EP0654077A1 - Method and reagent for treatment of animal diseases - Google Patents

Method and reagent for treatment of animal diseases

Info

Publication number
EP0654077A1
EP0654077A1 EP93918144A EP93918144A EP0654077A1 EP 0654077 A1 EP0654077 A1 EP 0654077A1 EP 93918144 A EP93918144 A EP 93918144A EP 93918144 A EP93918144 A EP 93918144A EP 0654077 A1 EP0654077 A1 EP 0654077A1
Authority
EP
European Patent Office
Prior art keywords
ribozyme
rna molecule
rna
ribozymes
enzymatic 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.)
Pending
Application number
EP93918144A
Other languages
German (de)
French (fr)
Other versions
EP0654077A4 (en
Inventor
Sean M. Sullivan
Kenneth G. Draper
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
Application filed by Ribozyme Pharmaceuticals Inc filed Critical Ribozyme Pharmaceuticals Inc
Priority to EP02005642A priority Critical patent/EP1251170A3/en
Priority to EP97101534A priority patent/EP0786522A2/en
Publication of EP0654077A1 publication Critical patent/EP0654077A1/en
Publication of EP0654077A4 publication Critical patent/EP0654077A4/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N15/1135Non-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 oncogenes or tumor suppressor genes
    • 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
    • C12N15/1136Non-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 growth factors, growth regulators, cytokines, lymphokines or hormones
    • 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
    • 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
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6489Metalloendopeptidases (3.4.24)
    • C12N9/6491Matrix metalloproteases [MMP's], e.g. interstitial collagenase (3.4.24.7); Stromelysins (3.4.24.17; 3.2.1.22); Matrilysin (3.4.24.23)
    • 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
    • C12N2310/11Antisense
    • C12N2310/111Antisense spanning the whole gene, or a large part of it
    • 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
    • C12N2310/12Type of nucleic acid catalytic nucleic acids, e.g. ribozymes
    • C12N2310/122Hairpin
    • 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
    • C12N2310/123Hepatitis delta
    • 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
    • C12N2310/126Type of nucleic acid catalytic nucleic acids, e.g. ribozymes involving RNAse P

Definitions

  • This invention relates to methods for inhibition of various animal diseases, including: a) inflammatory disease, in particular the inhibition of genetic expression which leads to the reduction or elimination of immune cell proliferation in inflammation or pre- inflammatory conditions; b) osteoarthritis, in particular, inhibition of genetic expression which leads to a reduction or elimination of extracellular matrix digestion by matrix etalloproteinases; c) a stenotic condition, in particular the inhibition of genetic expression which leads to the reduction or elimination of cellular proliferation in the areas of restenotic plaques; and d) cardiovascular disease such as hypertension, and in particular, those mediated by angiotensin converting enzyme activity.
  • Inflammatory disease in particular the inhibition of genetic expression which leads to the reduction or elimination of immune cell proliferation in inflammation or pre- inflammatory conditions
  • osteoarthritis in particular, inhibition of genetic expression which leads to a reduction or elimination of extracellular matrix digestion by matrix etalloproteinases
  • a stenotic condition in particular the inhibition of genetic expression which leads to the reduction or elimination of cellular proliferation in the areas of rest
  • Inflammation is a tissue reaction to irritation, infection or injury marked by localized heat, swelling, redness, pain and sometimes loss of function.
  • the afflicted cells synthesize and/or release chemicals (cytokines and cellular adhesion molecules) which attract lymphoid cells and accelerate cell growth which is required for healing.
  • the infiltration of lymphoid cells to the wound leads to the release of more biological response modifying molecules, which include vasoregulatory substances such as bradykinin and immunoregulatory molecules exemplified by the interleukin family of proteins.
  • vasoregulatory substances such as bradykinin and immunoregulatory molecules exemplified by the interleukin family of proteins.
  • a persistent inflammatory condition may be acute or chronic and is generically referred to as inflammatory disease.
  • Inflammatory disease is exemplified by psoriasis, which is a skin disease caused by excessive growth of basal keratinocytes in the epidermis. It is believed that this uncontrolled growth of the keratinocytes results from stimulation by infiltrating T-cells.
  • the conversion of normal skin to a psoriatic lesion is initiated by injury to the tissue and subsequent recognition of the injury by circulating immune cells.
  • An acute inflammatory reaction ensues and results in the release of various cytokines and growth factors, including tumor necrosis factor alpha
  • TNF- ⁇ interleukin-1
  • IL-1 interleukin-1
  • Induction of the acute phase inflammatory reactants results in the production of IL-1 and TNF-alpha; both of these molecules induce the appearance of the T-cell homing proteins, I-CAM, ELAM-1 and VCAM-1 on the cell surface of keratinocytes.
  • TNF- ⁇ also induces the production of TGF- ⁇ , IL-6 and IL-8 by the keratinocytes.
  • the cytokines and growth factors produced by the keratinocytes and infiltrating leukocytes cause uncontrolled keratinocyte proliferation. As the basal layer keratinocytes proliferate, the suprabasal keratinocytes are pushed closer to the surface of the skin where they normally form the cornified envelope layer and the stratum corneum.
  • the abnormal rate of proliferation by the basal keratinocytes results in improper formation of the epidermis and stratum corneum and the ultimate development of the red, scaly appearance which is characteristic of psoriatic skin.
  • the lack of keratinocyte growth regulation has been attributed to an unidentified mutation in a single gene or group of genes in which the keratinocytes no longer respond to the termination signal for wound healing.
  • corticosteroids There are 3 million patients in the United States afflicted with psoriasis. The available treatments for psoriasis are corticosteroids. The most widely prescribed are TEMOVATE (clobetasol propionate) , LIDEX (fluocinonide) , DIPROLENE (betamethasone propionate) , PSORCON (diflorasone diacetate) and TRIAMCINOLONE formulated for topical application. The mechanism of action of corticosteroids is multifactorial and probably not due to simple inhibition of cell replication. This is a palliative therapy because the underlying cause of the disease remains, and upon discontinuation of the treatment the disease returns.
  • Corticosteroids are not recommended for prolonged treatments or when treatment of large and/or inflamed areas is required.
  • Alternative treatments include retinoids, such as etretinate, which has been approved for treatment of severe, refractory psoriasis.
  • Alternative retinoid-based treatments are in advanced clinical trials. Retinoids act by converting keratinocytes to a differentiated state and restoration of normal skin development. Immunosuppressive drugs such as cyclosporine are also in the advanced stages of clinical trials.
  • Lymphocytes are present at the sites of tissue damage, but their role may be as mediators to amplify the eosinophilic response.
  • interleukin-5 which is released by T-lymphocytes, is important in retaining and priming eosinophil action in the airway.
  • osteoarthritis is a slowly progressive disease characterized by degeneration of articular cartilage with proliferation and remodeling of subchondral bone. It presents with a clinical picture of pain, deformity, and loss of joint motion.
  • Rheumatoid arthritis is a chronic systemic inflammatory disease. Rheumatoid arthritis may be mild and relapsing or severe and progressive, leading to joint deformity and incapacitation.
  • Arthritis is the major contributor to functional impairment among the older population. It is the major cause of disability and accounts for a large proportion of the hospitalizations and health care expenditures of the elderly. Arthritis is estimated to be the principal cause of total incapacitation for about one million persons aged 55 and older and is thought to be an important contributing cause for about one million more. Estimating the incidence of osteoarthritis is difficult for several reasons. First, osteoarthritis is diagnosed objectively on the basis of reading radiographs, but many people with radiologic evidence of disease have no obvious symptoms. Second, the estimates of prevalence are based upon clinical evaluations because radiographic data is not available for all afflicted joints.
  • rheumatoid arthritis has a world-wide distribution and affects all racial and ethnic groups. The exact prevalence in the US is unknown but has been estimated to range between 0.5% and 1.5%. Rheumatoid arthritis occurs at all age levels and generally increases in prevalence with advancing age. It is 2-3 times more prevalent in women than in men and peak incidence occurs between 40-60 years of age. In addition to i munological factors, environmental, occupational and psychosocial factors have been studied for potential etiologic roles in the disease.
  • the extracellular matrix of multicellular organisms plays an important role in the formation and maintenance of tissues.
  • the meshwork of the extracellular matrix is deposited by resident cells and provides a framework for cell adhesion and migration, as well as a permeability barrier in cell-cell communication.
  • Connective tissue turnover during normal growth and development or under pathological conditions is thought to be mediated by a family of neutral metalloproteinases, which are zinc-containing enzymes that require calcium for full activity.
  • the regulation of metalloproteinase expression is cell-type specific and may vary among species.
  • interstitial collagenase The best characterized of the matrix metalloproteinases, interstitial collagenase (MMP-1) , is specific for collagen types I, II, and III. MMP-1 cleaves all three ⁇ chains of the triple helix at a single point initiating sequential breakdown of the interstitial collagens. Interstitial collagenase activity has been observed in rheumatoid synovial cells as well as in the synovial fluid of patients with inflammatory arthritis.
  • Gelatinases (MMP-2) represent a subgroup of the metalloproteinases consisting of two distinct gene products; a 70 kDa gelatinase expressed by most connective tissue cells, and a 92 kDa gelatinase expressed by inflammatory phagocytes and tumor cells.
  • the larger enzyme is expressed by macrophages, SV-40 transformed fibroblasts, and neutrophils.
  • the smaller enzyme is secreted by H-ras transformed bronchial epithelial cells and tumor cells, as well as normal human skin fibroblasts. These enzymes degrade gelatin (denatured collagen) as well as native collagen type XI.
  • Stromelysin (MMP-3) has a wide spectrum of action on molecules composing the extracellular matrix. It digests proteoglycans, fibronectin, laminin, type IV and IX collagens and gelatin, and can remove the N-terminal propeptide region from procollagen, thus activating the collagenase. It has been found in human cartilage extracts, rheumatoid synovial cells, and in the synovium and chondrocytes of joints in rats with collagen-induced arthritis.
  • tissue growth factor- ⁇ has been shown to block epidermal growth factor (EGF) induction of stromelysin synthesis in vitro.
  • Experimental protocols involving gene therapy approaches include the controlled expression of the metalloproteinase inhibitors TIMP-1 and TIMP-2. Of the latter three approaches, only gamma-interferon treatment is currently feasible in a clinical application.
  • Stenosis is the occurrence of a blockage in a blood vessel. Such blockages may lead to impairment of functions or even death, dependent upon which vessel it occurs in and its size. These may be prevented as described below.
  • Restenosis is a disease state which occurs as a sequelae to percutaneous transluminal angioplasty (PCTA) or coronary artery bypass grafting (CABG) treatments of cardiovascular disease.
  • PCTA percutaneous transluminal angioplasty
  • CABG coronary artery bypass grafting
  • Acute or immediate restenosis occurs in approximately 10% of the patients undergoing PCTA and slower-developing restenosis (six month onset) occurs in approximately 30% of those patients.
  • the estimated number of PCTA patients in 1990 was 300,000 - 400,000. Thus, there are about 150,000 cases of restenosis per year in the United States.
  • the presently preferred chemotherapeutic treatment of patients is the use of streptokinase, urokinase or other thrombolytic compounds, such as fish oil, anticoagulants, ACE (angiotensin converting enzyme) inhibitors, aspirin and cholesterol lowering compounds; alternative treatment includes the surgical incorporation of endoluminal stents. It is reported that none of the current therapies have significantly impacted the rates of restenosis occurrence. A number of compounds are currently in preclinical evaluations. Platelet inhibitors include GR32191, Sultroban, Ketanserin, and fish oil.
  • Angiopeptin is being tested as a growth factor inhibitor and Lovostatin, Enoxaparin, RD Heparin, Cilazapril and Fosinopril are being investigated as smooth muscle cell proliferation inhibitors. While platelet inhibitors are being tested for prevention of restenosis, it appears that these compounds will not be efficacious as short-term treatments.
  • One of the biggest problems with current therapies is the occurrence of pharmacologic side-effects. These effects not only create other physiological problems, but also decrease the levels of patient compliance, thereby reducing the therapeutic efficacy of the treatments.
  • the proliferation of antherophils may be induced through a host of genetic activations, but the best candidate for targeting smooth muscle proliferation is the c-myb gene.
  • the c-myb protein binds DNA and activates DNA replication and cellular growth.
  • the role of c-myb in smooth muscle cell replication has been documented in bovine cells, and the expression of c-myb has been shown to activate cellular replication in chicken embryo fibroblasts and human T-lymphocytes.
  • NF-KB cascade NF-KB protein activates cellular transcription and induces increases in cellular synthetic pathways. In a resting cell, this protein is found in the cytoplasm, complexed with its inhibitor, 1KB.
  • NF-KB NF-KB protein
  • the complex Upon phosphorylation of the 1KB molecule, the complex dissociates and NFKB is released for transport to the nucleus, where it binds DNA and induces transcriptional activity in (NF-KB) -responsive genes.
  • One of the (NF-KB) -responsive genes is the NF-KB gene.
  • release of the NF-KB protein from the complex results in a cascade of gene expression which is auto- induced.
  • the invention features use of ribozy es to treat or prevent various animal diseases, in particular, those human diseases noted above.
  • psoriasis which can be treated, e.g. , by inhibiting the synthesis of tumor necrosis factor in activated lymphocytes, and basal keratinocytes.
  • the invention also features use of ribozymes to treat chronic asthma, e.g. , by inhibiting the synthesis of IL-5 in lymphocytes and preventing the recruitment and activation of eosinophils. Cleavage of targeted mRNAs (tumor necrosis factor and IL-5 mRNAs) expressed in keratinocytes, T-lymphocytes, monocytes or macrophages inhibits the synthesis of tumor necrosis factor and IL-5, respectively.
  • targeted mRNAs tumor necrosis factor and IL-5 mRNAs
  • cytokines may also be involved in the activation of inflammation in asthmatic patients, including platelet activating factor, IL-1, IL-3, IL-4, GM-CSF, TNF ⁇ , gamma interferon, ILAM-1, ELAM-1 and EoCSF.
  • platelet activating factor IL-1, IL-3, IL-4, GM-CSF, TNF ⁇ , gamma interferon, ILAM-1, ELAM-1 and EoCSF.
  • any cellular receptors which mediate the activities of the cytokines are also good targets for intervention in inflammatory diseases. These targets include, but are not limited to, the IL-1R and TNF ⁇ R on keratinocytes, epithelial and endothelial cells in airways. Recent data suggest that certain neuropeptides may play a role in asthmatic symptoms.
  • peptides include substance P, neurokinin A and calcitonin-gene-related peptides.
  • target genes may have more general roles in inflammatory diseases, but are currently assumed to have a role only in asthma.
  • Other genes which are considered to play a role in asthma are the c-myb and c-myc genes, which may be triggered to induce endothelial cell proliferation and contribute to blockage of the airways.
  • ribozymes which will reduce the risk or occurrence of inflammatory disease, such as the interleukins (1, 3, 4, 6, and 8), glycerol transferase, selectins (E-selectin, MEL-14) , cell adhesion molecules (ICAM-1, ELAM-1, VCAM-1, GMP-140, MAM), TGF- ⁇ , IL-1R, TGFCR, EoCSF, ⁇ -, ⁇ - or ⁇ -interferon, EoCSF, GM-CSF and protein kinase C (PKC) .
  • interleukins 1, 3, 4, 6, and 8
  • selectins E-selectin, MEL-14
  • IAM cell adhesion molecules
  • TGF- ⁇ IL-1R
  • TGFCR protein kinase C
  • EoCSF protein kinase C
  • a ribozyme to TNF ⁇ nucleotides 374 to 393 can be use to inhibit TNF ⁇ protein production.
  • This region may not a very good region to target because of secondary RNA structure in this region.
  • An adjoining region which begins at nucleotide 380 in the previous reference and extends to nucleotide 412 (408-440, our sequence numbers see below) appears to contain a relatively more accessible RNA structure, and therefore represents an improved target over the region between nucleotides 374-393 (402-421, our sequence numbers) .
  • Another disease is arthritis, which can be treated by inhibition of collagenase and stromelysin production in the synovial membrane of joints.
  • Ribozyme treatment can be a partner to current treatments which primarily target immune cells reacting to pre-existing tissue damage. Early ribozyme treatment which reduces the collagenase or stromelysin-induced damage can be followed by treatment with the anti-inflammatories or retinoids, if necessary. In this manner, expression of the proteinases can be controlled at both transcriptional and translational levels. Ribozyme treatment can be given to patients expressing radiological signs of osteoarthritis prior to the expression of clinical symptoms. Ribozyme treatment can impact the expression of stromelysin without introducing the non-specific effects upon gene expression which accompany treatment with the retinoids and dexamethasone.
  • stromelysin to activate procollagenase indicates that a ribozyme which reduces stromelysin expression can also be used in the treatment of both osteoarthritis (which is primarily a stromelysin- associated pathology) and rheumatoid arthritis (which is primarily related to enhanced collagenase activity) . While a number of cytokines and growth factors induce metalloproteinase activities during wound healing and tissue injury of a pre-osteoarthritic condition, these molecules are not preferred targets for therapeutic intervention. Primary emphasis is placed upon inhibiting the molecules which are responsible for the disruption of the extracellular matrix, because most people will be presenting radiologic or clinical symptoms prior to treatment.
  • the most versatile of the metalloproteinases is stromelysin. Additionally, this molecule can activate procollagenase, which in turn causes further damage to the collagen backbone of the extracellular matrix.
  • TIMPs tissue inhibitors of MMP
  • the invention features use of ribozymes to treat or prevent arthritis, particularly osteoarthritis, e.g. , by inhibiting the synthesis of the prostromelysin molecule in synovial cells, or by inhibition of other matrix metalloproteinases discussed above.
  • Cleavage of targeted mRNAs (stromelysin mRNAs) expressed in macrophages, neutrophils and synovial cells represses the synthesis of the zymogen form of stromelysin, prostromelysin.
  • ribozymes which will reduce the risk or occurrence of pathologic degradation of the extracellular matrix such as the collagenase and gelatinase metalloproteinases, other proteinases which can activate the proenzyme forms of the metalloproteinases in synovial fluid or cartilaginous cells, cytokines or growth factors which activate the expression of the metalloproteinases and adhesion molecules which attract macrophage and neutrophils to the areas of tissue injury.
  • Another disease is stenosis, which is treated, e.g. , by inhibiting the activation of smooth muscle proliferation by inhibiting the expression of the cellular c-myb gene.
  • Cleavage of targeted mRNAs c-myb mRNAs expressed in endothelial cells and smooth muscle cells represses activation of cellular replication and abnormal proliferation of the smooth muscle or endothelial cells.
  • Other potential targets suitable for treatment with ribozymes, which will reduce the risk or occurrence of cellular proliferation in the areas of restenotic risk include mRNAs encoding TGF- ⁇ , NF-KB, PDGF, bFGF, endothelium-derived relaxing factor, CGRP, and angiotensin II.
  • cardiovascular disease which can be treated, e.g. , by inhibiting the activation of angiotensin by angiotensin converting enzyme (ACE) , or by inhibition of the activity of endothelin converting enzyme (ECE) .
  • ACE angiotensin converting enzyme
  • ECE endothelin converting enzyme
  • ribozymes which will reduce the risk or occurrence of cardiovascular disease, such as mRNAs encoding HMG CoA reductase, renin, bradykinin, plasminogen, factors IX, X and II, 2-5A synthetase, ADH and fibrinogen.
  • Ribozymes are RNA molecules having an enzymatic activity which is able to repeatedly cleave other separate RNA molecules in a nucleotide base sequence specific manner. It is alleged that such enzymatic R ⁇ A molecules can be targeted to virtually any R ⁇ A transcript and efficient cleavage has been achieved in vitro . Kim et al., 84 Proc. ⁇ atl. Acad. Sci. USA 8788, 1987; Haseloff and Gerlach, 334 Nature 585, 1988; Cech, 260 JAMA 3030, 1988; and Jefferies et al., 17 Nucleic Acids Research 1371, 1989.
  • Ribozymes act by first binding to a target RNA. Such binding occurs through the target RNA binding portion of a ribozyme which is held in close proximity to an enzymatic portion of the RNA which acts to cleave the target RNA. Thus, the ribozyme 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 a ribozyme 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.
  • ribozyme The enzymatic nature of a ribozyme is advantageous over other technologies, such as antisense technology (where a nucleic acid molecule simply binds to a nucleic acid target to block its translation) since the effective concentration of ribozyme necessary to effect a therapeutic treatment is lower than that of an antisense oligonucleotide.
  • This advantage reflects the ability of the ribozyme to act enzymatically.
  • a single ribozyme molecule is able to cleave many molecules of target RNA.
  • the ribozyme is a highly specific inhibitor, with the specificity of inhibition depending not only on the base pairing mechanism of binding, but also on the mechanism by which the molecule inhibits the expression of the RNA to which it binds. That is, the inhibition is caused by cleavage of the RNA target and so specificity is defined as the ratio of the rate of cleavage of the targeted RNA over the rate of cleavage of non-targeted RNA. This cleavage mechanism is dependent upon factors additional to those involved in base pairing. Thus, it is thought that the specificity of action of a ribozyme is greater than that of antisense oligonucleotide binding the same R ⁇ A site.
  • This class of chemicals exhibits a high degree of specificity for cleavage of the intended target mR ⁇ A. Consequently, the ribozyme agent will only affect cells expressing that particular gene, and will not be toxic to normal tissues.
  • the invention can be used to treat or prevent (prophylactically) psoriasis, asthma and other inflammatory diseases, and restenosis or other cardiovascular conditions, including hypertension.
  • the preferred administration protocol is in vivo administration to reduce the level of those genes and the encoded mR ⁇ As noted above.
  • the invention features an enzymatic R ⁇ A molecule (or ribozyme) which cleaves mR ⁇ A associated with development or maintenance of a psoriatic or asthmatic condition, e.g. , mR ⁇ A encoding T ⁇ F- ⁇ , IL-5, IL-1, IL-3, IL-4, IL-6, IL-8, glycerol transferase, selectins, E-selectin, MEL-14, ICAM-1, ELAM-1, VCAM-1, GMP-140, MAM, TGF ⁇ , T ⁇ F ⁇ R, IL-lR, ⁇ -, ⁇ - or ⁇ -interferon, GM-CSF and protein kinase C, and in particular, those mRNA targets disclosed in Table 1.
  • the invention features and enzymatic RNA molecule which cleaves mRNA associated with development and maintenance of osteoarthritis or other pathological conditions which are mediated by metalloproteinase activation.
  • the preferred administration protocol is in vivo administration to reduce the level of stromelysin activity.
  • the invention features an enzymatic RNA molecule (or ribozyme) which cleaves mRNA associated with development or maintenance of an arthritic condition, e.g. , mRNA encoding stromelysin, and in particular, those mR ⁇ A targets disclosed in Table 2.
  • mRNA associated with development or maintenance of an arthritic condition e.g. , mRNA encoding stromelysin, and in particular, those mR ⁇ A targets disclosed in Table 2.
  • the invention features an enzymatic RNA molecule (or ribozyme) which cleaves mRNA associated with development or maintenance of a restenotic condition, e.g. , mRNA encoding c-myb (or other mRNAs noted above) , and in particular, those mRNA targets disclosed in Table 3.
  • a restenotic condition e.g. , mRNA encoding c-myb (or other mRNAs noted above)
  • the invention features an enzymatic RNA molecule (or ribozyme) which cleaves mRNA associated with development or maintenance of a cardiovascular condition, e.g. , mRNA encoding ACE or
  • enzymatic RNA molecule an RNA molecule which has complementarity in a substrate binding region to a specified mRNA target, and also has an enzymatic activity which is active to specifically cleave that mRNA. That is, the enzymatic R ⁇ A molecule is able to intermolecularly cleave mR ⁇ A and thereby inactivate a target mR ⁇ A molecule.
  • This complementarity functions to allow sufficient hybridization of the enzymatic R ⁇ A molecule to the target R ⁇ A to allow the cleavage to occur.
  • One hundred percent complementarity is preferred, but complementarity as low as 50-75% may also be useful in this invention. For in vivo treatment, complementarity between 30 and 45 bases is preferred.
  • the enzymatic RNA molecule is formed in a hammerhead motif, but may also be formed in the motif of a hairpin, hepatitis delta virus, group I intron or RNaseP-like RNA (in association with an RNA guide sequence) .
  • hammerhead motifs are described by Rossi et al., 8 Aids Research and Human Retroviruses 183, 1992, of hairpin motifs by Hampel and Tritz, 28 Biochemistry 4929, 1989; and Hampel et al., 18 Nucleic Acids Research 299, 1990, and an example of the hepatitis delta virus motif is described by Perrotta and Been, 31 Biochemistry 16,
  • the invention features a mammalian cell which includes an enzymatic RNA molecule as described above.
  • the mammalian cell is a human cell.
  • the invention features an expression vector which includes nucleic acid encoding an enzymatic RNA molecule described above, located in the vector, e.g. , in a manner which allows expression of that enzymatic RNA molecule within a mammalian cell.
  • the invention features a method for treatment of a disease noted above by administering to a patient an enzymatic RNA molecule as described above.
  • the invention provides a class of chemical cleaving agents which exhibit a high degree of specificity for the mRNA causative of a psoriatic or arthritic or cardiovascular condition. Such a condition includes any measurable indication of susceptibility to cardiac problems, and thus includes predisposition to such conditions or cardiovascular disease.
  • Such enzymatic RNA molecules can be delivered exogenously or endogenously to infected cells.
  • the small size (less than 40 nucleotides, preferably between 32 and 36 nucleotides in length) of the molecule allows the cost of treatment to be reduced.
  • ribozyme delivered for any type of treatment reported to date is an in vi tro transcript having a length of 142 nucleotides. Synthesis of ribozymes greater than 100 nucleotides in length is very difficult using automated methods, and the therapeutic cost of such molecules is prohibitive. Delivery of ribozymes by expression vectors is primarily feasible using only ex vivo treatments. This limits the utility of this approach.
  • an alternative approach uses smaller ribozyme motifs (e.g. , of the hammerhead structure, shown generally in Fig. 1) and exogenous delivery. The simple structure of these molecules also increases the ability of the ribozyme to invade targeted regions of the mRNA structure.
  • RNA molecules of this invention can be used to treat psoriatic or pre-psoriatic, asthmatic or pre-asthmatic, arthritic or prearthritic, stenotic or prestenotic conditions. Such treatment can also be extended to other related genes in nonhuman primates. Affected animals can be treated at the time of disease risk or detection, or in a prophylactic manner. This timing of treatment will reduce the chance of further disease damage.
  • Ribozymes of this invention may be used as diagnostic tools to examine genetic drift and mutations within diseased cells.
  • the close relationship between ribozyme activity and the structure of the target RNA allows the detection of mutations in any region of the molecule which alters the base-pairing and three- dimensional structure of the target RNA.
  • By using multiple ribozymes described in this invention one may 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 ribozymes may 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 may be defined as important mediators of the disease.
  • Fig. 1 is a diagrammatic representation of a hammerhead motif ribozyme showing stems I, II and III (marked (I) , (II) and (III) respectively) interacting with a target region. The 5' and 3' ends of both ribozyme and target are shown. Dashes indicate base- paired nucleotides. Target Sites
  • Ribozymes targeting selected regions of mRNA associated with a selected disease are chosen to cleave the target RNA in a manner which preferably inhibits translation of the RNA. Genes are selected such that inhibition of translation will preferably inhibit cell replication, e.g. , by inhibiting production of a necessary protein. Selection of effective target sites within these critical regions of mRNA entails testing the accessibility of the target RNA to hybridization with various oligonucleotide probes. These studies can be performed using RNA probes and assaying accessibility by cleaving the hybrid molecule with RNaseH (see below) .
  • ribozyme probes designed from secondary structure predictions of the mRNAs, and assaying cleavage products by polyacrylamide gel electrophoresis (PAGE) , to detect the presence of cleaved and uncleaved molecules.
  • PAGE polyacrylamide gel electrophoresis
  • the method involves identifying potential cleavage sites for a hammerhead ribozyme, and then testing each of these sites to determine their suitability as targets by ensuring that secondary structure formation is minimal.
  • the mRNA sequences are compared in an appropriate target region. Putative ribozyme cleavage sites are found. These sites represent the preferable sites for hammerhead ribozyme cleavage within these target mRNAs.
  • Short RNA substrates corresponding to each of the mRNA sites are designed.
  • Each substrate is composed of two to three nucleotides at the 5' and 3' ends that will not base pair with a corresponding ribozyme recognition region.
  • the unpaired regions flanked a central region of 12-14 nucleotides to which comple ⁇ mentary arms in the ribozyme are designed.
  • the structure of each substrate sequence is predicted using a standard PC fold computer program. Sequences which give a positive free energy of binding are accepted. Sequences which give a negative free energy are modified by trimming one or two bases from each of the ends. If the modified sequences are still predicted to have a strong secondary structure, they are rejected.
  • ribozymes are designed to each of the RNA substrates. Ribozyme folding is also analyzed using PC fold.
  • Ribozyme molecules are sought which form hammerhead motif stem II (see Fig. 1) regions and contain flanking arms which are devoid of intramolecular base pairing. Often the ribozymes are modified by trimming a base from the ends of the ribozyme, or by introducing additional base pairs in stem II to achieve the desired fold. Ribozymes with incorrect folding are rejected. After substrate/ribozyme pairs are found to contain correct intramolecular structures, the molecules are folded together to predict intermolecular interactions. A schematic representation of a ribozyme with its coordinate base pairing to its cognate target sequence is shown in Fig. 1.
  • ribonuclease H assay provides a quick test of the use of the target site without requiring synthesis of a ribozyme. It can be used to screen for sites most suited for ribozyme attack.
  • Ribozymes useful in this invention can be produced by gene transcription as described by Cech, supra, or by chemical synthesis. Chemical synthesis of RNA is similar to that for DNA synthesis.
  • RNA requires a different protecting group strategy to deal with selective 3'-5' internucleotide bond formation, and with RNA susceptibility to degradation in the presence of bases.
  • the recently developed method of RNA synthesis utilizing the t-butyldimethylsilyl group for the protection of the 2' hydroxyl is the most reliable method for synthesis of ribozymes.
  • the method reproducibly yields RNA with the correct 3'-5' internucleotide linkages, with average coupling yields in excess of 99%, and requires only a two-step deprotection of the polymer.
  • a method, based upon H-phosphonate chemistry of phosphoroamidites gives a relatively lower coupling efficiency than a method based upon phosphoroamidite chemistry. This is a problem for synthesis of DNA as well.
  • a promising approach to scale-up of automatic oligonucleotide synthesis has been described recently for the H-phosphonates.
  • a combination of a proper coupling time and additional capping of "failure" sequences gave high yields in the synthesis of oligodeoxynucleotides in scales in the range of 14 ⁇ moles with as little as 2 equivalents of a monomer in the coupling step.
  • Another alternative approach is to use soluble polymeric supports (e.g.
  • ribozyme structure can be made to enhance the utility of ribozymes. Such modifications will enhance shelf-life, half-life in vi tro, stability, and ease of introduction of such ribozymes to the target site, e.g. , to enhance penetration of cellular membranes, and confer the ability to recognize and bind to targeted cells.
  • Exogenous delivery of ribozymes benefits from chemical modification of the backbone, e.g. , by the overall negative charge of the ribozyme molecule being reduced to facilitate diffusion across the cell membrane.
  • the present strategies for reducing the oligonucleotide charge include: modification of internucleotide linkages by methylphosphonates, use of phosphoramidites, linking oligonucleotides to positively charged molecules, and creating complex packages composed of oligonucleotides, lipids and specific receptors or effectors for targeted cells. Examples of such modifications include sulfur-containing ribozymes containing phosphorothioates and phosphorodithioates as internucleotide linkages in RNA.
  • Ribozymes may also contain ribose modified ribonucleotides.
  • Pyrimidine analogues are prepared from uridine using a procedure employing diethylamino sulphur trifluoride (DAST) as a starting reagent.
  • Ribozymes can also be either electrostatically or covalently attached to polymeric cations for the purpose of reducing charge.
  • the polymer can be attached to the ribozyme by simply converting the 3'-end to a ribonucleoside dialdehyde which is obtained by a periodate cleavage of the terminal 2',3'-cis diol system.
  • other possible modifications may include different linker arms containing carboxyl, amino or thiol functionalities.
  • Yet further examples include use of methylphosphonates and 2'-O-methylribose and 5' or 3' capping or blocking with m 7 GpppG or m 3 2 ' 2,7 GpppG.
  • a kinased ribozyme is contacted with guanosine triphosphate and guanyltransferase to add an m 3 G cap to the ribozyme.
  • the ribozyme can be gel purified using standard procedure. To ensure that the ribozyme has the desired activity, it may be tested with and without the 5' cap using standard procedures to assay both its enzymatic activity and its stability.
  • Synthetic ribozymes including those containing various modifiers, can be purified by high pressure liquid chromatography (HPLC) .
  • HPLC high pressure liquid chromatography
  • Other liquid chromatography techniques, employing reverse phase columns and anion exchangers on silica and polymeric supports may also be used.
  • Synthesis is done on a 1 ⁇ mole scale using a 1 ⁇ ole RNA reaction column (Glen Research) .
  • the average coupling efficiencies are between 97% and 98% for the 394 model, and between 97% and 99% for the 380B model, as deter ⁇ mined by a calorimetric measurement of the released trityl cation.
  • Blocked ribozymes are cleaved from the solid support (e.g. , CPG) , and the bases and diphosphoester moiety deprotected in a sterile vial by dry ethanolic ammonia (2 mL) at 55°C for 16 hours. The reaction mixture is cooled on dry ice. Later, the cold liquid is transferred into a sterile screw cap vial and lyophilized.
  • the solid support e.g. , CPG
  • the residue is suspended in 1 M tetra-n-butylammonium fluoride in dry THF (TBAF) , using a 20 fold excess of the reagent for every silyl group, for 16 hours at ambient temperature (about 15- 25°C) .
  • THF dry THF
  • the reaction is quenched by adding an equal volume of sterile 1 M triethylamine acetate, pH 6.5.
  • the sample is cooled and concentrated on a SpeedVac to half the initial volume.
  • the ribozymes are purified in two steps by HPLC on a C4 300 A 5 mm DeltaPak column in an acetonitrile gradient.
  • the first step is a separation of 5'-DMT-protected ribozyme(s) from failure sequences lacking a 5'-DMT group.
  • Solvents used for this step are: A (0.1 M triethylammonium acetate, pH 6.8) and B (acetonitrile).
  • the elution profile is: 20% B for 10 minutes, followed by a linear gradient of 20% B to 50% B over 50 minutes, 50% B for 10 minutes, a linear gradient of 50% B to 100% B over 10 minutes, and a linear gradient of 100% B to 0% B over 10 minutes.
  • the second step is a purification of a completely deblocked ribozyme by a treatment of 2% trifluoroacetic acid on a C4 300 A 5 mm DeltaPak column in an acetonitrile gradient.
  • Solvents used for this second step are: A (0.1 M triethylammonium acetate, pH 6.8) and B (80% acetonitrile, 0.1 M triethylammonium acetate, pH 6.8).
  • the elution profile is: 5% B for 5 minutes, a linear gradient of 5% B to 15% B over 60 minutes, 15% B for 10 minutes, and a linear gradient of 15% B to 0% B over 10 minutes.
  • the fraction containing ribozyme is cooled and lyophilized on a SpeedVac. Solid residue is dissolved in a minimum amount of ethanol and sodium perchlorate in acetone. The ribozyme is collected by centrifugation, washed three times with acetone, and lyophilized.
  • ribozymes While synthetic ribozymes are preferred in this invention, those produced by expression vectors can also be used. In designing a suitable ribozyme expression vector the following factors are important to consider. The final ribozyme must be kept as small as possible to minimize unwanted secondary structure within the ribozyme.
  • a promoter e.g. , the human cytomegalovirus immediate early promoter or human beta actin promoter
  • Such a promoter should express the ribozyme at a level suitable to effect production of enough ribozyme to destroy a target RNA, but not at too high a level to prevent other cellular activities from occurring (unless cell death itself is desired) .
  • a hairpin at the 5' end of the ribozyme is useful to ensure that the required transcription initiation sequence (GG or GGG or GGGAG) does not bind to some other part of the ribozyme and thus affect regulation of the transcription process.
  • the 5' hairpin is also useful to protect the ribozyme from 5'-3' exonucleases.
  • a selected hairpin at the 3' end of the ribozyme gene is useful since it acts as a transcription termination signal, and protects the ribozyme from 3'-5' exonuclease activity.
  • a known termination signal is that present on the T7 RNA polymerase system. This signal is about 30 nucleotides in length.
  • Other 3' hairpins of shorter length can be used to provide good termination and RNA stability.
  • Such hairpins can be inserted within the vector sequences to allow standard ribozymes to be placed in an appropriate orientation and expressed with such sequences attached.
  • Poly(A) tails are also useful to protect the 3' end of the ribozyme.
  • ribozyme testing Once the desired ribozymes are selected, synthesized and purified, they are tested in kinetic and other experiments to determine their utility.
  • RNA is resuspended in 50-100 ⁇ l TE (TRIS 10 mM, EDTA 1 mM, pH 7.2). An aliquot of this solution is diluted 100-fold into 1 ml TE, half of which is used to spectrophotometrically quantitate the ribozyme solution. The concentration of this dilute stock is typically 150-800 nM. Purity of the ribozyme is confirmed by the presence of a single band on a denaturing polyacrylamide gel.
  • a ribozyme may advantageously be synthesized in two or more portions. Each portion of a ribozyme will generally have only limited or no enzymatic activity, and the activity will increase substantially (by at least 5-10 fold) when all portions are ligated (or otherwise juxtaposed) together.
  • a specific example of hammerhead ribozyme synthesis is provided below.
  • the method involves synthesis of two (or more) shorter "half" ribozymes and ligation of them together using T4 RNA ligase.
  • T4 RNA ligase For example, to make a 34 mer ribozyme, two 17 ers are synthesized, one is phosphorylated, and both are gel purified. These purified 17 mers are then annealed to a DNA splint strand complementary to the two 17 mers. (Such a splint is not always necessary.) This DNA splint has a sequence designed to locate the two 17 mer portions with one end of each adjacent each other. The juxtaposed RNA molecules are then treated with T4 RNA ligase in the presence of ATP. The 34 mer RNA so formed is then HPLC purified.
  • a substrate solution is made IX in assay buffer (75 mM Tris-HCl, pH 7.6; 0.1 mM EDTA, 10 mM MgCl 2 ) such that the concentration of substrate is less than 1 nM.
  • a ribozyme solution (typically 20 nM) is made IX in assay buffer and four dilutions are made using IX assay buffer. Fifteen ⁇ l of each ribozyme dilution
  • the reaction is started by mixing 15 ⁇ l of substrate into each ribozyme tube by rapid pipetting (note that final ribozyme concentrations are 10, 8, 6, 4, 2 nM) .
  • Five ⁇ l aliquots are removed at 15 or 30 second intervals and quenched with 5 ⁇ l stop solution (95% formamide, 20 mM EDTA xylene cyanol, and bromphenol blue dyes) .
  • 5 ⁇ l stop solution 95% formamide, 20 mM EDTA xylene cyanol, and bromphenol blue dyes
  • the samples are separated on either 15% or 20% polyacrylamide gels. Each gel is visualized and quantitated with an Ambis beta scanner (Ambis Systems, San Diego, CA) .
  • the substrates are prepared by transcription using T7 RNA polymerase and defined templates containing a T7 promoter, and DNA encoding appropriate nucleotides of the target RNA.
  • the substrate solution is made IX in assay buffer (75 mM Tris-HCl, pH 7.6; 0.1 mM EDTA; 10 mM MgCl 2 ) and contains 58 nanomolar concentration of the long RNA molecules.
  • the reaction is started by addition of gel purified ribozymes to 1 ⁇ M concentration. Aliquots are removed at 20, 40, 60, 80 and 100 minutes, then quenched by the addition of 5 ⁇ l stop solution. Cleavage products are separated using denaturing PAGE. The bands are visualized and quantitated with an Ambis beta scanner.
  • Kinetic Analysis A simple reaction mechanism for ribozyme- mediated cleavage is: k x k 2
  • the boxed step is important only in substrate excess.
  • the rate of the reaction is the rate of disappearance of substrate with time:
  • Lipid molecules are dissolved in a volatile organic solvent (CHC1 3 , methanol, diethylether, ethanol, etc.). The organic solvent is removed by evaporation.
  • the lipid is hydrated into suspension with O.lx phosphate buffered saline (PBS) , then freeze-thawed 3x using liquid nitrogen and incubation at room temperature.
  • PBS O.lx phosphate buffered saline
  • the suspension is extruded sequentially through a 0.4 ⁇ m, 0.2 ⁇ m and 0.1 ⁇ m polycarbonate filters at maximum pressure of 800 psi.
  • the ribozyme is mixed with the extruded liposome suspension and lyophilized to dryness.
  • the lipid/ribozyme powder is rehydrated with water to one-tenth the original volume.
  • the suspension is diluted to the minimum volume required for extrusion (0.4 ml for 1.5 ml barrel and 1.5 ml for 10 ml barrel) with lxPBS and re-extruded through 0.4 ⁇ m, 0.2 ⁇ m, 0.1 ⁇ m polycarbonate filters.
  • the liposome entrapped ribozyme is separated from untrapped ribozyme by gel filtration chromatography (SEPHAROSE CL-4B, BIOGEL A5M) .
  • the liposome extractions are pooled and sterilized by filtration through a 0.2 ⁇ m filter.
  • the free ribozyme is pooled and recovered by ethanol precipitation.
  • the liposome concentration is determined by incorporation of a radioactive lipid.
  • the ribozyme concentration is determined by labeling with 32 P. Rossi et al., 1992, supra (and references cited therein) describe other methods suitable for preparation of liposomes.
  • liposomes composed of DPPG/DPPC/Cholesterol (in a ratio of: 50/17/33) gave a punctate pattern of fluorescence, while DOPE/Egg PC/Cholesterol (30/37/33) gave a diffuse, homogeneous pattern of fluorescence in the cytoplasm.
  • Cell fractionation showed that 80% of the entrapped contents from the DPPG/DPPC/Cholesterol formulation was localized in the membrane fraction, whereas the DOPE/Egg PC/Cholesterol formulation was localized in the cytoplasm. Further characterization of the latter formulation showed that after 3 hours, 70% of the fluorescence was cytoplasmic and 30% was in the membrane. After 24 hours, uptake had increased 5 fold and the liposome contents were distributed 50/50 between the cytoplasmic and membrane fractions.
  • Liposomes containing 15 ribozymes ( 32 P-labeled) targeted to the HSV ICP4 mRNA were prepared and incubated with the cells. After 24 hours, 25% of the liposome dose was taken up with approximately 60,000 liposomes per cell. Thirty percent of the delivered ribozyme was intact after 24 hours. Cell fractionation studies showed 40% of the intact ribozyme to be in the membrane fraction and 52% of the intact ribozyme to be in the cytoplasmic fraction. In Vivo Assay
  • the efficacy of action of a chosen ribozyme may be tested in vivo using standard procedures in transformed cells or animals which express the target mRNA.
  • the accumulation of target mRNA in cells or the cleavage of the RNA by ribozymes or RNaseH can be quantified using an RNase protection assay.
  • antisense riboprobes are transcribed from template DNA using T7 RNA polymerase (U.S. Biochemical) in 20 ⁇ l reactions containing IX transcription buffer (supplied by the manufacturer), 0.2 mM ATP, GTP and UTP, 1 U/ ⁇ l pancreatic RNase inhibitor (Boehringer Mannheim Biochemicals) and 200 ⁇ Ci 32 P- labeled CTP (800 Ci/mmol, New England Nuclear) for 1 hour at 37°C.
  • Template DNA is digested with 1 U RNase- free DNasel (U.S. Biochemical, Cleveland, OH) at 37°C for 15 minutes and unincorporated nucleotides removed by G-50 SEPHADEX spin chromatography.
  • the target RNA can be transcribed in vi tro using a suitable DNA template.
  • the transcript is purified by standard methods and digested with ribozyme at 37°C according to methods described later.
  • afflicted (mRNA-expressing) cells are harvested into 1 ml of PBS) transferred to a 1.5 ml EPPENDORF tube, pelleted for 30 seconds at low speed in a microcentrifuge, and lysed in 70 ⁇ l of hybridization buffer (4 M guanidine isothiocyanate, 0.1% sarcosyl, 25 mM sodium citrate, pH 7.5).
  • Cell lysate 45 ⁇ l or defined amounts of in vi tro transcript (also in hybridization buffer) is then combined with 5 ⁇ l of hybridization buffer containing 5 x 10 5 cpm of each antisense riboprobe in 0.5 ml Eppendorf tubes, overlaid with 25 ⁇ l mineral oil, and hybridization accomplished by heating overnight at 55°C.
  • hybridization reactions are diluted into 0.5 ml RNase solution (20 U/ml RNaseA, 2 U/ml RNaseTl, 10 U/ml RNase-free DNasel in 0.4 M NaCl), heated for 30 minutes at 37°C, and 10 ⁇ l of 20% SDS and 10 ⁇ l of Proteinase K (10 mg/ml) added, followed by an additional 30 minutes incubation at 37°C.
  • Hybrids are partially purified by extraction with 0.5 ml of a 1:1 mixture of phenol/chloroform; aqueous phases are combined with 0.5 ml isopropanol, and RNase- resistant hybrids pelleted for 10 minutes at room temperature (about 20°C) in a microcentrifuge.
  • Pellets are dissolved in 10 ⁇ l loading buffer (95% formamide, IX TBE, 0.1% bromophenol blue, 0.1% xylene cylanol) , heated to 95°C for five minutes, cooled on ice, and analyzed on 4% polyacrylamide/7 M urea gels under denaturing conditions.
  • the chosen ribozyme can be tested to determine its stability, and thus its potential utility. Such a test can also be used to determine the effect of various chemical modifications (e.g. , addition of a poly(A) tail) on the ribozyme stability and thus aid selection of a more stable ribozyme.
  • a reaction mixture contains 1 to 5 pmoles of 5' (kinased) and/or 3' labeled ribozyme, 15 ⁇ g of cytosolic extract and 2.5 mM MgCl 2 in a total volume of 100 ⁇ l. The reaction is incubated at 37°C.
  • a 3'-labeled ribozyme can be formed by incorporation of the 32 P-labeled cordycepin at the 3' OH using poly(A) polymerase.
  • the poly(A) polymerase reaction contains 40 mM Tris, pH 8, 10 mM MgCl 2 , 250 mM NaCl, 2.5 mM MnCl 2 , ; 3 ⁇ l 32 P cordycepin, 500 Ci/mM; and 6 units poly(A) polymerase in a total volume of 50 ⁇ l. The reaction mixture is incubated for 30 minutes at 37°C. Effect of Base Substitution Upon Ribozyme Activity
  • minor base changes can be made in the substrate cleavage region recognized by a specific ribozyme.
  • the substrate sequences can be changed at the central "C" nucleotide, changing the cleavage site from a GUC to a GUA motif.
  • the K cat / ⁇ ⁇ -_ values for cleavage using each substrate are then analyzed to determine if such a change increases ribozyme cleavage rates.
  • Similar experiments can be performed to address the effects of changing bases complementary to the ribozyme binding arms. Changes predicted to maintain strong binding to the complementary substrate are preferred.
  • cleavage rates of ribozymes containing varied arm lengths, but targeted to the same length of short RNA substrate can be tested. Minimal arm lengths are required and effective cleavage varies with ribozyme/substrate combinations.
  • the cleavage activity of selected ribozymes can be assessed using target mRNA substrates.
  • the assays are performed in ribozyme excess and approximate K cat /K I . in values obtained. Comparison of values obtained with short and long substrates indicates utility in vivo of a ribozyme. Intracellular Stability of Liposome-delivered Ribozymes
  • Ribozymes are 32 P- end labeled, entrapped in liposomes and delivered to target mRNA-containing cells for three hours.
  • the cells are fractionated and ribozyme is purified by phenol/chloroform extraction.
  • cells lxlO 7 , T-175 flask
  • the cells are homogenized by douncing 35 times in 4 ml of TSE (10 mM Tris, pH 7.4, 0.25 M Sucrose, mM EDTA). Nuclei are pelleted at lOOxg for 10 minutes.
  • Subcellular organelles are pelleted at 200,000xg for two hours using an SW60 rotor.
  • the pellet is resuspended in 1 ml of H buffer (0.25 M Sucrose, 50 mM HEPES, pH 7.4).
  • the supernatant contains the cytoplasmic fraction (in approximately 3.7 ml).
  • the nuclear pellet is resuspended in 1 ml of 65% sucrose in TM (50 mM Tris, pH 7.4, 2.5 mM MgCl 2 ) and banded on a sucrose step gradient (1 ml nuclei in 65% sucrose TM, 1 ml 60% sucrose TM, 1 ml 55% sucrose TM, 50% sucrose TM, 300 ⁇ l 25% sucrose TM) for one hour at 37,000xg with an SW60 rotor.
  • the nuclear band is harvested and diluted to 10% sucrose with TM buffer. Nuclei are pelleted at 37,000xg using an SW60 rotor for 15 minutes and the pellet resuspended in 1 ml of TM buffer. Aliquots are size fractionated on denaturing polyacrylamide gels and the intracellular localization determined. By comparison to the migra' ion rate of newly synthesized ribozyme, the various fractions containing intact ribozyme can be determined.
  • the cells may be fractioned as above and the purity of each fraction assessed by assaying enzyme activity known to exist in that fraction.
  • the various cell fractions are frozen at -70°C and used to determine relative nuclease resistances of modified ribozyme molecules.
  • Ribozyme molecules may be synthesized with 5 phosphorothioate (ps) , or 2'-Omethyl (2'-OMe) modifications at each end of the molecule. These molecules and a phosphodiester version of the ribozyme are end-labeled with 32 P and ATP using T4 polynucleotide kinase.
  • Equal concentrations are added to the cell cytoplasmic extracts and aliquots of each taken at 10 minute intervals.
  • the samples are size fractionated by denaturing PAGE and relative rates of nuclease resistance analyzed by scanning the gel with an Ambis ⁇ -scanner. The results show whether the ribozymes are digested by the cytoplasmic extract, and which versions are relatively more nuclease resistant.
  • Modified ribozymes generally maintain 80-90% of the catalytic activity of the native ribozyme when short RNA substrates are employed.
  • Unlabeled, 5' end-labeled or 3' end-labeled ribozymes can be used in the assays. These experiments can also be performed with human cell extracts to verify the observations.
  • Vero or HeLa cells were grown to 90-95% confluency in 175 cm 2 tissue culture flasks, scraped into 10 ml of cold phosphate buffered saline
  • sucrose TM 50 mM Tris, pH 7.4; 2.5 mM MgCl 2
  • sucrose TM solutions were layered on top of the sample: 1 ml 60%, 1 ml 55%, and 25% sucrose to the top of the tube.
  • Subcellular organelles and membrane components in the post nuclear supernatant were separated from the cytoplasmic fraction by centrifugation at 200,000g for 2 hours in an SW60 rotor.
  • the pellet contained the membrane fraction, which was resuspended in 1 ml of H buffer (0.25 M sucrose; 50 mM HEPES, pH 7.4), and the supernatant contained the cytoplasmic fraction.
  • the assays were as follows: For N-acetyl-beta-hexosaminidase, the reaction mixture contained 0.3 mg/ l 4-methylumbelliferyl-N- acetyl-glucosaminide; 20 mM sodium citrate; pH 4.5; 0.01% Triton X-100; and 100 ⁇ l of sample in a final volume of 500 ⁇ l (Harding et al. , 64 Cell 393, 1991). The reactions were incubated at 37°C for 1 hour and stopped by the addition of 1.5 ml of stop buffer (0.13 M glycine, 0.07 M NaCl, 0.08 M sodium carbonate, pH 10.6). The reaction product was quantitated in a Hitachi F-4010 fluorescence spectrophotometer by excitation of the fluorophore at 360 nm and analysis of the emission at 448 nm.
  • the assay medium contained 25 mM CAPS (3-(Cyclohexylamino) - propanesulfonic acid), pH 10.6; 0.05% Triton X-100; 15 mM MgCl 2 ; 1.25 mg/ml Thymidine-5'-monophosphate-p- nitrophenyl ester; and 100 ⁇ l of sample in a total reaction volume of 200 ⁇ l.
  • the reactions were incubated at 37°C for 2 hours, then diluted to 1 ml with H 2 0 and the absorbance was measured at 400 nm (Razell and Khorana, 234 J. Biol. Chem. 739, 1959) .
  • the reaction contained 85 nM sodium citrate, pH 5.9; 0.12% Triton X- 100; 0.1% sodium taurocholate; 5 M 4-methylumbelliferyl ⁇ -D-glucopyranoside; and 125 ⁇ l of sample in a total volume of 250 ⁇ l (Kennedy and Cooper, 252 Biochem. J. 739, 1988) .
  • the reaction was incubated at 37°C for 1 hour and stopped by the addition of 0.75 ml of stop buffer.
  • Product formation was measured in a fluorescence spectrophotometer by using an excitation wavelength of 360 nm and analysis of the emission at 448 nm.
  • the cytoplasmic enzyme marker, lactate dehydrogenase was assayed in an assay mixture containing 0.2 M Tris; pH 7.4; 0.22 mM NADH; 1 mM sodium pyruvate; and 50 ⁇ l of sample in a final volume of 1.05 ml. Enzyme levels were determined by decreased absorbency at 350 nm resulting from the oxidation of
  • Lactate dehydrogenase was found predominantly in the cytoplasmic fractions of both Vero and HeLa cells, while ⁇ -glucocerebrosidase and alkaline phosphodiesterase were found almost exclusively in the membranous fractions.
  • the hexosaminidase activity in Vero cell fractions was concentrated in the membranous fraction (70%) with about 20% in the cytoplasmic fraction.
  • the isolation of enzyme markers with the appropriate cellular compartment demonstrated that cytoplasmic, membranous and nuclear fractions can be isolated with minimal intercompartmental contamination using this fractionation scheme.
  • the basic oligonucleotide digestion reaction contained substrate nucleic acid (an RNA oligonucleotide of 36 nucleotides) and cell fraction extract in a total volume of 100 ⁇ l. Aliquots (7 ⁇ l) were taken after various periods of incubation at 37°C and added to 7 ⁇ l of gel loading buffer (95% formamide, 0.1% bromophenol blue, 0.1% xylene cyanol, and 20 mM EDTA) . The samples were separated by electrophoresis on a 7 M urea, 20% polyacrylamide gel.
  • substrate nucleic acid an RNA oligonucleotide of 36 nucleotides
  • cell fraction extract in a total volume of 100 ⁇ l. Aliquots (7 ⁇ l) were taken after various periods of incubation at 37°C and added to 7 ⁇ l of gel loading buffer (95% formamide, 0.1% bromophenol blue, 0.1% xylene cyanol, and 20 mM EDTA) . The
  • Intact ribozymes were visualized either by staining with Stains-all (United States Biochemical, Cleveland, OH) , or autoradiography of 32 P-labeled ribozyme. The stained gels and X-ray films were scanned on a Bio 5000 density scanner (U.S. Biochemical). Ribozymes were 5' end- labeled with T4 polynucleotide kinase (U.S. Biochemical) using 10 ⁇ Ci of 32 P ⁇ -ATP (3,000 Ci/mmole, New England Nuclear, Boston, MA), and 20-25 pmoles of ribozymes. The unincorporated nucleotides were separated from the product by G-50 spin chromatography.
  • Nuclease assays contained 1-2 pmoles of 32 P-labeled ribozyme. All oligonucleotides were synthesized on an Applied Biosystems 394 DNA/RNA synthesizer (Applied Biosysterns Inc., Foster City, CA) according to manufacturer's protocols. The nuclear fractions were resuspended in a buffer containing 2.5 mM MgCl 2 . Experiments involving the nuclear fractions were performed in the presence of 1 mM Mg +2 , or in combination with 1 mM Mn +2 , Ca +2 , or Zn +2 .
  • RNA probes were produced from PCR-amplified template DNA using T7 RNA polymerase (U.S. Biochemical) in the presence of 32 P ⁇ -CTP (3,000 Ci/mmole, New England Nuclear, Boston, MA) . Template DNA was inactivated with 1 unit of RNase-free DNasel for 15 minutes at 37°C.
  • Hybridization reactions were covered with mineral oil and incubated at 55°C for 12-16 hours, after which the hybridization reaction was mixed with 500 ⁇ l of RNase buffer (0.4 M NaCl, 20 ⁇ g/ml RNaseA, 2 units/ml Tl RNase) and incubated for 30 minutes at 37°C. RNase activity was quenched by incubation with 10 ⁇ l of 20% SDS and 10 ⁇ l of proteinase K (20 mg/ml) , and the RNA was extracted using a phenol/chloroform mixture. The protected RNA fragment was purified by precipitation with an equal volume of isopropanol in the presence of 20 ⁇ g of carrier yeast tRNA.
  • RNase buffer 0.4 M NaCl, 20 ⁇ g/ml RNaseA, 2 units/ml Tl RNase
  • RNA pellets were resuspended in gel loading buffer, heated to 95°C for 5 minutes and separated by electrophoresis on a 5% polyacrylamide, 7 M urea gel. Protected fragments were visualized by autoradiography, and the films were scanned with a Bio 5000 density scanner.
  • the rate of digestion of ribozymes in Vero cell extracts was similar, demonstrating the lack of significant phosphatase activity in Vero cellular fractions. Similar results were observed with HeLa cellular fractions. In most extracts, ladders of digested fragments were observed; such ladders would not be expected if digestion was an artifact of phosphatase action.
  • digestion using 5' end-labeled ribozymes is an accurate assessment of nuclease action in cellular extracts.
  • ribozymes were incubated in various Vero and HeLa cellular fractions. Incubation of ribozymes in either membranous or nuclear fractions resulted in a linear decrease of intact molecules over time. In contrast, no digestion of ribozymes occurred during a 24 hour incubation in Vero cytoplasmic extracts, and HeLa cytoplasmic extracts exhibited a 20-30 minute delay in the onset of RNA digestion. After this refractory period, the rate of digestion was linear but not as rapid as the rates observed in any of the nuclear or membranous fractions.
  • the effect of four divalent cations (Mg +2 , Mn +2 , Ca +2 , and Zn +2 ) on the nuclease activity of the cellular fractions was assessed. Vero cytoplasmic extracts were stimulated by the addition of 1 mM Mg +2 or Mn +2 , while Ca +2 or Zn +2 had no effect. Nuclease activity in HeLa cytoplasmic extracts was enhanced only by the addition of 1 mM Zn +2 .
  • Vero and HeLa membranous fractions exhibited maximum nuclease activity with the addition of Mg +2 or Mn +2 ions, while the addition of Ca +2 significantly reduced activity of the HeLa membranous fraction and abolished nuclease activity in the Vero membranous fraction.
  • Addition of Zn +2 to both membranous fractions resulted in a loss of all RNase activity.
  • the Vero nuclear extract demonstrated roughly equivalent nuclease activity in the presence of either Mg +2 alone or a Mg +2 and Mn +2 ion combination, less in the presence of Mg +2 and Ca +2 , and no activity in the presence of Mg +2 and Zn +2 .
  • nuclease assays were performed using 1 mM Mg +2 in the presence and absence of 20 mM EDTA.
  • the Mg +2 was replaced with 1 mM Zn +2 .
  • the presence of 20 mM EDTA completely abolished nuclease activity in the Vero and HeLa cytoplasmic fractions and Vero nuclear fractions.
  • Nuclease activity in the HeLa membranous and nuclear fractions was partially inhibited by the addition of EDTA, while EDTA had no effect on the nuclease activity in the Vero membranous fraction.
  • reactions using DNA oligonucleotides were performed using different Vero fractions. All DNase activity in Vero cytoplasmic, membranous, and nuclear fractions was inhibited by 20 mM EDTA.
  • RNA oligonucleotides and HSV-1 mRNA were compared in the presence and absence of activity-enhancing divalent cations (1 mM Mg +2 , Vero cells; 1 mM Zn +2 , HeLa cells) .
  • Total cellular RNA from HSV-1 infected Vero cells (8 mg) and tracer amounts of 32 P-5'-end-labeled RNA oligonucleotides (1 pmole) were incubated with Vero or HeLa cytoplasmic extracts. In the absence of divalent cations, no substantial decrease of intact ribozymes was detected in assays, although mRNA was digested in both Vero and HeLa cytoplasmic extracts.
  • hammerhead ribozymes were compared in both Vero and HeLa cell cytoplasmic, membranous and nuclear fractions.
  • Vero cytoplasmic and nuclear fractions were found to require Mg +2 for optimal nuclease activity, while the membranous fraction was not altered by the addition of divalent cations.
  • HeLa membranous and nuclear fractions were also activated by Mg +2 , while the cytoplasmic fractions required Zn" 2 for nuclease activation.
  • Relative stabilities of ribozymes and mRNAs were compared in Vero and HeLa cytoplasmic fractions.
  • Selected ribozymes can be administered prophylactically, or to patients having disease conditions, e.g. , by exogenous delivery of the ribozyme to a desired tissue by means of an appropriate delivery vehicle, e.g. , a liposome, a controlled release vehicle, by use of iontophoresis, electroporation or ion paired molecules, or covalently attached adducts, and other pharmacologically approved methods of delivery.
  • routes of administration include intramuscular, aerosol, oral (tablet or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal.
  • Expression vectors for immunization with ribozymes and/or delivery of ribozymes are also suitable.
  • any selected ribozyme will depend on the use of the ribozyme. Generally, a specific delivery program for each ribozyme will focus on unmodified ribozyme uptake with regard to intracellular localization, followed by demonstration of efficacy. Alternatively, delivery to these same cells in an organ or tissue of an animal can be pursued. Uptake studies will include uptake assays to evaluate cellular ribozyme uptake, regardless of the delivery vehicle or strategy. Such assays will also determine the intracellular localization of the ribozyme following uptake, ultimately establishing the requirements for maintenance of steady-state concentrations within the cellular compartment containing the target sequence (nucleus and/or cytoplasm) .
  • Toxicity will not only include cell viability but also cell function.
  • Some methods of delivery that may be used include: a. encapsulation in liposomes, b. transduction by retroviral vectors, c. conjugation with cholesterol, d. localization to nuclear compartment utilizing nuclear targeting site found on most nuclear proteins, e. neutralization of charge of ribozyme by using nucleotide derivatives, and f. use of blood stem cells to distribute ribozymes throughout the body.
  • At least three types of delivery strategies are useful in the present invention, including: ribozyme modifications, particle carrier drug delivery vehicles, and retroviral expression vectors.
  • Unmodified ribozymes like most small molecules, are taken up by cells, albeit slowly. To enhance cellular uptake, the ribozyme may be modified essentially at random, in ways which reduce its charge but maintains specific functional groups. This results in a molecule which is able to diffuse across the cell membrane, thus removing the permeability barrier.
  • ribozymes Modification of ribozymes to reduce charge is just one approach to enhance the cellular uptake of these larger molecules.
  • the random approach is not advisable since ribozymes are structurally and functionally more complex than small drug molecules.
  • the structural requirements necessary to maintain ribozyme catalytic activity are well understood by those in the art. These requirements are taken into consideration when designing modifications to enhance cellular delivery. The modifications are also designed to reduce susceptibility to nuclease degradation. Both of these characteristics should greatly improve the efficacy of the ribozyme.
  • Cellular uptake can be increased by several orders of magnitude without having to alter the phosphodiester linkages necessary for ribozyme cleavage activity.
  • Drug delivery vehicles are effective for both systemic and topical administration. They can be designed to serve as a slow release reservoir, or to deliver their contents directly to the target cell.
  • An advantage of using direct delivery drug vehicles is that multiple molecules are delivered per uptake. Such vehicles have been shown to increase the circulation half-life of drugs which would otherwise be rapidly cleared from the blood stream.
  • Some examples of such specialized drug delivery vehicles which fall into this category are liposomes, hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres.
  • liposomes are preferred. Liposomes increase intracellular stability, increase uptake efficiency and improve biological activity.
  • Liposomes are hollow spherical vesicles composed of lipids arranged in a similar fashion as those lipids which make up the cell membrane. They have an internal aqueous space for entrapping water soluble compounds and range in size from 0.05 to several microns in diameter. Several studies have shown that liposomes can deliver RNA to cells and that the RNA remains biologically active.
  • Lipofectin a liposome delivery vehicle originally designed as a research tool, Lipofectin, has been shown to deliver intact mRNA molecules to cells yielding production of the corresponding protein.
  • Liposomes offer several advantages: They are non-toxic and biodegradable in composition; they display long circulation half-lives; and recognition molecules can be readily attached to their surface for targeting to tissues. Finally, cost effective manufacture of liposome-based pharmaceuticals, either in a liquid suspension or lyophilized product, has demonstrated the viability of this technology as an acceptable drug delivery system.
  • Other controlled release drug delivery systems such as nonoparticles and hydrogels may be potential delivery vehicles for a ribozyme. These carriers have been developed for chemotherapeutic agents and protein- based pharmaceuticals, and consequently, can be adapted for ribozyme delivery.
  • Topical administration of ribozymes is advantageous since it allows localized concentration at the site of administration with minimal systemic adsorption. This simplifies the delivery strategy of the ribozyme to the disease site and reduces the extent of toxicological characterization. Furthermore, the amount of material to be applied is far less than that required for other administration routes. Effective delivery requires the ribozyme to diffuse into the infected cells. Chemical modification of the ribozyme to neutralize negative charge may be all that is required for penetration. However, in the event that charge neutralization is insufficient, the modified ribozyme can be co-formulated with permeability enhancers, such as Azone or oleic acid, in a liposome.
  • permeability enhancers such as Azone or oleic acid
  • the liposomes can either represent a slow release presentation vehicle in which the modified ribozyme and permeability enhancer transfer from the liposome into the infected cell, or the liposome phospholipids can participate directly with the modified ribozyme and permeability enhancer in facilitating cellular delivery.
  • both the ribozyme and permeability enhancer can be formulated into a suppository formulation for slow release.
  • Ribozymes may also be systemically administered.
  • Systemic absorption refers to the accumulation of drugs in the blood stream followed by distribution throughout the entire body. Administration routes which lead to systemic absorption include: intravenous, subcutaneous, intraperitoneal, intranasal, intrathecal and ophthalmic.
  • Each of these administration routes expose the ribozyme to an accessible diseased tissue.
  • Subcutaneous administration drains into a localized lymph node which proceeds through the lymphatic network into the circulation.
  • the rate of entry into the circulation has been shown to be a function of molecular weight or size.
  • the use of a liposome or other drug carrier localizes the ribozyme at the lymph node.
  • the ribozyme can be modified to diffuse into the cell, or the liposome can directly participate in the delivery of either the unmodified or modified ribozyme to the cell.
  • a liposome formulation containing phosphatidyl- ethanolomidomethylthiosuccinimide which can deliver oligonucleotides to lymphocytes and macrophages is useful for certain conditions. Furthermore, a 200 nm diameter liposome of this composition was internalized as well as 100 nm diameter liposomes. The 200 nm liposomes exhibit a ten-fold greater packaging capacity than the 100 nm liposomes and can accommodate larger molecules such as a ribozyme expression vector. This ribozyme delivery system prevents mRNA expression in afflicted primary immune cells. Whole blood studies show that the formulation is taken up by 90% of the lymphocytes after 8 hours at 37°C.
  • Most preferred delivery methods include liposomes (10-400 nm) , hydrogels, controlled-release polymers, microinjection or electroporation (for ex vivo treatments) and other pharmaceutically applicable vehicles.
  • the dosage will depend upon the disease indication and the route of administration but should be between 100-200 mg/kg of body weight/day.
  • the duration of treatment will extend through the course of the disease symptoms, possibly continuously.
  • the number of doses will depend upon disease delivery vehicle and efficacy data from clinical trials.

Abstract

An enzymatic RNA molecule which cleaves mRNA associated with development of maintenance of an inflammatory disease, an arthritic condition, a stenotic condition, or a cardiovascular condition.

Description

DESCRIPTION
METHOD AND REAGENT FOR TREATMENT OF ANIMAL DISEASES
Background of the Invention This invention relates to methods for inhibition of various animal diseases, including: a) inflammatory disease, in particular the inhibition of genetic expression which leads to the reduction or elimination of immune cell proliferation in inflammation or pre- inflammatory conditions; b) osteoarthritis, in particular, inhibition of genetic expression which leads to a reduction or elimination of extracellular matrix digestion by matrix etalloproteinases; c) a stenotic condition, in particular the inhibition of genetic expression which leads to the reduction or elimination of cellular proliferation in the areas of restenotic plaques; and d) cardiovascular disease such as hypertension, and in particular, those mediated by angiotensin converting enzyme activity. Inflammatory disease
Inflammation is a tissue reaction to irritation, infection or injury marked by localized heat, swelling, redness, pain and sometimes loss of function. When tissues sustain physiological injury, the afflicted cells synthesize and/or release chemicals (cytokines and cellular adhesion molecules) which attract lymphoid cells and accelerate cell growth which is required for healing. The infiltration of lymphoid cells to the wound leads to the release of more biological response modifying molecules, which include vasoregulatory substances such as bradykinin and immunoregulatory molecules exemplified by the interleukin family of proteins. As healing occurs, the immune cell infiltration wanes and the inflammatory process ceases. Under certain physiological conditions, such as those observed in psoriasis, asthma, head injuries and systemic inflammatory response syndrome (SIRS) , the condition persists because of inappropriate cellular controls. A persistent inflammatory condition may be acute or chronic and is generically referred to as inflammatory disease.
Inflammatory disease is exemplified by psoriasis, which is a skin disease caused by excessive growth of basal keratinocytes in the epidermis. It is believed that this uncontrolled growth of the keratinocytes results from stimulation by infiltrating T-cells. The conversion of normal skin to a psoriatic lesion is initiated by injury to the tissue and subsequent recognition of the injury by circulating immune cells. An acute inflammatory reaction ensues and results in the release of various cytokines and growth factors, including tumor necrosis factor alpha
(TNF-α) and interleukin-1 (IL-1) . A number of the cytokines released by infiltrating leukocytes induce production of cell adhesion molecules on the surface of the keratinocytes, while others induce the proliferation of basal keratinocytes and result in the release of keratinocyte factors which either auto-stimulate further growth or inhibit normal controls which limit keratinocyte proliferation. The injury further results in the local activation of T-cells which return to the circulation and may cause subsequent psoriatic conditions which are quite distant from the original site of injury.
Induction of the acute phase inflammatory reactants results in the production of IL-1 and TNF-alpha; both of these molecules induce the appearance of the T-cell homing proteins, I-CAM, ELAM-1 and VCAM-1 on the cell surface of keratinocytes. TNF-α also induces the production of TGF-α, IL-6 and IL-8 by the keratinocytes. The cytokines and growth factors produced by the keratinocytes and infiltrating leukocytes cause uncontrolled keratinocyte proliferation. As the basal layer keratinocytes proliferate, the suprabasal keratinocytes are pushed closer to the surface of the skin where they normally form the cornified envelope layer and the stratum corneum. The abnormal rate of proliferation by the basal keratinocytes results in improper formation of the epidermis and stratum corneum and the ultimate development of the red, scaly appearance which is characteristic of psoriatic skin. The lack of keratinocyte growth regulation has been attributed to an unidentified mutation in a single gene or group of genes in which the keratinocytes no longer respond to the termination signal for wound healing.
There are 3 million patients in the United States afflicted with psoriasis. The available treatments for psoriasis are corticosteroids. The most widely prescribed are TEMOVATE (clobetasol propionate) , LIDEX (fluocinonide) , DIPROLENE (betamethasone propionate) , PSORCON (diflorasone diacetate) and TRIAMCINOLONE formulated for topical application. The mechanism of action of corticosteroids is multifactorial and probably not due to simple inhibition of cell replication. This is a palliative therapy because the underlying cause of the disease remains, and upon discontinuation of the treatment the disease returns. Discontinuation of treatment is often prompted by the appearance of adverse effects such as atrophy, telangiectasias and purpura. Corticosteroids are not recommended for prolonged treatments or when treatment of large and/or inflamed areas is required. Alternative treatments include retinoids, such as etretinate, which has been approved for treatment of severe, refractory psoriasis. Alternative retinoid-based treatments are in advanced clinical trials. Retinoids act by converting keratinocytes to a differentiated state and restoration of normal skin development. Immunosuppressive drugs such as cyclosporine are also in the advanced stages of clinical trials. Due to the nonspecific mechanism of action of corticosteroids, retinoids and immunosuppressives, all current treatments of psoriasis exhibit severe side effects and should not be used for extended periods of time unless the condition is life- threatening or disabling. There is a need for a less toxic, effective therapeutic agent in psoriatic patients. Asthma affects nearly 5% of the population in industrialized nations, yet it is underdiagnosed and undertreated. There is evidence that the incidence and prevalence of asthma are rising. These trends are occurring despite increases in the available therapies for asthma, which suggests that current methods of treating asthma are inadequate or not being utilized appropriately. Recently, it has been recognized that chronic asthma involves a characteristic inflammatory response in the airways.
Although it has long been acknowledged that fatal asthma is associated with inflammatory changes in the submucosal surfaces of the airways, it is now apparent that inflammation is present in patients with very mild asthma. Biopsies of patients have shown that infiltration of immune cells, especially eosinophils and lymphocytes, and epithelial shedding are prominent features. Further, there is a strong correlation between the degree of eosinophilia and the degree of bronchial hyperresponsiveness. Eosinophils are localized to areas of epithelial damage in the airways of patients. The basic proteins released by the eosinophils may be responsible for the damage observed in these patients. The role of mast cells and neutrophils in the disease is uncertain. Lymphocytes are present at the sites of tissue damage, but their role may be as mediators to amplify the eosinophilic response. In fact, interleukin-5, which is released by T-lymphocytes, is important in retaining and priming eosinophil action in the airway. Arthritis
There are several types of arthritis, with osteoarthritis and rheumatoid arthritis being predominant. Osteoarthritis is a slowly progressive disease characterized by degeneration of articular cartilage with proliferation and remodeling of subchondral bone. It presents with a clinical picture of pain, deformity, and loss of joint motion. Rheumatoid arthritis is a chronic systemic inflammatory disease. Rheumatoid arthritis may be mild and relapsing or severe and progressive, leading to joint deformity and incapacitation.
Arthritis is the major contributor to functional impairment among the older population. It is the major cause of disability and accounts for a large proportion of the hospitalizations and health care expenditures of the elderly. Arthritis is estimated to be the principal cause of total incapacitation for about one million persons aged 55 and older and is thought to be an important contributing cause for about one million more. Estimating the incidence of osteoarthritis is difficult for several reasons. First, osteoarthritis is diagnosed objectively on the basis of reading radiographs, but many people with radiologic evidence of disease have no obvious symptoms. Second, the estimates of prevalence are based upon clinical evaluations because radiographic data is not available for all afflicted joints. In the NHANESI survey of 1989, data were based upon a thorough musculoskeletal evaluation during which any abnormalities of the spine, knee, hips, and peripheral joints were noted as well as other specific diagnoses. Based on these observations, 12% of the US population between 25 and 74 years of age have osteoarthritis.
It is generally agreed that rheumatoid arthritis has a world-wide distribution and affects all racial and ethnic groups. The exact prevalence in the US is unknown but has been estimated to range between 0.5% and 1.5%. Rheumatoid arthritis occurs at all age levels and generally increases in prevalence with advancing age. It is 2-3 times more prevalent in women than in men and peak incidence occurs between 40-60 years of age. In addition to i munological factors, environmental, occupational and psychosocial factors have been studied for potential etiologic roles in the disease.
The extracellular matrix of multicellular organisms plays an important role in the formation and maintenance of tissues. The meshwork of the extracellular matrix is deposited by resident cells and provides a framework for cell adhesion and migration, as well as a permeability barrier in cell-cell communication. Connective tissue turnover during normal growth and development or under pathological conditions is thought to be mediated by a family of neutral metalloproteinases, which are zinc-containing enzymes that require calcium for full activity. The regulation of metalloproteinase expression is cell-type specific and may vary among species.
The best characterized of the matrix metalloproteinases, interstitial collagenase (MMP-1) , is specific for collagen types I, II, and III. MMP-1 cleaves all three α chains of the triple helix at a single point initiating sequential breakdown of the interstitial collagens. Interstitial collagenase activity has been observed in rheumatoid synovial cells as well as in the synovial fluid of patients with inflammatory arthritis. Gelatinases (MMP-2) represent a subgroup of the metalloproteinases consisting of two distinct gene products; a 70 kDa gelatinase expressed by most connective tissue cells, and a 92 kDa gelatinase expressed by inflammatory phagocytes and tumor cells. The larger enzyme is expressed by macrophages, SV-40 transformed fibroblasts, and neutrophils. The smaller enzyme is secreted by H-ras transformed bronchial epithelial cells and tumor cells, as well as normal human skin fibroblasts. These enzymes degrade gelatin (denatured collagen) as well as native collagen type XI. Stromelysin (MMP-3) has a wide spectrum of action on molecules composing the extracellular matrix. It digests proteoglycans, fibronectin, laminin, type IV and IX collagens and gelatin, and can remove the N-terminal propeptide region from procollagen, thus activating the collagenase. It has been found in human cartilage extracts, rheumatoid synovial cells, and in the synovium and chondrocytes of joints in rats with collagen-induced arthritis.
Both osteoarthritis and rheumatoid arthritis are treated mainly with compounds that inhibit cytokine or growth-factor induced synthesis of the matrix metalloproteinases which are involved in the extracellular matrix destruction observed in these diseases. Current clinical treatments rely upon dexamethasone and retinoid compounds, which are potent suppressors of a variety of metalloproteinases. The global effects of dexamethasone and retinoid treatment upon gene expression in treated cells make the development of alternative therapies desirable, especially for long term treatments. Recently, it was shown that ga ma-interferon suppressed lipopolysaccharide induced collagenase and stromelysin production in cultured macrophages. Also, tissue growth factor-β (TGF-β) has been shown to block epidermal growth factor (EGF) induction of stromelysin synthesis in vitro. Experimental protocols involving gene therapy approaches include the controlled expression of the metalloproteinase inhibitors TIMP-1 and TIMP-2. Of the latter three approaches, only gamma-interferon treatment is currently feasible in a clinical application. Stenosis
Stenosis is the occurrence of a blockage in a blood vessel. Such blockages may lead to impairment of functions or even death, dependent upon which vessel it occurs in and its size. These may be prevented as described below. One example of this condition is restenosis. Restenosis is a disease state which occurs as a sequelae to percutaneous transluminal angioplasty (PCTA) or coronary artery bypass grafting (CABG) treatments of cardiovascular disease. The condition is caused, primarily, by the proliferation of smooth muscle cells (atherophils) in the lamina propria layer of the intima in the vessel wall and secondarily, by the proliferation of other cell types present in the lamina propria. A secondary effect of the cellular proliferation is the increase in collagen and matrix protein synthesis. The cellular proliferation of the intimacytes and surrounding connective tissues results in intimal thickening, loss of vessel elasticity and reduction in blood flow through the afflicted region of the vessel. While inflammation may play a role in the pathology of restenosis, other pathologic mechanisms are involved and possibly represent the underlying cause of the disease. Pathologic mechanisms associated with restenosis can be divided into three categories based upon when the restenosis occurs. The three categories and their mechanisms are 1) pre- and post-PCTA: thrombosis, platelet activation and thrombin generation; 2) immediate, after PCTA: elastic recoil; and 3) delayed, after PCTA: fibrocellular proliferation. Acute or immediate restenosis occurs in approximately 10% of the patients undergoing PCTA and slower-developing restenosis (six month onset) occurs in approximately 30% of those patients. The estimated number of PCTA patients in 1990 was 300,000 - 400,000. Thus, there are about 150,000 cases of restenosis per year in the United States.
The presently preferred chemotherapeutic treatment of patients is the use of streptokinase, urokinase or other thrombolytic compounds, such as fish oil, anticoagulants, ACE (angiotensin converting enzyme) inhibitors, aspirin and cholesterol lowering compounds; alternative treatment includes the surgical incorporation of endoluminal stents. It is reported that none of the current therapies have significantly impacted the rates of restenosis occurrence. A number of compounds are currently in preclinical evaluations. Platelet inhibitors include GR32191, Sultroban, Ketanserin, and fish oil. Angiopeptin is being tested as a growth factor inhibitor and Lovostatin, Enoxaparin, RD Heparin, Cilazapril and Fosinopril are being investigated as smooth muscle cell proliferation inhibitors. While platelet inhibitors are being tested for prevention of restenosis, it appears that these compounds will not be efficacious as short-term treatments. One of the biggest problems with current therapies is the occurrence of pharmacologic side-effects. These effects not only create other physiological problems, but also decrease the levels of patient compliance, thereby reducing the therapeutic efficacy of the treatments.
The proliferation of antherophils may be induced through a host of genetic activations, but the best candidate for targeting smooth muscle proliferation is the c-myb gene. The c-myb protein binds DNA and activates DNA replication and cellular growth. The role of c-myb in smooth muscle cell replication has been documented in bovine cells, and the expression of c-myb has been shown to activate cellular replication in chicken embryo fibroblasts and human T-lymphocytes.
Cellular growth factors may also play a role in local proliferation of intimacytes. Trauma to the area may induce the release of many factors such as TGF-β, PDGF, bFGF, endothelium-derived relaxing factor, CGRP and angiotensin II. Each of these factors could play a role in the induction of cellular proliferation in restenotic plaques, but these factors are soluble proteins which exert their effects through secondary messenger systems in the target cells. One such messenger system is the NF-KB cascade. NF-KB protein activates cellular transcription and induces increases in cellular synthetic pathways. In a resting cell, this protein is found in the cytoplasm, complexed with its inhibitor, 1KB. Upon phosphorylation of the 1KB molecule, the complex dissociates and NFKB is released for transport to the nucleus, where it binds DNA and induces transcriptional activity in (NF-KB) -responsive genes. One of the (NF-KB) -responsive genes is the NF-KB gene. Thus, release of the NF-KB protein from the complex results in a cascade of gene expression which is auto- induced.
Summary of the Invention The invention features use of ribozy es to treat or prevent various animal diseases, in particular, those human diseases noted above.
One such disease is psoriasis, which can be treated, e.g. , by inhibiting the synthesis of tumor necrosis factor in activated lymphocytes, and basal keratinocytes. The invention also features use of ribozymes to treat chronic asthma, e.g. , by inhibiting the synthesis of IL-5 in lymphocytes and preventing the recruitment and activation of eosinophils. Cleavage of targeted mRNAs (tumor necrosis factor and IL-5 mRNAs) expressed in keratinocytes, T-lymphocytes, monocytes or macrophages inhibits the synthesis of tumor necrosis factor and IL-5, respectively.
A number of other cytokines may also be involved in the activation of inflammation in asthmatic patients, including platelet activating factor, IL-1, IL-3, IL-4, GM-CSF, TNFα, gamma interferon, ILAM-1, ELAM-1 and EoCSF. In addition to these cytokines, it is appreciated that any cellular receptors which mediate the activities of the cytokines are also good targets for intervention in inflammatory diseases. These targets include, but are not limited to, the IL-1R and TNFαR on keratinocytes, epithelial and endothelial cells in airways. Recent data suggest that certain neuropeptides may play a role in asthmatic symptoms. These peptides include substance P, neurokinin A and calcitonin-gene-related peptides. These target genes may have more general roles in inflammatory diseases, but are currently assumed to have a role only in asthma. Other genes which are considered to play a role in asthma are the c-myb and c-myc genes, which may be triggered to induce endothelial cell proliferation and contribute to blockage of the airways. Those skilled in the art will recognize the other potential targets noted above are also suitable for treatment with ribozymes, which will reduce the risk or occurrence of inflammatory disease, such as the interleukins (1, 3, 4, 6, and 8), glycerol transferase, selectins (E-selectin, MEL-14) , cell adhesion molecules (ICAM-1, ELAM-1, VCAM-1, GMP-140, MAM), TGF-α, IL-1R, TGFCR, EoCSF, α-, β- or γ-interferon, EoCSF, GM-CSF and protein kinase C (PKC) .
In particularly preferred embodiments, a ribozyme to TNFα nucleotides 374 to 393 can be use to inhibit TNFα protein production. This region may not a very good region to target because of secondary RNA structure in this region. An adjoining region which begins at nucleotide 380 in the previous reference and extends to nucleotide 412 (408-440, our sequence numbers see below) appears to contain a relatively more accessible RNA structure, and therefore represents an improved target over the region between nucleotides 374-393 (402-421, our sequence numbers) . Another disease is arthritis, which can be treated by inhibition of collagenase and stromelysin production in the synovial membrane of joints. Ribozyme treatment can be a partner to current treatments which primarily target immune cells reacting to pre-existing tissue damage. Early ribozyme treatment which reduces the collagenase or stromelysin-induced damage can be followed by treatment with the anti-inflammatories or retinoids, if necessary. In this manner, expression of the proteinases can be controlled at both transcriptional and translational levels. Ribozyme treatment can be given to patients expressing radiological signs of osteoarthritis prior to the expression of clinical symptoms. Ribozyme treatment can impact the expression of stromelysin without introducing the non-specific effects upon gene expression which accompany treatment with the retinoids and dexamethasone. The ability of stromelysin to activate procollagenase indicates that a ribozyme which reduces stromelysin expression can also be used in the treatment of both osteoarthritis (which is primarily a stromelysin- associated pathology) and rheumatoid arthritis (which is primarily related to enhanced collagenase activity) . While a number of cytokines and growth factors induce metalloproteinase activities during wound healing and tissue injury of a pre-osteoarthritic condition, these molecules are not preferred targets for therapeutic intervention. Primary emphasis is placed upon inhibiting the molecules which are responsible for the disruption of the extracellular matrix, because most people will be presenting radiologic or clinical symptoms prior to treatment. The most versatile of the metalloproteinases (the molecule which can do the most structural damage to the extracellular matrix, if not regulated) is stromelysin. Additionally, this molecule can activate procollagenase, which in turn causes further damage to the collagen backbone of the extracellular matrix. Under normal conditions, the conversion of prostromelysin to active stromelysin is regulated by the presence of inhibitors called TIMPs (tissue inhibitors of MMP) . Because the level of TIMP in synovial cells exceeds the level of prostromelysin and stromelysin activity is generally absent from the synovial fluid associated with non-arthritic tissues, the toxic effects of inhibiting stromelysin activity in non-target cells should be negligible.
Thus, the invention features use of ribozymes to treat or prevent arthritis, particularly osteoarthritis, e.g. , by inhibiting the synthesis of the prostromelysin molecule in synovial cells, or by inhibition of other matrix metalloproteinases discussed above. Cleavage of targeted mRNAs (stromelysin mRNAs) expressed in macrophages, neutrophils and synovial cells represses the synthesis of the zymogen form of stromelysin, prostromelysin. Those in the art will recognize the other potential targets discussed above are also suitable for treatment with ribozymes, which will reduce the risk or occurrence of pathologic degradation of the extracellular matrix such as the collagenase and gelatinase metalloproteinases, other proteinases which can activate the proenzyme forms of the metalloproteinases in synovial fluid or cartilaginous cells, cytokines or growth factors which activate the expression of the metalloproteinases and adhesion molecules which attract macrophage and neutrophils to the areas of tissue injury. Another disease is stenosis, which is treated, e.g. , by inhibiting the activation of smooth muscle proliferation by inhibiting the expression of the cellular c-myb gene. Cleavage of targeted mRNAs (c-myb mRNAs) expressed in endothelial cells and smooth muscle cells represses activation of cellular replication and abnormal proliferation of the smooth muscle or endothelial cells. Other potential targets suitable for treatment with ribozymes, which will reduce the risk or occurrence of cellular proliferation in the areas of restenotic risk include mRNAs encoding TGF-β, NF-KB, PDGF, bFGF, endothelium-derived relaxing factor, CGRP, and angiotensin II.
Yet another disease is cardiovascular disease, which can be treated, e.g. , by inhibiting the activation of angiotensin by angiotensin converting enzyme (ACE) , or by inhibition of the activity of endothelin converting enzyme (ECE) . Cleavage of targeted mRNAs (ACE mRNAs) expressed in endothelial cells elicits decreased levels of the vasoactive form of angiotensin. Those in the art will recognize many other potential targets suitable for treatment with ribozymes, which will reduce the risk or occurrence of cardiovascular disease, such as mRNAs encoding HMG CoA reductase, renin, bradykinin, plasminogen, factors IX, X and II, 2-5A synthetase, ADH and fibrinogen.
Ribozymes are RNA molecules having an enzymatic activity which is able to repeatedly cleave other separate RNA molecules in a nucleotide base sequence specific manner. It is alleged that such enzymatic RΝA molecules can be targeted to virtually any RΝA transcript and efficient cleavage has been achieved in vitro . Kim et al., 84 Proc. Νatl. Acad. Sci. USA 8788, 1987; Haseloff and Gerlach, 334 Nature 585, 1988; Cech, 260 JAMA 3030, 1988; and Jefferies et al., 17 Nucleic Acids Research 1371, 1989.
Ribozymes act by first binding to a target RNA. Such binding occurs through the target RNA binding portion of a ribozyme which is held in close proximity to an enzymatic portion of the RNA which acts to cleave the target RNA. Thus, the ribozyme 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 a ribozyme 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.
The enzymatic nature of a ribozyme is advantageous over other technologies, such as antisense technology (where a nucleic acid molecule simply binds to a nucleic acid target to block its translation) since the effective concentration of ribozyme necessary to effect a therapeutic treatment is lower than that of an antisense oligonucleotide. This advantage reflects the ability of the ribozyme to act enzymatically. Thus, a single ribozyme molecule is able to cleave many molecules of target RNA. In addition, the ribozyme is a highly specific inhibitor, with the specificity of inhibition depending not only on the base pairing mechanism of binding, but also on the mechanism by which the molecule inhibits the expression of the RNA to which it binds. That is, the inhibition is caused by cleavage of the RNA target and so specificity is defined as the ratio of the rate of cleavage of the targeted RNA over the rate of cleavage of non-targeted RNA. This cleavage mechanism is dependent upon factors additional to those involved in base pairing. Thus, it is thought that the specificity of action of a ribozyme is greater than that of antisense oligonucleotide binding the same RΝA site.
This class of chemicals exhibits a high degree of specificity for cleavage of the intended target mRΝA. Consequently, the ribozyme agent will only affect cells expressing that particular gene, and will not be toxic to normal tissues.
The invention can be used to treat or prevent (prophylactically) psoriasis, asthma and other inflammatory diseases, and restenosis or other cardiovascular conditions, including hypertension. The preferred administration protocol is in vivo administration to reduce the level of those genes and the encoded mRΝAs noted above.
Thus, in the first aspect, the invention features an enzymatic RΝA molecule (or ribozyme) which cleaves mRΝA associated with development or maintenance of a psoriatic or asthmatic condition, e.g. , mRΝA encoding TΝF-α, IL-5, IL-1, IL-3, IL-4, IL-6, IL-8, glycerol transferase, selectins, E-selectin, MEL-14, ICAM-1, ELAM-1, VCAM-1, GMP-140, MAM, TGFα, TΝFαR, IL-lR, α-, β- or γ-interferon, GM-CSF and protein kinase C, and in particular, those mRNA targets disclosed in Table 1.
Table 1
TΝF-α mRΝA Nucleotide
Number Sequence SEQ.ID.NO.
27 CAGCAGAGGACCAGCUA ID.NO. 01
56 GCAACUACAGACCC ID.NO. 02
70 CCCCUGAAAACAACCCUCAGACGC ID.NO. 03 94 CACAUCCCCUGACAAGCUGCCAGGCAGG ID.NO. 04
134 CACAUACUGACCCA ID.NO. 05
157 CCUCUCUCCCCUGGAAAGG ID.NO. 06 176 ACACCAUGAGCACUGAAAGCAUGAUCCGGGAC
253 GCCCCAGGGCUCCAGGC
270 GGUGCUUGUUCCUCAGCCUCUUC
311 CCACCACGCUCUUC 362 AAGAGUUCCCCAG
408 AGUCAGAUCAUCUUCUCGAACCCCGAGUGACAA
446 UAGCCCAUGUUGUAGCAAACCCUCAAGCUGAGGG
574 UACCUCAUCUACUCC
599 UCAAGGGCCAAGGCUGCCCCUC 621 CACCCAUGUGCUCCUCACCCA
652 CGCAUCGCCGUCUCCUACCAGACCAA
694 GCCAUCAAGAG
824 CCGACUAUCUCGACUUUGCC
869 UCAUUGCCCUGUGAGGAGGACGAACAUCCAACCU 991 CUUAGGGUCGGAACCCAA
1043 AAACCUGGGAUUCAGGAA
1084 CUGGCAACCACUAAGAAU
1115 UCCAGAACUCACUGG
1187 UGGCCAGAAUGCUGCAGGACUUGAGA 1213 AGACCUCACCUAGAAAUUGACACAAGU
1276 CUUCCUUGAGA
1322 UCUAUUUAUGUUUGCACUUGUG
1344 AUUAUUUAUUAUUUAUUUAUUAUUUAUUUAUUUA
1378 CAGAUGAAUGUAUUUAUUU 1402 ACCGGGGUAUCC
1419 GACCCAAUGUAGGAGCUGCCUUGGCUCAG
1495 AGCCCCCUGGC
1513 CCUUCUUUUGAUUAUGUUUUUUAAAAUAUUUAUC
1557 GUCUAAACAAUGCU 1587 GUCACUCAUUGCUGAG
1639 CUACUAUUCAGU
1656 GAAAUAAAGUUUGCUU
IL-5 mRNA Nucleotide Number Sequence SEP.ID.NO.
10 CUUUGCCAAAGGCAAAC ID.NO. 39 33 CGUUUCAGAGCCAUGAGGAUGC ID.NO. 40
61 AUUUGAGUUUGCUAGCUCUUGGAGCUG ID.NO. 41
88 CCUACGUGUAUGCCAUCC ID.NO. 42
139 AGACCUUGGCACUG ID.NO. 43 158 UACUCAUCGAACUCUGCUGAUA ID.NO. 44
209 UGUACAUAAAAA ID.NO. 45
275 AAACUGUGCAA ID.NO. 46
299 AAAGACUAUUCAAAAACUUGUCC ID.NO. 47
370 AGACGGAGAGUAAACCAAUUCCUAGACUACCUGC ID.NO. 48 522 AAGAAAGAGUCA ID.NO. 49
558 ACUUCAGAGGGAAAG ID.NO. 50
578 AUUUCAGGCAUACUGACACUUUGCCAGAAAGCA ID.NO. 51
635 AUAUCAGAAUCA ID.NO. 52
667 CAAAAUUGAUAUACUUUUUUCUUAUUUAA ID.NO. 53 738 GAAAUGGUUAAGAAUUUG ID.NO. 54
In a second aspect, the invention features and enzymatic RNA molecule which cleaves mRNA associated with development and maintenance of osteoarthritis or other pathological conditions which are mediated by metalloproteinase activation. The preferred administration protocol is in vivo administration to reduce the level of stromelysin activity.
Thus, in this aspect, the invention features an enzymatic RNA molecule (or ribozyme) which cleaves mRNA associated with development or maintenance of an arthritic condition, e.g. , mRNA encoding stromelysin, and in particular, those mRΝA targets disclosed in Table 2. Table 2
Nucleotide
Number Sequence SEQ. ID. NO.
20 UAGAGCUAAGUAAAGCCAG ID.NO. 01 126 ACACCAGCAUGAA ID.NO. 02
147 AGAAAUAUCUAGA ID.NO. 03
171 ACCUCAAAAAAGAUGUGAAACAGU ID.NO. 04 240 AAAUGCAGAAGUUC ID.NO. 05
287 GACACUCUGGAGGUGAUGCGCAAGCCCAGGUGU ID.NO. 06
327 CUGAUGUUGGUCACUUCAGAAC ID.NO. 07
357 GCAUCCCGAAGUGGAGGAAAACCCACCUUACAU ID.NO. 08 402 AUUAUACACCAGAUUUGCCAAAAGAUG ID.NO. 09
429 CUGUUGAUUCUGCUGUUGAGA ID.NO. 10
455 CUGAAAGUCUGGGAAGAGGUGA ID.NO. 11
513 CUGAUAUAAUGA ID.NO. 12
592 UGCCUAUGCCCC ID.NO. 13 624 AUGCCCACUUUGAUGAUGAUGAACAAUGGACA ID.NO. 14
679 AUUUCUCGUUGCUGCUCAUG ID.NO. 15
725 CACUCAGCCAACACUGA ID.NO. 16
801 AAGAUGAUAUAAAUGGCAUUCAGUCC ID.NO. 17
827 CUCUAUGGACCUCCCCCUGACUCCCCU ID.NO. 18 859 CCCCCUGGUACCCA ID.NO. 19
916 UCCUGCUUUGUCCUUUGAUGCUGUCAGCAC ID.NO. 20
958 AAUCCUGAUCUUUAAAGA ID.NO. 21
975 CAGGCACUUUUGGCGCAAAUCCC ID.NO. 22
1018 AUUGCAUUUGAUCUCUUCAUUUUGGCCAUC ID.NO. 23 1070 GCAUAUGAAGUUA ID.NO. 24
1203 AAAUCGAUGCAGCCAUUUCUGA ID.NO. 25
1274 UUUGAUGAGAAGAGAAAUUCCAUGGAGC ID.NO. 26
1302 CAGGCUUUCCCAAGCAAAUAGCUGAAGAC ID.NO. 27
1420 CCCAAAUGCAAAG ID.NO. 28 1485 AUGUAGAAGGCACAAUAUGGGCACUUUAAA ID.NO. 29
1623 UCUUGCCGGUCAUUUUUAUGUUAU ID.NO. 30
1665 GCUGCUGCUUAGC ID.NO. 31
1733 CAACAGACAAGUGACUGUAUCU ID.NO. 32
1769 CUUAUUUAAUA ID.NO. 33
In the third aspect, the invention features an enzymatic RNA molecule (or ribozyme) which cleaves mRNA associated with development or maintenance of a restenotic condition, e.g. , mRNA encoding c-myb (or other mRNAs noted above) , and in particular, those mRNA targets disclosed in Table 3. Table 3
Nucleotide
Number Seguence Seq. ID. No. 1 GGCGGCAGCGCCCUGCCGACGCCGGGG ID.NO.01
77 CCGCGGCUCUCGGC ID.NO.02
111 GCCAUGGCCCGAA ID.NO.03
129 CGGCACAGCAUAUAUAGCAGUGACGAGGA ID.NO.04
165 GACUUUGAGAUGUGUGACCAUGACUAUGAUGGG ID.NO.05 211 CUGGAAAGCGUC ID.NO.06
248 GGAAGAGGAUGAAAAACUGAAGAAG ID.NO.07
267 GAAGAACUGGUGGAACAGAAUGGAAC ID.NO.08
299 CUGGAAAGUUAUUGCCAA ID.NO.09
323 CCCGAAUCGAACAGAUGUGCAG ID.NO.10 362 GAAAGUACUAAACCCUGAG ID.NO.11
394 CUUGGACCAAAGAAGAAGAUCAGAGAGUGAUA ID.NO.12
433 ACAGAAAUACGGUCCGAAACGUUGGUCUG ID.NO.13
463 UUAUUGCCAAGCACUUAAAGGGGAGAAUUGGAA ID.NO.14
527 GAAUCCAGAAGUUAAGAA ID.NO.15 563 GGAAGACAGAAUUAUUUACCAGGCACA ID.NO.16
590 CAAGAGACUGGGGAACAGAU ID.NO.17
616 AAAUCGCAAAGCUA ID.NO.18
636 GGACGAACUGAUAAUGCUAUCAAGAACC ID.NO.19
664 ACUGGAAUUCUACAAUGCGUCGGAAGGUCGAACA ID.NO.20 732 CAGCCAGCAGUGGCCACAA ID.NO.21
768 CAUUUGAUGGGUUUUGCUCAGGCUCCGCCUACA ID.NO.22
801 GCUCAACUCCCUGCCACUGGCCAGCCC ID.NO.23
834 AACAACGACUAUUCCUAUUACCACA ID.NO.24
870 CAAAAUGUCUCCAGUCAUGUUCCAUACCCU ID.NO.25 914 AAAUAUAGUCAAUGUCCCUCAGCCAGCUGCCGCA ID.NO.26
955 AGAGACACUAUAAUGAUGAAGACCCUGAGAAGGA ID.NO.27
989 AAAGCGAAUAAAGGAAUUAGAAUUG ID.NO.28
3179 CGGUGUACUUACUGCC ID.NO.29
1036 AGCUAAAAGGACAGCAGGUGCUACCAACACAGAA ID.NO.30 1086 CCCGGGUGGCACAGCACCACCAUUGCCGACCACA ID.NO.31
1162 AACACCACUCCACUCCAUCUCUGCCAGCGGAUCC ID.NO.32
1204 UACCUGAAGAAA ID.NO.33 1236 AUGAUCGUCCACCAGGGCACCAUU ID.NO.34
1291 CAGAAACACUCCAAUUUA ID.NO.35
1343 AAACUCAGACU ID.NO.36
1359 AUGCCUUCUUUAAC ID.NO.37 1405 UUACAACACCA ID.NO.38
1440 ACUCAAAAGGAAAAUACUGUUUUUAGAACCC ID.NO.39
1471 CAGCUAUCAAAAGGUCAAUCUUAGAAAGCU ID.NO.40
1501 CUCCAAGAACUCCUACACCAUUCAA ID.NO.41
1526 ACAUGCACUUGCAGCUCAAGAA ID.NO.42 1554 UACGGUCCCCUGAAGAUGCUACCUCAGA ID.NO.43
1582 CACCCUCUCAUCUAGUAGAAGAUCUGCAGGA ID.NO.44
1618 UCAAACAGGAAUCUGAUGAAUCUGGA ID.NO.45
1660 AAGAAAAUGGA ID.NO.46
1676 CUUACUGAAGAAAAUCAAACAAGA ID.NO.47 1705 AAUCUCCAACUGAUAAAUCAG ID.NO.48
1738 GCUCACACCACUGGGA ID.NO.49
1789 CCUCGCCUGUGCGAGAUGCACCGAAUAUUC ID.NO.50
1838 GGCACCAGCAUCAGAAGAUGAAGAC ID.NO.51
1876 CAUUUACAGUACC ID.NO.52 1900 CCCUGGCGAGCCCCUUGCA ID.NO.53
1919 GCCUUGUAGCAGUACCUGGGA ID.NO.54
1984 GUCAAGCUCGUAAAUACGUGAA ID.NO.55
2067 GAACAGUUCAA ID.NO.56
2106 AUGAAACUUUUCAU ID.NO.57 2229 AAAAUAAAUAACAGUC ID.NO.58
2265 UGAAUUGUAGCC ID.NO.59
2282 UUAAUAUCUUAAU ID.NO.60
2325 AUUUAUCUGGUAUUUUAAAGGAUCCAACAGAUC ID.NO.61
2410 CCAGUAUUUCA ID.NO.62 2426 CUCGAUCACUAAACAUAUG ID.NO.63
2445 CAUAUAUUUUUAAAAAUC ID.NO.64
2695 UGCUAUGGUCUUAGCCU ID.NO.65
2726 AGUAUCAGAGG ID.NO.66
2776 UAGGUAAUUGACUAU ID.NO.67 2798 UAUUUCAGACUUUUUAAUUUUAUAUAUAUAUACA ID.NO.68
2847 CAAUACAUUUGAAAACUUGUUUGGGAGACUCUGC ID.NO.69
2891 GUGGUUUUUUUGUUAUUGUUGGUUU ID.NO.70 2935 UUCUUUUUUGGGAGAU ID.NO.71
2967 CUAUGUUUUGUUUUG ID.NO.72
2987 AGCCUGACUGUUUUAUA ID.NO.73
3016 UCGAUUUGAUC ID.NO.74 3072 UGGAUCCUGUGUU ID.NO.75
3111 UUGAUAGCCAGUCACUGCCUUAAGA ID.NO.76
3136 ACAUUUGAUGCAAGAUGGCCAGCACU ID.NO.77
In a fourth aspect, the invention features an enzymatic RNA molecule (or ribozyme) which cleaves mRNA associated with development or maintenance of a cardiovascular condition, e.g. , mRNA encoding ACE or
ECE, and in particular, those mRNA targets disclosed in Table 4. Table 4
Nucleotide
Number Sequence
38 GCUACUGCAGGACUUCCCAGC 59 CUCCUCUUCCUGCUGCUCGCUAGG
105 GCCAGGAGGCAUC
120 AACAGGUGACAGUCACCCAUG
210 CCCAGAGCCCAAACCUGGUGA
247 CAGCAAGUUUGUGGAGGAAUAUGA 271 CCGGACAUCCCA
292 GAACGAGUAUGCCGAGGCC
311 AACUGGAACUACAACAC
341 GAGACCAGCAAGAUUCUGCUG
369 ACAUGCAAAUAG 387 ACACCCUGAAGUACGGCACCCAG
424 GUGAACCAGUUGCAGAACACCACUA
474 AGGACCUAGAA
491 GCGCUGCCUGCCCAGGAGCUGGAG
515 GAGUACAACAAGAUCCUGUUGGA 535 GGAUAUGGAAACCACCUACAGC
564 CUGUGUGCCACCCGAAUGGC
598 CGAGCCAGAUCUGACGAAUGUGAUG 627 CAUCCCGGAAAUAUGAAGACCUG
646 CCUGUUAUGGGCAUGGG
667 CUGGCGAGACAAGGCGGG
706 CCCGAAAUACG 725 AUCAACCAGG
755 GUAGAUGCAGGGGACUC
775 AGGUCUAUGUACGAGACACCAUCC
831 AGCUGCAGCCACUCUACCUCAAC
844 CUGCAUGCCUACGUGCGCCG 899 CAGCAUCAAC
921 CCAUUCCUGCUCAC
956 CAGACCUGGUCCAAC
971 AUCUAUGACUUGGUGG
996 CUUCAGCCCCCUCGAUGGAC 1015 CACCACAGAGGCUAUGCUAA
1040 GGCUGGACGC
1054 GAGGAUGUUUAAGGAGGCUGAUGA
1071 CUGAUGAUUUCUUCACCUCC
1107 UGCCUCCUGAGUUCUGGAACA 1127 AAGUCGAUGCUGGAGAAG
1173 ACGCCUCGGCCUGGGACUUCUACAA
1203 AGGACUUCCGGAUCAAGCAGUGCA
1227 CCACCGUGAACUUGGAGGACCUGG
1275 AC UCCAGUAUUUC 1291 GCAGUACAAAGACUUACCUGUGG
1315 CUUGAGGGAGGGUGCCAACC
1335 CCGGCUUCCAUGAGGCCAUUGG
1358 GACGUGCUAG
1376 GUGUCUACGCCCAAGCACCUGCACA 1401 GUCUCAACCUGCUGAGCAG
1429 CAGCGACGAGCAUGACAUCAAC
1450 CUUUCUGAUGAAGAUGGCCCUUG
1469 CUUGACAAGAUCGCC
1500 ACCUCGUCGAUCAGUGGCG 1536 GAAGCAUCACC
1553 AACUAUAACCAGGAGUGG
1565 GCCUCAGGCUGAAGUA 1591 CCAGGGCCUCUGCCCCCCAG ID.NO. 57
1616 AGGACUCAAGGUGAC ID.NO. 58
1630 CUUUGACCCAGGGGCC ID.NO. 59
1662 CUAGCGUGCCUUAC ID.NO. 60 1699 CAUCCAGUUCCAGUUCCACG ID.NO. 61
1701 GUUCCACGAGGCACUG ID.NO. 62
1749 GCCCCCUGCACAAGUGUGACAUC ID.NO. 63
1771 CUACCAGUCCAAGGAG ID.NO. 64
1894 GAGCUACUUCAAGCUGCUGG ID.NO. 65 1915 GGACUGGCUCCGCACGG ID.NO. 66
1968 AGUACAACUGGACGCC ID.NO. 67
1984 GAACUCCGCUCGCUCAGAAGG ID.NO. 68
2005 GCCCCUCCCAGACAG ID.NO. 69
2046 ACCUGGAUGCGCAGCA ID.NO. 70 2076 AGUGGCUGCUGC ID.NO. 71
2101 CGCCCUGCUGGUAGCCACCC ID.NO. 72
2147 AUCCGCCACCGCAGCCUCC ID.NO. 73
2212 ACACUCCUGAGGUGACCCGG ID.NO. 74
2316 GCCCACCCUGC ID.NO. 75 2337 CUGUCCCUGUCCCCCUCCCC ID.NO. 76
2365 CUCCAGACCACC ID.NO. 77
2386 AGCCCCUUCUCCCAGCACAC ID.NO. 78
2408 CUGCCUGACACUGAGCCC ID.NO. 79
2426 CACCUCUCCAAGUCUCUCUG ID.NO. 80 2446 UGAAUACAAUUAAAGGUCCUG ID.NO. 81
By "enzymatic RNA molecule" it is meant an RNA molecule which has complementarity in a substrate binding region to a specified mRNA target, and also has an enzymatic activity which is active to specifically cleave that mRNA. That is, the enzymatic RΝA molecule is able to intermolecularly cleave mRΝA and thereby inactivate a target mRΝA molecule. This complementarity functions to allow sufficient hybridization of the enzymatic RΝA molecule to the target RΝA to allow the cleavage to occur. One hundred percent complementarity is preferred, but complementarity as low as 50-75% may also be useful in this invention. For in vivo treatment, complementarity between 30 and 45 bases is preferred. In preferred embodiments, the enzymatic RNA molecule is formed in a hammerhead motif, but may also be formed in the motif of a hairpin, hepatitis delta virus, group I intron or RNaseP-like RNA (in association with an RNA guide sequence) . Examples of such hammerhead motifs are described by Rossi et al., 8 Aids Research and Human Retroviruses 183, 1992, of hairpin motifs by Hampel and Tritz, 28 Biochemistry 4929, 1989; and Hampel et al., 18 Nucleic Acids Research 299, 1990, and an example of the hepatitis delta virus motif is described by Perrotta and Been, 31 Biochemistry 16,
1992, of the RNaseP motif by Guerrier-Takada et al., 35 Cell 849, 1983, and of the group I intron by Cech et al., U.S. Patent 4,987,071. These specific motifs are not limiting in the invention and those skilled in the art will recognize that all that is important in an enzymatic RNA 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.
In a related aspect, the invention features a mammalian cell which includes an enzymatic RNA molecule as described above. Preferably, the mammalian cell is a human cell.
In another related aspect, the invention features an expression vector which includes nucleic acid encoding an enzymatic RNA molecule described above, located in the vector, e.g. , in a manner which allows expression of that enzymatic RNA molecule within a mammalian cell. In yet another related aspect, the invention features a method for treatment of a disease noted above by administering to a patient an enzymatic RNA molecule as described above. The invention provides a class of chemical cleaving agents which exhibit a high degree of specificity for the mRNA causative of a psoriatic or arthritic or cardiovascular condition. Such a condition includes any measurable indication of susceptibility to cardiac problems, and thus includes predisposition to such conditions or cardiovascular disease. Such enzymatic RNA molecules can be delivered exogenously or endogenously to infected cells. In the preferred hammerhead motif the small size (less than 40 nucleotides, preferably between 32 and 36 nucleotides in length) of the molecule allows the cost of treatment to be reduced.
The smallest ribozyme delivered for any type of treatment reported to date (by Rossi et al. , 1992, supra) is an in vi tro transcript having a length of 142 nucleotides. Synthesis of ribozymes greater than 100 nucleotides in length is very difficult using automated methods, and the therapeutic cost of such molecules is prohibitive. Delivery of ribozymes by expression vectors is primarily feasible using only ex vivo treatments. This limits the utility of this approach. In this invention, an alternative approach uses smaller ribozyme motifs (e.g. , of the hammerhead structure, shown generally in Fig. 1) and exogenous delivery. The simple structure of these molecules also increases the ability of the ribozyme to invade targeted regions of the mRNA structure. Thus, unlike the situation when the hammerhead structure is included within longer transcripts, there are no non-ribozyme flanking sequences to interfere with correct folding of the ribozyme structure, as well as complementary binding of the ribozyme to the mRNA target. The enzymatic RNA molecules of this invention can be used to treat psoriatic or pre-psoriatic, asthmatic or pre-asthmatic, arthritic or prearthritic, stenotic or prestenotic conditions. Such treatment can also be extended to other related genes in nonhuman primates. Affected animals can be treated at the time of disease risk or detection, or in a prophylactic manner. This timing of treatment will reduce the chance of further disease damage. Ribozymes of this invention may be used as diagnostic tools to examine genetic drift and mutations within diseased cells. The close relationship between ribozyme activity and the structure of the target RNA allows the detection of mutations in any region of the molecule which alters the base-pairing and three- dimensional structure of the target RNA. By using multiple ribozymes described in this invention, one may 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 ribozymes may 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 may be defined as important mediators of the disease. These experiments will lead to better treatment of the disease progression by affording the possibility of combinational therapies (e.g. , multiple ribozymes targeted to different genes, ribozymes coupled with known small molecule inhibitors, or intermittent treatment with combinations of ribozymes and/or other chemical or biological molecules) .
Other features and advantages of the invention will be apparent from the following description of the preferred embodiments thereof, and from the claims. Description of the Preferred Embodiments The drawing will first briefly be described. Drawing
Fig. 1 is a diagrammatic representation of a hammerhead motif ribozyme showing stems I, II and III (marked (I) , (II) and (III) respectively) interacting with a target region. The 5' and 3' ends of both ribozyme and target are shown. Dashes indicate base- paired nucleotides. Target Sites
Ribozymes targeting selected regions of mRNA associated with a selected disease are chosen to cleave the target RNA in a manner which preferably inhibits translation of the RNA. Genes are selected such that inhibition of translation will preferably inhibit cell replication, e.g. , by inhibiting production of a necessary protein. Selection of effective target sites within these critical regions of mRNA entails testing the accessibility of the target RNA to hybridization with various oligonucleotide probes. These studies can be performed using RNA probes and assaying accessibility by cleaving the hybrid molecule with RNaseH (see below) . Alternatively, such a study can use ribozyme probes designed from secondary structure predictions of the mRNAs, and assaying cleavage products by polyacrylamide gel electrophoresis (PAGE) , to detect the presence of cleaved and uncleaved molecules.
The following is but one example of a method by which suitable target sites can be identified and is not limiting in this invention. Generally, the method involves identifying potential cleavage sites for a hammerhead ribozyme, and then testing each of these sites to determine their suitability as targets by ensuring that secondary structure formation is minimal. The mRNA sequences are compared in an appropriate target region. Putative ribozyme cleavage sites are found. These sites represent the preferable sites for hammerhead ribozyme cleavage within these target mRNAs.
Short RNA substrates corresponding to each of the mRNA sites are designed. Each substrate is composed of two to three nucleotides at the 5' and 3' ends that will not base pair with a corresponding ribozyme recognition region. The unpaired regions flanked a central region of 12-14 nucleotides to which comple¬ mentary arms in the ribozyme are designed. The structure of each substrate sequence is predicted using a standard PC fold computer program. Sequences which give a positive free energy of binding are accepted. Sequences which give a negative free energy are modified by trimming one or two bases from each of the ends. If the modified sequences are still predicted to have a strong secondary structure, they are rejected.
After substrates are chosen, ribozymes are designed to each of the RNA substrates. Ribozyme folding is also analyzed using PC fold.
Ribozyme molecules are sought which form hammerhead motif stem II (see Fig. 1) regions and contain flanking arms which are devoid of intramolecular base pairing. Often the ribozymes are modified by trimming a base from the ends of the ribozyme, or by introducing additional base pairs in stem II to achieve the desired fold. Ribozymes with incorrect folding are rejected. After substrate/ribozyme pairs are found to contain correct intramolecular structures, the molecules are folded together to predict intermolecular interactions. A schematic representation of a ribozyme with its coordinate base pairing to its cognate target sequence is shown in Fig. 1.
Those targets thought to be useful as ribozyme targets can be tested to determine accessibility to nucleic acid probes in a ribonuclease H assay (see below) . This assay provides a quick test of the use of the target site without requiring synthesis of a ribozyme. It can be used to screen for sites most suited for ribozyme attack. Synthesis of Ribozymes Ribozymes useful in this invention can be produced by gene transcription as described by Cech, supra, or by chemical synthesis. Chemical synthesis of RNA is similar to that for DNA synthesis. The additional 2'-OH group in RNA, however, requires a different protecting group strategy to deal with selective 3'-5' internucleotide bond formation, and with RNA susceptibility to degradation in the presence of bases. The recently developed method of RNA synthesis utilizing the t-butyldimethylsilyl group for the protection of the 2' hydroxyl is the most reliable method for synthesis of ribozymes. The method reproducibly yields RNA with the correct 3'-5' internucleotide linkages, with average coupling yields in excess of 99%, and requires only a two-step deprotection of the polymer.
A method, based upon H-phosphonate chemistry of phosphoroamidites gives a relatively lower coupling efficiency than a method based upon phosphoroamidite chemistry. This is a problem for synthesis of DNA as well. A promising approach to scale-up of automatic oligonucleotide synthesis has been described recently for the H-phosphonates. A combination of a proper coupling time and additional capping of "failure" sequences gave high yields in the synthesis of oligodeoxynucleotides in scales in the range of 14 μmoles with as little as 2 equivalents of a monomer in the coupling step. Another alternative approach is to use soluble polymeric supports (e.g. , polyethylene glycols) , instead of the conventional solid supports. This method can yield short oligonucleotides in hundred milligram quantities per batch utilizing about 3 equivalents of a monomer in a coupling step. Various modifications to ribozyme structure can be made to enhance the utility of ribozymes. Such modifications will enhance shelf-life, half-life in vi tro, stability, and ease of introduction of such ribozymes to the target site, e.g. , to enhance penetration of cellular membranes, and confer the ability to recognize and bind to targeted cells.
Exogenous delivery of ribozymes benefits from chemical modification of the backbone, e.g. , by the overall negative charge of the ribozyme molecule being reduced to facilitate diffusion across the cell membrane. The present strategies for reducing the oligonucleotide charge include: modification of internucleotide linkages by methylphosphonates, use of phosphoramidites, linking oligonucleotides to positively charged molecules, and creating complex packages composed of oligonucleotides, lipids and specific receptors or effectors for targeted cells. Examples of such modifications include sulfur-containing ribozymes containing phosphorothioates and phosphorodithioates as internucleotide linkages in RNA. Synthesis of such sulfur-modified ribozymes is achieved by use of the sulfur-transfer reagent, 3H-l,2-benzenedithiol-3-one 1,1- dioxide. Ribozymes may also contain ribose modified ribonucleotides. Pyrimidine analogues are prepared from uridine using a procedure employing diethylamino sulphur trifluoride (DAST) as a starting reagent. Ribozymes can also be either electrostatically or covalently attached to polymeric cations for the purpose of reducing charge. The polymer can be attached to the ribozyme by simply converting the 3'-end to a ribonucleoside dialdehyde which is obtained by a periodate cleavage of the terminal 2',3'-cis diol system. Depending on the specific requirements for delivery systems, other possible modifications may include different linker arms containing carboxyl, amino or thiol functionalities. Yet further examples include use of methylphosphonates and 2'-O-methylribose and 5' or 3' capping or blocking with m7GpppG or m3 2'2,7GpppG.
For example, a kinased ribozyme is contacted with guanosine triphosphate and guanyltransferase to add an m3G cap to the ribozyme. After such synthesis, the ribozyme can be gel purified using standard procedure. To ensure that the ribozyme has the desired activity, it may be tested with and without the 5' cap using standard procedures to assay both its enzymatic activity and its stability.
Synthetic ribozymes, including those containing various modifiers, can be purified by high pressure liquid chromatography (HPLC) . Other liquid chromatography techniques, employing reverse phase columns and anion exchangers on silica and polymeric supports may also be used.
There follows an example of the synthesis of one ribozyme. A solid phase phosphoramidite chemistry is employed. Monomers used are 2'-tert-butyl- dimethylsilyl cyanoethylphosphoramidities of uridine, N- benzoyl-cytosine, N-phenoxyacetyl adenosine and guanosine (Glen Research, Sterling, VA) . Solid phase synthesis is carried out on either an ABI 394 or 380B DNA/RNA synthesizer using the standard protocol provided with each machine. The only exception is that the coupling step is increased from 10 to 12 minutes. The phosphoramidite concentration is 0.1 M. Synthesis is done on a 1 μmole scale using a 1 μ ole RNA reaction column (Glen Research) . The average coupling efficiencies are between 97% and 98% for the 394 model, and between 97% and 99% for the 380B model, as deter¬ mined by a calorimetric measurement of the released trityl cation.
Blocked ribozymes are cleaved from the solid support (e.g. , CPG) , and the bases and diphosphoester moiety deprotected in a sterile vial by dry ethanolic ammonia (2 mL) at 55°C for 16 hours. The reaction mixture is cooled on dry ice. Later, the cold liquid is transferred into a sterile screw cap vial and lyophilized.
To remove the 2'-tert-butyl-dimethylsilyl groups from the ribozyme, the residue is suspended in 1 M tetra-n-butylammonium fluoride in dry THF (TBAF) , using a 20 fold excess of the reagent for every silyl group, for 16 hours at ambient temperature (about 15- 25°C) . The reaction is quenched by adding an equal volume of sterile 1 M triethylamine acetate, pH 6.5. The sample is cooled and concentrated on a SpeedVac to half the initial volume.
The ribozymes are purified in two steps by HPLC on a C4 300 A 5 mm DeltaPak column in an acetonitrile gradient.
The first step, or "trityl on" step, is a separation of 5'-DMT-protected ribozyme(s) from failure sequences lacking a 5'-DMT group. Solvents used for this step are: A (0.1 M triethylammonium acetate, pH 6.8) and B (acetonitrile). The elution profile is: 20% B for 10 minutes, followed by a linear gradient of 20% B to 50% B over 50 minutes, 50% B for 10 minutes, a linear gradient of 50% B to 100% B over 10 minutes, and a linear gradient of 100% B to 0% B over 10 minutes. The second step is a purification of a completely deblocked ribozyme by a treatment of 2% trifluoroacetic acid on a C4 300 A 5 mm DeltaPak column in an acetonitrile gradient. Solvents used for this second step are: A (0.1 M triethylammonium acetate, pH 6.8) and B (80% acetonitrile, 0.1 M triethylammonium acetate, pH 6.8). The elution profile is: 5% B for 5 minutes, a linear gradient of 5% B to 15% B over 60 minutes, 15% B for 10 minutes, and a linear gradient of 15% B to 0% B over 10 minutes. The fraction containing ribozyme is cooled and lyophilized on a SpeedVac. Solid residue is dissolved in a minimum amount of ethanol and sodium perchlorate in acetone. The ribozyme is collected by centrifugation, washed three times with acetone, and lyophilized. Expression Vector
While synthetic ribozymes are preferred in this invention, those produced by expression vectors can also be used. In designing a suitable ribozyme expression vector the following factors are important to consider. The final ribozyme must be kept as small as possible to minimize unwanted secondary structure within the ribozyme. A promoter (e.g. , the human cytomegalovirus immediate early promoter or human beta actin promoter) should be chosen to be a relatively strong promoter, and expressible both in vitro and in vivo (e.g. , the human cytomegalovirus immediate early promoter or human beta actin promoter) . Such a promoter should express the ribozyme at a level suitable to effect production of enough ribozyme to destroy a target RNA, but not at too high a level to prevent other cellular activities from occurring (unless cell death itself is desired) . A hairpin at the 5' end of the ribozyme is useful to ensure that the required transcription initiation sequence (GG or GGG or GGGAG) does not bind to some other part of the ribozyme and thus affect regulation of the transcription process. The 5' hairpin is also useful to protect the ribozyme from 5'-3' exonucleases. A selected hairpin at the 3' end of the ribozyme gene is useful since it acts as a transcription termination signal, and protects the ribozyme from 3'-5' exonuclease activity. One example of a known termination signal is that present on the T7 RNA polymerase system. This signal is about 30 nucleotides in length. Other 3' hairpins of shorter length can be used to provide good termination and RNA stability. Such hairpins can be inserted within the vector sequences to allow standard ribozymes to be placed in an appropriate orientation and expressed with such sequences attached. Poly(A) tails are also useful to protect the 3' end of the ribozyme. These can be provided by either including a poly(A) signal site in the expression vector (to signal a cell to add the poly(A) tail in vivo) , or by introducing a poly(A) sequence directly into the expression vector. In the first approach the signal must be located to prevent unwanted secondary structure formation with other parts of the ribozyme. In the second approach, the poly(A) stretch may reduce in size over time when expressed in vivo, and thus the vector may need to be checked over time. Care must be taken in addition of a poly(A) tail which binds poly(A) binding proteins which prevent the ribozyme from acting. Ribozyme Testing Once the desired ribozymes are selected, synthesized and purified, they are tested in kinetic and other experiments to determine their utility. An example of such a procedure is provided below. Preparation of Ribozyme Crude synthetic ribozyme (typically 350 μg at a time) is purified by separation on a 15% denaturing polyacrylamide gel (0.75 mm thick, 40 cm long) and visualized by UV shadowing. Once excised, gel slices containing full length ribozyme are soaked in 5 ml gel elution buffer (0.5 M NH4OAc, 1 mM EDTA) overnight with shaking at 4°C. The eluent is desalted over a C-18 matrix (Sep-Pak cartridges, Millipore, Milford, MA) and vacuum dried. The dried RNA is resuspended in 50-100 μl TE (TRIS 10 mM, EDTA 1 mM, pH 7.2). An aliquot of this solution is diluted 100-fold into 1 ml TE, half of which is used to spectrophotometrically quantitate the ribozyme solution. The concentration of this dilute stock is typically 150-800 nM. Purity of the ribozyme is confirmed by the presence of a single band on a denaturing polyacrylamide gel.
A ribozyme may advantageously be synthesized in two or more portions. Each portion of a ribozyme will generally have only limited or no enzymatic activity, and the activity will increase substantially (by at least 5-10 fold) when all portions are ligated (or otherwise juxtaposed) together. A specific example of hammerhead ribozyme synthesis is provided below.
The method involves synthesis of two (or more) shorter "half" ribozymes and ligation of them together using T4 RNA ligase. For example, to make a 34 mer ribozyme, two 17 ers are synthesized, one is phosphorylated, and both are gel purified. These purified 17 mers are then annealed to a DNA splint strand complementary to the two 17 mers. (Such a splint is not always necessary.) This DNA splint has a sequence designed to locate the two 17 mer portions with one end of each adjacent each other. The juxtaposed RNA molecules are then treated with T4 RNA ligase in the presence of ATP. The 34 mer RNA so formed is then HPLC purified.
Preparation of Substrates Approximately 10-30 pmoles of unpurified substrate is radioactively 5' end-labeled with T4 polynucleotide kinase using 25 pmoles of [γ-32P] ATP. The entire labeling mix is separated on a 20% denaturing polyacrylamide gel and visualized by autoradiography. The full length band is excised and soaked overnight at 4°C in 100 μl of TE (10 mM Tris-HCl pH 7.6, 0.1 mM EDTA) .
Kinetic Reactions
For reactions using short substrates (between 8 and 16 bases) a substrate solution is made IX in assay buffer (75 mM Tris-HCl, pH 7.6; 0.1 mM EDTA, 10 mM MgCl2) such that the concentration of substrate is less than 1 nM. A ribozyme solution (typically 20 nM) is made IX in assay buffer and four dilutions are made using IX assay buffer. Fifteen μl of each ribozyme dilution
(i.e., 20, 16, 12, 8 and 4 nM) is placed in a separate tube. These tubes and the substrate tube are pre- incubated at 37°C for at least five minutes.
The reaction is started by mixing 15 μl of substrate into each ribozyme tube by rapid pipetting (note that final ribozyme concentrations are 10, 8, 6, 4, 2 nM) . Five μl aliquots are removed at 15 or 30 second intervals and quenched with 5 μl stop solution (95% formamide, 20 mM EDTA xylene cyanol, and bromphenol blue dyes) . Following the final ribozyme time point, an aliquot of the remaining substrate is removed as a zero ribozyme control.
The samples are separated on either 15% or 20% polyacrylamide gels. Each gel is visualized and quantitated with an Ambis beta scanner (Ambis Systems, San Diego, CA) .
For the most active ribozymes, kinetic analyses are performed in substrate excess to determine K„, and Kcat values.
For kinetic reactions with long RNA substrates (greater than 15 bases in length) the substrates are prepared by transcription using T7 RNA polymerase and defined templates containing a T7 promoter, and DNA encoding appropriate nucleotides of the target RNA. The substrate solution is made IX in assay buffer (75 mM Tris-HCl, pH 7.6; 0.1 mM EDTA; 10 mM MgCl2) and contains 58 nanomolar concentration of the long RNA molecules. The reaction is started by addition of gel purified ribozymes to 1 μM concentration. Aliquots are removed at 20, 40, 60, 80 and 100 minutes, then quenched by the addition of 5 μl stop solution. Cleavage products are separated using denaturing PAGE. The bands are visualized and quantitated with an Ambis beta scanner. Kinetic Analysis A simple reaction mechanism for ribozyme- mediated cleavage is: kx k2
R + S ** [R : S] * [R : P] → R + P k_! where R = ribozyme, S = substrate, and P = products.
The boxed step is important only in substrate excess.
Because ribozyme concentration is in excess over substrate concentration, the concentration of the ribozyme-substrate complex ([R:S]) is constant over time except during the very brief time when the complex is being initially formed, i.e., : dfR:Sl = 0 dt where t = time, and thus:
(R) (S)kx = (RS) (k2 + kx) .
The rate of the reaction is the rate of disappearance of substrate with time:
Rate = ά^ = k2(RS)
Substituting these expressions:
(R) (S) i = l/k2 -d(S) (k2 + k. dt or: -d(S) = k,k, (R) dt
S (k2 + *._,)
Integrating this expression with respect to time yields:
-In S_ = k,k, (R) t S0 (k2 + kx) where S0 = initial substrate. Therefore, a plot of the negative log of fraction substrate uncut versus time (in minutes) yields a straight line with slope: slope = k.k, (R) = kobs (k2 + k where kobs = observed rate constant. A plot of slope (kobs) versus ribozyme concentration yields a straight line with a slope which is: slope = nk-, which is k^,. (k2 + kx) I ,
Using these equations the data obtained from the kinetic experiments provides the necessary information to determine which ribozyme tested is most useful, or active. Such ribozymes can be selected and tested in in vivo or ex vivo systems. Liposo e Preparation
Lipid molecules are dissolved in a volatile organic solvent (CHC13, methanol, diethylether, ethanol, etc.). The organic solvent is removed by evaporation. The lipid is hydrated into suspension with O.lx phosphate buffered saline (PBS) , then freeze-thawed 3x using liquid nitrogen and incubation at room temperature. The suspension is extruded sequentially through a 0.4 μm, 0.2 μm and 0.1 μm polycarbonate filters at maximum pressure of 800 psi. The ribozyme is mixed with the extruded liposome suspension and lyophilized to dryness. The lipid/ribozyme powder is rehydrated with water to one-tenth the original volume. The suspension is diluted to the minimum volume required for extrusion (0.4 ml for 1.5 ml barrel and 1.5 ml for 10 ml barrel) with lxPBS and re-extruded through 0.4 μm, 0.2 μm, 0.1 μm polycarbonate filters. The liposome entrapped ribozyme is separated from untrapped ribozyme by gel filtration chromatography (SEPHAROSE CL-4B, BIOGEL A5M) . The liposome extractions are pooled and sterilized by filtration through a 0.2 μm filter. The free ribozyme is pooled and recovered by ethanol precipitation. The liposome concentration is determined by incorporation of a radioactive lipid. The ribozyme concentration is determined by labeling with 32P. Rossi et al., 1992, supra (and references cited therein) describe other methods suitable for preparation of liposomes.
Examples of other useful liposome preparations which display similar degrees of uptake of both a radioactive lipid marker and an entrapped fluorophore by Vero cells showed different fluorescent staining patterns. Specifically, liposomes composed of DPPG/DPPC/Cholesterol (in a ratio of: 50/17/33) gave a punctate pattern of fluorescence, while DOPE/Egg PC/Cholesterol (30/37/33) gave a diffuse, homogeneous pattern of fluorescence in the cytoplasm. Cell fractionation showed that 80% of the entrapped contents from the DPPG/DPPC/Cholesterol formulation was localized in the membrane fraction, whereas the DOPE/Egg PC/Cholesterol formulation was localized in the cytoplasm. Further characterization of the latter formulation showed that after 3 hours, 70% of the fluorescence was cytoplasmic and 30% was in the membrane. After 24 hours, uptake had increased 5 fold and the liposome contents were distributed 50/50 between the cytoplasmic and membrane fractions.
Liposomes containing 15 ribozymes (32P-labeled) targeted to the HSV ICP4 mRNA were prepared and incubated with the cells. After 24 hours, 25% of the liposome dose was taken up with approximately 60,000 liposomes per cell. Thirty percent of the delivered ribozyme was intact after 24 hours. Cell fractionation studies showed 40% of the intact ribozyme to be in the membrane fraction and 52% of the intact ribozyme to be in the cytoplasmic fraction. In Vivo Assay
The efficacy of action of a chosen ribozyme may be tested in vivo using standard procedures in transformed cells or animals which express the target mRNA. Ribonuclease Protection Assay
The accumulation of target mRNA in cells or the cleavage of the RNA by ribozymes or RNaseH ( in vi tro or in vivo) can be quantified using an RNase protection assay.
In this method, antisense riboprobes are transcribed from template DNA using T7 RNA polymerase (U.S. Biochemical) in 20 μl reactions containing IX transcription buffer (supplied by the manufacturer), 0.2 mM ATP, GTP and UTP, 1 U/μl pancreatic RNase inhibitor (Boehringer Mannheim Biochemicals) and 200 μCi 32P- labeled CTP (800 Ci/mmol, New England Nuclear) for 1 hour at 37°C. Template DNA is digested with 1 U RNase- free DNasel (U.S. Biochemical, Cleveland, OH) at 37°C for 15 minutes and unincorporated nucleotides removed by G-50 SEPHADEX spin chromatography.
In a manner similar to the transcription of antisense probe, the target RNA can be transcribed in vi tro using a suitable DNA template. The transcript is purified by standard methods and digested with ribozyme at 37°C according to methods described later.
Alternatively, afflicted (mRNA-expressing) cells are harvested into 1 ml of PBS) transferred to a 1.5 ml EPPENDORF tube, pelleted for 30 seconds at low speed in a microcentrifuge, and lysed in 70 μl of hybridization buffer (4 M guanidine isothiocyanate, 0.1% sarcosyl, 25 mM sodium citrate, pH 7.5). Cell lysate (45 μl) or defined amounts of in vi tro transcript (also in hybridization buffer) is then combined with 5 μl of hybridization buffer containing 5 x 105 cpm of each antisense riboprobe in 0.5 ml Eppendorf tubes, overlaid with 25 μl mineral oil, and hybridization accomplished by heating overnight at 55°C. The hybridization reactions are diluted into 0.5 ml RNase solution (20 U/ml RNaseA, 2 U/ml RNaseTl, 10 U/ml RNase-free DNasel in 0.4 M NaCl), heated for 30 minutes at 37°C, and 10 μl of 20% SDS and 10 μl of Proteinase K (10 mg/ml) added, followed by an additional 30 minutes incubation at 37°C. Hybrids are partially purified by extraction with 0.5 ml of a 1:1 mixture of phenol/chloroform; aqueous phases are combined with 0.5 ml isopropanol, and RNase- resistant hybrids pelleted for 10 minutes at room temperature (about 20°C) in a microcentrifuge. Pellets are dissolved in 10 μl loading buffer (95% formamide, IX TBE, 0.1% bromophenol blue, 0.1% xylene cylanol) , heated to 95°C for five minutes, cooled on ice, and analyzed on 4% polyacrylamide/7 M urea gels under denaturing conditions.
Ribozyme Stability
The chosen ribozyme can be tested to determine its stability, and thus its potential utility. Such a test can also be used to determine the effect of various chemical modifications (e.g. , addition of a poly(A) tail) on the ribozyme stability and thus aid selection of a more stable ribozyme. For example, a reaction mixture contains 1 to 5 pmoles of 5' (kinased) and/or 3' labeled ribozyme, 15 μg of cytosolic extract and 2.5 mM MgCl2 in a total volume of 100 μl. The reaction is incubated at 37°C. Eight μl aliquots are taken at timed intervals and mixed with 8 μl of a stop mix (20 mM EDTA, 95% formamide) . Samples are separated on a 15% acrylamide sequencing gel, exposed to film, and scanned with an Ambis.
A 3'-labeled ribozyme can be formed by incorporation of the 32P-labeled cordycepin at the 3' OH using poly(A) polymerase. For example, the poly(A) polymerase reaction contains 40 mM Tris, pH 8, 10 mM MgCl2, 250 mM NaCl, 2.5 mM MnCl2, ; 3 μl 32P cordycepin, 500 Ci/mM; and 6 units poly(A) polymerase in a total volume of 50 μl. The reaction mixture is incubated for 30 minutes at 37°C. Effect of Base Substitution Upon Ribozyme Activity
To determine which primary structural characteristics could change ribozyme cleavage of substrate, minor base changes can be made in the substrate cleavage region recognized by a specific ribozyme. For example, the substrate sequences can be changed at the central "C" nucleotide, changing the cleavage site from a GUC to a GUA motif. The Kcat~-_ values for cleavage using each substrate are then analyzed to determine if such a change increases ribozyme cleavage rates. Similar experiments can be performed to address the effects of changing bases complementary to the ribozyme binding arms. Changes predicted to maintain strong binding to the complementary substrate are preferred. Minor changes in nucleotide content can alter ribozyme/substrate interactions in ways which are unpredictable based upon binding strength alone. Structures in the catalytic core region of the ribozyme recognize trivial changes in either substrate structure or the three dimensional structure of the ribozyme/substrate complex.
To begin optimizing ribozyme design, the cleavage rates of ribozymes containing varied arm lengths, but targeted to the same length of short RNA substrate can be tested. Minimal arm lengths are required and effective cleavage varies with ribozyme/substrate combinations.
The cleavage activity of selected ribozymes can be assessed using target mRNA substrates. The assays are performed in ribozyme excess and approximate Kcat/KI.in values obtained. Comparison of values obtained with short and long substrates indicates utility in vivo of a ribozyme. Intracellular Stability of Liposome-delivered Ribozymes
To test the stability of a chosen ribozyme in vivo the following test is useful. Ribozymes are 32P- end labeled, entrapped in liposomes and delivered to target mRNA-containing cells for three hours. The cells are fractionated and ribozyme is purified by phenol/chloroform extraction. Alternatively, cells (lxlO7, T-175 flask) are scraped from the surface of the flask and washed twice with cold PBS. The cells are homogenized by douncing 35 times in 4 ml of TSE (10 mM Tris, pH 7.4, 0.25 M Sucrose, mM EDTA). Nuclei are pelleted at lOOxg for 10 minutes. Subcellular organelles (the membrane fraction) are pelleted at 200,000xg for two hours using an SW60 rotor. The pellet is resuspended in 1 ml of H buffer (0.25 M Sucrose, 50 mM HEPES, pH 7.4). The supernatant contains the cytoplasmic fraction (in approximately 3.7 ml). The nuclear pellet is resuspended in 1 ml of 65% sucrose in TM (50 mM Tris, pH 7.4, 2.5 mM MgCl2) and banded on a sucrose step gradient (1 ml nuclei in 65% sucrose TM, 1 ml 60% sucrose TM, 1 ml 55% sucrose TM, 50% sucrose TM, 300 μl 25% sucrose TM) for one hour at 37,000xg with an SW60 rotor. The nuclear band is harvested and diluted to 10% sucrose with TM buffer. Nuclei are pelleted at 37,000xg using an SW60 rotor for 15 minutes and the pellet resuspended in 1 ml of TM buffer. Aliquots are size fractionated on denaturing polyacrylamide gels and the intracellular localization determined. By comparison to the migra' ion rate of newly synthesized ribozyme, the various fractions containing intact ribozyme can be determined.
To investigate modifications which would lengthen the half-life of ribozyme molecules intracellularly, the cells may be fractioned as above and the purity of each fraction assessed by assaying enzyme activity known to exist in that fraction. The various cell fractions are frozen at -70°C and used to determine relative nuclease resistances of modified ribozyme molecules. Ribozyme molecules may be synthesized with 5 phosphorothioate (ps) , or 2'-Omethyl (2'-OMe) modifications at each end of the molecule. These molecules and a phosphodiester version of the ribozyme are end-labeled with 32P and ATP using T4 polynucleotide kinase. Equal concentrations are added to the cell cytoplasmic extracts and aliquots of each taken at 10 minute intervals. The samples are size fractionated by denaturing PAGE and relative rates of nuclease resistance analyzed by scanning the gel with an Ambis β-scanner. The results show whether the ribozymes are digested by the cytoplasmic extract, and which versions are relatively more nuclease resistant.
Modified ribozymes generally maintain 80-90% of the catalytic activity of the native ribozyme when short RNA substrates are employed.
Unlabeled, 5' end-labeled or 3' end-labeled ribozymes can be used in the assays. These experiments can also be performed with human cell extracts to verify the observations.
In one example, Vero or HeLa cells were grown to 90-95% confluency in 175 cm2 tissue culture flasks, scraped into 10 ml of cold phosphate buffered saline
(PBS) , then washed once in 10 ml of cold PBS and once in 10 ml of cold TSE (10 mM Tris, pH 7.4; 0.25 M sucrose; 1 mM EDTA) . The cell pellets were resuspended in 4 ml of TSE, dounced 35x on ice, and the released nuclei pelleted by centrifugation at lOOOg for 10 minutes. The nuclear pellet was resuspended in 1 ml of 65% sucrose TM (50 mM Tris, pH 7.4; 2.5 mM MgCl2) and transferred to Beckman ultra-clear tubes. The following sucrose TM solutions were layered on top of the sample: 1 ml 60%, 1 ml 55%, and 25% sucrose to the top of the tube.
Gradients were spun in an SW60 rotor at 37,000g for 1 hour. HeLa nuclei banded at the 55-60% sucrose boundary and Vero nuclei banded at the 50-55% sucrose boundary. Nuclear bands were harvested, diluted to 10% sucrose with TM buffer, and pelleted by centrifugation at 37,000g for 15 minutes using an SW60 rotor. The nuclear pellet was resuspended in 1 ml of TM buffer.
Subcellular organelles and membrane components in the post nuclear supernatant were separated from the cytoplasmic fraction by centrifugation at 200,000g for 2 hours in an SW60 rotor. The pellet contained the membrane fraction, which was resuspended in 1 ml of H buffer (0.25 M sucrose; 50 mM HEPES, pH 7.4), and the supernatant contained the cytoplasmic fraction.
Purity of the various fractions was assessed using enzymatic markers specific for the cytoplasmic and membranous fractions. Three enzyme markers for the membranous fraction were used; hexosaminidase and β- glucocerebrosidase are localized in lysosomes, while alkaline phosphodiesterase is specific to endosomes. Specifically, the assays were as follows: For N-acetyl-beta-hexosaminidase, the reaction mixture contained 0.3 mg/ l 4-methylumbelliferyl-N- acetyl-glucosaminide; 20 mM sodium citrate; pH 4.5; 0.01% Triton X-100; and 100 μl of sample in a final volume of 500 μl (Harding et al. , 64 Cell 393, 1991). The reactions were incubated at 37°C for 1 hour and stopped by the addition of 1.5 ml of stop buffer (0.13 M glycine, 0.07 M NaCl, 0.08 M sodium carbonate, pH 10.6). The reaction product was quantitated in a Hitachi F-4010 fluorescence spectrophotometer by excitation of the fluorophore at 360 nm and analysis of the emission at 448 nm.
For Alkaline Phosphodiesterase, the assay medium contained 25 mM CAPS (3-(Cyclohexylamino) - propanesulfonic acid), pH 10.6; 0.05% Triton X-100; 15 mM MgCl2; 1.25 mg/ml Thymidine-5'-monophosphate-p- nitrophenyl ester; and 100 μl of sample in a total reaction volume of 200 μl. The reactions were incubated at 37°C for 2 hours, then diluted to 1 ml with H20 and the absorbance was measured at 400 nm (Razell and Khorana, 234 J. Biol. Chem. 739, 1959) .
For β-glucocerebrosidase, the reaction contained 85 nM sodium citrate, pH 5.9; 0.12% Triton X- 100; 0.1% sodium taurocholate; 5 M 4-methylumbelliferyl β-D-glucopyranoside; and 125 μl of sample in a total volume of 250 μl (Kennedy and Cooper, 252 Biochem. J. 739, 1988) . The reaction was incubated at 37°C for 1 hour and stopped by the addition of 0.75 ml of stop buffer. Product formation was measured in a fluorescence spectrophotometer by using an excitation wavelength of 360 nm and analysis of the emission at 448 nm. The cytoplasmic enzyme marker, lactate dehydrogenase, was assayed in an assay mixture containing 0.2 M Tris; pH 7.4; 0.22 mM NADH; 1 mM sodium pyruvate; and 50 μl of sample in a final volume of 1.05 ml. Enzyme levels were determined by decreased absorbency at 350 nm resulting from the oxidation of
NADH at room temperature (Silverstein and Boyer, 239 J. Biol. Chem. 3901, 1964) .
Lactate dehydrogenase was found predominantly in the cytoplasmic fractions of both Vero and HeLa cells, while β-glucocerebrosidase and alkaline phosphodiesterase were found almost exclusively in the membranous fractions. The hexosaminidase activity in Vero cell fractions was concentrated in the membranous fraction (70%) with about 20% in the cytoplasmic fraction. The isolation of enzyme markers with the appropriate cellular compartment demonstrated that cytoplasmic, membranous and nuclear fractions can be isolated with minimal intercompartmental contamination using this fractionation scheme. Nuclease Stability of Ribozymes and mRNA
The simplest and most sensitive way to monitor nuclease activity in cell fractions is to use end- labeled oligonucleotides. However, high levels of phosphatase activity in some biological extracts gives ambiguous results in nuclease experiments when 32P-5'- end-labeled oligonucleotides are used as substrates. To determine the phosphatase activity in the extracts, cellular fractions were incubated with cold ribozymes and trace amounts of 5'-end-labeled ribozyme in the presence of 1 mM Mg+2 (or Zn+2 with HeLa cytoplasmic extracts) to optimize digestion. After polyacrylamide gel electrophoresis of samples, digestion of the oligonucleotide was assessed both by staining and by autoradiography.
Specifically, the basic oligonucleotide digestion reaction contained substrate nucleic acid (an RNA oligonucleotide of 36 nucleotides) and cell fraction extract in a total volume of 100 μl. Aliquots (7 μl) were taken after various periods of incubation at 37°C and added to 7 μl of gel loading buffer (95% formamide, 0.1% bromophenol blue, 0.1% xylene cyanol, and 20 mM EDTA) . The samples were separated by electrophoresis on a 7 M urea, 20% polyacrylamide gel. Intact ribozymes were visualized either by staining with Stains-all (United States Biochemical, Cleveland, OH) , or autoradiography of 32P-labeled ribozyme. The stained gels and X-ray films were scanned on a Bio 5000 density scanner (U.S. Biochemical). Ribozymes were 5' end- labeled with T4 polynucleotide kinase (U.S. Biochemical) using 10 μCi of 32P γ-ATP (3,000 Ci/mmole, New England Nuclear, Boston, MA), and 20-25 pmoles of ribozymes. The unincorporated nucleotides were separated from the product by G-50 spin chromatography. Nuclease assays contained 1-2 pmoles of 32P-labeled ribozyme. All oligonucleotides were synthesized on an Applied Biosystems 394 DNA/RNA synthesizer (Applied Biosysterns Inc., Foster City, CA) according to manufacturer's protocols. The nuclear fractions were resuspended in a buffer containing 2.5 mM MgCl2. Experiments involving the nuclear fractions were performed in the presence of 1 mM Mg+2, or in combination with 1 mM Mn+2, Ca+2, or Zn+2.
To measure the stability of mRNA, Vero cells were infected with herpes simplex virus (HSV) at a M.O.I, of 5 and total RNA was extracted (Chomczynski and Sacchi, 162 Anal. Biochem. 156, 1987) . An RNase protection assay was used to detect mRNA after incubation of total infected cellular RNA in cytoplasmic extracts. RNA probes were produced from PCR-amplified template DNA using T7 RNA polymerase (U.S. Biochemical) in the presence of 32P α-CTP (3,000 Ci/mmole, New England Nuclear, Boston, MA) . Template DNA was inactivated with 1 unit of RNase-free DNasel for 15 minutes at 37°C. Unincorporated nucleotides were removed by G-50 spin chromatography. Samples (6 μl) were taken from the nuclease assays after various periods of incubation at 37°C, added to 40 μl of 4 M GUSCN buffer (4 M guanidinium thiocyanate; 25 mM sodium citrate, pH 7; 0.5% sarcosyl; and 0.1 M 2-mercaptoethanol) , and 5 μl of 32P-labeled RNA probe (5xl05 cpm/5 μl, specific activity of 1.8xl06 cpm/μg) in 4 M GUSCN buffer. Hybridization reactions were covered with mineral oil and incubated at 55°C for 12-16 hours, after which the hybridization reaction was mixed with 500 μl of RNase buffer (0.4 M NaCl, 20 μg/ml RNaseA, 2 units/ml Tl RNase) and incubated for 30 minutes at 37°C. RNase activity was quenched by incubation with 10 μl of 20% SDS and 10 μl of proteinase K (20 mg/ml) , and the RNA was extracted using a phenol/chloroform mixture. The protected RNA fragment was purified by precipitation with an equal volume of isopropanol in the presence of 20 μg of carrier yeast tRNA. The RNA pellets were resuspended in gel loading buffer, heated to 95°C for 5 minutes and separated by electrophoresis on a 5% polyacrylamide, 7 M urea gel. Protected fragments were visualized by autoradiography, and the films were scanned with a Bio 5000 density scanner. In experiments using these methods, the rate of digestion of ribozymes in Vero cell extracts was similar, demonstrating the lack of significant phosphatase activity in Vero cellular fractions. Similar results were observed with HeLa cellular fractions. In most extracts, ladders of digested fragments were observed; such ladders would not be expected if digestion was an artifact of phosphatase action. Thus, digestion using 5' end-labeled ribozymes is an accurate assessment of nuclease action in cellular extracts.
In other experiments, labeled ribozymes were incubated in various Vero and HeLa cellular fractions. Incubation of ribozymes in either membranous or nuclear fractions resulted in a linear decrease of intact molecules over time. In contrast, no digestion of ribozymes occurred during a 24 hour incubation in Vero cytoplasmic extracts, and HeLa cytoplasmic extracts exhibited a 20-30 minute delay in the onset of RNA digestion. After this refractory period, the rate of digestion was linear but not as rapid as the rates observed in any of the nuclear or membranous fractions. The effect of four divalent cations (Mg+2, Mn+2, Ca+2, and Zn+2) on the nuclease activity of the cellular fractions was assessed. Vero cytoplasmic extracts were stimulated by the addition of 1 mM Mg+2 or Mn+2, while Ca+2 or Zn+2 had no effect. Nuclease activity in HeLa cytoplasmic extracts was enhanced only by the addition of 1 mM Zn+2. Both Vero and HeLa membranous fractions exhibited maximum nuclease activity with the addition of Mg+2 or Mn+2 ions, while the addition of Ca+2 significantly reduced activity of the HeLa membranous fraction and abolished nuclease activity in the Vero membranous fraction. Addition of Zn+2 to both membranous fractions resulted in a loss of all RNase activity. The Vero nuclear extract demonstrated roughly equivalent nuclease activity in the presence of either Mg+2 alone or a Mg+2 and Mn+2 ion combination, less in the presence of Mg+2 and Ca+2, and no activity in the presence of Mg+2 and Zn+2. The effects of cation addition were not as dramatic with HeLa nuclear extracts. The nuclease activity of these fractions was greatest in the presence of Mg+2 alone or Mg+2 and Ca+2 and decreased slightly with the addition of Mn+2 or Zn+2 to the Mg+2 present in the extracts.
To verify that nuclease activity was dependent upon added divalent cations, nuclease assays were performed using 1 mM Mg+2 in the presence and absence of 20 mM EDTA. For the HeLa cytoplasmic fractions, the Mg+2 was replaced with 1 mM Zn+2. The presence of 20 mM EDTA completely abolished nuclease activity in the Vero and HeLa cytoplasmic fractions and Vero nuclear fractions. Nuclease activity in the HeLa membranous and nuclear fractions was partially inhibited by the addition of EDTA, while EDTA had no effect on the nuclease activity in the Vero membranous fraction. For comparative purposes, reactions using DNA oligonucleotides were performed using different Vero fractions. All DNase activity in Vero cytoplasmic, membranous, and nuclear fractions was inhibited by 20 mM EDTA.
The stability of RNA oligonucleotides and HSV-1 mRNA were compared in the presence and absence of activity-enhancing divalent cations (1 mM Mg+2, Vero cells; 1 mM Zn+2, HeLa cells) . Total cellular RNA from HSV-1 infected Vero cells (8 mg) and tracer amounts of 32P-5'-end-labeled RNA oligonucleotides (1 pmole) were incubated with Vero or HeLa cytoplasmic extracts. In the absence of divalent cations, no substantial decrease of intact ribozymes was detected in assays, although mRNA was digested in both Vero and HeLa cytoplasmic extracts. After addition of divalent cations, digestion of ribozymes occurred in both Vero and HeLa cytoplasmic fractions. The rate of ribozyme digestion in HeLa extracts increased to levels similar to those observed with mRNA, while the rate of mRNA digestion remained greater than the rate of ribozyme digestion in Vero cytoplasmic fractions.
Thus, the stability of hammerhead ribozymes were compared in both Vero and HeLa cell cytoplasmic, membranous and nuclear fractions. Vero cytoplasmic and nuclear fractions were found to require Mg+2 for optimal nuclease activity, while the membranous fraction was not altered by the addition of divalent cations. HeLa membranous and nuclear fractions were also activated by Mg+2, while the cytoplasmic fractions required Zn"2 for nuclease activation. Relative stabilities of ribozymes and mRNAs were compared in Vero and HeLa cytoplasmic fractions. In the absence of appropriate divalent cations, little ribozyme digestion was observed in either cytoplasmic preparation while mRNA was rapidly digested. The addition of Mg+2 to Vero cytoplasmic extracts and Zn+2 to the HeLa cytoplasmic extracts stimulated ribozyme degradation and enhanced mRNA digestion. These data show that the nuclease sensitivity of ribozymes is cell-type specific, varies with the intracellular compartment studied and may not be able to be predicted from studies with mRNA. Notably, however, ribozymes appear stable in such cellular fractions for a period of time potentially sufficient to have a therapeutically useful activity. Administration of Ribozyme
Selected ribozymes can be administered prophylactically, or to patients having disease conditions, e.g. , by exogenous delivery of the ribozyme to a desired tissue by means of an appropriate delivery vehicle, e.g. , a liposome, a controlled release vehicle, by use of iontophoresis, electroporation or ion paired molecules, or covalently attached adducts, and other pharmacologically approved methods of delivery. Routes of administration include intramuscular, aerosol, oral (tablet or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal. Expression vectors for immunization with ribozymes and/or delivery of ribozymes are also suitable.
The specific delivery route of any selected ribozyme will depend on the use of the ribozyme. Generally, a specific delivery program for each ribozyme will focus on unmodified ribozyme uptake with regard to intracellular localization, followed by demonstration of efficacy. Alternatively, delivery to these same cells in an organ or tissue of an animal can be pursued. Uptake studies will include uptake assays to evaluate cellular ribozyme uptake, regardless of the delivery vehicle or strategy. Such assays will also determine the intracellular localization of the ribozyme following uptake, ultimately establishing the requirements for maintenance of steady-state concentrations within the cellular compartment containing the target sequence (nucleus and/or cytoplasm) . Efficacy and cytotoxicity can then be tested. Toxicity will not only include cell viability but also cell function. Some methods of delivery that may be used include: a. encapsulation in liposomes, b. transduction by retroviral vectors, c. conjugation with cholesterol, d. localization to nuclear compartment utilizing nuclear targeting site found on most nuclear proteins, e. neutralization of charge of ribozyme by using nucleotide derivatives, and f. use of blood stem cells to distribute ribozymes throughout the body. At least three types of delivery strategies are useful in the present invention, including: ribozyme modifications, particle carrier drug delivery vehicles, and retroviral expression vectors. Unmodified ribozymes, like most small molecules, are taken up by cells, albeit slowly. To enhance cellular uptake, the ribozyme may be modified essentially at random, in ways which reduce its charge but maintains specific functional groups. This results in a molecule which is able to diffuse across the cell membrane, thus removing the permeability barrier.
Modification of ribozymes to reduce charge is just one approach to enhance the cellular uptake of these larger molecules. The random approach, however, is not advisable since ribozymes are structurally and functionally more complex than small drug molecules. The structural requirements necessary to maintain ribozyme catalytic activity are well understood by those in the art. These requirements are taken into consideration when designing modifications to enhance cellular delivery. The modifications are also designed to reduce susceptibility to nuclease degradation. Both of these characteristics should greatly improve the efficacy of the ribozyme. Cellular uptake can be increased by several orders of magnitude without having to alter the phosphodiester linkages necessary for ribozyme cleavage activity.
Chemical modifications of the phosphate backbone will reduce the negative charge allowing free diffusion across the membrane. This principle has been successfully demonstrated for antisense DNA technology. The similarities in chemical composition between DNA and RNA make this a feasible approach. In the body, maintenance of an external concentration will be necessary to drive the diffusion of the modified ribozyme into the cells of the tissue. Administration routes which allow the diseased tissue to be exposed to a transient high concentration of the drug, which is slowly dissipated by systemic adsorption are preferred. Intravenous administration with a drug carrier designed to increase the circulation half-life of the ribozyme can be used. The size and composition of the drug carrier restricts rapid clearance from the blood stream. The carrier, made to accumulate at the site of infection, can protect the ribozyme from degradative processes.
Drug delivery vehicles are effective for both systemic and topical administration. They can be designed to serve as a slow release reservoir, or to deliver their contents directly to the target cell. An advantage of using direct delivery drug vehicles is that multiple molecules are delivered per uptake. Such vehicles have been shown to increase the circulation half-life of drugs which would otherwise be rapidly cleared from the blood stream. Some examples of such specialized drug delivery vehicles which fall into this category are liposomes, hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres.
From this category of delivery systems, liposomes are preferred. Liposomes increase intracellular stability, increase uptake efficiency and improve biological activity.
Liposomes are hollow spherical vesicles composed of lipids arranged in a similar fashion as those lipids which make up the cell membrane. They have an internal aqueous space for entrapping water soluble compounds and range in size from 0.05 to several microns in diameter. Several studies have shown that liposomes can deliver RNA to cells and that the RNA remains biologically active.
For example, a liposome delivery vehicle originally designed as a research tool, Lipofectin, has been shown to deliver intact mRNA molecules to cells yielding production of the corresponding protein.
Liposomes offer several advantages: They are non-toxic and biodegradable in composition; they display long circulation half-lives; and recognition molecules can be readily attached to their surface for targeting to tissues. Finally, cost effective manufacture of liposome-based pharmaceuticals, either in a liquid suspension or lyophilized product, has demonstrated the viability of this technology as an acceptable drug delivery system. Other controlled release drug delivery systems, such as nonoparticles and hydrogels may be potential delivery vehicles for a ribozyme. These carriers have been developed for chemotherapeutic agents and protein- based pharmaceuticals, and consequently, can be adapted for ribozyme delivery.
Topical administration of ribozymes is advantageous since it allows localized concentration at the site of administration with minimal systemic adsorption. This simplifies the delivery strategy of the ribozyme to the disease site and reduces the extent of toxicological characterization. Furthermore, the amount of material to be applied is far less than that required for other administration routes. Effective delivery requires the ribozyme to diffuse into the infected cells. Chemical modification of the ribozyme to neutralize negative charge may be all that is required for penetration. However, in the event that charge neutralization is insufficient, the modified ribozyme can be co-formulated with permeability enhancers, such as Azone or oleic acid, in a liposome. The liposomes can either represent a slow release presentation vehicle in which the modified ribozyme and permeability enhancer transfer from the liposome into the infected cell, or the liposome phospholipids can participate directly with the modified ribozyme and permeability enhancer in facilitating cellular delivery. In some cases, both the ribozyme and permeability enhancer can be formulated into a suppository formulation for slow release. Ribozymes may also be systemically administered. Systemic absorption refers to the accumulation of drugs in the blood stream followed by distribution throughout the entire body. Administration routes which lead to systemic absorption include: intravenous, subcutaneous, intraperitoneal, intranasal, intrathecal and ophthalmic. Each of these administration routes expose the ribozyme to an accessible diseased tissue. Subcutaneous administration drains into a localized lymph node which proceeds through the lymphatic network into the circulation. The rate of entry into the circulation has been shown to be a function of molecular weight or size. The use of a liposome or other drug carrier localizes the ribozyme at the lymph node. The ribozyme can be modified to diffuse into the cell, or the liposome can directly participate in the delivery of either the unmodified or modified ribozyme to the cell.
A liposome formulation containing phosphatidyl- ethanolomidomethylthiosuccinimide which can deliver oligonucleotides to lymphocytes and macrophages is useful for certain conditions. Furthermore, a 200 nm diameter liposome of this composition was internalized as well as 100 nm diameter liposomes. The 200 nm liposomes exhibit a ten-fold greater packaging capacity than the 100 nm liposomes and can accommodate larger molecules such as a ribozyme expression vector. This ribozyme delivery system prevents mRNA expression in afflicted primary immune cells. Whole blood studies show that the formulation is taken up by 90% of the lymphocytes after 8 hours at 37°C. Preliminary biodistribution and pharmacokinetic studies yielded 70% of the injected dose/gm of tissue in the spleen after one hour following intravenous administration. Liposomes injected intravenously show accumulation in the liver, lung and spleen. The composition and size can be adjusted so that this accumulation represents 30% to 40% of the injected dose. The remaining dose circulates in the blood stream for up to 24 hours. The chosen method of delivery should result in cytoplasmic accumulation and molecules should have some nuclease-resistance for optimal dosing. Nuclear delivery may be used but is less preferable. Most preferred delivery methods include liposomes (10-400 nm) , hydrogels, controlled-release polymers, microinjection or electroporation (for ex vivo treatments) and other pharmaceutically applicable vehicles. The dosage will depend upon the disease indication and the route of administration but should be between 100-200 mg/kg of body weight/day. The duration of treatment will extend through the course of the disease symptoms, possibly continuously. The number of doses will depend upon disease delivery vehicle and efficacy data from clinical trials.
Establishment of therapeutic levels of ribozyme within the cell is dependent upon the rate of uptake and degradation. Decreasing the degree of degradation will prolong the intracellular half-life of the ribozyme. Thus, chemically modified ribozymes, e.g. , with modification of the phosphate backbone, or capping of the 5' and 3' ends of the ribozyme with nucleotide analogs may require different dosaging. Descriptions of useful systems are provided in the art cited above, all of which is hereby incorporated by reference herein. Other embodiments are within the following claims.

Claims

Claims
1. An enzymatic RNA molecule which cleaves mRNA associated with development or maintenance of an inflammatory, arthritic, stenotic or cardiovascular disease or condition.
2. The enzymatic RNA molecule of claim 1, which cleaves mRNA produced from: a) the genes encoding tumor necrosis factor-α, interleukin-5, -1, -3, -4, -6, -8, ICAM-1, ELAM-1, VCAM-1, TGF-α, TNFOR, IL-1R, α-, β- or γ-interferon, EoCSF, GM-CSF, glycerol transferase, the selectins, E-selectin, MEL-14, GMP-140, MAM and protein kinase C; b) from a gene encoding a matrix metalloproteinase; c) a gene selected from one encoding c-myb, TGF-β, NF-KB, PDGF, bFGF, CGRP, angiotensin II, and endothelium-derived relaxing factor; and d) the gene encoding ACE or ECE.
3. The enzymatic RNA molecule of claim 1, which cleaves target mRNA having a sequence selected from SEQ. ID. NOS. 1-54 in table 1; SEQ. ID. NOS 1-33 in table 2; SEQ. ID. NOS. 1-77 in table 3; and SEQ. ID. NOS. 1-81 in table 4.
4. The enzymatic RNA molecule of claims 1, 2 or 3, wherein said RNA molecule is in a hammerhead motif.
5. The enzymatic RNA molecule of claim 4, wherein said RNA molecule is in a hairpin, hepatitis Delta virus, group 1 intron, or RNaseP RNA motif.
6. The enzymatic RNA molecule of claim 4, wherein said ribozyme comprises between 5 and 23 bases complementary to said mRNA.
7. The enzymatic RNA molecule of claim 6, wherein said ribozyme comprises between 10 and 18 bases complementary to said mRNA.
8. A mammalian cell including an enzymatic RNA molecule of claims 1, 2 or 3.
9. The cell of claim 8, wherein said cell is a human cell.
10. An expression vector including nucleic acid encoding the enzymatic RNA molecule of claims 1, 2 or 3, in a manner which allows expression of that enzymatic RNA molecule within a mammalian cell.
11. A method for treatment of an inflammatory disease, an arthritic condition, a stenotic condition, or a cardiovascular condition by administering to a patient an enzymatic RNA molecule of claims 1, 2 or 3.
.12. The method of claim 11, wherein said patient is a human.
EP93918144A 1992-07-17 1993-07-02 Method and reagent for treatment of animal diseases. Pending EP0654077A4 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP02005642A EP1251170A3 (en) 1992-07-17 1993-07-02 Method and reagent for treatment of NF-kappaB dependent animal diseases
EP97101534A EP0786522A2 (en) 1992-07-17 1993-07-02 Enzymatic RNA molecules for treatment of stenotic conditions

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US91676392A 1992-07-17 1992-07-17
US916763 1992-07-17
US98984892A 1992-12-07 1992-12-07
US98984992A 1992-12-07 1992-12-07
US98713292A 1992-12-07 1992-12-07
US987132 1992-12-07
US989849 1992-12-07
US889593A 1993-01-19 1993-01-19
US8895 1993-01-19
PCT/US1993/006316 WO1994002595A1 (en) 1992-07-17 1993-07-02 Method and reagent for treatment of animal diseases
US989848 1997-12-12

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP02005642A Division EP1251170A3 (en) 1992-07-17 1993-07-02 Method and reagent for treatment of NF-kappaB dependent animal diseases
EP97101534.2 Division-Into 1997-01-31

Publications (2)

Publication Number Publication Date
EP0654077A1 true EP0654077A1 (en) 1995-05-24
EP0654077A4 EP0654077A4 (en) 1996-03-13

Family

ID=27533301

Family Applications (3)

Application Number Title Priority Date Filing Date
EP02005642A Withdrawn EP1251170A3 (en) 1992-07-17 1993-07-02 Method and reagent for treatment of NF-kappaB dependent animal diseases
EP93918144A Pending EP0654077A4 (en) 1992-07-17 1993-07-02 Method and reagent for treatment of animal diseases.
EP97101534A Withdrawn EP0786522A2 (en) 1992-07-17 1993-07-02 Enzymatic RNA molecules for treatment of stenotic conditions

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP02005642A Withdrawn EP1251170A3 (en) 1992-07-17 1993-07-02 Method and reagent for treatment of NF-kappaB dependent animal diseases

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP97101534A Withdrawn EP0786522A2 (en) 1992-07-17 1993-07-02 Enzymatic RNA molecules for treatment of stenotic conditions

Country Status (6)

Country Link
EP (3) EP1251170A3 (en)
JP (1) JPH07509133A (en)
AU (2) AU4769893A (en)
CA (1) CA2140343A1 (en)
MX (1) MX9304329A (en)
WO (1) WO1994002595A1 (en)

Families Citing this family (505)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6469158B1 (en) 1992-05-14 2002-10-22 Ribozyme Pharmaceuticals, Incorporated Synthesis, deprotection, analysis and purification of RNA and ribozymes
US5686599A (en) * 1992-05-14 1997-11-11 Ribozyme Pharmaceuticals, Inc. Synthesis, deprotection, analysis and purification of RNA and ribozymes
US5977343A (en) 1992-05-14 1999-11-02 Ribozyme Pharmaceuticals, Inc. Synthesis, deprotection, analysis and purification of RNA and ribozymes
US5804683A (en) * 1992-05-14 1998-09-08 Ribozyme Pharmaceuticals, Inc. Deprotection of RNA with alkylamine
US5646042A (en) * 1992-08-26 1997-07-08 Ribozyme Pharmaceuticals, Inc. C-myb targeted ribozymes
US5891684A (en) * 1992-10-15 1999-04-06 Ribozyme Pharmaceuticals, Inc. Base-modified enzymatic nucleic acid
US5658780A (en) * 1992-12-07 1997-08-19 Ribozyme Pharmaceuticals, Inc. Rel a targeted ribozymes
US5616488A (en) * 1992-12-07 1997-04-01 Ribozyme Pharmaceuticals, Inc. IL-5 targeted ribozymes
US5612215A (en) * 1992-12-07 1997-03-18 Ribozyme Pharmaceuticals, Inc. Stromelysin targeted ribozymes
US5837542A (en) * 1992-12-07 1998-11-17 Ribozyme Pharmaceuticals, Inc. Intercellular adhesion molecule-1 (ICAM-1) ribozymes
US5811300A (en) * 1992-12-07 1998-09-22 Ribozyme Pharmaceuticals, Inc. TNF-α ribozymes
DK0748382T3 (en) 1993-09-02 2003-02-17 Ribozyme Pharm Inc Enzymatic nucleic acid containing non-nucleotide
AU8088694A (en) * 1993-10-27 1995-05-22 Ribozyme Pharmaceuticals, Inc. 2'-amido and 2'-peptido modified oligonucleotides
CA2176035A1 (en) 1993-11-08 1995-05-18 Nassim Usman Base-modified enzymatic nucleic acid
ATE226254T1 (en) * 1993-11-12 2002-11-15 Ribozyme Pharm Inc REAGENT FOR THE TREATMENT OF ARTHRITIC CONDITIONS
US5693532A (en) * 1994-11-04 1997-12-02 Ribozyme Pharmaceuticals, Inc. Respiratory syncytial virus ribozymes
US5639647A (en) * 1994-03-29 1997-06-17 Ribozyme Pharmaceuticals, Inc. 2'-deoxy-2'alkylnucleotide containing nucleic acid
CA2183992A1 (en) * 1994-02-23 1995-08-31 Dan T. Stinchcomb Method and reagent for inhibiting the expression of disease related genes
US5902880A (en) * 1994-08-19 1999-05-11 Ribozyme Pharmaceuticals, Inc. RNA polymerase III-based expression of therapeutic RNAs
US5631359A (en) * 1994-10-11 1997-05-20 Ribozyme Pharmaceuticals, Inc. Hairpin ribozymes
US5834440A (en) * 1994-03-07 1998-11-10 Immusol Incorporated Ribozyme therapy for the inhibition of restenosis
US5627053A (en) * 1994-03-29 1997-05-06 Ribozyme Pharmaceuticals, Inc. 2'deoxy-2'-alkylnucleotide containing nucleic acid
US6103890A (en) * 1994-05-18 2000-08-15 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids that cleave C-fos
US20030026782A1 (en) * 1995-02-07 2003-02-06 Arthur M. Krieg Immunomodulatory oligonucleotides
US6350934B1 (en) 1994-09-02 2002-02-26 Ribozyme Pharmaceuticals, Inc. Nucleic acid encoding delta-9 desaturase
US5599706A (en) * 1994-09-23 1997-02-04 Stinchcomb; Dan T. Ribozymes targeted to apo(a) mRNA
US5716824A (en) * 1995-04-20 1998-02-10 Ribozyme Pharmaceuticals, Inc. 2'-O-alkylthioalkyl and 2-C-alkylthioalkyl-containing enzymatic nucleic acids (ribozymes)
CA2207593A1 (en) * 1994-12-13 1996-06-20 John Gustofson Method and reagent for treatment of arthritic conditions, induction of graft tolerance and reversal of immune responses
US5672501A (en) * 1994-12-23 1997-09-30 Ribozyme Pharmaceuticals, Inc. Base-modified enzymatic nucleic acid
US5705388A (en) * 1994-12-23 1998-01-06 Ribozyme Pharmaceuticals, Inc. CETP Ribozymes
US5663064A (en) * 1995-01-13 1997-09-02 University Of Vermont Ribozymes with RNA protein binding site
DE69633272T2 (en) 1995-02-08 2005-09-08 Takara Bio Inc., Otsu Methods for cancer treatment and control
US5877021A (en) 1995-07-07 1999-03-02 Ribozyme Pharmaceuticals, Inc. B7-1 targeted ribozymes
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
US7034009B2 (en) 1995-10-26 2006-04-25 Sirna Therapeutics, Inc. Enzymatic nucleic acid-mediated treatment of ocular diseases or conditions related to levels of vascular endothelial growth factor receptor (VEGF-R)
US5998203A (en) 1996-04-16 1999-12-07 Ribozyme Pharmaceuticals, Inc. Enzymatic nucleic acids containing 5'-and/or 3'-cap structures
WO1997032018A2 (en) * 1996-02-29 1997-09-04 Immusol, Inc. Hepatitis c virus ribozymes
JP2000509969A (en) * 1996-04-02 2000-08-08 コモンウェルス・サイエンティフィック・アンド・インダストリアル・リサーチ・オーガニゼーション Asymmetric hammerhead ribozyme
ATE449851T1 (en) 1996-10-01 2009-12-15 Geron Corp HUMAN TELOMERASE REVERSE TRANSCRIPTASE PROMOTER
US5807743A (en) * 1996-12-03 1998-09-15 Ribozyme Pharmaceuticals, Inc. Interleukin-2 receptor gamma-chain ribozymes
US7008776B1 (en) 1996-12-06 2006-03-07 Aventis Pharmaceuticals Inc. Compositions and methods for effecting the levels of high density lipoprotein (HDL) cholesterol and apolipoprotein AI very low density lipoprotein (VLDL) cholesterol and low density lipoprotein (LDL) cholesterol
US6248878B1 (en) 1996-12-24 2001-06-19 Ribozyme Pharmaceuticals, Inc. Nucleoside analogs
US6057156A (en) 1997-01-31 2000-05-02 Robozyme Pharmaceuticals, Inc. Enzymatic nucleic acid treatment of diseases or conditions related to levels of epidermal growth factor receptors
US6159951A (en) * 1997-02-13 2000-12-12 Ribozyme Pharmaceuticals Inc. 2'-O-amino-containing nucleoside analogs and polynucleotides
AU7071598A (en) * 1997-04-10 1998-10-30 Erasmus University Rotterdam Diagnosis method and reagents
US5942395A (en) * 1997-05-09 1999-08-24 Universite De Montreal Hybrid ribozymes and methods of use
US6548657B1 (en) 1997-06-09 2003-04-15 Ribozyme Pharmaceuticals, Inc. Method for screening nucleic acid catalysts
US6280936B1 (en) 1997-06-09 2001-08-28 Ribozyme Pharmaceuticals, Inc. Method for screening nucleic acid catalysts
US6183959B1 (en) 1997-07-03 2001-02-06 Ribozyme Pharmaceuticals, Inc. Method for target site selection and discovery
US6316612B1 (en) 1997-08-22 2001-11-13 Ribozyme Pharmaceuticals, Inc. Xylofuranosly-containing nucleoside phosphoramidites and polynucleotides
US6656731B1 (en) 1997-09-22 2003-12-02 Max Planck Gesellschaft Zur Forderung Der Wissenschaften E.V. Nucleic acid catalysts with endonuclease activity
CA2304813A1 (en) * 1997-09-22 1999-04-08 Fritz Eckstein Nucleic acid catalysts with endonuclease activity
AU9509798A (en) 1997-09-25 1999-04-12 University Of Florida Antisense oligonucleotide compositions targeted to angiotensi n converting enzy me mrna and methods of use
US6127535A (en) 1997-11-05 2000-10-03 Ribozyme Pharmaceuticals, Inc. Nucleoside triphosphates and their incorporation into oligonucleotides
US6617438B1 (en) 1997-11-05 2003-09-09 Sirna Therapeutics, Inc. Oligoribonucleotides with enzymatic activity
BR9814294B1 (en) 1997-12-18 2011-10-18 b cry3bb polypeptide, modified thuringiensis, composition comprising the same, process for producing said composition, polynucleotide, vector, virus, transgenic microorganism and methods for killing and controlling a coleopteran insect population and for preparing a transgenic plant resistant to beetles and a Coleoptera-resistant plant seed and a Coleoptera-resistant plant seed.
US20020147143A1 (en) 1998-03-18 2002-10-10 Corixa Corporation Compositions and methods for the therapy and diagnosis of lung cancer
US6200754B1 (en) 1998-03-19 2001-03-13 Variagenics, Inc. Inhibitors of alternative alleles of genes encoding products that mediate cell response to environmental changes
EP1071753A2 (en) 1998-04-20 2001-01-31 Ribozyme Pharmaceuticals, Inc. Nucleic acid molecules with novel chemical compositions capable of modulating gene expression
CA2230203A1 (en) 1998-04-29 1999-10-29 Universite De Sherbrooke Delta ribozyme for rna cleavage
US6228642B1 (en) * 1998-10-05 2001-05-08 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of tumor necrosis factor-(α) (TNF-α) expression
US6080580A (en) * 1998-10-05 2000-06-27 Isis Pharmaceuticals Inc. Antisense oligonucleotide modulation of tumor necrosis factor-α (TNF-α) expression
US6995259B1 (en) 1998-10-23 2006-02-07 Sirna Therapeutics, Inc. Method for the chemical synthesis of oligonucleotides
AU4807400A (en) 1999-04-30 2000-11-17 University Of Florida Adeno-associated virus-delivered ribozyme compositions and methods of use
US6831171B2 (en) 2000-02-08 2004-12-14 Yale University Nucleic acid catalysts with endonuclease activity
US20070026394A1 (en) 2000-02-11 2007-02-01 Lawrence Blatt Modulation of gene expression associated with inflammation proliferation and neurite outgrowth using nucleic acid based technologies
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)
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
US20040002068A1 (en) 2000-03-01 2004-01-01 Corixa Corporation Compositions and methods for the detection, diagnosis and therapy of hematological malignancies
US7153839B2 (en) 2000-03-24 2006-12-26 Biocell Laboratories Method of protecting erythricytes, in particular for improvement of blood cytopenia
US6737271B1 (en) * 2000-03-24 2004-05-18 Biocell Laboratories Compound, composition and method for the treatment of inflammatory and inflammatory-related disorders
GB0018307D0 (en) 2000-07-26 2000-09-13 Aventis Pharm Prod Inc Polypeptides
CA2404688C (en) 2000-03-31 2012-07-31 Aventis Pharmaceuticals Products Inc. Nuclear factor .kappa.b inducing factor
JP2004513615A (en) 2000-06-28 2004-05-13 コリクサ コーポレイション Compositions and methods for treatment and diagnosis of lung cancer
US7754435B2 (en) 2000-09-21 2010-07-13 Philadelphia Health And Education Corporation Prostate cancer-related compositions, methods, and kits based on DNA macroarray proteomics platforms
CA2441071C (en) 2001-03-14 2010-06-22 Myriad Genetics, Inc. Tsg101-gag interaction and use thereof
JP2005504513A (en) 2001-05-09 2005-02-17 コリクサ コーポレイション Compositions and methods for treatment and diagnosis of prostate cancer
US20050148530A1 (en) 2002-02-20 2005-07-07 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)
US9994853B2 (en) 2001-05-18 2018-06-12 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
US7205399B1 (en) 2001-07-06 2007-04-17 Sirna Therapeutics, Inc. Methods and reagents for oligonucleotide synthesis
ES2405790T3 (en) 2001-12-17 2013-06-03 Corixa Corporation Compositions and methods for therapy and diagnosis of inflammatory bowel disease
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)
AU2003207708A1 (en) 2002-02-20 2003-09-09 Sirna Therapeutics, Inc. Rna interference mediated inhibition of map kinase genes
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)
US7956176B2 (en) 2002-09-05 2011-06-07 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US20080260744A1 (en) 2002-09-09 2008-10-23 Omeros Corporation G protein coupled receptors and uses thereof
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
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
US7994149B2 (en) 2003-02-03 2011-08-09 Medtronic, Inc. Method for treatment of Huntington's disease through intracranial delivery of sirna
ES2394799T3 (en) 2003-12-31 2013-02-05 The Penn State Research Foundation Methods to predict and overcome chemotherapy resistance in ovarian cancer
EP1735009A4 (en) 2004-03-12 2011-03-30 Alnylam Pharmaceuticals Inc iRNA AGENTS TARGETING VEGF
CA2605574A1 (en) 2004-04-20 2005-11-03 Galapagos N.V. Methods, compositions and compound assays for inhibiting amyloid-beta protein production
EP2259063B1 (en) 2004-04-27 2012-05-23 Galapagos N.V. Compound screening assays for inducing differentiation of undifferentiated mammalian cells into osteoblasts
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
US10508277B2 (en) 2004-05-24 2019-12-17 Sirna Therapeutics, Inc. Chemically modified multifunctional short interfering nucleic acid molecules that mediate RNA interference
DE602005019156D1 (en) 2004-05-26 2010-03-18 Anima Cell Metrology METHOD OF EVALUATING RIBONUCLEOTIDE SEQUENCES
EP2270160A1 (en) 2004-06-14 2011-01-05 Galapagos N.V. Methods for identification, and compounds useful for the treatment of degenerative and inflammatory diseases
JP2008503712A (en) 2004-06-21 2008-02-07 ガラパゴス・ナムローゼ・フェンノートシャップ Methods and means for treatment of osteoarthritis
EP1758999B1 (en) 2004-06-22 2011-03-09 The Board Of Trustees Of The University Of Illinois METHODS OF INHIBITING TUMOR CELL PROLIFERATION WITH FOXM1 siRNA
USRE48960E1 (en) 2004-06-28 2022-03-08 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
SI2206781T1 (en) 2004-06-28 2016-05-31 The University Of Western Australia Antisense oligonucleotides for inducing exon skipping and methods of use thereof
WO2006020049A2 (en) 2004-07-15 2006-02-23 Amr Technology, Inc. Aryl-and heteroaryl-substituted tetrahydroisoquinolines and use thereof to block reuptake of norepinephrine, dopamine, and serotonin
US7485468B2 (en) 2004-10-15 2009-02-03 Galapagos Bv Molecular targets and compounds, and methods to identify the same, useful in the treatment of joint degenerative and inflammatory diseases
PL1866414T3 (en) 2005-03-31 2012-10-31 Calando Pharmaceuticals Inc Inhibitors of ribonucleotide reductase subunit 2 and uses thereof
ES2382814T3 (en) 2005-05-17 2012-06-13 Merck Sharp & Dohme Ltd. Cis-4 - [(4-chlorophenyl) sulfonyl] -4- (2,5-difluorophenyl) cyclohexanopropanoic acid for cancer treatment
AU2006261732B2 (en) 2005-06-27 2011-09-15 Alnylam Pharmaceuticals, Inc. RNAi modulation of HIF-1 and theraputic uses thereof
US9133517B2 (en) 2005-06-28 2015-09-15 Medtronics, Inc. Methods and sequences to preferentially suppress expression of mutated huntingtin
US7956050B2 (en) 2005-07-15 2011-06-07 Albany Molecular Research, Inc. Aryl- and heteroaryl-substituted tetrahydrobenzazepines and use thereof to block reuptake of norepinephrine, dopamine, and serotonin
US20070213292A1 (en) 2005-08-10 2007-09-13 The Rockefeller University Chemically modified oligonucleotides for use in modulating micro RNA and uses thereof
JP4879988B2 (en) 2005-09-29 2012-02-22 メルク・シャープ・エンド・ドーム・コーポレイション Acylated spiropiperidine derivatives as melanocortin-4 receptor modulators
EP3936621A1 (en) 2006-01-20 2022-01-12 Cell Signaling Technology, Inc. Translocation and mutant ros kinase in human non-small cell lung carcinoma
US20120208824A1 (en) 2006-01-20 2012-08-16 Cell Signaling Technology, Inc. ROS Kinase in Lung Cancer
GB0603041D0 (en) 2006-02-15 2006-03-29 Angeletti P Ist Richerche Bio Therapeutic compounds
US7846908B2 (en) 2006-03-16 2010-12-07 Alnylam Pharmaceuticals, Inc. RNAi modulation of TGF-beta and therapeutic uses thereof
KR101547579B1 (en) 2006-03-31 2015-08-27 알닐람 파마슈티칼스 인코포레이티드 DsRNA for inhibiting expression of Eg5 gene
US7741469B2 (en) 2006-04-05 2010-06-22 University Of Iowa Research Foundation Compositions for treating hearing loss and methods of use thereof
EP2010226B1 (en) 2006-04-07 2014-01-15 The Research Foundation of State University of New York Transcobalamin receptor polypeptides, nucleic acids, and modulators thereof, and related methods of use in modulating cell growth and treating cancer and cobalamin deficiency
EP3266867A1 (en) 2006-04-14 2018-01-10 Cell Signaling Technology, Inc. Gene defects and mutant alk kinase in human solid tumors
US8377448B2 (en) 2006-05-15 2013-02-19 The Board Of Trustees Of The Leland Standford Junior University CD47 related compositions and methods for treating immunological diseases and disorders
US9273356B2 (en) 2006-05-24 2016-03-01 Medtronic, Inc. Methods and kits for linking polymorphic sequences to expanded repeat mutations
US8598333B2 (en) 2006-05-26 2013-12-03 Alnylam Pharmaceuticals, Inc. SiRNA silencing of genes expressed in cancer
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
RU2553561C2 (en) 2006-07-21 2015-06-20 Сайленс Терапьютикс Аг Means for inhibiting proteinkinase 3 expression
EP1900749A1 (en) 2006-09-12 2008-03-19 Institut National De La Sante Et De La Recherche Medicale (Inserm) Nucleic acids for expressing a polynucleotide of interest in mammalian cancer cells
CA2770486C (en) 2006-09-22 2014-07-15 Merck Sharp & Dohme Corp. Use of platencin and platensimycin as fatty acid synthesis inhibitors to treat obesity, diabetes and cancer
EP2069501A2 (en) 2006-10-02 2009-06-17 Aprea Ab Rna interference against wrap53 to treat hyperproliferative diseases
EP2099467B1 (en) 2006-10-03 2017-05-10 University Of Medicine And Dentistry Of New Jersey Atap peptides, nucleic acids encoding the same and associated methods of use
US20110218176A1 (en) 2006-11-01 2011-09-08 Barbara Brooke Jennings-Spring Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
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
US8481506B2 (en) 2006-12-05 2013-07-09 Rosetta Genomics, Ltd. Nucleic acids involved in viral infection
PL2109608T3 (en) 2007-01-10 2011-08-31 Msd Italia Srl Amide substituted indazoles as poly(adp-ribose)polymerase (parp) inhibitors
WO2008092099A2 (en) 2007-01-26 2008-07-31 Rosetta Genomics, Ltd. Compositions and methods for treating hematopoietic malignancies
US9006206B2 (en) 2007-02-27 2015-04-14 Rosetta Genomics Ltd. Composition and methods for modulating cell proliferation and cell death
CA2699418A1 (en) 2007-02-27 2008-09-04 Moshe Oren Composition and methods for modulating cell proliferation and cell death
WO2008106551A2 (en) 2007-02-28 2008-09-04 The Govt. Of The U.S.A. As Represented By The Secretary Of The Dept. Of Health & Human Serv. Brachyury polypeptides and methods for use
EP2468864A1 (en) 2007-03-02 2012-06-27 Marina Biotech, Inc. Nucleic acid compounds for inhibiting VEGF family gene expression and uses thereof
AU2008233662B2 (en) 2007-04-02 2012-08-23 Msd K.K. Indoledione derivative
AU2008239594B2 (en) 2007-04-13 2013-10-24 Beth Israel Deaconess Medical Center Methods for treating cancer resistant to ErbB therapeutics
LT2494993T (en) 2007-05-04 2018-12-27 Marina Biotech, Inc. Amino acid lipids and uses thereof
CA2690685A1 (en) 2007-06-20 2008-12-24 Galapagos N.V. Molecular targets and compounds, and methods to identify the same, useful in the treatment of bone and joint degenerative diseases
JP5501227B2 (en) 2007-06-27 2014-05-21 メルク・シャープ・アンド・ドーム・コーポレーション 4-Carboxybenzylamino derivatives as histone deacetylase inhibitors
EP2188383B1 (en) 2007-10-09 2017-01-18 Anima Cell Metrology, Inc. Systems and methods for measuring translation activity in viable cells
CA2702686C (en) 2007-10-18 2023-04-04 Cell Signaling Technology, Inc. Translocation and mutant ros kinase in human non-small cell lung carcinoma
US9133522B2 (en) 2007-10-31 2015-09-15 Rosetta Genomics Ltd. Compositions and methods for the diagnosis and prognosis of mesothelioma
JP2011514881A (en) 2007-11-09 2011-05-12 ザ ソルク インスティテュート フォー バイオロジカル スタディーズ Use of TAM receptor inhibitors as immunopotentiators and use of TAM activators as immunosuppressants
CA2910760C (en) 2007-12-04 2019-07-09 Muthiah Manoharan Targeting lipids
EP2224912B1 (en) 2008-01-02 2016-05-11 TEKMIRA Pharmaceuticals Corporation Improved compositions and methods for the delivery of nucleic acids
JP2011515343A (en) 2008-03-03 2011-05-19 タイガー ファーマテック Tyrosine kinase inhibitor
CA2720473A1 (en) 2008-04-04 2009-10-08 Calando Pharmaceuticals, Inc. Compositions and use of epas1 inhibitors
CA2721183C (en) 2008-04-11 2019-07-16 Alnylam Pharmaceuticals, Inc. Site-specific delivery of nucleic acids by combining targeting ligands with endosomolytic components
NZ588583A (en) 2008-04-15 2012-08-31 Protiva Biotherapeutics Inc Novel lipid formulations for nucleic acid delivery
WO2009134443A2 (en) 2008-05-02 2009-11-05 The Brigham And Women's Hospital, Inc. Rna-induced translational silencing and cellular apoptosis
AU2009246134B2 (en) 2008-05-16 2016-03-03 The Children's Hospital Of Philadelphia Genetic alterations on chromosomes 21q, 6q and 15q and methods of use thereof for the diagnosis and treatment of type I diabetes
ATE550024T1 (en) 2008-05-30 2012-04-15 Univ Yale TARGETED OLIGONUCLEOTIDE COMPOSITIONS FOR MODIFYING GENE EXPRESSION
WO2009149182A1 (en) 2008-06-04 2009-12-10 The Board Of Regents Of The University Of Texas System Modulation of gene expression through endogenous small rna targeting of gene promoters
US9156812B2 (en) 2008-06-04 2015-10-13 Bristol-Myers Squibb Company Crystalline form of 6-[(4S)-2-methyl-4-(2-naphthyl)-1,2,3,4-tetrahydroisoquinolin-7-yl]pyridazin-3-amine
CA2736425A1 (en) 2008-09-11 2010-03-18 Galapagos Nv Method for identifying compounds useful for increasing the functional activity and cell surface expression of cf-associated mutant cystic fibrosis transmembrane conductance regulator
JP2012513953A (en) 2008-09-23 2012-06-21 アルニラム ファーマスーティカルズ インコーポレイテッド Chemical modification of monomers and oligonucleotides using cycloaddition
WO2010042877A1 (en) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Improved amino lipids and methods for the delivery of nucleic acids
AU2009305639B2 (en) 2008-10-16 2016-06-23 Marina Biotech, Inc. Processes and compositions for liposomal and efficient delivery of gene silencing therapeutics
WO2010048586A1 (en) 2008-10-24 2010-04-29 Avi Biopharma, Inc. Multiple exon skipping compositions for dmd
IT1391866B1 (en) * 2008-10-30 2012-01-27 Mastelli S R L INJECTIONABLE COMPOSITION OF POLYNUCLEOTIDS FOR THE TREATMENT OF OSTEOARTICULAR DISEASES.
KR20220150995A (en) 2008-11-10 2022-11-11 알닐람 파마슈티칼스 인코포레이티드 Novel lipids and compositions for the delivery of therapeutics
CA2751342C (en) 2009-01-29 2019-05-07 Alnylam Pharmaceuticals, Inc. Lipid formulations comprising cationic lipid and a targeting lipid comprising n-acetyl galactosamine for delivery of nucleic acid
PT2881402T (en) 2009-02-12 2017-08-23 Cell Signaling Technology Inc Mutant ros expression in human liver cancer
WO2010091878A2 (en) 2009-02-13 2010-08-19 Silence Therapeutics Ag Means for inhibiting the expression of opa1
US8829179B2 (en) 2009-02-18 2014-09-09 Silence Therapeutics Gmbh Means for inhibiting the expression of ANG2
EP2398481A2 (en) 2009-02-19 2011-12-28 Galapagos N.V. Methods for identifying and compounds useful for the diagnosis and treatment of diseases involving inflammation
EP2398479A2 (en) 2009-02-19 2011-12-28 Galapagos N.V. Methods for identifying and compounds useful for the diagnosis and treatment of diseases involving inflammation
WO2010094732A1 (en) 2009-02-19 2010-08-26 Biofocus Dpi B.V. Methods for identifying and compounds useful for the diagnosis and treatment of diseases involving inflammation
US8975389B2 (en) 2009-03-02 2015-03-10 Alnylam Pharmaceuticals, Inc. Nucleic acid chemical modifications
CA2754961C (en) 2009-03-11 2018-04-10 Promedior, Inc. Treatment and diagnostic methods for hypersensitive disorders
AU2010224172B2 (en) 2009-03-11 2016-01-21 Promedior, Inc. Treatment methods for autoimmune disorders
EP2408458A1 (en) 2009-03-19 2012-01-25 Merck Sharp&Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF SIGNAL TRANSDUCER AND ACTIVATOR OF TRANSCRIPTION 6 (STAT6) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
MX2011009724A (en) 2009-03-19 2011-10-14 Merck Sharp & Dohme RNA INTERFERENCE MEDIATED INHIBITION OF BTB AND CNC HOMOLOGY 1, BASIC LEUCINE ZIPPER TRANSCRIPTION FACTOR 1 (BACH 1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA) SEQUENCE LISTING.
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)
EP2408915A2 (en) 2009-03-19 2012-01-25 Merck Sharp&Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF GATA BINDING PROTEIN 3 (GATA3) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2010111471A2 (en) 2009-03-27 2010-09-30 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF SIGNAL TRANSDUCER AND ACTIVATOR OF TRANSCRIPTION 1 (STAT1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US20120004281A1 (en) 2009-03-27 2012-01-05 Merck Sharp & Dohme Corp RNA Interference Mediated Inhibition of the Nerve Growth Factor Beta Chain (NGFB) Gene Expression Using Short Interfering Nucleic Acid (siNA)
JP2012521765A (en) 2009-03-27 2012-09-20 メルク・シャープ・エンド・ドーム・コーポレイション RNA interference-mediated inhibition of intracellular adhesion molecule 1 (ICAM-1) gene expression using small interfering nucleic acids (siNA)
WO2010111464A1 (en) 2009-03-27 2010-09-30 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF APOPTOSIS SIGNAL-REGULATING KINASE 1 (ASK1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP2411520A2 (en) 2009-03-27 2012-02-01 Merck Sharp&Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF THE THYMIC STROMAL LYMPHOPOIETIN (TSLP) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2010112569A1 (en) 2009-03-31 2010-10-07 Robert Zimmermann Modulation of adipose triglyceride lipase for prevention and treatment of cachexia, loss of weight and muscle atrophy and methods of screening therefor
WO2010115825A2 (en) 2009-03-31 2010-10-14 Robert Zimmermann Modulation of adipose triglyceride lipase for prevention and treatment of cachexia, loss of weight and muscle atrophy and methods of screening therefor
EP3043179B1 (en) 2009-04-01 2020-05-06 Galapagos N.V. Methods and means for treatment of osteoarthritis
WO2010114780A1 (en) 2009-04-01 2010-10-07 Merck Sharp & Dohme Corp. Inhibitors of akt activity
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
AU2010245933B2 (en) 2009-05-05 2016-06-16 Arbutus Biopharma Corporation Methods of delivering oligonucleotides to immune cells
SG10201402054UA (en) 2009-05-05 2014-09-26 Muthiah Manoharan Lipid compositions
DK2429296T3 (en) 2009-05-12 2018-03-12 Albany Molecular Res Inc 7 - ([1,2,4,] TRIAZOLO [1,5, -A] PYRIDIN-6-YL) -4- (3,4-DICHLORPHENYL) -1,2,3,4- TETRAHYDROISOQUINOLINE AND USE thereof
KR20120034644A (en) 2009-05-12 2012-04-12 알바니 몰레큘라 리써치, 인크. Aryl, heteroaryl, and heterocycle substituted tetrahydroisoquinolines and use thereof
JP5739415B2 (en) 2009-05-12 2015-06-24 ブリストル−マイヤーズ スクウィブ カンパニー (S) -7-([1,2,4] triazolo [1,5-a] pyridin-6-yl) -4- (3,4-dichlorophenyl) -1,2,3,4-tetrahydroisoquinoline Crystal forms and uses thereof
UA110323C2 (en) 2009-06-04 2015-12-25 Promedior Inc Derivative of serum amyloid p and their receipt and application
CN104873464B (en) 2009-06-10 2018-06-22 阿布特斯生物制药公司 Improved lipid formulations
US8283333B2 (en) 2009-07-01 2012-10-09 Protiva Biotherapeutics, Inc. Lipid formulations for nucleic acid delivery
US9512164B2 (en) 2009-07-07 2016-12-06 Alnylam Pharmaceuticals, Inc. Oligonucleotide end caps
WO2011015572A1 (en) 2009-08-03 2011-02-10 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of neurodegenerative diseases
WO2011015573A1 (en) 2009-08-03 2011-02-10 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of neurodegenerative diseases
AU2010299444B2 (en) 2009-09-24 2015-12-10 Anima Cell Metrology, Inc. Systems and methods for measuring translation of target proteins in cells
US8859776B2 (en) 2009-10-14 2014-10-14 Merck Sharp & Dohme Corp. Substituted piperidines that increase p53 activity and the uses thereof
US20120270930A1 (en) 2009-10-29 2012-10-25 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Methods and compositions for dysferlin exon-skipping
KR102487132B1 (en) 2009-11-12 2023-01-10 더 유니버시티 오브 웨스턴 오스트레일리아 Antisense Molecules and Methods for Treating Pathologies
CN102712928B (en) 2009-11-13 2017-08-04 萨雷普塔治疗公司 Antisense antiviral compound and the method for treating influenza infection
CA3044884A1 (en) 2009-12-07 2011-06-16 Arbutus Biopharma Corporation Compositions for nucleic acid delivery
KR101692063B1 (en) 2009-12-09 2017-01-03 닛토덴코 가부시키가이샤 MODULATION OF hsp47 EXPRESSION
US20130017223A1 (en) 2009-12-18 2013-01-17 The University Of British Columbia Methods and compositions for delivery of nucleic acids
WO2011094580A2 (en) 2010-01-28 2011-08-04 Alnylam Pharmaceuticals, Inc. Chelated copper for use in the preparation of conjugated oligonucleotides
US9068185B2 (en) 2010-03-12 2015-06-30 Sarepta Therapeutics, Inc. Antisense modulation of nuclear hormone receptors
WO2011120023A1 (en) 2010-03-26 2011-09-29 Marina Biotech, Inc. Nucleic acid compounds for inhibiting survivin gene expression uses thereof
WO2011123621A2 (en) 2010-04-01 2011-10-06 Alnylam Pharmaceuticals Inc. 2' and 5' modified monomers and oligonucleotides
WO2011133584A2 (en) 2010-04-19 2011-10-27 Marina Biotech, Inc. Nucleic acid compounds for inhibiting hras gene expression and uses thereof
US9725479B2 (en) 2010-04-22 2017-08-08 Ionis Pharmaceuticals, Inc. 5′-end derivatives
WO2011133868A2 (en) 2010-04-22 2011-10-27 Alnylam Pharmaceuticals, Inc. Conformationally restricted dinucleotide monomers and oligonucleotides
US10913767B2 (en) 2010-04-22 2021-02-09 Alnylam Pharmaceuticals, Inc. Oligonucleotides comprising acyclic and abasic nucleosides and analogs
SG184876A1 (en) 2010-04-23 2012-11-29 Univ Florida Raav-guanylate cyclase compositions and methods for treating leber's congenital amaurosis-1 (lca1)
WO2011139843A2 (en) 2010-04-28 2011-11-10 Marina Biotech, Inc. Multi-sirna compositions for reducing gene expression
ES2816898T3 (en) 2010-05-13 2021-04-06 Sarepta Therapeutics Inc Compounds that modulate the signaling activity of interleukins 17 and 23
AU2011255203B2 (en) 2010-05-21 2016-01-21 Peptimed, Inc. Reagents and methods for treating cancer
EP2576579B1 (en) 2010-06-02 2018-08-08 Alnylam Pharmaceuticals, Inc. Compositions and methods directed to treating liver fibrosis
US20130236968A1 (en) 2010-06-21 2013-09-12 Alnylam Pharmaceuticals, Inc. Multifunctional copolymers for nucleic acid delivery
KR101553753B1 (en) 2010-06-24 2015-09-16 쿠아크 파마수티칼스 인코퍼레이티드 Double stranded rna compounds to rhoa and use thereof
EP2584903B1 (en) 2010-06-24 2018-10-24 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
PT2591114T (en) 2010-07-06 2016-08-02 Glaxosmithkline Biologicals Sa Immunisation of large mammals with low doses of rna
HUE047796T2 (en) 2010-07-06 2020-05-28 Glaxosmithkline Biologicals Sa Delivery of rna to trigger multiple immune pathways
US20130171241A1 (en) 2010-07-06 2013-07-04 Novartis Ag Liposomes with lipids having an advantageous pka-value for rna delivery
BR112013000392B8 (en) 2010-07-06 2022-10-04 Novartis Ag PHARMACEUTICAL COMPOSITION CONTAINING VIRION-LIKE DISTRIBUTION PARTICLE FOR SELF-REPLICATING RNA MOLECULES AND THEIR USE
EP2596352B1 (en) 2010-07-22 2016-08-31 Ramot at Tel Aviv University, Ltd. Systems and methods for detection of cellular stress
WO2012016188A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
WO2012016184A2 (en) 2010-07-30 2012-02-02 Alnylam Pharmaceuticals, Inc. Methods and compositions for delivery of active agents
JP6043285B2 (en) 2010-08-02 2016-12-14 サーナ・セラピューティクス・インコーポレイテッドSirna Therapeutics,Inc. RNA interference-mediated inhibition of catenin (cadherin-binding protein) β1 (CTNNB1) gene expression using small interfering nucleic acids (siNA)
CA2807440A1 (en) 2010-08-04 2012-02-09 Cizzle Biotechnology Limited Methods and compounds for the diagnosis and treatment of cancer
US20120101108A1 (en) 2010-08-06 2012-04-26 Cell Signaling Technology, Inc. Anaplastic Lymphoma Kinase In Kidney Cancer
RU2624045C2 (en) 2010-08-17 2017-06-30 Сирна Терапьютикс,Инк RNA INTERFERENCE MEDIATED INHIBITION OF HEPATITIS B VIRUS (HBV) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US8883801B2 (en) 2010-08-23 2014-11-11 Merck Sharp & Dohme Corp. Substituted pyrazolo[1,5-a]pyrimidines as mTOR inhibitors
KR20130137160A (en) 2010-08-24 2013-12-16 머크 샤프 앤드 돔 코포레이션 Single-stranded rnai agents containing an internal, non-nucleic acid spacer
WO2012027467A1 (en) 2010-08-26 2012-03-01 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF PROLYL HYDROXYLASE DOMAIN 2 (PHD2) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
DK4066819T3 (en) * 2010-08-31 2023-04-24 Glaxosmithkline Biologicals Sa Small liposomes for delivery of RNA encoding immunogen
ES2935009T3 (en) 2010-08-31 2023-03-01 Glaxosmithkline Biologicals Sa Pegylated liposomes for delivery of RNA encoding immunogen
EP2613782B1 (en) 2010-09-01 2016-11-02 Merck Sharp & Dohme Corp. Indazole derivatives useful as erk inhibitors
US9242981B2 (en) 2010-09-16 2016-01-26 Merck Sharp & Dohme Corp. Fused pyrazole derivatives as novel ERK inhibitors
CA2812940A1 (en) 2010-09-27 2012-04-12 The Children's Hospital Of Philadelphia Compositions and methods useful for the treatment and diagnosis of inflammatory bowel disease
SG188666A1 (en) 2010-09-30 2013-05-31 Agency Science Tech & Res Methods and reagents for detection and treatment of esophageal metaplasia
US20140134231A1 (en) 2010-10-11 2014-05-15 Sanford-Burnham Medical Research Institute Mir-211 expression and related pathways in human melanoma
AU2011316707A1 (en) 2010-10-11 2013-05-09 Novartis Ag Antigen delivery platforms
EP2632472B1 (en) 2010-10-29 2017-12-13 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using short interfering nucleic acids (sina)
SG190088A1 (en) 2010-11-01 2013-06-28 Peptimed Inc Compositions of a peptide targeting system for treating cancer
EP2648763A4 (en) 2010-12-10 2014-05-14 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of klf-1 and bcl11a genes
WO2012078967A2 (en) 2010-12-10 2012-06-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for increasing erythropoietin (epo) production
WO2012087772A1 (en) 2010-12-21 2012-06-28 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012093384A1 (en) 2011-01-03 2012-07-12 Rosetta Genomics Ltd. Compositions and methods for treatment of ovarian cancer
WO2012099755A1 (en) 2011-01-11 2012-07-26 Alnylam Pharmaceuticals, Inc. Pegylated lipids and their use for drug delivery
US20120263754A1 (en) 2011-02-15 2012-10-18 Immune Design Corp. Methods for Enhancing Immunogen Specific Immune Responses by Vectored Vaccines
CA2828002A1 (en) 2011-03-03 2012-09-07 Quark Pharmaceuticals, Inc. Compositions and methods for treating lung disease and injury
US8420617B2 (en) 2011-03-11 2013-04-16 Biocell Laboratories Multiantivirus compound, composition and method for treatment of virus diseases
JP6108628B2 (en) 2011-03-29 2017-04-05 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Alnylam Pharmaceuticals, Inc. Composition and method for inhibiting expression of TMPRSS6 gene
EA027236B1 (en) 2011-04-08 2017-07-31 Иммьюн Дизайн Корп. Immunogenic compositions and methods of using the compositions for inducing humoral and cellular immune responses
CN103732592A (en) 2011-04-21 2014-04-16 默沙东公司 Insulin-like growth factor-1 receptor inhibitors
US10196637B2 (en) 2011-06-08 2019-02-05 Nitto Denko Corporation Retinoid-lipid drug carrier
TWI658830B (en) 2011-06-08 2019-05-11 日東電工股份有限公司 Retinoid-liposomes for enhancing modulation of hsp47 expression
CA2839896A1 (en) 2011-06-21 2012-12-27 Alnylam Pharmaceuticals, Inc. Assays and methods for determining activity of a therapeutic agent in a subject
WO2012177595A1 (en) 2011-06-21 2012-12-27 Oncofactor Corporation Compositions and methods for the therapy and diagnosis of cancer
CN103890000B (en) 2011-06-21 2017-09-01 阿尔尼拉姆医药品有限公司 (ANGPTL3) the iRNA compositions of angiopoietin-like 3 and its application method
EP3366312A1 (en) 2011-06-23 2018-08-29 Alnylam Pharmaceuticals, Inc. Serpina 1 sirnas: compositions of matter and methods of treatment
US20140227293A1 (en) 2011-06-30 2014-08-14 Trustees Of Boston University Method for controlling tumor growth, angiogenesis and metastasis using immunoglobulin containing and proline rich receptor-1 (igpr-1)
US11896636B2 (en) 2011-07-06 2024-02-13 Glaxosmithkline Biologicals Sa Immunogenic combination compositions and uses thereof
WO2013012752A2 (en) 2011-07-15 2013-01-24 Sarepta Therapeutics, Inc. Methods and compositions for manipulating translation of protein isoforms from alternative initiation start sites
EP2557089A2 (en) 2011-07-15 2013-02-13 Fundació Institut d'Investigació Biomèdica de Bellvitge (IDIBELL) Compositions and methods for immunomodulation
US20140328811A1 (en) 2011-08-01 2014-11-06 Alnylam Pharmaceuticals, Inc. Method for improving the success rate of hematopoietic stem cell transplants
WO2013049328A1 (en) 2011-09-27 2013-04-04 Alnylam Pharmaceuticals, Inc. Di-aliphatic substituted pegylated lipids
US9023865B2 (en) 2011-10-27 2015-05-05 Merck Sharp & Dohme Corp. Compounds that are ERK inhibitors
JP6368243B2 (en) 2011-11-11 2018-08-08 フレッド ハッチンソン キャンサー リサーチ センター T-cell immunotherapy targeted to cyclin A1 for cancer
PT2788487T (en) 2011-12-08 2018-07-03 Sarepta Therapeutics Inc Oligonucleotide analogues targeting human lmna
DK2790736T3 (en) 2011-12-12 2018-05-07 Oncoimmunin Inc In vivo delivery of oligonucleotides
CA2858630A1 (en) 2012-01-12 2013-07-18 Quark Pharmaceuticals, Inc. Combination therapy for treating hearing and balance disorders
EP2617434A1 (en) 2012-01-20 2013-07-24 Laboratorios Del. Dr. Esteve, S.A. HIV-1 integrase deficient immunogens and methods for loading dendritic cells with said immunogens
WO2013123996A1 (en) 2012-02-24 2013-08-29 Astrazeneca Uk Limited Novel sirna inhibitors of human icam-1
KR102239887B1 (en) 2012-02-24 2021-04-13 아뷰터스 바이오파마 코포레이션 Trialkyl cationic lipids and methods of use thereof
US9133461B2 (en) 2012-04-10 2015-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the ALAS1 gene
EP3336181B1 (en) 2012-04-18 2022-01-12 Cell Signaling Technology, Inc. Egfr and ros1 in cancer
US9127274B2 (en) 2012-04-26 2015-09-08 Alnylam Pharmaceuticals, Inc. Serpinc1 iRNA compositions and methods of use thereof
US20150299696A1 (en) 2012-05-02 2015-10-22 Sirna Therapeutics, Inc. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
EP2872147B1 (en) 2012-07-13 2022-12-21 Wave Life Sciences Ltd. Method for making chiral oligonucleotides
WO2014018375A1 (en) 2012-07-23 2014-01-30 Xenon Pharmaceuticals Inc. Cyp8b1 and uses thereof in therapeutic and diagnostic methods
US9611474B2 (en) 2012-09-12 2017-04-04 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to DDIT4 and methods of use thereof
WO2014043292A1 (en) 2012-09-12 2014-03-20 Quark Pharmaceuticals, Inc. Double-stranded oligonucleotide molecules to p53 and methods of use thereof
CA2882950A1 (en) 2012-09-28 2014-04-03 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
RU2660349C2 (en) 2012-11-28 2018-07-05 Мерк Шарп И Доум Корп. Compositions and methods for treatment of malignant tumour
EP3336187A1 (en) 2012-12-05 2018-06-20 Alnylam Pharmaceuticals, Inc. Pcsk9 irna compositions and methods of use thereof
WO2014100065A1 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted imidazopyridines as hdm2 inhibitors
AU2013364158A1 (en) 2012-12-20 2015-07-09 Sarepta Therapeutics, Inc. Improved exon skipping compositions for treating muscular dystrophy
WO2014114649A1 (en) 2013-01-22 2014-07-31 Technische Universität Graz Lipase inhibitors
WO2014120748A1 (en) 2013-01-30 2014-08-07 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as hdm2 inhibitors
US20150366890A1 (en) 2013-02-25 2015-12-24 Trustees Of Boston University Compositions and methods for treating fungal infections
BR122020016865B1 (en) 2013-03-14 2022-12-27 Sarepta Therapeutics, Inc. ANTISENSE OLIGONUCLEOTIDE, PHARMACEUTICAL COMPOSITION INCLUDING THE SAME AND USE OF SAID COMPOSITION FOR THE TREATMENT OF DUCHENNE MUSCULAR DYSTROPHY (DMD)
WO2014139884A2 (en) 2013-03-14 2014-09-18 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of fibrosis
PL2970964T3 (en) 2013-03-14 2019-06-28 Sarepta Therapeutics, Inc. Exon skipping compositions for treating muscular dystrophy
JP2016522675A (en) 2013-03-14 2016-08-04 ガラパゴス・ナムローゼ・フェンノートシャップGalapagos N.V. Molecular targets useful in the treatment of diseases associated with epithelial-mesenchymal transition and inhibitors of said targets
WO2014139883A1 (en) 2013-03-14 2014-09-18 Galapagos Nv Molecular targets and compounds, and methods to identify the same, useful in the treatment of fibrotic diseases
HUE034987T2 (en) 2013-03-14 2018-05-02 Alnylam Pharmaceuticals Inc Complement component c5 irna compositions and methods of use thereof
CA2906812A1 (en) 2013-03-15 2014-09-18 Sarepta Therapeutics, Inc. Improved compositions for treating muscular dystrophy
CA2918194C (en) 2013-03-27 2022-12-06 The General Hospital Corporation Methods and agents for treating alzheimer's disease
AR096203A1 (en) 2013-05-06 2015-12-16 Alnylam Pharmaceuticals Inc DOSAGES AND METHODS FOR MANAGING NUCLEIC ACID MOLECULES FORMULATED IN LIPIDS
US20160186263A1 (en) 2013-05-09 2016-06-30 Trustees Of Boston University Using plexin-a4 as a biomarker and therapeutic target for alzheimer's disease
BR112015029276B1 (en) 2013-05-22 2022-07-12 Alnylam Pharmaceuticals, Inc DUAL STRIP IRNA AGENT CAPABLE OF INHIBITING THE EXPRESSION OF TMPRSS6, PHARMACEUTICAL COMPOSITION AND USE THEREOF
PT2999785T (en) 2013-05-22 2018-07-09 Alnylam Pharmaceuticals Inc Serpina1 irna compositions and methods of use thereof
WO2014203189A1 (en) 2013-06-18 2014-12-24 Rosetta Genomics Ltd. Nanocarrier system for micrornas and uses thereof
DK3019200T3 (en) 2013-07-11 2022-06-20 Alnylam Pharmaceuticals Inc OLIGONUCLEOTIDE-LIGAND CONJUGATES AND METHOD OF PREPARATION
WO2015020960A1 (en) 2013-08-09 2015-02-12 Novartis Ag Novel lncrna polynucleotides
EP3041938A1 (en) 2013-09-03 2016-07-13 Moderna Therapeutics, Inc. Circular polynucleotides
EP2853595A1 (en) 2013-09-30 2015-04-01 Soluventis GmbH NOTCH 1 specific siRNA molecules
EP3052626A1 (en) 2013-10-02 2016-08-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
UA124961C2 (en) 2013-10-04 2021-12-22 Елнілем Фармасьютикалз, Інк. Compositions and methods for inhibiting expression of the alas1 gene
CN114053395A (en) 2013-10-08 2022-02-18 普罗麦迪奥股份有限公司 Methods for treating fibrotic cancer
IL282401B (en) 2013-12-12 2022-08-01 Alnylam Pharmaceuticals Inc Complement component irna compositions and methods of use thereof
CN112274641A (en) 2014-01-28 2021-01-29 巴克老龄化研究所 Methods and compositions for killing senescent cells and for treating senescence-associated diseases and disorders
EP3960860A3 (en) 2014-02-11 2022-06-08 Alnylam Pharmaceuticals, Inc. Ketohexokinase (khk) irna compositions and methods of use thereof
ES2738582T3 (en) 2014-02-14 2020-01-23 Immune Design Corp Cancer immunotherapy through a combination of local and systemic immunostimulation
TW201607559A (en) 2014-05-12 2016-03-01 阿尼拉製藥公司 Methods and compositions for treating a SERPINC1-associated disorder
AU2015264038B2 (en) 2014-05-22 2021-02-11 Alnylam Pharmaceuticals, Inc. Angiotensinogen (AGT) iRNA compositions and methods of use thereof
CA2955015A1 (en) 2014-07-15 2016-01-21 Immune Design Corp. Prime-boost regimens with a tlr4 agonist adjuvant and a lentiviral vector
SG11201701402QA (en) 2014-08-27 2017-03-30 Peptimed Inc Anti-tumor compositions and methods
WO2016040589A1 (en) 2014-09-12 2016-03-17 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting complement component c5 and methods of use thereof
WO2016044271A2 (en) 2014-09-15 2016-03-24 Children's Medical Center Corporation Methods and compositions to increase somatic cell nuclear transfer (scnt) efficiency by removing histone h3-lysine trimethylation
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
WO2016061487A1 (en) 2014-10-17 2016-04-21 Alnylam Pharmaceuticals, Inc. Polynucleotide agents targeting aminolevulinic acid synthase-1 (alas1) and uses thereof
EP3904519A1 (en) 2014-10-30 2021-11-03 Genzyme Corporation Polynucleotide agents targeting serpinc1 (at3) and methods of use thereof
JOP20200092A1 (en) 2014-11-10 2017-06-16 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
AU2015350120B2 (en) 2014-11-17 2021-05-27 Alnylam Pharmaceuticals, Inc. Apolipoprotein C3 (APOC3) iRNA compositions and methods of use thereof
US20180057839A1 (en) 2014-11-26 2018-03-01 The Regents Of The University Of California Therapeutic compositions comprising transcription factors and methods of making and using the same
WO2016083624A1 (en) 2014-11-28 2016-06-02 Silence Therapeutics Gmbh Means for inhibiting the expression of edn1
CA2976445A1 (en) 2015-02-13 2016-08-18 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
EP3268475B1 (en) 2015-03-11 2020-10-21 Yissum Research and Development Company of the Hebrew University of Jerusalem Ltd. Decoy oligonucleotides for the treatment of diseases
WO2016164746A1 (en) 2015-04-08 2016-10-13 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
WO2016201301A1 (en) 2015-06-12 2016-12-15 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions and methods of use thereof
WO2016205323A1 (en) 2015-06-18 2016-12-22 Alnylam Pharmaceuticals, Inc. Polynucleotde agents targeting hydroxyacid oxidase (glycolate oxidase, hao1) and methods of use thereof
WO2016209862A1 (en) 2015-06-23 2016-12-29 Alnylam Pharmaceuticals, Inc. Glucokinase (gck) irna compositions and methods of use thereof
US10494632B2 (en) 2015-07-10 2019-12-03 Alnylam Pharmaceuticals, Inc. Insulin-like growth factor binding protein, acid labile subunit (IGFALS) compositions and methods of use thereof
AU2016295168B2 (en) 2015-07-17 2021-08-19 Alnylam Pharmaceuticals, Inc. Multi-targeted single entity conjugates
CN108697659A (en) 2015-08-21 2018-10-23 费城儿童医院 For treating and the composition being applied in combination and method of diagnosis of autoimmune disease
CN114525280A (en) 2015-09-02 2022-05-24 阿尔尼拉姆医药品有限公司 iRNA compositions of programmed cell death 1 ligand 1(PD-L1) and methods of use thereof
EP3653216A1 (en) 2015-09-30 2020-05-20 Sarepta Therapeutics, Inc. Methods for treating muscular dystrophy
WO2017069958A2 (en) 2015-10-09 2017-04-27 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
WO2017075451A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting pou2af1
EP3368689B1 (en) 2015-10-28 2020-06-17 The Broad Institute, Inc. Composition for modulating immune responses by use of immune cell gene signature
WO2017075465A1 (en) 2015-10-28 2017-05-04 The Broad Institute Inc. Compositions and methods for evaluating and modulating immune responses by detecting and targeting gata3
WO2017184586A1 (en) 2016-04-18 2017-10-26 The Trustees Of Columbia University In The City Of New York Therapeutic targets involved in the progression of nonalcoholic steatohepatitis (nash)
MA45295A (en) 2016-04-19 2019-02-27 Alnylam Pharmaceuticals Inc HIGH DENSITY LIPOPROTEIN BINDING PROTEIN (HDLBP / VIGILINE) RNA COMPOSITION AND METHODS FOR USING THEM
NZ747685A (en) 2016-04-29 2023-05-26 Sarepta Therapeutics Inc Oligonucleotide analogues targeting human lmna
JP2019518028A (en) 2016-06-10 2019-06-27 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. Complement component C5i RNA composition and its use for treating paroxysmal nocturnal hemoglobinuria (PNH)
AU2017290231A1 (en) 2016-06-30 2019-02-07 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
EP3269734A1 (en) 2016-07-15 2018-01-17 Fundació Privada Institut d'Investigació Oncològica de Vall-Hebron Methods and compositions for the treatment of cancer
US20190262399A1 (en) 2016-09-07 2019-08-29 The Broad Institute, Inc. Compositions and methods for evaluating and modulating immune responses
US20200016202A1 (en) 2016-10-07 2020-01-16 The Brigham And Women's Hospital, Inc. Modulation of novel immune checkpoint targets
TW202313978A (en) 2016-11-23 2023-04-01 美商阿尼拉製藥公司 Serpina1 irna compositions and methods of use thereof
EP3330276A1 (en) 2016-11-30 2018-06-06 Universität Bern Novel bicyclic nucleosides and oligomers prepared therefrom
KR20230166146A (en) 2016-12-16 2023-12-06 알닐람 파마슈티칼스 인코포레이티드 Methods for treating or preventing ttr-associated diseases using transthyretin(ttr) irna compositions
MD3554553T2 (en) 2016-12-19 2022-10-31 Sarepta Therapeutics Inc Exon skipping oligomer conjugates for muscular dystrophy
BR112019012664A2 (en) 2016-12-19 2020-01-21 Sarepta Therapeutics Inc exon jump oligomer conjugates for muscular dystrophy
MX2019006989A (en) 2016-12-19 2019-08-16 Sarepta Therapeutics Inc Exon skipping oligomer conjugates for muscular dystrophy.
KR20200015895A (en) 2017-04-18 2020-02-13 알닐람 파마슈티칼스 인코포레이티드 How to treat a subject infected with hepatitis B virus (HBV)
WO2019014530A1 (en) 2017-07-13 2019-01-17 Alnylam Pharmaceuticals Inc. Lactate dehydrogenase a (ldha) irna compositions and methods of use thereof
EA201991450A1 (en) 2017-09-22 2019-12-30 Сарепта Терапьютикс, Инк. OLIGOMER CONJUGATES FOR EXONISM SKIP IN MUSCULAR DYSTROPHY
US20200254002A1 (en) 2017-09-28 2020-08-13 Sarepta Therapeutics, Inc. Combination therapies for treating muscular dystrophy
JP2020536060A (en) 2017-09-28 2020-12-10 サレプタ セラピューティクス, インコーポレイテッド Combination therapy to treat muscular dystrophy
US20210145852A1 (en) 2017-09-28 2021-05-20 Sarepta Therapeutics, Inc. Combination Therapies for Treating Muscular Dystrophy
WO2019089922A1 (en) 2017-11-01 2019-05-09 Alnylam Pharmaceuticals, Inc. Complement component c3 irna compositions and methods of use thereof
EP3706742B1 (en) 2017-11-08 2023-03-15 Merck Sharp & Dohme LLC Prmt5 inhibitors
US20200385719A1 (en) 2017-11-16 2020-12-10 Alnylam Pharmaceuticals, Inc. Kisspeptin 1 (kiss1) irna compositions and methods of use thereof
WO2019100039A1 (en) 2017-11-20 2019-05-23 Alnylam Pharmaceuticals, Inc. Serum amyloid p component (apcs) irna compositions and methods of use thereof
JP2021508491A (en) 2017-12-18 2021-03-11 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. High Mobility Group Box-1 (HMGB1) iRNA Composition and How to Use It
CN112105625A (en) 2018-03-07 2020-12-18 赛诺菲 Nucleotide precursors, nucleotide analogs, and oligomeric compounds containing the same
TW202016304A (en) 2018-05-14 2020-05-01 美商阿尼拉製藥公司 Angiotensinogen (agt) irna compositions and methods of use thereof
US10765760B2 (en) 2018-05-29 2020-09-08 Sarepta Therapeutics, Inc. Exon skipping oligomer conjugates for muscular dystrophy
EP3806868A4 (en) 2018-06-13 2022-06-22 Sarepta Therapeutics, Inc. Exon skipping oligomers for muscular dystrophy
TW202020153A (en) 2018-07-27 2020-06-01 美商薩羅塔治療公司 Exon skipping oligomers for muscular dystrophy
US20210309688A1 (en) 2018-08-07 2021-10-07 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2020033282A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
SG11202100715WA (en) 2018-08-13 2021-02-25 Alnylam Pharmaceuticals Inc HEPATITIS B VIRUS (HBV) dsRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
TW202020157A (en) 2018-08-16 2020-06-01 美商艾爾妮蘭製藥公司 Compositions and methods for inhibiting expression of the lect2 gene
JP2022500003A (en) 2018-09-18 2022-01-04 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. Ketohexokinase (KHK) iRNA composition and its usage
US10913951B2 (en) 2018-10-31 2021-02-09 University of Pittsburgh—of the Commonwealth System of Higher Education Silencing of HNF4A-P2 isoforms with siRNA to improve hepatocyte function in liver failure
US20220025367A1 (en) 2018-11-23 2022-01-27 Sanofi Novel rna compositions and methods for inhibiting angptl8
EA202191601A1 (en) 2018-12-13 2022-01-19 Сарепта Терапьютикс, Инк. OLIGOMER CONJUGATES FOR EXON SKIP IN MUSCULAR DYSTROPHY
AU2019406186A1 (en) 2018-12-20 2021-07-15 Praxis Precision Medicines, Inc. Compositions and methods for the treatment of KCNT1 related disorders
TW202039000A (en) 2018-12-20 2020-11-01 瑞士商休曼斯生物醫藥公司 Combination hbv therapy
WO2020187998A1 (en) 2019-03-19 2020-09-24 Fundació Privada Institut D'investigació Oncològica De Vall Hebron Combination therapy with omomyc and an antibody binding pd-1 or ctla-4 for the treatment of cancer
MX2021011498A (en) 2019-03-28 2022-01-04 Sarepta Therapeutics Inc Methods for treating muscular dystrophy with casimersen.
US20220193246A1 (en) 2019-04-18 2022-06-23 Sarepta Therapeutics, Inc. Compositions for treating muscular dystrophy
US20220296633A1 (en) 2019-06-19 2022-09-22 Sarepta Therapeutics, Inc. Methods for treating muscular dystrophy
WO2021009273A1 (en) 2019-07-16 2021-01-21 Université Cote D'azur Method of assessing the sensitivity or resistance of a subject to an oncolytic virus, recombinant virus, preparation and uses thereof
WO2021022108A2 (en) 2019-08-01 2021-02-04 Alnylam Pharmaceuticals, Inc. CARBOXYPEPTIDASE B2 (CPB2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP4007812A1 (en) 2019-08-01 2022-06-08 Alnylam Pharmaceuticals, Inc. Serpin family f member 2 (serpinf2) irna compositions and methods of use thereof
WO2021030522A1 (en) 2019-08-13 2021-02-18 Alnylam Pharmaceuticals, Inc. SMALL RIBOSOMAL PROTEIN SUBUNIT 25 (RPS25) iRNA AGENT COMPOSITIONS AND METHODS OF USE THEREOF
ES2918001A2 (en) 2019-08-14 2022-07-13 Acuitas Therapeutics Inc Improved lipid nanoparticles for delivery of nucleic acids
EP4022061A1 (en) 2019-08-27 2022-07-06 Sanofi Compositions and methods for inhibiting pcsk9
US20220290152A1 (en) 2019-09-03 2022-09-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of the lect2 gene
JP2022547888A (en) 2019-09-05 2022-11-16 サノフイ Oligonucleotides containing nucleotide analogues
CA3147641A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
US20220348908A1 (en) 2019-09-23 2022-11-03 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4alpha) GENE EXPRESSION
US20220389429A1 (en) 2019-10-04 2022-12-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing ugt1a1 gene expression
AU2020363372A1 (en) 2019-10-07 2022-05-19 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
EP3808763A1 (en) 2019-10-17 2021-04-21 Fundació Institut d'Investigació Biomèdica de Bellvitge (IDIBELL) Compounds for immunomodulation
EP4045652A1 (en) 2019-10-18 2022-08-24 Alnylam Pharmaceuticals, Inc. Solute carrier family member irna compositions and methods of use thereof
TW202134435A (en) 2019-10-22 2021-09-16 美商阿尼拉製藥公司 Complement component c3 irna compositions and methods of use thereof
EP4051796A1 (en) 2019-11-01 2022-09-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajb1-prkaca fusion gene expression
TW202132567A (en) 2019-11-01 2021-09-01 美商阿尼拉製藥公司 Huntingtin (htt) irna agent compositions and methods of use thereof
MX2022005692A (en) 2019-11-13 2022-06-08 Alnylam Pharmaceuticals Inc Methods and compositions for treating an angiotensinogen- (agt-) associated disorder.
EP4061945A1 (en) 2019-11-22 2022-09-28 Alnylam Pharmaceuticals, Inc. Ataxin3 (atxn3) rnai agent compositions and methods of use thereof
KR20220115995A (en) 2019-12-13 2022-08-19 알닐람 파마슈티칼스 인코포레이티드 Human chromosome 9 open reading frame 72 (C9orf72) iRNA preparation compositions and methods of using the same
WO2021126734A1 (en) 2019-12-16 2021-06-24 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
US20230067811A1 (en) 2020-01-24 2023-03-02 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
WO2021154941A1 (en) 2020-01-31 2021-08-05 Alnylam Pharmaceuticals, Inc. Complement component c5 irna compositions for use in the treatment of amyotrophic lateral sclerosis (als)
IL295445A (en) 2020-02-10 2022-10-01 Alnylam Pharmaceuticals Inc Compositions and methods for silencing vegf-a expression
MX2022010052A (en) 2020-02-18 2022-09-05 Alnylam Pharmaceuticals Inc Apolipoprotein c3 (apoc3) irna compositions and methods of use thereof.
WO2021178736A1 (en) 2020-03-06 2021-09-10 Alnylam Pharmaceuticals, Inc. KETOHEXOKINASE (KHK) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021178778A1 (en) 2020-03-06 2021-09-10 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of transthyretin (ttr)
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
EP4121534A1 (en) 2020-03-18 2023-01-25 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating subjects having a heterozygous alanine-glyoxylate aminotransferase gene (agxt) variant
WO2021195307A1 (en) 2020-03-26 2021-09-30 Alnylam Pharmaceuticals, Inc. Coronavirus irna compositions and methods of use thereof
EP4127171A2 (en) 2020-03-30 2023-02-08 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing dnajc15 gene expression
CA3179411A1 (en) 2020-04-06 2021-10-14 Alnylam Pharmaceuticals, Inc. Compositions and methods for silencing myoc expression
WO2021206917A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. ANGIOTENSIN-CONVERTING ENZYME 2 (ACE2) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
WO2021206922A1 (en) 2020-04-07 2021-10-14 Alnylam Pharmaceuticals, Inc. Transmembrane serine protease 2 (tmprss2) irna compositions and methods of use thereof
CN116134135A (en) 2020-04-07 2023-05-16 阿尔尼拉姆医药品有限公司 Compositions and methods for silencing SCN9A expression
CA3181400A1 (en) 2020-04-27 2021-11-04 Alnylam Pharmaceuticals, Inc. Apolipoprotein e (apoe) irna agent compositions and methods of use thereof
WO2021222549A1 (en) 2020-04-30 2021-11-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
WO2021231692A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of otoferlin (otof)
EP4150077A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of transmembrane channel-like protein 1 (tmc1)
WO2021231673A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of leucine rich repeat kinase 2 (lrrk2)
EP4150078A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate lyase (asl)
WO2021231675A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of argininosuccinate synthetase (ass1)
WO2021231680A1 (en) 2020-05-15 2021-11-18 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of methyl-cpg binding protein 2 (mecp2)
EP4150089A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of retinoschisin 1 (rs1)
EP4150087A1 (en) 2020-05-15 2023-03-22 Korro Bio, Inc. Methods and compositions for the adar-mediated editing of gap junction protein beta 2 (gjb2)
EP4153746A1 (en) 2020-05-21 2023-03-29 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting marc1 gene expression
AR122534A1 (en) 2020-06-03 2022-09-21 Triplet Therapeutics Inc METHODS FOR THE TREATMENT OF NUCLEOTIDE REPEAT EXPANSION DISORDERS ASSOCIATED WITH MSH3 ACTIVITY
BR112022024420A2 (en) 2020-06-18 2023-01-17 Alnylam Pharmaceuticals Inc XANTHINE DEHYDROGENASE (XDH) IRNA COMPOSITIONS AND METHODS OF USE THEREOF
AU2021296848A1 (en) 2020-06-24 2023-02-09 Humabs Biomed Sa Engineered hepatitis B virus neutralizing antibodies and uses thereof
WO2022066847A1 (en) 2020-09-24 2022-03-31 Alnylam Pharmaceuticals, Inc. Dipeptidyl peptidase 4 (dpp4) irna compositions and methods of use thereof
EP4225917A1 (en) 2020-10-05 2023-08-16 Alnylam Pharmaceuticals, Inc. G protein-coupled receptor 75 (gpr75) irna compositions and methods of use thereof
US20240035029A1 (en) 2020-10-16 2024-02-01 Sanofi Rna compositions and methods for inhibiting lipoprotein(a)
JP2023546103A (en) 2020-10-16 2023-11-01 サノフイ Novel RNA compositions and methods for inhibiting ANGPTL3
CN116368146A (en) 2020-10-20 2023-06-30 赛诺菲 Novel ligands for asialoglycoprotein receptors
AU2021365822A1 (en) 2020-10-21 2023-06-08 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating primary hyperoxaluria
WO2022087329A1 (en) 2020-10-23 2022-04-28 Alnylam Pharmaceuticals, Inc. Mucin 5b (muc5b) irna compositions and methods of use thereof
IL302709A (en) 2020-11-13 2023-07-01 Alnylam Pharmaceuticals Inc COAGULATION FACTOR V (F5) iRNA COMPOSITIONS AND METHODS OF USE THEREOF
EP4256053A1 (en) 2020-12-01 2023-10-11 Alnylam Pharmaceuticals, Inc. Methods and compositions for inhibition of hao1 (hydroxyacid oxidase 1 (glycolate oxidase)) gene expression
WO2022125490A1 (en) 2020-12-08 2022-06-16 Alnylam Pharmaceuticals, Inc. Coagulation factor x (f10) irna compositions and methods of use thereof
EP4274896A1 (en) 2021-01-05 2023-11-15 Alnylam Pharmaceuticals, Inc. Complement component 9 (c9) irna compositions and methods of use thereof
JP2024508714A (en) 2021-02-12 2024-02-28 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 Superoxide dismutase 1 (SOD1) iRNA composition and method for using the same for treating or preventing superoxide dismutase 1- (SOD1-) related neurodegenerative diseases
JP2024509783A (en) 2021-02-25 2024-03-05 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 Prion protein (PRNP) IRNA compositions and methods of use thereof
KR20230150844A (en) 2021-02-26 2023-10-31 알닐람 파마슈티칼스 인코포레이티드 Ketohexokinase (KHK) iRNA compositions and methods of using the same
CA3212128A1 (en) 2021-03-04 2022-09-09 Alnylam Pharmaceuticals, Inc. Angiopoietin-like 3 (angptl3) irna compositions and methods of use thereof
WO2022192519A1 (en) 2021-03-12 2022-09-15 Alnylam Pharmaceuticals, Inc. Glycogen synthase kinase 3 alpha (gsk3a) irna compositions and methods of use thereof
WO2022212231A2 (en) 2021-03-29 2022-10-06 Alnylam Pharmaceuticals, Inc. Huntingtin (htt) irna agent compositions and methods of use thereof
IL307298A (en) 2021-03-31 2023-11-01 Entrada Therapeutics Inc Cyclic cell penetrating peptides
WO2022212153A1 (en) 2021-04-01 2022-10-06 Alnylam Pharmaceuticals, Inc. Proline dehydrogenase 2 (prodh2) irna compositions and methods of use thereof
IL307926A (en) 2021-04-26 2023-12-01 Alnylam Pharmaceuticals Inc Transmembrane protease, serine 6 (tmprss6) irna compositions and methods of use thereof
EP4330396A1 (en) 2021-04-29 2024-03-06 Alnylam Pharmaceuticals, Inc. Signal transducer and activator of transcription factor 6 (stat6) irna compositions and methods of use thereof
EP4330395A1 (en) 2021-04-30 2024-03-06 Sarepta Therapeutics, Inc. Treatment methods for muscular dystrophy
WO2022240721A1 (en) 2021-05-10 2022-11-17 Entrada Therapeutics, Inc. Compositions and methods for modulating interferon regulatory factor-5 (irf-5) activity
CA3217463A1 (en) 2021-05-10 2022-11-17 Ziqing QIAN Compositions and methods for modulating mrna splicing
IL308353A (en) 2021-05-10 2024-01-01 Entrada Therapeutics Inc Compositions and methods for modulating tissue distribution of intracellular therapeutics
WO2022245583A1 (en) 2021-05-18 2022-11-24 Alnylam Pharmaceuticals, Inc. Sodium-glucose cotransporter-2 (sglt2) irna compositions and methods of use thereof
EP4341405A1 (en) 2021-05-20 2024-03-27 Korro Bio, Inc. Methods and compositions for adar-mediated editing
WO2022256283A2 (en) 2021-06-01 2022-12-08 Korro Bio, Inc. Methods for restoring protein function using adar
EP4347823A1 (en) 2021-06-02 2024-04-10 Alnylam Pharmaceuticals, Inc. Patatin-like phospholipase domain containing 3 (pnpla3) irna compositions and methods of use thereof
CN117561334A (en) 2021-06-04 2024-02-13 阿尔尼拉姆医药品有限公司 Human chromosome 9 open reading frame 72 (C9 ORF 72) iRNA pharmaceutical compositions and methods of use thereof
WO2022260939A2 (en) 2021-06-08 2022-12-15 Alnylam Pharmaceuticals, Inc. Compositions and methods for treating or preventing stargardt's disease and/or retinal binding protein 4 (rbp4)-associated disorders
AU2022298774A1 (en) 2021-06-23 2023-12-14 Entrada Therapeutics, Inc. Antisense compounds and methods for targeting cug repeats
WO2023278410A1 (en) 2021-06-29 2023-01-05 Korro Bio, Inc. Methods and compositions for adar-mediated editing
US20230194709A9 (en) 2021-06-29 2023-06-22 Seagate Technology Llc Range information detection using coherent pulse sets with selected waveform characteristics
KR20240026203A (en) 2021-06-30 2024-02-27 알닐람 파마슈티칼스 인코포레이티드 Methods and compositions for treating angiotensinogen (AGT)-related disorders
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
WO2023283403A2 (en) 2021-07-09 2023-01-12 Alnylam Pharmaceuticals, Inc. Bis-rnai compounds for cns delivery
WO2023003805A1 (en) 2021-07-19 2023-01-26 Alnylam Pharmaceuticals, Inc. Methods and compositions for treating subjects having or at risk of developing a non-primary hyperoxaluria disease or disorder
AU2022314619A1 (en) 2021-07-21 2024-01-04 Alnylam Pharmaceuticals, Inc. Metabolic disorder-associated target gene irna compositions and methods of use thereof
WO2023003995A1 (en) 2021-07-23 2023-01-26 Alnylam Pharmaceuticals, Inc. Beta-catenin (ctnnb1) irna compositions and methods of use thereof
WO2023009687A1 (en) 2021-07-29 2023-02-02 Alnylam Pharmaceuticals, Inc. 3-hydroxy-3-methylglutaryl-coa reductase (hmgcr) irna compositions and methods of use thereof
WO2023014677A1 (en) 2021-08-03 2023-02-09 Alnylam Pharmaceuticals, Inc. Transthyretin (ttr) irna compositions and methods of use thereof
KR20240042016A (en) 2021-08-04 2024-04-01 알닐람 파마슈티칼스 인코포레이티드 iRNA compositions and methods for silencing angiotensinogen (AGT)
WO2023014938A1 (en) 2021-08-05 2023-02-09 Sanegene Bio Usa Inc. 1'-alkyl modified ribose derivatives and methods of use
IL310407A (en) 2021-08-13 2024-03-01 Alnylam Pharmaceuticals Inc Factor xii (f12) irna compositions and methods of use thereof
WO2023044370A2 (en) 2021-09-17 2023-03-23 Alnylam Pharmaceuticals, Inc. Irna compositions and methods for silencing complement component 3 (c3)
CA3232420A1 (en) 2021-09-20 2023-03-23 Alnylam Pharmaceuticals, Inc. Inhibin subunit beta e (inhbe) modulator compositions and methods of use thereof
US20230241224A1 (en) 2021-09-22 2023-08-03 Sanegene Bio Usa Inc. 2'-alkyl or 3'- alkyl modified ribose derivatives and methods of use
AU2022361238A1 (en) 2021-10-05 2024-04-11 Sanegene Bio Usa Inc. Polyhydroxylated cyclopentane derivatives and methods of use
WO2023064530A1 (en) 2021-10-15 2023-04-20 Alnylam Pharmaceuticals, Inc. Extra-hepatic delivery irna compositions and methods of use thereof
CA3234811A1 (en) 2021-10-20 2023-04-27 Steven Goldman Rejuvenation treatment of age-related white matter loss
CA3234835A1 (en) 2021-10-22 2023-04-27 Korro Bio, Inc. Methods and compositions for disrupting nrf2-keap1 protein interaction by adar mediated rna editing
CA3234636A1 (en) 2021-10-29 2023-05-04 Alnylam Pharmaceuticals, Inc. Complement factor b (cfb) irna compositions and methods of use thereof
TW202334418A (en) 2021-10-29 2023-09-01 美商艾拉倫製藥股份有限公司 Huntingtin (htt) irna agent compositions and methods of use thereof
WO2023091704A1 (en) 2021-11-18 2023-05-25 Circularis Biotechnologies, Inc. Compositions and methods for production of circular nucleic acid molecules
WO2023122762A1 (en) 2021-12-22 2023-06-29 Camp4 Therapeutics Corporation Modulation of gene transcription using antisense oligonucleotides targeting regulatory rnas
WO2023141314A2 (en) 2022-01-24 2023-07-27 Alnylam Pharmaceuticals, Inc. Heparin sulfate biosynthesis pathway enzyme irna agent compositions and methods of use thereof
WO2023144792A1 (en) 2022-01-31 2023-08-03 Genevant Sciences Gmbh Poly(alkyloxazoline)-lipid conjugates and lipid particles containing same
WO2023144798A1 (en) 2022-01-31 2023-08-03 Genevant Sciences Gmbh Ionizable cationic lipids for lipid nanoparticles
US20230346819A1 (en) 2022-02-22 2023-11-02 Sanegene Bio Usa Inc. 5'-modified carbocyclic ribonucleotide derivatives and methods of use
WO2023178230A1 (en) 2022-03-17 2023-09-21 Sarepta Therapeutics, Inc. Phosphorodiamidate morpholino oligomer conjugates
WO2023240277A2 (en) 2022-06-10 2023-12-14 Camp4 Therapeutics Corporation Methods of modulating progranulin expression using antisense oligonucleotides targeting regulatory rnas
WO2024015796A1 (en) 2022-07-11 2024-01-18 Sanegene Bio Usa Inc. Optimized 2'- modified ribose derivatives and methods of use
WO2024039776A2 (en) 2022-08-18 2024-02-22 Alnylam Pharmaceuticals, Inc. Universal non-targeting sirna compositions and methods of use thereof
WO2024059165A1 (en) 2022-09-15 2024-03-21 Alnylam Pharmaceuticals, Inc. 17b-hydroxysteroid dehydrogenase type 13 (hsd17b13) irna compositions and methods of use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994010301A1 (en) * 1992-11-03 1994-05-11 Gene Shears Pty. Limited TNF-α RIBOZYMES AND DEGRADATION RESISTANT mRNA DERIVATIVES LINKED TO TNF-α RIBOZYMES

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5168053A (en) * 1989-03-24 1992-12-01 Yale University Cleavage of targeted RNA by RNAase P
US5272263A (en) * 1989-04-28 1993-12-21 Biogen, Inc. DNA sequences encoding vascular cell adhesion molecules (VCAMS)
IE911115A1 (en) * 1990-04-10 1991-10-23 Canji Inc Gene therapy for cell proliferative diseases
GB9106678D0 (en) * 1991-03-28 1991-05-15 Ferguson Mark W J Wound healing
CA2183992A1 (en) * 1994-02-23 1995-08-31 Dan T. Stinchcomb Method and reagent for inhibiting the expression of disease related genes

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994010301A1 (en) * 1992-11-03 1994-05-11 Gene Shears Pty. Limited TNF-α RIBOZYMES AND DEGRADATION RESISTANT mRNA DERIVATIVES LINKED TO TNF-α RIBOZYMES

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
EDGINGTON, S.M.: "ribozymes: stop making sense" BIOTECHNOLOGY, vol. 10, pages 256-262, NEW YORK US *
KISICH, K. & ERICKSON, K.: "ANTI-TUMOR NECROSIS FACTOR - ALPHA RIBOZYMES CAN SPECIFICALLY CLEAVE TUMOR NECROSIS FACTOR - ALPHA MESSENGER RNA WITHIN MAMMALIAN CELLS." J. LEUKOCYTE BIOL. 0 (SUPPL. 2). 1991. 70, & TWENTY-EIGHTH NATIONAL MEETING OF THE SOCIETY FOR LEUKOCYTE BIOLOGY AND THE TWENTY-FIRST LEUKOCYTE CULTURE CONFERENCE, ASPEN, COLORADO, USA, 28 September 1991 (1991-09-28), *
KISICH, K. ET AL.: "REGULATION OF TUMOR NECROSIS FACTOR - ALPHA GENE EXPRESSION FOLLOWING RIBOZYME TRANSFECTION A GENETICALLY ENGINEERED MODEL FOR THE TREATMENT OF TOXIC SHOCK." CLIN RES 41 (2). 137A, & JOINT MEETING OF THE ASSOCIATION OF AMERICAN PHYSICIANS, THE AMERICAN SOCIETY FOR CLINICAL INVESTIGATION, AND THE AMERICAN FEDERATION FOR CLINICAL RESEARCH, WASHINGTON, DC, USA, APRIL 30-MAY 3, 1993., *
See also references of WO9402595A1 *
SIOUD, M. ET AL.: "Preformed ribozyme destroys tumor necrosis factor mRNA in human cells" J. MOL. BIOL. 223 (4), 831-5, 20 February 1992 (1992-02-20), *

Also Published As

Publication number Publication date
AU4769893A (en) 1994-02-14
EP1251170A3 (en) 2002-10-30
EP0786522A3 (en) 1997-08-27
EP0786522A2 (en) 1997-07-30
JPH07509133A (en) 1995-10-12
EP0654077A4 (en) 1996-03-13
CA2140343A1 (en) 1994-02-03
EP1251170A2 (en) 2002-10-23
WO1994002595A1 (en) 1994-02-03
AU5209698A (en) 1998-03-19
MX9304329A (en) 1994-05-31

Similar Documents

Publication Publication Date Title
EP1251170A2 (en) Method and reagent for treatment of NF-kappaB dependent animal diseases
US5989906A (en) Method and reagent for inhibiting P-glycoprotein (mdr-1-gene)
EP0725788B1 (en) 2'-amido and 2'-peptido modified oligonucleotides
US6159692A (en) Method and reagent for inhibiting human immunodeficiency virus replication
US6831171B2 (en) Nucleic acid catalysts with endonuclease activity
US5750390A (en) Method and reagent for treatment of diseases caused by expression of the bcl-2 gene
US6017756A (en) Method and reagent for inhibiting hepatitis B virus replication
AU687001B2 (en) Method and reagent for inhibiting cancer development
US5972699A (en) Method and reagent for inhibiting herpes simplex virus replication
US5616490A (en) Ribozymes targeted to TNF-α RNA
EP0728205B1 (en) Reagent for treatment of arthritic conditions
WO1995004818A1 (en) Method and reagent for inhibiting human immunodeficiency virus replication
US5599704A (en) ErbB2/neu targeted ribozymes
US5801158A (en) Enzymatic RNA with activity to RAS
WO1998032846A2 (en) Enzymatic nucleic acid treatment of diseases or conditions related to levels of c-fos
US5639655A (en) PML-RARA targeted ribozymes
CA2398750A1 (en) Nucleozymes with endonuclease activity
WO1994029452A2 (en) Enzymatic rna molecules and their application in the treatment of fibrosis and fibrous tissue disease
US6544755B1 (en) Method and reagent for treatment of diseases by expression of the c-Myc gene
US6492512B1 (en) Method and reagent for treatment of lung cancer and other malignancies caused by the deregulation of L-MYC gene expression
AU5661700A (en) Method and reagent for treatment of animal diseases
CA2168566A1 (en) Method and reagent for inhibiting human immunodeficiency virus replication
AU7375594A (en) Method and reagent for inhibiting human immunodeficiency virus replication
AU6879501A (en) Methods and compositions for treatment of restenosis and cancer using ribozymes

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19950216

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU MC NL PT SE

RHK1 Main classification (correction)

Ipc: C12N 15/52

A4 Supplementary search report drawn up and despatched
AK Designated contracting states

Kind code of ref document: A4

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU MC NL PT SE

17Q First examination report despatched

Effective date: 19991216

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN