WO2010059226A2 - Inhibition de map4k4 via arni - Google Patents

Inhibition de map4k4 via arni Download PDF

Info

Publication number
WO2010059226A2
WO2010059226A2 PCT/US2009/006211 US2009006211W WO2010059226A2 WO 2010059226 A2 WO2010059226 A2 WO 2010059226A2 US 2009006211 W US2009006211 W US 2009006211W WO 2010059226 A2 WO2010059226 A2 WO 2010059226A2
Authority
WO
WIPO (PCT)
Prior art keywords
rnai construct
nucleotides
sequence
construct
stranded
Prior art date
Application number
PCT/US2009/006211
Other languages
English (en)
Other versions
WO2010059226A3 (fr
Inventor
Tod M. Woolf
Joanne Kamens
Anastasia Khvorova
William Salomon
Original Assignee
Rxi Pharmaceuticals Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Rxi Pharmaceuticals Corporation filed Critical Rxi Pharmaceuticals Corporation
Priority to US13/130,194 priority Critical patent/US9074211B2/en
Publication of WO2010059226A2 publication Critical patent/WO2010059226A2/fr
Publication of WO2010059226A3 publication Critical patent/WO2010059226A3/fr
Priority to US14/728,764 priority patent/US11254940B2/en

Links

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
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/01Phosphotransferases with an alcohol group as acceptor (2.7.1)
    • C12Y207/01037Protein kinase (2.7.1.37)
    • 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
    • C12N2310/32Chemical 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
    • C12N2320/00Applications; Uses
    • C12N2320/10Applications; Uses in screening processes
    • C12N2320/11Applications; Uses in screening processes for the determination of target sites, i.e. of active nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications

Definitions

  • MAP4K4 is a mammalian serine/threonine protein kinase that belongs to a group of protein kinases related to Saccharomyces cerevisiae Sterile 20 (STE20).
  • MAP4K4 also known as NIK for Nek interacting kinase
  • was first identified in a mouse screen for proteins that interact with the SH3 domain of Nek (Su et al. (1997). Since its discovery, MAP4K4 has been and continues to be linked to wide range of physiological functions.
  • MAP4K4 is proposed to link protein tyrosine kinase signals to JNK activation and may play a role in cytoskeletal regulation (Xue et al., Development (2000), 128, 1559-1572).
  • MAP4K4 has been found to be essential for development in mammalian cells.
  • Nik "/- mouse embryos have been show to die postgastrulation. Patterning experiments in mice have suggested that NIK plays a critical and specific role in regulating the migration of cells that arise from the region of the primitive streak just posterior to the node (Xue et al., Development (2000), 128, 1559-1572).
  • MAP4K4 has also been shown to be involved in metabolic disorders. For example, silencing of MAP4K4 resulted in an increase in the expression of a nuclear hormone receptor, PPAR ⁇ , that regulates the expression of genes responsible for adipocyte differentiation (Tang et al., Proc. Natl. Acad. Sci. (2006), 103, 2087-2092). Such genes include, for example, the insulin-responsive facilitative glucose transporter isoform 4 (GLUT4), which mediates insulin-dependent glucose transport into both muscle and adipose tissue. Indeed, several studies have revealed that a MAP4K4-dependent signaling pathway potently inhibits PPAR ⁇ -responsive gene expression, adipogenesis, and insulin-stimulated glucose transport. SUMMARY OF THE INVENTION
  • the present invention is directed in some aspects to RNAi constructs comprising MAP4K4-specific sequences and methods pertaining to their use in MAP4K4 silencing. Accordingly, the present invention provides compositions and methods for increasing the efficiency of inhibiting MAP4K4 expression through RNA interference.
  • the present invention relates to an RNAi construct for inhibiting expression of a MAP4K4 gene, comprising a guide sequence that hybridizes to a target sequence on an mRNA of the MAP4K4 gene and inhibits the expression of the MAP4K4 gene through an RNA interference mechanism, wherein said target sequence is any one of SEQ ID NO. 1, 3, 5, 7, 9, 11, 12, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 53, 54, 55, 56 and 57.
  • the RNAi construct comprises a single-stranded polynucleotide that forms a hairpin structure which includes a double-stranded stem and a single-stranded loop, wherein the double-stranded stem can be cleaved by Dicer to produce an siRNA having a guide sequence that hybridizes to a sequence that is any one of SEQ ID NO. 1 , 3, 5, 7, 9, 11, 12, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 53, 54, 55, 56 and 57.
  • the RNAi construct comprises an siRNA having a guide sequence that hybridizes to a sequence that is any one of SEQ ID NO. 1, 3, 5, 7, 9, 11, 12, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 53, 54, 55, 56 and 57.
  • the RNAi construct comprises an siRNA having a guide sequence that is any one of SEQ ID NO. 2, 4, 6, 8, 10, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, or 52.
  • the RNAi construct comprises a single-stranded polynucleotide that forms a hairpin structure.
  • the hairpin structure includes a double-stranded stem and a single-stranded loop, wherein the double-stranded stem has a 5 '-stem sequence having a 5'-end, and a 3'-stem sequence having a 3'-end.
  • the 5'-stem sequence and at least a portion of said loop form the guide sequence that hybridizes to a sequence that is any one of SEQ ID NO. 1, 3, 5, 7, 9, 1 1, 12, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51 , 53, 54, 55, 56 and 57.
  • the 5'-stem sequence, the loop, and at least a portion of the 3'- stem sequence collectively form the guide sequence that hybridizes to a sequence that is any one of SEQ ID NO. 1, 3, 5, 7, 9, 11, 12, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 53, 54, 55, 56 and 57.
  • the single-stranded polynucleotide is an RNA.
  • At least 12 nucleotides from the 5'-end of the single-stranded polynucleotide are 100% complementary to the target sequence.
  • At least one nucleotide is modified to improve resistance to nucleases, serum stability, target specificity, tissue distribution, and/or cell-permeability of the polynucleotide.
  • the modified nucleotides are modified on the sugar moiety, the base, and/or the phosphodiester linkage.
  • the modification is at position 2 from the 5'-end of the polynucleotide.
  • the modification is a 2'-O-alkyl or 2'-halo group.
  • the modification comprises 2'-O-methyl modification at alternative nucleotides, starting from either the first or the second nucleotide from the 5'-end.
  • the modification comprises 2'-O-methyl modification of one or more randomly selected pyrimidine nucleotides (C or U).
  • the modification comprises 2'-O-methyl modification of one or more nucleotides within the loop.
  • the modification is either limited to one or more nucleotides within the loop, or additionally including 1, 2, 3, 4, 5, or 6 more nucleotide(s) of the guide sequence within the double-stranded stem region just 5' to said loop.
  • the modification comprises 2'-O-methyl modification, wherein no more than 4 consecutive nucleotides are modified.
  • all nucleotides in the 3'-end stem region are modified. In certain embodiments, all nucleotides in the sense sequence are modified.
  • the modification is a phosphate analog. In certain embodiments, the modification is a phosphorothioate linkage.
  • the phosphorothioate linkage is limited to one or more nucleotides within said loop. In another aspect, the phosphorothioate linkage is limited to one or more nucleotides within said loop, and 1, 2, 3, 4, 5, or 6 more nucleotide(s) of the guide sequence within the double-stranded stem region just 5' to said loop.
  • the modification comprises hydophobic modification to one or more bases.
  • said one or more bases are C or G.
  • the hydrophobic modification comprises an isobutyl group.
  • the guide sequence is about 15-21 nucleotides in length, or about 19-21 nucleotides in length.
  • the polynucleotide is 15-29 nucleotides in length, or about 25- 26 nucleotides in length.
  • the hairpin structure is not a substrate for Dicer.
  • the double-stranded stem of the hairpin structure is less than 21 base pairs in length. In another aspect, the double-stranded stem is less than about 20 base pairs in length, or is about 5-15 base pairs in length, or about 10 base pairs in length. In certain embodiments, the double-stranded stem is at least 11 base pairs in length, preferably at least 12 base pairs in length.
  • the single-stranded loop is 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 nucleotides in length.
  • the single-stranded polynucleotide comprises an overhang on the 3'-end and/or an overhang on the 5'-end.
  • the 3'-stem sequence is 100% complementary to the 5'-stem sequence. In other embodiments, the 3'-stem sequence is less than 100% complementary to the 5'-stem sequence.
  • the 3'-stem sequence of the double-stranded stem comprises one or more universal base-pairing nucleotides.
  • the RNAi constructs of the present invention comprise a double-stranded (dsRNA) construct of 25 base pairs in length, wherein the dsRNA comprises (1) a sense strand having a 5'-end and a 3'-end wherein said sense strand corresponds to a target sequence of an mRNA of the MAP4K4 gene, and (2) a guide sequence having a 5'-end and a 3'- end that hybridizes to the sense strand or to the target sequence that is any one of SEQ ID NO.
  • dsRNA double-stranded
  • the dsRNA is blunt-ended.
  • the sense strand comprises one or more consecutive 2'-modified ribose sugar at east of said 5'- and 3'-ends of said sense strand.
  • the sense strand comprises 12 and 10 consecutive 2'-modified ribose sugars at the 5'-end and the 3'- end nucleotides, respectively.
  • the sense strand comprises 4 consecutive 2'- modified ribose sugars at both ends.
  • the 2'-modified ribose sugars are 2'-O-alkyl nucleotides, T- deoxy-2'-fluoro nucleotides, 2'-deoxy nucleotides, 2'-H (deoxyribonucleotides), or combination thereof.
  • the guide sequence of the dsRNA further comprises a 2'- modified ribose sugar.
  • the 2'-ribose sugar is 2'-O- l alkyl nucleotides, 2'-deoxy-2'- fluoro nucleotides, 2'-deoxy nucleotides, 2'-H (deoxyribonucleotides), or combination thereof.
  • the guide sequence of the dsRNA comprises a 2'-modification at the 2 nd nucleotide from the 5'-end of the guide sequence.
  • the guide sequence comprises four consecutive 2 '-modification at the 3'-most end.
  • the MAP4K4 gene is present in a cell. In a related embodiment, the MAP4K4 gene is an endogenous gene.
  • the cell is of eukaryotic origin.
  • the cell is from a mammal, nematode, or insect.
  • the MAP4K4 gene is human or mouse.
  • the target sequence of the present invention is at least 95% identical between the mouse and human sequences.
  • the constructs of the present invention reduce the MAP4K.4 mRNA level by at least 60% of normal level.
  • the constructs of the present invention exhibit low off-target effects.
  • the construct exhibits a seed region frequency of less than 6000, preferably less than 350.
  • the construct of the present invention associates with RISC.
  • the invention provides a vector expressing the RNAi constructs of the present invention. In another aspect, the invention provides a cell comprising the vector expressing the RNAi constructs of the present invention. In another aspect, the invention provides a cell comprising any of the RNAi constructs of the present invention.
  • the cell is a mammalian cell in culture.
  • the cell is a human cell.
  • the present invention also provides a composition comprising any of the RNAi construct described herein and a pharmaceutically acceptable carrier or diluent. Also provided is a method for inhibiting the expression of a MAP4K4 gene in a mammalian cell, comprising contacting the mammalian cell with an RNAi construct according to the invention, or a vector that expresses an RNAi construct as described herein.
  • the mammalian cell is in culture.
  • the mammalian cell is a human cell.
  • the mammalian cell is contacted in the presence of a delivery agent.
  • the delivery reagent comprises a lipid.
  • the lipid is a cationic lipid.
  • the delivery reagent is a liposome.
  • the delivery reagent comprises beta-glucan, chitosan, and/or PEI.
  • RNAi constructs for inhibiting expression of a MAP4K4 gene comprising a guide sequence that hybridizes to a target sequence on an mRNA of the MAP4K4 gene and inhibits the expression of the MAP4K4 gene through an RNA interference mechanism, wherein the RNAi construct is a duplex selected from the group consisting of Duplex numbers 1-25 from Table 1 and 3 is provided.
  • the invention provides a method of treating a patient for a disease characterized by overexpression of a MAP4K4 gene, comprising administering to the patient an RNAi construct according to the present invention, wherein the RNAi construct mediates guide sequence-dependent reduction in MAP4K4 expression.
  • the invention also provides a method of inhibiting expression of a MAP4K4 gene with an RNAi construct according to the present invention, wherein the RNAi construct mediates guide sequence-dependent reduction in MAP4K4 expression.
  • the invention is a composition comprising an oligonucleotide that is any one of SEQ ID NO. 2, 4, 6, 8, 10, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, or 52.
  • the invention is a composition comprising an oligonucleotide that is any one of SEQ ID NO. 1, 3, 5, 7, 9, 11, 12, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 53, 54, 55, 56 or 57.
  • Figure 1 shows an RNA screen across the entire length of the human MAP4K4 gene (NM_004834.3).
  • HEK293 cells were transfected at 0.1 nM active RNA concentration and target mRNA was measured 24 hours post-transfection.
  • Three duplexes: 10, 20 and 21 corresponding to start sites 2337, 2926, and 3028, respectively, of MAP4K4 were chosen based on their activity and homology to the mouse MAP4K4 gene (NM 008696.2).
  • Figure 2A shows an EC 5O analysis of MAP4K4 siRNA duplexes containing sense strands corresponding to SEQ ID NOs:43 or 50 based off of sequences surrounding the duplex 21 site identified in the RNA screen.
  • MAP4K4 expression was measured using a bDNA hybridization assay (Panomics) 48 hours post-transfection.
  • A corresponds to an RNA duplex containing a sense strand corresponding to SEQ ID NO:43
  • “B” corresponds to an RNA duplex containing a sense strand corresponding to SEQ ID NO:50.
  • Figure 2B shows an EC 50 analysis of MAP4K.4 siRNA duplexes containing sense strands corresponding to SEQ ID NOs:41 , 56, 11 or 12 based off of sequences surrounding the duplex 20 site identified in the RNA screen.
  • MAP4K4 expression was measured using a bDNA hybridization assay (Panomics) 48 hours post-transfection.
  • FIG. 1 shows an EC 50 analysis of MAP4K4 siRNA duplexes containing sense strands corresponding to SEQ ID NOs:21, 53, 19 or 54 based off of sequences surrounding the duplex 10 site identified in the RNA screen.
  • MAP4K4 expression was measured using a bDNA hybridization assay (Panomics) 48 hours post-transfection.
  • A corresponds to an RNA duplex containing a sense strand corresponding to SEQ ID NO:21
  • B corresponds to an RNA duplex containing a sense strand corresponding to SEQ ID NO: 53
  • C corresponds to an RNA duplex containing a sense strand corresponding to SEQ ID NO: 19
  • D corresponds to an RNA duplex containing a sense strand corresponding to SEQ ID NO:54.
  • Figure 3 shows an EC 50 analysis of two lead MAP4K4 duplexes: 24 and 25. MAP4K4 expression was measured using a bDNA hybridization assay (Panomics) 48 hours post- transfection.
  • the present invention is related, in part, to compositions and therapeutic methods for the regulation of MAP4K4 activity.
  • the invention is directed to methods of identifying agents and the use of identified agents for regulating MAP4K4 in dysregulation, or in conditions that may benefit from silencing of MAP4K4 to below physiological levels.
  • MAP4K4 silencing through the use of certain MAP4K4 sequence-specific RNAi constructs which exhibit unexpectedly high gene silencing activity.
  • the present invention also includes methods for identifying sequences within a MAP4K4 gene that can be used to design effective RNAi constructs.
  • RNAi constructs of the present method are designed and manipulated according to the methods described herein to yield highly stable and potent constructs with low levels of off-targeting effects.
  • the invention is directed to RNAi constructs for inhibiting the expression of
  • RNAi construct comprises a guide sequence that hybridizes to a target sequence on an mRNA of the MAP4K4 gene and inhibits the expression of the MAP4K4 gene through an RNAi interference mechanism, and wherein the target sequence that is any one of SEQ ID NO. 1, 3, 5, 7, 9, 1 1, 12, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 53, 54, 55, 56 and 57. Described below are a few exemplary constructs that can be used in the context of the subject sequences and be used in the methods of the invention described herein. It is understood, however, that any given types of RNAi construct known in the art may be applicable.
  • the present invention relates to an RNAi construct that comprises a single-stranded polynucleotide that forms a hairpin structure, which includes a double-stranded stem and a single-stranded loop, wherein the double-stranded stem can be cleaved by Dicer to produce an siRNA having a guide sequence that hybridizes to a sequence that is any one of SEQ ID NO. 1, 3, 5, 7, 9, 11, 12, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 53, 54, 55, 56 and 57.
  • the present invention includes an siRNA (such as a duplex of 19 nucleotides, with or without 3 '-overhangs), as described in the art, comprising a guide sequence that hybridizes to a sequence that is any one of SEQ ID NO. 1, 3, 5, 7, 9, 11, 12, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 53, 54, 55, 56 and 57 under physiological condition of a cell or under high stringency hybridization condition.
  • siRNA such as a duplex of 19 nucleotides, with or without 3 '-overhangs
  • the invention provides a double-stranded RNA (dsRNA) construct, preferably of 25 base pairs in length, for inhibiting expression of a MAP4K4 gene.
  • dsRNA construct comprises a sense strand having a 5 '-end and a 3 '-end that corresponds to a region within an mRNA transcript of the MAP4K4 gene, and an antisense strand having a 5'-end and a 3 '-end that hybridizes to the sense strand under physiological conditions of a cell or under high stringency hybridization conditions.
  • the sense strand comprises a sequence that is any one of SEQ ID NO.
  • the dsRNA construct of the present invention inhibits expression of MAP4K4 in a sequence-dependent manner.
  • the sense strand comprises one or more consecutive 2'-modified ribose sugar at each of said 5'- and 3'-ends of said sense strand. In certain preferred embodiments, the sense strand comprises 12 and 10 consecutive 2'-modified ribose sugar at each of said 5'-end and 3'-end nucleotides, respectively. In certain embodiments, each end of the sense strand comprises a continuous stretch of 2'-modified ribose sugars, although each end may have the same number or different numbers of 2'-modified ribose sugars. In other aspects, the sense strand comprises 4 consecutive 2'-modified ribose sugars at both ends. For a 25-mer construct, each end of the sense strand may comprise, independently, 4-16
  • each end of the sense strand may comprise, independently, 4-18 2'- modified nucleotides and/or phosphothioate linkages, etc.
  • the antisense strand comprises an antisense sequence that is any one of SEQ ID NO. 2, 4, 6, 8, 10, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, or 52
  • the antisense strand is unmodified.
  • the antisense (guide) strand further comprises a 2'-modif ⁇ ed ribose sugar. Such T- modification of the antisense strand is at the 2 nd nucleotide from the 5'end of the antisense strand.
  • the antisense strand comprises four consecutive 2'-modification at the 3'-most end of the antisense strand.
  • 2'-modif ⁇ ed ribose sugar includes those ribose sugars that do not have a 2'-OH group.
  • the 2'-modified ribose sugar may be 2'-O-alkyl nucleotides, T- deoxy-2'-fluoro nucleotides, 2'-deoxy nucleotides, 2'-H (deoxyribonucleotides), or combination thereof.
  • the 2' -modified nucleotides are pyrimidine nucleotides (e.g., C AJ).
  • the 2'-O-alkyl nucleotides may be, for example, 2'-O-methyl nucleotides, or 2'-O-allyl nucleotides.
  • dsRNA constructs of the present invention comprise MAP4K4 sequences of human or mouse origin. Additionally, in certain preferred embodiments, the sense strands of the dsRNA correspond to regions of the MAP4K4 mRNA sequence that is at least about 80%, 85%, 90%, 95%, 97%, or 99% identical between the mouse and human sequences. In other aspects of the invention, the dsRNA construct significantly (e.g., at least about
  • the dsRNA construct reduces a MAP4K4 mRNA level by at least 60% of a normal level.
  • the dsRNA of the invention with the above-referenced modifications exhibits significantly (e.g., at least about 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or more) less "off-target” gene silencing when compared to similar constructs without the specified antisense modification, thus greatly improving the overall specificity of the RNAi reagent or therapeutics.
  • off-target gene silencing refers to unintended gene silencing due to, for example, spurious sequence homology between the antisense (guide) sequence and the unintended target mRNA sequence.
  • a suitable dsRNA MAP4K4 sequence to be used as an siRNA construct in the present invention adheres to what has been described as having a low or moderate seed complement frequency (SCF).
  • SCF seed complement frequency
  • siRNA sequences with low ( ⁇ 350) or moderate ( ⁇ 350-6000) incidents of complementation with the 3' UTR exhibit less off-targeting effects.
  • the subject MAP4K4-specific RNAi agents have low to moderate SCF. It should also be understood, however, that while low or moderate SCF is desirable, it is not essential for reduced off-targeting effects. As such, in certain embodiments, a sequence may fall within the range of high SCF (>6000) and still exhibits low off-targeting effects.
  • certain sense or antisense modifications further increase nuclease stability, and/or lower interferon induction, without a significant decrease in RNAi activity (or no decrease in RNAi activity at all).
  • the dsRNA construct associates with an RNA- induced silencing complex (RISC).
  • RISC RNA- induced silencing complex
  • the dsRNA construct of the present invention is blunt-ended.
  • 5'- and/or 3 '-end overhangs of 1-4 nucleotides may be present on one or both strands.
  • the present invention contemplates a composition comprising the dsRNA described herein and a pharmaceutically acceptable carrier or diluent.
  • Another aspect of the invention provides a method for inhibiting the expression of a MAP4K4 gene in a mammalian cell, comprising contacting the mammalian cell with any of the subject dsRNA constructs.
  • Another aspect of the invention provides a method for inhibiting the expression of a MAP4K4 gene in a mammalian cell, comprising contacting the mammalian cell with a vector expressing at least one strand of any of the subject dsRNA constructs. The method may be carried out in vitro or in vivo, in, for example, mammalian cells in culture, such as a human cell in culture.
  • the target cells may be contacted in the presence of a delivery reagent, such as a lipid (e.g., a cationic lipid), a liposome, beta-glucan, chitosan, polyethyleneimine (PEI), or any combination thereof.
  • a delivery reagent such as a lipid (e.g., a cationic lipid), a liposome, beta-glucan, chitosan, polyethyleneimine (PEI), or any combination thereof.
  • Another aspect of the invention provides a method for inhibiting the expression of a target gene in a mammalian cell, comprising contacting the mammalian cell with a vector expressing at least one strand of the dsRNA according to the present methods.
  • the antisense strand may comprise a 2'-modified ribose sugar, such as 2'-O-methyl modified nucleotide, at the 2 n nucleotide on the 5 '-end of the antisense strand and, preferably no other modified nucleotides.
  • the dsRNA having such modification may have enhanced target specificity or reduced off- target silencing compared to a similar construct without the 2'-O-methyl modification at said position.
  • the antisense strand may comprise at least four consecutive 2'-modified ribose sugars, such as 2'-O-methyl modified, 3'-end nucleotides with non-hydrolyzable internucleotide linkages, such as phosphothioate linkages.
  • the 5 '-end 12 nucleotides and the 3 '-end 10 nucleotides of a 25-mer may be 2'-modified ribose sugars.
  • the number of the modified bases may be adjusted depending on the overall length of the construct.
  • the 5'-end 12-14 nucleotides and the 3'-end 10-12 nucleotides may be 2'-modified nucleotides, etc.
  • the dsRNA may comprise a mismatch nucleotide at the 2 nd nucleotide from the 3 '-end of the sense strand.
  • certain sense strand sequences may be cleaved by the RISC complex loaded with the Dicer-resistant guide sequence, at the position where an equivalent mRNA is cleaved. While not wishing to be bound by any particular theory, this is partly because the sense strand share the same or similar sequence as the target mRNA. Therefore, the subject dsRNA constructs include those with a sense strand that can be cleaved between the 10th and 1 lth 3 '-end nucleotides.
  • the constructs of the invention may have different lengths.
  • the preferred lengths of the construct are 19-49 nucleotides in length.
  • the length of the construct is greater than or equal to 22 nucleotides in length.
  • the length of the construct is greater than or equal to 25 nucleotides in length.
  • the length of the construct is 26, 27, 28, 29, 30, or 31 -49 nucleotides in length.
  • Other lengths are also possible, so long as the lower length limit is the minimal length for a Dicer substrate, and the upper limit is a length that generally will not trigger PKR response in a target cell.
  • modifications may alter that upper limit such that longer lengths (such as 50, 60, 70, 80, 90, 100 bp) are tolerable.
  • the modified dsRNA may have improved stability in serum and/or cerebral spinal fluid compared to an unmodified dsRNA having the same sequence.
  • the dsRNA does not induce interferon response in primary cells, such as mammalian primary cells, including primary cells from human, mouse and other rodents, and other non-human mammals.
  • the dsRNA may also be used to inhibit expression of a target gene in an invertebrate organism.
  • either end of the sense strand and/or the 3 '-end of the antisense strand may be blocked by protective group(s).
  • protective groups such as inverted nucleotides, inverted abasic moieties, or amino-end modified nucleotides may be used.
  • Inverted nucleotides may comprise an inverted deoxynucleotide.
  • Inverted abasic moieties may comprise an inverted deoxyabasic moiety, such as a 3',3'-linked or 5',5'-linked deoxyabasic moiety.
  • alternating nucleotides on the ends of the sense and/or antisense strands comprise 2' -modified ribose sugars, and wherein each of the 2' -modified ribose sugars faces an unmodified nucleotide on the opposite strand.
  • the first T- modified antisense nucleotide is the most 5 '-end antisense nucleotide or the 2nd nucleotide from the 5 '-end of the antisense strand.
  • the subject double-stranded RNA may be chemically cross- linked at one or more points, or linked by a nucleotide loop structure at one or both ends (e.g. , a single-stranded hairpin structure or a circular structure).
  • the chemical cross-link or the loop of the hairpin structure is at the 3 '-end of the antisense strand (e.g., linking the 3'-end of the antisense strand to the 5'-end of the sense strand).
  • the chemical cross-link or the loop of the hairpin structure is at the 5 '-end of the antisense strand (e.g.
  • Another aspect of the invention provides a method for improving the gene silencing effect of a small interference RNA (siRNA), comprising modifying the sense and/or antisense nucleotides of the siRNA to become any of the subject dsRNA constructs.
  • siRNA small interference RNA
  • the subject MAP4K4 sequences are incorporated in the context of a hairpin structure. It has been previously described that a hairpin structure formed from a single-stranded polynucleotide does not require processing, and indeed is not processed by Dicer or other Dicer-like Rnase III enzymes to participate in (RISC- mediated) RNA interference. See U.S. Provisional Application No. 61/135,855, filed July 24, 2008, entitled “SHORT HAIRPIN RNAi CONSTRUCTS AND USES THEREOF” and U.S. Provisional Application filed on October 30, 2008, entitled “miniRNA CONSTRUCTS AND USES THEREOF", incorporated herein by reference in their entire contents.
  • the guide strand of such a hairpin structure becomes the single species of active RNAi reagent, and thus facilitates the development of RNAi reagents or therapeutics with higher target specificity, and better-defined biological activity and/or pharmacological property.
  • Another advantages of the miniRNA is the presence of single-stranded region (loop region).
  • single-stranded polynucleotides are a better substrates for cellular uptake.
  • the single-stranded (loop) region is chemically modified.
  • the chemical modification may comprise phosphothioate.
  • the chemical modification comprises 2'0ME or 2' Fluoro or 2' deoxy.
  • the chemical modification is a combination of phosphorothioates with 2' OMe and 2' Fluoro.
  • the loop may be completely or partially replaced by a chemical linker that is flexible enough to allow folding back of the duplex polynucleotide.
  • the invention provides a single-stranded polynucleotide miniRNA construct of 15-49 nucleotides in length, for inhibiting expression of a MAP4K4 gene, said polynucleotide comprising: (1) a 5'-stem sequence having a 5'-end, and (2) a 3'-stem sequence having a 3'-end, said 5'-stem sequence and 3'-stem sequence being sufficiently complementary to form a double-stranded stem, and (3) a single-stranded loop bridging the 5'- stem and 3'-stem sequences.
  • the single-stranded loop may be 2-15 nucleotides in length, or preferably 4, 5, 6, 7, 8, 9, 10, or 11 nucleotides in length.
  • the 5'-stem sequence (including any 5'-end overhangs) and at least a portion or all of said single-stranded loop form a guide strand / sequence that is complementary to a transcript of a target gene.
  • the single-stranded polynucleotide miniRNA structure may be (1) resistant to cleavage by Dicer, (2) associates with RISC, and/or (c) inhibits expression of the target gene in a guide sequence-dependent manner.
  • the invention provides a single-stranded polynucleotide miniRNA construct of 15-49 nucleotides in length, for inhibiting expression of a MAP4K4 gene, said polynucleotide comprising: (1) a 5'-stem sequence having a 5'-end, and (2) a 3'-stem sequence having a 3'-end, said 5'-stem sequence and 3'-stem sequence being sufficiently complementary to form a double-stranded stem, and (3) a single-stranded loop bridging the 5'-stem and 3'-stem sequences.
  • the single-stranded loop may be 2-15 nucleotides in length, or preferably 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 nucleotides in length, or more preferably 4, 5, 6, 7 nucleotides in length. In certain embodiments, the single-stranded loop is 4 or 6 nucleotides in length. In unmodified forms, the preferred length of the miniRNA stem region is around 10-13 bp. In certain embodiments, the double-stranded stem region is at least 11 bp in length, preferably at least 12 bp in length.
  • the guide sequence hybridizes to a target sequence on an mRNA of the MAP4K4 gene, wherein the target sequence is that is any one of SEQ ID NO. 1, 3, 5, 7, 9, 11, 12, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 50, 51, 53, 54, 55, 56 and 57.
  • duplex / stem length limitation may be partially defined by thermodynamic stability in cellular environments.
  • a group of chemical modifications known to enhance thermodynamic stability of a duplex region may be used to alter stem length.
  • a non-limiting example of these chemical modifications might be LNA (locked nucleic acid) or MGB (minor groove binder).
  • LNA locked nucleic acid
  • MGB mimerase binder
  • stabilizing chemical modifications might be applied to the duplex region of short miniRNAs and convert otherwise non-functional entities to functional ones.
  • the modification is in a non-guide sequence region.
  • IR inverted repeats
  • the 5'-stem sequence (including any 5'-end overhangs), the single-stranded loop, and at least a portion or all of the 3'-stem sequence form a guide strand / sequence that is complementary to a transcript of a MAP4K4 gene.
  • the single-stranded polynucleotide miniRNA structure may be (1) resistant to cleavage by Dicer, (2) associates with RISC, and/or (c) inhibits expression of the MAP4K4 gene in a guide sequence-dependent manner.
  • the polynucleotide does not contain any overhangs.
  • 5'- and/or 3'-end overhangs of 1-6 nucleotides may be present on one or both ends of the polynucleotide.
  • the number and/or sequence of nucleotides overhang on one end of the polynucleotide may be the same or different from the other end of the polynucleotide.
  • one or more of the overhang nucleotides may contain chemical modification(s), such as phosphorothioate or 2'-OMe modification.
  • the polynucleotide is unmodified. In other embodiments, at least one nucleotide is modified. In further embodiments, the modification includes a 2'-H or 2'-modified ribose sugar at the 2 nd nucleotide from the 5'-end of the guide sequence.
  • the "2nd nucleotide” is defined as the second nucleotide from the 5'-end of the polynucleotide.
  • 2' -modified ribose sugar includes those ribose sugars that do not have a 2'-OH group.
  • “2'-modified ribose sugar” does not include 2'-deoxyribose (found in unmodified canonical DNA nucleotides).
  • the 2'-modified ribose sugar may be T- O-alkyl nucleotides, 2'-deoxy-2'-fluoro nucleotides, 2'-deoxy nucleotides, or combination thereof.
  • the 2 > -modified nucleotides are pyrimidine nucleotides ⁇ e.g., C /U).
  • Examples of 2'-0-alkyl nucleotides include 2'-O-methyl nucleotides, or 2'-O-allyl nucleotides.
  • the miniRNA polynucleotide of the invention with the above- referenced 5'-end modification exhibits significantly ⁇ e.g., at least about 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or more) less "off-target" gene silencing when compared to similar constructs without the specified 5 '-end modification, thus greatly improving the overall specificity of the RNAi reagent or therapeutics.
  • certain guide strand modifications further increase nuclease stability, and/or lower interferon induction, without significantly decreasing RNAi activity (or no decrease in RNAi activity at all).
  • the 5'-stem sequence may comprise a 2' -modified ribose sugar, such as 2'-O-methyl modified nucleotide, at the 2" nucleotide on the 5'-end of the polynucleotide and, preferably no other modified nucleotides.
  • the hairpin structure having such modification may have enhanced target specificity or reduced off-target silencing compared to a similar construct without the 2'-O-methyl modification at said position.
  • the constructs of the invention may have different lengths.
  • the preferred lengths of the miniRNA construct are 15-49 nucleotides in length.
  • the length of the miniRNA construct is 25 or 26 nucleotides in length.
  • Other lengths are also possible, so long as the double-stranded stem does not exceed a maximum length causing it to be a Dicer substrate.
  • the maximum length of the double-stranded stem does not exceed 21 base pairs.
  • the maximum length of the double-stranded stem does not exceed 19 base pairs.
  • the double-stranded stem may be shorter than 10 base pairs without negatively affecting the RNAi capability of the hairpin construct.
  • the length of the single-stranded loop may be varied to allow for enhanced stability, for example.
  • the guide strand comprises a 2'-O-methyl modified nucleotide at the 2 nd nucleotide on the 5 '-end of the guide strand and no other modified nucleotides.
  • the modified hairpin structure may have improved stability in serum and/or cerebral spinal fluid compared to an unmodified hairpin structures having the same sequence.
  • At least the first 10 nucleotides from the 5 '-end of the polynucleotide are 100% complementary to the MAP4K4 gene transcript. More preferably, at least the first 12 nucleotides from the 5'-end of the polynucleotide are 100% complementary to the MAP4K4 gene transcript. In certain preferred embodiments, about the first 12 to 15 nucleotides from the 5'-end of the polynucleotide are 100% complementary to the MAP4K4 gene transcript.
  • nucleotides 2 to 17 of the guide sequence / strand is complementary to the MAP4K4 sequence.
  • the sequence complementarity may be partial, preferably, the guide sequence can hybridize to the MAP4K4 sequence under the physiological condition of the cell or under high stringency condition.
  • a nucleic acid molecule is "hybridizable" to another nucleic acid molecule, such as a cDNA, genomic DNA, or RNA, when a single-stranded form of the nucleic acid molecule can anneal to the other nucleic acid molecule under the appropriate conditions of temperature and solution ionic strength (see Sambrook et al. Molecular Cloning: A Laboratory Manual, Second Edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y.). The conditions of temperature and ionic strength determine the "stringency" of the hybridization.
  • Low stringency hybridization conditions correspond to a T m (melting temperature) of 55° C, e.g., 5> ⁇ SSC, 0.1% SDS, 0.25% milk, and no formamide; or 30% formamide, 5 ⁇ SSC, 0.5% SDS.
  • Moderate stringency hybridization conditions correspond to a higher T m , e.g., 40% formamide, with 5* or 6xSSC.
  • High stringency hybridization conditions correspond to the highest T m , e.g., 50% formamide, 5x or 6> ⁇ SSC.
  • SSC is 0.15 M NaCl, 0.015 M Na-citrate.
  • High stringency conditions are understood to encompass conditions of hybridization which allow hybridization of structurally related, but not structurally dissimilar, nucleic acids.
  • stringent is a term of art which is understood by the skilled artisan to describe any of a number of alternative hybridization and wash conditions which allow annealing of only highly complementary nucleic acids.
  • Exemplary high stringent hybridization conditions is equivalent to about 20-27° C. below the melting temperature (T m ) of the DNA duplex formed in about 1 M salt.
  • T m melting temperature
  • Hybridization requires that the two nucleic acids contain complementary sequences, although depending on the stringency of the hybridization, mismatches between bases are possible.
  • the appropriate stringency for hybridizing nucleic acids depends on the length of the nucleic acids and the degree of complementation, variables well known in the art. The greater the degree of similarity or homology between two nucleotide sequences, the greater the value of T m , for hybrids of nucleic acids having those sequences.
  • the relative stability (corresponding to higher T m ) of nucleic acid hybridizations decreases in the following order: RNA:RNA, DNA:RNA, DNA:DNA.
  • a minimum length for a hybridizable nucleic acid is at least about 10 nucleotides; preferably at least about 15 nucleotides; and more preferably the length is at least about 20 nucleotides.
  • standard hybridization conditions or “physiological conditions” refers to a T n , of about 55° C, and utilizes conditions as set forth above.
  • the T n is 60° C; in a more preferred embodiment, the T n , is 65° C.
  • high stringency refers to hybridization and/or washing conditions at 68° C. in 0.2 ⁇ SSC, at 42° C. in 50% formamide, 4> ⁇ SSC, or under conditions that afford levels of hybridization equivalent to those observed under either of these two conditions.
  • Suitable hybridization conditions for oligonucleotides are typically somewhat different than for full-length nucleic acids (e.g., full-length cDNA), because of the oligonucleotides' lower melting temperature. Because the melting temperature of oligonucleotides will depend on the length of the oligonucleotide sequences involved, suitable hybridization temperatures will vary depending upon the oligonucleotide molecules used. Exemplary temperatures may be 37° C. (for 14-base oligonucleotides), 48° C. (for 17-base oligonucleotides), 55° C.
  • oligonucleotides for 20-base oligonucleotides and 60° C. (for 23-base oligonucleotides).
  • exemplary suitable hybridization conditions for oligonucleotides include washing in 6 ⁇ SSC, 0.05% sodium pyrophosphate, or other conditions that afford equivalent levels of hybridization.
  • the hairpin structure does not induce interferon response in primary cells, such as mammalian primary cells, including primary cells from human, mouse and other rodents, and other non-human mammals.
  • the hairpin structure may also be used to inhibit expression of a MAP4K4 gene in an invertebrate organism.
  • the 3'-end of the hairpin structure may be blocked by protective group(s).
  • protective groups such as inverted nucleotides, inverted abasic moieties, or amino-end modified nucleotides may be used.
  • Inverted nucleotides may comprise an inverted deoxynucleotide.
  • Inverted abasic moieties may comprise an inverted deoxyabasic moiety, such as a 3',3'-linked or 5',5'-linked deoxyabasic moiety.
  • the invention includes methods to inhibit expression of a MAP4K4 gene either in a cell in vitro, or in vivo.
  • the polynucleotide hairpin constructs of the invention are useful for treating a patient with a disease characterized by the overexpression of a MAP4K.4 gene.
  • the polynucleotide hairpin constructs of the invention are useful for treating a condition that would benefit from inhibition of a MAP4K4 gene below physiological levels.
  • the MAP4K4 gene can be endogenous or exogenous (e.g., introduced into a cell by a virus or using recombinant DNA technology) to a cell.
  • Such methods may include introduction of RNA into a cell in an amount sufficient to inhibit expression of the MAP4K4 gene, where the RNA is a hairpin structure.
  • RNA may have a guide strand that is complementary to the nucleotide sequence of the MAP4K.4 gene, such that the composition inhibits expression of the MAP4K4 gene.
  • the guide strand may be formed by the 5'-stem sequence (including any 5'-end overhangs) and all or a portion of the single-stranded loop region.
  • the guide strand may be formed by the 5 '-stem sequence (including any 5 '-end overhangs), the entire loop region, and all or a portion of the 3'-stem sequence.
  • the invention also relates to vectors expressing the subject hairpin constructs, and cells comprising such vectors or the subject hairpin constructs.
  • the cell may be a mammalian cell in vivo or in culture, such as a human cell.
  • the invention further relates to compositions comprising the subject hairpin constructs, and a pharmaceutically acceptable carrier or diluent.
  • Another aspect of the invention provides a method for inhibiting the expression of a target gene in a mammalian cell, comprising contacting the mammalian cell with any of the subject hairpin constructs.
  • the method may be carried out in vitro, ex vivo, or in vivo, in, for example, mammalian cells in culture, such as a human cell in culture.
  • the target cells ⁇ e.g., mammalian cell
  • the target cells may be contacted in the presence of a delivery reagent, such as a lipid (e.g., a cationic lipid) or a liposome.
  • a delivery reagent such as a lipid (e.g., a cationic lipid) or a liposome.
  • RNAi structures of the present invention mediate sequence-dependent gene silencing by a microRNA mechanism.
  • miRNA small temporal RNAs
  • shRNAs small temporal RNAs
  • miRNA disorder shall refer to a disease or disorder characterized by an aberrant expression or activity of an miRNA.
  • microRNAs are involved in down-regulating target genes in critical pathways, such as development and cancer, in mice, worms and mammals. Gene silencing through a microRNA mechanism is achieved by specific yet imperfect base-pairing of the miRNA and its target messenger RNA (mRNA). Various mechanisms may be used in microRNA-mediated down- regulation of target mRNA expression.
  • miRNAs are noncoding RNAs of approximately 22 nucleotides which can regulate gene expression at the post transcriptional or translational level during plant and animal development.
  • miRNAs are all excised from an approximately 70 nucleotide precursor RNA stem-loop termed pre-miRNA, probably by Dicer, an RNase Ill-type enzyme, or a homolog thereof.
  • Naturally-occurring miRNAs are expressed by endogenous genes in vivo and are processed from a hairpin or stem-loop precursor (pre-miRNA or pri- miRNAs) by Dicer or other RNAses.
  • miRNAs can exist transiently in vivo as a double-stranded duplex but only one strand is taken up by the RISC complex to direct gene silencing.
  • RNA interference pathway Another pathway that uses small RNAs as sequence-specific regulators is the RNA interference (RNAi) pathway, which is an evolutionarily conserved response to the presence of double-stranded RNA (dsRNA) in the cell.
  • dsRNA double-stranded RNA
  • the dsRNAs are cleaved into ⁇ 20-base pair (bp) duplexes of small-interfering RNAs (siRNAs) by Dicer. These small RNAs get assembled into multiprotein effector complexes called RNA- induced silencing complexes (RJSCs).
  • RJSCs RNA- induced silencing complexes
  • RNAi constructs may mimic the dsRNA in the siRNA mechanism, or the microRNA in the miRNA mechanism.
  • a MAP4K4 gene or protein includes the full-length version as well as any naturally occurring fragments thereof. Additionally, a MAP4K4 gene or protein includes any naturally occurring isoforms of MAP4K4.
  • Double-stranded oligonucleotides of the invention may be formed by two separate complementary nucleic acid strands. Duplex formation can occur either inside or outside the cell containing the target gene.
  • double-stranded includes one or more nucleic acid molecules comprising a region of the molecule in which at least a portion of the nucleomonomers are complementary and hydrogen bond to form a duplex.
  • Double-stranded oligonucleotides of the invention may comprise a nucleotide sequence that is sense to a target gene and a complementary sequence that is antisense to the target gene.
  • the sense and antisense nucleotide sequences correspond to the target gene sequence, e.g., are identical or are sufficiently identical to effect target gene inhibition (e.g., are about at least about 98% identical, 96% identical, 94%, 90% identical, 85% identical, or 80% identical) to the target gene sequence.
  • the double-stranded oligonucleotide of the invention is double- stranded over its entire length, i.e., with no overhanging single-stranded sequence at either end of the molecule, i.e., is blunt-ended.
  • the individual nucleic acid molecules can be of different lengths.
  • a double-stranded oligonucleotide of the invention is not double-stranded over its entire length.
  • one of the molecules e.g., the first molecule comprising an antisense sequence, can be longer than the second molecule hybridizing thereto (leaving a portion of the molecule single-stranded).
  • a double-stranded oligonucleotide of the invention contains mismatches and/or loops or bulges, but is double-stranded over at least about 70% of the length of the oligonucleotide. In another embodiment, a double-stranded oligonucleotide of the invention is double-stranded over at least about 80% of the length of the oligonucleotide.
  • a double-stranded oligonucleotide of the invention is double-stranded over at least about 90%-95% of the length of the oligonucleotide. In another embodiment, a double- stranded oligonucleotide of the invention is double-stranded over at least about 96%-98% of the length of the oligonucleotide. In certain embodiments, the double-stranded oligonucleotide of the invention contains at least or up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, or 15 mismatches. Hairpin Characteristics
  • the hairpin structures of the present invention include a nucleic acid comprising a single-stranded RNA, such as a shRNA.
  • the hairpin structure can include a double-stranded stem region formed from a 5'-stem sequence having a 5'-end ("5'-stem sequence") and a 3'-stem sequence having a 3'-end ("3'-stem sequence") that is complementary to the 5'-stem sequence.
  • the hairpin structure can further include a single-stranded loop region.
  • the single-stranded polynucleotide is a DNA strand comprising one or more modified deoxyribonucleotides.
  • the single-stranded polynucleotide is an XNA strand, such as a peptide nucleic acid (PNA) strand or locked nucleic acid (LNA) strand.
  • the single-stranded polynucleotide is a DNA/RNA hybrid.
  • the 5 '-stem sequence and 3 '-stem sequence are at least substantially complementary to each other, and more preferably about 100% complementary to each other. More preferably, the 5'-stem sequence and 3'-stem sequence are each 5 tol9 nucleotides, inclusive, in length. Alternatively, the 5 '-stem sequence and 3 '-stem sequence are each 10 to 19 nucleotides, inclusive, in length.
  • the 5'-stem sequence and 3'-stem sequence within any hairpin of the invention can be the same length, or differ in length by less than about 5 bases, which as persons skilled in the art are aware can appear in a hairpin structure as one or more bulge(s).
  • the loop structure is about 2 to 15 nucleotides in length, and more preferably 4-11 nucleotides.
  • overhangs, if any comprise between 1 to 6 bases.
  • the overhangs can be unmodified, or can contain one or more specificity or stabilizing modifications, such as a halogen or O-alkyl modification of the 2' position, or internucleotide modifications such as phosphorothioate, phosphorodithioate, or methylphosphonate modifications.
  • the overhangs can be ribonucleic acid, deoxyribonucleic acid, or a combination of ribonucleic acid and deoxyribonucleic acid.
  • the modification(s) to the 5'-terminal nucleotide if any, does not affect the RNAi capability of the hairpin construct.
  • Such a modification can be, for example, a phosphorothioate.
  • double-stranded stem includes one or more nucleic acid molecules comprising a region of the molecule in which at least a portion of the nucleomonomers are complementary and hydrogen bond to form a double-stranded region.
  • the 3'-stem sequence comprises one or more universal base- pairing nucleotides.
  • a double-stranded stem of the hairpin construct contains mismatches and/or loops or bulges, but is double-stranded over at least about 50% of the length of the double-stranded stem.
  • a double-stranded stem of the invention is double-stranded over at least about 60% of the length of the stem.
  • a double-stranded stem of the hairpin construct is double-stranded over at least about 70% of the length of the stem.
  • a double-stranded stem of the hairpin is double- stranded over at least about 80% of the length of the stem.
  • a double- stranded stem of the hairpin construct is double-stranded over at least about 90%-95% of the length of the double-stranded stem. In another embodiment, a double-stranded stem of the hairpin construct is double-stranded over at least about 96%-98% of the length of the stem. In certain embodiments, the double-stranded stem of the hairpin construct contains at least or up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 mismatches.
  • the nucleotides of the invention may be modified at various locations, including the sugar moiety, the phosphodiester linkage, and/or the base.
  • Sugar moieties include natural, unmodified sugars, e.g., monosaccharide (such as pentose, e.g. , ribose, deoxyribose), modified sugars and sugar analogs.
  • possible modifications of nucleomonomers, particularly of a sugar moiety include, for example, replacement of one or more of the hydroxyl groups with a halogen, a heteroatom, an aliphatic group, or the functionalization of the hydroxyl group as an ether, an amine, a thiol, or the like.
  • modified nucleomonomers are 2'-O-methyl nucleotides. Such 2'-O-methyl nucleotides may be referred to as "methylated,” and the corresponding nucleotides may be made from unmethylated nucleotides followed by alkylation or directly from methylated nucleotide reagents. Modified nucleomonomers may be used in combination with unmodified nucleomonomers. For example, an oligonucleotide of the invention may contain both methylated and unmethylated nucleomonomers.
  • modified nucleomonomers include sugar- or backbone-modified ribonucleotides.
  • Modified ribonucleotides may contain a nonnaturally occurring base (instead of a naturally occurring base), such as uridines or cytidines modified at the 5'-position, e.g., 5'- (2-amino)propyl uridine and 5'-bromo uridine; adenosines and guanosines modified at the 8- position, e.g., 8-bromo guanosine; deaza nucleotides, e.g., 7-deaza-adenosine; and N-alkylated nucleotides, e.g., N6-methyl adenosine.
  • uridines or cytidines modified at the 5'-position, e.g., 5'- (2-amino)propyl uridine and 5'-bromo uridine
  • sugar-modified ribonucleotides may have the 2'- OH group replaced by a H, alxoxy (or OR), R or alkyl, halogen, SH, SR, amino (such as NH 2 , NHR, NR 2j ), or CN group, wherein R is lower alkyl, alkenyl, or alkynyl.
  • Modified ribonucleotides may also have the phosphoester group connecting to adjacent ribonucleotides replaced by a modified group, e.g., of phosphothioate group. More generally, the various nucleotide modifications may be combined.
  • sense oligomers may have 2 '-modifications on the ends (e.g., 2 on each end, 3 on each end, and 4 on each end, etc.; as well as 1 on one end, 2 on one end, 3 on one end, and 4 on one end, etc.; and even unbalanced combinations such as 12 on one end and 10 on the other end, etc.).
  • the antisense strand may have 2 '-modifications on the ends (1 on each end, 2 on each end, 3 on each end, and 4 on each end, and so on; as well as 1 on one end, 2 on one end, 3 on one end, and 4 on one end, and so on; and even unbalanced combinations such as 1 on one end and 2 on the other end, and so on).
  • the 2 '-modifications are 2'-O-methyl modifications in the sense RNA strand and/or the antisense strand.
  • the sense strand can tolerate many 2' -modifications (such as 2'-O-methyl modifications), so long as the central linkages are unmodified.
  • central is not limited to mean the geometric mid-point of the sense strand. Rather, it can include any location between the 5 '-end portion and the 3 '-end portion of the sense strand. The 5'-end portion and the 3'-end portion of the sense strand need not be symmetrical.
  • the sense strand is not completely modified (i. e. , at least one or more sense strand nucleotide(s) are unmodified).
  • the unmodified sense strand nucleotides are in the central portion of the sense strand, or between the streteh of modified sense strand nucleotides on the 5 '-end and the strerch of modified sense strand nucleotides on the 3 '-end.
  • the sense strand tolerance for 2 '-modification is not necessarily symmetrical. Rather, asymmetrical configurations may be desirable when using, for example, a sense strand of 25 or 26 nucleotides. 2'-mofications add nuclease stability, and reduce interferon induction, and are easier to synthesize. Thus it may be desirable to include more such 2 '-modified ribose sugars (especially 2'-O-methyl modified) on the sense strand, so long as the teachings of the instant invention is followed to preserve RNAi activity.
  • the subject highly modified sense strands may be combined with either unmodified or lightly modified antisense strands to allow maximum guide strand activity.
  • inter-nucleomonomer linkages other than phosphodiesters may be used.
  • end blocks may be used alone or in conjunction with phosphothioate linkages between the 2'-O-methly linkages.
  • Preferred T- modified nucleomonomers are 2'-modified end nucleotides.
  • the antisense strand may be substantially identical to at least a portion of the target MAP4K4 gene (or genes), at least with respect to the base pairing properties, the sequence need not be perfectly identical to be useful. Generally, higher homology can be used to compensate for the use of a shorter antisense gene. In some cases, the antisense strand generally will be substantially identical (although in antisense orientation) to the MAP4K4 target gene.
  • composition of the invention has end-blocks on both ends of a sense oligonucleotide and only the 3 '-end of an antisense oligonucleotide.
  • a 2'-O-modif ⁇ ed sense strand may work less well than its unmodified version, possibly because it is not efficiently unwound.
  • mismatches may be introduced into specific positions of the sense strand (modified 2'-O-methyl sense strand, or even unmodified sense strand) to facilitate easier loading into the RISC complex.
  • the length of the sense strand can be 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, or 18 nucleotides.
  • the length of the antisense strand can be 30, 29, 28, 27, 26, 25, 24, 23, 22, 21, 20, 19, or 18 nucleotides.
  • the resulting duplex may have blunt ends or overhangs of 0, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 nucleotides on one end or independently on each end.
  • double stranded nucleic acid molecules of the invention may be composed of a sense strand and an antisense strand wherein these strands are of lengths described above, and are of the same or different lengths, but share only 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides of sequence complementarity.
  • a double stranded nucleic acid molecules may be formed with a 10 nucleotide overhang on one end and a 5 nucleotide overhang on the other end.
  • RNA having 2'-O-methyl nucleomonomers may not be recognized by cellular machinery that is thought to recognize unmodified RNA.
  • the use of 2'-O-methylated or partially 2'-O-methylated RNA may avoid the interferon response to double-stranded nucleic acids, while maintaining target RNA inhibition. This may be useful, for example, for avoiding the interferon or other cellular stress responses, both in short RNAi (e.g., siRNA) sequences that induce the interferon response, and in longer RNAi sequences that may induce the interferon response.
  • modified sugars may include D-ribose, 2'-O-alkyl (including 2'-O-metbyl and
  • the sugar moiety can be a hexose and incorporated into an oligonucleotide as described (Augustyns, K., et al., Nucl. Acids. Res. 18:4711 (1992)).
  • Exemplary nucleomonomers can be found, e.g., in U.S. Pat. No. 5,849,902, incorporated by reference herein.
  • alkyl includes saturated aliphatic groups, including straight-chain alkyl groups (e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, etc.), branched-chain alkyl groups (isopropyl, tert-butyl, isobutyl, etc.), cycloalkyl (alicyclic) groups (cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl), alkyl substituted cycloalkyl groups, and cycloalkyl substituted alkyl groups.
  • straight-chain alkyl groups e.g., methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl,
  • a straight chain or branched chain alkyl has 6 or fewer carbon atoms in its backbone (e.g., Ci-Ce for straight chain, C 3 -C 6 for branched chain), and more preferably 4 or fewer.
  • preferred cycloalkyls have from 3-8 carbon atoms in their ring structure, and more preferably have 5 or 6 carbons in the ring structure.
  • CpC 6 includes alkyl groups containing 1 to 6 carbon atoms.
  • alkyl includes both "unsubstituted alkyls" and “substituted alkyls,” the latter of which refers to alkyl moieties having independently selected substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone.
  • substituents can include, for example, alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate, sul
  • Cycloalkyls can be further substituted, e.g., with the substituents described above.
  • An "alkylaryl” or an “arylalkyl” moiety is an alkyl substituted with an aryl (e.g., phenylmethyl (benzyl)).
  • the term “alkyl” also includes the side chains of natural and unnatural amino acids.
  • n-alkyl means a straight chain (i.e., unbranched) unsubstituted alkyl group.
  • alkenyl includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double bond.
  • alkenyl includes straight-chain alkenyl groups (e.g., ethylenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, decenyl, etc.), branched-chain alkenyl groups, cycloalkenyl (alicyclic) groups (cyclopropenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, cyclooctenyl), alkyl or alkenyl substituted cycloalkenyl groups, and cycloalkyl or cycloalkenyl substituted alkenyl groups.
  • a straight chain or branched chain alkenyl group has 6 or fewer carbon atoms in its backbone (e.g. , C 2 -C 6 for straight chain, C 3 -C 6 for branched chain).
  • cycloalkenyl groups may have from 3-8 carbon atoms in their ring structure, and more preferably have 5 or 6 carbons in the ring structure.
  • the term C 2 - C 6 includes alkenyl groups containing 2 to 6 carbon atoms.
  • alkenyl includes both "unsubstituted alkenyls" and “substituted alkenyls,” the latter of which refers to alkenyl moieties having independently selected substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone.
  • substituents can include, for example, alkyl groups, alkynyl groups, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate,
  • alkynyl includes unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but which contain at least one triple bond.
  • alkynyl includes straight-chain alkynyl groups (e.g., ethynyl, propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, decynyl, etc.), branched-chain alkynyl groups, and cycloalkyl or cycloalkenyl substituted alkynyl groups.
  • a straight chain or branched chain alkynyl group has 6 or fewer carbon atoms in its backbone (e.g., C 2 -Ce for straight chain, C 3 -C 6 for branched chain).
  • C 2 -C ⁇ includes alkynyl groups containing 2 to 6 carbon atoms.
  • alkynyl includes both "unsubstituted alkynyls" and “substituted alkynyls,” the latter of which refers to alkynyl moieties having independently selected substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone.
  • substituents can include, for example, alkyl groups, alkynyl groups, halogens, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulfhydryl, alkylthio, arylthio, thiocarboxylate,
  • lower alkyl as used herein means an alkyl group, as defined above, but having from one to five carbon atoms in its backbone structure.
  • Lower alkenyl and “lower alkynyl” have chain lengths of, for example, 2-5 carbon atoms.
  • alkoxy includes substituted and unsubstituted alkyl, alkenyl, and alkynyl groups covalently linked to an oxygen atom.
  • alkoxy groups include methoxy, ethoxy, isopropyloxy, propoxy, butoxy, and pentoxy groups.
  • substituted alkoxy groups include halogenated alkoxy groups.
  • the alkoxy groups can be substituted with independently selected groups such as alkenyl, alkynyl, halogen, hydroxyl, alkylcarbonyloxy, arylcarbonyloxy, alkoxycarbonyloxy, aryloxycarbonyloxy, carboxylate, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, alkylthiocarbonyl, alkoxyl, phosphate, phosphonato, phosphinato, cyano, amino (including alkyl amino, dialkylamino, arylamino, diarylamino, and alkylarylamino), acylamino (including alkylcarbonylamino, arylcarbonylamino, carbamoyl and ureido), amidino, imino, sulffiydryl, alkylthio, arylthio, thio
  • heteroatom includes atoms of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, sulfur and phosphorus.
  • hydroxy or "hydroxyl” includes groups with an -OH or -O " (with an appropriate counterion).
  • halogen includes fluorine, bromine, chlorine, iodine, etc.
  • perhalogenated generally refers to a moiety wherein all hydrogens are replaced by halogen atoms.
  • substituted includes independently selected substituents which can be placed on the moiety and which allow the molecule to perform its intended function.
  • substituents include alkyl, alkenyl, alkynyl, aryl, (CR'R")o- 3 NR'R", (CR'R")o- 3 CN, NO 2 , halogen, (CR'R")o-3C(halogen) 3; (CR'R")o- 3 CH(halogen) 2 , (CR'R")o- 3 CH 2 (halogen), (CR'R")o- 3 CONR'R", (CR'R")o.
  • each R' and R" are each independently hydrogen, a C 1 -C 5 alkyl, C 2 -Cs alkenyl, C 2 -C 5 alkynyl, or aryl group, or R' and R" taken together are a benzylidene group or a — (CH 2 ) 2 O(CH 2 ) 2 - group.
  • amine or “amino” includes compounds or moieties in which a nitrogen atom is covalently bonded to at least one carbon or heteroatom.
  • alkyl amino includes groups and compounds wherein the nitrogen is bound to at least one additional alkyl group.
  • dialkyl amino includes groups wherein the nitrogen atom is bound to at least two additional alkyl groups.
  • ether includes compounds or moieties which contain an oxygen bonded to two different carbon atoms or heteroatoms.
  • alkoxyalkyl refers to an alkyl, alkenyl, or alkynyl group covalently bonded to an oxygen atom which is covalently bonded to another alkyl group.
  • base includes the known purine and pyrimidine heterocyclic bases, deazapurines, and analogs (including heterocyclic substituted analogs, e.g., aminoethyoxy phenoxazine), derivatives (e.g., 1 -alkyl-, 1-alkenyl-, heteroaromatic- and 1-alkynyl derivatives) and tautomers thereof.
  • purines include adenine, guanine, inosine, diaminopurine, and xanthine and analogs (e.g. , 8-oxo-N 6 -methyladenine or 7-diazaxanthine) and derivatives thereof.
  • Pyrimidines include, for example, thymine, uracil, and cytosine, and their analogs (e.g., 5-methylcytosine, 5-methyluracil, 5-(l-propynyl)uracil, 5-(l-propynyl)cytosine and 4,4- ethanocytosine).
  • suitable bases include non-purinyl and non-pyrimidinyl bases such as 2-aminopyridine and triazines.
  • nucleomonomers of an oligonucleotide of the invention are RNA nucleotides.
  • the nucleomonomers of an oligonucleotide of the invention are modified RNA nucleotides.
  • the oligonucleotides contain modified RNA nucleotides.
  • the term "nucleoside” includes bases which are covalently attached to a sugar moiety, preferably ribose or deoxyribose. Examples of preferred nucleosides include ribonucleosides and deoxyribonucleosides. Nucleosides also include bases linked to amino acids or amino acid analogs which may comprise free carboxyl groups, free amino groups, or protecting groups. Suitable protecting groups are well known in the art (see P. G. M. Wuts and T. W. Greene, "Protective Groups in Organic Synthesis", 2 M Ed., Wiley-Interscience, New York, 1999).
  • nucleotide includes nucleosides which further comprise a phosphate group or a phosphate analog.
  • linkage includes a naturally occurring, unmodified phosphodiester moiety (-O-(PO 2 ⁇ )-O-) that covalently couples adjacent nucleomonomers.
  • substitute linkage includes any analog or derivative of the native phosphodiester group that covalently couples adjacent nucleomonomers. Substitute linkages include phosphodiester analogs, e.g., phosphorothioate, phosphorodithioate, and P- ethyoxyphosphodiester, P-ethoxyphosphodiester, P-alkyloxyphosphotriester, methylphosphonate, and nonphosphorus containing linkages, e.g., acetals and amides.
  • linkages are known in the art (e.g., Bjergarde et al. 1991. Nucleic Acids Res. 19:5843; Caruthers et al. 1991. Nucleosides Nucleotides. 10:47).
  • non- hydrolizable linkages are preferred, such as phosphorothioate linkages.
  • oligonucleotides of the invention comprise 3' and 5' termini (except for circular oligonucleotides).
  • the 3' and 5' termini of an oligonucleotide can be substantially protected from nucleases e.g., by modifying the 3 ' or 5' linkages (e.g., U.S. Pat. No. 5,849,902 and WO 98/13526).
  • oligonucleotides can be made resistant by the inclusion of a "blocking group.”
  • blocking group refers to substituents (e.g., other than OH groups) that can be attached to oligonucleotides or nucleomonomers, either as protecting groups or coupling groups for synthesis (e.g., FITC, propyl (CH 2 -CH 2 -CH 3 ), glycol (-0-CH 2 -CH 2 -O-) phosphate (PO 3 2" ), hydrogen phosphonate, or phosphoramidite).
  • Blocking groups also include “end blocking groups” or “exonuclease blocking groups” which protect the 5' and 3' termini of the oligonucleotide, including modified nucleotides and non-nucleotide exonuclease resistant structures.
  • Exemplary end-blocking groups include cap structures (e.g., a 7-methylguanosine cap), inverted nucleomonomers, e.g., with 3'-3' or 5'-5' end inversions (see, e.g., Ortiagao et al. 1992. Antisense Res. Dev.
  • the 3' terminal nucleomonomer can comprise a modified sugar moiety.
  • the 3' terminal nucleomonomer comprises a 3'-0 that can optionally be substituted by a blocking group that prevents 3 '-exonuclease degradation of the oligonucleotide.
  • the 3'-hydroxyl can be esterified to a nucleotide through a 3' ⁇ 3' internucleotide linkage.
  • the alkyloxy radical can be methoxy, ethoxy, or isopropoxy, and preferably, ethoxy.
  • the 3'— +3 'linked nucleotide at the 3' terminus can be linked by a substitute linkage.
  • the 5' most 3 ' ⁇ 5' linkage can be a modified linkage, e.g., a phosphorothioate or a P-alkyloxyphosphotriester linkage.
  • the two 5' most 3' ⁇ 5' linkages are modified linkages.
  • the 5' terminal hydroxy moiety can be esterified with a phosphorus containing moiety, e.g., phosphate, phosphorothioate, or P-ethoxyphosphate.
  • the sense strand of an oligonucleotide comprises a 5' group that allows for RNAi activity but which renders the sense strand inactive in terms of gene targeting.
  • a 5' modifying group is a phosphate group or a group larger than a phosphate group.
  • Oligonucleotides of this type often exhibit increased specificity for a target gene in a cell that corresponds to the nucleotide sequence of the antisense strand. This is because the sense strand in such an oligonucleotide is often rendered incapable of mediating cleavage of any nucleotide sequence it might bind to non-specifically and thus will not inactivate any other genes in the cell.
  • the invention provides a method of increasing the specificity of an oligonucleotide for a target MAP4K4 gene in a cell, wherein said oligonucleotide comprises a sense strand and an antisense strand, wherein both the sense strand and the antisense strand are capable of binding to corresponding nucleotide sequences if present in said cell, said method comprising the step of modifying the 5' terminal hydroxy moiety of said sense strand with a phosphate group or a group larger than a phosphate group prior to contacting said oligonucleotide with said cell so as to render said sense strand incapable of mediating cleavage of any nucleotide sequence it might bind to non-specifically and thus will not inactivate any other
  • Another way to increase target gene specificity, or to reduce off-target silencing effect is to introduce a 2 '-modification (such as the 2'-0 methyl modification) at a position corresponding to the second 5 '-end nucleotide of the Dicer-cleaved 21-mer.
  • a 2 '-modification such as the 2'-0 methyl modification
  • Applicants' discovery allows the positioning of this 2' -modification in the Dicer-resistant dsRNA, thus enabling one to design better siRNA constructs with less or no off-target silencing.
  • a double-stranded oligonucleotide of the invention can comprise (i.e., be a duplex of) one nucleic acid molecule which is DNA and one nucleic acid molecule which is RNA.
  • Antisense sequences of the invention can be "chimeric oligonucleotides" which comprise an RNA-like and a DNA-like region.
  • RNase H activating region includes a region of an oligonucleotide, e.g., a chimeric oligonucleotide, that is capable of recruiting RNase H to cleave the target RNA strand to which the oligonucleotide binds.
  • the RNase activating region contains a minimal core (of at least about 3-5, typically between about 3-12, more typically, between about 5-12, and more preferably between about 5-10 contiguous nucleomonomers) of DNA or DNA- like nucleomonomers. (See, e.g., U.S. Pat. No. 5,849,902).
  • the RNase H activating region comprises about nine contiguous deoxyribose containing nucleomonomers.
  • non-activating region includes a region of an antisense sequence, e.g., a chimeric oligonucleotide, that does not recruit or activate RNase H.
  • a non-activating region does not comprise phosphorothioate DNA.
  • the oligonucleotides of the invention comprise at least one non-activating region.
  • the non-activating region can be stabilized against nucleases or can provide specificity for the target by being complementary to the target and forming hydrogen bonds with the target nucleic acid molecule, which is to be bound by the oligonucleotide.
  • At least a portion of the contiguous polynucleotides are linked by a substitute linkage, e.g., a phosphorothioate linkage.
  • most or all of the sense strand nucleotides (2 '-modified or not) are linked by phosphorothioate linkages.
  • Such constructs tend to have improved pharmacokinetics due to their higher affinity for serum proteins.
  • the phosphorothioate linkages in the sense strand generally do not interfere with guide strand activity, once the latter is loaded into RISC.
  • Antisense sequences of the present invention may include "morpholino oligonucleotides.” Morpholino oligonucleotides are non-ionic and function by an RNase H- independent mechanism. Each of the 4 genetic bases (Adenine, Cytosine, Guanine, and Thymine/Uracil) of the morpholino oligonucleotides is linked to a 6-membered morpholine ring. Morpholino oligonucleotides are made by joining the 4 different subunit types by, e.g., non- ionic phosphorodiamidate inter-subunit linkages.
  • Morpholino oligonucleotides have many advantages including: complete resistance to nucleases (Antisense & Nucl. Acid Drug Dev. 1996. 6:267); predictable targeting (Biochemica Biophysica Acta. 1999. 1489:141); reliable activity in cells (Antisense & Nucl. Acid Drug Dev. 1997. 7:63); excellent sequence specificity (Antisense & Nucl. Acid Drug Dev. 1997. 7:151); minimal non-antisense activity (Biochemica Biophysica Acta. 1999. 1489:141); and simple osmotic or scrape delivery (Antisense & Nucl. Acid Drug Dev. 1997. 7:291).
  • Morpholino oligonucleotides are also preferred because of their non-toxicity at high doses. A discussion of the preparation of morpholino oligonucleotides can be found in Antisense & Nucl. Acid Drug Dev. 1997. 7:187.
  • Oligonucleotides of the invention can be synthesized by any method known in the art, e.g., using enzymatic synthesis and/or chemical synthesis.
  • the oligonucleotides can be synthesized in vitro (e.g., using enzymatic synthesis and chemical synthesis) or in vivo (using recombinant DNA technology well known in the art).
  • chemical synthesis is used for modified polynucleotides.
  • Oligonucleotides can be made by any of several different synthetic procedures including the phosphoramidite, phosphite triester, H-phosphonate, and phosphotriester methods, typically by automated synthesis methods.
  • Oligonucleotide synthesis protocols are well known in the art and can be found, e.g. , in U.S. Pat. No. 5,830,653; WO 98/13526; Stec et al. 1984. J. Am. Chem. Soc. 106:6077; Stec et al. 1985. J Org. Chem. 50:3908; Stec et al. J. Chromatog. 1985. 326:263; LaPlanche et al.
  • the synthesis method selected can depend on the length of the desired oligonucleotide and such choice is within the skill of the ordinary artisan.
  • the phosphoramidite and phosphite triester method can produce oligonucleotides having 175 or more nucleotides, while the H-phosphonate method works well for oligonucleotides of less than 100 nucleotides. If modified bases are incorporated into the oligonucleotide, and particularly if modified phosphodiester linkages are used, then the synthetic procedures are altered as needed according to known procedures. In this regard, Uhlmann et al.
  • linear oligonucleotides of defined sequence including some sequences with modified nucleotides, are readily available from several commercial sources.
  • oligonucleotides may be purified by polyacrylamide gel electrophoresis, or by any of a number of chromatographic methods, including gel chromatography and high pressure liquid chromatography.
  • oligonucleotides may be subjected to DNA sequencing by any of the known procedures, including Maxam and Gilbert sequencing, Sanger sequencing, capillary electrophoresis sequencing, the wandering spot sequencing procedure or by using selective chemical degradation of oligonucleotides bound to Hybond paper.
  • Sequences of short oligonucleotides can also be analyzed by laser desorption mass spectroscopy or by fast atom bombardment (McNeal, et al., 1982, J. Am. Chem. Soc. 104:976; Viari, et al, 1987, Biomed. Environ. Mass Spectrom. 14:83; Grotjahn et al., 1982, Nuc. Acid Res. 10:4671). Sequencing methods are also available for RNA oligonucleotides.
  • oligonucleotides synthesized can be verified by testing the oligonucleotide by capillary electrophoresis and denaturing strong anion HPLC (SAX-HPLC) using, e.g., the method of Bergot and Egan. 1992. J. Chrom. 599:35.
  • SAX-HPLC denaturing strong anion HPLC
  • the subject RNAi constructs or at least portions thereof are transcribed from expression vectors encoding the subject constructs. Any art recognized vectors may be use for this purpose.
  • the transcribed RNAi constructs may be isolated and purified, before desired modifications (such as replacing an unmodified sense strand with a modified one, etc.) are carried out.
  • Oligonucleotides and oligonucleotide compositions are contacted with (i.e., brought into contact with, also referred to herein as administered or delivered to) and taken up by one or more cells or a cell lysate.
  • the term "cells" includes prokaryotic and eukaryotic cells, preferably vertebrate cells, and, more preferably, mammalian cells.
  • the oligonucleotide compositions of the invention are contacted with human cells.
  • Oligonucleotide compositions of the invention can be contacted with cells in vitro, e.g., in a test tube or culture dish, (and may or may not be introduced into a subject) or in vivo, e.g., in a subject such as a mammalian subject. Oligonucleotides are taken up by cells at a slow rate by endocytosis, but endocytosed oligonucleotides are generally sequestered and not available, e.g., for hybridization to a target nucleic acid molecule. In one embodiment, cellular uptake can be facilitated by electroporation or calcium phosphate precipitation. However, these procedures are only useful for in vitro or ex vivo embodiments, are not convenient and, in some cases, are associated with cell toxicity.
  • delivery of oligonucleotides into cells can be enhanced by suitable art recognized methods including calcium phosphate, DMSO, glycerol or dextran, electroporation, or by transfection, e.g. , using cationic, anionic, or neutral lipid compositions or liposomes using methods known in the art (see e.g., WO 90/14074; WO 91/16024; WO 91/17424; U.S. Pat. No. 4,897,355; Bergan et al. 1993. Nucleic Acids Research. 21 :3567).
  • Enhanced delivery of oligonucleotides can also be mediated by the use of vectors (See e.g., Shi, Y. 2003.
  • oligonucleotides The optimal protocol for uptake of oligonucleotides will depend upon a number of factors, the most crucial being the type of cells that are being used. Other factors that are important in uptake include, but are not limited to, the nature and concentration of the oligonucleotide, the confluence of the cells, the type of culture the cells are in (e.g., a suspension culture or plated) and the type of media in which the cells are grown.
  • Conjugating agents bind to the oligonucleotide in a covalent manner.
  • oligonucleotides can be derivatized or chemically modified by binding to a conjugating agent to facilitate cellular uptake.
  • covalent linkage of a cholesterol moiety to an oligonucleotide can improve cellular uptake by 5- to 10-fold which in turn improves DNA binding by about 10-fold (Boutorin et ah, 1989, FEBS Letters 254:129-132).
  • Certain protein carriers can also facilitate cellular uptake of oligonucleotides, including, for example, serum albumin, nuclear proteins possessing signals for transport to the nucleus, and viral or bacterial proteins capable of cell membrane penetration. Therefore, protein carriers are useful when associated with or linked to the oligonucleotides. Accordingly, the present invention provides for derivatization of oligonucleotides with groups capable of facilitating cellular uptake, including hydrocarbons and non-polar groups, cholesterol, long chain alcohols (i. e.
  • conjugating agents are to increase the initial membrane interaction that leads to a greater cellular accumulation of oligonucleotides.
  • conjugating agents include various vitamins, such as fat soluble vitamins, which may be used as a conjugate to deliver RNAi constructs specifically into adipose tissue - the primary location where these vitamins are stored. These vitamin-based conjugating agents may be especially useful for targeting certain metabolic disease targets, such as diabetes / obesity.
  • fat soluble vitamins such as vitamins A, D, E, K, etc.
  • vitamin K may be preferred in some embodiments, as there is no known upper intake level (although large doses could lead to breakdown of red blood cells and possibly to liver disease).
  • vitamins A and D have more defined toxicity and established upper intake levels.
  • gamma carboxyglutamic acid residues may be conjugated to the subject RNAi constructs to increased their membrane stickiness, and/or to slow clearance and improve general uptake (infra).
  • conjugating agents that may be used with the instant constructs include those described in WO04048545A2 and US20040204377A1 (all incorporated herein by their entireties), such as a Tat peptide, a sequence substantially similar to the sequence of sequence ID number 12 of WO04048545A2 and US20040204377A1, a homeobox (hox) peptide, a MTS, VP22, MPG, at least one dendrimer (such as PAMAM), etc.
  • Tat peptide a sequence substantially similar to the sequence of sequence ID number 12 of WO04048545A2 and US20040204377A1
  • a homeobox (hox) peptide a MTS, VP22, MPG, at least one dendrimer (such as PAMAM), etc.
  • conjugating agents that may be used with the instant constructs include those described in WO07089607A2 (incorporated herein), which describes various nanotransporters and delivery complexes for use in delivery of nucleic acid molecules (such as the subject dsRNA constructs) and/or other pharmaceutical agents in vivo and in vitro.
  • the subject dsRNA can be delivered while conjugated or associated with a nanotransporter comprising a core conjugated with at least one functional surface group.
  • the core may be a nanoparticle, such as a dendrimer (e.g. , a polylysine dendrimer).
  • the core may also be a nanotube, such as a single walled nanotube or a multi-walled nanotube.
  • the functional surface group is at least one of a lipid, a cell type specific targeting moiety, a fluorescent molecule, and a charge controlling molecule.
  • the targeting moiety may be a tissue- selective peptide.
  • the lipid may be an oleoyl lipid or derivative thereof.
  • Exemplary nanotransporter include NOP-7 or HBOLD.
  • Encapsulating agents entrap oligonucleotides within vesicles.
  • an oligonucleotide may be associated with a carrier or vehicle, e.g., liposomes or micelles, although other carriers could be used, as would be appreciated by one skilled in the art.
  • Liposomes are vesicles made of a lipid bilayer having a structure similar to biological membranes. Such carriers are used to facilitate the cellular uptake or targeting of the oligonucleotide, or improve the oligonucleotide's pharmacokinetic or toxicologic properties.
  • the oligonucleotides of the present invention may also be administered encapsulated in liposomes, pharmaceutical compositions wherein the active ingredient is contained either dispersed or variously present in corpuscles consisting of aqueous concentric layers adherent to lipidic layers.
  • the oligonucleotides depending upon solubility, may be present both in the aqueous layer and in the lipidic layer, or in what is generally termed a liposomic suspension.
  • the hydrophobic layer generally but not exclusively, comprises phopholipids such as lecithin and sphingomyelin, steroids such as cholesterol, more or less ionic surfactants such as diacetylphosphate, stearylamine, or phosphatidic acid, or other materials of a hydrophobic nature.
  • phopholipids such as lecithin and sphingomyelin
  • steroids such as cholesterol
  • ionic surfactants such as diacetylphosphate, stearylamine, or phosphatidic acid
  • the diameters of the liposomes generally range from about 15 nm to about 5 microns.
  • Liposomes increase intracellular stability, increase uptake efficiency and improve biological activity.
  • Liposomes are hollow spherical vesicles composed of lipids arranged in a similar fashion as those lipids which make up the cell membrane. They have an internal aqueous space for entrapping water soluble compounds and range in size from 0.05 to several microns in diameter.
  • lipid delivery vehicle originally designed as a research tool, such as Lipofectin or LIPOFECTAMINETM 2000, can deliver intact nucleic acid molecules to cells.
  • liposomes are non-toxic and biodegradable in composition; they display long circulation half-lives; and recognition molecules can be readily attached to their surface for targeting to tissues.
  • cost-effective manufacture of liposome-based pharmaceuticals either in a liquid suspension or lyophilized product, has demonstrated the viability of this technology as an acceptable drug delivery system.
  • Methods and compositions for delivering nucleic acid molecules are also incorporated by reference from US Provisional Applications 61/192,954, filed on September 22, 2008, 61/149,946, filed on February 4, 2009 and 61/224,031, filed on July 8, 2009.
  • oligonucleotides of the invention can be complexed with a complexing agent to increase cellular uptake of oligonucleotides.
  • a complexing agent includes cationic lipids. Cationic lipids can be used to deliver oligonucleotides to cells.
  • cationic lipid includes lipids and synthetic lipids having both polar and non- polar domains and which are capable of being positively charged at or around physiological pH and which bind to polyanions, such as nucleic acids, and facilitate the delivery of nucleic acids into cells.
  • cationic lipids include saturated and unsaturated alkyl and alicyclic ethers and esters of amines, amides, or derivatives thereof.
  • Straight-chain and branched alkyl and alkenyl groups of cationic lipids can contain, e.g. , from 1 to about 25 carbon atoms.
  • Preferred straight chain or branched alkyl or alkene groups have six or more carbon atoms.
  • Alicyclic groups include cholesterol and other steroid groups.
  • Cationic lipids can be prepared with a variety of counterions (anions) including, e.g., CV, Br, I', F " , acetate, trifluoroacetate, sulfate, nitrite, and nitrate.
  • counterions e.g., CV, Br, I', F " , acetate, trifluoroacetate, sulfate, nitrite, and nitrate.
  • cationic lipids examples include polyethylenimine, polyamidoamine (PAMAM) starburst dendrimers, Lipofectin (a combination of DOTMA and DOPE), Lipofectase, LIPOFECTAMINETM (e.g. , LIPOFECTAMINETM 2000), DOPE, Cytofectin (Gilead Sciences, Foster City, Calif.), and Eufectins (JBL, San Luis Obispo, Calif.).
  • Exemplary cationic liposomes can be made from N-[I -(2,3 -dioleoloxy)-propyl] -N 5 N ,N-trimethylammonium chloride (DOTMA), N-[I -(2,3-dioleoloxy)-propyl]-N,N,N-trimethylammonium methylsulfate (DOTAP), 3 ⁇ -[N-(N',N'-dimethylaminoethane)carbamoyl]cholesterol (DC-Choi), 2,3,- dioleyloxy-N-[2(sperminecarboxamido)ethyl]-N,N-dimethyl-l-propanaminium trifluoroacetate (DOSPA), l ,2-dimyristyloxypropyl-3-dimethyl-hydroxyethyl ammonium bromide; and dimethyldioctadecylammoniurn bromide (DDAB).
  • DOTMA cationic lipid N-(I -(2,3- dioleyloxy)propyl)-N,N,N-trimethylammonium chloride
  • Cationic lipids have been used in the art to deliver oligonucleotides to cells (see, e.g., U.S. Pat. Nos. 5,855,910; 5,851,548; 5,830,430; 5,780,053; 5,767,099; Lewis et al. 1996. Proc. Na//. Acad. Sci. USA 93:3176; Hope et al. 1998. Molecular Membrane Biology 15:1).
  • Other lipid compositions which can be used to facilitate uptake of the instant oligonucleotides can be used in connection with the claimed methods.
  • other lipid compositions are also known in the art and include, e.g. , those taught in U.S. Pat. No.
  • lipid compositions can further comprise agents, e.g., viral proteins to enhance lipid-mediated transfections of oligonucleotides (Kamata, et al., 1994. Nucl. Acids. Res. 22:536).
  • oligonucleotides are contacted with cells as part of a composition comprising an oligonucleotide, a peptide, and a lipid as taught, e.g., in U.S. patent 5,736,392.
  • Improved lipids have also been described which are serum resistant (Lewis, et al., 1996. Proc. Natl. Acad. Sci. 93:3176).
  • Cationic lipids and other complexing agents act to increase the number of oligonucleotides carried into the cell through endocytosis.
  • N-substituted glycine oligonucleotides can be used to optimize uptake of oligonucleotides.
  • Peptoids have been used to create cationic lipid-like compounds for transfection (Murphy, et al., 1998. Proc. Natl. Acad Sci. 95:1517).
  • Peptoids can be synthesized using standard methods (e.g., Zuckermann, R. N., et al. 1992. J. Am. Chem. Soc. 114:10646; Zuckermann, R. N., et al. 1992. Int. J. Peptide Protein Res. 40:497).
  • Combinations of cationic lipids and peptoids, liptoids can also be used to optimize uptake of the subject oligonucleotides (Hunag, et al., 1998. Chemistry and Biology. 5:345).
  • Liptoids can be synthesized by elaborating peptoid oligonucleotides and coupling the amino terminal submonomer to a lipid via its amino group (Hunag, et al., 1998. Chemistry and Biology. 5:345). It is known in the art that positively charged amino acids can be used for creating highly active cationic lipids (Lewis et al. 1996. Proc. Natl. Acad. Sci. US. A. 93:3176).
  • a composition for delivering oligonucleotides of the invention comprises a number of arginine, lysine, histidine or ornithine residues linked to a lipophilic moiety (see e.g. , U.S. Pat. No. 5,777,153).
  • a composition for delivering oligonucleotides of the invention comprises a peptide having from between about one to about four basic residues. These basic residues can be located, e.g., on the amino terminal, C-terminal, or internal region of the peptide. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g.
  • aspartic acid, glutamic acid uncharged polar side chains (e.g., glycine (can also be considered non-polar), asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g. , alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g. , tyrosine, phenylalanine, tryptophan, histidine).
  • polar side chains e.g., glycine (can also be considered non-polar
  • asparagine, glutamine, serine, threonine, tyrosine, cysteine nonpolar side chains
  • nonpolar side chains e.g. , alanine, valine, leucine, iso
  • a majority or all of the other residues of the peptide can be selected from the non-basic amino acids, e.g., amino acids other than lysine, arginine, or histidine. Preferably a preponderance of neutral amino acids with long neutral side chains are used.
  • a composition for delivering oligonucleotides of the invention comprises a natural or synthetic polypeptide having one or more gamma carboxyglutamic acid residues, or ⁇ -Gla residues. These gamma carboxyglutamic acid residues may enable the polypeptide to bind to each other and to membrane surfaces.
  • a polypeptide having a series of ⁇ -Gla may be used as a general delivery modality that helps an RNAi construct to stick to whatever membrane to which it comes in contact. This may at least slow RNAi constructs from being cleared from the blood stream and enhance their chance of homing to the target.
  • the gamma carboxyglutamic acid residues may exist in natural proteins (for example, prothrombin has 10 ⁇ -Gla residues).
  • they can be introduced into the purified, recombinantly produced, or chemically synthesized polypeptides by carboxylation using, for example, a vitamin K-dependent carboxylase.
  • the gamma carboxyglutamic acid residues may be consecutive or non-consecutive, and the total number and location of such gamma carboxyglutamic acid residues in the polypeptide can be regulated / fine tuned to achieve different levels of "stickiness'Of the polypeptide.
  • the cells to be contacted with an oligonucleotide composition of the invention are contacted with a mixture comprising the oligonucleotide and a mixture comprising a lipid, e.g., one of the lipids or lipid compositions described supra for between about 12 hours to about 24 hours.
  • the cells to be contacted with an oligonucleotide composition are contacted with a mixture comprising the oligonucleotide and a mixture comprising a lipid, e.g., one of the lipids or lipid compositions described supra for between about 1 and about five days.
  • the cells are contacted with a mixture comprising a lipid and the oligonucleotide for between about three days to as long as about 30 days.
  • a mixture comprising a lipid is left in contact with the cells for at least about five to about 20 days.
  • a mixture comprising a lipid is left in contact with the cells for at least about seven to about 15 days.
  • an oligonucleotide composition can be contacted with cells in the presence of a lipid such as cytofectin CS or GSV (available from Glen Research; Sterling, Va.), GS3815, GS2888 for prolonged incubation periods as described herein.
  • a lipid such as cytofectin CS or GSV (available from Glen Research; Sterling, Va.), GS3815, GS2888 for prolonged incubation periods as described herein.
  • the incubation of the cells with the mixture comprising a lipid and an oligonucleotide composition does not reduce the viability of the cells.
  • the cells are substantially viable.
  • the cells are between at least about 70% and at least about 100% viable.
  • the cells are between at least about 80% and at least about 95% viable.
  • the cells are between at least about 85% and at least about 90% viable.
  • oligonucleotides are modified by attaching a peptide sequence that transports the oligonucleotide into a cell, referred to herein as a "transporting peptide.”
  • the composition includes an oligonucleotide which is complementary to a target nucleic acid molecule encoding the protein, and a covalently attached transporting peptide.
  • transporting peptide includes an amino acid sequence that facilitates the transport of an oligonucleotide into a cell.
  • Exemplary peptides which facilitate the transport of the moieties to which they are linked into cells are known in the art, and include, e.g., HIV TAT transcription factor, lactoferrin, Herpes VP22 protein, and fibroblast growth factor 2 (Pooga et al. 1998. Nature Biotechnology. 16:857; and Derossi et al. 1998. Trends in Cell Biology. 8:84; Elliott and O ⁇ are. 1997. Cell 88:223).
  • Oligonucleotides can be attached to the transporting peptide using known techniques, e.g., ( Prochiantz, A. 1996. Curr. Opirt. Neurobiol. 6:629; Derossi et al. 1998. Trends Cell Biol. 8:84; Troy et al. 1996. J. Neurosci. 16:253), Vives et al. 1997. J. Biol. Chem. 272: 16010).
  • oligonucleotides bearing an activated thiol group are linked via that thiol group to a cysteine present in a transport peptide (e.g., to the cysteine present in the ⁇ turn between the second and the third helix of the antennapedia homeodomain as taught, e.g. , in Derossi et al. 1998. Trends Cell Biol. 8:84; Prochiantz. 1996. Current Opinion in Neurobiol 6:629; Allinquant et al. 1995. J Cell Biol. 128:919).
  • a transport peptide e.g., to the cysteine present in the ⁇ turn between the second and the third helix of the antennapedia homeodomain as taught, e.g. , in Derossi et al. 1998. Trends Cell Biol. 8:84; Prochiantz. 1996. Current Opinion in Neurobiol 6:629; Allinquant et al. 1995. J Cell Biol. 128
  • a Boc-Cys- (Npys)OH group can be coupled to the transport peptide as the last (N-terminal) amino acid and an oligonucleotide bearing an SH group can be coupled to the peptide (Troy et al. 1996. J. Neurosci. 16:253).
  • a linking group can be attached to a nucleomonomer and the transporting peptide can be covalently attached to the linker.
  • a linker can function as both an attachment site for a transporting peptide and can provide stability against nucleases.
  • linkers examples include substituted or unsubstituted Ci-C 2O alkyl chains, C 2 -C 2O alkenyl chains, C 2 -C 2O alkynyl chains, peptides, and heteroatoms ⁇ e.g., S, O, NH, etc).
  • Other exemplary linkers include bifunctional crosslinking agents such as sulfosuccinimidyl-4- (maleimidophenyl)-butyrate (SMPB) (see, e.g., Smith et al. Biochem J 1991.276: 417-2).
  • oligonucleotides of the invention are synthesized as molecular conjugates which utilize receptor-mediated endocytotic mechanisms for delivering genes into cells (see, e.g., Bunnell et al. 1992. Somatic Cell and Molecular Genetics. 18:559, and the references cited therein).
  • the delivery of oligonucleotides can also be improved by targeting the oligonucleotides to a cellular receptor.
  • the targeting moieties can be conjugated to the oligonucleotides or attached to a carrier group (i.e., poly(L-lysine) or liposomes) linked to the oligonucleotides. This method is well suited to cells that display specific receptor-mediated endocytosis.
  • oligonucleotide conjugates to 6-phosphomannosylated proteins are internalized 20-fold more efficiently by cells expressing mannose 6-phosphate specific receptors than free oligonucleotides.
  • the oligonucleotides may also be coupled to a ligand for a cellular receptor using a biodegradable linker.
  • the delivery construct is mannosylated streptavidin which forms a tight complex with biotinylated oligonucleotides. Mannosylated streptavidin was found to increase 20-fold the internalization of biotinylated oligonucleotides. (Vlassov et al. 1994.
  • ligands can be conjugated to the polylysine component of polylysine- based delivery systems.
  • transferrin-polylysine, adenovirus-polylysine, and influenza virus hemagglutinin HA-2 N-terminal fusogenic peptides-polylysine conjugates greatly enhance receptor-mediated DNA delivery in eucaryotic cells.
  • Mannosylated glycoprotein conjugated to poly(L-lysine) in aveolar macrophages has been employed to enhance the cellular uptake of oligonucleotides. Liang et al. 1999. Pharmazie 54:559-566.
  • oligonucleotides can be used to target oligonucleotides to cancerous cells.
  • folic acid is linked to poly(L-lysine) enhanced oligonucleotide uptake is seen in promyelocytic leukaemia (HL-60) cells and human melanoma (M- 14) cells. Ginobbi et al. 1997. Anticancer Res. 17:29.
  • liposomes coated with maleylated bovine serum albumin, folic acid, or ferric protoporphyrin IX show enhanced cellular uptake of oligonucleotides in murine macrophages, KB cells, and 2.2.15 human hepatoma cells. Liang et al. 1999. Pharmazie 54:559-566.
  • Liposomes naturally accumulate in the liver, spleen, and reticuloendothelial system (so- called, passive targeting). By coupling liposomes to various ligands such as antibodies or protein A, they can be actively targeted to specific cell populations.
  • protein A- bearing liposomes may be pretreated with H-2K specific antibodies which are targeted to the mouse major histocompatibility complex-encoded H-2K protein expressed on L cells. (Vlassov et al. 1994. Biochimica et Biophysica Acta 1197:95-108). V. Administration
  • the optimal course of administration or delivery of the oligonucleotides may vary depending upon the desired result and/or on the subject to be treated.
  • administration refers to contacting cells with oligonucleotides and can be performed in vitro or in vivo.
  • the dosage of oligonucleotides may be adjusted to optimally reduce expression of a protein translated from a target nucleic acid molecule, e.g., as measured by a readout of RNA stability or by a therapeutic response, without undue experimentation.
  • RNA or protein levels in a cell or produced by a cell can be measured using any art recognized technique.
  • the effectiveness of the oligonucleotide in inducing the cleavage of a target RNA can be determined.
  • pharmaceutically acceptable carrier includes appropriate solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, it can be used in the therapeutic compositions. Supplementary active ingredients can also be incorporated into the compositions.
  • Oligonucleotides may be incorporated into liposomes or liposomes modified with polyethylene glycol or admixed with cationic lipids for parenteral administration. Incorporation of additional substances into the liposome, for example, antibodies reactive against membrane proteins found on specific target cells, can help target the oligonucleotides to specific cell types.
  • the present invention provides for administering the subject oligonucleotides with an osmotic pump providing continuous infusion of such oligonucleotides, for example, as described in Rataiczak et al. (1992 Proc. Natl. Acad. ScL USA 89:11823-11827).
  • Such osmotic pumps are commercially available, e.g., from Alzet Inc. (Palo Alto, Calif.). Topical administration and parenteral administration in a cationic lipid carrier are preferred.
  • the formulations of the present invention can be administered to a patient in a variety of forms adapted to the chosen route of administration, e.g., parenterally, orally, or intraperitoneal ⁇ .
  • Parenteral administration which is preferred, includes administration by the following routes: intravenous; intramuscular; interstitially; intraarterially; subcutaneous; intra ocular; intrasynovial; trans epithelial, including transdermal; pulmonary via inhalation; ophthalmic; sublingual and buccal; topically, including ophthalmic; dermal; ocular; rectal; and nasal inhalation via insufflation.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble or water-dispersible form.
  • suspensions of the active compounds as appropriate oily injection suspensions may be administered.
  • Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension include, for example, sodium carboxymethyl cellulose, sorbitol, or dextran, optionally, the suspension may also contain stabilizers.
  • the oligonucleotides of the invention can be formulated in liquid solutions, preferably in physiologically compatible buffers such as Hank's solution or Ringer's solution.
  • the oligonucleotides may be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms are also included in the invention.
  • compositions for topical administration include transdermal patches, ointments, lotions, creams, gels, drops, sprays, suppositories, liquids and powders.
  • conventional pharmaceutical carriers, aqueous, powder or oily bases, or thickeners may be used in pharmaceutical preparations for topical administration.
  • Pharmaceutical preparations for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, capsules, sachets or tablets.
  • thickeners, flavoring agents, diluents, emulsifiers, dispersing aids, or binders may be used in pharmaceutical preparations for oral administration.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are known in the art, and include, for example, for transmucosal administration bile salts and fusidic acid derivatives, and detergents.
  • Transmucosal administration may be through nasal sprays or using suppositories.
  • the oligonucleotides are formulated into conventional oral administration forms such as capsules, tablets, and tonics.
  • the oligonucleotides of the invention are formulated into ointments, salves, gels, or creams as known in the art.
  • Drug delivery vehicles can be chosen e.g., for in vitro, for systemic, or for topical administration. These vehicles can be designed to serve as a slow release reservoir or to deliver their contents directly to the target cell.
  • An advantage of using some direct delivery drug vehicles is that multiple molecules are delivered per uptake. Such vehicles have been shown to increase the circulation half-life of drugs that would otherwise be rapidly cleared from the blood stream.
  • Some examples of such specialized drug delivery vehicles which fall into this category are liposomes, hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres.
  • the described oligonucleotides may be administered systemically to a subject.
  • Systemic absorption refers to the entry of drugs into the blood stream followed by distribution throughout the entire body.
  • Administration routes which lead to systemic absorption include: intravenous, subcutaneous, intraperitoneal, and intranasal. Each of these administration routes delivers the oligonucleotide to accessible diseased cells.
  • the therapeutic agent drains into local lymph nodes and proceeds through the lymphatic network into the circulation.
  • the rate of entry into the circulation has been shown to be a function of molecular weight or size.
  • the use of a liposome or other drug carrier localizes the oligonucleotide at the lymph node.
  • the oligonucleotide can be modified to diffuse into the cell, or the liposome can directly participate in the delivery of either the unmodified or modified oligonucleotide into the cell.
  • the chosen method of delivery will result in entry into cells.
  • Preferred delivery methods include liposomes (10-400 nm), hydrogels, controlled-release polymers, and other pharmaceutically applicable vehicles, and microinjection or electroporation (for ex vivo treatments).
  • the pharmaceutical preparations of the present invention may be prepared and formulated as emulsions. Emulsions are usually heterogeneous systems of one liquid dispersed in another in the form of droplets usually exceeding 0.1 ⁇ m in diameter.
  • the emulsions of the present invention may contain excipients such as emulsifiers, stabilizers, dyes, fats, oils, waxes, fatty acids, fatty alcohols, fatty esters, humectants, hydrophilic colloids, preservatives, and anti- oxidants may also be present in emulsions as needed. These excipients may be present as a solution in either the aqueous phase, oily phase or itself as a separate phase.
  • excipients such as emulsifiers, stabilizers, dyes, fats, oils, waxes, fatty acids, fatty alcohols, fatty esters, humectants, hydrophilic colloids, preservatives, and anti- oxidants may also be present in emulsions as needed. These excipients may be present as a solution in either the aqueous phase, oily phase or itself as a separate phase.
  • Examples of naturally occurring emulsifiers that may be used in emulsion formulations of the present invention include lanolin, beeswax, phosphatides, lecithin and acacia. Finely divided solids have also been used as good emulsifiers especially in combination with surfactants and in viscous preparations. Examples of finely divided solids that may be used as emulsifiers include polar inorganic solids, such as heavy metal hydroxides, nonswelling clays such as bentonite, attapulgite, hectorite, kaolin, montmorillonite, colloidal aluminum silicate and colloidal magnesium aluminum silicate, pigments and nonpolar solids such as carbon or glyceryl tristearate.
  • polar inorganic solids such as heavy metal hydroxides, nonswelling clays such as bentonite, attapulgite, hectorite, kaolin, montmorillonite, colloidal aluminum silicate and colloidal magnesium aluminum silicate, pigments and
  • preservatives examples include methyl paraben, propyl paraben, quaternary ammonium salts, benzalkonium chloride, esters of p-hydroxybenzoic acid, and boric acid.
  • antioxidants examples include free radical scavengers such as tocopherols, alkyl gallates, butylated hydroxyanisole, butylated hydroxytoluene, or reducing agents such as ascorbic acid and sodium metabisulf ⁇ te, and antioxidant synergists such as citric acid, tartaric acid, and lecithin.
  • compositions of oligonucleotides are formulated as microemulsions.
  • a microemulsion is a system of water, oil and amphiphile which is a single optically isotropic and thermodynamically stable liquid solution.
  • microemulsions are prepared by first dispersing an oil in an aqueous surfactant solution and then adding a sufficient amount of a 4th component, generally an intermediate chain-length alcohol to form a transparent system.
  • Surfactants that may be used in the preparation of microemulsions include, but are not limited to, ionic surfactants, non-ionic surfactants, Brij 96, polyoxyethylene oleyl ethers, polyglycerol fatty acid esters, tetraglycerol monolaurate (ML310), tetraglycerol monooleate (MO310), hexaglycerol monooleate (PO310), hexaglycerol pentaoleate (PO500), decaglycerol monocaprate (MCA750), decaglycerol monooleate (MO750), decaglycerol sequioleate (S0750), decaglycerol decaoleate (DA0750), alone or in combination with cosurfactants.
  • ionic surfactants non-ionic surfactants
  • Brij 96 polyoxyethylene oleyl ethers
  • polyglycerol fatty acid esters tetraglycerol monolaurate
  • the cosurfactant usually a short-chain alcohol such as ethanol, 1-propanol, and 1-butanol, serves to increase the interfacial fluidity by penetrating into the surfactant film and consequently creating a disordered film because of the void space generated among surfactant molecules.
  • a short-chain alcohol such as ethanol, 1-propanol, and 1-butanol
  • Microemulsions may, however, be prepared without the use of cosurfactants and alcohol- free self-emulsifying microemulsion systems are known in the art.
  • the aqueous phase may typically be, but is not limited to, water, an aqueous solution of the drug, glycerol, PEG300, PEG400, polyglycerols, propylene glycols, and derivatives of ethylene glycol.
  • the oil phase may include, but is not limited to, materials such as Captex 300, Captex 355, Capmul MCM, fatty acid esters, medium chain (C 8 -Ci 2 ) mono, di, and tri-glycerides, polyoxyethylated glyceryl fatty acid esters, fatty alcohols, polyglycolized glycerides, saturated polyglycolized C 8 -CiO glycerides, vegetable oils and silicone oil.
  • materials such as Captex 300, Captex 355, Capmul MCM, fatty acid esters, medium chain (C 8 -Ci 2 ) mono, di, and tri-glycerides, polyoxyethylated glyceryl fatty acid esters, fatty alcohols, polyglycolized glycerides, saturated polyglycolized C 8 -CiO glycerides, vegetable oils and silicone oil.
  • Microemulsions are particularly of interest from the standpoint of drug solubilization and the enhanced absorption of drugs.
  • Lipid based microemulsions both oil/water and water/oil have been proposed to enhance the oral bioavailability of drugs.
  • Microemulsions offer improved drug solubilization, protection of drug from enzymatic hydrolysis, possible enhancement of drug absorption due to surfactant-induced alterations in membrane fluidity and permeability, ease of preparation, ease of oral administration over solid dosage forms, improved clinical potency, and decreased toxicity (Constantinides et al., Pharmaceutical Research, 1994, 11:1385; Ho et ai, J. Pharm. Sd., 1996, 85:138-143).
  • Microemulsions have also been effective in the transdermal delivery of active components in both cosmetic and pharmaceutical applications. It is expected that the microemulsion compositions and formulations of the present invention will facilitate the increased systemic absorption of oligonucleotides from the gastrointestinal tract, as well as improve the local cellular uptake of oligonucleotides within the gastrointestinal tract, vagina, buccal cavity and other areas of administration.
  • the present invention employs various penetration enhancers to affect the efficient delivery of nucleic acids, particularly oligonucleotides, to the skin of animals.
  • nucleic acids particularly oligonucleotides
  • the present invention employs various penetration enhancers to affect the efficient delivery of nucleic acids, particularly oligonucleotides, to the skin of animals.
  • non-lipophilic drugs may cross cell membranes if the membrane to be crossed is treated with a penetration enhancer.
  • penetration enhancers also act to enhance the permeability of lipophilic drugs.
  • Five categories of penetration enhancers that may be used in the present invention include: surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants.
  • agents may be utilized to enhance the penetration of the administered oligonucleotides include: glycols such as ethylene glycol and propylene glycol, pyrrols such as 2-15 pyrrol, azones, and terpenes such as limonene, and menthone.
  • the oligonucleotides, especially in lipid formulations, can also be administered by coating a medical device, for example, a catheter, such as an angioplasty balloon catheter, with a cationic lipid formulation. Coating may be achieved, for example, by dipping the medical device into a lipid formulation or a mixture of a lipid formulation and a suitable solvent, for example, an aqueous-based buffer, an aqueous solvent, ethanol, methylene chloride, chloroform and the like. An amount of the formulation will naturally adhere to the surface of the device which is subsequently administered to a patient, as appropriate. Alternatively, a lyophilized mixture of a lipid formulation may be specifically bound to the surface of the device. Such binding techniques are described, for example, in K. Ishihara et ah, Journal of Biomedical Materials Research, Vol. 27, pp. 1309-1314 (1993), the disclosures of which are incorporated herein by reference in their entirety.
  • the useful dosage to be administered and the particular mode of administration will vary depending upon such factors as the cell type, or for in vivo use, the age, weight and the particular animal and region thereof to be treated, the particular oligonucleotide and delivery method used, the therapeutic or diagnostic use contemplated, and the form of the formulation, for example, suspension, emulsion, micelle or liposome, as will be readily apparent to those skilled in the art.
  • dosage is administered at lower levels and increased until the desired effect is achieved.
  • the amount of lipid compound that is administered can vary and generally depends upon the amount of oligonucleotide agent being administered.
  • the weight ratio of lipid compound to oligonucleotide agent is preferably from about 1 : 1 to about 15:1, with a weight ratio of about 5 : 1 to about 10:1 being more preferred.
  • the amount of cationic lipid compound which is administered will vary from between about 0.1 milligram (mg) to about 1 gram (g).
  • mg milligram
  • g gram
  • the agents of the invention are administered to subjects or contacted with cells in a biologically compatible form suitable for pharmaceutical administration.
  • oligonucleotide is administered in a form in which any toxic effects are outweighed by the therapeutic effects of the oligonucleotide.
  • oligonucleotides can be administered to subjects. Examples of subjects include mammals, e.g. , humans and other primates; cows, pigs, horses, and farming (agricultural) animals; dogs, cats, and other domesticated pets; mice, rats, and transgenic non-human animals.
  • an active amount of an oligonucleotide of the present invention is defined as an amount effective, at dosages and for periods of time necessary to achieve the desired result.
  • an active amount of an oligonucleotide may vary according to factors such as the type of cell, the oligonucleotide used, and for in vivo uses the disease state, age, sex, and weight of the individual, and the ability of the oligonucleotide to elicit a desired response in the individual.
  • Establishment of therapeutic levels of oligonucleotides within the cell is dependent upon the rates of uptake and efflux or degradation. Decreasing the degree of degradation prolongs the intracellular half-life of the oligonucleotide.
  • chemically- modified oligonucleotides e.g., with modification of the phosphate backbone, may require different dosing.
  • an oligonucleotide and number of doses administered will depend upon the data generated experimentally and in clinical trials. Several factors such as the desired effect, the delivery vehicle, disease indication, and the route of administration, will affect the dosage. Dosages can be readily determined by one of ordinary skill in the art and formulated into the subject pharmaceutical compositions. Preferably, the duration of treatment will extend at least through the course of the disease symptoms. Dosage regime may be adjusted to provide the optimum therapeutic response. For example, the oligonucleotide may be repeatedly administered, e.g., several doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation. One of ordinary skill in the art will readily be able to determine appropriate doses and schedules of administration of the subject oligonucleotides, whether the oligonucleotides are to be administered to cells or to subjects.
  • the oligonucleotide compositions of the present invention can be used to treat any disease involving the expression of a MAP4K4 protein.
  • the inhibition of a MAP4K4 gene includes any fragments and isoforms of MAP4K4.
  • Diseases that can be treated by oligonucleotide compositions in accordance with the present methods include, just to illustrate: cancer, retinopathies, autoimmune diseases, inflammatory diseases (i.e., ICAM-I related disorders, Psoriasis, Ulcerative Colitus, Crohn's disease), viral diseases (i.e., HIV, Hepatitis C), and cardiovascular diseases.
  • Metabolic disorders or metabolic syndrome, is described by accepted synonyms, which includes, but is not limited to, syndrome X, insulin resistance syndrome, insulin-resistant hypertension, the metabolic hypertensive syndrome, dysmetabolic syndrome.
  • Components of the metabolic syndrome include, but is not limited to, glucose intolerance, impaired glucose tolerance, impaired fasting serum glucose, impaired fasting blood glucose, hyperinsulinemia, pre-diabetes, obesity, visceral obesity, hypertriglyceridemia, elevated serum concentrations of free fatty acids, elevated serum concentrations of C-reactive protein, elevated serum concentrations of lipoprotein(a), elevated serum concentrations of homocysteine, elevated serum concentrations of small, dense low- density lipoprotein (LDL)-cholesterol, elevated serum concentrations of lipoprotein-associated phospholipase (A2), reduced serum concentrations of high density lipoprotein (HDL)- cholesterol, reduced serum concentrations of HDL(2b)-cholesterol, reduced serum concentrations of adiponectin, adipogenesis, and albuminuria.
  • glucose intolerance impaired glucose tolerance
  • impaired fasting serum glucose impaired fasting blood glucose
  • hyperinsulinemia pre-diabetes
  • obesity visceral obesity
  • MAP4K4 silencing has been implicated in the inhibition of the migration of multiple carcinoma cell lines (Collins et al., 2006, Proc. Natl. Acad. ScI 103(10):3775-3780). Accordingly, it is contemplated that the use of MAP4K4 RNAi constructs according to the present invention may benefit certain types of cancers in which inhibition of rumor cell motility is desired.
  • in vitro treatment of cells with oligonucleotides can be used for ex vivo therapy of cells removed from a subject (e.g., for treatment of leukemia or viral infection) or for treatment of cells which did not originate in the subject, but are to be administered to the subject (e.g., to eliminate transplantation antigen expression on cells to be transplanted into a subject).
  • in vitro treatment of cells can be used in non-therapeutic settings, e.g., to evaluate gene function, to study gene regulation and protein synthesis or to evaluate improvements made to oligonucleotides designed to modulate gene expression or protein synthesis.
  • In vivo treatment of cells can be useful in certain clinical settings where it is desirable to inhibit the expression of a protein.
  • antisense therapy is reported to be suitable (see, e.g., U.S. Pat. No. 5,830,653) as well as respiratory syncytial virus infection (WO 95/22,553) influenza virus (WO 94/23,028), and malignancies (WO 94/08,003).
  • respiratory syncytial virus infection WO 95/22,553 influenza virus
  • malignancies WO 94/08,003
  • Other examples of clinical uses of antisense sequences are reviewed, e.g., in Glaser. 1996. Genetic Engineering News 16:1.
  • Exemplary targets for cleavage by oligonucleotides include, e.g., protein kinase Ca, ICAM-I, c-raf kinase, p53, c-myb, and the bcr/abl fusion gene found in chronic myelogenous leukemia.
  • HEK293 cells were transfected at 0.1 nM active RNA concentration. Target mRNA was measured 24 hours post-transfection using the methods described below. Three duplex sequences (10, 20 and 21) were chosen as lead hits based on their activity, seed complement frequency (SCF), and homology to the mouse MAP4K4 gene, as described below.
  • SCF seed complement frequency
  • duplexes used in the examples described herein are modified on the sense strand, but not on the antisense (guide) strand.
  • the sense strands have been modified to include 2'OMe on 12 of the 5'-most end nucleotides and on 10 of the 3'-most nucleotides.
  • the antisense strand complements the sense strand to form a blunt-ended duplex of 25 base-pairs.
  • RNAiMAX LIPOFECTAMINETM RNAiMAX.
  • Target mRNA levels were measured using a bDNA assay (Panomics QUANTIGENE ® ) 24 hours post-transfection.
  • MAP4K4 mRNA was normalized to Cyclophilin B (PPIB) mRNA.
  • the bDNA assay is a sandwich nucleic acid hybridization method that uses bDNA molecules to amplify signal from captured target RNA.
  • bDNA technology forms the basis of the FDA-approved clinical diagnostic VERSANT 3.0 assays for HIV, HCV and HBV viral load, that are offered commercially by Siemens and have been in use for over a decade.
  • Another advantage of bDNA assays is that RNA is measured directly from the sample source, without RNA purification or enzymatic manipulation, thereby avoiding inefficiencies and variability introduced by or errors inherent to these processes. The results were plotted and are shown in Figure 1 , which depicts the entire length of the MAP4K4 gene.
  • Each bar represents the silencing efficacy of a particular 25 base pair siRNA duplex which targets a region on MAP4K4 starting at the indicated start site. It can be seen, for example, that duplex sequences spanning the positions 2334 to 2345 of MAP4K4 are effective silencers. Generally, duplexes which effectively reduced target mRNA expression by more than 60% at a 0.1 nM dose were selected, which are listed in Table 1. However, sequences having low to moderate SCF are preferred, though not necessary, for the purpose of reducing potential off-targeting effects, as confirmed bioinformatically by Anderson et al. Additionally, to allow ease of pre-clinical testing in mice, sequences that he in regions that share high sequence homology between human and mouse sequences (e.g, those that are at least 95% homologous to the corresponding mouse sequence) are preferable.
  • Example 2 EC 50 Analysis of siRNA duplexes based off of lead sequences (duplexes 10, 20 and 21)
  • duplexes whose sequences are based off of select duplexes from Example 1 (i.e.,10, 20 and 21).
  • duplexes which effectively reduced target mRNA expression by more than 60% at a 0.1 nM dose were used to design and test additional duplexes. Additional selection factors are described in Example 1.
  • the additional sequences derived from the initial selected sequences are shown in Table 2, and the efficacy of each duplex is presented below. Table 2 compares each human sequence to the corresponding mouse sequence; mismatches, if any, are underlined and bolded in the mouse sequence.
  • Figure 2 A shows the results of duplexes that are based off of the duplex sequence 21 , namely, sequences containing sense strands corresponding to SEQ ID NOs:43 or 50 (see Table 2 below).
  • MAP4K4 expression was measured using cDNA hybridization assay as described above at 48 hours post-transfection.
  • the EC 50 values of these duplexes are as follows:
  • Figure 2B shows the results of duplexes that are based off of the duplex sequence 20, namely, sequences containing sense strands corresponding to SEQ ID NOs: 11, 12, 41 or 56 (see Table 2 below).
  • MAP4K4 expression was measured using cDNA hybridization assay as described above at 48 hours post-transfection.
  • the EC50 values of these duplexes are as follows: • Duplex containing SEQ ID NO:41 : 0.081 nM ⁇ 0.027 nM
  • FIG. 2C shows the results of duplexes that are based off of the duplex sequence 10, namely, sequences containing sense strands corresponding to SEQ ID NOs:21, 53, 19 or 54 (see Table 2 below).
  • MAP4K4 expression was measured using cDNA hybridization assay as described above at 48 hours post-transfection.
  • the EC 50 values of these duplexes are as follows:
  • Duplex containing SEQ ID NO:54 0.027 nM ⁇ 0.005 nM Table 2. Additional sequences derived from duplex sequences 10, 20, or 21 selected based on initial RNA screen. "Start Site (H)” and “Start Site (M)” indicate the starting position of each sequence in the human or mouse MAP4K4, respectively.
  • Table 3 Sequences of lead duplexes that were selected based on activity, mouse homology, and other beneficial attributes for development. The two duplexes were moved into mouse studies.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Genetics & Genomics (AREA)
  • Diabetes (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Hematology (AREA)
  • Biotechnology (AREA)
  • Obesity (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Endocrinology (AREA)
  • Cardiology (AREA)
  • Oncology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Emergency Medicine (AREA)
  • Communicable Diseases (AREA)
  • Ophthalmology & Optometry (AREA)
  • Transplantation (AREA)

Abstract

La présente invention concerne différentes constructions d’ARNi MAP4K4 ayant des activités inattendues de silençage de gène, et des utilisations de celles-ci. La construction a une région bicaténaire de 19 à 49 nucléotides, de préférence de 25, 26, ou 27 nucléotides, et de préférence à extrémités franches. La construction a des modifications minimales sélectives pour conférer un équilibre optimal d’activité biologique, toxicité, stabilité, et spécificité pour un gène cible. Par exemple, le brin sens peut être modifié (par exemple, une ou les deux extrémités du brin sens est/sont modifiées par quatre groupes 2'-O-méthyle), de sorte que la construction ne soit pas clivée par Dicer ou une autre ARNase III, et la longueur totale du brin antisens est chargée dans RISC. De plus, le brin antisens peut également être modifié par un groupe 2'-O-méthyle sur le 2ème nucléotide d’extrémité 5' pour réduire grandement le silençage hors cible. Les constructions de l’invention évitent grandement la réponse d’interféron et l’apoptose séquence-indépendante dans des cellules mammaliennes et présentent une meilleure stabilité sérique et une spécificité améliorée pour une cible.
PCT/US2009/006211 2008-11-19 2009-11-19 Inhibition de map4k4 via arni WO2010059226A2 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US13/130,194 US9074211B2 (en) 2008-11-19 2009-11-19 Inhibition of MAP4K4 through RNAI
US14/728,764 US11254940B2 (en) 2008-11-19 2015-06-02 Inhibition of MAP4K4 through RNAi

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US19966108P 2008-11-19 2008-11-19
US61/199,661 2008-11-19

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/130,194 A-371-Of-International US9074211B2 (en) 2008-11-19 2009-11-19 Inhibition of MAP4K4 through RNAI
US14/728,764 Continuation US11254940B2 (en) 2008-11-19 2015-06-02 Inhibition of MAP4K4 through RNAi

Publications (2)

Publication Number Publication Date
WO2010059226A2 true WO2010059226A2 (fr) 2010-05-27
WO2010059226A3 WO2010059226A3 (fr) 2010-07-15

Family

ID=42198720

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/006211 WO2010059226A2 (fr) 2008-11-19 2009-11-19 Inhibition de map4k4 via arni

Country Status (2)

Country Link
US (2) US9074211B2 (fr)
WO (1) WO2010059226A2 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9095504B2 (en) 2010-03-24 2015-08-04 Rxi Pharmaceuticals Corporation RNA interference in ocular indications
US10478503B2 (en) 2016-01-31 2019-11-19 University Of Massachusetts Branched oligonucleotides
US10633653B2 (en) 2015-08-14 2020-04-28 University Of Massachusetts Bioactive conjugates for oligonucleotide delivery
US11702659B2 (en) 2021-06-23 2023-07-18 University Of Massachusetts Optimized anti-FLT1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
US11753638B2 (en) 2016-08-12 2023-09-12 University Of Massachusetts Conjugated oligonucleotides
US11827882B2 (en) 2018-08-10 2023-11-28 University Of Massachusetts Modified oligonucleotides targeting SNPs

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10131904B2 (en) 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
WO2010008582A2 (fr) 2008-07-18 2010-01-21 Rxi Pharmaceuticals Corporation Système permettant d'administrer un médicament aux cellules phagocytaires
EP3336188B1 (fr) 2008-09-22 2020-05-06 Phio Pharmaceuticals Corp. Composés d'arni de taille réduite à auto-délivrance
WO2010059226A2 (fr) 2008-11-19 2010-05-27 Rxi Pharmaceuticals Corporation Inhibition de map4k4 via arni
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
EP2542058B1 (fr) 2010-03-02 2018-10-10 RXi Pharmaceuticals Corporation Dose à effet sensibilisant d'une composition topique immunomodulatrice gélifiée
KR20180044433A (ko) 2010-03-24 2018-05-02 알엑스아이 파마슈티칼스 코포레이션 진피 및 섬유증성 적응증에서의 rna 간섭
US9080171B2 (en) 2010-03-24 2015-07-14 RXi Parmaceuticals Corporation Reduced size self-delivering RNAi compounds
US9506033B2 (en) 2012-05-22 2016-11-29 University Of Massachusetts Compositions and methods for inducing myoblast differentiation and myotube formation
US9434951B2 (en) 2012-05-22 2016-09-06 University Of Massachusetts Compositions and methods for decreasing leukocyte extravasation and vessel fluid leakage
US10260089B2 (en) 2012-10-29 2019-04-16 The Research Foundation Of The State University Of New York Compositions and methods for recognition of RNA using triple helical peptide nucleic acids
RU2744194C2 (ru) 2013-12-02 2021-03-03 Фио Фармасьютикалс Корп Иммунотерапия рака
US11279934B2 (en) 2014-04-28 2022-03-22 Phio Pharmaceuticals Corp. Methods for treating cancer using nucleic acids targeting MDM2 or MYCN
JP6836987B2 (ja) 2014-09-05 2021-03-03 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. Tyrまたはmmp1を標的とする核酸を用いて老化および皮膚障害を処置するための方法
WO2017007825A1 (fr) 2015-07-06 2017-01-12 Rxi Pharmaceuticals Corporation Procédés pour le traitement de troubles neurologiques à l'aide d'une petite molécule synergique et approche thérapeutique utilisant des acides nucléiques
US11001845B2 (en) 2015-07-06 2021-05-11 Phio Pharmaceuticals Corp. Nucleic acid molecules targeting superoxide dismutase 1 (SOD1)
EP3365446A4 (fr) 2015-10-19 2019-06-26 Phio Pharmaceuticals Corp. Composés d'acides nucléiques de taille réduite à auto-administration ciblant des longs arn non codants

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5674683A (en) * 1995-03-21 1997-10-07 Research Corporation Technologies, Inc. Stem-loop and circular oligonucleotides and method of using
US6346416B1 (en) * 2000-08-29 2002-02-12 Isis Pharmaceuticals, Inc. Antisense inhibition of HPK/GCK-like kinase expression
US20050239731A1 (en) * 2001-05-18 2005-10-27 Sirna Therapeutics, Inc. RNA interference mediated inhibition of MAP kinase gene expression using short interfering nucleic acid (siNA)
US20070248659A1 (en) * 2005-11-18 2007-10-25 David Shanahan Individualized cancer therapy
US20080009012A1 (en) * 2006-03-16 2008-01-10 Dharmacon, Inc. Methods, libraries and computer program products for determining whether siRNA induced phenotypes are due to off-target effects

Family Cites Families (227)

* 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
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
US4201860A (en) 1978-05-09 1980-05-06 Bristol-Myers Company Purine derivatives
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4426330A (en) 1981-07-20 1984-01-17 Lipid Specialties, Inc. Synthetic phospholipid compounds
US5023243A (en) 1981-10-23 1991-06-11 Molecular Biosystems, Inc. Oligonucleotide therapeutic agent and method of making same
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US5643889A (en) 1984-07-11 1997-07-01 Temple University-Of The Commonwealth System Of Pennsylvania Cholesterol conjugates of 2'5'-oligoadenylate derivatives and antiviral uses thereof
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
US4737323A (en) 1986-02-13 1988-04-12 Liposome Technology, Inc. Liposome extrusion method
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5264423A (en) 1987-03-25 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Inhibitors for replication of retroviruses and for the expression of oncogene products
US4904582A (en) 1987-06-11 1990-02-27 Synthetic Genetics Novel amphiphilic nucleic acid conjugates
US5188897A (en) 1987-10-22 1993-02-23 Temple University Of The Commonwealth System Of Higher Education Encapsulated 2',5'-phosphorothioate oligoadenylates
US4924624A (en) 1987-10-22 1990-05-15 Temple University-Of The Commonwealth System Of Higher Education 2,',5'-phosphorothioate oligoadenylates and plant antiviral uses thereof
DE3738460A1 (de) 1987-11-12 1989-05-24 Max Planck Gesellschaft Modifizierte oligonukleotide
US5278302A (en) 1988-05-26 1994-01-11 University Patents, Inc. Polynucleotide phosphorodithioates
US5750666A (en) 1988-05-26 1998-05-12 Competitve Technologies, Inc. Polynucleotide phosphorodithioate compounds
ZA902710B (en) 1989-05-22 1991-12-24 Univ Georgia Res Found Enzyme luminescence assay
US4958013A (en) 1989-06-06 1990-09-18 Northwestern University Cholesteryl modified oligonucleotides
US5591722A (en) 1989-09-15 1997-01-07 Southern Research Institute 2'-deoxy-4'-thioribonucleosides and their antiviral activity
ATE190981T1 (de) 1989-10-24 2000-04-15 Isis Pharmaceuticals Inc 2'-modifizierte nukleotide
US5264562A (en) 1989-10-24 1993-11-23 Gilead Sciences, Inc. Oligonucleotide analogs with novel linkages
US5495009A (en) 1989-10-24 1996-02-27 Gilead Sciences, Inc. Oligonucleotide analogs containing thioformacetal linkages
US5486603A (en) 1990-01-08 1996-01-23 Gilead Sciences, Inc. Oligonucleotide having enhanced binding affinity
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US6395492B1 (en) 1990-01-11 2002-05-28 Isis Pharmaceuticals, Inc. Derivatized oligonucleotides having improved uptake and other properties
US5514786A (en) 1990-01-11 1996-05-07 Isis Pharmaceuticals, Inc. Compositions for inhibiting RNA activity
US6399754B1 (en) 1991-12-24 2002-06-04 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides
US5914396A (en) 1990-01-11 1999-06-22 Isis Pharmaceuticals, Inc. 2'-O-modified nucleosides and phosphoramidites
US5646265A (en) 1990-01-11 1997-07-08 Isis Pharmceuticals, Inc. Process for the preparation of 2'-O-alkyl purine phosphoramidites
US6153737A (en) 1990-01-11 2000-11-28 Isis Pharmaceuticals, Inc. Derivatized oligonucleotides having improved uptake and other properties
US6358931B1 (en) 1990-01-11 2002-03-19 Isis Pharmaceuticals, Inc. Compositions and methods for modulating RNA
US6005087A (en) 1995-06-06 1999-12-21 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5681941A (en) 1990-01-11 1997-10-28 Isis Pharmaceuticals, Inc. Substituted purines and oligonucleotide cross-linking
US5578718A (en) 1990-01-11 1996-11-26 Isis Pharmaceuticals, Inc. Thiol-derivatized nucleosides
US5658731A (en) 1990-04-09 1997-08-19 Europaisches Laboratorium Fur Molekularbiologie 2'-O-alkylnucleotides as well as polymers which contain such nucleotides
US5470967A (en) 1990-04-10 1995-11-28 The Dupont Merck Pharmaceutical Company Oligonucleotide analogs with sulfamate linkages
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5151510A (en) 1990-04-20 1992-09-29 Applied Biosystems, Inc. Method of synethesizing sulfurized oligonucleotide analogs
WO1991017424A1 (fr) 1990-05-03 1991-11-14 Vical, Inc. Acheminement intracellulaire de substances biologiquement actives effectue a l'aide de complexes de lipides s'auto-assemblant
US5602240A (en) 1990-07-27 1997-02-11 Ciba Geigy Ag. Backbone modified oligonucleotide analogs
JPH0874B2 (ja) 1990-07-27 1996-01-10 アイシス・ファーマシューティカルス・インコーポレーテッド 遺伝子発現を検出および変調するヌクレアーゼ耐性、ピリミジン修飾オリゴヌクレオチド
US5623070A (en) 1990-07-27 1997-04-22 Isis Pharmaceuticals, Inc. Heteroatomic oligonucleoside linkages
US5386023A (en) 1990-07-27 1995-01-31 Isis Pharmaceuticals Backbone modified oligonucleotide analogs and preparation thereof through reductive coupling
US5489677A (en) 1990-07-27 1996-02-06 Isis Pharmaceuticals, Inc. Oligonucleoside linkages containing adjacent oxygen and nitrogen atoms
JP3256539B2 (ja) 1990-08-28 2002-02-12 エポック・ファーマシューティカルズ・インコーポレイテッド 連結分子を介する3’―尾分子付オリゴヌクレオチドの固体支持体合成
US5561225A (en) 1990-09-19 1996-10-01 Southern Research Institute Polynucleotide analogs containing sulfonate and sulfonamide internucleoside linkages
CA2092002A1 (fr) 1990-09-20 1992-03-21 Mark Matteucci Liaisons internucleosidiques modifiees
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
US5599797A (en) 1991-10-15 1997-02-04 Isis Pharmaceuticals, Inc. Oligonucleotides having phosphorothioate linkages of high chiral purity
US5607923A (en) 1991-10-15 1997-03-04 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating cytomegalovirus having phosphorothioate linkages of high chiral purity
US5661134A (en) 1991-10-15 1997-08-26 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating Ha-ras or Ki-ras having phosphorothioate linkages of high chiral purity
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
US5594121A (en) 1991-11-07 1997-01-14 Gilead Sciences, Inc. Enhanced triple-helix and double-helix formation with oligomers containing modified purines
TW393513B (en) 1991-11-26 2000-06-11 Isis Pharmaceuticals Inc Enhanced triple-helix and double-helix formation with oligomers containing modified pyrimidines
US5359044A (en) 1991-12-13 1994-10-25 Isis Pharmaceuticals Cyclobutyl oligonucleotide surrogates
US5856455A (en) 1991-12-24 1999-01-05 Isis Pharmaceuticals, Inc. Gapped 2'-modified oligonucleotides
KR930016437A (ko) 1992-01-22 1993-08-26 귀틀라인, 슈미트 올리고뉴클레오티드 유사체, 이의 제조방법 및 용도
US5633360A (en) 1992-04-14 1997-05-27 Gilead Sciences, Inc. Oligonucleotide analogs capable of passive cell membrane permeation
US5817781A (en) 1992-06-01 1998-10-06 Gilead Sciences, Inc. Modified internucleoside linkages (II)
EP0577558A2 (fr) 1992-07-01 1994-01-05 Ciba-Geigy Ag Nucléosides carbocycliques contenant des noyaux bicycliques, oligonucléotides en dérivant, procédé pour leur préparation, leur application et des intermédiaires
US6346614B1 (en) 1992-07-23 2002-02-12 Hybridon, Inc. Hybrid oligonucleotide phosphorothioates
JP3351476B2 (ja) 1993-01-22 2002-11-25 三菱化学株式会社 リン脂質誘導体及びそれを含有するリポソーム
JPH08508491A (ja) 1993-03-31 1996-09-10 スターリング ウインスロップ インコーポレイティド ホスホジエステル結合をアミド結合に置き換えたオリゴヌクレオチド
AU6527094A (en) 1993-03-31 1994-10-24 Hybridon, Inc. Modified oligonucleotides having improved anti-influenza activity
FR2705099B1 (fr) 1993-05-12 1995-08-04 Centre Nat Rech Scient Oligonucléotides phosphorothioates triesters et procédé de préparation.
DE59407895D1 (de) 1993-05-12 1999-04-15 Novartis Ag Nukleoside und Oligonukleotide mit 2'-Ethergruppen
US6015886A (en) 1993-05-24 2000-01-18 Chemgenes Corporation Oligonucleotide phosphate esters
US5580972A (en) 1993-06-14 1996-12-03 Nexstar Pharmaceuticals, Inc. Purine nucleoside modifications by palladium catalyzed methods
US5428149A (en) 1993-06-14 1995-06-27 Washington State University Research Foundation Method for palladium catalyzed carbon-carbon coulping and products
US5534259A (en) 1993-07-08 1996-07-09 Liposome Technology, Inc. Polymer compound and coated particle composition
US5532130A (en) 1993-07-20 1996-07-02 Dyad Pharmaceutical Corporation Methods and compositions for sequence-specific hybridization of RNA by 2'-5' oligonucleotides
US5614621A (en) 1993-07-29 1997-03-25 Isis Pharmaceuticals, Inc. Process for preparing oligonucleotides using silyl-containing diamino phosphorous reagents
US5417978A (en) 1993-07-29 1995-05-23 Board Of Regents, The University Of Texas System Liposomal antisense methyl phosphonate oligonucleotides and methods for their preparation and use
CA2169645A1 (fr) 1993-09-02 1995-03-09 Nassim Usman Non-nucleotide renfermant un acide nucleique enzymatique
AU679566B2 (en) 1993-09-03 1997-07-03 Isis Pharmaceuticals, Inc. Amine-derivatized nucleosides and oligonucleosides
US5502177A (en) 1993-09-17 1996-03-26 Gilead Sciences, Inc. Pyrimidine derivatives for labeled binding partners
ES2127948T3 (es) 1993-10-27 1999-05-01 Ribozyme Pharm Inc Oligonucleotidos modificados en la posicion 2'-amido y 2'-peptido.
AU687492B2 (en) 1993-11-16 1998-02-26 Genta Incorporated Synthetic oligomers having phosphonate internucleosidyl linkages of undefined chirality mixed with non-phosphonate internucleosidyl linkages
US5646126A (en) 1994-02-28 1997-07-08 Epoch Pharmaceuticals Sterol modified oligonucleotide duplexes having anticancer activity
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
US5651981A (en) 1994-03-29 1997-07-29 Northwestern University Cationic phospholipids for transfection
US5625050A (en) 1994-03-31 1997-04-29 Amgen Inc. Modified oligonucleotides and intermediates useful in nucleic acid therapeutics
US5777153A (en) 1994-07-08 1998-07-07 Gilead Sciences, Inc. Cationic lipids
US5580731A (en) 1994-08-25 1996-12-03 Chiron Corporation N-4 modified pyrimidine deoxynucleotides and oligonucleotide probes synthesized therewith
US5591721A (en) 1994-10-25 1997-01-07 Hybridon, Inc. Method of down-regulating gene expression
US5681940A (en) 1994-11-02 1997-10-28 Icn Pharmaceuticals Sugar modified nucleosides and oligonucleotides
US5948767A (en) 1994-12-09 1999-09-07 Genzyme Corporation Cationic amphiphile/DNA complexes
US5767099A (en) 1994-12-09 1998-06-16 Genzyme Corporation Cationic amphiphiles containing amino acid or dervatized amino acid groups for intracellular delivery of therapeutic molecules
US5830430A (en) 1995-02-21 1998-11-03 Imarx Pharmaceutical Corp. Cationic lipids and the use thereof
US6420549B1 (en) 1995-06-06 2002-07-16 Isis Pharmaceuticals, Inc. Oligonucleotide analogs having modified dimers
US5981501A (en) 1995-06-07 1999-11-09 Inex Pharmaceuticals Corp. Methods for encapsulating plasmids in lipid bilayers
US5851548A (en) 1995-06-07 1998-12-22 Gen-Probe Incorporated Liposomes containing cationic lipids and vitamin D
WO1996040961A1 (fr) 1995-06-07 1996-12-19 Life Technologies, Inc. Transfection par lipides cationiques amelioree par des peptides
EP1489184A1 (fr) 1995-06-07 2004-12-22 Inex Pharmaceutical Corp. Particules d'acides nucléiques et de lipides préparées au moyen d'un intermédiaire de complexe hydrophobe d'acides nucléiques et de lipides et utilisation pour transférer des gènes
US5734041A (en) 1995-10-20 1998-03-31 Mcgill University Preparation of chiral phosphorothioate oligomers
US5705621A (en) 1995-11-17 1998-01-06 Isis Pharmaceuticals, Inc. Oligomeric phosphite, phosphodiester, Phosphorothioate and phosphorodithioate compounds and intermediates for preparing same
US5684143A (en) 1996-02-21 1997-11-04 Lynx Therapeutics, Inc. Oligo-2'-fluoronucleotide N3'->P5' phosphoramidates
US6331617B1 (en) 1996-03-21 2001-12-18 University Of Iowa Research Foundation Positively charged oligonucleotides as regulators of gene expression
US6444806B1 (en) 1996-04-30 2002-09-03 Hisamitsu Pharmaceutical Co., Inc. Conjugates and methods of forming conjugates of oligonucleotides and carbohydrates
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US5789416B1 (en) 1996-08-27 1999-10-05 Cv Therapeutics Inc N6 mono heterocyclic substituted adenosine derivatives
US6111085A (en) 1996-09-13 2000-08-29 Isis Pharmaceuticals, Inc. Carbamate-derivatized nucleosides and oligonucleosides
US6248878B1 (en) 1996-12-24 2001-06-19 Ribozyme Pharmaceuticals, Inc. Nucleoside analogs
CA2289702C (fr) 1997-05-14 2008-02-19 Inex Pharmaceuticals Corp. Encapsulation hautement efficace d'agents therapeutiques charges dans des vesicules lipidiques
US6028183A (en) 1997-11-07 2000-02-22 Gilead Sciences, Inc. Pyrimidine derivatives and oligonucleotides containing same
WO1999031073A1 (fr) 1997-12-15 1999-06-24 Yamanouchi Pharmaceutical Co., Ltd. Nouveaux derives de pyrimidine-5-carboxamide
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6020475A (en) 1998-02-10 2000-02-01 Isis Pharmeuticals, Inc. Process for the synthesis of oligomeric compounds
CA2326823A1 (fr) 1998-04-20 1999-10-28 Ribozyme Pharmaceuticals, Inc. Molecules d'acides nucleiques presentant de nouvelles compositions chimiques capables de moduler l'expression genique
EP1080103A4 (fr) 1998-05-21 2003-07-02 Isis Pharmaceuticals Inc Compositions et procedes pour l'administration non parenterale d'oligonucleotides
US20040241845A1 (en) 1998-05-22 2004-12-02 Luc Desgroseillers Mammalian staufen and use thereof
US6242589B1 (en) 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
US6277967B1 (en) 1998-07-14 2001-08-21 Isis Pharmaceuticals, Inc. Carbohydrate or 2′-modified oligonucleotides having alternating internucleoside linkages
US6271358B1 (en) 1998-07-27 2001-08-07 Isis Pharmaceuticals, Inc. RNA targeted 2′-modified oligonucleotides that are conformationally preorganized
US20040009938A1 (en) 1998-08-07 2004-01-15 Muthiah Manoharan Methods of enhancing renal uptake of oligonucleotides
US6043352A (en) 1998-08-07 2000-03-28 Isis Pharmaceuticals, Inc. 2'-O-Dimethylaminoethyloxyethyl-modified oligonucleotides
US6020483A (en) 1998-09-25 2000-02-01 Nexstar Pharmaceuticals, Inc. Nucleoside modifications by palladium catalyzed methods
US6455586B1 (en) 1998-12-15 2002-09-24 Leonard L. Kaplan Topical immunomodulating compositions for treatment of aids, Hepatitis B & C, other infectious diseases, and cancer
DE19956568A1 (de) 1999-01-30 2000-08-17 Roland Kreutzer Verfahren und Medikament zur Hemmung der Expression eines vorgegebenen Gens
US6465628B1 (en) 1999-02-04 2002-10-15 Isis Pharmaceuticals, Inc. Process for the synthesis of oligomeric compounds
TR200604211T1 (tr) 1999-02-12 2007-02-21 Daiichi Sankyo Company Limiteddaiichi Sankyo Company Limited Yeni nükleosid ve oligonükleotid analoglarıYeni nükleosid ve oligonükleotid analogları
US6207819B1 (en) 1999-02-12 2001-03-27 Isis Pharmaceuticals, Inc. Compounds, processes and intermediates for synthesis of mixed backbone oligomeric compounds
US6121437A (en) 1999-03-16 2000-09-19 Isis Pharmaceuticals, Inc. Phosphate and thiophosphate protecting groups
US6033910A (en) 1999-07-19 2000-03-07 Isis Pharmaceuticals Inc. Antisense inhibition of MAP kinase kinase 6 expression
GB9925459D0 (en) 1999-10-27 1999-12-29 Plant Bioscience Ltd Gene silencing
DE10160151A1 (de) 2001-01-09 2003-06-26 Ribopharma Ag Verfahren zur Hemmung der Expression eines vorgegebenen Zielgens
US7829693B2 (en) 1999-11-24 2010-11-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
US20050032733A1 (en) 2001-05-18 2005-02-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SiNA)
EP1272630A2 (fr) 2000-03-16 2003-01-08 Genetica, Inc. Procedes et compositions d'interference d'arn
ES2336887T5 (es) 2000-03-30 2019-03-06 Whitehead Inst Biomedical Res Mediadores de interferencia por ARN específicos de secuencias de ARN
AU2001273574A1 (en) 2000-07-24 2002-02-05 Krenitsky Pharmaceuticals Inc. Substituted 5-alkynyl pyrimidines having neurotrophic activity
US20020132788A1 (en) 2000-11-06 2002-09-19 David Lewis Inhibition of gene expression by delivery of small interfering RNA to post-embryonic animal cells in vivo
EP1873259B1 (fr) 2000-12-01 2012-01-25 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Interférence ARN à médiation par ARN 21 et 22nt
FR2818642B1 (fr) 2000-12-26 2005-07-15 Hoechst Marion Roussel Inc Nouveaux derives de la purine, leur procede de preparation, leur application a titre de medicaments, compositions pharmaceutiques et nouvelle utilistion
JP2004532616A (ja) 2000-12-28 2004-10-28 ジョンソン・アンド・ジョンソン・リサーチ・ピー・ティー・ワイ・リミテッド 二本鎖rna仲介遺伝子抑制
US20030077829A1 (en) 2001-04-30 2003-04-24 Protiva Biotherapeutics Inc.. Lipid-based formulations
WO2003070884A2 (fr) 2002-02-20 2003-08-28 Sirna Therapeutics, Inc. Inhibition de l'expression du gene de la mdr p-glycoproteine induite par l'interference d'arn, au moyen d'acide nucleique interferant court (sina)
US20070032441A1 (en) 2001-05-18 2007-02-08 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (sina)
US20030158403A1 (en) 2001-07-03 2003-08-21 Isis Pharmaceuticals, Inc. Nuclease resistant chimeric oligonucleotides
DE10133858A1 (de) 2001-07-12 2003-02-06 Aventis Pharma Gmbh Synthetische doppelsträngige Oligonucleotide zur gezielten Hemmung der Genexpression
AU2002329667A1 (en) 2001-07-30 2003-02-17 Uta Griesenbach Specific inhibition of gene expression by small double stranded rnas
EP1575976A4 (fr) 2001-11-02 2006-08-23 Insert Therapeutics Inc Procedes et compositions permettant l'utilisation therapeutique de l'interference arn
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
ATE508188T1 (de) 2002-02-01 2011-05-15 Life Technologies Corp Oligonukleotid-zusammensetzungen mit verbesserter wirksamkeit
EP1470144B1 (fr) 2002-02-01 2008-12-03 McGILL UNIVERSITY Oligonucleotides comportant des segments alternatifs et utilisations associees
WO2003064621A2 (fr) 2002-02-01 2003-08-07 Ambion, Inc. Courts fragments d'arn interferant haute activite visant a reduire l'expression de genes cibles
US20040014957A1 (en) 2002-05-24 2004-01-22 Anne Eldrup Oligonucleotides having modified nucleoside units
US20100009856A1 (en) 2002-06-21 2010-01-14 Sinogenomax Sompany LTD. Randomized dna libraries and double-stranded rna libraries, use and method of production thereof
EP1527176B2 (fr) 2002-08-05 2017-03-22 Silence Therapeutics GmbH Nouvelles formes de molecules d'arn interferant
WO2004027030A2 (fr) 2002-09-18 2004-04-01 Isis Pharmaceuticals, Inc. Reduction efficace d'arn cibles au moyen de composes oligomeres a brin simple et double
US20050020521A1 (en) 2002-09-25 2005-01-27 University Of Massachusetts In vivo gene silencing by chemically modified and stable siRNA
WO2004035765A2 (fr) 2002-10-18 2004-04-29 Nucleonics, Inc. Structures et constructions d'arn a double brin et procedes pour leur generation et leur utilisation
US20070254850A1 (en) 2002-10-30 2007-11-01 Judy Lieberman Methods for Treating and Preventing Apoptosis-Related Diseases Using Rna Interfering Agents
ES2334125T3 (es) 2002-11-04 2010-03-05 University Of Massachusetts Interferencia de arn especifico de alelos.
US9150605B2 (en) 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
PT2284266E (pt) * 2002-11-14 2013-12-17 Thermo Fisher Scient Biosciences Inc Siarn contra tp53
WO2004048545A2 (fr) 2002-11-26 2004-06-10 University Of Massachusetts Administration de sirnas
WO2004065600A2 (fr) 2003-01-17 2004-08-05 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Interference d'arn par des molecules d'arn palindromiques et marquees
DE10302421A1 (de) 2003-01-21 2004-07-29 Ribopharma Ag Doppelsträngige Ribonukleinsäure mit verbesserter Wirksamkeit
EP1589931A2 (fr) 2003-02-05 2005-11-02 University of Massachusetts Ciblage iarn de virus
US20040167090A1 (en) 2003-02-21 2004-08-26 Monahan Sean D. Covalent modification of RNA for in vitro and in vivo delivery
EP3450559A1 (fr) 2003-03-07 2019-03-06 Alnylam Pharmaceuticals, Inc. Compositions thérapeutiques
JP4605799B2 (ja) 2003-04-02 2011-01-05 ダーマコン, インコーポレイテッド Rna干渉において使用するための修飾ポリヌクレオチド
DK1620544T3 (en) 2003-04-17 2019-01-14 Alnylam Pharmaceuticals Inc MODIFIED iRNA AGENTS
US20060178327A1 (en) 2003-05-30 2006-08-10 Yeung Wah Hin A Inhibition of gene expression by delivery of specially selected double stranded or other forms of small interfering RNA precursors enabling the formation and function of small interfering RNA in vivo and in vitro
ES2357116T5 (es) 2003-06-02 2021-09-03 Univ Massachusetts Métodos y composiciones para mejorar la eficacia y especificad de FNAi
US7750144B2 (en) 2003-06-02 2010-07-06 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
EP3604537B1 (fr) 2003-06-13 2021-12-08 Alnylam Europe AG Acide ribonucléique double brin présentant une efficacité accrue dans un organisme
WO2005019430A2 (fr) 2003-08-22 2005-03-03 Research Development Foundation Selection in vitro de balises aptameres
ATE447024T1 (de) 2004-02-06 2009-11-15 Dharmacon Inc Stabilisierte rnas als transfektionskontrollen und silencing-reagentien
US20060069050A1 (en) 2004-02-17 2006-03-30 University Of Massachusetts Methods and compositions for mediating gene silencing
WO2005080582A1 (fr) 2004-02-20 2005-09-01 National Institute Of Advanced Industrial Science And Technology Adn et arn de localisation cytoplasmique
KR101147147B1 (ko) 2004-04-01 2012-05-25 머크 샤프 앤드 돔 코포레이션 Rna 간섭의 오프 타겟 효과 감소를 위한 변형된폴리뉴클레오타이드
MXPA06012076A (es) 2004-04-20 2007-01-25 Nastech Pharm Co Metodos y composiciones para mejorar el suministro de arn bicatenario o un acido nucleico hibrido bicatenario para regular la expresion genetica en celulas de mamifero.
AU2005252663B2 (en) * 2004-06-03 2011-07-07 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
US8138161B2 (en) 2004-10-01 2012-03-20 Novartis Vaccines And Diagnostics, Inc. Modified small interfering RNA molecules and methods of use
WO2006060743A2 (fr) 2004-12-02 2006-06-08 University Of Massachusetts Genes et polypeptides associes au transport du glucose et procedes d'utilisation de ceux-ci
US20100069620A1 (en) 2004-12-02 2010-03-18 Rxi Pharmaceuticals Corp. Novel compositions of chemically modified small interfering rna
JP2008524339A (ja) 2004-12-15 2008-07-10 メルク エンド カムパニー インコーポレーテッド Akt活性のインヒビター
EP1891217A2 (fr) 2005-05-27 2008-02-27 Sirna Therapeutics, Inc. Inhibition, mediee par l'interference d'arn, de l'expression genique du facteur 1 derive des cellules stromales (sdf-1), au moyen d'acide nucleique interferant court (sina)
WO2007030167A1 (fr) 2005-09-02 2007-03-15 Nastech Pharmaceutical Company Inc. Modification de molecules d'acide ribonucleique bicatenaire
WO2007056604A2 (fr) 2005-11-09 2007-05-18 Irm Llc Procédés et compositions servant à moduler la motilité cellulaire et à inhiber la métastase de tumeurs
JP2009519033A (ja) 2005-12-16 2009-05-14 ディアト 核酸を細胞に送達するための細胞貫通ペプチド結合体
US7825099B2 (en) 2006-01-20 2010-11-02 Quark Pharmaceuticals, Inc. Treatment or prevention of oto-pathologies by inhibition of pro-apoptotic genes
EP1981543A1 (fr) 2006-01-23 2008-10-22 Abbott Laboratories POLYMÈRE À POLYCATION CHIMIQUEMENT MODIFIÉ POUR ADMINISTRATION D'ARNsi
US7951784B2 (en) 2006-01-26 2011-05-31 University Of Massachusetts RNA interference agents for therapeutic use
WO2008036841A2 (fr) 2006-09-22 2008-03-27 Dharmacon, Inc. Complexes d'oligonucléotides tripartites et procédés de silençage de gènes par interférence arn
WO2008109353A1 (fr) 2007-03-02 2008-09-12 Mdrna, Inc. Composés d'acide nucléique permettant d'inhiber l'expression de gène map2k et utilisations de ceux-ci
WO2008109105A2 (fr) 2007-03-06 2008-09-12 Flagship Ventures Procédés et compositions pour obtenir des effets thérapeutiques améliorés avec l'arnsi
EP2205740A2 (fr) * 2007-10-02 2010-07-14 Rxi Pharmaceuticals Corp. CONSTRUCTIONS D'ARNi À STRUCTURE TRIPARTITE
BRPI0817605A2 (pt) 2007-10-03 2017-05-09 Quark Pharmaceuticals Inc novas estruturas de sirna
WO2009073809A2 (fr) 2007-12-04 2009-06-11 Alnylam Pharmaceuticals, Inc. Conjugués glucidiques utilisés en tant qu'agents d'administration pour des oligonucléotides
US10131904B2 (en) 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
WO2010006237A2 (fr) 2008-07-11 2010-01-14 Alnylam Pharmaceuticals, Inc. Oligonucléotides de phosphorothioate ou phosphorothiotes non nucléosidiques en tant qu'agents d’administration pour des agents d'arni
WO2010008582A2 (fr) * 2008-07-18 2010-01-21 Rxi Pharmaceuticals Corporation Système permettant d'administrer un médicament aux cellules phagocytaires
CA2731779A1 (fr) 2008-07-24 2010-01-28 Rxi Pharmaceuticals Corporation Constructions d'arni et leurs utilisations
MX2011002143A (es) 2008-08-25 2011-07-20 Excaliard Pharmaceuticals Inc Oligonucleotidos antisentido dirigidos contra el factor de crecimiento del tejido conectivo y usos de los mismos.
WO2010027830A2 (fr) 2008-08-25 2010-03-11 Excaliard Pharmaceuticals, Inc. Procédé de modulation de l'expression du facteur de croissance des tissus conjonctifs par administration d'oligonucléotides antisens par un système d'apport afin de réduire les cicatrices de cicatrisation et de traiter des maladies fibreuses
EP3336188B1 (fr) 2008-09-22 2020-05-06 Phio Pharmaceuticals Corp. Composés d'arni de taille réduite à auto-délivrance
WO2010059226A2 (fr) 2008-11-19 2010-05-27 Rxi Pharmaceuticals Corporation Inhibition de map4k4 via arni
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
EP2542058B1 (fr) 2010-03-02 2018-10-10 RXi Pharmaceuticals Corporation Dose à effet sensibilisant d'une composition topique immunomodulatrice gélifiée
WO2011109698A1 (fr) 2010-03-04 2011-09-09 Rxi Pharmaceuticals Corporation Formulations et procédés d'administration ciblée à des cellules phagocytaires
EP2550001B1 (fr) 2010-03-24 2019-05-22 Phio Pharmaceuticals Corp. Arn interférant dans des indications oculaires
US9080171B2 (en) 2010-03-24 2015-07-14 RXi Parmaceuticals Corporation Reduced size self-delivering RNAi compounds
KR20180044433A (ko) 2010-03-24 2018-05-02 알엑스아이 파마슈티칼스 코포레이션 진피 및 섬유증성 적응증에서의 rna 간섭
US20140072613A1 (en) 2012-09-10 2014-03-13 Cynthia Lander Compositions and Methods for Treating Cutaneous Scarring
RU2744194C2 (ru) 2013-12-02 2021-03-03 Фио Фармасьютикалс Корп Иммунотерапия рака
EP3077050B1 (fr) 2013-12-04 2020-10-21 Phio Pharmaceuticals Corp. Méthodes de traitement de cicatrisation à l'aide d'oligonucléotides chimiquement modifiés
US11279934B2 (en) 2014-04-28 2022-03-22 Phio Pharmaceuticals Corp. Methods for treating cancer using nucleic acids targeting MDM2 or MYCN
EP3137118A4 (fr) 2014-05-01 2017-11-22 RXi Pharmaceuticals Corporation Méthodes destinées à traiter les troubles affectant l'avant de l' il faisant appel à des molécules d'acide nucléique
JP6836987B2 (ja) 2014-09-05 2021-03-03 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. Tyrまたはmmp1を標的とする核酸を用いて老化および皮膚障害を処置するための方法
CA2969590A1 (fr) 2014-12-03 2016-06-09 Rxi Pharmaceuticals Corporation Methodes de traitement de l'alopecie en aires a l'aide d'approches de modulation genique
EP3277811B1 (fr) 2015-04-03 2020-12-23 University of Massachusetts Petit arn interférent asymétrique entièrement stabilisé
PT3277815T (pt) 2015-04-03 2021-11-11 Beth Israel Deaconess Medical Ct Inc Compostos de oligonucleótidos para o tratamento de pré-eclâmpsia e outros distúrbios angiogénicos
CA2980337A1 (fr) 2015-04-03 2016-10-06 University Of Massachusetts Composes oligonucleotidiques pour le ciblage de l'arnm de l'hungtingtine
WO2017007825A1 (fr) 2015-07-06 2017-01-12 Rxi Pharmaceuticals Corporation Procédés pour le traitement de troubles neurologiques à l'aide d'une petite molécule synergique et approche thérapeutique utilisant des acides nucléiques
US11001845B2 (en) 2015-07-06 2021-05-11 Phio Pharmaceuticals Corp. Nucleic acid molecules targeting superoxide dismutase 1 (SOD1)
CA2997949A1 (fr) 2015-09-11 2017-03-16 Rxi Pharmaceuticals Corporation Procedes de traitement de troubles et d'affections cutanes en utilisant des haptenes
EP3365446A4 (fr) 2015-10-19 2019-06-26 Phio Pharmaceuticals Corp. Composés d'acides nucléiques de taille réduite à auto-administration ciblant des longs arn non codants
WO2017173453A1 (fr) 2016-04-01 2017-10-05 The Brigham And Women's Hospital, Inc. Nanoparticules sensibles aux stimuli pour applications biomédicales
WO2018195355A1 (fr) 2017-04-19 2018-10-25 Rxi Pharmaceuticals Corporation Administration topique de composés d'acide nucléique
JP2020532955A (ja) 2017-08-07 2020-11-19 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. 化学修飾されたオリゴヌクレオチド

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5674683A (en) * 1995-03-21 1997-10-07 Research Corporation Technologies, Inc. Stem-loop and circular oligonucleotides and method of using
US6346416B1 (en) * 2000-08-29 2002-02-12 Isis Pharmaceuticals, Inc. Antisense inhibition of HPK/GCK-like kinase expression
US20050239731A1 (en) * 2001-05-18 2005-10-27 Sirna Therapeutics, Inc. RNA interference mediated inhibition of MAP kinase gene expression using short interfering nucleic acid (siNA)
US20070248659A1 (en) * 2005-11-18 2007-10-25 David Shanahan Individualized cancer therapy
US20080009012A1 (en) * 2006-03-16 2008-01-10 Dharmacon, Inc. Methods, libraries and computer program products for determining whether siRNA induced phenotypes are due to off-target effects

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9095504B2 (en) 2010-03-24 2015-08-04 Rxi Pharmaceuticals Corporation RNA interference in ocular indications
US10184124B2 (en) 2010-03-24 2019-01-22 Phio Pharmaceuticals Corp. RNA interference in ocular indications
US10662430B2 (en) 2010-03-24 2020-05-26 Phio Pharmaceuticals Corp. RNA interference in ocular indications
US11584933B2 (en) 2010-03-24 2023-02-21 Phio Pharmaceuticals Corp. RNA interference in ocular indications
US10633653B2 (en) 2015-08-14 2020-04-28 University Of Massachusetts Bioactive conjugates for oligonucleotide delivery
US10478503B2 (en) 2016-01-31 2019-11-19 University Of Massachusetts Branched oligonucleotides
US10799591B2 (en) 2016-01-31 2020-10-13 University Of Massachusetts Branched oligonucleotides
US11896669B2 (en) 2016-01-31 2024-02-13 University Of Massachusetts Branched oligonucleotides
US11753638B2 (en) 2016-08-12 2023-09-12 University Of Massachusetts Conjugated oligonucleotides
US11827882B2 (en) 2018-08-10 2023-11-28 University Of Massachusetts Modified oligonucleotides targeting SNPs
US11702659B2 (en) 2021-06-23 2023-07-18 University Of Massachusetts Optimized anti-FLT1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders

Also Published As

Publication number Publication date
US20120059046A1 (en) 2012-03-08
US9074211B2 (en) 2015-07-07
US20160115484A1 (en) 2016-04-28
WO2010059226A3 (fr) 2010-07-15
US11254940B2 (en) 2022-02-22

Similar Documents

Publication Publication Date Title
US10167471B2 (en) Inhibition of PCSK9 through RNAI
US11254940B2 (en) Inhibition of MAP4K4 through RNAi
JP7397019B2 (ja) サイズが減少した自己送達用RNAi化合物
US10633654B2 (en) Modified RNAi polynucleotides and uses thereof
US10479992B2 (en) RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US20110251258A1 (en) Rnai constructs and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09827879

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 13130194

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 09827879

Country of ref document: EP

Kind code of ref document: A2