17 Yeda Research And Development Co. Ltd. Citrin inhibitors for the treatment of cancer
EP3302053B1 (en) 2015-06-02 2021-03-17 Monsanto Technology LLC Compositions and methods for delivery of a polynucleotide into a plant
EP3302030A4 (en) 2015-06-03 2019-04-24 Monsanto Technology LLC Methods and compositions for introducing nucleic acids into plants
WO2016196897A1 (en) 2015-06-04 2016-12-08 Sarepta Therapeutics, Inc. Methods and compounds for treatment of lymphocyte-related diseases and conditions
EP3307316A1 (en) 2015-06-12 2018-04-18 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
US20180188257A1 (en) 2015-06-19 2018-07-05 University Of Rochester Septin proteins as novel biomarkers for detection and treatment of müllerian cancers
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
CA2990852A1 (en) 2015-06-26 2016-12-29 Beth Israel Deaconess Medical Center, Inc. Cancer therapy targeting tetraspanin 33 (tspan33) in myeloid derived suppressor cells
EP3314027A4 (en) 2015-06-29 2019-07-03 Caris Science, Inc. Therapeutic oligonucleotides
WO2017011286A1 (en) 2015-07-10 2017-01-19 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (igfals) and insulin-like growth factor 1 (igf-1) irna compositions and methods of use thereof
CA2989970A1 (en) 2015-07-17 2017-01-26 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
CA2993652A1 (en) 2015-07-28 2017-02-02 Caris Science, Inc. Targeted oligonucleotides
WO2017021963A1 (en) 2015-08-03 2017-02-09 Biokine Therapeutics Ltd. Cxcr4 binding agents for treatment of diseases
EP3332009A1 (en) 2015-08-04 2018-06-13 Yeda Research and Development Co., Ltd. Methods of screening for riboswitches and attenuators
CN108271387B (en) 2015-08-07 2023-06-27 箭头药业股份有限公司 RNAi therapy of hepatitis B virus infection
US10889813B2 (en) 2015-09-02 2021-01-12 Alnylam Pharmaceuticals, Inc. Programmed cell death 1 ligand 1 (PD-L1) iRNA compositions and methods of use thereof
PT3349802T (en) 2015-09-14 2021-10-15 Univ Texas Lipocationic dendrimers and uses thereof
CA2998287A1 (en) 2015-09-24 2017-04-20 Crispr Therapeutics Ag Novel family of rna-programmable endonucleases and their uses in genome editing and other applications
EP4285912A2 (en) 2015-09-25 2023-12-06 Ionis Pharmaceuticals, Inc. Compositions and methods for modulating ataxin 3 expression
US11905526B2 (en) 2015-10-13 2024-02-20 Duke University Genome engineering with Type I CRISPR systems in eukaryotic cells
US11369692B2 (en) 2015-10-28 2022-06-28 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of Duchenne Muscular Dystrophy
CA3001362C (en) 2015-10-30 2020-10-13 Genentech, Inc. Anti-htra1 antibodies and methods of use thereof
AU2016349954B2 (en) 2015-11-05 2022-08-25 Antisense Therapeutics Ltd Mobilizing leukemia cells
CN109328231A (en) 2015-11-06 2019-02-12 克里斯普治疗股份公司 For treating the material and method of 1A type glycogen storage disease
EP3373939A4 (en) 2015-11-10 2019-06-26 B.G. Negev Technologies and Applications Ltd., at Ben-Gurion University Means and methods for reducing tumorigenicity of cancer stem cells
CA3005878A1 (en) 2015-11-19 2017-05-26 The Brigham And Women's Hospital, Inc. Lymphocyte antigen cd5-like (cd5l)-interleukin 12b (p40) heterodimers in immunity
US20170145394A1 (en) 2015-11-23 2017-05-25 The Regents Of The University Of California Tracking and manipulating cellular rna via nuclear delivery of crispr/cas9
EP3967758A1 (en) 2015-12-01 2022-03-16 CRISPR Therapeutics AG Materials and methods for treatment of alpha-1 antitrypsin deficiency
WO2017096395A1 (en) 2015-12-04 2017-06-08 Ionis Pharmaceuticals, Inc. Methods of treating breast cancer
EP3387127A1 (en) 2015-12-07 2018-10-17 Erasmus University Medical Center Rotterdam Enzymatic replacement therapy and antisense therapy for pompe disease
WO2017106767A1 (en) 2015-12-18 2017-06-22 The Scripps Research Institute Production of unnatural nucleotides using a crispr/cas9 system
WO2017109757A1 (en) 2015-12-23 2017-06-29 Crispr Therapeutics Ag Materials and methods for treatment of amyotrophic lateral sclerosis and/or frontal temporal lobular degeneration
EP3400300A4 (en) 2016-01-05 2019-08-07 Ionis Pharmaceuticals, Inc. Methods for reducing lrrk2 expression
US10627396B2 (en) 2016-01-29 2020-04-21 Vanderbilt University Free-solution response function interferometry
US20190038771A1 (en) 2016-02-02 2019-02-07 Crispr Therapeutics Ag Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
CN109071625A (en) 2016-02-04 2018-12-21 柯瑞斯公司 Smooth mutant and its application method
EP3416689B1 (en) 2016-02-18 2023-01-18 CRISPR Therapeutics AG Materials and methods for treatment of severe combined immunodeficiency (scid) or omenn syndrome
JP7033072B2 (en) 2016-02-25 2022-03-09 ザ ブリガム アンド ウィメンズ ホスピタル インコーポレイテッド Treatment for fibrosis targeting SMOC2
EP3423581A4 (en) 2016-03-04 2020-03-04 Rhode Island Hospital Targeting microrna for cancer treatment
PT3430141T (en) 2016-03-14 2021-02-25 Hoffmann La Roche Oligonucleotides for reduction of pd-l1 expression
US11083799B2 (en) 2016-03-16 2021-08-10 Crispr Therapeutics Ag Materials and methods for treatment of hereditary haemochromatosis
WO2017161168A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Modulation of dyrk1b expression
WO2017161172A1 (en) 2016-03-16 2017-09-21 Ionis Pharmaceuticals, Inc. Methods of modulating keap1
IL303936A (en) 2016-03-18 2023-08-01 Caris Science Inc Oligonucleotide probes and uses thereof
EP3445388A2 (en) 2016-04-18 2019-02-27 CRISPR Therapeutics AG Materials and methods for treatment of hemoglobinopathies
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
WO2017191503A1 (en) 2016-05-05 2017-11-09 Crispr Therapeutics Ag Materials and methods for treatment of hemoglobinopathies
JP7080826B2 (en) 2016-05-16 2022-06-06 ザ ボード オブ リージェンツ オブ ザ ユニバーシティー オブ テキサス システム Cationic sulfonamide aminolipids and amphoteric zwitterionic aminolipids
IL298701B2 (en) 2016-05-25 2024-03-01 Caris Science Inc Oligonucleotide probes and uses thereof
US20190256845A1 (en) 2016-06-10 2019-08-22 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT C5 iRNA COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)
EP3471781A4 (en) 2016-06-17 2020-05-06 Ionis Pharmaceuticals, Inc. Modulation of gys1 expression
WO2017223528A1 (en) 2016-06-24 2017-12-28 The Scripps Research Institute Novel nucleoside triphosphate transporter and uses thereof
EP3478313B1 (en) 2016-06-29 2022-05-04 CRISPR Therapeutics AG Materials and methods for treatment of amyotrophic lateral sclerosis (als) and other related disorders
WO2018002812A1 (en) 2016-06-29 2018-01-04 Crispr Therapeutics Ag Materials and methods for treatment of myotonic dystrophy type 1 (dm1) and other related disorders
AU2017290614C1 (en) 2016-06-29 2024-01-18 Crispr Therapeutics Ag Materials and methods for treatment of friedreich ataxia and other related disorders
AU2017292173B2 (en) 2016-07-06 2022-01-13 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of pain related disorders
EP3481856A1 (en) 2016-07-06 2019-05-15 Crispr Therapeutics AG Materials and methods for treatment of pain related disorders
WO2018007871A1 (en) 2016-07-08 2018-01-11 Crispr Therapeutics Ag Materials and methods for treatment of transthyretin amyloidosis
US11253601B2 (en) 2016-07-11 2022-02-22 Translate Bio Ma, Inc. Nucleic acid conjugates and uses thereof
KR20230088522A (en) 2016-07-21 2023-06-19 맥스시티 인코포레이티드 Methods and compositions for modifying genomic dna
WO2018020323A2 (en) 2016-07-25 2018-02-01 Crispr Therapeutics Ag Materials and methods for treatment of fatty acid disorders
JOP20170161A1 (en) 2016-08-04 2019-01-30 Arrowhead Pharmaceuticals Inc RNAi Agents for Hepatitis B Virus Infection
NL2017295B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Antisense oligomeric compound for Pompe disease
NL2017294B1 (en) 2016-08-05 2018-02-14 Univ Erasmus Med Ct Rotterdam Natural cryptic exon removal by pairs of antisense oligonucleotides.
WO2018039629A2 (en) 2016-08-25 2018-03-01 Northwestern University Micellar spherical nucleic acids from thermoresponsive, traceless templates
MX2019002339A (en) 2016-09-02 2019-05-16 Dicerna Pharmaceuticals Inc 4'-phosphate analogs and oligonucleotides comprising the same.
US11400161B2 (en) 2016-10-06 2022-08-02 Ionis Pharmaceuticals, Inc. Method of conjugating oligomeric compounds
WO2018081817A2 (en) 2016-10-31 2018-05-03 University Of Massachusetts Targeting microrna-101-3p in cancer therapy
JOP20190104A1 (en) 2016-11-10 2019-05-07 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn3 expression
TW202313978A (en) 2016-11-23 2023-04-01 美商阿尼拉製藥公司 Serpina1 irna compositions and methods of use thereof
EP3330276A1 (en) 2016-11-30 2018-06-06 Universität Bern Novel bicyclic nucleosides and oligomers prepared therefrom
US11033570B2 (en) 2016-12-02 2021-06-15 Cold Spring Harbor Laboratory Modulation of Lnc05 expression
AU2017378153B2 (en) 2016-12-13 2024-03-28 Seattle Children's Hospital (dba Seattle Children's Research Institute) Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo
TWI790217B (en) 2016-12-16 2023-01-21 美商阿尼拉製藥公司 METHODS FOR TREATING OR PREVENTING TTR-ASSOCIATED DISEASES USING TRANSTHYRETIN (TTR) iRNA COMPOSITIONS
US11197928B2 (en) 2017-01-13 2021-12-14 Board Of Regents, The University Of Texas System Sustained production of high affinity antigen specific antibody by high dose BAFF receptor-targeting mAb-siRNA conjugate
RU2021133626A (en) 2017-01-23 2022-01-31 Регенерон Фармасьютикалз, Инк. HSD17B13 OPTIONS AND THEIR APPLICATIONS
WO2018154387A1 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Compositions and methods for gene editing
US20200216857A1 (en) 2017-02-22 2020-07-09 Crispr Therapeutics Ag Materials and methods for treatment of spinocerebellar ataxia type 2 (sca2) and other spinocerebellar ataxia type 2 protein (atxn2) gene related conditions or disorders
EP3585899A1 (en) 2017-02-22 2020-01-01 CRISPR Therapeutics AG Materials and methods for treatment of primary hyperoxaluria type 1 (ph1) and other alanine-glyoxylate aminotransferase (agxt) gene related conditions or disorders
US20200040061A1 (en) 2017-02-22 2020-02-06 Crispr Therapeutics Ag Materials and methods for treatment of early onset parkinson's disease (park1) and other synuclein, alpha (snca) gene related conditions or disorders
WO2018154439A1 (en) 2017-02-22 2018-08-30 Crispr Therapeutics Ag Materials and methods for treatment of spinocerebellar ataxia type 1 (sca1) and other spinocerebellar ataxia type 1 protein (atxn1) gene related conditions or disorders
WO2018165564A1 (en) 2017-03-09 2018-09-13 Ionis Pharmaceuticals, Inc. Morpholino modified oligomeric compounds
US20180284123A1 (en) 2017-03-30 2018-10-04 California Institute Of Technology Barcoded rapid assay platform useful for efficient analysis of candidate molecules and methods of making and using the platform
TWI801377B (en) 2017-04-18 2023-05-11 美商阿尼拉製藥公司 Methods for the treatment of subjects having a hepatitis b virus (hbv) infection
WO2018193428A1 (en) 2017-04-20 2018-10-25 Synthena Ag Modified oligomeric compounds comprising tricyclo-dna nucleosides and uses thereof
CN110536694A (en) 2017-04-20 2019-12-03 Atyr 医药公司 For treating pulmonary inflammatory composition and method
EP3612546B1 (en) 2017-04-20 2022-07-13 Synthena AG Modified oligomeric compounds comprising tricyclo-dna nucleosides and uses thereof
US20200384115A1 (en) 2017-04-21 2020-12-10 The Broad Institute , Inc. Targeted delivery to beta cells
US11453891B2 (en) 2017-05-10 2022-09-27 The Regents Of The University Of California Directed editing of cellular RNA via nuclear delivery of CRISPR/CAS9
BR112019023608A2 (en) 2017-05-12 2020-05-26 Crispr Therapeutics Ag MATERIALS AND METHODS FOR HANDLED CELLS AND THEIR USES IN IMMUNO-ONCOLOGY
TWI821192B (en) 2017-07-11 2023-11-11 美商新索思股份有限公司 Incorporation of unnatural nucleotides and methods thereof
JP2020526558A (en) 2017-07-13 2020-08-31 ノースウェスタン ユニバーシティ Common and direct methods for preparing oligonucleotide functionalized metal-organic framework nanoparticles
EP3652317A1 (en) 2017-07-13 2020-05-20 Alnylam Pharmaceuticals, Inc. Lactate dehydrogenase a (ldha) irna compositions and methods of use thereof
KR20200035092A (en) 2017-08-03 2020-04-01 신톡스, 인크. Cytokine conjugates for the treatment of autoimmune diseases
CA3071033A1 (en) 2017-08-18 2019-02-21 Ionis Pharmaceuticals, Inc. Modulation of the notch signaling pathway for treatment of respiratory disorders
EP4219715A3 (en) 2017-09-08 2023-09-06 MiNA Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
WO2019048631A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Hnf4a sarna compositions and methods of use
WO2019051173A1 (en) 2017-09-08 2019-03-14 Ionis Pharmaceuticals, Inc. Modulators of smad7 expression
JP2021508333A (en) 2017-09-19 2021-03-04 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Therapeutic compositions and methods for transthyretin (TTR) -mediated amyloidosis
MX2020004043A (en) 2017-10-17 2021-05-27 Crispr Therapeutics Ag Compositions and methods for gene editing for hemophilia a.
US20210180091A1 (en) 2017-10-26 2021-06-17 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
AU2018360697A1 (en) 2017-11-01 2020-05-14 Alnylam Pharmaceuticals, Inc. Complement component C3 iRNA compositions and methods of use thereof
EP3707155A2 (en) 2017-11-09 2020-09-16 Vertex Pharmaceuticals Incorporated Crispr/cas systems for treatment of dmd
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
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
US20190153440A1 (en) 2017-11-21 2019-05-23 Casebia Therapeutics Llp Materials and methods for treatment of autosomal dominant retinitis pigmentosa
WO2019113149A1 (en) 2017-12-05 2019-06-13 Crispr Therapeutics Ag Crispr-cas9 modified cd34+ human hematopoietic stem and progenitor cells and uses thereof
JP2021506251A (en) 2017-12-14 2021-02-22 クリスパー セラピューティクス アーゲー New RNA programmable endonuclease system, as well as its use in genome editing and other applications
MX2020006012A (en) 2017-12-18 2020-09-14 Alnylam Pharmaceuticals Inc High mobility group box-1 (hmgb1) irna compositions and methods of use thereof.
EP3728595A1 (en) 2017-12-21 2020-10-28 CRISPR Therapeutics AG Materials and methods for treatment of usher syndrome type 2a and/or non-syndromic autosomal recessive retinitis pigmentosa (arrp)
WO2019126641A2 (en) 2017-12-21 2019-06-27 Ionis Pharmaceuticals, Inc. Modulation of frataxin expression
EP3728594A1 (en) 2017-12-21 2020-10-28 CRISPR Therapeutics AG Materials and methods for treatment of usher syndrome type 2a
WO2019140330A1 (en) 2018-01-12 2019-07-18 Casebia Therapeutics Limited Liability Partnership Compositions and methods for gene editing by targeting transferrin
CN111902537A (en) 2018-01-15 2020-11-06 Ionis制药公司 Modulators of DNM2 expression
EP3740472A1 (en) 2018-01-19 2020-11-25 Synthena AG Tricyclo-dna nucleoside precursors and processes for preparing the same
WO2019147743A1 (en) 2018-01-26 2019-08-01 Massachusetts Institute Of Technology Structure-guided chemical modification of guide rna and its applications
EP3749768A1 (en) 2018-02-05 2020-12-16 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
EP3749767A1 (en) 2018-02-05 2020-12-16 Vertex Pharmaceuticals Incorporated Materials and methods for treatment of hemoglobinopathies
WO2019155465A1 (en) 2018-02-08 2019-08-15 Yeda Research And Development Co. Ltd. Methods of identifying and using agents for treating diseases associated with intestinal barrier dysfunction
EP3752616A1 (en) 2018-02-16 2020-12-23 CRISPR Therapeutics AG Compositions and methods for gene editing by targeting fibrinogen-alpha
AU2019224159A1 (en) 2018-02-26 2020-10-01 Synthorx, Inc. IL-15 conjugates and uses thereof
EP3759127A4 (en) 2018-03-02 2022-03-30 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
CN112105625A (en) 2018-03-07 2020-12-18 赛诺菲 Nucleotide precursors, nucleotide analogs, and oligomeric compounds containing the same
EP3768834A1 (en) 2018-03-19 2021-01-27 CRISPR Therapeutics AG Novel rna-programmable endonuclease systems and uses thereof
EP3768694A4 (en) 2018-03-22 2021-12-29 Ionis Pharmaceuticals, Inc. Methods for modulating fmr1 expression
CA3095545A1 (en) 2018-03-30 2019-10-03 Rheinische Friedrich-Wilhelms-Universitat Bonn Aptamers for targeted activaton of t cell-mediated immunity
JP2021520781A (en) 2018-04-06 2021-08-26 チルドレンズ メディカル センター コーポレーションChildren’S Medical Center Corporation Compositions and Methods for Somatic Cell Reprogramming and Imprinting Modulation
JP7275164B2 (en) 2018-04-11 2023-05-17 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 Regulators of EZH2 expression
EP3775211B1 (en) 2018-04-12 2023-04-05 MiNA Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2019204668A1 (en) 2018-04-18 2019-10-24 Casebia Therapeutics Limited Liability Partnership Compositions and methods for knockdown of apo(a) by gene editing for treatment of cardiovascular disease
WO2019217459A1 (en) 2018-05-07 2019-11-14 Alnylam Pharmaceuticals, Inc. Extrahepatic delivery
JP7438135B2 (en) 2018-05-09 2024-02-26 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 reducing expression of FXI
UY38225A (en) 2018-05-09 2019-11-29 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS TO REDUCE THE EXPRESSION OF ATXN3
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
BR112020025824A2 (en) 2018-06-28 2021-03-23 Crispr Therapeutics Ag compositions and methods for genome editing by inserting donor polynucleotides
SG11202100077PA (en) 2018-07-25 2021-02-25 Ionis Pharmaceuticals Inc Compounds and methods for reducing atxn2 expression
JP2021533767A (en) 2018-08-13 2021-12-09 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Hepatitis B virus (HBV) dsRNA substance composition and its usage
EP3837367A1 (en) 2018-08-16 2021-06-23 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
WO2020047229A1 (en) 2018-08-29 2020-03-05 University Of Massachusetts Inhibition of protein kinases to treat friedreich ataxia
AU2019339509A1 (en) 2018-09-14 2021-05-13 Northwestern University Programming protein polymerization with DNA
JP2022500003A (en) 2018-09-18 2022-01-04 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Ketohexokinase (KHK) iRNA composition and its usage
EP3856907A1 (en) 2018-09-28 2021-08-04 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof for treating or preventing ttr-associated ocular diseases
MX2021004455A (en) 2018-10-17 2021-08-11 Crispr Therapeutics Ag Compositions and methods for delivering transgenes.
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
TW202028222A (en) 2018-11-14 2020-08-01 美商Ionis製藥公司 Modulators of foxp3 expression
CN113646430A (en) 2018-11-15 2021-11-12 Ionis制药公司 Modulators of IRF5 expression
IL263184A (en) 2018-11-21 2020-05-31 Yarden Yosef Method of treating cancer and compositions for same
WO2020118259A1 (en) 2018-12-06 2020-06-11 Northwestern University Protein crystal engineering through dna hybridization interactions
AU2019406186A1 (en) 2018-12-20 2021-07-15 Praxis Precision Medicines, Inc. Compositions and methods for the treatment of KCNT1 related disorders
HUE064532T2 (en) 2018-12-20 2024-03-28 Vir Biotechnology Inc Combination hbv therapy
TW202043471A (en) 2019-01-16 2020-12-01 美商健贊公司 Serpinc1 irna compositions and methods of use thereof
SG11202107399WA (en) 2019-01-31 2021-08-30 Ionis Pharmaceuticals Inc Modulators of yap1 expression
SG11202107354WA (en) 2019-02-06 2021-08-30 Synthorx Inc Il-2 conjugates and methods of use thereof
CA3129532A1 (en) 2019-02-15 2020-08-20 Crispr Therapeutics Ag Gene editing for hemophilia a with improved factor viii expression
WO2020171889A1 (en) 2019-02-19 2020-08-27 University Of Rochester Blocking lipid accumulation or inflammation in thyroid eye disease
WO2020170240A1 (en) 2019-02-21 2020-08-27 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Method for reduction drug-induced nephrotoxicity
JP2022523214A (en) 2019-02-27 2022-04-21 アイオーニス ファーマシューティカルズ, インコーポレーテッド Modulator of MALAT1 expression
SG11202109741VA (en) 2019-03-12 2021-10-28 Crispr Therapeutics Ag Novel high fidelity rna-programmable endonuclease systems and uses thereof
TW202043251A (en) 2019-03-29 2020-12-01 美商Ionis製藥公司 Compounds and methods for modulating ube3a-ats
AU2020253823A1 (en) 2019-03-29 2021-10-14 Dicerna Pharmaceuticals, Inc. Compositions and methods for the treatment of KRAS associated diseases or disorders
WO2020208361A1 (en) 2019-04-12 2020-10-15 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
BR112021021686A2 (en) 2019-05-03 2022-03-22 Dicerna Pharmaceuticals Inc Double-stranded nucleic acid inhibitor molecules with short sense strands
WO2020225606A1 (en) 2019-05-08 2020-11-12 Crispr Therapeutics Ag Crispr/cas all-in-two vector systems for treatment of dmd
CN114126628A (en) 2019-05-13 2022-03-01 维尔生物科技有限公司 Compositions and methods for treating Hepatitis B Virus (HBV) infection
EP3969052A1 (en) 2019-05-17 2022-03-23 Alnylam Pharmaceuticals Inc. Oral delivery of oligonucleotides
EP3983543A4 (en) 2019-06-14 2023-05-03 The Scripps Research Institute Reagents and methods for replication, transcription, and translation in semi-synthetic organisms
CA3142521A1 (en) 2019-06-17 2020-12-24 Vertex Pharmaceuticals Incorporated Compositions and methods for editing beta-globin for treatment of hemaglobinopathies
TW202114731A (en) 2019-06-18 2021-04-16 愛爾蘭商健生科學愛爾蘭無限公司 COMBINATION OF HEPATITIS B VIRUS (HBV) VACCINES AND HBV-TARGETING RNAi
AU2020297008A1 (en) 2019-06-18 2022-02-17 Janssen Sciences Ireland Unlimited Company Combination of hepatitis B virus (HBV) vaccines and HBV-targeting RNAi
WO2021021673A1 (en) 2019-07-26 2021-02-04 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
EP4007811A2 (en) 2019-08-01 2022-06-08 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
CN114555128A (en) 2019-08-15 2022-05-27 新索思股份有限公司 Combination immunooncology therapy with IL-2 conjugates
KR20220062517A (en) 2019-08-15 2022-05-17 아이오니스 파마수티컬즈, 인코포레이티드 Linkage-modified oligomeric compounds and uses thereof
WO2021032777A1 (en) 2019-08-19 2021-02-25 Mina Therapeutics Limited Oligonucleotide conjugate compositions and methods of use
CA3148135A1 (en) 2019-08-23 2021-03-04 Carolina E. CAFFARO Il-15 conjugates and uses thereof
JP2022546570A (en) 2019-09-03 2022-11-04 アルナイラム ファーマシューティカルズ, インコーポレイテッド Compositions and methods for inhibiting expression of the LECT2 gene
JP2022547078A (en) 2019-09-10 2022-11-10 シンソークス, インコーポレイテッド IL-2 conjugates and methods of use for treating autoimmune diseases
EP4038189A1 (en) 2019-10-04 2022-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
WO2021076828A1 (en) 2019-10-18 2021-04-22 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
BR112022007540A2 (en) 2019-10-22 2022-07-12 Alnylam Pharmaceuticals Inc COMPONENTS COMPLEMENTARY C3 IRNA COMPOSITIONS AND METHODS OF USE THEREOF
MX2022004388A (en) 2019-11-01 2022-05-06 Alnylam Pharmaceuticals Inc HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF.
US20230040920A1 (en) 2019-11-01 2023-02-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
IL291786A (en) 2019-11-04 2022-06-01 Synthorx Inc Interleukin 10 conjugates and uses thereof
WO2021092145A1 (en) 2019-11-06 2021-05-14 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna composition and methods of use thereof for treating or preventing ttr-associated ocular diseases
US20230016929A1 (en) 2019-11-06 2023-01-19 Alnylam Pharmaceuticals, Inc. Extrahepatic delivery
TW202132568A (en) 2019-11-13 2021-09-01 美商阿尼拉製藥公司 Methods and compositions for treating an angiotensinogen- (agt-) associated disorder
US20230056569A1 (en) 2019-11-22 2023-02-23 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
JP2023503635A (en) 2019-11-27 2023-01-31 クリスパー・セラピューティクス・アクチェンゲゼルシャフト Methods of synthesizing RNA molecules
EP4073251A1 (en) 2019-12-13 2022-10-19 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
US20230054569A1 (en) 2019-12-18 2023-02-23 Alia Therapeutics Srl Compositions and methods for treating retinitis pigmentosa
MX2022008738A (en) 2020-01-15 2022-09-23 Dicerna Pharmaceuticals Inc 4'-o-methylene phosphonate nucleic acids and analogues thereof.
US20230057461A1 (en) 2020-01-27 2023-02-23 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Rab13 and net1 antisense oligonucleotides to treat metastatic cancer
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
WO2021163066A1 (en) 2020-02-10 2021-08-19 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing vegf-a expression
MX2022010052A (en) 2020-02-18 2022-09-05 Alnylam Pharmaceuticals Inc Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof.
PE20221913A1 (en) 2020-02-28 2022-12-23 Ionis Pharmaceuticals Inc COMPOUNDS AND METHODS FOR MODULATING SMN2
EP4114947A1 (en) 2020-03-05 2023-01-11 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
EP4114948A1 (en) 2020-03-06 2023-01-11 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
WO2021178778A1 (en) 2020-03-06 2021-09-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin (ttr)
EP4121534A1 (en) 2020-03-18 2023-01-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
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
WO2021202443A2 (en) 2020-03-30 2021-10-07 Alnylam Pharmaceucticals, Inc. Compositions and methods for silencing dnajc15 gene expression
BR112022020145A2 (en) 2020-04-06 2023-01-03 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR SILENCING THE MYOC EXPRESSION
EP4133076A1 (en) 2020-04-07 2023-02-15 Alnylam Pharmaceuticals, Inc. Angiotensin-converting enzyme 2 (ace2) irna compositions and methods of use thereof
EP4133077A1 (en) 2020-04-07 2023-02-15 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
EP4143319A1 (en) 2020-04-27 2023-03-08 Alnylam Pharmaceuticals, Inc. Apolipoprotein e (apoe) irna agent compositions and methods of use thereof
CA3181198A1 (en) 2020-04-30 2021-11-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
JP2023524065A (en) 2020-05-01 2023-06-08 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 ATXN1
WO2021231692A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
WO2021231680A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
WO2021231675A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
EP4150078A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
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)
EP4150086A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
EP4150077A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
EP4150089A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rs1)
WO2021237097A1 (en) 2020-05-21 2021-11-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
EP4153747A2 (en) 2020-05-22 2023-03-29 Wave Life Sciences Ltd. Double stranded oligonucleotide compositions and methods relating thereto
US11408000B2 (en) 2020-06-03 2022-08-09 Triplet Therapeutics, Inc. Oligonucleotides for the treatment of nucleotide repeat expansion disorders associated with MSH3 activity
EP4161552A1 (en) 2020-06-05 2023-04-12 The Broad Institute, Inc. Compositions and methods for treating neoplasia
WO2021252557A1 (en) 2020-06-09 2021-12-16 Alnylam Pharmaceuticals, Inc. Rnai compositions and methods of use thereof for delivery by inhalation
KR20230026455A (en) 2020-06-18 2023-02-24 알닐람 파마슈티칼스 인코포레이티드 Xanthine dehydrogenase (XDH) iRNA compositions and methods of use thereof
BR112022026316A2 (en) 2020-06-24 2023-03-07 Vir Biotechnology Inc ENGINEERED HEPATITIS B VIRUS NEUTRALIZING ANTIBODIES AND THEIR USES
TW202216203A (en) 2020-06-25 2022-05-01 美商欣爍克斯公司 Immuno oncology combination therapy with il-2 conjugates and anti-egfr antibodies
KR20230029837A (en) 2020-06-29 2023-03-03 아이오니스 파마수티컬즈, 인코포레이티드 Compounds and methods for modulating PLP1
WO2022011214A1 (en) 2020-07-10 2022-01-13 Alnylam Pharmaceuticals, Inc. Circular sirnas
IL299771A (en) 2020-07-10 2023-03-01 Inst Nat Sante Rech Med Methods and compositions for treating epilepsy
MX2023001541A (en) 2020-08-04 2023-03-08 Dicerna Pharmaceuticals Inc Systemic delivery of oligonucleotides.
EP4217489A1 (en) 2020-09-24 2023-08-02 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
EP4222264A1 (en) 2020-09-30 2023-08-09 CRISPR Therapeutics AG Materials and methods for treatment of amyotrophic lateral sclerosis
EP3978608A1 (en) 2020-10-05 2022-04-06 SQY Therapeutics Oligomeric compound for dystrophin rescue in dmd patients throughout skipping of exon-51
TW202229552A (en) 2020-10-05 2022-08-01 美商艾拉倫製藥股份有限公司 G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
TW202228786A (en) 2020-10-09 2022-08-01 美商欣爍克斯公司 Immuno oncology therapies with il-2 conjugates
AU2021356693A1 (en) 2020-10-09 2023-06-15 Synthorx, Inc. Immuno oncology combination therapy with il-2 conjugates and pembrolizumab
EP4228637A1 (en) 2020-10-15 2023-08-23 Yeda Research and Development Co. Ltd Method of treating myeloid malignancies
WO2022084331A2 (en) 2020-10-20 2022-04-28 Sanofi Novel ligands for asialoglycoprotein receptor
AU2021365822A1 (en) 2020-10-21 2023-06-08 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
EP4232582A1 (en) 2020-10-23 2023-08-30 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
CA3200595A1 (en) 2020-11-13 2022-05-19 Alnylam Pharmaceuticals, Inc. Coagulation factor v (f5) irna compositions and methods of use thereof
CA3201661A1 (en) 2020-11-18 2022-05-27 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
TW202237150A (en) 2020-12-01 2022-10-01 美商艾拉倫製藥股份有限公司 Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
WO2022125490A1 (en) 2020-12-08 2022-06-16 Alnylam Pharmaceuticals, Inc. Coagulation factor x (f10) irna compositions and methods of use thereof
JP2024501288A (en) 2020-12-23 2024-01-11 フラッグシップ パイオニアリング イノベーションズ シックス,エルエルシー Compositions of modified TREM and uses thereof
KR20230136130A (en) 2020-12-31 2023-09-26 알닐람 파마슈티칼스 인코포레이티드 Cyclic-disulfide modified phosphate-based oligonucleotide prodrug
WO2022147223A2 (en) 2020-12-31 2022-07-07 Alnylam Pharmaceuticals, Inc. 2'-modified nucleoside based oligonucleotide prodrugs
WO2022150260A1 (en) 2021-01-05 2022-07-14 Alnylam Pharmaceuticals, Inc. COMPLEMENT COMPONENT 9 (C9) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
TW202302148A (en) 2021-02-12 2023-01-16 美商欣爍克斯公司 Lung cancer combination therapy with il-2 conjugates and an anti-pd-1 antibody or antigen-binding fragment thereof
BR112023015761A2 (en) 2021-02-12 2023-11-07 Alnylam Pharmaceuticals Inc SUPEROXIDE DISMUTASE 1 (SOD1) IRNA COMPOSITIONS AND METHODS OF USE THEREOF TO TREAT OR PREVENT NEURODEGENERATIVE DISEASES ASSOCIATED WITH SUPEROXIDE DISMUTASE 1 (SOD1)
TW202245843A (en) 2021-02-12 2022-12-01 美商欣爍克斯公司 Skin cancer combination therapy with il-2 conjugates and cemiplimab
WO2022182864A1 (en) 2021-02-25 2022-09-01 Alnylam Pharmaceuticals, Inc. Prion protein (prnp) irna compositions and methods and methods of use thereof
EP4298218A1 (en) 2021-02-26 2024-01-03 Alnylam Pharmaceuticals, Inc. 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
US20220288181A1 (en) 2021-03-12 2022-09-15 Northwestern University Antiviral vaccines using spherical nucleic acids
EP4305169A1 (en) 2021-03-12 2024-01-17 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
CA3214137A1 (en) 2021-03-26 2022-09-29 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
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
EP4326873A1 (en) 2021-04-22 2024-02-28 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating cancer
BR112023022284A2 (en) 2021-04-26 2023-12-26 Alnylam Pharmaceuticals Inc COMPOSITIONS OF TRANSMEMBRANE PROTEASE IRNA, SERINE 6 (TMPRSS6) 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
WO2022235537A1 (en) 2021-05-03 2022-11-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating transthyretin (ttr) mediated amyloidosis
EP4341401A1 (en) 2021-05-18 2024-03-27 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
WO2022246023A1 (en) 2021-05-20 2022-11-24 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
TW202317762A (en) 2021-06-02 2023-05-01 美商艾拉倫製藥股份有限公司 Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
WO2022256534A1 (en) 2021-06-03 2022-12-08 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and pembrolizumab
IL308743A (en) 2021-06-04 2024-01-01 Alnylam Pharmaceuticals Inc HUMAN CHROMOSOME 9 OPEN READING FRAME 72 (C9ORF72) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
AR126070A1 (en) 2021-06-08 2023-09-06 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS FOR TREATING OR PREVENTING STARGARDT DISEASE AND/OR DISORDERS ASSOCIATED WITH RETINOL BORDER PROTEIN 4 (RBP4)
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
BR112023023768A2 (en) 2021-06-11 2024-02-27 Bayer Ag TYPE V RNA PROGRAMMABLE ENDONUCLEASE SYSTEMS
CA3223192A1 (en) 2021-06-18 2022-12-22 Ionis Pharmaceuticals, Inc. Compounds and methods for reducing ifnar1 expression
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
KR20240026203A (en) 2021-06-30 2024-02-27 알닐람 파마슈티칼스 인코포레이티드 Methods and compositions for treating angiotensinogen (AGT)-related disorders
WO2023283403A2 (en) 2021-07-09 2023-01-12 Alnylam Pharmaceuticals, Inc. Bis-rnai compounds for cns delivery
WO2023285431A1 (en) 2021-07-12 2023-01-19 Alia Therapeutics Srl Compositions and methods for allele specific treatment of retinitis pigmentosa
TW202333748A (en) 2021-07-19 2023-09-01 美商艾拉倫製藥股份有限公司 Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
WO2023003922A1 (en) 2021-07-21 2023-01-26 Alnylam Pharmaceuticals, Inc. Metabolic disorder-associated target gene irna compositions and methods of use thereof
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
IL310244A (en) 2021-08-03 2024-03-01 Alnylam Pharmaceuticals Inc Transthyretin (ttr) irna compositions and methods of use thereof
CA3228255A1 (en) 2021-08-04 2023-02-09 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing angiotensinogen (agt)
AR126771A1 (en) 2021-08-13 2023-11-15 Alnylam Pharmaceuticals Inc RNAi COMPOSITIONS AGAINST FACTOR XII (F12) AND THEIR METHODS OF USE
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
WO2023044370A2 (en) 2021-09-17 2023-03-23 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing complement component 3 (c3)
AU2022345881A1 (en) 2021-09-20 2024-03-21 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
WO2023064530A1 (en) 2021-10-15 2023-04-20 Alnylam Pharmaceuticals, Inc. Extra-hepatic delivery irna compositions and methods of use thereof
WO2023069603A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
TW202334418A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Huntingtin (htt) irna agent compositions and methods of use thereof
WO2023076451A1 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
WO2023099884A1 (en) 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use
WO2023122573A1 (en) 2021-12-20 2023-06-29 Synthorx, Inc. Head and neck cancer combination therapy comprising an il-2 conjugate and pembrolizumab
WO2023118349A1 (en) 2021-12-21 2023-06-29 Alia Therapeutics Srl Type ii cas proteins and applications thereof
WO2023118068A1 (en) 2021-12-23 2023-06-29 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2023122750A1 (en) 2021-12-23 2023-06-29 Synthorx, Inc. Cancer combination therapy with il-2 conjugates and cetuximab
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023170435A1 (en) 2022-03-07 2023-09-14 Mina Therapeutics Limited Il10 sarna compositions and methods of use
WO2023177866A1 (en) 2022-03-18 2023-09-21 Dicerna Pharmaceuticals, Inc. Decarboxylative acetoxylation using mn(ii) or mn(iii) reagent for synthesis of 4'-acetoxy- nucleoside and use thereof for synthesis of corresponding 4'-(dimethoxyphosphoryl)methoxy- nucleotide
WO2023194359A1 (en) 2022-04-04 2023-10-12 Alia Therapeutics Srl Compositions and methods for treatment of usher syndrome type 2a
WO2023220744A2 (en) 2022-05-13 2023-11-16 Alnylam Pharmaceuticals, Inc. Single-stranded loop oligonucleotides
WO2023233290A1 (en) 2022-05-31 2023-12-07 Janssen Sciences Ireland Unlimited Company Rnai agents targeting pd-l1
WO2023237587A1 (en) 2022-06-10 2023-12-14 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2024006999A2 (en) 2022-06-30 2024-01-04 Alnylam Pharmaceuticals, Inc. Cyclic-disulfide modified phosphate based oligonucleotide prodrugs
EP4311579A1 (en) 2022-07-29 2024-01-31 Association Française contre les Myopathies B cell-specific mab-sirna conjugates improve myasthenia
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
WO2024056880A2 (en) 2022-09-16 2024-03-21 Alia Therapeutics Srl Enqp type ii cas proteins and applications thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4908453A (en) * 1989-01-23 1990-03-13 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-biotinylated oligonucleotides
US4958013A (en) * 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5223618A (en) * 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
US5262536A (en) * 1988-09-15 1993-11-16 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-tagged oligonucleotides
US5391723A (en) * 1989-05-31 1995-02-21 Neorx Corporation Oligonucleotide conjugates

Family Cites Families (4)

* 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
EP1331011A3 (en) * 1991-10-24 2003-12-17 Isis Pharmaceuticals, Inc. Derivatized oligonucleotides having improved uptake and other properties
DE69331543T2 (en) * 1992-03-05 2002-09-26 Isis Pharmaceutical Inc COVALENTLY NETWORKED OLIGONUCLEOTIDES
IL107166A (en) * 1992-10-01 2000-10-31 Univ Columbia Complex combinatorial chemical libraries encoded with tags

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5262536A (en) * 1988-09-15 1993-11-16 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-tagged oligonucleotides
US4908453A (en) * 1989-01-23 1990-03-13 E. I. Du Pont De Nemours And Company Reagents for the preparation of 5'-biotinylated oligonucleotides
US5391723A (en) * 1989-05-31 1995-02-21 Neorx Corporation Oligonucleotide conjugates
US4958013A (en) * 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5223618A (en) * 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BIOCONJUGATE CHEMISTRY, Vol. 1, No. 3, issued May/June 1990, GOODCHILD, "Conjugates of Oligonucleotides and Modified Oligonucleotides: A Review of Their Synthesis and Properties", pages 165-186. *
BIOCONJUGATE CHEMISTRY, Vol. 2, No. 6, issued 1991, TUNG et al., "Preparation of Oligonucleotide-Peptide Conjugates", pages 464-465. *
CHEMICAL REVIEWS, Vol. 90, No. 4, issued 1990, UHLMANN et al., "Antisense Oligonucleotides: A New Therapeutic Principle", pages 544-584. *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6495553B1 (en) 1997-08-08 2002-12-17 Newbiotics, Inc. Methods and compositions for overcoming resistance to biologic and chemotherapy
US7465734B2 (en) 1997-08-08 2008-12-16 Celmed Oncology (Usa), Inc. Methods and compositions for overcoming resistance to biologic and chemotherapy
US7462605B2 (en) 1998-01-23 2008-12-09 Celmed Oncology (Usa), Inc. Phosphoramidate compounds and methods of use
US6339151B1 (en) 1998-01-23 2002-01-15 Newbiotics, Inc. Enzyme catalyzed therapeutic agents
US6245750B1 (en) * 1998-01-23 2001-06-12 Newbiotics, Inc. Enzyme catalyzed therapeutic agents
US7601703B2 (en) * 1998-01-23 2009-10-13 Celmed Oncology (Usa), Inc. Enzyme catalyzed therapeutic agents
US6683061B1 (en) 1999-07-22 2004-01-27 Newbiotics, Inc. Enzyme catalyzed therapeutic activation
US7605144B2 (en) 1999-07-22 2009-10-20 Celmed Oncology (Usa), Inc. Enzyme catalyzed therapeutic compounds
WO2001010882A1 (en) * 1999-08-10 2001-02-15 Adani, Alexander Depot forms of modified nucleoside 5'-hydrogenphosphates as inhibitors of reproduction of human immunodeficiency virus and human hepatitis b virus
US6620796B1 (en) 1999-11-08 2003-09-16 Micrologix Biotech Inc. Combinatorial library synthesis and pharmaceutically active compounds produced thereby
WO2001034622A1 (en) * 1999-11-08 2001-05-17 Origenix Technologies, Inc. Combinatorial library synthesis and pharmaceutically active compounds produced thereby
US7138388B2 (en) 2001-01-19 2006-11-21 Celmed Oncology (Usa), Inc. Methods to treat autoimmune and inflammatory conditions
US7256179B2 (en) 2001-05-16 2007-08-14 Migenix, Inc. Nucleic acid-based compounds and methods of use thereof
US7709449B2 (en) 2001-05-16 2010-05-04 Migenix, Inc. Nucleic acid-based compounds and methods of use thereof

Also Published As

Publication number Publication date
US5608046A (en) 1997-03-04
AU3892395A (en) 1996-04-19

Similar Documents

Publication Publication Date Title
US5998603A (en) 4'-desmethyl nucleoside analogs, and oligomers thereof
US5688941A (en) Methods of making conjugated 4' desmethyl nucleoside analog compounds
US5608046A (en) Conjugated 4'-desmethyl nucleoside analog compounds
EP0716604B1 (en) Thiol-derivatized nucleosides and oligonucleosides
US5677437A (en) Heteroatomic oligonucleoside linkages
US6087482A (en) Heteroatomic oligonucleoside linkages
EP0651759B1 (en) Novel 2'-o-alkyl nucleosides and phosphoramidites processes for the preparation and uses thereof
US6194598B1 (en) Aminooxy-modified oligonucleotide synthetic intermediates
US5914396A (en) 2'-O-modified nucleosides and phosphoramidites
US6001841A (en) Cyclobutyl oligonucleotide surrogates
US6531584B1 (en) 2'modified oligonucleotides
US6399754B1 (en) Sugar modified oligonucleotides
US6172209B1 (en) Aminooxy-modified oligonucleotides and methods for making same
KR0155574B1 (en) Back bone modified oligonucleotide analogs
US6369040B1 (en) Pyrimidine nucleosides
US6265558B1 (en) Thiol-derivatized nucleosides and oligonucleosides
JPH04500687A (en) Steroid modified oligonucleotide
KR20210008369A (en) Oligonucleotide conjugates comprising 7'-5'-alpha-anomer-bicyclic sugar nucleosides
US5852182A (en) Thiol-derivatized oligonucleosides
WO1995024185A1 (en) Novel pyrimidine nucleosides
JP7263236B2 (en) Novel bicyclic nucleosides and oligomers prepared therefrom
WO1994019023A1 (en) Cyclobutyl antisense oligonucleotides, methods of making and use thereof
EP0739902B1 (en) H-Phosphonate ribonucleotide derivatives
WO1994022886A1 (en) Heteroatomic oligonucleoside linkages
WO1994022886A9 (en) Heteroatomic oligonucleoside linkages

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS JP KE KG KP KR KZ LK LR LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TT UA UG US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 08809239

Country of ref document: US

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA