US20050119214A1 - Nuclease resistant double-stranded ribonucleic acid - Google Patents

Nuclease resistant double-stranded ribonucleic acid Download PDF

Info

Publication number
US20050119214A1
US20050119214A1 US10/936,115 US93611504A US2005119214A1 US 20050119214 A1 US20050119214 A1 US 20050119214A1 US 93611504 A US93611504 A US 93611504A US 2005119214 A1 US2005119214 A1 US 2005119214A1
Authority
US
United States
Prior art keywords
irna agent
modification
nucleotide
irna
strand
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/936,115
Inventor
Muthiah Manoharan
Kallanthottathil Rajeev
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Alnylam Pharmaceuticals Inc
Original Assignee
Individual
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
Priority claimed from PCT/US2004/007070 external-priority patent/WO2004080406A2/en
Priority claimed from PCT/US2004/010586 external-priority patent/WO2004090108A2/en
Priority claimed from PCT/US2004/011255 external-priority patent/WO2004091515A2/en
Priority claimed from PCT/US2004/011822 external-priority patent/WO2004094345A2/en
Application filed by Individual filed Critical Individual
Priority to US10/936,115 priority Critical patent/US20050119214A1/en
Assigned to ALNYLAM PHARMACEUTICALS, INC. reassignment ALNYLAM PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MANOHARAN, MUTHIAH, RAJEEV, KALLANTHOTTATHIL G.
Publication of US20050119214A1 publication Critical patent/US20050119214A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/554Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being a steroid plant sterol, glycyrrhetic acid, enoxolone or bile acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/02Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with ribosyl as saccharide radical
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/344Position-specific modifications, e.g. on every purine, at the 3'-end
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/351Conjugate
    • C12N2310/3515Lipophilic moiety, e.g. cholesterol
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • C12N2320/32Special delivery means, e.g. tissue-specific
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/50Methods for regulating/modulating their activity
    • C12N2320/51Methods for regulating/modulating their activity modulating the chemical stability, e.g. nuclease-resistance
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2330/00Production
    • C12N2330/30Production chemically synthesised

Definitions

  • This invention features modified iRNA agents having improved stability in cells and biological fluids, and methods of making and identifying iRNA agents having improved stability, and of using the iRNA agents to inhibit the expression or function of a target gene.
  • diseases arise from the abnormal expression or activity of a particular gene or group of genes.
  • disease can result through expression of a mutant form of protein, as well as from expression of viral genes that have been integrated into the genome of their host.
  • the therapeutic benefits of being able to selectively silence these abnormal or foreign genes are obvious.
  • Double-stranded RNA molecules can block gene expression by virtue of a highly conserved regulatory mechanism known as RNA interference (RNAi).
  • RNAi RNA interference
  • the RNA III Dicer enzyme processes dsRNA into small interfering RNA (siRNA) of approximately 22 nucleotides.
  • siRNA small interfering RNA
  • One strand of the siRNA serves as a guide sequence to induce cleavage of messenger RNAs (mRNAs) including a nucleotide sequence which is at least partially complementary to the sequence of the antisense strand by an RNA-induced silencing complex RISC (Hammond, S. M., et al., Nature (2000) 404:293-296).
  • RISC RNA-induced silencing complex
  • the invention features iRNA agents having improved stability in cells and biological fluids, methods of making and identifying iRNA agents having improved stability, as well as compositions and methods for inhibiting the expression of a target gene in a cell using the iRNA agent.
  • the invention also features compositions and methods for treating diseases caused by the expression or activity of the target gene.
  • the invention features iRNA agents that contain modifications that increase resistance of the iRNA agents to exonucleolytic degradation, including 5′ exonucleolytice and 3′ exonucleolytic degradation. The modifications render the iRNA agents more resistant to enzymatic or chemical degradation, and thus more stable and bioavailable than an otherwise identical iRNA agent that does not include the modification.
  • the invention features iRNA agents having improved stability when administered to a subject. These agents can have, e.g., improved stability in cells and biological samples, particularly serum.
  • the iRNA agent includes one or more of various modifications.
  • the iRNA agents can include a backbone modification to one or more of the terminal nucleotides on an iRNA strand.
  • a terminal nucleotide can be the nucleotide at the last position, and can include the nucleotide(s) at the second, third, fourth, and fifth positions from the 5′ or 3′-most ends of the sense or antisense strand of an iRNA agent.
  • an iRNA agent includes a phosphorothioate linkage or P-alkyl modification in the linkages between one or more of the terminal nucleotides of an iRNA agent.
  • one or more terminal nucleotides of an iRNA agent include a sugar modification, e.g., a 2′ or 3′ sugar modification.
  • Exemplary sugar modifications include, for example, a 2′-O-methylated nucleotide, a 2′-deoxy nucleotide, a 2′-deoxyfluoro nucleotide, a 2′-O-methoxyethyl nucleotide, a 2′-O-NMA, a 2′-DMAEOE, a 2′-AP, 2′-hydroxy, or a 2′-ara-fluoro or a locked nucleic acid (LNA), extended nucleic acid (ENA), hexose nucleic acid (HNA), or cyclohexene nucleic acid (CeNA).
  • a 2′ modification is preferably 2′OMe, more preferably, 2′fluoro. In one embodiment the modification is an LNA and the LNA is on the 5′ or 3′ end of the sense strand, and not on the 5′ or 3′ end of the antisense strand.
  • the iRNA agent includes a 3′ sugar modification, e.g., a 3′OMe modification.
  • a 3′OMe modification is on the 3′ or 5′ end of the sense strand of the iRNA agent.
  • an iRNA agent includes a 5′-methyl-pyrimidine (e.g., a 5′-methyl-uridine modification or a 5′-methyl-cytodine modification.
  • a 5′-methyl-pyrimidine e.g., a 5′-methyl-uridine modification or a 5′-methyl-cytodine modification.
  • At least one terminal nucleotide of an iRNA agent has a phosphorothioate linkage and a 2′ sugar modification, e.g., a 2′OMe or 2° F. modification.
  • at least one terminal nucleotide of an iRNA agent has a 5′ Me-pyrimidine and a 2′ sugar modification, e.g., a 2′F or 2′OMe modification.
  • an iRNA agent includes a nucleobase modification, such as a cationic modification, such as a 3′-abasic cationic modification.
  • the cationic modification can be e.g., an alkylamino-dT (e.g., a C6 amino-dT), an allylamino conjugate, a pyrrolidine conjugate, a pthalamido or a hydroxyprolinol conjugate, on one or more of the terminal nucleotides of the iRNA agent.
  • An alkylamino-dT conjugate is preferably attached to the 3′ end of the sense or antisense strand of an iRNA agent.
  • a pyrrolidine linker is preferably attached to the 3′ or 5′ end of the sense strand, or the 3′ end of the antisense strand.
  • An allyl amine uridine is preferably on the 3′ or 5′ end of the sense strand, and not on the 5′ end of the antisense strand.
  • the iRNA agent includes an conjugate on one or more of the terminal nucleotides of the iRNA agent.
  • the conjugate can be, for example, a lipophile, a terpene, a protein binding agent, a vitamin, a carbohydrate, or a peptide.
  • the conjugate can be naproxen, nitroindole (or another conjugate that contributes to stacking interactions), folate, ibuprofen, or a C5 pyrimidine linker.
  • the conjugates are glyceride lipid conjugates (e.g. a dialkyl glyceride derivatives), vitamin E conjugates, or thio-cholesterols.
  • conjugates are on the 3′ end of the antisense strand, or on the 5′ or 3′ end of the sense strand, and preferably the conjugates are not on the 3′ end of the antisense strand and on the 3′ end of the sense strand.
  • the conjugate is naproxen, and the conjugate is preferably on the 5′ or 3′ end of the sense or antisense strands.
  • the conjugate is cholesterol, and the conjugate is preferably on the 5′ or 3′ end of the sense strand and preferably not present on the antisense strand.
  • the cholesterol is conjugated to the iRNA agent by a pyrrolidine linker, or serinol linker, or hydroxyprolinol linker.
  • the conjugate is a dU-cholesterol, or cholesterol is conjugated to the iRNA agent by a disulfide linkage.
  • the conjugate is cholanic acid
  • the cholanic acid is attached to the 5′ or 3′ end of the sense strand, or the 3′ end of the antisense strand.
  • the cholanic acid is attached to the 3′ end of the sense strand and the 3′ end of the antisense strand.
  • one or more terminal nucleotides has a 2′-5′ linkage.
  • a 2′-5′ linkage occurs on the sense strand, e.g., the 5′ end of the sense strand.
  • the iRNA agent includes an L-sugar, preferably at the 5′ or 3′ end of the sense strand.
  • the iRNA agent includes a methylphosphonate at one or more terminal nucleotides to enhance exonuclease resistance, e.g., at the 3′ end of the sense or antisense strands of the iRNA agent.
  • an iRNA agent has been modified by replacing one or more ribonucleotides with deoxyribonucleotides.
  • adjacent deoxyribonucleotides are joined by phosphorothioate linkages, and the iRNA agent does not include more than four consecutive deoxyribonucleotides on the sense or the antisense strands.
  • an iRNA agent having increased stability in cells and biological samples includes a difluorotoluyl (DFT) modification, e.g., 2,4-difluorotoluyl uracil, or a guanidine to inosine substitution.
  • DFT difluorotoluyl
  • the iRNA agent includes a terminal 5′-uridine-adenine-3′ (5′-UA-3′) dinucleotide wherein the uridine is a 2′-modified nucleotide, or a terminal 5′-uridine-guanine-3′ (5′-UG-3′) dinucleotide, wherein the 5′-uridine is a 2′-modified nucleotide, or a terminal 5′-cytidine-adenine-3′ (5′-CA-3′) dinucleotide, wherein the 5′-cytidine is a 2′-modified nucleotide, or a terminal 5′-uridine-uridine-3′ (5′-UU-3′) dinucleotide, wherein the 5′-uridine is a 2′-modified nucleotide, or a terminal 5′-cytidine-cytidine-3′ (5′-CC-3′) dinucleotide, wherein the 5′-cytidine
  • the chemically modified nucleotide in the iRNA agent may be a 2′-O-methylated nucleotide.
  • the modified nucleotide can be a 2′-deoxy nucleotide, a 2′-deoxyfluoro nucleotide, a 2′-O-methoxyethyl nucleotide, a 2′-O-NMA, a 2′-DMAEOE, a 2′-AP, 2′-hydroxy, or a 2′-ara-fluoro, or a locked nucleic acid (LNA), extended nucleic acid (ENA), hexose nucleic acid (HNA), or cyclohexene nucleic acid (CeNA).
  • LNA locked nucleic acid
  • ENA extended nucleic acid
  • HNA hexose nucleic acid
  • CeNA cyclohexene nucleic acid
  • the iRNA agents including the modifications described herein are particularly stabilized against ex
  • an iRNA agent includes a cholesterol molecule on the 3′ end of an iRNA agent, and conjugated to the iRNA agent by a pyrrolidine linker, and at least one additional modification, e.g., a modification described herein, such as a phosphorothioate linkage, for inhibiting exonucleolytic degradation.
  • an iRNA agent includes a cholanic acid modification on one or both of the 3′ ends of the sense or antisense strands, and at least one additional modification, e.g., a modification described herein, such as a phosphorothioate linkage, for inhibiting exonucleolytic degradation.
  • the iRNA agent has a single overhang, e.g., one end of the iRNA agent has a 3′ or 5′ overhang and the other end of the iRNA agent is a blunt end.
  • the iRNA agent has a double overhang, e.g., both ends of the iRNA agent have a 3′ or 5′ overhang, such as a dinuclotide overhang.
  • both ends of the iRNA agent have blunt ends.
  • the iRNA agent may further include a sense RNA strand and an antisense RNA strand, wherein the antisense RNA strand is 25 or fewer nucleotides in length, and includes an antisense nucleotide sequence having 18-25 nucleotides in length.
  • the iRNA agent may further include a nucleotide overhang having 1 to 4 unpaired nucleotides, which may be at the 3′-end of the antisense RNA strand, and the nucleotide overhang may have the nucleotide sequence 5′-GC-3′ or 5′-CGC-3′.
  • the unpaired nucleotides may have at least one phosphorothioate dinucleotide linkage, and at least one of the unpaired nucleotides may be chemically modified in the 2′-position.
  • the doublestrand region of the iRNA agent includes phosphorothioate dinucleotide linkages on one or both of the sense and antisense strands.
  • the antisense RNA strand and the sense RNA strand are connected with a linker.
  • the chemical linker may be a hexaethylene glycol linker, a poly-(oxyphosphinico-oxy-1,3-propandiol) linker, an allyl linker, or a polyethylene glycol linker.
  • the invention in another aspect, relates to a method of making an iRNA agent.
  • the method includes preparing an antisense RNA strand and a sense RNA strand, wherein at least one of the RNA strands includes a modification described herein, e.g., a modification that confers an enhanced resistance to exonucleolytic cleavage.
  • the RNA strands may then be mixed to form a double stranded iRNA agent.
  • the RNA strands may be prepared using solid-phase synthesis in a 3′ to 5′ direction.
  • the invention in yet another aspect, relates to a method for inhibiting the expression of a target gene in a cell.
  • the method includes introducing an iRNA agent, as described above, into the cell, and maintaining the cell for a time sufficient to obtain iRNA agent-mediated downregulation, e.g., by degradation, of an mRNA transcript of the target gene.
  • the cell may be a mammalian cell, such as a human cell.
  • the invention relates to a pharmaceutical composition for inhibiting the expression of a target gene in an organism.
  • the compositions include an iRNA agent, as described above, and a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier may be an aqueous solution, such as phosphate buffered saline, or it may include a micellar structure, such as a liposome, capsid, capsoid, polymeric nanocapsule, or polymeric microcapsule.
  • the invention relates to a method for treating a disease caused by the expression of a target gene in a subject.
  • the method includes administering a pharmaceutical composition containing an iRNA agent, as described above, and a pharmaceutically acceptable carrier.
  • the subject may be a mammal, such as a human.
  • the invention in another aspect, relates to methods of identifying an iRNA agent having increased stability in a biological sample.
  • the method may include preparing a mixture of iRNA agent, incubating the mixture of iRNA agent in the biological sample, and identifying an iRNA agent exhibiting an increased stability as compared to other iRNA agent in the biological sample.
  • the mixture of iRNA agent may be prepared by chemical synthesis or by extraction from a biological sample.
  • the method may include introducing an iRNA agent into a cell, maintaining the cell under conditions suitable for expressing a protein encoded by a target gene, measuring an amount of the protein produced in the cell, comparing the amount of the protein produced in the cell to that in a control cell, and identifying an iRNA agent that causes a reduction in the amount of protein in the cell as compared to the control cell.
  • the cell may be a mammalian cell.
  • the invention in still another aspect, relates to a method of increasing the nuclease resistance of an iRNA agent.
  • the method includes forming an iRNA agent, as described above, wherein the nuclease resistance of the iRNA agent is increased compared to a control composition.
  • Such method includes the steps of incorporating into the iRNA agent, a modification described herein. The modification can be added to one or more of the 5′ or 3′ terminal nucleotides of the sense and antisense strands of the iRNA agent.
  • RNA interference using iRNA agent has been shown to be an effective means for selective gene silencing
  • RNA can have less than desired stability in some bodily fluids, particularly in serum.
  • RNA including iRNA agent, can be degraded between the time it is administered to a subject and the time it enters a target cell. Even within the cell, RNA can undergo rapid degradation by nucleases.
  • Methods of the invention can provide more stable or nuclease resistant iRNA agents that can offer better bioavailability and hence improved effectiveness. This is an improvement over the current insufficient methods for stabilizing iRNA agent against degradation.
  • FIG. 1 a general synthetic scheme for incorporation of RRMS monomers into an oligonucleotide.
  • FIG. 2A is a list of substituents that may be present on silicon in OFG 1 .
  • FIG. 2B is a list of substituents that may be present on the C2′-orthoester group.
  • FIG. 3 is list of representative RRMS cyclic carriers.
  • Panel 1 shows pyrroline-based RRMSs;
  • panel 2 shows 3-hydroxyproline-based RRMSs;
  • panel 3 shows piperidine-based RRMSs;
  • panel 4 shows morpholine and piperazine-based RRMSs;
  • panel 5 shows decalin-based RRMSs.
  • R1 is succinate or phosphoramidate and
  • R2 is H or a conjugate ligand.
  • FIG. 4 is a general reaction scheme for 3′ conjugation of peptide into iRNA.
  • FIG. 5 is a general reaction scheme for 5′ conjugation of peptide into iRNA.
  • FIG. 6 is a general reaction scheme for the synthesis of aza-peptides.
  • FIG. 7 is a general reaction scheme for the synthesis of N-methyl amino acids and peptides.
  • FIG. 8 is a general reaction scheme for the synthesis of ⁇ -methyl amino acids and Ant and Tat peptides.
  • FIG. 9 is a general reaction scheme for the synthesis of Ant and Tat oligocarbamates.
  • FIG. 10 is a a general reaction scheme for the synthesis of Ant and Tat oligoureas.
  • FIG. 11 is a schematic representation of peptide carriers.
  • FIG. 12 shows a gel electrophoretic separation of 3′ phosphorothiate modified dsRNAs following incubation in human serum.
  • FIG. 13 shows a gel electrophoretic separation of 3′ modified dsRNAs following incubation in human serum.
  • FIG. 14 shows a gel electrophoretic separation of 3′ abasic pyrrolidine conjugated dsRNAs following incubation in human serum.
  • FIG. 15 shows a gel electrophoretic separation of 3′ cationic C6 amino dT conjugated dsRNAs following incubation in human serum.
  • FIG. 16 is a graph showing luciferase gene expression activity in cells following transfection of 3′Naproxen modified dsRNAs.
  • FIG. 17 is a graph showing luciferase gene expression activity in cells following transfection of 3′abasic pyrrolidine modified dsRNAs.
  • FIG. 18 is a graph showing luciferase gene expression activity in cells following transfection of 3′ cationic C6 amino dT modified dsRNAs.
  • FIG. 19 shows a gel electrophoretic separation of 3′ naproxen conjugated dsRNAs following incubation in mouse serum.
  • FIG. 20 shows a gel electrophoretic separation of 3′ naproxen conjugated dsRNAs following incubation in liver extract.
  • FIG. 21 is a denaturing gel analysis of the human serum stability assay for AL-DUP-1000.
  • C is the 4 hour time point for siRNA duplex incubated in PBS buffer alone
  • OH— is the partial alkaline hydrolysis marker
  • *s/as represents siRNA duplex containing 5′ end-labeled sense RNA
  • s/*as represents duplex containing 5′ end-labeled antisense RNA.
  • Samples were incubated in 90% human serum and time points were assayed at 10 seconds, 5 min, 15 min, 30 min, 1 hour, 2 hours and 4 hours. Black lines to the right of bands indicate exonucleolytic degradation fragments and the red lines highlight a few of the endonucleolytic degradation fragment.
  • FIG. 22A is a denaturing gel analysis of the human serum stability assay for AL-DUP-1393.
  • C is the 4 hour time point for each siRNA duplex incubated in PBS buffer alone, *s/as represents siRNA duplex containing 5′ end-labeled sense RNA and s/*as represents duplex containing 5′ end-labeled antisense RNA. Samples were assayed at 10 seconds, 15 min, 30 min, 1 hour, 2 hours and 4 hours.
  • FIG. 22B is a denaturing gel analysis of the human serum stability assay for AL-DUP-1329. The lanes are labeled and the experiment was performed as described for FIG. 22A .
  • FIG. 23 is a denaturing gel analysis of AL-DUP-1036, AL-DUP-13ff, and AL-DUP-1363 (see Table 2 for sequences). Black vertical lines highlight regions where exonuclease cleavage is suppressed, stars indicate sites of strong endonucleolytic cleavage in the antisense strand and weaker endonucleolytic cleavage in the sense strand.
  • C is the 4 hour time point for each siRNA duplex incubated in PBS buffer alone, *s/as represents siRNA duplex containing 5′ end-labeled sense RNA and s/*as represents duplex containing 5′ end-labeled antisense RNA. Samples were assayed at 10 seconds, 15 min, 30 min, 1 hour, 2 hours and 4 hours.
  • FIG. 24 Human serum stability profile of siRNA duplexes containing cationic modifications. Denaturing gel analysis of AL-DUP-10aa (alkylamino-dT), AL-DUP-1ccc (abasic pyrrolidine cationic), and AL-DUP-1403 (see Table 3 for sequences). Black line highlights region where exonuclease cleavage is suppressed and red star indicates site of strong endonucleolytic cleavage in the antisense strand.
  • AL-DUP-10aa alkylamino-dT
  • AL-DUP-1ccc abasic pyrrolidine cationic
  • AL-DUP-1403 see Table 3 for sequences. Black line highlights region where exonuclease cleavage is suppressed and red star indicates site of strong endonucleolytic cleavage in the antisense strand.
  • C is the 4 hour time point for each siRNA duplex incubated in PBS buffer alone, *s/as represents siRNA duplex containing 5′ end-labeled sense RNA and s/*as represents duplex containing 5′ end-labeled antisense RNA. Samples were assayed at 10 seconds, 15 min, 30 min, 1 hour, 2 hours and 4 hours.
  • FIG. 25 is a denaturing gel analysis of the human serum stability assay for AL-DUP-1069.
  • the black vertical line highlights the region where exonuclease cleavage is suppressed.
  • C is the 4 hour time point for each siRNA duplex incubated in PBS buffer alone, *s/as represents siRNA duplex containing 5′ end-labeled sense RNA and s/*as represents duplex containing 5′ end-labeled antisense RNA. Samples were assayed at 10 seconds, 15 min, 30 min, 1 hour, 2 hours and 4 hours.
  • the invention is based, at least in part, on the discovery that particular modifications to an iRNA agent are effective at increasing resistance of the iRNA agent to exonucleolytic degradation.
  • the invention features iRNA agents that contain modifications that increase resistance of the iRNA agents to exonucleolytic degradation, including 5′ exonucleolytic and 3′ exonucleolytic degradation. These iRNA agents may have improved stability in cells and biological fluids.
  • the iRNA agents having improved stability can include one or more of various modifications.
  • the iRNA agent can include a backbone modification to one or more of the terminal nucleotides on an iRNA strand.
  • Exemplary backbone modifications include phosphorothioate linkages or P-alkyl linkages between one or more of the terminal nucleotides of an iRNA agent.
  • One or more terminal nucleotides of an iRNA agent can also include a sugar modification, e.g., a 2′ or 3′ sugar modification. Exemplary sugar modifications are described below.
  • An iRNA agent can include multiple modifications that confer an enhanced resistance to exonucleolytic cleavage.
  • one or more terminal nucleotides of an iRNA agent can have a phosphorothioate linkage and a 2′ sugar modification.
  • An iRNA agent having increased resistance to exonucleolytice degradation can include a nucleobase modification, a cationic modification on one or more of the terminal nucleotides of the iRNA agent.
  • R can also have the values for structures 1A-26.
  • the iRNA agent can include a conjugate (also called a ligand) on the 5′ or 3′ terminus of the sense or antisense strand.
  • a conjugate also called a ligand
  • exemplary ligands are shown below.
  • the conjugate can be attached to the 3′ or 5′ end.
  • a cholesterol conjugate is on the 5′ or 3′ end of the sense strand.
  • a cholesterol conjugate is not on the antisense strand.
  • Double-stranded directs the sequence-specific silencing of mRNA through a process known as RNA interference (RNAi).
  • RNAi RNA interference
  • the process occurs in a wide variety of organisms, including mammals and other vertebrates.
  • 21-23 nt fragments of dsRNA are sequence-specific mediators of RNA silencing, e.g., by causing RNA degradation. While not wishing to be bound by theory, it may be that a molecular signal, which may be merely the specific length of the fragments, present in these 21-23 nt fragments recruits cellular factors that mediate RNAi. Described herein are methods for preparing and administering these 21-23 nt fragments, and other iRNAs agents, and their use for specifically inactivating gene function.
  • iRNAs agents or recombinantly produced or chemically synthesized oligonucleotides of the same or similar nature
  • oligonucleotides of the same or similar nature
  • longer dsRNA agent fragments can also be used, e.g., as described below.
  • the length of the iRNA agent strands in an sRNA agent can be less than 31, 30, 28, 25, or 23 nt, e.g., sufficiently short to avoid inducing a deleterious interferon response.
  • a composition of sRNA agent e.g., formulated as described herein
  • use of a discrete species of iRNA agent can be used to selectively target one allele of a target gene, e.g., in a subject heterozygous for the allele.
  • the subject is a mammal such as a cow, horse, mouse, rat, dog, pig, goat, or a primate.
  • the subject can be a dairy mammal (e.g., a cow, or goat) or other farmed animal (e.g., a chicken, turkey, sheep, pig, fish, shrimp).
  • the subject is a human, e.g., a normal individual or an individual that has, is diagnosed with, or is predicted to have a disease or disorder.
  • halo refers to any radical of fluorine, chlorine, bromine or iodine.
  • alkyl refers to a hydrocarbon chain that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, C 1 -C 12 alkyl indicates that the group may have from 1 to 12 (inclusive) carbon atoms in it.
  • haloalkyl refers to an alkyl in which one or more hydrogen atoms are replaced by halo, and includes alkyl moieties in which all hydrogens have been replaced by halo (e.g., perfluoroalkyl). Alkyl and haloalkyl groups may be optionally inserted with O, N, or S.
  • aralkyl refers to an alkyl moiety in which an alkyl hydrogen atom is replaced by an aryl group.
  • Aralkyl includes groups in which more than one hydrogen atom has been replaced by an aryl group. Examples of “aralkyl” include benzyl, 9-fluorenyl, benzhydryl, and trityl groups.
  • alkenyl refers to a straight or branched hydrocarbon chain containing 2-8 carbon atoms and characterized in having one or more double bonds. Examples of a typical alkenyl include, but not limited to, allyl, propenyl, 2-butenyl, 3-hexenyl and 3-octenyl groups.
  • alkynyl refers to a straight or branched hydrocarbon chain containing 2-8 carbon atoms and characterized in having one or more triple bonds. Some examples of a typical alkynyl are ethynyl, 2-propynyl, and 3-methylbutynyl, and propargyl.
  • the sp 2 and sp 3 carbons may optionally serve as the point of attachment of the alkenyl and alkynyl groups, respectively.
  • alkylamino and dialkylamino refer to —NH(alkyl) and —N (alkyl) 2 radicals respectively.
  • aralkylamino refers to a —NH(aralkyl) radical.
  • alkoxy refers to an —O-alkyl radical
  • cycloalkoxy and “aralkoxy” refer to an —O-cycloalkyl and O-aralkyl radicals respectively.
  • sioxy refers to a R 3 SiO— radical.
  • mercapto refers to an SH radical.
  • thioalkoxy refers to an —S-alkyl radical.
  • alkylene refers to a divalent alkyl (i.e., —R—), e.g., —CH 2 —, —CH 2 CH 2 —, and —CH 2 CH 2 CH 2 —.
  • alkylenedioxo refers to a divalent species of the structure —O—R—O—, in which R represents an alkylene.
  • aryl refers to an aromatic monocyclic, bicyclic, or tricyclic hydrocarbon ring system, wherein any ring atom can be substituted.
  • aryl moieties include, but are not limited to, phenyl, naphthyl, anthracenyl, and pyrenyl.
  • cycloalkyl as employed herein includes saturated cyclic, bicyclic, tricyclic, or polycyclic hydrocarbon groups having 3 to 12 carbons, wherein any ring atom can be substituted.
  • the cycloalkyl groups herein described may also contain fused rings. Fused rings are rings that share a common carbon-carbon bond or a common carbon atom (e.g., spiro-fused rings). Examples of cycloalkyl moieties include, but are not limited to, cyclohexyl, adamantyl, and norbornyl.
  • heterocyclyl refers to a nonaromatic 3-10 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3,1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein any ring atom can be substituted.
  • the heterocyclyl groups herein described may also contain fused rings.
  • Fused rings are rings that share a common carbon-carbon bond or a common carbon atom (e.g., spiro-fused rings).
  • heterocyclyl include, but are not limited to tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholino, pyrrolinyl and pyrrolidinyl.
  • cycloalkenyl as employed herein includes partially unsaturated, nonaromatic, cyclic, bicyclic, tricyclic, or polycyclic hydrocarbon groups having 5 to 12 carbons, preferably 5 to 8 carbons, wherein any ring atom can be substituted.
  • the cycloalkenyl groups herein described may also contain fused rings. Fused rings are rings that share a common carbon-carbon bond or a common carbon atom (e.g., spiro-fused rings).
  • Examples of cycloalkenyl moieties include, but are not limited to cyclohexenyl, cyclohexadienyl, or norbornenyl.
  • heterocycloalkenyl refers to a partially saturated, nonaromatic 5-10 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein any ring atom can be substituted.
  • the heterocycloalkenyl groups herein described may also contain fused rings.
  • Fused rings are rings that share a common carbon-carbon bond or a common carbon atom (e.g., spiro-fused rings).
  • heterocycloalkenyl include but are not limited to tetrahydropyridyl and dihydropyran.
  • heteroaryl refers to an aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein any ring atom can be substituted.
  • the heteroaryl groups herein described may also contain fused rings that share a common carbon-carbon bond.
  • oxo refers to an oxygen atom, which forms a carbonyl when attached to carbon, an N-oxide when attached to nitrogen, and a sulfoxide or sulfone when attached to sulfur.
  • acyl refers to an alkylcarbonyl, cycloalkylcarbonyl, arylcarbonyl, heterocyclylcarbonyl, or heteroarylcarbonyl substituent, any of which may be further substituted by substituents.
  • substituted refers to a group “substituted” on an alkyl, cycloalkyl, alkenyl, alkynyl, heterocyclyl, heterocycloalkenyl, cycloalkenyl, aryl, or heteroaryl group at any atom of that group.
  • Suitable substituents include, without limitation, alkyl, alkenyl, alkynyl, alkoxy, halo, hydroxy, cyano, nitro, amino, SO 3 H, sulfate, phosphate, perfluoroalkyl, perfluoroalkoxy, methylenedioxy, ethylenedioxy, carboxyl, oxo, thioxo, imino (alkyl, aryl, aralkyl), S(O) n alkyl (where n is 0-2), S(O) n aryl (where n is 0-2), S(O) n heteroaryl (where n is 0-2), S(O) n heterocyclyl (where n is 0-2), amine (mono-, di-, alkyl, cycloalkyl, aralkyl, heteroaralkyl, and combinations thereof), ester (alkyl, aralkyl, heteroaralkyl), amide (mono
  • adeninyl, cytosinyl, guaninyl, thyminyl, and uracilyl refer to radicals of adenine, cytosine, guanine, thymine, and uracil.
  • a “protected” moiety refers to a reactive functional group, e.g., a hydroxyl group or an amino group, or a class of molecules, e.g., sugars, having one or more functional groups, in which the reactivity of the functional group is temporarily blocked by the presence of an attached protecting group.
  • Protecting groups useful for the monomers and methods described herein can be found, e.g., in Greene, T. W., Protective Groups in Organic Synthesis (John Wiley and Sons: New York), 1981, which is hereby incorporated by reference.
  • RNA agent e.g., an iRNA agent, containing a preferred, but nonlimiting ligand-conjugated monomer subunit is presented as formula (II) below and in the scheme in FIG. 1 .
  • the carrier also referred to in some embodiments as a “linker” can be a cyclic or acyclic moiety and includes two “backbone attachment points” (e.g., hydroxyl groups) and a ligand.
  • the ligand can be directly attached (e.g., conjugated) to the carrier or indirectly attached (e.g., conjugated) to the carrier by an intervening tether (e.g., an acyclic chain of one or more atoms; or a nucleobase, e.g., a naturally occurring nucleobase optionally having one or more chemical modifications, e.g., an unusual base; or a universal base).
  • the carrier therefore also includes a “ligand or tethering attachment point” for the ligand and tether/tethered ligand, respectively.
  • the ligand-conjugated monomer subunit may be the 5′ or 3′ terminal subunit of the RNA molecule, i.e., one of the two “W” groups may be a hydroxyl group, and the other “W” group may be a chain of two or more unmodified or modified ribonucleotides.
  • the ligand-conjugated monomer subunit may occupy an internal position, and both “W” groups may be one or more unmodified or modified ribonucleotides.
  • More than one ligand-conjugated monomer subunit may be present in a RNA molecule, e.g., an iRNA agent.
  • Preferred positions for inclusion of a tethered ligand-conjugated monomer subunits e.g., one in which a lipophilic moiety, e.g., cholesterol, is tethered to the carrier are at the 3′ terminus, the 5′ terminus, or an internal position of the sense strand.
  • the modified RNA molecule of formula (II) can be obtained using oligonucleotide synthetic methods known in the art.
  • the modified RNA molecule of formula (II) can be prepared by incorporating one or more of the corresponding monomer compounds (see, e.g., A, B, and C below and in the scheme in FIG. 1 ) into a growing sense or antisense strand, utilizing, e.g., phosphoramidite or H-phosphonate coupling strategies.
  • the monomers e.g., a ligand-conjugated monomer, generally include two differently functionalized hydroxyl groups (OFG 1 and OFG 2 ), which are linked to the carrier molecule (see A below and in FIG. 1 ), and a ligand/tethering attachment point.
  • the term “functionalized hydroxyl group” means that the hydroxyl proton has been replaced by another substituent.
  • one hydroxyl group (OFG 1 ) on the carrier is functionalized with a protecting group (PG).
  • the other hydroxyl group (OFG 2 ) can be functionalized with either (1) a liquid or solid phase synthesis support reagent (solid circle) directly or indirectly through a linker, L, as in B, or (2) a phosphorus-containing moiety, e.g., a phosphoramidite as in C.
  • the tethering attachment point may be connected to a hydrogen atom, a suitable protecting group, a tether, or a tethered ligand at the time that the monomer is incorporated into the growing sense or antisense strand (see variable “R” in A below).
  • the tethered ligand can be, but need not be attached to the monomer at the time that the monomer is incorporated into the growing strand.
  • the tether, the ligand or the tethered ligand may be linked to a “precursor” ligand-conjugated monomer subunit after a “precursor” ligand-conjugated monomer subunit has been incorporated into the strand.
  • the wavy line used below refers to a connection, and can represent a direct bond between the moiety and the attachment point or a tethering molecule which is interposed between the moiety and the attachment point.
  • Directly tethered means the moiety is bound directly to the attachment point. Indirectly tethered means that there is a tether molecule interposed between the attachment point and the moiety.
  • the (OFG 1 ) protecting group may be selected as desired, e.g., from T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991).
  • the protecting group is preferably stable under amidite synthesis conditions, storage conditions, and oligonucleotide synthesis conditions.
  • Hydroxyl groups, —OH are nucleophilic groups (i.e., Lewis bases), which react through the oxygen with electrophiles (i.e., Lewis acids).
  • Hydroxyl groups in which the hydrogen has been replaced with a protecting group are essentially unreactive as nucleophiles in displacement reactions.
  • a protecting group e.g., a triarylmethyl group or a trialkylsilyl group
  • the protected hydroxyl group is useful in preventing e.g., homocoupling of compounds exemplified by structure C during oligonucleotide synthesis.
  • a preferred protecting group is the dimethoxytrityl group.
  • a preferred protecting group is a silicon-based protecting group having the formula below:
  • X5′, X5′′, and X5′′′ can be selected from substituted or unsubstituted alkyl, cycloalkyl, aryl, araklyl, heteroaryl, alkoxy, cycloalkoxy, aralkoxy, aryloxy, heteroaryloxy, or siloxy (i.e., R 3 SiO—, the three “R” groups can be any combination of the above listed groups).
  • X 5′ , X 5 ′′, and X 5′′′ may all be the same or different; also contemplated is a combination in which two of X 5′ , X 5′′ , and X 5′′′ are identical and the third is different.
  • X 5′ , X 5′′ , and X 5′′′ include those that result in OFG 1 groups that meet the deprotection and stability criteria delineated below.
  • the group is preferably stable under amidite synthesis conditions, storage conditions, and oligonucleotide synthesis conditions. Rapid removal, i.e., less than one minute, of the silyl group from e.g., a support-bound oligonucleotide is desirable because it can reduce synthesis times and thereby reduce exposure time of the growing oligonucleotide chain to the reagents. Oligonucleotide synthesis can be improved if the silyl protecting group is visible during deprotection, e.g., from the addition of a chromophore silyl substituent.
  • silyl protecting groups can be complicated by the competing demands of the essential characteristics of stability and facile removal, and the need to balance these competitive goals. Most substituents that increase stability can also increase the reaction time required for removal of the silyl group, potentially increasing the level of difficulty in removal of the group.
  • alkoxy and siloxy substituents to OFG 1 silicon-containing protecting groups increases the susceptibility of the protecting groups to fluoride cleavage of the silylether bonds. Increasing the steric bulk of the substituents preserves stability while not decreasing fluoride lability to an equal extent. An appropriate balance of substituents on the silyl group makes a silyl ether a viable nucleoside protecting group.
  • Candidate OFG 1 silicon-containing protecting groups may be tested by exposing a tetrahydrofuran solution of a preferred carrier bearing the candidate OFG 1 group to five molar equivalents of tetrahydrofuran at room temperature. The reaction time may be determined by monitoring the disappearance of the starting material by thin layer chromatography.
  • the OFG 2 in B includes a linker, e.g., a relatively long organic linker, connected to a soluble or insoluble support reagent
  • a linker e.g., a relatively long organic linker
  • solution or solid phase synthesis techniques can be employed to build up a chain of natural and/or modified ribonucleotides once OFG 1 is deprotected and free to react as a nucleophile with another nucleoside or monomer containing an electrophilic group (e.g., an amidite group).
  • a natural or modified ribonucleotide or oligoribonucleotide chain can be coupled to monomer C via an amidite group or H-phosphonate group at OFG 2 .
  • OFG 1 can be deblocked, and the restored nucleophilic hydroxyl group can react with another nucleoside or monomer containing an electrophilic group.
  • R′ can be substituted or unsubstituted alkyl or alkenyl. In preferred embodiments, R′ is methyl, allyl or 2-cyanoethyl.
  • R′′ may a C 1 -C 10 alkyl group, preferably it is a branched group containing three or more carbons, e.g., isopropyl.
  • OFG 2 in B can be hydroxyl functionalized with a linker, which in turn contains a liquid or solid phase synthesis support reagent at the other linker terminus.
  • the support reagent can be any support medium that can support the monomers described herein.
  • the monomer can be attached to an insoluble support via a linker, L, which allows the monomer (and the growing chain) to be solubilized in the solvent in which the support is placed.
  • the solubilized, yet immobilized, monomer can react with reagents in the surrounding solvent; unreacted reagents and soluble by-products can be readily washed away from the solid support to which the monomer or monomer-derived products is attached.
  • the monomer can be attached to a soluble support moiety, e.g., polyethylene glycol (PEG) and liquid phase synthesis techniques can be used to build up the chain.
  • PEG polyethylene glycol
  • Linker and support medium selection is within skill of the art.
  • the linker may be —C(O)(CH 2 ) q C(O)—, or —C(O)(CH 2 ) q S—, in which q can be 0, 1, 2, 3, or 4; preferably, it is oxalyl, succinyl or thioglycolyl.
  • Standard control pore glass solid phase synthesis supports can not be used in conjunction with fluoride labile 5′ silyl protecting groups because the glass is degraded by fluoride with a significant reduction in the amount of full-length product.
  • Fluoride-stable polystyrene based supports or PEG are preferred.
  • the ligand/tethering attachment point can be any divalent, trivalent, tetravalent, pentavalent or hexavalent atom.
  • ligand/tethering attachment point can be a carbon, oxygen, nitrogen or sulfur atom.
  • a ligand/tethering attachment point precursor functional group can have a nucleophilic heteroatom, e.g., —SH, —NH 2 , secondary amino, ONH 2 , or NH 2 NH 2 .
  • the ligand/tethering attachment point precursor functional group can be an electrophilic moiety, e.g., an aldehyde.
  • the ligand/tethering attachment point can be an endocyclic atom (i.e., a constituent atom in the cyclic moiety, e.g., a nitrogen atom) or an exocyclic atom (i.e., an atom or group of atoms attached to a constituent atom in the cyclic moiety).
  • the carrier can be any organic molecule containing attachment points for OFG 1 , OFG 2 , and the ligand.
  • carrier is a cyclic molecule and may contain heteroatoms (e.g., O, N or S).
  • carrier molecules may include aryl (e.g., benzene, biphenyl, etc.), cycloalkyl (e.g., cyclohexane, cis or trans decalin, etc.), cycloalkenyl (e.g., cyclohexenyl), or heterocyclyl (tetrahydropyran, piperazine, pyrrolidine, etc.).
  • the carrier can be an acyclic moiety, e.g., based on serinol. Any of the above cyclic systems may include substituents in addition to OFG 1 , OFG 2 , and the ligand.
  • the carrier molecule is an oxygen containing heterocycle.
  • the carrier is a ribose sugar as shown in structure LCM-I.
  • the monomer e.g., a ligand-conjugated monomer is a nucleoside.
  • B represents a nucleobase, e.g., a naturally occurring nucleobase optionally having one or more chemical modifications, e.g., and unusual base; or a universal base.
  • nucleobase can include any one of the following:
  • a universal base can form base pairs with each of the natural DNA/RNA bases, exhibiting relatively little discrimination between them.
  • the universal bases are non-hydrogen bonding, hydrophobic, aromatic moieties which can stabilize e.g., duplex RNA or RNA-like molecules, via stacking interactions.
  • a universal base can also include hydrogen bonding substituents.
  • a “universal base” can include anthracenes, pyrenes or any one of the following:
  • B can form part of a tether that connects a ligand to the carrier.
  • the double bond is trans
  • the ligand is a substituted or unsubstituted cholesterolyl radical (e.g., attached through the D-ring side chain or the C-3 hydroxyl); an aralkyl moiety having at least one sterogenic center and at least one substituent on the aryl portion of the aralkyl group; or a nucleobase.
  • B in the tether described above, is uracilyl or a universal base, e.g., an aryl moiety, e.g., phenyl, optionally having additional substituents, e.g., one or more fluoro groups. B can be substituted at any atom with the remainder of the tether.
  • X 2 can include “oxy” or “deoxy” substituents in place of the 2′-OH; or be a ligand or a tethered ligand.
  • Preferred orthoesters have the general formula J.
  • the groups R 31 and R 32 may be the same or different and can be any combination of the groups listed in FIG. 2B .
  • a preferred orthoester is the “ACE” group, shown below as structure K.
  • X 3 is as described for OFG 2 above.
  • PG can be a triarylmethyl group (e.g., a dimethoxytrityl group) or Si(X 5′ )(X 5′′ )(X 5′′′ ) in which (X 5′ ), (X 5′′ ), and (X 5′′′ ) are as described elsewhere.
  • a triarylmethyl group e.g., a dimethoxytrityl group
  • Si(X 5′ )(X 5′′ )(X 5′′′ ) in which (X 5′ ), (X 5′′ ), and (X 5′′′ ) are as described elsewhere.
  • Cyclic sugar replacement-based monomers e.g., sugar replacement-based ligand-conjugated monomers, are also referred to herein as ribose replacement monomer subunit (RRMS) monomer compounds.
  • Preferred carriers have the general formula (LCM-2) provided below (In that structure preferred backbone attachment points can be chosen from R 1 or R 2 ; R 3 or R 4 ; or R 9 and R 10 if Y is CR 9 R 10 (two positions are chosen to give two backbone attachment points, e.g., R 1 and R 4 , or R 4 and R 9 )).
  • Preferred tethering attachment points include R 7 ; R 5 or R 6 when X is CH 2 .
  • the carriers are described below as an entity, which can be incorporated into a strand.
  • the structures also encompass the situations wherein one (in the case of a terminal position) or two (in the case of an internal position) of the attachment points, e.g., R 1 or R 2 ; R 3 or R 4 ; or R 9 or R 10 (when Y is CR 9 R 10 ), is connected to the phosphate, or modified phosphate, e.g., sulfur containing, backbone.
  • one of the above-named R groups can be —CH 2 —, wherein one bond is connected to the carrier and one to a backbone atom, e.g., a linking oxygen or a central phosphorus atom.
  • the carrier may be based on the pyrroline ring system or the 4-hydroxyproline ring system, e.g., X is N(CO)R 7 or NR 7 , Y is CR 9 R 10 , and Z is absent (D).
  • OFG 1 is preferably attached to a primary carbon, e.g., an exocyclic alkylene group, e.g., a methylene group, connected to one of the carbons in the five-membered ring (—CH 2 OFG 1 in D).
  • OFG 2 is preferably attached directly to one of the carbons in the five-membered ring (—OFG 2 in D).
  • —CH 2 OFG 1 may be attached to C-2 and OFG 2 may be attached to C-3; or —CH 2 OFG 1 may be attached to C-3 and OFG 2 may be attached to C-4.
  • CH 2 OFG 1 and OFG 2 may be geminally substituted to one of the above-referenced carbons.
  • —CH 2 OFG 1 may be attached to C-2 and OFG 2 may be attached to C-4.
  • the pyrroline- and 4-hydroxyproline-based monomers may therefore contain linkages (e.g., carbon-carbon bonds) wherein bond rotation is restricted about that particular linkage, e.g. restriction resulting from the presence of a ring.
  • CH 2 OFG 1 and OFG 2 may be cis or trans with respect to one another in any of the pairings delineated above Accordingly, all cis/trans isomers are expressly included.
  • the monomers may also contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of the monomers are expressly included (e.g., the centers bearing CH 2 OFG 1 and OFG 2 can both have the R configuration; or both have the S configuration; or one center can have the R configuration and the other center can have the S configuration and vice versa).
  • the tethering attachment point is preferably nitrogen.
  • Preferred examples of carrier D include the following:
  • the carrier may be based on the piperidine ring system (E), e.g., X is N(CO)R 7 or NR 7 , Y is CR 9 R 10 , and Z is CR 11 R 12 .
  • OFG 2 is preferably attached directly to one of the carbons in the six-membered ring (—OFG 2 in E).
  • —(CH 2 ) n OFG 1 and OFG 2 may be disposed in a geminal manner on the ring, i.e., both groups may be attached to the same carbon, e.g., at C-2, C-3, or C-4.
  • —(CH 2 ) n OFG 1 and OFG 2 may be disposed in a vicinal manner on the ring, i.e., both groups may be attached to adjacent ring carbon atoms, e.g.,
  • —(CH 2 ) n OFG 1 may be attached to C-2 and OFG 2 may be attached to C-3; —(CH 2 ) n OFG 1 may be attached to C-3 and OFG 2 may be attached to C-2; —(CH 2 ) n OFG 1 may be attached to C-3 and OFG 2 may be attached to C-4; or —(CH 2 ) n OFG 1 may be attached to C-4 and OFG 2 may be attached to C-3.
  • the piperidine-based monomers may therefore contain linkages (e.g., carbon-carbon bonds) wherein bond rotation is restricted about that particular linkage, e.g. restriction resulting from the presence of a ring.
  • —(CH 2 ) n OFG 1 and OFG 2 may be cis or trans with respect to one another in any of the pairings delineated above. Accordingly, all cis/trans isomers are expressly included.
  • the monomers may also contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of the monomers are expressly included (e.g., the centers bearing CH 2 OFG 1 and OFG 2 can both have the R configuration; or both have the S configuration; or one center can have the R configuration and the other center can have the S configuration and vice versa).
  • the tethering attachment point is preferably nitrogen.
  • the carrier may be based on the piperazine ring system (F), e.g., X is N(CO)R 7 or NR 7 , Y is NR 8 , and Z is CR 11 R 12 , or the morpholine ring system (G), e.g., X is N(CO)R 7 or NR 7 , Y is O, and Z is CR 11 R 12 .
  • OFG 1 is preferably attached to a primary carbon, e.g., an exocyclic alkylene group, e.g., a methylene group, connected to one of the carbons in the six-membered ring (—CH 2 OFG 1 in F or G).
  • OFG 2 is preferably attached directly to one of the carbons in the six-membered rings (—OFG 2 in F or G).
  • —CH 2 OFG 1 may be attached to C-2 and OFG 2 may be attached to C-3; or vice versa.
  • CH 2 OFG 1 and OFG 2 may be geminally substituted to one of the above-referenced carbons.
  • the piperazine- and morpholine-based monomers may therefore contain linkages (e.g., carbon-carbon bonds) wherein bond rotation is restricted about that particular linkage, e.g. restriction resulting from the presence of a ring.
  • CH 2 OFG 1 and OFG 2 may be cis or trans with respect to one another in any of the pairings delineated above. Accordingly, all cis/trans isomers are expressly included.
  • the monomers may also contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of the monomers are expressly included (e.g., the centers bearing CH 2 OFG 1 and OFG 2 can both have the R configuration; or both have the S configuration; or one center can have the R configuration and the other center can have the S configuration and vice versa).
  • R′′′ can be, e.g., C 1 -C 6 alkyl, preferably CH 3 .
  • the tethering attachment point is preferably nitrogen in both F and G.
  • OFG 2 is preferably attached directly to one of C-2, C-3, C-4, or C-5 (—OFG 2 in H).
  • —(CH 2 ) n OFG 1 and OFG 2 may be disposed in a geminal manner on the ring, i.e., both groups may be attached to the same carbon, e.g., at C-2, C-3, C-4, or C-5.
  • —(CH 2 ) n OFG 1 and OFG 2 may be disposed in a vicinal manner on the ring, i.e., both groups may be attached to adjacent ring carbon atoms, e.g., —(CH 2 ) n OFG 1 may be attached to C-2 and OFG 2 may be attached to C-3; —(CH 2 ) n OFG 1 may be attached to C-3 and OFG 2 may be attached to C-2; —(CH 2 ) n OFG 1 may be attached to C-3 and OFG 2 may be attached to C-4; or —(CH 2 ) n OFG 1 may be attached to C-4 and OFG 2 may be attached to C-3; —(CH 2 ) n OFG 1 may be attached to C-4 and OFG 2 may be attached to C-5; or —(CH 2 ) n OFG 1 may be attached to C-5 and OFG 2 may be attached to C-4.
  • the decalin or indane-based monomers may therefore contain linkages (e.g., carbon-carbon bonds) wherein bond rotation is restricted about that particular linkage, e.g. restriction resulting from the presence of a ring.
  • linkages e.g., carbon-carbon bonds
  • —(CH 2 ) n OFG 1 and OFG 2 may be cis or trans with respect to one another in any of the pairings delineated above. Accordingly, all cis/trans isomers are expressly included.
  • the monomers may also contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures.
  • the centers bearing CH 2 OFG 1 and OFG 2 can both have the R configuration; or both have the S configuration; or one center can have the R configuration and the other center can have the S configuration and vice versa).
  • the substituents at C-1 and C-6 are trans with respect to one another.
  • the tethering attachment point is preferably C-6 or C-7.
  • —(CH 2 ) n OFG 1 and OFG 2 may be cis or trans with respect to one another. Accordingly, all cis/trans isomers are expressly included.
  • the monomers may also contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of the monomers are expressly included (e.g., the centers bearing CH 2 OFG 1 and OFG 2 can both have the R configuration; or both have the S configuration; or one center can have the R configuration and the other center can have the S configuration and vice versa).
  • the tethering attachment point is preferably nitrogen.
  • FIG. 3 Representative cyclic, sugar replacement-based carriers are shown in FIG. 3 .
  • Acyclic sugar replacement-based monomers e.g., sugar replacement-based ligand-conjugated monomers
  • RRMS ribose replacement monomer subunit
  • Preferred acyclic carriers can have formula LCM-3 or LCM-4 below.
  • each of x, y, and z can be, independently of one another, 0, 1, 2, or 3.
  • the tertiary carbon can have either the R or S configuration.
  • x is zero and y and z are each 1 in formula LCM-3 (e.g., based on serinol), and y and z are each 1 in formula LCM-3.
  • Each of formula LCM-3 or LCM-4 below can optionally be substituted, e.g., with hydroxy, alkoxy, perhaloalkyl.
  • a moiety e.g., a ligand may be connected indirectly to the carrier via the intermediacy of an intervening tether.
  • Tethers are connected to the carrier at a tethering attachment point (TAP) and may include any C 1 -C 100 carbon-containing moiety, (e.g. C 1 -C 75 , C 1 -C 50 , C 1 -C 20 , C 1 -C 10 ; C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , or C 10 ), preferably having at least one nitrogen atom.
  • TAP tethering attachment point
  • the nitrogen atom forms part of a terminal amino or amido (NHC(O)—) group on the tether, which may serve as a connection point for the ligand.
  • Preferred tethers include TAP -(CH 2 ) n NH— ; TAP- C(O)(CH 2 ) n NH— ; TAP- NR′′′′(CH 2 ) n NH— , TAP- C(O—(CH 2 ) n —C(O)— ; TAP- C(O)—(CH 2 ) n —C(O)O— ; TAP- C(O)—O— ; TAP- C(O)— (CH 2 ) n —NH—C(O)— ; TAP- C(O)—(CH 2 ) n — ; TAP- C(O)—NH— ; TAP- C(O)— ; TAP- (CH 2 ) n —C(
  • n is 5, 6, or 11.
  • the nitrogen may form part of a terminal oxyamino group, e.g., —ONH 2 , or hydrazino group, —NHNH 2 .
  • the tether may optionally be substituted, e.g., with hydroxy, alkoxy, perhaloalkyl, and/or optionally inserted with one or more additional heteroatoms, e.g., N, O, or S.
  • Preferred tethered ligands may include, e.g., TAP- (CH 2 ) n NH(LIGAND) ; TAP- C(O)(CH 2 ) n NH(LIGAND) ; TAP- NR′′′′(CH 2 ) n NH(LIGAND) ; TAP- (CH 2 ) n ONH(LIGAND) ; TAP- C(O)(CH 2 ) n ONH(LIGAND) ; TAP- NR′′′′(CH 2 ) n ONH(LIGAND) ; TAP- (CH 2 ) n NHNH 2 (LIGAND) , TAP- C(O)(CH 2 ) n NHNH 2 (LIGAND) ; TAP- NR′′′′(CH 2 ) n —NHNH 2 (LIGAND) ; TAP- C(O)—(CH 2 ) n —C(O)(LIGAND) ; TAP- C
  • amino terminated tethers e.g., NH 2 , ONH 2 , NH 2 NH 2
  • amino terminated tethers can form an imino bond (i.e., C ⁇ N) with the ligand.
  • amino terminated tethers e.g., NH 2 , ONH 2 , NH 2 NH 2
  • the tether may optionally be substituted, e.g., with hydroxy, alkoxy, perhaloalkyl, and/or optionally inserted with one or more additional heteroatoms, e.g., N, O, or S.
  • the double bond can be cis or trans or E or Z.
  • the tether may include an electrophilic moiety, preferably at the terminal position of the tether.
  • electrophilic moieties include, e.g., an aldehyde, alkyl halide, mesylate, tosylate, nosylate, or brosylate, or an activated carboxylic acid ester, e.g. an NHS ester, or a pentafluorophenyl ester.
  • Preferred tethers include TAP- (CH 2 ) n CHO ; TAP- C(O)(CH 2 ) n CHO ; or TAP- NR′′′′(CH 2 ) n CHO , in which n is 1-6 and R′′′′ is C 1 -C 6 alkyl; or TAP- (CH 2 ) n C(O)ONHS ; TAP- C(O)(CH 2 ) n C(O)ONHS ; or TAP- NR′′′′(CH 2 ) n C(O)ONHS , in which n is 1-6 and R′′′′ is C 1 -C 6 alkyl; TAP- (CH 2 ) n C(O)OC 6 F 5 ; TAP- C(O)(CH 2 ) n C(O) OC 6 F 5 ; or TAP-NR′′′′(CH 2 ) n C(O)OC 6 F 5 , in which n is 1-11 and R′′′′ is C 1
  • the monomer can include a phthalimido group (K) at the terminal position of the tether.
  • other protected amino groups can be at the terminal position of the tether, e.g., alloc, monomethoxy trityl (MMT), itrifluoroacetyl, Fmoc, or aryl sulfonyl (e.g., the aryl portion can be ortho-nitrophenyl or ortho, para-dinitrophenyl).
  • MMT monomethoxy trityl
  • itrifluoroacetyl itrifluoroacetyl
  • Fmoc itrifluoroacetyl
  • aryl sulfonyl e.g., the aryl portion can be ortho-nitrophenyl or ortho, para-dinitrophenyl.
  • a wide variety of entities can be tethered to an iRNA agent, e.g., to the carrier of a ligand-conjugated monomer subunit. Examples are described below in the context of a ligand-conjugated monomer subunit but that is only preferred, entities can be coupled at other points to an iRNA agent.
  • Preferred moieties are ligands, which are coupled, preferably covalently, either directly or indirectly via an intervening tether, to the carrier.
  • the ligand is attached to the carrier via an intervening tether.
  • the ligand or tethered ligand may be present on the ligand-conjugated monomer ⁇ when the ligand-conjugated monomer is incorporated into the growing strand.
  • the ligand may be incorporated into a “precursor” ligand-conjugated monomer subunit after a “precursor” ligand-conjugated monomer subunit has been incorporated into the growing strand.
  • a monomer having, e.g., an amino-terminated tether, e.g., TAP-(CH 2 ) n NH 2 may be incorporated into a growing sense or antisense strand.
  • a ligand having an electrophilic group e.g., a pentafluorophenyl ester or aldehyde group, can subsequently be attached to the precursor ligand-conjugated monomer by coupling the electrophilic group of the ligand with the terminal nucleophilic group of the precursor ligand-conjugated monomer subunit tether.
  • a ligand alters the distribution, targeting or lifetime of an iRNA agent into which it is incorporated.
  • a ligand provides an enhanced affinity for a selected target, e.g, molecule, cell or cell type, compartment, e.g., a cellular or organ compartment, tissue, organ or region of the body, as, e.g., compared to a species absent such a ligand.
  • Preferred ligands can improve transport, hybridization, and specificity properties and may also improve nuclease resistance of the resultant natural or modified oligoribonucleotide, or a polymeric molecule comprising any combination of monomers described herein and/or natural or modified ribonucleotides.
  • Ligands in general can include therapeutic modifiers, e.g., for enhancing uptake; diagnostic compounds or reporter groups e.g., for monitoring distribution; cross-linking agents; nuclease-resistance conferring moieties; and natural or unusual nucleobases.
  • Lipophiles examples include lipophiles, lipids, steroids (e.g., cholesterol, uvaol, hecigenin, diosgenin), terpenes (e.g., triterpenes, e.g., sarsasapogenin, Friedelin, epifriedelanol derivatized lithocholic acid), vitamins (e.g., folic acid, vitamin A, vitamin E, biotin, pyridoxal), carbohydrates, proteins, protein binding agents, integrin targeting molecules, polycationics, peptides, polyamines, and peptide mimics.
  • steroids e.g., cholesterol, uvaol, hecigenin, diosgenin
  • terpenes e.g., triterpenes, e.g., sarsasapogenin, Friedelin, epifriedelanol derivatized lithocholic acid
  • vitamins e.g., folic acid,
  • the ligand can be a peptide or peptidomimetic.
  • a peptidomimetic also referred to herein as an oligopeptidomimetic is a molecule capable of folding into a defined three-dimensional structure similar to a natural peptide.
  • the attachment of peptide and peptidomimetics to iRNA agents can affect pharmacokinetic distribution of the iRNA, such as by enhancing cellular recognition and absorption.
  • the peptide or peptidomimetic moiety can be about 5-50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
  • the ligand can be one of the following triterpenes:
  • the ligand can be substituted or unsubstituted cholesterol, or a stereoisomer thereof or one of the following steroids:
  • the monomers described herein can be used to make oligonucleotides which are useful as iRNA agents, e.g., RNA molecules, (double-stranded; single-stranded) that mediate RNAi, e.g., with respect to an endogenous gene of a subject or to a gene of a pathogen.
  • the iRNA agent will incorporate momomers described herein together with naturally occuring nucleosides or nucleotides or with other modified nucleosides or nucleotides.
  • the modified monomers can be present at any position in the iRNA agent, e.g., at the terminii or in the middle region of an iRNA agent or in a duplex region or in an unpaired region.
  • iRNA agent can have any architecture, e.g., architecture described herein. E.g., it can be incorporated into an iRNA agent having an overhang structure, a hairpin or other single strand structure or a two-strand structure, as described herein.
  • RNA agent is an unmodified RNA, modified RNA, or nucleoside surrogate, all of which are defined herein (see, e.g., the section below entitled RNA Agents). While numerous modified RNAs and nucleoside surrogates are described, preferred examples include those which have greater resistance to nuclease degradation than do unmodified RNAs. Preferred examples include those which have a 2′ sugar modification, a modification in a single strand overhang, preferably a 3′ single strand overhang, or, particularly if single stranded, a 5′ modification which includes one or more phosphate groups or one or more analogs of a phosphate group.
  • RNA agent is an RNA agent which can, or which can be cleaved into an RNA agent which can, down regulate the expression of a target gene, preferably an endogenous or pathogen target RNA. While not wishing to be bound by theory, an iRNA agent may act by one or more of a number of mechanisms, including post-transcriptional cleavage of a target mRNA sometimes referred to in the art as RNAi, or pre-transcriptional or pre-translational mechanisms.
  • An iRNA agent can include a single strand or can include more than one strands, e.g., it can be a double stranded iRNA agent. If the iRNA agent is a single strand it is particularly preferred that it include a 5′ modification which includes one or more phosphate groups or one or more analogs of a phosphate group.
  • the RRMS-containing iRNA agent should include a region of sufficient homology to the target gene, and be of sufficient length in terms of nucleotides, such that the iRNA agent, or a fragment thereof, can mediate down regulation of the target gene.
  • nucleotide or ribonucleotide is sometimes used herein in reference to one or more monomeric subunits of an RNA agent.
  • ribonucleotide or “nucleotide”, herein can, in the case of a modified RNA or nucleotide surrogate, also refer to a modified nucleotide, or surrogate replacement moiety at one or more positions.
  • the iRNA agent is or includes a region which is at least partially, and in some embodiments fully, complementary to the target RNA.
  • RNAi cleavage of the target RNA e.g., mRNA.
  • an iRNA agent will often be modified or include nucleoside surrogates in addition to the ribose replacement modification subunit (RRMS).
  • Single stranded regions of an iRNA agent will often be modified or include nucleoside surrogates, e.g., the unpaired region or regions of a hairpin structure, e.g., a region which links two complementary regions, can have modifications or nucleoside surrogates.
  • Modification to stabilize one or more 3′- or 5′-terminus of an iRNA agent, e.g., against exonucleases, or to favor the antisense sRNA agent to enter into RISC are also favored.
  • Modifications can include C3 (or C6, C7, C12) amino linkers, thiol linkers, carboxyl linkers, non-nucleotidic spacers (C3, C6, C9, C12, abasic, triethylene glycol, hexaethylene glycol), special biotin or fluorescein reagents that come as phosphoramidites and that have another DMT-protected hydroxyl group, allowing multiple couplings during RNA synthesis.
  • an iRNA agent will preferably have one or more of the following properties:
  • a “single strand iRNA agent” as used herein, is an iRNA agent which is made up of a single molecule. It may include a duplexed region, formed by intra-strand pairing, e.g., it may be, or include, a hairpin or pan-handle structure.
  • Single strand iRNA agents are preferably antisense with regard to the target molecule.
  • single strand iRNA agents are 5′ phosphorylated or include a phosphoryl analog at the 5′ prime terminus. 5′-phosphate modifications include those which are compatible with RISC mediated gene silencing.
  • Suitable modifications include: 5′-monophosphate ((HO)2(O)P—O-5′); 5′-diphosphate ((HO) 2 (O)P—O—P(HO)(O)—O-5′); 5′-triphosphate ((HO)2(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-guanosine cap (7-methylated or non-methylated) (7m-G-O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-adenosine cap (Appp), and any modified or unmodified nucleotide cap structure (N—O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-monothiophosphate (phosphorothioate; (HO)2(S)P—O-5′); 5′
  • the antisense and sense strands of a double strand iRNA agent may be desirable to modify one or both of the antisense and sense strands of a double strand iRNA agent. In some cases they will have the same modification or the same class of modification but in other cases the sense and antisense strand will have different modifications, e.g., in some cases it is desirable to modify only the sense strand. It may be desirable to modify only the sense strand, e.g., to inactivate it, e.g., the sense strand can be modified in order to inactivate the sense strand and prevent formation of an active sRNA/protein or RISC.
  • Other modifications which prevent phosphorylation can also be used, e.g., simply substituting the 5′-OH by H rather than O-Me.
  • Antisense strand modifications include 5′ phosphorylation as well as any of the other 5′ modifications discussed herein, particularly the 5′ modifications discussed above in the section on single stranded iRNA molecules.
  • a ds iRNA agent contains sense and antisense strands, preferable paired to contain an overhang, e.g., one or two 5′ or 3′ overhangs but preferably a 3′ overhang of 2-3 nucleotides. Most embodiments will have a 3′ overhang.
  • Preferred sRNA agents will have single-stranded overhangs, preferably 3′ overhangs, of 1 or preferably 2 or 3 nucleotides in length at each end. The overhangs can be the result of one strand being longer than the other, or the result of two strands of the same length being staggered. 5′ ends are preferably phosphorylated.
  • Preferred lengths for the duplexed region is between 15 and 30, most preferably 18, 19, 20, 21, 22, and 23 nucleotides in length, e.g., in the sRNA agent range discussed above.
  • sRNA agents can resemble in length and structure the natural Dicer processed products from long dsRNAs.
  • Embodiments in which the two strands of the sRNA agent are linked, e.g., covalently linked are also included. Hairpin, or other single strand structures which provide the required double stranded region, and preferably a 3′ overhang are also within the invention.
  • nucleic acids are polymers of subunits or monomers
  • many of the modifications described below occur at a position which is repeated within a nucleic acid, e.g., a modification of a base, or a phosphate moiety, or the a non-linking 0 of a phosphate moiety.
  • the modification will occur at all of the subject positions in the nucleic acid but in many, and infact in most cases it will not.
  • a modification may only occur at a 3′ or 5′ terminal position, may only occur in a terminal regions, e.g. at a position on a terminal nucleotide or in the last 2, 3, 4, 5, or 10 nucleotides of a strand.
  • a modification may occur in a double strand region, a single strand region, or in both.
  • a modification may occur only in the double strand region of an RNA or may only occur in a single strand region of an RNA.
  • a phosphorothioate modification at a non-linking O position may only occur at one or both termini, may only occur in a terminal regions, e.g., at a position on a terminal nucleotide or in the last 2, 3, 4, 5, or 10 nucleotides of a strand, or may occur in double strand and single strand regions, particularly at termini.
  • the 5′ end or ends can be phosphorylated.
  • all or some of the bases in a 3′ or 5′ overhang will be modified, e.g., with a modification described herein.
  • Modifications can include, e.g., the use of modifications at the 2′ OH group of the ribose sugar, e.g., the use of deoxyribonucleotides, e.g., deoxythymidine, instead of ribonucleotides, and modifications in the phosphate group, e.g., phosphothioate modifications. Overhangs need not be homologous with the target sequence.
  • the scaffold presented above in Formula 1 represents a portion of a ribonucleic acid.
  • the basic components are the ribose sugar, the base, the terminal phosphates, and phosphate internucleotide linkers.
  • the bases are naturally occurring bases, e.g., adenine, uracil, guanine or cytosine
  • the sugars are the unmodified 2′ hydroxyl ribose sugar (as depicted) and W, X, Y, and Z are all O
  • Formula 1 represents a naturally occurring unmodified oligoribonucleotide.
  • Unmodified oligoribonucleotides may be less than optimal in some applications, e.g., unmodified oligoribonucleotides can be prone to degradation by e.g., cellular nucleases. Nucleases can hydrolyze nucleic acid phosphodiester bonds. However, chemical modifications to one or more of the above RNA components can confer improved properties, and, e.g., can render oligoribonucleotides more stable to nucleases. Umodified oligoribonucleotides may also be less than optimal in terms of offering tethering points for attaching ligands or other moieties to an iRNA agent.
  • Modified nucleic acids and nucleotide surrogates can include one or more of:
  • the actual electronic structure of some chemical entities cannot be adequately represented by only one canonical form (i.e. Lewis structure). While not wishing to be bound by theory, the actual structure can instead be some hybrid or weighted average of two or more canonical forms, known collectively as resonance forms or structures.
  • Resonance structures are not discrete chemical entities and exist only on paper. They differ from one another only in the placement or “localization” of the bonding and nonbonding electrons for a particular chemical entity. It can be possible for one resonance structure to contribute to a greater extent to the hybrid than the others.
  • the phosphate group is a negatively charged species.
  • the charge is distributed equally over the two non-linking oxygen atoms (i.e., X and Y in Formula 1 above).
  • the phosphate group can be modified by replacing one of the oxygens with a different substituent.
  • One result of this modification to RNA phosphate backbones can be increased resistance of the oligoribonucleotide to nucleolytic breakdown.
  • modified phosphate groups include phosphorothioate, phosphoroselenates, borano phosphates, borano phosphate esters, hydrogen phosphonates, phosphoroamidates, alkyl or aryl phosphonates and phosphotriesters.
  • Phosphorodithioates have both non-linking oxygens replaced by sulfur. Unlike the situation where only one of X or Y is altered, the phosphorus center in the phosphorodithioates is achiral which precludes the formation of oligoribonucleotides diastereomers. Diastereomer formation can result in a preparation in which the individual diastereomers exhibit varying resistance to nucleases.
  • RNA containing chiral phosphate groups can be lower relative to the corresponding unmodified RNA species.
  • modifications to both X and Y which eliminate the chiral center, e.g. phosphorodithioate formation may be desirable in that they cannot produce diastereomer mixtures.
  • X can be any one of S, Se, B, C, H, N, or OR (R is alkyl or aryl).
  • Y can be any one of S, Se, B, C, H, N, or OR (R is alkyl or aryl). Replacement of X and/or Y with sulfur is preferred.
  • the phosphate linker can also be modified by replacement of a linking oxygen (i.e., W or Z in Formula 1) with nitrogen (bridged phosphoroamidates), sulfur (bridged phosphorothioates) and carbon (bridged methylenephosphonates).
  • the replacement can occur at a terminal oxygen (position W (3′) or position Z (5′). Replacement of W with carbon or Z with nitrogen is preferred.
  • Candidate agents can be evaluated for suitability as described below.
  • a modified RNA can include modification of all or some of the sugar groups of the ribonucleic acid.
  • the 2′ hydroxyl group (OH) can be modified or replaced with a number of different “oxy” or “deoxy” substituents. While not being bound by theory, enhanced stability is expected since the hydroxyl can no longer be deprotonated to form a 2′ alkoxide ion.
  • the 2′ alkoxide can catalyze degradation by intramolecular nucleophilic attack on the linker phosphorus atom.
  • MOE methoxyethyl group
  • the sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose.
  • a modified RNA can include nucleotides containing e.g., arabinose, as the sugar.
  • Modified RNAs can also include “abasic” sugars, which lack a nucleobase at C-1′. These abasic sugars can also be further contain modifications at one or more of the constituent sugar atoms.
  • the 2′ modifications can be used in combination with one or more phosphate linker modifications (e.g., phosphorothioate).
  • phosphate linker modifications e.g., phosphorothioate
  • chimeric oligonucleotides are those that contain two or more different modifications.
  • the modificaton can also entail the wholesale replacement of a ribose structure with another entity at one or more sites in the iRNA agent.
  • the phosphate group can be replaced by non-phosphorus containing connectors (cf. Bracket I in Formula 1 above). While not wishing to be bound by theory, it is believed that since the charged phosphodiester group is the reaction center in nucleolytic degradation, its replacement with neutral structural mimics should impart enhanced nuclease stability. Again, while not wishing to be bound by theory, it can be desirable, in some embodiment, to introduce alterations in which the charged phosphate group is replaced by a neutral moiety.
  • moieties which can replace the phosphate group include siloxane, carbonate, carboxymethyl, carbamate, amide, thioether, ethylene oxide linker, sulfonate, sulfonamide, thioformacetal, formacetal, oxime, methyleneimino, methylenemethylimino, methylenehydrazo, methylenedimethylhydrazo and methyleneoxymethylimino.
  • Preferred replacements include the methylenecarbonylamino and methylenemethylimino groups.
  • Oligonucleotide-mimicking scaffolds can also be constructed wherein the phosphate linker and ribose sugar are replaced by nuclease resistant nucleoside or nucleotide surrogates (see Bracket II of Formula 1 above). While not wishing to be bound by theory, it is believed that the absence of a repetitively charged backbone diminishes binding to proteins that recognize polyanions (e.g. nucleases). Again, while not wishing to be bound by theory, it can be desirable in some embodiment, to introduce alterations in which the bases are tethered by a neutral surrogate backbone.
  • Examples include the mophilino, cyclobutyl, pyrrolidine and peptide nucleic acid (PNA) nucleoside surrogates.
  • a preferred surrogate is a PNA surrogate.
  • the 3′ and 5′ ends of an oligonucleotide can be modified. Such modifications can be at the 3′ end, 5′ end or both ends of the molecule. They can include modification or replacement of an entire terminal phosphate or of one or more of the atoms of the phosphate group.
  • the 3′ and 5′ ends of an oligonucleotide can be conjugated to other functional molecular entities such as labeling moieties, e.g., fluorophores (e.g., pyrene, TAMRA, fluorescein, Cy3 or Cy5 dyes) or protecting groups (based e.g., on sulfur, silicon, boron or ester).
  • labeling moieties e.g., fluorophores (e.g., pyrene, TAMRA, fluorescein, Cy3 or Cy5 dyes) or protecting groups (based e.g., on sulfur, silicon, boron or ester).
  • the functional molecular entities can be attached to the sugar through a phosphate group and/or a spacer.
  • the terminal atom of the spacer can connect to or replace the linking atom of the phosphate group or the C-3′ or C-5′ O, N, S or C group of the sugar.
  • the spacer can connect to or replace the terminal atom of a nucleotide surrogate (e.g., PNAs).
  • this array can substitute for a hairpin RNA loop in a hairpin-type RNA agent.
  • the 3′ end can be an —OH group. While not wishing to be bound by theory, it is believed that conjugation of certain moieties can improve transport, hybridization, and specificity properties. Again, while not wishing to be bound by theory, it may be desirable to introduce terminal alterations that improve nuclease resistance. Other examples of terminal modifications include dyes, intercalating agents (e.g. acridines), cross-linkers (e.g.
  • psoralene mitomycin C
  • porphyrins TPPC4, texaphyrin, Sapphyrin
  • polycyclic aromatic hydrocarbons e.g., phenazine, dihydrophenazine
  • artificial endonucleases e.g.
  • EDTA lipophilic carriers
  • lipophilic carriers e.g., cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, bomeol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, O3-(oleoyl)lithocholic acid, O3-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine) and peptide conjugates (e.g., antennapedia peptide, Tat peptide), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG] 2 , polyamino
  • biotin e.g., aspirin, vitamin E, folic acid
  • transport/absorption facilitators e.g., aspirin, vitamin E, folic acid
  • synthetic ribonucleases e.g., imidazole, bisimidazole, histamine, imidazole clusters, acridine-imidazole conjugates, Eu3+complexes of tetraazamacrocycles.
  • Terminal modifications can be added for a number of reasons, including as discussed elsewhere herein to modulate activity or to modulate resistance to degradation.
  • Terminal modifications useful for modulating activity include modification of the 5′ end with phosphate or phosphate analogs.
  • iRNA agents, especially antisense strands are 5′ phosphorylated or include a phosphoryl analog at the 5′ prime terminus.
  • 5′-phosphate modifications include those which are compatible with RISC mediated gene silencing.
  • Suitable modifications include: 5′-monophosphate ((HO)2(O)P—O-5′); 5′-diphosphate ((HO)2(O)P—O—P(HO)(O)—O-5′); 5′-triphosphate ((HO)2(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-guanosine cap (7-methylated or non-methylated) (7m-G-O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-adenosine cap (Appp), and any modified or unmodified nucleotide cap structure (N—O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-monothiophosphate (phosphorothioate; (HO)2(S)P—O-5′); 5′
  • Terminal modifications can also be useful for monitoring distribution, and in such cases the preferred groups to be added include fluorophores, e.g., fluorscein or an Alexa dye, e.g., Alexa 488. Terminal modifications can also be useful for enhancing uptake, useful modifications for this include cholesterol. Terminal modifications can also be useful for cross-linking an RNA agent to another moiety; modifications useful for this include mitomycin C.
  • Adenine, guanine, cytosine and uracil are the most common bases found in RNA. These bases can be modified or replaced to provide RNA's having improved properties.
  • nuclease resistant oligoribonucleotides can be prepared with these bases or with synthetic and natural nucleobases (e.g., inosine, thymine, xanthine, hypoxanthine, nubularine, isoguanisine, or tubercidine) and any one of the above modifications.
  • substituted or modified analogs of any of the above bases e.g., “unusual bases” and “universal bases” described herein, can be employed.
  • Examples include without limitation 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 5-halouracil, 5-(2-aminopropyl)uracil, 5-amino allyl uracil, 8-halo, amino, thiol, thioalkyl, hydroxyl and other 8-substituted adenines and guanines, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine, 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines,
  • base changes are less preferred for promoting stability, but they can be useful for other reasons, e.g., some, e.g., 2,6-diaminopurine and 2 amino purine, are fluorescent. Modified bases can reduce target specificity. This should be taken into consideration in the design of iRNA agents.
  • RNA agent e.g., a modified RNA
  • a candidate RNA agent for a selected property by exposing the agent or modified molecule and a control molecule to the appropriate conditions and evaluating for the presence of the selected property.
  • resistance to a degradent can be evaluated as follows.
  • a candidate modified RNA (and preferably a control molecule, usually the unmodified form) can be exposed to degradative conditions, e.g., exposed to a milieu, which includes a degradative agent, e.g., a nuclease.
  • a biological sample e.g., one that is similar to a milieu, which might be encountered, in therapeutic use, e.g., blood or a cellular fraction, e.g., a cell-free homogenate or disrupted cells.
  • the candidate and control could then be evaluated for resistance to degradation by any of a number of approaches.
  • the candidate and control could be labeled, preferably prior to exposure, with, e.g., a radioactive or enzymatic label, or a fluorescent label, such as Cy3 or Cy5.
  • Control and modified RNA's can be incubated with the degradative agent, and optionally a control, e.g., an inactivated, e.g., heat inactivated, degradative agent.
  • a physical parameter, e.g., size, of the modified and control molecules are then determined. They can be determined by a physical method, e.g., by polyacrylamide gel electrophoresis or a sizing column, to assess whether the molecule has maintained its original length, or assessed functionally. Alternatively, Northern blot analysis can be used to assay the length of an unlabeled modified molecule.
  • a functional assay can also be used to evaluate the candidate agent.
  • a functional assay can be applied initially or after an earlier non-functional assay, (e.g., assay for resistance to degradation) to determine if the modification alters the ability of the molecule to silence gene expression.
  • a cell e.g., a mammalian cell, such as a mouse or human cell, can be co-transfected with a plasmid expressing a fluorescent protein, e.g., GFP, and a candidate RNA agent homologous to the transcript encoding the fluorescent protein (see, e.g., WO 00/44914).
  • a modified dsRNA homologous to the GFP mRNA can be assayed for the ability to inhibit GFP expression by monitoring for a decrease in cell fluorescence, as compared to a control cell, in which the transfection did not include the candidate dsRNA, e.g., controls with no agent added and/or controls with a non-modified RNA added.
  • Efficacy of the candidate agent on gene expression can be assessed by comparing cell fluorescence in the presence of the modified and unmodified dsRNA agents.
  • a candidate dsRNA agent homologous to an endogenous mouse gene preferably a maternally expressed gene, such as c-mos
  • a phenotype of the oocyte e.g., the ability to maintain arrest in metaphase II, can be monitored as an indicator that the agent is inhibiting expression. For example, cleavage of c-mos mRNA by a dsRNA agent would cause the oocyte to exit metaphase arrest and initiate parthenogenetic development (Colledge et al.
  • RNA levels can be verified by Northern blot to assay for a decrease in the level of target mRNA, or by Western blot to assay for a decrease in the level of target protein, as compared to a negative control.
  • Controls can include cells in which with no agent is added and/or cells in which a non-modified RNA is added.
  • oligoribonucleotides and oligoribonucleosides used in accordance with this invention may be with solid phase synthesis, see for example “Oligonucleotide synthesis, a practical approach”, Ed. M. J. Gait, IRL Press, 1984; “Oligonucleotides and Analogues, A Practical Approach”, Ed. F.
  • phosphinate oligoribonucleotides The preparation of phosphinate oligoribonucleotides is described in U.S. Pat. No. 5,508,270. The preparation of alkyl phosphonate oligoribonucleotides is described in U.S. Pat. No. 4,469,863. The preparation of phosphoramidite oligoribonucleotides is described in U.S. Pat. No. 5,256,775 or U.S. Pat. No. 5,366,878. The preparation of phosphotriester oligoribonucleotides is described in U.S. Pat. No. 5,023,243. The preparation of borano phosphate oligoribonucleotide is described in U.S. Pat. Nos. 5,130,302 and 5,177,198.
  • 3′-Deoxy-3′-amino phosphoramidate oligoribonucleotides is described in U.S. Pat. No. 5,476,925.
  • 3′-Deoxy-3′-methylenephosphonate oligoribonucleotides is described in An, H, et al. J. Org. Chem. 2001, 66, 2789-2801.
  • Preparation of sulfur bridged nucleotides is described in Sproat et al. Nucleosides Nucleotides 1988, 7,651 and Crosstick et al. Tetrahedron Lett. 1989, 30, 4693.
  • MMI linked oligoribonucleosides also identified herein as MMI linked oligoribonucleosides, methylenedimethylhydrazo linked oligoribonucleosides, also identified herein as MDH linked oligoribonucleosides, and methylenecarbonylamino linked oligonucleosides, also identified herein as amide-3 linked oligoribonucleosides, and methyleneaminocarbonyl linked oligonucleosides, also identified herein as amide-4 linked oligoribonucleosides as well as mixed backbone compounds having, as for instance, alternating MMI and PO or PS linkages can be prepared as is described in U.S. Pat. Nos.
  • Formacetal and thioformacetal linked oligoribonucleosides can be prepared as is described in U.S. Pat. Nos. 5,264,562 and 5,264,564.
  • Ethylene oxide linked oligoribonucleosides can be prepared as is described in U.S. Pat. No. 5,223,618.
  • Siloxane replacements are described in Cormier, J. F. et al. Nucleic Acids Res. 1988, 16, 4583. Carbonate replacements are described in Tittensor, J.
  • Cyclobutyl sugar surrogate compounds can be prepared as is described in U.S. Pat. No. 5,359,044. Pyrrolidine sugar surrogate can be prepared as is described in U.S. Pat. No. 5,519,134. Morpholino sugar surrogates can be prepared as is described in U.S. Pat. Nos. 5,142,047 and 5,235,033, and other related patent disclosures.
  • Peptide Nucleic Acids (PNAs) are known per se and can be prepared in accordance with any of the various procedures referred to in Peptide Nucleic Acids (PNA): Synthesis, Properties and Potential Applications, Bioorganic & Medicinal Chemistry, 1996, 4, 5-23. They may also be prepared in accordance with U.S. Pat. No. 5,539,083.
  • N-2 substitued purine nucleoside amidites can be prepared as is described in U.S. Pat. No. 5,459,255.
  • 3-Deaza purine nucleoside amidites can be prepared as is described in U.S. Pat. No. 5,457,191.
  • 5,6-Substituted pyrimidine nucleoside amidites can be prepared as is described in U.S. Pat. No. 5,614,617.
  • 5-Propynyl pyrimidine nucleoside amidites can be prepared as is described in U.S. Pat. No. 5,484,908. Additional references can be disclosed in the above section on base modifications.
  • RNA agents have the following structure (see Formula 2 below):
  • R 1 , R 2 , and R 3 are each, independently, H, (i.e. abasic nucleotides), adenine, guanine, cytosine and uracil, inosine, thymine, xanthine, hypoxanthine, nubularine, tubercidine, isoguanisine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 5-halouracil, 5-(2-aminopropyl)uracil, 5-amino allyl uracil, 8-halo, amino, thio
  • H i.
  • R 4 , R 5 , and R 6 are each, independently, OR 8 , O(CH 2 CH 2 O) m CH 2 CH 2 OR 8 ; O(CH 2 ),R 9 ; O(CH 2 ) n OR 9 , H; halo; NH 2 ; NHR 8 ; N(R 8 ) 2 ; NH(CH 2 CH 2 NH) m CH 2 CH 2 NHR 9 ; NHC(O)R 8 ; cyano; mercapto, SR 8 ; alkyl-thio-alkyl; alkyl, aralkyl, cycloalkyl, aryl, heteroaryl, alkenyl, alkynyl, each of which may be optionally substituted with halo, hydroxy, oxo, nitro, haloalkyl, alkyl, alkaryl, aryl, aralkyl, alkoxy, aryloxy, amino, alkylamino, dialkylamino, heterocyclyl,
  • a 1 is:
  • a preferred A1 is chosen from 5′-monophosphate ((HO) 2 (O)P—O-5′), 5′-diphosphate ((HO) 2 (O)P—O—P(HO)(O)—O-5′), 5′-triphosphate ((HO) 2 (O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′), 5′-guanosine cap (7-methylated or non-methylated) (7m-G-O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′), 5′-adenosine cap (Appp), and any modified or unmodified nucleotide cap structure (N—O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′), 5′-monothiophosphate (phosphorothioate; (N—O-5′-(HO)(O
  • a 2 is:
  • a 3 is:
  • a 4 is:
  • W 1 is OH, (CH 2 ) n R 10 , (CH 2 ) n NHR 10 , (CH 2 ) n OR 10 , (CH 2 ) n SR 10 ; O(CH 2 ) n R 10 ; O(CH 2 ) n OR 10 , O(CH 2 ) n NR 10 , O(CH 2 ) n SR 10 ; O(CH 2 ) n SS(CH 2 ) n OR 10 , O(CH 2 ) n C(O)OR 10 , NH(CH 2 ) n R 10 ; NH(CH 2 ) n NR 10 ; NH(CH 2 ) n OR 10 , NH(CH 2 ) n SR 10 ; S(CH 2 ) n R 10 , S(CH 2 ) n NR 10 , S(CH 2 ) n OR 10 , S(CH 2 ) n SR 10 O(CH 2 CH 2 O) n CH 2 CH
  • X 1 , X 2 , X 3 , and X 4 are each, independently, O or S.
  • Y 1 , Y 2 , Y 3 , and Y 4 are each, independently, OH, O—, OR 8 , S, Se, BH 3 ⁇ , H, NHR 9 , N(R 9 ) 2 alkyl, cycloalkyl, aralkyl, aryl, or heteroaryl, each of which may be optionally substituted.
  • Z 1 , Z 2 , and Z 3 are each independently O, CH 2 , NH, or S.
  • Z 4 is OH, (CH 2 ) n R 10 , (CH 2 ) n NHR 10 , (CH 2 ) n OR 10 , (CH 2 ) n SR 10 ; O(CH 2 ) n R 10 ; O(CH 2 ) n OR 10 , O(CH 2 ) n NR 10 , O(CH 2 ) n SR 10 , O(CH 2 ) n SS(CH 2 ) n OR 10 , O(CH 2 ) n C(O)OR 10 ; NH(CH 2 ) n R 10 ; NH(CH 2 ) n NR 10 ; NH(CH 2 ) n OR 10 , NH(CH 2 ) n SR 10 ; S(CH 2 ) n R 10 , S(CH 2 ) n NR 10 , S(CH 2 ) n
  • x is 5-100, chosen to comply with a length for an RNA agent described herein.
  • R 7 is H; or is together combined with R 4 , R 5 , or R 6 to form an [—O—CH 2 —] covalently bound bridge between the sugar 2′ and 4′ carbons.
  • R 8 is alkyl, cycloalkyl, aryl, aralkyl, heterocyclyl, heteroaryl, amino acid, or sugar;
  • R 9 is NH 2 , alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid; and
  • R 10 is H; fluorophore (pyrene, TAMRA, fluorescein, Cy3 or Cy5 dyes); sulfur, silicon, boron or ester protecting group; intercalating agents (e.g. acridines), cross-linkers (e.g.
  • psoralene mitomycin C
  • porphyrins TPPC4, texaphyrin, Sapphyrin
  • polycyclic aromatic hydrocarbons e.g., phenazine, dihydrophenazine
  • artificial endonucleases e.g.
  • lipohilic carriers cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, bomeol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, O3-(oleoyl)lithocholic acid, 03-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine) and peptide conjugates (e.g., antennapedia peptide, Tat peptide), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG] 2 , polyamino; alkyl, cyclo
  • biotin e.g., aspirin, vitamin E, folic acid
  • synthetic ribonucleases e.g., imidazole, bisimidazole, histamine, imidazole clusters, acridine-imidazole conjugates, Eu3+complexes of tetraazamacrocycles
  • RNA agent e.g., m is 0-1,000,000, and n is 0-20.
  • Q is a spacer selected from the group consisting of abasic sugar, amide, carboxy, oxyamine, oxyimine, thioether, disulfide, thiourea, sulfonamide, or morpholino, biotin or fluorescein reagents.
  • RNA agents in which the entire phosphate group has been replaced have the following structure (see Formula 3 below):
  • a 10 -A 40 is L-G-L; A 10 and/or A 40 may be absent, in which L is a linker, wherein one or both L may be present or absent and is selected from the group consisting of CH 2 (CH 2 ) g ; N(CH 2 ) g ; O(CH 2 ) g ; S(CH 2 ) g .
  • G is a functional group selected from the group consisting of siloxane, carbonate, carboxymethyl, carbamate, amide, thioether, ethylene oxide linker, sulfonate, sulfonamide, thioformacetal, formacetal, oxime, methyleneimino, methylenemethylimino, methylenehydrazo, methylenedimethylhydrazo and methyleneoxymethylimino.
  • R 10 , R 20 , and R 30 are each, independently, H, (i.e. abasic nucleotides), adenine, guanine, cytosine and uracil, inosine, thymine, xanthine, hypoxanthine, nubularine, tubercidine, isoguanisine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 5-halouracil, 5-(2-aminopropyl)uracil, 5-amino allyl uracil, 8-halo, amino, thiol, thioal
  • R 40 , R 50 , and R 60 are each, independently, OR 8 , O(CH 2 CH 2 O) m CH 2 CH 2 OR 8 ; O(CH 2 ) n R 9 ; O(CH 2 ) n OR 9 , H; halo; NH 2 ; NHR 8 ; N(R 8 ) 2 ; NH(CH 2 CH 2 NH) m CH 2 CH 2 R 9 ; NHC(O)R 8 ; cyano; mercapto, SR 7 ; alkyl-thio-alkyl; alkyl, aralkyl, cycloalkyl, aryl, heteroaryl, alkenyl, alkynyl, each of which may be optionally substituted with halo, hydroxy, oxo, nitro, haloalkyl, alkyl, alkaryl, aryl, aralkyl, alkoxy, aryloxy, amino, alkylamino, dialkylamino, heterocyclyl
  • x is 5-100 or chosen to comply with a length for an RNA agent described herein.
  • R 70 is H; or is together combined with R 40 , R 50 , or R 60 to form an [—O—CH 2 —] covalently bound bridge between the sugar 2′ and 4′ carbons.
  • R 8 is alkyl, cycloalkyl, aryl, aralkyl, heterocyclyl, heteroaryl, amino acid, or sugar; and R 9 is NH 2 , alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid.
  • m is 0-1,000,000, n is 0-20, and g is 0-2.
  • Preferred nucleoside surrogates have the following structure (see Formula 4 below): SLR 100 -(M-SLR 200 ) x -M-sLR 300 FORMULA 4
  • S is a nucleoside surrogate selected from the group consisting of mophilino, cyclobutyl, pyrrolidine and peptide nucleic acid.
  • L is a linker and is selected from the group consisting of CH 2 (CH 2 ) g ; N(CH 2 ) g ; O(CH 2 ) g ; S(CH 2 ) g ; —C(O)(CH 2 ) n — or may be absent.
  • M is an amide bond; sulfonamide; sulfinate; phosphate group; modified phosphate group as described herein; or may be absent.
  • R 100 , R 200 , and R 300 are each, independently, H (i.e., abasic nucleotides), adenine, guanine, cytosine and uracil, inosine, thymine, xanthine, hypoxanthine, nubularine, tubercidine, isoguanisine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 5-halouracil, 5-(2-aminopropyl)uracil, 5-amino allyl uracil, 8-halo, amino, thiol, thioal
  • x is 5-100, or chosen to comply with a length for an RNA agent described herein; and g is 0-2.
  • RNA e.g., an iRNA agent
  • NAM nuclease resistant monomer
  • An iRNA agent can include monomers which have been modifed so as to inhibit degradation, e.g., by nucleases, e.g., endonucleases or exonucleases, found in the body of a subject. These monomers are referred to herein as NRMs, or nuclease resistance promoting monomers or modifications.
  • these modifications will modulate other properties of the iRNA agent as well, e.g., the ability to interact with a protein, e.g., a transport protein, e.g., serum albumin, or a member of the RISC(RNA-induced Silencing Complex), or the ability of the first and second sequences to form a duplex with one another or to form a duplex with another sequence, e.g., a target molecule.
  • a protein e.g., a transport protein, e.g., serum albumin, or a member of the RISC(RNA-induced Silencing Complex
  • modifications of the sugar, base, and/or phosphate backbone in an iRNA agent can enhance endonuclease and exonuclease resistance, and can enhance interactions with transporter proteins and one or more of the functional components of the RISC complex.
  • Preferred modifications are those that increase exonuclease and endonuclease resistance and thus prolong the half-life of the iRNA agent prior to interaction with the RISC complex, but at the same time do not render the iRNA agent resistant to endonuclease activity in the RISC complex.
  • An iRNA agent may include a duplex comprising a hybridized sense and antisense strand, in which the antisense strand and/or the sense strand may include one or more of the modifications described herein.
  • the anti sense strand may include modifications at the 3′ end and/or the 5′ end and/or at one or more positions that occur 1-6 (e.g., 1-5,1-4, 1-3,1-2) nucleotides from either end of the strand.
  • the sense strand may include modifications at the 3′ end and/or the 5′ end and/or at any one of the intervening positions between the two ends of the strand.
  • the iRNA agent may also include a duplex comprising two hybridized antisense strands.
  • the first and/or the second antisense strand may include one or more of the modifications described herein.
  • one and/or both antisense strands may include modifications at the 3′ end and/or the 5′ end and/or at one or more positions that occur 1-6 (e.g., 1-5,1-4, 1-3,1-2) nucleotides from either end of the strand. Particular configurations are discussed below.
  • Modifications that can be useful for producing iRNA agents that meet the preferred nuclease resistance criteria delineated above can include one or more of the following chemical and/or stereochemical modifications of the sugar, base, and/or phosphate backbone:
  • NRM modifications can be introduced into an iRNA agent or into a sequence of an iRNA agent.
  • An NRM modification can be used more than once in a sequence or in an iRNA agent. As some NRM's interfere with hybridization the total number incorporated, should be such that acceptable levels of iRNA agent duplex formation are maintained.
  • NRM modifications are introduced into the terminal the cleavage site or in the cleavage region of a sequence (a sense strand or sequence) which does not target a desired sequence or gene in the subject. This can reduce off-target silencing.
  • a modification can include the alteration, e.g., replacement, of one or both of the non-linking (X and Y) phosphate oxygens and/or of one or more of the linking (W and Z) phosphate oxygens.
  • Formula X depicts a phosphate moiety linking two sugar/sugar surrogate-base moieties, SB 1 and SB 2 .
  • one of the non-linking phosphate oxygens in the phosphate backbone moiety can be replaced by any one of the following: S, Se, BR 3 (R is hydrogen, alkyl, aryl, etc.), C (i.e., an alkyl group, an aryl group, etc.), H, NR 2 (R is hydrogen, alkyl, aryl, etc.), or OR (R is alkyl or aryl).
  • S, Se R is hydrogen, alkyl, aryl, etc.
  • C i.e., an alkyl group, an aryl group, etc.
  • H NR 2
  • OR R is alkyl or aryl
  • the phosphorus atom in an unmodified phosphate group is achiral.
  • the stereogenic phosphorus atom can possess either the “R” configuration (herein R p ) or the “S” configuration (herein S p ).
  • R p the “R” configuration
  • S p the “S” configuration
  • iRNA agents having phosphate groups in which a phosphate non-linking oxygen has been replaced by another atom or group of atoms, may contain a population of stereogenic phosphorus atoms in which at least about 50% of these atoms (e.g., at least about 60% of these atoms, at least about 70% of these atoms, at least about 80% of these atoms, at least about 90% of these atoms, at least about 95% of these atoms, at least about 98% of these atoms, at least about 99% of these atoms) have the S p configuration.
  • these atoms e.g., at least about 60% of these atoms, at least about 70% of these atoms, at least about 80% of these atoms, at least about 90% of these atoms, at least about 95% of these atoms, at least about 98% of these atoms, at least about 99% of these atoms
  • iRNA agents having phosphate groups in which a phosphate non-linking oxygen has been replaced by another atom or group of atoms may contain a population of stereogenic phosphorus atoms in which at least about 50% of these atoms (e.g., at least about 60% of these atoms, at least about 70% of these atoms, at least about 80% of these atoms, at least about 90% of these atoms, at least about 95% of these atoms, at least about 98% of these atoms, at least about 99% of these atoms) have the R p configuration.
  • the population of stereogenic phosphorus atoms may have the S p configuration and may be substantially free of stereogenic phosphorus atoms having the R p configuration. In still other embodiments, the population of stereogenic phosphorus atoms may have the R p configuration and may be substantially free of stereogenic phosphorus atoms having the S p configuration.
  • the phrase “substantially free of stereogenic phosphorus atoms having the R p configuration” means that moieties containing stereogenic phosphorus atoms having the R p configuration cannot be detected by conventional methods known in the art (chiral HPLC, 1 H NMR analysis using chiral shift reagents, etc.).
  • the phrase “substantially free of stereogenic phosphorus atoms having the S p configuration” means that moieties containing stereogenic phosphorus atoms having the S p configuration cannot be detected by conventional methods known in the art (chiral HPLC, 1 H NMR analysis using chiral shift reagents, etc.).
  • modified iRNA agents contain a phosphorothioate group, i.e., a phosphate groups in which a phosphate non-linking oxygen has been replaced by a sulfur atom.
  • the population of phosphorothioate stereogenic phosphorus atoms may have the S p configuration and be substantially free of stereogenic phosphorus atoms having the R p configuration.
  • Phosphorothioates may be incorporated into iRNA agents using dimers e.g., formulas X-1 and X-2.
  • the former can be used to introduce phosphorothioate at the 3′ end of a strand, while the latter can be used to introduce this modification at the 5′ end or at a position that occurs e.g., 1, 2, 3, 4, 5, or 6 nucleotides from either end of the strand.
  • Y can be 2-cyanoethoxy
  • W and Z can be 0,
  • R 2 can be, e.g., a substituent that can impart the C-3 endo configuration to the sugar (e.g., OH, F, OCH 3 ), DMT is dimethoxytrityl, and “BASE” can be a natural, unusual, or a universal base.
  • X-1 and X-2 can be prepared using chiral reagents or directing groups that can result in phosphorothioate-containing dimers having a population of stereogenic phosphorus atoms having essentially only the R p configuration (i.e., being substantially free of the S p configuration) or only the S p configuration (i.e., being substantially free of the R p configuration).
  • dimers can be prepared having a population of stereogenic phosphorus atoms in which about 50% of the atoms have the R p configuration and about 50% of the atoms have the S p configuration.
  • Dimers having stereogenic phosphorus atoms with the R p configuration can be identified and separated from dimers having stereogenic phosphorus atoms with the S p configuration using e.g., enzymatic degradation and/or conventional chromatography techniques.
  • Modifications can also include attachment of one or more cationic groups to the sugar, base, and/or the phosphorus atom of a phosphate or modified phosphate backbone moiety.
  • a cationic group can be attached to any atom capable of substitution on a natural, unusual or universal base.
  • a preferred position is one that does not interfere with hybridization, i.e., does not interfere with the hydrogen bonding interactions needed for base pairing.
  • a cationic group can be attached e.g., through the C2′ position of a sugar or analogous position in a cyclic or acyclic sugar surrogate.
  • NH 2 alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid
  • Modifications can also include the incorporation of nonphosphate linkages at the 5′ and/or 3′ end of a strand.
  • nonphosphate linkages which can replace the phosphate group include methyl phosphonate, hydroxylamino, siloxane, carbonate, carboxymethyl, carbamate, amide, thioether, ethylene oxide linker, sulfonate, sulfonamide, thioformacetal, formacetal, oxime, methyleneimino, methylenemethylimino, methylenehydrazo, methylenedimethylhydrazo and methyleneoxymethylimino.
  • Preferred replacements include the methyl phosphonate and hydroxylamino groups.
  • modifications can include replacement of one of the bridging or linking phosphate oxygens in the phosphate backbone moiety (W and Z). Unlike the situation where only one of X or Y is altered, the phosphorus center in the phosphorodithioates is achiral which precludes the formation of iRNA agents containing a stereogenic phosphorus atom.
  • Modifications can also include linking two sugars via a phosphate or modified phosphate group through the 2′ position of a first sugar and the 5′ position of a second sugar. Also contemplated are inverted linkages in which both a first and second sugar are eached linked through the respective 3′ positions.
  • Modified RNA's can also include “abasic” sugars, which lack a nucleobase at C-1′. The sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose.
  • a modified iRNA agent can include nucleotides containing e.g., arabinose, as the sugar.
  • the natural, unusual, or universal base may have the ⁇ -configuration.
  • Modifcations can also include L-RNA.
  • the prodrug groups may be decomposed via reaction first with carboxy esterases. The remaining ethyl thiolate group via intramolecular S N 2 displacement can depart as episulfide to afford the underivatized phosphate group.
  • Modification can also include the addition of conjugating groups described elseqhere herein, which are prefereably attached to an iRNA agent through any amino group available for conjugation.
  • Nuclease resistant modifications include some which can be placed only at the terminus and others which can go at any position. Generally the modifications that can inhibit hybridization so it is preferably to use them only in terminal regions, and preferrable to not use them at the cleavage site or in the cleavage region of an sequence which targets a subject sequence or gene. The can be used anywhere in a sense sequence, provided that sufficient hybridization between the two sequences of the iRNA agent is maintained. In some embodiments it is desirabable to put the NRM at the cleavage site or in the cleavage region of a sequence which does not target a subject sequence or gene, as it can minimize off-target silencing.
  • an iRNA agent described herein can have an overhang which does not form a duplex structure with the other sequence of the iRNA agent—it is an overhang, but it does hybridize, either with itself, or with another nucleic acid, other than the other sequence of the iRNA agent.
  • nuclease-resistance promoting modifications will be distributed differently depending on whether the sequence will target a sequence in the subject (often referred to as an anti-sense sequence) or will not target a sequence in the subject (often referred to as a sense sequence). If a sequence is to target a sequence in the subject, modifications which interfer with or inhibit endonuclease cleavage should not be inserted in the region which is subject to RISC mediated cleavage, e.g., the cleavage site or the cleavage region (As described in Elbashir et al., 2001, Genes and Dev.
  • cleavage of the target occurs about in the middle of a 20 or 21 nt guide RNA, or about 10 or 11 nucleotides upstream of the first nucleotide which is complementary to the guide sequence.
  • cleavage site refers to the nucleotide on either side of the cleavage site, on the target or on the iRNA agent strand which hybridizes to it.
  • Cleavage region means an nucleotide with 1, 2, or 3 nucletides of the cleave site, in either direction.
  • Such modifications can be introduced into the terminal regions, e.g., at the terminal position or with 2, 3, 4, or 5 positions of the terminus, of a sequence which targets or a sequence which does not target a sequence in the subject.
  • An iRNA agent can have a first and a second strand chosen from the following:
  • An iRNA agent can also target two sequences and can have a first and second strand chosen from:
  • RNA e.g., an iRNA agent
  • a ribose mimic such as those described herein and those described in copending co-owned U.S. Provisional Application Ser. No. 60/454,962, filed on Mar. 13, 2003, and International Application No. PCT/U.S.04/07070, both of which are hereby incorporated by reference.
  • an aspect of the invention features an iRNA agent that includes a secondary hydroxyl group, which can increase efficacy and/or confer nuclease resistance to the agent.
  • Nucleases e.g., cellular nucleases, can hydrolyze nucleic acid phosphodiester bonds, resulting in partial or complete degradation of the nucleic acid.
  • the secondary hydroxy group confers nuclease resistance to an iRNA agent by rendering the iRNA agent less prone to nuclease degradation relative to an iRNA which lacks the modification.
  • a secondary hydroxyl group on the iRNA agent can act as a structural mimic of a 3′ ribose hydroxyl group, thereby causing it to be less susceptible to degradation.
  • the secondary hydroxyl group refers to an “OH” radical that is attached to a carbon atom substituted by two other carbons and a hydrogen.
  • the secondary hydroxyl group that confers nuclease resistance as described above can be part of any acyclic carbon-containing group.
  • the hydroxyl may also be part of any cyclic carbon-containing group, and preferably one or more of the following conditions is met (1) there is no ribose moiety between the hydroxyl group and the terminal phosphate group or (2) the hydroxyl group is not on a sugar moiety which is coupled to a base.
  • the hydroxyl group is located at least two bonds (e.g., at least three bonds away, at least four bonds away, at least five bonds away, at least six bonds away, at least seven bonds away, at least eight bonds away, at least nine bonds away, at least ten bonds away, etc.) from the terminal phosphate group phosphorus of the iRNA agent. In preferred embodiments, there are five intervening bonds between the terminal phosphate group phosphorus and the secondary hydroxyl group.
  • Preferred iRNA agent delivery modules with five intervening bonds between the terminal phosphate group phosphorus and the secondary hydroxyl group have the following structure (see formula Y below):
  • A is an iRNA agent, including any iRNA agent described herein.
  • the iRNA agent may be connected directly or indirectly (e.g., through a spacer or linker) to “W” of the phosphate group.
  • the iRNA agents can have a terminal phosphate group that is unmodified (e.g., W, X, Y, and Z are O) or modified.
  • W and Z can be independently NH, O, or S; and
  • X and Y can be independently S, Se, BH 3 ⁇ , C 1 -C 6 alkyl, C 6 -C 10 aryl, H, O, O ⁇ , alkoxy or amino (including alkylamino, arylamino, etc.).
  • W, X and Z are 0 and Y is S.
  • R 1 and R 3 are each, independently, hydrogen; or C 1 -C 100 alkyl, optionally substituted with hydroxyl, amino, halo, phosphate or sulfate and/or may be optionally inserted with N, O, S, alkenyl or alkynyl.
  • R 2 is hydrogen; C 1 -C 100 alkyl, optionally substituted with hydroxyl, amino, halo, phosphate or sulfate and/or may be optionally inserted with N, O, S, alkenyl or alkynyl; or, when n is 1, R 2 may be taken together with with R 4 or R 6 to form a ring of 5-12 atoms.
  • R 4 is hydrogen; C 1 -C 100 alkyl, optionally substituted with hydroxyl, amino, halo, phosphate or sulfate and/or may be optionally inserted with N, O, S, alkenyl or alkynyl; or, when n is 1, R 4 may be taken together with with R 2 or R 5 to form a ring of 5-12 atoms.
  • R 5 is hydrogen, C 1 -C 100 alkyl optionally substituted with hydroxyl, amino, halo, phosphate or sulfate and/or may be optionally inserted with N, O, S, alkenyl or alkynyl; or, when n is 1, R 5 may be taken together with with R 4 to form a ring of 5-12 atoms.
  • R 6 is hydrogen, C 1 -C 100 alkyl, optionally substituted with hydroxyl, amino, halo, phosphate or sulfate and/or may be optionally inserted with N, O, S, alkenyl or alkynyl, or, when n is 1, R 6 may be taken together with with R 2 to form a ring of 6-10 atoms;
  • R 7 is hydrogen, C 1 -C 100 alkyl, or C(O)(CH 2 ) q C(O)NHR 9 ;
  • T is hydrogen or a functional group;
  • n and q are each independently 1-100;
  • R 8 is C 1 -C 10 alkyl or C 6 -C 10 aryl; and
  • R 9 is hydrogen, C1-C10 alkyl, C6-C10 aryl or a solid support agent.
  • Preferred embodiments may include one of more of the following subsets of iRNA agent delivery modules.
  • A can be connected directly or indirectly through a terminal 3′ or 5′ ribose sugar carbon of the RNA agent.
  • RNAi agent delivery modules In another subset of RNAi agent delivery modules, X, W, and Z are 0 and Y is S.
  • n is 1, and R 2 and R 6 are taken together to form a ring containing six atoms and R 4 and R 5 are taken together to form a ring containing six atoms.
  • the ring system is a trans-decalin.
  • the RNAi agent delivery module of this subset can include a compound of Formula (Y-1):
  • the functional group can be, for example, a targeting group (e.g., a steroid or a carbohydrate), a reporter group (e.g., a fluorophore), or a label (an isotopically labelled moiety).
  • a targeting group e.g., a steroid or a carbohydrate
  • a reporter group e.g., a fluorophore
  • a label an isotopically labelled moiety
  • the targeting group can further include protein binding agents, endothelial cell targeting groups (e.g., RGD peptides and mimetics), cancer cell targeting groups (e.g., folate Vitamin B12, Biotin), bone cell targeting groups (e.g., bisphosphonates, polyglutamates, polyaspartates), multivalent mannose (for e.g., macrophage testing), lactose, galactose, N-acetyl-galactosamine, monoclonal antibodies, glycoproteins, lectins, melanotropin, or thyrotropin.
  • endothelial cell targeting groups e.g., RGD peptides and mimetics
  • cancer cell targeting groups e.g., folate Vitamin B12, Biotin
  • bone cell targeting groups e.g., bisphosphonates, polyglutamates, polyaspartates
  • multivalent mannose for e.g., macrophage testing
  • lactose galactose
  • the invention relates to a pharmaceutical composition containing a modified iRNA agent, as described in the preceding sections, and a pharmaceutically acceptable carrier, as described below.
  • a pharmaceutical composition including the modified iRNA agent is useful for treating a disease caused by expression of a target gene.
  • the iRNA agent of the invention is formulated as described below.
  • the pharmaceutical composition is administered in a dosage sufficient to inhibit expression of the target gene.
  • compositions of the present invention are administered in dosages sufficient to inhibit the expression or activity of the target gene.
  • Compositions containing the iRNA agent of the invention can be administered at surprisingly low dosages.
  • a maximum dosage of 5 mg iRNA agent per kilogram body weight per day may be sufficient to inhibit or completely suppress the expression or activity of the target gene.
  • a suitable dose of modified iRNA agent will be in the range of 0.001 to 500 milligrams per kilogram body weight of the recipient per day (e.g., about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 100 milligrams per kilogram, about 1 milligrams per kilogram to about 75 milligrams per kilogram, about 10 micrograms per kilogram to about 50 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram).
  • the pharmaceutical composition may be administered once per day, or the iRNA agent may be administered as two, three, four, five, six or more sub-doses at appropriate intervals throughout the day.
  • the iRNA agent contained in each sub-dose must be correspondingly smaller in order to achieve the total daily dosage.
  • the dosage unit can also be compounded for delivery over several days, e.g., using a conventional sustained release formulation which provides sustained release of the iRNA agent over a several day period. Sustained release formulations are well known in the art. In this embodiment, the dosage unit contains a corresponding multiple of the daily dose.
  • treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or a series of treatments.
  • Estimates of effective dosages and in vivo half-lives for the individual iRNA agent encompassed by the invention can be made using conventional methodologies or on the basis of in vivo testing using an appropriate animal model, as described elsewhere herein.
  • mouse repositories can be found at The Jackson Laboratory, Charles River Laboratories, Taconic, Harlan, Mutant Mouse Regional Resource Centers (MMRRC) National Network and at the European Mouse Mutant Archive.
  • MMRRC Mutant Mouse Regional Resource Centers
  • Such models may be used for in vivo testing of iRNA agent, as well as for determining a therapeutically effective dose.
  • compositions encompassed by the invention may be administered by any means known in the art including, but not limited to oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), ocular, rectal, vaginal and topical (including buccal and sublingual) administration.
  • oral or parenteral routes including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), ocular, rectal, vaginal and topical (including buccal and sublingual) administration.
  • the pharmaceutical compositions are administered by intravenous or intraparenteral infusion or injection.
  • the pharmaceutical compositions can also be administered intraparenchymally, intrathecally, and/or by stereotactic injection.
  • the iRNA agent useful in the invention will generally be provided in the form of tablets or capsules, as a powder or granules, or as an aqueous solution or suspension.
  • Tablets for oral use may include the active ingredients mixed with pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives.
  • suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents.
  • Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc.
  • the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
  • Capsules for oral use include hard gelatin capsules in which the active ingredient is mixed with a solid diluent, and soft gelatin capsules wherein the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin or olive oil.
  • compositions of the invention will generally be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and isotonicity.
  • Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride.
  • the carrier consists exclusively of an aqueous buffer.
  • “exclusively” means no auxiliary agents or encapsulating substances are present which might affect or mediate uptake of iRNA agent in the cells that harbor the target gene or virus.
  • Such substances include, for example, micellar structures, such as liposomes or capsids, as described below.
  • Aqueous suspensions according to the invention may include suspending agents such as cellulose derivatives, sodium alginate, polyvinyl-pyrrolidone and gum tragacanth, and a wetting agent such as lecithin.
  • Suitable preservatives for aqueous suspensions include ethyl and n-propyl p-hydroxybenzoate.
  • the pharmaceutical compositions can also include encapsulated formulations to protect the iRNA agent against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • encapsulated formulations to protect the iRNA agent against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • the materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.
  • Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers.
  • Toxicity and therapeutic efficacy of iRNA agent can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
  • iRNA agents that exhibit high therapeutic indices are preferred.
  • the data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosages of compositions of the invention are preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range of the iRNA agent or, when appropriate, of the polypeptide product of a target sequence (e.g., achieving a decreased concentration of the polypeptide) that includes the IC50 (i.e., the concentration of the test iRNA agent which achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • a target sequence e.g., achieving a decreased concentration of the polypeptide
  • the IC50 i.e., the concentration of the test iRNA agent which achieves a half-maximal inhibition of symptoms
  • levels in plasma may be measured, for example, by high performance liquid chromatography.
  • iRNA agents relating to the invention can be administered in combination with other known agents effective in treating viral infections and diseases.
  • the administering physician can adjust the amount and timing of iRNA agent administration on the basis of results observed using standard measures of efficacy known in the art or described herein.
  • the iRNA agent useful in the invention will generally be provided in the form of tablets or capsules, as a powder or granules, or as an aqueous solution or suspension.
  • the invention relates to a method for treating a subject having a disease or at risk of developing a disease caused by the expression of a target gene.
  • iRNA agents can act as novel therapeutic agents for controlling one or more of cellular proliferative and/or differentiative disorders, disorders associated with bone metabolism, immune disorders, hematopoietic disorders, cardiovascular disorders, liver disorders, viral diseases, or metabolic disorders.
  • the method includes administering a pharmaceutical composition of the invention to the patient (e.g., a human), such that expression of the target gene is silenced. Because of their high efficiency and specificity, the iRNA agent of the present invention specifically target mRNA of target genes of diseased cells and tissues, as described below, and at surprisingly low dosages.
  • the pharmaceutical compositions are formulated as described in the preceding section, which is hereby incorporated by reference herein.
  • genes which can be targeted for treatment include, without limitation, an oncogene (Hanahan, D. and R. A. Weinberg, Cell (2000) 100:57; and Yokota, J., Carcinogenesis (2000) 21(3):497-503); a cytokine gene (Rubinstein, M., et al., Cytokine Growth Factor Rev . (1998) 9(2):175-81); a idiotype (Id) protein gene (Benezra, R., et al., Oncogene (2001) 20(58):8334-41; Norton, J. D., J. Cell Sci . (2000) 113(22):3897-905); a prion gene (Prusiner, S.
  • the target gene may be one which is required for initiation or maintenance of the disease, or which has been identified as being associated with a higher risk of contracting the disease.
  • the iRNA agent can be brought into contact with the cells or tissue exhibiting the disease.
  • iRNA agent substantially identical to all or part of a mutated gene associated with cancer, or one expressed at high levels in tumor cells may be brought into contact with or introduced into a cancerous cell or tumor gene.
  • Examples of cellular proliferative and/or differentiative disorders include cancer, e.g., a carcinoma, sarcoma, metastatic disorder or hematopoietic neoplastic disorder, such as a leukemia.
  • a metastatic tumor can arise from a multitude of primary tumor types, including but not limited to those of prostate, colon, lung, breast and liver origin.
  • the terms “cancer,” “hyperproliferative,” and “neoplastic” refer to cells having the capacity for autonomous growth, i.e., an abnormal state or condition characterized by rapidly proliferating cell growth.
  • Proliferative disorders also include hematopoietic neoplastic disorders, including diseases involving hyperplastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof.
  • compositions of the present invention can also be used to treat a variety of immune disorders, in particular those associated with overexpression or aberrant expression of a gene or expression of a mutant gene.
  • hematopoietic disorders or diseases include, without limitation, autoimmune diseases (including, for example, diabetes mellitus, arthritis (including rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, psoriatic arthritis), multiple sclerosis, encephalomyelitis, myasthenia gravis, systemic lupus erythematosis, automimmune thyroiditis, dermatitis (including atopic dermatitis and eczematous dermatitis), psoriasis, Sjogren's Syndrome, Crohn's disease, aphthous ulcer, ulceris, conjunctivitis, keratoconjunctivitis, ulcerative colitis, asthma, allergic asthma, cutaneous lupus erythematosus,
  • the invention relates to methods for treating viral diseases, including but not limited to hepatitis C, hepatitis B, herpes simplex virus (HSV), HIV-AIDS, poliovirus, and smallpox virus.
  • iRNA agent of the invention are prepared as described herein to target expressed sequences of a virus, thus ameliorating viral activity and replication.
  • the iRNA agents can be used in the treatment and/or diagnosis of viral infected tissue, both animal and plant. Also, such iRNA agent can be used in the treatment of virus-associated carcinoma, such as hepatocellular cancer.
  • the iRNA agent of the present invention are useful for treating a subject having an infection or a disease associated with the replication or activity of a (+) strand RNA virus having a 3′-UTR, such as HCV.
  • the iRNA agent can act as novel therapeutic agents for inhibiting replication of the virus.
  • the method includes administering a pharmaceutical composition of the invention to the patient (e.g., a human), such that viral replication is inhibited.
  • (+) strand RNA viruses which can be targeted for inhibition include, without limitation, picornaviruses, caliciviruses, nodaviruses, coronaviruses, arteriviruses, flaviviruses, and togaviruses.
  • picomaviruses examples include enterovirus (poliovirus 1), rhinovirus (human rhinovirus 1A), hepatovirus (hepatitis A virus), cardiovirus (encephalomyocarditis virus), aphthovirus (foot-and-mouth disease virus O), and parechovirus (human echovirus 22).
  • caliciviruses include vesiculovirus (swine vesicular exanthema virus), lagovirus (rabbit hemorrhagic disease virus), “Norwalk-like viruses” (Norwalk virus), “Sapporo-like viruses” (Sapporo virus), and “hepatitis E-like viruses” (hepatitis E virus).
  • Betanodavirus (striped jack nervous necrosis virus) is the representative nodavirus.
  • Coronaviruses include coronavirus (avian infections bronchitis virus) and torovirus (Beme virus).
  • Arterivirus (equine arteritis virus) is the representative arteriviridus.
  • Togavirises include alphavirus (Sindbis virus) and rubivirus (Rubella virus).
  • the flaviviruses include flavivirus (Yellow fever virus), pestivirus (bovine diarrhea virus), and hepacivirus (hepatitis C virus).
  • the virus is hepacivirus, the hepatitis C virus.
  • the present invention encompasses the compositions and methods for treating infections and diseases caused by any (+) strand RNA virus having a 3′-UTR, regardless of the host.
  • the invention encompasses the treatment of plant diseases caused by sequiviruses, comoviruses, potyviruses, sobemovirus, luteoviruses, tombusviruses, tobavirus, tobravirus, bromoviruses, and closteroviruses.
  • compositions encompassed by the invention may be administered by any means known in the art including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), ocular, rectal, vaginal, and topical (including buccal and sublingual) administration.
  • oral or parenteral routes including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), ocular, rectal, vaginal, and topical (including buccal and sublingual) administration.
  • the pharmaceutical compositions are administered by intravenous or intraparenteral infusion or injection.
  • the pharmaceutical compositions can also be administered intraparenchymally, intrathecally, and/or by stereotactic injection.
  • the invention relates to a method for inhibiting the expression of a target gene in a cell or organism.
  • the method includes administering the inventive iRNA agent or a pharmaceutical composition containing the iRNA agent to a cell or an organism, such as a mammal, such that expression of the target gene is silenced.
  • the iRNA agent of the present invention effectively inhibit expression or activity of target genes at surprisingly low dosages.
  • Compositions and methods for inhibiting the expression of a target gene using iRNA agent can be performed as described in the preceding sections, particularly Sections 4 and 5.
  • a pharmaceutical composition containing the iRNA agent may be administered by any means known in the art including, but not limited to oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), ocular, rectal, vaginal, and topical (including buccal and sublingual) administration.
  • the pharmaceutical compositions are administered by intravenous or intraparenteral infusion or injection.
  • the pharmaceutical compositions can also be administered intraparenchymally, intrathecally, and/or by stereotactic injection.
  • the methods for inhibiting the expression of a target gene can be applied to any gene one wishes to silence, thereby specifically inhibiting its expression, provided the cell or organism in which the target gene is expressed includes the cellular machinery which effects RNA interference.
  • genes which can be targeted for silencing include, without limitation, developmental genes including but not limited to adhesion molecules, cyclin kinase inhibitors, Wnt family members, Pax family members, Winged helix family members, Hox family members, cytokines/lymphokines and their receptors, growth/differentiation factors and their receptors, and neurotransmitters and their receptors; (2) oncogenes including but not limited to ABLI, BCL1, BCL2, BCL6, CBFA2, CBL, CSFIR, ERBA, ERBB, EBRB2, ETS 1, ETS 1, ETV6, FGR, FOS, FYN, HCR, HRAS, JUN, KRAS, LCK, LYN, MDM2, MLL, MYB, MYC, MYCL
  • the iRNA agent of the present invention are useful in a wide variety of in vitro applications.
  • Such in vitro applications include, for example, scientific and commercial research (e.g., elucidation of physiological pathways, drug discovery and development), and medical and veterinary diagnostics.
  • the method involves the introduction of the iRNA agent into a cell using known techniques (e.g., absorption through cellular processes, or by auxiliary agents or devices, such as electroporation and lipofection), then maintaining the cell for a time sufficient to obtain degradation of an mRNA transcript of the target gene.
  • the invention relates to methods for identifying iRNA agent having increased stability in biological tissues and fluids such as serum.
  • iRNA agent having increased stability have enhanced resistance to degradation, e.g., by chemicals or nucleases (particularly endonucleases) which normally degrade RNA molecules.
  • Methods for detecting increases in nucleic acid stability are well known in the art. Any assay capable of measuring or detecting differences between a test iRNA agent and a control iRNA agent in any measurable physical parameter may be suitable for use in the methods of the present invention.
  • the stability of a particular iRNA agent can be evaluated indirectly by observing or measuring a property associated with the expression of the gene.
  • the relative stability of an iRNA agent can be determined by observing or detecting (1) an absence or observable decrease in the level of the protein encoded by the target gene, (2) an absence or observable decrease in the level of mRNA product from the target gene, and (3) a change or loss in phenotype associated with expression of the target gene.
  • the stability of an iRNA agent may be evaluated based on the degree of the inhibition of expression or function of the target gene, which in turn may be assessed based on a change in the disease condition of the patient, such as reduction in symptoms, remission, or a change in disease state.
  • the method includes preparing an iRNA agent as described above (e.g., through chemical synthesis), incubating the iRNA agent with a biological sample, then analyzing and identifying those iRNA agent that exhibit an increased stability as compared to a control iRNA agent.
  • iRNA agent is produced in vitro by mixing/annealing complementary single-stranded RNA strands, preferably in a molar ratio of at least about 3:7, more preferably in a molar ratio of about 4:6, and most preferably in essentially equal molar amounts (e.g., a molar ratio of about 5:5).
  • the single-stranded RNA strands are denatured prior to mixing/annealing, and the buffer in which the mixing/annealing reaction takes place contains a salt, preferably potassium chloride.
  • Single-stranded RNA strands may be synthesized by solid phase synthesis using, for example, an Expedite 8909 synthesizer (Applied Biosystems, Applera Kunststoff GmbH, Darmstadt, Germany), as described above.
  • iRNA agent are incubated with a biological sample under the conditions sufficient or optimal for enzymatic function. After incubating with a biological sample, the stability of the iRNA agent is analyzed by means conventional in the art, for example using RNA gel electrophoresis as exemplified herein.
  • the iRNA agent may be incubated at a concentration of 1-10 ⁇ M, preferably 2-8 ⁇ M, more preferably 3-6 ⁇ M, and most preferably 4-5 ⁇ M.
  • the incubation temperature is preferably between 25° C. and 45° C., more preferably between 35° C. and 40° C., and most preferably about 37° C.
  • the biological sample used in the incubation step may be derived from tissues, cells, biological fluids or isolates thereof.
  • the biological sample may be isolated from a subject, such as a whole organism or a subset of its tissues or cells.
  • the biological sample may also be a component part of the subject, such as a body fluid, including but not limited to blood, serum, plasma, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine, vaginal fluid and semen.
  • the biological sample is a serum derived from a blood sample of a subject.
  • the subject is preferably a mammal, more preferably a human or a mouse.
  • the method includes selecting an iRNA agent having increased stability by measuring the mRNA and/or protein expression levels of a target gene in a cell following introduction of the iRNA agent.
  • an iRNA agent of the invention inhibits expression of a target gene in a cell, and thus the method includes selecting an iRNA agent that induces a measurable reduction in expression of a target gene as compared to a control iRNA agent.
  • Assays that measure gene expression by monitoring RNA and/or protein levels can be performed within about 24 hours following uptake of the iRNA agent by the cell.
  • RNA levels can be measured by Northern blot techniques, RNAse Protection Assays, or Quality Control-PCR (QC-PCR) (including quantitative reverse transcription coupled PCR (RT-PCR)) and analogous methods known in the art.
  • Protein levels can be assayed, for example, by Western blot techniques, flow cytometry, or reporter gene expression (e.g., expression of a fluorescent reporter protein, such as green fluorescent protein (GFP)).
  • RNA and/or protein levels resulting from target gene expression can be measured at regular time intervals following introduction of the test iRNA agent, and the levels are compared to those following introduction of a control iRNA agent into cells.
  • a control iRNA agent can be a nonsensical iRNA agent (i.e., an iRNA agent having a scrambled sequence that does not target any nucleotide sequence in the subject), an iRNA agent that can target a gene not present in the subject (e.g., a luciferase gene, when the iRNA agent is tested in human cells), or an iRNA agent otherwise previously shown to be ineffective at silencing the target gene.
  • the mRNA and protein levels of the test sample and the control sample can be compared.
  • the test iRNA agent is selected as having increased stability when there is a measurable reduction in expression levels following absorption of the test iRNA agent as compared to the control iRNA agent.
  • mRNA and protein measurements can be made using any art-recognized technique (see, e.g., Chiang, M. Y., et al., J. Biol. Chem . (1991) 266:18162-71; Fisher, T, et al., Nucl. Acids Res . (1993) 21:3857; and Chen et al., J. Biol. Chem . (1996)271:28259).
  • an iRNA agent composition of the invention to inhibit gene expression can be measured using a variety of techniques known in the art. For example, Northern blot analysis can be used to measure the presence of RNA encoding a target protein. The level of the specific mRNA produced by the target gene can be measured, e.g., using RT-PCR. Because iRNA agent directs the sequence-specific degradation of endogenous mRNA through RNAi, the selection methods of the invention encompass any technique that is capable of detecting a measurable reduction in the target RNA. In yet another example, Western blots can be used to measure the amount of target protein present. In still another embodiment, a phenotype influenced by the amount of the protein can be detected. Techniques for performing Western blots are well known in the art (see, e.g., Chen, et al., J. Biol. Chem . (1996) 271:28259).
  • the target gene When the target gene is to be silenced by an iRNA agent that targets a promoter sequence of the target gene, the target gene can be fused to a reporter gene, and reporter gene expression (e.g., transcription and/or translation) can be monitored.
  • reporter gene expression e.g., transcription and/or translation
  • a portion of the target gene e.g., a portion including the target sequence
  • a reporter gene By monitoring a change in the expression of the reporter gene in the presence of the iRNA agent, it is possible to determine the effectiveness of the iRNA agent in inhibiting the expression of the reporter gene.
  • the expression levels of the reporter gene in the presence of the test iRNA agent versus a control iRNA agent are then compared.
  • the test iRNA agent is selected as having increased stability when there is a measurable reduction in expression levels of the reporter gene as compared to the control iRNA agent.
  • reporter genes useful for use in the present invention include, without limitation, those coding for luciferase, GFP, chloramphenicol acetyl transferase (CAT), ⁇ -galactosidase, and alkaline phosphatase. Suitable reporter genes are described, for example, in Current Protocols in Molecular Biology , John Wiley & Sons, New York (Ausubel, F.
  • Double stranded RNAs (300 pmol, ca. 4.2 ⁇ g) were incubated at 37° C. in 60 ⁇ l 90% human serum/10% phosphate buffered saline for 0, 15, 30, 60, 120 or 240 minutes, or 8 hours, 16 hours or 24 hours. A 10 ⁇ l sample was taken from the incubation mixture, frozen in liquid nitrogen and stored at ⁇ 80° C. until analysis.
  • samples were thawed, their constituents were isolated by phenol-extraction and ethanol-precipitation, separated on denaturing 14% polyacrylamide gels (6M Urea, 20% formamide, Carl Roth GmbH & Co KG Düsseldorf, Germany) and detected by staining with the “stains-all” reagent (Sigma-Aldrich Chemie GmbH, Steinheim, Germany). Reaction mixtures from incubation of selected dsRNAs were further analysed covering the time points 8 hours, 16 hours and 24 hours.
  • HeLa SS6 cells were grown at 37° C. in Dulbecco's modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 units/mL penicillin, and 100 ⁇ g/mL streptomycin (Invitrogen, Carlsbad, Calif.). Cells were passaged regularly to maintain exponential growth. Twenty-four hours prior to siRNA transfection, cells were seeded on opaque, white 96-well plates (Costar, Corning, N.Y.) at a concentration of 15,000 cells/well in 150 ⁇ L antibiotic-free, phenol red-free DMEM (Invitrogen).
  • DMEM Dulbecco's modified Eagle medium
  • FBS fetal bovine serum
  • streptomycin 100 ⁇ g/mL streptomycin
  • siRNAs In vitro activity of siRNAs was determined using a high-throughput 96-well plate format luciferase silencing assay.
  • Cells were first transiently transfected with plasmids encoding firefly (target) and renilla (control) luciferase.
  • DNA transfections were performed using Lipofectamine 2000 (Invitrogen) (0.5 ⁇ L/ ⁇ g total DNA) and the plasmids gWiz-Luc (Aldevron, Fargo, N.Dak.) (200 ng/well) and pRL-CMV (Promega, Madison, Wis.) (200 ng/well).
  • siRNA transfections were performed using TransIT-TKO (Mirus, Madison, Wis.) (0.3 mL/well). After 24 h, cells were analyzed for both firefly and renilla luciferase expression using a plate luminometer (VICTOR 2 , PerkinElmer, Boston, Mass.) and the Dual-Glo Luciferase Assay kit (Promega). Firefly/renilla luciferase expression ratios were used to determine percent gene silencing relative to mock-treated (no siRNA) controls.
  • dsRNAs including phosphorothioate modifications were incubated in 90% human serum at 37° C. as described above. Gel electropheresis of the RNA products revealed that the phosphorothioate modifications enhanced stability ( FIG. 1 ). Similar results were observed when 3′-Naproxen, 5-Nitroindole, alkyamino-dT, abasic pyrrolidine, and cationic C6 amino dT were conjugated to the 3′ end of dsRNAs ( FIGS. 2, 3 , and 4 ).
  • FIGS. 5, 6 , and 7 show the silencing activity of 3′ conjugated Naproxen, abasic pyrrolidine, and cationic C6 amino dT, respectively.
  • FIGS. 8 and 9 show that the 3′Naproxen conjugated dsRNAs had enhanced exonuclease resistance in mouse serum and liver tissue, as well as in human serum (see FIG. 2 ).
  • siRNA duplexes were prepared at a stock concentration of 1 ⁇ M in which either the sense (S) or antisense strand (AS) contained a trace amount of 5′- 32 P labeled material (e.g. 32 P-S/AS and S/ 32 P-AS). The presence of the end-labeled sense or antisense strand allowed for monitoring of the individual strand within the context of the siRNA duplex. Therefore, two duplex preparations were made for each siRNA sequence tested. siRNA duplexes were incubated in 90% human serum at a final concentration of 100 nM duplex. Samples were removed and quenched in a stop mix at appropriate times.
  • S sense
  • AS antisense strand
  • 5′ end labeling is to place an internal 32 P or 33 P label within either the sense or antisense strand. This labeling method is much more laborious than 5′ end labeling and currently we have no evidence that dephosphorylation occurs during serum incubation.
  • Serum stability of unmodified parent duplex The unmodified parent duplex, AL-DUP-1000, was used to establish the serum stability baseline for evaluating the effect of chemical modifications on nuclease resistance.
  • AL-DUP-1000 5′-CUUACGCUGAGUACUUCGAdTdT-3′
  • ALN-SEQ-1000 SEQ ID NO: 3′ dTdTGAAUGCGACUCAUGAAGCU-5′
  • ALN-SEQ-1001 SEQ ID NO:
  • AL-DUP-1000 was subjected to the serum stability assay to evaluate its inherent nuclease resistance and to define its degradation pattern ( FIG. 21 ).
  • Denaturing gel electrophoresis was used analyze AL-DUP-1000 in a human serum stability assay.
  • An siRNA duplex containing 5′ end-labeled sense RNA (*s/as) and a duplex containing 5′ end-labeled antisense RNA (as/s*) were each incubated in 90% human serum and time points were assayed at 10 seconds, 5 min, 15 min, 30 min, 1 hour, 2 hours and 4 hours.
  • the control was a 4 hour time point for siRNA duplex incubated in PBS buffer alone, OH-was the partial alkaline hydrolysis marker. This unmodified duplex was observed to be degraded by both 3′-5′ exonucleases and endonucleases ( FIG. 21 ).
  • duplexes that contained additional phosphorothioates at their 3′ ends were similar results obtained for duplexes that contained additional phosphorothioates at their 3′ ends (data not shown).
  • the placement of phosphorothioates at the endonucleolytic cleavage sites did not inhibit endonucleolytic cleavage at these sites (data not shown).
  • a single phosphorothioate or methylphosphonate between the two 3′ terminal nucleotides was sufficient to protect the 3′ ends from exonuclease degradation. Additional phosphorothioates at the 3′ ends appear to enhance this effect, which may be necessary for long term exposure to serum nucleases. TABLE 1 siRNA duplexes containing backbone modifications.
  • FIG. 23 Star in s/*as gel, AL-DUP-13ff
  • the second, weaker endonucleolytic site FIG. 23 , black star in *s/as
  • FIG. 23 compare AL-DUP-13ff to AL-DUP-1036
  • Protection of both sites with 2′OMe (AL-DUP-1363) resulted in reduced endonucleolytic cleavage at both sites ( FIG. 23 ).
  • 2′OMe substitution is consistent with the mechanism of endonucleolytic cleavage, which requires the 2′OH as a nucleophile in the cleavage reaction.
  • 2′OMe modification will also be an effective means to protect the 3′ overhang of single overhang siRNA duplexes where the 3′ overhang is composed of ribonucleotides.
  • 2′OMe substitution can be used to block the possible loss of the terminal two nucleotides by endonucleolytic cleavage and phosphorothioate can be used to protect from exonuclease degradation.
  • Cationic modifications enhanced nuclease resistance The effect of three different cationic chemical modifications on nuclease resistance was tested and compared to the parent unmodified duplex. The structures of the three cationic modifications tested are shown below. TABLE 3 siRNA duplexes containing cationic substitutions.
  • FIG. 18 shows the serum stability data for the naproxen modified duplex (AL-DUP-1069) and similar results were obtained for AL-DUP1413. Presumably the conjugates inhibit exonucleolytic cleavage by sterically blocking the exonuclease from binding to the 3′ end of the siRNA duplex. Similar data was also obtained for AL-DUP-1069 in pooled mouse serum. TABLE 4 siRNA duplexes containing 3 conjugates.

Abstract

This invention features modified iRNA agents having improved stability in cells and biological fluids, and methods of making and identifying iRNA agents having improved stability, and of using the iRNA agents to inhibit the expression or function of a target gene.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of International Application No. PCT/U.S.2004/011829, filed on Apr. 16, 2004, which claims the benefit of U.S. Provisional Application No. 60/493,986, filed on Aug. 8, 2003; U.S. Provisional Application No. 60/494,597, filed on Aug. 11, 2003; U.S. Provisional Application No. 60/506,341, filed on Sep. 26, 2003; U.S. Provisional Application No. 60/518,453, filed on Nov. 7, 2003;U.S. Provisional Application No. 60/463, 772, filed on Apr. 17, 2003; U.S. Provisional Application No. 60/465,802, filed on Apr. 25, 2003; U.S. Provisional Application No. 60/469,612, filed on May 9, 2003; U.S. Provisional Application No. 60/510,246, filed on Oct. 9, 2003; U.S. Provisional Application No. 60/510,318, filed on Oct. 10, 2003; U.S. Provisional Application No. 60/503,414, filed on Sep. 15, 2003; U.S. Provisional Application No. 60/465,665, filed on Apr. 25, 2003; International Application No.: PCT/U.S.04/07070, filed on Mar. 8, 2004; International Application No.: PCT/U.S.2004/10586, filed on Apr. 5, 2004; International Application No.: PCT/U.S.2004/11255, filed on Apr. 9, 2004; and International Application No.: PCT/U.S.2004/011822, filed on Apr. 16, 2004. The contents of all of these prior applications are hereby incorporated by reference in their entireties.
  • TECHNICAL FIELD
  • This invention features modified iRNA agents having improved stability in cells and biological fluids, and methods of making and identifying iRNA agents having improved stability, and of using the iRNA agents to inhibit the expression or function of a target gene.
  • BACKGROUND
  • Many diseases (e.g., cancers, hematopoietic disorders, endocrine disorders, and immune disorders) arise from the abnormal expression or activity of a particular gene or group of genes. Similarly, disease can result through expression of a mutant form of protein, as well as from expression of viral genes that have been integrated into the genome of their host. The therapeutic benefits of being able to selectively silence these abnormal or foreign genes are obvious.
  • Double-stranded RNA molecules (dsRNA) can block gene expression by virtue of a highly conserved regulatory mechanism known as RNA interference (RNAi). Briefly, the RNA III Dicer enzyme processes dsRNA into small interfering RNA (siRNA) of approximately 22 nucleotides. One strand of the siRNA (the “antisense strand”) then serves as a guide sequence to induce cleavage of messenger RNAs (mRNAs) including a nucleotide sequence which is at least partially complementary to the sequence of the antisense strand by an RNA-induced silencing complex RISC (Hammond, S. M., et al., Nature (2000) 404:293-296). The antisense strand is not cleaved or otherwise degraded in this process, and the RISC including the antisense strand can subsequently effect the cleavage of further mRNAs.
  • SUMMARY
  • The invention features iRNA agents having improved stability in cells and biological fluids, methods of making and identifying iRNA agents having improved stability, as well as compositions and methods for inhibiting the expression of a target gene in a cell using the iRNA agent. The invention also features compositions and methods for treating diseases caused by the expression or activity of the target gene. The invention features iRNA agents that contain modifications that increase resistance of the iRNA agents to exonucleolytic degradation, including 5′ exonucleolytice and 3′ exonucleolytic degradation. The modifications render the iRNA agents more resistant to enzymatic or chemical degradation, and thus more stable and bioavailable than an otherwise identical iRNA agent that does not include the modification.
  • In one aspect, the invention features iRNA agents having improved stability when administered to a subject. These agents can have, e.g., improved stability in cells and biological samples, particularly serum. The iRNA agent includes one or more of various modifications. For example, the iRNA agents can include a backbone modification to one or more of the terminal nucleotides on an iRNA strand. A terminal nucleotide can be the nucleotide at the last position, and can include the nucleotide(s) at the second, third, fourth, and fifth positions from the 5′ or 3′-most ends of the sense or antisense strand of an iRNA agent. In one embodiment, an iRNA agent includes a phosphorothioate linkage or P-alkyl modification in the linkages between one or more of the terminal nucleotides of an iRNA agent. In another embodiment, one or more terminal nucleotides of an iRNA agent include a sugar modification, e.g., a 2′ or 3′ sugar modification. Exemplary sugar modifications include, for example, a 2′-O-methylated nucleotide, a 2′-deoxy nucleotide, a 2′-deoxyfluoro nucleotide, a 2′-O-methoxyethyl nucleotide, a 2′-O-NMA, a 2′-DMAEOE, a 2′-AP, 2′-hydroxy, or a 2′-ara-fluoro or a locked nucleic acid (LNA), extended nucleic acid (ENA), hexose nucleic acid (HNA), or cyclohexene nucleic acid (CeNA). A 2′ modification is preferably 2′OMe, more preferably, 2′fluoro. In one embodiment the modification is an LNA and the LNA is on the 5′ or 3′ end of the sense strand, and not on the 5′ or 3′ end of the antisense strand.
  • In one embodiment, the iRNA agent includes a 3′ sugar modification, e.g., a 3′OMe modification. Preferably a 3′OMe modificationis on the 3′ or 5′ end of the sense strand of the iRNA agent.
  • In some embodiments, an iRNA agent includes a 5′-methyl-pyrimidine (e.g., a 5′-methyl-uridine modification or a 5′-methyl-cytodine modification.
  • The modifications described herein can be combined onto a single iRNA agent. For example, in one embodiment, at least one terminal nucleotide of an iRNA agent has a phosphorothioate linkage and a 2′ sugar modification, e.g., a 2′OMe or 2° F. modification. In another embodiment, at least one terminal nucleotide of an iRNA agent has a 5′ Me-pyrimidine and a 2′ sugar modification, e.g., a 2′F or 2′OMe modification.
  • In one embodiment, an iRNA agent includes a nucleobase modification, such as a cationic modification, such as a 3′-abasic cationic modification. The cationic modification can be e.g., an alkylamino-dT (e.g., a C6 amino-dT), an allylamino conjugate, a pyrrolidine conjugate, a pthalamido or a hydroxyprolinol conjugate, on one or more of the terminal nucleotides of the iRNA agent. An alkylamino-dT conjugate is preferably attached to the 3′ end of the sense or antisense strand of an iRNA agent. A pyrrolidine linker is preferably attached to the 3′ or 5′ end of the sense strand, or the 3′ end of the antisense strand. An allyl amine uridine is preferably on the 3′ or 5′ end of the sense strand, and not on the 5′ end of the antisense strand.
  • In another embodiment, the iRNA agent includes an conjugate on one or more of the terminal nucleotides of the iRNA agent. The conjugate can be, for example, a lipophile, a terpene, a protein binding agent, a vitamin, a carbohydrate, or a peptide. For example, the conjugate can be naproxen, nitroindole (or another conjugate that contributes to stacking interactions), folate, ibuprofen, or a C5 pyrimidine linker. In other embodiments, the conjugates are glyceride lipid conjugates (e.g. a dialkyl glyceride derivatives), vitamin E conjugates, or thio-cholesterols. Preferably, conjugates are on the 3′ end of the antisense strand, or on the 5′ or 3′ end of the sense strand, and preferably the conjugates are not on the 3′ end of the antisense strand and on the 3′ end of the sense strand.
  • In one embodiment, the conjugate is naproxen, and the conjugate is preferably on the 5′ or 3′ end of the sense or antisense strands. In one embodiment, the conjugate is cholesterol, and the conjugate is preferably on the 5′ or 3′ end of the sense strand and preferably not present on the antisense strand. In some embodiments, the cholesterol is conjugated to the iRNA agent by a pyrrolidine linker, or serinol linker, or hydroxyprolinol linker. In other embodiments, the conjugate is a dU-cholesterol, or cholesterol is conjugated to the iRNA agent by a disulfide linkage. In a particularly prefered embodiment, the conjugate is cholanic acid, and the cholanic acid is attached to the 5′ or 3′ end of the sense strand, or the 3′ end of the antisense strand. In one embodiment, the cholanic acid is attached to the 3′ end of the sense strand and the 3′ end of the antisense strand.
  • In another embodiment, one or more terminal nucleotides has a 2′-5′ linkage. Preferably, a 2′-5′ linkage occurs on the sense strand, e.g., the 5′ end of the sense strand.
  • In one embodiment, the iRNA agent includes an L-sugar, preferably at the 5′ or 3′ end of the sense strand.
  • In one embodiment, the iRNA agent includes a methylphosphonate at one or more terminal nucleotides to enhance exonuclease resistance, e.g., at the 3′ end of the sense or antisense strands of the iRNA agent.
  • In one embodiment, an iRNA agent has been modified by replacing one or more ribonucleotides with deoxyribonucleotides. Preferably, adjacent deoxyribonucleotides are joined by phosphorothioate linkages, and the iRNA agent does not include more than four consecutive deoxyribonucleotides on the sense or the antisense strands.
  • In some embodiments, an iRNA agent having increased stability in cells and biological samples includes a difluorotoluyl (DFT) modification, e.g., 2,4-difluorotoluyl uracil, or a guanidine to inosine substitution.
  • In one embodiment the iRNA agent includes a terminal 5′-uridine-adenine-3′ (5′-UA-3′) dinucleotide wherein the uridine is a 2′-modified nucleotide, or a terminal 5′-uridine-guanine-3′ (5′-UG-3′) dinucleotide, wherein the 5′-uridine is a 2′-modified nucleotide, or a terminal 5′-cytidine-adenine-3′ (5′-CA-3′) dinucleotide, wherein the 5′-cytidine is a 2′-modified nucleotide, or a terminal 5′-uridine-uridine-3′ (5′-UU-3′) dinucleotide, wherein the 5′-uridine is a 2′-modified nucleotide, or a terminal 5′-cytidine-cytidine-3′ (5′-CC-3′) dinucleotide, wherein the 5′-cytidine is a 2′-modified nucleotide, or a terminal 5′-cytidine-uridine-3′ (5′-CU-3′) dinucleotide, wherein the 5′-cytidine is a 2′-modified nucleotide, or a terminal 5′-uridine-cytidine-3′ (5′-UC-3′) dinucleotide, wherein the 5′-uridine is a 2′-modified nucleotide. The chemically modified nucleotide in the iRNA agent may be a 2′-O-methylated nucleotide. In some embodiments, the modified nucleotide can be a 2′-deoxy nucleotide, a 2′-deoxyfluoro nucleotide, a 2′-O-methoxyethyl nucleotide, a 2′-O-NMA, a 2′-DMAEOE, a 2′-AP, 2′-hydroxy, or a 2′-ara-fluoro, or a locked nucleic acid (LNA), extended nucleic acid (ENA), hexose nucleic acid (HNA), or cyclohexene nucleic acid (CeNA). The iRNA agents including the modifications described herein are particularly stabilized against exonuclease activity.
  • In one particularly preferred embodiment, an iRNA agent includes a cholesterol molecule on the 3′ end of an iRNA agent, and conjugated to the iRNA agent by a pyrrolidine linker, and at least one additional modification, e.g., a modification described herein, such as a phosphorothioate linkage, for inhibiting exonucleolytic degradation. In another particularly preferred embodiment, an iRNA agent includes a cholanic acid modification on one or both of the 3′ ends of the sense or antisense strands, and at least one additional modification, e.g., a modification described herein, such as a phosphorothioate linkage, for inhibiting exonucleolytic degradation.
  • In one embodiment, the iRNA agent has a single overhang, e.g., one end of the iRNA agent has a 3′ or 5′ overhang and the other end of the iRNA agent is a blunt end. In another embodiment, the iRNA agent has a double overhang, e.g., both ends of the iRNA agent have a 3′ or 5′ overhang, such as a dinuclotide overhang. In another embodiment, both ends of the iRNA agent have blunt ends.
  • The iRNA agent may further include a sense RNA strand and an antisense RNA strand, wherein the antisense RNA strand is 25 or fewer nucleotides in length, and includes an antisense nucleotide sequence having 18-25 nucleotides in length. The iRNA agent may further include a nucleotide overhang having 1 to 4 unpaired nucleotides, which may be at the 3′-end of the antisense RNA strand, and the nucleotide overhang may have the nucleotide sequence 5′-GC-3′ or 5′-CGC-3′. The unpaired nucleotides may have at least one phosphorothioate dinucleotide linkage, and at least one of the unpaired nucleotides may be chemically modified in the 2′-position. In one embodiment, the doublestrand region of the iRNA agent includes phosphorothioate dinucleotide linkages on one or both of the sense and antisense strands. In one embodiment, the antisense RNA strand and the sense RNA strand are connected with a linker. The chemical linker may be a hexaethylene glycol linker, a poly-(oxyphosphinico-oxy-1,3-propandiol) linker, an allyl linker, or a polyethylene glycol linker.
  • In another aspect, the invention relates to a method of making an iRNA agent. The method includes preparing an antisense RNA strand and a sense RNA strand, wherein at least one of the RNA strands includes a modification described herein, e.g., a modification that confers an enhanced resistance to exonucleolytic cleavage. The RNA strands may then be mixed to form a double stranded iRNA agent. The RNA strands may be prepared using solid-phase synthesis in a 3′ to 5′ direction.
  • In yet another aspect, the invention relates to a method for inhibiting the expression of a target gene in a cell. The method includes introducing an iRNA agent, as described above, into the cell, and maintaining the cell for a time sufficient to obtain iRNA agent-mediated downregulation, e.g., by degradation, of an mRNA transcript of the target gene. The cell may be a mammalian cell, such as a human cell.
  • In still another aspect, the invention relates to a pharmaceutical composition for inhibiting the expression of a target gene in an organism. The compositions include an iRNA agent, as described above, and a pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier may be an aqueous solution, such as phosphate buffered saline, or it may include a micellar structure, such as a liposome, capsid, capsoid, polymeric nanocapsule, or polymeric microcapsule.
  • In yet a further aspect, the invention relates to a method for treating a disease caused by the expression of a target gene in a subject. The method includes administering a pharmaceutical composition containing an iRNA agent, as described above, and a pharmaceutically acceptable carrier. The subject may be a mammal, such as a human.
  • In another aspect, the invention relates to methods of identifying an iRNA agent having increased stability in a biological sample. The method may include preparing a mixture of iRNA agent, incubating the mixture of iRNA agent in the biological sample, and identifying an iRNA agent exhibiting an increased stability as compared to other iRNA agent in the biological sample. The mixture of iRNA agent may be prepared by chemical synthesis or by extraction from a biological sample. Alternatively, the method may include introducing an iRNA agent into a cell, maintaining the cell under conditions suitable for expressing a protein encoded by a target gene, measuring an amount of the protein produced in the cell, comparing the amount of the protein produced in the cell to that in a control cell, and identifying an iRNA agent that causes a reduction in the amount of protein in the cell as compared to the control cell. The cell may be a mammalian cell.
  • In still another aspect, the invention relates to a method of increasing the nuclease resistance of an iRNA agent. The method includes forming an iRNA agent, as described above, wherein the nuclease resistance of the iRNA agent is increased compared to a control composition. Such method includes the steps of incorporating into the iRNA agent, a modification described herein. The modification can be added to one or more of the 5′ or 3′ terminal nucleotides of the sense and antisense strands of the iRNA agent.
  • While RNA interference using iRNA agent has been shown to be an effective means for selective gene silencing, RNA can have less than desired stability in some bodily fluids, particularly in serum. Thus, RNA, including iRNA agent, can be degraded between the time it is administered to a subject and the time it enters a target cell. Even within the cell, RNA can undergo rapid degradation by nucleases. Methods of the invention can provide more stable or nuclease resistant iRNA agents that can offer better bioavailability and hence improved effectiveness. This is an improvement over the current insufficient methods for stabilizing iRNA agent against degradation.
  • The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
  • DESCRIPTION OF DRAWINGS
  • FIG. 1 a general synthetic scheme for incorporation of RRMS monomers into an oligonucleotide.
  • FIG. 2A is a list of substituents that may be present on silicon in OFG1.
  • FIG. 2B is a list of substituents that may be present on the C2′-orthoester group.
  • FIG. 3 is list of representative RRMS cyclic carriers. Panel 1 shows pyrroline-based RRMSs; panel 2 shows 3-hydroxyproline-based RRMSs; panel 3 shows piperidine-based RRMSs; panel 4 shows morpholine and piperazine-based RRMSs; and panel 5 shows decalin-based RRMSs. R1 is succinate or phosphoramidate and R2 is H or a conjugate ligand.
  • FIG. 4 is a general reaction scheme for 3′ conjugation of peptide into iRNA.
  • FIG. 5 is a general reaction scheme for 5′ conjugation of peptide into iRNA.
  • FIG. 6 is a general reaction scheme for the synthesis of aza-peptides.
  • FIG. 7 is a general reaction scheme for the synthesis of N-methyl amino acids and peptides.
  • FIG. 8 is a general reaction scheme for the synthesis of β-methyl amino acids and Ant and Tat peptides.
  • FIG. 9 is a general reaction scheme for the synthesis of Ant and Tat oligocarbamates.
  • FIG. 10 is a a general reaction scheme for the synthesis of Ant and Tat oligoureas.
  • FIG. 11 is a schematic representation of peptide carriers.
  • FIG. 12 shows a gel electrophoretic separation of 3′ phosphorothiate modified dsRNAs following incubation in human serum.
  • FIG. 13 shows a gel electrophoretic separation of 3′ modified dsRNAs following incubation in human serum.
  • FIG. 14 shows a gel electrophoretic separation of 3′ abasic pyrrolidine conjugated dsRNAs following incubation in human serum.
  • FIG. 15 shows a gel electrophoretic separation of 3′ cationic C6 amino dT conjugated dsRNAs following incubation in human serum.
  • FIG. 16 is a graph showing luciferase gene expression activity in cells following transfection of 3′Naproxen modified dsRNAs.
  • FIG. 17 is a graph showing luciferase gene expression activity in cells following transfection of 3′abasic pyrrolidine modified dsRNAs.
  • FIG. 18 is a graph showing luciferase gene expression activity in cells following transfection of 3′ cationic C6 amino dT modified dsRNAs.
  • FIG. 19 shows a gel electrophoretic separation of 3′ naproxen conjugated dsRNAs following incubation in mouse serum.
  • FIG. 20 shows a gel electrophoretic separation of 3′ naproxen conjugated dsRNAs following incubation in liver extract.
  • FIG. 21 is a denaturing gel analysis of the human serum stability assay for AL-DUP-1000. C is the 4 hour time point for siRNA duplex incubated in PBS buffer alone, OH— is the partial alkaline hydrolysis marker, *s/as represents siRNA duplex containing 5′ end-labeled sense RNA and s/*as represents duplex containing 5′ end-labeled antisense RNA. Samples were incubated in 90% human serum and time points were assayed at 10 seconds, 5 min, 15 min, 30 min, 1 hour, 2 hours and 4 hours. Black lines to the right of bands indicate exonucleolytic degradation fragments and the red lines highlight a few of the endonucleolytic degradation fragment.
  • FIG. 22A is a denaturing gel analysis of the human serum stability assay for AL-DUP-1393. C is the 4 hour time point for each siRNA duplex incubated in PBS buffer alone, *s/as represents siRNA duplex containing 5′ end-labeled sense RNA and s/*as represents duplex containing 5′ end-labeled antisense RNA. Samples were assayed at 10 seconds, 15 min, 30 min, 1 hour, 2 hours and 4 hours.
  • FIG. 22B is a denaturing gel analysis of the human serum stability assay for AL-DUP-1329. The lanes are labeled and the experiment was performed as described for FIG. 22A.
  • FIG. 23 is a denaturing gel analysis of AL-DUP-1036, AL-DUP-13ff, and AL-DUP-1363 (see Table 2 for sequences). Black vertical lines highlight regions where exonuclease cleavage is suppressed, stars indicate sites of strong endonucleolytic cleavage in the antisense strand and weaker endonucleolytic cleavage in the sense strand. C is the 4 hour time point for each siRNA duplex incubated in PBS buffer alone, *s/as represents siRNA duplex containing 5′ end-labeled sense RNA and s/*as represents duplex containing 5′ end-labeled antisense RNA. Samples were assayed at 10 seconds, 15 min, 30 min, 1 hour, 2 hours and 4 hours.
  • FIG. 24. Human serum stability profile of siRNA duplexes containing cationic modifications. Denaturing gel analysis of AL-DUP-10aa (alkylamino-dT), AL-DUP-1ccc (abasic pyrrolidine cationic), and AL-DUP-1403 (see Table 3 for sequences). Black line highlights region where exonuclease cleavage is suppressed and red star indicates site of strong endonucleolytic cleavage in the antisense strand. C is the 4 hour time point for each siRNA duplex incubated in PBS buffer alone, *s/as represents siRNA duplex containing 5′ end-labeled sense RNA and s/*as represents duplex containing 5′ end-labeled antisense RNA. Samples were assayed at 10 seconds, 15 min, 30 min, 1 hour, 2 hours and 4 hours.
  • FIG. 25 is a denaturing gel analysis of the human serum stability assay for AL-DUP-1069. The black vertical line highlights the region where exonuclease cleavage is suppressed. C is the 4 hour time point for each siRNA duplex incubated in PBS buffer alone, *s/as represents siRNA duplex containing 5′ end-labeled sense RNA and s/*as represents duplex containing 5′ end-labeled antisense RNA. Samples were assayed at 10 seconds, 15 min, 30 min, 1 hour, 2 hours and 4 hours.
  • Like reference symbols in the various drawings indicate like elements.
  • DETAILED DESCRIPTION
  • The invention is based, at least in part, on the discovery that particular modifications to an iRNA agent are effective at increasing resistance of the iRNA agent to exonucleolytic degradation. Thus, the invention features iRNA agents that contain modifications that increase resistance of the iRNA agents to exonucleolytic degradation, including 5′ exonucleolytic and 3′ exonucleolytic degradation. These iRNA agents may have improved stability in cells and biological fluids.
  • The iRNA agents having improved stability can include one or more of various modifications. For example, the iRNA agent can include a backbone modification to one or more of the terminal nucleotides on an iRNA strand. Exemplary backbone modifications include phosphorothioate linkages or P-alkyl linkages between one or more of the terminal nucleotides of an iRNA agent. One or more terminal nucleotides of an iRNA agent can also include a sugar modification, e.g., a 2′ or 3′ sugar modification. Exemplary sugar modifications are described below. An iRNA agent can include multiple modifications that confer an enhanced resistance to exonucleolytic cleavage. For example, one or more terminal nucleotides of an iRNA agent can have a phosphorothioate linkage and a 2′ sugar modification.
  • An iRNA agent having increased resistance to exonucleolytice degradation can include a nucleobase modification, a cationic modification on one or more of the terminal nucleotides of the iRNA agent.
  • Exemplary modifications for enhanced resistance to endonucleolytic cleavage are described below.
    Figure US20050119214A1-20050602-C00001
    Figure US20050119214A1-20050602-C00002
    Figure US20050119214A1-20050602-C00003
  • Most Preferred Sugar with 2′-Substituents and Backbones
    Figure US20050119214A1-20050602-C00004
    Figure US20050119214A1-20050602-C00005
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00006
    • R1, R2=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5H, Me, Et and R6=H, Me, Et or a ligand
    • X=OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X−O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00007
    • R1, R2=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00008
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R6=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00009
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R6=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Z=O, S, CH2, CHF, CF2
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00010
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8═H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00011
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Z1, Z2=H, OH, NR′R″, SR′″, OR″″
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00012
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R6=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00013
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Z1, Z2=O, S, NR′, CH2, CHF, CF
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00014
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8═H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Z=O, S, NR′, CH2, CHF, CF
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00015
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8═H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Z1=O, S, NR′, CH2, CHF, CF
    • Z2═H, OR′, SR′, NR′R″
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00016
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5H, Me, Et and R6=H, Me, Et or a ligand
        • Y=O, S, CH2, CHF, CF2
          Figure US20050119214A1-20050602-C00017
          Figure US20050119214A1-20050602-C00018
    • R=H, OH, OMe, F, O(CH2)2O(CH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X≡OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Z=O, S, NR′, CH2, CHF, CF2
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00019
    • R=H, OH, OMe, F, O(CH2)OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R6=H, Me, Et and R6=H, Me, Et or a ligand
    • X=OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Z=O, S, NR′, CH2, CHF, CF2
    • R′=H, Me, Et, (CH2)nOH; n=0-6
    • Chiral Rp and Sp isomers, P=S preferably Sp
      L-Nucleoside Derived from 1-15
      2′-5′ Linkage from 1-15
      Figure US20050119214A1-20050602-C00020
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
      Figure US20050119214A1-20050602-C00021
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
      Figure US20050119214A1-20050602-C00022
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X≡OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
      Figure US20050119214A1-20050602-C00023
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
      Figure US20050119214A1-20050602-C00024
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —N H-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
      Figure US20050119214A1-20050602-C00025
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
      Figure US20050119214A1-20050602-C00026
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R6=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00027
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Z=OH, OMe, SH, SMe, NH2, NHMe, NMe2, Aryl
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00028
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R5=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Z=OH, OMe, SH, SMe, NH2, NHMe, NMe2, Aryl, alkyl
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00029
    • R=H, OH, OMe, F, O(CH2)2OCH3, O(CH2)2SCH3, O(CH2)2ONMe2, O(CH2)2O(CH2)2NMe2, OCH2CONH2, O(CH2)nXR3, where X=O, S, NR4 and R3, R4=H, Me, Et,; OCH2CONR5R6, where R5, R6, where R6=H, Me, Et and R6=H, Me, Et or a ligand
    • X=—OH, —SH, -Me, -isopropyl, -tert-Butyl, -phenyl, —NH-Alkyl, —N(dialkyl), —N(alkyl)(Aryl), —(CH2)nXR7 or —NH(CH2)XR7, where X=O, S, NR8, R7=H, Me, Et, and R8=H, Me, Et, COMe and n=1-12
    • Y=O, S or Se
    • Chiral Rp and Sp isomers, P=S preferably Sp
      Figure US20050119214A1-20050602-C00030
      Figure US20050119214A1-20050602-C00031
      Figure US20050119214A1-20050602-C00032
    • R=H, OH or OMe, X=O, NH, NMe, S, CH2, or CF2; Y=O or S Q′, Q″=H; Q′=H and Q″=Me or Et; or Q′, Q″=Me or Et and n=0-5
      Figure US20050119214A1-20050602-C00033
      Figure US20050119214A1-20050602-C00034
      Figure US20050119214A1-20050602-C00035
    • R=H, OH or OMe; X=O, NH, NMe, S, CH2, or CF2; Y=O or S Q′, Q″=H; Q′=H and Q″=Me or Et; or Q′, Q″=Me or Et and n=0-5
      Figure US20050119214A1-20050602-C00036
      Figure US20050119214A1-20050602-C00037
    • R=H, OH or OMe; X=O, NH, NMe, S, CH2, or CF2; Y=O or S; Q′, Q″=H Q′=H and Q″=Me or Et ir Q′, Q″=Me or Et; p=0-6 and q=0-6
      Figure US20050119214A1-20050602-C00038
      Figure US20050119214A1-20050602-C00039
    • R=H, OH or OMe; X=O, NH, NMe, S, CH2, or CF2; Y=O or S; Q′, Q″=H; Q′=H and Q″=Me or Et or Q′, Q″=Me or Et; p=0-6 and q=0-6
      Figure US20050119214A1-20050602-C00040
      Figure US20050119214A1-20050602-C00041
    • R=H, OH or OMe; X=O, NH, NMe, S, CH2, or CF2; Y=O or S; Q′, Q″=H; Q′=H and Q″=Me or Et or Q′, Q″=Me or Et; p=0-6 and q=0-6
      Figure US20050119214A1-20050602-C00042
      Figure US20050119214A1-20050602-C00043
      Figure US20050119214A1-20050602-C00044
    • R=H, OH or OMe; X=O, NH, NMe, S, CH2, or CF2; Y=O or S; Q′, Q″=H; Q′=H and Q″=Me or Et or Q′, Q″=Me or Et; n=0-6; p=0-6; q=0-6 and r=0-50
      Figure US20050119214A1-20050602-C00045
      Figure US20050119214A1-20050602-C00046
      Figure US20050119214A1-20050602-C00047
    • R=H, OH or OMe
      Figure US20050119214A1-20050602-C00048
    • R=H, OH or OMe
      Figure US20050119214A1-20050602-C00049
      Figure US20050119214A1-20050602-C00050
    • R=H, OH or OMe
      Figure US20050119214A1-20050602-C00051
      Figure US20050119214A1-20050602-C00052
    • R=H, OH or OMe
      Figure US20050119214A1-20050602-C00053
      Figure US20050119214A1-20050602-C00054
    • R=H, OH or OMe
      Figure US20050119214A1-20050602-C00055
      Figure US20050119214A1-20050602-C00056
    • R=H, OH or OMe
      Figure US20050119214A1-20050602-C00057
    • R=H, OH or OMe
  • For structures 27-81, R can also have the values for structures 1A-26.
  • The iRNA agent can include a conjugate (also called a ligand) on the 5′ or 3′ terminus of the sense or antisense strand. Exemplary ligands are shown below. The conjugate can be attached to the 3′ or 5′ end. Preferably, a cholesterol conjugate is on the 5′ or 3′ end of the sense strand. Preferably, a cholesterol conjugate is not on the antisense strand.
  • Preferred Ligands
  • Figure US20050119214A1-20050602-C00058
      • Vitamins
      • Peptides
      • Carbohydrates
      • Terpenes
      • Steroids
      • Lipids
      • Amino acids
      • Naproxen
      • Ibuprofen
      • Amines (mono, di, tri, tetraalkyl or aryl)
      • Folate
      • Sugar (N-Acetylgalactosamine, galactosamine, galactose, Mannose)
      • —(CH2)nNQ1Q2, where n=0-40, Q1, Q2=H, Me or Et; Q1=H, Q2=H, Me, Et or aryl
      • (CH2)pCH=CH(CH2)qNQ1Q2, where p and/or q=0-40, Q1, Q2=H, Me or Et; Q1=H, Q2=H, Me, Et or aryl with E and/or Z configuration
      • —(CH2)pCH=CH(CH2)qNQ1Q2, where p and/or q=0-40, Q1, Q2=H, Me or Et; Q1=H, Q2=H, Me, Et or aryl
      • —(CH2)pCH=CH(CH2)qCH=CH(CH2)rNQ1Q2, where p, q and/or r=0-40, Q1, Q2=H, Me or Et; Q1=H, Q2=H, Me, Et or aryl with E and/or Z configuration
      • —O(CH2)m(OCH2CH2)n—OR, where m, n=0-40 and R=H, Me, NQ1Q2, —C(O)NR′R″—C(S)NR′R″
      • —NH(CH2)m(OCH2CH2)n—OR, where m, n=0-40 and R=H, Me, NQ1Q2, —C(O)NR′R″—C(S)NR′R″
      • —O(CH2)m(NHCH2CH2)n—R, where m, n=0-40 and R=H, OH, Me, NQ1Q2, —C(O)NR′R″—C(S)NR′R″
      • —NH(CH2)m(NHCH2CH2)n—R, where m, n=0-40 and R=H, OH, Me, NQ1Q2, —C(O)NR′R″—C(S)NR′R″
      • Dialkylglycerol (sn3, sn1, sn2 and racemic) with number of methylene varies from 0-40
      • Diacylglycerol (sn3, sn1, sn2 and racemic) with number of methylene varies from 0-40
      • Dialkylglycerol (sn3, sn1, sn2 and racemic) with number of methylene varies from 0-40 and the alkyl chian contains one or more double bonds with E and/or Z isomers
      • Dlacylglycerol (sn3, sn1, sn2 and racemic) with number of methylene varies from 0-40 and the alkyl chian contains one or more double bonds with E and/or Z isomers
  • Double-stranded (dsRNA) directs the sequence-specific silencing of mRNA through a process known as RNA interference (RNAi). The process occurs in a wide variety of organisms, including mammals and other vertebrates.
  • It has been demonstrated that 21-23 nt fragments of dsRNA are sequence-specific mediators of RNA silencing, e.g., by causing RNA degradation. While not wishing to be bound by theory, it may be that a molecular signal, which may be merely the specific length of the fragments, present in these 21-23 nt fragments recruits cellular factors that mediate RNAi. Described herein are methods for preparing and administering these 21-23 nt fragments, and other iRNAs agents, and their use for specifically inactivating gene function. The use of iRNAs agents (or recombinantly produced or chemically synthesized oligonucleotides of the same or similar nature) enables the targeting of specific mRNAs for silencing in mammalian cells. In addition, longer dsRNA agent fragments can also be used, e.g., as described below.
  • Although, in mammalian cells, long dsRNAs can induce the interferon response, which is frequently deleterious, sRNAs do not trigger the interferon response, at least not to an extent that is deleterious to the cell and host. In particular, the length of the iRNA agent strands in an sRNA agent can be less than 31, 30, 28, 25, or 23 nt, e.g., sufficiently short to avoid inducing a deleterious interferon response. Thus, the administration of a composition of sRNA agent (e.g., formulated as described herein) to a mammalian cell can be used to silence expression of a target gene while circumventing the interferon response. Further, use of a discrete species of iRNA agent can be used to selectively target one allele of a target gene, e.g., in a subject heterozygous for the allele.
  • In a typical embodiment, the subject is a mammal such as a cow, horse, mouse, rat, dog, pig, goat, or a primate. The subject can be a dairy mammal (e.g., a cow, or goat) or other farmed animal (e.g., a chicken, turkey, sheep, pig, fish, shrimp). In a much preferred embodiment, the subject is a human, e.g., a normal individual or an individual that has, is diagnosed with, or is predicted to have a disease or disorder.
  • Ligand-Conjugated Monomer Subunits and Monomers for Oligonucleotide Synthesis
  • Definitions
  • The term “halo” refers to any radical of fluorine, chlorine, bromine or iodine.
  • The term “alkyl” refers to a hydrocarbon chain that may be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, C1-C12 alkyl indicates that the group may have from 1 to 12 (inclusive) carbon atoms in it. The term “haloalkyl” refers to an alkyl in which one or more hydrogen atoms are replaced by halo, and includes alkyl moieties in which all hydrogens have been replaced by halo (e.g., perfluoroalkyl). Alkyl and haloalkyl groups may be optionally inserted with O, N, or S. The terms “aralkyl” refers to an alkyl moiety in which an alkyl hydrogen atom is replaced by an aryl group. Aralkyl includes groups in which more than one hydrogen atom has been replaced by an aryl group. Examples of “aralkyl” include benzyl, 9-fluorenyl, benzhydryl, and trityl groups.
  • The term “alkenyl” refers to a straight or branched hydrocarbon chain containing 2-8 carbon atoms and characterized in having one or more double bonds. Examples of a typical alkenyl include, but not limited to, allyl, propenyl, 2-butenyl, 3-hexenyl and 3-octenyl groups. The term “alkynyl” refers to a straight or branched hydrocarbon chain containing 2-8 carbon atoms and characterized in having one or more triple bonds. Some examples of a typical alkynyl are ethynyl, 2-propynyl, and 3-methylbutynyl, and propargyl. The sp2 and sp3 carbons may optionally serve as the point of attachment of the alkenyl and alkynyl groups, respectively.
  • The terms “alkylamino” and “dialkylamino” refer to —NH(alkyl) and —N (alkyl)2 radicals respectively. The term “aralkylamino” refers to a —NH(aralkyl) radical. The term “alkoxy” refers to an —O-alkyl radical, and the terms “cycloalkoxy” and “aralkoxy” refer to an —O-cycloalkyl and O-aralkyl radicals respectively. The term “siloxy” refers to a R3SiO— radical. The term “mercapto” refers to an SH radical. The term “thioalkoxy” refers to an —S-alkyl radical.
  • The term “alkylene” refers to a divalent alkyl (i.e., —R—), e.g., —CH2—, —CH2CH2—, and —CH2CH2CH2—. The term “alkylenedioxo” refers to a divalent species of the structure —O—R—O—, in which R represents an alkylene.
  • The term “aryl” refers to an aromatic monocyclic, bicyclic, or tricyclic hydrocarbon ring system, wherein any ring atom can be substituted. Examples of aryl moieties include, but are not limited to, phenyl, naphthyl, anthracenyl, and pyrenyl.
  • The term “cycloalkyl” as employed herein includes saturated cyclic, bicyclic, tricyclic, or polycyclic hydrocarbon groups having 3 to 12 carbons, wherein any ring atom can be substituted. The cycloalkyl groups herein described may also contain fused rings. Fused rings are rings that share a common carbon-carbon bond or a common carbon atom (e.g., spiro-fused rings). Examples of cycloalkyl moieties include, but are not limited to, cyclohexyl, adamantyl, and norbornyl.
  • The term “heterocyclyl” refers to a nonaromatic 3-10 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3,1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein any ring atom can be substituted. The heterocyclyl groups herein described may also contain fused rings. Fused rings are rings that share a common carbon-carbon bond or a common carbon atom (e.g., spiro-fused rings). Examples of heterocyclyl include, but are not limited to tetrahydrofuranyl, tetrahydropyranyl, piperidinyl, morpholino, pyrrolinyl and pyrrolidinyl.
  • The term “cycloalkenyl” as employed herein includes partially unsaturated, nonaromatic, cyclic, bicyclic, tricyclic, or polycyclic hydrocarbon groups having 5 to 12 carbons, preferably 5 to 8 carbons, wherein any ring atom can be substituted. The cycloalkenyl groups herein described may also contain fused rings. Fused rings are rings that share a common carbon-carbon bond or a common carbon atom (e.g., spiro-fused rings). Examples of cycloalkenyl moieties include, but are not limited to cyclohexenyl, cyclohexadienyl, or norbornenyl.
  • The term “heterocycloalkenyl” refers to a partially saturated, nonaromatic 5-10 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein any ring atom can be substituted. The heterocycloalkenyl groups herein described may also contain fused rings. Fused rings are rings that share a common carbon-carbon bond or a common carbon atom (e.g., spiro-fused rings). Examples of heterocycloalkenyl include but are not limited to tetrahydropyridyl and dihydropyran.
  • The term “heteroaryl” refers to an aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S (e.g., carbon atoms and 1-3, 1-6, or 1-9 heteroatoms of N, O, or S if monocyclic, bicyclic, or tricyclic, respectively), wherein any ring atom can be substituted. The heteroaryl groups herein described may also contain fused rings that share a common carbon-carbon bond.
  • The term “oxo” refers to an oxygen atom, which forms a carbonyl when attached to carbon, an N-oxide when attached to nitrogen, and a sulfoxide or sulfone when attached to sulfur.
  • The term “acyl” refers to an alkylcarbonyl, cycloalkylcarbonyl, arylcarbonyl, heterocyclylcarbonyl, or heteroarylcarbonyl substituent, any of which may be further substituted by substituents.
  • The term “substituents” refers to a group “substituted” on an alkyl, cycloalkyl, alkenyl, alkynyl, heterocyclyl, heterocycloalkenyl, cycloalkenyl, aryl, or heteroaryl group at any atom of that group. Suitable substituents include, without limitation, alkyl, alkenyl, alkynyl, alkoxy, halo, hydroxy, cyano, nitro, amino, SO3H, sulfate, phosphate, perfluoroalkyl, perfluoroalkoxy, methylenedioxy, ethylenedioxy, carboxyl, oxo, thioxo, imino (alkyl, aryl, aralkyl), S(O)nalkyl (where n is 0-2), S(O)naryl (where n is 0-2), S(O)n heteroaryl (where n is 0-2), S(O)n heterocyclyl (where n is 0-2), amine (mono-, di-, alkyl, cycloalkyl, aralkyl, heteroaralkyl, and combinations thereof), ester (alkyl, aralkyl, heteroaralkyl), amide (mono-, di-, alkyl, aralkyl, heteroaralkyl, and combinations thereof), sulfonamide (mono-, di-, alkyl, aralkyl, heteroaralkyl, and combinations thereof), unsubstituted aryl, unsubstituted heteroaryl, unsubstituted heterocyclyl, and unsubstituted cycloalkyl. In one aspect, the substituents on a group are independently any one single, or any subset of the aforementioned substituents.
  • The terms “adeninyl, cytosinyl, guaninyl, thyminyl, and uracilyl” and the like refer to radicals of adenine, cytosine, guanine, thymine, and uracil.
  • A “protected” moiety refers to a reactive functional group, e.g., a hydroxyl group or an amino group, or a class of molecules, e.g., sugars, having one or more functional groups, in which the reactivity of the functional group is temporarily blocked by the presence of an attached protecting group. Protecting groups useful for the monomers and methods described herein can be found, e.g., in Greene, T. W., Protective Groups in Organic Synthesis (John Wiley and Sons: New York), 1981, which is hereby incorporated by reference.
  • General
  • An RNA agent, e.g., an iRNA agent, containing a preferred, but nonlimiting ligand-conjugated monomer subunit is presented as formula (II) below and in the scheme in FIG. 1. The carrier (also referred to in some embodiments as a “linker”) can be a cyclic or acyclic moiety and includes two “backbone attachment points” (e.g., hydroxyl groups) and a ligand. The ligand can be directly attached (e.g., conjugated) to the carrier or indirectly attached (e.g., conjugated) to the carrier by an intervening tether (e.g., an acyclic chain of one or more atoms; or a nucleobase, e.g., a naturally occurring nucleobase optionally having one or more chemical modifications, e.g., an unusual base; or a universal base). The carrier therefore also includes a “ligand or tethering attachment point” for the ligand and tether/tethered ligand, respectively.
  • The ligand-conjugated monomer subunit may be the 5′ or 3′ terminal subunit of the RNA molecule, i.e., one of the two “W” groups may be a hydroxyl group, and the other “W” group may be a chain of two or more unmodified or modified ribonucleotides. Alternatively, the ligand-conjugated monomer subunit may occupy an internal position, and both “W” groups may be one or more unmodified or modified ribonucleotides. More than one ligand-conjugated monomer subunit may be present in a RNA molecule, e.g., an iRNA agent. Preferred positions for inclusion of a tethered ligand-conjugated monomer subunits, e.g., one in which a lipophilic moiety, e.g., cholesterol, is tethered to the carrier are at the 3′ terminus, the 5′ terminus, or an internal position of the sense strand.
    Figure US20050119214A1-20050602-C00059
  • The modified RNA molecule of formula (II) can be obtained using oligonucleotide synthetic methods known in the art. In a preferred embodiment, the modified RNA molecule of formula (II) can be prepared by incorporating one or more of the corresponding monomer compounds (see, e.g., A, B, and C below and in the scheme in FIG. 1) into a growing sense or antisense strand, utilizing, e.g., phosphoramidite or H-phosphonate coupling strategies.
  • The monomers, e.g., a ligand-conjugated monomer, generally include two differently functionalized hydroxyl groups (OFG1 and OFG2), which are linked to the carrier molecule (see A below and in FIG. 1), and a ligand/tethering attachment point. As used herein, the term “functionalized hydroxyl group” means that the hydroxyl proton has been replaced by another substituent. As shown in representative structures B and C below and in FIG. 1, one hydroxyl group (OFG1) on the carrier is functionalized with a protecting group (PG). The other hydroxyl group (OFG2) can be functionalized with either (1) a liquid or solid phase synthesis support reagent (solid circle) directly or indirectly through a linker, L, as in B, or (2) a phosphorus-containing moiety, e.g., a phosphoramidite as in C. The tethering attachment point may be connected to a hydrogen atom, a suitable protecting group, a tether, or a tethered ligand at the time that the monomer is incorporated into the growing sense or antisense strand (see variable “R” in A below). Thus, the tethered ligand can be, but need not be attached to the monomer at the time that the monomer is incorporated into the growing strand. In certain embodiments, the tether, the ligand or the tethered ligand may be linked to a “precursor” ligand-conjugated monomer subunit after a “precursor” ligand-conjugated monomer subunit has been incorporated into the strand. The wavy line used below (and elsewhere herein) refers to a connection, and can represent a direct bond between the moiety and the attachment point or a tethering molecule which is interposed between the moiety and the attachment point. Directly tethered means the moiety is bound directly to the attachment point. Indirectly tethered means that there is a tether molecule interposed between the attachment point and the moiety.
    Figure US20050119214A1-20050602-C00060
  • The (OFG1) protecting group may be selected as desired, e.g., from T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991). The protecting group is preferably stable under amidite synthesis conditions, storage conditions, and oligonucleotide synthesis conditions. Hydroxyl groups, —OH, are nucleophilic groups (i.e., Lewis bases), which react through the oxygen with electrophiles (i.e., Lewis acids). Hydroxyl groups in which the hydrogen has been replaced with a protecting group, e.g., a triarylmethyl group or a trialkylsilyl group, are essentially unreactive as nucleophiles in displacement reactions. Thus, the protected hydroxyl group is useful in preventing e.g., homocoupling of compounds exemplified by structure C during oligonucleotide synthesis. In some embodiments, a preferred protecting group is the dimethoxytrityl group. In other embodiments, a preferred protecting group is a silicon-based protecting group having the formula below:
    Figure US20050119214A1-20050602-C00061
  • X5′, X5″, and X5′″ can be selected from substituted or unsubstituted alkyl, cycloalkyl, aryl, araklyl, heteroaryl, alkoxy, cycloalkoxy, aralkoxy, aryloxy, heteroaryloxy, or siloxy (i.e., R3SiO—, the three “R” groups can be any combination of the above listed groups). X5′, X5″, and X5′″, may all be the same or different; also contemplated is a combination in which two of X5′, X5″, and X5′″are identical and the third is different. In certain embodiments X5′, X5″, and X5′″ include at least one alkoxy or siloxy groups and may be any one of the groups listed in FIG. 2A, a preferred combination includes X5′, X5″=trimethylsiloxy and X5′″=1,3-(triphenylmethoxy)-2-propoxy or cyclododecyloxy.
  • Other preferred combinations of X5′, X5″, and X5′″ include those that result in OFG1 groups that meet the deprotection and stability criteria delineated below. The group is preferably stable under amidite synthesis conditions, storage conditions, and oligonucleotide synthesis conditions. Rapid removal, i.e., less than one minute, of the silyl group from e.g., a support-bound oligonucleotide is desirable because it can reduce synthesis times and thereby reduce exposure time of the growing oligonucleotide chain to the reagents. Oligonucleotide synthesis can be improved if the silyl protecting group is visible during deprotection, e.g., from the addition of a chromophore silyl substituent.
  • Selection of silyl protecting groups can be complicated by the competing demands of the essential characteristics of stability and facile removal, and the need to balance these competitive goals. Most substituents that increase stability can also increase the reaction time required for removal of the silyl group, potentially increasing the level of difficulty in removal of the group.
  • The addition of alkoxy and siloxy substituents to OFG1 silicon-containing protecting groups increases the susceptibility of the protecting groups to fluoride cleavage of the silylether bonds. Increasing the steric bulk of the substituents preserves stability while not decreasing fluoride lability to an equal extent. An appropriate balance of substituents on the silyl group makes a silyl ether a viable nucleoside protecting group.
  • Candidate OFG1 silicon-containing protecting groups may be tested by exposing a tetrahydrofuran solution of a preferred carrier bearing the candidate OFG1 group to five molar equivalents of tetrahydrofuran at room temperature. The reaction time may be determined by monitoring the disappearance of the starting material by thin layer chromatography.
  • When the OFG2 in B includes a linker, e.g., a relatively long organic linker, connected to a soluble or insoluble support reagent, solution or solid phase synthesis techniques can be employed to build up a chain of natural and/or modified ribonucleotides once OFG1 is deprotected and free to react as a nucleophile with another nucleoside or monomer containing an electrophilic group (e.g., an amidite group). Alternatively, a natural or modified ribonucleotide or oligoribonucleotide chain can be coupled to monomer C via an amidite group or H-phosphonate group at OFG2. Subsequent to this operation, OFG1 can be deblocked, and the restored nucleophilic hydroxyl group can react with another nucleoside or monomer containing an electrophilic group. R′ can be substituted or unsubstituted alkyl or alkenyl. In preferred embodiments, R′ is methyl, allyl or 2-cyanoethyl. R″ may a C1-C10 alkyl group, preferably it is a branched group containing three or more carbons, e.g., isopropyl.
  • OFG2 in B can be hydroxyl functionalized with a linker, which in turn contains a liquid or solid phase synthesis support reagent at the other linker terminus. The support reagent can be any support medium that can support the monomers described herein. The monomer can be attached to an insoluble support via a linker, L, which allows the monomer (and the growing chain) to be solubilized in the solvent in which the support is placed. The solubilized, yet immobilized, monomer can react with reagents in the surrounding solvent; unreacted reagents and soluble by-products can be readily washed away from the solid support to which the monomer or monomer-derived products is attached. Alternatively, the monomer can be attached to a soluble support moiety, e.g., polyethylene glycol (PEG) and liquid phase synthesis techniques can be used to build up the chain. Linker and support medium selection is within skill of the art. Generally the linker may be —C(O)(CH2)qC(O)—, or —C(O)(CH2)qS—, in which q can be 0, 1, 2, 3, or 4; preferably, it is oxalyl, succinyl or thioglycolyl. Standard control pore glass solid phase synthesis supports can not be used in conjunction with fluoride labile 5′ silyl protecting groups because the glass is degraded by fluoride with a significant reduction in the amount of full-length product. Fluoride-stable polystyrene based supports or PEG are preferred.
  • The ligand/tethering attachment point can be any divalent, trivalent, tetravalent, pentavalent or hexavalent atom. In some embodiments, ligand/tethering attachment point can be a carbon, oxygen, nitrogen or sulfur atom. For example, a ligand/tethering attachment point precursor functional group can have a nucleophilic heteroatom, e.g., —SH, —NH2, secondary amino, ONH2, or NH2NH2. As another example, the ligand/tethering attachment point precursor functional group can be an olefin, e.g., —CH=CH2, and the precursor functional group can be attached to a ligand, a tether, or tethered ligand using, e.g., transition metal catalyzed carbon-carbon (for example olefin metathesis) processes or cycloadditions (e.g., Diels-Alder). As a further example, the ligand/tethering attachment point precursor functional group can be an electrophilic moiety, e.g., an aldehyde. When the carrier is a cyclic carrier, the ligand/tethering attachment point can be an endocyclic atom (i.e., a constituent atom in the cyclic moiety, e.g., a nitrogen atom) or an exocyclic atom (i.e., an atom or group of atoms attached to a constituent atom in the cyclic moiety).
  • The carrier can be any organic molecule containing attachment points for OFG1, OFG2, and the ligand. In certain embodiments, carrier is a cyclic molecule and may contain heteroatoms (e.g., O, N or S). E.g., carrier molecules may include aryl (e.g., benzene, biphenyl, etc.), cycloalkyl (e.g., cyclohexane, cis or trans decalin, etc.), cycloalkenyl (e.g., cyclohexenyl), or heterocyclyl (tetrahydropyran, piperazine, pyrrolidine, etc.). In other embodiments, the carrier can be an acyclic moiety, e.g., based on serinol. Any of the above cyclic systems may include substituents in addition to OFG1, OFG2, and the ligand.
  • Sugar-Based Monomers
  • In some embodiments, the carrier molecule is an oxygen containing heterocycle. Preferably the carrier is a ribose sugar as shown in structure LCM-I. In this embodiment, the monomer, e.g., a ligand-conjugated monomer is a nucleoside.
    Figure US20050119214A1-20050602-C00062
  • “B” represents a nucleobase, e.g., a naturally occurring nucleobase optionally having one or more chemical modifications, e.g., and unusual base; or a universal base.
  • As used herein, an “unusual” nucleobase can include any one of the following:
    • 2-methyladeninyl,
    • N6-methyladeninyl,
    • 2-methylthio-N-6-methyladeninyl,
    • N6-isopentenyladeninyl,
    • 2-methylthio-N-6-isopentenyladeninyl,
    • N6-(cis-hydroxyisopentenyl)adeninyl,
    • 2-methylthio-N-6-(cis-hydroxyisopentenyl)adeninyl,
    • N6-glycinylcarbamoyladeninyl,
    • N6-threonylcarbamoyladeninyl,
    • 2-methylthio-N-6-threonyl carbamoyladeninyl,
    • N6-methyl-N-6-threonylcarbamoyladeninyl,
    • N6-hydroxynorvalylcarbamoyladeninyl,
    • 2-methylthio-N-6-hydroxynorvalyl carbamoyladeninyl,
    • N6,N6-dimethyladeninyl,
    • 3-methylcytosinyl,
    • 5-methylcytosinyl,
    • 2-thiocytosinyl,
    • 5-formylcytosinyl,
      Figure US20050119214A1-20050602-C00063
    • N4-methylcytosinyl,
    • 5-hydroxymethylcytosinyl,
    • 1-methylguaninyl,
    • N2-methylguaninyl,
    • 7-methylguaninyl,
    • N2,N2-dimethylguaninyl,
      Figure US20050119214A1-20050602-C00064
      Figure US20050119214A1-20050602-C00065
      Figure US20050119214A1-20050602-C00066
    • N2,7-dimethylguaninyl,
    • N2,N2,7-trimethylguaninyl,
    • 1-methylguaninyl,
    • 7-cyano-7-deazaguaninyl,
    • 7-aminomethyl-7-deazaguaninyl,
    • pseudouracilyl,
    • dihydrouracilyl,
    • 5-methyluracilyl,
    • 1-methylpseudouracilyl,
    • 2-thiouracilyl,
    • 4-thiouracilyl,
    • 2-thiothyminyl
    • 5-methyl-2-thiouracilyl,
    • 3-(3-amino-3-carboxypropyl)uracilyl,
    • 5-hydroxyuracilyl,
    • 5-methoxyuracilyl,
    • uracilyl 5-oxyacetic acid,
    • uracilyl 5-oxyacetic acid methyl ester,
    • 5-(carboxyhydroxymethyl)uracilyl,
    • 5-(carboxyhydroxymethyl)uracilyl methyl ester,
    • 5-methoxycarbonylmethyluracilyl,
    • 5-methoxycarbonylmethyl-2-thiouracilyl,
    • 5-aminomethyl-2-thiouracilyl,
    • 5-methylaminomethyluracilyl,
    • 5-methylaminomethyl-2-thiouracilyl,
    • 5-methylaminomethyl-2-selenouracilyl,
    • 5-carbamoylmethyluracilyl,
    • 5-carboxymethylaminomethyluracilyl,
    • 5-carboxymethylaminomethyl-2-thiouracilyl,
    • 3-methyluracilyl,
    • 1-methyl-3-(3-amino-3-carboxypropyl) pseudouracilyl,
    • 5-carboxymethyluracilyl,
    • 5-methyldihydrouracilyl, or
    • 3-methylpseudouracilyl.
  • A universal base can form base pairs with each of the natural DNA/RNA bases, exhibiting relatively little discrimination between them. In general, the universal bases are non-hydrogen bonding, hydrophobic, aromatic moieties which can stabilize e.g., duplex RNA or RNA-like molecules, via stacking interactions. A universal base can also include hydrogen bonding substituents. As used herein, a “universal base” can include anthracenes, pyrenes or any one of the following:
    Figure US20050119214A1-20050602-C00067
    Figure US20050119214A1-20050602-C00068
  • In some embodiments, B can form part of a tether that connects a ligand to the carrier. For example, the tether can be B—CH=CH—C(O)NH—(CH2)5—NHC(O)-LIGAND. In a preferred embodiment, the double bond is trans, and the ligand is a substituted or unsubstituted cholesterolyl radical (e.g., attached through the D-ring side chain or the C-3 hydroxyl); an aralkyl moiety having at least one sterogenic center and at least one substituent on the aryl portion of the aralkyl group; or a nucleobase. In certain embodiments, B, in the tether described above, is uracilyl or a universal base, e.g., an aryl moiety, e.g., phenyl, optionally having additional substituents, e.g., one or more fluoro groups. B can be substituted at any atom with the remainder of the tether.
  • X2 can include “oxy” or “deoxy” substituents in place of the 2′-OH; or be a ligand or a tethered ligand.
  • Examples of “oxy”-substituents include alkoxy or aryloxy (OR, e.g., R=H, alkyl, cycloalkyl, aryl, aralkyl, heteroaryl, sugar, or protecting group); polyethyleneglycols (PEG), O(CH2CH2O)nCH2CH2OR; “locked” nucleic acids (LNA) in which the 2′ hydroxyl is connected, e.g., by a methylene bridge, to the 4′ carbon of the same ribose sugar; O—PROTECTED AMINE (AMINE=NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino, ethylene diamine, polyamino) and aminoalkoxy, O(CH2)nPROTECTED AMINE, (e.g., AMINE=NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino, ethylene diamine, polyamino), and orthoester. Amine protecting groups can include formyl, amido, benzyl, allyl, etc.
  • Preferred orthoesters have the general formula J. The groups R31 and R32 may be the same or different and can be any combination of the groups listed in FIG. 2B. A preferred orthoester is the “ACE” group, shown below as structure K.
    Figure US20050119214A1-20050602-C00069
  • “Deoxy” substituents include hydrogen (i.e. deoxyribose sugars); halo (e.g., fluoro); protected amino (e.g. NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid in which all amino are protected); fully protected polyamino (e.g., NH(CH2CH2NH)nCH2CH2-AMINE, wherein AMINE=NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino and all amino groups are protected), —NHC(O)R (R=alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar), cyano; alkyl-thio-alkyl; thioalkoxy; and alkyl, cycloalkyl, aryl, alkenyl and alkynyl, which may be optionally substituted with e.g., a protected amino functionality. Preferred substitutents are 2′-methoxyethyl, 2′-OCH3,2′-O-allyl, 2′-C— allyl, and 2′-fluoro.
  • X3 is as described for OFG2 above.
  • PG can be a triarylmethyl group (e.g., a dimethoxytrityl group) or Si(X5′)(X5″)(X5′″) in which (X5′), (X5″), and (X5′″) are as described elsewhere.
  • Sugar Replacement-Based Monomers, e.g., Ligand-Conjugated Monomers (Cyclic)
  • Cyclic sugar replacement-based monomers, e.g., sugar replacement-based ligand-conjugated monomers, are also referred to herein as ribose replacement monomer subunit (RRMS) monomer compounds. Preferred carriers have the general formula (LCM-2) provided below (In that structure preferred backbone attachment points can be chosen from R1 or R2; R3 or R4; or R9 and R10 if Y is CR9R10 (two positions are chosen to give two backbone attachment points, e.g., R1 and R4, or R4 and R9)). Preferred tethering attachment points include R7; R5 or R6 when X is CH2. The carriers are described below as an entity, which can be incorporated into a strand. Thus, it is understood that the structures also encompass the situations wherein one (in the case of a terminal position) or two (in the case of an internal position) of the attachment points, e.g., R1 or R2; R3 or R4; or R9 or R10 (when Y is CR9R10), is connected to the phosphate, or modified phosphate, e.g., sulfur containing, backbone. E.g., one of the above-named R groups can be —CH2—, wherein one bond is connected to the carrier and one to a backbone atom, e.g., a linking oxygen or a central phosphorus atom.)
    Figure US20050119214A1-20050602-C00070
      • in which,
      • X is N(CO)R7, NR7 or CH2;
      • Y is NR8, O, S, CR9R10;
      • Z is CR11R12 or absent;
      • Each of R1, R2, R3, R4, R9, and R10 is, independently, H, ORa, or (CH2)nORb provided that at least two of R1, R2, R3, R4, R9, and R10 are ORa and/or (CH2)nORb;
      • Each of R5, R6, R11, and R12 is, independently, a ligand, H, C1-C6 alkyl optionally substituted with 1-3 R13, or C(O)NHR7; or R5 and R11 together are C3-C8 cycloalkyl optionally substituted with R14;
      • R7 can be a ligand, e.g., R7 can be Rd, or R7 can be a ligand tethered indirectly to the carrier, e.g., through a tethering moiety, e.g., C1-C20 alkyl substituted with NRcRd; or C1-C20 alkyl substituted with NHC(O)Rd;
      • R8 is H or C1-C6 alkyl;
      • R13 is hydroxy, C1-C4 alkoxy, or halo;
      • R14 is NRcR7;
      • R15 is C1-C6 alkyl optionally substituted with cyano, or C2-C6 alkenyl;
      • R16 is C1-C10 alkyl;
      • R17 is a liquid or solid phase support reagent;
      • L is —C(O)(CH2)qC(O)—, or —C(O)(CH2)qS—;
      • Ra is a protecting group, e.g., CAr3; (e.g., a dimethoxytrityl group) or Si(X5′)(X5″)(X5′″) in which (X5′), (X5″), and (X5′″) are as described elsewhere.
      • Rb is P(O)(O)H, P(OR15)N(R6)2 or L-R17;
      • Rc is H or C1-C6 alkyl;
      • Rd is H or a ligand;
      • Each Ar is, independently, C6-C10 aryl optionally substituted with C1-C4 alkoxy;
      • n is 1-4; and q is 0-4.
  • Exemplary carriers include those in which, e.g., X is N(CO)R7 or NR7, Y is CR9R10, and Z is absent; or X is N(CO)R7 or NR7, Y is CR9R10, and Z is CR11R12; or X is N(CO)R7 or NR7, Y is NR8, and Z is CR11R12; or X is N(CO)R7 or NR7, Y is O, and Z is CR11R12; or X is CH2; Y is CR9R10; Z is CR11R12, and R5 and R11 together form C6 cycloalkyl (H, z=2), or the indane ring system, e.g., X is CH2; Y is CR9R10; Z is CR11R12, and R5 and R11 together form C5 cycloalkyl (H, z=1).
  • In certain embodiments, the carrier may be based on the pyrroline ring system or the 4-hydroxyproline ring system, e.g., X is N(CO)R7 or NR7, Y is CR9R10, and Z is absent (D). OFG1 is preferably attached to a primary carbon, e.g., an exocyclic alkylene
    Figure US20050119214A1-20050602-C00071

    group, e.g., a methylene group, connected to one of the carbons in the five-membered ring (—CH2OFG1 in D). OFG2 is preferably attached directly to one of the carbons in the five-membered ring (—OFG2 in D). For the pyrroline-based carriers, —CH2OFG1 may be attached to C-2 and OFG2 may be attached to C-3; or —CH2OFG1 may be attached to C-3 and OFG2 may be attached to C-4. In certain embodiments, CH2OFG1 and OFG2 may be geminally substituted to one of the above-referenced carbons. For the 3-hydroxyproline-based carriers, —CH2OFG1 may be attached to C-2 and OFG2 may be attached to C-4. The pyrroline- and 4-hydroxyproline-based monomers may therefore contain linkages (e.g., carbon-carbon bonds) wherein bond rotation is restricted about that particular linkage, e.g. restriction resulting from the presence of a ring. Thus, CH2OFG1 and OFG2 may be cis or trans with respect to one another in any of the pairings delineated above Accordingly, all cis/trans isomers are expressly included. The monomers may also contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of the monomers are expressly included (e.g., the centers bearing CH2OFG1 and OFG2 can both have the R configuration; or both have the S configuration; or one center can have the R configuration and the other center can have the S configuration and vice versa). The tethering attachment point is preferably nitrogen. Preferred examples of carrier D include the following:
    Figure US20050119214A1-20050602-C00072
  • In certain embodiments, the carrier may be based on the piperidine ring system (E), e.g., X is N(CO)R7 or NR7, Y is CR9R10, and Z is CR11R12. OFG1 is preferably
    Figure US20050119214A1-20050602-C00073

    attached to a primary carbon, e.g., an exocyclic alkylene group, e.g., a methylene group (n=1) or ethylene group (n=2), connected to one of the carbons in the six-membered ring [—(CH2)nOFG1 in E]. OFG2 is preferably attached directly to one of the carbons in the six-membered ring (—OFG2 in E). —(CH2)nOFG1 and OFG2 may be disposed in a geminal manner on the ring, i.e., both groups may be attached to the same carbon, e.g., at C-2, C-3, or C-4. Alternatively, —(CH2)nOFG1 and OFG2 may be disposed in a vicinal manner on the ring, i.e., both groups may be attached to adjacent ring carbon atoms, e.g.,
  • —(CH2)nOFG1 may be attached to C-2 and OFG2 may be attached to C-3; —(CH2)nOFG1 may be attached to C-3 and OFG2 may be attached to C-2; —(CH2)nOFG1 may be attached to C-3 and OFG2 may be attached to C-4; or —(CH2)nOFG1 may be attached to C-4 and OFG2 may be attached to C-3. The piperidine-based monomers may therefore contain linkages (e.g., carbon-carbon bonds) wherein bond rotation is restricted about that particular linkage, e.g. restriction resulting from the presence of a ring. Thus, —(CH2)nOFG1 and OFG2 may be cis or trans with respect to one another in any of the pairings delineated above. Accordingly, all cis/trans isomers are expressly included. The monomers may also contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of the monomers are expressly included (e.g., the centers bearing CH2OFG1 and OFG2 can both have the R configuration; or both have the S configuration; or one center can have the R configuration and the other center can have the S configuration and vice versa). The tethering attachment point is preferably nitrogen.
  • In certain embodiments, the carrier may be based on the piperazine ring system (F), e.g., X is N(CO)R7 or NR7, Y is NR8, and Z is CR11R12, or the morpholine ring system (G), e.g., X is N(CO)R7 or NR7, Y is O, and Z is CR11R12. OFG1 is preferably
    Figure US20050119214A1-20050602-C00074

    attached to a primary carbon, e.g., an exocyclic alkylene group, e.g., a methylene group, connected to one of the carbons in the six-membered ring (—CH2OFG1 in F or G). OFG2 is preferably attached directly to one of the carbons in the six-membered rings (—OFG2 in F or G). For both F and G, —CH2OFG1 may be attached to C-2 and OFG2 may be attached to C-3; or vice versa. In certain embodiments, CH2OFG1 and OFG2 may be geminally substituted to one of the above-referenced carbons. The piperazine- and morpholine-based monomers may therefore contain linkages (e.g., carbon-carbon bonds) wherein bond rotation is restricted about that particular linkage, e.g. restriction resulting from the presence of a ring. Thus, CH2OFG1 and OFG2 may be cis or trans with respect to one another in any of the pairings delineated above. Accordingly, all cis/trans isomers are expressly included. The monomers may also contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of the monomers are expressly included (e.g., the centers bearing CH2OFG1 and OFG2 can both have the R configuration; or both have the S configuration; or one center can have the R configuration and the other center can have the S configuration and vice versa). R′″ can be, e.g., C1-C6 alkyl, preferably CH3. The tethering attachment point is preferably nitrogen in both F and G.
  • In certain embodiments, the carrier may be based on the decalin ring system, e.g., X is CH2; Y is CR9R10; Z is CR11R12, and R5 and R11 together form C6 cycloalkyl (H, z=2), or the indane ring system, e.g., X is CH2; Y is CR9R10; Z is CR11R12, and R5 and R11 together form C5 cycloalkyl (H, z=1). OFG1 is preferably attached to a primary carbon,
    Figure US20050119214A1-20050602-C00075

    e.g., an exocyclic methylene group (n=1) or ethylene group (n=2) connected to one of C-2, C-3, C-4, or C-5 [—(CH2)nOFG1 in H]. OFG2 is preferably attached directly to one of C-2, C-3, C-4, or C-5 (—OFG2 in H). —(CH2)nOFG1 and OFG2 may be disposed in a geminal manner on the ring, i.e., both groups may be attached to the same carbon, e.g., at C-2, C-3, C-4, or C-5. Alternatively, —(CH2)nOFG1 and OFG2 may be disposed in a vicinal manner on the ring, i.e., both groups may be attached to adjacent ring carbon atoms, e.g., —(CH2)nOFG1 may be attached to C-2 and OFG2 may be attached to C-3; —(CH2)nOFG1 may be attached to C-3 and OFG2 may be attached to C-2; —(CH2)nOFG1 may be attached to C-3 and OFG2 may be attached to C-4; or —(CH2)nOFG1 may be attached to C-4 and OFG2 may be attached to C-3; —(CH2)nOFG1 may be attached to C-4 and OFG2 may be attached to C-5; or —(CH2)nOFG1 may be attached to C-5 and OFG2 may be attached to C-4. The decalin or indane-based monomers may therefore contain linkages (e.g., carbon-carbon bonds) wherein bond rotation is restricted about that particular linkage, e.g. restriction resulting from the presence of a ring. Thus, —(CH2)nOFG1 and OFG2 may be cis or trans with respect to one another in any of the pairings delineated above. Accordingly, all cis/trans isomers are expressly included. The monomers may also contain one or more asymmetric centers and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of the monomers are expressly included (e.g., the centers bearing CH2OFG1 and OFG2 can both have the R configuration; or both have the S configuration; or one center can have the R configuration and the other center can have the S configuration and vice versa). In a preferred embodiment, the substituents at C-1 and C-6 are trans with respect to one another. The tethering attachment point is preferably C-6 or C-7.
  • Other carriers may include those based on 3-hydroxyproline (J). Thus, —(CH2)nOFG1 and OFG2 may be cis or trans with respect to one another. Accordingly, all cis/trans isomers are expressly included. The monomers may also contain one or more asymmetric centers
    Figure US20050119214A1-20050602-C00076

    and thus occur as racemates and racemic mixtures, single enantiomers, individual diastereomers and diastereomeric mixtures. All such isomeric forms of the monomers are expressly included (e.g., the centers bearing CH2OFG1 and OFG2 can both have the R configuration; or both have the S configuration; or one center can have the R configuration and the other center can have the S configuration and vice versa). The tethering attachment point is preferably nitrogen.
  • Representative cyclic, sugar replacement-based carriers are shown in FIG. 3.
  • Sugar Replacement-Based Monomers (Acyclic)
  • Acyclic sugar replacement-based monomers, e.g., sugar replacement-based ligand-conjugated monomers, are also referred to herein as ribose replacement monomer subunit (RRMS) monomer compounds. Preferred acyclic carriers can have formula LCM-3 or LCM-4 below.
    Figure US20050119214A1-20050602-C00077
  • In some embodiments, each of x, y, and z can be, independently of one another, 0, 1, 2, or 3. In formula LCM-3, when y and z are different, then the tertiary carbon can have either the R or S configuration. In preferred embodiments, x is zero and y and z are each 1 in formula LCM-3 (e.g., based on serinol), and y and z are each 1 in formula LCM-3. Each of formula LCM-3 or LCM-4 below can optionally be substituted, e.g., with hydroxy, alkoxy, perhaloalkyl.
  • Tethers
  • In certain embodiments, a moiety, e.g., a ligand may be connected indirectly to the carrier via the intermediacy of an intervening tether. Tethers are connected to the carrier at a tethering attachment point (TAP) and may include any C1-C100 carbon-containing moiety, (e.g. C1-C75, C1-C50, C1-C20, C1-C10; C1, C2, C3, C4, C5, C6, C7, C8, C9, or C10), preferably having at least one nitrogen atom. In preferred embodiments, the nitrogen atom forms part of a terminal amino or amido (NHC(O)—) group on the tether, which may serve as a connection point for the ligand. Preferred tethers (underlined) include TAP-(CH 2 ) n NH—; TAP-C(O)(CH 2 ) n NH—; TAP-NR″″(CH 2 ) n NH—, TAP-C(O—(CH 2 ) n —C(O)—; TAP-C(O)—(CH 2 ) n —C(O)O—; TAP-C(O)—O—; TAP-C(O)— (CH 2 ) n —NH—C(O)—; TAP-C(O)—(CH 2 ) n ; TAP-C(O)—NH—; TAP-C(O)—; TAP-(CH 2 ) n —C(O)—; TAP-(CH 2 ) n —C(O)O—; TAP-(CH 2 ) n ; or TAP-(CH 2 ) n —NH—C(O)—; in which n is 1-20 (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) and R″″ is C1-C6 alkyl. Preferably, n is 5, 6, or 11. In other embodiments, the nitrogen may form part of a terminal oxyamino group, e.g., —ONH2, or hydrazino group, —NHNH2. The tether may optionally be substituted, e.g., with hydroxy, alkoxy, perhaloalkyl, and/or optionally inserted with one or more additional heteroatoms, e.g., N, O, or S. Preferred tethered ligands may include, e.g., TAP-(CH 2 ) n NH(LIGAND); TAP-C(O)(CH 2 ) n NH(LIGAND); TAP-NR″″(CH 2 ) n NH(LIGAND); TAP-(CH 2 ) n ONH(LIGAND); TAP-C(O)(CH 2 ) n ONH(LIGAND); TAP-NR″″(CH 2 ) n ONH(LIGAND); TAP-(CH 2 ) n NHNH 2 (LIGAND), TAP-C(O)(CH 2 ) n NHNH 2 (LIGAND); TAP-NR″″(CH 2 ) n —NHNH 2 (LIGAND); TAP-C(O)—(CH 2 ) n —C(O)(LIGAND); TAP-C(O)—(CH 2 ) n —C(O)O(LIGAND); TAP-C(O)—O(LIGAND); TAP-C(O)—(CH 2 ) n —NH—C(O)(LIGAND); TAP-C(O)—(CH 2 ) n (LIGAND); TAP-C(O)—NH(LIGAND); TAP-C(O)(LIGAND); TAP-(CH 2 ) n —C(O) (LIGAND); TAP-(CH 2 ) n —C(O)O(LIGAND); TAP-(CH 2 ) n (LIGAND); or TAP-(CH 2 ) n —NH—C(O)(LIGAND). In some embodiments, amino terminated tethers (e.g., NH2, ONH2, NH2NH2) can form an imino bond (i.e., C═N) with the ligand. In some embodiments, amino terminated tethers (e.g., NH2, ONH2, NH2NH2) can acylated, e.g., with C(O)CF3.
  • In some embodiments, the tether can terminate with a mercapto group (i.e., SH) or an olefin (e.g., CH=CH2). For example, the tether can be TAP-(CH 2 ) n —SH, TAP-C(O)(CH 2 ) n SH, TAP-(CH 2 ) n —(CH=CH 2 ), or TAP-C(O)(CH 2 ) n (CH=CH 2 ), in which n can be as described elsewhere. The tether may optionally be substituted, e.g., with hydroxy, alkoxy, perhaloalkyl, and/or optionally inserted with one or more additional heteroatoms, e.g., N, O, or S. The double bond can be cis or trans or E or Z.
  • In other embodiments the tether may include an electrophilic moiety, preferably at the terminal position of the tether. Preferred electrophilic moieties include, e.g., an aldehyde, alkyl halide, mesylate, tosylate, nosylate, or brosylate, or an activated carboxylic acid ester, e.g. an NHS ester, or a pentafluorophenyl ester. Preferred tethers (underlined) include TAP-(CH 2 ) n CHO; TAP-C(O)(CH 2 ) n CHO; or TAP-NR″″(CH 2 ) n CHO, in which n is 1-6 and R″″ is C1-C6 alkyl; or TAP-(CH 2 ) n C(O)ONHS; TAP-C(O)(CH 2 ) n C(O)ONHS; or TAP-NR″″(CH 2 ) n C(O)ONHS, in which n is 1-6 and R″″ is C1-C6 alkyl; TAP-(CH 2 ) n C(O)OC 6 F 5 ; TAP-C(O)(CH 2 ) n C(O) OC 6 F 5 ; or TAP-NR″″(CH 2 ) n C(O)OC 6 F 5 , in which n is 1-11 and R″″ is C1-C6 alkyl; or —(CH 2 ) n CH 2 LG; TAP-C(O)(CH 2 ) n CH 2 LG; or TAP-NR″″(CH 2 ) n CH 2 LG, in which n can be as described elsewhere and R″″ is C1-C6 alkyl (LG can be a leaving group, e.g., halide, mesylate, tosylate, nosylate, brosylate). Tethering can be carried out by coupling a nucleophilic group of a ligand, e.g., a thiol or amino group with an electrophilic group on the tether.
  • In other embodiments, it can be desirable for the monomer to include a phthalimido group (K) at the terminal position of the tether.
    Figure US20050119214A1-20050602-C00078
  • In other embodiments, other protected amino groups can be at the terminal position of the tether, e.g., alloc, monomethoxy trityl (MMT), itrifluoroacetyl, Fmoc, or aryl sulfonyl (e.g., the aryl portion can be ortho-nitrophenyl or ortho, para-dinitrophenyl).
  • Any of the tethers described herein may further include one or more additional linking groups, e.g., —O—(CH2)n—, —(CH2)n—SS—, —(CH2)n—, or —(CH=CH)—.
  • Tethered Ligands
  • A wide variety of entities, e.g., ligands, can be tethered to an iRNA agent, e.g., to the carrier of a ligand-conjugated monomer subunit. Examples are described below in the context of a ligand-conjugated monomer subunit but that is only preferred, entities can be coupled at other points to an iRNA agent.
  • Preferred moieties are ligands, which are coupled, preferably covalently, either directly or indirectly via an intervening tether, to the carrier. In preferred embodiments, the ligand is attached to the carrier via an intervening tether. As discussed above, the ligand or tethered ligand may be present on the ligand-conjugated monomer \ when the ligand-conjugated monomer is incorporated into the growing strand. In some embodiments, the ligand may be incorporated into a “precursor” ligand-conjugated monomer subunit after a “precursor” ligand-conjugated monomer subunit has been incorporated into the growing strand. For example, a monomer having, e.g., an amino-terminated tether, e.g., TAP-(CH2)nNH2 may be incorporated into a growing sense or antisense strand. In a subsequent operation, i.e., after incorporation of the precursor monomer subunit into the strand, a ligand having an electrophilic group, e.g., a pentafluorophenyl ester or aldehyde group, can subsequently be attached to the precursor ligand-conjugated monomer by coupling the electrophilic group of the ligand with the terminal nucleophilic group of the precursor ligand-conjugated monomer subunit tether.
  • In preferred embodiments, a ligand alters the distribution, targeting or lifetime of an iRNA agent into which it is incorporated. In preferred embodiments a ligand provides an enhanced affinity for a selected target, e.g, molecule, cell or cell type, compartment, e.g., a cellular or organ compartment, tissue, organ or region of the body, as, e.g., compared to a species absent such a ligand.
  • Preferred ligands can improve transport, hybridization, and specificity properties and may also improve nuclease resistance of the resultant natural or modified oligoribonucleotide, or a polymeric molecule comprising any combination of monomers described herein and/or natural or modified ribonucleotides.
  • Ligands in general can include therapeutic modifiers, e.g., for enhancing uptake; diagnostic compounds or reporter groups e.g., for monitoring distribution; cross-linking agents; nuclease-resistance conferring moieties; and natural or unusual nucleobases. General examples include lipophiles, lipids, steroids (e.g., cholesterol, uvaol, hecigenin, diosgenin), terpenes (e.g., triterpenes, e.g., sarsasapogenin, Friedelin, epifriedelanol derivatized lithocholic acid), vitamins (e.g., folic acid, vitamin A, vitamin E, biotin, pyridoxal), carbohydrates, proteins, protein binding agents, integrin targeting molecules, polycationics, peptides, polyamines, and peptide mimics.
  • The ligand can be a peptide or peptidomimetic. A peptidomimetic (also referred to herein as an oligopeptidomimetic) is a molecule capable of folding into a defined three-dimensional structure similar to a natural peptide. The attachment of peptide and peptidomimetics to iRNA agents can affect pharmacokinetic distribution of the iRNA, such as by enhancing cellular recognition and absorption. The peptide or peptidomimetic moiety can be about 5-50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
  • In some embodiments, the ligand can be one of the following triterpenes:
    Figure US20050119214A1-20050602-C00079
  • In some embodiments, the ligand can be substituted or unsubstituted cholesterol, or a stereoisomer thereof or one of the following steroids:
    Figure US20050119214A1-20050602-C00080
  • iRNA Agent Structure
  • The monomers described herein can be used to make oligonucleotides which are useful as iRNA agents, e.g., RNA molecules, (double-stranded; single-stranded) that mediate RNAi, e.g., with respect to an endogenous gene of a subject or to a gene of a pathogen. In most cases the iRNA agent will incorporate momomers described herein together with naturally occuring nucleosides or nucleotides or with other modified nucleosides or nucleotides. The modified monomers can be present at any position in the iRNA agent, e.g., at the terminii or in the middle region of an iRNA agent or in a duplex region or in an unpaired region. In a preferred embodiment iRNA agent can have any architecture, e.g., architecture described herein. E.g., it can be incorporated into an iRNA agent having an overhang structure, a hairpin or other single strand structure or a two-strand structure, as described herein.
  • An “RNA agent” as used herein, is an unmodified RNA, modified RNA, or nucleoside surrogate, all of which are defined herein (see, e.g., the section below entitled RNA Agents). While numerous modified RNAs and nucleoside surrogates are described, preferred examples include those which have greater resistance to nuclease degradation than do unmodified RNAs. Preferred examples include those which have a 2′ sugar modification, a modification in a single strand overhang, preferably a 3′ single strand overhang, or, particularly if single stranded, a 5′ modification which includes one or more phosphate groups or one or more analogs of a phosphate group.
  • An “iRNA agent” as used herein, is an RNA agent which can, or which can be cleaved into an RNA agent which can, down regulate the expression of a target gene, preferably an endogenous or pathogen target RNA. While not wishing to be bound by theory, an iRNA agent may act by one or more of a number of mechanisms, including post-transcriptional cleavage of a target mRNA sometimes referred to in the art as RNAi, or pre-transcriptional or pre-translational mechanisms. An iRNA agent can include a single strand or can include more than one strands, e.g., it can be a double stranded iRNA agent. If the iRNA agent is a single strand it is particularly preferred that it include a 5′ modification which includes one or more phosphate groups or one or more analogs of a phosphate group.
  • The RRMS-containing iRNA agent should include a region of sufficient homology to the target gene, and be of sufficient length in terms of nucleotides, such that the iRNA agent, or a fragment thereof, can mediate down regulation of the target gene. (For ease of exposition the term nucleotide or ribonucleotide is sometimes used herein in reference to one or more monomeric subunits of an RNA agent. It will be understood herein that the usage of the term “ribonucleotide” or “nucleotide”, herein can, in the case of a modified RNA or nucleotide surrogate, also refer to a modified nucleotide, or surrogate replacement moiety at one or more positions.) Thus, the iRNA agent is or includes a region which is at least partially, and in some embodiments fully, complementary to the target RNA. It is not necessary that there be perfect complementarity between the iRNA agent and the target, but the correspondence must be sufficient to enable the iRNA agent, or a cleavage product thereof, to direct sequence specific silencing, e.g., by RNAi cleavage of the target RNA, e.g., mRNA.
  • As discussed elsewhere herein, an iRNA agent will often be modified or include nucleoside surrogates in addition to the ribose replacement modification subunit (RRMS). Single stranded regions of an iRNA agent will often be modified or include nucleoside surrogates, e.g., the unpaired region or regions of a hairpin structure, e.g., a region which links two complementary regions, can have modifications or nucleoside surrogates. Modification to stabilize one or more 3′- or 5′-terminus of an iRNA agent, e.g., against exonucleases, or to favor the antisense sRNA agent to enter into RISC are also favored. Modifications can include C3 (or C6, C7, C12) amino linkers, thiol linkers, carboxyl linkers, non-nucleotidic spacers (C3, C6, C9, C12, abasic, triethylene glycol, hexaethylene glycol), special biotin or fluorescein reagents that come as phosphoramidites and that have another DMT-protected hydroxyl group, allowing multiple couplings during RNA synthesis.
  • In addition to homology to target RNA and the ability to down regulate a target gene, an iRNA agent will preferably have one or more of the following properties:
      • (1) it will be of the Formula 1, 2, 3, or 4 set out in the RNA Agent section below;
      • (2) if single stranded it will have a 5′ modification which includes one or more phosphate groups or one or more analogs of a phosphate group;
      • (3) it will, despite modifications, even to a very large number, or all of the nucleosides, have an antisense strand that can present bases (or modified bases) in the proper three dimensional framework so as to be able to form correct base pairing and form a duplex structure with a homologous target RNA which is sufficient to allow down regulation of the target, e.g., by cleavage of the target RNA;
      • (4) it will, despite modifications, even to a very large number, or all of the nucleosides, still have “RNA-like” properties, i.e., it will possess the overall structural, chemical and physical properties of an RNA molecule, even though not exclusively, or even partly, of ribonucleotide-based content. For example, an iRNA agent can contain, e.g., a sense and/or an antisense strand in which all of the nucleotide sugars contain e.g., 2′ fluoro in place of 2′ hydroxyl. This deoxyribonucleotide-containing agent can still be expected to exhibit RNA-like properties. While not wishing to be bound by theory, the electronegative fluorine prefers an axial orientation when attached to the C2′ position of ribose. This spatial preference of fluorine can, in turn, force the sugars to adopt a C3′-endo pucker. This is the same puckering mode as observed in RNA molecules and gives rise to the RNA-characteristic A-family-type helix. Further, since fluorine is a good hydrogen bond acceptor, it can participate in the same hydrogen bonding interactions with water molecules that are known to stabilize RNA structures. (Generally, it is preferred that a modified moiety at the 2′ sugar position will be able to enter into H-bonding which is more characteristic of the OH moiety of a ribonucleotide than the H moiety of a deoxyribonucleotide. A preferred iRNA agent will: exhibit a C3′-endo pucker in all, or at least 50, 75, 80, 85, 90, or 95% of its sugars; exhibit a C3′-endo pucker in a sufficient amount of its sugars that it can give rise to a the RNA-characteristic A-family-type helix; will have no more than 20, 10, 5, 4, 3, 2, or 1 sugar which is not a C3′-endo pucker structure. These limitations are particularly preferably in the antisense strand;
      • (4) regardless of the nature of the modification, and even though the RNA agent can contain deoxynucleotides or modified deoxynucleotides, particularly in overhang or other single strand regions, it is preferred that DNA molecules, or any molecule in which more than 50, 60, or 70% of the nucleotides in the molecule, or more than 50, 60, or 70% of the nucleotides in a duplexed region are deoxyribonucleotides, or modified deoxyribonucleotides which are deoxy at the 2′ position, are excluded from the definition of RNA agent.
  • A “single strand iRNA agent” as used herein, is an iRNA agent which is made up of a single molecule. It may include a duplexed region, formed by intra-strand pairing, e.g., it may be, or include, a hairpin or pan-handle structure. Single strand iRNA agents are preferably antisense with regard to the target molecule. In preferred embodiments single strand iRNA agents are 5′ phosphorylated or include a phosphoryl analog at the 5′ prime terminus. 5′-phosphate modifications include those which are compatible with RISC mediated gene silencing. Suitable modifications include: 5′-monophosphate ((HO)2(O)P—O-5′); 5′-diphosphate ((HO)2(O)P—O—P(HO)(O)—O-5′); 5′-triphosphate ((HO)2(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-guanosine cap (7-methylated or non-methylated) (7m-G-O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-adenosine cap (Appp), and any modified or unmodified nucleotide cap structure (N—O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-monothiophosphate (phosphorothioate; (HO)2(S)P—O-5′); 5′-monodithiophosphate (phosphorodithioate; (HO)(HS)(S)P—O-5′), 5′-phosphorothiolate ((HO)2(O)P—S-5′); any additional combination of oxygen/sulfur replaced monophosphate, diphosphate and triphosphates (e.g. 5′-alpha-thiotriphosphate, 5′-gamma-thiotriphosphate, etc.), 5′-phosphoramidates ((HO)2(O)P—NH-5′, (HO)(NH2)(O)P—O-5′), 5′-alkylphosphonates (R=alkyl=methyl, ethyl, isopropyl, propyl, etc., e.g. RP(OH)(O)—O-5′—, (OH)2(O)P-5′-CH2—), 5′-alkyletherphosphonates (R=alkylether=methoxymethyl (MeOCH2—), ethoxymethyl, etc., e.g. RP(OH)(O)—O-5′-). (These modifications can also be used with the antisense strand of a double stranded iRNA.)
  • It may be desirable to modify one or both of the antisense and sense strands of a double strand iRNA agent. In some cases they will have the same modification or the same class of modification but in other cases the sense and antisense strand will have different modifications, e.g., in some cases it is desirable to modify only the sense strand. It may be desirable to modify only the sense strand, e.g., to inactivate it, e.g., the sense strand can be modified in order to inactivate the sense strand and prevent formation of an active sRNA/protein or RISC. This can be accomplished by a modification which prevents 5′-phosphorylation of the sense strand, e.g., by modification with a 5′-O-methyl ribonucleotide (see Nykanen et al., (2001) ATP requirements and small interfering RNA structure in the RNA interference pathway. Cell 107, 309-321.) Other modifications which prevent phosphorylation can also be used, e.g., simply substituting the 5′-OH by H rather than O-Me. Alternatively, a large bulky group may be added to the 5′-phosphate turning it into a phosphodiester linkage, though this may be less desirable as phosphodiesterases can cleave such a linkage and release a functional sRNA 5′-end. Antisense strand modifications include 5′ phosphorylation as well as any of the other 5′ modifications discussed herein, particularly the 5′ modifications discussed above in the section on single stranded iRNA molecules.
  • It is preferred that the sense and antisense strands be chosen such that the ds iRNA agent includes a single strand or unpaired region at one or both ends of the molecule. Thus, a ds iRNA agent contains sense and antisense strands, preferable paired to contain an overhang, e.g., one or two 5′ or 3′ overhangs but preferably a 3′ overhang of 2-3 nucleotides. Most embodiments will have a 3′ overhang. Preferred sRNA agents will have single-stranded overhangs, preferably 3′ overhangs, of 1 or preferably 2 or 3 nucleotides in length at each end. The overhangs can be the result of one strand being longer than the other, or the result of two strands of the same length being staggered. 5′ ends are preferably phosphorylated.
  • Preferred lengths for the duplexed region is between 15 and 30, most preferably 18, 19, 20, 21, 22, and 23 nucleotides in length, e.g., in the sRNA agent range discussed above. sRNA agents can resemble in length and structure the natural Dicer processed products from long dsRNAs. Embodiments in which the two strands of the sRNA agent are linked, e.g., covalently linked are also included. Hairpin, or other single strand structures which provide the required double stranded region, and preferably a 3′ overhang are also within the invention.
  • As nucleic acids are polymers of subunits or monomers, many of the modifications described below occur at a position which is repeated within a nucleic acid, e.g., a modification of a base, or a phosphate moiety, or the a non-linking 0 of a phosphate moiety. In some cases the modification will occur at all of the subject positions in the nucleic acid but in many, and infact in most cases it will not. By way of example, a modification may only occur at a 3′ or 5′ terminal position, may only occur in a terminal regions, e.g. at a position on a terminal nucleotide or in the last 2, 3, 4, 5, or 10 nucleotides of a strand. A modification may occur in a double strand region, a single strand region, or in both. A modification may occur only in the double strand region of an RNA or may only occur in a single strand region of an RNA. E.g., a phosphorothioate modification at a non-linking O position may only occur at one or both termini, may only occur in a terminal regions, e.g., at a position on a terminal nucleotide or in the last 2, 3, 4, 5, or 10 nucleotides of a strand, or may occur in double strand and single strand regions, particularly at termini. The 5′ end or ends can be phosphorylated.
  • In some embodiments it is particularly preferred, e.g., to enhance stability, to include particular bases in overhangs, or to include modified nucleotides or nucleotide surrogates, in single strand overhangs, e.g., in a 5′ or 3′ overhang, or in both. E.g., it can be desirable to include purine nucleotides in overhangs. In some embodiments all or some of the bases in a 3′ or 5′ overhang will be modified, e.g., with a modification described herein. Modifications can include, e.g., the use of modifications at the 2′ OH group of the ribose sugar, e.g., the use of deoxyribonucleotides, e.g., deoxythymidine, instead of ribonucleotides, and modifications in the phosphate group, e.g., phosphothioate modifications. Overhangs need not be homologous with the target sequence.
  • Modifications and nucleotide surrogates are discussed below.
    Figure US20050119214A1-20050602-C00081
  • The scaffold presented above in Formula 1 represents a portion of a ribonucleic acid. The basic components are the ribose sugar, the base, the terminal phosphates, and phosphate internucleotide linkers. Where the bases are naturally occurring bases, e.g., adenine, uracil, guanine or cytosine, the sugars are the unmodified 2′ hydroxyl ribose sugar (as depicted) and W, X, Y, and Z are all O, Formula 1 represents a naturally occurring unmodified oligoribonucleotide.
  • Unmodified oligoribonucleotides may be less than optimal in some applications, e.g., unmodified oligoribonucleotides can be prone to degradation by e.g., cellular nucleases. Nucleases can hydrolyze nucleic acid phosphodiester bonds. However, chemical modifications to one or more of the above RNA components can confer improved properties, and, e.g., can render oligoribonucleotides more stable to nucleases. Umodified oligoribonucleotides may also be less than optimal in terms of offering tethering points for attaching ligands or other moieties to an iRNA agent.
  • Modified nucleic acids and nucleotide surrogates can include one or more of:
      • (i) alteration, e.g., replacement, of one or both of the non-linking (X and Y) phosphate oxygens and/or of one or more of the linking (W and Z) phosphate oxygens (When the phosphate is in the terminal position, one of the positions W or Z will not link the phosphate to an additional element in a naturally occurring ribonucleic acid. However, for simplicity of terminology, except where otherwise noted, the W position at the 5′ end of a nucleic acid and the terminal Z position at the 3′end of a nucleic acid, are within the term “linking phosphate oxygens” as used herein.);
      • (ii) alteration, e.g., replacement, of a constituent of the ribose sugar, e.g., of the 2′ hydroxyl on the ribose sugar, or wholesale replacement of the ribose sugar with a structure other than ribose, e.g., as described herein;
      • (iii) wholesale replacement of the phosphate moiety (bracket I) with “dephospho” linkers;
      • (iv) modification or replacement of a naturally occurring base;
      • (v) replacement or modification of the ribose-phosphate backbone (bracket II);
      • (vi) modification of the 3′ end or 5′ end of the RNA, e.g., removal, modification or replacement of a terminal phosphate group or conjugation of a moiety, e.g. a fluorescently labeled moiety, to either the 3′ or 5′ end of RNA.
  • The terms replacement, modification, alteration, and the like, as used in this context, do not imply any process limitation, e.g., modification does not mean that one must start with a reference or naturally occurring ribonucleic acid and modify it to produce a modified ribonucleic acid bur rather modified simply indicates a difference from a naturally occurring molecule.
  • It is understood that the actual electronic structure of some chemical entities cannot be adequately represented by only one canonical form (i.e. Lewis structure). While not wishing to be bound by theory, the actual structure can instead be some hybrid or weighted average of two or more canonical forms, known collectively as resonance forms or structures. Resonance structures are not discrete chemical entities and exist only on paper. They differ from one another only in the placement or “localization” of the bonding and nonbonding electrons for a particular chemical entity. It can be possible for one resonance structure to contribute to a greater extent to the hybrid than the others. Thus, the written and graphical descriptions of the embodiments of the present invention are made in terms of what the art recognizes as the predominant resonance form for a particular species. For example, any phosphoroamidate (replacement of a nonlinking oxygen with nitrogen) would be represented by X=O and Y=N in the above figure.
  • Specific modifications are discussed in more detail below.
  • The Phosphate Group
  • The phosphate group is a negatively charged species. The charge is distributed equally over the two non-linking oxygen atoms (i.e., X and Y in Formula 1 above). However, the phosphate group can be modified by replacing one of the oxygens with a different substituent. One result of this modification to RNA phosphate backbones can be increased resistance of the oligoribonucleotide to nucleolytic breakdown. Thus while not wishing to be bound by theory, it can be desirable in some embodiments to introduce alterations which result in either an uncharged linker or a charged linker with unsymmetrical charge distribution.
  • Examples of modified phosphate groups include phosphorothioate, phosphoroselenates, borano phosphates, borano phosphate esters, hydrogen phosphonates, phosphoroamidates, alkyl or aryl phosphonates and phosphotriesters. Phosphorodithioates have both non-linking oxygens replaced by sulfur. Unlike the situation where only one of X or Y is altered, the phosphorus center in the phosphorodithioates is achiral which precludes the formation of oligoribonucleotides diastereomers. Diastereomer formation can result in a preparation in which the individual diastereomers exhibit varying resistance to nucleases. Further, the hybridization affinity of RNA containing chiral phosphate groups can be lower relative to the corresponding unmodified RNA species. Thus, while not wishing to be bound by theory, modifications to both X and Y which eliminate the chiral center, e.g. phosphorodithioate formation, may be desirable in that they cannot produce diastereomer mixtures. Thus, X can be any one of S, Se, B, C, H, N, or OR (R is alkyl or aryl). Thus Y can be any one of S, Se, B, C, H, N, or OR (R is alkyl or aryl). Replacement of X and/or Y with sulfur is preferred.
  • The phosphate linker can also be modified by replacement of a linking oxygen (i.e., W or Z in Formula 1) with nitrogen (bridged phosphoroamidates), sulfur (bridged phosphorothioates) and carbon (bridged methylenephosphonates). The replacement can occur at a terminal oxygen (position W (3′) or position Z (5′). Replacement of W with carbon or Z with nitrogen is preferred.
  • Candidate agents can be evaluated for suitability as described below.
  • The Sugar Group
  • A modified RNA can include modification of all or some of the sugar groups of the ribonucleic acid. E.g., the 2′ hydroxyl group (OH) can be modified or replaced with a number of different “oxy” or “deoxy” substituents. While not being bound by theory, enhanced stability is expected since the hydroxyl can no longer be deprotonated to form a 2′ alkoxide ion. The 2′ alkoxide can catalyze degradation by intramolecular nucleophilic attack on the linker phosphorus atom. Again, while not wishing to be bound by theory, it can be desirable to some embodiments to introduce alterations in which alkoxide formation at the 2′ position is not possible.
  • Examples of “oxy”-2′ hydroxyl group modifications include alkoxy or aryloxy (OR, e.g., R=H, alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar); polyethyleneglycols (PEG), O(CH2CH2O)nCH2CH2OR; “locked” nucleic acids (LNA) in which the 2′ hydroxyl is connected, e.g., by a methylene bridge, to the 4′ carbon of the same ribose sugar; O-AMINE (AMINE=NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino, ethylene diamine, polyamino) and aminoalkoxy, O(CH2)nAMINE, (e.g., AMINE=NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino, ethylene diamine, polyamino). It is noteworthy that oligonucleotides containing only the methoxyethyl group (MOE), (OCH2CH2OCH3, a PEG derivative), exhibit nuclease stabilities comparable to those modified with the robust phosphorothioate modification.
  • “Deoxy” modifications include hydrogen (i.e. deoxyribose sugars, which are of particular relevance to the overhang portions of partially ds RNA); halo (e.g., fluoro); amino (e.g. NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid); NH(CH2CH2NH)nCH2CH2-AMINE (AMINE=NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino), —NHC(O)R(R=alkyl, cycloalkyl, aryl, aralkyl, heteroaryl or sugar), cyano; mercapto; alkyl-thio-alkyl; thioalkoxy; and alkyl, cycloalkyl, aryl, alkenyl and alkynyl, which may be optionally substituted with e.g., an amino functionality. Preferred substitutents are 2′-methoxyethyl, 2′-OCH3,2′-O-allyl, 2′-C— allyl, and 2′-fluoro.
  • The sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose. Thus, a modified RNA can include nucleotides containing e.g., arabinose, as the sugar.
  • Modified RNAs can also include “abasic” sugars, which lack a nucleobase at C-1′. These abasic sugars can also be further contain modifications at one or more of the constituent sugar atoms.
  • To maximize nuclease resistance, the 2′ modifications can be used in combination with one or more phosphate linker modifications (e.g., phosphorothioate). The so-called “chimeric” oligonucleotides are those that contain two or more different modifications.
  • The modificaton can also entail the wholesale replacement of a ribose structure with another entity at one or more sites in the iRNA agent.
  • Candidate modifications can be evaluated as described below.
  • Replacement of the Phosphate Group
  • The phosphate group can be replaced by non-phosphorus containing connectors (cf. Bracket I in Formula 1 above). While not wishing to be bound by theory, it is believed that since the charged phosphodiester group is the reaction center in nucleolytic degradation, its replacement with neutral structural mimics should impart enhanced nuclease stability. Again, while not wishing to be bound by theory, it can be desirable, in some embodiment, to introduce alterations in which the charged phosphate group is replaced by a neutral moiety.
  • Examples of moieties which can replace the phosphate group include siloxane, carbonate, carboxymethyl, carbamate, amide, thioether, ethylene oxide linker, sulfonate, sulfonamide, thioformacetal, formacetal, oxime, methyleneimino, methylenemethylimino, methylenehydrazo, methylenedimethylhydrazo and methyleneoxymethylimino. Preferred replacements include the methylenecarbonylamino and methylenemethylimino groups.
  • Candidate modifications can be evaluated as described below.
  • Replacement of Ribophosphate Backbone
  • Oligonucleotide-mimicking scaffolds can also be constructed wherein the phosphate linker and ribose sugar are replaced by nuclease resistant nucleoside or nucleotide surrogates (see Bracket II of Formula 1 above). While not wishing to be bound by theory, it is believed that the absence of a repetitively charged backbone diminishes binding to proteins that recognize polyanions (e.g. nucleases). Again, while not wishing to be bound by theory, it can be desirable in some embodiment, to introduce alterations in which the bases are tethered by a neutral surrogate backbone.
  • Examples include the mophilino, cyclobutyl, pyrrolidine and peptide nucleic acid (PNA) nucleoside surrogates. A preferred surrogate is a PNA surrogate.
  • Candidate modifications can be evaluated as described below.
  • Terminal Modifications
  • The 3′ and 5′ ends of an oligonucleotide can be modified. Such modifications can be at the 3′ end, 5′ end or both ends of the molecule. They can include modification or replacement of an entire terminal phosphate or of one or more of the atoms of the phosphate group. E.g., the 3′ and 5′ ends of an oligonucleotide can be conjugated to other functional molecular entities such as labeling moieties, e.g., fluorophores (e.g., pyrene, TAMRA, fluorescein, Cy3 or Cy5 dyes) or protecting groups (based e.g., on sulfur, silicon, boron or ester). The functional molecular entities can be attached to the sugar through a phosphate group and/or a spacer. The terminal atom of the spacer can connect to or replace the linking atom of the phosphate group or the C-3′ or C-5′ O, N, S or C group of the sugar. Alternatively, the spacer can connect to or replace the terminal atom of a nucleotide surrogate (e.g., PNAs). These spacers or linkers can include e.g., —(CH2)n—, —(CH2)nN—, —(CH2)nO—, —(CH2)nS—, O(CH2CH2O)nCH2CH2OH (e.g., n=3 or 6), abasic sugars, amide, carboxy, amine, oxyamine, oxyimine, thioether, disulfide, thiourea, sulfonamide, or morpholino, or biotin and fluorescein reagents. When a spacer/phosphate-functional molecular entity-spacer/phosphate array is interposed between two strands of iRNA agents, this array can substitute for a hairpin RNA loop in a hairpin-type RNA agent. The 3′ end can be an —OH group. While not wishing to be bound by theory, it is believed that conjugation of certain moieties can improve transport, hybridization, and specificity properties. Again, while not wishing to be bound by theory, it may be desirable to introduce terminal alterations that improve nuclease resistance. Other examples of terminal modifications include dyes, intercalating agents (e.g. acridines), cross-linkers (e.g. psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases (e.g. EDTA), lipophilic carriers (e.g., cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, bomeol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, O3-(oleoyl)lithocholic acid, O3-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine) and peptide conjugates (e.g., antennapedia peptide, Tat peptide), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG]2, polyamino, alkyl, substituted alkyl, radiolabeled markers, enzymes, haptens (e.g. biotin), transport/absorption facilitators (e.g., aspirin, vitamin E, folic acid), synthetic ribonucleases (e.g., imidazole, bisimidazole, histamine, imidazole clusters, acridine-imidazole conjugates, Eu3+complexes of tetraazamacrocycles).
  • Terminal modifications can be added for a number of reasons, including as discussed elsewhere herein to modulate activity or to modulate resistance to degradation. Terminal modifications useful for modulating activity include modification of the 5′ end with phosphate or phosphate analogs. E.g., in preferred embodiments iRNA agents, especially antisense strands, are 5′ phosphorylated or include a phosphoryl analog at the 5′ prime terminus. 5′-phosphate modifications include those which are compatible with RISC mediated gene silencing. Suitable modifications include: 5′-monophosphate ((HO)2(O)P—O-5′); 5′-diphosphate ((HO)2(O)P—O—P(HO)(O)—O-5′); 5′-triphosphate ((HO)2(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-guanosine cap (7-methylated or non-methylated) (7m-G-O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-adenosine cap (Appp), and any modified or unmodified nucleotide cap structure (N—O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′); 5′-monothiophosphate (phosphorothioate; (HO)2(S)P—O-5′); 5′-monodithiophosphate (phosphorodithioate; (HO)(HS)(S)P—O-5′), 5′-phosphorothiolate ((HO)2(O)P—S-5′); any additional combination of oxgen/sulfur replaced monophosphate, diphosphate and triphosphates (e.g. 5′-alpha-thiotriphosphate, 5′-gamma-thiotriphosphate, etc.), 5′-phosphoramidates ((HO)2(O)P—NH-5′, (HO)(NH2)(O)P—O-5′), 5′-alkylphosphonates (R=alkyl=methyl, ethyl, isopropyl, propyl, etc., e.g. RP(OH)(O)—O-5′—, (OH)2(O)P-5′-CH2—), 5′-alkyletherphosphonates (R=alkylether=methoxymethyl (MeOCH2—), ethoxymethyl, etc., e.g. RP(OH)(O)—O-5′—).
  • Terminal modifications can also be useful for monitoring distribution, and in such cases the preferred groups to be added include fluorophores, e.g., fluorscein or an Alexa dye, e.g., Alexa 488. Terminal modifications can also be useful for enhancing uptake, useful modifications for this include cholesterol. Terminal modifications can also be useful for cross-linking an RNA agent to another moiety; modifications useful for this include mitomycin C.
  • Candidate modifications can be evaluated as described below.
  • The Bases
  • Adenine, guanine, cytosine and uracil are the most common bases found in RNA. These bases can be modified or replaced to provide RNA's having improved properties. E.g., nuclease resistant oligoribonucleotides can be prepared with these bases or with synthetic and natural nucleobases (e.g., inosine, thymine, xanthine, hypoxanthine, nubularine, isoguanisine, or tubercidine) and any one of the above modifications. Alternatively, substituted or modified analogs of any of the above bases, e.g., “unusual bases” and “universal bases” described herein, can be employed. Examples include without limitation 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 5-halouracil, 5-(2-aminopropyl)uracil, 5-amino allyl uracil, 8-halo, amino, thiol, thioalkyl, hydroxyl and other 8-substituted adenines and guanines, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine, 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine, dihydrouracil, 3-deaza-5-azacytosine, 2-aminopurine, 5-alkyluracil, 7-alkylguanine, 5-alkyl cytosine, 7-deazaadenine, N6, N6-dimethyladenine, 2,6-diaminopurine, 5-amino-allyl-uracil, N3-methyluracil, substituted 1,2,4-triazoles, 2-pyridinone, 5-nitroindole, 3-nitropyrrole, 5-methoxyuracil, uracil-5-oxyacetic acid, 5-methoxycarbonylmethyluracil, 5-methyl-2-thiouracil, 5-methoxycarbonylmethyl-2-thiouracil, 5-methylaminomethyl-2-thiouracil, 3-(3-amino-3carboxypropyl)uracil, 3-methylcytosine, 5-methylcytosine, N4-acetyl cytosine, 2-thiocytosine, N6-methyladenine, N6-isopentyladenine, 2-methylthio-N6-isopentenyladenine, N-methylguanines, or O-alkylated bases. Further purines and pyrimidines include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in the Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. I., ed. John Wiley & Sons, 1990, and those disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613.
  • Generally, base changes are less preferred for promoting stability, but they can be useful for other reasons, e.g., some, e.g., 2,6-diaminopurine and 2 amino purine, are fluorescent. Modified bases can reduce target specificity. This should be taken into consideration in the design of iRNA agents.
  • Candidate modifications can be evaluated as described below.
  • Evaluation of Candidate RNA's
  • One can evaluate a candidate RNA agent, e.g., a modified RNA, for a selected property by exposing the agent or modified molecule and a control molecule to the appropriate conditions and evaluating for the presence of the selected property. For example, resistance to a degradent can be evaluated as follows. A candidate modified RNA (and preferably a control molecule, usually the unmodified form) can be exposed to degradative conditions, e.g., exposed to a milieu, which includes a degradative agent, e.g., a nuclease. E.g., one can use a biological sample, e.g., one that is similar to a milieu, which might be encountered, in therapeutic use, e.g., blood or a cellular fraction, e.g., a cell-free homogenate or disrupted cells. The candidate and control could then be evaluated for resistance to degradation by any of a number of approaches. For example, the candidate and control could be labeled, preferably prior to exposure, with, e.g., a radioactive or enzymatic label, or a fluorescent label, such as Cy3 or Cy5. Control and modified RNA's can be incubated with the degradative agent, and optionally a control, e.g., an inactivated, e.g., heat inactivated, degradative agent. A physical parameter, e.g., size, of the modified and control molecules are then determined. They can be determined by a physical method, e.g., by polyacrylamide gel electrophoresis or a sizing column, to assess whether the molecule has maintained its original length, or assessed functionally. Alternatively, Northern blot analysis can be used to assay the length of an unlabeled modified molecule.
  • A functional assay can also be used to evaluate the candidate agent. A functional assay can be applied initially or after an earlier non-functional assay, (e.g., assay for resistance to degradation) to determine if the modification alters the ability of the molecule to silence gene expression. For example, a cell, e.g., a mammalian cell, such as a mouse or human cell, can be co-transfected with a plasmid expressing a fluorescent protein, e.g., GFP, and a candidate RNA agent homologous to the transcript encoding the fluorescent protein (see, e.g., WO 00/44914). For example, a modified dsRNA homologous to the GFP mRNA can be assayed for the ability to inhibit GFP expression by monitoring for a decrease in cell fluorescence, as compared to a control cell, in which the transfection did not include the candidate dsRNA, e.g., controls with no agent added and/or controls with a non-modified RNA added. Efficacy of the candidate agent on gene expression can be assessed by comparing cell fluorescence in the presence of the modified and unmodified dsRNA agents.
  • In an alternative functional assay, a candidate dsRNA agent homologous to an endogenous mouse gene, preferably a maternally expressed gene, such as c-mos, can be injected into an immature mouse oocyte to assess the ability of the agent to inhibit gene expression in vivo (see, e.g., WO 01/36646). A phenotype of the oocyte, e.g., the ability to maintain arrest in metaphase II, can be monitored as an indicator that the agent is inhibiting expression. For example, cleavage of c-mos mRNA by a dsRNA agent would cause the oocyte to exit metaphase arrest and initiate parthenogenetic development (Colledge et al. Nature 370: 65-68, 1994; Hashimoto et al. Nature, 370:68-71, 1994). The effect of the modified agent on target RNA levels can be verified by Northern blot to assay for a decrease in the level of target mRNA, or by Western blot to assay for a decrease in the level of target protein, as compared to a negative control. Controls can include cells in which with no agent is added and/or cells in which a non-modified RNA is added.
  • References
  • General References
  • The oligoribonucleotides and oligoribonucleosides used in accordance with this invention may be with solid phase synthesis, see for example “Oligonucleotide synthesis, a practical approach”, Ed. M. J. Gait, IRL Press, 1984; “Oligonucleotides and Analogues, A Practical Approach”, Ed. F. Eckstein, IRL Press, 1991 (especially Chapter 1, Modern machine-aided methods of oligodeoxyribonucleotide synthesis, Chapter 2, Oligoribonucleotide synthesis, Chapter 3,2′-O—Methyloligoribonucleotide-s: synthesis and applications, Chapter 4, Phosphorothioate oligonucleotides, Chapter 5, Synthesis of oligonucleotide phosphorodithioates, Chapter 6, Synthesis of oligo-2′-deoxyribonucleoside methylphosphonates, and. Chapter 7, Oligodeoxynucleotides containing modified bases. Other particularly useful synthetic procedures, reagents, blocking groups and reaction conditions are described in Martin, P., Helv. Chim. Acta, 1995, 78, 486-504; Beaucage, S. L. and Iyer, R. P., Tetrahedron, 1992, 48, 2223-2311 and Beaucage, S. L. and Iyer, R. P., Tetrahedron, 1993, 49, 6123-6194, or references referred to therein.
  • Modification described in WO 00/44895, WO01/75164, or WO02/44321 can be used herein.
  • The disclosure of all publications, patents, and published patent applications listed herein are hereby incorporated by reference.
  • Phosphate Group References
  • The preparation of phosphinate oligoribonucleotides is described in U.S. Pat. No. 5,508,270. The preparation of alkyl phosphonate oligoribonucleotides is described in U.S. Pat. No. 4,469,863. The preparation of phosphoramidite oligoribonucleotides is described in U.S. Pat. No. 5,256,775 or U.S. Pat. No. 5,366,878. The preparation of phosphotriester oligoribonucleotides is described in U.S. Pat. No. 5,023,243. The preparation of borano phosphate oligoribonucleotide is described in U.S. Pat. Nos. 5,130,302 and 5,177,198. The preparation of 3′-Deoxy-3′-amino phosphoramidate oligoribonucleotides is described in U.S. Pat. No. 5,476,925. 3′-Deoxy-3′-methylenephosphonate oligoribonucleotides is described in An, H, et al. J. Org. Chem. 2001, 66, 2789-2801. Preparation of sulfur bridged nucleotides is described in Sproat et al. Nucleosides Nucleotides 1988, 7,651 and Crosstick et al. Tetrahedron Lett. 1989, 30, 4693.
  • Sugar Group References
  • Modifications to the 2′ modifications can be found in Verma, S. et al. Annu. Rev. Biochem. 1998, 67, 99-134 and all references therein. Specific modifications to the ribose can be found in the following references: 2′-fluoro (Kawasaki et. al., J. Med. Chem., 1993, 36, 831-841), 2′-MOE (Martin, P. Helv. Chim. Acta 1996, 79, 1930-1938), “LNA” (Wengel, J. Acc. Chem. Res. 1999, 32, 301-310).
  • Replacement of the Phosphate Group References
  • Methylenemethylimino linked oligoribonucleosides, also identified herein as MMI linked oligoribonucleosides, methylenedimethylhydrazo linked oligoribonucleosides, also identified herein as MDH linked oligoribonucleosides, and methylenecarbonylamino linked oligonucleosides, also identified herein as amide-3 linked oligoribonucleosides, and methyleneaminocarbonyl linked oligonucleosides, also identified herein as amide-4 linked oligoribonucleosides as well as mixed backbone compounds having, as for instance, alternating MMI and PO or PS linkages can be prepared as is described in U.S. Pat. Nos. 5,378,825, 5,386,023, 5,489,677 and in published PCT applications PCT/US92/04294 and PCT/US92/04305 (published as WO 92/20822 WO and 92/20823, respectively). Formacetal and thioformacetal linked oligoribonucleosides can be prepared as is described in U.S. Pat. Nos. 5,264,562 and 5,264,564. Ethylene oxide linked oligoribonucleosides can be prepared as is described in U.S. Pat. No. 5,223,618. Siloxane replacements are described in Cormier, J. F. et al. Nucleic Acids Res. 1988, 16, 4583. Carbonate replacements are described in Tittensor, J. R. J. Chem. Soc. C 1971, 1933. Carboxymethyl replacements are described in Edge, M. D. et al. J. Chem. Soc. Perkin Trans. 1 1972, 1991. Carbamate replacements are described in Stirchak, E. P. Nucleic Acids Res. 1989, 17, 6129.
  • Replacement of the Phosphate-Ribose Backbone References
  • Cyclobutyl sugar surrogate compounds can be prepared as is described in U.S. Pat. No. 5,359,044. Pyrrolidine sugar surrogate can be prepared as is described in U.S. Pat. No. 5,519,134. Morpholino sugar surrogates can be prepared as is described in U.S. Pat. Nos. 5,142,047 and 5,235,033, and other related patent disclosures. Peptide Nucleic Acids (PNAs) are known per se and can be prepared in accordance with any of the various procedures referred to in Peptide Nucleic Acids (PNA): Synthesis, Properties and Potential Applications, Bioorganic & Medicinal Chemistry, 1996, 4, 5-23. They may also be prepared in accordance with U.S. Pat. No. 5,539,083.
  • Terminal Modification References
  • Terminal modifications are described in Manoharan, M. et al. Antisense and Nucleic Acid Drug Development 12, 103-128 (2002) and references therein.
  • Bases References
  • N-2 substitued purine nucleoside amidites can be prepared as is described in U.S. Pat. No. 5,459,255. 3-Deaza purine nucleoside amidites can be prepared as is described in U.S. Pat. No. 5,457,191. 5,6-Substituted pyrimidine nucleoside amidites can be prepared as is described in U.S. Pat. No. 5,614,617. 5-Propynyl pyrimidine nucleoside amidites can be prepared as is described in U.S. Pat. No. 5,484,908. Additional references can be disclosed in the above section on base modifications.
  • Preferred iRNA Agents
  • Preferred RNA agents have the following structure (see Formula 2 below):
    Figure US20050119214A1-20050602-C00082
  • Referring to Formula 2 above, R1, R2, and R3 are each, independently, H, (i.e. abasic nucleotides), adenine, guanine, cytosine and uracil, inosine, thymine, xanthine, hypoxanthine, nubularine, tubercidine, isoguanisine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 5-halouracil, 5-(2-aminopropyl)uracil, 5-amino allyl uracil, 8-halo, amino, thiol, thioalkyl, hydroxyl and other 8-substituted adenines and guanines, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine, 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine, dihydrouracil, 3-deaza-5-azacytosine, 2-aminopurine, 5-alkyluracil, 7-alkylguanine, 5-alkyl cytosine, 7-deazaadenine, 7-deazaguanine, N6, N6-dimethyladenine, 2,6-diaminopurine, 5-amino-allyl-uracil, N3-methyluracil, substituted 1,2,4-triazoles, 2-pyridinone, 5-nitroindole, 3-nitropyrrole, 5-methoxyuracil, uracil-5-oxyacetic acid, 5-methoxycarbonylmethyluracil, 5-methyl-2-thiouracil, 5-methoxycarbonylmethyl-2-thiouracil, 5-methylaminomethyl-2-thiouracil, 3-(3-amino-3carboxypropyl)uracil, 3-methylcytosine, 5-methylcytosine, N4-acetyl cytosine, 2-thiocytosine, N6-methyladenine, N6-isopentyladenine, 2-methylthio-N-6-isopentenyladenine, N-methylguanines, or O-alkylated bases.
  • R4, R5, and R6 are each, independently, OR8, O(CH2CH2O)mCH2CH2OR8; O(CH2),R9; O(CH2)nOR9, H; halo; NH2; NHR8; N(R8)2; NH(CH2CH2NH)mCH2CH2NHR9; NHC(O)R8; cyano; mercapto, SR8; alkyl-thio-alkyl; alkyl, aralkyl, cycloalkyl, aryl, heteroaryl, alkenyl, alkynyl, each of which may be optionally substituted with halo, hydroxy, oxo, nitro, haloalkyl, alkyl, alkaryl, aryl, aralkyl, alkoxy, aryloxy, amino, alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, acylamino, alkylcarbamoyl, arylcarbamoyl, aminoalkyl, alkoxycarbonyl, carboxy, hydroxyalkyl, alkanesulfonyl, alkanesulfonamido, arenesulfonamido, aralkylsulfonamido, alkylcarbonyl, acyloxy, cyano, or ureido; or R4, R5, or R6 together combine with R7 to form an [—O—CH2—] covalently bound bridge between the sugar 2′ and 4′ carbons.
  • A1 is:
    Figure US20050119214A1-20050602-C00083
  • H; OH; OCH3; W1; an abasic nucleotide; or absent;
  • (a preferred A1, especially with regard to anti-sense strands, is chosen from 5′-monophosphate ((HO)2(O)P—O-5′), 5′-diphosphate ((HO)2(O)P—O—P(HO)(O)—O-5′), 5′-triphosphate ((HO)2(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′), 5′-guanosine cap (7-methylated or non-methylated) (7m-G-O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′), 5′-adenosine cap (Appp), and any modified or unmodified nucleotide cap structure (N—O-5′-(HO)(O)P—O—(HO)(O)P—O—P(HO)(O)—O-5′), 5′-monothiophosphate (phosphorothioate; (HO)2(S)P—O-5′), 5′-monodithiophosphate (phosphorodithioate; (HO)(HS)(S)P—O-5′), 5′-phosphorothiolate ((HO)2(O)P—S-5′); any additional combination of oxgen/sulfur replaced monophosphate, diphosphate and triphosphates (e.g. 5′-alpha-thiotriphosphate, 5′-gamma-thiotriphosphate, etc.), 5′-phosphoramidates ((HO)2(O)P—NH-5′, (HO)(NH2)(O)P—O-5′), 5′-alkylphosphonates (R=alkyl=methyl, ethyl, isopropyl, propyl, etc., e.g. RP(OH)(O)—O-5′-, (OH)2(O)P-5′-CH2—), 5′-alkyletherphosphonates (R=alkylether=methoxymethyl (MeOCH2—), ethoxymethyl, etc., e.g. RP(OH)(O)—O-5′-)).
  • A2 is:
    Figure US20050119214A1-20050602-C00084
  • A3 is:
    Figure US20050119214A1-20050602-C00085
  • and
  • A4 is:
    Figure US20050119214A1-20050602-C00086
  • H; Z4; an inverted nucleotide; an a basic nucleotide; or absent.
  • W1 is OH, (CH2)nR10, (CH2)nNHR10, (CH2)nOR10, (CH2)nSR10; O(CH2)nR10; O(CH2)nOR10, O(CH2)nNR10, O(CH2)nSR10; O(CH2)nSS(CH2)nOR10, O(CH2)nC(O)OR10, NH(CH2)nR10; NH(CH2)nNR10; NH(CH2)nOR10, NH(CH2)nSR10; S(CH2)nR10, S(CH2)nNR10, S(CH2)nOR10, S(CH2)nSR10 O(CH2CH2O)nCH2CH2OR10; O(CH2CH2O)mCH2CH2NHR10, NH(CH2CH2NH)mCH2CH2NHR10; Q-R10, O-Q-R10 N-Q-R10, S-Q-R10 or —O—. W4 is O, CH2, NH, or S.
  • X1, X2, X3, and X4 are each, independently, O or S.
  • Y1, Y2, Y3, and Y4 are each, independently, OH, O—, OR8, S, Se, BH3 , H, NHR9, N(R9)2 alkyl, cycloalkyl, aralkyl, aryl, or heteroaryl, each of which may be optionally substituted.
  • Z1, Z2, and Z3 are each independently O, CH2, NH, or S. Z4 is OH, (CH2)nR10, (CH2)nNHR10, (CH2)n OR10, (CH2)n SR10; O(CH2)nR10; O(CH2)nOR10, O(CH2)nNR10, O(CH2)nSR10, O(CH2)nSS(CH2)nOR10, O(CH2)nC(O)OR10; NH(CH2)nR10; NH(CH2)nNR10; NH(CH2)nOR10, NH(CH2)nSR10; S(CH2)nR10, S(CH2)nNR10, S(CH2)nOR10, S(CH2)nSR10, O(CH2CH2O)mCH2CH2OR10, O(CH2CH2O)mCH2CH2NHR10 NH(CH2CH2NH)mCH2CH2NHR10; Q-R10, O-Q-R10 N-Q-R10, S-Q-R10.
  • x is 5-100, chosen to comply with a length for an RNA agent described herein.
  • R7 is H; or is together combined with R4, R5, or R6 to form an [—O—CH2—] covalently bound bridge between the sugar 2′ and 4′ carbons.
  • R8 is alkyl, cycloalkyl, aryl, aralkyl, heterocyclyl, heteroaryl, amino acid, or sugar; R9 is NH2, alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid; and R10 is H; fluorophore (pyrene, TAMRA, fluorescein, Cy3 or Cy5 dyes); sulfur, silicon, boron or ester protecting group; intercalating agents (e.g. acridines), cross-linkers (e.g. psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases (e.g. EDTA), lipohilic carriers (cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, bomeol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, O3-(oleoyl)lithocholic acid, 03-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine) and peptide conjugates (e.g., antennapedia peptide, Tat peptide), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG]2, polyamino; alkyl, cycloalkyl, aryl, aralkyl, heteroaryl; radiolabelled markers, enzymes, haptens (e.g. biotin), transport/absorption facilitators (e.g., aspirin, vitamin E, folic acid), synthetic ribonucleases (e.g., imidazole, bisimidazole, histamine, imidazole clusters, acridine-imidazole conjugates, Eu3+complexes of tetraazamacrocycles); or an RNA agent. m is 0-1,000,000, and n is 0-20. Q is a spacer selected from the group consisting of abasic sugar, amide, carboxy, oxyamine, oxyimine, thioether, disulfide, thiourea, sulfonamide, or morpholino, biotin or fluorescein reagents.
  • Preferred RNA agents in which the entire phosphate group has been replaced have the following structure (see Formula 3 below):
    Figure US20050119214A1-20050602-C00087
  • Referring to Formula 3, A10-A40 is L-G-L; A10 and/or A40 may be absent, in which L is a linker, wherein one or both L may be present or absent and is selected from the group consisting of CH2(CH2)g; N(CH2)g; O(CH2)g; S(CH2)g. G is a functional group selected from the group consisting of siloxane, carbonate, carboxymethyl, carbamate, amide, thioether, ethylene oxide linker, sulfonate, sulfonamide, thioformacetal, formacetal, oxime, methyleneimino, methylenemethylimino, methylenehydrazo, methylenedimethylhydrazo and methyleneoxymethylimino.
  • R10, R20, and R30 are each, independently, H, (i.e. abasic nucleotides), adenine, guanine, cytosine and uracil, inosine, thymine, xanthine, hypoxanthine, nubularine, tubercidine, isoguanisine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 5-halouracil, 5-(2-aminopropyl)uracil, 5-amino allyl uracil, 8-halo, amino, thiol, thioalkyl, hydroxyl and other 8-substituted adenines and guanines, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine, 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine, dihydrouracil, 3-deaza-5-azacytosine, 2-aminopurine, 5-alkyluracil, 7-alkylguanine, 5-alkyl cytosine, 7-deazaadenine, 7-deazaguanine, N6, N6-dimethyladenine, 2,6-diaminopurine, 5-amino-allyl-uracil, N3-methyluracil substituted 1,2,4-triazoles, 2-pyridinone, 5-nitroindole, 3-nitropyrrole, 5-methoxyuracil, uracil-5-oxyacetic acid, 5-methoxycarbonylmethyluracil, 5-methyl-2-thiouracil, 5-methoxycarbonylmethyl-2-thiouracil, 5-methylaminomethyl-2-thiouracil, 3-(3-amino-3carboxypropyl)uracil, 3-methylcytosine, 5-methylcytosine, N4-acetyl cytosine, 2-thiocytosine, N6-methyladenine, N6-isopentyladenine, 2-methylthio-N-6-isopentenyladenine, N-methylguanines, or O-alkylated bases.
  • R40, R50, and R60 are each, independently, OR8, O(CH2CH2O)mCH2CH2OR8; O(CH2)nR9; O(CH2)nOR9, H; halo; NH2; NHR8; N(R8)2; NH(CH2CH2NH)mCH2CH2R9; NHC(O)R8; cyano; mercapto, SR7; alkyl-thio-alkyl; alkyl, aralkyl, cycloalkyl, aryl, heteroaryl, alkenyl, alkynyl, each of which may be optionally substituted with halo, hydroxy, oxo, nitro, haloalkyl, alkyl, alkaryl, aryl, aralkyl, alkoxy, aryloxy, amino, alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, acylamino, alkylcarbamoyl, arylcarbamoyl, aminoalkyl, alkoxycarbonyl, carboxy, hydroxyalkyl, alkanesulfonyl, alkanesulfonamido, arenesulfonamido, aralkylsulfonamido, alkylcarbonyl, acyloxy, cyano, and ureido groups; or R40, R50, or R60 together combine with R70 to form an [—O—CH2—] covalently bound bridge between the sugar 2′ and 4′ carbons.
  • x is 5-100 or chosen to comply with a length for an RNA agent described herein.
  • R70 is H; or is together combined with R40, R50, or R60 to form an [—O—CH2—] covalently bound bridge between the sugar 2′ and 4′ carbons.
  • R8 is alkyl, cycloalkyl, aryl, aralkyl, heterocyclyl, heteroaryl, amino acid, or sugar; and R9 is NH2, alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid. m is 0-1,000,000, n is 0-20, and g is 0-2.
  • Preferred nucleoside surrogates have the following structure (see Formula 4 below):
    SLR100-(M-SLR200)x-M-sLR300  FORMULA 4
  • S is a nucleoside surrogate selected from the group consisting of mophilino, cyclobutyl, pyrrolidine and peptide nucleic acid. L is a linker and is selected from the group consisting of CH2(CH2)g; N(CH2)g; O(CH2)g; S(CH2)g; —C(O)(CH2)n— or may be absent. M is an amide bond; sulfonamide; sulfinate; phosphate group; modified phosphate group as described herein; or may be absent.
  • R100, R200, and R300 are each, independently, H (i.e., abasic nucleotides), adenine, guanine, cytosine and uracil, inosine, thymine, xanthine, hypoxanthine, nubularine, tubercidine, isoguanisine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 5-halouracil, 5-(2-aminopropyl)uracil, 5-amino allyl uracil, 8-halo, amino, thiol, thioalkyl, hydroxyl and other 8-substituted adenines and guanines, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine, 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine, dihydrouracil, 3-deaza-5-azacytosine, 2-aminopurine, 5-alkyluracil, 7-alkylguanine, 5-alkyl cytosine, 7-deazaadenine, 7-deazaguanine, N6, N6-dimethyladenine, 2,6-diaminopurine, 5-amino-allyl-uracil, N3-methyluracil substituted 1, 2, 4,-triazoles, 2-pyridinones, 5-nitroindole, 3-nitropyrrole, 5-methoxyuracil, uracil-5-oxyacetic acid, 5-methoxycarbonylmethyluracil, 5-methyl-2-thiouracil, 5-methoxycarbonylmethyl-2-thiouracil, 5-methylaminomethyl-2-thiouracil, 3-(3-amino-3carboxypropyl)uracil, 3-methylcytosine, 5-methylcytosine, N4-acetyl cytosine, 2-thiocytosine, N6-methyladenine, N6-isopentyladenine, 2-methylthio-N-6-isopentenyladenine, N-methylguanines, or O-alkylated bases.
  • x is 5-100, or chosen to comply with a length for an RNA agent described herein; and g is 0-2.
  • Nuclease Resistant Monomers
  • The monomers and methods described herein can be used to prepare an RNA, e.g., an iRNA agent, that incorporates a nuclease resistant monomer (NRM), such as those described herein and those described in copending, co-owned U.S. Provisional Application Ser. No. 60/469,612, filed on May 9, 2003, and International Application No. PCT/US04/07070, both of which are hereby incorporated by reference.
  • An iRNA agent can include monomers which have been modifed so as to inhibit degradation, e.g., by nucleases, e.g., endonucleases or exonucleases, found in the body of a subject. These monomers are referred to herein as NRMs, or nuclease resistance promoting monomers or modifications. In many cases these modifications will modulate other properties of the iRNA agent as well, e.g., the ability to interact with a protein, e.g., a transport protein, e.g., serum albumin, or a member of the RISC(RNA-induced Silencing Complex), or the ability of the first and second sequences to form a duplex with one another or to form a duplex with another sequence, e.g., a target molecule.
  • While not wishing to be bound by theory, it is believed that modifications of the sugar, base, and/or phosphate backbone in an iRNA agent can enhance endonuclease and exonuclease resistance, and can enhance interactions with transporter proteins and one or more of the functional components of the RISC complex. Preferred modifications are those that increase exonuclease and endonuclease resistance and thus prolong the half-life of the iRNA agent prior to interaction with the RISC complex, but at the same time do not render the iRNA agent resistant to endonuclease activity in the RISC complex. Again, while not wishing to be bound by any theory, it is believed that placement of the modifications at or near the 3′ and/or 5′ end of antisense strands can result in iRNA agents that meet the preferred nuclease resistance criteria delineated above. Again, still while not wishing to be bound by any theory, it is believed that placement of the modifications at e.g., the middle of a sense strand can result in iRNA agents that are relatively less likely to undergo off-targeting.
  • Modifications described herein can be incorporated into any double-stranded RNA and RNA-like molecule described herein, e.g., an iRNA agent. An iRNA agent may include a duplex comprising a hybridized sense and antisense strand, in which the antisense strand and/or the sense strand may include one or more of the modifications described herein. The anti sense strand may include modifications at the 3′ end and/or the 5′ end and/or at one or more positions that occur 1-6 (e.g., 1-5,1-4, 1-3,1-2) nucleotides from either end of the strand. The sense strand may include modifications at the 3′ end and/or the 5′ end and/or at any one of the intervening positions between the two ends of the strand. The iRNA agent may also include a duplex comprising two hybridized antisense strands. The first and/or the second antisense strand may include one or more of the modifications described herein. Thus, one and/or both antisense strands may include modifications at the 3′ end and/or the 5′ end and/or at one or more positions that occur 1-6 (e.g., 1-5,1-4, 1-3,1-2) nucleotides from either end of the strand. Particular configurations are discussed below.
  • Modifications that can be useful for producing iRNA agents that meet the preferred nuclease resistance criteria delineated above can include one or more of the following chemical and/or stereochemical modifications of the sugar, base, and/or phosphate backbone:
      • (i) chiral (Sp) thioates. Thus, preferred NRMs include nucleotide dimers with an enriched or pure for a particular chiral form of a modified phosphate group containing a heteroatom at the nonbridging position, e.g., Sp or Rp, at the position X, where this is the position normally occupied by the oxygen. The atom at X can also be S, Se, Nr2, or Br3. When X is S, enriched or chirally pure Sp linkage is preferred. Enriched means at least 70, 80, 90, 95, or 99% of the preferred form. Such NRMs are discussed in more detail below;
      • (ii) attachment of one or more cationic groups to the sugar, base, and/or the phosphorus atom of a phosphate or modified phosphate backbone moiety. Thus, preferred NRMs include monomers at the terminal position derivatized at a cationic group. As the 5′ end of an antisense sequence should have a terminal —OH or phosphate group this NRM is preferably not used at the 5′ end of an anti-sense sequence. The group should be attached at a position on the base which minimizes interference with H bond formation and hybridization, e.g., away form the face which interacts with the complementary base on the other strand, e.g, at the 5′ position of a pyrimidine or a 7-position of a purine. These are discussed in more detail below;
      • (iii) nonphosphate linkages at the termini. Thus, preferred NRMs include Non-phosphate linkages, e.g., a linkage of 4 atoms which confers greater resistance to cleavage than does a phosphate bond. Examples include 3′ CH2—NCH3—O—CH2-5′ and 3′ CH2—NH—(O═)—CH2-5′.;
      • (iv) 3′-bridging thiophosphates and 5′-bridging thiophosphates. Thus, preferred NRM's can included these structures;
      • (v) L-RNA, 2′-5′ linkages, inverted linkages, a-nucleosides. Thus, other preferred NRM's include: L nucleosides and dimeric nucleotides derived from L-nucleosides; 2′-5′ phosphate, non-phosphate and modified phosphate linkages (e.g., thiophosphates, phosphoramidates and boronophosphates); dimers having inverted linkages, e.g., 3′-3′ or 5′-5′ linkages; monomers having an alpha linkage at the 1′ site on the sugar, e.g., the structures described herein having an alpha linkage;
      • (vi) conjugate groups. Thus, preferred NRM's can include e.g., a targeting moiety or a conjugated ligand described herein conjugated with the monomer, e.g., through the sugar, base, or backbone;
      • (vi) abasic linkages. Thus, preferred NRM's can include an abasic monomer, e.g., an abasic monomer as described herein (e.g., a nucleobaseless monomer); an aromatic or heterocyclic or polyheterocyclic aromatic monomer as described herein.; and
      • (vii) 5′-phosphonates and 5′-phosphate prodrugs. Thus, preferred NRM's include monomers, preferably at the terminal position, e.g., the 5′ position, in which one or more atoms of the phosphate group is derivatized with a protecting group, which protecting group or groups, are removed as a result of the action of a component in the subject's body, e.g, a carboxyesterase or an enzyme present in the subject's body. E.g., a phosphate prodrug in which a carboxy esterase cleaves the protected molecule resulting in the production of a thioate anion which attacks a carbon adjacent to the 0 of a phosphate and resulting in the production of an unprotected phosphate.
  • One or more different NRM modifications can be introduced into an iRNA agent or into a sequence of an iRNA agent. An NRM modification can be used more than once in a sequence or in an iRNA agent. As some NRM's interfere with hybridization the total number incorporated, should be such that acceptable levels of iRNA agent duplex formation are maintained.
  • In some embodiments NRM modifications are introduced into the terminal the cleavage site or in the cleavage region of a sequence (a sense strand or sequence) which does not target a desired sequence or gene in the subject. This can reduce off-target silencing.
  • Chiral Sp Thioates
  • A modification can include the alteration, e.g., replacement, of one or both of the non-linking (X and Y) phosphate oxygens and/or of one or more of the linking (W and Z) phosphate oxygens. Formula X below depicts a phosphate moiety linking two sugar/sugar surrogate-base moieties, SB1 and SB2.
    Figure US20050119214A1-20050602-C00088
  • In certain embodiments, one of the non-linking phosphate oxygens in the phosphate backbone moiety (X and Y) can be replaced by any one of the following: S, Se, BR3 (R is hydrogen, alkyl, aryl, etc.), C (i.e., an alkyl group, an aryl group, etc.), H, NR2 (R is hydrogen, alkyl, aryl, etc.), or OR (R is alkyl or aryl). The phosphorus atom in an unmodified phosphate group is achiral. However, replacement of one of the non-linking oxygens with one of the above atoms or groups of atoms renders the phosphorus atom chiral; in other words a phosphorus atom in a phosphate group modified in this way is a stereogenic center. The stereogenic phosphorus atom can possess either the “R” configuration (herein Rp) or the “S” configuration (herein Sp). Thus if 60% of a population of stereogenic phosphorus atoms have the Rp configuration, then the remaining 40% of the population of stereogenic phosphorus atoms have the Sp configuration.
  • In some embodiments, iRNA agents, having phosphate groups in which a phosphate non-linking oxygen has been replaced by another atom or group of atoms, may contain a population of stereogenic phosphorus atoms in which at least about 50% of these atoms (e.g., at least about 60% of these atoms, at least about 70% of these atoms, at least about 80% of these atoms, at least about 90% of these atoms, at least about 95% of these atoms, at least about 98% of these atoms, at least about 99% of these atoms) have the Sp configuration. Alternatively, iRNA agents having phosphate groups in which a phosphate non-linking oxygen has been replaced by another atom or group of atoms may contain a population of stereogenic phosphorus atoms in which at least about 50% of these atoms (e.g., at least about 60% of these atoms, at least about 70% of these atoms, at least about 80% of these atoms, at least about 90% of these atoms, at least about 95% of these atoms, at least about 98% of these atoms, at least about 99% of these atoms) have the Rp configuration. In other embodiments, the population of stereogenic phosphorus atoms may have the Sp configuration and may be substantially free of stereogenic phosphorus atoms having the Rp configuration. In still other embodiments, the population of stereogenic phosphorus atoms may have the Rp configuration and may be substantially free of stereogenic phosphorus atoms having the Sp configuration. As used herein, the phrase “substantially free of stereogenic phosphorus atoms having the Rp configuration” means that moieties containing stereogenic phosphorus atoms having the Rp configuration cannot be detected by conventional methods known in the art (chiral HPLC, 1H NMR analysis using chiral shift reagents, etc.). As used herein, the phrase “substantially free of stereogenic phosphorus atoms having the Sp configuration” means that moieties containing stereogenic phosphorus atoms having the Sp configuration cannot be detected by conventional methods known in the art (chiral HPLC, 1H NMR analysis using chiral shift reagents, etc.).
  • In a preferred embodiment, modified iRNA agents contain a phosphorothioate group, i.e., a phosphate groups in which a phosphate non-linking oxygen has been replaced by a sulfur atom. In an especially preferred embodiment, the population of phosphorothioate stereogenic phosphorus atoms may have the Sp configuration and be substantially free of stereogenic phosphorus atoms having the Rp configuration.
  • Phosphorothioates may be incorporated into iRNA agents using dimers e.g., formulas X-1 and X-2. The former can be used to introduce phosphorothioate
    Figure US20050119214A1-20050602-C00089

    at the 3′ end of a strand, while the latter can be used to introduce this modification at the 5′ end or at a position that occurs e.g., 1, 2, 3, 4, 5, or 6 nucleotides from either end of the strand. In the above formulas, Y can be 2-cyanoethoxy, W and Z can be 0, R2 can be, e.g., a substituent that can impart the C-3 endo configuration to the sugar (e.g., OH, F, OCH3), DMT is dimethoxytrityl, and “BASE” can be a natural, unusual, or a universal base.
  • X-1 and X-2 can be prepared using chiral reagents or directing groups that can result in phosphorothioate-containing dimers having a population of stereogenic phosphorus atoms having essentially only the Rp configuration (i.e., being substantially free of the Sp configuration) or only the Sp configuration (i.e., being substantially free of the Rp configuration). Alternatively, dimers can be prepared having a population of stereogenic phosphorus atoms in which about 50% of the atoms have the Rp configuration and about 50% of the atoms have the Sp configuration. Dimers having stereogenic phosphorus atoms with the Rp configuration can be identified and separated from dimers having stereogenic phosphorus atoms with the Sp configuration using e.g., enzymatic degradation and/or conventional chromatography techniques.
  • Cationic Groups
  • Modifications can also include attachment of one or more cationic groups to the sugar, base, and/or the phosphorus atom of a phosphate or modified phosphate backbone moiety. A cationic group can be attached to any atom capable of substitution on a natural, unusual or universal base. A preferred position is one that does not interfere with hybridization, i.e., does not interfere with the hydrogen bonding interactions needed for base pairing. A cationic group can be attached e.g., through the C2′ position of a sugar or analogous position in a cyclic or acyclic sugar surrogate. Cationic groups can include e.g., protonated amino groups, derived from e.g., O-AMINE (AMINE=NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino, ethylene diamine, polyamino); aminoalkoxy, e.g., O(CH2)nAMINE, (e.g., AMINE=NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino, ethylene diamine, polyamino); amino (e.g. NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, diheteroaryl amino, or amino acid); or NH(CH2CH2NH)nCH2CH2-AMINE (AMINE=NH2; alkylamino, dialkylamino, heterocyclyl, arylamino, diaryl amino, heteroaryl amino, or diheteroaryl amino).
  • Nonphosphate Linkages
  • Modifications can also include the incorporation of nonphosphate linkages at the 5′ and/or 3′ end of a strand. Examples of nonphosphate linkages which can replace the phosphate group include methyl phosphonate, hydroxylamino, siloxane, carbonate, carboxymethyl, carbamate, amide, thioether, ethylene oxide linker, sulfonate, sulfonamide, thioformacetal, formacetal, oxime, methyleneimino, methylenemethylimino, methylenehydrazo, methylenedimethylhydrazo and methyleneoxymethylimino. Preferred replacements include the methyl phosphonate and hydroxylamino groups.
  • 3′-Bridging Thiophosphates and 5′-Bridging Thiophosphates; Locked-RNA, 2′-5 Likages, Inverted Linkages, α-Nucleosides; Conjugate Groups; Abasic Linkages; and 5′-Phosphonates and 5′-Phosphate Prodrugs
  • Referring to formula X above, modifications can include replacement of one of the bridging or linking phosphate oxygens in the phosphate backbone moiety (W and Z). Unlike the situation where only one of X or Y is altered, the phosphorus center in the phosphorodithioates is achiral which precludes the formation of iRNA agents containing a stereogenic phosphorus atom.
  • Modifications can also include linking two sugars via a phosphate or modified phosphate group through the 2′ position of a first sugar and the 5′ position of a second sugar. Also contemplated are inverted linkages in which both a first and second sugar are eached linked through the respective 3′ positions. Modified RNA's can also include “abasic” sugars, which lack a nucleobase at C-1′. The sugar group can also contain one or more carbons that possess the opposite stereochemical configuration than that of the corresponding carbon in ribose. Thus, a modified iRNA agent can include nucleotides containing e.g., arabinose, as the sugar. In another subset of this modification, the natural, unusual, or universal base may have the α-configuration. Modifcations can also include L-RNA.
  • Modifications can also include 5′-phosphonates, e.g., P(O)(O)2—X—C5′-sugar (X=CH2, CF2, CHF and 5′-phosphate prodrugs, e.g., P(O)[OCH2CH2SC(O)R]2CH2C5′-sugar. In the latter case, the prodrug groups may be decomposed via reaction first with carboxy esterases. The remaining ethyl thiolate group via intramolecular S N2 displacement can depart as episulfide to afford the underivatized phosphate group.
  • Modification can also include the addition of conjugating groups described elseqhere herein, which are prefereably attached to an iRNA agent through any amino group available for conjugation.
  • Nuclease resistant modifications include some which can be placed only at the terminus and others which can go at any position. Generally the modifications that can inhibit hybridization so it is preferably to use them only in terminal regions, and preferrable to not use them at the cleavage site or in the cleavage region of an sequence which targets a subject sequence or gene. The can be used anywhere in a sense sequence, provided that sufficient hybridization between the two sequences of the iRNA agent is maintained. In some embodiments it is desirabable to put the NRM at the cleavage site or in the cleavage region of a sequence which does not target a subject sequence or gene, as it can minimize off-target silencing.
  • In addition, an iRNA agent described herein can have an overhang which does not form a duplex structure with the other sequence of the iRNA agent—it is an overhang, but it does hybridize, either with itself, or with another nucleic acid, other than the other sequence of the iRNA agent.
  • In most cases, the nuclease-resistance promoting modifications will be distributed differently depending on whether the sequence will target a sequence in the subject (often referred to as an anti-sense sequence) or will not target a sequence in the subject (often referred to as a sense sequence). If a sequence is to target a sequence in the subject, modifications which interfer with or inhibit endonuclease cleavage should not be inserted in the region which is subject to RISC mediated cleavage, e.g., the cleavage site or the cleavage region (As described in Elbashir et al., 2001, Genes and Dev. 15: 188, hereby incorporated by reference, cleavage of the target occurs about in the middle of a 20 or 21 nt guide RNA, or about 10 or 11 nucleotides upstream of the first nucleotide which is complementary to the guide sequence. As used herein cleavage site refers to the nucleotide on either side of the cleavage site, on the target or on the iRNA agent strand which hybridizes to it. Cleavage region means an nucleotide with 1, 2, or 3 nucletides of the cleave site, in either direction.)
  • Such modifications can be introduced into the terminal regions, e.g., at the terminal position or with 2, 3, 4, or 5 positions of the terminus, of a sequence which targets or a sequence which does not target a sequence in the subject.
  • An iRNA agent can have a first and a second strand chosen from the following:
      • a first strand which does not target a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end;
      • a first strand which does not target a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end;
      • a first strand which does not target a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end and which has a NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end;
      • a first strand which does not target a sequence and which has an NRM modification at the cleavage site or in the cleavage region;
      • a first strand which does not target a sequence and which has an NRM modification at the cleavage site or in the cleavage region and one or more of an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end, a NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end, or NRM modifications at or within 1, 2, 3, 4, 5, or 6 positions from both the 3′ and the 5′ end; and
      • a second strand which targets a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end;
      • a second strand which targets a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end (5′ end NRM modifications are preferentially not at the terminus but rather at a position 1, 2, 3, 4, 5, or 6 away from the 5′ terminus of an antisense strand);
      • a second strand which targets a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end and which has a NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end;
      • a second strand which targets a sequence and which preferably does not have an an NRM modification at the cleavage site or in the cleavage region;
      • a second strand which targets a sequence and which does not have an NRM modification at the cleavage site or in the cleavage region and one or more of an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end, a NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end, or NRM modifications at or within 1, 2, 3, 4, 5, or 6 positions from both the 3′ and the 5′ end (5′ end NRM modifications are preferentially not at the terminus but rather at a position 1, 2, 3, 4, 5, or 6 away from the 5′ terminus of an antisense strand).
  • An iRNA agent can also target two sequences and can have a first and second strand chosen from:
      • a first strand which targets a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end;
      • a first strand which targets a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end (5′ end NRM modifications are preferentially not at the terminus but rather at a position 1, 2, 3, 4, 5, or 6 away from the 5′ terminus of an antisense strand);
      • a first strand which targets a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end and which has a NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end;
      • a first strand which targets a sequence and which preferably does not have an an NRM modification at the cleavage site or in the cleavage region;
      • a first strand which targets a sequence and which dose not have an NRM modification at the cleavage site or in the cleavage region and one or more of an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end, a NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end, or NRM modifications at or within 1, 2, 3, 4, 5, or 6 positions from both the 3′ and the 5′ end (5′ end NRM modifications are preferentially not at the terminus but rather at a position 1, 2, 3, 4, 5, or 6 away from the 5′ terminus of an antisense strand) and a second strand which targets a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end;
      • a second strand which targets a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end (5′ end NRM modifications are preferentially not at the terminus but rather at a position 1, 2, 3, 4, 5, or 6 away from the 5′ terminus of an antisense strand);
      • a second strand which targets a sequence and which has an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end and which has a NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end;
      • a second strand which targets a sequence and which preferably does not have an an NRM modification at the cleavage site or in the cleavage region;
      • a second strand which targets a sequence and which dose not have an NRM modification at the cleavage site or in the cleavage region and one or more of an NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 3′ end, a NRM modification at or within 1, 2, 3, 4, 5, or 6 positions from the 5′ end, or NRM modifications at or within 1, 2, 3, 4, 5, or 6 positions from both the 3′ and the 5′ end (5′ end NRM modifications are preferentially not at the terminus but rather at a position 1, 2, 3, 4, 5, or 6 away from the 5′ terminus of an antisense strand).
    Ribose Mimics
  • The monomers and methods described herein can be used to prepare an RNA, e.g., an iRNA agent, that incorporates a ribose mimic, such as those described herein and those described in copending co-owned U.S. Provisional Application Ser. No. 60/454,962, filed on Mar. 13, 2003, and International Application No. PCT/U.S.04/07070, both of which are hereby incorporated by reference.
  • Thus, an aspect of the invention features an iRNA agent that includes a secondary hydroxyl group, which can increase efficacy and/or confer nuclease resistance to the agent. Nucleases, e.g., cellular nucleases, can hydrolyze nucleic acid phosphodiester bonds, resulting in partial or complete degradation of the nucleic acid. The secondary hydroxy group confers nuclease resistance to an iRNA agent by rendering the iRNA agent less prone to nuclease degradation relative to an iRNA which lacks the modification. While not wishing to be bound by theory, it is believed that the presence of a secondary hydroxyl group on the iRNA agent can act as a structural mimic of a 3′ ribose hydroxyl group, thereby causing it to be less susceptible to degradation.
  • The secondary hydroxyl group refers to an “OH” radical that is attached to a carbon atom substituted by two other carbons and a hydrogen. The secondary hydroxyl group that confers nuclease resistance as described above can be part of any acyclic carbon-containing group. The hydroxyl may also be part of any cyclic carbon-containing group, and preferably one or more of the following conditions is met (1) there is no ribose moiety between the hydroxyl group and the terminal phosphate group or (2) the hydroxyl group is not on a sugar moiety which is coupled to a base. The hydroxyl group is located at least two bonds (e.g., at least three bonds away, at least four bonds away, at least five bonds away, at least six bonds away, at least seven bonds away, at least eight bonds away, at least nine bonds away, at least ten bonds away, etc.) from the terminal phosphate group phosphorus of the iRNA agent. In preferred embodiments, there are five intervening bonds between the terminal phosphate group phosphorus and the secondary hydroxyl group.
  • Preferred iRNA agent delivery modules with five intervening bonds between the terminal phosphate group phosphorus and the secondary hydroxyl group have the following structure (see formula Y below):
    Figure US20050119214A1-20050602-C00090
  • Referring to formula Y, A is an iRNA agent, including any iRNA agent described herein. The iRNA agent may be connected directly or indirectly (e.g., through a spacer or linker) to “W” of the phosphate group. These spacers or linkers can include e.g., —(CH2)n—, —(CH2)nN—, —(CH2)nO—, —(CH2)nS—, O(CH2CH2O)nCH2CH2OH (e.g., n=3 or 6), abasic sugars, amide, carboxy, amine, oxyamine, oxyimine, thioether, disulfide, thiourea, sulfonamide, or morpholino, or biotin and fluorescein reagents.
  • The iRNA agents can have a terminal phosphate group that is unmodified (e.g., W, X, Y, and Z are O) or modified. In a modified phosphate group, W and Z can be independently NH, O, or S; and X and Y can be independently S, Se, BH3 , C1-C6 alkyl, C6-C10 aryl, H, O, O, alkoxy or amino (including alkylamino, arylamino, etc.). Preferably, W, X and Z are 0 and Y is S.
  • R1 and R3 are each, independently, hydrogen; or C1-C100 alkyl, optionally substituted with hydroxyl, amino, halo, phosphate or sulfate and/or may be optionally inserted with N, O, S, alkenyl or alkynyl.
  • R2 is hydrogen; C1-C100 alkyl, optionally substituted with hydroxyl, amino, halo, phosphate or sulfate and/or may be optionally inserted with N, O, S, alkenyl or alkynyl; or, when n is 1, R2 may be taken together with with R4 or R6 to form a ring of 5-12 atoms.
  • R4 is hydrogen; C1-C100 alkyl, optionally substituted with hydroxyl, amino, halo, phosphate or sulfate and/or may be optionally inserted with N, O, S, alkenyl or alkynyl; or, when n is 1, R4 may be taken together with with R2 or R5 to form a ring of 5-12 atoms.
  • R5 is hydrogen, C1-C100 alkyl optionally substituted with hydroxyl, amino, halo, phosphate or sulfate and/or may be optionally inserted with N, O, S, alkenyl or alkynyl; or, when n is 1, R5 may be taken together with with R4 to form a ring of 5-12 atoms.
  • R6 is hydrogen, C1-C100 alkyl, optionally substituted with hydroxyl, amino, halo, phosphate or sulfate and/or may be optionally inserted with N, O, S, alkenyl or alkynyl, or, when n is 1, R6 may be taken together with with R2 to form a ring of 6-10 atoms;
  • R7 is hydrogen, C1-C100 alkyl, or C(O)(CH2)qC(O)NHR9; T is hydrogen or a functional group; n and q are each independently 1-100; R8 is C1-C10 alkyl or C6-C10 aryl; and R9 is hydrogen, C1-C10 alkyl, C6-C10 aryl or a solid support agent.
  • Preferred embodiments may include one of more of the following subsets of iRNA agent delivery modules.
  • In one subset of RNAi agent delivery modules, A can be connected directly or indirectly through a terminal 3′ or 5′ ribose sugar carbon of the RNA agent.
  • In another subset of RNAi agent delivery modules, X, W, and Z are 0 and Y is S.
  • In still yet another subset of RNAi agent delivery modules, n is 1, and R2 and R6 are taken together to form a ring containing six atoms and R4 and R5 are taken together to form a ring containing six atoms. Preferably, the ring system is a trans-decalin. For example, the RNAi agent delivery module of this subset can include a compound of Formula (Y-1):
    Figure US20050119214A1-20050602-C00091
  • The functional group can be, for example, a targeting group (e.g., a steroid or a carbohydrate), a reporter group (e.g., a fluorophore), or a label (an isotopically labelled moiety). The targeting group can further include protein binding agents, endothelial cell targeting groups (e.g., RGD peptides and mimetics), cancer cell targeting groups (e.g., folate Vitamin B12, Biotin), bone cell targeting groups (e.g., bisphosphonates, polyglutamates, polyaspartates), multivalent mannose (for e.g., macrophage testing), lactose, galactose, N-acetyl-galactosamine, monoclonal antibodies, glycoproteins, lectins, melanotropin, or thyrotropin.
  • As can be appreciated by the skilled artisan, methods of synthesizing the compounds of the formulae herein will be evident to those of ordinary skill in the art. The synthesized compounds can be separated from a reaction mixture and further purified by a method such as column chromatography, high pressure liquid chromatography, or recrystallization. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof.
  • Pharmaceutical Compositions
  • In one embodiment, the invention relates to a pharmaceutical composition containing a modified iRNA agent, as described in the preceding sections, and a pharmaceutically acceptable carrier, as described below. A pharmaceutical composition including the modified iRNA agent is useful for treating a disease caused by expression of a target gene. In this aspect of the invention, the iRNA agent of the invention is formulated as described below. The pharmaceutical composition is administered in a dosage sufficient to inhibit expression of the target gene.
  • The pharmaceutical compositions of the present invention are administered in dosages sufficient to inhibit the expression or activity of the target gene. Compositions containing the iRNA agent of the invention can be administered at surprisingly low dosages. A maximum dosage of 5 mg iRNA agent per kilogram body weight per day may be sufficient to inhibit or completely suppress the expression or activity of the target gene.
  • In general, a suitable dose of modified iRNA agent will be in the range of 0.001 to 500 milligrams per kilogram body weight of the recipient per day (e.g., about 1 microgram per kilogram to about 500 milligrams per kilogram, about 100 micrograms per kilogram to about 100 milligrams per kilogram, about 1 milligrams per kilogram to about 75 milligrams per kilogram, about 10 micrograms per kilogram to about 50 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram). The pharmaceutical composition may be administered once per day, or the iRNA agent may be administered as two, three, four, five, six or more sub-doses at appropriate intervals throughout the day. In that case, the iRNA agent contained in each sub-dose must be correspondingly smaller in order to achieve the total daily dosage. The dosage unit can also be compounded for delivery over several days, e.g., using a conventional sustained release formulation which provides sustained release of the iRNA agent over a several day period. Sustained release formulations are well known in the art. In this embodiment, the dosage unit contains a corresponding multiple of the daily dose.
  • The skilled artisan will appreciate that certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the infection or disease, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or a series of treatments. Estimates of effective dosages and in vivo half-lives for the individual iRNA agent encompassed by the invention can be made using conventional methodologies or on the basis of in vivo testing using an appropriate animal model, as described elsewhere herein.
  • Advances in mouse genetics have generated a number of mouse models for the study of various human diseases. For example, mouse repositories can be found at The Jackson Laboratory, Charles River Laboratories, Taconic, Harlan, Mutant Mouse Regional Resource Centers (MMRRC) National Network and at the European Mouse Mutant Archive. Such models may be used for in vivo testing of iRNA agent, as well as for determining a therapeutically effective dose.
  • The pharmaceutical compositions encompassed by the invention may be administered by any means known in the art including, but not limited to oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), ocular, rectal, vaginal and topical (including buccal and sublingual) administration. In preferred embodiments, the pharmaceutical compositions are administered by intravenous or intraparenteral infusion or injection. The pharmaceutical compositions can also be administered intraparenchymally, intrathecally, and/or by stereotactic injection.
  • For oral administration, the iRNA agent useful in the invention will generally be provided in the form of tablets or capsules, as a powder or granules, or as an aqueous solution or suspension.
  • Tablets for oral use may include the active ingredients mixed with pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
  • Capsules for oral use include hard gelatin capsules in which the active ingredient is mixed with a solid diluent, and soft gelatin capsules wherein the active ingredient is mixed with water or an oil such as peanut oil, liquid paraffin or olive oil.
  • For intramuscular, intraperitoneal, subcutaneous and intravenous use, the pharmaceutical compositions of the invention will generally be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and isotonicity. Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride. In a preferred embodiment, the carrier consists exclusively of an aqueous buffer. In this context, “exclusively” means no auxiliary agents or encapsulating substances are present which might affect or mediate uptake of iRNA agent in the cells that harbor the target gene or virus. Such substances include, for example, micellar structures, such as liposomes or capsids, as described below. Although microinjection, lipofection, viruses, viroids, capsids, capsoids, or other auxiliary agents are required to introduce iRNA agent into cell cultures, surprisingly these methods and agents are not necessary for uptake of iRNA agent in vivo. The iRNA agent of the present invention are particularly advantageous in that they do not require the use of an auxiliary agent to mediate uptake of the iRNA agent into the cell, many of which agents are toxic or associated with deleterious side effects. Aqueous suspensions according to the invention may include suspending agents such as cellulose derivatives, sodium alginate, polyvinyl-pyrrolidone and gum tragacanth, and a wetting agent such as lecithin. Suitable preservatives for aqueous suspensions include ethyl and n-propyl p-hydroxybenzoate.
  • The pharmaceutical compositions can also include encapsulated formulations to protect the iRNA agent against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811; PCT publication WO 91/06309; and European patent publication EP-A-43075, which are incorporated by reference herein.
  • Toxicity and therapeutic efficacy of iRNA agent can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. iRNA agents that exhibit high therapeutic indices are preferred.
  • The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosages of compositions of the invention are preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any iRNA agent used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range of the iRNA agent or, when appropriate, of the polypeptide product of a target sequence (e.g., achieving a decreased concentration of the polypeptide) that includes the IC50 (i.e., the concentration of the test iRNA agent which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
  • In addition to their administration individually or as a plurality, as discussed above, iRNA agents relating to the invention can be administered in combination with other known agents effective in treating viral infections and diseases. In any event, the administering physician can adjust the amount and timing of iRNA agent administration on the basis of results observed using standard measures of efficacy known in the art or described herein.
  • For oral administration, the iRNA agent useful in the invention will generally be provided in the form of tablets or capsules, as a powder or granules, or as an aqueous solution or suspension.
  • Methods for Treating Diseases Caused by Expression of a Target Gene.
  • In one embodiment, the invention relates to a method for treating a subject having a disease or at risk of developing a disease caused by the expression of a target gene. In this embodiment, iRNA agents can act as novel therapeutic agents for controlling one or more of cellular proliferative and/or differentiative disorders, disorders associated with bone metabolism, immune disorders, hematopoietic disorders, cardiovascular disorders, liver disorders, viral diseases, or metabolic disorders. The method includes administering a pharmaceutical composition of the invention to the patient (e.g., a human), such that expression of the target gene is silenced. Because of their high efficiency and specificity, the iRNA agent of the present invention specifically target mRNA of target genes of diseased cells and tissues, as described below, and at surprisingly low dosages. The pharmaceutical compositions are formulated as described in the preceding section, which is hereby incorporated by reference herein.
  • Examples of genes which can be targeted for treatment include, without limitation, an oncogene (Hanahan, D. and R. A. Weinberg, Cell (2000) 100:57; and Yokota, J., Carcinogenesis (2000) 21(3):497-503); a cytokine gene (Rubinstein, M., et al., Cytokine Growth Factor Rev. (1998) 9(2):175-81); a idiotype (Id) protein gene (Benezra, R., et al., Oncogene (2001) 20(58):8334-41; Norton, J. D., J. Cell Sci. (2000) 113(22):3897-905); a prion gene (Prusiner, S. B., et al., Cell (1998) 93(3):337-48; Safar, J., and S. B. Prusiner, Prog. Brain Res. (1998) 117:421-34); a gene that expresses molecules that induce angiogenesis (Gould, V. E. and B. M. Wagner, Hum. Pathol. (2002) 33(11):1061-3); adhesion molecules (Chothia, C. and E. Y. Jones, Annu. Rev. Biochem. (1997) 66:823-62; Parise, L. V., et al., Semin. Cancer Biol. (2000) 10(6):407-14); cell surface receptors (Deller, M. C., and Y. E. Jones, Curr. Opin. Struct. Biol. (2000) 10(2):213-9); genes of proteins that are involved in metastasizing and/or invasive processes (Boyd, D., Cancer Metastasis Rev. (1996) 15(1):77-89; Yokota, J., Carcinogenesis (2000) 21(3):497-503); genes of proteases as well as of molecules that regulate apoptosis and the cell cycle (Matrisian, L. M., Curr. Biol. (1999) 9(20): R776-8; Krepela, E., Neoplasma (2001) 48(5):332-49; Basbaum and Werb, Curr. Opin. Cell Biol. (1996) 8:731-738; Birkedal-Hansen, et al., Crit. Rev. Oral Biol. Med. (1993) 4:197-250; Mignatti and Rifkin, Physiol. Rev. (1993) 73:161-195; Stetler-Stevenson, et al., Annu. Rev. Cell Biol. (1993) 9:541-573; Brinkerhoff, E., and L. M. Matrisan, Nature Reviews (2002) 3:207-214; Strasser, A., et al., Annu. Rev. Biochem. (2000) 69:217-45; Chao, D. T. and S. J. Korsmeyer, Annu. Rev. Immunol. (1998) 16:395-419; Mullauer, L., et al., Mutat. Res. (2001) 488(3):211-31; Fotedar, R., et al., Prog. Cell Cycle Res. (1996)2:147-63; Reed, J. C., Am. J. Pathol. (2000) 157(5):1415-30; D'Ari, R., Bioassays (2001) 23(7):563-5); genes that express the EGF receptor; Mendelsohn, J. and J. Baselga, Oncogene (2000) 19(56):6550-65; Normanno, N., et al., Front. Biosci. (2001) 6: D685-707); and the multi-drug resistance 1 gene, MDR1 gene (Childs, S., and V. Ling, Imp. Adv. Oncol. (1994) 21-36).
  • In the prevention of disease, the target gene may be one which is required for initiation or maintenance of the disease, or which has been identified as being associated with a higher risk of contracting the disease. In the treatment of disease, the iRNA agent can be brought into contact with the cells or tissue exhibiting the disease. For example, iRNA agent substantially identical to all or part of a mutated gene associated with cancer, or one expressed at high levels in tumor cells, may be brought into contact with or introduced into a cancerous cell or tumor gene.
  • Examples of cellular proliferative and/or differentiative disorders include cancer, e.g., a carcinoma, sarcoma, metastatic disorder or hematopoietic neoplastic disorder, such as a leukemia. A metastatic tumor can arise from a multitude of primary tumor types, including but not limited to those of prostate, colon, lung, breast and liver origin. As used herein, the terms “cancer,” “hyperproliferative,” and “neoplastic” refer to cells having the capacity for autonomous growth, i.e., an abnormal state or condition characterized by rapidly proliferating cell growth. These terms are meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness. Proliferative disorders also include hematopoietic neoplastic disorders, including diseases involving hyperplastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof.
  • The pharmaceutical compositions of the present invention can also be used to treat a variety of immune disorders, in particular those associated with overexpression or aberrant expression of a gene or expression of a mutant gene. Examples of hematopoietic disorders or diseases include, without limitation, autoimmune diseases (including, for example, diabetes mellitus, arthritis (including rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, psoriatic arthritis), multiple sclerosis, encephalomyelitis, myasthenia gravis, systemic lupus erythematosis, automimmune thyroiditis, dermatitis (including atopic dermatitis and eczematous dermatitis), psoriasis, Sjogren's Syndrome, Crohn's disease, aphthous ulcer, iritis, conjunctivitis, keratoconjunctivitis, ulcerative colitis, asthma, allergic asthma, cutaneous lupus erythematosus, scleroderma, vaginitis, proctitis, drug eruptions, leprosy reversal reactions, erythema nodosum leprosum, autoimmune uveitis, allergic encephalomyelitis, acute necrotizing hemorrhagic encephalopathy, idiopathic bilateral progressive sensorineural hearing, loss, aplastic anemia, pure red cell anemia, idiopathic thrombocytopenia, polychondritis, Wegener's granulomatosis, chronic active hepatitis, Stevens-Johnson syndrome, idiopathic sprue, lichen planus, Graves' disease, sarcoidosis, primary biliary cirrhosis, uveitis posterior, and interstitial lung fibrosis), graft-versus-host disease, cases of transplantation, and allergy.
  • In another embodiment, the invention relates to methods for treating viral diseases, including but not limited to hepatitis C, hepatitis B, herpes simplex virus (HSV), HIV-AIDS, poliovirus, and smallpox virus. iRNA agent of the invention are prepared as described herein to target expressed sequences of a virus, thus ameliorating viral activity and replication. The iRNA agents can be used in the treatment and/or diagnosis of viral infected tissue, both animal and plant. Also, such iRNA agent can be used in the treatment of virus-associated carcinoma, such as hepatocellular cancer.
  • For example, the iRNA agent of the present invention are useful for treating a subject having an infection or a disease associated with the replication or activity of a (+) strand RNA virus having a 3′-UTR, such as HCV. In this embodiment, the iRNA agent can act as novel therapeutic agents for inhibiting replication of the virus. The method includes administering a pharmaceutical composition of the invention to the patient (e.g., a human), such that viral replication is inhibited. Examples of (+) strand RNA viruses which can be targeted for inhibition include, without limitation, picornaviruses, caliciviruses, nodaviruses, coronaviruses, arteriviruses, flaviviruses, and togaviruses. Examples of picomaviruses include enterovirus (poliovirus 1), rhinovirus (human rhinovirus 1A), hepatovirus (hepatitis A virus), cardiovirus (encephalomyocarditis virus), aphthovirus (foot-and-mouth disease virus O), and parechovirus (human echovirus 22). Examples of caliciviruses include vesiculovirus (swine vesicular exanthema virus), lagovirus (rabbit hemorrhagic disease virus), “Norwalk-like viruses” (Norwalk virus), “Sapporo-like viruses” (Sapporo virus), and “hepatitis E-like viruses” (hepatitis E virus). Betanodavirus (striped jack nervous necrosis virus) is the representative nodavirus. Coronaviruses include coronavirus (avian infections bronchitis virus) and torovirus (Beme virus). Arterivirus (equine arteritis virus) is the representative arteriviridus. Togavirises include alphavirus (Sindbis virus) and rubivirus (Rubella virus). Finally, the flaviviruses include flavivirus (Yellow fever virus), pestivirus (bovine diarrhea virus), and hepacivirus (hepatitis C virus). In a preferred embodiment, the virus is hepacivirus, the hepatitis C virus. Although the foregoing list exemplifies vertebrate viruses, the present invention encompasses the compositions and methods for treating infections and diseases caused by any (+) strand RNA virus having a 3′-UTR, regardless of the host. For example, the invention encompasses the treatment of plant diseases caused by sequiviruses, comoviruses, potyviruses, sobemovirus, luteoviruses, tombusviruses, tobavirus, tobravirus, bromoviruses, and closteroviruses.
  • The pharmaceutical compositions encompassed by the invention may be administered by any means known in the art including, but not limited to, oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), ocular, rectal, vaginal, and topical (including buccal and sublingual) administration. In preferred embodiments, the pharmaceutical compositions are administered by intravenous or intraparenteral infusion or injection. The pharmaceutical compositions can also be administered intraparenchymally, intrathecally, and/or by stereotactic injection.
  • Methods for Inhibiting Expression of a Target Gene.
  • In yet another aspect, the invention relates to a method for inhibiting the expression of a target gene in a cell or organism. In one embodiment, the method includes administering the inventive iRNA agent or a pharmaceutical composition containing the iRNA agent to a cell or an organism, such as a mammal, such that expression of the target gene is silenced. Because of their surprisingly improved stability and bioavailability, the iRNA agent of the present invention effectively inhibit expression or activity of target genes at surprisingly low dosages. Compositions and methods for inhibiting the expression of a target gene using iRNA agent can be performed as described in the preceding sections, particularly Sections 4 and 5.
  • In this embodiment, a pharmaceutical composition containing the iRNA agent may be administered by any means known in the art including, but not limited to oral or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), ocular, rectal, vaginal, and topical (including buccal and sublingual) administration. In preferred embodiments, the pharmaceutical compositions are administered by intravenous or intraparenteral infusion or injection. The pharmaceutical compositions can also be administered intraparenchymally, intrathecally, and/or by stereotactic injection.
  • The methods for inhibiting the expression of a target gene can be applied to any gene one wishes to silence, thereby specifically inhibiting its expression, provided the cell or organism in which the target gene is expressed includes the cellular machinery which effects RNA interference. Examples of genes which can be targeted for silencing include, without limitation, developmental genes including but not limited to adhesion molecules, cyclin kinase inhibitors, Wnt family members, Pax family members, Winged helix family members, Hox family members, cytokines/lymphokines and their receptors, growth/differentiation factors and their receptors, and neurotransmitters and their receptors; (2) oncogenes including but not limited to ABLI, BCL1, BCL2, BCL6, CBFA2, CBL, CSFIR, ERBA, ERBB, EBRB2, ETS 1, ETS 1, ETV6, FGR, FOS, FYN, HCR, HRAS, JUN, KRAS, LCK, LYN, MDM2, MLL, MYB, MYC, MYCL1, MYCN, NRAS, PIM1, PML, RET, SRC, TAL1, TCL3 and YES; (3) tumor suppresser genes including but not limited to APC, BRCA1, BRCA2, MADH4, MCC, NF1, NF2, RB1, TP53 and WT1; and (4) enzymes including but not limited to ACP desaturases and hydroxylases, ADP-glucose pyrophorylases, ATPases, alcohol dehydrogenases, amylases, amyloglucosidases, catalases, cellulases, cyclooxygenases, decarboxylases, dextrinases, DNA and RNA polymerases, galactosidases, glucanases, glucose oxidases, GTPases, helicases, hemicellulases, integrases, invertases, isomerases, kinases, lactases, lipases, lipoxygenases, lysozymes, pectinesterases, peroxidases, phosphatases, phospholipases, phosphorylases, polygalacturonases, proteinases and peptideases, pullanases, recombinases, reverse transcriptases, topoisomerases, and xylanases.
  • In addition to in vivo gene inhibition, the skilled artisan will appreciate that the iRNA agent of the present invention are useful in a wide variety of in vitro applications. Such in vitro applications, include, for example, scientific and commercial research (e.g., elucidation of physiological pathways, drug discovery and development), and medical and veterinary diagnostics. In general, the method involves the introduction of the iRNA agent into a cell using known techniques (e.g., absorption through cellular processes, or by auxiliary agents or devices, such as electroporation and lipofection), then maintaining the cell for a time sufficient to obtain degradation of an mRNA transcript of the target gene.
  • Methods for Identifying iRNA Agent Having Increased Stability.
  • In yet another aspect, the invention relates to methods for identifying iRNA agent having increased stability in biological tissues and fluids such as serum. iRNA agent having increased stability have enhanced resistance to degradation, e.g., by chemicals or nucleases (particularly endonucleases) which normally degrade RNA molecules. Methods for detecting increases in nucleic acid stability are well known in the art. Any assay capable of measuring or detecting differences between a test iRNA agent and a control iRNA agent in any measurable physical parameter may be suitable for use in the methods of the present invention. In general, because the inhibitory effect of an iRNA agent on a target gene activity or expression requires that the molecule remain intact, the stability of a particular iRNA agent can be evaluated indirectly by observing or measuring a property associated with the expression of the gene. Thus, the relative stability of an iRNA agent can be determined by observing or detecting (1) an absence or observable decrease in the level of the protein encoded by the target gene, (2) an absence or observable decrease in the level of mRNA product from the target gene, and (3) a change or loss in phenotype associated with expression of the target gene. In the context of a medical treatment, the stability of an iRNA agent may be evaluated based on the degree of the inhibition of expression or function of the target gene, which in turn may be assessed based on a change in the disease condition of the patient, such as reduction in symptoms, remission, or a change in disease state.
  • In one embodiment, the method includes preparing an iRNA agent as described above (e.g., through chemical synthesis), incubating the iRNA agent with a biological sample, then analyzing and identifying those iRNA agent that exhibit an increased stability as compared to a control iRNA agent.
  • In an exemplified embodiment, iRNA agent is produced in vitro by mixing/annealing complementary single-stranded RNA strands, preferably in a molar ratio of at least about 3:7, more preferably in a molar ratio of about 4:6, and most preferably in essentially equal molar amounts (e.g., a molar ratio of about 5:5). Preferably, the single-stranded RNA strands are denatured prior to mixing/annealing, and the buffer in which the mixing/annealing reaction takes place contains a salt, preferably potassium chloride. Single-stranded RNA strands may be synthesized by solid phase synthesis using, for example, an Expedite 8909 synthesizer (Applied Biosystems, Applera Deutschland GmbH, Darmstadt, Germany), as described above.
  • iRNA agent are incubated with a biological sample under the conditions sufficient or optimal for enzymatic function. After incubating with a biological sample, the stability of the iRNA agent is analyzed by means conventional in the art, for example using RNA gel electrophoresis as exemplified herein. For example, when the sample is serum, the iRNA agent may be incubated at a concentration of 1-10 μM, preferably 2-8 μM, more preferably 3-6 μM, and most preferably 4-5 μM. The incubation temperature is preferably between 25° C. and 45° C., more preferably between 35° C. and 40° C., and most preferably about 37° C.
  • The biological sample used in the incubation step may be derived from tissues, cells, biological fluids or isolates thereof. For example, the biological sample may be isolated from a subject, such as a whole organism or a subset of its tissues or cells. The biological sample may also be a component part of the subject, such as a body fluid, including but not limited to blood, serum, plasma, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine, vaginal fluid and semen. Preferably, the biological sample is a serum derived from a blood sample of a subject. The subject is preferably a mammal, more preferably a human or a mouse.
  • In another embodiment, the method includes selecting an iRNA agent having increased stability by measuring the mRNA and/or protein expression levels of a target gene in a cell following introduction of the iRNA agent. In this embodiment, an iRNA agent of the invention inhibits expression of a target gene in a cell, and thus the method includes selecting an iRNA agent that induces a measurable reduction in expression of a target gene as compared to a control iRNA agent. Assays that measure gene expression by monitoring RNA and/or protein levels can be performed within about 24 hours following uptake of the iRNA agent by the cell. For example, RNA levels can be measured by Northern blot techniques, RNAse Protection Assays, or Quality Control-PCR (QC-PCR) (including quantitative reverse transcription coupled PCR (RT-PCR)) and analogous methods known in the art. Protein levels can be assayed, for example, by Western blot techniques, flow cytometry, or reporter gene expression (e.g., expression of a fluorescent reporter protein, such as green fluorescent protein (GFP)). RNA and/or protein levels resulting from target gene expression can be measured at regular time intervals following introduction of the test iRNA agent, and the levels are compared to those following introduction of a control iRNA agent into cells. A control iRNA agent can be a nonsensical iRNA agent (i.e., an iRNA agent having a scrambled sequence that does not target any nucleotide sequence in the subject), an iRNA agent that can target a gene not present in the subject (e.g., a luciferase gene, when the iRNA agent is tested in human cells), or an iRNA agent otherwise previously shown to be ineffective at silencing the target gene. The mRNA and protein levels of the test sample and the control sample can be compared. The test iRNA agent is selected as having increased stability when there is a measurable reduction in expression levels following absorption of the test iRNA agent as compared to the control iRNA agent. mRNA and protein measurements can be made using any art-recognized technique (see, e.g., Chiang, M. Y., et al., J. Biol. Chem. (1991) 266:18162-71; Fisher, T, et al., Nucl. Acids Res. (1993) 21:3857; and Chen et al., J. Biol. Chem. (1996)271:28259).
  • The ability of an iRNA agent composition of the invention to inhibit gene expression can be measured using a variety of techniques known in the art. For example, Northern blot analysis can be used to measure the presence of RNA encoding a target protein. The level of the specific mRNA produced by the target gene can be measured, e.g., using RT-PCR. Because iRNA agent directs the sequence-specific degradation of endogenous mRNA through RNAi, the selection methods of the invention encompass any technique that is capable of detecting a measurable reduction in the target RNA. In yet another example, Western blots can be used to measure the amount of target protein present. In still another embodiment, a phenotype influenced by the amount of the protein can be detected. Techniques for performing Western blots are well known in the art (see, e.g., Chen, et al., J. Biol. Chem. (1996) 271:28259).
  • When the target gene is to be silenced by an iRNA agent that targets a promoter sequence of the target gene, the target gene can be fused to a reporter gene, and reporter gene expression (e.g., transcription and/or translation) can be monitored. Similarly, when the target gene is to be silenced by an iRNA agent that targets a sequence other than a promoter, a portion of the target gene (e.g., a portion including the target sequence) can be fused with a reporter gene so that the reporter gene is transcribed. By monitoring a change in the expression of the reporter gene in the presence of the iRNA agent, it is possible to determine the effectiveness of the iRNA agent in inhibiting the expression of the reporter gene. The expression levels of the reporter gene in the presence of the test iRNA agent versus a control iRNA agent are then compared. The test iRNA agent is selected as having increased stability when there is a measurable reduction in expression levels of the reporter gene as compared to the control iRNA agent. Examples of reporter genes useful for use in the present invention include, without limitation, those coding for luciferase, GFP, chloramphenicol acetyl transferase (CAT), β-galactosidase, and alkaline phosphatase. Suitable reporter genes are described, for example, in Current Protocols in Molecular Biology, John Wiley & Sons, New York (Ausubel, F. A., et al., eds., 1989); Gould, S. J., and S. Subramani, Anal. Biochem. (1988) 7:404-408; Gorman, C. M., et al., Mol. Cell. Biol. (1982) 2:1044-1051; and Selden, R., et al., Mol. Cell. Biol. (1986) 6:3173-3179; each of which is hereby incorporated by reference.
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
  • EXAMPLES Example 1 Terminal Modifications Protect Sites of Exonucleolytic Cleavage
  • Serum Incubation Assay
  • Blood of 8 human volunteers (270 mL) was collected and kept at room temperature for 3 hours. The blood pool was then centrifuged at 20° C. and 3000 rcf using Megafuge 1.0 (Heraeus Instruments, Kendro Laboratory Products GmbH, Langenselbold) to separate the serum from the cellular fraction. The supernatant was stored in aliquots at −20° C. and used as needed. Human serum obtained from Sigma (Sigma-Aldrich Chemie GmbH, Taufkirchen, Germany) was employed in control assays. Assay results reported herein were consistent among the different serum sources tested.
  • Double stranded RNAs (300 pmol, ca. 4.2 μg) were incubated at 37° C. in 60 μl 90% human serum/10% phosphate buffered saline for 0, 15, 30, 60, 120 or 240 minutes, or 8 hours, 16 hours or 24 hours. A 10 μl sample was taken from the incubation mixture, frozen in liquid nitrogen and stored at −80° C. until analysis.
  • Analysis by Electrophoresis and “Stains all” Detection
  • For analysis, samples were thawed, their constituents were isolated by phenol-extraction and ethanol-precipitation, separated on denaturing 14% polyacrylamide gels (6M Urea, 20% formamide, Carl Roth GmbH & Co KG Karlsruhe, Germany) and detected by staining with the “stains-all” reagent (Sigma-Aldrich Chemie GmbH, Steinheim, Germany). Reaction mixtures from incubation of selected dsRNAs were further analysed covering the time points 8 hours, 16 hours and 24 hours.
  • Cell Culture
  • HeLa SS6 cells were grown at 37° C. in Dulbecco's modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 units/mL penicillin, and 100 μg/mL streptomycin (Invitrogen, Carlsbad, Calif.). Cells were passaged regularly to maintain exponential growth. Twenty-four hours prior to siRNA transfection, cells were seeded on opaque, white 96-well plates (Costar, Corning, N.Y.) at a concentration of 15,000 cells/well in 150 μL antibiotic-free, phenol red-free DMEM (Invitrogen).
  • Dual Luciferase Gene Silencing Assays
  • In vitro activity of siRNAs was determined using a high-throughput 96-well plate format luciferase silencing assay. Cells were first transiently transfected with plasmids encoding firefly (target) and renilla (control) luciferase. DNA transfections were performed using Lipofectamine 2000 (Invitrogen) (0.5 μL/μg total DNA) and the plasmids gWiz-Luc (Aldevron, Fargo, N.Dak.) (200 ng/well) and pRL-CMV (Promega, Madison, Wis.) (200 ng/well). After 2 h, the plasmid transfection medium was removed, and the firefly luciferase targeting siRNAs were added to the cells at 100 nM concentration. siRNA transfections were performed using TransIT-TKO (Mirus, Madison, Wis.) (0.3 mL/well). After 24 h, cells were analyzed for both firefly and renilla luciferase expression using a plate luminometer (VICTOR2, PerkinElmer, Boston, Mass.) and the Dual-Glo Luciferase Assay kit (Promega). Firefly/renilla luciferase expression ratios were used to determine percent gene silencing relative to mock-treated (no siRNA) controls.
  • Results
  • dsRNAs including phosphorothioate modifications were incubated in 90% human serum at 37° C. as described above. Gel electropheresis of the RNA products revealed that the phosphorothioate modifications enhanced stability (FIG. 1). Similar results were observed when 3′-Naproxen, 5-Nitroindole, alkyamino-dT, abasic pyrrolidine, and cationic C6 amino dT were conjugated to the 3′ end of dsRNAs (FIGS. 2, 3, and 4).
  • The dual luciferase assay revealed that modifications than enhanced resistance to exonuclease activity retained silencing activity. FIGS. 5, 6, and 7 show the silencing activity of 3′ conjugated Naproxen, abasic pyrrolidine, and cationic C6 amino dT, respectively.
  • FIGS. 8 and 9 show that the 3′Naproxen conjugated dsRNAs had enhanced exonuclease resistance in mouse serum and liver tissue, as well as in human serum (see FIG. 2).
  • Example 2 siRNA Modifications Enhanced Duplex Stability
  • Radiolabel method for monitoring serum stability of siRNA duplexes: siRNA duplexes were prepared at a stock concentration of 1 μM in which either the sense (S) or antisense strand (AS) contained a trace amount of 5′-32P labeled material (e.g. 32P-S/AS and S/32P-AS). The presence of the end-labeled sense or antisense strand allowed for monitoring of the individual strand within the context of the siRNA duplex. Therefore, two duplex preparations were made for each siRNA sequence tested. siRNA duplexes were incubated in 90% human serum at a final concentration of 100 nM duplex. Samples were removed and quenched in a stop mix at appropriate times. For a typical time course, 10 seconds, 15 minutes, 30 minutes, 1 hour, 2 hours and 4 hours time points were taken. Samples were analyzed by denaturing polyacrylamide gel electrophoresis along with a control sample (4 hour buffer-alone incubation) and a partial alkaline hydrolysis ladder of the labeled sense or antisense strand as a marker. The gel was imaged using a Fuji phosphorimager to detect the full length sense and antisense strands along with any degradation fragments that were generated by serum nucleases during incubation.
  • Since there is the possibility of losing the 5′ phosphate label due to phosphatase activity in the serum, an alternative to 5′ end labeling is to place an internal 32P or 33P label within either the sense or antisense strand. This labeling method is much more laborious than 5′ end labeling and currently we have no evidence that dephosphorylation occurs during serum incubation.
  • A series of chemical modifications that fall into the following categories; backbone modification, sugar modification, nucleobase modification and 3′ conjugate, were tested and showed enhanced serum stability as compared to a unmodified siRNA duplex. A description of each modification, its location within the siRNA duplex, and the serum stability data follows.
  • Serum stability of unmodified parent duplex: The unmodified parent duplex, AL-DUP-1000, was used to establish the serum stability baseline for evaluating the effect of chemical modifications on nuclease resistance.
    AL-DUP-1000
       5′-CUUACGCUGAGUACUUCGAdTdT-3′ ALN-SEQ-1000 SEQ ID NO:
    3′ dTdTGAAUGCGACUCAUGAAGCU-5′ ALN-SEQ-1001 SEQ ID NO:
  • AL-DUP-1000 was subjected to the serum stability assay to evaluate its inherent nuclease resistance and to define its degradation pattern (FIG. 21). Denaturing gel electrophoresis was used analyze AL-DUP-1000 in a human serum stability assay. An siRNA duplex containing 5′ end-labeled sense RNA (*s/as) and a duplex containing 5′ end-labeled antisense RNA (as/s*) were each incubated in 90% human serum and time points were assayed at 10 seconds, 5 min, 15 min, 30 min, 1 hour, 2 hours and 4 hours. The control was a 4 hour time point for siRNA duplex incubated in PBS buffer alone, OH-was the partial alkaline hydrolysis marker. This unmodified duplex was observed to be degraded by both 3′-5′ exonucleases and endonucleases (FIG. 21).
  • Cleavage of the 3′ end of both the sense and antisense strands by 3′-5′ exonucleases occurs within the first 5 minutes of incubation resulting in the loss of the 3′ terminal dT residues (top vertical lines in s*/as and s/as* panels of FIG. 21). In addition to exonuclease degradation, both strands were cleaved by endonucleases. There was a major endonuclease site at position sixteen of the antisense strand (bottom vertical lines in s*/as and s/as* panels of FIG. 21) that appears as early as 10 seconds. Very little full length sense or antisense strand was remaining after 1 hour in human serum. Chemical modifications were introduced within the context of the parent duplex to evaluate their effect on nuclease resistance. These chemical modifications fall within one of the following classes: backbone modification, sugar modification, nucleobase modification, cationic modification and conjugate.
  • Backbone modifications enhanced nuclease resistance: Specific phophodiester linkages of the siRNA duplex were replaced by either phosphorothioate or methylphosphonate and their stability was evaluated in the human serum stability assay. Table 1 contains the sequences of the duplexes tested. Substitution of the phosphodiester linkage at the 3′ end of both the sense and antisense strands inhibited exonucleolytic degradation of the 3′ overhangs (FIGS. 22A and 22B) as compared to the unmodified parent duplex (refer to FIG. 21). Full length starting material was present for four hours for both the sense and antisense strands. The endonucleolytic cleavage pattern seen in the unmodified duplex was unchanged. Similar results were obtained for duplexes that contained additional phosphorothioates at their 3′ ends (data not shown). The placement of phosphorothioates at the endonucleolytic cleavage sites (duplexes 1419, 1420 and 1421) did not inhibit endonucleolytic cleavage at these sites (data not shown). In summary, a single phosphorothioate or methylphosphonate between the two 3′ terminal nucleotides was sufficient to protect the 3′ ends from exonuclease degradation. Additional phosphorothioates at the 3′ ends appear to enhance this effect, which may be necessary for long term exposure to serum nucleases.
    TABLE 1
    siRNA duplexes containing backbone modifications.
    (* = phosphorothioate, mp = methylphosphonate)
    Alnylam Duplex Duplex Sequence Alnylam Sequence
    AL-DUP-1393     5′-CUUACGCUGAGUACUUCGAdT*dT-3′ AL-SEQ-1026
    3′-dT*dTGAAUGCGACUCAUGAAGCU-5′ AL-SEQ-1027
    AL-DUP-1394       5′-CUUACGCUGAGUACUUCGA*dT*dT-3′ AL-SEQ-1028
    3′-dT*dT*GAAUGCGACUCAUGAAGCU-5′ AL-SEQ-1029
    AL-DUP-1395       5′-CUUACGCUGAGUACUUCG*A*dT*dT-3′ AL-SEQ-1030
    3′-dT*dT*G*AAUGCGACUCAUGAAGCU-5′ AL-SEQ-1031
    AL-DUP-1396       5′-CUUACGCUGAGUACUUC*G*A*dT*dT-3′ AL-SEQ-1032
    3′-dT*dT*G*A*AUGCGACUCAUGAAGCU-5′ AL-SEQ-1033
    AL-DUP-1419     5′-CUUACGCUGAGU*ACUUCGAdTdT-3′ AL-SEQ-2182
    3′-dTdTGAAUGCGACUCA*UGAAGCU-5′ AL-SEQ-2184
    AL-DUP-1420     5′-CUU*ACGCUGAGU*ACUUCGAdTdT-3′ AL-SEQ-2183
    3′-dTdTGAA*UGCGACUCA*UGAAGCU-5′ AL-SEQ-2185
    AL-DUP-1421      5′-CUU*ACGCUGAGU*ACUUCGAdT*dT-3′ AL-SEQ-2186
    3′-dT*dTGAA*UGCGACUCA*UGAAGCU-5′ AL-SEQ-2188
    AL-DUP-1329     5′-CUUACGCUGAGUACUUCGAdTmpdT-3′ AL-SEQ-1038
    3′-dTmpdTGAAUGCGACUCAUGAAGCU-5′ AL-SEQ-1039
  • Sugar modifications enhanced nuclease resistance: The effect of replacing the 2′OH with 2′OMe was evaluated at the sites of endonucleolytic cleavage as well as at the 3′ ends of the siRNA duplex. The duplexes tested in the human serum stability assay are shown in Table 2. Some of these duplexes also contained phosphorothioate linkages to evaluate whether the combination of the two modifications enhance nuclease resistance more significantly. Substitution of the terminal dT residues with 2′OMe-U (AL-DUP-1027) reduced 3′-5′ exonuclease degradation slightly over the unmodified parent duplex (data not shown); however, the extent of exonuclease protection by 2′OMe-U was far less than that achieved by placing a
    TABLE 2
    siRNA duplexes containing 2′OMe substitutions.
    (U = 2′OMe-uridine, * = phosphorothioate)
    Alnylam Duplex Duplex Sequence Alnylam Sequence
    AL-DUP-1027   5′-CUUACGCUGAGUACUUCGAUU-3′ AL-SEQ-1006
    3′-UUGAAUGCGACUCAUGAAGCU-5′ AL-SEQ-1007
    AL-DUP-1036    5′-C*UUACGCUGAGUACUUCGAU*U-3′ AL-SEQ-1008
    3′-U*UGAAUGCGACUCAUGAAGC*U-5′ AL-SEQ-1009
    AL-DUP-13ff    5′-C*UUACGCUGAGUACUUCGAU*U-3′ AL-SEQ-gggg
    3′-U*UGAAUGCGACUCAUGAAGC*U-5′ AL-SEQ-hhhh
    AL-DUP-1363    5′-C*UUACGCUGAGUACUUCGAU*U-3′ AL-SEQ-1162
    3′-U*UGAAUGCGACUCAUGAAGC*U-5′ AL-SEQ-1163

    phosphorothioate between the two terminal dT residues (see FIG. 22A). Addition of a single phosphorothioate between the two terminal 2′OMe-uridine residues effectively inhibited 3′-5′ exonucleolytic cleavage as seen in FIG. 23 for duplexes AL-DUP-1036, AL-DUP-13ff, and AL-DUP-1363. 2′OMe substitution on its own was much more effective at protecting from endonucleolytic cleavage when placed at the internal cleavage sites. The parent duplex was cleaved 3′ of U at two UpA sites within the duplex. Both strands are cleaved due to the symmetry of this dinucleotide repeat and mapping data was used to confirm the sites of cleavage (data not shown). Placement of 2′OMe at the strong endonucleolytic site ((FIG. 23, star in s/*as gel, AL-DUP-13ff) resulted in inhibition of cleavage at this site. The second, weaker endonucleolytic site (FIG. 23, black star in *s/as), however, was slightly enhanced when the strong site was protected with 2′OMe (FIG. 23, compare AL-DUP-13ff to AL-DUP-1036). Protection of both sites with 2′OMe (AL-DUP-1363) resulted in reduced endonucleolytic cleavage at both sites (FIG. 23). The inhibitory effect of the 2′OMe substitution is consistent with the mechanism of endonucleolytic cleavage, which requires the 2′OH as a nucleophile in the cleavage reaction. 2′OMe modification will also be an effective means to protect the 3′ overhang of single overhang siRNA duplexes where the 3′ overhang is composed of ribonucleotides. In this situation, 2′OMe substitution can be used to block the possible loss of the terminal two nucleotides by endonucleolytic cleavage and phosphorothioate can be used to protect from exonuclease degradation.
  • Cationic modifications enhanced nuclease resistance: The effect of three different cationic chemical modifications on nuclease resistance was tested and compared to the parent unmodified duplex. The structures of the three cationic modifications tested are shown below.
    Figure US20050119214A1-20050602-C00092
    TABLE 3
    siRNA duplexes containing cationic substitutions.
    (aadT = alkylamine-dT, AbP = abasic pyrrolidine cationic,
    aaU = allylamino-U, * = phosphorothioate, U = 2′OMe-U)
    Alnylam Duplex Duplex Sequence Alnylam Sequence
    AL-DUP-10aa       5′-CUUACGCUGAGUACUUCGAdTaadT-3′ AL-SEQ-1017
    3′-aadTdTGAAUGCGACUCAUGAAGCU-5′ AL-SEQ-1018
    AL-DUP-10bb         5′-CUUACGCUGAGUACUUCGAaadTaadT-3′ AL-SEQ-1015
    3′-aadTaadTGAAUGCGACUCAUGAAGCU-5′ AL-SEQ-1016
    AL-DUP-1ccc        5′-CUUACGCUGAGUACUUCGAdTdTAbP-3′ AL-SEQ-dddd
    3′-AbPdTdTGAAUGCGACUCAUGAAGCU-5′ AL-SEQ-eeee
    AL-DUP-1403    5′-C*UaaUACGCUGAGUACUUCGAU*U-3′ AL-SEQ-2080
    3′-U*UGAAaaUGCGACUCAUGAAGC*U-5′ AL-SEQ-2081
    AL-DUP-1406    5′-C*UaaUACGCUGAGaaUACUUCGAU*U-3′ AL-SEQ-2082
    3′-U*UGAAaaUGCGACUCAaaUGAAGC*U-5′ AL-SEQ-2083

    The sequences of the duplexes assayed in the human serum stability assay are shown in Table 3. Both alkylamino-dT and abasic pyrrolidine cationic modifications were placed at the 3′ terminal overhang to evaluate their effect on 3′-5′ exonuclease degradation. Allylamino-uridines were placed at the internal endonucleolytic cleavage sites to evaluate their ability to inhibit endonucleolytic cleavage. As seen in FIG. 24, replacing the 3′ terminal dT residue with a single alkylamino-dT efficiently inhibited 3′-5′ exonucleolytic degradation (FIG. 24, AL-DUP-10aa, left gel image). Replacement of both dT residues in the overhang with alkylamino-dT resulted in a similar extent of inhibition (data not shown). Addition of an abasic pyrrolidine cationic modification at the 3′ terminus of each strand also protected against exonucleolytic degradation (FIG. 24, middle gel image). Both the alkylamino-dT and abasic pyrrolidine modifications protected from 3′-5′ exonucleolytic cleavage up to 23 hours (data not shown). Placement of allylamino-U at the internal cleavage sites inhibited endonucleolytic cleavage as shown in FIG. 24 for duplex AL-DUP-1403. The ends of this duplex were stabilized from exonucleolytic degradation by 2′OMe-U and phosphorothioate substitutions in order to separate the two different cleavage events. Endonucleolytic cleavage was inhibited at both internal cleavage sites by allylamino-U substitution for AL-DUP-1406 (data not shown).
  • 3′ conjugates enhanced nuclease resistance: Conjugation of naproxen and ibuprofen to the 3′ end of the siRNA were tested for their ability to inhibit 3′-5′ exonucleolytic degradation. The structure of naproxen is shown in below:
    Figure US20050119214A1-20050602-C00093
  • Table 4 lists the siRNAs that were tested in the human serum stability assay. Conjugation of either naproxen or ibuprofen to the 3′ end inhibited exonucleolytic degradation. FIG. 18 shows the serum stability data for the naproxen modified duplex (AL-DUP-1069) and similar results were obtained for AL-DUP1413. Presumably the conjugates inhibit exonucleolytic cleavage by sterically blocking the exonuclease from binding to the 3′ end of the siRNA duplex. Similar data was also obtained for AL-DUP-1069 in pooled mouse serum.
    TABLE 4
    siRNA duplexes containing 3 conjugates.
    (Nap = Naproxen, Ibu = Ibuprofen)
    Alnylam Duplex Duplex Sequence SEQ ID NO:
    AL-DUP-1069        5′-CUUACGCUGAGUACUUCGAdTdTNap-3′
    3′-NapdTdTGAAUGCGACUCAUGAAGCU-5′
    AL-DUP-1413        5′-CUUACGCUGAGUACUUCGAdTdTIbu-3′
    3′-NapdTdTGAAUGCGACUCAUGAAGCU-5′
  • A number of embodiments of the invention have been described. Nevertheless, it will be understood that various modifications may be made without departing from the spirit and scope of the invention. Other embodiments are in the claims.

Claims (35)

1. An iRNA agent comprising at least one ribonucleotide modification, wherein the modification confers improved exonuclease stability to the iRNA agent when the iRNA agent is contacted with a biological sample.
2. The iRNA agent of claim 1, wherein the iRNA agent comprises at least two ribonucleotide modifications.
3. The iRNA agent of claim 1, wherein the iRNA agent comprises at least three ribonucleotide modifications.
4. The iRNA agent of claim 1, wherein the iRNA agent comprises at least four ribonucleotide modifications.
5. The iRNA agent of claim 1, wherein the iRNA agent comprises at least five ribonucleotide modifications.
6. The iRNA agent of claim 1, wherein biological sample comprises serum.
7. The iRNA agent of claim 1, wherein the modification comprises a backbone modification between one or more of the terminal nucleotides on an iRNA strand of the iRNA agent.
8. The iRNA agent of claim 7, wherein the backbone modification comprises a phosphorothioate linkage between one or more of the terminal nucleotides of an iRNA agent.
9. The iRNA agent of claim 1, wherein the modification comprises a modification of a nucluotide sugar.
10. The iRNA agent of claim 9, wherein the nucleotide is a terminal nucleotide.
11. The iRNA agent of claim 10, wherein the terminal nucleotide comprises a 2′ or 3′ sugar modification.
12. The iRNA agent of claim 10, wherein the iRNA agent comprises a 2′-O-methylated nucleotide, a 2′-deoxy nucleotide, a 2′-deoxyfluoro nucleotide, a 2′-O-methoxyethyl nucleotide, a 2′-O-NMA nucleotide, a 2′-DMAEOE nucleotide, a 2′-AP nucleotide, 2′-hydroxy nucleotide, a 2′-ara-fluoro nucleotide, a locked nucleic acid, an extended nucleic acid, hexose nucleic acid, or cyclohexene nucleic acid.
13. The iRNA agent of claim 12, wherein the iRNA agent comprises a 2′-O-methylated nucleotide.
14. The iRNA agent of claim 12, wherein the iRNA agent comprises a 2′-deoxyfluoro nucleotide.
15. The iRNA agent of claim 12, wherein the iRNA agent comprises a locked nucleic acid and the locked nucleic acid is on a 5′ or 3′ end of a sense strand of the iRNA agent, and not on a 5′ or 3′ end of an antisense strand of the iRNA agent.
16. The iRNA agent of claim 11, wherein the iRNA agent comprises a 3′-O-methylated nucleotide.
17. The iRNA agent of claim 11, wherein the 3′-O-methylated nucleotide is on a 3′ or 5′ end of a sense strand of the iRNA agent.
18. The iRNA agent of claim 1, wherein the modification comprises a nucleobase modification.
19. The iRNA agent of claim 18, wherein the iRNA agent comprises a 5′-methyl-pyrimidine.
20. The iRNA agent of claim 19, wherein the iRNA agent comprises a 5′-methyl-uridine.
21. The iRNA agent of claim 19, wherein the iRNA agent comprises a 5′-methyl-cytodine.
22. The iRNA agent of claim 18, wherein the iRNA agent comprises at least one terminal nucleotide having a nucleobase modification.
23. The iRNA agent of claim 22, wherein the iRNA agent comprises a 3′-abasic cationic modification.
24. The iRNA agent of claim 23, wherein the cationic modification comprises an alkylamino-dT, an allylamino conjugate, a pyrrolidine conjugate, a pthalamido or a hydroxyprolinol conjugate, on one or more of the terminal nucleotides of the iRNA agent.
25. The iRNA agent of claim 1, wherein the iRNA agent comprises a comprises a backbone modification between one or more of the terminal nucleotides on an iRNA strand of the iRNA agent and a terminal nucleotide having a 2′ or 3′ sugar modification.
26. The iRNA agent of claim 1, wherein the iRNA agent comprises a terminal nucleotide having a nucleobase modification and a terminal nucleotide having a 2′ or 3′ sugar modification.
27. The iRNA agent of claim 1, wherein the iRNA agent comprises at least one conjugate.
28. The iRNA agent of claim 27, wherein the conjugate is attached to one or more terminal nucleotides of the iRNA agent.
29. The iRNA agent of claim 27, wherein the conjugate is selected from a lipophile, a terpene, a protein binding agent, a vitamin, a carbohydrate, or a peptide.
30. The iRNA agent of claim 27, wherein the conjugate is on a 3′ end of an antisense strand of the iRNA agent, or on a 5′ or 3′ end of a sense strand on the iRNA agent.
31. The iRNA agent of claim 30, the conjugate is not on the 3′ end of the antisense strand and on the 3′ end of the sense strand.
32. The iRNA agent of claim 27, wherein conjugate is naproxen.
32. The iRNA agent of claim 27, wherein the conjugate is cholesterol.
33. The iRNA agent of claim 32, wherein the cholesterol is conjugated to the iRNA agent by a pyrrolidine linker, or serinol linker, or hydroxyprolinol linker.
34. The iRNA agent of claim 27, wherein the conjugate is cholanic acid.
US10/936,115 2003-04-17 2004-09-07 Nuclease resistant double-stranded ribonucleic acid Abandoned US20050119214A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/936,115 US20050119214A1 (en) 2003-04-17 2004-09-07 Nuclease resistant double-stranded ribonucleic acid

Applications Claiming Priority (21)

Application Number Priority Date Filing Date Title
US46377203P 2003-04-17 2003-04-17
US46580203P 2003-04-25 2003-04-25
US46566503P 2003-04-25 2003-04-25
US46961203P 2003-05-09 2003-05-09
US49398603P 2003-08-08 2003-08-08
US49459703P 2003-08-11 2003-08-11
US50341403P 2003-09-15 2003-09-15
US50634103P 2003-09-26 2003-09-26
US51024603P 2003-10-09 2003-10-09
US51031803P 2003-10-10 2003-10-10
US51845303P 2003-11-07 2003-11-07
PCT/US2004/007070 WO2004080406A2 (en) 2003-03-07 2004-03-08 Therapeutic compositions
WOPCT/US04/07070 2004-03-08
WOPCT/US04/10586 2004-04-05
PCT/US2004/010586 WO2004090108A2 (en) 2003-04-03 2004-04-05 Irna conjugates
PCT/US2004/011255 WO2004091515A2 (en) 2003-04-09 2004-04-09 iRNA CONJUGATES
WOPCT/US04/11255 2004-04-09
WOPCT/US04/11822 2004-04-16
PCT/US2004/011829 WO2004094595A2 (en) 2003-04-17 2004-04-16 MODIFIED iRNA AGENTS
PCT/US2004/011822 WO2004094345A2 (en) 2003-04-17 2004-04-16 Protected monomers
US10/936,115 US20050119214A1 (en) 2003-04-17 2004-09-07 Nuclease resistant double-stranded ribonucleic acid

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/011829 Continuation-In-Part WO2004094595A2 (en) 2003-04-17 2004-04-16 MODIFIED iRNA AGENTS

Publications (1)

Publication Number Publication Date
US20050119214A1 true US20050119214A1 (en) 2005-06-02

Family

ID=35530789

Family Applications (7)

Application Number Title Priority Date Filing Date
US10/916,185 Active 2025-06-09 US7745608B2 (en) 2003-04-17 2004-08-10 Modified iRNA agents
US10/936,115 Abandoned US20050119214A1 (en) 2003-04-17 2004-09-07 Nuclease resistant double-stranded ribonucleic acid
US10/946,873 Abandoned US20050164235A1 (en) 2003-04-17 2004-09-21 Modified iRNA agents
US12/619,382 Active 2025-09-14 US8344125B2 (en) 2003-04-17 2009-11-16 Modified iRNA agents
US12/714,298 Active 2025-08-27 US8507661B2 (en) 2003-04-17 2010-02-26 Modified iRNA agents
US16/523,411 Active 2024-08-22 US11312957B2 (en) 2003-04-17 2019-07-26 Modified iRNA agents
US17/697,685 Pending US20220403377A1 (en) 2003-04-17 2022-03-17 MODIFIED iRNA AGENTS

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/916,185 Active 2025-06-09 US7745608B2 (en) 2003-04-17 2004-08-10 Modified iRNA agents

Family Applications After (5)

Application Number Title Priority Date Filing Date
US10/946,873 Abandoned US20050164235A1 (en) 2003-04-17 2004-09-21 Modified iRNA agents
US12/619,382 Active 2025-09-14 US8344125B2 (en) 2003-04-17 2009-11-16 Modified iRNA agents
US12/714,298 Active 2025-08-27 US8507661B2 (en) 2003-04-17 2010-02-26 Modified iRNA agents
US16/523,411 Active 2024-08-22 US11312957B2 (en) 2003-04-17 2019-07-26 Modified iRNA agents
US17/697,685 Pending US20220403377A1 (en) 2003-04-17 2022-03-17 MODIFIED iRNA AGENTS

Country Status (8)

Country Link
US (7) US7745608B2 (en)
EP (5) EP2660322A3 (en)
JP (1) JP4597976B2 (en)
AU (2) AU2004233092C9 (en)
CA (1) CA2522637C (en)
DK (1) DK1620544T3 (en)
ES (1) ES2702942T3 (en)
WO (1) WO2004094595A2 (en)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030166282A1 (en) * 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
US20040033602A1 (en) * 2002-06-12 2004-02-19 Ambion, Inc. Methods and compositions relating to polypeptides with RNase III domains that mediate RNA interference
US20040248094A1 (en) * 2002-06-12 2004-12-09 Ford Lance P. Methods and compositions relating to labeled RNA molecules that reduce gene expression
US20050164235A1 (en) * 2003-04-17 2005-07-28 Muthiah Manoharan Modified iRNA agents
US20060025366A1 (en) * 2004-07-02 2006-02-02 Protiva Biotherapeutics, Inc. Immunostimulatory siRNA molecules and uses therefor
US20060142228A1 (en) * 2004-12-23 2006-06-29 Ambion, Inc. Methods and compositions concerning siRNA's as mediators of RNA interference
US20060287260A1 (en) * 2004-06-30 2006-12-21 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a non-phosphate backbone linkage
US20070054873A1 (en) * 2005-08-26 2007-03-08 Protiva Biotherapeutics, Inc. Glucocorticoid modulation of nucleic acid-mediated immune stimulation
US20070135372A1 (en) * 2005-11-02 2007-06-14 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
WO2008067034A1 (en) * 2006-11-28 2008-06-05 Laird Technologies, Inc. Vehicle-mount antenna assemblies having snap-on outer cosmetic covers with compliant latching mechanisms for achieving zero-gap
US20080249046A1 (en) * 2006-06-09 2008-10-09 Protiva Biotherapeutics, Inc. MODIFIED siRNA MOLECULES AND USES THEREOF
US20090017124A1 (en) * 2007-04-17 2009-01-15 Baxter International Inc. Nucleic Acid Microparticles for Pulmonary Delivery
US20090298913A1 (en) * 2005-10-28 2009-12-03 Topigen Pharmaceuticals Inc. Small interfering oligonucleotides comprising arabinose modified nucleotides
US20090306178A1 (en) * 2006-03-27 2009-12-10 Balkrishen Bhat Conjugated double strand compositions for use in gene modulation
WO2010028079A2 (en) * 2008-09-02 2010-03-11 Alnylam Pharmaceuticals, Inc Synthetic methods and derivatives of triphosphate oligonucleotides
US20100075423A1 (en) * 2002-06-12 2010-03-25 Life Technologies Corporation Methods and compositions relating to polypeptides with rnase iii domains that mediate rna interference
US20100076056A1 (en) * 2003-04-17 2010-03-25 Alnylam Pharmaceuticals, Inc. MODIFIED iRNA AGENTS
US20100222414A1 (en) * 2007-09-19 2010-09-02 Applied Biosystems, Llc SiRNA Sequence-Independent Modification Formats for Reducing Off-Target Phenotypic Effects in RNAi, and Stabilized Forms Thereof
US20100267941A1 (en) * 2003-04-17 2010-10-21 Alnylam Pharmaceuticals, Inc. Irna agents with biocleavable tethers
US7851615B2 (en) 2003-04-17 2010-12-14 Alnylam Pharmaceuticals, Inc. Lipophilic conjugated iRNA agents
US20110009477A1 (en) * 2009-07-08 2011-01-13 Idera Pharmaceuticals, Inc. Oligonucleotide-based compounds as inhibitors of toll-like receptors
US20110028531A1 (en) * 2008-03-20 2011-02-03 Elena Feinstein Novel sirna compounds for inhibiting rtp801
US20110097707A1 (en) * 2004-07-21 2011-04-28 Alnylam Pharmaceuticals, Inc. RNAi Agents Comprising Universal Nucleobases
US20110189300A1 (en) * 2004-11-17 2011-08-04 Protiva Biotherapeutics, Inc. siRNA SILENCING OF APOLIPOPROTEIN B
US20130130378A1 (en) * 2010-04-22 2013-05-23 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
US8796436B2 (en) 2003-04-17 2014-08-05 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
US8815821B2 (en) 2002-02-01 2014-08-26 Life Technologies Corporation Double-stranded oligonucleotides
US20150322432A1 (en) * 2012-12-12 2015-11-12 1Massachusetts Institute Of Technology Compositions and methods for functional nucleic acid delivery
US9777275B2 (en) 2002-02-01 2017-10-03 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency

Families Citing this family (293)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7833992B2 (en) * 2001-05-18 2010-11-16 Merck Sharpe & Dohme Conjugates and compositions for cellular delivery
US7875612B2 (en) 2001-04-24 2011-01-25 Purdue Research Foundation Folate mimetics and folate-receptor binding conjugates thereof
US7910594B2 (en) * 2002-05-15 2011-03-22 Endocyte, Inc. Vitamin-mitomycin conjugates
TWI492750B (en) 2003-01-27 2015-07-21 安德賽特公司 Vitamin-receptor binding drug delivery conjugates, pharmaceutical compositions, uses and preparation processes
US20050233342A1 (en) * 2003-03-07 2005-10-20 Muthiah Manoharan Methods of preventing off-target gene silencing
US20050013855A1 (en) * 2003-04-09 2005-01-20 Biodelivery Sciences International, Inc. Cochleate compositions directed against expression of proteins
EP1660657A1 (en) * 2003-08-28 2006-05-31 Novartis AG Interfering rna duplex having blunt-ends and 3'-modifications
WO2005038013A1 (en) 2003-10-07 2005-04-28 Isis Pharmaceuticals, Inc. Artisense oligonucleotides optimized for kidney targeting
US20050191653A1 (en) * 2003-11-03 2005-09-01 Freier Susan M. Modulation of SGLT2 expression
CA2544349C (en) 2003-11-04 2020-02-18 Geron Corporation Rna amidates and thioamidates for rnai
US20090280567A1 (en) * 2004-02-06 2009-11-12 Dharmacon, Inc. Stabilized sirnas as transfection controls and silencing reagents
JP4937899B2 (en) 2004-03-12 2012-05-23 アルナイラム ファーマシューティカルズ, インコーポレイテッド IRNA substances targeting VEGF
KR101147147B1 (en) * 2004-04-01 2012-05-25 머크 샤프 앤드 돔 코포레이션 Modified polynucleotides for reducing off-target effects in rna interference
US20060014289A1 (en) * 2004-04-20 2006-01-19 Nastech Pharmaceutical Company Inc. Methods and compositions for enhancing delivery of double-stranded RNA or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells
WO2006012527A1 (en) 2004-07-23 2006-02-02 Endocyte, Inc. Bivalent linkers and conjugates thereof
EP1791567B1 (en) 2004-08-10 2015-07-29 Alnylam Pharmaceuticals Inc. Chemically modified oligonucleotides
AU2011250733C1 (en) * 2004-08-10 2013-09-12 Alnylam Pharmaceuticals, Inc. Chemically modified oligonucleotides
CN102628044A (en) 2004-09-24 2012-08-08 阿尔尼拉姆医药品有限公司 RNAi modulation of APOB and uses thereof
US7713944B2 (en) * 2004-10-13 2010-05-11 Isis Pharmaceuticals, Inc. Oligomers comprising activated disulfides which bind to plasma proteins and their use for delivery to cells
US7790878B2 (en) 2004-10-22 2010-09-07 Alnylam Pharmaceuticals, Inc. RNAi modulation of RSV, PIV and other respiratory viruses and uses thereof
US20060089324A1 (en) 2004-10-22 2006-04-27 Sailen Barik RNAi modulation of RSV, PIV and other respiratory viruses and uses thereof
US20060105979A1 (en) * 2004-11-08 2006-05-18 Hybridon, Inc. Synergistic inhibition of VEGF and modulation of the immune response
AU2005323303A1 (en) * 2004-11-24 2006-07-13 Alnylam Pharmaceuticals, Inc. RNAi modulation of the Bcr-Abl fusion gene and uses thereof
WO2006066158A2 (en) 2004-12-14 2006-06-22 Alnylam Pharmaceuticals, Inc. Rnai modulation of mll-af4 and uses thereof
US7507809B2 (en) 2005-01-07 2009-03-24 Alnylam Pharmaceuticals, Inc. RNAi modulation of RSV and therapeutic uses thereof
EP1841464B1 (en) * 2005-01-24 2012-06-27 Alnylam Pharmaceuticals Inc. Rnai modulation of the nogo-l or nogo-r gene and uses thereof
US20090016959A1 (en) * 2005-02-18 2009-01-15 Richard Beliveau Delivery of antibodies to the central nervous system
BRPI0607798A2 (en) * 2005-02-18 2010-06-22 Astrazeneca Ab method for predicting a patient's responsiveness to a chk1 inhibitor, use of a chk1 inhibitor, and kit for predicting a patient response to a chk1 inhibitor
US8044200B2 (en) * 2005-03-16 2011-10-25 Endocyte, Inc. Synthesis and purification of pteroic acid and conjugates thereof
US20060223777A1 (en) * 2005-03-29 2006-10-05 Dharmacon, Inc. Highly functional short hairpin RNA
CA2605068A1 (en) 2005-04-15 2006-10-26 The Board Of Regents Of The University Of Texas System Delivery of sirna by neutral lipid compositions
JP5116107B2 (en) 2005-06-27 2013-01-09 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 RNAi regulation of HIF-1 and its therapeutic use
JP2009502138A (en) 2005-07-21 2009-01-29 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 RNAi regulation of RhoA gene and use thereof
EP1937066A4 (en) * 2005-08-18 2008-12-24 Alnylam Pharmaceuticals Inc Methods and compositions for treating neurological disease
JP2009504783A (en) * 2005-08-19 2009-02-05 エンドサイト,インコーポレイテッド Ligand conjugates of vinca alkaloids, analogues and derivatives
JP5475992B2 (en) * 2005-08-19 2014-04-16 エンドサイト,インコーポレイテッド Multidrug ligand conjugate
WO2007029361A1 (en) * 2005-09-02 2007-03-15 Takeda Pharmaceutical Company Limited Sustained-release microsphere containing short chain deoxyribonucleic acid or short chain ribonucleic acid and method of producing the same
KR20080086440A (en) 2005-11-01 2008-09-25 알닐람 파마슈티칼스 인코포레이티드 Rnai inhibition of influenza virus replication
AU2006311730B2 (en) 2005-11-09 2010-12-02 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of Factor V Leiden mutant gene
WO2007068113A1 (en) * 2005-12-16 2007-06-21 Mcgill University 4'-thioarabinonucleotide-containing oligonucleotides, compounds and methods for their preparation and uses thereof
US7846908B2 (en) 2006-03-16 2010-12-07 Alnylam Pharmaceuticals, Inc. RNAi modulation of TGF-beta and therapeutic uses thereof
SG170780A1 (en) 2006-03-31 2011-05-30 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of eg5 gene
JP5814505B2 (en) * 2006-04-03 2015-11-17 ロシュ・イノベーション・センター・コペンハーゲン・アクティーゼルスカブRoche Innovation Center Copenhagen A/S Pharmaceutical composition comprising antimiRNA antisense oligonucleotide
US8007790B2 (en) 2006-04-03 2011-08-30 Stowers Institute For Medical Research Methods for treating polycystic kidney disease (PKD) or other cyst forming diseases
US7691824B2 (en) * 2006-04-28 2010-04-06 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a gene from the JC virus
GB0608838D0 (en) 2006-05-04 2006-06-14 Novartis Ag Organic compounds
EP2505646A1 (en) 2006-05-05 2012-10-03 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression of CRP
CA2672246A1 (en) * 2006-06-30 2008-03-13 Rosetta Genomics Ltd Detecting nucleic acids
CA2663601C (en) 2006-09-22 2014-11-25 Dharmacon, Inc. Duplex oligonucleotide complexes and methods for gene silencing by rna interference
WO2008061091A2 (en) 2006-11-13 2008-05-22 Grandis Jennifer R Antisense guanidinium peptide nucleic acid (gpna) oligonucleotides as antitumor agents
WO2009024834A2 (en) * 2006-12-05 2009-02-26 Rosetta Genomics Ltd Nucleic acids involved in viral infection
JP5252622B2 (en) * 2007-01-17 2013-07-31 独立行政法人産業技術総合研究所 Double-stranded RNA capable of expressing high nuclease resistance and excellent RNA interference effect
US8389486B2 (en) * 2007-01-26 2013-03-05 Rosetta Genomics, Ltd Methods for treating hematopoietic malignancies
WO2008101231A2 (en) 2007-02-16 2008-08-21 Endocyte, Inc. Methods and compositions for treating and diagnosing kidney disease
US9006206B2 (en) 2007-02-27 2015-04-14 Rosetta Genomics Ltd. Composition and methods for modulating cell proliferation and cell death
CA2699418A1 (en) * 2007-02-27 2008-09-04 Moshe Oren Composition and methods for modulating cell proliferation and cell death
US8067390B2 (en) * 2007-03-02 2011-11-29 The Board Of Regents Of The University Of Texas System Therapeutic targeting of interleukins using siRNA in neutral liposomes
EP2489372A3 (en) * 2007-03-14 2013-01-02 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
US20100273172A1 (en) * 2007-03-27 2010-10-28 Rosetta Genomics Ltd. Micrornas expression signature for determination of tumors origin
US20090105174A1 (en) * 2007-04-20 2009-04-23 Amgen Inc. Nucleic acids hybridizable to micro rna and precursors thereof
WO2008131419A2 (en) * 2007-04-23 2008-10-30 Alnylam Pharmaceuticals, Inc. Glycoconjugates of rna interference agents
WO2008147824A2 (en) 2007-05-22 2008-12-04 Mdrna, Inc. Hydroxymethyl substituted rna oligonucleotides and rna complexes
US9365634B2 (en) 2007-05-29 2016-06-14 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
US9877965B2 (en) 2007-06-25 2018-01-30 Endocyte, Inc. Vitamin receptor drug delivery conjugates for treating inflammation
EP2176293B1 (en) 2007-06-25 2019-04-03 Endocyte, Inc. Conjugates containing hydrophilic spacer linkers
CA2692287A1 (en) * 2007-06-29 2009-01-08 Newsouth Innovations Pty Limited Treatment of rheumatoid arthritis
WO2009009025A1 (en) * 2007-07-06 2009-01-15 Northeastern University Mixed micelles including amphipathic conjugates of rna agents, and uses thereof
EP2015071A1 (en) * 2007-07-13 2009-01-14 FUJIFILM Corporation Carrier, process for producing same, bioreactor, and chip for surface plasmon resonance analysis
EP2187965B1 (en) 2007-08-17 2019-10-09 Purdue Research Foundation Psma binding ligand-linker conjugates and methods for using
AR067997A1 (en) * 2007-08-24 2009-10-28 Novartis Ag ORGANIC COMPOUNDS
US8138318B2 (en) 2007-09-13 2012-03-20 Abbott Laboratories Hepatitis B pre-S2 nucleic acid
EP2209374B1 (en) 2007-10-25 2014-12-03 Endocyte, Inc. Tubulysins and processes for preparing
JP5252618B2 (en) * 2007-10-26 2013-07-31 独立行政法人産業技術総合研究所 Aromatic ring-modified siRNA with high RNA interference effect
AU2008320407A1 (en) * 2007-10-31 2009-05-07 Rosetta Genomics Ltd. Diagnosis and prognosis of specific cancers by means of differential detection of micro-RNAs/miRNAs
EP4074344A1 (en) 2007-12-04 2022-10-19 Arbutus Biopharma Corporation Targeting lipids
AU2008340354B2 (en) 2007-12-04 2014-04-17 Alnylam Pharmaceuticals, Inc. Folate-iRNA conjugates
EP2229459B1 (en) * 2007-12-13 2014-08-27 Alnylam Pharmaceuticals, Inc. Methods and compositions for prevention or treatment of RSV infection
CA2709635A1 (en) * 2007-12-20 2009-07-02 Angiochem Inc. Polypeptide-nucleic acid conjugates and uses thereof
AU2008342535B2 (en) 2007-12-27 2015-02-05 Arbutus Biopharma Corporation Silencing of polo-like kinase expression using interfering RNA
JP2011510623A (en) 2008-01-27 2011-04-07 ロゼッタ ゲノミックス エルティーディー. Methods and compositions for diagnosing complications of pregnancy
US20110118340A1 (en) * 2008-02-08 2011-05-19 Muthiah Manoharan Delivery of rnai constructs to oligodendrocytes
CN104975020B (en) * 2008-02-11 2020-01-17 菲奥医药公司 Modified RNAi polynucleotides and uses thereof
US8188060B2 (en) 2008-02-11 2012-05-29 Dharmacon, Inc. Duplex oligonucleotides with enhanced functionality in gene regulation
US8802923B2 (en) 2008-04-10 2014-08-12 A.B. Seeds Ltd. Compositions and methods for enhancing oil content in plants
WO2009127060A1 (en) 2008-04-15 2009-10-22 Protiva Biotherapeutics, Inc. Novel lipid formulations for nucleic acid delivery
AU2009236219B8 (en) 2008-04-15 2015-06-25 Arbutus Biopharma Corporation Silencing of CSN5 gene expression using interfering RNA
RU2518240C2 (en) * 2008-04-18 2014-06-10 Ангиочем Инк. Composition based on hydrophobic agents and method for preparing it (versions)
KR20220035504A (en) 2008-04-30 2022-03-22 이뮤노젠 아이엔씨 Cross-linkers and their uses
WO2009151620A2 (en) 2008-06-13 2009-12-17 Cornell Research Foundation, Inc. Wmc Pain treatment using erk2 inhibitors
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
US9068232B2 (en) 2008-08-06 2015-06-30 Rosetta Genomics Ltd. Gene expression signature for classification of kidney tumors
US20110160086A1 (en) 2008-08-06 2011-06-30 Rosetta Genomics Ltd. Gene expression signature for classification of kidney tumors
CN102405286A (en) 2008-09-22 2012-04-04 阿克赛医药公司 Reduced size self-delivering rnai compounds
EP2348848A4 (en) * 2008-10-03 2012-03-07 Charlotte Mecklenburg Hospital Treatment of hepatitis c infection with metalloporphyrins
EP2346896A4 (en) 2008-10-15 2014-06-25 Angiochem Inc Etoposide and doxorubicin conjugates for drug delivery
US8921314B2 (en) 2008-10-15 2014-12-30 Angiochem, Inc. Conjugates of GLP-1 agonists and uses thereof
WO2010045584A1 (en) * 2008-10-17 2010-04-22 Endocyte, Inc. Folate targeting of nucleotides
EP2350277A1 (en) * 2008-10-23 2011-08-03 Alnylam Pharmaceuticals, Inc. Methods and compositions for prevention or treatment of rsv infection using modified duplex rna molecules
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
US20100222417A1 (en) * 2008-11-26 2010-09-02 Alnylam Pharmaceuticals Compostions and methods for enhancing oligonucleotide delivery across and into epithelial tissues
CA2744093A1 (en) 2008-12-03 2010-06-10 Marina Biotech, Inc. Una oligomer structures for therapeutic agents
CA2745524C (en) 2008-12-05 2020-06-09 Angiochem Inc. Conjugates of neurotensin or neurotensin analogs and uses thereof
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
WO2010078536A1 (en) 2009-01-05 2010-07-08 Rxi Pharmaceuticals Corporation Inhibition of pcsk9 through rnai
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
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
KR101141544B1 (en) 2009-03-13 2012-05-03 한국과학기술원 Multi-conjugate of siRNA and preparing method thereof
US9173891B2 (en) 2009-04-20 2015-11-03 Angiochem, Inc. Treatment of ovarian cancer using an anticancer agent conjugated to an angiopep-2 analog
US9012225B2 (en) 2009-05-05 2015-04-21 Miragen Therapeutics Lipophilic polynucleotide conjugates
EP2438168B1 (en) 2009-06-01 2020-02-12 Halo-Bio Rnai Therapeutics, Inc. Polynucleotides for multivalent rna interference, compositions and methods of use thereof
SI2437790T1 (en) 2009-06-03 2019-07-31 Immunogen, Inc. Conjugation methods
CA2767127A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Novel lipid formulations for delivery of therapeutic agents to solid tumors
US8236943B2 (en) * 2009-07-01 2012-08-07 Protiva Biotherapeutics, Inc. Compositions and methods for silencing apolipoprotein B
US9161988B2 (en) 2009-07-02 2015-10-20 Angiochem Inc. Multimeric peptide conjugates and uses thereof
US8716464B2 (en) 2009-07-20 2014-05-06 Thomas W. Geisbert Compositions and methods for silencing Ebola virus gene expression
WO2011009624A1 (en) 2009-07-22 2011-01-27 Cenix Bioscience Gmbh Delivery system and conjugates for compound delivery via naturally occurring intracellular transport routes
WO2011021177A2 (en) 2009-08-19 2011-02-24 Rosetta Genomics Ltd. Compositions and methods for prognosis and treatment of prostate cancer
US9222086B2 (en) 2009-09-23 2015-12-29 Protiva Biotherapeutics, Inc. Compositions and methods for silencing genes expressed in cancer
WO2011041788A2 (en) * 2009-10-02 2011-04-07 The Trustees Of Columbia University In The City Of New York Turkey viral hepatitis virus and uses thereof
JP2013506709A (en) 2009-10-06 2013-02-28 イミュノジェン・インコーポレーテッド Effective conjugates and hydrophilic linkers
CA3177356A1 (en) 2009-11-02 2011-05-05 The Trustees Of The University Of Pennsylvania Foot and mouth disease virus (fmdv) consensus proteins, coding sequences therefor and vaccines made therefrom
US20130017223A1 (en) 2009-12-18 2013-01-17 The University Of British Columbia Methods and compositions for delivery of nucleic acids
CA2789629A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc. Cd20 antibodies and uses thereof
US9951324B2 (en) 2010-02-25 2018-04-24 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
EP2544719B1 (en) 2010-03-12 2019-07-03 Debiopharm International S.A. Cd37-binding molecules and immunoconjugates thereof
CN106074591B (en) 2010-03-24 2020-01-14 菲奥医药公司 RNA interference in ocular symptoms
KR101852210B1 (en) 2010-03-24 2018-04-25 알엑스아이 파마슈티칼스 코포레이션 Rna interference in dermal and fibrotic indications
RU2615143C2 (en) 2010-03-24 2017-04-04 Адвирна Self-delivered rnai compounds of reduced size
CA2792291A1 (en) 2010-03-29 2011-10-06 Kumamoto University Sirna therapy for transthyretin (ttr) related ocular amyloidosis
WO2011133871A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. 5'-end derivatives
HUE040278T2 (en) 2010-04-23 2019-02-28 Cold Spring Harbor Laboratory NOVEL STRUCTURALLY DESIGNED shRNAs
DE102010022937A1 (en) * 2010-06-04 2011-12-08 Universitätsklinikum Jena Cell-specific activatable biologically active molecules based on siRNA, methods for their activation and application kit for administration
WO2011159797A2 (en) 2010-06-15 2011-12-22 Cellular Dynamics International, Inc. A compendium of ready-built stem cell models for interrogation of biological response
WO2012016188A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
WO2012016184A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
WO2012019024A2 (en) 2010-08-04 2012-02-09 Immunogen, Inc. Her3-binding molecules and immunoconjugates thereof
KR20140008308A (en) 2010-10-29 2014-01-21 이뮤노젠 아이엔씨 Novel egfr-binding molecules and immunoconjugates thereof
CN105399831A (en) 2010-10-29 2016-03-16 伊缪诺金公司 Non-antagonistic egfr-binding molecules and immunoconjugates thereof
EP2643479B1 (en) 2010-11-22 2017-09-13 Rosetta Genomics Ltd Methods and materials for classification of tissue of origin of tumor samples
EP3202760B1 (en) 2011-01-11 2019-08-21 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
JP2014505064A (en) 2011-01-26 2014-02-27 セニックス バイオサイエンス ゲーエムベーハー Delivery systems and conjugates for delivering compounds via naturally occurring intracellular transport pathways
US9068184B2 (en) 2011-06-21 2015-06-30 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of protein C (PROC) genes
EP2723865B1 (en) 2011-06-21 2019-03-27 Alnylam Pharmaceuticals, Inc. METHODS FOR DETERMINING ACTIVITY OF RNAi IN A SUBJECT
MX344807B (en) 2011-06-21 2017-01-09 Alnylam Pharmaceuticals Inc Compositions and methods for inhibition of expression of apolipoprotein c-iii (apoc3) genes.
WO2013033230A1 (en) 2011-08-29 2013-03-07 Isis Pharmaceuticals, Inc. Oligomer-conjugate complexes and their use
EP2750681B1 (en) * 2011-08-30 2020-05-27 Quanta Biodesign, Ltd. Branched discrete peg constructs
US9701623B2 (en) 2011-09-27 2017-07-11 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
KR20140105765A (en) 2011-11-21 2014-09-02 이뮤노젠 아이엔씨 Method of treatment of tumors that are resistant to egfr therapies by egfr antibody cytotoxic agent conjugate
US9035039B2 (en) 2011-12-22 2015-05-19 Protiva Biotherapeutics, Inc. Compositions and methods for silencing SMAD4
WO2013124816A2 (en) 2012-02-22 2013-08-29 Brainstem Biotec Ltd. Generation of neural stem cells and motor neurons
US10080805B2 (en) 2012-02-24 2018-09-25 Purdue Research Foundation Cholecystokinin B receptor targeting for imaging and therapy
RS58077B1 (en) 2012-02-24 2019-02-28 Arbutus Biopharma Corp Trialkyl cationic lipids and methods of use thereof
US20140080175A1 (en) 2012-03-29 2014-03-20 Endocyte, Inc. Processes for preparing tubulysin derivatives and conjugates thereof
US20130259927A1 (en) * 2012-04-02 2013-10-03 Gradalis, Inc. Ewing's Sarcoma Bifunctional shRNA Design
US20150322428A1 (en) * 2012-06-18 2015-11-12 Isis Pharmaceuticals, Inc. Compounds and methods for improved cellular uptake of antisense compounds
WO2014018375A1 (en) 2012-07-23 2014-01-30 Xenon Pharmaceuticals Inc. Cyp8b1 and uses thereof in therapeutic and diagnostic methods
BR112015008365A2 (en) 2012-10-16 2017-07-04 Endocyte Inc compound of the formula bl (d) x, or a pharmaceutically acceptable salt thereof, pharmaceutical composition, use of a compound, unit dosage form or unit dose composition, composition for treating a cancer in a patient, and method for treating a cancer in a patient
CA2891476C (en) 2012-11-15 2022-07-05 Endocyte, Inc. Drug delivery conjugates, and methods for treating diseases caused by psma expressing cells
CA2895806C (en) 2013-03-15 2023-02-21 The Trustees Of The University Of Pennsylvania Foot and mouth disease virus (fmdv) consensus proteins, coding sequences therefor and vaccines made therefrom
JP6649248B2 (en) 2013-05-01 2020-02-19 レグルス セラピューティクス インコーポレイテッド Compounds and methods for enhancing cellular uptake
CA2908939C (en) 2013-05-01 2023-08-22 Balkrishen Bhat Microrna compounds and methods for modulating mir-122
BR112015027321A8 (en) 2013-05-01 2018-01-02 Isis Pharmaceuticals Inc COMPOUNDS AND COMPOSITIONS FOR MODULING APOLIPOPROTEIN(A) EXPRESSION AND THEIR USES
US20160145628A1 (en) 2013-06-18 2016-05-26 Ramot At Tel-Aviv University Ltd. Nanocarrier system for micrornas and uses thereof
WO2015006740A2 (en) 2013-07-11 2015-01-15 Alnylam Pharmaceuticals, Inc. Oligonucleotide-ligand conjugates and process for their preparation
EP3415489A1 (en) 2013-10-18 2018-12-19 Deutsches Krebsforschungszentrum Labeled inhibitors of prostate specific membrane antigen (psma), their use as imaging agents and pharmaceutical agents for the treatment of prostate cancer
JP6772062B2 (en) 2013-12-02 2020-10-21 フィオ ファーマシューティカルズ コーポレーションPhio Pharmaceuticals Corp. Cancer immunotherapy
US20160304875A1 (en) 2013-12-04 2016-10-20 Rxi Pharmaceuticals Corporation Methods for treatment of wound healing utilizing chemically modified oligonucleotides
CA2946719C (en) 2014-03-25 2023-09-26 Arcturus Therapeutics, Inc. Una oligomers having reduced off-target effects in gene silencing
US9856475B2 (en) 2014-03-25 2018-01-02 Arcturus Therapeutics, Inc. Formulations for treating amyloidosis
JP6771387B2 (en) 2014-03-25 2020-10-21 アークトゥラス・セラピューティクス・インコーポレイテッドArcturus Therapeutics,Inc. Transthyretin allele-selective UNA oligomer for gene silencing
US11279934B2 (en) 2014-04-28 2022-03-22 Phio Pharmaceuticals Corp. Methods for treating cancer using nucleic acids targeting MDM2 or MYCN
WO2015168172A1 (en) 2014-04-28 2015-11-05 Isis Pharmaceuticals, Inc. Linkage modified oligomeric compounds
KR102149571B1 (en) 2014-05-01 2020-08-31 아이오니스 파마수티컬즈, 인코포레이티드 Compositions and methods for modulating growth hormone receptor expression
US10098959B2 (en) 2014-05-01 2018-10-16 Ionis Pharmaceuticals, Inc. Method for synthesis of reactive conjugate clusters
HUE052931T2 (en) 2014-05-01 2021-05-28 Ionis Pharmaceuticals Inc Compositions and methods for modulating angiopoietin-like 3 expression
ES2745825T3 (en) 2014-05-01 2020-03-03 Ionis Pharmaceuticals Inc Compositions and methods for modulating expression of complement factor B
WO2015168605A1 (en) 2014-05-01 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treatment of disorders in the front of the eye utilizing nucleic acid molecules
BR112016022855B1 (en) 2014-05-01 2022-08-02 Ionis Pharmaceuticals, Inc COMPOUNDS AND COMPOSITIONS TO MODULATE THE EXPRESSION OF PKK AND ITS USES
WO2015179693A1 (en) 2014-05-22 2015-11-26 Isis Pharmaceuticals, Inc. Conjugated antisense compounds and their use
US10434174B2 (en) 2014-06-09 2019-10-08 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Combination therapies using platinum agents and agents that target tumor-associated stroma or tumor cells
US10758613B2 (en) 2014-06-09 2020-09-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services National Intstitutes Of Health Combination therapies using anti-metabolites and agents that target tumor-associated stroma or tumor cells
WO2015191583A2 (en) 2014-06-09 2015-12-17 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells and topoisomerase
WO2015191568A2 (en) 2014-06-09 2015-12-17 Biomed Valley Discoveries, Inc. Combination therapies using agents that target tumor-associated stroma or tumor cells and tumor vasculature
US10758526B2 (en) 2014-06-09 2020-09-01 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services National Institutes Of Health Combination therapies using agents that target tumor-associated stroma or tumor cells and other pathways
WO2015191590A2 (en) 2014-06-09 2015-12-17 Biomed Valley Discoveries, Inc. Combination therapies targeting tumor-associated stroma or tumor cells and microtubules
WO2016011324A2 (en) 2014-07-18 2016-01-21 Oregon Health & Science University 5'-triphosphate oligoribonucleotides
WO2016030863A1 (en) 2014-08-29 2016-03-03 Glaxosmithkline Intellectual Property Development Limited Compounds and methods for treating viral infections
KR102506169B1 (en) 2014-09-05 2023-03-08 피오 파마슈티칼스 코프. Methods for treating aging and skin disorders using nucleic acids targeting tyr or mmp1
US9950194B2 (en) 2014-09-09 2018-04-24 Mevion Medical Systems, Inc. Patient positioning system
KR20170066433A (en) * 2014-09-17 2017-06-14 더 유니버시티 오브 아이오와 리서치 파운데이션 Viral rna segments as immunomodulatory agents and vaccine components
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2016081444A1 (en) 2014-11-17 2016-05-26 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
US10188759B2 (en) 2015-01-07 2019-01-29 Endocyte, Inc. Conjugates for imaging
US9434947B2 (en) 2015-01-20 2016-09-06 Oregon Health & Science University Modulation of KCNH2 isoform expression by oligonucleotides as a therapeutic approach for long QT syndrome
WO2016161299A1 (en) 2015-04-01 2016-10-06 Arcturus Therapeutics, Inc. Therapeutic una oligomers and uses thereof
DK3307326T3 (en) 2015-06-15 2020-10-19 Angiochem Inc METHODS FOR THE TREATMENT OF LEPTOMENINGAL CARCINOMATOSIS
CA2988603A1 (en) 2015-06-15 2016-12-22 Mpeg La, Llc Defined multi-conjugate oligonucleotides
EP3313186A4 (en) 2015-06-29 2019-04-03 Biomed Valley Discoveries, Inc. Lpt-723 and immune checkpoint inhibitor combinations and methods of treatment
WO2017007825A1 (en) 2015-07-06 2017-01-12 Rxi Pharmaceuticals Corporation Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
CN108135923B (en) 2015-07-06 2021-03-02 菲奥医药公司 Nucleic acid molecules targeting superoxide dismutase 1(SOD1)
EP4092119A3 (en) 2015-07-10 2023-03-22 Ionis Pharmaceuticals, Inc. Modulators of diacyglycerol acyltransferase 2 (dgat2)
WO2017015671A1 (en) 2015-07-23 2017-01-26 Arcturus Therapeutics, Inc. Compositions for treating amyloidosis
CN108366966A (en) 2015-08-24 2018-08-03 光环生物干扰疗法公司 Polynucleotides nano particle and application thereof for adjusting gene expression
JP6666673B2 (en) * 2015-09-07 2020-03-18 キリンホールディングス株式会社 Intracellular delivery vehicle
US20180273577A1 (en) 2015-09-24 2018-09-27 Ionis Pharmaceuticals, Inc. Modulators of kras expression
CA3002744A1 (en) 2015-10-19 2017-04-27 Rxi Pharmaceuticals Corporation Reduced size self-delivering nucleic acid compounds targeting long non-coding rna
EP3371201A4 (en) 2015-11-06 2019-09-18 Ionis Pharmaceuticals, Inc. Conjugated antisense compounds for use in therapy
BR122023026882A2 (en) 2015-11-06 2024-01-23 Ionis Pharmaceuticals, Inc USE OF AN OLIGOMERIC COMPOUND
JP2019503394A (en) 2016-01-31 2019-02-07 ユニバーシティ・オブ・マサチューセッツUniversity Of Massachusetts Branched oligonucleotide
US10548881B2 (en) 2016-02-23 2020-02-04 Tarveda Therapeutics, Inc. HSP90 targeted conjugates and particles and formulations thereof
TWI783434B (en) 2016-03-07 2022-11-11 美商愛羅海德製藥公司 Targeting ligands for therapeutic compounds
US10036024B2 (en) 2016-06-03 2018-07-31 Purdue Research Foundation siRNA compositions that specifically downregulate expression of a variant of the PNPLA3 gene and methods of use thereof for treating a chronic liver disease or alcoholic liver disease (ALD)
UA124734C2 (en) 2016-06-14 2021-11-10 Рідженерон Фармасьютікалз, Інк. Anti-c5 antibodies and uses thereof
SI3484524T1 (en) 2016-07-15 2023-01-31 Ionis Pharmaceuticals, Inc. Compounds and methods for modulation of smn2
IL291323B2 (en) 2016-07-15 2023-09-01 Am Chemicals Llc Non-nucleosidic solid supports and phosphoramidite building blocks for oligonucleotide synthesis
EP3496758A4 (en) 2016-08-12 2020-11-11 University of Massachusetts Conjugated oligonucleotides
UY37376A (en) 2016-08-26 2018-03-23 Amgen Inc ARNI CONSTRUCTIONS TO INHIBIT EXPRESSION OF ASGR1 AND METHODS FOR USE
JP6989521B2 (en) 2016-09-02 2022-01-05 アローヘッド ファーマシューティカルズ インコーポレイテッド Targeting ligand
US10933081B2 (en) 2016-09-21 2021-03-02 Alnylam Pharmaceuticals, Inc. Myostatin iRNA compositions and methods of use thereof
EP3522898A4 (en) 2016-10-06 2020-05-27 Ionis Pharmaceuticals, Inc. Method of conjugating oligomeric compounds
US11447519B2 (en) 2016-11-10 2022-09-20 San Diego State University Research Foundation Compounds for fluorescence sensing of duplex formation
JOP20170192A1 (en) 2016-12-01 2019-01-30 Takeda Pharmaceuticals Co Cyclic dinucleotide
JP7183167B2 (en) 2017-02-06 2022-12-05 エムペグ エルエイ リミテッド ライアビリティ カンパニー Multimeric oligonucleotides with reduced renal clearance
JOP20190215A1 (en) 2017-03-24 2019-09-19 Ionis Pharmaceuticals Inc Modulators of pcsk9 expression
US11324820B2 (en) 2017-04-18 2022-05-10 Alnylam Pharmaceuticals, Inc. Methods for the treatment of subjects having a hepatitis b virus (HBV) infection
WO2019028055A1 (en) 2017-07-31 2019-02-07 The Trustees Of Columbia Univeristy In The City Of New York Compounds, compositionals, and methods for treating t-cell acute lymphoblastic leukemia
US11414665B2 (en) 2017-12-01 2022-08-16 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, composition and conjugate comprising the same, and preparation method and use thereof
EP3719126A4 (en) 2017-12-01 2021-10-20 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, composition and conjugate containing nucleic acid, preparation method therefor and use thereof
CN110997917A (en) 2017-12-01 2020-04-10 苏州瑞博生物技术有限公司 Nucleic acid, composition containing nucleic acid, conjugate, preparation method and application
WO2019105418A1 (en) 2017-12-01 2019-06-06 苏州瑞博生物技术有限公司 Double-stranded oligonucleotide, composition and conjugate comprising double-stranded oligonucleotide, preparation method therefor and use thereof
UY38003A (en) 2017-12-12 2019-06-28 Amgen Inc ARNI CONSTRUCTIONS TO INHIBIT THE EXPRESSION OF PNPLA3 AND METHODS OF USE OF THE SAME
KR20200098528A (en) 2017-12-13 2020-08-20 리제너론 파아마슈티컬스, 인크. Anti-C5 antibody combinations and uses thereof
KR102617947B1 (en) * 2017-12-29 2023-12-27 쑤저우 리보 라이프 사이언스 컴퍼니, 리미티드 Conjugates and their preparation and uses
JP2021511072A (en) 2018-01-15 2021-05-06 アイオニス・ファーマシューティカルズ・インコーポレイテッドIonis Pharmaceuticals,Inc. DNM2 expression modulator
CA3090901A1 (en) 2018-02-12 2019-08-15 Ionis Pharmaceuticals, Inc. Modified compounds and uses thereof
US11674141B2 (en) 2018-02-28 2023-06-13 National University Corporation Tokyo Medical And Dental University Ischemic-lesion-site-specific gene therapy
KR20210018267A (en) 2018-05-07 2021-02-17 알닐람 파마슈티칼스 인코포레이티드 Extrahepatic delivery
JOP20200280A1 (en) 2018-05-09 2020-11-05 Ionis Pharmaceuticals Inc Compounds and methods for reducing fxi expression
US10857174B2 (en) 2018-07-27 2020-12-08 United States Government As Represented By The Department Of Veterans Affairs Morpholino oligonucleotides useful in cancer treatment
US20210292768A1 (en) 2018-08-08 2021-09-23 Arcturus Therapeutics, Inc. Compositions and agents against nonalcoholic steatohepatitis
EP3833763A4 (en) 2018-08-10 2023-07-19 University of Massachusetts Modified oligonucleotides targeting snps
AU2019321375A1 (en) 2018-08-13 2021-03-11 Alnylam Pharmaceuticals, Inc. Hepatitis B virus (HBV) dsRNA agent compositions and methods of use thereof
EP3842534A4 (en) 2018-08-21 2022-07-06 Suzhou Ribo Life Science Co., Ltd. Nucleic acid, pharmaceutical composition and conjugate containing nucleic acid, and use thereof
TW202023573A (en) 2018-09-19 2020-07-01 美商Ionis製藥公司 Modulators of pnpla3 expression
AU2019350765A1 (en) 2018-09-28 2021-05-27 Alnylam Pharmaceuticals, Inc. Transthyretin (TTR) iRNA compositions and methods of use thereof for treating or preventing TTR-associated ocular diseases
CN111655297A (en) 2018-09-30 2020-09-11 苏州瑞博生物技术有限公司 siRNA conjugate and preparation method and application thereof
WO2020093322A1 (en) * 2018-11-08 2020-05-14 深圳华大生命科学研究院 Micro-fluidic chip and preparation method therefor, and dna synthesis method
AR117297A1 (en) 2018-12-10 2021-07-28 Amgen Inc ARNI CONSTRUCTS TO INHIBIT THE EXPRESSION OF PNPLA3 AND METHODS OF USE OF THEM
SG11202105900UA (en) 2018-12-10 2021-07-29 Amgen Inc CHEMICALLY-MODIFIED RNAi CONSTRUCTS AND USES THEREOF
US11116778B2 (en) 2019-01-15 2021-09-14 Empirico Inc. Prodrugs of ALOX-15 inhibitors and methods of using the same
WO2020225779A1 (en) 2019-05-09 2020-11-12 Istituto Pasteur Italia - Fondazione Cenci Bolognetti Rig-i agonists for cancer treatment and immunotherapy
EP3969052A1 (en) 2019-05-17 2022-03-23 Alnylam Pharmaceuticals Inc. Oral delivery of oligonucleotides
EP3976786A2 (en) 2019-05-30 2022-04-06 Amgen, Inc Rnai constructs for inhibiting scap expression and methods of use thereof
CA3150758A1 (en) 2019-08-13 2021-02-18 Amgen Inc. Rnai constructs for inhibiting slc30a8 expression and methods of use thereof
JP2022552249A (en) 2019-10-14 2022-12-15 アストラゼネカ・アクチエボラーグ Modulators of PNPLA3 expression
JP2023500661A (en) * 2019-11-01 2023-01-10 アルナイラム ファーマシューティカルズ, インコーポレイテッド HUNTINGTIN (HTT) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
EP4055165A1 (en) 2019-11-06 2022-09-14 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof for treating or preventing ttr-associated ocular diseases
MX2022005220A (en) 2019-11-06 2022-08-11 Alnylam Pharmaceuticals Inc Extrahepatic delivery.
CA3160657A1 (en) 2019-11-08 2021-05-14 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides targeting bromodomain containing protein 4 (brd4) for immunotherapy
US20230078200A1 (en) 2019-12-09 2023-03-16 Amgen Inc. RNAi CONSTRUCTS AND METHODS FOR INHIBITING LPA EXPRESSION
EP4085136A1 (en) 2019-12-31 2022-11-09 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides with improved systemic delivery
MX2022010602A (en) 2020-02-28 2022-09-09 Ionis Pharmaceuticals Inc Compounds and methods for modulating smn2.
WO2021178607A1 (en) 2020-03-05 2021-09-10 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof for treating or preventing complement component c3-associated diseases
EP4126967A1 (en) 2020-03-23 2023-02-08 Amgen Inc. Monoclonal antibodies to chemically-modified nucleic acids and uses thereof
KR20240004092A (en) 2020-06-01 2024-01-11 암젠 인크 RNAI constructs and methods of using the same to inhibit HSD17B13 expression
US20230257745A1 (en) 2020-07-10 2023-08-17 Alnylam Pharmaceuticals, Inc. Circular siRNAs
WO2022026531A1 (en) * 2020-07-29 2022-02-03 Alnylam Pharmaceuticals, Inc. Atxn2 irna compositions and methods of use thereof for treating or preventing atxn2-associated neurodegenerative diseases
IL300360A (en) 2020-08-13 2023-04-01 Amgen Inc RNAi CONSTRUCTS AND METHODS FOR INHIBITING MARC1 EXPRESSION
EP4136092A4 (en) 2020-11-18 2023-10-11 Ionis Pharmaceuticals, Inc. Compounds and methods for modulating angiotensinogen expression
WO2022140126A1 (en) 2020-12-21 2022-06-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Dna vector and uses thereof for detecting hiv and siv
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
EP4337197A1 (en) 2021-05-10 2024-03-20 Yissum Research Development Company of the Hebrew University of Jerusalem Ltd. Pharmaceutical compositions for treating neurological conditions
WO2022245583A1 (en) * 2021-05-18 2022-11-24 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
AU2022299169A1 (en) 2021-06-23 2024-02-08 Beth Israel Deaconess Medical Center, Inc. Optimized anti-flt1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
WO2023283403A2 (en) 2021-07-09 2023-01-12 Alnylam Pharmaceuticals, Inc. Bis-rnai compounds for cns delivery
WO2023003922A1 (en) 2021-07-21 2023-01-26 Alnylam Pharmaceuticals, Inc. Metabolic disorder-associated target gene irna compositions and methods of use thereof
WO2023015264A1 (en) 2021-08-04 2023-02-09 Phio Pharmaceuticals Corp. Immunotherapy of cancer utilizing natural killer cells treated with chemically modified oligonucleotides
WO2023015265A2 (en) 2021-08-04 2023-02-09 Phio Pharmaceuticals Corp. Chemically modified oligonucleotides
WO2023014938A1 (en) 2021-08-05 2023-02-09 Sanegene Bio Usa Inc. 1'-alkyl modified ribose derivatives and methods of use
WO2023049258A1 (en) 2021-09-22 2023-03-30 Sanegene Bio Usa Inc. 2'-alkyl or 3'- alkyl modified ribose derivatives for use in the in-vivo delivery of oligonucleotides
WO2023059629A1 (en) 2021-10-05 2023-04-13 Amgen Inc. Compositions and methods for enhancing gene silencing activity of oligonucleotide compounds
US20230227480A1 (en) 2021-10-05 2023-07-20 Sanegene Bio Usa Inc. Polyhydroxylated cyclopentane derivatives and methods of use
WO2023064530A1 (en) 2021-10-15 2023-04-20 Alnylam Pharmaceuticals, Inc. Extra-hepatic delivery irna compositions and methods of use thereof
WO2023069754A2 (en) 2021-10-22 2023-04-27 Amgen Inc. Rnai constructs for inhibiting gpam expression and methods of use thereof
WO2023144798A1 (en) 2022-01-31 2023-08-03 Genevant Sciences Gmbh Ionizable cationic lipids for lipid nanoparticles
WO2023144792A1 (en) 2022-01-31 2023-08-03 Genevant Sciences Gmbh Poly(alkyloxazoline)-lipid conjugates and lipid particles containing same
US20230346819A1 (en) 2022-02-22 2023-11-02 Sanegene Bio Usa Inc. 5'-modified carbocyclic ribonucleotide derivatives and methods of use
US20230340011A1 (en) * 2022-04-22 2023-10-26 Asteroid Therapeutics Steroidal compositions and methods of treating lipogenic cancers
WO2023220744A2 (en) 2022-05-13 2023-11-16 Alnylam Pharmaceuticals, Inc. Single-stranded loop oligonucleotides
WO2024006999A2 (en) 2022-06-30 2024-01-04 Alnylam Pharmaceuticals, Inc. Cyclic-disulfide modified phosphate based oligonucleotide prodrugs
WO2024015796A1 (en) 2022-07-11 2024-01-18 Sanegene Bio Usa Inc. Optimized 2'- modified ribose derivatives and methods of use
WO2024026258A2 (en) 2022-07-25 2024-02-01 Amgen Inc. Rnai constructs and methods for inhibiting fam13a expression

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6001991A (en) * 1996-10-04 1999-12-14 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of MDR P-glycoprotein gene expression

Family Cites Families (89)

* 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
BE795866A (en) 1972-02-29 1973-08-23 Basf Ag PROCESS FOR THE PREPARATION OF SOLID CHOLINE CHLORIDE FREE FLOW
DE3013839A1 (en) 1979-04-13 1980-10-30 Freunt Ind Co Ltd METHOD FOR PRODUCING AN ACTIVATED PHARMACEUTICAL COMPOSITION
US4465503A (en) 1979-08-07 1984-08-14 Ube Industries Ltd. Diphenyl ether derivatives, process for preparing the same and herbicidal compositions containing the same
US4469863A (en) 1980-11-12 1984-09-04 Ts O Paul O P Nonionic nucleic acid alkyl and aryl phosphonates and processes for manufacture and use thereof
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
JPS5921613A (en) 1982-07-28 1984-02-03 Takeda Chem Ind Ltd Pharmaceutical preparation for rectum administration
US4486435A (en) 1983-05-16 1984-12-04 Basf Wyandotte Corporation Spray-dried vitamin powders using hydrophobic silica
JPS6051189A (en) * 1983-08-30 1985-03-22 Sankyo Co Ltd Thiazolidine derivative and its preparation
US4897355A (en) 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
ATE81872T1 (en) 1985-03-15 1992-11-15 James Summerton STEROREOGULAR POLYNUCLEOTIDE BINDING POLYMERS.
US5235033A (en) 1985-03-15 1993-08-10 Anti-Gene Development Group Alpha-morpholino ribonucleoside derivatives and polymers thereof
FR2594693B1 (en) 1986-02-24 1990-01-05 Farah Nabil NOVEL PROCESSES FOR THE PREPARATION FROM DRY EMULSIONS OF SOLID ORAL FORMS WITH MODIFIED AND DELAYED RELEASE OF THEIR ACTIVE INGREDIENTS
SE8701479L (en) 1987-04-09 1988-10-10 Carbomatrix Ab METHOD FOR CONTAINING BIOLOGICALLY EFFECTIVE PREPARATIONS AND USE THEREOF
CH672048A5 (en) 1987-09-16 1989-10-31 Nestle Sa
US5328470A (en) 1989-03-31 1994-07-12 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
FR2645866B1 (en) 1989-04-17 1991-07-05 Centre Nat Rech Scient NEW LIPOPOLYAMINES, THEIR PREPARATION AND THEIR USE
US5256775A (en) 1989-06-05 1993-10-26 Gilead Sciences, Inc. Exonuclease-resistant oligonucleotides
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5264564A (en) 1989-10-24 1993-11-23 Gilead Sciences Oligonucleotide analogs with novel linkages
US5177198A (en) 1989-11-30 1993-01-05 University Of N.C. At Chapel Hill Process for preparing oligoribonucleoside and oligodeoxyribonucleoside boranophosphates
US5130302A (en) 1989-12-20 1992-07-14 Boron Bilogicals, Inc. Boronated nucleoside, nucleotide and oligonucleotide compounds, compositions and methods for using same
US5457191A (en) 1990-01-11 1995-10-10 Isis Pharmaceuticals, Inc. 3-deazapurines
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US5149797A (en) 1990-02-15 1992-09-22 The Worcester Foundation For Experimental Biology Method of site-specific alteration of rna and production of encoded polypeptides
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5000888A (en) 1990-05-23 1991-03-19 Basf Corporation Process for spray drying riboflavin to produce a granulate product having low binder content
JPH0436233A (en) 1990-05-29 1992-02-06 Biomaterial Universe Kk Sustained release preparation containing physiologically active substance and decomposable and absorbable in living body
US6147204A (en) * 1990-06-11 2000-11-14 Nexstar Pharmaceuticals, Inc. Nucleic acid ligand complexes
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5223618A (en) 1990-08-13 1993-06-29 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analog compounds
WO1992002258A1 (en) 1990-07-27 1992-02-20 Isis Pharmaceuticals, Inc. Nuclease resistant, pyrimidine modified oligonucleotides that detect and modulate gene expression
WO1992003464A1 (en) 1990-08-28 1992-03-05 Microprobe Corporation Solid support synthesis of 3'-tailed oligonucleotides via a linking molecule
US5283185A (en) 1991-08-28 1994-02-01 University Of Tennessee Research Corporation Method for delivering nucleic acids into cells
US5484908A (en) 1991-11-26 1996-01-16 Gilead Sciences, Inc. Oligonucleotides containing 5-propynyl pyrimidines
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
IL105914A0 (en) 1992-06-04 1993-10-20 Univ California Methods and compositions for in vivo gene therapy
CA2135313A1 (en) 1992-06-18 1994-01-06 Theodore Choi Methods for producing transgenic non-human animals harboring a yeast artificial chromosome
US5476925A (en) 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups
GB9304620D0 (en) 1993-03-06 1993-04-21 Ciba Geigy Ag Compounds
JPH07101882A (en) 1993-09-30 1995-04-18 Sanei Gen F F I Inc Preparation containing water-soluble hemicellulose
JPH07101881A (en) 1993-09-30 1995-04-18 Sanei Gen F F I Inc Preparation containing water-soluble hemicellulose
JP3342550B2 (en) 1993-10-01 2002-11-11 三栄源エフ・エフ・アイ株式会社 Formulations containing water-soluble hemicellulose
JPH07101884A (en) 1993-10-01 1995-04-18 Sanei Gen F F I Inc Prpearation containing water-soluble hemicellulose
US5801154A (en) * 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
US5652359A (en) * 1993-12-02 1997-07-29 Epoch Pharmaceuticals, Inc. Oligonucleotides containing n-methyl thiolated bases having antiviral activity
US5519134A (en) * 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5539083A (en) 1994-02-23 1996-07-23 Isis Pharmaceuticals, Inc. Peptide nucleic acid combinatorial libraries and improved methods of synthesis
US5646126A (en) * 1994-02-28 1997-07-08 Epoch Pharmaceuticals Sterol modified oligonucleotide duplexes having anticancer activity
JPH07242568A (en) 1994-03-04 1995-09-19 Eisai Co Ltd Pharmaceutical preparation for masking bitterness
US5691316A (en) 1994-06-01 1997-11-25 Hybridon, Inc. Cyclodextrin cellular delivery system for oligonucleotides
JPH0867666A (en) 1994-08-29 1996-03-12 Lion Corp Powdery pharmaceutical preparation containing carotenoid and its production
CA2220950A1 (en) 1995-05-26 1996-11-28 Somatix Therapy Corporation Delivery vehicles comprising stable lipid/nucleic acid complexes
US5994517A (en) 1995-11-22 1999-11-30 Paul O. P. Ts'o Ligands to enhance cellular uptake of biomolecules
IL118003A0 (en) * 1996-04-23 1996-08-04 Yeda Res & Dev Novel vip fragments and pharmaceutical compositions comprising them
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US6034135A (en) 1997-03-06 2000-03-07 Promega Biosciences, Inc. Dimeric cationic lipids
US6369237B1 (en) * 1997-03-07 2002-04-09 President And Fellows Of Harvard College DNA glycosylase inhibitors, and uses related thereto
US6509323B1 (en) 1998-07-01 2003-01-21 California Institute Of Technology Linear cyclodextrin copolymers
US6245427B1 (en) 1998-07-06 2001-06-12 DüZGüNES NEJAT Non-ligand polypeptide and liposome complexes as intracellular delivery vehicles
CA2361201A1 (en) 1999-01-28 2000-08-03 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded rna
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
EP1159285B1 (en) 1999-03-08 2005-05-25 Metrigen, Inc. Methods and compositions for economically synthesizing and assembling long dna sequences
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
EP1267939A2 (en) * 2000-03-17 2003-01-02 Cell Therapeutics, Inc. Polyglutamic acid-camptothecin conjugates and methods of preparation
AU2001249622B2 (en) * 2000-03-30 2007-06-07 Massachusetts Institute Of Technology RNA sequence-specific mediators of RNA interference
AR028948A1 (en) 2000-06-20 2003-05-28 Astrazeneca Ab NEW COMPOUNDS
ES2728168T3 (en) 2000-12-01 2019-10-22 Max Planck Gesellschaft Small RNA molecules that mediate RNA interference
TWI321054B (en) 2000-12-19 2010-03-01 California Inst Of Techn Compositions containing inclusion complexes
EP2415486B1 (en) * 2001-05-18 2017-02-22 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US20030170891A1 (en) * 2001-06-06 2003-09-11 Mcswiggen James A. RNA interference mediated inhibition of epidermal growth factor receptor gene expression using short interfering nucleic acid (siNA)
AU2002329667A1 (en) * 2001-07-30 2003-02-17 Uta Griesenbach Specific inhibition of gene expression by small double stranded rnas
SE0104251D0 (en) 2001-12-14 2001-12-14 Astrazeneca Ab Novel compounds
DE10302421A1 (en) * 2003-01-21 2004-07-29 Ribopharma Ag New double-stranded interfering RNA, useful for inhibiting hepatitis C virus, has one strand linked to a lipophilic group to improve activity and eliminate the need for transfection auxiliaries
ATE479752T1 (en) * 2003-03-07 2010-09-15 Alnylam Pharmaceuticals Inc THERAPEUTIC COMPOSITIONS
AU2004227414A1 (en) * 2003-04-03 2004-10-21 Alnylam Pharmaceuticals iRNA conjugates
WO2004091515A2 (en) * 2003-04-09 2004-10-28 Alnylam Pharmaceuticals, Inc. iRNA CONJUGATES
US20070179100A1 (en) * 2003-04-09 2007-08-02 Muthiah Manoharan Protected monomers
EP2660322A3 (en) 2003-04-17 2013-11-13 Alnylam Pharmaceuticals Inc. Modified iRNA agents
US8796436B2 (en) * 2003-04-17 2014-08-05 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
AU2004232964B2 (en) * 2003-04-17 2011-09-22 Alnylam Pharmaceuticals, Inc. Protected monomers
US7723509B2 (en) 2003-04-17 2010-05-25 Alnylam Pharmaceuticals IRNA agents with biocleavable tethers
SE0303541D0 (en) 2003-12-22 2003-12-22 Astrazeneca Ab New compounds
EP1791567B1 (en) * 2004-08-10 2015-07-29 Alnylam Pharmaceuticals Inc. Chemically modified oligonucleotides
EP4074344A1 (en) * 2007-12-04 2022-10-19 Arbutus Biopharma Corporation Targeting lipids
AU2009234266B2 (en) 2008-04-11 2015-08-06 Tekmira Pharmaceuticals Corporation Site-specific delivery of nucleic acids by combining targeting ligands with endosomolytic components
AU2009273878A1 (en) 2008-07-25 2010-01-28 Alnylam Pharmaceuticals, Inc. Enhancement of siRNA silencing activity using universal bases or mismatches in the sense strand

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6001991A (en) * 1996-10-04 1999-12-14 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of MDR P-glycoprotein gene expression

Cited By (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10626398B2 (en) 2002-02-01 2020-04-21 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US20030166282A1 (en) * 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
US8524680B2 (en) 2002-02-01 2013-09-03 Applied Biosystems, Llc High potency siRNAS for reducing the expression of target genes
US8815821B2 (en) 2002-02-01 2014-08-26 Life Technologies Corporation Double-stranded oligonucleotides
US9592250B2 (en) 2002-02-01 2017-03-14 Life Technologies Corporation Double-stranded oligonucleotides
US9777275B2 (en) 2002-02-01 2017-10-03 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US9796978B1 (en) 2002-02-01 2017-10-24 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US10036025B2 (en) 2002-02-01 2018-07-31 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US10106793B2 (en) 2002-02-01 2018-10-23 Life Technologies Corporation Double-stranded oligonucleotides
US10196640B1 (en) 2002-02-01 2019-02-05 Life Technologies Corporation Oligonucleotide compositions with enhanced efficiency
US20100075423A1 (en) * 2002-06-12 2010-03-25 Life Technologies Corporation Methods and compositions relating to polypeptides with rnase iii domains that mediate rna interference
US20040248094A1 (en) * 2002-06-12 2004-12-09 Ford Lance P. Methods and compositions relating to labeled RNA molecules that reduce gene expression
US20040033602A1 (en) * 2002-06-12 2004-02-19 Ambion, Inc. Methods and compositions relating to polypeptides with RNase III domains that mediate RNA interference
US8796436B2 (en) 2003-04-17 2014-08-05 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
US10119138B2 (en) 2003-04-17 2018-11-06 Alnylam Pharmaceuticals, Inc. iRNA agents with biocleavable tethers
US20100179309A1 (en) * 2003-04-17 2010-07-15 Alnylam Pharmaceuticals MODIFIED iRNA AGENTS
US8507661B2 (en) 2003-04-17 2013-08-13 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
US11312957B2 (en) 2003-04-17 2022-04-26 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
US8865677B2 (en) 2003-04-17 2014-10-21 Alnylam Pharmaceuticals, Inc. iRNA agents with biocleavable tethers
US8017762B2 (en) 2003-04-17 2011-09-13 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
US8344125B2 (en) 2003-04-17 2013-01-01 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
US20100076056A1 (en) * 2003-04-17 2010-03-25 Alnylam Pharmaceuticals, Inc. MODIFIED iRNA AGENTS
US10676740B2 (en) 2003-04-17 2020-06-09 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
US9476045B2 (en) 2003-04-17 2016-10-25 Alnylam Pharmaceuticals, Inc. iRNA agents with biocleavable tethers
US9394540B2 (en) 2003-04-17 2016-07-19 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
US8426377B2 (en) 2003-04-17 2013-04-23 Alnylam Pharmaceuticals, Inc. iRNA agents with biocleavable tethers
US11015194B2 (en) 2003-04-17 2021-05-25 Alnylam Pharmaceuticals, Inc. iRNA agents with biocleavable tethers
US20050164235A1 (en) * 2003-04-17 2005-07-28 Muthiah Manoharan Modified iRNA agents
US20100267941A1 (en) * 2003-04-17 2010-10-21 Alnylam Pharmaceuticals, Inc. Irna agents with biocleavable tethers
US20100292455A1 (en) * 2003-04-17 2010-11-18 Alnylam Pharmaceuticals MODIFIED iRNA AGENTS
US7851615B2 (en) 2003-04-17 2010-12-14 Alnylam Pharmaceuticals, Inc. Lipophilic conjugated iRNA agents
US7723512B2 (en) 2004-06-30 2010-05-25 Alnylam Pharmaceuticals Oligonucleotides comprising a non-phosphate backbone linkage
WO2006088490A3 (en) * 2004-06-30 2007-03-29 Alnylam Pharmaceuticals Inc Oligonucleotides comprising a non-phosphate backbone linkage
US8013136B2 (en) 2004-06-30 2011-09-06 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a non-phosphate backbone linkage
US20090318676A1 (en) * 2004-06-30 2009-12-24 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a non-phosphate backbone linkage
US20060287260A1 (en) * 2004-06-30 2006-12-21 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a non-phosphate backbone linkage
US7615618B2 (en) 2004-06-30 2009-11-10 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a non-phosphate backbone linkage
US7807815B2 (en) 2004-07-02 2010-10-05 Protiva Biotherapeutics, Inc. Compositions comprising immunostimulatory siRNA molecules and DLinDMA or DLenDMA
US20060025366A1 (en) * 2004-07-02 2006-02-02 Protiva Biotherapeutics, Inc. Immunostimulatory siRNA molecules and uses therefor
US20110097707A1 (en) * 2004-07-21 2011-04-28 Alnylam Pharmaceuticals, Inc. RNAi Agents Comprising Universal Nucleobases
US20110189300A1 (en) * 2004-11-17 2011-08-04 Protiva Biotherapeutics, Inc. siRNA SILENCING OF APOLIPOPROTEIN B
US8058255B2 (en) 2004-12-23 2011-11-15 Applied Biosystems, Llc Methods and compositions concerning siRNA's as mediators of RNA interference
US20060142228A1 (en) * 2004-12-23 2006-06-29 Ambion, Inc. Methods and compositions concerning siRNA's as mediators of RNA interference
US20100159591A1 (en) * 2004-12-23 2010-06-24 Life Technologies Corporation METHODS AND COMPOSITIONS CONCERNING siRNA'S AS MEDIATORS OF RNA INTERFERENCE
US20070054873A1 (en) * 2005-08-26 2007-03-08 Protiva Biotherapeutics, Inc. Glucocorticoid modulation of nucleic acid-mediated immune stimulation
US20090298913A1 (en) * 2005-10-28 2009-12-03 Topigen Pharmaceuticals Inc. Small interfering oligonucleotides comprising arabinose modified nucleotides
US8188263B2 (en) 2005-11-02 2012-05-29 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
US8513403B2 (en) 2005-11-02 2013-08-20 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
US20070135372A1 (en) * 2005-11-02 2007-06-14 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
US8101741B2 (en) 2005-11-02 2012-01-24 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
EP1948674A1 (en) * 2005-11-02 2008-07-30 Protiva Biotherapeutics Inc. Modified sirna molecules and uses thereof
EP1948674A4 (en) * 2005-11-02 2009-02-04 Protiva Biotherapeutics Inc Modified sirna molecules and uses thereof
US9074208B2 (en) 2005-11-02 2015-07-07 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
EP2395012A3 (en) * 2005-11-02 2012-03-14 Protiva Biotherapeutics Inc. Modified siRNA molecules and uses thereof
US20090306178A1 (en) * 2006-03-27 2009-12-10 Balkrishen Bhat Conjugated double strand compositions for use in gene modulation
US7915399B2 (en) 2006-06-09 2011-03-29 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
US20080249046A1 (en) * 2006-06-09 2008-10-09 Protiva Biotherapeutics, Inc. MODIFIED siRNA MOLECULES AND USES THEREOF
WO2008067034A1 (en) * 2006-11-28 2008-06-05 Laird Technologies, Inc. Vehicle-mount antenna assemblies having snap-on outer cosmetic covers with compliant latching mechanisms for achieving zero-gap
US8808747B2 (en) 2007-04-17 2014-08-19 Baxter International Inc. Nucleic acid microparticles for pulmonary delivery
US20090017124A1 (en) * 2007-04-17 2009-01-15 Baxter International Inc. Nucleic Acid Microparticles for Pulmonary Delivery
US9273312B2 (en) 2007-09-19 2016-03-01 Applied Biosystems, Llc SiRNA sequence-independent modification formats for reducing off-target phenotypic effects in RNAi, and stabilized forms thereof
US10329564B2 (en) 2007-09-19 2019-06-25 Applied Biosystems, Llc siRNA sequence-independent modification formats for reducing off-target phenotypic effects in RNAi, and stabilized forms thereof
US10900038B2 (en) 2007-09-19 2021-01-26 Applied Biosystems, Llc siRNA sequence-independent modification formats for reducing off-target phenotypic effects in RNAI, and stabilized forms thereof
US9284551B2 (en) 2007-09-19 2016-03-15 Applied Biosystems, Llc RNAi sequence-independent modification formats, and stabilized forms thereof
US20100222414A1 (en) * 2007-09-19 2010-09-02 Applied Biosystems, Llc SiRNA Sequence-Independent Modification Formats for Reducing Off-Target Phenotypic Effects in RNAi, and Stabilized Forms Thereof
US9771583B2 (en) 2007-09-19 2017-09-26 Applied Biosystems, Llc siRNA sequence-independent modification formats for reducing off-target phenotypic effects in RNAI, and stabilized forms thereof
US8524681B2 (en) 2007-09-19 2013-09-03 Applied Biosystems, Llc siRNA sequence-independent modification formats for reducing off-target phenotypic effects in RNAi, and stabilized forms thereof
US20110028531A1 (en) * 2008-03-20 2011-02-03 Elena Feinstein Novel sirna compounds for inhibiting rtp801
US9035041B2 (en) 2008-09-02 2015-05-19 Alnylam Pharmaceuticals, Inc. Process for triphosphate oligonucleotide synthesis
WO2010028079A2 (en) * 2008-09-02 2010-03-11 Alnylam Pharmaceuticals, Inc Synthetic methods and derivatives of triphosphate oligonucleotides
WO2010028079A3 (en) * 2008-09-02 2010-06-17 Alnylam Pharmaceuticals, Inc Synthetic methods and derivatives of triphosphate oligonucleotides
AU2009288078B2 (en) * 2008-09-02 2014-10-30 Alnylam Pharmaceuticals, Inc. Synthetic methods and derivatives of triphosphate oligonucleotides
US20110009477A1 (en) * 2009-07-08 2011-01-13 Idera Pharmaceuticals, Inc. Oligonucleotide-based compounds as inhibitors of toll-like receptors
US20130130378A1 (en) * 2010-04-22 2013-05-23 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
US10913767B2 (en) * 2010-04-22 2021-02-09 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
US9970002B2 (en) * 2012-12-12 2018-05-15 Massachusetts Institute Of Technology Compositions and methods for functional nucleic acid delivery
US20150322432A1 (en) * 2012-12-12 2015-11-12 1Massachusetts Institute Of Technology Compositions and methods for functional nucleic acid delivery

Also Published As

Publication number Publication date
US20050164235A1 (en) 2005-07-28
US20100179309A1 (en) 2010-07-15
US8344125B2 (en) 2013-01-01
AU2004233092C1 (en) 2010-01-14
CA2522637C (en) 2014-01-21
EP1620544A4 (en) 2011-03-02
US11312957B2 (en) 2022-04-26
US20100292455A1 (en) 2010-11-18
WO2004094595A3 (en) 2007-10-25
JP4597976B2 (en) 2010-12-15
DK1620544T3 (en) 2019-01-14
EP1620544A2 (en) 2006-02-01
US20190345498A1 (en) 2019-11-14
JP2007525454A (en) 2007-09-06
AU2004233092A1 (en) 2004-11-04
EP1620544B1 (en) 2018-09-19
US20050107325A1 (en) 2005-05-19
EP2660322A3 (en) 2013-11-13
US8507661B2 (en) 2013-08-13
CA2522637A1 (en) 2004-11-04
AU2004233092C9 (en) 2010-10-28
US7745608B2 (en) 2010-06-29
ES2702942T3 (en) 2019-03-06
US20220403377A1 (en) 2022-12-22
EP2669377A2 (en) 2013-12-04
WO2004094595A2 (en) 2004-11-04
AU2009213011B2 (en) 2012-01-19
EP2660322A2 (en) 2013-11-06
EP2669377A3 (en) 2015-10-14
AU2004233092B2 (en) 2009-07-09
EP2669377A8 (en) 2014-01-15
EP2664672A1 (en) 2013-11-20
AU2009213011A1 (en) 2009-10-08
EP2666858A1 (en) 2013-11-27

Similar Documents

Publication Publication Date Title
US20050119214A1 (en) Nuclease resistant double-stranded ribonucleic acid
US20190264203A1 (en) Glycoconjugates of rna interference agents
US11015194B2 (en) iRNA agents with biocleavable tethers
US7928217B2 (en) Nuclease resistant double-stranded ribonucleic acid
US8017762B2 (en) Modified iRNA agents
US8541385B2 (en) Chemically modified oligonucleotides for use in modulation micro RNA and uses thereof
US20140045919A1 (en) Folate Conjugates
US20110218334A1 (en) PHOSPHOROTHIOATE OLIGONUCLEOTIDES AND NON-NUCLEOSIDIC PHOSPHOROTHIOATES AS DELIVERY AGENTS FOR iRNA AGENTS
US20160264968A1 (en) MODIFIED iRNA AGENTS
EP3856907A1 (en) Transthyretin (ttr) irna compositions and methods of use thereof for treating or preventing ttr-associated ocular diseases
US20110166198A1 (en) Rna compositions for modulating immune response

Legal Events

Date Code Title Description
AS Assignment

Owner name: ALNYLAM PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MANOHARAN, MUTHIAH;RAJEEV, KALLANTHOTTATHIL G.;REEL/FRAME:015594/0731

Effective date: 20041129

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION