US20040058441A1 - Avian cell lines useful for the production of substances of interest - Google Patents

Avian cell lines useful for the production of substances of interest Download PDF

Info

Publication number
US20040058441A1
US20040058441A1 US10/625,847 US62584703A US2004058441A1 US 20040058441 A1 US20040058441 A1 US 20040058441A1 US 62584703 A US62584703 A US 62584703A US 2004058441 A1 US2004058441 A1 US 2004058441A1
Authority
US
United States
Prior art keywords
virus
cells
viruses
medium
serum
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
US10/625,847
Other languages
English (en)
Inventor
Bertrand Pain
Fabienne Guehenneux
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.)
Vivalis SA
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
Assigned to VIVALIS reassignment VIVALIS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GUEHENNEUX, FABIENNE, PAIN, BERTRAND
Publication of US20040058441A1 publication Critical patent/US20040058441A1/en
Priority to US12/350,781 priority Critical patent/US20090239297A1/en
Priority to US12/717,096 priority patent/US9382513B2/en
Priority to US13/206,056 priority patent/US20110294209A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • 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
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • C12N2500/92Medium free of human- or animal-derived components
    • 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
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • C12N2500/95Protein-free medium and culture conditions
    • 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
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production

Definitions

  • the present invention relates to a method for producing avian cell lines, in particular avian stem cells, comprising progressive or total withdrawal of growth factors, serum and/or feeder layer. These spontaneously established lines are adherent or nonadherent cells capable of proliferating indefinitely in a basic culture medium.
  • the invention also relates to the cells derived from such lines which are particularly useful for the production of vaccines and of substances of interest.
  • Stem cells are cells identified by their culture in vitro from an embryo, from part of an embryo or even from an adult tissue.
  • the expression stem cell is understood to mean any pluripotent cell of embryonic or adult origin which has a capacity for self-renewal and is capable of giving specialized differentiated cells.
  • any noncancerous cell capable of dividing indefinitely in culture and of giving a daughter cell having the same capacity for proliferation and differentiation as the mother cell from which it is derived.
  • These isolated cells exhibit particular morphological and immunocytochemical characteristics. It is also possible to distinguish the notion of:
  • CES cells embryonic stem cells
  • stem cells which have the characteristic feature of being obtained from culturing parts or all of a very early embryo (blastula stage).
  • CES cells exhibit in vitro all the characteristics of a stem cell, and in vivo the unique capacity of contributing to the morphogenesis of an embryo and of participating in germline colonization when they are reimplanted in any manner whatsoever in a recipient embryo.
  • somatic stem cells SSC
  • CES cells somatic stem cells
  • stem cells do not exhibit an easily identifiable characteristic state of morphological differentiation (fibroblasts, adipocytes, macrophage, and the like), but are rather characterized by a state of proliferation and of nondifferentiation. This state results in different behaviors such as a rapid proliferation in vitro, a characteristic morphology, the presence of different markers, variable requirements for growth factors and an ability to respond to particular stimuli for induction of differentiation. They are not sensitive to replicative senescence, a critical period for a large number of differentiated primary cells, including the fibroblasts for example.
  • This arrest often corresponds to a replicative senescence, known by the term Hayflick limit.
  • This stoppage is thought to be the result of the action of a true molecular clock of which one of the key components is thought to be the length of the telomeres.
  • the telomeres are repeat sequences situated at the end of the chromosomes. The shortening of these repetitive nucleotide structures is the consequence of the replication of DNA on a semiconservative mode.
  • telomeres In the absence of the telomerase enzyme, which is in charge of adding the repeat sequences at the end of the chromosomes, a point of no-return is reached with regard to the size of the telomeres, a point beyond which an as yet unknown molecular mechanism for activation of genes involved in controlling the cell cycle is triggered.
  • the cells are then thought to be blocked in the G1 phase in their divisions and are thought to stop proliferating. Numerous factors appear to be involved in this negative control of the cell cycle such as various cyclins, specific kinases, RB and P53 proteins, specific transcription factors such as E2F and many others (mdm2, BTG, p21, and the like).
  • the telomerase enzyme can therefore be viewed as a central factor in cell immortality because it maintains the length of the telomeres and therefore makes the cell insensitive to this loss caused by successive divisions.
  • telomere activity In an organism under development and during the life of this organism, only a few cell types, including certain lymphocytes, exhibit a permanent expression of telomerase. This activity also appears to be one of the characteristics of stem cells, both at the somatic level (SSC) and at the germline level. This property of expressing, of maintaining of expression and of “awakening” expression of the telomerase activity is also often associated with the immortal character of a cell maintained in vitro. To date, numerous cancer cells are also detected positive for telomerase activity. This activity is thought to be partially responsible for the capacity for uncontrolled proliferation of tumor cells in vivo.
  • telomere activity is, in all cases, an excellent marker for the stem cell character and for the germline lineage and for the capacity of a cell to become immortal. Two criteria are therefore used: the telomerase activity and the size of the telomeres.
  • the establishment of cell lines may be carried out according to two routes: a spontaneous establishment resulting from noninduced intrinsic genetic damage or a triggered establishment, induced by the use of viruses, retroviruses or by other means such as chemical agents, irradiation, UV (ultraviolet) radiation, and the like.
  • rodent (mouse, rat, and the like) cells is recognized as being fairly easy spontaneously; on the other hand, the situation is quite different for human cells regardless of their tissue origin (Smith and Pereira-Smith, 1996).
  • a first step leads the proliferating cell to the Hayflick limit which, depending on the cell types, is between 10 and 50 passages.
  • a first spontaneous mutational event then takes place which allows the cell to cross this first blockage, an event which often affects the p53 and pRb genes, and the like.
  • the cells therefore continue to proliferate until the moment when a second blockage occurs, which is in general lifted by new mutations in other genes and by the activation of telomerase, which is often observed.
  • lymphoblastoid lines DT40 and DT95 obtained in the presence of the avian leukosis virus (ALV) and in which the myc locus is activated (Baba et al., 1985, ATCC No. CRL 2111, CRL 2112),
  • lymphoblastoid line ConA-C1 established with the REV virus (reticuloendothelial virus, ATCC No. 12135, U.S. Pat. No. 5,691,200),
  • immortalizing genes adenovirus E1A gene, polyoma SV40 “large T”, and the like
  • gene fragments have also made it possible to obtain lines from already differentiated primary cells.
  • These components may be introduced into the cells by simple transfection of a vector allowing the expression of the immortalizing part, but may also be introduced via viruses or retroviruses which have been genetically modified to express these immortalizing components.
  • the origin of the immortalizing components may be avian or otherwise, viral or otherwise.
  • the tropism for avian cells can in fact be linked to the original virus or can also be modified.
  • the duck fibroblast line TDF-2A is thus obtained by introducing a first immortalizing gene and then an antiapoptotic gene (Guilhot et al., 1993, U.S. Pat. No. 6,255,108).
  • Other methods have been developed, such as the overexpression of p53 (Foster et al., U.S. Pat. No. 5,830,723).
  • the expression immortalization event is understood to mean various actions such as:
  • the present invention describes the production of lines which can become spontaneously nonadherent and for which the nonadherence is obtained by a withdrawal of the feeder layer. Because of their growth in suspension, these lines are perfectly suitable for industrial use for the production of substances of interest in bioreactors.
  • the present invention relates to a method for producing avian cell lines, characterized in that it comprises the following steps:
  • the expression “establishment of a line” is understood to mean maintaining cells in culture in vitro over a considerable period of time.
  • the cells derived from the lines obtained in step c) are capable of proliferating for at least 50 days, 100 days, 150 days, 300 days or preferably at least 600 days.
  • the 600 days do not constitute a time limit because the cell lines obtained are still alive after much longer time periods.
  • these lines are considered as being able to grow indefinitely in a basic culture medium free of exogenous growth factors, serum and/or inactivated feeder layer.
  • 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.
  • the cells derived from the lines according to the invention may be avian stem cells, in particular avian somatic stem cells.
  • the stem cells according to the invention can serve to obtain differentiated cell lines. Indeed, these stem cells have the property of being pluripotent, that is to say that they have the potential to be induced in multiple differentiation pathways which can be characterized by various specific markers.
  • These cells can also be precursor cells, which correspond to the partially differentiated cells of an adult or embryonic tissue, by contrast to a stem cell and which is capable of dividing and of giving more differentiated cells.
  • the expression “differentiated cell” is understood to mean any specialized cell of an adult or embryonic tissue, having specific markers or fulfilling specific physiological functions. It is possible, in a particular aspect of the invention, in particular for particular isolates or clones derived from a particular isolate obtained during establishment, for these stem cells to contribute to the germline. In this case, these stem cells established as lines are thought to be embryonic stem cells.
  • the invention relates to a method as defined above, in which the established lines are adherent stem cells which proliferate in the absence of inactivated feeder layer.
  • step b) consists in a withdrawal of the components of the medium (growth factors alone or serum alone or growth factors and then serum or alternatively serum and then growth factors).
  • the components of the medium growth factors alone or serum alone or growth factors and then serum or alternatively serum and then growth factors.
  • the invention relates to a method as defined above in which the established lines are nonadherent stem cells which proliferate in suspension in a medium free of exogenous growth factors.
  • step b) consists in a progressive or total withdrawal of the feeder layer and then optionally in a withdrawal of the other components of the medium (growth factors and serum).
  • the invention relates to a method as described above in which the established lines are nonadherent stem cells which proliferate in suspension in a medium free of serum (serum-free medium).
  • the invention relates to a method as defined above, in which the established lines are nonadherent stem cells which proliferate in suspension in a medium free of exogenous growth factors and serum.
  • step b) consists in a progressive or total withdrawal of the growth factors, optionally followed by a progressive withdrawal of the serum.
  • step b) consists in a progressive or total withdrawal of the growth factors and/or serum, optionally followed by a withdrawal of the feeder layer.
  • the established lines may be cells which proliferate in a serum-depleted medium, in particular in a medium free of serum.
  • serum-depleted is understood to mean a gradual reduction of the concentration of serum spread out over time. This method allows a selection of clones which adapt to these new, increasingly drastic conditions until stable lines are obtained which are capable of growing in a serum-depleted medium or in a medium completely free of serum.
  • the method described above may additionally comprise a step in which the cells obtained in step c) are subjected to a selection in culture media used for large-scale production so as to obtain clones suitable for the production of vaccines intended for human or animal therapy.
  • the cells according to the invention have at least one of the following characteristics:
  • the cells of the invention have all the abovementioned characteristics.
  • the invention relates to a method for producing avian lines, which is mentioned above, in which the cells derived from the lines obtained in step c) are modified in order to allow a better use in vitro such as the extension of the greater life span or growth densities or alternatively of the lower nutrient requirements.
  • the cells derived from established lines are modified in order to produce a substance of interest, in particular a polypeptide of interest, an antibody or an attenuated virus.
  • Said cells may be modified by any technique accessible to persons skilled in the art, in particular homologous, directed and/or conditional recombination (Cre-Lox or FLP-FRT system), by transformation with any vector, plasmid, in particular with the aid of retroviruses.
  • the medium used in step a) may comprise at least one factor selected from cytokines, in particular LIF, IL-11, IL-6, IL-6R, CNTF, Oncostatin and other factors such as SCF, IGF-1 and bFGF.
  • cytokines in particular LIF, IL-11, IL-6, IL-6R, CNTF, Oncostatin and other factors such as SCF, IGF-1 and bFGF.
  • the inactivated feeder layer used in step a) is preferably composed of fibroblasts, including mouse fibroblasts established as a line.
  • fibroblasts include the STO cells which may or may not be modified or transfected with expression vectors (Pain et al., 1996).
  • the cells used in step a) are cells obtained by suspending cells obtained from blastodermal disks of fertilized eggs in a culture medium comprising at least one cytokine, b-FGF, and SCF. Said cells are inoculated into a layer of feeder cells, incubated, and then collected.
  • Step b) consists in a progressive withdrawal of each growth factor added to the medium in step a), in particular a cytokine, b-FGF, and SCF, comprising a passage in a new medium free of at least one of said factors and in repeating various successive passages until the medium is free of all of said factors.
  • the expression progressive withdrawal is understood to mean a removal factor by factor from the culture medium.
  • the withdrawal of step b) may consist in progressively reducing the concentration of one or more factors or in culturing the avian stem cells directly in a medium free of one or more factors or alternatively free of all of said factors.
  • Step b) may also comprise the withdrawal of the serum.
  • the withdrawal may be progressive, by reducing the serum concentration during each passage, for example on passing from 10% to 7.5% and then 3.75% and 2%, tending toward 0% (serum-free medium).
  • a drastic withdrawal may be carried out.
  • Step b) may also comprise the withdrawal of the feeder layer.
  • the withdrawal of the feeder layer may also be gradual, by reducing the number of inactivated feeder cells during each passage. Alternatively, it is possible to carry out a drastic withdrawal.
  • the order of withdrawals can vary. For example, it is possible to start with the withdrawal of the growth factors and continue with the withdrawal of the feeder layer.
  • the invention relates to the established cell lines and to the cells derived from said lines which can be obtained from the method described above, said cells being capable of proliferating for at least 50 days, 100 days, 150 days, 300 days, or preferably at least 600 days in a medium free of exogenous growth factor, serum and/or feeder layer.
  • These cell lines and the cells derived therefrom are capable of proliferating for at least 50 days, 100 days, 150 days, 300 days, or preferably at least 600 days in a basal medium, in particular in a medium such as DMEM, GMEM, HamF12 or McCoy supplemented with various additives commonly used by persons skilled in the art.
  • a basal medium in particular in a medium such as DMEM, GMEM, HamF12 or McCoy supplemented with various additives commonly used by persons skilled in the art.
  • additives there may be mentioned nonessential amino acids, vitamins and sodium pyruvate.
  • the invention also relates to the cell lines and the cells derived from such lines described above, characterized in that they are avian stem cells, in particular avian somatic stem cells or avian embryonic stem cells.
  • stem cells may be adherent, while proliferating in the absence of the inactivated feeder layer.
  • these stem cells are nonadherent and proliferate in suspension in a basal medium mentioned above.
  • these cells are genetically modified so as to produce a substance of interest, in particular a polypeptide of interest, an antibody or an attenuated virus.
  • Cells of the invention can for example support the replication of live or attenuated viruses, in particular the viruses selected from the group of adenoviruses, hepadnaviruses, herpesviruses, orthomyxoviruses, papovaviruses, paramyxoviruses, picornaviruses, poxviruses, reoviruses and retroviruses.
  • viruses selected from the group of adenoviruses, hepadnaviruses, herpesviruses, orthomyxoviruses, papovaviruses, paramyxoviruses, picornaviruses, poxviruses, reoviruses and retroviruses.
  • the viruses belong to the family of orthomyxoviruses, in particular the influenza virus, to the family of paramyxoviruses, in particular the measles, mumps and rubella viruses.
  • the viruses replicated on these cells belong to the to the family of poxvirus, in particular canarypox virus, fowlpox virus as well as vaccinia virus.
  • the invention relates to the cell lines described above, the cells derived from said lines and also the cell lines obtained from cells which have been genetically modified.
  • the invention relates to the cell lines derived from step c) of the method described above, characterized in that they are avian stem cells capable of growing indefinitely in a basal medium free of exogenous growth factors, depleted of serum or free of serum and/or of feeder layer.
  • the cells obtained at the end of step c) may be genetically modified.
  • the invention also relates to a cell culture comprising cells derived from the cell lines described above, in particular avian stem cells or avian embryonic stem cells, and a basal medium free of exogenous growth factors depleted of serum or free of serum and/or of inactivated feeder layer.
  • the invention relates to the use of the cell lines and cells described above for the production of substances of interest, in particular of proteins of therapeutic interest, for the replication of live or attenuated viruses, in particular viruses chosen from the group of adenoviruses, hepadnaviruses, herpesviruses, orthomyxoviruses, papovaviruses, paramyxoviruses, picornaviruses, poxviruses, reoviruses and retroviruses.
  • viruses chosen from the group of adenoviruses, hepadnaviruses, herpesviruses, orthomyxoviruses, papovaviruses, paramyxoviruses, picornaviruses, poxviruses, reoviruses and retroviruses.
  • the cell lines and the cells described above are used for the production of viruses belonging to the family of orthomyxoviruses, in particular the influenza virus, and for the production of viruses belonging to the family of paramyxoviruses, in particular the measles, mumps and rubella viruses.
  • one best mode of the invention is to use the cells as defined above to produce live or attenuated vaccine, for example recombinant vaccine, comprises culturing the adherent or non adherent cell lines established in step c) according to the process described above, inoculating said cells with viral particles and culturing said cells in a basal medium as mentioned above until cell lysis occurs and newly produced viral particles are released in said medium.
  • the invention has shown to be particularly useful for the production of attenuated virus belonging to the family of poxvirus, in particular canarypoxvirus, fowlpoxvirus and vaccinia virus such as such as native or recombinant vaccinia virus (for example Modified Vaccinia virus Ankara, MVA (such as MVA available under ATCC Number VR-1508) or other orthopoxviruses) and is further described in the examples below.
  • canarypoxvirus fowlpoxvirus
  • vaccinia virus such as such as native or recombinant vaccinia virus (for example Modified Vaccinia virus Ankara, MVA (such as MVA available under ATCC Number VR-1508) or other orthopoxviruses) and is further described in the examples below.
  • the invention is aimed at the use of the cells according to the invention for producing recombinant viruses expressing antigens as vaccine against infection diseases such as smallpox and cancer (for example melanoma, prostate cancer, breast cancer, lung cancer, ovary cancer, liver cancer . . . ).
  • smallpox and cancer for example melanoma, prostate cancer, breast cancer, lung cancer, ovary cancer, liver cancer . . . ).
  • FIGS. 1 - 3 Growth curves for the cell lines of the invention (with withdrawal of serum (FIG. 2) and with withdrawal of feeder layer (FIG. 3).
  • FIG. 4 Photograph showing the characteristic morphology of avian stem cells.
  • N nucleus
  • n nucleolus
  • C cytoplasm
  • FIG. 5 Photograph showing the alkaline phosphatase activity of avian stem cell lines which are adherent or which are in suspension.
  • the cells After fixing (0.1% formaldehyde/0.5% glutaraldehyde, 30 minutes at 4° C.), the cells are rinsed twice in 1 ⁇ PBS and incubated for between 10 and 30 minutes at 37° C. in an NBT/BCIP (Nitro Blue Tetrazolium chloride 0.375 mg/ml, 5-bromo-4-chloro-3-indolyl phosphate 0.188 mg/ml, 0.1M Tris pH 9.5, 0.05M MgCl 2 , 0.1M Nac1) solution. The reaction is stopped by two 1 ⁇ PBS washes and the photographs are taken.
  • NBT/BCIP Niro Blue Tetrazolium chloride 0.375 mg/ml, 5-bromo-4-chloro-3-indolyl phosphate 0.188 mg/ml, 0.1M Tris pH 9.5, 0.05M MgCl 2 , 0.1M Nac1
  • A illustrates the characteristic violet coloration of the endogenous alkaline phosphatase activity obtained with the adherent line S86N45 p87, a line cultured with no feeder or factor ( ⁇ 40 magnification, Sony Cyber-shot digital camera).
  • [0103] B illustrates the violet coloration characteristic of the endogenous alkaline phosphatase activity obtained with the EB14 line maintained from 8 passages in suspension, line derived from the S86N45 cells, cultured in suspension with no feeder or factor ( ⁇ 20 magnification, Sony Cyber-shot digital camera).
  • the S86N strain a commercial strain intended for the production of chicken bearing a quality label
  • CNRs the strain intended for the production of chicken bearing a quality label
  • Marens a local strain which is genetically and phenotypically well characterized
  • White Leghorns a strain more intended for the production of eggs for consumption and a reference strain for research laboratories, and the like.
  • various origins have been tested including certain eggs (called Valo) obtained from the White Leghorn strain from Lohmann (Germany) considered to be “SPF” (Specific Pathogen Free) eggs kept under very particular health safety conditions. Numerous cell isolates were obtained from various strains, suggesting the general character of the method.
  • the eggs are opened, the yolk is separated from the egg white during the opening.
  • the embryos are removed from the yolk either directly or with the aid of a Pasteur pipette, or with the aid of a small absorbent filter paper (Whatmann 3M paper), cut out beforehand in the form of a perforated ring with the aid of a punch.
  • the diameter of the perforation is about 5 mm.
  • These small rings are sterilized using dry heat for about 30 minutes in an oven.
  • This small paper ring is deposited on the surface of the yolk and centered on the embryo which is thus surrounded by the paper ring.
  • the latter is then cut out with the aid of small pairs of scissors and the whole removed is placed in a Petri dish, filled with PBS or with a physiological saline.
  • the embryo thus carried away by the ring is cleaned of the excess yolk in the medium and the embryonic disk, thus freed of the excess vitellin, is collected with a Pasteur pipette.
  • the embryos are placed in a tube containing physiological medium (1 ⁇ PBS, Tris Glucose, medium, and the like). The embryos are then mechanically dissociated and inoculated on a “feeder” into defined culture medium.
  • the culture medium composed of MacCoy medium as basal medium supplemented with fetal calf serum at an initial concentration of 12 to 8%, with nonessential amino acids at 1%, with a mixture of vitamins of commercial origin at 1%, 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, with an initial mixture of antibiotics containing 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.
  • 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.
  • a mixture of nucleosides may also be added, this mixture being prepared as described above (Pain et al., 1996).
  • the basal media tested under these same conditions and which give similar results are the HamF12, Glasgow MEM and DMEM media, the latter supplemented with biotin at a final concentration of 8 mg/l.
  • the biotin concentration is 0.2 mg/l in the MacCoy medium, 0.0073 mg/l in the HamF12 and 0 in the commercial DMEM and GMEM media.
  • the growth factors and the cytokines added to the culture medium are preferably factors and cytokines which are recombinant, including mouse SCF at a final concentration of 1 ng/ml, IGF-1 at a final concentration of 1 to 5 ng/ml, CNTF at a final concentration of 1 ng/ml, IL-6 at a final concentration of 1 ng/ml, and the soluble IL-6 receptor at a final concentration of 0.5 ng/ml to 1 ng/ml.
  • some other factors may be added during the first passages. For example up to passage 3 or 10, it is possible to add bFGF to the medium at a final concentration of 1 ng/ml and IL-11 at a final concentration of 1 ng/ml.
  • the inoculation is carried out into this medium on the inactivated “feeder” composed of mouse fibroblasts established as lines, the STO cells.
  • these cells were transfected with simple expression vectors allowing the expression of growth factors such as avian SCF, constitutively in the STO cells.
  • this “feeder” produces the factor in a form which is soluble and/or attached in the plasma membrane of the cells.
  • the medium After initial inoculation of the cells directly into this medium, the medium is partially changed the next day, and then partially or completely during subsequent days, depending on the rate of adhesion observed for the primary cells. After about 4 to 7 days depending on the cases, the initial culture is dissociated and transferred into new dishes in the same initial medium on the inactivated feeder. After three to five passages, the cells are cultured on an inactivated feeder of STO cells which are nontransfected or transfected with an expression vector encoding a resistance to an antibiotic such as the gene for resistance to neomycin, to hygromycin, to puromycin and the like. After about twenty passages, the cells are progressively deprived of growth factors and cytokines.
  • the expression gradual withdrawal is understood to mean a removal factor by factor from the culture medium.
  • SCF is first of all removed, and then, two or three passages later, IGF-1. If the cells do not exhibit morphological alterations or a variation in their average rate of proliferation, the other factors, such as CNTF and IL-6, are then removed. This withdrawal may also be drastic. All the factors are in this case removed all at once. The cells are then observed and are only passaged several days later if their rate of proliferation is modified. The latter solution is generally that which is practiced.
  • very long periods of time is understood to mean periods of the order of several weeks with a minimum of 50 days, preferably periods greater than 200 to 400 days, without limitation in time. Periods greater than 600 days are observed.
  • all the cells which are adherent are dissociated with a proteolytic dissociation enzyme, such as pronase, collagenase, dispase, trypsin, and the like.
  • a proteolytic enzyme of bacterial origin is used in order to avoid any potential contaminant of animal origin.
  • These cells have the characteristics of embryonic stem cells with a specific morphology illustrated, by way of example, by the photograph of FIG. 4 i.e. a small size, a large nucleocytoplasmic ratio, a nucleus with at least one nucleolus which is clearly visible and a very small cytoplasm. These cells are characterized by growth in the form of more or less compact solid masses.
  • telomere activity component is also present and is an important factor in the “stem” nature of these cells.
  • Cells of different isolates are obtained and maintained for long periods of time. TABLE 1 Table 1 illustrates a few of the characteristics of these isolates Gener- Name Species Start “Stoppage” Days Passage ation S86N16 Chicken S86N 26-01-2000 May 8, 2001 559 207 692 WL3 Chicken WL 28-06-2000 Sep. 8, 2001 403 153 333 Valo4 Chicken Valo 26-09-2000 Jul. 2, 2002 401 135 317 S86N45 Chicken S86N 29-01-2001 Dec. 11, 2001 287 118 329
  • stoppage does not correspond to the end of the proliferation of the cells but to a deliberate stoppage of the cell cultures by the experimenter.
  • stem cells in particular somatic stem cells and embryonic stem cells, are their capacity to proliferate in vitro for considerable periods of time. In order to propagate and to passage the cells, the culture medium is changed and replaced with fresh medium a few hours before their passage.
  • the curve presented in FIG. 1 illustrates a profile of cell growth and establishment.
  • a mean division time can be calculated. For all the independent isolates obtained, the rate of proliferation increases slightly during successive passages, thus causing the average division time during the establishment of the cells to vary.
  • the cells are initially inoculated on an inactivated feeder layer and are passaged regularly at a constant initial inoculation density of 1 to 2 ⁇ 10 6 cells per 100 mm dish.
  • Table 2 illustrates the doubling time (d) and the mean division time (MDT in hour) for 3 established cell types as a function of the culture time. It is observed that the mean doubling time decreases during the establishment.
  • the mean division time (MDT) is then obtained in hours by dividing 24 hours by d.
  • Valo cells are passaged during this establishment on a plastic support without the presence of a feeder. The doubling time decreases and then increases again, when the cells become rehabituated to this new environment.
  • the culture media used are conventional culture media comprising a base (DMEM, GMEM, HamF12, McCoy, and the like) supplemented with various additives such as nonessential amino acids, vitamins, and sodium pyruvate.
  • This complex medium comprises fetal calf serum, which remains a central component of the culture, even though components of different origins, including plant components, can be gradually used.
  • a process for controlling and habituating the cells to relatively low proportions of fetal calf serum is presented. It is thus possible to maintain cells in high proliferation (division time>1) with low percentages of serum (2% for example in the case of the S86N16 cells).
  • the cytokines are mainly cytokines whose action is through a receptor which is associated with the gp130 protein.
  • LIF, interleukin 11, interleukin 6, CNTF, 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 heterodimeric form.
  • the combination of a soluble form of the receptors a form described inter alia for the receptors for interleukin 6 and CNTF, makes it possible to increase the proliferative effect observed. It has been previously shown that the addition of at least one of these cytokines appeared to be necessary for obtaining embryonic stem cells.
  • the trophic factors are mainly SCF, IGF-1 and bFGF, which are also used at the start of the culture, as described above. Their presence is also necessary for obtaining and amplifying the cells.
  • the cells are not obtained with the same frequencies. Comparison of the compositions of the media makes the identification of one of the components in particular difficult. It appears more likely that the whole combination allows an improvement in the physiology of the cells.
  • the preferred media the Ham F12 medium, the MacCoy medium, the DMEM medium and a DMEM medium enriched with biotin will be noted. Starting with such an isolate, adaptation trials are carried out in these different media.
  • telomere activity as presented in example 10 for the EB1, EB4 and EB5 cells.
  • the cells exhibit a high proliferation in different media.
  • the initial inoculation density and the very regular supply of fresh medium provides high densities which may range above 1 ⁇ 10 6 cells per ml.
  • Table 5 summarizes the main characteristics of a few isolates (parental cells, initial passage of the making into a suspension, number of days maintained in culture in suspension, number of passages and of generations obtained before voluntary stoppage of the maintenances). It can thus be noted that the passage for the making into a suspension can vary from one isolate to another (see isolate EB1 and EB14) and the proliferation rate (see isolate EB3 and EB14).
  • start corresponds to the cells being placed under nonadherence.
  • the stem cells maintained for long culture times are characterized with the same criteria as those described above (Pain et al., 1996). It is thus possible to regularly detect the endogenous alkaline phosphatase activity, illustrated by the photograph of FIG. 5, the endogenous telomerase activity and reactivity with specific antibodies such as the antibodies SSEA-1 (TEC-01) and EMA-1.
  • telomerase activity is detectable for the S86N16 cells, the S86N45 cells and for the EB1, EB4 and EB5 cells which are derived therefrom in a nonadherent form (see table 6).
  • the CEFs Chicken Embryonic Fibroblasts maintained in primary culture are considered as negative.
  • the threshold of an OD ⁇ 0.2 is the threshold recommended by the kit as the negative threshold. All the analyses were carried out on an equivalent of 2000 cells. TABLE 6 Assay of the telomerase activity in various lines at various passages Cells Passage Telomerase OD S86N16 p12 1.7 p29 2.8 p185 0.97 p204 0.95 S86N16 EB1 p134 1.1 S86N45 p50 0.87 p58 1.1 p66 0.96 p94 1.2 S86N45 EB4 p112 1.4 S86N45 EB5 p112 0.94 CEF* p4 0.07
  • the stem cells maintained in a growth over the long term are transfected with various expression plasmids. It has been shown that avian stem cells could be transfected (Pain et al., 1996). In particular, the nonadherent cells are transfected and various sorting systems make it possible to identify the stably transfected cells (cell sorting, limiting dilution, and the like). These genetic modifications can be made at the undifferentiated stage of the stem cell. Once this modification has been obtained, the cell is then induced to differentiate spontaneously or by addition of a differentiation inducer.
  • the cells can form embryoid bodies in suspension, which embryoid bodies can be caused to adhere to plastic after dissociation or nondissociation of the cells constituting them. These differentiated cells then proliferate but have a more limited capacity for proliferation over the long term. By targeting the genetic modification on a gene which influences the proliferation of the cells, it is possible to make these differentiated cells capable of proliferating over the long term.
  • the adherent and nonadherent cells can be infected with different viruses and retroviruses including avian viruses and retroviruses. These cells can thus serve as a replication support for the production of viral stocks intended for the production of live, attenuated or inactivated human and veterinary vaccines depending on the cases.
  • viruses of interest there may be mentioned those of the family of adenoviruses (such as Human Adenovirus C, Fowl Adenovirus A, Ovine Adenovirus D, Turkey Adenovirus B), circoviridae (such as Chicken Anemia Virus, CAV), certain coronaviruses, such as avian infectious bronchitis virus (IBV), flaviviruses (such as Yellow fever virus and hepatitis C virus), hepadnaviruses (such as Hepatitis B virus and Avihepadnaviruses such as Duck hepatitis B virus); herpesviruses (such as Gallid herpesvirus, HSV (Herpes simplex virus) and Human herpesvirus 1, 3 and 5), orthomyxoviruses (such as the influenza virus: Influenzavirus A, Influenzavirus B and Influenza-virus C), papovaviruses (such as polyomavirus and more particularly Simian virus 40), paramyxoviruses (such as
  • the EB1 or EB14 cells are inoculated into a medium, preferably MacCoy's 5A, HAMF12 or DMEM medium, or any other medium of interest, containing 5% serum at a concentration of 0.2 ⁇ 10 6 cells/ml for an initial volume of 50 ml in general. They are maintained in culture at 39° C. and at 7.5% CO 2 , with stirring. Fresh medium is added every day for the 3 to 4 days for which the amplification lasts in order to reach a cell concentration of 1 to 3 ⁇ 10 6 cells/ml for a final culture volume of 100 to 250 ml. The cells in suspension are collected and centrifuged for 10 min at 1000 rpm approximately.
  • the pellet is resuspended in 20 to 50 ml of 1 ⁇ PBS (Phosphate buffer Salt).
  • PBS Phosphate buffer Salt
  • the cells are then counted, centrifuged and the pelleted cells are taken up in a serum-free medium at a final concentration of 3 to 5 ⁇ 10 6 cells/ml.
  • Several tubes are then prepared under these conditions containing 3 to 5 ⁇ 10 6 cells per tube.
  • the viral stock having a known titer is rapidly thawed at 37° C. and diluted in serum-free medium at a titer of 10 ⁇ to 1000 ⁇ the concentration necessary for the final infection.
  • the cells are infected with the virus of interest at an m.o.i. (multiplicity of infection) of 0.01 to 0.5 according to the types of virus, which involves adding between 0.1 and 10% volume/volume of viral suspension to the cellular pellet. After incubating for 1 hour at an optimum temperature for the virus, in general from 33 to 37° C., the cells are again centrifuged and the medium removed with care. This step is found to be often necessary in order to limit the effect of the initial virus in the subsequent process.
  • One of the possibilities is to directly dilute the cells without centrifuging them again with serum-containing medium (5% of serum) at a final concentration of 0.2 to 1 ⁇ 10 6 cells/ml and incubated again.
  • the medium containing the cells or the cellular debris is harvested. Depending on the viruses, only the pellet or the supernatant may be of interest and contain the viral particles.
  • the cells are harvested and centrifuged. The collected supernatant is centrifuged again for 5 to 10 minutes at 2500 rpm, and stored at ⁇ 80° C. before purification of the particles. An aliquot is collected in order to carry out the titration.
  • the cellular pellet is taken up in 5 ml of serum-free medium, sonicated and centrifuged for 5 to 10 minutes at 2500 rpm. The supernatant obtained is stored at ⁇ 80° C. up to the purification and the titration of an aliquot.
  • the viral infection and production efficiencies are compared between the various conditions performed.
  • the titrations are in general carried out by the lysis plaque technique.
  • the cells are inoculated 48 hours before the infection into T150 flasks at a concentration of between 0.03 and 0.06 ⁇ 10 6 cells/cm 2 in a medium, preferably MacCoy's 5A, HAMF12 or DMEM medium, or any other medium of interest, containing 5% serum. They are maintained at 39° C. and 7.5% CO 2 .
  • the viral stock having a known titer is rapidly thawed at 37° C. and diluted in serum-free medium at a titer of 10 ⁇ to 1000 ⁇ the concentration necessary for the final infection.
  • the cells are infected with the virus of interest at an m.o.i. (multiplicity of infection) of 0.01 to 0.5 according to the types of virus, which involves adding between 0.1 and 10% volume/volume of viral suspension to the cellular pellet.
  • the infection is generally carried out in a minimum of medium (from 5 to 10 ml for a 75 cm 2 flask) in a medium containing 0% serum.
  • the cells After incubating for 1 hour at the optimum temperature for the virus, in general from 33 to 37° C., 20 ml of medium 5% are added to the flasks.
  • the cells can be washed with PBS in order to remove the particles which might be attached to the cells.
  • the cells are observed daily after the infection in order to monitor the appearance of the cell lysis plaque, which indicates good progress of the infection.
  • the medium containing the supernatant, the cells and the cellular debris are harvested. Depending on the viruses, only the pellet or the supernatant may be of interest and contain the viral particles.
  • the cells are harvested and centrifuged. The collected supernatant is centrifuged again for 5 to 10 minutes at 2500 rpm, and stored at ⁇ 80° C. before purification of the particles. An aliquot is collected in order to carry out the titration.
  • the cellular pellet is taken up in 5 ml of serum-free medium, sonicated and centrifuged for 5 to 10 minutes at 2500 rpm. The supernatant obtained is stored at ⁇ 80° C. up to the purification and the titration of an aliquot.
  • the viral infection and production efficiencies are compared between the various conditions performed.
  • the titrations are in general carried out by the lysis plaque technique.
  • Table 7 illustrates the results obtained. These results demonstrate the very satisfactory replication of the recombinant avipox on the EB1 stem cells.
  • the infectious titer progresses throughout the culture and the course of the infection, reaching a maximum of 7.2 PFU/cell (PFU: Plating Forming Unit) after 4 days of incubation. This titer is at least equivalent to that obtained for this same recombinant avipox on primary chicken embryo cells.
  • the MVA virus (titer 2,5 ⁇ 10 7 TCID50/ml in 0.5 ml vials) was received under frozen conditions. For safety reasons, the MVA virus and infected cells were kept under controlled conditions ( ⁇ 80° C. freezer) and the contaminated plastic material was placed into hypochloride solution for more than 1 hour and then place into a bag for full and complete autoclave inactivation.
  • the cells were infected 1 hour in 2 ml of PBS with the different m.o.i. of interest with no washing with PBS after the infection.
  • the medium was just added to the complete infectious medium, i.e. the added virus was not removed.
  • the non adherent EB14 cells were amplified. The cells were infected, not washed, and the complete medium directly added on the inocculum after 1 hour of contact with viral particles. After 3 days, a characteristic cell lysis was observed. The non infected cells used as the control were counted and a good growth was demonstrated, showing good culture conditions and therefore confirming an efficient lysis by the virus in the infected culture. Cells and supernatant are harvested and stored at ⁇ 80° C. before purification of particles and/or titration (see table 8). TABLE 8 Results of the titration Titer/mL Average Virus / based on Virus virus Cell pellet of 14 ml total yield / yield / Type M.O.I.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Gynecology & Obstetrics (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Reproductive Health (AREA)
  • Developmental Biology & Embryology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Feeding And Watering For Cattle Raising And Animal Husbandry (AREA)
  • Farming Of Fish And Shellfish (AREA)
  • Peptides Or Proteins (AREA)
  • Coloring Foods And Improving Nutritive Qualities (AREA)
US10/625,847 2002-03-08 2003-07-24 Avian cell lines useful for the production of substances of interest Abandoned US20040058441A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/350,781 US20090239297A1 (en) 2002-03-08 2009-01-08 Avian cell lines useful for the production of substrates of interest
US12/717,096 US9382513B2 (en) 2002-03-08 2010-03-03 Method of making an avian cell line
US13/206,056 US20110294209A1 (en) 2002-03-08 2011-08-09 Methods for producing nonadherent avian cell lines

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
FR0202945 2002-03-08
FR0202945A FR2836924B1 (fr) 2002-03-08 2002-03-08 Lignees de cellules aviaires utiles pour la production de substances d'interet
WOPCT/FR03/00735 2003-03-07
PCT/FR2003/000735 WO2003076601A1 (fr) 2002-03-08 2003-03-07 Lignées de cellules aviaires utiles pour la production de substances d'intérêt

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/FR2003/000735 Continuation WO2003076601A1 (fr) 2002-03-08 2003-03-07 Lignées de cellules aviaires utiles pour la production de substances d'intérêt
PCT/FR2003/000735 A-371-Of-International WO2003076601A1 (fr) 2002-03-08 2003-03-07 Lignées de cellules aviaires utiles pour la production de substances d'intérêt

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/350,781 Continuation US20090239297A1 (en) 2002-03-08 2009-01-08 Avian cell lines useful for the production of substrates of interest

Publications (1)

Publication Number Publication Date
US20040058441A1 true US20040058441A1 (en) 2004-03-25

Family

ID=27763651

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/625,847 Abandoned US20040058441A1 (en) 2002-03-08 2003-07-24 Avian cell lines useful for the production of substances of interest
US12/350,781 Abandoned US20090239297A1 (en) 2002-03-08 2009-01-08 Avian cell lines useful for the production of substrates of interest
US12/717,096 Expired - Lifetime US9382513B2 (en) 2002-03-08 2010-03-03 Method of making an avian cell line
US13/206,056 Pending US20110294209A1 (en) 2002-03-08 2011-08-09 Methods for producing nonadherent avian cell lines

Family Applications After (3)

Application Number Title Priority Date Filing Date
US12/350,781 Abandoned US20090239297A1 (en) 2002-03-08 2009-01-08 Avian cell lines useful for the production of substrates of interest
US12/717,096 Expired - Lifetime US9382513B2 (en) 2002-03-08 2010-03-03 Method of making an avian cell line
US13/206,056 Pending US20110294209A1 (en) 2002-03-08 2011-08-09 Methods for producing nonadherent avian cell lines

Country Status (15)

Country Link
US (4) US20040058441A1 (zh)
EP (2) EP1483369B1 (zh)
JP (2) JP5025888B2 (zh)
CN (2) CN100562567C (zh)
AT (1) ATE408004T1 (zh)
AU (1) AU2003227820B2 (zh)
CA (1) CA2478125C (zh)
DE (1) DE60323468D1 (zh)
DK (1) DK1483369T3 (zh)
ES (1) ES2312775T3 (zh)
FR (1) FR2836924B1 (zh)
HK (1) HK1142094A1 (zh)
PT (1) PT1483369E (zh)
SI (1) SI1483369T1 (zh)
WO (1) WO2003076601A1 (zh)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060233834A1 (en) * 2003-07-22 2006-10-19 Vivalis Production of poxviruses with adherent or non adherent avian cell lines
US20080274125A1 (en) * 2004-12-08 2008-11-06 Vivalis Human Stem Cell Lines Derived From Es Cells and Uses for Production of Vaccines and Recombinant Proteins
US20080317778A1 (en) * 2000-11-23 2008-12-25 Paul Chaplin Modified vaccinia ankara virus variant and cultivation method
US20090029459A1 (en) * 2002-09-05 2009-01-29 Bavarian Nordic A/S Method for the cultivation of primary cells and for the amplification of viruses under serum free conditions
US20090028831A1 (en) * 2007-07-23 2009-01-29 University Of Kentucky Research Foundation Stem cell regulator, compositions and methods of use
US20090081251A1 (en) * 2005-04-11 2009-03-26 Vivalis Production of Viral Vaccines in Suspension on Avian Embryonic Derived Stem Cell Lines
US20100221825A1 (en) * 2002-03-08 2010-09-02 Vivalis Avian cell lines useful for the production of substances of interest
US20100226912A1 (en) * 2007-05-21 2010-09-09 Vivalis RECOMBINANT PROTEIN PRODUCTION IN AVIAN EBx® CELLS
US20100235937A1 (en) * 2006-08-09 2010-09-16 Isabelle Valarche Production of transgenic avian organisms employing embryonic stem cells
WO2011051445A1 (en) 2009-10-30 2011-05-05 Glaxosmithkline Biologicals S.A. Process for preparing an influenza seed virus for vaccine manufacture
WO2011051235A1 (en) 2009-10-27 2011-05-05 Glaxosmithkline Biologicals, Niederlassung Der Smithkline Beecham Pharma Gmbh & Co. Kg Process for producing influenza vaccine
US20110165224A1 (en) * 2009-12-15 2011-07-07 University Of Saskatchewan Vaccines for inclusion body hepatitis
US20110182931A1 (en) * 2008-04-23 2011-07-28 Paul Michael Coussens Immortal avian cell line and methods of use
WO2012032521A2 (en) 2010-09-07 2012-03-15 Technion Research & Development Foundation Ltd. Novel methods and culture media for culturing pluripotent stem cells
US20130244246A1 (en) * 2007-07-03 2013-09-19 Transgene S.A.` Immortalized avian cell lines comprising e1a nucleic acid sequences
AU2011253998B2 (en) * 2005-04-11 2014-04-24 Valneva Process of manufacturing viral vaccines in suspension avian embryonic derived stem cell lines
US9260694B2 (en) 2007-04-24 2016-02-16 Valneva Generation of duck cell lines
US9441032B2 (en) 2010-04-07 2016-09-13 Agency For Science, Technology And Research Binding molecules against Chikungunya virus and uses thereof
WO2020234888A1 (en) 2019-05-22 2020-11-26 Hadasit Medical Research Services And Development Ltd. Methods of culturing human pluripotent cells
CN114480286A (zh) * 2022-02-16 2022-05-13 上海健士拜生物科技有限公司 无血清悬浮型lmh细胞系及其制备方法和应用
CN114554866A (zh) * 2019-09-10 2022-05-27 必要肉制品公司 用于生产食物产品的禽类干细胞

Families Citing this family (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060191026A1 (en) 2005-02-18 2006-08-24 Origen Therapeutics, Inc. Tissue specific expression of antibodies in chickens
US7145057B2 (en) 2002-02-01 2006-12-05 Origen Therapeutics, Inc. Chimeric bird from embryonic stem cells
AU2003296974A1 (en) * 2002-12-13 2004-07-14 Aventis Pasteur, Inc. Production of alvac on avian embryonic stem cells
EP1500699A1 (en) * 2003-07-22 2005-01-26 Vivalis Production of vaccinia virus with adherent or non adherent avian cell lines
US20070275010A1 (en) * 2003-09-18 2007-11-29 Mark Feinberg Mva Vaccines
EP2811027A1 (en) 2004-05-21 2014-12-10 Novartis Vaccines and Diagnostics, Inc. Alphavirus vectors for RSV and PIV vaccines
DE602005024827D1 (de) 2004-09-09 2010-12-30 Novartis Vaccines & Diagnostic Verminderung von potentiellen iatrogenen risiken in verbindung mit influenza impfstoffen
WO2006062637A2 (en) 2004-11-03 2006-06-15 Novartis Vaccines And Diagnostics Inc. Influenza vaccination
NZ567817A (en) 2005-11-01 2012-01-12 Novartis Vaccines & Diagnostic Cell-derived viral vaccines with low levels of residual cell DNA by beta-propiolactone treatment
EA014028B1 (ru) 2005-11-04 2010-08-30 Новартис Вэксинс Энд Диагностикс Срл Эмульсии, содержащие свободное поверхностно-активное вещество в водной фазе в качестве адъюванта сплит вакцин против гриппа
US20110180430A1 (en) 2005-11-04 2011-07-28 Novartis Vaccines And Diagnostics Srl Adjuvanted influenza vaccines including cytokine-inducing agents
WO2007052155A2 (en) 2005-11-04 2007-05-10 Novartis Vaccines And Diagnostics Srl Influenza vaccine with reduced amount of oil-in-water emulsion as adjuvant
NZ568211A (en) 2005-11-04 2011-11-25 Novartis Vaccines & Diagnostic Influenza vaccines including combinations of particulate adjuvants and immunopotentiators
CA2628152C (en) 2005-11-04 2016-02-02 Novartis Vaccines And Diagnostics S.R.L. Adjuvanted vaccines with non-virion antigens prepared from influenza viruses grown in cell culture
DK1976559T6 (da) 2006-01-27 2020-04-06 Seqirus Uk Ltd Influenzavacciner indeholdende hæmagglutinin og matrixproteiner
FR2898909A1 (fr) 2006-03-24 2007-09-28 Agronomique Inst Nat Rech Combinaison de marqueurs de cellules aviaires
EP2004226A1 (en) 2006-03-24 2008-12-24 Novartis Vaccines and Diagnostics GmbH & Co. KG Storage of influenza vaccines without refrigeration
GB0614460D0 (en) 2006-07-20 2006-08-30 Novartis Ag Vaccines
CA2663196A1 (en) 2006-09-11 2008-03-20 Novartis Ag Making influenza virus vaccines without using eggs
NZ577405A (en) 2006-12-06 2012-08-31 Novartis Ag Vaccines including antigen from four strains of influenza virus
CN100443588C (zh) * 2006-12-11 2008-12-17 中国农业科学院北京畜牧兽医研究所 静宁鸡胚成纤维细胞系及其培养方法
EA201070066A1 (ru) 2007-06-27 2010-06-30 Новартис Аг Вакцины против гриппа с низким содержанием добавок
GB0810305D0 (en) 2008-06-05 2008-07-09 Novartis Ag Influenza vaccination
CA2710480A1 (en) 2007-12-24 2009-07-02 Novartis Ag Assays for adsorbed influenza vaccines
US8506966B2 (en) 2008-02-22 2013-08-13 Novartis Ag Adjuvanted influenza vaccines for pediatric use
KR20140096162A (ko) * 2008-02-25 2014-08-04 백스터 인터내셔널 인코포레이티드 연속 세포주 제조 방법
EP2268309B1 (en) 2008-03-18 2015-01-21 Novartis AG Improvements in preparation of influenza virus vaccine antigens
EP2379598B1 (en) 2009-01-19 2015-03-11 Innate Pharma Anti-kir3d antibodies
JP2012517416A (ja) 2009-02-10 2012-08-02 ノバルティス アーゲー 増加した量のh3抗原を含むインフルエンザワクチン
DK2396032T3 (en) 2009-02-10 2016-12-19 Seqirus Uk Ltd Influenza vaccines with reduced amounts of squalene
US20120093859A1 (en) 2009-02-10 2012-04-19 Novartis Ag Influenza vaccine regimens for pandemic-associated strains
DE102010018462A1 (de) 2009-04-27 2011-04-07 Novartis Ag Impfstoffe zum Schutz gegen Influenza
DK3178490T3 (da) 2009-07-15 2022-06-20 Glaxosmithkline Biologicals Sa RSV F-proteinsammensætninger og fremgangsmåder til fremstilling af disse
CN102695523A (zh) 2009-09-10 2012-09-26 诺华有限公司 针对呼吸道疾病的组合疫苗
EP2531592A1 (en) 2010-02-04 2012-12-12 Vivalis 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
EP2545172B2 (en) 2010-03-08 2017-12-06 Novartis AG Methods of testing for intracellular pathogens
EP2374816B1 (en) 2010-04-07 2016-09-28 Agency For Science, Technology And Research Binding molecules against Chikungunya virus and uses thereof
WO2011127316A1 (en) 2010-04-07 2011-10-13 Novartis Ag Method for generating a parvovirus b19 virus-like particle
BR112012028146A2 (pt) 2010-05-06 2015-09-15 Novartis Ag compostos de peróxido orgânico para inativação de microorganismos
EP2571520B1 (en) 2010-05-21 2018-04-04 Seqirus UK Limited Influenza virus reassortment method
PL2575872T3 (pl) 2010-06-01 2021-02-22 Seqirus UK Limited Zatężanie antygenów szczepionkowych grypy bez liofilizacji
JP5976639B2 (ja) 2010-06-01 2016-08-23 ノバルティス アーゲー インフルエンザワクチン抗原の濃縮および凍結乾燥
US20130171185A1 (en) 2010-07-06 2013-07-04 Ethan Settembre Norovirus derived immunogenic compositions and methods
GB201011502D0 (en) 2010-07-08 2010-08-25 Glaxosmithkline Biolog Sa Novel process
AU2011290471B2 (en) 2010-08-20 2015-08-20 Novartis Ag Soluble needle arrays for delivery of influenza vaccines
FR2967072B1 (fr) 2010-11-05 2013-03-29 Univ Dundee Procede pour ameliorer la production de virus et semences vaccinales influenza
WO2012095514A1 (en) 2011-01-14 2012-07-19 Vivalis Recombinant protein production system
EP3527224A1 (en) 2011-01-26 2019-08-21 GlaxoSmithKline Biologicals S.A. Rsv immunization regimen
PT2707385T (pt) 2011-05-13 2017-12-19 Glaxosmithkline Biologicals Sa Antigénios de f de rsv pré-fusão
CA2841047A1 (en) 2011-07-06 2013-01-10 Novartis Ag Immunogenic compositions and uses thereof
EP3854413A1 (en) 2011-07-06 2021-07-28 GlaxoSmithKline Biologicals SA Immunogenic combination compositions and uses thereof
TWI575070B (zh) 2011-07-12 2017-03-21 傳斯堅公司 Hbv聚合酶突變體
TW201318637A (zh) 2011-09-29 2013-05-16 Transgene Sa 免疫療法組成物及用於治療c型肝炎病毒感染之療程(一)
WO2013045668A2 (en) 2011-09-29 2013-04-04 Transgene Sa Immunotherapy composition and regimen for treating hepatitis c virus infection
WO2013054199A2 (en) 2011-10-12 2013-04-18 Novartis Ag Cmv antigens and uses thereof
GB201216121D0 (en) 2012-09-10 2012-10-24 Novartis Ag Sample quantification by disc centrifugation
WO2013057715A1 (en) 2011-10-20 2013-04-25 Novartis Ag Adjuvanted influenza b virus vaccines for pediatric priming
EP2660316A1 (en) 2012-05-02 2013-11-06 Helmholtz-Zentrum für Infektionsforschung GmbH Avian cell line and its use in production of protein
EP2869842A1 (en) 2012-07-06 2015-05-13 Novartis AG Immunogenic compositions and uses thereof
JP6333814B2 (ja) 2012-07-10 2018-05-30 トランジェーヌ、ソシエテ、アノニムTransgene S.A. マイコバクテリア抗原ワクチン
WO2014009433A1 (en) 2012-07-10 2014-01-16 Transgene Sa Mycobacterium resuscitation promoting factor for use as adjuvant
GB201218195D0 (en) 2012-10-10 2012-11-21 Istituto Zooprofilattico Sperimentale Delle Venezie Composition
JP2016501196A (ja) 2012-11-20 2016-01-18 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 Rsvf融合前三量体
US20140255447A1 (en) * 2013-03-05 2014-09-11 Biomune Company Production of avian embryo cells
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
JP6605480B2 (ja) 2014-01-09 2019-11-13 トランスジェン・ソシエテ・アノニム ヘテロオリゴマーマイコバクテリア抗原の融合物
EP2974739A1 (en) 2014-07-15 2016-01-20 Novartis AG RSVF trimerization domains
EP3031822A1 (en) 2014-12-08 2016-06-15 Novartis AG Cytomegalovirus antigens
HUE048551T2 (hu) 2014-12-17 2020-08-28 Fundacion Para La Investig Medica Aplicada Nukleinsav-szerkezetek és génterápiás vektorok Wilson-kór és egyéb állapotok kezelésében történõ alkalmazásra
IL252917B2 (en) 2014-12-17 2023-10-01 Fundacion Para La Investig Medica Aplicada Vectors and vectors for gene therapy for use in the treatment of Wilson's disease
EP3047856A1 (en) 2015-01-23 2016-07-27 Novartis AG Cmv antigens and uses thereof
US11013795B2 (en) 2015-06-26 2021-05-25 Seqirus UK Limited Antigenically matched influenza vaccines
HUE051783T2 (hu) 2015-07-07 2021-03-29 Seqirus Uk Ltd Eljárás immunogén hemagglutinin mennyiségi meghatározására
MX2019011599A (es) 2017-03-30 2019-12-19 Univ Queensland Moleculas quimericas y usos de las mismas.
US20210180084A1 (en) 2018-05-14 2021-06-17 Vivet Therapeutics Gene therapy vectors comprising s/mar sequences
SG11202011280SA (en) 2018-05-30 2020-12-30 Inst Res Biomedicine Engineering b lymphocytes by utilizing endogenous activation-induced cytidine deaminase
EP3820884A1 (en) 2018-07-13 2021-05-19 Valneva SE Method for rescuing and producing a virus in avian cells
EP3863682B1 (en) 2018-10-12 2023-05-17 Vivet Therapeutics Codon-optimized transgene for the treatment of progressive familiar intrahepatic cholestasis type 3 (pfic3)
CA3117274A1 (en) 2018-11-07 2020-05-14 Vivet Therapeutics Codon-optimized abcb11 transgene for the treatment of progressive familial intrahepatic cholestasis type 2 (pfic2)
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
EP3884039A1 (en) 2018-11-23 2021-09-29 Valneva SE Food products comprising avian stem cells
AU2020299718A1 (en) 2019-07-02 2022-02-24 Fundacion Para La Investigacion Medica Aplicada cPLA2e inducing agents and uses thereof
JP2022542032A (ja) 2019-07-21 2022-09-29 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 治療用ウイルスワクチン
EP4004018A1 (en) 2019-07-24 2022-06-01 GlaxoSmithKline Biologicals SA Modified human cytomegalovirus proteins
IL293031A (en) 2019-11-18 2022-07-01 Seqirus Pty Ltd A method for producing reassortant influenza viruses
JP2023536080A (ja) 2020-08-06 2023-08-23 ファンダシオン パラ ラ インベスティガシオン メディカ アプリカダ 遺伝子治療によるアルツハイマー病などのタウオパチーの治療に使用するためのウイルス粒子
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
WO2022074105A1 (en) 2020-10-09 2022-04-14 UCB Biopharma SRL Nucleic acid constructs, viral vectors and viral particles
EP4267197A1 (en) 2020-12-23 2023-11-01 Vivet Therapeutics Minimal bile acid inducible promoters for gene therapy
EP4032547A1 (en) 2021-01-20 2022-07-27 GlaxoSmithKline Biologicals S.A. Hsv1 fce derived fragements for the treatment of hsv
IL312340A (en) 2021-10-28 2024-06-01 UCB Biopharma SRL Nucleic acid structures, viral vectors and viral particles
WO2023144665A1 (en) 2022-01-28 2023-08-03 Glaxosmithkline Biologicals Sa Modified human cytomegalovirus proteins
CN116656628A (zh) * 2023-07-31 2023-08-29 深圳市卫光生物制品股份有限公司 一种痘病毒载体疫苗的制备方法

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5162215A (en) * 1988-09-22 1992-11-10 Amgen Inc. Method of gene transfer into chickens and other avian species
US5340740A (en) * 1992-05-15 1994-08-23 North Carolina State University Method of producing an avian embryonic stem cell culture and the avian embryonic stem cell culture produced by the process
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
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

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4388298A (en) 1982-07-14 1983-06-14 The United States Of America As Represented By The Secretary Of Agriculture Propagation of hemorrhagic enteritis virus in a turkey cell line and vaccine produced
AU623922B2 (en) 1988-08-04 1992-05-28 Amrad Operations Pty. Limited In vitro propagation of embryonic stem cells
US5388680A (en) 1990-10-09 1995-02-14 Intellicall, Inc. Coin handling system with an improved coin chute
ES2137190T3 (es) 1991-07-12 1999-12-16 Pfizer Linea de celulas continua y vacuna contra los coccidios aviarios.
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
US5989805A (en) 1995-10-27 1999-11-23 Board Of Trustees Operating Michigan State University Immortal avian cell line to grow avian and animal viruses to produce vaccines
FR2749022B1 (fr) 1996-05-23 2001-06-01 Rhone Merieux Cellules aviaires immortelles
US5691200A (en) 1996-07-03 1997-11-25 The United States Of America As Represented By The Secretary Of Agriculture Method to produce granulocyte colony stimulating factor from immortalized avian T lymphocytes and method to produce immortalized cells
US5672485A (en) 1996-08-13 1997-09-30 Regents Of The University Of Minnesota Immortalized cell lines for virus growth
US5830723A (en) 1996-08-13 1998-11-03 Regents Of The University Of Minnesota Method for immortalizing chicken cells
US6103529A (en) 1996-10-10 2000-08-15 Life Technologies, Inc. Animal cell culture media comprising peptides derived from rice
PT841392E (pt) * 1996-11-12 2004-11-30 Univ Auburn Vacina viva atenuada de neospora
BR9811830A (pt) * 1997-08-04 2000-08-15 Univ Massachusetts Processo de cultivo que proporciona manutenção de células germinais primitivas de ave por perìodos prolongados, processo de produção de aves quiméricas, e, linhagem celular de pgc de ave
CA2300336A1 (en) * 1997-08-04 1999-02-11 University Of Massachusetts A Public Institution Of Higher Education Of The Commonwealth Of Massachusetts, As Represented By Its Amherst Campus Production of avian embryonic germ (eg) cell lines by prolonged culturing of pgcs, use thereof for cloning and chimerization
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
JP4972261B2 (ja) * 1999-08-27 2012-07-11 ライフ テクノロジーズ コーポレーション 金属結合化合物および細胞培養培地組成物中でのそれらの使用
DE19947407C2 (de) 1999-10-01 2002-08-01 Bayerische Motoren Werke Ag Datenbussystem für Kraftfahrzeuge
WO2002086073A2 (en) * 2001-04-20 2002-10-31 Memorial Sloan-Kettering Cancer Center Generation of differentiated tissue from nuclear transfer embryonic stem cells and methods of use
FR2836924B1 (fr) * 2002-03-08 2005-01-14 Vivalis Lignees de cellules aviaires utiles pour la production de substances d'interet
JP2005532057A (ja) * 2002-07-09 2005-10-27 バクスター・インターナショナル・インコーポレイテッド 細胞を培養するための動物性タンパク質非含有培地
EP1646715B1 (en) * 2003-07-22 2010-05-12 Vivalis Production of poxviruses with adherent or non adherent avian cell lines
EP1500699A1 (en) 2003-07-22 2005-01-26 Vivalis Production of vaccinia virus with adherent or non adherent avian cell lines
KR100552844B1 (ko) * 2003-12-31 2006-02-21 동부아남반도체 주식회사 반도체 소자의 제조 방법
FR2884255B1 (fr) 2005-04-11 2010-11-05 Vivalis Utilisation de lignees de cellules souches aviaires ebx pour la production de vaccin contre la grippe
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
WO2008017704A1 (en) * 2006-08-09 2008-02-14 Vivalis Method of production of transgenic avian using 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 (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5162215A (en) * 1988-09-22 1992-11-10 Amgen Inc. Method of gene transfer into chickens and other avian species
US5340740A (en) * 1992-05-15 1994-08-23 North Carolina State University Method of producing an avian embryonic stem cell culture and the avian embryonic stem cell culture produced by the process
US5656479A (en) * 1992-05-15 1997-08-12 North Carolina State University Avian embryonic stem cells
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
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
US6500668B2 (en) * 1994-10-21 2002-12-31 Jacques Samarut Culture medium for avian embryonic 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

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110217757A1 (en) * 2000-11-23 2011-09-08 Bavarian Nordic A/S Modified vaccinia ankara virus variant and cultivation method
US7964398B2 (en) 2000-11-23 2011-06-21 Bavarian Nordic A/S Modified vaccinia ankara virus variant and cultivation method
US20080317778A1 (en) * 2000-11-23 2008-12-25 Paul Chaplin Modified vaccinia ankara virus variant and cultivation method
US20090017536A1 (en) * 2000-11-23 2009-01-15 Paul Chaplin Modified vaccinia ankara virus variant and cultivation method
US7964395B2 (en) 2000-11-23 2011-06-21 Bavarian Nordic A/S Modified vaccinia ankara virus variant and cultivation method
US7964396B2 (en) 2000-11-23 2011-06-21 Bavarian Nordic A/S Modified vaccinia ankara virus variant and cultivation method
US8236560B2 (en) 2000-11-23 2012-08-07 Bavarian Nordic A/S Modified Vaccinia Ankara virus variant and cultivation method
US20100279386A1 (en) * 2000-11-23 2010-11-04 Bavarian Nordic A/S Modified vaccinia ankara virus variant and cultivation method
US8470598B2 (en) 2000-11-23 2013-06-25 Bavarian Nordic A/S Modified Vaccinia Ankara virus variant and cultivation method
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
US8329466B2 (en) 2002-09-05 2012-12-11 Bavarian Nordic A/S Method for the cultivation of primary cells and for the amplification of viruses under serum free conditions
US8673318B2 (en) 2002-09-05 2014-03-18 Bavarian Nordic A/S Method for the cultivation of primary cells and for the amplification of viruses under serum free conditions
US20110217749A1 (en) * 2002-09-05 2011-09-08 Bavarian Nordic A/S Method for the cultivation of primary cells and for the amplification of viruses under serum free conditions
US20090029459A1 (en) * 2002-09-05 2009-01-29 Bavarian Nordic A/S Method for the cultivation of primary cells and for the amplification of viruses under serum free conditions
US7964397B2 (en) 2002-09-05 2011-06-21 Bavarian Nordic A/S Method for the cultivation of primary cells and for the amplification of viruses under serum free conditions
US7771980B2 (en) 2003-07-22 2010-08-10 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
US20090239286A1 (en) * 2003-07-22 2009-09-24 Vivalis Production of poxviruses with adherent or non adherent avian cell lines
US20060233834A1 (en) * 2003-07-22 2006-10-19 Vivalis Production of poxviruses with adherent or non adherent avian cell lines
US7432101B2 (en) 2003-07-22 2008-10-07 Vivalis Production of poxviruses with adherent or non adherent avian cell lines
US20080274125A1 (en) * 2004-12-08 2008-11-06 Vivalis Human Stem Cell Lines Derived From Es Cells and Uses for Production of Vaccines and Recombinant Proteins
AU2011253998B2 (en) * 2005-04-11 2014-04-24 Valneva Process of manufacturing viral vaccines in suspension avian embryonic derived stem cell lines
US9701945B2 (en) 2005-04-11 2017-07-11 Valneva Production of viral vaccines in suspension on avian embryonic derived stem cell lines
US8148132B2 (en) 2005-04-11 2012-04-03 Vivalis Production of viral vaccines in suspension on avian embryonic derived stem cell lines
US9040296B2 (en) 2005-04-11 2015-05-26 Valneva Production of viral vaccines in suspension on avian embryonic derived stem cell lines
US20090081251A1 (en) * 2005-04-11 2009-03-26 Vivalis Production of Viral Vaccines in Suspension on Avian Embryonic Derived Stem Cell Lines
US20100235937A1 (en) * 2006-08-09 2010-09-16 Isabelle Valarche Production of transgenic avian organisms employing embryonic stem cells
US8962311B2 (en) 2006-08-09 2015-02-24 Valneva Method of obtaining chicken embryonic stem cells
US9822345B2 (en) 2007-04-24 2017-11-21 Valneva Method of making a virus using duck embryonic derived stem cell lines
US9260694B2 (en) 2007-04-24 2016-02-16 Valneva Generation of duck cell lines
US20100226912A1 (en) * 2007-05-21 2010-09-09 Vivalis RECOMBINANT PROTEIN PRODUCTION IN AVIAN EBx® CELLS
US8809056B2 (en) * 2007-07-03 2014-08-19 Transgene S.A. Immortalized avian cell lines comprising E1A nucleic acid sequences
US20130244246A1 (en) * 2007-07-03 2013-09-19 Transgene S.A.` Immortalized avian cell lines comprising e1a nucleic acid sequences
US20090028831A1 (en) * 2007-07-23 2009-01-29 University Of Kentucky Research Foundation Stem cell regulator, compositions and methods of use
US8716016B2 (en) 2008-04-23 2014-05-06 Board Of Trustees Of Michigan State University Immortal avian cell line and methods of use
US20110182931A1 (en) * 2008-04-23 2011-07-28 Paul Michael Coussens Immortal avian cell line and methods of use
WO2011051235A1 (en) 2009-10-27 2011-05-05 Glaxosmithkline Biologicals, Niederlassung Der Smithkline Beecham Pharma Gmbh & Co. Kg Process for producing influenza vaccine
WO2011051445A1 (en) 2009-10-30 2011-05-05 Glaxosmithkline Biologicals S.A. Process for preparing an influenza seed virus for vaccine manufacture
US9333253B2 (en) 2009-12-15 2016-05-10 University Of Saskatchewan Vaccines for inclusion body hepatitis
US8524249B2 (en) 2009-12-15 2013-09-03 University Of Saskatchewan Vaccines for inclusion body hepatitis
US20110165224A1 (en) * 2009-12-15 2011-07-07 University Of Saskatchewan Vaccines for inclusion body hepatitis
US9441032B2 (en) 2010-04-07 2016-09-13 Agency For Science, Technology And Research Binding molecules against Chikungunya virus and uses thereof
WO2012032521A2 (en) 2010-09-07 2012-03-15 Technion Research & Development Foundation Ltd. Novel methods and culture media for culturing pluripotent stem cells
WO2020234888A1 (en) 2019-05-22 2020-11-26 Hadasit Medical Research Services And Development Ltd. Methods of culturing human pluripotent cells
CN114554866A (zh) * 2019-09-10 2022-05-27 必要肉制品公司 用于生产食物产品的禽类干细胞
CN114480286A (zh) * 2022-02-16 2022-05-13 上海健士拜生物科技有限公司 无血清悬浮型lmh细胞系及其制备方法和应用

Also Published As

Publication number Publication date
JP5025888B2 (ja) 2012-09-12
EP1992687A2 (fr) 2008-11-19
PT1483369E (pt) 2008-12-17
WO2003076601A1 (fr) 2003-09-18
DK1483369T3 (da) 2008-12-01
EP1992687A3 (fr) 2009-03-25
US20120070893A9 (en) 2012-03-22
ES2312775T3 (es) 2009-03-01
CN100562567C (zh) 2009-11-25
CN101676388A (zh) 2010-03-24
CN1649999A (zh) 2005-08-03
US20110294209A1 (en) 2011-12-01
HK1142094A1 (en) 2010-11-26
US20100221825A1 (en) 2010-09-02
CA2478125C (fr) 2013-05-07
US20090239297A1 (en) 2009-09-24
AU2003227820A1 (en) 2003-09-22
FR2836924B1 (fr) 2005-01-14
SI1483369T1 (sl) 2008-12-31
CN101676388B (zh) 2013-09-04
WO2003076601A8 (fr) 2005-05-12
FR2836924A1 (fr) 2003-09-12
EP1992687B1 (fr) 2019-06-26
ATE408004T1 (de) 2008-09-15
JP5784521B2 (ja) 2015-09-24
AU2003227820B2 (en) 2008-04-03
EP1483369B1 (fr) 2008-09-10
JP2005525803A (ja) 2005-09-02
DE60323468D1 (de) 2008-10-23
EP1483369A1 (fr) 2004-12-08
US9382513B2 (en) 2016-07-05
CA2478125A1 (fr) 2003-09-18
JP2012110344A (ja) 2012-06-14

Similar Documents

Publication Publication Date Title
US9382513B2 (en) Method of making an avian cell line
US7432101B2 (en) Production of poxviruses with adherent or non adherent avian cell lines
PL215169B1 (pl) Sposób amplifikacji wirusów, kompozycja zawierajaca pokswirusa uzyskanego tym sposobem oraz jego zastosowanie do otrzymywania szczepionki
EP1500699A1 (en) Production of vaccinia virus with adherent or non adherent avian cell lines
US20080085554A1 (en) Culture Medium for Culturing Feeder Cells for Embryonic Stem Cells Culture and the Prepared Feeder Cells

Legal Events

Date Code Title Description
AS Assignment

Owner name: VIVALIS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PAIN, BERTRAND;GUEHENNEUX, FABIENNE;REEL/FRAME:014671/0010

Effective date: 20031030

STCB Information on status: application discontinuation

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