USRE41974E1 - Method for culturing Chinese hamster ovary cells - Google Patents

Method for culturing Chinese hamster ovary cells Download PDF

Info

Publication number
USRE41974E1
USRE41974E1 US11/640,428 US64042806A USRE41974E US RE41974 E1 USRE41974 E1 US RE41974E1 US 64042806 A US64042806 A US 64042806A US RE41974 E USRE41974 E US RE41974E
Authority
US
United States
Prior art keywords
cho cells
medium
growing
accordance
culturing
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.)
Expired - Lifetime
Application number
US11/640,428
Inventor
Michael John Keen
Nicholas Timothy Rapson
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.)
GlaxoSmithKline LLC
Original Assignee
GlaxoSmithKline LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=10683864&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=USRE41974(E1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by GlaxoSmithKline LLC filed Critical GlaxoSmithKline LLC
Priority to US11/640,428 priority Critical patent/USRE41974E1/en
Assigned to GLAXOSMITHKLINE LLC reassignment GLAXOSMITHKLINE LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: SMITHKLINE BEECHAM CORPORATION
Application granted granted Critical
Publication of USRE41974E1 publication Critical patent/USRE41974E1/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2893Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD52
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0018Culture media for cell or tissue culture
    • C12N5/0043Medium free of human- or animal-derived components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/6456Plasminogen activators
    • C12N9/6459Plasminogen activators t-plasminogen activator (3.4.21.68), i.e. tPA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21069Protein C activated (3.4.21.69)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/34Sugars
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/36Lipids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/70Undefined extracts
    • C12N2500/74Undefined extracts from fungi, e.g. yeasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/70Undefined extracts
    • C12N2500/76Undefined extracts from plants
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/135Platelet-derived growth factor [PDGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/33Insulin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • C12N2501/392Sexual steroids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/395Thyroid hormones

Definitions

  • the present invention relates to a biochemically defined culture medium for culturing Chinese hamster ovary (CHO) cell lines and cells adapted to grow in the culture medium.
  • CHO Chinese hamster ovary
  • CHO Chinese hamster ovary cells
  • Puck J. Exp. Med. 108, 945, 1958
  • CHO-K1 is proline-requiring and is diploid for the dihydrofolate reduotase (dhfr) gene.
  • a dhfr ⁇ CHO cell line (CHO DUK B11) was developed (PNAS 77, 1980, 4216-4220) which is characterised by the loss of dhfr function as a consequence of a mutation in one dhfr gene and the subsequent loss of the other gene. These cells are functionally dhfr ⁇ .
  • Other OHO DUK sub-lines have been derived which are also phenotypically dhfr ⁇ .
  • CHO cells which are dhfr ⁇ cannot grow without nucleotide precursors such as thymidine, hypoxanthine, or the equivalent nucleosides.
  • E. coli XGPRT gene J. Mol. App. Gen. 1981, 1, 165-175
  • human tissue-type plasminogen activator Mol. & Cell Biol. 5, 170-1759, 1985
  • human immune ( ⁇ ) interferon PNAS 80 pp 4654-4658
  • human beta interferon Molecular and Cellular Biology 4, 166-172, 1984.
  • a dhfr ⁇ CHO cell line is transfected with a product gene and a dhfr gene which enables selection of CHO cell transformants of the dhfr + phenotype.
  • Selection is carried out by culturing the colonies in media devoid of thymidine and hypoxanthine, the absence of which prevents untransformed cells from growing.
  • the transformants usually express low levels of the product gene by virtue of co-integration of both transfected genes.
  • the expression levels for the product gene may be increased by amplification using methotrexate.
  • This drug is a direct inhibitor of the dhfr enzyme and allows isolation of resistant colonies which have amplified their dhfr gene copy number sufficiently to survive under these conditions. Since the dhfr and product genes are usually closely linked in the original transformants, there is normally concomitant amplification resulting in increased expression of the desired product gene.
  • GS system glutamine synthetase selectable marker
  • Msx methionine sulphoximine
  • Engineered CHO cells are routinely grown in culture media containing serum, (References: J. Mol. App. Gen. 1981, 1, 165-175; Mol. & Cell Biol. 1985, 5, 1750-1759, 1985 ; PNAS 1983, 80, pp 4654-4658; Molecular and Cellular Biology 1984, 4, 166-172, 1984 ).
  • Fetal bovine serum (FBS) is probably the most extensively utilised serum for mammalian cell culture, although other mammalian sera are used.
  • serum poses a number of problems. Serum is an expensive commodity which is not readily available in amounts required for commercial production.
  • Serum is known to contain many major components including albumin and transferrin and also minor components many of which have not been fully identified nor their action determined, thus serum will differ from batch to batch possibly requiring testing to determine levels of the various components and their effect on the cells. Frequently, serum is contaminated with microorganisms such as viruses and mycoplasma many of which may be harmless but will represent an additional unknown factor. This problem has become more acute in recent years with the emergence of Bovine Spongiform Encephalopathy (BSE). Despite improvements in screening, regulatory authorities are likely to require the sourcing of bovine products from those areas which are free from (BSE) infections.
  • BSE Bovine Spongiform Encephalopathy
  • bovine serum albumin BSA
  • bovine antibody from the medium prior to use is possible but this and the additional product testing required, adds greatly to the everall overall cost of production of the product. Consequently, there has been much research into finding a culture medium devoid of animal components which will support cellular growth, especially of CHO cells. Unfortunately, the problems associated with the provision of such a medium are themselves numerous. CHO cells do not readily grow in serum-free conditions. In addition, the removal of serum may also remove those components that provide cell protection and detoxifying activity.
  • a culture medium which is serum-free but not free from animal components is described by Mendiaz et al (In Vitro Cellular & Development Biology Vol.22, No.2, 1986) for use in the culture of CHO K 1 cells.
  • the medium is a modification of the medium developed by Ham (Microbiology 53 1965 288-293) which is known as “Ham's F12”.
  • Other examples of media have been based on Ham's F12 medium for example as disclosed in EPA390327 and EP325190. These media contain transferrin as the serum substitute, but transferrin is derived from an animal source, so the resulting media do not overcome the contamination problems associated with the use of serum.
  • a further problem which arises with the use of serum-free media is that of supporting recombinant CHO cells to enable growth and expression of product.
  • Media based on Ham's F12 which are not supplemented with serum are generally not rich enough to support full growth or expression.
  • Engineered CHO cells are also difficult to grow in suspension. It is highly desirable to achieve growth in suspension when using the cells to express a product such as an antibody.
  • a product such as an antibody.
  • a suitable medium must be able to support the cells against sheer forces from blade impellers or turbines and from effects of sparging (ie: supplying air, oxygen and CO 2 in bubble form directly to the medium).
  • the present invention therefore provides a biochemically defined culture medium for culturing engineered CHO cells which is essentially free from protein, lipid and carbohydrate isolated from an animal source, comprising water, an osmolality regulator, a buffer, an energy source, amino acids including L-glutamine, an inorganic or recombinant iron source and a recombinant or synthetic growth factor and optionally non-ferrous metal ions, vitamins and cofactors.
  • the components of the medium are mostly inorganic, synthetic or recombinant and as such are not obtained directly from any animal source. Some components may be obtained from a plant or bacterial source. Recombinant components are prepared under highly pure conditions to minimise the risk of contamination from the parent tissue passing to the cells used to produce the components. Further purification steps may be employed to remove cell proteins. Thus, a medium which is essentially free from all protein, lipid and carbohydrate isolated from an animal source, can be achieved.
  • the preferred culture medium of the invention contains no protein, lipid and carbohydrate isolated from an animal source.
  • Osmolality regulators are generally salts. Those which may be used in the medium include NaCl, KCl, KNO 3 .
  • Buffers of use in the medium include carbonates such as NaHCO 3 ; also chlorides, sulphates and phosphates such as CaCl 2 2H 2 O, MgSO 4 7H 2 O, NaH 2 PO 4 2H 2 O, or sodium pyruvate, such buffers are generally present in an amount 50-500 mg/liter.
  • buffers such as N-[2-hydroxyethyl]piperazine-N′-[2-ethanesul-phonic acid] otherwise known as HEPES and 3-[N-Morpholino]-propanesul-fonic acid otherwise known as MOPS are generally present in an amount 1000-10,000 mg/liter.
  • the energy source of use in the medium is generally present in an amount 1000-10,000 mg/liter and is preferably a monosaccharide such as manose, fructose, galactose or maltose most preferably glucose, particularly D-glucose.
  • the non-ferous non-ferrous metal ions optionally of use in the medium include magnesium, copper and zinc; also sodium, potassium and selenium.
  • the ions are generally added to the medium in the form of salts such as chlorides and sulphates sulfates. The amounts are typically similar to those provided in the ISCOVES medium set out in Table 1 but clearly may be varied.
  • Vitamins and enzyme co-factor vitamins (co-factors) optionally of use in the medium include Vitamin B6 (pyridoxine). Vitamin B12 (cyanocobalamin) and Vitamin K (biotin) present in an amount 0.01-0.5 mg/liter; Vitamin C (ascorbic acid) present in an amount 10-30 mg/liter. Vitamin B2 (riboflavin) present in an amount 0.1-1.0 mg/liter and Vitamin B1 (thiamine), nicotin amide. Vitamin B5 (D calcium pentothenate), folic acid, i-inositol generally present in an amount 0.2-8.0 mg/liter.
  • Vitamin B6 pyridoxine
  • Vitamin B12 cyanocobalamin
  • Vitamin K biotin
  • Vitamin C ascorbic acid
  • Vitamin B2 riboflavin
  • Vitamin B1 thiamine
  • Vitamin B5 D calcium pentothenate
  • folic acid i-inositol generally present in an amount 0.2-8.0 mg/liter.
  • lipid factor such as choline chloride, lipoic acid, oleic acid, phosphatidylcholine or methyl lineoleate, generally in an amount 0.05-10 mg/liter.
  • lipid factor such as choline chloride, lipoic acid, oleic acid, phosphatidylcholine or methyl lineoleate
  • Compounds involved in lipid production for example alcoholamines such as ethanolamine may also be added.
  • Amino Acid Preferred mg/liter L-Alanine 20-50 L-Arginine (HCl) 50-100 L-Asparagine (H 2 O) 20-50 L-Aspartic Acid 20-50 L-Cystine (disodium salt) 50-100 L-Glutamic acid 50-100 L-Glutamine 400-600 Glycine 20-50 L-Histidine (HCl•H 2 O) 30-60 L-Isoleucine 50-150 L-Leucine 50-150 L-Lysine (HCl) 100-200 L-Methionine 20-50 L-Phenylalanine 40-80 L-Proline 30-60 L-Serine 30-60 L-Threonine 50-120 L-Tryptophan 10-20 L-Tyrosine (disodium salt) 50-120 L-Valine 80-120 The bracketed forms are preferred.
  • the amino acids are preferably of synthetic origin.
  • the amounts which are usually included vary for each amino acid but are generally in the range 10-150 mg/ml.
  • L-glutamine is generally present at much higher concentration preferably in the range 400-600 mg/ml mg/liter .
  • a pH indicator for example Phenol red sodium salt for example at 5-50 mg/liter.
  • Medium A as set out in Table 1, is an example of a medium which provides the preferred quantities of water, osmolality regulator, buffer, energy source, amino acids, non-ferrous metal ions, vitamins and co-factors as a basis for a culture medium according to the invention.
  • This medium does not contain any hypoxanthine or thymidine and is commercially available from GIBCO Ltd., Unit 4, Cowley Mill Td. Est., Uxbridge UB8 2YG. It is similar to a published culture medium (Iscoves and Melcher (1978) J. Exp. Med. 1, 47,923) but does not contain any bovine serum albumin, pure human transferrin or soyabean lecithin.
  • selenium (optionally in the form of sodium selenite) generally in an amount 0.01-0.2 mg/liter or L-Ascorbic acid generally in an amount 20-50 mg/liter to help minimise the potential toxic effects of ferrous or ferric ions, and oxygen.
  • chelating agents such as citrate or Ethylenediaminetetraacetic acid (EDTA) or a free radical scavenger such as ⁇ -Tocepherol (vitamin E) are advantageous in reducing free radical damage.
  • Antibiotics such as polymyxin, neomycin, penicillin or streptomycin may be added to-the medium to prevent bacterial contamination. These are usually included in an amount 10,000-100,000 Iu/liter.
  • Growth factors which may be added to the basal medium are synthetic or recombinant and include insulin.
  • Other factors such as platelet-derived growth factor (PDGF), thyroxtne thyroxine T 3 , thrombin, interleukins such as IL2 and IL6, progesterone, hydrocortisone and vitamin E may be included.
  • Folic acid, vitamin B6 and vitamin B12 which are involved in the folate pathway may be added to enhance the growth of cells.
  • the peptide hormone insulin (which in the present context includes analogues thereof such as Nucellin® (recombinant insulin, Eli Lilly) is advantageously obtained by recombinant DNA techniques but is not isolated from an animal source. It is preferably added to the medium in an amount 5 ⁇ g-5 mg/liter. Nucellin is the preferred form of insulin for use in the invention.
  • the non-animal derived iron source to supplement the medium is preferably inorganic and present in an amount 0.25-5 mg/liter.
  • examples include ferric and ferrous salts such as ferric citrate or ferrous sulphate.
  • the chelated salts such as ferric citrate and ferric ammonium citrate are preferred.
  • any iron source may be used which is not isolated from an animal source, for example, chemical iron chelators or recombinant protein iron carriers.
  • the concentration of ferric or ferrous ions should be carefully controlled as these may help generate superoxides and free radicals in the medium, which may damage not only the cells themselves, but medium components and the desired end product.
  • putrescine advantageously as a salt such as HCl, which is known to play a role in maintaining the structure of the endoplasmic reticulum and to be required by certain CHO cell lines to support growth.
  • Putrescine or a salt thereof is preferably added in an amount 0.01-1.0 mg/liter.
  • Serum-free media disclosed to date contain hypoxanthine or thymidine. This could bypass the selection pressure placed on the dhfr selection and amplification system as previously disclosed. The result may be loss of genetic material specifying the product and the dhfr genes. Therefore, in another aspect of the invention there is provided a culture medium for the growth of engineered dhfr ⁇ CHO cells in accordance with the invention, essentially free from hypoxanthine and/or thymidine.
  • the culture medium of the present invention supports CHO cell growth and when supplemented with an appropriate agent such as methotrexate for the dhfr system usually in an amount 0.1-5.0 ⁇ M, (or MSX for the GS system), allow full selection pressure to be exerted on the cells.
  • an appropriate agent such as methotrexate for the dhfr system usually in an amount 0.1-5.0 ⁇ M, (or MSX for the GS system)
  • hypoxanthine and thymidine at concentrations which are insufficient to bypass selection of the dhfr system may be present in the medium, but the presence of these two nucleotide precursors is not preferred for use with the present invention.
  • mammalian cells are particularly susceptible to sheer forces arising from the sparging of the vessel with gases and the mixing with the impeller.
  • a cell protectant such as polyethylene glycol, polyvinyl alcohols or pluronic polyols.
  • Pluronic® polyol, BASF Wyandotte Corp.
  • polyol F68 is preferred since unlike polyvinyl alcohols this is a non-toxic substance and unlike polyethylene glycols does not interfere with downstream purification.
  • a peptide digest such as Tryprone, casein hydrolysate, yeast extract, or preferably papain digested soya peptone.
  • the preferred amounts are 1%-0.025% w/v, most preferably 0.25% w/v.
  • the media of the invention for culturing recombinant CHO cells are capable of supporting the growth and secretion of product from such cells in suspension in small and large scale fermenters fermentors, static cultures and/or spinners.
  • the culture medium according to the invention is also capable of supporting growth of cells at high cell density namely greater than 1 ⁇ 10 5 cells/ml up to or greater than 1.5 ⁇ 10 6 cells/ml and product secretion of 30 mg/l up to greater than 150 mg/l.
  • the medium according to the invention is also capable of supporting this growth and product secretion over multiple passages lasting upto or greater than 6 months.
  • the medium is preferred for the production of all types of antibodies natural and altered.
  • the invention therefore includes production of human antibodies wherein the amino acid sequences of the heavy and light chains are homologous with those sequences of antibodies produced by human lymphocytes in vivo or in vitro by hybridomas.
  • hybrid antibodies in which the heavy and light chains are homologous to a natural antibody but are combined in a way that would not occur naturally.
  • a bispecific antibody has antigen binding sites specific to more than one antigen.
  • the constant region of the antibody may relate to one or other of the antigen binding regions or may be from a further antibody.
  • Altered antibodies, for example chimaeric antibodies have variable regions from one antibody and constant regions from another.
  • chimaeric antibodies may be species/species chimaeras or class/class chimaeras. Such chimaeric antibodies may have one or more further modifications to improve antigen binding ability or to alter effector functioning.
  • Humanised Humanized or CDR-grafted antibodies (EP 239400) are embraced within the invention, in particular Campath 1H (EP328404) (Campath is a TM of The Wellcome Foundation) also composite antibodies, wherein parts of the hypervariable regions in addition to the CDRs are tranferred transferred to the human framework. Additional amino acids in the framework or constant regions of such antibodies may be altered.
  • the invention further includes the production of Feb Fab fragments which are roughly equivalent to the Y branch portions of the heavy and light chains; this includes incomplete fragments or fragments including part of the Fc region.
  • an engineered CHO cell adapted to grow in a medium according to the invention.
  • a CHO cell engineered to express proteins such as tissue plasminogen activator or antibodies as defined above.
  • the invention provides a dhfr- CHO cell line transfected with a gene encoding a biologically active protein and a dhfr selectable marker gene, adapted to grow in a culture medium according to the invention.
  • the protein is preferably an antibody as defined above.
  • the ingredients of the culture medium may be added in any order but it is preferable to add the iron source and when used, tyrosine, last to avoid precipitation.
  • FIG. 1 shows growth of C1H 3D11* 44 in WCM5 (protein-free medium) in a 1 liter fermenterfermentor measured as cell count/ml over 90 days.
  • FIG. 2 shows antibody production from C1H 3D11* 44 cells in WCM5 in a 1 liter formenterfermentor measured as micrograms of antibody/ml over 80 days.
  • This medium does not contain hypoxanthine, thymidine or folinic acid which can bypass methotrexate selection.
  • the medium does contain glycine which cannot by itself bypass selection. Therefore, this medium maintains full selection for methotrexate resistance.
  • MedmiumMedium A (Iscoves modification of DMEM without BSA, transferrin or lecithin).
  • C1H 3D11* cells are genetically engineered CHO DUK B11 cells (Urlaub and Chasin PNAS ( 1980,) PNAS 77, 7 pp 4216-4220).
  • CHO DUK B11 cells cannot produce dihydrofolate reductase (dhfr).
  • dhfr dihydrofolate reductase
  • These cells were engineered to produce a humanised humanized IgG antibody, Campath 1H (Winter et al., Nature, 1988, 322, 323-327), using plasmid constructs to express heavy and light antibody chains and the mouse dhfr. Expression is amplified and maintained using the folate antagonist methotrate methotrexate.
  • H hypoxanthine
  • T thymidine
  • HT methotrexate
  • the flasks were pooled and added to an equal volume of WCM4+MTX without peptone or PEG, and were transferred to a 75 cm 2 flask.
  • these cells produced antibody in excess of 70 ⁇ g/ml and regularly achieved levels of 100 ⁇ g/ml or more.
  • the cells are denoted C1H 3D11* 44.
  • C1H 3D11*44 cells from Example 3 which had been growing serum-free for over 2 months were transferred to a SGi 1 liter fermenter fermentor with a stainless steel angled paddle turning at 70 rpm.
  • the temperature was set at 37° C., dO 2 at 10% and pH control to 7-7.2.
  • the fermenter fermentor was seeded on day 0 with 0.22 ⁇ 10 6 cells/ml in WCM4 (Example 1) with 0.1% polyethylene glycol (PEG) 10,000 and 0.25% soy peptone, and was top gassed with O 2 .
  • the cells were routinely passaged using fresh medium and a split rate typically between 1 to 2 and 1 to 4.
  • WCM5 (Example 2) was used together with peptone and PEG instead of WCM4.
  • Chinese hamster ovary cells CHO AJ19 MCB1, derived from CHO DUK cells. (Urlaub & Chasin PNAS, 1980, 77, 7, pp 4216-4220, 1980 ), were genetically engineered to produce tPA under methotrexate selection.
  • This cell line had been routinely grown in a fermenter fermentor as a suspension culture using normal growth medium consisting of RPMI 1640 medium (GIBCO), 2.5% acid hydrolysed hydrolyzed adult bovine serum (Imperial), 0.5% Tryptone, 50 IU/ml polymycin, 20 IU/ml neomycin, 500 nM methotrexate (MTX).
  • the yeast extract, Peptone and PEG were made up as 10% w/v solutions with water (Wellcome media production unit) and filtered through a 0.2 um disposable filter (Gelman, Supor Vac), then diluted for use.
  • the cells were incubated at 37° C. in a humidified incubator containing 5% CO 2 .
  • CHO AJ19 MCBI in WCM4 cells growing in normal growth medium were pelleted and washed as in Example 5 and were resuspended at 7 ⁇ 10 4 /ml in 500 ml of medium 46B. These cells were transferred to a Techne spinner flask and incubated, as above, stirring at 40 rpm. At various time intervals the cells were counted and subcultured using the same medium. A sample was taken for tPA assay and treated as in Example 5.

Abstract

A biochemically defined culture medium for culturing engineered Chinese hamster ovary (CHO) cell lines, which is essentially free from protein, lipid and carbohydrate isolated from an animal source, having water, an osmolality regulator, a buffer, an energy source, amino acids including L-glutamine, an inorganic or recombinant iron source, and a synthetic or recombinant growth factor, and optionally non-ferrous metal ions vitamins and cofactors. Also cells adapted to grow in such a culture medium.

Description

This is a continuation of application Ser. No. 07,991,717 filed Dec. 18, 1992, now U.S. Pat. No. 5,316,938 which is a continuation of Ser. No. 07/777,729, filed Oct. 16, 1991, now abandoned.Notice: more than one reissue application has been filed for reissue of U.S. Pat. No. 5,633,162. The reissue applications are application Ser. No. 11/640,428 (the present application), filed on 15 Dec. 2006; and application Ser. No. 10/995,010, filed on 22 Nov. 2004, now RE 39.792.
The present reissue application is a continuation of application Ser. No. 10/995,010, now Reissue No. RE 39,792, filed 22 Nov. 2004, which is a reissue application of U.S. Pat. No. 5,633,162, U.S. Pat. No. 5,633,162, filed as application Ser. No. 08/205,379 on 4 Mar. 1994, is a continuation of U.S. Ser. No. 07/991,717 filed 18 Dec. 1992, now U.S. Pat. No. 5,316,938, which is a continuation of U.S. Ser. No. 07/777,729 filed 16 Oct. 1991, now abandoned.
The present invention relates to a biochemically defined culture medium for culturing Chinese hamster ovary (CHO) cell lines and cells adapted to grow in the culture medium.
Chinese hamster ovary cells (CHO) were first cultured by Puck (J. Exp. Med. 108, 945, 1958) from a biopsy of an ovary from a female Chinese hamster. From these original cells various workers have cloned a number of sub-lines with various deficiencies, one of which, CHO-K1, is proline-requiring and is diploid for the dihydrofolate reduotase (dhfr) gene. From this cell line a dhfrCHO cell line (CHO DUK B11) was developed (PNAS 77, 1980, 4216-4220) which is characterised by the loss of dhfr function as a consequence of a mutation in one dhfr gene and the subsequent loss of the other gene. These cells are functionally dhfr. Other OHO DUK sub-lines have been derived which are also phenotypically dhfr. CHO cells which are dhfrcannot grow without nucleotide precursors such as thymidine, hypoxanthine, or the equivalent nucleosides.
Various proteins have been expressed in such CHO cells including E. coli XGPRT gene (J. Mol. App. Gen. 1981, 1, 165-175), human tissue-type plasminogen activator (Mol. & Cell Biol. 5, 170-1759, 1985), human immune (γ) interferon (PNAS 80 pp 4654-4658), and human beta interferon (Molecular and Cellular Biology 4, 166-172, 1984). A dhfrCHO cell line is transfected with a product gene and a dhfr gene which enables selection of CHO cell transformants of the dhfr+ phenotype. Selection is carried out by culturing the colonies in media devoid of thymidine and hypoxanthine, the absence of which prevents untransformed cells from growing. The transformants usually express low levels of the product gene by virtue of co-integration of both transfected genes. The expression levels for the product gene may be increased by amplification using methotrexate. This drug is a direct inhibitor of the dhfr enzyme and allows isolation of resistant colonies which have amplified their dhfr gene copy number sufficiently to survive under these conditions. Since the dhfr and product genes are usually closely linked in the original transformants, there is normally concomitant amplification resulting in increased expression of the desired product gene.
A different system of selection and amplification is provided by the glutamine synthetase selectable marker (or GS system) which is described in WO87/04462. CHO cells which have been successfully transfected with the gene encoding the GS enzyme and the desired antibody gene can be selected by culturing colonies in media devoid of glutamine and amplifying by the addition of methionine sulphoximine (Msx) as described in PCT published application number WO87/04462.
Engineered CHO cells (those in which a CHO cell line is transfected with a product gene and a selectable marker gene) are routinely grown in culture media containing serum, (References: J. Mol. App. Gen. 1981, 1, 165-175; Mol. & Cell Biol. 1985, 5, 1750-1759, 1985 ; PNAS 1983, 80, pp 4654-4658; Molecular and Cellular Biology 1984, 4, 166-172, 1984 ). Fetal bovine serum (FBS) is probably the most extensively utilised serum for mammalian cell culture, although other mammalian sera are used. However, the use of serum poses a number of problems. Serum is an expensive commodity which is not readily available in amounts required for commercial production. It is also a biochemically undefined material. Serum is known to contain many major components including albumin and transferrin and also minor components many of which have not been fully identified nor their action determined, thus serum will differ from batch to batch possibly requiring testing to determine levels of the various components and their effect on the cells. Frequently, serum is contaminated with microorganisms such as viruses and mycoplasma many of which may be harmless but will represent an additional unknown factor. This problem has become more acute in recent years with the emergence of Bovine Spongiform Encephalopathy (BSE). Despite improvements in screening, regulatory authorities are likely to require the sourcing of bovine products from those areas which are free from (BSE) infections. Furthermore, the presence of animal proteins in culture media can require lengthy purification procedures, in particular the presence of bovine antibodies in bovine serum albumin (BSA) makes purification of the desired antibodies expressed by the recombinant CHO cell line, extremely difficult. Removal of bovine antibody from the medium prior to use is possible but this and the additional product testing required, adds greatly to the everall overall cost of production of the product. Consequently, there has been much research into finding a culture medium devoid of animal components which will support cellular growth, especially of CHO cells. Unfortunately, the problems associated with the provision of such a medium are themselves numerous. CHO cells do not readily grow in serum-free conditions. In addition, the removal of serum may also remove those components that provide cell protection and detoxifying activity.
A culture medium which is serum-free but not free from animal components is described by Mendiaz et al (In Vitro Cellular & Development Biology Vol.22, No.2, 1986) for use in the culture of CHO K1 cells. The medium is a modification of the medium developed by Ham (Microbiology 53 1965 288-293) which is known as “Ham's F12”. Other examples of media have been based on Ham's F12 medium for example as disclosed in EPA390327 and EP325190. These media contain transferrin as the serum substitute, but transferrin is derived from an animal source, so the resulting media do not overcome the contamination problems associated with the use of serum.
A further problem which arises with the use of serum-free media is that of supporting recombinant CHO cells to enable growth and expression of product. Media based on Ham's F12 which are not supplemented with serum are generally not rich enough to support full growth or expression.
Engineered CHO cells are also difficult to grow in suspension. It is highly desirable to achieve growth in suspension when using the cells to express a product such as an antibody. For production of a biological protein on a commercial scale it is preferable to be able to support growth in fermenters fermentors which range from 1 liter glass vessels to multi-thousand liter stainless steel tanks. A suitable medium must be able to support the cells against sheer forces from blade impellers or turbines and from effects of sparging (ie: supplying air, oxygen and CO2 in bubble form directly to the medium).
The present invention therefore provides a biochemically defined culture medium for culturing engineered CHO cells which is essentially free from protein, lipid and carbohydrate isolated from an animal source, comprising water, an osmolality regulator, a buffer, an energy source, amino acids including L-glutamine, an inorganic or recombinant iron source and a recombinant or synthetic growth factor and optionally non-ferrous metal ions, vitamins and cofactors.
The components of the medium are mostly inorganic, synthetic or recombinant and as such are not obtained directly from any animal source. Some components may be obtained from a plant or bacterial source. Recombinant components are prepared under highly pure conditions to minimise the risk of contamination from the parent tissue passing to the cells used to produce the components. Further purification steps may be employed to remove cell proteins. Thus, a medium which is essentially free from all protein, lipid and carbohydrate isolated from an animal source, can be achieved. The preferred culture medium of the invention contains no protein, lipid and carbohydrate isolated from an animal source.
It is advantageous to maintain osmolality in the range 200-30 milli-Osmols (mOsm) preferably in the range 290-350 mOsm. Osmolality regulators are generally salts. Those which may be used in the medium include NaCl, KCl, KNO3.
Buffers of use in the medium to maintain the pH in the range 6.5-7.5 most preferably around pH 7.0. Buffers of use in the medium include carbonates such as NaHCO3; also chlorides, sulphates and phosphates such as CaCl22H2O, MgSO47H2O, NaH2PO42H2O, or sodium pyruvate, such buffers are generally present in an amount 50-500 mg/liter. Other buffers, such as N-[2-hydroxyethyl]piperazine-N′-[2-ethanesul-phonic acid] otherwise known as HEPES and 3-[N-Morpholino]-propanesul-fonic acid otherwise known as MOPS are generally present in an amount 1000-10,000 mg/liter.
The energy source of use in the medium is generally present in an amount 1000-10,000 mg/liter and is preferably a monosaccharide such as manose, fructose, galactose or maltose most preferably glucose, particularly D-glucose.
The non-ferous non-ferrous metal ions optionally of use in the medium include magnesium, copper and zinc; also sodium, potassium and selenium. The ions are generally added to the medium in the form of salts such as chlorides and sulphates sulfates. The amounts are typically similar to those provided in the ISCOVES medium set out in Table 1 but clearly may be varied.
Vitamins and enzyme co-factor vitamins (co-factors) optionally of use in the medium include Vitamin B6 (pyridoxine). Vitamin B12 (cyanocobalamin) and Vitamin K (biotin) present in an amount 0.01-0.5 mg/liter; Vitamin C (ascorbic acid) present in an amount 10-30 mg/liter. Vitamin B2 (riboflavin) present in an amount 0.1-1.0 mg/liter and Vitamin B1 (thiamine), nicotin amide. Vitamin B5 (D calcium pentothenate), folic acid, i-inositol generally present in an amount 0.2-8.0 mg/liter.
It is preferable to include in the basal medium a lipid factor such as choline chloride, lipoic acid, oleic acid, phosphatidylcholine or methyl lineoleate, generally in an amount 0.05-10 mg/liter. Compounds involved in lipid production for example alcoholamines such as ethanolamine may also be added.
It is preferable to include additional amino acids in the medium selected from:
Amino Acid Preferred mg/liter
L-Alanine 20-50
L-Arginine (HCl)  50-100
L-Asparagine (H2O) 20-50
L-Aspartic Acid 20-50
L-Cystine (disodium salt)  50-100
L-Glutamic acid  50-100
L-Glutamine 400-600
Glycine 20-50
L-Histidine (HCl•H2O) 30-60
L-Isoleucine  50-150
L-Leucine  50-150
L-Lysine (HCl) 100-200
L-Methionine 20-50
L-Phenylalanine 40-80
L-Proline 30-60
L-Serine 30-60
L-Threonine  50-120
L-Tryptophan 10-20
L-Tyrosine (disodium salt)  50-120
L-Valine  80-120

The bracketed forms are preferred.
The amino acids are preferably of synthetic origin. The amounts which are usually included vary for each amino acid but are generally in the range 10-150 mg/ml. However, L-glutamine is generally present at much higher concentration preferably in the range 400-600 mg/ml mg/liter.
It may be advantageous to include in the medium a pH indicator for example Phenol red sodium salt for example at 5-50 mg/liter.
Medium A as set out in Table 1, is an example of a medium which provides the preferred quantities of water, osmolality regulator, buffer, energy source, amino acids, non-ferrous metal ions, vitamins and co-factors as a basis for a culture medium according to the invention. This medium does not contain any hypoxanthine or thymidine and is commercially available from GIBCO Ltd., Unit 4, Cowley Mill Td. Est., Uxbridge UB8 2YG. It is similar to a published culture medium (Iscoves and Melcher (1978) J. Exp. Med. 1, 47,923) but does not contain any bovine serum albumin, pure human transferrin or soyabean lecithin.
TABLE 1
Medium A (modification of Iscoves' DMEM lacking albumin,
transferrin and lecithin)
Ingredient mg/liter
L-Alanine 25.00
L-Arginine HCl 84.00
L-Asparagine H2O 28.40
L-Aspartic Acid 30.00
L-Cystine 70.00
L-Glutamic acid 75.00
L-Glutamine 584.00
Glycine 30.00
L-Histidine HCl•H2O 42.00
L-Isoleucine 105.00
L-Leucine 105.00
L-Lysine HCl 146.00
L-Methionine 30.00
L-Phenylalanine 66.00
L-Proline 40.00
L-Serine 42.00
L-Threonine 95.00
L-Tryptophan 16.00
L-Tyrosine disodium salt 104.00
L-Valine 94.00
Biotin 0.013
D.Calcium Pantothenate 4.00
Choline chloride 4.00
Folic acid 4.00
i-Inositol 7.20
Nicotinamide 4.00
Pyridoxal HCl 4.00
Riboflavin 0.40
Thiamin HCl 4.00
Vitamin B 12 0.013
CaCl22H2O 219.00
KCl 330.00
KNO3 0.076
MgSO47H2O 200.00
NaCl 4505.00
NaHCO3 3024.00
NaH2PO42H2O 141.30
D-Glucose 4500.00
HEPES 5958.00
Phenol red sodium salt 15.00
Sodium pyruvate 110.00
Sodium selenite 0.017
DMEM modification of Iscoves N and Melcher (1978), J. Exp. Med. 1, 47, 923.
It is preferable to add to the medium, selenium (optionally in the form of sodium selenite) generally in an amount 0.01-0.2 mg/liter or L-Ascorbic acid generally in an amount 20-50 mg/liter to help minimise the potential toxic effects of ferrous or ferric ions, and oxygen. Further use of chelating agents such as citrate or Ethylenediaminetetraacetic acid (EDTA) or a free radical scavenger such as α-Tocepherol (vitamin E) are advantageous in reducing free radical damage.
Antibiotics such as polymyxin, neomycin, penicillin or streptomycin may be added to-the medium to prevent bacterial contamination. These are usually included in an amount 10,000-100,000 Iu/liter.
Growth factors which may be added to the basal medium are synthetic or recombinant and include insulin. Other factors such as platelet-derived growth factor (PDGF), thyroxtne thyroxine T3, thrombin, interleukins such as IL2 and IL6, progesterone, hydrocortisone and vitamin E may be included. Folic acid, vitamin B6 and vitamin B12 which are involved in the folate pathway may be added to enhance the growth of cells.
The peptide hormone insulin (which in the present context includes analogues thereof such as Nucellin® (recombinant insulin, Eli Lilly) is advantageously obtained by recombinant DNA techniques but is not isolated from an animal source. It is preferably added to the medium in an amount 5 μg-5 mg/liter. Nucellin is the preferred form of insulin for use in the invention.
The non-animal derived iron source to supplement the medium, is preferably inorganic and present in an amount 0.25-5 mg/liter. Examples include ferric and ferrous salts such as ferric citrate or ferrous sulphate. The chelated salts such as ferric citrate and ferric ammonium citrate are preferred. However, any iron source may be used which is not isolated from an animal source, for example, chemical iron chelators or recombinant protein iron carriers.
The concentration of ferric or ferrous ions should be carefully controlled as these may help generate superoxides and free radicals in the medium, which may damage not only the cells themselves, but medium components and the desired end product.
It is also preferable to add to the medium, a compound such as putrescine, advantageously as a salt such as HCl, which is known to play a role in maintaining the structure of the endoplasmic reticulum and to be required by certain CHO cell lines to support growth. Putrescine or a salt thereof is preferably added in an amount 0.01-1.0 mg/liter.
Serum-free media disclosed to date contain hypoxanthine or thymidine. This could bypass the selection pressure placed on the dhfr selection and amplification system as previously disclosed. The result may be loss of genetic material specifying the product and the dhfr genes. Therefore, In another aspect of the invention there is provided a culture medium for the growth of engineered dhfrCHO cells in accordance with the invention, essentially free from hypoxanthine and/or thymidine.
The culture medium of the present invention supports CHO cell growth and when supplemented with an appropriate agent such as methotrexate for the dhfr system usually in an amount 0.1-5.0 μM, (or MSX for the GS system), allow full selection pressure to be exerted on the cells. It will be understood that hypoxanthine and thymidine at concentrations which are insufficient to bypass selection of the dhfr system may be present in the medium, but the presence of these two nucleotide precursors is not preferred for use with the present invention.
In large scale fermenters fermentors, mammalian cells are particularly susceptible to sheer forces arising from the sparging of the vessel with gases and the mixing with the impeller. To minimise minimize the occurrence of cellular damage it is advantageous for the medium to contain a cell protectant such as polyethylene glycol, polyvinyl alcohols or pluronic polyols. Of these, Pluronic® (polyol, BASF Wyandotte Corp.) polyol F68 is preferred since unlike polyvinyl alcohols this is a non-toxic substance and unlike polyethylene glycols does not interfere with downstream purification.
Further improvements in CHO cell growth may be obtained by supplementing the medium with a peptide digest, hydrolysates or extracts, such as Tryprone, casein hydrolysate, yeast extract, or preferably papain digested soya peptone. The preferred amounts are 1%-0.025% w/v, most preferably 0.25% w/v.
The media of the invention for culturing recombinant CHO cells are capable of supporting the growth and secretion of product from such cells in suspension in small and large scale fermenters fermentors, static cultures and/or spinners. The culture medium according to the invention is also capable of supporting growth of cells at high cell density namely greater than 1×105 cells/ml up to or greater than 1.5×106 cells/ml and product secretion of 30 mg/l up to greater than 150 mg/l. The medium according to the invention is also capable of supporting this growth and product secretion over multiple passages lasting upto or greater than 6 months.
The medium is preferred for the production of all types of antibodies natural and altered. The invention therefore includes production of human antibodies wherein the amino acid sequences of the heavy and light chains are homologous with those sequences of antibodies produced by human lymphocytes in vivo or in vitro by hybridomas. Also provided are hybrid antibodies in which the heavy and light chains are homologous to a natural antibody but are combined in a way that would not occur naturally. For example, a bispecific antibody has antigen binding sites specific to more than one antigen. The constant region of the antibody may relate to one or other of the antigen binding regions or may be from a further antibody. Altered antibodies, for example chimaeric antibodies have variable regions from one antibody and constant regions from another. Thus, chimaeric antibodies may be species/species chimaeras or class/class chimaeras. Such chimaeric antibodies may have one or more further modifications to improve antigen binding ability or to alter effector functioning. Humanised Humanized or CDR-grafted antibodies (EP 239400) are embraced within the invention, in particular Campath 1H (EP328404) (Campath is a TM of The Wellcome Foundation) also composite antibodies, wherein parts of the hypervariable regions in addition to the CDRs are tranferred transferred to the human framework. Additional amino acids in the framework or constant regions of such antibodies may be altered. The invention further includes the production of Feb Fab fragments which are roughly equivalent to the Y branch portions of the heavy and light chains; this includes incomplete fragments or fragments including part of the Fc region.
In a further aspect of the invention there is provided an engineered CHO cell adapted to grow in a medium according to the invention. In particular a CHO cell engineered to express proteins such as tissue plasminogen activator or antibodies as defined above. In particular the invention provides a dhfr- CHO cell line transfected with a gene encoding a biologically active protein and a dhfr selectable marker gene, adapted to grow in a culture medium according to the invention. The protein is preferably an antibody as defined above.
The ingredients of the culture medium may be added in any order but it is preferable to add the iron source and when used, tyrosine, last to avoid precipitation.
Accompanying Figures are for illustration only.
FIG. 1 shows growth of C1H 3D11* 44 in WCM5 (protein-free medium) in a 1 liter fermenterfermentor measured as cell count/ml over 90 days.
FIG. 2 shows antibody production from C1H 3D11* 44 cells in WCM5 in a 1 liter formenterfermentor measured as micrograms of antibody/ml over 80 days.
EXAMPLE 1
Formulation for medium WCM4.
Medium A: (Iscoves modification of DMEM without BSA, transferrin and lecithin as set out in Table 1).
5 ml/liter+5 ml/liter 200 mM L glutamine
+50 mg/liter L proline
+50 mg/liter L threonine
+50 mg/liter L methionine
+50 mg/liter L cysteine
+50 mg/liter L tyrosine
+25 mg/liter ascorbic acid
+0.062 mg · liter vitamin B6
+1.36 mg · liter vitamin B12
+0.2 mg/liter lipoic acid
+0.088 mg/liter methyl linoleate
+1 mg · lit methotrexate
+1 IU/liter FeSO4
+1 IU/liter ZnSO4
+0.0025 μl/liter CuSO4
+5 mg/liter recombinant insulin (Nucellin)
+50,000 Iu/liter polymyxin
+20,000 Iu/liter neomycin
+0.16 mg/liter putrescine-2 HCL
This medium does not contain hypoxanthine, thymidine or folinic acid which can bypass methotrexate selection. The medium does contain glycine which cannot by itself bypass selection. Therefore, this medium maintains full selection for methotrexate resistance.
EXAMPLE 2
Formulation for Medium WGM5 WCM5
MedmiumMedium A: (Iscoves modification of DMEM without BSA, transferrin or lecithin).
+ 5 ml/liter 200 mM L glutamine
+ 50 mg/liter L proline
+ 50 mg/liter L threonine
+ 50 mg/liter L methionine
+ 50 mg/liter L cysteine
+ 50 mg/liter L tyrosine
+ 25 mg/liter L ascorbic acid
+ 0.062 mg · liter Vitamin B6
+ 1.36 mg · liter Vitamin B12
+ 2 mg/liter Ferric citrate
+ 1 mg/liter Zinc sulphate
+ 0.0025 mg · lit Copper sulphate
+ 50,000 IU/liter Polymyxin
+ 20,000 IU/liter Neomycin
+ 3 μl/liter Ethanolamine
+ 0.16 mg/liter Putrescine
+ 5 mg/liter Recombinant Insulin (Nucellin ®)
EXAMPLE 3
Growth of and Production from C1H 3D11* 44 in WCM4
C1H 3D11* cells are genetically engineered CHO DUK B11 cells (Urlaub and Chasin PNAS ( 1980,) PNAS 77, 7 pp 4216-4220). CHO DUK B11 cells cannot produce dihydrofolate reductase (dhfr). These cells were engineered to produce a humanised humanized IgG antibody, Campath 1H (Winter et al., Nature, 1988, 322, 323-327), using plasmid constructs to express heavy and light antibody chains and the mouse dhfr. Expression is amplified and maintained using the folate antagonist methotrate methotrexate. C1H 3D11* cells growing as a mono-layer in Isover+10% FBS Flow, non-essential amino acids, 10−6M Methotrexate and antibiotics were approximately 90% confluent. These cells were removed from the plastic with trypsin/versene, washed in Iscoves medium without supplements, centrifuged and resuspended at 5×104/ml in WCM4 medium+0.25% peptone+0.1% polyethylene glycol (PEG) 10,000+0.5% fetal bovine serum (FBS) without methotrexate (MTX). Three 25 cm 2 cm 3 flasks were set up with 10 ml of cell suspension+hypoxanthine (H), thymidine (T) or HT. These flasks were incubated at 36.5° C. in 5% CO2 incubator.
After six days, the flasks were pooled and added to an equal volume of WCM4+MTX without peptone or PEG, and were transferred to a 75 cm2 flask.
These cells were used to seed a 500 ml Techner spinner, incubated at 36.5° C. spinning at 40 rpm. Cells continued growing serum free for a period of over five months and although it was-found that the cells needed a period of adaptation, the growth rate and viability steadily improved. The population doubling time was calculated to be 73.1 hours over approximately 7 weeks; this decreased to 47.4 hours over the subsequent 20 days then stabilised. Antibody secretion remained high at levels in excess of 60 μg/ml. It was determined that the gene copy number in these cells did not decrease according to band intensity using Northern blot analysis.
In fermenters fermentors, these cells produced antibody in excess of 70 μg/ml and regularly achieved levels of 100 μg/ml or more. The cells are denoted C1H 3D11* 44.
EXAMPLE 4
Growth and Production of CIH C1H 3D11* 44 in WCM5 in a 1 liter fermenter fermentor.
C1H 3D11*44 cells from Example 3 which had been growing serum-free for over 2 months were transferred to a SGi 1 liter fermenter fermentor with a stainless steel angled paddle turning at 70 rpm. The temperature was set at 37° C., dO2 at 10% and pH control to 7-7.2. The fermenter fermentor was seeded on day 0 with 0.22×106 cells/ml in WCM4 (Example 1) with 0.1% polyethylene glycol (PEG) 10,000 and 0.25% soy peptone, and was top gassed with O2.The cells were routinely passaged using fresh medium and a split rate typically between 1 to 2 and 1 to 4.
On day 33 the top gassing was replaced with deep sparging which is can be expected to cause more physical damage to the cells.
On day 50 onwards WCM5 (Example 2) was used together with peptone and PEG instead of WCM4.
On day 53 the PEG was replaced with 0.1% Pluronic F68. The resulting growth and antibody levels achieved are shown in the the attached graphs (FIGS. 1 and 2), and demonstrate the capacity of the invention to allow protein-free production of antibody in excess of 100 μg/ml in fermenters fermentors.
EXAMPLE 5
Growth of CHO AJ19 MCB1 in WCM4 and compared to CHO AJ19 MCB1 grown in serum containing medium
Chinese hamster ovary cells, CHO AJ19 MCB1, derived from CHO DUK cells. (Urlaub & Chasin PNAS, 1980, 77, 7, pp 4216-4220, 1980 ), were genetically engineered to produce tPA under methotrexate selection. This cell line had been routinely grown in a fermenter fermentor as a suspension culture using normal growth medium consisting of RPMI 1640 medium (GIBCO), 2.5% acid hydrolysed hydrolyzed adult bovine serum (Imperial), 0.5% Tryptone, 50 IU/ml polymycin, 20 IU/ml neomycin, 500 nM methotrexate (MTX).
Medium WCM4 was formulated to which was added:
    • 46B 0.25% w/v N-Z Soy Peptone (Sigma P1265), 0.1% w/v Polyethylene glycol (PEG) 20,000 (Serva, Carbowax® 20M). 1 uM MTX.
    • 46C 0.25% w/v Yeast extract (Sigma Y0500), 0.1% w/v PEG 20,000 1 uM MTX. In this medium the Iscoves' in CM4 was replaced by RPMI 1640 medium (ICN FLOW).
    • 46D 0.25% w/v Yeast extract, 0.1% w/v PEG 20,000, 1 uM MTX.
    • 46E 0.25% w/v Yeast extract, 0.1% w/v PEG 20,000, 0.25% Fetal bovine serum (Imperial), 1 uM MTX.
The yeast extract, Peptone and PEG were made up as 10% w/v solutions with water (Wellcome media production unit) and filtered through a 0.2 um disposable filter (Gelman, Supor Vac), then diluted for use. The cells were incubated at 37° C. in a humidified incubator containing 5% CO2.
Cells growing in normal growth medium were pelleted by centrifugation at 1200 g +4° C. for 5 minutes, were washed in RPMI 1640 without supplements and pelleted again. The cells were then resuspended at 105 cell/ml in normal growth medium (46A) and the other media (46B, 46C, 46D or 46E). 24 well plates (Costar 16 mm wells) were seeded with 1 ml/well and incubated, at 37° C. in an incubator containing 5% CO2. On days 3, 4, 5 and 6 one well of each was counted using a haemcytometer and trypan blue exclusion. Two further wells of each were harvested, pooled and pelleted at 1200 g +4° C. 5 minutes. The supernatant was separated and stored at −20° C. These samples were subsequently assayed for tPA. On day 6 samples from 46A and 46D only were harvested.
RESULTS
tPA specific activities in various crude harvests
Crude material produced in the five different media were tested using a QA validated ELISA assay to measure the tPA antigen concentrations μg/ml using binding to a polyclonal antibody against tPA, and clot lysis assay to measure tPA activity in IU/ml. From these results (Table 2), the specific activities were calculated.
TABLE 2
MEAN tPA MEAN tPA
DAYS ACTIVITY CONTENT SPECIFIC
IN CELLCOUNT ×10−5 IU/ml ug/ml ACTIVITY
EXPERIMENT CULTURE VIABLE NONVIABLE (n = 3) (n = 3) MegIU/mg
46A 3 3.5 0.1 3051 10.51 0.290
46A 4 3.7 0.3 4841 14.85 0.326
46A 5 4.1 0.2 5306 15.52 0.335
46A 6 5.8 0.5 8235 23.22 0.355
46B 3 5.2 0.1 2552 10.44 0.244
46B 4 7.2 0.3 5310 18.58 0.286
46B 5 7.8 0.2 6230 22.19 0.281
46C 3 3.8 0.2 2779 9.61 0.289
46C 4 4.9 0.3 3536 16.54 0.214
46C 5 5.6 0.3 4639 19.88 0.233
46D 3 7.5 0.2 4650 17.66 0.263
46D 4 8.3 0.8 7369 25.99 0.285
46D 5 7.4 1.0 7882 24.26 0.325
46D 6 6.1 2.0 8095 27.06 0.299
46E 3 6.4 0.1 6262 23.85 0.263
46E 4 7.3 0.5 10180 29.70 0.343
46E 5 6.1 1.3 9080 34.25 0.265
From the above table there was no change of the specific activity in the five different crudes. The yield of tPA from protein free medium B, C and D was nearly equal to the yield of tPA from standard growth medium in group A and E.
Example 6 Continuous growth of CHO AJ19 MCBI in WCM4
CHO AJ19 MCBI in WCM4 cells growing in normal growth medium were pelleted and washed as in Example 5 and were resuspended at 7×104/ml in 500 ml of medium 46B. These cells were transferred to a Techne spinner flask and incubated, as above, stirring at 40 rpm. At various time intervals the cells were counted and subcultured using the same medium. A sample was taken for tPA assay and treated as in Example 5.
The specific activity of tPA in various cell subcultures
The specific activity of supernatants from different pass levels of cells grown in WCM4 with peptone and 0.1% PEG 20K were measured by a combination of ELISA and clot lysis assay. The specific activities of different cell passages are summarised in Table 3.
TABLE 3
tPA present in supernatant
tPA
conc. ACTIVITY SPECIFIC
CELLCOUNT ×10−5 SPLIT ug/ml IU/ml ACTIVITY
DAYS PASS VIABLE NONVIABLE RATE (n = 3) (n = 3) Meg.U/mg
7 1 9.75 0.65  1-10 ND ND ND
10 2 4.95 0.01 1-5 ND ND ND
13 3 6.35 0.0  1-10 22.2 8865 0.399
16 4 3.8 0.0  1-10 7.25 1914 0.264
21 5 7.2 0.8  1-10 15.08 4331 0.287
24 6 4.1 0.3  1-10 8.28 2040 0.246
30 7 5.3 0.4 1-6 7.30 2052 0.281
34 8 5.2 0.32 13.65 3518 0.256
36 8 7.95 0.10 1-8 18.60 5327 0.286
37 8 ND ND 20.68 5526 0.267
38 8 100% 19.10 5474 0.287
38 9 12.00 0.5 1-5 20.85 8348 0.400
43 10 5.5 0.12 1-5 7.38 1888 0.256
48 11 4.4 0.19 1-6 13.4 3143 0.235
12 Experiment terminated
ND = not done.

Over a 48 day period, base on the above split rate, one cell could have divided to give 3.77×108 cells. This is equivalent to 31.8 population doublings with a doubling time of 36 hours.
The results of the experiments conducted in Examples 5 and 6 demonstrate that the serum free media of the present invention is capable of supporting cell growth and tPA yield comparable to that achieved in serum containing media.

Claims (41)

1. A method for growing genetically engineered CHO cells which comprises in suspension comprising the step of:
culturing genetically engineered CHO cells in suspension under cell growing conditions effective to support secretion of a product from the genetically engineered CHO cells over multiple passages for at least 20 days, wherein the cell growing conditions are characterized as:
in the absencebeing free of both serum and transferrin; and
incomprising a medium comprising water, an osmolality regulator, a buffer, an energy source. L-glutamine and at least one additional amino acid, an inorganic, organic or recombinant iron source and a recombinant or synthetic growth factor wherein each component of said medium is obtained from a source other than directly from an animal source.
2. A The method for culturing growing CHO cells in accordance with claim 1, wherein the medium further comprises at least one component selected from the group consisting of: non-ferrous metals, vitamins or and cofactors.
3. A The method for culturing growing CHO cells in accordance with claim 1, wherein the osmolality regulator maintains the medium at 200-350 mOsm.
4. A The method for culturing growing CHO cells in accordance with claim 1, wherein the medium is maintained at a pH in the range of about 6.5 to about 7.5 by the buffer.
5. A The method for culturing growing CHO cells in accordance with claim 1, wherein the concentration of the energy source is within the range of 1000-10,000 mg/liter.
6. A The method for culturing growing CHO cells in accordance with claim 5, wherein the energy source is a monosaccharide.
7. A The method for culturing growing CHO cells in accordance with claim 1, wherein the additional amino acids are selected from the group consisting of L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cystine cysteine, L-glutamic acid, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine and L-valine.
8. A The method for culturing growing CHO cells in accordance with claim 1, wherein the concentration of L-glutamine is within the range of 400-600 mg/liter.
9. A The method for culturing growing CHO cells in accordance with claim 2, wherein the medium further comprises a lipid factor in an amount of 0.05-10 mg/liter.
10. A The method for culturing growing CHO cells in accordance with claim 1, wherein the iron source is an inorganic ferric or ferrous salt which is provided in a concentration of from 0.25-5 mg/liter.
11. A The method for culturing growing CHO cells in accordance with claim 1, wherein the growth factor comprises is selected from the group consisting of: recombinant or synthetic insulin, platelet derived growth factor, thyroxine T3, thrombin, interleukin, progesterone, hydrocortisone or and vitamin E.
12. A The method for culturing growing CHO cells in accordance with claim 11 1, wherein the growth factor is recombinant or synthetic insulin.
13. A The method for culturing growing CHO cells in accordance with claim 1, wherein the medium further comprises at least one component selected from the group consisting of: a peptide digest, peptide hydrolysate or and peptide extract.
14. A method for culturing cells in accordance with claim 1, A method for growing genetically engineered CHO cells in suspension, comprising the step of:
culturing genetically engineered CHO cells in suspension under cell growing conditions effective to support secretion of a product from the genetically engineered CHO cells over multiple passages for at least 20 days, wherein the cell growing conditions are characterized as:
being free of both serum and transferring; and
comprising a medium comprising water, an osmolality regulator, a buffer, an energy source, L-glutamine, and at least one additional amino acid, an inorganic, organic or recombinant iron source and a recombinant or synthetic growth factor, wherein each component of said medium is obtained from a source other than directly from an animal source, wherein the medium is essentially free of hypoxanthine and thymidine.
15. A The method for culturing growing CHO cells in accordance with claim 14, wherein the medium further comprises methotrexate.
16. A method for culturing growing genetically engineered CHO cells which comprises in suspension comprising the step of:
culturing and growing Chinese hamster ovary genetically engineered CHO cells in suspension under cell growing conditions effective to support secretion of a product from the genetically engineered CHO cells over multiple passages for at least 20 days, wherein the cell growing conditions are characterized as:
in the absencebeing free of both serum and transferrin;
incomprising a medium comprising:
an osmolality regulator to maintain the osmolality of the medium within the range of about 200-350 mOsm,
a buffer to maintain the pH of the medium within the range of about 6.5 to 7.5,
about 1000-10,000 mg of a monosaccharide,
about 400-600 mg of L-glutamine,
about 10-200 mg of at least one amino acid selected from the group consisting of L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cystine cysteine, L-glutamic acid, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine and L-valine,
about 0.25-5 mg of an inorganic or recombinant iron source,
about 5 μg-5 mg of a recombinant or synthetic insulin,
and sufficient water to provide one liter of medium.
17. A method for culturing CHO cells which comprises culturing and growing Chinese hamster ovary cells in the absence of serum in a medium comprising
a base medium containing the amino acids, non-ferrous metal ions, vitamins and cofactors essentially as set forth in Table 1,
an osmolality regulator selected from NaCl, KCl, and KNO3 in an amount sufficient to maintain the osmolality of the medium within the range of about 200-350 mOsm,
at least one buffer selected from CaCl2.2H2O, MgSO4.7H2O, NaH2PO4.2H 2O, sodium pyruvate, N-[2-hydroxyethyl]piperazine-N′-[2-ethanesulphonic acid (HEPES) and 3-[N-morpholino]-propanesulfonic acid (MOPS) in an amount sufficient to maintain the medium within the pH range of about 6.5-7.5,
about 1000-10,000 mg of mannose, fructose, glucose or maltose,
about 5 ml of 200 mM L-glutamine,
about 50 mg each of L-proline, L-threonine, L-methionine, L-cysteine and L-tyrosine,
about 20-50 mg of L-ascorbic acid,
about 0.01-0.5 mg each of Vitamin B6 and Vitamin B12,
about 0.25-5 mg of a ferric or ferrous salt,
about 1 mg of zinc sulfate,
about 2.5 μg of copper sulfate,
about 10,000-100,000 IU of at least one antibiotic selected from the group consisting of polymyxin, neomycin, penicillin and streptomycin,
about 3 μl of ethanolamine,
about 0.01-1.0 mg of putrescine,
about 5 μg-5 mg of recombinant insulin and sufficient water to comprise one liter of medium; wherein each component of said medium is obtained from a source other than directly from an animal source.
18. A The method for growing CHO cells in accordance with claim 1, wherein each component of the medium is selected from at least one source selected from the group consisting of: an inorganic, synthetic, recombinant, plant or and bacterial source.
19. A method for growing genetically engineered CHO cells in suspension, comprising the step of:
culturing genetically engineered CHO cells in suspension at a density greater than 1×10 5 cells/mL under cell growing conditions effective to support secretion of a product from the genetically engineered CHO cells over multiple passages for at least 20 days, wherein the cell growing conditions are characterized as:
being free of both serum and transferrin; and
comprising a medium comprising water, an osmolality regulator, a buffer, an energy source, L-glutamine and at least one additional amino acid, an inorganic, organic or recombinant iron source and a recombinant or synthetic growth factor, wherein each component of said medium is obtained from a source other than directly from an animal source.
20. The method for growing CHO cells in accordance with any one of claims 1 to 16, 18, or 19, wherein said culturing results in secretion of said product from the genetically engineered CHO cells into said medium.
21. The method according to any one of claims 1 to 16, 18 or 19, wherein said method results in growth of said genetically engineered CHO cells in suspension and secretion of said product over multiple passages for at least 30 days.
22. The method according to any one of claims 1 to 16, 18 or 19, wherein said method results in growth of said genetically engineered CHO cells in suspension and secretion of said product over multiple passages for at least 40 days.
23. The method according to any one of claims 1 to 16, 18 or 19, wherein said method results in growth of said genetically engineered CHO cells in suspension and secretion of said product over multiple passages for at least 50 days.
24. The method according to any one of claims 1 to 16, 18 or 19, wherein said method results in growth of said genetically engineered CHO cells in suspension and secretion of said product over multiple passages for at least 60 days.
25. The method according to any one of claims 1 to 16, 18 or 19, wherein said method results in growth of said genetically engineered CHO cells in suspension and secretion of said product over multiple passages for at least 70 days.
26. The method according to any one of claims 1 to 16, 18 or 19, wherein said method results in growth of said genetically engineered CHO cells in suspension and secretion of said product over multiple passages for at least 80 days.
27. The method according to any one of claims 1 to 16, 18 or 19, wherein said method results in growth of said genetically engineered CHO cells in suspension and secretion of said product over multiple passages for at least 6 months.
28. A method for growing genetically engineered CHO cells in suspension, comprising the step of:
culturing genetically engineered CHO cells in suspension at a density greater than 1×10 5 cells/mL under cell growing conditions effective to support secretion of a product from the genetically engineered CHO cells over multiple passages for at least 20 days, wherein the cell growing conditions are characterized as:
being free of both serum and transferrin; and
comprising a medium comprising water, an osmolality regulator, a buffer, an energy source, L-glutamine and at least one additional amino acid, an inorganic, organic or recombinant iron source and a recombinant or synthetic growth factor, wherein each component of said medium is obtained from a source other than directly from an animal source,
wherein said culturing results in secretion of at least 30 mg/L of said product from the genetically engineered CHO cells into said medium.
29. The method according to claim any one of claims 1 to 16, 18, 19, or 28, wherein said genetically engineered CHO cells are dhfr CHO cells.
30. The method for growing CHO cells in accordance with any one of claims 14, 16, 19, or 28 wherein the medium further comprises at least one component selected from the group consisting of: non-ferrous metals, vitamins and cofactors.
31. The method for growing CHO cells in accordance with any one of claims 14, 19, or 28, wherein the osmolality regulator maintains the medium at 200-350 mOsm.
32. The method for growing CHO cells in accordance with any one of claims 14, 19, or 28, wherein the medium is maintained at a pH in the range of about 6.5 to about 7.5 by the buffer.
33. The method for growing CHO cells in accordance with any one of claims 14, 19, or 28, wherein the concentration of the energy source is within the range of 1000-10,000 mg/liter.
34. The method for growing CHO cells in accordance with claim 33, wherein the energy source is a monosaccharide.
35. The method for growing CHO cells in accordance with any one of claims 14, 19, or 28, wherein the additional amino acids are selected from the group consisting of L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine and L-valine.
36. The method for growing CHO cells in accordance with any one of claims 14, 19, or 28, wherein the concentration of L-glutamine is within the range of 400-600 mg/liter.
37. The method for growing CHO cells in accordance with any one of claims 14, 16, 19, or 28, wherein the medium further comprises a lipid factor in an amount of 0.05-10 mg/liter.
38. The method for growing CHO cells in accordance with any one of claims 14, 19, or 28, wherein the iron source is an inorganic ferric or ferrous salt which is provided in a concentration of 0.25-5 mg/liter.
39. The method for growing CHO cells in accordance with any one of claims 14, 19, or 28, wherein the growth factor is selected from the group consisting of: recombinant or synthetic insulin, platelet derived growth factor, thyroxine T3 , thrombin, interleukin, progesterone, hydrocortisone and vitamin E.
40. The method for growing CHO cells in accordance with any one of claims 14, 19, or 28, wherein the growth factor is recombinant or synthetic insulin.
41. The method for growing CHO cells in accordance with any one of claims 14, 16, 19, or 28, wherein the medium further comprises at least one component selected from the group consisting of: a peptide digest, peptide hydrolysate and peptide extract.
US11/640,428 1990-10-17 2006-12-15 Method for culturing Chinese hamster ovary cells Expired - Lifetime USRE41974E1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/640,428 USRE41974E1 (en) 1990-10-17 2006-12-15 Method for culturing Chinese hamster ovary cells

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
GB9022545A GB9022545D0 (en) 1990-10-17 1990-10-17 Culture medium
US77772991A 1991-10-16 1991-10-16
US07/991,717 US5316938A (en) 1990-10-17 1992-12-18 Defined media for serum-free tissue culture
US08/205,379 US5633162A (en) 1990-10-17 1994-03-04 Method for culturing Chinese hamster ovary cells
US10/995,010 USRE39792E1 (en) 1990-10-17 2004-11-22 Method for culturing Chinese hamster ovary cells
US11/640,428 USRE41974E1 (en) 1990-10-17 2006-12-15 Method for culturing Chinese hamster ovary cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US08/205,379 Reissue US5633162A (en) 1990-10-17 1994-03-04 Method for culturing Chinese hamster ovary cells

Publications (1)

Publication Number Publication Date
USRE41974E1 true USRE41974E1 (en) 2010-11-30

Family

ID=10683864

Family Applications (4)

Application Number Title Priority Date Filing Date
US07/991,717 Expired - Lifetime US5316938A (en) 1990-10-17 1992-12-18 Defined media for serum-free tissue culture
US08/205,379 Ceased US5633162A (en) 1990-10-17 1994-03-04 Method for culturing Chinese hamster ovary cells
US10/995,010 Expired - Lifetime USRE39792E1 (en) 1990-10-17 2004-11-22 Method for culturing Chinese hamster ovary cells
US11/640,428 Expired - Lifetime USRE41974E1 (en) 1990-10-17 2006-12-15 Method for culturing Chinese hamster ovary cells

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US07/991,717 Expired - Lifetime US5316938A (en) 1990-10-17 1992-12-18 Defined media for serum-free tissue culture
US08/205,379 Ceased US5633162A (en) 1990-10-17 1994-03-04 Method for culturing Chinese hamster ovary cells
US10/995,010 Expired - Lifetime USRE39792E1 (en) 1990-10-17 2004-11-22 Method for culturing Chinese hamster ovary cells

Country Status (13)

Country Link
US (4) US5316938A (en)
EP (3) EP1221476B1 (en)
JP (1) JP2625302B2 (en)
AT (2) ATE382680T1 (en)
AU (1) AU645615B2 (en)
CA (1) CA2053586C (en)
DE (2) DE69133589T2 (en)
DK (2) DK0481791T3 (en)
ES (2) ES2298301T3 (en)
GB (1) GB9022545D0 (en)
IE (1) IE913559A1 (en)
NZ (1) NZ240248A (en)
ZA (1) ZA918249B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110008464A1 (en) * 2009-07-10 2011-01-13 Scott Iii Linzy O Methods and compositions for treating thyroid-related medical conditions with reduced folates
WO2013006960A1 (en) * 2011-07-13 2013-01-17 Foodchek Systems, Inc. Culture medium, method for culturing listeria, and method for detecting listeria
US9481861B2 (en) 2011-07-12 2016-11-01 Foodchek Systems, Inc. Culture medium, method for culturing Salmonella and E. coli and method for detecting Salmonella and E. coli

Families Citing this family (206)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5292655A (en) * 1990-01-29 1994-03-08 Wille Jr John J Method for the formation of a histologically-complete skin substitute
GB9022545D0 (en) 1990-10-17 1990-11-28 Wellcome Found Culture medium
GB9022543D0 (en) * 1990-10-17 1990-11-28 Wellcome Found Antibody production
JPH0789908B2 (en) * 1991-02-28 1995-10-04 倉敷紡績株式会社 Serum-free medium for animal cell culture
DE4115722A1 (en) * 1991-05-14 1992-11-19 Boehringer Mannheim Gmbh SERUM-FREE MEDIUM FOR CULTIVATING SUGAR CELLS
GB9118664D0 (en) * 1991-08-30 1991-10-16 Celltech Ltd Cell culture
US6231881B1 (en) 1992-02-24 2001-05-15 Anton-Lewis Usala Medium and matrix for long-term proliferation of cells
US6352707B1 (en) 1992-02-24 2002-03-05 Anton-Lewis Usala Transplant encapsulation in a hydrogel matrix to obscure immune recognition
IL102929A (en) * 1992-08-24 1996-11-14 Interpharm Lab Ltd Serum-free medium for mammalian cells
DE4313620A1 (en) * 1993-04-26 1994-10-27 Biotechnolog Forschung Gmbh Hamster cell lines and methods for glycoprotein recovery
USH1532H (en) * 1993-11-03 1996-05-07 Genetics Institute, Inc. Adaption of mammalian cell lines to high cell densities
EP0653487A1 (en) * 1993-11-07 1995-05-17 Ferruccio Dr. Messi Serum and protein-free growing cells
US5856179A (en) * 1994-03-10 1999-01-05 Genentech, Inc. Polypeptide production in animal cell culture
US5512477A (en) * 1994-04-21 1996-04-30 Genzyme Corporation Serum-free medium supplement
US6146873A (en) * 1994-11-10 2000-11-14 Baxter Aktiengesellschaft Production of orthomyxoviruses in monkey kidney cells using protein-free media
US5698433A (en) * 1994-11-10 1997-12-16 Immuno Ag Method for producing influenza virus and 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
US5705364A (en) 1995-06-06 1998-01-06 Genentech, Inc. Mammalian cell culture process
US6656466B1 (en) 1995-06-06 2003-12-02 Genetech, Inc. Human tumor necrosis factor—immunoglobulin(TNFR1-IgG1) chimera composition
US5721121A (en) * 1995-06-06 1998-02-24 Genentech, Inc. Mammalian cell culture process for producing a tumor necrosis factor receptor immunoglobulin chimeric protein
EP0891419A4 (en) * 1996-03-12 2000-03-01 Life Technologies Inc Hematopoietic cell culture nutrient supplement
US6180404B1 (en) * 1996-04-09 2001-01-30 The Board Of Trustees Of Southern Illinois University Cultural medium for maintaining neural cells in ambient atmosphere
EP2218775B1 (en) 1996-08-30 2015-01-28 Life Technologies Corporation Method for producing a polypeptide in vitro in mammalian cells in a protein-free and serum-free culture medium
US20040171152A1 (en) * 1996-10-10 2004-09-02 Invitrogen Corporation Animal cell culture media comprising non-animal or plant-derived nutrients
JP4543402B2 (en) * 1996-10-10 2010-09-15 ライフ テクノロジーズ コーポレーション Animal cell culture medium containing plant-derived nutrients
US6692961B1 (en) * 1996-10-11 2004-02-17 Invitrogen Corporation Defined systems for epithelial cell culture and use thereof
US20020012991A1 (en) * 1997-04-07 2002-01-31 Florence Chua Nee Ho Kit Fong Cell culture media for enhanced protein production
US6475725B1 (en) 1997-06-20 2002-11-05 Baxter Aktiengesellschaft Recombinant cell clones having increased stability and methods of making and using the same
US6989264B2 (en) 1997-09-05 2006-01-24 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
US6566118B1 (en) * 1997-09-05 2003-05-20 Targeted Genetics Corporation Methods for generating high titer helper-free preparations of released recombinant AAV vectors
WO1999057246A1 (en) * 1998-05-01 1999-11-11 Life Technologies, Inc. Animal cell culture media comprising non-animal or plant-derived nutrients
US6528286B1 (en) * 1998-05-29 2003-03-04 Genentech, Inc. Mammalian cell culture process for producing glycoproteins
US6406909B1 (en) * 1998-07-10 2002-06-18 Chugai Seiyaku Kabushiki Kaisha Serum-free medium for culturing animal cells
US7294481B1 (en) * 1999-01-05 2007-11-13 Immunex Corporation Method for producing recombinant proteins
ES2420835T3 (en) 1999-04-09 2013-08-27 Kyowa Hakko Kirin Co., Ltd. Procedure to control the activity of immunofunctional molecules
US20060286668A1 (en) * 1999-04-30 2006-12-21 Invitrogen Corporation Animal-cell culture media comprising non-animal or plant-derived nutrients
AT409379B (en) * 1999-06-02 2002-07-25 Baxter Ag MEDIUM FOR PROTEIN- AND SERUM-FREE CELL CULTURE
JO2291B1 (en) 1999-07-02 2005-09-12 اف . هوفمان لاروش ايه جي Erythopintin derivatives
CA2381173A1 (en) * 1999-08-05 2001-02-15 Baxter Aktiengesellschaft Recombinant stable cell clone, its production and use thereof
WO2001029246A1 (en) * 1999-10-19 2001-04-26 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
KR20010056451A (en) * 1999-12-15 2001-07-04 윤재승 Arginine-enriched medium used for mass-producing recombinant protein in animal cell culture
US6811776B2 (en) 2000-12-27 2004-11-02 The Regents Of The University Of Michigan Process for ex vivo formation of mammalian bone and uses thereof
EP1364002A2 (en) * 2000-08-23 2003-11-26 Pfizer Products Inc. Process for the preparation of neutrophil inhibitory factor
US20020099183A1 (en) * 2000-08-23 2002-07-25 Pluschkell Stefanie Beate Process for the preparation of neutrophil inhibitory factor
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
KR100645843B1 (en) 2000-12-20 2006-11-14 에프. 호프만-라 로슈 아게 Erythropoietin conjugates
US6506576B2 (en) 2001-03-14 2003-01-14 Board Of Trustees Of The University Of Arkansas Serum-and steroid-free culture media for cerebellar granule neurons
US20050069979A1 (en) * 2001-11-28 2005-03-31 Steffen Zeng Cell culture process
US20080254027A1 (en) * 2002-03-01 2008-10-16 Bernett Matthew J Optimized CD5 antibodies and methods of using the same
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US7662925B2 (en) * 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US20070148171A1 (en) * 2002-09-27 2007-06-28 Xencor, Inc. Optimized anti-CD30 antibodies
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US20080260731A1 (en) * 2002-03-01 2008-10-23 Bernett Matthew J Optimized antibodies that target cd19
JPWO2003084569A1 (en) 2002-04-09 2005-08-11 協和醗酵工業株式会社 Antibody composition-containing medicine
PL214284B1 (en) 2002-07-09 2013-07-31 Baxter Healthcare Sa Animal protein free media for cultivation of cells
US7067279B1 (en) 2002-08-23 2006-06-27 Immunex Corporation Cell culture performance with betaine
US20060235208A1 (en) * 2002-09-27 2006-10-19 Xencor, Inc. Fc variants with optimized properties
AU2003274011B2 (en) 2002-10-15 2010-03-04 Intercell Ag Nucleic acids coding for adhesion factor of group B streptococcus, adhesion factors of group B streptococcus and further uses thereof
DE10255508A1 (en) * 2002-11-27 2004-06-17 Forschungszentrum Jülich GmbH Process for cultivating cells for the production of substances
CN100402660C (en) 2002-12-23 2008-07-16 布里斯托尔-迈尔斯斯奎布公司 Product quality enhancement in mammalian cell culture processes for protein production
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US8388955B2 (en) * 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US20070275460A1 (en) * 2003-03-03 2007-11-29 Xencor.Inc. Fc Variants With Optimized Fc Receptor Binding Properties
US20090010920A1 (en) 2003-03-03 2009-01-08 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIb
GB0304799D0 (en) 2003-03-03 2003-04-09 Glaxosmithkline Biolog Sa Novel method
EP1622941A2 (en) * 2003-03-20 2006-02-08 ImClone Systems Incorporated Method of producing an antibody to epidermal growth factor receptor
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
GB2404665B (en) * 2003-08-08 2005-07-06 Cambridge Antibody Tech Cell culture
ATE359359T1 (en) * 2003-08-08 2007-05-15 Cambridge Antibody Tech MYELOMA CELL CULTURE IN A TRANSFERRIN-FREE MEDIUM WITH LOW IRON
CA2537462A1 (en) * 2003-09-18 2005-03-31 Raven Biotechnologies, Inc. Cell culture media comprising fructose as the primary energy source
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
US8101720B2 (en) * 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
EP1670821A4 (en) * 2003-10-09 2006-11-29 Daewoong Co Ltd Process for purifying human thrombopoietin with high content of sialic acid
AU2004280064A1 (en) * 2003-10-09 2005-04-21 Kyowa Hakko Kirin Co., Ltd. Genomically modified cell neutralized to serum-free system
BR122020007658B1 (en) * 2003-10-27 2021-07-27 Wyeth METHOD FOR REMOVING HIGH MOLECULAR WEIGHT AGGREGATE USING HYDROXIAPATITE CHROMATOGRAPHY
US20050249723A1 (en) * 2003-12-22 2005-11-10 Xencor, Inc. Fc polypeptides with novel Fc ligand binding sites
US7255288B2 (en) * 2004-03-08 2007-08-14 Wan Shan Chan Aroma therapy for fountain
EP2053062A1 (en) * 2004-03-24 2009-04-29 Xencor, Inc. Immunoglobin variants outside the Fc region
US20150010550A1 (en) 2004-07-15 2015-01-08 Xencor, Inc. OPTIMIZED Fc VARIANTS
TWI374935B (en) 2004-08-27 2012-10-21 Pfizer Ireland Pharmaceuticals Production of α-abeta
TWI364458B (en) 2004-08-27 2012-05-21 Wyeth Res Ireland Ltd Production of tnfr-lg
TWI384069B (en) 2004-08-27 2013-02-01 Pfizer Ireland Pharmaceuticals Production of polypeptides
US20060074225A1 (en) * 2004-09-14 2006-04-06 Xencor, Inc. Monomeric immunoglobulin Fc domains
AU2011221414B2 (en) * 2004-10-29 2012-09-20 Takeda Pharmaceutical Company Limited Animal Protein-Free Media for Cultivation of Cells
US20060094104A1 (en) * 2004-10-29 2006-05-04 Leopold Grillberger Animal protein-free media for cultivation of cells
ES2361559T3 (en) 2004-11-02 2011-06-20 Ares Trading S.A. MEANS OF GROWING CELLS EXEMPT FROM SERUM FOR MAMMALS.
BRPI0517942B8 (en) * 2004-11-02 2021-05-25 Ares Trading Sa process for the production of recombinant human growth hormone (rhgh) and uses of a serum-free cell culture medium
US8273553B2 (en) * 2004-11-02 2012-09-25 Ares Trading S.A. Production of growth hormone in serum-free cell culture medium for mammalian cells
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US8802820B2 (en) * 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US20070135620A1 (en) * 2004-11-12 2007-06-14 Xencor, Inc. Fc variants with altered binding to FcRn
EP2314618A3 (en) 2004-11-12 2011-10-19 Xencor Inc. Fc variants with altered binding to FcRn
CA2595169A1 (en) * 2005-01-12 2006-07-20 Xencor, Inc. Antibodies and fc fusion proteins with altered immunogenicity
ES2380485T3 (en) 2005-02-11 2012-05-14 Novo Nordisk Health Care Ag Production of a polypeptide in a serum-free culture liquid with vegetable protein hydrolyzate
JP2006310392A (en) * 2005-04-26 2006-11-09 Toshiba Corp Method and apparatus of electron beam lithography
JP2008541746A (en) * 2005-06-03 2008-11-27 ビオヴィトルム・アクチボラゲット(プブリクト) New process
TWI369401B (en) * 2005-07-05 2012-08-01 Ares Trading Sa Serum-free culture medium for the production of recombinant gonadotropins
CA2624189A1 (en) * 2005-10-03 2007-04-12 Xencor, Inc. Fc variants with optimized fc receptor binding properties
CA2625998C (en) 2005-10-06 2015-12-01 Xencor, Inc. Optimized anti-cd30 antibodies
PL1974014T3 (en) 2006-01-04 2017-09-29 Baxalta Incorporated Oligopeptide-free cell culture media
US20070190057A1 (en) 2006-01-23 2007-08-16 Jian Wu Methods for modulating mannose content of recombinant proteins
CA2657248C (en) 2006-07-13 2018-10-30 Wyeth Production of glycoproteins
CA2659990C (en) 2006-08-04 2016-03-22 Prolong Pharmaceuticals, Inc. Polyethylene glycol erythropoietin conjugates
DK2383297T5 (en) 2006-08-14 2022-07-04 Xencor Inc Optimized antibodies directed against CD19
CA2910619A1 (en) * 2006-09-13 2008-03-20 Abbvie Inc. Cell culture improvements
EP2500416A1 (en) * 2006-09-13 2012-09-19 Abbott Laboratories Cell culture improvements
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
CA2660795C (en) * 2006-09-18 2014-11-18 Xencor, Inc. Optimized antibodies that target hm1.24
EP2511705A1 (en) 2007-04-16 2012-10-17 Momenta Pharmaceuticals, Inc. Methods related to cell surface glycosylation
EP2135093B1 (en) * 2007-04-16 2015-04-15 Momenta Pharmaceuticals, Inc. Analysis of phosphorylated glycans, glcopeptides or glycoproteins by imac
EP2135089B1 (en) * 2007-04-16 2015-09-02 Momenta Pharmaceuticals, Inc. Comparative analysis of protein conformations by using 2d noesy nmr spectra
TW200902708A (en) 2007-04-23 2009-01-16 Wyeth Corp Methods of protein production using anti-senescence compounds
CN101663391A (en) 2007-04-26 2010-03-03 中外制药株式会社 Cell culture method using amino acid-enriched medium
GB0710614D0 (en) * 2007-06-04 2007-07-11 Lonza Biologics Plc Mammalian expression vector with a highly efficient secretory signal sequence
CN101319200B (en) * 2007-06-08 2010-05-19 中国科学院大连化学物理研究所 Non-serum medium suitable for microencapsulation CHO cell and uses thereof
US20100221823A1 (en) * 2007-06-11 2010-09-02 Amgen Inc. Method for culturing mammalian cells to improve recombinant protein production
US7580304B2 (en) * 2007-06-15 2009-08-25 United Memories, Inc. Multiple bus charge sharing
EP3327132A3 (en) * 2007-08-09 2018-07-18 Wyeth LLC Use of perfusion to enhance production of fed-batch cell culture in bioreactors
CA2701646C (en) * 2007-10-12 2019-02-26 F. Hoffmann-La Roche Ag Protein expression from multiple nucleic acids
ES2742268T3 (en) 2007-12-26 2020-02-13 Xencor Inc Fc variants with altered FcRn binding
CN106222220A (en) * 2007-12-27 2016-12-14 百深有限责任公司 Cell culture processes
EP2250251B1 (en) 2008-01-09 2017-11-22 Sartorius Stedim Cellca GmbH Improved culture media additive and process for using it
JP2011512877A (en) * 2008-03-12 2011-04-28 ワイス・エルエルシー Methods for identifying cells suitable for large-scale production of recombinant proteins
WO2009127098A1 (en) * 2008-04-18 2009-10-22 上海中信国健药业有限公司 A concentrated culture solution and application method thereof
DE102008002210A1 (en) 2008-06-04 2009-12-10 Evonik Degussa Gmbh Process for the fermentative production of erythropoietin
US8058407B2 (en) * 2008-10-31 2011-11-15 Wyeth Llc Purification of acidic proteins using ceramic hydroxyapatite chromatography
EP2356247B2 (en) * 2008-11-12 2019-05-15 Baxalta Incorporated Method of producing serum-free insulin-free factor vii
CA2745218A1 (en) * 2008-12-19 2010-06-24 Schering Corporation Feed supplement for mammalian cell culture and methods of use
BRPI0923191A2 (en) 2008-12-19 2016-02-16 Momenta Pharmaceuticals Inc Modified glycan related methods
HUE036415T2 (en) 2008-12-30 2018-07-30 Baxalta GmbH Method of enhancing cell growth using alkyl-amine-n-oxide (aanox)
SG173457A1 (en) * 2009-02-27 2011-09-29 Novartis Ag Methods for selecting eukaryotic cells expressing a heterologous protein
US8597943B2 (en) 2009-04-09 2013-12-03 Cellca Gmbh Method for improved single cell cloning
KR101573972B1 (en) 2009-07-31 2015-12-02 백스터 인터내셔널 인코포레이티드 Cell culture medium for adamts protein expression
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
US20120264688A1 (en) 2009-09-23 2012-10-18 Walter Hinderer Process for the purification of recombinant human erythropoietin (epo), epo thus purified and pharmaceutical compositions comprising same
KR101961254B1 (en) * 2009-10-26 2019-03-22 에프. 호프만-라 로슈 아게 Method for the production of a glycosylated immunoglobulin
EP2516624B1 (en) * 2009-12-23 2020-02-05 Merck Sharp & Dohme Corp. Cell line 3m
US8362210B2 (en) 2010-01-19 2013-01-29 Xencor, Inc. Antibody variants with enhanced complement activity
SG185038A1 (en) 2010-04-26 2012-11-29 Novartis Ag Improved cell culture medium
CN107254499B (en) 2010-04-26 2021-09-17 诺华股份有限公司 Improved cell culture process
CN102234627B (en) * 2010-04-30 2015-06-03 中国科学院广州生物医药与健康研究院 Culture medium additive and application thereof
BR112013000512A2 (en) 2010-07-08 2016-05-17 Baxter Healthcare Sa method for producing a recombinant adamts 13 (ra13) composition, cell culture supernatant, and recombinant adamts13 (ra13) composition
CN103080300B (en) 2010-08-05 2015-11-25 安姆根有限公司 Increase the productive rate of cell culture and the dipeptides of vigor
EP3147355B1 (en) * 2010-12-27 2020-07-22 Kyowa Kirin Co., Ltd. Method for preparing aqueous solution containing culture medium and chelating agent
US9133493B2 (en) 2011-04-21 2015-09-15 Amgen Inc. Method for culturing mammalian cells to improve recombinant protein production
EP2702077A2 (en) 2011-04-27 2014-03-05 AbbVie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
WO2012147053A1 (en) 2011-04-29 2012-11-01 Biocon Research Limited A method for reducing heterogeneity of antibodies and a process of producing the antibodies thereof
KR20170021919A (en) 2011-10-21 2017-02-28 화이자 인코포레이티드 Addition of iron to improve cell culture
AU2013222188A1 (en) 2012-02-22 2014-09-11 Amgen Inc. Autologous mammalian models derived from induced pluripotent stem cells and related methods
WO2013158279A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Protein purification methods to reduce acidic species
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
AU2013309506A1 (en) 2012-09-02 2015-03-12 Abbvie Inc. Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
HUE026516T2 (en) 2012-09-24 2016-06-28 Lonza Biologics Plc Expression vectors comprising chimeric cytomegalovirus promoter and enhancer sequences
CA2905010A1 (en) 2013-03-12 2014-09-18 Abbvie Inc. Human antibodies that bind human tnf-alpha and methods of preparing the same
US9217168B2 (en) 2013-03-14 2015-12-22 Momenta Pharmaceuticals, Inc. Methods of cell culture
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
ITTO20130493A1 (en) * 2013-06-14 2014-12-15 Determinants Of Metabolism Res Lab S R L COMPOSITION FOR THE ELIMINATION OF MANY ANIMALS
JP6195191B2 (en) * 2013-08-08 2017-09-13 極東製薬工業株式会社 Method for producing recombinant protein using cell line conditioned in protein-free and lipid-free medium
JP6190205B2 (en) * 2013-08-08 2017-08-30 極東製薬工業株式会社 Protein-free and lipid-free medium conditioned cell line, its production method and medium
EP3039129B1 (en) 2013-08-30 2018-06-27 Amgen Inc. High titer recombinant aav vector production in adherent and suspension cells
EP3052640A2 (en) 2013-10-04 2016-08-10 AbbVie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
WO2016049363A1 (en) * 2014-09-24 2016-03-31 Los Alamos National Security, Llc Bio-assessment device and method of making the device
US10202616B2 (en) 2014-10-15 2019-02-12 Amgen Inc. Promoter and regulatory elements for improved expression of heterologous genes in host cells
JP6432774B2 (en) * 2014-12-25 2018-12-05 江南化工株式会社 Cell activator
US11320422B2 (en) 2016-01-06 2022-05-03 Lonza Ltd. Inhibition of protein degradation for improved production
US10119117B2 (en) 2016-01-28 2018-11-06 Nanogen Pharmaceutical Biotechnology Co., Ltd Universal, glycosylation enhancer, completely chemically defined medium formulation
WO2017146646A1 (en) * 2016-02-22 2017-08-31 Agency For Science, Technology And Research Cell culture medium
US10689873B2 (en) 2016-03-10 2020-06-23 Lonza Ltd Customizable facility
BR112018068281A2 (en) 2016-03-10 2019-04-02 Lonza Ag customizable installation
KR102408260B1 (en) 2016-04-14 2022-06-10 론자 리미티드 Compositions and Methods for Detection of Host Cell Proteins
TWI734775B (en) * 2016-04-26 2021-08-01 美商美國泰福生技股份有限公司 Cell culture medium
KR102536221B1 (en) 2016-05-03 2023-05-23 론자 리미티드 Modulation of lipid metabolism for protein production
JP2019517272A (en) 2016-06-10 2019-06-24 ロンザ リミテッドLonza Limited Protein stabilization method
SG11201900528PA (en) 2016-08-02 2019-02-27 Lonza Ag Customizable facility
EP3485280A1 (en) 2016-08-12 2019-05-22 Lonza Ltd Proteomic analysis of host cell proteins
JP7170329B2 (en) 2016-09-16 2022-11-14 ロイコケア・アクチェンゲゼルシャフト Novel methods for stabilization of biopharmaceutical products during processing
CN106635958A (en) * 2016-12-24 2017-05-10 严志海 CHO (Chinese Hamster Ovary) cell culture medium
MX2019010575A (en) 2017-03-10 2019-10-15 Hoffmann La Roche Method for producing multispecific antibodies.
EP3622072A1 (en) 2017-06-16 2020-03-18 Lonza Ltd Universal self-regulating mammalian cell line platform for the production of biologics
JP2020533983A (en) 2017-09-15 2020-11-26 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Online biomass capacitance monitoring during large-scale production of the polypeptide of interest
AU2018381301A1 (en) 2017-12-05 2020-06-11 Lonza Ltd Methods of assaying tropolone
US20230174971A1 (en) 2018-02-02 2023-06-08 Lonza Ltd Methods of cell selection and modifying cell metabolism
CN111902160A (en) 2018-03-16 2020-11-06 百时美施贵宝公司 Metabolic enzyme activity and disulfide bond reduction during protein production
US20190367858A1 (en) 2018-06-01 2019-12-05 Lonza Ltd. Midscale Model For Organic Growth and Phasing
KR20210030949A (en) 2018-07-13 2021-03-18 론자 리미티드 How to improve production of biological products by reducing the level of endogenous proteins
WO2020028616A1 (en) 2018-08-02 2020-02-06 Lonza Ltd Methods for manufacturing recombinant protein comprising a disulfide bond
CN110343666B (en) * 2019-07-10 2023-05-30 通化东宝药业股份有限公司 Feed supplement culture medium for CHO cell culture and preparation method and application thereof
IL292890A (en) 2019-11-14 2022-07-01 Lonza Ag Methods of cell selection
MX2022006759A (en) 2019-12-06 2023-01-30 Regeneron Pharma Anti-vegf protein compositions and methods for producing the same.
JPWO2021193748A1 (en) * 2020-03-25 2021-09-30
JP2024503239A (en) 2020-12-22 2024-01-25 アムジェン インコーポレイテッド Cell culture method
WO2023045140A1 (en) * 2021-09-26 2023-03-30 上海迈泰君奥生物技术有限公司 Method for increasing expression quantity of antibody in host cell
CN113846051B (en) * 2021-09-28 2023-12-01 无锡多宁生物科技有限公司 Universal chemical composition limiting CHO cell subculture medium and application thereof

Citations (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4205126A (en) 1978-01-01 1980-05-27 Cartaya Oscar A Serum-free cell culture media
FR2543158A1 (en) 1983-03-24 1984-09-28 Inst Nat Sante Rech Med CULTURE MEDIUM OF ANIMAL CELLS WITHOUT SERUM, WITHOUT HORMONES AND WITHOUT GROWTH FACTORS AND METHODS OF PRIMARY CULTURE AND OBTAINING CELL LINES USING THE SAME
EP0164813A2 (en) 1984-06-14 1985-12-18 Teijin Limited Method of cultivating animal or plant cells
JPS6125480A (en) 1984-07-13 1986-02-04 Nitsusui Seiyaku Kk Serum-free synthetic medium for cell culture
WO1987000195A1 (en) 1985-06-28 1987-01-15 Celltech Limited Animal cell culture
WO1987001131A1 (en) 1985-08-19 1987-02-26 Gene Labs, Inc. Non-human primate monoclonal antibodies and methods
US4657866A (en) 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
EP0229530A2 (en) 1985-12-31 1987-07-22 Bass Public Limited Company The propagation of yeast, fermentation employing that yeast and products of that fermentation
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
EP0248656A2 (en) 1986-06-04 1987-12-09 Director-General of the Agency of Industrial Science and Technology Composition for cell cultivation and use thereof
JPS637780A (en) 1986-06-28 1988-01-13 Nippon Zenyaku Kogyo Kk Feeding method for iron to cell and serum-free synthetic culture medium used thereof
WO1988000967A1 (en) 1986-08-04 1988-02-11 The University Of New South Wales Serum free tissue culture medium containing polymeric cell-protective agent
GB2196348A (en) 1986-10-03 1988-04-27 Ceskoslovenska Akademie Ved Synthetic medium for hybridoma and myeloma cell cultivation
EP0274394A2 (en) 1987-01-08 1988-07-13 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
JPS63192381A (en) 1987-02-05 1988-08-09 Kanegafuchi Chem Ind Co Ltd Cell capable of subcultivation in serum-free culture medium and method for obtaining said cell
US4767704A (en) 1983-10-07 1988-08-30 Columbia University In The City Of New York Protein-free culture medium
WO1989000999A1 (en) 1987-07-24 1989-02-09 International Genetic Engineering, Inc. Modular assembly of antibody genes, antibodies prepared thereby and use
EP0307248A2 (en) 1987-09-11 1989-03-15 Genentech, Inc. Method for increasing the expression of polypeptides in recombinant cell culture
EP0307247A2 (en) 1987-09-11 1989-03-15 Genentech, Inc. A method for culturing recombinant cells
EP0314161A1 (en) 1987-10-28 1989-05-03 Bristol-Myers Squibb Company Human immunoglobulines produced by recombinant DNA techniques
EP0316068A1 (en) 1987-10-09 1989-05-17 Collaborative Research Inc. Modified low molecular weight plasminogen activator and method of preparation
EP0325190A2 (en) 1988-01-18 1989-07-26 Roche Diagnostics GmbH Pentosansulfate medium
EP0328404A1 (en) 1988-02-12 1989-08-16 Btg International Limited Modified antibodies
WO1990003429A1 (en) 1988-09-23 1990-04-05 Cetus Corporation Lipid microemulsions for culture media
EP0363703A1 (en) 1988-09-23 1990-04-18 Kenneth Alonso Method of producing human-human hybridomas, the production of monoclonal and polyclonal antibodies therefrom, and therapeutic use thereof
US4929706A (en) 1988-11-02 1990-05-29 W. R. Grace & Co.-Conn. Cell growth enhancers and/or antibody production stimulators comprising chemically modified hydrophilic polyurea-urethane prepolymers and polymers
EP0388151A1 (en) 1989-03-13 1990-09-19 Celltech Limited Modified antibodies
EP0389786A1 (en) 1989-03-03 1990-10-03 W.R. Grace & Co.-Conn. Very low protein nutrient medium for cell culture
EP0390327A2 (en) 1989-02-27 1990-10-03 Eli Lilly And Company improved tissue culture method
EP0404003A2 (en) 1989-06-19 1990-12-27 Xoma Corporation Chimeric mouse-human KM10 antibody with specificity to a human tumor cell antigen
WO1991004336A1 (en) 1989-09-19 1991-04-04 Centocor, Inc. Method for improving human monoclonal antibody production
US5019499A (en) 1986-04-14 1991-05-28 Daiichi Seiyaku Co., Ltd. Method of producing peptides by transforming myeloma cells with a recombinant plasmid
WO1991010722A2 (en) 1989-12-27 1991-07-25 Centocor, Inc. Chimeric immunoglobulin for cd4 receptors
US5045468A (en) 1986-12-12 1991-09-03 Cell Enterprises, Inc. Protein-free culture medium which promotes hybridoma growth
EP0481791A2 (en) 1990-10-17 1992-04-22 The Wellcome Foundation Limited Culture medium for CHO-cells and adapted CHO-cells
EP0481790A2 (en) 1990-10-17 1992-04-22 The Wellcome Foundation Limited Antibody production
WO1992007084A1 (en) 1990-10-17 1992-04-30 The Wellcome Foundation Limited Purified cdw52-specific antibodies
US5122469A (en) * 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
EP0513738A2 (en) 1991-05-14 1992-11-19 Roche Diagnostics GmbH Serum-free medium for mammalian cells cultivation
EP0523949A1 (en) 1991-07-15 1993-01-20 The Wellcome Foundation Limited Production of antibodies
WO1993002108A1 (en) 1991-07-25 1993-02-04 Idec Pharmaceuticals Corporation Recombinant antibodies for human therapy
WO1993007899A1 (en) 1991-10-15 1993-04-29 The Wellcome Foundation Limited CDw52 - SPECIFIC ANTIBODY FOR TREATMENT OF T-CELL MEDIATED INFLAMMATION OF THE JOINTS
US5232848A (en) 1987-03-24 1993-08-03 W. R. Grace & Co.-Conn. Basal nutrient medium for cell culture
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US5846534A (en) 1988-02-12 1998-12-08 British Technology Group Limited Antibodies to the antigen campath-1
US6100061A (en) 1997-06-20 2000-08-08 Immuno Aktiengesellschaft Recombinant cell clone having increased stability in serum- and protein-free medium and a method of recovering the stable cell clone and the production of recombinant proteins by using a stable cell clone
WO2001051615A1 (en) 2000-01-12 2001-07-19 Hypoxi Co. Ltd. Method for increasing survival rate of cells in animal cell culture under hypoxia condition
US20040259838A1 (en) 2001-12-11 2004-12-23 Joyce Joseph G. Staphylococcus aureus exopolysaccharide and process
USRE40070E1 (en) 1994-02-22 2008-02-19 Smithkline Beecham Corporation Antibody purification

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE65933C (en) Rheinische Gesellschaft für metall Valve device for alcohol stoves
ZA833174B (en) * 1982-05-05 1984-08-29 Genentech Inc Human tissue plasminogen activator
US4677704A (en) * 1986-04-22 1987-07-07 Huggins Richard A Cleaning system for static charged semiconductor wafer surface
JPS63196268A (en) * 1987-02-10 1988-08-15 Kanegafuchi Chem Ind Co Ltd Transformant cell capable of being subjected to passage multiplication in serum-free medium, breeding thereof and production of protein by said cell
JPH0322972A (en) * 1989-01-12 1991-01-31 Ajinomoto Co Inc Serum-free culture medium

Patent Citations (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4205126A (en) 1978-01-01 1980-05-27 Cartaya Oscar A Serum-free cell culture media
US4657866A (en) 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
FR2543158A1 (en) 1983-03-24 1984-09-28 Inst Nat Sante Rech Med CULTURE MEDIUM OF ANIMAL CELLS WITHOUT SERUM, WITHOUT HORMONES AND WITHOUT GROWTH FACTORS AND METHODS OF PRIMARY CULTURE AND OBTAINING CELL LINES USING THE SAME
US4767704A (en) 1983-10-07 1988-08-30 Columbia University In The City Of New York Protein-free culture medium
EP0164813A2 (en) 1984-06-14 1985-12-18 Teijin Limited Method of cultivating animal or plant cells
JPS6125480A (en) 1984-07-13 1986-02-04 Nitsusui Seiyaku Kk Serum-free synthetic medium for cell culture
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
WO1987000195A1 (en) 1985-06-28 1987-01-15 Celltech Limited Animal cell culture
WO1987001131A1 (en) 1985-08-19 1987-02-26 Gene Labs, Inc. Non-human primate monoclonal antibodies and methods
EP0229530A2 (en) 1985-12-31 1987-07-22 Bass Public Limited Company The propagation of yeast, fermentation employing that yeast and products of that fermentation
EP0239400A2 (en) 1986-03-27 1987-09-30 Medical Research Council Recombinant antibodies and methods for their production
US5019499A (en) 1986-04-14 1991-05-28 Daiichi Seiyaku Co., Ltd. Method of producing peptides by transforming myeloma cells with a recombinant plasmid
EP0248656A2 (en) 1986-06-04 1987-12-09 Director-General of the Agency of Industrial Science and Technology Composition for cell cultivation and use thereof
JPS637780A (en) 1986-06-28 1988-01-13 Nippon Zenyaku Kogyo Kk Feeding method for iron to cell and serum-free synthetic culture medium used thereof
WO1988000967A1 (en) 1986-08-04 1988-02-11 The University Of New South Wales Serum free tissue culture medium containing polymeric cell-protective agent
GB2196348A (en) 1986-10-03 1988-04-27 Ceskoslovenska Akademie Ved Synthetic medium for hybridoma and myeloma cell cultivation
US5045468A (en) 1986-12-12 1991-09-03 Cell Enterprises, Inc. Protein-free culture medium which promotes hybridoma growth
EP0274394A2 (en) 1987-01-08 1988-07-13 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
JPS63192381A (en) 1987-02-05 1988-08-09 Kanegafuchi Chem Ind Co Ltd Cell capable of subcultivation in serum-free culture medium and method for obtaining said cell
US5232848A (en) 1987-03-24 1993-08-03 W. R. Grace & Co.-Conn. Basal nutrient medium for cell culture
WO1989000999A1 (en) 1987-07-24 1989-02-09 International Genetic Engineering, Inc. Modular assembly of antibody genes, antibodies prepared thereby and use
EP0307247A2 (en) 1987-09-11 1989-03-15 Genentech, Inc. A method for culturing recombinant cells
EP0307248A2 (en) 1987-09-11 1989-03-15 Genentech, Inc. Method for increasing the expression of polypeptides in recombinant cell culture
EP0316068A1 (en) 1987-10-09 1989-05-17 Collaborative Research Inc. Modified low molecular weight plasminogen activator and method of preparation
EP0314161A1 (en) 1987-10-28 1989-05-03 Bristol-Myers Squibb Company Human immunoglobulines produced by recombinant DNA techniques
EP0325190A2 (en) 1988-01-18 1989-07-26 Roche Diagnostics GmbH Pentosansulfate medium
US5063157A (en) 1988-01-18 1991-11-05 Boehringer Mannheim Gmbh Serum-free culture medium for mammalian cells
EP0328404A1 (en) 1988-02-12 1989-08-16 Btg International Limited Modified antibodies
US5846534A (en) 1988-02-12 1998-12-08 British Technology Group Limited Antibodies to the antigen campath-1
WO1990003429A1 (en) 1988-09-23 1990-04-05 Cetus Corporation Lipid microemulsions for culture media
EP0363703A1 (en) 1988-09-23 1990-04-18 Kenneth Alonso Method of producing human-human hybridomas, the production of monoclonal and polyclonal antibodies therefrom, and therapeutic use thereof
US4929706A (en) 1988-11-02 1990-05-29 W. R. Grace & Co.-Conn. Cell growth enhancers and/or antibody production stimulators comprising chemically modified hydrophilic polyurea-urethane prepolymers and polymers
EP0390327A2 (en) 1989-02-27 1990-10-03 Eli Lilly And Company improved tissue culture method
EP0389786A1 (en) 1989-03-03 1990-10-03 W.R. Grace & Co.-Conn. Very low protein nutrient medium for cell culture
US5135866A (en) 1989-03-03 1992-08-04 W. R. Grace & Co.-Conn. Very low protein nutrient medium for cell culture
EP0388151A1 (en) 1989-03-13 1990-09-19 Celltech Limited Modified antibodies
EP0404003A2 (en) 1989-06-19 1990-12-27 Xoma Corporation Chimeric mouse-human KM10 antibody with specificity to a human tumor cell antigen
WO1991004336A1 (en) 1989-09-19 1991-04-04 Centocor, Inc. Method for improving human monoclonal antibody production
WO1991010722A2 (en) 1989-12-27 1991-07-25 Centocor, Inc. Chimeric immunoglobulin for cd4 receptors
US5122469A (en) * 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
WO1992007084A1 (en) 1990-10-17 1992-04-30 The Wellcome Foundation Limited Purified cdw52-specific antibodies
US5545405A (en) 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal suffering from cancer with a cho-glycosylated antibody
EP0481790A2 (en) 1990-10-17 1992-04-22 The Wellcome Foundation Limited Antibody production
EP0822255A2 (en) 1990-10-17 1998-02-04 The Wellcome Foundation Limited CHO (Chinese Hamster Ovary) glycosylated antibodies and their use in therapy
EP0481791A2 (en) 1990-10-17 1992-04-22 The Wellcome Foundation Limited Culture medium for CHO-cells and adapted CHO-cells
US5316938A (en) 1990-10-17 1994-05-31 Burroughs Wellcome Co. Defined media for serum-free tissue culture
US5633162A (en) 1990-10-17 1997-05-27 Glaxo Wellcome Inc. Method for culturing Chinese hamster ovary cells
US5545404A (en) 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal suffering from a T-cell medicated disorder with a CHO-Glycosylated antibody
US5545403A (en) 1990-10-17 1996-08-13 Burroughs Wellcome Co. Method for treating a mammal by administering a CHO-glycosylated antibody
EP0513738A2 (en) 1991-05-14 1992-11-19 Roche Diagnostics GmbH Serum-free medium for mammalian cells cultivation
EP0523949A1 (en) 1991-07-15 1993-01-20 The Wellcome Foundation Limited Production of antibodies
US5876961A (en) 1991-07-15 1999-03-02 Glaxo Wellcome Inc. Production of antibodies
WO1993002108A1 (en) 1991-07-25 1993-02-04 Idec Pharmaceuticals Corporation Recombinant antibodies for human therapy
EP0610447A1 (en) 1991-10-15 1994-08-17 The Wellcome Foundation Limited CDw52 - SPECIFIC ANTIBODY FOR TREATMENT OF T-CELL MEDIATED INFLAMMATION OF THE JOINTS
WO1993007899A1 (en) 1991-10-15 1993-04-29 The Wellcome Foundation Limited CDw52 - SPECIFIC ANTIBODY FOR TREATMENT OF T-CELL MEDIATED INFLAMMATION OF THE JOINTS
USRE40070E1 (en) 1994-02-22 2008-02-19 Smithkline Beecham Corporation Antibody purification
US6100061A (en) 1997-06-20 2000-08-08 Immuno Aktiengesellschaft Recombinant cell clone having increased stability in serum- and protein-free medium and a method of recovering the stable cell clone and the production of recombinant proteins by using a stable cell clone
WO2001051615A1 (en) 2000-01-12 2001-07-19 Hypoxi Co. Ltd. Method for increasing survival rate of cells in animal cell culture under hypoxia condition
US20040259838A1 (en) 2001-12-11 2004-12-23 Joyce Joseph G. Staphylococcus aureus exopolysaccharide and process

Non-Patent Citations (231)

* Cited by examiner, † Cited by third party
Title
1990 GIBCO BRL Catalogue & Refernce Guide (confirmation of availability attached).
Aathoon and Birch, Science, (1986) 232: 1390-1395.
Abstract of the USPTO trademark database regarding the trademark for Nucellin of Eli Lilly.
Ahmed, S. (2001) "Eating Human Hair by Another Name?," www.albalagh.net/hala/col2.shtml.
Ahrens, et al., Post Graduate Medicine vol. 80, pp. 181-187 (1988).
Alfred hahn's CV submitted in Opposition of EP0523949Email of Jan. 11, 2007 from Ingrid Haas submitted in Opposition of EP0523949.
Alsmadi et al. "Antibody-Dependant Cellular Cytotoxicity Directed against Cells Expreesing Human Immunodeficiency Virus Type 1 Envelope of Primary or Laboratory-Adapted Strains by Human and Chimpanzee Monoclonal Antibodies of Different Eptope Specificities", Journal of Virology, 72(1),:286-293 (1998).
Anthony Lubinicki, ESACT 9th Meeting, Editors Spier R.E. et al., pp. 85-92 (1989).
Barnes, Biotechniques, (1987) vol. 5, No. 6, pp. 534-542.
Bartholomew, et al. "functional analysis of the effects of a fully humanized anti-Cd4 antibody on resting and activated human T cells"Immunology 85:41-48 (1995).
Bebbington et al., Methods: A companion to Methods in Enzymology, vol. 2(2) pp. 136-145 (1991) Abstract.
Biech Z., reprinted with permission from MK News and Views, vol. IV (6) 2003.
Bobbington, et al., Biotechnology, vol. 10 pp. 169-175 (1992).
Bohak, et al., Biopolymers, (1987) 26:s205-213.
Broad, et al., In Vitro Cellular and Developmental Biology, (1986) 22 (2):66-74.
Brogden, et al., Drugs, vol. 34 pp. 350-371 (1987).
Brown et al., Emerging Infectious Diseases, vol. 7 (1) pp. 6-16 (2001).
Buck, et al., J. Virol. Meth., (1985) 10:171-184.
Bulens, et al., Uer. J. Biochem., (1991) 195:235-242.
Bullied, et al., Biochem Journal, vol. 268 pp. 777-781 (1990). *
C6852, Biochemicals and Reagents for Life Science Research, p. 600 (2002-2003) Sigma-Aldrich Company (Current website information also included).
C7880, Biochemicals and Reagents for Life Science Research, p. 600 (2002-2003) Sigma-Aldrich Company (Current website information also included).
C8503, Biochemicals and Reagents for Life Science Research, p. 508 (2002-2003) Sigma-Aldrich Company (Current website information also included).
Carter et al., PNAS, vol. 89 pp. 4285-4289 (May 1992).
Castro, et al., Bitoech Appl. BioChem (1985) 21:57-100.
Chandler, Josseph P., Cultivation of mammalian cells in serum-free medium, American Biotechology Journal 8(1), pp. 18-28, Jan. 1990.
Chotigeat, et al., Cytotechnology (1994) 15:217-221.
Cini, et al., "Molecular Basic for the Isozymes of bovine Glucose-6-Phosphate Isomerase," Arch Biochem. Biophys. 263(1) 96-106.
Colcher et al., Cancer Research, vol. 49 pp. 1738-1745 (1989).
Colomb, et al., Boochem. J., vol. 145 pp. 177-183 (1985).
Curling, Biochem. J., (1990) 272:333-337.
Curling, et al., Biochem Journal, vol. 272 pp. 333-337 (1990). *
Dafler, F., In Vitro Cell Dev. Bio., vol. 26 pp 769-778 (1990).
Darfler, In Vitro Cell. Dev. Bio., Vo 26 pp. 779-783 (1990).
Decision of Appeal of the European Patent Office dated Jan. 15, 2004 T0962/98—3.3.1 Appl. No. 94931395.1.
Decision of Boards of Appeal of the European Patent Office date Jan. 18, 2005 T0278/03—3.3.8 Appl. No. 91309595.6.
Decision of Boards of Appeal of The European Patent Office dated Aug. 26, 2005 T0039/03—3.4.2 Appl. No. 99100131.4.
Decision of Boards of Appeal of The European Patent Office dated Sep. 23, 2004 T0720/02—3.4.2 Appl. No. 97200957.5.
Decision of Technical Boards of Appeal 3.4.2 dated Jul. 13, 2004 T1158/01—3.4.2 Appl. No. 99126075.3.
Declaration of Dr. J Adair submitted in Opposition of EP0523949.
DeCourcy et al., Experimental Cell Research, vol. 192 pp. 52-60 (1991).
Deeds, et al., "Creating a New Medium to Meet the Variable Nutritional Requirements of Chinese Hamster Ovary (CHO) Cell Clones." Sigma Aldrich.com http://www.sigmaaldrich.com/sigma/general (Jul. 2003).
DeWaele et al., European Journal of Biochemistry, vol. 176 (2-3) pp. 287-295 (1988).
DiAugustine, et al. "Evidence of Isoaspartyl (Deamidated) Forms of Mouse Epidermal growth Factor." Anal. Biochem. (1987) 165(2):420-429.
Dickman, Nature, vol. 329 p. 93 (1987).
Dijkmans, et al., Journal of Biological Chemistry, vol. 262(6) pp. 2528-2535 (1987). *
Dulbecco and Freeman, Virology, vol 8 pp. 396-397 (1959) [composition of DMEM attached].
Dyer et al., Blood, vol. 73 pp. 1431-1439 (1989).
Dyer, et al., Leukemia and Lymophoma, (1989) 1:179-193.
Eagle, Science, vol. 130 pp. 432-437 (1959) [composition of MEM attached].
Ebert, Expression of Antibody C-DNA in CHO ("Chinese hamstet ovary") Cells, Dissertation Completed at the Institute for Applied Microbiology University for Soil Cultivation , Feb. 1991 (with Translation).
Ehrlich et al., Human Antibod. Hybridomas, vol. 1(1) pp. 23-26 (1990).
Ehrlich et al., Molecular Immunology, vol. 28(4-5) pp. 319-322 (1991).
E-mail from Diane Fedyk in respect of GIBCO catalogue.
English translation of Dissertation of Veronica Ebert Expression of Antibody c-DNA in CHO (Chinese Hamster Ovary) Cells University for Soil Conservation,Vienna Austria (1991).
Enlarged version of figure from Hamilton et al., In Vitro, vol. 13 (9) pp. 537-547 (1977.
EPO Communication in Opposition against European Patent No. 02 003 143.1, dated Jul. 21, 2010.
Examination Report of Apr. 1, 2005.
Expert declaration of Dr. Florian Ruker submitted in Opposition of EP0523949.
Feldman, G., (Oct. 2001), "Amino Acid Production and the Associated Theoretical Risk of BSE Transmission from their Use in the Production of Biologicals, Drugs, and Medical Devices," FDA TSA Advisory Committee Meeting <www.fda.gov/Ohrms/dockets/ac/01/slides/.
Felgenhauer, et al. "Nucleotide sequences of the cDNAs encoding the V-regions of H-and L-chains for a human monocalonal antibody specific to HIV-1-gp41," Nucleic Acids Research (1990) 18(16) 4927.
Feys et al, Int J Cancer, 1988, Supplement 2, pp. 26-27. *
Feys et al., International Journal of Cancer, vol. 2 pp. 26-27 (1988).
Feys, et al., Chemical Abstracts, vol. 108 (23) p. 514 (1988).
Forthal et al., "Antibody from Patients with Acute Human Immunodefciancy Virus (HIV) Infection Inhibitis Primary Strains of HIV in the presence of Natural-Killer Effector Cell", Journal of Virology, 75(15):6953-6961 (2001).
Fouser, et al., Biotechnology, vol. 10 pp. 1121-1127 (1992).
Freshney, Culture of Animal Cells, a Manual of Basic Technique, pp. 71, 74. and 119-128, 1992.
Freshney, Culture of Animal Cells, Second Edition, Wiley-Liss pp. 70-84 (1989).
Freshney, R.I. (1988). Culture of Animal Cells. Alan R. Liss. New York USA, pp. 70-83. *
Furukawa and Kotaba, Molecular Immunology, vol. 28 (12) pp. 133-1340 (1991). *
Gaboriau, et al., Biochemical Pharmacology, vol. 67 pp. 1627-1637 (2004).
Gardner-Lane et al, Decision on Preliminary Motions, Glaxo Welcome Inc v. Cabilly et al, 2002.
Gasser et al., In Vitro Cellular Development Biology, vol. 21 (10) pp. 588-592 (1985).
Gillies et al., Biotechnology, vol. 7 pp. 799-804 (1989).
Gillies et al., J. Imminological Methods, vol. 125 pp. 191-202 (1989).
Goeddel et al., Proceedings of the National Academy of Sciences USA, vol. 76 (1) pp. 106-110 (1979).
Grady, et al., Journal of Biological Chemistry, vol. 284(34) pp. 20221-20220 (1989).
Hale et al., Journal of Immunological Methods, vol. 103 pp. 59-67 (1987).
Hale et al., Mol. Biol. Med., vol. 1 pp. 305-319 (1983).
Hale et al., The Lancet, vol. 2 pp. 1394-1399 (1988).
Hale et al., Tissue Antigens, vol. 35 pp. 118-127 (1990).
Hale et al., Transplantation, vol. 45 pp. 753-759 (1988).
Hale, et al., The Lancet, vol. 332(8625) pp. 1394-1399 (1988).
Ham, Proceedings of the National Academy of Sciences, vol. 53 pp. 288-293 (1965) [composition of F12 attached].
Hamilton et al, in Vitro, 1977, vol. 13, No. 9, pp. 537-547. *
Handa-Corrigan et al., Enzyme Microbial Technology, vol. 11 ppl. 230-235 (1989).
Hata, et al., Cytotechnology (1992) vol. 10: pp. 9-14.
Haynes, et al., Nucleic Acids Res., 11, 687-706 (1983).
Hayter et al. "Recombinant Gamma-Interferon Production by CHO cells in Serum-free Medium." Advances in Cell Biology and Technology Bioprocessing. RE Spier et al (eds). Butterworths: Kent, England, 1989, pp. 280-282. *
Higuchi, K., Advances Applied Microbiology, vol. 16 pp. 111-136 (1973).
Holtta et al., Biochemica et Biophysica Acta, vol. 721 pp. 321-327 (1982).
Huang and Gorman, "The Simina Virus 40 Small-t Intron, Present in May Common Expression Vectors, Lead to Aberrant Splicing," Molecular and Cellular Biology (1990) 10(4):1805-1810.
I5500, Biochemicals and Reagents for Life Science Research, p. 1147 (2002-2003) Sigma-Aldrich Company (Current website information also included).
Isaacs et al., The Lancet, vol., 340 (8822) pp. 748-752 (1992).
Iscove and Melchers, The Journal of Experimental Medicine, vol. 147 pp. 923-933 (1978).
J. Cell. Biochem, 20th Annual Meeting (1991):122.
Jefferies. "Structure Function relationship in Human Immunoglobulans" neth J Med (1991), vol. 39, No. 3-4, pp. 188-198. Abstract only.
Jenkins, et al., Nature Biotechnology, (1996) 14:975-980.
Jungbauger, et al., Pilot scale production of a human monoclonal antibody against human immunodeficiency virus HIV-1, Journal of Biological and Biophysical Methods. (1989) 19:223-240.
K. Loren, Vibrant Life, vol. 2 (1) 13 pgs (1999).
Kalwy, S., et al., "Toward More Efficient Protein Expression," Molecular Biotechnology (2006) vol. 34 p. 151.
Kaqawa et al., Journal of Biochemistry, vol. 68 pp. 133-136 (1970).
Katsua & Takaoka, Methods of Cell Biology, vol. 6 pp. 1-42 (1973).
Katsuta and Takaoka, Journal of Experimental Medicine, vol. 30 pp. 235-259 (1960).
Katsuta and Takaoka, Methods of Cell Biology, vol. 6 pp. 1-42 (1973).
Kaufman et al., Molec. Cell Biol., vol. 5(7) pp. 1750-1759 (1985).
Kaufman, J. Mol. Biol., 159, 601-621 (1982).
Kaufman, R.J. et al. (1985), "Coamplification and coexpression of human tissue-type plasminogen . . . " Molec. Cell Biol. 5(7):1750-59. *
Keay, Biotechnology and Bioengineering, vol. XVIII pp. 363-382 (1976).
Kilburn, et al. Biotechnology and Bioengineering (1968) vol. X:801-814.
Kim, et al., In Vitro Cell Dev. Biology, vol. 38 pp. 314-319 (2002).
King, et al., Biochem Journal, vol. 281 pp. 317-323 (1992).
Knight et al., Human Antibody Hybridomas, vol. 3 pp. 129-136 (1992).
Köhrle, J., Biochimie, vol. 81 pp. 527-533 (1999).
Kunert, et al. "Molecular Characteristics of Five Neutralizinf Anti-HIV Type I Antibodies: Indentification of Nonconventional D. Segments in the Human Monoclonal Antibodies 2G12 and 2F5" Aids Research 14(3):1115-1128 (1998).
Kurano, et al., Journal of Biotechnology, vol. 15 pp. 101-112 (1990).
Kyle et al., Journal of Rheumatology, vol. 18 (11) pp. 1737-1738 (1991).
Large Scale Mammalian Cell Culturetechnology, published by Marcel Dekker, Inc., and edited by A.S. Lubiniecki (pp. 161-175, 417-449 and 515-541; Sep. 14, 1990).
Larrick, et al., Biotechnology, vol. 7 pp. 934-938 (1989).
Leatherbarrow, et al., Imuunology (1985) 22(4):401-413.
Letter form head librarian of Boku submitted in Opposition of EP0523949.
Letter form head librarian to "whom it may concern" submitted in Opposition of EP0523949.
Levy et al., Gene, vol. 54 pp. 167-173 (1987).
Lewis et al., Human Antibody Hybridomas, vol. 3 pp. 146-152 (1992).
Lipoldova, "T-cell receptor V beta 5 usage defines reactivity to9 a human T-call receptor monoclonal antobody," Immunogenetics (1989) 30(3):162-8—abstract.
Liu et al., J. Immunology, vol. 13(10) pp. 3521-3528 (1987).
Liu, et al., Gene, vol. 54(1) pp. 33-40 (1987).
Liu, et al., PNAS, vol 84(10) pp. 3439-3433 (1987).
Luff, cited in "The BSE Inquiry," established fot the British Government.
Madisen, et al., Growth Factors (1990) 3:129-138.
Marquis et al., Cytotechnology vol. 2 pp. 163-170 (1989).
Mather, et al., Methods of Enzymology, vol. 185 pp. 567-577 (1995).
McCormick et al., Molec. Cell Biol., vol. 4 (1) pp. 166-172 (1984).
McCormick, F., et al. (1984), "Inducible expression of amplified human beta . . . " Molec. Cell Biol. (4(1): 166-172. *
McKeehan et al., Proceedings of the National Academy of Sciences USA, vol. 73 pp. 2023-2027 (1976).
Mendiaz et al., In Vitro Cell. Dev. Biol., vol. 22 pp. 66-74 (1986).
Meri & Bonsdorff, ency. Of Imm., Delves & Roidd (eds), (1998) pp. 617-619.
Merten et al., Production of biologicals from animal cells in culture research, development, and achievements, 10th Mfg., Avignon, France (1990).
Mesa, et al., J. Interferon and Cytokine Res. (1995) 15:309-315.
Michel, et al., Proceedings of the National Academy of Sciences, vol. 81 (24) pp. 7708-7712 (1984).
Miyaji, et al., Expression of Human beta-interferon in Namalwa KJM-1 which was adapted to a serum-free medium., Cytotechnology vol. 3, pp. 133-140, 1990.
Miyazaki, et al. "Production of Monoclonal antibodies against human erythropoietin and their use in purification of human urinary erythropoietin" J. Immunol Methods. (Oct. 1988) 26:113(2)261-7 Abstract.
Mizouchi, et al., Biochem Journal, vol. 254 pp. 599-603 (1998). *
Moellering, et al., "Electrophoretic Differences in Mab Expressed in Three Media." BioPharm. (Feb. 1990) 30-38.
Morrison et al., PNAS, vol. 81 pp. 6851-6855 (1984).
Morrison, "Transfection Provide Novel Chimeric Antibodies," Science (1985) 229:1202-1205.
Morrison, "Trasfection Provide Novel Chimeric Antibodies," Science (1985) 229:1202-1205.
Mountain and Adair, Biotechnology and Genetic Engineering Reviews, vol. 10 pp. 1-142 (1992).
Murphy, Science, vol. 273 (5276 pp. 746-747 (1996).
Nakamori et al., Applied and Environmental Microbiology, vol. 64 (5) pp. 1607-1611 (1998).
Neidhardt, et al., J. Bacteriol., 119, No. 3, 3, 736-747 (1947).
Neuberger et al., Nucleic Acids Research, vol. 16(14) pp. 6713-6724 (1988).
Newman, et al., Biotechnology, vol. 10 pp. 1455-1460 (1992).
Nippon Zenyaku Industries, Reply of Applicant concerning EP92306420.8, May 28, 1998.
Nishimura, et al., Cancer research (1987) 47:999-1005.
Noda et al., Chem. Abs., vol. 110 (19) Abstract p. 652 (1988).
Ogata et al., Applied Mcrobiology Biotechnology, vol. 38 (4) pp. 520-525 (1993).
Oka et al., Bioprogress Technol., vol. 10 pp. 72-92 (1990).
Opposition Division's Decision of revocation in respect of parent case (EP-B-0481791).
Opposition Division's Preliminary Opinion in respect of parent case (EP-B-0481791).
Organic Chemistry, John Wiley & Sons Inc, vol. II Second Edition pp. 1129-1130 and 1136-1138 (1943).
Page and Sydenham, Biotechnology, vol. 21 (10) pp. 64-68 (1991).
Page et al., Biotechnology, vol. 9 pp. 64-68 (1991).
Painter, Encyclopedia Of Immunology, Second Ed. Delves et al., (1998):1208-1211.
Parekh, et al., Nature 316:452-457 (1985).
Patel, et al. "Different culture methods lead to differences in mrine IgG monoclonal antibody" Biochem J. 285, 839-845 (1992).
Patterson, et al., 1994: Appl Microbial Biotechnologiy 40: p. 691-698.
Paulson and Colley, Journal of Biological Chemistry, vol. 264(30) pp. 17615-17618 (1989). *
Paulson, TIBS Jul. 14, 1989. *
Pearson et al., The 19th Meeting of the European Society for Animal Cell Technology, pp. 1-11 (2005).
Persson et al. PNAS, vol. 88 pp. 2432-2436 (1991).
Phillips, et al., Cytotherapy (2001) 3 (3):233-242.
Phillpotts, Cytotechnology, vol. 2 pp. 161-162 (1989).
Prokop, et al., Editors, Annals of the New York Academic of Sciences, 646:Table of Contents and Prefaces, Dec. 27, 1991.
R.H. Kimberlin, Symposium of Virological Aspects of the Safety of Biological Products London, England 1990, Develop. Biol. Standard, vol. 75 pp. 75-82 (1991).
Rabbi S. Emanuel, MK Vaad Hair, vol. IV (7) 9 pgs (2003) .
Rabbi S. Emanuel, MK Vaad Hair, vol. IV (7) 9 pgs (2003) <www.mk.ca/page6_11.php>.
Ray, et al., ann. NY Aca Sci., (1990) 589:443-457.
Regenstein et al., E-Journal , Kosher Issues for Today's Dairy Industry, 2002.
Regenstein et al., E-Journal <www.Kashrut.com>, Kosher Issues for Today's Dairy Industry, 2002.
Response to Examination Report of Apr. 1, 2005.
Riechmann et al., Nature, vol. 322, pp. 323-327 (1988).
Ripka, et al., Archives of Biochemistry and Biophysics (Sep. 1986) 249(2):533-545.
Roberts, et al. J.Cell Biochem 20th Annual Meeting p. 122 (2001).
Robinson et al., Human Antibody Hybridomas, vol. 2 pp. 84-92 (1991).
Roitt Brostoff and Male, Immunology 6th edition, p. 74 (2001).
Rose et al., Molecular Immunology, vol. 29(1) pp. 131-144 (1992).
Rudd, et al., Molecular Immunology, vol. 28(12) pp. 1369-1378 (1991). *
Rüker, et al., Annals New York Acad. Sci., pp. 212-219 (1991).
Saban, T., (2004) "Food Additives From Islamic Perspective," Version 1.3 .
Saban, T., (2004) "Food Additives From Islamic Perspective," Version 1.3 <wwwsrvl.mycity.at/privat.9704236/Im/LM-en.html>.
Sakar, et al., Proceedings of the National Academy of Science, vol. 92 pp. 3323-3327 (1995).
Salahuddin et al., Journal of Experimental Medicine, vol. 155 pp. 1842-1857 (1982).
Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition pp. 8.8-8.9 (1989).
Sano et al., Cell Struct. Funct., vol. 13 (2) pp. 142-159 (1988).
Sano et al., Cell Structure and Function, vol. 13 (2) pp. 143-159 (1988).
Scanhill et al., Proceedings of the National Academy of Sciences, vol. 80 pp. 4654-4658 (1983).
Schneider & Laviox, Journal of Immunological Methods, vol. 129 pp. 251-268 (1990).
Schnider, Journal of Immunological Methods, vol. 116 pp. 65-77 (1989).
Sertich, et al., Journal of Cellular Physiology, vol. 127 pp. 114-120 (1986).
Sims, et al. "A Humanised CD18 Antibody can Block Fucntion without Cell Destruction" J. Immumol. 151:2296-2309 (1993).
Spellman, et al., Journal of Biological Chemistry, vol. 264(24) pp. 14100-14111 (1989). *
Spira et al., Trends in Animal Cell Culture Technology, Proc. Ann. Meeting Jpn. Tech., pp. 67-73 (1990) [Reporting conference meeting in 1989].
Takagi, et al., Journal of Bioscience and Bioengineering, vol. 90(5) pp. 509-514 (2001).
Taylor et al., Mutation Research, vol. 67 pp. 65-80 (1979).
Thilly, et al., Mammalian Cell Technology, pp. 26-29 (1986).
Titeux et al., Journal of Cellular Physiology, vol. 121 pp. 251-256 (1984).
Tsujimoto et al., Journal of Biochemistry, vol. 106 pp. 23-28 (1989).
U.S. Appl. No. 10/145,712, filed May 16, 2002, Page, et al.
U.S. Appl. No. 10/145,992, filed May 16, 2002, Page, et al.
U.S. Appl. No. 10/765,067, filed Jan. 28, 2004, Page, et al.
U.S. Appl. No. 11/289,714, filed Nov. 29, 2005, Crowe, et al.
U.S. Appl. No. 11/804,729, filed May 18, 2007, Shadle, et al.
U.S. Appl. No. 90/006,997, filed Apr. 5, 2004, Crowe, et al.
Ungemach et al., The 50th Meeting of the joint FAO/WHO Expert Committee on Food Additives (JECFA), World Health Organization, 1998.
Urban & Chasin, Proceedings of the National Academy of Sciences, vol. 77(7) pp. 4216-4220 (1980).
Urblan, G and L.A. Chasin (1980). "Isolation of Chinese hamster cell mutants . . . " Proc. Natl. Acad. Sci. USA 77(7):4216-20. *
Urlaub et al, Proceedings of the National Academy of Sciences of The United States of America, 1980, vol. 77, No. 7, pp. 4216-3220. *
Waldman, et al. "A Clonal Derivative of Tunicamysin-resiistant Chinese Hamster Ovary Cells with Increased N-Acetylglucosamine-Phosphate Transferase Activity Has Altered Asparagine-Linked Glycosylation" J. Cell. Physio. 131 pp. 302-317 (1987).
Weber et al., Journal of Neuroimmunology, vol. 22 pp. 1 to 9 (1989).
Weidle et al., Gene, vol. 51 pp. 21-29 (1987).
Weidle et al., Gene, vol. 60 pp. 205-216 (1987).
Whitaker et al., Biopharm, vol. 3 (8) p. 5 (Sep. 1990).
Whittle et al., Protein Eng., vol. 1 pp. 499-505 (1987).
Wiebe et al., ESACT 9th Meeting, Editors Spier R.E. et al., pp. 68-71 (1989).
Wood, et al, Journal of Immunology, vol. 145 pp. 3011-3016 (1990).
Yan, et al., TIBS Jul. 14, 1989. *
Yang et al., Proceedings of the National Academy of Sciences USA, vol. 81 pp. 2752-2756 (1984).
Zekauskas et al., J. Okla. State Med. Assoc. vol. 83 pp. 447-448 (1990).
Zettlemeissl et al., Biotechnology, vol. 5 pp. 720-725 (1987).
Zettlemeissl, et al., Journal of Biological Chemistry, vol. 264 (35) pp. 21153-21159 (1989).

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110008464A1 (en) * 2009-07-10 2011-01-13 Scott Iii Linzy O Methods and compositions for treating thyroid-related medical conditions with reduced folates
US8343974B2 (en) 2009-07-10 2013-01-01 Scott Iii Linzy O Methods and compositions for treating thyroid-related medical conditions with reduced folates
US8575171B2 (en) 2009-07-10 2013-11-05 Linzy O. Scott, III Methods and compositions for treating thyroid-related medical conditions with reduced folates
US9248130B2 (en) 2009-07-10 2016-02-02 Linzy O. Scott, III Methods and compositions for treating thyroid-related medical conditions with reduced folates
US9481861B2 (en) 2011-07-12 2016-11-01 Foodchek Systems, Inc. Culture medium, method for culturing Salmonella and E. coli and method for detecting Salmonella and E. coli
WO2013006960A1 (en) * 2011-07-13 2013-01-17 Foodchek Systems, Inc. Culture medium, method for culturing listeria, and method for detecting listeria
GB2497059A (en) * 2011-07-13 2013-05-29 Foodchek Systems Inc Culture medium,method for culturing listeria, and method for detecting listeria
AU2012283715B2 (en) * 2011-07-13 2015-09-17 Foodchek Systems, Inc. Culture medium, method for culturing Listeria, and method for detecting Listeria
GB2497059B (en) * 2011-07-13 2016-06-22 Foodchek Systems Inc Culture medium, method for culturing listeria,and method for dtecting listeria

Also Published As

Publication number Publication date
EP0481791B1 (en) 2003-08-27
US5316938A (en) 1994-05-31
DE69133303T2 (en) 2004-06-24
AU8591591A (en) 1992-05-07
NZ240248A (en) 1994-11-25
EP1221476A3 (en) 2003-09-17
ATE382680T1 (en) 2008-01-15
US5633162A (en) 1997-05-27
EP1849862A3 (en) 2008-02-13
JPH0670757A (en) 1994-03-15
EP1221476A2 (en) 2002-07-10
CA2053586A1 (en) 1992-04-18
DK1221476T3 (en) 2008-05-13
ES2204885T3 (en) 2004-05-01
DK0481791T3 (en) 2003-12-08
ZA918249B (en) 1993-04-16
GB9022545D0 (en) 1990-11-28
ES2298301T3 (en) 2008-05-16
JP2625302B2 (en) 1997-07-02
DE69133589T2 (en) 2009-01-08
AU645615B2 (en) 1994-01-20
EP1221476B1 (en) 2008-01-02
ATE248217T1 (en) 2003-09-15
EP1849862A2 (en) 2007-10-31
CA2053586C (en) 2003-07-29
EP0481791A2 (en) 1992-04-22
IE913559A1 (en) 1992-04-22
DE69133589D1 (en) 2008-02-14
EP0481791A3 (en) 1992-07-08
USRE39792E1 (en) 2007-08-21
DE69133303D1 (en) 2003-10-02

Similar Documents

Publication Publication Date Title
USRE41974E1 (en) Method for culturing Chinese hamster ovary cells
IE84174B1 (en) Culture medium for CHO cells and adapted CHO cells
US20060115901A1 (en) Method and media for single cell serum-free culture of CHO cells
EP1992697B1 (en) Production of TNFR-Fc
EP2357250B1 (en) Production of TNFR-Ig
US9982286B2 (en) Medium for the protein-free and serum-free cultivation of cells
EP1781803B1 (en) Production of anti-amyloid beta antibodies
EP0283942B1 (en) Basal nutrient medium for cell culture
EP1360314B1 (en) Chemically defined medium for cultured mammalian cells
US5122469A (en) Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
US4443546A (en) Process and composition for propagating mammalian cells
WO2000003000A2 (en) Serum-free medium for culturing animal cells
WO2008009642A1 (en) Cell culture media
EP0659880B1 (en) Medium for culturing animal cells or antibody-producing cells
Kim et al. Effects of supplementation of various medium components on Chinese hamster ovary cell cultures producing recombinant antibody
Gu et al. Influence of Primatone RL supplementation on sialylation of recombinant human interferon‐γ produced by Chinese hamster ovary cell culture using serum‐free media
Kim et al. Development of a serum-free medium for the production of humanized antibody from Chinese hamster ovary cells using a statistical design

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLAXOSMITHKLINE LLC, PENNSYLVANIA

Free format text: CHANGE OF NAME;ASSIGNOR:SMITHKLINE BEECHAM CORPORATION;REEL/FRAME:024377/0976

Effective date: 20091027