US20100062489A1 - Duck embryonic derived stem cell lines for the production of viral vaccines - Google Patents

Duck embryonic derived stem cell lines for the production of viral vaccines Download PDF

Info

Publication number
US20100062489A1
US20100062489A1 US12/597,486 US59748609A US2010062489A1 US 20100062489 A1 US20100062489 A1 US 20100062489A1 US 59748609 A US59748609 A US 59748609A US 2010062489 A1 US2010062489 A1 US 2010062489A1
Authority
US
United States
Prior art keywords
cells
virus
duck
cell
medium
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/597,486
Other languages
English (en)
Inventor
Fabienne Guehenneux
Karine Moreau
Magali Esnault
Majid Mehtali
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Valneva SE
Original Assignee
Vivalis 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 Vivalis SA filed Critical Vivalis SA
Publication of US20100062489A1 publication Critical patent/US20100062489A1/en
Assigned to VIVALIS reassignment VIVALIS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ESNAULT, MAGALI, GUEHENNEUX, FABIENNE, MEHTALI, MAJID, MOREAU, KARINE
Priority to US14/069,423 priority Critical patent/US9260694B2/en
Assigned to VALNEVA SE reassignment VALNEVA SE CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: VIVALIS SA
Assigned to VALNEVA reassignment VALNEVA CORRECTIVE ASSIGNMENT TO CORRECT THE CHANGE OF NAME RECORDATION PREVIOUSLY RECORDED ON REEL 031607 FRAME 0481. ASSIGNOR(S) HEREBY CONFIRMS THE CONVEYING PARTY: VIVALIS RECEIVING PARTY: VALNEVA. Assignors: VIVALIS
Priority to US15/013,079 priority patent/US9822345B2/en
Priority to US15/785,530 priority patent/US20180135026A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/145Orthomyxoviridae, e.g. influenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/155Paramyxoviridae, e.g. parainfluenza virus
    • A61K39/17Newcastle disease virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/275Poxviridae, e.g. avipoxvirus
    • A61K39/285Vaccinia virus or variola virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/10Inactivation or decontamination of a medicinal preparation prior to administration to an animal or a person
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/863Poxviral vectors, e.g. entomopoxvirus
    • C12N15/8636Vaccina virus vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/70Undefined extracts
    • C12N2500/80Undefined extracts from animals
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/105Insulin-like growth factors [IGF]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/125Stem cell factor [SCF], c-kit ligand [KL]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/13Nerve growth factor [NGF]; Brain-derived neurotrophic factor [BDNF]; Cilliary neurotrophic factor [CNTF]; Glial-derived neurotrophic factor [GDNF]; Neurotrophins [NT]; Neuregulins
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2306Interleukin-6 (IL-6)
    • 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
    • C12N2710/24134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2710/24151Methods of production or purification of viral material
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16051Methods of production or purification of viral material
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18111Avulavirus, e.g. Newcastle disease virus
    • C12N2760/18134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18111Avulavirus, e.g. Newcastle disease virus
    • C12N2760/18151Methods of production or purification of viral material
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18411Morbillivirus, e.g. Measles virus, canine distemper
    • C12N2760/18434Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/18011Paramyxoviridae
    • C12N2760/18411Morbillivirus, e.g. Measles virus, canine distemper
    • C12N2760/18451Methods of production or purification of viral material

Definitions

  • the present invention relates to the development and manufacturing of viral vaccines.
  • the invention relates to the field of industrial production of viral vectors and vaccines, more specifically to the use of duck cell lines derived from embryonic stem cells that are free of avian endogenous retrovirus, for the production of viral vectors and viruses.
  • the invention is particularly useful for the industrial production of viral vaccines to prevent viral infection of humans and animals.
  • Vaccines effectively reduce and prevent death and disease from many viral infections such as for example flu, measles, mumps, smallpox, yellow fever.
  • Both ALV-E and EAV are members of endogenous retrovirus families present in the chicken germ line.
  • ALV-E are expressed from ev loci, which are inheritable proviral elements. Based on their envelope sequences, ALV-E are differentiated from ALV subgroups A to D and J which are exogenously acquired infections. While exogenous ALVs cause several neoplastic diseases, such as myocarditis and osteopetrosis in infected chickens, ALV-E are not known to be pathogenic to chickens. The lack of oncogenic potential with ALV-E infections may be attributed to the absence of both a viral oncogene and enhancer activity in the endogenous long terminal repeat (LTR).
  • LTR long terminal repeat
  • Ev loci More than 20 different ev loci have been identified in White Leghorn chickens (ev-1 to ev-22). Ev loci designations are assigned in the order discovered and are phenotypically categorized with regard to the gene products they express and their capacity to generate infectious particles. ALV-E phenotypes conferred by ev loci range from structurally and enzymatically complete infectious particles to structurally or enzymatically (RT-) defective to no detectable viral protein expression. Most ev loci are structurally incomplete and therefore do not encode all sequences necessary for production of infectious virus particles. Chicken strain, named ev-0, has been obtained by breeding to be resistant to ALV-E.
  • Line-0 chickens are lacking ev loci (i.e ev-0) but EAV proviral sequences are present in the genome line 0 chickens (Dunwiddie and Far as, 1985, Proc Natl. Acad. Sci. USA, 82: 5097-5101).
  • EAV EAV-associated RT activity found in the supernatants of cultured chick embryo fibroblasts.
  • cell lines for the production of viral vaccines are MDCK (cells derived from the kidney of Madin-Darby dog), PerC6 (cells derived from human embryonic retinal cells genetically modified by inserting the E1 genes from the human adenovirus type 5) developed by CRUCELL (Netherland)), VERO (cells derived from epithelial cells of kidney from African green monkey ( Cercopithecus aethiops ) isolate at the Chiba University in Chiba, Japan), BHK21 (Cells immortalized from baby hamster kidney cells). None of the cell lines available fulfil all the medical, regulatory and industrial requirements.
  • the inventor has taken advantage of its expertise in avian biology and in avian embryonic stem (ES) cells to undertake the development of novel stable duck cell lines that enables the efficient replication of a large group of human and veterinarian vaccines and vaccine candidates.
  • ES avian embryonic stem
  • the inventor was able to generate a series of well characterized and documented duck cell lines (i.e the dEBx® cells) that are derived from duck ES cells, with no steps of genetic, chemical or viral immortalization and that do not produce replication-competent retroviruses in culture.
  • the instant invention provides a process for obtaining continuous diploid avian cell lines, named EBx, derived from avian embryonic stem cells (ES), wherein said avian cell lines do not produce replication-competent endogenous retrovirus particles.
  • the cell lines of the invention are “continuous” because they have the characteristics to be cultured in vitro over an extended period of time.
  • the cells of the invention are capable of proliferating for at least 50 generations, at least 75 generations, at least 100 generations, at least 125 generations, at least 150 generations, at least 175 generations, at least 200 generations, at least 250 generations.
  • the 250 generations do not constitute a time limit because the cells obtained are still alive and can still be passaged for additional passages.
  • the cells of the invention can be cultured “continuously” as long as telomerase is expressed by the cells. Indeed, it is assumed that the high level of telomerase expression of avian cells of the invention is responsible for genetic stability (i.e avian cells of the invention are diploid) and the continuous cell growth.
  • passage it is meant the transfer of transplantation of cells, with or without dilution, from one culture vessel to another. It is understood that any time cells are transferred from one vessel to another, a certain portion of the cells may be lost and therefore, dilution of cells, whether deliberate or not, may occur.
  • This term is synonymous with the term ‘subculture’.
  • the passage number is the number of times the cells in the culture, that grow either in suspension or in adherence, have been sub-cultured or passed in a new vessel. This term is not synonymous with population doubling or generation which is the time needed by a cell population to replicate one time; that is to say, roughly the time for each cells of a population to replicate.
  • Avian ES cells of step a) of the invention have a population doubling time (PDT) of around >40 hours.
  • the avian EBx cells of the invention have a PDT of around ⁇ 30 hours; usually for EBx® cells, there is one passage every 3 generations.
  • diploid it is mean that cells of the invention have two copies (2n) of each chromosome, usually one from the mother and one from the father.
  • avian EBx® cell lines of the invention are continuous and diploid (i.e genetically stable) constitutes a remarkable and unique feature because these terms are usually antagonist.
  • cancer cells and/or immortalized cells obtained by chemical, physical (U.V irradiation, X-ray or g-irradiation, . . . ) or genetic modification (virus transformation, oncogenes overexpression, . . . ) are continuous cells because they are able to replicate indefinitely into culture, but they are not genetically stable because they display polyplo ⁇ d karyotypes.
  • primary cells such as chicken embryonic fibroblasts, MRC5, WI38 which are non-transformed cells, are not continuous because they have a finite life-span after few generation, but they are genetically stable (i.e diploid) cells.
  • avian, “bird”, “ayes” or “ava” as used herein is intended to have the same meaning, and will be used indistinctly.
  • “Birds” refer to any species, subspecies or race of organism of the taxonomic class ⁇ ava>>.
  • “birds” refer to any animal of the taxonomix order:
  • endogenous retroviral particle or “endogenous retrovirus particle”, terms that could be used indistinctively, it is meant a retroviral particle or retrovirus encoded by and/or expressed from ALV-E or EAV proviral sequences present in some avian cell genomes.
  • ALV-E proviral sequences are known to be present in the genome of domestic chicken (except Line-0 chicken), red jungle fowl and Ringneck Pheasant.
  • EAV proviral sequences are known to be present in all genus gallus that includes domestic chicken, Line-0 chicken, red jungle fowl, green jungle fowl, grey jungle fowl, Ceylonese jungle fowl and allies) (see Resnick et al., 1990, J. Virol., 64:4640-4653).
  • the bird of the invention are selected among the birds that does not comprises ALV-E and EAV proviral sequences in its genome.
  • a man skilled in the art is able to determine whether ALV-E and EAV sequences are present in a bird genome (Johnson and Heneine, 2001; Weissmahr et al., 1996).
  • the bird is selected in the group comprising Anseriformes (i.e duck, goose, swan), turkeys, quails, Japanese quail, Guinea fowl, Pea Fowl. Therefore, cells derived from such bird do not produce replication-competent endogenous ALV-E and/or EAV particles.
  • the bird of the present invention is selected among the group comprising ducks, geese, swans, turkeys, quails and Japanese quails, Guinea Fowls and Pea Fowls.
  • the bird is a duck, more preferably a Pekin or Muscovy ducks.
  • the bird is a Pekin duck. Therefore, the instant invention provides a process for obtaining continuous diploid duck cell lines derived from embryonic stem cells (ES), wherein said duck cell lines do not produce replication-competent endogenous retrovirus particles.
  • ES embryonic stem cells
  • the bird of the invention are selected among the birds that does not comprises complete ALV-E proviral sequences in its genome but eventually EAV proviral sequences.
  • a man skilled in the art is able to determine whether partial or complete ALV-E and EAV sequences are present in a bird genome (Johnson and Heneine, 2001).
  • ev-0 strain Several chicken strains have been selected by breeding that do not contain complete ALV-E proviral sequences (i.e: ev-0 strain) and therefore do not produce infectious ALV-E retroparticles, such as:
  • the bird is an ev-0 domestic chicken ( Gallus Gallus subspecies domesticus ), preferably selected among ELL-0, DE and PE11.
  • the instant invention provides a process for obtaining continuous diploid chicken cell lines derived from embryonic stem cells (ES) of ev-0 chicken strains, wherein said ev-0 chicken cell lines do not produce replication-competent endogenous retrovirus particles.
  • ES embryonic stem cells
  • the bird of the invention are selected among the birds that comprise complete and/or incomplete ALV-E and EAV proviral sequences in its genome but that are unable to produce replication competent ALV-E and EAV retroparticles.
  • a man skilled in the art is able to determine whether ALV-E and/or EAV infectious and/or non-infectious retroparticles are produced from a bird cells (Johnson and Heneine, 2001; Weissmahr et al., 1996).
  • the bird is selected in the group comprising specific pathogen free (SPF) chicken, preferably from Valo strain (Lohman) or Line 22 (SPAFAS).
  • SPPF specific pathogen free
  • SPAFAS Line 22
  • retroviral particles are infectious, that is to say that such retroviral particules are able to infect and to replicate in avian cells of the invention.
  • EBx® The process of establishment of continuous diploid avian cell lines, named EBx®, of the invention comprises two steps:
  • the modification of the culture medium of step b) of the process of establishment EBx® cell lines, so as to obtain progressive or total withdrawal of growth factors, serum and feeder layer, can be made simultaneously, successively or separately.
  • the sequence of the weaning of the culture medium may be chosen among:
  • the sequence of the weaning is growth factors/feeder layer/serum.
  • the withdrawal of additives such as sodium pyruvate, non essential amino acids (NNEA), vitamins, yeastolate are performed after the weaning of feeder layer and before the weaning of serum.
  • the withdrawal of yeastolate is performed after the withdrawal of sodium pyruvate, NNEA and vitamins.
  • the avian embryonic stem cells according to step a) of the invention are collected from avian embryo at oviposition, that is to say when the egg is laid.
  • oviposition corresponds to the following development stages according to Eyal-Giladi's classification (EYAL-GILADI's classification: EYAL-GILADI and KOCHAN, 1976 , ⁇ From cleavage to primitive streack formation: a complementary normal table and a new look at the first stages of the development in the chick >>.
  • EYAL-GILADI's classification EYAL-GILADI and KOCHAN
  • the duck embryonic stem (ES) cells of step a) are obtained by dissociating Pekin duck embryo(s) at around stage VIII (oviposition) of Eyal-Giladi's classification. If the laid egg collected at oviposition is not enough developed to collect embryonic stem cells, the laid egg may be further incubated between several hours (overnight) to one to two days to mature the embryo.
  • the duck embryonic stem (ES) cells of step a) is from a Muscovy duck. At oviposition, Muscovy duck is not enough mature because it is around stage VII, therefore, the egg is incubated overnight to mature the egg up to stage VIII to X of Eyal-Giladi's classification.
  • the chicken embryonic stem (ES) cells preferably from ev-0 chicken strain, of step a) is obtained by dissociating embryo(s) at around stage X (oviposition) of Eyal-Giladi's classification.
  • the avian embryonic stem cells according to step a) of the invention are collected from embryo before oviposition.
  • the main limitations encountered before oviposition is the fact that the egg has to be surgically removed from hens and that the amount of ES cells per embryo is less important.
  • ES cells are not well individualized rendering difficult in vitro culture of ES cells. A man skilled in the Art will be able to define the timeframe prior egg laying that allows to collect avian ES cells.
  • the avian embryonic stem cells according to step a) of the invention may be collected from avian embryo after oviposition up to hatching.
  • avian embryonic stem cells will progressively enter into differentiation to generate differentiated tissues; therefore, it is preferred to collect avian ES not to long after the lay.
  • a man skilled in the Art will be able to define the timeframe after egg laying that allows to collect avian embryonic stem cells.
  • the cells of step a) are a population of embryonic stem cells enriched in primordial germ cells (PGC). More preferably, the avian ES cells of step a) are purified PGCs.
  • PGC primordial germ cells
  • Primordial Germ Cells arise from the central region of the blastoderm (Ginsburg and Eyal-Giladi, 1987 Development 101(2):209-19; Karagenc et al, 1996 Dev Genet. 19(4):290-301; Petitte et al, 1997 Poultry Sci. 76(8):1084-92). Then they move to an anterior, extra-embryonic site, the germinal crescent until collected by the vasculature between 2.5 and 5 days of embryonic development to reach the germinal ridge.
  • PGCs are collected from embryonic blood collected from the dorsal aorta of a chicken embryo at stage 12-14 of Hamburger & Hamilton's classification (Hamburger & Hamilton 1951 A series of normal stages in the development of chick embryo. J. Morphol. 88: 49-92).
  • PGCs were collected from the germinal crescent by mechanical dissection of chicken embryo or from the gonads.
  • others methods for isolating PGCs are known and can alternatively be used.
  • avian embryonic stem cells are characterized by a slow doubling time comprises between 48 to 72 hours in culture at 39° C.
  • the defined cell culture conditions of avian ES cells followed by the progressive weaning in grow factors, feeder layer, additives and serum allow to adapt and select cells that maintain most of the desirable feature of ES cells (stability of karyotype, indefinite proliferation, expression of ES markers) but in addition display industrial-friendly characteristics like growth in suspension up to high cell densities in serum-free medium.
  • Telomerase constitutes one of the most important ES markers. Due to the sustained and maintained telomerase expression over the cell passages, EBx® cell are continuous (i.e immortal) but in addition are genetically stable (i.e diploid).
  • the present invention provides a process for obtaining continuous diploid avian cell lines derived from ES cells, wherein said avian cell lines do not produce replication competent endogenous retroviral particles, said process comprising the following steps of:
  • the present invention relates to a process for obtaining continuous diploid avian cell lines, named EBx®, derived from avian embryonic stem cells (ES), wherein said avian cell lines do not produce replication-competent endogenous retrovirus particles, and said process comprising the steps of:
  • Step j) of adapting adherent avian EBx® cell lines to suspension culture conditions, when carried out, can be effected in another preferred embodiment before the step g) of progressively decreasing the concentration of mammalian serum in the culture medium.
  • the basal culture medium in step b) of the process for obtaining continuous diploid avian cell lines according to the present invention is further supplemented with a growth factor selected in the group comprising interleukin 6 (IL-6), interleukin 6 receptor (IL-6R), Stem cell Factor (SCF) and Fibroblast Growth Factor (FGF), and the said process further comprises a step d) of:
  • IL-6 interleukin 6
  • IL-6R interleukin 6 receptor
  • SCF Stem cell Factor
  • FGF Fibroblast Growth Factor
  • step e) of withdrawing IGF-1 and CNTF from the culture medium is effected after the step d) of withdrawing all the growth factors selected from the group comprising IL-6, IL-6R, SCF, FGF from the culture medium.
  • basal culture medium meant a culture medium with a classical media formulation that allows, by itself, at least cells survival, and even better, cell growth.
  • basal media are BME (basal Eagle Medium), MEM (minimum Eagle Medium), medium 199, DMEM (Dulbecco's modified Eagle Medium), GMEM (Glasgow modified Eagle medium), DMEM-HamF12, Ham-F12 and Ham-F10, Iscove's Modified Dulbecco's medium, MacCoy's 5A medium, RPMI 1640, GTM3.
  • Basal medium comprises inorganic salts (for examples: CaCl 2 , KCl, NaCl, NaHCO 3 , NaH 2 PO 4 , MgSO 4 , . . . ), amino-acids, vitamins (thiamine, riboflavin, folic acid, D-Ca panthothenate, . . . ) and others components such as glucose, beta-mercapto-ethanol, sodium pyruvate.
  • Basal medium is a synthetic medium. Table 1 gives the composition of DMEM/HAM F12:
  • basal medium of the invention may be complemented with additives selected in the following group:
  • the basal medium is preferably complemented with protein hydrolyzate of non-animal origin.
  • Protein hydrolyzates of non-animal origin are selected from the group consisting bacteria tryptone, yeast tryptone, plant hydrolyzates, such as soy hydrolyzates, or a mixture thereof.
  • the protein hydrolyzates of non-animal origin is yeast hydrolyzate.
  • the term “hydrolyzate” includes an enzymatic digest of soy peptone or yeast extract.
  • the hydrolysate can be obtained from a plurality of soy peptone or yeast extract preparations, respectively, which can be further enzymatically digested (for example, by papain), and/or formed by autolysis, thermolysis and/or plasmolysis.
  • Hydrolysates also may be obtained commercially, such as Yeastolate, Hy-Soy, Hy-Yeast 412 and Hi-Yeast 444, from sources such as SAFC BioSciences (formerly JRH) (Lenaxa, K A), Quest International (Norwich, N.Y.), OrganoTechnie S.A. (France) or Deutsche Hefewerke GmbH (Germany). Sources of yeast extracts also are disclosed in WO 98/15614.
  • Sources of yeast extracts and soy hydrolysates also are disclosed in WO00/03000.
  • the hydrolysates used in media of the invention are preferably purified from a crude fraction, because impurities which could interfere with efficient cultivation are preferably eliminated during this purification, thereby improving the consistency of the hydrolysate.
  • Purification can be by ultrafiltration or Sephadex chromatography (for example, with Sephadex G25 or Sephadex G10 or equivalent materials), ion-exchange chromatography, affinity chromatography, size exclusion chromatography or “reversed-phase” chromatography.
  • purification is performed by ultrafiltration utilizing a 10 kDa cut-off filter. These processes are known in the field.
  • fractions can be selected which contain soy or yeast hydrolysate of defined molecular weight.
  • the average molecular weights of the soy and yeast hydrolysates are preferably between about 220 and 375 daltons.
  • yeast hydrolyzate is present in the cell culture medium.
  • Yeast hydrolyzate 50 ⁇ (around 200 g/l) obtained for example from SAFC-BIOSCIENCES (Ref 58902C) is present in the cell culture medium at a final concentration comprises between around 0.1 ⁇ to 2 ⁇ , preferably around 0.5 ⁇ to around 1 ⁇ into the culture medium. Soy hydrolyzate may also be added to the cell culture medium.
  • soy hydrolyzate and yeast hydrolyzate may be added to the cell culture medium as described in US2004/0077086.
  • a preferred basal medium of the invention is DMEM-HamF12 that are complemented with 2 mM L-glutamin, 1 mM sodium pyruvate, 1% non-essential amino-acids, vitamins 1%, 0.16 mM beta-mercapto-ethanol, and optionally with 1 ⁇ yeast hydrolyzate.
  • complete culture medium it is meant a basal culture medium complemented or not, preferably a basal synthetic medium, supplemented with at least one growth factor and animal serum.
  • complete culture medium is described in WO 03/076601, WO 05/007840, EP 787180, U.S. Pat. No. 6,114,168, U.S. Pat. No. 5,340,740, U.S. Pat. No. 6,656,479, U.S. Pat. No. 5,830,510 and in Pain et al. (1996, Development 122:2339-2348).
  • the complete culture medium may a conditioned medium, preferably BRL conditioned medium.
  • BRL conditioned media is prepared according to art-recognized techniques, such as described by Smith and Hooper (1987, Dev. Biol. 121:1-9). BRL cells are available from ATCC accession number CRL-1442. Conditioned medium may be supplemented with exogenous growth factors and animal serum as described below.
  • growth factors as used herein meant growth factor necessary for the survival and the growth of the undifferentiated avian ES cells in culture in a basal culture medium. It is possible to schematically distinguish two families of growth factors: the cytokines and the trophic factors.
  • the cytokines are mainly cytokines whose action is through a receptor which is associated with the gp130 protein.
  • leukemia inhibitory factor (LIF) interleukin 11, interleukin 6, interleukin 6 receptor
  • CNTF Ciliary Neurotrophic factor
  • oncostatin and cardiotrophin have a similar mode of action with the recruitment at the level of the receptor of a specific chain and the combination of the latter with the gp130 protein in monomeric or sometimes hetero-dimeric form.
  • the trophic factors are mainly Stem cell Factor (SCF), Insulin Growth factor 1 (IGF-1) and Fibroblast Growth Factor (FGF), preferably basic FGF (bFGF) or human FGF (hFGF).
  • the complete culture medium according to the invention comprises basal culture medium, preferably basal synthetic medium, and at least one cytokine whose action is through a receptor which is associated with the gp130 protein and/or at least one trophic factors.
  • the complete culture medium according to the invention comprises basal medium and at least one growth factor selected in the group consisting of Leukemia Inhibitory factor (LIF), oncostatin, cardiotrophin, Insulin Growth factor 1 (IGF-1), Ciliary Neurotrophic factor (CNTF), Interleukin 6 (IL-6), interleukin 6 receptor (IL-6R), Stem cell Factor (SCF), Fibroblast Growth Factor (FGF), interleukin 11 (IL-11).
  • LIF Leukemia Inhibitory factor
  • IGF-1 Insulin Growth factor 1
  • IL-6 Interleukin 6
  • IL-6R interleukin 6 receptor
  • SCF Stem cell Factor
  • FGF Fibroblast Growth Factor
  • the complete culture medium is basal medium supplemented with animal serum and with at least IGF-1 and CNTF.
  • the complete culture medium is basal medium supplemented with animal serum and at least IGF-1, CNTF, IL-6 and IL-6R.
  • the complete culture medium is basal medium supplemented with animal serum and at least IGF-1, CNTF, IL-6, IL-6R, SCF, FGF.
  • the complete culture medium is a conditioned culture medium comprising growth factors (i.e expressed by BRL or STO cells for example) and optionally supplemented with at least one exogenous growth factors selected in the group comprising: LIF, IGF-1, CNTF, IL-6, IL-6R, SCF, FGF, IL-11.
  • the concentration of growth factors IGF-1, CNTF, IL-6, IL-6R, SCF, FGF, IL-11 in the basal medium or in the conditioned culture medium is comprised between about 0.01 to 10 ng/ml, preferably, 0.1 to 5 ng/ml, and more preferably about 1 ng/ml.
  • the culture medium of the invention may also comprise in addition antibiotics, such as for example, gentamicine, penicilline and streptomycine, to prevent bacterial contamination.
  • antibiotics such as for example, gentamicine, penicilline and streptomycine, to prevent bacterial contamination.
  • Antibiotics may be added to the culture medium at the early passages of ES cells culture. For example, gentamycin at a final concentration of 10 ng/ml, penicillin at a final concentration of 100 U/ml and streptomycin at a final concentration of 100 ⁇ g/ml may be added to the culture medium.
  • no antibiotics is added to the culture medium during the late steps of process of establishment of continuous diploid avian cell lines of the invention.
  • feeder cells are animal cells or cell lines cultured for the purpose of culturing avian ES cells.
  • the feeder cells can be substituted with extra-cellular matrix plus bound growth factors.
  • Feeder matrix will thereafter refers to either feeder cells or extra-cellular matrix.
  • a feeder matrix as used herein is constructed in accordance with procedures known in the art. As noted above, it is preferred that the feeder matrix be preconditioned. By the term “preconditioned” it is meant that the feeder matrix is cultured in the presence of media for a period of time prior to the depositing of cells originating from the blastoderm disk fertilized avian eggs in contact with the feeder matrix, e.g.
  • a feeder matrix is preconditioned by culturing the feeder matrix by itself for one to two days prior to the depositing of cells originating from the blastoderm disk fertilized avian eggs in contact with the feeder matrix.
  • the feeder cells preferably comprises mouse fibroblast cells. STO fibroblasts are preferred, but primary fibroblasts are also suitable. Also while the present invention has been described with respect to the use of mouse cell feeder matrices, it is contemplated that feeder matrices comprising cells from other murine species (e.g. rat); other mammalian species (e.g; ungulate, bovine, porcine species); or avian species (e.g.
  • feeder cells of the invention may be transfected with expression vector(s) allowing for example the constitutive expression of growth factors such as avian SCF in STO cells.
  • this “feeder” produces the factor in a form which is soluble and/or attached in the plasma membrane of the cells.
  • the culturing process of the present invention may optionally comprise establishing a monolayer of feeder cells.
  • Feeder cells are mitotically inactivated using standard techniques.
  • the feeder cells may be exposed to X or gamma radiation (e.g. 4000 Rads of gamma radiation) or may be treated with Mitomycin C (e.g.
  • Mono-layers may optionally be cultured to about 80% confluency, preferably to about 90% confluency, and more preferably about 100% confluency. While configuration of the feeder cells as a monolayer is the preferred configuration for the culture, any suitable configuration is contemplated to be within the scope of the present invention. Thus, for example, layers, mono-layers, clusters, aggregates or other associations or groupings of feeder cells are contemplated to fall within the scope of the present invention and are particularly contemplated to fall with the meaning of the term “matrix”.
  • the culture medium of the invention is supplemented with animal serum.
  • the animal serum preferably used is fetal animal serum. Fetal bovine serum is preferred. Also while the present invention has been described with respect to the use of fetal bovine serum, it is contemplated that animal serum comprising serum from other animal species (e.g. chicken, horse, porcine, ungulate, etc.) may also be used.
  • the final concentration of animal serum in the culture medium is comprises between approximately 1 to 25%, preferably between 5% to 20%, more preferably between 8% and 12%. In the preferred embodiment, the final concentration of animal serum in the culture medium is approximately 10%. According to a preferred embodiment, the culture medium comprises approximately 10% of fetal calf serum.
  • the bird of the present invention is selected in the Order of Anseriformes, and is preferably a duck, more preferably a Pekin Duck, and more preferably Pekin duck strain M14 or GL30.
  • the bird of the present invention is a Muscovy duck. Therefore, the instant invention provides a first process for obtaining continuous diploid duck cell lines derived from embryonic stem cells (ES), wherein said duck cell lines do not produce replication-competent endogenous retrovirus particles, and said process is comprising the steps of:
  • the animal serum concentration at step b) is preferably of 5 to 10%.
  • the concentration of with Insulin Growth factor 1 (IGF-1), Ciliary Neurotrophic factor (CNTF), interleukin 6 (IL-6), interleukin 6 receptor (IL-6R), Stem cell Factor (SCF) and Fibroblast Growth Factor (FGF) are preferably of about 1 ng/ml.
  • the instant invention also provides a second process for obtaining continuous diploid duck cell lines derived from embryonic stem cells (ES), wherein said duck cell lines do not produce replication-competent endogenous retrovirus particles, and said process is comprising the steps of:
  • the animal serum concentration at step b) is preferably of 5 to 10%.
  • the concentration of IGF-1, CNTF, IL-6, IL-6R, SCF and FGF are preferably of about 1 ng/ml.
  • the instant invention also provides a third process for obtaining continuous diploid duck cell lines derived from embryonic stem cells (ES), wherein said duck cell lines do not produce replication-competent endogenous retrovirus particles, and said process is comprising the steps of:
  • the animal serum concentration at step b) is preferably of 5 to 10%.
  • the concentration of with IGF-1 and CNTF are preferably of about 1 ng/ml.
  • the process of the invention may also comprises the additional step of adapting, duck EBx® cells to the growth in cell culture medium without protein hydrolyzate of non-animal origin, such as yeast hydrolyzates.
  • duck EBx® cell lines of the invention do not display reverse transcriptase activity by Q-PERT analysis. Moreover, no replication-competent endogenous retrovirus particles is produced by duck EBx® cells as demonstrated by co-culture experiments of duck EBx® cells of the invention with ALV replication competent cells, such as quail QT6 cells or chicken DF1 cells. In addition, transmission electronic microscopy (TEM) analysis also demonstrate the absence of replication-competent endogenous retrovirus particles in duck EBx® cells.
  • the duck EBx® cell line of the invention is selected among duck EB24, duck EB26 and duck EB66 as described hereinafter.
  • the bird of the present invention is selected in the Order of Galliformes and more preferably is a chicken, preferably an a ev-0 domestic chicken ( Gallus Gallus subspecies domesticus ). Therefore, the instant invention provides a process for obtaining continuous diploid ev-0 domestic chicken cell lines derived from embryonic stem cells (ES), wherein said ev-0 domestic chicken cell lines do not produce replication-competent endogenous ALV-E retrovirus particles, and said process is comprising the steps of:
  • the animal serum concentration at step b) is preferably of 5 to 10%.
  • the concentration of IGF-1, CNTF, IL-6, IL-6R, SCF and FGF are preferably of about 1 ng/ml.
  • the instant invention also provides a second process for obtaining continuous diploid ev-0 domestic chicken cell lines derived from embryonic stem cells (ES), wherein said ev-0 domestic chicken cell lines do not produce replication-competent endogenous ALV-E retrovirus particles, and said process is comprising the steps of:
  • the animal serum concentration at step b) is preferably of 5 to 10%.
  • the concentration of with IGF-1, CNTF, IL-6, IL-6R, SCF and FGF are preferably of about 1 ng/ml.
  • the instant invention also provides a third process for obtaining continuous diploid ev-0 domestic chicken cell lines derived from embryonic stem cells (ES), wherein said ev-0 domestic chicken cell lines do not produce replication-competent endogenous ALV-E retrovirus particles, and said process is comprising the steps of:
  • the animal serum concentration at step b) is preferably of 5 to 10%.
  • the concentration of with IGF-1 and CNTF are preferably of about 1 ng/ml.
  • the bird of the present invention is a domestic chicken ( Gallus Gallus subspecies domesticus ) obtained from a specific-pathogen-free (SPF) flock. More preferably, the chicken strain is White-Leghorn.
  • SPF chicken eggs has been screened for the absence of known chicken bacterial pathogens and viruses, including the reticuloendotheliosis virus (REV) and the avian exogenous leucosis virus (ALV-A, ALV-B, ALV-C, ALV-D, ALV-J).
  • the SPF egg of the invention may VALO eggs from LOHMANN (Cuxhaven, Germany) or L22 eggs from CHARLES RIVER (Spafas).
  • the instant invention also provides processes for obtaining continuous diploid chicken cell lines derived from embryonic stem cells (ES) obtained from SPF chicken eggs, like described with ev-0 chicken eggs.
  • ES embryonic stem cells
  • the chicken EBx® cell line obtained from SPF eggs is EBv13.
  • Chicken EBx® cell lines of the invention may display reverse transcriptase activity by Q-PERT analysis, but without producing replication-competent endogenous retrovirus particles.
  • the absence of replication-competent endogenous retrovirus particles may be demonstrated by co-culture experiments of chicken EBx® ev-0 cells of the invention with ALV replication competent cells, such as quail QT6 cells or chicken DF1 cells.
  • ALV replication competent cells such as quail QT6 cells or chicken DF1 cells.
  • the absence of endogenous retrovirus particles in chicken EBx® ev-0 cells may be also demonstrate by TEM.
  • the processes of the invention may also comprise the additional step of decreasing the cell culture temperature to 37° C. in order to adapt the avian cell lines of the invention to grow at 37° C.
  • the temperature adaptation is performed after feeder depletion and prior serum depletion.
  • the temperature adaptation is performed after the serum depletion step or after the step of adapting the cell lines to suspension culture.
  • the established lines EBx® of the invention have the characteristic to grow either as adherent cells or as suspension cells in a culture medium free of exogenous growth factors and animal serum and without feeder cells. Different techniques can be used alone or in combination to adapt cells to suspension culture, among them:
  • the EBx® cells preferably duck EBx® and chicken EBx® ev-0, can be in vitro cultured over a considerable period of time.
  • the adherent or anchorage-independent (i.e “suspension) EBx® cells obtained by the process of the invention are capable to proliferate for at least 50 generation, at least 75 generation, at least 100 generation, at least 125 generation, at least 150 generation, at least 175 generation, at least 200 generation, at least 250 generation.
  • the expression “line” is understood to mean any population of cells capable of proliferating indefinitely in culture in vitro while retaining to a greater or lesser degree the same morphological and phenotypic characteristics.
  • Clones may be obtained, for example by limit dilution, from EBx® cells of the invention. These clones are cells which are genetically identical to the cell from which they are derived by division.
  • the present invention also relates to the continuous diploid avian cell lines, named EBx®, obtainable by the process of the invention, said EBx® being small, round (i;e diameter around 10 um), individualized cells with a doubling time of around 30 hours or less at 37° C. or 39° C.
  • the avian EBx® cells preferably the duck EBx® or chicken EBx® ev-0, express an embryonic stem cell phenotype with the following characteristics:
  • Said cells do not produce replication competent endogenous retrovirus particles.
  • the avian EBx® cell lines of the invention are capable of proliferating indefinitely in a basal medium, in particular in a medium such as SAFC Excell media, DMEM, GMEM, DMEM-HamF12 or McCoy, free of exogenous growth factors, serum and/or inactivated feeder layer, optionally complemented with various additives commonly used by persons skilled in the art.
  • a medium such as SAFC Excell media, DMEM, GMEM, DMEM-HamF12 or McCoy
  • additives are non-essential amino acids, vitamins, sodium pyruvate and antibiotics.
  • Duck EBx® cells of the invention have the remarkable feature to grow in a basal culture medium that is not complemented with glutamine.
  • the present invention also relates to a cell culture medium to maintain pluri- or multipotent avian embryonic stem cells, preferably pluri- or multipotent duck embryonic stem (ES) cells, into culture in an undifferentiated state.
  • the present invention relates to cell culture medium for duck embryonic stem cells comprising a basal culture medium, supplemented with animal serum and supplemented with at least IGF-1 and CNTF.
  • the present invention relates to cell culture medium for duck embryonic stem cells comprising a basal culture medium supplemented with animal serum and supplemented with at least IGF-1, CNTF, II-6, II-6R.
  • the present invention relates to a cell culture medium for duck embryonic stem cells
  • a cell culture medium for duck embryonic stem cells comprising a basal culture medium supplemented with animal serum and supplemented with at least IGF-1, CNTF, II-6, II-6R, SCF and FGF.
  • Said media are sufficient for the maintenance of said duck ES cells into culture for at least 7 days, preferably for at least 20 days, preferably for at least 100 days in an undifferentiated state.
  • Said culture media of the invention may further comprise optionally at least one compound selected in the group comprising Interleukin-11, cardiotrophin, oncostatin and leukaemia inhibitory factor (LIF).
  • LIF leukaemia inhibitory factor
  • said culture media further comprise protein hydrolyzate of non-animal origin as previously described; more preferably it is yeast hydrolyzate at 1 ⁇ concentration.
  • the culture medium of avian (preferably duck) ES cells of the invention may further comprise a layer of feeder cells.
  • Said undifferentiated duck cells according to the invention are capable of maintaining said stem cell phenotype when grown on feeder cells in a cell culture medium for duck embryonic stem cells as previously described. Said undifferentiated duck cells are useful to produce chimeric or transgenic ducks.
  • the present invention also relates to a method of obtaining chimeric duck, said method comprising the steps of:
  • the present invention also relates to a method of obtaining genetically modified chimeric duck, comprising the steps of:
  • the present invention also relates to a method of obtaining a progeny of said chimeric duckling wherein said method comprises the following steps:
  • the invention may comprise the additional step of expressing an heterologous polypeptide encoded by an expression vector comprised in said genetically modified duck ES cells.
  • the heterologous polypeptide is delivered into biological fluid of duck, such as blood, sperm, urine, or the white of a developing avian egg produced by a female of the genetically modified duck.
  • the EBx® cells of the invention have all the above mentioned characteristics and are useful for the production of biologics such as viral vaccines and recombinant peptides and proteins.
  • the instant invention also provide a process of replicating a virus in the continuous diploid avian EBx® cell lines of the invention. More preferably, the invention provides a process of replicating a virus in the continuous diploid avian EBx® cell lines of the invention, preferably duck or chicken EBx® cell lines, that comprise the steps of:
  • said process comprises the steps of:
  • Said process of the invention may comprise the additional step of adding proteolytic enzyme in the culture medium in conditions that allow virus propagation.
  • the proteolytic enzyme is selected from the group consisting of trypsin, chymotrypsine, thermolysine, pepsine, pancreatine, papaIne, pronase, subtilisine A, elastase, furine and carboxypeptidase.
  • the enzyme is trypsin.
  • the proteolytic enzyme is a recombinant protein produced on a procaryotic host or on plants (i.e: trypzean).
  • the proteolytic enzyme may added before, during and/or after the virus infection.
  • the addition of proteolytic enzyme is performed after virus infection.
  • the addition of proteolytic enzyme in the culture medium may be performed one time per day, more than one time per day, or less than one time per day until the virus harvest.
  • virus as used herein includes not only naturally occurring viruses but also attenuated viruses, reassortant viruses, vaccine strains, as well as recombinant viruses and viral vectors derived thereof.
  • the virus of the invention are preferably selected from the group comprising poxviruses, orthomyxoviruses, paramyxoviruses, herpes viruses, hepadnaviruses, adenoviruses, parvoviruses, reoviruses, circoviruses, coronaviruses, flaviviruses, togaviruses, birnavriruses and retroviruses.
  • the viruses, the related viral vectors, viral particles and viral vaccines belong to the family of poxyiridae, and more preferably to the chordopoxyiridae.
  • the virus or the related viral vectors, viral particles and viral vaccines are a poxvirus, preferably an avipoxvirus selected among fowlpox virus (i.e TROVAC), canarypox virus (i.e ALVAC), juncopox virus, mynahpox virus, pigeonpox virus, psittacinepox virus, quailpoxvirus, sparrowpoxvirus, starling poxvirus, turkeypox virus.
  • fowlpox virus i.e TROVAC
  • canarypox virus i.e ALVAC
  • juncopox virus mynahpox virus
  • pigeonpox virus psittacinepox virus
  • quailpoxvirus sparrowpoxvirus
  • the virus is a vaccinia virus selected among Lister-Elstree vaccinia virus strain, modified vaccinia virus such as Modified Vaccinia virus Ankara (MVA) which can be obtained from ATCC (ATCC Number VR-1508), NYVAC (Tartaglia et al., 1992, Virology, 188:217-232), LC16 m8 (Sugimoto et Yamanouchi, 1994, Vaccine, 12:675-681), CVI178 (Kempe et al., 1968, Pediatrics 42:980-985) and other recombinant or non-recombinant vaccinia virus.
  • MVA Modified Vaccinia virus Ankara
  • influenza virus belongs to the family of ortho-myxoviridae, in particular influenza virus.
  • the influenza virus is selected from the group consisting of human influenza virus, avian influenza virus, equine influenza virus, swine influenza virus, feline influenza virus.
  • Influenza virus is preferably selected in strains A, B and C. Among strains A, one can recite viruses with different subtypes of haemagglutinin and neuraminidase, such as without limitation H1N1, H2N2, H3N2, H4N2, H4N6, H5N1, H5N2, H7N7 et H9N2.
  • H1N1 strains one can recite A/Porto Rico/8/34, A/New Calcdonia/20/99, A/Beijing/262/95, A/Johannesburg/282/96, A/Texas/36/91, A/Singapore, A/Solomon Islands/03/2006.
  • strains H3N2 one can recite A/Panama/2007/99, A/Moscow/10/99, A/Johannesburg/33/94, A/Wisconsin/10/04.
  • influenza Virus of the invention is selected among wild type virus, primary viral isolate obtained from infected individual, recombinant virus, attenuated virus, temperature sensitive virus, low-temperature adapted virus, reassortant virus, reverse genetic engineered virus.
  • the process of the invention comprises the additional step of adding proteolytic enzyme in the culture medium in conditions that allow virus propagation.
  • the enzyme is trypsin.
  • the final concentration of trypsin in cell culture medium is comprises between around 0.01 ⁇ g/ml up to 10 ⁇ g/ml. More preferably, the final concentration of trypsin in cell culture medium is comprised between 0.01 to 10 usp/ml (usp: US pharmacopea unit) preferably around between 0.05 to 2 usp/ml, more preferably around between 0.3 to 1 usp/ml and more preferably around 0.75 usp/ml.
  • the viruses, the related viral vectors, the viral particles and vaccines belong to the family of paramyxoviridae.
  • the virus is a naturally occurring paramyxovirus or a recombinant paramyxovirus selected in the group comprising measles virus, mumps virus, rubella virus, Sendai virus, Respiratory Syncythial virus (RSV), human para-influenza types I and III, Rinderpest virus, canine distemper virus, Newcastle disease virus, duck para-influenza virus.
  • the virus is measles virus or a recombinant measles virus.
  • the virus is Newcastle Disease virus (NDV) or a recombinant NDV.
  • Example of NDV strain is LaSota strain.
  • the process of the invention comprises preferably the additional step of adding proteolytic enzyme in the culture medium in conditions that allow virus propagation.
  • the enzyme is trypsin.
  • the final concentration of trypsin in cell culture medium is comprises between around 0.01 ⁇ g/ml up to 10 ⁇ g/ml. More preferably, the final concentration of trypsin in cell culture medium is comprised between 0.01 to 10 usp/ml (usp: US pharmacopea unit) preferably around between 0.3 to 1 usp/ml, more preferably around between 0.4 to 0.75 usp/ml.
  • the EBx® cell lines of the invention that may grow in adherence are useful to perform virus titration, and preferably NDV titration, on a plaque assay.
  • virus growth in EBx® cells leads to the formation of characteristic giant cells.
  • NDV viral particles may be determined by haemagglutination assay. Therefore, the invention also pertain to the use of EBx® cells of the invention for the titration of virus, such as NDV virus.
  • the viruses, the related viral vectors, the viral particles and vaccines belong to the family of togaviridae.
  • the virus is a naturally occurring alphavirus or a recombinant alphavirus selected in the group comprising Sinbis virus, Semliki forest virus, O'nyong'nyong virus, Chikungunya virus, Mayaro virus, Ross river virus, Eastern equine encephalitis virus, Western Equine encephalitis virus, Venezuelan Equine encephalitis virus.
  • the viruses, the related viral vectors, the viral particles and vaccines belong to the family of herpesviridae.
  • the virus is a naturally occurring Marek Disease virus or a recombinant Marek Disease virus.
  • the Marek Disease virus (MDV) is preferably selected among the license vaccine strains of MDV such as: FC126 (HTV), SB-1, 301B/1, CV1988 Clone C, CV1988/C/R6, CVI988/Rispens, R2/23 (Md11/75).
  • the viruses, the related viral vectors, the viral particles and vaccines belong to the family of hepadnaviridae.
  • the virus is a naturally occurring naturally occurring hepadnavirus or a recombinant hepadnavirus, preferably selected among avian and human hepadnavirus.
  • the avian hepadnavirus is preferably selected among the group consisting of duck hepatitis B virus (DHBV), heron hepatitis B virus (HHBV) and snow goose (SGHBV).
  • viruses, the related viral vectors, the viral particles and vaccines belong to the family of birnaviridae, in particular Infectious Bursal Disease virus.
  • the viruses, the related viral vectors, the viral particles and vaccines belong to the family of flaviviridae, in particular Dengue virus, Japanese encephalitis virus and West Nile virus.
  • viruses, the related viral vectors, the viral particles and vaccines belong to the family of coronaviridae, in particular Infectious Bronchitis virus.
  • viruses, the related viral vectors, the viral particles and vaccines belong to the family of circoviridae, in particular Chicken Anemia virus.
  • the viruses, the related viral vectors, the viral particles and vaccines belong to the family of retroviridae.
  • the virus is a naturally occurring retrovirus selected among reticulo-endotheliosis virus, duck infectious anemia virus, suck spleen necrosis virus, or a recombinant retrovirus thereof.
  • the viruses, the related viral vectors, the viral particles and vaccines belong to the family of parvoviridae.
  • the virus is a naturally occurring parvovirus such as duck parvovirus or a recombinant parvovirus thereof.
  • the viruses, the related viral vectors, the viral particles and vaccines belong to the family of adenoviridae.
  • the virus is a naturally occurring adenovirus preferably selected among fowl adenovirus, goose adenovirus, duck adenovirus and pigeon adenovirus or a recombinant adenovirus thereof.
  • Fowl adenovirus examples include Fowl adenovirus 1 (CELO), Fowl adenovirus 5 (340), Fowl adenovirus 4 (KR95), Fowl adenovirus 10 (CFA20), Fowl adenovirus 2 (P7-A), Fowl adenovirus 3 (75), Fowl adenovirus 9 (A2-A), Fowl adenovirus 11 (380), Fowl adenovirus 6 (CR119), Fowl adenovirus 7 (YR36), Fowl adenovirus 8a (TR59) Fowl adenovirus 8b (764) and Egg prop Syndrome virus.
  • CELO Fowl adenovirus 1
  • Fowl adenovirus 5 340
  • Fowl adenovirus 4 KR95
  • Fowl adenovirus 10 CFA20
  • Fowl adenovirus 2 P7-A
  • Examples of Goose adenovirus are Goose adenovirus 1, Goose adenovirus 2, Goose adenovirus 3.
  • Example of Duck adenovirus is Duck adenovirus 2.
  • Example of Pigeon adenovirus is Pigeon adenovirus 1.
  • Recombinant viruses include but are not limited to viral vectors comprising a heterologous gene.
  • a helper function(s) for replication of the viruses is provided by the host cell EBx®, a helper virus, or a helper plasmid.
  • Representative vectors include but are not limited to those that will infect avian or mammalian cells.
  • the instant invention also relates to the use of EBx® cells of the invention to replicate intracellular bacteria such as Chlamydia, Rickettsia or Coxiella.
  • the EBx® cells of the invention may also be used to produce recombinant proteins and peptides.
  • the invention also relates to a method of production of recombinant proteins and peptides, that include the steps of: (i) genetically modifying the EBx® cells of the invention by transient or stable transfection of an expression vector; (ii) optionally, selecting EBx® cells expressing said recombinant proteins or peptides; (iii) and purification of said peptides or proteins.
  • Peptides and proteins produced in EBx® cells are also included in the present invention.
  • the cultivation vessel of the invention is more preferably selected among continuous stirred tank bioreactor, WaveTM Bioreactor, BelloTM bioreactor, spinner flask, flask and a cell factory.
  • cells are scaled-up from a master or working cell bank vial through various sizes of T-flasks, roller bottles or WaveTM Bioreactor and, preferably, finally to bioreactors.
  • the resulting cell suspension is then typically fed into a seed production bioreactor (typically 20-30 L volume) for further cultivation, and in some embodiments, to a larger production bioreactor (typically 150-180 L volume and above).
  • the ratio of volume of the second (larger) bioreactor to the seed bioreactor depends upon the degree to which the cell line is propagated in the first bioreactor, but is typically from 3:1 to 10:1, e.g., in the range of (6-8):1.
  • the cultivation vessel is a continuous stirred tank bioreactor that allows control of temperature, aeration, pH and other controlled conditions and which is equipped with appropriate inlets for introducing the cells, sterile oxygen, various media for cultivation and outlets for removing cells and media and means for agitating the culture medium in the bioreactor.
  • “serum-free medium” meant a cell culture medium ready to use, that is to say that it does not required animal serum addition allowing cells survival and cell growth.
  • This medium is not necessary chemically defined, and may contained hydrolyzates of various origin, from plant or yeast for instance.
  • said SFM are “non animal origin” qualified, that is to say that it does not contain components of animal or human origin (FAO status: “free of animal origin”).
  • FEO status “free of animal origin”.
  • the native serum proteins are replaced by recombinant proteins.
  • SFM medium according to the invention does not contain protein (PF medium: “protein free medium”) and/or are chemically defined (CDM medium: “chemically defined medium”).
  • SFM media present several advantages: (i) the first of all being the regulatory compliance of such media (indeed there is no risk of contamination by adventitious agents such as BSE, viruses); (ii) the optimization of the purification process; (iii) the better reproducibility in the process because of the better defined medium.
  • SFM media examples include: VP SFM (InVitrogen Ref 11681-020, catalogue 2003), Opti Pro (InVitrogen Ref 12309-019, catalogue 2003), Episerf (InVitrogen Ref 10732-022, catalogue 2003), Pro 293 S-CDM (Cambrex ref 12765Q, catalogue 2003), LC17 (Cambrex Ref BESP302Q), Pro CHO 5-CDM (Cambrex ref12-766Q, catalogue 2003), HyQ SFM4-CHO (Hyclone Ref SH30515-02), HyQ SFM4-CHO-Utility (Hyclone Ref SH30516.02), HyQ PF293 (Hyclone ref SH30356.02), HyQ PF Vero (Hyclone Ref SH30352.02), Excell 293 medium (SAFC Biosciences ref 14570-1000M), Excell 325 PF CHO Protein free medium (SAFC Biosciences ref 14335-1000M),
  • the serum-free medium N o 1 and the serum-free medium N o 2 are the same medium.
  • the serum-free medium N o 1 and the serum-free medium N o 2 have a different composition.
  • the process of the invention encompasses the removal of the whole or a part of serum-free medium 1, followed by its replacement by serum-free medium N o 2. However, it is more convenient to remove a substantial fraction (e.g., up to about 50%) of the serum-free medium 1 and then replenish it with the serum-free medium N o 2 while still removing medium 1, e.g., through the spinfilter.
  • serum-free medium N o 2 is directly added to serum-free medium N o 1 without removal of a part of serum-free medium N o 1. Between 0.25 to 10 volumes of serum-free medium N o 2 is added to 1 volume of serum-free medium N o 1.
  • between around 0.5 to 8 volumes of serum-free medium N o 2 is added to 1 volume of serum-free medium N o 1. In a more preferred embodiment, between around 3 to 6 volumes of serum-free medium N o 2 is added to 1 volume of serum-free medium N o 1.
  • the serum-free medium N o 1 and/or the serum-free medium N o 2 may be supplemented with at least one ingredient selected from the group consisting of amino-acids, lipids, fatty acids, cholesterol, vitamins, carbohydrates, protein hydrolyzates of non-animal origin, and a mixture thereof.
  • the process of replicating a virus of the invention is a fed-batch process that comprises the additional step of feeding the cells with at least one ingredient selected from the group consisting of amino-acids, lipids, vitamins, carbohydrates, protein hydrolyzates of non-animal origin, surfactant and a mixture thereof.
  • the feeding occurs during steps a) to d) of the process of the invention of replicating a virus, alternatively only during the steps b) to d), or alternatively only during the steps d).
  • the feeding may occur either on a daily basis or on a continuous basis. When the feeding is discontinuous, the feeding may occur one time per day, more than one time per day, or less than one time per day.
  • the SFM media of the invention comprise a number of ingredients, including amino acids, vitamins, organic and inorganic salts, sources of carbohydrate, each ingredient being present in an amount which supports the cultivation of a cell in vitro.
  • additional ingredients are added to SFM media.
  • the choice of amino-acid(s) to add to the cell culture may be determined be an analysis of amino-acids consumption by the cells in the culture; such consumption varies according to cell species.
  • the amino-acids added to the medium may be selected from the group consisting of asparagine and glutamine, or a mixture thereof.
  • glutamine is added for chicken EBx cell culture and the feeding of glutamine is performed during step a) to d) to maintain the glutamine concentration in the medium between around 0.5 mM to around 5 mM, preferably between around 1 mM to around 3 mM, and most preferably around 2 mM.
  • the feeding of glutamine occur on a continuous basis.
  • duck EBx® cells do not consume much glutamine, because duck cells have the ability to synthetize glutamine. Therefore, glutamine may or may not be added for duck EBx cell culture.
  • the carbohydrates added to the medium are selected from the group consisting of D-glucose, D-sucrose and D-galactose or a mixture thereof.
  • the carbohydrate added is D-glucose.
  • the feeding of D-glucose is performed during step a) to d), more preferably between b) to d) to maintain the D-glucose concentration in the medium between around 0.5 g/l to 25 g/l of D-glucose, preferably between around 1 g/l to 10 g/l of D-glucose, preferably around 2 to 3 g/l of D-glucose.
  • the feeding of D-glucose occur on a continuous basis.
  • the lipids are selected from the group consisting of cholesterol, steroids, and fatty acids such as palmitic acid, palmitoleic acid, stearic acid, oleic acid, linoleic acid, linolenic acid, and their derivatives, or a mixture thereof. More preferably the fatty acids are from SIGMA-ALDRICH (Ref. F7050) and around 0.35 ⁇ l/ml of fatty acids solution is added to the culture medium.
  • fatty acids are from SIGMA-ALDRICH (Ref. F7050) and around 0.35 ⁇ l/ml of fatty acids solution is added to the culture medium.
  • the medium may contain auxiliary substances, such as buffer substances like sodium bicarbonate, oxidation stabilizers, stabilizers to counteract mechanical stress, or protease inhibitors.
  • auxiliary substances such as buffer substances like sodium bicarbonate, oxidation stabilizers, stabilizers to counteract mechanical stress, or protease inhibitors.
  • a non-ionic surfactant such as polypropylene glycol (PLURONIC F-61, PLURONIC F-68, SYNPERONIC F-68, PLURONIC F-71 or PLURONIC F-108) can be added to the medium as a de-foaming agent.
  • PLURONIC F-61, PLURONIC F-68, SYNPERONIC F-68, PLURONIC F-71 or PLURONIC F-108 can be added to the medium as a de-foaming agent.
  • the quantity of nonionic surfactant is preferably between about 0.05 and about 10 g/L, typically between about 0.1 and about 5 g/L.
  • the concentration of surfactant in cell culture medium may be modified to adapt (i.e increase or decrease) the size of the cell clumps.
  • the addition of serum-free medium N o 2 to the cell culture is performed after infection step b), preferably between around 0.5 to 4 hour after step b), and more preferably around 1 hour after step b).
  • the addition of serum-free medium N o 2 to the cell culture is performed before infection step b), preferably between around 0.5 to 4 hour after step b), and more preferably around 1 hour before step b).
  • the addition of serum-free medium N o 2 to the cell culture is performed simultaneously to infection step b.
  • the viral infection of step b) is carried out at an m.o.i (multiplicity of infection) of about 10 to 10 ⁇ 8 , preferably 10 ⁇ 1 to 10 ⁇ 6 , more preferably about 10 ⁇ 2 to 10 ⁇ 6 , and more preferably about 10 ⁇ 4 .
  • the man skilled in the art will determine the optimal m.o.i according to the virus type.
  • the infected cells are preferably cultured during at least 24 h, at least 48 h, at least 72 h, at least 96 h, at least 120 h, at least 144 h. When the virus is a poxvirus, the infected cells are cultured at least 144 h.
  • the cell culture of step a) is carried out by batch culture, repeated batch culture, fed-batch culture or perfusion culture. More preferably, the cell culture of step a) is performed by fed-batch culture.
  • the infection in step b) is performed when the cell density is at least around 4 million, preferably 6 million cells/ml, more preferably 8 million cells/ml in batch or fed-batch process.
  • the infection in step b) is performed when the cell density is of at least at least 8 million cells/ml, preferably around 9 to 10 million cells/ml, or even higher.
  • the pH of the serum-free culture medium in steps a), b), c) and d) is preferably monitored by the bioreactor.
  • the pH shall be in a range from 6.5 to 7.8, preferably around 6.8 to 7.5, and more preferably around 7.2.
  • step d) lasts for 1 to 10 days before the harvest.
  • step d) lasts for 2 to 5 days before the harvest.
  • the time of harvest (step e) is defined according to the cell density in the cultivation vessel. The inventor have now found that the optimal time for harvest the viruses is two days after the density of viable cells have reached its optimal level and have started to decrease because of viral infection.
  • the cell culture is performed at a temperature comprises between 32° C. to 39° C. depending of the virus type.
  • cell culture infection is preferably performed at 33° C.
  • EBx® cells have the ability to grow in suspension culture with cells clumped in loose aggregates of few cells, up to more than hundred(s) of cells.
  • the size of the clumps may vary according to the composition of cell culture medium. For example, presence of surfactant such as polypropylene glycol (PLURONIC F-61, PLURONIC F-68, SYNPERONIC F-68, PLURONIC F-71 or PLURONIC F-108), the stirring, the concentration of divalent ions, such as Mg2+ and Ca2+, may have an effect on the clumps size.
  • surfactant such as polypropylene glycol (PLURONIC F-61, PLURONIC F-68, SYNPERONIC F-68, PLURONIC F-71 or PLURONIC F-108)
  • the stirring the concentration of divalent ions, such as Mg2+ and Ca2+, may have an effect on the clumps size.
  • the viral yield may be increased by allowing the EBx® cells of the invention to aggregate to each others to form clumps during at least step a) of the process.
  • the suspension cells are generally passaged to a larger vessel, either by dilution into fresh medium or by centrifugation followed by a re-suspension of cell pellet into a fresh medium.
  • the inventor has found that during the cells passages, it is recommended to keep large cell clumps into the culture. To do so, it is better not to disrupt cells clumps in order to improve the replication of virus in EBx® cells.
  • step a) in T-flasks or roller-bottles, it is recommended to dilute the cell culture to passage the cells into larger vessel(s), and it is not recommended to centrifuge, nor to disrupt the cells clumps by pipetting or stirring.
  • too large clumps may be suboptimal for a high viral production. Consequently, the man skilled in the art will define whether a partial disruption of the clumps, by pipetting or stirring, during initial cell passages of step a) may improve viral yield.
  • poxviruses and preferably MVA, ALVAC and Fowlpox viruses are obtained by a process of the invention that include the step a) of proliferating clumped EBx® in loose aggregates of few cells, up to more than at least one hundred of cells, at least two hundred of cells, at least five hundred of cells, at least thousand(s) of cells.
  • size of EBx® cells clumps may be dependent of Mg2+ and/or Ca2+ ions concentration in anchorage-independent cell culture medium. Since too large clumps may be suboptimal for a high viral production, the size of clumps may be monitored by adjusting Mg2+ and Ca2+ concentration in cell culture medium.
  • the cell culture medium preferably contain Mg2+ concentration comprises between 0.5 mM and 2.5 mM, preferably around 1.6 mM, and Ca2+ concentration comprises between 0.01 mM and 0.5 mM, preferably around 0.1 mM.
  • the invention also relate to the virus obtainable by a process of the invention.
  • the instant invention also relates to the vaccine containing the virus of the invention.
  • the process of manufacturing a viral vaccine comprises the process of replicating a virus according to the invention wherein the step e) of virus harvest is comprising at least one step selected among filtering, concentrating, freezing and stabilizing by addition of stabilizing agent.
  • the virus harvest is performed according to technologies well-known to the man skilled in the art.
  • the step of harvesting said virus comprises collecting cell culture supernatant obtained from centrifugation of cell culture, then filtering, concentrating, freezing and stabilizing virus preparation by addition of stabilizing agent.
  • the process of manufacturing a viral vaccine according to the invention may also comprise the additional step of inactivation of harvested virus.
  • Inactivation is preferably performed by treatment with formaldehyde, beta-propiolactone, ether, ether and detergent (i.e such as Tween 80TM), cetyl-trimethyl ammonium bromide (CTAB) and Triton N102, sodium deoxycholate and tri(N-butyl)phosphate.
  • the invention also relates to a process of preparation of viral antigenic proteins from the virus obtainable by a process of the invention, said process comprises the additional steps of:
  • the virus in the vaccine may be present either as intact virus particles, or as disintegrated virus particles.
  • the vaccine is a killed or inactivated vaccine.
  • the vaccine is a live attenuated vaccine wherein said vaccines mainly comprises EBx cell culture supernatant obtainable by the process of the invention, preferably without serum, optionally filtered and/or concentrated and comprising said virus.
  • the vaccine is comprising viral antigenic proteins obtainable from a virus prepared according to the process of the invention.
  • the invention also pertain to provide a vaccine comprising an infected cell line EBx®, preferably duck or ev-0 chicken EBx®, obtainable by the process of the invention, and wherein infected cell line EBx®, preferably preferably duck or ev-0 chicken EBx®, are harvested in step d).
  • infected cell line EBx® preferably duck or ev-0 chicken EBx®
  • the vaccine of the invention may comprise the virus of the invention in combination with pharmaceutically acceptable substances which increase the immune response.
  • substances which increase the immune response comprises incomplete Freund adjuvant, saponine, aluminium hydroxide salts, lysolecithin, plutonic polyols, polyanions, peptides, bacilli Calmette-Guerin (BCG) and corynebacterium parvum .
  • BCG Bacilli Calmette-Guerin
  • Example of synthetic adjuvant is QS-21.
  • immuno-stimulating proteins immuno-stimulating proteins (interleukins II1, II2, IL3, IL4, IL12, IL13, granulocyte-macrophage-colony-stimulating factor, . . . ) may be used to enhance the vaccine immune response.
  • the vaccine of the invention is preferably a liquid formulation, a frozen preparation, a dehydrated and frozen preparation, optionally adapted to intra-nasal route of administration.
  • the vaccine of the invention is use for the prophylactic and/or therapeutic treatment of a human or an animal infected by a virus previously listed.
  • the viral vaccine of the invention is preferably use for the prophylactic and/or therapeutic treatment of a human infected by a virus selected among smallpox, influenza, measles, mumps, rubella viruses, RSV.
  • the vaccine of the invention is preferably use for the prophylactic and/or therapeutic treatment of a animal infected by a virus selected among influenza, Newcastle Disease Virus, Egg prop Syndrome Virus, Infectious Bursal Disease, Infectious Bronchitis Virus, Canine Distemper virus, Chicken Anemia Virus.
  • the recombinant viral vaccine of the invention may also be used for the prophylactic and/or therapeutic treatment of chronic diseases such as cancer and infectious diseases such as AIDS.
  • the EBx® cell lines of the invention are useful to generate and produce re-assorted virus.
  • the virus with a segmented genome such as influenza virus may be re-assorted.
  • a mix of segmented genome from two different strains is present in the same host cell.
  • Specific reassorted virus may thus be isolated by selecting or eliminating, with an antibody for example, virus with a desired traits (See Kilnourne E. D in Plotkin S A and Mortimer E. A. Eds, Vaccines 1994).
  • the EBx® cell lines of the invention are also usefull to generate and produce influenza virus by reverse genetics (See Enami, Proc. Natl. Acad. Sci. USA, 87:3802-3805 (1990); Enami et Palese, J. Virol. 65:2511-2513 (1991); Luytjes, Cell 59:1107-1113 (1989)).
  • the present invention also relates to the use of EBx® cell lines of the invention as a cell substrate to perform virus titration.
  • EBx® cells will efficiently in replace current cell system, such as embryonated eggs, CEFs, DF1 cells and others, used to determine the titer of a viral solution.
  • the viral titration is performed by TCID50 method (Reed L, Muench H, 1938. A simple method of estimating fifty percent endpoints. Am. J. Hyg. 27, 493-97).
  • the present invention also relates to the use of EBx® cell lines of the invention as a cell substrate to perform sanitary testing.
  • the invention also relates to the diagnostic composition containing viruses of the invention or constituents thereof.
  • FIG. 1 Anchorage-Independent Chicken EBx Cells
  • FIG. 1A Anchorage-Independent Chicken Valo EBv13 Cells in Serum Free-Medium.
  • EBv13 cells were cultured at 37° C. in suspension serum-free medium Excell 65319 (SAFC). EBv13 cells have an homogeneous size and grow in loose clumps into culture. The population doubling time is about 16-18 Hours and the cell density reached in agitated flask vessels were about 4-5 millions cells/ml.
  • SAFC suspension serum-free medium Excell 65319
  • FIG. 1B Anchorage-Independent Chicken EB Line 0 Cells in Serum Free-Medium
  • EB Line 0 cells were cultured at 39° C. in suspension serum-free medium Excell 66444 (SAFC). EB Line 0 cells have an homogeneous size and grow in loose clumps.
  • FIG. 2 Chicken Valo EBv13 Cells Express High Level of Telomerase
  • EBv13 cells at passage p 193 do express high level of telomerase in the same order of magnitude that chicken EB14-074 cells (see WO03/076601) at passage p 164 (Master cell Bank: MCB) or at passage p 184 (Workin Cell bank: WCB).
  • Murine embryonic stem cells (mES) were used as a positive control and mouse fibroblast (FED) were used as a negative control.
  • FIGS. 3A and 3B Susceptibility of Chicken Valo EBv13 to Poxvirus
  • EBv13 (passage 188) were seeded at 0.4 ⁇ 106 cells/ml in 100 mL F175 flasks either in 40 ml of SFM Excell Medium 65319 or G9916 SFM Medium (SAFC) supplemented with 4 mM Glutamine.
  • SAFC SFM Excell Medium
  • the cell growth and infection with MVA-GFP (MOI 10 ⁇ 2 TCID50/cell) were performed at 37° C. One hour post infection, 60 ml of fresh medium were added.
  • FIG. 3A cell density kinetics in SFM Excell Medium 65319 or G9916 SFM Medium (SAFC).
  • FIG. 3B MVA productivity expressed in TCID50/ml in SFM Excell Medium 65319 or G9916 SFM Medium (SAFC).
  • FIG. 4 Transmission Electronic Microscopy Analysis of Duck EBx Cells
  • Duck EBx cells display a typical embryonic stem cells morphology (i.e high nucleo-cytoplasmic ratio) that resemble the phenotype of murine embryonic stem cells and VIVALIS EB14 cells described in WO2006/108846.
  • Duck EBx cells are small round cells with a large nucleus and nucleolus, with short pseudopodia extending from the plasma membrane. They are highly metabolic active with a ribosome and mitochondria rich cytoplasm.
  • FIGS. 5A and 5B Telomerase Expression in Duck EBx Cell Lines
  • telomerase expression during different stages of establishment of duck EBx cells was investigated by using Roche telomerase detection kit (Telomerase OCR ELISA).
  • FIG. 5A Telomerase is found to be highly expressed in different adherent duck EBx cell lines just like in chicken EBv13 cells. Duck epithelial cells used as a negative control do not express telomerase.
  • FIG. 5B During the process of establishment of suspension duck EBx cells, high level of telomerase expression is maintained. High level of telomerase were investigated in duck EBx cells during feeder deprivation (with or without feeder cells), during the process of adapting duck EB26 cells to suspension and after serum deprivation of dEB24 et dEB26.
  • Duck EBx cells such as EB24 and EB26, express high level of telomerase just like chicken EB14 cells.
  • Duck EB66 also express high level of telomerase (Data not shown).
  • FIGS. 6A and 6B Duck EBx® Cells Display No Endogenous Reverse Transcriptase Activity
  • FIG. 6A Endogenous reverse transcriptase expression was investigated by direct F-PERT analysis (Lovett et al., 1999, J. Virol. Methods, 82:185-200) in Clean Cells (FRANCE).
  • Duck EBx® cell lines, EB26 and EB5-1 display no endogenous Reverse Transcriptase (RT) activity. High level of RT activity were detected in chicken EB14 and EBv13 cells culture (at different passages) as well as, to a lesser extend, in chicken embryonic fibroblast (CEF) derived from Specific Pathogen Free (SPF) chicken strain.
  • CEM cells which are RTase negative, were used as a negative control to set the detection limit of the assay.
  • FIG. 6B Presence of endogenous retroviral particles, either replicative (i.e replication competent) or non-replicative, in the cell culture supernatant of duck and chicken EBx cells were investigated by an ELISA assay detecting the avian leukosis major capsid antigen P27.
  • Duck EBx cell lines, EB26 and EB5-1, as well as chicken EBv13 do not secrete ALV p 27 antigen.
  • chicken EB14 cells do express ALV P27 antigen.
  • FIGS. 7A and 7B Duck EBx Cells do not Secrete Replicative Avian Leucosis Virus (ALV)
  • FIG. 7A described the principle of QT6 co-culture.
  • FIG. 7B The presence of replicative virus is detected by an ELISA assay detecting the avian leukosis major capsid antigen P27.
  • the assay demonstrates that none of duck EBx® cells tested (dEB26 and dEB51) secrete replicative ALV.
  • RAV-1 virus which is known to replicate in QT6, were used as a positive control.
  • FIG. 8 Cell Surface Expression of Receptors SA ⁇ 2-3 and SA ⁇ 2-6 in Duck EBx and Chicken EB14 Cell Lines
  • FIGS. 9A and 9B MVA-GFP Virus Propagation in Infected Duck EBx Cells
  • FIG. 9A Duck EBx® were allowed to form small clumps in T175 stirred tank flasks during cell proliferation in a cell growth SFM medium. Clumps were then infected with 10 ⁇ 2 TCID 50 /cell of MVA-GFP virus and the mixture was diluted in production SFM media. During a 6 days virus propagation period at 37° C., pictures of UV-exposed infected cells were taken daily. The pick of MVA infection was reached at day 4 post-infection (pi). At day 6 ⁇ l, the infected cells start to die.
  • FIG. 9B MVA-GFP virus titration propagated in duck EBx® cells in a 3 L fed-batch bioreactor.
  • Duck EBx-derived biomass was allowed to accumulate during cell proliferation phase in Excell growth medium (SAFC).
  • SAFC Excell growth medium
  • cell density reached 4 million cells/ml.
  • Cells were then infected with 10 ⁇ 1 TCID 50 /cell of MVA-GFP virus and the mixture was diluted in 1.5 L Excell medium.
  • samples were collected daily and TCID 50 titration (Right Panel) was performed at the end of the kinetic.
  • a yield of 8.5 log TCID50/ml were reached at 4 p.i. corresponding to a yield of 205 TCID 50 /Cell.
  • FIG. 10 Influence of Calcium and Magnesium Concentration in SFM Medium on the Size of EBx® Cells Clumps
  • FIG. 10A Chicken EBv13 cells were first cultured in the SFM medium from SAFC Biosciences that comprise a high concentration of calcium (Ca2+) (Approx. 0.79 mM) and magnesium (Mg2+) ions; in this medium, cells produce large aggregates in culture.
  • Ca2+ calcium
  • Mg2+ magnesium
  • the cells form smaller aggregates.
  • FIG. 10B Duck EB24, EB26 and EB66 cells were first cultured in the SFM medium from SAFC Biosciences that comprise a high concentration of calcium (Ca2+) (Approx. 0.79 mM) and magnesium (Mg2+) ions; in this medium, cells produce large aggregates in culture. Three days after having changed the cell culture medium with the same SFM medium that comprises a lower concentration of Ca2+ (0.03 mM final) and Mg2+ (1.6 mM final), the cells form smaller aggregates.
  • Ca2+ calcium
  • Mg2+ magnesium
  • FIGS. 11A and 11B Production of Influenza Virus Strains A in Duck EBx Cells in 3 L-Bioreactors
  • Duck EBx® biomass was allowed to accumulate at 37° C. during cell proliferation phase in a cell growth medium.
  • Cells were then infected with 10 ⁇ 4 TCID 50 /cell of A/H1N1/Beijing/262/95 or A/H3N2/New York/55/2004 influenza virus, the mixture was diluted in 1.5 L Excell production medium supplemented with 0.75 USP/mL of trypsin and temperature was lowered to 33° C.
  • samples were collected daily and stored at ⁇ 80° C.
  • FIG. 11A Growth kinetic of duck EBx cells infected with A/H1N1/Beijing/262/95 influenza virus strain
  • the viral yield reached 20 ug of Hemagglutinin per ml of culture supernatant.
  • FIG. 11B Growth kinetic of duck EBx cells infected with A/H3N2/New York/55/2004 influenza virus strain
  • the viral yield reached 30 ug of Hemagglutinin per ml of culture supernatant.
  • FIG. 12 Production of Influenza Virus Strain B in Duck EBx® Cells
  • Duck EBx® biomass was allowed to accumulate at 37° C. during cell proliferation phase in a cell growth medium.
  • Cells were then infected with 10 ⁇ 3 TCID 50 /cell of B/Jiangsu/10/2003 influenza virus, the mixture was diluted in 1.5 L Excell production medium supplemented with 0.75 USP/mL of trypsin and temperature was lowered to 33° C.
  • samples were collected daily and stored at ⁇ 80° C.
  • the viral yield reached 25 ug of Hemagglutinin per ml of culture supernatant.
  • FIG. 13 Analysis of NDV Productivity and Viral Protein Expression in Suspension Duck EB66 Cells (MOI 10 ⁇ 3 , 0.75 USP/mL Trypsin)
  • FIG. 13 right Panel showed NDV viral proteins (HN, Fo/F, NP & M) expression.
  • the viral proteins composition of NDV virus produced in duck EB66 cells are similar to the one obtained with NDV virus produced in chicken EB14 cells.
  • the kinetic of release for viruses produced in chicken and Duck EBx cells are similar.
  • FIG. 14 Analysis of Recombinant Measles Virus Replication in Suspension Duck EB66 Cells (MOI 10 ⁇ 1 or 10 ⁇ 2 ) in Tissue-Culture Flasks in Serum Free Medium.
  • Duck EB66 cells are at least as sensitive as VERO cells to infection by Measles Virus. Titers (in TCID50/ml) of recombinant Measles virus expressing Green Fluorescent Protein (GFP) produced in duck EB66 cells reach 10 7 TCID50/mL at day 6 post-infection.
  • GFP Green Fluorescent Protein
  • FIGS. 15A and 15B SSEA-1, EMA-1 & Telomerase Expression in Duck EB66 Cells
  • Telomerase expression at different passages of duck EB66 cultured in roller bottles was investigated by using Roche telomerase detection kit (Telomerase OCR ELISA). SSEA-1 and EMA-1 at different passages of duck EB66 cultured in roller bottles was investigated by FACS analysis.
  • FIG. 15A Telomerase is found to be highly expressed in suspension duck EB66 cell line at different passages (138, 144, 147, 150, 154).
  • FIG. 15B SSEA-1 and EMA-1 cell surface markers was found to be highly expressed in suspension duck EB66 cell line at different passages (138, 144, 147, 150, 154).
  • FIG. 16 Karyotype Analysis of Duck EB66 Cells
  • Duck EB66 cells karyotype was performed by Pr. Franck, ENVL, Lyon. EB66 cells are diploid cells.
  • SPF Specific Pathogen Free
  • the valo strain is a white Leghorn strain produced and delivered by Lohmann from Germany.
  • Those SPF chicken eggs, supplied with a certificate of analysis, are tested for: CAV, Avian adenoviruses (group 1, serotypes 1-12 and group 3), EDS, Avian Encephalomyelitis Virus, Avian Leukosis Viruses/RSV (including Serotype ALV-J), Avian Nephritis Virus, Avian Reoviruses, Fowlpox Virus, Infectious Bronchitis Virus, Infectious Bursitis Virus (IBDV), Infectious Laryngo Tracheitis Virus, Influenzavirus Type A, Marek's Disease Virus, Mycoplasmosis (Mg+Ms), Mycobacterium avium , Newcastle Disease Virus, Reticuloendotheliosis Virus, Salmonella pullorum, Other Salmonella Infections, Avian Rhino
  • EBv13 cells from murine origin (STO cells) were used as feeder layer to maintain the pluripotency of chicken stem cells.
  • Those feeder cells are mitotically inactivated by gamma irradiation (45 to 55 Grays) before seeding on plastic.
  • This dose of irradiation is a sub-lethal dose that induces a definitive arrest of the cell cycle but still permits the production of growth factors and extracellular matrix, necessary for the promotion of the cell growth of non differentiated cells.
  • the STO cell line was derived by A. Bernstein, Ontario Cancer Institute, Toronto, Canada from a continuous line of SIM (Sandos Inbred Mice) mouse embryonic fibroblasts and it was supplied by the American Type Culture Collection (ATCC) (STO Product number: CRL-1503, Batch number 1198713).
  • Fresh feeder layers were prepared twice a week, in general on monday and thursday. Exponentially cells were dissociated and counted. A part of cells were seeded for maintenance of viable cultures and another part was irradiated. For irradiation, we prepared a cell suspension at 10 ⁇ 10 6 cells/mL in tubes. Cells were exposed to a 45 to 55 grey dose and were seeded on plastic. After seeding, dishes or plates coated with inactivated feeder cells were used during a maximum of 5 days
  • the non irradiated serum used in the program was collected and produced in United States. Animals used for collection were USDA inspected and acceptable for slaughter. It was added in the medium during avian stem cells culture. This batch was not submitted to irradiation to avoid the destruction of critical proteins or components identified as essential for the maintenance of stem cells in culture.
  • the irradiated batch used in this program was also collected in United States. This irradiated batch was added as supplement in the DMEM medium used for the culture of STO or FED cells (feeder cells). Those cells do not require as stem cells a specific quality of serum for growth and maintenance in culture. To minimize high concentration of serum in the medium we have adapted the STO cells to grow in presence of 4% of FBS only.
  • Pronase is a recombinant protease manufactured by Roche Diagnostics, Germany, used for the dissociation of adherent avian stem cells.
  • Trypsine EDTA (Cambrex, cat n o BE17-161E)
  • Trypsine is used for the dissociation of STO or FED cells and at late passages for the dissociation of avian cells adapted to Serum Free Medium.
  • This enzyme of porcine origin is manufactured aseptically according to cGMP referential conditions by a validated sterile filtration method and tested according to current E.P.
  • the raw material, irradiated prior to formulation, is tested for porcine parvovirus in strict compliance with 9/CFR 113.53.
  • This agent of dissociation is a ready to use formulation used to gently detach cells from the growing surface of the culture vessel.
  • the formula contains no protein, and allows dislodging of cells without use of enzymes. Cellular proteins are preserved making possible immunochemical studies that are dependent upon the recognition of cell surface proteins.
  • This enzyme was used to detach cell before FACS analysis of biological markers like EMA-1 (Epithelial Membrane Antigen 1) and SSEA1 (Stage Specific Embryonic antigen-1).
  • the chicken Valo embryos were placed in a tube containing physiological medium (1 ⁇ PBS, Tris Glucose, medium, and the like). The Valo embryos were then mechanically dissociated and inoculated on a layer of feeder STO cells into complete culture medium at 39° C. The feeder cells were seeded in flask at around 2.7 ⁇ 10 4 cell/cm 2 .
  • the complete culture medium is composed of basal commercial medium DMEM-Ham F12 supplemented with 10% fetal calf serum, with IGF1 and CNTF at a final concentration of 1 ng/ml, and with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 1 mM, with beta-mercapto-ethanol at a final concentration of 0.2 mM, glutamine at a final concentration of 2.9 mM, with an initial mixture of antibiotics containing penicillin at a final concentration of 100 ⁇ ml and streptomycin at a final concentration of 100 ⁇ g/ml. Rapidly after the first passages of the cells, the mixture of antibiotics is no longer added to the medium. The expression rapidly is understood to mean after the first 3 to 5 passages in general.
  • the seeding of culture dishes was performed with around between 7 ⁇ 10 4 /cm 2 to 8 ⁇ 10 4 /cm 2 of avian ES cells in the complete culture medium.
  • the seeding is made with around 7.3 ⁇ 10 4 /cm 2 (4 ⁇ 10 6 cells/55 cm 2 or 4 ⁇ 10 6 cells/100 mm dish).
  • the avian cells preferably the avian embryonic cells of step a) are cultured during several passages in the complete medium.
  • the complete medium was depleted in growth factors IGF1 and CNTF. The depletion is made directly in one step, from one passage to another.
  • the embryonic stem cells preferably the avian embryonic cells are cultured during several passages in the complete medium without IGF1 and CNTF growth factors.
  • the flask were originally seeded with around 2.7 ⁇ 10 4 feeder cells/cm 2 , then around 2.2 ⁇ 10 4 feeder cells/cm 2 , then around 1.8 ⁇ 10 4 feeder cells/cm 2 , then around 1.4 ⁇ 10 4 feeder cells/cm 2 , then around 1.1 ⁇ 10 4 feeder cells/cm 2 , then around 0.9 ⁇ 10 4 feeder cells/cm 2 , then around 0.5 ⁇ 10 4 feeder cells/cm 2 . Then the flask were seeded with 6.5 ⁇ 10 4 avian cells/cm 2 to 7.5 ⁇ 10 4 avian cells/cm 2 and without feeder cells. The depletion of feeder cells started at around passage 21 and ended at around passage 65.
  • the chicken Valo ES cells were seeded in culture flask at a lower concentration than in step a), about around 4 ⁇ 10 4 cell/cm 2 to 5 ⁇ 10 4 cell/cm 2 .
  • the avian cells are cultured for additional passages with the same feeder cells concentration before to pursue the feeder cells depletion.
  • the serum depletion were performed after the growth factor and the feeder cells depletion.
  • the culture medium were composed of basal commercial medium DMEM-HamF12 supplemented with 10% fetal calf serum and with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 1 mM, with beta-mercaptoethanol at a final concentration of 0.2 mM, glutamine at a final concentration of 2.9 mM.
  • the chicken Valo cells were adapted to the growth in a serum free medium culture in a two steps process: first, the chicken Valo cells were rapidly adapted to a culture medium composed of commercial serum free medium (SFM), preferably ExCell 60947 (SAFC Biosciences) supplemented with 10% fetal calf serum and with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 1 mM, with beta-mercaptoethanol at a final concentration of 0.2 mM, glutamine at a final concentration of 2.9 mM.
  • SFM serum free medium
  • ExCell 60947 SAFC Biosciences
  • a second step consisting of a slow adaptation to decreasing concentration of animal serum in the SFM medium were initiated. Serum depletion was performed by a progressive decreasing starting from 10% serum, then 7.5%, then 5%, then 2.5%, then 1.25%, then 0.75% of serum concentration in SFM cell culture medium to finally reach 0% serum in SFM cell culture medium. Serum depletion started at passage 103 and ended at passage 135.
  • anchorage dependent chicken Valo cells named EBv13 were able to grow in absence of grow factors, in absence of feeder cells, in serum free medium. EBv13 Cells were then adapted to growth at 37° C., by progressively decreasing cell culture temperature of 0.5° C./day.
  • SPF Chicken Specific Pathogen Free (SPF) strain called ELL-0 (East Lansing Line 0) was provided by the Avian Disease and Oncology Laboratory (USDA-ARS-MWA, USA). Those SPF chicken eggs, are produced from a flock tested intensively to various poultry pathogens. Disease tested include: Salmonella pullorum, Salmonella gallinarum, mycoplasma gallisepticum , mycoplasma synoviae, Avian Leukosis virus A-D and J, Marek's disease virus, Reticuloendotheliosis virus, Avian adenovirus, Infectious bronchitis, Infectious bursal disease, Avian Influenza, Newcastle disease, Avian encephalomyelitis and Avian Reovirus. Line 0 chicken eggs were only submitted to a desinfection with the decontaminant to avoid any risk of contamination linked to the manipulation of eggs during transportation.
  • STO cells murine origin
  • Those feeder cells are mitotically inactivated by gamma irradiation (45 to 55 Grays) before seeding on plastic.
  • This dose of irradiation is a sub-lethal dose that induces a definitive arrest of the cell cycle but still permits the production of growth factors and extracellular matrix, necessary for the promotion of the cell growth of non differentiated cells.
  • the STO cell line was derived by A. Bernstein, Ontario Cancer Institute, Toronto, Canada from a continuous line of SIM (Sandos Inbred Mice) mouse embryonic fibroblasts and it was supplied by the American Type Culture Collection (ATCC) (STO Product number: CRL-1503, Batch number 1198713). Fresh feeder layers were prepared twice a week. Exponentially cells were dissociated and counted. A part of cells were seeded for maintenance of viable cultures and another part was irradiated. For irradiation, we prepared a cell suspension at 10 ⁇ 10 6 cells/mL in tubes. Cells were exposed to a 45 to 55 grey dose and were seeded on plastic. After seeding, dishes or plates coated with inactivated feeder cells were used during a maximum of 5 days.
  • Soluble IL6r is expressed in transfected HEK293 cells.
  • the non irradiated serum used in the program was collected and produced in Australia. Animals used for collection were USDA inspected and acceptable for slaughter. It was added in the medium during avian stem cells culture. This batch was not submitted to irradiation to avoid the destruction of critical proteins or components identified as essential for the maintenance of stem cells in culture.
  • the irradiated batch used in this program was collected in Australia. This irradiated batch was added as supplement in the DMEM medium used for the culture of STO or FED cells (feeder cells). Those cells do not require as stem cells a specific quality of serum for growth and maintenance in culture. To minimize high concentration of serum in the medium we have adapted the STO cells to grow in presence of 4% of FBS only.
  • Embryos from 13 eggs from Line 0 chicken were collected according to the process described in Example 1.2. Then, the Line 0 embryos were placed in a tube containing PBS1 ⁇ . Embryos were then mechanically dissociated and inoculated on a layer of feeder STO cells into complete culture medium at 39° C. The feeder cells were seeded in dishes at around 2.7 ⁇ 10 4 cell/cm 2 .
  • the complete culture medium is composed of basal commercial medium DMEM-Ham F12 supplemented with 10% fetal calf serum, with IGF1, CNTF, bFGF, IL6, IL6r and SCF at a final concentration of 1 ng/ml, and with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 1 mM, with beta-mercapto-ethanol at a final concentration of 0.2 mM, glutamine at a final concentration of 2.9 mM, with yeastolate 1 ⁇ and with an initial mixture of antibiotics containing penicillin at a final concentration of 100 ⁇ ml and streptomycin at a final concentration of 100 ⁇ g/ml. After 7 passages, the mixture of antibiotics is no longer added to the medium.
  • the seeding of culture dishes was performed with around between 7 ⁇ 10 4 /cm 2 to 8 ⁇ 10 4 /cm 2 of avian ES cells in the complete culture medium.
  • the seeding is made with around 7.3 ⁇ 10 4 /cm 2 (4 ⁇ 10 6 cells/55 cm 2 or 4 ⁇ 10 6 cells/100 mm dish).
  • the avian cells, preferably the avian embryonic cells of step a) are cultured during several passages in the complete medium supplemented with 10 or 15% of FBS. At passage 7, the complete medium was depleted in growth factors bFGF, IL6, IL6r and SCF.
  • the depletion was made directly in one step, from one passage to another.
  • the embryonic stem cells preferably the avian embryonic cells, were cultured during several passages in the complete medium without those 4 growth factors.
  • the 2 last factors IGF1 and CNTF were removed from the medium and cells were amplified without factor.
  • DMEM Ham F12 from passage 1 to passage 18
  • Exell GTM-3 from passage 18 to passage 26
  • Excell 66788 and Excell 66522 after passage 26.
  • Duck eggs from Peking strains GL30 were obtained from GRIMAUD FRERES SELECTION (La Corbière, Roussay France).
  • the parent ducks were vaccinated against Escherichia Coli (Autogenous vaccine Coli 01 & O 2 ), Pasteurella multocida (Landavax), Duck viral hepatitis (Hepatovax), Erysipelothrix rhusiopathiae (Ruvax), Avian metapneumovirus (Nemovac), Salmonella typhimurium & Enteridis (Autogenous vaccine), Riemerella antipestifer (Autovaccine Riemerella), Avian metapneumovirus (Nobilis RTV inactive) and Erysipelothrix rhusiopathiae (Ruvax).
  • fertilized Peking duck eggs were submitted to a disinfection in an hypochloryde bath followed by a decontamination with Fermacidal (Thermo) to avoid
  • STO cells cells from murine origin
  • Those feeder cells are mitotically inactivated by gamma irradiation (45 to 55 Grays) before seeding on plastic.
  • This dose of irradiation is a sub-lethal dose that induces a definitive arrest of the cell cycle but still permits the production of growth factors and extracellular matrix, necessary for the promotion of the cell growth of non differentiated cells.
  • the STO cell line was derived by A.
  • the non irradiated serum used in the program was collected and produced in Australia. Animals used for collection were USDA inspected and acceptable for slaughter. It was added in the medium during avian stem cells culture. This batch was not submitted to irradiation to avoid the destruction of critical proteins or components identified as essential for the maintenance of stem cells in culture.
  • the irradiated batch used in this program was collected in United States. This irradiated batch was added as supplement in the DMEM medium used for the culture of STO cells (feeder cells). Those cells do not require as stem cells a specific quality of serum for growth and maintenance in culture. To minimize high concentration of serum in the medium we have adapted the STO cells to grow in presence of 4% of FBS only.
  • Pronase is a recombinant protease manufactured by Roche Diagnostics, Germany, used for the dissociation of adherent avian stem cells.
  • Trypsine EDTA (Cambrex, cat n o BE17-161E)
  • Trypsine is used for the dissociation of STO cells and at late passages for the dissociation of avian cells adapted to Serum Free Medium.
  • This enzyme of porcine origin is manufactured aseptically according to cGMP referential conditions by a validated sterile filtration method and tested according to current E.P.
  • the raw material, irradiated prior to formulation, is tested for porcine parvovirus in strict compliance with 9/CFR 113.53.
  • Trypzean solution is formulated with a recombinant bovine trypsin, expressed in corn and manufactured by Sigma Aldrich utilizing ProdiGene's proprietary transgenic plant protein expression system. This product is optimized for cell dissociation in both serum free and serum-supplemented adherent cell cultures.
  • This agent of dissociation is a ready to use formulation used to gently detach cells from the growing surface of the culture vessel.
  • the formula contains no protein, and allows dislodging of cells without use of enzymes. Cellular proteins are preserved making possible immunochemical studies that are dependent upon the recognition of cell surface proteins.
  • This enzyme was used to detach cell before FACS analysis of biological markers like EMA-1 (Epithelial Membrane Antigen 1) and SSEA1 (Stage Specific Embryonic antigen-1).
  • the duck embryos were placed in 50 mL tubes containing PBS1 ⁇ . The duck embryos were then mechanically dissociated, washed with PBS, and seeded on an inactivated layer of feeder STO cells into complete culture medium at 39° C., 7.5% CO 2 . The feeder cells were seeded in 6 well plates or dishes at around 2.7 ⁇ 10 4 cell/cm 2 .
  • the complete culture medium is composed of serum free medium DMEM-Ham F12 supplemented with 10% fetal bovine serum, with IGF1, CNTF, at a final concentration of 1 ng/ml, and with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 0.1 mM, with beta-mercapto-ethanol at a final concentration of 0.5 mM, glutamine at a final concentration of 2.1 mM, penicillin at a final concentration of 100 U/ml, streptomycin at a final concentration of 100 ⁇ g/ml and yeastolate 1 ⁇ . Rapidly at the passage 4, the mixture of antibiotics is no longer added to the medium.
  • the duck ES cells were cultured in the DMEM-Ham F12 medium up to passage 4.
  • the base medium is modified and DMEM-Ham F12 complete medium is replaced by the SFM GTM-3 medium supplemented with 10% fetal bovine serum, with IGF1, CNTF, at a final concentration of 1 ng/ml, with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 0.1 mM, with beta-mercapto-ethanol at a final concentration of 0.5 mM, glutamine at a final concentration of 2.1 mM and yeastolate 1 ⁇ .
  • the duck ES cells were further cultured during 14 passages in this new medium of culture, then growth factors deprivation was performed at passage 18.
  • IGF1 and CNTF were simultaneously removed from the medium, thus from passage 19 to passage 24, the medium of culture was GTM-3 medium supplemented with 10% FBS, with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 0.1 mM, with beta-mercapto-ethanol at a final concentration of 0.5 mM, glutamine at a final concentration of 2.1 mM and yeastolate 1 ⁇ .
  • the seeding of culture dish was performed with around between 7 ⁇ 10 4 /cm 2 to 12 ⁇ 10 4 /cm 2 of duck ES cells in the complete culture medium.
  • the dishes were originally seeded with around 2.7 ⁇ 10 4 feeder cells/cm 2 , then around 1.8 ⁇ 10 4 feeder cells/cm 2 between passage 25 and 31, then around 1.4 ⁇ 10 4 cells/cm 2 between passage 32 and 35, then around 1 ⁇ 10 4 feeder cells/cm 2 between passage 36 and 41, then around 0.7 ⁇ 10 4 feeder cells/cm 2 between passage 42 and 44, and finally from passage 45 dishes were seeded only with avian cells and without feeder cells. At the end of the feeder depletion, the dishes are seeded with 9 ⁇ 10 4 avian cells/cm 2 to 12.7 ⁇ 10 4 avian cells/cm 2 .
  • the depletion of feeder cells started at passage 25 and ended at passage 45.
  • the duck ES cells are seeded in culture dishes at a higher concentration than in step a), about around 9 ⁇ 10 4 cell/cm 2 to 12.7 ⁇ 10 4 cell/cm 2 .
  • PDT Population Doubling Time
  • Density Density
  • the culture medium was Excell 65788 supplemented with 10% FBS, 2.5 mM glutamine and 1 ⁇ yeastolate.
  • 4 ⁇ 10 6 cells were transferred in a Ultra Low Attachment (ULA) dish maintained under constant agitation to initiate anchorage-independent cells growth.
  • ULA Ultra Low Attachment
  • the base medium was modified and percentage of serum was decreased from 10% to 5% for the seeding in the ULA dish.
  • the medium of culture was SFM GTM-3 supplemented with 5% FBS, 2.5 mM glutamine and 1 ⁇ yeastolate. Slow decrease of FBS was initiated on EB66 cell suspension after passage 85.
  • Serum depletion was performed by a progressive decreasing starting from 2.5% serum, then 1.5% of serum concentration in SFM cell culture medium to finally reach 0% serum in SFM cell culture medium. Serum depletion started at passage 86 and ended at passage 94. At the end of serum depletion, anchorage independent dEB66 cells were able to grow at 37° C. in absence of grow factors, in absence of feeder cells, in serum free medium.
  • suspension duck EB66 cell could also realized in presence or absence of yeastolate
  • Soluble IL6r is expressed in transfected HEK293 cells.
  • the duck embryos were collected as previously described with EB66.
  • the duck embryos were placed in 50 mL tubes containing PBS1 ⁇ .
  • the duck embryos were then mechanically dissociated, washed in PBS, and seeded on an inactivated layer of feeder STO cells into complete culture medium at 39° C., 7.5% CO 2 .
  • the feeder cells were seeded in 6 well plates or dishes at around 2.7 ⁇ 10 4 cell/cm 2 .
  • the complete culture medium is composed of serum free medium GTM-3 supplemented with 5% fetal bovine serum, with IGF1, CNTF, 11-6, II-6R, SCF and FGF at a final concentration of 1 ng/ml, and with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 0.1 mM, with beta-mercapto-ethanol at a final concentration of 0.5 mM, glutamine at a final concentration of 2.1 mM, penicillin at a final concentration of 100 U/ml, streptomycin at a final concentration of 100 ⁇ g/ml and yeastolate 1 ⁇ .
  • the duck ES cells were cultured in the complete medium up to passage 9. After passage 9, the complete medium is partially depleted in factors. Thus, between passage 10 and 13, SCF, IL6, IL6r and bFGF were removed for the medium and only recombinant IGF1 and CNTF were maintained at a concentration of 1 ng/mL. A simultaneous decease of concentration of IGF1 and CNTF is secondly performed between passage 13 and 16 to finally obtain cells able to grow without recombinant factors at passage 17. The factor depletion were made by a progressive adaptation to lower concentrations of factors.
  • the seeding of culture dish was performed with around between 7 ⁇ 10 4 /cm 2 to 12 ⁇ 10 4 /cm 2 of duck ES cells in the complete culture medium.
  • the seeding is made with around 7.3 ⁇ 10 4 /cm 2 (4 ⁇ 10 6 cells/55 cm 2 or 4 ⁇ 10 6 cells/100 mm dish).
  • a decrease of yeastolate were performed at passage 23 reaching the final concentration at 0.5 ⁇ .
  • depletion of feeder cells were performed by a progressive decrease of feeder cells concentration over several passages.
  • the dishes were originally seeded with around 207 ⁇ 10 4 feeder cells/cm 2 , then around 1.8 ⁇ 10 4 feeder cells/cm 2 between passage 32 and 38, then around 1.4 ⁇ 10 4 cells/cm 2 between passage 39 and 44, then around 1 ⁇ 10 4 feeder cells/cm 2 between passage 45 and 47, then around 0.7 ⁇ 10 4 feeder cells/cm 2 between passage 48 and 50, and finally from passage 51 dishes were seeded only with avian cells and without feeder cells.
  • the dishes are seeded with 9 ⁇ 10 4 avian cells/cm 2 to 12.7 ⁇ 10 4 avian cells/cm 2 .
  • the depletion of feeder cells started at passage 32 and ended at passage 51.
  • the duck ES cells are seeded in culture dishes at a higher concentration than in step a), about around 9 ⁇ 10 4 cell/cm 2 to 12.7 ⁇ 10 4 cell/cm 2 .
  • growth parameters Population Doubling Time (PDT) and Density
  • PDT Population Doubling Time
  • Density Density
  • the culture medium was supplemented with 5% FBS, 0.5 ⁇ yeastolate and 2.5 mM glutamine only.
  • the serum depletion is performed on cell suspensions already depleted in growth factor, feeder cells, vitamins, non essential amino acids, sodium pyruvate and beta-mercaptoethanol. Serum depletion was performed by a progressive decreasing starting from 5% serum, then 2.5%, then 1.5%, of serum concentration in SFM cell culture medium to finally reach 0% serum in SFM cell culture medium. Serum depletion started at passage 61 and ended at passage 79.
  • anchorage independent duck EB26 cells were able to grow at 39° C. in absence of grow factors, in absence of feeder cells, in serum free medium. EB26 cells were then adapted to growth in absence of 0.5 ⁇ yeastolate at 37° C., by decreasing cell culture temperature at passage 80.
  • Soluble IL6r is expressed in transfected HEK293 cells.
  • the duck embryos were collected as previously described with EB66.
  • the duck embryos were placed in 50 mL tubes containing PBS1 ⁇ .
  • the duck embryos are then mechanically dissociated and seeded on an inactivated layer of feeder STO cells into complete culture medium at 39° C., 7.5% CO 2 .
  • the feeder cells were seeded in 6 well plates or dishes at around 2.7 ⁇ 10 4 cell/cm 2 .
  • the complete culture medium is composed of serum free medium DMEM-Ham F12 supplemented with 10% fetal bovine serum, with IGF1, CNTF, II-6, II-6R, SCF and FGF at a final concentration of 1 ng/ml, and with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 0.1 mM, with beta-mercapto-ethanol at a final concentration of 0.5 mM, glutamine at a final concentration of 2.1 mM, penicillin at a final concentration of 100 U/ml, streptomycin at a final concentration of 100 ⁇ g/ml and 1 ⁇ yeastolate. Rapidly after the first passages of the cells, the mixture of antibiotics is no longer added to the medium. The expression rapidly is understood to mean after the first 3 to 9 passages in general.
  • the duck ES cells are cultured in the DMEM-Ham F12 complete medium up to passage 7.
  • the base medium is modified and DMEM-Ham F12 complete medium is replaced by the GTM-3 complete medium supplemented with 10% fetal bovine serum, with IGF1, CNTF, II-6, II-6R, SCF and FGF at a final concentration of 1 ng/ml, with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 0.1 mM, with beta-mercapto-ethanol at a final concentration of 0.5 mM, glutamine at a final concentration of 2.1 mM, penicillin at a final concentration of 100 U/ml, streptomycin at a final concentration of 100 ⁇ g/ml and yeastolate 1 ⁇ .
  • the serum concentration is decreased at 5% and SCF, IL6, IL6r and bFGF are removed for the medium.
  • the medium is composed of 5% FBS, with IGF1 and CNTF at a final concentration of 1 ng/mL with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 0.1 mM, with beta-mercapto-ethanol at a final concentration of 0.5 mM, glutamine at a final concentration of 2.1 mM, penicillin at a final concentration of 100 U/ml, streptomycin at a final concentration of 100 ⁇ g/ml and yeastolate 1 ⁇ .
  • a simultaneous withdrawal of IGF1 and CNTF is performed at passage 22. No recombinant factors are present in the GTM-3 culture medium after passage 22. Duck cells were maintained in a such medium between passage 23 and passage 28.
  • the seeding of culture dish was performed with around between 7 ⁇ 10 4 /cm 2 to 12 ⁇ 10 4 /cm 2 of duck ES cells in the complete culture medium.
  • the seeding is made with around 7.3 ⁇ 10 4 /cm 2 (4 ⁇ 10 6 cells/55 cm 2 or 4 ⁇ 10 6 cells/100 mm dish). Then, after passage 28, depletion of feeder cells is performed by a progressive decrease of feeder cells concentration over several passages.
  • the dishes were originally seeded with around 2.7 ⁇ 10 4 feeder cells/cm 2 , then around 1.8 ⁇ 10 4 feeder cells/cm 2 between passage 29 and 33, then around 1.4 ⁇ 10 4 cells/cm 2 between passage 34 and 37, then around 1 ⁇ 10 4 feeder cells/cm 2 between passage 38 and 42, then around 0.7 ⁇ 10 4 feeder cells/cm 2 between passage 43 and 46, and finally from passage 47 dishes were seeded only with avian cells and without feeder cells.
  • the dishes are seeded with 9 ⁇ 10 4 avian cells/cm 2 to 12.7 ⁇ 10 4 avian cells/cm 2 .
  • the depletion of feeder cells started at passage 29 and ended at passage 47.
  • the duck ES cells are seeded in culture dishes at a higher concentration than in step a), about around 9 ⁇ 10 4 cell/cm 2 to 12.7 ⁇ 10 4 cell/cm 2 .
  • growth parameters Population Doubling Time (PDT) and Density
  • PDT Population Doubling Time
  • Density Density
  • culture in suspension is initiated at passage 48. 8 ⁇ 10 6 cells were transferred in a Ultra Low attachment dish and maintained under constant agitation at around 50 to 70 rpm.
  • cells were seeded in T175 flasks (Sarsted, ref 831812502) at a concentration comprise between 0.4 to 0.5 ⁇ 10 6 cells/mL.
  • cell PDT decreased from around 248H to 128 hours and the next step of deprivation is then performed.
  • vitamines, non essential amino acids, sodium pyruvate and beta mercaptethanol are removed.
  • the serum deprivation was initiated.
  • the culture medium GTM-3 was supplemented with 5% FBS, 1 ⁇ yeastolate and 2.5 mM glutamine only.
  • the serum depletion is performed on cell suspensions already depleted in growth factors, feeder cells, vitamins, non essential amino acids, sodium pyruvate and beta-mercaptoethanol.
  • Serum depletion was performed by a progressive decreasing starting from 5% serum, then 2.5%, then 2%, then 1.5% of serum concentration in SFM cell culture medium to finally reach 0% serum in SFM cell culture medium. Serum depletion started at passage 57 and ended at passage 77.
  • Duck SPF eggs from Muscovy strains were obtained from Le Couvoir de Cerveloup (France). Those SPF duck eggs, are produced from a flock tested intensively to various poultry pathogens. Disease tested include: Salmonella gallinarum - pullorum, Mycoplasma synoviae, Mycoplasma meleagridis, Mycoplasma galliepticum , Marek's disease virus, Avian Influenza, Type 2 Paramyxovirus, Type 3 Paramyxovirus, Newcastle disease, Type 3 Adenovirus (EDS), Gumboro disease, Avian reovirus, Reticuloendotheliosis virus, Avian encephalomyelitis, infectious rhinotracheitis virus and Chlamydiosis. Muscovy duck eggs were only submitted to a disinfection with the decontaminant to avoid any risk of contamination linked to the manipulation of eggs during the transport.
  • the non irradiated serum used in the program was collected and produced in Australia. Animals used for collection were USDA inspected and acceptable for slaughter. It was added in the medium during avian stem cells culture. This batch was not submitted to irradiation to avoid the destruction of critical proteins or components identified as essential for the maintenance of stem cells in culture.
  • the irradiated batch used in this program was collected in Australia. This irradiated batch was added as supplement in the DMEM medium used for the culture of STO cells (feeder cells). Those cells do not require as stem cells a specific quality of serum for growth and maintenance in culture. To minimize high concentration of serum in the medium we have adapted the STO cells to grow in presence of 4% of FBS only.
  • Embryos from 20 fertilized SPF eggs from Muscovy ducks were collected according to the process described in Example 3.
  • the duck embryos were placed in 50 mL tubes containing PBS1 ⁇ .
  • the duck embryos were then mechanically dissociated, washed with PBS, and seeded in a well of a 12 well plate coated with an inactivated layer of feeder STO cells.
  • Duck Embryonic cells were seeded into complete culture medium and transferred at 39° C., 7.5%5% CO 2 .
  • the feeder cells were seeded at around 2.7 ⁇ 10 4 cell/cm 2 .
  • the complete culture medium used is composed of DMEM-Ham F12 supplemented with 10% fetal bovine serum, with IGF1, CNTF, at a final concentration of 1 ng/ml, and with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 0.1 mM, with beta-mercapto-ethanol at a final concentration of 0.5 mM, glutamine at a final concentration of 2.1 mM, penicillin at a final concentration of 100 U/ml, streptomycin at a final concentration of 100 ⁇ g/ml and yeastolate 1 ⁇ .
  • the DMEM-HamF12 base medium is replaced by GTM-3 base medium. The mixture of antibiotics is no longer added to the medium after passage 4.
  • the duck ES cells were cultured in the complete GTM-3 medium up to passage 8. After passage 8, concentration of IGF1 and CNTF are reduced to 0.5 ng/mL. The duck ES cells were further cultured during 2 passages in this new medium of culture, then growth factor deprivation was performed at passage 10. IGF1 and CNTF were simultaneously removed from the medium.
  • the medium of culture was GTM-3 medium supplemented with 10% FBS, with 1% non-essential amino acids, with 1% of mixture of vitamins of commercial origin, with sodium pyruvate at a final concentration of 0.1 mM, with beta-mercapto-ethanol at a final concentration of 0.5 mM, glutamine at a final concentration of 2.1 mM and yeastolate 1 ⁇ .
  • the seeding was performed with around 12 ⁇ 10 4 /cm 2 of duck ES cells in the culture medium.
  • Some conditionated medium can be occasionaly used for cell seeding to improve cell recovery post dissociation.
  • Anchorage-independent Muscovy duck EBx cells express ES cells markers, such as telomerase, SSEA-1 and EMEA-1 (data not shown).
  • Telomerase detection is achieved by using the Telo TAGGG Telomerase PCR ELISA developed by Roche Applied Science (Telomeric Repeat Amplification Protocol (TRAP)—Cat. No. 11 854 666 910) according to the supplier protocol.
  • the Telo TAGGG Telomerase PCR ELISA allows amplification of Telomerase-mediated elongation products combined with non radioactive detection following an ELISA protocol.
  • the assay is valid if absorbance value of the negative control is less than or equal to 0.25 A 450nm -A 690nm and if absorbance value of the positive control is higher than or equal to 1.5 A 450nm A 690nm when using 1 ⁇ 10 3 cell equivalents in the assay.
  • Two controls were used: the negative control is murine fibroblasts (FED cells) and the positive controls are FGB8 cells (Embryonic Stem cells established by Vivalis from 129 SV mouse embryos) and chicken EB14-O74 cells previously established in WO 03/076601.
  • FED cells murine fibroblasts
  • FGB8 cells Embryonic Stem cells established by Vivalis from 129 SV mouse embryos
  • chicken EB14-O74 cells previously established in WO 03/076601.
  • EBv13 cells do express high level of telomerase. At passage p 193 and 195, the telomerase activity is equivalent to the one of chicken EB14-O74 cells.
  • Embryonic stem cells are characterized by the expression of biological markers expressed on the cell membrane.
  • the expression of EMA-1 (Epithelial Membrane Antigen-1) and SSEA-1 (Stage Specific Embryonic Antigen-1) on EBv13 cells were evaluated by FACS analysis. After 10 minutes of fixation with PFA 4% (Para-formaldehyde), cell samples and controls are rinsed and pre-incubated with monoclonal antibodies specific of EMA-1 or SSEA-1. A second antibody conjugated to FITC is used for detection of cells expressing the 2 biological markers selected. Samples were analyzed by flow cytometry using a FACS (Flow Activated Cell Sorter) from Coulter.
  • FACS Flow Activated Cell Sorter
  • FED cells mouse fibroblasts cells
  • murine ES FGB8 cells as a positive control
  • chicken EB14-O74 cells as a positive control EBx cells and EBv13 cells.
  • FED cells do not express biological markers whereas FGB8 and EB14-O74 cells present an important staining, respectively of, 60.13% and 78.7 for EMA-1 and 94.45% and 95% for SSEA-1 (data not shown).
  • Chicken valo EBv13 cells population do not present any staining for EMA1 (2%) and a very light one for SSEA-1 (22%).
  • the inventors have found that the concentration of Calcium and Magnesium in the serum-free medium used for the EBx cells culture and infection have an impact on the clumps size.
  • FIG. 10 shows the decrease in clumps size when EBv13 cells are passed from a medium with a high to a low Ca2+ and Mg2+ concentration.
  • dEBx® cells Transmission Electronic Microscopy analysis of dEBx® cells were performed by Dr. A Rilude (Lyon, France).
  • Duck EBx® cells display a typical embryonic stem cells morphology (i.e high nucleo-cytoplasmic ratio) that resemble the phenotype of murine embryonic stem cells and VIVALIS EB14 cells described in WO2006/108846.
  • Duck EBx® cells are small round cells (diameter ⁇ 10 ⁇ m) with a large nucleus and nucleolus, with short pseudopodia extending from the plasma membrane ( FIG. 4 ). They are highly metabolic active with a ribosome and mitochondria rich cytoplasm. They contain numerous intracellular vacuoles, a very developed Golgi system and a granulous reticulum endoplasmic.
  • telomerase expression during different stages of establishment of in duck EBx® cells was investigated by using Roche telomerase detection kit (Telomerase OCR ELISA). Telomerase is found to be highly expressed in adherent duck EBx® cells, as well as during feeder deprivation, during the process of adapting duck EBx® cells to suspension and during feeder deprivation.
  • FIG. 5 shows that duck EB24 and EB26 express high level of telomerase, just like chicken EB14 cells. Duck EB66 also express high level of telomerase all along cell passages. This high telomerase activity is stable in EB66 cells after adaptation in different SFM ( FIG. 15 ).
  • RT activity were detected in chicken EB14 cells culture as well as, to a lesser extend, in chicken Embryonic fibroblast derived from Specific Pathogen Free (SPF) chicken strain.
  • SPF Specific Pathogen Free
  • FIG. 7A described the principle of QT6 co-culture.
  • the presence of replicative virus is detected by an ELISA assay detecting the avian leucosis major capsid antigen P27.
  • the assay demonstrates that none of duck EBx cells tested secrete replicative (i.e replication competent) ALV ( FIG. 7B ).
  • Chicken EB14 and duck EBx cells were washed in 10 mM HEPES, 150 mM NaCl pH7.5 and resuspended in the same buffer at a 5.10 6 final concentration. Cells were incubated 30 min on ice, then for an additional 15 to 30 minutes in presence of SNA or MAA. Lectin treated cells were washed in 10 mM HEPES, 150 mM NaCl pH7.5, prior to incubation on ice during 15 to 30 minutes with FITC-labelled anti-digoxygenin antibody. Then cells are washed in NaCl 0.9% and FACS analyzed.
  • Chicken EB14 and duck EBx cells express cell surface receptors comprising oligosaccharides with Sia ⁇ 2-6Gal and Sia ⁇ 2-3Gal residues ( FIG. 8 ).
  • FIG. 16 shows diploid karyotype of duck EBx66 cells ( FIG. 16 ).
  • EBv13 cells Susceptibility of EBv13 cells to infection with poxvirus was investigated using a recombinant Modified Vaccinia Ankara (MVA) encoding a GFP gene (Green Fluorescent Protein).
  • MVA Modified Vaccinia Ankara
  • Virus infectivity was measured through microscopic observation of global cytopathic effect (CPE) and UV-exposed infected cells. Then, TCID50 titers were calculated according the Reed and Muench method (1938, A simple method of estimating fifty percent endpoints. Am. J. Hyg. 27, 493-97). All along the experiment cell proliferation and viability are monitored. Chicken Valo EBv13 cells appear to be highly sensitive to MVA-GFP infection ( FIGS. 3A-3B ).
  • Duck EBx cells were stored in cryovials in liquid nitrogen at ⁇ 196° C. (20 ⁇ 10 6 cells/vial). The cryovial is directly thawed into a +37° C. pre-warmed water bath. The cell suspension is put into a 50 ml sterile tube with 30 ml pre-warmed culture medium. After centrifugation (5 min at 300 ⁇ 20 g, at room temperature), 15 ml of fresh culture medium is added on the pellet and gently homogenised. The sample is numbered using Trypan blue. Numeration has to be ⁇ 20 ⁇ 10 6 cells and viability has to be >70% to guarantee a good culture.
  • the cell suspension is plated into a T75 cm 2 flask and incubate at +37° C. under an 7.5% CO 2 atmosphere on an orbital shaker at 50 rpm. Fresh medium is then added daily.
  • the cells are then passaged to increase cells biomass to seed a 3 L-bioreactor. 320.10 6 cells are needed to inoculate a 3 L-bioreactor.
  • a sample is taken after gently mixing to perform a numeration using trypan blue to determine cell density.
  • a 150 mL cell mix is prepared in order to obtain a cell concentration of 0.4 ⁇ 10 6 cells.ml ⁇ 1 into the 800 ml final culture volume in the bioreactor.
  • the pH Prior to seed cells, the pH is set in the vessel to 7.2 (because pH will be decrease by CO 2 surface injection).
  • the pO 2 is set to 50% O 2 saturation (the mass flow controller is adjusted to 100% which correspond to a maximum sparger flow rate to 50 ml.min ⁇ 1 ).
  • the pH is maintained by CO 2 surface injection, later, it is controlled by addition of 7.5% NaHCO 3 .
  • the surface aeration is started with air at a flow rate of 0.3 ml.min ⁇ 1 . Cell numeration is performed on a routine basis.
  • cell density should be higher than 4 ⁇ 5 ⁇ 10 6 cells.ml ⁇ 1 .
  • the virus infection is performed at a MOI of 10 ⁇ 4 .
  • the vessel temperature is set to 33° C.
  • the virus strain is thawed on ice.
  • the infection mix is prepared in 10 ml of production medium. After inoculation of the infection mix into the bioreactor, viral adsorption is performed during 1 hour.
  • the final production medium is prepared: in 1.5 L of production medium, trypsin is added in order to obtain a final concentration in the vessel of 0.3 U.ml ⁇ 1 (2.3 L on the whole). The pre-warmed final production medium is then added.
  • a sample of approximatively 15 m 1 is collected from the bioreactor to perform cell numeration, cell morphology analysis and to observe CPE.
  • the metabolites such as glutamate, glutamine, lactate and glucose are analyzed all along the culture with the BioProfile Basic software. Concentration of the metabolites is adjusted if necessary. For example, glutamine concentration is adjusted to 2 mM if necessary. The glucose concentration is adjusted to 2 g.L ⁇ 1 if necessary.
  • Virus titration is carried-out at the end of the experiment using all collected samples.
  • Duck EBx® cells are routinely cultured in stirred-tank bioreactor.
  • Duck EBx®-derived biomass is allowed to accumulate at 37° C. in a cell growth medium until a cell density of 5-6.10 6 cells/mL was reached. Then the mixture is diluted from around 3 to 10 fold, and cell growth kinetic is followed-up over a 10 days period. In such conditions, cell density of 12 to 20 million cells/m 1 is routinely reached around day 5 to 8.
  • Duck EBx® cells display a range of splitting ratio that goes at least up to 10 to 15 fold.
  • the inventors have found that the concentration of Calcium and Magnesium in the serum-free medium used for the EBx cells culture and infection may have an impact on the clumps size.
  • the presence of small clumps of duck EBx cells improves virus infection and propagation, leading to high MVA virus titers ( FIG. 9A ).
  • Duck EBx®-derived biomass was allowed to accumulate during cell proliferation phase in Excell 66444 growth medium. Cells were then infected with 10 ⁇ 2 TCID 50 /cell of MVA-GFP virus and the mixture was diluted in Excell 66444 production medium. Following addition of fresh Excell medium, cell density dropped down on day 2, and at day 4, the cell density of infected cells increased and reached 12 million cell/ml. In such conditions, the MVA-GFP productivity is high. Since at day 4 post-infection, the MVA-GFP titer is around 10 8 TCID50/ml ( FIG. 9B ). A MVA-GFP yield of 205 TCID50/cell was obtained in duck EBx® cells.
  • the concentration of haemagglutinin in samples derived from influenza virus infected-EB14 cells was determined as described by Wood and colleagues*. Briefly, glass plates were coated with an agarose gel containing anti-Influenza serum (recommended concentration provided by NIBSC). After the gel has set, 10 ⁇ L of appropriate dilutions of the reference and the samples were loaded in 3 mm ⁇ punched wells. Following a 18-24 h incubation in a moist chamber at room temperature, plates were soaked in 0.9% NaCl and washed in distilled water. The gel was then pressed and dried.
  • Blots were blocked for 1 h at room temperature with a mixture composed of 5% fat dry milkpowder in TBST suplemented with 1% FCS (SAFC). Then, the blots were incubated overnight in blocking solution supplemented with specific polyclonal anti-HA sheep serum (1:500 (NIBSC). The blots were washed 6 times with TBST and incubated for 1 h at room temperature with a hrp-conjugated rabbit anti-sheep IgG polyclonal antibody (1:5000 (Rockland) in blocking solution. After 6 washes with TBST, the protein-conjugate complex was finally revealed using chemiluminescence (ECL kit, Amersham) and films (Hyperfilm, Amersham).
  • chemiluminescence ECL kit, Amersham
  • films Hyperfilm, Amersham
  • Virus titration, haemmaglutinin assays (HAU) and HA antigen quantifications are carry out at the end of the experiment using all collected samples.
  • the inventors demonstrate that duck EBx cells are a reliable and efficient cell substrate for the replication of various strains A and B of influenza virus.
  • Influenza virus production can be performed in various vessels, such as flasks and spinner (data not shown) and bioreactors.
  • Reproducible and efficient fedbatch process of production of influenza virus in 3 L and 30 L stirred tank bioreactors were obtained by the inventors.
  • Viral yield above 15 mg/l and up to 50 mg/l of haemagglutinin are routinely obtained in flasks and in bioreactors with strains A and B of influenza virus ( FIGS. 11 and 12 ).
  • Duck EBx® cells were grown in Excell medium (SFAC) in T175 flasks at 37° C. under 7.5% CO 2 atmosphere on an orbital shaker at 60 rpm. At day 0, cells are seeded at 0.4 ⁇ 10 6 cells/mL in 40 ml fresh medium. Cell culture was incubated at 37° C., 7.5% CO 2 under shaking (60 rpm). Cell growth kinetics were followed until cell density has reached a concentration between 4 ⁇ 10 6 to 6 ⁇ 10 6 cells/ml (usually at day 3 post seeding).
  • cells are inoculated with NDV La Sota strain at two different MOI (10 ⁇ 3 and 10 ⁇ 4 TCID 50 /cells) and incubated for one additional hour at 37° C., 7.5% CO 2 under shaking (60 RPM). Then the cell culture was diluted with the addition of 60 mL fresh viral production medium and the incubation pursued at 37° C. and 7.5% CO 2 under shaking (60 rpm). The cell growth and virus production kinetics were performed over 7 days. As a source of protease, recombinant trypsin (SAFC) was added every day in the culture medium; two concentration of trypsin (0.4 and 0.75 USP/mL) were tested. Daily aliquots were removed for cell numeration, virus titration and Western blotting analysis.
  • SAFC recombinant trypsin
  • Duck and chicken EBx cells are sensitive to and replicate NDV La Sota strain. Titers (in TCID50/ml) of NDV produced in duck EBx® cells increase from day 0 to day 2 ⁇ l to reach an average of 10 6.83 TCID50 /mL ( FIG. 13 left panel).
  • FIG. 13 right Panel showed NDV viral proteins (HN, Fo/F, NP & M) expression.
  • the viral proteins composition of NDV virus produced in duck EBx® cells are similar to the one obtained with NDV virus produced in chicken EBx® cells.
  • the kinetic of release for viruses produced in chicken and Duck EBx cells are similar.
  • EB66 cells were grown in Excell medium in T175 flasks at 37° C. under 7.5% CO 2 atmosphere on an orbital shaker at 60 rpm. At day 0, cells are seeded at 0.4 ⁇ 10 6 cells/mL in 40 ml fresh medium. Cell culture was incubated at 37° C., 7.5% CO 2 under shaking (60 rpm). Cell growth kinetics were followed until cell density has reached a concentration between 4 ⁇ 10 6 to 6 ⁇ 10 6 cells/ml (usually at day 3 post seeding).
  • cells are inoculated with recombinant measles virus at two different MOI (10 ⁇ 1 and 10 ⁇ 2 TCID 50 /cells) and incubated for one additional hour at 37° C., 7.5% CO 2 under shaking (60 RPM). Then the cell culture was diluted with the addition of 60 mL fresh viral production medium and the incubation pursued at 37° C. and 7.5% CO 2 under shaking (60 rpm). The cell growth and virus production kinetics were performed over 7 days. Daily aliquots were removed for cell numeration and virus titration.
  • EB66 cells are sensitive to and replicate measles virus.
  • titers (in TCID50/ml) of measles produced in EB66 cells reach an average of 10 7 TCID50/mL ( FIG. 14 ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Gynecology & Obstetrics (AREA)
  • Developmental Biology & Embryology (AREA)
  • Reproductive Health (AREA)
  • Pulmonology (AREA)
  • Cell Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Oncology (AREA)
  • Molecular Biology (AREA)
  • Communicable Diseases (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US12/597,486 2007-04-24 2008-04-23 Duck embryonic derived stem cell lines for the production of viral vaccines Abandoned US20100062489A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/069,423 US9260694B2 (en) 2007-04-24 2013-11-01 Generation of duck cell lines
US15/013,079 US9822345B2 (en) 2007-04-24 2016-02-02 Method of making a virus using duck embryonic derived stem cell lines
US15/785,530 US20180135026A1 (en) 2007-04-24 2017-10-17 Duck embryonic derived stem cell lines for the production of viral vaccines

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP07300979A EP1985305A1 (en) 2007-04-24 2007-04-24 Duck embryonic derived stem cell lines for the production of viral vaccines
EP07300979.7 2007-04-24
PCT/EP2008/054912 WO2008129058A1 (en) 2007-04-24 2008-04-23 Duck embryonic derived stem cell lines for the production of viral vaccines

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/054912 A-371-Of-International WO2008129058A1 (en) 2007-04-24 2008-04-23 Duck embryonic derived stem cell lines for the production of viral vaccines

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/069,423 Division US9260694B2 (en) 2007-04-24 2013-11-01 Generation of duck cell lines

Publications (1)

Publication Number Publication Date
US20100062489A1 true US20100062489A1 (en) 2010-03-11

Family

ID=38521810

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/597,486 Abandoned US20100062489A1 (en) 2007-04-24 2008-04-23 Duck embryonic derived stem cell lines for the production of viral vaccines
US14/069,423 Active US9260694B2 (en) 2007-04-24 2013-11-01 Generation of duck cell lines
US15/013,079 Active US9822345B2 (en) 2007-04-24 2016-02-02 Method of making a virus using duck embryonic derived stem cell lines
US15/785,530 Abandoned US20180135026A1 (en) 2007-04-24 2017-10-17 Duck embryonic derived stem cell lines for the production of viral vaccines

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/069,423 Active US9260694B2 (en) 2007-04-24 2013-11-01 Generation of duck cell lines
US15/013,079 Active US9822345B2 (en) 2007-04-24 2016-02-02 Method of making a virus using duck embryonic derived stem cell lines
US15/785,530 Abandoned US20180135026A1 (en) 2007-04-24 2017-10-17 Duck embryonic derived stem cell lines for the production of viral vaccines

Country Status (23)

Country Link
US (4) US20100062489A1 (sl)
EP (4) EP1985305A1 (sl)
JP (1) JP5539185B2 (sl)
KR (2) KR101624678B1 (sl)
CN (1) CN101668539B (sl)
AU (1) AU2008240708B2 (sl)
BR (1) BRPI0809842B8 (sl)
CA (1) CA2684845C (sl)
CY (1) CY1114736T1 (sl)
DK (1) DK2150275T3 (sl)
EA (1) EA021064B1 (sl)
ES (2) ES2574827T3 (sl)
HK (1) HK1140964A1 (sl)
HR (1) HRP20131203T1 (sl)
IL (1) IL201679A (sl)
MX (1) MX343247B (sl)
MY (1) MY151119A (sl)
NZ (1) NZ581321A (sl)
PL (1) PL2150275T3 (sl)
PT (1) PT2150275E (sl)
SI (1) SI2150275T1 (sl)
WO (1) WO2008129058A1 (sl)
ZA (1) ZA200908152B (sl)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100221825A1 (en) * 2002-03-08 2010-09-02 Vivalis Avian cell lines useful for the production of substances of interest
US20100235937A1 (en) * 2006-08-09 2010-09-16 Isabelle Valarche Production of transgenic avian organisms employing embryonic stem cells
WO2011051235A1 (en) 2009-10-27 2011-05-05 Glaxosmithkline Biologicals, Niederlassung Der Smithkline Beecham Pharma Gmbh & Co. Kg Process for producing influenza vaccine
US9260694B2 (en) 2007-04-24 2016-02-16 Valneva Generation of duck cell lines
US9381238B2 (en) 2010-11-05 2016-07-05 University Of Dundee Method for improving the production of influenza viruses and vaccine seeds
US9809800B2 (en) 2012-11-22 2017-11-07 Asahi Kasei Medical Co., Ltd. Method for producing parvovirus having high infectivity titer
US9840559B2 (en) 2013-02-01 2017-12-12 The Regents Of The University Of California Anti-CD83 antibodies and use thereof
CN112094819A (zh) * 2020-08-13 2020-12-18 浙江美保龙生物技术有限公司 一种鸡传染性法氏囊病毒的全悬浮培养方法
US10870704B2 (en) 2014-10-23 2020-12-22 Kira Biotech Pty Limited CD83 binding proteins and uses thereof

Families Citing this family (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102016027A (zh) * 2008-02-25 2011-04-13 巴克斯特国际公司 生产连续细胞系的方法
EP2361304A2 (en) * 2008-11-05 2011-08-31 GlaxoSmithKline Biologicals S.A. Novel method
ES2539045T3 (es) 2009-01-19 2015-06-25 Innate Pharma Anticuerpos anti-KIR3D
WO2010089339A1 (en) 2009-02-06 2010-08-12 Glaxosmithkline Biologicals S.A. Purification of virus or viral antigens by density gradient ultracentrifugation
EP2427547A1 (en) * 2009-05-08 2012-03-14 GlaxoSmithKline Biologicals S.A. Method for producing virus from cell culture involving homogenization
EP2429579A1 (en) * 2009-05-12 2012-03-21 Transgene SA Immortalized avian cell lines and use thereof
GB0919117D0 (en) * 2009-10-30 2009-12-16 Glaxosmithkline Biolog Sa Process
CA3177833A1 (en) 2009-12-15 2011-06-15 University Of Saskatchewan Vaccines for inclusion body hepatitis
CN102120983B (zh) * 2010-01-07 2013-04-03 中国农业科学院北京畜牧兽医研究所 北京鸭表皮干细胞的分离和培养方法
US20120309056A1 (en) 2010-02-04 2012-12-06 Leon Arnaud Fed-batch process using concentrated cell culture medium for the efficient production of biologics in eb66 cells
FR2956409A1 (fr) 2010-02-15 2011-08-19 Univ Claude Bernard Lyon Procedes pour optimiser la production virale par l'inactivation ou l'inhibition du gene hipk2 et cellules modifiees pour la production virale
CA2793772A1 (en) * 2010-03-26 2011-09-29 Emergent Product Development Gaithersburg Inc. Ectodomains of influenza matrix 2 protein, expression system, and uses thereof
US20130039943A1 (en) * 2010-05-03 2013-02-14 Bruno Rene Andre Novel method
GB201011502D0 (en) 2010-07-08 2010-08-25 Glaxosmithkline Biolog Sa Novel process
NZ604510A (en) 2010-08-17 2013-10-25 Csl Ltd Dilutable biocidal compositions and methods of use
CA2985099A1 (en) * 2010-10-15 2012-04-19 Tokyo Metropolitan Institute Of Medical Science Recombinant vaccinia virus having hemagglutinin protein genes derived from h1n1 influenza virus
WO2012095514A1 (en) 2011-01-14 2012-07-19 Vivalis Recombinant protein production system
JP2014518080A (ja) 2011-06-27 2014-07-28 バルネバ 細胞のスクリーニング方法
EP2760889A4 (en) 2011-09-30 2015-04-15 Teva Pharmaceuticals Australia Pty Ltd ANTIBODIES AGAINST TL1A AND USES THEREOF
AU2012327878A1 (en) 2011-10-28 2014-05-29 Patrys Limited PAT-LM1 epitopes and methods for using same
CN103157107A (zh) * 2011-12-14 2013-06-19 普莱柯生物工程股份有限公司 用连续传代细胞系生产鸡马立克氏病疫苗及方法
AR089231A1 (es) * 2011-12-15 2014-08-06 Amgen Inc Metodo de floculacion
EP2660316A1 (en) 2012-05-02 2013-11-06 Helmholtz-Zentrum für Infektionsforschung GmbH Avian cell line and its use in production of protein
CN117843785A (zh) 2013-02-07 2024-04-09 Csl有限公司 Il-11r结合蛋白及其应用
FR3008992A1 (fr) * 2013-07-25 2015-01-30 Agronomique Inst Nat Rech Procede de selection d'une lignee cellulaire permissive pour la replication de virus aviaires
JP6851827B2 (ja) * 2013-08-30 2021-03-31 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム 細胞培養中でのウイルスの大規模製造
CN105177176A (zh) * 2014-06-13 2015-12-23 亚宝药业太原制药有限公司 一种腺病毒滴度的检测方法
CA2954841A1 (en) 2014-07-16 2016-01-21 Transgene Sa Oncolytic virus for expression of immune checkpoint modulators
FR3025107B1 (fr) 2014-08-29 2018-10-05 Calixar Procede de preparation d'un antigene vaccinal, antigene vaccinal obtenu et utilisations
AU2015336946A1 (en) 2014-10-23 2017-04-13 La Trobe University Fn14-binding proteins and uses thereof
JP6865167B2 (ja) 2014-12-17 2021-04-28 フンダシオン パラ ラ インベスティガシオン メディカ アプリカダ ウィルソン病の処置に使用するための核酸構築物及び遺伝子治療ベクター
WO2016097218A1 (en) 2014-12-17 2016-06-23 Fundación Para La Investigación Mèdica Aplicada Nucleic acid constructs and gene therapy vectors for use in the treatment of wilson's disease and other conditions
WO2016131945A1 (en) 2015-02-20 2016-08-25 Transgene Sa Combination product with autophagy modulator
CN114716552B (zh) 2016-01-11 2024-05-24 四十七公司 人源化、小鼠或嵌合抗cd47单克隆抗体
EP3452081A1 (en) 2016-05-04 2019-03-13 Transgene SA Combination therapy with cpg tlr9 ligand
CN107137704B (zh) * 2016-08-20 2020-02-21 山东信得科技股份有限公司 一种鸭2型腺病毒灭活疫苗
RU2756534C2 (ru) * 2016-09-28 2021-10-01 Бавариан Нордик А/С Композиции и способы для повышения стабильности трансгенов в поксвирусах
US20190328869A1 (en) 2016-10-10 2019-10-31 Transgene Sa Immunotherapeutic product and mdsc modulator combination therapy
WO2018091680A1 (en) 2016-11-18 2018-05-24 Transgene Sa Cowpox-based oncolytic vectors
CN106692964A (zh) * 2016-12-05 2017-05-24 扬州大学 一种血清4型禽腺病毒灭活苗及其制备方法
KR20190097240A (ko) 2016-12-28 2019-08-20 트랜스진 에스.에이. 종양용해성 바이러스 및 치료 분자
CN107058243A (zh) * 2017-03-23 2017-08-18 华南农业大学 一种马立克氏病毒的悬浮培养方法
AU2018270375A1 (en) 2017-05-15 2019-10-31 Bavarian Nordic A/S Stable virus-containing composition
JP2020519666A (ja) 2017-05-15 2020-07-02 ヤンセン ファッシンズ アンド プリベンション ベーフェーJanssen Vaccines & Prevention B.V. 安定性のウイルス含有組成物
EP3634152A1 (en) 2017-06-07 2020-04-15 Wild Type, Inc. Ex vivo meat production
JP7334124B2 (ja) 2017-06-21 2023-08-28 トランジェーヌ 個別化ワクチン
WO2019020543A1 (en) 2017-07-28 2019-01-31 Transgene Sa ONCOLYTIC VIRUSES EXPRESSING AGENTS TARGETING METABOLIC IMMUNE MODULATORS
CN107794248A (zh) * 2017-10-24 2018-03-13 广州齐志生物工程设备有限公司 一种用微载体悬浮培养犬瘟热病毒的方法
KR102121042B1 (ko) * 2017-12-06 2020-06-09 이정복 조류의 에그로부터 분리한 세포인 배반엽 전분화능세포, 신경줄기세포, 배아섬유아세포 컨디션드 배지를 유효성분으로 하는 세포 증식, 피부재생 및 주름 개선용 배지 조성물
CN108220227A (zh) * 2017-12-27 2018-06-29 华农(肇庆)生物产业技术研究院有限公司 一种通过全悬浮传代细胞系悬浮培养新城疫病毒的方法
CN108159412A (zh) * 2018-01-12 2018-06-15 华农(肇庆)生物产业技术研究院有限公司 一种生产细胞源鸡新城疫、禽流感二联灭活疫苗的方法及其产品
CN108261543B (zh) * 2018-02-08 2019-03-08 华农(肇庆)生物产业技术研究院有限公司 传代细胞源nd、ib、ai三联灭活疫苗的制备方法及其应用
CN108187039A (zh) * 2018-03-29 2018-06-22 山东信得动物疫苗有限公司 一种禽流感灭活疫苗生产工艺及产品
WO2019219649A1 (en) 2018-05-14 2019-11-21 Vivet Therapeutics Gene therapy vectors comprising s/mar sequences
WO2020011754A1 (en) 2018-07-09 2020-01-16 Transgene Chimeric vaccinia viruses
CA3106286A1 (en) 2018-07-13 2020-01-16 Valneva Se Method for rescuing and producing a virus in avian cells
EP3617230A1 (en) 2018-09-03 2020-03-04 BioInvent International AB Novel antibodies and nucleotide sequences, and uses thereof
CN108977554B (zh) * 2018-09-05 2021-08-31 湖北省农业科学院畜牧兽医研究所 蛋鸭环状RNA circ_13034及其检测试剂、方法与应用
EP3847246A1 (en) 2018-09-06 2021-07-14 Bavarian Nordic A/S Storage improved poxvirus compositions
CA3116098A1 (en) 2018-10-12 2020-04-16 Vivet Therapeutics Codon-optimized transgene for the treatment of progressive familiar intrahepatic cholestasis type 3 (pfic3)
CN112955186A (zh) 2018-11-07 2021-06-11 维韦特治疗公司 用于治疗进行性家族性肝内胆汁淤积症2型(pfic2)的密码子优化的abcb11转基因
TW202039854A (zh) 2018-11-16 2020-11-01 美商編碼製藥公司 治療威爾遜氏病的組合物和方法
CA3120166A1 (en) 2018-11-23 2020-05-28 Valneva Se Food products comprising avian stem cells
WO2020108735A1 (en) 2018-11-26 2020-06-04 Blink Biomedical Sas Antibodies to cytomegalovirus interleukin-10
CN113423815A (zh) * 2018-12-06 2021-09-21 创赏有限公司 非动物来源培养基上的细菌生长
WO2020123876A1 (en) 2018-12-12 2020-06-18 Wild Type, Inc. Synthetic food compositions
CA3124773A1 (en) 2018-12-28 2020-07-02 Transgene M2-defective poxvirus
CN109593729A (zh) * 2018-12-29 2019-04-09 肇庆大华农生物药品有限公司 一种禽减蛋综合征病毒培养方法及其在疫苗中的应用
BR112021014116A2 (pt) * 2019-01-17 2021-09-21 Boehringer Ingelheim Animal Health USA Inc. Meio sem soro para produção de vacinas aviárias e seus usos
JP7212588B2 (ja) 2019-06-20 2023-01-25 オルガノ株式会社 水処理装置
JP2022544740A (ja) 2019-07-02 2022-10-21 フンダシオン・パラ・ラ・インベスティガシオン・メディカ・アプリカダ cPLA2e誘導剤及びその使用
IL296250A (en) 2020-03-12 2022-11-01 Bavarian Nordic As The compounds that improve the stability of the smallpox virus
WO2022013221A1 (en) 2020-07-13 2022-01-20 Transgene Treatment of immune depression
CA3181024A1 (en) 2020-08-06 2022-02-10 Fundacion Para La Investigacion Medica Aplicada Viral particles for use in treating tauopathies such as alzheimer's diseases by gene therapy
US20230265456A1 (en) 2020-08-10 2023-08-24 Fundacion Para La Investigacion Medica Aplicada Gene therapy vector expressing cyp27a1 for the treatment of cerebrotendinous xanthomatosis
BR112023004874A2 (pt) 2020-10-09 2023-05-02 UCB Biopharma SRL Construções de ácido nucléico, vetores virais e partículas virais
CN112442486B (zh) * 2020-11-05 2023-02-10 上海奥浦迈生物科技股份有限公司 一种维持体外培养cho dg44细胞后期活率的培养基及其应用
CN113699119A (zh) * 2020-12-10 2021-11-26 华农(肇庆)生物产业技术研究院有限公司 一种新城疫病毒及疫苗的无血清全悬浮培养制备方法
US20240091382A1 (en) 2020-12-23 2024-03-21 Vivet Therapeutics Minimal bile acid inducible promoters for gene therapy
WO2022148736A1 (en) 2021-01-05 2022-07-14 Transgene Vectorization of muc1 t cell engager
CN113278595B (zh) * 2021-04-28 2023-05-09 重庆永健生物技术有限责任公司 一种鸭腺病毒3型菌株、鸭腺病毒卵黄抗体及其制备方法和应用
WO2023025899A2 (en) 2021-08-26 2023-03-02 Transgene Delivery system for targeting genes of the interferon pathway
TW202321458A (zh) 2021-09-22 2023-06-01 瑞典商生物創新國際公司 新穎抗體組合及其用途
CA3234666A1 (en) 2021-10-28 2023-05-04 UCB Biopharma SRL Nucleic acid constructs, viral vectors and viral particles
WO2023213763A1 (en) 2022-05-02 2023-11-09 Transgene Poxvirus encoding a binding agent comprising an anti- pd-l1 sdab
WO2023213764A1 (en) 2022-05-02 2023-11-09 Transgene Fusion polypeptide comprising an anti-pd-l1 sdab and a member of the tnfsf
WO2024003353A1 (en) 2022-07-01 2024-01-04 Transgene Fusion protein comprising a surfactant-protein-d and a member of the tnfsf
CN115449502B (zh) * 2022-08-15 2023-10-20 广东省华晟生物技术有限公司 无血清全悬浮培养型bhk-21-c细胞株及其构建方法与应用
WO2024038175A1 (en) 2022-08-18 2024-02-22 Transgene Chimeric poxviruses

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US534740A (en) * 1895-02-26 Indicating door-bell
US5830510A (en) * 1992-05-15 1998-11-03 North Carolina State University Veterinary pharmaceutical formulation containing avian embryonic stem cells
US6114168A (en) * 1994-10-21 2000-09-05 Institute National De La Recherche Agronomique Active retinoic acid-free culture medium for chicken embryonic stem cells
WO2003076601A1 (fr) * 2002-03-08 2003-09-18 Vivalis Lignées de cellules aviaires utiles pour la production de substances d'intérêt
US6656479B2 (en) * 1996-11-12 2003-12-02 Pfizer Inc. Attenuated live neospora vaccine
US20040077086A1 (en) * 2002-07-09 2004-04-22 Manfred Reiter Animal protein free media for cultivation of cells
WO2005007840A1 (en) * 2003-07-22 2005-01-27 Vivalis Production of poxviruses with adherent or non adherent avian cell lines
US8148132B2 (en) * 2005-04-11 2012-04-03 Vivalis Production of viral vaccines in suspension on avian embryonic derived stem cell lines

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU623922B2 (en) 1988-08-04 1992-05-28 Amrad Operations Pty. Limited In vitro propagation of embryonic stem cells
US5162215A (en) 1988-09-22 1992-11-10 Amgen Inc. Method of gene transfer into chickens and other avian species
JPH05227947A (ja) 1992-01-14 1993-09-07 Rikagaku Kenkyusho 鳥類原始生殖細胞の分離方法
AU3597093A (en) 1992-01-27 1993-09-01 Embrex Inc. Gene transfer in poultry by introduction of embryo cells (in ovo)
WO1994003585A1 (en) 1992-08-04 1994-02-17 Commonwealth Scientific And Industrial Research Organisation A method for maintaining embryonic stem cells and avian factor useful for same
US5453357A (en) 1992-10-08 1995-09-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
US5589458A (en) 1992-11-13 1996-12-31 Thomas Jefferson University Compounds that inhibit T cell proliferation and methods for using the same
JPH0924997A (ja) 1995-07-11 1997-01-28 Tokico Ltd 出荷装置
US6103529A (en) 1996-10-10 2000-08-15 Life Technologies, Inc. Animal cell culture media comprising peptides derived from rice
WO1999006534A1 (en) 1997-08-04 1999-02-11 University Of Massachusetts, A Public Institution Of Higher Education Of The Commonwealth Of Massachusetts, Represented By Its Amherst Campus Production of avian embryonic germ (eg) cell lines by prolonged culturing of pgcs, use thereof for cloning and chimerization
JP2003532364A (ja) 1997-08-04 2003-11-05 ユニバシティ オブ マサチューセッツ、ア パブリック インスチチユーション オブ ハイアー エデュケイション オブ ザ コモンウエルス オブ マサチューセッツ、アズ リプリゼンテッド バイ イッツ アマースト キャンパス トリ始原生殖細胞(pgc)細胞系とその長期間培養法
US6406909B1 (en) 1998-07-10 2002-06-18 Chugai Seiyaku Kabushiki Kaisha Serum-free medium for culturing animal cells
DE60037107T2 (de) 1999-02-11 2008-09-25 Hanmi Pharm. Co., Ltd. Pluripotente embryonale keimzellinie aus vögeln
DE19947407C2 (de) 1999-10-01 2002-08-01 Bayerische Motoren Werke Ag Datenbussystem für Kraftfahrzeuge
EP1500699A1 (en) * 2003-07-22 2005-01-26 Vivalis Production of vaccinia virus with adherent or non adherent avian cell lines
EP1528101A1 (en) * 2003-11-03 2005-05-04 ProBioGen AG Immortalized avian cell lines for virus production
ATE477321T1 (de) * 2006-01-05 2010-08-15 Transgene Sa Aviäre telomerase reverse transkriptase
WO2007135133A1 (en) * 2006-05-19 2007-11-29 Vivalis Avian cell lines derived from primordial germ cells useful for the production of substances of interest
US8962311B2 (en) 2006-08-09 2015-02-24 Valneva Method of obtaining chicken embryonic stem cells
EP1985305A1 (en) 2007-04-24 2008-10-29 Vivalis Duck embryonic derived stem cell lines for the production of viral vaccines

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US534740A (en) * 1895-02-26 Indicating door-bell
US5830510A (en) * 1992-05-15 1998-11-03 North Carolina State University Veterinary pharmaceutical formulation containing avian embryonic stem cells
US6114168A (en) * 1994-10-21 2000-09-05 Institute National De La Recherche Agronomique Active retinoic acid-free culture medium for chicken embryonic stem cells
US6998266B2 (en) * 1994-10-21 2006-02-14 Institute National De La Recherche Agronomique Active retinoic acid-free culture medium for avian totipotent embryonic stem cells
US6656479B2 (en) * 1996-11-12 2003-12-02 Pfizer Inc. Attenuated live neospora vaccine
WO2003076601A1 (fr) * 2002-03-08 2003-09-18 Vivalis Lignées de cellules aviaires utiles pour la production de substances d'intérêt
US20110294209A1 (en) * 2002-03-08 2011-12-01 Vivalis Methods for producing nonadherent avian cell lines
US20040077086A1 (en) * 2002-07-09 2004-04-22 Manfred Reiter Animal protein free media for cultivation of cells
WO2005007840A1 (en) * 2003-07-22 2005-01-27 Vivalis Production of poxviruses with adherent or non adherent avian cell lines
US20100111999A1 (en) * 2003-07-22 2010-05-06 Vivalis Production of poxviruses with adherent or non adherent avian cell lines
US7771980B2 (en) * 2003-07-22 2010-08-10 Vivalis Production of poxviruses with adherent or non adherent avian cell lines
US8148132B2 (en) * 2005-04-11 2012-04-03 Vivalis Production of viral vaccines in suspension on avian embryonic derived stem cell lines

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Horiuchi, J Biol Chem. 279(23):24514-20, 2004 *
Pain (Development, 1996, Vol. 122, No. 8, pg 2339-2348) *
Petitte, Mech. Dev. 2004, Vol. 121, No. 9, pg 1159-68 *
Trentin (PNAS, March 30, 2004, Vol. 101, No. 13, pg 4495-4500) *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100221825A1 (en) * 2002-03-08 2010-09-02 Vivalis Avian cell lines useful for the production of substances of interest
US9382513B2 (en) 2002-03-08 2016-07-05 Valneva Method of making an avian cell line
US8962311B2 (en) 2006-08-09 2015-02-24 Valneva Method of obtaining chicken embryonic stem cells
US20100235937A1 (en) * 2006-08-09 2010-09-16 Isabelle Valarche Production of transgenic avian organisms employing embryonic stem cells
US9260694B2 (en) 2007-04-24 2016-02-16 Valneva Generation of duck cell lines
US9822345B2 (en) 2007-04-24 2017-11-21 Valneva Method of making a virus using duck embryonic derived stem cell lines
WO2011051235A1 (en) 2009-10-27 2011-05-05 Glaxosmithkline Biologicals, Niederlassung Der Smithkline Beecham Pharma Gmbh & Co. Kg Process for producing influenza vaccine
US9381238B2 (en) 2010-11-05 2016-07-05 University Of Dundee Method for improving the production of influenza viruses and vaccine seeds
US9809800B2 (en) 2012-11-22 2017-11-07 Asahi Kasei Medical Co., Ltd. Method for producing parvovirus having high infectivity titer
US9840559B2 (en) 2013-02-01 2017-12-12 The Regents Of The University Of California Anti-CD83 antibodies and use thereof
EP3626743A1 (en) 2013-02-01 2020-03-25 Kira Biotech Pty Limited Anti-cd83 antibodies and use thereof
US10781255B2 (en) 2013-02-01 2020-09-22 Dendrocyte Biotech Pty Ltd Anti-CD83 antibodies and use thereof
US11725054B2 (en) 2013-02-01 2023-08-15 Kira Biotech Pty Limited Anti-CD83 antibodies and use thereof
US10870704B2 (en) 2014-10-23 2020-12-22 Kira Biotech Pty Limited CD83 binding proteins and uses thereof
CN112094819A (zh) * 2020-08-13 2020-12-18 浙江美保龙生物技术有限公司 一种鸡传染性法氏囊病毒的全悬浮培养方法

Also Published As

Publication number Publication date
KR101624678B1 (ko) 2016-05-26
WO2008129058A1 (en) 2008-10-30
ES2574827T3 (es) 2016-06-22
HRP20131203T1 (hr) 2014-02-14
US20180135026A1 (en) 2018-05-17
EA021064B1 (ru) 2015-03-31
US20160257940A1 (en) 2016-09-08
KR20100017340A (ko) 2010-02-16
IL201679A (en) 2013-11-28
US9260694B2 (en) 2016-02-16
EP2150275B1 (en) 2013-11-13
ZA200908152B (en) 2010-04-28
HK1140964A1 (en) 2010-10-29
EP2572728A1 (en) 2013-03-27
PL2150275T3 (pl) 2014-03-31
EP2572727B1 (en) 2016-05-18
MY151119A (en) 2014-04-15
CA2684845C (en) 2017-12-05
NZ581321A (en) 2012-05-25
CN101668539B (zh) 2014-02-19
US20140154741A1 (en) 2014-06-05
CY1114736T1 (el) 2016-12-14
DK2150275T3 (da) 2014-01-13
BRPI0809842A2 (pt) 2014-09-23
EP1985305A1 (en) 2008-10-29
BRPI0809842B1 (pt) 2019-09-10
ES2442009T3 (es) 2014-02-07
KR20150036788A (ko) 2015-04-07
IL201679A0 (en) 2010-05-31
AU2008240708B2 (en) 2013-04-18
AU2008240708A1 (en) 2008-10-30
MX343247B (es) 2016-10-31
SI2150275T1 (sl) 2014-04-30
CN101668539A (zh) 2010-03-10
JP5539185B2 (ja) 2014-07-02
EP2150275A1 (en) 2010-02-10
PT2150275E (pt) 2014-01-10
MX2009011505A (es) 2009-11-09
JP2010524482A (ja) 2010-07-22
EP2572727A1 (en) 2013-03-27
US9822345B2 (en) 2017-11-21
EA200970996A1 (ru) 2010-04-30
BRPI0809842A8 (pt) 2019-07-16
CA2684845A1 (en) 2008-10-30
BRPI0809842B8 (pt) 2021-05-25

Similar Documents

Publication Publication Date Title
US9822345B2 (en) Method of making a virus using duck embryonic derived stem cell lines
RU2457253C2 (ru) Способ репликации вируса в птичьих эмбриональных стволовых клетках
AU2011253998B2 (en) Process of manufacturing viral vaccines in suspension avian embryonic derived stem cell lines

Legal Events

Date Code Title Description
AS Assignment

Owner name: VIVALIS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GUEHENNEUX, FABIENNE;MOREAU, KARINE;ESNAULT, MAGALI;AND OTHERS;REEL/FRAME:025185/0852

Effective date: 20091022

AS Assignment

Owner name: VALNEVA SE, FRANCE

Free format text: CHANGE OF NAME;ASSIGNOR:VIVALIS SA;REEL/FRAME:031607/0481

Effective date: 20130528

AS Assignment

Owner name: VALNEVA, FRANCE

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE CHANGE OF NAME RECORDATION PREVIOUSLY RECORDED ON REEL 031607 FRAME 0481. ASSIGNOR(S) HEREBY CONFIRMS THE CONVEYING PARTY: VIVALIS RECEIVING PARTY: VALNEVA;ASSIGNOR:VIVALIS;REEL/FRAME:031804/0451

Effective date: 20130528

STCB Information on status: application discontinuation

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