US20080274141A1 - Animal cells and processes for the replication of influenza viruses - Google Patents

Animal cells and processes for the replication of influenza viruses Download PDF

Info

Publication number
US20080274141A1
US20080274141A1 US12/146,689 US14668908A US2008274141A1 US 20080274141 A1 US20080274141 A1 US 20080274141A1 US 14668908 A US14668908 A US 14668908A US 2008274141 A1 US2008274141 A1 US 2008274141A1
Authority
US
United States
Prior art keywords
influenza viruses
mdck cells
serum
free medium
viruses
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
US12/146,689
Inventor
Albrecht Groner
Jurgen Vorlop
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.)
GSK Vaccines GmbH
Original Assignee
Novartis Vaccines and Diagnostics GmbH
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 Novartis Vaccines and Diagnostics GmbH filed Critical Novartis Vaccines and Diagnostics GmbH
Priority to US12/146,689 priority Critical patent/US20080274141A1/en
Publication of US20080274141A1 publication Critical patent/US20080274141A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • A61K2039/5252Virus inactivated (killed)
    • 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
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use 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
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16151Methods of production or purification of viral material

Definitions

  • the present invention relates to animal cells which can be infected by influenza viruses and are adapted to growth in suspension in serum-free medium, and to processes for the replication of influenza viruses in cell culture using these cells.
  • the present invention further relates to the influenza viruses obtainable by the process described and to vaccines which contain viruses of this type or constituents thereof.
  • All influenza vaccines which have been used since the 40s until today as permitted vaccines for the treatment of humans and animals consist of one or more virus strains which have been replicated in embryonate hens' eggs. These viruses are isolated from the allantoic fluid of infected hens' eggs and their antigens are used as vaccine either as intact virus particles or as virus particles disintegrated by detergents and/or solvents—so-called cleaved vaccine—or as isolated, defined virus proteins—so-called subunit vaccine. In all permitted vaccines, the viruses are inactivated by processes known to the person skilled in the art. The replication of live attenuated viruses, which are tested in experimental vaccines, is also carried out in embryonate hens' eggs.
  • the use of embryonate hens' eggs for vaccine production is time-, labor- and cost-intensive.
  • laborious purification processes substances from the hen's egg which lead to undesired side effects of the vaccine are separated from the viruses, and the viruses are concentrated.
  • eggs are not sterile (pathogen-free), it is additionally necessary to remove and/or to inactivate pyrogens and all pathogens which are possibly present.
  • Viruses of other vaccines such as, for example, rabies viruses, mumps, measles, rubella, polio viruses and FSME viruses can be replicated in cell cultures.
  • Economical vaccine production is possibly also achieved in that virus isolation and purification from a defined, sterile cell medium appears simpler than from the strongly protein-containing allantoic fluid.
  • the isolation and replication of influenza viruses in eggs leads to a selection of certain phenotypes, of which the majority differ from the clinical isolate.
  • virus replication in cell culture is also to be preferred from this aspect to that in eggs.
  • influenza viruses can be replicated in cell cultures. Beside hens' embryo cells and hamster cells (BHK21-F and HKCC), MDBK cells, and in particular MDCK cells have been described as suitable cells for the in-vitro replication of influenza viruses (Kilbourne, E. D., in: Influenza, pages 89-110, Plenum Medical Book Company—New York and London, 1987).
  • a prerequisite for a successful infection is the addition of proteases to the infection medium, preferably trypsin or similar serine proteases, as these proteases extracellularly cleave the precursor protein of hemagglutinin [HA 0 ] into active hemagglutinin [HA 1 and HA 2 ].
  • the U.S. Pat. No. 4,500,513 described the replication of influenza viruses in cell cultures of adherently growing cells. After cell proliferation, the nutrient medium is removed and fresh nutrient medium is added to the cells with infection of the cells with influenza viruses taking place simultaneously or shortly thereafter. A given time after the infection, protease (e.g. trypsin) is added in order to obtain an optimum virus replication. The viruses are harvested, purified and processed to give inactivated or attenuated vaccine.
  • protease e.g. trypsin
  • the serum necessary for the growth of the cells on the microcarriers (customarily fetal calf serum), however, contains trypsin inhibitors, so that even in this production method a change of medium to serum-free medium is necessary in order to achieve the cleavage of the influenza hemagglutinin by trypsin and thus an adequately high virus replication.
  • this methodology also requires opening of the culture vessels several times and thus brings with it the increased danger of contamination.
  • the present invention is thus based on the object of making available cells and processes which make possible simple and economical influenza virus replication in cell culture. This object is achieved by the provision of the embodiments indicated in the patent claims.
  • the invention thus relates to animal cells which can be infected by influenza viruses and which are adapted to growth in suspension in serum-free medium. It was found that it is possible with the aid of cells of this type to replicate influenza viruses in cell culture in a simple and economical manner.
  • a change of medium before infection to remove serum can be dispensed with an on the other hand the addition of protease can be carried out simultaneously to the infection.
  • the cells according to the invention are preferably vertebrate cells, e.g. avian cells, in particular hens' embryo cells.
  • the cells according to the invention are mammalian cells, e.g. from hamsters, cattle, monkeys or dogs, in particular kidney cells or cell lines derived from these. They are preferably cells which are derived from MDCK cells (ATCC CCL34 MDCK (NBL-2)), and particularly preferably cells of the cell line MDCK 33016. This cell line was deposited under the deposit number DSM ACC2219 on Jun.
  • the cell line MDCK 33016 is derived from the cell line MDCK by passaging and selection with respect to the capability of growing in suspension in serum-free medium and of replicating various viruses, e.g. orthomyxoviruses, paramyxoviruses, rhabdoviruses and flavoviruses. On account of these properties, these cells are suitable for economical replication of influenza viruses in cell culture by means of a simple and cost-effective process.
  • the present invention therefore also relates to a process for the replication of influenza viruses in cell culture, in which cells according to the invention are used, in particular a process which comprises the following steps:
  • the cells according to the invention can be cultured in the course of the process in various serum-free media known to the person skilled in the art (e.g. Iscove's medium, ultra CHO medium (BioWhittaker), EX-CELL (JRH Biosciences)). Otherwise, the cells for replication can also be cultured in the customary serum-containing media (e.g. MEM or DMEM medium with 0.5% to 10%, preferably 1.5% to 5%, of fetal calf serum) or protein-free media (e.g. PF-CHO (JRH Biosciences)).
  • Suitable culture vessels which can be employed in the course of the process according to the invention are all vessels known to the person skilled in the art, such as, for example, spinner bottles, roller bottles or fermenters.
  • the temperature for the proliferation of the cells before infection with influenza viruses is preferably 37° C.
  • Culturing for proliferation of the cells is carried out in a preferred embodiment of the process in a perfusion system, e.g. in a stirred vessel fermenter, using cell retention systems known to the person skilled in the art, such as, for example, centrifugation, filtration, spin filters and the like.
  • the cells are in this case preferably proliferated for 2 to 18 days, particularly preferably for 3 to 11 days. Exchange of the medium is carried out in the course of this, increasing from 0 to approximately 1 to 3 fermenter volumes per day.
  • the cells are proliferated up to very high cell densities in this manner, preferably up to approximately 2 ⁇ 10 7 cells/ml.
  • the perfusion rates during culture in the perfusion system can be regulated both via the cell count, the content of glucose, glutamine or lactate in the medium and via other parameters known to the person skilled in the art.
  • For infection with influenza viruses about 85% to 99%, preferably 93 to 97%, of the fermenter volume is transferred with cells to a further fermenter.
  • the cells remaining in the first fermenter can in turn be mixed with medium and replicated further in the perfusion system. In this manner, continuous cell culture for virus replication is available.
  • the cells in step (i) of the process according to the invention can preferably also be cultured in a batch process.
  • the cells according to the invention proliferate here at 37° C. with a generation time of 20 to 30 h up to a cell density of about 8 to 25 ⁇ 10 5 cells/ml.
  • the pH of the culture medium used in step (i) is regulated during culturing and is in the range from pH 6.6 to pH 7.8, preferably in the range from pH 6.8 to pH 7.3.
  • the pO 2 value is advantageously regulated in this step of the process and is preferably between 25% and 95%, in particular between 35% and 60% (based on the air saturation).
  • the infection of the cells cultured in suspension is preferably carried out when the cells in the batch process have achieved a cell density of about 8 to 25 ⁇ 10 5 cells/ml or about 5 to 20 ⁇ 10 6 cells/ml in the perfusion system.
  • the infection of the cells with influenza viruses is carried out at an m.o.i. (multiplicity of infection) of about 0.0001 to 10, preferably of 0.002 to 0.5.
  • the addition of the protease which brings about the cleavage of the precursor protein of hemagglutinin [HA 0 ] and thus the adsorption of the viruses on the cells can be carried out according to the invention shortly before, simultaneously to or shortly after the infection of the cells with influenza viruses. If the addition is carried out simultaneously to the infection, the protease can either be added directly to the cell culture to be infected or, for example, as a concentrate together with the virus inoculate.
  • the protease is preferably a serine protease, and particularly preferably trypsin.
  • trypsin is added to the cell culture to be infected up to a final concentration of 1 to 200 ⁇ g/ml, preferably 5 to 50 ⁇ g/ml, and particularly preferably 5 to 30 ⁇ g/ml in the culture medium.
  • trypsin reactivation can be carried out by fresh addition of trypsin in the case of the batch process or in the case of the perfusion system by continuous addition of a trypsin solution or by intermittent addition.
  • the trypsin concentration is preferably in the range from 1 ⁇ g/ml to 80 ⁇ g/ml.
  • the infected cell culture is cultured further to replicate the viruses, in particular until a maximum cytopathic effect or a maximum amount of virus antigen can be detected.
  • the culturing of the cells is carried out for 2 to 10 days, in particular for 3 to 7 days.
  • the culturing can in turn preferably be carried out in the perfusion system or in the batch process.
  • the cells are cultured at a temperature of 30° C. to 36° C., preferably of 32° C. to 34° C., after infection with influenza viruses.
  • the culturing of the infected cells at temperatures below 37° C., in particular in the temperature ranges indicated above, leads to the production of influenza viruses which after inactivation have an appreciably higher activity as vaccine, in comparison with influenza viruses which have been replicated at 37° C. in cell culture.
  • the culturing of the cells after infection with influenza viruses is in turn preferably carried out at regulated pH and pO 2 .
  • the pH in this case is preferably in the range from 6.6 to 7.8, particularly preferably from 6.8 to 7.2, and the pO 2 in the range from 25% to 150%, preferably from 30% to 75%, and particularly preferably in the range from 35% to 60% (based on the air saturation).
  • step (iv) of the process a substitution of the cell culture medium with freshly prepared medium, medium concentrate or with defined constituents such as amino acids, vitamins, lipid fractions, phosphates etc. for optimizing the antigen yield is also possible.
  • the cells can either be slowly diluted by further addition of medium or medium concentrate over several days or can be incubated during further perfusion with medium or medium concentrate decreasing from approximately 1 to 3 to 0 fermenter volumes/day.
  • the perfusion rates can in this case in turn be regulated by means of the cell count, the content of glucose, glutamine, lactate or lactate dehydrogenase in the medium or other parameters known to the person skilled in the art.
  • the harvesting and isolation of the replicated influenza viruses is carried out 2 to 10 days, preferably 3 to 7 days, after infection.
  • the cells or cell residues are separated from the culture medium by means of methods known to the person skilled in the art, for example by separators or filters.
  • concentration of the influenza viruses present in the culture medium is carried out by methods known to the person skilled in the art, such as, for example, gradient centrifugation, filtration, precipitation and the like.
  • influenza viruses which are obtainable by a process according to the invention. These can be formulated by known methods to give a vaccine for administration to humans or animals.
  • the immunogenicity or efficacy of the influenza viruses obtained as vaccine can be determined by methods known to the person skilled in the art, e.g. by means of the protection imparted in the loading experiment or as antibody titers of neutralizing antibodies.
  • the determination of the amount of virus or antigen produced can be carried out, for example, by the determination of the amount of hemagglutinin according to methods known to the person skilled in the art. It is known, for example, that cleaved hemagglutinin binds to erythrocytes of various species, e.g. to hens' erythrocytes. This makes possible a simple and rapid quantification of the viruses produced or of the antigen formed.
  • the invention also relates to vaccines which contain influenza viruses obtainable from the process according to the invention.
  • Vaccines of this type can optionally contain the additives customary for vaccines, in particular substances which increase the immune response, i.e. so-called adjuvants, e.g. hydroxide of various metals, constituents of bacterial cell walls, oils or saponins, and moreover customary pharmaceutically tolerable excipients.
  • adjuvants e.g. hydroxide of various metals, constituents of bacterial cell walls, oils or saponins, and moreover customary pharmaceutically tolerable excipients.
  • viruses can be present in the vaccines as intact virus particles, in particular as live attenuated viruses.
  • virus concentrates are adjusted to the desired titer and either lyophilized or stabilized in liquid form.
  • the vaccines according to the invention can contain disintegrated, i.e. inactivated, or intact, but inactivated viruses.
  • disintegrated i.e. inactivated, or intact, but inactivated viruses.
  • infectiousness of the viruses is destroyed by means of chemical and/or physical methods (e.g. by detergents or formaldehyde).
  • the vaccine is then adjusted to the desired amount of antigen and after possible admixture of adjuvants or after possible vaccine formulation, dispensed, for example, as liposomes, microspheres or “slow release” formulations.
  • the vaccines according to the invention can finally be present as subunit vaccine, i.e. they can contain defined, isolated virus constituents, preferably isolated proteins of the influenza virus. These constituents can be isolated from the influenza viruses by methods known to the person skilled in the art.
  • influenza viruses obtained by the process according to the invention can be used for diagnostic purposes.
  • the present invention also relates to diagnostic compositions which contain influenza viruses according to the invention or constituents of such viruses, if appropriate in combination with additives customary in this field and suitable detection agents.
  • the examples illustrate the invention.
  • a cell line which is adapted to growth in suspension culture and can be infected by influenza viruses is selected starting from MDCK cells (ATCC CCL34 MDCK (NBL-2), which had been proliferated by means of only a few passages or over several months in the laboratory. This selection was carried out by proliferation of the cells in roller bottles which were rotated at 16 rpm (instead of about 3 rpm as is customary for roller bottles having adherently growing cells). After several passages of the cells present suspended in the medium, cell strains growing in suspension were obtained. These cell strains were infected with influenza viruses and the strains were selected which produced the highest virus yield.
  • An increase in the rate of cells growing in suspension during the first passages at 16 rpm is achieved over 1 to 3 passages by the addition of selection systems known to the person skilled in the art, such as hypoxanthine, aminopterin and thymidine, or alanosine and adenine, individually or in combination.
  • selection systems known to the person skilled in the art, such as hypoxanthine, aminopterin and thymidine, or alanosine and adenine, individually or in combination.
  • the selection of cells growing in suspension is also possible in other agitated cell culture systems known to the person skilled in the art, such as stirred flasks.
  • highly virus-replicating cell clones can be established before selection as suspension cells by cell cloning in microtiter plates.
  • the adherently growing starting cells (after trypsinization) are diluted to a concentration of about 25 cells/ml with serum-containing medium and 100 ⁇ l each of this cell suspension are added to a well of a microtiter plate. If 100 ⁇ l of sterile-filtered medium from a 2 to 4-day old (homologous) cell culture (“conditioned medium”) are added to each well, the probability of growth of the cells inoculated at a very low cell density increases.
  • the wells are selected in which only one cell is contained; the cell lawn resulting therefrom is then passaged in larger cell culture vessels.
  • selection media e.g. hypoxanthine, aminopterin and thymidine, or alanosine and adenine, individually or in combination
  • the cell clones resulting in this way were selected with respect to their specific virus replication and then selected as suspension cells.
  • the selection of cells which are adapted to growth in serum-free medium can also be carried out by methods known to the person skilled in the art.
  • Examples of cells which are adapted to growth in serum-free medium in suspension and can be infected by influenza viruses are the cell lines MDCK 33016 (DSM ACC2219) and MDCK 13016, whose properties are described in the following examples.
  • the ratios indicated mean that a 1:X dilution of the virus harvest still has hemagglutinating properties.
  • the hemagglutinating properties can be determined, for example, as described in Mayer et al., Viryoghuren, [Virological Working Methods], Volume 1 (1974), pages 260-261 or in Grist, Diagnostic Methods in Clinical Virology, pages 72 to 75.
  • the cell line MDCK 33016 (DSM ACC2219) was replicated at 37° C. in Iscove's medium with a splitting rate of 1:8 to 1:12 twice weekly in a roller bottle which rotated at 16 rpm. Four days after transfer, a cell count of approximately 7.0 ⁇ 10 5 to 10 ⁇ 10 5 cells/ml was achieved. Simultaneously to the infection of the now 4-day old cell culture with various influenza virus strains (m.o.i. 0.1), the cell culture was treated with trypsin (25 ⁇ g/ml final concentration) and further incubated at 33° C., and the virus replication was determined on the 5th day after infection (Table III).
  • the cell line MDCK 33016 (DSM ACC2219) was inoculated in Iscove's medium with a cell inoculate of 1 ⁇ 10 5 cells/ml in a stirred vessel fermenter (working volume 8 l). At an incubation temperature of 37° C., a pO 2 of 50 ⁇ 10% (regulated) and a pH of 7.1 ⁇ 0.2 (regulated), the cells proliferated within 4 days to a cell density of 7 ⁇ 10 5 cells/ml.
  • virus stock solution 8 ml of virus stock solution (either A/PR/8/34 or A/Singapore/6/86 or A/Shanghai/11/87 or A/Beijing/1/87 or B/Massachusetts/71 or B/Yamagata/16/88 or B/Panama/45/90) and simultaneously 16 ml of a 1.25% strength trypsin solution were added to these cells and the inoculated cell culture was incubated further at 33° C. The virus replication was determined over 6 days (Table IV).
  • the cell line MDCK 13016 (obtained from an MDCK cell culture by selection pressure) was proliferated at 37° C. in ultra CHO medium with a splitting rate of 1:8 to 1:12 twice weekly in a roller bottle which rotated at 16 rpm. Four days after transfer, a cell count of approximately 7.0 ⁇ 10 5 to 10 ⁇ 10 5 cells/ml was achieved. The influence of the infective dose (m.o.i.) on the yield of antigen and infectiousness was investigated.
  • the cell culture was treated with trypsin (25 ⁇ g/ml final concentration) and incubated further at 37° C., and the virus replication was determined over 3 days (Table V).
  • the determination of the CCID 50 can in this case be carried out, for example, according to the method which is described in Paul, Zell-und Gewebekultur [Cell and tissue culture](1980), p. 395.
  • the cell line MDCK 33016 (DSM ACC2219) was proliferated at 37° C. in Iscove's medium with a splitting rate of 1:8 to 1:12 twice weekly in a roller bottle which rotated at 16 rpm. Four days after transfer, a cell count of approximately 7.0 ⁇ 10 5 to 10 ⁇ 10 5 cells/ml was achieved. The influence of a media substitution on the yield of antigen and infectiousness was investigated.
  • the cell line MDCK 33016 (ACC2219) was inoculated in Iscove's medium with a cell inoculate of 0.5 ⁇ 10 5 cells/ml in a stirred vessel fermenter (working volume 10 l). At an incubation temperature of 37° C., a pO 2 of 55 ⁇ 10% (regulated) and a pH of 7.1 ⁇ 0.2 (regulated), the cells proliferated within 4 days to a cell density of 7 ⁇ 10 5 cells/ml. 0.1 ml of virus stock solution (A/Singapore/6/86; m.o.i. about 0.0015) and simultaneously 16 ml of a 1.25% strength trypsin solution were added to these cells and the inoculated cell culture was incubated further at 33° C.
  • the virus replication was determined after 5 days and the virus was harvested. Cells and cell residues were removed by tangential flow filtration (Sarcoton Mini-Microsart Module with 0.45 ⁇ m pore size; filtration procedure according to the instructions of the manufacturer), no loss of antigen (measured as HA) being detectable in the filtrate.
  • the virus material was concentrated from 9.5 l to 600 ml by fresh tangential flow filtration (Sartocon Mini-Ultrasart Module with 100,000 NMWS (nominal molecular weight separation limit); filtration procedure according to the instructions of the manufacturer).
  • the amount of antigen in the concentrate was 5120 HA units (start 256 HA units; concentration factor 20), while the infectiousness in the concentrate was 9.2 log 10 CCID 50 (start 8.9 log 10 CCID 50 ; concentration factor 16); the loss of antigen and infectiousness was less than 1%, measured in the filtrate after the 100,000 NMWS filtration.
  • 1.6 ⁇ 10 8 cells of the cell line MDCK 33016 were suspended in UltraCHO medium (0.8 ⁇ 10 5 cells/ml) in the reactor vessel of Biostat MD (Braun Biotech Int., Melsungen, Germany) with an effective volume of 2000 ml and proliferated at 37° C. in perfusion operation with a rising flow rate (entry of oxygen by hose aeration (oxygen regulation 40 ⁇ 10% pO 2 ); pH regulation pH £7.2; cell retention by spin filter >95%).
  • the live cell count increased within 11 days by 200-fold to 175 ⁇ 10 5 cells/ml (Table VIIIa).
  • virus replication was carried out at 33° C. and the perfusion rate of 2 fermenter volumes/day was reduced to 0 within 7 days.
  • the trypsin necessary for virus replication was present in the UltraCHO medium which was used for the perfusion in a concentration of 10 ⁇ g/ml.
  • influenza virus A/PR/8/34 from Example 2—A/PR/8 replicated at 37° C.—(Experiment 2; vaccine A) and Example 4—A/PR/8 replicated at 33° C.—(vaccine B).
  • the influenza Viruses in the cell culture medium were separated from cells and cell fragments by low-speed centrifugation (2000 g, 20 min, 4° C.) and purified by a sucrose gradient centrifugation (10 to 50% (wt/wt) of linear sucrose gradient, 30,000 g, 2 h, 4° C.).
  • influenza virus-containing band was obtained, diluted 1:10 with PBS pH 7.2, and sedimented at 20,000 rpm, and the pellet was taken up in PBS (volume 50% of the original cell culture medium).
  • the influenza viruses were inactivated with formaldehyde (addition twice of 0.025% of a 35% strength formaldehyde solution at an interval of 24 h, incubation at 20° C. with stirring).
  • 10 NMRI mice each, 18 to 20 g in weight, were inoculated with 0.3 ml each of these inactivated experimental vaccines on day 0 and day 28 by subcutaneous injection.
  • mice were exposed 2 weeks after revaccination (6 weeks after the start of the experiment) by intranasal administration of 1000 LD 50 (lethal dose 50%). The results of the experiment are compiled in Table IX.
  • influenza viruses which had been replicated at 37° C. in cell culture with a high antigen yield (HA titer) only induced a low neutralizing antibody titer in the mouse and barely provided protection
  • influenza viruses which had been replicated at 33° C. in cell culture also with a high antigen yield (HA titer) induced very high neutralizing antibody titers in the mouse and led to very good protection.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Genetics & Genomics (AREA)
  • Pulmonology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Wood Science & Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Mycology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

Animal cells are described which can be infected by influenza viruses and which are adapted to growth in suspension in serum-free medium. Processes for the replication of influenza viruses in cell culture using these cells are furthermore described, as well as vaccines which contain the influenza viruses obtainable by the process or constituents thereof.

Description

  • This application is a continuation of U.S. Ser. No. 11/057,370, filed Feb. 15, 2005, which is a continuation of U.S. Ser. No. 10/670,788, filed Sep. 26, 2003, now abandoned, which is a continuation of U.S. Ser. No. 10/194,784, filed Jul. 12, 2002, now U.S. Pat. No. 6,656,720, which is a continuation of application U.S. Ser. No. 09/155,581, filed Sep. 29, 1998, now U.S. Pat. No. 6,455,298, which is a U.S. National Stage application under 35 U.S.C. § 371 of International Application No. PCT/IB97/00403, filed Apr. 1, 1997, which claims priority to German Application No. 196 12 966.4, filed Apr. 1, 1996. The disclosure of each of the foregoing is hereby incorporated by reference in its entirety.
  • The present invention relates to animal cells which can be infected by influenza viruses and are adapted to growth in suspension in serum-free medium, and to processes for the replication of influenza viruses in cell culture using these cells. The present invention further relates to the influenza viruses obtainable by the process described and to vaccines which contain viruses of this type or constituents thereof.
  • All influenza vaccines which have been used since the 40s until today as permitted vaccines for the treatment of humans and animals consist of one or more virus strains which have been replicated in embryonate hens' eggs. These viruses are isolated from the allantoic fluid of infected hens' eggs and their antigens are used as vaccine either as intact virus particles or as virus particles disintegrated by detergents and/or solvents—so-called cleaved vaccine—or as isolated, defined virus proteins—so-called subunit vaccine. In all permitted vaccines, the viruses are inactivated by processes known to the person skilled in the art. The replication of live attenuated viruses, which are tested in experimental vaccines, is also carried out in embryonate hens' eggs.
  • The use of embryonate hens' eggs for vaccine production is time-, labor- and cost-intensive. The eggs—from healthy flocks of hens monitored by veterinarians—have to be incubated before infection, customarily for 12 days. Before infection, the eggs have to be selected with respect to living embryos, as only these eggs are suitable for virus replication. After infection the eggs are again incubated, customarily for 2 to 3 days. The embryos still alive at this time are killed by cold and the allantoic fluid is then obtained from the individual eggs by aspiration. By means of laborious purification processes, substances from the hen's egg which lead to undesired side effects of the vaccine are separated from the viruses, and the viruses are concentrated. As eggs are not sterile (pathogen-free), it is additionally necessary to remove and/or to inactivate pyrogens and all pathogens which are possibly present.
  • Viruses of other vaccines such as, for example, rabies viruses, mumps, measles, rubella, polio viruses and FSME viruses can be replicated in cell cultures. As cell cultures originating from tested cell banks are pathogen-free and, in contrast to hens' eggs, are a defined virus replication system which (theoretically) is available in almost unlimited amounts, they make possible economical virus replication under certain circumstances even in the case of influenza viruses. Economical vaccine production is possibly also achieved in that virus isolation and purification from a defined, sterile cell medium appears simpler than from the strongly protein-containing allantoic fluid. The isolation and replication of influenza viruses in eggs leads to a selection of certain phenotypes, of which the majority differ from the clinical isolate. In contrast to this is the isolation and replication of the viruses in cell culture, in which no passage-dependent selection occurs (Oxford, J. S. et al., J. Gen. Virology 72 (1991), 185-189; Robertson, J. S. et al., J. Gen. Virology 74 (1993)2047-2051). For an effective vaccine, therefore, virus replication in cell culture is also to be preferred from this aspect to that in eggs.
  • It is known that influenza viruses can be replicated in cell cultures. Beside hens' embryo cells and hamster cells (BHK21-F and HKCC), MDBK cells, and in particular MDCK cells have been described as suitable cells for the in-vitro replication of influenza viruses (Kilbourne, E. D., in: Influenza, pages 89-110, Plenum Medical Book Company—New York and London, 1987). A prerequisite for a successful infection is the addition of proteases to the infection medium, preferably trypsin or similar serine proteases, as these proteases extracellularly cleave the precursor protein of hemagglutinin [HA0] into active hemagglutinin [HA1 and HA2]. Only cleaved hemagglutinin leads to the adsorption of the influenza viruses on cells with subsequent virus assimilation into the cells (Tobita, K. et al., Med Microbiol. Immunol., 162 (1975), 9-14; Lazarowitz, S. G. & Choppin, P. W., Virology, 68 (1975) 440-454; Klenk, H.-D. et al., Virology 68 (1975) 426-439) and thus to a further replication cycle of the virus in the cell culture.
  • The U.S. Pat. No. 4,500,513 described the replication of influenza viruses in cell cultures of adherently growing cells. After cell proliferation, the nutrient medium is removed and fresh nutrient medium is added to the cells with infection of the cells with influenza viruses taking place simultaneously or shortly thereafter. A given time after the infection, protease (e.g. trypsin) is added in order to obtain an optimum virus replication. The viruses are harvested, purified and processed to give inactivated or attenuated vaccine. Economical influenza virus replication as a prerequisite for vaccine production cannot be accomplished, however, using the methodology described in the patent mentioned, as the change of media, the subsequent infection as well as the addition of trypsin which is carried out later necessitates opening the individual cell culture vessels several times and is thus very labor-intensive. Furthermore, the danger increases of contamination of the cell culture by undesirable micro-organisms and viruses with each manipulation of the culture vessels. A more cost-effective alternative is cell proliferation in fermenter systems known to the person skilled in the art, the cells growing adherently on microcarriers. The serum necessary for the growth of the cells on the microcarriers (customarily fetal calf serum), however, contains trypsin inhibitors, so that even in this production method a change of medium to serum-free medium is necessary in order to achieve the cleavage of the influenza hemagglutinin by trypsin and thus an adequately high virus replication. Thus this methodology also requires opening of the culture vessels several times and thus brings with it the increased danger of contamination.
  • The present invention is thus based on the object of making available cells and processes which make possible simple and economical influenza virus replication in cell culture. This object is achieved by the provision of the embodiments indicated in the patent claims. The invention thus relates to animal cells which can be infected by influenza viruses and which are adapted to growth in suspension in serum-free medium. It was found that it is possible with the aid of cells of this type to replicate influenza viruses in cell culture in a simple and economical manner. By the use of the cells according to the invention, on the one hand a change of medium before infection to remove serum can be dispensed with an on the other hand the addition of protease can be carried out simultaneously to the infection. On the whole, only a single opening of the culture vessel for infection with influenza viruses is thus necessary, whereby the danger of the contamination of the cell cultures is drastically reduced. The expenditure of effort which would be associated with the change of medium, the infection and the subsequent protease addition is furthermore decreased. A further advantage is that the consumption of media is markedly decreased.
  • The cells according to the invention are preferably vertebrate cells, e.g. avian cells, in particular hens' embryo cells. In a particularly preferred embodiment, the cells according to the invention are mammalian cells, e.g. from hamsters, cattle, monkeys or dogs, in particular kidney cells or cell lines derived from these. They are preferably cells which are derived from MDCK cells (ATCC CCL34 MDCK (NBL-2)), and particularly preferably cells of the cell line MDCK 33016. This cell line was deposited under the deposit number DSM ACC2219 on Jun. 7, 1995 according to the requirements of the Budapest Convention for the International Recognition of the Deposition of Micro-organisms for the Purposes of Patenting in the German Collection of Micro-organisms (DSM), in Brunswick, Federal Republic of Germany, recognized as the international deposition site. The cell line MDCK 33016 is derived from the cell line MDCK by passaging and selection with respect to the capability of growing in suspension in serum-free medium and of replicating various viruses, e.g. orthomyxoviruses, paramyxoviruses, rhabdoviruses and flavoviruses. On account of these properties, these cells are suitable for economical replication of influenza viruses in cell culture by means of a simple and cost-effective process.
  • The present invention therefore also relates to a process for the replication of influenza viruses in cell culture, in which cells according to the invention are used, in particular a process which comprises the following steps:
  • i) proliferation of the cells according to the invention described above in serum-free medium in suspension;
  • ii) infection of the cells with influenza viruses;
  • iii) addition of protease shortly before, simultaneously to or shortly after infection; and
  • iv) further culturing of the infected cells and isolation of the replicated influenza viruses.
  • The cells according to the invention can be cultured in the course of the process in various serum-free media known to the person skilled in the art (e.g. Iscove's medium, ultra CHO medium (BioWhittaker), EX-CELL (JRH Biosciences)). Otherwise, the cells for replication can also be cultured in the customary serum-containing media (e.g. MEM or DMEM medium with 0.5% to 10%, preferably 1.5% to 5%, of fetal calf serum) or protein-free media (e.g. PF-CHO (JRH Biosciences)). Suitable culture vessels which can be employed in the course of the process according to the invention are all vessels known to the person skilled in the art, such as, for example, spinner bottles, roller bottles or fermenters.
  • The temperature for the proliferation of the cells before infection with influenza viruses is preferably 37° C. Culturing for proliferation of the cells (step (i)) is carried out in a preferred embodiment of the process in a perfusion system, e.g. in a stirred vessel fermenter, using cell retention systems known to the person skilled in the art, such as, for example, centrifugation, filtration, spin filters and the like.
  • The cells are in this case preferably proliferated for 2 to 18 days, particularly preferably for 3 to 11 days. Exchange of the medium is carried out in the course of this, increasing from 0 to approximately 1 to 3 fermenter volumes per day. The cells are proliferated up to very high cell densities in this manner, preferably up to approximately 2×107 cells/ml. The perfusion rates during culture in the perfusion system can be regulated both via the cell count, the content of glucose, glutamine or lactate in the medium and via other parameters known to the person skilled in the art. For infection with influenza viruses, about 85% to 99%, preferably 93 to 97%, of the fermenter volume is transferred with cells to a further fermenter. The cells remaining in the first fermenter can in turn be mixed with medium and replicated further in the perfusion system. In this manner, continuous cell culture for virus replication is available.
  • Alternatively to the perfusion system, the cells in step (i) of the process according to the invention can preferably also be cultured in a batch process. The cells according to the invention proliferate here at 37° C. with a generation time of 20 to 30 h up to a cell density of about 8 to 25×105 cells/ml.
  • In a preferred embodiment of the process according to the invention, the pH of the culture medium used in step (i) is regulated during culturing and is in the range from pH 6.6 to pH 7.8, preferably in the range from pH 6.8 to pH 7.3.
  • Furthermore, the pO2 value is advantageously regulated in this step of the process and is preferably between 25% and 95%, in particular between 35% and 60% (based on the air saturation). According to the invention, the infection of the cells cultured in suspension is preferably carried out when the cells in the batch process have achieved a cell density of about 8 to 25×105 cells/ml or about 5 to 20×106 cells/ml in the perfusion system.
  • In a further preferred embodiment, the infection of the cells with influenza viruses is carried out at an m.o.i. (multiplicity of infection) of about 0.0001 to 10, preferably of 0.002 to 0.5. The addition of the protease which brings about the cleavage of the precursor protein of hemagglutinin [HA0] and thus the adsorption of the viruses on the cells, can be carried out according to the invention shortly before, simultaneously to or shortly after the infection of the cells with influenza viruses. If the addition is carried out simultaneously to the infection, the protease can either be added directly to the cell culture to be infected or, for example, as a concentrate together with the virus inoculate. The protease is preferably a serine protease, and particularly preferably trypsin.
  • In a preferred embodiment, trypsin is added to the cell culture to be infected up to a final concentration of 1 to 200 μg/ml, preferably 5 to 50 μg/ml, and particularly preferably 5 to 30 μg/ml in the culture medium. During the further culturing of the infected cells according to step (iv) of the process according to the invention, trypsin reactivation can be carried out by fresh addition of trypsin in the case of the batch process or in the case of the perfusion system by continuous addition of a trypsin solution or by intermittent addition. In the latter case, the trypsin concentration is preferably in the range from 1 μg/ml to 80 μg/ml.
  • After infection, the infected cell culture is cultured further to replicate the viruses, in particular until a maximum cytopathic effect or a maximum amount of virus antigen can be detected. Preferably, the culturing of the cells is carried out for 2 to 10 days, in particular for 3 to 7 days. The culturing can in turn preferably be carried out in the perfusion system or in the batch process.
  • In a further preferred embodiment, the cells are cultured at a temperature of 30° C. to 36° C., preferably of 32° C. to 34° C., after infection with influenza viruses. The culturing of the infected cells at temperatures below 37° C., in particular in the temperature ranges indicated above, leads to the production of influenza viruses which after inactivation have an appreciably higher activity as vaccine, in comparison with influenza viruses which have been replicated at 37° C. in cell culture.
  • The culturing of the cells after infection with influenza viruses (step (iv)) is in turn preferably carried out at regulated pH and pO2. The pH in this case is preferably in the range from 6.6 to 7.8, particularly preferably from 6.8 to 7.2, and the pO2 in the range from 25% to 150%, preferably from 30% to 75%, and particularly preferably in the range from 35% to 60% (based on the air saturation).
  • During the culturing of the cells or virus replication according to step (iv) of the process, a substitution of the cell culture medium with freshly prepared medium, medium concentrate or with defined constituents such as amino acids, vitamins, lipid fractions, phosphates etc. for optimizing the antigen yield is also possible.
  • After infection with influenza viruses, the cells can either be slowly diluted by further addition of medium or medium concentrate over several days or can be incubated during further perfusion with medium or medium concentrate decreasing from approximately 1 to 3 to 0 fermenter volumes/day. The perfusion rates can in this case in turn be regulated by means of the cell count, the content of glucose, glutamine, lactate or lactate dehydrogenase in the medium or other parameters known to the person skilled in the art.
  • A combination of the perfusion system with a fed-batch process is further possible. In a preferred embodiment of the process, the harvesting and isolation of the replicated influenza viruses is carried out 2 to 10 days, preferably 3 to 7 days, after infection. To do this, for example, the cells or cell residues are separated from the culture medium by means of methods known to the person skilled in the art, for example by separators or filters. Following this the concentration of the influenza viruses present in the culture medium is carried out by methods known to the person skilled in the art, such as, for example, gradient centrifugation, filtration, precipitation and the like.
  • The invention further relates to influenza viruses which are obtainable by a process according to the invention. These can be formulated by known methods to give a vaccine for administration to humans or animals. The immunogenicity or efficacy of the influenza viruses obtained as vaccine can be determined by methods known to the person skilled in the art, e.g. by means of the protection imparted in the loading experiment or as antibody titers of neutralizing antibodies. The determination of the amount of virus or antigen produced can be carried out, for example, by the determination of the amount of hemagglutinin according to methods known to the person skilled in the art. It is known, for example, that cleaved hemagglutinin binds to erythrocytes of various species, e.g. to hens' erythrocytes. This makes possible a simple and rapid quantification of the viruses produced or of the antigen formed.
  • Thus the invention also relates to vaccines which contain influenza viruses obtainable from the process according to the invention. Vaccines of this type can optionally contain the additives customary for vaccines, in particular substances which increase the immune response, i.e. so-called adjuvants, e.g. hydroxide of various metals, constituents of bacterial cell walls, oils or saponins, and moreover customary pharmaceutically tolerable excipients.
  • The viruses can be present in the vaccines as intact virus particles, in particular as live attenuated viruses. For this purpose, virus concentrates are adjusted to the desired titer and either lyophilized or stabilized in liquid form.
  • In a further embodiment, the vaccines according to the invention can contain disintegrated, i.e. inactivated, or intact, but inactivated viruses. For this purpose, the infectiousness of the viruses is destroyed by means of chemical and/or physical methods (e.g. by detergents or formaldehyde). The vaccine is then adjusted to the desired amount of antigen and after possible admixture of adjuvants or after possible vaccine formulation, dispensed, for example, as liposomes, microspheres or “slow release” formulations.
  • In a further preferred embodiment, the vaccines according to the invention can finally be present as subunit vaccine, i.e. they can contain defined, isolated virus constituents, preferably isolated proteins of the influenza virus. These constituents can be isolated from the influenza viruses by methods known to the person skilled in the art.
  • Furthermore, the influenza viruses obtained by the process according to the invention can be used for diagnostic purposes. Thus the present invention also relates to diagnostic compositions which contain influenza viruses according to the invention or constituents of such viruses, if appropriate in combination with additives customary in this field and suitable detection agents. The examples illustrate the invention.
  • EXAMPLE 1 Preparation of Cell Lines which are Adapted to Growth in Suspension and can be Infected by Influenza Viruses
  • A cell line which is adapted to growth in suspension culture and can be infected by influenza viruses is selected starting from MDCK cells (ATCC CCL34 MDCK (NBL-2), which had been proliferated by means of only a few passages or over several months in the laboratory. This selection was carried out by proliferation of the cells in roller bottles which were rotated at 16 rpm (instead of about 3 rpm as is customary for roller bottles having adherently growing cells). After several passages of the cells present suspended in the medium, cell strains growing in suspension were obtained. These cell strains were infected with influenza viruses and the strains were selected which produced the highest virus yield. An increase in the rate of cells growing in suspension during the first passages at 16 rpm is achieved over 1 to 3 passages by the addition of selection systems known to the person skilled in the art, such as hypoxanthine, aminopterin and thymidine, or alanosine and adenine, individually or in combination. The selection of cells growing in suspension is also possible in other agitated cell culture systems known to the person skilled in the art, such as stirred flasks.
  • Alternatively, highly virus-replicating cell clones can be established before selection as suspension cells by cell cloning in microtiter plates. In this process, the adherently growing starting cells (after trypsinization) are diluted to a concentration of about 25 cells/ml with serum-containing medium and 100 μl each of this cell suspension are added to a well of a microtiter plate. If 100 μl of sterile-filtered medium from a 2 to 4-day old (homologous) cell culture (“conditioned medium”) are added to each well, the probability of growth of the cells inoculated at a very low cell density increases. By means of light-microscopic checking, the wells are selected in which only one cell is contained; the cell lawn resulting therefrom is then passaged in larger cell culture vessels. The addition of selection media (e.g. hypoxanthine, aminopterin and thymidine, or alanosine and adenine, individually or in combination) after the 1st cell passage leads over 1 to 3 passages to a greater distinguishability of the cell clones. The cell clones resulting in this way were selected with respect to their specific virus replication and then selected as suspension cells. The selection of cells which are adapted to growth in serum-free medium can also be carried out by methods known to the person skilled in the art.
  • Examples of cells which are adapted to growth in serum-free medium in suspension and can be infected by influenza viruses are the cell lines MDCK 33016 (DSM ACC2219) and MDCK 13016, whose properties are described in the following examples.
  • EXAMPLE 2 Replication of Influenza Viruses in the Cell Line MDCK 33016
  • The cell line MDCK 33016 (DSM ACC2219; obtained from an MDCK cell culture by selection pressure) was proliferated at 37° C. in Iscove's medium with a splitting rate of 1:8 to 1:12 twice weekly in a roller bottle which rotated at 16 rpm. Four days after transfer, a cell count of approximately 7.0×105 to 10×105 cells/ml was achieved. Simultaneously to the infection of the now 4-day old cell culture with the influenza virus strain A/PR/8/34 (m.o.i.=0.1), the cell culture was treated with trypsin (25 μg/ml final concentration) and cultured further at 37° C., and the virus replication was determined over 3 days (Table I).
  • TABLE I
    Replication of influenza A/PR/8/34 in roller bottles (cell line MDCK
    33016) after infection of a cell culture without change of medium,
    measured as antigen content (HA units)
    HA content after
    days after infection (dpi)
    1 dpi 2 dpi 3 dpi
    Experiment 1 1:64 1:512 1:1024
    Experiment 2 1:4 1:128 1:1024
    Experiment 3 1:8 1:32 1:512
  • The ratios indicated mean that a 1:X dilution of the virus harvest still has hemagglutinating properties. The hemagglutinating properties can be determined, for example, as described in Mayer et al., Virologische Arbeitsmethoden, [Virological Working Methods], Volume 1 (1974), pages 260-261 or in Grist, Diagnostic Methods in Clinical Virology, pages 72 to 75.
  • EXAMPLE 3 Replication of Influenza Viruses in the Cell Line MDCK 13016 in Spinner Bottles
  • The cell line MDCK 13016 was replicated at 37° C. in Iscove's medium with a splitting rate of 1:6 to 1:10 twice weekly in a spinner bottle (50 rpm). Four days after transfer, a cell count of 8.0×105 cells/ml was achieved. Simultaneously to the infection of the now 4 day old cell culture with the influenza virus strain A/PR/8/34 (m.o.i.=0.1), the cell culture was treated with trypsin (25 μg/ml final concentration) and incubated further at 33° C. and the virus replication was determined over 6 days (Table II).
  • TABLE II
    Replication of influenza A/PR/8/34 in spinner bottles (cell line MDCK
    13016) after infection of a cell culture without change of medium,
    measured as antigen content (HA units)
    HA content after days after infection (dpi)
    1 dpi 3 dpi 4 dpi 5 dpi 6 dpi
    Experiment 1 1:2 1:128 1:1024 1:1024 1:2048
    Experiment 2 1:4 1:512 1:2048 1:2048 1:1024
  • EXAMPLE 4 Replication of Various Influenza Strains in the Cell Line MDCK 33016 in Roller Bottles
  • The cell line MDCK 33016 (DSM ACC2219) was replicated at 37° C. in Iscove's medium with a splitting rate of 1:8 to 1:12 twice weekly in a roller bottle which rotated at 16 rpm. Four days after transfer, a cell count of approximately 7.0×105 to 10×105 cells/ml was achieved. Simultaneously to the infection of the now 4-day old cell culture with various influenza virus strains (m.o.i. 0.1), the cell culture was treated with trypsin (25 μg/ml final concentration) and further incubated at 33° C., and the virus replication was determined on the 5th day after infection (Table III).
  • TABLE III
    Replication of influenza strains in roller bottles (cell line MDCK 33016)
    after infection of a cell culture without change of medium, measured as
    antigen content (HA units)
    HA content 5 days
    after infection
    Influenza strain HA content
    A/Singapore/6/86 1:1024
    A/Sichuan/2/87 1:256
    A/Shanghai/11/87 1:256
    A/Guizhou/54/89 1:128
    A/Beijing/353/89 1:512
    B/Beijing/1/87 1:256
    B/Yamagata/16/88 1:512
    A/PR/8/34 1:1024
    A/Equi 1/Prague 1:512
    A/Equi 2/Miami 1:256
    A/Equi 2 Fontainebleau 1:128
    A/Swine/Ghent 1:512
    A/Swine/Iowa 1:1024
    A/Swine/Amsberg 1:512
  • EXAMPLE 5 Replication of Various Influenza Strains in MDCK 33016 Cells in the Fermenter
  • The cell line MDCK 33016 (DSM ACC2219) was inoculated in Iscove's medium with a cell inoculate of 1×105 cells/ml in a stirred vessel fermenter (working volume 8 l). At an incubation temperature of 37° C., a pO2 of 50±10% (regulated) and a pH of 7.1±0.2 (regulated), the cells proliferated within 4 days to a cell density of 7×105 cells/ml. 8 ml of virus stock solution (either A/PR/8/34 or A/Singapore/6/86 or A/Shanghai/11/87 or A/Beijing/1/87 or B/Massachusetts/71 or B/Yamagata/16/88 or B/Panama/45/90) and simultaneously 16 ml of a 1.25% strength trypsin solution were added to these cells and the inoculated cell culture was incubated further at 33° C. The virus replication was determined over 6 days (Table IV).
  • TABLE IV
    Replication of influenza virus strains in the fermenter (cell line
    MDCK 33016) after infection of a cell culture without change
    of medium, measured as antigen content (HA units)
    HA content after days after infection (dpi)
    1 dpi 3 dpi 4 dpi 5 dpi 6 dpi
    A/PR/8/34 1:64 1:512 1:1024 1:2048 1:2048
    A/Singapore 1:32 1:512 1:2048 1:2048 1:1024
    A/Shanghai 1:8 1:128 1:256 1:256 1:512
    A/Beijing 1:16 1:256 1:1024 1:1024 n.d.
    B/Yamagata 1:8 1:128 1:512 1:512 n.d.
    B/Massachusetts 1:4 1:128 1:256 1:512 n.d.
    B/Panama n.d. 1:128 1:256 n.d. 1:1024
  • EXAMPLE 6 Influence of the Infection Dose (m.o.i.) on Virus Replication
  • The cell line MDCK 13016 (obtained from an MDCK cell culture by selection pressure) was proliferated at 37° C. in ultra CHO medium with a splitting rate of 1:8 to 1:12 twice weekly in a roller bottle which rotated at 16 rpm. Four days after transfer, a cell count of approximately 7.0×105 to 10×105 cells/ml was achieved. The influence of the infective dose (m.o.i.) on the yield of antigen and infectiousness was investigated. Simultaneously to the infection of the now 4 day-old cell culture with the influenza virus stain A/PR/8/34 (m.o.i.=0.5 and m.o.i.=0.005), the cell culture was treated with trypsin (25 μg/ml final concentration) and incubated further at 37° C., and the virus replication was determined over 3 days (Table V).
  • TABLE V
    Replication of influenza virus strain PR/8/34 in the cell line
    MDCK 13016 in roller bottles after infection with an m.o.i. of
    0.5 or 0.005. The assessment of virus replication was carried
    out by antigen detection (HA) and infectiousness titer
    (CCID50 cell culture infective dose 50% in log10)
    Days after infection
    2 3 4 5
    PR/8/34 HA CCID50 HA CCID50 HA CCID50 HA CCID50
    m.o.i. = 0.5 128 5.1 256 5.7 512 5.3 1024 5.4
    m.o.i. = 0.005 64 4.9 512 8.0 512 8.3 1024 8.3
  • The determination of the CCID50 can in this case be carried out, for example, according to the method which is described in Paul, Zell-und Gewebekultur [Cell and tissue culture](1980), p. 395.
  • EXAMPLE 7 Influence of Media Substitution on Virus Replication
  • The cell line MDCK 33016 (DSM ACC2219) was proliferated at 37° C. in Iscove's medium with a splitting rate of 1:8 to 1:12 twice weekly in a roller bottle which rotated at 16 rpm. Four days after transfer, a cell count of approximately 7.0×105 to 10×105 cells/ml was achieved. The influence of a media substitution on the yield of antigen and infectiousness was investigated. The now 4-day old cell culture was infected with the influenza virus strain A/PR/8/34 (m.o.i.=0.05), the trypsin addition (20 μg/ml final concentration in the roller bottle) being carried out by mixing the virus inoculum with the trypsin stock solution. The cell culture was treated with additions of media and incubated further at 33° C., and the virus replication was determined over 5 days (Table VI).
  • TABLE VI
    Replication of influenza A/PR/8/34 in roller bottles (cell line MDCK
    33016); addition of 5% (final concentration) of a triple-concentrated
    Iscove's medium, of glucose (final concentration 3%) or glucose
    and casein hydrolysate (final concentration 3% or 0.1%) measured
    as antigen content (HA units)
    HA content after
    days after infection (dpi)
    Addition 1 dpi 3 dpi 4 dpi 5 dpi
    1:16 1:256 1:1024 1:1024
    (control)
    3x Iscove's 1:8 1:128 1:1024 1:2048
    Glucose 1:32 1:512 1:2048 1:2048
    Glucose/casein 1:8 1:128 1:512 1:1024
    hydrolysate
  • EXAMPLE 8 Replication of Influenza Viruses in MDCK 33016 Cells in the Fermenter and Obtainment of the Viruses
  • The cell line MDCK 33016 (ACC2219) was inoculated in Iscove's medium with a cell inoculate of 0.5×105 cells/ml in a stirred vessel fermenter (working volume 10 l). At an incubation temperature of 37° C., a pO2 of 55±10% (regulated) and a pH of 7.1±0.2 (regulated), the cells proliferated within 4 days to a cell density of 7×105 cells/ml. 0.1 ml of virus stock solution (A/Singapore/6/86; m.o.i. about 0.0015) and simultaneously 16 ml of a 1.25% strength trypsin solution were added to these cells and the inoculated cell culture was incubated further at 33° C. The virus replication was determined after 5 days and the virus was harvested. Cells and cell residues were removed by tangential flow filtration (Sarcoton Mini-Microsart Module with 0.45 μm pore size; filtration procedure according to the instructions of the manufacturer), no loss of antigen (measured as HA) being detectable in the filtrate. The virus material was concentrated from 9.5 l to 600 ml by fresh tangential flow filtration (Sartocon Mini-Ultrasart Module with 100,000 NMWS (nominal molecular weight separation limit); filtration procedure according to the instructions of the manufacturer). The amount of antigen in the concentrate was 5120 HA units (start 256 HA units; concentration factor 20), while the infectiousness in the concentrate was 9.2 log10 CCID50 (start 8.9 log10 CCID50; concentration factor 16); the loss of antigen and infectiousness was less than 1%, measured in the filtrate after the 100,000 NMWS filtration.
  • EXAMPLE 9 Replication of the Influenza Viruses in MDCK 33016 Cells in the Perfusion Fermenter
  • 1.6×108 cells of the cell line MDCK 33016 (DSM ACC2219) were suspended in UltraCHO medium (0.8×105 cells/ml) in the reactor vessel of Biostat MD (Braun Biotech Int., Melsungen, Germany) with an effective volume of 2000 ml and proliferated at 37° C. in perfusion operation with a rising flow rate (entry of oxygen by hose aeration (oxygen regulation 40±10% pO2); pH regulation pH £7.2; cell retention by spin filter >95%). The live cell count increased within 11 days by 200-fold to 175×105 cells/ml (Table VIIIa). 1990 ml of this cell culture were transferred to a 2nd perfusion fermenter (working volume 5 l), while the remaining cells were made up to 2000 ml again with medium and cell proliferation was carried out again in perfusion operation. In the 2nd perfusion fermenter (virus infection), the cells were infected with the influenza virus strain A/PR/8/34 (m.o.i.=0.01) with simultaneous addition of trypsin (10 μg/ml final concentration) and incubated for 1 h. The fermenter was then incubated further in perfusion operation (regulation of pO2: 40±10% and pH: £7.2). On the first day after infection, incubation was carried out at 37° C. and the virus harvest in the perfused cell culture supernatant was discarded. From the 2nd day after infection, virus replication was carried out at 33° C. and the perfusion rate of 2 fermenter volumes/day was reduced to 0 within 7 days. The trypsin necessary for virus replication was present in the UltraCHO medium which was used for the perfusion in a concentration of 10 μg/ml. The virus harvest (=perfused cell culture supernatant) was collected at 4° C. and the virus replication over 7 days was determined as the amount of antigen (Table VIIIb).
  • TABLE VIIIa
    Replication of MDCK 33016 cells in the perfusion fermenter
    Live Total
    cell count cell count Perfusion
    Day [105/ml] [105/ml] [1/day]
    0 0.6 0.6 0
    3 8.0 8.3 0
    4 14.6 17.5 1.1
    5 33.3 34.7 1.1
    6 49.8 53.6 2.1
    7 84.5 85.6 3.9
    9 82.6 84.9 4.0
    9 100.8 104.8 4.1
    10 148.5 151.0 4.0
    11 175.8 179.6 3.9
  • TABLE VIIIb
    Replication of influenza A/PR/8/34 in the perfusion fermenter
    (cell line MDCK 33016), measured as antigen content (HA units)
    in the cumulated perfused cell culture supernatant
    Day after HA content in virus Medium addition Total amount virus
    infection harvest (perfusion) harvest
    1 <4 41 01
    2 8 41 41
    3 64 31 71
    4 256 21 91
    5 2048 21 111
    6 4096 21 121
    7 4096 01 121
  • EXAMPLE 10 Preparation of an Experimental Influenza Vaccine
  • An experimental vaccine was prepared from influenza virus A/PR/8/34 from Example 2—A/PR/8 replicated at 37° C.—(Experiment 2; vaccine A) and Example 4—A/PR/8 replicated at 33° C.—(vaccine B). The influenza Viruses in the cell culture medium were separated from cells and cell fragments by low-speed centrifugation (2000 g, 20 min, 4° C.) and purified by a sucrose gradient centrifugation (10 to 50% (wt/wt) of linear sucrose gradient, 30,000 g, 2 h, 4° C.). The influenza virus-containing band was obtained, diluted 1:10 with PBS pH 7.2, and sedimented at 20,000 rpm, and the pellet was taken up in PBS (volume 50% of the original cell culture medium). The influenza viruses were inactivated with formaldehyde (addition twice of 0.025% of a 35% strength formaldehyde solution at an interval of 24 h, incubation at 20° C. with stirring). 10 NMRI mice each, 18 to 20 g in weight, were inoculated with 0.3 ml each of these inactivated experimental vaccines on day 0 and day 28 by subcutaneous injection. 2 and 4 weeks after the inoculation and also 1 and 2 weeks after revaccination, blood was taken from the animals to determine the titer of neutralizing antibodies against A/PR/8/34. To determine the protection rate, the mice were exposed 2 weeks after revaccination (6 weeks after the start of the experiment) by intranasal administration of 1000 LD50 (lethal dose 50%). The results of the experiment are compiled in Table IX.
  • TABLE IX
    Efficacy of experimental vaccines: for vaccine A the influenza
    virus A/PR/8/34 was replicated at 37° C. and for vaccine B at
    33° C. The titer of neutralizing antibodies against A/PR/8 and
    also the protection rate after exposure of the mice were investigated
    Protection
    Titer of neutralizing antibodies/ml* rate
    2 w Number
    pvacc 4 w pvacc 1 w prevacc 2 w prevacc living/total
    Vaccine A <28 56 676 1,620 1/10
    Vaccine B 112 1,549 44,670 112,200 9/10
    *Weeks after vaccination (w pvacc) and weeks after revaccination (w prevacc)
  • The experiments confirm that influenza viruses which had been replicated at 37° C. in cell culture with a high antigen yield (HA titer) only induced a low neutralizing antibody titer in the mouse and barely provided protection, while influenza viruses which had been replicated at 33° C. in cell culture also with a high antigen yield (HA titer) induced very high neutralizing antibody titers in the mouse and led to very good protection.

Claims (30)

1. A process for making a vaccine for administration to humans or animals, the process comprising:
(a) incubating MDCK cells in a serum-free medium;
(b) adding influenza viruses to the serum-free medium to infect the MDCK cells incubated in step (a), wherein protease is present in the serum-free medium or added to the serum-free medium before, during, or after infection with the influenza viruses;
(c) culturing the MDCK cells infected in step (b) to replicate the influenza viruses;
(d) isolating the influenza viruses replicated in step (c); and
(e) formulating the influenza viruses isolated in step (d) to provide the vaccine, wherein the vaccine contains disintegrated viruses.
2. The process of claim 1, wherein the MDCK cells incubated in step (a) are incubated in a perfusion system.
3. The process of claim 1, wherein the MDCK cells incubated in step (a) are incubated in a batch process.
4. The process of claim 1, wherein the MDCK cells infected in step (b) are cultured in step (c) at a regulated pH and pO2.
5. The process of claim 1, wherein the serum-free medium is a protein-free medium.
6. The process of claim 1, wherein the influenza viruses replicated in step (c) are isolated in step (d) by a process comprising centrifugation.
7. The process of claim 6, wherein the influenza viruses replicated in step (c) are isolated in step (d) by a process comprising sucrose gradient centrifugation.
8. The process of claim 1, wherein the MDCK cells are of the cell line MDCK 33016 (DSM ACC 2219).
9. The process of claim 1, wherein the influenza viruses isolated in step (d) are inactivated by formaldehyde.
10. The process of claim 1, wherein the vaccine comprises an adjuvant.
11. The process of claim 1, wherein the influenza viruses isolated in step (d) are disintegrated using a detergent, prior to step (e).
12. The process of claim 1, wherein the influenza viruses isolated in step (d) are disintegrated using a solvent, prior to step (e).
13. The process of claim 1, wherein the influenza viruses isolated in step (d) are disintegrated using a detergent and a solvent, prior to step (e).
14. The process of claim 1, wherein the vaccine is a subunit vaccine.
15. The process of claim 1, wherein step (e) comprises adjusting the vaccine containing disintegrated viruses to a desired amount of antigen.
16. The process of claim 1, wherein the protease is trypsin and the trypsin is present in the serum-free medium, or added to the serum-free medium before, during, or after infection with influenza viruses, at a concentration from 1 to 200 μg/ml.
17. The process of claim 1, wherein the influenza viruses have a genome segment from strain A/PR/8134.
18. The process of claim 1, wherein the MDCK cells are incubated adherently.
19. The process of claim 18, wherein the MDCK cells are incubated adherently on microcarriers.
20. The process of claim 1, wherein the MDCK cells are incubated in suspension.
21. The process of claim 1, wherein the MDCK cells infected in step (b) are cultured in step (c) at a temperature below 37° C.
22. The process of claim 21, wherein the MDCK cells infected in step (b) are cultured in step (c) at a temperature of 30° C. to 36° C.
23. The process of claim 21, wherein the MDCK cells infected in step (b) are cultured in step (c) for 2 to 10 days.
24. The process of claim 1, wherein the process is performed in a culture vessel that is not opened after adding the influenza viruses in step (b) or before isolating the influenza viruses in step (d).
25. The process of claim 1, wherein (i) the influenza viruses replicated in step (c) are isolated in step (d) by a process comprising centrifugation, (ii) the influenza viruses isolated in step (d) are inactivated by formaldehyde, and (iii) the MDCK cells are incubated adherently.
26. A process for making a vaccine for administration to humans or animals, the process comprising:
(a) incubating MDCK cells which can be infected by influenza viruses in a serum-free medium;
(b) adding the influenza viruses to the serum-free medium to infect the MDCK cells incubated in step (a), wherein protease is present in the serum-free medium or added to the serum-free medium before, during, or after infection with influenza viruses;
(c) culturing the MDCK cells infected in step (b) to replicate the influenza viruses;
(d) isolating the influenza viruses replicated in step (c) by a process comprising centrifugation; and
(e) formulating the influenza viruses replicated in step (c) to provide the vaccine, wherein the vaccine contains live attenuated viruses.
27. The process of claim 26, wherein step (e) comprises adjusting the influenza viruses isolated in step (d) to a desired titer.
28. The process of claim 26, wherein step (e) further comprises lyophilizing the viruses.
29. The process of claim 26, wherein step (e) further comprises stabilizing the viruses in liquid form.
30. A process for making a vaccine for administration to humans or animals, the process comprising:
(a) incubating MDCK cells adherently in a serum-free medium;
(b) adding influenza viruses, having a genome segment from strain A/PR/8/34, to the serum-free medium to infect the MDCK cells incubated in step (a), wherein protease is present in the serum-free medium or added to the serum-free medium before, during, or after infection with the influenza viruses;
(c) culturing the MDCK cells infected in step (b) to replicate the influenza viruses;
(d) isolating the influenza viruses replicated in step (c);
(e) inactivating the influenza viruses isolated in step (d) by formaldehyde; and
(f) formulating the influenza viruses inactivated in step (e) to provide the vaccine, wherein the vaccine contains disintegrated viruses.
US12/146,689 1996-04-01 2008-06-26 Animal cells and processes for the replication of influenza viruses Abandoned US20080274141A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/146,689 US20080274141A1 (en) 1996-04-01 2008-06-26 Animal cells and processes for the replication of influenza viruses

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
DE19612966A DE19612966B4 (en) 1996-04-01 1996-04-01 MDCK cells and methods of propagating influenza viruses
DE19612966.4 1996-04-01
PCT/IB1997/000403 WO1997037000A1 (en) 1996-04-01 1997-04-01 Animal cells and processes for the replication of influenza viruses
US09/155,581 US6455298B1 (en) 1996-04-01 1997-04-01 Animal cells and processes for the replication of influenza viruses
US10/194,784 US6656720B2 (en) 1996-04-01 2002-07-12 Animal cells and processes for the replication of influenza viruses
US67078803A 2003-09-26 2003-09-26
US11/057,370 US20050202553A1 (en) 1996-04-01 2005-02-15 Animal cells and processes for the replication of influenza viruses
US12/146,689 US20080274141A1 (en) 1996-04-01 2008-06-26 Animal cells and processes for the replication of influenza viruses

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/057,370 Continuation US20050202553A1 (en) 1996-04-01 2005-02-15 Animal cells and processes for the replication of influenza viruses

Publications (1)

Publication Number Publication Date
US20080274141A1 true US20080274141A1 (en) 2008-11-06

Family

ID=7790126

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/155,581 Expired - Lifetime US6455298B1 (en) 1996-04-01 1997-04-01 Animal cells and processes for the replication of influenza viruses
US10/194,784 Expired - Lifetime US6656720B2 (en) 1996-04-01 2002-07-12 Animal cells and processes for the replication of influenza viruses
US11/057,370 Abandoned US20050202553A1 (en) 1996-04-01 2005-02-15 Animal cells and processes for the replication of influenza viruses
US12/146,689 Abandoned US20080274141A1 (en) 1996-04-01 2008-06-26 Animal cells and processes for the replication of influenza viruses

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US09/155,581 Expired - Lifetime US6455298B1 (en) 1996-04-01 1997-04-01 Animal cells and processes for the replication of influenza viruses
US10/194,784 Expired - Lifetime US6656720B2 (en) 1996-04-01 2002-07-12 Animal cells and processes for the replication of influenza viruses
US11/057,370 Abandoned US20050202553A1 (en) 1996-04-01 2005-02-15 Animal cells and processes for the replication of influenza viruses

Country Status (10)

Country Link
US (4) US6455298B1 (en)
EP (2) EP0904351B1 (en)
JP (7) JP4243349B2 (en)
AT (1) ATE482267T1 (en)
DE (2) DE19612966B4 (en)
DK (1) DK0904351T3 (en)
ES (1) ES2352638T3 (en)
HK (1) HK1019233A1 (en)
PT (1) PT904351E (en)
WO (1) WO1997037000A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100183671A1 (en) * 2007-06-27 2010-07-22 Novartis Vaccines & Diagnostics GmbH & Co., KG Low-additive influenza vaccines
US9890363B2 (en) 2015-07-06 2018-02-13 Wisconsin Alumni Research Foundation (Warf) Influenza virus replication for vaccine development
US9926535B2 (en) 2006-03-31 2018-03-27 Wisconsin Alumni Research Foundation (Warf) High titer recombinant influenza viruses for vaccines
US9950057B2 (en) 2013-07-15 2018-04-24 Wisconsin Alumni Research Foundation (Warf) High titer recombinant influenza viruses with enhanced replication in MDCK or vero cells or eggs
US10053671B2 (en) 2014-06-20 2018-08-21 Wisconsin Alumni Research Foundation (Warf) Mutations that confer genetic stability to additional genes in influenza viruses
US10119124B2 (en) 2007-06-18 2018-11-06 Wisconsin Alumni Research Foundation (Warf) Influenza M2 protein mutant viruses as live influenza attenuated vaccines
US10130697B2 (en) 2010-03-23 2018-11-20 Wisconsin Alumni Research Foundation (Warf) Vaccines comprising mutant attenuated influenza viruses
US10633422B2 (en) 2015-06-01 2020-04-28 Wisconsin Alumni Research Foundation (Warf) Influenza virus replication by inhibiting microRNA lec7C binding to influenza viral cRNA and mRNA
US10808229B2 (en) 2009-10-26 2020-10-20 Wisconsin Alumni Research Foundation (“WARF”) High titer recombinant influenza viruses with enhanced replication in vero cells
US11197925B2 (en) 2016-02-19 2021-12-14 Wisconsin Alumni Research Foundation (Warf) Influenza B virus replication for vaccine development
US11241492B2 (en) 2019-01-23 2022-02-08 Wisconsin Alumni Research Foundation (Warf) Mutations that confer genetic stability to genes in influenza viruses
US11390649B2 (en) 2019-05-01 2022-07-19 Wisconsin Alumni Research Foundation (Warf) Influenza virus replication for vaccine development
US11807872B2 (en) 2019-08-27 2023-11-07 Wisconsin Alumni Research Foundation (Warf) Recombinant influenza viruses with stabilized HA for replication in eggs
US11851648B2 (en) 2019-02-08 2023-12-26 Wisconsin Alumni Research Foundation (Warf) Humanized cell line

Families Citing this family (117)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE19612967A1 (en) * 1996-04-01 1997-10-02 Behringwerke Ag Process for the propagation of influenza viruses in cell culture, and the influenza viruses obtainable by the process
DE19612966B4 (en) 1996-04-01 2009-12-10 Novartis Vaccines And Diagnostics Gmbh & Co. Kg MDCK cells and methods of propagating influenza viruses
HUP0004240A2 (en) * 1997-10-25 2001-03-28 Roche Diagnostics Gmbh Suspension packing cell lines for retrovirus vestors
IL136736A (en) 1997-12-24 2004-06-01 Duphar Int Res Preparation of cells for production of biologicals
KR100797547B1 (en) * 2000-03-03 2008-01-24 자이단호진 가가쿠오요비겟세이료호겐쿠쇼 Cell usable in serum-free culture and suspension culture and process for producing virus for vaccine by using the cell
JP3547715B2 (en) * 2001-03-06 2004-07-28 榮 新垣 Apparatus and method for vector purification
DE10144903A1 (en) * 2001-09-12 2003-03-27 Chiron Behring Gmbh & Co Replication of virus in cell cultures, useful for preparing vaccines and diagnostic reagents, where replication of cells and virus is simultaneous
DE10144906B4 (en) * 2001-09-12 2013-11-28 Novartis Vaccines And Diagnostics Gmbh Process for the large-scale production of vaccines
US20030078471A1 (en) * 2001-10-18 2003-04-24 Foley Frederick J. Manipulation of an organ
US6951752B2 (en) * 2001-12-10 2005-10-04 Bexter Healthcare S.A. Method for large scale production of virus antigen
EP1499348B1 (en) * 2002-04-26 2014-10-01 MedImmune, LLC Method for producing infectious influenza b virus in cell culture
US7465456B2 (en) 2002-04-26 2008-12-16 Medimmune, Llc Multi plasmid system for the production of influenza virus
KR101251902B1 (en) 2003-02-25 2013-04-08 메디뮨 엘엘씨 Methods of producing inflenza vaccine compositions
US20060110406A1 (en) 2003-02-25 2006-05-25 Medimmune Vaccines, Inc. Refrigerator-temperature stable influenza vaccine compositions
US7270990B2 (en) * 2003-06-20 2007-09-18 Microbix Biosystems, Inc. Virus production
WO2005026333A1 (en) * 2003-09-09 2005-03-24 Japan Science And Technology Agency Technique for suspension culture of adhesive cells using protease and method of producing virus with the use of the cells as host
CA2551489C (en) * 2003-12-23 2013-09-03 Gregory Duke Multi plasmid system for the production of influenza virus
US20080254065A1 (en) 2004-03-09 2008-10-16 Chiron Corporation Influenza Virus Vaccines
WO2005099751A2 (en) * 2004-04-01 2005-10-27 Alza Corporation Apparatus and method for transdermal delivery of influenza vaccine
MXPA06013411A (en) * 2004-05-20 2007-07-04 Id Biomedical Corp Process for the production of an influenza vaccine.
EP2179744B1 (en) 2004-09-09 2010-12-01 Novartis Vaccines and Diagnostics GmbH Decreasing potential latrogenic risks associated with influenza vaccines
ES2633471T3 (en) 2004-10-06 2017-09-21 Medimmune, Llc Cold-stable flu vaccine compositions
DE102004049290A1 (en) * 2004-10-09 2006-04-20 Bayer Healthcare Ag Process for the preparation of virus material
JP2008519042A (en) 2004-11-03 2008-06-05 ノバルティス ヴァクシンズ アンド ダイアグノスティクス, インコーポレイテッド Influenza vaccination
PT1824990E (en) 2004-12-08 2015-06-11 Medimmune Llc Methods of producing influenza vaccine compositions
ATE547511T1 (en) * 2004-12-23 2012-03-15 Medimmune Llc NON-TUMOR FORMING MDCK CELL LINE FOR REPRODUCTION OF VIRUSES
US7572620B2 (en) 2005-01-11 2009-08-11 Wisconsin Alumni Research Foundation H3 equine influenza A virus
FR2884255B1 (en) 2005-04-11 2010-11-05 Vivalis USE OF EBX AVIATION STEM CELL LINES FOR THE PRODUCTION OF INFLUENZA VACCINE
US20070111309A1 (en) * 2005-10-04 2007-05-17 Daelli Marcelo G Vero cell line adapted to grow in suspension
CN101365480B (en) 2005-11-01 2014-11-05 诺华疫苗和诊断有限两合公司 Cell-derived viral vaccines with low levels of residual cell DNA by beta-propiolactone treatment
PT1951299E (en) 2005-11-04 2012-02-28 Novartis Vaccines & Diagnostic Influenza vaccines including combinations of particulate adjuvants and immunopotentiators
US20090220541A1 (en) 2005-11-04 2009-09-03 Novartis Vaccines And Diagnostics Srl Emulsions with free aqueous-phase surfactant for adjuvanting split influenza vaccines
JP2009514850A (en) 2005-11-04 2009-04-09 ノバルティス ヴァクシンズ アンド ダイアグノスティクス エスアールエル Influenza vaccine with reduced amount of oil-in-water emulsion as adjuvant
DK2368572T3 (en) 2005-11-04 2020-05-25 Seqirus Uk Ltd Adjuvant vaccines with non-virion antigens produced from influenza viruses grown in cell culture
NZ592713A (en) 2005-11-04 2012-12-21 Novartis Vaccines & Diagnostic Adjuvanted influenza vaccines including a cytokine-inducing agents other than an agonist of Toll-Like Receptor 9
EP2478916B1 (en) 2006-01-27 2020-04-01 Seqirus UK Limited Influenza vaccines containing hemagglutinin and matrix proteins
EP2382987A1 (en) 2006-03-24 2011-11-02 Novartis Vaccines and Diagnostics GmbH Storage of influenza vaccines without refrigeration
WO2007132763A1 (en) 2006-05-11 2007-11-22 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Method for proliferation of influenza virus
GB0614460D0 (en) 2006-07-20 2006-08-30 Novartis Ag Vaccines
AU2007297178B2 (en) * 2006-09-11 2014-07-24 Novartis Ag Making influenza virus vaccines without using eggs
MX2009002741A (en) * 2006-09-15 2009-05-11 Medimmune Llc Mdck cell lines supporting viral growth to high titers and bioreactor process using the same.
CN101553252A (en) 2006-12-06 2009-10-07 诺华有限公司 Vaccines including antigen from four strains of influenza virus
US8673613B2 (en) 2007-06-18 2014-03-18 Medimmune, Llc Influenza B viruses having alterations in the hemaglutinin polypeptide
US20090042253A1 (en) * 2007-08-09 2009-02-12 Wyeth Use of perfusion to enhance production of fed-batch cell culture in bioreactors
GB0810305D0 (en) 2008-06-05 2008-07-09 Novartis Ag Influenza vaccination
CN101971030A (en) 2007-12-24 2011-02-09 诺华有限公司 Assays for adsorbed influenza vaccines
US8506966B2 (en) 2008-02-22 2013-08-13 Novartis Ag Adjuvanted influenza vaccines for pediatric use
AU2009227674C1 (en) 2008-03-18 2015-01-29 Seqirus UK Limited Improvements in preparation of influenza virus vaccine antigens
TW201012930A (en) * 2008-06-16 2010-04-01 Intervet Int Bv Method of replicating viruses in suspension
AU2009276304B2 (en) 2008-08-01 2012-10-11 Gamma Vaccines Pty Limited Influenza vaccines
RU2547587C2 (en) 2008-09-24 2015-04-10 Медиммун, Ллк Methods for cell culture, virus replication and purification
CN102215865B (en) 2008-09-24 2013-10-30 米迪缪尼有限公司 Methods for purification of viruses
WO2010079081A1 (en) 2009-01-07 2010-07-15 Glaxosmithkline Biologicals S.A. Methods for recovering a virus or a viral antigen produced by cell culture
JP5843615B2 (en) 2009-02-06 2016-01-13 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 Purification of viruses or viral antigens by density gradient ultracentrifugation
DK2396032T3 (en) 2009-02-10 2016-12-19 Seqirus Uk Ltd Influenza vaccines with reduced amounts of squalene
CA2752039A1 (en) 2009-02-10 2010-08-19 Novartis Ag Influenza vaccine regimens for pandemic-associated strains
EP2396031A1 (en) 2009-02-10 2011-12-21 Novartis AG Influenza vaccines with increased amounts of h3 antigen
FR2949344A1 (en) 2009-04-27 2011-03-04 Novartis Ag FLU PROTECTIVE VACCINES
SI2427576T1 (en) 2009-05-08 2016-10-28 Seqirus UK Limited Generic assays for detection of influenza viruses
SI2401384T1 (en) 2009-05-21 2013-01-31 Novartis Ag Reverse genetics using non-endogenous pol i promoters
CN103764160A (en) 2009-05-29 2014-04-30 诺华有限公司 Assays for influenza virus hemagglutinins
WO2011012999A1 (en) 2009-07-31 2011-02-03 Novartis Ag Reverse genetics systems
AU2010293902A1 (en) 2009-09-10 2012-03-22 Novartis Ag Combination vaccines against respiratory tract diseases
CA2778332A1 (en) 2009-10-20 2011-04-28 Novartis Ag Improved reverse genetics methods for virus rescue
PL2356983T3 (en) 2009-12-03 2013-08-30 Novartis Ag Circulation of components during homogenization of emulsions
DE102009056883B4 (en) 2009-12-03 2012-08-16 Novartis Ag Vaccine adjuvants and improved methods of making the same
EA025622B1 (en) 2009-12-03 2017-01-30 Новартис Аг Method for manufacture of a squalene-containing oil-in-water emulsion
SG10201501613SA (en) 2009-12-03 2015-04-29 Novartis Ag Arranging interaction and back pressure chambers for microfluidization
DE102009056871A1 (en) 2009-12-03 2011-06-22 Novartis AG, 4056 Vaccine adjuvants and improved methods of making the same
DE102009056884B4 (en) 2009-12-03 2021-03-18 Novartis Ag Vaccine Adjuvants and Improved Methods for Making Same
WO2011095596A1 (en) 2010-02-04 2011-08-11 Vivalis Fed-batch process using concentrated cell culture medium for the efficient production of biologics in eb66 cells
EP2545172B2 (en) 2010-03-08 2017-12-06 Novartis AG Methods of testing for intracellular pathogens
CA2797059A1 (en) 2010-05-03 2011-11-10 Glaxosmithkline Biologicals S.A. Novel method
KR20130079398A (en) 2010-05-06 2013-07-10 노파르티스 아게 Organic peroxide compounds for microorganism inactivation
PT3489211T (en) 2010-05-12 2020-09-22 Novartis Ag Improved methods for preparing squalene
AU2011254204B2 (en) 2010-05-21 2015-08-20 Seqirus UK Limited Influenza virus reassortment method
KR20130081659A (en) 2010-06-01 2013-07-17 노파르티스 아게 Concentration of vaccine antigens with lyophilization
PT2575872T (en) 2010-06-01 2020-11-19 Seqirus Uk Ltd Concentration of influenza vaccine antigens without lyophilization
AU2011307655B2 (en) * 2010-08-16 2015-12-17 Cevec Pharmaceuticals Gmbh Permanent human amniocyte cell lines for producing influenza viruses
US9051361B2 (en) 2010-08-17 2015-06-09 National Health Research Institutes Immunogenic compositions and uses thereof
WO2012023044A1 (en) 2010-08-20 2012-02-23 Novartis Ag Soluble needle arrays for delivery of influenza vaccines
US9657359B2 (en) 2010-09-07 2017-05-23 Novartis Ag Generic assays for detection of mamalian reovirus
WO2012100302A1 (en) 2011-01-27 2012-08-02 Gamma Vaccines Pty Limited Combination vaccines
AU2012221740B2 (en) 2011-02-25 2016-11-17 Novartis Ag Exogenous internal positive control
GB201216121D0 (en) 2012-09-10 2012-10-24 Novartis Ag Sample quantification by disc centrifugation
AU2012324398A1 (en) 2011-10-20 2014-05-01 Seqirus UK Limited Adjuvanted influenza B virus vaccines for pediatric priming
JP2016539314A (en) 2011-12-12 2016-12-15 ノバルティス アーゲー Assay method for influenza virus hemagglutinin
AU2014203051C1 (en) * 2012-03-02 2016-09-01 Seqirus UK Limited Influenza Virus Reassortment
JP6054883B2 (en) * 2012-03-02 2016-12-27 ノバルティス アーゲー Influenza virus reassembly
WO2013131898A1 (en) 2012-03-06 2013-09-12 Crucell Holland B.V. Improved vaccination against influenza
JP6152535B2 (en) * 2012-04-16 2017-06-28 公益財団法人ヒューマンサイエンス振興財団 Cell culture composition and method for producing influenza virus
US20150161359A1 (en) 2012-06-04 2015-06-11 Novartis Ag Methods for safety testing
GB201218195D0 (en) 2012-10-10 2012-11-21 Istituto Zooprofilattico Sperimentale Delle Venezie Composition
AU2013354219A1 (en) 2012-12-03 2015-07-02 Novartis Ag Reassortant influenza a viren
CA2905612A1 (en) 2013-03-13 2014-09-18 Novartis Ag Influenza virus reassortment
CN105828835A (en) 2013-05-10 2016-08-03 诺华股份有限公司 Avoiding narcolepsy risk in influenza vaccines
DE202013005100U1 (en) 2013-06-05 2013-08-26 Novartis Ag Influenza virus reassortment
DE202013005130U1 (en) 2013-06-05 2013-09-10 Novartis Ag Influenza virus reassortment
CN105722976A (en) 2013-06-06 2016-06-29 诺华股份有限公司 Influenza virus reassortment
KR101370512B1 (en) 2013-06-07 2014-03-06 재단법인 목암생명공학연구소 Mdck-derived cell lines suspension-cultured in a protein-free medium and method for propagating virus using the same
BE1022132B1 (en) 2013-08-30 2016-02-19 Glaxosmithkline Biologicals S.A. NEW METHOD.
JP6373376B2 (en) 2013-11-15 2018-08-15 ノバルティス アーゲー Removal of residual cell culture impurities
TWI743024B (en) 2014-06-06 2021-10-21 美商健臻公司 Perfusion culturing methods and uses thereof
TWI776235B (en) * 2014-06-09 2022-09-01 美商健臻公司 Seed train processes and uses thereof
AU2015252119A1 (en) 2014-11-07 2016-05-26 Takeda Vaccines, Inc. Hand, foot, and mouth vaccines and methods of manufacture and use thereof
AR102547A1 (en) 2014-11-07 2017-03-08 Takeda Vaccines Inc VACCINES AGAINST DISEASE OF HANDS, FEET AND MOUTH AND MANUFACTURING METHODS AND THEIR USE
US10238739B2 (en) 2014-12-02 2019-03-26 Novartis Ag Manufacture of surfactant-containing compositions with enhanced stability
WO2016207853A2 (en) 2015-06-26 2016-12-29 Seqirus UK Limited Antigenically matched influenza vaccines
AU2016290603B2 (en) 2015-07-07 2022-06-02 Seqirus UK Limited Influenza potency assays
TWI830290B (en) 2015-10-30 2024-01-21 財團法人國家衛生研究院 Mdck suspension cell lines in serum-free, chemically-defined media for vaccine production
US20190078056A1 (en) 2016-03-29 2019-03-14 The Research Foundation For Microbial Diseases Of Osaka University Method for culturing mdck cells
JP2017038622A (en) * 2016-11-21 2017-02-23 ウィスコンシン アルムニ リサーチ ファンデイション H3 equine influenza a virus
EP3703739A2 (en) 2017-11-03 2020-09-09 Takeda Vaccines, Inc. Zika vaccines and immunogenic compositions, and methods of using the same
JP2022533550A (en) 2019-05-08 2022-07-25 タケダ ワクチン,インコーポレイテッド Inactivated virus composition and Zika vaccine formulation
CN115380107A (en) 2019-11-18 2022-11-22 思齐乐私人有限公司 Method for producing reassortant influenza viruses
US20230218526A1 (en) 2020-06-30 2023-07-13 Seqirus UK Limited Cold filtration of oil-in-water emulsion adjuvants
WO2023154043A1 (en) 2022-02-09 2023-08-17 Takeda Vaccines, Inc. Zika vaccines and immunogenic compositions, and methods of using the same

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4064232A (en) * 1974-01-14 1977-12-20 Sandoz Ltd. Process for isolating the immunogenic components of influenza viruses
US4500513A (en) * 1979-05-15 1985-02-19 Miles Laboratories, Inc. Influenza vaccine production in liquid cell culture
US4783411A (en) * 1984-10-22 1988-11-08 Janis Gabliks Influenza-A virus vaccine from fish cell cultures
USRE33164E (en) * 1979-05-15 1990-02-13 Mobay Corporation Influenza vaccine production in liquid cell culture
US5013663A (en) * 1983-06-15 1991-05-07 American Home Products Corporation Canine corona virus vaccine
US5753489A (en) * 1994-11-10 1998-05-19 Immuno Ag Method for producing viruses and vaccines in serum-free culture
US5756341A (en) * 1994-11-10 1998-05-26 Immuno Ag Method for controlling the infectivity of viruses
US5762939A (en) * 1993-09-13 1998-06-09 Mg-Pmc, Llc Method for producing influenza hemagglutinin multivalent vaccines using baculovirus
US5911998A (en) * 1994-11-30 1999-06-15 Dyncorp Method of producing a virus vaccine from an African green monkey kidney cell line
US6455298B1 (en) * 1996-04-01 2002-09-24 Chiron Behring Gmbh & Co. Animal cells and processes for the replication of influenza viruses
US6514502B1 (en) * 1999-01-26 2003-02-04 Schering-Plough Veterinary Corporation Propagation of bovine cononavirus in chinese hamster ovary cells

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1278075B (en) * 1964-06-22 1968-09-19 Norden Lab Inc Use of tissue cultures to grow viruses for the production of vaccines
US3616203A (en) * 1969-12-11 1971-10-26 Norden Lab Inc Virus culture and method
CA1122527A (en) 1979-05-15 1982-04-27 Karen K. Brown Influenza vaccine production in liquid cell culture
EP0389925B1 (en) 1989-03-29 1995-12-27 The Research Foundation Of State University Of New York A method for purifying an outer membrane protein of Haemophilus influenzae
JP2827298B2 (en) * 1989-07-10 1998-11-25 三菱化学株式会社 Cell culture carrier
AT393277B (en) 1990-01-04 1991-09-25 Immuno Ag METHOD FOR PRODUCING EARLY SUMMER MENINGOENZEPHALITIS VIRUS (TBE VIRUS) ANTIGES
JPH0420284A (en) * 1990-05-12 1992-01-23 Mitsubishi Kasei Corp Micro-carrier for cell culture
JPH05207873A (en) * 1992-01-27 1993-08-20 Mitsubishi Kasei Corp Culture method for adhesive animal cell
JP3158157B2 (en) 1994-11-10 2001-04-23 バクスター・アクチエンゲゼルシャフト Biopharmaceutical production in protein-free culture
NZ296861A (en) 1994-11-16 1998-05-27 St Jude Childrens Res Hospital Replicating human influenza virus in a vero cell culture involving maintaining consistent minimum concentration of trypsin in the culture medium
EP0851873A2 (en) 1995-09-22 1998-07-08 Aventis Pasteur Limited Parainfluenza virus glycoproteins and vaccines
DE19612967A1 (en) 1996-04-01 1997-10-02 Behringwerke Ag Process for the propagation of influenza viruses in cell culture, and the influenza viruses obtainable by the process

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4064232A (en) * 1974-01-14 1977-12-20 Sandoz Ltd. Process for isolating the immunogenic components of influenza viruses
US4500513A (en) * 1979-05-15 1985-02-19 Miles Laboratories, Inc. Influenza vaccine production in liquid cell culture
USRE33164E (en) * 1979-05-15 1990-02-13 Mobay Corporation Influenza vaccine production in liquid cell culture
US5013663A (en) * 1983-06-15 1991-05-07 American Home Products Corporation Canine corona virus vaccine
US4783411A (en) * 1984-10-22 1988-11-08 Janis Gabliks Influenza-A virus vaccine from fish cell cultures
US5762939A (en) * 1993-09-13 1998-06-09 Mg-Pmc, Llc Method for producing influenza hemagglutinin multivalent vaccines using baculovirus
US5753489A (en) * 1994-11-10 1998-05-19 Immuno Ag Method for producing viruses and vaccines in serum-free culture
US5756341A (en) * 1994-11-10 1998-05-26 Immuno Ag Method for controlling the infectivity of viruses
US5911998A (en) * 1994-11-30 1999-06-15 Dyncorp Method of producing a virus vaccine from an African green monkey kidney cell line
US6455298B1 (en) * 1996-04-01 2002-09-24 Chiron Behring Gmbh & Co. Animal cells and processes for the replication of influenza viruses
US6656720B2 (en) * 1996-04-01 2003-12-02 Chiron Behring Gmbh & Co. Animal cells and processes for the replication of influenza viruses
US6514502B1 (en) * 1999-01-26 2003-02-04 Schering-Plough Veterinary Corporation Propagation of bovine cononavirus in chinese hamster ovary cells

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9926535B2 (en) 2006-03-31 2018-03-27 Wisconsin Alumni Research Foundation (Warf) High titer recombinant influenza viruses for vaccines
US10119124B2 (en) 2007-06-18 2018-11-06 Wisconsin Alumni Research Foundation (Warf) Influenza M2 protein mutant viruses as live influenza attenuated vaccines
US20100183671A1 (en) * 2007-06-27 2010-07-22 Novartis Vaccines & Diagnostics GmbH & Co., KG Low-additive influenza vaccines
US10808229B2 (en) 2009-10-26 2020-10-20 Wisconsin Alumni Research Foundation (“WARF”) High titer recombinant influenza viruses with enhanced replication in vero cells
US11007262B2 (en) 2010-03-23 2021-05-18 Wisconsin Alumni Research Foundation (Warf) Vaccines comprising mutant attenuated influenza viruses
US10130697B2 (en) 2010-03-23 2018-11-20 Wisconsin Alumni Research Foundation (Warf) Vaccines comprising mutant attenuated influenza viruses
US9950057B2 (en) 2013-07-15 2018-04-24 Wisconsin Alumni Research Foundation (Warf) High titer recombinant influenza viruses with enhanced replication in MDCK or vero cells or eggs
US10172934B2 (en) 2013-07-15 2019-01-08 Wisconsin Alumni Research Foundation (Warf) High titer recombinant influenza viruses with enhanced replication in MDCK or vero cells or eggs
US11046934B2 (en) 2014-06-20 2021-06-29 Wisconsin Alumni Research Foundation (Warf) Mutations that confer genetic stability to additional genes in influenza viruses
US10053671B2 (en) 2014-06-20 2018-08-21 Wisconsin Alumni Research Foundation (Warf) Mutations that confer genetic stability to additional genes in influenza viruses
US11802273B2 (en) 2014-06-20 2023-10-31 Wisconsin Alumni Research Foundation (Warf) Mutations that confer genetic stability to additional genes in influenza viruses
US10633422B2 (en) 2015-06-01 2020-04-28 Wisconsin Alumni Research Foundation (Warf) Influenza virus replication by inhibiting microRNA lec7C binding to influenza viral cRNA and mRNA
US10246686B2 (en) 2015-07-06 2019-04-02 Wisconsin Alumni Research Foundation (Warf) Influenza virus replication for vaccine development
US9890363B2 (en) 2015-07-06 2018-02-13 Wisconsin Alumni Research Foundation (Warf) Influenza virus replication for vaccine development
US11197925B2 (en) 2016-02-19 2021-12-14 Wisconsin Alumni Research Foundation (Warf) Influenza B virus replication for vaccine development
US11241492B2 (en) 2019-01-23 2022-02-08 Wisconsin Alumni Research Foundation (Warf) Mutations that confer genetic stability to genes in influenza viruses
US11851648B2 (en) 2019-02-08 2023-12-26 Wisconsin Alumni Research Foundation (Warf) Humanized cell line
US11390649B2 (en) 2019-05-01 2022-07-19 Wisconsin Alumni Research Foundation (Warf) Influenza virus replication for vaccine development
US11807872B2 (en) 2019-08-27 2023-11-07 Wisconsin Alumni Research Foundation (Warf) Recombinant influenza viruses with stabilized HA for replication in eggs

Also Published As

Publication number Publication date
US20050202553A1 (en) 2005-09-15
JP2000507448A (en) 2000-06-20
US20030073223A1 (en) 2003-04-17
DE19612966B4 (en) 2009-12-10
JP4243349B2 (en) 2009-03-25
JP2012016358A (en) 2012-01-26
JP2006230415A (en) 2006-09-07
EP2275535A1 (en) 2011-01-19
HK1019233A1 (en) 2000-01-28
EP0904351B1 (en) 2010-09-22
JP2009034115A (en) 2009-02-19
DK0904351T3 (en) 2010-11-01
JP2005211080A (en) 2005-08-11
DE19612966A1 (en) 1997-10-02
JP2012005502A (en) 2012-01-12
US6656720B2 (en) 2003-12-02
PT904351E (en) 2010-12-28
ES2352638T3 (en) 2011-02-22
JP2004331673A (en) 2004-11-25
ATE482267T1 (en) 2010-10-15
DE69740002D1 (en) 2010-11-04
WO1997037000A1 (en) 1997-10-09
US6455298B1 (en) 2002-09-24
EP0904351A1 (en) 1999-03-31

Similar Documents

Publication Publication Date Title
US6656720B2 (en) Animal cells and processes for the replication of influenza viruses
EP0891420B1 (en) Processes for the replication of influenza viruses in cell culture, and the influenza viruses obtainable by the process
US10329536B2 (en) Methods for producing an active constituent of a pharmaceutical or a diagnostic agent in an MDCK cell suspension culture
FI121117B (en) Process for producing biological products in protein-free culture
AU2002338666B2 (en) Multiplication of viruses in a cell culture
RU2162710C2 (en) Vaccine against swine and reproductive and respiratory syndrome
CA2250714C (en) Animal cells and processes for the replication of influenza viruses

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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