WO1990012087A1 - Particule virale infectieuse ciblee a replication defectueuse - Google Patents

Particule virale infectieuse ciblee a replication defectueuse Download PDF

Info

Publication number
WO1990012087A1
WO1990012087A1 PCT/US1990/001867 US9001867W WO9012087A1 WO 1990012087 A1 WO1990012087 A1 WO 1990012087A1 US 9001867 W US9001867 W US 9001867W WO 9012087 A1 WO9012087 A1 WO 9012087A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
gene
cells
genes
virus
Prior art date
Application number
PCT/US1990/001867
Other languages
English (en)
Inventor
Daniel R. Henderson
Original Assignee
Novacell 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 Novacell Corporation filed Critical Novacell Corporation
Publication of WO1990012087A1 publication Critical patent/WO1990012087A1/fr

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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70514CD4
    • 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
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13041Use of virus, viral particle or viral elements as a vector
    • C12N2740/13043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2740/13041Use of virus, viral particle or viral elements as a vector
    • C12N2740/13045Special targeting system for viral vectors
    • 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
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/40Vectors comprising a peptide as targeting moiety, e.g. a synthetic peptide, from undefined source
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/80Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/80Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates
    • C12N2810/85Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian
    • C12N2810/854Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian from hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/80Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates
    • C12N2810/85Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian
    • C12N2810/855Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian from receptors; from cell surface antigens; from cell surface determinants
    • 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
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/50Vectors comprising as targeting moiety peptide derived from defined protein
    • C12N2810/80Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates
    • C12N2810/85Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian
    • C12N2810/859Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian from immunoglobulins

Definitions

  • This invention relates to a binary vector, as well as methods for its preparation and use, comprising a packaging cell and a replication defective virion capable of introducing a cell growth inhibiting gene into a target mammalian cell.
  • antineoplastic or antiviral agents have varying degrees of specificity for tumor cells or virus-infected as opposed to normal cells for inhibiting the growth of and/or killing of tumor cells or viruses within virus-infected cells.
  • hormonally-related compounds such as hexestrol or cytotoxic agents such as methotrexate and nitrogen mustards have found use for treating cancer; drugs specific for inhibiting viral replication, such as AZT have found use for treating viral diseases.
  • drugs specific for inhibiting viral replication, such as AZT have found use for treating viral diseases.
  • the therapeutic index for these drugs tends to be narrow and the side effects of these treatments may be severe.
  • Essential to the success of inhibiting growth of or killing of a specific target cell in vivo is the introduction of the kill gene into the target cell under conditions whereby the gene may be expressed.
  • DNA and RNA as such is not taken up well by cells; by contrast, DNA and RNA in the form of virus particles is taken up very efficiently, mediated by receptors for the particular virus.
  • Special cells have been constructed that allow the preparation of virus particles capable of mediating the transfer of genes to target cells. These cells supply virus proteins necessary for the assembly of virus particles.
  • the genome, RNA or DNA, packaged into virus particles by these special cells can contain any gene; in this application a kill gene. The genes encoding virus proteins have been removed from the packaged virus genome. Thus, the virus particles produced cannot replicate.
  • the special cells so-called packaging cells, have evolved for both DNA and RNA viruses from constructs requiring an additional virus, a helper- virus, to sophisticated cells no longer requiring helper-viruses, to support virus growth.
  • Replication deficient helper-free RNA tumor viruses produced by packaging cells have been used to transform cells derived from the human reticulo- endothelial system. These viruses and their packag- ing cells have been derived from two strains of murine leukemia virus, Moloney murine leukemia virus (MoMuLV) that infects rodent cells, and strain 4070A that infects both murine and human cells.
  • Human reticuloendothelial cells genetically engineered with retroviruses have been proposed for correction of enzyme defects: gene therapy.
  • a bone marrow transplant In preparation for a transplant, the patient must be given a lethal dose of x-irradiation in preparation for the graft, to remove the resident bone marrow cells. Thus a bone marrow transplant involves significant expense, and uncertain survival.
  • Virus packaging systems have also been used to transfer genes into skin fibroblasts. Transformation of skin fibroblasts offers the advantage that engineered genes can be introduced into humans without the need for lethal whole body x-irradiation.
  • DNA viruses such as SV40, a papovaviruses, COS-7 cells have been used to construct helper-free, infectious, but replication defective SV40 virus (Gluzman, 1981).
  • COS-7 cells contain an SV40 genome without the ori, or origin of replication.
  • the cells produce SV40 T antigen, allowing for replacement of SV40 T antigen in the infecting virus genome with other desired genetic information.
  • COS-7 cells are inadequate packaging cells.
  • Early region SV40 gene replacement vectors (Tantigen replacement), expressed in COS-7 cells yield high levels of replication-deficient infectious helper- free virus, but also produce replication-competent infectious virus by homologous recombination with the endogenous viral genome.
  • Late region SV40 replacement vectors (replacing VP1, VP2 or VP3), require helper virus to be packaged and infectious.
  • the COS-7 cells are not capable of sufficient expression of the late genes, VPl, VP2 and VP3 to yield infectious virus.
  • Enhancers/promoters are DNA elements that bind certain regulatory proteins only found in specific cell types. This binding leads to cell-specific transcription of genes. Enhancers/promoters have been described for B cells, T cells, liver cells, liver cancer cells, HIV-1 infected cells, and many other tissues/organs. Thus coupling of tissue-organ- specific enhancers/promoters to toxin genes can kill specific cell types.
  • Retrovirus packaging cells capable of infecting human cells were made by incorporating the env gene from murine leukemia virus strain 4070A and the gag and pol genes from MoMuLV has been disclosed. These virus particles have been used to infect human reticuloendothelial cells, human hepatocytes and human keratinocytes. I.M. Ver a, A. Miller and R.M. Evans, W08600922, filed July 29, 1985 and I.R. Morgan and R.C. Mulligan, W08700201 filed June 30, 1986.
  • Virus particles nonspecifically packaging host cellular nonviral RNA in the presence of helper viruses is described in Linial, J. Virol (1981)
  • RNA tumor viruses have been used to establish cell lines producing fibronectin (Schwarzbauer, et al. , Proc Natl Acad Sci USA (1987) 4:754); Class II human major histocompatibility antigens (Kor an et al. , Proc Natl Acad Sci USA (1987) 84.:2150); growth hormone (Morgan, et al. , Science (1987) 237:1476) ; adenosine deaminase (Palmer, et al. , Proc Natl Acad Sci USA (1987) 8£:1055); Factor IX (St.
  • Packaging cells have also been isolated for adenoviruses (Shiroki and Shimojo, 1971; Graham et al. , 1977; and Grodzicker and Klessig 1980). Using these packaging cells, host-range mutants of Ad5 (Harrison et al. , 1977), deletion mutants of Ad5
  • packaging cells for Herpes Simplex virus have been isolated (Kimura et al. , 1974; Maenab and Timbury, 1976). Using Herpesvirus packaging cells. ts mutants of Herpes Simplex virus type 2 (Kimura et al. , 1974; Macnab and Timbury 1976), and Herpesvirus expression vectors of foreign genes (Roizman and Jenkins, 1985; Thomsen et al. , 1987) have been prepared.
  • Promoters and enhancers regulate genes that encode messenger RNA in mammalian cells. Promoters are located immediately upstream, 5', from the start site of transcription. Enhancers: increase the rate of transcription from promoters, act on cis-linked promoters at great distances, are orientation independent, and can be located both upstream, (5'), and downstream, (3'), from the transcription unit. Enhancers were first described for viruses. Later, enhancers indueible by hormones and metal ions and found only in specific tissues were described (Maniatis, T., S. Goodbourn, and J.A. Fisher. 1987, Science 236:1237-1245) . Proteins synthesized only on one tissue type are frequently regulated by tissue specific enhancers. For example, actin and myosin in muscle; lens crystalline in the eye. For the tissue specific expression of kill genes, tissue-specific enhancers are of particular interest.
  • B cell and T cell enhancers are the best characterized tissue-specific enhancers.
  • Im unoglobins are only synthesized in B cells.
  • IgG heavy and light chain enhancers have been found within the introns of both the mu heavy-chain a kappa light-chain genes, each with a minimal functional region of about 140 base pairs (Gillies, S.D., S.L. Morrison, V.T. Oi, S. Tonegawa, Cell 33:717-728) . These enhancers function only in B cells, not in T cells, colon cells, and other tissue types tested.
  • the immunoglobin chain enhancers are functional in B cells because enhancer binding proteins, only present in B cells, activate transcription from genes cis to the enhancer. Similarly, the T cell specific T cell receptor (TCR) is only found on T cells.
  • T cell enhancers the alpha chain enhancer (Winoto, A., and D. Baltimore, 1989, EMBO J.£:729-733) , and the delta chain enhancer (Redondo, J.M. , S. Hata, C. Brocklehurst, and M.S. Krangel. 1990. Science 247:1225-1229).
  • liver specific enhancers have been found for genes exclusively expressed in liver. These include: alphafetoprotein, transthyretin, alpha 1- antitryptsin, fibrinogen, and aldolase (Courtois, G., J.G. Morton, L.A. Campbell, G. Fourel and G.R. Crabtree, 1987, Science 238:688-692; Feuerman, M.H., R. Godbout, R.S. Ingram, and S.M. Tilghman. 1989, Molec. Cell Biol.9_:4204-4212) .
  • a novel replication-defective binary vector derived from an animal virus, conveniently a retro- virus or a papovavirus, together with methods for its preparation and use, is provided for introducing and expressing a kill gene in a mammalian target cell.
  • the binary vector comprises (1) a packaging cell containing in its chromosome a nucleic acid sequence encoding a trans acting virus polypeptide required for the maturation of new virus particles and (2) a mobile genetic element comprising a modified genome in which at least one kill gene has replaced the trans acting virus function induced by the packaging cell.
  • the modified genome of the mobile genetic element would contain a ⁇ sequence or an ori for a retrovirus and a papovavirus, respectively.
  • a modified ligand-receptor interaction introduced into the mobile genetic element by expression of an altered virus surface receptor moiety gene in the packaging cell, or by covalent attachment of one member of a specific binding pair to the virion, is used to direct the mobile genetic element to the target cell.
  • the packaging cell is grown to produce the mobile genetic element.
  • the kill gene is transferred to the target cell directly by infection with the mobile genetic element.
  • the packaging cells may produce the mobile genetic element in vivo to provide sustained release of the mobile genetic element which then infects the target cell. Tissue specific expression of the kill gene may be achieved by optional by introducing additional DNA sequences such as tissue specific enhancers/promoters to control expression of the kill genes.
  • Figure 1 shows the structure of the packaging cellpCRIPenv-.
  • Figure 2 shows the structure of the MoMuLV provirus in pNCl.
  • Figure 3 shows the structure of the Ndel-Nsil insert of pUC19-env-.
  • Figure 4 shows the structure of plasmids pNCSH-
  • Figure 5 shows the structure of pNC-CD4.
  • Figure 6 shows the three constructs having a second copy of gag, pol and env.
  • Figure 7 shows the structure of pNC-HSV-tk(2) .
  • Figure 8 shows the structure of pNC-HSV-AS.
  • Figure 9 shows the structure of pNC-HSV-X, where X is a protease, ribonuclease or DNAse.
  • Figure 10 shows the genetic map of SV40.
  • Figure 11 shows the three constructs for VP1,
  • Figure 12 shows the structure of pNCHSVkill.
  • Figure 13 shows the structure of pNCSVl.
  • Figure 14 shows the structure of the Diphtheria A tox insert.
  • Figure 15 shows a schematic representation of the SV40 and LPV genomes.
  • Figure 16 shows the structure of PNCLV2, and
  • Figure 17 shows the structure of a production minichromosome.
  • Figure 18 shows the structure of a T cell enhancer/promoter Dyphtheria A toxin.
  • Figure 19 shows the structure of the HIV-1 enhancer/promoter Dyphtheria A toxin.
  • Figure 20 shows the structure of the Hepatitis B virus enhancer/promoter Dyphtheria A toxin.
  • a novel binary vector, and methods of preparation and use, is provided for the introduction into a specific mammalian host cell, particularly a human cell, of a kill gene capable of transcription and expression in said host cell.
  • the binary vector comprises a packaging cell and a mobile genetic element.
  • the host range for cell attachment of the vector is determined by modification of the cell surface ligand-receptor interaction of the mobile genetic element of the vector. Modifications in cell attachment can be accomplished by incorporation of a modified virus surface receptor moiety gene, such as a retrovirus env gene or the VP1 gene of a papovavirus into the packaging cell.
  • the virion surface receptor moiety may be coupled to a targeting means comprising one member of a specific binding pair (sbp) such as an antibody to a cell surface antigen or a peptide hormone so that the vector binds to the cell surface antigen or peptide hormone receptor which comprises the other member of the sbp on the target cell.
  • a targeting means comprising one member of a specific binding pair (sbp) such as an antibody to a cell surface antigen or a peptide hormone so that the vector binds to the cell surface antigen or peptide hormone receptor which comprises the other member of the sbp on the target cell.
  • a targeting means comprising one member of a specific binding pair (sbp) such as an antibody to a cell surface antigen or a peptide hormone so that the vector binds to the cell surface antigen or peptide hormone receptor which comprises the other member of the sbp on the target cell.
  • Inhibition of the growth of or killing of the target host cell is provided by introduction of a
  • the packaging cells are grown to produce the mobile genetic element containing the minichromosome which is used to infect target mammalian cells under appropriate conditions for transfer of the minichromosome containing the nucleic acid sequence of interest into the target cell.
  • the result is inhibition of cell growth or killing of the target cell.
  • Tissue specific expression of the kill gene further narrows the specificity of target cell killing.
  • Tissue specific gene expression is achieved by the use of highly specific enhancers/promoters; that is DNA sequences to which are bound proteins that only exist in certain types of cells. These proteins are activators of transcription that regulate the expression of certain genes that are therefore expressed only in these cells.
  • tissue/organ-specific enhancers/promoters provides the opportunity for targeting anti-cancer and anti-viral agents to the specific organ or cell type where disease has arisen. This is done by (a) introducing into cells genes that encode toxic or potentially toxic proteins, and (b) controlling the expression of such genes with highly specific enhancers/promoters.
  • An object of the invention is to produce a binary vect ⁇ r capable of transmitting antiproliferative or cytotoxic genetic material, which will be referred to generally as a kill gene, in a one-way nonreversible manner to a specific target cell.
  • kill gene is intended a nucleic acid sequence which provides for inhibition of cell growth or cytotoxicity in any of a variety of ways, including the following.
  • the kill gene may affect an essential cell function directly, for example, by providing for synthesis of an anti-sense transcript of an essential cell component or of an oncogene.
  • the kill gene may also encode a polypeptide capable of directly inhibiting cell growth such as Diphtheria toxin or one which may catalyze formation of an inhibitory or toxic compound, such as, for example Herpes virus thymidine kinase (HSV-tk) which is part of a cascade to form acylo-GTP from acyclovir.
  • HSV-tk Herpes virus thymidine kinase
  • the binary vector consists of: (1) a packaging cell encoding a trans acting polypeptide(s) required in the maturation of new virus particles, and (2) a mobile genetic element comprising an infective yet replication incompetent virion and a minichromosome.
  • the binary vector is prepared as follows. A virus gene encoding the trans acting viral function(s) is removed from the wild-type virus genome and inserted into the chromosome of a packaging cell that is otherwise permissive for the virus.
  • packaging cell is intended a genetically constructed mammalian tissue culture cell that produces the necessary virus packaging proteins. The genetic material for the virus packaging proteins is not transferred with the mobile genetic element, hence the virus cannot replicate.
  • the virion produced by the packaging cell contains a modified genome or minichromosome in which genes designed to inhibit the growth of, or to kill, the target cell have replaced the trans acting function that has been incorporated into the chromosome of the packaging cell.
  • Packaging cells may be any animal cells permissive for the virus, or cells modified so as to be permissive for the virus; or the packaging cell may be one which is made permissive for the proviral construct, for example, with the use of a transforma- tion agent such as calcium phosphate.
  • Cell lines which can be used as packaging cells for retroviruses include rodent cells such as NIH/3T3 cells and other murine cells; a suspension cell line such as Chinese Hamster Ovary cells (CHO) or L929 cells; for papovavirus, cells such as BSC-1 or VERO cells from African Green Monkeys, or human cells, such as WI38; and for adenoviruses, KB cells and HeLa cells.
  • rodent cells such as NIH/3T3 cells and other murine cells
  • a suspension cell line such as Chinese Hamster Ovary cells (CHO) or L929 cells
  • for papovavirus cells such as BSC-1 or VERO cells from African Green Monkeys, or human cells, such as WI38
  • adenoviruses KB cells and HeLa cells.
  • packaging cells are cells, particularly human cells, deficient in glucocorticoid receptors.
  • the packaging cell capable of producing the desired replication-defective mobile genetic element at least a first and a second construct will be transformed into a packaging cell, the constructs being as exemplified below for a retrovirus.
  • the first construct, the mobile genetic element is prepared having the following components in the order of transcription:
  • P.S. intends a viral packaging sequence;
  • V.G. intends a trans acting gene, and
  • G.I. intends a kill gene and wherein the components of the construct are operably linked.
  • a second construct, and optionally third and fourth constructs, used to construct the packaging cell comprises the following components in the direction of transcription, a transcriptional initiation region, at least one of a gag, pol and an env gene and a transcriptional termination region, wherein the components are operably linked.
  • the general structure of these constructs is as follows: (5'-LTR) a —(P)b- (V.G.)c— .R wherein 5'LTR and V.G.
  • V.G. additionally may comprise a single copy or a plurality of copies of the same trans acting gene, for example, gag gag or pol pol.
  • the second construct will provide at least one copy of each of the viral gag and pol genes without the packaging ⁇ sequence and the 3'LTR.
  • a third construct will comprise a modified env gene without the ⁇ sequence and 3'LTR.
  • a fourth construct will provide at least one copy of the gag or pol gene, without the ⁇ sequence and the 3'LTR.
  • the desired replication defective virion produced by the packaging cells will have a genome containing the 5'LTR and ⁇ sequences, a kill gene and the 3'LTR.
  • the constructs which provide the ⁇ packing sequences and the gag, pol and modified env genes may be derived from any animal retrovirus, particularly Moloney murine leukemia virus, an avian leucosis virus, or an HTLV virus. Analogous constructs to those described above can be prepared for binary vectors employing papovavirus, where gag, pol and env are replaced with T-antigen VP1, VP2 and VP3.
  • the promoter is preferentially an inducible enhancer/promoter such as the metalothionein promoter, or murine mammary (MMTV) promote .
  • the preferred method for preparation of the packaging cells is to introduce at least two mutant viral genomes into the packaging cell, most prefer ⁇ ably three. This containment scheme will effectively prevent homologous recombination leading to reconstruction of infectious virus.
  • the probability of recombination occurring if two trans acting virus genes are used is less than 1 x 10-14; with the use of three separate mutant virus genes, the probability of recombination reconstruction of infectious virus is less than 1 x 10-21.
  • the gag, pol and env genes are transformed into the packaging cell on three separate plasmids, preferably sequentially, in any order, by any convenient means, for example, by the calcium phosphate precipitation protocol.
  • vectors such as those prepared from papovavirus and patterned after SV40, the SV40 ori, with or without the large T antigen could be maintained in the mobile element of a binary vector.
  • T antigen can also be supplied by the packaging cell, but the ori must be present in the mobile genetic element. If T antigen is present in the mobile genetic material, the minichromosome will replicate to high copy number in the target cell, increasing expression of the kill gene(s).
  • the minichromosomes will not replicate in target cells, papovavirus vectors patterned after SV40 are episomal and transitory in nature. Therefore, the kill genes encoded by the minichromosomes must be sufficiently lethal to kill the cell in an active manner. This restricts the kill genes to activities such as RNAse, DNAse, and protease. Since these vectors are transitory, they are particularly attractive for use in humans as they do not permanently genetically engineer the host cell.
  • high producing packaging cells are identified by screening the transfectants, for example by immunoassay, by PAGE, and the like, for packaging cells making quantitatively more gag, pol and env, or T antigen VP1, VP2 and VP3 expression products.
  • the high producing packaging cells may be transfected using the provirus construct comprising the kill gene.
  • the resulting transfectants may be selected, as described above, and the so-obtained packaging cells grown to express the viral genes and produce the desired substantially replication-defective virions.
  • the object is to produce a binary vector capable of transmitting genetic material to the target cell in a one-way nonreversible manner and for the gene of a trans acting polypeptide to be incorporated into the chromosome of a host cell otherwise permissive for the virus.
  • the chromosomally integrated gene(s) within the packaging cell must not be capable of being mobilized into super-infecting viral genomes, yet the minichromosomes must be capable of producing infectious virus.
  • Analogous vectors can also be derived from DNA viruses such as Adenovirus type, Herpesvirus, and Papovaviruses, such as SV40, lymphotropic papovavirus (LPV), BK virus (BKV) and JC virus (JCV) .
  • SV40 lymphotropic papovavirus
  • BKV BK virus
  • JCV JC virus
  • the minichromosome (the mobile DNA) packaging into the mobile genetic element also called pseudovirions
  • the minimum requirements for a construct are an ori and a circular genome within about 75-102% of 5243bp.
  • COS-7 cells can complement necessary trans acting genes, allowing for construction of helper-free, infectious, but replication defective SV40 virus (Gluzman, 1981).
  • COS-7 cells produce SV40 T antigen, thus the SV40 T antigen in the virus genome can be replaced with other genetic material.
  • the genome and distribution of trans acting genes of SV40, LPV, BK and JC viruses is well established.
  • site specific mutagenesis to affect the cell type of virus attachment as controlled by the outer capsid proteins could be readily evaluated.
  • DNA viruses in particular papovaviruses, may offer advantages over retroviruses as a source of mobile genetic material.
  • the best studied DNA virus, SV40 has many desirable features that would ease virus purification, chemical coupling procedures, manufacturing and quality control: the virus grows to titers of 10 8 to 109 pfu/ml in packaging cells o such as COS-7 and CMT4; the virus is stable at 4 C; the infectivity of the virus withstands harsh treatment such as deoxycholate and cesium chloride; fully packaged infectious virions can be readily isolated from defective empty capsids in cesium chloride; the virus does not cause disease in humans; SV40 minichromosomes are invariably lost from infected cells without positive selective pressure; T antigen mutants that are transformation-) yet replication(+) can be used to support SV40 minichromosome replication avoiding concerns of T antigen induced tumors; SV40 minichromosomes are genetically stable; and cell killing genes can be controlled by the early SV40 promoter which is active
  • papovavirus minichromosomes In considering ablative virus therapy, the transient expression of papovavirus minichromosomes represents a significant improvement in the concerns involved in genetic engineering of humans.
  • the goal in ablative virus therapy is to specifically affect a cell type without side effects or lasting residual genetic interference with the human chromosome. If the amphotropic retroviruses deliver proviruses to and transform the majority of human cells encountered, this goal is not accomplished. Many cells are needlessly permanently genetically engineered. With the use of, for example, papovavirus minichromosomes patterned after SV40, the tissue specific expression of genes can still be accomplished, yet no permanent genetic engineering of nontarget cells would occur. The viral therapeutic is metabolized and dilutes out in time as any conventional pharmaceutical product.
  • the host range of a virus generally is partially determined by a portion of the virus surface receptor moiety on the surface of the virion.
  • Virus host range is further defined by the unique molecular biology of the infected cell by enhancers/p romoters controlling gene expression. Some viruses attach to a specific cell type.
  • the virus produced by the packaging cell will also attach to a specific cell type, by use of a natural surface receptor moiety; it can also attach to cells using a receptor moiety which has been altered.
  • a glycoprotein receptor moiety may be provided, the receptor comprising one member of an sbp: The glyco ⁇ protein. gene can be modified so that the produced virion may be directed to a specific cell type.
  • Specific amino acid sequences such as, for example, the CD4 sequence that recognizes HIV gpl20 can be fused to the env gene of a retrovirus or the VPl gene of a papovavirus using methods known to those skilled in the art.
  • Gene fusions comprising heavy chain antibodies for the F(ab')2 fragment of a tumor- specific antigen fused to the constant region of the env gene or the VPl gene can also be used as a targeting means.
  • the fused gene can be inserted into an appropriate plasmid for transformation into the packaging cells. In this case the specific antibody light chains must also be cloned into the packaging cell.
  • virus surface receptor glycoprotein can be modified for example, by intro ⁇ ducing a mutation to give a lysine, histidine, or cysteine residue in the protruding portion of the glycoprotein yielding an amino group, a histidine free for coupling by organic chemical means, such as a sulfhydryl group.
  • Cell-specific reagents such as F(ab , )2 fragments of specific antibodies such as those to pancreatic cancer or stomach cancer, other tumor-specific antigens or membrane receptors can be coupled with glutaraldehyde, periodate, or maleimide compounds, or by other means known to those skilled in the art to the altered virus.
  • Such couplings may also be made directly to wild-type or unmodified virus env gene glycoprotein products, where coupling can be to a carbohydrate moiety, a sulfhydryl group, an amino group, or other group which may be available for binding.
  • Examples of other ligands which can be coupled by this method include the antibody to HIV gpl20 to target HIV infected lymphocytes, soluble CD4 protein to target HIV infected cells, transferrin to target transferrin receptors, and other members of sbps such as gastrin, secretin or cholecystokinin to target their respective cell receptors.
  • a preferred method of coupling the targeting means to the env gene or VPl gene encoded glycoprotein is to couple sulfhydryl groups of the hinge region of an antibody to the retrovirus envelope glycoprotein or the papovavirus VPl.
  • Other coupling groups include carbohydrate couplings and amino group couplings.
  • an antibody, or the immunoreactive Fab' or F(ab')2 portions of an antibody, particularly a monoclonal antibody (MAb) could also be coupled directly to the surface of the virus particle using methods well known to those skilled in the art.
  • the virion-MAbs conjugate reaches the endosomal compartment of the target cell, the virion must proceed into the normal virus infection pathway.
  • the covalently attached MAb may or may not hinder these processes. It is also possible to target a specific cell type by using the natural selectivity of a particular virus. For example, several papovaviruses exhibit a narrow host range. LPV infects only lymphocytes. Similarly, BKV infects only kidney and liver cells. JCV only infects glial cells.
  • LPV could be a candidate to deliver a mini chromosome to B lymphocytes.
  • LPV derived kill vectors can be envisioned to treat B cell disorders such as B cell leukemia.
  • a mutant of LPV has been isolated that also replicates in T lymphocytes. (Kanda, et al. J. Virol (1985) 55:96-100). This mutant LPV, LPV-76, has been sequenced, and the change is responsible for extending the host range of LPV-76 are three amino acid changes in the VPl.
  • LPV-76 or more specifically the VPl of LPV-76 would direct the virus vector exclusively to B and T lymphocytes.
  • the use of the tat responsive HIV-1 LTR would provide a 1,000-fold enhancement of transcriptional activity in HIV-1 infected cells as compared to non-HIV-1 infected cells of an appropriately designed transient expression minichromosome.
  • AIDS and lymphocytic disorders represent therapeutic targets of virus vectors designed as therapeutics.
  • Virus vectors containing minichromosomes can be developed using a papovavirus such as SV40 as follows.
  • An SV40 early replacement vector would be designed where a toxic gene would be controlled by the tat responsive region TAR of the 5'-HIV-l LTR. Thus this region would be transcriptionally silent in cells lacking the 14.5 Kd tat protein.
  • HIV-1 LTR contains binding sites for nuclear factor activated (NFAT-1) (Shaw, 1988), negative regulatory factor (NRF) (Ratner, 1988), Nuclear factor kb (NFkB) (Nabel, 1987; Tong, 1987), Spl (Jones, 1986), TATA- box transcription factor (TFIID) (Garcia, 1987), leader binding protein (LBP) (Jones, et al. Genes Development 2:1101-1114 (1988), and CCAAT transcription factor (CTF) (Jones, et al. (1988) supra) , and CCAAT transcription factor (CTF) (Jones, et al. (1988) supra; for review see Peterlin, et al. (1988) Molecular Biology of HIV, AIDS 2t (suppl. 1): 529-540. TAR found in mRNA transcribed from position
  • HIV-1 LTR -350 to +80 to control toxin expression yields an kill vector specifically active in HIV-1 virus infected cells.
  • Minichromosomes in which the major features are an SV40 ori with SV40 late functions intact, the HIV-
  • COS-7 Gluzman, Cell (1981) 23:175-182) or CMT4 (Gerard, et al. (1985) Molec. Cell Biol. 5:3231-3240) cells.
  • COS-7 cells are constitutive for wild type T antigen and VPl, VP2, and VP3 in trans.
  • CMT4 is inducible for wild- type T antigen by the metallothionein promoter.
  • the HIV-1 5'LTR should be transcriptionally silent in CV1 or VERO cells, or the cells used to construct the papovavirus packaging cells.
  • the kill gene can include not only the nontoxic HSV-tk gene product but more aggressive mechanisms such as angiogenin, a 14,000 dalton ribonuclease encoded by 369 bp of DNA (Kurachi, et al. (1985) Biochemistry 24:5494-5499), and the ricin or Diphtheria toxin A chain genes.
  • angiogenin inhibitor Lee, et al.
  • a new construct in CV1, CV1P, or VERO cells that includes a mutant SV40 T antigen that is SV40 ori replication positive but transformation negative, and also contains in a separate cellular chromosome integration site for one of the capsid proteins, perhaps VP2 or VP3 would solve this issue.
  • a nonamplifiable SV40 minichromosome can be delivered to a target cell by an SV40 capsid, and lymphocyte and HIV-1 infected cell specific transcription will occur. This transcription will encode a function designed to kill the target cell.
  • a disadvantage of a strategy that does not include T antigen in the minichromosomes is the lack of transient amplification of the infecting minichromosome. This could result in a shortfall of desired toxicity in the target cell.
  • To include an SV40 ori and a replication enabling T antigen would solve this problem.
  • Minichromosome DNA would be transiently amplified, gene products would be transiently amplified, and enhanced toxicity could be achieved.
  • the cell transforming and cell immortalization capabilities of SV40 T antigen must be addressed.
  • SV40 T antigen has long been known to be responsible for cell transformation in culture, and cell transformation has been associated with tumorigenicity in animals (Osborn, et al. J. Virol.
  • SV40 T antigen is a multifunctional 708-amino- acid phosphoprotein. Extensive mutational analyses of large T antigen has demonstrated that discrete regions of this multifunctional protein mediate these varied functions (Mohr, et al. J. Virol. (1989)
  • T antigen stabilization of p53 nonviral tumor antigen in mice while a logical candidate for the mechanism of T antigen induced cell transformation, is not required for transformation by SV40. Instead the Rb binding site seems to be involved.
  • the Rb suscepti ⁇ bility gene which when deleted or inactivated predisposes the development of retinoblastoma (Friend, et al. (1986) supra) binds SV40 T antigen (DeCaprio, et al. (1988) 54:275-283).
  • T antigen amino-acid residues 105-114 yielded a protein that failed to complex with the Rb susceptibility protein and failed to induce transformation as measured by focus formation and growth in soft agar.
  • mutant SV40 large T antigens that support minichromosome replication but not cellular transformation by virtue of mutation in AA 105-114 can be incorporated into minichromosome constructs.
  • packaging cells for a nonamplifiable SV40 minichromosome therapeutic As described in the construction of packaging cells for a nonamplifiable SV40 minichromosome therapeutic, another packaging cell construct is required for amplifiable minichromosome constructs in order to eliminate the possibility of recombination between mutant T antigens and the wild-type T antigen found in COS-7 and CMT4 cells. In this case the CV-1 or VERO derived cells would only contain capsid proteins.
  • the packaging cells for amplifying minichromosome constructs will contain the SV40 late region of VP2 and VP3 on separate constructs. Some of the constructs can be based on LPV, BKV and JC viruses, viruses closely related to SV40. Titers of q about 1 x 10 /ml can be expected from such a packaging cell where the virion proteins are driven by the metallothionein promoter (Gerard, et al. ,
  • VPl from LPV and BKV and enhancers from LPV, BKV, the T-cell receptor, and hepatitis B virus useful in targeting lymphocytic diseases and liver cancer respectively may also be prepared.
  • An HIV-1 therapeutic is dependent on the known activated lymphocyte transcriptional activators, and the known HIV-1 tat gene product. To develop ablative virus therapy into a more universal therapeutic approach will require specific knowledge of the target cell nuclear transcriptional activator factors(s) environment, or to target the virus to specific cell types through specific receptor interactions. Recently it has been reported that SV40 binds to protease and carbohydrate sensitive receptors at the apical surface of cells (Clayson, (1988) [reference]; Clayson, et al.
  • ICAM-1 immunoglobin supergene family member
  • Mahner et al. Cell (1989) 56:849-853
  • Greve et al. Cell (1989) 56:839-847
  • the putative virion surface binding site for picornaviruses, the canyon receptor is also highly conserved (Colonno, et al. Proc. Natl. Acad. Sci. USA (1988) 85:5449-5453; Rossman, et al. Virology (1988) 164:373-382).
  • SV40 infects most human cells
  • LPV infects B lymphocytes
  • attachment of a receptor protein to SV40 capsids could selectively direct the virus to target cells.
  • binding soluble CD4 would direct SV40 or LPV to HIV-1 infected lymphocytes.
  • An antibody fragment could also direct virions to p41 or gpl20 representing cells much like an antibody attached to a toxin has been used to selectively kill HIV-1 infected cells in vitro (Chaudhary, et al. Nature (1988) 355:369-372).
  • Receptor-mediated attachment to a cell of various ligands such as transferrin, monoclonal antibodies, and many viruses, is followed by transport of the receptor-ligand complex into a coated pit.
  • the coated pit invaginates and buds off the cell membrane into an internal vesicle. (Pastan and Willingham Science (1981) 214:504-509).
  • the internal vesicle then fuses with an endosome.
  • the fate of the endocytosed material in an endosome is diverse.
  • transferrin the pH environment of the endosome causes a conformational shift, releasing Fe+++.
  • the transferrin and transferrin receptor are then recycled to the cell surface for further use. If a MAb to the transferrin receptor is coupled to the ricin A chain, 50% of the MAb-ricin conjugate is recycled to the surface and released into the medium.
  • HA hemagglutinin
  • MAb directed viruses has the advantage of making use of natural adaptations of viruses to survive and pass through the endocytotic pathway.
  • proteolytic enzymes found in endosomes play a role in virion uncoating
  • it may be of use to include an amino acid sequence providing for proteolytic cleavage of the targeting portion of the virus in the endosomal vesicle by, for example, a cathepsin or a peptidase.
  • Cathepsin D a preglysoso- mal enzyme found in endosomes, cleaves between -Phe- Phe-.
  • the adjacent amino acids are not important.
  • a peptide bridge between, for example, a F(ab)' fragment and the virion, cleavable in the endosomal environment would be (AA- n -Phe-Phe-(AA)m where n + m is not more than about fifteen.
  • monoclonal antibodies conjugated to methotrexate have been disclosed by Umemoto et al. Cancer Immunol. Immunother. (1989) 28:9-16.
  • enhanced release of the targeted virion from the targeting moiety may be achieved by exploitation of the low pH environment of the endosome.
  • disulfide bonds are reduced at pH 5.5 in the presence of reducing agents.
  • the endosomal environment is also a reducing environment, disulfide bonds may be labile in endosomes.
  • use of a disulfide bridge between the F(ab') ⁇ fragment or other targeting moiety and the virion may provide for a bond which is labile in the endosome, thus releasing the virion.
  • Fc fragments have been retained in the MAb preparations to increase the circulating half-life of the antibody, to provide convenient chemical coupling sites for conjugate preparation, and to recruit Fc receptor mediated cytotoxic T cell killing.
  • retention of the Fc fragment also increases Fc receptor mediated binding to normal cells, and contributes to the strong humoral immune response that limits in vivo immune therapy to 2-4 I.V. injections.
  • the immune response to the F(ab')2 fragment has been shown to be minimal.
  • F(ab') 2 therapy is the short half- life of the fragment in vivo but by attaching F(ab') to a solid phase (such as a virion) the half-life and immunogenecity problems of current MAb therapy can be minimized.
  • MAbs, with or without attachment to therapeutic drugs have been shown to be cell specific in vivo in gastrointestinal malignancies and leukemia/lymphoma. Epenetos et al. The Lancet (1982) 2:999-1004.
  • immunohistochemical typing of tumor biopsies at various stages of malignant progression of tumor, with a panel of MAbs has confirmed the existence of antigenic heterogeneity within a given tumor type.
  • HBsAb Hepatitis B virus surface antigen
  • this issue can preferentially be addressed by use of a cocktail of MAbs.
  • this cocktail comprising a plurality of MAbs directed to different epitopic sites can be premixed prior to the coupling reaction of MAbs and virions.
  • the therapeutic virions containing the kill genes can all be identical, the mixture can be prepared after coupling.
  • a DNA sequence encoding the kill gene of interest can be synthesized employing conventional techniques such as overlapping single strands which may be ligated together to define the desired coding sequence.
  • the termini can be designed to provide restriction sites or one or both termini may be blunt-ended for ligation.
  • the kill gene may be isolated by various techniques.
  • mRNA can be isolated, the mRNA reversed transcribed, and the resulting single-stranded (ss)DNA used as a template to prepare double-stranded (ds)DNA and the (ds)DNA gene isolated.
  • Another technique is to isolate a piece of the DNA, and using a probe, appropriately degenerate, comprising a region of the most conserved sequences in the gene of interest, identify the sequences of interest.
  • the probe can be considerably less than the entire sequence, but should be at least 10, preferably at least 14, more preferably at least 20 nucleotides in length. Longer oligonucleotides are also useful up to the full length of the gene of interest. Both DNA and RNA probes can be used.
  • Kill genes of interest include the HSV-tk gene, and genes encoding RNAase, protease, and DNAase.
  • Other kill genes which may find use include anti- sense RNA directed at, for example, human oncogenies such as H-ras , K-ras, or N-ras genes.
  • the anti-sense RNA will generally be about 5 to 100 bp in length, preferably about 20 bp in length.
  • anti-sense RNA can be synthesized to various functional sites of the HIV genome (see, e.g., Gutch & Liddell (1988).
  • R region retrovirus RNA
  • RNA cleavage and polyadenylation regions provide for proper maturation for the mRNA transcripts and are necessary for efficient expression.
  • the native 3'- untranslated region may suffice, but the polyadenylation signal from, for example, SV40, particularly including a splice site which provides for more efficient expression, could also be used.
  • the 3'-untranslated region derived from a gene highly expressed in a particular cell type could be fused with the gene of interest.
  • the particular termination regions are not critical to this invention, being primarily for convenience, so long as they provide for expression of the gene of interest.
  • the resulting plasmids will be cloned and isolated and as required, the particular components of interest analyzed as to which sequence has been developed to ensure that the proper sequence has been obtained.
  • the desired sequence may be excised from the plasmid and introduced into a different vector or the plasmid may be restricted and the desired component(s) manipulated, as appropriate.
  • a shuttle vector will be employed where the vector is capable of replication in different hosts requiring different replication systems. This may or may not require additional markers which are functional in the two hosts. Where such markers are required, these can be included in the vector.
  • the plasmid containing the cassette, two replication systems and the marker(s) may be trans ⁇ ferred from one host to another as required.
  • any useful marker may be used. Desirably, resistance to an antibiotic or methotrexate is of interest.
  • a marker for selection is highly desirable for convenience, other procedures for screening transformed cells have been described. For example, transformed cells may be screened by the specific products they make, for example, by immunological or enzymatic methods. The various constructs of interest will then be purified.
  • the virions produced by the packaging cells and comprising the kill gene of interest and flanking regions providing for regulation of expression may be introduced into the intended host by any convenient means, such as intravenous or subcutaneous injection, and oral or intranasal administration.
  • the packaging cells themselves may be introduced into the host by grafting, for example, skin cells, and the like, where the cells grow and express the viral genes and produce the desired virions directly in the host.
  • the subject compositions can use a wide variety of toxins as kill genes. Genes which may be used include kill genes and anti-sense RNA.
  • the HSV-tk gene is a preferred cytotoxic gene in retrovirus constructs.
  • a specific retrovirus cell-targeting vector to deliver the HSV-tk gene to the target cells can be developed.
  • the target cells can then be killed after administration of acyclovir which is phosphorylated by HSV-tk but not by human thymidine kinase.
  • acyclovir monophosphatp acyclovir monophosphatp (acyclo-GMP)
  • normal cellular enzymes catalyze the sequential synthesis of acyclo-GDP and acyclo-GTP.
  • Acyclo-GTP functions in two ways: (1) as an inhibitor of DNA polymerase and (2) upon incorporation into DNA, acyclovir causes chain termination.
  • acyclovir for HSV-tk containing cells as compared to normal mammalian cells, and acyclovir is noted for its low human toxicity.
  • Acyclovir is available in capsules, ointment, and as a powder for intravenous use. IV administrations of acyclovir range up to 750 to 3000 mg/m2/day, although usual IV doses are 5 mg/kg yielding peak plasma levels of lOttg/ml.
  • the T ⁇ /2 is 2.5 hours in patients with normal renal function. Bioavailability of the oral administration is about 20% (Gil an, et al.
  • nucleotide analogs such as E5-(2-bromovinyl)-2' deoxyuridine (BVDU) , 9-[4- hydroxy-3-(hytroxymethyl)but-l-yl]guanine, 2'-fluoro- 5-iodo-ara-C (F1AC) (Harnden, et al. , J. Med. Chem. (1987) 10:1636-1642; Harada, et al. J. Med. Chem. (1987) £0:226-229; DeClercq, et al. Infectious Diseases (1981) 143:846-852) that function similarly are well documented and are available as pharmaceuticals.
  • BVDU E5-(2-bromovinyl)-2' deoxyuridine
  • F1AC 2'-fluoro- 5-iodo-ara-C
  • An advantage of this type of system is that when the genome comprising the HSV-tk gene is incorporated into a target cell, it will direct the synthesis of HSV-t , regardless of whether the target cell is an HIV infected T4 lymphocyte, or a lung cancer cell, a pancreatic cancer cell, or any other cell to which the packaging mechanisms can specifically direct the virus.
  • the Herpesvirus thymidine kinase Upon infection of the target cell the Herpesvirus thymidine kinase is produced intra-cellularly. It is only upon application of acyclovir that cell killing occurs.
  • Cancer chemotherapy has traditionally been directed at inhibition of DNA synthesis.
  • the target cell can also be killed independently of DNA synthesis. Potentially, this active chemotherapy could shorten the duration of therapy.
  • This is particularly desirable and the transient expression of many chromosomes derived from paporaviruses.
  • a ribosomal RNA specific ribonuclease such as angiogenin, or Diphtheria toxin that ADP ribosylates elongation factor 2 (EF-2) required for protein synthesis.
  • Toxins together with a provirus or minichromosome containing the mouse mammary tumor (MoMTV) virus 5'LTR which is inducible with dexamethasone, may be used. This type of construct is illustrated in Figure 9.
  • a human placental ribonuclease inhibitor (Shapiro and Vallee Pro. Nat. Acad. Sci. (1987) ££:2238-2241) can be cloned into a packaging cell which lacks a glucocorticoid receptor.
  • the ribonuclease inhibitor can be cloned so as to replace the defective env gene in pCRIPenv-. This would have the effect of protecting the packaging cell from the activity of the angiogenin contained in the retrovirus genome to be packaged.
  • the angiogenin gene can then be cloned downstream of the dexa ⁇ methasone inducible promoter enhancer derived from the U3 region of the MMTV LTR (Overhauser and Fan J ⁇ Virology (1985) 54:133144; Ponta, et al., Proc. Nat. Acad. Sci (1985)£:1020-1024) or the moloney murine sarcoma virus (MoMSV) enhancer located within MoMSV LTR (Miksicek, et al.. Cell (1986) 4£:283-290).
  • MoMSV moloney murine sarcoma virus
  • the effect of these constructions would be to provide a genome which is targeted to a specific glucocorticoid receptor-containing cell type, using any of the targeting methods described above.
  • a glucocorticoid such as dexamethasone
  • the target cell synthesizes angiogenin within the cytoplasm of the cell, thereby actively killing the cell.
  • the actual kill gene the gene encoding angiogenin
  • another ribonuclease gene such as that encoding pancreatic ribonuclease, so long as sufficient levels of the ribonuclease inhibitor are present in the packaging cell. Due to the glucocorticoid induction of the ribonuclease, conceivably the packaging cell would not express the ribonuclease in deleterious quantities.
  • Other ribonucleases and their inhibitors could also be used, for example, pancreatic ribonuclease and various of the natural cytoplasmic ribonuclease inhibitors (Blackburn and Moore, 1982).
  • ribonucleases and ribonuclease inhibitors can be altered to include proteases and protease inhibitors as well as DNAses and DNAse inhibitors (for example, restriction enzymes and specific methylases the ricin A chain, the diphtheria toxin A chain and the poliovirus AA2 protein).
  • proteases and protease inhibitors for example, restriction enzymes and specific methylases the ricin A chain, the diphtheria toxin A chain and the poliovirus AA2 protein.
  • trypsin could be inhibited by cloning bovine pancreatic trypsin inhibitor into the packaging cell construct (Gebhard, et al. (1986) Proteinase Inhibitors, Elsevier Science Publishers).
  • bdellins could be used to inhibit trypsin, plasmin and sperm acrosin, whereas eglins could be used to inhibit chymotrypsin and subtilisin (Seemuller et al. , Proteinase Inhibitors (1986) Elsevier Science Publishers) .
  • Other serine proteinase inhibitors include soybean trypsin inhibitor of the Kunitz and Bowman-Birk types (Ikenaka and Norioka Proteinase Inhibitors (1986) Elsevier Science Publishers).
  • Anti-sense RNA can also serve as a cell protecting or inhibiting gene.
  • Anti-sense RNA can be directed at the human oncogenies such as the human H-ras, K-ras, or N-ras genes.
  • pseudovirion yields of 1 x 10 /cell can be g expected. If 2 x 10 /cell are assumed per ml of culture fluid (Vero, microcarrier fermentation), yields will be nominally 2 x 10 /ml. A career q patient in complete remission has a residual 10 -
  • pCRIPenv- shown in figure 1, is a pBR322 amp resistant plasmid containing an engineered MoMuLV.
  • the MoMuLV contains the 5'LTR, is ⁇ - with a deletion between Ball (212) and Pstl (563) where the numbering is from the 5' end of MoMuLV.
  • the Sfil to Nsi fragment (5382-7054) has been replaced with the equivalent fragment containing a frame-shift mutation at position 6438 within env created by insertion of a ten base linker containing an EcoRI site (Colicelli, et al. (1985) Mol. Gen. Genet. 199:537-539). These packaging cells will be called ⁇ CRIPenv-cells.
  • Plasmid pMOV- ⁇ + (Mann, et al. (1983) Cell 11:153-159) is cut with Xhol (1560) and Sfil (5382). At the unique Xhol site (1560) Xhol-BamI adapters is added, and at the Sfil site (5382) BamHI-Sfil adapters are added:
  • BamHI-Sfi1 CATCCGGCCNNN GCCGGN
  • the BamHI fragment of HSV-tk (3.5 kb) from pxl is cloned in. This yields a ⁇ + MoMuLV- HSV-tk+ mutant that is packaged to high titer
  • Example II Mutagenesis of env Gene The unique Ndel-Nsil fragment (5401-7054) is subcl ⁇ ned for mutagenesis. Following mutagenesis, the Ndel fragment is replaced back into pNCl and transfected into CRIPenv-cells. The resultant virus particles are tested for desirable properties, including infectivity, host-range, host-range altera- tion by covalent attachment of specific receptors, and cell killing in response to acyclovir by virtue of the expressed HSV-tk.
  • env gene is not spliced properly in this construct (expression of both HSV-tk and env is not achieved)
  • other constructions of the two genes including a separate promotor for env, are made until the two genes are expressed properly.
  • Tests for virus production are by the XC overlay assay, and the reverse transcriptase activity assay (Rowe, et al. (1970) Virology 42:1136-1139).
  • Ndel (7054) is performed as follows: pUC19 contains a single Ndel site (183 within the pUC19 numbering system) and no Nsil sites. The Ndel site is removed by cutting pUC19 with Ndel, treating with SI nuclease and religation. The multiple cloning site in pUC19 is cut with EcoRI and Hindlll, to remove the polylinker. In its place is inserted a Ndel-Nsil adapter:
  • Ndel-Nsil insert is shown in Figure 3.
  • the env termination is at 7772.
  • env 5876-7054 there are codons for 15 cysteine residues. To test whether these cysteine residues are available for coupling with maleimide adducts, as well as the arbitrary mutations creating -SH groups, coupling is made in a manner analogous to that of
  • Analogous constructions which introduce lysine residues with NH-2 groups for coupling are created in pUC19-env-, transferred to pNCl where they are called pNC-NH-1, pNC-NH-2, etc. These plasmids are transferred into CRIPenv-cells, creating NC-NH-1, NC- NH-2 cells, etc., which in turn will produce envNH-1, envNH-2 helper-free virus particles containing the HSV-tk gene.
  • the packaging cells are designed to produce murine ecotropic viruses and are incapable of infecting human cells.
  • Example III Coupling of Specific Antibodies to Native or Modified env Glycoprotein
  • Tumor-specific antibodies are attached speci ⁇ fically to the protruding env glycoprotein, env-SH or env-NH2 group, so as to infect only tumor antigen presenting cells.
  • a mutation in the env gene is made yielding a sulfhydryl or amino group free for coupling by organic chemical means.
  • F(ab)'2 fragments of specific antibodies such as an antibody to pancreatic cancer or other tumor-specific antigens or hormone receptors is made to produce cell- specific reagents.
  • these cell-specific reagents are coupled by glutaraldehyde, periodate, or maleimide compounds to the virus gene delivery vehicle comprising the native or modified env glycoprotein.
  • Preferred reagents include maleimide reagents to couple with the sulfhydryl groups of the hinge region of the antibody. See, for example, Ishikawa, et al. (1983) J. Immunoassay 4:209.
  • Examples of other ligands which can be coupled by this method include the antibody to HIV gpl20 to target HIV infected lymphocytes, soluble CD4 protein to target HIV infected cells, transferrin to target transferrin receptors, and other members of specific binding pairs such as gastrin, secretin, cholecystokinin or somatomedin C to target their respective cell surface receptors.
  • Example IV Coupling of CD4 Protein to Modified env Glycoprotein A virus designed to infect HIV-infected T4 lymphocytes is also made by fusing the amino acid sequence of the CD4 protein that is recognized by the HIV gpl20 to the N-terminus of the env gene. In this way it is possible to direct infection by the HSV-tk containing provirus to T4 lymphocytes specifically elaborating the HIV gpl20 on the cell surface. Chandhery, et al. (1988) Nature 355:369-372. Preparation of pNC-CD4
  • CD4 is a polypeptide of 55 kDa, about 435 amino acids of which are expressed predominantly on the surface of helper T cells.
  • CD4 is a member of the immunoglobin superfamily and appears to be capable of regulating the proliferation of the CD4 subset of T cells.
  • the N-terminal 178 amino acids of CD4 fused to amino acids 1-3 and 256-613 of Pseudo onas exotoxin A, shown to specifically kill HIV infected cells (Chaudhery, et al. (1988) Nature 355:369-372) is replaced into pNCl and transfected into CRIPenv— cells. Virus is tested for infectivity on HIV infected T4 lymphocytes. Acyclovir is then tested for its ability to kill the HIV infected cells.
  • This linker is ligated to the roughly 250 bp Ndel-
  • Ndel partial cut removes the approximately 75 bp of domain II of Pseudomonas exotoxin A carried in pVC403. This linear pUC19-env- is then placed back in frame with the following Ndel-Ncol adapter. His His His
  • mutant viruses are identified as useful (i.e., capable of cell targeted interactions)
  • production binary vector system of ⁇ + provirus and packaging cells are prepared. Experimentally it is convenient to mutate the env gene in a proviral format so as to avoid selecting env+ cells prior to proviral transfection. However, in production, the env gene of choice is transferred to the chromosome of the packaging cell.
  • the desired Ndel-Nsil fragment from the pNC-SH series the pNC-NH series or pNC-CD4 described above is transferred to NIH/3T3 cells.
  • Nonsense mutations, or deletions in pCRIPgag-2 mutations are inserted not only at the Xhol site (1560), but also at Nrul (2120), Bell
  • NIH/3T3 cells are sequentially transformed with pCRIPenv- that has had the EcoRI-Clal fragment of env removed, and with the disabled pCRIPgag2.
  • the generalized format is a genome structure of 5'LTR, ⁇ -, trans virus gene product driven by the 5'LTR, a second copy of the same trans virus gene product driven by the 5'LTR splice donor and env splice acceptor, and the SV40 poly A.
  • pNCgag,gag The structure of pNCgag,gag is shown in Figure 6a. 5
  • the starting materials for this construct are pCRIPenv- and pMOV- ⁇ - (mana et al Cell (1983) 33:153-159).
  • pCRIPenv- is cut with BC1I (2740) downstream from the gag terminator (2235) .
  • BC1I 2740
  • the Ndel (5401) to Sbal (5766) containing the env splice 0 acceptor site is cloned into the Bell site.
  • the two strands of DNA are MoMuLV bp 615-739.
  • a dsDNA linker of up to about 15 bp to match the Pstl (739) of the gag gene.
  • a Pstl(739)-BClI(2740) 5 fragment from pMOV- ⁇ - is then added.
  • a dsDNA linker of up to about 10 bp designed to join Bell and Clal adds the SV40 poly A to pCRIPenv-.
  • pCRIPenv- is cut with Hindi11 and pMOV- ⁇ - is cut with Nrul (2120) and Hpal (5816).
  • the HindiII and Nrul sites are joined by a dsDNA adapter of up to about 15 bp.
  • the Hpal site is joined by another dsDNA adapter of up to about 15 bp to the Ndel (5401)
  • Xbal(5766) from pMOV- ⁇ -.
  • a dsDNA adapter not to exceed 15 bp, joins the Sbal (5766) to the Nrul(2120)-Hpal(5816) of the pol gene, again from pMOV- ⁇ -.
  • the structure of pNCenv,env is shown in Figure 6c.
  • the env gene comes from the desired 5 pNC-env-series: the pNC-SH-series, the pNC-NH- series, or the pNC-CD4-series, or analogous pNC-env- series constructs designed to change the cell- specific host-range of the virus.
  • the backbone of pNC-env-env is pCRIPgag-2 (Danos and Mulligan, 1988, supra.
  • pCRIPenv- is cut with HindiII and a dsDNA adapter used to join the NdeI(5 01-BSSH 11(8212) to the Sfil(5382)-Nsil(7054) env fragment which also contains the -env splice acceptor site.
  • Nsil(7054) site of the altered env gene from the desired pNC-env series is joined in reading frame to the Nsil(7054) of the env gene of pCRIPgag-2.
  • plasmids pNCgag,gag, pNCpol,pol, and pNCenv,env, are transfected into NIH/3T3 cells in a sequential manner, producing as a result the desired production packaging cell.
  • the second part of the binary vector system is the ⁇ + provirus designed to transfer the desired genes of intent.
  • the structure of pNC-HSV-tk(2) is shown in Figure 7.
  • Anti-sense RNA can also serve as a cell inhibiting, or cell activity inhibiting gene.
  • the gene therapy delivery vehicles described herein represent ideal vectors for programming a target cell to produce anti-sense RNA.
  • an anti-sense construct for HIV infected T4 lymphocytes is shown.
  • Goodchild, et al. (Goodchild, et al. (1988), Proc Natl Acad Sci, Var:5507-5511) have synthesized anti-sense RNA of 20 bp length to various functional sites of the HIV genome. Oligomers complementary to sites within or near the sequences repeated at the ends of the retrovirus RNA (R region) , to splice acceptor, and splice donor sites were found by Goodchild et al. to be most effective. Construction of pNC-tk-AS
  • oligonucleotides are synthesized on a DNA synthesizer and inserted into the first BamHI-BamHI insert of HSV-tk in pNC-HSV- tk(2).
  • Example VII Construction of an SV40 Binary Vector The genes encoding polypeptides required for maturation of new SV40 viral particles are VPl, VP2, and VP3 (see Figure 10). VPl, VP2, and VP3 are independently placed on high expression vectors. The constructs are prepared using the MuLV 5'LTR promoter, or the metallothionein promoter or any other strong promoter constructs in the host cell could be used. The structures of the three constructs for VPl, VP2 and VP3 are shown in Figures 11a, lib, and lie, respectively. These three constructs are based on pCRIPenv- and SV40, and are designated pNCVPl,VPl, pNCPV2,VP2, and ⁇ NCVP3,VP3.
  • pNCVPl,VPl The structure of pNCVPl,VPl is shown in figure 9a.
  • the numbering system is from MoMuLV unless otherwise indicated.
  • PCRIPenv- is cut from HindiII (212) and Clal (7674) and SV40 is cut with Rsal (SV40, 1467) and Bell (SV40, 2770).
  • the HindiII and Rsal sites are joined by a dsDNA adapter of up to about 15 bp.
  • the BC1I site is joined to the env splice acceptor (S.A.) site, Ndel-Xbal(5401-5766), with a Bell-Ndel adapter.
  • S.A. env splice acceptor
  • Ndel-Xbal(5401-5766) site
  • Bell-Ndel adapter a second copy of VPl is ligated into the Xbal site with a Xbal-Rsal adapter.
  • Figure lib shows the structure of pNCVP2,VP2.
  • pCRIPenv- is cut with Hindi!land Clal.
  • the large plasmid backbone is reconstructed with two copies of the SV40 VP2.
  • the small fragment is further cut with Ndel and Sbal to give the env splice acceptor (5401-5766).
  • the HindiII site is joined to the HpalI site of VP2 with an adapter.
  • the VP2 AceI (SV40, 1628) is joined to the S.A. with an adapter to Ndel.
  • the S.A. Xbal is joined with an adapter to a second copy of VP2 at the Hpall (SV40,344).
  • FIG. 1 The AceI (SV40,1628) is finally joined back to pCRIPenv- with an adapter to Clal (5674). None of the adapters in this construct need maintain a correct reading frame.
  • Figure lie shows the structure of pNCVP3,VP3. This construct is derived from pCRIPenv- and SV40. pCRIPenv- is cut with HindiII and Clal to yield the plasmid backbone. The small fragment is further cut with Ndel and Xbal (5401-5766) * to yield the S.A. The HindiII (212) site is joined to the SV40 Haell (832) site with an adapter. The SV40 AceI (1628) is joined with a synthesized dsDNA adapter to the 5 S.A.
  • Ndel (5401).
  • the S.A. Xbal (5766) is joined with an adapter to the VP3 Haell (32).
  • An adapter of dsDNA is synthesized to join the Accl-Clal (SV40 1628-7674) to complete the construct. None of these adapters need maintain a reading frame.
  • the three constructs described in figures 9a, 9b, and 9c are sequentially transfected into a permissive line for SV40 such as CV1, BSC-1 or VERO to yield SV40 packaging cells.
  • Site-specific mutagenesis of the VPl construct yields surface SH- or NH2- groups for covalent attachment of cell-specific receptors. These receptors define the host range of the mobile element of the SV40 binary vector. These constructs are patterned after Henderson, et al. Clinical Chemistry (1986) 32:1637-1641.
  • the SH series of constructs are known as pNCSV-SH-1, pNCSV-SH-2, etc., or alternatively for lysine substitutions, pNCSV-NH- 1, pNCSV-NH-2, etc.
  • the mobile element of the SV40 derived vector comprises SV40 nt 2533-5243-294 encoding T antigen and ori, which encodes both the early and late promoters. Downstream from the SV40 late promoter is the desired kill gene.
  • SV40 has a narrow range of DNA size that can be packaged, only a maximum of about 2250 nt can be accommodated. This is sufficient space to encode angiogenin (369 nt),
  • the pBR322 backbone Prior to transfeeting the packaging cell described above, the pBR322 backbone is removed by cutting with Ba HI and religation.
  • the different kill genes in these constructs are synthesized as small DNA fragments (about 100 bp), hybridized and ligated together into the appropriate gene (Henderson, et al. (1986), supra) .
  • the synthesized gene is ligated into the SV40 vector, pNCHSVkill.
  • the above-described binary viral vectors may be used for inhibiting growth of a specific target cell, where the targeting of the vector is provided by a modified virion surface glycoprotein. Growth of the target cell is inhibited either directly by the gene product of a kill gene in the viral vector, or by the action of the kill gene product to produce a cell inhibitory product.
  • the vector finds use both in vitro and in vivo.
  • SV40 pseudovirions are constructed in COS-7 cells (Guzman, (1981) Cell 23:175-182) .
  • the initial constructs are early T antigen replacement vectors containing the Diphtheria toxin A chain (DT-A) gene under control of heavy chain immunoglobin enhancers.
  • Cloning procedures are as described (Davis, (1980) Advanced Bacterial Genetics - A Manual for Genetic Engineering) Enzymes are used according to recommendations by the manufacturers.
  • DNA is synthesized on an Applied Biosysterns Model 380A.
  • pSV40 is prepared by cloning SV40 DNA in pBR322 at the unique EcoRI site.
  • BbvII fragments (nt 850-2904, and 2904-4416) of T antigen are removed.
  • the small polyA-containing SV40 BclI-BamHl fragment (nt 2770-2533) is duplicated and a second multiple cloning site, (polyA MCS is inserted in the joint between the duplication, creating pNCSVl shown in Figure 13.
  • the SV40 ori and late functions VPl, VP2, VP3, aqno gene, and late polyA site are intact.
  • the early region contains: a short region of the 5' coding region of T antigen, a terminator terminate the T antigen transcript, a MCS for the insertion of DT-A and regulatory enhancer and promoter elements (IgG enhancer 289 bp Alul fragment, and heavy chain promoter), the SV40 splice site, the SV40 early polyA site, and a second multiple cloning site, the polyA MCS.
  • the polyA MCS is for an additional Alul enhancer and an anti-sense T3 RNA polymerase promoter (O'Conner, et al. J.
  • SV40 can package minichromosomes 75-102% of the wild-type 5243 bp (Subra ani, Anal. Biochem. (1983) 135:1-15) .
  • SV40 early replacement vector is removed from this plasmid by cutting at the single EcoRI site, gel purified, and circularized by ligation. Transfection of DNA by electroporation shows the toxicity and cell specificity of this construct. Transfection of COS-7 cells with this yields pseudovirions. This construct is expected to show an 80-90% specificity for B cells as compared to other cell types.
  • Example IX Construction of Anti-Sense SV40 Pseudovirions SV40 pseudovirions are constructed as described in Example VIII. An anti-sense construct can be added, driven by the T3 RNA polymerase promoter (O'Conner, et al. J. Bacteriol. (1987) 169:4457- 4462).
  • T3 RNA polymerase and the T3 promoter are very specific and function in eukaryotic cells (Deuschle, et al. Proc. Nat. Acad. Sci. (1989) £6:5400-5404). Inclusion of the T3 RNA polymerase in COS-7 cells, and the T3 promoter at the 3'end of the DT-A gene yields anti-sense RNA inactivating DT-A mRNA. Anti-sense RNA is not present in the target tumor cells as they do not contain the T3 RNA polymerase. This construct is shown in Figure 14.
  • SV40 infects most cell types of monkey and human origin in vitro, and in vivo. SV40 efficiently infects human lymphocytes (Oppenheim, et al. Proc. Nat. Acad. Sci. (1986) £:6925-6929; Karlsson, et al. Proc. Nat. Acad. Sci. (1985) £:158-162).
  • the SV40 pseudovirions described above will provide early pseudovirion in vitro efficacy data.
  • the SV40 work can be used to obtain preliminary animal model in vivo therapeutic data.
  • LPV has a much narrower host range than SV40, infecting only B cell tumor cells of monkey and human origin.
  • the host range restriction is a function of both enhancer and VPl sequences.
  • LPV enhancers restrict gene expression to B and T cells (Mosthaf, et al. Nature (1985) £15:597-600; Chen, et al. J ⁇ Virol. (1989) £:2204-2214), whereas VPl restricts LPV to B cells (Kanda, et al. J. Virol. (1986) 5_£:531-534) .
  • Figure 15 shows schematic representations of the SV40 and LPV genomes (Fried, et al.
  • Packaging cells will be constructed using COS-1 and COS-7, Psi-2, Psi-AM cells, and two gene separation of virion functions on separate host cell chromosome inserts, as models (Guzman, et al. Cell (1981) £:175-182; Mann, et al. Cell (1983) £:153-159; Cone, et al. Proc. Nat. Acad. Sci. (1984) £1:6349-6353; Danos, et al. Proc. Nat. Acad. Sci. (1988) 8£:6460-6464) .
  • the goal is to produce the highest possible titer construct. This is achieved by designing an appropriate packaging cell.
  • Packaging cells encode virion proteins to be supplied in trans to pseudovirion production.
  • the highest pseudovirion titer may be achieved with lymphoma packaging cells (BJA-B) (Klein, et al. Proc. Nat. Acad. Sci. (1974) 21:3283-3286) in suspension culture producing LPV capsids.
  • BJA-B lymphoma packaging cells
  • an SV40-LPV hybrid capsid produced in CV-1, Vero, or BJA-B packaging cells may be best.
  • the change of ecotropic to amphitropic host range of Moloney murine leukemia virus gene delivery vehicles involved retaining the ecotropic gag and pol genes, but exchanged the env gene from the amphitropic murine leukemia 4070A strain.
  • the requirement for an LPV vector is to utilize the LPV VPl.
  • VP2, VP3, ori, and T antigen could well be from SV40.
  • T antigen, VPl, VP2, and VP3 expression in the packaging cells will be cloned into the chromosome of the packaging cells with the amphitropic retrovirus Psi-AM. These genes will be controlled by the inducible metallothionein promoter.
  • FIG. 16 shows pNCLVP2, an LPV ori miniplasmid construct prepared in BJA-B packaging cells containing LPV virion polypeptides. This appears similar to Figure 13, pNCSVl, except the sequences are LPV and the restriction cut sites are different, yet the functional regions are identical.
  • a minichromosome with an SV40 ori, SV40 T antigen, and LPV VPl, VP2, and VP3, will be constructed by exchanging the Smal-Ncol ori fragment of pNCLV2 for the Kpnl (nt 294)-EcoNl(nt 5027) ori fragment of pNCLVl.
  • Appropriate packaging cells will be produced from the results of this work as described above for retroviruses (See Example V) .
  • the requirements are an ori and a circular genome 75-102% of 5243 bp. Larger plasmids are not packaged.
  • the production minichromosome will contain two copies of the toxin insert shown in Figure 14 oriented in opposite directions as shown in Figure 17. Sequences on either side of the ori to the toxin MCS will be unrelated DNA composed of lac £ sequences to make an appropriate sized (5240 bp) construct.
  • the above-described binary viral vectors may be used for inhibiting growth of a specific target cell. Growth of the target cell is inhibited either directly by the gene product of the kill gene in the viral vector, or by the action of the kill gene product to produce a cell inhibitory product.
  • the compositions and methods may find use, for example, as vehicles to transiently express foreign genes in HIV-1 infected cells, and other cell types when the viral agent is a papovavirus.
  • the advantages of, for example, SV40 as an ablative therapeutic or more generically virus therapy are two-fold: (1) expression of SV40 minichromosomes as transient genetic engineering of target and non-target cells does not occur, and (2) SV40 is stable and manufacturable.
  • SV40 minichromosomes amplified by T antigen dependent replication as well as nonamplified constructs may find use.
  • Papovaviruses offer the additional advantage that they generally have a narrow host range and those might be used to develop tissue specific virus therapy.
  • Example XI Construction of LPV-76 LPV-76 is a mutant of LPV that infects both T cells and B cells of monkey and human origin (Kanda, et aJ _ J. Virol. (1986) .141:196-211).
  • the mutations in LPV-76, as compared to LPV, responsible for increasing the host range to include T cells are three amino acid changes in VPl. Otherwise the viruses are identical.
  • To build on the therapeutic described in Example X requires two additions: First the packaging cell must express the VPl of LPV-76 instead of the VPl or LPV. Second the DT-a toxin insert shown in Figure 14, and incorporated in the production minichromosome is Figure 17, must be redesigned to limit gene expression to T cells.
  • the construction of the packaging cell is as follows.
  • the appropriate mutations in VPl are removed from LPV-76 on a 3 kb PatI fragment (Kanda, et al. J. Virol. (1985) 55:96-100). This fragment is further reduced to contain only VPl.
  • Packaging cells are produced in CV-1, VERO, or BJA-B cells as described in Example X.
  • a T cell ablative therapeutic is constructed using DT-A toxin under the expression control of the T cell receptor (TCR) enhancers/promoters.
  • T cell receptor is a cell surface heterodimer molecule composed of either alpha + beta or gamma + delta chains.
  • the alpha gene is expressed in the classical helper and killer T cells (CD4+CD8- and CD4-CD8+) whereas the delta chain is expressed primarily in the CD4-Cd8- T cells.
  • a single locus containing both the alpha and delta genes is of interest because t cell derived acute and chronic lymphocytic leukemia (T-ALL and T-CLL) involve translocations of this locus.
  • This enhancer will control DT-A expression as shown in Figure 18.
  • the toxin insert shown in Figure 18 is inserted in the production plasmid shown in Figure 17 replacing the toxin insert shown therein.
  • Other T cell enhancers which can be used in this construction include the enhancers found for the genes encoding the TCR beta and the CD3 delta subunit (Georgopoulos et al. EMBO J. (1988) 2*2401-2407).
  • HIV-1 infected T cells A therapeutic exclusively effective against a subset of T cells, HIV-1 infected T cells, can be constructed.
  • HIV-1 is the causative agent of AIDS and infects CD4+ cells.
  • Unique to HIV-1 infected cells is the enhancer defined by the tat protein encoded by the HIV-1 genome, and TAR, the mRNA sequence responsive to the tat protein and encoded in the 0 to +80 transcription sequence of the HIV-1 5'-LTR.
  • the packaging cell used for this therapeutic uses the VP-1 from LPV-76 described earlier in this Example.
  • the enhancer/promoter insert is shown in Figure 19.
  • the HIV 5'-LTR from -350 to +80 is derived as a Kpnl-Hindlll fragment from pHIV-CAT (Peterlin et al. AIDS (1988) 2 suppl. :529-540) .
  • the insert in Figure 19 replaces the toxin insert in the production minichromosome shown in Figure 17. This therapeutic will kill only T cells infected with HIV-1.
  • BK virus is a human papovavirus isolated from the urine of a renal allograft recipient undergoing immunosuppressive therapy. BKV infects humans in childhood and persists in a latent from without causing any serious disease. BKV is routinely isolated from the urine of immunosuppressed patients and pregnant women. Molecular and biochemical studies have shown that BKV resembles SV40. Indeed the relationship between SV40 and LPV; the molecular organization of T antigen, VPl, VP2, and VP3 is identical; BKV and SV40 share 73% amino acid identity.
  • the tisssue tropisim of BKV for kidney and liver cells is a result of VPl and the transcriptional enhancers/promoters.
  • a packaging cell containing thhe VPl of BKV is required.
  • a new packaging cell expressing the VPl of BKV is constructed in CV-1, VERO, or BJA-B cells.
  • the ori, T antigen, VP2, and VP3 may be obtained from BKV or SV40 as described in Example X.
  • the enhancer/promoter controlling the Hepatitis virus B virus surface antigen is used, hepatitis B virus has been cloned and sequenced.
  • the large surface protein gene of Hepatitis B virus is controlled by enhancers/promoters functional only in hepatoma cells (Yee, Science (1989) 246:658-661) .
  • This promoter/enhancer is contained within the fragment 966-1402 of the Hepatitis B virus sequence (Chang, et al. Molec Cell Biol. (1989) £:5189-5197) .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Virology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • Cell Biology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

On décrit des procédés et composés nouveaux permettant d'introduire un gène tueur, à savoir un gène capable de tuer une cellule ou d'inhiber sa croissance, dans une cellule cible spécifique. On prépare notamment une particule virale à réplication sensiblement défecteuse ayant des capacités de ciblage pour un type spécifique de cellules. Soit la glycoprotéine superficielle de la particule virale native fournit le ciblage, soit on modifie ladite glycoprotéine superficielle de manière à introduire un élément d'une paire de liaison spécifique, tel qu'un anticorps, ou un fragment de celui-ci à un antigène spécifique de tumeur, une protéine virale exprimée par une cellule infectée par virus, ou un ligand de récepteur. La particule virale se dirigera donc vers la cellule cible normale pour le virus ou vers une cellule comprenant l'autre membre de la paire de liaison spécifique. C'est grâce au gène tueur intégré dans la particule virale, que cette dernière est capable d'inhiber la croissance.
PCT/US1990/001867 1989-04-05 1990-04-05 Particule virale infectieuse ciblee a replication defectueuse WO1990012087A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US33400689A 1989-04-05 1989-04-05
US334,006 1989-04-05

Publications (1)

Publication Number Publication Date
WO1990012087A1 true WO1990012087A1 (fr) 1990-10-18

Family

ID=23305150

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1990/001867 WO1990012087A1 (fr) 1989-04-05 1990-04-05 Particule virale infectieuse ciblee a replication defectueuse

Country Status (3)

Country Link
EP (1) EP0466815A4 (fr)
JP (1) JPH04504361A (fr)
WO (1) WO1990012087A1 (fr)

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992014829A1 (fr) * 1991-02-19 1992-09-03 The Regents Of The University Of California Particules virales agissant sur une gamme d'hotes modifiee
WO1992022645A1 (fr) * 1991-06-14 1992-12-23 Genpharm International, Inc. Animaux transgeniques non humains presentant une deficience immunitaire
WO1993000103A1 (fr) * 1991-06-21 1993-01-07 The Wistar Institute Of Anatomy And Biology Proteines d'enveloppe chimeriques pour le ciblage de virus
WO1993005147A1 (fr) * 1991-08-30 1993-03-18 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Particules defectives interferentes du vih avec un cd-env chimerique
WO1993009221A1 (fr) * 1991-10-30 1993-05-13 Thera Gene Hb Apport cible de vecteur viral dans des cellules de mammifere
WO1993008844A1 (fr) * 1991-10-30 1993-05-13 Universite Pierre Et Marie Curie (Paris Vi) Cellules transformees pour la prevention ou le traitement de maladies induites par des virus, notamment retrovirus pathogenes
WO1993022433A2 (fr) * 1992-04-28 1993-11-11 Frank Andreas Harald Meyer Medicament permettant de traiter l'homme, les animaux et les plantes par therapie genetique, dans le but notamment de bloquer la proliferation de virus et les carcinogenes et procede de fabrication dudit medicament
WO1994006920A1 (fr) * 1992-09-22 1994-03-31 Medical Research Council Virus recombines presentant un polypeptide non-viral sur leur surface externe
WO1994013823A1 (fr) * 1992-12-10 1994-06-23 Z. Company S.A. Sequence nucleotidique destinee au traitement du cancer et des infections
EP0633943A1 (fr) * 1992-04-03 1995-01-18 Alexander T. YOUNG Therapie genique utilisant des vecteurs viraux cibles
WO1995022617A1 (fr) * 1994-02-22 1995-08-24 Universite Pierre Et Marie Curie (Paris Vi) Systeme hote-vecteur utilisable en therapie genique
FR2716461A1 (fr) * 1994-02-22 1995-08-25 Univ Paris Curie Système hôte-vecteur utilisable en thérapie génique.
EP0672129A1 (fr) * 1992-11-20 1995-09-20 University Of Medicine & Dentistry Of New Jersey Transfert de genes specifique a un type cellulaire a l'aide de vecteurs retroviraux contenant des proteines de fusion enveloppe-anticorps
WO1995033842A1 (fr) * 1994-06-07 1995-12-14 Immunomedics, Inc. Vecteurs retroviraux sans danger, leur production et utilisation
EP0690129A1 (fr) * 1989-08-30 1996-01-03 The Wellcome Foundation Limited Substances nouvelles pour la thérapie des cancers
US5512421A (en) * 1991-02-19 1996-04-30 The Regents Of The University Of California Generation, concentration and efficient transfer of VSV-G pseudotyped retroviral vectors
EP0746625A1 (fr) * 1992-11-09 1996-12-11 UNITED STATES GOVERNMENT as represented by THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES Particules vectorielles pouvant etre ciblees
US5817491A (en) * 1990-09-21 1998-10-06 The Regents Of The University Of California VSV G pseusdotyped retroviral vectors
US5847096A (en) * 1991-08-30 1998-12-08 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services DNA constructs encoding CD4 fusion proteins
US5869331A (en) * 1992-11-20 1999-02-09 University Of Medicine & Dentistry Of New Jersey Cell type specific gene transfer using retroviral vectors containing antibody-envelope fusion proteins and wild-type envelope fusion proteins
US5885808A (en) * 1992-11-04 1999-03-23 Imperial Cancer Research Technology Limited Adenovirus with modified binding moiety specific for the target cells
US6057155A (en) * 1995-11-28 2000-05-02 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
US6071743A (en) * 1997-06-02 2000-06-06 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
US6153435A (en) * 1995-02-21 2000-11-28 Cornell Research Foundation, Inc. Nucleic acid that encodes a chimeric adenoviral coat protein
US6316210B1 (en) 1995-12-20 2001-11-13 Subsidiary No. 3, Inc. Genetic suppressor elements against human immunodeficiency virus
US6426412B1 (en) 1995-12-20 2002-07-30 Subsidiary No. 3, Inc. Nucleic acids encoding human immunodeficiency virus type 1 genetic suppressor elements
US6465253B1 (en) 1994-09-08 2002-10-15 Genvec, Inc. Vectors and methods for gene transfer to cells
US6534051B1 (en) 1992-11-20 2003-03-18 University Of Medicine And Dentistry Of New Jersey Cell type specific gene transfer using retroviral vectors containing antibody-envelope fusion proteins and wild-type envelope fusion proteins
US6537972B1 (en) 1997-06-02 2003-03-25 Subsidiary No. 3., Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
US6576456B2 (en) 1995-02-21 2003-06-10 Cornell Research Foundation, Inc. Chimeric adenovirus fiber protein
US6613506B1 (en) 2000-11-28 2003-09-02 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
US7361354B1 (en) 1999-11-25 2008-04-22 Viralytics Limited Methods for treating malignancies expressing ICAM-1 using coxsackie a group viruses
US7485292B2 (en) 2002-12-18 2009-02-03 Viralytics Limited Method of treating a malignancy in a subject via direct picornaviral-mediated oncolysis

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4687737A (en) * 1982-11-12 1987-08-18 Massachusetts Institute Of Technology Mammalian suppressor genes
US4707542A (en) * 1983-08-22 1987-11-17 Merck & Co., Inc. Immunogenic HbsAg derived from transformed yeast

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1310924C (fr) * 1986-04-24 1992-12-01 Francis P. Mccormick Systeme d'administration d'un medicament a l'aide de particules infectieuses
EP0273085A1 (fr) * 1986-12-29 1988-07-06 IntraCel Corporation Procédé pour faire entrer des acides nucléiques dans des cellules eucaryotes
WO1989009271A1 (fr) * 1988-03-21 1989-10-05 Viagene, Inc. Retrovirus recombinants
GB8919607D0 (en) * 1989-08-30 1989-10-11 Wellcome Found Novel entities for cancer therapy

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4687737A (en) * 1982-11-12 1987-08-18 Massachusetts Institute Of Technology Mammalian suppressor genes
US4707542A (en) * 1983-08-22 1987-11-17 Merck & Co., Inc. Immunogenic HbsAg derived from transformed yeast

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Science, Volume 236, Published 05 June 1987, (MANIATIS et al, "Regulation of Inducible and Tissue-Specific Gene Expression", Pages 1237-1245. See entire document. *
See also references of EP0466815A4 *
Transactions of the Association of American Physicians, Volume CI, 29 April- 02 May, 1988, MARKOWITZ et al, "Safe and efficient ecotropic and amphotropic packaging lines for use in gene transfer experiments", See page 213. *

Cited By (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0690129A1 (fr) * 1989-08-30 1996-01-03 The Wellcome Foundation Limited Substances nouvelles pour la thérapie des cancers
US5817491A (en) * 1990-09-21 1998-10-06 The Regents Of The University Of California VSV G pseusdotyped retroviral vectors
US5512421A (en) * 1991-02-19 1996-04-30 The Regents Of The University Of California Generation, concentration and efficient transfer of VSV-G pseudotyped retroviral vectors
US5670354A (en) * 1991-02-19 1997-09-23 The Regents Of The University Of California Use of VSV-G pseudotyped vectors for transfer of genes into embryos
WO1992014829A1 (fr) * 1991-02-19 1992-09-03 The Regents Of The University Of California Particules virales agissant sur une gamme d'hotes modifiee
WO1992022645A1 (fr) * 1991-06-14 1992-12-23 Genpharm International, Inc. Animaux transgeniques non humains presentant une deficience immunitaire
US5714316A (en) * 1991-06-21 1998-02-03 The Wistar Institute Of Anatomy & Biology Chimeric envelope proteins for viral targeting
WO1993000103A1 (fr) * 1991-06-21 1993-01-07 The Wistar Institute Of Anatomy And Biology Proteines d'enveloppe chimeriques pour le ciblage de virus
WO1993005147A1 (fr) * 1991-08-30 1993-03-18 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Particules defectives interferentes du vih avec un cd-env chimerique
US5847096A (en) * 1991-08-30 1998-12-08 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services DNA constructs encoding CD4 fusion proteins
WO1993009221A1 (fr) * 1991-10-30 1993-05-13 Thera Gene Hb Apport cible de vecteur viral dans des cellules de mammifere
US5695991A (en) * 1991-10-30 1997-12-09 Got-A-Gene Ab Targeted delivery of virus vector to mammalian cells
WO1993008844A1 (fr) * 1991-10-30 1993-05-13 Universite Pierre Et Marie Curie (Paris Vi) Cellules transformees pour la prevention ou le traitement de maladies induites par des virus, notamment retrovirus pathogenes
EP0633943A1 (fr) * 1992-04-03 1995-01-18 Alexander T. YOUNG Therapie genique utilisant des vecteurs viraux cibles
EP0633943A4 (fr) * 1992-04-03 1997-05-02 Alexander T Young Therapie genique utilisant des vecteurs viraux cibles.
WO1993022433A3 (fr) * 1992-04-28 1994-07-07 Frank Andreas Harald Meyer Medicament permettant de traiter l'homme, les animaux et les plantes par therapie genetique, dans le but notamment de bloquer la proliferation de virus et les carcinogenes et procede de fabrication dudit medicament
WO1993022433A2 (fr) * 1992-04-28 1993-11-11 Frank Andreas Harald Meyer Medicament permettant de traiter l'homme, les animaux et les plantes par therapie genetique, dans le but notamment de bloquer la proliferation de virus et les carcinogenes et procede de fabrication dudit medicament
US6297004B1 (en) 1992-09-22 2001-10-02 Cambridge Drug Discovery Holding, Ltd Recombinant viruses displaying a nonviral polypeptide on their external surface
US5723287A (en) * 1992-09-22 1998-03-03 Medical Research Council Recombinant viruses displaying a nonviral polypeptide on their external surface
WO1994006920A1 (fr) * 1992-09-22 1994-03-31 Medical Research Council Virus recombines presentant un polypeptide non-viral sur leur surface externe
AU689755B2 (en) * 1992-09-22 1998-04-09 Biofocus Discovery Limited Recombinant viruses displaying a nonviral polypeptide on their external surface
EP1038967A3 (fr) * 1992-11-04 2006-02-01 Transgene S.A. Virus comprenant une fraction de liaison modifiée spécifique contre des cellules cibles
US5885808A (en) * 1992-11-04 1999-03-23 Imperial Cancer Research Technology Limited Adenovirus with modified binding moiety specific for the target cells
EP1038967A2 (fr) * 1992-11-04 2000-09-27 Transgene S.A. Virus comprenant une fraction de liaison modifiée spécifique contre des cellules cibles
EP0746625A4 (fr) * 1992-11-09 1997-05-02 Us Health Particules vectorielles pouvant etre ciblees
EP0746625A1 (fr) * 1992-11-09 1996-12-11 UNITED STATES GOVERNMENT as represented by THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES Particules vectorielles pouvant etre ciblees
EP0672129A4 (fr) * 1992-11-20 1997-06-11 Univ New Jersey Med Transfert de genes specifique a un type cellulaire a l'aide de vecteurs retroviraux contenant des proteines de fusion enveloppe-anticorps.
US6534051B1 (en) 1992-11-20 2003-03-18 University Of Medicine And Dentistry Of New Jersey Cell type specific gene transfer using retroviral vectors containing antibody-envelope fusion proteins and wild-type envelope fusion proteins
EP0672129A1 (fr) * 1992-11-20 1995-09-20 University Of Medicine & Dentistry Of New Jersey Transfert de genes specifique a un type cellulaire a l'aide de vecteurs retroviraux contenant des proteines de fusion enveloppe-anticorps
US6146885A (en) * 1992-11-20 2000-11-14 University Of Medicine And Dentistry Of New Jersey Cell-type specific gene transfer using retroviral vectors containing antibody-envelope fusion proteins
US5869331A (en) * 1992-11-20 1999-02-09 University Of Medicine & Dentistry Of New Jersey Cell type specific gene transfer using retroviral vectors containing antibody-envelope fusion proteins and wild-type envelope fusion proteins
BE1006437A3 (fr) * 1992-12-10 1994-08-30 Company S A Z Sequence nucleotidique destinee au traitement du cancer et des infections.
WO1994013823A1 (fr) * 1992-12-10 1994-06-23 Z. Company S.A. Sequence nucleotidique destinee au traitement du cancer et des infections
FR2716459A1 (fr) * 1994-02-22 1995-08-25 Univ Paris Curie Système hôte-vecteur utilisable en thérapie génique.
WO1995022617A1 (fr) * 1994-02-22 1995-08-24 Universite Pierre Et Marie Curie (Paris Vi) Systeme hote-vecteur utilisable en therapie genique
EP0855185A3 (fr) * 1994-02-22 1998-11-18 Universite Pierre Et Marie Curie Paris Vi Système hote-vecteur utilisable en thérapie génique
EP0855185A2 (fr) * 1994-02-22 1998-07-29 Universite Pierre Et Marie Curie Paris Vi Système hote-vecteur utilisable en thérapie génique
US5948675A (en) * 1994-02-22 1999-09-07 Universite Pierre Et Marie Curie (Paris Vi) Host-vector system which can be used in gene therapy
FR2716461A1 (fr) * 1994-02-22 1995-08-25 Univ Paris Curie Système hôte-vecteur utilisable en thérapie génique.
WO1995033842A1 (fr) * 1994-06-07 1995-12-14 Immunomedics, Inc. Vecteurs retroviraux sans danger, leur production et utilisation
US6465253B1 (en) 1994-09-08 2002-10-15 Genvec, Inc. Vectors and methods for gene transfer to cells
US6951755B2 (en) 1994-09-08 2005-10-04 Genvec, Inc. Vectors and methods for gene transfer
US6576456B2 (en) 1995-02-21 2003-06-10 Cornell Research Foundation, Inc. Chimeric adenovirus fiber protein
US6153435A (en) * 1995-02-21 2000-11-28 Cornell Research Foundation, Inc. Nucleic acid that encodes a chimeric adenoviral coat protein
US6057155A (en) * 1995-11-28 2000-05-02 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
US6329190B1 (en) 1995-11-28 2001-12-11 Genvec, Inc. Targetting adenovirus with use of constrained peptide motifs
US6649407B2 (en) 1995-11-28 2003-11-18 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
US6426412B1 (en) 1995-12-20 2002-07-30 Subsidiary No. 3, Inc. Nucleic acids encoding human immunodeficiency virus type 1 genetic suppressor elements
US6316210B1 (en) 1995-12-20 2001-11-13 Subsidiary No. 3, Inc. Genetic suppressor elements against human immunodeficiency virus
US6326152B1 (en) 1997-06-02 2001-12-04 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
US6537972B1 (en) 1997-06-02 2003-03-25 Subsidiary No. 3., Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
US6071743A (en) * 1997-06-02 2000-06-06 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
US6436634B1 (en) 1997-06-02 2002-08-20 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
US7361354B1 (en) 1999-11-25 2008-04-22 Viralytics Limited Methods for treating malignancies expressing ICAM-1 using coxsackie a group viruses
US8722036B2 (en) 1999-11-25 2014-05-13 Viralytics Limited Methods for treating malignancies using coxsackieviruses
US6613506B1 (en) 2000-11-28 2003-09-02 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
US7485292B2 (en) 2002-12-18 2009-02-03 Viralytics Limited Method of treating a malignancy in a subject via direct picornaviral-mediated oncolysis

Also Published As

Publication number Publication date
JPH04504361A (ja) 1992-08-06
EP0466815A1 (fr) 1992-01-22
EP0466815A4 (en) 1992-09-02

Similar Documents

Publication Publication Date Title
WO1990012087A1 (fr) Particule virale infectieuse ciblee a replication defectueuse
Heard et al. An amino-terminal fragment of the Friend murine leukemia virus envelope glycoprotein binds the ecotropic receptor
EP0181929B1 (fr) Facteurs de transcription par trans-activation
EP0759087B1 (fr) Compositions et procedes de criblage de vecteurs d'apport de genes
Rosen et al. Construction of recombinant murine retroviruses that express the human T-cell leukemia virus type II and human T-cell lymphotropic virus type III trans activator genes
ES2246533T3 (es) Fibra adenovirica modificada y adenovirus dianas.
WO1993000103A1 (fr) Proteines d'enveloppe chimeriques pour le ciblage de virus
US5747323A (en) Retroviral vectors comprising a VL30-derived psi region
JP2008301826A (ja) ベクターおよびウイルスベクター、およびこれらを増殖するためのパッケージング細胞株
CA2307933A1 (fr) Constructions virales capables de replication ou de semi-replication, preparation et utilisations pour l'apport de genes
Stuhlmann et al. Construction and properties of replication-competent murine retroviral vectors encoding methotrexate resistance
JP2002522090A (ja) 抗体エンベロープ融合タンパク質および野生型エンベロープタンパク質を含有するレトロウイルスベクターを用いる細胞型特異的遺伝子移入
JPH06335392A (ja) Hiv阻害性アンチセンスおよび他のヌクレオチド配列を含有する発現構成体、レトロウイルスのベクターおよびそれを含有する組換えレトロウイルス
JP2002511751A (ja) スプライシング配列を含んでなる組換えアデノウイルスベクター
JPH09511405A (ja) レトロウイルスベクターハイブリッド及びその遺伝子転移のための使用
WO1995034639A1 (fr) Nouvelle enveloppe retrovirale et long segment terminal a repetitions (ltr) et particules de vecteur retroviral comprenant cette enveloppe et/ou le ltr
JPH10501403A (ja) 抗体−エンベロープ融合タンパク質および野生型エンベロープ融合タンパク質を含むレトロウィルスベクターを用いる細胞型特異的遺伝子移入
WO1999060110A2 (fr) Proteines a enveloppe stable pour des vecteurs retroviraux
Yanagawa et al. Extra mouse mammary tumor proviruses in DBA/2 mouse lymphomas acquire a selective advantage in lymphocytes by alteration in the U3 region of the long terminal repeat
NZ300795A (en) Retroviral Plasmid Vectors (enhancer region of LTR replaced, primer binding site replaced or mutated), packaging/transduced cells, use for gene therapy
US6132731A (en) Murine leukemia virus vectors
JP2001500021A (ja) 新規な内部リボソームエントリー部位およびそれを含有するベクター
Clark et al. Comparison of the transcriptional properties of the Friend and Moloney retrovirus long terminal repeats: importance of tandem duplications and of the core enhancer sequence
US6762031B2 (en) Targeting viral vectors to specific cells
EP1059356B1 (fr) Constructions rétrovirales capables de la réplication autonome, leur préparation et leurs utilisations pour le transfert de gènes

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB IT LU NL SE

WWE Wipo information: entry into national phase

Ref document number: 1990906533

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1990906533

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1990906533

Country of ref document: EP