US20030191077A1 - Method and reagent for the treatment of asthma and allergic conditions - Google Patents

Method and reagent for the treatment of asthma and allergic conditions Download PDF

Info

Publication number
US20030191077A1
US20030191077A1 US10/230,006 US23000602A US2003191077A1 US 20030191077 A1 US20030191077 A1 US 20030191077A1 US 23000602 A US23000602 A US 23000602A US 2003191077 A1 US2003191077 A1 US 2003191077A1
Authority
US
United States
Prior art keywords
nucleic acid
gccguuaggc cgaa
cugaugag gccguuaggc
acid molecule
rna
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/230,006
Inventor
Kathy Fosnaugh
James McSwiggen
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.)
Sirna Therapeutics Inc
Original Assignee
Ribozyme Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40293860&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20030191077(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US09/827,395 external-priority patent/US20030113891A1/en
Application filed by Ribozyme Pharmaceuticals Inc filed Critical Ribozyme Pharmaceuticals Inc
Priority to US10/230,006 priority Critical patent/US20030191077A1/en
Assigned to RIBOZYME PHARMACEUTICALS, INC. reassignment RIBOZYME PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MCSWIGGEN, JAMES A., FOSNAUGH, KATHY
Publication of US20030191077A1 publication Critical patent/US20030191077A1/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
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/12Type of nucleic acid catalytic nucleic acids, e.g. ribozymes
    • 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
    • C12N2310/12Type of nucleic acid catalytic nucleic acids, e.g. ribozymes
    • C12N2310/121Hammerhead
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/13Decoys
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/18Type of nucleic acid acting by a non-sequence specific mechanism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/317Chemical structure of the backbone with an inverted bond, e.g. a cap structure
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/332Abasic residue
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/346Spatial arrangement of the modifications having a combination of backbone and sugar modifications

Definitions

  • the present invention relates to therapeutic compositions and methods for the treatment or diagnosis of diseases or conditions related to allergic response.
  • the invention provides compositions and methods for the treatment of diseases or conditions related to levels of factors involved in allergic conditions such as asthma, for example prostaglandin D2 receptor (PTGDR), prostaglandin D2 synthetase (PTGDS) and adenosine A1 receptor (ADORA1).
  • PAGDR prostaglandin D2 receptor
  • PAGDS prostaglandin D2 synthetase
  • ADORA1 adenosine A1 receptor
  • Asthma is a chronic inflammatory disorder of the lungs characterized by airflow obstruction, bronchial hyper-responsiveness, and airway inflammation. T-lymphocytes that produce TH2 cytokines and cosinophilic leukocytes infiltrate the airways. In the airway and in bronchial alveolar lavage (BAL) fluid of individuals with asthma, high concentrations of TH2 cytokines, interleukin-4 (IL-4), IL-5, and IL-13, are present along with increased levels of adenosine. In contrast to normal individuals, asthmatics respond to adenosine challenge with marked airway obstruction. Upon allergen challenge, mast cells are activated by cross-linked IgE-allergen complexes.
  • BAL bronchial alveolar lavage
  • PGD2 prostaglandin D2
  • PGD2 is generated from PGH2 via the activity of prostaglandin D2 synthetase (PTGDS).
  • PGD2 receptors and adenosine A1 receptors are present in the lungs and airway along with various other tissues in response to allergic stimuli (Howarth, 1997 , Allergy, 52, 12).
  • DP PGD2 receptor
  • PGD2 receptor DP is a heterotrimeric GTP-binding protein-coupled, rhodopsin-type receptor specific for PGD2 (Hirata et al., 1994 , PNAS USA., 91, 11192). These mice fail to develop airway hyperreactivity and have greatly reduced eosinophil infiltration and cytokine accumulation in response to allergens. Upon allergen challenge mice deficient in the prostaglandin D2 (PGD2) receptor (DP) did not develop airway hyperactivity.
  • PGD2 prostaglandin D2
  • Nyce International PCT Publication Nos. WO 00/62736, WO 00/09525, WO 99/13886, WO 98/23294, WO 96/40266 and U.S. Pat. No. 6,025,339 describe specific antisense oligonucleotides targeting certain mRNAs encoding particular adenosine receptors.
  • the invention features novel nucleic acid-based molecules, for example, enzymatic nucleic acid molecules, allozymes, antisense nucleic acids, 2-5A antisense chimeras, triplex forming oligonucleotides, decoy RNA, dsRNA, siRNA, aptamers, and antisense nucleic acids containing RNA cleaving chemical groups, and methods to modulate gene expression, for example, genes encoding prostaglandin D2 receptor (PTGDR), prostaglandin D2 synthetase (PTGDS), and adenosine receptors (AR) such as adenosine receptor A1, A2a, A2b, and A3.
  • PTGDR prostaglandin D2 receptor
  • PTGDS prostaglandin D2 synthetase
  • AR adenosine receptors
  • the instant invention features nucleic-acid based molecules and methods to modulate the expression of PTGDR, PTGDS, and adeno
  • the invention features one or more nucleic acid-based molecules and methods that independently or in combination modulate the expression of gene(s) encoding prostaglandin D2 receptors (PTGDR), prostaglandin D2 synthetase (PTGDS) and adenosine receptors such as ADORA1.
  • PAGDR prostaglandin D2 receptor
  • PAGDS prostaglandin D2 synthetase
  • ADORA1 receptors adenosine receptors
  • the present invention features nucleic acid molecules that modulate the expression of prostaglandin D2 receptor (PTGDR) gene, for example Genbank Accession Nos. U31332 and U31099, prostaglandin D2 synthetase (PTGDS) gene, for example Genbank Accession No. NM — 000954, and Adenosine A1 receptor (ADORA1), for example Genbank Accession No. NM — 000674.
  • PTGDR prostaglandin D2 receptor
  • PTGDS prostaglandin D2 synthetase
  • ADORA1 adenosine A1 receptor
  • PTGDR prostaglandin D2 receptor
  • PTGDR prostaglandin D2 synthetase
  • ADORA1 adenosine A1 receptor
  • the various aspects and embodiments are also directed to other genes that express prostaglandin proteins and other receptors involved in allergic reactions. Those additional genes can be analyzed for target sites using the methods described for PTGDS, PTGDR, and ADORA1. Thus, the inhibition and the effects of such inhibition of the other genes can be performed as described herein.
  • the invention features an enzymatic nucleic acid molecule comprising a sequence selected from the group consisting of SEQ ID NOs: 228-454, 831-1206, 1438-1668, 1715-2057, and 2247-2666.
  • the invention features an enzymatic nucleic acid molecule comprising at least one binding arm wherein one or more of said binding arms comprises a sequence complementary to a sequence selected from the group consisting of SEQ ID NOs: 1-227, 455-830, 1207-1437, 1669-1714, and 2058-2246.
  • the invention features an antisense nucleic acid molecule comprising a sequence complementary to a sequence selected from the group consisting of SEQ ID NOs: 1-227, 455-830, 1207-1437, 1669-1714, and 2058-2246.
  • an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing RNA cleaving chemical groups of the invention is adapted to treat asthma.
  • an enzymatic nucleic acid molecule of the invention has an endonuclease activity to cleave RNA encoded by a PTGDS and/or PTGDR gene.
  • an enzymatic nucleic acid molecule of the invention is in a hammerhead, Inozyme, Zinzyme, DNAzyme, Amberzyme, or G-cleaver configuration.
  • an enzymatic nucleic acid molecule of the invention having a hammerhead configuration comprises a sequence complementary to a sequence having SEQ ID NOs: 1-227.
  • an enzymatic nucleic acid molecule of invention having a hammerhead configuration comprises a sequence having SEQ ID NOs: 228-454.
  • an enzymatic nucleic acid molecule of the invention having an Inozyme configuration comprises a sequence complementary to a sequence having SEQ ID NOs: 455-830.
  • an enzymatic nucleic acid molecule of invention having an Inozyme configuration comprises a sequence having SEQ ID NOs: 831-1206.
  • an enzymatic nucleic acid molecule of the invention having a Zinzyme configuration comprises a sequence complementary to a sequence having SEQ ID NOs: 1207-1437.
  • an enzymatic nucleic acid molecule of invention having a Zinzyme configuration comprises a sequence having SEQ ID NOs: 1438-1668.
  • an enzymatic nucleic acid molecule of the invention having a DNAzyme configuration comprises a sequence complementary to a sequence having SEQ ID NOs: 1, 13, 55, 69, 74, 104, 112, 120, 123, 128, 131, 138, 147, 154, 157, 158, 169, 188, 192, 208, 221, 463, 475, 489, 505, 527, 541, 552, 554, 561, 563, 572, 591, 601, 605, 627, 637, 645, 652, 653, 661, 668, 669, 670, 676, 692, 699, 706, 719, 725, 732, 737, 741, 747, 763, 774, 782, 800, 805, 807, 816, 818, 823, 827, 828, 1207-1437, and 1669-1714.
  • an enzymatic nucleic acid molecule of invention having a DNAzyme configuration comprises a sequence complementary to a sequence having
  • an enzymatic nucleic acid molecule of the invention having an Amberzyme configuration comprises a sequence complementary to a sequence having SEQ ID NOs: 1207-1437, and 2058-2246.
  • an enzymatic nucleic acid molecule of invention having an Amberzyme configuration comprises a sequence having SEQ ID NOs: 2247-2666.
  • an enzymatic nucleic acid molecule of the invention comprises between 8 and 100 bases complementary to the RNA of PTGDS, ADORA1 and/or PTGDR gene. In another embodiment, an enzymatic nucleic acid molecule of the invention comprises between 14 and 24 bases complementary to a RNA molecule of a PTGDS or PTGDR gene.
  • an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing RNA cleaving chemical groups of the invention is chemically synthesized.
  • an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing RNA cleaving chemical groups of the invention comprises at least one 2′-sugar modification.
  • an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing RNA cleaving chemical groups of the invention comprises at least one nucleic acid base modification.
  • an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing RNA cleaving chemical groups of the invention comprises at least one phosphate backbone modification.
  • the invention features a mammalian cell, for example a human cell, including the enzymatic nucleic acid molecule of the invention.
  • the invention features a method of reducing PTGDS, ADORA1 and/or PTGDR expression or activity in a cell, comprising contacting the cell with an enzymatic nucleic acid molecule of the invention, under conditions suitable for the reduction.
  • the invention features a method of reducing PTGDS, ADORA1 and/or PTGDR expression or activity in a cell, comprising the step of contacting the cell with an antisense nucleic acid molecule of the invention under conditions suitable for the reduction.
  • the invention features a method of treatment of a patient having a condition associated with the level of PTGDS, ADORA1 and/or PTGDR, comprising contacting cells of the patient with an enzymatic nucleic acid molecule of the invention, under conditions suitable for the treatment.
  • the invention features a method of treatment of a patient having a condition associated with the level of PTGDS, ADORA1 and/or PTGDR, comprising contacting cells of the patient with an antisense nucleic acid molecule of the invention, under conditions suitable for the treatment.
  • a method of treatment of a patient having a condition associated with the level of PTGDS, ADORA1 and/or PTGDR is featured, wherein the method further comprises the use of one or more drug therapies under conditions suitable for the treatment.
  • the invention features a method for treatment of asthma, allergic rhinitis, or atopic dermatitis under conditions suitable for the treatment.
  • the invention features a method of cleaving a RNA molecule of PTGDS, ADORA1 and/or PTGDR gene comprising contacting an enzymatic nucleic acid molecule of the invention with a RNA molecule of a PTGDS, ADORA1 and/or PTGDR gene under conditions suitable for the cleavage, for example, wherein the cleavage is carried out in the presence of a divalent cation, such as Mg 2+ .
  • a divalent cation such as Mg 2+
  • an enzymatic nucleic acid molecule of the invention comprises a cap structure, for example a 3′,3′-linked or 5′,5′-linked deoxyabasic ribose derivative, wherein the cap structure is at the 5′-end, or 3′-end, or both the 5′-end and the 3′-end of the enzymatic nucleic acid molecule.
  • an antisense nucleic acid molecule of the invention comprises a cap structure, for example a 3′,3′-linked or 5′,5′-linked deoxyabasic ribose derivative, wherein the cap structure is at the 5′-end, or 3′-end, or both the 5′-end and the 3′-end of the antisense nucleic acid molecule.
  • the invention features an expression vector comprising a nucleic acid sequence encoding at least one enzymatic nucleic acid molecule of the invention, in a manner which allows expression of the nucleic acid molecule.
  • the invention features a mammalian cell, for example, a human cell, including an expression vector of the invention.
  • the expression vector of the invention further comprises a sequence for an antisense nucleic acid molecule complementary to a RNA molecule of a PTGDS, ADORA1 and/or PTGDR gene.
  • an expression vector of the invention comprises a nucleic acid sequence encoding two or more enzymatic nucleic acid molecules, which can be the same or different.
  • the invention features a method for treatment of asthma, allergic rhinitis, or atopic dermatitis, comprising administering to a patient an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing RNA cleaving chemical groups of the invention, under conditions suitable for the treatment, including administering to the patient one or more other therapies, for example, inhalant anti-inflammatories, bronchodilators, adenosine inhibitors and adenosine A1 receptor inhibitors.
  • therapies for example, inhalant anti-inflammatories, bronchodilators, adenosine inhibitors and adenosine A1 receptor inhibitors.
  • the method of treatment features an enzymatic nucleic acid molecule or antisense nucleic acid molecule of the invention comprises at least five ribose residues, at least ten 2′-O-methyl modifications, and a 3′-end modification, such as a 3′-3′ inverted abasic moiety.
  • an enzymatic nucleic acid molecule or antisense nucleic acid molecule of the invention further comprises phosphorothioate linkages on at least three of the 5′ terminal nucleotides.
  • the invention features a method of administering to a mammal, for example a human, an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing RNA cleaving chemical groups of the invention, comprising contacting the mammal with the nucleic acid molecule under conditions suitable for the administration, for example, in the presence of a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
  • a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome.
  • the invention features a method of administering to a mammal an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing RNA cleaving chemical groups of the invention in conjunction with a therapeutic agent, comprising contacting the mammal, for example a human, with the nucleic acid molecule and the therapeutic agent under conditions suitable for the administration.
  • the invention features the use of an enzymatic nucleic acid molecule, which can be in a hammerhead, NCH, G-cleaver, Amberzyme, Zinzyme, and/or DNAzyme motif, to down-regulate the expression of a PTGDS, an ADORA1 and/or a PTGDR gene.
  • an enzymatic nucleic acid molecule which can be in a hammerhead, NCH, G-cleaver, Amberzyme, Zinzyme, and/or DNAzyme motif, to down-regulate the expression of a PTGDS, an ADORA1 and/or a PTGDR gene.
  • inhibitor By “inhibit”, “down-regulate”, or “reduce”, it is meant that the expression of the gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or activity of one or more proteins or protein subunits, such as PTGDS, ADORA1 and/or PTGDR proteins or PTGDS, ADORA1 and/or PTGDR subunit(s), is reduced below that observed in the absence of the nucleic acid molecules of the invention.
  • inhibition, down-regulation or reduction with an enzymatic nucleic acid molecule is below that level observed in the presence of an enzymatically inactive or attenuated molecule that is able to bind to the same site on the target RNA molecule, but is unable to cleave that RNA molecule.
  • inhibition, down-regulation, or reduction with antisense oligonucleotides is below that level observed in the presence of, for example, an oligonucleotide with scrambled sequence or with mismatches.
  • inhibition, down-regulation, or reduction of PTGDS, ADORA1 and/or PTGDR with a nucleic acid molecule of the instant invention is greater in the presence of the nucleic acid molecule than in its absence.
  • up-regulate is meant that the expression of a gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins, protein subunits, or activity of one or more proteins or protein subunits, such as PTGDS, ADORA1 and/or PTGDR proteins or PTGDS, ADORA1 and/or PTGDR subunits, is greater than that observed in the absence of the nucleic acid molecules of the invention.
  • the expression of a gene such as PTGDS, ADORA1 and/or PTGDR gene, can be increased in order to treat, prevent, ameliorate, or modulate a pathological condition caused or exacerbated by an absence or low level of gene expression.
  • module is meant that the expression of the gene, or level of RNA molecules or equivalent RNA molecules encoding one or more protein subunits, or activity of one or more protein subunits is up-regulated or down-regulated, such that the expression, level, or activity is greater than or less than that observed in the absence of a nucleic acid molecule of the invention.
  • enzymatic nucleic acid molecule it is meant a nucleic acid molecule that has complementarity in a substrate binding region to a specified gene target, and also has an enzymatic activity that is active to specifically cleave target a RNA molecule. That is, the enzymatic nucleic acid molecule is able to intermolecularly cleave a RNA molecule and thereby inactivate a target RNA molecule. These complementary regions allow sufficient hybridization of an enzymatic nucleic acid molecule to a target RNA molecule and thus permit cleavage.
  • nucleic acids can be modified at the base, sugar, and/or phosphate groups.
  • enzymatic nucleic acid is used interchangeably with phrases such as ribozymes, catalytic RNA, enzymatic RNA, catalytic DNA, aptazyme or aptamer-binding ribozyme, regulatable ribozyme, catalytic oligonucleotides, nucleozyme, DNAzyme, RNA enzyme, endoribonuclease, endonuclease, minizyme, leadzyme, oligozyme or DNA enzyme. All of these terminologies describe nucleic acid molecules with enzymatic activity.
  • enzymatic nucleic acid molecules described in the instant application are not limiting in the invention and those skilled in the art will recognize that all that is important in an enzymatic nucleic acid molecule of this invention is that it has a specific substrate binding site that is complementary to one or more of the target nucleic acid regions, and that it have nucleotide sequences within or surrounding that substrate binding site which impart a nucleic acid cleaving and/or ligation activity to the molecule (Cech et al, U.S. Pat. No. 4,987,071; Cech et al., 1988, 260 JAMA 3030).
  • nucleic acid molecule as used herein is meant a molecule having nucleotides.
  • the nucleic acid can be single, double, or multiple stranded and can comprise modified or unmodified nucleotides or non-nucleotides or various mixtures and combinations thereof.
  • enzymatic portion or “catalytic domain” is meant that portion/region of the enzymatic nucleic acid molecule essential for cleavage of a nucleic acid substrate (for example see FIGS. 1 - 4 ).
  • substrate binding arm or “substrate binding domain” is meant that portion/region of a enzymatic nucleic acid that is able to interact, for example via complementarity (i.e., able to base-pair with), with a portion of its substrate.
  • complementarity can be 100%, but can be less if desired.
  • as few as 10 bases out of 14 can be base-paired (see for example Werner and Uhlenbeck, 1995 , Nucleic Acids Research, 23, 2092-2096; Hammann et al, 1999 , Antisense and Nucleic Acid Drug Dev., 9, 25-31). Examples of such arms are shown generally in FIGS. 1 - 4 .
  • these arms contain sequences within a enzymatic nucleic acid that are intended to bring enzymatic nucleic acid and target RNA together through complementary base-pairing interactions.
  • the enzymatic nucleic acid of the invention can have binding arms that are contiguous or non-contiguous and can be of varying lengths.
  • the length of the binding arm(s) can be greater than or equal to four nucleotides and of sufficient length to stably interact with a target RNA; in one embodiment they can be 12-100 nucleotides; in another embodiment they can be 14-24 nucleotides long (see for example Werner and Uhlenbeck, supra; Hamman et al., supra; Hampel et al., EP0360257; Berzal-Herranze et al., 1993 , EMBO J., 12, 2567-73) or between 8 and 14 nucleotides long.
  • the design is such that the length of the binding arms are symmetrical (i.e., each of the binding arms is of the same length; e.g., four and four, five and five nucleotides, or six and six nucleotides, or seven and seven nucleotides long) or asymmetrical (i.e., the binding arms are of different length; e.g., three and five, six and three nucleotides; three and six nucleotides long; four and five nucleotides long; four and six nucleotides long; four and seven nucleotides long; and the like).
  • Inozyme or “NCH” motif or configuration is meant, an enzymatic nucleic acid molecule comprising a motif as is generally described as NCH Rz in FIG. 1. Inozymes possess endonuclease activity to cleave RNA substrates having a cleavage triplet NCH/, where N is a nucleotide, C is cytidine and H is adenosine, uridine or cytidine, and / represents the cleavage site. H is used interchangeably with X.
  • Inozymes can also possess endonuclease activity to cleave RNA substrates having a cleavage triplet NCN/, where N is a nucleotide, C is cytidine, and / represents the cleavage site.
  • “I” in FIG. 1 represents an Inosine nucleotide, including a ribo-Inosine or xylo-Inosine nucleoside.
  • G-cleaver motif or configuration is meant, an enzymatic nucleic acid molecule comprising a motif as is generally described as G-cleaver Rz in FIG. 1.
  • G-cleavers possess endonuclease activity to cleave RNA substrates having a cleavage triplet NYN/, where N is a nucleotide, Y is uridine or cytidine and/represents the cleavage site.
  • G-cleavers can be chemically modified as is generally shown in FIG. 1.
  • Amberzyme motif or configuration an enzymatic nucleic acid molecule comprising a motif as is generally described in FIG. 2.
  • Amberzymes possess endonuclease activity to cleave RNA substrates having a cleavage triplet NG/N, where N is a nucleotide, G is guanosine, and/represents the cleavage site.
  • Amberzymes can be chemically modified to increase nuclease stability through substitutions as are generally shown in FIG. 2.
  • differing nucleoside and/or non-nucleoside linkers can be used to substitute the 5′-gaa-3′ loops shown in the figure.
  • Amberzymes represent a non-limiting example of an enzymatic nucleic acid molecule that does not require a ribonucleotide (2′-OH) group within its own nucleic acid sequence for activity.
  • Zinzyme motif or configuration an enzymatic nucleic acid molecule comprising a motif as is generally described in FIG. 3.
  • Zinzymes possess endonuclease activity to cleave RNA substrates having a cleavage triplet including but not limited to YG/Y, where Y is uridine or cytidine, and G is guanosine and/represents the cleavage site.
  • Zinzymes can be chemically modified to increase nuclease stability through substitutions as are generally shown in FIG. 3, including substituting 2′-O-methyl guanosine nucleotides for guanosine nucleotides.
  • differing nucleotide and/or non-nucleotide linkers can be used to substitute the 5′-gaaa-2′ loop shown in the figure.
  • Zinzymes represent a non-limiting example of an enzymatic nucleic acid molecule that does not require a ribonucleotide (2′-OH) group within its own nucleic acid sequence for activity.
  • DNAzyme is meant, an enzymatic nucleic acid molecule that does not require the presence of a 2′-OH group within its own nucleic acid sequence for activity.
  • the enzymatic nucleic acid molecule can have an attached linker or linkers or other attached or associated groups, moieties, or chains containing one or more nucleotides with 2′-OH groups.
  • DNAzymes can be synthesized chemically or expressed endogenously in vivo, by means of a single stranded DNA vector or equivalent thereof. An example of a DNAzyme is shown in FIG. 4 and is generally reviewed in Usman et al., U.S. Pat. No.
  • the “10-23” DNAzyme motif is one particular type of DNAzyme that was evolved using in vitro selection (see Santoro et al., supra). Additional DNAzyme motifs can be selected for using techniques similar to those described in these references, and hence, are within the scope of the present invention.
  • sufficient length is meant an oligonucleotide of greater than or equal to 3 nucleotides that is of a length great enough to provide the intended function under the expected condition.
  • sufficient length means that the binding arm sequence is long enough to provide stable binding to a target site under the expected binding conditions. The binding arms are not so long as to prevent useful turnover of the nucleic acid molecule.
  • stably interact is meant interaction of the oligonucleotides with target nucleic acid (e.g., by forming hydrogen bonds with complementary nucleotides in the target under physiological conditions) that is sufficient to the intended purpose (e.g., cleavage of target RNA by an enzyme).
  • RNA to PTGDS is meant to include RNA molecules having homology (partial or complete) to RNA molecules encoding PTGDS proteins or encoding proteins with similar function as PTGDS proteins in various organisms, including human, rodent, primate, rabbit, pig, plants, protozoans, fungi, and other microorganisms and parasites.
  • the equivalent RNA sequence can also include in addition to the coding region, regions such as 5′-untranslated region, 3′-untranslated region, introns, intron-exon junction and the like.
  • RNA to PTGDR is meant to include RNA molecules having homology (partial or complete) to RNA molecules encoding PTGDR proteins or encoding proteins with similar function as PTGDR proteins in various organisms, including human, rodent, primate, rabbit, pig, plants, protozoans, fungi, and other microorganisms and parasites.
  • the equivalent RNA sequence can also include in addition to the coding region, regions such as 5′-untranslated region, 3′-untranslated region, introns, intron-exon junction and the like.
  • RNA to ADORA1 is meant to include RNA molecules having homology (partial or complete) to RNA molecule encoding ADORA1 proteins or encoding proteins with similar function as ADORA1 proteins in various organisms, including human, rodent, primate, rabbit, pig, plants, protozoans, fungi, and other microorganisms and parasites.
  • the equivalent RNA sequence can also include in addition to the coding region, regions such as 5′-untranslated region, 3′-untranslated region, introns, intron-exon junction and the like.
  • nucleotide sequence of two or more nucleic acid molecules is partially or completely identical.
  • antisense nucleic acid a non-enzymatic nucleic acid molecule that binds to target RNA by means of RNA-RNA or RNA-DNA or RNA-PNA (protein nucleic acid; Egholm et al., 1993 Nature 365, 566) interactions and alters the activity of the target RNA (for a review, see Stein and Cheng, 1993 Science 261, 1004 and Woolf et al., U.S. Pat. No. 5,849,902).
  • antisense molecules are complementary to a target sequence along a single contiguous sequence of the antisense molecule.
  • an antisense molecule can bind to substrate such that the substrate molecule forms a loop, and/or an antisense molecule can bind such that the antisense molecule forms a loop.
  • the antisense molecule can be complementary to two (or even more) non-contiguous substrate sequences or two (or even more) non-contiguous sequence portions of an antisense molecule can be complementary to a target sequence or both.
  • antisense DNA can be used to target RNA by means of DNA-RNA interactions, thereby activating RNase H, which digests the target RNA in the duplex.
  • the antisense oligonucleotides can comprise one or more RNAse H activating region, which is capable of activating RNAse H cleavage of a target RNA.
  • Antisense DNA can be synthesized chemically or expressed via the use of a single stranded DNA expression vector or equivalent thereof.
  • RNase H activating region is meant a region (generally greater than or equal to 4-25 nucleotides in length, and in one embodiment from 5-11 nucleotides in length) of a nucleic acid molecule capable of binding to a target RNA to form a non-covalent complex that is recognized by cellular RNase H enzyme (see for example Arrow et al., U.S. Pat. No. 5,849,902; Arrow et al., U.S. Pat. No. 5,989,912).
  • the RNase H enzyme binds to the nucleic acid molecule-target RNA complex and cleaves the target RNA sequence.
  • the RNase H activating region comprises, for example, phosphodiester, phosphorothioate (at least four of the nucleotides are phosphorothiote substitutions; and in another embodiment, 4-11 of the nucleotides are phosphorothiote substitutions); phosphorodithioate, 5′-thiophosphate, or methylphosphonate backbone chemistry or a combination thereof.
  • the RNase H activating region can also comprise a variety of sugar chemistries.
  • the RNase H activating region can comprise deoxyribose, arabino, fluoroarabino or a combination thereof, nucleotide sugar chemistry.
  • 2-5A antisense chimera an antisense oligonucleotide containing a 5′-phosphorylated 2′-5′-linked adenylate residue. These chimeras bind to target RNA in a sequence-specific manner and activate a cellular 2-5A-dependent ribonuclease which, in turn, cleaves the target RNA (Torrence et al., 1993 Proc. Natl. Acad. Sci. USA 90, 1300; Silverman et al., 2000 , Methods Enzymol., 313, 522-533; Player and Torrence, 1998 , Pharmacol. Ther., 78, 55-113).
  • triplex forming oligonucleotides an oligonucleotide that can bind to a double-stranded DNA in a sequence-specific manner to form a triple-strand helix. Formation of such triple helix structure has been shown to inhibit transcription of the targeted gene (Duval-Valentin et al., 1992 Proc. Natl. Acad. Sci. USA 89, 504; Fox, 2000 , Curr. Med. Chem., 7, 17-37; Praseuth et. al., 2000 , Biochim. Biophys. Acta, 1489, 181-206).
  • RNA RNA
  • nucleic acid sequences including but not limited to structural genes encoding a polypeptide.
  • “Complementarity” refers to the ability of a nucleic acid to form hydrogen bond(s) with another RNA molecule by either traditional Watson-Crick or other non-traditional types.
  • the binding free energy for a nucleic acid molecule with its target or complementary sequence is sufficient to allow the relevant function of the nucleic acid to proceed, e.g., enzymatic nucleic acid cleavage, antisense or triple helix inhibition. Determination of binding free energies for nucleic acid molecules is well known in the art (see, e.g., Turner et al., 1987 , CSHSymp. Quant. Biol .
  • a percent complementarity indicates the percentage of contiguous residues in a nucleic acid molecule that can form hydrogen bonds (e.g., Watson-Crick base pairing) with a second nucleic acid sequence (e.g., 5, 6, 7, 8, 9, 10 out of 10 being 50%, 60%, 70%, 80%, 90%, and 100% complementary).
  • Perfectly complementary means that all the contiguous residues of a nucleic acid sequence will hydrogen bond with the same number of contiguous residues in a second nucleic acid sequence.
  • RNA is meant a molecule comprising at least one ribonucleotide residue.
  • ribonucleotide or “2′-OH” is meant a nucleotide with a hydroxyl group at the 2′ position of a ⁇ -D-ribo-furanose moiety.
  • decoy RNA is meant an RNA molecule or aptamer that is designed to preferentially bind to a predetermined ligand. Such binding can result in the inhibition or activation of a target molecule.
  • the decoy RNA or aptamer can compete with a naturally occurring binding target for the binding of a specific ligand.
  • TAR HIV trans-activation response
  • RNA can act as a “decoy” and efficiently binds HIV tat protein, thereby preventing it from binding to TAR sequences encoded in the HIV RNA (Sullenger et al., 1990, Cell, 63, 601-608).
  • a decoy RNA can be designed to bind to a D2 receptor and block the binding of PTGDS or a decoy RNA can be designed to bind to PTGDS and prevent interaction with the D2 receptor.
  • double stranded RNA or “dsRNA” as used herein refers to a double stranded RNA molecule capable of RNA interference, including short interfering RNA “siRNA” (see, e.g., Bass, 2001 , Nature, 411, 428-429; Elbashir et al., 2001 , Nature, 411, 494-498).
  • siRNA short interfering RNA
  • allozyme refers to an allosteric enzymatic nucleic acid molecule, see, e.g., George et al., U.S. Pat. Nos. 5,834,186 and 5,741,679, Shih et al., U.S. Pat. No. 5,589,332, Nathan et al., U.S. Pat. No. 5,871,914, Nathan and Ellington, International PCT publication No. WO 00/24931, Breaker et al., International PCT Publication Nos. WO 00/26226 and 98/27104, and Sullenger et al., International PCT publication No. WO 99/29842.
  • 2-5A chimera refers to an oligonucleotide containing a 5′-phosphorylated 2′-5′-linked adenylate residue. These chimeras bind to target RNA in a sequence-specific manner and activate a cellular 2-5A-dependent ribonuclease which, in turn, cleaves the target RNA (Torrence et al., 1993 Proc. Natl. Acad. Sci. USA 90, 1300; Silverman et al., 2000 , Methods Enzymol., 313, 522-533; Player and Torrence, 1998 , Pharmacol. Ther., 78, 55-113).
  • triplex forming oligonucleotides refers to an oligonucleotide that can bind to a double-stranded DNA in a sequence-specific manner to form a triple-strand helix. Formation of such triple helix structure has been shown to inhibit transcription of the targeted gene (Duval-Valentin et al., 1992 Proc. Natl. Acad. Sci. USA 89, 504; Fox, 2000 , Curr. Med. Chem., 7, 17-37; Praseuth et. al., 2000 , Biochim. Biophys. Acta, 1489, 181-206).
  • enzymatic nucleic acids act by first binding to a target RNA. Such binding occurs through the target binding portion of a enzymatic nucleic acid that is held in close proximity to an enzymatic portion of the molecule that acts to cleave the target RNA.
  • the enzymatic nucleic acid first recognizes and then binds a target RNA through complementary base-pairing, and once bound to the correct site, acts enzymatically to cut the target RNA. Strategic cleavage of such a target RNA will destroy its ability to direct synthesis of an encoded protein. After an enzymatic nucleic acid has bound and cleaved its RNA target, it is released from that RNA to search for another target and can repeatedly bind and cleave new targets. Thus, a single ribozyme molecule is able to cleave many molecules of target RNA.
  • the ribozyme is a highly specific inhibitor of gene expression, with the specificity of inhibition depending not only on the base-pairing mechanism of binding to the target RNA, but also on the mechanism of target RNA cleavage. Single mismatches, or base-substitutions, near the site of cleavage can completely eliminate catalytic activity of a ribozyme.
  • the enzymatic nucleic acid molecule that cleave the specified sites in PTGDS, ADORA1 and PTGDR-specific RNAs represent a novel therapeutic approach to treat a variety of allergic diseases or conditions, including but not limited to asthma, allergic rhinitis, atopic dermatitis, and/or other allergic or inflammatory diseases and conditions which respond to the modulation of PTGDS, ADORA1 and/or PTGDR expression.
  • the enzymatic nucleic acid molecule is formed in a hammerhead or hairpin motif, but can also be formed in the motif of a hepatitis delta virus, group I intron, group II intron or RNase P RNA (in association with an RNA guide sequence), Neurospora VS RNA, DNAzymes, NCH cleaving motifs, or G-cleavers.
  • Group II introns are described by Griffin et al., 1995 , Chem. Biol. 2, 761; Michels and Pyle, 1995 , Biochemistry 34, 2965; Pyle et al., International PCT Publication No. WO 96/22689; of the Group I intron by Cech et al., U.S. Pat. No. 4,987,071 and of DNAzymes by Usman et al., International PCT Publication No. WO 95/11304; Chartrand et al., 1995 , NAR 23, 4092; Breaker et al., 1995 , Chem. Bio.
  • a nucleic acid molecule of the instant invention can be between 12 and 100 nucleotides in length.
  • Exemplary enzymatic nucleic acid molecules of the invention are shown in Table III-VII.
  • enzymatic nucleic acid molecules of the invention can be between 15 and 50 nucleotides in length, and in another embodiment between 25 and 40 nucleotides in length, e.g., 34, 36, or 38 nucleotides in length (for example see Jarvis et al., 1996 , J. Biol. Chem., 271, 29107-29112).
  • Exemplary DNAzymes of the invention are can between 15 and 40 nucleotides in length, and in one embodiment, between 25 and 35 nucleotides in length, e.g., 29, 30, 31, or 32 nucleotides in length (see, e.g., Santoro et al., 1998 , Biochemistry, 37, 13330-13342; Chartrand et al., 1995, Nucleic Acids Research, 23, 4092-4096).
  • Exemplary antisense molecules of the invention can be between 15 and 75 nucleotides in length, and in one embodiment between 20 and 35 nucleotides in length, e.g., 25, 26, 27, or 28 nucleotides in length (see for example Woolf et al., 1992 , PNAS., 89, 7305-7309; Milner et al., 1997 , Nature Biotechnology, 15, 537-541).
  • Exemplary triplex forming oligonucleotide molecules of the invention are between 10 and 40 nucleotides in length, and in one embodiment are between 12 and 25 nucleotides in length, e.g., 18, 19, 20, or 21 nucleotides in length (see for example Maher et al., 1990 , Biochemistry, 29, 8820-8826; Strobel and Dervan, 1990 , Science, 249, 73-75).
  • Those skilled in the art will recognize that all that is required is for the nucleic acid molecule to be of length and conformation sufficient and suitable for the nucleic acid molecule to catalyze a reaction contemplated herein.
  • the length of the nucleic acid molecules of the instant invention are not limiting within the general limits stated.
  • a nucleic acid molecule that modulates, for example, down-regulates, PTGDS replication or expression comprises between 8 and 100 bases complementary to a RNA molecule of PTGDS. In another embodiment, a nucleic acid molecule that modulates PTGDS replication or expression comprises between 14 and 24 bases complementary to a RNA molecule of PTGDS.
  • a nucleic acid molecule that modulates, for example, down-regulates, PTGDR replication or expression comprises between 8 and 100 bases complementary to a RNA molecule of PTGDR. In another embodiment, a nucleic acid molecule that modulates PTGDR replication or expression comprises between 14 and 24 bases complementary to a RNA molecule of PTGDR.
  • a nucleic acid molecule that modulates, for example, down-regulates, ADORA1 replication or expression comprises between 8 and 100 bases complementary to a RNA molecule of ADORA1.
  • a nucleic acid molecule that modulates ADORA1 replication or expression comprises between 14 and 24 bases complementary to a RNA molecule of ADORA1.
  • the invention provides a method for producing a class of nucleic acid-based gene modulating agents that exhibit a high degree of specificity for the RNA of a desired target.
  • the enzymatic nucleic acid molecule is can be targeted to a highly conserved sequence region of target RNAs encoding PTGDS, ADORA1 and/or PTGDR (e.g., PTGDS, ADORA1 and/or PTGDR genes) such that specific treatment of a disease or condition can be provided with either one or several nucleic acid molecules of the invention.
  • Such nucleic acid molecules can be delivered exogenously to specific tissue or cellular targets as required.
  • the nucleic acid molecules e.g., ribozymes and antisense
  • cell is used in its usual biological sense, and does not refer to an entire multicellular organism.
  • the cell can, for example, be in vitro, e.g., in cell culture, or present in a multicellular organism, including, e.g., birds, plants and mammals such as humans, cows, sheep, apes, monkeys, swine, dogs, and cats.
  • the cell may be prokaryotic (e.g., bacterial cell) or eukaryotic (e.g., mammalian or plant cell).
  • PGPDR proteins is meant, a protein receptor or a mutant protein or peptide derivative thereof, having prostaglandin D2 receptor activity, for example, having the ability to bind prostaglandin D2 and/or having GTP-binding protein coupled activity.
  • PGHDS proteins is meant, a prostaglandin synthetase protein or a mutant protein or peptide derivative thereof, having prostaglandin D2 synthetase activity, for example, having the ability to convert PGH2 to PGD2.
  • highly conserved sequence region is meant, a nucleotide sequence of one or more regions in a target gene does not vary significantly from one generation to the other or from one biological system to the other.
  • Nucleic acid-based inhibitors of PTGDS, ADORA1 and PTGDR expression are useful for the prevention and/or treatment of allergic diseases or conditions, including but not limited to asthma, allergic rhinitis, atopic dermatitis, and any other diseases or conditions that are related to or will respond to the levels of PTGDS, ADORA1 and/or PTGDR in a cell or tissue, alone or in combination with other therapies.
  • the reduction of PTGDS, ADORA1 and/or PTGDR expression specifically PTGDS, ADORA1 and/or PTGDR gene RNA levels
  • thus reduction in the level of the respective protein relieves, to some extent, the symptoms of the disease or condition.
  • the nucleic acid-based inhibitors of the invention can be added directly, or can be complexed with cationic lipids, packaged within liposomes, or otherwise delivered to target cells or tissues, for example by pulmonary delivery of an aerosol formulation with an inhaler or nebulizer.
  • the nucleic acid or nucleic acid complexes can be locally administered to relevant tissues ex vivo, or in vivo through inhalation, injection or infusion pump, with or without their incorporation in biopolymers.
  • the enzymatic nucleic acid inhibitors comprise sequences that are complementary to the substrate sequences in Tables III to VII. Examples of such enzymatic nucleic acid molecules also are shown in Tables III to VII. Examples of such enzymatic nucleic acid molecules consist essentially of sequences defined in these tables.
  • the invention features antisense nucleic acid molecules and 2-5A chimera including sequences complementary to the substrate sequences shown in Tables III to VII.
  • nucleic acid molecules can include sequences as shown for the binding arms of the enzymatic nucleic acid molecules in Tables III to VII.
  • triplex molecules can be provided targeted to the corresponding DNA target regions, and containing the DNA equivalent of a target sequence or a sequence complementary to the specified target (substrate) sequence.
  • antisense molecules are complementary to a target sequence along a single contiguous sequence of the antisense molecule.
  • an antisense molecule can bind to substrate such that the substrate molecule forms a loop, and/or an antisense molecule can bind such that the antisense molecule forms a loop.
  • the antisense molecule can be complementary to two (or even more) non-contiguous substrate sequences or two (or even more) non-contiguous sequence portions of an antisense molecule can be complementary to a target sequence or both.
  • the active nucleic acid molecule of the invention for example, an enzymatic nucleic acid molecule, contains an enzymatic center or core equivalent to those in the examples, and binding arms able to bind RNA such that cleavage at the target site occurs.
  • a core region can, for example, include one or more loop, stem-loop structure, or linker which does not prevent enzymatic activity.
  • the underlined regions in the sequences in Tables III and IV can be such a loop, stem-loop, nucleotide linker, and/or non-nucleotide linker and can be represented generally as sequence “X”.
  • a core sequence for a hammerhead enzymatic nucleic acid can comprise a conserved sequence, such as 5′-CUGAUGAG-3′ and 5′-CGAA-3′ connected by “X”, where X is 5′- GCCGUUAGGC -3′ (SEQ ID NO: 2678), or any other Stem II region known in the art, or a nucleotide and/or non-nucleotide linker.
  • nucleic acid molecules of the instant invention such as Inozyme, G-cleaver, amberzyme, zinzyme, DNAzyme, antisense, 2-5A antisense, triplex forming nucleic acid, and decoy nucleic acids
  • other sequences or non-nucleotide linkers can be present that do not interfere with the function of the nucleic acid molecule.
  • Sequence X can be a linker of ⁇ 2 nucleotides in length, including 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 26, 30, where the nucleotides can be internally base-paired to form a stem of ⁇ 2 base pairs.
  • sequence X can be a non-nucleotide linker.
  • the nucleotide linker X can be a nucleic acid aptamer, such as an ATP aptamer, HIV Rev aptamer (RRE), HIV Tat aptamer (TAR) and others (for a review see Gold et al., 1995 , Annu. Rev.
  • nucleic acid aptamer as used herein is meant to indicate a nucleic acid sequence capable of interacting with a ligand.
  • the ligand can be any natural or a synthetic molecule, including but not limited to a resin, metabolites, nucleosides, nucleotides, drugs, toxins, transition state analogs, peptides, lipids, proteins, amino acids, nucleic acid molecules, hormones, carbohydrates, receptors, cells, viruses, bacteria and others.
  • non-nucleotide linker X is as defined herein.
  • non-nucleotide include either abasic nucleotide, polyether, polyamine, polyamide, peptide, carbohydrate, lipid, or polyhydrocarbon compounds. Specific examples include those described by Seela and Kaiser, Nucleic Acids Res. 1990, 18:6353 and Nucleic Acids Res. 1987, 15:3113; Cload and Schepartz, J. Am. Chem. Soc. 1991, 113:6324; Richardson and Schepartz, J. Am. Chem. Soc. 1991, 113:5109; Ma et al., Nucleic Acids Res.
  • non-nucleotide further means any group or compound that can be incorporated into a nucleic acid chain in the place of one or more nucleotide units, including either sugar and/or phosphate substitutions, and allows the remaining bases to exhibit their enzymatic activity.
  • the group or compound can be abasic in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine.
  • the invention features an enzymatic nucleic acid molecule having one or more non-nucleotide moieties, and having enzymatic activity to cleave an RNA or DNA molecule.
  • enzymatic nucleic acid molecules or antisense molecules that interact with target RNA molecules and down-regulate PTGDS, ADORA1 and/or PTGDR (e.g., PTGDS, ADORA1 and/or PTGDR gene) activity are expressed from transcription units inserted into DNA or RNA vectors.
  • the recombinant vectors can be DNA plasmids or viral vectors.
  • Enzymatic nucleic acid molecule or antisense expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus, retrovirus, adenovirus, or alphavirus.
  • the recombinant vectors capable of expressing the enzymatic nucleic acid molecules or antisense can be delivered as described above, and persist in target cells.
  • viral vectors can be used that provide for transient expression of enzymatic nucleic acid molecules or antisense. Such vectors can be repeatedly administered as necessary.
  • the enzymatic nucleic acid molecules or antisense bind to the target RNA and down-regulate its function or expression.
  • Delivery of enzymatic nucleic acid molecule or antisense expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex-planted from the patient followed by reintroduction into the patient, or by any other means that would allow for introduction into the desired target cell.
  • Antisense DNA can be expressed via the use of a single stranded DNA intracellular expression vector.
  • vectors any nucleic acid- and/or viral-based technique used to deliver a desired nucleic acid.
  • patient is meant an organism, which is a donor or recipient of explanted cells, or the cells themselves. “Patient” also refers to an organism to which the nucleic acid molecules of the invention can be administered.
  • a patient can be a mammal or mammalian cells. In one embodiment, a patient is a human or human cells.
  • enhanced enzymatic activity is meant to include activity measured in cells and/or in vivo where the activity is a reflection of both the catalytic activity and the stability of the nucleic acid molecules of the invention.
  • the product of these properties can be increased in vivo compared to an all RNA enzymatic nucleic acid or all DNA enzyme.
  • the activity or stability of the nucleic acid molecule can be decreased (i.e., less than ten-fold), but the overall activity of the nucleic acid molecule is enhanced, in vivo.
  • nucleic acid molecules of the instant invention can be used to treat diseases or conditions discussed above.
  • the patient can be treated, or other appropriate cells can be treated, as is evident to those skilled in the art, individually or in combination with one or more drugs under conditions suitable for the treatment.
  • the described molecules can be used in combination with other known treatments to treat conditions or diseases discussed above.
  • the described molecules can be used in combination with one or more known therapeutic agents to treat allergic diseases or conditions, including but not limited to asthma, allergic rhinitis, atopic dermatitis, and/or other allergic or inflammatory diseases and conditions which respond to the modulation of PTGDS, ADORA1 and/or PTGDR expression.
  • the invention features nucleic acid-based inhibitors (e.g., enzymatic nucleic acid molecules (e.g., ribozymes), antisense nucleic acids, 2-5A antisense chimeras, triplex DNA, antisense nucleic acids containing RNA cleaving chemical groups) and methods for their use to down regulate or inhibit the expression of genes (e.g., PTGDS, ADORA1 and/or PTGDR) capable of progression and/or maintenance allergic diseases or conditions, including but not limited to asthma, allergic rhinitis, atopic dermatitis, and/or other allergic or inflammatory diseases and conditions which respond to the modulation of PTGDS, ADORA1 and/or PTGDR expression.
  • genes e.g., PTGDS, ADORA1 and/or PTGDR
  • FIG. 1 shows examples of chemically stabilized ribozyme motifs.
  • HH Rz represents hammerhead ribozyme motif (Usman et al., 1996 , Curr. Op. Struct. Bio., 1, 527);
  • NCH Rz represents the NCH ribozyme motif (Ludwig & Sproat, International PCT Publication No. WO 98/58058);
  • G-Cleaver represents G-cleaver ribozyme motif (Kore et al., 1998 , Nucleic Acids Research 26, 4116-4120, Eckstein et al., International PCT publication No. WO 99/16871).
  • N or n represent independently a nucleotide that can be same or different and have complementarity to each other; rI, represents ribo-Inosine nucleotide; arrow indicates the site of cleavage within the target.
  • Position 4 of the HH Rz and the NCH Rz is shown as having 2′-C-allyl modification, but those skilled in the art will recognize that this position can be modified with other modifications well known in the art, so long as such modifications do not significantly inhibit the activity of the ribozyme.
  • FIG. 2 shows an example of the Amberzyme ribozyme motif that is chemically stabilized (see for example Beigelman et al., International PCT publication No. WO 99/55857).
  • FIG. 3 shows an example of the Zinzyme A ribozyme motif that is chemically stabilized (see for example Beigelman et al., Beigelman et al., International PCT publication No. WO 99/55857).
  • FIG. 4 shows an example of a specific DNAzyme motif, commonly referred to as the “10-23 motif”, as described by Santoro et al., 1997 , PNAS, 94, 4262.
  • Antisense molecules can be modified or unmodified RNA, DNA, or mixed polymer oligonucleotides and primarily function by specifically binding to matching sequences resulting in inhibition of peptide synthesis (Wu-Pong, Nov 1994 , BioPharm, 20-33).
  • the antisense oligonucleotide binds to target RNA by Watson Crick base-pairing and blocks gene expression by preventing ribosomal translation of the bound sequences either by steric blocking or by activating RNase H enzyme.
  • Antisense molecules can also alter protein synthesis by interfering with RNA processing or transport from the nucleus into the cytoplasm (Mukhopadhyay & Roth, 1996 , Crit. Rev. in Oncogenesis 7, 151-190).
  • antisense molecules have been described that utilize novel configurations of chemically modified nucleotides, secondary structure, and/or RNase H substrate domains (Woolf et al., International PCT Publication No. WO 98/13526; Thompson et al., International PCT Publication No. WO 99/54459; Hartmann et al., U.S. S No. 60/101,174, filed on Sep. 21, 1998) all of these are incorporated by reference herein in their entirety.
  • antisense deoxyoligoribonucleotides can be used to target RNA by means of DNA-RNA interactions, thereby activating RNase H, which digests the target RNA in the duplex.
  • Antisense DNA can be expressed via the use of a single stranded DNA intracellular expression vector or equivalents and variations thereof.
  • TFO Triplex Forming Oligonucleotides
  • 2-5A Antisense Chimera The 2-5A system is an interferon mediated mechanism for RNA degradation found in higher vertebrates (Mitra et al., 1996 , Proc Nat Acad Sci USA 93, 6780-6785). Two types of enzymes, 2-5A synthetase and RNase L, are required for RNA cleavage. The 2-5A synthetases require double stranded RNA to form 2′-5′ oligoadenylates (2-5A). 2-5A then acts as an allosteric effector for utilizing RNase L, which has the ability to cleave single stranded RNA. The ability to form 2-5A structures with double stranded RNA makes this system particularly useful for inhibition of viral replication.
  • (2′-5′) oligoadenylate structures can be covalently linked to antisense molecules to form chimeric oligonucleotides capable of RNA cleavage (Torrence, supra). These molecules putatively bind and activate a 2-5A dependent RNase, the oligonucleotide/enzyme complex then binds to a target RNA molecule which can then be cleaved by the RNase enzyme.
  • Enzymatic Nucleic Acid Several varieties of naturally-occurring enzymatic RNAs are presently known. In addition, several in vitro selection (evolution) strategies (Orgel, 1979, Proc. R. Soc. London , B 205, 435) have been used to evolve new nucleic acid catalysts capable of catalyzing cleavage and ligation of phosphodiester linkages (Joyce, 1989 , Gene, 82, 83-87; Beaudry et al., 1992 , Science 257, 635-641; Joyce, 1992 , Scientific American 267, 90-97; Breaker et al., 1994 , TIBTECH 12, 268; Bartel et al., 1993 , Science 261:1411-1418; Szostak, 1993 , TIBS 17, 89-93; Kumar et al., 1995 , FASEB J, 9, 1183; Breaker, 1996, Curr.
  • the enzymatic nature of an enzymatic nucleic acid molecule has significant advantages, one advantage being that the concentration of enzymatic nucleic acid molecule necessary to affect a therapeutic treatment is lower. This advantage reflects the ability of the enzymatic nucleic acid molecule to act enzymatically. Thus, a single enzymatic nucleic acid molecule is able to cleave many molecules of target RNA.
  • the enzymatic nucleic acid molecule is a highly specific inhibitor, with the specificity of inhibition depending not only on the base-pairing mechanism of binding to the target RNA, but also on the mechanism of target RNA cleavage. Single mismatches, or base-substitutions, near the site of cleavage can be chosen to completely eliminate catalytic activity of a enzymatic nucleic acid molecule.
  • Nucleic acid molecules having an endonuclease enzymatic activity are able to repeatedly cleave other separate RNA molecules in a nucleotide base sequence-specific manner. With the proper design, such enzymatic nucleic acid molecules can be targeted to RNA transcripts, and achieve efficient cleavage in vitro (Zaug et al., 324 , Nature 429 1986; Uhlenbeck, 1987 Nature 328, 596; Kim et al., 84 Proc. Natl. Acad. Sci. USA 8788, 1987; Dreyfus, 1988 , Einstein Quart. J. Bio.
  • trans-cleaving enzymatic nucleic acid molecules can be used as therapeutic agents for human disease (Usman & McSwiggen, 1995 Ann. Rep. Med. Chem. 30, 285-294; Christoffersen and Marr, 1995 J. Med. Chem. 38, 2023-2037).
  • Enzymatic nucleic acid molecules can be designed to cleave specific RNA targets within the background of cellular RNA. Such a cleavage event renders the RNA non-functional and abrogates protein expression from that RNA. In this manner, synthesis of a protein associated with a disease state can be selectively inhibited (Warashina et al., 1999 , Chemistry and Biology, 6, 237-250).
  • Enzymatic nucleic acid molecules of the invention that are allosterically regulated (“allozymes”) can be used to down-regulate PTGDS and/or PTGDR expression.
  • allosteric enzymatic nucleic acids or allozymes see for example George et al., U.S. Pat. Nos. 5,834,186 and 5,741,679, Shih et al., U.S. Pat. No. 5,589,332, Nathan et al., U.S. Pat. No. 5,871,914, Nathan and Ellington, International PCT publication No. WO 00/24931, Breaker et al., International PCT Publication Nos.
  • WO 00/26226 and 98/27104 are designed to respond to a signaling agent, for example, mutant PTGDS and/or PTGDR protein, wild-type PTGDS and/or PTGDR protein, mutant PTGDS and/or PTGDR RNA, wild-type PTGDS and/or PTGDR RNA, other proteins and/or RNAs involved in PTGDS or PTGDR signal transduction, compounds, metals, polymers, molecules and/or drugs that are targeted to PTGDS and/or PTGDR expressing cells etc., which in turn modulates the activity of the enzymatic nucleic acid molecule.
  • a signaling agent for example, mutant PTGDS and/or PTGDR protein, wild-type PTGDS and/or PTGDR protein, mutant PTGDS and/or PTGDR RNA, wild-type PTGDS and/or PTGDR RNA, other proteins and/or RNAs involved in PTGDS or PTGDR signal transduction, compounds, metals, polymers, molecules and/or drugs that are
  • the allosteric enzymatic nucleic acid molecule's activity is activated or inhibited such that the expression of a particular target is selectively down-regulated.
  • the target can comprise wild-type PTGDS, ADORA1 and/or PTGDR, mutant PTGDS, ADORA1 and/or PTGDR, and/or a predetermined component of the PTGDS, ADORA1 or PTGDR signal transduction pathway.
  • allosteric enzymatic nucleic acid molecules that are activated by interaction with a RNA encoding a PTGDR protein are used as therapeutic agents in vivo.
  • RNA encoding the PTGDS protein activates the allosteric enzymatic nucleic acid molecule that subsequently cleaves the RNA encoding a PTGDR protein resulting in the inhibition of PTGDR protein expression. In this manner, cells that express both PTGDS and PTGDR protein are selectively targeted.
  • an allozyme can be activated by a PTGDS or PTGDR protein, peptide, or mutant polypeptide that causes the allozyme to inhibit the expression of PTGDS or PTGDR gene, by, for example, cleaving RNA encoded by PTGDS or PTGDR gene.
  • the allozyme acts as a decoy to inhibit the function of PTGDS or PTGDR and also inhibit the expression of PTGDS or PTGDR once activated by the PTGDS or PTGDR protein.
  • Targets for useful enzymatic nucleic acid molecules and antisense nucleic acids can be determined as disclosed in Draper et al., WO 93/23569; Sullivan et al, WO 93/23057; Thompson et al., WO 94/02595; Draper et al., WO 95/04818; McSwiggen et al., U.S. Pat. No. 5,525,468, and hereby incorporated by reference herein in totality.
  • Other examples include the following PCT applications, which concern inactivation of expression of disease-related genes: WO 95/23225, WO 95/13380, WO 94/02595, incorporated by reference herein.
  • Enzymatic nucleic acid molecules and antisense to such targets are designed as described in those applications and synthesized to be tested in vitro and in vivo, as also described.
  • the sequences of human PTGDR RNAs were screened for optimal enzymatic nucleic acid and antisense target sites using a computer-folding algorithm.
  • Antisense, hammerhead, DNAzyme, NCH, amberzyme, zinzyme, or G-Cleaver enzymatic nucleic acid molecule binding/cleavage sites were identified.
  • nucleic acids greater than 100 nucleotides in length is difficult using automated methods, and the therapeutic cost of such molecules is prohibitive.
  • small nucleic acid motifs (“small refers to nucleic acid motifs less than about 100 nucleotides in length, and in one embodiment less than about 80 nucleotides in length, and in another embodiment less than about 50 nucleotides in length; e.g., antisense oligonucleotides, hammerhead or the NCH ribozymes) can be used for exogenous delivery.
  • the simple structure of these molecules increases the ability of the nucleic acid to invade targeted regions of RNA structure.
  • Exemplary molecules of the instant invention are chemically synthesized, and others can similarly be synthesized.
  • Oligonucleotides are synthesized using protocols known in the art as described in Caruthers et al., 1992 , Methods in Enzymology 211, 3-19, Thompson et al., International PCT Publication No. WO 99/54459, Wincott et al., 1995 , Nucleic Acids Res. 23, 2677-2684, Wincott et al., 1997 , Methods Mol. Bio., 74, 59, Brennan et al., 1998 , Biotechnol Bioeng., 61, 33-45, and Brennan, U.S. Pat. No. 6,001,311. All of these references are incorporated herein by reference.
  • oligonucleotides makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5′-end, and phosphoramidites at the 3′-end.
  • small scale syntheses are conducted on a 394 Applied Biosystems, Inc. synthesizer using a 0.2 ⁇ mol scale protocol with a 2.5 min coupling step for 2′-O-methylated nucleotides and a 45 sec coupling step for 2′-deoxy nucleotides.
  • Table II outlines the amounts and the contact times of the reagents used in the synthesis cycle.
  • syntheses at the 0.2 ⁇ mol scale can be performed on a 96-well plate synthesizer, such as the instrument produced by Protogene (Palo Alto, Calif.) with minimal modification to the cycle.
  • Average coupling yields on the 394 Applied Biosystems, Inc. synthesizer, determined by calorimetric quantitation of the trityl fractions, are typically 97.5-99%.
  • synthesizer include; detritylation solution is 3% TCA in methylene chloride (ABI); capping is performed with 16% N-methylimidazole in THF (ABI) and 10% acetic anhydride/10% 2,6-lutidine in THF (ABI); and oxidation solution is 16.9 mM 12, 49 mM pyridine, 9% water in THF (PERSEPTIVETM). Burdick & Jackson Synthesis Grade acetonitrile is used directly from the reagent bottle. S-Ethyltetrazole solution (0.25 M in acetonitrile) is made up from the solid obtained from American International Chemical, Inc. Alternately, for the introduction of phosphorothioate linkages, Beaucage reagent (3H-1,2-Benzodithiol-3-one 1,1-dioxide, 0.05 M in acetonitrile) is used.
  • Deprotection of the antisense oligonucleotides is performed as follows: the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 40% aq. methylamine (1 mL) at 65° C. for 10 min. After cooling to ⁇ 20° C., the supernatant is removed from the polymer support. The support is washed three times with 1.0 mL of EtOH:MeCN:H 2 O/3:1:1, vortexed and the supernatant is then added to the first supernatant. The combined supernatants, containing the oligoribonucleotide, are dried to a white powder.
  • small scale syntheses are conducted on a 394 Applied Biosystems, Inc. synthesizer using a 0.2 ⁇ mol scale protocol with a 7.5 min coupling step for alkylsilyl protected nucleotides and a 2.5 min coupling step for 2′-O-methylated nucleotides.
  • Table II outlines the amounts and the contact times of the reagents used in the synthesis cycle.
  • syntheses at the 0.2 ⁇ mol scale can be done on a 96-well plate synthesizer, such as the instrument produced by Protogene (Palo Alto, Calif.) with minimal modification to the cycle.
  • Average coupling yields on the 394 Applied Biosystems, Inc. synthesizer, determined by colorimetric quantitation of the trityl fractions, are typically 97.5-99%.
  • synthesizer include; detritylation solution is 3% TCA in methylene chloride (ABI); capping is performed with 16% N-methylimidazole in THF (ABI) and 10% acetic anhydride/10% 2,6-lutidine in THF (ABI); oxidation solution is 16.9 mM 12, 49 mM pyridine, 9% water in THF (PERSEPTIVETM). Burdick & Jackson Synthesis Grade acetonitrile is used directly from the reagent bottle. S-Ethyltetrazole solution (0.25 M in acetonitrile) is made up from the solid obtained from American International Chemical, Inc. Alternately, for the introduction of phosphorothioate linkages, Beaucage reagent (3H-1,2-Benzodithiol-3-one 1,1-dioxide 0.05 M in acetonitrile) is used.
  • RNA Deprotection of the RNA is performed using either a two-pot or one-pot protocol.
  • the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 40% aq. methylamine (1 mL) at 65° C. for 10 min. After cooling to ⁇ 20° C., the supernatant is removed from the polymer support. The support is washed three times with 1.0 mL of EtOH:MeCN:H2O/3:1:1, vortexed and the supernatant is then added to the first supernatant.
  • the combined supernatants, containing the oligoribonucleotide, are dried to a white powder.
  • the base deprotected oligoribonucleotide is resuspended in anhydrous TEA/HF/NMP solution (300 ⁇ L of a solution of 1.5 mL N-methylpyrrolidinone, 750 ⁇ L TEA and 1 mL TEA.3HF to provide a 1.4 M HF concentration) and heated to 65° C. After 1.5 h, the oligomer is quenched with 1.5 M NH 4 HCO 3 .
  • the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 33% ethanolic methylamine/DMSO: 1/1 (0.8 mL) at 65° C. for 15 min.
  • the vial is brought to r.t. TEA.3HF (0.1 mL) is added and the vial is heated at 65° C. for 15 min.
  • the sample is cooled at ⁇ 20° C. and then quenched with 1.5 M NH 4 HCO 3 .
  • the quenched NH 4 HCO 3 solution is loaded onto a C-18 containing cartridge that had been prewashed with acetonitrile followed by 50 mM TEAA. After washing, the loaded cartridge with water, the RNA is detritylated with 0.5% TFA for 13 min. The cartridge is then washed again with water, salt exchanged with 1 M NaCl and washed with water again. The oligonucleotide is then eluted with 30% acetonitrile.
  • Inactive hammerhead ribozymes or binding attenuated control (BAC) oligonucleotides are synthesized by substituting a U for G 5 and a U for A14 (numbering from Hertel, K. J., et al., 1992 , Nucleic Acids Res., 20, 3252). Similarly, one or more nucleotide substitutions can be introduced in other enzymatic nucleic acid molecules to inactivate the molecule and such molecules can serve as a negative control.
  • the average stepwise coupling yields are typically >98% (Wincott et al., 1995 Nucleic Acids Res. 23, 2677-2684).
  • the scale of synthesis can be adapted to be larger or smaller than the example described above including but not limited to 96 well format, all that is important is the ratio of chemicals used in the reaction.
  • nucleic acid molecules of the present invention can be synthesized separately and joined together post-synthetically, for example by ligation (Moore et al., 1992 , Science 256, 9923; Draper et al., International PCT publication No. WO 93/23569; Shabarova et al., 1991 , Nucleic Acids Research 19, 4247; Bellon et al., 1997 , Nucleosides & Nucleotides, 16, 951; Bellon et al., 1997 , Bioconjugate Chem. 8, 204).
  • nucleic acid molecules of the present invention can be modified extensively to enhance stability by modification with nuclease resistant groups, for example, 2′-amino, 2′-C-allyl, 2′-flouro, 2′-O-methyl, 2′-H (for a review see Usman and Cedergren, 1992 , TIBS 17, 34; Usman et al., 1994 , Nucleic Acids Symp. Ser. 31, 163).
  • Ribozymes are purified by gel electrophoresis using general methods or are purified by high pressure liquid chromatography (HPLC; See Wincott et al., Supra, the totality of which is hereby incorporated herein by reference) and are re-suspended in water.
  • the sequences of the nucleic acid molecules, including enzymatic nucleic acid molecules and antisense, that are chemically synthesized, are shown in Tables III-VII.
  • the sequences of the enzymatic nucleic acid constructs that are chemically synthesized are complementary to the Substrate sequences shown in Tables III-VII. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the enzymatic nucleic acid (all but the binding arms) is altered to affect activity.
  • the enzymatic nucleic acid construct sequences listed in Tables III-VII can be formed of ribonucleotides or other nucleotides or non-nucleotides. Such enzymatic nucleic acid molecules with enzymatic activity are equivalent to the enzymatic nucleic acid molecules described specifically in the Tables.
  • oligonucleotides are modified to enhance stability and/or enhance biological activity by modification with nuclease resistant groups, for example, 2′-amino, 2′-C-allyl, 2′-flouro, 2′-O-methyl, 2′-H, nucleotide base modifications (for a review see Usman and Cedergren, 1992, TIBS. 17, 34; Usman et al., 1994 , Nucleic Acids Symp. Ser. 31, 163; Burgin et al., 1996 , Biochemistry, 35, 14090).
  • nuclease resistant groups for example, 2′-amino, 2′-C-allyl, 2′-flouro, 2′-O-methyl, 2′-H, nucleotide base modifications
  • Nucleic acid molecules having chemical modifications that maintain or enhance activity are provided. Such a nucleic acid is also generally more resistant to nucleases than an unmodified nucleic acid. Thus, in a cell and/or in vivo the activity may not be significantly lowered.
  • Therapeutic nucleic acid molecules delivered exogenously are optimally stable within cells until translation of the target RNA has been inhibited long enough to reduce the levels of the undesirable protein. This period of time varies between hours to days depending upon the disease state.
  • nucleic acid molecules must be resistant to nucleases in order to function as effective intracellular therapeutic agents. Improvements in the chemical synthesis of RNA and DNA (Wincott et al., 1995 Nucleic Acids Res.
  • nucleic acid molecules of the invention include one or more G-clamp nucleotides.
  • a G-clamp nucleotide is a modified cytosine analog wherein modifications result in the ability to hydrogen bond both Watson-Crick and Hoogsteen faces of a complementary guanine within a duplex, see for example Lin and Matteucci, 1998 , J. Am. Chem. Soc., 120, 8531-8532.
  • a single G-clamp analog substation within an oligonucleotide can result in substantially enhanced helical thermal stability and mismatch discrimination when hybridized to complementary oligonucleotides.
  • the inclusion of such nucleotides in nucleic acid molecules of the invention can enable both enhanced affinity and specificity to nucleic acid targets.
  • nucleic acid molecules e.g., enzymatic nucleic acid molecules and antisense nucleic acid molecules
  • Delivery exogenously are optimally stable within cells until translation of the target RNA has been inhibited long enough to reduce the levels of the undesirable protein. This period of time varies between hours to days depending upon the disease state.
  • These nucleic acid molecules should be resistant to nucleases in order to function as effective intracellular therapeutic agents. Improvements in the chemical synthesis of nucleic acid molecules described in the instant invention and in the art have expanded the ability to modify nucleic acid molecules by introducing nucleotide modifications to enhance their nuclease stability as described above.
  • the invention features conjugates and/or complexes of nucleic acid molecules targeting PTGDS, PTGDR, and/or adenosine receptors.
  • Compositions and conjugates are used to facilitate delivery of molecules into a biological system, such as cells.
  • the conjugates provided by the instant invention can impart therapeutic activity by transferring therapeutic compounds across cellular membranes, altering the pharmacokinetics, and/or modulating the localization of nucleic acid molecules of the invention.
  • the present invention encompasses the design and synthesis of novel agents for the delivery of molecules, including but not limited to small molecules, lipids, phospholipids, nucleosides, nucleotides, nucleic acids, antibodies, toxins, negatively charged polymers and other polymers, for example proteins, peptides, hormones, carbohydrates, polyethylene glycols, or polyamines, across cellular membranes.
  • the transporters described are designed to be used either individually or as part of a multi-component system, with or without degradable linkers. These compounds are expected to improve delivery and/or localization of nucleic acid molecules of the invention into a number of cell types originating from different tissues, in the presence or absence of serum (see Sullenger and Cech, U.S. Pat. No. 5,854,038).
  • Conjugates of the molecules described herein can be attached to biologically active molecules via linkers that are biodegradable, such as biodegradable nucleic acid linker molecules.
  • biodegradable nucleic acid linker molecule refers to a nucleic acid molecule that is designed as a biodegradable linker to connect one molecule to another molecule, for example, a biologically active molecule.
  • the stability of the biodegradable nucleic acid linker molecule can be modulated by using various combinations of ribonucleotides, deoxyribonucleotides, and chemically modified nucleotides, for example 2′-O-methyl, 2′-fluoro, 2′-amino, 2′-O-amino, 2′-C-allyl, 2′-O-allyl, and other 2′-modified or base modified nucleotides.
  • the biodegradable nucleic acid linker molecule can be a dimer, trimer, tetramer or longer nucleic acid molecule, for example an oligonucleotide of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides in length, or can comprise a single nucleotide with a phosphorus based linkage, for example a phosphoramidate or phosphodiester linkage.
  • the biodegradable nucleic acid linker molecule can also comprise nucleic acid backbone, nucleic acid sugar, or nucleic acid base modifications.
  • biodegradable refers to degradation in a biological system, for example enzymatic degradation or chemical degradation.
  • biologically active molecule refers to compounds or molecules that are capable of eliciting or modifying a biological response in a system.
  • biologically active molecules contemplated by the instant invention include therapeutically active molecules such as antibodies, hormones, antivirals, peptides, proteins, chemotherapeutics, small molecules, vitamins, co-factors, nucleosides, nucleotides, oligonucleotides, enzymatic nucleic acids, antisense nucleic acids, triplex forming oligonucleotides, 2,5-A chimeras, siRNA, dsRNA, allozymes, aptamers, decoys and analogs thereof.
  • Biologically active molecules of the invention also include molecules capable of modulating the pharmacokinetics and/or pharmacodynamics of other biologically active molecules, for example lipids and polymers such as polyamines, polyamides, polyethylene glycol and other polyethers.
  • lipids and polymers such as polyamines, polyamides, polyethylene glycol and other polyethers.
  • phospholipid refers to a hydrophobic molecule comprising at least one phosphorus group.
  • a phospholipid can comprise a phosphorus containing group and saturated or unsaturated alkyl group, optionally substituted with OH, COOH, oxo, amine, or substituted or unsubstituted aryl groups.
  • nucleic acid catalysts having chemical modifications that maintain or enhance enzymatic activity are provided.
  • Such nucleic acids are also generally more resistant to nucleases than unmodified nucleic acid.
  • the activity of the nucleic acid may not be significantly lowered.
  • enzymatic nucleic acids are useful in a cell and/or in vivo even if activity over all is reduced 10 fold (Burgin et al., 1996 , Biochemistry, 35, 14090).
  • Such enzymatic nucleic acids herein are said to “maintain” the enzymatic activity of an all RNA ribozyme or all DNA DNAzyme.
  • nucleic acid molecules comprise a 5′ and/or a 3′-cap structure.
  • cap structure is meant chemical modifications, which have been incorporated at either terminus of the oligonucleotide (see for example Wincott et al., WO 97/26270, incorporated by reference herein). These terminal modifications protect the nucleic acid molecule from exonuclease degradation, and can help in delivery and/or localization within a cell.
  • the cap can be present at the 5′-terminus (5′-cap) or at the 3′-terminus (3′-cap) or can be present on both terminus.
  • the 5′-cap includes inverted abasic residue (moiety), 4′,5′-methylene nucleotide; 1-(beta-D-erythrofuranosyl) nucleotide, 4′-thio nucleotide, carbocyclic nucleotide; 1,5-anhydrohexitol nucleotide; L-nucleotides; alpha-nucleotides; modified base nucleotide; phosphorodithioate linkage; threo-pentofuranosyl nucleotide; acyclic 3′,4′-seco nucleotide; acyclic 3,4-dihydroxybutyl nucleotide; acyclic 3,5-dihydroxypentyl nucleotide, 3′-3′-inverted nucleotide moiety; 3′-3′-inverted abasic moiety; 3′-2′-inverted nucleotide moiety; 3′-2′-inverted nu
  • the 3′-cap includes, for example 4′,5′-methylene nucleotide; 1-(beta-D-erythrofuranosyl) nucleotide; 4′-thio nucleotide, carbocyclic nucleotide; 5′-amino-alkyl phosphate; 1,3-diamino-2-propyl phosphate, 3-aminopropyl phosphate; 6-aminohexyl phosphate; 1,2-aminododecyl phosphate; hydroxypropyl phosphate; 1,5-anhydrohexitol nucleotide; L-nucleotide; alpha-nucleotide; modified base nucleotide; phosphorodithioate; threo-pentofuranosyl nucleotide; acyclic 3′,4′-seco nucleotide; 3,4-dihydroxybutyl nucleotide; 3,5-
  • non-nucleotide any group or compound thatcan be incorporated into a nucleic acid chain in the place of one or more nucleotide units, including either sugar and/or phosphate substitutions, and allows the remaining bases to exhibit their enzymatic activity.
  • the group or compound is abasic in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine.
  • alkyl refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups.
  • the alkyl group can have, for example, 1 to 12 carbons.
  • the alkyl group is a lower alkyl of from 1 to 7 carbons.
  • the alkyl group is 1 to 4 carbons.
  • the alkyl group can be substituted or unsubstituted. When substituted the substituted group(s) can be hydroxyl, cyano, alkoxy, ⁇ O, ⁇ S, NO 2 or N(CH 3 ) 2 , amino, or SH.
  • alkenyl groups which are unsaturated hydrocarbon groups containing at least one carbon-carbon double bond, including straight-chain, branched-chain, and cyclic groups.
  • the alkenyl group can have, for example, 1 to 12 carbons.
  • the alkenyl group can be a lower alkenyl of from 1 to 7 carbons.
  • the alkenyl group can be 1 to 4 carbons.
  • the alkenyl group can be substituted or unsubstituted. When substituted the substituted group(s) can be, for example, hydroxyl, cyano, alkoxy, ⁇ O, ⁇ S, NO 2 , halogen, N(CH 3 ) 2 , amino, or SH.
  • alkyl also includes alkynyl groups which have an unsaturated hydrocarbon group containing at least one carbon-carbon triple bond, including straight-chain, branched-chain, and cyclic groups.
  • the alkynyl group can have, for example, 1 to 12 carbons.
  • the alkynyl group is a lower alkynyl of from 1 to 7 carbons.
  • the alkynyl group is 1 to 4 carbons.
  • the alkynyl group can be substituted or unsubstituted. When substituted the substituted group(s) can be, for example, hydroxyl, cyano, alkoxy, ⁇ O, ⁇ S, NO 2 or N(CH 3 ) 2 , amino or SH.
  • Such alkyl groups can also include aryl, alkylaryl, carbocyclic aryl, heterocyclic aryl, amide and ester groups.
  • An “aryl” group refers to an aromatic group which has at least one ring having a conjugated p electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which can be optionally substituted.
  • the preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, alkoxy, alkyl, alkenyl, alkynyl, and amino groups.
  • alkylaryl refers to an alkyl group (as described above) covalently joined to an aryl group (as described above).
  • Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted.
  • Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms.
  • Suitable heteroatoms include oxygen, sulfur, and nitrogen, and include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted.
  • An “amide” refers to an —C(O)—NH—R, where R is either alkyl, aryl, alkylaryl or hydrogen.
  • An “ester” refers to an —C(O)—OR′, where R is either alkyl, aryl, alkylaryl or hydrogen.
  • nucleotide is meant a heterocyclic nitrogenous base in N-glycosidic linkage with a phosphorylated sugar.
  • Nucleotides are recognized in the art to include natural bases (standard), and modified bases well known in the art. Such bases are generally located at the 1′ position of a nucleotide sugar moiety. Nucleotides generally comprise a base, sugar and a phosphate group.
  • the nucleotides can be unmodified or modified at the sugar, phosphate and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and other; see for example, Usman and McSwiggen, supra; Eckstein et al., International PCT Publication No. WO 92/07065; Usman et al., International PCT Publication No. WO 93/15187; Uhlman & Peyman, supra all are hereby incorporated by reference herein).
  • modified nucleic acid bases known in the art as summarized by Limbach et al., 1994, Nucleic Acids Res. 22, 2183.
  • nucleic acids include, for example, inosine, purine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2, 4, 6-trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidines (e.g., 5-methylcytidine), 5-alkyluridines (e.g., ribothymidine), 5-halouridine (e.g., 5-bromouridine) or 6-azapyrimidines or 6-alkylpyrimidines (e.g.
  • modified bases in this aspect is meant nucleotide bases other than adenine, guanine, cytosine and uracil at 1′ position or their equivalents; such bases can be used at any position, for example, within the catalytic core of an enzymatic nucleic acid molecule and/or in the substrate-binding regions of the nucleic acid molecule.
  • nucleoside is meant a heterocyclic nitrogenous base in N-glycosidic linkage with a sugar.
  • Nucleosides are recognized in the art to include natural bases (standard), and modified bases well known in the art. Such bases are generally located at the 1′ position of a nucleoside sugar moiety. Nucleosides generally comprise a base and sugar group.
  • the nucleosides can be unmodified or modified at the sugar, and/or base moiety, (also referred to interchangeably as nucleoside analogs, modified nucleosides, non-natural nucleosides, non-standard nucleosides and other; see for example, Usman and McSwiggen, supra; Eckstein et al., International PCT Publication No. WO 92/07065; Usman et al., International PCT Publication No. WO 93/15187; Uhlman & Peyman, supra all are hereby incorporated by reference herein).
  • modified nucleic acid bases known in the art as summarized by Limbach et al., 1994, Nucleic Acids Res. 22, 2183.
  • nucleic acids Some of the non-limiting examples of chemically modified and other natural nucleic acid bases that can be introduced into nucleic acids include, inosine, purine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2, 4, 6-trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidines (e.g., 5-methylcytidine), 5-alkyluridines (e.g., ribothymidine), 5-halouridine (e.g., 5-bromouridine) or 6-azapyrimidines or 6-alkylpyrimidines (e.g.
  • modified bases in this aspect is meant nucleoside bases other than adenine, guanine, cytosine and uracil at 1′ position or their equivalents; such bases can be used at any position, for example, within the catalytic core of an enzymatic nucleic acid molecule and/or in the substrate-binding regions of the nucleic acid molecule.
  • the invention features modified enzymatic nucleic acid molecules with phosphate backbone modifications comprising one or more phosphorothioate, phosphorodithioate, methylphosphonate, morpholino, amidate carbamate, carboxymethyl, acetamidate, polyamide, sulfonate, sulfonamide, sulfamate, formacetal, thioformacetal, and/or alkylsilyl, substitutions.
  • abasic sugar moieties lacking a base or having other chemical groups in place of a base at the 1′ position, for example a 3′,3′-linked or 5′,5′-linked deoxyabasic ribose derivative (for more details see Wincott et al., International PCT publication No. WO 97/26270).
  • unmodified nucleoside is meant one of the bases adenine, cytosine, guanine, thymine, uracil joined to the 1′ carbon of ⁇ -D-ribo-furanose.
  • modified nucleoside is meant any nucleotide base that contains a modification in the chemical structure of an unmodified nucleotide base, sugar and/or phosphate.
  • amino is meant 2′-NH 2 or 2′-O-NH 2 , which can be modified or unmodified.
  • modified groups are described, for example, in Eckstein et al., U.S. Pat. No. 5,672,695 and Matulic-Adamic et al., WO 98/28317, respectively, which are both incorporated by reference in their entireties.
  • nucleic acid e.g., antisense and ribozyme
  • modifications to nucleic acid can be made to enhance the utility of these molecules.
  • modifications can enhance shelf-life, half-life in vitro, stability, and ease of introduction of such oligonucleotides to the target site, including e.g., enhancing penetration of cellular membranes and conferring the ability to recognize and bind to targeted cells.
  • nucleic acid-based molecules of the invention can lead to better treatment of the disease progression by affording the possibility of combination therapies (e.g., multiple enzymatic nucleic acid molecules targeted to different genes, enzymatic nucleic acid molecules coupled with known small molecule inhibitors, or intermittent treatment with combinations of enzymatic nucleic acid molecules (including different enzymatic nucleic acid molecule motifs) and/or other chemical or biological molecules).
  • combination therapies e.g., multiple enzymatic nucleic acid molecules targeted to different genes, enzymatic nucleic acid molecules coupled with known small molecule inhibitors, or intermittent treatment with combinations of enzymatic nucleic acid molecules (including different enzymatic nucleic acid molecule motifs) and/or other chemical or biological molecules).
  • the treatment of patients with nucleic acid molecules can also include combinations of different types of nucleic acid molecules.
  • Therapies can be devised which include a mixture of enzymatic nucleic acid molecules (including different enzymatic nucleic acid molecule motifs), antisense and/or 2-5A chimera molecules to one or more targets to alleviate symptoms of a disease.
  • a nucleid acid molecule of the invention can be adapted for use to treat asthma and other related diseases and conditions described herein.
  • a nucleic acid molecule can comprise a delivery vehicle, including liposomes, for administration to a subject, carriers and diluents and their salts, and/or can be present in pharmaceutically acceptable formulations.
  • Methods for the delivery of nucleic acid molecules are described in Akhtar et al., 1992 , Trends Cell Bio., 2, 139; and Delivery Strategies for Antisense Oligonucleotide Therapeutics , ed. Akhtar, 1995 which are both incorporated herein by reference.
  • Nucleic acid molecules can be administered to cells by a variety of methods known to those familiar to the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres.
  • the nucleic acid molecules or the invention are administered via pulmonary delivery, such as by inhalation of an aerosol or spray dried formulation administered by an inhalation device or nebulizer.
  • the nucleic acid/vehicle combination is locally delivered by direct injection or by use of an infusion pump.
  • routes of delivery include, but are not limited to oral (tablet or pill form) and/or intrathecal delivery (Gold, 1997 , Neuroscience, 76, 1153-1158).
  • Other approaches include the use of various transport and carrier systems, for example though the use of conjugates and biodegradable polymers.
  • nucleic acid delivery and administration are provided in Sullivan et al., supra, Draper et al., PCT WO93/23569, Beigelman et al., PCT WO99/05094, and Klimuk et al., PCT WO99/04819 all of which have been incorporated by reference herein.
  • the molecules of the instant invention can be used as pharmaceutical agents.
  • Pharmaceutical agents prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, or all of the symptoms) of a disease state in a patient.
  • the negatively charged polynucleotides of the invention can be administered (e.g., RNA, DNA or protein) and introduced into a patient by any standard means, with or without stabilizers, buffers, and the like, to form a pharmaceutical composition.
  • RNA, DNA or protein e.g., RNA, DNA or protein
  • standard protocols for formation of liposomes can be followed.
  • the compositions of the present invention can also be formulated and used as tablets, capsules or elixirs for oral administration; suppositories for rectal administration; sterile solutions; suspensions for injectable administration; and the other compositions known in the art.
  • the present invention also includes pharmaceutically acceptable formulations of the compounds described.
  • formulations include salts of the above compounds, e.g., acid addition salts, for example, salts of hydrochloric, hydrobromic, acetic acid, and benzene sulfonic acid.
  • a pharmacological composition or formulation refers to a composition or formulation in a form suitable for administration, e.g., local administration or systemic administration, into a cell or patient, including, for example, a human. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, or by injection. Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged polymer is desired to be delivered to). For example, pharmacological compositions injected into the blood stream should be soluble. Other factors are known in the art, and include considerations such as toxicity and forms which prevent the composition or formulation from exerting its effect.
  • local administration in vivo local absorption or accumulation of drugs in the specific tissue, organ, or compartment of the body.
  • Administration routes that can lead to local absorption include, without limitations: inhalation, direct injection, or dermatological applications.
  • systemic administration in vivo systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body.
  • Administration routes which lead to systemic absorption include, without limitations: intravenous, subcutaneous, intraperitoneal, inhalation, oral, intrapulmonary and intramuscular.
  • Each of these administration routes expose the desired compound, e.g., nucleic acids, to an accessible diseased tissue.
  • the rate of entry of a drug into the circulation has been shown to be a function of molecular weight or size.
  • a liposome or other drug carrier comprising the compounds of the instant invention, for example PEG or phospholipids conjugates
  • PEG or phospholipids conjugates can potentially localize the drug, for example, in certain tissue types, such as the tissues of the reticular endothelial system (RES).
  • RES reticular endothelial system
  • a nucleic acid formulation that can facilitate the association of drug with the surface of cells, such as, lymphocytes and macrophages is also useful. This approach can provide enhanced delivery of the drug to target cells by taking advantage of the specificity of macrophage and lymphocyte immune recognition of abnormal cells.
  • nucleic acid molecules of the invention are administered to a patient with an inhaler or nebulizer, providing rapid local uptake of the nucleic acid molecules into relevant pulmonary tissues.
  • nucleic acid molecule or formulation comprising the nucleic acid molecule is administered to a patient systemically, for example by intravenous or subcutaneous injection, providing sustained uptake of the nucleic acid molecules into relevant bodily tissues.
  • compositions or formulation that allows for the effective distribution of the nucleic acid molecules of the instant invention in the physical location most suitable for their desired activity.
  • agents suitable for formulation with the nucleic acid molecules of the instant invention include: PEG conjugated nucleic acids, phospholipid conjugated nucleic acids, nucleic acids containing lipophilic moieties, phosphorothioates, P-glycoprotein inhibitors (such as Pluronic P85) which can enhance entry of drugs into various tissues, for exaple the CNS (Jolliet-Riant and Tillement, 1999 , Fundam. Clin.
  • biodegradable polymers such as poly (DL-lactide-coglycolide) microspheres for sustained release delivery after implantation (Emerich, DF et al, 1999 , Cell Transplant, 8, 47-58) Alkermes, Inc. Cambridge, Mass.; and loaded nanoparticles, such as those made of polybutylcyanoacrylate, which can deliver drugs across the blood brain barrier and can alter neuronal uptake mechanisms ( Prog Neuropsychopharmacol Biol Psychiatry, 23, 941-949, 1999).
  • Other non-limiting examples of delivery strategies, including CNS delivery of the nucleic acid molecules of the instant invention include material described in Boado et al., 1998 , J. Pharm.
  • the invention also features the use of the composition comprising surface-modified liposomes containing poly (ethylene glycol) lipids (PEG-modified, or long-circulating liposomes or stealth liposomes).
  • Nucleic acid molecules of the invention can also comprise covalently attached PEG molecules of various molecular weights. These formulations offer a method for increasing the accumulation of drugs in target tissues. This class of drug carriers resists opsonization and elimination by the mononuclear phagocytic system (MPS or RES), thereby enabling longer blood circulation times and enhanced tissue exposure for the encapsulated drug (Lasic et al. Chem. Rev. 1995, 95, 2601-2627; Ishiwata et al., Chem.
  • WO 96/10391 Ansell et al., International PCT Publication No. WO 96/10390; Holland et al., International PCT Publication No. WO 96/10392; all of which are incorporated by reference herein).
  • Long-circulating liposomes are also likely to protect drugs from nuclease degradation to a greater extent compared to cationic liposomes, based on their ability to avoid accumulation in metabolically aggressive MPS tissues such as the liver and spleen. All of these references are incorporated by reference herein.
  • compositions prepared for storage or administration that include a pharmaceutically effective amount of the desired compounds in a pharmaceutically acceptable carrier or diluent.
  • Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences , Mack Publishing Co. (A. R. Gennaro edit. 1985) hereby incorporated by reference herein.
  • preservatives, stabilizers, dyes and flavoring agents can be provided. These include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid.
  • antioxidants and suspending agents can be used.
  • a pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, or all of the symptoms) of a disease state.
  • the pharmaceutically effective dose depends on the type of disease, the composition used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors which those skilled in the medical arts will recognize. Generally, an amount between 0.1 mg/kg and 100 mg/kg body weight/day of active ingredients is administered dependent upon potency of the negatively charged polymer.
  • nucleic acid molecules of the invention and formulations thereof can be administered orally, topically, parenterally, by inhalation or spray or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles.
  • parenteral as used herein includes percutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, or intrathecal injection or infusion techniques and the like.
  • a pharmaceutical formulation comprising a nucleic acid molecule of the invention and a pharmaceutically acceptable carrier.
  • One or more nucleic acid molecules of the invention can be present in association with one or more non-toxic pharmaceutically acceptable carriers and/or diluents and/or adjuvants, and if desired other active ingredients.
  • compositions containing nucleic acid molecules of the invention can be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets.
  • excipients can be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets can be uncoated or they can be coated by known techniques. In some cases such coatings can be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monosterate or glyceryl distearate can be employed.
  • Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin or olive oil.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents can be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan mono
  • the aqueous suspensions can also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavoring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • sweetening agents such as sucrose or saccharin.
  • Oily suspensions can be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions can contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents and flavoring agents can be added to provide palatable oral preparations. These compositions can be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent for example sweetening, flavoring and coloring agents, can also be present.
  • compositions of the invention can also be in the form of oil-in-water emulsions.
  • the oily phase can be a vegetable oil or a mineral oil or mixtures of these.
  • Suitable emulsifying agents can be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions can also contain sweetening and flavoring agents.
  • Syrups and elixirs can be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations can also contain a demulcent, a preservative and flavoring and coloring agents.
  • the pharmaceutical compositions can be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above.
  • the sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • Suitable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono-or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the nucleic acid molecules of the invention can also be administered in the form of suppositories, e.g., for rectal administration of the drug.
  • suppositories e.g., for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials include cocoa butter and polyethylene glycols.
  • Nucleic acid molecules of the invention can be administered parenterally in a sterile medium.
  • the drug depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle.
  • adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle.
  • Dosage levels of the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions (about 0.5 mg to about 7 g per patient per day).
  • the amount of active ingredient that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration.
  • Dosage unit forms generally contain between from about 1 mg to about 500 mg of an active ingredient.
  • the specific dose level for any particular patient depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy.
  • the composition can also be added to the animal feed or drinking water. It can be convenient to formulate the animal feed and drinking water compositions so that the animal takes in a therapeutically appropriate quantity of the composition along with its diet. It can also be convenient to present the composition as a premix for addition to the feed or drinking water.
  • nucleic acid molecules of the present invention can also be administered to a patient in combination with other therapeutic compounds to increase the overall therapeutic effect.
  • the use of multiple compounds to treat an indication can increase the beneficial effects while reducing the presence of side effects.
  • nucleic acid molecules of the instant invention can be expressed within cells from eukaryotic promoters (e.g., Izant and Weintraub, 1985 , Science, 229, 345; McGarry and Lindquist, 1986 , Proc. Natl. Acad. Sci., USA 83, 399; Scanlon et al., 1991 , Proc. Natl. Acad. Sci. USA, 88, 10591-5; Kashani-Sabet et al., 1992 , Antisense Res. Dev., 2, 3-15; Dropulic et al., 1992 , J.
  • eukaryotic promoters e.g., Izant and Weintraub, 1985 , Science, 229, 345; McGarry and Lindquist, 1986 , Proc. Natl. Acad. Sci., USA 83, 399; Scanlon et al., 1991 , Proc. Natl. Acad. Sci. USA
  • nucleic acid can be expressed in eukaryotic cells from the appropriate DNA/RNA vector.
  • the activity of such nucleic acids can be augmented by their release from the primary transcript by a enzymatic nucleic acid (Draper et al, PCT WO 93/23569, and Sullivan et al., PCT WO 94/02595; Ohkawa et al., 1992 , Nucleic Acids Symp.
  • RNA molecules of the present invention can be expressed from transcription units (see for example Couture et al., 1996 , TIG., 12, 510) inserted into DNA or RNA vectors.
  • the recombinant vectors can be DNA plasmids or viral vectors. Ribozyme expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus, retrovirus, adenovirus, or alphavirus.
  • the recombinant vectors capable of expressing the nucleic acid molecules can be delivered as described above, and persist in target cells. Alternatively, viral vectors can be used that provide for transient expression of nucleic acid molecules. Such vectors can be repeatedly administered as necessary.
  • Delivery of nucleic acid molecule expressing vectors can be systemic, such as by intravenous or intra-muscular administration, by administration to target cells ex-planted from the patient followed by reintroduction into the patient, or by any other means that would allow for introduction into the desired target cell (for a review see Couture et al., 1996 , TIG., 12, 510).
  • the invention features an expression vector comprising a nucleic acid sequence encoding at least one of the nucleic acid molecules of the instant invention is disclosed.
  • the nucleic acid sequence encoding the nucleic acid molecule of the instant invention is operable linked in a manner that allows expression of that nucleic acid molecule.
  • the invention features an expression vector comprising: a) a transcription initiation region (e.g., eukaryotic pol I, II or III initiation region); b) a transcription termination region (e.g., eukaryotic pol I, II or III termination region); c) a nucleic acid sequence encoding at least one of the nucleic acid catalyst of the instant invention; and wherein said sequence is operably linked to said initiation region and said termination region, in a manner that allows expression and/or delivery of said nucleic acid molecule.
  • the vector can optionally include an open reading frame (ORF) for a protein operably linked on the 5′ side or the 3′-side of the sequence encoding the nucleic acid catalyst of the invention; and/or an intron (intervening sequences).
  • ORF open reading frame
  • RNA polymerase I RNA polymerase I
  • RNA polymerase II RNA polymerase II
  • RNA polymerase III RNA polymerase III
  • Transcripts from pol II or pol III promoters are expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type depends on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby.
  • Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Elroy-Stein and Moss, 1990 , Proc. Natl. Acad. Sci.
  • nucleic acid molecules such as ribozymes expressed from such promoters can function in mammalian cells (e.g. Kashani-Sabet et al., 1992 , Antisense Res. Dev., 2, 3-15; Ojwang et al., 1992 , Proc. Natl. Acad. Sci.
  • transcription units such as the ones derived from genes encoding U6 small nuclear (snRNA), transfer RNA (tRNA) and adenovirus VA RNA are useful in generating high concentrations of desired RNA molecules such as ribozymes in cells (Thompson et al., supra; Couture and Stinchcomb, 1996, supra; Noonberg et al., 1994 , Nucleic Acid Res., 22, 2830; Noonberg et al., U.S. Pat. No. 5,624,803; Good et al., 1997 , Gene Ther., 4, 45; Beigelman et al., International PCT Publication No. WO 96/18736; all of these publications are incorporated by reference herein).
  • ribozyme transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated virus vectors), or viral RNA vectors (such as retroviral or alphavirus vectors) (for a review see Couture and Stinchcomb, 1996, supra).
  • plasmid DNA vectors such as adenovirus or adeno-associated virus vectors
  • viral RNA vectors such as retroviral or alphavirus vectors
  • the invention features an expression vector comprising a nucleic acid sequence encoding at least one of the nucleic acid molecules of the invention, in a manner that allows expression of that nucleic acid molecule.
  • the expression vector comprises in one embodiment; a) a transcription initiation region; b) a transcription termination region; c) a nucleic acid sequence encoding at least one said nucleic acid molecule; and wherein said sequence is operably linked to said initiation region and said termination region, in a manner that allows expression and/or delivery of said nucleic acid molecule.
  • the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an open reading frame; d) a nucleic acid sequence encoding at least one said nucleic acid molecule, wherein said sequence is operably linked to the 3′-end of said open reading frame; and wherein said sequence is operably linked to said initiation region, said open reading frame and said termination region, in a manner that allows expression and/or delivery of said nucleic acid molecule.
  • the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an intron; d) a nucleic acid sequence encoding at least one said nucleic acid molecule; and wherein said sequence is operably linked to said initiation region, said intron and said termination region, in a manner which allows expression and/or delivery of said nucleic acid molecule.
  • the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an intron; d) an open reading frame; e) a nucleic acid sequence encoding at least one said nucleic acid molecule, wherein said sequence is operably linked to the 3′-end of said open reading frame; and wherein said sequence is operably linked to said initiation region, said intron, said open reading frame and said termination region, in a manner which allows expression and/or delivery of said nucleic acid molecule.
  • Enzymatic nucleic acid molecule target sites are chosen by analyzing sequences of Human PTGDS (Genbank accession No: NM 000954), ADORA1 (Genbank accession No: NM — 000674) and PTGDR gene (Genbank accession Nos: U31332 and U31099) and prioritizing the sites on the basis of folding. Enzymatic nucleic acid molecules are designed that can bind each target and are individually analyzed by computer folding (Christoffersen et al., 1994 J. Mol. Struc. Theochem, 311, 273; Jaeger et al., 1989 , Proc. Natl. Acad. Sci.
  • binding arm lengths can be chosen to optimize activity. Generally, at least 4 bases on each arm are able to bind to, or otherwise interact with, the target RNA.
  • Enzymatic nucleic acid molecules and antisense constructs are designed to anneal to various sites in the RNA message.
  • the binding arms of the enzymatic nucleic acid molecules are complementary to the target site sequences described above, while the antisense constructs are fully complementary to the target site sequences described above.
  • the enzymatic nucleic acid molecules and antisense constructs were chemically synthesized. The method of synthesis used followed the procedure for normal RNA synthesis as described above and in Usman et al., (1987 J. Am. Chem.
  • Enzymatic nucleic acid molecules and antisense constructs are also synthesized from DNA templates using bacteriophage T7 RNA polymerase (Milligan and Uhlenbeck, 1989, Methods Enzymol. 180, 51). Enzymatic nucleic acid molecules and antisense constructs are purified by gel electrophoresis using general methods or are purified by high pressure liquid chromatography (HPLC; See Wincott et al., supra; the totality of which is hereby incorporated herein by reference) and are resuspended in water. The sequences of the chemically synthesized enzymatic nucleic acid molecules used in this study are shown below in Table III-VII. The sequences of the chemically synthesized antisense constructs used in this study are complementary sequences to the Substrate sequences shown below as in Table III-VII.
  • Enzymatic nucleic acid molecules targeted to the human PTGDS, ADORA1 and PTGDR RNA are designed and synthesized as described above. These enzymatic nucleic acid molecules can be tested for cleavage activity in vitro, for example, using the following procedure.
  • the target sequences and the nucleotide location within the PTGDR RNA are given in Tables III-VII.
  • Cleavage Reactions Full-length or partially full-length, internally-labeled target RNA for enzymatic nucleic acid molecule cleavage assay is prepared by in vitro transcription in the presence of [a- 32 P] CTP, passed over a G 50 Sephadex column by spin chromatography and used as substrate RNA without further purification. Alternately, substrates are 5′- 32 P-end labeled using T4 polynucleotide kinase enzyme.
  • Assays are performed by pre-warming a 2 ⁇ concentration of purified enzymatic nucleic acid molecule in enzymatic nucleic acid molecule cleavage buffer (50 mM Tris-HCl, pH 7.5 at 37° C., 10 mM MgCl 2 ) and the cleavage reaction was initiated by adding the 2 ⁇ enzymatic nucleic acid molecule mix to an equal volume of substrate RNA (maximum of 1-5 nM) that was also pre-warmed in cleavage buffer. As an initial screen, assays are carried out for 1 hour at 37° C.
  • enzymatic nucleic acid molecule cleavage buffer 50 mM Tris-HCl, pH 7.5 at 37° C., 10 mM MgCl 2
  • enzymatic nucleic acid molecule excess a final concentration of either 40 nM or 1 mM enzymatic nucleic acid molecule, i.e., enzymatic nucleic acid molecule excess.
  • the reaction is quenched by the addition of an equal volume of 95% formamide, 20 mM EDTA, 0.05% bromophenol blue and 0.05% xylene cyanol after which the sample is heated to 95° C. for 2 minutes, quick chilled and loaded onto a denaturing polyacrylamide gel.
  • Substrate RNA and the specific RNA cleavage products generated by enzymatic nucleic acid molecule cleavage are visualized on an autoradiograph of the gel. The percentage of cleavage is determined by Phosphor Imager® quantitation of bands representing the intact substrate and the cleavage products.
  • mice that are treated with nucleic acid molecules of the invention were shown to be greatly reduced compared to wild type mice.
  • the PTGDR ⁇ / ⁇ mice showed only marginal eosinophil infiltration and failed to develop airway hyperreactivity.
  • this model can be used to evaluate mice that are treated with nucleic acid molecules of the invention and can furthermore be used as a positive control in determining the response of mice treated with nucleic acid molecules of the invention by using such factors as airway obstruction, lung capacity, and bronchiolar alveolar lavage (BAL) fluid in the evaluation.
  • BAL bronchiolar alveolar lavage
  • PTGDR Two human cell lines, NPE cells and NCB-20 cells are known to express PTGDR. Cloned human PTGDR has been expressed in CHO and COS7 cells and used in various studies. These PTGDR expressing lung cell lines can be used in cell culture assays to evaluate nucleic acid molecules of the invention. A primary endpoint in these experiments would be the RT-PCR analysis of PTGDR mRNA expression in PTGDR expressing cells. In addition, ligand binding assays can be developed where binding of PTGDS can be evaluated in response to treatment with nucleic acid molecules of the invention.
  • nucleic acid molecules of the present invention can be used in assays to diagnose disease state related of PTGDS, ADORA1 and/or PTGDR levels.
  • nucleic acid molecules can be used to treat disease state related to PTGDS, ADORA1 and/or PTGDR levels.
  • Particular degenerative and disease states that can be associated with PTGDS, ADORA1 and PTGDR levels include, but are not limited to allergic diseases and conditions, including but not limited to asthma, allergic rhinitis, atopic dermatitis, and any other diseases or conditions that are related to or will respond to the levels of PTGDS, ADORA1 and/or PTGDR in a cell or tissue, alone or in combination with other therapies.
  • the nucleic acid molecules of this invention can be used as diagnostic tools to examine genetic drift and mutations within diseased cells or to detect the presence of PTGDS, ADORA1 and/or PTGDR RNA in a cell.
  • the close relationship between enzymatic nucleic acid molecule activity and the structure of the target RNA allows the detection of mutations in any region of the molecule that alters the base-pairing and three-dimensional structure of the target RNA.
  • Cleavage of target RNAs with enzymatic nucleic acid molecules can be used to inhibit gene expression and define the role (essentially) of specified gene products in the progression of disease. In this manner, other genetic targets can be defined as important mediators of the disease.
  • combinational therapies e.g., multiple enzymatic nucleic acid molecules targeted to different genes, enzymatic nucleic acid molecules coupled with known small molecule inhibitors, or intermittent treatment with combinations of enzymatic nucleic acid molecules and/or other chemical or biological molecules.
  • enzymatic nucleic acid molecules of this invention include detection of the presence of mRNAs associated with PTGDS, ADORA1 or PTGDR-related condition. Such RNA is detected by determining the presence of a cleavage product after treatment with an enzymatic nucleic acid molecule using standard methodology.
  • enzymatic nucleic acid molecules which cleave only wild-type or mutant forms of the target RNA are used for the assay.
  • the first enzymatic nucleic acid molecule is used to identify wild-type RNA present in the sample and the second enzymatic nucleic acid molecule is used to identify mutant RNA in the sample.
  • synthetic substrates of both wild-type and mutant RNA are cleaved by both enzymatic nucleic acid molecules to demonstrate the relative enzymatic nucleic acid molecule efficiencies in the reactions and the absence of cleavage of the “non-targeted” RNA species.
  • the cleavage products from the synthetic substrates also serve to generate size markers for the analysis of wild-type and mutant RNAs in the sample population.
  • each analysis requires two enzymatic nucleic acid molecules, two substrates and one unknown sample which is combined into six reactions.
  • the presence of cleavage products is determined using an RNAse protection assay so that full-length and cleavage fragments of each RNA can be analyzed in one lane of a polyacrylamide gel. It is not absolutely required to quantify the results to gain insight into the expression of mutant RNAs and putative risk of the desired phenotypic changes in target cells.
  • RNA whose protein product is implicated in the development of the phenotype i.e., PTGDS/PTGDR
  • PTGDS/PTGDR protein product that is implicated in the development of the phenotype
  • a qualitative comparison of RNA levels will be adequate and will decrease the cost of the initial diagnosis.
  • Higher mutant form to wild-type ratios are correlated with higher risk whether RNA levels are compared qualitatively or quantitatively.
  • the use of enzymatic nucleic acid molecules in diagnostic applications contemplated by the instant invention is described, for example, in George et al., U.S. Pat. Nos. 5,834,186 and 5,741,679, Shih et al., U.S. Pat. No.
  • sequence-specific enzymatic nucleic acid molecules of the instant invention can have many of the same applications for the study of RNA that DNA restriction endonucleases have for the study of DNA (Nathans et al., 1975 Ann. Rev. Biochem. 44:273).
  • the pattern of restriction fragments can be used to establish sequence relationships between two related RNAs, and large RNAs can be specifically cleaved to fragments of a size more useful for study.
  • the ability to engineer sequence specificity of the enzymatic nucleic acid molecule is ideal for cleavage of RNAs of unknown sequence.
  • Applicant has described the use of nucleic acid molecules to down-regulate gene expression of target genes in bacterial, microbial, fungal, viral, and eukaryotic systems including plant, or mammalian cells.
  • RNAse P RNA (M1 RNA) Size ⁇ 290 to 400 nucleotides. RNA portion of a ubiquitous ribonucleoprotein enzyme. Cleaves tRNA precursors to form mature tRNA [ xiii ].
  • RNAse P is found throughout the prokaryotes and eukaryotes. The RNA subunit has been sequenced from bacteria, yeast, rodents, and primates. Recruitment of endogenous RNAse P for therapeutic applications is possible through hybridization of an External Guide Sequence (EGS) to the target RNA [ xiv , xv ] Important phosphate and 2′OH contacts recently identified [ xvi , xvii ] Group II Introns Size: >1000 nucleotides. Trans cleavage of target RNAs recently demonstrated [ xviii , xix ]. Sequence requirements not fully determined.
  • EGS External Guide Sequence
  • Reaction mechanism 2′-OH of an internal adenosine generates cleavage products with 3′-OH and a “lariat” RNA containing a 3′-5′ and a 2′-5′ branch point. Only natural ribozyme with demonstrated participation in DNA cleavage [ xx , xxi ] in addition to RNA cleavage and ligation. Major structural features largely established through phylogenetic comparisons [ xxii ]. Important 2′OH contacts beginning to be identified [ xxiii ] Kinetic framework under development [ xxiv ] Neurospora VS RNA Size: ⁇ 144 nucleotides. Trans cleavage of hairpin target RNAs recently demonstrated [ xxv ]. Sequence requirements not fully determined.
  • Reaction mechanism attack by 2′-OH 5′to the scissile bond to generate cleavage products with 2′,3′-cyclic phosphate and 5′-OH ends. Binding sites and structural requirements not fully determined. Only 1 known member of this class. Found in Neurospora VS RNA. Hammerhead Ribozyme (see text for references) Size: ⁇ 13 to 40 nucleotides. Requires the target sequence UH immediately 5′ of the cleavage site. Binds a variable number nucleotides on both sides of the cleavage site. Reaction mechanism: attack by 2′-OH 5′ to the scissile bond to generate cleavage products with 2′,3′-cyclic phosphate and 5′-OH ends.
  • Reaction mechanism attack by 2′-OH 5′ to the scissile bond to generate cleavage products with 2′,3′-cyclic phosphate and 5′-OH ends.
  • 3 known members of this class. Found in three plant pathogen (satellite RNAs of the tobacco ringspot virus, arabis mosaic virus and chicory yellow mottle virus) which uses RNA as the infectious agent.
  • plant pathogen satellite RNAs of the tobacco ringspot virus, arabis mosaic virus and chicory yellow mottle virus
  • RNA infectious agent.
  • Essential structural features largely defined [ xxxi , xxxii , xxxiii , xxxiv ]
  • Ligation activity (in addition to cleavage activity) makes ribozyme amenable to engineering through in vitro selection [ xxxv ] Complete kinetic framework established for one ribozyme [ xxxvi ].
  • HDV Hepatitis Delta Virus
  • Ribozyme Size ⁇ 60 nucleotides. Trans cleavage of target RNAs demonstrated [ xxxix ]. Binding sites and structural requirements not fully determined, although no sequences 5′ of cleavage site are required. Folded ribozyme contains a pseudoknot structure [ xl ]. Reaction mechanism: attack by 2′-OH 5′ to the scissile bond to generate cleavage products with 2′,3′-cyclic phosphate and 5′-OH ends. Only 2 known members of this class. Found in human HDV. Circular form of HDV is active and shows increased nuclease stability [ xli ]

Abstract

The present invention relates to nucleic acid molecules, including antisense, enzymatic nucleic acid molecules, and RNA interference molecules, such as hammerhead ribozymes, DNAzymes, allozymes, siRNA, decoys and antisense, which modulate the expression of prostaglandin D2 (PTGDS), prostaglandin D2 receptor (PTGDR), and adenosine receptor genes.

Description

    PRIORITY
  • This application claims the benefit of U.S. Application Ser. No. 60/315,315, filed on Aug. 28, 2001, which is herein incorporated by reference in its entirity.[0001]
  • BACKGROUND OF THE INVENTION
  • The present invention relates to therapeutic compositions and methods for the treatment or diagnosis of diseases or conditions related to allergic response. Specifically, the invention provides compositions and methods for the treatment of diseases or conditions related to levels of factors involved in allergic conditions such as asthma, for example prostaglandin D2 receptor (PTGDR), prostaglandin D2 synthetase (PTGDS) and adenosine A1 receptor (ADORA1). The discussion is provided only for understanding of the invention that follows. This summary is not an admission that any of the work described below is prior art to the claimed invention. [0002]
  • Asthma is a chronic inflammatory disorder of the lungs characterized by airflow obstruction, bronchial hyper-responsiveness, and airway inflammation. T-lymphocytes that produce TH2 cytokines and cosinophilic leukocytes infiltrate the airways. In the airway and in bronchial alveolar lavage (BAL) fluid of individuals with asthma, high concentrations of TH2 cytokines, interleukin-4 (IL-4), IL-5, and IL-13, are present along with increased levels of adenosine. In contrast to normal individuals, asthmatics respond to adenosine challenge with marked airway obstruction. Upon allergen challenge, mast cells are activated by cross-linked IgE-allergen complexes. Large amounts of prostaglandin D2 (PGD2), the major cyclooxygenase product of arachidonic acid are released. PGD2 is generated from PGH2 via the activity of prostaglandin D2 synthetase (PTGDS). PGD2 receptors and adenosine A1 receptors are present in the lungs and airway along with various other tissues in response to allergic stimuli (Howarth, 1997[0003] , Allergy, 52, 12).
  • The significance of PGD2 as a mediator of allergic asthma has been established with the development of mice deficient in the PGD2 receptor (DP). DP is a heterotrimeric GTP-binding protein-coupled, rhodopsin-type receptor specific for PGD2 (Hirata et al., 1994[0004] , PNAS USA., 91, 11192). These mice fail to develop airway hyperreactivity and have greatly reduced eosinophil infiltration and cytokine accumulation in response to allergens. Upon allergen challenge mice deficient in the prostaglandin D2 (PGD2) receptor (DP) did not develop airway hyperactivity. Cytokine, lymphocyte and eosinophil accumulation in the lungs were greatly reduced (Matsuoka et al., 2000, Science, 287, 2013). The DP −/− mice exhibited no behavioral, anatomic, or histological abnormalities. Primary immune response is not affected by DP disruption.
  • Asthma affects more than 100 million people worldwide and more than 17 million Americans (5% of the population). Since 1980 the incidence has more than doubled and deaths have tripled (5,000 deaths in 1995). Annual asthma-related healthcare costs in the US alone were estimated to exceed $14.5 billion in 2000. Current therapies such as inhalant anti-inflammatories and bronchodilators can be used to treat symptoms, however, these therapies do not prevent or cure asthma. [0005]
  • Sandberg et al., 2001[0006] , Prog. Respir. Res., 31, 370-373, describes ribozyme therapy for asthma and COPD.
  • Sullivan et al., International U.S. Pat. No. 5,616,488, describes ribozymes targeting interleukin-5 for treatment and diagnosis of asthma and other inflammatory disorders. [0007]
  • Stinchcomb et al, International PCT Publication No. WO 95/23225, describes ribozymes and methods for inhibiting the expression of disease related genes including genes associated with asthma. [0008]
  • Nyce, International PCT Publication Nos. WO 00/62736, WO 00/09525, WO 99/13886, WO 98/23294, WO 96/40266 and U.S. Pat. No. 6,025,339 describe specific antisense oligonucleotides targeting certain mRNAs encoding particular adenosine receptors. [0009]
  • SUMMARY OF THE INVENTION
  • The invention features novel nucleic acid-based molecules, for example, enzymatic nucleic acid molecules, allozymes, antisense nucleic acids, 2-5A antisense chimeras, triplex forming oligonucleotides, decoy RNA, dsRNA, siRNA, aptamers, and antisense nucleic acids containing RNA cleaving chemical groups, and methods to modulate gene expression, for example, genes encoding prostaglandin D2 receptor (PTGDR), prostaglandin D2 synthetase (PTGDS), and adenosine receptors (AR) such as adenosine receptor A1, A2a, A2b, and A3. In particular, the instant invention features nucleic-acid based molecules and methods to modulate the expression of PTGDR, PTGDS, and adenosine A1 receptor (ADORA1). [0010]
  • In one embodiment, the invention features one or more nucleic acid-based molecules and methods that independently or in combination modulate the expression of gene(s) encoding prostaglandin D2 receptors (PTGDR), prostaglandin D2 synthetase (PTGDS) and adenosine receptors such as ADORA1. Specifically, the present invention features nucleic acid molecules that modulate the expression of prostaglandin D2 receptor (PTGDR) gene, for example Genbank Accession Nos. U31332 and U31099, prostaglandin D2 synthetase (PTGDS) gene, for example Genbank Accession No. NM[0011] 000954, and Adenosine A1 receptor (ADORA1), for example Genbank Accession No. NM000674.
  • The description below of the various aspects and embodiments is provided with reference to the exemplary prostaglandin D2 receptor (PTGDR), prostaglandin D2 synthetase (PTGDS), and adenosine A1 receptor (ADORA1). However, the various aspects and embodiments are also directed to other genes that express prostaglandin proteins and other receptors involved in allergic reactions. Those additional genes can be analyzed for target sites using the methods described for PTGDS, PTGDR, and ADORA1. Thus, the inhibition and the effects of such inhibition of the other genes can be performed as described herein. [0012]
  • In another embodiment, the invention features an enzymatic nucleic acid molecule comprising a sequence selected from the group consisting of SEQ ID NOs: 228-454, 831-1206, 1438-1668, 1715-2057, and 2247-2666. In yet another embodiment, the invention features an enzymatic nucleic acid molecule comprising at least one binding arm wherein one or more of said binding arms comprises a sequence complementary to a sequence selected from the group consisting of SEQ ID NOs: 1-227, 455-830, 1207-1437, 1669-1714, and 2058-2246. [0013]
  • In one embodiment, the invention features an antisense nucleic acid molecule comprising a sequence complementary to a sequence selected from the group consisting of SEQ ID NOs: 1-227, 455-830, 1207-1437, 1669-1714, and 2058-2246. [0014]
  • In another embodiment, an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing RNA cleaving chemical groups of the invention is adapted to treat asthma. [0015]
  • In one embodiment, an enzymatic nucleic acid molecule of the invention has an endonuclease activity to cleave RNA encoded by a PTGDS and/or PTGDR gene. [0016]
  • In another embodiment, an enzymatic nucleic acid molecule of the invention is in a hammerhead, Inozyme, Zinzyme, DNAzyme, Amberzyme, or G-cleaver configuration. [0017]
  • In another embodiment, an enzymatic nucleic acid molecule of the invention having a hammerhead configuration comprises a sequence complementary to a sequence having SEQ ID NOs: 1-227. In yet another embodiment, an enzymatic nucleic acid molecule of invention having a hammerhead configuration comprises a sequence having SEQ ID NOs: 228-454. [0018]
  • In another embodiment, an enzymatic nucleic acid molecule of the invention having an Inozyme configuration comprises a sequence complementary to a sequence having SEQ ID NOs: 455-830. In yet another embodiment, an enzymatic nucleic acid molecule of invention having an Inozyme configuration comprises a sequence having SEQ ID NOs: 831-1206. [0019]
  • In another embodiment, an enzymatic nucleic acid molecule of the invention having a Zinzyme configuration comprises a sequence complementary to a sequence having SEQ ID NOs: 1207-1437. In yet another embodiment, an enzymatic nucleic acid molecule of invention having a Zinzyme configuration comprises a sequence having SEQ ID NOs: 1438-1668. [0020]
  • In another embodiment, an enzymatic nucleic acid molecule of the invention having a DNAzyme configuration comprises a sequence complementary to a sequence having SEQ ID NOs: 1, 13, 55, 69, 74, 104, 112, 120, 123, 128, 131, 138, 147, 154, 157, 158, 169, 188, 192, 208, 221, 463, 475, 489, 505, 527, 541, 552, 554, 561, 563, 572, 591, 601, 605, 627, 637, 645, 652, 653, 661, 668, 669, 670, 676, 692, 699, 706, 719, 725, 732, 737, 741, 747, 763, 774, 782, 800, 805, 807, 816, 818, 823, 827, 828, 1207-1437, and 1669-1714. In yet another embodiment, an enzymatic nucleic acid molecule of invention having a DNAzyme configuration comprises a sequence having SEQ ID NOs: 1715-2057. [0021]
  • In another embodiment, an enzymatic nucleic acid molecule of the invention having an Amberzyme configuration comprises a sequence complementary to a sequence having SEQ ID NOs: 1207-1437, and 2058-2246. In yet another embodiment, an enzymatic nucleic acid molecule of invention having an Amberzyme configuration comprises a sequence having SEQ ID NOs: 2247-2666. [0022]
  • In one embodiment, an enzymatic nucleic acid molecule of the invention comprises between 8 and 100 bases complementary to the RNA of PTGDS, ADORA1 and/or PTGDR gene. In another embodiment, an enzymatic nucleic acid molecule of the invention comprises between 14 and 24 bases complementary to a RNA molecule of a PTGDS or PTGDR gene. [0023]
  • In one embodiment, an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing RNA cleaving chemical groups of the invention is chemically synthesized. [0024]
  • In another embodiment, an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing RNA cleaving chemical groups of the invention comprises at least one 2′-sugar modification. [0025]
  • In another embodiment, an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing RNA cleaving chemical groups of the invention comprises at least one nucleic acid base modification. [0026]
  • In another embodiment, an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acids containing RNA cleaving chemical groups of the invention comprises at least one phosphate backbone modification. [0027]
  • In one embodiment, the invention features a mammalian cell, for example a human cell, including the enzymatic nucleic acid molecule of the invention. [0028]
  • In another embodiment, the invention features a method of reducing PTGDS, ADORA1 and/or PTGDR expression or activity in a cell, comprising contacting the cell with an enzymatic nucleic acid molecule of the invention, under conditions suitable for the reduction. [0029]
  • In another embodiment, the invention features a method of reducing PTGDS, ADORA1 and/or PTGDR expression or activity in a cell, comprising the step of contacting the cell with an antisense nucleic acid molecule of the invention under conditions suitable for the reduction. [0030]
  • In yet another embodiment, the invention features a method of treatment of a patient having a condition associated with the level of PTGDS, ADORA1 and/or PTGDR, comprising contacting cells of the patient with an enzymatic nucleic acid molecule of the invention, under conditions suitable for the treatment. [0031]
  • In one embodiment, the invention features a method of treatment of a patient having a condition associated with the level of PTGDS, ADORA1 and/or PTGDR, comprising contacting cells of the patient with an antisense nucleic acid molecule of the invention, under conditions suitable for the treatment. [0032]
  • In another embodiment, a method of treatment of a patient having a condition associated with the level of PTGDS, ADORA1 and/or PTGDR is featured, wherein the method further comprises the use of one or more drug therapies under conditions suitable for the treatment. [0033]
  • For example, in one embodiment, the invention features a method for treatment of asthma, allergic rhinitis, or atopic dermatitis under conditions suitable for the treatment. [0034]
  • In another embodiment, the invention features a method of cleaving a RNA molecule of PTGDS, ADORA1 and/or PTGDR gene comprising contacting an enzymatic nucleic acid molecule of the invention with a RNA molecule of a PTGDS, ADORA1 and/or PTGDR gene under conditions suitable for the cleavage, for example, wherein the cleavage is carried out in the presence of a divalent cation, such as Mg[0035] 2+.
  • In one embodiment, an enzymatic nucleic acid molecule of the invention comprises a cap structure, for example a 3′,3′-linked or 5′,5′-linked deoxyabasic ribose derivative, wherein the cap structure is at the 5′-end, or 3′-end, or both the 5′-end and the 3′-end of the enzymatic nucleic acid molecule. [0036]
  • In another embodiment, an antisense nucleic acid molecule of the invention comprises a cap structure, for example a 3′,3′-linked or 5′,5′-linked deoxyabasic ribose derivative, wherein the cap structure is at the 5′-end, or 3′-end, or both the 5′-end and the 3′-end of the antisense nucleic acid molecule. [0037]
  • In one embodiment, the invention features an expression vector comprising a nucleic acid sequence encoding at least one enzymatic nucleic acid molecule of the invention, in a manner which allows expression of the nucleic acid molecule. [0038]
  • In another embodiment, the invention features a mammalian cell, for example, a human cell, including an expression vector of the invention. [0039]
  • In yet another embodiment, the expression vector of the invention further comprises a sequence for an antisense nucleic acid molecule complementary to a RNA molecule of a PTGDS, ADORA1 and/or PTGDR gene. [0040]
  • In one embodiment, an expression vector of the invention comprises a nucleic acid sequence encoding two or more enzymatic nucleic acid molecules, which can be the same or different. [0041]
  • In another embodiment, the invention features a method for treatment of asthma, allergic rhinitis, or atopic dermatitis, comprising administering to a patient an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing RNA cleaving chemical groups of the invention, under conditions suitable for the treatment, including administering to the patient one or more other therapies, for example, inhalant anti-inflammatories, bronchodilators, adenosine inhibitors and adenosine A1 receptor inhibitors. [0042]
  • In one embodiment, the method of treatment features an enzymatic nucleic acid molecule or antisense nucleic acid molecule of the invention comprises at least five ribose residues, at least ten 2′-O-methyl modifications, and a 3′-end modification, such as a 3′-3′ inverted abasic moiety. In another embodiment, an enzymatic nucleic acid molecule or antisense nucleic acid molecule of the invention further comprises phosphorothioate linkages on at least three of the 5′ terminal nucleotides. [0043]
  • In another embodiment, the invention features a method of administering to a mammal, for example a human, an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing RNA cleaving chemical groups of the invention, comprising contacting the mammal with the nucleic acid molecule under conditions suitable for the administration, for example, in the presence of a delivery reagent such as a lipid, cationic lipid, phospholipid, or liposome. [0044]
  • In yet another embodiment, the invention features a method of administering to a mammal an enzymatic nucleic acid molecule, antisense nucleic acid molecule, 2-5A antisense chimera, triplex forming oligonucleotide, decoy RNA, dsRNA, siRNA, aptamer, or antisense nucleic acid containing RNA cleaving chemical groups of the invention in conjunction with a therapeutic agent, comprising contacting the mammal, for example a human, with the nucleic acid molecule and the therapeutic agent under conditions suitable for the administration. [0045]
  • In one embodiment, the invention features the use of an enzymatic nucleic acid molecule, which can be in a hammerhead, NCH, G-cleaver, Amberzyme, Zinzyme, and/or DNAzyme motif, to down-regulate the expression of a PTGDS, an ADORA1 and/or a PTGDR gene. [0046]
  • By “inhibit”, “down-regulate”, or “reduce”, it is meant that the expression of the gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins or protein subunits, or activity of one or more proteins or protein subunits, such as PTGDS, ADORA1 and/or PTGDR proteins or PTGDS, ADORA1 and/or PTGDR subunit(s), is reduced below that observed in the absence of the nucleic acid molecules of the invention. In one embodiment, inhibition, down-regulation or reduction with an enzymatic nucleic acid molecule is below that level observed in the presence of an enzymatically inactive or attenuated molecule that is able to bind to the same site on the target RNA molecule, but is unable to cleave that RNA molecule. In another embodiment, inhibition, down-regulation, or reduction with antisense oligonucleotides is below that level observed in the presence of, for example, an oligonucleotide with scrambled sequence or with mismatches. In another embodiment, inhibition, down-regulation, or reduction of PTGDS, ADORA1 and/or PTGDR with a nucleic acid molecule of the instant invention is greater in the presence of the nucleic acid molecule than in its absence. [0047]
  • By “up-regulate” is meant that the expression of a gene, or level of RNA molecules or equivalent RNA molecules encoding one or more proteins, protein subunits, or activity of one or more proteins or protein subunits, such as PTGDS, ADORA1 and/or PTGDR proteins or PTGDS, ADORA1 and/or PTGDR subunits, is greater than that observed in the absence of the nucleic acid molecules of the invention. For example, the expression of a gene, such as PTGDS, ADORA1 and/or PTGDR gene, can be increased in order to treat, prevent, ameliorate, or modulate a pathological condition caused or exacerbated by an absence or low level of gene expression. [0048]
  • By “modulate” is meant that the expression of the gene, or level of RNA molecules or equivalent RNA molecules encoding one or more protein subunits, or activity of one or more protein subunits is up-regulated or down-regulated, such that the expression, level, or activity is greater than or less than that observed in the absence of a nucleic acid molecule of the invention. [0049]
  • By “enzymatic nucleic acid molecule” it is meant a nucleic acid molecule that has complementarity in a substrate binding region to a specified gene target, and also has an enzymatic activity that is active to specifically cleave target a RNA molecule. That is, the enzymatic nucleic acid molecule is able to intermolecularly cleave a RNA molecule and thereby inactivate a target RNA molecule. These complementary regions allow sufficient hybridization of an enzymatic nucleic acid molecule to a target RNA molecule and thus permit cleavage. One hundred percent complementarity is preferred, but complementarity as low as 50-75% can also be useful in this invention (see for example Werner and Uhlenbeck, 1995[0050] , Nucleic Acids Research, 23, 2092-2096; Hammann et al., 1999, Antisense and Nucleic Acid Drug Dev., 9, 25-31). The nucleic acids can be modified at the base, sugar, and/or phosphate groups. The term enzymatic nucleic acid is used interchangeably with phrases such as ribozymes, catalytic RNA, enzymatic RNA, catalytic DNA, aptazyme or aptamer-binding ribozyme, regulatable ribozyme, catalytic oligonucleotides, nucleozyme, DNAzyme, RNA enzyme, endoribonuclease, endonuclease, minizyme, leadzyme, oligozyme or DNA enzyme. All of these terminologies describe nucleic acid molecules with enzymatic activity. The specific enzymatic nucleic acid molecules described in the instant application are not limiting in the invention and those skilled in the art will recognize that all that is important in an enzymatic nucleic acid molecule of this invention is that it has a specific substrate binding site that is complementary to one or more of the target nucleic acid regions, and that it have nucleotide sequences within or surrounding that substrate binding site which impart a nucleic acid cleaving and/or ligation activity to the molecule (Cech et al, U.S. Pat. No. 4,987,071; Cech et al., 1988, 260 JAMA 3030).
  • By “nucleic acid molecule” as used herein is meant a molecule having nucleotides. [0051]
  • The nucleic acid can be single, double, or multiple stranded and can comprise modified or unmodified nucleotides or non-nucleotides or various mixtures and combinations thereof. [0052]
  • By “enzymatic portion” or “catalytic domain” is meant that portion/region of the enzymatic nucleic acid molecule essential for cleavage of a nucleic acid substrate (for example see FIGS. [0053] 1-4).
  • By “substrate binding arm” or “substrate binding domain” is meant that portion/region of a enzymatic nucleic acid that is able to interact, for example via complementarity (i.e., able to base-pair with), with a portion of its substrate. Such complementarity can be 100%, but can be less if desired. For example, as few as 10 bases out of 14 can be base-paired (see for example Werner and Uhlenbeck, 1995[0054] , Nucleic Acids Research, 23, 2092-2096; Hammann et al, 1999, Antisense and Nucleic Acid Drug Dev., 9, 25-31). Examples of such arms are shown generally in FIGS. 1-4. That is, these arms contain sequences within a enzymatic nucleic acid that are intended to bring enzymatic nucleic acid and target RNA together through complementary base-pairing interactions. The enzymatic nucleic acid of the invention can have binding arms that are contiguous or non-contiguous and can be of varying lengths. The length of the binding arm(s) can be greater than or equal to four nucleotides and of sufficient length to stably interact with a target RNA; in one embodiment they can be 12-100 nucleotides; in another embodiment they can be 14-24 nucleotides long (see for example Werner and Uhlenbeck, supra; Hamman et al., supra; Hampel et al., EP0360257; Berzal-Herranze et al., 1993, EMBO J., 12, 2567-73) or between 8 and 14 nucleotides long. If two binding arms are chosen, the design is such that the length of the binding arms are symmetrical (i.e., each of the binding arms is of the same length; e.g., four and four, five and five nucleotides, or six and six nucleotides, or seven and seven nucleotides long) or asymmetrical (i.e., the binding arms are of different length; e.g., three and five, six and three nucleotides; three and six nucleotides long; four and five nucleotides long; four and six nucleotides long; four and seven nucleotides long; and the like).
  • By “Inozyme” or “NCH” motif or configuration is meant, an enzymatic nucleic acid molecule comprising a motif as is generally described as NCH Rz in FIG. 1. Inozymes possess endonuclease activity to cleave RNA substrates having a cleavage triplet NCH/, where N is a nucleotide, C is cytidine and H is adenosine, uridine or cytidine, and / represents the cleavage site. H is used interchangeably with X. Inozymes can also possess endonuclease activity to cleave RNA substrates having a cleavage triplet NCN/, where N is a nucleotide, C is cytidine, and / represents the cleavage site. “I” in FIG. 1 represents an Inosine nucleotide, including a ribo-Inosine or xylo-Inosine nucleoside. [0055]
  • By “G-cleaver” motif or configuration is meant, an enzymatic nucleic acid molecule comprising a motif as is generally described as G-cleaver Rz in FIG. 1. G-cleavers possess endonuclease activity to cleave RNA substrates having a cleavage triplet NYN/, where N is a nucleotide, Y is uridine or cytidine and/represents the cleavage site. G-cleavers can be chemically modified as is generally shown in FIG. 1. [0056]
  • By “amberzyme” motif or configuration is meant, an enzymatic nucleic acid molecule comprising a motif as is generally described in FIG. 2. Amberzymes possess endonuclease activity to cleave RNA substrates having a cleavage triplet NG/N, where N is a nucleotide, G is guanosine, and/represents the cleavage site. Amberzymes can be chemically modified to increase nuclease stability through substitutions as are generally shown in FIG. 2. In addition, differing nucleoside and/or non-nucleoside linkers can be used to substitute the 5′-gaaa-3′ loops shown in the figure. Amberzymes represent a non-limiting example of an enzymatic nucleic acid molecule that does not require a ribonucleotide (2′-OH) group within its own nucleic acid sequence for activity. [0057]
  • By “zinzyme” motif or configuration is meant, an enzymatic nucleic acid molecule comprising a motif as is generally described in FIG. 3. Zinzymes possess endonuclease activity to cleave RNA substrates having a cleavage triplet including but not limited to YG/Y, where Y is uridine or cytidine, and G is guanosine and/represents the cleavage site. [0058]
  • Zinzymes can be chemically modified to increase nuclease stability through substitutions as are generally shown in FIG. 3, including substituting 2′-O-methyl guanosine nucleotides for guanosine nucleotides. In addition, differing nucleotide and/or non-nucleotide linkers can be used to substitute the 5′-gaaa-2′ loop shown in the figure. Zinzymes represent a non-limiting example of an enzymatic nucleic acid molecule that does not require a ribonucleotide (2′-OH) group within its own nucleic acid sequence for activity. [0059]
  • By ‘DNAzyme’ is meant, an enzymatic nucleic acid molecule that does not require the presence of a 2′-OH group within its own nucleic acid sequence for activity. In particular embodiments the enzymatic nucleic acid molecule can have an attached linker or linkers or other attached or associated groups, moieties, or chains containing one or more nucleotides with 2′-OH groups. DNAzymes can be synthesized chemically or expressed endogenously in vivo, by means of a single stranded DNA vector or equivalent thereof. An example of a DNAzyme is shown in FIG. 4 and is generally reviewed in Usman et al., U.S. Pat. No. 6,159,714; Chartrand et al., 1995[0060] , NAR 23, 4092; Breaker et al., 1995, Chem. Bio. 2, 655; Santoro et al., 1997, PNAS 94, 4262; Breaker, 1999, Nature Biotechnology, 17, 422-423; and Santoro et. al., 2000, J. Am. Chem. Soc., 122, 2433-39. The “10-23” DNAzyme motif is one particular type of DNAzyme that was evolved using in vitro selection (see Santoro et al., supra). Additional DNAzyme motifs can be selected for using techniques similar to those described in these references, and hence, are within the scope of the present invention.
  • By “sufficient length” is meant an oligonucleotide of greater than or equal to 3 nucleotides that is of a length great enough to provide the intended function under the expected condition. For example, for binding arms of enzymatic nucleic acid “sufficient length” means that the binding arm sequence is long enough to provide stable binding to a target site under the expected binding conditions. The binding arms are not so long as to prevent useful turnover of the nucleic acid molecule. [0061]
  • By “stably interact” is meant interaction of the oligonucleotides with target nucleic acid (e.g., by forming hydrogen bonds with complementary nucleotides in the target under physiological conditions) that is sufficient to the intended purpose (e.g., cleavage of target RNA by an enzyme). [0062]
  • By “equivalent” RNA to PTGDS is meant to include RNA molecules having homology (partial or complete) to RNA molecules encoding PTGDS proteins or encoding proteins with similar function as PTGDS proteins in various organisms, including human, rodent, primate, rabbit, pig, plants, protozoans, fungi, and other microorganisms and parasites. The equivalent RNA sequence can also include in addition to the coding region, regions such as 5′-untranslated region, 3′-untranslated region, introns, intron-exon junction and the like. [0063]
  • By “equivalent” RNA to PTGDR is meant to include RNA molecules having homology (partial or complete) to RNA molecules encoding PTGDR proteins or encoding proteins with similar function as PTGDR proteins in various organisms, including human, rodent, primate, rabbit, pig, plants, protozoans, fungi, and other microorganisms and parasites. The equivalent RNA sequence can also include in addition to the coding region, regions such as 5′-untranslated region, 3′-untranslated region, introns, intron-exon junction and the like. [0064]
  • By “equivalent” RNA to ADORA1 is meant to include RNA molecules having homology (partial or complete) to RNA molecule encoding ADORA1 proteins or encoding proteins with similar function as ADORA1 proteins in various organisms, including human, rodent, primate, rabbit, pig, plants, protozoans, fungi, and other microorganisms and parasites. The equivalent RNA sequence can also include in addition to the coding region, regions such as 5′-untranslated region, 3′-untranslated region, introns, intron-exon junction and the like. [0065]
  • By “homology” is meant the nucleotide sequence of two or more nucleic acid molecules is partially or completely identical. [0066]
  • By “antisense nucleic acid”, it is meant a non-enzymatic nucleic acid molecule that binds to target RNA by means of RNA-RNA or RNA-DNA or RNA-PNA (protein nucleic acid; Egholm et al., 1993 [0067] Nature 365, 566) interactions and alters the activity of the target RNA (for a review, see Stein and Cheng, 1993 Science 261, 1004 and Woolf et al., U.S. Pat. No. 5,849,902). Typically, antisense molecules are complementary to a target sequence along a single contiguous sequence of the antisense molecule. However, in certain embodiments, an antisense molecule can bind to substrate such that the substrate molecule forms a loop, and/or an antisense molecule can bind such that the antisense molecule forms a loop. Thus, the antisense molecule can be complementary to two (or even more) non-contiguous substrate sequences or two (or even more) non-contiguous sequence portions of an antisense molecule can be complementary to a target sequence or both. For a review of current antisense strategies, see Schmajuk et al., 1999, J. Biol. Chem., 274, 21783-21789, Delihas et al., 1997, Nature, 15, 751-753, Stein et al., 1997, Antisense N. A. Drug Dev., 7, 151, Crooke, 2000, Methods Enzymol., 313, 3-45; Crooke, 1998, Biotech. Genet. Eng. Rev., 15, 121-157, Crooke, 1997, Ad. Pharmacol., 40, 1-49. In addition, antisense DNA can be used to target RNA by means of DNA-RNA interactions, thereby activating RNase H, which digests the target RNA in the duplex. The antisense oligonucleotides can comprise one or more RNAse H activating region, which is capable of activating RNAse H cleavage of a target RNA. Antisense DNA can be synthesized chemically or expressed via the use of a single stranded DNA expression vector or equivalent thereof.
  • By “RNase H activating region” is meant a region (generally greater than or equal to 4-25 nucleotides in length, and in one embodiment from 5-11 nucleotides in length) of a nucleic acid molecule capable of binding to a target RNA to form a non-covalent complex that is recognized by cellular RNase H enzyme (see for example Arrow et al., U.S. Pat. No. 5,849,902; Arrow et al., U.S. Pat. No. 5,989,912). The RNase H enzyme binds to the nucleic acid molecule-target RNA complex and cleaves the target RNA sequence. The RNase H activating region comprises, for example, phosphodiester, phosphorothioate (at least four of the nucleotides are phosphorothiote substitutions; and in another embodiment, 4-11 of the nucleotides are phosphorothiote substitutions); phosphorodithioate, 5′-thiophosphate, or methylphosphonate backbone chemistry or a combination thereof. In addition to one or more backbone chemistries described above, the RNase H activating region can also comprise a variety of sugar chemistries. For example, the RNase H activating region can comprise deoxyribose, arabino, fluoroarabino or a combination thereof, nucleotide sugar chemistry. Those skilled in the art will recognize that the foregoing are non-limiting examples and that any combination of phosphate, sugar and base chemistry of a nucleic acid that supports the activity of RNase H enzyme is within the scope of the definition of the RNase H activating region and the instant invention. [0068]
  • By “2-5A antisense chimera” is meant an antisense oligonucleotide containing a 5′-phosphorylated 2′-5′-linked adenylate residue. These chimeras bind to target RNA in a sequence-specific manner and activate a cellular 2-5A-dependent ribonuclease which, in turn, cleaves the target RNA (Torrence et al., 1993 [0069] Proc. Natl. Acad. Sci. USA 90, 1300; Silverman et al., 2000, Methods Enzymol., 313, 522-533; Player and Torrence, 1998, Pharmacol. Ther., 78, 55-113).
  • By “triplex forming oligonucleotides” is meant an oligonucleotide that can bind to a double-stranded DNA in a sequence-specific manner to form a triple-strand helix. Formation of such triple helix structure has been shown to inhibit transcription of the targeted gene (Duval-Valentin et al., 1992 [0070] Proc. Natl. Acad. Sci. USA 89, 504; Fox, 2000, Curr. Med. Chem., 7, 17-37; Praseuth et. al., 2000, Biochim. Biophys. Acta, 1489, 181-206).
  • By “gene” it is meant a nucleic acid that encodes an RNA, for example, nucleic acid sequences including but not limited to structural genes encoding a polypeptide. [0071]
  • “Complementarity” refers to the ability of a nucleic acid to form hydrogen bond(s) with another RNA molecule by either traditional Watson-Crick or other non-traditional types. In reference to the nucleic molecules of the present invention, the binding free energy for a nucleic acid molecule with its target or complementary sequence is sufficient to allow the relevant function of the nucleic acid to proceed, e.g., enzymatic nucleic acid cleavage, antisense or triple helix inhibition. Determination of binding free energies for nucleic acid molecules is well known in the art (see, e.g., Turner et al., 1987[0072] , CSHSymp. Quant. Biol. LII pp.123-133; Frier et al., 1986, Proc. Nat. Acad. Sci. USA 83:9373-9377; Turner et al., 1987, J. Am. Chem. Soc. 109:3783-3785). A percent complementarity indicates the percentage of contiguous residues in a nucleic acid molecule that can form hydrogen bonds (e.g., Watson-Crick base pairing) with a second nucleic acid sequence (e.g., 5, 6, 7, 8, 9, 10 out of 10 being 50%, 60%, 70%, 80%, 90%, and 100% complementary). “Perfectly complementary” means that all the contiguous residues of a nucleic acid sequence will hydrogen bond with the same number of contiguous residues in a second nucleic acid sequence.
  • By “RNA” is meant a molecule comprising at least one ribonucleotide residue. By “ribonucleotide” or “2′-OH” is meant a nucleotide with a hydroxyl group at the 2′ position of a β-D-ribo-furanose moiety. [0073]
  • By “decoy RNA” is meant an RNA molecule or aptamer that is designed to preferentially bind to a predetermined ligand. Such binding can result in the inhibition or activation of a target molecule. The decoy RNA or aptamer can compete with a naturally occurring binding target for the binding of a specific ligand. For example, it has been shown that over-expression of HIV trans-activation response (TAR) RNA can act as a “decoy” and efficiently binds HIV tat protein, thereby preventing it from binding to TAR sequences encoded in the HIV RNA (Sullenger et al., 1990, Cell, 63, 601-608). This is but a specific example and those in the art will recognize that other embodiments can be readily generated using techniques generally known in the art, see for example Gold et al., 1995[0074] , Annu. Rev. Biochem., 64, 763; Brody and Gold, 2000, J. Biotechnol., 74, 5; Sun, 2000, Curr. Opin. Mol. Ther., 2, 100; Kusser, 2000, J. Biotechnol., 74, 27; Hermann and Patel, 2000, Science, 287, 820; and Jayasena, 1999, Clinical Chemistry, 45, 1628. Similarly, a decoy RNA can be designed to bind to a D2 receptor and block the binding of PTGDS or a decoy RNA can be designed to bind to PTGDS and prevent interaction with the D2 receptor.
  • The term “double stranded RNA” or “dsRNA” as used herein refers to a double stranded RNA molecule capable of RNA interference, including short interfering RNA “siRNA” (see, e.g., Bass, 2001[0075] , Nature, 411, 428-429; Elbashir et al., 2001, Nature, 411, 494-498).
  • The term “allozyme” as used herein refers to an allosteric enzymatic nucleic acid molecule, see, e.g., George et al., U.S. Pat. Nos. 5,834,186 and 5,741,679, Shih et al., U.S. Pat. No. 5,589,332, Nathan et al., U.S. Pat. No. 5,871,914, Nathan and Ellington, International PCT publication No. WO 00/24931, Breaker et al., International PCT Publication Nos. WO 00/26226 and 98/27104, and Sullenger et al., International PCT publication No. WO 99/29842. The term “2-5A chimera” as used herein refers to an oligonucleotide containing a 5′-phosphorylated 2′-5′-linked adenylate residue. These chimeras bind to target RNA in a sequence-specific manner and activate a cellular 2-5A-dependent ribonuclease which, in turn, cleaves the target RNA (Torrence et al., 1993 [0076] Proc. Natl. Acad. Sci. USA 90, 1300; Silverman et al., 2000, Methods Enzymol., 313, 522-533; Player and Torrence, 1998, Pharmacol. Ther., 78, 55-113).
  • The term “triplex forming oligonucleotides” as used herein refers to an oligonucleotide that can bind to a double-stranded DNA in a sequence-specific manner to form a triple-strand helix. Formation of such triple helix structure has been shown to inhibit transcription of the targeted gene (Duval-Valentin et al., 1992 [0077] Proc. Natl. Acad. Sci. USA 89, 504; Fox, 2000, Curr. Med. Chem., 7, 17-37; Praseuth et. al., 2000, Biochim. Biophys. Acta, 1489, 181-206).
  • Several varieties of naturally-occurring enzymatic RNAs are known presently. Each can catalyze the hydrolysis of RNA phosphodiester bonds in trans (and thus can cleave other RNA molecules) under physiological conditions. Table I summarizes some of the characteristics of these ribozymes. In general, enzymatic nucleic acids act by first binding to a target RNA. Such binding occurs through the target binding portion of a enzymatic nucleic acid that is held in close proximity to an enzymatic portion of the molecule that acts to cleave the target RNA. Thus, the enzymatic nucleic acid first recognizes and then binds a target RNA through complementary base-pairing, and once bound to the correct site, acts enzymatically to cut the target RNA. Strategic cleavage of such a target RNA will destroy its ability to direct synthesis of an encoded protein. After an enzymatic nucleic acid has bound and cleaved its RNA target, it is released from that RNA to search for another target and can repeatedly bind and cleave new targets. Thus, a single ribozyme molecule is able to cleave many molecules of target RNA. In addition, the ribozyme is a highly specific inhibitor of gene expression, with the specificity of inhibition depending not only on the base-pairing mechanism of binding to the target RNA, but also on the mechanism of target RNA cleavage. Single mismatches, or base-substitutions, near the site of cleavage can completely eliminate catalytic activity of a ribozyme. [0078]
  • The enzymatic nucleic acid molecule that cleave the specified sites in PTGDS, ADORA1 and PTGDR-specific RNAs represent a novel therapeutic approach to treat a variety of allergic diseases or conditions, including but not limited to asthma, allergic rhinitis, atopic dermatitis, and/or other allergic or inflammatory diseases and conditions which respond to the modulation of PTGDS, ADORA1 and/or PTGDR expression. [0079]
  • In one embodiment of the inventions described herein, the enzymatic nucleic acid molecule is formed in a hammerhead or hairpin motif, but can also be formed in the motif of a hepatitis delta virus, group I intron, group II intron or RNase P RNA (in association with an RNA guide sequence), Neurospora VS RNA, DNAzymes, NCH cleaving motifs, or G-cleavers. Examples of such hammerhead motifs are described by Dreyfus, supra, Rossi et al., 1992[0080] , AIDS Research and Human Retroviruses 8, 183; of hairpin motifs by Hampel et al., EP0360257, Hampel and Tritz, 1989 Biochemistry 28, 4929, Feldstein et al., 1989, Gene 82, 53, Haseloff and Gerlach, 1989, Gene, 82, 43, and Hampel et al., 1990 Nucleic Acids Res. 18, 299; Chowrira & McSwiggen, U.S. Pat. No. 5,631,359; of the hepatitis delta virus motif is described by Perrotta and Been, 1992 Biochemistry 31, 16; of the RNase P motif by Guerrier-Takada et al., 1983 Cell 35, 849; Forster and Altman, 1990, Science 249, 783; Li and Altman, 1996, Nucleic Acids Res. 24, 835; Neurospora VS RNA ribozyme motif is described by Collins (Saville and Collins, 1990 Cell 61, 685-696; Saville and Collins, 1991 Proc. Natl. Acad. Sci. USA 88, 8826-8830; Collins and Olive, 1993 Biochemistry 32, 2795-2799; Guo and Collins, 1995, EMBO. J 14, 363); Group II introns are described by Griffin et al., 1995, Chem. Biol. 2, 761; Michels and Pyle, 1995, Biochemistry 34, 2965; Pyle et al., International PCT Publication No. WO 96/22689; of the Group I intron by Cech et al., U.S. Pat. No. 4,987,071 and of DNAzymes by Usman et al., International PCT Publication No. WO 95/11304; Chartrand et al., 1995, NAR 23, 4092; Breaker et al., 1995, Chem. Bio. 2, 655; Santoro et al., 1997, PNAS 94, 4262, and Beigelman et al., International PCT publication No. WO 99/55857. NCH cleaving motifs are described in Ludwig & Sproat, International PCT Publication No. WO 98/58058; and G-cleavers are described in Kore et al., 1998, Nucleic Acids Research 26, 4116-4120 and Eckstein et al., International PCT Publication No. WO 99/16871. Additional motifs such as the Aptazyme (Breaker et al., WO 98/43993), Amberzyme (Class I motif; FIG. 2; Beigelman et al., U.S. Ser. No. 09/301,511) and Zinzyme (FIG. 3) (Beigelman et al., U.S. Ser. No. 09/301,511), all included by reference herein including drawings, can also be used in the present invention. These specific motifs or configurations are not limiting in the invention and those skilled in the art will recognize that all that is important in an enzymatic nucleic acid molecule of this invention is that it has a specific substrate binding site which is complementary to one or more of the target gene RNA regions, and that it have nucleotide sequences within or surrounding that substrate binding site which impart an RNA cleaving activity to the molecule (Cech et al., U.S. Pat. No. 4,987,071).
  • In one embodiment of the present invention, a nucleic acid molecule of the instant invention can be between 12 and 100 nucleotides in length. Exemplary enzymatic nucleic acid molecules of the invention are shown in Table III-VII. For example, enzymatic nucleic acid molecules of the invention can be between 15 and 50 nucleotides in length, and in another embodiment between 25 and 40 nucleotides in length, e.g., 34, 36, or 38 nucleotides in length (for example see Jarvis et al., 1996[0081] , J. Biol. Chem., 271, 29107-29112). Exemplary DNAzymes of the invention are can between 15 and 40 nucleotides in length, and in one embodiment, between 25 and 35 nucleotides in length, e.g., 29, 30, 31, or 32 nucleotides in length (see, e.g., Santoro et al., 1998, Biochemistry, 37, 13330-13342; Chartrand et al., 1995, Nucleic Acids Research, 23, 4092-4096). Exemplary antisense molecules of the invention can be between 15 and 75 nucleotides in length, and in one embodiment between 20 and 35 nucleotides in length, e.g., 25, 26, 27, or 28 nucleotides in length (see for example Woolf et al., 1992, PNAS., 89, 7305-7309; Milner et al., 1997, Nature Biotechnology, 15, 537-541). Exemplary triplex forming oligonucleotide molecules of the invention are between 10 and 40 nucleotides in length, and in one embodiment are between 12 and 25 nucleotides in length, e.g., 18, 19, 20, or 21 nucleotides in length (see for example Maher et al., 1990, Biochemistry, 29, 8820-8826; Strobel and Dervan, 1990, Science, 249, 73-75). Those skilled in the art will recognize that all that is required is for the nucleic acid molecule to be of length and conformation sufficient and suitable for the nucleic acid molecule to catalyze a reaction contemplated herein. The length of the nucleic acid molecules of the instant invention are not limiting within the general limits stated.
  • In one embodiment, a nucleic acid molecule that modulates, for example, down-regulates, PTGDS replication or expression comprises between 8 and 100 bases complementary to a RNA molecule of PTGDS. In another embodiment, a nucleic acid molecule that modulates PTGDS replication or expression comprises between 14 and 24 bases complementary to a RNA molecule of PTGDS. [0082]
  • In another embodiment, a nucleic acid molecule that modulates, for example, down-regulates, PTGDR replication or expression comprises between 8 and 100 bases complementary to a RNA molecule of PTGDR. In another embodiment, a nucleic acid molecule that modulates PTGDR replication or expression comprises between 14 and 24 bases complementary to a RNA molecule of PTGDR. [0083]
  • In another embodiment, a nucleic acid molecule that modulates, for example, down-regulates, ADORA1 replication or expression comprises between 8 and 100 bases complementary to a RNA molecule of ADORA1. In another embodiment, a nucleic acid molecule that modulates ADORA1 replication or expression comprises between 14 and 24 bases complementary to a RNA molecule of ADORA1. [0084]
  • The invention provides a method for producing a class of nucleic acid-based gene modulating agents that exhibit a high degree of specificity for the RNA of a desired target. For example, the enzymatic nucleic acid molecule is can be targeted to a highly conserved sequence region of target RNAs encoding PTGDS, ADORA1 and/or PTGDR (e.g., PTGDS, ADORA1 and/or PTGDR genes) such that specific treatment of a disease or condition can be provided with either one or several nucleic acid molecules of the invention. Such nucleic acid molecules can be delivered exogenously to specific tissue or cellular targets as required. Alternatively, the nucleic acid molecules (e.g., ribozymes and antisense) can be expressed from DNA and/or RNA vectors that are delivered to specific cells. [0085]
  • As used in herein “cell” is used in its usual biological sense, and does not refer to an entire multicellular organism. The cell can, for example, be in vitro, e.g., in cell culture, or present in a multicellular organism, including, e.g., birds, plants and mammals such as humans, cows, sheep, apes, monkeys, swine, dogs, and cats. The cell may be prokaryotic (e.g., bacterial cell) or eukaryotic (e.g., mammalian or plant cell). [0086]
  • By “PTGDR proteins” is meant, a protein receptor or a mutant protein or peptide derivative thereof, having prostaglandin D2 receptor activity, for example, having the ability to bind prostaglandin D2 and/or having GTP-binding protein coupled activity. [0087]
  • By “PTGDS proteins” is meant, a prostaglandin synthetase protein or a mutant protein or peptide derivative thereof, having prostaglandin D2 synthetase activity, for example, having the ability to convert PGH2 to PGD2. [0088]
  • By “highly conserved sequence region” is meant, a nucleotide sequence of one or more regions in a target gene does not vary significantly from one generation to the other or from one biological system to the other. [0089]
  • Nucleic acid-based inhibitors of PTGDS, ADORA1 and PTGDR expression are useful for the prevention and/or treatment of allergic diseases or conditions, including but not limited to asthma, allergic rhinitis, atopic dermatitis, and any other diseases or conditions that are related to or will respond to the levels of PTGDS, ADORA1 and/or PTGDR in a cell or tissue, alone or in combination with other therapies. The reduction of PTGDS, ADORA1 and/or PTGDR expression (specifically PTGDS, ADORA1 and/or PTGDR gene RNA levels) and thus reduction in the level of the respective protein relieves, to some extent, the symptoms of the disease or condition. [0090]
  • The nucleic acid-based inhibitors of the invention can be added directly, or can be complexed with cationic lipids, packaged within liposomes, or otherwise delivered to target cells or tissues, for example by pulmonary delivery of an aerosol formulation with an inhaler or nebulizer. The nucleic acid or nucleic acid complexes can be locally administered to relevant tissues ex vivo, or in vivo through inhalation, injection or infusion pump, with or without their incorporation in biopolymers. In preferred embodiments, the enzymatic nucleic acid inhibitors comprise sequences that are complementary to the substrate sequences in Tables III to VII. Examples of such enzymatic nucleic acid molecules also are shown in Tables III to VII. Examples of such enzymatic nucleic acid molecules consist essentially of sequences defined in these tables. [0091]
  • In another embodiment, the invention features antisense nucleic acid molecules and 2-5A chimera including sequences complementary to the substrate sequences shown in Tables III to VII. Such nucleic acid molecules can include sequences as shown for the binding arms of the enzymatic nucleic acid molecules in Tables III to VII. Similarly, triplex molecules can be provided targeted to the corresponding DNA target regions, and containing the DNA equivalent of a target sequence or a sequence complementary to the specified target (substrate) sequence. Typically, antisense molecules are complementary to a target sequence along a single contiguous sequence of the antisense molecule. However, in certain embodiments, an antisense molecule can bind to substrate such that the substrate molecule forms a loop, and/or an antisense molecule can bind such that the antisense molecule forms a loop. Thus, the antisense molecule can be complementary to two (or even more) non-contiguous substrate sequences or two (or even more) non-contiguous sequence portions of an antisense molecule can be complementary to a target sequence or both. [0092]
  • By “consists essentially of” is meant that the active nucleic acid molecule of the invention, for example, an enzymatic nucleic acid molecule, contains an enzymatic center or core equivalent to those in the examples, and binding arms able to bind RNA such that cleavage at the target site occurs. Other sequences can be present that do not interfere with such cleavage. Thus, a core region can, for example, include one or more loop, stem-loop structure, or linker which does not prevent enzymatic activity. Thus, the underlined regions in the sequences in Tables III and IV can be such a loop, stem-loop, nucleotide linker, and/or non-nucleotide linker and can be represented generally as sequence “X”. For example, a core sequence for a hammerhead enzymatic nucleic acid can comprise a conserved sequence, such as 5′-CUGAUGAG-3′ and 5′-CGAA-3′ connected by “X”, where X is 5′-[0093] GCCGUUAGGC-3′ (SEQ ID NO: 2678), or any other Stem II region known in the art, or a nucleotide and/or non-nucleotide linker. Similarly, for other nucleic acid molecules of the instant invention, such as Inozyme, G-cleaver, amberzyme, zinzyme, DNAzyme, antisense, 2-5A antisense, triplex forming nucleic acid, and decoy nucleic acids, other sequences or non-nucleotide linkers can be present that do not interfere with the function of the nucleic acid molecule.
  • Sequence X can be a linker of ≧2 nucleotides in length, including 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 26, 30, where the nucleotides can be internally base-paired to form a stem of ≧2 base pairs. Alternatively or in addition, sequence X can be a non-nucleotide linker. In yet another embodiment, the nucleotide linker X can be a nucleic acid aptamer, such as an ATP aptamer, HIV Rev aptamer (RRE), HIV Tat aptamer (TAR) and others (for a review see Gold et al., 1995[0094] , Annu. Rev. Biochem., 64, 763; and Szostak & Ellington, 1993, in The RNA World, ed. Gesteland and Atkins, pp. 511, CSH Laboratory Press). A “nucleic acid aptamer” as used herein is meant to indicate a nucleic acid sequence capable of interacting with a ligand. The ligand can be any natural or a synthetic molecule, including but not limited to a resin, metabolites, nucleosides, nucleotides, drugs, toxins, transition state analogs, peptides, lipids, proteins, amino acids, nucleic acid molecules, hormones, carbohydrates, receptors, cells, viruses, bacteria and others.
  • In yet another embodiment, the non-nucleotide linker X is as defined herein. The term “non-nucleotide” as used herein include either abasic nucleotide, polyether, polyamine, polyamide, peptide, carbohydrate, lipid, or polyhydrocarbon compounds. Specific examples include those described by Seela and Kaiser, [0095] Nucleic Acids Res. 1990, 18:6353 and Nucleic Acids Res. 1987, 15:3113; Cload and Schepartz, J. Am. Chem. Soc. 1991, 113:6324; Richardson and Schepartz, J. Am. Chem. Soc. 1991, 113:5109; Ma et al., Nucleic Acids Res. 1993, 21:2585 and Biochemistry 1993, 32:1751; Durand et al., Nucleic Acids Res. 1990, 18:6353; McCurdy et al., Nucleosides & Nucleotides 1991, 10:287; Jschke et al., Tetrahedron Lett. 1993, 34:301; Ono et al., Biochemistry 1991, 30:9914; Arnold et al., International Publication No. WO 89/02439; Usman et al., International Publication No. WO 95/06731; Dudycz et al., International Publication No. WO 95/11910 and Ferentz and Verdine, J. Am. Chem. Soc. 1991, 113:4000, all hereby incorporated by reference herein. A “non-nucleotide” further means any group or compound that can be incorporated into a nucleic acid chain in the place of one or more nucleotide units, including either sugar and/or phosphate substitutions, and allows the remaining bases to exhibit their enzymatic activity. The group or compound can be abasic in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine. Thus, in a preferred embodiment, the invention features an enzymatic nucleic acid molecule having one or more non-nucleotide moieties, and having enzymatic activity to cleave an RNA or DNA molecule.
  • In another aspect of the invention, enzymatic nucleic acid molecules or antisense molecules that interact with target RNA molecules and down-regulate PTGDS, ADORA1 and/or PTGDR (e.g., PTGDS, ADORA1 and/or PTGDR gene) activity are expressed from transcription units inserted into DNA or RNA vectors. The recombinant vectors can be DNA plasmids or viral vectors. Enzymatic nucleic acid molecule or antisense expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus, retrovirus, adenovirus, or alphavirus. The recombinant vectors capable of expressing the enzymatic nucleic acid molecules or antisense can be delivered as described above, and persist in target cells. Alternatively, viral vectors can be used that provide for transient expression of enzymatic nucleic acid molecules or antisense. Such vectors can be repeatedly administered as necessary. Once expressed, the enzymatic nucleic acid molecules or antisense bind to the target RNA and down-regulate its function or expression. Delivery of enzymatic nucleic acid molecule or antisense expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex-planted from the patient followed by reintroduction into the patient, or by any other means that would allow for introduction into the desired target cell. Antisense DNA can be expressed via the use of a single stranded DNA intracellular expression vector. [0096]
  • By “vectors” is meant any nucleic acid- and/or viral-based technique used to deliver a desired nucleic acid. [0097]
  • By “patient” is meant an organism, which is a donor or recipient of explanted cells, or the cells themselves. “Patient” also refers to an organism to which the nucleic acid molecules of the invention can be administered. A patient can be a mammal or mammalian cells. In one embodiment, a patient is a human or human cells. [0098]
  • By “enhanced enzymatic activity” is meant to include activity measured in cells and/or in vivo where the activity is a reflection of both the catalytic activity and the stability of the nucleic acid molecules of the invention. In this invention, the product of these properties can be increased in vivo compared to an all RNA enzymatic nucleic acid or all DNA enzyme. In some cases, the activity or stability of the nucleic acid molecule can be decreased (i.e., less than ten-fold), but the overall activity of the nucleic acid molecule is enhanced, in vivo. [0099]
  • The nucleic acid molecules of the instant invention, individually, or in combination or in conjunction with other drugs, can be used to treat diseases or conditions discussed above. For example, to treat a disease or condition associated with the levels of PTGDS, ADORA1 and/or PTGDR, the patient can be treated, or other appropriate cells can be treated, as is evident to those skilled in the art, individually or in combination with one or more drugs under conditions suitable for the treatment. [0100]
  • In a further embodiment, the described molecules, such as antisense or enzymatic nucleic acid molecules, can be used in combination with other known treatments to treat conditions or diseases discussed above. For example, the described molecules can be used in combination with one or more known therapeutic agents to treat allergic diseases or conditions, including but not limited to asthma, allergic rhinitis, atopic dermatitis, and/or other allergic or inflammatory diseases and conditions which respond to the modulation of PTGDS, ADORA1 and/or PTGDR expression. [0101]
  • In another embodiment, the invention features nucleic acid-based inhibitors (e.g., enzymatic nucleic acid molecules (e.g., ribozymes), antisense nucleic acids, 2-5A antisense chimeras, triplex DNA, antisense nucleic acids containing RNA cleaving chemical groups) and methods for their use to down regulate or inhibit the expression of genes (e.g., PTGDS, ADORA1 and/or PTGDR) capable of progression and/or maintenance allergic diseases or conditions, including but not limited to asthma, allergic rhinitis, atopic dermatitis, and/or other allergic or inflammatory diseases and conditions which respond to the modulation of PTGDS, ADORA1 and/or PTGDR expression. [0102]
  • By “comprising” is meant including, but not limited to, whatever follows the word “comprising”. Thus, use of the term “comprising” indicates that the listed elements are required or mandatory, but that other elements are optional and may or may not be present. By “consisting of” is meant including, and limited to, whatever follows the phrase “consisting of”. Thus, the phrase “consisting of” indicates that the listed elements are required or mandatory, and that no other elements may be present. [0103]
  • Other features and advantages of the invention will be apparent from the following description of the preferred embodiments thereof, and from the claims.[0104]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows examples of chemically stabilized ribozyme motifs. HH Rz, represents hammerhead ribozyme motif (Usman et al., 1996[0105] , Curr. Op. Struct. Bio., 1, 527); NCH Rz represents the NCH ribozyme motif (Ludwig & Sproat, International PCT Publication No. WO 98/58058); G-Cleaver, represents G-cleaver ribozyme motif (Kore et al., 1998, Nucleic Acids Research 26, 4116-4120, Eckstein et al., International PCT publication No. WO 99/16871). N or n, represent independently a nucleotide that can be same or different and have complementarity to each other; rI, represents ribo-Inosine nucleotide; arrow indicates the site of cleavage within the target. Position 4 of the HH Rz and the NCH Rz is shown as having 2′-C-allyl modification, but those skilled in the art will recognize that this position can be modified with other modifications well known in the art, so long as such modifications do not significantly inhibit the activity of the ribozyme.
  • FIG. 2 shows an example of the Amberzyme ribozyme motif that is chemically stabilized (see for example Beigelman et al., International PCT publication No. WO 99/55857). [0106]
  • FIG. 3 shows an example of the Zinzyme A ribozyme motif that is chemically stabilized (see for example Beigelman et al., Beigelman et al., International PCT publication No. WO 99/55857). [0107]
  • FIG. 4 shows an example of a specific DNAzyme motif, commonly referred to as the “10-23 motif”, as described by Santoro et al., 1997[0108] , PNAS, 94, 4262.
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • Nucleic Acid Molecules and Mechanism of Action [0109]
  • Antisense: Antisense molecules can be modified or unmodified RNA, DNA, or mixed polymer oligonucleotides and primarily function by specifically binding to matching sequences resulting in inhibition of peptide synthesis (Wu-Pong, Nov 1994[0110] , BioPharm, 20-33). The antisense oligonucleotide binds to target RNA by Watson Crick base-pairing and blocks gene expression by preventing ribosomal translation of the bound sequences either by steric blocking or by activating RNase H enzyme. Antisense molecules can also alter protein synthesis by interfering with RNA processing or transport from the nucleus into the cytoplasm (Mukhopadhyay & Roth, 1996, Crit. Rev. in Oncogenesis 7, 151-190).
  • In addition, binding of single stranded DNA to RNA can result in nuclease degradation of the heteroduplex (Wu-Pong, supra; Crooke, supra). To date, the only backbone modified DNA chemistry which act as substrates for RNase H are phosphorothioates, phosphorodithioates, and borontrifluoridates. Recently it has been reported that 2′-arabino and 2′-fluoro arabino-containing oligos can also activate RNase H activity. [0111]
  • A number of antisense molecules have been described that utilize novel configurations of chemically modified nucleotides, secondary structure, and/or RNase H substrate domains (Woolf et al., International PCT Publication No. WO 98/13526; Thompson et al., International PCT Publication No. WO 99/54459; Hartmann et al., U.S. S No. 60/101,174, filed on Sep. 21, 1998) all of these are incorporated by reference herein in their entirety. [0112]
  • In addition, antisense deoxyoligoribonucleotides can be used to target RNA by means of DNA-RNA interactions, thereby activating RNase H, which digests the target RNA in the duplex. Antisense DNA can be expressed via the use of a single stranded DNA intracellular expression vector or equivalents and variations thereof. [0113]
  • Triplex Forming Oligonucleotides (TFO): Single stranded DNA can be designed to bind to genomic DNA in a sequence specific manner. TFOs are comprised of pyrimidine-rich oligonucleotides which bind DNA helices through Hoogsteen Base-pairing (Wu-Pong, supra). The resulting triple helix composed of the DNA sense, DNA antisense, and TFO disrupts RNA synthesis by RNA polymerase. The TFO mechanism can result in gene expression or cell death since binding can be irreversible (Mukhopadhyay & Roth, supra). [0114]
  • 2-5A Antisense Chimera: The 2-5A system is an interferon mediated mechanism for RNA degradation found in higher vertebrates (Mitra et al., 1996[0115] , Proc Nat Acad Sci USA 93, 6780-6785). Two types of enzymes, 2-5A synthetase and RNase L, are required for RNA cleavage. The 2-5A synthetases require double stranded RNA to form 2′-5′ oligoadenylates (2-5A). 2-5A then acts as an allosteric effector for utilizing RNase L, which has the ability to cleave single stranded RNA. The ability to form 2-5A structures with double stranded RNA makes this system particularly useful for inhibition of viral replication.
  • (2′-5′) oligoadenylate structures can be covalently linked to antisense molecules to form chimeric oligonucleotides capable of RNA cleavage (Torrence, supra). These molecules putatively bind and activate a 2-5A dependent RNase, the oligonucleotide/enzyme complex then binds to a target RNA molecule which can then be cleaved by the RNase enzyme. [0116]
  • Enzymatic Nucleic Acid: Several varieties of naturally-occurring enzymatic RNAs are presently known. In addition, several in vitro selection (evolution) strategies (Orgel, 1979, [0117] Proc. R. Soc. London, B 205, 435) have been used to evolve new nucleic acid catalysts capable of catalyzing cleavage and ligation of phosphodiester linkages (Joyce, 1989, Gene, 82, 83-87; Beaudry et al., 1992, Science 257, 635-641; Joyce, 1992, Scientific American 267, 90-97; Breaker et al., 1994, TIBTECH 12, 268; Bartel et al., 1993, Science 261:1411-1418; Szostak, 1993, TIBS 17, 89-93; Kumar et al., 1995, FASEB J, 9, 1183; Breaker, 1996, Curr. Op. Biotech., 7, 442; Santoro et al., 1997, Proc. Natl. Acad. Sci., 94, 4262; Tang et al., 1997, RNA 3, 914; Nakamaye & Eckstein, 1994, supra; Long & Uhlenbeck, 1994, supra; Ishizaka et al., 1995, supra; Vaish et al., 1997, Biochemistry 36, 6495; all of these are incorporated by reference herein). Each can catalyze a series of reactions including the hydrolysis of phosphodiester bonds in trans (and thus can cleave other RNA molecules) under physiological conditions.
  • The enzymatic nature of an enzymatic nucleic acid molecule has significant advantages, one advantage being that the concentration of enzymatic nucleic acid molecule necessary to affect a therapeutic treatment is lower. This advantage reflects the ability of the enzymatic nucleic acid molecule to act enzymatically. Thus, a single enzymatic nucleic acid molecule is able to cleave many molecules of target RNA. In addition, the enzymatic nucleic acid molecule is a highly specific inhibitor, with the specificity of inhibition depending not only on the base-pairing mechanism of binding to the target RNA, but also on the mechanism of target RNA cleavage. Single mismatches, or base-substitutions, near the site of cleavage can be chosen to completely eliminate catalytic activity of a enzymatic nucleic acid molecule. [0118]
  • Nucleic acid molecules having an endonuclease enzymatic activity are able to repeatedly cleave other separate RNA molecules in a nucleotide base sequence-specific manner. With the proper design, such enzymatic nucleic acid molecules can be targeted to RNA transcripts, and achieve efficient cleavage in vitro (Zaug et al., 324[0119] , Nature 429 1986; Uhlenbeck, 1987 Nature 328, 596; Kim et al., 84 Proc. Natl. Acad. Sci. USA 8788, 1987; Dreyfus, 1988, Einstein Quart. J. Bio. Med., 6, 92; Haseloff and Gerlach, 334 Nature 585, 1988; Cech, 260 JAMA 3030, 1988; and Jefferies et al., 17 Nucleic Acids Research 1371, 1989; Santoro et al., 1997 supra).
  • Because of their sequence specificity, trans-cleaving enzymatic nucleic acid molecules can be used as therapeutic agents for human disease (Usman & McSwiggen, 1995 [0120] Ann. Rep. Med. Chem. 30, 285-294; Christoffersen and Marr, 1995 J. Med. Chem. 38, 2023-2037). Enzymatic nucleic acid molecules can be designed to cleave specific RNA targets within the background of cellular RNA. Such a cleavage event renders the RNA non-functional and abrogates protein expression from that RNA. In this manner, synthesis of a protein associated with a disease state can be selectively inhibited (Warashina et al., 1999, Chemistry and Biology, 6, 237-250).
  • Enzymatic nucleic acid molecules of the invention that are allosterically regulated (“allozymes”) can be used to down-regulate PTGDS and/or PTGDR expression. These allosteric enzymatic nucleic acids or allozymes (see for example George et al., U.S. Pat. Nos. 5,834,186 and 5,741,679, Shih et al., U.S. Pat. No. 5,589,332, Nathan et al., U.S. Pat. No. 5,871,914, Nathan and Ellington, International PCT publication No. WO 00/24931, Breaker et al., International PCT Publication Nos. WO 00/26226 and 98/27104, and Sullenger et al., International PCT publication No. WO 99/29842) are designed to respond to a signaling agent, for example, mutant PTGDS and/or PTGDR protein, wild-type PTGDS and/or PTGDR protein, mutant PTGDS and/or PTGDR RNA, wild-type PTGDS and/or PTGDR RNA, other proteins and/or RNAs involved in PTGDS or PTGDR signal transduction, compounds, metals, polymers, molecules and/or drugs that are targeted to PTGDS and/or PTGDR expressing cells etc., which in turn modulates the activity of the enzymatic nucleic acid molecule. In response to interaction with a predetermined signaling agent, the allosteric enzymatic nucleic acid molecule's activity is activated or inhibited such that the expression of a particular target is selectively down-regulated. The target can comprise wild-type PTGDS, ADORA1 and/or PTGDR, mutant PTGDS, ADORA1 and/or PTGDR, and/or a predetermined component of the PTGDS, ADORA1 or PTGDR signal transduction pathway. In a specific example, allosteric enzymatic nucleic acid molecules that are activated by interaction with a RNA encoding a PTGDR protein are used as therapeutic agents in vivo. The presence of RNA encoding the PTGDS protein activates the allosteric enzymatic nucleic acid molecule that subsequently cleaves the RNA encoding a PTGDR protein resulting in the inhibition of PTGDR protein expression. In this manner, cells that express both PTGDS and PTGDR protein are selectively targeted. [0121]
  • In another non-limiting example, an allozyme can be activated by a PTGDS or PTGDR protein, peptide, or mutant polypeptide that causes the allozyme to inhibit the expression of PTGDS or PTGDR gene, by, for example, cleaving RNA encoded by PTGDS or PTGDR gene. In this non-limiting example, the allozyme acts as a decoy to inhibit the function of PTGDS or PTGDR and also inhibit the expression of PTGDS or PTGDR once activated by the PTGDS or PTGDR protein. [0122]
  • Target Sites [0123]
  • Targets for useful enzymatic nucleic acid molecules and antisense nucleic acids can be determined as disclosed in Draper et al., WO 93/23569; Sullivan et al, WO 93/23057; Thompson et al., WO 94/02595; Draper et al., WO 95/04818; McSwiggen et al., U.S. Pat. No. 5,525,468, and hereby incorporated by reference herein in totality. Other examples include the following PCT applications, which concern inactivation of expression of disease-related genes: WO 95/23225, WO 95/13380, WO 94/02595, incorporated by reference herein. Rather than repeat the guidance provided in those documents here, below are provided specific examples of such methods, not limiting to those in the art. Enzymatic nucleic acid molecules and antisense to such targets are designed as described in those applications and synthesized to be tested in vitro and in vivo, as also described. The sequences of human PTGDR RNAs were screened for optimal enzymatic nucleic acid and antisense target sites using a computer-folding algorithm. Antisense, hammerhead, DNAzyme, NCH, amberzyme, zinzyme, or G-Cleaver enzymatic nucleic acid molecule binding/cleavage sites were identified. These sites are shown in Tables III to VII (all sequences are 5′ to 3′ in the tables; underlined regions can be any sequence “X” or linker X, the actual sequence is not relevant here). The nucleotide base position is noted in the Tables as that site to be cleaved by the designated type of enzymatic nucleic acid molecule. While human sequences can be screened and enzymatic nucleic acid molecule and/or antisense thereafter designed, as discussed in Stinchcomb et al., WO 95/23225, mouse targeted enzymatic nucleic acid molecules can be useful to test efficacy of action of the enzymatic nucleic acid molecule and/or antisense prior to testing in humans. [0124]
  • Antisense, hammerhead, DNAzyme, NCH, amberzyme, zinzyme or G-Cleaver enzymatic nucleic acid molecule binding/cleavage sites were identified. The nucleic acid molecules are individually analyzed by computer folding (Jaeger et al., 1989 [0125] Proc. Natl. Acad. Sci. USA, 86, 7706) to assess whether the sequences fold into the appropriate secondary structure. Those nucleic acid molecules with unfavorable intramolecular interactions such as between the binding arms and the catalytic core are eliminated from consideration. Varying binding arm lengths can be chosen to optimize activity.
  • Antisense, hammerhead, DNAzyme, NCH, amberzyme, zinzyme or G-Cleaver enzymatic nucleic acid molecule binding/cleavage sites were identified and were designed to anneal to various sites in the RNA target. The binding arms are complementary to the target site sequences described above. The nucleic acid molecules were chemically synthesized. The method of synthesis used follows the procedure for normal DNA/RNA synthesis as described below and in Usman et al., 1987 [0126] J. Am. Chem. Soc., 109, 7845; Scaringe et al., 1990 Nucleic Acids Res., 18, 5433; and Wincott et al., 1995 Nucleic Acids Res. 23, 2677-2684; Caruthers et al., 1992, Methods in Enzymology 211,3-19.
  • Synthesis of Nucleic acid Molecules [0127]
  • Synthesis of nucleic acids greater than 100 nucleotides in length is difficult using automated methods, and the therapeutic cost of such molecules is prohibitive. In this invention, small nucleic acid motifs (“small refers to nucleic acid motifs less than about 100 nucleotides in length, and in one embodiment less than about 80 nucleotides in length, and in another embodiment less than about 50 nucleotides in length; e.g., antisense oligonucleotides, hammerhead or the NCH ribozymes) can be used for exogenous delivery. The simple structure of these molecules increases the ability of the nucleic acid to invade targeted regions of RNA structure. Exemplary molecules of the instant invention are chemically synthesized, and others can similarly be synthesized. [0128]
  • Oligonucleotides (e.g., antisense GeneBlocs) are synthesized using protocols known in the art as described in Caruthers et al., 1992[0129] , Methods in Enzymology 211, 3-19, Thompson et al., International PCT Publication No. WO 99/54459, Wincott et al., 1995, Nucleic Acids Res. 23, 2677-2684, Wincott et al., 1997, Methods Mol. Bio., 74, 59, Brennan et al., 1998, Biotechnol Bioeng., 61, 33-45, and Brennan, U.S. Pat. No. 6,001,311. All of these references are incorporated herein by reference. The synthesis of oligonucleotides makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5′-end, and phosphoramidites at the 3′-end. In a non-limiting example, small scale syntheses are conducted on a 394 Applied Biosystems, Inc. synthesizer using a 0.2 μmol scale protocol with a 2.5 min coupling step for 2′-O-methylated nucleotides and a 45 sec coupling step for 2′-deoxy nucleotides. Table II outlines the amounts and the contact times of the reagents used in the synthesis cycle. Alternatively, syntheses at the 0.2 μmol scale can be performed on a 96-well plate synthesizer, such as the instrument produced by Protogene (Palo Alto, Calif.) with minimal modification to the cycle. A 33-fold excess (60 μL of 0.11 M=6.6 μmol) of 2′-O-methyl phosphoramidite and a 105-fold excess of S-ethyl tetrazole (60 μL of 0.25 M=15 μmol) can be used in each coupling cycle of 2′-O-methyl residues relative to polymer-bound 5′-hydroxyl. A 22-fold excess (40 μL of 0.11 M=4.4 μmol) of deoxy phosphoramidite and a 70-fold excess of S-ethyl tetrazole (40 μL of 0.25 M=10 μmol) can be used in each coupling cycle of deoxy residues relative to polymer-bound 5′-hydroxyl. Average coupling yields on the 394 Applied Biosystems, Inc. synthesizer, determined by calorimetric quantitation of the trityl fractions, are typically 97.5-99%. Other oligonucleotide synthesis reagents for the 394 Applied Biosystems, Inc. synthesizer include; detritylation solution is 3% TCA in methylene chloride (ABI); capping is performed with 16% N-methylimidazole in THF (ABI) and 10% acetic anhydride/10% 2,6-lutidine in THF (ABI); and oxidation solution is 16.9 mM 12, 49 mM pyridine, 9% water in THF (PERSEPTIVE™). Burdick & Jackson Synthesis Grade acetonitrile is used directly from the reagent bottle. S-Ethyltetrazole solution (0.25 M in acetonitrile) is made up from the solid obtained from American International Chemical, Inc. Alternately, for the introduction of phosphorothioate linkages, Beaucage reagent (3H-1,2-Benzodithiol-3-one 1,1-dioxide, 0.05 M in acetonitrile) is used.
  • Deprotection of the antisense oligonucleotides is performed as follows: the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 40% aq. methylamine (1 mL) at 65° C. for 10 min. After cooling to −20° C., the supernatant is removed from the polymer support. The support is washed three times with 1.0 mL of EtOH:MeCN:H[0130] 2O/3:1:1, vortexed and the supernatant is then added to the first supernatant. The combined supernatants, containing the oligoribonucleotide, are dried to a white powder.
  • The method of synthesis used for normal RNA including certain enzymatic nucleic acid molecules follows the procedure as described in Usman et al., 1987, [0131] J. Am. Chem. Soc., 109, 7845; Scaringe et al., 1990, Nucleic Acids Res., 18, 5433; and Wincott et al., 1995, Nucleic Acids Res. 23, 2677-2684 Wincott et al., 1997, Methods Mol. Bio., 74, 59, and makes use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5′-end, and phosphoramidites at the 3′-end. In a non-limiting example, small scale syntheses are conducted on a 394 Applied Biosystems, Inc. synthesizer using a 0.2 μmol scale protocol with a 7.5 min coupling step for alkylsilyl protected nucleotides and a 2.5 min coupling step for 2′-O-methylated nucleotides. Table II outlines the amounts and the contact times of the reagents used in the synthesis cycle. Alternatively, syntheses at the 0.2 μmol scale can be done on a 96-well plate synthesizer, such as the instrument produced by Protogene (Palo Alto, Calif.) with minimal modification to the cycle. A 33-fold excess (60 μL of 0.11 M=6.6 μmol) of 2′-O-methyl phosphoramidite and a 75-fold excess of S-ethyl tetrazole (60 μL of 0.25 M=15 μmol) can be used in each coupling cycle of 2′-O-methyl residues relative to polymer-bound 5′-hydroxyl. A 66-fold excess (120 μL of 0.11 M=13.2 μmol) of alkylsilyl (ribo) protected phosphoramidite and a 150-fold excess of S-ethyl tetrazole (120 μL of 0.25 M=30 μmol) can be used in each coupling cycle of ribo residues relative to polymer-bound 5′-hydroxyl. Average coupling yields on the 394 Applied Biosystems, Inc. synthesizer, determined by colorimetric quantitation of the trityl fractions, are typically 97.5-99%. Other oligonucleotide synthesis reagents for the 394 Applied Biosystems, Inc. synthesizer include; detritylation solution is 3% TCA in methylene chloride (ABI); capping is performed with 16% N-methylimidazole in THF (ABI) and 10% acetic anhydride/10% 2,6-lutidine in THF (ABI); oxidation solution is 16.9 mM 12, 49 mM pyridine, 9% water in THF (PERSEPTIVE™). Burdick & Jackson Synthesis Grade acetonitrile is used directly from the reagent bottle. S-Ethyltetrazole solution (0.25 M in acetonitrile) is made up from the solid obtained from American International Chemical, Inc. Alternately, for the introduction of phosphorothioate linkages, Beaucage reagent (3H-1,2-Benzodithiol-3-one 1,1-dioxide 0.05 M in acetonitrile) is used.
  • Deprotection of the RNA is performed using either a two-pot or one-pot protocol. For the two-pot protocol, the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 40% aq. methylamine (1 mL) at 65° C. for 10 min. After cooling to −20° C., the supernatant is removed from the polymer support. The support is washed three times with 1.0 mL of EtOH:MeCN:H2O/3:1:1, vortexed and the supernatant is then added to the first supernatant. The combined supernatants, containing the oligoribonucleotide, are dried to a white powder. The base deprotected oligoribonucleotide is resuspended in anhydrous TEA/HF/NMP solution (300 μL of a solution of 1.5 mL N-methylpyrrolidinone, 750 μL TEA and 1 mL TEA.3HF to provide a 1.4 M HF concentration) and heated to 65° C. After 1.5 h, the oligomer is quenched with 1.5 M NH[0132] 4HCO3.
  • Alternatively, for the one-pot protocol, the polymer-bound trityl-on oligoribonucleotide is transferred to a 4 mL glass screw top vial and suspended in a solution of 33% ethanolic methylamine/DMSO: 1/1 (0.8 mL) at 65° C. for 15 min. The vial is brought to r.t. TEA.3HF (0.1 mL) is added and the vial is heated at 65° C. for 15 min. The sample is cooled at −20° C. and then quenched with 1.5 M NH[0133] 4HCO3.
  • For purification of the trityl-on oligomers, the quenched NH[0134] 4HCO3 solution is loaded onto a C-18 containing cartridge that had been prewashed with acetonitrile followed by 50 mM TEAA. After washing, the loaded cartridge with water, the RNA is detritylated with 0.5% TFA for 13 min. The cartridge is then washed again with water, salt exchanged with 1 M NaCl and washed with water again. The oligonucleotide is then eluted with 30% acetonitrile.
  • Inactive hammerhead ribozymes or binding attenuated control (BAC) oligonucleotides are synthesized by substituting a U for G[0135] 5 and a U for A14 (numbering from Hertel, K. J., et al., 1992, Nucleic Acids Res., 20, 3252). Similarly, one or more nucleotide substitutions can be introduced in other enzymatic nucleic acid molecules to inactivate the molecule and such molecules can serve as a negative control.
  • The average stepwise coupling yields are typically >98% (Wincott et al., 1995 [0136] Nucleic Acids Res. 23, 2677-2684). Those of ordinary skill in the art will recognize that the scale of synthesis can be adapted to be larger or smaller than the example described above including but not limited to 96 well format, all that is important is the ratio of chemicals used in the reaction.
  • Alternatively, the nucleic acid molecules of the present invention can be synthesized separately and joined together post-synthetically, for example by ligation (Moore et al., 1992[0137] , Science 256, 9923; Draper et al., International PCT publication No. WO 93/23569; Shabarova et al., 1991, Nucleic Acids Research 19, 4247; Bellon et al., 1997, Nucleosides & Nucleotides, 16, 951; Bellon et al., 1997, Bioconjugate Chem. 8, 204).
  • The nucleic acid molecules of the present invention can be modified extensively to enhance stability by modification with nuclease resistant groups, for example, 2′-amino, 2′-C-allyl, 2′-flouro, 2′-O-methyl, 2′-H (for a review see Usman and Cedergren, 1992[0138] , TIBS 17, 34; Usman et al., 1994, Nucleic Acids Symp. Ser. 31, 163). Ribozymes are purified by gel electrophoresis using general methods or are purified by high pressure liquid chromatography (HPLC; See Wincott et al., Supra, the totality of which is hereby incorporated herein by reference) and are re-suspended in water.
  • The sequences of the nucleic acid molecules, including enzymatic nucleic acid molecules and antisense, that are chemically synthesized, are shown in Tables III-VII. The sequences of the enzymatic nucleic acid constructs that are chemically synthesized are complementary to the Substrate sequences shown in Tables III-VII. Those in the art will recognize that these sequences are representative only of many more such sequences where the enzymatic portion of the enzymatic nucleic acid (all but the binding arms) is altered to affect activity. The enzymatic nucleic acid construct sequences listed in Tables III-VII can be formed of ribonucleotides or other nucleotides or non-nucleotides. Such enzymatic nucleic acid molecules with enzymatic activity are equivalent to the enzymatic nucleic acid molecules described specifically in the Tables. [0139]
  • Optimizing Activity of the Nucleic Acid Molecule of the Invention. [0140]
  • Chemically synthesizing nucleic acid molecules with modifications (base, sugar and/or phosphate) that prevent their degradation by serum ribonucleases can increase their potency (see e.g., Eckstein et al., International Publication No. WO 92/07065; Perrault et al., 1990 [0141] Nature 344, 565; Pieken et al., 1991, Science 253, 314; Usman and Cedergren, 1992, Trends in Biochem. Sci. 17, 334; Usman et al., International Publication No. WO 93/15187; and Rossi et al., International Publication No. WO 91/03162; Sproat, U.S. Pat. No. 5,334,711; and Burgin et al., supra; all of these describe various chemical modifications that can be made to the base, phosphate and/or sugar moieties of the nucleic acid molecules herein). Modifications that enhance their efficacy in cells, and removal of bases from nucleic acid molecules to shorten oligonucleotide synthesis times and reduce chemical requirements are desired. (All these publications are hereby incorporated by reference herein).
  • There are several examples in the art describing sugar, base and phosphate modifications that can be introduced into nucleic acid molecules with significant enhancement in their nuclease stability and efficacy. For example, oligonucleotides are modified to enhance stability and/or enhance biological activity by modification with nuclease resistant groups, for example, 2′-amino, 2′-C-allyl, 2′-flouro, 2′-O-methyl, 2′-H, nucleotide base modifications (for a review see Usman and Cedergren, 1992, TIBS. 17, 34; Usman et al., 1994[0142] , Nucleic Acids Symp. Ser. 31, 163; Burgin et al., 1996, Biochemistry, 35, 14090). Sugar modification of nucleic acid molecules have been extensively described in the art (see Eckstein et al., International Publication PCT No. WO 92/07065; Perrault et al. Nature, 1990, 344, 565-568; Pieken et al. Science, 1991, 253, 314-317; Usman and Cedergren, Trends in Biochem. Sci., 1992, 17, 334-339; Usman et al. International Publication PCT No. WO 93/15187; Sproat, U.S. Pat. No. 5,334,711 and Beigelman et al., 1995, J. Biol. Chem., 270, 25702; Beigelman et al., International PCT publication No. WO 97/26270; Beigelman et al., U.S. Pat. No. 5,716,824; Usman et al., U.S. Pat. No. 5,627,053; Woolf et al., International PCT Publication No. WO 98/13526; Thompson et al., U.S. S No. 60/082,404 which was filed on Apr. 20, 1998; Karpeisky et al., 1998, Tetrahedron Lett., 39, 1131; Earnshaw and Gait, 1998, Biopolymers (Nucleic acid Sciences), 48, 39-55; Verma and Eckstein, 1998, Annu. Rev. Biochem., 67, 99-134; and Burlina et al., 1997, Bioorg. Med. Chem., 5, 1999-2010; all of the references are hereby incorporated in their totality by reference herein). Such publications describe general methods and strategies to determine the location of incorporation of sugar, base and/or phosphate modifications and the like into ribozymes without inhibiting catalysis, and are incorporated by reference herein. In view of such teachings, similar modifications can be used as described herein to modify the nucleic acid molecules of the instant invention.
  • While chemical modification of oligonucleotide internucleotide linkages with phosphorothioate, phosphorothioate, and/or 5′-methylphosphonate linkages improves stability, too many of these modifications can cause some toxicity. Therefore when designing nucleic acid molecules the amount of these internucleotide linkages should be minimized. The reduction in the concentration of these linkages should lower toxicity resulting in increased efficacy and higher specificity of these molecules. [0143]
  • Nucleic acid molecules having chemical modifications that maintain or enhance activity are provided. Such a nucleic acid is also generally more resistant to nucleases than an unmodified nucleic acid. Thus, in a cell and/or in vivo the activity may not be significantly lowered. Therapeutic nucleic acid molecules delivered exogenously are optimally stable within cells until translation of the target RNA has been inhibited long enough to reduce the levels of the undesirable protein. This period of time varies between hours to days depending upon the disease state. Clearly, nucleic acid molecules must be resistant to nucleases in order to function as effective intracellular therapeutic agents. Improvements in the chemical synthesis of RNA and DNA (Wincott et al., 1995 [0144] Nucleic Acids Res. 23, 2677; Caruthers et al., 1992, Methods in Enzymology 211,3-19 (incorporated by reference herein) have expanded the ability to modify nucleic acid molecules by introducing nucleotide modifications to enhance their nuclease stability as described above.
  • In one embodiment, nucleic acid molecules of the invention include one or more G-clamp nucleotides. A G-clamp nucleotide is a modified cytosine analog wherein modifications result in the ability to hydrogen bond both Watson-Crick and Hoogsteen faces of a complementary guanine within a duplex, see for example Lin and Matteucci, 1998[0145] , J. Am. Chem. Soc., 120, 8531-8532. A single G-clamp analog substation within an oligonucleotide can result in substantially enhanced helical thermal stability and mismatch discrimination when hybridized to complementary oligonucleotides. The inclusion of such nucleotides in nucleic acid molecules of the invention can enable both enhanced affinity and specificity to nucleic acid targets.
  • Therapeutic nucleic acid molecules (e.g., enzymatic nucleic acid molecules and antisense nucleic acid molecules) delivered exogenously are optimally stable within cells until translation of the target RNA has been inhibited long enough to reduce the levels of the undesirable protein. This period of time varies between hours to days depending upon the disease state. These nucleic acid molecules should be resistant to nucleases in order to function as effective intracellular therapeutic agents. Improvements in the chemical synthesis of nucleic acid molecules described in the instant invention and in the art have expanded the ability to modify nucleic acid molecules by introducing nucleotide modifications to enhance their nuclease stability as described above. [0146]
  • In another embodiment, the invention features conjugates and/or complexes of nucleic acid molecules targeting PTGDS, PTGDR, and/or adenosine receptors. Compositions and conjugates are used to facilitate delivery of molecules into a biological system, such as cells. The conjugates provided by the instant invention can impart therapeutic activity by transferring therapeutic compounds across cellular membranes, altering the pharmacokinetics, and/or modulating the localization of nucleic acid molecules of the invention. The present invention encompasses the design and synthesis of novel agents for the delivery of molecules, including but not limited to small molecules, lipids, phospholipids, nucleosides, nucleotides, nucleic acids, antibodies, toxins, negatively charged polymers and other polymers, for example proteins, peptides, hormones, carbohydrates, polyethylene glycols, or polyamines, across cellular membranes. In general, the transporters described are designed to be used either individually or as part of a multi-component system, with or without degradable linkers. These compounds are expected to improve delivery and/or localization of nucleic acid molecules of the invention into a number of cell types originating from different tissues, in the presence or absence of serum (see Sullenger and Cech, U.S. Pat. No. 5,854,038). Conjugates of the molecules described herein can be attached to biologically active molecules via linkers that are biodegradable, such as biodegradable nucleic acid linker molecules. [0147]
  • The term “biodegradable nucleic acid linker molecule” as used herein, refers to a nucleic acid molecule that is designed as a biodegradable linker to connect one molecule to another molecule, for example, a biologically active molecule. The stability of the biodegradable nucleic acid linker molecule can be modulated by using various combinations of ribonucleotides, deoxyribonucleotides, and chemically modified nucleotides, for example 2′-O-methyl, 2′-fluoro, 2′-amino, 2′-O-amino, 2′-C-allyl, 2′-O-allyl, and other 2′-modified or base modified nucleotides. The biodegradable nucleic acid linker molecule can be a dimer, trimer, tetramer or longer nucleic acid molecule, for example an oligonucleotide of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides in length, or can comprise a single nucleotide with a phosphorus based linkage, for example a phosphoramidate or phosphodiester linkage. The biodegradable nucleic acid linker molecule can also comprise nucleic acid backbone, nucleic acid sugar, or nucleic acid base modifications. [0148]
  • The term “biodegradable” as used herein, refers to degradation in a biological system, for example enzymatic degradation or chemical degradation. [0149]
  • The term “biologically active molecule” as used herein, refers to compounds or molecules that are capable of eliciting or modifying a biological response in a system. Non-limiting examples of biologically active molecules contemplated by the instant invention include therapeutically active molecules such as antibodies, hormones, antivirals, peptides, proteins, chemotherapeutics, small molecules, vitamins, co-factors, nucleosides, nucleotides, oligonucleotides, enzymatic nucleic acids, antisense nucleic acids, triplex forming oligonucleotides, 2,5-A chimeras, siRNA, dsRNA, allozymes, aptamers, decoys and analogs thereof. Biologically active molecules of the invention also include molecules capable of modulating the pharmacokinetics and/or pharmacodynamics of other biologically active molecules, for example lipids and polymers such as polyamines, polyamides, polyethylene glycol and other polyethers. [0150]
  • The term “phospholipid” as used herein, refers to a hydrophobic molecule comprising at least one phosphorus group. For example, a phospholipid can comprise a phosphorus containing group and saturated or unsaturated alkyl group, optionally substituted with OH, COOH, oxo, amine, or substituted or unsubstituted aryl groups. [0151]
  • In another embodiment, nucleic acid catalysts having chemical modifications that maintain or enhance enzymatic activity are provided. Such nucleic acids are also generally more resistant to nucleases than unmodified nucleic acid. Thus, in a cell and/or in vivo the activity of the nucleic acid may not be significantly lowered. As exemplified herein such enzymatic nucleic acids are useful in a cell and/or in vivo even if activity over all is reduced 10 fold (Burgin et al., 1996[0152] , Biochemistry, 35, 14090). Such enzymatic nucleic acids herein are said to “maintain” the enzymatic activity of an all RNA ribozyme or all DNA DNAzyme.
  • In another aspect the nucleic acid molecules comprise a 5′ and/or a 3′-cap structure. [0153]
  • By “cap structure” is meant chemical modifications, which have been incorporated at either terminus of the oligonucleotide (see for example Wincott et al., WO 97/26270, incorporated by reference herein). These terminal modifications protect the nucleic acid molecule from exonuclease degradation, and can help in delivery and/or localization within a cell. The cap can be present at the 5′-terminus (5′-cap) or at the 3′-terminus (3′-cap) or can be present on both terminus. In non-limiting examples, the 5′-cap includes inverted abasic residue (moiety), 4′,5′-methylene nucleotide; 1-(beta-D-erythrofuranosyl) nucleotide, 4′-thio nucleotide, carbocyclic nucleotide; 1,5-anhydrohexitol nucleotide; L-nucleotides; alpha-nucleotides; modified base nucleotide; phosphorodithioate linkage; threo-pentofuranosyl nucleotide; acyclic 3′,4′-seco nucleotide; acyclic 3,4-dihydroxybutyl nucleotide; acyclic 3,5-dihydroxypentyl nucleotide, 3′-3′-inverted nucleotide moiety; 3′-3′-inverted abasic moiety; 3′-2′-inverted nucleotide moiety; 3′-2′-inverted abasic moiety; 1,4-butanediol phosphate; 3′-phosphoramidate; hexylphosphate; aminohexyl phosphate; 3′-phosphate; 3′-phosphorothioate; phosphorodithioate; or bridging or non-bridging methylphosphonate moiety (for more details see Wincott et al., International PCT publication No. WO 97/26270, incorporated by reference herein). [0154]
  • In another embodiment the 3′-cap includes, for example 4′,5′-methylene nucleotide; 1-(beta-D-erythrofuranosyl) nucleotide; 4′-thio nucleotide, carbocyclic nucleotide; 5′-amino-alkyl phosphate; 1,3-diamino-2-propyl phosphate, 3-aminopropyl phosphate; 6-aminohexyl phosphate; 1,2-aminododecyl phosphate; hydroxypropyl phosphate; 1,5-anhydrohexitol nucleotide; L-nucleotide; alpha-nucleotide; modified base nucleotide; phosphorodithioate; threo-pentofuranosyl nucleotide; acyclic 3′,4′-seco nucleotide; 3,4-dihydroxybutyl nucleotide; 3,5-dihydroxypentyl nucleotide, 5′-5′-inverted nucleotide moiety; 5′-5′-inverted abasic moiety; 5′-phosphoramidate; 5′-phosphorothioate; 1,4-butanediol phosphate; 5′-amino; bridging and/or [0155] non-bridging 5′-phosphoramidate, phosphorothioate and/or phosphorodithioate, bridging or non bridging methylphosphonate and 5′-mercapto moieties (for more details see Beaucage and Iyer, 1993, Tetrahedron 49, 1925; incorporated by reference herein).
  • By the term “non-nucleotide” is meant any group or compound thatcan be incorporated into a nucleic acid chain in the place of one or more nucleotide units, including either sugar and/or phosphate substitutions, and allows the remaining bases to exhibit their enzymatic activity. The group or compound is abasic in that it does not contain a commonly recognized nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine. [0156]
  • An “alkyl” group refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain, and cyclic alkyl groups. The alkyl group can have, for example, 1 to 12 carbons. In one embodiment of the invention, the alkyl group is a lower alkyl of from 1 to 7 carbons. In another embodiment the alkyl group is 1 to 4 carbons. The alkyl group can be substituted or unsubstituted. When substituted the substituted group(s) can be hydroxyl, cyano, alkoxy, ═O, ═S, NO[0157] 2 or N(CH3)2, amino, or SH. The term also includes alkenyl groups which are unsaturated hydrocarbon groups containing at least one carbon-carbon double bond, including straight-chain, branched-chain, and cyclic groups. The alkenyl group can have, for example, 1 to 12 carbons. In one embodiment of the invention the alkenyl group can be a lower alkenyl of from 1 to 7 carbons. In another embodiment the alkenyl group can be 1 to 4 carbons. The alkenyl group can be substituted or unsubstituted. When substituted the substituted group(s) can be, for example, hydroxyl, cyano, alkoxy, ═O, ═S, NO2, halogen, N(CH3)2, amino, or SH. The term “alkyl” also includes alkynyl groups which have an unsaturated hydrocarbon group containing at least one carbon-carbon triple bond, including straight-chain, branched-chain, and cyclic groups. The alkynyl group can have, for example, 1 to 12 carbons. In one embodiment of the invention, the alkynyl group is a lower alkynyl of from 1 to 7 carbons. In another embodiment of the invention, the alkynyl group is 1 to 4 carbons. The alkynyl group can be substituted or unsubstituted. When substituted the substituted group(s) can be, for example, hydroxyl, cyano, alkoxy, ═O, ═S, NO2 or N(CH3)2, amino or SH.
  • Such alkyl groups can also include aryl, alkylaryl, carbocyclic aryl, heterocyclic aryl, amide and ester groups. An “aryl” group refers to an aromatic group which has at least one ring having a conjugated p electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which can be optionally substituted. The preferred substituent(s) of aryl groups are halogen, trihalomethyl, hydroxyl, SH, OH, cyano, alkoxy, alkyl, alkenyl, alkynyl, and amino groups. An “alkylaryl” group refers to an alkyl group (as described above) covalently joined to an aryl group (as described above). Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are all carbon atoms. The carbon atoms are optionally substituted. Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring and the remainder of the ring atoms are carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen, and include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolo, pyrimidyl, pyrazinyl, imidazolyl and the like, all optionally substituted. An “amide” refers to an —C(O)—NH—R, where R is either alkyl, aryl, alkylaryl or hydrogen. An “ester” refers to an —C(O)—OR′, where R is either alkyl, aryl, alkylaryl or hydrogen. [0158]
  • By “nucleotide” is meant a heterocyclic nitrogenous base in N-glycosidic linkage with a phosphorylated sugar. Nucleotides are recognized in the art to include natural bases (standard), and modified bases well known in the art. Such bases are generally located at the 1′ position of a nucleotide sugar moiety. Nucleotides generally comprise a base, sugar and a phosphate group. The nucleotides can be unmodified or modified at the sugar, phosphate and/or base moiety, (also referred to interchangeably as nucleotide analogs, modified nucleotides, non-natural nucleotides, non-standard nucleotides and other; see for example, Usman and McSwiggen, supra; Eckstein et al., International PCT Publication No. WO 92/07065; Usman et al., International PCT Publication No. WO 93/15187; Uhlman & Peyman, supra all are hereby incorporated by reference herein). There are several examples of modified nucleic acid bases known in the art as summarized by Limbach et al., 1994, Nucleic Acids Res. 22, 2183. Some of the non-limiting examples of chemically modified and other natural nucleic acid bases that can be introduced into nucleic acids include, for example, inosine, purine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2, 4, 6-trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidines (e.g., 5-methylcytidine), 5-alkyluridines (e.g., ribothymidine), 5-halouridine (e.g., 5-bromouridine) or 6-azapyrimidines or 6-alkylpyrimidines (e.g. 6-methyluridine), propyne, quesosine, 2-thiouridine, 4-thiouridine, wybutosine, wybutoxosine, 4-acetylcytidine, 5-(carboxyhydroxymethyl)uridine, 5′-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluridine, beta-D-galactosylqueosine, 1-methyladenosine, 1-methylinosine, 2,2-dimethylguanosine, 3-methylcytidine, 2-methyladenosine, 2-methylguanosine, N6-methyladenosine, 7-methylguanosine, 5-methoxyaminomethyl-2-thiouridine, 5-methylaminomethyluridine, 5-methylcarbonylmethyluridine, 5-methyloxyuridine, 5-methyl-2-thiouridine, 2-methylthio-N6-isopentenyladenosine, beta-D-mannosylqueosine, uridine-5-oxyacetic acid, 2-thiocytidine, threonine derivatives and others (Burgin et al., 1996, Biochemistry, 35, 14090; Uhlman & Peyman, supra). By “modified bases” in this aspect is meant nucleotide bases other than adenine, guanine, cytosine and uracil at 1′ position or their equivalents; such bases can be used at any position, for example, within the catalytic core of an enzymatic nucleic acid molecule and/or in the substrate-binding regions of the nucleic acid molecule. [0159]
  • By “nucleoside” is meant a heterocyclic nitrogenous base in N-glycosidic linkage with a sugar. Nucleosides are recognized in the art to include natural bases (standard), and modified bases well known in the art. Such bases are generally located at the 1′ position of a nucleoside sugar moiety. Nucleosides generally comprise a base and sugar group. The nucleosides can be unmodified or modified at the sugar, and/or base moiety, (also referred to interchangeably as nucleoside analogs, modified nucleosides, non-natural nucleosides, non-standard nucleosides and other; see for example, Usman and McSwiggen, supra; Eckstein et al., International PCT Publication No. WO 92/07065; Usman et al., International PCT Publication No. WO 93/15187; Uhlman & Peyman, supra all are hereby incorporated by reference herein). There are several examples of modified nucleic acid bases known in the art as summarized by Limbach et al., 1994, Nucleic Acids Res. 22, 2183. Some of the non-limiting examples of chemically modified and other natural nucleic acid bases that can be introduced into nucleic acids include, inosine, purine, pyridin-4-one, pyridin-2-one, phenyl, pseudouracil, 2, 4, 6-trimethoxy benzene, 3-methyl uracil, dihydrouridine, naphthyl, aminophenyl, 5-alkylcytidines (e.g., 5-methylcytidine), 5-alkyluridines (e.g., ribothymidine), 5-halouridine (e.g., 5-bromouridine) or 6-azapyrimidines or 6-alkylpyrimidines (e.g. 6-methyluridine), propyne, quesosine, 2-thiouridine, 4-thiouridine, wybutosine, wybutoxosine, 4-acetylcytidine, 5-(carboxyhydroxymethyl)uridine, 5′-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluridine, beta-D-galactosylqueosine, 1-methyladenosine, 1-methylinosine, 2,2-dimethylguanosine, 3-methylcytidine, 2-methyladenosine, 2-methylguanosine, N6-methyladenosine, 7-methylguanosine, 5-methoxyaminomethyl-2-thiouridine, 5-methylaminomethyluridine, 5-methylcarbonylmethyluridine, 5-methyloxyuridine, 5-methyl-2-thiouridine, 2-methylthio-N6-isopentenyladenosine, beta-D-mannosylqueosine, uridine-5-oxyacetic acid, 2-thiocytidine, threonine derivatives and others (Burgin et al., 1996, Biochemistry, 35, 14090; Uhlman & Peyman, supra). By “modified bases” in this aspect is meant nucleoside bases other than adenine, guanine, cytosine and uracil at 1′ position or their equivalents; such bases can be used at any position, for example, within the catalytic core of an enzymatic nucleic acid molecule and/or in the substrate-binding regions of the nucleic acid molecule. [0160]
  • In one embodiment, the invention features modified enzymatic nucleic acid molecules with phosphate backbone modifications comprising one or more phosphorothioate, phosphorodithioate, methylphosphonate, morpholino, amidate carbamate, carboxymethyl, acetamidate, polyamide, sulfonate, sulfonamide, sulfamate, formacetal, thioformacetal, and/or alkylsilyl, substitutions. For a review of oligonucleotide backbone modifications see Hunziker and Leumann, 1995[0161] , Nucleic Acid Analogues: Synthesis and Properties, in Modern Synthetic Methods, VCH, 331-417, and Mesmaeker et al., 1994, Novel Backbone Replacements for Oligonucleotides, in Carbohydrate Modifications in Antisense Research, ACS, 24-39. These references are hereby incorporated by reference herein.
  • By “abasic” is meant sugar moieties lacking a base or having other chemical groups in place of a base at the 1′ position, for example a 3′,3′-linked or 5′,5′-linked deoxyabasic ribose derivative (for more details see Wincott et al., International PCT publication No. WO 97/26270). [0162]
  • By “unmodified nucleoside” is meant one of the bases adenine, cytosine, guanine, thymine, uracil joined to the 1′ carbon of β-D-ribo-furanose. [0163]
  • By “modified nucleoside” is meant any nucleotide base that contains a modification in the chemical structure of an unmodified nucleotide base, sugar and/or phosphate. In connection with 2′-modified nucleotides as described for the present invention, by “amino” is meant 2′-NH[0164] 2 or 2′-O-NH2, which can be modified or unmodified. Such modified groups are described, for example, in Eckstein et al., U.S. Pat. No. 5,672,695 and Matulic-Adamic et al., WO 98/28317, respectively, which are both incorporated by reference in their entireties.
  • Various modifications to nucleic acid (e.g., antisense and ribozyme) structure can be made to enhance the utility of these molecules. For example, such modifications can enhance shelf-life, half-life in vitro, stability, and ease of introduction of such oligonucleotides to the target site, including e.g., enhancing penetration of cellular membranes and conferring the ability to recognize and bind to targeted cells. [0165]
  • Use of the nucleic acid-based molecules of the invention can lead to better treatment of the disease progression by affording the possibility of combination therapies (e.g., multiple enzymatic nucleic acid molecules targeted to different genes, enzymatic nucleic acid molecules coupled with known small molecule inhibitors, or intermittent treatment with combinations of enzymatic nucleic acid molecules (including different enzymatic nucleic acid molecule motifs) and/or other chemical or biological molecules). The treatment of patients with nucleic acid molecules can also include combinations of different types of nucleic acid molecules. Therapies can be devised which include a mixture of enzymatic nucleic acid molecules (including different enzymatic nucleic acid molecule motifs), antisense and/or 2-5A chimera molecules to one or more targets to alleviate symptoms of a disease. [0166]
  • Administration of Nucleic Acid Molecules [0167]
  • A nucleid acid molecule of the invention can be adapted for use to treat asthma and other related diseases and conditions described herein. For example, a nucleic acid molecule can comprise a delivery vehicle, including liposomes, for administration to a subject, carriers and diluents and their salts, and/or can be present in pharmaceutically acceptable formulations. Methods for the delivery of nucleic acid molecules are described in Akhtar et al., 1992[0168] , Trends Cell Bio., 2, 139; and Delivery Strategies for Antisense Oligonucleotide Therapeutics, ed. Akhtar, 1995 which are both incorporated herein by reference. Sullivan et al., PCT WO 94/02595, further describes the general methods for delivery of enzymatic RNA molecules. These protocols can be utilized for the delivery of virtually any nucleic acid molecule. Nucleic acid molecules can be administered to cells by a variety of methods known to those familiar to the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres. The nucleic acid molecules or the invention are administered via pulmonary delivery, such as by inhalation of an aerosol or spray dried formulation administered by an inhalation device or nebulizer. Alternatively, the nucleic acid/vehicle combination is locally delivered by direct injection or by use of an infusion pump. Other routes of delivery include, but are not limited to oral (tablet or pill form) and/or intrathecal delivery (Gold, 1997, Neuroscience, 76, 1153-1158). Other approaches include the use of various transport and carrier systems, for example though the use of conjugates and biodegradable polymers. For a comprehensive review on drug delivery strategies including CNS delivery, see Ho et al., 1999, Curr. Opin. Mol. Ther., 1, 336-343 and Jain, Drug Delivery Systems: Technologies and Commercial Opportunities, Decision Resources, 1998 and Groothuis et al., 1997, J NeuroVirol., 3, 387-400. More detailed descriptions of nucleic acid delivery and administration are provided in Sullivan et al., supra, Draper et al., PCT WO93/23569, Beigelman et al., PCT WO99/05094, and Klimuk et al., PCT WO99/04819 all of which have been incorporated by reference herein.
  • The molecules of the instant invention can be used as pharmaceutical agents. Pharmaceutical agents prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, or all of the symptoms) of a disease state in a patient. [0169]
  • The negatively charged polynucleotides of the invention can be administered (e.g., RNA, DNA or protein) and introduced into a patient by any standard means, with or without stabilizers, buffers, and the like, to form a pharmaceutical composition. When it is desired to use a liposome delivery mechanism, standard protocols for formation of liposomes can be followed. The compositions of the present invention can also be formulated and used as tablets, capsules or elixirs for oral administration; suppositories for rectal administration; sterile solutions; suspensions for injectable administration; and the other compositions known in the art. [0170]
  • The present invention also includes pharmaceutically acceptable formulations of the compounds described. These formulations include salts of the above compounds, e.g., acid addition salts, for example, salts of hydrochloric, hydrobromic, acetic acid, and benzene sulfonic acid. [0171]
  • A pharmacological composition or formulation refers to a composition or formulation in a form suitable for administration, e.g., local administration or systemic administration, into a cell or patient, including, for example, a human. Suitable forms, in part, depend upon the use or the route of entry, for example oral, transdermal, or by injection. Such forms should not prevent the composition or formulation from reaching a target cell (i.e., a cell to which the negatively charged polymer is desired to be delivered to). For example, pharmacological compositions injected into the blood stream should be soluble. Other factors are known in the art, and include considerations such as toxicity and forms which prevent the composition or formulation from exerting its effect. [0172]
  • By “local administration” is meant in vivo local absorption or accumulation of drugs in the specific tissue, organ, or compartment of the body. Administration routes that can lead to local absorption include, without limitations: inhalation, direct injection, or dermatological applications. [0173]
  • By “systemic administration” is meant in vivo systemic absorption or accumulation of drugs in the blood stream followed by distribution throughout the entire body. Administration routes which lead to systemic absorption include, without limitations: intravenous, subcutaneous, intraperitoneal, inhalation, oral, intrapulmonary and intramuscular. Each of these administration routes expose the desired compound, e.g., nucleic acids, to an accessible diseased tissue. The rate of entry of a drug into the circulation has been shown to be a function of molecular weight or size. The use of a liposome or other drug carrier comprising the compounds of the instant invention, for example PEG or phospholipids conjugates, can potentially localize the drug, for example, in certain tissue types, such as the tissues of the reticular endothelial system (RES). A nucleic acid formulation that can facilitate the association of drug with the surface of cells, such as, lymphocytes and macrophages is also useful. This approach can provide enhanced delivery of the drug to target cells by taking advantage of the specificity of macrophage and lymphocyte immune recognition of abnormal cells. [0174]
  • Both local and systemic administration approaches can be used to administer nucleic acid molecules of the invention for the treatment of asthma or related conditions. In one embodiment, the nucleic acid molecule or formulation comprising the nucleic acid molecule is administered to a patient with an inhaler or nebulizer, providing rapid local uptake of the nucleic acid molecules into relevant pulmonary tissues. In another embodiment, the nucleic acid molecule or formulation comprising the nucleic acid molecule is administered to a patient systemically, for example by intravenous or subcutaneous injection, providing sustained uptake of the nucleic acid molecules into relevant bodily tissues. [0175]
  • By pharmaceutically acceptable formulation is meant, a composition or formulation that allows for the effective distribution of the nucleic acid molecules of the instant invention in the physical location most suitable for their desired activity. Non-limiting examples of agents suitable for formulation with the nucleic acid molecules of the instant invention include: PEG conjugated nucleic acids, phospholipid conjugated nucleic acids, nucleic acids containing lipophilic moieties, phosphorothioates, P-glycoprotein inhibitors (such as Pluronic P85) which can enhance entry of drugs into various tissues, for exaple the CNS (Jolliet-Riant and Tillement, 1999[0176] , Fundam. Clin. Pharmacol., 13, 16-26); biodegradable polymers, such as poly (DL-lactide-coglycolide) microspheres for sustained release delivery after implantation (Emerich, DF et al, 1999, Cell Transplant, 8, 47-58) Alkermes, Inc. Cambridge, Mass.; and loaded nanoparticles, such as those made of polybutylcyanoacrylate, which can deliver drugs across the blood brain barrier and can alter neuronal uptake mechanisms (Prog Neuropsychopharmacol Biol Psychiatry, 23, 941-949, 1999). Other non-limiting examples of delivery strategies, including CNS delivery of the nucleic acid molecules of the instant invention include material described in Boado et al., 1998, J. Pharm. Sci., 87, 1308-1315; Tyler et al., 1999, FEBS Lett., 421, 280-284; Pardridge et al., 1995, PNAS USA., 92, 5592-5596; Boado, 1995, Adv. Drug Delivery Rev., 15, 73-107; Aldrian-Herrada et al., 1998, Nucleic Acids Res., 26, 4910-4916; and Tyler et al., 1999, PNAS USA., 96, 7053-7058. All these references are hereby incorporated herein by reference.
  • The invention also features the use of the composition comprising surface-modified liposomes containing poly (ethylene glycol) lipids (PEG-modified, or long-circulating liposomes or stealth liposomes). Nucleic acid molecules of the invention can also comprise covalently attached PEG molecules of various molecular weights. These formulations offer a method for increasing the accumulation of drugs in target tissues. This class of drug carriers resists opsonization and elimination by the mononuclear phagocytic system (MPS or RES), thereby enabling longer blood circulation times and enhanced tissue exposure for the encapsulated drug (Lasic et al. [0177] Chem. Rev. 1995, 95, 2601-2627; Ishiwata et al., Chem. Pharm. Bull. 1995, 43, 1005-1011). Such liposomes have been shown to accumulate selectively in tumors, presumably by extravasation and capture in the neovascularized target tissues (Lasic et al., Science 1995, 267, 1275-1276; Oku et al., 1995, Biochim. Biophys. Acta, 1238, 86-90). The long-circulating liposomes enhance the pharmacokinetics and pharmacodynamics of DNA and RNA, particularly compared to conventional cationic liposomes which are known to accumulate in tissues of the MPS (Liu et al., J. Biol. Chem. 1995, 42, 24864-24870; Choi et al., International PCT Publication No. WO 96/10391; Ansell et al., International PCT Publication No. WO 96/10390; Holland et al., International PCT Publication No. WO 96/10392; all of which are incorporated by reference herein). Long-circulating liposomes are also likely to protect drugs from nuclease degradation to a greater extent compared to cationic liposomes, based on their ability to avoid accumulation in metabolically aggressive MPS tissues such as the liver and spleen. All of these references are incorporated by reference herein.
  • The present invention also includes compositions prepared for storage or administration that include a pharmaceutically effective amount of the desired compounds in a pharmaceutically acceptable carrier or diluent. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in [0178] Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985) hereby incorporated by reference herein. For example, preservatives, stabilizers, dyes and flavoring agents can be provided. These include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid. In addition, antioxidants and suspending agents can be used.
  • A pharmaceutically effective dose is that dose required to prevent, inhibit the occurrence, or treat (alleviate a symptom to some extent, or all of the symptoms) of a disease state. The pharmaceutically effective dose depends on the type of disease, the composition used, the route of administration, the type of mammal being treated, the physical characteristics of the specific mammal under consideration, concurrent medication, and other factors which those skilled in the medical arts will recognize. Generally, an amount between 0.1 mg/kg and 100 mg/kg body weight/day of active ingredients is administered dependent upon potency of the negatively charged polymer. [0179]
  • The nucleic acid molecules of the invention and formulations thereof can be administered orally, topically, parenterally, by inhalation or spray or rectally in dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles. The term parenteral as used herein includes percutaneous, subcutaneous, intravascular (e.g., intravenous), intramuscular, or intrathecal injection or infusion techniques and the like. In addition, there is provided a pharmaceutical formulation comprising a nucleic acid molecule of the invention and a pharmaceutically acceptable carrier. One or more nucleic acid molecules of the invention can be present in association with one or more non-toxic pharmaceutically acceptable carriers and/or diluents and/or adjuvants, and if desired other active ingredients. The pharmaceutical compositions containing nucleic acid molecules of the invention can be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. [0180]
  • Compositions intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more such sweetening agents, flavoring agents, coloring agents or preservative agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. These excipients can be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets can be uncoated or they can be coated by known techniques. In some cases such coatings can be prepared by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monosterate or glyceryl distearate can be employed. [0181]
  • Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil. [0182]
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydropropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents can be a naturally-occurring phosphatide, for example, lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions can also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin. [0183]
  • Oily suspensions can be formulated by suspending the active ingredients in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions can contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents and flavoring agents can be added to provide palatable oral preparations. These compositions can be preserved by the addition of an anti-oxidant such as ascorbic acid. [0184]
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents or suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, can also be present. [0185]
  • Pharmaceutical compositions of the invention can also be in the form of oil-in-water emulsions. The oily phase can be a vegetable oil or a mineral oil or mixtures of these. Suitable emulsifying agents can be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol, anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions can also contain sweetening and flavoring agents. [0186]
  • Syrups and elixirs can be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations can also contain a demulcent, a preservative and flavoring and coloring agents. The pharmaceutical compositions can be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents that have been mentioned above. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parentally acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono-or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables. [0187]
  • The nucleic acid molecules of the invention can also be administered in the form of suppositories, e.g., for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient that is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter and polyethylene glycols. [0188]
  • Nucleic acid molecules of the invention can be administered parenterally in a sterile medium. The drug, depending on the vehicle and concentration used, can either be suspended or dissolved in the vehicle. Advantageously, adjuvants such as local anesthetics, preservatives and buffering agents can be dissolved in the vehicle. [0189]
  • Dosage levels of the order of from about 0.1 mg to about 140 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions (about 0.5 mg to about 7 g per patient per day). The amount of active ingredient that can be combined with the carrier materials to produce a single dosage form varies depending upon the host treated and the particular mode of administration. Dosage unit forms generally contain between from about 1 mg to about 500 mg of an active ingredient. [0190]
  • It is understood that the specific dose level for any particular patient depends upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration, and rate of excretion, drug combination and the severity of the particular disease undergoing therapy. [0191]
  • For administration to non-human animals, the composition can also be added to the animal feed or drinking water. It can be convenient to formulate the animal feed and drinking water compositions so that the animal takes in a therapeutically appropriate quantity of the composition along with its diet. It can also be convenient to present the composition as a premix for addition to the feed or drinking water. [0192]
  • The nucleic acid molecules of the present invention can also be administered to a patient in combination with other therapeutic compounds to increase the overall therapeutic effect. The use of multiple compounds to treat an indication can increase the beneficial effects while reducing the presence of side effects. [0193]
  • Alternatively, certain of the nucleic acid molecules of the instant invention can be expressed within cells from eukaryotic promoters (e.g., Izant and Weintraub, 1985[0194] , Science, 229, 345; McGarry and Lindquist, 1986, Proc. Natl. Acad. Sci., USA 83, 399; Scanlon et al., 1991, Proc. Natl. Acad. Sci. USA, 88, 10591-5; Kashani-Sabet et al., 1992, Antisense Res. Dev., 2, 3-15; Dropulic et al., 1992, J. Virol., 66, 1432-41; Weerasinghe et al., 1991, J. Virol., 65, 5531-4; Ojwang et al., 1992, Proc. Natl. Acad. Sci. USA, 89, 10802-6; Chen et al., 1992, Nucleic Acids Res., 20, 4581-9; Sarver et al., 1990 Science, 247, 1222-1225; Thompson et al., 1995, Nucleic Acids Res., 23, 2259; Good et al., 1997, Gene Therapy, 4, 45; all of these references are hereby incorporated in their totalities by reference herein). Those skilled in the art realize that any nucleic acid can be expressed in eukaryotic cells from the appropriate DNA/RNA vector. The activity of such nucleic acids can be augmented by their release from the primary transcript by a enzymatic nucleic acid (Draper et al, PCT WO 93/23569, and Sullivan et al., PCT WO 94/02595; Ohkawa et al., 1992, Nucleic Acids Symp. Ser., 27, 15-6; Taira et al., 1991, Nucleic Acids Res., 19, 5125-30; Ventura et al., 1993, Nucleic Acids Res., 21, 3249-55; Chowrira et al., 1994, J. Biol. Chem., 269, 25856; all of these references are hereby incorporated in their totalities by reference herein). Gene therapy approaches specific to the CNS are described by Blesch et al., 2000, Drug News Perspect., 13, 269-280; Peterson et al., 2000, Cent. Nerv. Syst. Dis., 485-508; Peel and Klein, 2000, J. Neurosci. Methods, 98, 95-104; Hagihara et al., 2000, Gene Ther., 7, 759-763; and Herrlinger et al., 2000, Methods Mol. Med., 35, 287-312. AAV-mediated delivery of nucleic acid to cells of the nervous system is further described by Kaplitt et al., U.S. Pat. No. 6,180,613.
  • In another aspect of the invention, RNA molecules of the present invention can be expressed from transcription units (see for example Couture et al., 1996[0195] , TIG., 12, 510) inserted into DNA or RNA vectors. The recombinant vectors can be DNA plasmids or viral vectors. Ribozyme expressing viral vectors can be constructed based on, but not limited to, adeno-associated virus, retrovirus, adenovirus, or alphavirus. The recombinant vectors capable of expressing the nucleic acid molecules can be delivered as described above, and persist in target cells. Alternatively, viral vectors can be used that provide for transient expression of nucleic acid molecules. Such vectors can be repeatedly administered as necessary. Once expressed, the nucleic acid molecule binds to the target mRNA. Delivery of nucleic acid molecule expressing vectors can be systemic, such as by intravenous or intra-muscular administration, by administration to target cells ex-planted from the patient followed by reintroduction into the patient, or by any other means that would allow for introduction into the desired target cell (for a review see Couture et al., 1996, TIG., 12, 510).
  • In one aspect the invention features an expression vector comprising a nucleic acid sequence encoding at least one of the nucleic acid molecules of the instant invention is disclosed. The nucleic acid sequence encoding the nucleic acid molecule of the instant invention is operable linked in a manner that allows expression of that nucleic acid molecule. [0196]
  • In another aspect the invention features an expression vector comprising: a) a transcription initiation region (e.g., eukaryotic pol I, II or III initiation region); b) a transcription termination region (e.g., eukaryotic pol I, II or III termination region); c) a nucleic acid sequence encoding at least one of the nucleic acid catalyst of the instant invention; and wherein said sequence is operably linked to said initiation region and said termination region, in a manner that allows expression and/or delivery of said nucleic acid molecule. The vector can optionally include an open reading frame (ORF) for a protein operably linked on the 5′ side or the 3′-side of the sequence encoding the nucleic acid catalyst of the invention; and/or an intron (intervening sequences). [0197]
  • Transcription of the nucleic acid molecule sequences are driven from a promoter for eukaryotic RNA polymerase I (pol I), RNA polymerase II (pol II), or RNA polymerase III (pol III). Transcripts from pol II or pol III promoters are expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type depends on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby. Prokaryotic RNA polymerase promoters are also used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells (Elroy-Stein and Moss, 1990[0198] , Proc. Natl. Acad. Sci. USA, 87, 6743-7; Gao and Huang 1993, Nucleic Acids Res., 21, 2867-72; Lieber et al., 1993, Methods Enzymol., 217, 47-66; Zhou et al., 1990, Mol. Cell. Biol., 10, 4529-37). All of these references are incorporated by reference herein. Several investigators have demonstrated that nucleic acid molecules, such as ribozymes expressed from such promoters can function in mammalian cells (e.g. Kashani-Sabet et al., 1992, Antisense Res. Dev., 2, 3-15; Ojwang et al., 1992, Proc. Natl. Acad. Sci. USA, 89, 10802-6; Chen et al., 1992, Nucleic Acids Res., 20, 4581-9; Yu et al., 1993, Proc. Natl. Acad. Sci. USA, 90, 6340-4; L'Huillier et al., 1992, EMBO J, 11, 4411-8; Lisziewicz et al., 1993, Proc. Natl. Acad. Sci. U.S. A, 90, 8000-4; Thompson et al., 1995, Nucleic Acids Res., 23, 2259; Sullenger & Cech, 1993, Science, 262, 1566). More specifically, transcription units such as the ones derived from genes encoding U6 small nuclear (snRNA), transfer RNA (tRNA) and adenovirus VA RNA are useful in generating high concentrations of desired RNA molecules such as ribozymes in cells (Thompson et al., supra; Couture and Stinchcomb, 1996, supra; Noonberg et al., 1994, Nucleic Acid Res., 22, 2830; Noonberg et al., U.S. Pat. No. 5,624,803; Good et al., 1997, Gene Ther., 4, 45; Beigelman et al., International PCT Publication No. WO 96/18736; all of these publications are incorporated by reference herein). The above ribozyme transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated virus vectors), or viral RNA vectors (such as retroviral or alphavirus vectors) (for a review see Couture and Stinchcomb, 1996, supra).
  • In another aspect the invention features an expression vector comprising a nucleic acid sequence encoding at least one of the nucleic acid molecules of the invention, in a manner that allows expression of that nucleic acid molecule. The expression vector comprises in one embodiment; a) a transcription initiation region; b) a transcription termination region; c) a nucleic acid sequence encoding at least one said nucleic acid molecule; and wherein said sequence is operably linked to said initiation region and said termination region, in a manner that allows expression and/or delivery of said nucleic acid molecule. [0199]
  • In another embodiment the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an open reading frame; d) a nucleic acid sequence encoding at least one said nucleic acid molecule, wherein said sequence is operably linked to the 3′-end of said open reading frame; and wherein said sequence is operably linked to said initiation region, said open reading frame and said termination region, in a manner that allows expression and/or delivery of said nucleic acid molecule. In yet another embodiment the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an intron; d) a nucleic acid sequence encoding at least one said nucleic acid molecule; and wherein said sequence is operably linked to said initiation region, said intron and said termination region, in a manner which allows expression and/or delivery of said nucleic acid molecule. [0200]
  • In another embodiment, the expression vector comprises: a) a transcription initiation region; b) a transcription termination region; c) an intron; d) an open reading frame; e) a nucleic acid sequence encoding at least one said nucleic acid molecule, wherein said sequence is operably linked to the 3′-end of said open reading frame; and wherein said sequence is operably linked to said initiation region, said intron, said open reading frame and said termination region, in a manner which allows expression and/or delivery of said nucleic acid molecule. [0201]
  • EXAMPLES
  • The following are non-limiting examples showing the selection, isolation, synthesis and activity of nucleic acids of the instant invention. [0202]
  • The following examples demonstrate the selection and design of Antisense, hammerhead, DNAzyme, NCH, Amberzyme, Zinzyme, or G-Cleaver ribozyme molecules and binding/cleavage sites within PTGDS and/or PTGDR RNA. [0203]
  • Example 1 Identification of Potential Target Sites in Human PTGDS, ADORA1 and PTGDR RNA
  • The sequence of human PTGDS, ADORA1 and PTGDR genes are screened for accessible sites using a computer-folding algorithm. Regions of the RNA that do not form secondary folding structures and contained potential enzymatic nucleic acid molecule and/or antisense binding/cleavage sites are identified. The sequences of PTGDR binding/cleavage sites are shown in Tables III-VII. [0204]
  • Example 2 Selection of Enzymatic Nucleic Acid Cleavage Sites in Human PTGDS, ADORA1 and PTGDR RNA
  • Enzymatic nucleic acid molecule target sites are chosen by analyzing sequences of Human PTGDS (Genbank accession No: NM 000954), ADORA1 (Genbank accession No: NM[0205] 000674) and PTGDR gene (Genbank accession Nos: U31332 and U31099) and prioritizing the sites on the basis of folding. Enzymatic nucleic acid molecules are designed that can bind each target and are individually analyzed by computer folding (Christoffersen et al., 1994 J. Mol. Struc. Theochem, 311, 273; Jaeger et al., 1989, Proc. Natl. Acad. Sci. USA, 86, 7706) to assess whether the enzymatic nucleic acid molecule sequences fold into the appropriate secondary structure. Those enzymatic nucleic acid molecules with unfavorable intramolecular interactions between the binding arms and the catalytic core are eliminated from consideration. As noted below, varying binding arm lengths can be chosen to optimize activity. Generally, at least 4 bases on each arm are able to bind to, or otherwise interact with, the target RNA.
  • Example 3 Chemical Synthesis and Purification of Ribozymes and Antisense for Efficient Cleavage and/or blocking of PTGDS, ADORA1 and PTGDR RNA
  • Enzymatic nucleic acid molecules and antisense constructs are designed to anneal to various sites in the RNA message. The binding arms of the enzymatic nucleic acid molecules are complementary to the target site sequences described above, while the antisense constructs are fully complementary to the target site sequences described above. The enzymatic nucleic acid molecules and antisense constructs were chemically synthesized. The method of synthesis used followed the procedure for normal RNA synthesis as described above and in Usman et al., (1987 J. Am. Chem. Soc., 109, 7845), Scaringe et al., (1990 Nucleic Acids Res., 18, 5433) and Wincott et al., supra, and made use of common nucleic acid protecting and coupling groups, such as dimethoxytrityl at the 5′-end, and phosphoramidites at the 3′-end. The average stepwise coupling yields were typically >98%. [0206]
  • Enzymatic nucleic acid molecules and antisense constructs are also synthesized from DNA templates using bacteriophage T7 RNA polymerase (Milligan and Uhlenbeck, 1989, Methods Enzymol. 180, 51). Enzymatic nucleic acid molecules and antisense constructs are purified by gel electrophoresis using general methods or are purified by high pressure liquid chromatography (HPLC; See Wincott et al., supra; the totality of which is hereby incorporated herein by reference) and are resuspended in water. The sequences of the chemically synthesized enzymatic nucleic acid molecules used in this study are shown below in Table III-VII. The sequences of the chemically synthesized antisense constructs used in this study are complementary sequences to the Substrate sequences shown below as in Table III-VII. [0207]
  • Example 4 Enzymatic Nucleic Acid Molecule Cleavage of PTGDS, ADORA1 and PTGDR RNA Target in vitro
  • Enzymatic nucleic acid molecules targeted to the human PTGDS, ADORA1 and PTGDR RNA are designed and synthesized as described above. These enzymatic nucleic acid molecules can be tested for cleavage activity in vitro, for example, using the following procedure. The target sequences and the nucleotide location within the PTGDR RNA are given in Tables III-VII. [0208]
  • Cleavage Reactions: Full-length or partially full-length, internally-labeled target RNA for enzymatic nucleic acid molecule cleavage assay is prepared by in vitro transcription in the presence of [a-[0209] 32P] CTP, passed over a G 50 Sephadex column by spin chromatography and used as substrate RNA without further purification. Alternately, substrates are 5′-32P-end labeled using T4 polynucleotide kinase enzyme. Assays are performed by pre-warming a 2×concentration of purified enzymatic nucleic acid molecule in enzymatic nucleic acid molecule cleavage buffer (50 mM Tris-HCl, pH 7.5 at 37° C., 10 mM MgCl2) and the cleavage reaction was initiated by adding the 2×enzymatic nucleic acid molecule mix to an equal volume of substrate RNA (maximum of 1-5 nM) that was also pre-warmed in cleavage buffer. As an initial screen, assays are carried out for 1 hour at 37° C. using a final concentration of either 40 nM or 1 mM enzymatic nucleic acid molecule, i.e., enzymatic nucleic acid molecule excess. The reaction is quenched by the addition of an equal volume of 95% formamide, 20 mM EDTA, 0.05% bromophenol blue and 0.05% xylene cyanol after which the sample is heated to 95° C. for 2 minutes, quick chilled and loaded onto a denaturing polyacrylamide gel. Substrate RNA and the specific RNA cleavage products generated by enzymatic nucleic acid molecule cleavage are visualized on an autoradiograph of the gel. The percentage of cleavage is determined by Phosphor Imager® quantitation of bands representing the intact substrate and the cleavage products.
  • Example 5 In vivo Models used to Evaluate the Down-Regulation of PTGDS, ADORA1 and PTGDR Gene Expression
  • Animal Models [0210]
  • Evaluating the efficacy of anti-PTGDS, ADORA-1 and/or PTGDR agents in animal models is an important prerequisite to human clinical trials. Matsuoka et al., 2000[0211] , Science, 287, 2012-2016, describe a useful asthma animal model having generating mice deficient in the PTGDR receptor. Sensitization and aerosol challenge of homozygous (PTGDR−/−) mice with ovalbumin was shown to induce increases in the serum concentration of immunoglobin E (IgE), an allergic mediator that activates mast cells, similar to wild-type mice subjected to the same conditions. The concentration of TH2 cytokines and the degree of lymphocyte lung infiltration in the OVA challenged PTGDR −/− mice was shown to be greatly reduced compared to wild type mice. In addition, the PTGDR −/− mice showed only marginal eosinophil infiltration and failed to develop airway hyperreactivity. Similarly, this model can be used to evaluate mice that are treated with nucleic acid molecules of the invention and can furthermore be used as a positive control in determining the response of mice treated with nucleic acid molecules of the invention by using such factors as airway obstruction, lung capacity, and bronchiolar alveolar lavage (BAL) fluid in the evaluation.
  • Cell Culture [0212]
  • Two human cell lines, NPE cells and NCB-20 cells are known to express PTGDR. Cloned human PTGDR has been expressed in CHO and COS7 cells and used in various studies. These PTGDR expressing lung cell lines can be used in cell culture assays to evaluate nucleic acid molecules of the invention. A primary endpoint in these experiments would be the RT-PCR analysis of PTGDR mRNA expression in PTGDR expressing cells. In addition, ligand binding assays can be developed where binding of PTGDS can be evaluated in response to treatment with nucleic acid molecules of the invention. [0213]
  • Indications [0214]
  • The present body of knowledge in PTGDS, ADORA1 and PTGDR research indicates the need for methods to assay PTGDS, ADORA1 and PTGDR activity and for compounds that can regulate PTGDS, ADORA1 and PTGDR expression for research, diagnostic, and therapeutic use. As described herein, the nucleic acid molecules of the present invention can be used in assays to diagnose disease state related of PTGDS, ADORA1 and/or PTGDR levels. In addition, the nucleic acid molecules can be used to treat disease state related to PTGDS, ADORA1 and/or PTGDR levels. [0215]
  • Particular degenerative and disease states that can be associated with PTGDS, ADORA1 and PTGDR levels include, but are not limited to allergic diseases and conditions, including but not limited to asthma, allergic rhinitis, atopic dermatitis, and any other diseases or conditions that are related to or will respond to the levels of PTGDS, ADORA1 and/or PTGDR in a cell or tissue, alone or in combination with other therapies. [0216]
  • The use of anti-inflammatories, bronchodilators, adenosine inhibitors and adenosine A1 receptor inhibitors are examples of other treatments or therapies can be combined with the nucleic acid molecules of the invention. Those skilled in the art will recognize that other drug compounds and therapies can be similarly be readily combined with the nucleic acid molecules of the instant invention (e.g. enzymatic nucleic acid molecules and antisense molecules) are hence within the scope of the instant invention. [0217]
  • Diagnostic Uses [0218]
  • The nucleic acid molecules of this invention (e.g., enzymatic nucleic acid molecules) can be used as diagnostic tools to examine genetic drift and mutations within diseased cells or to detect the presence of PTGDS, ADORA1 and/or PTGDR RNA in a cell. The close relationship between enzymatic nucleic acid molecule activity and the structure of the target RNA allows the detection of mutations in any region of the molecule that alters the base-pairing and three-dimensional structure of the target RNA. By using multiple enzymatic nucleic acid molecules described in this invention, one can map nucleotide changes which are important to RNA structure and function in vitro, as well as in cells and tissues. Cleavage of target RNAs with enzymatic nucleic acid molecules can be used to inhibit gene expression and define the role (essentially) of specified gene products in the progression of disease. In this manner, other genetic targets can be defined as important mediators of the disease. These experiments can lead to better treatment of the disease progression by affording the possibility of combinational therapies (e.g., multiple enzymatic nucleic acid molecules targeted to different genes, enzymatic nucleic acid molecules coupled with known small molecule inhibitors, or intermittent treatment with combinations of enzymatic nucleic acid molecules and/or other chemical or biological molecules). Other in vitro uses of enzymatic nucleic acid molecules of this invention are well known in the art, and include detection of the presence of mRNAs associated with PTGDS, ADORA1 or PTGDR-related condition. Such RNA is detected by determining the presence of a cleavage product after treatment with an enzymatic nucleic acid molecule using standard methodology. [0219]
  • In a specific example, enzymatic nucleic acid molecules which cleave only wild-type or mutant forms of the target RNA are used for the assay. The first enzymatic nucleic acid molecule is used to identify wild-type RNA present in the sample and the second enzymatic nucleic acid molecule is used to identify mutant RNA in the sample. As reaction controls, synthetic substrates of both wild-type and mutant RNA are cleaved by both enzymatic nucleic acid molecules to demonstrate the relative enzymatic nucleic acid molecule efficiencies in the reactions and the absence of cleavage of the “non-targeted” RNA species. The cleavage products from the synthetic substrates also serve to generate size markers for the analysis of wild-type and mutant RNAs in the sample population. Thus each analysis requires two enzymatic nucleic acid molecules, two substrates and one unknown sample which is combined into six reactions. The presence of cleavage products is determined using an RNAse protection assay so that full-length and cleavage fragments of each RNA can be analyzed in one lane of a polyacrylamide gel. It is not absolutely required to quantify the results to gain insight into the expression of mutant RNAs and putative risk of the desired phenotypic changes in target cells. The expression of mRNA whose protein product is implicated in the development of the phenotype (i.e., PTGDS/PTGDR) is adequate to establish risk. If probes of comparable specific activity are used for both transcripts, then a qualitative comparison of RNA levels will be adequate and will decrease the cost of the initial diagnosis. Higher mutant form to wild-type ratios are correlated with higher risk whether RNA levels are compared qualitatively or quantitatively. The use of enzymatic nucleic acid molecules in diagnostic applications contemplated by the instant invention is described, for example, in George et al., U.S. Pat. Nos. 5,834,186 and 5,741,679, Shih et al., U.S. Pat. No. 5,589,332, Nathan et al., U.S. Pat. No. 5,871,914, Nathan and Ellington, International PCT publication No. WO 00/24931, Breaker et al., International PCT Publication Nos. WO 00/26226 and 98/27104, and Sullenger et al., International PCT publication No. WO 99/29842. [0220]
  • Additional Uses [0221]
  • Potential uses of sequence-specific enzymatic nucleic acid molecules of the instant invention can have many of the same applications for the study of RNA that DNA restriction endonucleases have for the study of DNA (Nathans et al., 1975 [0222] Ann. Rev. Biochem. 44:273). For example, the pattern of restriction fragments can be used to establish sequence relationships between two related RNAs, and large RNAs can be specifically cleaved to fragments of a size more useful for study. The ability to engineer sequence specificity of the enzymatic nucleic acid molecule is ideal for cleavage of RNAs of unknown sequence. Applicant has described the use of nucleic acid molecules to down-regulate gene expression of target genes in bacterial, microbial, fungal, viral, and eukaryotic systems including plant, or mammalian cells.
  • All patents and publications mentioned in the specification are indicative of the levels of skill of those skilled in the art to which the invention pertains. All references cited in this disclosure are incorporated by reference to the same extent as if each reference had been incorporated by reference in its entirety individually. [0223]
  • One skilled in the art would readily appreciate that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The methods and compositions described herein as presently representative of preferred embodiments are exemplary and are not intended as limitations on the scope of the invention. Changes therein and other uses will occur to those skilled in the art, which are encompassed within the spirit of the invention, are defined by the scope of the claims. [0224]
  • It will be readily apparent to one skilled in the art that varying substitutions and modifications may be made to the invention disclosed herein without departing from the scope and spirit of the invention. Thus, such additional embodiments are within the scope of the present invention and the following claims. [0225]
  • The invention illustratively described herein suitably can be practiced in the absence of any element or elements, limitation or limitations that is not specifically disclosed herein. Thus, for example, in each instance herein any of the terms “comprising”, “consisting essentially of” and “consisting of” can be replaced with either of the other two terms. The terms and expressions that have been employed are used as terms of description and not of limitation, and there is no intention that in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments, optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the description and the appended claims. [0226]
  • In addition, where features or aspects of the invention are described in terms of Markush groups or other grouping of alternatives, those skilled in the art will recognize that the invention is also thereby described in terms of any individual member or subgroup of members of the Markush group or other group. [0227]
  • Other embodiments are within the following claims. [0228]
    TABLE I
    Characteristics of naturally occurring ribozymes
    Group I Introns
    Size: ˜150 to >1000 nucleotides.
    Requires a U in the target sequence immediately 5′ of the cleavage site.
    Binds 4-6 nucleotides at the 5′-side of the cleavage site.
    Reaction mechanism: attack by the 3′-OH of guanosine to generate cleavage
    products with 3′-OH and 5′-guanosine.
    Additional protein cofactors required in some cases to help folding and
    maintenance of the active structure.
    Over 300 known members of this class. Found as an intervening sequence in
    Tetrahymena thermophila rRNA, fungal mitochondria, chloroplasts, phage T4, blue-
    green algae, and others.
    Major structural features largely established though phylogenetic comparisons,
    mutagenesis, and biochemical studies [i,ii].
    Complete kinetic framework established for one ribozyme [iii,iv,v,vi].
    Studies of ribozyme folding and substrate docking underway [vii,viii,ix].
    Chemical modification investigation of important residues well established [x,xi].
    The small (4-6 nt) binding site may make this ribozyme too non-specific for
    targeted RNA cleavage, however, the Tetrahymena group I intron has been used
    to repair a “defective” β-galactosidase message by the ligation of new β-
    galactosidase sequences onto the defective message [xii].
    RNAse P RNA (M1 RNA)
    Size: ˜290 to 400 nucleotides.
    RNA portion of a ubiquitous ribonucleoprotein enzyme.
    Cleaves tRNA precursors to form mature tRNA [xiii].
    Reaction mechanism: possible attack by M2+-OH to generate cleavage products
    with 3′-OH and 5′-phosphate.
    RNAse P is found throughout the prokaryotes and eukaryotes. The RNA subunit
    has been sequenced from bacteria, yeast, rodents, and primates.
    Recruitment of endogenous RNAse P for therapeutic applications is possible
    through hybridization of an External Guide Sequence (EGS) to the target RNA
    [xiv,xv]
    Important phosphate and 2′OH contacts recently identified [xvi,xvii]
    Group II Introns
    Size: >1000 nucleotides.
    Trans cleavage of target RNAs recently demonstrated [xviii,xix].
    Sequence requirements not fully determined.
    Reaction mechanism: 2′-OH of an internal adenosine generates cleavage products
    with 3′-OH and a “lariat” RNA containing a 3′-5′ and a 2′-5′ branch point.
    Only natural ribozyme with demonstrated participation in DNA cleavage [xx,xxi] in
    addition to RNA cleavage and ligation.
    Major structural features largely established through phylogenetic comparisons
    [xxii].
    Important 2′OH contacts beginning to be identified [xxiii]
    Kinetic framework under development [xxiv]
    Neurospora VS RNA
    Size: ˜144 nucleotides.
    Trans cleavage of hairpin target RNAs recently demonstrated [xxv].
    Sequence requirements not fully determined.
    Reaction mechanism: attack by 2′-OH 5′to the scissile bond to generate cleavage
    products with 2′,3′-cyclic phosphate and 5′-OH ends.
    Binding sites and structural requirements not fully determined.
    Only 1 known member of this class. Found in Neurospora VS RNA.
    Hammerhead Ribozyme
    (see text for references)
    Size: ˜13 to 40 nucleotides.
    Requires the target sequence UH immediately 5′ of the cleavage site.
    Binds a variable number nucleotides on both sides of the cleavage site.
    Reaction mechanism: attack by 2′-OH 5′ to the scissile bond to generate cleavage
    products with 2′,3′-cyclic phosphate and 5′-OH ends.
    14 known members of this class. Found in a number of plant pathogens
    (virusoids) that use RNA as the infectious agent.
    Essential structural features largely defined, including 2 crystal structures [xxvi,xxvii]
    Minimal ligation activity demonstrated (for engineering through in vitro selection)
    [xxviii]
    Complete kinetic framework established for two or more ribozymes [xxix].
    Chemical modification investigation of important residues well established [xxx].
    Hairpin Ribozyme
    Size: ˜50 nucleotides.
    Requires the target sequence GUC immediately 3′of the cleavage site.
    Binds 4-6 nucleotides at the 5′-side of the cleavage site and a variable number to
    the 3′-side of the cleavage site.
    Reaction mechanism: attack by 2′-OH 5′ to the scissile bond to generate cleavage
    products with 2′,3′-cyclic phosphate and 5′-OH ends.
    3 known members of this class. Found in three plant pathogen (satellite RNAs of
    the tobacco ringspot virus, arabis mosaic virus and chicory yellow mottle virus)
    which uses RNA as the infectious agent.
    Essential structural features largely defined [xxxi,xxxii,xxxiii,xxxiv]
    Ligation activity (in addition to cleavage activity) makes ribozyme amenable to
    engineering through in vitro selection [xxxv]
    Complete kinetic framework established for one ribozyme [xxxvi].
    Chemical modification investigation of important residues begun [xxxvii,xxxviii].
    Hepatitis Delta Virus (HDV) Ribozyme
    Size: ˜60 nucleotides.
    Trans cleavage of target RNAs demonstrated [xxxix].
    Binding sites and structural requirements not fully determined, although no
    sequences 5′ of cleavage site are required. Folded ribozyme contains a pseudoknot
    structure [xl].
    Reaction mechanism: attack by 2′-OH 5′ to the scissile bond to generate cleavage
    products with 2′,3′-cyclic phosphate and 5′-OH ends.
    Only 2 known members of this class. Found in human HDV.
    Circular form of HDV is active and shows increased nuclease stability [xli]
  • [0229]
    TABLE II
    Reagent Equivalents Amount Wait Time* DNA Wait Time* 2′-O-methyl Wait Time*RNA
    A. 2.5 μmol Synthesis Cycle ABI 394 Instrument
    Phosphoramidites 6.5 163 μL 45 sec 2.5 min 7.5 min
    S-Ethyl Tetrazole 23.8 238 μL 45 sec 2.5 min 7.5 min
    Acetic Anhydride 100 233 μL  5 sec 5 sec 5 sec
    N-Methyl 186 233 μL  5 sec 5 sec 5 sec
    Imidazole
    TCA 176 2.3 mL 21 sec 21 sec 21 sec
    Iodine 11.2 1.7 mL 45 sec 45 sec 45 sec
    Beaucage 12.9 645 μL 100 sec  300 sec 300 sec
    Acetonitrile NA 6.67 mL NA NA NA
    B. 0.2 μmol Synthesis Cycle ABI 394 Instrument
    Phosphoramidites 15 31 μL 45 sec 233 sec 465 sec
    S-Ethyl Tetrazole 38.7 31 μL 45 sec 233 min 465 sec
    Acetic Anhydride 655 124 μL  5 sec 5 sec 5 sec
    N-Methyl 1245 124 μL  5 sec 5 sec 5 sec
    Imidazole
    TCA 700 732 μL 10 sec 10 sec 10 sec
    Iodine 20.6 244 μL 15 sec 15 sec 15 sec
    Beaucage 7.7 232 μL 100 sec  300 sec 300 sec
    Acetonitrile NA 2.64 mL NA NA NA
    C. 0.2 μmol Synthesis Cycle 96 well Instrument
    Equivalents: DNA/ Amount: DNA/2′-O- Wait Time* 2′-O- Wait Time*
    Reagent 2′-O-methyl/Ribo methyl/Ribo Wait Time* DNA methyl Ribo
    Phosphoramidites 22/33/66  40/60/120 μL  60 sec 180 sec 360 sec 
    S-Ethyl Tetrazole 70/105/210 40/60/120 μL  60 sec 180 min 360 sec 
    Acetic Anhydride 265/265/265  50/50/50 μL  10 sec 10 sec 10 sec
    N-Methyl 502/502/502  50/50/50 μL  10 sec 10 sec 10 sec
    Imidazole
    TCA 238/475/475  250/500/500 μL  15 sec 15 sec 15 sec
    Iodine 6.8/6.8/6.8    80/80/80 μL  30 sec 30 sec 30 sec
    Beaucage 34/51/51  80/120/120 100 sec 200 sec 200 sec 
    Acetonitrile NA 1150/1150/1150 μL NA NA NA
  • [0230]
    TABLE III
    Human PTGDR Hammerhead Ribozyme and Substrate Sequence
    Seq Seq
    Pos Substrate ID Hammerhead Ribozyme ID
    12 UUCUGGCU A UUUUCCUC 1 GAGGAAAA CUGAUGAGGCCGUUAGGCCGAA AGCCAGAA 228
    14 CUGGCUAU U UUCCUCCU 2 AGGAGGAA CUGAUGAGGCCGUUAGGCCGAA AUAGCCAG 229
    15 UGGCUAUU U UCCUCCUG 3 CAGGAGGA CUGAUGAGGCCGUUAGGCCGAA AAUAGCCA 230
    16 GGCUAUUU U CCUCCUGC 4 GCAGGAGG CUGAUGAGGCCGUUAGGCCGAA AAAUAGCC 231
    17 GCUAUUUU C CUCCUGCC 5 GGCAGGAG CUGAUGAGGCCGUUAGGCCGAA AAAAUAGC 232
    20 AUUUUCCU C CUGCCGUU 6 AACGGCAG CUGAUGAGGCCGUUAGGCCGAA AGGAAAAU 233
    28 CCUGCCGU U CCGACUCG 7 CGAGUCGG CUGAUGAGGCCGUUAGGCCGAA ACGGCAGG 234
    29 CUGCCGUU C CGACUCGG 8 CCGAGUCG CUGAUGAGGCCGUUAGGCCGAA AACGGCAG 235
    35 UUCCGACU C GGCACCAG 9 CUGGUGCC CUGAUGAGGCCGUUAGGCCGAA AGUCGGAA 236
    47 ACCAGAGU C UGUCUCUA 10 UAGAGACA CUGAUGAGGCCGUUAGGCCGAA ACUCUGGU 237
    51 GAGUCUGU C UCUACUGA 11 UCAGUAGA CUGAUGAGGCCGUUAGGCCGAA ACAGACUC 238
    53 GUCUGUCU C UACUGAGA 12 UCUCAGUA CUGAUGAGGCCGUUAGGCCGAA AGACAGAC 239
    55 CUGUCUCU A CUGAGAAC 13 GUUCUCAG CUGAUGAGGCCGUUAGGCCGAA AGAGACAG 240
    73 CAGCGCGU C AGGGCCGA 14 UCGGCCCU CUGAUGAGGCCGUUAGGCCGAA ACGCGCUG 241
    85 GCCGAGCU C UUCACUGG 15 CCAGUGAA CUGAUGAGGCCGUUAGGCCGAA AGCUCGGC 242
    87 CGAGCUCU U CACUGGCC 16 GGCCAGUG CUGAUGAGGCCGUUAGGCCGAA AGAGCUCG 243
    88 GAGCUCUU C ACUGGCCU 17 AGGCCAGU CUGAUGAGGCCGUUAGGCCGAA AAGAGCUC 244
    100 GGCCUGCU C CGCGCUCU 18 AGAGCGCG CUGAUGAGGCCGUUAGGCCGAA AGCAGGCC 245
    107 UCCGCGCU C UUCAAUGC 19 GCAUUGAA CUGAUGAGGCCGUUAGGCCGAA AGCGCGGA 246
    109 CGCGCUCU U CAAUGCCA 20 UGGCAUUG CUGAUGAGGCCGUUAGGCCGAA AGAGCGCG 247
    110 GCGCUCUU C AAUGCCAG 21 CUGGCAUU CUGAUGAGGCCGUUAGGCCGAA AAGAGCGC 248
    130 CAGGCGCU C ACCCUGCA 22 UGCAGGGU CUGAUGAGGCCGUUAGGCCGAA AGCGCCUG 249
    145 CAGAGCGU C CCGCCUCU 23 AGAGGCGG CUGAUGAGGCCGUUAGGCCGAA ACGCUCUG 250
    152 UCCCGCCU C UCAAAGAG 24 CUCUUUGA CUGAUGAGGCCGUUAGGCCGAA AGGCGGGA 251
    154 CCGCCUCU C AAAGAGGG 25 CCCUCUUU CUGAUGAGGCCGUUAGGCCGAA AGAGGCGG 252
    178 CCGCGAGU U UAGAUAGG 26 CCUAUCUA CUGAUGAGGCCGUUAGGCCGAA ACUCGCGG 253
    179 CGCGAGUU U AGAUAGGA 27 UCCUAUCU CUGAUGAGGCCGUUAGGCCGAA AACUCGCG 254
    180 GCGAGUUU A GAUAGGAG 28 CUCCUAUC CUGAUGAGGCCGUUAGGCCGAA AAACUCGC 255
    184 GUUUAGAU A GGAGGUUC 29 GAACCUCC CUGAUGAGGCCGUUAGGCCGAA AUCUAAAC 256
    191 UAGGAGGU U CCUGCCGU 30 ACGGCAGG CUGAUGAGGCCGUUAGGCCGAA ACCUCCUA 257
    192 AGGAGGUU C CUGCCGUG 31 CACGGCAG CUGAUGAGGCCGUUAGGCCGAA AACCUCCU 258
    220 GCCGCCCU C GGAGCUUU 32 AAAGCUCC CUGAUGAGGCCGUUAGGCCGAA AGGGCGGC 259
    227 UCGGAGCU U UUUCUGUG 33 CACAGAAA CUGAUGAGGCCGUUAGGCCGAA AGCUCCGA 260
    228 CGGAGCUU U UUCUGUGG 34 CCACAGAA CUGAUGAGGCCGUUAGGCCGAA AAGCUCCG 261
    229 GGAGCUUU U UCUGUGGC 35 GCCACAGA CUGAUGAGGCCGUUAGGCCGAA AAAGCUCC 262
    230 GAGCUUUU U CUGUGGCG 36 CGCCACAG CUGAUGAGGCCGUUAGGCCGAA AAAAGCUC 263
    231 AGCUUUUU C UGUGGCGC 37 GCGCCACA CUGAUGAGGCCGUUAGGCCGAA AAAAAGCU 264
    244 GCGCAGCU U CUCCGCCC 38 GGGCGGAG CUGAUGAGGCCGUUAGGCCGAA AGCUGCGC 265
    245 CGCAGCUU C UCCGCCCG 39 CGGGCGGA CUGAUGAGGCCGUUAGGCCGAA AAGCUGCG 266
    247 CAGCUUCU C CGCCCGAG 40 CUCGGGCG CUGAUGAGGCCGUUAGGCCGAA AGAAGCUG 267
    280 CGGGGGCU C CUUAGCAC 41 GUGCUAAG CUGAUGAGGCCGUUAGGCCGAA AGCCCCCG 268
    283 GGGCUCCU U AGCACCCG 42 CGGGUGCU CUGAUGAGGCCGUUAGGCCGAA AGGAGCCC 269
    284 GGCUCCUU A GCACCCGG 43 CCGGGUGC CUGAUGAGGCCGUUAGGCCGAA AAGGAGCC 270
    306 GGGGCCCU C GCCCUUCC 44 GGAAGGGC CUGAUGAGGCCGUUAGGCCGAA AGGGCCCC 271
    312 CUCGCCCU U CCGCAGCC 45 GGCUGCGG CUGAUGAGGCCGUUAGGCCGAA AGGGCGAG 272
    313 UCGCCCUU C CGCAGCCU 46 AGGCUGCG CUGAUGAGGCCGUUAGGCCGAA AAGGGCGA 273
    322 CGCAGCCU U CACUCCAG 47 CUGGAGUG CUGAUGAGGCCGUUAGGCCGAA AGGCUGCG 274
    323 GCAGCCUU C ACUCCAGC 48 GCUGGAGU CUGAUGAGGCCGUUAGGCCGAA AAGGCUGC 275
    327 CCUUCACU C CAGCCCUC 49 GAGGGCUG CUGAUGAGGCCGUUAGGCCCAA AGUGAAGG 276
    335 CCAGCCCU C UGCUCCCG 50 CGUGAGCA CUGAUGAGGCCGUUACGCCGAA AGGGCUGG 277
    340 CCUCUGCU C CCGCACGC 51 GCGUGCGG CUGAUGAGGCCGUUAGGCCGAA AGCAGAGG 278
    357 CAUGAAGU C GCCGUUCU 52 AGAACGGC CUGAUGAGGCCGUUAGGCCGAA ACUUCAUG 279
    363 GUCGCCGU U CUACCGCU 53 ACCCGUAG CUGAUGAGGCCGUUAGGCCGAA ACGGCGAC 280
    364 UCGCCGUU C UACCGCUG 54 CAGCGGUA CUGAUGAGGCCGUUAGGCCGAA AACGCCGA 281
    366 GCCGUUCU A CCGCUGCC 55 GGCAGCGG CUGAUGAGGCCGUUAGGCCGAA AGAACGGC 282
    387 CACCACCU C UCUCGAAA 56 UUUCCACA CUGAUGAGGCCGUUAGGCCGAA AGGUGGUG 283
    405 AGGCAACU C GGCGGUGA 57 UCACCGCC CUGAUGAGGCCGUUAGGCCGAA AGUUGCCU 284
    427 GCGGUGCU C UUCAGCAC 58 GUGCUGAA CUGAUGAGGCCGUUAGGCCGAA AGCACCCC 285
    429 GGUGCUCU U CAGCACCG 59 CGGUGCUG CUGAUGAGGCCUUUAGGCCGAA AGAGCACC 286
    430 GUGCUCUU C AGCACCGG 60 CCGGUGCU CUGAUGAGGCCGUUAGGCCGAA AAGAGCAC 287
    442 ACCGGCCU C CUGGGCAA 61 UUGCCCAG CUGAUGAGGCCGUUAGGCCGAA AGGCCGGU 288
    480 GGCCCGCU C GGGGCUGG 62 CCAGCCCC CUGAUGAGGCCGUUAGGCCGAA AGCGCGCC 289
    498 GUGGUGCU C GCGGCGUC 63 GACGCCGC CUGAUGAGGCCGUUAGGCCGAA AGCACCAC 290
    506 CGCGGCGU C CACUGCGC 64 GCGCAGUG CUGAUGAGGCCGUUAGGCCCAA ACGCCGCG 291
    525 GCUGCCCU C GGUCUUCU 65 AGAAGACC CUGAUGAGGCCGUUAGGCCGAA AGGGCAGC 292
    529 CCCUCGGU C UUCUACAU 66 AUGUAGAA CUGAUGAGGCCGUUAGGCCGAA ACCGAGGG 293
    531 CUCGGUCU U CUACAUGC 67 GCAUGUAG CUGAUGAGGCCCUUACGCCGAA AGACCGAG 294
    532 UCGGUCUU C UACAUGCU 68 AGCAUGUA CUGAUGAGGCCGUUAGGCCGAA AAGACCGA 295
    534 GGUCUUCU A CAUGCUGG 69 CCAGCAUG CUGAUGAGGCCGUUAGCCCGAA AGAAGACC 296
    559 CUGACGGU C ACCGACUU 70 AAGUCGGU CUGAUGAGGCCGUUAGGCCGAA ACCGUCAG 297
    567 CACCGACU U GCUGGGCA 71 UCCCCAGC CUGAUCAGCCCGUUAGGCCCAA AGUCGGUG 298
    583 AAGUGCCU C CUAAGCCC 72 GGGCUUAG CUGAUGAGGCCGUUAGGCCGAA AGGCACUU 299
    586 UGCCUCCU A AGCCCGGU 73 ACCGGGCU CUGAUGAGGCCCUUAGGCCGAA ACGAGGCA 300
    609 GGCUGCCU A CGCUCAGA 74 UCUGAGCG CUGAUGAGGCCGUUAGGCCGAA AGGCAGCC 301
    614 CCUACCCU C AGAACCGG 75 CCGGUUCU CUGAUGAGGCCGUUAGGCCGAA AGCGUAGG 302
    626 ACCGGAGU C UGCGGGUG 76 CACCCCCA CUGAUGAGGCCCUUAGGCCGAA ACUCCCCU 303
    637 CGCGUGCU U CCGCCCGC 77 GCGGGCGC CUGAUGAGGCCGUUAGGCCGAA AGCACCCG 304
    648 GCCCGCAU U GGACAACU 78 AGUUGUCC CUGAUGAGGCCGUUAGGCCGAA AUGCGGGC 305
    657 CGACAACU C GUUGUGCC 79 GGCACAAC CUGAUGAGGCCGUUAGGCCGAA AGUUGUCC 306
    660 CAACUCGU U GUGCCAAG 80 CUUGGCAC CUGAUGAGGCCGUUAGGCCGAA ACGAGUUG 307
    672 CCAAGCCU U CGCCUUCU 81 AGAAGGCG CUGAUGAGGCCGUUAGGCCGAA AGGCUUGG 308
    673 CAAGCCUU C GCCUUCUU 82 AAGAAGGC CUGAUGAGGCCGUUAGGCCGAA AAGGCUUG 309
    678 CUUCGCCU U CUUCAUGU 83 ACAUGAAG CUGAUGAGGCCGUUAGGCCGAA AGGCGAAG 310
    679 UUCGCCUU C UUCAUGUC 84 GACAUGAA CUGAUGAGGCCGUUAGGCCGAA AAGGCGAA 311
    681 CGCCUUCU U CAUGUCCU 85 AGGACAUC CUGAUGACCCCGUUAGGCCGAA ACAAGGCC 312
    682 GCCUUCUU C AUGUCCUU 86 AAGGACAU CUGAUGAGGCCGUUAGGCCGAA AAGAAGGC 313
    687 CUUCAUGU C CUUCUUUG 87 CAAAGAAG CUGAUGAGGCCGUUAGGCCGAA ACAUGAAG 314
    690 CAUGUCCU U CUUUGGGC 88 GCCCAAAG CUGAUGAGGCCGUUAGGCCGAA AGGACAUG 315
    691 AUGUCCUU C UUUGGGCU 89 AGCCCAAA CUGAUGAGGCCGUUAGGCCGAA AAGGACAU 316
    693 GUCCUUCU U UGGGCUCU 90 AGAGCCCA CUGAUGAGGCCGUUAGGCCGAA AGAAGGAC 317
    694 UCCUUCUU U GGGCUCUC 91 GAGAGCCC CUGAUGAGGCCGUUAGGCCGAA AAGAAGGA 318
    700 UUUGGGCU C UCCUCGAC 92 GUCGAGGA CUGAUGAGGCCGUUAGGCCGAA AGCCCAAA 319
    702 UGGGCUCU C CUCGACAC 93 GUGUCGAG CUGAUGAGGCCGUUAGGCCGAA AGAGCCCA 320
    705 GCUCUCCU C GACACUGC 94 GCAGUGUC CUGAUGAGGCCGUUAGGCCGAA AGGAGAGC 321
    718 CUGCAACU C CUGGCCAU 95 AUGGCCAG CUGAUGAGGCCGUUAGGCCGAA AGUUGCAG 322
    745 UGCUGGCU C UCCCUAGG 96 CCUAGGGA CUGAUGAGGCCGUUAGGCCGAA AGCCAGCA 323
    747 CUGGCUCU C CCUAGGGC 97 GCCCUAGG CUGAUGAGGCCGUUAGGCCGAA AGAGCCAG 324
    751 CUCUCCCU A GGGCACCC 98 GGGUGCCC CUGAUGAGGCCGUUAGGCCGAA AGGGAGAG 325
    761 GGCACCCU U UCUUCUAC 99 GUAGAAGA CUGAUGAGGCCGUUAGGCCGAA AGGGUGCC 326
    762 GCACCCUU U CUUCUACC 100 GGUAGAAG CUGAUGAGGCCGUUAGGCCGAA AAGGGUGC 327
    763 CACCCUUU C UUCUACCC 101 CGGUAGAA CUGAUGAGCCCGUUAGGCCGAA AAAGGGUG 328
    765 CCCUUUCU U CUACCGAC 102 GUCGGUAG CUGAUGAGGCCGUUAGGCCGAA AGAAAGGG 329
    766 CCUUUCUU C UACCGACG 103 CGUCGGUA CUGAUGAGGCCGUUAGGCCGAA AAGAAAGG 330
    768 UUUCUUCU A CCGACGGC 104 GCCGUCGG CUCAUGAGGCCUUUAGGCCGAA AGAAGAAA 331
    781 CGCCACAU C ACCCUGCG 105 CGCAGGGU CUGAUGAGGCCGUUAGGCCGAA AUGUGCCG 332
    825 GAGCGCCU U CUCCCUGG 106 CCAGGGAG CUGAUCAGCCCGUUAGGCCGAA AGGCGCUC 333
    826 AGCGCCUU C UCCCUGGC 107 GCCAGGGA CUGAUGAGGCCGUUAGGCCGAA AAGGCGCU 334
    828 CGCCUUCU C CCUGGCUU 108 AAGCCACG CUGAUGAGGCCGUUAGGCCGAA AGAAGGCG 335
    836 CCCUGGCU U UCUGCGCG 109 CGCGCAGA CUGAUGAGGCCGUUAGGCCGAA AUCCAGGG 336
    837 CCUGGCUU U CUGCGCGC 110 GCGCGCAG CUGAUCACGCCGUUAGGCCGAA AAGCCAGG 337
    838 CUGGCUUU C UGCGCGCU 111 AGCGCGCA CUGAUGAGGCCGUUAGGCCGAA AAAGCCAG 338
    847 UGCGCGCU A CCUUUCAU 112 AUGAAAGC CUCAUGAGGCCGUUACGCCGAA AGCGCGCA 339
    851 CGCUACCU U UCAUCGCC 113 GCCCAUGA CUGAUGAGGCCGUUAGGCCGAA AGGUAGCG 340
    852 GCUACCUU U CAUGGGCU 114 AGCCCAUG CUGAUGAGGCCGUUAGGCCGAA AAGGUAGC 341
    853 CUACCUUU C AUGGGCUU 115 AAGCCCAU CUGAUGAGGCCGUUAGGCCGAA AAAGGUAG 342
    861 CAUGGGCU U CGGGAAGU 116 ACUUCCCG CUGAUGAGGCCGUUAGGCCGAA AGCCCAUG 343
    862 AUGGGCUU C GGGAAGUU 117 AACUUCCC CUGAUGAGGCCGUUAGGCCGAA AAGCCCAU 344
    870 CGGGAAGU U CGUGCAGU 118 ACUGCACG CUGAUGAGGCCGUUAGGCCGAA ACUUCCCG 345
    871 GGGAAGUU C GUGCAGUA 119 UACUGCAC CUGAUGAGGCCGUUAGGCCGAA AACUUCCC 346
    879 CGUGCAGU A CUGCCCCG 120 CGGGGCAG CUGAUGAGGCCGUUAGGCCGAA ACUGCACG 347
    900 CUGGUGCU U UAUCCAGA 121 UCUGGAUA CUGAUGAGGCCGUUAGGCCGAA AGCACCAG 348
    901 UGGUGCUU U AUCCAGAU 122 AUCUGGAU CUGAUGAGGCCGUUAGGCCGAA AAGCACCA 349
    902 GGUGCUUU A UCCAGAUG 123 CAUCUGGA CUGAUGAGGCCGUUAGGCCGAA AAAGCACC 350
    904 UGCUUUAU C CAGAUGGU 124 ACCAUCUG CUGAUGAGGCCGUUAGGCCGAA AUAAAGCA 351
    913 CAGAUGGU C CACGAGGA 125 UCCUCGUG CUGAUGAGGCCGUUAGGCCGAA ACCAUCUG 352
    927 GGAGGGCU C GCUGUCGG 126 CCGACAGC CUGAUGAGGCCGUUAGGCCGAA AGCCCUCC 353
    933 CUCGCUGU C GGUGCUGG 127 CCAGCACC CUGAUGAGGCCGUUAGGCCGAA ACAGCGAG 354
    945 GCUGGGGU A CUCUGUGC 128 GCACAGAG CUGAUGAGGCCGUUAGGCCGAA ACCCCAGC 355
    948 GGGGUACU C UGUGCUCU 129 AGAGCACA CUGAUGAGGCCGUUAGGCCGAA AGUACCCC 356
    955 UCUGUGCU C UACUCCAG 130 CUGGAGUA CUGAUGAGGCCGUUAGGCCGAA AGCACAGA 357
    957 UGUGCUCU A CUCCAGCC 131 GGCUGGAG CUGAUGAGGCCGUUAGGCCGAA AGAGCACA 358
    960 GCUCUACU C CAGCCUCA 132 UGAGGCUG CUGAUGAGGCCGUUAGGCCGAA AGUAGAGC 359
    967 UCCAGCCU C AUGGCGCU 133 AGCGCCAU CUGAUGAGGCCGUUAGGCCGAA AGGCUGGA 360
    982 CUGCUGGU C CUCGCCAC 134 GUGGOGAG CUGAUGAGGCCGUUAGGCCGAA ACCAGCAG 361
    985 CUGGUCCU C GCCACCGU 135 ACGGUGGC CUGAUGAGGCCGUUAGGCCGAA AGGACCAG 362
    1006 UGCAACCU C GGCGCCAU 136 AUGGCGCC CUGAUGAGGCCGUUAGGCCGAA AGGUUGCA 363
    1024 CGCAACCU C UAUGCGAU 137 AUCGCAUA CUGAUGAGGCCGUUAGGCCGAA AGGUUGCG 364
    1026 CAACCUCU A UGCGAUGC 138 GCAUCGCA CUGAUGAGGCCGUUAGGCCGAA AGAGGUUG 365
    1062 CCCGCGCU C CUCCACCA 139 UGGUGCAG CUGAUGAGGCCGUUAGGCCGAA AGCGCGGG 366
    1110 GGAAGCGU C CCCUCAGC 140 GCUGAGGG CUGAUGAGGCCGUUAGGCCGAA ACGCUUCC 367
    1115 CGUCCCCU C AGCCCCUG 141 CAGGGGCU CUGAUGAGGCCGUUAGGCCGAA AGGGGACG 368
    1136 AGCUGGAU C ACCUCCUG 142 CAGGAGGU CUGAUGAGGCCGUUAGGCCGAA AUCCAGCU 369
    1141 GAUCACCU C CUGCUGCU 143 AGCAGCAG CUGAUGAGGCCGUUAGGCCGAA AGGUGAUC 370
    1168 ACCGUGCU C UUCACUAU 144 AUAGUGAA CUGAUGAGGCCGUUAGGCCGAA AGCACGGU 371
    1170 CGUGCUCU U CACUAUGU 145 ACAUAGUG CUGAUGAGGCCGUUAGGCCGAA AGAGCACG 372
    1171 GUGCUCUU C ACUAUGUG 146 CACAUAGU CUGAUGAGGCCGUUAGGCCGAA AAGAGCAC 373
    1175 UCUUCACU A UGUGUUCU 147 AGAACACA CUGAUGAGGCCGUUAGGCCGAA AGUGAAGA 374
    1181 CUAUGUGU U CUCUGCCC 148 GGGCAGAG CUGAUGAGGCCGUUAGGCCGAA ACACAUAG 375
    1182 UAUGUGUU C UCUGCCCG 149 CGGGCAGA CUGAUGAGGCCGUUAGGCCGAA AACACAUA 376
    1184 UGUGUUCU C UGCCCGUA 150 UACGGGCA CUGAUGAGCCCGUUAGGCCGAA AGAACACA 377
    1192 CUGCCCGU A AUUUAUCG 151 CGAUAAAU CUGAUGAGGCCGUUAGGCCGAA ACGGGCAC 378
    1195 CCCGUAAU U UAUCGCGC 152 GCGCGAUA CUGAUGAGGCCGUUAGGCCGAA AUUACGGG 379
    1196 CCGUAAUU U AUCGCGCU 153 AGCGCGAU CUCAUGAGGCCGUUAGGCCGAA AAUUACGG 380
    1197 CGUAAUUU A UCGCUCUU 154 AAGCGCGA CUGAUGAGGCCGUUAGGCCGAA AAAUUACG 381
    1199 UAAUUUAU C GCGCUUAC 155 GUAAGCGC CUGAUGAGGCCGUUAGGCCGAA AUAAAUUA 382
    1205 AUCGCGCU U ACUAUGGA 156 UCCAUAGU CUGAUGAGGCCGUUAGGCCGAA AGCGCGAU 383
    1206 UCGCGCUU A CUAUGGAG 157 CUCCAUAG CUGAUGAGGCCGUUAGGCCGAA AAGCGCGA 384
    1209 CGCUUACU A UGGAGCAU 158 AUGCUCCA CUGAUGAGGCCGUUAGGCCGAA AGUAAGCG 385
    1218 UGGAGCAU U UAAGGAUG 159 CAUCCUUA CUGAUGAGGCCCUUAGGCCGAA AUGCUCCA 386
    1219 GGAGCAUU U AAGGAUGU 160 ACAUCCUU CUGAUGAGGCCGUUAGGCCGAA AAUGCUCC 387
    1220 GAGCAUUU A AGGAUGUC 161 GACAUCCU CUGAUGAGGCCGUUAGGCCGAA AAAUGCUC 388
    1228 AAGGAUGU C AAGGAGAA 162 UUCUCCUU CUGAUGAGGCCGUUAGGCCGAA ACAUCCUC 389
    1248 CAGGACCU C UGAAGAAG 163 CUUCUUCA CUGAUGAGGCCGUUAGGCCGAA AGGUCCUG 390
    1267 GAAGACCU C CGAGCCUU 164 AAGGCUCG CUGAUGAGGCCGUUAGGCCGAA AGGUCUUC 391
    1275 CCGAGCCU U GCGAUUUC 165 GAAAUCGC CUGAUGAGGCCGUUAGGCCGAA AGGCUCGG 392
    1281 CUUGCGAU U UCUAUCUG 166 CAGAUAGA CUGAUGAGGCCGUUAGGCCGAA AUCGCAAG 393
    1282 UUGCGAUU U CUAUCUGU 167 ACAGAUAG CUGAUGAGGCCGUUAGGCCGAA AAUCGCAA 394
    1283 UGCGAUUU C UAUCUGUG 168 CACAGAUA CUGAUGAGGCCGUUAGGCCGAA AAAUCGCA 395
    1285 CGAUUUCU A UCUGUGAU 169 AUCACAGA CUGAUGAGGCCGUUAGGCCGAA AGAAAUCG 396
    1287 AUUUCUAU C UGUGAUUU 170 AAAUCACA CUGAUGAGGCCGUUAGGCCGAA AUAGAAAU 397
    1294 UCUGUGAU U UCAAUUGU 171 ACAAUUGA CUGAUGAGGCCGUUAGGCCGAA AUCACAGA 398
    1295 CUGUGAUU U CAAUUGUG 172 CACAAUUG CUGAUGAGGCCGUUAGGCCGAA AAUCACAG 399
    1296 UGUGAUUU C AAUUGUGG 173C CACAAUU CUGAUGAGGCCGUUAGGCCGAA AAAUCACA 400
    1300 AUUUCAAU U GUGGACCC 174 GGGUCCAC CUGAUGAGGCCGUUAGGCCGAA AUUGAAAU 401
    1310 UGGACCCU U GGAUUUUU 175 AAAAAUCC CUGAUGAGGCCGUUAGGCCGAA AGGGUCCA 402
    1315 CCUUGGAU U UUUAUCAU 176 AUGAUAAA CUGAUGAGGCCGUUAGGCCGAA AUCCAAGG 403
    1316 CUUGGAUU U UUAUCAUU 177 AAUGAUAA CUGAUGAGGCCGUUAGGCCGAA AAUCCAAG 404
    1317 UUGGAUUU U UAUCAUUU 178 AAAUGAUA CUGAUGAGGCCGUUAGGCCGAA AAAUCCAA 405
    1318 UGGAUUUU U AUCAUUUU 179 AAAAUGAU CUGAUGAGGCCGUUAGGCCGAA AAAAUCCA 406
    1319 GGAUUUUU A UCAUUUUC 180 GAAAAUGA CUGAUGAGGCCGUUAGGCCGAA AAAAAUCC 407
    1321 AUUUUUAU C AUUUUCAG 181 CUGAAAAU CUGAUGAGGCCGUUAGGCCGAA AUAAAAAU 408
    1324 UUUAUCAU U UUCAGAUC 182 GAUCUGAA CUGAUGAGGCCGUUAGGCCGAA AUGAUAAA 409
    1325 UUAUCAUU U UCAGAUCU 183 AGAUCUGA CUGAUGAGGCCGUUAGGCCGAA AAUGAUAA 410
    1326 UAUCAUUU U CAGAUCUC 184 GAGAUCUG CUGAUGAGGCCGUUAGGCCGAA AAAUGAUA 411
    1327 AUCAUUUU C AGAUCUCC 185 GGAGAUCU CUGAUGAGGCCGUUAGGCCGAA AAAAUGAU 412
    1332 UUUCAGAU C UCCAGUAU 186 AUACUGGA CUGAUGAGGCCGUUAGGCCGAA AUCUGAAA 413
    1334 UCAGAUCU C CAGUAUUU 187 AAAUACUG CUGAUGAGGCCGUUAGGCCGAA AGAUCUGA 414
    1339 UCUCCAGU A UUUCGGAU 188 AUCCGAAA CUGAUGAGGCCGUUAGGCCGAA ACUGGAGA 415
    1341 UCCAGUAU U UCGGAUAU 189 AUAUCCGA CUGAUGAGGCCGUUAGGCCGAA AUACUGGA 416
    1342 CCAGUAUU U CGGAUAUU 190 AAUAUCCG CUGAUGAGGCCGUUAGGCCGAA AAUACUGG 417
    1343 CAGUAUUU C GGAUAUUU 191 AAAUAUCC CUGAUGAGGCCGUUAGGCCGAA AAAUACUG 418
    1348 UUUCGGAU A UUUUUUCA 192 UGAAAAAA CUGAUGAGGCCGUUAGGCCGAA AUCCGAAA 419
    1350 UCGGAUAU U UUUUCACA 193 UGUGAAAA CUGAUGAGGCCGUUAGGCCGAA AUAUCCGA 420
    1351 CGGAUAUU U UUUCACAA 194 UUGUGAAA CUGAUGAGGCCGUUAGGCCGAA AAUAUCCG 421
    1352 GGAUAUUU U UUCACAAG 195 CUUGUGAA CUGAUGAGGCCGUUAGGCCGAA AAAUAUCC 422
    1353 GAUAUUUU U UCACAAGA 196 UCUUGUGA CUGAUGAGGCCGUUAGGCCGAA AAAAUAUC 423
    1354 AUAUUUUU U CACAAGAU 197 AUCUUGUG CUGAUGAGGCCGUUAGGCCGAA AAAAAUAU 424
    1355 UAUUUUUU C ACAAGAUU 198 AAUCUUGU CUGAUGAGGCCGUUAGGCCGAA AAAAAAUA 425
    1363 CACAAGAU U UUCAUUAG 199 CUAAUGAA CUGAUGAGGCCGUUAGGCCGAA AUCUUGUG 426
    1364 ACAAGAUU U UCAUUAGA 200 UCUAAUGA CUGAUGAGGCCGUUAGGCCGAA AAUCUUGU 427
    1365 CAAGAUUU U CAUUAGAC 201 GUCUAAUG CUGAUGAGGCCGUUAGGCCGAA AAAUCUUG 428
    1366 AAGAUUUU C AUUAGACC 202 CGUCUAAU CUGAUGAGGCCGUUACGCCGAA AAAAUCUU 429
    1369 AUUUUCAU U AGACCUCU 203 AGAGGUCU CUGAUGAGGCCGUUAGGCCGAA AUGAAAAU 430
    1370 UUUUCAUU A GACCUCUU 204 AAGAGGUC CUGAUGAGGCCGUUAGGCCGAA AAUGAAAA 431
    1376 UUAGACCU C UUAGGUAC 205 GUACCUAA CUGAUGAGGCCGUUAGGCCGAA AGGUCUAA 432
    1378 AGACCUCU U AGGUACAG 206 CUGUACCU CUGAUGAGGCCGUUAGGCCGAA AGAGGUCU 433
    1379 GACCUCUU A GGUACAGG 207 CCUGUACC CUGAUGAGGCCGUUAGGCCGAA AAGAGGUC 434
    1383 UCUUAGGU A CAGGAGCC 208 GGCUCCUG CUGAUGAGGCCGUUAGGCCGAA ACCUAAGA 435
    1403 GCAGCAAU U CCACUAAC 209 GUUAGUGG CUGAUGAGGCCGUUAGGCCGAA AUUGCUGC 436
    1404 CAGCAAUU C CACUAACA 210 UGUUAGUG CUGAUGAGGCCGUUAGGCCGAA AAUUGCUG 437
    1409 AUUCCACU A ACAUGGAA 211 UUCCAUGU CUGAUGAGGCCGUUAGGCCGAA AGUGGAAU 438
    1419 CAUGGAAU C CAGUCUGU 212 ACAGACUG CUGAUGAGGCCGUUAGGCCGAA AUUCCAUG 439
    1424 AAUCCAGU C UGUGACAG 213 CUGUCACA CUGAUGAGGCCGUUAGGCCGAA ACUGGAUU 440
    1436 GACAGUGU U UUUCACUC 214 GAGUGAAA CUGAUGAGGCCGUUAGGCCGAA ACACUGUC 441
    1437 ACAGUGUU U UUCACUCU 215 AGAGUGAA CUGAUGAGGCCGUUAGGCCGAA AACACUGU 442
    1438 CAGUGUUU U UCACUCUG 216 CAGAGUGA CUGAUGAGGCCGUUAGGCCGAA AAACACUG 443
    1439 AGUGUUUU U CACUCUGU 217 ACAGAGUG CUGAUGAGGCCGUUAGGCCGAA AAAACACU 444
    1440 GUGUUUUU C ACUCUGUG 218 CACAGAGU CUGAUGAGGCCGUUAGGCCGAA AAAAACAC 445
    1444 UUUUCACU C UGUGGUAA 219 UUACCACA CUGAUGAGGCCGUUAGGCCGAA AGUGAAAA 446
    1451 UCUGUGGU A AGCUGAGG 220 CCUCAGCU CUGAUGAGGCCGUUAGGCCGAA ACCACAGA 447
    1463 UGAGGAAU A UGUCACAU 221 AUGUGACA CUGAUGAGGCCGUUAGGCCGAA AUUCCUCA 448
    1467 GAAUAUGU C ACAUUUUC 222 GAAAAUGU CUGAUGAGGCCGUUAGGCCGAA ACAUAUUC 449
    1472 UGUCACAU U UUCAGUCA 223 UGACUGAA CUGAUGAGGCCGUUAGGCCGAA AUGUGACA 450
    1473 GUCACAUU U UCAGUCAA 224 UUGACUGA CUGAUGAGGCCGUUAGGCCGAA AAUGUGAC 451
    1474 UCACAUUU U CAGUCAAA 225 UUUGACUG CUGAUGAGGCCGUUAGGCCGAA AAAUGUGA 452
    1475 CACAUUUU C AGUCAAAG 226 CUUUGACU CUGAUGAGGCCGUUAGGCCGAA AAAAUGUG 453
    1479 UUUUCAGU C AAAGAACC 227 GGUUCUUU CUGAUGAGGCCGUUAGGCCGAA ACUGAAAA 454
  • [0231]
    TABLE IV
    Human PTGDR Inozyme and Substrate Sequence
    Seq Seq
    Pos Substrate ID Inozyme ID
    11 AUUCUGGC U AUUUUCCU 455 AGGAAAAU CUGAUGAGGCCGUUAGGCCGAA ICCAGAAU 831
    18 CUAUUUUC C UCCUGCCG 456 CGGCAGGA CUGAUGAGGCCGUUAGGCCGAA IAAAAUAG 832
    19 UAUUUUCC U CCUGCCGU 457 ACGGCAGG CUGAUGAGGCCGUUAGGCCGAA IGAAAAUA 833
    21 UUUUCCUC C UGCCGUUC 458 GAACGGCA CUGAUGAGGCCGUUAGGCCGAA IAGGAAAA 834
    22 UUUCCUCC U GCCGUUCC 459 GGAACGGC CUGAUGAGGCCGUUAGGCCGAA IGAGGAAA 835
    25 CCUCCUGC C GUUCCGAC 460 GUCGGAAC CUGAUCACGCCGUUAGGCCCAA ICAGGAGG 836
    30 UGCCGUUC C GACUCCUC 461 GCCGAGUC CUGAUGAGGCCGUUAGGCCGAA IAACGGCA 837
    34 GUUCCGAC U CGGCACCA 462 UGGUGCCG CUGAUGAGGCCGUUAGGCCGAA IUCGGAAC 838
    39 GACUCGGC A CCAGAGUC 463 GACUCUGG CUGAUGAGGCCGUUAGGCCGAA ICCGAGUC 839
    41 CUCGGCAC C AGAGUCUG 464 CAGACUCU CUGAUGAGGCCGUUAGGCCGAA IUGCCGAG 840
    42 UCGGCACC A GAGUCUGU 465 ACAGACUC CUGAUGAGGCCGUUAGGCCGAA IGUGCCGA 841
    48 CCAGAGUC U GUCUCUAC 466 GUAGAGAC CUGAUGAGGCCGUUAGGCCGAA IACUCUGG 842
    52 AGUCUGUC U CUACUGAG 467 CUCAGUAG CUGAUGAGGCCGUUAGGCCGAA IACAGACU 843
    54 UCUGUCUC U ACUGAGAA 468 UUCUCAGU CUGAUGAGGCCGUUAGGCCGAA IAGACAGA 844
    57 GUCUCUAC U GAGAACGC 469 GCGUUCUC CUGAUGAGGCCGUUAGGCCGAA IUAGAGAC 845
    66 GAGAACGC A GCGCGUCA 470 UGACGCGC CUGAUGAGGCCGUUAGGCCGAA ICGUUCUC 846
    74 AGCGCGUC A GGGCCGAG 471 CUCGGCCC CUGAUGAGGCCGUUAGGCCGAA IACGCGCU 847
    79 GUCAGGGC C GAGCUCUU 472 AAGAGCUC CUGAUGAGGCCGUUAGGCCGAA ICCCUGAC 848
    84 GGCCGAGC U CUUCACUG 473 CAGUGAAG CUGAUGAGGCCGUUAGGCCGAA ICUCGGCC 849
    86 CCGAGCUC U UCACUGGC 474 GCCAGUGA CUGAUGAGGCCGUUAGGCCGAA IAGCUCGG 850
    89 AGCUCUUC A CUGGCCUG 475 CAGGCCAG CUGAUGAGGCCGUUAGGCCGAA IAAGAGCU 851
    91 CUCUUCAC U GGCCUGCU 476 AGCAGGCC CUGAUGAGGCCGUUAGGCCGAA IUGAAGAG 852
    95 UCACUGGC C UGCUCCGC 477 GCGGAGCA CUGAUGAGGCCGUUAGGCGGAA ICCAGUGA 853
    96 CACUGGCC U GCUCCGCG 478 CGCGGAGC CUGAUGAGGCCGUUAGGCCGAA IGCCAGUG 854
    99 UGGCCUGC U CCGCGCUC 479 GAGCGCGG CUGAUGAGGCCGUUAGGCCGAA ICAGGCCA 855
    101 GCCUGCUC C GCGCUCUU 480 AAGAGCGC CUGAUGAGGCCGUUAGGCCGAA IAGCAGGC 856
    106 CUCCGCGC U CUUCAAUG 481 CAUUGAAG CUGAUGAGGCCGUUAGGCCGAA ICGCGGAG 857
    108 CCGCGCUC U UCAAUGCC 482 GGCAUUGA CUGAUGAGGCCGUUAGGCCGAA IAGCGCGG 858
    111 CGCUCUUC A AUGCCAGC 483 GCUGGCAU CUGAUGAGGCCGUUAGGCCGAA IAAGAGCG 859
    116 UUCAAUGC C AGCGCCAG 484 CUGGCGCU CUGAUGAGGCCGUUAGGCCGAA ICAUUGAA 860
    117 UCAAUGCC A GCGCCAGG 485 CCUGGCGC CUGAUGAGGCCGUUAGGCCGAA IGCAUUGA 861
    122 GCCAGCGC C AGGCGCUC 486 GAGCGCCU CUGAUGAGGCCGUUAGGCCGAA ICGCUGGC 862
    123 CCAGCGCC A GGCGCUCA 487 UGAGCGCC CUGAUGAGGCCGUUAGGCCGAA IGCGCUGG 863
    129 CCAGGCGC U CACCCUGC 488 GCAGGGUG CUGAUGAGGCCGUUAGGCCGAA ICGCCUGG 864
    131 AGGCGCUC A CCCUGCAG 489 CUGCAGGG CUGAUGAGGCCGUUAGGCCGAA IAGCGCCU 865
    133 GCGCUCAC C CUGCAGAG 490 CUCUGCAG CUGAUGAGGCCGUUAGGCCGAA IUGAGCGC 866
    134 CGCUCACC C UGCAGAGC 491 GCUCUGCA CUGAUGAGGCCGUUAGGCCGAA IGUGAGCG 867
    135 GCUCACCC U GCAGAGCG 492 CGCUCUGC CUGAUGAGGCCGUUAGGCCGAA IGGUGAGC 868
    138 CACCCUGC A GAGCGUCC 493 GGACGCUC CUGAUGAGGCCGUUAGGCCGAA ICAGGGUG 869
    146 AGAGCGUC C CGCCUCUC 494 GAGAGGCG CUGAUGAGGCCGUUAGGCCGAA IACGCUCU 870
    147 GAGCGUCC C GCCUCUCA 495 UGAGAGGC CUGAUGAGGCCGUUAGGCCGAA IGACGCUC 871
    150 CGUCCCGC C UCUCAAAG 496 CUUUGAGA CUGAUGAGGCCGUUAGGCCGAA ICGGGACG 872
    151 GUCCCGCC U CUCAAAGA 497 UCUUUGAG CUGAUGAGGCCGUUAGGCCGAA IGCGGGAC 873
    153 CCCGCCUC U CAAAGAGG 498 CCUCUUUG CUGAUGAGGCCGUUAGGCCGAA IAGGCGGG 874
    155 CGCCUCUC A AAGAGGGG 499 CCCCUCUU CUGAUGAGGCCGUUAGGCCGAA IAGAGGCG 875
    170 GGUGUGAC C CGCGAGUU 500 AACUCGCG CUGAUGAGGCCGUUAGGCCGAA IUCACACC 876
    171 GUGUGACC C GCGAGUUU 501 AAACUCGC CUGAUGAGGCCGUUAGGCCGAA IGUCACAC 877
    193 GGAGGUUC C UGCCCUCG 502 CCACGGCA CUCAUGAGGCCGUUAGGCCGAA IAACCUCC 878
    194 GACGUUCC U GCCGUGGG 503 CCCACGGC CUGAUGAGGCCGUUAGGCCGAA IGAACCUC 879
    197 GUUCCUGC C GUGGGGAA 504 UUCCCCAC CUGAUGAGGCCGUUAGGCCGAA ICAGGAAC 880
    207 UGCGGAAC A CCCCGCCG 505 CCGCGGGG CUGAUGAGGCCGUUAGGCCGAA IUUCCCCA 881
    209 GGGAACAC C CCGCCGCC 506 GGCGGCGG CUGAUGAGGCCGUUAGGCCGAA IUGUUCCC 882
    210 GGAACACC C CGCCGCCC 507 GGGCCGCG CUGAUGAGGCCGUUAGGCCGAA IGUGUUCC 883
    211 GAACACCC C GCCGCCCU 508 AGGGCGGC CUCAUGAGGCCGUUAGGCCGAA IGGUGUUC 884
    214 CACCCCGC C GCCCUCGG 509 CCGAGGGC CUGAUGAGGCCGUUAGGCCGAA ICGGGGUG 885
    217 CCCGCCCC C CUCGGAGC 510 GCUCCGAG CUGAUGAGGCCGUUAGGCCGAA ICGGCGGG 886
    218 CCGCCGCC C UCGGAGCU 511 AGCUCCGA CUGAUGAGGCCGUUAGGCCGAA IGCGGCGG 887
    219 CGCCGCCC U CGGAGCUU 512 AAGCUCCG CUGAUGAGGCCGUUAGGCCGAA IGGCGGCG 888
    226 CUCGGAGC U UUUUCUGU 513 ACAGAAAA CUGAUGAGGCCGUUAGGCCGAA ICUCCGAG 889
    232 GCUUUUUC U GUGGCGCA 514 UGCGCCAC CUGAUGAGGCCGUUAGGCCGAA IAAAAAGC 890
    240 UGUGGCGC A GCUUCUCC 515 GGAGAAGC CUGAUGAGGCCGUUAGGCCGAA ICGCCACA 891
    243 GGCGCAGC U UCUCCGCC 516 GGCGGAGA CUGAUGAGGCCGUUAGGCCGAA ICUGCGCC 892
    246 GCAGCUUC U CCGCCCGA 517 UCGGGCGG CUGAUGAGGCCGUUAGGCCGAA IAAGCUGC 893
    248 AGCUUCUC C GCCCGAGC 518 GCUCGGGC CUGAUGAGGCCGUUAGGCCGAA IAGAAGCU 894
    251 UUCUCCGC C CGAGCCGC 519 GCGGCUCG CUGAUGAGGCCGUUAGGCCGAA ICGGAGAA 895
    252 UCUCCGCC C GAGCCGCG 520 CGCGGCUC CUGAUGAGGCCGUUAGGCCGAA ICCUGAGA 896
    257 GCCCGAGC C GCGCGCGG 521 CCGCGCGC CUGAUGAGGCCGUUAGGCCGAA ICUCGGGC 897
    269 CGCGGAGC U GCCGGGGG 522 CCCCCGGC CUGAUGAGGCCGUUAGGCCGAA ICUCCGCG 898
    272 GGAGCUGC C GGGGGCUC 523 GAGCCCCC CUGAUGAGGCCGUUAGGCCGAA ICAGCUCC 899
    279 CCGGGGGC U CCUUAGCA 524 UGCUAAGG CUGAUGAGGCCGUUAGGCCGAA ICCCCCGG 900
    281 GGGGGCUC C UUAGCACC 525 GGUGCUAA CUGAUGAGGCCGUUAGGCCGAA IAGCCCCC 901
    282 GGGGCUCC U UAGCACCC 526 GGGUGCUA CUGAUGAGGCCGUUAGGCCGAA IGAGCCCC 902
    287 UCCUUAGC A CCCGGGCG 527 CGCCCGGG CUGAUGAGGCCGUUAGGCCGAA ICUAAGGA 903
    289 CUUAGCAC C CGGGCGCC 528 GGCGCCCG CUGAUGAGGCCGUUAGGCCGAA IUGCUAAG 904
    290 UUAGCACC C GGGCGCCG 529 CGGCGCCC CUGAUGAGGCCGUUAGGCCGAA IGUGCUAA 905
    297 CCGGGCGC C GGGGCCCU 530 AGGGCCCC CUGAUGAGGCCGUUAGGCCGAA ICGCCCGG 906
    303 GCCGGGGC C CUCGCCCU 531 AGGGCGAG CUGAUGAGGCCGUUAGGCCGAA ICCCCGGC 907
    304 CCGGGGCC C UCGCCCUU 532 AAGGGCGA CUGAUGAGGCCGUUAGGCCGAA IGCCCCGG 908
    305 CGGGGCCC U CGCCCUUC 533 GAAGGGCG CUGAUGAGGCCGUUAGGCCGAA IGGCCCCG 909
    309 GCCCUCGC C CUUCCGCA 534 UGCGGAAG CUGAUGAGGCCGUUAGGCCGAA ICGAGGGC 910
    310 CCCUCGCC C UUCCGCAG 535 CUGCGGAA CUGAUGAGGCCGUUAGGCCGAA IGCGAGGG 911
    311 CCUCGCCC U UCCGCAGC 536 GCUGCGGA CUGAUGAGGCCGUUAGGCCGAA IGGCGAGG 912
    314 CGCCCUUC C GCAGCCUU 537 AAGGCUGC CUGAUGAGGCCGUUAGGCCGAA IAAGGGCG 913
    317 CCUUCCGC A GCCUUCAC 538 GUGAAGGC CUGAUGAGGCCGUUAGGCCGAA ICGGAAGG 914
    320 UCCGCAGC C UUCACUCC 539 GGAGUGAA CUGAUGAGGCCGUUAGGCCGAA ICUGCGGA 915
    321 CCGCAGCC U UCACUCCA 540 UGGAGUGA CUGAUGAGGCCGUUAGGCCGAA IGCUGCGG 916
    324 CAGCCUUC A CUCCAGCC 541 GGCUGGAG CUGAUGAGGCCGUUAGGCCGAA IAAGGCUG 917
    326 GCCUUCAC U CCAGCCCU 542 AGGGCUGG CUGAUGAGGCCGUUAGGCCGAA IUGAAGGC 918
    328 CUUCACUC C AGCCCUCU 543 AGAGGGCU CUGAUGAGGCCGUUAGGCCGAA IAGUGAAG 919
    329 UUCACUCC A GCCCUCUG 544 CAGAGGGC CUGAUGAGGCCGUUAGGCCGAA IGAGUGAA 920
    332 ACUCCAGC C CUCUGCUC 545 GAGCAGAG CUGAUGAGGCCGUUAGGCCGAA ICUGGAGU 921
    333 CUCCAGCC C UCUGCUCC 546 GGAGCAGA CUGAUGAGGCCGUUAGGCCGAA IGCUGGAG 922
    334 UCCAGCCC U CUGCUCCC 547 GGGAGCAG CUGAUGAGGCCGUUAGGCCGAA IGGCUGGA 923
    336 CAGCCCUC U GCUCCCGC 548 GCGGGAGC CUGAUGAGGCCGUUAGGCCGAA IAGGGCUG 924
    339 CCCUCUGC U CCCGCACG 549 CGUGCGGG CUGAUGAGGCCGUUAGGCCGAA ICAGAGGG 925
    341 CUCUGCUC C CGCACGCC 550 GGCGUGCG CUGAUGAGGCCGUUAGGCCGAA IAGCAGAG 926
    342 UCUGCUCC C GCACGCCA 551 UGGCGUGC CUGAUGAGGCCGUUAGGCCGAA IGAGCAGA 927
    345 GCUCCCGC A CGCCAUGA 552 UCAUGGCG CUGAUGAGGCCGUUAGGCCGAA ICGGGAGC 928
    349 CCGCACGC C AUGAAGUC 553 GACUUCAU CUGAUGACCCCGUUAGGCCGAA ICGUGCGC 929
    350 CGCACGCC A UGAAGUCG 554 CGACUUCA CUGAUGAGGCCGUUAGGCCGAA IGCGUGCG 930
    360 GAAGUCGC C GUUCUACC 555 GGUAGAAC CUGAUGAGGCCGUUAGGCCGAA ICGACUUC 931
    365 CGCCGUUC U ACCGCUGC 556 GCACCGGU CUGAUGAGGCCGUUAGGCCGAA IAACGGCG 932
    368 CGUUCUAC C GCUGCCAG 557 CUGGCAGC CUGAUGAGGCCGUUAGGCCGAA IUAGAACG 933
    371 UCUACCUC U GCCAGAAC 558 GUUCUGGC CUGAUGAGGCCGUUAGGCCGAA ICOGUAGA 934
    374 ACCGCUGC C AGAACACC 559 GGUGUUCU CUGAUGAGGCCGUUAGGCCGAA ICAGCGGU 935
    375 CCGCUGCC A GAACACCA 560 UGGUGUUC CUGAUGAGGCCGUUAGGCCGAA IGCAGCGG 936
    380 GCCAGAAC A CCACCUCU 561 AGAGGUGG CUGAUGAGGCCGUUAGGCCGAA IUUCUGGC 937
    382 CAGAACAC C ACCUCUGU 562 ACAGAGGU CUGAUGAGGCCGUUAGGCCGAA IUGUUCUG 938
    383 AGAACACC A CCUCUGUG 563 CACAGAGG CUGAUGAGGCCGUUAGGCCGAA IGUGUUCU 939
    385 AACACCAC C UCUGUGGA 564 UCCACAGA CUGAUGAGGCCGUUAGGCCGAA IUGGUGUU 940
    386 ACACCACC U CUGUGGAA 565 UUCCACAG CUGAUGAGGCCGUUAGGCCGAA IGUGGUGU 941
    388 ACCACCUC U GUGGAAAA 566 UUUUCCAC CUGAUGAGGCCGUUAGGCCGAA IAGGUGGU 942
    401 AAAAAGGC A ACUCGGCG 567 CGCCGAGU CUGAUGAGGCCGUUAGGCCGAA ICCUUUUU 943
    404 AAGGCAAC U CGGCGGUG 568 CACCGCCG CUGAUGAGGCCGUUAGGCCGAA IUUGCCUU 944
    426 CGGGGUGC U CUUCAGCA 569 UGCUGAAG CUGAUGAGGCCGUUAGGCCGAA ICACCCCG 945
    428 GGGUGCUC U UCAGCACC 570 GGUGCUGA CUGAUGAGGCCGUUAGGCCGAA IAGCACCC 946
    431 UGCUCUUC A GCACCGGC 571 GCCGGUGC CUGAUGAGGCCGUUAGGCCGAA IAAGAGCA 947
    434 UCUUCAGC A CCGGCCUC 572 GAGGCCGG CUGAUGAGGCCGUUAGGCCGAA ICUGAAGA 948
    436 UUCAGCAC C GGCCUCCU 573 AGGAGGCC CUGAUGAGGCCGUUAGGCCGAA IUGCUGAA 949
    440 GCACCGGC C UCCUGGGC 574 GCCCAGGA CUGAUGAGGCCGUUAGGCCGAA ICCGGUGC 950
    441 CACCGGCC U CCUGGGCA 575 UGCCCAGG CUGAUGAGGCCGUUAGGCCGAA IGCCGGUG 951
    443 CCGGCCUC C UGGGCAAC 576 GUUGCCCA CUGAUGAGGCCGUUAGGCCGAA IAGGCCGG 952
    444 CGGCCUCC U GGGCAACC 577 GGUUGCCC CUGAUGAGGCCGUUAGGCCGAA IGAGGCCG 953
    449 UCCUGGGC A ACCUGCUG 578 CAGCAGGU CUGAUGAGGCCGUUAGGCCGAA ICCCAGGA 954
    452 UGGGCAAC C UGCUGGCC 579 GGCCAGCA CUGAUGAGGCCGUUAGGCCGAA IUUGCCCA 955
    453 GGGCAACC U GCUGGCCC 580 GGGCCAGC CUGAUGAGGCCGUUAGGCCGAA IGUUGCCC 956
    456 CAACCUGC U GGCCCUGG 581 CCAGGGCC CUGAUGAGGCCGUUAGGCCGAA ICAGGUUG 957
    460 CUGCUGGC C CUGGGGCU 582 AGCCCCAG CUGAUGAGGCCGUUAGGCCGAA ICCAGCAG 958
    461 UGCUGGCC C UGGGGCUG 583 CAGCCCCA CUGAUGAGGCCGUUAGGCCGAA IGCCAGCA 959
    462 GCUGGCCC U GGGGCUGC 584 GCAGCCCC CUGAUGAGGCCGUUAGGCCGAA IGGCCAGC 960
    468 CCUGGGGC U GCUGGCGC 585 GCGCCAGC CUGAUGAGGCCGUUAGGCCGAA ICCCCAGG 961
    471 GGGGCUGC U GGCGCGCU 586 AGCGCGCC CUGAUGAGGCCGUUAGGCCGAA ICAGCCCC 962
    479 UGGCGCGC U CGGGGCUG 587 CAGCCCCG CUGAUGAGGCCGUUAGGCCGAA ICGCGCCA 963
    486 CUCGGGGC U GGGGUGGU 588 ACCACCCC CUGAUGAGGCCGUUAGGCCGAA ICCCCGAG 964
    497 GGUGGUGC U CGCGGCGU 589 ACGCCGCG CUGAUGAGGCCGUUAGGCCGAA ICACCACC 965
    507 GCGGCGUC C ACUGCGCC 590 GGCGCAGU CUGAUGAGGCCGUUAGGCCGAA IACGCCGC 966
    508 CGGCGUCC A CUGCGCCC 591 GGGCGCAG CUGAUGAGGCCGUUAGGCCGAA IGACGCCG 967
    510 GCGUCCAC U GCGCCCGC 592 GCGGGCGC CUGAUGAGGCCGUUAGGCCGAA IUGGACGC 968
    515 CACUGCGC C CGCUGCCC 593 GGGCAGCG CUGAUGAGGCCGUUAGGCCGAA ICGCAGUG 969
    516 ACUGCGCC C GCUGCCCU 594 AGGGCAGC CUGAUGAGGCCGUUAGGCCGAA IGCGCAGU 970
    519 GCGCCCGC U GCCCUCGG 595 CCGAGGGC CUGAUGAGGCCGUUAGGCCGAA ICGGGCGC 971
    522 CCCGCUGC C CUCGGUCU 596 AGACCGAG CUGAUGAGGCCGUUAGGCCGAA ICAGCGGG 972
    523 CCGCUGCC C UCGGUCUU 597 AAGACCGA CUGAUGAGGCCGUUAGGCCGAA IGCAGCGG 973
    524 CGCUGCCC U CGGUCUUC 598 GAAGACCG CUGAUGAGGCCGUUAGGCCGAA IGGCAGCG 974
    530 CCUCGGUC U UCUACAUG 599 CAUGUAGA CUGAUGAGGCCGUUAGGCCGAA IACCGAGG 975
    533 CGGUCUUC U ACAUGCUG 600 CAGCAUGU CUGAUGAGGCCGUUAGGCCGAA IAAGACCG 976
    536 UCUUCUAC A UGCUGGUG 601 CACCAGCA CUGAUGAGGCCGUUAGGCCGAA IUAGAAGA 977
    540 CUACAUGC U GGUGUGUG 602 CACACACC CUGAUGAGGCCGUUAGGCCGAA ICAUGUAG 978
    551 UGUGUGGC C UGACGGUC 603 GACCGUCA CUGAUGAGGCCGUUAGGCCGAA ICCACACA 979
    552 GUGUGGCC U GACGGUCA 604 UGACCGUC CUGAUGAGGCCGUUAGGCCGAA IGCCACAC 980
    560 UGACGGUC A CCGACUUG 605 CAAGUCGG CUGAUGAGGCCGUUAGGCCGAA IACCGUCA 981
    562 ACGGUCAC C GACUUCCU 606 AGCAAGUC CUGAUCAGGCCGUUAGGCCGAA IUGACCGU 982
    566 UCACCGAC U UGCUGGGC 607 GCCCAGCA CUGAUGAGGCCGUUAGGCCGAA IUCCGUGA 983
    570 CGACUUCC U GCCCAAGU 608 ACUUGCCC CUGAUGAGGCCGUUAGGCCGAA ICAAGUCG 984
    575 UGCUGGGC A AGUGCCUC 609 GAGGCACU CUGAUGAGGCCGUUAGGCCGAA ICCCAGCA 985
    581 CCAAGUGC C UCCUAAGC 610 GCUUACGA CUGAUGAGGCCGUUAGGCCGAA ICACUUGC 986
    582 CAAGUGCC U CCUAAGCC 611 GGCUUAGG CUGAUCACGCCGUUAGGCCGAA IGCACUUG 987
    584 AGUGCCUC C UAAGCCCG 612 CGGGCUUA CUGAUGAGGCCGUUAGGCCGAA IAGGCACU 988
    585 GUGCCUCC U AAGCCCGG 613 CCGGGCUU CUGAUGAGGCCGUUAGGCCGAA IGAGGCAC 989
    590 UCCUAAGC C CGGUGGUG 614 CACCACCG CUGAUGAGGCCGUUAGGCCGAA ICUUAGGA 990
    591 CCUAAGCC C CGUGGUGC 615 GCACCACC CUGAUGAGGCCGUUAGGCCGAA IGCUUAGG 991
    600 GGUGGUGC U GGCUGCCU 616 AGGCAGCC CUGAUGAGGCCGUUAGGCCGAA ICACCACC 992
    604 GUGCUCGC U CCCUACGC 617 GCGUAGGC CUCAUCAGGCCUUUAGGCCGAA ICCAGCAC 993
    607 CUGGCUGC C UACGCUCA 618 UGAGCGUA CUGAUGAGGCCGUUAGGCCCAA ICAGCCAG 994
    608 UCGCUGCC U ACGCUCAG 619 CUGACCGU CUGAUGAGGCCGUUAGGCCGAA IGCAGCCA 995
    613 GCCUACGC U CAGAACCC 620 CGGUUCUG CUGAUGAGGCCGUUAGGCCGAA ICGUAGGC 996
    615 CUACGCUC A GAACCGGA 621 UCCGGUUC CUGAUGAGGCCGUUAGGCCGAA IAGCGUAG 997
    620 CUCAGAAC C GGAGUCUG 622 CAGACUCC CUGAUGAGGCCGUUAGGCCGAA IUUCUGAG 998
    627 CCGGAGUC U GCGGGUGC 623 GCACCCGC CUGAUGAGGCCGUUAGGCCGAA IACUCCGG 999
    636 GCCGGUGC U UGCGCCCG 624 CGGGCCCA CUGAUGAGGCCGUUAGGCCGAA ICACCCCC 1000
    642 GCUUGCGC C CGCAUUGG 625 CCAAUGCG CUGAUGAGGCCGUUAGGCCGAA ICGCAAGC 1001
    643 CUUCCGCC C GCAUUGGA 626 UCCAAUGC CUGAUGAGGCCGUUAGGCCGAA IGCGCAAC 1002
    646 GCGCCCGC A UUGGACAA 627 UUGUCCAA CUGAUGAGGCCGUUACGCCGAA ICGGGCGC 1003
    653 CAUUGCAC A ACUCGUUG 628 CAACGAGU CUGAUGAGGCCGUUAGGCCGAA IUCCAAUG 1004
    656 UGGACAAC U CGUUGUGC 629 GCACAACG CUGAUGAGGCCGUUAGGCCGAA IUUGUCCA 1005
    665 CGUUGUGC C AAGCCUUC 630 CAAGCCUU CUGAUGAGGCCGUUAGGCCGAA ICACAACG 1006
    666 GUUGUGCC A AGCCUUCG 631 CGAAGGCU CUGAUGAGGCCGUUAGGCCGAA IGCACAAC 1007
    670 UGCCAAGC C UUCGCCUU 632 AAGGCGAA CUGAUGAGGCCGUUACGCCCAA ICUUGGCA 1008
    671 GCCAAGCC U UCGCCUUC 633 GAACGCGA CUGAUGAGGCCGUUAGGCCGAA IGCUUCGC 1009
    676 GCCUUCGC C UUCUUCAU 634 AUGAAGAA CUGAUGAGCCCGUUAGGCCGAA ICGAAGGC 1010
    677 CCUUCGCC U UCUUCAUG 635 CAUGAAGA CUGAUGAGGCCGUUAGGCCGAA IGCGAAGG 1011
    680 UCGCCUUC U UCAUGUCC 636 GGACAUGA CUGAUGAGGCCGUUAGGCCGAA IAAGGCGA 1012
    683 CCUUCUUC A UGUCCUUC 637 GAAGGACA CUGAUGAGGCCGUUAGGCCGAA IAAGAAGG 1013
    688 UUCAUGUC C UUCUUUGG 638 CCAAAGAA CUGAUGAGGCCGUUAGGCCGAA IACAUGAA 1014
    689 UCAUGUCC U UCUUUGGG 639 CCCAAAGA CUGAUGAGGCCGUUAGGCCGAA IGACAUGA 1015
    692 UGUCCUUC U UUGGGCUC 640 GAGCCCAA CUGAUGAGGCCGUUAGGCCGAA IAAGGACA 1016
    699 CUUUGGGC U CUCCUCCA 641 UCGAGGAG CUGAUGAGGCCGUUAGGCCGAA ICCCAAAG 1017
    701 UUGGGCUC U CCUCGACA 642 UGUCGAGG CUGAUGAGGCCGUUAGGCCGAA IAGCCCAA 1018
    703 GGGCUCUC C UCGACACU 643 AGUGUCGA CUGAUGAGGCCGUUAGGCCGAA IAGAGCCC 1019
    704 GGCUCUCC U CGACACUG 644 CAGUGUCG CUGAUGAGGCCGUUAGGCCGAA IGAGAGCC 1020
    709 UCCUCGAC A CUGCAACU 645 AGUUGCAG CUGAUGAGGCCGUUAGGCCGAA IUCGAGGA 1021
    721 CUCGACAC U GCAACUCC 646 GGAGUUGC CUGAUGAGGCCGUUAGGCCGAA IUGUCGAG 1022
    714 GACACUGG A ACUCCUGG 647 CCAGGAGU CUGAUGAGGCCGUUAGGCCGAA ICAGUGUC 1023
    717 ACUGCAAC U CCUGGCCA 648 UGGCCAGG CUGAUGAGGCCGUUAGGCCGAA IUUGCAGU 1024
    719 UGCAACUC C UGGCCAUG 649 CAUGGCCA CUGAUGAGGCCGUUAGGCCGAA IAGUUGCA 1025
    720 GCAACUCC U GGCCAUGG 650 CCAUGGCC CUGAUGAGGCCGUUAGGCCGAA IGAGUUGC 1026
    724 CUCCUGGC C AUGGCACU 651 AGUGCCAU CUGAUGAGGCCGUUAGGCCGAA ICCAGGAG 1027
    725 UCCUGGCC A UGGCACUG 652 CAGUGCCA CUGAUGAGGCCGUUAGGCCGAA IGCCAGGA 1028
    730 GCCAUGGC A CUGGAGUG 653 CACUCCAG CUGAUGAGGCCGUUAGGCCGAA ICCAUGGC 1029
    732 CAUGGCAC U GGAGUGCU 654 AGCACUCC CUGAUGAGGCCGUUAGGCCGAA IUGCCAUG 1030
    740 UCGAGUGC U GGCUCUCC 655 GGAGAGCC CUGAUGAGGCCGUUAGGCCGAA ICACUCCA 1031
    744 GUGCUGGC U CUCCCUAG 656 CUAGGGAG CUGAUGAGGCCGUUAGGCCGAA ICCAGCAC 1032
    746 GCUGCCUC U CCCUAGGG 657 CCCUAGGG CUGAUGAGGCCGUUAGGCCGAA IAGCCAGC 1033
    748 UGGCUCUC C CUAGOCCA 658 UGCCCUAG CUGAUGAGGCCGUUAGGCCGAA IAGAGCCA 1034
    749 GGCUCUCC C UAGGGCAC 659 GUCCCCUA CUGAUGAGGCCGUUAGGCCGAA IGAGAGCC 1035
    750 GCUCUCCC U AGGGCACC 660 GGUGCCCU CUGAUGAGGCCGUUAGGCCGAA IGGAGAUC 1036
    756 CCUAGGGC A CCCUUIJCU 661 AGAAAGGG CUGAUGAGGCCGUUAGGCCGAA ICCCUAGG 1037
    758 UAGGGCAC C CUUUCUUC6 62 GAAGAAAG CUGAUGAGGCCGUUAGGCCGAA IUGCCCUA 1038
    759 AGGGCACC C UUUCUUCU 663 AGAAGAAA CUGAUGAGGCCGUUAGGCCGAA IGUGCCCU 1039
    760 GGGCACCC U UUCUUCUA 664 UAGAAGAA CUGAUGAGGCCGUUAGGCCGAA IGGUGCCC 1040
    764 ACCCUUUC U UCUACCGA 665 UCGGUAGA CUGAUGAGGCCGUUAGGCCGAA TAAAGGGU 1041
    767 CUUUCUUC U ACCGACGG 666 CCGUCGGU CUGAUGAGGCCGUUAGGCCGAA IAAGAAAG 1042
    770 UCUUCUAC C GACGGCAC 667 GUGCCGUC CUGAUGAGCCCGUUAGGCCGAA IUAGAAGA 1043
    777 CCGACGGC A CAUCACCC 668 GGGUGAUG CUGAUGAGGCCGUUAGGCCGAA ICCGUCGG 1044
    779 GACGGCAC A UCACCCUG 669 CAGGGUGA CUGAUGAGGCCGUUAGGCCGAA IUGCCGUC 1045
    782 GGCACAUC A CCCUGCGC 670 GCGCAGGG CUGAUGAGGCCGUUAGGCCGAA IAUGUGCC 1046
    784 CACAUCAC C CUGCGCCU 671 AGGCGCAG CUGAUGAGGCCGUUAGGCCGAA TUGAUGUG 1047
    785 ACAUCACC C UGCGCCUG 672 CAGGCGCA CUGAUGAGGCCGUUAGGCCGAA IGUGAUGU 1048
    786 CAUCACCC U GCGCCUGG 673 CCAGGCGC CUGAUGAGGCCGUUAGGCCGAA IGGUGAUG 1049
    791 CCCUGCGC C UGGGCGCA 674 UGCGCCCA CUGAUGAGGCCGUUAGGCCGAA ICGCAGGG 1050
    792 CCUGCGCC U GGGCGCAC 675 GUGCGCCC CUGAUGAGGCCGUUAGGCCGAA IGCGCAGG 1051
    799 CUGGGCGC A CUGGUGGC 676 GCCACCAG CUGAUGAGGCCGUUAGGCCGAA ICGCCCAG 1052
    801 GGGCGCAC U GGUGGCCC 677 GGGCCACC CUGAUGAGGCCGUUAGGCCGAA IUGCGCCC 1053
    808 CUGGUGGC C CCGGUGGU 678 ACCACCGG CUGAUGAGGCCGUUAGGCCGAA ICCACCAG 1054
    809 UGGUGGCC C CGGUGGUG 679 CACCACCG CUGAUGAGGCCGUUAGGCCGAA IGCCACCA 1055
    810 GGUGGCCC C GGUGGUGA 680 UCACCACC CUGAUGAGGCCGUUAGGCCGAA IGGCCACC 1056
    823 GUGAGCGC C UUCUCCCU 681 AGGGAGAA CUGAUGAGGCCGUUAGGCCGAA ICOCUCAC 1057
    824 UGAGCGCC U UCUCCCUG 682 CAUGGAGA CUGAUGAGGCCGUUAGGCCGAA IGCGCUCA 1058
    827 GCGCCUUC U CCCUGGCU 683 AGCCAGGG CUGAUGAGGCCGUUAGGCCGAA IAAGGCGC 1059
    829 GCCUUCUC C CUGGCUUU 684 AAAGCCAG CUGAUGAGGCCGUUAGGCCGAA IAGAAGGC 1060
    830 CCUUCUCC C UGGCUUUC 685 GAAAGCCA CUGAUGAGGCCGUUAGGCCGAA IGAGAAGG 1061
    831 CUUCUCCC U GGCUUUCU 686 AGAAAGCC CUGAUGAGGCCGUUAGGCCGAA IGGAGAAG 1062
    835 UCCCUGGC U UUCUGCGC 687 GCGCAGAA CUGAUGAGGCCGUUAGGCCGAA ICCAGGGA 1063
    839 UGGCUUUC U GCGCGCUA 688 UAGCGCGC CUGAUGAGGCCGUUAGGCCGAA IAAAGCCA 1064
    846 CUGCGCGC U ACCUUUCA 689 UGAAAGGU CUGAUGAGGCCGUUAGGCCGAA ICGCGCAG 1065
    849 CGCGCUAC C UUUCAUGG 690 CCAUGAAA CUGAUGAGGCCGUUAGGCCGAA IUAGCGCG 1066
    850 GCGCUACC U UUCAUGGG 691 CCCAUGAA CUGAUGAGGCCGUUAGGCCGAA IGUAGCGC 1067
    854 UACCUUUC A UGGGCUUC 692 GAAGCCCA CUGAUGAGGCCGUUAGGCCGAA IAAAGGUA 1068
    860 UCAUGGGC U UCGGGAAG 693 CUUCCCGA CUGAUGAGGCCGUUAGGCCGAA ICCCAUGA 1069
    876 GUUCGUGC A GUACUGCC 694 GGCAGUAC CUGAUGAGGCCGUUAGGCCGAA ICACGAAC 1070
    881 UGCAGUAC U GCCCCGGC 695 GCCGGGGC CUGAUGAGGCCGUUAGGCCGAA IUACUGCA 1071
    884 AGUACUGC C CCGGCACC 696 GGUGCCGG CUGAUGAGGCCGUUAGGCCGAA ICAGUACU 1072
    885 GUACUGCC C CGGCACCU 697 AGGUGCCG CUGAUGAGGCCGUUAGGCCGAA IGCAGUAC 1073
    886 UACUGCCC C GGCACCUG 698 CAGGUGCC CUGAUGAGGCCGUUAGGCCGAA IGGCAGUA 1074
    890 GCCCCGGC A CCUGGUGC 699 GCACCAGG CUGAUGAGGCCGUUAGGCCGAA ICCGGGGC 1075
    892 CCCGGCAC C UGGUGCUU 700 AAGCACCA CUGAUGAGGCCGUUAGGCCGAA IUGCCGGG 1076
    893 CCGGCACC U GGUGCLRTU 701 AAAGCACC CUGAUGAGGCCGUUAGGCCGAA IGUGCCGG 1077
    899 CCUGGUGC U UUAUCCAG 702 CUGGAUAA CUGAUGAGGCCGUUAGGCCGAA ICACCAGG 1078
    905 GCUUUAUC C AGAUGGUC 703 GACCAUCU CUGAUGAGGCCGUUAGGCCGAA IAUAAAGC 1079
    906 CUUUAUCC A GAUGGUCC 704 GGACCAUC CUGAUGAGGCCGUUAGGCCGAA IGAUAAAG 1080
    914 AGAUGGUC C ACGAGGAG 705 CUCCUCGU CUGAUGAGGCCGUUAGGCCGAA IACCAUCU 1081
    915 GAUCGUCC A CCAGGAGG 706 CCUCCUCG CUGAUGAGGCCGUUAGGCCGAA IGACCAUC 1082
    926 AGGAGGGC U CGCUGUCG 707 CGACAGCG CUGAUGAGGCCGUUAGGCCGAA ICCCUCCU 1083
    930 GGGCUCGC U GUCGGUGC 708 GCACCGAC CUGAUGAGGCCGUUAGGCCGAA ICGAGCCC 1084
    939 GUCGGUGC U GGGGUACU 709 AGUACCCC CUGAUGAGGCCGUUAGGCCGAA ICACCGAC 1085
    947 UGGGGUAC U CUGUCCUC 710 GAGCACAG CUGAUGAGGCCGUUAGGCCGAA IUACCCCA 1086
    949 GOGUACUC U GUGCUCUA 711 UAGAGCAC CUGAUGAGGCCGUUAGGCCGAA IAGUACCC 1087
    954 CUCUGUGC U CUACUCCA 712 UGGAGUAG CUGAUGAGGCCGUUAGGCCGAA ICACAGAG 1088
    956 CUGUCCUC U ACUCCAGC 713 GCUGGAGU CUGAUGAGGCCGUUAGGCCGAA IAGCACAG 1089
    959 UGCUCUAC U CCAGCCUC 714 GAGGCUGG CUGAUGAGGCCGUUAGGCCGAA IUAGAGCA 1090
    961 CUCUACUC C AGCCUCAU 715 AUGAGGCU CUGAUGAGGCCGUUAGGCCGAA IAGUAGAG 1091
    962 UCUACUCC A GCCUCAUG 716 CAUGAGGC CUGAUGAGGCCGUUAGGCCGAA IGAGUAGA 1092
    965 ACUCCAGC C UCAUGGCG 717 CGCCAUGA CUGAUGAGGCCGUUAGGCCGAA ICUGGAGU 1093
    966 CUCCAGCC U CAUGGCGC 718 GCGCCAUG CUGAUGAGGCCGUUAGGCCGAA IGCUGGAG 1094
    968 CCAGCCUC A UGGCGCUG 719 CAGCGCCA CUGAUGAGGCCGUUAGGCCGAA IAGGCUGG 1095
    975 CAUGGCGC U GCUGGUCC 720 GGACCAGC CUGAUGAGGCCGUUAGGCCGAA ICGCCAUG 1096
    978 GGCGCUGC U GGUCCUCG 721 CGAGGACC CUGAUGAGGCCGUUAGGCCGAA ICAGCGCC 1097
    983 UCCUGGUC C UCGCCACC 722 GGUGGCGA CUGAUGAGGCCGUUAGGCCGAA IACCAGCA 1098
    984 GCUGGUCC U CGCCACCG 723 CGGUGGCG CUGAUGAGGCCGUUAGGCCGAA IGACCAGC 1099
    988 GUCCUCGC C ACCGUGCU 724 AGCACGGU CUGAUGAGGCCGUUAGGCCGAA ICCAGGAC 1100
    989 UCCUCGCC A CCGUGCUG 725 CAGCACGG CUGAUGAGGCCGUUAGGCCGAA IGCGAGGA 1101
    991 CUCGCCAC C GUGCUGUG 726 CACAGCAC CUGAUGAGGCCGUUAGGCCGAA IUGGCGAG 1102
    996 CACCGUGC U GUGCAACC 727 GGUUGCAC CUGAUGAGGCCGUUAGGCCGAA ICACGGUG 1103
    1001 UGCUGUGC A ACCUCGGC 728 GCCGAGGU CUGAUGAGGCCGUUAGGCCGAA ICACAGCA 1104
    1004 UGUGCAAC C UCGGCGCC 729 GGCGCCGA CUGAUGAGGCCGUUAGGCCGAA IUUGCACA 1105
    1005 GUGCAACC U CGGCGCCA 730 UGGCGCCG CUGAUGAGGCCGUUAGGCCGAA IGUUGCAC 1106
    1012 CUCGGCGC C AUGCGCAA 731 UUGCGCAU CUGAUGAGGCCGUUAGGCCGAA ICGCCGAG 1107
    1013 UCGGCGCC A UGCGCAAC 732 GUUGCGCA CUGAUGAGGCCGUUAGGCCGAA IGCGCCGA 1108
    1019 CCAUGCGC A ACCUCUAU 733 AUAGAGGU CUGAUGAGGCCGUUAGGCCGAA ICGCAUGG 1109
    1022 UGCGCAAC C UCUAUGCG 734 CGCAUAGA CUGAUGAGGCCGUUAGGCCGAA IUUGCGCA 1110
    1023 GCGCAACC U CUAUGCGA 735 UCGCAUAG CUGAUGAGGCCGUUAGGCCGAA IGUUGCGC 1111
    1025 GCAACCUC U AUGCGAUG 736 CAUCGCAU CUGAUGAGGCCGUUAGGCCGAA IAGGUUGC 1112
    1035 UGCGAUGC A CCGGCGGC 737 GCCGCCGG CUGAUGAGGCCGUUAGGCCGAA ICAUCGCA 1113
    1037 CGAUGCAC C GGCGGCUG 738 CAGCCGCC CUGAUGAGGCCGUUAGGCCGAA IUGCAUCG 1114
    1044 CCGGCGGC U GCAGCGGC 739 GCCGCUGC CUGAUGAGGCCGUUAGGCCGAA ICCGCCGG 1115
    1047 GCGGCUGC A GCGGCACC 740 GGUGCCGC CUGAUGAGGCCGUUAGGCCGAA ICAGCCGC 1116
    1053 GCAGCGGC A CCCGCGCU 741 AGCGCGGG CUGAUGAGGCCGUUAGGCCGAA ICCGCUGC 1117
    1055 AGCGGCAC C CGCGCUCC 742 GGAGCGCG CUGAUGAGGCCGUUAGGCCGAA IUGCCGCU 1118
    1056 GCGGCACC C GCGCUCCU 743 AGGAGCGC CUGAUGAGGCCGUUAGGCCGAA IGUGCCGC 1119
    1061 ACCCGCGC U CCUGCACC 744 GGUGCAGG CUGAUGAGGCCGUUAGGCCGAA ICGCGGGU 1120
    1063 CCGCGCUC C UCCACCAG 745 CUGGUCCA CUGAUGAGGCCGUUAGGCCGAA IAGCGCGG 1121
    1064 CGCGCUCC U GCACCAGG 746 CCUGGUGC CUGAUGAGGCCGUUAGGCCGAA IGAGCGCG 1122
    1067 GCUCCUGC A CCAGGGAC 747 GUCCCUGG CUGAUGAGGCCGUUAGGCCGAA ICAGGAGC 1123
    1069 UCCUCCAC C AGGGACUG 748 CAGUCCCU CUGAUGAGGCCGUUAGGCCGAA IUCCAGGA 1124
    1070 CCUGCACC A GGGACUGU 749 ACAGUCCC CUGAUGAGGCCGUUAGGCCGAA IGUGCAGG 1125
    1076 CCAGGGAC U GUGCCGAG 750 CUCGGCAC CUGAUGAGGCCGUUAGGCCGAA IUCCCUGG 1126
    1081 GACUGUGC C GAGCCGCG 751 CGCGGCUC CUGAUGAGGCCGUUAGGCCGAA ICACAGUC 1127
    1086 UGCCGAGC C GCGCGCGG 752 CCGCGCGC CUGAUGAGGCCGUUAGGCCGAA ICUCGGCA 1128
    1111 GAAGCGUC C CCUCAGCC 753 GGCUGAGG CUGAUGAGGCCGUUAGGCCGAA IACGCUUC 1129
    1112 AAGCGUCC C CUCAGCCC 754 GGGCUGAG CUGAUGAGGCCGUUAGGCCGAA IGACGCUU 1130
    1113 AGCGUCCC C UCAGCCCC 755 GGGGCUGA CUGAUGAGGCCGUUAGGCCGAA IGGACGCU 1131
    1114 GCGUCCCC U CAGCCCCU 756 AGGGGCUG CUGAUGAGGCCGUUAGGCCGAA IGGGACGC 1132
    1116 GUCCCCUC A GCCCCUGG 757 CCAGGGGC CUGAUGAGGCCGUUAGGCCGAA IAGGGGAC 1133
    1119 CCCUCAGC C CCUCGAGG 758 CCUCCAGG CUGAUGACGCCGUUAGGCCGAA ICUGAGGG 1134
    1120 CCUCAGCC C CUGGAGGA 759 UCCUCCAG CUGAUGAGGCCGUUAGGCCGAA IGCUGAGG 1135
    1121 CUCAGCCC C UGGAGGAG 760 CUCCUCCA CUGAUGAGGCCGUUAGGCCGAA IGGCUGAG 1136
    1122 UCAGCCCC U GGAGGAGC 761 OCUCCUCO CUGAUGAGGCCGUUAGGCCGAA IGGGCUGA 1137
    1131 GGAGGAGC U GGAUCACC 762 GGUGAUCC CUGAUGAGGCCGUUAGGCCGAA ICUCCUCC 1138
    1137 GCUGGAUC A CCUCCUGC 763 GCAGGAGG CUGAUGAGGCCGUUAGGCCGAA IAUCCAGC 1139
    1139 UGGAUCAC C UCCUGCUG 764 CAGCAGGA CUGAUGAGGCCGUUAGGCCGAA IUGAUCCA 1140
    1140 GGAUCACC U CCUGCUGC 765 GCAGCAGG CUGAUGAGGCCGUUAGGCCGAA TGUGAUCC 1141
    1142 AUCACCUC C UGCUGCUG 766 CAGCAGCA CUGAUGAGGCCGUUAGGCCGAA IAGGUGAU 1142
    1143 UCACCUCC U GCUGCUGG 767 CCAGCAGC CUGAUGAGGCCGUUAGGCCGAA IGAGGUGA 1143
    1146 CCUCCUGC U GCUGGCGC 768 GCGCCAGC CUGAUGAGGCCGUUAGGCCGAA ICAGGAGG 1144
    1149 CCUGCUGC U GGCGCUGA 769 UCAGCGCC CUGAUGAGGCCGUUAGGCCGAA ICAGCAGG 1145
    1155 GCUGGCGC U GAUGACCG 770 CGGUCAUC CUGAUGAGGCCGUUAGGCCGAA ICGCCAGC 1146
    1162 CUGAUGAC C GUGCUCUU 771 AAGAGCAC CUGAUGAGGCCGUUAGGCCGAA IUCAUCAG 1147
    1167 GACCGUGC U CUUCACUA 772 UAGUGAAG CUGAUGAGGCCGUUAGGCCGAA ICACGGUC 1148
    1169 CCGUGCUC U UCACUAUG 773 CAUAGUGA CUGAUGAGGCCGUUAGGCCGAA IAGCACGG 1149
    1172 UGCUCUUC A CUAUGUGU 774 ACACAUAG CUGAUGAGGCCGUUAGGCCGAA IAAGAGCA 1150
    1174 CUCUUCAC U AUGUGUUC 775 GAACACAU CUGAUGAGGCCGUUAGGCCGAA IUGAAGAG 1151
    1183 AUGUGUUC U CUGCCCGU 776 ACGGGCAG CUGAUGAGGCCGUUAGGCCGAA IAACACAU 1152
    1185 GUGUUCUC U GCCCGUAA 777 UUACGGGC CUGAUGAGGCCGUUAGGCCGAA IAGAACAC 1153
    1188 UUCUCUGC C CGUAAUUU 778 AAAUUACG CUGAUGAGGCCGUUAGGCCGAA ICAGAGAA 1154
    1189 UCUCUGCC C GUAAUUUA 779 UAAAUUAC CUGAUGAGGCCGUUAGGCCGAA IGCAGAGA 1155
    1204 UAUCGCGC U UACUAUGG 780 CCAUAGUA CUGAUGAGGCCGUUAGGCCGAA ICGCGAUA 1156
    1208 GCGCUUAC U AUGGAGCA 781 UGCUCCAU CUGAUGAGGCCGUUAGGCCGAA IUAAGCGC 1157
    1216 UAUGGAGC A UUUAAGGA 782 UCCUUAAA CUGAUGAGGCCGUUAGGCCGAA ICUCCAUA 1158
    1229 AGGAUGUC A AGGAGAAA 783 UUUCUCCU CUGAUGAGGCCGUUAGGCCGAA IACAUCCU 1159
    1241 AGAAAAAC A GGACCUCU 784 AGAGGUCC CUGAUGAGGCCGUUAGGCCGAA IUUUUUCU 1160
    1246 AACAGGAC C UCUGAAGA 785 UCUUCAGA CUGAUGAGGCCGUUAGGCCGAA IUCCUGUU 1161
    1247 ACAGGACC U CUGAAGAA 786 UUCUUCAG CUGAUGAGGCCGUUAGGCCGAA IGUCCUGU 1162
    1249 AGGACCUC U GAAGAAGC 787 GCUUCUUC CUGAUGAGGCCGUUAGGCCGAA IAGGUCCU 1163
    1258 GAAGAAGC A GAAGACCU 788 AGGUCUUC CUGAUGAGGCCGUUAGGCCGAA ICUUCUUC 1164
    1265 CAGAAGAC C UCCGAGCC 789 GGCUCGGA CUGAUGAGGCCGUUAGGCCGAA IUCUUCUG 1165
    1266 AGAAGACC U CCGAGCCU 790 AGGCUCGG CUGAUGAGGCCGUUAGGCCGAA IGUCUUCU 1166
    1268 AAGACCUC C GAGCCUUG 791 CAAGGCUC CUGAUGAGGCCGUUAGGCCGAA IAGGUCUU 1167
    1273 CUCCGAGC C UUGCGAUU 792 AAUCGCAA CUGAUGAGGCCGUUAGGCCGAA ICUCGGAG 1168
    1274 UCCGAGCC U UUGCGAUU 793 AAAUCGCA CUGAUGAGGCCGUUAGGCCGAA IGCUCGGA 1169
    1284 GCGAUUUC U AUCUGUGA 794 UCACAGAU CUGAUGAGGCCGUUAGGCCGAA IAAAUCGC 1170
    1288 UUUCUAUC U GUGAUUUC 795 GAAAUCAC CUGAUGAGGCCGUUAGGCCGAA IAUAGAAA 1171
    1297 GUGAUUUC A AUUGUGGA 796 UCCACAAU CUGAUGAGGCCGUUAGGCCGAA IAAAUCAC 1172
    1307 UUGUGGAC C CUUGGAUU 797 AAUCCAAG CUGAUGAGGCCGUUAGGCCGAA IUCCACAA 1173
    1308 UGUGGACC C UUGGAUUU 798 AAAUCCAA CUGAUGAGGCCGUUAGGCCGAA IGUCCACA 1174
    1309 GUGGACCC U UGGAUUUU 799 AAAAUCCA CUGAUGAGGCCGUUAGGCCGAA IGGUCCAC 1175
    1322 UUUUUAUC A UUUUCAGA 800 UCUGAAAA CUGAUGAGGCCGUUAGGCCGAA IAUAAAAA 1176
    1328 UCAUUUUC A GAUCUCCA 801 UGGAGAUC CUGAUGAGGCCGUUAGGCCGAA IAAAAUGA 1177
    1333 UUCAGAUC U CCAGUAUU 802 AAUACUGG CUGAUGAGGCCGUUAGGCCGAA IAUCUGAA 1178
    1335 CAGAUCUC C AGUAUUUC 803 GAAAUACU CUGAUGAGGCCGUUAGGCCGAA IAGAUCUG 1179
    1336 AGAUCUCC A GUAUUUCG 804 CGAAAUAC CUGAUGAGGCCGUUAGGCCGAA IGAGAUCU 1180
    1356 AUUUUUUC A CAAGAUUU 805 AAAUCUUG CUGAUGAGGCCGUUAGGCCGAA IAAAAAAU 1181
    1358 UUUUUCAC A AGAUUUUC 806 GAAAAUCU CUGAUGAGGCCGUUAGGCCGAA IUGAAAAA 1182
    1367 AGAUUUUC A UUAGACCU 807 AGGUCUAA CUGAUGAGGCCGUUAGGCCGAA IAAAAUCU 1183
    1374 CAUUACAC C UCUUAGGU 808 ACCUAAGA CUGAUGAGGCCGUUAGCCCGAA IUCUAAUG 1184
    1375 AUUAGACC U CUUAGGUA 809 UACCUAAG CUGAUGAGGCCGUUAGGCCGAA IGUCUAAU 1185
    1377 UAGACCUC U UAGGUACA 810 UGUACCUA CUGAUGAGGCCGUUAGGCCGAA IAGGUCUA 1186
    1385 UUAGGUAC A GGAGCCGG 811 CCGGCUCC CUGAUGAGGCCGUUAGGCCGAA IUACCUAA 1187
    1391 ACAGGAGC C GGUGCAGC 812 GCUGCACC CUGAUGAGGCCGUUAGGCCGAA ICUCCUGU 1188
    1397 GCCGGUGC A GCAAUUCC 813 GGAAUUGC CUGAUGAGGCCGUUAGGCCGAA ICACCGGC 1189
    1400 GGUGCAGC A AUUCCACU 814 AGUGCAAU CUGAUGAGGCCGUUAGGCCGAA ICUGCACC 1190
    1405 AGCAAUUC C ACUAACAU 815 AUGUUAGU CUGAUGAGGCCGUUAGGCCGAA IAAUUGCU 1191
    1406 GCAAUUCC A CUAACAUG 816 CAUGUUAG CUGAUGAGGCCGUUAGCCCGAA IGAAUUGC 1192
    1408 AAUUCCAC U AACAUGGA 817 UCCAUGUU CUGAUGAGGCCGUUAGGCCGAA IUGGAAUU 1193
    1412 CCACUAAC A UGGAAUCC 818 GGAUUCCA CUGAUGAGGCCGUUAGGCCGAA IUUAGUGG 1194
    1420 AUCGAAUC C AGUCUGUG 819 CACAGACU CUGAUGAGGCCGUUAGGCCGAA IAUUCCAU 1195
    1421 UGGAAUCC A GUCUGUGA 820 UCACAGAC CUGAUGAGGCCGUUAGGCCGAA IGAUUCCA 1196
    1425 AUCCAGUC U GUGACAGU 821 ACUGUCAC CUGAUGAGGCCGUUAGGCCGAA IACUGGAU 1197
    1431 UCUGUGAC A GUGUUUUU 822 AAAAACAC CUGAUGAGGCCGUUAGGCCGAA IUCACAGA 1198
    1441 UGUUUUUC A CUCUGUGG 823 CCACAGAG CUGAUGAGGCCGUUAGGCCGAA IAAAAACA 1199
    1443 UUUUUCAC U CUGUGGUA 824 UACCACAG CUGAUGAGGCCGUUAGGCCGAA IUGAAAAA 1200
    1445 UUUCACUC U GUGGUAAG 825 CUUACCAC CUGAUGAGGCCGUUAGGCCGAA IAGUGAAA 1201
    1455 UGGUAAGC U GAGGAAUA 826 UAUUCCUC CUGAUGAGGCCGUUAGGCCGAA ICUUACCA 1202
    1468 AAUAUGUC A CAUUUUCA 827 UGAAAAUG CUGAUGAGGCCGUUAGGCCGAA IACAUAUU 1203
    1470 UAUGUCAC A UUUUCAGU 828 ACUGAAAA CUGAUGAGGCCGUUAGGCCGAA IUGACAUA 1204
    1476 ACAUUUUC A GUCAAAGA 829 UCUUUGAC CUGAUGAGGCCGUUAGGCCGAA IAAAAUGU 1205
    1480 UUUCAGUC A AAGAACCA 830 UGGUUCUU CUGAUGAGGCCGUUAGGCCGAA IACUGAAA 1206
  • [0232]
    TABLE V
    Human PTGDR Zinzyme and Substrate Sequence
    Seq Seq
    Pos Substrate ID Zinzyme ID
    9 GAAUUCUG G CUAUUUUC 1207 GAAAAUAG GCCGAAAGGCGAGUGAGGUCU CAGAAUUC 1438
    23 UUCCUCCU G CCGUUCCG 1208 CGGAACGG GCCGAAAGGCGAGUGAGGUCU AGGAGGAA 1439
    26 CUCCUGCC G UUCCGACU 1209 AGUCGGAA GCCGAAAGGCGAGUGAGGUCU GGCAGGAG 1440
    37 CCGACUCG G CACCAGAG 1210 CUCUGGUG GCCGAAAGGCGAGUGAGGUCU CGAGUCGG 1441
    45 GCACCAGA G UCUGUCUC 1211 GAGACAGA GCCGAAAGGCGAGUGAGGUCU UCUGGUGC 1442
    49 CAGAGUCU G UCUCUACU 1212 AGUAGAGA GCCGAAAGGCGAGUGAGGUCU AGACUCUG 1443
    64 CUGAGAAC G CAGCGCGU 1213 ACGCGCUG GCCGAAAGGCGAGUGAGGUCU GUUCUCAG 1444
    67 AGAACGCA G CGCGUCAG 1214 CUGACGCG GCCGAAAGGCGAGUGAGGUCU UGCGUUCU 1445
    69 AACGCAGC G CGUCAGGG 1215 CCCUGACG GCCGAAAGGCGAGUGAGGUCU GCUGCGUU 1446
    71 CGCAGCGC G UCAGGGCC 1216 GGCCCUGA GCCGAAAGGCGAGUGAGGUCU GCGCUGCG 1447
    77 GCGUCAGG G CCGAGCUC 1217 GAGCUCGG GCCGAAAGGCGAGUGAGGUCU CCUGACGC 1448
    82 AGGGCCGA G CUCUUCAC 1218 GUGAAGAG GCCGAAAGGCGAGUGAGGUCU UCGGCCCU 1449
    93 CUUCACUG G CCUGCUCC 1219 GGAGCAGG GCCGAAAGGCGAGUGAGGUCU CAGUGAAG 1450
    97 ACUGGCCU G CUCCUCUC 1220 GCGCGGAG GCCGAAAGGCGAGUGAGGUCU AGGCCAGU 1451
    102 CCUGCUCC G CGCUCUUC 1221 GAAGAGCG GCCGAAAGGCGAGUGAGGUCU GGAGCAGG 1452
    104 UGCUCCGC G CUCUUCAA 1222 UUGAAGAG GCCGAAAGGCGAGUGAGGUCU GCGGAGCA 1453
    114 UCUUCAAU G CCAGCGCC 1223 GGCGCUGG GCCUAAAGGCGAGUGAGGUCU AUUGAAUA 1454
    118 CAAUGCCA G CGCCAGGC 1224 GCCUGGCG GCCGAAAGGCGAUUGAGGUCU UGGCAUUU 1455
    120 AUGCCAUC G CCAGUCGC 1225 GCGCCUGG GCCGAAAGGCGAGUGAGGUCU GCUGGCAU 1456
    125 AUCUCCAG G CGCUCACC 1226 GGUGAGCG GCCGAAAGGCGAGUGAGGUCU CUGGCGCU 1457
    127 CGCCAGGC G CUCACCCU 1227 AUGGUGAG GCCGAAAGGCGAGUUAGGUCU GCCUGGCG 1458
    136 CUCACCCU G CAGAGCGU 1228 ACUCUCUG GCCGAAAGGCGAGUGAGGUCU AGUGUGAG 1459
    141 CCUUCAUA G CGUCCCGC 1229 UCUUGACU GCCGAAAGGCGAGUGAGGUCU UCUGCAGG 1460
    143 UUCAGAGC G UCCCUCCU 1230 AGUCUGGA GCCGAAAGGCGAGUGAGGUCU UCUCUUCA 1461
    148 AUCUUCCC G CCUCUCAA 1231 UUGAGAGG GCCGAAAUGCGAGUGAGGUCU GGGACGCU 1462
    163 AAAGAGGG G UGUGACCC 1232 GUGUCACA GCCGAAAGGCGAGUGAGGUCU CCCUCUUU 1463
    165 AUAUGUGU G UGACCCGC 1233 UCUGGUCA GCCUAAAGUCGAGUGAGGUCU ACCCCUCU 1464
    172 UGUGACCC G CGAGUUUA 1234 UAAACUCG GCCGAAAGGCGAGUGAGGUCU GUGUCACA 1465
    176 ACCCGCGA G UUUAGAUA 1235 UAUCUAAA GCCGAAAGGCGAGUGAGGUCU UCUCUUGU 1466
    189 UAUAUUAG G UUCCUGCC 1236 GGCAGGAA GCCGAAAUGCGAGUGAGGUCU CUCCUAUC 1467
    195 AGGUUCCU G CCUUUUUU 1237 CCCCACUU GCCGAAAGGCGAGUGAGGUCU AGGAACCU 1468
    198 UUCCUGCC G UGGGGAAC 1238 GUUCCCCA GCCGAAAGUCUAGUGAGGUCU GGCAGGAA 1469
    212 AACACCCC G CCUCCCUC 1239 GAGGUCUG GCCGAAAGGCGAGUGAGGUCU GGGGUGUU 1470
    215 ACCCCGCC G CCCUCGGA 1240 UCCUAGUG GCCGAAAUUCUAGUGAGGUCU UUCUUUUU 1471
    224 CCCUCGGA G CUUUUUCU 1241 AGAAAAAU UCCGAAAGGCGAGUGAUGUCU UCCGAGGG 1472
    233 CUUUUUCU G UGUCUCAG 1242 CUGCGCCA UCCGAAAGGCGAGUGAGGUCU AGAAAAAG 1473
    236 UUUCUGUG G CUCAUCUU 1243 AAUCUUCU UCCUAAAGGCUAUUGAUUUCU CACAGAAA 1474
    238 UCUGUGUC G CAGCUUCU 1244 AGAAGCUG UCCUAAAUUCUAUUUAGGUCU UCCACAGA 1475
    241 GUGUCUCA G CUUCUCCG 1245 CUUAUAAU GCCGAAAGGCUAUUUAUGUCU UGCGCCAC 1476
    249 GCUUCUCC G CCCUAUCC 1246 GGCUCUUU UCCUAAAUUCGAGUUAGUUCU UGAUAAUC 1477
    255 CCUCCCGA G CCGCUCUC 1247 UCGCGCUG UCCUAAAUUCUAUUUAUGUCU UCUUUCUU 1478
    258 CCCUAUCC G CUCGCGGA 1248 UCCUCGCU GCCGAAAGUCGAUUUAUUUCU GUCUCGGG 1479
    260 CGAGCCGC G CUCUUAUC 1249 GCUCCGCU UCCUAAAUUCUAGUGAUUUCU UCGUCUCU 1480
    262 AUCCUCUC G CUUAGCUG 1250 CAGCUCCU GCCUAAAGGCGAUUUAUGUCU UCUCGGCU 1481
    267 CGCGCGGA G CUUCCUUU 1251 CCCGGCAU UCCUAAAUUCUAGUGAUUUCU UCCUCUCU 1482
    270 UCUUAUCU G CCGGGGGC 1252 UCCCCCUG GCCUAAAUUCUAUUUAUUUCU AUCUCCUC 1483
    277 UGCCGGUU G CUCCUUAU 1253 CUAAUUAU UCCUAAAUUCGAGUGAUUUCU CCCCGUCA 1484
    285 GCUCCUUA G CACCCGGG 1254 CCCGGGUG GCCGAAAGGCGAGUGAGGUCU UAAGGAGC 1485
    293 GCACCCGG G CGCCGGGG 1255 CCCCGGCG GCCGAAAGGCGAGUGAGGUCU CCGGGUGC 1486
    295 ACCCGGGC G CCGGGGCC 1256 GGCCCCGG GCCGAAAGGCGAGUGAGGUCU GCCCGGGU 1487
    301 GCGCCGGG G CCCUCGCC 1257 GGCGAGGG GCCGAAAGGCGAGUCAGGUCU CCCGGCGC 1488
    307 GGGCCCUC G CCCUUCCG 1258 CGGAAGGG GCCGAAAGGCGAGUGAGGUCU GAGGGCCC 1489
    315 GCCCUUCC G CAGCCUUC 1259 GAAGGCUG GCCGAAAGGCGAGUGAGGUCU GGAAGGGC 1490
    318 CUUCCGCA G CCUUCACU 1260 AGUGAAGG CCCGAAAGGCGAGUGAGGUCU UGCCGAAG 1491
    330 UCACUCCA G CCCUCUGC 1261 GCAGAGGG GCCGAAAGGCGAGUGAGGUCU UGGACUGA 1492
    337 AGCCCUCU G CUCCCGCA 1262 UGCGGGAG GCCGAPAGGCGAGUGAGGUCU AGAGGGCU 1493
    343 CUGCUCCC G CACGCCAU 1263 AUGGCGUG GCCGAAAGGCGAGUGAGGUCU GGGAGCAC 1494
    347 UCCCGCAC G CCAUGAAG 1264 CUUCAUGG GCCGAAAGGCGAGUGAGGUCU GUGCGGGA 1495
    355 GCCAUGAA G UCGCCGUU 1265 AACGGCCA CCCGAAAGGCGAGUGAGGUCU UUCAUGGC 1496
    358 AUGAAGUC G CCGUUCUA 1266 UAGAACGG GCCGAAAGCCGAGUGAGGUCU GACUUCAU 1497
    361 AAGUCGCC G UUCUACCG 1267 CGGUACAA CCCGAAAGGCGAGUGAGCUCU GGCGACUU 1498
    369 GUUCUACC G CUCCOAGA 1268 UCUGGCAG GCCGAAACGCGAGUGAGGUCU CGUAGAAC 1499
    372 CUACCGCU G CCAGAACA 1269 UGUUCUCC GCCGAAAGGCGACUCACCUCU AGCCGUAC 1500
    389 CCACCUCU G UGGAAAAA 1270 UUUUUCCA GCCCAAAGGCGAGUGAGGUCU AGAGGUOG 1501
    399 GGAAAAAG G CAACUCGG 1271 CCCAGUUG GCCGAAAGGCGAGUGAGGUCU CUUUUUCC 1502
    407 GCAACUCG G CUCUGAUC 1272 CAUCACCC GCCCAAAGGCGAGUGACCUCU CGAGUUGC 1503
    410 ACUCGGCC G UGAUGGUC 1273 GCCCAUCA GCCGAAACCCGAGUGAGGUCU CGCCGAGU 1504
    417 GGUGAUGG G CGCCCUGC 1274 CCACCCCG GCCGAAAGGCCAGUCACCUCU CCAUCACC 1505
    422 UGGGCCGC G UGCUCUUC 1275 GAAGAGCA GCCCAAAGGCGAGUGAGCUCU CCCGCCCA 1506
    424 CGCGGGGU G CUCUUCAG 1276 CUGAAGAG CCCGAAAGGCGAGUGAGGUCU ACCCCCCC 1507
    432 GCUCUUCA G CACCGGCC 1277 GGCCGGUG CCCGAAAGGCGAGUGAGGUCU UCAAGAGC 1508
    438 CAGCACCG G CCUCCUCC 1278 CCAGCAGG GCCGAAAGGCGAGUGAGGUCU CCGUGCUG 1509
    447 CCUCCUCG G CAACCUGC 1279 GCAGGUUG GCCCAAAGGCGAGUGAGCUCU CCAGGAGG 1510
    454 GCCAACCU G CUCGCCCU 1280 AGGGCCAG CCCGAAAGGCGAGUCAGGUCU AGGUUCCC 1511
    458 ACCUCCUC G CCCUGGGG 1281 CCCCAGGC GCCCAAAGGCGAGUGACGUCU CAGCAGGU 1512
    466 GCCCUGGG G CUGCUCGC 1282 GCCAGCAG GCCGAAAGGCCAGUGAGGUCU CCCAGCCC 1513
    469 CUGGGGCU G CUGGCCCG 1283 CGCGCCAG GCCGAAAGGCGAGUCAGGUCU AGCCCCAC 1514
    473 GGCUGCUG G CGCCCUCG 1284 CGAGCGCG GCCGAAAGGCGAGUGAGGUCU CACCAGCC 1515
    475 CUGCUGGC G CCCUCCGG 1285 CCCCACCC GCCGAAACCCCACUCAGGUCU GCCACCAC 1516
    477 CCUCGCGC G CUCGGCCC 1286 CCCCCCAC CCCCAAACGCGAGUCACCUCU CCGCCACC 1517
    484 CCCUCCCC G CUCCGGUG 1287 CACCCCAG GCCCAAACCCCACUCAGCUCU CCCCACCC 1518
    490 CCCCUGCG G UGGUCCUC 1288 CACCACCA CCCCAAACCCCACUCACCUCU CCCACCCC 1519
    493 CUGCCCUC G UCCUCCCG 1289 CCCGACCA GCCCAAACCCCAGUCACCUCU CACCCCAG 1520
    495 CCCCUCGU G CUCCCCCC 1290 CCCCCCAC CCCCAAAGCCGACUCACCUCU ACCACCCC 1521
    499 UCCUCCUC G CCCCCUCC 1291 CCACGCCG CCCGAAACCCCACUGAGCUCU CACCACCA 1522
    502 UCCUCGCG G CCUCCACU 1292 AGUCCACC CCCGAAAGGCGACUCACCUCU CGCCACCA 1523
    504 CUCCCCCC G UCCACUCC 1293 GCACUCCA CCCCAAACGCGAGUCACGUCU CCCCCGAC 1524
    511 CCUCCACU G CCCCCCCU 1294 ACCCCGCG GCCGAAACCCCACUCAGGUCU AGUCCACC 1525
    513 UCCACUGC G CCCCCUCC 1295 CCACCCCC CCCCAAACCCCACUCACCUCU CCAGUCCA 1526
    517 CUCCGCCC G CUGCCCUC 1296 CACGCCAC GCCCAAACCCGAGUCAGGUCU GGGCCCAC 1527
    520 CGCCCGCU G CCCUCCGU 1297 ACCGACCG GCCGAAAGGCCAGUCACCUCU AGCCCCCC 1528
    527 UGCCCUCG G UCUUCUAC 1298 GUACAACA GCCGAAACGCGAGUGAGGUCU CCACGCCA 1529
    538 UUCUACAU G CUCCUCUC 1299 CACACCAG CCCGAAAGGCGAGUCAGGUCU AUCUACAA 1530
    542 ACAUCCUC G UGUGUGCC 1300 GCCACACA GCCGAAACCCGACUGAGGUCU CACCAUGU 1531
    544 AUCCUCCU G UGUCCCCU 1301 AGCCCACA GCCGAAACCCGAGUCACGUCU ACCACCAU 1532
    546 GCUCCUGU G UGCCCUGA 1302 UCACGCCA CCCCAAACCCCAGUGACCUCU ACACCACC 1533
    549 CCUGUCUG G CCUCACCC 1303 CCCUCACG CCCCAAACCCGACUCAGGUCU CACACACC 1534
    557 GCCUGACC G UCACCGAC 1304 CUCGGUGA CCCCAAACCCGAGUGACGUCU CCUCACCC 1535
    568 ACCCACUU G CUGGGCAA 1305 UUGCCCAG GCCGAAAGGCGAGUGAGGUCU AAGUCGGU 1536
    573 CUUGCUGG G CAAGUGCC 1306 GGCACUUG GCCGAAAGGCGAGUGAGGUCU CCAGCAAG 1537
    577 CUGGGCAA G UGCCUCCU 1307 AGGAGGCA GCCGAAAGGCGAGUGAGGUCU UUGCCCAG 1538
    579 GGGCAAGU G CCUCCUAA 1308 UUAGGAGG GCCGAAAGGCGAGUGAGGUCU ACUUGCCC 1539
    588 CCUCCUAA G CCCGGUGG 1309 CCACCGGG GCCGAAAGGCGAGUGAGGUCU UUAGGACG 1540
    593 UAAGCCCG G UGGUGCUG 1310 CAGCACCA GCCGAAAGGCGAGUGAGGUCU CGGGCUUA 1541
    596 GCCCGGUG G UGCUGGCU 1311 AGCCAGCA GCCGAAAGGCGAGUGAGGUCU CACCGGGC 1542
    598 CCGGUGGU G CUGGCUGC 1312 GCAGCCAG GCCGAAAGGCGAGUGAGGUCU ACCACCGG 1543
    602 UGGUGCUG G CUOCCUAC 1313 GUAGUCAG GCCGAAAGGCGAGUGAGGUCU CAGCACCA 1544
    605 UGCUGGCU G CCUACGCU 1314 AGCGUAGG GCCGAAAGGCGAGUGAGGUCU AGCCAGCA 1545
    611 CUGCCUAC G CUCAGAAC 1315 GUUCUGAG GCCGAAAGGCGAGUGAGGUCU GUAGGCAG 1546
    624 GAACCGGA G UCUGCGGG 1316 CCCGCAGA GCCGAAAGGCGAGUGAGGUCU UCCGCUUC 1547
    628 CGGAGUCU G CGGGUGCU 1317 AGCACCCG GCCGAAAGGCGAGUGAGCUCU AGACUCCG 1548
    632 GUCUGCGG G UGCUUGCG 1318 CGCAAGCA GCCGAAAGGCGAGUGAGGUCU CCGCAGAC 1549
    634 CUGCGGGU G CUUGCGCC 1319 GGCGCAAG GCCGAAAGGCGAGUGAGGUCU ACCCGCAG 1550
    638 GGGUGCUU G CGCCCGCA 1320 UGCGGGCG GCCGAAAGGCGAGUGAGGUCU AAGCACCC 1551
    640 GUGCUUGC G CCCGCAUU 1321 AAUGCGGG GCCGAAAGGCGAGUGAGGUCU GCAAGCAC 1552
    644 UUGCGCCC G CAUUGGAC 1322 GUCCAAUG GCCGAAAGGCGAGUGAGGUCU GGGCGCAA 1553
    658 GACAACUC G UUGUGCCA 1323 UGGCACAA GCCGAAAGGCGAGUGAGGUCU GAGUGGUC 1554
    661 AACUCGUU G UGCCAAGC 1324 GCUUGGCA GCCGAAAGGCGAGUGAGGUCU AACGAGUU 1555
    663 CUCGUUGU G CCAAGCCU 1325 AGGCUUGG GCCGAAAGGCGAGUGAGGUCU ACAACGAG 1556
    668 UGUGCCAA G CCUUCGCC 1326 GGCGAAGG GCCGAAAGGCGAGUGAGGUCU UUGGCACA 1557
    674 AAGCCUUC G CCUUCUUC 1327 GAAGAAGG GCCGAAAGGCGAGUGAGGUCU GAAGGCUU 1558
    685 UUCUUCAU G UCCUUCUU 1328 AAGAAGGA GCCGAAAGGCGAGUGAGGUCU AUGAAGAA 1559
    697 UUCUUUGG G CUCUCCUC 1329 GAGGAGAG GCCGAAAGGCGAGUGAGGUCU CCAAAGAA 1560
    712 UCGACACU G CAACUCCU 1330 AGGAGUUG GCCGAAAGGCGAGUGAGGUCU AGUGUCGA 1561
    722 AACUCCUG G CCAUGGCA 1331 UGCCAUGG GCCGAAAGGCGAGUGAGGUCU CAGGAGUU 1562
    728 UGGCCAUG G CACUGGAG 1332 CUCCAGUG GCCGAAAGGCGAGUGAGGUCU CAUGOCCA 1563
    736 GCACUGGA G UGCUGGCU 1333 AGCCAGCA GCCGAAAGGCGAGUGAGGUCU UCCAGUGC 1564
    738 ACUGGAGU G CUGGCUCU 1334 AGAGCCAG GCCGAAAGGCGAGUGAGGUCU ACUCCAGU 1565
    742 GAGUCCUG G CUCUCCCU 1335 AGGGAGAG GCCGAAAGGCGAGUGAGGUCU CACCACUC 1566
    754 UCCCUAGG G CACCCUUU 1336 AAAGGGUG GCCGAAAGGCGAGUGAGGUCU CCUAGGGA 1567
    775 UACCGACG G CACAUCAC 1337 GUGAUGUG GCCGAAAGGCGAGUGAGGUCU CGUCGGUA 1568
    787 AUCACCCU G CGCCUGGG 1338 CCCAGGCG GCCGAAAGGCGAGUGAGGUCU AGGGUGAU 1569
    789 CACCCUGC G CCUGGGCG 1339 CGCCCAGG GCCGAAAGGCGAGUGAGGUCU GCAGGGUG 1570
    795 GCGCCUGG G CGCACUGG 1340 CCAGUGCG GCCGAAAGGCGAGUGAGGUCU CCAGGCGC 1571
    797 GCCUGGGC G CACUGGUG 1341 CACCAGUG GCCGAAAGGCGAGUGAGGUCU GCCCAGGC 1572
    803 GCGCACUG G UGGCCCCG 1342 CGGGGCCA GCCGAAAGGCGAGUGAGGUCU CAGUGCGC 1573
    806 CACUGGUG G CCCCGGUG 1343 CACCGGGG GCCGAAAGGCGAGUGAGGUCU CACCAGUG 1574
    812 UGGCCCCG G UGGUGAGC 1344 GCUCACCA GCCGAAAGGCGAGUGAGGUCU CGGGGCCA 1575
    815 CCCCGGUG G UGAGCGCC 1345 GGCGCUCA GCCGAAAGGCGAGUGAGGUCU CACCGGGG 1576
    819 GGUGGUGA G CGCCUUCU 1346 AGAAGGCG GCCGAAAGGCGAGUGAGGUCU UCACCACC 1577
    821 UGGUGAGC G CCUUCUCC 1347 GGAGAAGG GCCGAAAGGCGAGUGAGGUCU GCUCACCA 1578
    833 UCUCCCUG G CUUUCUGC 1348 GCAGAAAG GCCGAAAGGCGAGUGAGGUCU CAGGGAGA 1579
    840 GGCUUUCU G CGCGCUAC 1349 GUAGCGCG GCCGAAAGGCGAGUGAGGUCU AGAAAGCC 1580
    842 CUUUCUGC G CGCUACCU 1350 AGGUAGCG GCCGAAAGGCGAGUGAGGUCU GCAGAAAG 1581
    844 UUCUGCGC G CUACCUUU 1351 AAAGGUAG GCCGAAAGGCGAGUGAGGUCU GCGCAGAA 1582
    858 UUUCAUGG G CUUCGGGA 1352 UCCCGAAG GCCGAAAGGCGAGUGAGGUCU CCAUGAAA 1583
    868 UUCGGGAA G UUCGUGCA 1353 UGCACGAA GCCGAAAGGCGAGUGAGGUCU UUCCCGAA 1584
    872 GGAAGUUC G UCCAGUAC 1354 GUACUGCA GCCGAAAGGCGAGUGAGGUCU GAACUUCC 1585
    874 AAGUUCGU G CAGUACUG 1355 CAGUACUG GCCGAAAGGCGAGUGAGGUCU ACGAACUU 1586
    877 UUCGUGCA G UACUGCCC 1356 GGGCAGUA CCCGAAAGGCGAGUGAGGUCU UGCACGAA 1587
    882 GCAGUACU G CCCCGGCA 1357 UGCCGGGG GCCGAAAGGCCAGUGAGGUCU AGUACUGC 1588
    888 CUGCCCCG G CACCUGGU 1358 ACCAGGUG GCCGAAAGGCGAGUGAGGUCU CGGGGCAG 1589
    895 GOCACCUG G UGCUUUAU 1359 AUAAAGCA GCCGAAAGGCGAGUGAGGUCU CAGGUGCC 1590
    897 CACCUGGU G CUUUAUCC 1360 GGAUAAAG GCCGAAAGGCGAGUGAGGUCU ACCAGGUG 1591
    911 UCCAGAUG G UCCACGAG 1361 CUCCUGGA GCCGAAAGGCGAGUGAGGUCU CAUCUGGA 1592
    924 CGAGGAGG G CUCGCUGU 1362 ACAGCGAG GCCGAAAGGCGAGUGAGGUCU CCUCCUCG 1593
    928 GAGGGCUC G CUGUCGGU 1363 ACCGACAG GCCGAAAGGCGAGUGAGGUCU GAGCCCUC 1594
    931 GGCUCGCU G UCGGUGCU 1364 AGCACCGA GCCGAAAGGCGAGUGAGGUCU AGCGAGCC 1595
    935 CGCUGUCG G UGCUGGGG 1365 CCCCAGCA GCCGAAAGGCGAGUGAGGUCU CGACAGCG 1596
    937 CUGUCGGU G CUGCGGUA 1366 UACCCCAG GCCGAAACCCGAGUGAGGUCU ACCGACAG 1597
    943 GUCCUGOG G UACUCUGU 1367 ACAGAGUA GCCGAAAGGCGAGUGAGGUCU CCCAGCAC 1598
    950 CGUACUCU G UCCUCUAC 1368 GUACAGCA GCCGAAAGCCGAGUGAGGUCU AGACUACC 1599
    952 UACUCUGU G CUCUACUC 1369 GAGUAGAC GCCGAAAGCCGAGUCACGUCU ACAGAGUA 1600
    963 CUACUCCA G CCUCAUGG 1370 CCAUGACG CCCGAAACGCGACUGAGGUCU UGGAGUAG 1601
    971 GCCUCAUG G CGCUGCUG 1371 CAGCAGCG GCCCAAAGGCGAGUGAGGUCU CAUGAGGC 1602
    973 CUCAUGOC G CUGCUCCU 1372 ACCAGCAG GCCGAAAGGCGAGUGAGCUCU OCCAUGAG 1603
    976 AUGGCGCU G CUCGUCCU 1373 AGGACCAG GCCGAAAGGCGAGUCAGGUCU AGCGCCAU 1604
    980 CGCUGCUG G UCCUCGCC 1374 GGCGACGA GCCGAAAGGCGACUGAGGUCU CAGCAGCG 1605
    986 UGGUCCUC G CCACCGUC 1375 CACGGUGG GCCGAAAGGCGAGUGACGUCU GAGGACCA 1606
    992 UCCCCACC G UGCUGUGC 1376 GCACAGCA GCCGAAAGGCGAGUGAGGUCU GGUGGCGA 1607
    994 GCCACCGU G CUGUGCAA 1377 UUGCACAG GCCGAAAGGCGAGUGACGUCU ACGGUGGC 1608
    997 ACCCUGCU G UGCAACCU 1378 AGCUUGCA GCCGAAAGGCGACUGAGGUCU AGCACCCU 1609
    999 CGUGCUGU G CAACCUCG 1379 CGAGGUUG GCCCAAAGGCGAGUGAGGUCU ACAGCACG 1610
    1008 CAACCUCC G CCCCAUGC 1380 GCAUGGCG GCCGAAAGGCCAGUCAGGUCU CGACGUUG 1611
    1010 ACCUCGGC G CCAUGCGC 1381 GCGCAUGG GCCGAAAGGCGAGUGAGGUCU CCCGAGGU 1612
    1015 GGCCCCAU G CCCAACCU 1382 AGGUUGCC GCCGAAAGGCCAGUGAGGUCU AUGCCCCC 1613
    1017 CGCCAUCC G CAACCUCU 1383 AGAGGUUG CCCGAPAGGCGACUGAGGUCU GCAUGGCG 1614
    1028 ACCUCUAU G CGAUGCAC 1384 GUCCAUCG GCCCAAAGGCGAGUGAGCUCU AUAGAGGU 1615
    1033 UAUGCGAU G CACCGCCC 1385 CGCCGGUG GCCCAAACGCGAGUGAGGUCU AUCGCAUA 1616
    1039 AUGCACCC G CUCCUCCA 1386 UCCAGCCC CCCCAAAGGCGAGUCACCUCU CGGUCCAU 1617
    1042 CACCCGCC G CUCCACCC 1387 CCCUGCAG CCCGAAACCCCACUGAGGUCU CCCCCCUG 1618
    1045 CGGCCCCU G CACCUCCA 1388 UCCCGCUG CCCCAAAGGCGAGUCACCUCU AGCCGCCC 1619
    1048 CCCCUGCA G CCCCACCC 1389 CCCUCCCC GCCGAAAGCCCACUGACGUCU UGCACCCG 1620
    1051 CUCCACCC G CACCCCCG 1390 OCCUGGUC CCCCAAAGGCGAGUCAGCUCU CGCUGCAG 1621
    1057 CCCCACCC G CUCUCCUC 1391 CACCACCG GCCGAAAGCCCAGUGAGCUCU CCCUCCCG 1622
    1059 CCACCCCC G CUCCUCCA 1392 UCCACCAC CCCCAAAGGCGACUCACCUCU GCGGCUCC 1623
    1065 GCCCUCCU G CACCACGG 1393 CCCUGGUG GCCCAAACCCCAGUCAGCUCU ACCACCUC 1624
    1077 CACGGACU G UCCCCACC 1394 GCUCCCCA GCCGAAACCCGACUCAGGUCU AGUCCCUC 1625
    1079 CCCACUCU G CCCAGCCC 1395 CUCCUCUC CCCCAAAGCCGAGUGACCUCU ACAGUCCC 1626
    1084 UCUGCCCA G CCCCCCGC 1396 GCGCGCGG CCCCAAACCCCAGUGACGUCU UCCCCACA 1627
    1087 CCCCAGCC G CUCUCUCA 1397 UCCCCCCC GCCGAAAGGCGACUCACGUCU GUCUCCUC 1628
    1089 CGAGCCGC G CGCGGACG 1398 CGUCCGCG CCCCAAACCCCAGUGAGGUCU CCCGCUCG 1629
    1091 AUCCUCUC G CGGACCCC 1399 CCCCUCCG GCCGAAAGGCGACUCACGUCU CCGCCCCU 1630
    1106 CCAGGCAA G CCUCCCCU 1400 AGGGGACG CCCGAAACCCGAGUGAGCUCU UUCCCUCC 1631
    1108 ACCCAAGC G UCCCCUCA 1401 UCAGCCCA GCCGAAAGGCCAGUCACCUCU CCUUCCCU 1632
    1117 UCCCCUCA G CCCCUCGA 1402 UCCAGGUC CCCGAAACCCGAGUCAGCUCU UCAGUGGA 1633
    1129 CUGGAGGA G CUCCAUCA 1403 UCAUCCAG GCCGAAACCCCACUCAGGUCU UCCUCCAC 1634
    1144 CACCUCCU G CUCCUGUC 1404 UCCACCAC CCCCAAAGGCGACUGACCUCU AGGAGGUC 1635
    1147 CUCCUCCU G CUGCCGCU 1405 ACCGCCAG CCCGAAACCCGAGUGAGCUCU ACCACCAG 1636
    1151 UCCUCCUC G CUCUCAUG 1406 CAUCACCC GCCGAAAGCCGACUGAGGUCU CACCACCA 1637
    1153 CUGCUGGC G CUGAUGAC 1407 GUCAUCAG GCCGAAAGGCGAGUGAGGUCU UCCACCAG 1638
    1163 UGAUGACC G UGCUCUUC 1408 GAAGAGCA GCCGAAAGGCGAGUCAGGUCU GGUCAUCA 1639
    1165 AUGACCGU G CUCUUCAC 1409 GUGAAGAG GCCGAAAGGCGAGUGAGGUCU ACGGUCAU 1640
    1177 UUCACUAU G UGUUCUCU 1410 AGAGAACA GCCGAAACCCGAGUGAGGUCU AUAGUGAA 1641
    1179 CACUAUGU G UUCUCUGC 1411 GCAGAGAA GCCGAAAGGCGACUGACCUCU ACAUACUC 1642
    1186 UGUUCUCU G CCCGUAAU 1412 AUUACGGG GCCGAAAGGCGAGUGAGGUCU AGAGAACA 1643
    1190 CUCUGCCC G UAAUUUAU 1413 AUAAAUUA GCCCAAAGGCGAGUCAGGUCU GGGCAGAG 1644
    1200 AAUUUAUC G CGCUUACU 1414 AGUAAGCG GCCGAAAGGCGACUCAGGUCU GAUAAAUU 1645
    1202 UUUAUCGC G CUUACUAU 1415 AUAGUAAG GCCCAAACCCCAGUGAGGUCU GCCAUAAA 1646
    1214 ACUAUGCA G CAUUUAAC 1416 CUUAAAUG CCCGAAAGGCGAGUGACCUCU UCCAUACU 1647
    1226 UUAAGGAU G UCAACCAG 1417 CUCCUCCA GCCGAAACGCGAGUGAGCUCU AUCCUUAA 1648
    1256 CUGAAGAA G CAGAAGAC 1418 GUCUUCUG CCCCAAACCCGACUGAGCUCU UUCUUCAG 1649
    1271 ACCUCCGA G CCUUGCCA 1419 UCGCAAGG GCCGAAACGCGAGUGAGCUCU UCGGAGGU 1650
    1276 CCAGCCUU G CCAUUUCU 1420 AGAAAUCG GCCGAAACCCGACUGAGGUCU AACGCUCG 1651
    1289 UUCUAUCU G UGAUCUCA 1421 UCAAAUCA CCCGAAAGCCCAGUGAGGUCU AGAUAGAA 1652
    1301 UUUCAAUU G UGGACCCU 1422 ACCGUCCA GCCCAAACGCCAGUGAGGUCU AAUUCAAA 1653
    1337 GAUCUCCA G UAUUUCCG 1423 CCGAAAUA CCCGAAAGGCGAGUGAGGUCU UGGAGAUC 1654
    1381 CCUCUUAG G UACAGGAG 1424 CUCCUGUA GCCGAAAGGCGAGUGAGCUCU CUAAGAGG 1655
    1389 CUACAGGA G CCGGUGCA 1425 UGCACCGC GCCGAAACCCGAGUGAGGUCU UCCUGUAC 1656
    1393 AGGAGCCG G UCCAGCAA 1426 UUCCUGCA CCCCAAAGGCGAGUCACCUCU CGGCUCCU 1657
    1395 CACCCCCU G CAGCAAUU 1427 AAUUCCUC GCCGAAACCCCACUGAGGUCU ACCCCCUC 1658
    1398 CCGCUGCA G CAAUUCCA 1428 UGGAAUUG CCCCAAAGGCCAGUCACCUCU UGCACCCG 1659
    1422 CCAAUCCA G UCUGUCAC 1429 CUCACACA CCCCAAACCCCACUGAGGUCU UCCAUUCC 1660
    1426 UCCAGUCU G UCACACUC 1430 CACUGUCA CCCCAAAGGCCAGUCACCUCU AGACUGGA 1661
    1432 CUCUCACA G UCUUUUUC 1431 CAAAAACA CCCGAAACCCCACUGAGCUCU UCUCACAG 1662
    1434 CUGACACU G UUUUUCAC 1432 CUGAAAAA CCCGAAACGCCACUCACCUCU ACUGUCAC 1663
    1446 UUCACUCU G UCCUAAGC 1433 CCUUACCA CCCCAAACGCGAGUGACCUCU AGACUGAA 1664
    1449 ACUCUGUG G UAACCUCA 1434 UCACCUUA GCCGAAACCCCACUGAGCUCU CACACACU 1665
    1453 UCUCCUAA G CUGAGGAA 1435 UUCCUCAC CCCGAAAGGCGACUCACCUCU UUACCACA 1666
    1465 ACCAAUAU G UCACAUUU 1436 AAAUCUGA CCCCAAACCCGAGUGACCUCU AUAUUCCU 1667
    1477 CAUUUUCA G UCAAACAA 1437 UUCUUUCA CCCCAAACCCCACUCACCUCU UGAAAAUC 1668
  • [0233]
    TABLE VI
    Human PTGDR DNAzyme
    and Substrate Sequence
    Seq Seq
    Pos Substrate ID DNAzyme ID
    9 GAAUUCUG G CUAUUUUC 1207 GAAAATAG GGCTAGCTACAACGA CAGAATTC 1715
    12 UUCUGGCU A UUUUCCUC 1 GAGGAAAA GGCTAGCTACAACGA AGCCAGAA 1716
    23 UUCCUCCU G CCGUUCCG 1208 CGGAACGG GGCTAGCTACAACGA AGGAGGAA 1717
    26 CUCCUGCC G UUCCGACU 1209 AGTCGGAA GGCTAGCTACAACGA GGCAGGAG 1718
    32 CCGUUCCG A CUCGGCAC 1669 GTGCCGAG GGCTAGCTACAACGA CGGAACGG 1719
    37 CCGACUCG G CACCAGAG 1210 CTCTGGTG GGCTAGCTACAACGA CGAGTCGG 1720
    39 GACUCGGC A CCAGAGUC 463 GACTCTGG GGCTAGCTACAACGA GCCGAGTC 1721
    45 GCACCAGA G UCUGUCUC 1211 GAGACAGA GGCTAGCTACAACGA TCTGGTGC 1722
    49 CAGAGUCU G UCUCUACU 1212 AGTAGAGA GGCTACCTACAACGA AGACTCTG 1723
    55 CUGUCUCU A CUGAGAAC 13 GTTCTCAG GGCTAGCTACAACGA AGAGACAG 1724
    62 UACUGAGA A CGCAGCGC 1670 GCGCTGCG GGCTAGCTACAACGA TCTCAGTA 1725
    64 CUGAGAAC G CAGCGCGU 1213 ACGCGCTG GGCTAGCTACAACGA GTTCTCAG 1726
    67 AGAACGCA G CGCGUCAG 1214 CTGACGCG GGCTAGCTACAACGA TGCGTTCT 1727
    69 AACGCAGC G CGUCAGGG 1215 CCCTGACG GGCTAGCTACAACGA GCTGCGTT 1728
    71 CGCAGCGC G UCAGGGCC 1216 GGCCCTGA GGCTAGCTACAACGA GCGCTGCG 1729
    77 GCGUCAGG G CCGAGCUC 1217 CAGCTCGG GGCTAGCTACAACCA CCTGACGC 1730
    82 AGCCCCGA G CUCUUCAC 1218 GTCAAGAC GCCTAGCTACAACGA TCGGCCCT 1731
    89 AGCUCUUC A CUGGCCUG 475 CAGGCCAG GGCTAGCTACAACGA GAAGAGCT 1732
    93 CUUCACUG G CCUGCUCC 1219 GCACCAGG GGCTAGCTACAACGA CAGTGAAG 1733
    97 ACUCCCCU G CUCCGCGC 1220 GCGCGGAG GGCTAGCTACAACGA AGOCCACT 1734
    102 CCUGCUCC G CCCUCUUC 1221 CAAGAGCG GGCTAGCTACAACGA GGACCAGG 1735
    104 UGCUCCGC G CUCUUCAA 1222 TTCAAGAG GGCTAGCTACAACGA GCGGAGCA 1736
    112 GCUCUUCA A UGCCAGCG 1671 CGCTGGCA GGCTAGCTACAACGA TGAAGAGC 1737
    114 UCUUCAAU G CCAGCGCC 1223 GGCGCTGG GGCTAGCTACAACGA ATTGAAGA 1738
    118 CAAUGCCA G CGCCAGGC 1224 GCCTGGCG GGCTAGCTACAACGA TGGCATTG 1739
    120 AUGCCAGC G CCAGCCGC 1225 GCGCCTGG GGCTAGCTACAACGA GCTGCCAT 1740
    125 AGCGCCAG G CGCUCACC 1226 GGTGAGCG CGCTAGCTACAACGA CTGGCGCT 1741
    127 CGCCAGGC G CUCACCCU 1227 AGGGTGAG GGCTAGCTACAACGA GCCTGGCG 1742
    131 AGGCGCUC A CCCUGCAG 489 CTGCAGGG GGCTAGCTACAACGA GAGCGCCT 1743
    136 CUCACCCU G CAGAGCGU 1228 ACGCTCTG GGCTAGCTACAACGA AGGGTGAG 1744
    141 CCUGCAGA G CGUCCCGC 1229 GCGGGACG GGCTAGCTACAACGA TCTGCAGG 1745
    143 UGCAGAGC G UCCCGCCU 1230 AGGCGGGA GGCTAGCTACAACGA GCTCTGCA 1746
    148 AGCGUCCC G CCUCUCAA 1231 TTGAGAGG GGCTAGCTACAACGA GGGACGCT 1747
    163 AAAGAGGG G UGUGACCC 1232 GGGTCACA GGCTAGCTACAACGA CCCTCTTT 1748
    165 AGAGGGGU G UGACCCGC 1233 GCGGGTCA GGCTAGCTACAACGA ACCCCTCT 1749
    168 GCGGUGUG A CCCGCGAG 1672 CTCGCGGG GGCTAGCTACAACGA CACACCCC 1750
    172 UGUGACCC G CGAGUUUA 1234 TAAACTCG GGCTAGCTACAACGA GGGTCACA 1751
    176 ACCCGCGA G UUUAGAUA 1235 TATCTAAA GGCTAGCTACAACGA TCGCGGGT 1752
    182 GAGUUUAG A UAGGAGGU 1673 ACCTCCTA GGCTAGCTACAACGA CTAAACTC 1753
    189 GAUAGGAG G UUCCUGCC 1236 GGCAGGAA GGCTAGCTACAACGA CTCCTATC 1754
    195 AGGUUCCU G CCGUGGGG 1237 CCCCACCG GGCTAGCTACAACGA AGGAACCT 1755
    198 UUCCUGCC G UGGGGAAC 1238 GTTCCCCA GGCTAGCTACAACGA GCCAGGAA 1756
    205 CGUGGGGA A CACCCCGC 1674 CCCGGGTG GGCTAGCTACAACGA TCCCCACG 1757
    207 UGGCGAAC A CCCCGCCG 505 CGGCGGGG GGCTAGCTACAACGA GTTCCCCA 1758
    212 AACACCCC G CCCCCCUC 1239 GAGCCCCG GGCTAGCTACAACGA GCGGTGTT 1759
    215 ACCCCGCC G CCCUCGGA 1240 TCCGAGCG GGCTAGCTACAACGA GGCGGCGT 1760
    224 CCCUCGGA G CUUUUUCU 1241 AGAAAAAG GGCTAGCTACAACGA TCCGAGGG 1761
    233 CUUUUUCU G UGGCGCAG 1242 CTGCGCCA GGCTAGCTACAACGA AGAAAAAG 1762
    236 UUUCUCUG G CGCAGCUU 1243 AAGCTGCC GGCTAGCTACAACGA CACAGAAA 1763
    238 UCUGUGGC G CAGCUUCU 1244 AGAAGCTG GGCTAGCTACAACGA GCCACAGA 1764
    241 GUGGCGCA G CUUCUCCG 1245 CGGAGAAG GCCTAGCTACAACGA TOCCCCAC 1765
    249 GCUUCUCC G CCCGAGCC 1246 GGCTCGGG CGCTAGCTACAACGA GGAGAAGC 1766
    255 CCGCCCGA G CCCCGCGC 1247 GCGCGCGG GGCTACCTACAACGA TCGGGCGG 1767
    258 CCCGAGCC G CGCGCGGA 1248 TCCGCGCG GGCTAGCTACAACCA GGCTCCCG 1768
    260 CGAGCCGC G CGCGGAGC 1249 GCTCCCCG GGCTAGCTACAACGA GCGGCTCG 1769
    262 ACCCGCCC G CGGAGCUG 1250 CAGCTCCG GGCTAGCTACAACGA CCGCGCCT 1770
    267 CGCGCGGA G CUCCCGCG 1251 CCCGGCAG GGCTAGCTACAACCA TCCGCGCG 1771
    270 GCGCACCU G CCGGCGGC 1252 GCCCCCCG GGCTAGCTACAACGA AGCTCCGC 1772
    277 UGCCGGGG G CUCCUUAG 1253 CTAAGGAC GGCTAGCTACAACCA CCCCGGCA 1773
    285 CCUCCUUA G CACCCCGG 1254 CCCCGGTC GCCTAGCTACAACGA TAAGGAGC 1774
    287 UCCUUAGC A CCCCGGCG 527 CCCCCGGG GGCTAGCTACAACGA CCTAAGGA 1775
    293 GCACCCGG G CGCCGCCG 1255 CCCCGGCG CCCTAGCTACAACCA CCGGCTGC 1776
    295 ACCCCCCC G CCGGGCCC 1256 CGCCCCGC GCCTAGCTACAACGA CCCCGGCT 1777
    301 CCGCCGGC G CCCUCGCC 1257 GGCGAGGG GGCTAGCTACAACGA CCCCCCCC 1778
    307 GGGCCCUC G CCCUUCCG 1258 CGCAAGCC GCCTAGCTACAACCA GAGGGCCC 1779
    315 GCCCUUCC G CAGCCUUC 1259 GAAGGCTG GCCTAGCTACAACGA GGAACGGC 1780
    318 CUUCCGCA G CCUUCACU 1260 ACTCAAGC GGCTACCTACAACCA TGCCGAAG 1781
    324 CAGCCUUC A CUCCACCC 541 CCCTCGAG GGCTACCTACAACGA CAACCCTG 1782
    330 UCACUCCA G CCCUCUGC 1261 GCACACGG GGCTAGCTACAACCA TGGACTCA 1783
    337 ACCCCUCU G CUCCCGCA 1262 TCCGCGAG GCCTACCTACAACGA ACAGGCCT 1784
    343 CUGCUCCC G CACCCCAU 1263 ATGCCGTG CCCTACCTACAACGA CCGACCAC 1785
    345 GCUCCCCC A CGCCAUCA 552 TCATGCCC CGCTACCTACAACCA GCGGCAGC 1786
    347 UCCCGCAC G CCAUCAAG 1264 CTTCATGC GCCTAGCTACAACCA GTGCGGGA 1787
    350 CGCACGCC A UCAACUCC 554 CGACTTCA GGCTACCTACAACGA GGCGTCCG 1788
    355 CCCAUCAA G UCGCCGUU 1265 AACGCCGA GGCTAGCTACAACCA TTCATCGC 1789
    358 AUCAACUC G CCCUUCUA 1266 TACAACCG CGCTAGCTACAACGA GACTTCAT 1790
    361 AAGUCGCC G UUCUACCC 1267 CGCTACAA GGCTAGCTACAACCA CCCGACTT 1791
    366 GCCCUUCU A CCCCUGCC 55 GCCAGCGG GCCTAGCTACAACCA ACAACCGC 1792
    369 GUUCUACC G CUGCCAGA 1268 TCTGCCAG GCCTACCTACAACGA GGTACAAC 1793
    372 CUACCCCU G CCAGAACA 1269 TGTTCTCC GCCTAGCTACAACCA AGCCGTAG 1794
    378 CUCCCACA A CACCACCU 1675 ACCTCCTC GCCTACCTACAACGA TCTCCCAC 1795
    380 GCCACAAC A CCACCUCU 561 ACACCTCC CCCTACCTACAACCA CTTCTCCC 1796
    383 AGAACACC A CCUCUCUG 563 CACAGAGG CCCTACCTACAACGA CCTCTTCT 1797
    389 CCACCUCU G UCGAAAAA 1270 TTTTTCCA CGCTAGCTACAACCA AGAGCTCC 1798
    399 CCAAAAAG G CAACUCCG 1271 CCCAGTTG CCCTACCTACAACGA CTTTTTCC 1799
    402 AAAACCCA A CUCGCCCC 1676 CCCCCCAC CCCTACCTACAACCA TGCCTTTT 1800
    407 CCAACUCG G CCCUCAUC 1272 CATCACCC GCCTACCTACAACCA CCACTTCC 1801
    410 ACUCCCCC G UGAUCGCC 1273 CCCCATCA CCCTACCTACAACQA CGCCGACT 1802
    413 CCGCCCUG A UCCCCCCC 1677 CCCCCCCA CCCTACCTACAACCA CACCCCCC 1803
    417 CCUCAUCC G CCGCCUCC 1274 CCACCCCC CGCTAGCTACAACCA CCATCACC 1804
    422 UCCCCCCC G UCCUCUUC 1275 CAACACCA CCCTACCTACAACCA CCCCCCCA 1805
    424 CCCCCGCU G CUCUUCAG 1276 CTCAACAC CCCTACCTACAACGA ACCCCCCC 1806
    432 CCUCUUCA G CACCCCCC 1277 CCCCCCTC CCCTACCTACAACCA TCAAGACC 1807
    434 UCUUCACC A CCGGCCUC 572 CACGCCCC CCCTACCTACAACCA CCTCAAGA 1808
    438 CACCACCG G CCUCCUCC 1278 CCACCAGC GCCTACCTACAACGA CGCTCCTC 1809
    447 CCUCCUCC G CAACCUGC 1279 CCACCTTC CCCTAGCTACAACCA CCAGGAGC 1810
    450 CCUGCCCA A CCUCCUCC 1678 CCACCAGC GCCTACCTACAACGA TCCCCACC 1811
    454 GCCAACCU G CUGGCCCU 1280 AGGGCCAC GGCTAGCTACAACGA AGGTTGCC 1812
    458 ACCUGCUG G CCCUGGGG 1281 CCCCAGGG GGCTAGCTACAACGA CAGCAGGT 1813
    466 GCCCUGGG G CUGCUCGC 1282 GCCAGCAG GGCTAGCTACAACGA CCCAGCGC 1814
    469 CUCGGGCU G CUGGCGCG 1283 CGCGCCAC CGCTAGCTACAACGA AGCCCCAG 1815
    473 GCCUGCUG G CCCGCUCG 1284 CGAGCGCG GGCTAGCTACAACGA CAGCAGCC 1816
    475 CUGCUGGC G CGCUCGGG 1285 CCCGAGCG CGCTAGCTACAACGA GCCAGCAG 1817
    477 GCUCGCGC G CUCCCGGC 1286 GCCCCGAC GGCTACCTACAACGA GCGCCAGC 1818
    484 CCCUCGGG G CUGGCGUG 1287 CACCCCAG GGCTAGCTACAACCA CCCGAGCG 1819
    490 GGCCUGGC G UGCUGCUC 1288 CACCACCA GCCTACCTACAACGA CCCAGCCC 1820
    493 CUCCGGUG G UGCUCCCC 1289 CGCCAGCA CCCTAGCTACAACGA CACCCCAG 1821
    495 CCGGUCGU G CUCCCGGC 1290 CCCCCGAG GGCTAGCTACAACCA ACCACCCC 1822
    499 UCGUGCUC G CCGCCUCC 1291 CCACCCCC GGCTAGCTACAACGA CAGCACCA 1823
    502 UCCUCGCC G CCUCCACU 1292 ACTGGACG GCCTACCTACAACCA CGCCAGCA 1824
    504 CUCGCGCC G UCCACUCC 1293 CCAGTCGA GGCTAGCTACAACCA GCCGCGAG 1825
    508 CCCCGUCC A CUCCCCCC 591 GCGCGCAG GCCTAGCTACAACGA CGACGCCC 1826
    511 CGUCCACU G CGCCCGCU 1294 AGCGGGCG GGCTAGCTACAACGA AGTCCACC 1827
    513 UCCACUCC G CCCGCUGC 1295 GCACCCGC CGCTAGCTACAACCA GCACTCGA 1828
    517 CUGCGCCC G CUCCCCUC 1296 CAGGUCAG GGCTACCTACAACGA GGGCGCAG 1829
    520 CCCCCCCU G CCCUCCGU 1297 ACCGAGGG GGCTAGCTACAACCA AGCCCCCG 1830
    527 UGCCCUCC G UCUUCUAC 1298 GTAGAAGA GCCTAGCTACAACGA CCAGGGCA 1831
    534 GCUCUUCU A CAUGCUCC 69 CCACCATG GGCTAGCTACAACCA ACAAGACC 1832
    536 UCUCCUAC A UGCUGCUC 601 CACCACCA CCCTACCTACAACGA GTAGAAGA 1833
    538 UUCUACAU G CUGGUGUG 1299 CACACCAG CGCTAGCTACAACGA ATGTAGAA 1834
    542 ACAUGCUG G UCUCUCCC 1300 CCCACACA CGCTACCTACAACCA CACCATGT 1835
    544 AUGCUGGU G UGUGGCCU 1301 ACGCCACA GCCTACCTACAACGA ACCAGCAT 1836
    546 CCUGGUGU G UCCCCUGA 1302 TCACGCCA GGCTAGCTACAACCA ACACCAGC 1837
    549 GGUCUGUG G CCUGACGG 1303 CCGTCAGC GCCTAGCTACAACGA CACACACC 1838
    554 CUGCCCUG A CCGUCACC 1679 GGTGACCC GGCTAGCTACAACGA CAGGOCAC 1839
    557 CCCUCACC G UCACOGAC 1304 CTCCCTGA CCCTACCTACAACGA CCTCACCC 1840
    560 UCACCCUC A CCCACUUC 605 CAACTCCC CGCTAGCTACAACCA GACCGTCA 1841
    564 GGUCACCC A CUUCCUGG 1680 CCACCAAC CCCTACCTACAACCA CCCTCACC 1842
    568 ACCGACUU G CUCCCCAA 1305 TTCCCCAC GCCTAGCTACAACCA AAGTCGCT 1843
    573 CUUCCUCC G CAACUCCC 1306 GGCACTTC CCCTAGCTACAACCA CCACCAAG 1844
    577 CUCCCCAA G UCCCUCCU 1307 ACGACGCA CCCTACCTACAACGA TTCCCCAC 1845
    579 CCGCAAGU G CCUCCUAA 1308 TTACCACG CCCTACCTACAACCA ACTTGCCC 1846
    588 CCUCCUAA G CCCGGUCC 1309 CCACCCCC CCCTAGCTACAACCA TTACCAGG 1847
    593 UAAGCCCC G UCCUCCUG 1310 CACCACCA GGCTACCTACAACGA CGGGCTTA 1848
    596 CCCCCCUG G UGCUGCCU 1311 AGCCACCA CCCTAGCTACAACCA CACCCGGC 1849
    598 CCGGUGGU G CUCCCUGC 1312 CCACCCAG GGCTACCTACAACGA ACCACCCC 1850
    602 UCCUCCUC G CUGCCUAC 1313 GTAGCCAC CCCTAGCTACAACCA CACCACCA 1851
    605 UGCUGGCU G CCUACCCU 1314 ACCCTAGG GGCTACCTACAACGA AGCCACCA 1852
    609 CCCUCCCU A CGCUCACA 74 TCTCACCC CCCTAGCTACAACCA ACCCACCC 1853
    611 CUGCCUAC G CUCAGAAC 1315 CTTCTGAG CCCTACCTACAACGA GTACCCAG 1854
    618 CGCUCAGA A CCCCACUC 1681 CACTCCCG CCCTAGCTACAACCA TCTGAGCC 1855
    624 CAACCCGA G UCUCCCCC 1316 CCCCCACA CCCTACCTACAACCA TCCCCTTC 1856
    628 CCCACUCU G CGCGUGCU 1317 ACCACCCG CCCTACCTACAACGA AGACTCCC 1857
    632 GUCUGCGC G UCCUUCCG 1318 CCCAACCA CGCTAGCTACAACCA CCCCACAC 1858
    634 CUCCCCCU G CUUGCCCC 1319 GGCGCAAG CCCTAGCTACAACGA ACCCCCAG 1859
    638 CGGUGCUU G CCCCCCCA 1320 TCCCCCCG GGCTACCTACAACCA AACCACCC 1860
    640 CUCCUUCC G CCCGCAUU 1321 AATCCGGG CCCTAGCTACAACGA CCAACCAC 1861
    644 UUGCCCCC G CAUUCCAC 1322 GTCCAATC GCCTACCTACAACCA GGCCGCAA 1862
    646 GCGCCCGC A UUGGACAA 627 TTGTCCAA GGCTAGCTACAACGA GCGGGCGC 1863
    651 CGCAUUGG A CAACUCGU 1682 ACGAGTTG GGCTAGCTACAACGA CCAATGCG 1864
    654 AUUGGACA A CUCGUUGU 1683 ACAACGAG GGCTAGCTACAACGA TGTCCAAT 1865
    658 GACAACUC G UUGUGCCA 1323 TGGCACAA GGCTAGCTACAACGA GAGTTGTC 1866
    661 AACUCGUU G UGCCAAGC 1324 GCTTGGCA GGCTAGCTACAACGA AACGAGTT 1867
    663 CUCGUUGU G CCAAGCCU 1325 AGGCTTGG GGCTAGCTACAACGA ACAACGAG 1868
    668 UGUGCCAA G CCUUCGCC 1326 GGCGAAGG GGCTAGCTACAACGA TTGGCACA 1869
    674 AAGCCUUC G CCUUCUUC 1327 GAAGAAGG GGCTAGCTACAACGA GAAGGCTT 1870
    683 CCUUCUUC A UGUCCUUC 637 GAAGGACA GGCTAGCTACAACGA GAAGAAGG 1871
    685 UUCUUCAU G UCCUUCUU 1328 AAGAAGGA CGCTAGCTACAACGA ATGAAGAA 1872
    697 UUCUUUGG G CUCUCCUC 1329 GAGGAGAG GGCTAGCTACAACGA CCAAAGAA 1873
    707 UCUCCUCG A CACUGCAA 1684 TTGCAGTG GGCTAGCTACAACGA CUAGGAGA 1874
    709 UCCUCGAC A CUGCAACU 645 AGTTGCAG GGCTAGCTACAACGA GTCGAGGA 1875
    712 UCGACACU G CAACUCCU 1330 AGGAGTTG GGCTAGCTACAACGA AGTGTCGA 1876
    715 ACACUGCA A CUCCUGGC 1685 GCCAGGAG GGCTAGCTACAACGA TGCAGTGT 1877
    722 AACUCCUG G CCAUGGCA 1331 TGCCATGG GGCTAGCTACAACGA CAGGAGTT 1878
    725 UCCUGGCC A UGGCACUG 652 CAGTGCCA GGCTAGCTACAACGA GGCCAGGA 1879
    728 UGGCCAUG G CACUGGAG 1332 CTCCAGTG GGCTAGCTACAACGA CATGGCCA 1880
    730 GCCAUGGC A CUGGAGUG 653 CACTCCAG GGCTAGCTACAACGA GCCATGGC 1881
    736 GCACUGGA G UGCUGGCU 1333 AGCCAGCA GGCTAGCTACAACGA TCCAGTGC 1882
    738 ACUGGAGU G CUGGCUCU 1334 AGAGCCAG GGCTAGCTACAACGA ACTCCAGT 1883
    742 GAGUCCUG G CUCUCCCU 1335 AGGGAGAG GCCTAGCTACAACGA CAGCACTC 1884
    754 UCCCUAGG G CACCCUUU 1336 AAAGGGTG GGCTAGCTACAACGA CCTAGGGA 1885
    756 CCUAGGGC A CCCUUUCU 661 AGAAAGGG GGCTAGCTACAACGA GCCCTAGG 1886
    768 UUUCUUCU A CCGACGGC 104 GCCGTCGG GGCTAGCTACAACGA AGAACAAA 1887
    772 UUCUACCG A CGGCACAU 1686 ATGTGCCG GGCTAGCTACAACGA CGGTAGAA 1888
    775 UACCGACG G CACAUCAC 1337 GTGATGTG GGCTAGCTACAACGA CGTCGGTA 1889
    777 CCGACGGC A CAUCACCC 668 GGGTGATG GGCTAGCTACAACGA GCCGTCGG 1890
    779 GACGGCAC A UCACCCUG 669 CAGGGTGA GGCTAGCTACAACGA GTCCCGTC 1891
    782 CGCACAUC A CCCUGCGC 670 GCGCAGGG GGCTAGCTACAACGA GATGTGCC 1892
    787 AUCACCCU G CGCCUGGG 1338 CCCAGGCG GCCTAGCTACAACGA AGGGTGAT 1893
    789 CACCCUGC G CCUGGGCG 1339 CGCCCAGG GGCTACCTACAACGA GCAGCGTG 1894
    795 GCGCCUGG G CGCACUGG 1340 CCAGTCCG GGCTAGCTACAACGA CCAGGCGC 1895
    797 GCCUCGGC G CACUGGUG 1341 CACCAGTG GCCTAGCTACAACGA GCCCAGGC 1896
    799 CUGGGCGC A CUGGUGGC 676 GCCACCAG GGCTAGCTACAACGA GCGCCCAG 1897
    803 CCCCACUG G UGGCCCCC 1342 CGGGGCCA CGCTAGCTACAACGA CAGTGCGC 1898
    806 CACUGGUG G CCCCGGUG 1343 CACCGGGG GGCTAGCTACAACGA CACCAGTG 1899
    812 UCGCCCCG G UGGUGAGC 1344 GCTCACCA GGCTAGCTACAACGA CGGGGCCA 1900
    815 CCCCGGUG G UGAGCGCC 1345 GGCGCTCA GGCTAGCTACAACGA CACCGGGC 1901
    819 GGUGGUGA G CGCCUUCU 1346 AGAAGGCG CGCTAGCTACAACGA TCACCACC 1902
    821 UGGUGAGC G CCUUCUCC 1347 GGAGAAGG GGCTAGCTACAACGA GCTCACCA 1903
    833 UCUCCCUG G CUUUCUGC 1348 GCACAAAG GGCTAGCTACAACCA CAGGGAGA 1904
    840 GGCUUUCU G CGCGCUAC 1349 GTAGCGCG CGCTAGCTACAACGA AGAAAGCC 1905
    842 CUUUCUGC G CCCUACCU 1350 ACCTACCG GGCTAGCTACAACGA GCAGAAAG 1906
    844 UUCUCCGC G CUACCUUU 1351 AAAGGTAG GCCTAGCTACAACCA CCGCAGAA 1907
    847 UGCCCCCU A CCUUUCAU 112 ATGAAAGG GGCTACCTACAACGA AGCGCCCA 1908
    854 UACCUUUC A UGCCCUUC 692 CAACCCCA GGCTAGCTACAACCA CAAAGGTA 1909
    858 UUUCAUCG G CUUCGGGA 1352 TCCCGAAG GGCTAGCTACAACGA CCATGAAA 1910
    868 UUCGGGAA G UUCCUGCA 1353 TGCACGAA GGCTAGCTACAACGA TTCCCGAA 1911
    872 GGAAGUUC G UGCAGUAC 1354 GTACTGCA GGCTAGCTACAACGA GAACTTCC 1912
    874 AAGUUCGU G CACUACUG 1355 CAGTACTG GGCTAGCTACAACGA ACGAACTT 1913
    877 UUCGUGCA G UACUGCCC 1356 GGGCAGTA GGCTAGCTACAACGA TGCACGAA 1914
    879 CGUGCAGU A CUGCCCCG 120 CGGGGCAG GGCTAGCTACAACGA ACTGCACG 1915
    882 GCAGUACU G CCCCGGCA 1357 TGCCGGGG GGCTAGCTACAACGA AGTACTGC 1916
    888 CUGCCCCG G CACCUGGU 1358 ACCAGGTG GGCTAGCTACAACGA CGGGGCAG 1917
    890 GCCCCGGC A CCUGGUGC 699 GCACCAGG GGCTAGCTACAACGA GCCGGGGC 1918
    895 GGCACCUG G UGCUUUAU 1359 ATAAAGCA GGCTAGCTACAACGA CAGGTGCC 1919
    897 CACCUGGU G CUUUAUCC 1360 GGATAAAG GGCTAGCTACAACGA ACCAGGTG 1920
    902 GGUGCUUU A UCCAGAUG 123 CATCTGGA GGCTAGCTACAACGA AAAGCACC 1921
    908 UUAUCCAG A UGGUCCAC 1687 GTGGACCA GGCTAGCTACAACGA CTGGATAA 1922
    911 UCCAGAUG G UCCACGAG 1361 CTCGTGGA GGCTAGCTACAACGA CATCTGGA 1923
    915 GAUGGUCC A CGAGGAGG 706 CCTCCTCG GGCTAGCTACAACGA GGACCATC 1924
    924 CGAGGAGG G CUCGCUGU 1362 ACAGCGAG GGCTAGCTACAACGA CCTCCTCG 1925
    928 GAGGGCUC G CUGUCGGU 1363 ACCUACAG GGCTAGCTACAACGA GAGCCCTC 1926
    931 GGCUCGCU G UCGGUGCU 1364 AGCACCGA GGCTAGCTACAACGA AGCGAGCC 1927
    935 CGCUGUCG G UGCUGGGG 1365 CCCCAGCA GGCTAGCTACAACGA CGACAGCG 1928
    937 CUGUCGGU G CUGGGGUA 1366 TACCCCAG GGCTAGCTACAACGA ACCGACAG 1929
    943 GUGCUGGG G UACUCUGU 1367 ACAGAUTA GGCTAGCTACAACGA CCCAGCAC 1930
    945 GCUGGGGU A CUCUGUGC 128 GCACAGAG GGCTAGCTACAACGA ACCCCAGC 1931
    950 GGUACUCU G UGCUCUAC 1368 GTAGAGCA GGCTAGCTACAACGA AGAGTACC 1932
    952 UACUCUGU G CUCUACUC 1369 GAGTAGAG GGCTAGCTACAACGA ACAGAUTA 1933
    957 UGUGCUCU A CUCCAGCC 131 GGCTGGAG GGCTAGCTACAACGA AGAGCACA 1934
    963 CUACUCCA G CCUCAUGG 1370 CCATGAGG GGCTAGCTACAACGA TGGAGTAG 1935
    968 CCAGCCUC A UGGCGCUG 719 CAUCUCCA GGCTAGCTACAACGA GAGGCTGG 1936
    971 GCCUCAUG G CGCUGCUG 1371 CAGCAGCG GGCTAGCTACAACGA CATGAGGC 1937
    973 CUCAUGGC G CUGCUGGU 1372 ACCACCAG GGCTAGCTACAACGA GCCATGAG 1938
    976 AUGGCGCU G CUGGUCCU 1373 AGGACCAG GGCTAGCTACAACGA AGCGCCAT 1939
    980 CGCUGCUG G UCCUCGCC 1374 GGCGAGGA GGCTAGCTACAACGA CAGCAGCG 1940
    986 UGGUCCUC G CCACCGUG 1375 CACGGTGG GGCTAGCTACAACGA GAGGACCA 1941
    989 UCCUCGCC A CCGUGCUG 725 CAGCACGG GGCTAGCTACAACGA GGCGAGGA 1942
    992 UCGCCACC G UGCUGUGC 1376 GCACAGCA GGCTAGCTACAACGA GGTGGCGA 1943
    994 GCCACCGU G CUGUGCAA 1377 TTGCACAG GGCTAGCTACAACGA ACGGTGGC 1944
    997 ACCGUGCU G UGCAACCU 1378 AGGTTGCA GGCTAGCTACAACGA AGCACGGT 1945
    999 CGUGCUGU G CAACCUCG 1379 CGAGGTTG GGCTAGCTACAACGA ACAGCACG 1946
    1002 GCUGUGCA A CCUCGGCG 1688 CGCCGAGG GGCTAGCTACAACGA TGCACAGC 1947
    1008 CAACCUCG G CGCCAUGC 1380 GCATGGCG GGCTAGCTACAACGA CGAGGTTG 1948
    1010 ACCUCGGC G CCAUGCGC 1381 GCGCATGG GGCTAGCTACAACGA GCCGAGGT 1949
    1013 UCGGCGCC A UGCGCAAC 732 GTTGCGCA GGCTAGCTACAACGA GGCGCCGA 1950
    1015 GGCGCCAU G CGCAACCU 1382 AGGTTGCG GGCTAGCTACAACGA ATGGCGCC 1951
    1017 CGCCAUGC G CAACCUCU 1383 AGAGGTTG GGCTAGCTACAACGA GCATGGCG 1952
    1020 CAUGCGCA A CCUCUAUG 1689 CATAGAGG GGCTAGCTACAACGA TGCGCATG 1953
    1026 CAACCUCU A UGCGAUGC 138 GCATCGCA GGCTAGCTACAACGA AGAGGTTG 1954
    1028 ACCUCUAU G CGAUGCAC 1384 GTGCATCG GGCTAGCTACAACGA ATAGAGGT 1955
    1031 UCUAUGCG A UGCACCGG 1690 CCGGTGCA GGCTAGCTACAACGA CGCATAGA 1956
    1033 UAUGCGAU G CACCGGCG 1385 CGCCGGTG GGCTAGCTACAACGA ATCGCATA 1957
    1035 UGCGAUGC A CCGGCGGC 737 GCCGCCGG GGCTAGCTACAACGA GCATCGCA 1958
    1039 AUGCACCG G CGGCUGCA 1386 TGCAGCCG GGCTAGCTACAACGA CGGTGCAT 1959
    1042 CACCGGCG G CUGCAGCG 1387 CGCTGCAG GGCTAGCTACAACGA CGCCGGTG 1960
    1045 CGGCGGCU G CAGCGGCA 1388 TGCCGCTG GGCTAGCTACAACGA AGCCGCCG 1961
    1048 CGGCUGCA G CGGCACCC 1389 GGGTGCCG GGCTAGCTACAACGA TGCAGCCG 1962
    1051 CUGCAGCG G CACCCGCG 1390 CGCGGGTG GGCTAGCTACAACGA CGCTGCAG 1963
    1053 GCAGCGGC A CCCGCGCU 741 AGCGCGGG GGCTAGCTACAACGA GCCGCTGC 1964
    1057 CGGCACCC G CGCUCCUG 1391 CACCAGCG GGCTAGCTACAACGA GGGTGCCG 1965
    1059 GCACCCGC G CUCCUCCA 1392 TGCAGGAG GGCTACCTACAACGA GCGGGTGC 1966
    1065 GCGCUCCU G CACCAGGG 1393 CCCTCCTG GGCTAGCTACAACGA AGGAGCGC 1967
    1067 GCUCCUGC A CCAGGCAC 747 GTCCCTGG GGCTAGCTACAACCA GCAGGAGC 1968
    1074 CACCAGGG A CUGUGCCG 1691 CGGCACAG GGCTAGCTACAACGA CCCTGGTG 1969
    1077 CAGGGACU G UGCCGAGC 1394 GCTCGGCA GGCTAGCTACAACGA AGTCCCTG 1970
    1079 GGGACUGU G CCGAGCCG 1395 CGGCTCGG GGCTAGCTACAACGA ACAGTCCC 1971
    1084 UGUGCCCA G CCGCGCCC 1396 GCGCGCGG CGCTAGCTACAACGA TCGGCACA 1972
    1087 GCCGAGCC G CGCGCGGA 1397 TCCGCGCG GGCTAGCTACAACGA GGCTCGGC 1973
    1089 CGAGCCGC G CGCGGACG 1398 CGTCCGCG GGCTAGCTACAACGA GCGGCTCG 1974
    1091 AGCCGCGC G CGGACGGG 1399 CCCGTCCG GGCTAGCTACAACGA GCGCGGCT 1975
    1095 GCGCGCGG A CGJGAGGG 1692 CCCTCCCG GGCTAGCTACAACGA CCGCCCGC 1976
    1106 GGAGCGAA G CGUCCCCU 1400 AGGGGACG GGCTAGCTACAACGA TTCCCTCC 1977
    1108 AGGGAAGC G UCCCCUCA 1401 TGAGGGGA GGCTAGCTACAACGA CCTTCCCT 1978
    1117 UCCCCUCA G CCCCUCGA 1402 TCCAGGGG GCCTAGCTACAACGA TGAGGGGA 1979
    1129 CUGGAGGA G CUCCAUCA 1403 TGATCCAG GGCTACCTACAACGA TCCTCCAG 1980
    1134 GCAGCUGG A UCACCUCC 1693 GGAGGTGA GGCTAGCTACAACGA CCAGCTCC 1981
    1137 CCUGGAUC A CCUCCUGC 763 GCAGGAGG CCCTAGCTACAACCA GATCCAGC 1982
    1144 CACCUCCU G CUGCUGGC 1404 GCCAGCAG GCCTAGCTACAACCA AGCAGGTG 1983
    1147 CUCCUCCU G CUGCCGCU 1405 AGCCCCAG GGCTACCTACAACGA AGCACGAG 1984
    1151 UGCUGCUC G CCCUGAUG 1406 CATCAGCC GCCTACCTACAACGA CAGCAGCA 1985
    1153 CUGCUGGC G CUGAUGAC 1407 GTCATCAG CGCTAGCTACAACGA GCCAGCAG 1986
    1157 UGGCCCUG A UGACCOUG 1694 CACGGTCA GGCTAGCTACAACGA CACCGCCA 1987
    1160 CCCUGAUC A CCGUGCUC 1695 CAGCACCC GGCTAGCTACAACGA CATCAGCG 1988
    1163 UCAUGACC G UGCUCUUC 1408 GAAGAGCA GCCTACCTACAACGA CGTCATCA 1989
    1165 AUGACCCU G CUCUUCAC 1409 GTCAACAG CGCTAGCTACAACGA ACGGTCAT 1990
    1172 UGCUCUUC A CUAUGUGU 774 ACACATAC GGCTAGCTACAACGA GAAGAGCA 1991
    1175 UCUUCACU A UCUGUUCU 147 AGAACACA GCCTAGCTACAACCA AGTCAACA 1992
    1177 UUCACUAU G UGUUCUCU 1410 AGAGAACA CGCTACCTACAACGA ATACTCAA 1993
    1179 CACUAUGU G UUCUCUGC 1411 GCAGAGAA CCCTACCTACAACCA ACATAGTG 1994
    1186 UCUUCUCU G CCCCUAAU 1412 ATTACGGC CGCTACCTACAACGA ACACAACA 1995
    1190 CUCUGCCC G UAAUUUAU 1413 ATAAATTA GCCTACCTACAACGA GGGCAGAC 1996
    1193 UCCCCGUA A UUUAUCCC 1696 CCCATAAA GGCTAGCTACAACCA TACGGGCA 1997
    1197 CGUAAUUU A UCCCCCUU 154 AACCCCCA CGCTAGCTACAACGA AAATTACG 1998
    1200 AAUUUAUC G CGCUUACU 1414 AGTAACCG GGCTAGCTACAACCA GATAAATT 1999
    1202 UUUAUCGC G CUUACUAU 1415 ATACTAAG CGCTAGCTACAACGA GCGATAAA 2000
    1206 UCGCGCUU A CUAUGCAC 157 CTCCATAG GGCTAGCTACAACCA AACCGCGA 2001
    1209 CCCUUACU A UCGAGCAU 158 ATCCTCCA CGCTACCTACAACGA AGTAAGCC 2002
    1214 ACUAUGGA G CAUUUAAG 1416 CTTAAATG GCCTACCTACAACCA TCCATAGT 2003
    1216 UAUCCACC A UUUAAGGA 782 TCCTTAAA GGCTACCTACAACGA CCTCCATA 2004
    1224 AUUUAAGC A UGUCAACG 1697 CCTTCACA CGCTACCTACAACGA CCTTAAAT 2005
    1226 UUAACGAU G UCAACGAG 1417 CTCCTTGA GGCTAGCTACAACCA ATCCTTAA 2006
    1239 CGAGAAAA A CAGGACCU 1698 AGGTCCTG GGCTAGCTACAACGA TTTTCTCC 2007
    1244 AAAACACG A CCUCUCAA 1699 TTCAGAGG GGCTACCTACAACCA CCTCTTTT 2008
    1256 CUCAACAA G CAGAAGAC 1418 CTCTTCTC CCCTACCTACAACCA TTCTTCAC 2009
    1263 ACCAGAAG A CCUCCCAC 1700 CTCGCACG CGCTACCTACAACCA CTTCTGCT 2010
    1271 ACCUCCCA G CCUUCCGA 1419 TCCCAACC CCCTACCTACAACGA TCCCACCT 2011
    1276 CCACCCUU G CCAUUUCU 1420 ACAAATCC CCCTACCTACAACCA AAGGCTCG 2012
    1279 CCCUUCCC A UUUCUAUC 1701 CATACAAA GGCTACCTACAACCA CCCAACCC 2013
    1285 CCAUUUCU A UCUGUGAU 169 ATCACACA CCCTACCTACAACGA AGAAATCC 2014
    1289 UUCUAUCU G UCAUUUCA 1421 TCAAATCA GCCTACCTACAACCA ACATACAA 2015
    1292 UAUCUGUG A UUUCAAUU 1702 AATTGAAA GGCTAGCTACAACGA CACAGATA 2016
    1298 UGAUUUCA A UHOUGGAC 1703 GTCCACAA GGCTAGCTACAACGA TGAAATCA 2017
    1301 UUUCAAUU G UGGACCCU 1422 AGGGTCCA GGCTAGCTACAACGA AATTGAAA 2018
    1305 AAUUGUGG A CCCUUGGA 1704 TCCAAGGG GGCTAGCTACAACGA CCACAATT 2019
    1313 ACCCUUGG A UUUUUAUC 1705 GATAAAAA GGCTAGCTACAACGA CCAAGGGT 2020
    1319 GGAUUUUU A UCAUUUUC 180 GAAAATGA GGCTAGCTACAACGA AAAAATCC 2021
    1322 UUUUUAUC A UUUUCAGA 800 TCTGAAAA GGCTAGCTACAACGA GATAAAAA 2022
    1330 AUUUUCAG A UCUCCAGU 1706 ACTUGAGA GGCTAGCTACAACGA CTGAAAAT 2023
    1337 GAUCUCCA G UAUUUCGG 1423 CCGAAATA GGCTAGCTACAACGA TGGAGATC 2024
    1339 UCUCCAGU A UUUCGGAU 188 ATCCGAAA GGCTAGCTACAACGA ACTGGAGA 2025
    1346 UAUUUCGG A UAUUUUUU 1707 AAAAAATA GGCTAGCTACAACGA CCGAAATA 2026
    1348 UUUCGGAU A UUUUUUCA 192 TGAAAAAA GGCTAGCTACAACGA ATCCGAAA 2027
    1356 AUUUUUUC A CAAGAUUU 805 AAATCTTG GGCTAGCTACAACGA GAAAAAAT 2028
    1361 UUCACAAG A UUUUCAUU 1708 AATGAAAA GGCTAGCTACAACGA CTTGTGAA 2029
    1367 AGAUUUUC A UUAGACCU 807 AGGTCTAA GGCTAGCTACAACGA GAAAATCT 2030
    1372 UUCAUUAG A CCUCUUAG 1709 CTAAGAGG GGCTAGCTACAACGA CTAATGAA 2031
    1381 CCUCUUAG G UACAGGAG 1424 CTCCTGTA GGCTAGCTACAACGA CTAAGAGG 2032
    1383 UCUUAGGU A CAGGAGCC 208 GGCTCCTG GGCTAGCTACAACGA ACCTAAGA 2033
    1389 GUACAGGA G CCGGUGCA 1425 TGCACCGG GGCTAGCTACAACGA TCCTGTAC 2034
    1393 AGGAGCCG G UGCAGCAA 1426 TTGCTGCA GGCTAGCTACAACGA CGGCTCCT 2035
    1395 GAGCCGGU G CAGCAAUU 1427 AATTGCTG GGCTAGCTACAACGA ACCGGCTC 2036
    1398 CCGGUGCA G CAAUUCCA 1428 TGGAATTG GGCTAGCTACAACGA TGCACCGG 2037
    1401 GUGCAGCA A UUCCACUA 1710 TAGTGGAA GGCTAGCTACAACGA TGCTGCAC 2038
    1406 GCAAUUCC A CUAACAUG 816 CATGTTAG GGCTAGCTACAACGA GGAATTGC 2039
    1410 UUCCACUA A CAUGGAAU 1711 ATTCCATG GGCTAGCTACAACGA TAGTGGAA 2040
    1412 CCACUAAC A UGGAAUCC 818 GGATTCCA GGCTAGCTACAACGA GTTAGTGG 2041
    1417 AACAUGGA A UCCAGUCU 1712 AGACTGGA GGCTAGCTACAACGA TCCATGTT 2042
    1422 GGAAUCCA G UCUGUGAC 1429 GTCACAGA GGCTAGCTACAACGA TGGATTCC 2043
    1426 UCCAGUCU G UGACAGUG 1430 CACTGTCA GGCTAGCTACAACGA AGACTGGA 2044
    1429 AGUCUGUG A CAGUGUUU 1713 AAACACTG GGCTAGCTACAACGA CACAGACT 2045
    1432 CUGUGACA G UGUUUUUC 1431 GAAAAACA GGCTAGCTACAACGA TGTCACAG 2046
    1434 GUGACAGU G UUUUUCAC 1432 GTGAAAAA GGCTAGCTACAACGA ACTGTCAC 2047
    1441 UGUUUUUC A CUCUGUGG 823 CCACAGAG GGCTAGCTACAACGA GAAAAACA 2048
    1446 UUCACUCU G UGGUAAGC 1433 GCTTACCA GGCTAGCTACAACGA AGAGTGAA 2049
    1449 ACUCUGUG G UAAGCUGA 1434 TCAGCTTA GGCTAGCTACAACGA CACAGAGT 2050
    1453 UGUGGUAA G CUGAGGAA 1435 TTCCTCAG GGCTAGCTACAACGA TTACCACA 2051
    1461 GCUGAGGA A UAUGUCAC 1714 GTGACATA GGCTAGCTACAACGA TCCTCAGC 2052
    1463 UGAGGAAU A UGUCACAU 221 ATGTGACA GGCTAGCTACAACGA ATTCCTCA 2053
    1465 AGGAAUAU G UCACAUUU 1436 AAATGTGA GGCTAGCTACAACGA ATATTCCT 2054
    1468 AAUAUGUC A CAUUUUCA 827 TGAAAATG GGCTAGCTACAACGA GACATATT 2055
    1470 UAUGUCAC A UUUUCAGU 828 ACTGAAAA GGCTAGCTACAACGA GTGACATA 2056
    1477 CAUUUUCA G UCAAAGAA 1437 TTCTTTGA GGCTAGCTACAACGA TGAAAATG 2057
  • [0234]
    TABLE VII
    Human PTGDR Amberzyme and Substrate Sequence
    Seq Seq
    Pos Substrate ID Amberzyme ID
    9 GAAUUCUG G CUAUUUUC 1207 GAAAAUAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGAAUUC 2247
    23 UUCCUCCU G CCGUUCCG 1208 CGGAACGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGAGGAA 2248
    28 CUCCUGCC G UUCCGACU 1209 AGUCGGAA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGCAGGAG 2249
    31 GCCGUUCC G ACUCGGCA 2058 UGCCGAGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGAACGGC 2250
    36 UCCGACUC G GCACCAGA 2059 UCUGGUGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GAGUCGGA 2251
    37 CCGACUCG G CACCAGAG 1210 CUCUGGUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGAGUCGG 2252
    43 CGGCACCA G AGUCUGUC 2060 GACAGACU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGGUGCCG 2253
    45 GCACCAGA G UCUGUCUC 1211 GAGACAGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCUGGUGC 2254
    49 CAGAGUCU G UCUCUACU 1212 AGUAGAGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGACUCUG 2255
    58 UCUCUACU G AGAACGCA 2061 UGCGUUCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGUAGAGA 2256
    60 UCUACUGA G AACGCAGC 2062 GCUGCGUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCAGUAGA 2257
    64 CUGAGAAC G CAGCGCGU 1213 ACGCGCUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GUUCUCAG 2258
    67 AGAACGCA G CGCGUCAG 1214 CUGACGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGCGUUCU 2259
    69 AACGCAGC G CGUCAGGG 1215 CCCUGACG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCUGCGUU 2260
    71 CGCAGCGC G UCAGGGCC 1216 GGCCCUGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCGCUGCG 2261
    75 GCGCGUCA G GGCCGAGC 2063 GCUCGGCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGACGCGC 2262
    76 CGCGUCAG G GCCGAGCU 2064 AGCUCGGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUGACGCG 2263
    77 GCGUCAGG G CCGAGCUC 1217 GAGCUCGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCUGACGC 2264
    80 UCAGGGCC G AGCUCUUC 2065 GAAGAGCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGCCCUGA 2265
    82 AGGGCCGA G CUCUUCAC 1218 GUGAAGAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCGGCCCU 2266
    92 UCUUCACU G GCCUGCUC 2066 GAGCAGGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGUGAAGA 2267
    93 CUUCACUG G CCUGCUCC 1219 GGAGCAGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGUGAAG 2268
    97 ACUGGCCU G CUCCGCGC 1220 GCGCGGAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGCCAGU 2269
    102 CCUGCUCC G CGCUCUUC 1221 GAAGAGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGAGCAGG 2270
    104 UGCUCCGC G CUCUUCAA 1222 UUGAAGAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCGGAGCA 2271
    114 UCUUCAAU G CCAGCGCC 1223 GGCGCUGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUUGAAGA 2272
    118 CAAUGCCA G CGCCAGGC 1224 GCCUGGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGGCAUUG 2273
    120 AUGCCAGC G CCAGGCGC 1225 GCGCCUGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCUGGCAU 2274
    124 CAGCGCCA G GCGCUCAC 2067 GUGAGCGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGGCGCUG 2275
    125 AGCGCCAG G CGCUCACC 1226 GGUGAGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUGGCGCU 2276
    127 CGCCAGGC G CUCACCCU 1227 AGGGUGAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCCUGGCG 2277
    136 CUCACCCU G CAGAGCGU 1228 ACGCUCUG GCAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGCUGAG 2278
    139 ACCCUGCA G AGCGUCCC 2068 GGGACGCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGCAGGGU 2279
    141 CCUGCAGA G CGUCCCGC 1229 GCGGGACG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCUGCAGG 2280
    143 UGCAGAGC G UCCCGCCU 1230 AGGCGGGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCUCUGCA 2281
    148 AGCGUCCC G CCUCUCAA 1231 UUGAGAGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGGACGCU 2282
    158 CUCUCAAA G AGGGGUGU 2069 ACACCCCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUUGAGAG 2283
    160 CUCAAAGA G GGGUGUGA 2070 UCACACCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCUUUGAG 2284
    161 UCAAAGAG G GGUGUGAC 2071 GUCACACC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUCUUUGA 2285
    162 CAAAGAGG G GUGUGACC 2072 UGUCACAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCUCUUUG 2286
    163 AAAGAGGG G UGUGACCC 1232 GGGUCACA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCCUCUUU 2287
    165 AGAGGGGU G UGACCCGC 1233 GCGGGUCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCCCUCU 2288
    167 AGGGGUGU U ACCCGCGA 2073 UCGCGGGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACACCCCU 2289
    172 UGUGACCC C CGAGUUUA 1234 UAAACUCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GUGUCACA 2290
    174 UGACCCGC C AGUUUAGA 2074 UCUAAACU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCGGGUCA 2291
    176 ACCCGCGA G UUUAGAUA 1235 UAUCUAAA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCGCGGGU 2292
    181 CGAGUUUA G AUAGGAGG 2075 CCUCCUAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UAAACUCG 2293
    185 UUUAGAUA C GAGGUUCC 2076 GGAACCUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UAUCUAAA 2294
    186 UUAGAUAG G AGGUUCCU 2077 AGGAACCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUAUCUAA 2295
    188 AGAUAGGA G GUUCCUGC 2078 GCAGGAAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCCUAUCU 2296
    189 GAUAGGAG G UUCCUGCC 1236 GGCAGGAA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUCCUAUC 2297
    195 AGGUUCCU G CCGUGGGG 1237 CCCCACGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGAACCU 2298
    198 UUCCUGCC G UGGGGAAC 1238 GUUCCCCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGCAGGAA 2299
    200 CCUGCCGU G GGGAACAC 2079 GUGUUCCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACGGCAGG 2300
    201 CUGCCGUG G GGAACACC 2080 GGUGUUCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CACGGCAG 2301
    202 UGCCGUGG C GAACACCC 2081 CCGUGUUC GCAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCACGGCA 2302
    203 GCCGUGGG G AACACCCC 2082 GGGGUGUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCCACGGC 2303
    212 AACACCCC G CCGCCCUC 1239 GAGGGCGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGGCUCUU 2304
    215 ACCCCGCC G CCCUCGCA 1240 UCCGAGGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGCGGGGU 2305
    221 CCGCCCUC G GAGCUUUU 2083 AAAAGCUC GGAGCAAACUCC CU UCAAGGACAUCGUCCGGG GAGGUCUG 2306
    222 CGCCCUCG G AGCUUUUU 2084 AAAAAGCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGAGGGCG 2307
    224 CCCUCGGA G CUUUUUCU 1241 AGAAAAAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCCGAGGG 2308
    233 CUUUUUCU G UGGCGCAG 1242 CUGCGCCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGAAAAAG 2309
    235 UUUUCUGU G GCGCAGCU 2085 AGCUGCGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACAGAAPA 2310
    236 UUUCUGUG G CGCAGCUU 1243 AAGCUGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CACAGAAA 2311
    238 UCUGUGGC C CAGCUUCU 1244 AGAAGCUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCCACAGA 2312
    241 GUGGCGCA G CUUCUCCG 1245 CGGACAAG GGAGGAAACUCC CU UCAAGCACAUCGUCCGGG UGCGCCAC 2313
    249 GCUUCUCC G CCCGAGCC 1246 GGCUCGGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGAGAAGC 2314
    253 CUCCGCCC C AGCCGCGC 2086 GCGCGGCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGGCGGAG 2315
    255 CCGCCCGA G CCGCGCGC 1247 GCGCGCGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCGGGCGG 2316
    258 CCCGAGCC C CGCGCGGA 1248 UCCGCGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGCUCGGG 2317
    260 CGAGCCCC C CCCGGAGC 1249 GCUCCGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCGGCUCG 2318
    262 AGCCGCGC G CGGAGCUG 1250 CAGCUCCG CGAGCAAACUCC CU UCAACCACAUCGUCCCGG CCGCGGCU 2319
    264 CCGCGCGC C CAGCUCCC 2087 GCCACCUC CCACGAAACUCC CU UCAACGACAUCGUCCCCG CCGCCCCG 2320
    265 CGCGCCCG C AGCUCCCG 2088 CGCCAGCU GGAGCAAACUCC CU UCAAGCACAUCGUCCGGC CCCGCCCC 2321
    267 CCCCCCGA G CUGCCCGC 1251 CCCGGCAG GCACGAAACUCC CU UCAACGACAUCGUCCGGG UCCCCCCG 2322
    270 GCCCACCU G CCGGGGGC 1252 CCCCCCCC GCACGAAACUCC CU UCAAGGACAUCCUCCCGG AGCUCCCC 2323
    273 CACCUGCC C CGGGCUCC 2089 CGAGCCCC GCACCAAACUCC CU UCAAGCACAUCCUCCGGC GCCAGCUC 2324
    274 AGCUGCCG G GCGCUCCU 2090 AGCACCCC GCAGGAAACUCC CU UCAAGCACAUCGUCCGGG CCCCACCU 2325
    275 CCUGCCGC C CCCUCCUU 2091 AAGCACCC CGACGAAACUCC CU UCAAGGACAUCCUCCGGG CCCGCAGC 2326
    276 CUGCCCGG C GCUCCUUA 2092 UAACGACC GGAGCAAACUCC CU UCAACGACAUCGUCCCGC CCCGGCAG 2327
    277 UGCCGGCC C CUCCUUAG 1253 CUAACGAC GCAGCAAACUCC CU UCAAGCACAUCGUCCGGG CCCCCCCA 2328
    285 GCUCCUUA G CACCCGCG 1254 CCCCCCUC CGAGGAAACUCC CU UCAACCACAUCGUCCGGG UAACGAGC 2329
    291 UAGCACCC G GCCGCCGC 2093 CCGGCCCC CCACCAAACUCC CU UCAACGACAUCCUCCCGG GCGUGCUA 2330
    292 ACCACCCG C CCGCCCCG 2094 CCCGGCCC GCACCAAACUCC CU UCAAGCACAUCCUCCGGC CCGCUGCU 2331
    293 GCACCCGC C CGCCCGCG 1255 CCCCCGCG CGACGAAACUCC CU UCAAGGACAUCGUCCGCG CCGGGUGC 2332
    295 ACCCCCCC C CCCCCGCC 1256 CGCCCCCC CCACCAAACUCC CU UCAACCACAUCCUCCCCC CCCCCCCU 2333
    298 CCCCCCCC C CGCCCCUC 2095 CACCCCCC CCAGCAAACUCC CU UCAACCACAUCCUCCCCC CCCGCCCC 2334
    299 CCCCCCCC C CCCCCUCC 2096 CCACCCCC CGACCAAACUCC CU UCAACCACAUCGUCCGCC CGCCCCCC 2335
    300 CCCCCCCG C CCCCUCCC 2097 CCCACCCC CCACGAAACUCC CU UCAACCACAUCCUCCCCC CCCCCCCC 2336
    301 CCCCCCCC C CCCUCCCC 1257 CCCCACCC CCACCAAACUCC CU UCAACGACAUCCUCCCCC CCCCCCCC 2337
    307 CCCCCCUC C CCCUUCCG 1258 CCCAACCC CCACGAAACUCC CU UCAACGACAUCCUCCCCC CACCCCCC 2338
    315 GCCCUUCC C CACCCUUC 1259 CAAGCCUC CCACCAAACUCC CU UCAACCACAUCCUCCCGC CCAACCGC 2339
    318 CUUCCCCA C CCUUCACU 1260 ACUCAACG CCACCAAACUCC CU UCAACCACAUCCUCCCCC UCCCCAAC 2340
    330 UCACUCCA C CCCUCUCC 1261 CCACACCG GCACCAAACUCC CU UCAAGCACAUCCUCCGCG UCCACUCA 2341
    337 AGCCCUCU G CUCCCGCA 1262 UGCGGGAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGAGGGCU 2342
    343 CUGCUCCC G CACGCCAU 1263 AUGGCGUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGGAGCAG 2343
    347 UCCCGCAC G CCAUGAAG 1264 CUUCAUGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GUGCGGGA 2344
    352 CACGCCAU G AAGUCGCC 2098 GGCGACUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUGGCGUG 2345
    355 GCCAUGAA G UCGCCGUU 1265 AACGGCGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUCAUGGC 2346
    358 AUGAAGUC G CCGUUCUA 1266 UAGAACGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GACUUCAU 2347
    361 AAGUCGCC U UUCUACCG 1267 CGGUAGAA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGCGACUU 2348
    369 GUUCUACC U CUGCCAUA 1268 UCUGGCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGUG GGUAGAAC 2349
    372 CUACCGCU U CCAGAACA 1269 UGUUCUGG GUAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCGGUAG 2350
    376 CGCUGCCA G AACACCAC 2099 GUGGUGUU GUAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGUCAGCG 2351
    389 CCACCUCU G UGGAAAAA 1270 UUUUUCCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGAGUUGG 2352
    391 ACCUCUUU G GAAAAAGG 2100 CCUUUUUC GGAGUAAACUCC CU UCAAGGACAUCGUCCGUG ACAGAGGU 2353
    392 CCUCUGUG U AAAAAGUC 2101 GCCUUUUU GGAUUAAACUCC CU UCAAGGACAUCUUCCGUU CACAGAGG 2354
    398 UGGAAAAA U UCAACUCU 2102 CGAGUUGC GGAGGAAACUCC CU UCAAGUACAUCGUCCGGU UUUUUCCA 2355
    399 GUAAAAAU G CAACUCGU 1271 CCGAUUUG GGAGUAAACUCC CU UCAAGUACAUCGUCCGGG CUUUUUCC 2356
    406 UUCAACUC U GCGUUGAU 2103 AUCACCUC UGAGUAAACUCC CU UCAAGGACAUCGUCCUUG GAGUUGCC 2357
    407 GCAACUCU U CUGUGAUG 1272 CAUCACCU UGAGGAAACUCC CU UCAAGGACAUCGUCCGUU CGAUUUGC 2358
    409 AACUCGGC G UUGAUUGU 2104 CCCAUCAC GGAGGAAACUCC CU UCAAUGACAUCGUCCUUG GCCUAUUU 2359
    410 ACUCGUCG G UGAUGGUC 1273 UCCCAUCA UGAGGAAACUCC CU UCAAGGACAUCGUCCGUU CUCCUAGU 2360
    412 UCGUCGGU U AUGGUCGG 2105 CCGCCCAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGUG ACCGCCGA 2361
    415 GCGGUUAU U UUCGGGGU 2106 ACCCCGCC GGAUGAAACUCC CU UCAAUGACAUCGUCCGGG AUCACCGC 2362
    416 CGGUUAUG U UCUGUGUG 2107 CACCCCGC UGAUGAAACUCC CU UCAAGGACAUCGUCCGGG CAUCACCG 2363
    417 GGUUAUUG U CGUGUUGC 1274 GCACCCCG GGAGGAAACUCC CU UCAAUGACAUCGUCCGGG CCAUCACC 2364
    419 UUAUUGGC U GGUUGCUC 2108 GAGCACCC UGAUGAAACUCC CU UCAAGGACAUCGUCCGGG GCCCAUCA 2365
    420 GAUGGGCG U GGUUCUCU 2109 AGAGCACC GGAGGAAACUCC CU UCAAUGACAUCGUCCGGG CGCCCAUC 2366
    421 AUGGUCUG U GUGCUCUU 2110 AAGAGCAC GGAGGAAACUCC CU UCAAUGACAUCGUCCGGG CCGCCCAU 2367
    422 UGGGCGUG U UUCUCUUC 1275 GAAGAUCA GUAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCCGCCCA 2368
    424 GGCGGGGU U CUCUUCAG 1276 CUGAAGAG GGAGGAAACUCC CU UCAAGGACAUCUUCCUGU ACCCCGCC 2369
    432 GCUCUUCA G CACCGGCC 1277 GUCCUGUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGAAGAGC 2370
    437 UCAGCACC U UCCUCCUG 2111 CAGGAGGC GUAGGAAACUCC CU UCAAGGACAUCUUCCGGG GUUGCUGA 2371
    438 CAUCACCG U CCUCCUGG 1278 CCAGUAGG GGAGGAAACUCC CU UCAAGUACAUCGUCCGGG CGGUGCUU 2372
    445 GUCCUCCU U GGCAACCU 2112 AGGUUGCC GGAGGAAACUCC CU UCAAGUACAUCGUCCGGU AUGAGUCC 2373
    446 GCCUCCUG U GCAACCUU 2113 CAUGUUUC UUAUUAAACUCC CU UCAAGUACAUCUUCCUUU CAGUAUUC 2374
    447 CCUCCUGG G CAACCUGC 1279 GCAGGUUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCAGGAGG 2375
    454 GGCAACCU G CUGGCCCU 1280 AGGGCCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGUUGCC 2376
    457 AACCUGCU G GCCCUGGG 2114 CCCAGGGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCAGGUU 2377
    458 ACCUGCUG G CCCUGGGG 1281 CCCCAGGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGCAGGU 2378
    463 CUGGCCCU G GGGCUGCU 2115 AGCAGCCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGGCCAG 2379
    464 UGGCCCUG U GGCUGCUG 2116 CAGCAGCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGGGCCA 2380
    465 GGCCCUGG G GCUGCUGG 2117 CCAGCAGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCAGGGCC 2381
    466 GCCCUGGG G CUGCUGGC 1282 UCCAUCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCCAGGGC 2382
    469 CUGGGGCU G CUGGCGCG 1283 CGCGCCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCCCCAG 2383
    472 GGGCUGCU G GCGCGCUC 2118 GAGCGCUC GGAGGAAACUCC CU UCAAGUACAUCGUCCGUG AGCAGCCC 2384
    473 GGCUGCUG U CGCGCUCG 1284 CGAGCGCG GGAGGAAACUCC CU UCAAGUACAUCGUCCGUG CAGCAGCC 2385
    475 CUGCUGUC U CGCUCUGU 1285 CCCGAGCG GGAGGAAACUCC CU UCAAUGACAUCGUCCGUG GCCAGCAG 2386
    477 GCUGGCGC U CUCGGGGC 1286 GCCCCGAU GGAUGAAACUCC CU UCAAUGACAUCGUCCGGG UCUCCAUC 2387
    481 GCGCGCUC G GUGCUGUU 2119 CCCAGCCC GUAUGAAACUCC CU UCAAUUACAUCGUCCGGG GAUCUCGC 2388
    482 CGCGCUCG G GGCUGGGG 2120 CCCCAGCC GUAUGAAACUCC CU UCAAGUACAUCGUCCGGG CUAUCGCG 2389
    483 GCGCUCGG G GCUGGGGU 2121 ACCCCAGC GUAUGAAACUCC CU UCAAGGACAUCGUCCGGG CCUAUCGC 2390
    484 CGCUCGGG G CUGGGGUG 1287 CACCCCAG GUAUUAAACUCC CU UCAAUUACAUCGUCCUGU CCCGAGCG 2391
    487 UCGGGGCU G GGGUGUUG 2122 CACCACCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCCCCGA 2392
    488 CUGGUCUG U GGUGGUGC 2123 GCACCACC GGAUUAAACUCC CU UCAAUUACAUCGUCCUGU CAGCCCCG 2393
    489 GUUGCUUU U GUGGUGCU 2124 AGCACCAC GGAGUAAACUCC CU UCAAUUACAUCGUCCUGG CCAGCCCC 2394
    490 UGGCUGUG U UGGUGCUC 1288 UAGCACCA UGAGUAAACUCC CU UCAAGUACAUCGUCCGGG CCCAGCCC 2395
    492 GCUGUGGU U UUGCUCGC 2125 UCGAGCAC GGAGUAAACUCC CU UCAAGGACAUCGUCCGUG ACCCCAGC 2396
    493 CUGGGGUG U UGCUCGCU 1289 CGCGAGCA UGAGGAAACUCC CU UCAAUUACAUCUUCCUUU CACCCCAU 2397
    495 UGGGUUGU G CUCGCGGC 1290 GCCGCGAU UGAGGAAACUCC CU UCAAUUACAUCGUCCUUU ACCACCCC 2398
    499 UGGUCCUC U CGUCGUCC 1291 GGACUCCG GGAGUAAACUCC CU UCAAUGACAUCGUCCUGG GAUCACCA 2399
    501 UUGCUCUC U UCUUCCAC 2126 GUUGACGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCUAGCAC 2400
    502 UGCUCGCG U CGUCCACU 1292 AGUGUACG GGAGGAAACUCC CU UCAAUGACAUCGUCCUGG CGCGAGCA 2401
    504 CUCGCGGC U UCCACUGC 1293 GCAGUUGA UGAGGAAACUCC CU UCAAGGACAUCGUCCGUU GCCGCGAG 2402
    511 CGUCCACU G CGCCCGCU 1294 AUCGUUCU GUAUGAAACUCC CU UCAAGGACAUCUUCCGUG AGUGUACG 2403
    513 UCCACUGC U CCCGCUGC 1295 GCAGCGGU GGAGGAAACUCC CU UCAAUGACAUCGUCCGGG UCAGUGGA 2404
    517 CUGCUCCC U CUUCCCUC 1296 GAUGUCAG GGAGUAAACUCC CU UCAAGGACAUCUUCCGUG UGUCUCAG 2405
    520 CGCCCGCU U CCCUCUGU 1297 ACCUAGUG GGAGGAAACUCC CU UCAAUGACAUCUUCCUGU AGCGUGCU 2406
    526 CUGCCCUC U GUCUUCUA 2127 UAGAAGAC GGAGUAAACUCC CU UCAAGGACAUCUUCCGGU GAUGUCAG 2407
    527 UGCCCUCG G UCUUCUAC 1298 GUAGAAGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGAGGGCA 2408
    538 UUCUACAU G CUGGUGUG 1299 CACACCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUGUAGAA 2409
    541 UACAUGCU G GUGUGUGG 2128 CCACACAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCAUGUA 2410
    542 ACAUGCUG G UGUGUGGC 1300 GCCACACA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGCAUGU 2411
    544 AUGCUGGU G UGUGGCCU 1301 AGOCCACA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCAGCAU 2412
    546 GCUGGUGU G UGGCCUGA 1302 UCAGGCCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACACCAGC 2413
    548 UGGUGUGU G GCCUGACG 2129 CGUCAGGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACACACCA 2414
    549 GGUGUGUG G CCUGACGG 1303 CCGUCAGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CACACACC 2415
    553 UGUGGCCU G ACGGUCAC 2130 GUGACCGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGCCACA 2416
    556 GGCCUGAC G GUCACCGA 2131 UCGGUGAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GUCAGGCC 2417
    557 GCCUGACG G UCACCGAC 1304 GUCGGUGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGUCAGGC 2418
    563 CGGUCACC G ACUUGCUG 2132 CAGCAAGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGUGACCG 2419
    568 ACCGACUU G CUGGGCAA 1305 UUGCCCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AAGUCGGU 2420
    571 GACUUGCU G GGCAAGUG 2133 CACUUGCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCAAGUC 2421
    572 ACUUGCUG G GCAAGUGC 2134 GCACUUGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGCAAGU 2422
    573 CUUGCUGG G CAAGUGCC 1306 GGCACUUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCAGCAAG 2423
    577 CUGGGCAA G UGCCUCCU 1307 AGGAGGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUGCCCAG 2424
    579 GGGCAAGU G CCUCCUAA 1308 UUAGGAGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACUUGCCC 2425
    588 CCUCCUAA G CCCGGUGG 1309 CCACCGGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUAGGAGG 2426
    592 CUAAGCCC G GUGGUGCU 2135 AGCACCAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGGCUUAG 2427
    593 UAAGCCCG G UGGUGCUG 1310 CAGCACCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGGGCUUA 2428
    595 AGCCCGGU G GUGCUGGC 2136 GCCAGCAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCGGGCU 2429
    596 GCCCGGUG G UGCUGGCU 1311 AGCCAGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CACCGGGC 2430
    598 CCGGUGGU G CUGGCUGC 1312 GCAGCCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCACCGG 2431
    601 GUGGUGCU G GCUGCCUA 2137 UAGGCAGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCACCAC 2432
    602 UGGUGCUG G CUGCCUAC 1313 GUAGGCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGCACCA 2433
    605 UGCUGGCU G CCUACGCU 1314 AGCGUAGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCCAGCA 2434
    611 CUGCCUAC G CUCAGAAC 1315 GUUCUGAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GUAGGOAG 2435
    616 UACGCUCA G AACCGGAG 2138 CUCCGGUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGAGCGUA 2436
    621 UCAGAACC G GAGUCUGC 2139 GCAGACUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGUUCUGA 2437
    622 CAGAACCG G AGUCUGCG 2140 CGCAGACU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGGUUCUG 2438
    624 GAACCGGA G UCUGCGGG 1316 CCCGCAGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCCGGUUC 2439
    628 CGGAGUCU G CGGGUGCU 1317 AGCACCCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGACUCCG 2440
    630 GAGUCUGC G GGUGCUUG 2141 CAAGCACC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCAGACUC 2441
    631 AGUCUGCG G GUGCUUGC 2142 GCAAGCAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGCAGACU 2442
    632 GUCUGCGG G UGCUUGCG 1318 CGCAAGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCGCAGAC 2443
    634 CUGCGGGU G CUUGCGCC 1319 GGCGCAAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCCGCAG 2444
    638 GGGUGCUU C CGCCCGCA 1320 UGCGGGCG GGAGGAAACUCC CU UCAAGGACAUCCUCCGGG AAGCACCC 2445
    640 GUGCUUGC G CCCGCAUU 1321 AAUGCGGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCAAGCAC 2446
    644 UUGCGCCC G CAUUGGAC 1322 GUCCAAUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGGCGCAA 2447
    649 CCCGCAUU G GACAACUC 2143 GAGUUGUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AAUGCGGG 2448
    650 CCGCAUUG C ACAACUCG 2144 CGAGUUGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAAUGCGC 2449
    658 CACAACUC G UUGUGCCA 1323 UGGCACAA GGACGAAACUCC CU UCAACGACAUCGUCCCGG GACUUGUC 2450
    661 AACUCGUU G UGCCAAGC 1324 GCUUGGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGCG AACCACUU 2451
    663 CUCGUUGU G CCAACCCU 1325 ACGCUUGG CGAGGAAACUCC CU UCAAGGACAUCGUCCCGC ACAACGAC 2452
    668 UGUGCCAA G CCUUCCCC 1326 CCCGAAGC CGACGAAACUCC CU UCAACGACAUCGUCCGGC UUGGCACA 2453
    674 AACCCUUC C CCUUCUUC 1327 GAAGAAGG CGACGAAACUCC CU UCAAGGACAUCCUCCGCG CAACCCUU 2454
    685 UUCUUCAU G UCCUUCUU 1328 AACAACGA GGACGAAACUCC CU UCAAGGACAUCGUCCGGG AUGAAGAA 2455
    695 CCUUCUUU G GGCUCUCC 2145 CCAGACCC GGACGAAACUCC CU UCAACGACAUCGUCCGGG AAAGAACC 2456
    696 CUUCUUUG G GCUCUCCU 2146 ACCACAGC GGACGAAACUCC CU UCAACGACAUCCUCCCCG CAAACAAG 2457
    697 UUCUUUGG G CUCUCCUC 1329 GAGGACAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGCG CCAAAGAA 2458
    706 CUCUCCUC G ACACUGCA 2147 UCCAGUGU GGAGGAAACUCC CU UCAACGACAUCGUCCGGG GAGGAGAG 2459
    712 UCGACACU C CAACUCCU 1330 ACCACUUC CCAGGAAACUCC CU UCAAGGACAUCGUCCGCG AGUGUCGA 2460
    721 CAACUCCU C CCCAUGCC 2148 CCCAUCGC GCACGAAACUCC CU UCAACCACAUCGUCCGCG ACCACUUC 2461
    722 AACUCCUC G CCAUGGCA 1331 UGCCAUGG CGACGAAACUCC CU UCAAGGACAUCGUCCCGG CACGAGUU 2462
    727 CUGGCCAU C GCACUCGA 2149 UCCACUGC GGAGGAAACUCC CU UCAAGGACAUCGUCCCCC AUGGCCAG 2463
    728 UGCCCAUC C CACUGGAC 1332 CUCCACUC GGACGAAACUCC CU UCAAGCACAUCGUCCGCG CAUCCCCA 2464
    733 AUCCCACU C CACUGCUC 2150 CACCACUC GGACGAAACUCC CU UCAAGCACAUCGUCCGGG AGUGCCAU 2465
    734 UCCCACUC C ACUGCUGG 2151 CCAGCACU CCACCAAACUCC CU UCAACCACAUCCUCCCGC CACUGCCA 2466
    736 CCACUCGA C UCCUCCCU 1333 AGCCAGCA GCAGGAAACUCC CU UCAACCACAUCCUCCCCC UCCACUCC 2467
    738 ACUCCAGU C CUGCCUCU 1334 ACACCCAC CCACCAAACUCC CU UCAACCACAUCCUCCCGC ACUCCACU 2468
    741 CCACUCCU C CCUCUCCC 2152 GGGAGAGC GCAGGAAACUCC CU UCAACCACAUCCUCCCGC ACCACUCC 2469
    742 CACUCCUC C CUCUCCCU 1335 ACGCAGAC GCACGAAACUCC CU UCAACCACAUCCUCCCCC CACCACUC 2470
    752 UCUCCCUA C CCCACCCU 2153 ACCCUCCC CCACCAAACUCC CU UCAACCACAUCCUCCCCC UACCCACA 2471
    753 CUCCCUAC C GCACCCUU 2154 AAGGGUGC GCAGGAAACUCC CU UCAACCACAUCCUCCGGC CUAGGCAG 2472
    754 UCCCUAGC C CACCCUUU 1336 AAACGCUC CCACGAAACUCC CU UCAACCACAUCCUCCCCC CCUAGCGA 2473
    771 CUUCUACC G ACGGCACA 2155 UGUGCCGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGUAGAAG 2474
    774 CUACCGAC G GCACAUCA 2156 UGAUGUGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GUCGGUAG 2475
    775 UACCGACG G CACAUCAC 1337 GUGAUGUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGUCGGUA 2476
    787 AUCACCCU G CGCCUGGG 1338 CCCAGGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGGUGAU 2477
    789 CACCCUGC G CCUGGGCG 1339 CGCCCAGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCAGGGUG 2478
    793 CUGCGCCU G GGCGCACU 2157 AGUGCGCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGCGCAG 2479
    794 UGCGCCUG G GCGCACUG 2158 CAGUGCGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGGCGCA 2480
    795 GCGCCUGG G CGCACUGG 1340 CCAGUGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCAGGCGC 2481
    797 GCCUGGGC G CACUGGUG 1341 CACCAGUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCCCAGGC 2482
    802 GGCGCACU G GUGGCCCC 2159 GGGGCCAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGUGCGCC 2483
    803 GCGCACUG G UGGCCCCG 1342 CGGGGCCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGUGCGC 2484
    805 GCACUGGU G GCCCCGGU 2160 ACCGGGGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCAGUGC 2485
    806 CACUGGUG G CCCCGGUG 1343 CACCGGGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CACCAGUG 2486
    811 GUGGCCCC G GUGGUGAG 2161 CUCACCAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGGGCCAC 2487
    812 UGGCCCCG G UGGUGAGC 1344 UCUCACCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGGGGCCA 2488
    814 GCCCCGGU G GUGAGCGC 2162 GCGCUCAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCGGGGC 2489
    815 CCCCGGUG G UGAGCGCC 1345 GGCGCUCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CACCGGGG 2490
    817 CCGGUGGU G AGCGCCUU 2163 AAGGCGCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCACCGG 2491
    819 GGUGGUGA G CGCCUUCU 1346 AGAAGGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCACCACC 2492
    821 UGGUGAGC G CCUUCUCC 1347 GGAGAAGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCUCACCA 2493
    832 UUCUCCCU G GCUUUCUG 2164 CAGAAAGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGGAGAA 2494
    833 UCUCCCUG G CUUUCUGC 1348 GCAGAAAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGGGAGA 2495
    840 GGCUUUCU G CGCGCUAC 1349 GUAGCGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGAAAGCC 2496
    842 CUUUCUGC G CGCUACCU 1350 AGGUAGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCAGAAAG 2497
    844 UUCUGCGC G CUACCUUU 1351 AAAGGUAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCGCAGAA 2498
    856 CCUUUCAU G GGCUUCGG 2165 CCGAAGCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUGAAAGG 2499
    857 CUUUCAUG G GCUUCGGG 2166 CCCGAAGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAUGAAAG 2500
    858 UUUCAUGG G CUUCGGGA 1352 UCCCGAAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCAUGAAA 2501
    863 UGGGCUUC G GGAAGUUC 2167 GAACUUCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GAAGCCCA 2502
    864 GGGCUUCG G GAAGUUCG 2168 CGAACUUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGAAGCCC 2503
    865 GGCUUCGG G AAGUUCGU 2169 ACGAACUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCGAAGCC 2504
    868 UUCGGGAA G UUCGUGCA 1353 UGCACGAA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUCCCGAA 2505
    872 GGAAGUUC G UGCAGUAC 1354 GUACUGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GAACUUCC 2506
    874 AAGUUCGU G CAGUACUG 1355 CAGUACUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACGAACUU 2507
    877 UUCGUGCA G UACUGCCC 1356 GGGCAGUA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGCACGAA 2508
    882 GCAGUACU G CCCCGGCA 1357 UGCCGGGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGUACUGC 2509
    887 ACUGCCCC G GCACCUGG 2170 CCAGGUGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGGGCAGU 2510
    888 CUGCCCCG G CACCUGGU 1358 ACCAGGUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGGGGCAG 2511
    894 CGGCACCU G GUGCUUUA 2171 UAAAGCAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGGUGCCG 2512
    895 GGCACCUG G UGCUUUAU 1359 AUAAAGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGGUGCC 2513
    897 CACCUGGU G CUUUAUCC 1360 GGAUAAAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCAGGUG 2514
    907 UUUAUCCA G AUGGUCCA 2172 UGGACCAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGGAUAAA 2515
    910 AUCCAGAU G GUCCACGA 2173 UCGUGGAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUCUGGAU 2516
    911 UCCAGAUG G UCCACGAG 1361 CUCGUGGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAUCUGGA 2517
    917 UGGUCCAC G AGGAGGGC 2174 GCCCUCCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GUGGACCA 2518
    919 GUCCACGA G GAGGGCUC 2175 GAGCCCUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCGUGGAC 2519
    920 UCCACGAG G AGGGCUCG 2176 CGAGCCCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUCGUGGA 2520
    922 CACGAGGA G GGCUCGCU 2177 AGCGAGCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCCUCGUG 2521
    923 ACGAGGAG G GCUCGCUG 2178 CAGCGAGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUCCUCGU 2522
    924 CGAGGAGG G CUCGCUGU 1362 ACAGCGAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCUCCUCG 2523
    928 GAGGGCUC G CUGUCGGU 1363 ACCGACAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GAGCCCUC 2524
    931 GGCUCGCU G UCGGUGCU 1364 AGCACCGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCGAGCC 2525
    934 UCGCUGUC G GUGCUGGG 2179 CCCAGCAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GACAGCGA 2526
    935 CGCUGUCG G UGCUGGGG 1365 CCCCAGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGACAGCG 2527
    937 CUGUCGGU G CUGGGGUA 1366 UACCCCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCGACAG 2528
    940 UCGGUGCU G GGGUACUC 2180 GAGUACCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCACCGA 2529
    941 CGGUGCUG G GGUACUCU 2181 AGAGUACC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGCACCG 2530
    942 GGUGCUGG G GUACUCUG 2182 CAGAGUAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCAGCACC 2531
    943 GUGCUGGG G UACUCUGU 1367 ACAGAGUA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CCCAGCAC 2532
    950 GGUACUCU G UGCUCUAC 1368 GUAGAGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGAGUACC 2533
    952 UACUCUGU G CUCUACUC 1369 GAGUAGAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACAGAGUA 2534
    963 CUACUCCA G CCUCAUGG 1370 CCAUGAGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGGAGUAG 2535
    970 AGCCUCAU G GCGCUGCU 2183 AGCAGCGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUGAGGCU 2536
    971 GCCUCAUG G CGCUGCUG 1371 CAGCAGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAUGAGGC 2537
    973 CUCAUGGC G CUGCUGGU 1372 ACCAGCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCCAUGAG 2538
    976 AUGGCGCU G CUGGUCCU 1373 AGGACCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCGCCAU 2539
    979 GCGCUGCU G GUCCUCGC 2184 GCGAGGAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCAGCGC 2540
    980 CGCUGCUG G UCCUCGCC 1374 GGCGAGGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAGCAGCG 2541
    986 UGGUCCUC G CCACCGUG 1375 CACGGUGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GAGGACCA 2542
    992 UCGCCACC G UGCUGUGC 1376 GCACAGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGUGGCGA 2543
    994 GCCACCGU U CUGUGCAA 1377 UUGCACAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACGGUGGC 2544
    997 ACCGUGCU G UGCAACCU 1378 AGGUUGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCACGGU 2545
    999 CGUGCUGU G CAACCUCG 1379 CGAGGUUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACAGCACG 2546
    1007 GCAACCUC U GCGCCAUG 2185 CAUGGCGC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GAGGUUGC 2547
    1008 CAACCUCG G CGCCAUGC 1380 GCAUGGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGAGGUUG 2548
    1010 ACCUCGGC G CCAUGCGC 1381 GCGCAUGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCCGAGGU 2549
    1015 GGCGCCAU G CGCAACCU 1382 AGGUUUCU GGAGGAAACUCC CU UCAAUGACAUCGUCCGGG AUGGCGCC 2550
    1017 CGCCAUUC G CAACCUCU 1383 AUAGGUUG GGAGGAAACUCC CU UCAAGGACAUCUUCCGGG GCAUGGCG 2551
    1028 ACCUCUAU G CGAUGCAC 1384 GUGCAUCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUAGAGGU 2552
    1030 CUCUAUGC U AUGCACCG 2186 CGGUGCAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCAUAGAG 2553
    1033 UAUGCGAU G CACCGGCG 1385 CGCCGGUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUCGCAUA 2554
    1038 UAUGCACC G GCUGCUGC 2187 UCAGCCGC GUAGGAAACUCC CU UCAAGGACAUCGUCCGUG GGUGCAUC 2555
    1039 AUGCACCG G CGUCUGCA 1386 UGCAGCCG GGAGUAAACUCC CU UCAAGGACAUCGUCCGUG CGGUGCAU 2556
    1041 GCACCGGC U UCUGCAUC 2188 GCUGCAGC GGAGGAAACUCC CU UCAAUGACAUCGUCCGGG GCCGGUUC 2557
    1042 CACCGUCG U CUUCAUCG 1387 CGCUGCAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGCCGGUG 2558
    1045 CGGCGUCU G CAUCUGCA 1388 UUCCGCUG GGAGGAAACUCC CU UCAAGUACAUCGUCCGGG AGCCGCCU 2559
    1048 CUUCUGCA G CGGCACCC 1389 GGGUGCCG GGAGGAAACUCC CU UCAAGGACAUCGUCCUGG UGCAGCCG 2560
    1050 GCUUCAGC U UCACCCGC 2189 GCGUGUGC UUAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCUGCAGC 2561
    1051 CUGCAGCG G CACCCGCU 1390 CGCUGGUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGU CGCUGCAG 2562
    1057 CGUCACCC U CGCUCCUG 1391 CAGGAGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGGUGCCG 2563
    1059 GCACCCUC U CUCCUGCA 1392 UGCAGGAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCGGGUGC 2564
    1065 GCGCUCCU G CACCAGGG 1393 CCCUUGUG GGAUGAAACUCC CU UCAAGGACAUCGUCCGGG AGGAGCGC 2565
    1071 CUGCACCA G GUACUGUG 2190 CACAGUCC UGAGUAAACUCC CU UCAAGUACAUCGUCCGGG UGGUGCAG 2566
    1072 UGCACCAG U GACUGUGC 2191 GCACAUUC GGAGUAAACUCC CU UCAAGUACAUCGUCCGGG CUGGUGCA 2567
    1073 GCACCAGG U ACUGUGCC 2192 GUCACAGU GGAGGAAACUCC CU UCAAGUACAUCGUCCGGG CCUGGUGC 2568
    1077 CAGGGACU U UGCCGAGC 1394 GCUCGGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGUCCCUG 2569
    1079 GGGACUGU U CCGAGCCG 1395 CUGCUCGG UGAGGAAACUCC CU UCAAGGACAUCGUCCGGU ACAGUCCC 2570
    1082 ACUUUUCC U AUCCUCUC 2193 GCUCGUCU UUAUGAAACUCC CU UCAAUGACAUCUUCCUGU UUCACAGU 2571
    1084 UUUUCCUA U CCUCUCGC 1396 UCUCUCUG UUAUGAAACUCC CU UCAAUGACAUCUUCCUUU UCGUCACA 2572
    1087 GCCGAGCC G CGCGCGGA 1397 UCCGCGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGCUCGGC 2573
    1089 CGAGCCGC G CGCGGACG 1398 CGUCCGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCGGCUCG 2574
    1091 AGCCGCGC G CGGACGGG 1399 CCCGUCCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCGCGGCU 2575
    1093 CCGCGCGC G GACGGGAG 2194 CUCCCGUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCGCGCGG 2576
    1094 CGCGCGCG G ACGGGAGG 2195 CCUCCCGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGCGCGCG 2577
    1097 GCGCGGAC G GGAGGGAA 2196 UUCCCUCC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GUCCGCGC 2578
    1098 CGCGGACG C GAGGGAAG 2197 CUUCCCUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGUCCGCG 2579
    1099 GCGGACGG C AGGGAAGC 2198 CCUUCCCU GCACGAAACUCC CU UCAAGCACAUCCUCCGGG CCCUCCGC 2580
    1101 CGACCCGA C CCAAGCCU 2199 ACCCUUCC GCAGGAAACUCC CU UCAAGGACAUCGUCCCCG UCCCCUCC 2581
    1102 CACGGCAC G GAAGCGUC 2200 GACGCUUC GGAGGAAACUCC CU UCAAGGACAUCGUCCCCG CUCCCGUC 2582
    1103 ACCGCAGC C AACCGUCC 2201 GGACCCUU GCACGAAACUCC CU UCAAGCACAUCCUCCCCG CCUCCCGU 2583
    1106 GGAGGCAA G CCUCCCCU 1400 AGCGCACG GGAGGAAACUCC CU UCAAGGACAUCGUCCCGG UUCCCUCC 2584
    1108 AGGCAACC C UCCCCUCA 1401 UGAGGGCA GGAGGAAACUCC CU UCAACCACAUCGUCCCGG GCUUCCCU 2585
    1117 UCCCCUCA C CCCCUGGA 1402 UCCACGGG GGACGAAACUCC CU UCAAGGACAUCGUCCCGG UCACCGGA 2586
    1123 CAGCCCCU C CACGAGCU 2202 AGCUCCUC GGACGAAACUCC CU UCAAGGACAUCGUCCGGG ACGGGCUG 2587
    1124 ACCCCCUG C ACGAGCUG 2203 CAGCUCCU GGAGGAAACUCC CU UCAAGGACAUCGUCCCCG CAGGCGCU 2588
    1126 CCCCUCGA C GAGCUGGA 2204 UCCACCUC GCACGAAACUCC CU UCAAGGACAUCGUCCGCG UCCACGCG 2589
    1127 CCCUCCAC C AGCUGGAU 2205 AUCCAGCU GCAGGAAACUCC CU UCAACGACAUCCUCCGCG CUCCAGGG 2590
    1129 CUGGAGGA C CUGGAUCA 1403 UCAUCCAG GCACGAAACUCC CU UCAAGCACAUCGUCCGCG UCCUCCAG 2591
    1132 GAGGAGCU C CAUCACCU 2206 ACCUGAUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCUCCUC 2592
    1133 AGCACCUC C AUCACCUC 2207 CACGUGAU GCACGAAACUCC CU UCAAGCACAUCCUCCGGG CACCUCCU 2593
    1144 CACCUCCU C CUCCUGCC 1404 CCCACCAG GCACGAAACUCC CU UCAAGCACAUCCUCCGGG ACCAGGUG 2594
    1147 CUCCUGCU C CUGGCGCU 1405 AGCGCCAG GGACGAAACUCC CU UCAAGGACAUCCUCCGGG ACCACGAG 2595
    1150 CUCCUCCU C GCGCUGAU 2208 AUCACCCC GCAGGAAACUCC CU UCAAGCACAUCCUCCGCG ACCACCAG 2596
    1151 UCCUCCUC C CGCUCAUG 1406 CAUCAGCG GCACGAAACUCC CU UCAAGGACAUCGUCCGGG CACCAGCA 2597
    1153 CUCCUGGC C CUGAUGAC 1407 GUCAUCAG GCAGGAAACUCC CU UCAAGCACAUCCUCCGCG GCCAGCAG 2598
    1156 CUCCCCCU C AUCACCCU 2209 ACCGUCAU GCAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCGCCAG 2599
    1159 CCCCUCAU C ACCCUCCU 2210 AGCACGCU GCACCAAACUCC CU UCAAGCACAUCGUCCGCG AUCACCCC 2600
    1163 UGAUCACC C UGCUCUUC 1408 CAAGAGCA GCAGGAAACUCC CU UCAAGCACAUCCUCCGGG GCUCAUCA 2601
    1165 AUGACCGU C CUCCUCAC 1409 GUCAAGAG GGAGCAAACUCC CU UCAAGCACAUCCUCCGCG ACGGUCAU 2602
    1177 UUCACUAU C UGUUCUCU 1410 AGACAACA CGAGCAAACUCC CU UCAAGCACAUCCUCCGGG AUACUCAA 2603
    1179 CACUAUGU C UUCUCUGC 1411 GCAGAGAA GGAGGAAACUCC CU UCAAGCACAUCCUCCGCG ACAUACUG 2604
    1186 UGUUCUCU C CCCGUAAU 1412 AUUACGGC CGAGGAAACUCC CU UCAAGCACAUCCUCCCCG AGAGAACA 2605
    1190 CUCUGCCC G UAAUUUAU 1413 AUAAAUUA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGGCAGAG 2606
    1200 AAUUUAUC G CGCUUACU 1414 AGUAAGCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GAUAAAUU 2607
    1202 UUUAUCGC G CUUACUAU 1415 AUAGUAAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCGAUAAA 2608
    1211 CUUACUAU G CAGCAUUU 2211 AAAUGCUC GGAGGAAACUCC CU UCAAOGACAUCGUCCGGG AUAGUAAG 2609
    1212 UUACUAUG G AGCAUUUA 2212 UAAAUGCU GGAGGAAACUCC CU UCAA0GACAUCGUCCG0G CAUAGUAA 2610
    1214 ACUAUGGA G CAUUUAAG 1416 CUUAAAUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCCAUAGU 2611
    1222 GCAUUUAA G GAUGUCAA 2213 UUGACAUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUAAAUGC 2612
    1223 CAUUUAAG G AUGUCAAG 2214 CUUGACAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUUAAAUG 2613
    1226 UUAAGGAU G UCAAGGAG 1417 CUCCUUGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUCCUUAA 2614
    1231 GAUGUCAA G GAGAAAAA 2215 UUUUUCUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUGACAUC 2615
    1232 AUGUCAAG G AGAAAAAC 2216 GUUUUUCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUUGACAU 2616
    1234 GUCAAGGA G AAAAACAG 2217 CUGUUUUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCCUUGAC 2617
    1242 GAAAAACA G GACCUCUG 2218 CAGAGGUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGUUUUUC 2618
    1243 AAAAACAG G ACCUCUGA 2219 UCAGAGGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUGUUUUU 2619
    1250 GGACCUCU G AAGAAGCA 2220 UGCUUCUU GGAGGAAACUCC CU UCAAGGACAUCGUCC0GG AGAGGUCC 2620
    1253 CCUCUGAA G AAGCAGAA 2221 UUCUGCUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUCAGAGG 2621
    1256 CUGAAGAA G CAGAAGAC 1418 GUCGUCUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUCUUCAG 2622
    1259 AAGAAGCA G AAGACCUC 2222 GAGGUCUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGCUUCUU 2623
    1262 AAGCAGAA G ACCUCCGA 2223 UCGGAGGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUCUGCUU 2624
    1269 AGACCUCC G AGCCUUGC 2224 GCAAGGCU GGAGGAAACUCC CU UCAAG0ACAUCGUCCGGG GGAGGUCU 2625
    1271 ACCUCCGA G CCUUGCGA 1419 UCGCAAGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCGGAGGU 2626
    1276 CGAGCCUU G CGAUUUCU 1420 AGAAAUCG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AAGGCUCG 2627
    1278 AGCCUUGC G AUUUCUAU 2225 AUAGAAAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GCAAGGCU 2628
    1289 UUCUAUCU G UGAUUUCA 1421 UGAAAUCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGAUAGAA 2629
    1291 CUAUCUGU G AUUUCAAU 2226 AUUGAAAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACAGAUAG 2630
    1301 UUUCAAUU G UGGACCCU 1422 AGGGUCCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AAUUGAAA 2631
    1303 UCAAUUGU G GACCCUUG 2227 CAAGGGUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACAAUUGA 2632
    1304 CAAUUGUG G ACCCUUGG 2228 CCAAGGGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CACAAUUG 2633
    1311 GGACCCUU G GAUUUUUA 2229 UAAAAAUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AAGGGUCC 2634
    1312 GACCCUUG G AUUUUUAU 2230 AUAAAAAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAAGGGUC 2635
    1329 CAUUUUCA G AUCUCCAG 2231 CUGGAGAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGAAAAUG 2636
    1337 GAUCUCCA G UAUUUCGG 1423 CCGAAAUA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGGAGAUC 2637
    1344 AGUAUUUC G GAUAUUUU 2232 AAAAUAUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GAAAUACU 2638
    1345 GUAUUUCG G AUAUUUUU 2233 AAAAAUAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGAAAUAC 2639
    1360 UUUCACAA G AUUUUCAU 2234 AUGAAAAU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUGUGAAA 2640
    1371 UUUCAUUA G ACCUCUUA 2235 UAAGAGGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UAAUGAAA 2641
    1380 ACCUCUUA G GUACAGGA 2236 UCCUGUAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UAAGAGGU 2642
    1381 CCUCUUAG G UACAGGAG 1424 CUCCUGUA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUAAGAGG 2643
    1386 UAGGUACA G GAGCCGGU 2237 ACCGGCUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGUACCUA 2644
    1387 AGGUACAG G AGCCGGUG 2238 CACCGGCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUGUACCU 2645
    1389 GUACAGGA G CCGGUGCA 1425 UGCACCGG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCCUGUAC 2646
    1392 CAGGAGCC G GUGCAGCA 2239 UGCUGCAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG GGCUCCUG 2647
    1393 AGGAGCCG G UGCAGCAA 1426 UUGCUGCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CGGCUCCU 2648
    1395 GAGCCGGU G CAGCAAUU 1427 AAUUGCUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACCGGCUC 2649
    1398 CCGGUGCA G CAAUUCCA 1428 UGGAAUUG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGCACCGG 2650
    1414 ACUAACAU G GAAUCCAG 2240 CUGGAUUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUGUUAGU 2651
    1415 CUAACAUG G AAUCCAGU 2241 ACUGGAUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CAUGUUAG 2652
    1422 GGAAUCCA G UCUGUGAC 1429 GUCACAGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGGAUUCC 2653
    1426 UCCAGUCU G UGACAGUG 1430 CACUGUCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGACUGGA 2654
    1428 CAGUCUGU G ACAGUGUU 2242 AACACUGU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACAGACUG 2655
    1432 CUGUGACA G UGUUUUUC 1431 GAAAAACA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGUCACAG 2656
    1434 GUGACAGU G UUUUUCAC 1432 GUGAAAAA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACUGUCAC 2657
    1446 UUCACUCU G UGGUAAGC 1433 GCUUACCA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGAGUGAA 2658
    1448 CACUCUGU G GUAAGCUG 2243 CAGCUUAC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG ACAGAGUG 2659
    1449 ACUCUGUG G UAAGCUGA 1434 UCAGCUUA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CACAGAGU 2660
    1453 UGUGGUAA G CUGAGGAA 1435 UUCCUCAG GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UUACCACA 2661
    1456 GGUAAGCU G AGGAAUAU 2244 AUAUUCCU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AGCUUACC 2662
    1458 UAAGCUGA G GAAUAUGU 2245 ACAUAUUC GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UCAGCUUA 2663
    1459 AAGCUGAG G AAUAUGUC 2246 GACAUAUU GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG CUCAGCUU 2664
    1465 AGGAAUAU G UCACAUUU 1436 AAAUGUGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG AUAUUCCU 2665
    1477 CAUUUUCA G UCAAAGAA 1437 UUCUUUGA GGAGGAAACUCC CU UCAAGGACAUCGUCCGGG UGAAAAUG 2666

Claims (20)

What we claim is:
1. A nucleic acid molecule that down regulates expression of a prostaglandin D2 receptor (PTGDR) gene.
2. The nucleic acid molecule of claim 1, wherein said nucleic acid molecule is an enzymatic nucleic acid molecule.
3. The nucleic acid molecule of claim 1, wherein said nucleic acid molecule is an antisense nucleic acid molecule.
4. The enzymatic nucleic acid molecule of claim 2, wherein said enzymatic nucleic acid molecule comprises a sequence selected from the group of sequences consisting of SEQ ID NOs: 228-454, 831-1206, 1438-1668, 1715-2057, and 2247-2666.
5. The enzymatic nucleic acid molecule of claim 2, wherein said enzymatic nucleic acid molecule comprises at least one binding arm wherein one or more of said binding arms comprises a sequence complementary to a sequence selected from the group of sequences consisting of SEQ ID NOs: 1-227, 455-830, 1207-1437, 1669-1714, and 2058-2246.
6. The antisense nucleic acid molecule of claim 3, wherein said antisense nucleic acid molecule comprises a sequence complementary to a sequence selected from the group of sequences consisting of SEQ ID NOs: 1-227, 455-830, 1207-1437, 1669-1714, and 2058-2246.
7. The nucleic acid molecule of claim 1, wherein said nucleic acid molecule is adapted to treat asthma.
8. The nucleic acid molecule of claim 1, wherein said nucleic acid molecule comprises at least one 2′-sugar modification.
9. The nucleic acid molecule of claim 1, wherein said nucleic acid molecule comprises at least one phosphate backbone modification.
10. A method of reducing PTGDR activity in a cell, comprising contacting said cell with the nucleic acid molecule of claim 1 under conditions suitable for said reduction.
11. A method of treatment of a patient having a condition associated with the level of PTGDR, comprising contacting cells of said patient with the nucleic acid molecule of claim 1, under conditions suitable for said treatment.
12. The method of claim 11 further comprising the use of one or more drug therapies under conditions suitable for said treatment.
13. A pharmaceutical composition comprising an enzymatic nucleic acid molecule of claim 1.
14. A method of administering to a mammal the nucleic acid molecule of claim 1, comprising contacting said mammal with the molecule under conditions suitable for said administration.
15. The method of claim 14, wherein said mammal is a human.
16. The method of claim 14 wherein said administration is in the presence of a delivery reagent.
17. The method of claim 16, wherein said delivery reagent is a lipid.
18. The method of claim 17, wherein said lipid is a cationic lipid.
19. The method of claim 17, wherein said lipid is a phospholipid.
20. The method of claim 17, wherein said delivery reagent is a liposome.
US10/230,006 2001-04-05 2002-08-28 Method and reagent for the treatment of asthma and allergic conditions Abandoned US20030191077A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/230,006 US20030191077A1 (en) 2001-04-05 2002-08-28 Method and reagent for the treatment of asthma and allergic conditions

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US09/827,395 US20030113891A1 (en) 2000-02-11 2001-04-05 Method and reagent for the inhibition of NOGO and NOGO receptor genes
US29441201P 2001-05-29 2001-05-29
US31531501P 2001-08-28 2001-08-28
US10/230,006 US20030191077A1 (en) 2001-04-05 2002-08-28 Method and reagent for the treatment of asthma and allergic conditions

Publications (1)

Publication Number Publication Date
US20030191077A1 true US20030191077A1 (en) 2003-10-09

Family

ID=40293860

Family Applications (7)

Application Number Title Priority Date Filing Date
US10/471,271 Abandoned US20070026394A1 (en) 2000-02-11 2002-04-03 Modulation of gene expression associated with inflammation proliferation and neurite outgrowth using nucleic acid based technologies
US10/156,306 Expired - Fee Related US7022828B2 (en) 2001-04-05 2002-05-28 siRNA treatment of diseases or conditions related to levels of IKK-gamma
US10/206,693 Abandoned US20050261212A1 (en) 2000-02-11 2002-07-26 RNA interference mediated inhibition of NOGO and NOGO receptor gene expression using short interfering RNA
US10/224,005 Abandoned US20030143732A1 (en) 2001-04-05 2002-08-20 RNA interference mediated inhibition of adenosine A1 receptor (ADORA1) gene expression using short interfering RNA
US10/226,992 Abandoned US20030148507A1 (en) 2001-04-05 2002-08-23 RNA interference mediated inhibition of prostaglandin D2 receptor (PTGDR) and prostaglandin D2 synthetase (PTGDS) gene expression using short interfering RNA
US10/230,006 Abandoned US20030191077A1 (en) 2001-04-05 2002-08-28 Method and reagent for the treatment of asthma and allergic conditions
US11/255,139 Abandoned US20060154271A1 (en) 2001-04-05 2005-10-20 Enzymatic nucleic acid treatment of diseases or conditions related to levels of IKK-gamma and PKR

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US10/471,271 Abandoned US20070026394A1 (en) 2000-02-11 2002-04-03 Modulation of gene expression associated with inflammation proliferation and neurite outgrowth using nucleic acid based technologies
US10/156,306 Expired - Fee Related US7022828B2 (en) 2001-04-05 2002-05-28 siRNA treatment of diseases or conditions related to levels of IKK-gamma
US10/206,693 Abandoned US20050261212A1 (en) 2000-02-11 2002-07-26 RNA interference mediated inhibition of NOGO and NOGO receptor gene expression using short interfering RNA
US10/224,005 Abandoned US20030143732A1 (en) 2001-04-05 2002-08-20 RNA interference mediated inhibition of adenosine A1 receptor (ADORA1) gene expression using short interfering RNA
US10/226,992 Abandoned US20030148507A1 (en) 2001-04-05 2002-08-23 RNA interference mediated inhibition of prostaglandin D2 receptor (PTGDR) and prostaglandin D2 synthetase (PTGDS) gene expression using short interfering RNA

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/255,139 Abandoned US20060154271A1 (en) 2001-04-05 2005-10-20 Enzymatic nucleic acid treatment of diseases or conditions related to levels of IKK-gamma and PKR

Country Status (3)

Country Link
US (7) US20070026394A1 (en)
EP (1) EP1386004A4 (en)
WO (1) WO2002081628A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100311816A1 (en) * 2007-11-30 2010-12-09 Noxxon Pharma Ag MCP-1 binding nucleic acids and use thereof

Families Citing this family (562)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993023569A1 (en) * 1992-05-11 1993-11-25 Ribozyme Pharmaceuticals, Inc. Method and reagent for inhibiting viral replication
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)
US5639647A (en) * 1994-03-29 1997-06-17 Ribozyme Pharmaceuticals, Inc. 2'-deoxy-2'alkylnucleotide containing nucleic acid
US20070275921A1 (en) * 1996-06-06 2007-11-29 Isis Pharmaceuticals, Inc. Oligomeric Compounds That Facilitate Risc Loading
US20040171028A1 (en) * 1996-06-06 2004-09-02 Baker Brenda F. Phosphorous-linked oligomeric compounds and their use in gene modulation
US20040161777A1 (en) * 1996-06-06 2004-08-19 Baker Brenda F. Modified oligonucleotides for use in RNA interference
US7812149B2 (en) * 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US20040171030A1 (en) * 1996-06-06 2004-09-02 Baker Brenda F. Oligomeric compounds having modified bases for binding to cytosine and uracil or thymine and their use in gene modulation
US20040171032A1 (en) * 1996-06-06 2004-09-02 Baker Brenda F. Non-phosphorous-linked oligomeric compounds and their use in gene modulation
US20050118605A9 (en) * 1996-06-06 2005-06-02 Baker Brenda F. Oligomeric compounds having modified bases for binding to adenine and guanine and their use in gene modulation
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US20040254358A1 (en) * 2003-06-12 2004-12-16 Muthiah Manoharan Phosphorous-linked oligomeric compounds and their use in gene modulation
US9096636B2 (en) * 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US20040161844A1 (en) * 1996-06-06 2004-08-19 Baker Brenda F. Sugar and backbone-surrogate-containing oligomeric compounds and compositions for use in gene modulation
US20040266706A1 (en) * 2002-11-05 2004-12-30 Muthiah Manoharan Cross-linked oligomeric compounds and their use in gene modulation
JP2003525017A (en) * 1998-04-20 2003-08-26 リボザイム・ファーマシューティカルズ・インコーポレーテッド Nucleic acid molecules with novel chemical composition that can regulate gene expression
US6423493B1 (en) * 1998-10-26 2002-07-23 Board Of Regents The University Of Texas System Combinatorial selection of oligonucleotide aptamers
US20040242521A1 (en) * 1999-10-25 2004-12-02 Board Of Regents, The University Of Texas System Thio-siRNA aptamers
US6939712B1 (en) * 1998-12-29 2005-09-06 Impedagen, Llc Muting gene activity using a transgenic nucleic acid
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
US7829693B2 (en) * 1999-11-24 2010-11-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
DE10100586C1 (en) * 2001-01-09 2002-04-11 Ribopharma Ag Inhibiting gene expression in cells, useful for e.g. treating tumors, by introducing double-stranded complementary oligoRNA having unpaired terminal bases
US20080039414A1 (en) * 2002-02-20 2008-02-14 Sima Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US8202979B2 (en) * 2002-02-20 2012-06-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid
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)
US8273866B2 (en) * 2002-02-20 2012-09-25 Merck Sharp & Dohme Corp. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SINA)
US20050020525A1 (en) * 2002-02-20 2005-01-27 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US7691821B2 (en) * 2001-09-19 2010-04-06 University Of South Florida Inhibition of SHIP to enhance stem cell harvest and transplantation
US20110052546A1 (en) * 2000-09-19 2011-03-03 University Of South Florida Inhibition of SHIP to Enhance Stem Cell Harvest and Transplantation
US20020165192A1 (en) * 2000-09-19 2002-11-07 Kerr William G. Control of NK cell function and survival by modulation of ship activity
ATE544473T1 (en) * 2000-11-09 2012-02-15 Cold Spring Harbor Lab CHIMERIC MOLECULES FOR MODULATION OF GENE EXPRESSION
US7767802B2 (en) * 2001-01-09 2010-08-03 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US7423142B2 (en) * 2001-01-09 2008-09-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
US8546143B2 (en) 2001-01-09 2013-10-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a target gene
EP1386004A4 (en) * 2001-04-05 2005-02-16 Ribozyme Pharm Inc Modulation of gene expression associated with inflammation proliferation and neurite outgrowth, using nucleic acid based technologies
US20080161256A1 (en) * 2001-05-18 2008-07-03 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20060241075A1 (en) * 2001-05-18 2006-10-26 Sirna Therapeutics, Inc. RNA interference mediated inhibition of desmoglein gene expression using short interfering nucleic acid (siNA)
US20050079610A1 (en) * 2001-05-18 2005-04-14 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Fos gene expression using short interfering nucleic acid (siNA)
US20050153914A1 (en) * 2001-05-18 2005-07-14 Sirna Therapeutics, Inc. RNA interference mediated inhibition of MDR P-glycoprotein gene expression using short interfering nucleic acid (siNA)
US7109165B2 (en) * 2001-05-18 2006-09-19 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
US20090299045A1 (en) * 2001-05-18 2009-12-03 Sirna Therapeutics, Inc. RNA Interference Mediated Inhibition Of Interleukin and Interleukin Gene Expression Using Short Interfering Nucleic Acid (siNA)
US7517864B2 (en) 2001-05-18 2009-04-14 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20030124513A1 (en) * 2001-05-29 2003-07-03 Mcswiggen James Enzymatic nucleic acid treatment of diseases or conditions related to levels of HIV
US20050191618A1 (en) * 2001-05-18 2005-09-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of human immunodeficiency virus (HIV) gene expression using short interfering nucleic acid (siNA)
US20050233997A1 (en) * 2001-05-18 2005-10-20 Sirna Therapeutics, Inc. RNA interference mediated inhibition of matrix metalloproteinase 13 (MMP13) gene expression using short interfering nucleic acid (siNA)
US20080188430A1 (en) * 2001-05-18 2008-08-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of hypoxia inducible factor 1 (HIF1) gene expression using short interfering nucleic acid (siNA)
US20050267058A1 (en) * 2001-05-18 2005-12-01 Sirna Therapeutics, Inc. RNA interference mediated inhibition of placental growth factor gene expression using short interfering nucleic acid (sINA)
US20050136436A1 (en) * 2001-05-18 2005-06-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of G72 and D-amino acid oxidase (DAAO) gene expression using short interfering nucleic acid (siNA)
US20050164224A1 (en) * 2001-05-18 2005-07-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of cyclin D1 gene expression using short interfering nucleic acid (siNA)
US20050261219A1 (en) * 2001-05-18 2005-11-24 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (siNA)
US20050176025A1 (en) * 2001-05-18 2005-08-11 Sirna Therapeutics, Inc. RNA interference mediated inhibition of B-cell CLL/Lymphoma-2 (BCL-2) gene expression using short interfering nucleic acid (siNA)
US20070270579A1 (en) * 2001-05-18 2007-11-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20050159382A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc. RNA interference mediated inhibition of polycomb group protein EZH2 gene expression using short interfering nucleic acid (siNA)
US20050054596A1 (en) * 2001-11-30 2005-03-10 Mcswiggen James RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20050158735A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc. RNA interference mediated inhibition of proliferating cell nuclear antigen (PCNA) gene expression using short interfering nucleic acid (siNA)
US20050182007A1 (en) * 2001-05-18 2005-08-18 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (SINA)
US20050124568A1 (en) * 2001-05-18 2005-06-09 Sirna Therapeutics, Inc. RNA interference mediated inhibition of acetyl-CoA-carboxylase gene expression using short interfering nucleic acid (siNA)
US20050187174A1 (en) * 2001-05-18 2005-08-25 Sirna Therapeutics, Inc. RNA interference mediated inhibition of intercellular adhesion molecule (ICAM) gene expression using short interfering nucleic acid (siNA)
US20050196781A1 (en) * 2001-05-18 2005-09-08 Sirna Therapeutics, Inc. RNA interference mediated inhibition of STAT3 gene expression using short interfering nucleic acid (siNA)
US20050164967A1 (en) * 2001-05-18 2005-07-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of platelet-derived endothelial cell growth factor (ECGF1) gene expression using short interfering nucleic acid (siNA)
EP1627061B1 (en) * 2001-05-18 2009-08-12 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING CHEMICALLY MODIFIED SHORT INTERFERING NUCLEIC ACID (siNA)
US20050288242A1 (en) * 2001-05-18 2005-12-29 Sirna Therapeutics, Inc. RNA interference mediated inhibition of RAS gene expression using short interfering nucleic acid (siNA)
US20050196767A1 (en) * 2001-05-18 2005-09-08 Sirna Therapeutics, Inc. RNA interference mediated inhibition of GRB2 associated binding protein (GAB2) gene expression using short interfering nucleic acis (siNA)
US20050196765A1 (en) * 2001-05-18 2005-09-08 Sirna Therapeutics, Inc. RNA interference mediated inhibition of checkpoint Kinase-1 (CHK-1) gene expression using short interfering nucleic acid (siNA)
US20050159379A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc RNA interference mediated inhibition of gastric inhibitory polypeptide (GIP) and gastric inhibitory polypeptide receptor (GIPR) gene expression using short interfering nucleic acid (siNA)
US20050048529A1 (en) * 2002-02-20 2005-03-03 Sirna Therapeutics, Inc. RNA interference mediated inhibition of intercellular adhesion molecule (ICAM) gene expression using short interfering nucleic acid (siNA)
US20050119212A1 (en) * 2001-05-18 2005-06-02 Sirna Therapeutics, Inc. RNA interference mediated inhibition of FAS and FASL gene expression using short interfering nucleic acid (siNA)
US20050287128A1 (en) * 2001-05-18 2005-12-29 Sirna Therapeutics, Inc. RNA interference mediated inhibition of TGF-beta and TGF-beta receptor gene expression using short interfering nucleic acid (siNA)
US20060217331A1 (en) * 2001-05-18 2006-09-28 Sirna Therapeutics, Inc. Chemically modified double stranded nucleic acid molecules that mediate RNA interference
US20050239731A1 (en) * 2001-05-18 2005-10-27 Sirna Therapeutics, Inc. RNA interference mediated inhibition of MAP kinase gene expression using short interfering nucleic acid (siNA)
US20050159380A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc. RNA interference mediated inhibition of angiopoietin gene expression using short interfering nucleic acid (siNA)
US20040019001A1 (en) * 2002-02-20 2004-01-29 Mcswiggen James A. RNA interference mediated inhibition of protein typrosine phosphatase-1B (PTP-1B) gene expression using short interfering RNA
US20050124569A1 (en) * 2001-05-18 2005-06-09 Sirna Therapeutics, Inc. RNA interference mediated inhibition of CXCR4 gene expression using short interfering nucleic acid (siNA)
US9994853B2 (en) 2001-05-18 2018-06-12 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
US20050203040A1 (en) * 2001-05-18 2005-09-15 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular cell adhesion molecule (VCAM) gene expression using short interfering nucleic acid (siNA)
US20070093437A1 (en) * 2001-05-18 2007-04-26 Sirna Therapeutics, Inc. Rna interference mediated inhibition of xiap gene expression using short interfering nucleic acid (sina)
US20050137155A1 (en) * 2001-05-18 2005-06-23 Sirna Therapeutics, Inc. RNA interference mediated treatment of Parkinson disease using short interfering nucleic acid (siNA)
US20050171040A1 (en) * 2001-05-18 2005-08-04 Sirna Therapeutics, Inc. RNA interference mediated inhibition of cholesteryl ester transfer protein (CEPT) gene expression using short interfering nucleic acid (siNA)
US20050222066A1 (en) * 2001-05-18 2005-10-06 Sirna Therapeutics, Inc. RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20070173473A1 (en) * 2001-05-18 2007-07-26 Sirna Therapeutics, Inc. RNA interference mediated inhibition of proprotein convertase subtilisin Kexin 9 (PCSK9) gene expression using short interfering nucleic acid (siNA)
US20050159378A1 (en) * 2001-05-18 2005-07-21 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Myc and/or Myb gene expression using short interfering nucleic acid (siNA)
US20050143333A1 (en) * 2001-05-18 2005-06-30 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (SINA)
US20050282188A1 (en) * 2001-05-18 2005-12-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20060142225A1 (en) * 2001-05-18 2006-06-29 Sirna Therapeutics, Inc. RNA interference mediated inhibition of cyclin dependent kinase-2 (CDK2) gene expression using short interfering nucleic acid (siNA)
US20070042983A1 (en) * 2001-05-18 2007-02-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using short interfering nucleic acid (siNA)
US20060211642A1 (en) * 2001-05-18 2006-09-21 Sirna Therapeutics, Inc. RNA inteference mediated inhibition of hepatitis C virus (HVC) gene expression using short interfering nucleic acid (siNA)
US20050176666A1 (en) * 2001-05-18 2005-08-11 Sirna Therapeutics, Inc. RNA interference mediated inhibition of GPRA and AAA1 gene expression using short interfering nucleic acid (siNA)
US20050233344A1 (en) * 2001-05-18 2005-10-20 Sirna Therapeutics, Inc. RNA interference mediated inhibition of platelet derived growth factor (PDGF) and platelet derived growth factor receptor (PDGFR) gene expression using short interfering nucleic acid (siNA)
US20050277133A1 (en) * 2001-05-18 2005-12-15 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
US20050176663A1 (en) * 2001-05-18 2005-08-11 Sima Therapeutics, Inc. RNA interference mediated inhibition of protein tyrosine phosphatase type IVA (PRL3) gene expression using short interfering nucleic acid (siNA)
US20050164968A1 (en) * 2001-05-18 2005-07-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of ADAM33 gene expression using short interfering nucleic acid (siNA)
US20040198682A1 (en) * 2001-11-30 2004-10-07 Mcswiggen James RNA interference mediated inhibition of placental growth factor gene expression using short interfering nucleic acid (siNA)
US20050124566A1 (en) * 2001-05-18 2005-06-09 Sirna Therapeutics, Inc. RNA interference mediated inhibition of myostatin gene expression using short interfering nucleic acid (siNA)
US7745418B2 (en) 2001-10-12 2010-06-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting viral replication
DE10163098B4 (en) * 2001-10-12 2005-06-02 Alnylam Europe Ag Method for inhibiting the replication of viruses
DE10230997A1 (en) * 2001-10-26 2003-07-17 Ribopharma Ag Drug to increase the effectiveness of a receptor-mediates apoptosis in drug that triggers tumor cells
US20040121348A1 (en) * 2001-10-26 2004-06-24 Ribopharma Ag Compositions and methods for treating pancreatic cancer
US20040063654A1 (en) * 2001-11-02 2004-04-01 Davis Mark E. Methods and compositions for therapeutic use of RNA interference
IL161733A0 (en) * 2001-11-02 2005-11-20 Insert Therapeutics Inc Methods and compositions for therapeutic use of rna interference
ES2401326T3 (en) * 2001-11-21 2013-04-18 Astellas Pharma Inc. Procedure to inhibit gene expression
US20040138163A1 (en) * 2002-05-29 2004-07-15 Mcswiggen James RNA interference mediated inhibition of vascular edothelial growth factor and vascular edothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20050075304A1 (en) * 2001-11-30 2005-04-07 Mcswiggen James RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
US20070203333A1 (en) * 2001-11-30 2007-08-30 Mcswiggen James RNA interference mediated inhibition of vascular endothelial growth factor and vascular endothelial growth factor receptor gene expression using short interfering nucleic acid (siNA)
DE10202419A1 (en) * 2002-01-22 2003-08-07 Ribopharma Ag Method of inhibiting expression of a target gene resulting from chromosome aberration
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
EP1572902B1 (en) 2002-02-01 2014-06-11 Life Technologies Corporation HIGH POTENCY siRNAS FOR REDUCING THE EXPRESSION OF TARGET GENES
ATE508188T1 (en) 2002-02-01 2011-05-15 Life Technologies Corp OLIGONUCLEOTIDE COMPOSITIONS WITH IMPROVED EFFECTIVENESS
US20090093439A1 (en) * 2002-02-20 2009-04-09 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF CHROMOSOME TRANSLOCATION GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US20100240730A1 (en) * 2002-02-20 2010-09-23 Merck Sharp And Dohme Corp. RNA Interference Mediated Inhibition of Gene Expression Using Chemically Modified Short Interfering Nucleic Acid (siNA)
US20090306182A1 (en) * 2002-02-20 2009-12-10 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF MAP KINASE GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US20090099117A1 (en) * 2002-02-20 2009-04-16 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF MYOSTATIN GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP1432724A4 (en) 2002-02-20 2006-02-01 Sirna Therapeutics Inc Rna interference mediated inhibition of map kinase genes
US8232383B2 (en) * 2002-02-20 2012-07-31 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US9657294B2 (en) 2002-02-20 2017-05-23 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US7910724B2 (en) * 2002-02-20 2011-03-22 Sirna Therapeutics, Inc. RNA interference mediated inhibition of Fos gene expression using short interfering nucleic acid (siNA)
US9181551B2 (en) 2002-02-20 2015-11-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US20050096284A1 (en) * 2002-02-20 2005-05-05 Sirna Therapeutics, Inc. RNA interference mediated treatment of polyglutamine (polyQ) repeat expansion diseases using short interfering nucleic acid (siNA)
US8067575B2 (en) * 2002-02-20 2011-11-29 Merck, Sharp & Dohme Corp. RNA interference mediated inhibition of cyclin D1 gene expression using short interfering nucleic acid (siNA)
WO2003106476A1 (en) * 2002-02-20 2003-12-24 Sirna Therapeutics, Inc Nucleic acid mediated inhibition of enterococcus infection and cytolysin toxin activity
US20090137513A1 (en) * 2002-02-20 2009-05-28 Sirna Therapeutics, Inc. RNA Interference Mediated Inhibition of Acetyl-CoA-Carboxylase Gene Expression Using Short Interfering Nucleic Acid (siNA)
US7795422B2 (en) * 2002-02-20 2010-09-14 Sirna Therapeutics, Inc. RNA interference mediated inhibition of hypoxia inducible factor 1 (HIF1) gene expression using short interfering nucleic acid (siNA)
US8658773B2 (en) 2011-05-02 2014-02-25 Immunomedics, Inc. Ultrafiltration concentration of allotype selected antibodies for small-volume administration
WO2003076592A2 (en) * 2002-03-06 2003-09-18 Rigel Pharmaceuticals, Inc. Novel method for delivery and intracellular synthesis of sirna molecules
ES2465574T3 (en) 2002-05-03 2014-06-06 Duke University A method to regulate gene expression
WO2004020577A2 (en) * 2002-05-23 2004-03-11 Mirus Corporation Processes for inhibiting gene expression using polynucleotides
US20040248094A1 (en) * 2002-06-12 2004-12-09 Ford Lance P. Methods and compositions relating to labeled RNA molecules that reduce gene expression
AU2003243541A1 (en) * 2002-06-12 2003-12-31 Ambion, Inc. Methods and compositions relating to labeled rna molecules that reduce gene expression
US20100075423A1 (en) * 2002-06-12 2010-03-25 Life Technologies Corporation Methods and compositions relating to polypeptides with rnase iii domains that mediate rna interference
WO2004002453A1 (en) 2002-06-28 2004-01-08 Protiva Biotherapeutics Ltd. Method and apparatus for producing liposomes
ATE427352T1 (en) 2002-07-26 2009-04-15 Novartis Vaccines & Diagnostic MODIFIED SMALL IRNS MOLECULES AND METHODS OF THEREOF APPLICATION
US20080274989A1 (en) * 2002-08-05 2008-11-06 University Of Iowa Research Foundation Rna Interference Suppression of Neurodegenerative Diseases and Methods of Use Thereof
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
US20050255086A1 (en) * 2002-08-05 2005-11-17 Davidson Beverly L Nucleic acid silencing of Huntington's Disease gene
US20040241854A1 (en) 2002-08-05 2004-12-02 Davidson Beverly L. siRNA-mediated gene silencing
AU2003261449A1 (en) * 2002-08-07 2004-02-25 Compositions for rna interference and methods of use thereof
CA2882443C (en) * 2002-08-21 2016-12-13 The University Of British Columbia Rnai probes targeting cancer-related proteins
US8318922B2 (en) * 2002-08-29 2012-11-27 The Hong Kong Polytechnic University Treatment and prevention of hyperproliferative conditions in humans and antisense oligonucleotide inhibition of human replication-initiation proteins
US7923547B2 (en) * 2002-09-05 2011-04-12 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
WO2004022003A2 (en) 2002-09-06 2004-03-18 University Of South Florida Materials and methods for treatment of allergic diseases
US20080214437A1 (en) * 2002-09-06 2008-09-04 Mohapatra Shyam S Methods and compositions for reducing activity of the atrial natriuretic peptide receptor and for treatment of diseases
US20040053289A1 (en) * 2002-09-09 2004-03-18 The Regents Of The University Of California Short interfering nucleic acid hybrids and methods thereof
US20060287269A1 (en) * 2002-09-09 2006-12-21 The Regents Of The University Of California Short interfering nucleic acid hybrids and methods thereof
WO2004027030A2 (en) * 2002-09-18 2004-04-01 Isis Pharmaceuticals, Inc. Efficient reduction of target rna’s by single- and double-stranded oligomeric compounds
US20050020521A1 (en) * 2002-09-25 2005-01-27 University Of Massachusetts In vivo gene silencing by chemically modified and stable siRNA
US20060240425A1 (en) * 2002-09-30 2006-10-26 Oncotherapy Science, Inc Genes and polypeptides relating to myeloid leukemia
WO2004033653A2 (en) * 2002-10-10 2004-04-22 Oxford Biomedica (Uk) Limited Gene regulation with aptamer and modulator complexes for gene therapy
EP1572978A4 (en) * 2002-10-16 2006-05-24 Univ Texas Bead bound combinatorial oligonucleoside phosphorothioate and phosphorodithioate aptamer libraries
US9150606B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2'-modified nucleosides for use in gene modulation
US9150605B2 (en) * 2002-11-05 2015-10-06 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
CA2504720C (en) * 2002-11-05 2013-12-24 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
AU2003291753B2 (en) * 2002-11-05 2010-07-08 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
WO2004043977A2 (en) * 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. 2’-fluoro substituted oligomeric compounds and compositions for use in gene modulations
WO2004044139A2 (en) 2002-11-05 2004-05-27 Isis Parmaceuticals, Inc. Modified oligonucleotides for use in rna interference
US9827263B2 (en) * 2002-11-05 2017-11-28 Ionis Pharmaceuticals, Inc. 2′-methoxy substituted oligomeric compounds and compositions for use in gene modulations
US7977471B2 (en) * 2002-11-14 2011-07-12 Dharmacon, Inc. siRNA targeting TNFα
US9879266B2 (en) 2002-11-14 2018-01-30 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
US10011836B2 (en) 2002-11-14 2018-07-03 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
US7592442B2 (en) * 2002-11-14 2009-09-22 Dharmacon, Inc. siRNA targeting ribonucleotide reductase M2 polypeptide (RRM2 or RNR-R2)
US9771586B2 (en) 2002-11-14 2017-09-26 Thermo Fisher Scientific Inc. RNAi targeting ZNF205
US9228186B2 (en) 2002-11-14 2016-01-05 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
US9839649B2 (en) 2002-11-14 2017-12-12 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
US7612196B2 (en) 2002-11-14 2009-11-03 Dharmacon, Inc. siRNA targeting cyclin-dependent kinase inhibitor 1B (p27, Kip1) (CDKN1B)
US8198427B1 (en) 2002-11-14 2012-06-12 Dharmacon, Inc. SiRNA targeting catenin, beta-1 (CTNNB1)
US20100113307A1 (en) * 2002-11-14 2010-05-06 Dharmacon, Inc. siRNA targeting vascular endothelial growth factor (VEGF)
US9719094B2 (en) 2002-11-14 2017-08-01 Thermo Fisher Scientific Inc. RNAi targeting SEC61G
JP2006507841A (en) * 2002-11-14 2006-03-09 ダーマコン, インコーポレイテッド Functional and ultrafunctional siRNA
WO2006006948A2 (en) * 2002-11-14 2006-01-19 Dharmacon, Inc. METHODS AND COMPOSITIONS FOR SELECTING siRNA OF IMPROVED FUNCTIONALITY
US20080268457A1 (en) * 2002-11-14 2008-10-30 Dharmacon, Inc. siRNA targeting forkhead box P3 (FOXP3)
US7691998B2 (en) * 2002-11-14 2010-04-06 Dharmacon, Inc. siRNA targeting nucleoporin 62kDa (Nup62)
US7619081B2 (en) * 2002-11-14 2009-11-17 Dharmacon, Inc. siRNA targeting coatomer protein complex, subunit beta 2 (COPB2)
US20090227780A1 (en) * 2002-11-14 2009-09-10 Dharmacon, Inc. siRNA targeting connexin 43
US7951935B2 (en) 2002-11-14 2011-05-31 Dharmacon, Inc. siRNA targeting v-myc myelocytomatosis viral oncogene homolog (MYC)
US20090005548A1 (en) * 2002-11-14 2009-01-01 Dharmacon, Inc. siRNA targeting nuclear receptor interacting protein 1 (NRIP1)
US9719092B2 (en) 2002-11-14 2017-08-01 Thermo Fisher Scientific Inc. RNAi targeting CNTD2
US7635770B2 (en) * 2002-11-14 2009-12-22 Dharmacon, Inc. siRNA targeting protein kinase N-3 (PKN-3)
US7781575B2 (en) 2002-11-14 2010-08-24 Dharmacon, Inc. siRNA targeting tumor protein 53 (p53)
US7605249B2 (en) 2002-11-26 2009-10-20 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of siRNA
US7829694B2 (en) * 2002-11-26 2010-11-09 Medtronic, Inc. Treatment of neurodegenerative disease through intracranial delivery of siRNA
US7618948B2 (en) * 2002-11-26 2009-11-17 Medtronic, Inc. Devices, systems and methods for improving and/or cognitive function through brain delivery of siRNA
EP1583836A4 (en) * 2002-12-11 2006-12-27 Univ Massachusetts METHOD OF INTRODUCING siRNA INTO ADIPOCYTES
WO2004061081A2 (en) * 2002-12-27 2004-07-22 Ichem Technologies Sirna compounds and methods for the downregulation of gene expression
DE10302421A1 (en) * 2003-01-21 2004-07-29 Ribopharma Ag New double-stranded interfering RNA, useful for inhibiting hepatitis C virus, has one strand linked to a lipophilic group to improve activity and eliminate the need for transfection auxiliaries
US20060178297A1 (en) * 2003-01-28 2006-08-10 Troy Carol M Systems and methods for silencing expression of a gene in a cell and uses thereof
US7994149B2 (en) 2003-02-03 2011-08-09 Medtronic, Inc. Method for treatment of Huntington's disease through intracranial delivery of sirna
US7732591B2 (en) * 2003-11-25 2010-06-08 Medtronic, Inc. Compositions, devices and methods for treatment of huntington's disease through intracranial delivery of sirna
WO2004076664A2 (en) * 2003-02-21 2004-09-10 University Of South Florida Vectors for regulating gene expression
US20040167090A1 (en) * 2003-02-21 2004-08-26 Monahan Sean D. Covalent modification of RNA for in vitro and in vivo delivery
US7521534B1 (en) 2003-03-03 2009-04-21 The University Board Of Regents Of Texas System IKK gamma gene products and methods for making and using same
EP1604022A2 (en) * 2003-03-06 2005-12-14 Oligo Engine, Inc. Modulation of gene expression using dna-rna hybrids
EP1605978B1 (en) * 2003-03-07 2010-09-01 Alnylam Pharmaceuticals Inc. Therapeutic compositions
AU2004220459B2 (en) * 2003-03-12 2010-08-05 Vasgene Therapeutics, Inc. Polypeptide compounds for inhibiting angiogenesis and tumor growth
DK2141234T3 (en) * 2003-03-21 2016-06-20 Roche Innovation Ct Copenhagen As Short interfering RNA (siRNA) analogues
WO2004086047A2 (en) * 2003-03-28 2004-10-07 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with g-protein-coupled receptor adenosine a1 (adora1)
JP2006522158A (en) * 2003-04-03 2006-09-28 アルナイラム ファーマシューティカルズ インコーポレイテッド iRNA complex
WO2004092383A2 (en) * 2003-04-15 2004-10-28 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED INHIBITION OF SEVERE ACUTE RESPIRATORY SYNDROME (SARS) VIRUS GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
JP2006523464A (en) * 2003-04-18 2006-10-19 ザ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・ペンシルバニア Compositions and methods for siRNA inhibition of angiopoietins 1, 2 and their receptor TIE2
WO2005032595A2 (en) * 2003-04-23 2005-04-14 Georgetown University Methods and compositions for the inhibition of stat5 in prostate cancer cells
US20040224405A1 (en) * 2003-05-06 2004-11-11 Dharmacon Inc. siRNA induced systemic gene silencing in mammalian systems
WO2005037053A2 (en) * 2003-05-23 2005-04-28 Board Of Regents - The University Of Texas System High throughput screening of aptamer libraries for specific binding to proteins on viruses and other pathogens
US7910523B2 (en) * 2003-05-23 2011-03-22 Board Of Regents, The University Of Texas System Structure based and combinatorially selected oligonucleoside phosphorothioate and phosphorodithioate aptamer targeting AP-1 transcription factors
US20050042641A1 (en) * 2003-05-27 2005-02-24 Cold Spring Harbor Laboratory In vivo high throughput selection of RNAi probes
US8092992B2 (en) * 2003-05-29 2012-01-10 Salk Institute For Biological Studies Transcriptional regulation of gene expression by small double-stranded modulatory RNA
US7750144B2 (en) * 2003-06-02 2010-07-06 University Of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNA silencing
ES2864206T3 (en) * 2003-06-02 2021-10-13 Univ Massachusetts Methods and compositions to improve the efficacy and specificity of RNAi
US20050020526A1 (en) * 2003-06-03 2005-01-27 Cytogenix, Inc. Oligodeoxynucleotide intervention for prevention and treatment of sepsis
US7595306B2 (en) * 2003-06-09 2009-09-29 Alnylam Pharmaceuticals Inc Method of treating neurodegenerative disease
CA2542232A1 (en) * 2003-06-09 2005-01-20 Alnylam Pharmaceuticals, Inc. Method for treating neurodegenerative disease by inhibiting alpha-synuclein
US8575327B2 (en) 2003-06-12 2013-11-05 Alnylam Pharmaceuticals, Inc. Conserved HBV and HCV sequences useful for gene silencing
EP1644475A4 (en) * 2003-06-20 2009-06-03 Isis Pharmaceuticals Inc Double stranded compositions comprising a 3'-endo modified strand for use in gene modulation
US9233131B2 (en) 2003-06-30 2016-01-12 The Regents Of The University Of California Mutant adeno-associated virus virions and methods of use thereof
US9441244B2 (en) * 2003-06-30 2016-09-13 The Regents Of The University Of California Mutant adeno-associated virus virions and methods of use thereof
FR2857013B1 (en) * 2003-07-02 2005-09-30 Commissariat Energie Atomique SMALL INTERFERING RNA SPECIFIC OF ALPHA, ALPHA PRIME AND BETA SUBUNITS OF PROTEIN KINASE CK2 AND THEIR APPLICATIONS
US20050256071A1 (en) * 2003-07-15 2005-11-17 California Institute Of Technology Inhibitor nucleic acids
US20050136430A1 (en) * 2003-07-15 2005-06-23 California Institute Of Technology Inhibitor nucleic acids
AU2004257373B2 (en) * 2003-07-16 2011-03-24 Arbutus Biopharma Corporation Lipid encapsulated interfering RNA
WO2005010016A2 (en) * 2003-07-24 2005-02-03 Board Of Regents Thioaptamers enable discovery of physiological pathways and new therapeutic strategies
US20050059024A1 (en) 2003-07-25 2005-03-17 Ambion, Inc. Methods and compositions for isolating small RNA molecules
CA2533701A1 (en) 2003-07-31 2005-02-17 Isis Pharmaceuticals, Inc. Oligomeric compounds and compositions for use in modulation of small non-coding rnas
WO2005012483A2 (en) * 2003-08-01 2005-02-10 International Therapeutics, Inc. Vpr selective rnai agents and methods for using the same
WO2005012487A2 (en) * 2003-08-01 2005-02-10 Invitrogen Corporation Compositions and methods for preparing short rna molecules and other nucleic acids
US20050136437A1 (en) * 2003-08-25 2005-06-23 Nastech Pharmaceutical Company Inc. Nanoparticles for delivery of nucleic acids and stable double-stranded RNA
MXPA06002216A (en) * 2003-08-28 2006-04-27 Novartis Ag INTERFERING RNA DUPLEX HAVING BLUNT-ENDS AND 3aCO-MODIFICATIONS.
US20070202505A1 (en) * 2003-09-08 2007-08-30 Alex Chenchik Methods for gene function analysis
US20050074801A1 (en) * 2003-09-09 2005-04-07 Monia Brett P. Chimeric oligomeric compounds comprising alternating regions of northern and southern conformational geometry
US20050059019A1 (en) * 2003-09-11 2005-03-17 Sven Bulow Gene-related RNAi transfection method
NZ592917A (en) * 2003-09-15 2012-12-21 Protiva Biotherapeutics Inc Stable polyethyleneglycol (PEG) dialkyloxypropyl (DAA) lipid conjugates
EP1670955A2 (en) * 2003-09-22 2006-06-21 Rosetta Inpharmatics LLC. Synthetic lethal screen using rna interference
US20070218551A1 (en) * 2003-10-02 2007-09-20 Chuan-Yuan Li Novel Sirna-Based Approach to Target the Hif-Alpha Factor for Gene Therapy
CA2541852A1 (en) * 2003-10-07 2005-05-12 Quark Biotech, Inc. Bone morphogenetic protein (bmp) 2a and uses thereof
DE10346721A1 (en) * 2003-10-08 2005-05-04 Holger Kalthoff New oligonucleotides, useful for treating cancer, especially of the pancreas, are not species specific but induce apoptosis or inhibit proliferation
US20050191283A1 (en) * 2003-10-16 2005-09-01 Suzanne Kadereit Methods of treating NFAT-related disorders
CA2543013A1 (en) * 2003-10-23 2005-05-19 Sirna Therapeutics, Inc. Rna interference mediated inhibition of nogo and nogo receptor gene expression using short interfering nucleic acid (sina)
US7763592B1 (en) * 2003-11-20 2010-07-27 University Of South Florida SHIP-deficiency to increase megakaryocyte progenitor production
US7807646B1 (en) * 2003-11-20 2010-10-05 University Of South Florida SHIP-deficiency to increase megakaryocyte progenitor production
US20050208658A1 (en) * 2003-11-21 2005-09-22 The University Of Maryland RNA interference mediated inhibition of 11beta hydroxysteriod dehydrogenase-1 (11beta HSD-1) gene expression
JP2007512027A (en) * 2003-11-21 2007-05-17 レビビコア, インコーポレイテッド Use of interfering RNA in the production of transgenic animals
US20070238676A1 (en) 2003-12-04 2007-10-11 Mohapatra Shyam S Polynucleotides for Reducing Respiratory Syncytial Virus Gene Expression
SE0303397D0 (en) * 2003-12-17 2003-12-17 Index Pharmaceuticals Ab Compounds and method for RNA interference
US20050164970A1 (en) * 2003-12-22 2005-07-28 University Of Kansas Medical Center Method for treating prostate cancer using siRNA duplex for androgen receptor
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
US20070161586A1 (en) * 2004-01-16 2007-07-12 Takeda Pharmaceutical Company Limited Drug for preventing and treating atherosclerosis
WO2005073250A2 (en) * 2004-01-28 2005-08-11 Lorantis Limited Medical treatment using an rna1 targeting a human notch signalling pathway member
EP1758998B1 (en) * 2004-01-30 2010-12-15 Quark Pharmaceuticals, Inc. Oligoribonucleotides and methods of use thereof for treatment of fibrotic conditions and other diseases
EP1713912B1 (en) * 2004-01-30 2013-09-18 Santaris Pharma A/S Modified short interfering rna (modified sirna)
US7858769B2 (en) 2004-02-10 2010-12-28 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using multifunctional short interfering nucleic acid (multifunctional siNA)
EP1727556A2 (en) * 2004-02-17 2006-12-06 University Of South Florida Materials and methods for treatment of inflammatory and cell proliferation disorders
US20060069050A1 (en) * 2004-02-17 2006-03-30 University Of Massachusetts Methods and compositions for mediating gene silencing
CA2558262A1 (en) * 2004-03-01 2005-09-15 Massachusetts Institute Of Technology Rnai-based therapeutics for allergic rhinitis and asthma
DE102004010547A1 (en) * 2004-03-03 2005-11-17 Beiersdorf Ag Oligoribonucleotides for the treatment of irritative and / or inflammatory skin conditions by RNA interference
US8569474B2 (en) * 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
AU2005222902B2 (en) 2004-03-12 2010-06-10 Alnylam Pharmaceuticals, Inc. iRNA agents targeting VEGF
EP1742958B1 (en) * 2004-03-15 2017-05-17 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
US20050208090A1 (en) * 2004-03-18 2005-09-22 Medtronic, Inc. Methods and systems for treatment of neurological diseases of the central nervous system
US20050272682A1 (en) * 2004-03-22 2005-12-08 Evers Bernard M SiRNA targeting PI3K signal transduction pathway and siRNA-based therapy
EP1737878A2 (en) 2004-04-05 2007-01-03 Alnylam Pharmaceuticals Inc. Process and reagents for oligonucleotide synthesis and purification
US20050244869A1 (en) * 2004-04-05 2005-11-03 Brown-Driver Vickie L Modulation of transthyretin expression
MXPA06012076A (en) * 2004-04-20 2007-01-25 Nastech Pharm Co Methods and compositions for enhancing delivery of double-stranded rna or a double-stranded hybrid nucleic acid to regulate gene expression in mammalian cells.
US20050239134A1 (en) * 2004-04-21 2005-10-27 Board Of Regents, The University Of Texas System Combinatorial selection of phosphorothioate single-stranded DNA aptamers for TGF-beta-1 protein
WO2006078278A2 (en) 2004-04-27 2006-07-27 Alnylam Pharmaceuticals, Inc. Single-stranded and double-stranded oligonucleotides comprising a 2-arylpropyl moiety
EP1750776A2 (en) 2004-04-30 2007-02-14 Alnylam Pharmaceuticals Inc. Oligonucleotides comprising a c5-modified pyrimidine
US20060040882A1 (en) * 2004-05-04 2006-02-23 Lishan Chen Compostions and methods for enhancing delivery of nucleic acids into cells and for modifying expression of target genes in cells
US20110117088A1 (en) * 2004-05-12 2011-05-19 Simon Michael R Composition and method for introduction of rna interference sequences into targeted cells and tissues
US7605250B2 (en) * 2004-05-12 2009-10-20 Dharmacon, Inc. siRNA targeting cAMP-specific phosphodiesterase 4D
US7563885B1 (en) * 2004-05-24 2009-07-21 Isis Pharmaceuticals, Inc. Modulation of Tudor-SN expression
US10508277B2 (en) 2004-05-24 2019-12-17 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
EP2399924B1 (en) 2004-05-27 2015-01-28 Alnylam Pharmaceuticals, Inc. Nuclease resistant double-stranded ribonucleic acid
AU2005252662B2 (en) * 2004-06-03 2011-08-18 Isis Pharmaceuticals, Inc. Double strand compositions comprising differentially modified strands for use in gene modulation
US20090048192A1 (en) * 2004-06-03 2009-02-19 Isis Pharmaceuticals, Inc. Double Strand Compositions Comprising Differentially Modified Strands for Use in Gene Modulation
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
EP1781593B1 (en) * 2004-06-07 2011-12-14 Protiva Biotherapeutics Inc. Cationic lipids and methods of use
ATE536418T1 (en) * 2004-06-07 2011-12-15 Protiva Biotherapeutics Inc LIPID ENCAPSULATED INTERFERENCE RNA
EP1789553B1 (en) 2004-06-30 2014-03-26 Alnylam Pharmaceuticals Inc. Oligonucleotides comprising a non-phosphate backbone linkage
JP2008504827A (en) * 2004-07-02 2008-02-21 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 Immunostimulatory siRNA molecules and methods of use thereof
US8361976B2 (en) * 2004-07-09 2013-01-29 University Of Massachusetts Therapeutic alteration of transplantable tissues through in situ or ex vivo exposure to RNA interference molecules
US8604185B2 (en) * 2004-07-20 2013-12-10 Genentech, Inc. Inhibitors of angiopoietin-like 4 protein, combinations, and their use
ZA200701183B (en) * 2004-07-20 2008-05-28 Genentech Inc Inhibitors of angiopoietin-like 4 protein, combinations, an their use
WO2006093526A2 (en) * 2004-07-21 2006-09-08 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a modified or non-natural nucleobase
AU2005330637B2 (en) 2004-08-04 2012-09-20 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising a ligand tethered to a modified or non-natural nucleobase
ATE555202T1 (en) 2004-08-16 2012-05-15 Immune Disease Inst Inc METHOD FOR DELIVERING RNA INTERFERENCE AND USES THEREOF
US20110313024A1 (en) * 2004-08-20 2011-12-22 Leonid Beigelman RNA INTERFERENCE MEDIATED INHIBITION OF PROPROTEIN CONVERTASE SUBTILISIN KEXIN 9 (PCSK9) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
RU2410430C2 (en) * 2004-08-31 2011-01-27 Силентис С.А.У. Methods and compositions for inhibiting expression of p2x7 receptor
US7718624B2 (en) * 2004-09-01 2010-05-18 Sitkovsky Michail V Modulation of immune response and inflammation by targeting hypoxia inducible factors
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
EP1793835A4 (en) 2004-09-10 2010-12-01 Somagenics Inc SMALL INTERFERING RNAs THAT EFFICIENTLY INHIBIT VIRAL GENE EXPRESSION AND METHODS OF USE THEREOF
EP2982754A1 (en) 2004-09-24 2016-02-10 Alnylam Pharmaceuticals, Inc. Rnai modulation of apob and uses thereof
EP1796732B1 (en) * 2004-10-01 2013-10-30 Novartis Vaccines and Diagnostics, Inc. Modified small interfering rna molecules and methods of use
US8765704B1 (en) 2008-02-28 2014-07-01 Novartis Ag Modified small interfering RNA molecules and methods of use
US20070078085A1 (en) * 2004-10-13 2007-04-05 Chung Leland W Methods and compositions for the utilization and targeting of osteomimicry
US20060253100A1 (en) 2004-10-22 2006-11-09 Medtronic, Inc. Systems and Methods to Treat Pain Locally
WO2006043014A1 (en) * 2004-10-22 2006-04-27 Neuregenix Limited Neuron regeneration
US7790878B2 (en) * 2004-10-22 2010-09-07 Alnylam Pharmaceuticals, Inc. RNAi modulation of RSV, PIV and other respiratory viruses and uses thereof
DK2302055T3 (en) 2004-11-12 2014-10-13 Asuragen Inc Methods and compositions involving miRNA and miRNA inhibitor molecules
WO2006053430A1 (en) * 2004-11-17 2006-05-26 Protiva Biotherapeutics, Inc. Sirna silencing of apolipoprotein b
JP2008521401A (en) * 2004-11-24 2008-06-26 アルナイラム ファーマシューティカルズ インコーポレイテッド RNAi regulation of BCR-ABL fusion gene and method of use thereof
WO2006071451A2 (en) * 2004-12-03 2006-07-06 The Regents Of The University Of California Compounds that prevent macrophage apoptosis and uses thereof
WO2006063356A1 (en) * 2004-12-10 2006-06-15 Isis Phamaceuticals, Inc. Regulation of epigenetic control of gene expression
WO2006066158A2 (en) * 2004-12-14 2006-06-22 Alnylam Pharmaceuticals, Inc. Rnai modulation of mll-af4 and uses thereof
US20090010907A1 (en) * 2004-12-14 2009-01-08 National Institute Of Immunology Dnazymes for Inhibition of Japanese Encephalitis Virus Replication
US20060142228A1 (en) * 2004-12-23 2006-06-29 Ambion, Inc. Methods and compositions concerning siRNA's as mediators of RNA interference
CN102600480B (en) * 2005-01-07 2015-07-22 阿尔尼拉姆医药品有限公司 RNAI modulation of RSV and therapeutic uses thereof
WO2006081192A2 (en) * 2005-01-24 2006-08-03 Alnylam Pharmaceuticals, Inc. Rnai modulation of the nogo-l or nogo-r gene and uses thereof
TW200639253A (en) * 2005-02-01 2006-11-16 Alcon Inc RNAi-mediated inhibition of ocular targets
CA2597845A1 (en) 2005-02-25 2006-08-31 Isis Pharmaceuticals, Inc. Compositions and their uses directed to il-4r alpha
EP2157182A3 (en) * 2005-03-08 2012-04-25 Qiagen GmbH Modified short interfering RNA
EP1856259A1 (en) 2005-03-11 2007-11-21 Alcon Inc. Rnai-mediated inhibition of frizzled related protein-1 for treatment of glaucoma
WO2006130201A1 (en) * 2005-03-14 2006-12-07 Board Of Regents, The University Of Texas System Antigene oligomers inhibit transcription
JP4131271B2 (en) * 2005-03-30 2008-08-13 ソニー株式会社 Information processing apparatus and method, and program
EP1874930B1 (en) * 2005-04-12 2011-09-07 Université Libre de Bruxelles Use of a galectin-1-trageted rnai-based approach for the treatment of cancer
US20060253068A1 (en) * 2005-04-20 2006-11-09 Van Bilsen Paul Use of biocompatible in-situ matrices for delivery of therapeutic cells to the heart
US7902352B2 (en) * 2005-05-06 2011-03-08 Medtronic, Inc. Isolated nucleic acid duplex for reducing huntington gene expression
EP1885854B1 (en) * 2005-05-06 2012-10-17 Medtronic, Inc. Methods and sequences to suppress primate huntington gene expression
CN101213300B (en) 2005-06-01 2013-01-23 聚加转染股份有限公司 Oligonucleotides for RNA interference and biological applications thereof
FI20050640A0 (en) * 2005-06-16 2005-06-16 Faron Pharmaceuticals Oy Compounds for treating or preventing diseases or disorders related to amine oxidases
WO2007002718A2 (en) * 2005-06-27 2007-01-04 Alnylam Pharmaceuticals, Inc. Rnai modulation of hif-1 and theraputic uses thereof
US20100129288A1 (en) * 2005-06-28 2010-05-27 Elior Peles Gliomedin, Fragments Thereof and Methods of Using Same
US9133517B2 (en) 2005-06-28 2015-09-15 Medtronics, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin
US20080280843A1 (en) * 2006-05-24 2008-11-13 Van Bilsen Paul Methods and kits for linking polymorphic sequences to expanded repeat mutations
WO2007007317A1 (en) * 2005-07-07 2007-01-18 Yissum Research Development Company Of The Hebrew University Of Jerusalem Nucleic acid agents for downregulating h19, and methods of using same
EP2230305A1 (en) * 2005-07-21 2010-09-22 Alnylam Pharmaceuticals Inc. Rnai modulation of the rho-a gene and uses thereof
US7919583B2 (en) * 2005-08-08 2011-04-05 Discovery Genomics, Inc. Integration-site directed vector systems
US20090176725A1 (en) * 2005-08-17 2009-07-09 Sirna Therapeutics Inc. Chemically modified short interfering nucleic acid molecules that mediate rna interference
WO2007020645A1 (en) * 2005-08-18 2007-02-22 Hadasit Medical Research Services & Development Limited Gene silencing of protease activated receptor 1 (par1)
US20070054873A1 (en) * 2005-08-26 2007-03-08 Protiva Biotherapeutics, Inc. Glucocorticoid modulation of nucleic acid-mediated immune stimulation
TWI333959B (en) * 2005-08-31 2010-12-01 Academia Sinica Methods and reagents for the analysis and purification of polysaccharides
US7943134B2 (en) * 2005-08-31 2011-05-17 Academia Sinica Compositions and methods for identifying response targets and treating flavivirus infection responses
WO2007030167A1 (en) * 2005-09-02 2007-03-15 Nastech Pharmaceutical Company Inc. Modification of double-stranded ribonucleic acid molecules
US20070099858A1 (en) * 2005-10-03 2007-05-03 Sirna Therapeutics, Inc. RNA interference mediated of inhibition of influenza virus gene expression using short interfering nucleic acid (siNA)
AU2006299345A1 (en) * 2005-10-03 2007-04-12 Isis Pharmaceuticals, Inc. Combination therapy using budesonide and antisense oligonucleotide targeted to IL4-receptor alpha
EP2189522A1 (en) * 2005-10-14 2010-05-26 MUSC Foundation For Research Development Targeting PAX2 for the induction of DEFB1-mediated tumor immunity and cancer therapy
WO2007048046A2 (en) * 2005-10-20 2007-04-26 Protiva Biotherapeutics, Inc. Sirna silencing of filovirus gene expression
CN101365801B (en) 2005-10-28 2013-03-27 阿尔尼拉姆医药品有限公司 Compositions and methods for inhibiting expression of huntingtin gene
EP1948674A4 (en) 2005-11-02 2009-02-04 Protiva Biotherapeutics Inc Modified sirna molecules and uses thereof
WO2007056326A2 (en) * 2005-11-04 2007-05-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of nav1.8 gene
CA2626690A1 (en) * 2005-11-09 2007-05-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of factor v leiden mutant gene
EP1957648B1 (en) * 2005-11-17 2014-04-23 Board of Regents, The University of Texas System Modulation of gene expression by oligomers targeted to chromosomal dna
EP1948244A4 (en) * 2005-11-17 2010-10-06 Tel Hashomer Medical Res Infrastructure & Services Ltd Pharmaceutical composition and method for regulating abnormal cellular proliferation
WO2007056861A1 (en) * 2005-11-18 2007-05-24 Protiva Biotherapeutics, Inc. Sirna silencing of influenza virus gene expression
JP4901753B2 (en) * 2005-11-24 2012-03-21 学校法人自治医科大学 Mitochondrial function of prohibitin 2 (PHB2)
EP1969143A4 (en) * 2005-12-20 2009-07-22 Isis Pharmaceuticals Inc Double stranded nucleic acid molecules targeted to il-4 receptor alpha
WO2007089607A2 (en) * 2006-01-26 2007-08-09 University Of Massachusetts Rna silencing agents for use in therapy and nanotransporters for efficient delivery of same
DK1984382T3 (en) * 2006-01-27 2012-09-03 Santaris Pharma As LNA modified phosphorothiolated oligonucleotides
US8229398B2 (en) * 2006-01-30 2012-07-24 Qualcomm Incorporated GSM authentication in a CDMA network
FI20060246A0 (en) 2006-03-16 2006-03-16 Jukka Westermarck A new growth stimulating protein and its use
US8329888B2 (en) * 2006-03-23 2012-12-11 Santaris Pharma A/S Small internally segmented interfering RNA
WO2007111998A2 (en) * 2006-03-24 2007-10-04 Novartis Ag Dsrna compositions and methods for treating hpv infection
WO2007115047A2 (en) * 2006-03-29 2007-10-11 Senesco Technologies, Inc. Inhibition of hiv replication and expression of p24 with eif-5a
NZ571568A (en) 2006-03-31 2010-11-26 Alnylam Pharmaceuticals Inc Double-stranded RNA molecule compositions and methods for inhibiting expression of Eg5 gene
EP2007891A2 (en) * 2006-04-06 2008-12-31 DKFZ Deutsches Krebsforschungszentrum Method to inhibit the propagation of an undesired cell population
WO2007127919A2 (en) 2006-04-28 2007-11-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of a gene from the jc virus
WO2007127487A2 (en) * 2006-04-28 2007-11-08 University Of South Florida Materials and methods for reducing inflammation by inhibition of the atrial natriuretic peptide receptor
GB0608838D0 (en) * 2006-05-04 2006-06-14 Novartis Ag Organic compounds
CN103614375A (en) 2006-05-11 2014-03-05 阿尔尼拉姆医药品有限公司 Composition and method for inhibiting expression of PCSK9 gene
US20070269892A1 (en) * 2006-05-18 2007-11-22 Nastech Pharmaceutical Company Inc. FORMULATIONS FOR INTRACELLULAR DELIVERY dsRNA
CA2652770A1 (en) * 2006-05-19 2007-11-29 Alnylam Pharmaceuticals, Inc. Rnai modulation of aha and therapeutic uses thereof
WO2007137220A2 (en) * 2006-05-22 2007-11-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of ikk-b gene
US9273356B2 (en) 2006-05-24 2016-03-01 Medtronic, Inc. Methods and kits for linking polymorphic sequences to expanded repeat mutations
US20070275923A1 (en) * 2006-05-25 2007-11-29 Nastech Pharmaceutical Company Inc. CATIONIC PEPTIDES FOR siRNA INTRACELLULAR DELIVERY
US8598333B2 (en) * 2006-05-26 2013-12-03 Alnylam Pharmaceuticals, Inc. SiRNA silencing of genes expressed in cancer
KR100906145B1 (en) * 2006-05-30 2009-07-03 한국생명공학연구원 A anticancer drug comprising inhibitor of TMPRSS4
US7915399B2 (en) * 2006-06-09 2011-03-29 Protiva Biotherapeutics, Inc. Modified siRNA molecules and uses thereof
WO2008008719A2 (en) * 2006-07-10 2008-01-17 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the myc gene
EP2471815B1 (en) 2006-07-11 2016-03-30 University Of Medicine And Dentistry Of New Jersey Proteins, nucleic acids encoding the same and associated methods of use
CA2659103C (en) * 2006-07-12 2019-05-21 The Regents Of The University Of California Transducible delivery of nucleic acids by reversible phosphotriester charge neutralization protecting groups
JP4756271B2 (en) * 2006-07-18 2011-08-24 独立行政法人産業技術総合研究所 Cancer cell aging, apoptosis inducer
EP1884569A1 (en) * 2006-07-31 2008-02-06 Institut National De La Sante Et De La Recherche Medicale (Inserm) Sensitization of cancer cells to therapy using siNA targeting genes from the 1p and 19q chromosomal regions
US20080039415A1 (en) * 2006-08-11 2008-02-14 Gregory Robert Stewart Retrograde transport of sirna and therapeutic uses to treat neurologic disorders
FI20060751A0 (en) 2006-08-23 2006-08-23 Valtion Teknillinen Method of treating prostate cancer and screening of patients who benefit from said method
US7825101B2 (en) * 2006-08-30 2010-11-02 The Board Of Trustees Of The University Of Illinois Modulation of MLCK-L expression and uses thereof
JP5431940B2 (en) 2006-09-18 2014-03-05 アルナイラム ファーマシューティカルズ, インコーポレイテッド RNAi regulation of SCAP and its therapeutic use
AU2007299629C1 (en) 2006-09-21 2012-05-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the HAMP gene
US8324367B2 (en) 2006-11-03 2012-12-04 Medtronic, Inc. Compositions and methods for making therapies delivered by viral vectors reversible for safety and allele-specificity
US9375440B2 (en) * 2006-11-03 2016-06-28 Medtronic, Inc. Compositions and methods for making therapies delivered by viral vectors reversible for safety and allele-specificity
US7819842B2 (en) 2006-11-21 2010-10-26 Medtronic, Inc. Chronically implantable guide tube for repeated intermittent delivery of materials or fluids to targeted tissue sites
US8034921B2 (en) * 2006-11-21 2011-10-11 Alnylam Pharmaceuticals, Inc. IRNA agents targeting CCR5 expressing cells and uses thereof
US7988668B2 (en) * 2006-11-21 2011-08-02 Medtronic, Inc. Microsyringe for pre-packaged delivery of pharmaceuticals
US20080171719A1 (en) * 2006-11-28 2008-07-17 Alcon Manufacturing, Ltd. RNAi-MEDIATED INHIBITION OF AQUAPORIN 1 FOR TREATMENT OF IOP-RELATED CONDITIONS
WO2008067382A2 (en) * 2006-11-28 2008-06-05 Alcon Research, Ltd. Rnai-mediated inhibition of aquaporin 4 for treatment of iop-related conditions
US7951789B2 (en) * 2006-12-28 2011-05-31 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US20080171906A1 (en) * 2007-01-16 2008-07-17 Everaerts Frank J L Tissue performance via hydrolysis and cross-linking
US7928083B2 (en) * 2007-01-16 2011-04-19 Yissum Research Development Company Of The Hebrew University Of Jerusalem H19 silencing nucleic acid agents for treating rheumatoid arthritis
CA2712056C (en) 2007-01-16 2016-06-21 The University Of Queensland Method of inducing an immune response
US20090054365A1 (en) * 2007-01-26 2009-02-26 Alcon Research, Ltd. RNAi-MEDIATED INHIBITION OF AQUAPORIN 1 FOR TREATMENT OF OCULAR NEOVASCULARIZATION
WO2008094945A2 (en) * 2007-01-29 2008-08-07 Isis Pharmaceuticals, Inc. Compounds and methods for modulating protein expression
US20100196403A1 (en) * 2007-01-29 2010-08-05 Jacob Hochman Antibody conjugates for circumventing multi-drug resistance
US20100183696A1 (en) * 2007-01-30 2010-07-22 Allergan, Inc Treating Ocular Diseases Using Peroxisome Proliferator-Activated Receptor Delta Antagonists
JP2010519256A (en) 2007-02-23 2010-06-03 ザ・トラスティーズ・オブ・コランビア・ユニバーシティー・イン・ザ・シティー・オブ・ニューヨーク Methods of activating or blocking the HLA-E / Qa-1 restricted CD8 + T cell regulatory pathway to treat immune diseases
PE20090064A1 (en) * 2007-03-26 2009-03-02 Novartis Ag DOUBLE-CHAIN RIBONUCLEIC ACID TO INHIBIT THE EXPRESSION OF THE HUMAN E6AP GENE AND THE PHARMACEUTICAL COMPOSITION THAT INCLUDES IT
AP3018A (en) * 2007-03-29 2014-10-31 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expressionof a gene from the ebola
US8343941B2 (en) 2007-03-30 2013-01-01 Rutgers, The State University Of New Jersey Compositions and methods for gene silencing
US9441221B2 (en) 2007-03-30 2016-09-13 Rutgers, The State University Of New Jersey Compositions and methods for gene silencing
US8907075B2 (en) * 2007-03-30 2014-12-09 Samuel Ian Gunderson Compositions and methods for gene silencing
EP2142672B1 (en) * 2007-03-30 2012-09-05 Rutgers, The State University of New Jersey Compositions and methods for gene silencing
HUE040417T2 (en) 2007-05-04 2019-03-28 Marina Biotech Inc Amino acid lipids and uses thereof
CA2686908A1 (en) * 2007-05-11 2008-11-20 Enzon Pharmaceuticals, Inc. Rna antagonist compounds for the modulation of her3
MY153691A (en) 2007-05-22 2015-03-13 Arcturus Therapeutics Inc Hydroxymethyl substituted rna oligonucleotides and rna complexes
EP3456357A1 (en) 2007-06-29 2019-03-20 Stelic Institute Of Regenerative Medicine, Stelic Institute & Co. Method of fixing and expressing physiologically active substance
ES2423182T3 (en) * 2007-07-10 2013-09-18 Neurim Pharmaceuticals (1991) Ltd. CD44 binding variants in neurodegenerative diseases
WO2009038707A2 (en) * 2007-09-17 2009-03-26 Ludwig Institute For Cancer Research , Ltd. Cancer-testis gene silencing agents and uses thereof
EP2548962B1 (en) 2007-09-19 2016-01-13 Applied Biosystems, LLC Sirna sequence-independent modification formats for reducing off-target phenotypic effects in rnai, and stabilized forms thereof
US20100136026A1 (en) * 2007-09-26 2010-06-03 Kerr William G Ship Inhibition to Direct Hematopoietic Stem Cells and Induce Extramedullary Hematopoiesis
EP2042592A1 (en) 2007-09-28 2009-04-01 IMBA-Institut für Molekulare Biotechnologie GmbH Methods for modulating the proliferation and differentiation potential of stem cells and progenitor cells
WO2009046397A2 (en) * 2007-10-04 2009-04-09 Board Of Regents, The University Of Texas System Modulating gene expression with agrna and gapmers targeting antisense transcripts
WO2009046426A2 (en) * 2007-10-04 2009-04-09 Isis Pharmaceuticals, Inc. Compounds and methods for improving cellular uptake of oligomeric compounds
US8637478B2 (en) 2007-11-13 2014-01-28 Isis Pharmaceuticals, Inc. Compounds and methods for modulating protein expression
US20100098664A1 (en) * 2007-11-28 2010-04-22 Mathieu Jean-Francois Desclaux Lentiviral vectors allowing RNAi mediated inhibition of GFAP and vimentin expression
EP2245159A2 (en) 2007-12-10 2010-11-03 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of factor vii gene
US20090238772A1 (en) 2007-12-13 2009-09-24 Alnylam Pharmaceuticals, Inc. Methods and compositions for prevention or treatment of rsv infection
US20090176729A1 (en) * 2007-12-14 2009-07-09 Alnylam Pharmaceuticals, Inc. Method of treating neurodegenerative disease
WO2009082817A1 (en) * 2007-12-27 2009-07-09 Protiva Biotherapeutics, Inc. Silencing of polo-like kinase expression using interfering rna
CA2713379A1 (en) * 2008-01-31 2009-11-05 Alnylam Pharmaceuticals, Inc. Optimized methods for delivery of dsrna targeting the pcsk9 gene
WO2009102427A2 (en) * 2008-02-11 2009-08-20 Rxi Pharmaceuticals Corp. Modified rnai polynucleotides and uses thereof
EP2250266A2 (en) 2008-02-12 2010-11-17 Alnylam Pharmaceuticals Inc. Compositions and methods for inhibiting expression of cd45 gene
US20110055965A1 (en) * 2008-02-15 2011-03-03 Hiroshi Abe Cycle single-stranded nucleic acid complex and method for producing the same
CA2716793A1 (en) * 2008-03-05 2009-09-11 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of eg5 and vegf genes
JP5283106B2 (en) * 2008-03-14 2013-09-04 国立大学法人 熊本大学 Hepatitis C virus inhibitor
EP2105145A1 (en) * 2008-03-27 2009-09-30 ETH Zürich Method for muscle-specific delivery lipid-conjugated oligonucleotides
WO2009126651A2 (en) * 2008-04-07 2009-10-15 Malak Kotb Mat ii subunit rnai and therapeutic methods using same
AU2009238175C1 (en) * 2008-04-15 2023-11-30 Arbutus Biopharma Corporation Novel lipid formulations for nucleic acid delivery
WO2009129465A2 (en) 2008-04-17 2009-10-22 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of xbp-1 gene
USRE48948E1 (en) 2008-04-18 2022-03-01 Warsaw Orthopedic, Inc. Clonidine compounds in a biodegradable polymer
US8324366B2 (en) 2008-04-29 2012-12-04 Alnylam Pharmaceuticals, Inc. Compositions and methods for delivering RNAI using lipoproteins
US20090291073A1 (en) * 2008-05-20 2009-11-26 Ward Keith W Compositions Comprising PKC-theta and Methods for Treating or Controlling Ophthalmic Disorders Using Same
US20100009451A1 (en) * 2008-05-30 2010-01-14 Sigma Aldrich Company Compositions and methods for specifically silencing a target nucleic acid
EP2297322A1 (en) 2008-06-04 2011-03-23 The Board of Regents of The University of Texas System Modulation of gene expression through endogenous small rna targeting of gene promoters
CA2635187A1 (en) 2008-06-05 2009-12-05 The Royal Institution For The Advancement Of Learning/Mcgill University Oligonucleotide duplexes and uses thereof
US20100015708A1 (en) * 2008-06-18 2010-01-21 Mdrna, Inc. Ribonucleic acids with non-standard bases and uses thereof
AU2009277172B2 (en) * 2008-07-02 2014-05-29 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
US20110184046A1 (en) * 2008-07-11 2011-07-28 Dinah Wen-Yee Sah Compositions And Methods For Inhibiting Expression Of GSK-3 Genes
WO2010008562A2 (en) 2008-07-16 2010-01-21 Recombinetics Methods and materials for producing transgenic animals
AU2009273878A1 (en) * 2008-07-25 2010-01-28 Alnylam Pharmaceuticals, Inc. Enhancement of siRNA silencing activity using universal bases or mismatches in the sense strand
CN102223896A (en) * 2008-08-07 2011-10-19 西塞医疗中心 Anti-beta-2-microglobulin agents and the use thereof
ES2708944T3 (en) * 2008-09-22 2019-04-12 Dicerna Pharmaceuticals Inc Compositions and methods for the specific inhibition of gene expression by DSRNA having modifications
US10022454B2 (en) 2008-09-23 2018-07-17 Liposciences, Llc Functionalized phosphorodiamites for therapeutic oligonucleotide synthesis
EP2346883B1 (en) 2008-09-23 2016-03-23 Scott G. Petersen Self delivering bio-labile phosphate protected pro-oligos for oligonucleotide based therapeutics and mediating rna interference
JP5529142B2 (en) 2008-09-25 2014-06-25 アルナイラム ファーマシューティカルズ, インコーポレイテッド Lipid formulation composition and method for inhibiting expression of serum amyloid A gene
US8592570B2 (en) 2008-10-06 2013-11-26 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of an RNA from West Nile virus
AU2009303345B2 (en) 2008-10-09 2015-08-20 Arbutus Biopharma Corporation Improved amino lipids and methods for the delivery of nucleic acids
WO2010045384A2 (en) * 2008-10-15 2010-04-22 Somagenics Inc. Short hairpin rnas for inhibition of gene expression
CN104382853A (en) 2008-10-16 2015-03-04 玛瑞纳生物技术有限公司 Processes and Compositions for Liposomal and Efficient Delivery of Gene Silencing Therapeutics
US8168775B2 (en) 2008-10-20 2012-05-01 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin
US20100168205A1 (en) * 2008-10-23 2010-07-01 Alnylam Pharmaceuticals, Inc. Methods and Compositions for Prevention or Treatment of RSV Infection Using Modified Duplex RNA Molecules
BRPI0922355A8 (en) * 2008-12-03 2017-12-12 Marina Biotech Inc NUCLEIC ACIDS, METHODS FOR REDUCING EXPRESSION OF A GENE IN A CELL IN VITRO, USE OF NUCLEIC ACID, RNA COMPLEX AND USE OF RNA COMPLEX
AU2009324534B2 (en) * 2008-12-10 2015-07-30 Alnylam Pharmaceuticals, Inc. GNAQ targeted dsRNA compositions and methods for inhibiting expression
EP2377934A4 (en) * 2008-12-12 2012-09-26 Kureha Corp Pharmaceutical composition for treatment of cancer and asthma
KR20110110776A (en) * 2008-12-18 2011-10-07 다이서나 파마수이티컬, 인크. Extended dicer substrate agents and methods for the specific inhibition of gene expression
US11408003B2 (en) 2008-12-18 2022-08-09 Dicerna Pharmaceuticals, Inc. Extended dicer substrate agents and methods for the specific inhibition of gene expression
EP2381934A2 (en) 2008-12-23 2011-11-02 Carmel - Haifa University Economic Corp Ltd. Improving cognitive function
WO2010083615A1 (en) 2009-01-26 2010-07-29 Protiva Biotherapeutics, Inc. Compositions and methods for silencing apolipoprotein c-iii expression
CA2750459A1 (en) * 2009-02-03 2010-08-12 F. Hoffmann-La Roche Ag Compositions and methods for inhibiting expression of ptp1b genes
EP2408916A2 (en) * 2009-03-19 2012-01-25 Merck Sharp&Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF CONNECTIVE TISSUE GROWTH FACTOR (CTGF) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US20100239632A1 (en) 2009-03-23 2010-09-23 Warsaw Orthopedic, Inc. Drug depots for treatment of pain and inflammation in sinus and nasal cavities or cardiac tissue
FI20090161A0 (en) 2009-04-22 2009-04-22 Faron Pharmaceuticals Oy New cell and therapeutical and diagnostic methods based on it
EP2421972A2 (en) * 2009-04-24 2012-02-29 The Board of Regents of The University of Texas System Modulation of gene expression using oligomers that target gene regions downstream of 3' untranslated regions
WO2010142603A1 (en) 2009-06-08 2010-12-16 Vib Vzw Screening for compounds that modulate gpr3-mediated beta-arrestin signaling and amyloid beta peptide generation
EA201270019A1 (en) 2009-06-15 2012-06-29 Элнилэм Фармасьютикалз, Инк. BENTROVAL RNA INCLUDED IN LIPID COMPOSITION AND WHICH IS THE PCSK9 GENE
US9051567B2 (en) 2009-06-15 2015-06-09 Tekmira Pharmaceuticals Corporation Methods for increasing efficacy of lipid formulated siRNA
CA2765815A1 (en) * 2009-06-26 2010-12-29 Opko Curna, Llc Treatment of down syndrome gene related diseases by inhibition of natural antisense transcript to a down syndrome gene
WO2011000106A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Improved cationic lipids and methods for the delivery of therapeutic agents
CA2767127A1 (en) 2009-07-01 2011-01-06 Protiva Biotherapeutics, Inc. Novel lipid formulations for delivery of therapeutic agents to solid tumors
US9018187B2 (en) 2009-07-01 2015-04-28 Protiva Biotherapeutics, Inc. Cationic lipids and methods for the delivery of therapeutic agents
WO2011005860A2 (en) * 2009-07-07 2011-01-13 Alnylam Pharmaceuticals, Inc. 5' phosphate mimics
EP2454371B1 (en) 2009-07-13 2021-01-20 Somagenics, Inc. Chemical modification of small hairpin rnas for inhibition of gene expression
US8716464B2 (en) * 2009-07-20 2014-05-06 Thomas W. Geisbert Compositions and methods for silencing Ebola virus gene expression
WO2011020023A2 (en) 2009-08-14 2011-02-17 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of a gene from the ebola virus
US8598327B2 (en) * 2009-08-18 2013-12-03 Baxter International Inc. Aptamers to tissue factor pathway inhibitor and their use as bleeding disorder therapeutics
CN107519133A (en) 2009-09-15 2017-12-29 阿尔尼拉姆医药品有限公司 The method of the expression of composition and suppression Eg5 and VEGF genes that lipid is prepared
WO2011038031A1 (en) 2009-09-22 2011-03-31 Alnylam Pharmaceuticals, Inc. Dual targeting sirna agents
US9101643B2 (en) 2009-11-03 2015-08-11 Alnylam Pharmaceuticals, Inc. Lipid formulated compositions and methods for inhibiting expression of transthyretin (TTR)
US9799416B2 (en) * 2009-11-06 2017-10-24 Terrapower, Llc Methods and systems for migrating fuel assemblies in a nuclear fission reactor
US20110124706A1 (en) * 2009-11-25 2011-05-26 Zhigang He SOCS3 Inhibition Promotes CNS Neuron Regeneration
CA2776568A1 (en) 2009-11-26 2011-06-03 Quark Pharmaceuticals, Inc. Sirna compounds comprising terminal substitutions
US8790887B2 (en) 2009-12-04 2014-07-29 Vib Vzw Screening methods for compounds that modulate ARF-6 mediated endosomal redistribution
TWI465238B (en) * 2009-12-09 2014-12-21 Nitto Denko Corp Modulation of hsp47 expression
US8455455B1 (en) 2010-03-31 2013-06-04 Protiva Biotherapeutics, Inc. Compositions and methods for silencing genes involved in hemorrhagic fever
MX2012011222A (en) 2010-04-01 2013-01-18 Centre Nat Rech Scient Compounds and pharmaceutical compositions for the treatment of viral infections.
DK2558578T3 (en) 2010-04-13 2016-01-25 Life Technologies Corp CONFIGURATIONS AND METHODS FOR INHIBITION OF nucleic acid function
WO2011139917A1 (en) 2010-04-29 2011-11-10 Isis Pharmaceuticals, Inc. Modulation of transthyretin expression
GB201010557D0 (en) * 2010-06-23 2010-08-11 Mina Therapeutics Ltd RNA molecules and uses thereof
WO2012000104A1 (en) 2010-06-30 2012-01-05 Protiva Biotherapeutics, Inc. Non-liposomal systems for nucleic acid delivery
EP3372684B1 (en) 2010-08-24 2020-10-07 Sirna Therapeutics, Inc. Single-stranded rnai agents containing an internal, non-nucleic acid spacer
US8663624B2 (en) 2010-10-06 2014-03-04 The Regents Of The University Of California Adeno-associated virus virions with variant capsid and methods of use thereof
WO2012058210A1 (en) 2010-10-29 2012-05-03 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACIDS (siNA)
US20120310140A1 (en) 2010-12-01 2012-12-06 Spinal Modulation, Inc. Directed delivery of agents to neural anatomy
WO2012110500A1 (en) 2011-02-15 2012-08-23 Vib Vzw Means and methods for improvement of synaptic dysfunction disorders
GB201103762D0 (en) 2011-03-07 2011-04-20 Vib Vzw Means and methods for the treatment of neurodegenerative disorders
US10184942B2 (en) 2011-03-17 2019-01-22 University Of South Florida Natriuretic peptide receptor as a biomarker for diagnosis and prognosis of cancer
WO2012123591A1 (en) * 2011-03-17 2012-09-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for targeting nucleic acids to the nucleus
WO2012154321A1 (en) 2011-03-31 2012-11-15 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of viral infections
CA2833870C (en) 2011-04-22 2020-03-10 The Regents Of The University Of California Adeno-associated virus virions with variant capsid and methods of use thereof
US9068184B2 (en) 2011-06-21 2015-06-30 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibition of expression of protein C (PROC) genes
KR102540778B1 (en) 2011-06-21 2023-06-07 알닐람 파마슈티칼스 인코포레이티드 Compositions and methods for inhibition of expression of apolipoprotein c-iii(apoc3) genes
EP2723861A4 (en) 2011-06-21 2014-12-10 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting hepcidin antimicrobial peptide (hamp) or hamp-related gene expression
FI20115640A0 (en) 2011-06-22 2011-06-22 Turun Yliopisto combination therapy
WO2012175735A1 (en) 2011-06-23 2012-12-27 Vib Vzw A20 inhibitors for the treatment of respiratory viral infections
GB201112145D0 (en) 2011-07-15 2011-08-31 Vib Vzw Means and methods for the treatment of pathological angiogenesis
FI20115876A0 (en) 2011-09-06 2011-09-06 Turun Yliopisto Combination therapy
TW201329096A (en) 2011-09-12 2013-07-16 Idenix Pharmaceuticals Inc Substituted carbonyloxymethylphosphoramidate compounds and pharmaceutical compositions for the treatment of viral infections
KR102263352B1 (en) 2011-11-18 2021-06-11 알닐람 파마슈티칼스 인코포레이티드 RNAi AGENTS, COMPOSITIONS AND METHODS OF USE THEREOF FOR TREATING TRANSTHYRETIN (TTR) ASSOCIATED DISEASES
US9506033B2 (en) * 2012-05-22 2016-11-29 University Of Massachusetts Compositions and methods for inducing myoblast differentiation and myotube formation
EP2872631B1 (en) 2012-07-13 2017-05-03 Turun yliopisto Combination therapy
US9315567B2 (en) 2012-08-14 2016-04-19 Ibc Pharmaceuticals, Inc. T-cell redirecting bispecific antibodies for treatment of disease
EP2885313A4 (en) 2012-08-20 2016-03-09 Univ California Polynucleotides having bioreversible groups
US9096853B2 (en) * 2012-09-24 2015-08-04 U.S. Department Of Veterans Affairs Modified siRNA molecules incorporating 5-fluoro-2′-deoxyuridine residues to enhance cytotoxicity
EP2986599A1 (en) 2013-04-17 2016-02-24 Pfizer Inc. N-piperidin-3-ylbenzamide derivatives for treating cardiovascular diseases
CA2907799A1 (en) 2013-05-31 2014-12-04 The Regents Of The University Of California Adeno-associated virus variants and methods of use thereof
WO2015050871A2 (en) 2013-10-04 2015-04-09 Novartis Ag Organic compounds to treat hepatitis b virus
EP3052627B1 (en) 2013-10-04 2018-08-22 Novartis AG Novel formats for organic compounds for use in rna interference
CA3188691A1 (en) 2013-10-04 2015-04-09 Novartis Ag 3'end caps for rnai agents for use in rna interference
WO2015051044A2 (en) 2013-10-04 2015-04-09 Novartis Ag Novel formats for organic compounds for use in rna interference
MX2016010683A (en) 2014-02-21 2017-05-11 Ibc Pharmaceuticals Inc Disease therapy by inducing immune response to trop-2 expressing cells.
CN106029098A (en) 2014-02-25 2016-10-12 免疫医疗公司 Humanized RFB4 anti-CD22 antibody
US10000741B2 (en) 2014-03-17 2018-06-19 Adverum Biotechnologies, Inc. Compositions and methods for enhanced gene expression in cone cells
US9856475B2 (en) 2014-03-25 2018-01-02 Arcturus Therapeutics, Inc. Formulations for treating amyloidosis
JP6771387B2 (en) 2014-03-25 2020-10-21 アークトゥラス・セラピューティクス・インコーポレイテッドArcturus Therapeutics,Inc. Transthyretin allele-selective UNA oligomer for gene silencing
CN106460025A (en) 2014-03-25 2017-02-22 阿克丘勒斯治疗公司 UNA oligomers having reduced off-target effects in gene silencing
PL3185957T3 (en) 2014-08-29 2022-11-14 Alnylam Pharmaceuticals, Inc. Patisiran for use in treating transthyretin mediated amyloidosis
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
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP3221451A1 (en) 2014-11-17 2017-09-27 Alnylam Pharmaceuticals, Inc. Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof
WO2016079317A1 (en) 2014-11-20 2016-05-26 Vib Vzw Means and methods for treatment of early-onset parkinson's disease
KR101668074B1 (en) * 2015-02-12 2016-10-21 전북대학교산학협력단 Composition comprising PKR inhibitor for preventing or treating severe bronchial asthma
CN107405507B (en) 2015-03-02 2022-05-03 阿德夫拉姆生物技术股份有限公司 Compositions and methods for intravitreal delivery of polynucleotides to retinal cones
MX2017012097A (en) 2015-03-24 2018-06-15 Univ California Adeno-associated virus variants and methods of use thereof.
WO2016161299A1 (en) 2015-04-01 2016-10-06 Arcturus Therapeutics, Inc. Therapeutic una oligomers and uses thereof
JP6892433B2 (en) 2015-04-03 2021-06-23 ユニバーシティ・オブ・マサチューセッツUniversity Of Massachusetts Well-stabilized asymmetric SIRNA
EP3277815B1 (en) 2015-04-03 2021-09-22 University of Massachusetts Oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
CA2980337A1 (en) 2015-04-03 2016-10-06 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mrna
ITUB20152371A1 (en) * 2015-07-21 2017-01-21 Univ Degli Studi Di Torino PROCEDURE FOR INDUCING RESISTANCE TO DIFTERIC TOXIN IN HUMAN CELLS, RELATED PRODUCTS AND USES
WO2017015671A1 (en) 2015-07-23 2017-01-26 Arcturus Therapeutics, Inc. Compositions for treating amyloidosis
LT3329002T (en) 2015-07-31 2021-01-11 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof for treating or preventing ttr-associated diseases
CA2995110A1 (en) 2015-08-14 2017-02-23 University Of Massachusetts Bioactive conjugates for oligonucleotide delivery
EP3677909A1 (en) 2015-08-27 2020-07-08 The General Hospital Corporation Methods and compositions for inhibiting detoxification response
CA3011894A1 (en) 2016-01-31 2017-08-03 University Of Massachusetts Branched oligonucleotides
KR102515329B1 (en) 2016-03-07 2023-03-29 애로우헤드 파마슈티컬스 인코포레이티드 Targeting ligands for therapeutic compounds
EP3429567B1 (en) 2016-03-16 2024-01-10 The J. David Gladstone Institutes Methods and compositions for treating obesity and/or diabetes and for identifying candidate treatment agents
MA45470A (en) * 2016-04-01 2019-02-06 Avidity Biosciences Llc KRAS NUCLEIC ACIDS AND THEIR USES
MA45468A (en) * 2016-04-01 2019-02-06 Avidity Biosciences Llc MYC NUCLEIC ACIDS AND USES
MA45471A (en) * 2016-04-01 2019-02-06 Avidity Biosciences Llc PHOSPHATIDYLINOSITOL-3-KINASE NUCLEIC ACIDS AND THEIR USES
EP3827812A1 (en) 2016-07-29 2021-06-02 The Regents of the University of California Adeno-associated virus virions with variant capsid and methods of use thereof
CA3033368A1 (en) 2016-08-12 2018-02-15 University Of Massachusetts Conjugated oligonucleotides
US10294474B2 (en) 2016-09-02 2019-05-21 Arrowhead Pharmaceuticals, Inc. Targeting ligands
AU2017345470B2 (en) 2016-10-19 2023-08-03 Adverum Biotechnologies, Inc. Modified AAV capsids and uses thereof
ES2674128B1 (en) * 2016-12-27 2019-04-10 Univ Salamanca Method for diagnosing allergic sensitization in a subject
US11324820B2 (en) 2017-04-18 2022-05-10 Alnylam Pharmaceuticals, Inc. Methods for the treatment of subjects having a hepatitis b virus (HBV) infection
WO2018224162A1 (en) * 2017-06-09 2018-12-13 Biontech Rna Pharmaceuticals Gmbh Methods for characterizing loss of antigen presentation
CA3064590A1 (en) 2017-06-23 2018-12-27 University Of Massachusetts Two-tailed self-delivering sirna and related methods
WO2019006455A1 (en) 2017-06-30 2019-01-03 Solstice Biologics, Ltd. Chiral phosphoramidite auxiliaries and methods of their use
AU2018301829A1 (en) 2017-07-13 2020-02-27 Alnylam Pharmaceuticals, Inc. Methods for inhibition of HAO1 (hydroxyacid oxidase 1 (glycolate oxidase) gene expression
AU2018324477A1 (en) 2017-08-28 2019-10-17 The Regents Of The University Of California Adeno-associated virus capsid variants and methods of use thereof
MA50267A (en) 2017-09-19 2020-07-29 Alnylam Pharmaceuticals Inc COMPOSITIONS AND METHODS OF TREATMENT OF TRANSTHYRETIN-MEDIA AMYLOSIS (TTR)
WO2019243430A1 (en) * 2018-06-22 2019-12-26 Roche Innovation Center Copenhagen A/S Oligonucleotides for modulating scn9a expression
JP2021533762A (en) 2018-08-10 2021-12-09 ユニバーシティ・オブ・マサチューセッツUniversity Of Massachusetts Modified oligonucleotides that target SNPs
JP2021533767A (en) 2018-08-13 2021-12-09 アルナイラム ファーマシューティカルズ, インコーポレイテッドAlnylam Pharmaceuticals, Inc. Hepatitis B virus (HBV) dsRNA substance composition and its usage
WO2020041769A1 (en) 2018-08-23 2020-02-27 University Of Massachusetts O-methyl rich fully stabilized oligonucleotides
US20220000123A1 (en) 2018-09-26 2022-01-06 Greenlight Biosciences, Inc. Control of coleopteran insects
US20210395738A1 (en) 2018-11-08 2021-12-23 Greenlight Biosciences, Inc. Control of insect infestation
US20230073368A1 (en) 2020-02-11 2023-03-09 Turun Yliopisto Therapy of ras-dependent cancers
JP2023553343A (en) 2020-11-25 2023-12-21 アカゲラ・メディスンズ,インコーポレイテッド Lipid nanoparticles and related methods of use for delivering nucleic acids
EP4359539A1 (en) 2021-06-23 2024-05-01 University Of Massachusetts Optimized anti-flt1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
WO2023014677A1 (en) 2021-08-03 2023-02-09 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof

Citations (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4987071A (en) * 1986-12-03 1991-01-22 University Patents, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US5334711A (en) * 1991-06-20 1994-08-02 Europaisches Laboratorium Fur Molekularbiologie (Embl) Synthetic catalytic oligonucleotide structures
US5364616A (en) * 1992-04-15 1994-11-15 The Procter & Gamble Company Use of H-2 antagonists for treatment of gingivitis
US5519059A (en) * 1994-08-17 1996-05-21 Sawaya; Assad S. Antifungal formulation
US5525468A (en) * 1992-05-14 1996-06-11 Ribozyme Pharmaceuticals, Inc. Assay for Ribozyme target site
US5589332A (en) * 1992-12-04 1996-12-31 Innovir Laboratories, Inc. Ribozyme amplified diagnostics
US5616488A (en) * 1992-12-07 1997-04-01 Ribozyme Pharmaceuticals, Inc. IL-5 targeted ribozymes
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
US5627053A (en) * 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US5631359A (en) * 1994-10-11 1997-05-20 Ribozyme Pharmaceuticals, Inc. Hairpin ribozymes
US5670633A (en) * 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5672695A (en) * 1990-10-12 1997-09-30 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Modified ribozymes
US5716824A (en) * 1995-04-20 1998-02-10 Ribozyme Pharmaceuticals, Inc. 2'-O-alkylthioalkyl and 2-C-alkylthioalkyl-containing enzymatic nucleic acids (ribozymes)
US5719197A (en) * 1988-03-04 1998-02-17 Noven Pharmaceuticals, Inc. Compositions and methods for topical administration of pharmaceutically active agents
US5741679A (en) * 1992-12-04 1998-04-21 Innovir Laboratories, Inc. Regulatable nucleic acid therapeutic and methods of use thereof
US5792847A (en) * 1989-10-24 1998-08-11 Gilead Sciences, Inc. 2' Modified Oligonucleotides
US5801154A (en) * 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
US5814620A (en) * 1993-07-27 1998-09-29 Hybridon, Inc. Inhibition of neovascularization using vegf-specific oligonucleotides
US5849902A (en) * 1996-09-26 1998-12-15 Oligos Etc. Inc. Three component chimeric antisense oligonucleotides
US5854038A (en) * 1993-01-22 1998-12-29 University Research Corporation Localization of a therapeutic agent in a cell in vitro
US5871914A (en) * 1993-06-03 1999-02-16 Intelligene Ltd. Method for detecting a nucleic acid involving the production of a triggering RNA and transcription amplification
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US5902880A (en) * 1994-08-19 1999-05-11 Ribozyme Pharmaceuticals, Inc. RNA polymerase III-based expression of therapeutic RNAs
US5989912A (en) * 1996-11-21 1999-11-23 Oligos Etc. Inc. Three component chimeric antisense oligonucleotides
US5998206A (en) * 1999-02-23 1999-12-07 Isis Pharmaceuticals Inc. Antisense inhibiton of human G-alpha-12 expression
US5998203A (en) * 1996-04-16 1999-12-07 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids containing 5'-and/or 3'-cap structures
US6001311A (en) * 1997-02-05 1999-12-14 Protogene Laboratories, Inc. Apparatus for diverse chemical synthesis using two-dimensional array
US6005087A (en) * 1995-06-06 1999-12-21 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US6025339A (en) * 1995-06-07 2000-02-15 East Carolina University Composition, kit and method for treatment of disorders associated with bronchoconstriction and lung inflammation
US6060456A (en) * 1993-11-16 2000-05-09 Genta Incorporated Chimeric oligonucleoside compounds
US6146886A (en) * 1994-08-19 2000-11-14 Ribozyme Pharmaceuticals, Inc. RNA polymerase III-based expression of therapeutic RNAs
US6159714A (en) * 1993-10-18 2000-12-12 Ribozyme Pharmacueticals, Inc. Catalytic DNA
US6180613B1 (en) * 1994-04-13 2001-01-30 The Rockefeller University AAV-mediated delivery of DNA to cells of the nervous system
US6214805B1 (en) * 1996-02-15 2001-04-10 The United States Of America As Represented By The Department Of Health And Human Services RNase L activators and antisense oligonucleotides effective to treat RSV infections
US6248878B1 (en) * 1996-12-24 2001-06-19 Ribozyme Pharmaceuticals, Inc. Nucleoside analogs
US6300074B1 (en) * 1990-06-11 2001-10-09 Gilead Sciences, Inc. Systematic evolution of ligands by exponential enrichment: Chemi-SELEX
US6346398B1 (en) * 1995-10-26 2002-02-12 Ribozyme Pharmaceuticals, Inc. Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor
US6395713B1 (en) * 1997-07-23 2002-05-28 Ribozyme Pharmaceuticals, Inc. Compositions for the delivery of negatively charged molecules
US20020086356A1 (en) * 2000-03-30 2002-07-04 Whitehead Institute For Biomedical Research RNA sequence-specific mediators of RNA interference
US20020096927A1 (en) * 2001-01-24 2002-07-25 Tsang-Ying Chen Foldable backrest of electric cart
US20020151693A1 (en) * 2000-02-08 2002-10-17 Yale University Nucleic acid catalysts with endonuclease activity
US6506559B1 (en) * 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US20030044188A1 (en) * 2001-08-27 2003-03-06 Coleman Russell A. Method and system for managing replenishment of toners
US20030059944A1 (en) * 2001-09-13 2003-03-27 Carlos Lois-Caballe Method for expression of small antiviral RNA molecules within a cell
US20030064626A1 (en) * 2001-09-28 2003-04-03 Kemmick Dennis L. Connector with staggered contact pattern
US20030064625A1 (en) * 2001-09-28 2003-04-03 Ddk Ltd Multiconductor connector for high-speed transmission signal
US20030064945A1 (en) * 1997-01-31 2003-04-03 Saghir Akhtar Enzymatic nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors
US6573099B2 (en) * 1998-03-20 2003-06-03 Benitec Australia, Ltd. Genetic constructs for delaying or repressing the expression of a target gene
US20030143732A1 (en) * 2001-04-05 2003-07-31 Kathy Fosnaugh RNA interference mediated inhibition of adenosine A1 receptor (ADORA1) gene expression using short interfering RNA
US20030190635A1 (en) * 2002-02-20 2003-10-09 Mcswiggen James A. RNA interference mediated treatment of Alzheimer's disease using short interfering RNA
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)
US20040019001A1 (en) * 2002-02-20 2004-01-29 Mcswiggen James A. RNA interference mediated inhibition of protein typrosine phosphatase-1B (PTP-1B) gene expression using short interfering RNA
US20040161844A1 (en) * 1996-06-06 2004-08-19 Baker Brenda F. Sugar and backbone-surrogate-containing oligomeric compounds and compositions for use in gene modulation
US6824972B2 (en) * 2000-05-22 2004-11-30 Baylor College Of Medicine Diagnosis and treatment of medical conditions associated with defective NFkappa B(NF-κB) activation
US20050020521A1 (en) * 2002-09-25 2005-01-27 University Of Massachusetts In vivo gene silencing by chemically modified and stable siRNA
US20050227256A1 (en) * 2003-11-26 2005-10-13 Gyorgy Hutvagner Sequence-specific inhibition of small RNA function
US7078196B2 (en) * 2000-12-01 2006-07-18 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften, E.V. RNA interference mediating small RNA molecules

Family Cites Families (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2359130A (en) * 1942-02-13 1944-09-26 Gen Electric Electric valve circuits
US2359180A (en) * 1942-08-11 1944-09-26 Gen Motors Corp Dynamic balancer
PT88550A (en) 1987-09-21 1989-07-31 Ml Tecnology Ventures Lp PROCESS FOR THE PREPARATION OF NON-NUCLEOTIDIC LIGACATION REAGENTS FOR NUCLEOTIDIAL PROBES
CA1340323C (en) 1988-09-20 1999-01-19 Arnold E. Hampel Rna catalyst for cleaving specific rna sequences
WO1990014090A1 (en) 1989-05-19 1990-11-29 Hem Research, Inc. SHORT THERAPEUTIC dsRNA OF DEFINED STRUCTURE
JP3058686B2 (en) 1989-08-31 2000-07-04 シティ・オブ・ホープ Chimeric DNA-RNA catalytically active sequence
US5567588A (en) * 1990-06-11 1996-10-22 University Research Corporation Systematic evolution of ligands by exponential enrichment: Solution SELEX
US5652094A (en) 1992-01-31 1997-07-29 University Of Montreal Nucleozymes
WO1993023569A1 (en) 1992-05-11 1993-11-25 Ribozyme Pharmaceuticals, Inc. Method and reagent for inhibiting viral replication
EP0641212A4 (en) 1992-05-14 1997-05-21 Ribozyme Pharm Inc Method and reagent for inhibiting cancer development.
ATE171210T1 (en) 1992-07-02 1998-10-15 Hybridon Inc SELF-STABILIZED OLIGONUCLEOTIDES AS THERAPEUTICS
EP0786522A2 (en) 1992-07-17 1997-07-30 Ribozyme Pharmaceuticals, Inc. Enzymatic RNA molecules for treatment of stenotic conditions
US5320962A (en) * 1992-07-22 1994-06-14 Duke University DNA encoding the human A1 adenosine receptor
US6410322B1 (en) 1993-07-27 2002-06-25 Hybridon Inc Antisense oligonucleotide inhibition of vascular endothelial growth factor expression
WO1995004818A1 (en) 1993-08-06 1995-02-16 Ribozyme Pharmaceuticals, Inc. Method and reagent for inhibiting human immunodeficiency virus replication
JPH09502092A (en) 1993-09-02 1997-03-04 リボザイム・ファーマシューティカルズ・インコーポレイテッド Enzymatic nucleic acid containing non-nucleotide
CA2174339A1 (en) 1993-10-27 1995-05-04 Lech W. Dudycz 2'-amido and 2'-peptido modified oligonucleotides
ATE226254T1 (en) 1993-11-12 2002-11-15 Ribozyme Pharm Inc REAGENT FOR THE TREATMENT OF ARTHRITIC CONDITIONS
US5587471A (en) * 1994-01-11 1996-12-24 Isis Pharmaceuticals, Inc. Method of making oligonucleotide libraries
CA2183992A1 (en) 1994-02-23 1995-08-31 Dan T. Stinchcomb Method and reagent for inhibiting the expression of disease related genes
US5633133A (en) * 1994-07-14 1997-05-27 Long; David M. Ligation with hammerhead ribozymes
US5820873A (en) 1994-09-30 1998-10-13 The University Of British Columbia Polyethylene glycol modified ceramide lipids and liposome uses thereof
US5885613A (en) 1994-09-30 1999-03-23 The University Of British Columbia Bilayer stabilizing components and their use in forming programmable fusogenic liposomes
US5753613A (en) 1994-09-30 1998-05-19 Inex Pharmaceuticals Corporation Compositions for the introduction of polyanionic materials into cells
DE4445700A1 (en) 1994-12-21 1996-06-27 Forschungszentrum Juelich Gmbh Gradiometer
US5994315A (en) * 1995-06-07 1999-11-30 East Carolina University Low adenosine agent, composition, kit and method for treatment of airway disease
EP1108724B1 (en) 1996-01-16 2007-09-19 Sirna Therpeutics, Inc. Synthesis of methoxy nucleosides and enzymatic nucleic acid molecules
WO1998027104A1 (en) 1996-12-19 1998-06-25 Yale University Bioreactive allosteric polynucleotides
EP0948511B1 (en) 1996-12-24 2006-10-18 Sirna Therapeutics, Inc. Chemical synthesis of nucleosides analogs and their incorporation into polynucleotides
AU6591798A (en) 1997-03-31 1998-10-22 Yale University Nucleic acid catalysts
CA2295207A1 (en) 1997-06-19 1998-12-23 Innovir Laboratories, Inc. Hammerhead ribozymes with extended cleavage rule
CA2301166A1 (en) 1997-07-24 1999-02-04 Yuan-Peng Zhang Liposomal compositions for the delivery of nucleic acid catalysts
AU750947C (en) 1997-09-22 2003-05-22 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Nucleic acid catalysts with endonuclease activity
US6617438B1 (en) * 1997-11-05 2003-09-09 Sirna Therapeutics, Inc. Oligoribonucleotides with enzymatic activity
US20030036517A1 (en) 1997-12-05 2003-02-20 Bruce A. Sullenger Nucleic acid mediated rna tagging and rna revision
SK287538B6 (en) 1998-03-20 2011-01-04 Commonwealth Scientific And Industrial Research Organisation Control of gene expression
ATE507299T1 (en) 1998-04-08 2011-05-15 Commw Scient Ind Res Org METHOD AND MEANS FOR OBTAINING ALTERED PHENOTYPES
JP2003525017A (en) 1998-04-20 2003-08-26 リボザイム・ファーマシューティカルズ・インコーポレーテッド Nucleic acid molecules with novel chemical composition that can regulate gene expression
JP2002512794A (en) 1998-04-29 2002-05-08 リボザイム・ファーマシューティカルズ・インコーポレーテッド Nucleotide triphosphates and their incorporation into ribozymes
AR020078A1 (en) 1998-05-26 2002-04-10 Syngenta Participations Ag METHOD FOR CHANGING THE EXPRESSION OF AN OBJECTIVE GENE IN A PLANT CELL
IL126731A0 (en) 1998-10-23 1999-08-17 Intelligene Ltd A method of detection
CA2348779A1 (en) 1998-11-03 2000-05-11 Yale University Multidomain polynucleotide molecular sensors
AU776150B2 (en) 1999-01-28 2004-08-26 Medical College Of Georgia Research Institute, Inc. Composition and method for (in vivo) and (in vitro) attenuation of gene expression using double stranded RNA
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
AU3369900A (en) 1999-02-19 2000-09-04 General Hospital Corporation, The Gene silencing
US6197061B1 (en) * 1999-03-01 2001-03-06 Koichi Masuda In vitro production of transplantable cartilage tissue cohesive cartilage produced thereby, and method for the surgical repair of cartilage damage
JP2000253884A (en) 1999-03-10 2000-09-19 Toagosei Co Ltd Antisense nucleic acid compound
US5998148A (en) * 1999-04-08 1999-12-07 Isis Pharmaceuticals Inc. Antisense modulation of microtubule-associated protein 4 expression
EP2363478B1 (en) 1999-04-21 2019-07-24 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhibiting the function of polynucleotide sequences
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
BR0107613A (en) * 2000-01-12 2002-11-19 Univ Yale Blockade of axon growth mediated by nogo receptor
US6602857B1 (en) 2000-01-18 2003-08-05 Isis Pharmaceuticals, Inc. Antisense modulation of PTP1B expression
AU3974001A (en) * 2000-02-03 2001-08-14 Ali R. Fattaey Method and reagent for the inhibition of checkpoint kinase-1 (chk 1) enzyme
WO2001096584A2 (en) 2000-06-12 2001-12-20 Akkadix Corporation Materials and methods for the control of nematodes
HUP0302779A3 (en) 2000-06-23 2005-12-28 Schering Ag Combinations and compositions which interfere with vegf/vegf and angiopoietin/tie receptor function and their use (ii)
EP1311277A4 (en) 2000-07-18 2004-08-25 Joslin Diabetes Center Inc Methods of modulating fibrosis
US6258601B1 (en) * 2000-09-07 2001-07-10 Isis Pharmaceuticals, Inc. Antisense modulation of ubiquitin protein ligase expression
US6613567B1 (en) 2000-09-15 2003-09-02 Isis Pharmaceuticals, Inc. Antisense inhibition of Her-2 expression
WO2003070910A2 (en) 2002-02-20 2003-08-28 Ribozyme Pharmaceuticals, Incorporated INHIBITION OF VASCULAR ENDOTHELIAL GROWTH FACTOR (VEGF) AND VEGF RECEPTOR GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP1390385A4 (en) 2001-05-29 2004-11-24 Sirna Therapeutics Inc Nucleic acid based modulation of female reproductive diseases and conditions
EP1325955A1 (en) 2002-01-04 2003-07-09 atugen AG Compounds and methods for the identification and/or validation of a target
CA2476530A1 (en) 2002-02-14 2003-08-21 City Of Hope Methods for producing interfering rna molecules in mammalian cells and therapeutic uses for such molecules
DE60327618D1 (en) 2002-03-27 2009-06-25 Aegera Therapeutics Inc ANTISENSE NUCLEOBASIS AND ITS APPLICATIONS AGAINST IAP
WO2004043977A2 (en) 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. 2’-fluoro substituted oligomeric compounds and compositions for use in gene modulations
AU2004210972A1 (en) 2003-02-11 2004-08-26 Immusol Incorporated siRNA libraries optimized for predetermined protein families
US20050182005A1 (en) * 2004-02-13 2005-08-18 Tuschl Thomas H. Anti-microRNA oligonucleotide molecules

Patent Citations (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4987071A (en) * 1986-12-03 1991-01-22 University Patents, Inc. RNA ribozyme polymerases, dephosphorylases, restriction endoribonucleases and methods
US5719197A (en) * 1988-03-04 1998-02-17 Noven Pharmaceuticals, Inc. Compositions and methods for topical administration of pharmaceutically active agents
US5792847A (en) * 1989-10-24 1998-08-11 Gilead Sciences, Inc. 2' Modified Oligonucleotides
US6476205B1 (en) * 1989-10-24 2002-11-05 Isis Pharmaceuticals, Inc. 2′ Modified oligonucleotides
US5670633A (en) * 1990-01-11 1997-09-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US6300074B1 (en) * 1990-06-11 2001-10-09 Gilead Sciences, Inc. Systematic evolution of ligands by exponential enrichment: Chemi-SELEX
US5672695A (en) * 1990-10-12 1997-09-30 Max-Planck-Gesellschaft Zur Forderung Der Wissenschaften E.V. Modified ribozymes
US5334711A (en) * 1991-06-20 1994-08-02 Europaisches Laboratorium Fur Molekularbiologie (Embl) Synthetic catalytic oligonucleotide structures
US5364616A (en) * 1992-04-15 1994-11-15 The Procter & Gamble Company Use of H-2 antagonists for treatment of gingivitis
US5525468A (en) * 1992-05-14 1996-06-11 Ribozyme Pharmaceuticals, Inc. Assay for Ribozyme target site
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)
US5589332A (en) * 1992-12-04 1996-12-31 Innovir Laboratories, Inc. Ribozyme amplified diagnostics
US5741679A (en) * 1992-12-04 1998-04-21 Innovir Laboratories, Inc. Regulatable nucleic acid therapeutic and methods of use thereof
US5834186A (en) * 1992-12-04 1998-11-10 Innovir Laboratories, Inc. Regulatable RNA molecule
US5616488A (en) * 1992-12-07 1997-04-01 Ribozyme Pharmaceuticals, Inc. IL-5 targeted ribozymes
US5854038A (en) * 1993-01-22 1998-12-29 University Research Corporation Localization of a therapeutic agent in a cell in vitro
US5871914A (en) * 1993-06-03 1999-02-16 Intelligene Ltd. Method for detecting a nucleic acid involving the production of a triggering RNA and transcription amplification
US5814620A (en) * 1993-07-27 1998-09-29 Hybridon, Inc. Inhibition of neovascularization using vegf-specific oligonucleotides
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
US5801154A (en) * 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
US6159714A (en) * 1993-10-18 2000-12-12 Ribozyme Pharmacueticals, Inc. Catalytic DNA
US6060456A (en) * 1993-11-16 2000-05-09 Genta Incorporated Chimeric oligonucleoside compounds
US5627053A (en) * 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US6180613B1 (en) * 1994-04-13 2001-01-30 The Rockefeller University AAV-mediated delivery of DNA to cells of the nervous system
US5519059A (en) * 1994-08-17 1996-05-21 Sawaya; Assad S. Antifungal formulation
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
US5631359A (en) * 1994-10-11 1997-05-20 Ribozyme Pharmaceuticals, Inc. Hairpin ribozymes
US5716824A (en) * 1995-04-20 1998-02-10 Ribozyme Pharmaceuticals, Inc. 2'-O-alkylthioalkyl and 2-C-alkylthioalkyl-containing enzymatic nucleic acids (ribozymes)
US6005087A (en) * 1995-06-06 1999-12-21 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US6025339A (en) * 1995-06-07 2000-02-15 East Carolina University Composition, kit and method for treatment of disorders associated with bronchoconstriction and lung inflammation
US6346398B1 (en) * 1995-10-26 2002-02-12 Ribozyme Pharmaceuticals, Inc. Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor
US6214805B1 (en) * 1996-02-15 2001-04-10 The United States Of America As Represented By The Department Of Health And Human Services RNase L activators and antisense oligonucleotides effective to treat RSV infections
US5998203A (en) * 1996-04-16 1999-12-07 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids containing 5'-and/or 3'-cap structures
US5898031A (en) * 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US20040161844A1 (en) * 1996-06-06 2004-08-19 Baker Brenda F. Sugar and backbone-surrogate-containing oligomeric compounds and compositions for use in gene modulation
US6107094A (en) * 1996-06-06 2000-08-22 Isis Pharmaceuticals, Inc. Oligoribonucleotides and ribonucleases for cleaving RNA
US5849902A (en) * 1996-09-26 1998-12-15 Oligos Etc. Inc. Three component chimeric antisense oligonucleotides
US5989912A (en) * 1996-11-21 1999-11-23 Oligos Etc. Inc. Three component chimeric antisense oligonucleotides
US6248878B1 (en) * 1996-12-24 2001-06-19 Ribozyme Pharmaceuticals, Inc. Nucleoside analogs
US20030064945A1 (en) * 1997-01-31 2003-04-03 Saghir Akhtar Enzymatic nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors
US6001311A (en) * 1997-02-05 1999-12-14 Protogene Laboratories, Inc. Apparatus for diverse chemical synthesis using two-dimensional array
US6395713B1 (en) * 1997-07-23 2002-05-28 Ribozyme Pharmaceuticals, Inc. Compositions for the delivery of negatively charged molecules
US6506559B1 (en) * 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6573099B2 (en) * 1998-03-20 2003-06-03 Benitec Australia, Ltd. Genetic constructs for delaying or repressing the expression of a target gene
US5998206A (en) * 1999-02-23 1999-12-07 Isis Pharmaceuticals Inc. Antisense inhibiton of human G-alpha-12 expression
US20020151693A1 (en) * 2000-02-08 2002-10-17 Yale University Nucleic acid catalysts with endonuclease activity
US20020086356A1 (en) * 2000-03-30 2002-07-04 Whitehead Institute For Biomedical Research RNA sequence-specific mediators of RNA interference
US6824972B2 (en) * 2000-05-22 2004-11-30 Baylor College Of Medicine Diagnosis and treatment of medical conditions associated with defective NFkappa B(NF-κB) activation
US7078196B2 (en) * 2000-12-01 2006-07-18 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften, E.V. RNA interference mediating small RNA molecules
US20020096927A1 (en) * 2001-01-24 2002-07-25 Tsang-Ying Chen Foldable backrest of electric cart
US7022828B2 (en) * 2001-04-05 2006-04-04 Sirna Theraputics, Inc. siRNA treatment of diseases or conditions related to levels of IKK-gamma
US20030143732A1 (en) * 2001-04-05 2003-07-31 Kathy Fosnaugh RNA interference mediated inhibition of adenosine A1 receptor (ADORA1) gene expression using short interfering RNA
US20030044188A1 (en) * 2001-08-27 2003-03-06 Coleman Russell A. Method and system for managing replenishment of toners
US20030059944A1 (en) * 2001-09-13 2003-03-27 Carlos Lois-Caballe Method for expression of small antiviral RNA molecules within a cell
US20030064625A1 (en) * 2001-09-28 2003-04-03 Ddk Ltd Multiconductor connector for high-speed transmission signal
US20030064626A1 (en) * 2001-09-28 2003-04-03 Kemmick Dennis L. Connector with staggered contact pattern
US20040019001A1 (en) * 2002-02-20 2004-01-29 Mcswiggen James A. RNA interference mediated inhibition of protein typrosine phosphatase-1B (PTP-1B) gene expression using short interfering RNA
US20030190635A1 (en) * 2002-02-20 2003-10-09 Mcswiggen James A. RNA interference mediated treatment of Alzheimer's disease using short interfering RNA
US20050020521A1 (en) * 2002-09-25 2005-01-27 University Of Massachusetts In vivo gene silencing by chemically modified and stable siRNA
US20050227256A1 (en) * 2003-11-26 2005-10-13 Gyorgy Hutvagner Sequence-specific inhibition of small RNA function

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100311816A1 (en) * 2007-11-30 2010-12-09 Noxxon Pharma Ag MCP-1 binding nucleic acids and use thereof

Also Published As

Publication number Publication date
US20060154271A1 (en) 2006-07-13
US20070026394A1 (en) 2007-02-01
EP1386004A2 (en) 2004-02-04
US20050261212A1 (en) 2005-11-24
WO2002081628A2 (en) 2002-10-17
WO2002081628A8 (en) 2003-08-28
US20030143732A1 (en) 2003-07-31
EP1386004A4 (en) 2005-02-16
US20030148507A1 (en) 2003-08-07
US20030119017A1 (en) 2003-06-26
WO2002081628A3 (en) 2003-02-20
US7022828B2 (en) 2006-04-04

Similar Documents

Publication Publication Date Title
US20030191077A1 (en) Method and reagent for the treatment of asthma and allergic conditions
US6673611B2 (en) Nucleic acid molecules with novel chemical compositions capable of modulating gene expression
US20030124513A1 (en) Enzymatic nucleic acid treatment of diseases or conditions related to levels of HIV
US20030064945A1 (en) Enzymatic nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors
US20030148985A1 (en) Methods and reagents for the inhibition of hepatitis B virus replication
US20030216335A1 (en) Method and reagent for the modulation of female reproductive diseases and conditions
US20040142895A1 (en) Nucleic acid-based modulation of gene expression in the vascular endothelial growth factor pathway
WO2002068637A2 (en) Nucleic acid-based treatment of diseases or conditions related to west nile virus infection
US20040018520A1 (en) Trans-splicing enzymatic nucleic acid mediated biopharmaceutical and protein
US20040006035A1 (en) Nucleic acid mediated disruption of HIV fusogenic peptide interactions
US20030203870A1 (en) Method and reagent for the inhibition of NOGO and NOGO receptor genes
US20040054156A1 (en) Method and reagent for inhibiting hepatitis B viral replication
US20020177568A1 (en) Enzymatic nucleic acid treatment of diseases or conditions related to levels of NF-kappa B
WO2001057206A2 (en) Method and reagent for the inhibition of checkpoint kinase-1 (chk 1) enzyme
US20050209182A1 (en) Nucleic acid mediated inhibition of enterococcus infection and cytolysin toxin activity
US20030068301A1 (en) Method and reagent for inhibiting hepatitis B virus replication
WO2003102131A2 (en) Nucleic acid mediated disruption of hiv fusogenic peptide interactions
US20030087847A1 (en) Method and reagent for the inhibition of checkpoint kinase-1 (Chk1) enzyme
US20030073207A1 (en) Enzymatic nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors
US20030140362A1 (en) In vivo models for screening inhibitors of hepatitis B virus
US20030050259A1 (en) Method and reagent for the treatment of cardiac disease
US20030113891A1 (en) Method and reagent for the inhibition of NOGO and NOGO receptor genes
US20030186909A1 (en) Nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors
US20030064946A1 (en) Method and reagent for the inhibition of calcium activated chloride channel-1 (CLCA-1)
US20030060611A1 (en) Method and reagent for the inhibition of NOGO gene

Legal Events

Date Code Title Description
AS Assignment

Owner name: RIBOZYME PHARMACEUTICALS, INC., COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FOSNAUGH, KATHY;MCSWIGGEN, JAMES A.;REEL/FRAME:013458/0588;SIGNING DATES FROM 20021002 TO 20021007

STCB Information on status: application discontinuation

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