US20140154215A1 - Herpes virus strains - Google Patents

Herpes virus strains Download PDF

Info

Publication number
US20140154215A1
US20140154215A1 US14/174,521 US201414174521A US2014154215A1 US 20140154215 A1 US20140154215 A1 US 20140154215A1 US 201414174521 A US201414174521 A US 201414174521A US 2014154215 A1 US2014154215 A1 US 2014154215A1
Authority
US
United States
Prior art keywords
virus
strain
gene
functional
cells
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.)
Abandoned
Application number
US14/174,521
Inventor
Robert S. Coffin
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.)
Biovex Ltd
Amgen Inc
Original Assignee
Amgen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27447756&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20140154215(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from GB0001475A external-priority patent/GB0001475D0/en
Priority claimed from GB0002854A external-priority patent/GB0002854D0/en
Priority claimed from GB0100288A external-priority patent/GB0100288D0/en
Priority claimed from GB0100430A external-priority patent/GB0100430D0/en
Application filed by Amgen Inc filed Critical Amgen Inc
Priority to US14/174,521 priority Critical patent/US20140154215A1/en
Publication of US20140154215A1 publication Critical patent/US20140154215A1/en
Assigned to BIOVEX LIMITED reassignment BIOVEX LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: COFFIN, ROBERT S.
Priority to US14/700,036 priority patent/US20150232812A1/en
Priority to US16/376,089 priority patent/US20200032219A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/763Herpes virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction 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
    • C12N15/869Herpesviral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55516Proteins; Peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16021Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16033Use of viral protein as therapeutic agent other than vaccine, e.g. apoptosis inducing or anti-inflammatory
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16632Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16611Simplexvirus, e.g. human herpesvirus 1, 2
    • C12N2710/16641Use of virus, viral particle or viral elements as a vector
    • C12N2710/16643Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • the present invention relates to herpes virus strains with improved anti-tumour activity as compared to previously known strains.
  • viruses have been shown to have utility in a variety of applications in biotechnology and medicine on many occasions. Each is due to the unique ability of viruses to enter cells at high efficiency. This is followed in such applications by either virus gene expression and replication and/or expression of an inserted heterologous gene.
  • viruses can either deliver and express genes in cells (either viral or other genes) which may be useful in for example gene therapy or the development of vaccines, or they may be useful in selectively killing cells by lytic replication or the action of a delivered gene in for example cancer.
  • Herpes simplex virus has been suggested to be of use for the oncolytic treatment of cancer.
  • the virus must however be disabled such that it is no longer pathogenic, i.e. does not replicate in and kill non-tumor cells, but such that it can still enter and kill tumor cells.
  • HSV Herpes simplex virus
  • a number of mutations to HSV have been identified which still allow the virus to replicate in culture or in actively dividing cells in vivo (e.g. in tumors), but which prevent significant replication in normal tissue.
  • Such mutations include disruption of the genes encoding ICP34.5, ICP6, and thymidine kinase.
  • viruses with mutations to ICP34.5, or ICP34.5 together with mutation of e.g. ICP6 have so far shown the most favourable safety profile.
  • Viruses deleted for only ICP34.5 have been shown to replicate in many tumor cell types in vitro and to selectively replicate in artificially induced brain tumors in mice while sparing surrounding tissue. Early stage clinical trials have also shown their safety in man.
  • HSV ICP47 protein specifically inhibits antigen presentation in HSV infected cells (Hill et al 1995), and the product of the UL43 gene and the vhs protein reduce the immune-stimulating abilities of dendritic cells infected with HSV.
  • ICP47 and/or dendritic cell-inactivating genes might therefore usefully be deleted from an oncolytic HSV mutant virus used for the treatment of cancer, particularly if immune effects are to be enhanced through the use of GM-CSF or other immunostimulatory cytokine or chemokine GM-CSF has recently been shown to give an enhanced anti-tumor immune effect if expressed from within a tumor cell rather than administered systemically (Shi et al 1999).
  • an oncolytic HSV mutant would be inoculated into a primary or secondary tumor where replication and oncolytic destruction of the tumor would occur.
  • Immune responses would also be stimulated against the HSV infected cells, and also to tumor cells elsewhere which had spread from the primary tumor site.
  • the present invention provides viruses with improved capabilities for the lytic destruction of tumor cells in vivo.
  • herpes simplex virus strains are constructed using a strain of HSV1 or HSV2 in which the genes encoding ICP34.5 and ICP47 have been inactivated such that a functional ICP34.5 or ICP47 protein cannot be expressed and which also carries a gene encoding an immunomodulatory protein.
  • the virus may also be mutated for any additional gene(s) which may be involved in inhibiting the function of dendritic cells including the UL43 gene and/or the gene encoding vhs.
  • the present invention therefore provides viruses capable of the oncolytic destruction of tumor cells and in which anti-tumor immune effects will have been maximised.
  • FIG. 1 Viruses
  • diagrams show: laboratory HSV1 strain 17+, clinical HSV1 strain JS1, strain 17+/ICP34.5-, strain JS1/ICP34.5-, strain JS1/ICP34.5-/IC47-/hGMCSF, strain JS1/ICP34.5-/ICP47-/mGMCSF.
  • a herpes virus of the invention is capable of efficiently infecting target tumor cells and the genes encoding ICP34.5 and ICP47 are inactivated in the virus. Mutation of ICP34.5 allows selective oncolytic activity. Such mutations are described in Chou et al 1990 and Maclean et al 1991, although any mutation in which ICP34.5 is non-functional may be used.
  • the genes encoding ICP6 and/or thymidine kinase may additionally be inactivated, as may other genes if such inactivation does significantly reduce the oncolytic effect, or if such deletion enhances oncolytic or other desirable properties of the virus.
  • ICP47 usually functions to block antigen presentation in HSV-infected cells so its disruption leads to a virus that does not confer on infected tumour cells properties that might protect them from the host's immune system when infected with HSV.
  • Viruses of the invention additionally encode an immunomodulatory protein, preferably GM-CSF, but may also encode other cytokines, chemokines such as RANTES, or other immune-modulatory proteins such as B7.1, B7.2 or CD40L. Genes encoding immunomodulatory proteins may be included individually or in combination.
  • Viral regions altered for the purposes described above may be either eliminated (completely or partly), or made non-functional, or substituted by other sequences, in particular by a gene for an immunomodulatory protein such as GM-CSF.
  • the virus of the invention may be derived from a HSV1 or HSV2 strain, or from a derivative thereof, preferably HSV1.
  • Derivatives include inter-type recombinants containing DNA from HSV1 and HSV2 strains. Such inter-type recombinants are described in the art, for example in Thompson et al, 1988 and Meignier et al, 1988.
  • Derivatives preferably have at least 70% sequence homology to either the HSV1 or HSV2 genomes, more preferably at least 80%, even more preferably at least 90 or 95%. More preferably, a derivative has at least 70% sequence identity to either the HSV1 or HSV2 genome, more preferably at least 80% identity, even more preferably at least 90%, 95% or 98% identity.
  • the UWGCG Package provides the BESTFIT program which can be used to calculate homology (for example used on its default settings) (Devereux et al. (1984) Nucleic Acids Research 12, p387-395).
  • the PILEUP and BLAST algorithms can be used to calculate homology or line up sequences (typically on their default settings), for example as described in Altschul (1993) J. Mol. Evol. 36:290-300; Altschul et al. (1990) J. Mol. Biol. 215:403-10.
  • HSPs high scoring sequence pair
  • Extensions for the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment.
  • the BLAST algorithm performs a statistical analysis of the similarity between two sequences; see e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-5787.
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a sequence is considered similar to another sequence if the smallest sum probability in comparison of the first sequence to the second sequence is less than about 1, preferably less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
  • a derivative may have the sequence of a HSV1 or HSV2 genome modified by nucleotide substitutions, for example from 1, 2 or 3 to 10, 25, 50 or 100 substitutions.
  • the HSV1 or HSV2 genome may alternatively or additionally be modified by one or more insertions and/or deletions and/or by an extension at either or both ends.
  • Virus strains of the invention may be “non-laboratory” strains. These can also be referred to as “clinical” strains. A person of skill in the art will readily be able to distinguish between a laboratory strain and a non-laboratory, or clinical, strain. Further guidance on the properties likely to be exhibited by virus strains is given below.
  • the key distinction between a laboratory and non-laboratory strain is that laboratory strains currently in common use have been maintained for long periods, many years in some cases, in culture.
  • the culture of viruses such as HSV involves a technique known as serial passage. To grow and maintain viruses, suitable cells are infected with the virus, the virus replicates within the cell and the virus is then harvested; fresh cells are then re-infected, this process constitutes one cycle of serial passage. Each such cycle may take, for example, a few days in the case of HSV.
  • serial passaging may lead to changes in the properties of the virus strain, in that selection takes places for properties that favour growth in culture (e.g. rapid replication), as opposed to properties useful for practical applications, e.g. maintenance of the capacity to travel along axons in the case of HSV or to infect human cells.
  • Virus strains of the invention are may be non-laboratory strains in that they are derived from strains recently isolated from infected individuals. Strains of the invention are modified compared to the original clinical isolates, and may have spent a time in culture, but any time spent in culture will be comparatively short. Strains of the invention are prepared in such a manner as to retain substantially the desirable properties of the original clinical isolates from which they are derived.
  • a virus strain of the invention is derived from a parental virus strain if the parental virus strain is mutated to produce the virus.
  • a virus of the invention may be derived from the clinical isolate JSI.
  • the parental strain of such a JSI-derived virus may be JSI or another HSV1 strain derived from JSI.
  • a virus of the invention may be a JSI virus comprising a gene encoding an immunomodulatory protein and which lacks a functional ICP34.5 encoding gene and a functional ICP47 encoding gene.
  • such a virus may contain any other mutation as mentioned herein.
  • a virus of the invention is capable of efficiently infecting target human cancer cells.
  • a virus When such a virus is a non-laboratory or clinical strain it will have been recently isolated from an HSV infected individual and then screened for the desired ability of enhanced replication, infection or killing of tumour and/or other cells in vitro and/or in vivo in comparison to standard laboratory strains.
  • Such viruses of the invention with improved properties as compared to laboratory virus strains are then engineered such that they lack functional ICP34.5 and ICP47 genes and encode a gene(s) for an immunomodulatory protein(s) such as GM-CSF under the control of a suitable promoter(s).
  • Other genes encoding proteins which interfere with the function of dendritic cells such as UL43 and/or vhs may also be inactivated.
  • a non-laboratory virus strain of the invention has undergone three years or less in culture since isolation of its unmodified clinical precursor strain from its host. More preferably, the strain has undergone one year or less in culture, for example nine months or less, six months or less, three months or less, or two months or less, one month or less, two weeks or less, or one week or less.
  • time in culture is meant time actually spent in culture.
  • it is a common practice to freeze virus strains in order to preserve them.
  • preserving by freezing or in an equivalent manner does not qualify as maintaining the strain in culture.
  • time spent frozen or otherwise preserved is not included in the above definitions of time spent in culture.
  • Time spent in culture is typically time actually spent undergoing serial passage, i.e. time during which selection for undesirable characteristics can occur.
  • a non-laboratory virus strain has undergone 1,000 or less cycles or serial passage since isolation of its unmodified clinical precursor strain from its host. More preferably, it has undergone 500 or less, 100 or less, 90 or less, 80 or less, 70 or less, 60 or less, 50 or less, 40 or less, 30 or less, 20 or less, or 10 or less such cycles.
  • a non-laboratory virus has a greater ability, as measured by standard statistical tests, than a reference laboratory strain with the equivalent modifications to perform certain functions useful in the application at hand.
  • a non-laboratory virus strain of the invention will preferably have a greater ability than a reference laboratory strain with equivalent modifications to infect or replicate in tumour cells, to kill tumour cells or to spread between cells in tissue. More preferably, such greater ability is a statistically significantly greater ability.
  • a non-laboratory strain of the invention may have up to 1.1 fold, 1.2 fold, 1.5 fold, 2 fold, 5 fold, 10 fold, 20 fold, 50 fold, or 100 fold the capacity of the reference strain in respect of the property being tested.
  • viruses of the invention infect and replicate in tumour cells, subsequently killing the tumour cells.
  • viruses are replication competent.
  • they are selectively replication competent in tumour cells. This means that either they replicate in tumour cells and not in non-tumour cells, or that they replicate more effectively in tumour cells than in non-tumour cells.
  • Cells in which the virus is able to replicate are permissive cells. Measurement of selective replication competence can be carried out by the tests described herein for measurement of replication and tumour cell-killing capacity, and also analysed by the statistical techniques mentioned herein if desired.
  • a virus of the invention preferably has a greater ability than an unmodified parent strain to infect or replicate in a tumour cell, to kill tumour cells or to spread between cells in tissues. Preferably this ability is a statistically significant greater ability.
  • a virus according to the invention may have up to 1.1 fold, 1.2 fold, 1.5 fold, 2 fold, 5 fold, 10 fold, 20 fold, 50 fold or 100 fold the capacity of the unmodified parent strain in respect of the property being tested.
  • the properties of the virus strain in respect of tumour cells can be measured in any manner known in the art.
  • the capacity of a virus to infect a tumour cell can be quantified by measuring the dose of virus required to measure a given percentage of cells, for example 50% or 80% of cells.
  • the capacity to replicate in a tumour cell can be measured by growth measurements such as those carried out in the Examples, e.g. by measuring virus growth in cells over a period of 6, 12, 24, 36, 48 or 72 hours or longer.
  • tumour cells The ability of a virus to kill tumour cells can be roughly quantitated by eye or more exactly quantitated by counting the number of live cells that remain over time for a given time point and MOI for given cell type. For example, comparisons may be made over 24, 48 or 72 hours and using any known tumour cell type.
  • HT29 colorectal adenocarcinoma, LNCaP.FGC prostate adenocarcinoma, MDA-MB-231 breast adenocarcinoma, SK-MEL-28 malignant melanoma or U-87 MG glioblastoma astrocytoma cells can be used. Any one of these cell types or any combination of these cell types can be used, as may other tumour cell types.
  • tumour cell types it may be desirable to construct a standard panel of tumour cell types for this purpose.
  • the number of trypan blue-excluding cells i.e. live cells
  • Quantitation may also be carried out by fluorescence activated cell sorting (FACS) or MTT assay.
  • FACS fluorescence activated cell sorting
  • MTT assay MTT assay.
  • Tumour cell-killing ability may also be measured in vivo, e.g. by measuring the reduction in tumour volume engendered by a particular virus.
  • a standard laboratory reference strain for comparison. Any suitable standard laboratory reference strain may be used.
  • HSV it is preferred to use one or more of HSV1 strain 17+, HSV1 strain F or HSV1 strain KOS.
  • the reference strain will typically have equivalent modifications to the strain of the invention being tested.
  • the reference strain will typically have equivalent modifications gene deletions and, such as heterologous gene insertions.
  • ICP34.5 and ICP47-encoding genes have been rendered non-functional, then they will also have been rendered non-functional in the reference strain.
  • the modifications made to the reference strain may be identical to those made to the strain of the invention.
  • the gene disruptions in the reference strain will be in exactly equivalent positions to those in the strain of the invention, e.g. deletions will be of the same size and in the same place.
  • heterologous genes will be inserted in the same place, driven by the same promoter, etc.
  • the various genes referred to may be rendered functionally inactive by several techniques well known in the art. For example, they may be rendered functionally inactive by deletion(s), substitution(s) or insertion(s), preferably by deletion. Deletions may remove one or more portions of the gene or the entire gene. For example, deletion of only one nucleotide may be made, resulting in a frame shift. However, preferably a larger deletion( ) is made, for example at least 25%, more preferably at least 50% of the total coding and non-coding sequence (or alternatively, in absolute terms, at least 10 nucleotides, more preferably at least 100 nucleotides, most preferably, at least 1000 nucleotides). It is particularly preferred to remove the entire gene and some of the flanking sequences. Where two or more copies of the gene are present in the viral genome it is preferred that both copies of the gene are rendered functionally inactive.
  • HSV genomic DNA is transfected together with a vector, preferably a plasmid vector, comprising the mutated sequence flanked by homologous HSV sequences.
  • the mutated sequence may comprise a deletion(s), insertion(s) or substitution(s), all of which may be constructed by routine techniques.
  • Insertions may include selectable marker genes, for example lacZ or green fluorescent protein (GFP), for screening recombinant viruses, for example, Thgalactosidase activity or fluorescence.
  • GFP green fluorescent protein
  • the viruses of the invention may be modified to carry a heterologous gene encoding an immunomodulatory protein.
  • the immunomodulatory protein will enhance the anti-tumour activity of the virus. More preferably the protein is GM-CSF or another cytokine, a chemokine such as RANTES, or another immunomodulatory molecule such as B7.1, B7.2 or CD40L. Most preferably the immunomodulatory molecule is GM-CSF.
  • the immunomodulatory gene may be any allelic variant of a wild-type gene, or it may be a mutant gene.
  • the immunomodulatory gene will be derived from a mammal, preferably a rodent or primate, more preferably a human.
  • the immunomodulatory gene is preferably operably linked to a control sequence permitting expression of said gene in a cell in vivo.
  • Viruses of the invention may thus be used to deliver the immunomodulatory gene (or genes) to a cell in vivo where it will be expressed.
  • the immunomodulatory gene may be inserted into the viral genome by any suitable technique such as homologous recombination of HSV strains with, for example, plasmid vectors carrying the gene flanked by HSV sequences.
  • the GM-CSF gene, or other immunomodulatory gene may be introduced into a suitable plasmid vector comprising herpes viral sequences using cloning techniques well-known in the art.
  • the gene may be inserted into the viral genome at any location provided that oncolytic properties are still retained.
  • Immunomodulatory genes may be inserted at multiple sites within the virus genome. For example, from 2 to 5 genes may be inserted into the genome.
  • the transcribed sequence of the immunomodulatory gene is preferably operably linked to a control sequence permitting expression of the gene in a tumour cell.
  • operably linked refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner.
  • a control sequence “operably linked” to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequence.
  • the control sequence comprises a promoter allowing expression of the immunomodulatory gene and a signal for termination of transcription.
  • the promoter is selected from promoters which are functional in mammalian, preferably human tumour cells.
  • the promoter may be derived from promoter sequences of eukaryotic genes.
  • the promoter may be derived from the genome of a cell in which expression of the heterologous gene is to occur, preferably a mammalian, preferably a human tumour cell.
  • eukaryotic promoters they may be promoters that function in a ubiquitous manner (such as promoters of ⁇ -actin, tubulin) or, alternatively, in a tumour-specific manner.
  • Viral promoters may also be used, for example the Moloney murine leukaemia virus long terminal repeat (MMLV LTR) promoter or other retroviral promoters, the human or mouse cytomegalovirus (CMV) IE promoter, or promoters of herpes virus genes including those driving expression of the latency associated transcripts.
  • MMLV LTR Moloney murine leukaemia virus long terminal repeat
  • CMV human or mouse cytomegalovirus
  • herpes virus genes including those driving expression of the latency associated transcripts.
  • Expression cassettes and other suitable constructs comprising the immunomodulatory gene and control sequences can be made using routine cloning techniques known to persons skilled in the art (see, for example, Sambrook et al., 1989, Molecular Cloning—A laboratory manual; Cold Spring Harbor Press).
  • a virus of the invention may further comprise a hererologous gene encoding the tet repressor/VP16 transcriptional activator fusion protein under the control of a strong promoter (e.g. the CMV IE promoter) and the immunomodulatory gene may be under the control of a promoter responsive to the tet repressor VP16 transcriptional activator fusion protein previously reported (Gossen and Bujard, 1992, Gossen et al, 1995).
  • a strong promoter e.g. the CMV IE promoter
  • expression of the immunomodulatory gene would depend on the presence or absence of tetracycline.
  • a virus of the invention may comprise two or more immunomodulatory genes, for example from 2 to 3, 4 or 5 immunomodulatory genes. More than one gene and associated control sequences could be introduced into a particular HSV strain either at a single site or at multiple sites in the virus genome. Alternatively pairs of promoters (the same or different promoters) facing in opposite orientations away from each other, each driving the expression of an immunomodulatory gene may be used.
  • viruses of the invention may be used in methods of cancer therapy of the human or animal body.
  • viruses of the invention may be used in the oncolytic treatment of cancer, either with or without additional pro-drug therapy or stimulation of an anti-tumour immune response.
  • Viruses of the invention may be used in the therapeutic treatment of any solid tumour in a mammal, preferably in a human.
  • viruses of the invention may be administered to a subject with prostate, breast, lung, liver, endometrial, bladder, colon or cervical carcinoma; adenocarcinoma; melanoma; lymphoma; glioma; or sarcomas such as soft tissue and bone sarcomas.
  • the invention provides the use of a herpes virus according to the invention in the manufacture of a medicament for the treatment of cancer.
  • the medicament may be used in a patient, preferably a human patient, in need of treatment.
  • a patient in need of treatment is an individual suffering from cancer, preferably an individual with a solid tumour.
  • the aim of therapeutic treatment using the medicament is to improve the condition of a patient.
  • therapeutic treatment using a virus of the invention allieviates the symptoms of the cancer.
  • Administration of the medicament to an individual suffering from a tumour will typically kill the cells of the tumour thus decreasing the size of the tumour and/or preventing spread of malignant cells from the tumour.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the virus and a pharmaceutically acceptable carrier or diluent.
  • Suitable carriers and diluents include isotonic saline solutions, for example phosphate-buffered saline.
  • Therapeutic treatment may be carried out following direct injection of the virus composition into target tissue which may be the tumour or a blood vessel supplying the tumour.
  • the amount of virus administered is in the case of HSV in the range of from 10 4 to 10 10 pfu, preferably from 10 5 to 10 8 pfu, more preferably about 10 6 to 10 8 pfu.
  • up to 500:1 typically from 1 to 200 ⁇ l preferably from 1 to 10 ⁇ l of a pharmaceutical composition consisting essentially of the virus and a pharmaceutically acceptable suitable carrier or diluent would be used for injection.
  • larger volumes up to 10 ml may also be used, depending on the tumour and the inoculation site.
  • the routes of administration and dosages described are intended only as a guide since a skilled practitioner will be able to determine readily the optimum route of administration and dosage.
  • the dosage may be determined according to various parameters, especially according to the location of the tumour, the size of the tumour, the age, weight and condition of the patient to be treated and the route of administration.
  • the virus is administered by direct injection into the tumour.
  • the virus may also be administered systemically or by injection into a blood vessel supplying the tumour.
  • the optimum route of administration will depend on the location and size of the tumour.
  • Herpes simplex type-1 virus in which the neurovirulence factor ICP34.5 is inactivated has previously been shown to direct tumour specific cell lysis in tumour models both in vitro and in vivo. Such viruses have also been shown to be safe in Phase I clinical trials by direct intra-cerebral injection in late stage glioma patients.
  • HSV 1 viruss derived from HSV1 strain 17+ or HSV1 strain F
  • HSV 1 viruses derived from HSV1 strain 17+ or HSV1 strain F
  • the viruses used were either based on HSV1 strain 17+ (a standard laboratory strain) or a clinical isolate derived from cold sores from a frequent re-activator of HSV1.
  • This clinical, or “non-laboratory”, strain is named JS1.
  • ICP34.5 was completely deleted from strain 17+ and JS1 together with the insertion of a CMV-GFP cassette.
  • JS1 was then also further engineered by the insertion of human GM-CSF (hGM-CSF) or mouse GM-CSF (mGM-CSF) so as to replace the ICP34.5 gene and by the deletion of ICP47.
  • hGM-CSF human GM-CSF
  • mGM-CSF mouse GM-CSF
  • Lytic (cell killing) capabilities were enhanced with the JS1-derived non-laboratory strains derived virus in all tumour cell lines tested as compared with the 17+ derived strains. More particularly, the JS1/34.5-virus, i.e. JS1 with ICP34.5 removed by deletion, showed enhanced lytic capabilities in HT29 colorectal adenocarcinoma, LNCaP.FGC prostate adenocarcinoma, MDA-MB-231 breast adenocarcinoma, SK-MEL-28 malignant melanoma and U-87 MG glioblastoma astrocytoma cells.
  • viruses are then used to deliver genes with anti-tumour activity.
  • genes include those encoding pro-drug activators or immunostimulatory proteins.
  • HSV1 strain JS1 has been deposited at the European Collection of Cell Cultures (ECACC), CAMR, Sailsbury, Wiltshire SP4 0JG, United Kingdom, on 2 Jan. 2001 under provisional accession number 01010209 .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Biotechnology (AREA)
  • Virology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Communicable Diseases (AREA)
  • Psychiatry (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention provides a herpes virus with improved oncolytic properties which comprises a gene encoding an immunomodulatory cytokine and which lacks a functional ICP34.5 gene and a functional ICP47 encoding gene.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This is a continuation application of U.S. patent application Ser. No. 13/600,711, filed Aug. 31, 2012, which is a continuation application of U.S. patent application Ser. No. 11/738,807, filed Apr. 23, 2007, now U.S. Pat. No. 8,277,818, which is a continuation application of U.S. patent application Ser. No. 10/181,697, filed Oct. 2, 2002, now U.S. Pat. No. 7,223,593, which is the U.S. National Phase of International Application PCT/GB01/00225, filed Jan. 22, 2001, which claims priority to United Kingdom Patent Application Numbers: 0001475.3, filed Jan. 21, 2000; 0002854.8, filed Feb. 8, 2000; 0100288.0, filed Jan. 5, 2001; and 0100430.8, filed Jan. 6, 2001, which are incorporated by reference herein in their entireties.
  • FIELD OF THE INVENTION
  • The present invention relates to herpes virus strains with improved anti-tumour activity as compared to previously known strains.
  • BACKGROUND TO THE INVENTION
  • Viruses have been shown to have utility in a variety of applications in biotechnology and medicine on many occasions. Each is due to the unique ability of viruses to enter cells at high efficiency. This is followed in such applications by either virus gene expression and replication and/or expression of an inserted heterologous gene. Thus viruses can either deliver and express genes in cells (either viral or other genes) which may be useful in for example gene therapy or the development of vaccines, or they may be useful in selectively killing cells by lytic replication or the action of a delivered gene in for example cancer.
  • Herpes simplex virus (HSV) has been suggested to be of use for the oncolytic treatment of cancer. Here the virus must however be disabled such that it is no longer pathogenic, i.e. does not replicate in and kill non-tumor cells, but such that it can still enter and kill tumor cells. For the oncolytic treatment of cancer, which may also include the delivery of gene(s) enhancing the therapeutic effect, a number of mutations to HSV have been identified which still allow the virus to replicate in culture or in actively dividing cells in vivo (e.g. in tumors), but which prevent significant replication in normal tissue. Such mutations include disruption of the genes encoding ICP34.5, ICP6, and thymidine kinase. Of these, viruses with mutations to ICP34.5, or ICP34.5 together with mutation of e.g. ICP6 have so far shown the most favourable safety profile. Viruses deleted for only ICP34.5 have been shown to replicate in many tumor cell types in vitro and to selectively replicate in artificially induced brain tumors in mice while sparing surrounding tissue. Early stage clinical trials have also shown their safety in man.
  • However, while promise has been shown for various viruses including HSV for the oncolytic treatment of cancer, the majority of this work has used virus strains which do not carry a heterologous gene which may enhance the anti-tumor effect. We propose that the combined use of HSV with an inactivating mutation in the gene encoding ICP34.5 together with the delivery of the gene encoding an immunomodulatory protein such as granulocyte macrophage colony stimulating factor (GM-CSF) encoded in the disabled virus genome may have optimal immune stimulating properties against the tumor to be treated, particularly if functions in the virus which usually reduce immune responses to HSV infected cells have also been inactivated. For example the HSV ICP47 protein specifically inhibits antigen presentation in HSV infected cells (Hill et al 1995), and the product of the UL43 gene and the vhs protein reduce the immune-stimulating abilities of dendritic cells infected with HSV. ICP47 and/or dendritic cell-inactivating genes might therefore usefully be deleted from an oncolytic HSV mutant virus used for the treatment of cancer, particularly if immune effects are to be enhanced through the use of GM-CSF or other immunostimulatory cytokine or chemokine GM-CSF has recently been shown to give an enhanced anti-tumor immune effect if expressed from within a tumor cell rather than administered systemically (Shi et al 1999). Thus in such use an oncolytic HSV mutant would be inoculated into a primary or secondary tumor where replication and oncolytic destruction of the tumor would occur. Immune responses would also be stimulated against the HSV infected cells, and also to tumor cells elsewhere which had spread from the primary tumor site.
  • SUMMARY OF THE INVENTION
  • The present invention provides viruses with improved capabilities for the lytic destruction of tumor cells in vivo. Here herpes simplex virus strains are constructed using a strain of HSV1 or HSV2 in which the genes encoding ICP34.5 and ICP47 have been inactivated such that a functional ICP34.5 or ICP47 protein cannot be expressed and which also carries a gene encoding an immunomodulatory protein. The virus may also be mutated for any additional gene(s) which may be involved in inhibiting the function of dendritic cells including the UL43 gene and/or the gene encoding vhs. The present invention therefore provides viruses capable of the oncolytic destruction of tumor cells and in which anti-tumor immune effects will have been maximised.
  • Accordingly the invention provides:
      • a herpes virus which comprises a gene encoding an immunomodulatory protein, which lacks a functional ICP34.5 encoding gene and a functional ICP47 encoding gene and which is a replication competent in tumour cells;
      • a herpes virus of the invention for use in a method of treatment of the human or animal body by therapy;
      • use of a virus of the invention in the manufacture of a medicament for the treatment of cancer;
      • a pharmaceutical composition comprising as active ingredient a virus according to the invention and a pharmaceutically acceptable carrier or diluent; and
      • HSVI strain JSI as deposited at the European Collection of Cell Cultures (ECACC) under provisional accession number 01010209, or an HSV1 strain derived therefrom.
    BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1. Viruses
  • From top to bottom, diagrams show: laboratory HSV1 strain 17+, clinical HSV1 strain JS1, strain 17+/ICP34.5-, strain JS1/ICP34.5-, strain JS1/ICP34.5-/IC47-/hGMCSF, strain JS1/ICP34.5-/ICP47-/mGMCSF.
  • DETAILED DESCRIPTION OF THE INVENTION A. Viruses
  • A herpes virus of the invention is capable of efficiently infecting target tumor cells and the genes encoding ICP34.5 and ICP47 are inactivated in the virus. Mutation of ICP34.5 allows selective oncolytic activity. Such mutations are described in Chou et al 1990 and Maclean et al 1991, although any mutation in which ICP34.5 is non-functional may be used. The genes encoding ICP6 and/or thymidine kinase may additionally be inactivated, as may other genes if such inactivation does significantly reduce the oncolytic effect, or if such deletion enhances oncolytic or other desirable properties of the virus. ICP47 usually functions to block antigen presentation in HSV-infected cells so its disruption leads to a virus that does not confer on infected tumour cells properties that might protect them from the host's immune system when infected with HSV. Viruses of the invention additionally encode an immunomodulatory protein, preferably GM-CSF, but may also encode other cytokines, chemokines such as RANTES, or other immune-modulatory proteins such as B7.1, B7.2 or CD40L. Genes encoding immunomodulatory proteins may be included individually or in combination.
  • Viral regions altered for the purposes described above may be either eliminated (completely or partly), or made non-functional, or substituted by other sequences, in particular by a gene for an immunomodulatory protein such as GM-CSF.
  • The virus of the invention may be derived from a HSV1 or HSV2 strain, or from a derivative thereof, preferably HSV1. Derivatives include inter-type recombinants containing DNA from HSV1 and HSV2 strains. Such inter-type recombinants are described in the art, for example in Thompson et al, 1988 and Meignier et al, 1988. Derivatives preferably have at least 70% sequence homology to either the HSV1 or HSV2 genomes, more preferably at least 80%, even more preferably at least 90 or 95%. More preferably, a derivative has at least 70% sequence identity to either the HSV1 or HSV2 genome, more preferably at least 80% identity, even more preferably at least 90%, 95% or 98% identity.
  • For example the UWGCG Package provides the BESTFIT program which can be used to calculate homology (for example used on its default settings) (Devereux et al. (1984) Nucleic Acids Research 12, p387-395). The PILEUP and BLAST algorithms can be used to calculate homology or line up sequences (typically on their default settings), for example as described in Altschul (1993) J. Mol. Evol. 36:290-300; Altschul et al. (1990) J. Mol. Biol. 215:403-10.
  • Software for performing BLAST analyses is publicly available through the National Centre for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). This algorithm involves first identifying high scoring sequence pair (HSPs) by identifying short words of length W in the query sequence that either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighbourhood word score threshold (Altschul et al., 1990). These initial neighbourhood word hits act as seeds for initiating searches to find HSPs containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Extensions for the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment. The BLAST program uses as defaults a word length (W) of 11, the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1992) Proc. Natl. Acad. Sci. USA 89: 10915-10919) alignments (B) of 50, expectation (E) of 10, M=5, N=4, and a comparison of both strands.
  • The BLAST algorithm performs a statistical analysis of the similarity between two sequences; see e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-5787. One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a sequence is considered similar to another sequence if the smallest sum probability in comparison of the first sequence to the second sequence is less than about 1, preferably less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
  • A derivative may have the sequence of a HSV1 or HSV2 genome modified by nucleotide substitutions, for example from 1, 2 or 3 to 10, 25, 50 or 100 substitutions. The HSV1 or HSV2 genome may alternatively or additionally be modified by one or more insertions and/or deletions and/or by an extension at either or both ends.
  • Virus strains of the invention may be “non-laboratory” strains. These can also be referred to as “clinical” strains. A person of skill in the art will readily be able to distinguish between a laboratory strain and a non-laboratory, or clinical, strain. Further guidance on the properties likely to be exhibited by virus strains is given below.
  • The key distinction between a laboratory and non-laboratory strain is that laboratory strains currently in common use have been maintained for long periods, many years in some cases, in culture. The culture of viruses such as HSV involves a technique known as serial passage. To grow and maintain viruses, suitable cells are infected with the virus, the virus replicates within the cell and the virus is then harvested; fresh cells are then re-infected, this process constitutes one cycle of serial passage. Each such cycle may take, for example, a few days in the case of HSV. As discussed above, such serial passaging may lead to changes in the properties of the virus strain, in that selection takes places for properties that favour growth in culture (e.g. rapid replication), as opposed to properties useful for practical applications, e.g. maintenance of the capacity to travel along axons in the case of HSV or to infect human cells.
  • Virus strains of the invention are may be non-laboratory strains in that they are derived from strains recently isolated from infected individuals. Strains of the invention are modified compared to the original clinical isolates, and may have spent a time in culture, but any time spent in culture will be comparatively short. Strains of the invention are prepared in such a manner as to retain substantially the desirable properties of the original clinical isolates from which they are derived.
  • A virus strain of the invention is derived from a parental virus strain if the parental virus strain is mutated to produce the virus. For example, a virus of the invention may be derived from the clinical isolate JSI. The parental strain of such a JSI-derived virus may be JSI or another HSV1 strain derived from JSI. Thus a virus of the invention may be a JSI virus comprising a gene encoding an immunomodulatory protein and which lacks a functional ICP34.5 encoding gene and a functional ICP47 encoding gene. In addition, such a virus may contain any other mutation as mentioned herein.
  • A virus of the invention is capable of efficiently infecting target human cancer cells. When such a virus is a non-laboratory or clinical strain it will have been recently isolated from an HSV infected individual and then screened for the desired ability of enhanced replication, infection or killing of tumour and/or other cells in vitro and/or in vivo in comparison to standard laboratory strains. Such viruses of the invention with improved properties as compared to laboratory virus strains are then engineered such that they lack functional ICP34.5 and ICP47 genes and encode a gene(s) for an immunomodulatory protein(s) such as GM-CSF under the control of a suitable promoter(s). Other genes encoding proteins which interfere with the function of dendritic cells such as UL43 and/or vhs may also be inactivated.
  • Preferably, a non-laboratory virus strain of the invention has undergone three years or less in culture since isolation of its unmodified clinical precursor strain from its host. More preferably, the strain has undergone one year or less in culture, for example nine months or less, six months or less, three months or less, or two months or less, one month or less, two weeks or less, or one week or less. By these definitions of time in culture, is meant time actually spent in culture. Thus, for example, it is a common practice to freeze virus strains in order to preserve them. Evidently, preserving by freezing or in an equivalent manner does not qualify as maintaining the strain in culture. Thus, time spent frozen or otherwise preserved is not included in the above definitions of time spent in culture. Time spent in culture is typically time actually spent undergoing serial passage, i.e. time during which selection for undesirable characteristics can occur.
  • Preferably, a non-laboratory virus strain has undergone 1,000 or less cycles or serial passage since isolation of its unmodified clinical precursor strain from its host. More preferably, it has undergone 500 or less, 100 or less, 90 or less, 80 or less, 70 or less, 60 or less, 50 or less, 40 or less, 30 or less, 20 or less, or 10 or less such cycles.
  • Preferably, a non-laboratory virus has a greater ability, as measured by standard statistical tests, than a reference laboratory strain with the equivalent modifications to perform certain functions useful in the application at hand. In the case of an oncolytic virus for tumour treatment, a non-laboratory virus strain of the invention will preferably have a greater ability than a reference laboratory strain with equivalent modifications to infect or replicate in tumour cells, to kill tumour cells or to spread between cells in tissue. More preferably, such greater ability is a statistically significantly greater ability. For example, according to the invention, a non-laboratory strain of the invention may have up to 1.1 fold, 1.2 fold, 1.5 fold, 2 fold, 5 fold, 10 fold, 20 fold, 50 fold, or 100 fold the capacity of the reference strain in respect of the property being tested.
  • Statistical analysis of the properties described herein may be carried out by standard tests, for example, t-tests, ANOVA, or Chi squared tests. Typically, statistical significance will be measured to a level of p=0.05 (5%), more preferably p=0.01p, p=0.001, p=0.0001, p=0.000001.
  • Viruses of the invention infect and replicate in tumour cells, subsequently killing the tumour cells. Thus, such viruses are replication competent. Preferably, they are selectively replication competent in tumour cells. This means that either they replicate in tumour cells and not in non-tumour cells, or that they replicate more effectively in tumour cells than in non-tumour cells. Cells in which the virus is able to replicate are permissive cells. Measurement of selective replication competence can be carried out by the tests described herein for measurement of replication and tumour cell-killing capacity, and also analysed by the statistical techniques mentioned herein if desired.
  • A virus of the invention preferably has a greater ability than an unmodified parent strain to infect or replicate in a tumour cell, to kill tumour cells or to spread between cells in tissues. Preferably this ability is a statistically significant greater ability. For example, a virus according to the invention may have up to 1.1 fold, 1.2 fold, 1.5 fold, 2 fold, 5 fold, 10 fold, 20 fold, 50 fold or 100 fold the capacity of the unmodified parent strain in respect of the property being tested.
  • The properties of the virus strain in respect of tumour cells can be measured in any manner known in the art. For example, the capacity of a virus to infect a tumour cell can be quantified by measuring the dose of virus required to measure a given percentage of cells, for example 50% or 80% of cells. The capacity to replicate in a tumour cell can be measured by growth measurements such as those carried out in the Examples, e.g. by measuring virus growth in cells over a period of 6, 12, 24, 36, 48 or 72 hours or longer.
  • The ability of a virus to kill tumour cells can be roughly quantitated by eye or more exactly quantitated by counting the number of live cells that remain over time for a given time point and MOI for given cell type. For example, comparisons may be made over 24, 48 or 72 hours and using any known tumour cell type. In particular, HT29 colorectal adenocarcinoma, LNCaP.FGC prostate adenocarcinoma, MDA-MB-231 breast adenocarcinoma, SK-MEL-28 malignant melanoma or U-87 MG glioblastoma astrocytoma cells can be used. Any one of these cell types or any combination of these cell types can be used, as may other tumour cell types. It may be desirable to construct a standard panel of tumour cell types for this purpose. To count the number of live cells remaining at a given time point, the number of trypan blue-excluding cells (i.e. live cells) can be counted. Quantitation may also be carried out by fluorescence activated cell sorting (FACS) or MTT assay. Tumour cell-killing ability may also be measured in vivo, e.g. by measuring the reduction in tumour volume engendered by a particular virus.
  • In order to determine the properties of viruses of the invention, it will generally be desirable to use a standard laboratory reference strain for comparison. Any suitable standard laboratory reference strain may be used. In the case of HSV, it is preferred to use one or more of HSV1 strain 17+, HSV1 strain F or HSV1 strain KOS. The reference strain will typically have equivalent modifications to the strain of the invention being tested. Thus, the reference strain will typically have equivalent modifications gene deletions and, such as heterologous gene insertions. In the case of a virus of the invention, where the ICP34.5 and ICP47-encoding genes have been rendered non-functional, then they will also have been rendered non-functional in the reference strain. The modifications made to the reference strain may be identical to those made to the strain of the invention. By this, it is meant that the gene disruptions in the reference strain will be in exactly equivalent positions to those in the strain of the invention, e.g. deletions will be of the same size and in the same place. Similarly, in these embodiments, heterologous genes will be inserted in the same place, driven by the same promoter, etc. However, it is not essential that identical modifications be made. What is important is that the reference gene has functionally equivalent modifications, e.g. that the same genes are rendered non-functional and/or the same heterologous gene or genes is inserted.
  • B. Methods of Mutation
  • The various genes referred to may be rendered functionally inactive by several techniques well known in the art. For example, they may be rendered functionally inactive by deletion(s), substitution(s) or insertion(s), preferably by deletion. Deletions may remove one or more portions of the gene or the entire gene. For example, deletion of only one nucleotide may be made, resulting in a frame shift. However, preferably a larger deletion( ) is made, for example at least 25%, more preferably at least 50% of the total coding and non-coding sequence (or alternatively, in absolute terms, at least 10 nucleotides, more preferably at least 100 nucleotides, most preferably, at least 1000 nucleotides). It is particularly preferred to remove the entire gene and some of the flanking sequences. Where two or more copies of the gene are present in the viral genome it is preferred that both copies of the gene are rendered functionally inactive.
  • Mutations are made in the herpes viruses by homologous recombination methods well known to those skilled in the art. For example, HSV genomic DNA is transfected together with a vector, preferably a plasmid vector, comprising the mutated sequence flanked by homologous HSV sequences. The mutated sequence may comprise a deletion(s), insertion(s) or substitution(s), all of which may be constructed by routine techniques. Insertions may include selectable marker genes, for example lacZ or green fluorescent protein (GFP), for screening recombinant viruses, for example, Thgalactosidase activity or fluorescence.
  • C. Heterologous Genes and Promoters
  • The viruses of the invention may be modified to carry a heterologous gene encoding an immunomodulatory protein. Preferably the immunomodulatory protein will enhance the anti-tumour activity of the virus. More preferably the protein is GM-CSF or another cytokine, a chemokine such as RANTES, or another immunomodulatory molecule such as B7.1, B7.2 or CD40L. Most preferably the immunomodulatory molecule is GM-CSF. The immunomodulatory gene may be any allelic variant of a wild-type gene, or it may be a mutant gene. The immunomodulatory gene will be derived from a mammal, preferably a rodent or primate, more preferably a human. The immunomodulatory gene is preferably operably linked to a control sequence permitting expression of said gene in a cell in vivo. Viruses of the invention may thus be used to deliver the immunomodulatory gene (or genes) to a cell in vivo where it will be expressed.
  • The immunomodulatory gene may be inserted into the viral genome by any suitable technique such as homologous recombination of HSV strains with, for example, plasmid vectors carrying the gene flanked by HSV sequences. The GM-CSF gene, or other immunomodulatory gene, may be introduced into a suitable plasmid vector comprising herpes viral sequences using cloning techniques well-known in the art. The gene may be inserted into the viral genome at any location provided that oncolytic properties are still retained. Immunomodulatory genes may be inserted at multiple sites within the virus genome. For example, from 2 to 5 genes may be inserted into the genome.
  • The transcribed sequence of the immunomodulatory gene is preferably operably linked to a control sequence permitting expression of the gene in a tumour cell. The term “operably linked” refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner. A control sequence “operably linked” to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequence.
  • The control sequence comprises a promoter allowing expression of the immunomodulatory gene and a signal for termination of transcription. The promoter is selected from promoters which are functional in mammalian, preferably human tumour cells. The promoter may be derived from promoter sequences of eukaryotic genes. For example, the promoter may be derived from the genome of a cell in which expression of the heterologous gene is to occur, preferably a mammalian, preferably a human tumour cell. With respect to eukaryotic promoters, they may be promoters that function in a ubiquitous manner (such as promoters of ∃-actin, tubulin) or, alternatively, in a tumour-specific manner. They may also be promoters that respond to specific stimuli, for example promoters that bind steroid hormone receptors. Viral promoters may also be used, for example the Moloney murine leukaemia virus long terminal repeat (MMLV LTR) promoter or other retroviral promoters, the human or mouse cytomegalovirus (CMV) IE promoter, or promoters of herpes virus genes including those driving expression of the latency associated transcripts.
  • Expression cassettes and other suitable constructs comprising the immunomodulatory gene and control sequences can be made using routine cloning techniques known to persons skilled in the art (see, for example, Sambrook et al., 1989, Molecular Cloning—A laboratory manual; Cold Spring Harbor Press).
  • It may also be advantageous for the promoters to be inducible so that the levels of expression of the immunomodulatory gene(s) can be regulated during the life-time of the tumour cell. Inducible means that the levels of expression obtained using the promoter can be regulated. For example, a virus of the invention may further comprise a hererologous gene encoding the tet repressor/VP16 transcriptional activator fusion protein under the control of a strong promoter (e.g. the CMV IE promoter) and the immunomodulatory gene may be under the control of a promoter responsive to the tet repressor VP16 transcriptional activator fusion protein previously reported (Gossen and Bujard, 1992, Gossen et al, 1995). Thus, in this example, expression of the immunomodulatory gene would depend on the presence or absence of tetracycline.
  • Multiple heterologous genes can be accommodated in the herpes virus genome. Therefore, a virus of the invention may comprise two or more immunomodulatory genes, for example from 2 to 3, 4 or 5 immunomodulatory genes. More than one gene and associated control sequences could be introduced into a particular HSV strain either at a single site or at multiple sites in the virus genome. Alternatively pairs of promoters (the same or different promoters) facing in opposite orientations away from each other, each driving the expression of an immunomodulatory gene may be used.
  • D. Therapeutic Uses
  • Viruses of the invention may be used in methods of cancer therapy of the human or animal body. In particular, viruses of the invention may be used in the oncolytic treatment of cancer, either with or without additional pro-drug therapy or stimulation of an anti-tumour immune response. Viruses of the invention may be used in the therapeutic treatment of any solid tumour in a mammal, preferably in a human. For example viruses of the invention may be administered to a subject with prostate, breast, lung, liver, endometrial, bladder, colon or cervical carcinoma; adenocarcinoma; melanoma; lymphoma; glioma; or sarcomas such as soft tissue and bone sarcomas.
  • E. Administration
  • The invention provides the use of a herpes virus according to the invention in the manufacture of a medicament for the treatment of cancer. The medicament may be used in a patient, preferably a human patient, in need of treatment. A patient in need of treatment is an individual suffering from cancer, preferably an individual with a solid tumour. The aim of therapeutic treatment using the medicament is to improve the condition of a patient. Typically therapeutic treatment using a virus of the invention allieviates the symptoms of the cancer. Administration of the medicament to an individual suffering from a tumour will typically kill the cells of the tumour thus decreasing the size of the tumour and/or preventing spread of malignant cells from the tumour.
  • The invention provides a pharmaceutical composition comprising the virus and a pharmaceutically acceptable carrier or diluent. Suitable carriers and diluents include isotonic saline solutions, for example phosphate-buffered saline.
  • Therapeutic treatment may be carried out following direct injection of the virus composition into target tissue which may be the tumour or a blood vessel supplying the tumour. The amount of virus administered is in the case of HSV in the range of from 104 to 1010 pfu, preferably from 105 to 108 pfu, more preferably about 106 to 108 pfu. Typically up to 500:1, typically from 1 to 200 μl preferably from 1 to 10 μl of a pharmaceutical composition consisting essentially of the virus and a pharmaceutically acceptable suitable carrier or diluent would be used for injection. However for some oncolytic therapy applications larger volumes up to 10 ml may also be used, depending on the tumour and the inoculation site.
  • The routes of administration and dosages described are intended only as a guide since a skilled practitioner will be able to determine readily the optimum route of administration and dosage. The dosage may be determined according to various parameters, especially according to the location of the tumour, the size of the tumour, the age, weight and condition of the patient to be treated and the route of administration. Preferably the virus is administered by direct injection into the tumour. The virus may also be administered systemically or by injection into a blood vessel supplying the tumour. The optimum route of administration will depend on the location and size of the tumour.
  • The following Examples illustrates the invention.
  • Herpes simplex type-1 virus (HSV1) in which the neurovirulence factor ICP34.5 is inactivated has previously been shown to direct tumour specific cell lysis in tumour models both in vitro and in vivo. Such viruses have also been shown to be safe in Phase I clinical trials by direct intra-cerebral injection in late stage glioma patients.
  • Previous work has used serially passaged laboratory isolates of HSV1 (viruses derived from HSV1 strain 17+ or HSV1 strain F) which might be anticipated to be attenuated in their lytic capability in human tumour cells as compared to more recent clinical isolates.
  • In work aimed at producing ICP34.5 deleted HSV with enhanced oncolytic and anti-tumour potential, we have deleted ICP47 and ICP34.5 from HSV1 strain JS1 and have inserted the immunomodulatory gene for GM-CSF.
  • Virus Construction (See FIG. 1)
  • The viruses used were either based on HSV1 strain 17+ (a standard laboratory strain) or a clinical isolate derived from cold sores from a frequent re-activator of HSV1. This clinical, or “non-laboratory”, strain is named JS1. ICP34.5 was completely deleted from strain 17+ and JS1 together with the insertion of a CMV-GFP cassette. JS1 was then also further engineered by the insertion of human GM-CSF (hGM-CSF) or mouse GM-CSF (mGM-CSF) so as to replace the ICP34.5 gene and by the deletion of ICP47. The derivatives of JS1 discussed herein are also non-laboratory strains, i.e. modified non-laboratory strains of the invention.
  • Lytic Capabilities of Viruses
  • Lytic (cell killing) capabilities were enhanced with the JS1-derived non-laboratory strains derived virus in all tumour cell lines tested as compared with the 17+ derived strains. More particularly, the JS1/34.5-virus, i.e. JS1 with ICP34.5 removed by deletion, showed enhanced lytic capabilities in HT29 colorectal adenocarcinoma, LNCaP.FGC prostate adenocarcinoma, MDA-MB-231 breast adenocarcinoma, SK-MEL-28 malignant melanoma and U-87 MG glioblastoma astrocytoma cells.
  • Thus, to provide increased oncolytic activity, the use of recent clinical virus strains is likely to enhance the anti-tumour capabilities of such viruses when used in human patients for cancer treatment.
  • Further Enhanced Anti-Tumour Activity
  • Further enhanced activity may also be anticipated if these viruses are then used to deliver genes with anti-tumour activity. Such genes include those encoding pro-drug activators or immunostimulatory proteins.
  • An ICP34.5 deleted clinical isolated of HSV1 which expresses human or mouse GM-CSF was produced from JS1. GM-CSF is a potent immune stimulator. These virus are designed to enhance anti-tumour immune responses following intra-tumoral injection. These viruses were demonstrated to express human or mouse GM-CSF using ELISA assay kits (Biotrak, RTM Amersham) when the viruses are produced in BHK cells in culture. Individual wells of a six well plate produced 0.56 or 0.54 microgrammes of human or mouse GM-CSF respectively 24 hrs after infection of confluent BHK cells at MOI=0.5.
  • Deposit Information
  • HSV1 strain JS1 has been deposited at the European Collection of Cell Cultures (ECACC), CAMR, Sailsbury, Wiltshire SP4 0JG, United Kingdom, on 2 Jan. 2001 under provisional accession number 01010209.
  • REFERENCES
    • Hill et al. 1995, Nature 375; 411-415
    • Shi et al. 1999, Cancer-Gene-Ther 6: 81-88
    • Chou et al. 1990, Science 250: 1262-1266
    • Maclean et al. 1991, J. Gen. Virol. 72: 631-639
    • Gossen M & Bujard H, 1992, PNAS 89: 5547-5551
    • Gossen M et al. 1995, Science 268: 1766-1769
    • Thompson et al. 1988, Virus Genes 1(3); 275-286
    • Meignier et al. 1988, Infect. Dis. 159; 602-614

Claims (43)

1. A herpes virus which comprises a gene encoding an immunomodulatory protein, which lacks a functional ICP34.5 encoding gene and a functional ICP47 encoding gene and which is replication competent in tumour cells.
2. A virus according to claim 1 wherein said immunomodulatory gene is a cytokine, a chemokine or a protein capable of regulating T cell proliferation.
3. A virus according to claim 2 wherein said cytokine is GM-CSF, said chemokine is RANTES or said protein is B7.1, B7.2 or CD40L.
4. A virus according to claim 1 which encodes two or more immunomodulatory proteins.
5. A virus according to claim 1 which further lacks a functional gene encoding ICP6, glycoprotein H or thymidine kinase.
6. A virus according to claim 1 which further lacks a gene encoding functional copy of a protein capable of inhibiting dendritic cell function.
7. A virus according to claim 6 in which said protein capable of inhibiting dendritic cell function is UL43 or vhs.
8. A virus according to claim 1 which is a strain of herpes simplex virus 1 or 2.
9. A virus according to claim 1 which is a non-laboratory virus strain.
10. A virus according to claim 9 wherein said non-laboratory strain: (a) has undergone one year or less in culture since isolation of its unmodified precursor strain from its host, or (b) has undergone 100 or less cycles of serial passage since isolation of unmodified precursor strain from its host, or (c) has a greater ability than a reference laboratory strain with equivalent modifications to infect or replicate in a tumour cell, to kill tumour cells, or to spread between cells in tissue, or (d) has substantially the ability of its unmodified precursor strain in respect of one or more of the properties defined in (c).
11. A virus according to claim 10 wherein, in (c), said greater ability in a statistically significant greater ability; or wherein, in (d), substantially the same ability is the same ability or at an ability not statistically different.
12. A virus according to claim 10 wherein the non-laboratory strain is an HSV strain and the reference strain is HSV1 strain 17+, HSV1 strain F as HSV1 strain KOS with equivalent modifications to the non-laboratory strain.
13. A virus according to claim 1 which is derived from HSV1 JSI as deposited at the European collection of cell cultures (ECAAC) under provisional accession number 01010209.
14. A virus according to claim 1 for use in a method of treatment of the human or animal body by therapy.
15. Use of a virus according to claim 1 in the manufacture of a medicament for the treatment of cancer.
16. Use according to claim 15 wherein said medicament is for direct intra-tumoral inoculation.
17. A pharmaceutical composition comprising as active ingredient a virus according to claim 1 and a pharmaceutically acceptable carrier or diluent.
18. A method of treating a tumour in an individual in need thereof by administering to said individual an effective amount of a virus according to claim 1.
19. An agent for treating cancer comprising a herpes virus which comprises a gene encoding an immunomodulatory cytokine, which lacks a functional ICP34.5-encoding gene and a functional ICP47 encoding gene and which is replication competent in tumour cells.
20. HSV1 strain JSI as deposited as the European Collection of Cell. Cultures (ECACC) under provisional accession number 01010209, or an HSV1 strain derived therefrom.
21. A method of treating cancer in an individual in need thereof by administering to a tumor in said individual a therapeutically effective amount of a herpes simplex virus which: (i) comprises an immunostimulatory protein; (ii) lacks a functional ICP34.5 encoding gene and a functional ICP47 encoding gene; and (iii) is replication competent in infected tumor cells.
22. The method of claim 21 wherein said virus kills infected tumor cells by oncolysis and stimulates an anti-tumor immune response.
23. The method of claim 21 wherein tumor cells infected with said virus and non-infected tumor cells are killed.
24. The method of claim 23 wherein said virus is administered to a primary tumor and tumor cells in the primary tumor and tumor cells elsewhere that have spread from the primary tumor site are killed.
25. The method of claim 23 wherein said cancer is selected from the group consisting of prostate, breast, liver, endometrial, bladder, colon or cervical carcinoma, melanoma, lymphoma, glioma and sarcoma.
26. The method of claim 23 wherein said cancer is selected from the group consisting of prostate, breast, lung, liver, endometrial, bladder, colon or cervical carcinoma, adenocarcinoma, melanoma, lymphoma, glioma and sarcoma.
27. The method of claim 21 wherein said virus is administered by direct intratumoral inoculation.
28. The method of claim 21 wherein said virus is administered to a blood vessel supplying the tumor.
29. The method of claim 21 wherein said immunostimulatory protein is GM-CSF.
30. The method of claim 21 wherein said immunostimulatory protein is a cytokine, chemokine or protein capable of regulating T cell proliferation.
31. The method of claim 21 wherein said virus further lacks a functional gene encoding ICP6, glycoprotein H or thymidine kinase.
32. The method of claim 21 wherein said virus is a strain of herpes simplex virus 1 or 2.
33. The method of claim 21 wherein said virus is a clinical isolate.
34. The method of claim 33 wherein said clinical isolate is derived from HSV1 JS1 as deposited at the European collection of cell cultures (ECAAC) under accession number 01010209.
35. A herpes simplex virus which: (i) comprises a gene encoding an immunostimulatory protein selected from GM-CSF and a chemokine; (ii) lacks a functional ICP34.5 encoding gene and a functional ICP47 encoding gene; and (iii) is replication competent in infected tumor cells.
36. The virus of claim 35 wherein said immunostimulatory protein is GM-CSF.
37. The virus of claim 35 which comprises a functional UL43 gene and/or a functional gene encoding vhs.
38. The virus of claim 35 which further lacks a functional gene encoding ICP6, glycoprotein H or thymidine kinase.
39. The virus of claim 35 which is a strain of herpes simplex virus 1 or 2.
40. The virus of claim 35 which is a clinical isolate.
41. A pharmaceutical composition comprising as active ingredient a virus according to claim 35 and a pharmaceutically acceptable carrier or diluent.
42. A pharmaceutical composition comprising as active ingredient a virus according to claim 36 and a pharmaceutically acceptable carrier or diluent.
43. A pharmaceutical composition according to claim 42, wherein the virus is derived from HSV1 JS1 as deposited at the European collection of cell cultures (ECAAC) under accession number 01010209.
US14/174,521 2000-01-21 2014-02-06 Herpes virus strains Abandoned US20140154215A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/174,521 US20140154215A1 (en) 2000-01-21 2014-02-06 Herpes virus strains
US14/700,036 US20150232812A1 (en) 2000-01-21 2015-04-29 Herpes virus strains
US16/376,089 US20200032219A1 (en) 2000-01-21 2019-04-05 Herpes virus strains

Applications Claiming Priority (13)

Application Number Priority Date Filing Date Title
GB0001475A GB0001475D0 (en) 2000-01-21 2000-01-21 Virus strains
GB0001475.3 2000-01-21
GB0002854A GB0002854D0 (en) 2000-02-08 2000-02-08 Virus strains
GB0002854.8 2000-02-08
GB0100288.0 2001-01-05
GB0100288A GB0100288D0 (en) 2001-01-05 2001-01-05 Virus strains
GB0100430.8 2001-01-06
GB0100430A GB0100430D0 (en) 2001-01-06 2001-01-06 Virus strains
PCT/GB2001/000225 WO2001053505A2 (en) 2000-01-21 2001-01-22 Herpes virus strains for gene therapy
US10/181,697 US7223593B2 (en) 2000-01-21 2001-01-22 Herpes virus strains for gene therapy
US11/738,807 US8277818B2 (en) 2000-01-21 2007-04-23 Herpes virus strains for gene therapy
US13/600,711 US8680068B2 (en) 2000-01-21 2012-08-31 Herpes virus strains
US14/174,521 US20140154215A1 (en) 2000-01-21 2014-02-06 Herpes virus strains

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/600,711 Continuation US8680068B2 (en) 2000-01-21 2012-08-31 Herpes virus strains

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/700,036 Continuation US20150232812A1 (en) 2000-01-21 2015-04-29 Herpes virus strains

Publications (1)

Publication Number Publication Date
US20140154215A1 true US20140154215A1 (en) 2014-06-05

Family

ID=27447756

Family Applications (12)

Application Number Title Priority Date Filing Date
US10/181,697 Active 2026-10-28 US7223593B2 (en) 2000-01-21 2001-01-22 Herpes virus strains for gene therapy
US10/181,692 Expired - Lifetime US7063835B2 (en) 2000-01-21 2001-01-22 Virus strains
US11/372,613 Expired - Lifetime US7537924B2 (en) 2000-01-21 2006-03-10 Virus strains
US11/738,807 Expired - Lifetime US8277818B2 (en) 2000-01-21 2007-04-23 Herpes virus strains for gene therapy
US12/425,952 Abandoned US20090220460A1 (en) 2000-01-21 2009-04-17 Virus strains
US13/399,868 Abandoned US20120164108A1 (en) 2000-01-21 2012-02-17 Virus strains
US13/600,711 Expired - Fee Related US8680068B2 (en) 2000-01-21 2012-08-31 Herpes virus strains
US14/174,521 Abandoned US20140154215A1 (en) 2000-01-21 2014-02-06 Herpes virus strains
US14/175,855 Expired - Fee Related US10301600B2 (en) 2000-01-21 2014-02-07 Virus strains
US14/700,036 Abandoned US20150232812A1 (en) 2000-01-21 2015-04-29 Herpes virus strains
US16/372,800 Abandoned US20200032218A1 (en) 2000-01-21 2019-04-02 Virus strains
US16/376,089 Abandoned US20200032219A1 (en) 2000-01-21 2019-04-05 Herpes virus strains

Family Applications Before (7)

Application Number Title Priority Date Filing Date
US10/181,697 Active 2026-10-28 US7223593B2 (en) 2000-01-21 2001-01-22 Herpes virus strains for gene therapy
US10/181,692 Expired - Lifetime US7063835B2 (en) 2000-01-21 2001-01-22 Virus strains
US11/372,613 Expired - Lifetime US7537924B2 (en) 2000-01-21 2006-03-10 Virus strains
US11/738,807 Expired - Lifetime US8277818B2 (en) 2000-01-21 2007-04-23 Herpes virus strains for gene therapy
US12/425,952 Abandoned US20090220460A1 (en) 2000-01-21 2009-04-17 Virus strains
US13/399,868 Abandoned US20120164108A1 (en) 2000-01-21 2012-02-17 Virus strains
US13/600,711 Expired - Fee Related US8680068B2 (en) 2000-01-21 2012-08-31 Herpes virus strains

Family Applications After (4)

Application Number Title Priority Date Filing Date
US14/175,855 Expired - Fee Related US10301600B2 (en) 2000-01-21 2014-02-07 Virus strains
US14/700,036 Abandoned US20150232812A1 (en) 2000-01-21 2015-04-29 Herpes virus strains
US16/372,800 Abandoned US20200032218A1 (en) 2000-01-21 2019-04-02 Virus strains
US16/376,089 Abandoned US20200032219A1 (en) 2000-01-21 2019-04-05 Herpes virus strains

Country Status (22)

Country Link
US (12) US7223593B2 (en)
EP (4) EP1568779A1 (en)
JP (6) JP4810042B2 (en)
KR (2) KR100768408B1 (en)
CN (2) CN1418255A (en)
AT (2) ATE312189T1 (en)
AU (2) AU2695101A (en)
BE (1) BE2016C033I2 (en)
BR (3) BRPI0107736B8 (en)
CA (2) CA2398335C (en)
CY (2) CY2016021I1 (en)
DE (2) DE60107203T3 (en)
DK (2) DK1252323T3 (en)
ES (2) ES2254359T3 (en)
FR (1) FR16C0026I2 (en)
GB (2) GB2375113B (en)
HK (2) HK1047451B (en)
IL (4) IL150678A0 (en)
LU (2) LU93101I2 (en)
NL (2) NL300821I2 (en)
PT (1) PT1252322E (en)
WO (2) WO2001053505A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150202290A1 (en) * 2012-08-30 2015-07-23 Amgen Inc. Method for treating melanoma using a herpes simplex virus and an immune checkpoint inhibitor
WO2020109389A1 (en) 2018-11-28 2020-06-04 Innovative Molecules Gmbh Helicase primase inhibitors for treating cancer in a combination therapy with oncolytic viruses

Families Citing this family (174)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AUPQ425699A0 (en) 1999-11-25 1999-12-23 University Of Newcastle Research Associates Limited, The A method of treating a malignancy in a subject and a pharmaceutical composition for use in same
DE60107203T3 (en) * 2000-01-21 2009-07-23 Biovex Ltd. HERPES VIRUSES FOR GENE THERAPY
WO2002076216A1 (en) 2001-03-27 2002-10-03 Medigene, Inc. Viral vectors and their use in therapeutic methods
GB0203285D0 (en) 2002-02-12 2002-03-27 Brown Susanne M An herpes simplex virus complex
JP2004099584A (en) * 2002-05-02 2004-04-02 Keio Gijuku Antitumor agent using hsv
AU2002953436A0 (en) 2002-12-18 2003-01-09 The University Of Newcastle Research Associates Limited A method of treating a malignancy in a subject via direct picornaviral-mediated oncolysis
GB0317511D0 (en) * 2003-07-25 2003-08-27 Biovex Ltd Viral vectors
US7897146B2 (en) 2003-11-17 2011-03-01 Crusade Laboratories Limited Treatment using herpes simplex virus
GB0326798D0 (en) * 2003-11-17 2003-12-24 Crusade Lab Ltd Methods for generating mutant virus
EP1694852B1 (en) * 2003-11-17 2010-10-13 Crusade Laboratories Limited Mutant herpes simplex virus and use thereof in the treatment of squamous cell cancer
WO2005094867A1 (en) * 2004-03-31 2005-10-13 Tomoki Todo Anticancer activity enhancer of viral therapy and method of cancer prevention or treatment
CA2561714A1 (en) * 2004-04-08 2005-10-27 Sangamo Biosciences, Inc. Methods and compositions for treating neuropathic and neurodegenerative conditions
US9273326B2 (en) * 2004-04-30 2016-03-01 The Brigham And Women's Hospital, Inc. Tetracycline-regulated gene expression in HSV-1 vectors
KR20130114758A (en) * 2005-05-27 2013-10-17 오스페달레 산 라파엘 에스.알.엘. Gene vector comprising mi-rna
US20080008686A1 (en) * 2006-07-10 2008-01-10 The Brigham And Women's Hospital, Inc. Tetracycline repressor regulated oncolytic viruses
US8450106B2 (en) * 2007-10-17 2013-05-28 The Ohio State University Research Foundation Oncolytic virus
US8313896B2 (en) * 2008-04-04 2012-11-20 The General Hospital Corporation Oncolytic herpes simplex virus immunotherapy in the treatment of brain cancer
CA2689707A1 (en) 2009-11-16 2011-05-16 Jean-Simon Diallo Identification of the novel small molecule viral sensitizer vse1 using high-throughput screening
AU2010333749B2 (en) 2009-12-21 2015-07-30 The Brigham And Women's Hospital, Inc. Herpes simplex virus vaccines
CN102146418B (en) * 2010-02-09 2014-01-15 武汉滨会生物科技有限公司 Recombinant II type herpes simplex virus vector, preparation method of recombinant II type herpes simplex virus vector, recombinant virus, medicinal composition and application
WO2011101912A1 (en) * 2010-02-19 2011-08-25 国立大学法人東京大学 Recombinant herpes virus and pharmaceutical composition containing recombinant herpes virus
WO2011119628A2 (en) 2010-03-23 2011-09-29 The Regents Of The University Of California Compositions and methods for self-adjuvanting vaccines against microbes and tumors
US20130202639A1 (en) * 2010-03-25 2013-08-08 Konstantin G. Kousoulas Synthetic Herpes Simplex Viruses for Treatment of Cancers
EP2670426B1 (en) 2011-01-31 2017-05-10 The General Hospital Corporation Multimodal trail molecules and uses in cellular therapies
ES2704632T3 (en) 2011-09-08 2019-03-19 Univ New York Oncolytic herpes simplex virus and its therapeutic uses
EP2764119A2 (en) 2011-10-05 2014-08-13 Genelux Corporation Method for detecting replication or colonization of a biological therapeutic
WO2013138522A2 (en) 2012-03-16 2013-09-19 Genelux Corporation Methods for assessing effectiveness and monitoring oncolytic virus treatment
WO2013158265A1 (en) 2012-04-20 2013-10-24 Genelux Corporation Imaging methods for oncolytic virus therapy
US9862932B2 (en) 2012-07-24 2018-01-09 The General Hospital Corporation Oncolytic virus therapy for resistant tumors
US9605074B2 (en) 2012-08-30 2017-03-28 The General Hospital Corporation Multifunctional nanobodies for treating cancer
WO2014055960A1 (en) 2012-10-05 2014-04-10 Genelux Corporation Energy absorbing-based diagnostic and therapeutic methods employing nucleic acid molecules encoding chromophore-producing enzymes
CN105637092B (en) * 2013-07-17 2021-06-11 匹兹堡大学-属高等教育联邦体系 Non-toxic HSV vectors for efficient gene delivery applications and complementing cells for their production
WO2015061386A1 (en) 2013-10-24 2015-04-30 Amgen Inc. Injector and method of assembly
EP3060281B1 (en) 2013-10-24 2019-01-30 Amgen Inc. Drug delivery system with temperature-sensitive control
RS57043B1 (en) 2013-10-25 2018-05-31 Psioxus Therapeutics Ltd Oncolytic adenoviruses armed with heterologous genes
WO2015089280A1 (en) 2013-12-11 2015-06-18 The General Hospital Corporation Stem cell delivered oncolytic herpes simplex virus and methods for treating brain tumors
WO2015103438A2 (en) 2014-01-02 2015-07-09 Genelux Corporation Oncolytic virus adjunct therapy with agents that increase virus infectivity
US10994112B2 (en) 2014-02-05 2021-05-04 Amgen Inc. Drug delivery system with electromagnetic field generator
MA39818A (en) 2014-03-30 2017-02-08 Benevir Biopharm Inc Exogenous tap inhibitor "armed" oncolytic viruses and therapeutic uses thereof
EP3785749A1 (en) 2014-05-07 2021-03-03 Amgen Inc. Autoinjector with shock reducing elements
WO2015187797A1 (en) 2014-06-03 2015-12-10 Amgen Inc. Controllable drug delivery system and method of use
JP6625627B2 (en) 2014-10-14 2019-12-25 ハロザイム インコーポレイテッド Compositions of adenosine deaminase-2 (ADA2), variants thereof and methods of using the same
US10695506B2 (en) 2014-10-14 2020-06-30 Amgen Inc. Drug injection device with visual and audio indicators
EP3689394A1 (en) 2014-12-19 2020-08-05 Amgen Inc. Drug delivery device with live button or user interface field
ES2898469T3 (en) 2014-12-19 2022-03-07 Amgen Inc Medication administration device with proximity sensor
ES2952055T3 (en) 2015-01-26 2023-10-26 Ottawa Hospital Res Inst Compositions and methods for viral sensitization
WO2016133947A1 (en) 2015-02-17 2016-08-25 Amgen Inc. Drug delivery device with vacuum assisted securement and/or feedback
WO2016138434A1 (en) 2015-02-27 2016-09-01 Amgen Inc. Drug delivery device having a needle guard mechanism with a tunable threshold of resistance to needle guard movement
GB201505860D0 (en) 2015-04-07 2015-05-20 Agalimmune Ltd Therapeutic compositions and methods of use for treating cancer
US20180112006A1 (en) 2015-04-17 2018-04-26 The General Hospital Corporation Agents, systems and methods for treating cancer
BR112017023171A2 (en) * 2015-04-30 2018-07-17 Psioxus Therapeutics Limited oncolytic adenovirus encoding protein b7
WO2016205429A1 (en) 2015-06-15 2016-12-22 New York University Method of treatment using oncolytic viruses
WO2017039786A1 (en) 2015-09-02 2017-03-09 Amgen Inc. Syringe assembly adapter for a syringe
CN105219738A (en) * 2015-09-21 2016-01-06 北京神源德生物科技有限公司 Recombinant herpes simplex virus and its infect and prepare its host cell and their application
JP2018532810A (en) 2015-11-07 2018-11-08 マルチビア インコーポレイテッド Composition comprising tumor suppressor gene therapy and immune checkpoint therapy for the treatment of cancer
JP7082568B2 (en) 2015-12-09 2022-06-08 アムジエン・インコーポレーテツド Automatic syringe with signal transduction cap
EA201891021A1 (en) 2015-12-17 2018-11-30 Псайоксус Терапьютикс Лимитед ADENOVIRUS GROUP B, ENCODING ANTIBODY TO THE TCR COMPLEX OR FRAGMENT OF THE INDICATED ANTIBODY
US11154661B2 (en) 2016-01-06 2021-10-26 Amgen Inc. Auto-injector with signaling electronics
ES2831080T5 (en) 2016-01-08 2023-10-30 Replimune Ltd Modified oncolytic virus
WO2017143308A1 (en) 2016-02-19 2017-08-24 Virogin Biotech Canada Ltd Compositions and methods of using stat1/3 inhibitors with oncolytic herpes virus
ES2814287T3 (en) 2016-03-15 2021-03-26 Amgen Inc Reduce the likelihood of glass breakage in drug delivery devices
EP4212169A1 (en) 2016-03-25 2023-07-19 PeriphaGen, Inc. High-transducing hsv vectors
EP3443000A2 (en) 2016-04-15 2019-02-20 Alpine Immune Sciences, Inc. Cd80 variant immunomodulatory proteins and uses thereof
EP3442999A2 (en) 2016-04-15 2019-02-20 Alpine Immune Sciences, Inc. Icos ligand variant immunomodulatory proteins and uses thereof
WO2017189754A1 (en) * 2016-04-26 2017-11-02 Salk Institute For Biological Studies Hsv--1 oncolytic virus therapies that specificallyh kill alt dependent cancers
WO2017190112A1 (en) 2016-04-29 2017-11-02 Virogin Biotech Canada Ltd Hsv vectors with enhanced replication in cancer cells
US11541168B2 (en) 2016-04-29 2023-01-03 Amgen Inc. Drug delivery device with messaging label
US11389588B2 (en) 2016-05-02 2022-07-19 Amgen Inc. Syringe adapter and guide for filling an on-body injector
EP3455142B1 (en) 2016-05-13 2023-08-09 Amgen Inc. Vial sleeve assembly
EP3458988B1 (en) 2016-05-16 2023-10-18 Amgen Inc. Data encryption in medical devices with limited computational capability
US11541176B2 (en) 2016-06-03 2023-01-03 Amgen Inc. Impact testing apparatuses and methods for drug delivery devices
EP3478342A1 (en) 2016-07-01 2019-05-08 Amgen Inc. Drug delivery device having minimized risk of component fracture upon impact events
US11471488B2 (en) 2016-07-28 2022-10-18 Alpine Immune Sciences, Inc. CD155 variant immunomodulatory proteins and uses thereof
EP3491013A1 (en) 2016-07-28 2019-06-05 Alpine Immune Sciences, Inc. Cd155 variant immunomodulatory proteins and uses thereof
US11834490B2 (en) 2016-07-28 2023-12-05 Alpine Immune Sciences, Inc. CD112 variant immunomodulatory proteins and uses thereof
JP7200104B2 (en) 2016-08-01 2023-01-06 ヴァイロジン バイオテック カナダ リミテッド Oncolytic Herpes Simplex Virus Vectors Expressing Immune System Stimulatory Molecules
US20190328965A1 (en) 2016-08-17 2019-10-31 Amgen Inc. Drug delivery device with placement detection
GB201713765D0 (en) 2017-08-28 2017-10-11 Psioxus Therapeutics Ltd Modified adenovirus
CN110022889B (en) 2016-10-03 2024-05-10 渥太华医院研究所 Compositions and methods for enhancing the growth, transmission and oncolytic and immunotherapeutic effects of oncolytic RNA viruses
WO2018075447A1 (en) 2016-10-19 2018-04-26 The Trustees Of Columbia University In The City Of New York Combination of braf inhibitor, talimogene laherparepvec, and immune checkpoint inhibitor for use in the treatment cancer (melanoma)
KR20190099194A (en) 2016-10-20 2019-08-26 알파인 이뮨 사이언시즈, 인코포레이티드 Secretory variant immunomodulatory proteins and engineered cell therapy
WO2018081234A1 (en) 2016-10-25 2018-05-03 Amgen Inc. On-body injector
CN110381997A (en) 2016-12-12 2019-10-25 茂体外尔公司 For treating and preventing the method and composition comprising gene-virus therapy and immunologic test point inhibitor of cancer and infectious diseases
WO2018118967A1 (en) 2016-12-21 2018-06-28 Memgen, Llc Armed replication-competent oncolytic adenoviruses
US11298420B2 (en) 2016-12-21 2022-04-12 Memgen, Llc Armed oncolytic viruses
AU2018210301A1 (en) 2017-01-17 2019-08-01 Amgen Inc. Injection devices and related methods of use and assembly
WO2018148462A1 (en) 2017-02-09 2018-08-16 Indapta Therapeutics, Inc. Engineered natural killer (nk) cells and compositions and methods thereof
MX2019009625A (en) 2017-02-17 2019-10-09 Amgen Inc Drug delivery device with sterile fluid flowpath and related method of assembly.
JP7280189B2 (en) 2017-02-17 2023-05-23 アムジエン・インコーポレーテツド Insertion mechanism for drug delivery device
MX2019010544A (en) 2017-03-06 2019-10-21 Amgen Inc Drug delivery device with activation prevention feature.
SG11201908058UA (en) 2017-03-07 2019-09-27 Amgen Inc Needle insertion by overpressure
MX2019010671A (en) 2017-03-09 2019-10-21 Amgen Inc Insertion mechanism for drug delivery device.
AU2018235944B2 (en) 2017-03-15 2024-01-04 Amgen Inc. Use of oncolytic viruses, alone or in combination with a checkpoint inhibitor, for the treatment of cancer
AU2018235838B2 (en) 2017-03-16 2023-12-14 Alpine Immune Sciences, Inc. CD80 variant immunomodulatory proteins and uses thereof
EP3596116B1 (en) 2017-03-16 2023-09-06 Alpine Immune Sciences, Inc. Pd-l1 variant immunomodulatory proteins and uses thereof
WO2018170023A1 (en) 2017-03-16 2018-09-20 Alpine Immune Sciences, Inc. Pd-l2 variant immunomodulatory proteins and uses thereof
EP3600491B1 (en) 2017-03-28 2023-08-09 Amgen Inc. Plunger rod and syringe assembly system and method
BR112019022488A2 (en) 2017-04-28 2020-06-16 Merck Sharp & Dohme Corp. BIOMARCHERS FOR CANCER THERAPEUTICS
CN106974942A (en) * 2017-05-03 2017-07-25 武汉滨会生物科技股份有限公司 Application of the recombination oncolytic II herpes simplex virus types in anti-lymphadenoma, cancer of the esophagus, breast cancer, pancreatic cancer drug is prepared
AU2018280054B2 (en) 2017-06-08 2023-07-13 Amgen Inc. Syringe assembly for a drug delivery device and method of assembly
MX2019014615A (en) 2017-06-08 2020-02-07 Amgen Inc Torque driven drug delivery device.
CN107354136A (en) * 2017-06-15 2017-11-17 杭州睿可特生物科技有限公司 Recombinant herpes simplex virus and its preparation method and application
JP7195276B2 (en) 2017-06-22 2022-12-23 アムジエン・インコーポレーテツド Collision/shock reduction due to device activation
WO2018237225A1 (en) 2017-06-23 2018-12-27 Amgen Inc. Electronic drug delivery device comprising a cap activated by a switch assembly
ES2972207T3 (en) 2017-07-14 2024-06-11 Amgen Inc Needle insertion-retraction system with double torsion spring system
IL271173B2 (en) 2017-07-21 2024-04-01 Amgen Inc Gas permeable sealing member for drug container and methods of assembly
JP7242562B2 (en) 2017-07-25 2023-03-20 アムジエン・インコーポレーテツド Drug delivery device with container access system and associated method of assembly
MA49677A (en) 2017-07-25 2021-04-21 Amgen Inc DRUG DELIVERY DEVICE WITH GEAR MODULE AND ASSOCIATED ASSEMBLY PROCESS
CN118126157A (en) 2017-08-03 2024-06-04 美国安进公司 Interleukin-21 muteins and methods of treatment
EP3664844A1 (en) 2017-08-07 2020-06-17 Amgen Inc. Treatment of triple negative breast cancer or colorectal cancer with liver metastases with an anti pd-l1 antibody and an oncolytic virus
EP3664863A2 (en) 2017-08-09 2020-06-17 Amgen Inc. Hydraulic-pneumatic pressurized chamber drug delivery system
EP3668567A1 (en) 2017-08-18 2020-06-24 Amgen Inc. Wearable injector with sterile adhesive patch
US11103636B2 (en) 2017-08-22 2021-08-31 Amgen Inc. Needle insertion mechanism for drug delivery device
EP4141005B1 (en) 2017-09-08 2024-04-03 Amgen Inc. Inhibitors of kras g12c and methods of using the same
CN111065732A (en) 2017-09-11 2020-04-24 Imba-莫利库尔生物技术研究所 Tumor organoid model
US11759565B2 (en) 2017-10-04 2023-09-19 Amgen Inc. Flow adapter for drug delivery device
EP3691716B1 (en) 2017-10-06 2023-11-29 Amgen Inc. Drug delivery device with interlock assembly and related method of assembly
WO2019074579A1 (en) 2017-10-09 2019-04-18 Amgen Inc. Drug delivery device with drive assembly and related method of assembly
MA50360A (en) 2017-10-10 2020-08-19 Alpine Immune Sciences Inc CTLA-4 VARIANTS IMMUNOMODULATOR PROTEINS AND THEIR USES
CA3078517A1 (en) 2017-10-18 2019-04-25 Alpine Immune Sciences, Inc. Variant icos ligand immunomodulatory proteins and related compositions and methods
JP2021501150A (en) 2017-10-27 2021-01-14 メルク・シャープ・エンド・ドーム・コーポレイション Compositions and Methods for Treating Liver Cancer
EP3703779A1 (en) 2017-11-03 2020-09-09 Amgen Inc. Systems and approaches for sterilizing a drug delivery device
JP2021501616A (en) 2017-11-06 2021-01-21 アムジエン・インコーポレーテツド Drug delivery device with placement and flow detection
WO2019090303A1 (en) 2017-11-06 2019-05-09 Amgen Inc. Fill-finish assemblies and related methods
CN116832271A (en) 2017-11-10 2023-10-03 安进公司 Plunger for a drug delivery device
AU2018368340B2 (en) 2017-11-16 2024-03-07 Amgen Inc. Door latch mechanism for drug delivery device
US20230101432A1 (en) 2018-01-03 2023-03-30 Alpine Immune Sciences, Inc. Multi-domain immunomodulatory proteins and methods of use thereof
TW201930344A (en) 2018-01-12 2019-08-01 美商安進公司 Anti-PD-1 antibodies and methods of treatment
WO2019152821A1 (en) 2018-02-05 2019-08-08 The Brigham And Women's Hospital, Inc. Recombinant herpes simplex virus-2 expressing glycoprotein b and d antigens
CN112020510A (en) 2018-03-19 2020-12-01 茂体外尔公司 Methods and compositions comprising tumor suppressor gene therapy and CD122/CD132 agonists for treating cancer
EP3774877A1 (en) * 2018-03-28 2021-02-17 Bioxodes Anticoagulant fusion proteins and uses thereof
AU2019262056A1 (en) * 2018-05-01 2020-10-29 Albert Einstein College Of Medicine HSV-2-delta-gD vaccines and methods for their production and use
US10835685B2 (en) 2018-05-30 2020-11-17 Amgen Inc. Thermal spring release mechanism for a drug delivery device
US11083840B2 (en) 2018-06-01 2021-08-10 Amgen Inc. Modular fluid path assemblies for drug delivery devices
AU2019282239B2 (en) 2018-06-04 2024-06-06 Calidi Biotherapeutics (Nevada), Inc. Cell-based vehicles for potentiation of viral therapy
US11505782B2 (en) 2018-06-04 2022-11-22 Calidi Biotherapeutics, Inc. Cell-based vehicles for potentiation of viral therapy
AU2019282755A1 (en) * 2018-06-08 2021-01-28 The Board Of Trustees Of University Of Illinois Recombinant Herpes Simplex Virus for cancer immunotherapy
US20210253655A1 (en) * 2018-06-15 2021-08-19 Children's Hospital Medical Center Polypeptides, nucleic acid molecules, compositions, and related methods
US20210363219A1 (en) 2018-06-15 2021-11-25 Alpine Immune Sciences, Inc. Pd-1 variant immunomodulatory proteins and uses thereof
EP3826699A1 (en) 2018-07-24 2021-06-02 Amgen Inc. Delivery devices for administering drugs
MX2021000748A (en) 2018-07-24 2021-03-26 Amgen Inc Delivery devices for administering drugs.
WO2020023336A1 (en) 2018-07-24 2020-01-30 Amgen Inc. Hybrid drug delivery devices with grip portion
US20210260279A1 (en) 2018-07-24 2021-08-26 Amgen Inc. Hybrid drug delivery devices with optional grip portion and related method of preparation
CA3103105A1 (en) 2018-07-31 2020-02-06 Amgen Inc. Fluid path assembly for a drug delivery device
CN109161561A (en) * 2018-08-09 2019-01-08 湖北科技学院 A kind of the novel oncolytic virus and its construction method of selectively killing prostate gland cancer cell
US11242528B2 (en) 2018-08-28 2022-02-08 Actym Therapeutics, Inc. Engineered immunostimulatory bacterial strains and uses thereof
CA3106452A1 (en) 2018-09-24 2020-04-02 Amgen Inc. Interventional dosing systems and methods
IL281469B1 (en) 2018-09-28 2024-04-01 Amgen Inc Muscle wire escapement activation assembly for a drug delivery device
TW202027806A (en) 2018-10-02 2020-08-01 美商安進公司 Injection systems for drug delivery with internal force transmission
AR116607A1 (en) 2018-10-05 2021-05-26 Amgen Inc DRUG ADMINISTRATION DEVICE WITH DOSE INDICATOR
MA53913A (en) 2018-10-15 2022-01-19 Amgen Inc METHOD OF ASSEMBLING DRUG DELIVERY DEVICE PLATFORM
CN112789073B (en) 2018-10-15 2023-09-15 安进公司 Drug delivery device with damping mechanism
WO2020091956A1 (en) 2018-11-01 2020-05-07 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
TWI831847B (en) 2018-11-01 2024-02-11 美商安進公司 Drug delivery devices with partial needle retraction and methods for operating the same
US20220031939A1 (en) 2018-11-01 2022-02-03 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
JP7329593B2 (en) 2018-11-06 2023-08-18 カリディ・バイオセラピューティクス・インコーポレイテッド An enhanced system for cell-mediated oncolytic virus therapy
EP3884041A2 (en) 2018-11-21 2021-09-29 Indapta Therapeutics, Inc. Methods for expansion of natural killer (nk) cell subset and related compositions and methods
JP2022510276A (en) 2018-11-30 2022-01-26 アルパイン イミューン サイエンシズ インコーポレイテッド CD86 variant immunomodulatory protein and its use
JP2022516077A (en) 2018-12-27 2022-02-24 アムジェン インコーポレイテッド Freeze-dried virus preparation
CA3176660A1 (en) 2019-02-27 2020-09-03 Actym Therapeutics, Inc. Immunostimulatory bacteria engineered to colonize tumors, tumor-resident immune cells, and the tumor microenvironment
US12024709B2 (en) 2019-02-27 2024-07-02 Actym Therapeutics, Inc. Immunostimulatory bacteria engineered to colonize tumors, tumor-resident immune cells, and the tumor microenvironment
EP3935182A1 (en) 2019-03-05 2022-01-12 Amgen Inc. Use of oncolytic viruses for the treatment of cancer
EP3938524A1 (en) * 2019-03-14 2022-01-19 Massachusetts Institute of Technology Engineered herpes simplex virus-1 (hsv-1) vectors and uses thereof
TW202102543A (en) 2019-03-29 2021-01-16 美商安進公司 Use of oncolytic viruses in the neoadjuvant therapy of cancer
JP7510952B2 (en) 2019-04-24 2024-07-04 アムジエン・インコーポレーテツド Syringe sterility verification assembly and method
JP2022545227A (en) 2019-08-23 2022-10-26 アムジエン・インコーポレーテツド Drug delivery device with configurable needle shield-engaging component and related methods
WO2021113644A1 (en) 2019-12-05 2021-06-10 Multivir Inc. Combinations comprising a cd8+ t cell enhancer, an immune checkpoint inhibitor and radiotherapy for targeted and abscopal effects for the treatment of cancer
WO2021127524A1 (en) * 2019-12-20 2021-06-24 Krystal Biotech, Inc. Compositions and methods for gene delivery to the airways and/or lungs
JP2023523429A (en) 2020-04-22 2023-06-05 インダプタ セラピューティクス インコーポレイテッド Natural killer (NK) cell compositions and methods for producing same
WO2022015407A1 (en) * 2020-07-16 2022-01-20 Massachusetts Institute Of Technology Simultaneous delivery of cancer treatment programs to tumor and immune cells
WO2022147481A1 (en) 2020-12-30 2022-07-07 Ansun Biopharma Inc. Combination therapy of an oncolytic virus delivering a foreign antigen and an engineered immune cell expressing a chimeric receptor targeting the foreign antigen
WO2022170219A1 (en) 2021-02-05 2022-08-11 Iovance Biotherapeutics, Inc. Adjuvant therapy for cancer
KR20240011135A (en) 2021-05-21 2024-01-25 암젠 인크 How to Optimize Filling Recipes for Drug Containers
CN115707781A (en) * 2021-08-20 2023-02-21 广东东阳光药业有限公司 HSV (herpes simplex virus) vector and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5585096A (en) * 1994-06-23 1996-12-17 Georgetown University Replication-competent herpes simplex virus mediates destruction of neoplastic cells
US6428968B1 (en) * 1999-03-15 2002-08-06 The Trustees Of The University Of Pennsylvania Combined therapy with a chemotherapeutic agent and an oncolytic virus for killing tumor cells in a subject
US7223593B2 (en) * 2000-01-21 2007-05-29 Biovex Limited Herpes virus strains for gene therapy

Family Cites Families (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5163949A (en) * 1990-03-02 1992-11-17 Bonutti Peter M Fluid operated retractors
US6610287B1 (en) 1990-04-16 2003-08-26 The General Hospital Corporation Transfer and expression of gene sequences into nervous system cells using herpes simplex virus mutants with deletions in genes for viral replication
US5328688A (en) * 1990-09-10 1994-07-12 Arch Development Corporation Recombinant herpes simplex viruses vaccines and methods
GB9102126D0 (en) 1991-01-31 1991-03-13 Smithkline Beecham Biolog Novel vaccine
GB9202933D0 (en) 1992-02-12 1992-03-25 Smithkline Beecham Biolog Vaccines
JP3974167B2 (en) 1992-03-31 2007-09-12 アーチ デベロプメント コーポレイション Methods and compositions for treating genes, tumors and viral infections, and preventing programmed cell death (apoptosis)
GB9325496D0 (en) 1993-12-14 1994-02-16 Smithkline Beecham Biolog Vaccines
US5728379A (en) 1994-06-23 1998-03-17 Georgetown University Tumor- or cell-specific herpes simplex virus replication
GB9423663D0 (en) * 1994-11-23 1995-01-11 Cantab Pharma Res Viral preparations, immunogens, and vaccines
NL9500216A (en) * 1995-02-06 1996-09-02 Bio Pharma Sciences Bv Pharmaceutical composition for the treatment of herpes.
ATE324435T1 (en) * 1995-02-21 2006-05-15 Cantab Pharmaceuticals Res Ltd VIRAL PREPARATIONS, VECTORS, IMMUNOGENS AND VACCINES
ZA966287B (en) 1995-07-27 1998-03-09 American Cyanamid Co Avirulent herpetic viruses useful as tumoricidal agents and vaccines.
US6344445B1 (en) * 1995-10-19 2002-02-05 Cantab Pharmaceutical Research Limited Herpes virus vectors and their uses
GB9524973D0 (en) * 1995-12-06 1996-02-07 Lynxvale Ltd Viral vectors
EP1314430A1 (en) 1996-01-25 2003-05-28 The University Court Of The University Of Glasgow Treatment of non-neuronal cancer using HSV mutant
US5824318A (en) 1996-07-24 1998-10-20 American Cyanamid Company Avirulent herpetic viruses useful as tumoricidal agents and vaccines
US5876923A (en) 1996-07-26 1999-03-02 Arch Development Corporation Herpes simplex virus ICP4 as an inhibitor of apoptosis
GB9615794D0 (en) * 1996-07-26 1996-09-04 Medical Res Council Mutant herpes simplex virus strains and uses thereof
GB2322130B (en) 1997-02-13 2000-12-20 Secr Defence A vaccine against Simian Herpes B virus
GB9704046D0 (en) * 1997-02-27 1997-04-16 Univ Leeds Arrestable therapeutic
US6051428A (en) * 1997-03-21 2000-04-18 Sloan-Kettering Institute For Cancer Research Rapid production of autologous tumor vaccines
US5998174A (en) 1997-05-12 1999-12-07 University Of Pittsburgh Of The Commonwealth System Of Higher Education Multigene vectors
US6379674B1 (en) 1997-08-12 2002-04-30 Georgetown University Use of herpes vectors for tumor therapy
US20030044384A1 (en) * 1997-10-09 2003-03-06 Pro-Virus, Inc. Treatment of neoplasms with viruses
GB9801930D0 (en) 1998-01-29 1998-03-25 Univ London Mutant herpes simplex viruses and uses thereof
EP1061806A4 (en) 1998-03-12 2001-09-12 Univ Pennsylvania Producer cells for replication selective viruses in the treatment of malignancy
GB9810904D0 (en) 1998-05-20 1998-07-22 Univ London Mutant herpes simplex viruses and uses thereof
GB9816781D0 (en) 1998-07-31 1998-09-30 Univ London Herpes virus vectors for dendritic cells
US6713067B2 (en) * 1998-07-31 2004-03-30 Biovex Limited Herpes viruses for immune modulation
US6375952B1 (en) 1998-08-07 2002-04-23 University Of Washington Immunological herpes simplex virus antigens and methods for use thereof
EP1141338A4 (en) 1998-12-31 2002-09-25 Arch Dev Corp Recombinant herpes simplex virus useful for treating neoplastic disease
US6764675B1 (en) * 1999-06-08 2004-07-20 The Uab Research Foundation Herpes simplex virus expressing foreign genes and method for treating cancers therewith
AU6120600A (en) 1999-06-08 2000-12-28 Uab Research Foundation Herpes simplex virus expressing foreign genes and method for treating cancers therewith
IL131212A0 (en) * 1999-08-03 2001-01-28 Yissum Res Dev Co Recombinant virus and live-virus vaccines
GB9930418D0 (en) 1999-12-22 2000-02-16 Neurovex Ltd Replication incompetent herpes virus vectors
GB9930419D0 (en) 1999-12-22 2000-02-16 Neurovex Ltd Replication incompetent herpes virus vectors
GB0001476D0 (en) 2000-01-21 2000-03-15 Neurovex Ltd Herpes virus strains
US7063851B2 (en) 2000-04-12 2006-06-20 Biovex Limited Herpes viruses for immune modulation
GB0317511D0 (en) * 2003-07-25 2003-08-27 Biovex Ltd Viral vectors
ES2831080T5 (en) * 2016-01-08 2023-10-30 Replimune Ltd Modified oncolytic virus

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5585096A (en) * 1994-06-23 1996-12-17 Georgetown University Replication-competent herpes simplex virus mediates destruction of neoplastic cells
US6428968B1 (en) * 1999-03-15 2002-08-06 The Trustees Of The University Of Pennsylvania Combined therapy with a chemotherapeutic agent and an oncolytic virus for killing tumor cells in a subject
US7223593B2 (en) * 2000-01-21 2007-05-29 Biovex Limited Herpes virus strains for gene therapy
US7537924B2 (en) * 2000-01-21 2009-05-26 Biovex Limited Virus strains
US8277818B2 (en) * 2000-01-21 2012-10-02 Biovex Limited Herpes virus strains for gene therapy
US8680068B2 (en) * 2000-01-21 2014-03-25 Biovex Limited Herpes virus strains

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Rekabdar et al. (1999) Clin. Diag. Lab. Immunol., Vol. 6 (6), 826-231 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150202290A1 (en) * 2012-08-30 2015-07-23 Amgen Inc. Method for treating melanoma using a herpes simplex virus and an immune checkpoint inhibitor
US10034938B2 (en) * 2012-08-30 2018-07-31 Amgen Inc. Method for treating melanoma using a herpes simplex virus and an immune checkpoint inhibitor
WO2020109389A1 (en) 2018-11-28 2020-06-04 Innovative Molecules Gmbh Helicase primase inhibitors for treating cancer in a combination therapy with oncolytic viruses

Also Published As

Publication number Publication date
BRPI0107737B1 (en) 2016-11-16
US20120164108A1 (en) 2012-06-28
IL150677A0 (en) 2003-02-12
JP2019048864A (en) 2019-03-28
CN1250732C (en) 2006-04-12
HK1047451A1 (en) 2003-02-21
IL150678A0 (en) 2003-02-12
BRPI0107737B8 (en) 2021-05-25
DK1252323T3 (en) 2006-04-03
US8680068B2 (en) 2014-03-25
CA2398335A1 (en) 2001-07-26
US8277818B2 (en) 2012-10-02
US20070264282A1 (en) 2007-11-15
DK1252322T4 (en) 2009-06-08
JP2017132779A (en) 2017-08-03
BR0107737A (en) 2002-11-19
WO2001053505A3 (en) 2001-12-27
LU93100I2 (en) 2016-08-08
IL150677A (en) 2008-12-29
US20140154216A1 (en) 2014-06-05
US20120321599A1 (en) 2012-12-20
ES2254359T3 (en) 2006-06-16
NL300821I2 (en) 2016-10-27
WO2001053506A3 (en) 2002-05-23
LU93101I2 (en) 2016-08-08
US20200032219A1 (en) 2020-01-30
ES2233600T3 (en) 2005-06-16
GB2375113B (en) 2004-10-20
JP2012072156A (en) 2012-04-12
US20030091537A1 (en) 2003-05-15
EP1252323A2 (en) 2002-10-30
ES2233600T5 (en) 2009-06-22
FR16C0026I1 (en) 2016-07-22
EP1252322B1 (en) 2004-11-17
GB0219030D0 (en) 2002-09-25
GB2374873C (en) 2011-07-27
CN1418255A (en) 2003-05-14
JP2003520044A (en) 2003-07-02
US20030113348A1 (en) 2003-06-19
CY2016020I2 (en) 2016-12-14
WO2001053505A2 (en) 2001-07-26
CY2016021I2 (en) 2016-12-14
EP1252322A2 (en) 2002-10-30
WO2001053506A2 (en) 2001-07-26
JP2015221813A (en) 2015-12-10
JP2003520789A (en) 2003-07-08
US20070003571A1 (en) 2007-01-04
BRPI0107736B1 (en) 2019-12-17
GB0219033D0 (en) 2002-09-25
HK1047297A1 (en) 2003-02-14
KR20020080383A (en) 2002-10-23
KR100802403B1 (en) 2008-02-13
BRPI0107736B8 (en) 2021-05-25
AU2001226951B8 (en) 2006-10-26
JP4810042B2 (en) 2011-11-09
KR20030032913A (en) 2003-04-26
AU2695101A (en) 2001-07-31
EP1252323B1 (en) 2005-12-07
GB2374873A (en) 2002-10-30
AU2694701A (en) 2001-07-31
CN1425073A (en) 2003-06-18
EP2177619A1 (en) 2010-04-21
FR16C0026I2 (en) 2018-04-27
DE60115600T2 (en) 2006-07-20
HK1047451B (en) 2005-03-18
US7537924B2 (en) 2009-05-26
AU2001226951B2 (en) 2006-10-05
US20200032218A1 (en) 2020-01-30
EP1568779A1 (en) 2005-08-31
JP4921669B2 (en) 2012-04-25
CA2398343A1 (en) 2001-07-26
US20090220460A1 (en) 2009-09-03
PT1252322E (en) 2005-03-31
CY2016020I1 (en) 2016-12-14
JP6424145B2 (en) 2018-11-14
DE60107203D1 (en) 2004-12-23
US20150232812A1 (en) 2015-08-20
CA2398335C (en) 2010-12-07
EP1252322B2 (en) 2009-03-04
GB2374873B (en) 2004-05-26
US10301600B2 (en) 2019-05-28
GB2375113A (en) 2002-11-06
ATE282708T1 (en) 2004-12-15
AU782659B2 (en) 2005-08-18
BR0107736A (en) 2002-11-19
DE60107203T3 (en) 2009-07-23
DE60107203T2 (en) 2005-12-01
HK1047297B (en) 2004-10-21
BE2016C033I2 (en) 2020-05-20
NL300820I2 (en) 2016-11-17
KR100768408B1 (en) 2007-10-18
ATE312189T1 (en) 2005-12-15
DE60115600D1 (en) 2006-01-12
CY2016021I1 (en) 2016-12-14
US7223593B2 (en) 2007-05-29
DK1252322T3 (en) 2005-03-14
US7063835B2 (en) 2006-06-20
IL150678A (en) 2008-12-29
JP6644378B2 (en) 2020-02-12

Similar Documents

Publication Publication Date Title
US20200032219A1 (en) Herpes virus strains
US7981669B2 (en) Viral vectors
AU2001226951A1 (en) Virus strains for the oncolytic treatment of cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: BIOVEX LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:COFFIN, ROBERT S.;REEL/FRAME:035361/0770

Effective date: 20070210

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION