WO1999011292A2 - Compositions and methods for treating arthritis utilizing gene therapy - Google Patents

Compositions and methods for treating arthritis utilizing gene therapy Download PDF

Info

Publication number
WO1999011292A2
WO1999011292A2 PCT/US1998/018308 US9818308W WO9911292A2 WO 1999011292 A2 WO1999011292 A2 WO 1999011292A2 US 9818308 W US9818308 W US 9818308W WO 9911292 A2 WO9911292 A2 WO 9911292A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
recombinant
gag
pol
vector
Prior art date
Application number
PCT/US1998/018308
Other languages
French (fr)
Other versions
WO1999011292A3 (en
WO1999011292A9 (en
Inventor
James E. Mccormack
Sunil Chada
Douglas J. Jolly
Original Assignee
Chiron Corporation
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 Chiron Corporation filed Critical Chiron Corporation
Priority to JP2000508393A priority Critical patent/JP2001514235A/en
Priority to EP98945862A priority patent/EP1009444A2/en
Publication of WO1999011292A2 publication Critical patent/WO1999011292A2/en
Publication of WO1999011292A3 publication Critical patent/WO1999011292A3/en
Publication of WO1999011292A9 publication Critical patent/WO1999011292A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction 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
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • A61K38/57Protease inhibitors from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13051Methods of production or purification of viral material
    • C12N2740/13052Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles

Definitions

  • the present invention relates generally to pharmaceutical compositions and methods, and more specifically, to compositions and methods for treating arthritis utilizing gene therapy.
  • rheumatoid arthritis is a debilitating, chronic inflammatory disease affecting 1 to 2% of the world's population. This disease causes pain, swelling and destruction of multiple joints in the body. The disease can also result in damage to other organs such as the lungs and kidneys.
  • rheumatoid arthritis is an autoimmune disease and that many different stimuli can trigger an inappropriate immune response of an immunologically susceptible individual. Examples of potential stimuli include both infectious agents (e.g., viruses) and endogenous host proteins (e.g., collagen).
  • the stimuli i.e., an antigen
  • an antigen is taken up by antigen-presenting cells (macrophages or dendritic cells in the synovial membrane), processed, and presented to T lymphocytes.
  • This presentation initiates an immunological cascade mediated by numerous factors, ultimately resulting in the pain, swelling and cartilage destruction which are associated with rheumatoid arthritis.
  • Therapeutic regimens which are presently being utilized include both steroidal and non-steroidal anti-arthritic agents.
  • anti-cancer drugs such as methotrexate have become the first line of therapy.
  • Other drugs such as cyclosporin and azathioprine (alone or in combination) have also been employed. Many of these agents however, are highly toxic, have significant side effects.
  • the present invention discloses novel compositions and methods for treating arthritis, and further provides other related advantages.
  • the present invention provides compositions and methods for treating or preventing a variety of forms of arthritis, including for example, rheumatoid arthritis, osteoarthritis, and reactive forms of arthritis (e.g., due to a disease process such as Lyme disease, see generally, Goldenberg, Textbook of Rheumatology, and Cooke and Dattwyler, Ann. Rev. Med. 43:93-103, 1992).
  • rheumatoid arthritis e.g., osteoarthritis, and reactive forms of arthritis
  • reactive forms of arthritis e.g., due to a disease process such as Lyme disease, see generally, Goldenberg, Textbook of Rheumatology, and Cooke and Dattwyler, Ann. Rev. Med. 43:93-103, 1992.
  • methods for treating or preventing arthritis comprising the general step of administering to a mammal a therapeutically effective amount of a recombinant retrovirus which directs the expression of an anti-arthritic agent, wherein the recombinant retrovirus is administered at a titer of greater than 10 7 cfu/ml, 10 8 cfu/ml, 10 9 cfu/ml, or, 10 10 cfu/ml.
  • a titer of greater than 10 7 cfu/ml, 10 8 cfu/ml, 10 9 cfu/ml, or, 10 10 cfu/ml.
  • mammals may be treated utilizing such techniques, including for example, humans, horses, dogs, cats, rats and mice.
  • the present invention also provides a variety of anti-arthritic agents that may be utilized, including for example, pro-drug converting enzymes, inhibitors of tumor necrosis factor, IL-1 Receptor antagonists and inhibitors of matrix metalloproteinase expression such as TIMP-1 or TIMP-2.
  • the recombinant retrovirus is administered intraarticularly.
  • Figure 1 is a description of all modifications carried out on retroviral vector as shown in A), resulting in the cross-less retroviral vector shown in B).
  • the cross-less retroviral backbone cloned into a prokaryotic vector is called pBA-5.
  • Retroviral vector and “recombinant retroviral vector” refers to a nucleic acid construct which carries, and within certain embodiments, is capable of directing the expression of a nucleic acid molecule of interest.
  • the retroviral vector must include at least one transcriptional promoter/enhancer or locus defining element(s), or other elements which control gene expression by other means such as alternate splicing, nuclear RNA export, post-translational modification of messenger, or post-transcriptional modification of protein.
  • Such vector constructs must also include a packaging signal, long terminal repeats (LTRs) or portion thereof, and positive and negative strand primer binding sites appropriate to the retrovirus used (if these are not already present in the retroviral vector).
  • LTRs long terminal repeats
  • the recombinant retroviral vector may also include a signal which directs polyadenylation, selectable markers such as Neomycin resistance, TK, hygromycin resistance, phleomycin resistance histidinol resistance, or DHFR, as well as one or more restriction sites and a translation termination sequence.
  • selectable markers such as Neomycin resistance, TK, hygromycin resistance, phleomycin resistance histidinol resistance, or DHFR
  • restriction sites and a translation termination sequence typically include a 5' LTR, a tRNA binding site, a packaging signal, an origin of second strand DNA synthesis, and a 3' LTR or a portion thereof.
  • Retroviral gene delivery vehicle and “retroviral vector particle” as utilized within the present invention refers to a retrovirus which carries at least one gene of interest.
  • the retrovirus may also contain a selectable marker.
  • the recombinant retrovirus is capable of reverse transcribing its genetic material into DNA and incorporating this genetic material into a host cell's DNA upon infection.
  • Prodrug activating enzyme refers to a compound that is capable of activating an otherwise inactive compound into an active compound. Representative examples of prodrug activating enzymes include herpes simplex thymidine kinase and cytosine deaminase.
  • Anti-arthritic agent refers to a molecule which is capable of (1) preventing or lessening the pathological and/or clinical symptoms associated with arthritis; (2) downregulating the white blood cell response which initiates the inflammatory cascade and results in pain, swelling and cartilage destruction; (3) inhibiting the proliferation of synoviocytes; (4) decreasing the production/activity of matrix metalloproteinases; or, (5) inhibiting blood vessel formation.
  • Such molecules include, for example, proteins (e.g., inhibitors of TNF ⁇ or the IL-1 Receptor), antisense and ribozyme nucleic acid sequences.
  • the present invention provides compositions and methods for treating arthritis, comprising the general step of administering to a patient a therapeutically effective amount of a recombinant retrovirus which directs the expression of an anti-arthritic agent, wherein the recombinant retrovirus is administered at a titer of greater than 10 7 cfu/ml. That administration of such recombinant retroviruses can provide a significant therapeutic advantage is surprising, particularly in light of published results which suggest that such a method would be ineffective for the treatment or prevention of arthritis (see Nita et al, Arthritis and Rheumatism 3P(5):820- 828, 1996).
  • the present invention provides recombinant retroviruses which are constructed to carry or express a selected nucleic acid molecule of interest.
  • numerous retroviral gene delivery vehicles may be utilized within the context of the present invention, including for example those described in EP 0,415,731; WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; U.S. Patent No. 5,219,740; WO 9311230; WO 9310218; Vile and Hart, Cancer Res. 53:3860-3864, 1993; Vile and Hart, Cancer Res. 53:962-967, 1993; Ram et al., Cancer Res.
  • retroviruses include those described in WO 91/02805, as well as lentiviral vectors based upon, for example, HIV (see, e.g., PCT Application No.
  • Retroviral gene delivery vehicles of the present invention may be readily constructed from a wide variety of retroviruses, including for example, B, C, and D type retroviruses as well as spumaviruses and lentiviruses (see RNA Tumor Viruses, Second Edition, Cold Spring Harbor Laboratory, 1985).
  • Preferred retroviruses for the preparation or construction of retroviral gene delivery vehicles of the present invention include retroviruses selected from the group consisting of Avian Leukosis Virus, Bovine Leukemia Virus, Murine Leukemia Virus, Mink-Cell Focus-Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis virus and Rous Sarcoma Virus.
  • retroviruses selected from the group consisting of Avian Leukosis Virus, Bovine Leukemia Virus, Murine Leukemia Virus, Mink-Cell Focus-Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis virus and Rous Sarcoma Virus.
  • Particularly preferred Murine Leukemia Viruses include 4070A and 1504A (Hartley and Rowe, J. Virol. 19:19-25, 1976), Abelson (ATCC No. VR-999), Friend (ATCC No. VR-245), Graffi, Gross (ATCC
  • retroviruses may be readily obtained from depositories or collections such as the American Type Culture Collection ("ATCC”; Rockville, Maryland), or isolated from known sources using commonly available techniques.
  • retroviruses may be readily utilized in order to assemble or construct recombinant retroviral vectors and recombinant retroviruses given the disclosure provided herein, and standard recombinant techniques (e.g., Sambrook et al, Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, 1989; Kunkle, N4S 52:488, 1985).
  • portions of the recombinant retroviral vectors or viruses may be derived from different retroviruses.
  • retroviral vector LTRs may be derived from a Murine Sarcoma Virus, a tR ⁇ A binding site from a Rous Sarcoma Virus, a packaging signal from a Murine Leukemia Virus, and an origin of second strand synthesis from an Avian Leukosis Virus.
  • a recombinant retroviral vector as discussed above which lacks certain elements such as gag/pol or env that are required for the production of infectious retrovirus.
  • Such defective retroviral vectors can then be introduced into a cell (termed a "packaging cell") which contains those elements necessary for production of infectious recombinant retrovirus that are lacking in the retroviral vector. The result is to produce a recombinant retrovirus which is infectious, but which lacks those elements required to required to replicate.
  • retroviral vectors may be utilized within the present invention in order to prepare recombinant retroviruses.
  • retroviral vector constructs comprising a 5' LTR, a tR ⁇ A binding site, a packaging signal, one or more heterologous sequences, an origin of second strand D ⁇ A synthesis and a 3' LTR, wherein the vector construct lacks gag/pol or env coding sequences.
  • LTRs Long Terminal Repeats
  • LTRs may be readily identified in the pro virus due to their precise duplication at either end of the genome.
  • a 5' LTR should be understood to include a 5' promoter element and sufficient LTR sequence to allow reverse transcription and integration of the DNA form of the vector.
  • the 3' LTR should be understood to include a polyadenylation signal, and sufficient LTR sequence to allow reverse transcription and integration of the DNA form of the vector.
  • the tRNA binding site and origin of second strand DNA synthesis are also important for a retrovirus to be biologically active, and may be readily identified by one of skill in the art.
  • retroviral tRNA binds to a tRNA binding site by Watson-Crick base pairing, and is carried with the retrovirus genome into a viral particle.
  • the tRNA is then utilized as a primer for DNA synthesis by reverse transcriptase.
  • the tRNA binding site may be readily identified based upon its location just downstream from the 5' LTR.
  • the origin of second strand DNA synthesis is, as its name implies, important for the second strand DNA synthesis of a retrovirus. This region, which is also referred to as the poly-purine tract, is located just upstream of the 3' LTR.
  • retroviral vector constructs which are provided herein also comprise a packaging signal, as well as one or more nucleic acid molecules (e.g., heterologous sequences), each of which is discussed in more detail below.
  • retroviral vector constructs are provided which lack both gag/pol and env coding sequences.
  • the phrase "lacks gag/pol or env coding sequences" should be understood to mean that the retroviral vector does not contain at least 20, preferably at least 15, more preferably at least 10, and most preferably at least 8 consecutive nucleotides which are found in gag/pol or env genes, and in particular, within gag/pol or env expression cassettes that are used to construct packaging cell lines for the retroviral vector construct.
  • Packaging cell lines suitable for use with the above-described retroviral vector constructs may be readily prepared (see U.S. Serial No. 08/240,030, filed May 9, 1994; see also WO 92/05266 and WO 95/30763), and utilized to create producer cell lines (also termed vector cell lines or "VCLs") for the production of recombinant vector particles.
  • producer cell lines also termed vector cell lines or "VCLs”
  • packaging cell lines are made from human (e.g., HT1080 cells) or mink parent cell lines, thereby allowing production of recombinant retroviruses that are capable of surviving inactivation in human serum (e.g., they are "complement resistant").
  • packaging cell lines that produce greater than recombinant retroviral particles at titers greater than 10 ⁇ cfu/ml may readily be obtained.
  • titers are generally obtained from titer assays on HT1080 cells, which produce a three-fold lower titer than titers obtained on murine 3T3 cells.
  • anti-arthritic agents can be delivered (and expressed) by the recombinant retroviruses of the present invention, including not only proteins, but antisense and ribozymes sequences.
  • the recombinant retrovirus directs the expression of a cytotoxic gene.
  • cytotoxic genes include the genes which encode proteins such as ricin (Lamb et al., Ewr.
  • recombinant retroviruses which direct the expression of a gene product that activates a compound with little or no cytotoxicity (i.e., a "prodrug activating or converting enzyme" into a toxic product.
  • gene products include varicella zoster virus thymidine kinase (VZVTK), herpes simplex virus thymidine kinase (HSVTK) (Field et al, J. Gen. Virol. 49:115-124, 1980; Munir et al., Protein Engineering 7(l):83-89, 1994; Black and Loeb, Biochem 32(43): 11618-11626, 1993), and E.
  • VZVTK varicella zoster virus thymidine kinase
  • HSVTK herpes simplex virus thymidine kinase
  • interleukin- 1 antagonists such as Interleukin- 1 Receptor Antagonist Protein or 'TRAP
  • 'TRAP Interleukin- 1 Receptor Antagonist Protein
  • ⁇ isenberg et al Nature 343(6256):341-346, 1990, Carter et al, Nature 344(6267):633-638, 1990, Carrier et al, Cytokine 4(2):83- 89, 1992, Eisenberg et al, PNAS USA 55(12):5232-5236, 1991
  • type II IL-1 receptors e.g., Genebank Accession Nos.
  • Poxvirus "decoy” receptors naturally occurring proteins similar to the type II "decoy” receptor, which are used by the virus to help regulate local immune responses
  • anti-IL- 1 antibodies which bind to and neutralize the biological activity of IL-1
  • antisense or ribozyme molecules which target interleukin- 1.
  • TNF antagonists such as sTNFR; sTNFR/Ig (e.g., a construct of the extracellular portion of the TNF-R which is linked to an immunoglobulin constant region); anti-TNF antibodies (e.g., antibodies which bind to and neutralize the biological activity of TNF); and antisense or ribozyme molecules which target TNF (see generally Nophar et al, EMBO J. 9(10):3269-3278, 1990, Gray et al, PNAS USA 57:7380-7384, 1990; Smith et al, Science 245(4958): 1019- 1023, 1990; Genebank Accession Nos. X55313, M60275, M37764 and M32315).
  • gene products which can also be utilized for treating or preventing arthritis include gene products which switch an immune response from helper 1 (inflammatory) to a helper 2 response (e.g., IL-4 (Klein et al, Immunogenetics 41(1):51, 1995, Arai et al, J. Immunol. 742(l):274-282, 1989, Yokota et al, PNAS USA 53(16):5894-5898, 1986, Genebank Accession Nos. X81851, M23442 and M13982), IL-10 (Kube, Immunogenetics 45(l):82-82, 1996, Vieira et al, PNAS USA 55(44): 1172-1176, 1991, Genebank Accession Nos.
  • IL-4 Kerbein et al, Immunogenetics 41(1):51, 1995, Arai et al, J. Immunol. 742(l):274-282, 1989, Yokota et al, PNAS USA 53(16):58
  • chrodrocyte growth factors and stimulators such as IGF-1 (EP 0155655, Tobin et al, Mol Endocrinol 4(12): 1914- 1920, 1990, Genebank Accession Nos. A29117, E00602, A04811, A04812, AA542914 AA456717 and M37484); TGF- ⁇ (JP 1986219395, EP 0542679, EP 0433225, Kim et al, J. biol Chem. 264(l):402-408, 1989, Genebank Accession Nos. E00973 A18277 and A23751); and human growth hormone.
  • IGF-1 EP 0155655, Tobin et al, Mol Endocrinol 4(12): 1914- 1920, 1990, Genebank Accession Nos. A29117, E00602, A04811, A04812, AA542914 AA456717 and M37484
  • TGF- ⁇ JP 1986219395, EP 0542679, EP
  • Recombinant retrovirus of the present invention is preferably concentrated, purified, and formulated so as to preserve the infectious capability of the virus upon reconstitution (see generally, U.S. Serial No. 08/153,342 and PCT Publication Nos. WO 95/16582 and WO 95/16852). More specifically, recombinant retrovirus of the present invention can be purified and concentrated as generally described in PCT publication No. WO 95/16582, and then preserved by adding a sufficient amount of a formulation buffer to the media containing the recombinant retrovirus, in order to form an aqueous suspension.
  • the formulation buffer is an aqueous solution that contains a saccharide, a high molecular weight structural additive, and a buffering component in water.
  • a "buffering compound” or “buffering component” should be understood to refer to a substance that functions to maintain the aqueous suspension at a desired pH.
  • the aqueous solution may also contain one or more amino acids.
  • the recombinant retrovirus can be preserved prior to the addition of the formulation buffer. Briefly, the crude recombinant retrovirus is clarified by passing it through a filter, and then concentrated, such as by a cross flow concentrating system (Filtron Technology Corp., Nortborough, MA). Within one embodiment, DNase is added to the concentrate to digest exogenous DNA.
  • the digest is then diafiltrated to remove excess media components and establish the recombinant retrovirus in a more desirable buffered solution.
  • the diafiltrate is then passed over a Sephadex S-500 gel column and a purified recombinant retrovirus is eluted.
  • a sufficient amount of formulation buffer is added to this eluate to reach a desired final concentration of the constituents and to minimally dilute the recombinant retrovirus, and the aqueous suspension is then stored, preferably at -70°C or immediately dried.
  • the formulation buffer can be an aqueous solution that contains a saccharide, a high molecular weight structural additive, and a buffering component in water.
  • the aqueous solution may also contain one or more amino acids.
  • the crude recombinant retrovirus can also be purified by ion exchange column chromatography. This method is described in more detail in U.S. Patent Application Serial No. 08/093,436.
  • the crude recombinant retrovirus is clarified by passing it through a filter, and the filtrate loaded onto a column containing a highly sulfonated cellulose matrix.
  • the recombinant retrovirus is eluted from the column in purified form by using a high salt buffer.
  • the high salt buffer is then exchanged for a more desirable buffer by passing the eluate over a molecular exclusion column.
  • a sufficient amount of formulation buffer is then added, as discussed above, to the purified recombinant retrovirus and the aqueous suspension is either dried immediately or stored, preferably at -70°C.
  • the aqueous suspension in crude or purified form can be dried by lyophilization or evaporation at ambient temperature.
  • lyophilization involves the steps of cooling the aqueous suspension below the glass transition temperature or below the eutectic point temperature of the aqueous suspension, and removing water from the cooled suspension by sublimation to form a lyophilized retrovirus. Briefly, aliquots of the formulated recombinant retrovirus are placed into an Edwards Refrigerated Chamber (3 shelf RC3S unit) attached to a freeze dryer (Supermodulyo 12K). A multistep freeze drying procedure as described by Phillips et al.
  • the recombinant retrovirus which directs the expression of one or more anti-arthritic agents is then administered in a therapeutically effective amount to a patient in order to treat or prevent the arthritis.
  • efficacious administration of the recombinant retroviruses described herein may be assessed in several ways, including: (1) by preventing or lessening the pathological and/or clinical symptoms associated with the arthritis; (2) by downregulating the inflammatory response which initiates the inflammatory cascade and results in pain, swelling and cartilage destruction; (3) by inhibiting the proliferation of synoviocytes; (4) by decreasing the production/activity of matrix metalloproteinases; and (5) by inhibiting blood vessel formation.
  • assays may be utilized to determine whether a particular therapeutic regimen or anti-arthritic agent will prevent or lessen the pathological and/or clinical symptoms associated with arthritis, prior to use in humans.
  • animal models such as adjuvant-induced arthritis (see Kaklamanis, Clin. Rheumatol 11:41-41, 1992; Houri and O'Sullivan, Curr. Opin. Rheumatol 7:201- 205, 1995), rat streptococcal cell wall arthritis, collagen induced arthritis (Trentham et al., J. Exp. Med. 746:857, 1977; Stuart et al., Ann. Rev. Immunol. 2:199-218, 1984; and Wooley et al., J. Exp.
  • disease progression may be monitored through measurements of inflammation (e.g., volume of affected paws, or the diameter of affected joints), through radiologic evaluation, or by histological assessment of joint damage.
  • inflammation e.g., volume of affected paws, or the diameter of affected joints
  • assays for determining efficacious agents and administration may likewise be readily determined prior to human clinical trials.
  • downregulation of the inflammatory response may be readily determined by assaying synovial fluid from animals which have been treated with the anti-arthritic agent (or recombinant retrovirus) (e.g., by white cell counts or neutrophil activity).
  • inhibition of synoviocyte proliferation may be readily determined by assaying the effect of the anti-arthritic agent or recombinant retroviruson on tritiated thymidine incorporation of synoviocytes, or by directly scoring pannus formation.
  • Assessment of matrix metalloproteinase inhibition may be accomplished by measuring inhibition of collagenase activity utilizing commercially available kits (e.g., available from Boehringer Mannheim).
  • Inhibition of blood vessel formation may likewise be readily determined based upon standard assays, for example, the Chick Chorioallantoic Membrane or "CAM” assay (see the Examples below).
  • the above assays may be readily practiced by one of ordinary skill in the art given the disclosure provided herein (see generally, Kelley et al., eds., Textbook of Rheumatology (4th edition), W.B. Saunders Company, Philadelphia, 1993).
  • one or more injections of the recombinant retrovirus is administered at a titer of greater than 10 7 cfu/ml, 10 8 cfu ml, 10 9 cfu ml, or, 10 10 cfu/ml.
  • the recombinant retrovirus may be delivered at a variety of sites an locations (e.g., intravenously) in order to systemically treat or prevent the arthritis, or locally (e.g., intraarticularly by direct injection or under artheroscopic guidance) to selected joints.
  • the synovial fluid prior to administration of the recombinant retrovirus or anti-arthritic agent, may be aspirated from the joint, and the joint flushed or otherwise treated with one or more enzymes capable of breaking down extracellular matrix proteins.
  • enzymes include collagenases, elastase, hyaluronidase, dispase, papain, pronase, and trypsin.
  • a complement resistant recombinant retrovirus in order to help prevent or limit the amount of retrovirus that is degraded due to the action of complement.
  • the recombinant retroviral preparation be 'clean', in that it should contain low levels of foreign antigens (e.g., fetal calf serum) and low levels of endotoxin.
  • This example describes modification of the extended packaging signal ( ⁇ +) by PCR on the template KT-1 (see WO 95/16582) using primers that introduce two stop codons in the extended packaging signal sequence.
  • the template pKT-1 contains the modification ATT at the normal ATG start site of gag (position 621-623 of SEQ ID NO: 1).
  • the start site was further modified to the stop codon, TAA, and an additional stop codon TGA was added to replace the codon TTA at position 645-647 of SEQ ID NO: 1.
  • PCR reactions were carried out on pKTl, each introducing one stop codon.
  • the primers for the PCR were designed such that the two PCR products had overlapping regions and a splice-overlap extension PCR (SOE-PCR) was carried out with the two PCR products in order to combine the two introduced stop codons on one strand.
  • SOE-PCR splice-overlap extension PCR
  • the first set of ohgonucleotides introducing the change from ATT to TAA were 5'-GGG-AGT-GGT-AAC-AGT-CTG-GCC-TTA-ATT-CTC-AG (SEQ ID NO: _) and 5'-CGG-TCG-ACC-TCG-AGA-ATT-AAT-TC (SEQ ID NO: _ and the second set of ohgonucleotides introducing the change from TTA to TGA were 5'CTG-GGA-GAC-GTC-CCA-GGG-ACT-TC (SEQ ID NO: __) and 5'GGC-CAG- ACT-GTT-ACC-ACT-CCC-TGA-AGT-TTG-AC (SEQ ID NO: __).
  • the flanking primers of the final 708 base pair PCR product introduced the Aat ⁇ l and the Xhol sites, at the 5' and 3', respectively.
  • the ends of the 708 base pair product were blunted and phosphorylated and the product introduced into the Smal and Ec RV digested vector pBluescript SK- (Stratagene, San Diego, Calif.).
  • the resulting plasmid was designated pBA-2, and is shown diagrammatically in Figure 1.
  • Retroviral envelope sequence was removed upstream of the 3' LTR between the Clal site and the TAG stop codon of the envelope coding sequence.
  • the DNA sequence was modified by PCR such that the TAG stop codon was replaced by a Clal site and the 97 nucleotides upstream from this new Clal site to the original Clal site were deleted, as well as the 212 base pairs of retroviral sequence downstream of the 3' LTR.
  • the PCR product was cloned into pPCRII (Invitrogen, San Diego, Calif.) using the TA cloning kit (Invitrogen, San Diego, Calif.) and called pBA-1.
  • pBA-1 was digested with Clal and Apal resulting in a 640 base pair fragment that was cloned into the Clal and Apal digested pBA-3 resulting in the plasmid pBA-4.
  • This plasmid combines the above described 5' LTR and the packaging signal with the 3' LTR.
  • pBA-4 was digested with Apal and Ec RI, ends blunted and religated in order to remove extraneous 3' polylinker sites, resulting in plasmid ⁇ BA-5a.
  • pBA-5a was cut with NotI (blunted) and EcoRI and introduced into Smal and EcoRI digested pUC18 (GIBCO/BRL, Gaithersburg, MD) resulting in pBA-5b. This construct moved the retroviral vector from a pBluescript into an alternate pUC18 vector.
  • HSV-TK Recombinant Vector Containing HSV-TK
  • pBH-1 PCT#UU 091-02805
  • Xhol and EcoRI Xhol and EcoRI
  • BstBI EcoRI and BstBI resulting in a 1.3 kb fragment. Both fragments were cloned into a Xhol and Clal digested pBA-5b resulting in a retroviral vector designated "pMO-TK”.
  • the cloning of a human TIMP-1 gene into recombinant retroviral vectors can be accomplished by PCR amplification using two 38-mer ohgonucleotides.
  • the ohgonucleotides contain 25 nt corresponding to the TIMP-1 gene template, plus additional 5'-flanking sequences comprising a Pme I recognition site and buffer sequence, as follows:
  • TIMP-1 cDNA clone Docherty et al., Nature 318:66, 1985
  • Thermalase thermostable DNA polymerase is performed in a reaction containing 1.5 mM MgCl2 provided by the supplier, 5% DMSO, and Hot Start Wax beads according to the following protocol:
  • the approximately 700 bp TIMP-I gene amplicon is digested with Pme I (or Xho I and Cla I) and ligated into a retroviral vector backbone (pKT-lL, pKT-3BL, pKT-3BCL or pBA-5b) that have been digested with Pme I and treated with CIAP.
  • a retroviral vector backbone pKT-lL, pKT-3BL, pKT-3BCL or pBA-5b
  • Proper orientation of the insert sequence is determined by restriction endonuclease digests. Production of packaged vector particles is accomplished using the methods described below. Other related gene products from this group are readily cloned using these approaches and ohgonucleotide primers which contain appropriate restriction sites, and are specific for their respective gene.
  • This example describes the gag/pol expression plasmid cassette that contains wobbled non-coding sequences upstream from the gag start site, thereby reducing recombination potential between the gag/pol expression element and the extended packaging signal of a retroviral vector construct, and inhibiting co-packaging of the gag/pol expression element along with the retroviral vector.
  • a 406 bp DNA fragment with a Clal site at the 5' end (underlined) and a Narl site at the 3' end (underlined) was synthesized by Operon (Operon Technologies Inc, Alameda CA). The sequence of the 406 bp DNA fragment was verified and is provided in Table 1. The synthesized DNA was transferred to a shuttle plasmid as a Clal-Narl fragment to create the plasmid pWOB.
  • the Clal-Narl fragment from pWOB was isolated by Clal-Narl digest, and the 406 bp fragment cloned into the Clal-Narl site of pSCVIO to create the plasmid pSCVlO/wob (-Narl fragment) which resulted in the loss of the 481 bp Narl-Narl fragment just downstream of the wobbled Clal-Narl fragment.
  • the PCR fragment was digested with Clal and PstI and the 131 bp fragment cloned into pSCVIO replacing the existing C/ T-Pst/ DNA fragment to create the plasmid pSCV10/5'truncated g/p.
  • This example describes the construction of the 5' truncated gag/pol construct analogous to that described under section B above in the pCI (Promega Corp, Madison, WI) vector backbone.
  • fragments were prepared for a three-way ligation as follows: pCI was digested with Smal and NotI and the 4 kb fragment was isolated. pSCVIO was digested with Xhol and NotI and the 4.7 kb fragment was isolated. pSCVl 0/5' truncated g/p as described in section B was digested with Clal, filled in with Klenow to blunt, then digested with Xhol and the 0.95 kb fragment was isolated. These three fragments were then ligated together to give the final plasmid pCI/5 'truncated g/p.
  • the wobbled gag/pol sequence (0.47 kb) was retrieved from plasmid pSCVlO/wob (-Narl fragment) as described in section A above by digestion with Clal and Xhol. This fragment was cloned into the Clal-Xhol digested pBluescript SK- (Stratagene, San Diego, CA) to create pBluescript/wob (- Narl fragment).
  • This plasmid was digested with EcoRI and N ⁇ r7 to retrieve the wobbled gag sequence and the EcoRI-Narl fragment cloned into the EcoRI-Narl digested pCI/5 1 truncated g/p described in section C above in order to create pCI/5 'truncated wob g/p.
  • This example describes the construction of the 5' and 3' truncated gag/pol construct in the pCI vector where the 5' truncation as described in section C is combined with the following 3'truncation upstream of the stop codon eliminating the D ⁇ A sequence coding for the last 28 amino acids of the pol protein.
  • the plasmid pCI/5'truncated g/p described in section C was linearized with the restriction enzyme Smal which is located 84 bases upstream of the gag/pol termination codon in the open reading frame of gag/pol.
  • the linearized plasmid was ligated to the ohgonucleotide 5' TAAGCGGCCG CTTA (SEQ ID NO: ).
  • This ohgonucleotide is self-complementary and forms a palindromic duplex containing a TAA termination codon and a NotI restriction endonuclease site.
  • the reaction was purified by GeneClean and digested with Smal to recut any vector that did not contain an insert. The reaction was used to transform XL1 Blue E. coli (Stratagene, San Diego, CA) and plasmid DNA from a correct clone was then digested with NotI.
  • NotI cuts in the inserted ohgonucleotide as well as just upstream of the SV40 termination/polyadenylation site of the pCI vector.
  • the digested plasmids were purified by Geneclean and religated to recircularize. Bacteria were transformed and clones were identified which deleted the sequences between the NotI site introduced by the ohgonucleotide and the Not7 site in the pCI vector. These sequences include sequences encoding the last 28 amino acids of gag/pol as well as MoMLV sequences and vector sequences carried over from pSCVIO.
  • the resulting gag/pol construct was named pCI- GPM.
  • the identically shortened gag/pol region was cloned by standard techniques into a pSCVIO background expression cassette. This expression plasmid was named pSCV10/5',3'tr.
  • This example describes the construction of the 5' and 3' truncated and wobbled gag/pol construct in the pCI vector combining the 5' truncation and wobbled gag/pol sequence from section D above with the 3'truncation described in section E above.
  • pCI/5 'truncated wob g/p was linearized with Smal and all further steps leading to the 3'truncation of gag/pol were carried out as described in section E above, leading to the 5' and 3' truncated and wobbled gag/pol construct in pCI named pCI-WGPM.
  • PCLs with the gag/pol expression plasmid cassette pCI-WGPM described in Example 3 F and the env expression plasmid pCMV-b/envam result in producer cell lines where sequence overlap between all three areas of homology is completely eliminated.
  • the cell lines HT 1080 (ATCC #CCL 121) and D17 (ATCC #CCL 183) were used as parent cell lines to establish the PCLs.
  • gag/pol plasmid pCI-WGPM was co-transfected together with a phleomycin r expressing marker plasmid into HT 1080 and D17 cells, selected and dilution cloned as described above.
  • HT 1080 and D17 derived clones were isolated and analyzed for intracellular p30 expression levels as described above. Results of the p30 Western are shown below in Table 2.
  • Table 2 HT 1080 and D17 derived clones screened for intracellular p30 levels after introduction of gag/pol expression cassette pCI-WGPM
  • HT-1080g/p 96 26 3-4 clones have p30 levels (pCI-WGPM) (27%) that are comparable to HTSCV21
  • pCI-WGPM 12 HT 1080 and 22 Dl 7 gag/pol intermediates with the highest p30 expression levels were analyzed for titer potential as described above.
  • the titer results for the HT 1080 gag/pol intermediates are shown below in Table 3.
  • the titer potential measured within the range indicates decreases in titer potential of 10-200 fold in most clones.
  • D17 derived PCL clones named HAIIwob and DAIIwob, respectively, were isolated and analyzed for titer potential. Briefly, several rounds of titer potential analysis were carried out using various retroviral vectors. The DA or HA PCL (PCT #WO 92/05266) controls were included as a reference for high titer potential PCLs.
  • the PCL clones were transduced in 24-well plates with the ⁇ -gal coding retroviral vector DX/ND7 (WO 95/16852) at an moi of 5-10 in the presence of 8 ⁇ g/ml polybrene, transient supernatants harvested, filtered (0.45 ⁇ m), HT 1080 target cells transduced and transient ⁇ -gal titer determined using a standard Galactolight transfer of expression procedure described previously.
  • the same transduction assay as described for the first round was repeated with the top clones from round one using standardized PCL cell numbers.
  • the top clones from round two were used to transduce with several retroviral vectors, supernatant from transient and stable pools harvested, filtered, HT 1080 target cells transduced and titers determined.
  • Transient ⁇ -gal titer on VCL pools from transduced HAII and DAII PCLs determined by Galactolight readout.
  • the best DAIIwob PCL clone shows a 4-fold reduction in titer but most clones show >10-fold reduction.
  • the best HAIIwob PCL clone shows a 50-fold reduced titer potential and most HAIIwob clones have >100-fold reduced titer potential.
  • the DAII wob and HAIIwob PCL clones gave in average about a 5-50 fold lower titer potential when compared to DAII and HAII PCLs.
  • DA amphotropic cell line derived from D 17 cells ATCC No. 183, WO 92/05266
  • DA amphotropic cell line derived from D 17 cells ATCC No. 183, WO 92/05266
  • cells are seeded at 5.0 x 10 5 cells/10 cm tissue culture dish in 10 ml DMEM and 10% FBS, 4 ⁇ g/ml polybrene (Sigma, St. Louis, MO) on day 1.
  • FBS 4 ⁇ g/ml polybrene
  • 3 ml, 1.0 ml and 0.2 ml of the freshly collected virus-containing HX media is added to the cells.
  • the cells are incubated with the virus overnight at 37°C.
  • the retroviral also encodes a selectable marker, e.g., neomycin resistance
  • a selectable marker e.g., neomycin resistance
  • the media is removed and 1.0 ml DMEM, 10% FBS with 800 ⁇ g/ml G418 is added to the plate. Only cells that have been transduced with the vector and expressing the selectable marker will survive. In the case of neomycin resistance, G418 resistant pools can be generated over a period of a week.
  • a pool of cells is then dilution cloned by removing the cells from the plate and counting the cell suspension, diluting the cells suspension down to 10 cells/ml and adding 0.1 ml to each well (1 cell/well) of a 96 well plate (Corning, Corning, NY). Cells are incubated for 14 days at 37°C, 10% CO2- As many as twenty-four clones are selected and expanded up to 24 well plates, 6 well plates then 10 cm plates at which time the clones are assayed for expression and the supernatant are collected and assayed for viral titer.
  • the titer of the individual clones is determined by infection of HT 1080 cells, (ATCC No. CCL 121). On day 1, 5.0 x 10 5 HT1080 cells are plated on each well of a 6 well microtiter plate in 3.0 ml DMEM, 10%> FBS and 4 ⁇ g/ml polybrene. On day 2, the supernatant from each clone is serially diluted 10 fold and used to infect the HT1080 cells in 1.0 ml aliquots.
  • the media is replaced with fresh DMEM, 10% FBS media, and the cells incubated with the vector overnight at 37°C, 10%o CO2- On day 3, selection of transduced cells is performed (assuming the presence of a selectable marker in the recombinant vector) by replacing the media with fresh DMEM, 10% FBS media containing the appropriate selection agent, for instance, 800 ⁇ g/ml G418 in the case of neomycin resistance.
  • Cells are incubated at 37°C, 10% CO2 for 14 days at which time G418 resistant colonies are scored at each dilution to determine the viral titer of each clone as cfu/ml.
  • cell lines are derived that produce greater than or equal to 1.0 x 10° " cfu/ml in culture.
  • selectable markers other than drug resistance or when no selectable marker is encoded by the recombinant vector, other titer methods, such as antibody-based assays, PCR assays, etc., may be employed.
  • the packaging cell line HX is transduced with vector generated from the
  • a sample of RIPA lysate containing a total protein concentration of 20 mg and a commercial molecular weight (MW) marker (Amersham, Chicago, IL) are mixed 1 :1 with 2x sample buffer (4% (w/v) SDS, 50 mM Tris-HCl pH 7.0, 24% (v/v) glycerol, 0.1% (w/v) bromophenol blue and 0.05% b-mercaptoethanol) and heated to 65°C for 10 minutes. After heating the sample and MW marker are placed on ice.
  • 2x sample buffer 4% (w/v) SDS, 50 mM Tris-HCl pH 7.0, 24% (v/v) glycerol, 0.1% (w/v) bromophenol blue and 0.05% b-mercaptoethanol
  • Tris HC1 based minigel BioRad, Hercules, CA
  • running buffer 3.0 gm Tris-HCl pH 8.6, 1.0 gm SDS and 14.4 gm glycine to 1.0 L
  • sample and MW marker are loaded onto the gel.
  • Approximately 70 to 120V is applied to the gel until the marker reaches the bottom of the gel.
  • the protein bands are transferred from the gel to an Immobilon-PTM membrane (Millipore, Bedford, MA) by immersing the gel in CAPS buffer pH 11.0 with 5%> (v/v) methanol for 5 minutes.
  • the Hoefer HSI TTE transfer apparatus Hoefer Scientific Instruments, San Francisco, CA is used to transfer proteins from the gel to the membrane. Approximately 70V is applied to the gel for 1.5 hours.
  • the Immobilon-PTM membrane is blocked for 30 minutes at room temperature with 0.5% BM blocking reagent (Boehringer Mannheim, Chicago, IL) containing 3%> BSA, heated slowly in a microwave.
  • the membrane is then probed with a primary mouse antibody that reacts specifically with the protein being detected at a 1 :2000 dilution in BM blocking solution containing a 3% BSA and incubated for 1 hour at room temperature.
  • the membrane is washed three times with 40 ml of PBST (0.2% Tween-20 in PBS) for 10 min and a secondary goat anti-mouse, HRP-labeled antibody (Jackson, Bar Harbor, MA) at a 1 :20,000 dilution in 3% BSA solution (Sigma, St.
  • the membrane is then washed three times with 40 ml of PBST for 10 minutes. After washing, the membrane is submersed in ECL developing solution (Amersham, Chicago, IL) for 1 minute and then wrapped in plastic wrap and exposed to Hyperfilm (Amersham, Chicago, IL) for 5 seconds. The film is then developed and the protein bands are analyzed.
  • ECL developing solution Amersham, Chicago, IL
  • Hyperfilm Amersham, Chicago, IL
  • the microtiter plate is washed four times with fresh FACS buffer. After the final wash, 150 ⁇ l from each well is transferred to a FACS tube (Falcon 2052 tubes) and 100 ⁇ l of FACS buffer is added. The fluorescent signal is detected by a FACScan counter.
  • Frozen tissue sections are dried at room temperature for 30 minutes. The dried tissue sections are fixed with anhydrous acetone for 15 minutes at room temperature and allowed to air dry. The sections are rehydrated with PBS for several minutes. Tissue sections are then incubated in a humid chamber for 45 to 120 minutes at room temperature with 50 ⁇ l of primary antibody that specifically reacts with the protein to be detected (Marek et al., Cancer 67:1377, 1991). The tissue sections are then washed with PBS and allowed to soak in PBS for several minutes.
  • the sections are incubated with 50 ⁇ l of HRP conjugated anti-mouse antibody or HRP-conjugated strept-avidin (DAKO, Carpenteria, CA) for 45 to 120 minutes at room temperature.
  • the tissue sections are then washed with PBS and allowed to soak in PBS for several minutes.
  • the sections are incubated with 60 to 100 ⁇ l substrate solution containing 0.01% H2O2 and 3,3 diaminodenzidine (DAB) for 8 minutes or for 20 minutes with substrate solution containing 100 ⁇ l of a-ethyl carbazole (60 mg of 3-amino-9-ethylcarbazole in 25 ml DMF) in 1 ml of acetate buffer (0.68 g sodium acetate in H2O pH 5.2 adjusted to a volume of 250 mis) and 0.01% H2O2.
  • the tissue sections are washed with water for 2-5 minutes and then stained with haemotoxylin for 15 seconds.
  • Anti-angiogenic activity of various polypeptides useful in accordance with certain aspects of this invention may be assayed on the chorioallantoic membrane (CAM) as described by Takigawa et al, (Biochem. Int. 14:351, 1987). Briefly, B16 melanoma cells are inoculated subcutaneously into the loins of C57BL/6N mice. When the tumors reach approximately 1 cm in diameter, they are excised, cut into pieces of 2 mg and placed on sterile Whatman GF/B glass fiber filter disks (6 mm in diameter; Reeve- Angel, Clifton, NJ) to which 30 ⁇ l of transduced cells have been added.
  • CAM chorioallantoic membrane
  • CAM 10-day-old chicken embryos through windows made in the egg shells on day 8 of inoculation.
  • the embryos are killed 5 days later by injection of 10% formalin in PBS.
  • the CAM is excised, fixed in 10%> formalin in PBS inverted, and examined under a stereo-microscope.
  • Angiogenesis is assayed by measuring the number and thickness of capillaries beneath the filter. A thick capillary, a middle sized capillary, a small capillary, and 5 minute capillaries are given 3, 2, 1 and 1 points, respectively, and the average number of points is defined as the angiogenic activity.
  • the diameters of tumors on the filters are measured in three dimensions and the tumor size is calculated as (p/6) abc mm- (a, b, and c: length, width, and height, respectively). Low average number of points and decreased tumor size indicates anti- angiogenic activity.
  • HSVTK vector transduced cells to gancyclovir may be used to determine the activity of expressed HSVTK expressed in cells treated according to the disclosed methods. Briefly, cells that are transduced with pTK-3 are seeded into six plates at a density of 2.5 x 10° " per plate. In addition, untransduced CT26 and CT26 b-gal (this cell line was transduced with a virus carrying the reporter gene b- galactosidase from E. coll), are also seeded into six plates as controls.
  • Crude recombinant retrovirus is obtained from a Celligan bioreactor (New Brunswick, New Brunswick, NJ) containing DA cells transformed with the recombinant retrovirus (U.S.S.N. 07/395,932) bound to the beads of the bioreactor matrix.
  • the cells release the recombinant retrovirus into the growth media that is passed over the cells in a continuous flow process.
  • the media exiting the bioreactor is collected and passed initially through a 0.8 micron filter then through a 0.65 micron filter to clarify the crude recombinant retrovirus.
  • the filtrate is concentrated utilizing a cross flow concentrating system (Filtron, Boston, MA).
  • DNase Intergen, New York, NY
  • concentrate Approximately 50 units of DNase (Intergen, New York, NY) per ml of concentrate is added to digest exogenous DNA.
  • the digest is diafiltrated using the same cross flow system to 150 mM NaCl, 25 mM tromethamine, pH 7.2.
  • the diafiltrate is loaded onto a Sephadex S-500 gel column (Pharmacia, Piscataway, NJ), equilibrated in 50 mM NaCl, 25 mM tromethamine, pH 7.4.
  • the purified recombinant retrovirus is eluted from the Sephadex S-500 gel column in 50 mM NaCl, 25 mM tromethamine, pH 7.4.
  • the formulation buffer containing lactose was prepared at a 2x concentrated stock solution.
  • the formulation buffer contains 25 mM tromethamine, 70 mM NaCl, 2 mg/ml arginine, 10 mg/ml human serum albumin (HSA), and 100 mg/ml lactose in a final volume of 100 mis at a pH 7.4.
  • the purified recombinant retrovirus is formulated by adding one part 2x lactose formulation buffer to one part S-500 purified recombinant retrovirus.
  • the formulated recombinant retrovirus can be stored at -70°C to -80°C or dried.
  • the formulated retrovirus is lyophilized in an Edwards Refrigerated Chamber (3 Shelf RC3S unit) attached to a Supermodulyo 12K freeze dryer (Edwards High Vacuum, Tonawanda, NY).
  • a Supermodulyo 12K freeze dryer Edwards High Vacuum, Tonawanda, NY.
  • the lyophilized recombinant retrovirus is reconstituted with 1.0 ml water.
  • the infectivity of the reconstituted recombinant retrovirus is determined by a titer activity assay.
  • the assay is conducted on HT 1080 fibroblasts or 3T3 mouse fibroblast cell line (ATCC CCL 163). Specifically, 1.0 x 10 ⁇ cells are plated onto 6 cm plates and incubated overnight at 37°C, 10% CO2- Ten microliters of a dilution series of reconstituted recombinant retroviruses are added to the cells in the presence of 4 mg/mL polybrene (Sigma, St.
  • cells that have been transduced with a recombinant vector which encodes the neo resistance gene are selected for neomycin resistance in G418 containing media and incubated for 5 days at 37°C, 10% CO2- Following initial selection, the cells are re- fed with fresh media containing G418 and incubated for 5 to 6 days. After final selection, the cells are stained with Commassie blue for colony detection. The titer of the sample is determined from the number of colonies, the dilution and the volume used.
  • Mus dunni NIH NIAID Bethesda, MD
  • small scale cocultivations are performed by mixing of 5.0 x 10 ⁇ Mus dunni cells with 5.0 x 10 ⁇ producer cells and seeding the mixture into 10 cm plates (10 ml standard culture media/plate, 4 ⁇ g/ml polybrene) at day 0. Every 3 to 4 days the cultures are split at a 1 :10 ratio and 5.0 x 10 ⁇ Mus dunni cells are added to each culture plate to effectively dilute out the producer cell line and provide maximum amplification of RCR.
  • culture supernatant is harvested, passed through a 0.45 ⁇ cellulose-acetate filter, and tested in the MdH marker rescue assay.
  • Large scale co-cultivations are performed by seeding a mixture of 1.0 x 10 ⁇ Mus dunni cells and 1.0 x 10 ⁇ producer cells into a total of twenty T-150 flasks (30 ml standard culture media flask, 4 ⁇ g/ml polybrene). Cultures are split at a ratio of 1 :10 on days 3, 6, and 13 and at a ratio of 1 :20 on day 9.
  • the final supernatant is harvested, filtered and a portion of each is tested in the MdH marker rescue assay.
  • the MdH marker rescue cell line is cloned from a pool of Mus dunni cells transduced with LHL, a retroviral vector encoding the hygromycin B resistance gene (Palmer et al., PNAS 84: 1055-1059, 1987).
  • the retroviral vector can be rescued from MdH cells upon infection of the cells with RCR.
  • One ml of test sample is added to a well of a 6-well plate containing 1.0 x 10 ⁇ MdH cells in 2 ml standard culture medium (DMEM with 10%> FBS, 1% 200 mM L-glutamine, 1% non-essential amino acids) containing 4 ⁇ g/ml polybrene.

Abstract

Methods are provided for treating or preventing arthritis, comprising the general step of administering to a mammal a therapeutically effective amount of a retroviral vector which directs the expression of an anti-arthritic agent, wherein the retroviral vector is administered at a titer of greater than 107 cfu/ml.

Description

COMPOSITIONS AND METHODS FOR TREATING ARTHRITIS UTILIZING GENE THERAPY
TECHNICAL FIELD The present invention relates generally to pharmaceutical compositions and methods, and more specifically, to compositions and methods for treating arthritis utilizing gene therapy.
BACKGROUND OF THE INVENTION
Due in large part to the rapidly increasing number of aged individuals, arthritis has become a serious health problem worldwide. For example, one form of arthritis, rheumatoid arthritis, is a debilitating, chronic inflammatory disease affecting 1 to 2% of the world's population. This disease causes pain, swelling and destruction of multiple joints in the body. The disease can also result in damage to other organs such as the lungs and kidneys. Briefly, it is generally believed that rheumatoid arthritis is an autoimmune disease and that many different stimuli can trigger an inappropriate immune response of an immunologically susceptible individual. Examples of potential stimuli include both infectious agents (e.g., viruses) and endogenous host proteins (e.g., collagen). In particular, the stimuli (i.e., an antigen) is taken up by antigen-presenting cells (macrophages or dendritic cells in the synovial membrane), processed, and presented to T lymphocytes. This presentation initiates an immunological cascade mediated by numerous factors, ultimately resulting in the pain, swelling and cartilage destruction which are associated with rheumatoid arthritis. Therapeutic regimens which are presently being utilized include both steroidal and non-steroidal anti-arthritic agents. For many patients with severe disease, anti-cancer drugs such as methotrexate have become the first line of therapy. Other drugs, such as cyclosporin and azathioprine (alone or in combination) have also been employed. Many of these agents however, are highly toxic, have significant side effects.
The present invention discloses novel compositions and methods for treating arthritis, and further provides other related advantages.
SUMMARY OF THE INVENTION Briefly stated, the present invention provides compositions and methods for treating or preventing a variety of forms of arthritis, including for example, rheumatoid arthritis, osteoarthritis, and reactive forms of arthritis (e.g., due to a disease process such as Lyme disease, see generally, Goldenberg, Textbook of Rheumatology, and Cooke and Dattwyler, Ann. Rev. Med. 43:93-103, 1992). Within one aspect of the present invention, methods are provided for treating or preventing arthritis comprising the general step of administering to a mammal a therapeutically effective amount of a recombinant retrovirus which directs the expression of an anti-arthritic agent, wherein the recombinant retrovirus is administered at a titer of greater than 107 cfu/ml, 108 cfu/ml, 109 cfu/ml, or, 1010 cfu/ml. A wide variety of mammals may be treated utilizing such techniques, including for example, humans, horses, dogs, cats, rats and mice.
The present invention also provides a variety of anti-arthritic agents that may be utilized, including for example, pro-drug converting enzymes, inhibitors of tumor necrosis factor, IL-1 Receptor antagonists and inhibitors of matrix metalloproteinase expression such as TIMP-1 or TIMP-2. Within certain embodiments of the invention, the recombinant retrovirus is administered intraarticularly.
These and other aspects of the present invention will become evident upon reference to the following detailed description and attached drawings. In addition, various references are set forth herein which describe in more detail certain procedures or compositions (e.g., plasmids, etc.), and are therefore incorporated by reference in their entirety.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a description of all modifications carried out on retroviral vector as shown in A), resulting in the cross-less retroviral vector shown in B). The cross-less retroviral backbone cloned into a prokaryotic vector is called pBA-5.
DETAILED DESCRIPTION OF THE INVENTION
DEFINITIONS Prior to setting forth the invention, it may be helpful to an understanding thereof to first set forth definitions of certain terms that will be used hereinafter.
"Retroviral vector" and "recombinant retroviral vector" refers to a nucleic acid construct which carries, and within certain embodiments, is capable of directing the expression of a nucleic acid molecule of interest. The retroviral vector must include at least one transcriptional promoter/enhancer or locus defining element(s), or other elements which control gene expression by other means such as alternate splicing, nuclear RNA export, post-translational modification of messenger, or post-transcriptional modification of protein. Such vector constructs must also include a packaging signal, long terminal repeats (LTRs) or portion thereof, and positive and negative strand primer binding sites appropriate to the retrovirus used (if these are not already present in the retroviral vector). Optionally, the recombinant retroviral vector may also include a signal which directs polyadenylation, selectable markers such as Neomycin resistance, TK, hygromycin resistance, phleomycin resistance histidinol resistance, or DHFR, as well as one or more restriction sites and a translation termination sequence. By way of example, such vectors typically include a 5' LTR, a tRNA binding site, a packaging signal, an origin of second strand DNA synthesis, and a 3' LTR or a portion thereof.
"Recombinant retrovirus". "retroviral gene delivery vehicle" and "retroviral vector particle" as utilized within the present invention refers to a retrovirus which carries at least one gene of interest. The retrovirus may also contain a selectable marker. The recombinant retrovirus is capable of reverse transcribing its genetic material into DNA and incorporating this genetic material into a host cell's DNA upon infection. "Prodrug activating enzyme" refers to a compound that is capable of activating an otherwise inactive compound into an active compound. Representative examples of prodrug activating enzymes include herpes simplex thymidine kinase and cytosine deaminase.
"Anti-arthritic agent" refers to a molecule which is capable of (1) preventing or lessening the pathological and/or clinical symptoms associated with arthritis; (2) downregulating the white blood cell response which initiates the inflammatory cascade and results in pain, swelling and cartilage destruction; (3) inhibiting the proliferation of synoviocytes; (4) decreasing the production/activity of matrix metalloproteinases; or, (5) inhibiting blood vessel formation. Such molecules include, for example, proteins (e.g., inhibitors of TNFα or the IL-1 Receptor), antisense and ribozyme nucleic acid sequences.
As noted above, the present invention provides compositions and methods for treating arthritis, comprising the general step of administering to a patient a therapeutically effective amount of a recombinant retrovirus which directs the expression of an anti-arthritic agent, wherein the recombinant retrovirus is administered at a titer of greater than 107 cfu/ml. That administration of such recombinant retroviruses can provide a significant therapeutic advantage is surprising, particularly in light of published results which suggest that such a method would be ineffective for the treatment or prevention of arthritis (see Nita et al, Arthritis and Rheumatism 3P(5):820- 828, 1996).
In order to further an understanding of the invention, a brief discussion is provided below of: (1) the preparation of recombinant retroviral vectors, packaging cell lines, producer cell lines and recombinant retroviruses; (2) anti-arthritic agents that can be utilized within the context of the present invention; and (3) the preparation, formulation and administration of recombinant retroviruses.
PREPARATION OF RECOMBINANT RETROVIRAL VECTORS. PACKAGING CELLS. PRODUCER CELLS AND RECOMBINANT RETROVIRUSES
As noted above, the present invention provides recombinant retroviruses which are constructed to carry or express a selected nucleic acid molecule of interest. Briefly, numerous retroviral gene delivery vehicles may be utilized within the context of the present invention, including for example those described in EP 0,415,731; WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; U.S. Patent No. 5,219,740; WO 9311230; WO 9310218; Vile and Hart, Cancer Res. 53:3860-3864, 1993; Vile and Hart, Cancer Res. 53:962-967, 1993; Ram et al., Cancer Res. 53:83-88, 1993; Takamiya et al., J. Neurosci. Res. 33:493-503, 1992; Baba et al., J. Neurosurg. 79:729-735, 1993 (U.S. Patent No. 4,777,127, GB 2,200,651, EP 0,345,242 and WO 91/02805). Particularly preferred recombinant retroviruses include those described in WO 91/02805, as well as lentiviral vectors based upon, for example, HIV (see, e.g., PCT Application No. PCT/US98/14996; and Naldini et al., Science 272:263, 1996), FIV (see, e.g., U.S. Application No. 60/086,825; and Poeschla et al, Nature Med. 4:354, 1998), and foamy viruses (see, e.g., Russell and Miller, J. Virol. 70:211, 1996). Retroviral gene delivery vehicles of the present invention may be readily constructed from a wide variety of retroviruses, including for example, B, C, and D type retroviruses as well as spumaviruses and lentiviruses (see RNA Tumor Viruses, Second Edition, Cold Spring Harbor Laboratory, 1985). Preferred retroviruses for the preparation or construction of retroviral gene delivery vehicles of the present invention include retroviruses selected from the group consisting of Avian Leukosis Virus, Bovine Leukemia Virus, Murine Leukemia Virus, Mink-Cell Focus-Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis virus and Rous Sarcoma Virus. Particularly preferred Murine Leukemia Viruses include 4070A and 1504A (Hartley and Rowe, J. Virol. 19:19-25, 1976), Abelson (ATCC No. VR-999), Friend (ATCC No. VR-245), Graffi, Gross (ATCC No. VR-590), Kirsten, Harvey Sarcoma Virus and Rauscher (ATCC No. VR-998), and Moloney Murine Leukemia Virus (ATCC No. VR- 190). Such retroviruses may be readily obtained from depositories or collections such as the American Type Culture Collection ("ATCC"; Rockville, Maryland), or isolated from known sources using commonly available techniques.
Any of the above retroviruses may be readily utilized in order to assemble or construct recombinant retroviral vectors and recombinant retroviruses given the disclosure provided herein, and standard recombinant techniques (e.g., Sambrook et al, Molecular Cloning: A Laboratory Manual, 2d ed., Cold Spring Harbor Laboratory Press, 1989; Kunkle, N4S 52:488, 1985). In addition, within certain embodiments of the invention, portions of the recombinant retroviral vectors or viruses may be derived from different retroviruses. For example, within one embodiment of the invention, retroviral vector LTRs may be derived from a Murine Sarcoma Virus, a tRΝA binding site from a Rous Sarcoma Virus, a packaging signal from a Murine Leukemia Virus, and an origin of second strand synthesis from an Avian Leukosis Virus.
In order to generate a recombinant retrovirus (and in particular, a recombinant retrovirus which is replication incompetent), it is generally preferred to construct a recombinant retroviral vector as discussed above which lacks certain elements such as gag/pol or env that are required for the production of infectious retrovirus. Such defective retroviral vectors can then be introduced into a cell (termed a "packaging cell") which contains those elements necessary for production of infectious recombinant retrovirus that are lacking in the retroviral vector. The result is to produce a recombinant retrovirus which is infectious, but which lacks those elements required to required to replicate. As discussed in more detail below, a wide variety of retroviral vectors may be utilized within the present invention in order to prepare recombinant retroviruses. For example, within one aspect of the present invention retroviral vector constructs are provided comprising a 5' LTR, a tRΝA binding site, a packaging signal, one or more heterologous sequences, an origin of second strand DΝA synthesis and a 3' LTR, wherein the vector construct lacks gag/pol or env coding sequences. Briefly, Long Terminal Repeats ("LTRs") are subdivided into three elements, designated U5, R and U3. These elements contain a variety of signals which are responsible for the biological activity of a retrovirus, including for example, promoter and enhancer elements which are located within U3. LTRs may be readily identified in the pro virus due to their precise duplication at either end of the genome. As utilized herein, a 5' LTR should be understood to include a 5' promoter element and sufficient LTR sequence to allow reverse transcription and integration of the DNA form of the vector. The 3' LTR should be understood to include a polyadenylation signal, and sufficient LTR sequence to allow reverse transcription and integration of the DNA form of the vector.
The tRNA binding site and origin of second strand DNA synthesis are also important for a retrovirus to be biologically active, and may be readily identified by one of skill in the art. For example, retroviral tRNA binds to a tRNA binding site by Watson-Crick base pairing, and is carried with the retrovirus genome into a viral particle. The tRNA is then utilized as a primer for DNA synthesis by reverse transcriptase. The tRNA binding site may be readily identified based upon its location just downstream from the 5' LTR. Similarly, the origin of second strand DNA synthesis is, as its name implies, important for the second strand DNA synthesis of a retrovirus. This region, which is also referred to as the poly-purine tract, is located just upstream of the 3' LTR.
In addition to a 5' and 3' LTR, tRNA binding site, and origin of second strand DNA synthesis, certain preferred retroviral vector constructs which are provided herein also comprise a packaging signal, as well as one or more nucleic acid molecules (e.g., heterologous sequences), each of which is discussed in more detail below. Within one aspect of the invention, retroviral vector constructs are provided which lack both gag/pol and env coding sequences. As utilized herein, the phrase "lacks gag/pol or env coding sequences" should be understood to mean that the retroviral vector does not contain at least 20, preferably at least 15, more preferably at least 10, and most preferably at least 8 consecutive nucleotides which are found in gag/pol or env genes, and in particular, within gag/pol or env expression cassettes that are used to construct packaging cell lines for the retroviral vector construct.
Packaging cell lines suitable for use with the above-described retroviral vector constructs may be readily prepared (see U.S. Serial No. 08/240,030, filed May 9, 1994; see also WO 92/05266 and WO 95/30763), and utilized to create producer cell lines (also termed vector cell lines or "VCLs") for the production of recombinant vector particles. Within particularly preferred embodiments of the present invention packaging cell lines are made from human (e.g., HT1080 cells) or mink parent cell lines, thereby allowing production of recombinant retroviruses that are capable of surviving inactivation in human serum (e.g., they are "complement resistant").
In addition, for those vectors which are derived from viruses that carry regulatory genes (such as lentiviral vectors and vectors derived from foamy or spuma- viruses), such regulatory genes can be inserted into either a packaging cell line or vector as desired or necessary. Utilizing the methods of the present invention as disclosed herein, packaging cell lines that produce greater than recombinant retroviral particles at titers greater than 10^ cfu/ml may readily be obtained. In addition, it should be noted that such titers are generally obtained from titer assays on HT1080 cells, which produce a three-fold lower titer than titers obtained on murine 3T3 cells.
ANTI-ARTHRITIC AGENTS As noted above, a wide variety of anti-arthritic agents can be delivered (and expressed) by the recombinant retroviruses of the present invention, including not only proteins, but antisense and ribozymes sequences.
For example, within one embodiment of the invention the recombinant retrovirus directs the expression of a cytotoxic gene. Representative examples of cytotoxic genes include the genes which encode proteins such as ricin (Lamb et al., Ewr.
J. Biochem. 148:265-210, 1985), abrin (Wood et al., Eur. J. Biochem. 198:123-132, 1991; Εvensen et al., J. ofBiol. Chem. 266:6848-6852, 1991 : Collins et al., J. ofBiol. Chem. 265:8665-8669, 1990; Chen et al., Fed. of Eur. Biochem Soc. 309:115-118, 1992), diphtheria toxin (Tweten et al., J. Biol. Chem. 260:10392-10394, 1985), cholera toxin (Mekalanos et al, Nature 306:551-551, 1983; Sanchez & Holmgren, PNAS 56:481-485, 1989), gelonin (Stirpe et al., J. Biol. Chem. 255:6947-6953, 1980), pokeweed (Irvin, Pharmac. Ther. 27:371-387, 1983), antiviral protein (Barbieri et al., Biochem. J. 203:55-59, 1982; Irvin et al., Arch. Biochem. & Biophys. 200:418-425, 1980; Irvin, Arch. Biochem. & Biophys. 169:522-52%, 1975), tritin, Shigella toxin (Calderwood et al, PNAS 54:4364-4368, 1987; Jackson et al, Microb. Path. 2:147-153, 1987), and Pseudomonas exotoxin A (Carroll and Collier, J. Biol. Chem. 262:8101- 8711, 1987).
Within other aspects of the invention, recombinant retroviruses are provided which direct the expression of a gene product that activates a compound with little or no cytotoxicity (i.e., a "prodrug activating or converting enzyme") into a toxic product. Representative examples of such gene products include varicella zoster virus thymidine kinase (VZVTK), herpes simplex virus thymidine kinase (HSVTK) (Field et al, J. Gen. Virol. 49:115-124, 1980; Munir et al., Protein Engineering 7(l):83-89, 1994; Black and Loeb, Biochem 32(43): 11618-11626, 1993), and E. coll guanine phosphoribosyl transferase (see U.S. Patent Application Serial No. 08/155,944, entitled "Compositions and Methods for Utilizing Conditionally Lethal Genes," filed November 18, 1993; see also WO 93/10218 entitled "Vectors Including Foreign Genes and Negative Selection Markers", WO 93/01281 entitled "Cytosine Deaminase Negative Selection System for Gene Transfer Techniques and Therapies", WO 93/08843 entitled "Trapped Cells and Use Thereof as a Drug", WO 93/08844 entitled "Transformant Cells for the Prophylaxis or Treatment of Diseases Caused by Viruses, Particularly Pathogenic Retroviruses", and WO 90/07936 entitled "Recombinant Therapies for Infection and Hyperproliferative Disorders.")
Other gene products which may also be expressed by the recombinant retroviruses of the present invention include interleukin- 1 antagonists such as Interleukin- 1 Receptor Antagonist Protein or 'TRAP" (see Genebank Accession Nos. X52015, X53296, X64532 and M63099, Εisenberg et al, Nature 343(6256):341-346, 1990, Carter et al, Nature 344(6267):633-638, 1990, Carrier et al, Cytokine 4(2):83- 89, 1992, Eisenberg et al, PNAS USA 55(12):5232-5236, 1991), type II IL-1 receptors (e.g., Genebank Accession Nos. X59770, McMahan et al, EMBO, J i0(10):2821-2832 (1991)), soluble or mutant type I IL-1 receptors (see generally, Genebank Accession Nos. M27492, X16896, Gubler, Nucleic Acids Res. 77(23):10114, 1989, Sims et al., PNAS USA 86(22):8946-8950, 1989, Dower et al, J. Immunol. 142:4314-4320, 1989); Poxvirus "decoy" receptors (naturally occurring proteins similar to the type II "decoy" receptor, which are used by the virus to help regulate local immune responses); anti-IL- 1 antibodies which bind to and neutralize the biological activity of IL-1; and antisense or ribozyme molecules which target interleukin- 1.
Other useful gene products include TNF antagonists such as sTNFR; sTNFR/Ig (e.g., a construct of the extracellular portion of the TNF-R which is linked to an immunoglobulin constant region); anti-TNF antibodies (e.g., antibodies which bind to and neutralize the biological activity of TNF); and antisense or ribozyme molecules which target TNF (see generally Nophar et al, EMBO J. 9(10):3269-3278, 1990, Gray et al, PNAS USA 57:7380-7384, 1990; Smith et al, Science 245(4958): 1019- 1023, 1990; Genebank Accession Nos. X55313, M60275, M37764 and M32315).
Other gene products which can also be utilized for treating or preventing arthritis include gene products which switch an immune response from helper 1 (inflammatory) to a helper 2 response (e.g., IL-4 (Klein et al, Immunogenetics 41(1):51, 1995, Arai et al, J. Immunol. 742(l):274-282, 1989, Yokota et al, PNAS USA 53(16):5894-5898, 1986, Genebank Accession Nos. X81851, M23442 and M13982), IL-10 (Kube, Immunogenetics 45(l):82-82, 1996, Vieira et al, PNAS USA 55(44): 1172-1176, 1991, Genebank Accession Nos. X78437 M57627 and U16720), and IL-13 (Dolganov et al, Blood 57(8):3316-3326, 1996, Smirnov et al, Gene 155(2):211- 281, 1995, McKenzie et al, J. Immunol. 750:5436-5444, 1993, Genebank Accession Nos. L13209, U31120 and U10307)), as well as gene products which reverse synovial hyperproliferation or joint damage (e.g., prodrugs as discussed above, or metalloprotease inhibitors such as TIMP-1 or -2). Yet other gene products that may be particularly useful for treating certain forms of arthritis, such as osteoarthritis, include chrodrocyte growth factors and stimulators such as IGF-1 (EP 0155655, Tobin et al, Mol Endocrinol 4(12): 1914- 1920, 1990, Genebank Accession Nos. A29117, E00602, A04811, A04812, AA542914 AA456717 and M37484); TGF-β (JP 1986219395, EP 0542679, EP 0433225, Kim et al, J. biol Chem. 264(l):402-408, 1989, Genebank Accession Nos. E00973 A18277 and A23751); and human growth hormone.
PREPARATION AND FORMULATION OF RECOMBINANT RETROVIRUSES
Recombinant retrovirus of the present invention is preferably concentrated, purified, and formulated so as to preserve the infectious capability of the virus upon reconstitution (see generally, U.S. Serial No. 08/153,342 and PCT Publication Nos. WO 95/16582 and WO 95/16852). More specifically, recombinant retrovirus of the present invention can be purified and concentrated as generally described in PCT publication No. WO 95/16582, and then preserved by adding a sufficient amount of a formulation buffer to the media containing the recombinant retrovirus, in order to form an aqueous suspension. Within certain embodiments of the invention, the formulation buffer is an aqueous solution that contains a saccharide, a high molecular weight structural additive, and a buffering component in water. (As utilized within the context of the present invention, a "buffering compound" or "buffering component" should be understood to refer to a substance that functions to maintain the aqueous suspension at a desired pH. The aqueous solution may also contain one or more amino acids.) Within other embodiments, the recombinant retrovirus can be preserved prior to the addition of the formulation buffer. Briefly, the crude recombinant retrovirus is clarified by passing it through a filter, and then concentrated, such as by a cross flow concentrating system (Filtron Technology Corp., Nortborough, MA). Within one embodiment, DNase is added to the concentrate to digest exogenous DNA. The digest is then diafiltrated to remove excess media components and establish the recombinant retrovirus in a more desirable buffered solution. The diafiltrate is then passed over a Sephadex S-500 gel column and a purified recombinant retrovirus is eluted. A sufficient amount of formulation buffer is added to this eluate to reach a desired final concentration of the constituents and to minimally dilute the recombinant retrovirus, and the aqueous suspension is then stored, preferably at -70°C or immediately dried. As noted above, the formulation buffer can be an aqueous solution that contains a saccharide, a high molecular weight structural additive, and a buffering component in water. The aqueous solution may also contain one or more amino acids.
The crude recombinant retrovirus can also be purified by ion exchange column chromatography. This method is described in more detail in U.S. Patent Application Serial No. 08/093,436. In general, the crude recombinant retrovirus is clarified by passing it through a filter, and the filtrate loaded onto a column containing a highly sulfonated cellulose matrix. The recombinant retrovirus is eluted from the column in purified form by using a high salt buffer. The high salt buffer is then exchanged for a more desirable buffer by passing the eluate over a molecular exclusion column. A sufficient amount of formulation buffer is then added, as discussed above, to the purified recombinant retrovirus and the aqueous suspension is either dried immediately or stored, preferably at -70°C.
The aqueous suspension in crude or purified form can be dried by lyophilization or evaporation at ambient temperature. Specifically, lyophilization involves the steps of cooling the aqueous suspension below the glass transition temperature or below the eutectic point temperature of the aqueous suspension, and removing water from the cooled suspension by sublimation to form a lyophilized retrovirus. Briefly, aliquots of the formulated recombinant retrovirus are placed into an Edwards Refrigerated Chamber (3 shelf RC3S unit) attached to a freeze dryer (Supermodulyo 12K). A multistep freeze drying procedure as described by Phillips et al. (Cryobiology 18:414, 1981) is used to lyophilize the formulated recombinant retrovirus, preferably from a temperature of -40°C to -45°C. The resulting composition contains less than 10% water by weight of the lyophilized retrovirus. Once lyophilized, the recombinant retrovirus is stable and may be stored at -20°C to 25°C. ADMINISTRATION As noted above, the recombinant retrovirus which directs the expression of one or more anti-arthritic agents is then administered in a therapeutically effective amount to a patient in order to treat or prevent the arthritis. As utilized within the context of the present invention, it should be understood that efficacious administration of the recombinant retroviruses described herein may be assessed in several ways, including: (1) by preventing or lessening the pathological and/or clinical symptoms associated with the arthritis; (2) by downregulating the inflammatory response which initiates the inflammatory cascade and results in pain, swelling and cartilage destruction; (3) by inhibiting the proliferation of synoviocytes; (4) by decreasing the production/activity of matrix metalloproteinases; and (5) by inhibiting blood vessel formation.
Briefly, a wide variety of assays may be utilized to determine whether a particular therapeutic regimen or anti-arthritic agent will prevent or lessen the pathological and/or clinical symptoms associated with arthritis, prior to use in humans. Examples include animal models such as adjuvant-induced arthritis (see Kaklamanis, Clin. Rheumatol 11:41-41, 1992; Houri and O'Sullivan, Curr. Opin. Rheumatol 7:201- 205, 1995), rat streptococcal cell wall arthritis, collagen induced arthritis (Trentham et al., J. Exp. Med. 746:857, 1977; Stuart et al., Ann. Rev. Immunol. 2:199-218, 1984; and Wooley et al., J. Exp. Med. 754:688, 1981), and antigen-driven arthritis (Dumonde and Glynn, Br. J. Exp. Pathol 43:373-383, 1962). Within such methods, disease progression may be monitored through measurements of inflammation (e.g., volume of affected paws, or the diameter of affected joints), through radiologic evaluation, or by histological assessment of joint damage.
Other assays for determining efficacious agents and administration may likewise be readily determined prior to human clinical trials. For example, downregulation of the inflammatory response may be readily determined by assaying synovial fluid from animals which have been treated with the anti-arthritic agent (or recombinant retrovirus) (e.g., by white cell counts or neutrophil activity). Similarly, inhibition of synoviocyte proliferation may be readily determined by assaying the effect of the anti-arthritic agent or recombinant retroviruson on tritiated thymidine incorporation of synoviocytes, or by directly scoring pannus formation. Assessment of matrix metalloproteinase inhibition may be accomplished by measuring inhibition of collagenase activity utilizing commercially available kits (e.g., available from Boehringer Mannheim). Inhibition of blood vessel formation may likewise be readily determined based upon standard assays, for example, the Chick Chorioallantoic Membrane or "CAM" assay (see the Examples below). The above assays may be readily practiced by one of ordinary skill in the art given the disclosure provided herein (see generally, Kelley et al., eds., Textbook of Rheumatology (4th edition), W.B. Saunders Company, Philadelphia, 1993).
Typically, one or more injections of the recombinant retrovirus is administered at a titer of greater than 107 cfu/ml, 108 cfu ml, 109 cfu ml, or, 1010 cfu/ml. The recombinant retrovirus may be delivered at a variety of sites an locations (e.g., intravenously) in order to systemically treat or prevent the arthritis, or locally (e.g., intraarticularly by direct injection or under artheroscopic guidance) to selected joints. Within certain embodiments, prior to administration of the recombinant retrovirus or anti-arthritic agent, the synovial fluid may be aspirated from the joint, and the joint flushed or otherwise treated with one or more enzymes capable of breaking down extracellular matrix proteins. Representative examples of such enzymes include collagenases, elastase, hyaluronidase, dispase, papain, pronase, and trypsin.
Irrespective of the route or method of administration, it is generally preferred to administer a complement resistant recombinant retrovirus in order to help prevent or limit the amount of retrovirus that is degraded due to the action of complement. In addition, it is generally preferred that the recombinant retroviral preparation be 'clean', in that it should contain low levels of foreign antigens (e.g., fetal calf serum) and low levels of endotoxin. The following examples are offered by way of illustration, and not by way of limitation.
EXAMPLES
EXAMPLE 1
CONSTRUCTION OF RETROVIRAL VECTOR BACKBONES
This example describes the construction of retroviral vectors which have decreased sequence homology to the retroviral gag/pol and envelope expression constructs. In addition, two stop codons are introduced in the DNA sequence of the packaging signal sequence in order to increase the safety of these vectors. These modifications are generally described in Figure 1.
A. Substitution of Nonsense Codons in the Extended Packaging Sequence (Ψ+)
This example describes modification of the extended packaging signal (Ψ+) by PCR on the template KT-1 (see WO 95/16582) using primers that introduce two stop codons in the extended packaging signal sequence. In particular, the template pKT-1 contains the modification ATT at the normal ATG start site of gag (position 621-623 of SEQ ID NO: 1). Here the start site was further modified to the stop codon, TAA, and an additional stop codon TGA was added to replace the codon TTA at position 645-647 of SEQ ID NO: 1.
Briefly, two sets of PCR reactions were carried out on pKTl, each introducing one stop codon. The primers for the PCR were designed such that the two PCR products had overlapping regions and a splice-overlap extension PCR (SOE-PCR) was carried out with the two PCR products in order to combine the two introduced stop codons on one strand. The first set of ohgonucleotides introducing the change from ATT to TAA were 5'-GGG-AGT-GGT-AAC-AGT-CTG-GCC-TTA-ATT-CTC-AG (SEQ ID NO: _) and 5'-CGG-TCG-ACC-TCG-AGA-ATT-AAT-TC (SEQ ID NO: _ and the second set of ohgonucleotides introducing the change from TTA to TGA were 5'CTG-GGA-GAC-GTC-CCA-GGG-ACT-TC (SEQ ID NO: __) and 5'GGC-CAG- ACT-GTT-ACC-ACT-CCC-TGA-AGT-TTG-AC (SEQ ID NO: __). The flanking primers of the final 708 base pair PCR product introduced the Aatϊl and the Xhol sites, at the 5' and 3', respectively. The ends of the 708 base pair product were blunted and phosphorylated and the product introduced into the Smal and Ec RV digested vector pBluescript SK- (Stratagene, San Diego, Calif.). The resulting plasmid was designated pBA-2, and is shown diagrammatically in Figure 1.
B. Removal of Retroviral Sequences Upstream and Downstream from the 3' LTR and Upstream and within the 5' LTR
Retroviral envelope sequence was removed upstream of the 3' LTR between the Clal site and the TAG stop codon of the envelope coding sequence. The DNA sequence was modified by PCR such that the TAG stop codon was replaced by a Clal site and the 97 nucleotides upstream from this new Clal site to the original Clal site were deleted, as well as the 212 base pairs of retroviral sequence downstream of the 3' LTR.
Briefly, the following two ohgonucleotides were used for the PCR: 5'-
CATCGATAAA ATAAAAGATT TTATTTAGTC (SΕQ ID NO: _ and 5'- CAAATGAAAG ACCCCCGCTG AC (SΕQ ID NO: _) and the template was pKTl.
The PCR product was cloned into pPCRII (Invitrogen, San Diego, Calif.) using the TA cloning kit (Invitrogen, San Diego, Calif.) and called pBA-1.
Subsequently, pBA-2 (described in section A above) was digested with
Xbal and Aatll which deleted a part of the multiple cloning site and into this linearized vector the 780 base pair fragment from Nhel to Aatϊl from pKTl was cloned, resulting in the plasmid pBA-3. This plasmid pBA-3 combined the shortened 5' LTR with the above described packaging region including the two introduced stop codons.
Subsequently, pBA-1 was digested with Clal and Apal resulting in a 640 base pair fragment that was cloned into the Clal and Apal digested pBA-3 resulting in the plasmid pBA-4. This plasmid combines the above described 5' LTR and the packaging signal with the 3' LTR.
Subsequently, pBA-4 was digested with Apal and Ec RI, ends blunted and religated in order to remove extraneous 3' polylinker sites, resulting in plasmid ρBA-5a. Subsequently, pBA-5a was cut with NotI (blunted) and EcoRI and introduced into Smal and EcoRI digested pUC18 (GIBCO/BRL, Gaithersburg, MD) resulting in pBA-5b. This construct moved the retroviral vector from a pBluescript into an alternate pUC18 vector.
EXAMPLE 2
INSERTION OF GENES OF INTEREST INTO CROSS-LESS
RETROVIRAL VECTOR BACKBONE PBA-5B
A. Construction of Recombinant Vector Containing HSV-TK In order to insert a gene of interest such as HSV-TK into a retroviral vector backbone, the HSV-TK gene is first retrieved by digesting pBH-1 (PCT#UU 091-02805) with Xhol and EcoRI, resulting in a 1.2 kb fragment. The neomycin gene driven by the SV40 promoter was also retrieved by digesting pKTl with EcoRI and BstBI resulting in a 1.3 kb fragment. Both fragments were cloned into a Xhol and Clal digested pBA-5b resulting in a retroviral vector designated "pMO-TK".
B. Construction of Recombinant Vectors Containing Tissue Inhibitor of Metalloproteinase-I
The cloning of a human TIMP-1 gene into recombinant retroviral vectors can be accomplished by PCR amplification using two 38-mer ohgonucleotides. The ohgonucleotides contain 25 nt corresponding to the TIMP-1 gene template, plus additional 5'-flanking sequences comprising a Pme I recognition site and buffer sequence, as follows:
HTIMP-F:
(Sequence ID No.
5'-TATATGTTTAAACCACCAGAGAACCCACCATGGCCCCC-3' HTIMP-R:
(Sequence ID No.:
5'-ATATAGTTTAAACCCACTCCGGGCAGGATTCAGGCTAT-3'
PCR amplification using these primers, a TIMP-1 cDNA clone (Docherty et al., Nature 318:66, 1985) as template and Thermalase thermostable DNA polymerase, is performed in a reaction containing 1.5 mM MgCl2 provided by the supplier, 5% DMSO, and Hot Start Wax beads according to the following protocol:
Figure imgf000021_0001
Following amplification, the approximately 700 bp TIMP-I gene amplicon is digested with Pme I (or Xho I and Cla I) and ligated into a retroviral vector backbone (pKT-lL, pKT-3BL, pKT-3BCL or pBA-5b) that have been digested with Pme I and treated with CIAP. Proper orientation of the insert sequence is determined by restriction endonuclease digests. Production of packaged vector particles is accomplished using the methods described below. Other related gene products from this group are readily cloned using these approaches and ohgonucleotide primers which contain appropriate restriction sites, and are specific for their respective gene. EXAMPLE 3
CONSTRUCTION OF VARIOUS GAG/POL EXPRESSION PLASMIDS WITH
PARTIALLY OR COMPLETELY REDUCED SEQUENCE OVERLAP TO THE
CROSS-LESS RETROVIRAL BACKBONE AND ENVELOPE
This example describes several modifications of the MoMLV gag/pol expression plasmid pSCVIO (PCT/US91/06852, WO 92/05266) resulting in decreased or eliminated sequence homology to the retroviral backbone and envelope expression constructs.
A. Creation of New Codons for the 5 'Gag
This example describes the gag/pol expression plasmid cassette that contains wobbled non-coding sequences upstream from the gag start site, thereby reducing recombination potential between the gag/pol expression element and the extended packaging signal of a retroviral vector construct, and inhibiting co-packaging of the gag/pol expression element along with the retroviral vector. In order to construct such an expression cassette a 406 bp DNA fragment with a Clal site at the 5' end (underlined) and a Narl site at the 3' end (underlined) was synthesized by Operon (Operon Technologies Inc, Alameda CA). The sequence of the 406 bp DNA fragment was verified and is provided in Table 1. The synthesized DNA was transferred to a shuttle plasmid as a Clal-Narl fragment to create the plasmid pWOB.
Table 1
ATCCzATACCATGGGGCAAACCGTGACTACCCCTCTGTCCCTCACACTGGGCC ATTGGAAGGACGTGGAAAGAATTGCCCATAATCAAAGCGTGGACGTCAAAA AACGCAGGTGGGTGACATTTTGTAGCGCCGAGTGGCCCACATTCAATGTTG GCTGGCCTAGGGATGGAACTTTCAATCGCGATCTGATTACTCAAGTGAAAA TTAAAGTGTTCAGCCCCGGACCCCACGGCCATCCCGATCAAGTTCCTTATAT TGTCACATGGGAGGCTCTCGCTTTCGATCCACCACCTTGGGTGAAACCATTC GTGCATCCCAAACCACCTCCACCCCTCCCACCCAGCGCTCCTAGCCTGCCCT TGGAGCCCCCACGAAGCACACCACCCAGGAGCAGCTTGTACCCTGCTCTGA CCCCCAGCCTCGGCGCC (SEQ ID NO: _
The Clal-Narl fragment from pWOB was isolated by Clal-Narl digest, and the 406 bp fragment cloned into the Clal-Narl site of pSCVIO to create the plasmid pSCVlO/wob (-Narl fragment) which resulted in the loss of the 481 bp Narl-Narl fragment just downstream of the wobbled Clal-Narl fragment.
B. Creation of a 5' truncated gag/pol construct without MoMLV sequence upstream of the start codon in pSCVIO This example describes the gag/pol expression plasmid cassette that eliminated the MoMLV sequence upstream of the ATG start codon in order to prevent sequence overlap to the retroviral backbone.
Briefly, a new C/α7 site followed by an ideal Kozak translational start sequence was introduced upstream of the start codon of the gag/pol construct pSCVIO by PCR using the forward primer 5' CGAATCGATA CCATGGGCCA GACTGTTACC AC (SEQ ID NO: _J (the Clal site is underlined) and the reverse primer 5' CATTCTGCAG AGCAGAAGGT AAC (SEQ ID NO: _ containing a restriction site for PstI (underlined). The PCR fragment was digested with Clal and PstI and the 131 bp fragment cloned into pSCVIO replacing the existing C/ T-Pst/ DNA fragment to create the plasmid pSCV10/5'truncated g/p.
C. Creation of a 5' truncated gag/pol construct without MoMLV sequence upstream of the start codon in pCI
This example describes the construction of the 5' truncated gag/pol construct analogous to that described under section B above in the pCI (Promega Corp, Madison, WI) vector backbone.
Briefly, fragments were prepared for a three-way ligation as follows: pCI was digested with Smal and NotI and the 4 kb fragment was isolated. pSCVIO was digested with Xhol and NotI and the 4.7 kb fragment was isolated. pSCVl 0/5' truncated g/p as described in section B was digested with Clal, filled in with Klenow to blunt, then digested with Xhol and the 0.95 kb fragment was isolated. These three fragments were then ligated together to give the final plasmid pCI/5 'truncated g/p.
D. Creation of a 5' truncated and wobbled gag/pol construct in pCI This example describes the construction of the 5' truncated and wobbled gag/pol construct in the pCI vector where the 5' truncation as described in section C and the wobbled gag sequences as described in section A were combined.
Briefly, the wobbled gag/pol sequence (0.47 kb) was retrieved from plasmid pSCVlO/wob (-Narl fragment) as described in section A above by digestion with Clal and Xhol. This fragment was cloned into the Clal-Xhol digested pBluescript SK- (Stratagene, San Diego, CA) to create pBluescript/wob (- Narl fragment). This plasmid was digested with EcoRI and Nαr7 to retrieve the wobbled gag sequence and the EcoRI-Narl fragment cloned into the EcoRI-Narl digested pCI/51 truncated g/p described in section C above in order to create pCI/5 'truncated wob g/p.
E. Creation of a 5' and 3' truncated gag/pol construct in pCI and pSCV 10
This example describes the construction of the 5' and 3' truncated gag/pol construct in the pCI vector where the 5' truncation as described in section C is combined with the following 3'truncation upstream of the stop codon eliminating the DΝA sequence coding for the last 28 amino acids of the pol protein. Briefly, the plasmid pCI/5'truncated g/p described in section C was linearized with the restriction enzyme Smal which is located 84 bases upstream of the gag/pol termination codon in the open reading frame of gag/pol. The linearized plasmid was ligated to the ohgonucleotide 5' TAAGCGGCCG CTTA (SEQ ID NO: ). This ohgonucleotide is self-complementary and forms a palindromic duplex containing a TAA termination codon and a NotI restriction endonuclease site. After ligation of lOOng vector and 5μM oligo in the presence of T4 DNA ligase, the reaction was purified by GeneClean and digested with Smal to recut any vector that did not contain an insert. The reaction was used to transform XL1 Blue E. coli (Stratagene, San Diego, CA) and plasmid DNA from a correct clone was then digested with NotI. NotI cuts in the inserted ohgonucleotide as well as just upstream of the SV40 termination/polyadenylation site of the pCI vector. The digested plasmids were purified by Geneclean and religated to recircularize. Bacteria were transformed and clones were identified which deleted the sequences between the NotI site introduced by the ohgonucleotide and the Not7 site in the pCI vector. These sequences include sequences encoding the last 28 amino acids of gag/pol as well as MoMLV sequences and vector sequences carried over from pSCVIO. The resulting gag/pol construct was named pCI- GPM. The identically shortened gag/pol region was cloned by standard techniques into a pSCVIO background expression cassette. This expression plasmid was named pSCV10/5',3'tr.
F. Creation of a 5' and 3' truncated and wobbled gag/pol construct in pCI
This example describes the construction of the 5' and 3' truncated and wobbled gag/pol construct in the pCI vector combining the 5' truncation and wobbled gag/pol sequence from section D above with the 3'truncation described in section E above.
Briefly, pCI/5 'truncated wob g/p was linearized with Smal and all further steps leading to the 3'truncation of gag/pol were carried out as described in section E above, leading to the 5' and 3' truncated and wobbled gag/pol construct in pCI named pCI-WGPM.
EXAMPLE 4
CONSTRUCTION AND TESTING TITER POTENTIAL OF PCLs WITH VARIOUS COMBINATIONS
OF GAG/POL AND ENV EXPRESSION CASSETTES RESULTING IN PCLs WITH VARIOUS
DEGREES OF DNA SEQUENCE OVERLAP BETWEEN THE RETROVIRAL COMPONENTS: GA G/POL, ENV AND RETROVIRAL VECTOR
This example describes the production of PCLs with the gag/pol expression plasmid cassette pCI-WGPM described in Example 3 F and the env expression plasmid pCMV-b/envam (see WO 95/30763). PCLs with these gag/pol and env expression plasmids in conjunction with the retroviral vector derived from pBA-5b (Example 1) result in producer cell lines where sequence overlap between all three areas of homology is completely eliminated. The cell lines HT 1080 (ATCC #CCL 121) and D17 (ATCC #CCL 183) were used as parent cell lines to establish the PCLs.
Briefly, gag/pol plasmid pCI-WGPM was co-transfected together with a phleomycinr expressing marker plasmid into HT 1080 and D17 cells, selected and dilution cloned as described above. HT 1080 and D17 derived clones were isolated and analyzed for intracellular p30 expression levels as described above. Results of the p30 Western are shown below in Table 2.
Table 2: HT 1080 and D17 derived clones screened for intracellular p30 levels after introduction of gag/pol expression cassette pCI-WGPM
Gag/pol #clones #clones p30 expression levels intermediates screened positive for for p30 p30 - (%)
D-17g/p 82 36 3-4 clones have p30 levels (pCI-WGPM) (44%) comparable to Dl 7 4-15
HT-1080g/p 96 26 3-4 clones have p30 levels (pCI-WGPM) (27%) that are comparable to HTSCV21 The 12 HT 1080 and 22 Dl 7 gag/pol intermediates with the highest p30 expression levels were analyzed for titer potential as described above. The titer results for the HT 1080 gag/pol intermediates are shown below in Table 3.
Table 3:
Transient β-gal titers from transduced pools of HT 1080 gag/pol intermediates (pCI- WGPM)
Clone# Transient β-gal x-fold titer decrease titer (CFU/ml) (HTSCV21 :HT 1080 gag/pol intermediate)
10 217 >9
12 28 >71
23 670 >3
29 565 >4
34 950 >2
35 398 >5
45 280 >7
52 670 >3
53 600 >3
71 590 >3
86 480 >4
87 55 >36
HTSCV21 >200
The Galactolight readout for HTSCV10 was out of the range with >2000, therefore the above shown decrease in titer potential is likely to be higher. The titer results for the D17 gag/pol intermediates are shown below in Table 4. Table 4:
Transient β-gal titers from transduced pools of D17gag/pol (D17 g/p) intermediates and stable β-gal titers from transduced and G-418 selected pools of D17gag/pol intermediates
Clone# Transient x-fold Stable β- x-fold Transient x-fold β-gal titer decrease gal titer decrease β-gal decrease
(CFU/ml) (D17 4- (CFU/ml) (D17 4-15: (CFU/ml) (D17 4-
15:D17g/ D17g/p 15:D17g/p p inter.) inter.) inter.)
1 0 -
40 >100
20 >200
14 10 >400 1 10 >400 2 1380 >3 800 >5 2.1E4 9 7 30 >133 1 100 >40 7 30 >133 730 >6 90 2111 8 30 >133
49 500 >8 680 >6 9.4E3 20
50 140 >29
51 10 >400
56 600 >7 320 >13 1.8E3 105
57 230 >17 1.3E3 146
60 380 >11 580 >7 1.0E4 190
65 0 -
66 470 >9 330 >12 1.1E3 172
70 30 >133
73 320 >13 1.05E4 0
76 40 >100
79 20 >200
D17 4-15 >4000* >4000* 1.9E5 =out of range
The titer potential measured within the range indicates decreases in titer potential of 10-200 fold in most clones.
A total of 6 D17 and 4 HT 1080 gag/pol intermediates with the highest titer potential were co-transfected with the env expression plasmid pCMV-b/envam, pools selected and dilution cloned as described above. Several hundred HT 1080 and
D17 derived PCL clones named HAIIwob and DAIIwob, respectively, were isolated and analyzed for titer potential. Briefly, several rounds of titer potential analysis were carried out using various retroviral vectors. The DA or HA PCL (PCT #WO 92/05266) controls were included as a reference for high titer potential PCLs. In the first round, the PCL clones were transduced in 24-well plates with the β-gal coding retroviral vector DX/ND7 (WO 95/16852) at an moi of 5-10 in the presence of 8 μg/ml polybrene, transient supernatants harvested, filtered (0.45 μm), HT 1080 target cells transduced and transient β-gal titer determined using a standard Galactolight transfer of expression procedure described previously. In the second round, the same transduction assay as described for the first round was repeated with the top clones from round one using standardized PCL cell numbers. In the third round, the top clones from round two were used to transduce with several retroviral vectors, supernatant from transient and stable pools harvested, filtered, HT 1080 target cells transduced and titers determined.
Data on the titer potential analysis of the first and second round of screening is shown below in Table 5 on a small selection of representative DAII and HAII PCL clones.
Table 5:
Transient β-gal titer on VCL pools from transduced HAII and DAII PCLs determined by Galactolight readout.
Clone# Transient β-gal titer x-fold decrease in titer
(Galactolight, light units) potential (DA:DAIIwob or HA:HAII ob)
D-17 based PCLs called DAIIwob
DAIIwob (pCI-WGPM#60):
7 21 27
11 6 93
21 2 279
30 14 40
33 51 1 1
41 30 19
DA 558
DAIIwob (pCI-WGPM#22):
5 148 0
8 28 5
28 14 1 1
56 15 10
78 39 4
97 10 15
DA 153
HT-1080 based PCLs called HAIIwob
HAIIwob (pCI-WGPM)#34:
4 8 128
7 9 114
35 7 147
43 4 257
53 5 205
65 9 114
66 10 103
77 19 54
79 6 171
80 4 257
95 4 257
105 2 500
115 9 114
118 6 171
HA 1026 The best DAIIwob PCL clone shows a 4-fold reduction in titer but most clones show >10-fold reduction. The best HAIIwob PCL clone shows a 50-fold reduced titer potential and most HAIIwob clones have >100-fold reduced titer potential. In general, the DAII wob and HAIIwob PCL clones gave in average about a 5-50 fold lower titer potential when compared to DAII and HAII PCLs.
EXAMPLE 5 TRANSDUCTION OF PACKAGING CELL LINES
HX AND DA WITH THE VECTOR CONSTRUCTS
TO increase retroviral titers produced from packaging cells, it is preferable to transduce another packaging cell line with retroviral vectors transiently produced from another cell line. For example, DA (an amphotropic cell line derived from D 17 cells ATCC No. 183, WO 92/05266) cells are seeded at 5.0 x 105 cells/10 cm tissue culture dish in 10 ml DMEM and 10% FBS, 4 μg/ml polybrene (Sigma, St. Louis, MO) on day 1. On day 2, 3.0 ml, 1.0 ml and 0.2 ml of the freshly collected virus-containing HX media is added to the cells. The cells are incubated with the virus overnight at 37°C. In those instances when the retroviral also encodes a selectable marker, e.g., neomycin resistance, on day 3, the media is removed and 1.0 ml DMEM, 10% FBS with 800 μg/ml G418 is added to the plate. Only cells that have been transduced with the vector and expressing the selectable marker will survive. In the case of neomycin resistance, G418 resistant pools can be generated over a period of a week. Typically, a pool of cells is then dilution cloned by removing the cells from the plate and counting the cell suspension, diluting the cells suspension down to 10 cells/ml and adding 0.1 ml to each well (1 cell/well) of a 96 well plate (Corning, Corning, NY). Cells are incubated for 14 days at 37°C, 10% CO2- As many as twenty-four clones are selected and expanded up to 24 well plates, 6 well plates then 10 cm plates at which time the clones are assayed for expression and the supernatant are collected and assayed for viral titer.
The titer of the individual clones is determined by infection of HT 1080 cells, (ATCC No. CCL 121). On day 1, 5.0 x 105 HT1080 cells are plated on each well of a 6 well microtiter plate in 3.0 ml DMEM, 10%> FBS and 4 μg/ml polybrene. On day 2, the supernatant from each clone is serially diluted 10 fold and used to infect the HT1080 cells in 1.0 ml aliquots. The media is replaced with fresh DMEM, 10% FBS media, and the cells incubated with the vector overnight at 37°C, 10%o CO2- On day 3, selection of transduced cells is performed (assuming the presence of a selectable marker in the recombinant vector) by replacing the media with fresh DMEM, 10% FBS media containing the appropriate selection agent, for instance, 800 μg/ml G418 in the case of neomycin resistance. Cells are incubated at 37°C, 10% CO2 for 14 days at which time G418 resistant colonies are scored at each dilution to determine the viral titer of each clone as cfu/ml. Using these procedures, cell lines are derived that produce greater than or equal to 1.0 x 10°" cfu/ml in culture. In addition, as those in the art will appreciate, in those instances where selectable markers other than drug resistance, or when no selectable marker is encoded by the recombinant vector, other titer methods, such as antibody-based assays, PCR assays, etc., may be employed. The packaging cell line HX is transduced with vector generated from the
DA vector producing cell line in the same manner as described for transduction of the DA cells from HX supernatant.
Transduction of the DA or HX cells with vectors lacking a neo selectable marker was performed as described above. However, instead of adding G418 to the cells on day 3, the cells are cloned by limiting dilution. Titer is analyzed as described above. EXAMPLE 6 DETERMINATION OF PROTEIN EXPRESSION
A. Western Blotting
i. Preparation of RIPA-Lysates Cells transduced with the recombinant vectors of this invention are harvested with trypsin/EDTA and washed twice with cold PBS. The cells are lysed with 50 to 400 ml RIPA buffer (10 mM Tris-HCl pH 7.0; 1% (v/v) NP40; 0.1% (w/v) SDS; and 150 mM NaCl) and incubated for 15 minutes at room temperature. The lysate is centrifuged at full speed in an Eppendorf centrifuge for 5 minutes. The supernatant is removed and stored at -20°C. A Bradford protein assay is performed to determine the total protein concentration.
ii. SDS-Page
A sample of RIPA lysate containing a total protein concentration of 20 mg and a commercial molecular weight (MW) marker (Amersham, Chicago, IL) are mixed 1 :1 with 2x sample buffer (4% (w/v) SDS, 50 mM Tris-HCl pH 7.0, 24% (v/v) glycerol, 0.1% (w/v) bromophenol blue and 0.05% b-mercaptoethanol) and heated to 65°C for 10 minutes. After heating the sample and MW marker are placed on ice. The slots of a precast 7.5% Tris HC1 based minigel (BioRad, Hercules, CA) are rinsed with running buffer (3.0 gm Tris-HCl pH 8.6, 1.0 gm SDS and 14.4 gm glycine to 1.0 L) and the sample and MW marker are loaded onto the gel. Approximately 70 to 120V is applied to the gel until the marker reaches the bottom of the gel.
iii. Transfer
The protein bands are transferred from the gel to an Immobilon-P™ membrane (Millipore, Bedford, MA) by immersing the gel in CAPS buffer pH 11.0 with 5%> (v/v) methanol for 5 minutes. The Hoefer HSI TTE transfer apparatus (Hoefer Scientific Instruments, San Francisco, CA) is used to transfer proteins from the gel to the membrane. Approximately 70V is applied to the gel for 1.5 hours.
iv. Immunodetection
The Immobilon-P™ membrane is blocked for 30 minutes at room temperature with 0.5% BM blocking reagent (Boehringer Mannheim, Chicago, IL) containing 3%> BSA, heated slowly in a microwave. The membrane is then probed with a primary mouse antibody that reacts specifically with the protein being detected at a 1 :2000 dilution in BM blocking solution containing a 3% BSA and incubated for 1 hour at room temperature. The membrane is washed three times with 40 ml of PBST (0.2% Tween-20 in PBS) for 10 min and a secondary goat anti-mouse, HRP-labeled antibody (Jackson, Bar Harbor, MA) at a 1 :20,000 dilution in 3% BSA solution (Sigma, St. Louis, MO). The membrane is then washed three times with 40 ml of PBST for 10 minutes. After washing, the membrane is submersed in ECL developing solution (Amersham, Chicago, IL) for 1 minute and then wrapped in plastic wrap and exposed to Hyperfilm (Amersham, Chicago, IL) for 5 seconds. The film is then developed and the protein bands are analyzed.
B. Indirect Immunofluorescence Staining and FACS Analysis
The indirect immunofluorescence staining and FACS analysis method of Amlot et al. (Lymphocytes: A Practical Approach, GGB Klaus (ed.), IRL Press (1987), pp 77-72) is used to detect cell surface expression of heterologous protein antigens from recombinant vector transduced cells. Fifty microliters of primary antibody stock, 62 μg/ml of antibody that specifically react with the protein to be detected IN FACS buffer PBSA containing 0.2%> BSA and 0.2%> NaNβ are added to each well in the first column of a 96 well U-shaped microtiter plate. The antibody in this column is then serially diluted to the wells of the remaining columns. Approximately 50 μl of target cell suspension, 2.0 x 10^ to 4.0 x 10^ cells/ml, in FACS buffer are placed in each well (the antibody final concentrations are 31, 10, 3.1, 0.31, and 0.1 μg/ml) of the microtiter plate and incubated for 15 minutes at room temperature. Following incubation, 100 μl of FACS buffer is added to each well and the microtiter plate is centrifuged at 2000 rpm for 1 minute. The supernatant is removed and 200 μl of fresh FACS buffer is added to each well. This process is repeated 3 additional times. After the final wash, the secondary antibody solution containing 85%o FACS buffer, 10% normal mouse serum, and 5% FITC labeled anti-mouse antibody is added. The microtiter plate is washed four times with fresh FACS buffer. After the final wash, 150 μl from each well is transferred to a FACS tube (Falcon 2052 tubes) and 100 μl of FACS buffer is added. The fluorescent signal is detected by a FACScan counter.
C. Simple Indirect Immuno-peroxidase Staining on Frozen Sections Frozen tissue sections are dried at room temperature for 30 minutes. The dried tissue sections are fixed with anhydrous acetone for 15 minutes at room temperature and allowed to air dry. The sections are rehydrated with PBS for several minutes. Tissue sections are then incubated in a humid chamber for 45 to 120 minutes at room temperature with 50 μl of primary antibody that specifically reacts with the protein to be detected (Marek et al., Cancer 67:1377, 1991). The tissue sections are then washed with PBS and allowed to soak in PBS for several minutes. Following soaking, the sections are incubated with 50 μl of HRP conjugated anti-mouse antibody or HRP-conjugated strept-avidin (DAKO, Carpenteria, CA) for 45 to 120 minutes at room temperature. The tissue sections are then washed with PBS and allowed to soak in PBS for several minutes. Following soaking, the sections are incubated with 60 to 100 μl substrate solution containing 0.01% H2O2 and 3,3 diaminodenzidine (DAB) for 8 minutes or for 20 minutes with substrate solution containing 100 μl of a-ethyl carbazole (60 mg of 3-amino-9-ethylcarbazole in 25 ml DMF) in 1 ml of acetate buffer (0.68 g sodium acetate in H2O pH 5.2 adjusted to a volume of 250 mis) and 0.01% H2O2. The tissue sections are washed with water for 2-5 minutes and then stained with haemotoxylin for 15 seconds. The tissue sections are then washed with water for 10 seconds and mounted in a Crystal Mount™ (Biomeda, Alameda, CA) according to manufacturer's instructions. EXAMPLE 7 DETERMINATION OF ACTIVITY OF THE EXPRESSED PROTEIN
A. Determination of Anti-Angiogenic Activity Anti-angiogenic activity of various polypeptides useful in accordance with certain aspects of this invention may be assayed on the chorioallantoic membrane (CAM) as described by Takigawa et al, (Biochem. Int. 14:351, 1987). Briefly, B16 melanoma cells are inoculated subcutaneously into the loins of C57BL/6N mice. When the tumors reach approximately 1 cm in diameter, they are excised, cut into pieces of 2 mg and placed on sterile Whatman GF/B glass fiber filter disks (6 mm in diameter; Reeve- Angel, Clifton, NJ) to which 30 μl of transduced cells have been added. They are placed upside down on the CAM of 10-day-old chicken embryos through windows made in the egg shells on day 8 of inoculation. The embryos are killed 5 days later by injection of 10% formalin in PBS. The CAM is excised, fixed in 10%> formalin in PBS inverted, and examined under a stereo-microscope. Angiogenesis is assayed by measuring the number and thickness of capillaries beneath the filter. A thick capillary, a middle sized capillary, a small capillary, and 5 minute capillaries are given 3, 2, 1 and 1 points, respectively, and the average number of points is defined as the angiogenic activity. The diameters of tumors on the filters are measured in three dimensions and the tumor size is calculated as (p/6) abc mm- (a, b, and c: length, width, and height, respectively). Low average number of points and decreased tumor size indicates anti- angiogenic activity.
B. Determination of the Herpes Simplex Virus Thymidine Kinase Activity
The sensitivity of HSVTK vector transduced cells to gancyclovir may be used to determine the activity of expressed HSVTK expressed in cells treated according to the disclosed methods. Briefly, cells that are transduced with pTK-3 are seeded into six plates at a density of 2.5 x 10°" per plate. In addition, untransduced CT26 and CT26 b-gal (this cell line was transduced with a virus carrying the reporter gene b- galactosidase from E. coll), are also seeded into six plates as controls. Five plates of each cell type are treated twice per day for four consecutive days with medium containing ganciclovir concentrations of 100 μg/ml, 50 μg/ml, 25 μg/ml, 12.5 μg/ml, and 6.25 μg/ml. One plate of each cell type is left untreated. Following this treatment, the cells are removed from each dish using trypsin ΕDTA, resuspended in DMΕM with 10%) FBS and counted. Similar protocols can be employed for other pro-drug activating enzymes.
EXAMPLE 8
FORMULATION OF THE RETROVIRAL VECTOR
Crude recombinant retrovirus is obtained from a Celligan bioreactor (New Brunswick, New Brunswick, NJ) containing DA cells transformed with the recombinant retrovirus (U.S.S.N. 07/395,932) bound to the beads of the bioreactor matrix. The cells release the recombinant retrovirus into the growth media that is passed over the cells in a continuous flow process. The media exiting the bioreactor is collected and passed initially through a 0.8 micron filter then through a 0.65 micron filter to clarify the crude recombinant retrovirus. The filtrate is concentrated utilizing a cross flow concentrating system (Filtron, Boston, MA). Approximately 50 units of DNase (Intergen, New York, NY) per ml of concentrate is added to digest exogenous DNA. The digest is diafiltrated using the same cross flow system to 150 mM NaCl, 25 mM tromethamine, pH 7.2. The diafiltrate is loaded onto a Sephadex S-500 gel column (Pharmacia, Piscataway, NJ), equilibrated in 50 mM NaCl, 25 mM tromethamine, pH 7.4. The purified recombinant retrovirus is eluted from the Sephadex S-500 gel column in 50 mM NaCl, 25 mM tromethamine, pH 7.4.
The formulation buffer containing lactose was prepared at a 2x concentrated stock solution. The formulation buffer contains 25 mM tromethamine, 70 mM NaCl, 2 mg/ml arginine, 10 mg/ml human serum albumin (HSA), and 100 mg/ml lactose in a final volume of 100 mis at a pH 7.4. The purified recombinant retrovirus is formulated by adding one part 2x lactose formulation buffer to one part S-500 purified recombinant retrovirus. The formulated recombinant retrovirus can be stored at -70°C to -80°C or dried.
The formulated retrovirus is lyophilized in an Edwards Refrigerated Chamber (3 Shelf RC3S unit) attached to a Supermodulyo 12K freeze dryer (Edwards High Vacuum, Tonawanda, NY). When the freeze drying cycle is completed, the vials are stoppered under a vacuum following a slight nitrogen gas bleeding. Upon removal, vials are crimped with aluminum seals.
The lyophilized recombinant retrovirus is reconstituted with 1.0 ml water. The infectivity of the reconstituted recombinant retrovirus is determined by a titer activity assay. The assay is conducted on HT 1080 fibroblasts or 3T3 mouse fibroblast cell line (ATCC CCL 163). Specifically, 1.0 x 10^ cells are plated onto 6 cm plates and incubated overnight at 37°C, 10% CO2- Ten microliters of a dilution series of reconstituted recombinant retroviruses are added to the cells in the presence of 4 mg/mL polybrene (Sigma, St. Louis, MO) and incubated overnight at 37°C, 10% CO2- Following incubation, cells that have been transduced with a recombinant vector which encodes the neo resistance gene are selected for neomycin resistance in G418 containing media and incubated for 5 days at 37°C, 10% CO2- Following initial selection, the cells are re- fed with fresh media containing G418 and incubated for 5 to 6 days. After final selection, the cells are stained with Commassie blue for colony detection. The titer of the sample is determined from the number of colonies, the dilution and the volume used.
EXAMPLE 9
DETECTION OF REPLICATION COMPETENT RETROVIRUSES: CO-CULTIVATION OF PRODUCER LINES AND MDH MARKER RESCUE ASSAY
In order to detect the presence of replication competent retrovirus in a vector-producing cell line, producer cells are cocultivated with an equivalent number of Mus dunni (NIH NIAID Bethesda, MD) cells. Briefly, small scale cocultivations are performed by mixing of 5.0 x 10^ Mus dunni cells with 5.0 x 10^ producer cells and seeding the mixture into 10 cm plates (10 ml standard culture media/plate, 4 μg/ml polybrene) at day 0. Every 3 to 4 days the cultures are split at a 1 :10 ratio and 5.0 x 10^ Mus dunni cells are added to each culture plate to effectively dilute out the producer cell line and provide maximum amplification of RCR. On day 14, culture supernatant is harvested, passed through a 0.45 μ cellulose-acetate filter, and tested in the MdH marker rescue assay. Large scale co-cultivations are performed by seeding a mixture of 1.0 x 10^ Mus dunni cells and 1.0 x 10^ producer cells into a total of twenty T-150 flasks (30 ml standard culture media flask, 4 μg/ml polybrene). Cultures are split at a ratio of 1 :10 on days 3, 6, and 13 and at a ratio of 1 :20 on day 9. On day 15, the final supernatant is harvested, filtered and a portion of each is tested in the MdH marker rescue assay.
The MdH marker rescue cell line is cloned from a pool of Mus dunni cells transduced with LHL, a retroviral vector encoding the hygromycin B resistance gene (Palmer et al., PNAS 84: 1055-1059, 1987). The retroviral vector can be rescued from MdH cells upon infection of the cells with RCR. One ml of test sample is added to a well of a 6-well plate containing 1.0 x 10^ MdH cells in 2 ml standard culture medium (DMEM with 10%> FBS, 1% 200 mM L-glutamine, 1% non-essential amino acids) containing 4 μg/ml polybrene. Media is replaced after 24 hours with standard culture medium without polybrene. Two days later, the entire volume of MdH culture supernatant is passed through a 0.45 μ cellulose-acetate filter and transferred to a well of a 6-well plate containing 5.0 x 10^ Mus dunni target cells in 2 ml standard culture medium containing polybrene. After 24 hours, supernatant is replaced with standard culture media containing 250 μg/ml of hygromycin B and subsequently replaced on days 2 and 5 with media containing 200 μg/ml of hygromycin B. Colonies resistant to hygromycin B appear and are visualized on day 9 post-selection by staining with 0.2% Coomassie blue. From the foregoing, it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.
SEQUENCE LISTING
(1) GENERAL INFORMATION COMPOSITIONS AND METHODS FOR TREATING ARTHRITIS UTILIZING GENE THERAPY
(i) APPLICANTS MCCORMACK, JAMES E CHADA, SU IL JOLLY, DOUGLAS J
(n) TITLE OF INVENTION
(in) NUMBER OF SEQUENCES
(iv) CORRESPONDENCE ADDRESS
(A) ADDRESSEE CHIRON CORPORATION
(B) STREET INTELLECTUAL PROPERTY R440
P 0 BOX 8097
(C) CITY EMERYVILLE
(D) STATE CA
(E) COUNTRY USA
(F) ZIP 94662-8097
(v) COMPUTER READABLE FORM
(A) MEDIUM TYPE Floppy disk
(B) COMPUTER IBM PC compatible
(C) OPERATING SYSTEM PC DOS/MS DOS
(D) SOFTWARE Patentin Release #10 Version #125
(vi) CURRENT APPLICATION DATA
(A) APPLICATION NUMBER
(B) FILING DATE
(C) CLASSIFICATION
(vm) ATTORNEY/AGENT INFORMATION
(A) NAME MCMASTERS, DAVID D
(B) REGISTRATION NUMBER 33,963
(C) REFERENCE/DOCKET NUMBER 930049463P1
(ix) TELECOMMUNICATION INFORMATION
(A) TELEPHONE (206)622-4900
(B) TELEFAX (206)682-6031
(2) INFORMATION FOR SEQ ID NO 1
(i) SEQUENCE CHARACTERISTICS
(A) LENGTH 8332 base pairs
(B) TYPE nucleic acid
(C) STRANDEDNESS single
(D) TOPOLOGY linear 0991 33331111393391V9319V V91W333913W313V3333939991V91 V939999391
0091 99399VW9V 1399V9V11313919993W 3VWW9W9 V9933V91391313V99991
OWΪ 19139V39V31913V93V999133V3339V 31V333V31V 3131191319 V931V91313
08GI 9V3V913WV 199V331V9V V9131II11311333W1W 1VWW9913 W3V11133V
0281 91311313313119339913 V1W31139V 3V993VW99 V99V39393313333311V3
0921 99V393133V 13V3313V93399191333339V9993V9V 999193V13393131V3991
0021 W333313333V9933V399 V9V9993913333V939W9 V991991VW 993V999V3V
Ot/TT 9331133333 V33V9W333 V999V1V11339333333V9 W9V3V113V 133V931V31
0801 393399999919V3V9131113119W3133VW133VW 339399V1313113313V31
0201 3339V331V1113331331V 9313393333 V931193133133W911333331313193
096 3339331V331331131331339331339V V1333V3V191113339W31999133313
006 33333V9111139911339V V999133V91931V3V13333199V33V9V 333V3V991V
0^8 3933399133 V311113199 VV31V9W1199V333V31V 3133V9V933 W11133V39
08/ 93V9V93933991V993193 W11133W33991W9V3913139131133V11999119
02Z 3V9V9W9W 3191V9V199319V33W3V 313931V9939V93191V9V W9913V319
099 9V1I33V91119W1133313V33V11913 V9V339991V 1W9V91319111V191311
009 191913V913191313191191913LL931 V39V39139131911319393933939339
Ot/g W933V9991119931113911111W91319331339333119V3WW 133W9V93V
08fr 9V99V199131199191V1V 999V99V9V11333333V39199111313V 991111931V
02fr 93331VWW 3319V9133V 9333991911111933999993113V999V 333193V9V9
098 991333W393399333V3V V993119V93 V913W9919919333V99399131V1913
008 139V13W139V119V13V199319391339391V1111V 913V91V131919V131911 0^2 V93319131919131V113V V39V3399139W199V99933V33V333V 933V33V999 081 V333913333 V9V99931V999331931399999111V3111319999939V319333V1 021 3V911V919V 913133131999V999113311913931319919113V9331V39119V3
09 91131333W V1W331V1919333V19993339319V913V911V93313319V33939
:χ:0N 01 03S :N0Ildiy0S3α 33N3D03S (PO
Ot
£8 l/86Sπ/XDd Z6ZI 1/66 OΛV 01/28 33V39V331V 9933113V9V 39V133V9V9 V3V3913V399V91V911191333V33319
0818 V3VWW3LL 1999V3V33313V9V33V99133V911W3 V99V3131W 9991V9V9V3
0218 31V9V9V9919V91113393113131339V 319V33V3333V3313V9V91339131111
0908 13391V99W V111V911391913V3V19919V33V3331933V3331399939V9LL31
0008 33W3V11333W33391933V3333V331 V3V9W99199939W3W319W9V9V91
01/62 31V99V331913399V1V11 V91W13V999V33VW9W 119333V1391333393V3V
0882 V9913333319V3391333V 19913V1W999V33V9911913V9V9V3V 1V3V33339V
0282 V31V999913 V9V339W9V V3V3191V3333V1W3VW V1V33191933333V13130
09Z2 W39VW913133V1V31V913133139W 3393119V39913V9991V39999933VW
002 99399913399V3133H11 V9131913991V3V331999 V131311191 V9V339V9W
0W2 W3133V9V91V3V139931 V39V91V9W 9V1V1VW1333V911919V V39133339V
0892 399991W33 V9991V1199 V3139V99V31V999V91113V331VW3339WW13W
0292 V13V9139111V9V9W99V 119131331V 13331913V9 V33V191V33133111313V
09^2 333V3199W 19933V139V 131V3V19W V3931V993V 33V9913931 V19939VW9
00^ 9V9913V139999W33199913391319V V1V919W13333V991331 WW333V91
01782 39193313V3 W333999913V1V9V1991331133V319333W3999999319VW31
0822 333W1V99V 333W91333333339V99V 31999V3199 V999V13V91 V9V1333V91
0222 331333133V 9V3333V9W 33V99993133V99V93V33 VW9W3331911V9VW13
0912 99913V39999WW9VW3913V13391919V33V93931V9313W333199V99W9
0012 3W9V99V999V3V9V1V99 V3VW9V3V9919V11931913V339911V 139W39V91
OtOS V9V9V9V1V3 V9V99V1931 V9V9VW9W V9V9VW9V39V91V99V9V 3V99V19339
0861 3W9W9VW W99V9V3W V9V9V99V31 V193W99V9 V9VW9W993333VW9V9
0261 3VW1W11131V9WW9V 399V9V9V1199111V9V9911393V9W3 WWW1LLV
0981 9W9V1199V 9V9V119VW 9V99911V3V 9V333391319V399111V31113191V13
0081 19191W13V W9W3999V 333V99V91333V91V113313V3V199V3931V1339W
OUl 99W113V9V 9V9V133113399313331319V91W333999W3V3W 1W99WW1
0891 99W3399111W33V33339W9V3999393WW3313199939V133139119V339
0291 31V13V3319 V133V33W99V199V399V 333V33V3V11V99913V9V 333939V931
It €8I/86Sn/IDd Z6ZII/660Λ__ 098V 99933W91391V3V13V133319V3V339 W9V9V9V133131399W91VWW3131
008V 139V3133V313V9139V31 V31131113V 9V11V11W911113V1119 V33V913391
OVV V919133VW V99W33V13199911V19V V9VW3WW 1V91V111V3399999119V
08917 V33W133V99VW1V1V913V919V3V3V 11V31111V3 W9V3133V3 V1333V31V3
029V 11WW9V1V 31331333V13133V3V9V3313V9V9V3V 31V339V399 VW9333993
09917 9W33V913991V9933W399V9V1399V 933939V3V3 V999WW31 V3V99V3319
00917 11V331W1V 39V113V9W W333913111313339VW V13V1333991131V9V93V
OVVV 9VW1VWW 31V9V9VW399W9V31V3 V31391199919399W9V3 V1V1VW9V9
088V 91V331V1V333913V1391111391V119339V1V913V 1V11191VW 139W9W19
028V 9W9V3991V 9VW133399 V333V313V39V1V913W91399939V3139331V3V99
092V 9339V33913339VW13999131W199V 933V9V933V 33V919939139V999399V
002V V1939V3V999V9W3V1131319V39W991V993V3V199133V3V33 V93393V9V3
OVIV 33139339V33V993W133 V9333V9333 W993V3339 W933991331V1V911339
080V 13W3V3W3913999W99 V9133913V3331391393V 1399333W913339V1991
020V 9933V993119V33199933 V993V3V99111139113399V31V13V313V91V99333
0968 93W3311139913933V9333333W3W V319V13V399V9V19V391 V333333991
0068 311V319V13 V339V3V9991V33W139V V399V391V99VW3V913V 1933911V33
0V88 9V39V1991V 993V13391133333991999139V39V19 V333V9V139 VWW33191
08Z8 33V1339919933993193991133V99913WVW393V V13319199V W3393V139
02Z8 99V39W9V93V9319111313W91113339W13V9111V9V33911999913339V3
0998 333913W13113139W39 W31VW9W 31V13399W W3W33V9V 333999911V
0098 V11191313V 99993WW33V3131333V 1911333339 V3991VW9V 3911199913
0V98 331V991313393191311399V3993V999V133119V999W13W3V 9313333V9V
08V8 V93313V1339V399991V91913V9V9W W9V3399V913V913991V 9V9V31999V
02V8 9WW131131V19999131 V19W3199V 3VW9V339111VW3339V W9W33993
0988 13399931V19993133W999V1333VW 3V1191333999313V199V V3W33913V
0088 9V139V913113V339339913913V113V 91V99193V19V3V1391331V9113V9V3
ZV
8 l/86S_T/lDd Z6ZI 1/66 OΛV 0V99 1V999111V3 V113999V331V9W3V993313191V1911193V113999913V11V3V
08V9 99V3V33V99133113V119993V9V9993393V9V3V11199311V119 V113333W3
02V9 919919W1V V1V9VW391 V199V33191399V33V913133V3131W 3W3VW19V
0989 3V31V3111V 9991V31V313313339W9913V1139V9 V19933W3V 9V91913999
0089 911V13391913V13113313V9V33199999191V319V V331W9V933333933V33
0V29 339993333911191V1111 V999V91VW 31VW1V313 W3V9V33V9 V139W313V
0819 9V3W99133913V3W39199313333V31333133W11133W9W93913V9V9V3
02T9 31191399V3339V39V399999V313911911333339999333331311311111333
0909 31W31V1W 9V139999911V1131V33V 991V33V3339V1191V191 V111V9V333
0009 3V1133V913399199133V 9919131333 V33W39913113VV39991 V1993V9V99
0V69 931V9V991V V33W199V999133V31V1 W1V1319W 31V313319V 3313993339
0889 3113913V19 V319999V9V 31391V91311311W1133331W13333399V939333
0289 W1199W1V WW113333 WW3131393W3119393991V3V913V 9V131331V3
09Z9 3V991999993333V93391399W9193V 33393393V3 V1V991139V 3931V3993V
00Z9 9V19VW31333933V33333V33V913913319V3V3V1133V99VW991393133W
0V99 9V133W9W 13V9V33V3V 93393319991919V3V3V9399319V933 V11333V313
0899 3V19919933 V933V9913V V3W9W33V 1339V39939913133V9V991319W93V
0299 39V3319V113V13131399 V3V113V313 9W331313133339V3W13V119V9W3
09V9 V91V3V91333V91333113 VW19113333933333V399991V1V1131V9V91V1V3
00V9 333V3133991V333339993393V3W393339V933V1913339V113 33313V1331
0V89 39199913V9 V9V13139913W3911393 W11VW13V V1113V9V99 W31V33W9
0829 V1W91W9V W9V199V3399V3139VW 3333V9V3V1 V391911V3V 11VW9911V
0229 911V9999119131V9339919V3V9V319 V9199W3313193113391339991W3V
0919 913VV99911 V199V313391V39931199 V33331131V 9V99V9V13V 139W9W33
00T9 W193199W 33933VW9V W9W33W33311339W9 V1V99139913111133V1V
0V09 9V1V111119 V131131V1V W1V13991V 1911V993339VW1V9V933V3111V931
086V V9V99911V313V3993339931V399939331999V13V V999V3VW1193391319V
026V V39V3393W 319W3V39191139VW39133V9V913V 31V1VWW313V3W931V
εv
80€8T/86Sri/X3d Z6ZIΪ/66 OΛV 0918 91VW9133V 99W333391999V33JJL191V9V31V33V V9V9V131119V39V31333
00T8 9V33199391 V9V33331991V9V3W9W 33999V3139933339133119V39W199
0V08 19131V1V99 V3VW339991V1W9139V 3W991V9V3 W9W33999 V313993333
086Z 9133119V39 W19919131 V1V99V3VW 339991V1W 9139V3W991V9V3W99V
026Z 3199W31V9 V3119W9V9 V1W9V913V V1V3V1WW V991V399W 391111V339
098Z 3W19W1139V139W39911199V19133V3333V9W V91W999999VWW9V33
008Z 1319V111V1111V9VWV1 WW1V9V1V 339V93V19V 9V1V1339W 9139V33V31
OVZZ V1W3W313 V911119V131399V3319919V31V1V99 V3V9VW119 LL1W3319V
089Z 11V931W11311V391333 V9931131391111V91W13313V1911V 313333V999
029Z 1V11V33V131V1V91133V 33V1LL991133331V9V3V V111913V999V9111991V
099Z 99W313W319V9111913 VW9V3V9V9 V31W11399 V9V9V9V911 VW33991V3
009Z 9V3V9V9V919V13V99V3V 3V33V99391 V1311391191W9W9VW V131391391
OVVZ 91913999V99W9VVW13 _L_U.V_U.9il3 V9V1339999 W931W9V3 V13319119V
088Z V913191333113V3131319WW9V133 W13131W31VWW91199V999V3131
028Z V91V99V3V19V39339W33139V39V3311W39V313 V33991W1313913V13V9
092Z 99V3W99V1 W9913933911W9999991V33W13V99199911V11 V133399133
002Z 3W11931919933W9V9V VW3V3V933 W331V9V9V 91119133993V11191V13
OVIZ 9V3333311V 31V133V319 V9W33991313W93191131191911V11V913V33V1
080Z 133W913V1 V33V33V33131V391V3313V11399933 V19V19113999191V33V1
020Z 99V3V133339119V131V11V1331999V 9319V39W3 V9V333V33V 1W191V133
0969 399V31V3V3 WW333119 V39V99V1V391313V999V 3V9933V919 W93319133
0069 3V9119W3V 3W33313399193313913 W339V3313913133V1V33W3313V13
0V89 3V199913319339119999 W93V13V1333333V99939V19V131919119913919
08Z9 V9W333VW V3V913319V 33V3133W313339W33V 1339V993V9 V19V113VW
02Z9 1391399V1V VW993W99939933V33113W333V13333131313313V99919V3
0999 33V33VW33 V319V311339319W119133VW3339V V331313V339V3W33V9V
0099 399131193333VW33999 V1W33319393333V99V131WW33V1 V9V313V933
VV
80€8I/86Sfl/XDd Z6ZII/66 OΛV ACCCTGTGCC TTATTTGAAC TAACCAATCA GTTCGCTTCT CGCπCTGπ CGCGCGCπC 8220 TGCTCCCCGA GCTCAATAAA AGAGCCCACA ACCCCTCACT CGGGGCGCCA GTCCTCCGAT 8280 TGACTGAGTC GCCCGGGTAC CCGTGTATCC AATAAACCCT CTTGCAGTTG CA 8332

Claims

CLAIMS We claim:
1. A method for treating or preventing arthritis, comprising, administering to a mammal a therapeutically effective amount of a recombinant retrovirus which directs the expression of an anti-arthritic agent, wherein said recombinant retrovirus is administered at a titer of greater than 107 cfu/ml.
2. The method according to claim 1 wherein said arthritis is rheumatoid arthritis.
3. The method according to claim 1 wherein said arthritis is osteoarthritis.
4. The method according to claim 1 wherein said anti-arthritic agent is a pro-drug converting enzyme.
5. The method according to claim 1 wherein said anti-arthritic agent is an inhibitor of tumor necrosis factor.
6. The method according to claim 1 wherein said anti-arthritic agent is a IL-1 Receptor antagonist.
7. The method according to claim 1 wherein said anti-arthritic agent is an inhibitor of matrix metalloproteinase expression.
8. The method according to claim 7 wherein said inhibitor of matrix metalloproteinase expression is TIMP-1 or TIMP-2.
9. The method according to claim 1 wherein said recombinant retrovirus is administered intraarticularly.
PCT/US1998/018308 1997-09-02 1998-09-02 Compositions and methods for treating arthritis utilizing gene therapy WO1999011292A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
JP2000508393A JP2001514235A (en) 1997-09-02 1998-09-02 Compositions and methods for treating arthritis utilizing gene therapy
EP98945862A EP1009444A2 (en) 1997-09-02 1998-09-02 Compositions and methods for treating arthritis utilizing gene therapy

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US5603997P 1997-09-02 1997-09-02
US60/056,039 1997-09-02

Publications (3)

Publication Number Publication Date
WO1999011292A2 true WO1999011292A2 (en) 1999-03-11
WO1999011292A3 WO1999011292A3 (en) 1999-07-01
WO1999011292A9 WO1999011292A9 (en) 1999-08-05

Family

ID=22001771

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/018308 WO1999011292A2 (en) 1997-09-02 1998-09-02 Compositions and methods for treating arthritis utilizing gene therapy

Country Status (3)

Country Link
EP (1) EP1009444A2 (en)
JP (1) JP2001514235A (en)
WO (1) WO1999011292A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003528923A (en) * 2000-04-05 2003-09-30 イーペーエフ ファルマシューティカルス ゲゼルシャフト ミット ベシュレンクテル ハフツング Drug containing a tissue inhibitor of metalloproteinase-2 (TIMP-2) as a bone synthesis metabolic active substance

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0334301A1 (en) * 1988-03-21 1989-09-27 Chiron Viagene, Inc. Recombinant retroviruses
WO1992011359A1 (en) * 1990-12-20 1992-07-09 University Of Pittsburgh Of The Commonwealth System Of Higher Education A truncated interleukin-1 receptor gene for the treatment of arthritis
WO1994020517A1 (en) * 1993-03-08 1994-09-15 University Of Pittsburgh Of The Commonwealth System Of Higher Education Gene transfer for treating a connective tissue of a mammalian host
WO1995016353A1 (en) * 1993-12-14 1995-06-22 University Of Pittsburgh Of The Commonwealth System Of Higher Education Systemic gene treatment of connective tissue diseases
WO1996006941A1 (en) * 1994-08-26 1996-03-07 Hoechst Aktiengesellschaft Genetic therapy of diseases caused by the immune system, said therapy using a cell-specific active substance regulated by the cell cycle
WO1996021014A2 (en) * 1994-12-30 1996-07-11 Chiron Corporation Production and administration of high titer recombinant retroviruses
WO1998000541A2 (en) * 1996-07-03 1998-01-08 Chiron Corporation Methods for administration of recombinant gene delivery vehicles for treatment of human disease

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0334301A1 (en) * 1988-03-21 1989-09-27 Chiron Viagene, Inc. Recombinant retroviruses
WO1992011359A1 (en) * 1990-12-20 1992-07-09 University Of Pittsburgh Of The Commonwealth System Of Higher Education A truncated interleukin-1 receptor gene for the treatment of arthritis
WO1994020517A1 (en) * 1993-03-08 1994-09-15 University Of Pittsburgh Of The Commonwealth System Of Higher Education Gene transfer for treating a connective tissue of a mammalian host
WO1995016353A1 (en) * 1993-12-14 1995-06-22 University Of Pittsburgh Of The Commonwealth System Of Higher Education Systemic gene treatment of connective tissue diseases
WO1996006941A1 (en) * 1994-08-26 1996-03-07 Hoechst Aktiengesellschaft Genetic therapy of diseases caused by the immune system, said therapy using a cell-specific active substance regulated by the cell cycle
WO1996021014A2 (en) * 1994-12-30 1996-07-11 Chiron Corporation Production and administration of high titer recombinant retroviruses
WO1998000541A2 (en) * 1996-07-03 1998-01-08 Chiron Corporation Methods for administration of recombinant gene delivery vehicles for treatment of human disease

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BANDARA G ET AL: "GENE TRANSFER TO SYNOVIOCYTES: PROSPECTS FOR GENE TREATMENT OF ARTHRITIS" DNA AND CELL BIOLOGY, vol. 11, no. 3, 1 April 1992, pages 227-231, XP000577115 *
EVANS C H ET AL: "GENE TRANSFER TO JOINTS FOR ARTHRITIS THERAPY" JOURNAL OF CELLULAR BIOCHEMISTRY, no. SUPPL. 16F, 1 January 1992, page 46 XP000566580 *
HUNG G L ET AL: "SUPPRESSION OF INTRA-ARTICULAR RESPONSES TO INTERLEUKIN-1 BY TRANSFER OF THE INTERLEUKIN-1 RECEPTOR ANTAGONIST GENE TO SYNOVIUM" GENE THERAPY, vol. 1, no. 1, 1 January 1994, pages 64-69, XP000671119 *
ROBBINS P D ET AL: "RETROVIRAL VECTORS FOR USE IN HUMAN GENE THERAPY FOR CANCER, GAUCHER DISEASE, AND ARTHRITIS" ANNALS OF THE NEW YORK ACADEMY OF SCIENCES, vol. 716, 31 May 1994, pages 72-89, XP000565748 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2003528923A (en) * 2000-04-05 2003-09-30 イーペーエフ ファルマシューティカルス ゲゼルシャフト ミット ベシュレンクテル ハフツング Drug containing a tissue inhibitor of metalloproteinase-2 (TIMP-2) as a bone synthesis metabolic active substance
JP4939715B2 (en) * 2000-04-05 2012-05-30 イーペーエフ ファルマシューティカルス ゲゼルシャフト ミット ベシュレンクテル ハフツング Agents containing tissue inhibitors of metalloproteinase-2 (TIMP-2) as osteosynthesis metabolic active substances

Also Published As

Publication number Publication date
JP2001514235A (en) 2001-09-11
EP1009444A2 (en) 2000-06-21
WO1999011292A3 (en) 1999-07-01
WO1999011292A9 (en) 1999-08-05

Similar Documents

Publication Publication Date Title
EP0702084B1 (en) Recombinant retroviruses
Brown et al. A retrovirus vector expressing the putative mammary oncogene int-1 causes partial transformation of a mammary epithelial cell line
US8034620B2 (en) Lentiviral packaging cells and uses therefor
Blömer et al. Highly efficient and sustained gene transfer in adult neurons with a lentivirus vector
JP3150340B2 (en) Bifunctional selectable fusion gene
US6761884B1 (en) Vectors including foreign genes and negative selective markers
US6485965B1 (en) Replicating or semi-replicating viral constructs, preparation and uses for gene delivery
Hafenrichter et al. Quantitative evaluation of liver-specific promoters from retroviral vectors after in vivo transduction of hepatocytes
US5747323A (en) Retroviral vectors comprising a VL30-derived psi region
WO1996021036A2 (en) Nucleic acid condensing agents with reduced immunogenicity
Wilson et al. Sustained expression of α1-antitrypsin after transplantation of manipulated hematopoietic stem cells
EP0632129B1 (en) Recombinant foamy virus vectors for medicinal and diagnostic uses, and processes for preparing recombinant foamy virus vectors
JP3959117B2 (en) Generating retroviral vectors for gene therapy
US20070077233A1 (en) Inhibition of pathological angiogenesis in vivo
US7056730B2 (en) Expression of heterologous genes from an IRES translational cassette in retroviral vectors
JP3908274B2 (en) Vector having a therapeutic gene encoding an antimicrobial peptide for gene therapy
KR20010015624A (en) Self-deleting Vectors for Cancer Therapy
JP2001500021A (en) Novel internal ribosome entry site and vector containing it
EP1009444A2 (en) Compositions and methods for treating arthritis utilizing gene therapy
Milan et al. Activator-dependent and activator-independent defective recombinant retroviruses from bovine leukemia virus
EP1059356B1 (en) Replicating retroviral constructs, preparation and uses for gene delivery
JPH10510163A (en) Packaging cells and expression vectors for transcomplementation of deleted retroviral vectors
DE69829174T2 (en) EXPRESSION OF A MODIFIED "FOAMY VIRUS ENVELOPE PROTEIN" (SLEEP PROTEIN)
JPH11508441A (en) Novel recombinant DNA vector for gene therapy
KR100353260B1 (en) A triple fusion genes carrying drug resistance gene, green fluorescent protein gene and drug-sensitizing gene and gene expression vector harboring the gene

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): CA JP

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): CA JP

AL Designated countries for regional patents

Kind code of ref document: A3

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

AK Designated states

Kind code of ref document: C2

Designated state(s): CA JP

AL Designated countries for regional patents

Kind code of ref document: C2

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

COP Corrected version of pamphlet

Free format text: PAGES 1-38, DESCRIPTION, REPLACED BY NEW PAGES 1-35; PAGE 46, CLAIMS, REPLACED BY NEW PAGES 36 AND 37; PAGE 1/1, DRAWINGS, REPLACED BY NEW PAGE 1/1; PAGES 39-45, SEQUENCE LISTING, REPLACE BY NEW PAGES 1-6; DUE TO LATE TRANSMITTAL BY THE RECEIVING OFFICE

ENP Entry into the national phase in:

Ref country code: JP

Ref document number: 2000 508393

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1998945862

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1998945862

Country of ref document: EP

NENP Non-entry into the national phase in:

Ref country code: CA

WWW Wipo information: withdrawn in national office

Ref document number: 1998945862

Country of ref document: EP