WO2004015107A2 - Further novel forms of interfering rna molecules - Google Patents

Further novel forms of interfering rna molecules Download PDF

Info

Publication number
WO2004015107A2
WO2004015107A2 PCT/EP2003/008666 EP0308666W WO2004015107A2 WO 2004015107 A2 WO2004015107 A2 WO 2004015107A2 EP 0308666 W EP0308666 W EP 0308666W WO 2004015107 A2 WO2004015107 A2 WO 2004015107A2
Authority
WO
WIPO (PCT)
Prior art keywords
strand
ribonucleic acid
nucleotides
acid according
group
Prior art date
Application number
PCT/EP2003/008666
Other languages
French (fr)
Other versions
WO2004015107A3 (en
Inventor
Klaus Giese
Jörg Kaufmann
Anke Klippel-Giese
Original Assignee
Atugen Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=31720998&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2004015107(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from EP02017601A external-priority patent/EP1389637B1/en
Priority to CA2494930A priority Critical patent/CA2494930C/en
Priority to AU2003260370A priority patent/AU2003260370B2/en
Application filed by Atugen Ag filed Critical Atugen Ag
Priority to JP2004526885A priority patent/JP4705370B2/en
Priority to KR1020057002008A priority patent/KR101201664B1/en
Priority to EP06027129.3A priority patent/EP1857547B2/en
Priority to IL16654603A priority patent/IL166546A0/en
Priority to EP03784183.0A priority patent/EP1527176B2/en
Priority to MXPA05001355A priority patent/MXPA05001355A/en
Priority to SI200330726T priority patent/SI1527176T1/en
Priority to EP10012263.9A priority patent/EP2258847B2/en
Priority to BRPI0313202A priority patent/BRPI0313202A8/en
Priority to EP17160954.8A priority patent/EP3222724B1/en
Priority to DE60310944.6T priority patent/DE60310944T3/en
Priority to DK03784183.0T priority patent/DK1527176T4/en
Priority to ES03784183.0T priority patent/ES2280826T5/en
Publication of WO2004015107A2 publication Critical patent/WO2004015107A2/en
Publication of WO2004015107A3 publication Critical patent/WO2004015107A3/en
Priority to ZA2005/00459A priority patent/ZA200500459B/en
Priority to IL16654605A priority patent/IL166546A/en
Priority to CY20071100439T priority patent/CY1106422T1/en
Priority to AU2008207337A priority patent/AU2008207337A1/en
Priority to CY181101252T priority patent/CY1120919T1/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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P3/08Drugs 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/111General methods applicable to biologically active non-coding nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • C12Y207/11001Non-specific serine/threonine protein kinase (2.7.11.1), i.e. casein kinase or checkpoint kinase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/03Phosphoric monoester hydrolases (3.1.3)
    • C12Y301/03048Protein-tyrosine-phosphatase (3.1.3.48)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2310/13Decoys
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/317Chemical structure of the backbone with an inverted bond, e.g. a cap structure
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/318Chemical structure of the backbone where the PO2 is completely replaced, e.g. MMI or formacetal
    • C12N2310/3183Diol linkers, e.g. glycols or propanediols
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/332Abasic residue
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/343Spatial arrangement of the modifications having patterns, e.g. ==--==--==--
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/346Spatial arrangement of the modifications having a combination of backbone and sugar modifications
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • C12N2310/3521Methyl
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/352Nature of the modification linked to the nucleic acid via a carbon atom
    • C12N2310/3525MOE, methoxyethoxy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • 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
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • C12N2310/531Stem-loop; Hairpin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/50Methods for regulating/modulating their activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/50Methods for regulating/modulating their activity
    • C12N2320/51Methods for regulating/modulating their activity modulating the chemical stability, e.g. nuclease-resistance

Definitions

  • the present invention is related to a ribonucleic acid comprising a double-stranded structure whereby the double-stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to the target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid, the use of such ribonucleic acid, a cell and an organism, respectively, comprising such ribonucleic acid, a composition containing such ribonucleic acid, a pharmaceutical composition containing such ribonucleic acid and a method for inhibiting expression of a targeted gene.
  • RNA-mediated interference is a post-transcriptional gene silencing mechanism initiated by double stranded RNA (dsRNA) homologous in sequence to the silenced gene (Fire (1999), Trends Genet 15, 358-63, Tuschl, et al. (1999), Genes Dev 13, 3191-7, , Waterhouse, et al. (2001), Nature 411, 834-42, Elbashir, et al. (2001), Nature 411, 494-8, for review see Sharp (2001), Genes Dev 15, 485-90, Barstead (2001), Curr Opin Chem Biol 5, 63-6).
  • dsRNA double stranded RNA
  • RNAi has been used extensively to determine gene function in a number of organisms, including plants (Baulcombe (1999), Curr Opin Plant Biol 2, 109-13), nematodes (Montgomery, et al. (1998), Proc Natl Acad Sci U S A 95, 15502-7), Drosophila (Kennerdell, et al. (1998), Cell 95, 1017-26, Kennerdell, et al. (2000), Nat Biotechnol 18, 896-8). In the nematode C. elegans about one third ofthe genome has already been subjected to functional analysis by RNAi (Kim (2001), Curr Biol 11, R85-7, Maeda, et al. (2001), Curr Biol 11, 171-6).
  • RNAi in mammalian cells was not generally applicable, with the exception of early mouse development (Wianny, et al. (2000), Nat Cell Biol 2, 70-5).
  • the discovery that transfection of duplexes of 21-nt into mammalian cells interfered with gene expression and did not induce a sequence independent interferon-driven anti-viral response usually obtained with long dsRNA led to new potential application in differentiated mammalian cells (Elbashir et al. (2001), Nature 411, 494-8).
  • siRNAs small interfering RNAs resemble the processing products from long dsRNAs suggesting a potential bypassing mechanism in differentiated mammalian cells.
  • the Dicer complex a member of the RNAse III family, necessary for the initial dsRNA processing has been identified (Bernstein, et al. (2001), Nature 409, 363-6, Billy, et al. (2001), Proc Natl Acad Sci U S A 98, 14428-33).
  • One of the problems previously encountered when using unmodified ribooligonucleotides was the rapid degradation in cells or even in the serum-containing medium (Wickstrom (1986), J Biochem Biophys Methods 13, 97-102, Cazenave, et al. (1987), Nucleic Acids Res 15, 10507-21). It will depend on the particular gene function and assay systems used whether the respective knock down induced by transfected siRNA will be maintained long enough to achieve a phenotypic change.
  • the problem underlying the present invention was to provide synthetic interfering RNA molecules which are both stable and active in a biochemical environment such as a living cell.
  • a ribonucleic acid comprising a double stranded structure whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid, and whereby the double stranded structure is blunt ended.
  • a ribonucleic acid comprising a double stranded structure whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides, whereby said second stretch is at least partially identical to a target nucleic acid, whereby the first stretch and/or the second stretch have a length of 18 or 19 nucleotides.
  • the first stretch and/or the second stretch have a length of 18 or 19 nucleotides.
  • the double stranded structure is blunt ended on both sides ofthe double strand.
  • the double stranded structure is blunt ended on the double stranded structure which is defined by the 5'-end ofthe first strand and the 3 '-end ofthe second strand.
  • the double stranded structure is blunt ended on the double stranded structure which is defined by the 3 '-end ofthe first strand and the 5 '-end ofthe second strand.
  • a ribonucleic acid comprising a double stranded structure whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides and whereby said second stretch is at least partially identical to a target nucleic acid, and whereby at least one ofthe two strands has an overhang of at least one nucleotide at the 5 '-end.
  • the overhang consists of at least one nucleotide which is selected from the group comprising ribonucleotides and deoxyribonucleotides.
  • the nucleotide has a modification whereby said modification is preferably selected from the group comprising nucleotides being an inverted abasic and nucleotides having an NH 2 -modification at the 2 '-position.
  • At least one of the strands has an overhang of at least one nucleotide at the 3 '-end consisting of ribonucleotide or deoxyribonucleotide.
  • the first stretch and/or the second stretch have a length of 18 or 19 nucleotides.
  • the double-stranded structure has a length of 17 to 21 nucleotides, preferably 18 to 19 nucleotides.
  • the overhang at the 5 '-end is on the second strand.
  • the first strand comprises also an overhang, preferably at the 5 '-end.
  • the 3 '-end ofthe first strand comprises an overhang.
  • the overhang at the 5 '-end is on the first strand.
  • the second strand also comprises an overhang, preferably at the 5 '-end.
  • the 3 '-end ofthe first strand comprises an overhang.
  • At least one nucleotide of the ribonucleic acid has a modification at the 2'-position and the modification is preferably selected from the group comprising amino, fluoro, methoxy, alkoxy and alkyl.
  • a ribonucleic acid comprising a double stranded structure, whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides and whereby said second stretch is at least partially identical to a target nucleic acid,
  • said first strand and/or said second strand comprises a plurality of groups of modified nucleotides having a modification at the 2'-position whereby within the strand each group of modified nucleotides is flanked on one or both sides by a flanking group of nucleotides whereby the flanking nucleotides forming the flanking group of nucleotides is either an unmodified nucleotide or a nucleotide having a modification different from the modification of the modified nucleotides.
  • the ribonucleic acid is the ribonucleic acid according to the first, second or third aspect of the present invention.
  • said first strand and/or said second strand comprise said plurality of modified nucleotides.
  • said first strand comprises said plurality of groups of modified nucleotides.
  • said second strand comprises said plurality of groups of modified nucleotides.
  • the group of modified nucleotides and or the group of flanking nucleotides comprises a number of nucleotides whereby the number is selected from the group comprising one nucleotide to 10 nucleotides.
  • each range discloses any individual integer between the respective figures used to define the range including said two figures defining said range.
  • the group thus comprises one nucleotide, two nucleotides, three nucleotides, four nucleotides, five nucleotides, six nucleotides, seven nucleotides, eight nucleotides, nine nucleotides and ten nucleotides.
  • the pattern of modified nucleotides of said first strand is the same as the pattern of modified nucleotides of said second strand.
  • the pattern of said first strand aligns with the pattern of said second strand.
  • the pattern of said first strand is shifted by one or more nucleotides relative to the pattern ofthe second strand.
  • the modification is selected from the group comprising amino, fluoro, methoxy, alkoxy and alkyl.
  • the double stranded structure is blunt ended.
  • the double stranded structure is blunt ended on both sides.
  • the double stranded structure is blunt ended on the double stranded structure's side which is defined by the 5 '-end ofthe first strand and the 3 '-end ofthe second strand.
  • the double stranded structure is blunt ended on the double stranded structure's side which is defined by at the 3 '-end ofthe first strand and the 5'-end ofthe second strand.
  • At least one ofthe two strands has an overhang of at least one nucleotide at the 5'-end.
  • the overhang consists of at least one deoxyribonucleotide.
  • At least one ofthe strands has an overhang of at least one nucleotide at the 3 '-end.
  • the length of the double-stranded structure is from about 17 to 21 and more preferably 18 or 19 bases
  • the length of said first strand andor the length of said second strand is independently from each other selected from the group comprising the ranges of from about 15 to about 23 bases, 17 to 21 bases and 18 or 19 bases.
  • the complementarity between said first strand and the target nucleic acid is perfect.
  • the duplex formed between the first strand and the target nucleic acid comprises at least 15 nucleotides wherein there is one mismatch or two mismatches between said first strand and the target nucleic acid forming said double-stranded structure.
  • both the first strand and the second strand each comprise at least one group of modified nucleotides and at least one flanking group of nucleotides, whereby each group of modified nucleotides comprises at least one nucleotide and whereby each flanking group of nucleotides comprising at least one nucleotide with each group of modified nucleotides of the first strand being aligned with a flanking group of nucleotides on the second strand, whereby the most terminal 5' nucleotide ofthe first strand is a nucleotide ofthe group of modified nucleotides, and the most terminal 3' nucleotide of the second strand is a nucleotide of the flanking group of nucleotides.
  • each group of modified nucleotides consists of a single nucleotide and/or each flanking group of nucleotides consists of a single nucleotide.
  • the nucleotide forming the flanking group of nucleotides is an unmodified nucleotide which is arranged in a 3' direction relative to the nucleotide forming the group of modified nucleotides, and wherein on the second strand the nucleotide forming the group of modified nucleotides is a modified nucleotide which is arranged in 5' direction relative to the nucleotide forming the flanking group of nucleotides.
  • the first strand comprises eight to twelve, preferably nine to eleven, groups of modified nucleotides, and wherein the second strand comprises seven to eleven, preferably eight to ten, groups of modified nucleotides.
  • the target gene is selected from the group comprising structural genes, housekeeping genes, transcription factors, motility factors, cell cycle factors, cell cycle inhibitors, enzymes, growth factors, cytokines and tumor suppressors.
  • the first strand and the second strand are linked by a loop structure.
  • the loop structure is comprised of a non-nucleic acid polymer.
  • non-nucleic acid polymer is polyethylene glycol.
  • the loop structure is comprised of a nucleic acid.
  • the 5'-terminus ofthe first strand is linked to the 3'-terminus ofthe second strand.
  • the 3 '-end ofthe first strand is linked to the 5 '-terminus ofthe second strand.
  • the problem underlying the present invention is solved by the use of a ribonucleic acid according to any ofthe aspects ofthe present invention, for target validation.
  • the problem underlying the present invention is solved by the use of a ribonucleic acid according to any ofthe aspects ofthe present invention, for the manufacture of a medicament.
  • the medicament is for the treatment of a disease or of a condition which is selected from the group comprising glioblastoma, prostate cancer, breast cancer, lung cancer, liver cancer, colon cancer, pancreatic cancer and leukaemia, diabetes, obesity, cardiovascular diseases, and metabolic diseases.
  • a cell preferably a knockdown cell, containing a ribonucleic acid according to any of the aspects of the present invention.
  • an eighth aspect the problem underlying the present invention is solved by an organism, preferably a knockdown organism, containing a ribonucleic acid according to any of the aspects ofthe present invention.
  • composition containing a ribonucleic acid according to any ofthe aspects ofthe present invention.
  • the problem underlying the present invention is solved by a pharmaceutical composition containing a ribonucleic acid according to any of the aspects of the present invention, and a pharmaceutically acceptable carrier.
  • the problem underlying the present invention is solved by a method for inhibiting the expression of a target gene in a cell or derivative thereof comprising introducing of a ribonucleic acid according to any of the aspects of the present invention into the cell in an amount sufficient to inhibit expression of the target gene, wherein the target gene is the target gene of a ribonucleic acid according to any ofthe aspects ofthe present invention.
  • the present invention is based on the surprising finding that small interfering RNAs can be designed such as to be both highly specific and active as well as stable under the reaction conditions typically encountered in biological systems such as biochemical assays or cellular environments.
  • the various interfering RNAs described in the prior art such as by Tuschl et al. (international patent application WO 01/75164) provide for a length of 21 to 23 nucleotides and a modification at the 3' end of the double-stranded RNA.
  • RNAi small interfering RNA
  • RNAi consists of a ribonucleic acid comprising a double-stranded structure.
  • Said double-stranded structure is formed by a first strand and a second strand.
  • Said first strand comprises a stretch of contiguous nucleotides, also referred to as first stretch of contiguous nucleotides herein, and this first stretch is at least partially complementary to a target nucleic acid.
  • Said second strand comprises also a stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid.
  • the very basic structure of this ribonucleic acid is schematically shown in Fig. 1.
  • Said first strand and said second strand are preferably hybridised to each other and form the double-stranded structure.
  • the hybridisation typically occurs by Watson Crick base pairing.
  • the inventive ribonucleic acid is not necessarily limited in its length to said double- stranded structure. There might be further nucleotides added to each strand and/or to each end of any of the strands forming the RNAi.
  • the hybridisation or base pairing is not necessarily complete or perfect, which means that the first and the second stretch are not 100 % base paired due to mismatches. There might also be one or more mismatches within the duplex.
  • RNAi activity if placed outside a stretch of preferably 15, 16 or 17 matching nucleotides. If mismatches are placed to yield only 15 or less contiguous matching nucleotides, the RNAi molecule typically shows a reduced activity in down regulating mRNA for a given target compared to a 17 matching nucleotide duplex.
  • the first stretch of contiguous nucleotides of the first strand is essentially complementary to a target nucleic acid, more preferably to a part of the target nucleic acid.
  • Complementary as used herein preferably means that the nucleotide sequence ofthe first strand is hybridising to a nucleic acid sequence of a target nucleic acid sequence or a part thereof.
  • the target nucleic acid sequence or target nucleic acid is, in accordance with the mode of action of interfering ribonucleic acids, a single stranded RNA, more preferably an mRNA. Such hybridisation occurs most likely through Watson Crick base pairing, however, is not necessarily limited thereto.
  • the extent to which said first strand and more particularly the first stretch of contiguous nucleotides of said first strand is complementary to a target nucleic acid sequence can be as high as 100% and be as little as 80%, preferably 80-100%, more preferably 85-100%, most preferably 90-
  • Optimum complementarity seems to be 95-100%.
  • Complementarity in this sense means that the aforementioned range of nucleotides, such as, e. g., 80%-100%, depending on the particular range, of the nucleotides are perfect by Watson Crick base pairing. It is shown in one aspect ofthe present invention that the complementarity between said first stretch of nucleotides and the target RNA has to be 18-19 nucleotides, stretches of as little as 17 nucleotides even with two sequence specific overhangs are not functional in mediating RNAi.
  • 17 nucleotides or nucleotide base pairs would be allowable and functionally active.
  • the extent of complementarity required for a length of a duplex, i. e. of a double stranded structure can also be based on the melting temperature of the complex formed by either the double stranded structure as described herein or by the complex of the first stretch ofthe first strand and the target nucleic acid.
  • RNA mediated interference such as, for example, described in international patent applications WO 99/32619, WO 00/44895 and WO 01/75164.
  • the first strategy according to which an interfering ribonucleic acid molecule may be designed according to the present invention is to have an optimum length of 18 or 19 nucleotides of the stretch which is complementary to the target nucleic acid. It is also within the present invention that said optimum length of 18 or 19 nucleotides is the length ofthe double stranded structure in the RNAi used. This length requirement is clearly different from the technical teaching of the prior art such as, for example, international patent application WO 01/75164. It is within the present invention that any further design, both according to the present invention and as described in the prior art, can be realised in connection with an interfering ribonucleic acid having said length characteristics, i.e. a length of 18 or 19 nucleotides.
  • the second strategy according to which an interfering ribonucleic acid molecule may be designed is to have a free 5' hydroxyl group, also referred to herein as free 5' OH-group, at the first strand.
  • a free 5' OH-group means that the most terminal nucleotide forming the first strand is present and is thus not modified, particularly not by an end modification.
  • the terminal 5 '-hydroxy group of the second strand, respectively, is also present in an unmodified manner.
  • the 3 '-end of the first strand and first stretch, respectively is unmodified such as to present a free OH-group which is also referred to herein as free 3 'OH-group, whereby the design of the 5' terminal nucleotide is the one of any ofthe afore- described embodiments.
  • a free OH-group is also present at the 3 '-end of the second strand and second stretch, respectively.
  • the 3 '-end of the first strand and first stretch, respectively, and/or the 3 '-end of the second strand and second stretch, respectively may have an end modification at the 3' end.
  • free 5'OH-group and 3'OH-group also indicate that the respective most terminal nucleotide at the 5 'end and the 3 ' end of the polynucleotide, respectively, presents an OH-group.
  • Such OH-group may stem from either the sugar moiety of the nucleotide, more preferably from the 5 'position in case of the 5'OH-group and from the 3 'position in case of the 3'OH-group, or from a phosphate group attached to the sugar moiety of the respective terminal nucleotide.
  • the phosphate group may in principle be attached to any OH-group of the sugar moiety of the nucleotide.
  • the phosphate group is attached to the 5'OH-group of the sugar moiety in case of the free 5'OH-group and/or to the 3'OH-group of the sugar moiety in case ofthe free 3'OH-group still providing what is referred to herein as free 5' or 3' OH-group.
  • end modification means a chemical entity added to the most 5' or 3' nucleotide of the first and/or second strand.
  • examples for such end modifications include, but are not limited to, inverted (deoxy) abasics, amino, fluoro, chloro, bromo, CN, CF, methoxy, imidazole, caboxylate, thioate, d to Cio lower alkyl, substituted lower alkyl, alkaryl or aralkyl, OCF 3 , OCN, O-, S-, or N-alkyl; O-, S-, or N-alkenyl; SOCH 3 ; SO 2 CH 3 ; ONO 2 ; NO 2 , N 3 ; heterozycloalkyl; heterozycloalkaryl; aminoalkylamino; polyalkylamino or substituted silyl, as, among others, described in European patents EP 0 5
  • a further end modification is a biotin group.
  • Such biotin group may preferably be attached to either the most 5' or the most 3' nucleotide of the first and/or second strand or to both ends.
  • the biotin group is coupled to a polypeptide or a protein. It is also within the scope ofthe present invention that the polypeptide or protein is attached through any of the other aforementioned end modifications.
  • the polypeptide or protein may confer further characteristics to the inventive nucleic acid molecules. Among others the polypeptide or protein may act as a ligand to another molecule. If said other molecule is a receptor the receptor's function and activity may be activated by the binding ligand. The receptor may show an internalization activity which allows an effective transfection of the ligand bound inventive nucleic acid molecules.
  • An example for the ligand to be coupled to the inventive nucleic acid molecule is VEGF and the corresponding receptor is the NEGF receptor.
  • Table 1 Various embodiments o the interfering ribonucleic acid according to the present invention
  • the various end modifications as disclosed herein are preferably located at the ribose moiety of a nucleotide ofthe ribonucleic acid. More particularly, the end modification may be attached to or replace any of the OH-groups of the ribose moiety, including but not limited to the 2'OH, 3'OH and 5'OH position, provided that the nucleotide thus modified is a terminal nucleotide.
  • Inverted abasics are nucleotides, either desoxyribonucleotides or ribonucleotides which do not have a nucleobase moiety. This kind of compound is, among others, described in Sternberger et al.(2002), Antisense. Nucl. Ac. Drug Dev. in press.
  • any of the aforementioned end modifications may be used in connection with the various embodiments of RNAi depicted in table 1.
  • any of the RNAi forms or embodiments disclosed herein with the sense strand being inactivated, preferably by having an end modification more preferably at the 5' end are particularly advantageous. This arises from the inactivation of the sense strand which corresponds to the second strand of the ribonucleic acids described herein, which might otherwise interfere with an unrelated single-stranded RNA in the cell.
  • the expression and more particularly the translation pattern of the transcriptome of a cell is more specifically influenced. This effect is also referred to as off-target effect.
  • a third strategy subject to the present invention is to realise a ribonucleic acid comprising a double-stranded structure whereby the double-stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid whereby the double-stranded structure is blunt-ended.
  • the term double-stranded structure is also referred to as duplex. This design of RNAi is thus clearly different from, e.
  • overhang refers to a double-stranded structure whereby at least one end of one strand is longer than the corresponding end of the other strand forming the double-stranded structure, which is also referred to herein as the counter strand.
  • the first stretch is identical to the first strand and the second stretch is identical to the second strand.
  • blunt-ended RNAi Taken the effectiveness of blunt-ended RNAi and the advantages of an end modification of either the first or the second strand or both, respectively, of the respective ribonucleic acid it is preferred to have a combination of both design principles. In other words, it is within the present invention to have blunt-ended RNAi carrying any end modification scheme as depicted in table 1.
  • the fourth strategy subject to the present invention is to have an overhang at the 5 '-end of the ribonucleic acid. More particularly, such overhang may in principle be present at either or both the first strand and second strand of the ribonucleic acid according to the present invention.
  • the length of said overhang may be as little as one nucleotide and as long as 2 to 8 nucleotides, preferably 2, 4, 6 or 8 nucleotides. It is within the present invention that the 5' overhang may be located on the first strand and/or the second strand of the ribonucleic acid according to the present application.
  • the nucleotide(s) forming the overhang may be (a) desoxyribonucleotide(s), (a) ribonucleotide(s) or a continuation thereof.
  • the overhang preferably comprises at least one desoxyribonucleotide, whereby said one desoxyribonucleotide is preferably the most 5 '-terminal one. It is within the present invention that the 3 '-end ofthe respective counter-strand ofthe inventive ribonucleic acid does not have an overhang, more preferably not a desoxyribonucleotide overhang.
  • any of the inventive ribonucleic acids may comprise an end modification scheme as outlined in connection with table 1 and/or and end modification as outlined herein.
  • the fifth strategy in the design of interfering ribonucleic acids subject to the present application resides in the formation of a certain pattern of modified nucleotides on at least one ofthe strands and more particularly on one ofthe stretches of contiguous nucleotides ofthe ribonucleic acid(s) according to the present invention.
  • the kind of modification of said nucleotides may be the same as discussed in connection with the other strategies for designing interfering RNA as disclosed herein and more particularly the kind of modification described herein for or as an end modification, such as, e.g., inverted abasics, methoxy, or amino and the like at the ribose moiety of at least one nucleotide forming the ribonucleotide acids according to the present application.
  • the modification of said nucleotides may be any form of modification described herein, more particularly the kind of modification as described herein as end modification with the particularity that the so-called end modification is not necessarily located at terminal nucleotides.
  • the modification is occurring at a non-terminal nucleotide.
  • the modification is preferably attached to the ribose moiety of the - to be - modified nucleotide and even more preferably to the 2 'position ofthe ribose moiety.
  • any ribonucleic acid designed according to this strategy may also have the features conferred to a ribonucleic acid according to the present application by any of the other design strategies disclosed herein.
  • the interfering ribonucleic acid having a pattern of modified " nucleotides may have an end modification, an end modification scheme, may be blunt ended or may have a 5' overhang or any combination of two or more of these elements or characteristics.
  • the ribonucleic acid backbone as such may be further modified by forming different links between the nucleotides.
  • Such different links are, among others, described in European patent EP 0 586 520 BI and European patent EP 0 618 925 BI.
  • Of particular interest here are internal modification(s) ofthe ribonucleic acid backbone which have been shown to confer higher nuclease resistance of ribooligonucleotides.
  • the modification of the modified nucleotide is a methoxylation of the 2 '-OH-group ofthe ribose moiety ofthe nucleotide.
  • both strands, and more particularly both the first stretch and the second stretch show this kind of modification of the nucleotides forming said strands and stretches, respectively.
  • first strand and first stretch, respectively, or the second strand and second stretch, respectively show this particular pattern of modification of the nucleotides.
  • group of modified nucleotide or flanking group of nucleotide may comprise or represent as little nucleotides as one nucleotide, i. e. one or more nucleotides.
  • a pattern of modification of the nucleotides forming the stretch may be realised such that a single nucleotide or group of nucleotides which are covalently linked to each other via standard phosphorodiester bonds or, at least partially, through phosphorothioate bonds, show such kind of modification.
  • nucleotide or group of nucleotides which is also referred to herein as group of modified nucleotides, is not forming the 5 '-end or 3 '-end of said stretch a nucleotide or group of nucleotides follows on both sides ofthe nucleotide which does not have the modification of the preceding nucleotide or group of nucleotides. It is to be noted that this kind of nucleotide or group of nucleotides, however, may have a different modification. This kind of nucleotide or group of nucleotides is also referred to herein as the flanking group of nucleotides.
  • This sequence of modified nucleotide and group of modified nucleotides, respectively, and unmodified or differently modified nucleotide or group of unmodified or differently modified nucleotides may be repeated one or several times. Preferably, the sequence is repeated more than one time. For reason of clarity the pattern is discussed in more detail in the following, generally referring to a group of modified nucleotides or a group of unmodified nucleotides whereby each of said group may actually comprise as little as a single nucleotide.
  • Unmodified nucleotide as used herein means either not having any ofthe afore-mentioned modifications at the nucleotide forming the respective nucleotide or group of nucleotides, or having a modification which is different from the one ofthe modified nucleotide and group of nucleotides, respectively.
  • the modification of the unmodified nucleotide(s) wherein such unmodified nucleotide(s) is/are actually modified in a way different from the modification of the modified nucleotide(s), can be the same or even different for the various nucleotides forming said unmodified nucleotides or for the various flanking groups of nucleotides.
  • the pattern of modified and unmodified nucleotides may be such that the 5 '-terminal nucleotide of the strand or of the stretch starts with a modified group of nucleotides or starts with an unmodified group of nucleotides.
  • the 5 '-terminal nuleotide is formed by an unmodified group of nucleotides.
  • This kind of pattern may be realised either on the first stretch or the second stretch of the interfering RNA or on both. It has to be noted that a 5' phosphate on the target-complementary strand of the siRNA duplex is required for siRNA function, suggesting that cells check the authenticity of siRNAs through a free 5' OH (which can be phosphorylated) and allow only such bona fide siRNAs to direct target RNA destruction (Nykanen, et al. (2001), Cell 107, 309-21).
  • the first stretch shows a kind of pattern of modified and unmodified groups of nucleotides, i. e. of grou ⁇ (s) of modified nucleotides and flanking group(s) of nucleotides, whereas the second stretch does not show this kind of pattern.
  • This may be useful insofar as the first stretch is actually the more important one for the target-specific degradation process underlying the interference phenomenon of RNA so that for specificity reasons the second stretch can be chemically modified so it is not functional in mediating RNA interference.
  • both the first stretch and the second stretch have this kind of pattern.
  • the pattern of modification and non-modification is the same for both the first stretch and the second stretch.
  • the group of nucleotides forming the second stretch and corresponding to the modified group of nucleotides ofthe first stretch are also modified whereas the unmodified group of nucleotides of or forming the second stretch correspond to the unmodified group of nucleotides of or forming the first stretch.
  • Fig. 2A Another alternative is that there is a phase shift of the pattern of modification of the first stretch and first strand, respectively, relative to the pattern of modification of the second stretch and second strand, respectively.
  • the shift is such that the modified group of nucleotides of the first strand corresponds to the unmodified group of nucleotides of the second strand and vice versa.
  • Fig. 2B This possibility is shown in Fig. 2B.
  • the phase shift ofthe pattern of modification is not complete but overlapping as illustrated in Fig. 2C.
  • the second nucleotide at the terminus of the strand and stretch, respectively is an unmodified nucleotide or the beginning of group of unmodified nucleotides.
  • this unmodified nucleotide or unmodified group of nucleotides is located at the 5'- end of the first and second strand, respectively, and even more preferably ofthe first strand.
  • the unmodified nucleotide or unmodified group of nucleotide is located at the 5 '-end of the first strand and first stretch, respectively.
  • the pattern consists of alternating single modified and unmodified nucleotides.
  • the interfering ribonucleic acid subject comprises two strands, whereby a 2'-O-methyl modified nucleotide and a non-modified nucleotide, preferably a nucleotide which is not 2'-O-methyl modified, are incorporated on both strands in an alternate manner which means that every second nucleotide is a 2'-O-methyl modified and a non-modified nucleotide, respectively.
  • the second, and optionally fourth, sixth, eighth and/or similar position(s) at the 5' terminal end ofthe antisense strand which should not comprise any modification, whereas the most 5' terminal nucleotide, i. e. the first 5' terminal nucleotide of the antisense strand may - exhibit such modification with any uneven positions such as first, optionally third, fifth and similar position(s) at the antisense strand may be modified.
  • the modification and non-modification, respectively, of the modified and non-modified nucleotide(s), respectively may be anyone as described herein.
  • the double-stranded structure of the inventive ribonucleic acid which is also referred to as duplex, is formed by the first strand and second strand, respectively, or the first and second stretch of contiguous nucleotides.
  • the length of the first stretch and second stretch, respectively is typically about 15 to about 23, preferably 17 to 21, and more preferably 18 or 19 bases.
  • a length of less than 30 nucleotides, preferably less than 21 nucleotides does not cause any biological system which is basically capable of showing RNA interference and also interferon response, to develop an interferon response.
  • the ribonucleic acid comprises a double-stranded structure whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid whereby one terminus of the first strand and one terminus of the second strand are linked by a loop structure.
  • the loop structure is comprised of a non-nucleic acid polymer.
  • non- nucleic acid polymer may be polyethylene glycol or similar polymers.
  • the non-nucleic acid polymers may in principle be chosen from the group comprising polymers which do not comprise a polynucleotide and allow that the two strands to be linked may actually hybridize to each other.
  • the molecule or moiety of the molecule linking the two stretches hybridizing with each other has to have a certain molecular structure or molecular flexibility to allow the bending of the molecule so as to allow that both stretches get in close contact and in a three-dimensional orientation which permits hybridization.
  • Such molecule or moiety factually acts as a hinge.
  • any molecule which complies with this requirement may be used in connection with the present invention.
  • polyethylene glycol amino acid based molecules may be used.
  • Such amino acid based molecules may be either homopolymers or hetereopolymers.
  • a useful example is a homopolymer consisting of seven glycine residues which allows the generation of a hinge as required to bring the two stretches to hybridize in the close proximity as needed.
  • This glycine based hinge is described, e. g., in Guan K. L. and Dixon J. E. (1991), Anal Biochem. 192, 262.
  • the hinge may be formed by crown ethers known in the art.
  • the loop is comprised of a nucleic acid.
  • LNA as described in Elayadi and Corey (2001)Curr Opin Investig Drugs. 2(4):558-61. Review; Orum and Wengel (2001) Curr Opin Mol Ther. 3(3):239-43; and PNA are regarded as nucleic acids and may also be used as loop forming polymers.
  • the 5 '-terminus of the first strand may be linked to the 3 '-terminus of the second strand.
  • the 3 '-end of the first strand may be linked to the 5 '-terminus of the second strand.
  • the nucleotide sequence forming said loop structure is regarded as in general not being critical. However, the length of the nucleotide sequence forming such loop seems to be critical for sterical reasons. Accordingly, a minimum length of four nucleotides seems to be appropriate to form the required loop structure.
  • the maximum number of nucleotides forming the hinge or the link between both stretches to be hybridized is not limited. However, the longer a polynucleotide is, the more likely secondary and tertiary structures are formed and thus the required orientation of the stretches affected.
  • a maximum number of nucleotides forming the hinge is about 12 nucleotides. It is within the disclosure of this application that any ofthe designs described above may be combined with the present sixth strategy, i. e. by linking the two strands covalently in a manner that back folding (loop) can occur through a loop structure or similar structure.
  • the present inventors have surprisingly found that if the loop is placed 3' ofthe antisense strand, i. e. the first strand of the ribonucleic acid(s) according to the present invention, the activities of this kind of RNAi are higher compared to the placement of the loop 5' of the antisense strand. Accordingly, the particular arrangement of the loop relative to the " antisense strand and sense strand, i. e. the first strand and the second strand, respectively, is crucial and is thus in contrast to the understanding as expressed in the prior art where the orientation is said to be of no relevance. However, this seems not true given the experimental results presented herein.
  • RNAi RNAi-binding protein
  • the vector comprises a promoter for the expression of RNAi.
  • the respective promoter is pol III and more preferably the promoters are the U6, HI, 7SK promoter as described in Good et al. (1997) Gene Ther, 4, 45-54.
  • any gene producing such RNA may be modified in its expression by using any ofthe ribonucleic acid molecule according to the present invention. Because of this basic and generally applicable mechanism any application based thereon can be realised which imply the knockdown or knockout of a gene.
  • a preferred application is the use of the inventive ribonucleic acid for target validation.
  • target validation shall mean a process that involves taking steps to prove that a DNA, RNA, or protein molecule is directly involved in a biological process, preferably in a process - causally - involved in a disease or non-standard condition and is therefore a suitable target for development of a new therapeutic compound.
  • Sequence homology studies have successfully classified genes into target families. The enormous task of deciphering which of these targets are key players in diseases and which should be used for subsequent drug development needs to be addressed in an efficient manner. Therefore, the knockdown of gene expression should be reduced by 50-100%o, preferably by 90% to see significant effects on the phenotype. In other cases depending on the gene, a knockdown of as little as 20% might be sufficient to yield a phenotype.
  • a phenotype will be defined by comparison of cells containing functional RNAi molecules with cells containing non functional RNAi molecules. This will ensure a significant readout even under conditions where the protein function is inhibited only partially. Generally there is no linear correlation between the degree of rnRNA reduction and the extent of the change in phenotype. It has to be acknowledged that for some proteins a reduction of about 20 % of the protein is sufficient to create a change in the phenotype whereas in case of other genes and mRNA, respectively, as little as 5 to 10 % remaining protein is sufficient to maintain " the observed phenotype.
  • a further use of the ribonucleic acid molecules according to the present invention is its use for the manufacture of a medicament or its use as a medicament.
  • Such medicament could either be used for the treatment and/or prevention of diseases or conditions such as any type of cancer where a gene and its product have been linked to the onset, cause or progression of this disease. Additionally, such medicament could be used to treat diseases where the presence or overexpression of a gene product is causing a pathological phenotype.
  • the disease is characterised by a gain of function and may be remedied through application or administration of the corresponding, biologically active RNAi.
  • Diseases or conditions which may be treated by the medicament comprising a ribonucleic acid as disclosed herein may be selected from the group comprising cancer, heart diseases, metabolic diseases, dermatological diseases, inflammatory diseases, immune system disorders and autoimmune disorders.
  • the various forms of cancer include, but are not limited to, solid tumors and tumors of the hematopoietic system, such as glioblastoma, prostate cancer, breast cancer, lung cancer, liver cancer, pancreatic cancer and leukaemia.
  • Metabolic diseases include, but are not limited to, obesity and diabetes.
  • Dermatological diseases include, but are not limited to, psoriasis.
  • the ribonucleic acid molecules according to the present invention may be used as diagnostics, preferably for those diseases as specified in connection with the above-mentioned diseases and conditions.
  • diagnosis could be based on the observation that upon applying the ribonucleic acid molecules according to the present invention to a sample which preferably contains cells, a change in the expression pattern of the sample occurs.
  • a sample comprises cells from a subject from whom it is assumed that it may exhibit said disease or condition to be treated or a predisposition therefor.
  • a further application of the nucleic acids according to the present invention resides in their use in the screening of pharmaceutically active compounds and/or lead optimization.
  • the latter is done such as to monitor or determine the effect of candidate drugs such as small molecules and compare the effect created by said candidate drugs with the effect observed upon administering specific RNAi designed on the basis of the principles disclosed herein.
  • candidate drugs having off-target effects may be eliminated from the screening process whereas those candidate drugs which create a similar or identical phenotype are deemed as highly relevant lead compound or may even be a pharmaceutically active compound themselves.
  • the highly specific RNAi molecules act as gold standard against which candidate drugs are measured.
  • the invention is related to a cell, preferably a knockdown cell, which_ contains a ribonucleic acid as disclosed herein.
  • a cell preferably a knockdown cell, which_ contains a ribonucleic acid as disclosed herein.
  • Such cell is preferably a cell which is either isolated or contained in a tissue or even organ which again preferably is not contained in an organism. However, the cell may also be contained in an organism.
  • the cell is preferably a cell which is involved in the disease or condition which is to be treated by the inventive ribonucleic acids.
  • This kind of knock-down cells may be used to generate an expression profile based on, e. g., mRNA or protein, in order to elucidate functional relationship and to determine downstream targets.
  • the invention is related to an organism containing a ribonucleic acid as disclosed herein.
  • a ribonucleic acid as disclosed herein.
  • such organism is a vertebrate organism and more preferably the vertebrate organism is a mammal.
  • a mammal as used herein is, among others and not limited thereto, an ape, a dog, a cat, a goat, a sheep, a pig, a guinea pig, a rabbit, a mouse, a rat and a human being.
  • the present invention is related to a composition containing a ribonucleic acid according to the present invention.
  • a composition containing a ribonucleic acid according to the present invention.
  • Such composition comprises negative and positive controls either in combination with the effective ribonucleic acid or separated therefrom.
  • Such composition may further comprise a solvent, preferably a buffer.
  • the present invention is related to a pharmaceutical composition containing a ribonucleic acid according to the present invention and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are known to the one skilled in the art and comprise, among others, diluent, buffers and the like.
  • the pharmaceutical composition may comprise further pharmaceutically active compounds.
  • the disease or condition to be treated using the ribonucleic acid molecules according to the present invention are preferably those which are already now used in connection with the treatment of said diseases or conditions. Due to the different mode of action ofthe ribonucleic acid molecules according to the present invention and the medicaments used for the treatment of said diseases and conditions according to the prior art, synergistic effects will happen.
  • Fig. 1 shows a schematic illustration defining the terminology as used herein.
  • the upper of the two strand is the first strand and the antisense strand of the targeted nucleic acid such as mRNA.
  • the second strand is the one which essentially corresponds in its sequence to the targeted nucleic acid and thus forms the sense strand.
  • Both, the first strand and second strand form a double- stranded structure, typically through Watson Crick base pairing.
  • Fig. 2 illustrates some embodiments of the ribonucleic acid molecules of the present invention with patterns of modified and unmodified groups of nucleotides which is also referred to herein as pattern of modification.
  • the modified groups of nucleotides are also referred to herein as group of modified nucleotides.
  • the unmodified nucleotides or unmodified groups of nucleotides referred to as flanking group(s) of nucleotides herein, as used herein may also have one or several of the modif ⁇ cation(s) as disclosed herein which, however, is/are different from the modification of the nucleotides forming the group(s) of modified nucleotides.
  • flanking group(s) of nucleotides as used herein may also have one or several of the modif ⁇ cation(s) as disclosed herein which, however, is/are different from the modification of the nucleotides forming the group(s) of modified nucleotides.
  • the modified and unmodified groups of nucleotides i. e. the groups of modified nucleotides and the flanking groups of nucleotides on both the first stretch and the second stretch are located on corresponding parts of the stretches and are thus aligned to each other (groups of modified nucleotides on the first strand aligned with groups of modified nucleotides on the second strand and flanking groups of nucleotides on the first strand aligned with flanking group of nucleotides on the second strand), whereas in Fig.
  • the pattern realised on the first strand is also realised on the second strand, however, with a phase shift such that the modified group of nucleotides of the first stretch is base pairing with an unmodified group of nucleotides ofthe second stretch and vice versa so that a group of modified nucleotides on the first strand alignes with a flanking group of nucleotides on the second strand.
  • Fig. 2C a further possibility of arranging the modified and unmodified groups of nucleotides is realised.
  • the pattern of the first stretch is independent from the pattern of the second stretch and that both patterns partially overlap in terms of relative position to each other in the double-stranded structure defined by base pairing.
  • the extent of this overlapping can vary over the length ofthe stretch(es) and strand(s), respectively.
  • Fig. 3 shows the result of a knockdown experiment using RNAi molecules with different end protection groups. More particularly Fig. 3A shows that the various forms of end protected RNAi molecules are functional on the knockdown of PTEN mRNA.
  • Fig. 3B shows the sequence ofthe different RNAi molecules used in the experiment the result of which is depicted in Fig. 3A.
  • Fig. 3C shows the result of an immunoblot analysis of PTEN protein after treatment with modified RNAi molecules in comparison to PTEN specific antisense constructs.
  • Fig. 4 shows that the 3' overhang of RNAi molecules is not important for RNA interference. More particularly, Fig. 4A shows a dose response curve of different RNAi molecules and Fig. 4B shows the sequence of the RNAi molecules used in the experiment the result of which is shown in Fig. 4A.
  • Fig. 5 shows that duplex length ofthe RNAi molecules has to be at least 18-19 nucleotide. More particularly, Fig. 5B shows the sequence of the PTEN specific RNAi molecules used in the experiment the result of which is depicted in Fig. 5 A as dose response curve.
  • Fig. 6 shows that four terminal mismatched nucleotides in RNAi molecules with a length of 19 nucleotides are still functional in mediating Aktl knockdown. More particularly, Fig. 6B shows the sequence of the RNAi molecules used in the experiment the result of which is depicted in Fig. 6A.
  • Fig. 7 shows further results on duplex length requirements and tolerance for mutation in siRNAs. More particularly, Fig. 7A shows the various constructs used (left panel) and the respective impact on inhibition of Aktl mRNA expression in HeLa cells relative to the expression of pi 10a used in the indicated amounts of siRNA molecules (right panel). The nucleotide changes in the mismatch siRNA molecules are indicated by arrows; the 3' desoxynucleotide, if any, are indicated in capital letter.
  • Fig. 7B shows the various PTEN specific siRNAs (left panel), the inhibition of PTEN mRNA expression in HeLA cells expressed as ratio PTEN/pl 10a, at various amounts of siRNA (middle panel) and Fig. 7C a Western Blot analysis depicting the inhibition of PTEN protein expression using PTEN specific siRNA (30nM) and respective mismatch siRNA after 48 and 96 hours, respectively, with plOO ⁇ being used as loading control.
  • Fig. 8 shows the result of studies on the stability in serum conferred to RNAi molecules by 2'-O- methylation and that end modifications have no beneficial effects on RNAi stability. More particularly, Fig. 8A shows the result of a gel electrophoresis of the various RNAi molecules depicted in Fig. 8B being subject to incubation with fetal calf serum.
  • Fig. 9 shows that an amino end modification results in loss of activity.
  • Fig. 9B shows the particular RNAi molecules used in the experiments the result of which is shown in Fig. 9A expressed as PTEN/pl 10a expression level ratio.
  • Fig. 9C shows the design principles which may be deduced from the results depicted in Fig. 9A.
  • functional means functionally active in the particular assay system as described in the example and "not functional" means not functionally active in said system.
  • Fig. 10 shows that 2'-O-Alkyl (methyl) modifications stabilize RNAi molecules but also result in reduction of their activity. More particularly, Fig. 10C shows the sequence of the RNAi molecules used in the experiment the result of which is depicted as a dose response curve in Fig. 10 A. Fig. 10B shows the result of a gel electrophoresis of the various RNAi molecules depicted in Fig. 10C being subject to a two hour incubation in fetal calf serum.
  • Fig. 11 shows the result of an experiment on the efficacy of RNAi molecules with blocks of 2'- O-methyl modifications with Fig. 11 A graphically depicting the results of said experiments as a dose response curve and with Fig. 1 IC showing the sequences ofthe particular RNAi molecules used in said experiments.
  • Fig. 11B shows the result of a gel electrophoresis of the various RNAi molecules depicted in Fig. 1 IC being subject to a two hour incubation in fetal calf serum.
  • Fig. 12 shows that alternating 2'-O-methyl modification result in activity of the modified RNAi molecules compared to unmodified forms. More particularly, Fig. 12B shows the sequence ofthe RNAi molecules used in this experiment the result of which is depicted in Fig. 12A. Fig. 12C shows the stability of said RNAi molecules following incubation in serum for two hours, whereas Fig. 12D shows an immunoblot for PTEN protein upon application of different RNAi molecules to HeLa cells. As may be taken therefrom RNAi molecules with alternating modifications are stabilized against endonuclease degradation and active in mediating a PTEN protein knock down.
  • Fig. 13 shows the result of a Western Blot analysis to determine the time course of PTEN protein knock down.
  • Cells were continuously transfected with 2'-O-Methyl modified versus unmodified RNAi molecules using cationic lipids for 72 h. Protein lysates were prepared and analysed by immunoblot after 48 and 120 h. For the 96 h and 120 h timepoints the cells were splitted, replated and incubated in the absence of RNAi molecules for additional 24 and 48 h. .
  • Fig. 14 shows a Western Blot depicting the protein knock down of PTEN being persistent using alternating modified RNAi molecules versus unmodified RNAi molecules. Transfection were performed for only 5 h and new medium without transfection reagents was added. Lysates were analysed by immunoblot 72h and 96h post transfection with the indicated RNAi molecules.
  • Fig. 15 shows that siRNA molecules with distinct 2 '-O-methyl ribonucleotides modifications show increased stability in serum and mediate protein knock-down in HeLa cells. More particularly, Fig. 15A indicates the various siRNA molecule constructs used (left panel), whereby 2'-O-methyl ribonucleotides modifications are underlined and indicated by bold letters in the sequence. Inhibition of PTEN mRNA expression in HeLa cells transfected with the indicated amounts of modified siRNA molecules is expressed as ratio PTEN/pllO ⁇ and indicated on the right panel. Fig.
  • Fig. 15B shows on the left panel the various siRNA constructs used and on the right panel a PAA gel electrophoresis of modified and unmodified siRNA molecules after incubation in serum; the various constructs with 2'-O-methyl ribonucleotides are indicated by underlining and bold printing.
  • Fig. 15C shows an SDS-PAGE based immunoblot illustrating the inhibition of PTEN protein expression using various of the siRNA constructs (30 nM) as depicted in Fig. 15A and 15B, respectively. Again, pllO ⁇ is used as loading control.
  • Fig. 15D is an immunoblot indicating a prolonged protein knock-down, i. e. the inhibition of PTEN protein expression, upon administration of siRNA molecules (30 nM) with distinct 2'-O- methylribonucleotides modifications after 48 and 128 hours. As in Fig. 15C, p 110a is used as loading control.
  • Fig. 16 shows that siRNA molecules with distinct 2'-O-methylribonucleotides modifications which are specific for Aktl and pllO ⁇ mRNA show increased stability in serum and mediate protein knock-down in HeLa cells. More particularly, Fig. 16A indicates on the left panel the various constructs used whereby again 2'-O-methylribonucleotides are underlined and printed in bold. The integrity of the indicated siRNA molecules after incubation in serum is shown in the right panel. Fig. 16B shows an immunoblot of Aktl, Akt2 and Akt phosphorylation and pllO being used as a loading control upon transfection of the cells with the indicated siRNAs (30 mM). Fig. 16C shows various pi lO ⁇ specific siRNA constructs (left panel) with the 2'-O-methyl modifications being underlined and printed in bold, and the result of an immunoblot analysis
  • pi 10a (right panel) of the inhibition of the phosphorylation of the downstream kinase Aktl by said siRNA constructs, pi 10a has been used as a loading control.
  • Fig. 17 shows the efficacy of various RNAi molecules with hairpin structures as dose response curve while Fig. 17B shows the structure of said RNAi molecules the result of which is depicted in Fig. 17 A.
  • Synthetic siRNAs with different loops are functional in reducing the pllO ⁇ , Aktl and Akt 2 expression.
  • (14A) Inhibition of pllO ⁇ mRNA expression in siRNA transfected HeLa cells. Samples were analyzed in parallel for the level of pl lO ⁇ mRNA expression 24h after transfection of the indicated siRNAs.
  • the transfected bimolecular siRNAs (2 Imer with 3' TT overhangs, molecule 1 AB) or the monomolecular siRNAs with loop structures are schematically shown.
  • RNA was prepared und subjected to real time RT-PCR (Taqman) analysis.
  • pl lO ⁇ mRNA levels are shown relative to the mRNA levels of pl lO ⁇ , which serve as an internal reference.
  • Each bar represents triplicate transfections ( ⁇ standard deviation).
  • HeLa cells were transfected at 50% confluency (2500 cells per 96 well) with siRNAs at the indicated concentrations in growth medium.
  • Fig. 18 shows the efficacy of various RNAi molecules with intermolecular and intramolecular loop structures as dose response curves.
  • (18A) Inhibition of Aktl mRNA expression in siRNA transfected HeLa cells. Samples were analysed in parallel for the level of Aktl and Akt2 mRNA expression 24h after transfection ofthe indicated siRNAs. The different loops (A-loops; GAGA- loop and a polyethylenglycol (PEG)-linker) and their putative secondary structure are shown schematically.
  • the siRNA molecule 9A is specific for Akt2 and serves as a negative control. Note that 10 A and 10B do not contain self-complementary sequences and are transfected in combination.
  • Aktl mRNA levels is shown relative to the mRNA levels of pllO ⁇ , which served as internal control.
  • the Aktl specific molecule 7A serves here as a negative control.
  • Fig. 18 C shows a Western Blot analysis on Akt protein depicting the functionality of synthetic siRNAs with different loops in specifically reducing the Aktl and Akt 2 expression.
  • Inhibition of Aktl and Akt2 protein expression were analysed by immunoblot.
  • the cells were harvested 48h after transfection of the indicated hairpin siRNAs (20nM).
  • Cell extracts were separated by SDS- PAGE and analysed by immunoblotting using anti-pllO antibody, anti Akt 1/2. Similar results were obtained with an antibody specific for the phosphorylated form of Aktl.
  • the positions of pllO ⁇ , another catalytic subunit of PI 3-kinase, which was used as a loading control, and of Aktl, Akt2 and phosphorylated Akt (P*-Akt) are indicated on the left.
  • Fig. 19 shows an NH 2 modification, also referred to herein as amino modification, which may be present at either the 3'-OH terminal nucleotide or the 5' terminal nucleotide.
  • the amino group is attached to the phosphate which in turn is attached to the OH group ofthe sugar moiety, through an alkyl group comprising an alkyl chain of 1 to 8, preferably 6 C atoms, whereby the second C atom close to the phosphate group has a CH 2 OH group attached thereo.
  • the linker may be formed by an ether whereby the ether is comprised of two alcohols whereby one alcohol is an amino alcohol and the other is a dialcohol with one alcohol group involved in the formation of the ether group and the other one being an OH group located at either of the C atoms, preferably at the second C atom relative to the phosphate group.
  • siRNAs were purchased from Biospring (Frankfurt, Germany). The ribo-oligonucleotides were resuspended in RNase free TE to a final concentration of 50 ⁇ M. In the case of bimolecular siRNA molecules equal aliquots (100 ⁇ M) were combined to a final concentration of 50 ⁇ M. For the formation of intramolecular duplexes the siRNAs were incubated at 50 °C for 2 min in annealing buffer (25mM NaCl; 5mM MgCl 2 ) and were cooled down to RT.
  • annealing buffer 25mM NaCl; 5mM MgCl 2
  • RNAi molecules were transfected by adding preformed 5x concentrated complex of annealed RNAi and lipid in serum-free medium to cells in complete medium. Prior to transfection 2500 HeLa cells were plated per well 15 - 18 hours before transfection for the 96 well format.
  • the total transfection volume was 100 ⁇ l for cells plated in 96-wells and 10 ml for cells in 10 cm plates.
  • the final lipid concentration was 0.8 to 1.2 ⁇ g/ml depending on cell density; the RNAi concentration is indicated in each experiment.
  • RNA isolation was allowed to take place for 30 min at 37°C. Complexes were added to cells to yield a final lx concentration of both lipid and RNAi.
  • cells were lysed using a standard cell lysis buffer for protein extraction (Klippel, A., Escobedo, J. A., Hirano, M. and Williams, L. T. (1994). Mol Cell Biol 14, 2675-2685) or a denaturing buffer for RNA isolation according to the RNA isolation kit (Invitek, Berlin (Germany) 24 to 48 hours post transfection for RNA analysis and 48 to 72 hours post transfection for protein analysis by Western Blot.
  • RNA of cells transfected in 96-wells was isolated and purified using the Invisorb RNA HTS 96 kit (InVitek GmbH, Berlin). Inhibition of PTEN mRNA expression was detected by real time RT-PCR (Taqman) analysis using 300 nM PTEN 5' primer CACCGCCAAATTTAACTGCAGA, 300 nM PTEN 3' primer
  • the Akt primers and probes are determined in Sternberger et al. (Sternberger, a.a.O.) and are used according to the manufacturer's instructions (Applied Biosystem; use of Amplicon Set).
  • primers and probes may be designed using the software program Primer Express (Applied Biosystem).
  • the reaction was carried out in 50 ⁇ l and assayed on the ABI PRISM 7700 Sequence detector (Applied Biosystems) according to the manufacturer's instructions under the following conditions: 48°C for 30 min, 95°C for 10 min, followed by 40 cycles of 15 sec at 95°C and 1 min at 60°C.
  • RNA knockdown is shown by real time RT-PCR analysis of HeLa cells transfected with 21 nt long siRNA duplex molecules unmodified and modified with either NH 2 or inverted Abasics groups at the 5 '-end at a lipid carrier concentration of 1.0 ⁇ g/ml. Cell density was 2000 cells/well. Modifications on the 3 '-end are either RNA overhangs, RNA overhangs with amino groups or DNA overhangs.
  • Lysates were cleared by centrifugation at 14,000 x g for 5 minutes and aliquots of the cell extracts containing equal amounts of protein were analyzed for protein expression by Western-blotting: Samples were separated by SDS-PAGE and transferred to nitrocellulose-filters (Schleicher & Schuell). Filters were blocked in TBST buffer (10 mM Tris-HCl (pH 7.5), 150 mM NaCl, 0.05% (vol/vol) Tween 20, 0.5% (wt/vol) sodium azide) containing 5% (wt/vol) dried milk. The respective antibodies were added in TBST at appropriate dilutions.
  • Bound antibody was detected using anti-mouse- or anti-rabbit-conjugated horse radish peroxidase (Transduction Laboratories) in TBST, washed, and developed using the SuperSignal West Dura (Pierce) or ECL (Amersham) chemoluminescence substrates (c.f. Sternbergeret al. (2002). Antisense. Nucl. Ac. Drug Dev. in press..
  • the murine monoclonal anti-pl lO antibody U3A and the murine monoclonal anti- p85 antibody N7B have been described (Klippel et al., 1994, aaO).
  • Rabbit polyclonal anti-Akt and anti-phospho Akt (S473) antibodies were obtained from Cell Signaling Technology.
  • the murine monoclonal anti-PTEN antibody was from Santa Cruz Biotechnology.
  • the PTEN 53 specific antisense molecule, i. e. geneBloc, is described in Sternberg et al.
  • RNAi molecules used in this and other examples disclosed herein are also referred to as small interfering RNA molecules siRNA. It is to be noted that in any of the figures contained herein the upper strand is the antisense or first strand, whereas the lower strand is the sense or second strand ofthe interfering RNA molecule.
  • amino end modifications such as amino modification and inverted abasics modification of the terminal OH group of the nucleic acid are as potent as unmodified ends when the modification is located at the 3' end ofthe antisense strand (see also Fig. 8A; 8B). Therefore chemical modification to stabilize or with other beneficial properties (delivery) will be tolerated without activity loss when located at the 3' OH; especially when the 3 'OH is located on an overhanging nucleotide.
  • RNAi modified by a NH 2 group at the 3 '-position ofthe ribose moiety or by an inverted abasic at said positions.
  • the first construct is designated as siRNA-NH 2 (3A3B)
  • siRNA-iB 4A4B
  • the sequence of both molecules is depicted in Fig. 3B.
  • 3A3B indicates that the interfering ribonucleic acid consists of strand 3 A as the antisense strand and strand 3B as the sense strand.
  • GB53 Steinberger et al., supra
  • RNAi molecules depicted in Fig.3B are functional in yielding a PTEN protein knockdown.
  • RNAi 18 is composed of strand 18A as antisense strand and strand 18B as sense strand.
  • Blunt ended molecules are compared to molecules with 3'-overhangs (RNAi 18) and 5'- overhangs (RNAi 30 and RNAi 31) in their activity to knockdown PTEN mRNA in HeLa cells.
  • the activity of blunt ended molecules (RNAi 28) and molecules with 5 '-overhangs is comparable to the activity of molecules with 3 '-overhangs. This shows that 3 '-overhangs are not essential for RNAi activity.
  • Example 3 Duplex length requirements of interfering RNA molecules for RNAi activity in vivo
  • the experimental approach was similar to the one outlined in connection with example 1 except that the interfering RNA molecules were directed against the mRNA of Aktl.
  • the negative control to show the specificity ofthe RNAi molecules was again pi 10 mRNA.
  • the experimental results are shown in Fig. 5A with the particularities of the interfering RNAi molecules used being represented in Fig. 5B.
  • Similar experiments were carried out with further siRNA constructs which are depicted in Fig. 7A, left panel, whereby the arrows indicate mismatches and desoxyribonucleotides are expressed in capital letters.
  • the inhibition of Aktl mRNA expression in HeLa cells transfected with the indicated amounts of siRNA molecules is depicted on the right panel of Fig. 7 A.
  • RNAi 51 A/5 IB referred to as RNAi 51 corresponds to the one as described in international patent application WO 01/75164.
  • the RNAi molecule 55A/55B comprises a stretch of 17 nucleotides and has a clearly decreased activity in terms of degradation of Aktl mRNA.
  • nt long duplexes are highly efficient in reducing Aktl mRNA levels independent of the nature (desoxy- or ribonucleotides) of the 3' overhang (compare molecules 1AB, 2AB, 3AB, 4AB).
  • the 17 nucleotide long siRNA (molecule 5AB) showed a dramatically reduced silencing activity confirming the above expressed understanding that active siRNA duplexes should be at least 18 nt or longer. Without being wished to be bound by any theory this result may be explained mechanistically by two different requirements.
  • RNA-induced silencing complex requires a minimum length of the siRNA duplex.
  • a 19 nt long siRNA duplex molecule with one and two terminal mutations (CG and UA inversion) relative to the wildtype sequence was synthesised (molecules 6AB and 7AB). Both molecules, even the molecule with a stretch of only 15 nt base pairing to the target mRNA were functional in inducing the Aktl mRNA level.
  • the duplex length itself but not the base pairing of the antisense siRNA with the target mRNA seems to determine the minimum length of functional siRNAs. This suggests that the length of the double-stranded helix is an important determinant for the incorporation into the RISC complex. The introduced mismatches at the terminal ends ofthe siRNA duplexes had little effect on RNA interference.
  • RNAi mediated interference is thus a duplex length of 18 or 19 nucleotides, independent of the further design of the RNAi molecules such as blunt end or 5 '-overhang or any other form as disclosed herein, but generally applicable to RNAi molecules.
  • the particular design of the RNAi molecules may confer further advantages to said molecules, such as, e. g., increased efficiency and increased stability, respectively.
  • Example 4 Target -antisense homology requirements for RNAi in vivo
  • RNAi is specific for Aktl.
  • a PTEN specific interfering RNA molecule was designed and used as negative control.
  • the results are shown in Fig. 6A and Fig. 6B.
  • Fig. 7B Basically the same experiment was carried out using further siRNA molecules as depicted in Fig. 7B with the results being indicated in Fig. 7B (right panel) and Fig. 7C, respectively.
  • the present invention is reducing this off-target problem of siRNA by two approaches. First by reducing the molecule length of the siRNA molecules to the minimal requirements (18-19 nt) and thereby reducing the chance of homology to off-targets. Second, by inactivation of the sense strand to prevent a unwanted RNA silencing caused by accidential complemetarity ofthe sense strand to a unrelated target RNA (see also Example 6).
  • Example 5 Stability of modified RNAi molecules in serum
  • Oligonucleotides were incubated in human serum for 15 min and two hours and loaded on 10% polyacrylamide gel with untreated controls. The results are shown in Fig. 8 A. The various RNAi molecules used are shown and described in more detail in Fig. 8B.
  • RNAi duplex of RNA molecules with all nucleotides modified with 2 '-O-methyl groups RNAi molecules 79A79B and 28A28B
  • end protection e.g. iB or Amino
  • a further or additional modification of the nucleotides may be the use of a phosphorothioate backbone of the RNAi molecules which may be either complete or partial in order to inhibit endonuclease function.
  • a complete phosphorothioate backbone means that any of the nucleotides exhibits a phosphorothioate group whereas a partial phosphorothioate backbone means that not all of the nucleotides forming the RNAi molecule have a phosphorothioate modification.
  • This modification is suitable to increase the lifetime of RNAi molecules irrespective of the further design of RNAi molecules.
  • RNAi a partially or completely phosphorothioate modified RNAi is subject to the present invention which may be realized in connection with the different strategies for the design of interfering RNA molecules as disclosed herein or with any ofthe designs known in the art.
  • Example 6 Inactivation of the sense strand by NH 2 end protection groups on the 5' and
  • the experimental set-up was similar to the one described in connection with example 1 with the target nucleic acid sequence being PTEN mRNA.
  • the concentration of HeLa cells was 2,000 cells per well.
  • RNA of PTEN was analysed in Taqman assays after transfection of differently modified RNAi molecules.
  • the different interfering RNA molecules used are depicted in Fig. 9B whereas the experimental results are shown in Fig. 9A.
  • RNAi molecules are functional when the sense strand, i. e. the second stand, is modified on both ends with amino groups. Particularly effective are RNAi molecules 20A26B, 18A26B, and 28A26B. The lowest activity is shown by RNAi molecule 26A26B which corresponds to end modification on all 4 ends ofthe duplex (Tuschl is 18AB) .
  • RNAi activity is also achieved when the antisense strand, i. e. the first strand, is modified only at the 3' end leaving a free OH group at the 5' end (RNAi constructs 22A26B; 20A26B). There is no activity when the antisense strand is modified with amino groups on both the 5' and the 3' end (26A26B). This leads to the conclusion that any end ofthe antisense strand and more particularly the 5' end of the antisense should be kept without modifications.
  • the NH 2 end modification can be used to inactivate the sense strand on the 5' and 3' end and therefore reduce off-target effects mediated by an otherwise functional sense strand which results in a significantly increased specificity of the RNAi molecule which is advantageous for target validation as well as for any medical use ofthe RNAi molecule.
  • RNAi are accordingly those not having an amino modification at the antisense strand or having an amino modification only at the 3' end of the antisense strand whereas an amino modification at both ends of the antisense strand is not functional, i. e. does not result in a knockdown ofthe target mRNA.
  • Example 7 Impact of 2 '-O-methyl modification of RNAi molecules for endonuclease protection.
  • RNA knockdown is again shown using the real time RT-PCR analysis on HeLa cells transfected with RNAi duplex molecules directed against the PTEN mRNA as represented in Fig. 10 A.
  • Experimental procedures were basically the same as specified in example 1.
  • the nucleotides printed in bold are those having a 2'-O-methyl modification.
  • RNAi molecules in Fig. 10A It is illustrated by the dose response curves shown for various RNAi molecules in Fig. 10A that internal 2'-O-alkyl groups are reducing RNAi activity.
  • Preferably such 2'-O-alkyl groups are 2'- O-methyl or 2'-O-ethyl groups.
  • molecules with unmodified nucleotides in combination with 2'-O-alkyl modification show significant activity.
  • Fig. 10A there is no activity when the antisense strand is all modified with 2 '-O-methyl groups and the sense strand is not modified (c. f, e. g., RNAi molecule 79A28B).
  • RNAi molecules has to balance stability against activity which makes it important to be aware of the various design principles as disclosed in the present application.
  • Example 8 Impact of blocks of internal 2 '-O-methyl modifications on the stability of RNAi molecules in serum.
  • RNAi molecules were incubated in serum for two hours and analysed on a 10% polyacrylamide gel
  • Figs. 11A to 11C show the results of this study.
  • Fig. 11A shows the dose response of the various RNAi molecules depicted in Fig. 1 IC
  • Fig. 1 IB shows the result of a stability test using some of the RNAi molecules depicted in Fig. llC.
  • nucleotides written in bold in Fig. 11C are the ones carrying a modification which is in the present case of a 2 '-O-methyl modification ofthe ribose moiety ofthe nucleotides.
  • RNAi molecules There is a dose dependent inhibition by the unmodified RNAi molecules. It is also shown that the 2 '-O-methyl modification of the core 9 nucleotides makes the RNAi stable in serum and allows activity of the duplex in the meaning of mediating the interference phenomenon leading to a degradation of the PTEN mRNA. Total modification of the sense strand makes the RNAi molecule stable in serum and allows certain activity.
  • Alternating blocks of 5 nucleotides with 2 '-O-methyl modification renders the RNAi molecule stable in serum and allows activity on PTEN RNA as shown by incubating the RNAi duplex in serum for two hours and loading the samples on a 10% polyacrylamide gel.
  • the duplex comprising strands 80A and 80B is strongly degraded after incubation in serum for two hours.
  • the duplex consisting of strands 82A and 82B confirms the result that the 5 '-end of the first strand which comprises the antisense strand should not be modified at the 5 '-terminal nucleoties (compare 82A82B with the reverse orientated 81A81B).
  • Example 9 The impact of alternating internal 2'-O-alkyI modification on serum stability of RNAi molecules.
  • RNAi molecules depicted in Fig. 12B and RNA knockdown was demonstrated using real time RT-PCR on PTEN RNA in a dose-dependent manner (Fig. 12A).
  • Fig. 12C The stability of the various RNAi molecules after 15 min and two hours in serum at 37°C is depicted in Fig. 12C and a Western Blot for pi 10 and PTEN as the target-protein ofthe various RNAi molecules is depicted in Fig. 12D with the RNAi molecules tested being the same in both the experiments underlying Fig. 12C and Fig. 12D.
  • nucleotides modified with 2 '-O-methyl groups alternating with unmodified nucleotides render RNAi molecules stable in serum while still allowing them to be active in the sense of interfering with the target mRNA. It is shown that incubation of RNAi duplex molecules for 15 min and two hours in serum will degrade the unmodified duplex and the duplex where the 10 most 5 '-positioned nucleotides are unmodified.
  • RNAi molecules represented in Fig. 12B various patterns of modified and unmodified nucleotides are realised.
  • the RNAi molecule 94A1/94B1 comprises a structure wherein a modified nucleotide is flanked by an unmodified nucleotide with the unmodified nucleotide being located at the 5' end of the first strand.
  • the RNAi molecule comprised of strands 94A2 and 94B2 is another example where the modified nucleotides and the unmodified nucleotides of the first and the second strand are located at opposing sites.
  • the RNAi molecule comprised of strands 94A1 and 94B2 have the same pattern of modified and unmodified nucleotides.
  • the two RNAi molecules comprised of strands 94A1 and 94B1 and strands 94A2 and 94B2 differ from each other such that in the first case the first strand starts with an unmodified nucleotide and the corresponding first nucleotide of the second strand, i. e. the nucleotide at the 3' end ofthe second strand, starts with an unmodified nucleotide with the arrangement being opposite to this in the RNAi molecule comprised of 94A2 and 94B2.
  • alternatingly modified RNAi molecules as depicted in Fig. 12B are functional in mediating a PTEN protein knock down as shown in Fig. 12D but only when the second 5' and second 3' terminal nucleotide is not modified (see 94A294B1 and 94A294B2).
  • 94A294B1 and 94A294B2 Taken together these data show that the most stable and most active RNAi molecules do have alternating 2'Alkyl modified and unmodified nucleotide residues. It should be noted that these molecules do show a very similar mRNA reduction when compared to unmodified siRNA molecules which being stable in serum allows increased or easier handling.
  • Example 10 Functional Protein Knockdown mediated by internally modified RNAi molecules
  • RNAi were transfected by adding pre-formed 5x concentrated complex of siRNAs and lipid in serum-free medium to cells in complete medium.
  • the total transfection volume was 100 ⁇ l for cells plated in 96-wells and 10 ml for cells in 10 cm plates.
  • the final lipid concentration was 0.8 to 1.2 ⁇ g/ml depending on cell density; the siRNA concentration is indicated in each experiment.
  • modified RNAi molecules of the 94A2B1 and 94A2B2 version yield a longer lasting PTEN protein knock down as unmodified molecules.
  • the lack of protein knock down also seen in Fig. 12 with molecules of the 94A1B1 and 94A1B2 version is confirmed in this experiment.
  • Unmodified molecules (80AB) are not as potent in supporting long lasting protein knock down when the cells are not continuously transfected.
  • the experimental approach was similar to the one outlined in connection with example 10 with the exception that the transfection was terminated after 5 h by replacing the transfection medium with new medium.
  • the protocol was slightly modified such that for each of the RNAi molecules a 40 nM concentration was realized using a stock solution of 1 ⁇ g RNAi / ml cationic lipid as described in connection with example 1. 5 hours after transfection the medium was withdrawn and fresh EMEM added. The cells were split after 72, with half of the cells being lysed and the other half newly plated and lysed 24 h later (96h post transfection).
  • the result of a Western Blot analysis using 3 different RNAi molecules 80AB, 94A1/B2, 94A2/B1 are depicted in Fig. 14.
  • RNAi molecules 94A1B2 As a positive control untreated cells were used.
  • Fig. 14 shows the expression of PTEN after 72 h and 96 h, respectively. Taken the structural particularities of the various RNAi molecules it can be taken from Fig. 14 that protein knockdown is persistent with alternating molecules of the 94A2B1 kind even over 96 h after splitting and replating cells compared to unmodified RNAi molecules (such as 80AB) and RNAi molecule 94A1B2.
  • Fig. 15A A further experiment was carried out using the siRNA constructs as depicted in Fig. 15A (left panel). From the results as depicted as ratio of PTEN/pl lO ⁇ mRNA degradation at the various concentrations of siRNA constructs administered to the test system, it can be taken that siRNA molecules with either one or both strands consisting of 2 '-O-methyl residues were not able to induce RNA interference in the mammalian system (Fig. 15 A, molecules V2, V5, V6). However, the decrease in activity was less pronounced when only parts of the strands were modified.
  • Fig. 15B (right panel with the various sequences indicated on the left panel of Fig. 15B).
  • Fig. 15B compare molecule AB with VI.
  • siRNA molecules with partial 2'-O-methyl modification showed as well an increased stability when compared to unmodified siRNAs. Especially molecules with alternating modifications on both strands showed a significant improve in instability (Fig.
  • RNA interference activity assay an unexpected preference for molecules was observed which were modified at every second nucleotide beginning with the most 5' terminal nucleotide ofthe antisense strand (molecules V15 and V12). Molecules which contained modifications beginning with the second nucleotide at the 5' end of the antisense strand were more stable but had a strongly reduced activity in gene silencing (molecules VI 3, V14).
  • Aktl As a target an efficient protein knock-down as well as a dramatic reduction in phospho-Akt levels was observed with blunt, unmodified siRNAs (Fig. 16A, right panel). From the different versions of molecules with modifications on every second other nucleotide only one was efficiently mediating RNAi (Fig. 16 A, molecule V5).
  • This siRNA molecule contained an antisense strand which was modified at the most terminal 5' and 3' nucleotides. The sense strand started with the unmodified nucleotides at the most terminal position, resulting in a structure in which the modified and unmodified ribonucleotides of both strands are facing each other. As expected, this molecule was also protected against serum-derived nucleases as depicted in Fig. 16B (molecule V5).
  • the reduced activity of the molecules N6 might be explained by reduced duplex stability since the same structure was active in the PTE ⁇ knockdown experiment with 2 Imer siRNAs. Although it is known that 2 '-O-methyl modification on both strands facing each other will reduce the stability of nucleic acid duplexes, the difference between the activity of siR ⁇ A molecules V4 and V5 (Figs. 16B and C) is probably not due to differences in duplex stability since the number of base pairing of modified and unmodified nucleotides is identical. This difference in activity might be due to specific requirements in the interacting proteins involved in the degradation of the target mR ⁇ A. Also it can be taken from these experiments that the most terminal nucleotides of the antisense strand can be modified at the 2 '-OH-group with significant loss of silencing activity.
  • Example 12 Different loop structures are functional in mediating R ⁇ A interference
  • R ⁇ Ai molecules preferably synthetic R ⁇ Ai molecules with self complementary structures can inhibit gene expression as efficiently as standard double stranded siR ⁇ A molecules
  • HeLa cells were transfected with pl lO ⁇ specific synthetic siRNAs.
  • Transfections were carried out in 96 well or 10-cm plates (at 30% to 50% confluency) by using various cationic lipids such as Oligofectamine, Lipofectamine (Life Technologies), GeneBlocs were transfected by adding pre-formed 5x concentrated complex of GB and lipid in serum-free medium to cells in complete medium.
  • the total transfection volume was 100 ⁇ l for cells plated in 96-wells and 10 ml for cells in 10 cm plates.
  • the final lipid concentration was 0.8 to 1.2 ⁇ g/ml depending on cell density; the RNAi molecule concentration is indicated in each experiment.
  • a dose dependent titration showed no significant difference in efficiency of mRNA knock-down achieved by the standard bimolecular double strand 2 Imer and the corresponding monomolecular molecules as analysed by realtime PCR (Taqman) (Fig. 17A).
  • Two different loop structures a (A) ⁇ 2 loop and a HTN derived pA-loop were tested in parallel with similar results.
  • a comparision of the relative position of the antisense sequence and the loop structure revealed an improved knock-down efficiency with the antisense sequence being located 3 ' to the loop (Fig. 17B; compare construct 3 A, 3B and 4A, 4B).
  • Example 13 Studies on the efficacy of different intramolecular hairpin loops and intermolecular "bubbles"
  • Akt2-specific sequences within the same molecule (Fig. 18B, constructs 10 A, 10B) and were designed to be restricted to form a bimolecular duplex ("bubble").
  • this molecule efficiently mediated both Aktl and Akt2 mRNA knock-down as well as protein knock-down when transfected after annealing of both strands.

Abstract

The present invention is related to a ribonucleic acid comprising a double stranded structure whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid, and whereby the double stranded structure is blunt ended.

Description

Further novel forms of interfering RNA molecules
The present invention is related to a ribonucleic acid comprising a double-stranded structure whereby the double-stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to the target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid, the use of such ribonucleic acid, a cell and an organism, respectively, comprising such ribonucleic acid, a composition containing such ribonucleic acid, a pharmaceutical composition containing such ribonucleic acid and a method for inhibiting expression of a targeted gene.
RNA-mediated interference (RNAi) is a post-transcriptional gene silencing mechanism initiated by double stranded RNA (dsRNA) homologous in sequence to the silenced gene (Fire (1999), Trends Genet 15, 358-63, Tuschl, et al. (1999), Genes Dev 13, 3191-7, , Waterhouse, et al. (2001), Nature 411, 834-42, Elbashir, et al. (2001), Nature 411, 494-8, for review see Sharp (2001), Genes Dev 15, 485-90, Barstead (2001), Curr Opin Chem Biol 5, 63-6). RNAi has been used extensively to determine gene function in a number of organisms, including plants (Baulcombe (1999), Curr Opin Plant Biol 2, 109-13), nematodes (Montgomery, et al. (1998), Proc Natl Acad Sci U S A 95, 15502-7), Drosophila (Kennerdell, et al. (1998), Cell 95, 1017-26, Kennerdell, et al. (2000), Nat Biotechnol 18, 896-8). In the nematode C. elegans about one third ofthe genome has already been subjected to functional analysis by RNAi (Kim (2001), Curr Biol 11, R85-7, Maeda, et al. (2001), Curr Biol 11, 171-6).
Until recently RNAi in mammalian cells was not generally applicable, with the exception of early mouse development (Wianny, et al. (2000), Nat Cell Biol 2, 70-5). The discovery that transfection of duplexes of 21-nt into mammalian cells interfered with gene expression and did not induce a sequence independent interferon-driven anti-viral response usually obtained with long dsRNA led to new potential application in differentiated mammalian cells (Elbashir et al. (2001), Nature 411, 494-8). Interestingly these small interfering RNAs (siRNAs ) resemble the processing products from long dsRNAs suggesting a potential bypassing mechanism in differentiated mammalian cells. The Dicer complex, a member of the RNAse III family, necessary for the initial dsRNA processing has been identified (Bernstein, et al. (2001), Nature 409, 363-6, Billy, et al. (2001), Proc Natl Acad Sci U S A 98, 14428-33). One of the problems previously encountered when using unmodified ribooligonucleotides was the rapid degradation in cells or even in the serum-containing medium (Wickstrom (1986), J Biochem Biophys Methods 13, 97-102, Cazenave, et al. (1987), Nucleic Acids Res 15, 10507-21). It will depend on the particular gene function and assay systems used whether the respective knock down induced by transfected siRNA will be maintained long enough to achieve a phenotypic change.
The problem underlying the present invention was to provide synthetic interfering RNA molecules which are both stable and active in a biochemical environment such as a living cell.
In a first aspect ofthe present invention the problem is solved by a ribonucleic acid comprising a double stranded structure whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid, and whereby the double stranded structure is blunt ended.
In a second aspect the problem underlying the present invention is solved by a ribonucleic acid comprising a double stranded structure whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides, whereby said second stretch is at least partially identical to a target nucleic acid, whereby the first stretch and/or the second stretch have a length of 18 or 19 nucleotides.
In an embodiment of the ribonucleic acid according to the first aspect of the invention the first stretch and/or the second stretch have a length of 18 or 19 nucleotides.
In a further embodiment of the ribonucleic acid according to the first aspect ofthe invention the double stranded structure is blunt ended on both sides ofthe double strand. In an alternative embodiment ofthe ribonucleic acid according to the first aspect ofthe invention the double stranded structure is blunt ended on the double stranded structure which is defined by the 5'-end ofthe first strand and the 3 '-end ofthe second strand.
In a further alternative embodiment of the ribonucleic acid according to the first and the second aspect of the invention the double stranded structure is blunt ended on the double stranded structure which is defined by the 3 '-end ofthe first strand and the 5 '-end ofthe second strand.
In a third aspect the problem underlying the present invention is solved by a ribonucleic acid comprising a double stranded structure whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides and whereby said second stretch is at least partially identical to a target nucleic acid, and whereby at least one ofthe two strands has an overhang of at least one nucleotide at the 5 '-end.
In an embodiment of the ribonucleic acid according to the third aspect of the present invention the overhang consists of at least one nucleotide which is selected from the group comprising ribonucleotides and deoxyribonucleotides.
In a more preferred embodiment of the ribonucleic acid according to the third aspect of the present invention the nucleotide has a modification whereby said modification is preferably selected from the group comprising nucleotides being an inverted abasic and nucleotides having an NH2-modification at the 2 '-position.
In a preferred embodiment of the ribonucleic acid according to the third aspect of the present invention at least one of the strands has an overhang of at least one nucleotide at the 3 '-end consisting of ribonucleotide or deoxyribonucleotide.
In another preferred embodiment of the ribonucleic acid according to the third aspect of the present invention the first stretch and/or the second stretch have a length of 18 or 19 nucleotides.
In an embodiment of the ribonucleic acid according to any aspect of the present invention the double-stranded structure has a length of 17 to 21 nucleotides, preferably 18 to 19 nucleotides. In an embodiment of the ribonucleic acid according to the third aspect of the present invention the overhang at the 5 '-end is on the second strand.
In a preferred embodiment of the ribonucleic acid according to the third aspect of the present invention the first strand comprises also an overhang, preferably at the 5 '-end.
In an embodiment of the ribonucleic acid according to the third aspect of the present invention the 3 '-end ofthe first strand comprises an overhang.
In an alternative embodiment ofthe ribonucleic acid according to the third aspect of the present invention the overhang at the 5 '-end is on the first strand.
In a preferred embodiment thereof the second strand also comprises an overhang, preferably at the 5 '-end.
In an embodiment of the ribonucleic acid according to the third aspect of the present invention the 3 '-end ofthe first strand comprises an overhang.
In an embodiment ofthe ribonucleic acid according to any aspect ofthe present invention at least one nucleotide of the ribonucleic acid has a modification at the 2'-position and the modification is preferably selected from the group comprising amino, fluoro, methoxy, alkoxy and alkyl.
In a fourth aspect the problem underlying the present invention is solved by a ribonucleic acid comprising a double stranded structure, whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides and whereby said second stretch is at least partially identical to a target nucleic acid,
whereby
said first strand and/or said second strand comprises a plurality of groups of modified nucleotides having a modification at the 2'-position whereby within the strand each group of modified nucleotides is flanked on one or both sides by a flanking group of nucleotides whereby the flanking nucleotides forming the flanking group of nucleotides is either an unmodified nucleotide or a nucleotide having a modification different from the modification of the modified nucleotides.
In an embodiment of the ribonucleic acid according to the fourth aspect of the present invention the ribonucleic acid is the ribonucleic acid according to the first, second or third aspect of the present invention.
In a further embodiment of the ribonucleic acid according to the fourth aspect of the present invention said first strand and/or said second strand comprise said plurality of modified nucleotides.
In another embodiment of the ribonucleic acid according to the fourth aspect of the present invention said first strand comprises said plurality of groups of modified nucleotides.
In yet another embodiment of the ribonucleic acid according to the fourth aspect of the present invention said second strand comprises said plurality of groups of modified nucleotides.
In a preferred embodiment of the ribonucleic acid according to the fourth aspect of the present invention the group of modified nucleotides and or the group of flanking nucleotides comprises a number of nucleotides whereby the number is selected from the group comprising one nucleotide to 10 nucleotides. In connection with any ranges specified herein it is to be understood that each range discloses any individual integer between the respective figures used to define the range including said two figures defining said range. In the present case the group thus comprises one nucleotide, two nucleotides, three nucleotides, four nucleotides, five nucleotides, six nucleotides, seven nucleotides, eight nucleotides, nine nucleotides and ten nucleotides.
In another embodiment of the ribonucleic acid according to the fourth aspect of the present invention the pattern of modified nucleotides of said first strand is the same as the pattern of modified nucleotides of said second strand.
In a preferred embodiment of the ribonucleic acid according to the fourth aspect of the present invention the pattern of said first strand aligns with the pattern of said second strand. In an alternative embodiment ofthe ribonucleic acid according to the fourth aspect ofthe present invention the pattern of said first strand is shifted by one or more nucleotides relative to the pattern ofthe second strand.
In an embodiment of the ribonucleic acid according to the fourth aspect of the present invention the modification is selected from the group comprising amino, fluoro, methoxy, alkoxy and alkyl.
In another embodiment of the ribonucleic acid according to the fourth aspect of the present invention the double stranded structure is blunt ended.
In a preferred embodiment of the ribonucleic acid according to the fourth aspect of the present invention the double stranded structure is blunt ended on both sides.
In another embodiment of the ribonucleic acid according to the fourth aspect of the present invention the double stranded structure is blunt ended on the double stranded structure's side which is defined by the 5 '-end ofthe first strand and the 3 '-end ofthe second strand.
In still another embodiment of the ribonucleic acid according to the fourth aspect of the present invention the double stranded structure is blunt ended on the double stranded structure's side which is defined by at the 3 '-end ofthe first strand and the 5'-end ofthe second strand.
In another embodiment of the ribonucleic acid according to the fourth aspect of the present invention at least one ofthe two strands has an overhang of at least one nucleotide at the 5'-end.
In a preferred embodiment of the ribonucleic acid according to the fourth aspect of the present invention the overhang consists of at least one deoxyribonucleotide.
In a further embodiment of the ribonucleic acid according to the fourth aspect of the present invention at least one ofthe strands has an overhang of at least one nucleotide at the 3 '-end. In an embodiment ofthe ribonucleic acid according to any ofthe aspects ofthe present invention the length of the double-stranded structure is from about 17 to 21 and more preferably 18 or 19 bases
In another embodiment of the ribonucleic acid according to any of the aspects of the present invention the length of said first strand andor the length of said second strand is independently from each other selected from the group comprising the ranges of from about 15 to about 23 bases, 17 to 21 bases and 18 or 19 bases.
In a preferred embodiment ofthe ribonucleic acid according to any ofthe aspects ofthe present invention the complementarity between said first strand and the target nucleic acid is perfect.
In an embodiment ofthe ribonucleic acid according to any ofthe aspects ofthe present invention the duplex formed between the first strand and the target nucleic acid comprises at least 15 nucleotides wherein there is one mismatch or two mismatches between said first strand and the target nucleic acid forming said double-stranded structure.
In an embodiment ofthe ribonucleic acid according to any ofthe aspects ofthe present invention both the first strand and the second strand each comprise at least one group of modified nucleotides and at least one flanking group of nucleotides, whereby each group of modified nucleotides comprises at least one nucleotide and whereby each flanking group of nucleotides comprising at least one nucleotide with each group of modified nucleotides of the first strand being aligned with a flanking group of nucleotides on the second strand, whereby the most terminal 5' nucleotide ofthe first strand is a nucleotide ofthe group of modified nucleotides, and the most terminal 3' nucleotide of the second strand is a nucleotide of the flanking group of nucleotides.
In a preferred embodiment of the ribonucleic acid according to the fourth aspect, each group of modified nucleotides consists of a single nucleotide and/or each flanking group of nucleotides consists of a single nucleotide.
Tji a further embodiment ofthe ribonucleic acid according to the fourth aspect, on the first strand the nucleotide forming the flanking group of nucleotides is an unmodified nucleotide which is arranged in a 3' direction relative to the nucleotide forming the group of modified nucleotides, and wherein on the second strand the nucleotide forming the group of modified nucleotides is a modified nucleotide which is arranged in 5' direction relative to the nucleotide forming the flanking group of nucleotides.
In a another embodiment of the ribonucleic acid according to the fourth aspect, the first strand comprises eight to twelve, preferably nine to eleven, groups of modified nucleotides, and wherein the second strand comprises seven to eleven, preferably eight to ten, groups of modified nucleotides.
In a preferred embodiment of the ribonucleic acid according to any of the aspects of the present invention the target gene is selected from the group comprising structural genes, housekeeping genes, transcription factors, motility factors, cell cycle factors, cell cycle inhibitors, enzymes, growth factors, cytokines and tumor suppressors.
In a further embodiment of the ribonucleic acid according to any of the aspects of the present invention the first strand and the second strand are linked by a loop structure.
In a preferred embodiment of the ribonucleic acid according to any of the aspects of the present invention the loop structure is comprised of a non-nucleic acid polymer.
In a preferred embodiment thereof the non-nucleic acid polymer is polyethylene glycol.
In an alternative embodiment thereof the loop structure is comprised of a nucleic acid.
In an embodiment ofthe ribonucleic acid according to any ofthe aspects ofthe present invention the 5'-terminus ofthe first strand is linked to the 3'-terminus ofthe second strand.
In a further embodiment of the ribonucleic acid according to any of the aspects of the present invention the 3 '-end ofthe first strand is linked to the 5 '-terminus ofthe second strand.
In a fifth aspect the problem underlying the present invention is solved by the use of a ribonucleic acid according to any ofthe aspects ofthe present invention, for target validation. In a sixth aspect the problem underlying the present invention is solved by the use of a ribonucleic acid according to any ofthe aspects ofthe present invention, for the manufacture of a medicament.
In a preferred embodiment of the use according to the sixth aspect of the present invention the medicament is for the treatment of a disease or of a condition which is selected from the group comprising glioblastoma, prostate cancer, breast cancer, lung cancer, liver cancer, colon cancer, pancreatic cancer and leukaemia, diabetes, obesity, cardiovascular diseases, and metabolic diseases.
In a seventh aspect the problem underlying the present invention is solved by a cell, preferably a knockdown cell, containing a ribonucleic acid according to any of the aspects of the present invention.
fn an eighth aspect the problem underlying the present invention is solved by an organism, preferably a knockdown organism, containing a ribonucleic acid according to any of the aspects ofthe present invention.
In a ninth aspect the problem underlying the present invention is solved by a composition containing a ribonucleic acid according to any ofthe aspects ofthe present invention.
In a tenth aspect the problem underlying the present invention is solved by a pharmaceutical composition containing a ribonucleic acid according to any of the aspects of the present invention, and a pharmaceutically acceptable carrier.
In an eleventh aspect the problem underlying the present invention is solved by a method for inhibiting the expression of a target gene in a cell or derivative thereof comprising introducing of a ribonucleic acid according to any of the aspects of the present invention into the cell in an amount sufficient to inhibit expression of the target gene, wherein the target gene is the target gene of a ribonucleic acid according to any ofthe aspects ofthe present invention.
The present invention is based on the surprising finding that small interfering RNAs can be designed such as to be both highly specific and active as well as stable under the reaction conditions typically encountered in biological systems such as biochemical assays or cellular environments. The various interfering RNAs described in the prior art such as by Tuschl et al. (international patent application WO 01/75164) provide for a length of 21 to 23 nucleotides and a modification at the 3' end of the double-stranded RNA. It has been surprisingly found by the present inventors that the problem of stability of interfering RNA, including small interfering RNA (siRNA) which is generally referred to herein in the following as RNAi, actually resides in the attack of endonucleases rather than exonucleases as thought earlier. Based on this finding several strategies have been perceived by the present inventors which are subject to the present application.
The present invention is thus related to new forms of interfering RNA. RNAi consists of a ribonucleic acid comprising a double-stranded structure. Said double-stranded structure is formed by a first strand and a second strand. Said first strand comprises a stretch of contiguous nucleotides, also referred to as first stretch of contiguous nucleotides herein, and this first stretch is at least partially complementary to a target nucleic acid. Said second strand comprises also a stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid. The very basic structure of this ribonucleic acid is schematically shown in Fig. 1. Said first strand and said second strand are preferably hybridised to each other and form the double-stranded structure. The hybridisation typically occurs by Watson Crick base pairing. The inventive ribonucleic acid, however, is not necessarily limited in its length to said double- stranded structure. There might be further nucleotides added to each strand and/or to each end of any of the strands forming the RNAi. Depending on the particular sequence of the first stretch and the second stretch, the hybridisation or base pairing is not necessarily complete or perfect, which means that the first and the second stretch are not 100 % base paired due to mismatches. There might also be one or more mismatches within the duplex. Said mismatches have no effect on the RNAi activity if placed outside a stretch of preferably 15, 16 or 17 matching nucleotides. If mismatches are placed to yield only 15 or less contiguous matching nucleotides, the RNAi molecule typically shows a reduced activity in down regulating mRNA for a given target compared to a 17 matching nucleotide duplex.
The first stretch of contiguous nucleotides of the first strand is essentially complementary to a target nucleic acid, more preferably to a part of the target nucleic acid. Complementary as used herein preferably means that the nucleotide sequence ofthe first strand is hybridising to a nucleic acid sequence of a target nucleic acid sequence or a part thereof. Typically, the target nucleic acid sequence or target nucleic acid is, in accordance with the mode of action of interfering ribonucleic acids, a single stranded RNA, more preferably an mRNA. Such hybridisation occurs most likely through Watson Crick base pairing, however, is not necessarily limited thereto. The extent to which said first strand and more particularly the first stretch of contiguous nucleotides of said first strand is complementary to a target nucleic acid sequence can be as high as 100% and be as little as 80%, preferably 80-100%, more preferably 85-100%, most preferably 90-
100%. Optimum complementarity seems to be 95-100%. Complementarity in this sense means that the aforementioned range of nucleotides, such as, e. g., 80%-100%, depending on the particular range, of the nucleotides are perfect by Watson Crick base pairing. It is shown in one aspect ofthe present invention that the complementarity between said first stretch of nucleotides and the target RNA has to be 18-19 nucleotides, stretches of as little as 17 nucleotides even with two sequence specific overhangs are not functional in mediating RNAi. Accordingly, given a duplex having a length of 19 nucleotides or base pairs a minimum complementarity of 17 nucleotides or nucleotide base pairs would be acceptable allowing for a mismatch of two nucleotides. In case of a duplex consisting of 20 nucleotides or base pairs a complementarity of
17 nucleotides or nucleotide base pairs would be allowable and functionally active. The same applies to a duplex of 21 nucleotides or base pairs with a total of 17 complementary nucleotides or base pairs. Basically, the extent of complementarity required for a length of a duplex, i. e. of a double stranded structure, can also be based on the melting temperature of the complex formed by either the double stranded structure as described herein or by the complex of the first stretch ofthe first strand and the target nucleic acid.
It is to be understood that all of the ribonucleic acids of the present invention are suitable to cause or being involved in RNA mediated interference such as, for example, described in international patent applications WO 99/32619, WO 00/44895 and WO 01/75164.
The first strategy according to which an interfering ribonucleic acid molecule may be designed according to the present invention is to have an optimum length of 18 or 19 nucleotides of the stretch which is complementary to the target nucleic acid. It is also within the present invention that said optimum length of 18 or 19 nucleotides is the length ofthe double stranded structure in the RNAi used. This length requirement is clearly different from the technical teaching of the prior art such as, for example, international patent application WO 01/75164. It is within the present invention that any further design, both according to the present invention and as described in the prior art, can be realised in connection with an interfering ribonucleic acid having said length characteristics, i.e. a length of 18 or 19 nucleotides. The second strategy according to which an interfering ribonucleic acid molecule may be designed is to have a free 5' hydroxyl group, also referred to herein as free 5' OH-group, at the first strand. A free 5' OH-group means that the most terminal nucleotide forming the first strand is present and is thus not modified, particularly not by an end modification. Typically, the terminal 5 '-hydroxy group of the second strand, respectively, is also present in an unmodified manner. In a more preferred embodiment, also the 3 '-end of the first strand and first stretch, respectively, is unmodified such as to present a free OH-group which is also referred to herein as free 3 'OH-group, whereby the design of the 5' terminal nucleotide is the one of any ofthe afore- described embodiments. Preferably such free OH-group is also present at the 3 '-end of the second strand and second stretch, respectively. In other embodiments of the ribonucleic acid molecules as described previously according to the present invention the 3 '-end of the first strand and first stretch, respectively, and/or the 3 '-end of the second strand and second stretch, respectively, may have an end modification at the 3' end.
As used herein the terms free 5'OH-group and 3'OH-group also indicate that the respective most terminal nucleotide at the 5 'end and the 3 ' end of the polynucleotide, respectively, presents an OH-group. Such OH-group may stem from either the sugar moiety of the nucleotide, more preferably from the 5 'position in case of the 5'OH-group and from the 3 'position in case of the 3'OH-group, or from a phosphate group attached to the sugar moiety of the respective terminal nucleotide. The phosphate group may in principle be attached to any OH-group of the sugar moiety of the nucleotide. Preferably, the phosphate group is attached to the 5'OH-group of the sugar moiety in case of the free 5'OH-group and/or to the 3'OH-group of the sugar moiety in case ofthe free 3'OH-group still providing what is referred to herein as free 5' or 3' OH-group.
As used herein with any strategy for the design of RNAi or any embodiment of RNAi disclosed herein, the term end modification means a chemical entity added to the most 5' or 3' nucleotide of the first and/or second strand. Examples for such end modifications include, but are not limited to, inverted (deoxy) abasics, amino, fluoro, chloro, bromo, CN, CF, methoxy, imidazole, caboxylate, thioate, d to Cio lower alkyl, substituted lower alkyl, alkaryl or aralkyl, OCF3, OCN, O-, S-, or N-alkyl; O-, S-, or N-alkenyl; SOCH3; SO2CH3; ONO2; NO2, N3; heterozycloalkyl; heterozycloalkaryl; aminoalkylamino; polyalkylamino or substituted silyl, as, among others, described in European patents EP 0 586 520 BI or EP 0 618 925 BI. As used herein, alkyl or any term comprising "alkyl" means any carbon atom chain comprising 1 to 12, preferably 1 to 6 and more, preferably 1 to 2 C atoms.
A further end modification is a biotin group. Such biotin group may preferably be attached to either the most 5' or the most 3' nucleotide of the first and/or second strand or to both ends. In a more preferred embodiment the biotin group is coupled to a polypeptide or a protein. It is also within the scope ofthe present invention that the polypeptide or protein is attached through any of the other aforementioned end modifications. The polypeptide or protein may confer further characteristics to the inventive nucleic acid molecules. Among others the polypeptide or protein may act as a ligand to another molecule. If said other molecule is a receptor the receptor's function and activity may be activated by the binding ligand. The receptor may show an internalization activity which allows an effective transfection of the ligand bound inventive nucleic acid molecules. An example for the ligand to be coupled to the inventive nucleic acid molecule is VEGF and the corresponding receptor is the NEGF receptor.
Various possible embodiments of the RΝAi of the present invention having different kinds of end modification(s) are presented in the following table 1.
Table 1: Various embodiments o the interfering ribonucleic acid according to the present invention
Figure imgf000016_0001
The various end modifications as disclosed herein are preferably located at the ribose moiety of a nucleotide ofthe ribonucleic acid. More particularly, the end modification may be attached to or replace any of the OH-groups of the ribose moiety, including but not limited to the 2'OH, 3'OH and 5'OH position, provided that the nucleotide thus modified is a terminal nucleotide. Inverted abasics are nucleotides, either desoxyribonucleotides or ribonucleotides which do not have a nucleobase moiety. This kind of compound is, among others, described in Sternberger et al.(2002), Antisense. Nucl. Ac. Drug Dev. in press.
Any of the aforementioned end modifications may be used in connection with the various embodiments of RNAi depicted in table 1. In connection therewith it is to be noted that any of the RNAi forms or embodiments disclosed herein with the sense strand being inactivated, preferably by having an end modification more preferably at the 5' end, are particularly advantageous. This arises from the inactivation of the sense strand which corresponds to the second strand of the ribonucleic acids described herein, which might otherwise interfere with an unrelated single-stranded RNA in the cell. Thus the expression and more particularly the translation pattern of the transcriptome of a cell is more specifically influenced. This effect is also referred to as off-target effect. Referring to table 1 those embodiments depicted as embodiments 7 and 8 are particularly advantageous in the above sense as the modification results in an inactivation of the - target unspecific - part of the RNAi (which is the second strand) thus reducing any unspecific interaction of the second strand with single-stranded RNA in a cellular or similar system where the RNAi according to the present invention is going to be used to knock down specific ribonucleic acids and proteins, respectively.
A third strategy subject to the present invention is to realise a ribonucleic acid comprising a double-stranded structure whereby the double-stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid whereby the double-stranded structure is blunt-ended. As used in connection with the present description the term double-stranded structure is also referred to as duplex. This design of RNAi is thus clearly different from, e. g., the one of Tuschl et al. as specified in international patent application WO 01/75164 which discloses a 3 '-overhang. As used herein overhang refers to a double-stranded structure whereby at least one end of one strand is longer than the corresponding end of the other strand forming the double-stranded structure, which is also referred to herein as the counter strand. Preferably, the first stretch is identical to the first strand and the second stretch is identical to the second strand.
Taken the effectiveness of blunt-ended RNAi and the advantages of an end modification of either the first or the second strand or both, respectively, of the respective ribonucleic acid it is preferred to have a combination of both design principles. In other words, it is within the present invention to have blunt-ended RNAi carrying any end modification scheme as depicted in table 1.
The fourth strategy subject to the present invention is to have an overhang at the 5 '-end of the ribonucleic acid. More particularly, such overhang may in principle be present at either or both the first strand and second strand of the ribonucleic acid according to the present invention. The length of said overhang may be as little as one nucleotide and as long as 2 to 8 nucleotides, preferably 2, 4, 6 or 8 nucleotides. It is within the present invention that the 5' overhang may be located on the first strand and/or the second strand of the ribonucleic acid according to the present application. The nucleotide(s) forming the overhang may be (a) desoxyribonucleotide(s), (a) ribonucleotide(s) or a continuation thereof.
The overhang preferably comprises at least one desoxyribonucleotide, whereby said one desoxyribonucleotide is preferably the most 5 '-terminal one. It is within the present invention that the 3 '-end ofthe respective counter-strand ofthe inventive ribonucleic acid does not have an overhang, more preferably not a desoxyribonucleotide overhang. Here again, any of the inventive ribonucleic acids may comprise an end modification scheme as outlined in connection with table 1 and/or and end modification as outlined herein.
The fifth strategy in the design of interfering ribonucleic acids subject to the present application resides in the formation of a certain pattern of modified nucleotides on at least one ofthe strands and more particularly on one ofthe stretches of contiguous nucleotides ofthe ribonucleic acid(s) according to the present invention. The kind of modification of said nucleotides may be the same as discussed in connection with the other strategies for designing interfering RNA as disclosed herein and more particularly the kind of modification described herein for or as an end modification, such as, e.g., inverted abasics, methoxy, or amino and the like at the ribose moiety of at least one nucleotide forming the ribonucleotide acids according to the present application. It is to be noted that the modification of said nucleotides may be any form of modification described herein, more particularly the kind of modification as described herein as end modification with the particularity that the so-called end modification is not necessarily located at terminal nucleotides. Rather the modification is occurring at a non-terminal nucleotide. Under such conditions the modification is preferably attached to the ribose moiety of the - to be - modified nucleotide and even more preferably to the 2 'position ofthe ribose moiety.
It is also within the present invention that any ribonucleic acid designed according to this strategy may also have the features conferred to a ribonucleic acid according to the present application by any of the other design strategies disclosed herein. Accordingly, the interfering ribonucleic acid having a pattern of modified" nucleotides may have an end modification, an end modification scheme, may be blunt ended or may have a 5' overhang or any combination of two or more of these elements or characteristics.
Apart from the aforementioned modifications which may be presented either as end modifications or as modification pattern, the ribonucleic acid backbone as such may be further modified by forming different links between the nucleotides. Such different links are, among others, described in European patent EP 0 586 520 BI and European patent EP 0 618 925 BI. Of particular interest here are internal modification(s) ofthe ribonucleic acid backbone which have been shown to confer higher nuclease resistance of ribooligonucleotides. hi a preferred embodiment the modification of the modified nucleotide is a methoxylation of the 2 '-OH-group ofthe ribose moiety ofthe nucleotide.
In a preferred embodiment, both strands, and more particularly both the first stretch and the second stretch show this kind of modification of the nucleotides forming said strands and stretches, respectively. However, it is also within the present invention that either the first strand and first stretch, respectively, or the second strand and second stretch, respectively, show this particular pattern of modification of the nucleotides. As used herein, the term group of modified nucleotide or flanking group of nucleotide may comprise or represent as little nucleotides as one nucleotide, i. e. one or more nucleotides.
Taken the stretch of contiguous nucleotides a pattern of modification of the nucleotides forming the stretch may be realised such that a single nucleotide or group of nucleotides which are covalently linked to each other via standard phosphorodiester bonds or, at least partially, through phosphorothioate bonds, show such kind of modification. In case such nucleotide or group of nucleotides which is also referred to herein as group of modified nucleotides, is not forming the 5 '-end or 3 '-end of said stretch a nucleotide or group of nucleotides follows on both sides ofthe nucleotide which does not have the modification of the preceding nucleotide or group of nucleotides. It is to be noted that this kind of nucleotide or group of nucleotides, however, may have a different modification. This kind of nucleotide or group of nucleotides is also referred to herein as the flanking group of nucleotides. This sequence of modified nucleotide and group of modified nucleotides, respectively, and unmodified or differently modified nucleotide or group of unmodified or differently modified nucleotides may be repeated one or several times. Preferably, the sequence is repeated more than one time. For reason of clarity the pattern is discussed in more detail in the following, generally referring to a group of modified nucleotides or a group of unmodified nucleotides whereby each of said group may actually comprise as little as a single nucleotide. Unmodified nucleotide as used herein means either not having any ofthe afore-mentioned modifications at the nucleotide forming the respective nucleotide or group of nucleotides, or having a modification which is different from the one ofthe modified nucleotide and group of nucleotides, respectively.
It is also within the present invention that the modification of the unmodified nucleotide(s) wherein such unmodified nucleotide(s) is/are actually modified in a way different from the modification of the modified nucleotide(s), can be the same or even different for the various nucleotides forming said unmodified nucleotides or for the various flanking groups of nucleotides.
The pattern of modified and unmodified nucleotides may be such that the 5 '-terminal nucleotide of the strand or of the stretch starts with a modified group of nucleotides or starts with an unmodified group of nucleotides. However, in an alternative embodiment it is also possible that the 5 '-terminal nuleotide is formed by an unmodified group of nucleotides.
This kind of pattern may be realised either on the first stretch or the second stretch of the interfering RNA or on both. It has to be noted that a 5' phosphate on the target-complementary strand of the siRNA duplex is required for siRNA function, suggesting that cells check the authenticity of siRNAs through a free 5' OH (which can be phosphorylated) and allow only such bona fide siRNAs to direct target RNA destruction (Nykanen, et al. (2001), Cell 107, 309-21).
Preferably, the first stretch shows a kind of pattern of modified and unmodified groups of nucleotides, i. e. of grouρ(s) of modified nucleotides and flanking group(s) of nucleotides, whereas the second stretch does not show this kind of pattern. This may be useful insofar as the first stretch is actually the more important one for the target-specific degradation process underlying the interference phenomenon of RNA so that for specificity reasons the second stretch can be chemically modified so it is not functional in mediating RNA interference.
However, it is also within the present invention that both the first stretch and the second stretch have this kind of pattern. Preferably, the pattern of modification and non-modification is the same for both the first stretch and the second stretch. In a preferred embodiment the group of nucleotides forming the second stretch and corresponding to the modified group of nucleotides ofthe first stretch are also modified whereas the unmodified group of nucleotides of or forming the second stretch correspond to the unmodified group of nucleotides of or forming the first stretch. This possibility is schematically depicted in Fig. 2A. Another alternative is that there is a phase shift of the pattern of modification of the first stretch and first strand, respectively, relative to the pattern of modification of the second stretch and second strand, respectively. Preferably, the shift is such that the modified group of nucleotides of the first strand corresponds to the unmodified group of nucleotides of the second strand and vice versa. This possibility is shown in Fig. 2B. It is also within the present invention that the phase shift ofthe pattern of modification is not complete but overlapping as illustrated in Fig. 2C.
In a preferred embodiment the second nucleotide at the terminus of the strand and stretch, respectively, is an unmodified nucleotide or the beginning of group of unmodified nucleotides. Preferably, this unmodified nucleotide or unmodified group of nucleotides is located at the 5'- end of the first and second strand, respectively, and even more preferably ofthe first strand. In a further preferred embodiment the unmodified nucleotide or unmodified group of nucleotide is located at the 5 '-end of the first strand and first stretch, respectively. In a preferred embodiment the pattern consists of alternating single modified and unmodified nucleotides.
In a further preferred embodiment of this aspect of the present invention the interfering ribonucleic acid subject comprises two strands, whereby a 2'-O-methyl modified nucleotide and a non-modified nucleotide, preferably a nucleotide which is not 2'-O-methyl modified, are incorporated on both strands in an alternate manner which means that every second nucleotide is a 2'-O-methyl modified and a non-modified nucleotide, respectively. This means that on the first strand one 2'-O-methyl modified nucleotide is followed by a non-modified nucleotide which in turn is followed by 2'-O-methyl modified nucleotide and so on. The same sequence of 2'-O- methyl modification and non-modification exists on the second strand, whereby there is preferably a phase shift such that the 2'-O-methyl modified nucleotide on the first strand base pairs with a non-modified nucleotide(s) on the second strand and vice versa. This particular arrangement, i. e. base pairing of 2'-O-methyl modified and non-modified nucleotide(s) on both strands is particularly preferred in case of short interfering ribonucleic acids, i. e. short base paired double-stranded ribonucleic acids because it is assumed, although the present inventors do not wish to be bound by that theory, that a certain repulsion exists between two base-pairing 2'- O-methyl modified nucleotides which would destabilise such duplex, preferably short duplexes. About the particular arrangement, it is preferred if the antisense strand starts with a 2 '-O-methyl modified nucleotide at the 5' end whereby consequently the second nucleotide is non-modified, the third, fifth, seventh and so on nucleotides are thus again 2'-O-methyl modified whereas the second, fourth, sixth, eighth and the like nucleotides are non-modified nucleotides. Again, not wishing to be bound by any theory, it seems that a particular importance may be ascribed to the second, and optionally fourth, sixth, eighth and/or similar position(s) at the 5' terminal end ofthe antisense strand which should not comprise any modification, whereas the most 5' terminal nucleotide, i. e. the first 5' terminal nucleotide of the antisense strand may - exhibit such modification with any uneven positions such as first, optionally third, fifth and similar position(s) at the antisense strand may be modified. In further embodiments the modification and non-modification, respectively, of the modified and non-modified nucleotide(s), respectively, may be anyone as described herein.
Although not limited thereto, the double-stranded structure of the inventive ribonucleic acid, which is also referred to as duplex, is formed by the first strand and second strand, respectively, or the first and second stretch of contiguous nucleotides. The length of the first stretch and second stretch, respectively, is typically about 15 to about 23, preferably 17 to 21, and more preferably 18 or 19 bases. In connection with this it is to be noted that a length of less than 30 nucleotides, preferably less than 21 nucleotides does not cause any biological system which is basically capable of showing RNA interference and also interferon response, to develop an interferon response. The reason for this resides in the observation that a given cell is experiencing profound physiological changes when double-stranded RNA longer than 30 base pairs binds and activates the protein kinase PKR and 2',5'-oligoadenylate synthetase. Activated PKR stalls translation via phosphorylation of eIF2a, activated 2',5'-AS causes mRNA degradation. These effects are not desired in target validation and animal models because they overrule the effect of the target specific knockdown on the phenotype.
According to a sixth strategy in the design of interfering ribonucleic acids subject to the present application the ribonucleic acid comprises a double-stranded structure whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides whereby said second stretch is at least partially identical to a target nucleic acid whereby one terminus of the first strand and one terminus of the second strand are linked by a loop structure.
In an embodiment the loop structure is comprised of a non-nucleic acid polymer. Such non- nucleic acid polymer may be polyethylene glycol or similar polymers. The non-nucleic acid polymers may in principle be chosen from the group comprising polymers which do not comprise a polynucleotide and allow that the two strands to be linked may actually hybridize to each other. To allow for such hybridization the molecule or moiety of the molecule linking the two stretches hybridizing with each other, has to have a certain molecular structure or molecular flexibility to allow the bending of the molecule so as to allow that both stretches get in close contact and in a three-dimensional orientation which permits hybridization. Such molecule or moiety factually acts as a hinge. In principle any molecule which complies with this requirement may be used in connection with the present invention. Besides polyethylene glycol amino acid based molecules may be used. Such amino acid based molecules may be either homopolymers or hetereopolymers. A useful example is a homopolymer consisting of seven glycine residues which allows the generation of a hinge as required to bring the two stretches to hybridize in the close proximity as needed. This glycine based hinge is described, e. g., in Guan K. L. and Dixon J. E. (1991), Anal Biochem. 192, 262. In another embodiment the hinge may be formed by crown ethers known in the art.
In an alternative embodiment the loop is comprised of a nucleic acid. As used herein, LNA as described in Elayadi and Corey (2001)Curr Opin Investig Drugs. 2(4):558-61. Review; Orum and Wengel (2001) Curr Opin Mol Ther. 3(3):239-43; and PNA are regarded as nucleic acids and may also be used as loop forming polymers. Basically, the 5 '-terminus of the first strand may be linked to the 3 '-terminus of the second strand. As an alternative, the 3 '-end of the first strand may be linked to the 5 '-terminus of the second strand. The nucleotide sequence forming said loop structure is regarded as in general not being critical. However, the length of the nucleotide sequence forming such loop seems to be critical for sterical reasons. Accordingly, a minimum length of four nucleotides seems to be appropriate to form the required loop structure. In principle, the maximum number of nucleotides forming the hinge or the link between both stretches to be hybridized is not limited. However, the longer a polynucleotide is, the more likely secondary and tertiary structures are formed and thus the required orientation of the stretches affected. Preferably, a maximum number of nucleotides forming the hinge is about 12 nucleotides. It is within the disclosure of this application that any ofthe designs described above may be combined with the present sixth strategy, i. e. by linking the two strands covalently in a manner that back folding (loop) can occur through a loop structure or similar structure.
The present inventors have surprisingly found that if the loop is placed 3' ofthe antisense strand, i. e. the first strand of the ribonucleic acid(s) according to the present invention, the activities of this kind of RNAi are higher compared to the placement of the loop 5' of the antisense strand. Accordingly, the particular arrangement of the loop relative to the" antisense strand and sense strand, i. e. the first strand and the second strand, respectively, is crucial and is thus in contrast to the understanding as expressed in the prior art where the orientation is said to be of no relevance. However, this seems not true given the experimental results presented herein. The understanding as expressed in the prior art is based on the assumption that any RNAi, is subject to a processing during which non-loop linked RNAi is generated. However, if this was the case, the clearly observed increased activity of those structures having the loop placed 3' of the antisense could not be explained. Insofar a preferred arrangement in 5' — » 3' direction of this kind of small interfering RNAi is second strand - loop - first strand. The respective constructs may be incorporated into suitable vector systems. Preferably the vector comprises a promoter for the expression of RNAi. Preferably the respective promoter is pol III and more preferably the promoters are the U6, HI, 7SK promoter as described in Good et al. (1997) Gene Ther, 4, 45-54.
Because of the general applicability of the concept of interfering RNA and thus the knockdown or knockout for a coding nucleotide such as an mRNA any gene producing such RNA may be modified in its expression by using any ofthe ribonucleic acid molecule according to the present invention. Because of this basic and generally applicable mechanism any application based thereon can be realised which imply the knockdown or knockout of a gene. A preferred application is the use of the inventive ribonucleic acid for target validation. As used herein, target validation shall mean a process that involves taking steps to prove that a DNA, RNA, or protein molecule is directly involved in a biological process, preferably in a process - causally - involved in a disease or non-standard condition and is therefore a suitable target for development of a new therapeutic compound. Sequence homology studies have successfully classified genes into target families. The enormous task of deciphering which of these targets are key players in diseases and which should be used for subsequent drug development needs to be addressed in an efficient manner. Therefore, the knockdown of gene expression should be reduced by 50-100%o, preferably by 90% to see significant effects on the phenotype. In other cases depending on the gene, a knockdown of as little as 20% might be sufficient to yield a phenotype. A phenotype will be defined by comparison of cells containing functional RNAi molecules with cells containing non functional RNAi molecules. This will ensure a significant readout even under conditions where the protein function is inhibited only partially. Generally there is no linear correlation between the degree of rnRNA reduction and the extent of the change in phenotype. It has to be acknowledged that for some proteins a reduction of about 20 % of the protein is sufficient to create a change in the phenotype whereas in case of other genes and mRNA, respectively, as little as 5 to 10 % remaining protein is sufficient to maintain" the observed phenotype.
A further use of the ribonucleic acid molecules according to the present invention is its use for the manufacture of a medicament or its use as a medicament. Such medicament could either be used for the treatment and/or prevention of diseases or conditions such as any type of cancer where a gene and its product have been linked to the onset, cause or progression of this disease. Additionally, such medicament could be used to treat diseases where the presence or overexpression of a gene product is causing a pathological phenotype. In a preferred embodiment the disease is characterised by a gain of function and may be remedied through application or administration of the corresponding, biologically active RNAi. Diseases or conditions which may be treated by the medicament comprising a ribonucleic acid as disclosed herein, may be selected from the group comprising cancer, heart diseases, metabolic diseases, dermatological diseases, inflammatory diseases, immune system disorders and autoimmune disorders. The various forms of cancer include, but are not limited to, solid tumors and tumors of the hematopoietic system, such as glioblastoma, prostate cancer, breast cancer, lung cancer, liver cancer, pancreatic cancer and leukaemia. Metabolic diseases include, but are not limited to, obesity and diabetes. Dermatological diseases include, but are not limited to, psoriasis.
In another aspect the ribonucleic acid molecules according to the present invention may be used as diagnostics, preferably for those diseases as specified in connection with the above-mentioned diseases and conditions. Such diagnosis could be based on the observation that upon applying the ribonucleic acid molecules according to the present invention to a sample which preferably contains cells, a change in the expression pattern of the sample occurs. Preferably such sample comprises cells from a subject from whom it is assumed that it may exhibit said disease or condition to be treated or a predisposition therefor.
A further application of the nucleic acids according to the present invention resides in their use in the screening of pharmaceutically active compounds and/or lead optimization. The latter is done such as to monitor or determine the effect of candidate drugs such as small molecules and compare the effect created by said candidate drugs with the effect observed upon administering specific RNAi designed on the basis of the principles disclosed herein. In doing so candidate drugs having off-target effects may be eliminated from the screening process whereas those candidate drugs which create a similar or identical phenotype are deemed as highly relevant lead compound or may even be a pharmaceutically active compound themselves. In this approach, the highly specific RNAi molecules act as gold standard against which candidate drugs are measured.
In a further aspect the invention is related to a cell, preferably a knockdown cell, which_ contains a ribonucleic acid as disclosed herein. Such cell is preferably a cell which is either isolated or contained in a tissue or even organ which again preferably is not contained in an organism. However, the cell may also be contained in an organism. The cell is preferably a cell which is involved in the disease or condition which is to be treated by the inventive ribonucleic acids. This kind of knock-down cells may be used to generate an expression profile based on, e. g., mRNA or protein, in order to elucidate functional relationship and to determine downstream targets.
In a further aspect the invention is related to an organism containing a ribonucleic acid as disclosed herein. Preferably such organism is a vertebrate organism and more preferably the vertebrate organism is a mammal. A mammal as used herein is, among others and not limited thereto, an ape, a dog, a cat, a goat, a sheep, a pig, a guinea pig, a rabbit, a mouse, a rat and a human being.
In a still further aspect the present invention is related to a composition containing a ribonucleic acid according to the present invention. Preferably such composition comprises negative and positive controls either in combination with the effective ribonucleic acid or separated therefrom. Such composition may further comprise a solvent, preferably a buffer.
In a further aspect the present invention is related to a pharmaceutical composition containing a ribonucleic acid according to the present invention and a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are known to the one skilled in the art and comprise, among others, diluent, buffers and the like. The pharmaceutical composition may comprise further pharmaceutically active compounds. In case the disease or condition to be treated using the ribonucleic acid molecules according to the present invention are preferably those which are already now used in connection with the treatment of said diseases or conditions. Due to the different mode of action ofthe ribonucleic acid molecules according to the present invention and the medicaments used for the treatment of said diseases and conditions according to the prior art, synergistic effects will happen.
The invention is now further illustrated by reference to the figures and examples from which further features, embodiments and advantages ofthe present invention may be taken.
Fig. 1 shows a schematic illustration defining the terminology as used herein. The upper of the two strand is the first strand and the antisense strand of the targeted nucleic acid such as mRNA. The second strand is the one which essentially corresponds in its sequence to the targeted nucleic acid and thus forms the sense strand. Both, the first strand and second strand form a double- stranded structure, typically through Watson Crick base pairing.
Fig. 2 illustrates some embodiments of the ribonucleic acid molecules of the present invention with patterns of modified and unmodified groups of nucleotides which is also referred to herein as pattern of modification. The modified groups of nucleotides are also referred to herein as group of modified nucleotides. The unmodified nucleotides or unmodified groups of nucleotides referred to as flanking group(s) of nucleotides herein, as used herein may also have one or several of the modifιcation(s) as disclosed herein which, however, is/are different from the modification of the nucleotides forming the group(s) of modified nucleotides. In Fig. 2A the modified and unmodified groups of nucleotides, i. e. the groups of modified nucleotides and the flanking groups of nucleotides on both the first stretch and the second stretch are located on corresponding parts of the stretches and are thus aligned to each other (groups of modified nucleotides on the first strand aligned with groups of modified nucleotides on the second strand and flanking groups of nucleotides on the first strand aligned with flanking group of nucleotides on the second strand), whereas in Fig. 2B the pattern realised on the first strand is also realised on the second strand, however, with a phase shift such that the modified group of nucleotides of the first stretch is base pairing with an unmodified group of nucleotides ofthe second stretch and vice versa so that a group of modified nucleotides on the first strand alignes with a flanking group of nucleotides on the second strand. In Fig. 2C a further possibility of arranging the modified and unmodified groups of nucleotides is realised. It is also within the present invention that the pattern of the first stretch is independent from the pattern of the second stretch and that both patterns partially overlap in terms of relative position to each other in the double-stranded structure defined by base pairing. In a further embodiment the extent of this overlapping can vary over the length ofthe stretch(es) and strand(s), respectively.
Fig. 3 shows the result of a knockdown experiment using RNAi molecules with different end protection groups. More particularly Fig. 3A shows that the various forms of end protected RNAi molecules are functional on the knockdown of PTEN mRNA.
Fig. 3B shows the sequence ofthe different RNAi molecules used in the experiment the result of which is depicted in Fig. 3A. Fig. 3C shows the result of an immunoblot analysis of PTEN protein after treatment with modified RNAi molecules in comparison to PTEN specific antisense constructs.
Fig. 4 shows that the 3' overhang of RNAi molecules is not important for RNA interference. More particularly, Fig. 4A shows a dose response curve of different RNAi molecules and Fig. 4B shows the sequence of the RNAi molecules used in the experiment the result of which is shown in Fig. 4A.
Fig. 5 shows that duplex length ofthe RNAi molecules has to be at least 18-19 nucleotide. More particularly, Fig. 5B shows the sequence of the PTEN specific RNAi molecules used in the experiment the result of which is depicted in Fig. 5 A as dose response curve.
Fig. 6 shows that four terminal mismatched nucleotides in RNAi molecules with a length of 19 nucleotides are still functional in mediating Aktl knockdown. More particularly, Fig. 6B shows the sequence of the RNAi molecules used in the experiment the result of which is depicted in Fig. 6A.
Fig. 7 shows further results on duplex length requirements and tolerance for mutation in siRNAs. More particularly, Fig. 7A shows the various constructs used (left panel) and the respective impact on inhibition of Aktl mRNA expression in HeLa cells relative to the expression of pi 10a used in the indicated amounts of siRNA molecules (right panel). The nucleotide changes in the mismatch siRNA molecules are indicated by arrows; the 3' desoxynucleotide, if any, are indicated in capital letter. Fig. 7B shows the various PTEN specific siRNAs (left panel), the inhibition of PTEN mRNA expression in HeLA cells expressed as ratio PTEN/pl 10a, at various amounts of siRNA (middle panel) and Fig. 7C a Western Blot analysis depicting the inhibition of PTEN protein expression using PTEN specific siRNA (30nM) and respective mismatch siRNA after 48 and 96 hours, respectively, with plOOα being used as loading control.
Fig. 8 shows the result of studies on the stability in serum conferred to RNAi molecules by 2'-O- methylation and that end modifications have no beneficial effects on RNAi stability. More particularly, Fig. 8A shows the result of a gel electrophoresis of the various RNAi molecules depicted in Fig. 8B being subject to incubation with fetal calf serum.
Fig. 9 shows that an amino end modification results in loss of activity. Fig. 9B shows the particular RNAi molecules used in the experiments the result of which is shown in Fig. 9A expressed as PTEN/pl 10a expression level ratio. Fig. 9C shows the design principles which may be deduced from the results depicted in Fig. 9A. As used in Fig. 9C the term functional means functionally active in the particular assay system as described in the example and "not functional" means not functionally active in said system.
Fig. 10 shows that 2'-O-Alkyl (methyl) modifications stabilize RNAi molecules but also result in reduction of their activity. More particularly, Fig. 10C shows the sequence of the RNAi molecules used in the experiment the result of which is depicted as a dose response curve in Fig. 10 A. Fig. 10B shows the result of a gel electrophoresis of the various RNAi molecules depicted in Fig. 10C being subject to a two hour incubation in fetal calf serum.
Fig. 11 shows the result of an experiment on the efficacy of RNAi molecules with blocks of 2'- O-methyl modifications with Fig. 11 A graphically depicting the results of said experiments as a dose response curve and with Fig. 1 IC showing the sequences ofthe particular RNAi molecules used in said experiments. Fig. 11B shows the result of a gel electrophoresis of the various RNAi molecules depicted in Fig. 1 IC being subject to a two hour incubation in fetal calf serum.
Fig. 12 shows that alternating 2'-O-methyl modification result in activity of the modified RNAi molecules compared to unmodified forms. More particularly, Fig. 12B shows the sequence ofthe RNAi molecules used in this experiment the result of which is depicted in Fig. 12A. Fig. 12C shows the stability of said RNAi molecules following incubation in serum for two hours, whereas Fig. 12D shows an immunoblot for PTEN protein upon application of different RNAi molecules to HeLa cells. As may be taken therefrom RNAi molecules with alternating modifications are stabilized against endonuclease degradation and active in mediating a PTEN protein knock down.
Fig. 13 shows the result of a Western Blot analysis to determine the time course of PTEN protein knock down. Cells were continuously transfected with 2'-O-Methyl modified versus unmodified RNAi molecules using cationic lipids for 72 h. Protein lysates were prepared and analysed by immunoblot after 48 and 120 h. For the 96 h and 120 h timepoints the cells were splitted, replated and incubated in the absence of RNAi molecules for additional 24 and 48 h. .
Fig. 14 shows a Western Blot depicting the protein knock down of PTEN being persistent using alternating modified RNAi molecules versus unmodified RNAi molecules. Transfection were performed for only 5 h and new medium without transfection reagents was added. Lysates were analysed by immunoblot 72h and 96h post transfection with the indicated RNAi molecules.
Fig. 15 shows that siRNA molecules with distinct 2 '-O-methyl ribonucleotides modifications show increased stability in serum and mediate protein knock-down in HeLa cells. More particularly, Fig. 15A indicates the various siRNA molecule constructs used (left panel), whereby 2'-O-methyl ribonucleotides modifications are underlined and indicated by bold letters in the sequence. Inhibition of PTEN mRNA expression in HeLa cells transfected with the indicated amounts of modified siRNA molecules is expressed as ratio PTEN/pllOα and indicated on the right panel. Fig. 15B shows on the left panel the various siRNA constructs used and on the right panel a PAA gel electrophoresis of modified and unmodified siRNA molecules after incubation in serum; the various constructs with 2'-O-methyl ribonucleotides are indicated by underlining and bold printing. Fig. 15C shows an SDS-PAGE based immunoblot illustrating the inhibition of PTEN protein expression using various of the siRNA constructs (30 nM) as depicted in Fig. 15A and 15B, respectively. Again, pllOα is used as loading control. Finally, Fig. 15D is an immunoblot indicating a prolonged protein knock-down, i. e. the inhibition of PTEN protein expression, upon administration of siRNA molecules (30 nM) with distinct 2'-O- methylribonucleotides modifications after 48 and 128 hours. As in Fig. 15C, p 110a is used as loading control.
Fig. 16 shows that siRNA molecules with distinct 2'-O-methylribonucleotides modifications which are specific for Aktl and pllOβ mRNA show increased stability in serum and mediate protein knock-down in HeLa cells. More particularly, Fig. 16A indicates on the left panel the various constructs used whereby again 2'-O-methylribonucleotides are underlined and printed in bold. The integrity of the indicated siRNA molecules after incubation in serum is shown in the right panel. Fig. 16B shows an immunoblot of Aktl, Akt2 and Akt phosphorylation and pllO being used as a loading control upon transfection of the cells with the indicated siRNAs (30 mM). Fig. 16C shows various pi lOβ specific siRNA constructs (left panel) with the 2'-O-methyl modifications being underlined and printed in bold, and the result of an immunoblot analysis
(right panel) of the inhibition of the phosphorylation of the downstream kinase Aktl by said siRNA constructs, pi 10a has been used as a loading control.
Fig. 17 shows the efficacy of various RNAi molecules with hairpin structures as dose response curve while Fig. 17B shows the structure of said RNAi molecules the result of which is depicted in Fig. 17 A. Synthetic siRNAs with different loops are functional in reducing the pllOβ , Aktl and Akt 2 expression. (14A) Inhibition of pllOβ mRNA expression in siRNA transfected HeLa cells. Samples were analyzed in parallel for the level of pl lOβ mRNA expression 24h after transfection of the indicated siRNAs. The transfected bimolecular siRNAs (2 Imer with 3' TT overhangs, molecule 1 AB) or the monomolecular siRNAs with loop structures are schematically shown. Note that the position of the loops (HIV derived pA-loop; (A)ι2-loop) relative to the antisense sequence is reversed in 3A, 4A relative to 3B, 4B. The 2AB siRNA molecule contains 6 mismatches in the 21mer duplex and serves as a negative control together with the untreated sample. RNA was prepared und subjected to real time RT-PCR (Taqman) analysis. pl lOβ mRNA levels are shown relative to the mRNA levels of pl lOα, which serve as an internal reference. Each bar represents triplicate transfections (± standard deviation). HeLa cells were transfected at 50% confluency (2500 cells per 96 well) with siRNAs at the indicated concentrations in growth medium.
Fig. 18 shows the efficacy of various RNAi molecules with intermolecular and intramolecular loop structures as dose response curves. (18A) Inhibition of Aktl mRNA expression in siRNA transfected HeLa cells. Samples were analysed in parallel for the level of Aktl and Akt2 mRNA expression 24h after transfection ofthe indicated siRNAs. The different loops (A-loops; GAGA- loop and a polyethylenglycol (PEG)-linker) and their putative secondary structure are shown schematically. The siRNA molecule 9A is specific for Akt2 and serves as a negative control. Note that 10 A and 10B do not contain self-complementary sequences and are transfected in combination. Aktl mRNA levels is shown relative to the mRNA levels of pllOβ , which served as internal control. (18B) Inhibition of Akt2 mRNA expression in HeLa cells transfected with the indicated siRNA molecules. Akt2 mRNA levels is shown relative to the mRNA levels of pi lOβ. The Aktl specific molecule 7A serves here as a negative control.
Fig. 18 C shows a Western Blot analysis on Akt protein depicting the functionality of synthetic siRNAs with different loops in specifically reducing the Aktl and Akt 2 expression. Inhibition of Aktl and Akt2 protein expression were analysed by immunoblot. The cells were harvested 48h after transfection of the indicated hairpin siRNAs (20nM). Cell extracts were separated by SDS- PAGE and analysed by immunoblotting using anti-pllO antibody, anti Akt 1/2. Similar results were obtained with an antibody specific for the phosphorylated form of Aktl. The positions of pllOα, another catalytic subunit of PI 3-kinase, which was used as a loading control, and of Aktl, Akt2 and phosphorylated Akt (P*-Akt) are indicated on the left.
Fig. 19 shows an NH2 modification, also referred to herein as amino modification, which may be present at either the 3'-OH terminal nucleotide or the 5' terminal nucleotide. The amino group is attached to the phosphate which in turn is attached to the OH group ofthe sugar moiety, through an alkyl group comprising an alkyl chain of 1 to 8, preferably 6 C atoms, whereby the second C atom close to the phosphate group has a CH2OH group attached thereo. As an alternative the linker may be formed by an ether whereby the ether is comprised of two alcohols whereby one alcohol is an amino alcohol and the other is a dialcohol with one alcohol group involved in the formation of the ether group and the other one being an OH group located at either of the C atoms, preferably at the second C atom relative to the phosphate group.
Example 1 : Dose response of synthetic duplex RNAi molecules
In this example the impact of NH2 end protection groups on the activity of duplex RNAi molecules was investigated. Synthetic siRNAs were purchased from Biospring (Frankfurt, Germany). The ribo-oligonucleotides were resuspended in RNase free TE to a final concentration of 50 μM. In the case of bimolecular siRNA molecules equal aliquots (100 μM) were combined to a final concentration of 50 μM. For the formation of intramolecular duplexes the siRNAs were incubated at 50 °C for 2 min in annealing buffer (25mM NaCl; 5mM MgCl2) and were cooled down to RT. Transfections were carried out in 96 well or 10-cm plates (at 30% to 50% confluency) by using various cationic lipids such as Oligofectamine, Lipofectamine (Life Technologies), NC388 (Ribozyme Pharmaceuticals, Inc., Boulder, CO), or FuGene 6 (Roche) according to the manufacturer's instructions. RNAi molecules were transfected by adding preformed 5x concentrated complex of annealed RNAi and lipid in serum-free medium to cells in complete medium. Prior to transfection 2500 HeLa cells were plated per well 15 - 18 hours before transfection for the 96 well format.
The total transfection volume was 100 μl for cells plated in 96-wells and 10 ml for cells in 10 cm plates. The final lipid concentration was 0.8 to 1.2 μg/ml depending on cell density; the RNAi concentration is indicated in each experiment.
Complex formation was allowed to take place for 30 min at 37°C. Complexes were added to cells to yield a final lx concentration of both lipid and RNAi. Depending on the analysis performed following transfection, cells were lysed using a standard cell lysis buffer for protein extraction (Klippel, A., Escobedo, J. A., Hirano, M. and Williams, L. T. (1994). Mol Cell Biol 14, 2675-2685) or a denaturing buffer for RNA isolation according to the RNA isolation kit (Invitek, Berlin (Germany) 24 to 48 hours post transfection for RNA analysis and 48 to 72 hours post transfection for protein analysis by Western Blot.
Determination of the relative amounts of RNA levels by Taqman analysis:
24h post transfection the RNA of cells transfected in 96-wells was isolated and purified using the Invisorb RNA HTS 96 kit (InVitek GmbH, Berlin). Inhibition of PTEN mRNA expression was detected by real time RT-PCR (Taqman) analysis using 300 nM PTEN 5' primer CACCGCCAAATTTAACTGCAGA, 300 nM PTEN 3' primer
AAGGGTTTGATAAGTTCTAGCTGT and 100 nM of the PTEN Taqman probe Fam- TGCACAGTATCCTTTTGAAGACCATAACCCA-Tamra in combination with 40 nM β-actin 5' primer GTTTGAGACCTTCAACACCCCA, 40 nM β-actin 3' primer GACCAGAGGCATACAGGGACA and 100 nM of the β-actin Taqman probe Vic- CCATGTACGTAGCCATCCAGGCTGTG-Tamra. The Akt primers and probes are determined in Sternberger et al. (Sternberger, a.a.O.) and are used according to the manufacturer's instructions (Applied Biosystem; use of Amplicon Set). Also said primers and probes may be designed using the software program Primer Express (Applied Biosystem). The reaction was carried out in 50 μl and assayed on the ABI PRISM 7700 Sequence detector (Applied Biosystems) according to the manufacturer's instructions under the following conditions: 48°C for 30 min, 95°C for 10 min, followed by 40 cycles of 15 sec at 95°C and 1 min at 60°C.
RNA knockdown is shown by real time RT-PCR analysis of HeLa cells transfected with 21 nt long siRNA duplex molecules unmodified and modified with either NH2 or inverted Abasics groups at the 5 '-end at a lipid carrier concentration of 1.0 μg/ml. Cell density was 2000 cells/well. Modifications on the 3 '-end are either RNA overhangs, RNA overhangs with amino groups or DNA overhangs.
Preparation of cell extracts and immunoblotting. Cells were washed twice with cold phosphate-buffered saline and lysed at 4°C in lysis buffer containing 20 mM Tris (pH 7.5), 137 mM NaCl, 15% (vol/vol) glycerol, 1% (vol/vol) Nonidet P-40 (NP-40), 2 mM phenylmethylsulfonyl fluoride, 10 mg aprotinin per ml, 20 mM leupeptin, 2 mM benzamidine, 1 mM sodium vanadate, 25 mM β-glycerol phosphate, 50 mM NaF and 10 mM NaPPi. Lysates were cleared by centrifugation at 14,000 x g for 5 minutes and aliquots of the cell extracts containing equal amounts of protein were analyzed for protein expression by Western-blotting: Samples were separated by SDS-PAGE and transferred to nitrocellulose-filters (Schleicher & Schuell). Filters were blocked in TBST buffer (10 mM Tris-HCl (pH 7.5), 150 mM NaCl, 0.05% (vol/vol) Tween 20, 0.5% (wt/vol) sodium azide) containing 5% (wt/vol) dried milk. The respective antibodies were added in TBST at appropriate dilutions. Bound antibody was detected using anti-mouse- or anti-rabbit-conjugated horse radish peroxidase (Transduction Laboratories) in TBST, washed, and developed using the SuperSignal West Dura (Pierce) or ECL (Amersham) chemoluminescence substrates (c.f. Sternbergeret al. (2002). Antisense. Nucl. Ac. Drug Dev. in press..
Antibodies. The murine monoclonal anti-pl lO antibody U3A and the murine monoclonal anti- p85 antibody N7B have been described (Klippel et al., 1994, aaO). Rabbit polyclonal anti-Akt and anti-phospho Akt (S473) antibodies were obtained from Cell Signaling Technology. The murine monoclonal anti-PTEN antibody was from Santa Cruz Biotechnology. The PTEN 53 specific antisense molecule, i. e. geneBloc, is described in Sternberg et al. [Sternberger, supra] having the following sequence (ucuccuuTTGTTTCTGcuaacga), whereby the nucleotide depicted in lower case are ribonucleotides whereas the nucleotide in capital letters are desoxyribonucleotides. This antisense molecule is also identical to RNAi 1 A without TT.
The results are shown in Fig. 3A and the respective RNAi molecules in Fig. 3B which are directed to the mRNA of PTEN. The nucleotides written in lower case letters represent ribonucleotides whereas capital letters represent desoxyribonucleotides. The term NH2 indicates that the 3' '-position of the ribonucleotide was modified by an amino group. The RNAi molecules used in this and other examples disclosed herein are also referred to as small interfering RNA molecules siRNA. It is to be noted that in any of the figures contained herein the upper strand is the antisense or first strand, whereas the lower strand is the sense or second strand ofthe interfering RNA molecule.
As can be taken from Fig. 3A amino end modifications such as amino modification and inverted abasics modification of the terminal OH group of the nucleic acid are as potent as unmodified ends when the modification is located at the 3' end ofthe antisense strand (see also Fig. 8A; 8B). Therefore chemical modification to stabilize or with other beneficial properties (delivery) will be tolerated without activity loss when located at the 3' OH; especially when the 3 'OH is located on an overhanging nucleotide.
For the experiment shown in Fig. 3C similar conditions as outlined above were used. The first strand and the second strand ofthe RNAi were either modified by a NH2 group at the 3 '-position ofthe ribose moiety or by an inverted abasic at said positions. The first construct is designated as siRNA-NH2 (3A3B) the second as siRNA-iB (4A4B). The sequence of both molecules is depicted in Fig. 3B. The term 3A3B indicates that the interfering ribonucleic acid consists of strand 3 A as the antisense strand and strand 3B as the sense strand. For reason of comparison an antisense oligonucleotide designated GB53 (Steinberger et al., supra) was generated which was directed against the PTEN mRNA as well. The particularities of this latter experiment were as follows.
As may be taken from Fig. 3C end protected RNAi molecules depicted in Fig.3B are functional in yielding a PTEN protein knockdown.
From this example it can be taken that both end protection groups render RNAi molecules active in knocking down PTEN protein. This inhibition is as efficient as inhibition with antisense constructs but at lower concentrations used which is a clear advantage over the already very powerful antisense technology.
Example 2: Overhang requirements for RNAi duplex activity in vivo
The experimental procedures were the same as depicted in connection with example 1 except that the PTEN mRNA targeting interfering RNAi molecules were differently designed. The results are shown in Fig. 4A as dose response curves with Fig. 4B showing the particular sequence and modifications ofthe interfering RNAi molecules used to generate the data depicted in Fig. 4A. The nomenclature is such that, e. g., RNAi 18 is composed of strand 18A as antisense strand and strand 18B as sense strand.
Blunt ended molecules are compared to molecules with 3'-overhangs (RNAi 18) and 5'- overhangs (RNAi 30 and RNAi 31) in their activity to knockdown PTEN mRNA in HeLa cells. The activity of blunt ended molecules (RNAi 28) and molecules with 5 '-overhangs is comparable to the activity of molecules with 3 '-overhangs. This shows that 3 '-overhangs are not essential for RNAi activity.
Example 3: Duplex length requirements of interfering RNA molecules for RNAi activity in vivo
The experimental approach was similar to the one outlined in connection with example 1 except that the interfering RNA molecules were directed against the mRNA of Aktl. The negative control to show the specificity ofthe RNAi molecules was again pi 10 mRNA. The experimental results are shown in Fig. 5A with the particularities of the interfering RNAi molecules used being represented in Fig. 5B. Similar experiments were carried out with further siRNA constructs which are depicted in Fig. 7A, left panel, whereby the arrows indicate mismatches and desoxyribonucleotides are expressed in capital letters. The inhibition of Aktl mRNA expression in HeLa cells transfected with the indicated amounts of siRNA molecules is depicted on the right panel of Fig. 7 A. Taqman analysis on Akt RNA from HeLa cells transfected with different RNAi molecules shows that the doublestrand duplex ofthe siRNA molecules has to be longer than 17 base pairs to show activity whereas molecules with 17 base pair long duplexes or shorter are not functional even if sequence-specific overhangs are added. The shortest RNAi molecules successfully tested were 18 to 19 nucleotides or base pairs in length. It is to be noted that the design of the interfering RNA molecule 51 A/5 IB referred to as RNAi 51 corresponds to the one as described in international patent application WO 01/75164. The RNAi molecule 55A/55B comprises a stretch of 17 nucleotides and has a clearly decreased activity in terms of degradation of Aktl mRNA.
As may be taken from Fig. 7A 19 nt long duplexes are highly efficient in reducing Aktl mRNA levels independent of the nature (desoxy- or ribonucleotides) of the 3' overhang (compare molecules 1AB, 2AB, 3AB, 4AB). The 17 nucleotide long siRNA (molecule 5AB) showed a dramatically reduced silencing activity confirming the above expressed understanding that active siRNA duplexes should be at least 18 nt or longer. Without being wished to be bound by any theory this result may be explained mechanistically by two different requirements. First, a minimum base pairing of 18 nt between the antisense ofthe siRNA and the target mRNA may be obligatory, or second, incorporation into the RNA-induced silencing complex (RISC) requires a minimum length of the siRNA duplex. To address this question a 19 nt long siRNA duplex molecule with one and two terminal mutations (CG and UA inversion) relative to the wildtype sequence was synthesised (molecules 6AB and 7AB). Both molecules, even the molecule with a stretch of only 15 nt base pairing to the target mRNA were functional in inducing the Aktl mRNA level. Therefore, it can be concluded that the duplex length itself, but not the base pairing of the antisense siRNA with the target mRNA seems to determine the minimum length of functional siRNAs. This suggests that the length of the double-stranded helix is an important determinant for the incorporation into the RISC complex. The introduced mismatches at the terminal ends ofthe siRNA duplexes had little effect on RNA interference.
Given the experimental results, the minimum requirement for optimum RNAi mediated interference is thus a duplex length of 18 or 19 nucleotides, independent of the further design of the RNAi molecules such as blunt end or 5 '-overhang or any other form as disclosed herein, but generally applicable to RNAi molecules. However, it has to be acknowledged that the particular design of the RNAi molecules may confer further advantages to said molecules, such as, e. g., increased efficiency and increased stability, respectively. Example 4: Target -antisense homology requirements for RNAi in vivo
The experimental set-up was similar to the one described in example 1, whereby the RNAi is specific for Aktl. In addition, a PTEN specific interfering RNA molecule was designed and used as negative control. The results are shown in Fig. 6A and Fig. 6B. Basically the same experiment was carried out using further siRNA molecules as depicted in Fig. 7B with the results being indicated in Fig. 7B (right panel) and Fig. 7C, respectively.
Having established the minimal duplex length of 18 or more than 18 nucleotides for functional siRNA molecules we have asked the question how many matching nucleotides between target mRNA and siRNA are necessary for silencing activity.As shown by Taqman analysis on Aktl RNA a stretch of 19 to 15 nucleotides perfectly matching to the target RNA, in the present case Akt 1, is sufficient to mediate RNAi activity. A PTEN specific RNAi molecule does not reduce RNA amounts of Aktl thus confirming the specificity of this approach. Mismatches of one or two nucleotides at any or both ends of a strand are functional suggesting that a homolog stretch of 15 nt between a target mRNA and RNAi is sufficient for gene silencing. It can be concluded from these data that unspecific gene silencing can occur by chance through unspecific binding to unrelated targets. This is based on the understanding that a stretch of 15 to 17 matching base pairs is not specific for a single gene and will occur by chance considering the complexitiy and size ofthe genome or transcriptosome of vertrebrates. Apart from the afore-describe experiments also the location ofthe mismatch was subsequently analysed. For this purpose a 19 nt long blunt siRNA directed against PTEN mRNA was used. The sequence changes in one siRNA strand were compensated by complementary changes in the other strand to avoid disrupting duplex formation. As may be taken form both Figs. 7B and C, respectively, a siRNA with only one point mutation in the centre ofthe molecule was severely compromised in its ability to use mRNA and protein expression levels. This result indicates that the RNA machinery is highly discriminative between perfect and imperfect base pairing between target mRNA and siRNA in the centre ofthe duplex. This extreme dependence on a perfect complementarity between target and siRNA has already been described for RNAi interference in the Drosophila system, however, not yet in connection with mammalian systems such as HeLa.
Based on this observation the present invention is reducing this off-target problem of siRNA by two approaches. First by reducing the molecule length of the siRNA molecules to the minimal requirements (18-19 nt) and thereby reducing the chance of homology to off-targets. Second, by inactivation of the sense strand to prevent a unwanted RNA silencing caused by accidential complemetarity ofthe sense strand to a unrelated target RNA (see also Example 6).
Example 5: Stability of modified RNAi molecules in serum
Oligonucleotides were incubated in human serum for 15 min and two hours and loaded on 10% polyacrylamide gel with untreated controls. The results are shown in Fig. 8 A. The various RNAi molecules used are shown and described in more detail in Fig. 8B.
From this example it can be taken that the RNAi duplex of RNA molecules with all nucleotides modified with 2 '-O-methyl groups (RNAi molecules 79A79B and 28A28B) have higher stability in serum. It is also shown that a blunt duplex is more stable than the duplex molecule with overhangs. From this the conclusion may be drawn that end protection (e.g. iB or Amino) is not increasing the stability in serum.
In addition, it can also be concluded that in contrast to the understanding in the art before the filing of this application endonucleases rather than exonucleases are more important in the protection of RNAi molecules.
In view of this, in addition to the various modifications or designs of the inventive RNAi molecules as disclosed in this application a further or additional modification of the nucleotides may be the use of a phosphorothioate backbone of the RNAi molecules which may be either complete or partial in order to inhibit endonuclease function. A complete phosphorothioate backbone means that any of the nucleotides exhibits a phosphorothioate group whereas a partial phosphorothioate backbone means that not all of the nucleotides forming the RNAi molecule have a phosphorothioate modification. This modification is suitable to increase the lifetime of RNAi molecules irrespective of the further design of RNAi molecules. In this regard, a partially or completely phosphorothioate modified RNAi is subject to the present invention which may be realized in connection with the different strategies for the design of interfering RNA molecules as disclosed herein or with any ofthe designs known in the art. Example 6: Inactivation of the sense strand by NH2 end protection groups on the 5' and
3' ends
The experimental set-up was similar to the one described in connection with example 1 with the target nucleic acid sequence being PTEN mRNA. The concentration of HeLa cells was 2,000 cells per well. RNA of PTEN was analysed in Taqman assays after transfection of differently modified RNAi molecules. The different interfering RNA molecules used are depicted in Fig. 9B whereas the experimental results are shown in Fig. 9A.
As may be taken from the dose response curves of various RNAi molecules depicted in Fig. 8 A RNAi molecules are functional when the sense strand, i. e. the second stand, is modified on both ends with amino groups. Particularly effective are RNAi molecules 20A26B, 18A26B, and 28A26B. The lowest activity is shown by RNAi molecule 26A26B which corresponds to end modification on all 4 ends ofthe duplex (Tuschl is 18AB) .
However, RNAi activity is also achieved when the antisense strand, i. e. the first strand, is modified only at the 3' end leaving a free OH group at the 5' end (RNAi constructs 22A26B; 20A26B). There is no activity when the antisense strand is modified with amino groups on both the 5' and the 3' end (26A26B). This leads to the conclusion that any end ofthe antisense strand and more particularly the 5' end of the antisense should be kept without modifications. Additionally, it is worth stating that the NH2 end modification can be used to inactivate the sense strand on the 5' and 3' end and therefore reduce off-target effects mediated by an otherwise functional sense strand which results in a significantly increased specificity of the RNAi molecule which is advantageous for target validation as well as for any medical use ofthe RNAi molecule.
The further generalisation ofthe results from this experiment is depicted in Fig. 9C. Functionally active RNAi are accordingly those not having an amino modification at the antisense strand or having an amino modification only at the 3' end of the antisense strand whereas an amino modification at both ends of the antisense strand is not functional, i. e. does not result in a knockdown ofthe target mRNA. Example 7: Impact of 2 '-O-methyl modification of RNAi molecules for endonuclease protection.
RNA knockdown is again shown using the real time RT-PCR analysis on HeLa cells transfected with RNAi duplex molecules directed against the PTEN mRNA as represented in Fig. 10 A. Experimental procedures were basically the same as specified in example 1. The structure ofthe RNAi molecules investigated and their dose responses, which are depicted in Fig. 10 A, are shown in Fig. IOC. The nucleotides printed in bold are those having a 2'-O-methyl modification.
It is illustrated by the dose response curves shown for various RNAi molecules in Fig. 10A that internal 2'-O-alkyl groups are reducing RNAi activity. Preferably such 2'-O-alkyl groups are 2'- O-methyl or 2'-O-ethyl groups. However, molecules with unmodified nucleotides in combination with 2'-O-alkyl modification show significant activity. As is also depicted in Fig. 10A there is no activity when the antisense strand is all modified with 2 '-O-methyl groups and the sense strand is not modified (c. f, e. g., RNAi molecule 79A28B). Taken the results of a stability test such as incubation ofthe various RNAi molecules in serum, as depicted in Fig. 10B, shows that 2'-O-alkyl modifications stabilize RNAi molecules against degradation. This clearly beneficial effect, however, is at least to a certain degree counterbalanced by the effect that 2'-O- alkyl modifications generally result in a reduced knockdown activity. Accordingly, the design of RNAi molecules has to balance stability against activity which makes it important to be aware of the various design principles as disclosed in the present application.
Example 8: Impact of blocks of internal 2 '-O-methyl modifications on the stability of RNAi molecules in serum.
The experimental approach in connection with this study was actually the same as depicted in example 1. Again, PTEN RNA is analysed by real time RT-PCR on HeLa cells at a density of 2000 cells/well which were transfected with different doses of RNAi molecules. RNAi molecules were incubated in serum for two hours and analysed on a 10% polyacrylamide gel The results of this study are illustrated in Figs. 11A to 11C, whereby Fig. 11A shows the dose response of the various RNAi molecules depicted in Fig. 1 IC and Fig. 1 IB shows the result of a stability test using some of the RNAi molecules depicted in Fig. llC. It is to be acknowledged that the nucleotides written in bold in Fig. 11C are the ones carrying a modification which is in the present case of a 2 '-O-methyl modification ofthe ribose moiety ofthe nucleotides.
There is a dose dependent inhibition by the unmodified RNAi molecules. It is also shown that the 2 '-O-methyl modification of the core 9 nucleotides makes the RNAi stable in serum and allows activity of the duplex in the meaning of mediating the interference phenomenon leading to a degradation of the PTEN mRNA. Total modification of the sense strand makes the RNAi molecule stable in serum and allows certain activity.
Alternating blocks of 5 nucleotides with 2 '-O-methyl modification renders the RNAi molecule stable in serum and allows activity on PTEN RNA as shown by incubating the RNAi duplex in serum for two hours and loading the samples on a 10% polyacrylamide gel. As may be taken from Fig. 11B the duplex comprising strands 80A and 80B is strongly degraded after incubation in serum for two hours. The duplex consisting of strands 82A and 82B confirms the result that the 5 '-end of the first strand which comprises the antisense strand should not be modified at the 5 '-terminal nucleoties (compare 82A82B with the reverse orientated 81A81B). This is also confirmed by the results obtained having the duplex consisting of the strands 86A and 86B which is both active and stabilised in serum. It is noteworthy that molecules with unmodified blocks at the terminal 5' of the antisense strand are more active whereby the 5 'terminal OH group is preferably not derivatized.
Further experiments were carried out using different modification patterns of 2'O-methyl modification of the nucleotides. The results thereof are shown in Fig. 12A to 12C and further discussed herein in example 9.
Example 9: The impact of alternating internal 2'-O-alkyI modification on serum stability of RNAi molecules.
The experimental set-up for performing this kind of study was the same as used in connection with the studies reported in example 1 and example 8, respectively, with the targeted nucleic acid being again PTEN mRNA. HeLa cells were transfected with the different RNAi molecules depicted in Fig. 12B and RNA knockdown was demonstrated using real time RT-PCR on PTEN RNA in a dose-dependent manner (Fig. 12A). The stability of the various RNAi molecules after 15 min and two hours in serum at 37°C is depicted in Fig. 12C and a Western Blot for pi 10 and PTEN as the target-protein ofthe various RNAi molecules is depicted in Fig. 12D with the RNAi molecules tested being the same in both the experiments underlying Fig. 12C and Fig. 12D.
As illustrated in Fig. 12A and Fig. 12C nucleotides modified with 2 '-O-methyl groups alternating with unmodified nucleotides render RNAi molecules stable in serum while still allowing them to be active in the sense of interfering with the target mRNA. It is shown that incubation of RNAi duplex molecules for 15 min and two hours in serum will degrade the unmodified duplex and the duplex where the 10 most 5 '-positioned nucleotides are unmodified.
In the RNAi molecules represented in Fig. 12B various patterns of modified and unmodified nucleotides are realised. The RNAi molecule 94A1/94B1 comprises a structure wherein a modified nucleotide is flanked by an unmodified nucleotide with the unmodified nucleotide being located at the 5' end of the first strand. The RNAi molecule comprised of strands 94A2 and 94B2 is another example where the modified nucleotides and the unmodified nucleotides of the first and the second strand are located at opposing sites. In contrast to this the RNAi molecule comprised of strands 94A1 and 94B2 have the same pattern of modified and unmodified nucleotides. However, there is a phase shift such that the modified nucleotide forms base paring with an unmodified nucleotide. The two RNAi molecules comprised of strands 94A1 and 94B1 and strands 94A2 and 94B2 differ from each other such that in the first case the first strand starts with an unmodified nucleotide and the corresponding first nucleotide of the second strand, i. e. the nucleotide at the 3' end ofthe second strand, starts with an unmodified nucleotide with the arrangement being opposite to this in the RNAi molecule comprised of 94A2 and 94B2.
Additionally, alternatingly modified RNAi molecules as depicted in Fig. 12B are functional in mediating a PTEN protein knock down as shown in Fig. 12D but only when the second 5' and second 3' terminal nucleotide is not modified (see 94A294B1 and 94A294B2). Taken together these data show that the most stable and most active RNAi molecules do have alternating 2'Alkyl modified and unmodified nucleotide residues. It should be noted that these molecules do show a very similar mRNA reduction when compared to unmodified siRNA molecules which being stable in serum allows increased or easier handling. Example 10: Functional Protein Knockdown mediated by internally modified RNAi molecules
The experimental approach was similar to the one outlined in connection with example 1.
Western Blots were performed on HeLa cells harvested at various time points following transfection (48, 72, 96 and 120 hours) with alternatingly modified RNAi molecules as depicted in Fig. 12B. For experimental reasons it is noteworthy that at the 96 hour time point cells have been split and half the population was replated. A total of 40 nM ofthe various RNAi molecules were applied to the cells. Cells were continuously transfected for 72h with cationic lipids as described in example 1; then replated in the absence of transfection reagents.
Transfections were carried out in 96 well or 10-cm plates (at 30% to 50% confluency) by using various cationic lipids such as Oligofectamine, Lipofectamine (Life Technologies), NC388, L8 (Atugen, Berlin), RNAi were transfected by adding pre-formed 5x concentrated complex of siRNAs and lipid in serum-free medium to cells in complete medium. The total transfection volume was 100 μl for cells plated in 96-wells and 10 ml for cells in 10 cm plates. The final lipid concentration was 0.8 to 1.2 μg/ml depending on cell density; the siRNA concentration is indicated in each experiment.
The result of the Western Blot analysis is depicted in Fig. 13. As can be taken from this Figure, modified RNAi molecules of the 94A2B1 and 94A2B2 version yield a longer lasting PTEN protein knock down as unmodified molecules. The lack of protein knock down also seen in Fig. 12 with molecules of the 94A1B1 and 94A1B2 version is confirmed in this experiment. Unmodified molecules (80AB) are not as potent in supporting long lasting protein knock down when the cells are not continuously transfected.
Example 11: Persistent PTEN protein knock down with alternating 2 '-O-methyl modifications of RNAi molecules
The experimental approach was similar to the one outlined in connection with example 10 with the exception that the transfection was terminated after 5 h by replacing the transfection medium with new medium.The protocol was slightly modified such that for each of the RNAi molecules a 40 nM concentration was realized using a stock solution of 1 μg RNAi / ml cationic lipid as described in connection with example 1. 5 hours after transfection the medium was withdrawn and fresh EMEM added. The cells were split after 72, with half of the cells being lysed and the other half newly plated and lysed 24 h later (96h post transfection). The result of a Western Blot analysis using 3 different RNAi molecules (80AB, 94A1/B2, 94A2/B1 are depicted in Fig. 14. As a positive control untreated cells were used. Fig. 14 shows the expression of PTEN after 72 h and 96 h, respectively. Taken the structural particularities of the various RNAi molecules it can be taken from Fig. 14 that protein knockdown is persistent with alternating molecules of the 94A2B1 kind even over 96 h after splitting and replating cells compared to unmodified RNAi molecules (such as 80AB) and RNAi molecule 94A1B2.
A further experiment was carried out using the siRNA constructs as depicted in Fig. 15A (left panel). From the results as depicted as ratio of PTEN/pl lOα mRNA degradation at the various concentrations of siRNA constructs administered to the test system, it can be taken that siRNA molecules with either one or both strands consisting of 2 '-O-methyl residues were not able to induce RNA interference in the mammalian system (Fig. 15 A, molecules V2, V5, V6). However, the decrease in activity was less pronounced when only parts of the strands were modified. Interestingly, a molecule having an unmodified antisense strand (which is the upper strand in the representation throughout this specification if not indicated otherwise) and a completely modified sense strand was significantly more active when compared to the reversed version (Fig. 5 A, molecules V5 and V6). This result suggests that the antisense strand of the siRNA seems to be more critical and sensitive to modification. The most efficient molecules inducing PTEN mRNA had only stretches of modifications leaving the 5' end unmodified or were modified on alternating positions on both strands (Fig. 15A, molecules V10, V12).
To test the nuclease resistance the different siRNA versions were incubated in serum followed PAA gelelectrophoresis. The result is shown in Fig. 15B (right panel with the various sequences indicated on the left panel of Fig. 15B). As shown earlier, blunt ended siRNA molecules with unmodified ribonucleotides were very rapidly degraded whereas a complete substitution with 2'- O-methyl nucleotides mediated resistance against serum-derived nucleases (Fig. 15B, compare molecule AB with VI). siRNA molecules with partial 2'-O-methyl modification showed as well an increased stability when compared to unmodified siRNAs. Especially molecules with alternating modifications on both strands showed a significant improve in instability (Fig. 15B, molecules V13, V14, VI 5 and V12). More importantly, transfection of three of these molecules into HeLa cells resulted in a significant down regulation of PTEN protein expression as depicted in Fig. 15C, length 6, 9 and 10). In this RNA interference activity assay an unexpected preference for molecules was observed which were modified at every second nucleotide beginning with the most 5' terminal nucleotide ofthe antisense strand (molecules V15 and V12). Molecules which contained modifications beginning with the second nucleotide at the 5' end of the antisense strand were more stable but had a strongly reduced activity in gene silencing (molecules VI 3, V14). This result points towards highly "specific interactions between the involved enzymes and precise nucleotides in the siRNA duplex. Taken together the data shown herein demonstrate that 2 '-O-methyl modifications at particularly selected positions in the siRNA duplex can increase nuclease resistance and do not necessarily abolish RNAi completely.
Although an increased stability of synthetic siRNA has primarily implication for in vivo application, it was also analysed whether the particular modification can also lead to an extended protein knock-down in cell culture systems. In accordance therewith HeLa cells were transiently transfected for six hours using different versions of PTEN specific siRNAs. The lipid siRNA complex was then washed away and the PTEN protein knock-down was analysed 48 hours and 120 hours later. Although knock-down experiments without continued transfection of siRNAs are complicated due to rapid growth of untransfected cells in this time period resulting in a very transient knock-down, the present inventors were able to demonstrate a prolonged PTEN protein knock-down with siRNA molecules stabilised by the described 2 '-O-methyl modification. At 48 hours post transfection the unmodified siRNA (AB) showed the biggest reduction in PTEN protein levels, however, at 120 hours post transfection the reduction in PTEN protein expression is superior with the siRNA stabilised by alternating 2 '-O-methyl modifications (Fig. 15D, compare lane 2 with lanes 4, 6 and 7).
From the results it can also be taken that preferably the starting nucleotide position of the alternating modification seems to be important. To test this preference in more detail two additional series of siRNAs were synthesised, one specific for the kinase Aktl and the other one specific for pllOβ which is one of the two catalytic subunits of PI(3-) kinase. The particular constructs are shown in Fig. 16 A. As may be taken therefrom only 19 nt long siRNA duplexes either without any or with 2'-O-methyl modification on every second other nucleotide were used. Using Aktl as a target an efficient protein knock-down as well as a dramatic reduction in phospho-Akt levels was observed with blunt, unmodified siRNAs (Fig. 16A, right panel). From the different versions of molecules with modifications on every second other nucleotide only one was efficiently mediating RNAi (Fig. 16 A, molecule V5). This siRNA molecule contained an antisense strand which was modified at the most terminal 5' and 3' nucleotides. The sense strand started with the unmodified nucleotides at the most terminal position, resulting in a structure in which the modified and unmodified ribonucleotides of both strands are facing each other. As expected, this molecule was also protected against serum-derived nucleases as depicted in Fig. 16B (molecule V5).
Interestingly, a very similar 19 nt long siRNA molecule (V4) with modifications beginning at the second nucleotide of the antisense strand showed no RNA interference activation in the particular assay used. The version V6 in which the modified nucleotides of the antisense strand are facing modified nucleotides on the sense strand, was also inactive in this experiment. An identical series of 19 nt long siRNA molecules specific for pl lOβ confirmed these observations as depicted in Fig. 16C. Again the similarily modified siRNA molecule (N5) was the most active, as indicated by reducing Akt phosphorylation, which is indicative for a reduced P (I)-3 kinase activity due to reduced pl lOβ levels. The reduced activity of the molecules N6 might be explained by reduced duplex stability since the same structure was active in the PTEΝ knockdown experiment with 2 Imer siRNAs. Although it is known that 2 '-O-methyl modification on both strands facing each other will reduce the stability of nucleic acid duplexes, the difference between the activity of siRΝA molecules V4 and V5 (Figs. 16B and C) is probably not due to differences in duplex stability since the number of base pairing of modified and unmodified nucleotides is identical. This difference in activity might be due to specific requirements in the interacting proteins involved in the degradation of the target mRΝA. Also it can be taken from these experiments that the most terminal nucleotides of the antisense strand can be modified at the 2 '-OH-group with significant loss of silencing activity.
Example 12: Different loop structures are functional in mediating RΝA interference
To test whether RΝAi molecules, preferably synthetic RΝAi molecules with self complementary structures can inhibit gene expression as efficiently as standard double stranded siRΝA molecules, HeLa cells were transfected with pl lOβ specific synthetic siRNAs. Transfections were carried out in 96 well or 10-cm plates (at 30% to 50% confluency) by using various cationic lipids such as Oligofectamine, Lipofectamine (Life Technologies), GeneBlocs were transfected by adding pre-formed 5x concentrated complex of GB and lipid in serum-free medium to cells in complete medium. The total transfection volume was 100 μl for cells plated in 96-wells and 10 ml for cells in 10 cm plates. The final lipid concentration was 0.8 to 1.2 μg/ml depending on cell density; the RNAi molecule concentration is indicated in each experiment.
A dose dependent titration showed no significant difference in efficiency of mRNA knock-down achieved by the standard bimolecular double strand 2 Imer and the corresponding monomolecular molecules as analysed by realtime PCR (Taqman) (Fig. 17A). Two different loop structures a (A)ι2 loop and a HTN derived pA-loop were tested in parallel with similar results. A comparision of the relative position of the antisense sequence and the loop structure revealed an improved knock-down efficiency with the antisense sequence being located 3 ' to the loop (Fig. 17B; compare construct 3 A, 3B and 4A, 4B).
Example 13: Studies on the efficacy of different intramolecular hairpin loops and intermolecular "bubbles"
The influence of different loop structures on inhibition of mRΝA and protein expression was tested. For these experiments Aktl and Akt2 were chosen as targets. The experimental approach was similar to the one described in example 12.
Significantly, the reduction in Aktl mRΝA as depicted in Fig. 18A and Fig. 18B as well as Aktl protein levels as depicted in Fig. 18C was completely independent of the loop structure tested (compare molecules 5A, 6A, 7A, 8A) (the structure of the RΝAi molecule tested is always depicted underneath the bar diagram). Even a molecule containing a rather unphysiological structure such as a polyethylenglycol linker (PEG) as a loop efficiently reduced Aktl expression indicating that size and nucleotide sequence of the loop is not crucial (Fig. 18 A; molecule 8A). A synthetic siRΝA molecule specific for Akt2 (9 A) was used to control for specificity, and had no effect on Aktl levels as shown in Fig. 15 A. This molecule, however, efficiently silenced Akt2 expression (Fig. 18B; Fig. 18C). Self-complementary RΝA molecules with loop structures have the possibility to anneal as doublestrands in monomolecular or bimolecular structures under physiological hybridization conditions (Fig. 18B, loop or bubble structure). To address the question of whether the siRΝA molecules exert their function via adapting an intra-molecular loop or an inter-molecular "bubble" (schematically shown in Fig. 18B) two molecules not capable of folding back on themselves were transfected. These constructs contained Aktl- and
Akt2-specific sequences within the same molecule (Fig. 18B, constructs 10 A, 10B) and were designed to be restricted to form a bimolecular duplex ("bubble"). Surprisingly, this molecule efficiently mediated both Aktl and Akt2 mRNA knock-down as well as protein knock-down when transfected after annealing of both strands.
Whether loop and bubble structures are indeed substrates for RNA processing enzymes, e.g. Dicer, is not clear at this point. A recent study by Paddison and coworkers suggests that hairpin containing siRNAs are more dependent on Dicer activity than double stranded siRNAs. However, these data demonstrating RNA interference activity using a PEG linker molecule indicate that the linker sequence is likely to be irrelevant.
The features of the present invention disclosed in the specification, the claims and/or the drawings may both separately and in any combination thereof be material for realizing the invention in various forms thereof.

Claims

Claims
1. A ribonucleic acid comprising a double stranded structure, whereby the double- stranded structure comprises a first strand and a second strand, whereby the first strand comprises a first stretch of contiguous nucleotides and whereby said first stretch is at least partially complementary to a target nucleic acid, and the second strand comprises a second stretch of contiguous nucleotides and whereby said second stretch is at "least partially identical to a target nucleic acid,
characterised in
that said first strand and/or said second strand comprises a plurality of groups of modified nucleotides having a modification at the 2'-position whereby within the strand each group of modified nucleotides is flanked on one or both sides by a flanking group of nucleotides whereby the flanking nucleotides forming the flanking group of nucleotides is either an unmodified nucleotide or a nucleotide having a modification different from the modification ofthe modified nucleotides.
2. The ribonucleic acid according to claim 1, wherein said first strand and/or said second strand comprise said plurality of modified nucleotides.
3. The ribonucleic acid according to claims 1 or 2, wherein said first strand comprises said plurality of groups of modified nucleotides.
4. The ribonucleic acid according to claims 1 or 2, wherein said second strand comprises said plurality of groups of modified nucleotides.
5. The ribonucleic acid according to any of claims 1 to 4, wherein the group of modified nucleotides and/or the group of flanking nucleotides comprises a number of nucleotides whereby the number is selected from the group comprising one nucleotide to 10 nucleotides.
6. The ribonucleic acid according to any of claims 1 to 5, wherein the pattern of modified nucleotides of said first strand is the same as the pattern of modified nucleotides of said second strand.
7. The ribonucleic acid according to any of claims 1 to 6, wherein the pattern of said first strand aligns with the pattern of said second strand.
8. The ribonucleic acid according to any of claims 1 to 6, wherein the pattern of said first strand is shifted by one or more nucleotides relative to the pattern ofthe second strand.
9. The ribonucleic acid according to any of claims 1 to 8, wherein the modification is selected from the group comprising amino, fluoro, methoxy, alkoxy and alkyl.
10. The ribonucleic acid according to any of claims 1 to 9, wherein the double stranded structure is blunt ended.
11. The ribonucleic acid according to any of claims 1 to 10, wherein the double stranded structure is blunt ended on both sides.
12. The ribonucleic acid according to any of claims 1 to 11, wherein the double stranded structure is blunt ended on the side on the double stranded structure which is defined by the 5'-end ofthe first strand and the 3'-end ofthe second strand.
13. The nucleic acid according to any of claims 1 to 12, wherein the double stranded structure is blunt ended on the side on the double stranded structure which is defined by the 3'-end ofthe first strand and the 5'-end ofthe second strand.
14. The ribonucleic acid according to claim 1 to 13, wherein at least one of the two strands has an overhang of at least one nucleotide at the 5 '-end.
15. The ribonucleic acid according to claim 14, wherein the overhang consists of at least one nucleotide which is selected from the group comprising ribonucleotides and deoxyribonucleotides.
16. The ribonucleic acid according to claim 15, wherein the nucleotide has a modification, whereby said modification is preferably selected from the group comprising nucleotides being an inverted abasic and nucleotides having an NH2-modification at the 2 '-position.
17. The ribonucleic acid according to any of claims 14 to 16, wherein the overhang at the 5'- end is on the second strand.
18. The ribonucleic acid according to claim 17, characterised in that the first strand comprises also an overhang, preferably at the 5 '-end.
19. The ribonucleic acid according to any of claims 14 to 18, wherein the overhang at the 5'- end is on the first strand.
20. The ribonucleic acid according to claim 19, characterised in that the second strand also comprise an overhang, preferably at the 5 '-end.
21. The ribonucleic acid according to any of claims 14 to 20, wherein at least one of the strands has an overhang of at least one nucleotide at the 3 '-end.
22. The ribonucleic acid according to claim 21, wherein the nucleotide at the 3'-end is selected from the group comprising ribonucleotides and deoxyribonucleotides.
23. The ribonucleic acid according to any of claims 14 to 22, wherein the 3 '-end of the first strand comprises an overhang.
24. The ribonucleic acid according to any of claims 1 to 23, wherein the length ofthe double- stranded structure has a length from about 17 to 21 and more preferably 18 or 19 bases.
25. The ribonucleic acid according to any of claims 1 to 24, wherein the length of said first strand and/or the length of said second strand is independently from each other selected from the group comprising the ranges of from about 15 to about 23 bases, 17 to 21 bases and 18 or 19 bases.
26. The ribonucleic acid according to any of claims 1 to 25, wherein the complementarity between said first strand and the target nucleic acid is perfect.
27. The ribonucleic acid according to any of claims 1 to 26, wherein the duplex formed between the first strand and the target nucleic acid comprises at least 15 nucleotides wherein there is one mismatch or two mismatches between said first strand and the target nucleic acid forming said double-stranded structure.
28. The ribonucleic acid according to any of claims 1 to 27, wherein
both the first strand and the second strand each comprise at least one group of modified nucleotides and at least one flanking group of nucleotides,
whereby each group of modified nucleotides comprises at least one nucleotide and whereby each flanking group of nucleotides comprising at least one nucleotide;
with each group of modified nucleotides of the first strand being aligned with a flanking group of nucleotides on the second strand, whereby
the most terminal 5' nucleotide ofthe first strand is a nucleotide ofthe group of modified nucleotides, and
the most terminal 3' nucleotide ofthe second strand is a nucleotide ofthe flanking group of nucleotides.
29. The ribonucleic acid according to claim 28, wherein each group of modified nucleotides consists of a single nucleotide and/or each flanking group of nucleotides consists of a single nucleotide.
30. The ribonucleic acid according to claim 29, wherein on the first strand the nucleotide forming the flanking group of nucleotides is an unmodified nucleotide which is arranged in a 3' direction relative to the nucleotide forming the group of modified nucleotides, and wherein on the second strand the nucleotide forming the group of modified nucleotides is a modified nucleotide which is arranged in 5' direction relative to the nucleotide forming the flanking group of nucleotides.
31. The ribonucleic acid according to any of claims 28 to 30, wherein the first strand comprises eight to twelve, preferably nine to eleven, groups of modified nucleotides, and wherein the second strand comprises seven to eleven," preferably eight to ten, groups of modified nucleotides.
32. The ribonucleic acid according to any of claims 1 to 31, wherein the target gene is selected from the group comprising structural genes, housekeeping genes, transcription factors, motility factors, cell cycle factors, cell cycle inhibitors, enzymes, growth factors, cytokines and tumor suppressors.
33. The ribonucleic acid according to any of claims 1 to 32, whereby the first strand and the second strand are linked by a loop structure.
34. The ribonucleic acid according to claim 33, wherein the loop structure is comprised of a non-nucleic acid polymer.
35. The ribonucleic acid according to claim 34, characterised in that the non-nucleic acid polymer is polyethylene glycol.
36. The ribonucleic acid according to any of claims 33 to 35, wherein the loop structure is comprised of a nucleic acid.
37. The ribonucleic acid according to any of claims 33 to 36, characterised in that the 5'- terminus ofthe first strand is linked to the 3 '-terminus ofthe second strand.
38. The ribonucleic acid according to any of claims 33 to 37, characterised in that the 3 '-end ofthe first strand is linked to the 5 '-terminus ofthe second strand.
39. Use of a ribonucleic acid according to any of claims 1 to 38, for target validation.
40. Use of a ribonucleic acid according to any of claims 1 to 38 for the manufacture of a medicament.
41. The use according to claim 40, wherein the medicament is for the treatment of a disease or of a condition which is selected from the group comprising glioblastoma, prostate cancer, breast cancer, lung cancer, liver cancer, colon cancer, pancreatic cancer and leukaemia, diabetes, obesity, cardiovascular diseases, and metabolic diseases.
42. A cell, preferably a knockdown cell, containing a ribonucleic acid according to any of claims 1 to 38.
43. An organism, preferably a knockdown organism, containing a ribonucleic acid according to any of claims 1 to 38.
44. A composition containing a ribonucleic acid according to any of claims 1 to 38.
45. A pharmaceutical composition containing a ribonucleic acid according to any of claims 1 to 38 and a pharmaceutically acceptable carrier.
46. A method for inhibiting the expression of a target gene in a cell or derivative thereof comprising introducing a ribonucleic acid according to any of claims 1 to 38 into the cell in an amount sufficient to inhibit expression ofthe target gene, wherein the target gene is the target gene ofthe a ribonucleic acid according to any of claims 1 to 38.
PCT/EP2003/008666 2002-08-05 2003-08-05 Further novel forms of interfering rna molecules WO2004015107A2 (en)

Priority Applications (20)

Application Number Priority Date Filing Date Title
AU2003260370A AU2003260370B2 (en) 2002-08-05 2003-08-05 Further novel forms of interfering RNA molecules
ES03784183.0T ES2280826T5 (en) 2002-08-05 2003-08-05 Additional new forms of interfering RNA molecules
BRPI0313202A BRPI0313202A8 (en) 2002-08-05 2003-08-05 ADDITIONAL WAYS TO INTERFERE WITH RNA MOLECULES
EP10012263.9A EP2258847B2 (en) 2002-08-05 2003-08-05 Futher novel forms of interfering RNA molecules
JP2004526885A JP4705370B2 (en) 2002-08-05 2003-08-05 Newer forms of interfering RNA molecules
KR1020057002008A KR101201664B1 (en) 2002-08-05 2003-08-05 Further novel forms of interfeing RNA molecules
EP06027129.3A EP1857547B2 (en) 2002-08-05 2003-08-05 Further novel forms of interfering RNA molecules
IL16654603A IL166546A0 (en) 2002-08-05 2003-08-05 Further novel forms of interfering ran molecules
EP03784183.0A EP1527176B2 (en) 2002-08-05 2003-08-05 Further novel forms of interfering rna molecules
MXPA05001355A MXPA05001355A (en) 2002-08-05 2003-08-05 Further novel forms of interfering rna molecules.
EP17160954.8A EP3222724B1 (en) 2002-08-05 2003-08-05 Further novel forms of interfering rna molecules
CA2494930A CA2494930C (en) 2002-08-05 2003-08-05 Modified forms of interfering rna molecules
DK03784183.0T DK1527176T4 (en) 2002-08-05 2003-08-05 ADDITIONAL NEW FORMS OF INTERFERRING RNA MOLECULES
SI200330726T SI1527176T1 (en) 2002-08-05 2003-08-05 Further novel forms of interfering rna molecules
DE60310944.6T DE60310944T3 (en) 2002-08-05 2003-08-05 OTHER NEW FORMS OF INTERFERING RNS MOLECULES
ZA2005/00459A ZA200500459B (en) 2002-08-05 2005-01-18 Further novel forms of interfering rna molecules
IL16654605A IL166546A (en) 2002-08-05 2005-01-27 Double stranded ribonucleic acid, composition containing same, use thereof in the preparation of medicaments and an in vitro method for inhibiting the expression of a target gene
CY20071100439T CY1106422T1 (en) 2002-08-05 2007-03-29 ADDITIONAL NEW FORMS OF INTERFERING RNA
AU2008207337A AU2008207337A1 (en) 2002-08-05 2008-08-15 Further novel forms of interfering RNA molecules
CY181101252T CY1120919T1 (en) 2002-08-05 2018-11-27 ADDITIONAL NEW FORMS OF INTERFERED RNA MOLECULES

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP02017601.2 2002-08-05
EP02017601A EP1389637B1 (en) 2002-08-05 2002-08-05 Blunt-ended interfering RNA molecules
US40254102P 2002-08-12 2002-08-12
US60/402,541 2002-08-12
EP03008383 2003-04-10
EP03008383.6 2003-04-10

Publications (2)

Publication Number Publication Date
WO2004015107A2 true WO2004015107A2 (en) 2004-02-19
WO2004015107A3 WO2004015107A3 (en) 2004-04-29

Family

ID=31720998

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2003/008666 WO2004015107A2 (en) 2002-08-05 2003-08-05 Further novel forms of interfering rna molecules

Country Status (19)

Country Link
US (15) US7452987B2 (en)
EP (6) EP1857547B2 (en)
JP (4) JP5490655B2 (en)
KR (1) KR101201664B1 (en)
CN (1) CN100543137C (en)
AT (1) ATE350473T2 (en)
AU (2) AU2003260370B2 (en)
BR (1) BRPI0313202A8 (en)
CA (1) CA2494930C (en)
CY (4) CY1106422T1 (en)
DE (1) DE60310944T3 (en)
DK (4) DK1527176T4 (en)
ES (1) ES2280826T5 (en)
IL (2) IL166546A0 (en)
MX (1) MXPA05001355A (en)
PT (1) PT1527176E (en)
SI (3) SI1857547T1 (en)
WO (1) WO2004015107A2 (en)
ZA (1) ZA200500459B (en)

Cited By (120)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005021749A1 (en) * 2003-08-28 2005-03-10 Novartis Ag Interfering rna duplex having blunt-ends and 3’-modifications
WO2005097992A3 (en) * 2004-04-01 2006-02-02 Dharmacon Inc Modified polynucleotides for reducing off-target effects in rna interference
WO2006035434A2 (en) 2004-09-28 2006-04-06 Quark Biotech, Inc. Oligoribonucleotides and methods of use thereof for treatment of alopecia, acute renal failure and other diseases
EP1644475A2 (en) * 2003-06-20 2006-04-12 Isis Pharmaceuticals, Inc. DOUBLE STRANDED COMPOSITIONS COMPRISING A 3’-ENDO MODIFIED STRAND FOR USE IN GENE MODULATION
WO2005121372A3 (en) * 2004-06-03 2006-04-13 Isis Pharmaceuticals Inc Double strand compositions comprising differentially modified strands for use in gene modulation
EP1766052A1 (en) * 2004-06-03 2007-03-28 Isis Pharmaceuticals, Inc. Chimeric gapped oligomeric compositions
WO2007039268A1 (en) * 2005-10-03 2007-04-12 Novartis Forschungsstiftung, Zweigniederlassung Methods for regulating metabolism and diagnosing disease
EP1791568A2 (en) * 2004-08-16 2007-06-06 Quark Biotech, Inc. Therapeutic uses of inhibitors of rtp801
JP2007151544A (en) * 2005-11-08 2007-06-21 Ehime Univ siRNA SPECIFIC FOR Akt GENE
EP1897941A1 (en) * 2006-09-11 2008-03-12 Max-Delbrück-Centrum Für Molekulare Medizin Means for increasing intracellular levels of protoporphyrin IX and use thereof
EP1984003A2 (en) * 2006-01-20 2008-10-29 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of rtp801
US7452987B2 (en) 2002-08-05 2008-11-18 Silence Therapeutics Aktiengesellschaft (Ag) Interfering RNA molecules
JP2009514877A (en) * 2005-11-04 2009-04-09 エムディーアールエヌエー,インコーポレイテッド Peptide-Dither substrate RNA conjugates as siRNA delivery vehicles
WO2009044392A2 (en) 2007-10-03 2009-04-09 Quark Pharmaceuticals, Inc. Novel sirna structures
EP2076526A2 (en) * 2006-10-25 2009-07-08 Quark Pharmaceuticals, Inc. Novel sirnas and methods of use thereof
US7626015B2 (en) 2006-06-09 2009-12-01 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801L
WO2009147684A2 (en) 2008-06-06 2009-12-10 Quark Pharmaceuticals, Inc. Compositions and methods for treatment of ear disorders
EP2155772A1 (en) * 2007-05-01 2010-02-24 City of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US7695902B2 (en) 1996-06-06 2010-04-13 Isis Pharmaceuticals, Inc. Oligoribonucleotides and ribonucleases for cleaving RNA
US7750144B2 (en) 2003-06-02 2010-07-06 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
WO2010080452A2 (en) 2008-12-18 2010-07-15 Quark Pharmaceuticals, Inc. siRNA COMPOUNDS AND METHODS OF USE THEREOF
JP2010172344A (en) * 2003-04-02 2010-08-12 Dharmacon Inc Modified polynucleotide for use in rna interference
WO2010091878A2 (en) 2009-02-13 2010-08-19 Silence Therapeutics Ag Means for inhibiting the expression of opa1
WO2010094491A1 (en) 2009-02-18 2010-08-26 Silence Therapeutics Ag Means for inhibiting the expression of ang2
US7812002B2 (en) 2007-03-21 2010-10-12 Quark Pharmaceuticals, Inc. Oligoribonucleotide inhibitors of NRF2 and methods of use thereof for treatment of cancer
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US7825099B2 (en) 2006-01-20 2010-11-02 Quark Pharmaceuticals, Inc. Treatment or prevention of oto-pathologies by inhibition of pro-apoptotic genes
EP2256191A1 (en) * 2008-02-15 2010-12-01 Riken Cyclic single-stranded nucleic acid complex and method for producing the same
EP2268316A2 (en) * 2008-03-20 2011-01-05 Quark Pharmaceuticals, Inc. NOVEL siRNA COMPOUNDS FOR INHIBITING RTP801
US7872119B2 (en) 2007-02-26 2011-01-18 Quark Pharmaceuticals, Inc. Inhibitors of RTP801 and their use in disease treatment
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
US7892793B2 (en) 2002-11-04 2011-02-22 University Of Massachusetts Allele-specific RNA interference
EP2285385A2 (en) * 2008-04-15 2011-02-23 Quark Pharmaceuticals, Inc. siRNA COMPOUNDS FOR INHIBITING NRF2
EP2292739A1 (en) 2006-03-24 2011-03-09 Institut National De La Recherche Agronomique Method for preparing differentiated avian cells and genes involved in the maintenance of pluripotency
US7910566B2 (en) 2006-03-09 2011-03-22 Quark Pharmaceuticals Inc. Prevention and treatment of acute renal failure and other kidney diseases by inhibition of p53 by siRNA
US7923206B2 (en) 2004-11-22 2011-04-12 Dharmacon, Inc. Method of determining a cellular response to a biological agent
US7923207B2 (en) 2004-11-22 2011-04-12 Dharmacon, Inc. Apparatus and system having dry gene silencing pools
US7935811B2 (en) 2004-11-22 2011-05-03 Dharmacon, Inc. Apparatus and system having dry gene silencing compositions
US7947658B2 (en) 2003-09-12 2011-05-24 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
WO2011066475A1 (en) 2009-11-26 2011-06-03 Quark Pharmaceuticals, Inc. Sirna compounds comprising terminal substitutions
EP2330111A2 (en) 2004-01-30 2011-06-08 Quark Pharmaceuticals, Inc. Oligoribonucleotides and methods of use thereof for treatment of fibrotic conditions and other diseases
WO2011072091A1 (en) 2009-12-09 2011-06-16 Quark Pharmaceuticals, Inc. Methods and compositions for treating diseases, disorders or injury of the cns
WO2011084193A1 (en) 2010-01-07 2011-07-14 Quark Pharmaceuticals, Inc. Oligonucleotide compounds comprising non-nucleotide overhangs
US20110229557A1 (en) * 2008-10-22 2011-09-22 Elena Feinstein Methods for treating eye disorders
US8084600B2 (en) 2006-05-04 2011-12-27 Novartis Ag Short interfering ribonucleic acid (siRNA) with improved pharmacological properties
WO2011163436A1 (en) 2010-06-24 2011-12-29 Quark Pharmaceuticals, Inc. Double stranded rna compounds to rhoa and use thereof
EP2407558A1 (en) 2006-10-31 2012-01-18 Noxxon Pharma AG Methods for the detection of a single- or double-stranded nucleic acid molecule
EP2412724A1 (en) 2010-07-29 2012-02-01 Centre National de la Recherche Scientifique (C.N.R.S) Regulation of Glypican 4 activity to modulate the fate of stem cells and uses thereof
WO2012037254A1 (en) * 2010-09-15 2012-03-22 Alnylam Pharmaceuticals, Inc. MODIFIED iRNA AGENTS
US8188060B2 (en) 2008-02-11 2012-05-29 Dharmacon, Inc. Duplex oligonucleotides with enhanced functionality in gene regulation
WO2012078536A2 (en) 2010-12-06 2012-06-14 Quark Pharmaceuticals, Inc. Double stranded oligonucleotide compounds comprising positional modifications
US8252755B2 (en) 2006-09-22 2012-08-28 Dharmacon, Inc. Duplex oligonucleotide complexes and methods for gene silencing by RNA interference
WO2012118911A1 (en) 2011-03-03 2012-09-07 Quark Pharmaceuticals, Inc. Oligonucleotide modulators of the toll-like receptor pathway
US8309704B2 (en) 2003-06-02 2012-11-13 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNAi
US8329463B2 (en) 2000-12-01 2012-12-11 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
EP2546337A1 (en) 2006-07-21 2013-01-16 Silence Therapeutics AG Means for inhibiting the expression of protein kinase 3
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
US8394628B2 (en) 2000-03-30 2013-03-12 University Of Massachusetts RNA sequence-specific mediators of RNA interference
WO2013070821A1 (en) 2011-11-08 2013-05-16 Quark Pharmaceuticals, Inc. Methods and compositions for treating diseases, disorders or injury of the nervous system
WO2013098813A1 (en) 2012-01-01 2013-07-04 Qbi Enterprises Ltd. Endo180-targeted particles for selective delivery of therapeutic and diagnostic agents
WO2013106494A1 (en) 2012-01-12 2013-07-18 Quark Pharmaceuticals, Inc. Combination therapy for treating hearing and balance disorders
WO2013074974A3 (en) * 2011-11-18 2013-08-08 Alnylam Pharmaceuticals, Inc. Modified rnai agents
WO2013123996A1 (en) 2012-02-24 2013-08-29 Astrazeneca Uk Limited Novel sirna inhibitors of human icam-1
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
WO2013166043A1 (en) 2012-05-02 2013-11-07 Children's Hospital Medical Center Rejuvenation of precursor cells
US8604183B2 (en) 2002-11-05 2013-12-10 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
WO2014043289A2 (en) 2012-09-12 2014-03-20 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to ddit4 and methods of use thereof
WO2014043292A1 (en) 2012-09-12 2014-03-20 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to p53 and methods of use thereof
US8680063B2 (en) 2003-09-12 2014-03-25 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
US8785408B2 (en) 2007-06-27 2014-07-22 Quark Pharmaceuticals, Inc. Compositions and methods for reducing or protecting against delayed graft function (DGF)
WO2014176259A1 (en) 2013-04-22 2014-10-30 Icahn School Of Medicine At Mount Sinai Mutations in pdgfrb and notch3 as causes of autosomal dominant infantile myofibromatosis
WO2015005253A1 (en) 2013-07-08 2015-01-15 第一三共株式会社 Novel lipid
WO2015015498A1 (en) 2013-07-31 2015-02-05 Qbi Enterprises Ltd. Methods of use of sphingolipid polyalkylamine oligonucleotide compounds
WO2015015496A1 (en) 2013-07-31 2015-02-05 Qbi Enterprises Ltd. Sphingolipid-polyalkylamine-oligonucleotide compounds
EP2853595A1 (en) 2013-09-30 2015-04-01 Soluventis GmbH NOTCH 1 specific siRNA molecules
EP2865758A1 (en) * 2013-10-22 2015-04-29 Sylentis, S.A.U. siRNA and their use in methods and compositions for inhibiting the expression of the ORAI1 gene
EP2865757A1 (en) * 2013-10-22 2015-04-29 Sylentis, S.A.U. siRNA and their use in methods and compositions for inhibiting the expression of the PDK1 gene.
EP2865756A1 (en) * 2013-10-22 2015-04-29 Sylentis, S.A.U. siRNA and their use in methods and compositions for inhibiting the expression of the FLAP gene.
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
EP2933333A1 (en) * 2010-07-08 2015-10-21 Bonac Corporation Single-strand nucleic acid molecule for controlling gene expression
US9181551B2 (en) 2002-02-20 2015-11-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9200278B2 (en) 2010-08-03 2015-12-01 Bonac Corporation Single-stranded nucleic acid molecule having nitrogen-containing alicyclic skeleton
US9243246B2 (en) 2010-08-24 2016-01-26 Sirna Therapeutics, Inc. Single-stranded RNAi agents containing an internal, non-nucleic acid spacer
US9260471B2 (en) 2010-10-29 2016-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
EP3018202A1 (en) 2005-11-11 2016-05-11 Korea Research Institute of Bioscience and Biotechnology E2EPF Ubiquitin carrier protein-von-Hippel-Lindau interaction and uses of thereof
WO2016083624A1 (en) 2014-11-28 2016-06-02 Silence Therapeutics Gmbh Means for inhibiting the expression of edn1
EP2363479B1 (en) 1999-01-30 2016-07-13 Alnylam Europe AG Oligoribonucucleotide for inhibiting the expression of a predefined gene
US9399775B2 (en) 2011-11-18 2016-07-26 Alnylam Pharmaceuticals, Inc. RNAi agents, compositions and methods of use thereof for treating transthyretin (TTR) associated diseases
WO2016149020A1 (en) * 2015-03-17 2016-09-22 Arrowhead Research Corporation Rna interference agents
WO2017079442A1 (en) 2015-11-04 2017-05-11 Icahn School Of Medicine At Mount Sinai Methods of treating tumors and cancer, and identifying candidate subjects for such treatment
US9657294B2 (en) 2002-02-20 2017-05-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
JP2017523788A (en) * 2014-08-07 2017-08-24 財團法人工業技術研究院Industrial Technology Research Institute Small interfering RNA for suppressing galectin-12 expression and / or promoting lipolysis and pharmaceutical composition comprising the same, and for inhibiting galectin-12 expression and / or promoting lipolysis How to
US9796979B2 (en) 2011-03-03 2017-10-24 Quark Pharmaceuticals Inc. Oligonucleotide modulators of the toll-like receptor pathway
WO2017189730A1 (en) 2016-04-26 2017-11-02 Icahn School Of Medicine At Mount Sinai Treatment of hippo pathway mutant tumors and methods of identifying subjects as candidates for treatment
US9879253B2 (en) 2003-12-22 2018-01-30 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of single and double blunt-ended siRNA
EP3276004A2 (en) 2009-06-08 2018-01-31 Quark Pharmaceuticals, Inc. Methods for treating chronic kidney disease
US9885036B2 (en) 2008-07-01 2018-02-06 Daiichi Sankyo Company, Limited Double-stranded polynucleotide
US9914924B2 (en) 2005-08-18 2018-03-13 University Of Massachusetts Methods and compositions for treating neurological disease
US10208307B2 (en) 2015-07-31 2019-02-19 Alnylam Pharmaceuticals, Inc. Transthyretin (TTR) iRNA compositions and methods of use thereof for treating or preventing TTR-associated diseases
WO2019048645A1 (en) * 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
US10238752B2 (en) 2012-05-26 2019-03-26 Bonac Corporation Single-stranded nucleic acid molecule for regulating expression of gene having delivering function
US10364429B2 (en) 2003-06-02 2019-07-30 University Of Massachusetts Methods and compositions for controlling efficacy of RNA silencing
WO2019246262A2 (en) 2018-06-21 2019-12-26 University Of Rochester Methods of treating or inhibiting onset of huntington's disease
WO2019246112A1 (en) 2018-06-18 2019-12-26 University Of Rochester Methods of treating schizophrenia and other neuropsychiatric disorders
US10612020B2 (en) 2013-12-26 2020-04-07 Tokyo Medical University Artificial mimic miRNA for controlling gene expression, and use of same
WO2020123663A1 (en) 2018-12-11 2020-06-18 University Of Rochester Methods of treating schizophrenia and other neuropsychiatric disorders
US10695362B2 (en) 2010-05-14 2020-06-30 Tagcyx Biotechnologies Inc. Stabilization method of functional nucleic acid
WO2020167822A2 (en) 2019-02-13 2020-08-20 University Of Rochester Gene networks that mediate remyelination of the human brain
WO2021011936A2 (en) 2019-07-18 2021-01-21 University Of Rochester Cell-type selective immunoprotection of cells
US10934542B2 (en) 2013-12-27 2021-03-02 Bonac Corporation Artificial match-type miRNA for controlling gene expression and use therefor
WO2021049504A1 (en) 2019-09-10 2021-03-18 第一三共株式会社 Galnac-oligonucleotide conjugate for liver-targeted delivery use, and method for producing same
EP3584319A4 (en) * 2017-02-06 2021-04-14 Nissan Chemical Corporation Single-stranded oligonucleotide
US11027023B2 (en) 2014-12-27 2021-06-08 Bonac Corporation Natural type miRNA for controlling gene expression, and use of same
EP3105331B1 (en) 2014-02-11 2021-06-23 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
US11142769B2 (en) 2015-03-27 2021-10-12 Bonac Corporation Single-stranded nucleic acid molecule having delivery function and gene expression regulating ability
US11530409B2 (en) 2016-01-26 2022-12-20 Nissan Chemical Corporation Single-stranded oligonucleotide
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023150553A1 (en) 2022-02-01 2023-08-10 University Of Rochester Gpr17 promoter-based targeting and transduction of glial progenitor cells
US11806360B2 (en) 2017-09-19 2023-11-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating transthyretin (TTR) mediated amyloidosis
US11959081B2 (en) 2021-08-03 2024-04-16 Alnylam Pharmaceuticals, Inc. Transthyretin (TTR) iRNA compositions and methods of use thereof

Families Citing this family (139)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8273866B2 (en) 2002-02-20 2012-09-25 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SINA)
US20050020525A1 (en) * 2002-02-20 2005-01-27 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US8202979B2 (en) * 2002-02-20 2012-06-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid
US20050054836A1 (en) * 2000-11-09 2005-03-10 Cold Spring Harbor Laboratory Chimeric molecules to modulate gene expression
US7767802B2 (en) 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US20050282188A1 (en) * 2001-05-18 2005-12-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
JP2005515780A (en) 2002-02-01 2005-06-02 セクイター インコーポレイテッド Double stranded oligonucleotide
EP1572902B1 (en) 2002-02-01 2014-06-11 Life Technologies Corporation HIGH POTENCY siRNAS FOR REDUCING THE EXPRESSION OF TARGET GENES
US8232383B2 (en) * 2002-02-20 2012-07-31 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US7928218B2 (en) * 2002-02-20 2011-04-19 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of polycomb group protein EZH2 gene expression using short interfering nucleic acid (siNA)
US20100240730A1 (en) * 2002-02-20 2010-09-23 Merck Sharp And Dohme Corp. RNA Interference Mediated Inhibition of Gene Expression Using Chemically Modified Short Interfering Nucleic Acid (siNA)
US7910724B2 (en) * 2002-02-20 2011-03-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Fos gene expression using short interfering nucleic acid (siNA)
AU2003207708A1 (en) 2002-02-20 2003-09-09 Sirna Therapeutics, Inc. Rna interference mediated inhibition of map kinase genes
US20090099117A1 (en) * 2002-02-20 2009-04-16 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF MYOSTATIN GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US20090093439A1 (en) * 2002-02-20 2009-04-09 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF CHROMOSOME TRANSLOCATION GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
AU2003261449A1 (en) 2002-08-07 2004-02-25 Compositions for rna interference and methods of use thereof
US7956176B2 (en) * 2002-09-05 2011-06-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
AU2003273336A1 (en) * 2002-09-18 2004-04-08 Isis Pharmaceuticals, Inc. Efficient reduction of target rna's by single- and double-stranded oligomeric compounds
US9150606B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
US9150605B2 (en) 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
US9827263B2 (en) * 2002-11-05 2017-11-28 Ionis Pharmaceuticals, Inc. 2′-methoxy substituted oligomeric compounds and compositions for use in gene modulations
DK1606406T4 (en) 2003-03-21 2013-12-16 Santaris Pharma As Short Interfering RNA (siRNA) Analogues
US8575327B2 (en) 2003-06-12 2013-11-05 Alnylam Pharmaceuticals, Inc. Conserved HBV and HCV sequences useful for gene silencing
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
US20050136437A1 (en) * 2003-08-25 2005-06-23 Nastech Pharmaceutical Company Inc. Nanoparticles for delivery of nucleic acids and stable double-stranded RNA
US20050074801A1 (en) * 2003-09-09 2005-04-07 Monia Brett P. Chimeric oligomeric compounds comprising alternating regions of northern and southern conformational geometry
WO2005073378A1 (en) * 2004-01-30 2005-08-11 Santaris Pharma A/S MODIFIED SHORT INTERFERING RNA (MODIFIED siRNA)
US20160230185A1 (en) 2013-03-14 2016-08-11 Monsanto Technology Llc Compositions and methods for controlling diabrotica
EP2471922A1 (en) 2004-05-28 2012-07-04 Asuragen, Inc. Methods and compositions involving microRNA
US20090048192A1 (en) * 2004-06-03 2009-02-19 Isis Pharmaceuticals, Inc. Double Strand Compositions Comprising Differentially Modified Strands for Use in Gene Modulation
SI1781787T1 (en) 2004-08-23 2017-08-31 Sylentis S.A.U. Treatment of eye disorders characterized by an elevated intraocular pressure by sirnas
EP2322616A1 (en) 2004-11-12 2011-05-18 Asuragen, Inc. Methods and compositions involving miRNA and miRNA inhibitor molecules
US20060160110A1 (en) * 2004-12-02 2006-07-20 Takayuki Mizutani Methods of designing small interfering RNAs, antisense polynucleotides, and other hybridizing polynucleotides
WO2006063356A1 (en) * 2004-12-10 2006-06-15 Isis Phamaceuticals, Inc. Regulation of epigenetic control of gene expression
WO2006131925A2 (en) * 2005-06-10 2006-12-14 Quark Pharmaceuticals, Inc. Oligoribonucleotides and methods of use thereof for treatment of fibrotic conditions and other diseases
GB0521351D0 (en) 2005-10-20 2005-11-30 Genomica Sau Modulation of TRPV expression levels
GB0521716D0 (en) 2005-10-25 2005-11-30 Genomica Sau Modulation of 11beta-hydroxysteriod dehydrogenase 1 expression for the treatment of ocular diseases
US8129515B2 (en) * 2006-01-27 2012-03-06 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for the use in modulation of microRNAs
JP2009524419A (en) * 2006-01-27 2009-07-02 サンタリス ファーマ アー/エス LNA modified phosphorothiolated oligonucleotides
EP1989307B1 (en) * 2006-02-08 2012-08-08 Quark Pharmaceuticals, Inc. NOVEL TANDEM siRNAS
WO2007107162A2 (en) * 2006-03-23 2007-09-27 Santaris Pharma A/S Small internally segmented interfering rna
JP2010504350A (en) * 2006-09-19 2010-02-12 アシュラジェン インコーポレイテッド Genes and pathways regulated by miR-200 as targets for therapeutic intervention
US20100292301A1 (en) * 2007-02-28 2010-11-18 Elena Feinstein Novel sirna structures
WO2008143774A2 (en) * 2007-05-01 2008-11-27 University Of Massachusetts Methods and compositions for locating snp heterozygosity for allele specific diagnosis and therapy
KR20100024410A (en) * 2007-05-11 2010-03-05 엔즌 파마슈티칼스, 인코포레이티드 Rna antagonist compounds for the modulation of her3
US20100273854A1 (en) * 2007-06-15 2010-10-28 Hagar Kalinski Compositions and methods for inhibiting nadph oxidase expression
US8361714B2 (en) 2007-09-14 2013-01-29 Asuragen, Inc. Micrornas differentially expressed in cervical cancer and uses thereof
JP5723154B2 (en) 2007-09-19 2015-05-27 アプライド バイオシステムズ リミテッド ライアビリティー カンパニー SiRNA sequence-independent modification format for reducing the influence of off-target phenotype in RNAi and its stabilized form
US20100317714A1 (en) * 2007-11-29 2010-12-16 Suzhou Ribo Life Science Co., Ltd Complex molecule interfering the expression of target genes and its preparing methods
US8071562B2 (en) 2007-12-01 2011-12-06 Mirna Therapeutics, Inc. MiR-124 regulated genes and pathways as targets for therapeutic intervention
US20110105584A1 (en) * 2007-12-12 2011-05-05 Elena Feinstein Rtp80il sirna compounds and methods of use thereof
US8614311B2 (en) 2007-12-12 2013-12-24 Quark Pharmaceuticals, Inc. RTP801L siRNA compounds and methods of use thereof
KR100949791B1 (en) * 2007-12-18 2010-03-30 이동기 Novel siRNA Structure for Minimizing Off-target Effects and Relaxing Saturation of RNAi Machinery and the Use Thereof
WO2009090639A2 (en) * 2008-01-15 2009-07-23 Quark Pharmaceuticals, Inc. Sirna compounds and methods of use thereof
US10131904B2 (en) 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
US8258111B2 (en) 2008-05-08 2012-09-04 The Johns Hopkins University Compositions and methods related to miRNA modulation of neovascularization or angiogenesis
CA2635187A1 (en) 2008-06-05 2009-12-05 The Royal Institution For The Advancement Of Learning/Mcgill University Oligonucleotide duplexes and uses thereof
WO2010008562A2 (en) 2008-07-16 2010-01-21 Recombinetics Methods and materials for producing transgenic animals
WO2010008582A2 (en) 2008-07-18 2010-01-21 Rxi Pharmaceuticals Corporation Phagocytic cell drug delivery system
US9433684B2 (en) 2008-08-19 2016-09-06 Nektar Therapeutics Conjugates of small-interfering nucleic acids
ES2708944T3 (en) * 2008-09-22 2019-04-12 Dicerna Pharmaceuticals Inc Compositions and methods for the specific inhibition of gene expression by DSRNA having modifications
JP2012502991A (en) 2008-09-22 2012-02-02 アールエックスアイ ファーマシューティカルズ コーポレーション RNA interference in dermal applications
EP2447274B1 (en) 2008-10-24 2017-10-04 Ionis Pharmaceuticals, Inc. Oligomeric compounds and methods
AU2009308217B2 (en) 2008-10-24 2016-01-21 Ionis Pharmaceuticals, Inc. 5' and 2' bis-substituted nucleosides and oligomeric compounds prepared therefrom
WO2010059226A2 (en) 2008-11-19 2010-05-27 Rxi Pharmaceuticals Corporation Inhibition of map4k4 through rnai
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
WO2010090762A1 (en) 2009-02-04 2010-08-12 Rxi Pharmaceuticals Corporation Rna duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
EP2408916A2 (en) * 2009-03-19 2012-01-25 Merck Sharp&Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF CONNECTIVE TISSUE GROWTH FACTOR (CTGF) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2010111198A1 (en) 2009-03-23 2010-09-30 Quark Pharmaceuticals, Inc. Compounds compositions and methods of treating cancer and fibrotic diseases
GB0910723D0 (en) 2009-06-22 2009-08-05 Sylentis Sau Novel drugs for inhibition of gene expression
WO2011035065A1 (en) 2009-09-17 2011-03-24 Nektar Therapeutics Monoconjugated chitosans as delivery agents for small interfering nucleic acids
WO2011034811A1 (en) 2009-09-17 2011-03-24 Sigma-Aldrich Co. Short rna mimetics
CN102666856B (en) 2009-11-08 2016-04-06 夸克制药公司 Be directed to the purposes of double-stranded RNA compound in the medicine manufacturing treatment neuropathic pain of RhoA target gene
KR101692063B1 (en) 2009-12-09 2017-01-03 닛토덴코 가부시키가이샤 MODULATION OF hsp47 EXPRESSION
WO2011119871A1 (en) 2010-03-24 2011-09-29 Rxi Phrmaceuticals Corporation Rna interference in ocular indications
BR112012024049A2 (en) 2010-03-24 2017-03-01 Rxi Pharmaceuticals Corp rna interference on dermal and fibrotic indications
WO2011119852A1 (en) 2010-03-24 2011-09-29 Rxi Pharmaceuticals Corporation Reduced size self-delivering rnai compounds
EP2625186B1 (en) 2010-04-28 2016-07-27 Ionis Pharmaceuticals, Inc. 5' modified nucleosides and oligomeric compounds prepared therefrom
CN103154014B (en) 2010-04-28 2015-03-25 Isis制药公司 Modified nucleosides, modified nucleosides-like and oligomeric compounds prepared therefrom
EP2390327A1 (en) 2010-05-27 2011-11-30 Sylentis S.A. siRNA and their use in methods and compositions for the treatment and/or prevention of eye conditions
CA2818662C (en) 2010-10-22 2021-07-06 Sungkyunkwan University Foundation For Corporate Collaboration Nucleic acid molecule inducing rna interference, and uses thereof
CA2828002A1 (en) 2011-03-03 2012-09-07 Quark Pharmaceuticals, Inc. Compositions and methods for treating lung disease and injury
US10196637B2 (en) 2011-06-08 2019-02-05 Nitto Denko Corporation Retinoid-lipid drug carrier
TWI658830B (en) 2011-06-08 2019-05-11 日東電工股份有限公司 Retinoid-liposomes for enhancing modulation of hsp47 expression
TW202244278A (en) 2011-06-30 2022-11-16 美商艾羅海德製藥公司 Compositions and methods for inhibiting gene expression of hepatitis b virus
US9644241B2 (en) 2011-09-13 2017-05-09 Interpace Diagnostics, Llc Methods and compositions involving miR-135B for distinguishing pancreatic cancer from benign pancreatic disease
KR102011048B1 (en) 2011-10-18 2019-08-14 다이서나 파마수이티컬, 인크. Amine cationic lipids and uses thereof
DK2853597T3 (en) * 2012-05-22 2019-04-08 Olix Pharmaceuticals Inc RNA INTERFERENCE-INducing NUCLEIC ACID MOLECULES WITH CELL PENETENING EQUIPMENT AND USE THEREOF
WO2014018375A1 (en) 2012-07-23 2014-01-30 Xenon Pharmaceuticals Inc. Cyp8b1 and uses thereof in therapeutic and diagnostic methods
GB201215857D0 (en) 2012-09-05 2012-10-24 Sylentis Sau siRNA and their use in methods and compositions for the treatment and/or prevention of eye conditions
WO2014037686A1 (en) 2012-09-05 2014-03-13 Sylentis S.A.U. Sirna and their use in methods and compositions for the treatment and/or prevention of eye conditions
CA3120574A1 (en) 2013-03-14 2014-09-25 Dicerna Pharmaceuticals, Inc. Process for formulating an anionic agent
WO2015051366A2 (en) * 2013-10-04 2015-04-09 Novartis Ag Novel formats for organic compounds for use in rna interference
RU2744194C2 (en) 2013-12-02 2021-03-03 Фио Фармасьютикалс Корп Cancer immunotherapy
US20160333346A1 (en) * 2014-01-17 2016-11-17 Kyowa Hakko Kirin Co., Ltd. Nucleic acid capable of inhibiting expression of beta-2gpi
US10011837B2 (en) 2014-03-04 2018-07-03 Sylentis Sau SiRNAs and their use in methods and compositions for the treatment and/or prevention of eye conditions
CA2947270A1 (en) 2014-04-28 2015-11-05 Rxi Pharmaceuticals Corporation Methods for treating cancer using nucleic acids targeting mdm2 or mycn
US10900039B2 (en) 2014-09-05 2021-01-26 Phio Pharmaceuticals Corp. Methods for treating aging and skin disorders using nucleic acids targeting Tyr or MMP1
FR3026409B1 (en) * 2014-09-26 2018-03-30 Centre National De La Recherche Scientifique (Cnrs) METHOD FOR SCREENING INTERFERENT MOLECULES
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2016094845A2 (en) 2014-12-12 2016-06-16 Woolf Tod M Compositions and methods for editing nucleic acids in cells utilizing oligonucleotides
WO2017007825A1 (en) 2015-07-06 2017-01-12 Rxi Pharmaceuticals Corporation Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
CN108135923B (en) 2015-07-06 2021-03-02 菲奥医药公司 Nucleic acid molecules targeting superoxide dismutase 1(SOD1)
KR20180038465A (en) 2015-08-07 2018-04-16 애로우헤드 파마슈티컬스 인코포레이티드 RNAi therapy for hepatitis B virus infection
MA44908A (en) 2015-09-08 2018-07-18 Sylentis Sau ARNSI MOLECULES AND THEIR USE IN PROCESSES AND COMPOSITIONS TO INHIBIT NRARP GENE EXPRESSION
JP2018531037A (en) 2015-10-19 2018-10-25 アールエックスアイ ファーマシューティカルズ コーポレーション Reduced size self-delivering nucleic acid compounds targeting long non-coding RNAs
KR20180071362A (en) 2015-11-16 2018-06-27 올릭스 주식회사 Treatment of age-related macular degeneration with RNA complex targeting MyD88 or TLR3
KR20180104692A (en) 2016-02-02 2018-09-21 올릭스 주식회사 Treatment of angiogenesis related diseases using RNA complexes targeting ANGPT2 and PDGFB
WO2017134525A1 (en) 2016-02-02 2017-08-10 Olix Pharmaceuticals, Inc. TREATMENT OF ATOPIC DERMATITIS AND ASTHMA USING RNA COMPLEXES THAT TARGET lL4Rα, TRPA1, OR F2RL1
MA45328A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc NUCLEIC ACID-POLYPEPTIDE COMPOSITIONS AND USES THEREOF
CN109072238B (en) 2016-04-11 2022-05-24 奥利克斯医药有限公司 Treatment of idiopathic pulmonary fibrosis using RNA complexes targeting connective tissue growth factor
WO2017181088A1 (en) 2016-04-14 2017-10-19 University Of Florida Research Foundation, Incorporated Use of mir-223-3p as a cancer therapeutic and method for treating cancer using the same
EP3452055A4 (en) 2016-05-06 2019-11-06 Tod M. Woolf Improved methods for genome editing with and without programmable nucleases
KR101916652B1 (en) 2016-06-29 2018-11-08 올릭스 주식회사 Compounds improving RNA interference of small interfering RNA and use thereof
JOP20170161A1 (en) 2016-08-04 2019-01-30 Arrowhead Pharmaceuticals Inc RNAi Agents for Hepatitis B Virus Infection
EP3315125A1 (en) 2016-10-31 2018-05-02 Silence Therapeutics (London) Ltd Lipid nanoparticle formulation
AU2017368050A1 (en) 2016-11-29 2019-06-20 Puretech Lyt, Inc. Exosomes for delivery of therapeutic agents
MX2019008199A (en) 2017-01-06 2019-11-25 Avidity Biosciences Llc Nucleic acid-polypeptide compositions and methods of inducing exon skipping.
WO2018146557A2 (en) 2017-02-10 2018-08-16 Dong Ki Lee Long double-stranded rna for rna interference
US11324820B2 (en) 2017-04-18 2022-05-10 Alnylam Pharmaceuticals, Inc. Methods for the treatment of subjects having a hepatitis b virus (HBV) infection
GB201711809D0 (en) 2017-07-21 2017-09-06 Governors Of The Univ Of Alberta Antisense oligonucleotide
EP3679139B1 (en) * 2017-09-08 2022-11-02 MiNA Therapeutics Limited Stabilized hnf4a sarna compositions and methods of use
MX2020004898A (en) * 2017-11-13 2020-10-05 Silence Therapeutics Gmbh Nucleic acids for inhibiting expression of a target gene comprising phosphorodithioate linkages.
EP3720448A4 (en) 2017-12-06 2021-11-03 Avidity Biosciences, Inc. Compositions and methods of treating muscle atrophy and myotonic dystrophy
US11648260B2 (en) 2018-03-29 2023-05-16 Technion Research And Development Foundation Limitted Vesicles comprising a PTEN inhibitor and uses of same
SG11202100715WA (en) 2018-08-13 2021-02-25 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) dsRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
JP2022513482A (en) 2018-12-21 2022-02-08 アビディティー バイオサイエンシーズ,インク. Anti-transferrin receptor antibody and its use
CN115666589A (en) 2020-03-19 2023-01-31 艾维迪提生物科学公司 Compositions and methods for treating facioscapulohumeral muscular dystrophy
CN115697418A (en) 2020-03-27 2023-02-03 艾维迪提生物科学公司 Compositions and methods for treating muscular dystrophy
WO2021255262A1 (en) 2020-06-19 2021-12-23 Sylentis Sau siRNA AND COMPOSITIONS FOR PROPHYLACTIC AND THERAPEUTIC TREATMENT OF VIRUS DISEASES
US11459567B2 (en) 2020-06-24 2022-10-04 Patricia Virginia Elizalde Specific siRNA molecules, composition and use thereof for the treatment of triple negative breast cancer
EP4015634A1 (en) 2020-12-15 2022-06-22 Sylentis, S.A.U. Sirna and compositions for prophylactic and therapeutic treatment of virus diseases
WO2022162155A1 (en) 2021-01-30 2022-08-04 E-Therapeutics Plc Nucleic acids containing abasic nucleotides
AU2022214281A1 (en) 2021-01-30 2023-07-27 E-Therapeutics Plc Conjugated oligonucleotide compounds, methods of making and uses thereof
WO2022162161A1 (en) 2021-01-30 2022-08-04 E-Therapeutics Plc Conjugated oligonucleotide compounds, methods of making and uses thereof
WO2022162157A1 (en) 2021-01-30 2022-08-04 E-Therapeutics Plc Conjugated oligonucleotide compounds, methods of making and uses thereof
CA3204317A1 (en) 2021-01-30 2022-08-04 Ahmad Ali MORTAZAVI Conjugated oligonucleotide compounds, methods of making and uses thereof
CA3231330A1 (en) 2021-09-16 2023-03-23 Avidity Biosciences, Inc. Compositions and methods of treating facioscapulohumeral muscular dystrophy

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995013834A1 (en) * 1993-11-16 1995-05-26 Genta, Incorporated Chimeric oligonucleoside compounds
WO1997041141A1 (en) * 1996-05-01 1997-11-06 Thomas Jefferson University Methods and compounds for curing diseases caused by mutations
WO2000044895A1 (en) * 1999-01-30 2000-08-03 Roland Kreutzer Method and medicament for inhibiting the expression of a defined gene
WO2002044321A2 (en) * 2000-12-01 2002-06-06 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Rna interference mediating small rna molecules
WO2002055693A2 (en) * 2001-01-09 2002-07-18 Ribopharma Ag Method for inhibiting the expression of a target gene
WO2003070912A2 (en) * 2001-06-06 2003-08-28 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF EPIDERMAL GROWTH FACTOR RECEPTOR GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)

Family Cites Families (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB992744A (en) 1962-01-18 1965-05-19 Dennis Harold Ward Plastic containers and lifting means therefore
US4217294A (en) 1979-03-30 1980-08-12 Union Carbide Corporation Method for producing mercaptan-containing organosilicon compounds
EP0942000B1 (en) 1989-10-24 2004-06-23 Isis Pharmaceuticals, Inc. 2'-Modified oligonucleotides
US6153737A (en) 1990-01-11 2000-11-28 Isis Pharmaceuticals, Inc. Derivatized oligonucleotides having improved uptake and other properties
US6399754B1 (en) 1991-12-24 2002-06-04 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides
US6005087A (en) 1995-06-06 1999-12-21 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US5914396A (en) 1990-01-11 1999-06-22 Isis Pharmaceuticals, Inc. 2'-O-modified nucleosides and phosphoramidites
US5670633A (en) 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5955589A (en) * 1991-12-24 1999-09-21 Isis Pharmaceuticals Inc. Gapped 2' modified oligonucleotides
US5872232A (en) 1990-01-11 1999-02-16 Isis Pharmaceuticals Inc. 2'-O-modified oligonucleotides
US5811533A (en) 1990-06-11 1998-09-22 Nexstar Pharmaceuticals, Inc. High-affinity oligonucleotide ligands to vascular endothelial growth factor (VEGF)
US5378825A (en) 1990-07-27 1995-01-03 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogs
CA2093664C (en) 1990-10-12 2003-07-29 Fritz Eckstein Modified ribozymes
FR2675803B1 (en) 1991-04-25 1996-09-06 Genset Sa CLOSED, ANTISENSE AND SENSE OLIGONUCLEOTIDES AND THEIR APPLICATIONS.
FR2678512B1 (en) 1991-07-03 1995-06-30 Novacell INTERNALIZING MACHINE.
EP1695979B1 (en) 1991-12-24 2011-07-06 Isis Pharmaceuticals, Inc. Gapped modified oligonucleotides
HUT69981A (en) * 1992-07-02 1995-09-28 Hybridon Inc Self-stabilized oligonucleotides as therapeutic agents
CA2105364A1 (en) * 1993-09-01 1995-03-02 Eric T. Kool Stem-loop oligonucleotides containing parallel and antiparallel binding domains
DE4338704A1 (en) 1993-11-12 1995-05-18 Hoechst Ag Stabilized oligonucleotides and their use
US6060456A (en) * 1993-11-16 2000-05-09 Genta Incorporated Chimeric oligonucleoside compounds
GB9425118D0 (en) 1994-12-13 1995-02-08 Henkel Kgaa Two component natural hair colorant
CA2207593A1 (en) 1994-12-13 1996-06-20 John Gustofson Method and reagent for treatment of arthritic conditions, induction of graft tolerance and reversal of immune responses
US5716824A (en) 1995-04-20 1998-02-10 Ribozyme Pharmaceuticals, Inc. 2'-O-alkylthioalkyl and 2-C-alkylthioalkyl-containing enzymatic nucleic acids (ribozymes)
DE19618797C2 (en) 1996-05-10 2000-03-23 Bertling Wolf Vehicle for the transport of molecular substances
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
DE19631919C2 (en) 1996-08-07 1998-07-16 Deutsches Krebsforsch Anti-sense RNA with secondary structure
US6284267B1 (en) 1996-08-14 2001-09-04 Nutrimed Biotech Amphiphilic materials and liposome formulations thereof
GB9710475D0 (en) 1997-05-21 1997-07-16 Zeneca Ltd Gene silencing
US6133246A (en) * 1997-08-13 2000-10-17 Isis Pharmaceuticals Inc. Antisense oligonucleotide compositions and methods for the modulation of JNK proteins
GB9720148D0 (en) 1997-09-22 1997-11-26 Innes John Centre Innov Ltd Gene silencing materials and methods
US6129912A (en) 1997-09-26 2000-10-10 Uab Research Foundation Polyethylene glycol-protein compositions which reduce the antigenic display of red blood cells
US6617438B1 (en) * 1997-11-05 2003-09-09 Sirna Therapeutics, Inc. Oligoribonucleotides with enzymatic activity
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
EP1071762A4 (en) 1998-03-20 2003-09-24 Benitec Australia Ltd Control of gene expression
EP1068311B2 (en) 1998-04-08 2020-12-09 Commonwealth Scientific and Industrial Research Organisation Methods and means for obtaining modified phenotypes
EP1071753A2 (en) 1998-04-20 2001-01-31 Ribozyme Pharmaceuticals, Inc. Nucleic acid molecules with novel chemical compositions capable of modulating gene expression
JP2002512794A (en) 1998-04-29 2002-05-08 リボザイム・ファーマシューティカルズ・インコーポレーテッド Nucleotide triphosphates and their incorporation into ribozymes
AR020078A1 (en) 1998-05-26 2002-04-10 Syngenta Participations Ag METHOD FOR CHANGING THE EXPRESSION OF AN OBJECTIVE GENE IN A PLANT CELL
GB9827152D0 (en) 1998-07-03 1999-02-03 Devgen Nv Characterisation of gene function using double stranded rna inhibition
CA2361201A1 (en) 1999-01-28 2000-08-03 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded rna
US6200924B1 (en) 1999-01-29 2001-03-13 E. I. Du Pont De Nemours And Company Porous highly fluorinated acidic polymer catalyst
JP2002542263A (en) 1999-04-21 2002-12-10 ワイス Methods and compositions for inhibiting the function of a polynucleotide sequence
DE60026313D1 (en) 1999-07-23 2006-04-27 Uutech Ltd SENSITIZATION OF RED BLOOD BODIES AGAINST ULTRASOUND BY IMPACT OF AN ELECTRIC FIELD
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
US7829693B2 (en) 1999-11-24 2010-11-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
WO2003070918A2 (en) 2002-02-20 2003-08-28 Ribozyme Pharmaceuticals, Incorporated Rna interference by modified short interfering nucleic acid
EP2345742B1 (en) 2000-03-30 2014-06-11 The Whitehead Institute for Biomedical Research RNA sequence-specific mediators of RNA interference
EP1309706A2 (en) 2000-08-19 2003-05-14 Axordia Limited Modulation of stem cell differentiation
US20030190635A1 (en) 2002-02-20 2003-10-09 Mcswiggen James A. RNA interference mediated treatment of Alzheimer's disease using short interfering RNA
US20060142226A1 (en) 2001-05-18 2006-06-29 Sirna Therapeutics, Inc. RNA interference mediated inhibition of cholesteryl ester transfer protein (CETP) gene expression using short interfering nucleic acid (siNA)
EP1390472A4 (en) 2001-05-29 2004-11-17 Sirna Therapeutics Inc Nucleic acid treatment of diseases or conditions related to levels of ras, her2 and hiv
US7074863B2 (en) 2001-06-22 2006-07-11 Borealis Technology Oy Metallocene catalysts containing an idenyl moiety substituted at the 4,-5,-6- or 7-position by a siloxy or germyloxy group
DE10163098B4 (en) 2001-10-12 2005-06-02 Alnylam Europe Ag Method for inhibiting the replication of viruses
US20030135033A1 (en) 2002-01-04 2003-07-17 Anke Klippel-Giese Compounds and methods for the identification and/ or validation of a target
DE10202419A1 (en) 2002-01-22 2003-08-07 Ribopharma Ag Method of inhibiting expression of a target gene resulting from chromosome aberration
CA2474414C (en) * 2002-02-01 2018-03-06 Mcgill University Oligonucleotides comprising alternating segments and uses thereof
EP1572902B1 (en) 2002-02-01 2014-06-11 Life Technologies Corporation HIGH POTENCY siRNAS FOR REDUCING THE EXPRESSION OF TARGET GENES
AU2003207708A1 (en) * 2002-02-20 2003-09-09 Sirna Therapeutics, Inc. Rna interference mediated inhibition of map kinase genes
US7148342B2 (en) 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
DE60310944T3 (en) 2002-08-05 2017-08-03 Silence Therapeutics Gmbh OTHER NEW FORMS OF INTERFERING RNS MOLECULES
EP1393742A1 (en) 2002-08-14 2004-03-03 atugen AG Use of protein kinase N beta
KR101295939B1 (en) 2002-08-14 2013-09-09 사일런스 테라퓨틱스 아게 Use of protein kinase n beta
PT1551868E (en) 2002-10-18 2009-04-06 Silence Therapeutics Ag Factor involved in metastasis and uses thereof
US7289445B2 (en) * 2002-11-25 2007-10-30 Intel Corporation Managing a protocol control block cache in a network device
DK3604537T3 (en) 2003-06-13 2022-02-28 Alnylam Europe Ag Double-stranded ribonucleic acid with increased efficiency in an organism
CA2530125A1 (en) 2003-06-27 2005-01-06 Atugen Ag Use of double-stranded ribonucleic acid for inducing cell lysis
EP1735009A4 (en) 2004-03-12 2011-03-30 Alnylam Pharmaceuticals Inc iRNA AGENTS TARGETING VEGF
US20070265220A1 (en) 2004-03-15 2007-11-15 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US20050217329A1 (en) * 2004-04-02 2005-10-06 Wen-Kwei Chang Anti-drilling structure of a latch
HUE036916T2 (en) 2004-05-05 2018-08-28 Silence Therapeutics Gmbh Lipids, lipid complexes and use thereof
EP1765847A4 (en) 2004-05-27 2010-10-20 Alnylam Pharmaceuticals Inc Nuclease resistant double-stranded ribonucleic acid
AP2007003921A0 (en) 2004-08-16 2007-02-28 Atugen Ag Therapeutic uses of inhibitors of rtp801
EP1830888B1 (en) 2004-12-27 2015-08-05 Silence Therapeutics GmbH Lipid complexes coated with peg and their use
US20070027097A1 (en) 2005-07-28 2007-02-01 Lewis David L Small interfering RNA with improved activity
MX2008005508A (en) 2005-10-28 2008-11-18 Topigen Pharmaceuticals Inc Small interfering ribonucleic acid duplexes comprising arabinose modified nucleotides.
EP2395012B8 (en) 2005-11-02 2018-06-06 Arbutus Biopharma Corporation Modified siRNA molecules and uses thereof
NL2000439C2 (en) 2006-01-20 2009-03-16 Quark Biotech Therapeutic applications of inhibitors of RTP801.
US7541994B2 (en) 2006-05-17 2009-06-02 Raytheon Company Refractive compact range
US8614309B2 (en) 2007-10-03 2013-12-24 Quark Pharmaceuticals, Inc. Double-stranded RNA directed to CASP2 and methods of use thereof

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995013834A1 (en) * 1993-11-16 1995-05-26 Genta, Incorporated Chimeric oligonucleoside compounds
WO1997041141A1 (en) * 1996-05-01 1997-11-06 Thomas Jefferson University Methods and compounds for curing diseases caused by mutations
WO2000044895A1 (en) * 1999-01-30 2000-08-03 Roland Kreutzer Method and medicament for inhibiting the expression of a defined gene
WO2002044321A2 (en) * 2000-12-01 2002-06-06 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Rna interference mediating small rna molecules
WO2002055693A2 (en) * 2001-01-09 2002-07-18 Ribopharma Ag Method for inhibiting the expression of a target gene
WO2003070912A2 (en) * 2001-06-06 2003-08-28 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF EPIDERMAL GROWTH FACTOR RECEPTOR GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CZAUDERNA FRANK ET AL: "Structural variations and stabilising modifications of synthetic siRNAs in mammalian cells." NUCLEIC ACIDS RESEARCH, vol. 31, no. 11, 1 June 2003 (2003-06-01), pages 2705-2716, XP002270732 ISSN: 0305-1048 *
PARRISH S ET AL: "Functional anatomy of a dsRNA trigger: Differential requirement for the two trigger strands in RNA interference" MOLECULAR CELL, vol. 6, no. 5, November 2000 (2000-11), pages 1077-1087, XP002226298 ISSN: 1097-2765 *
STERNBERGER M ET AL: "GeneBlocs are powerful tools to study and delineate signal transduction processes that regulate cell growth and transformation" ANTISENSE & NUCLEIC ACID DRUG DEVELOPMENT, vol. 12, no. 3, June 2002 (2002-06), pages 131-143, XP002226299 ISSN: 1087-2906 cited in the application *

Cited By (274)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US7695902B2 (en) 1996-06-06 2010-04-13 Isis Pharmaceuticals, Inc. Oligoribonucleotides and ribonucleases for cleaving RNA
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US9902955B2 (en) 1999-01-30 2018-02-27 Alnylam Pharmaceuticals, Inc. Method and medicament for inhibiting the expression of a given gene
EP1798285B1 (en) 1999-01-30 2017-03-15 Alnylam Europe AG Methods and medicament for inhibition the expression of a defined gene
EP2363479B1 (en) 1999-01-30 2016-07-13 Alnylam Europe AG Oligoribonucucleotide for inhibiting the expression of a predefined gene
US8394628B2 (en) 2000-03-30 2013-03-12 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US8552171B2 (en) 2000-03-30 2013-10-08 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US9012621B2 (en) 2000-03-30 2015-04-21 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA sequence-specific mediators of RNA interference
US8790922B2 (en) 2000-03-30 2014-07-29 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA sequence-specific mediators of RNA interference
US8420391B2 (en) 2000-03-30 2013-04-16 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US9012138B2 (en) 2000-03-30 2015-04-21 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA sequence-specific mediators of RNA interference
US8632997B2 (en) 2000-03-30 2014-01-21 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US9193753B2 (en) 2000-03-30 2015-11-24 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US10472625B2 (en) 2000-03-30 2019-11-12 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA sequence-specific mediators of RNA interference
US8742092B2 (en) 2000-03-30 2014-06-03 University Of Massachusetts RNA sequence-specific mediators of RNA interference
US8329463B2 (en) 2000-12-01 2012-12-11 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8445237B2 (en) 2000-12-01 2013-05-21 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8993745B2 (en) 2000-12-01 2015-03-31 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8895718B2 (en) 2000-12-01 2014-11-25 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US10633656B2 (en) 2000-12-01 2020-04-28 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. RNA interference mediating small RNA molecules
US8372968B2 (en) 2000-12-01 2013-02-12 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8895721B2 (en) 2000-12-01 2014-11-25 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8933044B2 (en) 2000-12-01 2015-01-13 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8853384B2 (en) 2000-12-01 2014-10-07 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8765930B2 (en) 2000-12-01 2014-07-01 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8362231B2 (en) 2000-12-01 2013-01-29 Max-Planck-Gesellschaft zur Föderung der Wissenschaften E.V. RNA interference mediating small RNA molecules
US8796016B2 (en) 2000-12-01 2014-08-05 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US8778902B2 (en) 2000-12-01 2014-07-15 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. RNA interference mediating small RNA molecules
US9732344B2 (en) 2002-02-20 2017-08-15 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US10351852B2 (en) 2002-02-20 2019-07-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US10000754B2 (en) 2002-02-20 2018-06-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US10889815B2 (en) 2002-02-20 2021-01-12 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9181551B2 (en) 2002-02-20 2015-11-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9957517B2 (en) 2002-02-20 2018-05-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9657294B2 (en) 2002-02-20 2017-05-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9738899B2 (en) 2002-02-20 2017-08-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9771588B2 (en) 2002-02-20 2017-09-26 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US10662428B2 (en) 2002-02-20 2020-05-26 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US10329568B2 (en) 2002-08-05 2019-06-25 Silence Therapeutics Gmbh Interfering RNA molecules
US7452987B2 (en) 2002-08-05 2008-11-18 Silence Therapeutics Aktiengesellschaft (Ag) Interfering RNA molecules
US11578328B2 (en) 2002-08-05 2023-02-14 Silence Therapeutics Gmbh Interfering RNA molecules
US8933215B2 (en) 2002-08-05 2015-01-13 Silence Therapeutics Aktiengesellschaft (Ag) Interfering RNA molecules
US9222092B2 (en) 2002-08-05 2015-12-29 Silence Therapeutics Gmbh Interfering RNA molecules
US10323246B2 (en) 2002-08-05 2019-06-18 Silence Therapeutics Gmbh Interfering RNA molecules
US8324370B2 (en) 2002-08-05 2012-12-04 Silence Therapeutics Aktiengesellschaft (Ag) Interfering RNA molecules
US9695423B2 (en) 2002-08-05 2017-07-04 Silence Therapeutics Gmbh Interfering RNA molecules
US10266829B2 (en) 2002-08-05 2019-04-23 Silence Therapeutics Gmbh Interfering RNA molecules
US10774332B2 (en) 2002-08-05 2020-09-15 Silence Therapeutics Gmbh Interfering RNA molecules
US9790501B2 (en) 2002-08-05 2017-10-17 Silence Therapeutics Gmbh Interfering RNA molecules
US9790505B2 (en) 2002-08-05 2017-10-17 Silence Therapeutics Gmbh Interfering RNA molecules
US9783802B2 (en) 2002-08-05 2017-10-10 Silence Therapeutics Gmbh Interfering RNA molecules
US7893245B2 (en) 2002-08-05 2011-02-22 Silence Therapeutics Aktiengesellschaft (Ag) Interfering RNA molecules
US9758784B1 (en) 2002-08-05 2017-09-12 Silence Therapeutics Gmbh Interfering RNA molecules
US7892793B2 (en) 2002-11-04 2011-02-22 University Of Massachusetts Allele-specific RNA interference
US8604183B2 (en) 2002-11-05 2013-12-10 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
US7834171B2 (en) 2003-04-02 2010-11-16 Dharmacon, Inc. Modified polynucleotides for reducing off-target effects in RNA interference
JP2010172344A (en) * 2003-04-02 2010-08-12 Dharmacon Inc Modified polynucleotide for use in rna interference
US10604754B2 (en) 2003-06-02 2020-03-31 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
US8304530B2 (en) 2003-06-02 2012-11-06 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
US8309705B2 (en) 2003-06-02 2012-11-13 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
US8309704B2 (en) 2003-06-02 2012-11-13 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNAi
US10364429B2 (en) 2003-06-02 2019-07-30 University Of Massachusetts Methods and compositions for controlling efficacy of RNA silencing
US11459562B2 (en) 2003-06-02 2022-10-04 University Of Massachusetts Methods and compositions for controlling efficacy of RNA silencing
US8329892B2 (en) 2003-06-02 2012-12-11 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
US7750144B2 (en) 2003-06-02 2010-07-06 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
US9121018B2 (en) 2003-06-02 2015-09-01 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
EP1644475A4 (en) * 2003-06-20 2009-06-03 Isis Pharmaceuticals Inc Double stranded compositions comprising a 3'-endo modified strand for use in gene modulation
EP1644475A2 (en) * 2003-06-20 2006-04-12 Isis Pharmaceuticals, Inc. DOUBLE STRANDED COMPOSITIONS COMPRISING A 3’-ENDO MODIFIED STRAND FOR USE IN GENE MODULATION
EP2338995A3 (en) * 2003-08-28 2012-01-11 Novartis AG Interfering RNA duplex having blunt-ends and 3'-modifications
US8097716B2 (en) 2003-08-28 2012-01-17 Novartis Ag Interfering RNA duplex having blunt-ends and 3′-modifications
WO2005021749A1 (en) * 2003-08-28 2005-03-10 Novartis Ag Interfering rna duplex having blunt-ends and 3’-modifications
US11299734B2 (en) 2003-09-12 2022-04-12 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
US7947658B2 (en) 2003-09-12 2011-05-24 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
US10344277B2 (en) 2003-09-12 2019-07-09 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
US9434943B2 (en) 2003-09-12 2016-09-06 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
US8680063B2 (en) 2003-09-12 2014-03-25 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
US10385339B2 (en) 2003-12-22 2019-08-20 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of single and double blunt-ended siRNA
US9879253B2 (en) 2003-12-22 2018-01-30 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of single and double blunt-ended siRNA
EP2330111A2 (en) 2004-01-30 2011-06-08 Quark Pharmaceuticals, Inc. Oligoribonucleotides and methods of use thereof for treatment of fibrotic conditions and other diseases
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
JP2007531520A (en) * 2004-04-01 2007-11-08 ダーマコン, インコーポレイテッド Modified polynucleotides for reducing off-target effects in RNA interference
KR101147147B1 (en) 2004-04-01 2012-05-25 머크 샤프 앤드 돔 코포레이션 Modified polynucleotides for reducing off-target effects in rna interference
AU2005230850B2 (en) * 2004-04-01 2009-09-17 Dharmacon, Inc. Modified polynucleotides for reducing off-target effects in RNA interference
WO2005097992A3 (en) * 2004-04-01 2006-02-02 Dharmacon Inc Modified polynucleotides for reducing off-target effects in rna interference
EP1766071A4 (en) * 2004-06-03 2009-11-11 Isis Pharmaceuticals Inc Double strand compositions comprising differentially modified strands for use in gene modulation
EP1765416A2 (en) * 2004-06-03 2007-03-28 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
EP1766052A4 (en) * 2004-06-03 2009-12-16 Isis Pharmaceuticals Inc Chimeric gapped oligomeric compositions
EP1765416A4 (en) * 2004-06-03 2010-03-24 Isis Pharmaceuticals Inc Double strand compositions comprising differentially modified strands for use in gene modulation
EP1765415A4 (en) * 2004-06-03 2010-03-24 Isis Pharmaceuticals Inc Oligomeric compounds that facilitate risc loading
EP1766071A2 (en) * 2004-06-03 2007-03-28 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
AU2005252663B2 (en) * 2004-06-03 2011-07-07 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
EP1765415A2 (en) * 2004-06-03 2007-03-28 Isis Pharmaceuticals, Inc. Oligomeric compounds that facilitate risc loading
WO2005121370A3 (en) * 2004-06-03 2006-05-26 Isis Pharmaceuticals Inc Oligomeric compounds that facilitate risc loading
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
JP2008501694A (en) * 2004-06-03 2008-01-24 アイシス ファーマシューティカルズ、インク. Double-stranded composition having individually modified strands for use in gene regulation
JP2008501335A (en) * 2004-06-03 2008-01-24 アイシス ファーマシューティカルズ、インク. Chimeric gapped oligomer composition
JP2008501693A (en) * 2004-06-03 2008-01-24 アイシス ファーマシューティカルズ、インク. Double-stranded composition with individually regulated strands for use in gene regulation
EP1766052A1 (en) * 2004-06-03 2007-03-28 Isis Pharmaceuticals, Inc. Chimeric gapped oligomeric compositions
WO2005121372A3 (en) * 2004-06-03 2006-04-13 Isis Pharmaceuticals Inc Double strand compositions comprising differentially modified strands for use in gene modulation
US8642571B2 (en) 2004-08-06 2014-02-04 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
US8168607B2 (en) 2004-08-06 2012-05-01 Quark Pharmaceuticals Inc. Methods of treating eye diseases in diabetic patients
US7741299B2 (en) 2004-08-16 2010-06-22 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
EP1791568A2 (en) * 2004-08-16 2007-06-06 Quark Biotech, Inc. Therapeutic uses of inhibitors of rtp801
CN101123994B (en) * 2004-08-16 2012-11-14 夸克医药公司 Therapeutic uses of inhibitors of rtp801
US8309532B2 (en) 2004-08-16 2012-11-13 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
EP1791568A4 (en) * 2004-08-16 2008-12-10 Quark Pharmaceuticals Inc Therapeutic uses of inhibitors of rtp801
KR101094617B1 (en) * 2004-08-16 2011-12-15 사일런스 테라퓨틱스 아게 Therapeutic uses of inhibitors of rtp801
NO342934B1 (en) * 2004-08-16 2018-09-03 Silence Therapeutics Ag THERAPEUTIC USES OF RTP801 INHIBITORS
EP2319925A3 (en) * 2004-08-16 2011-09-21 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
EP2319925A2 (en) 2004-08-16 2011-05-11 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
US8765699B2 (en) 2004-09-28 2014-07-01 Quark Pharmaceuticals, Inc. Oligoribonucleotides and methods of use thereof for treatment of alopecia, acute renal failure and other diseases
EP1799269A2 (en) * 2004-09-28 2007-06-27 Quark Biotech, Inc. Oligoribonucleotides and methods of use thereof for treatment of alopecia, acute renal failure and other diseases
US8148342B2 (en) 2004-09-28 2012-04-03 Quark Pharmaceuticals Inc. Oligoribonucleotides and methods of use thereof for treatment of alopecia, acute renal failure and other diseases
EP1799269A4 (en) * 2004-09-28 2012-01-18 Quark Pharmaceuticals Inc Oligoribonucleotides and methods of use thereof for treatment of alopecia, acute renal failure and other diseases
US7842674B2 (en) 2004-09-28 2010-11-30 Quark Pharmaceuticals Inc. Methods of treatment of acute renal failure
WO2006035434A2 (en) 2004-09-28 2006-04-06 Quark Biotech, Inc. Oligoribonucleotides and methods of use thereof for treatment of alopecia, acute renal failure and other diseases
US7923207B2 (en) 2004-11-22 2011-04-12 Dharmacon, Inc. Apparatus and system having dry gene silencing pools
US7923206B2 (en) 2004-11-22 2011-04-12 Dharmacon, Inc. Method of determining a cellular response to a biological agent
US7935811B2 (en) 2004-11-22 2011-05-03 Dharmacon, Inc. Apparatus and system having dry gene silencing compositions
US9914924B2 (en) 2005-08-18 2018-03-13 University Of Massachusetts Methods and compositions for treating neurological disease
US8309533B2 (en) 2005-09-30 2012-11-13 University Of Massachusetts Allele-specific RNA interference
WO2007039268A1 (en) * 2005-10-03 2007-04-12 Novartis Forschungsstiftung, Zweigniederlassung Methods for regulating metabolism and diagnosing disease
JP2009514877A (en) * 2005-11-04 2009-04-09 エムディーアールエヌエー,インコーポレイテッド Peptide-Dither substrate RNA conjugates as siRNA delivery vehicles
JP2007151544A (en) * 2005-11-08 2007-06-21 Ehime Univ siRNA SPECIFIC FOR Akt GENE
EP3018202A1 (en) 2005-11-11 2016-05-11 Korea Research Institute of Bioscience and Biotechnology E2EPF Ubiquitin carrier protein-von-Hippel-Lindau interaction and uses of thereof
EP1984003A2 (en) * 2006-01-20 2008-10-29 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of rtp801
EP2402443A2 (en) 2006-01-20 2012-01-04 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of rtp801
NL2000439C2 (en) * 2006-01-20 2009-03-16 Quark Biotech Therapeutic applications of inhibitors of RTP801.
US8067570B2 (en) 2006-01-20 2011-11-29 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
EP1984003A4 (en) * 2006-01-20 2011-01-05 Quark Pharmaceuticals Inc Therapeutic uses of inhibitors of rtp801
US7825099B2 (en) 2006-01-20 2010-11-02 Quark Pharmaceuticals, Inc. Treatment or prevention of oto-pathologies by inhibition of pro-apoptotic genes
EP2402443A3 (en) * 2006-01-20 2015-10-07 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of rtp801
US8404654B2 (en) 2006-01-20 2013-03-26 Quark Pharmaceuticals, Inc. Treatment or prevention of oto-pathologies by inhibition of pro-apoptotic genes
US9056903B2 (en) 2006-01-20 2015-06-16 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
US7910566B2 (en) 2006-03-09 2011-03-22 Quark Pharmaceuticals Inc. Prevention and treatment of acute renal failure and other kidney diseases by inhibition of p53 by siRNA
EP2292739A1 (en) 2006-03-24 2011-03-09 Institut National De La Recherche Agronomique Method for preparing differentiated avian cells and genes involved in the maintenance of pluripotency
US8404831B2 (en) 2006-05-04 2013-03-26 Novartis Ag Short interfering ribonucleic acid (siRNA) for oral administration
US8344128B2 (en) 2006-05-04 2013-01-01 Novartis Ag Short interfering ribonucleic acid (siRNA) for oral administration
US8957041B2 (en) 2006-05-04 2015-02-17 Novartis Ag Short interfering ribonucleic acid (siRNA) for oral administration
US9493771B2 (en) 2006-05-04 2016-11-15 Novartis Ag Short interfering ribonucleic acid (siRNA) for oral administration
US8084600B2 (en) 2006-05-04 2011-12-27 Novartis Ag Short interfering ribonucleic acid (siRNA) with improved pharmacological properties
US8404832B2 (en) 2006-05-04 2013-03-26 Novartis Ag Short interfering ribonucleic acid (siRNA) for oral administration
US7626015B2 (en) 2006-06-09 2009-12-01 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801L
US8017764B2 (en) 2006-06-09 2011-09-13 Quark Pharmaceuticals Inc. Therapeutic uses of inhibitors of RTP801L
EP2546337A1 (en) 2006-07-21 2013-01-16 Silence Therapeutics AG Means for inhibiting the expression of protein kinase 3
WO2008031569A1 (en) * 2006-09-11 2008-03-20 Max-Delbrück-Centrum für Molekulare Medizin Means for increasing intracellular levels of protoporphyrin ix and use thereof
EP1897941A1 (en) * 2006-09-11 2008-03-12 Max-Delbrück-Centrum Für Molekulare Medizin Means for increasing intracellular levels of protoporphyrin IX and use thereof
US8252755B2 (en) 2006-09-22 2012-08-28 Dharmacon, Inc. Duplex oligonucleotide complexes and methods for gene silencing by RNA interference
EP2371958A1 (en) 2006-10-25 2011-10-05 Quark Pharmaceuticals, Inc. Novel siRNAs and methods of use thereof
EP2076526A4 (en) * 2006-10-25 2010-12-29 Quark Pharmaceuticals Inc Novel sirnas and methods of use thereof
US9446062B2 (en) 2006-10-25 2016-09-20 Quark Pharmaceuticals, Inc. Methods of treating ischemia-reperfusion injury with siRNAs
JP2010507387A (en) * 2006-10-25 2010-03-11 クアーク・ファーマスーティカルス、インコーポレイテッド Novel siRNA and method of using the same
EP2076526A2 (en) * 2006-10-25 2009-07-08 Quark Pharmaceuticals, Inc. Novel sirnas and methods of use thereof
EP2407558A1 (en) 2006-10-31 2012-01-18 Noxxon Pharma AG Methods for the detection of a single- or double-stranded nucleic acid molecule
US7872119B2 (en) 2007-02-26 2011-01-18 Quark Pharmaceuticals, Inc. Inhibitors of RTP801 and their use in disease treatment
US7812002B2 (en) 2007-03-21 2010-10-12 Quark Pharmaceuticals, Inc. Oligoribonucleotide inhibitors of NRF2 and methods of use thereof for treatment of cancer
US8410069B2 (en) 2007-03-21 2013-04-02 Quark Pharmaceuticals, Inc. Oligoribonucleotide inhibitors of Nrf2 and methods of use thereof for treatment of cancer
EP2155772A4 (en) * 2007-05-01 2011-04-06 Hope City Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US9873875B2 (en) 2007-05-01 2018-01-23 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US8883996B2 (en) 2007-05-01 2014-11-11 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US9441227B2 (en) 2007-05-01 2016-09-13 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US10233450B2 (en) 2007-05-01 2019-03-19 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
EP2155772A1 (en) * 2007-05-01 2010-02-24 City of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US8785408B2 (en) 2007-06-27 2014-07-22 Quark Pharmaceuticals, Inc. Compositions and methods for reducing or protecting against delayed graft function (DGF)
WO2009044392A2 (en) 2007-10-03 2009-04-09 Quark Pharmaceuticals, Inc. Novel sirna structures
US8188060B2 (en) 2008-02-11 2012-05-29 Dharmacon, Inc. Duplex oligonucleotides with enhanced functionality in gene regulation
EP2256191A1 (en) * 2008-02-15 2010-12-01 Riken Cyclic single-stranded nucleic acid complex and method for producing the same
EP2256191A4 (en) * 2008-02-15 2011-07-06 Riken Cyclic single-stranded nucleic acid complex and method for producing the same
EP2268316A4 (en) * 2008-03-20 2011-05-25 Quark Pharmaceuticals Inc NOVEL siRNA COMPOUNDS FOR INHIBITING RTP801
EP2268316A2 (en) * 2008-03-20 2011-01-05 Quark Pharmaceuticals, Inc. NOVEL siRNA COMPOUNDS FOR INHIBITING RTP801
EP2285385A2 (en) * 2008-04-15 2011-02-23 Quark Pharmaceuticals, Inc. siRNA COMPOUNDS FOR INHIBITING NRF2
EP2285385A4 (en) * 2008-04-15 2013-01-16 Quark Pharmaceuticals Inc siRNA COMPOUNDS FOR INHIBITING NRF2
WO2009147684A2 (en) 2008-06-06 2009-12-10 Quark Pharmaceuticals, Inc. Compositions and methods for treatment of ear disorders
US9885036B2 (en) 2008-07-01 2018-02-06 Daiichi Sankyo Company, Limited Double-stranded polynucleotide
US8765931B2 (en) * 2008-10-22 2014-07-01 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide compound for down-regulating the expression of CASP2 gene
US20110229557A1 (en) * 2008-10-22 2011-09-22 Elena Feinstein Methods for treating eye disorders
US9121020B2 (en) 2008-10-22 2015-09-01 Quark Pharmaceuticals, Inc. Methods for treating eye disorders
US9701961B2 (en) 2008-10-22 2017-07-11 Quark Pharmaceuticals, Inc. Methods for treating eye disorders
WO2010080452A2 (en) 2008-12-18 2010-07-15 Quark Pharmaceuticals, Inc. siRNA COMPOUNDS AND METHODS OF USE THEREOF
WO2010091878A2 (en) 2009-02-13 2010-08-19 Silence Therapeutics Ag Means for inhibiting the expression of opa1
WO2010094491A1 (en) 2009-02-18 2010-08-26 Silence Therapeutics Ag Means for inhibiting the expression of ang2
EP3276004A2 (en) 2009-06-08 2018-01-31 Quark Pharmaceuticals, Inc. Methods for treating chronic kidney disease
US10494631B2 (en) 2009-11-26 2019-12-03 Quark Pharmaceuticals, Inc. siRNA compounds comprising terminal substitutions
US8796239B2 (en) 2009-11-26 2014-08-05 Quark Pharmaceuticals, Inc. Sirna compounds comprising terminal substitutions
WO2011066475A1 (en) 2009-11-26 2011-06-03 Quark Pharmaceuticals, Inc. Sirna compounds comprising terminal substitutions
US9701960B2 (en) 2009-11-26 2017-07-11 Quark Pharmaceuticals, Inc. siRNA compounds comprising terminal substitutions
EP2862929A1 (en) 2009-12-09 2015-04-22 Quark Pharmaceuticals, Inc. Compositions and methods for treating diseases, disorders or injury of the CNS
WO2011072091A1 (en) 2009-12-09 2011-06-16 Quark Pharmaceuticals, Inc. Methods and compositions for treating diseases, disorders or injury of the cns
WO2011084193A1 (en) 2010-01-07 2011-07-14 Quark Pharmaceuticals, Inc. Oligonucleotide compounds comprising non-nucleotide overhangs
WO2011085056A1 (en) 2010-01-07 2011-07-14 Quark Pharmaceuticals, Inc. Oligonucleotide compounds comprising non-nucleotide overhangs
US10695362B2 (en) 2010-05-14 2020-06-30 Tagcyx Biotechnologies Inc. Stabilization method of functional nucleic acid
WO2011163436A1 (en) 2010-06-24 2011-12-29 Quark Pharmaceuticals, Inc. Double stranded rna compounds to rhoa and use thereof
EP2933333A1 (en) * 2010-07-08 2015-10-21 Bonac Corporation Single-strand nucleic acid molecule for controlling gene expression
WO2012013784A1 (en) 2010-07-29 2012-02-02 Centre National De La Recherche Scientifique (C.N.R.S) Regulation of glypican 4 activity to modulate the fate of stem cells and uses thereof
EP2412724A1 (en) 2010-07-29 2012-02-01 Centre National de la Recherche Scientifique (C.N.R.S) Regulation of Glypican 4 activity to modulate the fate of stem cells and uses thereof
US9200278B2 (en) 2010-08-03 2015-12-01 Bonac Corporation Single-stranded nucleic acid molecule having nitrogen-containing alicyclic skeleton
US9206422B2 (en) 2010-08-03 2015-12-08 Bonac Corporation Single-stranded nucleic acid molecule having nitrogen-containing alicyclic skeleton
US10584335B2 (en) 2010-08-24 2020-03-10 Sirna Therapeutics, Inc. Single-stranded RNAi agents containing an internal, non-nucleic acid spacer
US9845466B2 (en) 2010-08-24 2017-12-19 Sirna Therapeutics, Inc. Single-stranded RNAi agents containing an internal, non-nucleic acid spacer
US9243246B2 (en) 2010-08-24 2016-01-26 Sirna Therapeutics, Inc. Single-stranded RNAi agents containing an internal, non-nucleic acid spacer
US9290760B2 (en) 2010-09-15 2016-03-22 Alnylam Pharmaceuticals, Inc. Modified iRNA agents
WO2012037254A1 (en) * 2010-09-15 2012-03-22 Alnylam Pharmaceuticals, Inc. MODIFIED iRNA AGENTS
US9260471B2 (en) 2010-10-29 2016-02-16 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
US9970005B2 (en) 2010-10-29 2018-05-15 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
US11193126B2 (en) 2010-10-29 2021-12-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
US11932854B2 (en) 2010-10-29 2024-03-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acids (siNA)
WO2012078536A2 (en) 2010-12-06 2012-06-14 Quark Pharmaceuticals, Inc. Double stranded oligonucleotide compounds comprising positional modifications
US9796979B2 (en) 2011-03-03 2017-10-24 Quark Pharmaceuticals Inc. Oligonucleotide modulators of the toll-like receptor pathway
WO2012118911A1 (en) 2011-03-03 2012-09-07 Quark Pharmaceuticals, Inc. Oligonucleotide modulators of the toll-like receptor pathway
US9487778B2 (en) 2011-03-03 2016-11-08 Quark Pharmaceuticals, Inc. Oligonucleotide modulators of the toll-like receptor pathway
WO2013070821A1 (en) 2011-11-08 2013-05-16 Quark Pharmaceuticals, Inc. Methods and compositions for treating diseases, disorders or injury of the nervous system
US9399775B2 (en) 2011-11-18 2016-07-26 Alnylam Pharmaceuticals, Inc. RNAi agents, compositions and methods of use thereof for treating transthyretin (TTR) associated diseases
WO2013074974A3 (en) * 2011-11-18 2013-08-08 Alnylam Pharmaceuticals, Inc. Modified rnai agents
KR102534909B1 (en) 2011-11-18 2023-05-30 알닐람 파마슈티칼스 인코포레이티드 MODIFIED RNAi AGENTS
US10668170B2 (en) 2011-11-18 2020-06-02 Alnylam Pharmaceuticals, Inc. Modified RNAi agents
US9796974B2 (en) 2011-11-18 2017-10-24 Alnylam Pharmaceuticals, Inc. Modified RNAi agents
US11406716B2 (en) 2011-11-18 2022-08-09 Alnylam Pharmaceuticals, Inc. Modified RNAi agents
KR20220061279A (en) * 2011-11-18 2022-05-12 알닐람 파마슈티칼스 인코포레이티드 MODIFIED RNAi AGENTS
US10570391B2 (en) 2011-11-18 2020-02-25 Alnylam Pharmaceuticals, Inc. RNAi agents, compositions and methods of use thereof for treating transthyretin (TTR) associated diseases
WO2013098813A1 (en) 2012-01-01 2013-07-04 Qbi Enterprises Ltd. Endo180-targeted particles for selective delivery of therapeutic and diagnostic agents
WO2013106494A1 (en) 2012-01-12 2013-07-18 Quark Pharmaceuticals, Inc. Combination therapy for treating hearing and balance disorders
WO2013123996A1 (en) 2012-02-24 2013-08-29 Astrazeneca Uk Limited Novel sirna inhibitors of human icam-1
WO2013166043A1 (en) 2012-05-02 2013-11-07 Children's Hospital Medical Center Rejuvenation of precursor cells
US10238752B2 (en) 2012-05-26 2019-03-26 Bonac Corporation Single-stranded nucleic acid molecule for regulating expression of gene having delivering function
US9611474B2 (en) 2012-09-12 2017-04-04 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to DDIT4 and methods of use thereof
WO2014043289A3 (en) * 2012-09-12 2014-06-05 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to ddit4 and methods of use thereof
WO2014043292A1 (en) 2012-09-12 2014-03-20 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to p53 and methods of use thereof
WO2014043289A2 (en) 2012-09-12 2014-03-20 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to ddit4 and methods of use thereof
US10421962B2 (en) 2012-09-12 2019-09-24 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to DDIT4 and methods of use thereof
WO2014176259A1 (en) 2013-04-22 2014-10-30 Icahn School Of Medicine At Mount Sinai Mutations in pdgfrb and notch3 as causes of autosomal dominant infantile myofibromatosis
WO2015005253A1 (en) 2013-07-08 2015-01-15 第一三共株式会社 Novel lipid
WO2015015496A1 (en) 2013-07-31 2015-02-05 Qbi Enterprises Ltd. Sphingolipid-polyalkylamine-oligonucleotide compounds
WO2015015498A1 (en) 2013-07-31 2015-02-05 Qbi Enterprises Ltd. Methods of use of sphingolipid polyalkylamine oligonucleotide compounds
EP2853595A1 (en) 2013-09-30 2015-04-01 Soluventis GmbH NOTCH 1 specific siRNA molecules
WO2015043768A1 (en) 2013-09-30 2015-04-02 Soluventis Gmbh Notch 1 specific sirna molecule
EP2865756A1 (en) * 2013-10-22 2015-04-29 Sylentis, S.A.U. siRNA and their use in methods and compositions for inhibiting the expression of the FLAP gene.
WO2015059122A3 (en) * 2013-10-22 2015-08-20 Sylentis Sau Sirna and their use in methods and compositions for inhibiting the expression of the orai1 gene
EP2865757A1 (en) * 2013-10-22 2015-04-29 Sylentis, S.A.U. siRNA and their use in methods and compositions for inhibiting the expression of the PDK1 gene.
US10167473B2 (en) 2013-10-22 2019-01-01 Sylentis Sau SiRNA and their use in methods and compositions for inhibiting the expression of the ORAI1 gene
EP2865758A1 (en) * 2013-10-22 2015-04-29 Sylentis, S.A.U. siRNA and their use in methods and compositions for inhibiting the expression of the ORAI1 gene
US10017766B2 (en) 2013-10-22 2018-07-10 Sylentis Sau SiRNA and their use in methods and compositions for inhibiting the expression of the FLAP gene
US9951338B2 (en) 2013-10-22 2018-04-24 Sylentis Sau siRNA and their use in methods and compositions for inhibiting the expression of the PDK1 gene
WO2015059116A3 (en) * 2013-10-22 2015-08-20 Sylentis Sau Sirna and their use in methods and compositions for inhibiting the expression of the flap gene
WO2015059124A1 (en) * 2013-10-22 2015-04-30 Sylentis Sau Sirna and their use in methods and compositions for inhibiting the expression of the pdk1 gene
US10612020B2 (en) 2013-12-26 2020-04-07 Tokyo Medical University Artificial mimic miRNA for controlling gene expression, and use of same
US10934542B2 (en) 2013-12-27 2021-03-02 Bonac Corporation Artificial match-type miRNA for controlling gene expression and use therefor
EP3105331B1 (en) 2014-02-11 2021-06-23 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
JP2017523788A (en) * 2014-08-07 2017-08-24 財團法人工業技術研究院Industrial Technology Research Institute Small interfering RNA for suppressing galectin-12 expression and / or promoting lipolysis and pharmaceutical composition comprising the same, and for inhibiting galectin-12 expression and / or promoting lipolysis How to
WO2016083624A1 (en) 2014-11-28 2016-06-02 Silence Therapeutics Gmbh Means for inhibiting the expression of edn1
US11027023B2 (en) 2014-12-27 2021-06-08 Bonac Corporation Natural type miRNA for controlling gene expression, and use of same
WO2016149020A1 (en) * 2015-03-17 2016-09-22 Arrowhead Research Corporation Rna interference agents
US11142769B2 (en) 2015-03-27 2021-10-12 Bonac Corporation Single-stranded nucleic acid molecule having delivery function and gene expression regulating ability
US10683501B2 (en) 2015-07-31 2020-06-16 Alnylam Pharmaceuticals, Inc. Transthyretin (TTR) iRNA compositions and methods of use thereof for treating or preventing TTR-associated diseases
US11286486B2 (en) 2015-07-31 2022-03-29 Alnylam Pharmaceuticals, Inc. Transthyretin (TTR) iRNA compositions and methods of use thereof for treating or preventing TTR-associated diseases
US10208307B2 (en) 2015-07-31 2019-02-19 Alnylam Pharmaceuticals, Inc. Transthyretin (TTR) iRNA compositions and methods of use thereof for treating or preventing TTR-associated diseases
WO2017079442A1 (en) 2015-11-04 2017-05-11 Icahn School Of Medicine At Mount Sinai Methods of treating tumors and cancer, and identifying candidate subjects for such treatment
US11530409B2 (en) 2016-01-26 2022-12-20 Nissan Chemical Corporation Single-stranded oligonucleotide
WO2017189730A1 (en) 2016-04-26 2017-11-02 Icahn School Of Medicine At Mount Sinai Treatment of hippo pathway mutant tumors and methods of identifying subjects as candidates for treatment
EP3584319A4 (en) * 2017-02-06 2021-04-14 Nissan Chemical Corporation Single-stranded oligonucleotide
US11572558B2 (en) 2017-02-06 2023-02-07 Nissan Chemical Corporation Single-stranded oligonucleotide
EP4219715A3 (en) * 2017-09-08 2023-09-06 MiNA Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
WO2019048645A1 (en) * 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
US11806360B2 (en) 2017-09-19 2023-11-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating transthyretin (TTR) mediated amyloidosis
WO2019246112A1 (en) 2018-06-18 2019-12-26 University Of Rochester Methods of treating schizophrenia and other neuropsychiatric disorders
WO2019246262A2 (en) 2018-06-21 2019-12-26 University Of Rochester Methods of treating or inhibiting onset of huntington's disease
WO2020123663A1 (en) 2018-12-11 2020-06-18 University Of Rochester Methods of treating schizophrenia and other neuropsychiatric disorders
WO2020167822A2 (en) 2019-02-13 2020-08-20 University Of Rochester Gene networks that mediate remyelination of the human brain
WO2021011936A2 (en) 2019-07-18 2021-01-21 University Of Rochester Cell-type selective immunoprotection of cells
WO2021049504A1 (en) 2019-09-10 2021-03-18 第一三共株式会社 Galnac-oligonucleotide conjugate for liver-targeted delivery use, and method for producing same
US11959081B2 (en) 2021-08-03 2024-04-16 Alnylam Pharmaceuticals, Inc. Transthyretin (TTR) iRNA compositions and methods of use thereof
WO2023031394A1 (en) 2021-09-03 2023-03-09 CureVac SE Novel lipid nanoparticles for delivery of nucleic acids
WO2023150553A1 (en) 2022-02-01 2023-08-10 University Of Rochester Gpr17 promoter-based targeting and transduction of glial progenitor cells

Also Published As

Publication number Publication date
CY1119021T1 (en) 2018-01-10
US10323246B2 (en) 2019-06-18
US20110118456A1 (en) 2011-05-19
BRPI0313202A8 (en) 2016-08-16
EP3222725A1 (en) 2017-09-27
US20170247691A1 (en) 2017-08-31
DK1527176T4 (en) 2017-07-03
ES2280826T3 (en) 2007-09-16
US20170247707A1 (en) 2017-08-31
CY1120128T1 (en) 2018-12-12
KR20050035877A (en) 2005-04-19
DE60310944D1 (en) 2007-02-15
MXPA05001355A (en) 2005-09-30
JP2011024592A (en) 2011-02-10
US20170247697A1 (en) 2017-08-31
DK2258847T3 (en) 2017-06-26
DK3222724T3 (en) 2018-12-03
IL166546A (en) 2014-01-30
US8933215B2 (en) 2015-01-13
EP1857547B1 (en) 2018-01-17
US20130102769A1 (en) 2013-04-25
US9783802B2 (en) 2017-10-10
DE60310944T3 (en) 2017-08-03
SI2258847T1 (en) 2017-08-31
US9222092B2 (en) 2015-12-29
EP1857547A2 (en) 2007-11-21
CN1675359A (en) 2005-09-28
AU2008207337A1 (en) 2008-09-04
US20210062198A1 (en) 2021-03-04
EP3222726A1 (en) 2017-09-27
WO2004015107A3 (en) 2004-04-29
CA2494930A1 (en) 2004-02-19
US9758784B1 (en) 2017-09-12
US8324370B2 (en) 2012-12-04
ES2280826T5 (en) 2017-08-03
US20190024089A1 (en) 2019-01-24
AU2003260370B2 (en) 2008-05-22
US7452987B2 (en) 2008-11-18
US20180223286A1 (en) 2018-08-09
US20160130587A1 (en) 2016-05-12
EP1527176B1 (en) 2007-01-03
DK1527176T3 (en) 2007-05-07
US20150105545A1 (en) 2015-04-16
PT1527176E (en) 2007-04-30
EP2258847B2 (en) 2020-07-01
JP2013179944A (en) 2013-09-12
US20190390201A1 (en) 2019-12-26
BR0313202A (en) 2005-06-21
EP3222724B1 (en) 2018-10-31
EP1857547A3 (en) 2011-05-18
US20170247698A1 (en) 2017-08-31
EP3222724A1 (en) 2017-09-27
ZA200500459B (en) 2005-08-31
CA2494930C (en) 2021-03-09
US10774332B2 (en) 2020-09-15
JP2016144451A (en) 2016-08-12
KR101201664B1 (en) 2012-11-15
DK1857547T3 (en) 2018-04-23
JP2017163995A (en) 2017-09-21
IL166546A0 (en) 2006-01-15
AU2003260370A1 (en) 2004-02-25
EP2258847A2 (en) 2010-12-08
EP2258847B1 (en) 2017-03-15
CY1120919T1 (en) 2019-12-11
US9790505B2 (en) 2017-10-17
US20090186845A1 (en) 2009-07-23
DE60310944T2 (en) 2007-10-11
EP1527176A2 (en) 2005-05-04
JP5490655B2 (en) 2014-05-14
CY1106422T1 (en) 2011-10-12
US7893245B2 (en) 2011-02-22
EP1857547B2 (en) 2020-12-02
US20040180351A1 (en) 2004-09-16
EP1527176B2 (en) 2017-03-22
EP2258847A3 (en) 2011-05-18
SI3222724T1 (en) 2019-03-29
US9695423B2 (en) 2017-07-04
US10329568B2 (en) 2019-06-25
US20180291381A1 (en) 2018-10-11
ATE350473T2 (en) 2007-01-15
JP6069096B2 (en) 2017-01-25
US10266829B2 (en) 2019-04-23
SI1857547T1 (en) 2018-05-31
US9790501B2 (en) 2017-10-17
CN100543137C (en) 2009-09-23
US11578328B2 (en) 2023-02-14

Similar Documents

Publication Publication Date Title
US11578328B2 (en) Interfering RNA molecules
EP1389637A1 (en) Novel forms of interfering RNA molecules
AU2018200311B2 (en) Further novel forms of interfering rna molecules
AU2012216354B2 (en) Further novel forms of interfering RNA molecules

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2005/00459

Country of ref document: ZA

Ref document number: 200500459

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2003260370

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2003784183

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 166546

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: PA/a/2005/001355

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1020057002008

Country of ref document: KR

Ref document number: 427/DELNP/2005

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2494930

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 20038188678

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2004526885

Country of ref document: JP

WWP Wipo information: published in national office

Ref document number: 1020057002008

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2003784183

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2003784183

Country of ref document: EP