WO2019020543A1 - Virus oncolytiques exprimant des agents ciblant des modulateurs immunitaires métaboliques - Google Patents

Virus oncolytiques exprimant des agents ciblant des modulateurs immunitaires métaboliques Download PDF

Info

Publication number
WO2019020543A1
WO2019020543A1 PCT/EP2018/069867 EP2018069867W WO2019020543A1 WO 2019020543 A1 WO2019020543 A1 WO 2019020543A1 EP 2018069867 W EP2018069867 W EP 2018069867W WO 2019020543 A1 WO2019020543 A1 WO 2019020543A1
Authority
WO
WIPO (PCT)
Prior art keywords
virus
oncolytic virus
oncolytic
cancer
cells
Prior art date
Application number
PCT/EP2018/069867
Other languages
English (en)
Inventor
Sylvain Robin
Original Assignee
Transgene Sa
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Transgene Sa filed Critical Transgene Sa
Publication of WO2019020543A1 publication Critical patent/WO2019020543A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/50Hydrolases (3) acting on carbon-nitrogen bonds, other than peptide bonds (3.5), e.g. asparaginase
    • 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
    • A61K35/768Oncolytic viruses not provided for in groups A61K35/761 - A61K35/766
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y305/00Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5)
    • C12Y305/04Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5) in cyclic amidines (3.5.4)
    • C12Y305/04004Adenosine deaminase (3.5.4.4)
    • 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
    • C12N2710/24011Poxviridae
    • C12N2710/24111Orthopoxvirus, e.g. vaccinia virus, variola
    • C12N2710/24132Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent

Definitions

  • the present invention is in the field of oncolytic viruses and agents targeting metabolic immune modulators.
  • the invention provides oncolytic viruses comprising, inserted in their genomes, a nucleotide sequence encoding at least one agent targeting one or more metabolic immune modulator(s). More precisely, said agents play a role in the adenosine pathway, by decreasing the adenosine concentration. More particularly, said agents have an activity of degradation of adenosine. Even more particularly, said agents are adenosine deaminases (ADA) or have an adenosine deaminase activity.
  • ADA adenosine deaminases
  • These oncolytic viruses can be used for the prevention and/or the treatment of proliferative diseases like cancers.
  • Oncolytic viruses are a class of therapeutic agents that have the unique property of tumour- dependent self-perpetuation (Hermiston et al., 2006, Curr. Opin. Mol. Ther., 8(4):322-30). The benefit of using these viruses is that as they replicate, they kill their host cells by lysis. Oncolytic viruses are capable of selective replication in dividing cells (e.g. cancer cells) while leaving non- dividing cells (e.g. normal cells) unharmed. As the infected dividing cells are destroyed by lysis, they release new infectious particles to infect the surrounding dividing cells. Therefore, oncolytic viruses offer a new area for treating cancer, optionally in association with conventional treatments for cancer (Fisher et al., 2006, Curr. Opin. Mol.
  • Cancer cells are ideal hosts for many viruses because they have the antiviral interferon pathway inactivated or have mutated tumour suppressor genes that enable viral replication to proceed unhindered (Chernajovsky et al., 2006, BMJ, 332(7534):170-2).
  • viruses including adenovirus, reovirus, measles, herpes simplex, Newcastle disease virus and vaccinia virus have now been clinically tested as oncolytic agents.
  • viruses are naturally oncolytic and have an innate ability to selectively infect and kill tumour cells.
  • oncolytic viruses can also be engineered by modifying naturally occurring viruses.
  • the main strategies used currently to modify the viruses include: functional deletions in viral genes, the use of tumour- or tissue-specific promoters to control the expression of these viral genes, tropism modification to redirect virus to the cancer cell surface, among many other possibilities.
  • Vaccinia virus a Poxviridae
  • Viruses can also be engineered by introducing modifications in the genome in order to improve their ability to infect and lyse tumor cells notably in the in vivo context while preserving safety.
  • Several gene modifications have already been considered in the art to enhance safety in the host organism.
  • increasing safety is often detrimental to efficiency in dividing cells.
  • TK thymidine kinase deleted virus showed a decreased lytic activity in non-dividing cells compared to wild type virus (Buller et al., 1985, Nature, 317(6040):813-5). Therefore, armament of oncolytic viruses has been considered for several decades and various enzyme-prodrug systems have been widely used to enhance the oncolytic efficacy of the virus therapy.
  • armament with the so-called FCUl suicide gene encoding a bifunctional chimeric protein that combines the enzymatic activities of cytidine deaminase (CDase) and uracil phosphoribosyl transferase (UPRTase), and catalyses the conversion of nontoxic 5-fluorocytosine (5-FC) into the toxic metabolites 5-fluorouracil (5-FU) and 5-fluorouridine-5'monophosphate (5-FU P), was shown to improve antitumoral effect in the presence of 5-FC, to exert a strong bystander effect, thus permitting elimination of neighbouring uninfected tumor cells and to bypass the natural resistance of certain human tumor cells to 5-fluorouracil (Erbs et al., 2000, Cancer Res., 60(14):3813).
  • a TK gene-deleted VV expressing the FCUl gene showed potent anti-tumour effect both in vitro and in vivo in a murine model of
  • TME tumor microenvironement
  • adenosine This purine nucleotide exerts pleiotropic functions in the cell metabolism (Kumar et al., 2009, Eur J Pharmacol., 616(l-3):7-15). Notably, intracellular adenosine is involved in energy metabolism, nucleic acid metabolism, and the methionine cycle, while extracellular adenosine plays an important role in intercellular signalling, affecting diverse physiological functions including neurological, cardiovascular, and immunological systems (Ohta A., 2016, Front. Immunol., 7:109).
  • extracellular adenosine Upon binding with adenosine receptors present on the surface of various immune cells, extracellular adenosine suppresses pro-inflammatory activities and upregulates a number of anti-inflammatory molecules and immunoregulatory cells (e.g. T cells, NK cells, neutrophils, etc.,), leading to the establishment of a long-lasting immunosuppressive environment (Ohta A., 2016, Front. Immunol., 7:109).
  • immunoregulatory cells e.g. T cells, NK cells, neutrophils, etc.
  • Adenosine has also been shown to constitute one of the important mechanisms used by solid tumours to evade the immune system.
  • the adenosine pathway also regulates cancer growth and dissemination by interfering with cancer cell proliferation, apoptosis and angiogenesis via adenosine receptors that are expressed on cancer cells and endothelial cells, respectively.
  • Solid tumours express high levels of CD39 and CD73, as well as low levels of nucleoside transporters (NTs), ecto-adenosine deaminase and its cofactor CD26, which lead to an increase in adenosine signalling in the cancer environment (Antonioli et al., 2013, Nature Reviews Cancer 13, 842-857).
  • NTs nucleoside transporters
  • ecto-adenosine deaminase ecto-adenosine deaminase and its cofactor CD26
  • adenosinergic machinery As a result, several components of the adenosinergic machinery actually constitute potential therapeutic targets in immune-oncology, in order to promote anti-tumour immunity and to enhance the activity of some anti-cancer treatments.
  • Different ways have been explored to reduce the adenosine effects: blocking of adenosine receptors (e.g. A2A, A2B), modulation of CD39, CD73, or of the activity or concentration of adenosine-degradating enzymes such as the adenosine deaminase enzyme (ADA), etc., the two major targets being the blockade of A2A and CD73.
  • A2A-deficient mice had increased anti-tumour immunity, and that A2A blockade was an effective approach to re-instate anti-tumour immunity (Ohta et al., 2001, Nature 414(6866), 916-920).
  • This pathway was confirmed by several groups who showed that A2A activation on CD8 + anti-tumour T cells and NK cells was the predominant pathway exploited by adenosine-rich tumour microenvironment to dampen anti-tumour immunity.
  • Antagonists against A2A receptor are currently being evaluated: PBF-509, a small molecule inhibitor of A2A entered phase I in 2015 for the treatment of NSCLC.
  • CD73 is another target of the adenosinergic machinery. This enzyme, which converts AMP to adenosine, has immunosuppressive functions and is highly expressed in various types of solid tumours. High rates of CD73 have been correlated with lower survival in colorectal cancers (Wu et al., 2012, J Surg Oncol, DOI: 10.1002/jso.23056) and in gastric cancers (Lu et al., 2013, World J Gastroenterol, 19(12): 1912-1918). The blockade of CD73 is now considered as clinically relevant approach for cancer immunotherapy (Allard et al., 2016, Immunotherapy, DOI: 10.2217/imt.l5.106).
  • MEDI9447 a blocking anti-CD73 monoclonal antibody, is being evaluated in phase 1 clinical trial in adults with advanced solid tumours, as single agent and in combination with anti-PDLl mAb durvalumab.
  • ADA activity levels have been described in different types of cancer and in inflammatory diseases such as rheumatoid arthritis, celiac disease, ulcerative colitis, systemic lupus erythematosus, visceral leishmaniasis and inflammatory obesity or infections such as tuberculosis in human serum, saliva or sputum samples (Ungerer et al, 1992, Clin Chem 38/7, 1322- 1326).
  • ADA deficiency causes severe combined immunodeficiency disease (ADA-SCID), in which both B-cell and T-cell development is impaired.
  • ADA-SCID severe combined immunodeficiency disease
  • PEG-ADA has been approved for use for treating inherited ADA deficiency (Chaffee et al., 1992, J Clin Invest, 89:1643-1651). More recently, PEG-ADA2 (human adenosine deaminase 2) has been explored for treating solid tumours (Wang et al., 2006, Abstract 1472, AACR Annual Meeting) and reduction of lung metastasis was observed in PEG-ADA2 treated 4T1 animal model, as well as reduction in TME adenosine levels.
  • the inventors have now generated an oncolytic virus engineered to express an adenosine degrading agent. Against all expectations, this adenosine deaminase (ADA)-expressing oncolytic vaccinia virus is able to secrete an active enzyme and, upon administration to animals, to delay tumor growth and increase animal survival in two different tumor models compared to empty oncolytic viruses.
  • the inventors thus provide an effective anti-tumoral therapy allowing a combined effect of tumour cells lysis and modification of the TME, resulting in a significant reduction of tumour progression. Based on these results, one may anticipate that the oncolytic viruses of the invention may be successfully used as an alternative to other existing oncolytic viruses and may have a better efficiency profile.
  • the oncolytic viruses of the invention can also be exploited in combination with additional anticancer therapy/ies.
  • One aspect of the invention relates to an oncolytic virus comprising a nucleotide sequence encoding at least one agent degrading one or more metabolic immune modulator(s).
  • said metabolic immune modulator is an adenosine
  • said nucleotide sequence encodes an adenosine deaminase (ADA) polypeptide, or a polypeptide having an adenosine deaminase activity.
  • ADA adenosine deaminase
  • the oncolytic virus of the invention is a poxvirus belonging to the Orthopoxvirus and especially a vaccinia virus or a cowpox virus.
  • the oncolytic virus of the invention is defective in the thymidine kinase (TK-) encoding J2R locus. In still another embodiment, the oncolytic virus of the invention is defective in the I4L and/or F4L locus (alternatively or in combination with a defective TK- locus).
  • TK- thymidine kinase
  • the present invention further provides a composition comprising the oncolytic virus of the present invention and a pharmaceutically acceptable vehicle.
  • the oncolytic virus is preferably formulated for intravenous or intra-tumoral administration.
  • the present invention also concerns a process for preparing the oncolytic virus, comprising at least the steps of introducing said oncolytic virus into a producer cell, culturing the producer cell under conditions that are appropriate for enabling said oncolytic virus to be produced and recovering the produced virus from the cell culture.
  • the recovered oncolytic virus can be purified at least partially.
  • the present invention provides the oncolytic virus or the composition of the invention, for use for the prophylaxis and/or the treatment of a disease.
  • said disease is a proliferative disease such as cancers and restenosis.
  • Said cancer is preferably selected from the group consisting in renal cancer, bladder cancer, prostate cancer, breast cancer, colorectal cancer, lung cancer, hepatic cancer, gastric cancer, pancreatic cancer, melanoma, ovarian cancer and glioblastoma, and especially metastatic ones.
  • said disease is a disease associated with an increased osteoclast activity like rheumatoid arthritis and osteoporosis.
  • the present invention provides a method of treating a disease which comprises the administration into a host organism in need thereof of a therapeutically effective amount of the oncolytic virus or the composition of the invention.
  • Said method of treatment may be used in conjunction with one or more additional therapies such as ones selected from the group consisting of surgery, radiotherapy, chemotherapy, cryotherapy, hormonal therapy, toxin therapy, immunotherapy or cytokine therapy.
  • Figure 1 Schematic representation of the genetic constructs for the expression of ADAs by vaccinia virus WR TK- RR- and COP TK- RR-
  • WR western reserve
  • COP Copenhagen
  • huADAl human ADA1
  • huADA2 human ADA2
  • cuADA culex quinquefasciatus ADA
  • pllk7.5 promoter
  • SS signal sequence
  • FLAG FLAG sequence.
  • MCA205 murine fibrosarcoma MCA205 (Fig 2A) and murine colon carcinoma CT26 (Fig 2B) cell lines by ADA expressing viruses WRTG19009, WRTG19010 and WRTG19021 or the empty vector WRTG18011.
  • MCA205 were infected at OI of 0.01 and as CT26 cells are more resistant to vaccinia virus infection, they were infected at MOI of 0.1.
  • the adenosine degradation is given as the quantity of adenosine deaminated in nmoles per minute per millilitre of culture supernatant. Values are represented in mean ( ⁇ standard error of mean) of three individual determinations.
  • MCA205 cells were implanted subcutaneousiy (s.c.) in C57BL/6 mice. Tumours were then treated with two intratumoral (i.t.) administrations at day 7 and 10 with vehicle alone (Fig 5A) or with lxlO 7 PFU of TK and RR-defective virus (empty WRTG18011; Fig 5B), expressing the human ADA1 (WRTG19009; Fig 5C) or expressing the human ADA2 (WRTG19021; Fig 5D). Each plot represents one of five treated animals.
  • huADAl and huADA2 were detected by western blotting on culture supernatants after infection of HCT 116 cells with VVTG17989, COPTG19183 and COPTG19185.
  • the presence of huADAl (41 kDa) and huADA2 (56 kDa) is indicated (arrows).
  • the specific adenosine deaminase activity was measured on culture supernatants after 72 hours infection of human HCT 116 and human LoVo cell lines by huADA expressing viruses COPTG19183 and COPTG19185 or the empty vector VVTG17989.
  • HCT 116 were infected at MOI of 0.001.
  • LoVo cells are more resistant to vaccinia virus infection, they were infected at MOI of 0.01.
  • the adenosine degradation is given as the quantity of inosine produced from adenosine in pmoles per minute per microlitres of culture supernatant.
  • the enzymatic reaction was initiated with addition of adenosine at 2 mM (Fig 10A and Fig 10B) or 20 ⁇ (Fig IOC and Fig 10D). Values are represented in mean ( ⁇ standard error of mean) of three individual determinations.
  • the intratumoral replication efficacy was evaluated in HCT 116 tumors collected 7 days after treatment with one single intravenous injection of 10 7 pfu of COPTG19183 and COPTG19185 or the empty vector VVTG17989. Values are represented in mean ( ⁇ standard error of mean) of three individual determinations.
  • huADAl The specific detection of huADAl was performed by western blotting on interstitial fluids collected from HCT 116 tumors 7 days after treatment with one single intravenous injection of 10 7 pfu of VVTG17989, COPTG19183. The presence of huADAl is indicated (arrow).
  • the specific adenosine deaminase activity was measured in interstitial fluids collected from HCT 116 tumors 7 days after treatment with one single intravenous injection of 10 7 pfu of VVTG17989, COPTG19183 and COPTG19185.
  • the adenosine degradation is given as the quantity of inosine produced from adenosine in pmoles per minute per microlitres of culture supernatant.
  • the enzymatic reaction was initiated with addition of adenosine at 2 mM (Fig 13A) or 20 ⁇ (Fig 13B). Values are represented in mean ( ⁇ standard error of mean) of three individual determinations.
  • HCT 116 cells were implanted subcutaneously (s.c.) in Swiss nude mice. Tumours were then treated with a single intravenous (i.v.) administration at day 14 with vehicle alone (Fig 14A) or with lxlO 7 PFU of TK and RR-defective virus (empty VVTG11989; Fig 14B), expressing the human ADAl (COPTG19183; Fig 14C) or expressing the human ADA2 (COPTG19185; Fig 14D). Each plot represents one of fifteen treated animals.
  • an oncolytic virus encompasses a single oncolytic 20 virus as well as a plurality of oncolytic viruses, including mixtures thereof.
  • one or more refers to either one or a number above one (e.g. 2, 3, 4, etc.).
  • the terms “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are open- 30 ended and do not exclude additional, un-recited elements or method steps.
  • the expression “consisting essentially of” means excluding other components or steps of any essential significance. Thus, a composition consisting essentially of the recited components would not exclude traces, contaminants and pharmaceutically acceptable carriers. "Consisting of” shall mean excluding more than trace elements of other components or steps.
  • polypeptide refers to polymers of amino acid residues which comprise at least nine or more amino acids bonded via peptide bonds.
  • the polymer can be linear, branched or cyclic and may comprise naturally occurring and/or amino acid analogues and it may be interrupted by non-amino acids.
  • amino acid polymer is more than 50 amino acid residues, it is preferably referred to as a polypeptide or a protein whereas if it is 50 amino acids long or less, it is referred to as a "peptide”.
  • nucleic acid refers to any length of either polydeoxyribonucleotides (DNA) (e.g. cDNA, genomic DNA, plasmids, vectors, viral genomes, isolated DNA, probes, primers and any mixture thereof) or polyribonucleotides (RNA) (e.g. mRNA, antisense RNA, siRNA) or mixed polyribo- polydeoxyribonucleotides.
  • DNA polydeoxyribonucleotides
  • RNA e.g. mRNA, antisense RNA, siRNA
  • mixed polyribo- polydeoxyribonucleotides encompass single or double-stranded, linear or circular, natural or synthetic, modified or unmodified polynucleotides.
  • a polynucleotide may comprise non- naturally occurring nucleotides and may be interrupted by non-nucleotide components.
  • agent refers to a substance that induces or is capable of inducing an effect, like a biological effect or a chemical reaction.
  • Said agents can be biological agents, chemical agents or pharmaceutical agents.
  • agents include, but are not limited to, polypeptides (e.g.
  • nucleic acids e.g.: DNA, RNA, miRNA, siRNA, etc.
  • gene regulators lipids, small molecules, cytotoxins, drugs, acids, bases, oxidizing agents, reducing agents, alkaline agents, nucleophilic agents, electrophilic agents, polymers, synthetic materials, viruses, bacterias, archaea, protozoa, fungi, algae, and combinations thereof.
  • the agents encoded by the oncolytic are polypeptides.
  • degradation refers to the transformation or reduction of a compound to one less complex, as by splitting off one or more groups.
  • One of a result of the degradation of a compound is the diminution of said compound concentration.
  • degradation reactions include, but are not limited to hydrolysis, deamination, dephosphorilation, Reactive Oxygene Species degradations, etc.
  • said degradation consists in a deamination, or the removal of at least one amino group from an amino acid or another compound.
  • host cell should be understood broadly without any limitation concerning particular organization in tissue, organ, or isolated cells.
  • Such cells may be of a unique type of cells or a group of different types of cells such as cultured cell lines, primary cells and dividing cells.
  • the term "host cells” include prokaryotic cells, lower eukaryotic cells such as yeast, and other eukaryotic cells such as insect cells, plant and mammalian (e.g. human or non-human) cells as well as cells capable of producing the oncolytic virus of the invention. This term also includes cells which can be or has been the recipient of the virus described herein as well as progeny of such cells.
  • virus refers to a vehicle comprising at least one element of a wild-type virus genome and may be packaged into a viral particle or to a viral particle.
  • viral particles may or may not contain nucleic acid (i.e. the viral genome) it is preferred that a virus comprises a DNA or RNA viral genome packaged into a viral particle (or virion) and is infectious (i.e. capable of infecting and entering a host cell or subject).
  • the virus of this invention is associated with a DNA genome, and most preferably a double-stranded DNA genome.
  • a "virus” includes wild-type and engineered viruses.
  • Naturally occurring or wild-type or “native” is used to describe a biological molecule or organism that can be found in nature as distinct from being artificially produced by man.
  • a virus which can be isolated from a source in nature is wild-type.
  • the present invention also encompasses wild-type viruses that can be obtained from specific collections (e.g. ECCAC, ATCC, CNCM, etc.).
  • ECCAC ECCAC
  • ATCC ATCC
  • CNCM etc.
  • non- naturally occurring viruses include, among many others, recombinant viruses engineered by insertion of one or more gene(s) of interest in the viral genome and mutated viruses engineered by total or partial deletion of a viral gene to make the modified virus defective for the encoded gene product (e.g. TK- virus) as well as chimeric viruses containing genomic fragments obtained from different virus origins.
  • recombinant viruses engineered by insertion of one or more gene(s) of interest in the viral genome and mutated viruses engineered by total or partial deletion of a viral gene to make the modified virus defective for the encoded gene product (e.g. TK- virus) as well as chimeric viruses containing genomic fragments obtained from different virus origins.
  • originating or “originate” is used to identify the original source of a component (e.g. polypeptide, nucleic acid molecule, virus, etc.) but is not meant to limit the method by which the component is made which can be, for example, by chemical synthesis or recombinant means.
  • a component e.g. polypeptide, nucleic acid molecule, virus, etc.
  • oncolytic virus refers to a virus capable of selectively replicating in dividing cells (e.g. a proliferative cell such as a cancer cell) with the aim of slowing the growth and/or lysing said dividing cell, either in vitro or in vivo, while showing no or minimal replication in non-dividing cells (e.g. primary cells).
  • dividing cells e.g. a proliferative cell such as a cancer cell
  • the term “oncolytic virus” encompasses any virus naturally occurring, engineered or otherwise modified.
  • treatment encompasses prophylaxis (e.g. preventive measure in a subject at risk of having the pathological condition to be treated) and/or therapy (e.g. in a subject diagnosed as having the pathological condition), eventually in association with conventional therapeutic modalities.
  • the result of the treatment is to slow down, cure, ameliorate or control the progression of the targeted pathological condition.
  • a subject is successfully treated for a cancer if after administration of an oncolytic virus as described herein, alone or in combination, the subject shows an observable improvement of its clinical status.
  • administering refers to the delivery to a subject of a therapeutic agent such as the oncolytic virus described herein.
  • a therapeutic agent such as the oncolytic virus described herein.
  • proliferative disease encompasses any disease or condition resulting from uncontrolled cell growth and spread including cancers and some cardiovascular diseases (restenosis that results from the proliferation of the smooth muscle cells of the blood vessel wall, etc.).
  • cancer may be used interchangeably with any of the terms “tumour”, “malignancy”, “neoplasm”, etc. These terms are meant to include any type of tissue, organ or cell, any stage of malignancy (e.g. from a pre-lesion to stage IV).
  • disease associated with an increased osteoclast activity encompasses any disease or condition resulting in bone resorption or destruction (e.g. rheumatoid arthritis, osteoporosis, etc.).
  • subject generally refers to an organism for whom any product and method of the invention is needed or may be beneficial.
  • the organism is a mammal, particularly a mammal selected from the group consisting of domestic animals, farm animals, sport animals, and primates.
  • the subject is a human who has been diagnosed as having or at risk of having a proliferative disease such as a cancer.
  • subject and patients may be used interchangeably when referring to a human organism and encompasses male and female.
  • the subject to be treated may be a new-born, an infant, a young adult, an adult or an elderly.
  • combinatorial treatment may be used interchangeably and refer to a treatment of a subject with an oncolytic virus as described herein and at least an additional therapeutic modality.
  • the additional therapeutic modality may be selected from the group consisting of surgery, radiotherapy, chemotherapy, cryotherapy, hormonal therapy, toxin therapy, immunotherapy, cytokine therapy, targeted cancer therapy, gene therapy, photodynamic therapy, transplantation etc.
  • a combinatorial treatment may include a third or even further therapeutic modality.
  • optimal concentration of each component of the combination can be determined by the artisan skilled in the art.
  • the oncolytic virus of the invention e.g.
  • wild type oncolytic virus, or a modified derivative oncolytic virus may be delivered in combination with adenosine deaminase, wherein said metabolic immune modulator, or said adenosine deaminase is not encoded by the oncolytic virus.
  • Said modulator or adenosine deaminase may be in the form of a polypeptide (e.g. a recombinantly produced adenosine deaminase, or analogue thereof) or of a nucleic acid molecule (e.g.
  • adenosine deaminase(s) carried by a vector engineered for expressing such adenosine deaminase(s)), as well as mixture of polypeptide(s) and nucleic acid molecule(s) (e.g. adenosine deaminase(s) and expressing vector(s)).
  • the "oncolytic virus” of the present invention can be obtained from any member of virus identified at present time provided that it is oncolytic by a higher propensity to replicate in and kill dividing cells as compared to non-dividing cells. It may be a native virus that is naturally oncolytic or may be engineered by modifying one or more viral genes so-as to increase tumour selectivity and/or preferential replication in dividing cells, such as those involved in DNA replication, nucleic acid metabolism, host tropism, surface attachment, virulence, lysis and spread (see for example Kirn et al., 2001, Nat. Med. 7: 781; Wong et al., 2010, Viruses 2: 78-106). One may also envisage placing one or more viral gene(s) under the control of event or tissue-specific regulatory elements (e.g. promoter).
  • event or tissue-specific regulatory elements e.g. promoter
  • Exemplary oncolytic viruses include without limitation reovirus, Seneca Valley virus (SVV), vesicular stomatitis virus (VSV), Newcastle disease virus (NDV), herpes simplex virus (HSV), morbillivirus, adenovirus, poxvirus, retrovirus, measles virus, foamy virus, alpha virus, lentivirus, influenza virus, Sinbis virus, myxoma virus, rhabdovirus, picornavirus, coxsackievirus, parvovirus or the like.
  • SVV Seneca Valley virus
  • VSV vesicular stomatitis virus
  • NDV Newcastle disease virus
  • HSV herpes simplex virus
  • morbillivirus morbillivirus
  • adenovirus adenovirus
  • poxvirus poxvirus
  • retrovirus measles virus
  • foamy virus alpha virus
  • lentivirus influenza virus
  • Sinbis virus myxoma virus
  • rhabdovirus pi
  • the oncolytic virus of the present invention is obtained from a reovirus.
  • a representative example includes Reolysin (under development by Oncolytics Biotech; NCT01166542).
  • the oncolytic virus of the present invention is obtained from a Seneca Valley virus.
  • a representative example includes NTX-010 (Rudin et al., 2011, Clin. Cancer. Res. 17(4): 888-95).
  • the oncolytic virus of the present invention is obtained from a vesicular 5 stomatitis virus (VSV).
  • VSV vesicular 5 stomatitis virus
  • Representative examples are described in the literature (e.g. Stojdl et al., 2000, Nat. Med. 6(7): 821-5; Stojdl et al., 2003, Cancer Cell 4(4): 263-75).
  • the oncolytic virus of the present invention is obtained from a Newcastle disease virus.
  • Representative examples include without limitation the 73-T PV701 and HDV-HUJ strains as well as those described in the literature (e.g. Phuangsab et al., 2001, Cancer 10 Lett. 172(l):27-36; Lorence et al., 2007, Curr. Cancer Drug Targets 7(2):157-67; Freeman et al., 2006, Mol. Ther. 13(l):221-8).
  • the oncolytic virus of the present invention is obtained from a herpes virus.
  • the Herpesviridae are a large family of DNA viruses that all share a common structure and are composed of relatively large double-stranded, linear DNA genomes encoding 100-200 genes
  • the oncolytic herpes virus can be derived from different types of HSV, particularly preferred are HSV1 and HSV2.
  • the herpes virus may be genetically modified so-as to restrict viral replication in tumours or reduce its cytotoxicity in non-dividing cells.
  • any viral gene involved in nucleic acid metabolism may be inactivated, such as thymidine kinase (Martuza et al., 1991, Science
  • ribonucleotide reductase (RR) (Boviatsis et al., Gene Ther. 1: 323-31; Mineta et al., 1994, Cancer Res. 54: 3363-66), or uracil-N-glycosylase (Pyles et al., 1994, J. Virol. 68: 4963-72).
  • RR ribonucleotide reductase
  • uracil-N-glycosylase uracil-N-glycosylase
  • Another aspect involves viral mutants with defects in the function of genes encoding virulence factors such as the ICP34.5 gene (Chambers et al., 1995, Proc. Natl. Acad. Sci. USA 92: 1411-5).
  • Representative examples of oncolytic herpes virus include NV1020 (e.g. Geevarghese et al., 2010,
  • the oncolytic virus of the present invention is obtained from a morbillivirus which can be obtained from the paramyxoviridae family, with a specific preference for measles virus.
  • oncolytic measles viruses include without limitation MV- 30 Edm (McDonald et al., 2006; Breast Cancer Treat. 99(2): 177-84) and HMWMAA (Kaufmann et al., 2013, J. Invest. Dermatol. 133(4): 1034-42)
  • the oncolytic virus of the present invention is obtained from an adenovirus.
  • Methods are available in the art to engineer oncolytic adenoviruses.
  • An advantageous strategy includes the replacement of viral promoters with tumour-selective promoters or modifications of the El adenoviral gene product(s) to inactivate its/their binding function with p53 or retinoblastoma (Rb) protein that are altered in tumour cells.
  • the adenovirus ElB55kDa gene cooperates with another adenoviral product to inactivate p53 (p53 is frequently dysregulated in cancer cells), thus preventing apoptosis.
  • Representative examples of oncolytic adenovirus include ONYX-015 (e.g. Khuri et al., 2000, Nat. Med 6(8): 879-85) and H101 also named Oncorine (Xia et al., 2004, Ai Zheng 23(12): 1666-70).
  • the oncolytic virus of the present invention is a poxvirus.
  • poxvirus or "poxviral vector” refers to a virus belonging to the Poxviridae family, with a specific preference for a poxvirus belonging to the Chordopoxviridae subfamily and more preferably to the Orthopoxvirus genus.
  • Sequences of the genome of various poxviruses for example, the vaccinia virus, cowpox virus, canarypox virus, ectromelia virus, myxoma virus genomes are available in the art and specialized databases such as Genbank (accession number NC_006998, NC_003663 or AF482758.2, NC_005309, NC_004105, NC_001132 respectively).
  • the oncolytic poxvirus is an oncolytic cowpox virus, and can derive from any cowpox strain, like for example CPXV_GER1980_EP4 (Genbank HQ420895), CPXV_GER2002_MKY (Genbank HQ420898), CPXV_GER1991_3 (Genbank DQ 437593), CPXV_FRA2001_NANCY (Genbank HQ420894), CPXV_GER1990_2 (Genbank HQ420896), CPXV_UK2000_K2984 (Genbank HQ420900), CPXV_BR (Genbank AF482758.2 or NC 003663) and CPXV_NOR1994-MAN (Genbank HQ420899), CPXV_GER1998_2 (Genbank HQ420897), CPXV_gri (Genbank X94355), CPXV_FIN2000_MAN (Genbank HQ420893) and CPXV_A
  • the oncolytic poxvirus is an oncolytic vaccinia virus.
  • Vaccinia viruses are members of the poxvirus family characterized by a 200 kb double-stranded DNA genome that encodes numerous viral enzymes and factors that enable the virus to replicate independently from the host cell machinery.
  • the majority of vaccinia virus particles is intracellular (IMV for intracellular mature virion) with a single lipid envelop and remains in the cytosol of infected cells until lysis.
  • the other infectious form is a double enveloped particle (EEV for extracellular enveloped virion) that buds out from the infected cell without lysing it.
  • EEV extracellular enveloped virion
  • the oncolytic vaccinia virus of the present invention is modified by altering one or more viral gene(s).
  • Said modification(s) preferably lead(s) to the synthesis of a defective protein unable to ensure the activity of the protein produced under normal conditions by the unmodified gene (or lack of synthesis).
  • Exemplary modifications are disclosed in the literature with the goal of altering viral genes involved in DNA metabolism, host virulence, IFN pathway (e.g. Guse et al., 2011, Expert Opinion Biol. Ther.ll(5): 595-608) and the like.
  • Modifications for altering a viral locus encompass deletion, mutation and/or substitution of one or more nucleotide(s) (contiguous or not) within the viral gene or its regulatory elements. Modification(s) can be made by a number of ways known to those skilled in the art using conventional recombinant techniques.
  • the oncolytic virus of the present invention is modified by altering the thymidine kinase-encoding gene (locus J2R).
  • locus J2R thymidine kinase-encoding gene
  • the TK enzyme is involved in the synthesis of deoxyribonucleotides. TK is needed for viral replication in normal cells as these cells have generally low concentration of nucleotides whereas it is dispensable in dividing cells which contain high nucleotide concentration.
  • the oncolytic virus of the present invention is modified by altering at least one gene or both genes encoding ribonucleotide reductase (RR).
  • RR ribonucleotide reductase
  • this enzyme catalyses the reduction of ribonucleotides to deoxyribonucleotides that represents a crucial step in DNA biosynthesis.
  • the viral enzyme is similar in subunit structure to the mammalian enzyme, being composed of two heterologous subunits, designed Rl and R2 encoded respectively by the I4L and F4L locus.
  • VGF for VV growth factor
  • a representative example of suitable modification includes alteration of the VGF-encoding gene from the viral genome.
  • VGF for VV growth factor
  • A56R gene coding for hemagglutinin is a secreted protein which is expressed early after cell infection and its function seems important for virus spread in normal cells.
  • Another example is the alteration of the A56R gene coding for hemagglutinin, eventually in combination with TK deletion (Zhang et al., 2007, Cancer Res. 67:10038-46).
  • Alteration of interferon modulating gene(s) may also be advantageous (e.g. the B8R or B18R gene) or the caspase-1 inhibitor B13R gene.
  • Another suitable modification comprises the alteration of the F2L gene which encodes the viral dUTPase involved in both maintaining the fidelity of DNA replication and providing the precursor for the production of TMP by thymidylate synthase (Broyles et al., 1993, Virol. 195: 863-5). Sequence of the vaccinia virus F2L gene is available in Genbank via accession number M25392 and a poxvirus defective in F2L locus is available from WO2009/065547.
  • the oncolytic virus of this invention is a poxvirus defective for TK activity, resulting from alteration of the J2R locus.
  • the oncolytic virus of this invention is a poxvirus defective in both TK and RR activities resulting from alteration of both the J2R locus and at least one of the RR-encoding I4L and/or F4L locus carried by the viral genome (e.g. as described in WO2009/065546 and Foloppe et al., 2008, Gene Ther., 15: 1361-71).
  • the oncolytic virus of this invention is a poxvirus defective for dUTPase resulting from alteration of the F2L gene (e.g. as described in WO2009/065547), eventually in combination with alteration of at least one of TK and RR activities or both (resulting in a virus with alterations in the F2L; F2L and J2R gene; F2L and I4L; or F2L, J2R and I4L).
  • the oncolytic virus described herein comprises a nucleotide sequence encoding at least one agent targeting one or more metabolic immune modulator(s).
  • metabolic immune modulator(s) also designated as “metabolic immune checkpoint(s)” refers to any metabolite (e.g. amino acids, nucleotides, nucleosides, alcohols, vitamins, polyols, organic acids) that may directly or indirectly modulate the functioning of the immune system.
  • metabolic immune modulator(s) refers to any metabolite directly or indirectly involved in an immune pathway that under normal physiological conditions is crucial for preventing uncontrolled immune reactions and thus for the maintenance of self-tolerance and/or tissue protection.
  • the one or more metabolic immune modulator(s) described herein may independently act by blocking the activation and effectiveness of stimulatory immune cells (e.g.
  • NK cells macrophages, dendritic cells, Thl cells
  • inhibitory immune cells e.g. regulatory T-cells (T-regs)
  • T-regs regulatory T-cells
  • each of these reactions are regulated by counterbalancing stimulatory and inhibitory signals that in fine tune the response.
  • a preferred metabolic immune modulator in the context of the invention is a nucleoside, and more precisely, a purine nucleoside with a specific preference for adenosine.
  • adenosine is composed of a molecule of adenine attached to a ribose sugar molecule (ribofuranose) moiety via a ⁇ -Ng-glycosidic bond.
  • ribofuranose ribofuranose
  • the present invention also encompasses adenosine analogue such as 2'deoxyadenosine.
  • Agents targeting metabolic immune modulators have, in the context of the invention, the ability to modify the activity or the concentration of said one or more metabolic immune modulator(s).
  • said agents exert a degradation activity on the modulator(s).
  • the agent for use herein is an enzymatic polypeptide exerting a degradative action on adenosine.
  • a degradative action can result from any reaction which inhibits adenosine effects, and especially its action on the immune system (e.g. adenosine degradation, inactivation, capture, complexation, sequestration, etc.).
  • Suitable agents include, but are not limited to, adenosine deaminase, adenosine monophosphate deaminase, adenosine kinase, adenosine nucleosidase, etc. or any analogue thereof.
  • a particularly appropriate agent in the context of the invention, is an adenosine deaminase (ADA) polypeptide, or a polypeptide having an adenosine deaminase activity, encompassing naturally-occurring adenosine deaminase or analogues thereof.
  • an analogue refers to a polypeptide retaining a substantial adenosine deaminase enzymatic activity (at least 50% of the wild-type counterpart).
  • the term "analogue” encompasses fragment of ADA (e.g. a truncated ADA) as well as mutated ADA comprising one or more amino acid modification(s) preferably outside the catalytic site.
  • ADA analogues are ADA2-K374D and ADA2-E182T (Wang et al., 2006, Abstract 1472, AACR Annual Meeting).
  • analogues that retain a degree of sequence identity of at least 80%, advantageously at least 85%, preferably at least 90%, more preferably at least 95%, and even more preferably at least 98% identity with the sequence of the naturally occuring counterpart.
  • identity refers to an amino acid to amino acid, or nucleotide to nucleotide correspondence between two polypeptides or nucleic acid sequences. The percentage of identity between two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps which need to be introduced for optimal alignment and the length of each gap.
  • Adenosine deaminase - also termed “ADA”, “adenosine deaminase polypeptide” or “adenosine aminohydrolase” - is a polymorphic enzyme of the purine metabolism which catalyses the irreversible deamination of adenosine and deoxyadenosine to inosine and deoxyinosine, respectively (EC 3.5.4.4).
  • ADA is involved in the differentiation and maturation of the immune cells including lymphocytes and monocyte-macrophage cell lines.
  • Adenosine deaminase is present in mammalians and in a wide variety of microorganisms, plants, and invertebrates (Cristalli et al. 2001, Med Res Rev 21(2):105-28), and two different isoenzymes of ADA, designated as "ADAl” and "ADA2", were found in mammals and lower vertebrates (Zavialov et Engstrom, 2005, Biochem J. 391(Pt 1): 51-57). It is the case for example in chickens, which livers contain two kinetically distinct ADA isozymes with different molecular weights.
  • ADAl has a molecular mass of 35 kDa, an optimal pH of 7-7.5, a Km of 5.2xl0 "5 M, and a similar affinity for both adenosine and 2'-deoxyadenosine. It is present in all tissues, in erythrocytes and, in a lower amount, in plasma.
  • ADAl is also present in the cytosol and nucleus of lymphocytes and macrophages, and on the cell membrane as an ecto-form (ADAl complexed with a dimer of DPPIV or CD26). Two molecules of ADAl can also be connected via a combining protein, forming dimers of 280 kDa (Ungerer et al, 1992, Clin Chem 38/7, 1322-1326). Human ADA2 originates mainly from the monocyte-macrophage system. It is not ubiquitous but is more abundant than ADAl in serum. ADA2 has a molecular mass of 100 kDa, an optimum pH of 6.5, a Km of 200xl0 "5 M, and a weak affinity for 2'deoxyadenosine.
  • ADAl In humans, both ADAl and ADA2 have similar affinity for adenosine (Gakis et al., 1996, Eur Respir J, 9, 632-633) while ADAl also guarantees the downregulation of 2'deoxyadenosine.
  • adenosine deaminases can be used in the context of the present invention, including mammalian (e.g. human, bovine, guinea-pig, dog, cat, rabbit, rat, mouse), insects (e.g. mosquito, drosophila, phlebotomus), bacterial (e.g. E. coli) or lower eukaryotic ADA.
  • mammalian e.g. human, bovine, guinea-pig, dog, cat, rabbit, rat, mouse
  • insects e.g. mosquito, drosophila, phlebotomus
  • bacterial e.g. E. coli
  • lower eukaryotic ADA lower eukaryotic ADA.
  • the oncolytic virus of the present invention encodes or is used in combination with an ADA of human origin.
  • adenosine deaminases include, without limitation, human ADA1 (Genbank access. NO X02994), human ADA2 (Genbank access NO AF190746) and any human ADA analogues as defined above.
  • the oncolytic virus encodes or is used in combination with an ADA of insect origin.
  • insect adenosine deaminases include, but are not limited to, Culex quinquefasciatus ADA (Genbank access. NO. AF298886), Drosophila melanogaster ADA (Genbank access. NO. NM_141609.3) Phlebotomus duboscqi ADA (Genbank access. NO. DQ835383.1, and DQ835357.1) and Lutzomyia longipalpis ADA (Genbank access. NO. AF234182.1).
  • Adenosine deaminase can also originate from microorganisms, like fungi, yeast, virus or bacteria.
  • Examples of non-human adenosine deaminase originate from S. cerevisiae (GenBank access NO: Z46843), Candida glabrata (GenBank access NO: CR380956), E. coli (GenBank access NO: M59033), etc.
  • the oncolytic virus of the invention encodes the human adenosine deaminase (huADAl), wherein said huADAl comprises an amino acid sequence having at least 80%, preferably greater than 90%, and more preferably greater than 95% identity with SEQ ID NO: 1.
  • said oncolytic virus comprises a nucleic acid sequence having at least 80%, preferably greater than 90%, and more preferably greater than 95% identity with SEQ ID NO: 2.
  • the oncolytic virus of the invention encodes the human adenosine deaminase 2 (huADA2), wherein said huADA2 comprises an amino acid sequence having at least 80%, preferably greater than 90%, and more preferably greater than 95% identity with SEQ ID NO: 3.
  • said oncolytic virus comprises a nucleic acid sequence having at least 80%, preferably greater than 90%, and more preferably greater than 95% identity with SEQ ID NO: 4
  • the concentration of ADA expressed from the oncolytic virus of the present invention can be measured using high performance liquid chromatography or enzymatic or colorimetric techniques.
  • colorimetric technique is the measurement of the ammonia released from adenosine when broken down to inosine (NewLife BioChemEX LLC). For example, one may proceed by incubating a sample of infected host cells (e.g. cultured host cells or supernatant) with a buffered solution of adenosine, the ammonia is reacted with a Berthelot reagent to form a blue colour which is proportionate to the amount of enzyme activity.
  • the "adenosine deaminase encoding nucleic acid molecule” may be easily obtained by cloning, by PCR or by chemical synthesis based on the information provided herein and the general knowledge of the skilled person.
  • the ADA-encoding nucleic acid molecule for use herein may be native ADA-encoding sequences (e.g. cDNA) or analogue thereof derived from the latter by mutation, deletion, substitution and/or addition of one or more nucleotides.
  • the ADA- encoding nucleic acid molecule can be optimized for providing high level expression in a particular host cell or subject as described hereinafter.
  • the nucleic acid molecules encoding human adenosine deaminase can be inserted at any location of the viral genome with a specific preference for a non-essential locus and placed under the control of appropriate regulatory elements as described hereinafter.
  • the J2R locus, the I4L locus, the F4L locus or intergenic zones are particularly appropriate for insertion in oncolytic vaccinia virus.
  • the present invention relates to a vaccinia virus defective in viral TK and RR-encoded activities and encoding a human ADA, such as WTK-RR-/huADAl or VVTK-RR-/huADA2 illustrated in the Example section.
  • the present invention relates to a vaccinia virus defective in viral TK and RR-encoded activities and encoding a human ADA1, such as VVTK-RR-/huADAl.
  • the oncolytic virus of the invention may further comprise, inserted in its genome, one or more nucleic acid(s) of interest different from ADA- encoding nucleic acid molecule.
  • the nucleic acid(s) of interest can be homologous or heterologous to the host organism into which it is introduced. More specifically, it can originate from Prokaryotes (comprising the kingdoms of Bacteria, Archaea), Acaryotes (comprising the viruses) or Eukaryotes (comprising the kingdoms of Protista, Fungi, Plantae, Animalia).
  • said nucleic acid of interest encodes all or part of a polypeptide.
  • a polypeptide is understood to be any translational product of a polynucleotide regardless of size, and whether glycosylated or not, and includes peptides and proteins.
  • the nucleic acid of interest encodes a polypeptide of therapeutic or prophylactic interest which is capable of providing a biological activity when administered appropriately to a subject or which is expected to cause a beneficial effect on the course or a symptom of the pathological condition to be treated.
  • a vast number of nucleic acid of interest may be envisaged in the context of the invention such as those encoding polypeptides that can compensate for defective or deficient proteins in the subject, or those that act through toxic effects to limit or remove harmful cells from the body or those that encode immunity conferring polypeptides.
  • polypeptides of therapeutic interest include, without limitation, polypeptides capable of potentiating anti-tumor efficacy such as immunostimulatory polypeptides and antigens (for inducing or activating an immune humoral and/or cellular response), as well as polypeptides encoded by suicide genes which are capable of reinforcing the oncolytic nature of the virus of the present invention.
  • a specific embodiment of the invention is directed to an oncolytic virus further comprising an immunostimulatory polypeptide.
  • immunostimulatory polypeptide refers to a polypeptide, or protein, which has the ability to stimulate the immune system, in a specific or non-specific way. A vast number of proteins are known in the art for their ability to exert an immunostimulatory effect.
  • immunostimulatory proteins include, without limitation, immune checkpoint inhibitors, including, but not limited to anti-PDl, anti-PDLl, anti-PDL-2, anti-CTLA4, anti-Tim3, anti-LAG3, anti-BTLA; cytokines, like alpha, beta or gamma interferon, interleukins or tumour necrosis factor; agents that affect the regulation of cell surface receptors such as, e.g.
  • inhibitors of Epidermal Growth Factor Receptor in particular cetuximab, panitumumab, zalutumumab, nimotuzumab, matuzumab, gefitinib, erlotinib or lapatinib
  • inhibitors of Human Epidermal Growth Factor Receptor-2 in particular trastuzumab
  • agents that affect angiogenesis such as, e.g. inhibitor of Vascular Endothelial Growth Factor (in particular bevacizumab or ranibizumab)
  • agents that stimulates stem cells to produce granulocytes, macrophages such as, e.g. granulocyte macrophage - colony stimulating factor and B7 proteins.
  • antigen generally refers to a substance that is recognized and selectively bound by an antibody or by a T cell antigen receptor, in order to trigger an immune response. It is contemplated that the term antigen encompasses native antigen as well as fragment (e.g. epitopes, immunogenic domains, etc.) and analogue thereof, provided that such fragment or analogue is capable of being the target of an immune response.
  • Suitable antigens in the context of the invention are preferably polypeptides (e.g.
  • peptides, polypeptides, post translationally modified polypeptides, etc. including one or more B cell epitope(s) or one or more T cell epitope(s) or both B and T cell epitope(s) and capable of raising an immune response, preferably, a humoral or cell response that can be specific for that antigen.
  • the one or more antigen(s) is selected in connection with the disease to treat.
  • Preferred antigens for use herein are cancer antigens and antigens of tumour-inducing pathogens.
  • the antigen(s) contained in or encoded by the oncolytic virus is/are cancer antigen(s) (also called tumour-associated antigens) that is associated with and/or serve as markers for cancers.
  • Cancer antigens encompass various categories of polypeptides, e.g. those which are normally silent (i.e. not expressed) in normal cells, those that are expressed only at low levels or at certain stages of differentiation and those that are temporally expressed such as embryonic and foetal antigens as well as those resulting from mutation of cellular genes, such as oncogenes (e.g. activated ras oncogene), proto-oncogenes (e.g. ErbB family), or proteins resulting from chromosomal translocations.
  • oncogenes e.g. activated ras oncogene
  • proto-oncogenes e.g. ErbB family
  • the cancer antigens also encompass antigens encoded by pathogenic organisms (bacteria, viruses, parasites, fungi, viroids or prions) that are capable of inducing a malignant condition in a subject (especially chronically infected subject) such as RNA and DNA tumour viruses (e.g. HPV, HCV, EBV, etc.) and bacteria (e.g. Helicobacter pilori).
  • pathogenic organisms bacteria, viruses, parasites, fungi, viroids or prions
  • cancer antigens include, without limitation, MART-l/Melan- A, gplOO, Dipeptidyl peptidase IV (DPPIV), cyclophilin b, Colorectal associated antigen, Carcinoembryonic Antigen (CEA) , Prostate Specific Antigen (PSA) , prostate-specific membrane antigen (PSMA), T-cell receptor/CD3-zeta chain, MAGE-family of tumour antigens, GAGE-family of tumour antigens, BAGE, RAGE, LAGE-1, NAG, GnT-V, MUM-1, CDK4, tyrosinase, p53, MUC family (e.g.
  • antigens suitable for use in this invention are marker antigens (beta-galactosidase, luciferase, green fluorescent proteins, etc.).
  • the present invention also encompasses oncolytic viruses expressing two or more polypeptides of interest as described herein, e.g. at least two antigens, at least one antigen and one cytokine, at least two antigens and one cytokine, etc.
  • the oncolytic virus of the invention may further comprise at least a suicide gene.
  • suicide gene refers to a gene coding for a polypeptide able to convert a precursor of a drug, also named “prodrug”, into a cytotoxic compound. Examples of suicide genes suitable for use herein and corresponding prodrugs are disclosed in the following table: Suicide gene prodrug
  • the oncolytic virus of the invention carries in its genome a suicide gene encoding a polypeptide having at least cytosine deaminase (CDase) activity.
  • the oncolytic virus of the invention carries in its viral genome a suicide gene encoding a polypeptide having uracil phosphoribosyl transferase (UPRTase) activity.
  • CDase converts 5-fluorocytosine (5-FC), thereby forming cytotoxic 5-fluorouracil (5-FU), which is then converted into the even more toxic 5- fluoro-UMP (5-FUMP).
  • the suicide gene inserted in the viral genome of the oncolytic virus of the present invention encodes a polypeptide engineered by fusion of two enzymatic domains, one having the CDase activity and the second having the UPRTase activity.
  • Exemplary polypeptides include without limitation fusion polypeptides codA::upp, FCY1::FUR1 and FCYl::FURl[Delta] 105 (FCUl) and FCUl-8 described in W096/16183, EP998568 and WO2005/07857.
  • FCUl suicide gene (or FCYl::FURl[Delta] 105 fusion) encoding a polypeptide comprising the amino acid sequence represented in the sequence identifier SEQ ID NO: 1 of WO2009/065546.
  • nucleic acids of interest include, but not limited to: - Nucleoside pool modulators (e.g.: cytidine deaminase) apoptotic genes, including pro-apoptotic genes inhibitors of pro-apoptotic genes anti- apoptotic genes (and inhibitors of anti-apoptotic genes,
  • Nucleoside pool modulators e.g.: cytidine deaminase
  • apoptotic genes including pro-apoptotic genes inhibitors of pro-apoptotic genes anti- apoptotic genes (and inhibitors of anti-apoptotic genes,
  • nucleic acid coding for endonuclease like restriction enzymes, C ISPR/Cas9;
  • RNA including but not limited to target-specific miRNA, shRNA, siRNA.
  • the nucleic acid(s) of interest sequences may be easily obtained by cloning, by PCR or by chemical synthesis using conventional techniques. They may be native nucleic acid(s) sequences (e.g. cDNA) or sequences derived from the latter by mutation, deletion, substitution and/or addition of one or more nucleotides. Moreover, their sequences are described in the literature which can be consulted by persons skilled in the art.
  • the nucleic acids sequences can be inserted at any location of the viral genome, with a specific preference for a non-essential locus, at the same location of the ADA- encoding nucleic acid (e.g. within J2R locus) or at another location (within I4L locus).
  • nucleic acid(s) sequences The ADA nucleic acid sequence and, if any, nucleic acid(s) of interest can be independently optimized for providing high level expression in a particular host cell or subject. It has been indeed observed that, the codon usage patterns of organisms are highly non-random, and the use of codons may be markedly different between different hosts. As such nucleic acid(s) might be from bacterial or lower eukaryote origin, they may have an inappropriate codon usage pattern for efficient expression in higher eukaryotic cells (e.g. human). Typically, codon optimization is performed by replacing one or more "native" (e.g.
  • expression in the host cell or subject can further be improved through additional modifications of the nucleic acid sequence.
  • negative sequence elements include without limitation the regions having very high (>80%) or very low ( ⁇ 30%) GC content; AT -rich or GC-rich sequence stretches; unstable direct or inverted repeat sequences; and/or internal cryptic regulatory elements such as internal TATA-boxes, chi-sites, ribosome entry sites, and/or splicing donor/acceptor sites.
  • the oncolytic virus comprises the elements necessary for the expression of the ADA-encoding nucleic acid molecule(s) and/or the nucleic acid(s) of interest in a host cell subject.
  • nucleic acid(s) is/are operably linked to suitable regulatory elements that allow, contribute or modulate expression in a given host cell or subject, including replication, duplication, transcription, splicing, translation, stability and/or transport of the nucleic acid(s) or its derivative (i.e. mRNA).
  • "operably linked” means that the elements being linked are arranged so that they function in concert for their intended purposes.
  • a promoter is operably linked to a nucleic acid molecule if the promoter effects transcription from the transcription initiation to the terminator of said nucleic acid molecule.
  • the choice of the regulatory sequences can depend on such factors as the nucleic acid molecule itself, the virus into which it is inserted, the host cell or subject, the level of expression desired, etc.
  • the promoter is of special importance. In the context of the invention, it can be constitutive directing expression of the encoded product (e.g. ADA and/or polypeptide(s) of interest) in many types of host cells or specific to certain host cells (e.g. liver-specific regulatory sequences) or regulated in response to specific events or exogenous factors (e.g. by temperature, nutrient additive, hormone, etc.) or according to the phase of a viral cycle (e.g. late or early).
  • CMV cytomegalovirus
  • RSV cytomegalovirus
  • PGK phosphoglycero kinase
  • TK thymidine kinase promoter of herpes simplex virus
  • T7 polymerase promoter WO98/10088
  • Vaccinia virus promoters are particularly adapted for expression in oncolytic poxviruses.
  • Representative examples include without limitation the vaccinia 7.5K, H5R, 11K7.5 (Erbs et al., 2008, Cancer Gene Ther. 15(1): 18-28), TK, p28, pll Prl3.5 (WO2014/063832), pB8R, pFUL, pA44L, pCllR (WO2011/128704) and KIL promoter, as well as synthetic promoters such as those described in Chakrabarti et al. (1997, Biotechniques, 23: 1094-7; Hammond et al., 1997, J. Virol.
  • the ADA nucleic acid molecule is inserted in the TK locus of the oncolytic virus of the invention and placed under the control of the vaccinia pllK7.5 promoter.
  • the ADA nucleic acid molecule is inserted in the TK locus of the oncolytic virus of the invention and placed under the control of the vaccinia p7.5K or pH5R promoter.
  • the regulatory elements controlling the nucleic acid expression may further comprise additional elements for proper initiation, regulation and/or termination of transcription (e.g. a transcription termination sequences), mRNA transport (e.g. nuclear localization signal sequences), processing (e.g. splicing signals), and stability (e.g. introns and non-coding 5' and 3' sequences), translation (e.g. an initiator Met, tripartite leader sequences, IRES ribosome binding sites, signal peptides, etc.), targeting sequences, transport sequences, secretion signal, and sequences involved in replication or integration. Said sequences have been reported in the literature and can be readily obtained by those skilled in the art.
  • the invention also relates to a process for preparing an oncolytic virus of the invention, in which process:
  • an oncolytic virus of the invention is introduced into a producer cell
  • said producer cell is cultured under conditions which are appropriate for enabling said oncolytic virus to be produced
  • said oncolytic virus is recovered from the cell culture.
  • the oncolytic virus of the present invention is produced into a suitable host cell line using conventional techniques including culturing the transfected or infected host cell under suitable conditions so-as to allow the production of infectious viral particles and recovering the produced infectious viral particles from the culture of said cell and optionally purifying said recovered infectious viral particles.
  • Suitable host cells for production of the oncolytic virus include without limitation human cell lines such as HeLa (ATCC), 293 cells (Graham et al., 1997, J. Gen. Virol. 36: 59-72), HER96, PER-C6 (Fallaux et al., 1998, Human Gene Ther.
  • Monkey cells such as Vero (ATCC CCL-081), CV1(ATCC CCL-70) and BSC1(ATCC CCL-26) cell lines, avian cells such as those described in WO2005/042728, WO2006/108846, WO2008/129058, WO2010/130756, WO2012/001075, etc.), hamster cell lines such as BHK-21 (ATCC CCL-10) as well as primary chicken embryo fibroblasts (CEF) prepared from chicken embryos obtained from fertilized eggs.
  • Host cells are preferably cultivated in a medium free of animal-or human-derived products, using a chemically defined medium with no product of animal or human origin.
  • Culturing is carried out at a temperature, pH and oxygen content appropriate for the producer cell. Such culturing conditions are within the expertise of one of ordinary skill in the art. If growth factors are present, they are preferably recombinantly produced and not purified from animal material. Suitable animal-free medium media are commercially available, for example VP-SFM medium (Invitrogen) for culturing CEF producer cells. Producer cells are preferably cultivated at a temperature comprised between +30°C and +38°C (more preferably at about +37°C) for between 1 and 8 days (preferably for 1 to 5 days for CEF and 2 to 7 days for immortalized cells) before infection. If needed, several passages of 1 to 8 days may be made in order to increase the total number of cells.
  • VP-SFM medium Invitrogen
  • Producer cells are infected by the oncolytic virus with an appropriate multiplicity of infection (MOI), which can be as low as 0.001 (more preferably between 0.05 and 5) to permit productive infection.
  • MOI multiplicity of infection
  • infected producer cells are then cultured under appropriate conditions well known to those skilled in the art until progeny viral vector is produced.
  • Culture of infected producer cells is also preferably performed in a chemically defined medium (which may be the same as or different from the medium used for culture of producer cells and/or for infection step) free of animal- or human-derived products at a temperature between +30°C and +37°C, for 1 to 5 days.
  • the viral particles may be collected from the culture supernatant and/or the producer cells.
  • Recovery from producer cells may require a step allowing the disruption of the producer cell membrane to allow the liberation of the virus from producer cells.
  • the disruption of the producer cell membrane can be induced by various techniques well known to those skilled in the art, including but not limited to, freeze/thaw, hypotonic lysis, sonication, microfluidization, or high-speed homogenization.
  • the recovered oncolytic virus can be at least partially purified before being used according to the present invention.
  • Various purification steps can be envisaged, including clarification, enzymatic treatment (e.g. endonuclease such as benzonase, protease), ultracentrifugation (e.g. sucrose gradient or caesium chloride gradient), chromatographic and filtration steps.
  • enzymatic treatment e.g. endonuclease such as benzonase, protease
  • ultracentrifugation e.g. sucrose gradient or caesium chloride gradient
  • chromatographic and filtration steps e.g. WO2007/147528; WO2008/138533, WO2009/100521, WO2010/130753, WO2013/022764.
  • Oncolytic virus composition e.g. WO2007/147528; WO2008/138533, WO2009/100521, WO2010/130753, WO2013/022764
  • the invention also relates to a composition which comprises a therapeutically effective amount of the oncolytic virus of the present invention or prepared according to the process described herein.
  • the composition further comprises a pharmaceutically acceptable vehicle.
  • a “therapeutically effective amount” corresponds to the amount of each of the active agents comprised in the composition of the invention that is sufficient for producing one or more beneficial results.
  • a therapeutically effective amount may vary as a function of various parameters, e.g. the mode of administration; the disease state; the age and weight of the subject; the ability of the subject to respond to the treatment; kind of concurrent treatment; the frequency of treatment; and/or the need for prevention or therapy.
  • the composition of the invention is administered at a dose sufficient to prevent or to delay the onset and/or establishment and/or relapse of a pathologic condition (e.g. a proliferative disease such as cancer), especially in a subject at risk.
  • a pathologic condition e.g. a proliferative disease such as cancer
  • the composition of the invention is administered to a subject diagnosed as having a pathological condition (e.g. a proliferative disease such as cancer) with the goal of treating the disease, eventually in association with one or more conventional therapeutic modalities.
  • a therapeutically effective amount could be that amount necessary to cause an observable improvement of the clinical status over the baseline status or over the expected status if not treated, as described hereinafter.
  • An improvement of the clinical status can be easily assessed by any relevant clinical measurement typically used by physicians and skilled healthcare staff. For example, techniques routinely used in laboratories (e.g. flow cytometry, histology, imaging techniques, etc.) may be used to perform tumour surveillance.
  • a therapeutically effective amount could also be the amount necessary to cause the development of an effective non-specific (innate) and/or specific anti-tumour response.
  • development of an immune response in particular a T cell response, can be evaluated in vitro, in suitable animal models or using biological samples collected from the subject.
  • the appropriate dosage of oncolytic virus can be adapted as a function of various parameters and may be routinely determined by a practitioner in the light of the relevant circumstances.
  • individual doses for the oncolytic virus may vary within a range extending from approximately 10 3 to approximately 10 12 vp (viral particles), iu (infectious unit) or PFU (plaque- forming units) depending on the virus and the quantitative technique used.
  • a suitable dose of oncolytic vaccinia virus for human use is comprised between approximately 10 4 to approximately 10 11 PFU, preferably between approximately 10 5 PFU to approximately 10 10 PFU; doses of approximately 10 6 PFU to approximately 5xl0 9 PFU being particularly preferred (e.g.
  • the quantity of virus present in a sample can be determined by routine titration techniques, e.g. by counting the number of plaques following infection of permissive cells (e.g. BHK-21 or CEF), immunostaining (e.g. using anti-virus antibodies; Caroll et al., 1997, Virology 238: 198-211), by measuring the A260 absorbance (vp titers), or still by quantitative immunofluorescence (iu titers).
  • permissive cells e.g. BHK-21 or CEF
  • immunostaining e.g. using anti-virus antibodies; Caroll et al., 1997, Virology 238: 198-211
  • A260 absorbance vp titers
  • iu titers quantitative immunofluorescence
  • pharmaceutically acceptable vehicle is intended to include any and all carriers, solvents, diluents, excipients, adjuvants, dispersion media, coatings, antibacterial and antifungal agents, absorption agents and the like compatible with administration in mammals and in particular human subjects.
  • the oncolytic virus of the invention can independently be placed in a solvent or diluent appropriate for human or animal use.
  • the solvent or diluent is preferably isotonic, hypotonic or weakly hypertonic and has a relatively low ionic strength.
  • Representative examples include sterile water, physiological saline (e.g. sodium chloride), Ringer's solution, glucose, trehalose or saccharose solutions, Hank's solution, and other aqueous physiologically balanced salt solutions (see for example the most current edition of Remington: The Science and Practice of Pharmacy, A. Gennaro, Lippincott, Williams&Wilkins).
  • oncolytic viruses are suitably buffered for human use.
  • Suitable buffers include without limitation phosphate buffer (e.g. PBS), bicarbonate buffer and/or Tris buffer capable of maintaining a physiological or slightly basic pH (e.g. from approximately pH 7 to approximately pH 9).
  • composition of the invention may also contain other pharmaceutically acceptable excipients for providing desirable pharmaceutical or pharmacodynamic properties, including for example osmolarity, viscosity, clarity, colour, sterility, stability, rate of dissolution of the formulation, modifying or maintaining release or absorption into a human or animal subject, promoting transport across the blood barrier or penetration in a particular organ.
  • composition of the invention may be adjuvanted to further enhance immunity (especially a T cell-mediated immunity) or facilitate infection of tumour cells upon administration.
  • Suitable adjuvants include, without limitation, alum, mineral oil emulsion such as, Freunds complete and incomplete (IFA), lipopolysaccharide or a derivative thereof (Ribi et al., 1986, Plenum Publ. Corp.,407-419), saponins such as QS21 (Sumino et al., 1998, J.Virol. 72: 4931; W098/56415), imidazo-quinoline compounds such as Imiquimod (Suader, 2000, J. Am Acad Dermatol. 43: S6), S-27609 (Smorlesi, 2005, Gene Ther.
  • alum mineral oil emulsion
  • IFA Freunds complete and incomplete
  • lipopolysaccharide or a derivative thereof Ribi et al., 1986, Plenum Publ. Corp.,407-419
  • saponins such as QS21 (Sumino et al., 1998, J.Virol. 72: 4931;
  • the composition of the invention may be formulated with the goal of improving its stability, in particular under the conditions of manufacture and long-term storage (i.e. for at least 6 months, with a preference for at least two years) at freezing (e.g. -70°C, -20°C), refrigerated (e.g. 4°C) or ambient temperatures.
  • freezing e.g. -70°C, -20°C
  • refrigerated e.g. 4°C
  • ambient temperatures e.g. Various virus formulations are available in the art either in frozen, liquid form or lyophilized form (e.g. WO98/02522, WO01/66137, WO03/053463, WO2007/056847 and WO2008/114021, etc.).
  • Solid (e.g. dry powdered or lyophilized) compositions can be obtained by a process involving vacuum drying and freeze-drying.
  • buffered formulations including NaCI and/or sugar are particularly adapted to the preservation of viruses (e.g. Tris 10 mM pH 8 with sucrose 5 % (W/V), Sodium glutamate 10 mM, and NaCI 50 mM or phosphate-buffered saline with glycerol (10%) and NaCI).
  • viruses e.g. Tris 10 mM pH 8 with sucrose 5 % (W/V), Sodium glutamate 10 mM, and NaCI 50 mM or phosphate-buffered saline with glycerol (10%) and NaCI.
  • the oncolytic virus composition is preferably formulated in a way adapted to the mode of administration to ensure proper distribution and release in vivo.
  • gastro-resistant capsules and granules are particularly appropriate for oral administration, suppositories for rectal or vaginal administration, eventually in combination with absorption enhancers useful to increase the pore size of the mucosal membranes.
  • absorption enhancers are typically substances having structural similarities to the phospholipid domains of the mucosal membranes (such as sodium deoxycholate, sodium glycocholate, dimethyl-beta-cyclodextrin, lauryl-l-lysophosphatidylcholine).
  • Another and particularly appropriate example is a formulation adapted to the administration through microneedle means (e.g. transcutaneous or intradermal patches).
  • Such a formulation may comprise resuspension of the immunotherapeutic product in endotoxin-free phosphate-buffered saline (PBS).
  • PBS endotoxin-
  • the oncolytic virus, or the composition of the invention may be administered in a single dose or multiple doses. If multiples doses are contemplated, administrations may be performed by the same or different routes and may take place at the same site or at alternative sites. Intervals between each administration can be from several hours to 8 weeks (e.g. 24h, 48h, 72h, weekly, every two or three weeks, monthly, etc.). Intervals can also be irregular. It is also possible to proceed via sequential cycles of administrations that are repeated after a rest period (e.g. cycles of 3 to 6 weekly administrations followed by a rest period of 3 to 6 weeks). The dose can vary for each administration within the range described above.
  • Parenteral routes are intended for administration as an injection or infusion and encompass systemic as well as local routes.
  • Common parenteral injection types are intravenous (into a vein), intra-arterial (into an artery), intradermal (into the dermis), subcutaneous (under the skin), intramuscular (into muscle) and intratumoural (into a tumour or at its proximity). Infusions typically are given by intravenous route.
  • Topical administration can be performed using transdermal means (e.g. patch and the like).
  • Mucosal administrations include without limitation oral/alimentary, intranasal, intratracheal, intrapulmonary, intravaginal or intra-rectal route.
  • intranasal, intrapulmonary and intratracheal routes it is advantageous for administration to take place by means of an aerosol or by means of instillation.
  • Preferred routes of administration for the oncolytic virus of the invention include intravenous and intratumoral routes.
  • Administrations may use conventional syringes and needles (e.g. Quadrafuse injection needles) or any compound or device available in the art capable of facilitating or improving delivery of the active agent(s) in the subject.
  • Transdermal systems are also appropriate, e.g. using solid, hollow, coated or dissolvable microneedles (e.g. Van der Maaden et al., 2012, J. Control release 161: 645-55) and preferred are silicon and sucrose microneedle patches (see, e.g., Carrey et al., 2014, Sci Rep 4: 6154 doi 10.1038; and Carrey et al., 2011, PLoS ONE, 6(7) e22442).
  • solid, hollow, coated or dissolvable microneedles e.g. Van der Maaden et al., 2012, J. Control release 161: 645-55
  • silicon and sucrose microneedle patches see, e.g., Carrey et al.,
  • a particularly preferred composition comprises 10 6 PFU to 5xl0 9 PFU of an oncolytic virus comprising a ADA-encoding nucleic acid molecule and preferably an oncolytic vaccinia virus defective in J2R locus (TK-), in I4L and/or F4L locus (RR-) or both in J2R locus (TK-) and in I4L/ F4L locus (TK- RR-), with a specific preference for a vaccinia having the ADA nucleic acid molecule inserted in place of the TK locus and placed under the pllK7.5 promoter such as VVTK-RR-/ADA1, VVTK-RR-/ADA2 or VVTK-RR-/cuADA described herein; formulated for intravenous or intratumoral administration.
  • the present invention provides an oncolytic virus or a composition thereof (in particular a pharmaceutical composition) for use for treating or preventing a disease or a pathologic condition in a subject in need thereof.
  • the present invention also relates to the use of an oncolytic virus or composition thereof for the manufacture of a medicament for treating or preventing a disease or a pathologic condition in a subject in need thereof.
  • the present invention also relates to a method of treatment comprising administering the oncolytic virus or the composition thereof in an amount sufficient for treating or preventing a disease or a pathologic condition in a subject in need thereof.
  • the present invention also relates to the use of an oncolytic virus or composition thereof for treating or preventing a disease or a pathologic condition in a subject in need thereof.
  • a "disease” (and any form of disease such as "disorder” or "pathological condition”) is typically characterized by identifiable symptoms.
  • diseases that may be prevented or treated using the oncolytic virus of the invention or the composition thereof include proliferative diseases such as cancers, tumours or restenosis and diseases associated to an increased osteoclast activity such as rheumatoid arthritis and osteoporosis.
  • the present invention is particularly suited for treating or preventing cancers, and particularly Adrenocortical Carcinoma, Adrenal Cortex Cancer, Anal Cancer, Gastrointestinal Carcinoid Tumors (for example Appendix Cancer and Carcinoid Tumor), Bile Duct Cancer (for example Cholangiocarcinoma), Bladder Cancer, Bone Cancer (for example Ewing Sarcoma, Malignant Fibrous Histiocytoma of Bone and Osteosarcoma), Brain Tumors (for example Astrocytomas, Embryonal Tumors, Germ Cell Tumors, Central Nervous System Atypical Teratoid/Rhabdoid Tumor, Craniopharyngioma, Ependymoma, Gliomas and Glioblastoma), Breast Cancer (for example Ductal Carcinoma In Situ), Bronchial Tumors, Carcinoma of Unknown Primary, Cardiac (Heart) Tumors, Cervical Cancer, Chordoma, Chronic
  • the oncolytic virus or a composition of the invention is used for treating gastro-intestinal cancers, comprising cancers of oesophagus, gallbladder, liver, pancreas, stomach, small intestine, bowel (large intestine or colon and rectum), and anus.
  • a particularly preferred method comprises 1 to 6 intravenous or intratumoral administrations of the oncolytic virus of the invention or composition thereof given at weekly to monthly intervals with a specific preference for 3 bi-weekly administrations (e.g.
  • composition comprising 10 6 to 5xl0 9 PFU of an oncolytic vaccinia virus comprising inserted in its genome a ADA-encoding nucleic acid molecule (e.g. placed under the pllK7.5 promoter), preferably defective in J2R locus (TK-), in I4L and/or F4L locus (RR-) or both in J2R locus (TK-) and in I4L/ F4L locus (TK- RR-), such as VVTK-RR-/huADAl or VVTK-RR-/huADA2.
  • the beneficial effects provided by the methods of the present invention can be evidenced by an observable improvement of the clinical status over the baseline status or over the expected status if not treated according to the modalities described herein.
  • An improvement of the clinical status can be easily assessed by any relevant clinical measurement typically used by physicians and skilled healthcare staff.
  • the therapeutic benefit can be transient (for one or a couple of months after cessation of administration) or sustained (for several months or years).
  • the natural course of clinical status which may vary considerably from a subject to another, it is not required that the therapeutic benefit be observed in each subject treated but in a significant number of subjects (e.g.
  • such methods can be correlated with one or more of the followings: inhibiting or slowing tumour growth, proliferation and metastasis, preventing or delaying tumour invasion (spread of tumour cells in neighbouring tissues), reducing the tumour number; reducing the tumour size, reducing the number or extent of metastases, providing a prolonged overall survival rate (OS), increasing progression free survival (PFS), increasing the length of remission, stabilizing (i.e. not worsening) the state of disease, providing a better response to the standard treatment, improving quality of life and/or inducing an anti-tumour response (e.g. nonspecific (innate) and/or specific such as a cytotoxic T cell response) in the subject treated in accordance with the present invention.
  • an anti-tumour response e.g. nonspecific (innate) and/or specific such as a cytotoxic T cell response
  • the present invention also relates to a method for treating a disease or a pathologic condition in a subject in need thereof comprising administering the oncolytic virus or the composition of the present invention or prepared according to the process described herein.
  • said disease is a proliferative disease such as cancers, tumours and restenosis.
  • said disease is a disease associated with an increased osteoclast activity like rheumatoid arthritis and osteoporosis.
  • the present invention relates to a method for inhibiting tumour cell growth in vivo comprising administering an oncolytic virus or a composition thereof in a subject in need thereof so-as to inhibit the growth of a tumour.
  • inhibition of tumour cell growth can be evaluated routinely, for example by radiography means.
  • the administration(s) of the oncolytic virus or a composition thereof desirably result(s) in at least a 10% decrease of the tumour mass.
  • the oncolytic virus or composition can be administered in association with any conventional therapeutic modalities which are available for treating or preventing the targeted disease or pathological condition.
  • conventional therapies include, without limitation, surgery, radiotherapy, chemotherapy, cryotherapy, hormonal therapy, toxin therapy, immunotherapy, cytokine therapy, transplantation (e.g. stem cell), hyperthermia and photodynamic therapy.
  • Such conventional therapy/ies is/are administered to the subject in accordance with standard practice before, after, essentially simultaneously or in an interspersed manner with the oncolytic virus or composition thereof.
  • the oncolytic virus, composition or method according to the invention can be used in association with radiotherapy.
  • radiotherapy Those skilled in the art can readily formulate appropriate radiation therapy protocols and parameters (see for example Perez and Brady, 1992, Principles and Practice of Radiation Oncology, 2nd Ed. JB Lippincott Co; using appropriate adaptations and modifications as will be readily apparent to those skilled in the field).
  • the types of radiation that may be used are well known in the art and include electron beams, high-energy photons from a linear accelerator or from radioactive sources such as cobalt or caesium, protons, and neutrons.
  • Dosage ranges for radio-isotopes can be defined by the practitioners depending on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells.
  • Regular X-rays doses for prolonged periods of time (3 to 6 weeks), or high single doses are contemplated by the present invention.
  • the methods may be used in conjunction with surgery.
  • the oncolytic virus or composition thereof may be administered upon excision of the tumour (e.g. by local application within the excised zone for example).
  • the oncolytic virus or composition thereof may be used in combination with one or more substances effective in anticancer therapy, like chemotherapeutic drugs or immunotherapeutic products.
  • the oncolytic virus or composition of the invention may be used in conjunction with chemotherapeutic drugs currently used for treating cancer.
  • any anticancer chemotherapy drug may be used in combination with the oncolytic virus of the present invention or the composition thereof, there may be mentioned more specifically alkylating agents, topoisomerase I inhibitors, topoisomerase II inhibitors, platinum derivatives, inhibitors of tyrosine kinase receptors, antimetabolites and antimitotic agents.
  • the oncolytic virus or composition of the invention may be used in conjunction with immunotherapy, and especially with anti-neoplastic antibodies as well as siRNA and antisense polynucleotides.
  • anti-neoplastic antibodies as well as siRNA and antisense polynucleotides.
  • Representative examples include, among others, monoclonal antibodies blocking immune checkpoint (e.g.
  • Monoclonal antibodies blocking Epidermal Growth Factor Receptor in particular cetuximab, panitumumab, zalutumumab, nimotuzumab, matuzumab, trastuzumab (HerceptinTM), gefitinib, eriotinib, lapatinib, etc.
  • monoclonal antibodies blocking Vascular Endothelial Growth Factor in particular bevacizumab and ranibizumab.
  • Other representative examples of such immunotherapeutic products suitable for use in the present invention are plasmid DNA vector, vaccinia virus (e.g.
  • Another aspect of the invention concerns the combination of an oncolytic virus (e.g. wild type oncolytic virus, or modified derivative oncolytic virus) with at least one agent degrading a metabolic immune modulator (e.g.
  • said degrading agent is a polypeptide, more specifically an ADA polypeptide, or a polypeptide having an adenosine deaminase activity.
  • ADA may be encoded by another vector, like for example DNA molecules (plasmids, non-oncolytic viral vectors, cosmids and artificial chromosomes), RNA molecules, nanoparticles, etc.
  • Adenosine deaminase may also be in its polypeptide form.
  • the polypeptide can be of any origin, e.g. human, humanized, animal, insects, microorganisms or chimeric.
  • the polypeptide may be glycosylated or non- glycosylated.
  • the oncolytic virus or composition thereof may also be used in combination with substances which enhance adenosine deaminase activity.
  • the course of treatment may be routinely determined by a practitioner and various protocols are encompassed by the present invention.
  • 1 to 10 administrations of the oncolytic virus e.g. 3 to 6 biweekly injections
  • All vaccinia viruses used in this study are of the Western Reserve strain or Copenhagen strain with disrupted TK and RR genes for enhanced cancer cell specificity.
  • Fragments containing huADAl, huADA2 and cuADA were generated synthetically (Geneart, Germany) and cloned in in the vaccinia transfer plasmid pTG18495 restricted by Pst ⁇ and fcoRI to give pTG19009, pTG19021 and pTG19010 respectively.
  • the vaccinia transfer plasmid pTG18495, is designed to permit insertion of the nucleotide sequence to be transferred by homologous recombination in TK gene of the VV genome. It contains the flanking sequences (BRGTK and BRDTK) surrounding the J2R gene and the pllK7.5 promoter followed by multiple cloning sites.
  • amino acid sequences of the huADAl, huADA2 and cuADA are given in SEQ ID NO: 1,
  • SEQ ID NO: 3 and SEQ ID NO: 5 respectively.
  • the nucleic acid sequence of the huADAl, huADA2 and cuADA are given in SEQ ID NO: 2, SEQ ID NO: 4 and SEQ ID NO: 6.
  • Generation of the WR vaccinia viruses was performed by homologous recombination in primary chicken embryos fibroblasts (CEF) infected with a RR-deleted WR and transfected by nucleofection with pTG19009, pTG19010 or pTG19021 (according to Amaxa Nucleofector technology).
  • Viral selection was performed by plaque purification after growth in Thymidine kinase- 5 deficient (TK ⁇ ) 143B cells cultured in the presence of bromodeoxyuridine. This selection allows only TK ⁇ recombinant WR to remain viable. Absence of contamination by parental WR was verified by PCR.
  • TK ⁇ Thymidine kinase- 5 deficient
  • Culture supernatants were collected after 24, 48 or 72 hours of infection at 37°C.
  • the absorbance (Abs) at 265 nm was read at 60 s intervals using an Infinite MIOOOPro microplate reader thermostated to 30 °C (Tecan, Switzerland), after orbital mixing.
  • WRTG19009 and WRTG19021 encoded huADAl and huADA2 were well secreted by MCA205 (Figure 2A) and CT26 ( Figure 2B) and active in the culture supernatant.
  • the enzymatic activity was accumulating in the culture medium during the extended culture which showed a certain level of stability of the virus encoded human ADAs.
  • the lower permissivity of 30 the CT26 cell line resulted in overall lower level of active enzymes produced by the infected cells.
  • the oncolytic activity of the different viruses WR expressing huADAl, huADA2 and cuADA was evaluated by cell viability measurements in both MCA205 or CT26 infected cells.
  • a total of 5xl0 4 cells per well were plated in six-well culture dishes in 2 mL of culture medium and infected with either WRTG18011, WRTG19009, WRTG19010 or WRTG19021 at a MOI of either 0.1, 0.01, 0.001 or 0.0001.
  • TK-RR-deleted WR vaccinia virus without transgene and TK-RR deleted WR virus expressing the two human enzymes showed similar cytotoxicity in tumor cells, whereas in contrary, the expression of cuADA did not affect the oncolytic activity of the vaccinia virus towards the cultured tumor cells.
  • ADAs expression on viral replication was assessed in cultured cells.
  • 2xl0 5 cells per well of MCA205 and CT26 cells were plated in six-well culture dishes in 2 mL of culture medium and infected with WRTG18011, WRTG19009, WRTG19010 or WRTG19021 at a MOI of 0.01 and 0.001.
  • Cells and medium were harvested after 24, 48 or 72 hours after infection and frozen at -80°C until use.
  • Cell suspensions were thawed and sonicated and treated with benzonase (Novagen) to eliminate non-encapsidated DNA.
  • the virus capsids were disrupted by addition of an equal volume of Tris 20 mM, EDTA 10 mM, SDS 1 % pH7.4 followed by 160 ⁇ g of Proteinase K (Qiagen) and an incubation of 30 min at 65°C. After inactivation of Proteinase K, the MVA genome DNA was quantified using a q-PCR. The absolute quantification was allowed using a series of dilutions from 10 6 to 10 2 copies of purified MVA genome.
  • a set of oligonucleotides primers and probe that hybridize into the gene encoding for the secreted chemokine-binding protein (SCBP) of MVA were used (forward primer 5'- CGATGATGGAGTAATAAGTGGTAGGA-3' (SEQ ID NO: 7); reverse primer 5'- CACCGACCGATGATAAGATTTG-3' (SEQ ID NO: 8); probe 5' FAM - ACTG ATTCC ACCTCG G G - 3' (SEQ ID NO:9; MGB / NFQ).
  • the Quantitect Multiplex PCR kit from Qiagen was used to perform the q-PCR reactions on a 7300 real-time system from Applied Biosystem.
  • mice As shown in Figure 6, the survival of mice is also increased by ADA-expression. More specifically, all mice treated with the vehicle were dead at 32 days (0% survival rate), whereas at this time, 40, 60 and 80 % survival rate were obtained respectively in empty virus control group, huADAl and huADA2 expressing viruses treated group.
  • vaccinia viruses Copenhagen strain (COP) expressing adenosine deaminase was performed by homologous recombination in primary chicken embryos fibroblasts (CEF) infected with a RR-deleted COP virus and transfected by nucleofection with pTG19183 or pTG19185 (according to Amaxa Nucleofector technology).
  • Viral selection was performed by plaque purification after growth in Thymidine kinase-deficient (TK " ) 143B cells cultured in the presence of bromodeoxyuridine. This selection allows only TIC recombinant WR to remain viable. Absence of contamination by parental COP was verified by PCR.
  • the oncolytic activity of the COP viruses expressing huADAl and huADA2 was evaluated by cell viability measurements in both human colorectal carcinoma HCT116 (ATCC ® CCL-247TM) or human colorectal adenocarcinoma LoVo (ATCC ® CCL-229TM) infected cells.
  • a total of 5xl0 4 cells per well were plated in six-well culture dishes in 2 mL of culture medium and infected with either VVTG17989 (empty virus), COPTG19183 and COPTG19185 at a MOI of either 0.1, 0.01, 0.001 or 0.0001.
  • VVTG17989 empty virus
  • COPTG19183 COPTG19185
  • MOI 0.1, 0.01, 0.001 or 0.0001.
  • TK-RR-deleted COP vaccinia virus without transgene and TK-RR- deleted COP virus expressing the two human enzymes showed similar cytotoxicity in tumor cells. There is no impact of huADAl and huADA2 vectorization on in vitro oncolytic activity of VVCOP.
  • a set of oligonucleotides primers and probe that hybridize into the gene encoding for the secreted chemokine-binding protein (SCBP) of MVA were used (forward primer 5'-CGATGATGGAGTAATAAGTGGTAGGA-3' (SEQ ID NO: 7); reverse primer 5'- CACCGACCGATGATAAGATTTG-3' (SEQ ID NO: 8); probe 5' FAM - ACTGATTCCACCTCGGG - 3' (SEQ ID NO:9; MGB / NFQ).
  • the Quantitect Multiplex PCR kit from Qiagen was used to perform the q-PCR reactions on a 7300 real-time system from Applied Biosystem. As shown in Figure 8, no substantial difference could be detected for the replication of all the different viruses. Therefore, the expression of the adenosine deaminases has little or no effect on the infection and multiplication of the recombinant viruses.
  • the secreted adenosine deaminase activity was measured on the culture supernatants by quantifying the production of inosine from an excess of adenosine using HPLC and a diode array UV-visible detector. Briefly, the supernatants were diluted into 50 mmol.l "1 NaH 2 P0 4 buffer, pH 7.2, and the reaction was initiated by the addition of adenosine at a final concentration of 20 ⁇ , equivalent to physiological conditions, or 2 mM. After incubation at 30°C for 20 min. the reaction was stopped, and the proteins precipitated on ice by adding trifluoroacetic acid at a final concentration of 17%.
  • Inosine and adenosine were separated using a Sinergy Fusion RP C18 column (Phenomenex) using a mobile phase composed of 50 mmol.l 1 sodium acetate buffer at pH 4.5 and acetonitrile up to 50%. Inosine produced was quantified from the area under peak using a standard curve.
  • FIG. 10 shows that infection of HCT116 or LoVo cells by either of the two viruses encoding an ADA leads to the accumulation of adenosine deaminase activity in the culture supernatant.
  • adenosine is added in large excess
  • cellules infected by COPTG19185, encoding the huADA2 show higher levels of specific activity.
  • Figures IOC and 10D infection by COPTG19183, encoding the huADAl enzyme, results in higher specific ADA activity than after infection by COPTG19185.
  • the former ADA isoform presents a higher affinity for adenosine than the latter (Km equals to 20 ⁇ and 2.5 mM respectively) that largely compensate the lower level of secretion of this enzyme.
  • This higher affinity for the targeted metabolite results in higher degrading efficacy for the virus COPTG19183.
  • mice were injected subcutaneously into the right flanks of 15 Swiss nude mice. Seventeen days after inoculation, when tumours volume reached between 100 and 200 mm 3 , mice were injected once intravenously with vehicle or 10 7 pfu of COPTG19183 or COPTG19185. Tumours volume were monitored twice per week by calliper measurements for over 120 days and mice were sacrificed when tumour size reached 2000 mm 3 . After 3, 7 and 14 days, tumors were surgically removed from three mice per treated group and promptly snap-frozen for further analysis.
  • the adenosine deaminase activity was measured in the tumors after 7 days after the single virus injection.
  • the frozen tumors were thawed on ice, washed with PBS, and the interstitial fluids were collected by centrifugation for 10 min. at 500g after placing the tissues on a cell strainer with nylon mesh presenting 70 ⁇ pores.
  • the ADA specific activity was quantified by HPLC, as described above, with an adenosine concentration of 2 mM and 20 ⁇ .
  • Figure 13A illustrates the amount of inosine produced per minutes per microliter of tumor interstitial fluid from a large excess of adenosine.
  • mice As shown in Figure 15, the survival of mice is also significantly increased by the treatment with the oncolytic viruses. More specifically, all mice treated with the vehicle were dead at 57 days (0% survival rate), whereas at this time, 78%, 89% and 100 % survival rates were obtained respectively in empty virus control group, huADAl and huADA2 expressing viruses treated group respectively.
  • the use of xenograft models based on human cancer cells implanted in nude immunodeficient animals allows for the evaluation of the oncolytic activity of the VV constructs, as those cells are relatively permissive to those viruses, compared to mice cell lines. Nevertheless, the impaired adaptive immune response associated to those models prevent to appreciate the overall activity of the viruses armed with immunomodulators.
  • Antitumour activity of the ADA-expressing vaccinia viruses described above may be tested in immunocompetent preclinical models after implantation of tumour followed by injection of the constructs.
  • said anti-tumour activity can be tested in a mouse model composed of female NOD/Shi-scid/IL-2Rpull immunodeficient mouse strain (NOG) humanized using hematopoietic stem cells (CD34 + ) isolated from human cord blood and engrafted subcutaneously with human derived cancer cell line (e.g. HCT116).
  • NOG NOD/Shi-scid/IL-2Rpull immunodeficient mouse strain
  • CD34 + hematopoietic stem cells isolated from human cord blood and engrafted subcutaneously with human derived cancer cell line (e.g. HCT116).
  • VVTG18058 or COPTG19183 are injected (10 7 ⁇ /50 ⁇ - ⁇ ⁇ 5 ⁇ ).
  • Body weight and tumor volume are monitored three times a week.
  • tumors are harvested to evaluate viral replication, adenosine deaminase activity, the immune landscape by measuring immune cells populations (e.g.: CD4+, CD8+ T cells, T reg, NK cells, MDSCs, M1/M2 macrophages, etc.); soluble markers of the intratumoral immune response are also evaluated (e.g. IFNg, Granzyme B, IL12, VEGF, etc.).
  • immune cells populations e.g.: CD4+, CD8+ T cells, T reg, NK cells, MDSCs, M1/M2 macrophages, etc.
  • soluble markers of the intratumoral immune response are also evaluated (e.g. IFNg, Granzyme B, IL12, VEGF, etc.).
  • ADA-expressing vaccinia virus on the immune microenvironment can be assessed in an ex vivo environment reconstituted from human tumors by quantifying leucocytes (e.g. granulocytes, monocytes, T lymphocytes, T-helper cells, T regulatory cells, cytotoxic T cells, B lymphocytes, thrombocytes, NK cells) by using markers (ex: CD45 + , CD3 + , CD4 + , CD8 + , etc.), quantifying intracellular Thl cytokines (e.g. IFNy, TNFa), assessing the activation status of myeloid cells (e.g. DC, MDSC), etc.
  • leucocytes e.g. granulocytes, monocytes, T lymphocytes, T-helper cells, T regulatory cells, cytotoxic T cells, B lymphocytes, thrombocytes, NK cells
  • markers ex: CD45 + , CD3 + , CD4 + , CD8 + , etc.

Abstract

La présente invention concerne un virus oncolytique codant au moins un agent qui dégrade un ou plusieurs modulateur(s) immunitaires métaboliques, une composition le comprenant, ainsi que leur utilisation à des fins prophylactiques ou thérapeutiques, et plus particulièrement pour le traitement du cancer. La présente invention concerne également une méthode de traitement d'une maladie ou d'un état pathologique comprenant l'administration d'un tel virus oncolytique ou d'une composition de celui-ci et un procédé de préparation d'un tel virus oncolytique.
PCT/EP2018/069867 2017-07-28 2018-07-23 Virus oncolytiques exprimant des agents ciblant des modulateurs immunitaires métaboliques WO2019020543A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP17306012 2017-07-28
EP17306012.0 2017-07-28

Publications (1)

Publication Number Publication Date
WO2019020543A1 true WO2019020543A1 (fr) 2019-01-31

Family

ID=59761881

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/069867 WO2019020543A1 (fr) 2017-07-28 2018-07-23 Virus oncolytiques exprimant des agents ciblant des modulateurs immunitaires métaboliques

Country Status (1)

Country Link
WO (1) WO2019020543A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110747174A (zh) * 2019-10-30 2020-02-04 青岛宁逸生物科技有限公司 一种用于肿瘤治疗的重组病毒
EP3725323A1 (fr) * 2019-04-17 2020-10-21 Targovax Asa Vecteur adénoviral oncolytique exprimant un membre de la famille b7 des ligands costimulants et l'ada
WO2022076756A1 (fr) * 2020-10-07 2022-04-14 Devacell, Inc. Virus recombinés, systèmes d'administration modifiés en surface et méthodes associées

Citations (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
WO1996016183A1 (fr) 1994-11-17 1996-05-30 Cayla Genes suicide et associations d'analogues de nucleobases et nucleosides pyrimidiques avec des genes suicide en vue d'une therapie genique
WO1998002522A1 (fr) 1996-07-16 1998-01-22 Transgene S.A. Procede de conservation de virus recombinants infectieux, suspension aqueuse virale et utilisation comme medicament
WO1998004727A1 (fr) 1996-07-25 1998-02-05 Therion Biologics Corporation Poxvirus recombinant pour l'immunisation contre des antigenes associes aux tumeurs
WO1998010088A1 (fr) 1996-09-06 1998-03-12 Trustees Of The University Of Pennsylvania Procede inductible de production de virus adeno-associes recombines au moyen de la polymerase t7
WO1998037095A2 (fr) 1997-02-24 1998-08-27 Therion Biologics Corporation Poxvirus recombine destine a une immunisation contre l'antigene associe aux tumeurs muc1
WO1998056415A1 (fr) 1997-06-11 1998-12-17 Aquila Biopharmaceuticals, Inc. Saponines purifiees servant d'adjuvants oraux
US6054438A (en) 1987-01-07 2000-04-25 Imperial Cancer Research Technology Limited Nucleic acid fragments encoding portions of the core protein of the human mammary epithelial mucin
EP0998568A1 (fr) 1998-04-17 2000-05-10 Transgene S.A. Mutant ayant une activite uracile phosphoribosyl transferase
WO2001066137A1 (fr) 2000-03-07 2001-09-13 Merck & Co., Inc. Formulations d'adenovirus
WO2002090501A2 (fr) * 2001-05-08 2002-11-14 Gary Gamerman Vecteurs dont la replication, l'immunogenicite et/ou la pathogenicite sont regulees par le promoteur du stress et utilisation de ceux-ci
WO2003053463A2 (fr) 2001-12-10 2003-07-03 Bavarian Nordic A/S Preparations contenant un poxvirus et procede de preparation de compositions stables contenant un poxvirus
WO2005007857A1 (fr) 2003-07-21 2005-01-27 Transgene S.A. Polypeptide a activite cytosine-desaminase amelioree
WO2005042728A2 (fr) 2003-11-03 2005-05-12 Probiogen Ag Lignees de cellules aviaires immortalisees pour la production de virus
WO2006108846A1 (fr) 2005-04-11 2006-10-19 Vivalis Procede de fabrication de vaccins viraux dans des lignees de cellules souches embryonnaires aviaires derivees en suspension
WO2007056847A1 (fr) 2005-11-21 2007-05-24 Sanofi Pasteur Limited Formulations de stabilisation pour virus recombinants
WO2007147528A1 (fr) 2006-06-20 2007-12-27 Transgene S.A. Procédé de production de poxvirus et compositions de poxvirus
WO2007147529A2 (fr) 2006-06-20 2007-12-27 Transgene S.A. Vaccin viral recombinant
WO2008114021A1 (fr) 2007-03-19 2008-09-25 Stabilitech Ltd. Procédé de conservation de particules virales
WO2008129058A1 (fr) 2007-04-24 2008-10-30 Vivalis Lignées de cellules souches dérivées de l'embryon de canard pour la production de vaccins viraux
WO2008138533A1 (fr) 2007-05-14 2008-11-20 Bavarian Nordic A/S Purification de vaccins à base de virus vaccinia et de virus vaccinia recombinés
WO2009065546A1 (fr) 2007-11-19 2009-05-28 Transgene Sa Vecteurs oncolytiques dérivés de poxvirus
WO2009065547A2 (fr) 2007-11-19 2009-05-28 Transgene Sa Vecteurs oncolytiques poxviraux
WO2009100521A1 (fr) 2008-02-12 2009-08-20 Sanofi Pasteur Limited Procédés d’utilisation de chromatographie d'échange d'ions et de chromatographie d'exclusion diffusion pour la purification du poxvirus
WO2010130756A1 (fr) 2009-05-12 2010-11-18 Transgene Sa Lignées cellulaires aviaires immortalisées et applications associées
WO2011128704A1 (fr) 2010-04-16 2011-10-20 Isis Innovation Limited Système d'expression de poxvirus
WO2012001075A2 (fr) 2010-07-02 2012-01-05 Transgene Lignees cellulaire aviaires immortalisées
WO2013022764A1 (fr) 2011-08-05 2013-02-14 David Kirn Procédés et compositions de production du virus de la vaccine
US8394385B2 (en) 2009-03-13 2013-03-12 Bavarian Nordic A/S Optimized early-late promoter combined with repeated vaccination favors cytotoxic T cell response against recombinant antigen in MVA vaccines
WO2014063832A1 (fr) 2012-10-28 2014-05-01 Bavarian Nordig A/S Promoteur pr13.5 pour lymphocytes t robustes et réponses d'anticorps
US8772023B2 (en) 2008-11-27 2014-07-08 Bavarian Nordic A/S Promoters for recombinant viral expression
WO2016061286A2 (fr) 2014-10-14 2016-04-21 Halozyme, Inc. Compositions d'adénosine désaminase-2 (ada2), variants de cette dernière et leurs procédés d'utilisation
WO2016191283A2 (fr) * 2015-05-22 2016-12-01 University Of Houston System Immunomodulation enzymatique de tumeurs solides et utilisateur associé

Patent Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
US6054438A (en) 1987-01-07 2000-04-25 Imperial Cancer Research Technology Limited Nucleic acid fragments encoding portions of the core protein of the human mammary epithelial mucin
WO1996016183A1 (fr) 1994-11-17 1996-05-30 Cayla Genes suicide et associations d'analogues de nucleobases et nucleosides pyrimidiques avec des genes suicide en vue d'une therapie genique
WO1998002522A1 (fr) 1996-07-16 1998-01-22 Transgene S.A. Procede de conservation de virus recombinants infectieux, suspension aqueuse virale et utilisation comme medicament
WO1998004727A1 (fr) 1996-07-25 1998-02-05 Therion Biologics Corporation Poxvirus recombinant pour l'immunisation contre des antigenes associes aux tumeurs
WO1998010088A1 (fr) 1996-09-06 1998-03-12 Trustees Of The University Of Pennsylvania Procede inductible de production de virus adeno-associes recombines au moyen de la polymerase t7
WO1998037095A2 (fr) 1997-02-24 1998-08-27 Therion Biologics Corporation Poxvirus recombine destine a une immunisation contre l'antigene associe aux tumeurs muc1
WO1998056415A1 (fr) 1997-06-11 1998-12-17 Aquila Biopharmaceuticals, Inc. Saponines purifiees servant d'adjuvants oraux
EP0998568A1 (fr) 1998-04-17 2000-05-10 Transgene S.A. Mutant ayant une activite uracile phosphoribosyl transferase
WO2001066137A1 (fr) 2000-03-07 2001-09-13 Merck & Co., Inc. Formulations d'adenovirus
WO2002090501A2 (fr) * 2001-05-08 2002-11-14 Gary Gamerman Vecteurs dont la replication, l'immunogenicite et/ou la pathogenicite sont regulees par le promoteur du stress et utilisation de ceux-ci
WO2003053463A2 (fr) 2001-12-10 2003-07-03 Bavarian Nordic A/S Preparations contenant un poxvirus et procede de preparation de compositions stables contenant un poxvirus
WO2005007857A1 (fr) 2003-07-21 2005-01-27 Transgene S.A. Polypeptide a activite cytosine-desaminase amelioree
WO2005042728A2 (fr) 2003-11-03 2005-05-12 Probiogen Ag Lignees de cellules aviaires immortalisees pour la production de virus
WO2006108846A1 (fr) 2005-04-11 2006-10-19 Vivalis Procede de fabrication de vaccins viraux dans des lignees de cellules souches embryonnaires aviaires derivees en suspension
WO2007056847A1 (fr) 2005-11-21 2007-05-24 Sanofi Pasteur Limited Formulations de stabilisation pour virus recombinants
WO2007147528A1 (fr) 2006-06-20 2007-12-27 Transgene S.A. Procédé de production de poxvirus et compositions de poxvirus
WO2007147529A2 (fr) 2006-06-20 2007-12-27 Transgene S.A. Vaccin viral recombinant
WO2008114021A1 (fr) 2007-03-19 2008-09-25 Stabilitech Ltd. Procédé de conservation de particules virales
WO2008129058A1 (fr) 2007-04-24 2008-10-30 Vivalis Lignées de cellules souches dérivées de l'embryon de canard pour la production de vaccins viraux
WO2008138533A1 (fr) 2007-05-14 2008-11-20 Bavarian Nordic A/S Purification de vaccins à base de virus vaccinia et de virus vaccinia recombinés
WO2009065546A1 (fr) 2007-11-19 2009-05-28 Transgene Sa Vecteurs oncolytiques dérivés de poxvirus
WO2009065547A2 (fr) 2007-11-19 2009-05-28 Transgene Sa Vecteurs oncolytiques poxviraux
WO2009100521A1 (fr) 2008-02-12 2009-08-20 Sanofi Pasteur Limited Procédés d’utilisation de chromatographie d'échange d'ions et de chromatographie d'exclusion diffusion pour la purification du poxvirus
US8772023B2 (en) 2008-11-27 2014-07-08 Bavarian Nordic A/S Promoters for recombinant viral expression
US8394385B2 (en) 2009-03-13 2013-03-12 Bavarian Nordic A/S Optimized early-late promoter combined with repeated vaccination favors cytotoxic T cell response against recombinant antigen in MVA vaccines
WO2010130753A1 (fr) 2009-05-12 2010-11-18 Transgene Sa Procédé de préparation et de purification d'orthopoxvirus
WO2010130756A1 (fr) 2009-05-12 2010-11-18 Transgene Sa Lignées cellulaires aviaires immortalisées et applications associées
WO2011128704A1 (fr) 2010-04-16 2011-10-20 Isis Innovation Limited Système d'expression de poxvirus
WO2012001075A2 (fr) 2010-07-02 2012-01-05 Transgene Lignees cellulaire aviaires immortalisées
WO2013022764A1 (fr) 2011-08-05 2013-02-14 David Kirn Procédés et compositions de production du virus de la vaccine
WO2014063832A1 (fr) 2012-10-28 2014-05-01 Bavarian Nordig A/S Promoteur pr13.5 pour lymphocytes t robustes et réponses d'anticorps
WO2016061286A2 (fr) 2014-10-14 2016-04-21 Halozyme, Inc. Compositions d'adénosine désaminase-2 (ada2), variants de cette dernière et leurs procédés d'utilisation
WO2016191283A2 (fr) * 2015-05-22 2016-12-01 University Of Houston System Immunomodulation enzymatique de tumeurs solides et utilisateur associé

Non-Patent Citations (114)

* Cited by examiner, † Cited by third party
Title
"Genbank", Database accession no. AF190746
"Genbank", Database accession no. AF234182.1
"Genbank", Database accession no. AF298886
"Genbank", Database accession no. AF482758.2
"GenBank", Database accession no. CR380956
"Genbank", Database accession no. DQ 437593
"Genbank", Database accession no. DQ835357.1
"Genbank", Database accession no. DQ835383.1
"Genbank", Database accession no. HQ407377
"Genbank", Database accession no. HQ420893
"Genbank", Database accession no. HQ420894
"Genbank", Database accession no. HQ420895
"Genbank", Database accession no. HQ420896
"Genbank", Database accession no. HQ420897
"Genbank", Database accession no. HQ420898
"Genbank", Database accession no. HQ420899
"Genbank", Database accession no. HQ420900
"GenBank", Database accession no. M59033
"Genbank", Database accession no. NC 003663
"Genbank", Database accession no. NC_001132
"Genbank", Database accession no. NC_003663
"Genbank", Database accession no. NC_004105
"Genbank", Database accession no. NC_005309
"Genbank", Database accession no. NC_006998
"Genbank", Database accession no. NM_141609.3
"Genbank", Database accession no. X02994
"Genbank", Database accession no. X94355
"GenBank", Database accession no. Z46843
A. GENNARO: "Remington: The Science and Practice of Pharmacy", LIPPINCOTT, WILLIAMS&WILKINS
ADRA ET AL., GENE, vol. 60, 1987, pages 65 - 74
ALLARD ET AL., IMMUNOTHERAPY, 2016
ALLARD ET AL., INTERNATIONAL JOURNAL OF CANCER, vol. 134, no. 6, 2014, pages 1466 - 1473
ANDERSEN ET AL., EUROPEAN J. BIOCHEM., vol. 204, 1992, pages 51 - 56
ANTONIOLI ET AL., NATURE REVIEWS CANCER, vol. 13, 2013, pages 842 - 857
BALICKI D ET AL: "GENE THERAPY OF HUMAN DISEASE", MEDIC, WILLIAMS AND WILKINS, BALTIMORE, US, vol. 81, no. 1, 1 January 2002 (2002-01-01), pages 69 - 86, XP008050587, ISSN: 0025-7974, DOI: 10.1097/00005792-200201000-00005 *
BEAVIS ET AL., ONCOIMMUNOLOGY, vol. 2, no. 12, 2013, pages e26705
BOVIATSIS ET AL., GENE THER., vol. 1, pages 323 - 31
BROYLES ET AL., VIROL., vol. 195, 1993, pages 863 - 5
BULLER ET AL., NATURE, vol. 317, no. 6040, 1985, pages 813 - 5
BULLER R M L ET AL: "DECREASED VIRULENCE OF RECOMBINANT VACCINIA VIRUS EXPRESSION VECTORS IS ASSOCIATED WITH A THYMIDINE KINASE-NEGATIVE PHENOTYPE", NATURE, MACMILLAN JOURNALS LTD, LONDON, GB, vol. 317, no. 6040, 1 January 1985 (1985-01-01), pages 813 - 815, XP002255895, ISSN: 0028-0836, DOI: 10.1038/317813A0 *
CAROLL ET AL., VIROLOGY, vol. 238, 1997, pages 198 - 211
CARREY ET AL., PLOS ONE, vol. 6, no. 7, 2011, pages e22442
CARREY ET AL., SCI REP, vol. 4, 2014, pages 6154
CHAFFEE ET AL., J CLIN INVEST, vol. 89, 1992, pages 1643 - 1651
CHAKRABARTI ET AL., BIOTECHNIQUES, vol. 23, 1997, pages 1094 - 7
CHAMBERS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 92, 1995, pages 1411 - 5
CHAMBERS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 92, pages 1411 - 5
CHERNAJOVSKY ET AL., BMJ, vol. 332, no. 7534, 2006, pages 170 - 2
CHU ET AL., J. EXP. MED., vol. 186, 1997, pages 1623
CRISTALLI ET AL., MED RES REV, vol. 21, no. 2, 2001, pages 105 - 28
DAVID ALLARD ET AL: "CD73-adenosine: a next-generation target in immuno-oncology", IMMUNOTHERAPY, vol. 8, no. 2, 1 February 2016 (2016-02-01), GB, pages 1 - 19, XP055420607, ISSN: 1750-743X, DOI: 10.2217/imt.15.106 *
DAYHOFFED: "Atlas of Protein Sequence and Structure", 1981, pages: 482 - 9
ERBS ET AL., CANCER GENE THER., vol. 15, no. 1, 2008, pages 18 - 28
ERBS ET AL., CANCER RES., vol. 60, no. 14, 2000, pages 3813
ERBS PHILIPPE ET AL: "In vivo cancer gene therapy by adenovirus-mediated transfer of a bifunctional yeast cytosine deaminase/uracil phosphoribosyltransferase fusion gene", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 60, no. 14, 15 July 2000 (2000-07-15), pages 3813 - 3822, XP002355011, ISSN: 0008-5472 *
FALLAUX ET AL., HUMAN GENE THER., vol. 9, 1998, pages 1909 - 17
FEND ET AL., CANCER RES., vol. 77, no. 15, 2017, pages 4146 - 4157
FISHER ET AL., CURR. OPIN. MOL. THER., vol. 8, no. 4, 2006, pages 301 - 13
FOLOPPE ET AL., GENE THER., vol. 15, 2008, pages 1361 - 71
FREEMAN ET AL., MOL. THER., vol. 13, no. 1, 2006, pages 221 - 8
GAKIS ET AL., EUR RESPIR J, vol. 9, 1996, pages 632 - 633
GEEVARGHESE ET AL., HUM. GENE THER., vol. 21, no. 9, 2010, pages 1119 - 28
GOUBRAN ET AL., CANCER GROWTH METASTASIS, vol. 7, 2014, pages 9 - 18
GRAHAM ET AL., J. GEN. VIROL., vol. 36, 1997, pages 59 - 72
GUSE ET AL., EXPERT OPINION BIOL. THER., vol. ll, no. 5, 2011, pages 595 - 608
HAMMOND ET AL., J. VIROL. METHODS, vol. 66, 1997, pages 135 - 8
HARRINGTON ET AL., EXPERT REV. ANTICANCER THER., vol. 15, no. 12, 2015, pages 1389 - 1403
HAUSLER ET AL., CANCER IMMUNOLOGY, IMMUNOTHERAPY, vol. CII 60, no. 10, 2011, pages 1405 - 1418
HAUSLER ET AL., CANCER IMMUNOLOGY, IMMUNOTHERAPY, vol. Cll 60, no. 10, 2011, pages 1405 - 1418
HERMISTON ET AL., CURR. OPIN. MOL. THER., vol. 8, no. 4, 2006, pages 322 - 30
JOYCE JA; POLLARD JW., NAT REV CANCER, vol. 9, no. 4, 2009, pages 239 - 252
KAUFMANN ET AL., J. INVEST. DERMATOL., vol. 133, no. 4, 2013, pages 1034 - 42
KERN ET AL., GENE, vol. 88, 1990, pages 149 - 57
KHURI ET AL., NAT. MED, vol. 6, no. 8, 2000, pages 879 - 85
KIRN ET AL., NAT. MED., vol. 7, 2001, pages 781
KRITSCH ET AL., J. CHROMATOGR. ANAL. TECHNOL. BIOMED. LIFE SCI., vol. 822, 2005, pages 263 - 70
KUMAR ET AL., EUR J PHARMACOL., vol. 616, no. 1-3, 2009, pages 7 - 15
KUMAR; BOYLE, VIROLOGY, vol. 179, 1990, pages 151 - 8
LANNONE ET AL., AMERICAN JOURNAL OF CANCER RESEARCH, vol. 4, no. 2, 2014, pages 172 - 181
LORENCE ET AL., CURR. CANCER DRUG TARGETS, vol. 7, no. 2, 2007, pages 157 - 67
LU ET AL., WORLD J GASTROENTEROL, vol. 19, no. 12, 2013, pages 1912 - 1918
MARTINUSSEN ET AL., J. BACTERIOL., vol. 176, 1994, pages 6457 - 63
MARTINUSSEN ET AL., J. BACTERIOL., vol. 177, 1995, pages 271 - 4
MARTUZA ET AL., SCIENCE, vol. 252, 1991, pages 854 - 6
MCDONALD ET AL., BREAST CANCER TREAT., vol. 99, no. 2, 2006, pages 177 - 84
MINETA ET AL., CANCER RES., vol. 54, 1994, pages 3363 - 66
OHTA A., FRONT. IMMUNOL., vol. 7, 2016, pages 109
OHTA ET AL., NATURE, vol. 414, no. 6866, 2001, pages 916 - 920
OLDHAM; DILLMAN: "Principles of cancer biotherapy", 2009, SPRINGER
PEREZ; BRADY: "Principles and Practice of Radiation Oncology", 1992, JB LIPPINCOTT CO
PHUANGSAB ET AL., CANCER LETT., vol. 172, no. 1, 2001, pages 27 - 36
PYLES ET AL., J. VIROL., vol. 68, 1994, pages 4963 - 72
QUAIL DF; JOYCE JA., NAT. MED., vol. 19, no. 11, 2013, pages 1423 - 1437
RIBEIRO ET AL., J. EXP. BIOL., vol. 204, 2001, pages 2001 - 2010
ROSENBERG ET AL., J IMMUNOL, vol. 135, no. 4, 1985, pages 2895 - 2903
RUDIN ET AL., CLIN. CANCER. RES., vol. 17, no. 4, 2011, pages 888 - 95
SMORLESI, GENE THER., vol. 12, 2005, pages 1324
STERN, B.; L. OLSEN, TRENDS CELL MOL. BIOL, vol. 2, 2007, pages 1 - 17
STOJDL ET AL., CANCER CELL, vol. 4, no. 4, 2003, pages 263 - 75
STOJDL ET AL., NAT. MED., vol. 6, no. 7, 2000, pages 821 - 5
SUADER, J. AM ACAD DERMATOL., vol. 43, 2000, pages 6
SUMINO ET AL., J.VIROL., vol. 72, 1998, pages 4931
THORNE ET AL., CURR. OPIN. MOL. THER., vol. 7, no. 4, 2005, pages 359 - 65
TRITEL ET AL., J. IMMUNOL., vol. 171, 2003, pages 2358
UNGERER ET AL., CLIN CHEM, vol. 38/7, 1992, pages 1322 - 1326
VAN DER MAADEN ET AL., J. CONTROL RELEASE, vol. 161, 2012, pages 645 - 55
WANG ET AL., AACR ANNUAL MEETING, 2006
WANG LIN ET AL: "Enzymatic depletion of adenosine by pegylated, engineered adenosine deaminase 2 (PEG-ADA2): A novel immunotherapeutic approach to treat solid tumors", CANCER RESEARCH, vol. 76, no. Suppl. 14, July 2016 (2016-07-01), & 107TH ANNUAL MEETING OF THE AMERICAN-ASSOCIATION-FOR-CANCER-RESEARCH (AACR); NEW ORLEANS, LA, USA; APRIL 16 -20, 2016, pages 1472, XP055510255 *
WIRTH THOMAS ET AL: "History of gene therapy", GENE, ELSEVIER, AMSTERDAM, NL, vol. 525, no. 2, 23 April 2013 (2013-04-23), pages 162 - 169, XP028576972, ISSN: 0378-1119, DOI: 10.1016/J.GENE.2013.03.137 *
WONG ET AL., VIRUSES, vol. 2, 2010, pages 78 - 106
WU ET AL., J SURG ONCOL, 2012
XIA ET AL., AI ZHENG, vol. 23, no. 12, 2004, pages 1666 - 70
ZAVIALOV; ENGSTROM, BIOCHEM J., vol. 391, 2005, pages 51 - 57
ZHANG ET AL., CANCER RES., vol. 67, 2007, pages 10038 - 46

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3725323A1 (fr) * 2019-04-17 2020-10-21 Targovax Asa Vecteur adénoviral oncolytique exprimant un membre de la famille b7 des ligands costimulants et l'ada
WO2020212503A1 (fr) * 2019-04-17 2020-10-22 Targovax Asa Vecteur adénoviral oncolytique exprimant un membre de la famille b7 de ligands costimulateurs et d'ada
CN114007630A (zh) * 2019-04-17 2022-02-01 塔格瓦克斯公司 表达b7家族共刺激配体成员和ada的溶瘤腺病毒载体
CN110747174A (zh) * 2019-10-30 2020-02-04 青岛宁逸生物科技有限公司 一种用于肿瘤治疗的重组病毒
WO2022076756A1 (fr) * 2020-10-07 2022-04-14 Devacell, Inc. Virus recombinés, systèmes d'administration modifiés en surface et méthodes associées

Similar Documents

Publication Publication Date Title
EP3562946B1 (fr) Virus oncolytiques et molécules thérapeutiques
Guse et al. Oncolytic vaccinia virus for the treatment of cancer
AU2018215558B2 (en) Oncolytic virus therapy
Bourke et al. The emerging role of viruses in the treatment of solid tumours
CN101868546B (zh) 痘病毒溶瘤载体
US11806374B2 (en) Isolated recombinant oncolytic adenoviruses, pharmaceutical compositions, and uses thereof for drugs for treatment of tumors and/or cancers
Abd-Aziz et al. Development of oncolytic viruses for cancer therapy
Vachani et al. Gene therapy for mesothelioma and lung cancer
Irwin et al. Targeting nucleotide biosynthesis: a strategy for improving the oncolytic potential of DNA viruses
WO2019020543A1 (fr) Virus oncolytiques exprimant des agents ciblant des modulateurs immunitaires métaboliques
KR20200026894A (ko) 개인 맞춤형 백신
Goradel et al. Oncolytic virotherapy as promising immunotherapy against cancer: mechanisms of resistance to oncolytic viruses
WO2018091680A1 (fr) Vecteurs oncolytiques à base de la variole bovine
WO2020011754A1 (fr) Virus de la vaccine chimériques
EP3129037B1 (fr) Vecteurs oncolytiques poxviraux
Mohamadi et al. The important role of oncolytic viruses in common cancer treatments
Thoidingjam et al. Oncolytic virus-based suicide gene therapy for cancer treatment: a perspective of the clinical trials conducted at Henry Ford Health
Chen et al. Progress of oncolytic virotherapy for neuroblastoma
Vidal et al. Reovirus and other oncolytic viruses for the targeted treatment of cancer
Garcia-Aragoncillo et al. Design of virotherapy for effective tumor treatment
WO2024038175A1 (fr) Poxvirus chimériques
TW202413636A (zh) 嵌合痘病毒
KR20220116191A (ko) 4-1bbl 아쥬반트화 재조합 변형 백시니아 바이러스 앙카라 (mva)의 의약적 용도
Wongthida Understanding, and exploiting, the contribution of the immune system to the therapeutic efficacy of oncolytic virotherapy with vesicular stomatitis virus (VSV)

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18740612

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18740612

Country of ref document: EP

Kind code of ref document: A1