US20100004436A1 - Methods and compositions for the specific inhibition of gene expression by double-stranded rna - Google Patents

Methods and compositions for the specific inhibition of gene expression by double-stranded rna Download PDF

Info

Publication number
US20100004436A1
US20100004436A1 US12/549,680 US54968009A US2010004436A1 US 20100004436 A1 US20100004436 A1 US 20100004436A1 US 54968009 A US54968009 A US 54968009A US 2010004436 A1 US2010004436 A1 US 2010004436A1
Authority
US
United States
Prior art keywords
nucleic acid
stranded nucleic
double stranded
terminus
strand
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/549,680
Inventor
John J. Rossi
Mark A. Behlke
Dongho Kim
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Integrated DNA Technologies Inc
City of Hope
Original Assignee
Integrated DNA Technologies Inc
City of Hope
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=34994243&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20100004436(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Integrated DNA Technologies Inc, City of Hope filed Critical Integrated DNA Technologies Inc
Priority to US12/549,680 priority Critical patent/US20100004436A1/en
Publication of US20100004436A1 publication Critical patent/US20100004436A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-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
    • 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
    • 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
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/02Drugs for genital or sexual disorders; Contraceptives for disorders of the vagina
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • 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
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/08Antibacterial agents for leprosy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • 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
    • 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
    • C12N2310/33Chemical structure of the base
    • 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
    • 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
    • C12N2310/51Physical structure in polymeric form, e.g. multimers, concatemers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2330/00Production
    • C12N2330/30Production chemically synthesised

Definitions

  • compositions and methods for gene-specific inhibition of gene expression by double-stranded ribonucleic acid (dsRNA) effector molecules are useful in modulating gene expression in a variety of applications, including therapeutic, diagnostic, target validation, and genomic discovery.
  • dsRNA double-stranded RNA
  • the protein kinase PKR is activated by dsRNAs of greater than 30 bp long (Manche et al., 1992, Mol Cell Biol., 12:5238-48) and results in phosphorylation of translation initiation factor eIF2 ⁇ which leads to arrest of protein synthesis and activation of 2′5′-oligoadenylate synthetase (2′-5′-OAS), which leads to RNA degradation (Minks et al., 1979, J. Biol. Chem. 254:10180-10183).
  • dsRNAs of 150 bp length or greater were seen to induce RNA interference while shorter dsRNAs were ineffective (Tuschl et al., 1999, Genes & Dev., 13:3191-3197).
  • Long double-stranded RNA is not the active effecter molecule; long dsRNAs are degraded by an RNase III class enzyme called Dicer (Bernstein et al., 2001, Nature, 409:363-366) into very short 21-23 bp duplexes that have 2-base 3′-overhangs (Zamore et al., 2000, Cell, 101:25-33).
  • siRNAs direct the RNAi response in vivo and transfection of short chemically synthesized siRNA duplexes of this design permits use of RNAi methods to suppress gene expression in mammalian cells without triggering unwanted interferon responses (Elbashir et al., 2001, Nature, 411:494-498).
  • the antisense strand of the siRNA duplex serves as a sequence-specific guide that directs activity of an endoribonuclease function in the RNA induced silencing complex (RISC) to degrade target mRNA (Martinez et al., 2002, Cell, 110:563-574).
  • RISC RNA induced silencing complex
  • RNAi In studying the size limits for RNAi in Drosophila embryo extracts in vitro, a lower threshold of around 38 bp double-stranded RNA was established for activation of RNA interference using exogenously supplied double-stranded RNA and duplexes of 36, 30, and 29 bp length were without effect (Elbashir et al., 2001, Genes & Dev., 15:188-200). The short 30-base RNAs were not cleaved into active 21-23-base siRNAs and therefore were deemed inactive for use in RNAi (Elbashir et al., 2001, Genes & Dev., 15:188-200).
  • RNA duplexes of 21-bp length with 2-base 3′-overhangs were most effective, duplexes of 20, 22, and 23-bp length had slightly decreased potency but did result in RNAi mediated mRNA degradation, and 24 and 25-bp duplexes were inactive (Elbashir et al., 2001, EMBO J., 20:6877-6888).
  • elegans found that although a microinjected 26-bp RNA duplex could function to suppress gene expression, it required a 250-fold increase in concentration compared with an 81-bp duplex (Parrish et al., 2000, Mol. Cell, 6:1077-1087).
  • RNAi molecules are capable of targeting the destruction of their complementary RNAs in a cell.
  • complex sets of rules have been developed for designing RNAi molecules that will be effective.
  • Those having skill in the art expect to test multiple siRNA molecules to find functional compositions.
  • Some artisans pool several siRNA preparations together to increase the chance of obtaining silencing in a single study.
  • Such pools typically contain 20 nM of a mixture of siRNA oligonucleotide duplexes or more (Ji et al.
  • RNAi treatment duration is limited to about 4 days (Holen et al. 2002).
  • researchers must carry out siRNA experiments within 2-3 days of transfection with an siRNA duplex or work with plasmid or viral expression vectors to obtain longer term silencing.
  • RNAi active oligonucleotide duplexes To more completely characterize the structural requirements of RNAi active oligonucleotide duplexes to identify more potent and longer lasting compositions and/or methods that simplify site-selection difficulties. These studies should also include a detailed analysis of the interferon response. Ideally, such studies will be useful in identifying new RNAi active compounds that are more potent, that simplify the site selection process, and decrease “off target effects.”
  • RNAi compositions with increased potency, duration of action, and decreased “off target effects” that do not activate the interferon response and provides methods for their use.
  • the compositions ease site selection criteria and provide a duration of action that is about twice as long as prior known compositions.
  • the invention provides improved compositions and methods for selectively reducing the expression of a gene product from a desired target gene in a eukaryotic cell, as well as for treating diseases caused by the expression of the gene.
  • the method involves introducing into the environment of a cell an amount of a double-stranded RNA (dsRNA) such that a sufficient portion of the dsRNA can enter the cytoplasm of the cell to cause a reduction in the expression of the target gene.
  • the dsRNA has a first oligonucleotide sequence that is between 26 and about 30 nucleotides in length and a second oligonucleotide sequence that anneals to the first sequence under biological conditions, such as the conditions found in the cytoplasm of a cell.
  • a region of one of the sequences of the dsRNA having a sequence length of from about 19 to about 23 nucleotides is complementary to a nucleotide sequence of the RNA produced from the target gene.
  • a dsRNA composition of the invention is at least as active as any isolated 19, 20, 21, 22, or 23 basepair sequence that is contained within it.
  • Pharmaceutical compositions containing the disclosed dsRNA compositions are also contemplated. The compositions and methods give a surprising increase in the potency and duration of action of the RNAi effect.
  • dsRNA serves as a substrate for Dicer which appears to facilitate incorporation of one sequence from the dsRNA into the RISC complex that is directly responsible for destruction of the RNA from the target gene.
  • FIG. 1 provides a comparison of RNAi efficacy using several dsRNAs having variable length and formats including a two nucleotide 3′ overhang (+2), a two nucleotide 5′ overhang ( ⁇ 2), and blunt ends (+0).
  • the sequences are disclosed in the Example 2.
  • 200 ⁇ g of reporter vector was co-transfected with the indicated concentration of dsRNA.
  • Each bar represents the average of three duplicate experiments.
  • 50 nM of each dsRNA was used.
  • FIG. 1B 1 nM of each dsRNA was used.
  • FIG. 1C 200 pM of each dsRNA was used.
  • 50 pM of each dsRNA was used.
  • FIG. 2 shows an RNAi assay in 3T3 cells expressing endogenous EGFP.
  • the experimental procedure is described in Example 2. Measurements were made 4 days after treatment.
  • the dose response curves for 21-mer duplex with 2-base 3′-overhang (SEQ ID No. 6/7), 25-mer duplex with 2-base 5′-overhang (SEQ ID Nos. 16/17), and blunt 27-mer duplex (SEQ ID Nos. 30/31) are shown.
  • FIG. 3 shows RNAi assays of various 27-mer RNA duplex formats as outlined in Example 2.
  • Duplex 27+0UU SEQ ID Nos. 30/31) was most potent.
  • FIG. 4 shows RNAi assays on HEK 293 cells that were either mock transfected (negative control), transfected with 200 ng EGFP reporter plasmid alone (positive control), or reporter plasmid+RNA duplexes at varying concentrations as described in Example 3.
  • FIG. 5 shows superior knockout of the HNRPH1 gene by a 27-mer of the invention as compared to a 21-mer directed to the same target.
  • Western blots obtained from HEK 293 cells after transfection with EGFP-specific siRNA (SEQ ID No. 6/7; C) (negative control) and an HNRPH1 specific 21-mer siRNA duplex (SEQ ID Nos. 51/52; 21+2) at varying concentrations, or with an HNRPH1 specific 27-mer siRNA duplex (SEQ ID Nos. 53/54; 27+0) at varying concentrations, as described in Example 4.
  • FIG. 6 shows the reaction of Dicer with various length RNA duplexes as described in Example 5.
  • Dicer was able to digest 25-29-mers primarily into about a 21 basepair duplex but dd not digest the 21 nucleotide long test duplex.
  • FIG. 7 shows the relative expression of EGFP after RNAi assays using a 27-mer dsRNA versus shorter 21-mer siRNAs contained within the 27-mer sequence as described in more detail in Example 6. As shown a blunt ended 27-mer that covers a poor site for a 21 nucleotide RNAi can effectively target that site.
  • FIG. 8 shows the results of RNAi assays after treatment by various effector dsRNA molecules and pools of molecules as set forth in Example 6.
  • FIG. 9 shows the time course study of the duration of the RNAi effect with various effector molecules as described in Example 7.
  • the study shows the duration of the RNAi effect is at least about twice as long with the 27-mer dsRNA of the invention as with 21-mers.
  • the “27+0 UU” sequences are set forth in SEQ ID NOs:28 and 29.
  • the “Mut-16” sequences are set forth in SEQ ID NOs:70 and 71.
  • the “Mut-16,17” sequences are set forth in SEQ ID NOs:72 and 73.
  • the “Mut-15,16,17” sequences are set forth in SEQ ID NOs:74 and 75.
  • FIG. 10 shows the images of cells in a time course study of the duration of the RNAi effect with various effector molecules as described in Example 7.
  • the study shows the duration of the RNAi effect is at least about twice as long with the 27-mer dsRNA of the invention as with 21-mers.
  • FIG. 11 shows that neither interferon alpha ( FIG. 11A ) or interferon beta ( FIG. 11B ) are induced by the 27-mer dsRNA of the invention as described in more detail in Example 8.
  • FIG. 12 shows the results of a PKR activation assay in which long dsRNA resulted in strong PKR activation (positive control) while all of the short synthetic RNAs showed no evidence for PKR activation.
  • the invention is directed to compositions that contain double stranded RNA (“dsRNA”), and methods for preparing them, that are capable of reducing the expression of target genes in eukaryotic cells.
  • dsRNA double stranded RNA
  • One of the strands of the dsRNA contains a region of nucleotide sequence that has a length that ranges from about 19 to about 23 nucleotides that can direct the destruction of the RNA transcribed from the target gene.
  • a suitable dsRNA contains one oligonucleotide sequence, a first sequence, that is at least 25 nucleotides in length and no longer than about 30 nucleotides. More preferably this sequence of RNA is between about 26 and 29 nucleotides in length. Still more preferably this sequence is about 27 or 28 nucleotides in length, 27 nucleotides is most preferred.
  • the second sequence of the dsRNA can be any sequence that anneals to the first sequence under biological conditions, such as within the cytoplasm of a eukaryotic cell.
  • the second oligonucleotide sequence will have at least 19 complementary base pairs with the first oligonucleotide sequence, more typically the second oligonucleotides sequence will have about 21 or more complementary base pairs, and more preferably about 25 or more complementary base pairs with the first oligonucleotide sequence.
  • the second sequence is the same length as the first sequence.
  • the double-stranded RNA structure the first and second oligonucleotide sequences exist on separate oligonucleotide strands which can be and typically are chemically synthesized. In preferred embodiments both strands are between 26 and 30 nucleotides in length. In one preferred embodiment both strands are 27 nucleotides in length, are completely complementary and have blunt ends.
  • the dsRNA can be from a single RNA oligonucleotide that undergoes intramolecular annealing or, more typically, the first and second sequences exist on separate RNA oligonucleotides.
  • Suitable dsRNA compositions that contain two separate oligonucleotides can be chemically linked outside their annealing region by chemical linking groups.
  • Many suitable chemical linking groups are known in the art and can be used. Suitable groups will not block Dicer activity on the dsRNA and will not interfere with the directed destruction of the RNA transcribed from the target gene.
  • the first and second oligonucleotide sequences are not required to be completely complimentary.
  • the 3′-terminus of the sense strand contains one or more mismatches. It is more preferred that two mismatches be incorporated at the 3′terminus.
  • the dsRNA of the invention is a double stranded RNA molecule containing two RNA oligonucleotides each of which is 27 nucleotides in length and, when annealed to each other, have blunt ends and a two nucleotide mismatch on the 3′-terminus of the sense strand (the 5′-terminus of the antisense strand).
  • dsRNA compositions of the invention can serve as a substrate for Dicer.
  • the dsRNA compositions of this invention will not have been treated with Dicer, other RNAses, or extracts that contain them. Such treatments could digest the dsRNA to lengths of less than 25 nucleotides that are no longer Dicer substrates.
  • Dicer activity can be measured in vitro using the Recombinant Dicer Enzyme Kit (GTS, San Diego, Calif.) according to the manufacturer's instructions.
  • Dicer activity can be measured in vivo by treating cells with dsRNA and maintaining them for 24 h before harvesting them and isolating their RNA.
  • RNa can be isolated using standard methods, such as with the RNeasyTM Kit (Qiagen) according to the manufacturer's instructions. The isolated RNA can be separated on a 10% PAGE gel which is used to prepare a standard RNA blot that can be probed with a suitable labeled deoxyoligonucleotide, such as an oligonucleotide labeled with the StarfireTM Oligo Labeling System (Integrated DNA Technologies, Inc., Coralville, Iowa).
  • a suitable labeled deoxyoligonucleotide such as an oligonucleotide labeled with the StarfireTM Oligo Labeling System (Integrated DNA Technologies, Inc., Coralville, Iowa).
  • dsRNA compositions of the invention are at least as active as any isolated 23 nucleotide dsRNA sequence contained within them and in preferred embodiments more active. Without wishing to be bound by the underlying theory of the invention, it is thought that the longer dsRNA species serve as a substrate for the enzyme Dicer in the cytoplasm of a cell.
  • Dicer In addition to cleaving the dsRNA of the invention into shorter segments, Dicer is thought to facilitate the incorporation of a single-stranded cleavage product derived from the cleaved dsRNA into the RISC complex that is responsible for the destruction of the cytoplasmic RNA derived from the target gene. Studies have shown that the cleavability of a dsRNA species by Dicer corresponds with increased potency and duration of action of the dsRNA species.
  • Suitable dsRNA compositions of this invention do not induce apoptosis in the cells in which they are used.
  • Apoptosis or “programmed cell death,” includes any non-necrotic, cell-regulated form of cell death, as defined by criteria well established in the art. Cells undergoing apoptosis show characteristic morphological and biochemical features.
  • morphological and physiological changes include cell shrinkage, chromatin condensation, nuclear and cytoplasmic condensation, membrane blebbing, partitioning of cytoplasm and nucleus into membrane bound vesicles which contain ribosomes (apoptotic bodies), and DNA degradation into a characteristic oligonucleosomal ladder composed of multiples of 200 base pairs, leading eventually to cell death.
  • apoptotic bodies are rapidly recognized and phagocytized by either macrophages or adjacent epithelial cells.
  • the apoptotic bodies as well as the remaining cell fragments ultimately swell and finally lyse. This terminal phase of in vitro cell death has been termed “secondary necrosis.”
  • a dsRNA has on a cell can depend upon the cell itself. In some circumstances a dsRNA could induce apoptosis or gene silencing in one cell type and not another. Thus, it is possible that a dsRNA could be suitable for use in one cell and not another. To be considered “suitable” a dsRNA composition need not be suitable under all possible circumstances in which it might be used, rather it need only be suitable under a particular set of circumstances.
  • Modifications can be included in the disclosed dsRNA so long as the dsRNA remains sufficiently chemically stable, does not induce apoptosis, does not substantially interrupt annealing of the first and second strands, and otherwise does not substantially interfere with the directed destruction of the RNA transcribed from the target gene. Modifications can be incorporated in the 3′-terminal region, the 5′-terminal region, in both the 3′-terminal and 5′-terminal region or in some instances throughout the sequence. With the restrictions noted above in mind any number and combination of modifications can be incorporated into the dsRNA. Where multiple modifications are present, they may be the same or different. Modifications to bases, sugar moieties, the phosphate backbone, and their combinations are contemplated.
  • either the 3′ or 5′ terminal regions of the sequences in a dsRNA can be phosphorylated or biotinylated.
  • modifications contemplated for the phosphate backbone include phosphonates, including methylphosphonate, phosphorothioate, and phosphotriester modifications such as alkylphosphotriesters, and the like.
  • modifications contemplated for the sugar moiety include 2′-alkyl pyrimidine, such as 2′-O-methyl, 2′-fluoro, amino, and deoxy modifications and the like.
  • base groups examples include abasic sugars, 2-O-alkyl modified pyrimidines, 4-thiouracil, 5-bromouracil, 5-iodouracil, and 5-(3-aminoallyl)-uracil and the like. Many other modifications are known and can be used so long as the above criteria are satisfied
  • the double-stranded RNA sample can be suitably formulated and introduced into the environment of the cell by any means that allows for a sufficient portion of the sample to enter the cell to induce gene silencing, if it is to occur.
  • Many formulations for dsRNA are known in the art and can be used so long as dsRNA gains entry to the target cells so that it can act.
  • dsRNA can be formulated in buffer solutions such as phosphate buffered saline solutions, liposomes, micellar structures, and capsids.
  • Formulations of dsRNA with cationic lipids can be used to facilitate transfection of the dsRNA into cells.
  • Suitable lipids include Oligofectamine, Lipofectamine (Life Technologies), NC388 (Ribozyme Pharmaceuticals, Inc., Boulder, Colo.), or FuGene 6 (Roche) all of which can be used according to the manufacturer's instructions.
  • the method of introducing dsRNA into the environment of the cell will depend on the type of cell and the make up of its environment.
  • a lipid formulation such as in lipofectamine and the dsRNA can be added directly to the liquid environment of the cells.
  • Lipid formulations can also be administered to animals such as by intravenous, intramuscular, or intraperitoneal injection, or orally or by inhalation or other methods as are known in the art.
  • the formulation is suitable for administration into animals such as mammals and more specifically humans, the formulation is also pharmaceutically acceptable.
  • Pharmaceutically acceptable formulations for administering oligonucleotides are known and can be used.
  • dsRNA Suitable amounts of dsRNA must be introduced and these amounts can be empirically determined using standard methods. Typically, effective concentrations of individual dsRNA species in the environment of a cell will be about 50 nanomolar or less 10 nanomolar or less, more preferred are compositions in which concentrations of about 1 nanomolar or less can be used. Even more preferred are methods that utilize a concentration of about 200 picomolar or less and even a concentration of about 50 picomolar or less can be used in many circumstances.
  • the method can be carried out by addition of the dsRNA compositions to any extracellular matrix in which cells can live provided that the dsRNA composition is formulated so that a sufficient amount of the dsRNA can enter the cell to exert its effect.
  • the method is amenable for use with cells present in a liquid such as a liquid culture or cell growth media, in tissue explants, or in whole organisms, including animals, such as mammals and especially humans.
  • RNAi methods are applicable to a wide variety of genes in a wide variety of organisms and the disclosed compositions and methods can be utilized in each of these contexts.
  • genes which can be targeted by the disclosed compositions and methods include endogenous genes which are genes that are native to the cell or to genes that are not normally native to the cell. Without limitation these genes include oncogenes, cytokine genes, idiotype (Id) protein genes, prion genes, genes that expresses molecules that induce angiogenesis, genes for adhesion molecules, cell surface receptors, proteins involved in metastasis, proteases, apoptosis genes, cell cycle control genes, genes that express EGF and the EGF receptor, multi-drug resistance genes, such as the MDR1 gene.
  • Expression of a target gene can be determined by any suitable method now known in the art or that is later developed. It can be appreciated that the method used to measure the expression of a target gene will depend upon the nature of the target gene. For example, when the target gene encodes a protein the term “expression” can refer to a protein or transcript derived from the gene. In such instances the expression of a target gene can be determined by measuring the amount of mRNA corresponding to the target gene or by measuring the amount of that protein. Protein can be measured in protein assays such as by staining or immunoblotting or, if the protein catalyzes a reaction that can be measured, by measuring reaction rates. All such methods are known in the art and can be used.
  • RNA species expression can be measured by determining the amount of RNA corresponding to the gene product.
  • Several specific methods for detecting gene expression are described in Example 1. The measurements can be made on cells, cell extracts, tissues, tissue extracts or any other suitable source material.
  • the determination of whether the expression of a target gene has been reduced can be by any suitable method that can reliably detect changes in gene expression. Typically, the determination is made by introducing into the environment of a cell undigested dsRNA such that at least a portion of that dsRNA enters the cytoplasm and then measuring the expression of the target gene. The same measurement is made on identical untreated cells and the results obtained from each measurement are compared. When the method appears to reduce the expression of the target gene by about 10% or more (which is equivalent to about 90% or less) of the level in an untreated organism, for purposes of this invention, the method is considered to reduce the expression of the target gene. Typically, the method can be used to reduce the expression of a target gene by far more than 10%.
  • the method can be used to reduce the expression by about 50% or more, in more preferred methods the expression is reduced by about 75% or more, still more preferable are methods that reduce the expression by about 90% or more, or even about 95% or more, or about 99% or more or even by completely eliminating expression of the target gene.
  • the dsRNA can be formulated as a pharmaceutical composition which comprises a pharmacologically effective amount of a dsRNA and pharmaceutically acceptable carrier.
  • a pharmacologically or therapeutically effective amount refers to that amount of a dsRNA effective to produce the intended pharmacological, therapeutic or preventive result.
  • the phrases “pharmacologically effective amount” and “therapeutically effective amount” or simply “effective amount” refer to that amount of an RNA effective to produce the intended pharmacological, therapeutic or preventive result.
  • a therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 20% reduction in that parameter.
  • pharmaceutically acceptable carrier refers to a carrier for the administration of a therapeutic agent.
  • exemplary carriers include saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
  • pharmaceutically acceptable carriers include, but are not limited to pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives.
  • suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents.
  • Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract.
  • the pharmaceutically acceptable carrier of the disclosed dsRNA composition may be micellar structures, such as a liposomes, capsids, capsoids, polymeric nanocapsules, or polymeric microcapsules.
  • Polymeric nanocapsules or microcapsules facilitate transport and release of the encapsulated or bound dsRNA into the cell. They include polymeric and monomeric materials, especially including polybutylcyanoacrylate. A summary of materials and fabrication methods has been published (see J. Kreuter, (1991) Nanoparticles-preparation and applications. In: M. Donbrow (Ed.) Microcapsules and nanoparticles in medicine and pharmacy. CRC Press, Boca Raton, Fla., pp. 125-14).
  • polymeric materials which are formed from monomeric and/or oligomeric precursors in the polymerization/nanoparticle generation step, are per se known from the prior art, as are the molecular weights and molecular weight distribution of the polymeric material which a person skilled in the field of manufacturing nanoparticles may suitably select in accordance with the usual skill.
  • compositions of this invention can be administered by any means known in the art such as by parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), rectal, vaginal and topical (including buccal and sublingual) administration.
  • parenteral routes including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), rectal, vaginal and topical (including buccal and sublingual) administration.
  • a suitable dosage unit of dsRNA will be in the range of 0.001 to 0.25 milligrams per kilogram body weight of the recipient per day, preferably in the range of 0.01 to 20 micrograms per kilogram body weight per day, more preferably in the range of 0.01 to 10 micrograms per kilogram body weight per day, even more preferably in the range of 0.10 to 5 micrograms per kilogram body weight per day, and most preferably in the range of 0.1 to 2.5 micrograms per kilogram body weight per day.
  • pharmaceutical composition comprising the dsRNA is administered once daily.
  • the therapeutic agent may also be dosed in dosage units containing two, three, four, five, six or more sub-doses administered at appropriate intervals throughout the day.
  • the dsRNA contained in each sub-dose must be correspondingly smaller in order to achieve the total daily dosage unit.
  • the dosage unit can also be compounded for a single dose over several days, e.g., using a conventional sustained release formulation which provides sustained and consistent release of the dsRNA over a several day period. Sustained release formulations are well known in the art.
  • the dosage unit contains a corresponding multiple of the daily dose.
  • the pharmaceutical composition must contain dsRNA in a quantity sufficient to inhibit expression of the target gene in the animal or human being treated.
  • the composition can be compounded in such a way that the sum of the multiple units of dsRNA together contain a sufficient dose.
  • Data can be obtained from cell culture assays and animal studies to formulate a suitable dosage range for humans.
  • the dosage of compositions of the invention lies, preferably, within a range of circulating concentrations that include the ED50 (as determined by known methods) with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range of the compound that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC50 concentration of the test compound which achieves a half-maximal inhibition of symptoms
  • levels of dsRNA in plasma may be measured by standard methods, for example, by high performance liquid chromatography.
  • RNA oligonucleotides were synthesized using solid phase phosphoramidite chemistry, deprotected and desalted on NAP-5 columns (Amersham Pharmacia Biotech, Piscataway, N.J.) using standard techniques (Damha and Olgivie, Methods Mol Biol 1993, 20:81-114; Wincott et al., Nucleic Acids Res 1995, 23:2677-84).
  • the oligomers were purified using ion-exchange high performance liquid chromatography (IE-HPLC) on an Amersham Source 15Q column (1.0 cm ⁇ 25 cm) (Amersham Pharmacia Biotech, Piscataway, N.J.) using a 15 min step-linear gradient.
  • IE-HPLC ion-exchange high performance liquid chromatography
  • the gradient varied from 90:10 Buffers A:B to 52:48 Buffers A:B, where Buffer A was 100 mM Tris pH 8.5 and Buffer B was 100 mM Tris pH 8.5, 1 M NaCl. Samples were monitored at 260 nm and peaks corresponding to the full-length oligonucleotide species were collected, pooled, desalted on NAP-5 columns, and lyophilized.
  • each oligomer was determined by capillary electrophoresis (CE) on a Beckman PACE 5000 (Beckman Coulter, Inc., Fullerton, Calif.).
  • the CE capillaries had a 100 ⁇ m inner diameter and contained ssDNA 100R Gel (Beckman-Coulter).
  • Typically, about 0.6 nmole of oligonucleotide was injected into a capillary, ran in an electric field of 444 V/cm and detected by UV absorbance at 260 nm.
  • Denaturing Tris-Borate-7 M-urea running buffer was purchased from Beckman-Coulter. Oligoribonucleotides were at least 90% pure as assessed by CE for use in experiments described below.
  • ssRNA Single-Stranded RNA Oligomers were resuspended at 100 ⁇ M concentration in duplex buffer consisting of 100 mM potassium acetate, 30 mM HEPES, pH 7.5. Complementary sense and antisense strands were mixed in equal molar amounts to yield a final solution of 50 ⁇ M duplex. Samples were heated to 95° C. for 5′ and allowed to cool to room temperature before use. Double-stranded RNA (dsRNA) oligomers were stored at ⁇ 20° C. Single-stranded RNA oligomers were stored lyophilized or in nuclease-free water at ⁇ 80° C.
  • dsRNA Double-stranded RNA
  • duplexes contains two RNA strands both of which are 21 nucleotides in length, also termed a 21-mer siRNA duplex, and having a 2 base 3′-overhang.
  • a “21-2” design is a 21-mer siRNA duplex with a 2 base 5′-overhang.
  • a 21-0 design is a 21-mer siRNA duplex with no overhangs (blunt).
  • a “21+2UU” is a 21-mer duplex with 2-base 3′-overhang and the terminal 2 bases at the 3′-ends are both U residues (which may result in mismatch with target sequence).
  • dsRNAs having strands that are 25 nucleotides in length or longer have surprisingly increased potency in mammalian systems than known 21-23-mer siRNAs.
  • HEK Human embryonic kidney
  • FBS fetal bovine serum
  • Opti-MEM media 50 ⁇ l was mixed with nucleic acids, including siRNA duplexes and/or 100-200 ng plasmid pEGFP-C1 (Clontech, Palo Alto, Calif.) for 5 min.
  • Nucleic acids were then mixed with 50 ⁇ l of Opti-MEM media that had been pre-mixed with 1.5 ⁇ l of Lipofectamine 2000 and incubated at room temperature for 15 min.
  • the lipid-nucleic acid mixtures were added to cells after removal of old media and swirled and then an additional 0.4 ml of media pre-warmed to 37° C. was added. Incubation was continued at 37° C. and cells were assayed for fluorescence at the times indicated. Each assay was performed in triplicate.
  • EGFP expression levels were measured by direct fluorescence in a fluorescence-activated cell sorter (FACS) (Moslo-MLS, Dako Cytomation, Fort Collins, Colo.) in the City of Hope Cytometrics Core Facility (Duarte, Calif.). EGFP expression was measured as the percentage of cells showing detectable fluorescence above background (mock-transfected negative control cells).
  • FACS fluorescence-activated cell sorter
  • NIH 3T3 cells that stably expressed EGFP were grown in DMEM media supplemented with 10% FBS.
  • Cells were plated at 30% density on 24-well plates and transfected with siRNA alone without reporter plasmid using the same method described above. Media was changed at 24 h and EGFP assays were performed at 3, 6 and 9 days post-transfection. At 3 days post-transfection, 1 ⁇ 10 5 cells were used for extract preparation and 1 ⁇ 10 4 cells were re-plated and continued incubation for later assay. At day 6, 1 ⁇ 10 5 cells were used for extract preparation, and 1 ⁇ 10 4 cells were re-plated and continued incubation for later assay.
  • 1 ⁇ 10 5 cells were used for extract preparation.
  • 1 ⁇ 10 5 cells were suspended in 300 ⁇ l phosphate buffered saline (PBS) and sonicated for 10 sec. Cells were centrifuged at 14,000 g for 2 min and cell supernatant was recovered for fluorometry.
  • EGFP fluorescence was examined using a VersaFluor Cuvette Fluorometer (Bio-Rad, Hercules, Calif.) using excitation filter D490 and emission filter D520. Percentage of EGFP expression was determined relative to extract prepared from non-transfected control cells.
  • the reporter system employed EGFP either as a transfection plasmid vector pEGFP-C1 (Clontech, Palo Alto, Calif.) or as a stable transformant in an NIH 3T3 cell line.
  • the coding sequence of EGFP is shown below, from Genbank accession #U55763. The ATG start codon and TAA stop codons are highlighted in bold font and sites target by siRNA reagents are underscored.
  • Site-1 used for siRNA targeting in EGFP was:
  • Site-2 used for siRNA targeting in EGFP was:
  • RNA duplexes were synthesized and prepared as described in Example 1. RNA duplexes targeting EGFP Site-1 are summarized in Table 1 below. Some sequences had the dinucleotide sequence “UU” placed at the 3′-end of the sense strand (Elbashir et al., 2001, EMBO J., 20:6877-6888; Hohjoh, 2002, FEBS Lett., 521:195-199). Mismatches that resulted from including 3′-terminal “UU” or where a mismatch was intentionally positioned are highlighted in bold and underscored.
  • HEK 293 cells were mock transfected (negative control), transfected with 200 ng EGFP reporter plasmid alone (positive control), or reporter plasmid+siRNA duplexes at varying concentrations. EGFP expression was assessed using the FACS assay at 24 h post-transfection. Results are shown in FIG. 1 . At 50 nM concentration ( FIG. 1A ), a 21-mer siRNA duplex with 2-base 3′-overhang (21+2 design) (SEQ ID NoS. 6/7) showed about a 70% reduction in EGFP expression while longer duplexes were more potent. 25-mer siRNA duplexes (SEQ ID Nos. 16/17 and 18/19) and longer (SEQ ID Nos.
  • 21-mer siRNA oligos are employed at 10-100 nM concentration by those skilled in the art.
  • 1 nM concentration FIG. 1B
  • a concentration much lower than is typically employed today, the 21-mer duplex showed only about a 40% reduction in EGFP expression while longer duplexes continued to suppress EGFP below detection limits.
  • 200 ⁇ M concentration FIG. 1C
  • the 21-mer duplex had no effect on EGFP expression while the blunt 27-mer duplex continued to suppress EGFP below detection limits.
  • FIG. 1D the 27-mer blunt duplex (SEQ ID No.
  • dsRNAs of the invention have about a 100-fold or more higher potency than traditional 21-mer siRNAs.
  • the enhanced effect was first seen at about a 25-mer length and maximal potency was achieved with a 27-mer.
  • Potent RNAi effects were observed for 30-mer duplexes (the longest compounds tested herein), with no apparent toxicity to either the HEK 293 cells or NIH 3T3 cells.
  • duplex length was increased above 25-mer length (presumably when the duplex is sufficiently long to be a Dicer substrate), a 2-base 3′-overhang (as taught in the prior art) is no longer necessary.
  • duplexes that included base mismatches between the sense and antisense strands were more potent than the duplex having perfect complementarity (SEQ ID Nos. 28/29).
  • These duplexes (27+0UU, 27+2UU, and 27 ⁇ 2UU) had 1 or 2 mismatches at the 3′-end of the sense strand.
  • the set of 27-mer duplexes were compared for effective suppression of EGFP expression in the HEK 293 cell transient transfection assay and the results are shown in FIG. 3 .
  • Duplex 27+0UU SEQ ID Nos. 30/31) was most potent.
  • RNAi effector molecules To overcome the need to pool or engage in large scale empiric testing, complex design rules and algorithms have been devised to increase the likelihood of obtaining active RNAi effector molecules (Schwarz et al., 2003, Cell, 115:199-208; Khvorova et al., 2003, Cell, 115:209-216; Ui-Tei et al., 2004, Nucleic Acids Res., 32:936-948; Reynolds et al., 2004, Nat. Biotechnol., 22:326-330). These design rules significantly limit the number of sites amenable to RNAi knockdown within a given target gene. In fact, the design can be overly restrictive in situations demanding the suppression of specific alleles or isoforms.
  • RNA duplexes targeting EGFP Site-2 are summarized in Table 2 below.
  • Duplex EGFPS2-27+0 mm was a blunt 27-mer duplex with a 2 base mismatch at the terminal 2 bases of the sense strand. These bases are shown in bold and underscored.
  • HEK 293 cells were mock transfected (negative control), transfected with 200 ng EGFP reporter plasmid alone (positive control), or reporter plasmid+RNA duplexes at varying concentrations as described previously. EGFP expression was assessed using the FACS assay at 24 h post-transfection. Results are shown in FIG. 4 . At 10 nM concentration, the traditional 21-mer siRNA duplex with 2-base 3′-overhangs targeted to Site-2 in the EGFP gene (SEQ ID Nos. 44/45) did not detectably reduce EGFP expression. In contrast, a 10 nM concentration of the longer 27-mer duplex RNA (SEQ ID No.
  • dsRNAs of the invention can efficiently target sites within the EGFP gene that were previously considered poor targets by previously known methods. Use of the method of the invention will therefore simplify site selection and design criteria for RNAi. This example also shows that the intentional placement of mismatches at the 3′-terminus of the sense strand increases the potency of the 27-mer duplex.
  • This example demonstrates the use of the disclosed dsRNA duplexes to reduce expression of the human HNRPH1 gene in HEK 293 cells.
  • HEK 293 cells were cultured in a 6-well plate. At 30% confluence, cells were transfected with iRNA duplexes as outlined in Example 2 above except that all reagents were used at 5-fold higher volume due to the larger scale of the cultures.
  • Cells were harvested at 72 h in 300 ⁇ l phosphate buffered saline (PBS) and sonicated for 10 sec. Cell lysates was centrifuged for 2 min at 14,000 g and the supernatant was collected. Aliquots of 2 ⁇ l were taken from the cleared lysates which were run on a 10% SDS-PAGE gel.
  • PBS phosphate buffered saline
  • the HNRPH1 gene product was detected using a rabbit polyclonal anti-HNRPH1 antiserum and an anti-rabbit antibody conjugated with alkaline phosphatase (Sigma, St. Louis, Mo.).
  • ⁇ -actin was detected by a murine anti-human actin antibody (Sigma, St. Louis, Mo.) and anti-mouse antibody conjugated with alkaline phosphatase (Sigma, St. Louis, Mo.), as previously described (Markovtsov et al., 2000, Mol. Cell. Biol., 20:7463-79).
  • HNRPH1 Homo sapiens heterogeneous nuclear ribonucleoprotein H1 (HNRPH1) mRNA is shown (Genbank accession No. NM — 005520) below. The ATG start codon and TAA stop codons are highlighted in bold font and site target by siRNA reagents is underscored.
  • RNA duplexes were synthesized and prepared as described in Example 1. RNA duplexes targeting HNRPH1 are summarized in Table 3 below.
  • HNRPH1 Site-1 Sequence Name SEQ ID No. 5′ UGAACUUGAAUCAGAAGAUGAAGUCAAAUUGGC 3′ HNRPH1 Site-1 SEQ ID NO: 69 5′ CUUGAAUCAGAAGAUGAAGUU HNRPH1-21 + 2 SEQ ID No. 51 3′ UUGAACUUAGUCUUCUACUUC SEQ ID No. 52 5′ AACUUGAAUCAGAAGAUGAAGUCAAAU HNRPH1-27 + 0 SEQ ID No. 53 3′ UUGAACUUAGUCUUCUACUUCAGUUUA SEQ ID No. 54
  • HEK 293 cells were transfected with EGFP-specific siRNA (SEQ ID No. 6/7) (negative control) and an HNRPH1 specific 21-mer siRNA duplex (SEQ ID Nos. 51/52) at varying concentrations, or with an HNRPH1 specific 27-mer siRNA duplex (SEQ ID Nos. 53/54) at varying concentrations, as described previously.
  • HNRPH1 expression was assessed by Western Blot assay at 72 h post-transfection. Results are shown in FIG. 5 .
  • This example demonstrates a method for determining whether a dsRNA serves as s substrate for Dicer.
  • RNA duplex RNA oligonucleotides In vitro Dicer assay. Recombinant human Dicer enzyme (Gene Therapy Systems, San Diego, Calif.) was incubated with synthetic duplex RNA oligonucleotides according to the manufacturer's instructions. Briefly, 2 units of Dicer was incubated in a buffer supplied by the manufacturer with 250 pmoles of RNA duplex in a 50 ⁇ l volume (5 ⁇ M RNA concentration) for 18 h at 37° C. Half of each reaction was separated on non-denaturing PAGE (10% acrylamide) and visualized using ethidium bromide staining with UV excitation.
  • RNA duplexes tested included 21-mer (SEQ ID No. 6/7), 25-mer (SEQ ID No. 18/19), 26-mer (SEQ ID No. 24/25), 27-mer (SEQ ID No. 30/31), and 29-mer (SEQ ID No. 40/41).
  • the duplexes were subjected to Dicer digestion in vitro and visualized by PAGE. Results are shown in FIG. 6 .
  • the 21-mer RNA duplex did not react with Dicer.
  • the 25-mer, 26-mer, 27-mer, and 29-mer duplexes all reacted with Dicer and were digested to a 21-mer size product, predominantly.
  • RNA duplexes used in the method of the invention are substrates for the Dicer endoribonuclease.
  • 21-mer siRNAs could result from the action of Dicer on longer duplex RNAs.
  • 7 different 21-mer species could result from degradation of a 27-mer sequence. It is possible that one of these 21-mers (or a combination of 21-mers) accounts for the activity observed with the previously tested 27-mer dsRNA. This example shows that no single 21-mer duplex or mixture of 21-mers resulting from degradation of a 27-mer sequence functions as effectively as its parent 27-mer duplex at reducing EGFP expression.
  • RNA duplexes were prepared as described in Example 1. The sequences of a set of 21-mer RNA duplexes from within EGFP Site-1 were prepared. The duplexes are listed below in Table 4. The 21-mer duplexes are aligned beneath the parent 27-mer to illustrate their relative positioning. The 27-mer blunt duplex (SEQ ID No. 28/29) and the 21-mer duplex 21+2(7) (SEQ ID No. 6/7) are shown in Example 2 (Table 1) and were also used.
  • the each of the 21-mer duplexes from Table 4 was transfected individually or together as a pool into HEK 293 cells with 200 ng of EGFP reporter plasmid as described previously. The result from each transfection was compared with the 27-mer duplex (SEQ ID No. 28/29). The relative EGFP expression from each experiment is shown in FIG. 7 . At concentrations of 50 or 200 ⁇ M, none of the individual 21-mer duplexes or the pooled set of 7 21-mer duplexes showed activity comparable with the 27-mer duplex. For pools, 50 ⁇ M and 200 ⁇ M represent the total concentration of all RNAs transfected together, rather than for individual duplexes. FIG. 7 shows that the potency of the 27-mer duplex was much higher than for any of the shorter 21-mer sequences, which included every possible 21-mer duplex that could result from degradation of the parent 27-mer.
  • the activity of “diced” products made from digestion of the 27-mer duplex with recombinant Dicer enzyme in vitro was compared with the parent 27-mer compound in RNAi assays.
  • the 27-mer duplex (SEQ ID No. 28/29) was degraded using Dicer as described in Example 5 above and fragments (“diced” products) were diluted and directly used in transfection experiments.
  • EGFP expression levels were measured following transfection of HEK 293 cells with 200 ng EGFP reporter plasmid with a 21-mer duplex (SEQ ID No. 6/7), a 27-mer duplex (SEQ ID No.
  • This example provides another demonstration that the improved potency of dsRNA 27-mers is not derived from a highly active individual or a pooled set of short 21-mer duplexes.
  • RNAi Suppression of gene expression using synthetic siRNA typically has a duration of 3-4 days in tissue culture (Chiu and Rana, 2002, Mol. Cell, 10:549-561). Methods that increase the duration of the RNAi effect would improve the functional utility of RNAi as an experimental tool in tissue culture and would be beneficial for use of RNAi in vivo.
  • NIH 3T3 cells were transfected with 5 nM of either the 21-mer duplex (SEQ ID No. 6/7) or the 27-mer duplex (SEQ ID No. 30/31).
  • Cell extracts were prepared and measured for EGFP protein expression in a cuvette fluorometer (as described in Example 2 above) at 2, 4, 6, 8, and 10 days post-transfection. Results are shown in FIG. 9 .
  • images of cells that were transfected in parallel using 1 nM siRNAs were obtained using fluorescence microscopy (as described in Example 2) and the images are shown in FIG. 10 .
  • EGFP expression was suppressed to about 70% of control levels at day 4 but returned to about 80% of control levels at day 6 and was at control levels at day 8.
  • suppression using the 27-mer duplex was about 90% or more at day 8 and was still at about 70% on day 10.
  • the 27-mer duplexes used in the present example demonstrates suppression of gene expression for at least twice the duration seen using 21-mer duplexes.
  • RNA I experiments because, in addition to suppressing gene expression, they avoid interferon activation. Because the more active double stranded RNAs of this invention are longer than known siRNA duplexes, they were examined further to show that they do not activate interferon. Duplexes of up to about 30-mer lengths were tested.
  • HEK 293 cells were transfected with 20 nM T7 ssRNA (Kim et al., 2004, Nat. Biotechnol., 22:321-325), 21-mer RNA duplex (SEQ ID No. 6/7), or 27-mer RNA duplex (SEQ ID No. 30/31) as described in Example 2 above.
  • Culture medium was collected at 24 h and subjected to interferon alpha and beta ELISA assays (Research Diagnostics, Inc., Flanders, N.J.) according to the manufacturer's instructions, as previously described (Kim et al., 2004, Nat. Biotechnol., 22:321-325).
  • dsRNA Human double-stranded RNA
  • PSR protein kinase
  • HEK 293 cells were transfected with ssRNA, 21-mer duplex, 27-mer duplex, or no RNA (negative control, mock transfection) as described above. As shown in FIG. 11 , high levels of interferon alpha ( FIG. 1A ) and interferon beta ( FIG. 1B ) were detected after transfection with ssRNA however no interferon was detected when 21-mer or 27-mer RNAs were transfected.
  • HEK 293 cells were transfected with 400 bp EGFP dsRNA (at 20 nM), or 20 nM of chemically synthesized short RNA duplexes including 21+2 (SEQ ID No. 6/7), 25+2 (SEQ ID No. 18/19), 25-2 (SEQ ID No. 16/17), 27+2 (SEQ ID No. 34/35), and 27-2 (SEQ ID No. 32/33).
  • Results of the PKR activation assay are shown in FIG. 12 .
  • the long dsRNA resulted in strong PKR activation (positive control) while all of the short synthetic RNAs showed no evidence for PKR activation.
  • RNAi mediated suppression of gene expression do not activate interferon responses and therefore should be usable in a wide variety of mammalian systems.
  • a therapeutically effective amount of a composition containing a sequence that encodes a dsRNA is an amount that inhibits expression of the product of the target gene by at least 10 percent. Higher percentages of inhibition, e.g., 20, 50, 90 95, 99 percent or higher may be preferred in certain circumstances.
  • Exemplary doses include milligram or microgram amounts of the molecule per kilogram of subject or sample weight (e.g., about 1 microgram per kilogram to about 5 milligrams per kilogram, about 100 micrograms per kilogram to about 0.5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram).
  • compositions can be administered one or more times per week for between about 1 to 10 weeks, e.g., between 2 to 8 weeks, or between about 3 to 7 weeks, or for about 4, 5, or 6 weeks, as deemed necessary by the attending physician.
  • Treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or a series of treatments.
  • Appropriate doses of a particular dsRNA composition depend upon the potency of the molecule with respect to the expression or activity to be modulated.
  • One or more of these molecules can be administered to an animal, particularly a mammal, and especially humans, to modulate expression or activity of one or more target genes.
  • a physician may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained.
  • the specific dose level for any particular subject will depend upon a variety of other factors including the severity of the disease, previous treatment regimen, other diseases present, off-target effects of the active agent, age, body weight, general health, gender, and diet of the patient, the time of administration, the route of administration, the rate of excretion, any drug combination, and the degree of expression or activity to be modulated.
  • the efficacy of treatment can be monitored by measuring the amount of the target gene mRNA (e.g. using real time PCR) or the amount of product encoded by the target gene such as by Western blot analysis.
  • the attending physician can monitor the symptoms associated with the disease or disorder afflicting the patient and compare with those symptoms recorded prior to the initiation of treatment

Abstract

The invention provides compositions and methods for selectively reducing the expression of a gene product from a desired target gene, as well as treating diseases caused by expression of the gene. The method involves introducing into the environment of a cell an amount of a double-stranded RNA (dsRNA) such that a sufficient portion of the dsRNA can enter the cytoplasm of the cell to cause a reduction in the expression of the target gene. The dsRNA has a first oligonucleotide sequence that is between 26 and about 30 nucleotides in length and a second oligonucleotide sequence that anneals to the first sequence under biological conditions. In addition, a region of one of the sequences of the dsRNA having a sequence length of from about 19 to about 23 nucleotides is complementary to a nucleotide sequence of the RNA produced from the target gene.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • The present application is a division of U.S. patent application Ser. No. 12/137,914 filed 12 Jun. 2008, which in turn is a division of U.S. patent application Ser. No. 11/079,476 filed 15 Mar. 2005, which in turn is related to and claims priority under 35 U.S.C. § 119(e) to U.S. provisional patent application No. 60/553,487 filed 15 Mar. 2004. Each application is incorporated herein by reference.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
  • This invention was made in part with Government support under Grant Numbers A129329 and HL074704 awarded by the National Institute of Health. The Government has certain rights in this invention.
  • FIELD OF THE INVENTION
  • This invention pertains to compositions and methods for gene-specific inhibition of gene expression by double-stranded ribonucleic acid (dsRNA) effector molecules. The compositions and methods are useful in modulating gene expression in a variety of applications, including therapeutic, diagnostic, target validation, and genomic discovery.
  • BACKGROUND OF THE INVENTION
  • Suppression gene expression by double-stranded RNA (dsRNA) has been demonstrated in a variety of systems including plants (post-transcriptional gene suppression) (Napoli et al., 1990, Plant Cell. 2:279-289), fungi (quelling) (Romano and Marcino, 1992, Mol. Microbiol. 6:3343-53), and nematodes (RNA interference) (Fire et al., 1998, Nature 391:806-811). Early attempts to similarly suppress gene expression using long dsRNAs in mammalians systems failed due to activation of interferon pathways that do not exist in lower organisms. Interferon responses are triggered by dsRNAs (Stark et al., 1998, Annu. Rev. Biochem., 67:227-264). In particular, the protein kinase PKR is activated by dsRNAs of greater than 30 bp long (Manche et al., 1992, Mol Cell Biol., 12:5238-48) and results in phosphorylation of translation initiation factor eIF2α which leads to arrest of protein synthesis and activation of 2′5′-oligoadenylate synthetase (2′-5′-OAS), which leads to RNA degradation (Minks et al., 1979, J. Biol. Chem. 254:10180-10183).
  • In Drosophila cells and cell extracts, dsRNAs of 150 bp length or greater were seen to induce RNA interference while shorter dsRNAs were ineffective (Tuschl et al., 1999, Genes & Dev., 13:3191-3197). Long double-stranded RNA, however, is not the active effecter molecule; long dsRNAs are degraded by an RNase III class enzyme called Dicer (Bernstein et al., 2001, Nature, 409:363-366) into very short 21-23 bp duplexes that have 2-base 3′-overhangs (Zamore et al., 2000, Cell, 101:25-33). These short RNA duplexes, called siRNAs, direct the RNAi response in vivo and transfection of short chemically synthesized siRNA duplexes of this design permits use of RNAi methods to suppress gene expression in mammalian cells without triggering unwanted interferon responses (Elbashir et al., 2001, Nature, 411:494-498). The antisense strand of the siRNA duplex serves as a sequence-specific guide that directs activity of an endoribonuclease function in the RNA induced silencing complex (RISC) to degrade target mRNA (Martinez et al., 2002, Cell, 110:563-574).
  • In studying the size limits for RNAi in Drosophila embryo extracts in vitro, a lower threshold of around 38 bp double-stranded RNA was established for activation of RNA interference using exogenously supplied double-stranded RNA and duplexes of 36, 30, and 29 bp length were without effect (Elbashir et al., 2001, Genes & Dev., 15:188-200). The short 30-base RNAs were not cleaved into active 21-23-base siRNAs and therefore were deemed inactive for use in RNAi (Elbashir et al., 2001, Genes & Dev., 15:188-200). Continuing to work in the Drosophila embryo extract system, the same group later carefully mapped the structural features needed for short chemically synthesized RNA duplexes to function as siRNAs in RNAi pathways. RNA duplexes of 21-bp length with 2-base 3′-overhangs were most effective, duplexes of 20, 22, and 23-bp length had slightly decreased potency but did result in RNAi mediated mRNA degradation, and 24 and 25-bp duplexes were inactive (Elbashir et al., 2001, EMBO J., 20:6877-6888).
  • Some of the conclusions of these earlier studies may be specific to the Drosophila system employed. Other investigators established that longer siRNAs can work in human cells. However, duplexes in the 21-23-bp range have been shown to be more active and have become the accepted design (Caplen et al., 2001, Proc. Natl. Acad. Sci. USA, 98:9742-9747). Essentially, chemically synthesized duplex RNAs that mimicked the natural products that result from Dicer degradation of long duplex RNAs were identified to be the preferred compound for use in RNAi. Approaching this problem from the opposite direction, investigators studying size limits for RNAi in C. elegans found that although a microinjected 26-bp RNA duplex could function to suppress gene expression, it required a 250-fold increase in concentration compared with an 81-bp duplex (Parrish et al., 2000, Mol. Cell, 6:1077-1087).
  • Despite the attention given to RNAi research recently, the field is still in the early stages of development. Not all siRNA molecules are capable of targeting the destruction of their complementary RNAs in a cell. As a result, complex sets of rules have been developed for designing RNAi molecules that will be effective. Those having skill in the art expect to test multiple siRNA molecules to find functional compositions. (Ji et al. 2003) Some artisans pool several siRNA preparations together to increase the chance of obtaining silencing in a single study. (Ji et al. 2003) Such pools typically contain 20 nM of a mixture of siRNA oligonucleotide duplexes or more (Ji et al. 2003), despite the fact that a siRNA molecule can work at concentrations of 1 nM or less (Holen et al. 2002). This technique can lead to artifacts caused by interactions of the siRNA sequences with other cellular RNAs (“off target effects”). (Scherer et al. 2003) Off target effects can occur when the RNAi oligonucleotides have homology to unintended targets or when the RISC complex incorporates the unintended strand from and RNAi duplex. (Scherer et al. 2003) Generally, these effects tend to be more pronounced when higher concentrations of RNAi duplexes are used. (Scherer et al. 2003)
  • In addition, the duration of the effect of an effective RNAi treatment is limited to about 4 days (Holen et al. 2002). Thus, researchers must carry out siRNA experiments within 2-3 days of transfection with an siRNA duplex or work with plasmid or viral expression vectors to obtain longer term silencing.
  • Additional physical studies are needed to more completely characterize the structural requirements of RNAi active oligonucleotide duplexes to identify more potent and longer lasting compositions and/or methods that simplify site-selection difficulties. These studies should also include a detailed analysis of the interferon response. Ideally, such studies will be useful in identifying new RNAi active compounds that are more potent, that simplify the site selection process, and decrease “off target effects.”
  • The invention provides RNAi compositions with increased potency, duration of action, and decreased “off target effects” that do not activate the interferon response and provides methods for their use. In addition, the compositions ease site selection criteria and provide a duration of action that is about twice as long as prior known compositions. These and other advantages of the invention, as well as additional inventive features, will be apparent from the description of the invention provided herein.
  • BRIEF SUMMARY OF THE INVENTION
  • The invention provides improved compositions and methods for selectively reducing the expression of a gene product from a desired target gene in a eukaryotic cell, as well as for treating diseases caused by the expression of the gene. The method involves introducing into the environment of a cell an amount of a double-stranded RNA (dsRNA) such that a sufficient portion of the dsRNA can enter the cytoplasm of the cell to cause a reduction in the expression of the target gene. The dsRNA has a first oligonucleotide sequence that is between 26 and about 30 nucleotides in length and a second oligonucleotide sequence that anneals to the first sequence under biological conditions, such as the conditions found in the cytoplasm of a cell. In addition, a region of one of the sequences of the dsRNA having a sequence length of from about 19 to about 23 nucleotides is complementary to a nucleotide sequence of the RNA produced from the target gene. A dsRNA composition of the invention is at least as active as any isolated 19, 20, 21, 22, or 23 basepair sequence that is contained within it. Pharmaceutical compositions containing the disclosed dsRNA compositions are also contemplated. The compositions and methods give a surprising increase in the potency and duration of action of the RNAi effect. Although the invention is not intended to be limited by the underlying theory on which it is believed to operate, it is thought that this increase in potency and duration of action are caused by the fact the dsRNA serves as a substrate for Dicer which appears to facilitate incorporation of one sequence from the dsRNA into the RISC complex that is directly responsible for destruction of the RNA from the target gene.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 provides a comparison of RNAi efficacy using several dsRNAs having variable length and formats including a two nucleotide 3′ overhang (+2), a two nucleotide 5′ overhang (−2), and blunt ends (+0). The sequences are disclosed in the Example 2. In each panel A-D 200 μg of reporter vector was co-transfected with the indicated concentration of dsRNA. Each bar represents the average of three duplicate experiments. In FIG. 1A, 50 nM of each dsRNA was used. In FIG. 1B, 1 nM of each dsRNA was used. In FIG. 1C, 200 pM of each dsRNA was used. In FIG. 1D, 50 pM of each dsRNA was used.
  • FIG. 2 shows an RNAi assay in 3T3 cells expressing endogenous EGFP. The experimental procedure is described in Example 2. Measurements were made 4 days after treatment. The dose response curves for 21-mer duplex with 2-base 3′-overhang (SEQ ID No. 6/7), 25-mer duplex with 2-base 5′-overhang (SEQ ID Nos. 16/17), and blunt 27-mer duplex (SEQ ID Nos. 30/31) are shown.
  • FIG. 3 shows RNAi assays of various 27-mer RNA duplex formats as outlined in Example 2. Duplex 27+0UU (SEQ ID Nos. 30/31) was most potent.
  • FIG. 4 shows RNAi assays on HEK 293 cells that were either mock transfected (negative control), transfected with 200 ng EGFP reporter plasmid alone (positive control), or reporter plasmid+RNA duplexes at varying concentrations as described in Example 3.
  • FIG. 5 shows superior knockout of the HNRPH1 gene by a 27-mer of the invention as compared to a 21-mer directed to the same target. Western blots obtained from HEK 293 cells after transfection with EGFP-specific siRNA (SEQ ID No. 6/7; C) (negative control) and an HNRPH1 specific 21-mer siRNA duplex (SEQ ID Nos. 51/52; 21+2) at varying concentrations, or with an HNRPH1 specific 27-mer siRNA duplex (SEQ ID Nos. 53/54; 27+0) at varying concentrations, as described in Example 4.
  • FIG. 6 shows the reaction of Dicer with various length RNA duplexes as described in Example 5. Dicer was able to digest 25-29-mers primarily into about a 21 basepair duplex but dd not digest the 21 nucleotide long test duplex.
  • FIG. 7 shows the relative expression of EGFP after RNAi assays using a 27-mer dsRNA versus shorter 21-mer siRNAs contained within the 27-mer sequence as described in more detail in Example 6. As shown a blunt ended 27-mer that covers a poor site for a 21 nucleotide RNAi can effectively target that site.
  • FIG. 8 shows the results of RNAi assays after treatment by various effector dsRNA molecules and pools of molecules as set forth in Example 6.
  • FIG. 9 shows the time course study of the duration of the RNAi effect with various effector molecules as described in Example 7. The study shows the duration of the RNAi effect is at least about twice as long with the 27-mer dsRNA of the invention as with 21-mers. The “27+0 UU” sequences are set forth in SEQ ID NOs:28 and 29. The “Mut-16” sequences are set forth in SEQ ID NOs:70 and 71. The “Mut-16,17” sequences are set forth in SEQ ID NOs:72 and 73. The “Mut-15,16,17” sequences are set forth in SEQ ID NOs:74 and 75.
  • FIG. 10 shows the images of cells in a time course study of the duration of the RNAi effect with various effector molecules as described in Example 7. The study shows the duration of the RNAi effect is at least about twice as long with the 27-mer dsRNA of the invention as with 21-mers.
  • FIG. 11 shows that neither interferon alpha (FIG. 11A) or interferon beta (FIG. 11B) are induced by the 27-mer dsRNA of the invention as described in more detail in Example 8.
  • FIG. 12 shows the results of a PKR activation assay in which long dsRNA resulted in strong PKR activation (positive control) while all of the short synthetic RNAs showed no evidence for PKR activation.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention is directed to compositions that contain double stranded RNA (“dsRNA”), and methods for preparing them, that are capable of reducing the expression of target genes in eukaryotic cells. One of the strands of the dsRNA contains a region of nucleotide sequence that has a length that ranges from about 19 to about 23 nucleotides that can direct the destruction of the RNA transcribed from the target gene.
  • For purposes of the invention a suitable dsRNA contains one oligonucleotide sequence, a first sequence, that is at least 25 nucleotides in length and no longer than about 30 nucleotides. More preferably this sequence of RNA is between about 26 and 29 nucleotides in length. Still more preferably this sequence is about 27 or 28 nucleotides in length, 27 nucleotides is most preferred. The second sequence of the dsRNA can be any sequence that anneals to the first sequence under biological conditions, such as within the cytoplasm of a eukaryotic cell. Generally, the second oligonucleotide sequence will have at least 19 complementary base pairs with the first oligonucleotide sequence, more typically the second oligonucleotides sequence will have about 21 or more complementary base pairs, and more preferably about 25 or more complementary base pairs with the first oligonucleotide sequence. In a preferred embodiment the second sequence is the same length as the first sequence.
  • In certain embodiments the double-stranded RNA structure the first and second oligonucleotide sequences exist on separate oligonucleotide strands which can be and typically are chemically synthesized. In preferred embodiments both strands are between 26 and 30 nucleotides in length. In one preferred embodiment both strands are 27 nucleotides in length, are completely complementary and have blunt ends. The dsRNA can be from a single RNA oligonucleotide that undergoes intramolecular annealing or, more typically, the first and second sequences exist on separate RNA oligonucleotides.
  • Suitable dsRNA compositions that contain two separate oligonucleotides can be chemically linked outside their annealing region by chemical linking groups. Many suitable chemical linking groups are known in the art and can be used. Suitable groups will not block Dicer activity on the dsRNA and will not interfere with the directed destruction of the RNA transcribed from the target gene.
  • The first and second oligonucleotide sequences are not required to be completely complimentary. In fact, it is preferred that the 3′-terminus of the sense strand contains one or more mismatches. It is more preferred that two mismatches be incorporated at the 3′terminus. In a most preferred embodiment the dsRNA of the invention is a double stranded RNA molecule containing two RNA oligonucleotides each of which is 27 nucleotides in length and, when annealed to each other, have blunt ends and a two nucleotide mismatch on the 3′-terminus of the sense strand (the 5′-terminus of the antisense strand).
  • One feature of the dsRNA compositions of the invention is that they can serve as a substrate for Dicer. Typically, the dsRNA compositions of this invention will not have been treated with Dicer, other RNAses, or extracts that contain them. Such treatments could digest the dsRNA to lengths of less than 25 nucleotides that are no longer Dicer substrates. Several methods are known and can be used for determining whether a dsRNA composition serves as a substrate for Dicer. For example, Dicer activity can be measured in vitro using the Recombinant Dicer Enzyme Kit (GTS, San Diego, Calif.) according to the manufacturer's instructions. Dicer activity can be measured in vivo by treating cells with dsRNA and maintaining them for 24 h before harvesting them and isolating their RNA. RNa can be isolated using standard methods, such as with the RNeasy™ Kit (Qiagen) according to the manufacturer's instructions. The isolated RNA can be separated on a 10% PAGE gel which is used to prepare a standard RNA blot that can be probed with a suitable labeled deoxyoligonucleotide, such as an oligonucleotide labeled with the Starfire™ Oligo Labeling System (Integrated DNA Technologies, Inc., Coralville, Iowa).
  • It has been found empirically that these longer dsRNA species of from 25 to about 30 nucleotides give unexpectedly improved results in terms of increased potency and increased duration of action over shorter prior art RNAi compositions. The dsRNA compositions of the invention are at least as active as any isolated 23 nucleotide dsRNA sequence contained within them and in preferred embodiments more active. Without wishing to be bound by the underlying theory of the invention, it is thought that the longer dsRNA species serve as a substrate for the enzyme Dicer in the cytoplasm of a cell. In addition to cleaving the dsRNA of the invention into shorter segments, Dicer is thought to facilitate the incorporation of a single-stranded cleavage product derived from the cleaved dsRNA into the RISC complex that is responsible for the destruction of the cytoplasmic RNA derived from the target gene. Studies have shown that the cleavability of a dsRNA species by Dicer corresponds with increased potency and duration of action of the dsRNA species.
  • Suitable dsRNA compositions of this invention do not induce apoptosis in the cells in which they are used. Apoptosis or “programmed cell death,” includes any non-necrotic, cell-regulated form of cell death, as defined by criteria well established in the art. Cells undergoing apoptosis show characteristic morphological and biochemical features. Once the process is triggered, or the cells are committed to undergoing apoptosis, morphological and physiological changes include cell shrinkage, chromatin condensation, nuclear and cytoplasmic condensation, membrane blebbing, partitioning of cytoplasm and nucleus into membrane bound vesicles which contain ribosomes (apoptotic bodies), and DNA degradation into a characteristic oligonucleosomal ladder composed of multiples of 200 base pairs, leading eventually to cell death. In vivo, these apoptotic bodies are rapidly recognized and phagocytized by either macrophages or adjacent epithelial cells. In vitro, the apoptotic bodies as well as the remaining cell fragments ultimately swell and finally lyse. This terminal phase of in vitro cell death has been termed “secondary necrosis.”
  • The effect that a dsRNA has on a cell can depend upon the cell itself. In some circumstances a dsRNA could induce apoptosis or gene silencing in one cell type and not another. Thus, it is possible that a dsRNA could be suitable for use in one cell and not another. To be considered “suitable” a dsRNA composition need not be suitable under all possible circumstances in which it might be used, rather it need only be suitable under a particular set of circumstances.
  • Modifications can be included in the disclosed dsRNA so long as the dsRNA remains sufficiently chemically stable, does not induce apoptosis, does not substantially interrupt annealing of the first and second strands, and otherwise does not substantially interfere with the directed destruction of the RNA transcribed from the target gene. Modifications can be incorporated in the 3′-terminal region, the 5′-terminal region, in both the 3′-terminal and 5′-terminal region or in some instances throughout the sequence. With the restrictions noted above in mind any number and combination of modifications can be incorporated into the dsRNA. Where multiple modifications are present, they may be the same or different. Modifications to bases, sugar moieties, the phosphate backbone, and their combinations are contemplated.
  • For example, either the 3′ or 5′ terminal regions of the sequences in a dsRNA can be phosphorylated or biotinylated. Examples of modifications contemplated for the phosphate backbone include phosphonates, including methylphosphonate, phosphorothioate, and phosphotriester modifications such as alkylphosphotriesters, and the like. Examples of modifications contemplated for the sugar moiety include 2′-alkyl pyrimidine, such as 2′-O-methyl, 2′-fluoro, amino, and deoxy modifications and the like. Examples of modifications contemplated for the base groups include abasic sugars, 2-O-alkyl modified pyrimidines, 4-thiouracil, 5-bromouracil, 5-iodouracil, and 5-(3-aminoallyl)-uracil and the like. Many other modifications are known and can be used so long as the above criteria are satisfied
  • The double-stranded RNA sample can be suitably formulated and introduced into the environment of the cell by any means that allows for a sufficient portion of the sample to enter the cell to induce gene silencing, if it is to occur. Many formulations for dsRNA are known in the art and can be used so long as dsRNA gains entry to the target cells so that it can act. For example, dsRNA can be formulated in buffer solutions such as phosphate buffered saline solutions, liposomes, micellar structures, and capsids. Formulations of dsRNA with cationic lipids can be used to facilitate transfection of the dsRNA into cells. Suitable lipids include Oligofectamine, Lipofectamine (Life Technologies), NC388 (Ribozyme Pharmaceuticals, Inc., Boulder, Colo.), or FuGene 6 (Roche) all of which can be used according to the manufacturer's instructions.
  • It can be appreciated that the method of introducing dsRNA into the environment of the cell will depend on the type of cell and the make up of its environment. For example, when the cells are found within a liquid, one preferable formulation is with a lipid formulation such as in lipofectamine and the dsRNA can be added directly to the liquid environment of the cells. Lipid formulations can also be administered to animals such as by intravenous, intramuscular, or intraperitoneal injection, or orally or by inhalation or other methods as are known in the art. When the formulation is suitable for administration into animals such as mammals and more specifically humans, the formulation is also pharmaceutically acceptable. Pharmaceutically acceptable formulations for administering oligonucleotides are known and can be used. In some instances, it may be preferable to formulate dsRNA in a buffer or saline solution and directly inject the formulated dsRNA into cells, as in studies with oocytes. The direct injection of dsRNA duplexes
  • Suitable amounts of dsRNA must be introduced and these amounts can be empirically determined using standard methods. Typically, effective concentrations of individual dsRNA species in the environment of a cell will be about 50 nanomolar or less 10 nanomolar or less, more preferred are compositions in which concentrations of about 1 nanomolar or less can be used. Even more preferred are methods that utilize a concentration of about 200 picomolar or less and even a concentration of about 50 picomolar or less can be used in many circumstances.
  • The method can be carried out by addition of the dsRNA compositions to any extracellular matrix in which cells can live provided that the dsRNA composition is formulated so that a sufficient amount of the dsRNA can enter the cell to exert its effect. For example, the method is amenable for use with cells present in a liquid such as a liquid culture or cell growth media, in tissue explants, or in whole organisms, including animals, such as mammals and especially humans.
  • As is known, RNAi methods are applicable to a wide variety of genes in a wide variety of organisms and the disclosed compositions and methods can be utilized in each of these contexts. Examples of genes which can be targeted by the disclosed compositions and methods include endogenous genes which are genes that are native to the cell or to genes that are not normally native to the cell. Without limitation these genes include oncogenes, cytokine genes, idiotype (Id) protein genes, prion genes, genes that expresses molecules that induce angiogenesis, genes for adhesion molecules, cell surface receptors, proteins involved in metastasis, proteases, apoptosis genes, cell cycle control genes, genes that express EGF and the EGF receptor, multi-drug resistance genes, such as the MDR1 gene.
  • Expression of a target gene can be determined by any suitable method now known in the art or that is later developed. It can be appreciated that the method used to measure the expression of a target gene will depend upon the nature of the target gene. For example, when the target gene encodes a protein the term “expression” can refer to a protein or transcript derived from the gene. In such instances the expression of a target gene can be determined by measuring the amount of mRNA corresponding to the target gene or by measuring the amount of that protein. Protein can be measured in protein assays such as by staining or immunoblotting or, if the protein catalyzes a reaction that can be measured, by measuring reaction rates. All such methods are known in the art and can be used. Where the gene product is an RNA species expression can be measured by determining the amount of RNA corresponding to the gene product. Several specific methods for detecting gene expression are described in Example 1. The measurements can be made on cells, cell extracts, tissues, tissue extracts or any other suitable source material.
  • The determination of whether the expression of a target gene has been reduced can be by any suitable method that can reliably detect changes in gene expression. Typically, the determination is made by introducing into the environment of a cell undigested dsRNA such that at least a portion of that dsRNA enters the cytoplasm and then measuring the expression of the target gene. The same measurement is made on identical untreated cells and the results obtained from each measurement are compared. When the method appears to reduce the expression of the target gene by about 10% or more (which is equivalent to about 90% or less) of the level in an untreated organism, for purposes of this invention, the method is considered to reduce the expression of the target gene. Typically, the method can be used to reduce the expression of a target gene by far more than 10%. In some instances the method can be used to reduce the expression by about 50% or more, in more preferred methods the expression is reduced by about 75% or more, still more preferable are methods that reduce the expression by about 90% or more, or even about 95% or more, or about 99% or more or even by completely eliminating expression of the target gene.
  • The dsRNA can be formulated as a pharmaceutical composition which comprises a pharmacologically effective amount of a dsRNA and pharmaceutically acceptable carrier. A pharmacologically or therapeutically effective amount refers to that amount of a dsRNA effective to produce the intended pharmacological, therapeutic or preventive result. The phrases “pharmacologically effective amount” and “therapeutically effective amount” or simply “effective amount” refer to that amount of an RNA effective to produce the intended pharmacological, therapeutic or preventive result. For example, if a given clinical treatment is considered effective when there is at least a 20% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 20% reduction in that parameter.
  • The phrase pharmaceutically acceptable carrier refers to a carrier for the administration of a therapeutic agent. Exemplary carriers include saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. For drugs administered orally, pharmaceutically acceptable carriers include, but are not limited to pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract. The pharmaceutically acceptable carrier of the disclosed dsRNA composition may be micellar structures, such as a liposomes, capsids, capsoids, polymeric nanocapsules, or polymeric microcapsules.
  • Polymeric nanocapsules or microcapsules facilitate transport and release of the encapsulated or bound dsRNA into the cell. They include polymeric and monomeric materials, especially including polybutylcyanoacrylate. A summary of materials and fabrication methods has been published (see J. Kreuter, (1991) Nanoparticles-preparation and applications. In: M. Donbrow (Ed.) Microcapsules and nanoparticles in medicine and pharmacy. CRC Press, Boca Raton, Fla., pp. 125-14). The polymeric materials which are formed from monomeric and/or oligomeric precursors in the polymerization/nanoparticle generation step, are per se known from the prior art, as are the molecular weights and molecular weight distribution of the polymeric material which a person skilled in the field of manufacturing nanoparticles may suitably select in accordance with the usual skill.
  • Suitably formulated pharmaceutical compositions of this invention can be administered by any means known in the art such as by parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), rectal, vaginal and topical (including buccal and sublingual) administration.
  • In general a suitable dosage unit of dsRNA will be in the range of 0.001 to 0.25 milligrams per kilogram body weight of the recipient per day, preferably in the range of 0.01 to 20 micrograms per kilogram body weight per day, more preferably in the range of 0.01 to 10 micrograms per kilogram body weight per day, even more preferably in the range of 0.10 to 5 micrograms per kilogram body weight per day, and most preferably in the range of 0.1 to 2.5 micrograms per kilogram body weight per day. Preferably, pharmaceutical composition comprising the dsRNA is administered once daily. However, the therapeutic agent may also be dosed in dosage units containing two, three, four, five, six or more sub-doses administered at appropriate intervals throughout the day. In that case, the dsRNA contained in each sub-dose must be correspondingly smaller in order to achieve the total daily dosage unit. The dosage unit can also be compounded for a single dose over several days, e.g., using a conventional sustained release formulation which provides sustained and consistent release of the dsRNA over a several day period. Sustained release formulations are well known in the art. In this embodiment, the dosage unit contains a corresponding multiple of the daily dose. Regardless of the formulation, the pharmaceutical composition must contain dsRNA in a quantity sufficient to inhibit expression of the target gene in the animal or human being treated. The composition can be compounded in such a way that the sum of the multiple units of dsRNA together contain a sufficient dose.
  • Data can be obtained from cell culture assays and animal studies to formulate a suitable dosage range for humans. The dosage of compositions of the invention lies, preferably, within a range of circulating concentrations that include the ED50 (as determined by known methods) with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range of the compound that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels of dsRNA in plasma may be measured by standard methods, for example, by high performance liquid chromatography.
  • The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope.
  • Example 1
  • This example demonstrates the preparation of double-stranded RNA oligonucleotides
  • Oligonucleotide synthesis and purification. RNA oligonucleotides were synthesized using solid phase phosphoramidite chemistry, deprotected and desalted on NAP-5 columns (Amersham Pharmacia Biotech, Piscataway, N.J.) using standard techniques (Damha and Olgivie, Methods Mol Biol 1993, 20:81-114; Wincott et al., Nucleic Acids Res 1995, 23:2677-84). The oligomers were purified using ion-exchange high performance liquid chromatography (IE-HPLC) on an Amersham Source 15Q column (1.0 cm×25 cm) (Amersham Pharmacia Biotech, Piscataway, N.J.) using a 15 min step-linear gradient. The gradient varied from 90:10 Buffers A:B to 52:48 Buffers A:B, where Buffer A was 100 mM Tris pH 8.5 and Buffer B was 100 mM Tris pH 8.5, 1 M NaCl. Samples were monitored at 260 nm and peaks corresponding to the full-length oligonucleotide species were collected, pooled, desalted on NAP-5 columns, and lyophilized.
  • The purity of each oligomer was determined by capillary electrophoresis (CE) on a Beckman PACE 5000 (Beckman Coulter, Inc., Fullerton, Calif.). The CE capillaries had a 100 μm inner diameter and contained ssDNA 100R Gel (Beckman-Coulter). Typically, about 0.6 nmole of oligonucleotide was injected into a capillary, ran in an electric field of 444 V/cm and detected by UV absorbance at 260 nm. Denaturing Tris-Borate-7 M-urea running buffer was purchased from Beckman-Coulter. Oligoribonucleotides were at least 90% pure as assessed by CE for use in experiments described below. Compound identity was verified by matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectroscopy on a Voyager DE™ Biospectometry Work Station (Applied Biosystems, Foster City, Calif.) following the manufacturer's recommended protocol. Relative molecular masses of all oligomers were within 0.2% of expected molecular mass.
  • Preparation of Duplexes. Single-Stranded RNA (ssRNA) Oligomers were resuspended at 100 μM concentration in duplex buffer consisting of 100 mM potassium acetate, 30 mM HEPES, pH 7.5. Complementary sense and antisense strands were mixed in equal molar amounts to yield a final solution of 50 μM duplex. Samples were heated to 95° C. for 5′ and allowed to cool to room temperature before use. Double-stranded RNA (dsRNA) oligomers were stored at −20° C. Single-stranded RNA oligomers were stored lyophilized or in nuclease-free water at −80° C.
  • Nomenclature. For consistency, the following nomenclature has been employed throughout the Examples. Names given to duplexes indicate the length of the oligomers and the presence or absence of overhangs. A “21+2” duplex contains two RNA strands both of which are 21 nucleotides in length, also termed a 21-mer siRNA duplex, and having a 2 base 3′-overhang. A “21-2” design is a 21-mer siRNA duplex with a 2 base 5′-overhang. A 21-0 design is a 21-mer siRNA duplex with no overhangs (blunt). A “21+2UU” is a 21-mer duplex with 2-base 3′-overhang and the terminal 2 bases at the 3′-ends are both U residues (which may result in mismatch with target sequence).
  • Example 2
  • This example demonstrates that dsRNAs having strands that are 25 nucleotides in length or longer have surprisingly increased potency in mammalian systems than known 21-23-mer siRNAs.
  • Cell Culture, Transfection, and EGFP Assays. Human embryonic kidney (HEK) 293 cells were grown in DMEM medium supplemented with 10% fetal bovine serum (FBS) (Irvine Scientific, Santa Ana, Calif.). Transfections were done at 90% confluence in 24-well plates using Lipofectamine 2000 (Invitrogen, Carlsbad, Calif.) according to the manufacturer's instructions. Briefly, 50 μl of Opti-MEM media was mixed with nucleic acids, including siRNA duplexes and/or 100-200 ng plasmid pEGFP-C1 (Clontech, Palo Alto, Calif.) for 5 min. Nucleic acids were then mixed with 50 μl of Opti-MEM media that had been pre-mixed with 1.5 μl of Lipofectamine 2000 and incubated at room temperature for 15 min. The lipid-nucleic acid mixtures were added to cells after removal of old media and swirled and then an additional 0.4 ml of media pre-warmed to 37° C. was added. Incubation was continued at 37° C. and cells were assayed for fluorescence at the times indicated. Each assay was performed in triplicate. EGFP expression levels were measured by direct fluorescence in a fluorescence-activated cell sorter (FACS) (Moslo-MLS, Dako Cytomation, Fort Collins, Colo.) in the City of Hope Cytometrics Core Facility (Duarte, Calif.). EGFP expression was measured as the percentage of cells showing detectable fluorescence above background (mock-transfected negative control cells).
  • NIH 3T3 cells that stably expressed EGFP (Kim and Rossi, 2003, Antisense Nucleic Acid Drug Dev., 13:151-155) were grown in DMEM media supplemented with 10% FBS. Cells were plated at 30% density on 24-well plates and transfected with siRNA alone without reporter plasmid using the same method described above. Media was changed at 24 h and EGFP assays were performed at 3, 6 and 9 days post-transfection. At 3 days post-transfection, 1×105 cells were used for extract preparation and 1×104 cells were re-plated and continued incubation for later assay. At day 6, 1×105 cells were used for extract preparation, and 1×104 cells were re-plated and continued incubation for later assay. At day 9, 1×105 cells were used for extract preparation. For extract preparation, 1×105 cells were suspended in 300 μl phosphate buffered saline (PBS) and sonicated for 10 sec. Cells were centrifuged at 14,000 g for 2 min and cell supernatant was recovered for fluorometry. EGFP fluorescence was examined using a VersaFluor Cuvette Fluorometer (Bio-Rad, Hercules, Calif.) using excitation filter D490 and emission filter D520. Percentage of EGFP expression was determined relative to extract prepared from non-transfected control cells.
  • In addition, cells were directly examined by fluorescence microscopy using a Nikon Eclipse TE2000-S (Nikon Instech Co., Kanagawa, JP) using the program Spot v3.5.8. Images were digitally captured with identical exposure times so that comparisons between cells samples could be made.
  • Nucleic Acid Reagents. The reporter system employed EGFP either as a transfection plasmid vector pEGFP-C1 (Clontech, Palo Alto, Calif.) or as a stable transformant in an NIH 3T3 cell line. The coding sequence of EGFP is shown below, from Genbank accession #U55763. The ATG start codon and TAA stop codons are highlighted in bold font and sites target by siRNA reagents are underscored.
  • SEQ ID NO. 1:
    atggtgagcaagggcgaggagctgttcaccggggtggtgcccatcctggtcgagctggacggcgacgtaaacggcca
    caagttcagcgtgtccggcgagggcgagggcgatgccacctacggcaagctgaccctgaagttcatctgcaccaccg
    gcaagctgcccgtgccctggcccaccctcgtgaccaccctgacctacggcgtgcagtgcttcagccgctaccccgac
    cacatgaagcagcacgacttcttcaagtccgccatgcccgaaggctacgtccaggagcgcaccatcttcttcaagga
    cgacggcaactacaagacccgcgccgaggtgaagttcgagggcgacaccctggtgaaccgcatcgagctgaagggca
    tcgacttcaaggaggacggcaacatcctggggcacaagctggagtacaactacaacagccacaacgtctatatcatg
    gccgacaagcagaagaacggcatcaaggtgaacttcaagatccgccacaacatcgaggacggcagcgtgcagctcgc
    cgaccactaccagcagaacacccccatcggcgacggccccgtgctgctgcccgacaaccactacctgagcacccagt
    ccgccctgagcaaagaccccaacgagaagcgcgatcacatggtcctgctggagttcgtgaccgccgccgggatcact
    ctcggcatggacgagctgtacaagtaa
  • Site-1 used for siRNA targeting in EGFP was:
  • SITE 1:
    (SEQ ID NO: 67)
    5′ GCAAGCUGACCCUGAAGUUCAUCUGCACCACCGGCAAGC 3′
  • Site-2 used for siRNA targeting in EGFP was:
  • SITE 2:
    (SEQ ID NO: 68)
    5′ UGAAGCAGCACGACUUCUUCAAGUCCGCCAUG 3′
  • RNA duplexes were synthesized and prepared as described in Example 1. RNA duplexes targeting EGFP Site-1 are summarized in Table 1 below. Some sequences had the dinucleotide sequence “UU” placed at the 3′-end of the sense strand (Elbashir et al., 2001, EMBO J., 20:6877-6888; Hohjoh, 2002, FEBS Lett., 521:195-199). Mismatches that resulted from including 3′-terminal “UU” or where a mismatch was intentionally positioned are highlighted in bold and underscored.
  • TABLE 1
    Summary of Oligonucleotide Reagents, EGFP Site-1
    Sequence Name SEQ ID NOo.
    5′ GCAAGCUGACCCUGAAGUUCAUCUGCACCACCGGCAAGC 3′ EGFP Site-1 SEQ ID NO: 67
    5′ GCAAGCUGACCCUGAAGUUCA EGFPS1-21 − 2 SEQ ID No. 2
    3′ UUCGACUGGGACUUCAAGUAG SEQ ID NO. 3
    5′ AAGCUGACCCUGAAGUUCAUC EGFPS1-21 + 0 SEQ ID No. 4
    3′ UUCGACUGGGACUUCAAGUAG SEQ ID NO. 5
    5′ GCUGACCCUGAAGUUCAUCUG EGFPS1-21 + 2 (7) SEQ ID No. 6
    3′ UUCGACUGGGACUUCAAGUAG SEQ ID NO. 7
    5′ GCAAGCUGACCCUGAAGUUCAU U EGFPS1-23 − 2UU SEQ ID No. 8
    3′ UUCGACUGGGACUUCAAGUAGAC SEQ ID NO. 9
    5′ GCUGACCCUGAAGUUCAUCUGUU EGFPS1-23 + 2UU SEQ ID No. 10
    3′ UUCGACUGGGACUUCAAGUAGAC SEQ ID NO. 11
    5′ GCAAGCUGACCCUGAAGUUCAU U U EGFPS1-24 − 2UU SEQ ID No. 12
    3′ UUCGACUGGGACUUCAAGUAGACG SEQ ID NO. 13
    5′ GCUGACCCUGAAGUUCAUCUGCUU EGFPS1-24 + 2UU SEQ ID No. 14
    3′ UUCGACUGGGACUUCAAGUAGACG SEQ ID NO. 15
    5′ GCAAGCUGACCCUGAAGUUCAUCU U EGFPS1-25 − 2UU SEQ ID No. 16
    3′ UUCGACUGGGACUUCAAGUAGACGU SEQ ID NO. 17
    5′ GCUGACCCUGAAGUUCAUCUGCAUU EGFPS1-25 + 2UU SEQ ID No. 18
    3′ UUCGACUGGGACUUCAAGUAGACGU SEQ ID NO. 19
    5′ AAGCUGACCCUGAAGUUCAUCUGCAC EGFPS1-26 + 0 SEQ ID No. 20
    3′ UUCGACUGGGACUUCAAGUAGACGUG SEQ ID NO. 21
    5′ AAGCUGACCCUGAAGUUCAUCUGC UU EGFPS1-26 + 0UU SEQ ID No. 22
    3′ UUCGACUGGGACUUCAAGUAGACGUG SEQ ID NO. 23
    5′ GCAAGCUGACCCUGAAGUUCAUCU UU EGFPS1-26 − 2UU SEQ ID No. 24
    3′ UUCGACUGGGACUUCAAGUAGACGUG SEQ ID NO. 25
    5′ GCUGACCCUGAAGUUCAUCUGCACUU EGFPS1-26 + 2UU SEQ ID No. 26
    3′ UUCGACUGGGACUUCAAGUAGACGUG SEQ ID NO. 27
    5′ AAGCUGACCCUGAAGUUCAUCUGCACC EGFPS1-27 + 0 SEQ ID No. 28
    3′ UUCGACUGGGACUUCAAGUAGACGUGG SEQ ID NO. 29
    5′ AAGCUGACCCUGAAGUUCAUCUGCA UU EGFPS1-27 + 0UU SEQ ID No. 30
    3′ UUCGACUGGGACUUCAAGUAGACGUGG SEQ ID NO. 31
    5′ GCAAGCUGACCCUGAAGUUCAUCUG UU EGFPS1-27 − 2UU SEQ ID No. 32
    3′ UUCGACUGGGACUUCAAGUAGACGUGG SEQ ID NO. 33
    5′ GCUGACCCUGAAGUUCAUCUGCAC A UU EGFPS1-27 + 2UU, mut SEQ ID No. 34
    3′ UUCGACUGGGACUUCAAGUAGACGUGG SEQ ID NO. 35
    5′ AAGCUGACCCUGUUCAUCAUCUGCACC EGFPS1-27 + 0-mut SEQ ID No. 36
    3′ UUCGACUGGGACAAGUAGUAGACGUGG SEQ ID NO. 37
    5′ AAGCUGACCCUGAAGUUCAUCUGCACCA EGFPS1-28 + 0 SEQ ID No. 38
    3′ UUCGACUGGGACUUCAAGUAGACGUGGU SEQ ID NO. 39
    5′ AAGCUGACCCUGAAGUUCAUCUGCACCAC EGFPS1-29 + 0 SEQ ID No. 40
    3′ UUCGACUGGGACUUCAAGUAGACGUGGUG SEQ ID NO. 41
    5′ AAGCUGACCCUGAAGUUCAUCUGCACCACC EGFPS1-30 + 0 SEQ ID No. 42
    3′ UUCGACUGGGACUUCAAGUAGACGUGGUGG SEQ ID NO. 43
  • Results. HEK 293 cells were mock transfected (negative control), transfected with 200 ng EGFP reporter plasmid alone (positive control), or reporter plasmid+siRNA duplexes at varying concentrations. EGFP expression was assessed using the FACS assay at 24 h post-transfection. Results are shown in FIG. 1. At 50 nM concentration (FIG. 1A), a 21-mer siRNA duplex with 2-base 3′-overhang (21+2 design) (SEQ ID NoS. 6/7) showed about a 70% reduction in EGFP expression while longer duplexes were more potent. 25-mer siRNA duplexes (SEQ ID Nos. 16/17 and 18/19) and longer (SEQ ID Nos. 20/21, 24/25, 26/27, 30/31, 32/33, and 34/35) suppressed EGFP below detection limits. Typically, 21-mer siRNA oligos are employed at 10-100 nM concentration by those skilled in the art. At 1 nM concentration (FIG. 1B), a concentration much lower than is typically employed today, the 21-mer duplex showed only about a 40% reduction in EGFP expression while longer duplexes continued to suppress EGFP below detection limits. At 200 μM concentration (FIG. 1C), the 21-mer duplex had no effect on EGFP expression while the blunt 27-mer duplex continued to suppress EGFP below detection limits. At 50 μM concentration (FIG. 1D), the 27-mer blunt duplex (SEQ ID No. 30/31) suppressed EGFP expression by about 90% or more. The longer and shorter duplexes tested (26-mers SEQ ID No. 22/23, 24/25, 26/27; 29-mer SEQ ID No. 40/41; or 30-mer SEQ ID No. 42/43) were slightly less effective than the blunt 27-mer.
  • This experiment was repeated using NIH 3T3 cells that stably express EGFP. EGFP protein was detected in cellular extracts using a cuvette fluorometer as described above. The dose response curves for 21-mer duplex with 2-base 3′-overhang (SEQ ID No. 6/7), 25-mer duplex with 2-base 5′-overhang (SEQ ID Nos. 16/17), and blunt 27-mer duplex (SEQ ID Nos. 30/31) are shown in FIG. 2. The exact level of suppression varied between experiments done using stably transfected 3T3 cells compared with transiently transfected HEK 293 cells (FIG. 1), however qualitative trends were identical.
  • This example demonstrates that the longer dsRNAs of the invention have about a 100-fold or more higher potency than traditional 21-mer siRNAs. The enhanced effect was first seen at about a 25-mer length and maximal potency was achieved with a 27-mer. Potent RNAi effects were observed for 30-mer duplexes (the longest compounds tested herein), with no apparent toxicity to either the HEK 293 cells or NIH 3T3 cells. Furthermore, as duplex length was increased above 25-mer length (presumably when the duplex is sufficiently long to be a Dicer substrate), a 2-base 3′-overhang (as taught in the prior art) is no longer necessary. In the present experiments 25-mer duplexes with a 2-base 5′-overhang had similar potency as did blunt ended duplexes or duplexes with a 2-base 3′-overhang. In the current experimental system, the 27-mer blunt duplex showed greatest potency.
  • Within the set of 27-mer RNA duplexes tested in this example, duplexes that included base mismatches between the sense and antisense strands (SEQ ID Nos. 30/31, 32/33, 34/35) were more potent than the duplex having perfect complementarity (SEQ ID Nos. 28/29). These duplexes (27+0UU, 27+2UU, and 27−2UU) had 1 or 2 mismatches at the 3′-end of the sense strand. The set of 27-mer duplexes were compared for effective suppression of EGFP expression in the HEK 293 cell transient transfection assay and the results are shown in FIG. 3. Duplex 27+0UU (SEQ ID Nos. 30/31) was most potent.
  • The use of mismatches or decreased thermodynamic stability (specifically at the 3′-sense/5′-antisense position) has been proposed to facilitate or favor entry of the antisense strand into RISC (Schwarz et al., 2003, Cell, 115:199-208; Khvorova et al., 2003, Cell, 115:209-216), presumably by affecting some rate-limiting unwinding steps that occur with entry of the siRNA into RISC. Because of this terminal base composition has been included in design algorithms for selecting active 21-mer siRNA duplexes (Ui-Tei et al., 2004, Nucleic Acids Res., 32:936-948; Reynolds et al., 2004, Nat. Biotechnol., 22:326-330). It has been proposed that the 27-mer duplexes employed in this example do not directly enter RISC but first are cleaved by Dicer into 21-mer siRNAs. With Dicer cleavage, the small end-terminal sequence which contains the mismatches will either be left unpaired with the antisense strand (become part of a 3′-overhang) or be cleaved entirely off the final 21-mer siRNA. These “mismatches”, therefore, do not persist as mismatches in the final RNA component of RISC. It was surprising to find that base mismatches or destabilization of segments at the 3′-end of the sense strand of a Dicer substrate improve the potency of synthetic duplexes in RNAi, presumably by facilitating processing by Dicer.
  • Example 3
  • This example demonstrates that the use of 25-30 nucleotide RNA duplexes allows gene targeting at a site that could not be effectively targeted using traditional siRNA 21-mer designs.
  • It is currently expected in the art that the majority of 21-mer siRNA duplexes targeted to sites within a given target gene sequence will be ineffective (Holen et al., 2002, Nucleic Acids Res., 30:1757-1766). Consequently, a variety of sites are commonly tested in parallel or pools containing several distinct siRNA duplexes specific to the same target with the hope that one of the reagents will be effective (Ji et al., 2003, FEBS Lett., 552:247-252). To overcome the need to pool or engage in large scale empiric testing, complex design rules and algorithms have been devised to increase the likelihood of obtaining active RNAi effector molecules (Schwarz et al., 2003, Cell, 115:199-208; Khvorova et al., 2003, Cell, 115:209-216; Ui-Tei et al., 2004, Nucleic Acids Res., 32:936-948; Reynolds et al., 2004, Nat. Biotechnol., 22:326-330). These design rules significantly limit the number of sites amenable to RNAi knockdown within a given target gene. In fact, the design can be overly restrictive in situations demanding the suppression of specific alleles or isoforms. Moreover, the rules are not perfect and do not always provide active siRNA effector molecules. This example shows that the use of dsRNA duplexes of the present invention allow RNAi targeting at sites that were ineffectively targeted by previously known 21-mer siRNA reagents. This result minimizes the need for empirically testing multiple sites or using pooled reagent sets.
  • Nucleic Acid Reagents. The reporter system employed EGFP as in SEQ ID No. 1 above. Site-2 in EGFP, as shown in Example 1, was targeted. RNA duplexes were synthesized and prepared as described in Example 1. RNA duplexes targeting EGFP Site-2 are summarized in Table 2 below. Duplex EGFPS2-27+0 mm was a blunt 27-mer duplex with a 2 base mismatch at the terminal 2 bases of the sense strand. These bases are shown in bold and underscored.
  • TABLE 2
    Summary of Oligonucleotide Reagents, EGFP Site-2
    Sequence Name SEQ ID No.
    5′ UGAAGCAGCACGACUUCUUCAAGUCCGCCAUG 3′ EGFP Site-2 SEQ ID NO: 68
    5′ GCAGCACGACUUCUUCAAGUU EGFPS2-21 + 2 SEQ ID No. 44
    3′ UUCGUCGUGCUGAAGAAGUUC SEQ ID No. 45
    5′ AAGCAGCACGACUUCUUCAAGUCCGCC EGFPS2-27 + 0 SEQ ID No. 46
    3′ UUCGUCGUGCUGAAGAAGUUCAGGCGG SEQ ID No. 47
    5′ AAGCAGCACGACUUCUUCAAGUCCG GG EGFPS2-27 + 0 mm SEQ ID No. 48
    3′ UUCGUCGUGCUGAAGAAGUUCAGGCGG SEQ ID No. 49
  • Results. HEK 293 cells were mock transfected (negative control), transfected with 200 ng EGFP reporter plasmid alone (positive control), or reporter plasmid+RNA duplexes at varying concentrations as described previously. EGFP expression was assessed using the FACS assay at 24 h post-transfection. Results are shown in FIG. 4. At 10 nM concentration, the traditional 21-mer siRNA duplex with 2-base 3′-overhangs targeted to Site-2 in the EGFP gene (SEQ ID Nos. 44/45) did not detectably reduce EGFP expression. In contrast, a 10 nM concentration of the longer 27-mer duplex RNA (SEQ ID No. 46/47) reduced EGFP by about 80% or more and 10 nM of a related 27-mer (SEQ ID No. 48/49) reduced EGFP by about 90% or more. As in Example 2 above, 3′- or 5′-overhangs did not improve activity over the blunt ended version. The 27-mer with the 2-base mismatch in the sense strand (SEQ ID Nos. 48/49) showed improved activity as compared to the perfectly matched 27-mer (SEQ ID Nos. 46/47). It is possible that destabilization of the RNA duplex at this position improves efficiency of cleavage by Dicer.
  • This example demonstrates that dsRNAs of the invention can efficiently target sites within the EGFP gene that were previously considered poor targets by previously known methods. Use of the method of the invention will therefore simplify site selection and design criteria for RNAi. This example also shows that the intentional placement of mismatches at the 3′-terminus of the sense strand increases the potency of the 27-mer duplex.
  • Example 4
  • This example demonstrates the use of the disclosed dsRNA duplexes to reduce expression of the human HNRPH1 gene in HEK 293 cells.
  • Western Blot. HEK 293 cells were cultured in a 6-well plate. At 30% confluence, cells were transfected with iRNA duplexes as outlined in Example 2 above except that all reagents were used at 5-fold higher volume due to the larger scale of the cultures. Cells were harvested at 72 h in 300 μl phosphate buffered saline (PBS) and sonicated for 10 sec. Cell lysates was centrifuged for 2 min at 14,000 g and the supernatant was collected. Aliquots of 2 μl were taken from the cleared lysates which were run on a 10% SDS-PAGE gel. The HNRPH1 gene product was detected using a rabbit polyclonal anti-HNRPH1 antiserum and an anti-rabbit antibody conjugated with alkaline phosphatase (Sigma, St. Louis, Mo.). As control, β-actin was detected by a murine anti-human actin antibody (Sigma, St. Louis, Mo.) and anti-mouse antibody conjugated with alkaline phosphatase (Sigma, St. Louis, Mo.), as previously described (Markovtsov et al., 2000, Mol. Cell. Biol., 20:7463-79).
  • Nucleic Acid Reagents. The coding sequence of Homo sapiens heterogeneous nuclear ribonucleoprotein H1 (HNRPH1) mRNA is shown (Genbank accession No. NM005520) below. The ATG start codon and TAA stop codons are highlighted in bold font and site target by siRNA reagents is underscored.
  • SEQ ID No. 50:
    ttttttttttcgtcttagccacgcagaagtcgcgtgtctagtttgtttcgacgccggaccgcgtaagagacgatgat
    gttgggcacggaaggtggagagggattcgtggtgaaggtccggggcttgccctggtcttgctcggccgatgaagtgc
    agaggtttttttctgactgcaaaattcaaaatggggctcaaggtattcgtttcatctacaccagagaaggcagacca
    agtggcgaggcttttgttgaacttgaatcagaagatgaagtcaaattggccctgaaaaaagacagagaaactatggg
    acacagatatgttgaagtattcaagtcaaacaacgttgaaatggattgggtgttgaagcatactggtccaaatagtc
    ctgacacggccaatgatggctttgtacggcttagaggacttccctttggatgtagcaaggaagaaattgttcagttc
    ttctcagggttggaaatcgtgccaaatgggataacattgccggtggacttccaggggaggagtacgggggaggcctt
    cgtgcagtttgcttcacaggaaatagctgaaaaggctctaaagaaacacaaggaaagaatagggcacaggtatattg
    aaatctttaagagcagtagagctgaagttagaactcattatgatccaccacgaaagcttatggccatgcagcggcca
    ggtccttatgacagacctggggctggtagagggtataacagcattggcagaggagctggctttgagaggatgaggcg
    tggtgcttatggtggaggctatggaggctatgatgattacaatggctataatgatggctatggatttgggtcagata
    gatttggaagagacctcaattactgtttttcaggaatgtctgatcacagatacggggatggtggctctactttccag
    agcacaacaggacactgtgtacacatgcggggattaccttacagagctactgagaatgacatttataattttttttc
    accgctcaaccctgtgagagtacacattgaaattggtcctgatggcagagtaactggtgaagcagatgtcgagttcg
    caactcatgaagatgctgtggcagctatgtcaaaagacaaagcaaatatgcaacacagatatgtagaactcttcttg
    aattctacagcaggagcaagcggtggtgcttacgaacacagatatgtagaactcttcttgaattctacagcaggagc
    aagcggtggtgcttatggtagccaaatgatgggaggcatgggcttgtcaaaccagtccagctacgggggcccagcca
    gccagcagctgagtgggggttacggaggcggctacggtggccagagcagcatgagtggatacgaccaagttttacag
    gaaaactccagtgattttcaatcaaacattgcataggtaaccaaggagcagtgaacagcagctactacagtagtgga
    agccgtgcatctatgggcgtgaacggaatgggagggttgtctagcatgtccagtatgagtggtggatggggaatgta
    attgatcgatcctgatcactgactcttggtcaacctttttttttttttttttttctttaagaaaacttcagtttaac
    agtttctgcaatacaagcttgtgatttatgcttactctaagtggaaatcaggattgttatgaagacttaaggcccag
    tatttttgaatacaatactcatctaggatgtaacagtgaagctgagtaaactataactgttaaacttaagttccagc
    ttttctcaagttagttataggatgtacttaagcagtaagcgtatttaggtaaaagcagttgaattatgttaaatgtt
    gccctttgccacgttaaattgaacactgttttggatgcatgttgaaagacatgcttttattttttttgtaaaacaat
    ataggagctgtgtctactattaaaagtgaaacattttggcatgtttgttaattctagtttcatttaataacctgtaa
    ggcacgtaagtttaagctttttttttttttaagttaatgggaaaaatttgagacgcaataccaatacttaggatttt
    ggtcttggtgtttgtatgaaattctgaggccttgatttaaatctttcattgtattgtgatttccttttaggtatatt
    gcgctaagtgaaacttgtcaaataaatcctccttttaaaaactgc
  • RNA duplexes were synthesized and prepared as described in Example 1. RNA duplexes targeting HNRPH1 are summarized in Table 3 below.
  • TABLE 3
    Summary of Oligonucleotide Reagents, HNRPH1 Site-1
    Sequence Name SEQ ID No.
    5′ UGAACUUGAAUCAGAAGAUGAAGUCAAAUUGGC 3′ HNRPH1 Site-1 SEQ ID NO: 69
    5′ CUUGAAUCAGAAGAUGAAGUU HNRPH1-21 + 2 SEQ ID No. 51
    3′ UUGAACUUAGUCUUCUACUUC SEQ ID No. 52
    5′ AACUUGAAUCAGAAGAUGAAGUCAAAU HNRPH1-27 + 0 SEQ ID No. 53
    3′ UUGAACUUAGUCUUCUACUUCAGUUUA SEQ ID No. 54
  • HEK 293 cells were transfected with EGFP-specific siRNA (SEQ ID No. 6/7) (negative control) and an HNRPH1 specific 21-mer siRNA duplex (SEQ ID Nos. 51/52) at varying concentrations, or with an HNRPH1 specific 27-mer siRNA duplex (SEQ ID Nos. 53/54) at varying concentrations, as described previously. HNRPH1 expression was assessed by Western Blot assay at 72 h post-transfection. Results are shown in FIG. 5. As shown, only a slight decrease in HNRPH1 protein levels occurred after treatment with 20 nM of the 21-mer siRNA (SEQ ID Nos 51/52) while significant inhibition was seen using 1 nM of the 27-mer dsRNA (SEQ ID Nos 53/54) and almost complete elimination of HNRPH1 protein was achieved using 5 nM of the 27-mer RNA duplex. Improved reduction in gene expression by RNAi methods is therefore also seen for human genes using the method of the invention.
  • Example 5
  • This example demonstrates a method for determining whether a dsRNA serves as s substrate for Dicer.
  • In vitro Dicer assay. Recombinant human Dicer enzyme (Gene Therapy Systems, San Diego, Calif.) was incubated with synthetic duplex RNA oligonucleotides according to the manufacturer's instructions. Briefly, 2 units of Dicer was incubated in a buffer supplied by the manufacturer with 250 pmoles of RNA duplex in a 50 μl volume (5 μM RNA concentration) for 18 h at 37° C. Half of each reaction was separated on non-denaturing PAGE (10% acrylamide) and visualized using ethidium bromide staining with UV excitation.
  • Results. RNA duplexes tested included 21-mer (SEQ ID No. 6/7), 25-mer (SEQ ID No. 18/19), 26-mer (SEQ ID No. 24/25), 27-mer (SEQ ID No. 30/31), and 29-mer (SEQ ID No. 40/41). The duplexes were subjected to Dicer digestion in vitro and visualized by PAGE. Results are shown in FIG. 6. As shown, the 21-mer RNA duplex did not react with Dicer. The 25-mer, 26-mer, 27-mer, and 29-mer duplexes all reacted with Dicer and were digested to a 21-mer size product, predominantly.
  • This example shows that the longer RNA duplexes used in the method of the invention are substrates for the Dicer endoribonuclease.
  • Example 6
  • This example demonstrates that 27-mer duplexes have more RNAi activity than any of the shorter 21-mer duplexes that they encompass.
  • Theoretically, a variety of short 21-mer siRNAs could result from the action of Dicer on longer duplex RNAs. For example, based upon the antisense strand, 7 different 21-mer species could result from degradation of a 27-mer sequence. It is possible that one of these 21-mers (or a combination of 21-mers) accounts for the activity observed with the previously tested 27-mer dsRNA. This example shows that no single 21-mer duplex or mixture of 21-mers resulting from degradation of a 27-mer sequence functions as effectively as its parent 27-mer duplex at reducing EGFP expression.
  • Nucleic Acid Reagents. RNA duplexes were prepared as described in Example 1. The sequences of a set of 21-mer RNA duplexes from within EGFP Site-1 were prepared. The duplexes are listed below in Table 4. The 21-mer duplexes are aligned beneath the parent 27-mer to illustrate their relative positioning. The 27-mer blunt duplex (SEQ ID No. 28/29) and the 21-mer duplex 21+2(7) (SEQ ID No. 6/7) are shown in Example 2 (Table 1) and were also used.
  • TABLE 4
    Summary of Oligonucleotide Reagents, EGFP Site-1, Tiled Set
    Sequence Name SEQ ID No.
    5′ GCAAGCUGACCCUGAAGUUCAUCUGCACCACCGGCAAGC 3′ EGFP Site-1 SEQ ID NO: 67
    5′ AAGCUGACCCUGAAGUUCAUCUGCACC EGFPS1-27 + 0 SEQ ID No. 28
    3′ UUCGACUGGGACUUCAAGUAGACGUGG SEQ ID No. 29
    5′ CCUGAAGUUCAUCUGCACCAC EGFPS1-21 + 2 (1) SEQ ID No. 55
    3′ UGGGACUUCAAGUAGACGUGG SEQ ID No. 56
    5′ CCCUGAAGUUCAUCUGCACCA EGFPS1-21 + 2 (2) SEQ ID No. 57
    3′ CUGGGACUUCAAGUAGACGUG SEQ ID No. 58
    5′ ACCCUGAAGUUCAUCUGCACC EGFPS1-21 + 2 (3) SEQ ID No. 59
    3′ ACUGGGACUUCAAGUAGACGU SEQ ID No. 60
    5′ GACCCUGAAGUUCAUCUGCAC EGFPS1-21 + 2 (4) SEQ ID No. 61
    3′ GACUGGGACUUCAAGUAGACG SEQ ID No. 62
    5′ UGACCCUGAAGUUCAUCUGCA EGFPS1-21 + 2 (5) SEQ ID No. 63
    3′ CGACUGGGACUUCAAGUAGAC SEQ ID No. 64
    5′ CUGACCCUGAAGUUCAUCUGC EGFPS1-21 + 2 (6) SEQ ID No. 65
    3′ UCGACUGGGACUUCAAGUAGA SEQ ID No. 66
    5′ GCUGACCCUGAAGUUCAUCUG EGFPS1-21 + 2 (7) SEQ ID No. 6
    3′ UUCGACUGGGACUUCAAGUAG SEQ ID No. 7
  • The each of the 21-mer duplexes from Table 4 was transfected individually or together as a pool into HEK 293 cells with 200 ng of EGFP reporter plasmid as described previously. The result from each transfection was compared with the 27-mer duplex (SEQ ID No. 28/29). The relative EGFP expression from each experiment is shown in FIG. 7. At concentrations of 50 or 200 μM, none of the individual 21-mer duplexes or the pooled set of 7 21-mer duplexes showed activity comparable with the 27-mer duplex. For pools, 50 μM and 200 μM represent the total concentration of all RNAs transfected together, rather than for individual duplexes. FIG. 7 shows that the potency of the 27-mer duplex was much higher than for any of the shorter 21-mer sequences, which included every possible 21-mer duplex that could result from degradation of the parent 27-mer.
  • The activity of “diced” products made from digestion of the 27-mer duplex with recombinant Dicer enzyme in vitro was compared with the parent 27-mer compound in RNAi assays. The 27-mer duplex (SEQ ID No. 28/29) was degraded using Dicer as described in Example 5 above and fragments (“diced” products) were diluted and directly used in transfection experiments. EGFP expression levels were measured following transfection of HEK 293 cells with 200 ng EGFP reporter plasmid with a 21-mer duplex (SEQ ID No. 6/7), a 27-mer duplex (SEQ ID No. 28/29), products of in vitro Dicer degradation (“diced” products), a mutant 27-mer with 4 base central mismatch (SEQ ID No. 36/37), and the pooled set of 7 21-mer duplexes (SEQ ID Nos. 6/7 and 55/56, 57/58, 59/60, 61/62, 63/64, 65/66). Results are shown in FIG. 8. Again, the 27-mer duplex was the most potent reagent in reducing EGFP expression. The “diced” products were more effective than the set of pooled 21-mer duplexes. One explanation for this result is that the in vitro dicing reaction was incomplete and some intact 27-mer remains even after 18 h incubation (residual 27-mer is seen in FIG. 6).
  • This example provides another demonstration that the improved potency of dsRNA 27-mers is not derived from a highly active individual or a pooled set of short 21-mer duplexes.
  • Example 7
  • This example demonstrates that gene suppression using the 27-mer duplexes of the invention last twice as long as suppression achieved using 21-mer duplexes.
  • Suppression of gene expression using synthetic siRNA typically has a duration of 3-4 days in tissue culture (Chiu and Rana, 2002, Mol. Cell, 10:549-561). Methods that increase the duration of the RNAi effect would improve the functional utility of RNAi as an experimental tool in tissue culture and would be beneficial for use of RNAi in vivo.
  • NIH 3T3 cells were transfected with 5 nM of either the 21-mer duplex (SEQ ID No. 6/7) or the 27-mer duplex (SEQ ID No. 30/31). Cell extracts were prepared and measured for EGFP protein expression in a cuvette fluorometer (as described in Example 2 above) at 2, 4, 6, 8, and 10 days post-transfection. Results are shown in FIG. 9. In addition, images of cells that were transfected in parallel using 1 nM siRNAs were obtained using fluorescence microscopy (as described in Example 2) and the images are shown in FIG. 10. EGFP expression was suppressed to about 70% of control levels at day 4 but returned to about 80% of control levels at day 6 and was at control levels at day 8. In contrast, suppression using the 27-mer duplex was about 90% or more at day 8 and was still at about 70% on day 10.
  • The 27-mer duplexes used in the present example demonstrates suppression of gene expression for at least twice the duration seen using 21-mer duplexes.
  • Example 8
  • This example demonstrates that the dsRNA duplexes of the invention do not activate the interferon response.
  • Historically, long double stranded RNA was considered to be ineffective as an agent for reducing gene expression in mammalian cells because it tends to activate interferon pathway responses and lead to a variety of metabolic disturbances in cells which are not sequence specific. Short 21-mer siRNAs were considered useful for RNA I experiments because, in addition to suppressing gene expression, they avoid interferon activation. Because the more active double stranded RNAs of this invention are longer than known siRNA duplexes, they were examined further to show that they do not activate interferon. Duplexes of up to about 30-mer lengths were tested.
  • Interferon and PKR assays. HEK 293 cells were transfected with 20 nM T7 ssRNA (Kim et al., 2004, Nat. Biotechnol., 22:321-325), 21-mer RNA duplex (SEQ ID No. 6/7), or 27-mer RNA duplex (SEQ ID No. 30/31) as described in Example 2 above. Culture medium was collected at 24 h and subjected to interferon alpha and beta ELISA assays (Research Diagnostics, Inc., Flanders, N.J.) according to the manufacturer's instructions, as previously described (Kim et al., 2004, Nat. Biotechnol., 22:321-325).
  • Human double-stranded RNA (dsRNA)-dependent protein kinase (PKR) was assayed using the PKR activation assay (Gunnery et al., 1998, Methods, 15:189-98) in HEK 293 cell extracts. HEK 293 cells were transfected as described in Example 2 with 20 nM of each RNA and extracts were prepared 18 h post-transfection.
  • Results. HEK 293 cells were transfected with ssRNA, 21-mer duplex, 27-mer duplex, or no RNA (negative control, mock transfection) as described above. As shown in FIG. 11, high levels of interferon alpha (FIG. 1A) and interferon beta (FIG. 1B) were detected after transfection with ssRNA however no interferon was detected when 21-mer or 27-mer RNAs were transfected.
  • HEK 293 cells were transfected with 400 bp EGFP dsRNA (at 20 nM), or 20 nM of chemically synthesized short RNA duplexes including 21+2 (SEQ ID No. 6/7), 25+2 (SEQ ID No. 18/19), 25-2 (SEQ ID No. 16/17), 27+2 (SEQ ID No. 34/35), and 27-2 (SEQ ID No. 32/33). Results of the PKR activation assay are shown in FIG. 12. The long dsRNA resulted in strong PKR activation (positive control) while all of the short synthetic RNAs showed no evidence for PKR activation.
  • We conclude that the longer synthetic RNAs used in the invention for improved RNAi mediated suppression of gene expression do not activate interferon responses and therefore should be usable in a wide variety of mammalian systems.
  • Example 9
  • This example demonstrates a method for determining an effective dose of the dsRNA of the invention in a mammal. A therapeutically effective amount of a composition containing a sequence that encodes a dsRNA, (i.e., an effective dosage), is an amount that inhibits expression of the product of the target gene by at least 10 percent. Higher percentages of inhibition, e.g., 20, 50, 90 95, 99 percent or higher may be preferred in certain circumstances. Exemplary doses include milligram or microgram amounts of the molecule per kilogram of subject or sample weight (e.g., about 1 microgram per kilogram to about 5 milligrams per kilogram, about 100 micrograms per kilogram to about 0.5 milligrams per kilogram, or about 1 microgram per kilogram to about 50 micrograms per kilogram). The compositions can be administered one or more times per week for between about 1 to 10 weeks, e.g., between 2 to 8 weeks, or between about 3 to 7 weeks, or for about 4, 5, or 6 weeks, as deemed necessary by the attending physician. Treatment of a subject with a therapeutically effective amount of a composition can include a single treatment or a series of treatments.
  • Appropriate doses of a particular dsRNA composition depend upon the potency of the molecule with respect to the expression or activity to be modulated. One or more of these molecules can be administered to an animal, particularly a mammal, and especially humans, to modulate expression or activity of one or more target genes. A physician may, for example, prescribe a relatively low dose at first, subsequently increasing the dose until an appropriate response is obtained. In addition, it is understood that the specific dose level for any particular subject will depend upon a variety of other factors including the severity of the disease, previous treatment regimen, other diseases present, off-target effects of the active agent, age, body weight, general health, gender, and diet of the patient, the time of administration, the route of administration, the rate of excretion, any drug combination, and the degree of expression or activity to be modulated.
  • The efficacy of treatment can be monitored by measuring the amount of the target gene mRNA (e.g. using real time PCR) or the amount of product encoded by the target gene such as by Western blot analysis. In addition, the attending physician can monitor the symptoms associated with the disease or disorder afflicting the patient and compare with those symptoms recorded prior to the initiation of treatment
  • All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
  • The use of the terms “a” and “an” and “the” and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The terms “comprising,” “having,” “including,” and “containing” are to be construed as open-ended terms (i.e., meaning “including, but not limited to,”) unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
  • Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.
  • REFERENCES
    • Amarzguioui et al., Tolerance for Mutation and Chemical Modifications in a siRNA, Nucleic Acids Research, 2003, Vol. 31, No. 2, 589-595.
    • Bernstein, E., Caudy, A. A., Hammond, S. M., and Hannon, G. J. (2001) Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature, 409:363-366.
    • Braasch et al., RNA Interference in Mammalian Cells by Chemically-Modified RNA, Biochemistry, 2003, 42, 7967-7975.
    • Bridge et al., Induction of an Interferon Response by RNAi Vectors in Mammalian Cells, Nature Genetics, 2003, Vol. 34, No. 3, 263-264.
    • Caplen, N. J., Parrish, S., Imani, F., Fire, A., and Morgan, R. A. (2001) Specific inhibition of gene expression by small double-stranded RNAs in invertebrate and vertebrate systems. Proc. Natl. Acad. Sci. USA, 98:9742-9747.
    • Chang et al., Gene Expression from Both Intronless and Intron-Containing Rous Sarcoma Virus Clones is Specifically Inhibited by Anti-Sense RNA, Molecular and Cellular Biology, 1985, Vol. 5, No. 9, 2341-2348.
    • Check, RNA to the Rescue?, Nature, 2003, 425, 10-12.
    • CHIU et al., siRNA Function in RNAi: A Chemical Modification Analysis, RNA, 2003, 9:1034-1048.
    • Chiu, Y. L., and Rana, T. R. (2002) RNAi in human cells: basic structural and functional features of small interfering RNA. Mol. Cell, 10:549-561.
    • Czauderna et al., Structural Variations and Stabilising Modifications of Synthetic siRNAs in Mammalian Cells, Nucleic Acids Research, 2003, Vol. 31, No. 11, 2705-2716.
    • Damha, M. J., and Ogilvie, K. K. (1993) Oligoribonucleotide synthesis. The silyl-phosphoramidite method. Methods Mol. Biol. 20:81-114.
    • Donbrow (Ed.) Microcapsules and nanoparticles in medicine and pharmacy. CRC Press, Boca Raton, Fla., pp. 125-14.
    • Elbashir et al., Functional Anatomy of siRNAs for Mediating Efficient RNAi in Drosophila melanogaster Embryo Lysate, EMBO Journal, 2001, Vol. 20, No. 33, 6877-6888.
    • Elbashir et al., RNA Interference is Mediated by 21- and 22-Nucleotide RNAs, Genes & Development, 2001, 15:188-200.
    • Elbashir, S. M., Harborth, J., Lendeckel, W., Yalcin, A., Weber, K., and Tuschl, T. (2001) Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature, 411:494-498.
    • Elbashir, S. M., Lendeckel, W., and Tuschl, T. (2001) RNA interference is mediated by 21- and 22-nucleotide RNAs. Genes & Dev., 15:188-200.
    • Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A., Driver, S. E., and Mellow, C. C. (1998) Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 391:806-811.
    • Graessmann, M., Michaels, G., Berg, B., and Graessmann, A. (1991) Inhibition of SV40 gene expression by microinjected small antisense RNA and DNA molecules. Nucleic Acids Res. 19:53-59.
    • Gunnery, S., and Mathews, M. B. (1998) RNA binding and modulation of PKR activity. Methods, 15:189-98.
    • Hamada et al., Effects on RNA Interference in Gene Expression (RNAi) in Cultured Mammalian Cells of Mismatches and the Introduction of Chemical Modifications at the 3′-Ends of siRNAs, Antisense and Nucleic Acid Drug Development, 2002, 12:301-309
  • Hannon, RNA Interference, Nature, 2002, 418, 244-251.
    • Harborth, J., Elbashir, S. M., Vandenburgh, K., Manning a, H., Scaringe, S. A., Weber, K., and Tuschl, T. (2003) Sequence, chemical, and structural variation of small interfering RNAs and short hairpin RNAs and the effect on mammalian gene silencing. Antisense Nucleic Acid Drug Dev., 13:83-105.
    • Hohjoh, J. (2002) RNA interference (RNAi) induction with various types of synthetic oligonucleotide duplexes in cultured human cells. FEBS Lett., 521:195-199.
    • Holen, T., Amarzguioui, M., Wilger, M. T., Babaie, E., and Prydz, H. (2002) Positional effects of short interfering RNAs targeting the human coagulation trigger tissue factor. Nucleic Acids Res., 30:1757-1766.
    • Ji, J., Wernli, M., Klimkait, T., and Erb, P. (2003) Enhanced gene silencing by the application of multiple specific small interfering RNAs. FEBS Lett., 552:247-252.
    • Khvorova, A., Reynolds, A., and Jayasena, S. D. (2003) Functional siRNAs and miRNAs exhibit strand bias. Cell, 115:209-216.
    • Kim, D. H., and Rossi, J. J. (2003) Coupling of RNAi-mediated target downregulation with gene replacement. Antisense Nucleic Acid Drug Dev., 13:151-155.
    • Kim, D. H., Longo, M., Han, Y., Lundberg, P., Cantin, E., and Rossi, J. J. (2004) Interferon induction by siRNAs and ssRNAs synthesized by phage polymerase. Nat. Biotechnol., 22:321-325.
    • Kretshmer-Kazemi Far et al., The Activity of siRNA in Mammalian Cells is Related to Structural Target Accessibility: A Comparison with Antisense Oligonucleotides, Nucleic Acids Research, 2003, Vol. 31, No. 15, 4417-4424.
    • Liu et al., R2D2, a Bridge Between the Initiator and Effector Steps of the Drosophila RNAi Pathway, Science, 2003, Vol. 301, 1921-1925.
    • Manche, L., Green, S. R., Schmedt, C., and Mathews, M. B. (1992) Interactions between double-stranded RNA regulators and the protein kinase DAI. Mol. Cell. Biol. 12:5238-5248.
    • Markovtsov, V., Nikolic, J. M., Goldman, J. A., Turck, C. W., Chou, M. Y., and Black, D. L. (2000) Cooperative assembly of an hnRNP complex induced by a tissue specific homolog of polypyrimidine tract binding protein. Mol. Cell. Biol., 20:7463-79.
    • Martinez, J., Patkaniowska, A., Urlaub, H., Luhrmann, R., and Tuschl, T. (2002) Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell, 110:563-574.
    • McManus et al., Gene Silencing in Mammals by Small Interfering RNAs, Nature Reviews Genetics, 2002, Vol. 3, 737-747.
    • Melton, D. A. (1985) Injected anti-sense RNAs specifically block messenger RNA translation in vivo. Proc. Natl. Acad. Sci. USA. 82:144-148.
    • Minks, M. A., West, D. K., Benvin, S., and Baglioni, C. (1979) Structural requirements of double-stranded RNA for the activation of the 2′-5′-oligo(A) polymerase and protein kinase of interferon-treated HeLa cells. J. Biol. Chem. 254:10180-10183.
    • Napoli, C., Lemieux, C., and Jorgensen, R. (1990) Introduction of a chimeric chalcone synthase gene into petunia results in reversible co-suppression of homologous genes in trans. Plant Cell. 2:279-289.
    • Ngo et al., Double-Stranded RNA Induces mRNA Degradation in Trypanosoma brucei, Proceedings of the National Academy of Sciences of the United States of America, 1998, Vol. 95, 14687-14692.
    • Parrish, S., Fleenor, J., Xu, S., Mello, C., and Fire, A. (2000) Functional anatomy of a dsRNA trigger: differential requirements for the two trigger strands in RNA interference. Mol. Cell, 6:1077-1087.
    • Pellino et al., R2D2 Leads the Silencing Trigger to mRNA's Death Star, Cell, 2003, 115:132-133.
    • Reynolds et al., Rational siRNA Design for RNA Interference, Nature Biotechnology, 2004, 22, 1-5.
    • Reynolds, A., Leake, D., Boese, Q., Scaringe, S., Marshall, W. S., and Khvorova, A. (2004) Rational siRNA design for RNA interference. Nat. Biotechnol. 22:326-330.
    • Romano, N., and Macino, G. (1992) Quelling: transient inactivation of gene expression in Neurospora crassa by transformation with homologous sequences. Mol. Microbiol. 6:3343-53.
    • Rossi, presentation at Integrated DNA Technologies, 2003.
    • Scherer et al., Approaches for the Sequence-Specific Knockdown of mRNA, Nature Biotechnology, 2003, Vol. 21, No. 12, 1457-1465.
    • Schwarz, D. S., Hutvagner, G., Du, T., Xu, Z., Aronin, N., and Zamore, P. D. (2003) Asymmetry in the assembly of the RNAi enzyme complex. Cell, 115:199-208.
    • Skipper, Elegant Tour de Force, Nature Reviews Genetics, 2003, Vol. 4, 79-80.
    • Sledz et al., Activation of the Interferon System by Short-Interfering RNAs, Nature Cell Biology, 2003, Vol. 5, No. 9, 834-839.
    • Stark, G. R., Kerr, I. M., Williams, B. R., Silverman, R. H., and Schreiber, R. D. (1998) How cells respond to interferons. Annu. Rev. Biochem. 67:227-264.
    • Tuschl, T., Zamore, P. D., Lehmann, R., Bartel, D. P., and Sharp, P. A. (1999) Targeted mRNA degradation by double-stranded RNA in vitro. Genes & Dev., 13:3191-3197.
    • Ui-Tei, K., Naito, Y., Takahashi, F., Haraguchi, T., Ohki-Hamazaki, H., Juni, A., Ueda, R., and Saigo, K. (2004) Guidelines for the selection of highly effective siRNA sequences for mammalian and chick RNA interference. Nucleic Acids Res., 32:936-948.
    • Waterhouse et al., Exploring Plant Genomes by RNA-Induced Gene Silencing, Nature Reviews Genetics, 2003, Vol. 4, 29-38.
    • Wincott, F., DiRenzo, A., Shaffer, C., Grimm, S., Tracz, D., Workman, C., Sweedler, D., Gonzalez, C., Scaringe, S., and Usman, N. (1995) Synthesis, deprotection, analysis and purification of RNA and ribozymes. Nucleic Acids Res., 23:2677-84.
    • Xu et al., Effective Small Interfering RNAs and Phosphorothioate Antisense DNAs Have Different Preferences for Target Sites in the Luciferase mRNAs, Biochemical and Biophysical Research Communications, 2003, 306, 712-717.
    • Zamore, P. D., Tuschl, T., Sharp, P. A., and Bartel, D. P. (2000) RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell, 101:25-33.

Claims (28)

1. A method for preparing an isolated double stranded nucleic acid that corresponds to a selected target sequence of a target gene, comprising:
synthesizing first and second oligonucleotide strands, wherein each of said first and said second strands has a 5′ terminus and a 3′ terminus, consists of the same number of nucleotide residues and is 25-30 nucleotides, wherein said first and second strands are complementary to each other such that they form a duplex of at least 25 nucleotides, said double stranded nucleic acid comprises blunt ends and the ultimate and penultimate residues of said 3′ terminus of said first strand and the ultimate and penultimate residues of said 5′ terminus of said second strand form one or two mismatched base pairs when said duplex is formed, and wherein said second strand comprises a nucleotide sequence complementary to the selected target sequence such that it anneals to said target sequence under biological conditions, and
annealing said first and said second oligonucleotide strands to form a double stranded nucleic acid, wherein said double stranded nucleic acid reduces target gene expression when introduced into a mammalian cell,
thereby preparing said isolated double stranded nucleic acid.
2. The method of claim 1, wherein said biological conditions are conditions as found in the cytoplasm of a mammalian cell.
3. The method of claim 1, wherein said double stranded nucleic acid is then formulated into a pharmaceutically acceptable excipient.
4. The method of claim 1, wherein the selected target sequence of said target gene comprises at least 19 nucleotides.
5. The method of claim 1, wherein each of said first and second strands of said isolated double stranded nucleic acid has a length which is at least 26 and at most 30 nucleotides.
6. The method of claim 1, wherein said first and second strands of said isolated double stranded nucleic acid are, independently, 27 nucleotide residues in length.
7. The method of claim 1, wherein the ultimate and penultimate residues of said 3′ terminus of said first strand and the ultimate and penultimate residues of said 5′ terminus of said second strand of said isolated double stranded nucleic acid form two mismatched base pairs.
8. The method of claim 1, wherein said 5′ terminus of each of said first and said second strands of said isolated double stranded nucleic acid comprises a 5′ phosphate.
9. The method of claim 1, wherein said second strand is fully complementary to the selected target sequence.
10. The method of claim 1, wherein said isolated double stranded nucleic acid comprises a modified nucleotide selected from the group consisting of a deoxyribonucleotide, a dideoxyribonucleotide, an acyclonucleotide, a 3′-deoxyadenosine (cordycepin), a 3′-azido-3′-deoxythymidine (AZT), a 2′,3′-dideoxyinosine (ddI), a 2′,3′-dideoxy-3′-thiacytidine (3TC), a 2′,3′-didehydro-2′,3′-dideoxythymidine (d4T), a monophosphate nucleotide of 3′-azido-3′-deoxythymidine (AZT), a 2′,3′-dideoxy-3′-thiacytidine (3TC) and a monophosphate nucleotide of 2′,3′-didehydro-2′,3′-dideoxythymidine (d4T), a 4-thiouracil, a 5-bromouracil, a 5-iodouracil, a 5-(3-aminoallyl)-uracil, a 2′-O-alkyl ribonucleotide, a 2′-O-methyl ribonucleotide, a 2′-amino ribonucleotide, a 2′-fluoro ribonucleotide, and a locked nucleic acid.
11. The method of claim 1, wherein said double stranded nucleic acid comprises a phosphate backbone modification selected from the group consisting of a phosphonate, a phosphorothioate and a phosphotriester.
12. The method of claim 1, wherein said double stranded nucleic acid is cleaved endogenously in a mammalian cell to produce a double-stranded nucleic acid of a length in the range of 19-23 nucleotides that reduces target gene expression.
13. The method of claim 1, wherein said double stranded nucleic acid reduces target gene expression when introduced into a mammalian cell in vitro by an amount (expressed by %) selected from the group consisting of at least 10%, at least 50% and at least 80%.
14. The method of claim 1, wherein the first and second strands of said isolated double stranded nucleic acid are joined by a chemical linker.
15. The method of claim 1, wherein said 3′ terminus of said first strand and said 5′ terminus of said second strand of said isolated double stranded nucleic acid are joined by a chemical linker.
16. A method for preparing an isolated double stranded nucleic acid that corresponds to a selected target sequence of a target gene, comprising:
synthesizing first and second oligonucleotide strands, wherein each of said first and said second strands has a 5′ terminus and a 3′ terminus and is 27 nucleotides in length, wherein said first and second strands are complementary to each other such that they form a duplex, said double stranded nucleic acid comprises blunt ends and the ultimate and penultimate residues of said 3′ terminus of said first strand and the ultimate and penultimate residues of said 5′ terminus of said second strand form two mismatched base pairs when said duplex is formed, and wherein said second strand comprises a nucleotide sequence complementary to the selected target sequence such that it anneals to said target sequence under biological conditions, and
annealing said first and said second oligonucleotide strands to form a double stranded nucleic acid, wherein said double stranded nucleic acid reduces target gene expression when introduced into a mammalian cell,
thereby preparing said isolated double stranded nucleic acid.
17. The method of claim 16, wherein said isolated double stranded nucleic acid comprises a modified nucleotide selected from the group consisting of a deoxyribonucleotide, a dideoxyribonucleotide, an acyclonucleotide, a 3′-deoxyadenosine (cordycepin), a 3′-azido-3′-deoxythymidine (AZT), a 2′,3′-dideoxyinosine (ddI), a 2′,3′-dideoxy-3′-thiacytidine (3TC), a 2′,3′-didehydro-2′,3′-dideoxythymidine (d4T), a monophosphate nucleotide of 3′-azido-3′-deoxythymidine (AZT), a 2′,3′-dideoxy-3′-thiacytidine (3TC) and a monophosphate nucleotide of 2′,3′-didehydro-2′,3′-dideoxythymidine (d4T), a 4-thiouracil, a 5-bromouracil, a 5-iodouracil, a 5-(3-aminoallyl)-uracil, a 2′-O-alkyl ribonucleotide, a 2′-O-methyl ribonucleotide, a 2′-amino ribonucleotide, a 2′-fluoro ribonucleotide, and a locked nucleic acid.
18. The method of claim 16, wherein said double stranded nucleic acid comprises a phosphate backbone modification selected from the group consisting of a phosphonate, a phosphorothioate and a phosphotriester.
19. The method of claim 16, wherein said double stranded nucleic acid reduces target gene expression when introduced into a mammalian cell in vitro by an amount (expressed by %) selected from the group consisting of at least 10%, at least 50% and at least 80%.
20. A method for preparing an isolated double stranded nucleic acid that corresponds to a selected target sequence of a target gene, comprising:
synthesizing first and second oligonucleotide strands, wherein each of said first and said second strands has a 5′ terminus and a 3′ terminus, consists of the same number of nucleotide residues and is 25-30 nucleotides, wherein said first and second strands are complementary to each other such that they form a duplex of at least 25 nucleotides, said double stranded nucleic acid comprises blunt ends and the ultimate and penultimate residues of said 5′ terminus of said first strand and the ultimate and penultimate residues of said 3′ terminus of said second strand form one or two mismatched base pairs when said duplex is formed, and wherein said second strand comprises a nucleotide sequence complementary to the selected target sequence such that it anneals to said target sequence under biological conditions, and
annealing said first and said second oligonucleotide strands to form a double stranded nucleic acid, wherein said double stranded nucleic acid reduces target gene expression when introduced into a mammalian cell,
thereby preparing said isolated double stranded nucleic acid.
21. The method of claim 20, wherein the ultimate and penultimate residues of said 5′ terminus of said first strand and the ultimate and penultimate residues of said 3′ terminus of said second strand form two mismatched base pairs.
22. The method of claim 20, wherein said isolated double stranded nucleic acid comprises a modified nucleotide selected from the group consisting of a deoxyribonucleotide, a dideoxyribonucleotide, an acyclonucleotide, a 3′-deoxyadenosine (cordycepin), a 3′-azido-3′-deoxythymidine (AZT), a 2′,3′-dideoxyinosine (ddI), a 2′,3′-dideoxy-3′-thiacytidine (3TC), a 2′,3′-didehydro-2′,3′-dideoxythymidine (d4T), a monophosphate nucleotide of 3′-azido-3′-deoxythymidine (AZT), a 2′,3′-dideoxy-3′-thiacytidine (3TC) and a monophosphate nucleotide of 2′,3′-didehydro-2′,3′-dideoxythymidine (d4T), a 4-thiouracil, a 5-bromouracil, a 5-iodouracil, a 5-(3-aminoallyl)-uracil, a 2′-O-alkyl ribonucleotide, a 2′-O-methyl ribonucleotide, a 2′-amino ribonucleotide, a 2′-fluoro ribonucleotide, and a locked nucleic acid.
23. The method of claim 20, wherein said double stranded nucleic acid comprises a phosphate backbone modification selected from the group consisting of a phosphonate, a phosphorothioate and a phosphotriester.
24. The method of claim 20, wherein said double stranded nucleic acid reduces target gene expression when introduced into a mammalian cell in vitro by an amount (expressed by %) selected from the group consisting of at least 10%, at least 50% and at least 80%.
25. A method for preparing an isolated double stranded nucleic acid that corresponds to a selected target sequence of a target gene, comprising:
synthesizing first and second oligonucleotide strands, wherein each of said first and said second strands has a 5′ terminus and a 3′ terminus and is 27 nucleotides in length, wherein said first and second strands are complementary to each other such that they form a duplex, said double stranded nucleic acid comprises blunt ends and the ultimate and penultimate residues of said 5′ terminus of said first strand and the ultimate and penultimate residues of said 3′ terminus of said second strand form two mismatched base pairs when said duplex is formed, and wherein said second strand comprises a nucleotide sequence complementary to the selected target sequence such that it anneals to said target sequence under biological conditions, and
annealing said first and said second oligonucleotide strands to form a double stranded nucleic acid, wherein said double stranded nucleic acid reduces target gene expression when introduced into a mammalian cell,
thereby preparing said isolated double stranded nucleic acid.
26. The method of claim 25, wherein said isolated double stranded nucleic acid comprises a modified nucleotide selected from the group consisting of a deoxyribonucleotide, a dideoxyribonucleotide, an acyclonucleotide, a 3′-deoxyadenosine (cordycepin), a 3′-azido-3′-deoxythymidine (AZT), a 2′,3′-dideoxyinosine (ddI), a 2′,3′-dideoxy-3′-thiacytidine (3TC), a 2′,3′-didehydro-2′,3′-dideoxythymidine (d4T), a monophosphate nucleotide of 3′-azido-3′-deoxythymidine (AZT), a 2′,3′-dideoxy-3′-thiacytidine (3TC) and a monophosphate nucleotide of 2′,3′-didehydro-2′,3′-dideoxythymidine (d4T), a 4-thiouracil, a 5-bromouracil, a 5-iodouracil, a 5-(3-aminoallyl)-uracil, a 2′-O-alkyl ribonucleotide, a 2′-O-methyl ribonucleotide, a 2′-amino ribonucleotide, a 2′-fluoro ribonucleotide, and a locked nucleic acid.
27. The method of claim 25, wherein said double stranded nucleic acid comprises a phosphate backbone modification selected from the group consisting of a phosphonate, a phosphorothioate and a phosphotriester.
28. The method of claim 25, wherein said double stranded nucleic acid reduces target gene expression when introduced into a mammalian cell in vitro by an amount (expressed by %) selected from the group consisting of at least 10%, at least 50% and at least 80%.
US12/549,680 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna Abandoned US20100004436A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/549,680 US20100004436A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US55348704P 2004-03-15 2004-03-15
US11/079,476 US20050277610A1 (en) 2004-03-15 2005-03-15 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US12/137,914 US20090018321A1 (en) 2004-03-15 2008-06-12 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,680 US20100004436A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/137,914 Division US20090018321A1 (en) 2004-03-15 2008-06-12 Methods and compositions for the specific inhibition of gene expression by double-stranded rna

Publications (1)

Publication Number Publication Date
US20100004436A1 true US20100004436A1 (en) 2010-01-07

Family

ID=34994243

Family Applications (26)

Application Number Title Priority Date Filing Date
US11/079,476 Abandoned US20050277610A1 (en) 2004-03-15 2005-03-15 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US11/079,906 Active 2027-07-17 US8084599B2 (en) 2004-03-15 2005-03-15 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US12/137,914 Abandoned US20090018321A1 (en) 2004-03-15 2008-06-12 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/138,215 Abandoned US20090043083A1 (en) 2004-03-15 2008-06-12 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,013 Abandoned US20090326046A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/548,948 Abandoned US20100069465A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/548,965 Abandoned US20100004317A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,058 Abandoned US20090325285A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/548,986 Abandoned US20100004434A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,099 Abandoned US20090325181A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,707 Abandoned US20090325286A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,680 Abandoned US20100004436A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,641 Abandoned US20100004318A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,730 Abandoned US20100003758A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,666 Abandoned US20100004435A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/855,223 Abandoned US20100324121A1 (en) 2004-03-15 2010-08-12 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/955,241 Abandoned US20110065908A1 (en) 2004-03-15 2010-11-29 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US13/152,686 Active US8658356B2 (en) 2004-03-15 2011-06-03 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US13/177,051 Active US8796444B2 (en) 2004-03-15 2011-07-06 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US13/178,806 Active US8809515B2 (en) 2004-03-15 2011-07-08 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US13/178,844 Abandoned US20110301224A1 (en) 2004-03-15 2011-07-08 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US13/749,110 Active US8691786B2 (en) 2004-03-15 2013-01-24 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US14/019,911 Active US9365849B2 (en) 2004-03-15 2013-09-06 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US14/463,427 Active US9518262B2 (en) 2004-03-15 2014-08-19 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US15/181,162 Active US9988630B2 (en) 2004-03-15 2016-06-13 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US15/375,730 Active US10106792B2 (en) 2004-03-15 2016-12-12 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA

Family Applications Before (11)

Application Number Title Priority Date Filing Date
US11/079,476 Abandoned US20050277610A1 (en) 2004-03-15 2005-03-15 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US11/079,906 Active 2027-07-17 US8084599B2 (en) 2004-03-15 2005-03-15 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US12/137,914 Abandoned US20090018321A1 (en) 2004-03-15 2008-06-12 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/138,215 Abandoned US20090043083A1 (en) 2004-03-15 2008-06-12 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,013 Abandoned US20090326046A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/548,948 Abandoned US20100069465A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/548,965 Abandoned US20100004317A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,058 Abandoned US20090325285A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/548,986 Abandoned US20100004434A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,099 Abandoned US20090325181A1 (en) 2004-03-15 2009-08-27 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,707 Abandoned US20090325286A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna

Family Applications After (14)

Application Number Title Priority Date Filing Date
US12/549,641 Abandoned US20100004318A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,730 Abandoned US20100003758A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/549,666 Abandoned US20100004435A1 (en) 2004-03-15 2009-08-28 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/855,223 Abandoned US20100324121A1 (en) 2004-03-15 2010-08-12 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US12/955,241 Abandoned US20110065908A1 (en) 2004-03-15 2010-11-29 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US13/152,686 Active US8658356B2 (en) 2004-03-15 2011-06-03 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US13/177,051 Active US8796444B2 (en) 2004-03-15 2011-07-06 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US13/178,806 Active US8809515B2 (en) 2004-03-15 2011-07-08 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US13/178,844 Abandoned US20110301224A1 (en) 2004-03-15 2011-07-08 Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US13/749,110 Active US8691786B2 (en) 2004-03-15 2013-01-24 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US14/019,911 Active US9365849B2 (en) 2004-03-15 2013-09-06 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US14/463,427 Active US9518262B2 (en) 2004-03-15 2014-08-19 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US15/181,162 Active US9988630B2 (en) 2004-03-15 2016-06-13 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US15/375,730 Active US10106792B2 (en) 2004-03-15 2016-12-12 Methods and compositions for the specific inhibition of gene expression by double-stranded RNA

Country Status (6)

Country Link
US (26) US20050277610A1 (en)
EP (2) EP2514758B2 (en)
JP (1) JP5243789B2 (en)
AU (2) AU2005222965B8 (en)
CA (1) CA2559955C (en)
WO (1) WO2005089287A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050244858A1 (en) * 2004-03-15 2005-11-03 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
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

Families Citing this family (175)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1470148B1 (en) 2002-02-01 2012-07-18 Life Technologies Corporation Double-stranded oligonucleotides
US20030166282A1 (en) 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
US7892793B2 (en) * 2002-11-04 2011-02-22 University Of Massachusetts Allele-specific RNA interference
US9879266B2 (en) 2002-11-14 2018-01-30 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
US9228186B2 (en) 2002-11-14 2016-01-05 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
AU2004252442B2 (en) * 2003-06-02 2010-04-08 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of FNAi
ES2712695T3 (en) 2003-06-02 2019-05-14 Univ Massachusetts Methods and compositions to control the efficiency 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
US8680063B2 (en) * 2003-09-12 2014-03-25 University Of Massachusetts RNA interference for the treatment of gain-of-function disorders
EP2821085B1 (en) 2003-09-12 2020-04-29 University of Massachusetts Rna interference for the treatment of gain-of-function disorders
US20060134787A1 (en) 2004-12-22 2006-06-22 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of single and double blunt-ended siRNA
WO2006085987A2 (en) * 2004-07-09 2006-08-17 University Of Iowa Research Foundation Rna interference in respiratory epitheial cells
US20060142228A1 (en) * 2004-12-23 2006-06-29 Ambion, Inc. Methods and compositions concerning siRNA's as mediators of RNA interference
GB2421948A (en) * 2004-12-30 2006-07-12 Ist Superiore Sanita Retrotransposon inhibition to treat cancer
US20070213293A1 (en) * 2005-04-08 2007-09-13 Nastech Pharmaceutical Company Inc. Rnai therapeutic for respiratory virus infection
US8252756B2 (en) 2005-06-14 2012-08-28 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
CA2619534A1 (en) 2005-08-18 2007-02-22 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating neurological disease
WO2007139935A1 (en) * 2006-05-24 2007-12-06 Genolution Pharmaceuticals, Inc. Innate immune response inducer
US8598333B2 (en) * 2006-05-26 2013-12-03 Alnylam Pharmaceuticals, Inc. SiRNA silencing of genes expressed in cancer
HUE037173T2 (en) 2006-08-08 2018-08-28 Univ Bonn Rheinische Friedrich Wilhelms Structure and use of 5' phosphate oligonucleotides
WO2008049078A1 (en) * 2006-10-18 2008-04-24 Nastech Pharmaceutical Company Inc. Nicked or gapped nucleic acid molecules and uses thereof
CA2691066C (en) 2007-02-09 2018-07-31 Northwestern University Particles for detecting intracellular targets
JP5311854B2 (en) 2007-03-23 2013-10-09 キヤノン株式会社 Electrophotographic image forming apparatus, developing device, and coupling member
WO2008143774A2 (en) * 2007-05-01 2008-11-27 University Of Massachusetts Methods and compositions for locating snp heterozygosity for allele specific diagnosis and therapy
KR101750640B1 (en) * 2007-05-22 2017-06-23 아크투루스 쎄라퓨틱스, 인크. Oligomer for therapeutics
AU2013273719B2 (en) * 2007-05-22 2016-04-14 Arcturus Therapeutics, Inc. UNA amidites for therapeutic oligomers
EP2826863B1 (en) 2007-05-30 2017-08-23 Northwestern University Nucleic acid functionalized nanoparticles for therapeutic applications
AR071146A1 (en) 2007-08-03 2010-06-02 Alcon Res Ltd INHIBITION RELATED TO ARNI OF THE TNF ALFA SIGNAL ROUTE FOR THE TREATMENT OF EYE ANGIOGENESIS, USE OF AN INTERFERENCE RNA MOLECULA AND INTERFERENCE RNA MOLECULA
EP2193140B1 (en) * 2007-08-27 2016-11-02 1Globe Health Institute LLC Compositions of asymmetric interfering rna 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
TW200927177A (en) * 2007-10-24 2009-07-01 Nat Inst Of Advanced Ind Scien Lipid-modified double-stranded RNA having potent RNA interference effect
US8030290B2 (en) * 2007-12-07 2011-10-04 City Of Hope Cell-type specific aptamer-siRNA delivery system for HIV-1 Therapy
EP2297323A1 (en) 2008-05-21 2011-03-23 Hartmann, Gunther 5' triphosphate oligonucleotide with blunt end and uses thereof
EP2315831A2 (en) * 2008-08-05 2011-05-04 Marina Biotech, Inc. Nucleic acid compounds for inhibiting plk1 gene expression and uses thereof
US9433684B2 (en) 2008-08-19 2016-09-06 Nektar Therapeutics Conjugates of small-interfering nucleic acids
JP2012504389A (en) * 2008-09-22 2012-02-23 ダイセルナ ファーマシューティカルズ, インコーポレイテッド Compositions and methods for specific inhibition of gene expression by dsRNA with modifications
CA2744207C (en) 2008-11-24 2019-05-28 Northwestern University Polyvalent rna-nanoparticle compositions
CA2744093A1 (en) 2008-12-03 2010-06-10 Marina Biotech, Inc. Una oligomer structures for therapeutic agents
CA3151965A1 (en) * 2008-12-18 2010-07-15 Dicerna Pharmaceuticals, Inc. Extended dicer substrate agents and methods for the specific inhibition of gene expression
US11414664B2 (en) 2008-12-18 2022-08-16 Dicerna Pharmaceuticals, Inc. Extended dicer substrate agents and methods for the specific inhibition of gene expression
US20100233270A1 (en) 2009-01-08 2010-09-16 Northwestern University Delivery of Oligonucleotide-Functionalized Nanoparticles
WO2010093788A2 (en) 2009-02-11 2010-08-19 Dicerna Pharmaceuticals, Inc. Multiplex dicer substrate rna interference molecules having joining sequences
CN103555722B (en) 2009-04-03 2016-08-31 戴瑟纳制药公司 Asymmetric double-stranded RNA is utilized specifically to suppress the method and composition of KRAS
EP2756845B1 (en) 2009-04-03 2017-03-15 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of KRAS by asymmetric double-stranded RNA
CN102753187A (en) * 2009-06-03 2012-10-24 戴瑟纳制药公司 Peptide dicer substrate agents and methods for their specific inhibition of gene expression
WO2010141933A1 (en) * 2009-06-05 2010-12-09 Dicerna Pharmaceuticals, Inc. Specific inhibition of gene expression by nucleic acid containing a dicer substrate
WO2010144058A1 (en) 2009-06-10 2010-12-16 Temasek Life Sciences Laboratory Limited Virus induced gene silencing (vigs) for functional analysis of genes in cotton.
JP5766188B2 (en) 2009-07-01 2015-08-19 プロチバ バイオセラピューティクス インコーポレイティッド Lipid formulations for delivering therapeutic agents to solid tumors
WO2011011447A1 (en) 2009-07-20 2011-01-27 Protiva Biotherapeutics, Inc. Compositions and methods for silencing ebola virus gene expression
WO2011032100A1 (en) 2009-09-11 2011-03-17 Government Of The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Inhibitors of kshv vil6 and human il6
US8916693B2 (en) 2009-09-17 2014-12-23 Nektar Therapeutics Monoconjugated chitosans as delivery agents for small interfering nucleic acids
US9222086B2 (en) 2009-09-23 2015-12-29 Protiva Biotherapeutics, Inc. Compositions and methods for silencing genes expressed in cancer
CA2779099C (en) 2009-10-30 2021-08-10 Northwestern University Templated nanoconjugates
TWI543763B (en) 2009-12-09 2016-08-01 日東電工股份有限公司 Modulation of hsp47 expression
JP5257796B2 (en) * 2009-12-28 2013-08-07 株式会社村田製作所 Solid electrolytic capacitor element and manufacturing method thereof
WO2011109294A1 (en) * 2010-03-01 2011-09-09 Dicerna Pharmaceuticals, Inc. Lipid delivery formulations
EP2554669B1 (en) 2010-03-26 2018-09-19 Kyowa Hakko Kirin Co., Ltd. Novel antibody having modification site introduced therein, and antibody fragment
WO2011133878A2 (en) * 2010-04-22 2011-10-27 University Of Utah Research Foundation Compositions and methods for selectively producing sirna
JP5902616B2 (en) 2010-04-28 2016-04-13 協和発酵キリン株式会社 Cationic lipid
JP5902617B2 (en) 2010-04-28 2016-04-13 協和発酵キリン株式会社 Cationic lipid
DK3587579T3 (en) 2010-07-06 2021-04-26 Dicerna Pharmaceuticals Inc METHODS AND COMPOSITIONS FOR SPECIFIC INHIBITION OF BETA-CATENIN USING DOUBLE-STRANDED RNA
WO2012006241A2 (en) 2010-07-06 2012-01-12 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of androgen receptor by double-stranded rna
WO2012051491A1 (en) 2010-10-14 2012-04-19 The United States Of America, As Represented By The Secretary National Institutes Of Health Compositions and methods for controlling neurotropic viral pathogenesis by micro-rna targeting
JP6000287B2 (en) 2011-03-03 2016-09-28 クォーク ファーマシューティカルズ インコーポレーティッドQuark Pharmaceuticals,Inc. Compositions and methods for treating lung disease and injury
EP2508530A1 (en) 2011-03-28 2012-10-10 Rheinische Friedrich-Wilhelms-Universität Bonn Purification of triphosphorylated oligonucleotides using capture tags
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
MY166780A (en) 2011-06-10 2018-07-23 Temasek Life Sciences Laboratory Ltd Genetic manipulation and expression system for pucciniomycotina and ustilaginomycotina subphyla
EP2755692B1 (en) 2011-09-14 2020-11-25 Northwestern University Nanoconjugates able to cross the blood-brain barrier
EP3597644B1 (en) 2011-10-18 2021-09-29 Dicerna Pharmaceuticals, Inc. Amine cationic lipids and uses thereof
BR112014010013A2 (en) * 2011-10-27 2017-04-25 Occlutech Holding Ag medical implant, kit and method of manufacturing a 3d filament tissue for forming a medical implant
JP2013095755A (en) 2011-11-02 2013-05-20 Kyowa Hakko Kirin Co Ltd Cationic lipid
JP6182458B2 (en) 2011-12-12 2017-08-16 協和発酵キリン株式会社 Lipid nanoparticles containing a combination of cationic lipids
EP2792367A4 (en) 2011-12-12 2015-09-30 Kyowa Hakko Kirin Co Ltd Lipid nanoparticles for drug delivery system containing cationic lipids
US9035039B2 (en) 2011-12-22 2015-05-19 Protiva Biotherapeutics, Inc. Compositions and methods for silencing SMAD4
DK2817287T3 (en) 2012-02-24 2019-01-02 Arbutus Biopharma Corp TRIALKYL CATIONIC LIPID AND METHODS FOR USING IT
WO2013158032A1 (en) 2012-04-19 2013-10-24 Temasek Life Sciences Laboratory Limited Methods for increasing cotton fiber length
US9272016B2 (en) 2012-04-20 2016-03-01 University Of Iowa Research Foundation Methods to enhance RNAi oligonucleotide delivery to respiratory epithelial cells
TW201726600A (en) 2012-07-06 2017-08-01 協和醱酵麒麟有限公司 Cationic lipid
EP3878962A1 (en) * 2012-07-17 2021-09-15 Stelic Institute & Co. Mucosal healing promoter
WO2014018375A1 (en) 2012-07-23 2014-01-30 Xenon Pharmaceuticals Inc. Cyp8b1 and uses thereof in therapeutic and diagnostic methods
US10378011B2 (en) 2012-08-31 2019-08-13 Kyowa Hakko Kirin Co., Ltd. Oligonucleotide
AU2013318338B2 (en) 2012-09-21 2017-05-25 Intensity Therapeutics, Inc Method of treating cancer
EP2712870A1 (en) 2012-09-27 2014-04-02 Rheinische Friedrich-Wilhelms-Universität Bonn Novel RIG-I ligands and methods for producing them
WO2014055825A1 (en) 2012-10-04 2014-04-10 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services A formulation of mycobacterial components as an adjuvant for inducing th17 responses
WO2014093746A2 (en) 2012-12-14 2014-06-19 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of ckap5 by double-stranded rna
WO2014113541A1 (en) 2013-01-16 2014-07-24 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Attenuated chlamydia vaccine
WO2014134255A2 (en) 2013-02-28 2014-09-04 Novartis Ag Organic compositions to treat epas1-related diseases
CN105142614A (en) 2013-03-14 2015-12-09 迪克纳制药公司 Process for formulating an anionic agent
US9546364B2 (en) 2013-03-15 2017-01-17 Carnegie Mellon University Synthetic lariat RNA for RNA interference
EP3444350B1 (en) 2013-07-03 2021-12-01 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of alpha-1 antitrypsin by double-stranded rna
JP6064251B2 (en) * 2013-07-19 2017-01-25 ホアウェイ・テクノロジーズ・カンパニー・リミテッド Signaling and transport of metadata information in dynamic adaptive hypertext transfer protocol streaming
SG11201600379TA (en) 2013-08-07 2016-02-26 Arrowhead Res Corp Polyconjugates for delivery of rnai triggers to tumor cells in vivo
EP3077511A4 (en) 2013-12-06 2017-07-05 Dicerna Pharmaceuticals Inc. Methods and compositions for the specific inhibition of transthyretin (ttr) by double-stranded rna
AU2014369850B2 (en) 2013-12-27 2021-04-08 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of Glycolate Oxidase (HAO1) by double-stranded RNA
CN105980560A (en) 2014-01-17 2016-09-28 协和发酵麒麟株式会社 Nucleic acid capable of inhibiting expression of [beta]2GPI
CN110846316B (en) 2014-03-25 2023-10-31 阿克丘勒斯治疗公司 UNA oligomers with reduced off-target effects in gene silencing
US9856475B2 (en) 2014-03-25 2018-01-02 Arcturus Therapeutics, Inc. Formulations for treating amyloidosis
WO2015148582A1 (en) 2014-03-25 2015-10-01 Arcturus Therapeutics, Inc. Transthyretin allele selective una oligomers for gene silencing
US9987299B2 (en) 2014-05-05 2018-06-05 University Of Iowa Research Foundation Methods of improving RNAi in well-differentiated airway epithelia
WO2015171827A1 (en) 2014-05-06 2015-11-12 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Triggering rna interference with rna-dna and dna-rna nanoparticles
CN106536056B (en) 2014-06-13 2021-07-16 儿童医学中心公司 Products and methods for isolating mitochondria
TW201620526A (en) 2014-06-17 2016-06-16 愛羅海德研究公司 Compositions and methods for inhibiting gene expression of alpha-1 antitrypsin
KR101670254B1 (en) * 2014-07-09 2016-10-28 서울대학교산학협력단 siRNA molecule having increased specificity for suppression of target gene expression
JP2016071140A (en) 2014-09-30 2016-05-09 富士フイルム株式会社 Zoom lens and image capturing device
JOP20200115A1 (en) * 2014-10-10 2017-06-16 Alnylam Pharmaceuticals Inc Compositions And Methods For Inhibition Of HAO1 (Hydroxyacid Oxidase 1 (Glycolate Oxidase)) Gene Expression
CN107257858B (en) 2014-10-10 2022-03-08 戴瑟纳制药公司 Therapeutic inhibition of lactate dehydrogenase and reagents therefor
SG11201703132UA (en) 2014-10-22 2017-05-30 Temasek Life Sciences Lab Ltd Terpene synthases from ylang ylang (cananga odorata var. fruticosa)
CN107106493A (en) 2014-11-21 2017-08-29 西北大学 The sequence-specific cellular uptake of spherical nucleic acid nano particle conjugate
JP7105065B2 (en) 2014-12-15 2022-07-22 ダイセルナ ファーマシューティカルズ, インコーポレイテッド Ligand-modified double-stranded nucleic acid
US10264976B2 (en) 2014-12-26 2019-04-23 The University Of Akron Biocompatible flavonoid compounds for organelle and cell imaging
US10036017B2 (en) 2015-02-17 2018-07-31 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of complement component 5(C5) by double-stranded RNA
WO2016149313A1 (en) 2015-03-17 2016-09-22 Arrowhead Research Corporation Improved disulfide-containing alkyne linking agents
JP2018509913A (en) 2015-03-17 2018-04-12 アローヘッド ファーマシューティカルズ インコーポレイテッド Compositions and methods for inhibiting factor XII gene expression
US10519447B2 (en) 2015-04-01 2019-12-31 Arcturus Therapeutics, Inc. Therapeutic UNA oligomers and uses thereof
WO2016183009A2 (en) 2015-05-08 2016-11-17 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of antithrombin 3 (at3) by double-stranded rna
MX2017014641A (en) 2015-05-29 2018-01-23 Arrowhead Pharmaceuticals Inc Compositions and methods for inhibiting gene expression of hif2alpha.
JP6966427B2 (en) 2015-05-29 2021-11-17 アローヘッド ファーマシューティカルズ インコーポレイテッド Biologically cleavable tetrapeptide linking agent
WO2017015671A1 (en) 2015-07-23 2017-01-26 Arcturus Therapeutics, Inc. Compositions for treating amyloidosis
KR20180038465A (en) 2015-08-07 2018-04-16 애로우헤드 파마슈티컬스 인코포레이티드 RNAi therapy for hepatitis B virus infection
JOP20210043A1 (en) 2015-10-01 2017-06-16 Arrowhead Pharmaceuticals Inc Compositions and Methods for Inhibiting Gene Expression of LPA
WO2017095946A1 (en) 2015-11-30 2017-06-08 Flagship Pioneering, Inc. Methods and compositions relating to chondrisomes
BR112018008344A2 (en) 2015-12-13 2018-10-30 Nitto Denko Corp nucleic acid molecule, pharmaceutical composition, vector or cell, method for preventing, treating or ameliorating a disease, and use of a composition
JP6774965B2 (en) 2015-12-25 2020-10-28 協和キリン株式会社 Compounds as cationic lipids
CN105462932A (en) * 2015-12-29 2016-04-06 同济大学苏州研究院 Tumor metastasis resistance medicine screening cell model constructed by adopting lentivirus technology and application method thereof
BR112018067914A2 (en) 2016-02-25 2019-01-29 Applied Biological Laboratories Inc compositions and methods for protection against airborne pathogens and irritants
US20170360815A1 (en) 2016-02-25 2017-12-21 Applied Biological Laboratories, Inc. Compositions and methods for protecting against airborne pathogens and irritants
MA45328A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc NUCLEIC ACID-POLYPEPTIDE COMPOSITIONS AND USES THEREOF
MA45349A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc EGFR NUCLEIC ACIDS AND THEIR USES
MA45469A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc BETA-CATENIN NUCLEIC ACIDS AND THEIR USES
MA45470A (en) 2016-04-01 2019-02-06 Avidity Biosciences Llc KRAS NUCLEIC ACIDS AND THEIR USES
CN107333099B (en) * 2016-04-28 2019-11-19 瑞昱半导体股份有限公司 Network camera with wireless relay function
US11091795B2 (en) 2016-07-11 2021-08-17 Arizona Board Of Regents On Behalf Of The University Of Arizona Compositions and methods for diagnosing and treating arrhythmias
JOP20170161A1 (en) 2016-08-04 2019-01-30 Arrowhead Pharmaceuticals Inc RNAi Agents for Hepatitis B Virus Infection
BR112019014282A2 (en) 2017-01-10 2020-03-03 Arrowhead Pharmaceuticals, Inc. ANTITHRIPSIN RNAI AGENTS (AAT) ALPHA-1, COMPOSITIONS INCLUDING AAT RNAI AGENTS, AND METHODS OF USE
US11241436B2 (en) 2017-01-25 2022-02-08 Northwestern University Autophagy inducers for treatment of CNS conditions
JPWO2018164186A1 (en) 2017-03-09 2020-01-09 協和キリン株式会社 Nucleic acids that suppress MASP2 expression
WO2018187373A1 (en) 2017-04-03 2018-10-11 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Functionally-interdependent shape switching nucleic acid nanoparticles
AU2018259859B2 (en) * 2017-04-28 2024-02-15 Kyowa Kirin Co., Ltd. Oligonucleotide derivative or salt thereof
WO2018209270A1 (en) 2017-05-11 2018-11-15 Northwestern University Adoptive cell therapy using spherical nucleic acids (snas)
CA3065149A1 (en) 2017-05-31 2018-12-06 Kyowa Kirin Co., Ltd. Nucleic acid suppressing expression of apcs
CA3066750A1 (en) 2017-06-13 2018-12-20 Flagship Pioneering Innovations V, Inc. Compositions comprising curons and uses thereof
AU2018301829A1 (en) 2017-07-13 2020-02-27 Alnylam Pharmaceuticals, Inc. Methods for inhibition of HAO1 (hydroxyacid oxidase 1 (glycolate oxidase) gene expression
US11633365B2 (en) 2017-08-04 2023-04-25 Kyowa Kirin Co., Ltd. Nucleic acid-containing lipid nanoparticle
EP3679141B1 (en) 2017-10-13 2023-06-07 Novo Nordisk Health Care AG Methods and compositions for inhibiting expression of ldha
US10815484B2 (en) 2017-11-22 2020-10-27 The Regents Of The University Of Michigan Compositions and methods for treating cancer
KR102443358B1 (en) 2017-12-06 2022-09-14 어비디티 바이오사이언시스 인크. Compositions and methods for treating muscular dystrophy and myotonic dystrophy
US20200306286A1 (en) 2017-12-15 2020-10-01 Flagship Pioneering Innovations Vi, Llc Compositions comprising circular polyribonucleotides and uses thereof
JP2022512077A (en) 2018-11-16 2022-02-02 日東電工株式会社 RNA interference delivery products and methods for malignant tumors
AU2019390097A1 (en) 2018-11-30 2021-07-15 Kyowa Kirin Co., Ltd. Nucleic acid conjugate
MX2021011928A (en) 2019-03-29 2022-01-04 Dicerna Pharmaceuticals Inc Compositions and methods for the treatment of kras associated diseases or disorders.
BR112022003861A2 (en) 2019-08-30 2022-05-24 Dicerna Pharmaceuticals Inc Ligand-modified nucleic acids, synthesis thereof, and intermediates therefor
CN114391040A (en) 2019-09-23 2022-04-22 欧米茄治疗公司 Compositions and methods for modulating apolipoprotein B (APOB) gene expression
JP2022548399A (en) 2019-09-23 2022-11-18 オメガ セラピューティクス, インコーポレイテッド Compositions and methods for modulating hepatocyte nuclear factor 4-alpha (HNF4α) gene expression
US20230056569A1 (en) 2019-11-22 2023-02-23 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
TW202142689A (en) 2020-01-29 2021-11-16 美商旗艦先鋒創新有限責任公司 Compositions for translation and methods of use thereof
AU2021234302A1 (en) 2020-03-11 2022-11-10 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
KR20220156867A (en) 2020-03-19 2022-11-28 어비디티 바이오사이언시스 인크. Compositions and methods for the treatment of facial scapular brachial muscular dystrophy
EP4100028A4 (en) 2020-08-20 2023-07-26 A2 Biotherapeutics, Inc. Compositions and methods for treating mesothelin positive cancers
EP4058474A1 (en) 2020-08-20 2022-09-21 A2 Biotherapeutics, Inc. Compositions and methods for treating egfr positive cancers
AU2021329375A1 (en) 2020-08-20 2023-04-20 A2 Biotherapeutics, Inc. Compositions and methods for treating ceacam positive cancers
EP4267157A1 (en) 2020-12-23 2023-11-01 Flagship Pioneering Innovations V, Inc. In vitro assembly of anellovirus capsids enclosing rna
KR20230147109A (en) 2021-02-16 2023-10-20 에이투 바이오쎄라퓨틱스, 인크. Compositions and methods for treating HER2 positive cancer
WO2022215758A1 (en) 2021-04-09 2022-10-13 相互薬工株式会社 Lipid and composition
WO2022263931A2 (en) 2021-06-17 2022-12-22 Oslo Universitetssykehus Hf Compositions and methods for treating cancer
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
US20230107967A1 (en) 2021-08-25 2023-04-06 Dicerna Pharmaceuticals, Inc. Compositions and methods for inhibiting alpha-1 antitrypsin expression
WO2023069498A1 (en) 2021-10-22 2023-04-27 Senda Biosciences, Inc. Mrna vaccine composition
US20230159930A1 (en) 2021-11-19 2023-05-25 Sanegene Bio Usa Inc. Double stranded rna targeting angiopoietin-like 3 (angptl-3) and methods of use thereof
WO2023096858A1 (en) 2021-11-23 2023-06-01 Senda Biosciences, Inc. A bacteria-derived lipid composition and use thereof
WO2023122080A1 (en) 2021-12-20 2023-06-29 Senda Biosciences, Inc. Compositions comprising mrna and lipid reconstructed plant messenger packs
WO2023220561A1 (en) 2022-05-09 2023-11-16 Sanegene Bio Usa Inc. Double stranded rna targeting 17-beta hydroxysteroiddehydrogenase 13 (hsd17b13) and methods of use thereof
WO2023220729A2 (en) 2022-05-13 2023-11-16 Flagship Pioneering Innovations Vii, Llc Double stranded dna compositions and related methods
US20240052348A1 (en) 2022-08-05 2024-02-15 Sanegene Bio Usa Inc. Double stranded rna targeting angiotensinogen (agt) and methods of use thereof

Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5624803A (en) * 1993-10-14 1997-04-29 The Regents Of The University Of California In vivo oligonucleotide generator, and methods of testing the binding affinity of triplex forming oligonucleotides derived therefrom
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US20020086356A1 (en) * 2000-03-30 2002-07-04 Whitehead Institute For Biomedical Research RNA sequence-specific mediators of RNA interference
US20020114784A1 (en) * 1999-01-28 2002-08-22 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded RNA
US20040002077A1 (en) * 2001-11-28 2004-01-01 Center For Advanced Science And Technology Incubation, Ltd. siRNA expression system and method for producing functional gene knock-down cell using the system
US20040014956A1 (en) * 2002-02-01 2004-01-22 Sequitur, Inc. Double-stranded oligonucleotides
US20040023390A1 (en) * 2002-08-05 2004-02-05 Davidson Beverly L. SiRNA-mediated gene silencing with viral vectors
US20040147476A1 (en) * 1999-04-21 2004-07-29 Wyeth Methods and compositions for inhibiting the function of polynucleotide sequences
US20040152117A1 (en) * 2001-01-31 2004-08-05 Tony Giordano Use of post-transcriptional gene silencing for identifying nucleic acid sequences that modulate the function of a cell
US20040175703A1 (en) * 1999-11-24 2004-09-09 Ribopharma Ag Compositions and methods for inhibiting expression of a target gene
US20040180438A1 (en) * 2002-04-26 2004-09-16 Pachuk Catherine J. Methods and compositions for silencing genes without inducing toxicity
US20040192626A1 (en) * 2002-02-20 2004-09-30 Mcswiggen James RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US20040203145A1 (en) * 2002-08-07 2004-10-14 University Of Massachusetts Compositions for RNA interference and methods of use thereof
US20040224405A1 (en) * 2003-05-06 2004-11-11 Dharmacon Inc. siRNA induced systemic gene silencing in mammalian systems
US20040248299A1 (en) * 2002-12-27 2004-12-09 Sumedha Jayasena RNA interference
US20040259247A1 (en) * 2000-12-01 2004-12-23 Thomas Tuschl Rna interference mediating small rna molecules
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)
US20050032733A1 (en) * 2001-05-18 2005-02-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SiNA)
US20050039224A1 (en) * 2001-10-22 2005-02-17 Pachuk Catherine J. Transfection kinetics and structural promoters
US20050042646A1 (en) * 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
US20050186586A1 (en) * 2003-06-02 2005-08-25 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNAi
US20050233329A1 (en) * 2002-02-20 2005-10-20 Sirna Therapeutics, Inc. Inhibition of gene expression using duplex forming oligonucleotides
US20050239728A1 (en) * 2002-07-31 2005-10-27 Pachuk Catherine J Double stranded rna structures and constructs, and methods for generating and using the same
US20050244858A1 (en) * 2004-03-15 2005-11-03 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
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
US20060009402A1 (en) * 2001-07-12 2006-01-12 Zamore Phillip D In vivo production of small interfering rnas that mediate gene silencing
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
US20080146788A1 (en) * 2004-06-03 2008-06-19 Isis Pharmaceuticals, Inc. Positionally Modified Sirna Constructs

Family Cites Families (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5208149A (en) * 1983-10-20 1993-05-04 The Research Foundation Of State University Of New York Nucleic acid constructs containing stable stem and loop structures
EP0331939B1 (en) * 1988-02-16 2001-11-14 Greatbatch Gen-Aid, Ltd Modified cells having resistance to retroviral infection
US20030206887A1 (en) * 1992-05-14 2003-11-06 David Morrissey RNA interference mediated inhibition of hepatitis B virus (HBV) using short interfering nucleic acid (siNA)
CA2156288C (en) 1993-02-19 2005-10-18 Junichi Yano Glycerol derivative, device and pharmaceutical composition
US6291438B1 (en) 1993-02-24 2001-09-18 Jui H. Wang Antiviral anticancer poly-substituted phenyl derivatized oligoribonucleotides and methods for their use
US5858988A (en) 1993-02-24 1999-01-12 Wang; Jui H. Poly-substituted-phenyl-oligoribo nucleotides having enhanced stability and membrane permeability and methods of use
US5902880A (en) * 1994-08-19 1999-05-11 Ribozyme Pharmaceuticals, Inc. RNA polymerase III-based expression of therapeutic RNAs
US6146886A (en) * 1994-08-19 2000-11-14 Ribozyme Pharmaceuticals, Inc. RNA polymerase III-based expression of therapeutic RNAs
US5994316A (en) 1996-02-21 1999-11-30 The Immune Response Corporation Method of preparing polynucleotide-carrier complexes for delivery to cells
US5684143A (en) 1996-02-21 1997-11-04 Lynx Therapeutics, Inc. Oligo-2'-fluoronucleotide N3'->P5' phosphoramidates
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
AU783681B2 (en) 1999-07-09 2005-11-24 Wyeth Methods and compositions for preventing the formation of aberrant RNA during transcription of a plasmid sequence
DE10160151A1 (en) * 2001-01-09 2003-06-26 Ribopharma Ag Inhibiting expression of target gene, useful e.g. for inhibiting oncogenes, by administering double-stranded RNA complementary to the target and having an overhang
US20070026394A1 (en) * 2000-02-11 2007-02-01 Lawrence Blatt Modulation of gene expression associated with inflammation proliferation and neurite outgrowth using nucleic acid based technologies
US20020114764A1 (en) 2001-02-20 2002-08-22 Berryman Michelle S. Tattoo method and system for medical and surgical applications
CA2372450A1 (en) * 2001-05-10 2001-09-19 Pharmaceutical Partners Of Canada Inc. Liquid injectable formulation of disodium pamidronate
US20050054598A1 (en) 2002-02-20 2005-03-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition hairless (HR) gene expression using short interfering nucleic acid (siNA)
CA2526831C (en) 2001-05-18 2012-07-31 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (sina)
US10590418B2 (en) 2001-07-23 2020-03-17 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for RNAi mediated inhibition of gene expression in mammals
WO2003016572A1 (en) 2001-08-17 2003-02-27 Eli Lilly And Company Oligonucleotide therapeutics for treating hepatitis c virus infections
GB0224341D0 (en) 2002-10-19 2002-11-27 Qinetiq Ltd Mobile radio base station
DE10202419A1 (en) * 2002-01-22 2003-08-07 Ribopharma Ag Method of inhibiting expression of a target gene resulting from chromosome aberration
US20030166282A1 (en) 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
WO2003079757A2 (en) * 2002-03-20 2003-10-02 Massachusetts Institute Of Technology Hiv therapeutic
US20040121353A1 (en) * 2002-05-23 2004-06-24 Ceptyr, Inc. Modulation of TCPTP signal transduction by RNA interference
GB0212302D0 (en) 2002-05-28 2002-07-10 Isis Innovation Method of selecting targets for gene silencing by RNA interference
CA2486658C (en) * 2002-05-31 2014-07-29 The Regents Of The University Of California Method for efficient rna interference in mammalian cells
US7148342B2 (en) * 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
CA2493949C (en) * 2002-07-26 2015-06-02 Chiron Corporation Modified small interfering rna molecules and methods of use
AU2003299531A1 (en) * 2002-08-05 2004-06-07 University Of Massachusetts Compounds for modulating rna interference
AU2003277005A1 (en) 2002-09-26 2004-04-19 The Regents Of The University Of California Conditional gene expression using rnai
US20060035344A1 (en) 2002-10-18 2006-02-16 Pachuk Catherine J Double-stranded rna structures and constructs, and methods for generating and using the same
EP2284266B1 (en) 2002-11-14 2013-11-06 Thermo Fisher Scientific Biosciences Inc. siRNA targeting tp53
US6960568B2 (en) * 2002-12-17 2005-11-01 Yale University Nucleosides and related processes, pharmaceutical compositions and methods
JP3813934B2 (en) 2003-02-26 2006-08-23 共同カイテック株式会社 Green body
US20060263764A1 (en) 2003-02-27 2006-11-23 Nucleonics Inc. Methods and constructs for evaluation of rnai targets and effector molecules
US20050042641A1 (en) 2003-05-27 2005-02-24 Cold Spring Harbor Laboratory In vivo high throughput selection of RNAi probes
BRPI0411219A (en) 2003-06-12 2006-07-18 Nucleonics Inc conserved hbv and hcv sequences useful for gene silencing
TW200523265A (en) 2003-07-31 2005-07-16 Basf Ag A process for the preparation of acylphosphines
WO2005019463A1 (en) 2003-08-11 2005-03-03 Therapeutic Human Polyclonals, Inc. Improved transgenesis with humanized immunoglobulin loci
EP1656453A2 (en) 2003-08-22 2006-05-17 Nucleonics, Inc. Eukaryotic expression systems for expression of inhibitory rna in multiple intracellular compartments
EP2004049A1 (en) 2003-08-26 2008-12-24 Scuola Superiore Di Studi Universitari E Di Perfezionamento S. Anna A wearable mechatronic device for the analysis of joint biomechanics
JPWO2005068630A1 (en) * 2003-12-16 2007-07-26 独立行政法人産業技術総合研究所 Double-stranded RNA for interference
JP5196900B2 (en) 2007-07-30 2013-05-15 三洋電機株式会社 Laminated battery
JP2010021493A (en) 2008-07-14 2010-01-28 Toshiba Corp Semiconductor device and method of manufacturing the same

Patent Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5624803A (en) * 1993-10-14 1997-04-29 The Regents Of The University Of California In vivo oligonucleotide generator, and methods of testing the binding affinity of triplex forming oligonucleotides derived therefrom
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US6107094A (en) * 1996-06-06 2000-08-22 Isis Pharmaceuticals, Inc. Oligoribonucleotides and ribonucleases for cleaving RNA
US20020114784A1 (en) * 1999-01-28 2002-08-22 Medical College Of Georgia Research Institute, Inc. Composition and method for in vivo and in vitro attenuation of gene expression using double stranded RNA
US20040147476A1 (en) * 1999-04-21 2004-07-29 Wyeth Methods and compositions for inhibiting the function of polynucleotide sequences
US20040175703A1 (en) * 1999-11-24 2004-09-09 Ribopharma Ag Compositions and methods for inhibiting expression of a target gene
US20020086356A1 (en) * 2000-03-30 2002-07-04 Whitehead Institute For Biomedical Research RNA sequence-specific mediators of RNA interference
US20030108923A1 (en) * 2000-03-30 2003-06-12 Whitehead Institute For Biomedical Research RNA sequence-specific mediators of RNA interference
US20040259247A1 (en) * 2000-12-01 2004-12-23 Thomas Tuschl Rna interference mediating small rna molecules
US20040152117A1 (en) * 2001-01-31 2004-08-05 Tony Giordano Use of post-transcriptional gene silencing for identifying nucleic acid sequences that modulate the function of a cell
US20050282188A1 (en) * 2001-05-18 2005-12-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20050032733A1 (en) * 2001-05-18 2005-02-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SiNA)
US20060009402A1 (en) * 2001-07-12 2006-01-12 Zamore Phillip D In vivo production of small interfering rnas that mediate gene silencing
US20050039224A1 (en) * 2001-10-22 2005-02-17 Pachuk Catherine J. Transfection kinetics and structural promoters
US20050048647A1 (en) * 2001-11-28 2005-03-03 Kazunari Taira Sirna expression system and process for producing functional gene knockdown cell or the like using the same
US20040002077A1 (en) * 2001-11-28 2004-01-01 Center For Advanced Science And Technology Incubation, Ltd. siRNA expression system and method for producing functional gene knock-down cell using the system
US20060009409A1 (en) * 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
US20040014956A1 (en) * 2002-02-01 2004-01-22 Sequitur, Inc. Double-stranded oligonucleotides
US20040192626A1 (en) * 2002-02-20 2004-09-30 Mcswiggen James 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)
US20050233329A1 (en) * 2002-02-20 2005-10-20 Sirna Therapeutics, Inc. Inhibition of gene expression using duplex forming oligonucleotides
US20040180438A1 (en) * 2002-04-26 2004-09-16 Pachuk Catherine J. Methods and compositions for silencing genes without inducing toxicity
US20050239728A1 (en) * 2002-07-31 2005-10-27 Pachuk Catherine J Double stranded rna structures and constructs, and methods for generating and using the same
US20050042646A1 (en) * 2002-08-05 2005-02-24 Davidson Beverly L. RNA interference suppresion of neurodegenerative diseases and methods of use thereof
US20040023390A1 (en) * 2002-08-05 2004-02-05 Davidson Beverly L. SiRNA-mediated gene silencing with viral vectors
US20040203145A1 (en) * 2002-08-07 2004-10-14 University Of Massachusetts Compositions for RNA interference and methods of use thereof
US20040248299A1 (en) * 2002-12-27 2004-12-09 Sumedha Jayasena RNA interference
US20040224405A1 (en) * 2003-05-06 2004-11-11 Dharmacon Inc. siRNA induced systemic gene silencing in mammalian systems
US20050186586A1 (en) * 2003-06-02 2005-08-25 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNAi
US20090326046A1 (en) * 2004-03-15 2009-12-31 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20100004435A1 (en) * 2004-03-15 2010-01-07 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded rna
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
US20100069465A1 (en) * 2004-03-15 2010-03-18 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090018321A1 (en) * 2004-03-15 2009-01-15 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20100004434A1 (en) * 2004-03-15 2010-01-07 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20100003758A1 (en) * 2004-03-15 2010-01-07 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20100004318A1 (en) * 2004-03-15 2010-01-07 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20050277610A1 (en) * 2004-03-15 2005-12-15 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US20090043083A1 (en) * 2004-03-15 2009-02-12 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20100004317A1 (en) * 2004-03-15 2010-01-07 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090325286A1 (en) * 2004-03-15 2009-12-31 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20050244858A1 (en) * 2004-03-15 2005-11-03 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US20090325285A1 (en) * 2004-03-15 2009-12-31 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090325181A1 (en) * 2004-03-15 2009-12-31 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20080146788A1 (en) * 2004-06-03 2008-06-19 Isis Pharmaceuticals, Inc. Positionally Modified Sirna Constructs
US20090043085A1 (en) * 2007-05-01 2009-02-12 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090035854A1 (en) * 2007-05-01 2009-02-05 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090036661A1 (en) * 2007-05-01 2009-02-05 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090029466A1 (en) * 2007-05-01 2009-01-29 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090029936A1 (en) * 2007-05-01 2009-01-29 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8809515B2 (en) 2004-03-15 2014-08-19 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US20090043083A1 (en) * 2004-03-15 2009-02-12 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090018321A1 (en) * 2004-03-15 2009-01-15 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20100003758A1 (en) * 2004-03-15 2010-01-07 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US10106792B2 (en) 2004-03-15 2018-10-23 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US9988630B2 (en) 2004-03-15 2018-06-05 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US9518262B2 (en) 2004-03-15 2016-12-13 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US20100004317A1 (en) * 2004-03-15 2010-01-07 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US9365849B2 (en) 2004-03-15 2016-06-14 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US20090325181A1 (en) * 2004-03-15 2009-12-31 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090325286A1 (en) * 2004-03-15 2009-12-31 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090325285A1 (en) * 2004-03-15 2009-12-31 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090326046A1 (en) * 2004-03-15 2009-12-31 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20100069465A1 (en) * 2004-03-15 2010-03-18 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20100004318A1 (en) * 2004-03-15 2010-01-07 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20100004434A1 (en) * 2004-03-15 2010-01-07 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20050244858A1 (en) * 2004-03-15 2005-11-03 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
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
US20100004435A1 (en) * 2004-03-15 2010-01-07 Integrated Dna Technologies, Inc. Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US8796444B2 (en) 2004-03-15 2014-08-05 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US8084599B2 (en) 2004-03-15 2011-12-27 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US8658356B2 (en) 2004-03-15 2014-02-25 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US8691786B2 (en) 2004-03-15 2014-04-08 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US20100240734A1 (en) * 2007-05-01 2010-09-23 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090029466A1 (en) * 2007-05-01 2009-01-29 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
US20090043085A1 (en) * 2007-05-01 2009-02-12 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
US20090036661A1 (en) * 2007-05-01 2009-02-05 City Of Hope 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
US20090035854A1 (en) * 2007-05-01 2009-02-05 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20090029936A1 (en) * 2007-05-01 2009-01-29 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

Also Published As

Publication number Publication date
US20130345291A1 (en) 2013-12-26
US9365849B2 (en) 2016-06-14
AU2010202321A1 (en) 2010-07-01
US20160340676A1 (en) 2016-11-24
JP5243789B2 (en) 2013-07-24
US20110301229A1 (en) 2011-12-08
EP1742958A2 (en) 2007-01-17
US20050277610A1 (en) 2005-12-15
US20090325285A1 (en) 2009-12-31
US20110288158A1 (en) 2011-11-24
AU2005222965B2 (en) 2010-03-04
AU2010202321B2 (en) 2013-03-28
US20140357700A1 (en) 2014-12-04
US20110065908A1 (en) 2011-03-17
US20100004434A1 (en) 2010-01-07
US9518262B2 (en) 2016-12-13
US20100003758A1 (en) 2010-01-07
CA2559955A1 (en) 2005-09-29
WO2005089287A2 (en) 2005-09-29
US8084599B2 (en) 2011-12-27
AU2005222965B8 (en) 2010-07-01
US20090326046A1 (en) 2009-12-31
US20100004318A1 (en) 2010-01-07
US8809515B2 (en) 2014-08-19
EP1742958B1 (en) 2017-05-17
AU2005222965A1 (en) 2005-09-29
US20100004435A1 (en) 2010-01-07
US20170159054A1 (en) 2017-06-08
US9988630B2 (en) 2018-06-05
US20100004317A1 (en) 2010-01-07
US8796444B2 (en) 2014-08-05
US20090018321A1 (en) 2009-01-15
WO2005089287A3 (en) 2006-11-30
US8658356B2 (en) 2014-02-25
US20050244858A1 (en) 2005-11-03
US20110257384A1 (en) 2011-10-20
US20100324121A1 (en) 2010-12-23
EP1742958A4 (en) 2009-08-05
US20090325181A1 (en) 2009-12-31
US8691786B2 (en) 2014-04-08
EP2514758B1 (en) 2017-04-19
US20100069465A1 (en) 2010-03-18
EP2514758A1 (en) 2012-10-24
US10106792B2 (en) 2018-10-23
CA2559955C (en) 2016-02-16
US20090325286A1 (en) 2009-12-31
US20090043083A1 (en) 2009-02-12
JP2007529224A (en) 2007-10-25
US20130273652A1 (en) 2013-10-17
US20110301224A1 (en) 2011-12-08
EP2514758B2 (en) 2021-06-23
WO2005089287A9 (en) 2017-03-02

Similar Documents

Publication Publication Date Title
US9988630B2 (en) Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US10233450B2 (en) Methods and compositions for the specific inhibition of gene expression by double-stranded RNA

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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