EP0549723A1 - Packaging cells - Google Patents

Packaging cells

Info

Publication number
EP0549723A1
EP0549723A1 EP91919095A EP91919095A EP0549723A1 EP 0549723 A1 EP0549723 A1 EP 0549723A1 EP 91919095 A EP91919095 A EP 91919095A EP 91919095 A EP91919095 A EP 91919095A EP 0549723 A1 EP0549723 A1 EP 0549723A1
Authority
EP
European Patent Office
Prior art keywords
cells
vector
packaging
cell line
retroviral
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP91919095A
Other languages
German (de)
English (en)
French (fr)
Inventor
Douglas J. Jolly
Jack R. Barber
James G. Respess
Stephen M. W. Chang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novartis Vaccines and Diagnostics Inc
Original Assignee
Chiron Viagene Inc
Chiron Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chiron Viagene Inc, Chiron Corp filed Critical Chiron Viagene Inc
Priority to EP02019446A priority Critical patent/EP1285967A3/en
Priority to EP97121096A priority patent/EP0854193A1/en
Publication of EP0549723A1 publication Critical patent/EP0549723A1/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/033Rearing or breeding invertebrates; New breeds of invertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/217IFN-gamma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/13Tumour cells, irrespective of tissue of origin
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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
    • C12N2740/13022New 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13023Virus like particles [VLP]
    • 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
    • C12N2740/13051Methods of production or purification of viral material
    • C12N2740/13052Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • 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
    • C12N2740/16051Methods of production or purification of viral material
    • C12N2740/16052Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • 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
    • C12N2740/16111Human Immunodeficiency Virus, HIV concerning HIV env
    • C12N2740/16122New 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
    • 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

Definitions

  • the present invention relates generally to retroviruses, and more specifically, to recombinant retroviruses which are capable of delivering vector constructs to susceptible target cells.
  • These vector constructs are typically designed to express desired proteins in target cells, including proteins which can have a therapeutic effect in a number of ways, and hence, constitute a "drug" transport system for allowing transport of proteins (or RNA) into cells.
  • the specificity of proteins (and RNA) for enzymatic reaction, for binding of cellular components, for immunological action, or for other biological effects allows for correspondingly specific actions on target cells if the protein or RNA molecule can be transported into the cells.
  • Such actions include the repair of genetic defects, production of antisense RNA to block cellular process, the enzymatic potentiation of prodrugs, and stimulation of the cellular immune system, as well as many other therapies based on the intracellular production of proteins.
  • Retroviruses are RNA viruses which can replicate and integrate into a host cell's genome through a DNA intermediate. This DNA intermediate, or provirus, may be stably integrated into the host's cellular DNA. Due to their efficiency at integrating into host cells, retroviruses are considered to be one of the most promising vectors for use in human gene therapy. These vectors have a number of properties that lead them to be considered as one of the most promising techniques for genetic therapy of disease.
  • a foreign gene of interest may be incorporated into the retrovirus in place of the normal retroviral RNA.
  • the retrovirus injects its RNA into a cell, the foreign gene is also introduced into the cell, and may then be integrated into the host's cellular DNA as if it were the retrovirus itself. Expression of this foreign gene within the host results in expression of the foreign protein by the host cell.
  • retroviruses which have been developed for gene therapy are murine retroviruses. Briefly, these retroviruses exist in two forms, as proviruses integrated into a host's cellular DNA, or as free virions.
  • the virion form of the virus contains the structural and enzymatic proteins of the retrovirus (including reverse transcriptase) , two RNA copies of the viral genome, and portions of the cell's plasma membrane in which is embedded the viral envelope glycoprotein.
  • the genome is organized into four main regions: the Long Terminal Repeat (LTR) , and the gag, pol, and env genes.
  • LTR Long Terminal Repeat
  • the LTR may be found at both ends of the proviral genome, is a composite of the 5' and 3' ends of the RNA genome, and contains cis-acting elements necessary for the initiation and termination of transcription.
  • the three genes gag, pol, and env are located between the terminal LTRs.
  • the gag and pol genes encode, respectively, internal viral structures and enzymatic proteins.
  • the env gene encodes the envelope glycoprotein which confers infectivity and host range specificity of the virus.
  • retroviral vectors contain long terminal repeats (LTRs) , the foreign DNA to be transferred and a packaging sequence ( ) .
  • LTRs long terminal repeats
  • This retroviral vector will not reproduce by itself because the genes which encode structural and envelope proteins are not included within the vector.
  • the packaging cell contains genes encoding the gag, pol, and env proteins, but does not contain the packaging signal " ⁇ ". Thus, a packaging cell can only form empty virion particles by itself.
  • the retroviral vector is introduced into the packaging cell, thereby creating a "producer cell.”
  • This producer cell manufactures virion particles containing only the retroviral vector's (foreign) DNA, and therefore has previously been considered to be a safe retrovirus for therapeutic use.
  • Live virus i.e., competent replicating retrovirus
  • Preparations of human therapeutics which are contaminated with retroviruses are not currently considered suitable for use in human therapy. For example, extreme ' measures are taken to exclude retroviral contamination of monoclonal antibodies for imaging and therapy.
  • Live virus can in conventional producer cells when: (1) The vector genome and the helper genomes recombine with each other; (2) The vector genome or helper genome recombines with homologous cryptic endogenous retroviral elements in the producer cell; or (3) Cryptic endogenous retroviral elements reactivate (e.g., xenotropic retroviruses in mouse cells).
  • Another issue is the propensity of mouse based producer lines to package endogenous retroviral-vector- like elements (which can contain one gene sequences) at efficiencies close to that with which they package the desired vector. Such elements, because of their vector ⁇ like structure, are transmitted to the target treatment cell at frequencies that parallel its transfer of the desired vector sequence.
  • a third issue is the ability to make sufficient vector particles at a suitable concentration to:
  • helper cell lines that have been described to date have been limited to a host cell range of murine, avian, rat and dog cells. While later helper cell lines have been generated using amphotropic retroviral vector systems, which can infect human cells as well as a broad range of other mammalian cells (see, Sorge et al.. Mol. Cell. Biol. 4:1720-1737 r 1984), amphotropic packaging lines developed thus far have retained portions of one or more of the viral LTRs, and, thus, even when multiple mutations are present, have remained capable of generating a replication-competent genome. Amphotropic vector systems with multiple mutations and reduced propensities toward generating infectious virus generally exhibit unsatisfactorily low titres of retroviral particles.
  • helper virus One of the more recent approaches to constructing safer packaging cell lines involves the use of complementary portions of helper virus, divided among two separate plasmids, one containing gag and pol , and the other containing env (see, Markowitz et al., J. Virol. 62.:1120-1124; and Markowitz et al.. Virology 167: 600-606, 1988.
  • One benefit of this double-plasmid system is that three recombination events are required to generate a replication competent genome. Nonetheless, these double- plasmid vectors have also suffered from the drawback of including portions of the retroviral LTRs, and therefore remain capable of producing infectious virus.
  • Cell lines containing both 3' and 5' LTR deletions have been constructed, but have thus far not proven useful since they produce relatively low titers (Daugherty et al., J. Virol. 63:3209-3212. 1989).
  • the present invention overcomes difficulties of prior packaging cell lines, and further provides other related advantages.
  • the present invention provides a method for producing recombinant retroviruses in which the retroviral genome is packaged in a capsid and envelope, preferably through the use of a packaging cell.
  • the packaging cells are provided with viral protein-coding sequences, preferably in the form of two plasmids integrated into the genome of the cell, which produce all proteins necessary for production of viable retroviral particles, a DNA viral construct which codes for an RNA which will carry the desired gene, along with a packaging signal which will direct packaging of the RNA into the retroviral particles.
  • the present invention additionally provides a number of techniques for producing recombinant retroviruses which can facilitate: i) the production of higher titres from packaging cells; ii) the production of higher titres of helper free recombinant retrovirus from packaging cell lines that are non-murine (to avoid production of recombinant or endogenously activated retroviruses, and to avoid packaging of defective murine retroviral sequences) and which will infect human cells; i ⁇ ) the production of helper free recombinant retroviruses with higher titres using alternative non-hybrid envelopes such as xenotropic or polytropic envelope proteins (to allow infection of cells poorly infectable with amphotropic recombinant retroviruses or to allow specificity of cell type infection) .
  • alternative non-hybrid envelopes such as xenotropic or polytropic envelope proteins
  • packaging of vector constructs by means not involving the use of packaging cells; v) the production of recombinant retroviruses which can be targeted for preselected cell lines; vi) the construction of retroviral vectors with tissue-specific (e.g., tumor) promoters; and vii) the integration of the proviral construct into a preselected site or sites in a cell's genome.
  • tissue-specific e.g., tumor
  • One technique for producing higher titres from packaging cells takes advantage of the discovery that of the many factors which can limit titre from a packaging cell, one of the most limiting is the level of expression of the packaging proteins, namely, the gag, pol, and env proteins, as well as the level of expression of the retroviral vector RNA from the proviral vector. This technique allows the selection of packaging cells which have higher levels of expression (i.e., produce higher concentrations) of the foregoing packaging proteins and vector construct RNA.
  • this technique allows selection of packaging cells which produce high levels of what is referred to herein as a "primary agent,” which is either a packaging protein (e.g., gag, pol, or env proteins) or a gene of interest to be carried into the genome of target cells (typically as a vector construct) .
  • a packaging protein e.g., gag, pol, or env proteins
  • a gene of interest to be carried into the genome of target cells (typically as a vector construct) .
  • This is accomplished by providing in packaging cells a genome carrying a gene (the "primary gene") which expresses the primary agent in the packaging cells, along with a selectable gene, preferably downstream from the primary gene.
  • the selectable gene expresses a selectable protein in the packaging cells, preferably one which conveys resistance to an otherwise cytotoxic drug.
  • the cells are then exposed to a selecting agent, preferably the cytotoxic drug, which enables identification of those cells which express the selectable protein at a critical level (i.e., in the case of a cytotoxic drug, by killing those cells which do not produce a level of resistance protein required for survival) .
  • a selecting agent preferably the cytotoxic drug
  • the expression of both the selectable and primary genes is controlled by the same promoter.
  • this technique is first used to select packaging cells expressing high levels of all the required packaging proteins, and then is used to select which of these cells, following transfection with the desired proviral construct, produce the highest titres of the recombinant retrovirus.
  • Techniques are also provided to select cells that produce higher titres of helper free recombinant retroviruses in non-murine cells. These cell lines produce recombinant retroviruses capable of efficiently infecting human cells. These techniques involve screening potential parent cells for their ability to produce recombinant retroviruses in the presence of a replicating virus. Subsequently, uninfected cultures of candidate cell lines chosen by the above procedure are infected with a vector expressing a retroviral gag/pol, and clones which synthesize high levels of gag/pol are identified. A clone of this type is then reinfected with a vector expressing env, and clones expressing high level of env (and gag/pol) are identified.
  • high levels means discernibly greater than that seen in the standard mouse packaging line, PA317 on a Western blot analysis.
  • Many non-mouse cell lines such as human or dog have never been known to spontaneously generate competent retrovirus, do not carry possible recombination partners for recombinant murine retroviral packaging or gene sequences; and do not carry genes which make RNA which may be packaged by the MLV system.
  • Techniques are provided to generate cell lines which produce high titres of recombinant retroviruses using alternative envelopes such as xenotropic or polytropic by techniques similar to those described above.
  • retroviruses may be used in infecting amphotropic resistant cells (xenotropic envelope) or infecting only a subset of cells (polytropic) .
  • a technique suitable for producing recombinant retroviruses which can be targeted for preselected cell lines utilizes recombinant retroviruses having one or more of the following: an env gene comprised of a cytoplasmic segment of a first retroviral phenotype, and an extracel ⁇ lular binding segment exogenous to the first retroviral phenotype (the binding segment being from a second viral phenotype or from another protein with desired binding properties which is selected to be expressed as a peptide which will bind to the desired target) ; another viral envelope protein; another ligand molecule in place of the normal envelope protein; or another ligand molecule along with an envelope protein that does not lead to infection of the target cell type.
  • an env gene comprised of a cytoplasmic segment of a retroviral phenotype is combined with an exogenous gene encoding a protein having a receptor-binding domain to improve the ability of the recombinant retrovirus to bind specifically to a targeted cell type, e.g., a tumor cell.
  • a receptor-binding domain which binds to receptors expressed at high levels on the surface of the target cell (e.g., growth factor receptors in tumor cells) or alternatively, a receptor-binding domain binding to receptors expressed at a relatively higher level in one tissue cell type (e.g., epithelial cells, ductal epithelial cells, etc., in breast cancer).
  • a tissue cell type e.g., epithelial cells, ductal epithelial cells, etc., in breast cancer.
  • Techniques for integrating a retroviral genome at a specific site in the DNA of a target cell involve the use of homologous recombination, or alternatively, the use of a modified integrase enzyme which will recognize a specific site on the target cell genome.
  • site- specific insertion allows genes to be inserted at sites on the target cells' DNA, which will minimize the chances of insertional mutagenesis, minimize interference from other sequences on the DNA, and allow insertion of sequences at specific target sites so as to reduce or eliminate the expression of an undesirable gene (such as a viral gene) in the DNA of the target cell.
  • Figure 1A depicts four plasmids designed to express retroviral proteins in mammalian cells.
  • pSVgp and pRSVenv are cotransfected with a selectable marker, while pSVgp-DHFR and pRSVenv-phleo are the equivalent plasmids with the selectable marker placed downstream of the viral protein-coding regions.
  • Figure IB depicts vectors which lead to expression of: 1. MLV core proteins (pSCVIO) ; 2-4 MLV amphotropic env (pCMVenv AmDra, pCMVenv AnNhe, pMLPenv
  • Figure 1C depicts the results of the screening procedure for assessing the intrinsic ability of cell lines to make retroviral vectors in the presence of helper virus (Example 2B) .
  • Figure ID depicts the results of selecting clones of cells into which pSCVIO had been introduced and examining these clones for gag production as compared to
  • Figure IE depicts the results of Western blot experiments to compare levels of amphotropic env in cell lysates from DA, CA, 2A and PA317.
  • Figure IF depicts the results of Western blot experiments to compare levels of xenotropic env in cell lysates from transient transfections of CF gag/pol and permanently expressing lines XF7, X6, X10 and PA317 (ampho env) .
  • Figure 1G depicts the results of Western blot experiments to compare levels of MCF (polytropic) env in HT1080 derived clones, PA317 (amphotropic env) and HX (xenotropic env) .
  • Figure 2 depicts three sites of fusion of HIV env and MoMLV env after site-directed mutagenesis.
  • the joint at the extracellular margin of the transmembrane region is designated as A, while B and C indicate loca ⁇ tions of joints at the middle of the transmembrane region and cytoplasmic margin, respectively.
  • the numbering is according to nucleotide numbers (RNA Tumor Viruses. Vol. II, Cold Spring Harbor, 1985).
  • ST, SR, SE are the starts of tat, rev and env while TT, TR, and TE are the corresponding termination sites.
  • Figure 3 depicts the substitution of U3 in a 5' LTR by a heterologous promoter/enhancer in which can be fused to either the Sac I, Bssh II or other site in the region.
  • Figure 4 illustrates a representative method for crossing transgenic mice expressing viral protein or vector RNA.
  • the present invention is based, in part, upon the discovery of the major causes of low recombinant virus titres from packaging cell lines (PCL) , and of techniques to correct those causes. Basically, at least five factors may be postulated as causes for low recombinant virus titres:
  • the limited availability of viral packaging proteins is the initial limiting factor in recombinant retrovirus production from packaging cells.
  • titre increases to about 10 5 infectious units/milliliter, following which increasing packaging protein level has no further effect on titres.
  • titres can be further augmented by also increasing the level of retroviral vector genome available for packaging.
  • it is advantageous to select producer cells that manufacture the maximum levels of packaging proteins and retroviral vector genomes. It has been discovered that the methods of identifying, and thus selecting, packaging cells and producer cells, described earlier under the section entitled "Background of the Invention,” tend to lead to selection of many producer cells which produce low titres for the reasons described below.
  • the present invention takes advantage of the previously disadvantageous fact that the protein expression level of a gene downstream from the 5' LTR or other promoter, and spaced therefrom by an intervening gene, is substantially less than if the intervening gene were absent.
  • the selectable gene is placed downstream from a gene of the packaging genome or the gene of interest carried by the vector construct, but is still transcribed under the control of the viral 5' LTR or other promoter without any splice donor or splice acceptor sites. This accomplishes two things. First, since the packaging genes or genes of interest are now upstream with no intervening gene between themselves and the promoter, their corresponding proteins (packaging protein or protein of interest) will be expressed at a higher level (five- to twentyfold) than the selectable protein.
  • the selectable protein will be expressed on average at a lower level, with the distribution of level of expression shifting toward lower levels.
  • the selection level for resistance to phleomycin remains the same, so that only the top-end expressing cells survive.
  • the levels of the packaging protein or of the protein of interest will still be proportional, only in this case, a higher level of selectable protein corresponds to a much higher level of packaging protein or protein of interest.
  • the foregoing procedure is performed using a plasmid carrying one of the proviral gag/pol or env packaging genes, along with a first selectable gene. These cells are then screened for the cells producing the highest levels of protein by reaction with an antibody against gag/pol (or possibly env) , a second enzyme or labelled antibody, and then sorted on a fluorescence- activated cell sorter (FACS) or detected on a western blot. Alternatively, other tests for protein level may be used. Subsequently, the procedure and screening are repeated using those selected cells, and the other of the gag/pol or env packaging genes.
  • FACS fluorescence- activated cell sorter
  • a second selectable gene (different from the first) would be required downstream from the packaging gene and the cells producing the largest amount of the second viral protein selected.
  • This cell line is a packaging cell line (PCL) that may be used with any available vector.
  • PCL packaging cell line
  • the procedure and screening are then repeated using the surviving cells, with a plasmid carrying the proviral vector construct bearing the gene of interest and a third selectable gene, different from the first or second selectable gene.
  • cells producing high titres of the desired recombinant retrovirus will be selected, and these can be cultured as required to supply recombinant retrovirus.
  • gag and pol can be independently introduced and selected.
  • Example 1 describes the construction of gag/pol and env plasmids designed to use these procedures.
  • a 0.7 kb Bam HI to BstE II fragment from the plasmid pUT507 (Mulsant et al., Somat. Cell. Mol. Genet. 14.:243, 1988) with the BstE II end filled in carries the phleo resistance coding sequence.
  • the 4.2 kb Bam HI to Xho I fragment, the contiguous 1.6 kb Xho I to Xba I (Xba I filled in) from RSVenv, and the phleo fragment were ligated to give pRSVenv-phleo.
  • pSVgpDHFR was made using the following fragments: the 3.6 kb Hind III to Sal I fragment from pSVgp containing the SV40 promoter plus MLV gag and some pol sequences; the 2.1 kb Sal I to Sea I fragment from pMLV-K with the rest of the pol gene, the 3.2 kb Xba I (Xba I filled-in) to Pst I fragment from pF400 with the DHFR gene plus poly A site, pBR322 origin and half the ampicillin resistance gene; the 0.7 kb Pst I to Hind III fragment from pBR322 with the other half of the ampicillin resistance gene. This gives pSVgp-DHFR. All these constructs are shown in Figure 1. These plasmids can be transfected into 3T3 cells or other cells and high levels of gag, pol or env obtained.
  • An additional method for accomplishing selection is to use a gene selection in one round and its antisense in a subsequent round.
  • gag/pol may be introduced into an HPRT-deficient cell with the HPRT gene and selected for the presence of this gene using that media which requires HPRT for the salvage of purines.
  • the antisense to HPRT could be delivered downstream to env and the cell selected in 6 thioguanine for the HPRT-deficient phenotype. Large amounts of antisense HPRT would be required in order to inactivate the HPRT gene transcripts, assuming no reversion occurred.
  • a further method of accomplishing selection is described below.
  • gag/pol in particular
  • Example 2 The relative ease, sensitivity, and reproducibility of immunoblotting allowed rapid, quantitative analysis of a large number of cell clones necessary to identify over-expressors of the MLV structural proteins (gag/pol in particular) (see Example 2) .
  • gag/pol expressing constructs which begin at nucleotide 563 of MoMLV
  • several others can be constructed which contain upstream lead sequences. It has been observed by Prats et al. (RNA Tumor Viruses Meeting. Cold Spring Harbor, N.Y., 1988) that a glycosylated form of the gag protein initiates at nucleotide 357 and a translation enhancer maps in the region between nucleotides 200-270.
  • gag/pol expressing constructs may be made beginning at the Bal I site (nucleotide 212) or Eag I site (nucleotide 346) to include these upstream elements and enhance vector production.
  • a preferred method of accomplishing this is to include degenerate mutations to inactivate the packaging signal found here, without affecting the coding potential of the nucleic acid.
  • gag/pol and env function separately allows for manipulation of these functions independently.
  • a cell line that expresses ample amounts of gag/pol can be used, for example, to address questions of titre with regard to env.
  • One factor resulting in low titres is the density of appropriate receptor molecules on the target cell or tissue.
  • a second factor is the affinity of the receptor for the viral envelope protein.
  • Envelope proteins from one retrovirus can often substitute, to varying degrees, for that of another retrovirus.
  • the envelope of murine virus 4070A, HTLV I, GALV, and BLV can each substitute for that of MoMLV, albeit with a lower efficiency (Cone and Mulligan, Proc. Natl. Acad. Sci., USA £:6349-53, 1984; Wilson et al., J. Virol. 63. 2374-78, 1989; Ban et al. , J__ Gen. Virol. 70.:1987-93, 1989).
  • PCLs were generated which individually express either amphotropic, xenotropic, or polytropic envelopes.
  • Vector produced from any of these PCLs can be used to infect any cell which contains the corresponding distinct receptor (Rein and Schultz, Virology 136:144-52. 1984). Some cell types may, for instance, lack the amphotropic receptor and thus be resistant to infection with amphotropic vector, but express the xenotropic receptor and therefore be infectable by xenotropic vector.
  • xenotropic vector in the presence of replication- complement xenotropic virus, may more effectively infect human hematopoietic progenitor cells (Eglitis et al., Biochem. Biophvs. Res. Comm. 151:201-206, 1988).
  • Xenotropic vector in the presence of replication- competent xenotropic virus, also infects cells from other species which are not easily infectable by amphotropic virus such as bovine, porcine, and equine (Delouis et al., Biochem. Biophvs Res. Comm. 169:80-14, 1990).
  • amphotropic virus such as bovine, porcine, and equine
  • the xenotropic PCLs will therefore be useful for veterinary purposes in these species.
  • Another example would be utilization of the spleen focus-forming virus (SFFV) envelope gene which may allow targeting to cells containing the erythropoietin receptor (J.P. Li et al.. Nature 343:762-764. 1990).
  • SFFV spleen focus-forming virus
  • amphotropic PCLs described herein canine and human fibroblasts
  • amphotropic vector were infectable by xenotropic vector but were resistant to infection by amphotropic vector, presumably due to the phenomenon of "viral interference" (cf. A. Rein, Virology
  • the xenotropic PCL therefore allows the facile infection of these amphotropic PCLs, which in turn produces 10-lOOx higher titre than PCLs whose vector has been introduced by other means (Miller et al., So at.
  • PCL expressing any viral envelope which can function with the MLV vector and packaging system and whose corresponding cellular receptor is found in a given PCL, is useful for allowing vector infection of that PCL.
  • Vector produced from the polytropic PCL described herein has a more restricted host range on human cells than vector produced from either amphotropic or xenotropic PCLs (see data below) .
  • the polytropic PCL may therefore be particularly useful for targeting vector to a specific human cell type.
  • envelopes from nonmurine retrovirus sources can be used for pseudotyping a vector.
  • the exact rules for pseudotyping i.e., which envelope proteins will interact with the nascent vector particle at the cytoplasmic side of the cell membrane to give a viable viral particle (Tato, Virology £8.:71, 1978) and which will not (Vana, Nature 336:36. 1988), are not well characterized.
  • a piece of cell membrane buds off to form the viral envelope molecules normally in the membrane are carried along on the viral envelope.
  • a number of different potential ligands can be put on the surface of viral vectors by manipulating the cell line making gag and pol in which the vectors are produced or choosing various types of cell lines with particular surface markers.
  • the pathogenic virus displays on the infected cell surface its virally specific protein (e.g., env) that normally interacts with the cell surface marker or receptor to give viral infection. This reverses the specificity of the infection of the vector with respect to the potentially pathogenic virus by using the same viral protein-receptor interaction, but with the receptors on the vector and the viral protein on the cell. It may be desirable to include a gene which encodes for an irrelevant envelope protein which does not lead to infection of target cells by the vector so produced, but does facilitate the formation of infectious viral particles. For example, one could use human Sup Tl cells as a helper line.
  • This human T-cell line expresses CD4 molecules at high levels on its surface. Conversion of this into a helper line can be achieved by expressing gag/pol with appropriate expression vectors and also, if necessary, the Moloney ecotropic env gene product as an irrelevant (for human cells) envelope protein (the Moloney ecotropic env only leads to infection of mouse cells) . Vectors produced from such a helper line would have CD4 molecules on their surfaces and therefore be capable of infecting only cells which express HIV env, such as HIV- infected cells.
  • hybrid envelopes (as described below) can be used in this system as well, to tailor the tropism (and effectively increase titres) of a retroviral vector.
  • a cell line that expresses ample amounts of a given envelope gene can be employed to address questions of titre with regard to gag and pol.
  • ligand molecules exogenously to the viral particles which either incorporate themselves in the lipid envelope or can be linked chemically to the lipid or protein constituents.
  • non-murine cell lines are potential parents for packaging lines. These include Vero cells which are used in Europe to prepare polio vaccine, WI38 which are used in the U.S. in vaccine production, CHO cells which are used in the U.S. for TPA preparation, D17 or other dog cells that may have no endogenous viruses, and those described in Example 2.
  • retroviral vectors The most important safety concern for the production of retroviral vectors is the inherent propensity of retroviral PCLs to generate replication- competent retrovirus after introduction of a vector (Munchau et al. , Virology 176:262-65. 1990).
  • homologous recombination can occur between the therapeutic proviral DNA and the DNA encoding the MoMLV structural genes ("gag/pol” and “env") present in the PCL (discussed below under "Generation of PCLs”) ; and 2) generation of replication-competent virus by homologous recombination of the proviral DNA with the very large number of defective endogenous proviruses found in murine cells (Steffen and Wei ' nberg, Cell 15:1003-10, 1978) ; Canaani and Aaronson, Proc. Natl. Acad. Sci.. USA 7j5:1677-81, 1979; Sto e and Coffin, J. Virol.
  • murine cell lines lacking vector can produce virus spontaneously or after induction, (e.g., xenotropic virus which can replicate in human cells, Aaronson and Dunn, J. Virol. .13.:181-85, 1974; Stephenson and Aaronson, Proc. Natl. Acad. Sci.. USA 71:4925-29, 1974; Aaronson and Stephenson, Biochem. Biophvs. Acta 458:323-54. 1976).
  • PCLs have been generated exclusively from non-murine cell lines (e.g., canine and human cell lines) which are known to lack genomic sequences homologous to that of MoMLV by hybridization analysis (data not shown) (Martin et al., Proc. Natl. Acad. Sci.. USA 7_8:4892-96, 1981).
  • non-murine cell lines e.g., canine and human cell lines
  • genomic sequences homologous to that of MoMLV by hybridization analysis (data not shown) (Martin et al., Proc. Natl. Acad. Sci.. USA 7_8:4892-96, 1981).
  • packaging cells described herein will have a low, but inherent capability of packaging random RNA molecules. Such RNA molecules will not be permanently transmitted to the pseudo-infected target cell.
  • the choice of host cell line for the PCL is of importance because many of the physical (such as stability) and biological properties (such as titre) of retroviral particles are dictated by the properties of the host cell.
  • the host cell must efficiently express (transcribe) the vector RNA genome, prime the vector for first strand synthesis with a cellular tRNA, tolerate and covalently modify the MLV structural proteins (proteolysis, glycosylation, myristylation, and phosphorylation) , and the maturing virion buds from the cell membrane, carrying many of the membrane components with it.
  • vector made from the mouse packaging line PA317 is retained by a 0.3 micron filter, while that made from the CA line described herein will pass through.
  • MLV structural gene expression vectors To decrease the possibility of replication- competent virus being generated by genetic interactions between the MLV proviral vector DNA and the structural genes of the PCL, separate expression vectors, each lacking the viral LTR, were generated to express the gag/pol and env genes independently. To further decrease the possibility of homologous recombination with MLV vectors and the resultant generation of replication- competent virus, minimal sequences other than the protein coding sequences were used. In order to express high levels of the MLV structural proteins in the host cells, strong transcriptional promoters (CMV early and Ad5 major late promoters) were utilized. An example of the construction of a MoMLV gag/pol expression vector (pSCVIO, see Figure 1B.1) follows: 1. The 0.7 Kb HinCII/Xmalll fragment encompassing the human cytomegalovirus (CMV) early transcriptional promoter (Boshart et al.. Cell .41:521-30, 1985) was isolated.
  • CMV cytomegalovirus
  • the CMV early promoter described for the gag/pol expression above (pSCVIO) was ligated in the plasmid pUCl ⁇ in the order CMV promoter- envelope (no added transcriptional termination signal) .
  • MLP amphotropic envelope
  • SV40 termination signal were ligated in plasmid pBR322 in the order MLP-envelope-SV40.
  • An example of the construction of an MLV xenotropic envelope expression vector (pCMMVxeno, see Figure IB.5) follows.
  • CMV early promoter and SV40 late termination signal described for the gag/pol expression above were ligated in the order CMV promoter-envelope-termination signal.
  • the vector pLRNL vector (Emi et al., J. Virol. __5 :1202-1207, 1991) was digested with BamHI.
  • Host Cell Selection Host cell lines were screened for their ability to efficiently (high titre) rescue a drug resistance retroviral vector (A alpha N2) using replication competent retrovirus to produce the gag/pol and env structural genes ("MA" virus). Titre was measured from confluent onolayers 16 h after a medium change by adding filtered supernatants (0.45 urn filters) to 5xl0 4 NIH 3T3 TK * cells on a 6 cm tissue culture plate in the presence of 4 ug/ml polybrene followed by selection in G418. Data from the screening process is shown in
  • Figure 2 Among the non-murine cell lines which demonstrate the ability to package MoMLV-based vector with high titre are the cell lines CF2, D17, 293, and HT1080. These cell lines were used herein as examples, although any other cells may be tested by such means.
  • D17, 293, and HT1080 were co-transfected with 1 ug of the methotrexate resistance vector, pFR400 (Graham and van der Eb, Virology 52.:456-67, 1973), and 10 ug of the MoMLV gag/pol expression vector, pSCVIO (above) by calcium phosphate co- precipitation (D17 and HT1080, see Graham and van der Eb, Virology 52_:456-67, 1973), or lipofection (293, see Feigner et al., Proc. Natl. Acad. Sci.. USA 84:7413-17. 1987) .
  • LARNL see Figure IB
  • Neo + marker which confers resistance to the drug, G418.
  • co-expression of gag/pol in the cell line and env from the vector allowed efficient packaging of the vector as determined by cell-free transfer of G418 resistance to 3T3 cells (titre) .
  • Titre was measured from confluent monolayers 16 h after a medium change by adding filtered supernatants (0.45 urn filters) to 5xl0 4 NIH3T3 TK "" cells on a 6 cm tissue culture plate in the presence of 4 ug/ml polybrene followed by selection in G418.
  • the vector titres from the cell lines correlated with the levels of p30 ga ⁇ (Table 1) . Since the level of env should be the same in each clone and is comparable to the level found in PA317 (data not shown) , this indicates that titre was limited by the lower levels of gag/pol in these cells (including PA317) . The titre correlated more closely with the levels of p30 gag than with the levels of RT.
  • PCLs the gag/pol over-expressors for 293 (termed 2-3) and D17 (termed 4-15) were co-transfected by the same techniques described above except that 1 ug of the phleomycin resistance vector, pUT507 (Mulsant et al., Somat. Cell Mol. Genet. 14:243-52, 1988), and 10 ug of the amphotropic envelope expression vectors, pMLPenvAmSph (for 2-3) or pCMVenvAmNhe (for 4-15) were used.
  • PCL, CF2 cells were electroporated (cf. Chu et al., Nucl. Acids Res. .15:1311-26, 1987) with 2 ug of the phleomycin resistance marker, pUT507, 10 ug of pSCVIO (above), and 10 ug of pCMVenvAmNhe (above) .
  • individual drug resistant cell colonies were expanded and analyzed for intracellular expression of MLV p30 gag and gp80 env proteins by western blot using specific antisera. A clone was identified which expressed relatively high levels of both gag/pol and ampho env ( Figure IE) .
  • Ten ug of the vector DNA of interest was co- transfected with 10 ug of DNA which expresses either xeno env (pCMVxeno, above) or a VSV G protein expression vector, MLP G, onto a cell line which expresses high levels of MoMLV gag/pol such as 2-3 cell (see above) .
  • the resultant vector containing xenotropic env or VSV G protein, respectively was produced transiently in the co ⁇ transfected cells and after 2 days cell free supernatants were added to the potential PCLs, and vector-infected cells were identified by selection in G418.
  • the gag/pol over-expressors for D17 (4-15) and HT1080 (SCV21) were co-transfected by the same techniques described above except that 1 ug of either the phleomycin resistance vector, pUT507 (for SCV21) , or the hygromycin B resistance marker, pY3 (for 4-15, see Blochlinger and Diggelmann, Mol. Cell Biol. 4.:2929-31, 1984), and 10 ug of the xenotropic envelope expression vector, pCMVxeno (above) was used.
  • vector containing VSV G protein was produced transiently in 2-3 cells. After 2 days, cell free supernatants were added to the xeno PCLs and after G418 selection cell free supernatants were collected from the confluent monolayers and titred as described above except that HT1080 cells, which are infectable by xeno vector, was used instead of NIH 3T3 TK " cells which are resistant to xeno vector.
  • the cell clones with the highest titre were chosen as PCLs and referred to as DX (D17 xeno) and HX (HT1080 xeno) , respectively.
  • the propensity of the PCLs described above to generate helper virus was tested even more stringently by co-cultivating ampho and xeno PCLs containing the vector, N2. Since ampho vector can infect the xeno PCLs and vice versa, this allows continuous cross-infection events, each of which increases the probability of generating helper virus.
  • 2A cells containing N2 were co- cultivated with HX cells containing N2. After 23 days, the cultures were still free of ampho and xeno viruses as judged by a vector rescue assay on 293 or Mus dunni cells, both of which can detect ampho and xeno viruses (Table 4) .
  • the gag/pol over-expressor for HT1080 (SCV21) was co ⁇ transfected by the same techniques described above, except that 1 ug of the phleomycin resistance vector, pUT507, and 10 ug of the polytropic envelope expression vector, pCMVMCF (above) was used. After selection for transfected cells in the presence of phleomycin, individual drug resistant cell colonies were expanded and analyzed for intracellular expression of MLV gp70 env protein by western blot using specific antiserum. Clones were identified which expressed relatively high levels of both gag/pol (not shown) and polytropic env ( Figure 1G) .
  • This cell clone was chosen as PCL and referred to as HP (HT1080 poly) .
  • HP HT1080 poly
  • vector containing VSV G protein was produced transiently in 2-3 cells and after 26 days, cell free supernatants were added to the polytropic PCL (HP) . After G418 selection, cell free supernatants were collected from the confluent monolayers and titred as described above on a variety of cell lines. The infection of human cells was very restricted, with all cell lines tested being negative with the exception of 293 cells.
  • retroviruses may be adapted for markedly improved growth in cell types in which initial growth is poor, simply by continual reinfection and growth of the virus in that cell type (the adapter cell) .
  • This can also be achieved using viral vectors that encode some viral functions (e.g., env), and which are passed continuously in cells of a particular type which have been engineered to have the functions necessary to complement those of the vector to give out infectious vector particles (e.g., gag/pol).
  • the virus is cloned out by any of a number of standard methods, the clone is checked for activity (i.e., the ability to give the same maximal growth characteristic on transfection into the adapter cell type) and this genome used to make defective helper genomes and/or vectors which in turn, in an appropriately manufactured helper or producer line, will lead to production of viral vector particles which infect and express in the adapter cell type with high efficiency (10 7 - 10 9 infectious units/ml) .
  • activity i.e., the ability to give the same maximal growth characteristic on transfection into the adapter cell type
  • Two additional alternative systems can be used to produce recombinant retroviruses carrying the vector construct.
  • Each of these systems takes advantage of the fact that the insect virus, baculovirus, and the mammalian viruses, vaccinia and adenovirus, have been adapted recently to make large amounts of any given protein for which the gene has been cloned. For example, see Smith et al. (Mol. Cell. Biol. 2:12, 1983); Piccini et al. (Meth. Enzvmologv. 153:545, 1987); and Mansour et al. (Proc. Natl. Acad. Sci. USA 82:1359, 1985).
  • viral vectors can be used to produce proteins in tissue culture cells by insertion of appro ⁇ priate genes into the viral vector and, hence, could be adapted to make retroviral vector particles.
  • Adenovirus vectors are derived from nuclear replicating viruses and can be defective. Genes can be inserted into vectors and used to express proteins in mammalian cells either by in vitro construction (Ballay et al. , EMBO J. 4.:3861, 1985) or by recombination in cells (Thummel et al., J. Mol. APPI. Genetics 1:435, 1982).
  • One preferred method is to construct plasmids using the adenovirus Major Late Promoter (MLP) driving: (1) gag/pol, (2) env, (3) a modified viral vector construct.
  • MLP adenovirus Major Late Promoter
  • a modified viral vector construct is possible because the U3 region of the 5' LTR, which contains the viral vector promoter, can be replaced by other promoter sequences (see, for example, Hartman, Nucl. Acids Res. 16:9345, 1988). This portion will be replaced after one round of reverse transcriptase by the U3 from the 3' LTR.
  • plasmids can then be used to make adenovirus genomes in vitro (Ballay et al., op. cit.), and these transfected in 293 cells (a human cell line making adenovirus E1A protein) , for which the adenoviral vectors are defective, to yield pure stocks of gag/pol, env and retroviral vector carried separately in defective adenovirus vectors. Since the titres of such vectors are typically 10 7 -10 1:L /ml, these stocks can be used to infect tissue culture cells simultaneously at high multiplicity. The cells will then be programmed to produce retroviral proteins and retroviral vector genomes at high levels.
  • retroviral vectors can be harvested from the cell supernatants.
  • Other viral vectors such as those derived from unrelated retroviral vectors (e.g., RSV, MMTV or HIV) can be used in the same manner to generate vectors from primary cells.
  • these adenoviral vectors are used in conjunction with primary cells, giving rise to retroviral vector preparations from primary cells.
  • HIV rev protein might be included to prevent splicing of HIV env or HIV gag/pol MLV vectors or HIV sor might increase the infectivity of T cells by free virus as it does with HIV (See Fischer et al.. Science 237:888-893. 1987).
  • gag/pol and env proteins made in the baculovirus system in a similar manner as described in Smith et al. (supra) (or in other protein production systems, such as yeast or E__ coli) ;
  • viral vector RNA made in the known T7 or
  • tRNA made as in (2) or purified from yeast or mammalian tissue culture cells; 4. liposomes (with embedded env protein); and
  • cell extract or purified necessary components (when identified) (typically from mouse cells) to provide env processing, and any or other necessary cell-derived functions.
  • (1) , (2) and (3) are mixed, and then env protein, cell extract and pre-liposome mix (lipid in a suitable solvent) added. It may, however, be necessary to earlier embed the env protein in the liposomes prior to adding the resulting liposome-embedded env to the mixture of (1) , (2) , and (3) .
  • the mix is treated (e.g., by sonication, temperature manipulation, or rotary dialysis) to allow encapsidation of the nascent viral particles with lipid plus embedded env protein in a manner similar to that for liposome encapsidation of pharmaceuticals, as described in Gould-Fogerite et al., Anal. Biochem. 148:15. 1985).
  • This procedure allows the production of high titres of replication incompetent recombinant retroviruses without contamination with pathogenic retroviruses or replication-competent retroviruses.
  • the host cell range specificity of a retrovirus is determined in part by the env gene products.
  • Coffin, J. RNA Tumor Viruses 2.:25-27, Cold Spring Harbor, 1985
  • MMV murine leukemia virus
  • RSV Rous Sarcoma virus
  • the cytoplasmic domain of envelope proteins are understood to play a role in virion formation. While pseudotyping (i.e., the encapsidation of viral RNA from one species by viral proteins of another species) does occur at a low frequency, the envelope protein has some specificity for virion formation of a given retrovirus.
  • the present invention recognizes that by creating a hybrid env gene product (i.e., specifically, an env protein having cytoplasmic regions and exogenous binding regions which are not in the same protein molecule in nature) the host range specificity may be changed independently from the cytoplasmic function. Thus, recombinant retroviruses can be produced which will specifically bind to preselected target cells.
  • a hybrid env gene product i.e., specifically, an env protein having cytoplasmic regions and exogenous binding regions which are not in the same protein molecule in nature
  • Example 10 describes the construction of a hybrid env gene which expresses a protein with the CD4 binding portion of the HIV envelope protein coupled to the cytoplasmic domain of the MLV envelope protein.
  • Hybrid HIV-MLV Envelopes A hybrid envelope gene is prepared using in vitro mutagenesis (Kunkel, Proc. Natl. Acad. Sci. USA £:488-492, 1985) to introduce a new restriction site at an appropriate point of junction. Alternatively, if the two envelope sequences are on the same plasmid, they can be joined directly at any desired point using in vitro mutagenesis. The end result in either case is a hybrid gene containing the 5' end of the HIV gp 160 and the 3' end of MLV pl5E.
  • the hybrid protein expressed by the resulting recombinant gene is illustrated in Figure 2 and contains the HIV gpl20 (CD4 receptor binding protein) , the extracellular portion of HIV gp 41 (the gp 120 binding and fusigenic regions) , and the cytoplasmic portion of MLV pl5E, with the joint occurring at any of several points within the host membrane.
  • a hybrid with a fusion joint at the cytoplasmic surface causes syncytia when expressed in Sup Tl cells.
  • the number of apparent syncytia are approximately one-fifth that of the nonhybrid HIV envelope gene in the same expression vector.
  • Syncytia with the hybrid occurs only when the rev protein is co-expressed in trans.
  • hybrid with a fusion joint at the extracellular surface gives no syncytia while hybrid B (in the middle of the transmembrane regions) gives approximately five-fold less syncytium on Sup Tl cells than hybrid C.
  • Example 3 illustrates one hybrid protein produced from two different retroviruses
  • the possibili ⁇ ties are not limited to retroviruses or other viruses.
  • the receptor binding portion of human inter- leukin-2 may be combined with the envelope protein of MLV to target vectors to cells with IL-2 receptors.
  • a recombination would preferably be located in the gp 70 portion of the MLV env gene, leaving an intact pl5E protein.
  • the foregoing technique may be used to create a recombinant retrovirus with an envelope protein which recognizes antibody Fc segments.
  • Monoclonal antibodies which recognize only preselected target cells only could then be bound to such a recombinant retrovirus exhibiting such envelope proteins so that the retrovirus would bind to and infect only those preselected target cells.
  • a hybrid envelope with the binding domain of avidin would be useful for targeting cells' "images" in a patient or animal with biotinylated antibodies or other ligands. The patient would first be flooded with antibodies, and then antibody binding nonspecifically allowed to clear from the patient's system, before administering the vector.
  • the high affinity (10 "15 ) of the avidin binding site for biotin would then allow accurate and efficient targeting to the original tissue identified by the monoclonal "image.”
  • the approach may also be used to achieve tumor- specific targeting and killing by taking advantage of three levels of retroviral vector specificity; namely, cell entry, gene expression, and choice of protein expressed. Retroviral vectors enter cells and exert their effects at intracellular sites. In this respect their action is quite unique. Using this property, and the three levels of natural retroviral specificity (above) , retroviral vectors may be engineered to target and kill tumor cells.
  • the overall goal of targeting of retrovirus to tumor cells may be accomplished by two major experimental routes; namely, a) selection in tissue culture (or in animals) for retroviruses that grow preferentially in tumor cells; or b) construction of retroviral vectors with tissue (tumor) -specific promoters with improvements being made by .in vitro passage, and negative and positive drug- sensitivity selection.
  • Vectors suitable for selectively infecting selected cell types such as a tumor cell, may generally be prepared by (a) continuously passaging a virus in cells of the selected cell type until the virus has genetically mutated and a predominant fast growing strain has evolved;
  • At least four selective protocols may be utilized to select for retrovirus which grow preferentially in tumor cells; namely, 1) "Env Selection by Passage In Vitro,” wherein selection of retrovirus with improved replicative growth ability is accomplished by repeated passage in tumor cells; 2) “Selection with a Drug Resistance Gene,” wherein genetic selection for tumor “specific” retroviruses is based on viral constructs containing a linked drug resistance gene; 3) “Hybrid-Env,” wherein selection (by protocol ⁇ l or #2, above) of retrovirus with tumor-"specificity” is initiated from a construct containing a hybrid envelope gene which is a fusion of a tumor receptor gene (i.e., an anti-tumor antibody H-chain V-region gene fused with env; or, a growth receptor fused with env) ; in this case selection begins at a favorable starting point, e.g., an env which has some specificity for tumor cells; or 4) "Selection by Passage In Vitro and Counter Selection by Co-cul
  • tissue (tumor) -specific promoters biochemical markers with different levels of tissue-specificity are well known, and genetic control through tissue-specific promoters is understood in some systems.
  • tissue-specific promoters There are a number of genes whose transcriptional promoter elements are relatively active in rapidly growing cells (i.e., transferring receptor, thymidine kinase, etc.) and others whose promoter/enhancer elements are tissue specific (i.e., HBV enhancer for liver, PSA promoter for prostate).
  • Retroviral vectors and tissue-specific promoters (present either as an internal promoter or within the LTR) which can drive the expression of selectable markers and cell cycle genes (i.e., drug sensitivity, Eco gpt; or HSVTK in TK-cells) . Expression of these genes can be selected for in media containing mycophenolic acid or HAT, respectively. In this manner, tumor cells containing integrated provirus which actively expresses the drug resistance gene will survive. Selection in this system may involve selection for both tissue-specific promoters and viral LTRs.
  • specific expression in tumor cells can be counter- selected by periodically passaging virus onto normal cells, and selecting against virus that express Eco gpt or HSVtk (drug sensitivity) in those cells (by thioxanthine or acyclovir) .
  • Infected cells containing integrated provirus which express Eco gpt or tk phenotype will die and thus virus in that cell type will be selected against.
  • Another technique for integrating a vector construct into specific, preselected sites of a target cell's genome involves integrase modification.
  • the retrovirus pol gene product is generally processed into four parts: (i) a protease which processes the viral gag and pol products; (ii) the reverse transcriptase; and (iii) RNase H, which degrades RNA of an RNA/DNA duplex; and (iv) the endonuclease or "integrase.”
  • such "fingers” allow the protein to bind to DNA at particular sequences.
  • One illustrative example is the steroid receptors.
  • the estrogen receptor, responding to estrogens have the effect of a glucocorticoid receptor, responding to glucocorticoids, simply by substituting the glucocorticoid receptor "finger" (i.e., DNA binding segment) in place of the estrogen receptor finger segment in the estrogen receptor gene.
  • the position in the genome to which the proteins are targeted has been changed.
  • Such directing sequences can also be substituted into the integrase gene in place of the present zinc finger.
  • the segment coding for the DNA binding region of the human estrogen receptor gene may be substituted in place of the DNA binding region of the integrase in a packaging genome.
  • specific integration would be tested by means of an n vitro integration system (Brown et al.. Cell £9.:347-356, 1987).
  • this packaging genome is used to make infectious vector particles, and infection of and integration into estrogen- sensitive and estrogen-nonsensitive cells compared in culture.
  • incoming viral vectors may be directed to integrate into preselected sites on the target cell's genome, dictated by the genome- binding properties of site-specific DNA-binding protein segments spliced into the integrase genome.
  • the integration site must, in fact, be receptive to the fingers of the modified integrase.
  • most cells are sensitive to glucocorticoids and hence their chromatin has sites for glucocorticoid receptors.
  • a modified integrase having a glucocorti ⁇ coid receptor finger would be suitable to integrate the proviral vector construct at those glucocorticoid receptor-binding sites.
  • the vector to be delivered is also inserted into a line of transgenic animals with a tissue-specific or housekeeping promoter. As discussed above, the vector can be driven off such a promoter substituting for the U3 region of the 5' LTR ( Figure 3) . This transgene could be inducible or ubiquitous in its expression. This vector, however, is not packaged.
  • These lines of animals are then mated to the gag/pol/env animal and subsequent progeny produce packaged vector.
  • the progeny which are essentially identical, are characterized and offer an unlimited source of primary producing cells.
  • primary cells making gag/pol and env and derived from transgenic animals can be infected or transfected in bulk with retrovirus vectors to make a primary cell producer line.
  • Many different transgenic animals or insects could produce these vectors, such as mice, rats, chickens, swine, rabbits, cows, sheep, fish and flies.
  • the vector and packaging genomes would be tailored for species infection specificity and tissue-specific expression through the use of tissue-specific promoters and different envelope proteins.
  • An example of such a procedure is illustrated in Figure 4. Although the following examples of transgenic production of primary packaging lines are described only for mice, these procedures can be extended to other species by those skilled in the art.
  • These transgenic animals may be produced by microinjection or gene transfer techniques. Given the homology to MLV sequences in mice genome, the final preferred animals would not be mice.
  • HPRT is a purine salvage enzyme which expresses in all tissues.
  • This promoter is inserted in front of various gag/pol fragments (e.g., Bal I/Sca I; Aat Il/Sca I; Pst I/Sca I of MoMLV; see RNA Tumor Viruses _., Cold Spring Harbor Laboratory, 1985) that are cloned in Bluescript plasmids (Strategene, Inc.) using recombinant DNA techniques (see Maniatis et al., Molecular Cloning: A Laboratory Manual. Cold Spring Harbor, 1982) . The resulting plasmids are purified (Maniatis et al. , op. cit.) and the relevant genetic information isolated using Geneclean (Bio 101) or electroelution (see Hogan et al. (eds.), Manipulating the Mouse Embryo: A
  • Transgenic-positive animals are characterized for expression levels of gag/pol proteins by immunoprecipita- tion of radiolabeled primary cells, such as fibroblast
  • the envelope proteins can be from any retrovirus that is capable of complementing the relevant gag/pol, in this case that of MLV. Examples are ecotropic MLV, amphotrophic MLV, xenotropic MLV, polytropic MLV, or hybrid envelopes. As above, the envelope gene is cloned behind the HPRT promoter using recombinant DNA techniques
  • the resulting "minigene” is isolated (see Hogan et al., op. cit.), and expression of envelope protein is determined (Harlow et al., op. cit.).
  • the transgenic envelope animals are bred to homozygosity to establish a well-characterized envelope animal.
  • gag/pol-env Animals Using Housekeeping Promoters for Ubiquitous Expression in Transgenic Animals This uses the well-characterized gag/pol animals, as well as the animals for the establishment of a permanent gag/pol/envelope animal line. This involves breeding to homozygosity and the establishment of a well- characterized line. These lines are then used to estab ⁇ lish primary mouse embryo lines that can be used for packaging vectors in tissue culture. Furthermore, animals containing the retroviral vector are bred into this line.
  • This example illustrates high level expression of the gag/pol, envelope, or hybrid envelope in specific tissues, such as T-cells.
  • the 1.5 kb Bam HI/Hind III fragment from the CD2 gene is inserted in front of gag/pol, envelope, or hybrid envelope fragments using recombinant DNA techniques. These genes are inserted into fertilized mouse ova by microinjection.
  • Transgenic animals are characterized as before. Expression in T-cells is established, and animals are bred to homozygosity to establish we11-characterized lines of transgenic animals.
  • Gag/pol animals are mated to envelope animals to establish gag/pol-env animals expressing only in T-cells.
  • the T-cells of these animals are then a source for T-cells capable . of packaging retroviral vectors.
  • vector animals can be bred into these gag/pol-env animals to establish T-cells expressing the vector.
  • tissue-specific promoters such as milk-specific (whey) , pancre- atic (insulin or elastase) , or neuronal (myelin basic protein) promoters.
  • promoters such as milk-specific promoters
  • recombinant retroviruses may be isolated directly from the biological fluid of the progeny.
  • EXAMPLE 8 Production of Either Housekeeping or Tissue-Specific Retroviral Vectors in Transgenic Animals
  • the insertion of retroviruses or retroviral vectors into the germ line of transgenic animals results in little or no expression.
  • This effect described by Jaenisch (see Jahner et al. , Nature 298:623-628. 1982), is attributed to methylation of 5' retroviral LTR sequences.
  • This technique would overcome the methylation effect by substituting either a housekeeping or tissue-specific promoter to express the retroviral vector/retrovirus.
  • the U3 region of the 5' LTR, which contains the enhancer elements, is replaced with regulatory sequences from housekeeping or tissue-specific promoters (see Figure 20) .
  • the 3' LTR is fully retained, as it contains sequences necessary for polyadenylation of the viral RNA and integration.
  • the U3 region of the 5' LTR of the integrated provirus is generated by the U3 region of the 3' LTR of the infecting virus.
  • the 3' is necessary, while the 5' 113 is dispensable.
  • Substitution of the 5' LTR U3 sequences with promoters and insertion into the germ line of transgenic animals results in lines of animals capable of producing retroviral vector transcripts. These animals would then be mated to gag/pol-env animals to generate retroviral-producing animals (see Figure 4) .
EP91919095A 1990-09-21 1991-09-20 Packaging cells Withdrawn EP0549723A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP02019446A EP1285967A3 (en) 1990-09-21 1991-09-20 Human retroviral packaging cell line
EP97121096A EP0854193A1 (en) 1990-09-21 1991-09-20 Human retroviral packaging cell line

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US58660390A 1990-09-21 1990-09-21
US586603 1990-09-21

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP97121096A Division EP0854193A1 (en) 1990-09-21 1991-09-20 Human retroviral packaging cell line

Publications (1)

Publication Number Publication Date
EP0549723A1 true EP0549723A1 (en) 1993-07-07

Family

ID=24346413

Family Applications (3)

Application Number Title Priority Date Filing Date
EP02019446A Withdrawn EP1285967A3 (en) 1990-09-21 1991-09-20 Human retroviral packaging cell line
EP97121096A Ceased EP0854193A1 (en) 1990-09-21 1991-09-20 Human retroviral packaging cell line
EP91919095A Withdrawn EP0549723A1 (en) 1990-09-21 1991-09-20 Packaging cells

Family Applications Before (2)

Application Number Title Priority Date Filing Date
EP02019446A Withdrawn EP1285967A3 (en) 1990-09-21 1991-09-20 Human retroviral packaging cell line
EP97121096A Ceased EP0854193A1 (en) 1990-09-21 1991-09-20 Human retroviral packaging cell line

Country Status (5)

Country Link
EP (3) EP1285967A3 (es)
JP (3) JPH06500923A (es)
AU (2) AU665176B2 (es)
CA (1) CA2092195C (es)
WO (1) WO1992005266A2 (es)

Families Citing this family (120)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5662896A (en) * 1988-03-21 1997-09-02 Chiron Viagene, Inc. Compositions and methods for cancer immunotherapy
US6027722A (en) * 1990-10-25 2000-02-22 Nature Technology Corporation Vectors for gene transfer
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
WO1993020221A1 (en) * 1992-04-03 1993-10-14 Young Alexander T Gene therapy using targeted viral vectors
AU4259193A (en) * 1992-04-28 1993-11-29 Frank Andreas Harald Meyer Medicament for the gene-therapeutic treatment of human beings, animals and plants, especially to block virus multiplication and carcinogenes and process for producing the medicament
AU689755B2 (en) * 1992-09-22 1998-04-09 Biofocus Discovery Limited Recombinant viruses displaying a nonviral polypeptide on their external surface
CA2148934C (en) * 1992-11-09 2005-08-02 French W. Anderson Targetable vector particles
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
DE69434860T2 (de) * 1993-02-22 2007-03-15 The Rockefeller University Herstellung von helfer-freien retroviren mit hohem titer mittels transienter transfektion
AU699706C (en) 1993-03-12 2000-01-13 Creighton University Improved vectors for gene therapy
FR2707091B1 (fr) 1993-06-30 1997-04-04 Cohen Haguenauer Odile Vecteur rétroviral pour le transfert et l'expression de gènes dans des cellules eucaryotes.
FR2716461A1 (fr) * 1994-02-22 1995-08-25 Univ Paris Curie Système hôte-vecteur utilisable en thérapie génique.
FR2716459B1 (fr) * 1994-02-22 1996-05-10 Univ Paris Curie Système hôte-vecteur utilisable en thérapie génique.
US6013517A (en) 1994-05-09 2000-01-11 Chiron Corporation Crossless retroviral vectors
EP1881075B1 (en) 1994-05-09 2013-07-24 Oxford BioMedica (UK) Limited Retroviral vectors having a reduced recombination rate
EP0763123A4 (en) * 1994-06-07 1998-08-12 Immunomedics Inc SAFE RETROVIRAL VECTORS, THEIR PRODUCTION AND USE
US5952225A (en) * 1994-08-17 1999-09-14 Genetic Therapy, Inc. Retroviral vectors produced by producer cell lines resistant to lysis by human serum
US5698446A (en) * 1994-09-07 1997-12-16 Chiron Corporation Methods and compositions for inhibiting production of replication competent virus
DK0787191T4 (da) 1994-10-20 2012-01-30 Pasteur Institut Nukleotidsekvenser af HIV-1 type (eller undertype) O retrovirusantigener
FR2727429B1 (fr) * 1994-11-30 1997-11-28 Haguenauer Odile Cohen Lignees d'encapsidation et vecteurs d'expression pour la transcomplementation de vecteurs retroviraux defectifs
US5681746A (en) * 1994-12-30 1997-10-28 Chiron Viagene, Inc. Retroviral delivery of full length factor VIII
US6818439B1 (en) 1994-12-30 2004-11-16 Chiron Corporation Methods for administration of recombinant gene delivery vehicles for treatment of hemophilia and other disorders
US5910434A (en) * 1995-12-15 1999-06-08 Systemix, Inc. Method for obtaining retroviral packaging cell lines producing high transducing efficiency retroviral supernatant
US6365344B1 (en) 1996-01-23 2002-04-02 The Board Of Trustees Of The Leland Stanford Junior University Methods for screening for transdominant effector peptides and RNA molecules
US6455247B1 (en) 1996-01-23 2002-09-24 Board Of Trustees Of The Leland Stanford Junior University Methods for screening for transdominant effector peptides and RNA molecules
US6723531B2 (en) 1996-04-05 2004-04-20 The Salk Institute For Biological Studies Method for modulating expression of exogenous genes in mammalian systems, and products related thereto
EP0953052B1 (en) * 1996-05-06 2009-03-04 Oxford BioMedica (UK) Limited Crossless retroviral vectors
US5928638A (en) * 1996-06-17 1999-07-27 Systemix, Inc. Methods for gene transfer
US5739018A (en) * 1996-08-07 1998-04-14 The Regents Of The University Of California Packaging cell lines for pseudotyped retroviral vectors
US6133027A (en) * 1996-08-07 2000-10-17 City Of Hope Inducible expression system
EP2298900A1 (en) 1996-09-17 2011-03-23 Novartis Vaccines and Diagnostics, Inc. Compositions and methods for treating intracellular diseases
US6544523B1 (en) 1996-11-13 2003-04-08 Chiron Corporation Mutant forms of Fas ligand and uses thereof
US6300488B1 (en) 1997-07-10 2001-10-09 The Salk Institute For Biological Studies Modified lepidopteran receptors and hybrid multifunctional proteins for use in transcription and regulation of transgene expression
DE69838584T2 (de) 1997-08-04 2008-06-26 Cell Genesys, Inc., Foster City Enhancer des menschlichen glandulären kallikreingens, vektoren die ihn enthalten und methoden für seine verwendung
EP2278006A3 (en) 1997-11-06 2011-03-02 Novartis Vaccines and Diagnostics S.r.l. Neisserial antigens
US6914131B1 (en) 1998-10-09 2005-07-05 Chiron S.R.L. Neisserial antigens
CA2317815A1 (en) 1998-01-14 1999-07-22 Chiron S.P.A. Neisseria meningitidis antigens
EP1645631B1 (en) 1998-05-01 2007-10-24 Novartis Vaccines and Diagnostics, Inc. Neisseria antigens and compositions
US6333318B1 (en) 1998-05-14 2001-12-25 The Salk Institute For Biological Studies Formulations useful for modulating expression of exogenous genes in mammalian systems, and products related thereto
DE69938190T2 (de) 1998-10-15 2009-03-05 Novartis Vaccines and Diagnostics, Inc., Emeryville Gene mit veränderter expression in metastatischen brust- oder dickdarm- krebszellen
US20030035798A1 (en) 2000-08-16 2003-02-20 Fang Fang Humanized antibodies
ES2310055T3 (es) 1998-12-16 2008-12-16 Novartis Vaccines & Diagnostic Quinasa humana dependiente de ciclina (hpnqalre).
WO2000055343A2 (en) * 1999-03-15 2000-09-21 Chiron Corporation Retrovirus producting cells utilizing a high multiplicity of transduction
ES2477194T3 (es) 1999-04-30 2014-07-16 Novartis Vaccines And Diagnostics S.R.L. Ant�genos neisseriales conservados
GB9911683D0 (en) 1999-05-19 1999-07-21 Chiron Spa Antigenic peptides
US7504253B2 (en) 1999-06-11 2009-03-17 The Burnham Institute For Medical Research Nucleic acid encoding proteins involved in protein degradation, products and methods related thereof
GB9916529D0 (en) 1999-07-14 1999-09-15 Chiron Spa Antigenic peptides
US7057015B1 (en) 1999-10-20 2006-06-06 The Salk Institute For Biological Studies Hormone receptor functional dimers and methods of their use
DK2275552T3 (en) 1999-10-29 2015-12-07 Glaxosmithkline Biolog Sa Neisserial antigenic peptides
ATE446365T1 (de) 1999-11-18 2009-11-15 Novartis Vaccines & Diagnostic Menschliches fgf-21 gen und genexpressionsprodukte
DK2289545T3 (en) 2000-01-17 2016-09-05 Glaxosmithkline Biologicals Sa Supplemented OMV vaccine against meningococcus
BR0108711A (pt) 2000-02-28 2004-06-22 Chiron Spa Expressão heteróloga de proteìnas de neisseria
US20020082205A1 (en) 2000-03-08 2002-06-27 Nobuyuki Itoh Human FGF-23 gene and gene expression products
CA2403205A1 (en) 2000-03-13 2001-09-20 Engene, Inc. Compositions and methods of regulated protein expression in gut endocrine k cells
GB0009760D0 (en) 2000-04-19 2000-06-07 Oxford Biomedica Ltd Method
GB0024550D0 (es) 2000-10-06 2000-11-22 Oxford Biomedica Ltd
CA2881568C (en) 2000-10-27 2019-09-24 Novartis Vaccines And Diagnostics, Inc. Nucleic acids and proteins from streptococcus groups a & b
GB0107661D0 (en) 2001-03-27 2001-05-16 Chiron Spa Staphylococcus aureus
GB0107658D0 (en) 2001-03-27 2001-05-16 Chiron Spa Streptococcus pneumoniae
WO2002081639A2 (en) 2001-04-06 2002-10-17 Georgetown University Gene brcc2 and diagnostic and therapeutic uses thereof
AU2002305151A1 (en) 2001-04-06 2002-10-21 Georgetown University Gene scc-112 and diagnostic and therapeutic uses thereof
WO2002081642A2 (en) 2001-04-06 2002-10-17 Georgetown University Gene brcc-3 and diagnostic and therapeutic uses thereof
CA2344208A1 (en) 2001-04-30 2002-10-30 Oxford Biomedica (Uk) Limited Method
CN100515494C (zh) 2001-12-12 2009-07-22 启龙有限公司 抗沙眼衣原体的免疫
ES2411981T3 (es) 2002-02-01 2013-07-09 Oxford Biomedica (Uk) Limited Vector lentivírico
US7138512B2 (en) 2002-04-10 2006-11-21 Georgetown University Gene SHINC-2 and diagnostic and therapeutic uses thereof
US7244565B2 (en) 2002-04-10 2007-07-17 Georgetown University Gene shinc-3 and diagnostic and therapeutic uses thereof
WO2004046177A2 (en) 2002-11-15 2004-06-03 Chiron Srl Unexpected surface proteins in neisseria meningitidis
GB0308198D0 (en) 2003-04-09 2003-05-14 Chiron Srl ADP-ribosylating bacterial toxin
ES2407465T3 (es) 2004-03-29 2013-06-12 Galpharma Co., Ltd. Nueva proteína galectina 9 modificada y uso de la misma
WO2006007539A1 (en) * 2004-07-01 2006-01-19 Virxsys Corporation Vector packaging cell line
CA2524619A1 (en) 2005-11-22 2007-05-22 Ottawa Health Research Institute Novel stem cells, nucleotide sequences and proteins therefrom
TWI461436B (zh) 2005-11-25 2014-11-21 Kyowa Hakko Kirin Co Ltd 人類cd134(ox40)之人類單株抗體及其製造及使用方法
WO2008020335A2 (en) 2006-06-09 2008-02-21 Novartis Ag Immunogenic compositions for streptococcus agalactiae
JP2010510772A (ja) 2006-11-27 2010-04-08 パトリス リミテッド 新生細胞における新規なグリコシル化ペプチド標的
JP5829377B2 (ja) 2007-02-06 2015-12-09 ジュン ヨー,タイ 遺伝子治療を使用する神経変性疾患の治療および予防
EP2139447A2 (en) 2007-03-20 2010-01-06 Harold Brem Gm-csf cosmeceutical compositions and methods of use thereof
EP2185694A2 (en) 2007-08-02 2010-05-19 California Stem Cell, Inc. Neuronal progenitor cells and methods of derivation and purification of neuronal progenitor cells from embryonic stem cells
US9593340B2 (en) 2007-10-15 2017-03-14 Admedus Vaccines Pty Ltd. Expression system for modulating an immune response
US8324182B2 (en) 2008-02-15 2012-12-04 Tufts University Humanized model of membrane attack complex (MAC) formation on murine retina and compositions, kits and methods for treatment of macular degeneration
US8877896B2 (en) 2008-02-15 2014-11-04 Tufts University Compositions, methods and kits for modeling, diagnosing, and treating complement disorders
US8999919B2 (en) 2008-10-22 2015-04-07 Trustees Of Dartmouth College Compositions and methods for inhibiting the interaction between CFTR and CAL
US8211487B2 (en) 2008-11-26 2012-07-03 Srinivasan Damodaran Inhibition of ice crystal growth
ES2571235T3 (es) 2009-04-10 2016-05-24 Kyowa Hakko Kirin Co Ltd Procedimiento para el tratamiento de un tumor sanguíneo que utiliza el anticuerpo anti-TIM-3
KR101732201B1 (ko) 2009-04-27 2017-05-02 교와 핫꼬 기린 가부시키가이샤 혈액 종양 치료를 목적으로 하는 항IL-3Rα 항체
EP3546583A1 (en) * 2009-06-17 2019-10-02 Tocagen Inc. Producer cells for replication competent retroviral vectors
US9556272B2 (en) 2009-11-11 2017-01-31 The Trustees Of The University Of Pennsylvania Anti-TEM1 antibodies and uses thereof
GB0920775D0 (en) 2009-11-26 2010-01-13 King S College Cells
US9795658B2 (en) 2010-04-20 2017-10-24 Admedus Vaccines Pty Ltd Expression system for modulating an immune response
CA2843684A1 (en) 2010-08-13 2012-02-16 Tufts University Compositions, kits and methods for treatment of macular degeneration using soluble membrane-independent cd59 protein
WO2012075243A2 (en) 2010-12-01 2012-06-07 The University Of North Carolina At Chapel Hill Methods and compositions for targeting sites of neovascular growth
CN103998057A (zh) 2011-09-15 2014-08-20 美国加州大学洛杉矶海滨分校医学中心的洛杉矶生物医学研究所 利用CotH的毛霉菌病的免疫疗法和诊断
WO2013162748A1 (en) 2012-04-27 2013-10-31 The Trustees Of The University Of Pennsylvania Anti-tumor endothelial marker-1 (tem1) antibody variants and uses thereof
US9790269B2 (en) 2013-02-08 2017-10-17 Misfolding Diagnostics, Inc. Transthyretin antibodies and uses thereof
US10166300B2 (en) 2013-06-04 2019-01-01 Virginia Commonwealth University Tripartite cancer theranostic nucleic acid constructs
WO2014197535A1 (en) 2013-06-04 2014-12-11 Virginia Commonwealth University Recombinant cancer therapeutic cytokine
EP3004872B1 (en) 2013-06-04 2017-10-18 Virginia Commonwealth University Mda-9/syntenin promoter to image and treat metastatic cancer cells
EP3003022B1 (en) 2013-06-04 2017-11-22 Virginia Commonwealth University Use of a truncated ccn1 promoter for cancer diagnostics, therapeutics and theranostics
EP3004357A4 (en) 2013-06-04 2017-01-11 The Johns Hopkins University Peg-prom mediated surface expression of avidin/streptavidin
WO2014210142A1 (en) 2013-06-25 2014-12-31 Temple University-Of The Commonwealth System Of Higher Education Cortical bone-derived stem cells
GB201318804D0 (en) 2013-10-24 2013-12-11 Adaptimmune Ltd Vectors for transgene expression
CA2943640A1 (en) 2014-03-26 2015-10-01 Tocagen Inc. A retroviral vector having immune-stimulating activity
EP3186269B1 (en) 2014-08-28 2021-03-10 Tufts University Compositions, methods and kits for treating complement related disorders
GB201415344D0 (en) 2014-08-29 2014-10-15 Ucl Business Plc Protein
US9901639B2 (en) 2015-02-13 2018-02-27 Temple University—Of the Commonwealth System of Higher Education Bone marrow origin progenitor cell or endothelial progenitor cell in combination with DNMT1 gene therapy for vascular repair in metabolic disease
US10149887B2 (en) 2015-10-23 2018-12-11 Canbas Co., Ltd. Peptides and peptidomimetics in combination with t cell activating and/or checkpoint inhibiting agents for cancer treatment
AU2016367712B2 (en) 2015-12-09 2021-10-07 Jingang Medicine (Australia) Pty Ltd Immunomodulating composition for treatment
GB201603372D0 (en) * 2016-02-26 2016-04-13 Ucl Business Plc Cell
EP3299460A1 (en) 2016-09-26 2018-03-28 Johann Wolfgang Goethe-Universität Frankfurt am Main Novel compounds and methods for modulating ubiquitination
GB201710973D0 (en) 2017-07-07 2017-08-23 Avacta Life Sciences Ltd Scaffold proteins
US20210052743A1 (en) 2018-02-12 2021-02-25 Trustees Of Tufts College Cd59 for inhibiting inflammasome activation
MX2022001732A (es) 2019-08-12 2022-05-06 Purinomia Biotech Inc Metodos y composiciones para promover y potenciar la respuesta inmunitaria mediada por linfocitos t dirigida a la adcc de las celulas con expresion de cd39.
TW202128775A (zh) 2019-10-16 2021-08-01 英商阿法克塔生命科學有限公司 PD-L1抑制劑-TGFβ抑制劑雙特異性藥物部分
DE102020106710A1 (de) 2020-03-11 2021-09-16 Immatics US, Inc. Wpre-mutantenkonstrukte, zusammensetzungen und zugehörige verfahren
DE102020111571A1 (de) 2020-03-11 2021-09-16 Immatics US, Inc. Wpre-mutantenkonstrukte, zusammensetzungen und zugehörige verfahren
GB202101299D0 (en) 2020-06-09 2021-03-17 Avacta Life Sciences Ltd Diagnostic polypetides and methods
WO2022234003A1 (en) 2021-05-07 2022-11-10 Avacta Life Sciences Limited Cd33 binding polypeptides with stefin a protein
WO2023057946A1 (en) 2021-10-07 2023-04-13 Avacta Life Sciences Limited Serum half-life extended pd-l1 binding polypeptides
TW202334196A (zh) 2021-10-07 2023-09-01 英商阿法克塔生命科學有限公司 Pd-l1結合多肽
WO2023089564A1 (en) 2021-11-19 2023-05-25 Janssen Biotech, Inc. Method of treating geographic atrophy with a gene therapy vector expressing soluble cd59

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1988008454A1 (en) * 1987-04-23 1988-11-03 State Of Oregon Acting By And Through The State Bo Process for amplifying expression and transmission of cloned genes in eukaryotic cells
JPH03504079A (ja) * 1988-03-21 1991-09-12 カイロン コーポレイション 組換えレトロウィルス
JP3140757B2 (ja) * 1989-02-06 2001-03-05 デイナ・フアーバー・キヤンサー・インステイテユート パッケージング欠陥hivプロウイルス、細胞系及びその使用
EP0845537A1 (en) * 1989-08-18 1998-06-03 Chiron Corporation Recombinant retroviruses delivering vector constructs to target cells
WO1991019803A1 (en) * 1990-06-19 1991-12-26 Applied Biotechnology, Incorporated Self assembled, defective, nonself-propagating viral particles

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9205266A2 *

Also Published As

Publication number Publication date
EP0854193A1 (en) 1998-07-22
AU8842491A (en) 1992-04-15
JP2003159062A (ja) 2003-06-03
EP1285967A3 (en) 2005-03-02
WO1992005266A3 (en) 1992-07-23
AU4798496A (en) 1996-06-20
JP4092278B2 (ja) 2008-05-28
AU690427B2 (en) 1998-04-23
CA2092195A1 (en) 1992-03-22
CA2092195C (en) 2000-04-18
EP1285967A2 (en) 2003-02-26
JP2004041234A (ja) 2004-02-12
WO1992005266A2 (en) 1992-04-02
JPH06500923A (ja) 1994-01-27
AU665176B2 (en) 1995-12-21

Similar Documents

Publication Publication Date Title
AU665176B2 (en) Packaging cells
US5716832A (en) Packaging cells
US5817491A (en) VSV G pseusdotyped retroviral vectors
US7070994B2 (en) Packaging cells
US5856185A (en) Method for making reflection defective retroviral vectors for infecting human cells
JP3866760B2 (ja) 一過性トランスフェクションによる高力価のヘルパーフリーのレトロウイルスの生産
EP1030861A1 (en) Modification of virus tropism and host range by viral genome shuffling
US6096534A (en) Retrovirus vectors derived from avian sarcoma leukosis viruses permitting transfer of genes into mammalian cells
US5766945A (en) 10A1 Retroviral packaging cells and uses thereof
Somia Gene transfer by retroviral vectors: an overview
US20030051259A1 (en) Packaging cells
AU4312699A (en) Lentiviral vectors
Schambach et al. Retroviral vectors for cell and gene therapy
AU773576B2 (en) Production of high titer helper-free retroviruses by transient transfection
US20030229903A1 (en) Novel system for the evaluation of the activity and/or specificity of a viral component
WO1998050538A1 (en) Mus dunni endogenous retroviral packaging cell lines
Garoff et al. Alphavirus-Retrovirus Vectors Henrik Garoff and Kejun Li Karolinska Institutet, Dept. of Biosciences at Novum 141 57 HUDDINGE, Sweden

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19930326

AK Designated contracting states

Kind code of ref document: A1

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

17Q First examination report despatched

Effective date: 19930715

18D Application deemed to be withdrawn

Effective date: 19941206

D18D Application deemed to be withdrawn (deleted)
RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CHIRON VIAGENE, INC.

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CHIRON CORPORATION

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

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

R18D Application deemed to be withdrawn (corrected)

Effective date: 19980217