CA2157782A1 - Gene transfer for treating a connective tissue of a mammalian host - Google Patents

Gene transfer for treating a connective tissue of a mammalian host

Info

Publication number
CA2157782A1
CA2157782A1 CA002157782A CA2157782A CA2157782A1 CA 2157782 A1 CA2157782 A1 CA 2157782A1 CA 002157782 A CA002157782 A CA 002157782A CA 2157782 A CA2157782 A CA 2157782A CA 2157782 A1 CA2157782 A1 CA 2157782A1
Authority
CA
Canada
Prior art keywords
interleukin
gene
employing
mammalian host
including employing
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.)
Expired - Lifetime
Application number
CA002157782A
Other languages
French (fr)
Inventor
Joseph C. Glorioso
Christopher H. Evans
Paul D. Robbins
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.)
University of Pittsburgh
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2157782A1 publication Critical patent/CA2157782A1/en
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/545IL-1
    • 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
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/15Humanized animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/02Animal zootechnically ameliorated
    • A01K2267/025Animal producing cells or organs for transplantation
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Veterinary Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • General Engineering & Computer Science (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Epidemiology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The subject invention concerns a method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host including employing recombinant to produce a DNA vector molecule which contains the gene encoding for the product and infecting the connective cell of the mammalian host using the DNA vector molecule using the gene coding for the product. A method is provided for introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host employing non-viral means. A method to produce an animal model for the study of connective tissue pathology is also disclosed. Additionally, this invention provides a method of using in vivo a gene encoding an extracellular interleukin-1 binding domain of an interleukin-1 receptor.

Description

GBN13: TR~N~FB~ FOR q'R~:ATING A CON~-~v~; TI881JE
OF A MA~'`r-T~N HO8T

~ROSS-~EF~RENCE TO p~F~T.~TF~n APPLICATION
This is a continuation-in-part application of United States Application Serial No. 07/630,981, filed Decem~er 20, 1990, now r~n~ing.
sAcKGRouND OF 'rR~ lN V~llON
Field Of The Invent;on The present invention relates to a method of i~.LLoducing at least one gene ~nco~;ng a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host. This method discloses employing DNA vector molecules cont~ining a gene encoding the product and infecting the conn~çtive tissue cells of the mammalian host using the DNA vector molecule. This invention provides a method of introducing at least one gene ~nco~ing a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian hcst including employing non-viral means for effecting such introduction.
The present invention also relates to a method to produce an animal model for the study of connective tissue pathology.
The present invention further relates to a method of using a gene ~nco~;ng a truncated interleukin-l receptor to resist the deleterious pathological changes associated with arthritis. More specifically, this invention provides a method wherein a gene coding for an extracellular interleukin-1 binding domain of an interleukin-l receptor is introduced into synovial cells of a mammalian host in vivo for neutralizing the destructive activity of interleukin-l upon cartilage and other soft tissues. As an alternative, the patients own synovial cells are transduced in vitro and WO94/20517 PCT~S94/0~14 .

introduced back into the affected joint, using transplantation procedures such as for example, intra-articular injection.
As an alternative to the in vitro manipulation of synovia, the gene encoding the product of interest is introduced into liposomes and injected directly into the area of the joint, where the liposomes fuse with synovial cells, resulting in an in vivo gene transfer to synovial tissue. As an additional alternative ~o the n vitro manipulation of synovia, the gene encoding the product of interest is introduced into the area of the joint as naked DNA. The naked DNA enters the synovial cell, resulting in an in vivo gene transfer to synovial tissue.
As an another alternative, hematopoietic progenitor cells or the mature lymphoid or myeloid cells may be transfected n vitro, recovered and injested into the bone marrow of the patient using tec-hniques known to the skilled artisan.
Brief Description Of The Related Art Arthritis involves inflammation of a joint that is usually accompanied by pain and frequently changes in struc-ture. Arthritis may result from or be associated with a number of conditions including infection, immunological disturbances, trauma and degenerative joint diseases such as, for example, osteoarthritis. The biochemistry of cartilage degradation in joints and cellular changes have received considerable investigation.
In a healthy joint, cells in cartilage (chondrocytes) and the surrounding synovium (synoviocytes) are in a resting state. In this resting state, these cells secrete basal levels of prostaglandin E2 and various neutral proteinases, such as, for example, collagenase, gelatinase and stromelysin, with the ability to degrade cartilage. During the development of an arthritic condition, these cells become activated. In the activated state, synoviocytes and WO94/20517 PCT~S94/0~14 .

chondrocytes synthesize and secrete large amounts of prostaglandin E2 and neutral proteinases.
In efforts to identify pathophysiologically relevant cell activators, it has been known that the cytokine interleukin-1 activates chondrocytes and synoviocytes and induces cartilage breakdown n vitro and in vivo.
Additionally, interleukin-1 is a growth factor for synoviocytes and promotes their synthesis of matrix, two properties suggesting the involvement of interleukin-1 in the synovial hypertrophy that accompanies arthritis. In contrast, interleukin-1 inhibits cartilaginous matrix synthesis by chondrocytes, thereby suppressing repair of cartilage.
Interleukin-1 also induces bone resorption and thus may account for the loss of bone density seen in rheumatoid arthritis. Interleukin-1 is inflammatory, serves as a growth factor for lymphocytes, is a chemotactic factor and a possible activator of polymorphonuclear leukocytes (PMNs). When present in a sufficient concentration, interleukin-1 may cause fever, muscle wasting and sleepiness.
The major source of interleukin-1 in the joint is the synovium. Interleukin-1 is secreted by the resident synoviocytes, which are joined under inflammatory conditions by macrophages and other white blood cells.
Much attention has been devoted to the development of a class of agents identified as the "Non-Steroidal Anti-Inflammatory Drugs" (hereinafter "NSAIDs"). The NSAIDs inhibit cartilage synthesis and repair and control inflamma-tion. The mechanism of action of the NSAIDs appears to be associated principally with the inhibition of prostaglandin synthesis in body tissues. Most of this development has involved the synthesis of better inhibitors of cyclo-oxygenase, a key enzyme that catalyzes the formation of prostaglandin precursors (endoperoxides) from arachidonic acid. The anti-inflammatory eEfect of the NSAIDs is thought WO94/20~17 PCT~S94/0~14 ~- 2i~ 82 to be due in part to inhibition of prostaglandin synthesis and release during inflammation. Prostagl~n~;n~ are also believed to play a role in modulating the rate and extent of leukocyte infiltration during inflammation. The NSAIDs include, such as, for example, acetylsalicylic acid (aspirin), fenoprofen calcium (Nalfon~ Pulvules~, Dista Products Company), ibuprofen (Motrin~, The Upjohn Company), and indomethacin (Indocin~, Merck, Sharp & Dohme).
In contrast, the studies upon which the present invention is based show that production of the various neutral proteinases with the ability to degrade cartilage occurs even if prostaglandin synthesis is completely blocked.
Therapeutic intervention in arthritis is hindered by the inability to target drugs, such as the NSAIDs, to specific areas within a mammalian host, such as, for example a joint.
Traditional routes of drug delivery, such as for example, oral, intravenous or intramuscular a~r;n;stration, depend upon vascular perfusion of the synovium to carry the drug to the joint. This is inefficient because transynovial transfer of small molecules from the synovial capillaries to the joint space occurs generally by passive diffusion. This diffusion is less efficient with increased size of the target molecule.
Thus, the access of large drug molecules, for example, proteins, to the joint space is substantially restricted.
Intra-articular injection of drugs circumvents those limita-tions; however, the half-life of drugs administered intra-articularly is generally short. Another disadvantage of intra-articular injection of drugs is that frequent repeated injections are necessary to obtain acceptable drug levels at the joint spaces for treating a chronic condition such as, for example, arthritis. Because therapeutic agents heretofore could not be selectively targeted to joints, it was necessary to expose the mammalian host to systemically high concentrations of drugs in order to achieve a sustained, WO94t20517 21 S 7 7 8 2 PCT~S94/0~14 intra-articular therapeutic dose. Exposure of non-target organs in this manner exacerbated the tendency of anti-arthritis drugs to produce serious side effects, such as for example, gastrointestinal upset and changes in the hemato-logical, cardiovascular, hepatic and renal systems of the mammalian host.
It has been shown that genetic material can be introduced into mammalian cells by chemical or biologic means.
Moreover, the introduced genetic material can be expressed so that high levels of a specific protein can be synthesized by the host cell. Cells retaining the introduced genetic material may include an antibiotic resistance gene thus providing a selectable marker for preferential growth of the transduced cell in the presence of the corresponding antibiotic. Chemical compounds for inhibiting the production of interleukin-1 are also known.
U.S. Patent No. 4,778,806 discloses a method of inhibiting the production of interleukin-l by monocytes and/or macrophages in a human by administering through the parenteral route a 2-2'-[1,3-propan-2-onediyl-bis (thio)] bis-l H-imidazole or a pharmaceutically acceptable salt thereof. This patent discloses a chemical compound for inhibiting the production of interleukin-l. By contrast, in one embodiment of the present invention, gene therapy is employed that is capable of binding to and neutralizing interleukin-1.
U.S. Patent No. 4,780,470 discloses a method of inhibiting the production of interleukin-1 by monocytes in a human by administering a 4,5-diaryl-2 (substituted) imidazole.
This patent also discloses a chemical compound for inhibiting the production of interleukin-1.
U.S. Patent No. 4,794,114 discloses a method of inhibiting the 5-lipoxygenase pathway in a human by administering a diaryl-substituted imidazole fused to a thiazole, pyrrolidine or piperidine rirg or a pharmaceutically WOg4/20517 PCT~S94/0~14 21~7782 acceptable salt thereof. This patent also discloses a chemical compound for inhibiting the production of interleukin-l.
U.S. Pater.t No. 4,870,101 discloses a method for inhibiting the release of interleukin-1 and for alleviating interleukin-l mediated condit~ns by administering an effective amount of a pharmaceutically acceptable anti-oxidant compound such as disulfiram, tetrakis t3-(2,6-di-tert-butyl-4-hydroxyphenyl) propionyloxy methyl] methane or 2,4-di-isobutyl-~-(N,N-dimethylamino methyl)-phenol. This patent discloses a chemical compound for inhibiting the release of interleukin-1.
U.S. Patent No. 4,816,436 discloses a process for the use of interleukin-1 as an anti-arthritic agent. This patent states that interleukin-1, in association with a pharmaceutical carrier, may be administered by intra-articular injection for the treatment of arthritis or inflammation. In contrast, the present invention discloses a method of using and preparing a gene that is capable of binding to and neutralizing interleukin-1 as a method of resisting arthritis.
U.S. Patent No. 4,935,343 discloses an immunoassay method for the detection of interleukin-1 beta that employs a monoclonal antibody that binds to interleukin-1 beta but does not bind to interleukin-1 beta. This patent discloses that the monoclonal antibody binds to interleukin-1 beta and blocks the binding of interleukin-1 beta to nterleukin-1 receptors, and thus blocking the biological activity of interleukin-1 beta. The monoclonal antibody disclosed in this patent may be obtained by production of an immunogen through genetic engineering using recombinant DNA technology. The immunogen is injected into a mouse and thereafter spleen cells of the mouse are immortalized by fusing the spleen cells with myeloma cells. The resulting cells include the hybrid continuous cell lines (hybridomas) that may be later screened for monoclonal WO94/20517 PCT~S94/0~14 .

antibodies. This patent states that the monoclonal antibodies of the invention may be used therapeutically, such as for example, in the immunization of a patient, or the monoclonal antibodies may be bound to a toxin to form an immunotoxin or to a radioactive material or drug tc form a radio pharmaceutical or pharmaceutical.
U.S. Patent No. 4,766,069 discloses a recombinant DNA cloning vehicle having a DNA sequence comprising the human interleukin-l gene DNA sequence. This patent provides a process for preparing human interleukin-l beta, and recovering the human interleukin-l beta. This patent discloses use of interleukin-l as an immunological reagent in humans because of its ability to stimulate T-cells and B-cells and increase immunoglobulin synthesis.
U.S. Patent No. 4,396,601 discloses a method for providing mammalian hosts with additional genetic capability.
This patent provides that host cells capable of regeneration are removed from the host and treated with genetic material including at least one marker which allows for selective advantage for the host cells in which the genetic material is capable of expression and replication. This patent states that the modified host cells are then returned to the host under regenerative conditions. In the present invention, genetic material may be directly introduced (a) into host cells in vivo or (b) into synoviocytes in vitro for subsequent transplantation back into the patient's joints.
U.S. Patent No. 4,968,607 discloses a DNA sequence encoding a mammalian interleukin-l receptor protein which exhibits interleukin-l binding activity.
In spite of these prior art disclosures, there remains a very real and substantia7 need for a method or introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host in vitro, or alternatively ln vivo, for use in treating the mammalian WO94/20517 PCT~S94/0~14 .
2 15 ~ 7 8 host. Further, there is a need for a process wherein a gene encoding a truncated interleukin-1 receptor is used to resist the deleterious pathological changes associated with arthritis. More specifically there is a need for such a process where a gene coding for ~he extracellular interleukin-1 binding domain of the interleukin-1 receptor, capable of binding to and neutralizing interleukin-1 is expressed in host synovial cells in vivo.
SUMMA~Y OF THE INVENTION
The present invention has met the hereinbefore described need. A method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host is provided for in the present invention. This method includes employing recombinant t~c-hniques to produce a DNA
vector molecule containing the gene encoding for the product and infecting the connective tissue cell of the mammalian host using the DNA vector molecule containing the gene coding for the product. The DNA vector molecule can be any DNA molecule capable of being delivered and maintained within the target cell or tissue such that the gene encoding the product of interest can be stably expressed. The DNA vector molecule preferably utilized in the present invention is either a viral DNA vector molecule or a plasmid DNA viral molecule. This method preferably includes introducing the gene encoding the product into the cell of the mammalian connective tissue for a therapeutic use.
More specifically, this method includes employing as the gene a gene capable of encoding at least one of the materials which is selected from the group which includes (a) a human interleukin-l receptor antagonist protein or a biologically active derivative or fragment thereof, (b) a Lac Z marker gene capable of encoding a beta-galactosidase protein or a biologically active derivative or fragment WO94/20517 . PCT~S94/0~14 .

21~7782 thereof, (c) a soluble interleukin-l receptor protein or a biologically active derivative or fragment thereof, (d) a prot~;n~e inhibitor, and (e) a cytokine, and employing as the viral vector at least one vector which is selected from the group which includes (a) a retroviral vector including at least one of the materials selected from the group which includes MFG and BAG, (b) an adeno-associated virus, (c) an adenovirus, and (d) a herpes virus, including but not limited to herpes simplex 1 or herpes simplex 2.
A further embodiment of the present invention includes employing as the gene a gene capable of encoding at least one of the materials which is selected from the group which includes (a) a human interleukin-l receptor antagonist protein or a biologically active derivative or fragment thereof, (b) a Lac Z marker gene capable of encoding a beta-galactosidase protein or a biologically active derivative or fragment thereof, (c) a soluble interleukin-l receptor protein or a biologically active derivative or fragment thereof, (d) a proteinase inhibitor, and (e) a cytokine, and employing as the DNA plasmid vector any DNA plasmid vector known to one of ordinary skill in the art capable of stable maintenance within the targeted cell or tissue upon delivery, regardless of the method of delivery utilized. One such method is the direct delivery of tne DNA vector molecule, whether it be a viral or plasmid DNA vector molecule, to the target cell or tissue. This method also includes employing as the gene a gene capable of encoding at least one of the materials selected from the group which includes (a) a human interleukin-l receptor antagonist protein or biologicallv 30 active derivative or fragment thereof, (b) a Lac Z marker gene capable of encoding a beta-galactosidase protein or biologically active derivative or fragment thereof, (c) a soluble interleukin-1 receptor protein or biologically active derivative or fragment thereof, (d) a p_oteinase inhibitor and WO94/20~17 PCT~S94/0~14 .

215~782 (e) a cytokine. In a specific method disclosed as an example, and not as a limitation to the present invention, a DNA plasmid vector containing the interleukin-1 beta coding sequence was ligated downstream of the cytomegalovirus (CMV) promoter. This DNA plasmid construction was encapsulated within liposomes and injected intra-articularly into the knee joints of recipient rabbits. Interleukin-1 beta was expressed and significant amounts of interleukin-1 beta was recovered from the synovial tissue. An alternative is injection of the naked plasmid DNA into the knee joint, allowing direct transfection of the DNA into the synovial tissue.
Another embodiment of this invention provides a method for introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host. This method includes employing non-viral means for introducing the gene encoding for the product into the connective tissue cell.
More specifically, this method includes employing non-viral means which is selected from at least one of the group which includes (a) at least one liposome, (b) Ca3 (P04) 2' (c) electroporation, and (d) DEAE-dextran, and includes employing as the gene a gene capable of encoding at least one of the materials selected from the group which includes (a) a human interleukin-1 receptor antagonist protein or biologically active derivative or fragment thereof, (b) a Lac Z marker gene capable of encoding a beta-galactosidase protein or biologically active derivative or fragment thereof, (c) a soluble interleukin-1 receptor protein or biologically active derivative or fragment thereof, (d) a proteinase inhibitor (e) a soluble tumor necrosis factor receptor protein or biologically active derivative or fragment thereof, and (f) a cytokine.
A further embodiment of this invention provides an additional method for introducing at least one gene encoding WO94/20517 PCT~S94/0~14 a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host. This additional method includes employing the biologic means of utilizing a virus to deliver the DNA vector molecule to the target cell or tissue. Preferably, the virus is a psuedovirus, the genome having been altered such that the psuedovirus is capable only of delivery and stable maintenance within the target cell; but not retaining an ability to replicate within the target cell or tissue. The altered viral genome is further manipulated by recombinant DNA techniques such that the viral genome acts as a DNA vector molecule which contains the heterologous gene of interest to be expressed within the target cell or tissue. This method also includes employing as the gene a gene capable of encoding at least one of the materials selected from the group which includes (a) a human interleukin-1 receptor antagonist protein or biologically active derivative or fragment thereof, (b) a Lac Z marker gene capable of encoding a beta-galactosidase protein or biologically active derivative or fragmen~ thereof, (c) a soluble interleukin-1 receptor protein or biologically active derivative or fragment thereof, (d) a proteinase inhibitor and (e) a cytokine.
A further embodiment of this invention includes a method to produce an animal model for the study of connective tissue pathology which includes introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host.
Another embodiment of this invention provides a method of using the gene encoding an extracellular interleukin-1 binding domain of the interleukin-1 receptor.
This gene is capable of binding to and neutralizing interleukin-1 in vivo to substantially resist the degradation of cartilage in a mammalian host. Unlike previous pharmacological efforts, the method of this invention employs WO94120~17 PCT~S94/0~14 21S~782 gene therapy in vivo to address the chronic debilitating effects of arthritis.
A preferred method of using the gene coding for the truncated interleukin-l receptor-of this invention involves employing recombinant t~hn;ques to generate a cell line which produces infectious retroviral particles containing the gene coding for the truncated interleukin-l receptor. The producer cell line is generated by inserting the gene coding into a retroviral vector under the regulation of a suitable eukaryotic promoter, transfecting the retroviral vector containing the gene coding into the retroviral packaging cell line for the production of a viral particle that is capable of expressing the gene coding for the truncated interleukin-l receptor, and infecting the synovial cells of a mammalian host using the viral particle.
More specifically, the method of using the hereinbefore described gene coding for the truncated interleukin-l receptor involves introducing the viral particles obtained from the retroviral packaging cell line directly by intra-articular injection into a joint space of a mammalian host that is lined with synovial cells. In a preferred embodiment, synoviocytes recovered from the knee joint are cultured in vitro for subsequent utilization as a delivery system for gene therapy. It will be apparent that Applicants are not limited to the use of the specific synovial tissue disclosed. It would be possible to utilize other tissue sources, such as skin cells, for in vitro culture tec-hn;ques. The method of using the gene of this invention may be employed both prophylactically and in the therapeutic treatment of arthritis. It will also be apparent that Applicants are not limited to prophylactic or therapeutic applications in treating only the knee joint. It would be possible to utilize the present invention either WO94/20517 PCT~S9410~14 .

21~7782 prophylactically or therapeutically to treat arthritis in any susceptible joint.
In another embodiment of this invention, a method of using the hereinbefore described gene coding for the truncated interleukin-l receptor involves infecting synovial cells in culture with the viral particles and subsequently transplanting the infected synovial cells back into the joint.
This method of using the gene of this invention may also be employed prophylactically and in the therapeutic treatment of arthritis in any area susceptible to the disorder.
In another embodiment of this invention, a method of using the gene coding for an extracellular interleukin-l binding domain of the interleukin-l receptor that is capable of binding to and neutralizing interleukin-l includes employing recombinant techniques to produce a retrovirus vector carrying two genes. The first gene encodes the extracellular interleukin-l binding domain of the interleukin receptor, and the second gene encodes for selectable antibiotic resistance. This method of use involves transfecting the retrovirus vector into a retrovirus packaging cell line to obtain a cell line producing infectious retroviral particles carrying the gene.
Another embodiment of this invention provides a method of preparing a gene encoding an extracellular interleukin-l binding domain of the interleukin-l receptor including synthesizing the gene by a polymerase chain reaction, introducing the amplified interleukin-l receptor coding sequence into a retroviral vector, transfecting the retroviral vector into a retrovirus packaging cell line and collecting viral particles from the retrovirus packaging cell line.
In another embodiment of this invention, a compound for parenteral administration to a patient in a therapeutically effective amount is provided for that contains WO94/20517 PCT~S94/0~14 2157~8~ --a gene encoding an extracellular interleukin-l binding domain of the interleukin-1 receptor and a suitable pharmaceutical carrier.
Another emkodiment of this invention provides for a compound for parenteral~a~; n; ~tration to a patient in a prophylactically effective amount that includes a gene encoding an extracellular interleukin-1 binding domain of the interleukin-l receptor and a suitable pharmaceutical carrier.
An additional embodiment of the invention involves transfection of hematopoietic progenitor cells or mature lymphoid or myeloid cells with a DNA vector molecule containing any of the gene or genes disclosed throughout the specification. The transfected cells are recovered and injected into the bone marrow of the patient using techni~ues known and available to one of ordinary skill in the art. It will be possible, within the scope of this method, to use cells derived from donor bone marrow instead of cells derived from recipient bone marrow so as to modify rejection.
In another embodiment of the invention, synoviocytes are transfected in vivo subsequent to direct intra-articular injection of a DNA molecule containing the gene of interest into the joint. Transfection of the recipient synovial cells bypasses the requirement of removal, culturing, in vitro transfection, selection and transplanting the DNA vector containing - synoviocytes (as disclosed in the Example section) to promote stable expression of the heterologous gene of interest. Methods of injecting the DNA molecule into the joint includes, but is not limited to, encapsulation of the DNA molecule into cationic liposomes or the direct injection of the DNA molecule itself into the joint. The DNA molecule, regardless of the form of presentation to the knee joint, is preferably presented as a DNA vector molecule, either as viral DNA vector molecule, or preferably, a DNA plasmid vector molecule. Expression of the heterologous gene of interest is WO94/20517 PCT~S94/0~14 ensured by inserting a promoter fragment act~ve in eukaryotic cells directly upstream of the coding region of the heterologous gene. = One of ordinary skill in the art may utilize known strategies and t~n;ques of vector construction to ensure appropriate levels of expression subsequent to entry of the DNA molecule into the synovial tissue.
It is an object of the present invention to provide a method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host.
It is an object of the invention to provide a method of i~troducing a gene encoding a product into at least one cell of a connective tissue of a mammalian host for a therapeutic use.
It is an object of the present invention to provide a method of introducing into the synovial lining cells of a mammalian arthritic joint at least one gene which codes for proteins having therapeutic properties.
It is an object of the present invention to provide an animal model for the study of connective tissue pathology.
It is an object of the present invention to provide a method of using in vivo a gene coding for the extracellular interleukin-1 binding domain of the interleukin-1 receptor that is capable of binding to and neutralizing subs'antially all isoforms of interleukin-1, including interleukin-1 alpha and interleukin-~ beta.
It is an object of the present invention to provide a method of using a gene in vivo in a mammalian host that is capable of binding to and neutralizing substantially all isoforms of interleukin-1 and thus, substantially resist t~e degradation of cartilage and protect surrounding soft tissues of the joint space.
It is an object of the present invention to provide a method of using ~n vivo a gene coding for the extracellular WO94/20517 PCT~S9410~14 21~7~8Z

interleukin-l binding domain of the interleukin-l receptor that is capable of binding to and neutralizing substantially all isoforms of interleukin-l for the prevention of arthritis in patients that demonstrate a high susceptibility for developing the disease.
It is an object of the present invention to provide a method of using in vivo a gene coding for an extracellular interleukin-l binding domain of an interleukin-l receptor that is capable of binding to and neutralizing substantially all isoforms of interleukin-l for the treatment of patients with arthritis.
It is an object of the present invention to provide a method of using in vivo a gene or genes that address the chronic debilitating pathophysiology of arthritis.
It is a further object of the present invention to provide a compound for parenteral administration to a patient which comprises a gene encoding an extracellular interleukin-l binding domain of the interleukin-l receptor and a suitable pharmaceutical carrier.
These and other objects of the invention will be more fully understood from the following description of the invention, the referenced drawings attached hereto and the claims appended hereto.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure l shows the structure of the cDNA encoding the human interleukin-l receptor antagonist protein (IRAP) gene inserted into the NcoI and BamHI cloning sites of the retroviral vector MFG.
Figure 2 shows the structure of the cDNA encoding the human interleukin-l receptor antagonist protein (IRAP) gene with a selectable neo marker inserted into the retroviral vector MFG.
Figure 3 shows a micrograph of synovium recovered from the knee of a rabbit approximately one month after WO94/20517 PCT~S94/0~14 2157782 ~ I

intra-articular injection of Lac Z+ neo synoviocytes employing the methods of this invention.
Figure 4 shows a Western blot demonstrating the production of interleukin-1 receptor antagonist protein by four cultures of HIG-82 cells (Georgescu 1988) infected using the method of this invention employing the MFG-IRAP viral vector.
Figure 5 shows data demonstrating the inhibition of chondrocytes by the addition of medium conditioned by MFG-IRAP
infected HIG-82 cells.
Figure 6 shows the uptake and expression of the Lac Z gene by synoviocytes using lipofection. Well 1 -Control cells, treated with liposomes alone; Well 2 - Control cells, treated with DNA alone; Well 3 - DNA + 150 nmole liposomes; Well 4 - DNA + 240 nmole liposomes; Well 5 -DNA + 300 nmole liposomes; Well 6 - DNA + 600 nmole liposomes.
Figure 7 shows the interleukin-1 binding domain amino acid arrangement.
Figures 8A-8C show the amino acid and nucleotide sequence of the human and mouse interleukin-1 receptors.
Figure 9 shows gene encoding a truncated interleukin-1 receptor inserted into a retroviral vector.
DETAILED DESCRIPTION OF THE INVENTION
As used herein, the term "patient" includes members of the animal kingdom including but not limited to human beings.
As used herein, the term "mammalian host" includes members of the animal kingdom including but not limited to human beings.
As used herein, the term "connective tissue"
includes but is not limited to a ligament, a cartilage, a tendon, and a synovium of a mammalian host.
-WO94/20517 . PCT~S94/0~14 ~,~5~782 As used herein, the term "DC-chol" means a cationic liposome con~; n; ng cationic cholesterol derivatives. The "DC-chol" molecule includes a tertiary amino group, a medium length spacer arm (tuo atoms) and a carbamyol linker bond as described in Biochem. Biophys. ~es. Commun., 179:280-285 (l99l), X. Gao and L. Huang.
As used herein, "SF-chol" is defined as a type of cationic liposome.
As used herein, the term "biologically active" used in relation to liposomes denotes the ability to introduce functional DNA and/or proteins into the target cell.
As used herein, the term "biologically active" in reference to a nucleic acid, protein, protein fragment or derivative thereof is defined as an ability of the nucleic acid or amino acid sequence to mimic a known biological function elicited by the wild type form of the nucleic acid or protein.
As used herein, the term "maintenance", when used in the context of liposome delivery, denotes the ability of the introduced DNA to remain present in the cell. When used in other contexts, it means the ability of targeted DNA to remain present in the targeted cell or tissue so as to impart a therapeutic effect.
Connective tissues are difficult organs to target therapeutically. Intravenous and oral routes of drug delivery that are known in the art provide poor access to these connective tissues and have the disadvantage of exposing the mammalian host body systemically to the therapeutic agent.
More specifically, known intra-articular injection of joints provides direct access to a joint. However, most of the injected drugs have a short intra-articular half-life. The present invention solves these probiems by introducing into the connective tissue of a mammalian host genes encoding for proteins that may be used to treat the mammalian host. More WO94/20S17 PCT~S94/0~14 21~7782 specifically, this invention provides a method for introducing into the connective tissue of a mammalian host genes encoding for proteins with anti-arthritic properties.
The present invention provides a method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host which comprises employing recombinant tec-hniques to produce a viral vector which contains the gene encoding for the product, and infecting the connective tissue cell of the mammalian host using the viral vector containing the gene coding or the product. This method preferably includes introducing the gene encoding the product into at least one cell of the connective tissue of the mammalian host for a therapeutic use.
In one embodiment of this invention, the method as hereinbefore described includes employing as the gene a gene capable of encoding a human interleukin-1 receptor antagonist protein (IRAP).
In another embodiment of this invention, the method as hereinbefore described includes employing as the gene a Lac Z marker gene capable of encoding a beta-galactosidase.
In another embodiment of this invention, the method as hereinbefore described includes employing as the gene a gene capable of encoding a soluble interleukin-1 receptor.
Another embodiment of this invention includes the method as hereinbefore described including employing as the gene a gene capable of encoding at least proteinase inhibitor.
More specifically, this method preferably includes employing a tissue inhibitor of a metalloproteinases as the proteinase inhibitor.
Another embodiment of this invention ir.cludes the method as hereinbefore described including employing as the gene a gene capable of encoding at least one cytokine. More specifically, this method includes employing as the cytokine 21~7782 at least one material selected from the group consisting of interleukin-1 alpha, interleukin-1 beta, interleukin-2, interleukin-3, interleukin-4, interleukin-5, interleukin-6, interleukin-7, interleukin-8, interleukin-9, interleukin-10, interleukin-11, interleukin-i2, tumor necrosis factor c~, and tumor necrosis factor B.
A further embodiment of this invention includes a method as hereinbefore described including employing as the cytokine at least one transforming growth factor. More specifically, this method includes employing as the trans-forming growth factor a growth factor selected from the group consisting of TGF-betal, TGF-beta2, TGF-beta3, and TGF-alpha.
Each transforming growth factor is commercially available from R & D Systems, 614 McKinley Place, N.E., Minneapolis, MN
55413.
In another embodiment of this invention, the method as hereinbefore described includes employing as the cytokine at least one fibroblast growth factor The fibroblast growth factors are also commercially available from R & D Systems, 614 McKinley Place, N.E., Minneapolis, MN 55413.
Another embodiment of this invention includes the method as hereinbefore described including employing as the viral vector a retroviral vector. More specifically, this method includes employing as the retroviral vector at least one material selected from the group consisting of MFG and BAG. A preferred embodiment of this invention includes providing the method as hereinbefore described including employing as the gene a gene capable of encoding a human interleukin-1 receptor antagonist protein and employing MFG as the retroviral vector.
Another preferred embodiment of this invention includes the method as hereinbefore described including employing a Lac Z marker gene as the gene capable of encoding WO94120517 PCT~S94/0~14 a beta-galactosidase and employing MFG as the retroviral vector.
Another preferred embodiment of this invention provides the method as hereinbefore described including employing a Lac Z neo marker gene as the gene capable of encoding a beta-galactosidase and employing BAG as the retroviral vector.
In a most preferred embo~;rent of this invention, the method as hereinbefore described includes employing a retroviral vector selected from the group consisting of MFG
and BAG and includes employing as the gene a gene capable of encoding a soluble interleukin-l receptor.
In another embodiment of this invention, a method as hereinbefore described is provided including employing as the gene a gene capable of encoding at least one proteinase inhibitor and including employing as the retroviral vector at least one material selected from the group consisting of MFG
and BAG.
In another embodiment of this invention, a method as hereinbefore described is provided which includes employing as the retroviral vector at least one material selected from the group consisting of MFG and BAG and including employing as the gene a gene capable of encoding at least one cytokine as hereinbefore described.
In another embodiment of this invention, a method is provided for introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host which compr ses employing recombinant techniques to produce a viral vector which contains the gene encoding for the product and infecting the connective tissue cell of the mammalian host using the viral vector containing the gene coding for the product, wherein the viral vector is at least one vector selected from the group consisting of an adeno--~ssociated virus, an WO94/20517 PCT~S94/0~14 2157~8% ,~

adenovirus, and a herpes virus, such as herpes simplex type-1 or herpes simplex type-2.
This method includes employing as the gene a gene capable of encoding at least one material selected from the group which includes (a) a human interleukin-1 receptor antagonist protein, (b) a soluble interleukin-1 receptor, (c) a Lac Z
marker gene capable of encoding a beta-galactosidase, (d) at least one proteinase inhibitor and (e) at least one cytokine.
More specifically, this method includes employing a tissue inhibitor of metalloproteinases as the proteinase inhibitor and includes employing as the cytokine at least one of the materials selected from the group which includes (a) at least one transforming growth factor selected from the group consisting of TGF-beta" TGF-beta2, TGF-beta3, and TGF-alpha, (b) at least one fibroblast growth factor, (c) interleukin-1 alpha, (d) interleukin-l beta, (e) interleukin-2, (f) interleukin-3, (g) interleukin-4, (h) interleukin-5, (i) interleukin-6, (j) interleukin-7, (k) interleukin-8, (l) interleukin-9, (m) interleukin-10, (n) interleukin-11, and (o) interleukin-12 (p) tumor necrosis factor a, and (q) tumor necrosis factor B.
Another embodiment of this invention includes the method as hereinbefore described including introducing the gene into a connective tissue which tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium. It is preferable that this method includes employing a cruciate ligament as the ligament. Most prefer-able this method includes employing as the cruciate ligament a ligament selected from the group consisting of an anterior cruciate ligament and a posterior cruciate ligament.
Another embodiment of this invention includes the method as hereinbefore described including employing as the gene a gene having DNA that is capable of maintenance and expression.

WO94/20517 PCT~S94/0~14 ~ 21S7782 A further embodiment of this inventior. includes the method as hereinbefore described including introducing the gene into the cell in vitro. This method includes subsequently transplanting the infected cell into the mammalian host. This method also includes after effecting the infecting of the connective tissue cell but before the transplanting of the infected cell into the mammalian host, storing the infected connective tissue cell. It will be appreciated by those skilled in the art that the infected connective tissue cell may be stored frozen in 10 percent DMSO
in liquid nitrogen. This method inc udes employing a method to substantially prevent the development of arthritis in a mammalian host having a high susceptibility of developing arthritis.
The method of this invention includes employing the method on an arthritic ~rAlian host for a therapeutic use.
This method includes employing a method to repair and regenerate the connective tissue whicn tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium. This method includes employing the method on a m~Alian host that is a human being.
Another embodiment of this invention includes a method of introducing at least one gene encoding a product into at least one cell of a cor.nective tissue of a mammalian host for use in treating the mammalian host as hereinbefore described including effecting in vivo the infection of the cell by introducing the viral vector containing the gene coding for the product directly into the mammalian host.
Pre~erably, this method includes effecting the direct introduction into the mammalian host by intra-articular injection. This method includes employing the method to substantially prevent a developmen' of arthritis in mammalian host having a high susceptibility of developing arthritis. This method also includes employing the method on WO94/20517 PCT~S94/0~14 2l~7~82 an arthritic mammalian host for therapeutic use. Further this method as includes employing the method to repair and regenerate the connective tissue a$ hereinbefore defined.
In yet another embo~ rnt of this invention, a method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host includes employing non-viral means for introducing the gene encoding for the product into the connective tissue cell. This method includes employing non-viral means selected from the group consisting of at least one liposome, Ca3(PO4)2, electroporation, and DEAE-dextran. This method includes employing as the liposome a material selected from the group consisting of DC-chol and SF-chol.
It will be understood that the method of this invention of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host that includes employing non-viral means for introducing the gene encoding for the product into the connective tissue cell is a non-infectious delivery system. An advantage of the use of a non-infectious delivery system is the elimination of insertional mutagenesis and virally induced disease.
It will be appreciated by those skilled in the art, that the viral vectors employing a liposome are not limited by cell division as is required for the retroviruses to effect infection and integration of connective tissue cells. This method employing non viral means as hereinbefore described includes employing as the gene a gene capable of encoding at least one of the following materials selected from the group which includes (a) a human interleukin-1 receptor antagonist protein, (b) a Lac Z marker gene capable of encoding a beta-galactosidase, (c) a soluble interleukin-l receptor, (d) at least one proteinase inhibitor, (e) at least one .

WO94/20517 PCT~S94/0~14 transforming growth factor, and (f) at least one cytokine.
More specifically, this method includes employing as the cytokine a cytokine selected from the group which includes interleukin-l alpha, interleukin-l beta, interleukin-2, interleukin-3, interleukin-4, interleukin-5, interleukin-6, interleukin-7, interleukin-8, interleukin-9, interleukin-lO, interleukin-ll, interleukin-12, tumor necrosis factor ~, tumor necrosis factor B, at least one fibroblast growth factor, and at least one transforming growth factor. Preferably, this method includes employing as the transforming growth factor a growth factor selected from the group consisting of TGF-betal, TGF-betal, TGF-beta3, and TGF-alpha.
Another preferred embodiment of this invention includes providing the method employing non-viral means as hereinbefore described which includes employing a tissue inhibitor of metalloproteinases as the proteinase inhibitor.
This method employing non-viral means for introducing the gene encoding for the product into the connective tissue cell as hereinbefore described includes introducing the gene into the connective tissue which tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium. Preferably, this method includes employing a cruciate ligament as the ligament. The cruciate ligament is selected from the group consisting of an anterior cruciate ligament and an posterior cruciate ligament.
Another embodiment of this invention provides the method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mamma;ian host for use in treating the mammalian host which includes employing non-viral means as hereinbefore described and includes employing as the gene a gene having DNA that is capable of maintenance and expression.
In yet a further embodiment of this invention, the method of introducing at least one gene encoding a product WO94/20517 PCT~S94/0~14 ~ls77s~ --into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host is provided that includes employing non-viral means for introducing the gene encoding for the product into the connective tissue cell n vitro and includes subsequently transplanting the cell having the gene into the mammalian host. Another embodiment of this invention provides a method including after introducing the gene encoding for the product in the connective tissue cell and before the transplanting of the connective tissue cell having the gene into the mammalian host, storing the connective tissue cell having the gene.
This method includes storing connective tissue cell frozen in lO percent DMS0 in liquid nitrogen. This method includes employing a method to substantially prevent the development of arthritis in a mammalian host having a high susceptibility of developing arthritis. Further, this method includes employing the method on an arthritic mammalian host for a therapeutic use. This method includes employing the method to repair and regenerate the connective tissue which tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium.
A further embodiment of this invention provides a method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host which includes employing non-viral means in vivo for directly introducing the gene encoding for the product into the connective tissue cell of the mammalian host. The non-viral means is selected from the group consisting of at least one liposome ~ ca3 (P04) 2 and DEAE-dextran. Preferably, this method includes effecting the in vivo introduction into the mammalian host by intra-articular injection. This method includes employing the method to substantiall~ prevent the development of arthritis in a mammalian host having a high susceptibility of developing W094120517 PCT~S94/0~14 21577~o~

arthritis. Further, this method includes employlng the method on an arthritic mammalian host for a therapeutic use. This method also includes employing the method to repair and regenerate the connective tissue which tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium.
Another embodiment of the present invention is a method to produce an animal model for the study of connective tissue pathology. As will be understood by those skilled in the art, over-expression of interleukin-l in the joint of a mammalian host is generally responsible for the induction of an arthritic condition. This invention provides a method for producing an animal model using the hereinbefore described gene transfer technology of this invention. Preferably, the method of this invention provides a method for producing an animal model using the hereinbefore described gene transfer technology of this invention to effect an animal model for arthritis. For example, constitutive expression of interleukin-l in the joint of a rabbit following the method of gene transfer provided for by this invention leads to the onset of an arthritic condition. It will be appreciated by those skilled in the art that this rabbit model is suitable for use for the testing of therapeutic agents. This method includes introducing at least one gene encoding a product into at least one cell of a connective tissue of a ~r~lian host comprising (a) employing reccmbinant techniques to produce a viral vector which contains the gene encoding for the product and (b) infecting the connective tissue cell of the ~rr~lian host using the viral vector containing the gene coding for the product for effecting the animal model. This method includes employing as the gene a material selected from the group consisting of a cytokine and a proteinase. This method includes employing as the cytokine a material selected from the group consisting of interleukin-l alpha, WO94120517 PCT~S94/0~14 2~5~782 interleukin-l be_a, and tumor necrosis factor-~ (TNF-~). This method includes employing as the proteinase a matrix metalloproteinase. The matrix metalloprGteinase is an enzyme selected from the group consisting of a collagenase, a gelatinase and a stromelysin. It will be apparent that use of the term "a collogenase, a gelatinase and a stromolysin" is meant to include the plural, and not be limited to the singular. It is well known in the art that numerous collagenases, gelatinases and stromolysins could be employed as a matrix metalloproteinase in the present invention. A
further embodiment of this invention provides a method to produce an animal model for the study of connective tissue pathology which includes employing non-viral means for introducing at least one gene encoding a product into at least lS one cell of a connective tissue of a mammalian host for effecting the animal model. The non-viral means is selected from the group consisting of at least one liposome, Ca3(PO4)2, electroporation, and DEAE-dextran. This method includes employing as the gene a material selected from the group consisting of a cytokine and a proteinase. This method includes employing as the cytokine a material selected from the group consisting of interleukin-l alpha, interleukin-l beta, and TNF-~. This method also includes employing as the proteinase a matrix metalloproteinase. The matrix metalloproteinase includes an enzyme selected from at least one of the group consisting of a collagenase, a gelatinase, and a stromelysin.
A further embodiment of the present invention includes employing as the gene a gene capable of encoding at least one of the materials which is selected from the group which includes (a~ a human interleukin-l receptor antagonist protein or a biologically active derivative or fragment thereof, (b) a Lac Z marker gene capable of encoding a beta-galactosidase protein or a biologically active derivative WO94/20517 PCT~S94/0~14 ~1~7782 or fragment thereof, (c) a soluble interleukin-l receptor protein or a biologically active derivative or fragment thereof, (d) a proteinase inhibitor, (e) a soluble tumor necrosis factor receptor protein or a biologically active derivative or fragment thereof and (f) a cytokine, and employing as the DNA vector any DNA vector, preferably a plasmid or viral vector, known to one of ordinary skill in the art capable of stable maintenance within the targeted cell or tissue upon delivery, regardless of the method of delivery utilized. In one embodiment of the invention, synoviocytes are transfected n vivo subsequent to direct intra-articular injection of a DNA molecule containing the gene of interest into the joint. Transfection of the recipient synovial cells bypasses the requirement of removai, culturing, in vitro transfection, selection and transplanting the DNA vector containing - synoviocytes (as disclosed in the Example section) to promote stable expression of the heterologous gene of interest. Methods of injecting the DNA molecule into the joint includes, but is not limited to, encapsulation of the DNA molecule into cationic liposomes or the direct injection of the DNA molecule itself into the joint. Expression of the heterologous gene of interest subsequent to n vivo transfection of the synovial tissue is ensured by inserting a promoter fragment active in eukaryotic cells directly upstream of the coding region of the heterologous gene. One of ordinary skill in the art may utilize known strategies and t~chn;ques of vector construction to ensure appropriate levels of expression subsequent to entry of the DNA molecule into the synovial tissue. As an example, and not a limitation, of the present invention, a DNA plasmid vector containing the interleukin-l beta coding sequence ligated downstream of the - CMV promoter was encapsulated within liposomes and injected into the knee joints of recipient rabbits. Interleukin-l beta was expressed in synovial tissue, as significant amounts of WO94/20517 PCT~S94/0~14 ~
21S~ ~ 82 interleukin-l beta was recovered from the synovial tissue within the region of intra-articular injection.
A further embodiment~ of this invention provides an additional method for introdùcing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating the mammalian host. This additional method includes employing the biologic means of utilizing a virus to deliver the DNA vector molecule to the target cell or tissue. Preferably, the virus is a psuedovirus, the genome having been altered such that the psuedovirus is capable only of delivery and stable maintenance within the target cell; but not retaining an ability to replicate within the target cell or tissue. The altered viral genome is further manipulated by recombinant DNA techniques such that the viral genome acts as a DNA vector molecule which contains the heterologous gene of interest to be expressed within the target cell or tissue. This method also includes employing as the gene a gene capable of encoding at least one of the materials selected from the group which includes (a) a human interleukin-1 receptor antagonist protein or biologically active derivative or fragment thereof, (b) a Lac Z marker gene capable of encoding a beta-galactosidase protein or biologically active derivative or fragment thereof, (c) a soluble interleukin-1 receptor protein or biologically active derivative or fragment thereof, (d) a proteinase inhibitor and (e) a soluble tumor necrosis factor receptor protein or a biologically active derivative or fragment thereof and ~f) a cytokine.
The following examples are offered by way of illustration of the present inventicn, and not by way of limitation.

WO94/20517 PCT~S94/0~14 ~IS~:~8~

EXAMPLE I
Packaginq of AAV
The only cis-acting sequences required for replication and packaging of recombinant adeno-associated virus (AAV) vector are the AAV terminal repeats. Up to 4 kb of DNA can be inserted between the terminal repeats without effecting viral replication or packaging. The virus rep proteins and viral capsid proteins are required in trans for virus replication as is an adeno-associated virus helper. To package a recombinant AAV vector, the plasmid containing the terminal repeats and the therapeutic gene is co-transfected into cells with a plasmid that expresses the rep and capsid proteins. The transfected cells are then infected with adeno-associated virus and virus isolated from the cells about 48-72 hours post-transfection. The supernatants are heated to about 56 Centigrade to inactivate the adeno-associated virus, leaving a pure virus stock of recombinant AAV.
EXAMPLE II
Electro~oration The connective tissue cells to be electroporated are placed into Hepes buffer saline (HBS) at a concentration of about 107 cells per ml. The DNA to be electroporated is added at a concentration of about 5-20 ug/ml of HBS. The mixture is placed into a cuvette and inse~ted into the cuvette holder that accompanies the Bio-RAD electroporation device (1414 Harbour Way South, Richmond, CA 94804). A range between about 250 and 300 volts at a capacitance of about 960 ufarads is required for introduction of DNA into most eukaryotic cell types. Once the DNA and the cells are inserted into the Bio-RAD holder, a button is pushed and the set voltage is delivered to the cell-DNA solution. The cells are removed ~ from the cuvette and replated on plastic dishes.

WOg4/20517 ~ PCT~S9~/0~14 Z1577~2 EXAMPLE III
The cDNA encoding the human interleukin-1 receptor antagonist (IRAP) was inserted into the NcoI and BamHI cloning sites of the retroviral véctor MFG as shown in Figure 1.
Specifically, a Pstl to Ba~HI fragmen' from the IRAP cDNA was linked to a synthetic oligonucleotide adapter from the NcoI
site (representing the start site of translation for IRAP) to the Pstl site (approximately 12 base pairs downstream from the NcoI site) to the MFG backbone digested at NcoI and BamHI in a three part ligation reaction. This three part ligation involving a synthetic oligo and two DNA fragments is well known by those skilled in the art of cloning. LTR means long terminal repeats, 5'SD means 5' splice donor, 3'SA means 3' splice acceptor. The straight arrow and the crooked arrow in Figure 1 represent unspliced and spliced messenger RNAs respectively. IRAP is encoded by the spliced message.
Figure 2 shows the cDNA encoding the human interleukin-1 receptor antagonist protein (IRAP) with a selectable neo gene marker. Figure 3 shows a low power micrograph of synovium recovered from the knee of a rabbit one month after intra-articular injection of Lac Z+, neo+ synoviocytes. Tissue was stained histochemically for the presence of beta galactosidase. This micrograph counterstained with eosin revealed an area of intensely stained, transplanted cells demonstrating that these cells have colonized the synovial lining of the recipient joint.
EXAMPLE IV
Animal Models The methods of this invention of transferring genes to the synovia of mammalian joints permit the production and analysis of joint pathologies that were not previously possible. This is because the only other way of delivering potentially arthriotogenic compounds to the joint is by intra-articular injection. Not only are such compounds WO94120517 PCT~S94/0~14 ~ 21~7782 quickly cleared from joints, but the effects of bolus injections of these compounds do not accurately mimic physiological conditions where they are constantly produced over a long period of time. In contrast, the gene transfer technologies of this invention permit selected proteins of known or suspected involvement in the arthritic process to be expressed intra-articularly over an extended period of time, such as for example, at least a three month period. The animal models of this invention therefore permits the importance of each gene product to the arthritic process to be evaluated individually. Candidate genes include, but are not restricted to, those coding ~or cytokines such as interleukin-l (IL-l) alpha, IL-l beta, and TNF-alpha, and matrix metalloproteinases such as collagenases, gelatinases and stromelysins.
Additionally, the gene transfer techniques of this invention are suitable for use in the screening of potentially therapeutic proteins. In this use, the animal models of the invention are initiated in joints whose synovia express gene coding for potential anti-arthritic proteins. Candidate proteins include, but are not restricted to, inhibitors of proteinases such as, for example, the tissue inhibitor of metalloproteinases, and cytokines such as, for example, trans-forming growth factor-beta.
EXAMPLE V
Method For Using Synoviocytes As A Delivery System For Gene TherapY
Rabbits are killed by intravenous injection of 4 ml nembutol, and their knees quickly shaved. Synovia are surgically removed from each knee under aseptic conditions !
and the cells removed from their surrounding matrix by - sequential digestion with trypsin and collagenase (0.2% w/v in Gey's Balanced Salt Solution) for about 30 minutes and about 2 hours, respectively. The cells recovered in this way are WO94/20517 PCT~S94/0~14 ~

215~8~

seeded into 25 cm2 culture flasks with about 4 ml of Ham's Fl2 nutrient medium supplemented with 10% fetal bovine serum, about 100 U/ml penicillin and about 100 ) g/ml streptomycin, and incubated at about 37 in an atmosphere of 95% air, 5% CO2.
Following about 3-4 days incubation, the cells attain confluence. At this stage, the culture medium is removed and the cell sheet washed twice with approximately 5 mls of Grey's Balanced Salt Solution to remove non-adherent cells such as lymphocytes. The adherent cells are then treated with ~rypsin (0.25% w/v in balanced salt solution). This treatment detaches the fibroblastic, Type B synoviocytes, but leaves macrophages, polymorphonuclear leukocytes and the Type A
synoviocytes attached to the culture vessel. The detached cells are recovered, re-seeded into 25 cm2 culture vessels at a 1:2 split ratio, medium is added and the culture returned to the incubator. At confluence this procedure is repeated.
After the third such passage, the cells are uniformly fibroblastic and comprise a homogeneous population of Type B synoviocytes. At this stage, cells are infected with the retroviral vector.
Following infection, cells are transferred to fresh nutrient medium supplemented with about 1 mg/ml G418 (GIBCO/BRL, P.O. Box 68, Grand Island, NY 14072-0068) and returned to the incubator. Medium is changed every three days as neo- cells die and the neo+ cells proliferate and attain confluency. When confluent, the cells are trypsinized and subcultured as described above. One flask is set aside for staining with X-gal to confirm that the neo+ cells are also Lac Z+. When the subcultures are confluent, the medium is recovered and tested for the presence of IRAP, soluble IL-lR or other appropriate gene products as hereinbefore described. Producing synoviocyte cultures are then ready for transplantation.

~ WO94/20517 PCT~S94/0~14 The day before transplantation, the cells are recovered by trypsinizing, as hereinbefore described. These cells are then suspended in nutrient medium, and incubated overnight in an untreated plastic centrifuge tube. Under these conditions, the cells do not adhere, bu. they regenerate their cell surface proteins that were removed by trypsinizing.
The following morning, the cells are recovered by centrifuging, washed several times by resuspension in Gey's Balanced Salt Solution and finally resuspended at a concentra-tion of about 106-107 cells/ml in Grey's solution.
Approximately 1 ml of this suspension is then introduced into the knee joint of a recipient rabbit by intra-articular injection. For this purpose a 1 ml syringe with a 25-gauge hypodermic needle is used. Injection is carried out through the patellar tendon. Experiments in which radioopaque dye was injected have confirmed that this method successfully intro-duces material into all parts of the joint.
EXAMPLE VI
The method of Example V for producing generally uniformly fibroblastic cells of a homogeneous population of Type B synoviocytes was followed to effect growing cultures of lapine synovial fibroblasts. These growing cultures of lapine synovial fibroblasts were subsequently infected with an amphotropic retroviral vector carrying marker genes coding for beta-galactosidase (Lac Z) and resistance to the neomycin analogue G418 (neo+). Following infection and growth in selective medium containing about 1 mg/ml G418, all cells stained positively in a histochemical stain for beta-galactosidase.
- 30 Neo selected cells carrying the Lac Z marker gene were transplanted back into the knees of recipient rabbits to examine the persistence and expression of these genes in vivo.
Two weeks following transplantation, islands of Lac Z+ cells within the synoviu~. of recipient knees were observed. This WO94/20517 PCT~S94/0~14 ~

~ 2~5~

confirmed the akility of the method of this invention to introduce marker genes into rabbit synovia and to express them n situ.
EXAMPLE VII
Neo-selected, Lac Z+ synoviocytes were recovered from cell culture, suspended in Gey's Balanced Salt Solution and injected intra-articularly into the knee joints of recipient rabbits (about lOs _ 107 cells per knee). Contra-lateral control knees received only a carrier solution. At intervals up to 3 months following transplant, the rabbits were killed and their synovia and surrounding capsule recovered. Each sample may be analyzed in three ways. A
third of the synovium was stained histochemically en masse for the presence of beta-galactosidase. A second portion may be lS used for immunocytochemistry using antibodies specific for bacterial beta-galactosidase. The final portion may be digested with trypsin and collagenase, and the cells thus recovered cultured in the presence of G418.
Staining of the bulk synovial tissue revealed extensive areas of Lac Z+ cells, visible to the naked eye.
Control synovia remained colorless. Histochemical examination of synovia revealed the presence of islands of cells staining intensely positive for beta-galactosidase. These cells were present on the superficial layer of the synovial lining, and were absent from control synovia. From such tissue it was possible to grow Lac Z+, neo+ cells. Cells recovered from control tissue were Lac Z~ and died when G418 was added to the culture. This indicates that the transplanted, transduced synovial fibroblasts have successfully recolonized the synovia of recipient joints, and continue to express the two marker genes, Lac Z and neo. Maintaining intra-articular Lac Z and neo expression in transplanted synoviocytes has been effected for 3 months using primary cells and one month using the HIG-82 cell line.

~ WO94/20517 PCT~S94/0~14 21~7782 EXAMPLE VIII
Based upon the methods of the hereinbefore presented examples, and employing stAn~rd recombinant techniques well known by those skilled in the art, the human IRAP gene was incorporated into an MFG vector as shown in Figure 1.
Following the infection of synoviocyte cultures of rabbit origin with this viral vector, IRAP was secreted into the culture medium.
Western blotting, well known by those skilled in the art, was carried out using an IRAP-specific rabbit polyclonai antibody that does not recognize human or rabbit IL-1 alpha or IL-1 beta, or rabbit IRAP. Figure 4 shows a Western blot which sets forth the production of IRAP by four cultures of HIG-82 cells infected with MFG-IRAP. Three forms of the IRAP
are present: a non-glycosylated form which runs with recombinant standards, and two larger glycosylated forms. The results of the Western blotting shown in Figure 4 demonstrated that IRAP was produced by HIG-82 synoviocyte cell line (Georgescu, 1988) following infection with the MFG-IRAP vector of this invention. The Western blotting of Figure 4 shows the IRAP concentration of the conditioned medium is as high as 50 ng/ml. This is approximately equal to 500 ng IRAP/106 cells/day. Lane 1 and Lane 2 of Figure 4 show that the recipient synovia tissue secrete substantial amounts of HIG-IRAP at 3 days (Lane 2) and 6 days (Lane 1). Lane 3 shows human recombinant IRAP. Lane 6 indicates that rabbit synovial cells produce a larger glycosylated version of this molecule after infection with MFG-IRAP. Lane 7 indicates that native rabbit synovial celis do not produce this glycosylated form.
Figure 5 shows that medium conditioned by IRAP+
synoviocytes blocks the induction of neutral metallopro-teinases in articular chondrocytes exposed to recombinant human IL-l beta. Chondrocytes normally secrete 1 U/106 cells, or less, gelatinase into their culture media. Figure 5 shows WO94/20517 PCT~S94/0~14 ~

P 215~2 that when to about 5 U/ml or lO U/ml IL-1 are added, gelatinase production increases to over 4 U and 6U/108 cells, respectively. Addition of medium conditioned by MFG-IRAP-infected HIG-82 cells employed by the method of this invention suppressed gelatinase production by IL-1 treated chondrocytes.
With 5 U/ml IL-l (Figure 5, right panel) inhibition was 100%
for one culture and 41% for the other. With 10 U/ml IL-l, inhibition was reduced to 38% and 18% (Figure 5, left panel) as is expected of a compeiitive inhibitor. These data demonstrate that the IRAP produced by HIG-82 cells infected with MFG-IRAP is biologically active.
EXAMPLE IX
This example demonstrates the uptake and expression of Lac Z gene by synoviocytes using infection by a liposome (lipofection). A six well plate containing synoviocyte cultures were transduced with the Lac Z gene by lipofection.
The content of each well is as follows:
Well 1 Control cells, treated with liposomes alone Well 2 Control cells, treated with DNA alone Well 3 DNA + 150 nmole liposomes Well 4 DNA + 240 nmole liposomes Well 5 DNA + 300 nmole liposomes Well 6 DNA + 600 nmole liposomes Wells 3-6 containing sub-confluent cultures of synovial fibroblasts were infected with 6 ug of DNA complexed with 150-600 nmoles/well of "DC-chol" liposome or in the alternative, with "SF-chol". Three days later, cells were stained histochemically for expression of beta-galactosidase (Figure 6).
Table 1 shows the results Gf using the liposomes "DC-chol" and "SF-chol" in converting synoviocyte cultures to the Lac Z+ phenotype without selection. Table 1 sets forth that the "DC-chol" liposome in a concentration of about 300 nmole/well converted generally 30% of the synovial cells in WO94/20517 PCT~S94/0~14 synoviocyte cultures to the Lac Z+ phenotype without selection. Reduced expression was shown in Well 6 for "DC-chol" due to the toxic effect of the high liposome concentration.

% Lac Z+ Cells Liposome, nmole/wellDC-chol SF-chol 150 10 0.5 240 22 1.0 300 30 2.8 600 NA 3.5 In another embodiment of this invention, a gene and method of using this gene provides for the neutralization of interleukin-l. Interleukin-l is a key mediator of cartilage destruction in arthritis. Interleukin-l also causes inflammation and is a very powerful inducer of bone resorption. Many of these effects result from the ability of interleukin-l to increase enormously the cellular synthesis of prostaglandin E2, the neutral proteinases -- collagenase, gelatinase, and stromelysin, and plasminogen activator. Th~
catabolic effects of interleukin-l upon cartilage are exacerbated by its ability to suppress the synthesis of the cartilaginous matrix by chondrocytes. Interleukin-l is present at high concentrations in synovial fluids aspirated from arthritic joints and it has been demonstrated that intra-articular injection of recombinant interleukin-l in animals causes cartilage breakdown and inflammation.
Interleukin-1 exists as several species, such as unglycosylated polypeptide of 17,000 Daltons. Two species have previously been cloned, interleukin-l alpha and WO94/20517 PCT~S94/0~14 ~

21S7~82 interleukin-l beta. The alpha form has a pI of approximately 5, and the beta form has a pI aro`und 7. Despite the existence of these isoforms, interleukin-l alpha and interleukin-1 beta have substantially identical biological properties and share common cell surface receptors. The type I interleukin-1 receptor is a 80kDa (kilodalton) glycoprotein and contains an extracellular, interleukin-l binding portion of 319 amino acids which are arranged in three immunoglobulin-like domains held together by disulfide bridges as shown in Figure 7. A 21 amino acid trans-membrane domain joins the extracellular portion to the 217 amino acid cytoplasmic domain. Figures 8A-8C show the amino acid and nucleotide sequence of the human and mouse interleukin-1 receptors. In Figure 8B, the 21 amino acid trans-membrane region of the interleukin-l receptor is marked by the thicker solid line. In Figures 8A and 8B, the position of the 5' and 3' oligonucleotides for PCR are marked by thinner short lines, respectively. The lysine amino acid just 5' to the trans-membrane domain to be mutated to a stop codon is marked by a solid circle in Figure 8B.
Synovium is by far the major, and perhaps the only, intra-articular source of interleukin-1 in the arthritic joint. Snyovia recovered from arthritic joints secrete high levels of interleukin-l. Both the resident synoviocytes and infiltrating blood mononuclear cells within the synovial lining produce interleukin-1.
The present invention provides a method of using in vivo a gene coding for a truncated form of the interleukin-1 receptor which retains its ability to bind interleukin-1 wi~h high affinity but which is released extracellularly and therefore inactive in signal transduction. The binding of this truncated and modified receptor to interleukin-1 inhibits the intra-articular activity of interleukin-1.
This method of using a gene encoding the extracellular interleukin-1 binding domain of an interleukin-1 WO94/20517 - PCT~S94/0~14 receptor that is capable of binding to and neutralizing interleukin-l includes employing a retroviral vector carrying a truncated interleukin-1 receptor gene which encodes a truncated and soluble active form of the receptor. The expression of the novel interleukin-1 receptor gene is controlled by regulatory se~l~nc~c contained within the vector that are active in eukaryotic cells. This recombinant viral vector is transfected into cell lines stably expressing the viral proteins in trans required for ~roduction of infectious virus particles carrying the recombinant vector. These viral particles are used to deliver the recombinant interleukin-1 receptor to the recipient synovial cells by direct virus infection in vivo.
The soluble human interleukin-1 receptor to be inserted into the retroviral vector may be generated by a polymerase chain reaction (PCR). An oligonucleotide complementary to the 5' leader sequence of the human interleukin-1 receptor (GCGGATCCCCTCCTAGAAGCT) and an oligonucleotide complementary to a region just upstream from the transmembrane domain of the interleukin-l receptor (GCGGATCCCATGTGCTACTGG) are used as primers for PCR. The primer for the region of the interleukin-1 receptor adjacent to the trans-membrane domain contains a single base change so that the lys codon at amino acid 319 (AAG) is changed to a stop codon (TAG). By inserting a translation stop codon just upstream from the transmembrane domain, a truncated form of interleukin-1 receptor that is secreted by the cell is generated. A BamHI recognition sequence (GGATCC) is added to the 5' end of the PCR primers, and following amplification, the resulting interleukin-1 receptor fragment is cloned into a BamHI site. A cDNA library from human T-cells is used as a source for the interleukin-1 receptor cDNA. To amplify the appropriate region of the interleukin-1 receptor from the cDNA
library, the complementary primers are added to the DNA and 50 WO94/20517 PCT~S94/0~14 ~
~S~2 cycles of annealing, primer extension and denaturation are performed using a thermocycler and standard PCR reaction conditions well known by tho~e~ persons skilled in the art.
Following amplification of t~e interleukin-1 soluble receptor using the PCR process, the resulting fragment is digested with BamHI and inserted into the pLJ retroviral vector. The pLJ
retroviral vector is available from A. J. Korman and R. C.
Mulligan. See also Proc. Natl. Acad. Sci., Vol. 84, pp. 2150-2154 (April 1987) co-authored by Alan J. Korman, J. Daniel Frantz, Jack L. Strominger and Richard C. Mulligan.
Restriction analysis was performed to determine the correct orientation of the insert.
The retrovirus vector carrying the truncated interleukin-1 receptor is transferred into the CRIP
(Proc. Natl. Acad. Sci., Vol. 85, pp. 6460-6464 (1988), 0. Danos and R. C. Mulligan) packaging cell line using a standard CaP04 transfection procedure and cells wherein the viral vector is stably integrated and is selected on the basis of resistance to the antibiotic G418. The viral vector cont~ining the neomycin resistant (neo-r) gene is capable of imparting resistance of the cell line to G418. The CRIP cell line expresses the three viral proteins required for packaging the vector viral RNAs into infectious particles. Moreover, the viral particles produced by the CRIP cell line are able to efficiently infect a wide variety of mammalian cell types including human cells. All retroviral particles produced by this cell line are defective for replication but retain the ability to stably integrate into synovial cells thereby becoming an heritable trait of these cells. Virus stocks produced by this method are substantially free of cont~in~ting helper-virus particles and are also non-pathogenic.
More specifically, the truncated interleukin-1 gene can be inserted into a retroviral vector under the regulation WO94120517 PCT~S94/0~14 of a suitable eukaryotic promoter such as the retroviral promoter already contained within the gene transfer vector, such as for example, the pLJ vector shown in Figure 9.
Figure 9 shows the structure of the pLJ interleukin receptor retroviral vector and partial restriction endonuclease map.
Reference numeral lO shows the interleukin-l receptor inserted into a retroviral vector. Reference numeral 12 indicates long terminal repeats (LTR's) at eac~ end of the structure of the pLJ interleukin receptor retroviral vector shown in Figure 8.
These LTR's regulate the viral transcription and expression of the interleukin-l receptor. Bacterial gene encoding resistance to the antibiotic neomycin (neo-r) is shown at reference numeral 16. The Simian Virus 40 enhancer promoter (SV 40) is indicated at reference numeral 18, and regulates the expression of the neo-r gene. Reference numbers 20 and 22, respectively, show the sites wherein the resulting interleukin receptor fragment is cloned. It will be understood by those persons skilled in the art that other vectors containing different eukaryotic promoters may also be utilized to obtain a generally maximal level of interleukin-l receptor expression. The vectors containing the truncated, and modified interleukin-l receptor will be introduced into a retroviral packaging cell line (CRIP) by transfection and stable transformants isolated by selection for the expression of the neomycin resistance gene also carried by the pLJ
vector. The CRIP cell line expresses all the proteins required for packaging of the exogenous retroviral RNA. Viral particles produced by the G418-selected CRIP cell lines wi l carry a recombinant retrovirus able to infect mammalian cells and stably express the interleukin-l truncated receptor. The viral particles are used to infect synovial cells directly in vivo by injecting the virus into the ~oint space.
Another embodiment of this invention provides a method for using the hereinbefore described viral particles to WO94/20517 PCT~S94/0~14 ~

2~S7~ 8~
-infect in culture synovial cells obtained from the lining of the joint of a mammalian host. The advantage of the infection of synovial cells in culture is that infected cells harboring the interleukin-l receptor retroviral construct can be 5 selected using G418 for expression of the neomycin resistance gene. The infected synovial c~lls expressing the interleukin-1 receptor can then be transplanted back into the joint by intra-articular injection. The transplanted cells will express high levels of soluble interleukin-1 receptor in the joint space thereby binding to and neutralizing substantially all isoforms of interleukin-l, including interleukin-1 alpha and interleukin-l beta.
The method used for transplantation of the synovial cells within the joint is a routine and relatively minor procedure used in the treatment of chronic inflammatory joint disease. Although synovium can be recovered from the joint during open surgery, it is now common to perform synovectomies, especially of the knee, through the arthroscope. The arthroscope is a small, hollow rod inserted into the knee via a small puncture wound. In addition to permitting the intra-articular insertion of a fibre-option system, the arthroscope allows access to surgical instruments, such that snyovial tissue can be removed arthroscopically.
Such procedures can be carried out under "spinal" anesthetic and the patient allowed home the same day. In this manner sufficient synovium can be obtained from patients who will receive this gene therapy.
The synovial cells (synoviocytes) contained within the excised tissue may be aseptically recovered by enzymic digestion of the connective tissue matrix. Generally, the synovium is cut into pieces of approximately 1 millimeter diameter and digested sequentially with trypsin (0.2% w/v in Grey's Balanced Salt Solution) for 30 minutes at 37 Centigrade, and collagenase (0.2% w/v in Grey's Balanced Salt .

WO94/20517 PCT~S94/0~14 2157~82 Solution) for 2 hours at 37 Centigrade. Cells recovered from this digestion are seeded into plastic culture dishes at a concentration of 104 - 105 cells per square centimeter with Hank's F12 medium supplemented with 10% fetal bovine serum and antibiotics After 3-7 days, the culture medium is withdrawn.
Non-adherent cells such as lymphocytes are removed by washing with Gey's Balanced Salt Solution and fresh medium added. The adherent cells can now be used as they are, allowed to grow to confluency or taken through one or more subcultures.
Subcultivating expands the cell number and removes non-dividing cells such as macrophages.
Following genetic manipulation of the cells thus recovered, they can be removed from the culture dish by trypsinizing, scraping or other m~ans, and made into a st~n~rd suspension. Gey's Balanced Salt Solution or other isotonic salt solutions of suitable composition, or saline solution are suitable carriers. A suspension of cells can then be injected into the recipient mammalian joint.
Intra-articular injections of this type are routine and easily carried out in the doctor's office. No surgery is necessary.
Very large numbers of cells can be introduced in this way and repeat injections carried out as needed.
Another embodiment of this invention is the gene produced by the hereinbefore described method of preparation.
This gene carried by the retrovirus may be incorporated in a suitable pharmaceutical carrier, such as for example, buffered physiologic saline, for parenteral administration. This gene may be administered to a patient in a therapeutically effective dose. More specifically, this gene may be incorporated in a suitable pharmaceutical carrier at a therapeutically effective dose and administered by intra-articular injection.
In another embodiment of this invention, this gene may be administered to patients as a prophylactic measure to WO94/20517 PCT~S94/0~14 ~

215~78~

prevent the development of arthritis in those patients determined to be highly susceptible of developing this disease. More speçifically, this gene carried by the retrovirus may be incorporated in a suitable pharmaceutical carrier at a prophylactically effective dose and administered by parenteral injection, including intra-articular injection.
EXAMPLE X
Fifty mi~L Gy r ams of a DNA plasmid vector molecule containing the interleukin-l beta coding sequence ligated downstream of the CMV promoter was encapsulated within cationic liposomes, mixed with Geys biological buffer and injected intra-articularly into the knee joints of a rabbit.
Fourty eight hours subsequent to injection one nanogram of interleukin-1 beta was recovered from the knee joint area.
Therefore, injection of the DNA cont~i n; ng liposome solution within the region of the synovial tissue prompted fusion of the liposomes to the synovial cells, transfer of the DNA
plasmid vector into synovial cells and subsequent expression of the IL-1 beta gene. Additionally, it is possible to inject non-encapsulated (i.e., naked) DNA into the joint area and monitor transfection of the DNA vector into the synovial cells as determined by subsequent expression of the IL-1 beta gene in synovial cells. Therefore, either method may be utilized as a plausible alternative to the n vitro manipulation of synovia also exemplified in the present invention.
It will be appreciated by those skilled in the art that this invention provides a method of introducing into a connective tissue cell of a mammalian host n vitro, or in the alternative in vivo, at least one gene which codes for proteins with therapeutic properties. This method includes employing genes having DNA that is capable of maintenance and expression.
It will be appreciated by those skilled in the art that this invention provides a method of introducing at least WO94/20517 215 7 7 8 2 PCT~S94/0~14 one gene encoding a product into at least one cell of the connective tissue of a mammalian host for treating an arthritic condition of the mammalian host.
It will be understood by those skilled in the art that this invention provides a method to repair and regenerate the connective tissue of a mammalian host.
It will be further understood that this invention provides a method to produce an animal model for the study of connective tissue pathology.
It will be appreciated by those persons skilled in the art that this invention provides a method of using and a method of preparing a gene encoding an extra cellular interleukin-l binding domain of an interleukin-l receptor that is capable of binding to and neutralizing substantially all isoforms of interleukin-l, and thus substantially protect cartilage of a mammalian host from pathological degradation.
In addition, it will be understood by those persons skilled in the art that the method of using the gene of this invention will reduce inflammation, protect soft tissues of the joint and suppress the loss of bone that occurs in patients suffering with arthritis.
It will be appreciated by those persons skilled in the art that the viral vectors employed in the hereinbefore described invention may be employed to transfect synovial cells in vivo or in culture, such as by direct intra-articular injection or transplantation of autologous synovial cells from the patient transduced with the retroviral vector carrying the truncated interleukin-l receptor gene.
Whereas particular embodiments of this invention have been described above for purposes of illustration, it will be evident to those persons skilled in the art that numerous variations of the details of the present invention may be made without departing from the invention as defined in the appended claims.
-WO94/20517 PCT~S94/0~14 2iS~2 SEQUENCE LISTING
(1) GENERAL INFORMATION: ~
(i) APPLICANT: University of Pittsburgh of the Commonwealth System of Higher Education (ii) TITLE OF INVENTION: Gene Transfer For Treating a Connective Tissue of a Mammalian Host (iii) NUMBER OF SEQUENCES: 6 (iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Eckert Seamans Cherin ~ Mellott (B) STREET: 1700 Market Street Suite 3232 (C) CITY: Philadelphia (D) STATE: PA
(E) COUNTRY: USA
(F) ZIP: 19103 (v) COM~Ul~ READABLE FORM:
(A) MEDIUM TYPE: Floppy disk (B) CO~U'l'~K: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Release #1.0, Version ~1.25 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Gould, Jr., Lewis F.
(B) REGISTRATION NUMBER: 25,057 (C) REFERENCE/DOCKET NUMBER: 109070-9 (ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (215) 575-6000 (B) TELEFAX: (215) 575-6015 (C) TELEX: 866172 (2) INFORMATION FOR SEQ ID NO:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1770 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: double (D) TOPOLOGY: linear WO94/20517 . PCT~S94/0~14 ~ 2I57782 (ii) MOLECULE TYPE: cDNA
(iii) HYPG~ CAL: NO
- (iv) ANTI-SENSE: NO

(vii) IMMEDIATE SOURCE:
(A) LIBRARY: Human T-cell cDNA Library (B) CLONE: Human Interleukin-1 Receptor (ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 55..1764 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:
C~lC~lGAGA AGCTGGACCC CTTGGTAAAA GACAAGGCCT TCTC~AA~-AA GAAT ATG 57 Met AAA GTG TTA CTC AGA CTT ATT TGT TTC ATA GCT CTA CTG ATT TCT TCT lOS
Ly~ Val Leu Leu Arg Leu Ile Cy~ Phe Ile Ala Leu Leu Ile Ser Ser 5 lO 15 Leu Glu Ala Asp Lys Cys Ly~ Glu Arg Glu Glu Lys Ile Ile Leu Val Ser Ser Ala A~n Glu Ile Asp Val Arg Pro Cys Pro Leu Asn Pro Asn Glu His Lys Gly Thr Ile Thr Trp Tyr Lys A~p Asp Ser Lys Thr Pro Val Ser Thr Glu Gln Ala Ser Arg Ile Hi~ Gln Hi~ LYB Glu LYB Leu Trp Phe Val Pro Ala Lys Val Glu A8p Ser Gly Hi~ Tyr Tyr Cys Val Val Arg Asn Ser Ser Tyr CYB Leu Arg Ile Lys Ile Ser Ala Lys Phe Val Glu A~n Glu Pro Asn Leu Cy8 Tyr A~n Ala Gln Ala Ile Phe Lys Gln Lys Leu Pro Val Ala Gly Asp Gly Gly Leu Val Cys Pro Tyr Met -WO 94120517 . PCT/US94/02414 2~1$~

GAG TTT TTT AAA AAT GAA A~T AAT GAG TTA CCT AaA TTA CAG TGG TAT 537 Glu Phe Phe Lys Asn Glu A~n A~n Glu Leu Pro Ly~ Leu Gln Trp Tyr Lys Asp Cy~ Ly~ Pro Leu Leu Leu Asp Asn Ile Hi~ Phe Ser Gly Val Lys Asp Arg Leu Ile Val Met Asn Val Ala Glu Ly~ His Arg Gly Asn Tyr Thr Cys His Ala Ser Tyr Thr Tyr Leu Gly Lys Gln Tyr Pro Ile Thr Arg V~l Ile Glu Phe Ile Thr Leu Glu Glu Asn Ly~ Pro Thr Arg Pro Val Ile Val Ser Pro Ala Asn Glu Thr Met Glu Val Asp Leu Gly Ser Gln Ile Gln Leu Ile Cys Asn Val Thr Gly Gln Leu Ser Asp Ile Ala Tyr Trp Ly~ Trp Asn Gly Ser Val Ile Asp Glu Asp Asp Pro Val Leu Gly Glu Asp Tyr Tyr Ser Val Glu Asn Pro Ala Asn Ly~ Arg Arg AGT ACC CTC ATC ACA GTG CTT AAT ATA TCG GA~ ATT GAA AGT AGA TTT 969 Ser Thr Leu Ile Thr Val Leu Asn Ile Ser Glu Ile Glu Ser Arg Phe Tyr Lys His Pro Phe Thr Cys Phe Ala Lys Asn Thr His Gly Ile Asp Ala Ala Tyr Ile Gln Leu Ile Tyr Pro Val Thr Asn Phe Gln Ly~ His Met Ile Gly Ile Cys Val Thr Leu Thr Val Ile Ile Val Cys Ser Val TTC ATC TAT AAA ATC TTC A~G ATT GAC ATT GTG CTT TGG TAC AGG GAT 1161 Phe Ile Tyr Lys Ile Phe Lys Ile Asp Ile Val Leu Trp Tyr Arg Asp Ser Cys Tyr Asp Phe Leu Pro Ile Lys Ala Ser Asp Gly Ly~ Thr Tyr -WO94120517 PCT~S94/0~14 ~ 2157782 A~p Ala Tyr Ile Leu Tyr Pro Lys Thr Val Gly Glu Gly Ser Thr Ser GAC TGT GAT ATT ~TT GTG TTT AAA GTC TTG CCT GAG GTC TTG GAA AAA 1305 Agp Cy8 Asp Ile Phe Val Phe Lys Val Leu Pro Glu Val Leu Glu Lys Gln Cy~ Gly Tyr Lys Leu Phe Ile Tyr Gly Arg Asp Asp Tyr Val Gly Glu Asp Ile Val Glu Val Ile Asn Glu A~n Val Ly~ Ly~ Ser Arg Arg Leu Ile Ile Ile Leu Val Arg Glu Thr Ser Gly Phe Ser Trp Leu Gly Gly Ser Ser Glu Glu Gln Ile Ala Met Tyr A~n Ala Leu Val Gln Asp Gly Ile Lys Val Val Leu Leu Glu Leu Glu Ly~ Ile Gln A~p Tyr Glu Ly~ Met Pro Glu Ser Ile Ly~ Phe Ile Ly~ Gln Lys His Gly Ala Ile Arg Trp Ser Gly Asp Phe Thr Gln Gly Pro Gln Ser Ala Lys Thr Arg Phe Trp Lys A~n Val Arg Tyr His Met Pro Val Gln Arg Arg Ser Pro Ser Ser Ly~ His Gln Leu Leu Ser Pro Ala Thr Ly~ Glu Ly~ Leu Gln Arg Glu Ala His Val Pro Leu Gly (2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 569 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:

WO 94/20517 PCT/US94/02414 ~

~,~5~

-- 52 ~
Met Lys Val Leu Leu Arg Leu Ile Cys Phe Ile Ala Leu Leu Ile Ser .10 15 Ser Leu Glu Ala A~p Lys Cys L-ys Glu Arg Glu Glu Lys I le I le Leu Val Ser Ser Ala Asn Glu Ile Asp Val Arg Pro CYD Pro Leu Asn Pro Asn Glu His Lys Gly Thr Ile Thr Trp Tyr Lys Asp Asp Ser Ly~ Thr Pro Val Ser Thr Glu Gln Ala Ser Arg Ile His Gln His Lys Glu Lys Leu Trp Phe Val Pro Ala Lys Val Glu Asp Ser Gly His Tyr Tyr Cys Val Val Arg Asn Ser Ser Tyr Cys Leu Arg Ile Lys Ile Ser Ala Lys Phe Val Glu Asn Glu Pro Asn Leu Cys Tyr Asn Ala Gln Ala I le Phe Lys Gln Lys Leu Pro Val Ala Gly Asp Gly Gly Leu Val Cys Pro Tyr Met Glu Phe Phe Lys Asn Glu Asn Asn Glu Leu Pro Lys Leu Gln Trp Tyr Lys Asp Cys Lys Pro Leu Leu Leu A~p Asn Ile His Phe Ser Gly Val Lys Asp Arg Leu Ile Val Met Asn Val Ala Glu Ly~ His Arg Gly Asn Tyr Thr Cys His Ala Ser Tyr Thr Tyr Leu Gly Lys Gln Tyr Pro Ile Thr Arg Val Ile Glu Phe Ile Thr Leu Glu Glu Asn Lys Pro Thr Arg Pro Val Ile Val Ser Pro Ala Asn Glu Thr Met Glu Val Asp Leu Gly Ser Gln Ile Gln Leu Ile Cys Asn Val Thr Gly Gln Leu Ser Asp Ile Ala Tyr Trp Lys Trp Asn Gly Ser Val Ile Asp Glu Asp Asp Pro Val Leu Gly Glu Asp Tyr Tyr Ser Val Glu Asn Pro Ala Asn Lys Arg Arg Ser Thr Leu Ile Thr Val Leu Afin Ile Ser Glu Ile Glu Ser Arg Phe Tyr Lys His Pro Phe Thr Cys Phe Ala Lys Asn Thr His Gly Ile Asp Ala Ala Tyr Ile Gln Leu Ile Tyr Pro Val Thr Asn Phe Gln Lys W094n0517 . PCT~S94/0~14 His Met Ile Gly Ile Cy~ Val Thr Leu Thr Val Ile Ile Val Cys Ser Val Phe Ile Tyr Lys Ile Phe Ly~ Ile ARP Ile Val Leu Trp Tyr Arg Asp Ser Cy~ Tyr Asp Phe Leu Pro Ile Ly~ Ala Ser Asp Gly Lys Thr Tyr A~p Ala Tyr Ile Leu Tyr Pro Lys Thr Val Gly Glu Gly Ser Thr Ser Asp Cys Asp Ile Phe Val Phe Lys Val Leu Pro Glu Val Leu Glu 405 410 . 415 Lys Gln Cys Gly Tyr Lys Leu Phe Ile Tyr Gly Arg Asp Asp Tyr Val Gly Glu Asp Ile Val Glu Val Ile Asn Glu Asn Val Lys Lys Ser Arg Arg Leu Ile Ile Ile Leu Val Arg Glu Thr Ser Gly Phe Ser Trp Leu Gly Giy Ser Ser Glu Glu Gln Ile Ala Met Tyr A~n Ala Leu Val Gln Asp Gly Ile Ly~ Val Val Leu Leu Glu Leu Glu Lys Ile Gln Asp Tyr Glu Ly~ Met Pro Glu Ser Ile Ly~ Phe Ile Lys Gln Lys His Gly Ala Ile Arg Trp Ser Gly A~p Phe Thr Gln Gly Pro Gln Ser Ala Lys Thr Arg Phe Trp Lys Asn Val Arg Tyr His Met Pro Val Gln Arg Arg Ser Pro Ser Ser Lys His C-ln Leu Leu Ser Pro Ala Thr Lys Glu Ly~ Leu Gln Arg Glu Ala His Val Pro Leu Gly (2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 178~ base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: double (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA

(iii) HYPOTHETICAL: NO

WOg4/20517 . PCT~S94/0~14 ~

2~,S~Ss~ ' ' (iv) ANTI-SENSE: NO

(vii) IMMEDIATE SOURCE:
(A) LIBRARY: Mouse T-cell cDNA Library (B) CLONE: ~ouse Interleukin-1 Receptor (ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 46..1776 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
GGATGTCATC AGAGTTCCCA GTGCCCC~-~A CCGTGAACAA CACAA ATG GAG AAT 54 Met Glu Asn Met Lys Val Leu Leu Gly Leu Ile Cys Leu Met Val Pro Leu Leu Ser Leu Glu Ile Asp Val Cys Thr Glu Tyr Pro Asn Gln Ile Val Leu Phe Leu Ser Val Asn Glu Ile Asp Ile Arg Lys Cys Pro Leu Thr Pro Asn LYB Met His Gly Asp Thr ILe Ile Trp Tyr Ly~ Asn Asp Ser Lys Thr Pro Ile Ser Ala Asp Arg Asp Ser Arg Ile His Gln Gln Asn Glu His Leu Trp Phe Val Pro Ala Ly~ Val Glu Asp Ser Gly Tyr Tyr Tyr Cys Ile Val Arg Asn Ser Thr Tyr Cys Leu Lys Thr Lys Val Thr Val Thr GTG TTA GAG A~T GAC CCT GGC TTG TGT TAC AGC ACA CAG GCC ACC TTC 438 Val Leu Glu Asn Asp Pro Gly Leu Cys Tyr Ser Thr Gln Ala Thr Phe Pro Gln Arg Leu His Ile Ala Gly Asp Gly Ser Leu Val Cys Pro Tyr Val Ser Tyr Phe Lys Asp Glu Asn Asn Glu Leu Pro Glu Val Gln Trp -- 2157~82 -- 55 ~
Tyr LYB A~n Cy~ Lys Pro Leu Leu Leu A~p Ann Val Ser Phe Phe Gly GTA AAA GAT AP~A CTG TTG GTG AGG AAT GTG GCT GAA GAG CAC AGA GGG 630 Val LYB ABP LYB Leu Leu Val Arg Asn Val Ala Glu Glu His Arq Gly Asp Tyr Ile Cys Arg Met Ser Tyr Thr Phe Arg Gly Lys Gln Tyr Pro Val Thr Arg Val Ile Gln Phe Ile Thr Ile ABP Glu AYn Lys Arg Asp AGA CCT GTT ATC CTG AGC CCT CGG AAT GAG ACG ATC GAA GCT GAC CCA 7?4 Arg Pro Val Ile Leu Ser Pro Arg Asn Glu Thr Ile Glu Ala Asp Pro Gly Ser Met Ile Gln Leu Ile Cy8 Asn Val Thr Gly Gln Phe Ser Asp Leu Val Tyr Trp Ly~ Trp Asn Gly Ser Glu Ile Glu Trp Asn Asp Pro Phe Leu Ala Glu Asp Tyr Gln Phe Val Glu His Pro Ser Thr Lys Arg Lys Tyr Thr Leu Ile Thr Thr Leu Asn Ile Ser Glu Val Lys Ser Gln Phe l'yr Arg Tyr Pro Phe I le Cys Val Val Lys Asn Thr Asn I le Phe Glu Ser Ala His Val Gln Leu Ile Tyr Pro Val Pro Asp Phe Ly~ Asn Tyr Leu Ile Gly Gly Phe Ile Ile Leu Thr Ala Thr Ile Val Cy~ Cys GTG TGC ATC TAT AP~A GTC TTC AAG GTT GAC ATA GTG CTT TGG TAC AGG 1158 Val Cy~ Ile Tyr Lys Val Phe LYB Val Asp Ile Val Leu Trp Tyr Arg Asp Ser Cys Ser Gly Phe Leu Pro Ser Lys Ala Ser Asp Gly Lys Thr Tyr Asp Ala Tyr Ile Leu Tyr Pro Lys Thr Leu Gly Glu Gly Ser Phe TCA GAC TTA GAT ACT TTT GTT TTT A~A CT-', TTG CCT GAG GTC TTG GAG 1302 Ser Asp Leu Asp Thr Phe Val Phe Lys Leu Leu Pro Glu Val Leu Glu W O 94120517 PCTruS94/024l4 ~

2ls~8~

GGA CAG TTT GGA TAC AAG CTG TTC ATT~TAT GGA AGG GAT GAC TAT GTT 1350 Gly Gln Phe Gly Tyr Ly~ Leu Phe Iie Tyr Gly Arg Asp Asp Tyr Val GGA GAA GAT ACC ATC GAG GTT ACT AAT GAA AAT GTA AAG A~A AGC AGG 1398 Gly Glu A~p Thr Ile Glu Val Thr Asn Glu A~n Val Lys Ly~ Ser Arg Arg Leu Ile Ile Ile Leu Val Arg A~p Met Gly Gly Phe Ser Trp Leu Gly Gln Ser Ser Glu Glu ~ln Ile Ala Ile Tyr Asn Ala Leu Ile Gln GAA GGA ATT AAA ATC GTC CTG CTT GAG TTG GAG A~A ATC CAA GAC TAT 1542 Glu Gly Ile Lys Ile Val Leu Leu Glu Leu Glu Ly~ Ile Gln A~p Tyr Glu Lys Met Pro Asp Ser Ile Gln Phe Ile Lys Gln Lys His Gly Val Ile Cy~ Trp Ser Gly Asp Phe Gln Glu Arg Pro Gln Ser Ala Lys Thr Arg Phe Trp Ly~ Asn Leu Arg Tyr Gln Met Pro Ala Gln Arg Arg Ser Pro Leu Ser Ly~ His Arg Leu Leu Thr Leu Asp Pro Val Arg A~p Thr AAG GAG A~A CTG CCG GCA GCA ACA CAC TTA CCA CTC GGC TAGCATGGC 1782 Lys Glu Lys Leu Pro Ala Ala Thr His Leu Pro Leu Gly (2) INFORMATION FOR SEQ ID NO:4:
(i) SEQ'JENCE CHARACTERISTICS:
(A) LENGTH: 576 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
Met Glu Asn Met Lys Val Leu Leu Gly Leu Ile Cys Leu Met Val Pro Leu Leu Ser Leu Glu Ile Asp Val Cy8 Thr Glu Tyr Pro Asn Gln Ile WO 94/20517 . PCT/US94/02414 Val Leu Phe Leu Ser Val Asn Glu Ile Asp Ile Arg Lys Cy5 Pro Leu Thr Pro Asn Lys Met His Gly Asp Thr Ile Ile Trp Tyr Lys A~n A~p Ser Lys Thr Pro Ile Ser Ala Asp Arq Asp Ser Arg Ile His Gln Gln A~n Glu His Leu Trp Phe Val Pro Ala Ly~ Val Glu Asp Ser Gly Tyr Tyr Tyr Cys Ile Val Arg Asn Ser Thr Tyr Cy8 Leu Lys Thr Lys Val Thr Val Thr Val Leu Glu Asn Asp Pro Gly Leu Cys Tyr Ser Thr Gln Ala Thr Phe Pro Gln Arg Leu His Ile Ala Gly Asp Gly Ser Leu Val Cys Pro Tyr Val Ser Tyr Phe Lys Asp Glu Asn Asn Glu Leu Pro Glu Val Gln Trp Tyr Lys Asn Cys Lys Pro Leu Leu Leu Asp Asn Val Ser Phe Phe Gly Val Lys Asp Lys Leu Leu Val Arg Asn Val Ala Glu Glu Hi~- Arg Gly Asp Tyr Ile Cys Arg Met Ser Tyr Thr Phe Arg Gly Lys Gln Tyr Pro Val Thr Arg Val Ile Gln Phe Ile Thr Ile A~p Glu Asn Lys Arg Asp Arg Pro Val Ile Leu Ser Pro Arg Asn Glu Thr Ile Glu Ala Asp Pro Gly Ser Met Ile Gln Leu Ile Cy5 Asn Val Thr Gly Gln Phe Ser Asp Leu Val Tyr Trp Lys Trp Asn Gly Ser Glu Ile Glu Trp Asn Asp Pro Phe Leu Ala Glu Asp Tyr Gln Phe Val Glu His Pro Ser Thr Lys Arg Ly~ Tyr Thr Leu Ile Thr Thr Leu Asn Ile Ser Glu Val Lys Ser Gln Phe Tyr Arg Tyr Pro Phe Ile Cys Val Val Lys Asn Thr Asn Ile Phe Glu Ser Ala His Val Gln Leu Ile Tyr Pro Val Pro Asp Phe Lys Asn Tyr Leu Ile Gly Gly Phe Ile Ile Leu Thr Ala Thr Ile Val Cys Cys Val Cys Ile Tyr Lys Val Phe Ly~ Val Asp Ile Val Leu WO94/20517 ~ ~ PCT~S94/0~14 Trp Tyr Arg A~p Ser Cy~ Ser Gly Phe Leu P~o Ser Ly~ Ala Ser Asp Gly Ly~ Thr Tyr Anp Ala Tyr Il~ ~eu Tyr Pro Lys Thr Leu Gly Glu Gly Ser Phe Ser A~p Leu A3p Thr Phe Val Phe Ly~ Leu Leu Pro Glu Val Leu Glu Gly Gln Phe Gly Tyr Lys Leu Phe Ile Tyr Gly Arg Asp Asp Tyr Val Gly Glu Asp Thr Ile Glu Val Thr Asn Glu A6n Val Lys Ly~ Ser Arg Arg Leu Ile Ile Ile Leu Val Arg Asp Met Gly Gly Phe Ser Trp Leu Gly Gln Ser Ser Glu Glu Gln Ile Ala Ile Tyr Asn Ala Leu Ile Gln Glu Gly Ile Ly~ Ile Val Leu Leu Glu Leu Glu Lys Ile Gln A~p Tyr Glu Lys Met Pro Asp Ser Ile Gln Phe Ile Lys Gln Lys Hi~ Gly Val Ile Cys Trp Ser Gly A6p Phe Gln Glu Arg Pro Gln Ser Ala Ly~ Thr Arq Phe Trp Ly~ A~n Leu Arg Tyr Gln Met Pro Ala Gln Arg Arg Ser Pro Leu Ser Lys His Arg Leu Leu Thr Leu Asp Pro Val Arg Asp Thr Lys Glu Lys Leu Pro Ala Ala Thr His Leu Pro Leu Gly (2) INFORMATION FO~ SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 22 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: Primer oligonucleotide (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO

(vii) IMMEDIATE SOURCE:

WO94120517 PCT~Ss4/0~14 21577$2 (B) CLONE: Primer Oligonuleotide to 5' Leader Sequence of IL-~ Receptor (xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:

(2) INFORMATION FOR SEQ ID NO:6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 2l base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY. linear (ii) MOLECULE TYPE: Primer oligonucleotide (iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO

(vii) IMMEDIATE SOURCE:
(B) CLONE: Primer Oligonucleotide Upstream of Transmembrane Portion of IL-l Receptor (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:

Claims (130)

WE CLAIM:
1. A method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating said mammalian host which comprises:
employing recombinant techniques to produce a viral vector which contains said gene encoding for said product; and infecting said connective tissue cell of said mammalian host using said viral vector containing said gene coding for said product.
2. The method of Claim 1, including introducing said gene encoding said product into at least one cell of said connective tissue of said mammalian host for a therapeutic use.
3. The method of Claim 1, including employing as said gene a gene encoding a human interleukin-1 receptor antagonist protein.
4. The method of Claim 1, including employing as said gene a gene encoding a soluble interleukin-1 receptor.
5. The method of Claim 1, including employing as said gene a gene encoding at least one proteinase inhibitor.
6. The method of Claim 5, including employing a tissue inhibitor of metalloproteinases as said proteinase inhibitor.
7. The method of Claim 1, including employing as said gene a gene encoding at least one cytokine.
8. The method of Claim 7, including employing as said cytokine at least one material selected from the group consisting of interleukin-2, interleukin-3, interleukin-4, interleukin-5, interleukin-6, interleukin-7, interleukin-8, interleukin-9, interleukin-10, interleukin-11, and interleukin-12.
9. The method of Claim 7, including employing as said cytokine at least one transforming growth factor.
10.The method of Claim 9, including employing as said transforming growth factor a growth factor selected from the group consisting of TGF-beta1, TGF-beta2, TGF-beta3, and TGF-alpha.
11. The method of Claim 7, including employing as said cytokine at least one fibroblast growth factor.
12. The method of Claim 2, including employing as said viral vector a retroviral vector.
13. The method of Claim 12, including employing as said retroviral vector at least one material selected from the group consisting of MFG and BAG.
14. The method of Claim 13 including employing as said gene a gene encoding a human interleukin-1 receptor antagonist protein and employing MFG as said retroviral vector.
15. The method of Claim 13, including employing as said gene a gene encoding a soluble interleukin-1 receptor.
16. The method of Claim 13, including employing as said gene a gene encoding at least one proteinase inhibitor.
17. The method of Claim 16, including employing a tissue inhibitor of metalloproteinases as said proteinase inhibitor.
18. The method of Claim 13, including employing as said gene a gene encoding at least one cytokine.
19. The method of Claim 18, including employing as said cytokine at least one material selected from the group consisting of interleukin-2, interleukin-3, interleukin-4, interleukin-5, interleukin-6, interleukin-7, interleukin-8, interleukin-9, interleukin-10, interleukin-11, and interleukin-12.
20. The method of Claim 18, including employing as said cytokine at least one transforming growth factor.
21. The method of Claim 20, including employing as said transforming growth factor a growth factor selected from the group consisting of TGF-beta1, TGF-beta2, TGF-beta3, and TGF-alpha.
22. The method of Claim 18, including employing as said cytokine at least one fibroblast growth factor.
23. The method of Claim 1, including employing as said viral vector at least one vector from the group consisting of an adeno-associated virus, adenovirus and a herpes virus.
24. The method of Claim 23, including employing as said gene a gene encoding a human interleukin-1 receptor antagonist protein.
25. The method of Claim 23, including employing as said gene a gene encoding a soluble interleukin-1 receptor.
26. The method of Claim 23, including employing as said gene a gene encoding at least one proteinase inhibitor.
27. The method of Claim 26, including employing a tissue inhibitor of metallproteinases as said proteinase inhibitor.
28. The method of Claim 23, including employing as said gene a gene encoding at least one cytokine.
29. The method of Claim 28, including employing as said cytokine at least one material selected from the group consisting of interleukin-2, interleukin-3, interleukin-4, interleukin-5, interleukin-6, interleukin-7, interleukin-8, interleukin-9, interleukin-10, interleukin-11, and interleukin-12.
30. The method of Claim 28, including employing as said cytokine at least one transforming growth factor.
31. The method of Claim 30, including employing as said transforming growth factor a growth factor selected from the group consisting of TGF-beta1, TGF-beta2, TGF-beta3, and TGF-alpha.
32. The method of Claim 28, including employing as said cytokine at least one fibroblast growth factor.
33. The method of Claim 1, including introducing said gene into said connective tissue which tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium.
34. The method of Claim 33, including employing a cruciate ligament as said ligament.
35. The method of Claim 34, including employing as said cruciate ligament a ligament selected from the group consisting of an anterior cruciate ligament and a posterior-cruciate ligament.
36. The method of Claim 1, including employing as said gene a gene having DNA that is capable of maintenance and expression.
37. The method of Claim 1, including introducing said gene into said cell in vitro.
38. The method of Claim 37, including subsequently transplanting said infected cell into said mammalian host.
39. The method of Claim 37, including after said infecting of said connective tissue cell and before said transplanting of said infected cell into said mammalian host, storing said infected connective tissue cell.
40. The method of Claim 39, including storing said infected connective tissue cell in 10% DMSO under liquid nitrogen.
41. The method of Claim 38, including employing said method to substantially prevent the development of arthritis in a mammalian host having a high susceptibility of developing arthritis.
42. The method of Claim 38, including employing said method on an arthritic mammalian host for a therapeutic use.
43. The method of Claim 38, including employing said method to repair and regenerate said connective tissue which tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium.
44. The method of Claim 43, including employing said method on a mammalian host that is a human being.
45. The method of Claim 14 including effecting in vivo said infection of said cell by introducing said viral vector containing said gene coding for said product directly into said mammalian host.
46. The method of Claim 45, including effecting said direct introduction into said mammalian host by intra-articular injection.
47. The method of Claim 45, including employing said method to substantially prevent the development of arthritis in a mammalian host having a high susceptibility of developing arthritis.
48. The method of Claim 45, including employing said method on an arthritic mammalian host for a therapeutic use.
49. The method of Claim 45, including employing said method to repair and regenerate said connective tissue which tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium.
50. The method of Claim 45, including employing said method on a mammalian host that is a human being.
51. A method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating said mammalian host which comprises:
employing non-viral means for introducing said gene encoding for said product into said connective tissue cell, said non-viral means being selected from the group consisting of at least one liposome, Ca3(PO4)2, electroporation and DEAE-dextran.
52. The method of Claim 51, including employing as said liposome a material selected from the group consisting of DC-cholesterol and SF-cholesterol.
53. The method of Claim 51, including employing as said gene a gene encoding a human interleukin-1 receptor antagonist protein.
54. The method of Claim 51, including employing as said gene a gene encoding an interleukin-1 soluble receptor.
55. The method of Claim 51, including employing as said gene a gene encoding at least one proteinase inhibitor.
56. The method of Claim 55, including employing a tissue inhibitor of metalloproteinases as said proteinase inhibitor.
57. The method of Claim 51, including employing as said gene a gene encoding at least one cytokine.
58. The method of Claim 57, including employing as said cytokine at least one material selected from the group consisting of interleukin-2, interleukin-3, interleukin-4, interleukin-5, interleukin-6, interleukin-7, interleukin-8, interleukin-9, interleukin-10, interleukin-11, and interleukin-12.
59. The method of Claim 57, including employing as said cytokine at least one transforming growth factor.
60. The method of Claim 59, including employing as said transforming growth factor a growth factor selected from the group consisting of TGF-beta1, TGF-beta2, TGF-beta3, and TGF-alpha.
61. The method of Claim 57, including employing as said cytokine at least one fibroblast growth factor.
62. The method of Claim 51, including introducing said gene into connective tissue which tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium.
63. The method of Claim 62, including employing a cruciate ligament as said ligament.
64. The method of Claim 63, including employing as said cruciate ligament a ligament selected from the group consisting of an anterior cruciate ligament and a posterior cruciate ligament.
65. The method of Claim 51, including employing as said gene a gene having DNA that is capable of maintenance and expression.
66. The method of Claim 51, including introducing said gene into said cell in vitro.
67. The method of Claim 66, including subsequently transplanting said cell having said gene into said mammalian host.
68. The method of Claim 67, including after said introducing of said gene encoding for said product into said connective tissue cell and before said transplanting of said connective tissue cell having said gene into said mammalian host, storing said connective tissue cell having said gene.
69. The method of Claim 68, including storing said connective tissue cell having said gene in 10% DMSO under liquid nitrogen.
70. The method of Claim 67, including employing said method to substantially prevent the development of arthritis in a mammalian host having a high susceptibility of developing arthritis.
71. The method of Claim 67, including employing said method on an arthritic mammalian host for a therapeutic use.
72. The method of Claim 67, including employing said method to repair and regenerate said connective tissue which tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium.
73. A method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating said mammalian host which comprises:
employing non-viral means in vivo for directly introducing said gene encoding for said product into said connective tissue cell of said mammalian host, said non-viral means selected from the group consisting of at least one liposome, Ca3(PO4)2 and DEAE-dextran.
74. The method of Claim 73, including effecting said in vivo introduction into said mammalian host by intra-articular injection.
75. The method of Claim 73, including employing said method to substantially prevent the development of arthritis in a mammalian host having a high susceptibility of developing arthritis.
76. The method of Claim 73, including employing said method on an arthritic mammalian host for a therapeutic use.
77. The method of Claim 73, including employing said method to repair and regenerate said connective tissue which tissue is selected from the group consisting of a ligament, a cartilage, a tendon, and a synovium.
78. A method to produce an animal model for the study of connective tissue pathology which comprises:
introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host comprising (a) employing recombinant techniques to produce a viral vector which contains said gene encoding for said product and(b) infecting said connective tissue cell of said mammalian host using said viral vector containing said gene coding for said product for effecting said animal model.
79. The method of Claim 78, including employing as said gene a material selected from the group consisting of a cytokine and a proteinase.
80. The method of Claim 79, including employing as said cytokine a material selected from the group consisting of interleukin-1 alpha, interleukin-1 beta, and TNF-alpha.
81. The method of Claim 79, including employing as said proteinase a matrix metalloproteinase.
82. The method of Claim 78, including employing as said matrix metalloproteinase an enzyme selected from the group consisting of a collagenase, a gelatinase, and a stromelysin.
83. A method to produce an animal model for the study of connective tissue pathology which comprises:
employing non-viral means for introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for effecting said animal model, said non-viral means being selected from the group consisting of at least one liposome, Ca3(PO4)2, electroporation, and DEAE-dextran.
84. The method of Claim 83, including employing as said gene a material selected from the group consisting of a cytokine and a proteinase.
85. The method of Claim 84, including employing as said cytokine a material selected from the group consisting of interleukin-1 alpha, interleukin-1 beta, and TNF-alpha.
86. The method of Claim 84, including employing as said proteinase a matrix metalloproteinase.
87. The method of Claim 86, including employing as said matrix metalloproteinase an enzyme selected from the group consisting of a collagenase, a gelatinase, and a stromelysin.
88. A method of using a gene coding for a truncated interleukin-1 receptor to resist deletrious pathological changes associated with arthritis which comprises:
employing recombinant techniques to produce a retroviral packaging cell line which contain said gene coding for said truncated interleukin-1 receptor;
inserting said gene coding for said truncated interleukin-1 receptor into a retroviral vector wherein said retroviral vector is under the regulation of a suitable eukaryotic promoter;
transfecting said retroviral vector containing said gene coding for said truncated interleukin-1 receptor into said retroviral packaging cell line for the production of a viral particle that is capable of expressing said gene coding for said truncated interleukin-1 receptor; and infecting synovial cells of an inflamed joint of a mammalian host using said viral particle obtained from said retroviral packaging cell line.
89. The method of Claim 88, employing said gene having DNA that replicates and is expressed in said synovial cells lining a joint space of said mammalian host.
90. The method of Claim 88, including employing said method to substantially prevent the development of arthritis in a patient having a high susceptibility of developing arthritis.
91. The method of Claim 88, including employing said method to treat an arthritic patient.
92. The method of Claim 88, including effecting the infection of said synovial cells of a mammalian host by introducing said viral particle directly into said synovial cells lining said inflamed joint space of said mammalian host.
93. The method of Claim 92, including effecting said introduction of said viral particle by parenteral injection.
94. The method of Claim 92, including effecting said introduction of said viral particle by intra-articular injection.
95. The method of Claim 130, including effecting said transplantation of said transduced synovial cells into a patient's joint by employing intra-articular injection.
96. The method of Claim 130, including effecting said infection of said synovial cells of a mammalian host by introducing said viral particles into other synovial cells.
97. A method of using a gene coding for a extracellular interleukin-1 binding domain of an interleukin-1 receptor that is capable of binding to and neutralizing interleukin-1 which comprises:
employing recombinant techniques to produce a retrovirus vector carrying two genes wherein a first gene encodes said extracellular interleukin-1 binding domain of said interleukin-1 receptor and a second gene encodes for selectable antibiotic resistance; and transfecting said retrovirus vector into a retrovirus packaging cell line to obtain a cell line producing nonpathogenic, replication deficient but integration competent, amphitrophic infectious retroviral particles carrying said gene.
98. The method of Claim 97, including initiating introduction of said gene by infection with said retroviral particles from said cell line directly into synovial cells lining an inflamed joint space of a mammalian host.
99. The method of Claim 97, including initiating introduction of said gene by transduction of autologous synovial cells in culture, selecting a synoviocyte cell line by treatment of cultures with antibiotic, and transplanting said selected synoviocyte cells into an affected mammalian joint.
100. The method of Claim 97, wherein effecting said introduction of said viral particles is by parenteral injection.
101. The method of Claim 97, wherein effecting said introduction of said viral particles is by intra-articular injection.
102. A method for preparing a gene encoding an extracellular interleukin-1 binding domain of an interleukin-1 receptor that is capable of binding to and neutralizing interleukin-1 which comprises:
synthesizing said gene by a polymerase chain reaction of said extracellular interleukin-1 binding domain including a signal sequence for secretion of a protein;
introducing amplified interleukin-1 receptor coding sequence into a retroviral vector;
transfecting said retroviral vector into a amphitrophic retrovirus packaging cell line; and collecting viral particles obtained from said retrovirus packaging cell line, wherein said viral particles contain said gene.
103. The gene prepared by the process of Claim 102.
104. A compound for parenteral administration to a patient in a therapeutically effective amount which comprises a gene encoding an extracellular interleukin-1 binding domain of an interleukin-1 receptor and a suitable pharmaceutical carrier.
105. A compound for parenteral administration to a patient in a prophylacticallyeffective amount which comprises a gene encoding an extracellular interleukin-1 binding domain of an interleukin-1 receptor and a suitable pharmaceutical carrier.
106. A method of introducing at least one gene encoding a product into at least one cell of a synovial tissue of a mammalian host for use in treating said mammalian host which comprises:

employing recombinant DNA techniques to produce a DNA vector molecule which contains said gene encoding said product; and injecting said DNA vector molecule into a joint of said mammalian host, said DNA vector molecule subsequently contacting said synovial cell.
107. The method of claim 106 wherein said DNA vector molecule is introduced prophylactically.
108. The method of claim 106 wherein the DNA vector molecule is encapsulated within liposomes prior to said injection into said joint.
109. The method of claim 107 wherein the DNA vector molecule is encapsulated within liposomes prior to said injection into said joint.
110. The method of claim 108, wherein the DNA vector molecule is a plasmid.
111. The method of claim 109, wherein the DNA vector molecule is a plasmid.
112. The method of Claim 106, including employing as said gene a gene encoding a human interleukin-1 receptor antagonist protein.
113. The method of Claim 106, including employing as said gene a gene encoding a soluble interleukin-1 receptor.
114. The method of Claim 106, including employing as said gene a gene encoding at least one proteinase inhibitor.
115. The method of Claim 114, including employing a tissue inhibitor of metalloproteinases as said proteinase inhibitor.
116. The method of Claim 106, including employing as said gene a gene encoding at least one cytokine.
117. The method of Claim 116, including employing as said cytokine at least one material selected from the group consisting of interleukin-2, interleukin-3,interleukin-4, interleukin-5, interleukin-6, interleukin-7, interleukin-8, interleukin-9, interleukin-10, interleukin-11, and interleukin-12.
118. The method of Claim 116, including employing as said cytokine at least one transforming growth factor.
119. The method of Claim 118, including employing as said transforming growth factor a growth factor selected from the group consisting of TGF-beta1, TGF-beta2, TGF-beta3, and TGF-alpha.
120. The method of claim 106, including employing as said gene a gene encoding a soluble tumor necrosis factor receptor.
121. A method of introducing at least one gene encoding a product into at least one cell of a connective tissue of a mammalian host for use in treating said mammalian host which comprises:
employing recombinant techniques to produce a DNA vector molecule which contains said gene encoding for said product; and infecting said connective tissue cell of said mammalian host using a pseudoviruscontaining said DNA vector molecule.
122. The method of Claim 121 wherein the DNA vector is an altered viral genomic molecule, such that said DNA vector contains the heterologous gene of interest to be expressed in at least one cell of said connective tissue of said mammalian host.
123. The method of claim 23 wherein said herpes simplex virus vector is selected from the group consisting of herpes simplex type 1 and herpes simplex type 2.
124. The method of claim 130 wherein the synovial cells are synoviocytes removed from the joint.
125. The method of claim 123 wherein the synoviocytes are removed from the knee joint.
126. The method of claim 130 wherein the synovial cells are skin cells.
127. The method of claim 1, including employing as said gene a gene encoding a soluble tumor necrosis factor receptor.
128. The method of claim 13, including employing as said gene a gene encoding a soluble tumor necrosis factor receptor.
129. The method of claim 23, including employing as said gene a gene encoding a soluble tumor necrosis factor receptor.
130. A method of using a gene coding for a truncated interleukin-1 receptor to resist deleterious pathological changes associated with which comprises:

employing recombinant techniques to produce a retroviral packaging cell line which contain said gene coding for said truncated interleukin-1 receptor;
inserting said gene coding for said truncated interleukin-1 receptor into a retroviral vector whereas said retroviral vector is under regulation of a suitable eukaryotic promoter;
transfecting said retroviral vector containing said gene coding for said truncated interlukin-1 receptor into said retroviral packaging cell line for the production of a viral practice that is capable of expressing said gene coding for said truncated interleukin-1 receptor; and infecting synovial cells of a mammalian host by introducing said viral particlesdirectly into synovial cells in culture to form transduced synovial cells for transplantation into the joint space of said mammalian host.
CA002157782A 1993-03-08 1994-03-07 Gene transfer for treating a connective tissue of a mammalian host Expired - Lifetime CA2157782A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US2775093A 1993-03-08 1993-03-08
US027,750 1993-03-08

Publications (1)

Publication Number Publication Date
CA2157782A1 true CA2157782A1 (en) 1994-09-15

Family

ID=21839568

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002157782A Expired - Lifetime CA2157782A1 (en) 1993-03-08 1994-03-07 Gene transfer for treating a connective tissue of a mammalian host

Country Status (4)

Country Link
EP (1) EP0701563A4 (en)
JP (1) JPH08511507A (en)
CA (1) CA2157782A1 (en)
WO (1) WO1994020517A1 (en)

Families Citing this family (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6159460A (en) * 1988-05-27 2000-12-12 Amgen Inc. Method for treating interleukin-1 mediated diseases
US5075222A (en) 1988-05-27 1991-12-24 Synergen, Inc. Interleukin-1 inhibitors
US6858409B1 (en) 1988-05-27 2005-02-22 Amgen Inc. Nucleic acids encoding interleukin-1 inhibitors and processes for preparing interleukin-1 inhibitors
US6413511B1 (en) * 1990-12-20 2002-07-02 University Of Pittsburgh Of The Commonwealth System Of Higher Education Cartilage alterations by administering to joints chondrocytes comprising a heterologous polynucleotide
US5792751A (en) * 1992-04-13 1998-08-11 Baylor College Of Medicine Tranformation of cells associated with fluid spaces
AU684050B2 (en) * 1992-07-13 1997-12-04 Baylor College Of Medicine Targeting somatic gene therapy to joints
EP0690673A4 (en) * 1993-12-14 1996-05-29 Univ Pittsburgh Systemic gene treatment of connective tissue diseases
US6239268B1 (en) 1994-09-09 2001-05-29 Neurocrine Biosciences, Inc. Interleukin-1 type 3 receptors
CA2199609A1 (en) * 1994-09-09 1996-03-14 Errol B. Desouza Interleukin-1 type 3 receptors
ES2099029B1 (en) * 1994-12-29 1998-01-01 Boeheringer Ingelheim Espana S USE OF THE PLASMIDE PMAMAFGF TO PREPARE VASORRELAJANTOS AND ANTI-HYPERTENSIVE DRUGS.
US6281010B1 (en) 1995-06-05 2001-08-28 The Trustees Of The University Of Pennsylvania Adenovirus gene therapy vehicle and cell line
US5756283A (en) * 1995-06-05 1998-05-26 The Trustees Of The University Of Pennsylvania Method for improved production of recombinant adeno-associated viruses for gene therapy
US5866552A (en) * 1996-09-06 1999-02-02 The Trustees Of The University Of Pennsylvania Method for expressing a gene in the absence of an immune response
ATE406176T1 (en) 1996-12-06 2008-09-15 Amgen Inc IL-1 INHIBITOR IN COMBINATION THERAPY FOR THE TREATMENT OF IL-1-MEDIATED DISEASES
US6294170B1 (en) 1997-08-08 2001-09-25 Amgen Inc. Composition and method for treating inflammatory diseases
US6013253A (en) * 1997-08-15 2000-01-11 Amgen, Inc. Treatment of multiple sclerosis using consensus interferon and IL-1 receptor antagonist
EP1009444A2 (en) * 1997-09-02 2000-06-21 Chiron Corporation Compositions and methods for treating arthritis utilizing gene therapy
EP1939300A1 (en) 1999-05-28 2008-07-02 Targeted Genetics Corporation Methods and compositions for lowering the level of tumor necrosis factor (TNF) in TNF-associated disorders
ATE407214T1 (en) 1999-05-28 2008-09-15 Targeted Genetics Corp METHODS AND COMPOSITIONS FOR REDUCING TUMOR NECROSIS FACTOR (TNF) LEVELS IN TNF-ASSOCIATED DISEASES
US8106098B2 (en) 1999-08-09 2012-01-31 The General Hospital Corporation Protein conjugates with a water-soluble biocompatible, biodegradable polymer
PT2087908T (en) 2001-06-26 2018-07-16 Amgen Inc Antibodies to opgl
NZ593428A (en) 2002-09-06 2013-01-25 Amgen Inc Therapeutic human anti-il-1r1 monoclonal antibody
AR056806A1 (en) 2005-11-14 2007-10-24 Amgen Inc RANKL- PTH / PTHRP ANTIBODY CHEMICAL MOLECULES
US7767206B2 (en) 2006-10-02 2010-08-03 Amgen Inc. Neutralizing determinants of IL-17 Receptor A and antibodies that bind thereto
CN101918018B (en) 2007-11-14 2012-12-05 再生医药有限公司 Methods of using interleukin-1 receptor antagonist as a myeloprotective agent
TW201117824A (en) 2009-10-12 2011-06-01 Amgen Inc Use of IL-17 receptor a antigen binding proteins
CN101690801B (en) 2009-10-26 2012-08-01 上海交通大学 Application of interleukin-1 receptor antagonist and medicinal composition thereof
US20140234330A1 (en) 2011-07-22 2014-08-21 Amgen Inc. Il-17 receptor a is required for il-17c biology
JP6440148B2 (en) * 2013-12-12 2018-12-19 国立大学法人三重大学 Transgenic non-human mammal expressing human MMP2
US20160000936A1 (en) 2014-06-10 2016-01-07 Abbvie Inc. Biomarkers for inflammatory disease and methods of using same
WO2018022982A1 (en) 2016-07-29 2018-02-01 Trustees Of Boston University Age-associated clonal hematopoiesis accelerates cardio-metabolic disease development
WO2020035482A1 (en) 2018-08-13 2020-02-20 Iltoo Pharma Combination of interleukin-2 with an interleukin 1 inhibitor, conjugates and therapeutic uses thereof
WO2023222565A1 (en) 2022-05-16 2023-11-23 Institut National de la Santé et de la Recherche Médicale Methods for assessing the exhaustion of hematopoietic stems cells induced by chronic inflammation

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1282721C (en) * 1984-06-04 1991-04-09 Bernard Roizman Herpes simplex virus as a vector
US5319071A (en) * 1987-11-25 1994-06-07 Immunex Corporation Soluble interleukin-1 receptors
AU659824B2 (en) * 1990-10-31 1995-06-01 Cell Genesys, Inc. Retroviral vectors useful for gene therapy
EP0563239A4 (en) * 1990-12-20 1994-10-12 Univ Pittsburgh A truncated interleukin-1 receptor gene for the treatment of arthritis.
US5180182A (en) * 1991-08-26 1993-01-19 Haworth James R Trailer hitch alignment device

Also Published As

Publication number Publication date
JPH08511507A (en) 1996-12-03
EP0701563A1 (en) 1996-03-20
WO1994020517A1 (en) 1994-09-15
EP0701563A4 (en) 1997-07-02

Similar Documents

Publication Publication Date Title
CA2157782A1 (en) Gene transfer for treating a connective tissue of a mammalian host
AU720762B2 (en) Gene transfer for treating a connective tissue of a mammalian host
US5858355A (en) IRAP gene as treatment for arthritis
US7037492B2 (en) Gene transfer for studying and treating a connective tissue of a mammalian host
Bandara et al. Intraarticular expression of biologically active interleukin 1-receptor-antagonist protein by ex vivo gene transfer.
US6048524A (en) In vivo production and delivery of erythropoietin for gene therapy
ES2294781T3 (en) DNA TRANSFER MEDIATED BY IMPROVED VIRUSES.
US6670178B1 (en) In Vivo production and delivery of insulinotropin for gene therapy
KR100506569B1 (en) Methods of Enhancing Virus-Mediated DNA Delivery Using Molecules Having Virus- and Cell-Binding Domains
US6159464A (en) Viral vectors to inhibit leukocyte infiltration or cartilage degradation of joints
CA2098848A1 (en) Truncated interleukin-1 receptor gene for the treatment of arthritis
AU1266692A (en) Retroviral vectors useful for gene therapy
US6228356B1 (en) Viral vectors to inhibit leukocyte infiltration or cartilage degradation of joints
Evans et al. Gene therapy for arthritis
Ueblacker et al. In vivo analysis of retroviral gene transfer to chondrocytes within collagen scaffolds for the treatment of osteochondral defects
JPH10501140A (en) Retroviral gene therapy vector and therapeutic method based thereon
ES2303372T3 (en) PROCEDURES FOR CONTROL OF DIFFERENTIATION AND CELL GROWTH THAT USE A FUSION PROTEIN AND A PHARMACO.
US6531124B1 (en) In vivo production and delivery of insulinotropin for gene therapy

Legal Events

Date Code Title Description
EEER Examination request
EEER Examination request

Effective date: 20010307

MKEX Expiry

Effective date: 20140307

MKEX Expiry

Effective date: 20140307