WO1995024202A1 - Ribozyme therapy for restenosis - Google Patents

Ribozyme therapy for restenosis Download PDF

Info

Publication number
WO1995024202A1
WO1995024202A1 PCT/US1995/002926 US9502926W WO9524202A1 WO 1995024202 A1 WO1995024202 A1 WO 1995024202A1 US 9502926 W US9502926 W US 9502926W WO 9524202 A1 WO9524202 A1 WO 9524202A1
Authority
WO
WIPO (PCT)
Prior art keywords
ribozyme
cell
smooth muscle
vector
dna
Prior art date
Application number
PCT/US1995/002926
Other languages
French (fr)
Inventor
Tsvi Goldenberg
Original Assignee
Immusol, 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 Immusol, Inc. filed Critical Immusol, Inc.
Priority to AU20990/95A priority Critical patent/AU2099095A/en
Priority to EP95913613A priority patent/EP0750503A4/en
Priority to JP7523634A priority patent/JPH09510101A/en
Publication of WO1995024202A1 publication Critical patent/WO1995024202A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • 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
    • C12YENZYMES
    • C12Y104/00Oxidoreductases acting on the CH-NH2 group of donors (1.4)
    • C12Y104/03Oxidoreductases acting on the CH-NH2 group of donors (1.4) with oxygen as acceptor (1.4.3)
    • C12Y104/03013Protein-lysine 6-oxidase (1.4.3.13), i.e. lysyl-oxidase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/11Protein-serine/threonine kinases (2.7.11)
    • C12Y207/11022Cyclin-dependent kinase (2.7.11.22)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C12N2310/30Chemical structure
    • C12N2310/35Nature of the modification
    • C12N2310/353Nature of the modification linked to the nucleic acid via an atom other than carbon
    • C12N2310/3531Hydrogen
    • 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
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/022Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from an adenovirus
    • 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
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/025Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a parvovirus

Definitions

  • SMC smooth muscle cell
  • this invention provides ribozymes and ribozyme delivery systems useful to inhibit abnormal smooth muscle cell proliferation in vascular tissue. Methods of producing ribozymes and gene therapy utilizing these ribozymes also are provided.
  • Restenosis is a major clinical problem and as the result of a need for repeat hospitalization, repeat angioplasty or bypass surgery, restenosis costs the nation's health care system in excess of $1 billion per year. Restenosis is now understood to comprise three important components. First, myointimal proliferation of vascular smooth muscle cells and the subsequent deposition of ECM results in a fibrocellular mass which can encroach upon the vascular lumen. Second, following acute angioplasty, there may be significant elastic recoil of the artery which contributes to a late loss of luminal dimension. Finally, platelets and thrombus adherent to the vascular wall may, over time, organize into a fibrocellular mass.
  • Smooth muscle cells have been demonstrated capable of producing and responding to a variety of growth factors including platelet-derived growth factor (PDGF) , transforming growth factor (TGF) , fibroblast growth factor (FGF) , insulin-like growth factor (IGF), and interleukins. These same factors have been found in human restenotic lesions.
  • PDGF platelet-derived growth factor
  • TGF transforming growth factor
  • FGF fibroblast growth factor
  • IGF insulin-like growth factor
  • interleukins interleukins.
  • This invention provides such by providing ribozymes and methods of using ribozymes that directly block the production of the growth factors, oncogenes and cell regulatory proteins involved with SMC growth following vascular injury.
  • ribozymes are intended to include RNA molecules that contain anti-sense sequences for specific recognition, and a RNA-cleaving enzymatic activity.
  • the catalytic strand cleaves a specific site in a target RNA at greater than stoichio etric concentration.
  • Two "types" of ribozymes are particularly useful in this invention, the hammerhead ribozyme (Rossi, J.J. et al., Pharmac. Ther. 50:245-254 (1991) incorporated herein by reference) and the hairpin ribozyme. (Ha pel et al., Nucl. Acids Res. 18:299-304 (1990) and U.S. Patent No.
  • this invention provides a ribozyme having the ability to inhibit pathologic smooth muscle proliferation and restenosis.
  • This ribozyme can be a hammerhead (for example, as described by Forster and Symons (1987) Cell 48:211-220; Haseloff and Gerlach (1988) Nature 328:596-600; Walbot and Bruening (1988) Nature 334:196; Haseloff and Gerlach (1988) Nature 334:585, each incorporated herein by reference) or a hairpin ribozyme (for example, as described by Haseloff et al., U.S. Patent No. 5,254,678, issued October 19, 1993 and Hempel et al., European Patent Publication No.
  • the ribozyme has the ability to inhibit the activity of a growth factor responsible for abnormal smooth muscle cell proliferation in vascular tissue.
  • growth factors include, but are not limited to platelet derived growth factor, fibroblast growth factor, insulin-like growth factor, c-myc, c-myb, c-fos, cdc2 kinase, TGF- ⁇ , TGF- ⁇ , interleukin, and components of the extracellular matrix.
  • abnormal smooth muscle cell proliferation shall mean any small cell proliferation and deposition of extracellular matrix occurring in response to vascular injury or trauma involved by any angioplasty, stent, balloon angioplasty, atherectomy, laser surgery, endovascular or surgical procedure.
  • the sequence requirement for the hairpin ribozyme is any RNA sequence consisting of NNNG/CN*GUCNNNNNNNNNN (where N*G is the cleavage site, and where N is any of G, U, C, or A) .
  • the sequence requirement at the cleavage site for the hammerhead ribozyme is any RNA sequence consisting of NUX (where N is any of G, U, C, or A and X represents C, U or A) can be targeted. Accordingly, the same target within the hairpin leader sequence, GUC, is useful for the hammerhead ribozyme.
  • the additional nucleotides of the hammerhead ribozyme or hairpin ribozyme is determined by the target flanking nucleotides and the hammerhead consensus sequence. This information, and the published sequence of nucleic acids coding for c-myc, c-myb, c-fos, cdc2 kinase, TGF- ⁇ and TGF- ⁇ proteins enables the production of the ribozymes of this invention. Appropriate base changes in the ribozyme is made to maintain the necessary base pairing with the target RNA sequences. Cech et al. (U.S. Patent No.
  • the ribozymes of this invention hybridize efficiently to target sequences at physiological temperatures, making them suitable for use in vivo, not merely as research tools (see, column 15, lines 18-42, of Cech et al., U.S. Patent No. 4,987,071).
  • the ribozymes of this invention and DNA encoding the ribozymes, described in more detail below, can be chemically synthesized using methods well known in the art. (For example, according to recommended protocols of Promega, Madison, Wis., USA, incorporated herein by reference) .
  • the ribozymes also can be prepared from a DNA molecule (that upon transcription, yields an RNA molecule) operably linked to an RNA polymerase promoter, e.g., the promoter for T7 RNA polymerase or SP6 RNA polymerase.
  • an RNA polymerase promoter e.g., the promoter for T7 RNA polymerase or SP6 RNA polymerase.
  • a nucleic acid molecule i.e., DNA or cDNA, coding for the ribozymes of this invention.
  • the ribozyme can be produced in vitro upon incubation with RNA polymerase and nucleotides.
  • the DNA is inserted into an expression cassette as described in Cotten and Birnstiel (1989) EMBO J. 8(12) :3861-3866 and in Hempel et al.. Biochemistry 28:4929- 4933 (1989), each incorporated herein by reference.
  • a more detailed discussion of molecular biology methodology is disclosed in Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, incorporated herein by reference.
  • the RNA molecule can be modified by ligation to a DNA molecule having the ability to stabilize the ribozyme and make it resistant to RNase.
  • the ribozyme can be modified to the phosphothio analog for use in liposome delivery systems. This modification also renders the ribozyme resistant to endonuclease activity.
  • the DNA molecule also can be in a host procaryotic or eucaryotic cell in culture or in the cells of an organism.
  • Appropriate procaryotic and eucaryotic cells can be transfected with an appropriate transfer vector containing the DNA molecule encoding a ribozyme of this invention.
  • the RNA can be produced in the host cell when the host cell is grown under suitable conditions favoring transcription of the DNA molecule.
  • the vector can be, but is not limited to a plasmid, a virus, a retrotransposon or a cos id. Examples of such vectors are disclosed in U.S. Patent No. 5,166,320, incorporated herein by reference.
  • a suitable adenoviral vector such as adeno-associated vector type 1 ("AAV-1") or adeno-associated vector type 2 (“AAV-2”) (see Chatterjee et al., (1992) Science Vol. 258:1485-1488, incorporated herein by reference) are particularly useful.
  • Methods of gene therapy are well known in the art, see, for example, Larrick, J.W. and Burck, K.L. Gene Therapy: Application of Molecular Biology, Elsevier Science Publishing Co., Inc. New York, New York (1991) and Kreigler, M. Gene Transfer and Expression: A Laboratory Manual, W.H. Freeman and Company, New York (1990), each incorporated herein by reference.
  • the nucleotide sequences coding for ribozymes are placed under the control of a strong promoter such as the lac, SV40 late, SV40 early, or lambda promoters. Ribozymes are then produced directly from the transfer vector in vivo.
  • the viral vector is a retrovirus, such as a non-virulent vaccinia based viral vector, a Moloney murine leukemia virus, or an HIV vector as described in Lever, A. (1989) J. Virol. 63:4085-4087, incorporated herein by reference. Retroviral vectors are particularly useful for gene therapy of restinosis.
  • An appropriate retroviral vector for such gene therapy is a self-replicating retrovirus having inserted between the 5' and 3' long terminal repeat (LTR) regions of the retrovirus a foreign nucleic acid (DNA or cDNA) sequence, under the control of a promoter, for example, the retroviral LTR or an inserted pol III promoter such as the human tRNA Val promoter or the adenovirus VA1 promoter.
  • LTR long terminal repeat
  • the retroviral vector then stably expresses the ribozyme in the host cell.
  • retroviral vectors having inserted between the 5' and 3' LTR more than one nucleic acid molecule encoding a ribozyme of this invention, each molecule under the control of a separate pol III promoter or alternatively, under the control of single pol III promoter or the retroviral LTR.
  • retroviral vectors provide the advantage of providing multi-functional therapy against restenosis, each specific therapy working in synergy.
  • the number of integration sites in the host cell are reduced thereby reducing the possibility of activation of host cellular DNA sequences by the inserted retroviral vector.
  • Host procaryotic and eucaryotic cells stably transduced with the vectors described above also are provided by this invention.
  • Suitable host cells include bacterial cells, mouse cells, rat cells, mouse cells, and human cells, for example, vascular smooth muscle cells.
  • a method of inhibiting abnormal smooth cell vascular tissue proliferation or restenosis is provided by this invention.
  • This method requires contacting a suitable cell or tissue with an effective amount ribozyme of this invention or alternatively, by transducing the cell with an effective amount vector having a nucleic acid molecule encoding the ribozyme.
  • Effective amounts are easily determined by those of skill in the art using well known methodology.
  • the RNA molecule can be embedded within a stable RNA molecule or in another form of protective environment, such as a liposome. Alternatively, the RNA can be embedded within RNase resistant DNA counterparts.
  • Cellular uptake of the exogenous ribozyme can be enhanced by attaching chemical groups to the DNA ends, such as cholesteryl moieties (Letsinger et al., P.N.A.S. , U.S.A. (1989), incorporated herein by reference).
  • chemical groups such as cholesteryl moieties (Letsinger et al., P.N.A.S. , U.S.A. (1989), incorporated herein by reference).
  • the target cell is transduced under conditions favoring insertion of the vector into the target cell and stable expression of the nucleic acid encoding the specific ribozyme.
  • the target cell can include but is not limited to a vascular smooth muscle cell or a cell responsible for the deposition of proteins involved in the formation of extracellular matrix. Such cells are well known to those of skill in the art. Accordingly, another aspect of this invention is a method for interfering with or preventing abnormal smooth muscle proliferation in a suitable cell by reacting the target RNA sequence with a ribozyme of this invention.
  • a transfer vector as described above encoding one or more ribozymes is transfected into a cell or cells using methods described in Llwewllyn et al., (1987) J. Mol. Biol. 195:115-123 and Hanahan et al. (1983) 166:557-580, each incorporated herein by reference.
  • the transfer vector replicates and the DNA coding for the ribozyme is transcribed by cellular polymerases to produce ribozymes which then inactivate factors responsible for abnormal smooth muscle cell proliferation, abnormal extracellular matrix deposition and restinosis.
  • Micromanipulation techniques such as microinjection also can be used to insert the vector into the cell so that the transfer vector or a part thereof is integrated into the genome of the cell. Transcription of the integrated material gives rise to ribozymes which then inactivate the target proteins.
  • the term "inactivate” is intended to mean interfere with the production of the protein product such as c-myc or TGF- ⁇ .
  • compositions also are provided by this invention. These compositions contain any of the above described ribozymes, DNA molecules, vectors or host cells. In one aspect of this invention, the compositions also contain a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents.
  • Host cell containing the ribozymes, nucleic acids and/or vectors described above also are within the scope of this invention.
  • These host cells can be procaryotic cells, for example bacterial cells or eucaryotic cells, such as mammalian, human, rat, or mouse cells.
  • the host cells transduced with nucleic acids encoding the ribozmyes are useful to recombinantly produce the ribozymes.
  • DNA encoding the ribozyme is provided to the cell, the DNA being under the transcriptional control of a promoter, using methods well known to those of skill in the art. See Sambrook et al. Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory (1989), incorporated herein by reference..
  • the DNA is then transcribed in the cell to produce the ribozyme.
  • the ribozyme can be purified or isolated from the cell by using methods well known in the art.
  • the DNA can be transferred in a carrier or in a vector in a carrier a number of ways.
  • the DNA can be administered by transluminal delivery to the vascular wall, or exoluminally.
  • the active ingredient can be embedded in a biodegradable polymer or sphere and administered by vascular stent. Alternatively, it can be delivered in a pleuronic gel.
  • An alternative method of inhibiting abnormal smooth muscle cell proliferation in vascular tissue consists of introducing into a cell an effective amount of DNA encoding a ribozyme described above, under conditions favoring transcription of the DNA to produce the ribozyme. This method also is useful to prevent abnormal SMC and abnormal extracellular matrix deposition and thus, prevent restenosis.
  • Also provided by this invention is a method of inhibiting or preventing abnormal smooth muscle cell proliferation in vascular tissue in a subject, which comprises administering to the subject an effective amount of a ribozyme having the ability to inhibit abnormal smooth muscle cell proliferation in vascular tissue.
  • the ribozyme is delivered to the smooth muscle cell exoluminally, transluminally, by stent, by a biodegradable polymer or sphere or in a pleuronic gel.

Abstract

As an effective therapy for restenosis, this invention provides ribozymes and ribozyme delivery systems useful to inhibit abnormal smooth muscle cell proliferation in vascular tissue. Methods of producing ribozymes and gene therapy utilizing these ribozymes also are provided.

Description

RIBOZYME THERAPY FOR RESTENOSIS
BACKGROUND OF THE INVENTION
In 1992, an excess of 300,000 angioplasties were performed in the United States. Restenosis is a major complication following angioplasty, occurring in 30-60% of patients. Indeed, restenosis is the single most significant problem in interventional cardiology and costs the health care system in excess of $ 1 billion per year.
Restenosis following angioplasty is the result of local vascular injury characterized by local infiltration of platelets, macrophages and local activation of the clotting system. These factors result in the elaboration of the number of biologic mediators of smooth muscle cell (SMC) migration and proliferation. Medial SMCs migrate into the vascular intima and begin to proliferate and produce extracellular matrix (ECM) , resulting in the formation of a fibrocellular mass which can obstruct blood flow.
The role of growth factors and cyto ines as mediators of SMC migration and proliferation has gained widespread attention. Further, injury has been shown to induce the expression of a variety of oncogenes that are believed to play a role in the cellular response to this injury. Thus, a need exists for an effective therapy to prevent and treat restenosis. This invention satisfies this need and provides related advantages as well. SUMMARY OF THE INVENTION
As an effective therapy for restenosis, this invention provides ribozymes and ribozyme delivery systems useful to inhibit abnormal smooth muscle cell proliferation in vascular tissue. Methods of producing ribozymes and gene therapy utilizing these ribozymes also are provided.
DETAILED DESCRIPTION OF THE INVENTION
Restenosis is a major clinical problem and as the result of a need for repeat hospitalization, repeat angioplasty or bypass surgery, restenosis costs the nation's health care system in excess of $1 billion per year. Restenosis is now understood to comprise three important components. First, myointimal proliferation of vascular smooth muscle cells and the subsequent deposition of ECM results in a fibrocellular mass which can encroach upon the vascular lumen. Second, following acute angioplasty, there may be significant elastic recoil of the artery which contributes to a late loss of luminal dimension. Finally, platelets and thrombus adherent to the vascular wall may, over time, organize into a fibrocellular mass.
Smooth muscle cells (SMCs) have been demonstrated capable of producing and responding to a variety of growth factors including platelet-derived growth factor (PDGF) , transforming growth factor (TGF) , fibroblast growth factor (FGF) , insulin-like growth factor (IGF), and interleukins. These same factors have been found in human restenotic lesions. In addition, a variety of oncogenes (e.g., c-myc, c-fos, and c-myb) have been found to be involved in smooth muscle cell migration and proliferation as well as deposition of ECM that is associated with post-vascular injury. Smooth muscle cells themselves are capable of regulating their own growth by local autocrine and paracrine mechanisms. Therefore, by interfering with the local production and action of the growth factors, oncogenes and cell regulatory proteins involved with SMC growth following vascular injury, one can effectively treat and prevent restenosis. This invention provides such by providing ribozymes and methods of using ribozymes that directly block the production of the growth factors, oncogenes and cell regulatory proteins involved with SMC growth following vascular injury.
As used herein, "ribozymes" are intended to include RNA molecules that contain anti-sense sequences for specific recognition, and a RNA-cleaving enzymatic activity. The catalytic strand cleaves a specific site in a target RNA at greater than stoichio etric concentration. Two "types" of ribozymes are particularly useful in this invention, the hammerhead ribozyme (Rossi, J.J. et al., Pharmac. Ther. 50:245-254 (1991) incorporated herein by reference) and the hairpin ribozyme. (Ha pel et al., Nucl. Acids Res. 18:299-304 (1990) and U.S. Patent No. 5,254,678, issued October 19, 1993, each incorporated herein by reference.) Because both hammerhead and hairpin ribozymes are catalytic molecules having antisense and endoribonucleotidase activity; ribozyme technology has emerged as a potentially powerful extension of the antisense approach to gene inactivation.
Accordingly, this invention provides a ribozyme having the ability to inhibit pathologic smooth muscle proliferation and restenosis. This ribozyme can be a hammerhead (for example, as described by Forster and Symons (1987) Cell 48:211-220; Haseloff and Gerlach (1988) Nature 328:596-600; Walbot and Bruening (1988) Nature 334:196; Haseloff and Gerlach (1988) Nature 334:585, each incorporated herein by reference) or a hairpin ribozyme (for example, as described by Haseloff et al., U.S. Patent No. 5,254,678, issued October 19, 1993 and Hempel et al., European Patent Publication No. 0 360 257, published March 26, 1990, each incorporated herein by reference) having the ability to specifically target, cleave and inactivate factors responsible for restinosis. In one embodiment, the ribozyme has the ability to inhibit the activity of a growth factor responsible for abnormal smooth muscle cell proliferation in vascular tissue. Such growth factors include, but are not limited to platelet derived growth factor, fibroblast growth factor, insulin-like growth factor, c-myc, c-myb, c-fos, cdc2 kinase, TGF-α, TGF-β, interleukin, and components of the extracellular matrix. As used herein, the term "abnormal smooth muscle cell proliferation" shall mean any small cell proliferation and deposition of extracellular matrix occurring in response to vascular injury or trauma involved by any angioplasty, stent, balloon angioplasty, atherectomy, laser surgery, endovascular or surgical procedure.
The sequence requirement for the hairpin ribozyme is any RNA sequence consisting of NNNG/CN*GUCNNNNNNNN (where N*G is the cleavage site, and where N is any of G, U, C, or A) . The sequence requirement at the cleavage site for the hammerhead ribozyme is any RNA sequence consisting of NUX (where N is any of G, U, C, or A and X represents C, U or A) can be targeted. Accordingly, the same target within the hairpin leader sequence, GUC, is useful for the hammerhead ribozyme. The additional nucleotides of the hammerhead ribozyme or hairpin ribozyme is determined by the target flanking nucleotides and the hammerhead consensus sequence. This information, and the published sequence of nucleic acids coding for c-myc, c-myb, c-fos, cdc2 kinase, TGF-α and TGF-β proteins enables the production of the ribozymes of this invention. Appropriate base changes in the ribozyme is made to maintain the necessary base pairing with the target RNA sequences. Cech et al. (U.S. Patent No. 4,987,071, issued January 22, 1991) has disclosed the preparation and use of certain synthetic ribozymes which have endoribonuclease activity. These ribozymes are based on the properties of the Tetrahymena ribosomal RNA self-splicing reaction and require an eight base pair target site. A temperature optimum of 50°C is reported for the endoribonuclease activity. The fragments that arise from cleavage contain 5 ' phosphate and 3' hydroxyl groups and a free guanosine nucleotide added to the 5' end of the cleaved RNA. In contrast, the ribozymes of this invention hybridize efficiently to target sequences at physiological temperatures, making them suitable for use in vivo, not merely as research tools (see, column 15, lines 18-42, of Cech et al., U.S. Patent No. 4,987,071).
The ribozymes of this invention and DNA encoding the ribozymes, described in more detail below, can be chemically synthesized using methods well known in the art. (For example, according to recommended protocols of Promega, Madison, Wis., USA, incorporated herein by reference) . The ribozymes also can be prepared from a DNA molecule (that upon transcription, yields an RNA molecule) operably linked to an RNA polymerase promoter, e.g., the promoter for T7 RNA polymerase or SP6 RNA polymerase. Accordingly, also provided by this invention is a nucleic acid molecule, i.e., DNA or cDNA, coding for the ribozymes of this invention. When the vector also contains an RNA polymerase promoter operably linked to the DNA molecule, the ribozyme can be produced in vitro upon incubation with RNA polymerase and nucleotides. In a separate embodiment, the DNA is inserted into an expression cassette as described in Cotten and Birnstiel (1989) EMBO J. 8(12) :3861-3866 and in Hempel et al.. Biochemistry 28:4929- 4933 (1989), each incorporated herein by reference. A more detailed discussion of molecular biology methodology is disclosed in Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, incorporated herein by reference. After synthesis, the RNA molecule can be modified by ligation to a DNA molecule having the ability to stabilize the ribozyme and make it resistant to RNase. Alternatively, the ribozyme can be modified to the phosphothio analog for use in liposome delivery systems. This modification also renders the ribozyme resistant to endonuclease activity.
The DNA molecule also can be in a host procaryotic or eucaryotic cell in culture or in the cells of an organism. Appropriate procaryotic and eucaryotic cells can be transfected with an appropriate transfer vector containing the DNA molecule encoding a ribozyme of this invention. When the DNA molecule is operatively linked to a promoter for RNA transcription, the RNA can be produced in the host cell when the host cell is grown under suitable conditions favoring transcription of the DNA molecule. The vector can be, but is not limited to a plasmid, a virus, a retrotransposon or a cos id. Examples of such vectors are disclosed in U.S. Patent No. 5,166,320, incorporated herein by reference. A suitable adenoviral vector such as adeno-associated vector type 1 ("AAV-1") or adeno-associated vector type 2 ("AAV-2") (see Chatterjee et al., (1992) Science Vol. 258:1485-1488, incorporated herein by reference) are particularly useful. Methods of gene therapy are well known in the art, see, for example, Larrick, J.W. and Burck, K.L. Gene Therapy: Application of Molecular Biology, Elsevier Science Publishing Co., Inc. New York, New York (1991) and Kreigler, M. Gene Transfer and Expression: A Laboratory Manual, W.H. Freeman and Company, New York (1990), each incorporated herein by reference.
To produce the ribozymes with a vector, the nucleotide sequences coding for ribozymes are placed under the control of a strong promoter such as the lac, SV40 late, SV40 early, or lambda promoters. Ribozymes are then produced directly from the transfer vector in vivo.
In a separate embodiment, the viral vector is a retrovirus, such as a non-virulent vaccinia based viral vector, a Moloney murine leukemia virus, or an HIV vector as described in Lever, A. (1989) J. Virol. 63:4085-4087, incorporated herein by reference. Retroviral vectors are particularly useful for gene therapy of restinosis. An appropriate retroviral vector for such gene therapy is a self-replicating retrovirus having inserted between the 5' and 3' long terminal repeat (LTR) regions of the retrovirus a foreign nucleic acid (DNA or cDNA) sequence, under the control of a promoter, for example, the retroviral LTR or an inserted pol III promoter such as the human tRNAVal promoter or the adenovirus VA1 promoter. The retroviral vector then stably expresses the ribozyme in the host cell.
Further provided by this invention are retroviral vectors having inserted between the 5' and 3' LTR more than one nucleic acid molecule encoding a ribozyme of this invention, each molecule under the control of a separate pol III promoter or alternatively, under the control of single pol III promoter or the retroviral LTR. These retroviral vectors provide the advantage of providing multi-functional therapy against restenosis, each specific therapy working in synergy. In addition, because only a limited number of vectors are used, the number of integration sites in the host cell are reduced thereby reducing the possibility of activation of host cellular DNA sequences by the inserted retroviral vector.
Host procaryotic and eucaryotic cells stably transduced with the vectors described above also are provided by this invention. Suitable host cells include bacterial cells, mouse cells, rat cells, mouse cells, and human cells, for example, vascular smooth muscle cells.
A method of inhibiting abnormal smooth cell vascular tissue proliferation or restenosis is provided by this invention. This method requires contacting a suitable cell or tissue with an effective amount ribozyme of this invention or alternatively, by transducing the cell with an effective amount vector having a nucleic acid molecule encoding the ribozyme. Effective amounts are easily determined by those of skill in the art using well known methodology. When exogenously delivering the ribozyme, the RNA molecule can be embedded within a stable RNA molecule or in another form of protective environment, such as a liposome. Alternatively, the RNA can be embedded within RNase resistant DNA counterparts. Cellular uptake of the exogenous ribozyme can be enhanced by attaching chemical groups to the DNA ends, such as cholesteryl moieties (Letsinger et al., P.N.A.S. , U.S.A. (1989), incorporated herein by reference).
In a separate embodiment, the target cell is transduced under conditions favoring insertion of the vector into the target cell and stable expression of the nucleic acid encoding the specific ribozyme. The target cell can include but is not limited to a vascular smooth muscle cell or a cell responsible for the deposition of proteins involved in the formation of extracellular matrix. Such cells are well known to those of skill in the art. Accordingly, another aspect of this invention is a method for interfering with or preventing abnormal smooth muscle proliferation in a suitable cell by reacting the target RNA sequence with a ribozyme of this invention. Within the cell or within the cells of an organism, a transfer vector as described above encoding one or more ribozymes is transfected into a cell or cells using methods described in Llwewllyn et al., (1987) J. Mol. Biol. 195:115-123 and Hanahan et al. (1983) 166:557-580, each incorporated herein by reference. Inside the cell, the transfer vector replicates and the DNA coding for the ribozyme is transcribed by cellular polymerases to produce ribozymes which then inactivate factors responsible for abnormal smooth muscle cell proliferation, abnormal extracellular matrix deposition and restinosis. Micromanipulation techniques such as microinjection also can be used to insert the vector into the cell so that the transfer vector or a part thereof is integrated into the genome of the cell. Transcription of the integrated material gives rise to ribozymes which then inactivate the target proteins. As used herein, the term "inactivate" is intended to mean interfere with the production of the protein product such as c-myc or TGF-β.
Compositions also are provided by this invention. These compositions contain any of the above described ribozymes, DNA molecules, vectors or host cells. In one aspect of this invention, the compositions also contain a pharmaceutically acceptable carrier. As used herein, the term "pharmaceutically acceptable carrier" encompasses any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents.
Host cell containing the ribozymes, nucleic acids and/or vectors described above also are within the scope of this invention. These host cells can be procaryotic cells, for example bacterial cells or eucaryotic cells, such as mammalian, human, rat, or mouse cells. The host cells transduced with nucleic acids encoding the ribozmyes are useful to recombinantly produce the ribozymes. Thus, also provided by this invention is a method for producing a ribozyme, in vitro or in vivo, the ribozyme being able to inhibit abnormal smooth muscle cell proliferation in vascular tissue. DNA encoding the ribozyme is provided to the cell, the DNA being under the transcriptional control of a promoter, using methods well known to those of skill in the art. See Sambrook et al. Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory (1989), incorporated herein by reference.. The DNA is then transcribed in the cell to produce the ribozyme. When produced in vitro, the ribozyme can be purified or isolated from the cell by using methods well known in the art. The DNA can be transferred in a carrier or in a vector in a carrier a number of ways. For example, the DNA can be administered by transluminal delivery to the vascular wall, or exoluminally. In another aspect, the active ingredient can be embedded in a biodegradable polymer or sphere and administered by vascular stent. Alternatively, it can be delivered in a pleuronic gel.
Further provided by this invention are methods of inhibiting abnormal smooth muscle cell proliferation in vascular tissue, by introducing into the cell one or more of the ribozymes described above, in an amount effective to inhibit SMC proliferation or abnormal extracellular matrix deposition.
An alternative method of inhibiting abnormal smooth muscle cell proliferation in vascular tissue consists of introducing into a cell an effective amount of DNA encoding a ribozyme described above, under conditions favoring transcription of the DNA to produce the ribozyme. This method also is useful to prevent abnormal SMC and abnormal extracellular matrix deposition and thus, prevent restenosis.
Also provided by this invention is a method of inhibiting or preventing abnormal smooth muscle cell proliferation in vascular tissue in a subject, which comprises administering to the subject an effective amount of a ribozyme having the ability to inhibit abnormal smooth muscle cell proliferation in vascular tissue. The ribozyme is delivered to the smooth muscle cell exoluminally, transluminally, by stent, by a biodegradable polymer or sphere or in a pleuronic gel.
It should be understood to those skilled in the art that "effective amounts" are administered. These amounts are easily determined using methods well-known to those of skill in the art.
Although the invention has been described with reference to the presently preferred embodiment, it should be understood that various modifications can be made without departing from the spirit of the invention. Accordingly, the invention is limited only by the following claims.

Claims

What is claimed is:
1. A ribozyme having the ability to inhibit abnormal smooth muscle cell proliferation in vascular tissue.
2. The ribozyme of claim 1, wherein the ribozyme has the ability to inhibit the activity of a growth factor responsible for abnormal smooth muscle cell proliferation in vascular tissue.
3. The ribozyme of claim 2, wherein the growth factor is platelet derived growth factor, fibroblast growth factor, TGF-α, TGF-β, c- myc, c-fos, c-myb, cdc2-kinase, insulin-like growth factor or an interleukin.
4. The ribozyme of claim 2, wherein the ribozyme specifically cleaves a protein responsible for the production of an extracellular matrix protein.
5. The ribozyme of claim 1, wherein the ribozyme has the ability to prevent abnormal deposition of extracellular matrix in vascular tissue.
6. A nucleic acid molecule encoding the ribozyme of claim 1.
7. The nucleic acid molecule of claim 6, wherein the nucleic acid is DNA or cDNA.
8. The nucleic acid molecule of claim 6, under the control of a promoter to transcribe the nucleic acid.
9. A host cell comprising the ribozyme of claim 1.
10. A vector comprising the nucleic acid of claim 6.
11. A host cell comprising the vector of claim
10.
12. The vector of claim 10, wherein the vector is a plasmid, a virus, retrotransposon or a cosmid.
13. The vector of claim 12, wherein the vector is an adenoviral vector or an AAV.
14. A host cell stably transformed with the vector of claim 13.
15. The host cell of claim 9 or 14, wherein the host cell is a human cell.
16. A method for producing a ribozyme, the ribozyme being able to inhibit abnormal smooth muscle cell proliferation in vascular tissue, which comprises providing DNA encoding the ribozyme under the transcriptional control of a promoter, transcribing the DNA to produce the ribozyme.
17. The method of claim 16, wherein the ribozyme is produced in vitro.
18. The method of claim 17, further comprising purifying the ribozyme produced.
19. The method of claim 16, wherein the ribozyme is produced in vivo.
20. A method of inhibiting abnormal smooth muscle cell proliferation in vascular tissue, which comprises introducing into the cell an effective amount of the ribozyme of claim 1.
21. A method of inhibiting abnormal smooth muscle cell proliferation in vascular tissue, which comprises introducing into the cell an effective amount of the DNA of claim 6 under conditions favoring transcription of the DNA to produce the ribozyme.
22. The method of claim 20 or 21, wherein the cell is a human cell.
23. A method of preventing abnormal smooth muscle cell proliferation in vascular tissue, which comprises introducing into the cell an effective amount of the DNA of claim 6 under conditions favoring transcription of the DNA to produce the ribozyme.
24. The method of claim 23, wherein the cell is a human cell.
25. A method of inhibiting or preventing abnormal smooth muscle cell proliferation in vascular tissue in a subject, which comprises administering to the subject an effective amount of the ribozyme of claim 1.
26. The method of claim 25, wherein the ribozyme is delivered to the smooth muscle cell exoluminally, transluminally, by stent, by a biodegradable polymer or sphere or in a pleuronic gel.
PCT/US1995/002926 1994-03-07 1995-03-06 Ribozyme therapy for restenosis WO1995024202A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU20990/95A AU2099095A (en) 1994-03-07 1995-03-06 Ribozyme therapy for restenosis
EP95913613A EP0750503A4 (en) 1994-03-07 1995-03-06 Ribozyme therapy for restenosis
JP7523634A JPH09510101A (en) 1994-03-07 1995-03-06 Ribozyme therapy for restenosis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US20764994A 1994-03-07 1994-03-07
US08/207,649 1994-03-07

Publications (1)

Publication Number Publication Date
WO1995024202A1 true WO1995024202A1 (en) 1995-09-14

Family

ID=22771442

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/002926 WO1995024202A1 (en) 1994-03-07 1995-03-06 Ribozyme therapy for restenosis

Country Status (5)

Country Link
EP (1) EP0750503A4 (en)
JP (1) JPH09510101A (en)
AU (1) AU2099095A (en)
CA (1) CA2184989A1 (en)
WO (1) WO1995024202A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997010334A2 (en) * 1995-09-12 1997-03-20 Immusol, Inc. Ribozyme therapy for the treatment and/or prevention of restenosis
WO1997011163A1 (en) * 1995-09-18 1997-03-27 Hans Peter Blankenborg Prydz Pskh-1 ribozymes and uses in disease treatment
WO1997032990A1 (en) * 1996-03-04 1997-09-12 Targeted Genetics Corporation Methods for transducing cells in blood vessels using recombinant aav vectors

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991002595A1 (en) * 1989-08-15 1991-03-07 Creo-Uni Co., Ltd. Nozzle for jetting self-vibratory cleaning fluid stream containing solid particles
WO1991015580A1 (en) * 1990-04-10 1991-10-17 Research Development Foundation Gene therapy for cell proliferative diseases
WO1991018012A1 (en) * 1990-05-14 1991-11-28 Biospan Corporation Cell internalizable conjugates and complexes including intracellularly cleavable moieties
WO1992000990A1 (en) * 1990-07-12 1992-01-23 Nova Pharmaceutical Corporation Interleukin receptor expression inhibiting antisense oligonucleotides
WO1992017206A1 (en) * 1991-03-28 1992-10-15 The Victoria University Of Manchester Wound healing
WO1993020691A1 (en) * 1992-04-20 1993-10-28 Tykocinski Mark L Method for inducing tumor immunity
WO1993023057A1 (en) * 1992-05-14 1993-11-25 Ribozyme Pharmaceuticals, Inc. Method and reagent for inhibiting cancer development

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69125374T2 (en) * 1990-06-19 1997-10-23 Gene Shears Pty Ltd ENDONUCLEASES
AU3129993A (en) * 1991-11-08 1993-06-07 Massachusetts Institute Of Technology Localized oligonucleotide therapy
US5646042A (en) * 1992-08-26 1997-07-08 Ribozyme Pharmaceuticals, Inc. C-myb targeted ribozymes

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991002595A1 (en) * 1989-08-15 1991-03-07 Creo-Uni Co., Ltd. Nozzle for jetting self-vibratory cleaning fluid stream containing solid particles
WO1991015580A1 (en) * 1990-04-10 1991-10-17 Research Development Foundation Gene therapy for cell proliferative diseases
WO1991018012A1 (en) * 1990-05-14 1991-11-28 Biospan Corporation Cell internalizable conjugates and complexes including intracellularly cleavable moieties
WO1992000990A1 (en) * 1990-07-12 1992-01-23 Nova Pharmaceutical Corporation Interleukin receptor expression inhibiting antisense oligonucleotides
WO1992017206A1 (en) * 1991-03-28 1992-10-15 The Victoria University Of Manchester Wound healing
WO1993020691A1 (en) * 1992-04-20 1993-10-28 Tykocinski Mark L Method for inducing tumor immunity
WO1993023057A1 (en) * 1992-05-14 1993-11-25 Ribozyme Pharmaceuticals, Inc. Method and reagent for inhibiting cancer development

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
ADVAN. ENZYME REGUL., Volume 32, issued 1992, FUNATO et al., "The Utility of an antifos Ribozyme in Reversing Cisplatin Resistance in Human Carcinomas", pages 195-209. *
J. CELL. BIOCHEM. SUPPL., Vol. 17, Part B, issued 1993, CONTENT et al., "Construction of a Ribozyme Cleaving Human IL-6 mRNA", page 110, abstract no. E 502. *
JOURNAL INTERFERON RESEARCH, Volume 12, Supplement 1, issued September 1992, MAHIEU et al., "Construction of a Ribozyme Cleaving Human IL-6 mRNA", page S79, abstract no. P4-6. *
JOURNAL OF MOLECULAR BIOLOGY, Vol. 242, No. 5, issued 1994, SIOUD, "Interaction Between Tumour Necrosis Factor alpha Ribozyme and Cellular Proteins. Involvement in Ribozyme Stability and Activity", pages 619-629. *
PROC. AM. ASSOC. CANCER RES. ANNU. MEET., Volume 31, issued March 1990, SCANLON et al., "Development of a c-fos Ribozyme to Circumvent Cisplatin Resistance in Human Ovarian Carcinoma Cells", page 404, abstract no. 2399. *
PROC. NATL. ACAD. SCI. U.S.A., Vol. 88, issued December 1991, SCANLON et al., "Ribozyme-mediated Cleavage of c-fos mRNA Reduces Gene Expression of DNA Synthesis Enzymes and Metallothionein", pages 10591-10595. *
See also references of EP0750503A4 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5834440A (en) * 1994-03-07 1998-11-10 Immusol Incorporated Ribozyme therapy for the inhibition of restenosis
WO1997010334A2 (en) * 1995-09-12 1997-03-20 Immusol, Inc. Ribozyme therapy for the treatment and/or prevention of restenosis
WO1997010334A3 (en) * 1995-09-12 1997-06-05 Immusol Inc Ribozyme therapy for the treatment and/or prevention of restenosis
WO1997011163A1 (en) * 1995-09-18 1997-03-27 Hans Peter Blankenborg Prydz Pskh-1 ribozymes and uses in disease treatment
US5856463A (en) * 1995-09-18 1999-01-05 Prydz; Hans Peter Blankenborg PSKH-1 ribozymes
WO1997032990A1 (en) * 1996-03-04 1997-09-12 Targeted Genetics Corporation Methods for transducing cells in blood vessels using recombinant aav vectors

Also Published As

Publication number Publication date
EP0750503A1 (en) 1997-01-02
EP0750503A4 (en) 1999-07-21
JPH09510101A (en) 1997-10-14
CA2184989A1 (en) 1995-09-14
AU2099095A (en) 1995-09-25

Similar Documents

Publication Publication Date Title
Marschall et al. Inhibition of gene expression with ribozymes
US5646042A (en) C-myb targeted ribozymes
Ohkawa et al. Importance of independence in ribozyme reactions: kinetic behavior of trimmed and of simply connected multiple ribozymes with potential activity against human immunodeficiency virus.
US6100087A (en) Ribozymes targeted to human CCR5 mRNA
US20030036517A1 (en) Nucleic acid mediated rna tagging and rna revision
Rossi Therapeutic antisense and ribozymes
US5834440A (en) Ribozyme therapy for the inhibition of restenosis
Santiago et al. Nucleic acid based strategies as potential therapeutic tools: mechanistic considerations and implications to restenosis
WO2000032765A2 (en) Ribozyme therapy for the treatment and/or prevention of restenosis
US6103890A (en) Enzymatic nucleic acids that cleave C-fos
Hauswirth et al. Ribozyme uses in retinal gene therapy
EP0750503A1 (en) Ribozyme therapy for restenosis
AU757789B2 (en) Method and reagents for the treatment of diseases or conditions related to molecules involved in angiogenic responses
ROSSI et al. Exploring the use of antisense, enzymatic RNA molecules (ribozymes) as therapeutic agents
WO1995022600A1 (en) Ribozyme therapy for hepatitis b infection
Phylactou et al. Potential therapy paradigms for Marfan syndrome
Simons Antisense approach to restenosis
Poggi et al. Ribozymes as therapeutic agents and genetic tools
WO1999055856A2 (en) Nucleic acid enzyme for rna cleavage
KR20100083575A (en) Ribozyme and dnazyme that specifically cleave tel-aml1 chimeric mrna
MXPA00009431A (en) Method and reagents for the treatment of diseases or conditions related to molecules involved in angiogenic responses
Lavrovsky et al. Ribozyme Technology and Drug Development
AU3918899A (en) Methods and compositions for treatment of restenosis and cancer using ribozymes
AU2005242129A1 (en) Catalytic molecules

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AU BB BG BR BY CA CN CZ EE FI GE HU JP KG KP KR KZ LK LR LT LV MD MG MN MW MX NO NZ PL RO RU SI SK TJ TT UA UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2184989

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1995913613

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1995913613

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1995913613

Country of ref document: EP