US20070254358A1 - Culture medium for cell growth and transfection - Google Patents

Culture medium for cell growth and transfection Download PDF

Info

Publication number
US20070254358A1
US20070254358A1 US11/552,783 US55278306A US2007254358A1 US 20070254358 A1 US20070254358 A1 US 20070254358A1 US 55278306 A US55278306 A US 55278306A US 2007254358 A1 US2007254358 A1 US 2007254358A1
Authority
US
United States
Prior art keywords
cells
medium
cell
culture
acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/552,783
Inventor
Valentina CICCARONE
Dale Gruber
Shelly Bennett
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.)
Life Technologies Corp
Original Assignee
Invitrogen Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Invitrogen Corp filed Critical Invitrogen Corp
Priority to US11/552,783 priority Critical patent/US20070254358A1/en
Publication of US20070254358A1 publication Critical patent/US20070254358A1/en
Assigned to BANK OF AMERICA, N.A., AS COLLATERAL AGENT reassignment BANK OF AMERICA, N.A., AS COLLATERAL AGENT SECURITY AGREEMENT Assignors: Life Technologies Corporation
Assigned to Life Technologies Corporation reassignment Life Technologies Corporation LIEN RELEASE Assignors: BANK OF AMERICA, N.A.
Priority to US13/968,157 priority patent/US9879243B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/24Hydrolases (3) acting on glycosyl compounds (3.2)
    • C12N9/2402Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1)
    • C12N9/2468Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1) acting on beta-galactose-glycoside bonds, e.g. carrageenases (3.2.1.83; 3.2.1.157); beta-agarase (3.2.1.81)
    • C12N9/2471Beta-galactosidase (3.2.1.23), i.e. exo-(1-->4)-beta-D-galactanase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • 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
    • 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
    • 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
    • C12N5/005Protein-free medium
    • 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
    • C12N2500/22Zinc; Zn chelators
    • 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
    • C12N2500/24Iron; Fe chelators; Transferrin
    • 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
    • C12N2501/999Small molecules not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production

Definitions

  • the present invention relates to the field of cell culture.
  • the present invention provides media suitable for the culture and transfection of cells.
  • Cell culture media provide the nutrients necessary to maintain and grow cells in a controlled, artificial and in vitro environment. Characteristics and formulations of cell culture media vary depending upon the particular cellular requirements. Important parameters include osmolarity, pH, and nutrient compositions.
  • Typical components of cell culture media include amino acids, organic and inorganic salts, vitamins, trace metals, sugars, lipids and nucleic acids, the types and amounts of which may vary depending upon the particular requirements of a given cell or tissue type.
  • Cultivated cells have many uses including the study of physiological processes and the production of useful biological substances.
  • useful products include monoclonal antibodies, hormones, growth factors, enzymes and the like.
  • Such products have many commercial and therapeutic applications and, with the advent of recombinant DNA technology, cells can be engineered to produce large quantities of these products.
  • Cultured cells are also routinely used for the isolation, identification and growth of viruses which can be used as vectors and/or vaccines.
  • the ability to cultivate cells in vitro is not only important for the study of cell physiology, but is also necessary for the production of useful substances which may not otherwise be obtained by cost-effective means.
  • the epithelium lines the internal and external surfaces of the organs and glands of higher organisms. Because of this localization at the external interface between the environment and the organism (e.g., the skin) or at the internal interface between an organ and the interstitial space (e.g., the intestinal mucosal lining), the epithelium has a major role in the maintenance of homeostasis.
  • the epithelium carries out this function, for example, by regulating transport and permeability of nutrients and wastes (Freshney, R. I., in: Culture of Epithelial Cells , Freshney, R. I., ed., New York: Wiley-Liss, pp. 1-23 (1992)).
  • epithelial cells The cells making up the epithelium are generically termed epithelial cells. These cells can be present in multiple layers as in the skin, or in a single layer as in the lung alveoli. As might be expected, the structure, function and physiology of epithelial cells are often tissue-specific. For example, the epidermal epithelial cells of the skin are organized as stratified squamous epithelium and are primarily involved in forming a protective barrier for the organism, while the secretory epithelial cells of many glands are often found in single layers of cuboidal cells that have a major role in producing secretory proteins and glycoproteins. Regardless of their location or function, however, epithelial cells are usually regenerative.
  • epithelial cells are capable of dividing or growing. This regenerative capacity has facilitated the in vitro manipulation of epithelial cells, to the point where a variety of primary epithelial cells and cell lines have been successfully cultivated in vitro (Freshney, Id.).
  • the human embryonic kidney cell line 293 (“293 cells”), which exhibits epithelial morphology, has proven particularly useful for studies of the expression of exogenous ligand receptors, production of viruses and expression of allogeneic and xenogeneic recombinant proteins.
  • 293 cells human embryonic kidney cell line 293
  • U.S. Pat. No. 5,166,066 describes the construction of a stable 293 cell line comprising functional GABA receptors that include a benzodiazepine binding site that have proven useful in identification and screening of candidate psychoactive drugs.
  • 293 cells have also been used to produce viruses such as natural and recombinant adenoviruses (Garnier, A., et al., Cytotechnol. 15:145-155 (1994); Bout, A., et al., Cancer Gene Therapy 3(6):S24, abs. P-52 (1996); Wang, J.-W., et al., Cancer Gene Therapy 3(6):S24, abs. P-53 (1996)), which can be used for vaccine production or construction of adenovirus vectors for recombinant protein expression.
  • 293 cells have proven useful in large-scale production of a variety of recombinant human proteins (Berg, D.
  • Fibroblasts have been isolated from many different tissues and are understood to be connective tissue cells. It is clearly possible to cultivate cell lines, loosely termed fibroblastic cells, from embryonic and adult tissues. Fibroblasts characteristically have a “spindle” appearance. Fibroblast-like cells have morphological characteristics typical of fibroblast cells. Under a light microscope the cells appear pointed and elongated (“spindle shaped”) when they grow as a monolayer on the surface of a culture vessel. Cell lines can be regarded as fibroblast or fibroblast-like after confirmation with appropriate markers, such as collagen, type I ((Freshney, R. I., in: Culture of Epithelial Cells , Freshney, R. I., ed., New York: Wiley-Liss, pp. 1-23 (1987)).
  • CHO cells have been classified as both epithelial and fibroblast cells derived from the Chinese hamster ovary.
  • a cell line started from Chinese hamster ovary (CHO-K1) (Kao, F.-T. And Puck, T. T., Proc. Natl. Acad. Sci. USA 60:1275-1281 (1968) has been in culture for many years but its identity is still not confirmed.
  • suspension cultures grow in a three-dimensional space.
  • Monolayer cultures in similar-sized vessels can only grow two-dimensionally on the vessel surface.
  • suspension cultures can result in higher cell yields and, correspondingly, higher yields of biologicals (e.g., viruses, recombinant polypeptides, etc.) compared to monolayer cultures.
  • suspension cultures are often easier to feed and scale-up, via simple addition of fresh culture media (dilution subculturing) to the culture vessel rather than trypsinization and centrifugation as is often required with monolayer cultures.
  • fresh culture media diution subculturing
  • centrifugation as is often required with monolayer cultures.
  • the ease of feeding and the ease with which suspension cultures can be scaled up represent a substantial saving in time and labor for handling a comparable number of cells.
  • anchorage-dependent cells such as primary epithelial cells, primary fibroblast cells, epithelial cell lines, and fibroblast cell lines, however, are not easily adapted to suspension culture. Since they are typically dependent upon anchorage to a substrate for optimal growth, growth of these cells in suspension can require their attachment to microcarriers such as latex or collagen beads. Thus, cells grown in this fashion, while capable of higher density culture than traditional monolayer cultures, are still technically attached to a surface; subculturing of these cells therefore requires similar steps as those used for the subculturing of monolayer cultures.
  • cell culture media formulations are supplemented with a range of additives, including undefined components such as fetal bovine serum (FBS) (5-20% v/v) or extracts from animal embryos, organs or glands (0.5-10% v/v). While FBS is the most commonly applied supplement in animal cell culture media, other serum sources are also routinely used, including newborn calf, horse and human. Organs or glands that have been used to prepare extracts for the supplementation of culture media include submaxillary gland (Cohen, S., J. Biol. Chem. 237:1555-1565 (1961)), pituitary (Peehl, D. M., and Ham, R.
  • FBS fetal bovine serum
  • these supplements provide carriers or chelators for labile or water-insoluble nutrients; bind and neutralize toxic moieties; provide hormones and growth factors, protease inhibitors and essential, often unidentified or undefined low molecular weight nutrients; and protect cells from physical stress and damage.
  • serum or organ/gland extracts are commonly used as relatively low-cost supplements to provide an optimal culture medium for the cultivation of animal cells.
  • undefined components such as serum or animal extracts also prevents the true definition and elucidation of the nutritional and hormonal requirements of the cultured cells, thus eliminating the ability to study, in a controlled way, the effect of specific growth factors or nutrients on cell growth and differentiation in culture.
  • undefined supplements prevent the researcher from studying aberrant growth and differentiation and the disease-related changes in cultured cells.
  • serum and organ/gland extract supplementation of culture media can complicate and increase the costs of the purification of the desired substances from the culture media due to nonspecific co-purification of serum or extract proteins.
  • serum-free media can still contain one or more of a variety of animal-derived components, including albumin, fetuin, various hormones and other proteins. The presence of proteins or peptides makes purification of recombinant protein difficult, time-consuming, and expensive.
  • defined culture media Since the components (and concentrations thereof) in such culture media are precisely known, these media are generally referred to as “defined culture media.”
  • defined culture media Sometimes used interchangeably with “defined culture media” is the term “serum-free media” or “SFM.”
  • SFM serum-free media
  • a number of SFM formulations are commercially available, such as those designed to support the culture of endothelial cells, keratinocytes, monocytes/macrophages, lymphocytes, hematopoietic stem cells, fibroblasts, chondrocytes or hepatocytes which are available from Invitrogen Corporation, Carlsbad, Calif.
  • SFM serum and protein fractions
  • other undefined components such as organ/gland extracts.
  • SFM serum and protein fractions
  • keratinocytes Boyce, S. T., and Ham, R. G., J. Invest. Dermatol. 81:33 (1983); Wille, J. J., et al., J. Cell. Physiol. 121:31 (1984); Pittelkow, M. R., and Scott, R. E., Mayo Clin. Proc.
  • defined media generally provide several distinct advantages to the user. For example, the use of defined media facilitates the investigation of the effects of a specific growth factor or other medium component on cellular physiology, which can be masked when the cells are cultivated in serum- or extract-containing media.
  • defined media typically contain much lower quantities of protein (indeed, defined media are often termed “low protein media”) than those containing serum or extracts, rendering purification of biological substances produced by cells cultured in defined media far simpler and less expensive.
  • basal media Some extremely simple defined media, which consist essentially of vitamins, amino acids, organic and inorganic salts and buffers have been used for cell culture. Such media (often called “basal media”), however, are usually seriously deficient in the nutritional content required by most animal cells. Accordingly, most defined media incorporate into the basal media additional components to make the media more nutritionally complex, but to maintain the serum-free and low protein content of the media.
  • bovine serum albumin or human serum albumin (HSA)
  • certain growth factors derived from natural (animal) or recombinant sources such as epidermal growth factor (EGF) or fibroblast growth factor (FGF)
  • lipids such as fatty acids, sterols and phospholipids
  • lipid derivatives and complexes such as phosphoethanolamine, ethanolamine and lipoproteins
  • protein and steroid hormones such as insulin, hydrocortisone and progesterone
  • nucleotide precursors and certain trace elements (reviewed by Waymouth, C., in: Cell Culture Methods for Molecular and Cell Biology, Vol.
  • animal protein supplements in cell culture media, however, also has certain drawbacks. For example, there is a risk that the culture medium and/or products purified from it can be immunogenic, particularly if the supplements are derived from an animal different from the source of the cells to be cultured. If biological substances to be used as therapeutics are purified from such culture media, certain amounts of these immunogenic proteins or peptides can be co-purified and can induce an immunological reaction, up to and including anaphylaxis, in an animal receiving such therapeutics.
  • animal cell culture media that are completely free of animal proteins.
  • some culture media have incorporated extracts of yeast cells into the basal medium (see, for example, U.K. Patent Application No. GB 901673; Keay, L., Biotechnol. Bioengin. 17:745-764 (1975)) to provide sources of nitrogen and other essential nutrients.
  • hydrolysates of wheat gluten have been used, with or without addition of yeast extract, to promote in vitro growth of animal cells (Japanese Patent Application No. JP 2-49579).
  • Transferrin functions in vivo to deliver iron to cells.
  • the mechanism of iron uptake by mammalian cells has been reviewed (Qian, Z. M. and Tang, P. L. (1995) Biochim. Biophys. Acta 1269, 205-214).
  • serum-free media are often supplemented with transferrin in order to deliver the requisite iron for the successful cultivation of most cells in vitro.
  • Simple carriers such as chelating agents (e.g., EDTA) and certain acids or salts thereof (e.g., citrate, picolinate, and derivatives of benzoic acid or hydroxamic acid) have been shown to be useful in certain serum-free growth media (see U.S. Pat. Nos. 5,045,454 and 5,118,513; Testa et al., Brit. J. Haematol. 60:491-502, (1985); Ganeshaguru et al., Biochem. Pharmacol. 29:1275-1279 (1980); White et al., Blood 48:923-929 (1976)).
  • chelating agents e.g., EDTA
  • acids or salts thereof e.g., citrate, picolinate, and derivatives of benzoic acid or hydroxamic acid
  • a critical step in the effective production and purification of biological substances is the introduction of one or more macromolecules (e.g., peptides, proteins, nucleic acids, etc.) into the cell in which the material will be produced. This can be accomplished by a variety of methods.
  • One widely used method to introduce macromolecules into a cell is known as transfection.
  • the target cell is grown to a desired cell density in a cell culture medium optimized for growth of the cell.
  • the medium is exchanged for a medium optimized for the transfection process.
  • the medium used for transfection does not support the growth of the cells but the transfection medium is merely used for the purpose of introducing nucleic acids into the cells.
  • the process generally requires collecting the cells from the culture, usually by centrifugation, washing the cells to remove traces of the growth medium, suspending the cells in a transfection medium in the presence of the macromolecule of interest, incubating the cells in the transfection medium for a period of time sufficient for the uptake of the macromolecule, optionally, removing the transfection medium and washing the remnants of the transfection medium from the cells and then re-suspending the transfected cells in a growth medium.
  • the steps of exchanging the growth media for transfection media, washing the cells, and exchanging the transfection media back to a growth media require a great deal of hands-on manipulation of the cells thereby adding substantially to the time and expense of recombinant DNA technology.
  • 293 cells have been cultivated in monolayer cultures in a serum-supplemented version of a complex medium (i.e., DMEM).
  • DMEM complex medium
  • 293 cells When grown in suspension, 293 cells have a tendency to aggregate into large clusters of cells. The formation of these large cell aggregates reduces the viability of the cells. Since the cells in the center of the aggregates are not directly exposed to the medium, these cells have limited access to nutrients in the medium and have difficulty in exchanging waste into the medium. In addition, this reduced access to the medium makes cells in clusters unsuitable for genetic manipulation by factors introduced into the medium (i.e., for transformation by nucleic acids). As a result of these difficulties, 293 cells have not generally been used in suspension culture for the production of biological materials.
  • Such a medium should preferably be a serum-free and/or chemically defined and/or protein-free medium and/or a medium lacking animal derived materials which facilitates the growth of mammalian cells to high density and/or increases the level of expression of recombinant protein, reduces cell clumping, and which does not require supplementation with animal proteins, such as serum, transferrin, insulin and the like.
  • a medium of this type will permit the suspension cultivation of mammalian cells that are normally anchorage-dependent, including epithelial cells and fibroblast cells, such as 293 cells and CHO cells.
  • mammalian cells that are normally anchorage-dependent, including epithelial cells and fibroblast cells, such as 293 cells and CHO cells.
  • Such culture media will facilitate studies of the effects of growth factors and other stimuli on cellular physiology, will allow easier and more cost-effective production and purification of biological substances (e.g., viruses, recombinant proteins, etc.) produced by cultured mammalian cells in the biotechnology industry, and will provide more consistent results in methods employing the cultivation of mammalian cells.
  • the present invention provides a cell culture medium, wherein the medium supports introduction of one or more macromolecules into at least one eukaryotic cell in culture and supports cultivation of the at least one cell subsequent to the introduction, wherein growth of the at least one cell continues in the medium in the absence of the medium being with fresh medium.
  • the medium supports introduction of one or more macromolecules into at least one eukaryotic cell in culture and supports cultivation of the at least one cell subsequent to the introduction, wherein growth of the at least one cell continues in the medium in the absence of the medium being with fresh medium.
  • growth is accomplished in cultivation in a volume of medium that is about the same volume up to no more than about 10 times the volume of the medium in which the introduction occurred.
  • the present invention also provides a method of making a medium comprising admixing water and at least one ingredient selected from the group consisting of an amino acid, a sugar, a fatty acid (such as linoleic acid, linolenic acid, and especially fatty acids of 12, 14, 16, 18, 19, 20 or 24 carbon atoms, each carbon chain branched or unbranched), arachadonic acid, palmitoleic acid, oleic acid, polyenoic acid (e.g., palmitoleic acid, oleic acid, linoleic acid and/or linolenic acid), a vitamin (such as pyridoxine, niacinamide, thiamine, etc.), a pH buffer, a surfactant, a trace metal or salt or hydrate thereof, an amine compound, a growth factor, an agent to control osmolarity and/or ionic strength and/or maintain membrane potential, a flavin, a compound that participates in or is a
  • the present invention also provides the medium obtained by admixing water and at least one ingredient selected from the group consisting of an amino acid, a sugar, a fatty acid (such as linoleic acid, linolenic acid, and especially fatty acids of 12, 14, 16, 18, 19, 20 or 24 carbon atoms, each carbon chain branched or unbranched), arachadonic acid, palmitoleic acid, oleic acid, polyenoic acid (e.g., palmitoleic acid, oleic acid, linoleic acid and/or linolenic acid), a vitamin (such as pyridoxine, niacinamide, thiamine, etc.), a pH buffer, a surfactant, a trace metal or salt or hydrate thereof, an amine compound, a growth factor, an agent to control osmolarity and/or ionic strength and/or maintain membrane potential, a flavin, a compound that participates in or is a product of the glycolytic pathway
  • the present invention also provides a method of cultivating eukaryotic cells comprising: (a) contacting the cells with the cell culture medium of the present invention; (b) maintaining the cells under conditions suitable to support cultivation of the cells in culture; and (c) optionally expressing a nucleic acid to form a protein product.
  • the present invention also provides a method for introducing one or more macromolecules into at least one eukaryotic cell in culture, the method comprising: (a) culturing at least one eukaryotic cell in the medium of claim 1 in culture; (b) introducing at least one macromolecule into the culture under conditions sufficient to cause one or more of the at least one macromolecule to be introduced in the at least one cell; and (c) cultivating the at least one cell in the medium to produce a product whose production is controlled by the at least one molecule, wherein growth of the at least one cell continues in the medium in the absence of the medium being with fresh medium, wherein it is not necessary to remove medium used during the introduction from the presence of the at least one cell to support growth of the at least one cell, and/or wherein after the introduction, growth is accomplished in cultivation in a volume of medium that is about the same volume up to no more than about 10 times the volume of the medium in which the introduction occurred.
  • the present invention also provides a kit for the cultivation and transfection of cells in vitro, the kit comprising the cell culture medium of the present invention, and optionally further comprising one or more of: one or more agents for the introduction of at least one molecule into a cell, one or more macromolecules, at least one cell, and instructions for culturing the at least one cell in culture and/or for introducing at least one macromolecule into at least one cell in culture.
  • the present invention also provides a composition
  • a composition comprising the cell culture medium of the present invention and at least one component selected from the group consisting of at least one eukaryotic cell, one or more agents for the introduction of at least one macromolecule into at least one cell, and one or more macromolecules.
  • FIG. 1A is a bar graph showing a plot of the number of 293-F cells/ml as a function of passage number in the medium of the present invention.
  • FIG. 1B is a bar graph showing a plot of the number of 293-H cells/ml as a function of passage number in the medium of the present invention.
  • FIG. 2A is a graph showing the number of viable 293-F cells/ml as a function of the number of days cultured in the medium of the invention.
  • FIG. 2B is a graph showing the number of viable 293-H cells/ml as a function of the number of days cultured in the medium of the invention.
  • Growth for 293-F and 293-H cells cultured in 293 SFM II medium (Invitrogen Corp., Carlsbad, Calif.) vs. prototype media A, B and C. 15 ml cultures were seeded at 3 ⁇ 10 5 viable cells per ml in 125 ml shaker flasks in triplicate and 1 ml samples taken each day for viabilities and counts.
  • Cells were cultured in 293 SFM II medium supplemented with 4 mM L-Glutamine or in one of the tested media A, B or C.
  • FIG. 3 is a bar graph showing a plot of the number of 293-F cells/ml after each of three passages in the medium of the invention supplemented with various salts.
  • the three bars correspond, from left to right, to passage 1 (p1), passage 2, (p2) and passage 3 (p3).
  • FIG. 4A is a bar graph showing a plot of 293-F cell density (cells/ml) in pilot lot 1 (PL1), pilot lot 2 (PL2) and pilot lot (PL) 000458 media.
  • FIG. 4B is a bar graph showing a plot of % viable 293-F cells in PL1, PL-2 and PL000458 media.
  • FIG. 4C is a bar graph showing a plot of 293-F cell density (cells/ml) in PL-2 and PL-2A (+insulin) media.
  • FIG. 4D is a bar graph showing a plot of % viable 293-F cells in PL-2 and PL-2A (+insulin) media.
  • the present invention provides improved medium formulations for the growth of both eukaryotic and prokaryotic cells.
  • the inventive media support cell growth, introduction of macromolecules into cells in culture and cell cultivation without requiring replenishment, replacement, supplementation, or changing medium between growth, introduction and/or cultivation.
  • the media of the present invention can be used to support or enhance the growth and cultivation of any cell.
  • the present invention also provides compounds that can be used as substitutes or to replace one or more undesired components, e.g., animal derived components.
  • the replacement compounds provide at least one desired function of the undesired component.
  • introduction of a macromolecule or compound into culture refers to the provision of the macromolecule or compound into the culture medium.
  • introduction of a macromolecule or compound into at least one cell refers to the provision of a macromolecule or compound to a cell, such that the macromolecule or compound becomes internalized in the cell.
  • a macromolecule or compound can be introduced into a cell using transfection, transformation, injection, and/or liposomal introduction, and may also be introduced into a cell using other methods known to those of ordinary skill in the art.
  • a macromolecule or compound is introduced into a cell by liposomal introduction.
  • the macromolecule is preferably a protein, peptide, polypeptide, or nucleic acid.
  • the macromolecule may a protein.
  • the macromolecule may be a peptide.
  • the macromolecule may be a polypeptide.
  • the macromolecule may also be a nucleic acid.
  • macromolecule encompasses biomolecules.
  • the term macromolecule refers to nucleic acid.
  • the term macromolecule refers to deoxyribonucleic acid (DNA) and ribonucleic acid (RNA). More preferably, the term macromolecule refers to DNA. More preferably, the term macromolecule refers to complementary DNA (cDNA).
  • a macromolecule can be charged or uncharged.
  • a DNA molecule is an example of a charged macromolecule.
  • nucleic acid refers to any nucleic acid, including deoxyribonucleic acid (DNA) and ribronucleic acid (RNA).
  • nucleic acid refers to DNA, including genomic DNA, complementary DNA (cDNA), and oligonucleotides, including oligoDNA. More preferably, “nucleic acid’ refers to genomic DNA and/or cDNA.
  • nucleic acid refers to the replication of the nucleic acid in a cell, to transcription of DNA to messenger RNA, to translation of RNA to protein, to post-translational modification of protein, and/or to protein trafficking in the cell, or variations or combinations thereof.
  • the term “ingredient” refers to any compound, whether of chemical or biological origin, that can be used in cell culture media to maintain or promote the growth or proliferation of cells.
  • component e.g., fetal calf serum
  • ingredient can be used interchangeably and are all meant to refer to such compounds.
  • Typical ingredients that are used in cell culture media include amino acids, salts, metals, sugars, lipids, nucleic acids, hormones, vitamins, fatty acids, proteins and the like.
  • Other ingredients that promote or maintain cultivation of cells ex vivo can be selected by those of skill in the art, in accordance with the particular need.
  • Media of the present invention can include one or more components selected from the group consisting of bovine serum albumin (BSA) or human serum albumin (HSA), a one or more growth factors derived from natural (animal) or recombinant sources such as epidermal growth factor (EGF) or fibroblast growth factor (FGF), one or more lipids, such as fatty acids, sterols and phospholipids, one or more lipid derivatives and complexes, such as phosphoethanolamine, ethanolamine and lipoproteins, one or more proteins, one or more and teroid hormones, such as insulin, hydrocortisone and progesterone, one or more nucleotide precursors; and one or more trace elements.
  • BSA bovine serum albumin
  • HSA human serum albumin
  • growth factors derived from natural (animal) or recombinant sources such as epidermal growth factor (EGF) or fibroblast growth factor (FGF)
  • EGF epidermal growth factor
  • FGF fibroblast growth factor
  • cell refers includes all types of eukaryotic and prokaryotic cells. In preferred embodiments, the term refers to eukaryotic cells, especially mammalian cells.
  • cell culture or “culture” is meant the maintenance of cells in an artificial, in vitro environment.
  • cultivation is meant the maintenance of cells in vitro under conditions favoring growth and/or differentiation and/or or continued viability. “Cultivation” can be used interchangeably with “cell culture.” Cultivation is assessed by number of viable cells/ml culture medium. Cultivation after introduction of a macromolecule preferably includes production of a product, for example, a protein product on a virus.
  • fresh medium refers to adding a volume of fresh cell culture medium to medium that was already present in culture and/or replacing medium that was already present in culture with fresh medium, and/or supplementing medium already present in culture with new medium.
  • Fresh medium is medium that does not contain the one or more macromolecules or compounds to be introduced into at least one cell or medium that has not been in contact with cells to support their growth on cultivation.
  • the skilled artisan can determine whether there is an advantage from or a need to remove and/or replenish, replace or supplement medium by monitoring cell growth and/or viability by techniques known in the art, such as cell counting (manual or automated), trypan blue exclusion, production of protein or other substance, alamar blue assay, presence or concentration of one or more metabolic products, cell adhesion, morphological appearance, analysis of spent medium, etc.
  • One or a combination of monitoring techniques can be used to determine whether the medium needs to be to support growth, introduction of at least one macromolecule and/or cultivation after introduction of at least one macromolecule.
  • Recombinant protein refers to protein that is encoded by a nucleic acid that is introduced into a host cell.
  • the host cell expresses the nucleic acid.
  • the term “expressing a nucleic acid” is synonymous with “expressing a protein of an RNA encoded by a nucleic acid.”
  • Protein as used herein broadly refers to polymerized amino acids, e.g., peptides, polypeptides, proteins, lipoproteins, glycoproteins, etc.
  • protein yield refers to the amount of protein expressed by cultured cells, and can be measured, for example, in terms of grams of protein produced/ml medium. If the protein is not secreted by the cells, the protein can be isolated from the interior of the cells by methods known to those of ordinary skill in the art. If the protein is secreted by the cells, the protein can be isolated from the culture medium by methods known to those of ordinary skill in the art. The amount of protein expressed by the cell can readily be determined by those of ordinary skill in the art.
  • the protein may be a recombinant protein.
  • a “protein product” is a product associated with production or an action by a protein.
  • a protein product may be a protein.
  • a protein product may also be a product resulting from action of a protein by one or more other substances to produce a product.
  • An example of such action is enzymatic action by a protein.
  • “suspension culture” is meant cell culture in which the majority or all of cells in a culture vessel are present in suspension, and the minority or none of the cells in the culture vessel are attached to the vessel surface or to another surface within the vessel.
  • “suspension culture” has greater than 75% of the cells in the culture vessel are in suspension, not attached to a surface on or in the culture vessel. More preferably, a “suspension culture” has greater than 85% of the cells in the culture vessel are present in suspension, not attached to a surface on or in the culture vessel. Even more preferred is a “suspension culture” with greater than 95% of the cells in the culture vessel present in suspension, not attached to a surface on or in the culture vessel.
  • the medium, methods, kit and composition of the present invention are suitable for either monolayer or suspension culture, transfection, and cultivation of cells, and for expression of protein in cells in monolayer or suspension culture.
  • the medium, methods, kit and composition of the present invention are for suspension culture, transfection, and cultivation of cells, and for expression of protein product in cells in suspension culture.
  • culture vessel any container, for example, a glass, plastic, or metal container, that can provide an aseptic environment for culturing cells.
  • cell culture medium tissue culture medium
  • culture medium plural “media” in each case
  • medium formulation refers to a nutritive solution for cultivating cells or tissues. These phrases can be used interchangeably.
  • combining refers to the mixing or admixing of ingredients.
  • Derivative of a molecule includes these compounds that comprise the base cyclic or heterocyclic molecule, but have additional or modified side groups.
  • a “derivative” can be formed by reacting the base molecule with only 1, but possibly 2, 3, 4, 5, 6, etc. reactant molecules.
  • a single step reaction is preferred, but multi-step, e.g., 2, 3, 4, 5, 6, etc. reactions are known in the art to form derivatives.
  • Substitution, condensation and hydrolysis reactions are preferred and may be combined to form the derivative compound.
  • a derivative compound may be a compound that preferably in 1, but possibly 2, 3, 4, 5, 6, etc. reactions can form the base cyclic or heterocyclic compound or a substitution or condensation product thereto.
  • a cell culture medium is composed of a number of ingredients and these ingredients can vary from medium to medium. Each ingredient used in a cell culture medium has its unique physical and chemical characteristics. Compatibility and stability of ingredients are determined in part by the “solubility” of the ingredients in aqueous solution. The terms “solubility” and “soluble” refer to the ability of an ingredient to form and remain in solution with other ingredients. Ingredients are thus compatible if they can be maintained in solution without forming a measurable or detectable precipitate.
  • compatible ingredients are also meant those media components which can be maintained together in solution and form a “stable” combination.
  • a solution containing “compatible ingredients” is said to be “stable” when the ingredients do not precipitate, degrade or decompose substantially such that the concentration of one or more of the components available to the cells from the media is reduced to a level that no longer supports the optimum or desired growth of the cells.
  • Ingredients are also considered “stable” if degradation cannot be detected or when degradation occurs at a slower rate when compared to decomposition of the same ingredient in a 1 ⁇ cell culture media formulation. For example, in 1 ⁇ media formulations glutamine is known to degrade into pyrolidone carboxylic acid and ammonia.
  • Glutamine in combination with divalent cations are considered “compatible ingredients” since little or no decomposition of the glutamine can be detected over time in solutions or combinations in which both glutamine and divalent cations are present. See U.S. Pat. No. 5,474,931.
  • compatible ingredients refers to the combination of particular culture media ingredients which, when mixed in solution either as concentrated or 1 ⁇ formulations, are “stable” and “soluble.”
  • the term “1 ⁇ formulation” is meant to refer to any aqueous solution that contains some or all ingredients found in a cell culture medium at working concentrations.
  • the “1 ⁇ formulation” can refer to, for example, the cell culture medium or to any subgroup of ingredients for that medium.
  • the concentration of an ingredient in a 1 ⁇ solution is about the same as the concentration of that ingredient found in a cell culture formulation used for maintaining or cultivating cells in vitro.
  • a cell culture medium used for the in vitro cultivation of cells is a 1 ⁇ formulation by definition. When a number of ingredients are present, each ingredient in a 1 ⁇ formulation has a concentration about equal to the concentration of each respective ingredient in a medium during cell culturing.
  • RPMI-1640 culture medium contains, among other ingredients, 0.2 g/L L-arginine, 0.05 g/L L-asparagine, and 0.02 g/L L-aspartic acid.
  • a “1 ⁇ formulation” of these amino acids contains about the same concentrations of these ingredients in solution.
  • each ingredient in solution has the same or about the same concentration as that found in the cell culture medium being described.
  • concentrations of ingredients in a 1 ⁇ formulation of cell culture medium are well known to those of ordinary skill in the art. See, for example, Methods For Preparation of Media, Supplements and Substrate For Serum - Free Animal Cell Culture Allen R. Liss, N.Y.
  • a “10 ⁇ formulation” is meant to refer to a solution wherein the concentration of each ingredient in that solution is about 10 times more than the concentration of each respective ingredient in a medium during cell culturing.
  • a 10 ⁇ formulation of RPMI-1640 culture medium can contain, among other ingredients, 2.0 g/L L-arginine, 0.5 g/L L-asparagine, and 0.2 g/L L-aspartic acid (compare 1 ⁇ formulation, above).
  • a “10 ⁇ formulation” can contain a number of additional ingredients at a concentration about 10 times that found in the 1 ⁇ culture formulation.
  • “25 ⁇ formulation,” “50 ⁇ formulation,” “100 ⁇ formulation,” “500 ⁇ formulation,” and “1000 ⁇ formulation” designate solutions that contain ingredients at about 25-, 50-, 100-, 500-, or 1000-fold concentrations, respectively, as compared to a 1 ⁇ cell culture formulation.
  • the osmolarity and pH of the medium formulation and concentrated solution can vary.
  • trace element or “trace element moiety” refers to a moiety which is present in a cell culture medium in only very low (i.e., “trace”) amounts or concentrations, relative to the amounts or concentrations of other moieties or components present in the culture medium.
  • these terms encompass Ag + , Al 3+ , Ba 2+ , Cd 2 , Co 2+ , Cr 3+ , Cu 1+ , Cu 2+ , Fe 2+ , Fe 3+ , Ge 4+ , Se 4+ , Br ⁇ , I ⁇ , Mn 2+ , F ⁇ , Si 4+ , V 5+ , Mo 6+ , Ni 2+ , Rb + , Sn 2+ and Zr 4+ and salts thereof.
  • the following salts can be used as trace elements in the culture media of the invention: AgNO 3 , AlCl 3 .6H 2 O, Ba(C 2 H 3 O 2 ) 2 , CdSO 4 .8H 2 O, CoCl 2 .6H 2 O, Cr 2 (SO 4 ) 3 .1H 2 O, GeO 2 , Na 2 SeO 3 , H 2 SeO 3 , KBr, KI, MnCl 2 .4H 2 O, NaF, Na 2 SiO 3 .9H 2 O, NaVO 3 , (NH 4 ) 6 Mo 7 O 24 .4H 2 O, NiSO 4 .6H 2 O, RbCl, SnCl 2 , and ZrOCl 2 .8H 2 O.
  • concentrations of trace element moieties can be determined by one of ordinary skill in the art using only routine experimentation.
  • amino acid refers to amino acids or their derivatives (e.g., amino acid analogs), as well as their D- and L-forms.
  • amino acids include glycine, L-alanine, L-asparagine, L-cysteine, L-aspartic acid, L-glutamic acid, L-phenylalanine, L-histidine, L-isoleucine, L-lysine, L-leucine, L-glutamine, L-arginine, L-methionine, L-proline, L-hydroxyproline, L-serine, L-threonine, L-tryptophan, L-tyrosine, and L-valine, N-acetyl cysteine.
  • a “serum-free medium” is a medium that contains no serum (e.g., fetal bovine serum (FBS), calf serum, horse serum, goat serum, human serum, etc.) and is generally designated by the letters SFM.
  • FBS fetal bovine serum
  • SFM serum-free medium
  • serum-free culture conditions and “serum-free conditions” refer to cell culture conditions that exclude serum of any type. These terms can be used interchangeably.
  • a “chemically defined” medium is one in which each chemical species and its respective quantity is known prior to its use in culturing cells.
  • a chemically defined medium is made without lysates or hydrolysates whose chemical species are not known and/or quantified.
  • a chemically defined medium is one preferred embodiment of the medium of the present invention.
  • protein-free culture media refers to culture media that contain no protein (e.g., no serum proteins such as serum albumin or attachment factors, nutritive proteins such as growth factors, or metal ion carrier proteins such as transferrin, ceruloplasmin, etc.).
  • no protein e.g., no serum proteins such as serum albumin or attachment factors, nutritive proteins such as growth factors, or metal ion carrier proteins such as transferrin, ceruloplasmin, etc.
  • the peptides are smaller peptides, e.g., di- or tri-peptides.
  • peptides of deca-peptide length or greater are less than about 1%, more preferably less than about 0.1%, and even more preferably less than about 0.01% of the amino acids present in the protein free medium.
  • low-protein culture media refers to media that contain only low amounts of protein (typically less than about 10%, less than about 5%, less than about 1%, less than about 0.5%, or less than about 0.1%, of the amount or concentration of total protein found in culture media containing standard amounts of protein, such as standard basal medium supplemented with 5-10% serum).
  • animal derived material refers to material that is derived in whole or in part from an animal source, including recombinant animal DNA or recombinant animal protein DNA. Preferred media contain no animal desired material.
  • transition element or “transition metal” (which can be used interchangeably) is meant an element in which an inner electron valence shell, rather than an outer shell, is only partially filled, such that the element acts as a transitional link between the most and least electropositive in a given series of elements. Transition elements are typically characterized by high melting points, high densities, high dipole or magnetic moments, multiple valencies, and the ability to form stable complex ions.
  • transition elements useful in the present invention include scandium (Sc), titanium (Ti), vanadium (V), chromium (Cr), manganese (Mn), iron (Fe), cobalt (Co), nickel (Ni), copper (Cu), zinc (Zn), yttrium (Y), zirconium (Zr), niobium (Nb), molybdenum (Mo), technetium (Tc), rubidium (Ru), rhodium (Rh), palladium (Pd), silver (Ag), cadmium (Cd), lanthanum (La), hafnium (Hf), tantalum (Ta), tungsten (W), rhenium (Re), osmium (Os), iridium (Ir), platinum (Pt), gold (Au), mercury (Hg), and actinium (Ac).
  • reagents include, for example, calcium phosphate, DEAE-dextran and lipids.
  • reagents include, for example, calcium phosphate, DEAE-dextran and lipids.
  • numerous references texts are available for example, Current Protocols in Molecular Biology , Chapter 9, Ausubel, et al. Eds., John Wiley and Sons, 1998.
  • Lipid aggregates such as liposomes have been found to be useful as agents for the delivery of macromolecules into cells.
  • lipid aggregates comprising one or more cationic lipids have been demonstrated to be extremely efficient at the delivery of anionic macromolecules (for example, nucleic acids) into cells.
  • anionic macromolecules for example, nucleic acids
  • One commonly used cationic lipid is N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA).
  • DOTMA N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride
  • DOPE dioleoylphosphatidylethanolamine
  • DOTMA 1,2-bis(oleoyl-oxy)-3-3-(trimethylammonia) propane
  • DOTAP 1,2-bis(oleoyl-oxy)-3-3-(trimethylammonia) propane
  • DOTAP differs from DOTMA in that the oleoyl moieties are linked to the propylamine backbone via ether bonds in DOTAP whereas they are linked via ester bonds in DOTMA.
  • DOTAP is believed to be more readily degraded by the target cells.
  • a structurally related group of compounds wherein one of the methyl groups of the trimethylammonium moiety is replaced with a hydroxyethyl group are similar in structure to the Rosenthal inhibitor (RI) of phospholipase A (see Rosenthal, et al., (1960) J. Biol. Chem. 233:2202-2206.).
  • the RI has stearoyl esters linked to the propylamine core.
  • the dioleoyl analogs of RI are commonly abbreviated DOR1-ether and DOR1-ester, depending upon the linkage of the lipid moiety to the propylamine core.
  • the hydroxyl group of the hydroxyethyl moiety can be further derivatized, for example, by esterification to carboxyspermine.
  • DPES dipalmitoylphosphatidylethanolamine 5-carboxyspermylamide
  • DOGS 5-carboxyspermylglycine dioctadecylamide
  • a cationic derivative of cholesterol (3 ⁇ -[N—(N′,N′-dimethylaminoethane)-carbamoyl] cholesterol, DC-Chol) has been synthesized and formulated into liposomes with DOPE (see Gao, et al., (1991) BBRC 179(1):280-285.) and used to introduce DNA into cells.
  • the liposomes thus formulated were reported to efficiently introduce DNA into the cells with a low level of cellular toxicity.
  • Lipopolylysine formed by conjugating polylysine to DOPE (see Zhou, et al., (1991) BBA 1065:8-14), has been reported to be effective at introducing nucleic acids into cells in the presence of serum.
  • cationic lipids that have been used to introduce nucleic acids into cells include highly packed polycationic ammonium, sulfonium and phosphonium lipids such as those described in U.S. Pat. Nos. 5,674,908 and 5,834,439, and international application no. PCT/US99/26825, published as WO 00/27795.
  • One preferred agent for delivery of macromolecules is LIPOFECTAMINE 2000TM which is available from Invitrogen. See U.S. international application no. PCT/US99/26825, published as WO 00/27795.
  • a “reagent for the introduction of macromolecules” into cells is any material known to those of skill in the art which facilitates the entry of a macromolecule into a cell.
  • the reagent can be a “transfection reagent” and can be any compound and/or composition that increases the uptake of one or more nucleic acids into one or more target cells.
  • transfection reagents are known to those skilled in the art.
  • Suitable transfection reagents can include, but are not limited to, one or more compounds and/or compositions comprising cationic polymers such as polyethyleneimine (PEI), polymers of positively charged amino acids such as polylysine and polyarginine, positively charged dendrimers and fractured dendrimers, cationic ⁇ -cyclodextrin containing polymers (CD-polymers), DEAE-dextran and the like.
  • a reagent for the introduction of macromolecules into cells can comprise one or more lipids which can be cationic lipids and/or neutral lipids.
  • Preferred lipids include, but are not limited to, N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethylamonium chloride (DOTMA), dioleoylphosphatidylcholine (DOPE),1,2-Bis(oleoyloxy)-3-(4′-trimethylammonio) propane (DOTAP), 1,2-dioleoyl-3-(4′-trimethylammonio) butanoyl-sn-glycerol (DOTB), 1,2-dioleoyl-3-succinyl-sn-glycerol choline ester (DOSC), cholesteryl (4′-trimethylammonio)butanoate (ChoTB), cetyltrimethylammonium bromide (CTAB), 1,2-dioleoyl-3-dimethyl-hydroxyethyl ammonium bromide (DORI), 1,2-dioleyloxypropyl-3-dimethyl-hydroxye
  • lipids have been shown to be particularly suited for the introduction of nucleic acids into cells for example a 3:1 (w/w) combination of DOSPA and DOPE is available from Invitrogen Corporation, Carlsbad, Calif. under the trade name LIPOFECTAMINETM, a 1:1 (w/w) combination of DOTMA and DOPE is available from Invitrogen Corporation, Carlsbad, Calif. under the trade name LIPOFECTIN®, a 1:1 (M/M) combination of DMRIE and cholesterol is available from Invitrogen Corporation, Carlsbad, Calif.
  • LIPOFECTAMINETM a 1:1 (w/w) combination of DOTMA and DOPE is available from Invitrogen Corporation, Carlsbad, Calif.
  • LIPOFECTIN® a 1:1 (M/M) combination of DMRIE and cholesterol is available from Invitrogen Corporation, Carlsbad, Calif.
  • DMRIE-C reagent a 1:1.5 (M/M) combination of TM-TPS and DOPE is available from Invitrogen Corporation, Carlsbad, Calif. under the trade name CellFECTIN® and a 1:2.5 (w/w) combination of DDAB and DOPE is available from Invitrogen Corporation, Carlsbad, Calif. under the trade name LipfectACE®.
  • other formulations comprising lipids in admixture with other compounds, in particular, in admixture with peptides and proteins comprising nuclear localization sequences, are known to those skilled in the art. For example, see international application no. PCT/US99/26825, published as WO 00/27795.
  • the present invention is directed to a culture medium that supports (a) the introduction of at least one macromolecule into eukaryotic cells in culture, (b) the cultivation of cells into which at least one macromolecule is introduced, and optionally (c) the production of protein product or expression of the nucleic acid in cells into which at least one macromolecule is introduced, wherein medium containing the macromolecule does not need to be removed from the culture and replaced with fresh medium after introduction of at least one macromolecule into cells and prior to cultivation and production of protein product or expression of nucleic acid.
  • the medium of the present invention it is not necessary to replenish, replace or supplement the medium after one has introduced at least on emacromolecule into at least one cell, and before cells into which at least one macromolecule has been introduced are further cultured to produces protein product or express a nucleic acid.
  • the medium is a serum-free medium and/or a chemically defined medium and/or protein free or low protein medium and/or a medium that does not contain animal derived components, or a medium having combinations of these features.
  • CD CHO medium (Invitrogen Corporation, Carlsbad, Calif.) can be used to transfect CHO cells in suspension culture.
  • transfection of CHO cells in CD CHO medium works only if the cells are transfected with nucleic acid in a relatively much smaller volume of CD CHO medium than the transfected cells are to be cultured in after transfection.
  • the culture medium of the present invention facilitates higher cell transformation efficiency than does CD CHO medium, and/or does not require transfecting in a smaller volume than cells are to be cultured in after transfection, and/or facilitates higher cell viability than does CD CHO medium, and/or facilitates higher cell density (i.e., cells/ml of culture medium) than does CD CHO medium, and/or facilitates a higher level of recombinant protein expression in cells in culture than does CD CHO medium.
  • the same volume of medium is used for introduction of at least one macromolecule or transfection and subsequent cultivation.
  • the cells are divided or medium volume is increased less from about 2, about 5, about 8 or about 10 times.
  • the culture medium of the present invention is not CD CHO medium.
  • the medium, methods, kit and composition of the present invention are intended to be used to introduce at least one macromolecule or to transfect and culture cells in any volume of culture medium. Such introduction is preferably accomplished in 0.1 to 10 times the amount of medium used to culture cells to be transfected.
  • the cell culture volume is greater than about one milliliter. More preferably, the cell culture volume is from about one milliliter to 250 liters. More preferably, the cell culture volume is from about 30 ml to about 50 liters. More preferably, the cell culture volume is from about 100 ml to about 50 liters. More preferably, the cell culture volume is from about 500 ml to about 50 liters. More preferably, the cell culture volume is from about 500 ml to about 25 liters.
  • the cell culture volume is from about 500 ml to about 10 liters. More preferably, the cell culture volume is from about 500 ml to about 5 liters. More preferably, the cell culture volume is from about 500 ml to about 1 liter.
  • the medium preferably does not contain compounds that can interfere with introduction of macromolecules or transfection, e.g., polyanionic compounds such as polysulfonated and/or polysulfated compounds.
  • the medium does not contain dextran sulfate.
  • the medium, methods, kit and composition of the present invention permit the introduction of compounds or macromolecules (particularly macromolecules, for example nucleic acids, proteins and peptides) into the cultured cells (for example by transfection) without the need to change the medium.
  • the present invention provides a medium for the cultivation and transfection of eukaryotic cells.
  • macromolecules or compounds e.g., nucleic acid
  • the macromolecule or compound e.g., nucleic acid
  • the macromolecule or compound is introduced into at least about 20 percent of the cells. More preferably, the macromolecule or compound (e.g., nucleic acid) is introduced into about 20 to about 100 percent of the cells. More preferably, the macromolecule or compound (e.g., nucleic acid) is introduced into about 30 to about 100 percent of the cells. More preferably, the macromolecule or compound (e.g., nucleic acid) is introduced into about 50 to about 100 percent of the cells.
  • the macromolecule or compound might be introduced into about 20% to about 90% of the cells, about 20% to about 80% of the cells, about 30% to about 60, 70, 80 or 90% of the cells, about 20, 30, 40 or 50% to about 70, 75, 80, 85, 90, 95 or 98% of the cells, etc. Even about 60, 70, 75 or 80 to about 90% or close to 100% of the cells may contain the introduced molecule or compound.
  • one or more undesirable components i.e., one or more serum components, one or more undefined components, one or more protein components and/or one or more animal derived components
  • Replacement compounds of the invention can be metal binding compounds and/or one or more transition element complexes, said complexes comprising one or more transition elements or a salts or ions thereof, in a complex with one or more metal-binding compounds.
  • the medium is capable of supporting the cultivation of a cell in vitro in the absence of one or more naturally derived metal carriers, such as transferrin, or other animal derived proteins or extracts.
  • the metal binding compound can be in a complex with a transition metal prior to addition of the metal binding compound to the medium. In other embodiments, the metal binding compound is not in a complex with a transition metal prior to addition of the metal binding compound to the media.
  • the medium of the present invention does not contain transferrin and/or does not contain insulin.
  • the present invention also relates to a cell culture medium obtained by combining a medium with one or more replacement compounds.
  • the medium can be a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or can be a medium lacking animal derived components.
  • the medium preferably does not contain transferrin and/or does not contain insulin.
  • the medium can be capable of supporting the cultivation of a cell in vitro and/or can permit the introduction of macromolecules into the cell.
  • one or more of the replacement compounds can be a metal binding compound and/or can be a transition element complex, said complex comprising at least one transition element or a salt or ion thereof complexed to at least one metal-binding compound.
  • Preferred transition elements, metal-binding compounds, and transition element complexes for use in this aspect of the invention include those described in detail herein.
  • Replacement compounds of the present invention can facilitate the delivery of transition metals to cells cultured in vitro.
  • the replacement compounds can deliver iron and replace transferrin.
  • a preferred replacement compound is a hydroxypyridine derivative.
  • the hydroxypyridine derivative is selected from the group consisting of 2-hydroxypyridine-N-oxide, 3-hydroxy-4-pyrone, 3-hydroxypypyrid-2-one, 3-hydroxypyrid-2-one, 3-hydroxypyrid-4-one, 1-hydroxypyrid-2-one, 1,2-dimethyl-3-hydroxypyrid-4-one, 1-methyl-3-hydroxypyrid-2-one, 3-hydroxy-2(1H)-pyridinone, and pyridoxal isonicotinyl hydrazone, nicotinic acid-N-oxide, 2-hydroxy-nicotinic acid.
  • the hydroxypyridine derivative is 2-hydroxypyridine-N-oxide.
  • the replacement compounds of the present invention can be used with any media, including media for cultivating or growing eukaryotic and/or prokaryotic cells, tissues, organs, etc.
  • media include, but are not limited to, Dulbecco's Modified Eagle's Medium (DMEM), Minimal Essential Medium (MEM), Basal Medium Eagle (BME), RPMI-1640, Ham's F-10, Ham's F-12, ⁇ Minimal Essential Medium ( ⁇ MEM), Glasgow's Minimal Essential Medium (G-MEM), and Iscove's Modified Dulbecco's Medium (IMDM).
  • the present invention also provides a method for introducing macromolecules into cells, comprising culturing cells in a medium of the invention and contacting the cells in the medium with one or more macromolecules under conditions causing the macromolecules to be taken up by one or more of the cells.
  • the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or can be a medium lacking animal derived components.
  • Preferred cells include eukaryotic cells. More preferably, the cells are mammalian cells.
  • the medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin. In some preferred embodiments, the medium permits the growth and transfection of the cell in the same medium.
  • the macromolecules can comprise one or more nucleic acids and conditions causing the nucleic acid molecules to be taken up by the cells include contacting the nucleic acid with a reagent which causes the nucleic acid to be introduced into one or more cells.
  • the present invention also provides a composition comprising a medium of the invention and a cell.
  • the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or a medium lacking animal derived components.
  • Preferred cells include eukaryotic cells. More preferably, the cells are mammalian cells.
  • the medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin.
  • the medium supports the growth and transfection of the cell in the same medium.
  • the present invention also provides compositions comprising a medium of the present invention and one or more reagents for the introduction of macromolecules into one or more cells.
  • the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or a medium lacking animal derived components.
  • the medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin.
  • the medium contains a transfection reagent and the macromolecules are nucleic acids.
  • the macromolecules might also be proteins and/or peptides.
  • the reagent comprises one or more lipids of which one or more can be cationic lipids. More preferably, the reagent comprises a mixture of neutral and cationic lipids.
  • the reagent comprises one or more peptides and/or proteins which can be provided alone or in admixture with one or more lipids.
  • the present invention also provides compositions comprising a medium of the invention and one or more macromolecules to be introduced into a cell.
  • the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or a medium lacking animal derived components.
  • the medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin.
  • the macromolecules can be, for example, nucleic acids and/or proteins and/or peptides and can be uncomplexed or can be in the form of a complex with one or more reagents for the introduction of macromolecules into cells.
  • the macromolecules are nucleic acids and can be in the form of a complex with one or more transfection reagents.
  • the present invention also provides a composition comprising at least one component (or combination thereof) selected from the group consisting of a medium of the present invention, at least one cell, at least one macromolecule, at least one reagene for introducing at least one macromoleule into at least one cell.
  • the cells are eukaryotic cells. More preferably, the cells are mammalian cells.
  • the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or a medium lacking animal derived components.
  • the medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin.
  • the reagent is a transfection reagent and the macromolecules are nucleic acids, for example RNA and/or DNA. Alternatively, the macromolecules are proteins and/or peptides.
  • the reagent comprises one or more lipids of which one or more can be cationic lipids. More preferably, the reagent comprises a mixture of neutral and cationic lipids. In some embodiments, the reagent comprises one or more peptides and/or proteins which can be provided alone or in admixture with one or more lipids. In preferred embodiments, the reagent complexes with the macromolecule to introduce the macromolecule into the cell.
  • kits for the culture and transfection of cells comprising at least one container comprising a medium for the culture and transfection of cells.
  • kits may also comprise at least one component (or a combination thereof) selected from the group consisting of a medium of the present invention, at least one cell, at least one macromolecule, at least one reagent for introducing at least one macromolecule into at least one cell, at least one buffer or buffering salt, and instructions for using the kit to introduce at least one macromolecule into at least one cell.
  • the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or a medium lacking animal derived components.
  • the medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin and/or does not contain an animal growth factor.
  • the medium can comprise one or more replacement compounds which can be metal binding compounds and/or can comprise one or more complexes comprising one or more replacement compounds.
  • the medium can comprise one or more complexes, said complex comprising one or more transition elements or salts or ions thereof complexed one or more replacement compounds which can be metal-binding compounds.
  • said medium is capable of supporting the cultivation of a cell in vitro and permits transfection of cells cultured therein.
  • kits of the invention can further comprise at least one container comprising a lipid for transfecting cells.
  • the kits of the invention can comprise at least one container comprising a nucleic acid.
  • a transition element is preferably selected from the group consisting of scandium, titanium, vanadium, chromium, manganese, iron, cobalt, nickel, copper, zinc, yttrium, zirconium, niobium, molybdenum, technetium, rubidium, rhodium, palladium, silver, cadmium, lanthanum, hafnium, tantalum, tungsten, rhenium, osmium, iridium, platinum, gold, mercury, and actinium, or salts or ions thereof, and is preferably an iron salt.
  • Suitable iron salts include, but are not limited to, FeCl 3 , Fe(NO 3 ) 3 or FeSO 4 or other compounds that contain Fe +++ or Fe ++ ions.
  • Preferred replacement compounds include, but are not limited to, metal-binding compounds. See, for example, international patent application no. PCT/US00/23580, publication no. WO 01/16294.
  • Metal binding compounds of the present invention include any macromolecules which can interact with or bind with transition elements and facilitate their uptake by cells. Such interaction/binding can be covalent or non-covalent in nature.
  • the metal-binding compound used in this aspect of the invention is preferably selected from the group consisting of a polyol, a hydroxypyridine derivative, 1,3,5-N,N′,N′′-tris(2,3-dihydroxybenzoyl)amino-methylbenzene, ethylenediamine-N,N′-tetramethylenephosphonic acid, trisuccin, an acidic saccharide (e.g., ferrous gluconate), a glycosaminoglycan, diethylenetriaminepentaacetic acid, nicotinic acid-N-oxide, 2-hydroxy-nicotinic acid, mono-, bis-, or tris-substituted 2,2′-bipyridine, a hydroxamate derivative (e.g.
  • the metal-binding compound is a polyol such as sorbitol or dextran, and particularly sorbitol.
  • the metal-binding compound is a hydroxypyridine derivative, such as 2-hydroxypyridine-N-oxide, 3-hydroxy-4-pyrone, 3-hydroxypypyrid-2-one, 3-hydroxypyrid-2-one, 3-hydroxypyrid-4-one, 1-hydroxypyrid-2-one, 1,2-dimethyl-3-hydroxypyrid-4-one, 1-methyl-3-hydroxypyrid-2-one, 3-hydroxy-2(1H)-pyridinone, ethyl maltol or pyridoxal isonicotinyl hydrazone, and is preferably 2-hydroxypyridine-N-oxide.
  • 2-hydroxypyridine-N-oxide such as 2-hydroxypyridine-N-oxide, 3-hydroxy-4-pyrone, 3-hydroxypypyrid-2-one, 3-hydroxypyrid-2-one, 3-hydroxypyrid-4-one, 1-hydroxypyrid-2-one, 1,2-dimethyl-3-hydroxypyrid-4-one, 1-methyl-3-hydroxypyrid-2-one, 3-hydroxy-2(1H)
  • the transition metal complex can be a sorbitol-iron complex or 2-hydroxypyridine-N-oxide-iron complex.
  • the metal binding compounds of the present invention can also bind divalent cations such as Ca ++ and Mg ++ .
  • the invention relates to cell culture media comprising one or more replacement compounds which can be metal-binding compounds and further comprising one or more ingredients selected from the group of ingredients consisting of at least one amino acid (such as L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid, L-glutamine, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine or L-valine, N-acetyl-cysteine), at least one vitamin (such as biotin, choline chloride, D-Ca ++ -pantothenate, folic acid, i-inositol, niacinamide, pyridoxine, riboflavin,
  • the culture media of the present invention can optionally include one or more buffering agents.
  • Suitable buffering agents include, but are not limited to, N-[2-hydroxyethyl]-piperazine-N′-[2-ethanesulfonic acid] (HEPES), MOPS, MES, phosphate, bicarbonate and other buffering agents suitable for use in cell culture applications.
  • a suitable buffering agent is one that provides buffering capacity without substantial cytotoxicity to the cells cultured. The selection of suitable buffering agents is within the ambit of ordinary skill in the art of cell culture.
  • a medium suitable for use in forming the cell culture media of the invention can comprise one or more ingredients, and can be obtained, for example, by combining one or more ingredients selected from the group consisting of adenine, ethanolamine, D-glucose, heparin, a buffering agent, hydrocortisone, lipoic acid, phenol red, phosphoethanolamine, putrescine, sodium pyruvate, tri-iodothyronine, thymidine, L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid, L-glutamine, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, L-va
  • the invention is also directed to a cell culture medium comprising ingredients selected from ethanolamine, D-glucose, HEPES, insulin, linoleic acid, lipoic acid, phenol red, PLURONIC F68, putrescine, sodium pyruvate, transferrin, L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid, L-glutamine, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, L-valine, biotin, choline chloride, D-Ca ++ -pantothenate, folic acid, i-inositol, niacinamide, pyridoxine,
  • the invention is also directed to such media which can optionally further comprise one or more supplements selected from the group consisting of one or more cytokines, heparin, one or more animal peptides, one or more yeast peptides and one or more plant peptides (most preferably one or more of rice, aloevera, soy, maize, wheat, pea, squash, spinach, carrot, potato, sweet potato, tapioca, avocado, barley, coconut and/or green bean, and/or one or more other plants), e.g., see international application no. PCT/US97/18255, published as WO 98/15614.
  • one or more supplements selected from the group consisting of one or more cytokines, heparin, one or more animal peptides, one or more yeast peptides and one or more plant peptides (most preferably one or more of rice, aloevera, soy, maize, wheat, pea, squash, spinach, carrot, potato, sweet potato, tapioca, avocado, barley
  • the media provided by the present invention can be protein-free, and can be a 1 ⁇ formulation or concentrated as, for example, a 10 ⁇ , 20 ⁇ , 25 ⁇ , 50 ⁇ , 10 ⁇ , 500 ⁇ , or 1000 ⁇ medium formulation.
  • the media of the invention can also be prepared in different forms, such as dry powder media (“DPM”), a granulated preparation (which requires addition of water, but not other processing, such as pHing), liquid media or as media concentrates.
  • DPM dry powder media
  • a granulated preparation which requires addition of water, but not other processing, such as pHing
  • liquid media or as media concentrates.
  • the basal medium that is a medium useful only for maintenance, but not for growth or production of product, can comprise a number of ingredients, including amino acids, vitamins, organic and inorganic salts, sugars and other components, each ingredient being present in an amount which supports the cultivation of a mammalian epithelial cell in vitro.
  • the medium can be used to culture a variety of cells.
  • the medium is used to culture eukaryotic cells. More preferably, the medium is used to culture plant and/or animal cells. More preferably, the medium is used to culture mammalian cells, fish cells, insect cells, amphibian cells or avian cells. More preferably, the medium is used to culture mammalian cells.
  • the medium is used to culture mammalian cells, including primary epithelial cells (e.g., keratinocytes, cervical epithelial cells, bronchial epithelial cells, tracheal epithelial cells, kidney epithelial cells and retinal epithelial cells) and established cell lines and their strains (e.g., 293 embryonic kidney cells, BHK cells, HeLa cervical epithelial cells and PER-C6 retinal cells, MDBK (NBL-1) cells, 911 cells, CRFK cells, MDCK cells, CHO cells, BeWo cells, Chang cells, Detroit 562 cells, HeLa 229 cells, HeLa S3 cells, Hep-2 cells, KB cells, LS180 cells, LS174T cells, NCI-H-548 cells, RPMI 2650 cells, SW-13 cells, T24 cells, WI-28 VA13, 2RA cells, WISH cells, BS-C-I cells, LLC-MK 2 cells, Clone M-3 cells, 1-10 cells, R
  • the medium is used to culture mammalian cells selected from the group consisting of 293 cells, PER-C6 cells, CHO hells, COS cells and Sp2/0 cells. More preferably, the medium is used to culture 293 cells. Preferably, the medium is used to culture cells in suspension.
  • Cells supported by the medium of the present invention can be derived from any animal, preferably a mammal, and most preferably a mouse or a human.
  • the cells cultivated in the present media can be normal cells or abnormal cells (i.e., transformed cells, established cells, or cells derived from diseased tissue samples).
  • the present invention also provides methods of cultivating mammalian epithelial or fibroblast cells using the culture medium formulations disclosed herein, comprising (a) contacting the cells with the cell culture media of the invention; and (b) cultivating the cells under conditions suitable to support cultivation of the cells.
  • the methods of the present invention can optionally include a step of contacting the cultured cells with a solution comprising one or more macromolecules (preferably comprising one or more nucleic acids) under conditions causing the introduction of one or more of the macromolecules into one or more of the cells.
  • cells cultivated according to these methods are cultivated in suspension.
  • compositions comprising the culture media of the present invention, which optionally can further comprise one or more mammalian epithelial or fibroblast cells, such as those described above, particularly one or more 293 embryonic kidney cells, PER-C6 retinal cells, and CHO cells.
  • mammalian epithelial or fibroblast cells such as those described above, particularly one or more 293 embryonic kidney cells, PER-C6 retinal cells, and CHO cells.
  • the present invention further relates to methods of cultivating mammalian cells (particularly those described above and most particularly 293 embryonic kidney epithelial cells, PER-C6, and CHO cells) in suspension comprising (a) obtaining a mammalian cell to be cultivated in suspension; and (b) contacting the cell with the culture media of the invention under conditions sufficient to support the cultivation of the cell in suspension.
  • mammalian cells particularly those described above and most particularly 293 embryonic kidney epithelial cells, PER-C6, and CHO cells
  • the present invention further relates to methods of producing a virus, and to viruses produced by these methods, the methods comprising (a) obtaining a mammalian cell, preferably a mammalian cell described above, e.g., a primate, especially simian and human, and most preferably a 293 embryonic kidney epithelial cell, PER-C6 cell, 911 cell, or CHO cell, to be infected with a virus; (b) contacting the cell with a virus under conditions suitable to promote the infection of the cell by the virus; and (c) cultivating the cell in the culture medium of the invention under conditions suitable to promote the production of the virus by the cell.
  • Viruses which can be produced according to these methods include adenoviruses, adeno-associated viruses and retroviruses.
  • the present invention further relates to methods of producing a polypeptide, and to polypeptides produced by these methods, the methods comprising (a) obtaining a cell, preferably a mammalian cell described above and most preferably a 293 embryonic kidney epithelial cell, PER-C6, or CHO cell; (b) contacting the cell with a solution comprising a nucleic acid encoding the polypeptide under conditions causing the introduction of the nucleic acid into the cell; and (c) cultivating the cell in the culture medium of the invention under conditions favoring the expression of the desired polypeptide by the cell.
  • the present invention is also directed to compositions, particularly cell culture media, comprising one or more replacement compounds.
  • the replacement compounds can be metal binding compounds and/or one or more transition elements in a complex with one or more metal binding compounds.
  • Such cell culture media of the invention can be used to grow or cultivate plant cells, animal cells (particularly human cells), insect cells, bacterial cells, yeast cells and more generally any type of eukaryotic or prokaryotic cells.
  • the replacement compounds of the present invention can be added to any type or kind of culture media, and are preferably used to replace naturally derived metal carriers (e.g., animal derived proteins or extracts such as transferrin) in such media.
  • the invention is also directed to methods of use of such compositions, including, for example, methods for the cultivation of eukaryotic cells, particularly animal cells, in vitro.
  • the invention also relates to compositions comprising such culture media and one or more cells, especially those cells specifically referenced herein, and to kits comprising one or more of the above-described compositions.
  • the invention in another aspect, relates to a kit for the cultivation of cells in vitro.
  • the kit comprise one or more containers, wherein a first container contains the culture medium of the present invention.
  • the kit can further comprise one or more additional containers, each container containing one or more supplements selected from the group consisting of one or more cytokines, heparin, one or more animal or animal-derived peptides, one or more yeast peptides and one or more plant peptides (which are preferably one or more peptides from rice, aloevera, soy, maize, wheat, pea, squash, spinach, carrot, potato, sweet potato, tapioca, avocado, barley coconut and/or green bean, and/or one or more other plants).
  • the kit of the present invention can further comprise one or more containers comprising a nucleic acid and/or a reagent that facilitates the introduction of at least one macromolecule, e.g., a nucleic acid into cells cultured in the media of the present invention, i.e., a transfection reagent.
  • a transfection reagent include, but are not limited to, cationic lipids and the like.
  • kits according to one aspect of the invention can comprise one or more of the culture media of the invention, one or more replacement compounds, which can be one or more metal binding compounds, and/or one or more transition element complexes, and can optionally comprise one or more nucleic acids and transfection reagents.
  • Kits according to another aspect of the invention can comprise one or more cell culture media (one of which can be a basal medium) and one or more replacement compounds.
  • Preferred replacement compounds include, but are not limited to, metal binding compounds and/or transition element complexes, said complexes comprising at least one transition element or a salt or ion thereof complexed to at least one metal-binding compound.
  • Preferred transition elements, metal-binding compounds, and transition element complexes for use in the kits according to this aspect of the invention include those described in detail herein.
  • the kit of the present invention can also contain instructions for using the kit to culture cells and/or introduce macromolecules or compounds (e.g., nucleic acid, such as DNA), into cells.
  • macromolecules or compounds e.g., nucleic acid, such as DNA
  • 293 Cells Semi-confluent adherent cultures of 293 Cells (ATCC, CRL 1573) are readily adapted to suspension culture.
  • the cells are first detached with a solution of Trypsin-EDTA (0.05% Trypsin, 0.53 mM Na 4 EDTA), and then resuspended in conventional medium supplemented with 10% FBS to inhibit the trypsin.
  • the resuspended cells are centrifuged at 200 ⁇ g for five minutes.
  • the cell pellet is resuspended in 293 SFM (available from Invitrogen Corporation, Carlsbad, Calif.) the formulation of which is described in WO 98/08934 which is specifically incorporated herein by reference.
  • the cells can be detached with VERSENE (Na 4 EDTA, 0.53 mM) and resuspended in 293 SFM.
  • the initial seeding density of the 293 cells after conversion to suspension culture is 1 ⁇ 10 6 cells/mL.
  • the cells are shaken on a rotary shaker at 150 rpm in a 37° C. incubator equilibrated with 8% CO 2 -92% air.
  • the cells can be vortexed vigorously for approximately 45 seconds to obtain a predominantly single cell suspension at the time of passaging and counting. After several passages in suspension culture, the maximum achievable density can be determined.
  • the 293 cells which are described here were grown to approximately 3-4 ⁇ 10 6 cells/mL in suspension culture.
  • 293 SFM medium was used.
  • 293 SFM medium is no longer available from Invitrogen (Carlsbad, Calif.), but similar results would be expected using 293 SFM II medium (Invitrogen, Carlsbad, Calif.).
  • Positive control cultures contained 5 ag/mL human holo-transferrin while negative control cultures were established in the absence of either transferrin or replacement compound.
  • Replacement compound stocks were prepared at 0.1M-0.2M in ddH 2 O and solubilized when necessary using 5N NaOH or HCl.
  • Iron complexes were established using 0.2M replacement compound and iron stocks mixed 1:1 (v/v) and incubated for 5-10 minutes at 22° C. All solutions containing replacement compounds were filter sterilized using a 0.22 ⁇ m Millex-GV filter prior to addition to transferrin and serum-free media.
  • Replacement compound stocks were prepared at 0.1M-0.2M in ddH 2 O and solubilized when necessary using 5N NaOH or HCl. Iron complexes were established using 0.2M replacement compound and iron stocks mixed 1:1 (v/v) and incubated for 5-10 minutes at 22° C. All solutions containing replacement compounds were filter sterilized using a 0.22 ⁇ m Millex-GV filter prior to addition to transferrin and serum-free media.
  • 293-F cells are a clone of 293 cells obtained from Robert Horlick at Pharmacopeia, Princeton, N.J. Cells were adapted to the test media for several passages (>4) prior to transfection experiments. When cultured in suspension, cells were split to 3 ⁇ 10 5 viable cells per ml at each passage. On the week of transfection cells were seeded at 3 ⁇ 10 5 viable cells per ml. After ⁇ 48 hrs of growth (day 2) cells were vortexed for 30 seconds and counted and appropriate volumes used for transfection as described in procedures below.
  • the osmolarity is adjusted to from about 275-310 mOsm, preferably about 280 mOsm and the pH is adjusted to from about 7.0 to about 7.45 preferably about 7.4 respectively.
  • the above media is referred to as 293 SFM w/o insulin, w/o transferrin, w/o citrate chelate, w/o dextran sulfate, with L-glutamine and with PLURONIC F-68.
  • one or more of the replacement compounds of the invention and one or more metals can be added, for example, 25 ⁇ M 2-hydroxypyridine-N-oxide and 0.375 mg/L ZnSO 4 .7H 2 0 can be added for the transfection media.
  • the nucleic acid was prepared for transfection by combining 700 ⁇ l test media+60 ⁇ l DNA (the DNA used was plasmid pCMV-SPORT- ⁇ -gal available from Invitrogen Corporation, Carlsbad, Calif. Cat# 10586-014)+240 ⁇ l LIPOFECTAMINE 2000 (available from Invitrogen Corporation, Carlsbad, Calif. Cat# 11668-019) in a microfuge tube and incubating the mixture for 30 minutes at room temperature in order to form complexes of nucleic acid and transfection reagent.
  • the additions were always done by first adding the DNA to the tested media, then adding the LIPOFECTAMINE 2000 to the DNA-tested media mix.
  • the final cell concentration was 1 ⁇ 10 6 viable cells per ml
  • the final DNA concentration was 11 g per 1 ⁇ 10 6 viable cells
  • the final LIPOFECTAMINE 2000 concentration was 8 ⁇ l per 1 ⁇ 10 6 viable cells.
  • the final cell concentration in the suspension in the shaker flasks was 1 ⁇ 10 6 viable cells per ml, the final DNA concentration was 1 ⁇ g per 1 ⁇ 10 6 viable cells and the final LIPOFECTAMINE 2000 concentration was 8 ⁇ l per 1 ⁇ 10 6 viable cells.
  • samples were collected for ⁇ -galactosidase quantitation.
  • the contents of duplicate wells were pooled (i.e. ⁇ 2 ⁇ 10 6 cells for the assay) for each sample and transferred to a microfuge tube.
  • the cells were centrifuged, the supernatant was removed and the cells were re-suspended in 1 ml of D-PBS.
  • the cells were pelleted a second time and re-suspended in 1 ml of D-PBS.
  • the samples were then stored at ⁇ 70° C. Prior to the assay for ⁇ -galactosidase, samples were freeze-thawed a total of five times. Aliquots (15 ⁇ l and 25 ⁇ l) of each sample were assayed and the activity determined by comparison to a standard curve ranging from 0-70 ng ⁇ -galactosidase.
  • the third well of each treatment was used for in situ staining for ⁇ -galactosidase activity (i.e., ⁇ 1 ⁇ 10 6 cells).
  • 293 cells were grown in monolayer culture in the presence of the various replacement compounds indicated and transfection experiments performed as described above. The results are shown in Table 8 and the values are ⁇ -galactosidase activity expressed as nanograms of ⁇ -galactosidase per 2 ⁇ 10 6 cells. A comparison was made between the transfection efficiency of DNA complexes formed in D-MEM medium and DNA complexes formed in test medium. The replacement compounds were present at a concentration of 25 ⁇ M.
  • Transfection efficiency was generally better when test media rather than D-MEM was used for complexing the DNA and lipid.
  • the transferrin control (ID# A) worked best, reflecting that transferrin facilitates transfections. It should also be noted that cell growth in the above test media was comparable to growth in media containing transferrin and to growth in 293 SFM-II.
  • Base media for test media was 293-II+insulin+L-glutamine+PLURONIC F-68 ( ⁇ ) dextran sulfate ( ⁇ ) transferrin (+) replacement compound (A-M for test media, see table).
  • 293 cells were grown in monolayer culture in base medium supplemented with the indicated replacement compounds at the concentrations specified in Table 9.
  • the top number in the concentration column is the concentration of the first supplement and the second number in the concentration column is the concentration of the second supplement.
  • the effects of the medium used to form DNA complexes were tested. The results are presented as nanograms of ⁇ -galactosidase per 2 ⁇ 10 6 cells.
  • Ethyl maltol did not support growth at 100 ⁇ M. Growth in 50 ⁇ M was a little better than 25 ⁇ M but better for transfection in the presence of insulin (ID# B, C, H, P and Q) and about the same in presence of ZnSO 4 .
  • DPTA.FeSO 4 supported growth better in the presence of insulin than in the presence of ZnSO 4 , however, growth was acceptable in the presence of ZnSO 4 . Transfection results were much better when the chelate was present in combination with ZnSO 4 instead of insulin (see ID# F and L).
  • X-gal staining showed a transfection efficiency of ⁇ 40% in media with 1 ⁇ ZnSO 4 and of ⁇ 50-60% in media with insulin.
  • 2-hydroxypyridine at a concentration of 25 ⁇ M supported growth at concentrations of 1 ⁇ , 2 ⁇ and 5 ⁇ ZnSO 4 , but not at 0.5 ⁇ . Expression was about the same at 1 ⁇ , 2 ⁇ and 5 ⁇ ZnSO 4 (see H, I, and J). X-gal staining was only done on sample H (1 ⁇ ZnSO 4 ) and showed a transfection efficiency at 85-95%.
  • 2-hydroxypyridine at a concentration of 10 ⁇ M (sample K) supported growth in combination with 1 ⁇ ZnSO 4 . Expression was slightly lower than when 2-hydroxypyridine is at 25 ⁇ M (compare H to K).
  • DPTA.FeSO 4 supported growth at concentrations of 1 ⁇ , 2 ⁇ and 5 ⁇ ZnSO 4 , but not at 0.5 ⁇ . Expression decreased with increasing concentrations of ZnSO 4 , with 1 ⁇ being the best. X-gal staining showed a transfection efficiency of 50% (ID# N).
  • 293 cells were grown in monloayer culture in base media supplemented with the indicated chelator and metal salt at the indicated concentrations.
  • the concentration of the chelator is given in ⁇ moles/l and the concentration of the metal salt is given in ⁇ grams/ml.
  • a 10 ml flask of 293 cells in suspension culture in test media supplemented with +2-hydroxypyridine-N-oxide at 25 ⁇ M and ZnSO 4 at 0.375 ⁇ g/ml was transfected.
  • the concentration of the cells in the media was 1 ⁇ 10 6 cell/ml
  • the concentration of DNA used was 1 ⁇ g DNA/10 6 cells
  • the concentration of LIPOFECTAMINE 2000 used was 8 ⁇ l/10 6 cells.
  • Table 12 The results are shown in Table 12.
  • ⁇ -galactosidase expression was lower overall than in previous experiments but the trend seems similar. Again the data show that D-PBS should not be used for complexing in these experiments.
  • the ⁇ -galactosidase expression in the cells in suspension culture in shaker flasks was substantially higher than that seen in monolayer culture. It should be emphasized that the flask was put on a shaker platform (125 rpm) from the time of transfection to the time of collection 24 hrs later. The improved transfection and resulting expression can be due to the cells being in better shape in the shaker flask than in the 24-well plates over the incubation period.
  • Expression in media A was lower than in the previous experiments where expression was 29118 ng/ ⁇ 2 ⁇ 10 6 cells.
  • the volume used in that experiment was 10 ml in a 125 ml shaker flask.
  • Growth data for the adaptation of the cells to the test media shows that cells densities are similar for all media A-F.
  • X-gal staining of cells shows similar transfection efficiencies.
  • FIGS. 1A and 1B show the cell density at each of the first 3 passages in the test media.
  • FIG. 1A shows the cell densities of the 293-F cells while FIG. 1B shows the densities of the 293-H cells
  • the 293-F cells adapted well into the test media. There was some clumping but vortexing was enough to get a single cell suspension.
  • the 293-H cells grew to similar densities in the test media as in the 293SFM II. However, there was a lot of clumping that could not be resolved by vortexing.
  • FIGS. 2A and 2B a growth curve comparing the growth of 293-F&H cells in a single passage is shown. 25 ml cultures in 125 ml flasks were seeded with 3 ⁇ 10 5 viable cells/ml in triplicate and 1 ml aliquots were taken each day and the viable cells counted.
  • Catalog media was 293 SFM TI supplemented with 4 mM glutamine and test media A, B and C were as in Table 14.
  • FIG. 2A shows the results obtained with 293-F cells and FIG. 2B shows the results obtained with 293-H cells. In all cases, the test media supported cell growth more effectively than the catalog media.
  • Cells were seeded at a density of 3 ⁇ 10 5 cells/ml and counted after 4 days of growth in media containing in varying amounts of ZnSO 4 , ZnCl 2 or Na 2 SO 4 in order to determine which ion Zn or SO 4 2 ⁇ in zinc sulfate promotes cell growth.
  • Test media supplemented with 25 ⁇ m 2-hydroxypyridine-N-oxide and the indicated salts were used to assess the effects of zinc ion and sulfate ion in suspension culture. As shown in FIG. 3 , ZnCl 2 could effectively substitute for zinc sulfate whereas Na 2 SO 4 resulted in less cell growth.
  • 293 cells in suspension were grown in the indicated amount of media in 125 ml shake flasks. Results below are averages of duplicate flasks (A-25 m 1 is for single flask only). The cell densities (at time of set-up) in all cases was 1 ⁇ 10 6 viable cells per ml. The DNA and lipid concentrations used were 1 ⁇ g and 8 ⁇ g1 respectively per 1 ⁇ 10 6 viable cells.
  • Flasks set-up in media A were for testing effect of cell suspension volume in the flasks and its effects on transfections. There appears to be no significant difference between a 10 ml and 15 ml volume. However the decline when at 20 ml and then increase at 25 m 1 is not explained. Also note that 25 ml was not set-up in duplicate.
  • Conditioned media consisted of the media the culture had been growing in until day of transfection set-up. This was done to evaluate whether transfections can be carried out without having to change the culture medium (i.e. spin cells down, re-suspend, etc.). The results show more ⁇ -galactosidase produced when in conditioned media for B and comparable results in media C thus supporting the possibility of just adding the lipid-DNA complex to culture when a certain cell density has been reached.
  • the first protocol required the cells to be suspended in a reduced volume of medium and contacted with a nucleic acid and transfection reagent. After an incubation period to allow uptake of the nucleic acid, additional medium was added to bring the culture volume up to a final volume.
  • the second protocol called for the introduction of a nucleic acid and transfection reagent into the culture at the final volume. Since the second protocol requires one less medium addition step, it requires less manipulation of the cells and less hands-on time.
  • Nucleic acid:transfection reagent complexes were prepared as follows: 30 ⁇ g DNA was added to 1 ml of minimal medium (OPTIMEM, Invitrogen Corporation, Carlsbad, Calif.) 200 ⁇ l transfection reagent (LIPOFECTAMINE 2000, Invitrogen Corporation, Carlsbad, Calif.) was added to 1 ml minimal medium. The diluted DNA and transfection reagent were combined, mixed gently and incubated at room temperature for 20 minutes. The complexes were added to the cells and mixed. Cells were incubated at 37° C.
  • PROTOCOL 1 24 hours mg protein/ 0.8 0.9 10 6 cells ⁇ g ⁇ -gal/ 22.7 25.0 10 6 cells ⁇ g ⁇ -gal/ 28.7 28.7 mg protein expected mg 22.7 25 ⁇ -gal/L cells 48 hours mg protein/ 1.1 1.1 10 6 cells ⁇ g ⁇ -gal/ 50.1 52.9 10 6 cells ⁇ g ⁇ -gal/ 46.4 46.8 mg protein expected mg 50.1 52.9 ⁇ -gal/L cells mg ⁇ -gal/ 1.5 1.6 30 ml culture
  • protocol 2 was used in the following experiment.
  • transformations were conducted using 100 ⁇ l, 150 ⁇ l and 200 ⁇ l of LIPOFECTAMINE 2000.
  • the indicated amount of transfection reagent was diluted in 1 ml minimal medium and 30 ⁇ g DNA was diluted in 1 ml medium as described above.
  • the dilutions were combined and mixed gently and incubated 20 minutes at room temperature.
  • the nucleic acid complexes were added to 3 ⁇ 10 7 cells in 28 ml of medium. Aliquots were taken at 25 hours and 49 hours and assayed above. The results are presented in Table 17.
  • transfection reagent did not seem to dramatically affect the expression levels of ⁇ -galactosidase (compare 1.2 mg/30 ml of culture at 100 ⁇ l transfection reagent to 1.3 mg/30 ml of culture at 200 ⁇ l of transfection reagent). In contrast, the presence of butyrate in the culture medium more than doubled the production of ⁇ -galactosidase from 1.3 mg/30 ml of culture to 2.7 mg/30 ml of culture.

Abstract

The present invention is directed generally to cell culture media (particularly serum free, non animal derived, and/or chemically defined media) which are useful for introducing macromolecules and compounds (e.g., nucleic acid molecules) into cells (e.g., eukaryotic cells). According to the invention, such introduction can take place in the presence of said medium. Cells containing such introduced materials can then be cultured in the medium and the effect of the introduced materials on the cells can be measured or determined. In particular, the invention allows introduction of nucleic acid molecules (e.g., vectors) into cells (particularly eukaryotic cells) and expression of proteins encoded by the nucleic acid molecules in the cells. The invention obviates the need to change the cell culture medium each time a different procedure is performed with the cells (e.g., culturing cells vs. transfecting cells). The invention thus provides efficient and high throughput methods to transform/transfect culture and cells avoiding the need for multiple manipulations and transfers of cells during transfection and expression studies. The invention also relates to compositions and kits useful for culturing and transforming/transfecting cells.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
    • This is a continuation application of U.S. application Ser. No. 10/105,937, filed Mar. 26, 2002, which claims the benefit of Provisional Application No. 60/278,754 filed Mar. 27, 2001, all of which are incorporated herein by reference in their entireties.
    BACKGROUND OF THE INVENTION
  • 1. Field of the Invention
  • The present invention relates to the field of cell culture. In particular, the present invention provides media suitable for the culture and transfection of cells.
  • 2. Related Art
  • Cell culture media provide the nutrients necessary to maintain and grow cells in a controlled, artificial and in vitro environment. Characteristics and formulations of cell culture media vary depending upon the particular cellular requirements. Important parameters include osmolarity, pH, and nutrient compositions.
  • Cell culture medium formulations have been well documented in the literature and a large number of media are commercially available. In early cell culture work, medium formulations were based upon the chemical composition and physicochemical properties (e.g., osmolality, pH, etc.) of blood and were referred to as “physiological solutions” (Ringer, S., J. Physiol. 3:380-393 (1880); Waymouth, C., In: Cells and Tissues in Culture, Vol. 1, Academic Press, London, pp. 99-142 (1965); Waymouth, C., In Vitro 6:109-127 (1970)). However, cells in different tissues of a mammalian body are exposed to different microenvironments with respect to oxygen/carbon dioxide partial pressure and concentrations of nutrients, vitamins, and trace elements; accordingly, successful in vitro culture of different cell types may require the use of different medium formulations. Typical components of cell culture media include amino acids, organic and inorganic salts, vitamins, trace metals, sugars, lipids and nucleic acids, the types and amounts of which may vary depending upon the particular requirements of a given cell or tissue type.
  • Medium formulations have been used to cultivate a number of cell types including animal, plant and bacterial cells. Cultivated cells have many uses including the study of physiological processes and the production of useful biological substances. Examples of such useful products include monoclonal antibodies, hormones, growth factors, enzymes and the like. Such products have many commercial and therapeutic applications and, with the advent of recombinant DNA technology, cells can be engineered to produce large quantities of these products. Cultured cells are also routinely used for the isolation, identification and growth of viruses which can be used as vectors and/or vaccines. Thus, the ability to cultivate cells in vitro is not only important for the study of cell physiology, but is also necessary for the production of useful substances which may not otherwise be obtained by cost-effective means.
  • Among the various cell types that have been grown using in vitro cell culture media, of particular interest are cells derived from the epithelium. The epithelium lines the internal and external surfaces of the organs and glands of higher organisms. Because of this localization at the external interface between the environment and the organism (e.g., the skin) or at the internal interface between an organ and the interstitial space (e.g., the intestinal mucosal lining), the epithelium has a major role in the maintenance of homeostasis. The epithelium carries out this function, for example, by regulating transport and permeability of nutrients and wastes (Freshney, R. I., in: Culture of Epithelial Cells, Freshney, R. I., ed., New York: Wiley-Liss, pp. 1-23 (1992)).
  • The cells making up the epithelium are generically termed epithelial cells. These cells can be present in multiple layers as in the skin, or in a single layer as in the lung alveoli. As might be expected, the structure, function and physiology of epithelial cells are often tissue-specific. For example, the epidermal epithelial cells of the skin are organized as stratified squamous epithelium and are primarily involved in forming a protective barrier for the organism, while the secretory epithelial cells of many glands are often found in single layers of cuboidal cells that have a major role in producing secretory proteins and glycoproteins. Regardless of their location or function, however, epithelial cells are usually regenerative. That is, under normal conditions, or in response to injury or other activating stimulus, epithelial cells are capable of dividing or growing. This regenerative capacity has facilitated the in vitro manipulation of epithelial cells, to the point where a variety of primary epithelial cells and cell lines have been successfully cultivated in vitro (Freshney, Id.).
  • While the isolation and use of a variety of epithelial cells and epithelial cell lines have been reported in the literature, the human embryonic kidney cell line 293 (“293 cells”), which exhibits epithelial morphology, has proven particularly useful for studies of the expression of exogenous ligand receptors, production of viruses and expression of allogeneic and xenogeneic recombinant proteins. For example, U.S. Pat. No. 5,166,066 describes the construction of a stable 293 cell line comprising functional GABA receptors that include a benzodiazepine binding site that have proven useful in identification and screening of candidate psychoactive drugs. 293 cells have also been used to produce viruses such as natural and recombinant adenoviruses (Garnier, A., et al., Cytotechnol. 15:145-155 (1994); Bout, A., et al., Cancer Gene Therapy 3(6):S24, abs. P-52 (1996); Wang, J.-W., et al., Cancer Gene Therapy 3(6):S24, abs. P-53 (1996)), which can be used for vaccine production or construction of adenovirus vectors for recombinant protein expression. Finally, 293 cells have proven useful in large-scale production of a variety of recombinant human proteins (Berg, D. T., et al., BioTechniques 14(6):972-978 (1993); Peshwa, M. V., et al., Biotechnol. Bioeng. 41:179-187 (1993); Garnier, A., et al., Cytotechnol. 15:145-155 (1994)).
  • Cells loosely called fibroblasts have been isolated from many different tissues and are understood to be connective tissue cells. It is clearly possible to cultivate cell lines, loosely termed fibroblastic cells, from embryonic and adult tissues. Fibroblasts characteristically have a “spindle” appearance. Fibroblast-like cells have morphological characteristics typical of fibroblast cells. Under a light microscope the cells appear pointed and elongated (“spindle shaped”) when they grow as a monolayer on the surface of a culture vessel. Cell lines can be regarded as fibroblast or fibroblast-like after confirmation with appropriate markers, such as collagen, type I ((Freshney, R. I., in: Culture of Epithelial Cells, Freshney, R. I., ed., New York: Wiley-Liss, pp. 1-23 (1987)).
  • CHO cells have been classified as both epithelial and fibroblast cells derived from the Chinese hamster ovary. A cell line started from Chinese hamster ovary (CHO-K1) (Kao, F.-T. And Puck, T. T., Proc. Natl. Acad. Sci. USA 60:1275-1281 (1968) has been in culture for many years but its identity is still not confirmed.
  • Most primary mammalian epithelial cells, mammalian fibroblast cells, epithelial cell lines, and fibroblast cell lines are typically grown in monolayer culture. For some applications, however, it would be advantageous to cultivate such cells as suspension cultures. For example, suspension cultures grow in a three-dimensional space. Monolayer cultures in similar-sized vessels, however, can only grow two-dimensionally on the vessel surface. Thus, suspension cultures can result in higher cell yields and, correspondingly, higher yields of biologicals (e.g., viruses, recombinant polypeptides, etc.) compared to monolayer cultures. In addition, suspension cultures are often easier to feed and scale-up, via simple addition of fresh culture media (dilution subculturing) to the culture vessel rather than trypsinization and centrifugation as is often required with monolayer cultures. The ease of feeding and the ease with which suspension cultures can be scaled up represent a substantial saving in time and labor for handling a comparable number of cells.
  • Many anchorage-dependent cells, such as primary epithelial cells, primary fibroblast cells, epithelial cell lines, and fibroblast cell lines, however, are not easily adapted to suspension culture. Since they are typically dependent upon anchorage to a substrate for optimal growth, growth of these cells in suspension can require their attachment to microcarriers such as latex or collagen beads. Thus, cells grown in this fashion, while capable of higher density culture than traditional monolayer cultures, are still technically attached to a surface; subculturing of these cells therefore requires similar steps as those used for the subculturing of monolayer cultures. Furthermore, when large batch or fermenter cultures are established, a large volume of microcarriers often settles to the bottom of the culture vessel, thereby requiring a more complicated agitation mechanism to keep the microcarriers (and thus, the cells) in suspension without causing shear damage to the cells (Peshwa, M. V., et al., Biotechnol. Bioeng. 41:179-187 (1993)).
  • Although many transformed cells are capable of being grown in suspension (Freshney, R. I., Culture of Animal Cells: A Manual of Basic Technique, New York: Alan R. Liss, Inc., pp. 123-125 (1983)), successful suspension cultures often require relatively high-protein media or supplementation of the media with serum or serum components (such as the attachment factors fibronectin and/or vitronectin), or sophisticated perfusion culture control systems (Kyung, Y.-S., et al., Cytotechnol. 14:183-190 (1994)), which can be disadvantageous. In addition, many epithelial cells when grown in suspension form aggregates or “clumps” which can interfere with successful subculturing and reduce growth rate and production of biologicals by the cultures. When clumping occurs, the overall cellular surface area exposed to medium is decreased and the cells are deprived of nutrition and are unable to efficiently exchange waste into the medium. As a result, growth slows, diminished cell densities are obtained, and protein expression is compromised.
  • Typically, cell culture media formulations are supplemented with a range of additives, including undefined components such as fetal bovine serum (FBS) (5-20% v/v) or extracts from animal embryos, organs or glands (0.5-10% v/v). While FBS is the most commonly applied supplement in animal cell culture media, other serum sources are also routinely used, including newborn calf, horse and human. Organs or glands that have been used to prepare extracts for the supplementation of culture media include submaxillary gland (Cohen, S., J. Biol. Chem. 237:1555-1565 (1961)), pituitary (Peehl, D. M., and Ham, R. G., In Vitro 16:516-525 (1980); U.S. Pat. No. 4,673,649), hypothalamus (Maciag, T., et al., Proc. Natl. Acad. Sci. USA 76:5674-5678 (1979); Gilchrest, B. A., et al., J. Cell Physiol. 120:377-383 (1984)), ocular retina (Barretault, D., et al., Differentiation 18:29-42 (1981)) and brain (Maciag, T., et al., Science 211:1452-1454 (1981)). These types of chemically undefined supplements serve several useful functions in cell culture media (Lambert, K. J. et al., In: Animal Cell Biotechnology, Vol. 1, Spier, R. E. et al., Eds., Academic Press New York, pp. 85-122 (1985)). For example, these supplements provide carriers or chelators for labile or water-insoluble nutrients; bind and neutralize toxic moieties; provide hormones and growth factors, protease inhibitors and essential, often unidentified or undefined low molecular weight nutrients; and protect cells from physical stress and damage. Thus, serum or organ/gland extracts are commonly used as relatively low-cost supplements to provide an optimal culture medium for the cultivation of animal cells.
  • Unfortunately, the use of serum or organ/gland extracts in tissue culture applications has several drawbacks (Lambert, K. J. et al., In: Animal Cell Biotechnology, Vol. 1, Spier, R. E. et al., Eds., Academic Press New York, pp. 85-122 (1985)). For example, the chemical compositions of these supplements and sera vary between lots, even from a single manufacturer. The supplements can also be contaminated with infectious agents (e.g., mycoplasma and viruses) which can seriously undermine the health of the cultured cells and the quality of the final product. The use of undefined components such as serum or animal extracts also prevents the true definition and elucidation of the nutritional and hormonal requirements of the cultured cells, thus eliminating the ability to study, in a controlled way, the effect of specific growth factors or nutrients on cell growth and differentiation in culture. Moreover, undefined supplements prevent the researcher from studying aberrant growth and differentiation and the disease-related changes in cultured cells. Finally and most importantly to those employing cell culture media in the industrial production of biological substances, serum and organ/gland extract supplementation of culture media can complicate and increase the costs of the purification of the desired substances from the culture media due to nonspecific co-purification of serum or extract proteins.
  • Improved levels of recombinant protein expression are obtained from cells grown in serum-free medium, relative to the level of expression seen in cells grown in medium supplemented with serum (Battista, P. J. et al., Am. Biotech. Lab. 12:64-68 (1994)). However, serum-free media can still contain one or more of a variety of animal-derived components, including albumin, fetuin, various hormones and other proteins. The presence of proteins or peptides makes purification of recombinant protein difficult, time-consuming, and expensive.
  • To overcome these drawbacks of the use of serum or organ/gland extracts, a number of so-called “defined” media have been developed. These media, which often are specifically formulated to support the culture of a single cell type, contain no undefined supplements and instead incorporate defined quantities of purified growth factors, proteins, lipoproteins and other substances usually provided by the serum or extract supplement. Since the components (and concentrations thereof) in such culture media are precisely known, these media are generally referred to as “defined culture media.” Sometimes used interchangeably with “defined culture media” is the term “serum-free media” or “SFM.” A number of SFM formulations are commercially available, such as those designed to support the culture of endothelial cells, keratinocytes, monocytes/macrophages, lymphocytes, hematopoietic stem cells, fibroblasts, chondrocytes or hepatocytes which are available from Invitrogen Corporation, Carlsbad, Calif. The distinction between SFM and defined media, however, is that SFM are media devoid of serum and protein fractions (e.g., serum albumin), but not necessarily of other undefined components such as organ/gland extracts. Indeed, several SFM that have been reported or that are available commercially contain such undefined components, including several formulations supporting in vitro culture of keratinocytes (Boyce, S. T., and Ham, R. G., J. Invest. Dermatol. 81:33 (1983); Wille, J. J., et al., J. Cell. Physiol. 121:31 (1984); Pittelkow, M. R., and Scott, R. E., Mayo Clin. Proc. 61:771 (1986); Pirisi, L., et al., J. Virol. 61:1061 (1987); Shipley, G. D., and Pittelkow, M. R., Arch. DermatoL 123:1541 (1987); Shipley, G. D., et al., J. Cell. Physiol. 138:511-518 (1989); Daley, J. P., et al., FOCUS (GIBCO/LTI) 12:68 (1990); U.S. Pat. Nos. 4,673,649 and 4,940,666). SFM thus cannot be considered to be defined media in the true definition of the term.
  • Defined media generally provide several distinct advantages to the user. For example, the use of defined media facilitates the investigation of the effects of a specific growth factor or other medium component on cellular physiology, which can be masked when the cells are cultivated in serum- or extract-containing media. In addition, defined media typically contain much lower quantities of protein (indeed, defined media are often termed “low protein media”) than those containing serum or extracts, rendering purification of biological substances produced by cells cultured in defined media far simpler and less expensive.
  • Some extremely simple defined media, which consist essentially of vitamins, amino acids, organic and inorganic salts and buffers have been used for cell culture. Such media (often called “basal media”), however, are usually seriously deficient in the nutritional content required by most animal cells. Accordingly, most defined media incorporate into the basal media additional components to make the media more nutritionally complex, but to maintain the serum-free and low protein content of the media. Examples of such components include bovine serum albumin (BSA) or human serum albumin (HSA); certain growth factors derived from natural (animal) or recombinant sources such as epidermal growth factor (EGF) or fibroblast growth factor (FGF); lipids such as fatty acids, sterols and phospholipids; lipid derivatives and complexes such as phosphoethanolamine, ethanolamine and lipoproteins; protein and steroid hormones such as insulin, hydrocortisone and progesterone; nucleotide precursors; and certain trace elements (reviewed by Waymouth, C., in: Cell Culture Methods for Molecular and Cell Biology, Vol. 1: Methods for Preparation of Media, Supplements, and Substrata for Serum-Free Animal Cell Culture, Barnes, D. W., et al., eds., New York: Alan R. Liss, Inc., pp. 23-68 (1984), and by Gospodarowicz, D., Id., at pp 69-86 (1984)).
  • The use of animal protein supplements in cell culture media, however, also has certain drawbacks. For example, there is a risk that the culture medium and/or products purified from it can be immunogenic, particularly if the supplements are derived from an animal different from the source of the cells to be cultured. If biological substances to be used as therapeutics are purified from such culture media, certain amounts of these immunogenic proteins or peptides can be co-purified and can induce an immunological reaction, up to and including anaphylaxis, in an animal receiving such therapeutics.
  • To obviate this potential problem, supplements derived from the same species as the cells to be cultured can be used. For example, culture of human cells can be facilitated using HSA as a supplement, while media for the culture of bovine cells would instead use BSA. This approach, however, runs the risks of introducing contaminants and adventitious pathogens into the culture medium (such as Creutzfeld-Jakob Disease (CJD) from HSA preparations, or
    • Bovine Spongiform Encephalopathy (“Mad Cow Disease”) prion from BSA preparations), which can obviously negatively impact the use of such media in the preparation of animal and human therapeutics. In fact, for such safety reasons, the biotechnology industry and government agencies are increasingly regulating, discouraging and even forbidding the use of cell culture media containing animal-derived proteins which can contain such pathogens.
  • To overcome the limitations of the use of animal proteins in SFM, several attempts have been made to construct animal cell culture media that are completely free of animal proteins. For example, some culture media have incorporated extracts of yeast cells into the basal medium (see, for example, U.K. Patent Application No. GB 901673; Keay, L., Biotechnol. Bioengin. 17:745-764 (1975)) to provide sources of nitrogen and other essential nutrients. In another approach, hydrolysates of wheat gluten have been used, with or without addition of yeast extract, to promote in vitro growth of animal cells (Japanese Patent Application No. JP 2-49579). Still other media have been developed in which serum is replaced by enzymatic digests of meat, or of proteins such as α-lactalbumin or casein (e.g., peptone), which have been traditionally used in bacterial culture (Lasfargues, E. Y., et al., In Vitro 8(6):494-500 (1973); Keay, L., Biotechnol. Bioeng. 17:745-764 (1975); Keay, L., Biotechnol. Bioeng. 19:399-411 (1977); Schlager, E.-J., J. Immunol. Meth. 194:191-199 (1996)). None of these approaches, however, provided a culture medium optimal for the cultivation of a variety of animal cells. Moreover, extracts from certain plants, including wheat, barley, rye and oats have been shown to inhibit protein synthesis in cell-free systems derived from animal cells (Coleman, W. H., and Roberts, W. K., Biochim. Biophys. Acta 696:239-244 (1982)), suggesting that the use of peptides derived from these plants in cell culture media can actually inhibit, rather than stimulate, the growth of animal cells in vitro. More recently, animal cell culture SFM formulations comprising rice peptides have been described and shown to be useful in cultivation of a variety of normal and transformed animal cells (see U.S. Pat. No. 6,103,529, incorporated herein by reference in its entirety).
  • Notwithstanding the potential difficulties posed by the addition of animal derived supplements to cell culture media, such supplements are in routine use. One such supplement that is frequently added to defined media is transferrin. Transferrin functions in vivo to deliver iron to cells. The mechanism of iron uptake by mammalian cells has been reviewed (Qian, Z. M. and Tang, P. L. (1995) Biochim. Biophys. Acta 1269, 205-214). As iron is required as a co-factor in numerous metabolic processes including energy generation and oxidative respiration, serum-free media are often supplemented with transferrin in order to deliver the requisite iron for the successful cultivation of most cells in vitro. Concern about various potential adventitious agents in preparations of transferrin has stimulated a search for other natural iron carrier compounds which can be used as a substitute for transferrin. This search is complicated by the fact that the natural iron carriers are often derived from serum and thus are subject to the above-described limitations of serum supplementation.
  • To overcome the limitations of using naturally derived metal carriers, certain metal binding compounds are being explored for use in supplying metals, particularly zinc, iron, manganese and magnesium, to cultured cells. Simple carriers such as chelating agents (e.g., EDTA) and certain acids or salts thereof (e.g., citrate, picolinate, and derivatives of benzoic acid or hydroxamic acid) have been shown to be useful in certain serum-free growth media (see U.S. Pat. Nos. 5,045,454 and 5,118,513; Testa et al., Brit. J. Haematol. 60:491-502, (1985); Ganeshaguru et al., Biochem. Pharmacol. 29:1275-1279 (1980); White et al., Blood 48:923-929 (1976)).
  • Although these references disclose some metal carriers, the interpretation of the data is complicated by several experimental factors. The data were gathered from a limited number of cell lines and show results of a single passage. In addition, the media were supplemented with serum. Serum inherently contains transferrin and other potential iron carriers. There is a “carry-over effect” on growth of cells which have been cultured in serum-supplemented medium, even after one or two passages in the absence of serum or transferrin (see, for example, Keenan, J. and Clynes, M. (1996) In Vitro Cell Dev. Biol-Animal 32, 451-453). Other known metal binding compounds have been used medicinally to remove iron from the body and not for delivery. Unfortunately, many of these simple iron chelating compounds do not provide sufficient iron availability to, or uptake by, cultured cells.
  • Once a suitable medium formulation for the growth of a particular cell type has been determined, it is frequently necessary to alter the cell in question so as to optimize the production of a desired biological substance. A critical step in the effective production and purification of biological substances is the introduction of one or more macromolecules (e.g., peptides, proteins, nucleic acids, etc.) into the cell in which the material will be produced. This can be accomplished by a variety of methods. One widely used method to introduce macromolecules into a cell is known as transfection.
  • Typically, the target cell is grown to a desired cell density in a cell culture medium optimized for growth of the cell. Once the desired density is reached, the medium is exchanged for a medium optimized for the transfection process. Under most circumstances, the medium used for transfection does not support the growth of the cells but the transfection medium is merely used for the purpose of introducing nucleic acids into the cells. As a result, the process generally requires collecting the cells from the culture, usually by centrifugation, washing the cells to remove traces of the growth medium, suspending the cells in a transfection medium in the presence of the macromolecule of interest, incubating the cells in the transfection medium for a period of time sufficient for the uptake of the macromolecule, optionally, removing the transfection medium and washing the remnants of the transfection medium from the cells and then re-suspending the transfected cells in a growth medium. The steps of exchanging the growth media for transfection media, washing the cells, and exchanging the transfection media back to a growth media require a great deal of hands-on manipulation of the cells thereby adding substantially to the time and expense of recombinant DNA technology.
  • As an historical example, 293 cells have been cultivated in monolayer cultures in a serum-supplemented version of a complex medium (i.e., DMEM). When grown in suspension, 293 cells have a tendency to aggregate into large clusters of cells. The formation of these large cell aggregates reduces the viability of the cells. Since the cells in the center of the aggregates are not directly exposed to the medium, these cells have limited access to nutrients in the medium and have difficulty in exchanging waste into the medium. In addition, this reduced access to the medium makes cells in clusters unsuitable for genetic manipulation by factors introduced into the medium (i.e., for transformation by nucleic acids). As a result of these difficulties, 293 cells have not generally been used in suspension culture for the production of biological materials.
  • Thus, there still remains a need in the art for a cell medium that permits the growth of eukaryotic cells in suspension while permitting the transfection of the cells with a reduced amount of manipulation. Such a medium should preferably be a serum-free and/or chemically defined and/or protein-free medium and/or a medium lacking animal derived materials which facilitates the growth of mammalian cells to high density and/or increases the level of expression of recombinant protein, reduces cell clumping, and which does not require supplementation with animal proteins, such as serum, transferrin, insulin and the like. Preferably a medium of this type will permit the suspension cultivation of mammalian cells that are normally anchorage-dependent, including epithelial cells and fibroblast cells, such as 293 cells and CHO cells. Such culture media will facilitate studies of the effects of growth factors and other stimuli on cellular physiology, will allow easier and more cost-effective production and purification of biological substances (e.g., viruses, recombinant proteins, etc.) produced by cultured mammalian cells in the biotechnology industry, and will provide more consistent results in methods employing the cultivation of mammalian cells. These needs and others are met by the present invention.
  • SUMMARY OF THE INVENTION
  • The present invention provides a cell culture medium, wherein the medium supports introduction of one or more macromolecules into at least one eukaryotic cell in culture and supports cultivation of the at least one cell subsequent to the introduction, wherein growth of the at least one cell continues in the medium in the absence of the medium being with fresh medium. In a preferred embodiment, it is not necessary to remove medium used during the introduction from the presence of the at least one cell to support growth of the at least one cell. In another preferred embodiment, after the introduction, growth is accomplished in cultivation in a volume of medium that is about the same volume up to no more than about 10 times the volume of the medium in which the introduction occurred. Using the medium of the present invention, it is not necessary to replenish, replace or supplement the medium after one has introduced nucleic acid into cells, and before cells into which nucleic acid has been introduced are further cultured to express the nucleic acid.
  • The present invention also provides a method of making a medium comprising admixing water and at least one ingredient selected from the group consisting of an amino acid, a sugar, a fatty acid (such as linoleic acid, linolenic acid, and especially fatty acids of 12, 14, 16, 18, 19, 20 or 24 carbon atoms, each carbon chain branched or unbranched), arachadonic acid, palmitoleic acid, oleic acid, polyenoic acid (e.g., palmitoleic acid, oleic acid, linoleic acid and/or linolenic acid), a vitamin (such as pyridoxine, niacinamide, thiamine, etc.), a pH buffer, a surfactant, a trace metal or salt or hydrate thereof, an amine compound, a growth factor, an agent to control osmolarity and/or ionic strength and/or maintain membrane potential, a flavin, a compound that participates in or is a product of the glycolytic pathway, an alcoholamine, a cyclic alcohol, a phospholipid or portion thereof (such as choline chloride), a salt of selenious acid (such as sodium selenite), a divalent or trivalent cation (such as manganese, calcium, zinc, magnesium, copper or iron), a pteridine derivative (such as folic acid), a valeric acid (such as lipoic acid) and/or a coenzyme (such as riboflavin), wherein the medium supports (a) the introduction of at least one macromolecule into eukaryotic cells in culture, (b) the cultivation of cells into which at least one macromolecule is introduced, and optionally (c) the expression of a nucleic acid in cells into which a macromolecule is introduced to form a protein product, wherein medium containing the at least one molecule does not need to be removed from the culture and replaced with fresh medium after introduction of the at least one molecule into cells and prior to cultivation and optimal expression of the nucleic acid.
  • The present invention also provides the medium obtained by admixing water and at least one ingredient selected from the group consisting of an amino acid, a sugar, a fatty acid (such as linoleic acid, linolenic acid, and especially fatty acids of 12, 14, 16, 18, 19, 20 or 24 carbon atoms, each carbon chain branched or unbranched), arachadonic acid, palmitoleic acid, oleic acid, polyenoic acid (e.g., palmitoleic acid, oleic acid, linoleic acid and/or linolenic acid), a vitamin (such as pyridoxine, niacinamide, thiamine, etc.), a pH buffer, a surfactant, a trace metal or salt or hydrate thereof, an amine compound, a growth factor, an agent to control osmolarity and/or ionic strength and/or maintain membrane potential, a flavin, a compound that participates in or is a product of the glycolytic pathway, an alcoholamine, a cyclic alcohol, a phospholipid or portion thereof (such as choline chloride), a salt of selenious acid (such as sodium selenite), a divalent or trivalent cation (such as manganese, calcium, zinc, magnesium, copper or iron), a pteridine derivative (such as folic acid), a valeric acid (such as lipoic acid) and/or a coenzyme (such as riboflavin), wherein the medium supports (a) the introduction at least one macromolecule into at least one eukaryotic cell in culture, (b) the cultivation of at least one cell into which at least one macromolecule is introduced, and (c) the expression of a nucleic acid in cells into which a macromolecule is introduced, wherein medium containing the at least one macromolecule does not need to be removed from the culture and replaced with fresh medium after introduction of the at least one macromolecule into cells and prior to cultivation and optimal expression of the nucleic acid.
  • The present invention also provides a method of cultivating eukaryotic cells comprising: (a) contacting the cells with the cell culture medium of the present invention; (b) maintaining the cells under conditions suitable to support cultivation of the cells in culture; and (c) optionally expressing a nucleic acid to form a protein product.
  • The present invention also provides a method for introducing one or more macromolecules into at least one eukaryotic cell in culture, the method comprising: (a) culturing at least one eukaryotic cell in the medium of claim 1 in culture; (b) introducing at least one macromolecule into the culture under conditions sufficient to cause one or more of the at least one macromolecule to be introduced in the at least one cell; and (c) cultivating the at least one cell in the medium to produce a product whose production is controlled by the at least one molecule, wherein growth of the at least one cell continues in the medium in the absence of the medium being with fresh medium, wherein it is not necessary to remove medium used during the introduction from the presence of the at least one cell to support growth of the at least one cell, and/or wherein after the introduction, growth is accomplished in cultivation in a volume of medium that is about the same volume up to no more than about 10 times the volume of the medium in which the introduction occurred.
  • The present invention also provides a kit for the cultivation and transfection of cells in vitro, the kit comprising the cell culture medium of the present invention, and optionally further comprising one or more of: one or more agents for the introduction of at least one molecule into a cell, one or more macromolecules, at least one cell, and instructions for culturing the at least one cell in culture and/or for introducing at least one macromolecule into at least one cell in culture.
  • The present invention also provides a composition comprising the cell culture medium of the present invention and at least one component selected from the group consisting of at least one eukaryotic cell, one or more agents for the introduction of at least one macromolecule into at least one cell, and one or more macromolecules.
  • Other embodiments of the present invention will be apparent to one of ordinary skill in light of the following drawings and description of the invention, and of the claims.
  • BRIEF DESCRIPTION OF THE DRAWINGS/FIGURES
  • FIG. 1A is a bar graph showing a plot of the number of 293-F cells/ml as a function of passage number in the medium of the present invention. FIG. 1B is a bar graph showing a plot of the number of 293-H cells/ml as a function of passage number in the medium of the present invention. Growth for 293-F and 293-H cells cultured in 293 SFM II medium (Invitrogen Corp., Carlsbad, Calif.) vs. prototype media A, B and C. 15 ml cultures were seeded at 3×105 viable cells per ml in 125 ml shaker flasks in the media one passage after recovery from cryopreservation. Cells were counted and split every four days to 3×105 viable cells per ml. At passage 3, cells were used to seed cultures for a growth curve. In FIG. 1A, at each passage number, the respective bars correspond, from left to right, to 293 SFM II medium, medium A, medium B, and medium C. In FIG. 1B, at each passage number, the respective bars correspond, from left to right, to 293 SFM II medium, and medium A.
  • FIG. 2A is a graph showing the number of viable 293-F cells/ml as a function of the number of days cultured in the medium of the invention. FIG. 2B is a graph showing the number of viable 293-H cells/ml as a function of the number of days cultured in the medium of the invention. Growth for 293-F and 293-H cells cultured in 293 SFM II medium (Invitrogen Corp., Carlsbad, Calif.) vs. prototype media A, B and C. 15 ml cultures were seeded at 3×105 viable cells per ml in 125 ml shaker flasks in triplicate and 1 ml samples taken each day for viabilities and counts. Cells were cultured in 293 SFM II medium supplemented with 4 mM L-Glutamine or in one of the tested media A, B or C.
  • FIG. 3 is a bar graph showing a plot of the number of 293-F cells/ml after each of three passages in the medium of the invention supplemented with various salts. For each medium, the three bars correspond, from left to right, to passage 1 (p1), passage 2, (p2) and passage 3 (p3).
  • FIG. 4A is a bar graph showing a plot of 293-F cell density (cells/ml) in pilot lot 1 (PL1), pilot lot 2 (PL2) and pilot lot (PL) 000458 media. FIG. 4B is a bar graph showing a plot of % viable 293-F cells in PL1, PL-2 and PL000458 media. FIG. 4C is a bar graph showing a plot of 293-F cell density (cells/ml) in PL-2 and PL-2A (+insulin) media. FIG. 4D is a bar graph showing a plot of % viable 293-F cells in PL-2 and PL-2A (+insulin) media.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides improved medium formulations for the growth of both eukaryotic and prokaryotic cells. The inventive media support cell growth, introduction of macromolecules into cells in culture and cell cultivation without requiring replenishment, replacement, supplementation, or changing medium between growth, introduction and/or cultivation. The media of the present invention can be used to support or enhance the growth and cultivation of any cell. The present invention also provides compounds that can be used as substitutes or to replace one or more undesired components, e.g., animal derived components. The replacement compounds provide at least one desired function of the undesired component.
  • Definitions
  • In the description that follows, a number of terms used in cell culture and recombinant DNA technology are utilized extensively. In order to provide a clear and more consistent understanding of the specification and claims, including the scope to be given such terms, the following definitions are provided.
  • The term “introduction” of a macromolecule or compound into culture refers to the provision of the macromolecule or compound into the culture medium.
  • The term “introduction” of a macromolecule or compound into at least one cell refers to the provision of a macromolecule or compound to a cell, such that the macromolecule or compound becomes internalized in the cell. For example, a macromolecule or compound can be introduced into a cell using transfection, transformation, injection, and/or liposomal introduction, and may also be introduced into a cell using other methods known to those of ordinary skill in the art. Preferably, a macromolecule or compound is introduced into a cell by liposomal introduction. The macromolecule is preferably a protein, peptide, polypeptide, or nucleic acid. The macromolecule may a protein. Alternatively, the macromolecule may be a peptide. Alternatively, the macromolecule may be a polypeptide. The macromolecule may also be a nucleic acid.
  • The term “macromolecule,” as used herein, encompasses biomolecules. In one embodiment, the term macromolecule refers to nucleic acid. In a preferred embodiment, the term macromolecule refers to deoxyribonucleic acid (DNA) and ribonucleic acid (RNA). More preferably, the term macromolecule refers to DNA. More preferably, the term macromolecule refers to complementary DNA (cDNA). A macromolecule can be charged or uncharged. A DNA molecule is an example of a charged macromolecule.
  • The term “nucleic acid” as used herein refers to any nucleic acid, including deoxyribonucleic acid (DNA) and ribronucleic acid (RNA). In preferred embodiments, “nucleic acid” refers to DNA, including genomic DNA, complementary DNA (cDNA), and oligonucleotides, including oligoDNA. More preferably, “nucleic acid’ refers to genomic DNA and/or cDNA.
  • The term “expression of nucleic acid,” as used herein, refers to the replication of the nucleic acid in a cell, to transcription of DNA to messenger RNA, to translation of RNA to protein, to post-translational modification of protein, and/or to protein trafficking in the cell, or variations or combinations thereof.
  • The term “ingredient” refers to any compound, whether of chemical or biological origin, that can be used in cell culture media to maintain or promote the growth or proliferation of cells. The terms “component,” “nutrient” and “ingredient” can be used interchangeably and are all meant to refer to such compounds. Typical ingredients that are used in cell culture media include amino acids, salts, metals, sugars, lipids, nucleic acids, hormones, vitamins, fatty acids, proteins and the like. Other ingredients that promote or maintain cultivation of cells ex vivo can be selected by those of skill in the art, in accordance with the particular need. Media of the present invention can include one or more components selected from the group consisting of bovine serum albumin (BSA) or human serum albumin (HSA), a one or more growth factors derived from natural (animal) or recombinant sources such as epidermal growth factor (EGF) or fibroblast growth factor (FGF), one or more lipids, such as fatty acids, sterols and phospholipids, one or more lipid derivatives and complexes, such as phosphoethanolamine, ethanolamine and lipoproteins, one or more proteins, one or more and teroid hormones, such as insulin, hydrocortisone and progesterone, one or more nucleotide precursors; and one or more trace elements.
  • The term “cell” as used herein refers includes all types of eukaryotic and prokaryotic cells. In preferred embodiments, the term refers to eukaryotic cells, especially mammalian cells.
  • By “cell culture” or “culture” is meant the maintenance of cells in an artificial, in vitro environment.
  • By “cultivation” is meant the maintenance of cells in vitro under conditions favoring growth and/or differentiation and/or or continued viability. “Cultivation” can be used interchangeably with “cell culture.” Cultivation is assessed by number of viable cells/ml culture medium. Cultivation after introduction of a macromolecule preferably includes production of a product, for example, a protein product on a virus.
  • The term “replenishing, replacing, or supplementing medium” as used herein refers to adding a volume of fresh cell culture medium to medium that was already present in culture and/or replacing medium that was already present in culture with fresh medium, and/or supplementing medium already present in culture with new medium. Fresh medium is medium that does not contain the one or more macromolecules or compounds to be introduced into at least one cell or medium that has not been in contact with cells to support their growth on cultivation. The skilled artisan can determine whether there is an advantage from or a need to remove and/or replenish, replace or supplement medium by monitoring cell growth and/or viability by techniques known in the art, such as cell counting (manual or automated), trypan blue exclusion, production of protein or other substance, alamar blue assay, presence or concentration of one or more metabolic products, cell adhesion, morphological appearance, analysis of spent medium, etc. One or a combination of monitoring techniques can be used to determine whether the medium needs to be to support growth, introduction of at least one macromolecule and/or cultivation after introduction of at least one macromolecule.
  • “Recombinant protein” refers to protein that is encoded by a nucleic acid that is introduced into a host cell. The host cell expresses the nucleic acid. The term “expressing a nucleic acid” is synonymous with “expressing a protein of an RNA encoded by a nucleic acid.” “Protein” as used herein broadly refers to polymerized amino acids, e.g., peptides, polypeptides, proteins, lipoproteins, glycoproteins, etc.
  • The term “protein yield” refers to the amount of protein expressed by cultured cells, and can be measured, for example, in terms of grams of protein produced/ml medium. If the protein is not secreted by the cells, the protein can be isolated from the interior of the cells by methods known to those of ordinary skill in the art. If the protein is secreted by the cells, the protein can be isolated from the culture medium by methods known to those of ordinary skill in the art. The amount of protein expressed by the cell can readily be determined by those of ordinary skill in the art. The protein may be a recombinant protein.
  • A “protein product” is a product associated with production or an action by a protein. A protein product may be a protein. A protein product may also be a product resulting from action of a protein by one or more other substances to produce a product. An example of such action is enzymatic action by a protein.
  • By “suspension culture” is meant cell culture in which the majority or all of cells in a culture vessel are present in suspension, and the minority or none of the cells in the culture vessel are attached to the vessel surface or to another surface within the vessel. Preferably, “suspension culture” has greater than 75% of the cells in the culture vessel are in suspension, not attached to a surface on or in the culture vessel. More preferably, a “suspension culture” has greater than 85% of the cells in the culture vessel are present in suspension, not attached to a surface on or in the culture vessel. Even more preferred is a “suspension culture” with greater than 95% of the cells in the culture vessel present in suspension, not attached to a surface on or in the culture vessel.
  • The medium, methods, kit and composition of the present invention are suitable for either monolayer or suspension culture, transfection, and cultivation of cells, and for expression of protein in cells in monolayer or suspension culture. Preferably, the medium, methods, kit and composition of the present invention are for suspension culture, transfection, and cultivation of cells, and for expression of protein product in cells in suspension culture.
  • By “culture vessel” is meant any container, for example, a glass, plastic, or metal container, that can provide an aseptic environment for culturing cells.
  • The phrases “cell culture medium,” “tissue culture medium,” “culture medium” (plural “media” in each case) and “medium formulation” refer to a nutritive solution for cultivating cells or tissues. These phrases can be used interchangeably.
  • The term “combining” refers to the mixing or admixing of ingredients.
  • Derivative of a molecule includes these compounds that comprise the base cyclic or heterocyclic molecule, but have additional or modified side groups. Preferably a “derivative” can be formed by reacting the base molecule with only 1, but possibly 2, 3, 4, 5, 6, etc. reactant molecules. A single step reaction is preferred, but multi-step, e.g., 2, 3, 4, 5, 6, etc. reactions are known in the art to form derivatives. Substitution, condensation and hydrolysis reactions are preferred and may be combined to form the derivative compound. Alternatively, a derivative compound may be a compound that preferably in 1, but possibly 2, 3, 4, 5, 6, etc. reactions can form the base cyclic or heterocyclic compound or a substitution or condensation product thereto.
  • A cell culture medium is composed of a number of ingredients and these ingredients can vary from medium to medium. Each ingredient used in a cell culture medium has its unique physical and chemical characteristics. Compatibility and stability of ingredients are determined in part by the “solubility” of the ingredients in aqueous solution. The terms “solubility” and “soluble” refer to the ability of an ingredient to form and remain in solution with other ingredients. Ingredients are thus compatible if they can be maintained in solution without forming a measurable or detectable precipitate.
  • By “compatible ingredients” is also meant those media components which can be maintained together in solution and form a “stable” combination. A solution containing “compatible ingredients” is said to be “stable” when the ingredients do not precipitate, degrade or decompose substantially such that the concentration of one or more of the components available to the cells from the media is reduced to a level that no longer supports the optimum or desired growth of the cells. Ingredients are also considered “stable” if degradation cannot be detected or when degradation occurs at a slower rate when compared to decomposition of the same ingredient in a 1× cell culture media formulation. For example, in 1× media formulations glutamine is known to degrade into pyrolidone carboxylic acid and ammonia. Glutamine in combination with divalent cations are considered “compatible ingredients” since little or no decomposition of the glutamine can be detected over time in solutions or combinations in which both glutamine and divalent cations are present. See U.S. Pat. No. 5,474,931. Thus, the term “compatible ingredients” as used herein refers to the combination of particular culture media ingredients which, when mixed in solution either as concentrated or 1× formulations, are “stable” and “soluble.”
  • The term “1× formulation” is meant to refer to any aqueous solution that contains some or all ingredients found in a cell culture medium at working concentrations. The “1× formulation” can refer to, for example, the cell culture medium or to any subgroup of ingredients for that medium. The concentration of an ingredient in a 1× solution is about the same as the concentration of that ingredient found in a cell culture formulation used for maintaining or cultivating cells in vitro. A cell culture medium used for the in vitro cultivation of cells is a 1× formulation by definition. When a number of ingredients are present, each ingredient in a 1× formulation has a concentration about equal to the concentration of each respective ingredient in a medium during cell culturing. For example, RPMI-1640 culture medium contains, among other ingredients, 0.2 g/L L-arginine, 0.05 g/L L-asparagine, and 0.02 g/L L-aspartic acid. A “1× formulation” of these amino acids contains about the same concentrations of these ingredients in solution. Thus, when referring to a “1× formulation,” it is intended that each ingredient in solution has the same or about the same concentration as that found in the cell culture medium being described. The concentrations of ingredients in a 1× formulation of cell culture medium are well known to those of ordinary skill in the art. See, for example, Methods For Preparation of Media, Supplements and Substrate For Serum-Free Animal Cell Culture Allen R. Liss, N.Y. (1984), Handbook of Microbiological Media, Second Ed., Ronald M. Atlas, ed. Lawrence C. Parks (1997) CRC Press, Boca Raton, Fla. and Plant Culture Media, Vol. 1: Formulations and Uses E. F. George, D. J. M. Puttock, and H. J. George (1987) Exegetics Ltd. Edington, Westbury, Wilts, BA13 4QG England each of which is incorporated by reference herein in its entirety. The osmolarity and/or pH, however, can differ in a 1× formulation compared to the culture medium, particularly when fewer ingredients are contained in the 1× formulation.
  • A “10× formulation” is meant to refer to a solution wherein the concentration of each ingredient in that solution is about 10 times more than the concentration of each respective ingredient in a medium during cell culturing. For example, a 10× formulation of RPMI-1640 culture medium can contain, among other ingredients, 2.0 g/L L-arginine, 0.5 g/L L-asparagine, and 0.2 g/L L-aspartic acid (compare 1× formulation, above). A “10× formulation” can contain a number of additional ingredients at a concentration about 10 times that found in the 1× culture formulation. As will be readily apparent, “25× formulation,” “50× formulation,” “100× formulation,” “500× formulation,” and “1000× formulation” designate solutions that contain ingredients at about 25-, 50-, 100-, 500-, or 1000-fold concentrations, respectively, as compared to a 1× cell culture formulation. Again, the osmolarity and pH of the medium formulation and concentrated solution can vary.
  • The term “trace element” or “trace element moiety” refers to a moiety which is present in a cell culture medium in only very low (i.e., “trace”) amounts or concentrations, relative to the amounts or concentrations of other moieties or components present in the culture medium. In the present invention, these terms encompass Ag+, Al3+, Ba2+, Cd2, Co2+, Cr3+, Cu1+, Cu2+, Fe2+, Fe3+, Ge4+, Se4+, Br, I, Mn2+, F, Si4+, V5+, Mo6+, Ni2+, Rb+, Sn2+ and Zr4+ and salts thereof. For example, the following salts can be used as trace elements in the culture media of the invention: AgNO3, AlCl3.6H2O, Ba(C2H3O2)2, CdSO4.8H2O, CoCl2.6H2O, Cr2(SO4)3.1H2O, GeO2, Na2SeO3, H2SeO3, KBr, KI, MnCl2.4H2O, NaF, Na2SiO3.9H2O, NaVO3, (NH4)6Mo7O24.4H2O, NiSO4.6H2O, RbCl, SnCl2, and ZrOCl2.8H2O. Suitable concentrations of trace element moieties can be determined by one of ordinary skill in the art using only routine experimentation.
  • The term “amino acid” refers to amino acids or their derivatives (e.g., amino acid analogs), as well as their D- and L-forms. Examples of such amino acids include glycine, L-alanine, L-asparagine, L-cysteine, L-aspartic acid, L-glutamic acid, L-phenylalanine, L-histidine, L-isoleucine, L-lysine, L-leucine, L-glutamine, L-arginine, L-methionine, L-proline, L-hydroxyproline, L-serine, L-threonine, L-tryptophan, L-tyrosine, and L-valine, N-acetyl cysteine.
  • A “serum-free medium” is a medium that contains no serum (e.g., fetal bovine serum (FBS), calf serum, horse serum, goat serum, human serum, etc.) and is generally designated by the letters SFM.
  • The terms “serum-free culture conditions” and “serum-free conditions” refer to cell culture conditions that exclude serum of any type. These terms can be used interchangeably.
  • A “chemically defined” medium is one in which each chemical species and its respective quantity is known prior to its use in culturing cells. A chemically defined medium is made without lysates or hydrolysates whose chemical species are not known and/or quantified. A chemically defined medium is one preferred embodiment of the medium of the present invention.
  • The phrase “protein-free” culture media refers to culture media that contain no protein (e.g., no serum proteins such as serum albumin or attachment factors, nutritive proteins such as growth factors, or metal ion carrier proteins such as transferrin, ceruloplasmin, etc.). Preferably, if peptides are present, the peptides are smaller peptides, e.g., di- or tri-peptides. Preferably, peptides of deca-peptide length or greater are less than about 1%, more preferably less than about 0.1%, and even more preferably less than about 0.01% of the amino acids present in the protein free medium.
  • The phrase “low-protein” culture media as used herein refers to media that contain only low amounts of protein (typically less than about 10%, less than about 5%, less than about 1%, less than about 0.5%, or less than about 0.1%, of the amount or concentration of total protein found in culture media containing standard amounts of protein, such as standard basal medium supplemented with 5-10% serum).
  • The term “animal derived” material as used herein refers to material that is derived in whole or in part from an animal source, including recombinant animal DNA or recombinant animal protein DNA. Preferred media contain no animal desired material.
  • By “transition element” or “transition metal” (which can be used interchangeably) is meant an element in which an inner electron valence shell, rather than an outer shell, is only partially filled, such that the element acts as a transitional link between the most and least electropositive in a given series of elements. Transition elements are typically characterized by high melting points, high densities, high dipole or magnetic moments, multiple valencies, and the ability to form stable complex ions. Examples of such transition elements useful in the present invention include scandium (Sc), titanium (Ti), vanadium (V), chromium (Cr), manganese (Mn), iron (Fe), cobalt (Co), nickel (Ni), copper (Cu), zinc (Zn), yttrium (Y), zirconium (Zr), niobium (Nb), molybdenum (Mo), technetium (Tc), rubidium (Ru), rhodium (Rh), palladium (Pd), silver (Ag), cadmium (Cd), lanthanum (La), hafnium (Hf), tantalum (Ta), tungsten (W), rhenium (Re), osmium (Os), iridium (Ir), platinum (Pt), gold (Au), mercury (Hg), and actinium (Ac). Of particular interest as a transition element for use in culture media compositions, including those of the present invention, are ions, chelates, salts, and complexes of iron (Fe++ or Fe+++).
  • A variety of techniques and reagents are available for the introduction of macromolecules into a target cell. Commonly used reagents include, for example, calcium phosphate, DEAE-dextran and lipids. For examples of detailed protocols for the use of reagents of these types, numerous references texts are available for example, Current Protocols in Molecular Biology, Chapter 9, Ausubel, et al. Eds., John Wiley and Sons, 1998.
  • Lipid aggregates such as liposomes have been found to be useful as agents for the delivery of macromolecules into cells. In particular, lipid aggregates comprising one or more cationic lipids have been demonstrated to be extremely efficient at the delivery of anionic macromolecules (for example, nucleic acids) into cells. One commonly used cationic lipid is N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium chloride (DOTMA). Liposomes comprising DOTMA alone or as a 1:1 mixture with dioleoylphosphatidylethanolamine (DOPE) have been used to introduce nucleic acids into cells. A 1:1 mixture of DOTMA:DOPE is commercially available from Invitrogen Corporation, Carlsbad, Calif. under the trade name of LIPOFECTIN®. Another cationic lipid that has been used to introduce nucleic acids into cells is 1,2-bis(oleoyl-oxy)-3-3-(trimethylammonia) propane (DOTAP). DOTAP differs from DOTMA in that the oleoyl moieties are linked to the propylamine backbone via ether bonds in DOTAP whereas they are linked via ester bonds in DOTMA. DOTAP is believed to be more readily degraded by the target cells. A structurally related group of compounds wherein one of the methyl groups of the trimethylammonium moiety is replaced with a hydroxyethyl group are similar in structure to the Rosenthal inhibitor (RI) of phospholipase A (see Rosenthal, et al., (1960) J. Biol. Chem. 233:2202-2206.). The RI has stearoyl esters linked to the propylamine core. The dioleoyl analogs of RI are commonly abbreviated DOR1-ether and DOR1-ester, depending upon the linkage of the lipid moiety to the propylamine core. The hydroxyl group of the hydroxyethyl moiety can be further derivatized, for example, by esterification to carboxyspermine.
  • Another class of compounds which has been used for the introduction of macromolecules into cells comprise a carboxyspermine moiety attached to a lipid (see, Behr, et al., (1989) Proceedings of the National Academy of Sciences, USA 86:6982-6986 and EPO 0 394 111). Examples of compounds of this type include dipalmitoylphosphatidylethanolamine 5-carboxyspermylamide (DPPES) and 5-carboxyspermylglycine dioctadecylamide (DOGS). DOGS is commercially available from Promega, Madison, Wis. under the trade name of TRANSFECTAM™.
  • A cationic derivative of cholesterol (3β-[N—(N′,N′-dimethylaminoethane)-carbamoyl] cholesterol, DC-Chol) has been synthesized and formulated into liposomes with DOPE (see Gao, et al., (1991) BBRC 179(1):280-285.) and used to introduce DNA into cells. The liposomes thus formulated were reported to efficiently introduce DNA into the cells with a low level of cellular toxicity. Lipopolylysine, formed by conjugating polylysine to DOPE (see Zhou, et al., (1991) BBA 1065:8-14), has been reported to be effective at introducing nucleic acids into cells in the presence of serum.
  • Other types of cationic lipids that have been used to introduce nucleic acids into cells include highly packed polycationic ammonium, sulfonium and phosphonium lipids such as those described in U.S. Pat. Nos. 5,674,908 and 5,834,439, and international application no. PCT/US99/26825, published as WO 00/27795. One preferred agent for delivery of macromolecules is LIPOFECTAMINE 2000™ which is available from Invitrogen. See U.S. international application no. PCT/US99/26825, published as WO 00/27795.
  • A “reagent for the introduction of macromolecules” into cells is any material known to those of skill in the art which facilitates the entry of a macromolecule into a cell. For example, see U.S. Pat. No. 5,279,833. In some embodiments, the reagent can be a “transfection reagent” and can be any compound and/or composition that increases the uptake of one or more nucleic acids into one or more target cells. A variety of transfection reagents are known to those skilled in the art. Suitable transfection reagents can include, but are not limited to, one or more compounds and/or compositions comprising cationic polymers such as polyethyleneimine (PEI), polymers of positively charged amino acids such as polylysine and polyarginine, positively charged dendrimers and fractured dendrimers, cationic β-cyclodextrin containing polymers (CD-polymers), DEAE-dextran and the like. In some embodiments, a reagent for the introduction of macromolecules into cells can comprise one or more lipids which can be cationic lipids and/or neutral lipids. Preferred lipids include, but are not limited to, N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethylamonium chloride (DOTMA), dioleoylphosphatidylcholine (DOPE),1,2-Bis(oleoyloxy)-3-(4′-trimethylammonio) propane (DOTAP), 1,2-dioleoyl-3-(4′-trimethylammonio) butanoyl-sn-glycerol (DOTB), 1,2-dioleoyl-3-succinyl-sn-glycerol choline ester (DOSC), cholesteryl (4′-trimethylammonio)butanoate (ChoTB), cetyltrimethylammonium bromide (CTAB), 1,2-dioleoyl-3-dimethyl-hydroxyethyl ammonium bromide (DORI), 1,2-dioleyloxypropyl-3-dimethyl-hydroxyethyl ammonium bromide (DORIE), 1,2-dimyristyloxypropyl-3-dimethyl-hydroxyethyl ammonium bromide (DMRIE), O,O′-didodecyl-N-[p(2-trimethylammonioethyloxy)benzoyl]-N,N,N-trimethylammonium chloride, spermine conjugated to one or more lipids (for example, 5-carboxyspermylglycine dioctadecylamide (DOGS), N,NI,NII,NIII-tetramethyl-N,NI,NII,NIII-tetrapalmitylspermine (TM-TPS) and dipalmitoylphasphatidylethanolamine 5-carboxyspermylaminde (DPPES)), lipopolylysine (polylysine conjugated to DOPE), TRIS (Tris(hydroxymethyl)aminomethane, tromethamine) conjugated fatty acids (TFAs) and/or peptides such as trilysyl-alanyl-TRIS mono-, di-, and tri-palmitate, (3β-[N—(N′,N′-dimethylaminoethane)-carbamoyl] cholesterol (DC-Chol), N-(α-trimethylammonioacetyl)-didodecyl-D-glutamate chloride (TMAG), dimethyl dioctadecylammonium bromide (DDAB), 2,3-dioleyloxy-N-[2(spermine-carboxamido)ethyl]-N,N-dimethyl-1-propanamininiumtrifluoroacetate (DOSPA) and combinations thereof.
  • Those skilled in the art will appreciate that certain combinations of the above mentioned lipids have been shown to be particularly suited for the introduction of nucleic acids into cells for example a 3:1 (w/w) combination of DOSPA and DOPE is available from Invitrogen Corporation, Carlsbad, Calif. under the trade name LIPOFECTAMINE™, a 1:1 (w/w) combination of DOTMA and DOPE is available from Invitrogen Corporation, Carlsbad, Calif. under the trade name LIPOFECTIN®, a 1:1 (M/M) combination of DMRIE and cholesterol is available from Invitrogen Corporation, Carlsbad, Calif. under the trade name DMRIE-C reagent, a 1:1.5 (M/M) combination of TM-TPS and DOPE is available from Invitrogen Corporation, Carlsbad, Calif. under the trade name CellFECTIN® and a 1:2.5 (w/w) combination of DDAB and DOPE is available from Invitrogen Corporation, Carlsbad, Calif. under the trade name LipfectACE®. In addition to the above-mentioned lipid combinations, other formulations comprising lipids in admixture with other compounds, in particular, in admixture with peptides and proteins comprising nuclear localization sequences, are known to those skilled in the art. For example, see international application no. PCT/US99/26825, published as WO 00/27795.
  • The present invention is directed to a culture medium that supports (a) the introduction of at least one macromolecule into eukaryotic cells in culture, (b) the cultivation of cells into which at least one macromolecule is introduced, and optionally (c) the production of protein product or expression of the nucleic acid in cells into which at least one macromolecule is introduced, wherein medium containing the macromolecule does not need to be removed from the culture and replaced with fresh medium after introduction of at least one macromolecule into cells and prior to cultivation and production of protein product or expression of nucleic acid. Using the medium of the present invention, it is not necessary to replenish, replace or supplement the medium after one has introduced at least on emacromolecule into at least one cell, and before cells into which at least one macromolecule has been introduced are further cultured to produces protein product or express a nucleic acid. In the medium, methods, kit and composition of the present invention, the medium is a serum-free medium and/or a chemically defined medium and/or protein free or low protein medium and/or a medium that does not contain animal derived components, or a medium having combinations of these features.
  • CD CHO medium (Invitrogen Corporation, Carlsbad, Calif.) can be used to transfect CHO cells in suspension culture. However, transfection of CHO cells in CD CHO medium works only if the cells are transfected with nucleic acid in a relatively much smaller volume of CD CHO medium than the transfected cells are to be cultured in after transfection. Thus, for cells to be cultured in CD CHO medium after they are transfected with nucleic acid, it is necessary to transfect cells in a relatively small volume, and then dilute the transfected cells in a relatively much larger volume of CD CHO medium for subsequent culturing.
  • In a preferred embodiment, with respect to the introduction of compounds or macromolecules (e.g., nucleic acid) into cells in culture, the culture medium of the present invention facilitates higher cell transformation efficiency than does CD CHO medium, and/or does not require transfecting in a smaller volume than cells are to be cultured in after transfection, and/or facilitates higher cell viability than does CD CHO medium, and/or facilitates higher cell density (i.e., cells/ml of culture medium) than does CD CHO medium, and/or facilitates a higher level of recombinant protein expression in cells in culture than does CD CHO medium. Preferably, the same volume of medium is used for introduction of at least one macromolecule or transfection and subsequent cultivation. Alternatively, the cells are divided or medium volume is increased less from about 2, about 5, about 8 or about 10 times. In preferred embodiments, the culture medium of the present invention is not CD CHO medium.
  • The medium, methods, kit and composition of the present invention are intended to be used to introduce at least one macromolecule or to transfect and culture cells in any volume of culture medium. Such introduction is preferably accomplished in 0.1 to 10 times the amount of medium used to culture cells to be transfected. Preferably, the cell culture volume is greater than about one milliliter. More preferably, the cell culture volume is from about one milliliter to 250 liters. More preferably, the cell culture volume is from about 30 ml to about 50 liters. More preferably, the cell culture volume is from about 100 ml to about 50 liters. More preferably, the cell culture volume is from about 500 ml to about 50 liters. More preferably, the cell culture volume is from about 500 ml to about 25 liters. More preferably, the cell culture volume is from about 500 ml to about 10 liters. More preferably, the cell culture volume is from about 500 ml to about 5 liters. More preferably, the cell culture volume is from about 500 ml to about 1 liter.
  • In the medium, methods, kit and composition of the present invention, the medium preferably does not contain compounds that can interfere with introduction of macromolecules or transfection, e.g., polyanionic compounds such as polysulfonated and/or polysulfated compounds. Preferably, the medium does not contain dextran sulfate.
  • The medium, methods, kit and composition of the present invention permit the introduction of compounds or macromolecules (particularly macromolecules, for example nucleic acids, proteins and peptides) into the cultured cells (for example by transfection) without the need to change the medium. In one preferred embodiment, the present invention provides a medium for the cultivation and transfection of eukaryotic cells.
  • Using the medium, methods, kit and composition of the present invention, those of ordinary skill in the art can introduce macromolecules or compounds (e.g., nucleic acid) into cells in culture. Preferably, the macromolecule or compound (e.g., nucleic acid) is introduced into at least about 20 percent of the cells. More preferably, the macromolecule or compound (e.g., nucleic acid) is introduced into about 20 to about 100 percent of the cells. More preferably, the macromolecule or compound (e.g., nucleic acid) is introduced into about 30 to about 100 percent of the cells. More preferably, the macromolecule or compound (e.g., nucleic acid) is introduced into about 50 to about 100 percent of the cells. Practically, the macromolecule or compound might be introduced into about 20% to about 90% of the cells, about 20% to about 80% of the cells, about 30% to about 60, 70, 80 or 90% of the cells, about 20, 30, 40 or 50% to about 70, 75, 80, 85, 90, 95 or 98% of the cells, etc. Even about 60, 70, 75 or 80 to about 90% or close to 100% of the cells may contain the introduced molecule or compound.
  • In preferred embodiments of the medium, methods, kit and composition of the present invention, one or more undesirable components (i.e., one or more serum components, one or more undefined components, one or more protein components and/or one or more animal derived components) have been substituted or replaced in one or more functions by one or more replacement compounds. Replacement compounds of the invention can be metal binding compounds and/or one or more transition element complexes, said complexes comprising one or more transition elements or a salts or ions thereof, in a complex with one or more metal-binding compounds. Preferably, the medium is capable of supporting the cultivation of a cell in vitro in the absence of one or more naturally derived metal carriers, such as transferrin, or other animal derived proteins or extracts. The metal binding compound can be in a complex with a transition metal prior to addition of the metal binding compound to the medium. In other embodiments, the metal binding compound is not in a complex with a transition metal prior to addition of the metal binding compound to the media. Preferably, the medium of the present invention does not contain transferrin and/or does not contain insulin.
  • The present invention also relates to a cell culture medium obtained by combining a medium with one or more replacement compounds. Preferably, the medium can be a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or can be a medium lacking animal derived components. The medium preferably does not contain transferrin and/or does not contain insulin. In some preferred embodiments, the medium can be capable of supporting the cultivation of a cell in vitro and/or can permit the introduction of macromolecules into the cell. In some embodiments, one or more of the replacement compounds can be a metal binding compound and/or can be a transition element complex, said complex comprising at least one transition element or a salt or ion thereof complexed to at least one metal-binding compound. Preferred transition elements, metal-binding compounds, and transition element complexes for use in this aspect of the invention include those described in detail herein.
  • Replacement compounds of the present invention can facilitate the delivery of transition metals to cells cultured in vitro. In preferred embodiments, the replacement compounds can deliver iron and replace transferrin. A preferred replacement compound is a hydroxypyridine derivative. Preferably, the hydroxypyridine derivative is selected from the group consisting of 2-hydroxypyridine-N-oxide, 3-hydroxy-4-pyrone, 3-hydroxypypyrid-2-one, 3-hydroxypyrid-2-one, 3-hydroxypyrid-4-one, 1-hydroxypyrid-2-one, 1,2-dimethyl-3-hydroxypyrid-4-one, 1-methyl-3-hydroxypyrid-2-one, 3-hydroxy-2(1H)-pyridinone, and pyridoxal isonicotinyl hydrazone, nicotinic acid-N-oxide, 2-hydroxy-nicotinic acid. Most preferably, the hydroxypyridine derivative is 2-hydroxypyridine-N-oxide.
  • The replacement compounds of the present invention can be used with any media, including media for cultivating or growing eukaryotic and/or prokaryotic cells, tissues, organs, etc. Such media include, but are not limited to, Dulbecco's Modified Eagle's Medium (DMEM), Minimal Essential Medium (MEM), Basal Medium Eagle (BME), RPMI-1640, Ham's F-10, Ham's F-12, αMinimal Essential Medium (αMEM), Glasgow's Minimal Essential Medium (G-MEM), and Iscove's Modified Dulbecco's Medium (IMDM). Other media that are commercially available (e.g., from Invitrogen Corporation, Carlsbad, Calif.) or that are otherwise known in the art can be equivalently used in accordance with the present invention including, but not limited to, 293 SFM, CD-CHO medium, VP SFM, BGJb medium, Brinster's BMOC-3 medium, cell culture freezing medium, CMRL media, EHAA medium, eRDF medium, Fischer's medium, Gamborg's B-5 medium, GLUTAMAX™ supplemented media, Grace's insect cell media, HEPES buffered media, Richter's modified MEM, IPL-41 insect cell medium, Leibovitz's L-15 media, McCoy's 5A media, MCDB 131 medium, Media 199, Modified Eagle's Medium (MEM), Medium NCTC-109, Schneider's Drosophila medium, TC-100 insect medium, Waymouth's MB 752/1 media, William's Media E, protein free hybridoma medium II (PFHM II), AIM V media, Keratinocyte SFM, defined Keratinocyte SFM, STEMPRO® SFM, STEMPRO® complete methylcellulose medium, HepatoZYME-SFM, Neurobasal™ medium, Neurobasal-A medium, Hibernate™ A medium, Hibernate E medium, Endothelial SFM, Human Endothelial SFM, Hybridoma SFM, PFHM II, Sf 900 medium, Sf 900 TI SFM, EXPRESS FIVE® medium, CHO-S-SFM, AMINOMAX-II complete medium, AMINOMAX-C100 complete medium, AMINOMAX-C 100 basal medium, PB-MAX™ karyotyping medium, KARYOMAX bone marrow karyotyping medium, KNOCKOUT D-MEM and CO2 independent medium. The above media are obtained from manufacturers known to those of ordinary skill in the art, such as JRH, Sigma, HyClone, and BioWhittaker. Additional examples of media suitable for use in the practice of the present invention can be found in U.S. Pat. Nos. 5,135,866 and 5,232,848 as well as in international publications nos. WO 88/02774, WO 98/15614, WO 98/08934 and European patent no. 0 282 942, the entireties of which are specifically incorporated herein by reference.
  • The present invention also provides a method for introducing macromolecules into cells, comprising culturing cells in a medium of the invention and contacting the cells in the medium with one or more macromolecules under conditions causing the macromolecules to be taken up by one or more of the cells. Preferably, the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or can be a medium lacking animal derived components. Preferred cells include eukaryotic cells. More preferably, the cells are mammalian cells. The medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin. In some preferred embodiments, the medium permits the growth and transfection of the cell in the same medium. In some embodiments, the macromolecules can comprise one or more nucleic acids and conditions causing the nucleic acid molecules to be taken up by the cells include contacting the nucleic acid with a reagent which causes the nucleic acid to be introduced into one or more cells.
  • The present invention also provides a composition comprising a medium of the invention and a cell. Preferably, the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or a medium lacking animal derived components. Preferred cells include eukaryotic cells. More preferably, the cells are mammalian cells. The medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin. Preferably, the medium supports the growth and transfection of the cell in the same medium.
  • The present invention also provides compositions comprising a medium of the present invention and one or more reagents for the introduction of macromolecules into one or more cells. Preferably, the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or a medium lacking animal derived components. The medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin. Preferably, the medium contains a transfection reagent and the macromolecules are nucleic acids. The macromolecules might also be proteins and/or peptides. In some embodiments, the reagent comprises one or more lipids of which one or more can be cationic lipids. More preferably, the reagent comprises a mixture of neutral and cationic lipids. In some embodiments, the reagent comprises one or more peptides and/or proteins which can be provided alone or in admixture with one or more lipids.
  • The present invention also provides compositions comprising a medium of the invention and one or more macromolecules to be introduced into a cell. Preferably, the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or a medium lacking animal derived components. The medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin. The macromolecules can be, for example, nucleic acids and/or proteins and/or peptides and can be uncomplexed or can be in the form of a complex with one or more reagents for the introduction of macromolecules into cells. Preferably, the macromolecules are nucleic acids and can be in the form of a complex with one or more transfection reagents.
  • The present invention also provides a composition comprising at least one component (or combination thereof) selected from the group consisting of a medium of the present invention, at least one cell, at least one macromolecule, at least one reagene for introducing at least one macromoleule into at least one cell. Preferably, the cells are eukaryotic cells. More preferably, the cells are mammalian cells. Preferably, the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or a medium lacking animal derived components. The medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin. In some preferred embodiments, the reagent is a transfection reagent and the macromolecules are nucleic acids, for example RNA and/or DNA. Alternatively, the macromolecules are proteins and/or peptides.
  • In some embodiments, the reagent comprises one or more lipids of which one or more can be cationic lipids. More preferably, the reagent comprises a mixture of neutral and cationic lipids. In some embodiments, the reagent comprises one or more peptides and/or proteins which can be provided alone or in admixture with one or more lipids. In preferred embodiments, the reagent complexes with the macromolecule to introduce the macromolecule into the cell.
  • The present invention also provides kits for the culture and transfection of cells comprising at least one container comprising a medium for the culture and transfection of cells. Such kits may also comprise at least one component (or a combination thereof) selected from the group consisting of a medium of the present invention, at least one cell, at least one macromolecule, at least one reagent for introducing at least one macromolecule into at least one cell, at least one buffer or buffering salt, and instructions for using the kit to introduce at least one macromolecule into at least one cell. Preferably, the medium is a serum-free medium and/or a chemically defined medium and/or a protein-free or low protein medium and/or a medium lacking animal derived components. The medium can comprise one or more replacement compounds and preferably does not contain transferrin and/or does not contain insulin and/or does not contain an animal growth factor. The medium can comprise one or more replacement compounds which can be metal binding compounds and/or can comprise one or more complexes comprising one or more replacement compounds. In some embodiments, the medium can comprise one or more complexes, said complex comprising one or more transition elements or salts or ions thereof complexed one or more replacement compounds which can be metal-binding compounds. In some embodiments, said medium is capable of supporting the cultivation of a cell in vitro and permits transfection of cells cultured therein. In some embodiments, kits of the invention can further comprise at least one container comprising a lipid for transfecting cells. In some embodiments, the kits of the invention can comprise at least one container comprising a nucleic acid.
  • According to one aspect of the invention, a transition element is preferably selected from the group consisting of scandium, titanium, vanadium, chromium, manganese, iron, cobalt, nickel, copper, zinc, yttrium, zirconium, niobium, molybdenum, technetium, rubidium, rhodium, palladium, silver, cadmium, lanthanum, hafnium, tantalum, tungsten, rhenium, osmium, iridium, platinum, gold, mercury, and actinium, or salts or ions thereof, and is preferably an iron salt. Suitable iron salts include, but are not limited to, FeCl3, Fe(NO3)3 or FeSO4 or other compounds that contain Fe+++ or Fe++ ions.
  • Preferred replacement compounds include, but are not limited to, metal-binding compounds. See, for example, international patent application no. PCT/US00/23580, publication no. WO 01/16294.
  • Metal binding compounds of the present invention include any macromolecules which can interact with or bind with transition elements and facilitate their uptake by cells. Such interaction/binding can be covalent or non-covalent in nature. The metal-binding compound used in this aspect of the invention is preferably selected from the group consisting of a polyol, a hydroxypyridine derivative, 1,3,5-N,N′,N″-tris(2,3-dihydroxybenzoyl)amino-methylbenzene, ethylenediamine-N,N′-tetramethylenephosphonic acid, trisuccin, an acidic saccharide (e.g., ferrous gluconate), a glycosaminoglycan, diethylenetriaminepentaacetic acid, nicotinic acid-N-oxide, 2-hydroxy-nicotinic acid, mono-, bis-, or tris-substituted 2,2′-bipyridine, a hydroxamate derivative (e.g. acetohydroxamic acid), an amino acid derivative, deferoxamine, ferrioxamine, iron basic porphine and derivatives thereof, DOTA-lysine, a texaphyrin, a sapphyrin, a polyaminocarboxylic acid, an α-hydroxycarboxylic acid, a polyethylenecarbamate, ethyl maltol, 3-hydroxy-2-pyridine, and IRC011. In one preferred embodiment, the metal-binding compound is a polyol such as sorbitol or dextran, and particularly sorbitol. In a related embodiment, the metal-binding compound is a hydroxypyridine derivative, such as 2-hydroxypyridine-N-oxide, 3-hydroxy-4-pyrone, 3-hydroxypypyrid-2-one, 3-hydroxypyrid-2-one, 3-hydroxypyrid-4-one, 1-hydroxypyrid-2-one, 1,2-dimethyl-3-hydroxypyrid-4-one, 1-methyl-3-hydroxypyrid-2-one, 3-hydroxy-2(1H)-pyridinone, ethyl maltol or pyridoxal isonicotinyl hydrazone, and is preferably 2-hydroxypyridine-N-oxide. In particularly preferred embodiments according to this aspect of the invention, the transition metal complex can be a sorbitol-iron complex or 2-hydroxypyridine-N-oxide-iron complex. The metal binding compounds of the present invention can also bind divalent cations such as Ca++ and Mg++.
  • The invention relates to cell culture media comprising one or more replacement compounds which can be metal-binding compounds and further comprising one or more ingredients selected from the group of ingredients consisting of at least one amino acid (such as L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid, L-glutamine, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine or L-valine, N-acetyl-cysteine), at least one vitamin (such as biotin, choline chloride, D-Ca++-pantothenate, folic acid, i-inositol, niacinamide, pyridoxine, riboflavin, thiamine or vitamin B12), at least one inorganic salt (such as a calcium salt, CuSO4, FeSO4, Fe(NO3)3, FeCl3, KCl, a magnesium salt, a manganese salt, sodium acetate, NaCl, NaHCO3, Na2HPO4, Na2SO4, a selenium salt, a silicon salt, a molybdenum salt, a vanadium salt, a nickel salt, a tin salt, ZnCl2, ZnSO4 or other zinc salts), adenine, ethanolamine, D-glucose, one or more cytokines, heparin, hydrocortisone, lipoic acid, phenol red, phosphoethanolamine, putrescine, sodium pyruvate, tri-iodothyronine, PLURONIC F68, and thymidine.
  • The culture media of the present invention can optionally include one or more buffering agents. Suitable buffering agents include, but are not limited to, N-[2-hydroxyethyl]-piperazine-N′-[2-ethanesulfonic acid] (HEPES), MOPS, MES, phosphate, bicarbonate and other buffering agents suitable for use in cell culture applications. A suitable buffering agent is one that provides buffering capacity without substantial cytotoxicity to the cells cultured. The selection of suitable buffering agents is within the ambit of ordinary skill in the art of cell culture.
  • According to the invention, a medium suitable for use in forming the cell culture media of the invention can comprise one or more ingredients, and can be obtained, for example, by combining one or more ingredients selected from the group consisting of adenine, ethanolamine, D-glucose, heparin, a buffering agent, hydrocortisone, lipoic acid, phenol red, phosphoethanolamine, putrescine, sodium pyruvate, tri-iodothyronine, thymidine, L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid, L-glutamine, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, L-valine, N-acetyl-cysteine, biotin, choline chloride, D-Ca++-pantothenate, folic acid, i-inositol, niacinamide, pyridoxine, riboflavin, thiamine, vitamin B12, Pluronic F68, recombinant insulin, a calcium salt, CuSO4, FeSO4, FeCl3, Fe(NO3)3, KCl, a magnesium salt, a manganese salt, sodium acetate, NaCl, NaHCO3, Na2HPO4, Na2SO4, a selenium salt, a silicon salt, a molybdenum salt, a vanadium salt, a nickel salt, a tin salt, ZnCl2, ZnSO4 or other zinc salts, wherein each ingredient is added in an amount which supports the cultivation of a cell in vitro.
  • The invention is also directed to a cell culture medium comprising ingredients selected from ethanolamine, D-glucose, HEPES, insulin, linoleic acid, lipoic acid, phenol red, PLURONIC F68, putrescine, sodium pyruvate, transferrin, L-alanine, L-arginine, L-asparagine, L-aspartic acid, L-cysteine, L-glutamic acid, L-glutamine, glycine, L-histidine, L-isoleucine, L-leucine, L-lysine, L-methionine, L-phenylalanine, L-proline, L-serine, L-threonine, L-tryptophan, L-tyrosine, L-valine, biotin, choline chloride, D-Ca++-pantothenate, folic acid, i-inositol, niacinamide, pyridoxine, riboflavin, thiamine, vitamin B12, one or more calcium salts, Fe(NO3)3, KCl, one or more magnesium salts, one or more manganese salts, NaCl, NaHCO3, Na2HPO4, one or more selenium salts, one or more vanadium salts and one or more zinc salts, wherein each ingredient is present in an amount which supports the suspension cultivation of a mammalian epithelial cell in vitro. The invention is also directed to such media which can optionally further comprise one or more supplements selected from the group consisting of one or more cytokines, heparin, one or more animal peptides, one or more yeast peptides and one or more plant peptides (most preferably one or more of rice, aloevera, soy, maize, wheat, pea, squash, spinach, carrot, potato, sweet potato, tapioca, avocado, barley, coconut and/or green bean, and/or one or more other plants), e.g., see international application no. PCT/US97/18255, published as WO 98/15614.
  • The above ingredients listed in the following Tables 1-3, when admixed together in solution, form a complete culture medium of the present invention. These complete media are suitable for use in the culture of a variety of mammalian cells, as described in more detail herein. Based on the information obtained in Tables 1-3, and knowledge possessed by those of ordinary skill in the art, one of ordinary skill in the art can obtain operative media formulations without undue experimentation.
    TABLE 1
    Preferred Ranges of Ingredients
    Gram(s)/L Gram(s)/L
    (unless noted) (unless noted)
    Component about: Component about:
    L-Arginine HCl 0.05-2.50 Ammonium Meta 0.0000001-0.000003
    Vanadate
    L-Asparagine 0.005-0.05  Manganous Chloride 0.00000001-0.0000005
    4H2O
    L-Aspartic Acid 0.03-0.8  Pyridoxine HCl 0.0002-0.005
    L-Cysteine HCl H2O 0.03-0.8  Thiamine HCl 0.0002-0.006
    L-Glutamic Acid 0.002-0.05  Ferric Nitrate 9H2O 0.0002-0.004
    L-Histidine HCl H2O 0.01-0.3  Magnesium Sulfate 0.005-0.1 
    L-Isoleucine 0.04-1   Zinc Sulfate 0.00008-0.002 
    L-Leucine 0.06-1.5  Zinc Chloride 0.00005-0.001 
    L-Lysine HCl 0.05-1.25 Ascorbic Acid 0.00000005-0.0000003
    L-Methionine 0.02-0.6  D-Calcium Pantothenate 0.0002-0.006
    L-Phenylalanine 0.02-0.5  Calcium Chloride 0.002-0.06
    L-Serine 0.07-2   Magnesium Chloride 0.02-0.4
    L-Threonine 0.02-0.6  Potassium Chloride 0.06-1.2
    L-Tryptophan 0.008-0.2   Sodium Selenite 0.0000000001-0.00000003 
    L-Valine .03-1   Vitamin B12 0.00002-0.005 
    L-Tyrosine 2Na 2 H2O 0.02-0.5  Choline Chloride 0.003-0.07
    L-Glutamine 0.1-3   i-Inositol 0.004-0.09
    (recommended
    addition, formulation
    does not contain)
    D-Glucose (Dextrose)  1-25 Niacinamide 0.0002-0.006
    Lipoic Acid 0.0004-0.01  Ethanolamine HCl 0.001-0.3 
    Linoleic Acid 0.00001-0.0003  Putrescine 2HCl 0.00003-0.008 
    Folic Acid 0.0001-003   Sodium Pyruvate 0.02-0.6
    HEPES 0.5-15  Riboflavin 0.00004-0.001 
    2-Hydroxypyridine-n- 0.0006-0.02  Biotin 0.00002-0.0005
    oxide
    Pluronic F-68* 0.06-2   Sodium Bicarbonate 0.5-2 
    Sodium Phosphate 0.03-0.6  Sodium Chloride***    0.6-16***
    Monobasic H2O

    ***Added to adjust osmolarity. Quantity will vary somewhat between formulations. NaCL is added to adjust osmo from ˜160 to a value within 275 +/− 5 mOsmo
  • TABLE 2
    Exemplary Medium
    Gram(s)/L Gram(s)/L
    (unless noted) (unless noted)
    Component about: Component about:
    L-Arginine HCl 0.4 Ammonium Meta 0.0000006
    Vanadate
    L-Asparagine 0.03 Manganous Chloride 0.0000001
    4H2O
    L-Aspartic Acid 0.2 Pyridoxine HCl 0.001
    L-Cysteine HCl H2O 0.2 Thiamine HCl 0.001
    L-Glutamic Acid 0.01 Ferric Nitrate 9H2O 0.0008
    L-Histidine HCl H2O 0.06 Magnesium Sulfate 0.02
    L-Isoleucine 0.2 Zinc Sulfate 0.0004
    L-Leucine 0.3 Zinc Chloride 0.0003
    L-Lysine HCl 0.2 Ascorbic Acid 0.0000003
    L-Methionine 0.1 D-Calcium Pantothenate 0.001
    L-Phenylalanine 0.1 Calcium Chloride 0.01
    L-Serine 0.4 Magnesium Chloride 0.08
    L-Threonine 0.1 Potassium Chloride 0.3
    L-Tryptophan 0.04 Sodium Selenite 0.000000007
    L-Valine 0.2 Vitamin B12 0.001
    L-Tyrosine 2Na 2H2O 0.1 Choline Chloride 0.01
    L-Glutamine 0.6 i-Inositol 0.02
    (recommended
    addition, formulation
    does not contain)
    D-Glucose (Dextrose) 5 Niacinamide 0.001
    Lipoic Acid 0.002 Ethanolamine HCl 0.005
    Linoleic Acid 0.00006 Putrescine 2HCl 0.0002
    Folic Acid 0.0005 Sodium Pyruvate 0.1
    HEPES 3.0 Riboflavin 0.0002
    2-Hydroxypyridine-n- 0.003 Biotin 0.0001
    oxide
    Pluronic F-68* 0.3 Sodium Bicarbonate 2.0
    Sodium Phosphate 0.1 Sodium Chloride*** 3.0***
    Monobasic H2O

    ***Added to adjust osmolarity. Quantity will vary somewhat between formulations. NaCL is added to adjust osmo from ˜160 to a value within 275 +/− 5 mOsmo
  • TABLE 3
    Well Defined Medium
    Gram(s)/L Gram(s)/L
    (unless noted) (unless noted)
    Component about: Component about:
    L-Arginine HCl 0.43 Ammonium Meta Vanadate 0.0000006
    L-Asparagine 0.0264 Manganous Chloride 4H2O 0.0000001
    L-Aspartic Acid 0.15 Pyridoxine HCl 0.00103
    L-Cysteine HCl H2O 0.15 Thiamine HCl 0.00112
    L-Glutamic Acid 0.01 Ferric Nitrate 9H2O 0.00081
    L-Histidine HCl 0.06 Magnesium Sulfate 0.0241
    H2O
    L-Isoleucine 0.2 Zinc Sulfate 0.000375
    L-Leucine 0.3 Zinc Chloride 0.00025
    L-Lysine HCl 0.25 Ascorbic Acid 0.00000025
    L-Methionine 0.115 D-Calcium Pantothenate 0.00119
    L-Phenylalanine 0.1 Calcium Chloride 0.0111
    L-Serine 0.388 Magnesium Chloride 0.0762
    L-Threonine 0.12 Potassium Chloride 0.2763
    L-Tryptophan 0.04 Sodium Selenite 0.0000000067
    L-Valine 0.19 Vitamin B12 0.00103
    L-Tyrosine 2Na 0.1 Choline Chloride 0.014
    2H2O
    L-Glutamine 0.584 i-Inositol 0.018
    (recommended
    addition,
    formulation does not
    contain)
    D-Glucose 5 Niacinamide 0.00122
    (Dextrose)
    Lipoic Acid 0.002 Ethanolamine HCl 0.005
    Linoleic Acid 0.00006 Putrescine 2HCl 0.00016
    Folic Acid 0.0005 Sodium Pyruvate 0.11
    HEPES 2.98 Riboflavin 0.00022
    2-Hydroxypyridine- 0.003 Biotin 0.000097
    n-oxide
    Pluronic F-68* 0.3 Sodium Bicarbonate 2.4
    Sodium Phosphate 0.125 Sodium Chloride*** 3.126***
    Monobasic H2O

    ***Added to adjust osmolarity. Quantity will vary somewhat between formulations. NaCL is added to adjust osmo from ˜160 to a value within 275 +/− 5 mOsmo
  • The media provided by the present invention can be protein-free, and can be a 1× formulation or concentrated as, for example, a 10×, 20×, 25×, 50×, 10×, 500×, or 1000× medium formulation.
  • The media of the invention can also be prepared in different forms, such as dry powder media (“DPM”), a granulated preparation (which requires addition of water, but not other processing, such as pHing), liquid media or as media concentrates.
  • The basal medium that is a medium useful only for maintenance, but not for growth or production of product, can comprise a number of ingredients, including amino acids, vitamins, organic and inorganic salts, sugars and other components, each ingredient being present in an amount which supports the cultivation of a mammalian epithelial cell in vitro.
  • In the medium, methods, kit and composition of the present invention, the medium can be used to culture a variety of cells. Preferably, the medium is used to culture eukaryotic cells. More preferably, the medium is used to culture plant and/or animal cells. More preferably, the medium is used to culture mammalian cells, fish cells, insect cells, amphibian cells or avian cells. More preferably, the medium is used to culture mammalian cells. More preferably, the medium is used to culture mammalian cells, including primary epithelial cells (e.g., keratinocytes, cervical epithelial cells, bronchial epithelial cells, tracheal epithelial cells, kidney epithelial cells and retinal epithelial cells) and established cell lines and their strains (e.g., 293 embryonic kidney cells, BHK cells, HeLa cervical epithelial cells and PER-C6 retinal cells, MDBK (NBL-1) cells, 911 cells, CRFK cells, MDCK cells, CHO cells, BeWo cells, Chang cells, Detroit 562 cells, HeLa 229 cells, HeLa S3 cells, Hep-2 cells, KB cells, LS180 cells, LS174T cells, NCI-H-548 cells, RPMI 2650 cells, SW-13 cells, T24 cells, WI-28 VA13, 2RA cells, WISH cells, BS-C-I cells, LLC-MK2 cells, Clone M-3 cells, 1-10 cells, RAG cells, TCMK-1 cells, Y-1 cells, LLC-PK1 cells, PK(15) cells, GH1 cells, GH3 cells, L2 cells, LLC-RC 256 cells, MH1C1 cells, XC cells, MDOK cells, VSW cells, and TH-I, B1 cells, or derivatives thereof), fibroblast cells from any tissue or organ (including but not limited to heart, liver, kidney, colon, intestines, esophagus, stomach, neural tissue (brain, spinal cord), lung, vascular tissue (artery, vein, capillary), lymphoid tissue (lymph gland, adenoid, tonsil, bone marrow, and blood), spleen, and fibroblast and fibroblast-like cell lines (e.g., CHO cells, TRG-2 cells, IMR-33 cells, Don cells, GHK-21 cells, citrullinemia cells, Dempsey cells, Detroit 551 cells, Detroit 510 cells, Detroit 525 cells, Detroit 529 cells, Detroit 532 cells, Detroit 539 cells, Detroit 548 cells, Detroit 573 cells, HEL 299 cells, IMR-90 cells, MRC-5 cells, WI-38 cells, WI-26 cells, MiCl1 cells, CHO cells, CV-1 cells, COS-1 cells, COS-3 cells, COS-7 cells, Vero cells, DBS-FrhL-2 cells, BALB/3T3 cells, F9 cells, SV-T2 cells, M-MSV-BALB/3T3 cells, K-BALB cells, BLO-11 cells, NOR-10 cells, C3H/IOTI/2 cells, HSDM1C3 cells, KLN2O5 cells, McCoy cells, Mouse L cells, Strain 2071 (Mouse L) cells, L-M strain (Mouse L) cells, L-MTK (Mouse L) cells, NCTC clones 2472 and 2555, SCC-PSA1 cells, Swiss/3T3 cells, Indian muntjac cells, SIRC cells, CII cells, and Jensen cells, or derivatives thereof). More preferably, the medium is used to culture mammalian cells selected from the group consisting of 293 cells, PER-C6 cells, CHO hells, COS cells and Sp2/0 cells. More preferably, the medium is used to culture 293 cells. Preferably, the medium is used to culture cells in suspension.
  • Cells supported by the medium of the present invention can be derived from any animal, preferably a mammal, and most preferably a mouse or a human. The cells cultivated in the present media can be normal cells or abnormal cells (i.e., transformed cells, established cells, or cells derived from diseased tissue samples).
  • The present invention also provides methods of cultivating mammalian epithelial or fibroblast cells using the culture medium formulations disclosed herein, comprising (a) contacting the cells with the cell culture media of the invention; and (b) cultivating the cells under conditions suitable to support cultivation of the cells. In some embodiments, the methods of the present invention can optionally include a step of contacting the cultured cells with a solution comprising one or more macromolecules (preferably comprising one or more nucleic acids) under conditions causing the introduction of one or more of the macromolecules into one or more of the cells. Preferably, cells cultivated according to these methods (which can include any of the cells described above) are cultivated in suspension.
  • The invention further provides compositions comprising the culture media of the present invention, which optionally can further comprise one or more mammalian epithelial or fibroblast cells, such as those described above, particularly one or more 293 embryonic kidney cells, PER-C6 retinal cells, and CHO cells.
  • The present invention further relates to methods of cultivating mammalian cells (particularly those described above and most particularly 293 embryonic kidney epithelial cells, PER-C6, and CHO cells) in suspension comprising (a) obtaining a mammalian cell to be cultivated in suspension; and (b) contacting the cell with the culture media of the invention under conditions sufficient to support the cultivation of the cell in suspension.
  • The present invention further relates to methods of producing a virus, and to viruses produced by these methods, the methods comprising (a) obtaining a mammalian cell, preferably a mammalian cell described above, e.g., a primate, especially simian and human, and most preferably a 293 embryonic kidney epithelial cell, PER-C6 cell, 911 cell, or CHO cell, to be infected with a virus; (b) contacting the cell with a virus under conditions suitable to promote the infection of the cell by the virus; and (c) cultivating the cell in the culture medium of the invention under conditions suitable to promote the production of the virus by the cell. Viruses which can be produced according to these methods include adenoviruses, adeno-associated viruses and retroviruses.
  • The present invention further relates to methods of producing a polypeptide, and to polypeptides produced by these methods, the methods comprising (a) obtaining a cell, preferably a mammalian cell described above and most preferably a 293 embryonic kidney epithelial cell, PER-C6, or CHO cell; (b) contacting the cell with a solution comprising a nucleic acid encoding the polypeptide under conditions causing the introduction of the nucleic acid into the cell; and (c) cultivating the cell in the culture medium of the invention under conditions favoring the expression of the desired polypeptide by the cell.
  • The present invention is also directed to compositions, particularly cell culture media, comprising one or more replacement compounds. In some embodiments, the replacement compounds can be metal binding compounds and/or one or more transition elements in a complex with one or more metal binding compounds. Such cell culture media of the invention can be used to grow or cultivate plant cells, animal cells (particularly human cells), insect cells, bacterial cells, yeast cells and more generally any type of eukaryotic or prokaryotic cells. Thus, the replacement compounds of the present invention can be added to any type or kind of culture media, and are preferably used to replace naturally derived metal carriers (e.g., animal derived proteins or extracts such as transferrin) in such media. The invention is also directed to methods of use of such compositions, including, for example, methods for the cultivation of eukaryotic cells, particularly animal cells, in vitro. The invention also relates to compositions comprising such culture media and one or more cells, especially those cells specifically referenced herein, and to kits comprising one or more of the above-described compositions.
  • In another aspect, the invention relates to a kit for the cultivation of cells in vitro. The kit comprise one or more containers, wherein a first container contains the culture medium of the present invention. The kit can further comprise one or more additional containers, each container containing one or more supplements selected from the group consisting of one or more cytokines, heparin, one or more animal or animal-derived peptides, one or more yeast peptides and one or more plant peptides (which are preferably one or more peptides from rice, aloevera, soy, maize, wheat, pea, squash, spinach, carrot, potato, sweet potato, tapioca, avocado, barley coconut and/or green bean, and/or one or more other plants).
  • The kit of the present invention can further comprise one or more containers comprising a nucleic acid and/or a reagent that facilitates the introduction of at least one macromolecule, e.g., a nucleic acid into cells cultured in the media of the present invention, i.e., a transfection reagent. Preferred transfection reagents include, but are not limited to, cationic lipids and the like.
  • A kit according to one aspect of the invention can comprise one or more of the culture media of the invention, one or more replacement compounds, which can be one or more metal binding compounds, and/or one or more transition element complexes, and can optionally comprise one or more nucleic acids and transfection reagents. Kits according to another aspect of the invention can comprise one or more cell culture media (one of which can be a basal medium) and one or more replacement compounds. Preferred replacement compounds include, but are not limited to, metal binding compounds and/or transition element complexes, said complexes comprising at least one transition element or a salt or ion thereof complexed to at least one metal-binding compound. Preferred transition elements, metal-binding compounds, and transition element complexes for use in the kits according to this aspect of the invention include those described in detail herein.
  • The kit of the present invention can also contain instructions for using the kit to culture cells and/or introduce macromolecules or compounds (e.g., nucleic acid, such as DNA), into cells.
  • It will be readily apparent to one of ordinary skill in the relevant arts that other suitable modifications and adaptations to the methods and applications described herein are obvious and can be made without departing from the scope of the invention or any embodiment thereof. Having now described the present invention in detail, the same will be more clearly understood by reference to the following examples, which are included herewith for purposes of illustration only and are not intended to be limiting of the invention.
  • EXAMPLE 1
  • Semi-confluent adherent cultures of 293 Cells (ATCC, CRL 1573) are readily adapted to suspension culture. The cells are first detached with a solution of Trypsin-EDTA (0.05% Trypsin, 0.53 mM Na4EDTA), and then resuspended in conventional medium supplemented with 10% FBS to inhibit the trypsin. The resuspended cells are centrifuged at 200×g for five minutes. The cell pellet is resuspended in 293 SFM (available from Invitrogen Corporation, Carlsbad, Calif.) the formulation of which is described in WO 98/08934 which is specifically incorporated herein by reference. Alternatively, the cells can be detached with VERSENE (Na4EDTA, 0.53 mM) and resuspended in 293 SFM.
  • The initial seeding density of the 293 cells after conversion to suspension culture is 1×106 cells/mL. The cells are shaken on a rotary shaker at 150 rpm in a 37° C. incubator equilibrated with 8% CO2-92% air. When the cells reach a density of 1.5×106 cells/mL they are diluted with 293 SFM to a density of 3.0×105 cells/mL. Because 293 cells have a tendency to aggregate, the cells can be vortexed vigorously for approximately 45 seconds to obtain a predominantly single cell suspension at the time of passaging and counting. After several passages in suspension culture, the maximum achievable density can be determined. The 293 cells which are described here were grown to approximately 3-4×106 cells/mL in suspension culture.
  • The ability of various replacement compounds to support cellular growth was evaluated using 293 cells maintained in 293 SFM (Invitrogen Corporation, Carlsbad, Calif.) without transferrin or replacement compound. Stock cultures containing 5 μg/mL human holo-transferrin were prepared in 293 SFM. For replacement compound evaluation, 293 cells were established in 125 mL shaker flasks at an initial viable seeding density of 2×105 cells/mL in a final volume of 20 mL. All cultures were maintained at 37° C. in humidified air containing 8% CO2. To eliminate transferrin carry-over effects, cells were subcultured at 4 day intervals for a total of three passages. At each subculturing, cells were seeded at a density of 2×105 cells/mL. Positive control cultures contained 5 μg/mL human holo-transferrin while negative control cultures were established in the absence of either transferrin or replacement compound. Replacement compound stocks were prepared at 0.1M-0.2M in ddH2O and solubilized when necessary using 5N NaOH or HCl. Iron complexes were established using 0.2 M replacement compound and iron stocks mixed 1:1 (v/v) and incubated for 5-10 minutes at 22° C. All solutions containing replacement compounds were filter sterilized using a 0.22 μm Millex-GV filter prior to addition to transferrin and serum-free media.
  • Except where noted, replacement compounds were evaluated either alone or in combination with ferric chloride or ferrous sulfate at 25 μM, 50 EM and 100 μM. When used in combination, the replacement compounds were used in equimolar concentration with iron ions. Values in Table 4 represent the mean percentage of control growth for duplicate cultures determined at the third passage. Media that was not supplemented, i.e., did not contain either transferrin or replacement compound, failed to support the growth of the cells over three passages.
  • The results of 293 cells grown in the presence of various replacement compounds are shown in Table 4. When added to the medium formulation un-complexed, the replacement compound is listed alone, when added as a complex with a transition metal, the source of the transition metal is listed with the replacement compound.
  • In this and in other examples, 293 SFM medium was used. 293 SFM medium is no longer available from Invitrogen (Carlsbad, Calif.), but similar results would be expected using 293 SFM II medium (Invitrogen, Carlsbad, Calif.).
    TABLE 4
    Effect of Replacement Compounds on the Growth of 293 Cells
    Replacement Concentration
    compound tested 25 μM 50 μM 100 μM
    2-Hydroxypyridine-N-Oxide 117 150 Not
    (293 tested at 10-50 μM) Tested
    3-Hydroxypyridine-N- 4 4 4
    Oxide
    3-Hydroxypyridine-N- 77 72 81
    Oxide•Ferric Chloride
    3-Hydroxypyridine-N- 104 58 0
    Oxide•Ferrous Sulfate
    Sorbitol•Ferrous 58 69 83
    Sulfate
    Deferoxamine Mesylate•Ferric 101 120 127
    Chloride (5 μM) (10 μM) (20 μM)
    Acetohydroxamic Acid•Ferric 83 22 0
    Chloride
    Serine Hydroxamate•Ferric 68 65 51
    Chloride
    Glycine•Ferric Chloride 81 71 72
    Nitriloacetic Acid•Ferric 100 118 74
    Chloride
    Nitriloacetic Acid•Ferrous 75 95 0
    Sulfate
    3-Hydroxy-2-Methyl-4-Pyrone 0 28 81
    (Maltol)
    2-Ethyl-3-Hydroxy-4-Pyrone 59 120 126
    (Ethyl Maltol)
    2,2-Dipyridylamine•Ferric 118 96 101
    Chloride
    2,2-Dipyridyl•Ferric Chloride 51 96 0
    Diethylenetriamine Penta- 79 96 102
    Acetic Acid•Ferric Chloride
    Diethylenetriamine Penta- 77 107 85
    Acetic Acid•Ferrous Sulfate
    Nicotinic Acid-N-Oxide•Ferric 104 80 79
    Chloride
    2-Hydroxynicotinic Acid•Ferric 0 0 99
    Chloride
    (293 tested at 100, 150, 200 μM)
    Ferrous Gluconate•Ascorbic 86 109 101
    Acid Phosphate
    Aspartic Acid•Ferric Chloride 81 86 100
    Glutamic Acid•Ferric Chloride 102 68 83
    N-Acetyl-Cysteine•Ferric 88 82 52
    Chloride
    4-Pyridoxic Acid•Ferric 109 85 99
    Chloride
    2-Pyridinecarboxylic 67 48 0
    Acid•Ferrous Sulfate
    Myo-Inositol•Ferric Chloride 79 87 85
    Mimosine•Ferric Chloride 64 55 80
    Ferrous Sulfate 71 60 41
    Ferric Chloride 87 91 109
  • EXAMPLE 2
  • The ability of replacement compounds to support cellular growth in the absence of transferrin was evaluated using CHO cells maintained in CD CHO medium (Invitrogen Corporation, Carlsbad, Calif., the formulation of which is described in WO 98/08934). Stock cultures containing 5 μg/mL human holo-transferrin were prepared in CD CHO medium. For replacement compound evaluation, CHO cells were established in 125 mL shaker flasks at an initial viable seeding density of 1×105 cells/mL in a final volume of 20 mL. All cultures were maintained at 37° C. in humidified air containing 8% CO2. To eliminate transferrin carry-over effects, cells were subcultured at 4 day intervals for a total of three passages. Positive control cultures contained 5 ag/mL human holo-transferrin while negative control cultures were established in the absence of either transferrin or replacement compound. Replacement compound stocks were prepared at 0.1M-0.2M in ddH2O and solubilized when necessary using 5N NaOH or HCl. Iron complexes were established using 0.2M replacement compound and iron stocks mixed 1:1 (v/v) and incubated for 5-10 minutes at 22° C. All solutions containing replacement compounds were filter sterilized using a 0.22 μm Millex-GV filter prior to addition to transferrin and serum-free media.
  • Except where noted, replacement compounds were evaluated either alone or in combination with ferric chloride or ferrous sulfate at 25 μM, 50 μM and 100 μM. Values in the table represent the mean percentage of control growth for duplicate cultures determined at the third passage. Media that was not supplemented, i.e., did not contain either transferrin or replacement compounds, failed to support the growth of the cells over three passages.
  • The ability of various replacement compounds to substitute for transferrin in the culture of CHO cells was determined. The results are shown in Table 5. When added to the medium formulation un-complexed, the replacement compound is listed alone, when added as a complex with a transition metal, the source of the transition metal is listed with the metal binding compound.
    TABLE 5
    Effect of Replacement Compounds on the Growth of CHO Cells
    Concentration
    Replacement compound tested 25 μM 50 μM 100 μM
    2-Hydroxypyridine-N-Oxide 127 117 83
    3-Hydroxypyridine-N-Oxide•Ferric 77 66 101
    Chloride
    Sorbitol•Ferric Chloride 100 107 112
    Deferoxamine Mesylate•Ferric 50 74 52
    Chloride (5 μM) (10 μM) (20 μM)
    Serine Hydroxamate•Ferric Chloride 106 121 124
    Lysine•Ferric Chloride 51 79 98
    Nitriloacetic Acid•Ferric Chloride 127 107 107
    2-Ethyl-3-Hydroxy-4-Pyrone 9 60 113
    2,2-Dipyridylamine•Ferric Chloride 40 48 65
    Diethylenetriamine Penta-Acetic 135 137 147
    Acid•Ferrous Sulfate
    Nicotinic Acid-N-Oxide•Ferric 60 84 105
    Chloride
    Ferrous Gluconate•Ascorbic Acid 91 61 28
    Phosphate
    Aspartic Acid•Ferric Chloride 77 89 112
    Cysteine•Ferrous Sulfate 83 64 45
    4-Pyridoxic Acid•Ferric Chloride 55 63 100
    2-Pyridinecarboxylic Acid•Ferric 88 89 100
    Chloride
    Kojic Acid
    0 0 0
    Kojic Acid•Ferric Chloride 89 88 85
    2,3-Dihydrobenzoic Acid•Ferric 71 0 0
    Chloride
    Ferrous Sulfate 88 79 38
    Ferric Chloride 56 85 101
  • EXAMPLE 3
  • The ability of replacement compounds to support cellular growth in the absence of transferrin was evaluated using Sp2/0 cells maintained in CD Hybridoma medium (Invitrogen Corporation, Carlsbad, Calif.).
  • Stock cultures containing 5 μg/mL human holo-transferrin were prepared in CD Hybridoma medium. For replacement compound evaluation, Sp2/0 cells were established at an initial viable cell density of 0.5×105 cells/mL in stationary culture using 75 cm tissue culture flasks in a final volume of 20 mL. All cultures were maintained at 37° C. in humidified air containing 8% CO2. To eliminate transferrin carry-over effects, cells were subcultured at the same seeding density at 4 day intervals for a total of three passages. Positive control cultures contained 5 μg/mL human holo-transferrin while negative control cultures were established in the absence of either transferrin or replacement compound. Replacement compound stocks were prepared at 0.1M-0.2M in ddH2O and solubilized when necessary using 5N NaOH or HCl. Iron complexes were established using 0.2M replacement compound and iron stocks mixed 1:1 (v/v) and incubated for 5-10 minutes at 22° C. All solutions containing replacement compounds were filter sterilized using a 0.22 μm Millex-GV filter prior to addition to transferrin and serum-free media.
  • Except where otherwise noted, replacement compounds were evaluated either alone or in combination with ferric chloride or ferrous sulfate at 25 μM, 50 μM and 100 μM. Values in the table represent the mean percentage of control growth for duplicate cultures determined at the third passage. Media that was not supplemented, i.e., did not contain either transferrin or replacement compounds, failed to support the growth of the cells over three passages.
  • The ability of various replacement compounds to substitute for transferrin in the culture of Sp2/0 cells was determined and the results are seen in Table 6. When added to the medium formulation un-complexed, the replacement compound is listed alone, when added as a complex with a transition metal, the source of the transition metal is listed with the replacement compound.
    TABLE 6
    Effect of Replacement Compounds on the Growth of Sp2/0 Cells
    Concentration
    Replacement compound tested 25 μM 50 μM 100 μM
    2-Hydroxypyridine-N-Oxide 98 93 89
    3-Hydroxypyridine-N-Oxide•Ferric 55 54 57
    Chloride
    Sorbitol•Ferric Chloride 94 55 60
    Deferoxamine Mesylate•Ferric 0 0 0
    Chloride (5 μM) (10 μM) (20 μM)
    (All lines tested at 5, 10, 20 μM)
    Acetohydroxamic Acid•Ferric Chloride 40 48 47
    (Sp2 tested at 5, 10, 20 μM)
    Serine Hydroxamate•Ferric Chloride 46 66 62
    Glycine•Ferric Chloride 34 61 56
    Nitriloacetic Acid•Ferric Chloride 88 87 70
    Nitriloacetic Acid 0 0 0
    3-Hydroxy-2-Methyl-4-Pyrone (Maltol) 0 0 0
    3-Hydroxy-2-Methyl-4-Pyrone•Ferric 60 71 75
    Chloride
    2-Ethyl-3-Hydroxy-4-Pyrone (Ethyl 0 75 116
    Maltol)
    Diethylenetriamine Penta-Acetic 54 90 91
    Acid•Ferrous Sulfate
    2-Hydroxynicotinic Acid•Ferric 64 82 85
    Chloride)
    Ferrous Gluconate•Ascorbic Acid 92 94 93
    Phosphate
    Glutamine•Ferric Chloride 36 55 65
    Asparagine•Ferric Chloride 36 51 54
    Cysteine•Ferrous Sulfate 85 79 67
    4-Pyridoxic Acid•Ferric Chloride 40 73 76
    2-Pyridinecarboxylic Acid•Ferric 0 30 48
    Chloride
    Morpholine•Ferric Chloride 54 64 81
    3-Hydroxy-2-Nitropyridine•Ferric 52 62 72
    Chloride
    Kojic Acid
    0 0 0
    Kojic Acid•Ferric Chloride 0 0 0
    Ferrous Sulfate 91 103 94
    Ferric Chloride 55 73 74
  • EXAMPLE 4 Protocol Used for Transfections
  • Cells used were 293-F cells cultured either in suspension or attached. 293-F cells are a clone of 293 cells obtained from Robert Horlick at Pharmacopeia, Princeton, N.J. Cells were adapted to the test media for several passages (>4) prior to transfection experiments. When cultured in suspension, cells were split to 3×105 viable cells per ml at each passage. On the week of transfection cells were seeded at 3×105 viable cells per ml. After ˜48 hrs of growth (day 2) cells were vortexed for 30 seconds and counted and appropriate volumes used for transfection as described in procedures below.
  • The composition of the base media used for the transfection experiments is shown in Table 7.
    TABLE 7
    COMP.
    GP ID# COMPONENTS GM/100 L GM/L
    1 8101009 L-Arginine HCl 43 0.430
    1 8101013 L-Asparagine Anhyd 2.64 0.026
    1 8101016 L-Aspartic Acid 7.5 0.075
    1 8101035 L-Cysteine HCl H2O 7.5 0.075
    1 8101048 L-Glutamic Acid 2.94 0.029
    1 8101062 L-Histidine HCl H2O 5.7 0.057
    1 8101072 L-Isoleucine 19 0.190
    1 8101077 L-Leucine 28 0.280
    1 8101083 L-Lysine HCl 25.5 0.255
    1 8101086 L-Methionine 11.5 0.115
    1 8101095 L-Phenylalanine 7 0.070
    1 8101101 L-Serine 25 0.250
    1 8101104 L-Threonine 6 0.060
    1 8101110 L-Tryptophan 2 0.020
    1 8101116 L-Valine 19 0.190
    1 8801985 L-Tyrosine Disodium Salt 10 0.100
    2 8205023 D-Glucose (Dextrose) 200 2.00
    2 8503100 Vitamin B12 0.103 0.0010
    2 8503140 Biotin 0.0097 0.000097
    2 8503200 Choline Chloride 1.4 0.014
    2 8503370 Folic Acid 0.5 0.005
    2 8503390 i-Inositol 1.8 0.018
    2 8503450 Niacinamide 0.122 0.0012
    2 8503460 D Calcium Pantothenate 0.119 0.0012
    2 8503510 Pyridoxine HCl 0.103 0.0010
    2 8503520 Riboflavin 0.022 0.0002
    2 8503540 Thiamine HCl 0.112 0.0011
    2 8800517 Ethanolamine HCl 0.5 0.005
    2 8801680 Putrescine 2HCl 0.016 0.0002
    2 8901840 Sodium Pyruvate 11 0.110
    3 8205023 D-Glucose (Dextrose) 200 2.00
    3 8503560 DL Lipoic Acid Thioctic (see 0.2 0.0020
    Step 1 process instruction
    below)
    3 8801225 Linoleic Acid (see Step 1 0.006
    process instruction below)
    4 0855845 Opti-MEM (100000X) TES 1 0.010
    4 8800615 Ferric Nitrate 9H2O (see Step 0.081 0.0008
    2 process instruction below)
    4 8801314 Magnesium Chloride Anhyd 7.62 0.076
    4 8801340 Magnesium Sulfate Anhyd 2.41 0.024
    4 8801635 Potassium Chloride 27.6 0.276
    4 8802626 Zinc Sulfate H2O (see Step 2 0.00874
    process instruction below)
    4 8951830 Sodium Chloride 441 4.41
    4 8951837 Sodium Phosphate 12.5 0.125
    Monobasic H2O
    5 8800320 Calcium Chloride Anhyd 1.47 0.015
    6 8205023 D-Glucose (Dextrose) 50 0.500
    6 8701160 Phenol Red 0.1 0.0010

    In addition, the base media comprises 2.4 g/L NaHCO3, 2.98 g/L Hepes, 4 mM L-glutamine and 3 ml/L PLURONIC F-68 (Invitrogen Corp., Carlsbad, Calif.)
  • The osmolarity is adjusted to from about 275-310 mOsm, preferably about 280 mOsm and the pH is adjusted to from about 7.0 to about 7.45 preferably about 7.4 respectively. The above media is referred to as 293 SFM w/o insulin, w/o transferrin, w/o citrate chelate, w/o dextran sulfate, with L-glutamine and with PLURONIC F-68. To that media, one or more of the replacement compounds of the invention and one or more metals can be added, for example, 25 μM 2-hydroxypyridine-N-oxide and 0.375 mg/L ZnSO4.7H20 can be added for the transfection media.
  • For transfection experiments in 15 ml shaker flasks, a volume of cells corresponding to 1.5×107 viable cells was centrifuged and the supernatant was removed. The pellets were suspended in 14.5 ml fresh test media. Each medium was tested in duplicate.
  • The nucleic acid was prepared for transfection by combining 700 μl test media+60 μl DNA (the DNA used was plasmid pCMV-SPORT-β-gal available from Invitrogen Corporation, Carlsbad, Calif. Cat# 10586-014)+240 μl LIPOFECTAMINE 2000 (available from Invitrogen Corporation, Carlsbad, Calif. Cat# 11668-019) in a microfuge tube and incubating the mixture for 30 minutes at room temperature in order to form complexes of nucleic acid and transfection reagent. In this and all subsequent experiments, when complexing DNA and lipid for transfections, the additions were always done by first adding the DNA to the tested media, then adding the LIPOFECTAMINE 2000 to the DNA-tested media mix. After complex formation, 5001 of the complex was added to each of the duplicate shaker flasks. The final cell concentration was 1×106 viable cells per ml, the final DNA concentration was 11 g per 1×106 viable cells and the final LIPOFECTAMINE 2000 concentration was 8 μl per 1×106 viable cells.
  • When 10 ml shaker flasks were used, a volume of cells corresponding to 1.0×107 viable cells was centrifuged and the supernatant was removed. The pellets were re-suspended in 9.5 ml fresh test media. In a microfuge tube 800 μl test media+40 μl DNA+160 μl LIPOFECTAMINE 2000 were combined and incubated for 30 minutes to permit complex formation and 500 μl of the complex added to each of the shaker flasks. The final cell concentration in the suspension in the shaker flasks was 1×106 viable cells per ml, the final DNA concentration was 1 μg per 1×106 viable cells and the final LIPOFECTAMINE 2000 concentration was 8 μl per 1×106 viable cells.
  • After 24 hrs, 2 ml samples were removed and the β-galactosidase activity determined. Each 2 ml sample was centrifuged, the supernatant removed and the pelleted cells were re-suspended pellets in 1 ml of Dulbecco's-PBS (D-PBS, Invitrogen Corporation, Carlsbad, Calif.). The cells were then centrifuged and re-suspended a second time in 1 ml of D-PBS. The samples were stored at −70° C. Prior to the assay for β-galactosidase, samples were subjected to cycles of freezing and thawing a total of five times. Due to the high amounts of β-galactosidase activity, 10 μl and 20 μl samples were run against a standard curve ranging from 0-700 ng β-galactosidase.
  • For determination of β-galactosidase activity by in situ staining, 1 ml samples (i.e., ˜1×106 cells) were used.
  • Protocol Used To Transfect 293-F Cells In 24-well plates:
  • Three wells were set up for each treatment. A volume of media corresponding to 3×106 viable cells was centrifuged. The supernatant was removed and the cells re-suspended in 6001 test media. Each well of a 24 well plate received 200 μl of the re-suspended cells.
  • To prepare the nucleic acid for the transfection experiments, 93 μl test media+3 μl DNA+24 μl LIPOFECTAMINE 2000 were combined in a microfuge tube and complexed for 30 minutes. A 40 μl aliquot of complex was added to each well. 250 μl test media was added to each well five hours after addition of complex. The final cell concentration was 1×106 viable cells per well; the final DNA concentration was 1 μg per 1×106 viable cells and the final LIPOFECTAMINE 2000 concentration was 8 μl per 1×106 viable cells.
  • 24 hrs post-transfection, samples were collected for β-galactosidase quantitation. The contents of duplicate wells were pooled (i.e. ˜2×106 cells for the assay) for each sample and transferred to a microfuge tube. The cells were centrifuged, the supernatant was removed and the cells were re-suspended in 1 ml of D-PBS. The cells were pelleted a second time and re-suspended in 1 ml of D-PBS. The samples were then stored at −70° C. Prior to the assay for β-galactosidase, samples were freeze-thawed a total of five times. Aliquots (15 μl and 25 μl) of each sample were assayed and the activity determined by comparison to a standard curve ranging from 0-70 ng β-galactosidase.
  • The third well of each treatment was used for in situ staining for β-galactosidase activity (i.e., ˜1×106 cells).
  • EXAMPLE 5 Comparison of Transfection Efficiencies of Cells Grown in the Presence of Various Replacement Compounds
  • 293 cells were grown in monolayer culture in the presence of the various replacement compounds indicated and transfection experiments performed as described above. The results are shown in Table 8 and the values are β-galactosidase activity expressed as nanograms of β-galactosidase per 2×106 cells. A comparison was made between the transfection efficiency of DNA complexes formed in D-MEM medium and DNA complexes formed in test medium. The replacement compounds were present at a concentration of 25 μM.
    TABLE 8
    Test Media
    D-MEM used for used for
    complexing, Test complexing
    Replacement compound in media to and to re-
    ID test media: re-suspend cells suspend cells
    A Human Transferrin (as 326 3279
    control)
    B 2-hydroxypyridine-N- 354 1314
    oxide
    C 3-hydroxypyridine-N- 176 317
    Oxide•Ferrous sulfate
    D Deferoxamine 154 301
    Mesylate•Ferric Chloride
    E 2-ethyl-3-hydroxy-4- 378 1827
    pyrone (ethyl maltol)
    F 2,2-dipyridylamine•ferric 347 459
    chloride
    G Diethylene-triamine-penta 131 574
    acetic acid•Ferric chloride
    H Diethylene-triamine-penta 234 720
    acetic acid•Ferrous sulfate
    I Ferrous 116 275
    Gluconate•Ascorbic Acid
    J Ferric Chloride 18 0
    K 2-methyl-3-hydroxy-4- 103 52
    pyrone
    L Nitrilotriacetic acid•ferric 45 33
    chloride
    M Ferrous sulfate 258 406
    Stock Stock (293 SFM II 3 0
    (293-II) medium)
    D-MEM + D-MEM + 10% FBS + 1054 1464
    10% FBS 0•1 mM NEAA
  • Transfection efficiency was generally better when test media rather than D-MEM was used for complexing the DNA and lipid. The transferrin control (ID# A) worked best, reflecting that transferrin facilitates transfections. It should also be noted that cell growth in the above test media was comparable to growth in media containing transferrin and to growth in 293 SFM-II.
  • Base media for test media was 293-II+insulin+L-glutamine+PLURONIC F-68 (−) dextran sulfate (−) transferrin (+) replacement compound (A-M for test media, see table).
  • EXAMPLE 6 Effect of Varying Concentration of Replacement Compound and Effect of Media Used to Form Nucleic Acid Complexes
  • 293 cells were grown in monolayer culture in base medium supplemented with the indicated replacement compounds at the concentrations specified in Table 9. The top number in the concentration column is the concentration of the first supplement and the second number in the concentration column is the concentration of the second supplement. The effects of the medium used to form DNA complexes were tested. The results are presented as nanograms of β-galactosidase per 2×106 cells.
    TABLE 9
    Test Media
    D-MEM used used for
    for complexing, complexing
    Compound added to Concentration Test media to re- and to re-
    ID base media added suspend cells suspend cells
    A +Human Transferrin (as 5 μg/ml 401 550
    control) +
    Insulin 10 μg/ml
    B +2-ethyl-3-hydroxy-4- 50 μM 245 997
    pyrone (ethyl maltol) +
    insulin 10 μg/ml
    C +2-ethyl-3-hydroxy-4- 100 μM Discontinued at p1 of adaptation to
    pyrone (ethyl maltol) + media due to v. poor growth
    insulin
    10 μg/ml
    D +2-hydroxypyridine-N- 25 μM 361 2054
    oxide +
    insulin 10 μg/ml
    E +2-hydroxypyridine-N- 50 μM Discontinued at p1 of adaptation to
    oxide + media due to v. poor growth
    insulin
    10 μg/ml
    F +Diethylene-triamine- 50 μM 262 1029
    penta acetic
    acid•Ferrous sulfate
    (DPTA•FeSO4) +
    insulin 10 μg/ml
    G +Transferrin + 5 μg/ml 635 1700
    ZnSO4 0.375 μg/ml
    H +2-ethyl-3-hydroxy-4- 50 μM 242 395
    pyrone (ethyl maltol) +
    ZnSO4 0.375 μg/ml
    I +2-ethyl-3-hydroxy-4- 100 μM Discontinued at p1 of adaptation to
    pyrone (ethyl maltol) + media due to v. poor growth
    ZnSO4 0.375 μg/ml
    J +2-hydroxypyridine-N- 25 μM 1066 2937
    oxide +
    ZnSO4 0.375 μg/ml
    K +2-hydroxypyridine-N- 50 μM Discontinued at p1 of adaptation to
    oxide + media due to v. poor growth
    ZnSO4 0.375 μg/ml
    L +Diethylene-triamine- 50 μM 378 2372
    penta acetic
    acid•Ferrous sulfate
    (DPTA•FeSO4) +
    ZnSO4 0.375 μg/ml
    M +2-ethyl-3-hydroxy-4- 50 μM 94 218
    pyrone (ethyl maltol) +
    ZnSO4 0.188 μg/ml
    N +2-ethyl-3-hydroxy-4- 50 μM 482 624
    pyrone (ethyl maltol) +
    ZnSO4 0.750 μg/ml
    O +2-ethyl-3-hydroxy-4- 50 μM Discontinued at p1 of adaptation to
    pyrone (ethyl maltol) + media due to v. poor growth
    ZnSO4 0.375 μg/ml
    +CaCl2 0.1 mg/ml
    P +2-ethyl-3-hydroxy-4- 25 μM 220 282
    pyrone (ethyl maltol) +
    insulin 10 μg/ml
    Q +2-ethyl-3-hydroxy-4- 25 μM 142 484
    pyrone (ethyl maltol) +
    ZnSO4 0.375 μg/ml
    Stock in 293SFM II 0 0
    (Catalogue)
    D-MEM +10% FBS Out of range, too Out of range,
    high* too high*

    *Note: there were only about 1 × 106 viable cells/2-wells in the wells for cells grown in D-MEM +10% FBS vs. 2 × 106 viable cells/2-wells for all other samples.
  • Ethyl maltol did not support growth at 100 μM. Growth in 50 μM was a little better than 25 μM but better for transfection in the presence of insulin (ID# B, C, H, P and Q) and about the same in presence of ZnSO4.
  • 2-hydroxypyridine did not support growth at 50 μM. Transfection was better when this chelate was present in combination with ZnSO4 instead of insulin (see ID# D, E and J).
  • DPTA.FeSO4 supported growth better in the presence of insulin than in the presence of ZnSO4, however, growth was acceptable in the presence of ZnSO4. Transfection results were much better when the chelate was present in combination with ZnSO4 instead of insulin (see ID# F and L).
  • EXAMPLE 7 Effect of Zinc Sulfate Concentration on Transfection Efficiency of Attached Cells
  • The transfection efficiency of cells grown in the presence of ZnSO4+ chelating agent as a replacement for insulin was tested at various concentrations of chelator and ZnSO4 and the results are shown in Table 10. Cells were prepared in monolayer culture in base medium containing the indicated supplements and transfected and assayed as described above.
    TABLE 10
    X-gal stain %
    Compound added to Concentration β-gal expression transfection
    ID base media added (ng/˜2 × 106 cells) efficiency
    A +Human Transferrin 5 μg/ml 1495 ˜50-60
    (as control) +
    Insulin 10 μg/ml
    B +2-ethyl-3-hydroxy-4- 50 μM 1106 ˜50-60
    pyrone (ethyl maltol) +
    insulin 10 μg/ml
    C +2-ethyl-3-hydroxy-4- 50 μM 1157 ˜40
    pyrone (ethyl maltol) +
    ZnSO4 (1x) 0.375 μg/ml
    D +2-ethyl-3-hydroxy-4- 50 μM 570 N/A
    pyrone (ethyl maltol) +
    ZnSO4 (2x) 0.750 μg/ml
    E +2-ethyl-3-hydroxy-4- 50 μM 210 N/A
    pyrone (ethyl maltol) +
    ZnSO4 (5x) 1.875 μg/ml
    F +2-hydroxypyridine- 25 μM 1089 N/A
    N-oxide +
    insulin 10 μg/ml
    G +2-hydroxypyridine- 25 μM Discontinued at p3 of adaptation
    N-oxide + due to poor growth
    ZnSO4 (0.5x) 0.188 μg/ml
    H +2-hydroxypyridine- 25 μM 1531 ˜85-95
    N-oxide +
    ZnSO4 (1x) 0.375 μg/ml
    I +2-hydroxypyridine- 25 μM 1540 N/A
    N-oxide +
    ZnSO4 (2x) 0.750 μg/ml
    J +2-hydroxypyridine- 25 μM 1474 N/A
    N-oxide +
    ZnSO4 (5x) 1.875 μg/ml
    K +2-hydroxypyridine- 10 μM 1285 N/A
    N-oxide +
    ZnSO4 (1x) 0.375 μg/ml
    L +Diethylene-triamine- 50 μM Discontinued at p3 of adaptation
    penta acetic due to poor growth
    acid•Ferrous sulfate +
    insulin 10 μg/ml
    M +Diethylene-triamine- 50 μM Discontinued at p3 of adaptation
    penta acetic due to poor growth
    acid•Ferrous sulfate +
    ZnSO4 (0.5x) 0.188 μg/ml
    N +Diethylene-triamine- 50 μM 836 ˜50
    penta acetic
    acid•Ferrous sulfate +
    ZnSO4 (1x) 0.375 μg/ml
    O +Diethylene-triamine- 50 μM 710 N/A
    penta acetic
    acid•Ferrous sulfate +
    ZnSO4 (2x) 0.750 μg/ml
    P +Diethylene-triamine- 50 μM 467 N/A
    penta acetic
    acid•Ferrous sulfate +
    ZnSO4 (5x) 1.875 μg/ml
    Q +Human Transferrin + 5 μg/ml 766 N/A
    ZnSO4 (1x) 0.375 μg/ml
    Cat. Catalog 293 SFM II 0 N/A
    D- D-MEM +10% FBS + Out of range (too ˜85-95
    Mem 0.1 mM NEAA high >5000
  • Ethyl Maltol (samples B-E) supported growth at all concentrations of ZnSO4 tested (1×=0.188 μg/ml, 2×=0.375 μg/ml and 5×=1.875 μg/ml); however, expression decreased with increase in ZnSO4 concentration (see C, D and E) with 1× ZnSO4 being the best. X-gal staining showed a transfection efficiency of ˜40% in media with 1× ZnSO4 and of ˜50-60% in media with insulin.
  • 2-hydroxypyridine at a concentration of 25 μM (samples F-J) supported growth at concentrations of 1×, 2× and 5× ZnSO4, but not at 0.5×. Expression was about the same at 1×, 2× and 5× ZnSO4 (see H, I, and J). X-gal staining was only done on sample H (1× ZnSO4) and showed a transfection efficiency at 85-95%.
  • 2-hydroxypyridine at a concentration of 10 μM (sample K) supported growth in combination with 1× ZnSO4. Expression was slightly lower than when 2-hydroxypyridine is at 25 μM (compare H to K).
  • DPTA.FeSO4 supported growth at concentrations of 1×, 2× and 5× ZnSO4, but not at 0.5×. Expression decreased with increasing concentrations of ZnSO4, with 1× being the best. X-gal staining showed a transfection efficiency of 50% (ID# N).
  • Cells in D-MEM+10% FBS+0.1 mM NEAA had very high expression which was out of the range of the standard curve and estimated at >5000 ng/˜2×106 cells. X-gal staining showed transfection efficiency to be at about 85-95%.
  • Cells in all test media were very clumped both at 5 hrs post transfection (right before feeding) and at 24 hrs (prior to collection). Cells in catalog control were not clumped and uniformly covered the bottom of the well. Also, the amount of cells appeared to be too many for the well resulting in cells sitting on top of each other in several layers.
  • EXAMPLE 8 Effect of Varying Zinc Sulfate Concentration in the Presence of 2-Hydroxypyridine-N-Oxide
  • 293 cells were grown in monolayer culture in base medium in the presence of the indicated supplements. The cells were transfected and assayed as above. The results are shown in Table 11.
    TABLE 11
    Concentration β-gal expression
    ID Compound added to base media added (ng/˜2 × 106 cells)
    H +2-hydroxypyridine-N-oxide + 25 μM 1671
    ZnSO4 (1x) 0.375 μg/ml
    I +2-hydroxypyridine-N-oxide + 25 μM 1844
    ZnSO4 (2x) 0.750 μg/ml
    J +2-hydroxypyridine-N-oxide + 25 μM 2933
    ZnSO4 (5x) 1.875 μg/ml
    K +2-hydroxypyridine-N-oxide + 10 μM 2258
    ZnSO4 (1x) 0.375 μg/ml
    8E5
    8 × 105 cells/well in media H above  1607*
    6E5 6 × 105 cells/well in media H above  1290*
    4E5 4 × 105 cells/well in media H above  462*
    2E5 2 × 105 cells/well in media H above   0*
    R-H Same as H above, but on rotator  1138*
    R- Same as 6E5 above but on rotator  1143*
    6E5

    *Values are the total nanograms of β-galactosidase produced by two wells, each well set up at the concentration of cells indicated. For example, for 8E5, the number is the nanograms of β-galactosidase produced by two wells at 8 × 105 cells/well, i.e., 1607 ng/˜1.6 × 106 cells.
  • Continuous rotation of cells during the transfection did not improve transfection results.
  • Increasing concentrations of ZnSO4 seemed to increase expression. This is in contrast to the results present in the preceding example where additional ZnSO4 did not seem to affect expression levels.
  • Lower concentration of 2-hydroxypyridine (10 μM) increases expression (compare H to K). Again, this was not seen in the last experiment were K was actually a little lower than H.
  • EXAMPLE 9 Comparison of Transfection Efficiencies for Nucleic Acids Complexed In D-PBS and Media of the Present Invention
  • 293 cells were grown in monloayer culture in base media supplemented with the indicated chelator and metal salt at the indicated concentrations. The concentration of the chelator is given in μmoles/l and the concentration of the metal salt is given in μgrams/ml. For comparison, a 10 ml flask of 293 cells in suspension culture in test media supplemented with +2-hydroxypyridine-N-oxide at 25 μM and ZnSO4 at 0.375 μg/ml was transfected. The concentration of the cells in the media was 1×106 cell/ml, the concentration of DNA used was 1 μg DNA/106 cells, the concentration of LIPOFECTAMINE 2000 used was 8 μl/106 cells. The results are shown in Table 12.
    TABLE 12
    Exp. B Exp. C
    Exp. A complex in complex
    complex & D-PBS & &resuspend
    Compound added to Concentration resuspend in resuspend in test
    ID base media added test media in test media media
    A +2-hydroxypyridine- 25 μM 913 159
    N-oxide
    +ZnSO4 (1x) 0.375 μg/ml
    B +2-hydroxypyridine- 25 μM 732 221
    N-oxide
    +ZnSO4 (2x) 0.750 μg/ml
    C +2-hydroxypyridine- 25 μM 1054 262
    N-oxide
    +ZnSO4 (5x) 1.875 μg/ml
    D +2-hydroxypyridine- 10 μM 738 123
    N-oxide
    +ZnSO4 (1x) 0.375 μg/ml
    E +2-hydroxypyridine- 10 μM 569 92
    N-oxide
    +ZnSO4 (2x) 0.750 μg/ml
    F +2-hydroxypyridine- 10 μM 624 78
    N-oxide
    +ZnSO4 (5x) 1.875 μg/ml
    G +2-hydroxypyridine- 5 μM 536 291
    N-oxide
    +ZnSO4 (1x) 0.375 μg/ml
    Flask +2-hydroxypyridine- 25 μM 29118
    N-oxide
    +ZnSO4 (1x) 0.375 μg/ml
  • In situ staining for β-galactosidase was done and showed that D-PBS should not be used for complexing, as efficiencies were less than or equal to 20%. Transformation efficiencies of the samples transfected with DNA complexed in test media were in the range of 50-85% transfected. The best efficiency seen so far was in the cells from the shaker flask at 98-100% transfection efficiency.
  • β-galactosidase expression was lower overall than in previous experiments but the trend seems similar. Again the data show that D-PBS should not be used for complexing in these experiments. The β-galactosidase expression in the cells in suspension culture in shaker flasks was substantially higher than that seen in monolayer culture. It should be emphasized that the flask was put on a shaker platform (125 rpm) from the time of transfection to the time of collection 24 hrs later. The improved transfection and resulting expression can be due to the cells being in better shape in the shaker flask than in the 24-well plates over the incubation period.
  • EXAMPLE 10 Effects of Varying Zinc Sulfate and 2-hydroxypyridine-N-oxide Concentrations on Transfection Efficiencies of Cells in Suspension
  • A comparison of transfection results obtained with 293 cells in suspension culture versus monolayer culture was performed. Cells in test media A were transfected in a 15 ml volume and cells in test media B-F were transfected in 10 ml volume. All cell densities were 1×106 viable cells per ml at transfection set-up. DNA and lipid concentrations used were 1 μg DNA and 8 μl LIPOFECTAMINE 2000 per 1×106 viable cells. Duplicate samples of cells in test media A were transfected and the results from both replicates are shown in table 13.
    TABLE 13
    β-gal X-gal stain
    Compound added Concentration expression (ng/ % transfection
    ID to base media added ˜2 × 106 cells) efficiency
    A +2- 25 μM 17218 96
    hydroxypyridine-
    N-oxide +
    ZnSO4 (1x) 0.375 μg/ml 15283 98
    B +2- 25 μM 29335 99
    hydroxypyridine-
    N-oxide +
    ZnSO4 (2x) 0.750 μg/ml
    C +2- 25 μM 28972 99
    hydroxypyridine-
    N-oxide +
    ZnSO4 (5x) 1.875 μg/ml
    D +2- 10 μM 25018 99
    hydroxypyridine-
    N-oxide +
    ZnSO4 (1x) 0.375 μg/ml
    E +2- 10 μM 23319 98
    hydroxypyridine-
    N-oxide +
    ZnSO4 (2x) 0.750 μg/ml
    F +2- 10 μM 25991 95
    hydroxypyridine-
    N-oxide +
    ZnSO4 (5x) 1.875 μg/ml
  • For cells transfected in 24-well plates, cells densities used were 1×106 viable cells per well. DNA and lipid concentrations used were 1 μg DNA and 8 μl LIPOFECTAMINE 2000 per 1×106 viable cells and the results are shown in Table 14.
    TABLE 14
    β-gal
    expression X-gal stain
    Compound added to Concentration (ng/˜2 × 106 % transfection
    ID base media added cells) efficiency
    A +2-hydroxypyridine- 25 μM 1374 10
    N-oxide +
    ZnSO4 (1x) 0.375 μg/ml
    B +2-hydroxypyridine- 25 μM 1240 20
    N-oxide +
    ZnSO4 (2x) 0.750 μg/ml
    C +2-hydroxypyridine- 25 μM 1357 60
    N-oxide +
    ZnSO4 (5x) 1.875 μg/ml
  • Transfection efficiencies and β-galactosidase expression levels were much better when transfections were done in shaker flasks at 125 rpm rather than in stationary plates.
  • Expression in media A was lower than in the previous experiments where expression was 29118 ng/˜2×106 cells. The volume used in that experiment was 10 ml in a 125 ml shaker flask. Growth data for the adaptation of the cells to the test media shows that cells densities are similar for all media A-F. X-gal staining of cells shows similar transfection efficiencies.
  • EXAMPLE 11 Comparison Of The Growth Of Cells In Transfection Media
  • Frozen aliquots of 293-F cells and 293-H cells were thawed and suspended in 293 SFM II (Invitrogen Corporation, Carlsbad, Calif.) for 1 passage before being counted and suspended at a concentration 3×105 viable cells/ml in test media.
  • The cells were suspended in 15 ml of test media in a 125 ml shaker flask. The cells were shaken at 125 rpm on a rotary shaker in an atmosphere of 8% CO2/92% air. The compositions of media A, B and C were as shown in Table 14; catalog media was 293 SFM II (Invitrogen Corporation, Carlsbad, Calif.). Cells were counted every four days and split to 3×105 viable cells/ml at that time. FIGS. 1A and 1B show the cell density at each of the first 3 passages in the test media. FIG. 1A shows the cell densities of the 293-F cells while FIG. 1B shows the densities of the 293-H cells
  • The 293-F cells adapted well into the test media. There was some clumping but vortexing was enough to get a single cell suspension.
  • The 293-H cells grew to similar densities in the test media as in the 293SFM II. However, there was a lot of clumping that could not be resolved by vortexing.
  • In FIGS. 2A and 2B, a growth curve comparing the growth of 293-F&H cells in a single passage is shown. 25 ml cultures in 125 ml flasks were seeded with 3×105 viable cells/ml in triplicate and 1 ml aliquots were taken each day and the viable cells counted. Catalog media was 293 SFM TI supplemented with 4 mM glutamine and test media A, B and C were as in Table 14. FIG. 2A shows the results obtained with 293-F cells and FIG. 2B shows the results obtained with 293-H cells. In all cases, the test media supported cell growth more effectively than the catalog media.
  • EXAMPLE 12 Effects of Sulfate Ion and Zinc Ion on Growth of Cells
  • Cells were seeded at a density of 3×105 cells/ml and counted after 4 days of growth in media containing in varying amounts of ZnSO4, ZnCl2 or Na2SO4 in order to determine which ion Zn or SO4 2− in zinc sulfate promotes cell growth. Test media supplemented with 25 μm 2-hydroxypyridine-N-oxide and the indicated salts were used to assess the effects of zinc ion and sulfate ion in suspension culture. As shown in FIG. 3, ZnCl2 could effectively substitute for zinc sulfate whereas Na2SO4 resulted in less cell growth.
  • EXAMPLE 13 Evaluation of Flask Volume and Conditioned Media in Transfection Efficiency
  • 293 cells in suspension were grown in the indicated amount of media in 125 ml shake flasks. Results below are averages of duplicate flasks (A-25 m1 is for single flask only). The cell densities (at time of set-up) in all cases was 1×106 viable cells per ml. The DNA and lipid concentrations used were 1 μg and 8 μg1 respectively per 1×106 viable cells.
  • In addition, the effects of conditioned media on transformation were assessed. The results are presented in Table 15.
    TABLE 15
    β-gal expression X-gal stain
    Sample (ng/˜1 × 106 cells) % transfection efficiency
    A - 10 ml 9250 90-95
    A - 15 ml 8037 90-95
    A - 20 ml 4605 80
    A - 25 ml 20287 99
    B - 15 ml 4224 75
    B - 15 ml in conditioned 8902 85-90
    media
    B - 15 ml (used LTI Cat. 5899 75
    DNA)
    C - 15 ml 6965 90
    C - 15 ml in conditioned 7312 95
    media

    Note:

    β-galactosidase production in this experiment is expressed as ng/˜1 × 106 cells instead of ng/˜2 × 106 cells as in previous experiments.
  • Flasks set-up in media A were for testing effect of cell suspension volume in the flasks and its effects on transfections. There appears to be no significant difference between a 10 ml and 15 ml volume. However the decline when at 20 ml and then increase at 25 m1 is not explained. Also note that 25 ml was not set-up in duplicate.
  • For samples set-up in B and C the aim was to compare transfectability in fresh media vs. conditioned media. Conditioned media consisted of the media the culture had been growing in until day of transfection set-up. This was done to evaluate whether transfections can be carried out without having to change the culture medium (i.e. spin cells down, re-suspend, etc.). The results show more β-galactosidase produced when in conditioned media for B and comparable results in media C thus supporting the possibility of just adding the lipid-DNA complex to culture when a certain cell density has been reached.
  • Comparison of the 15 ml samples for A, B and C show no significant difference between A and B and somewhat lower results for C as for as β-gal quantification and similar results for x-gal staining.
  • EXAMPLE 14 Effect of Culture Volume on Transfection Efficiency
  • In order to determine whether the volume of the culture medium affects the efficiency of transfection for a given number of cells, two protocols were tested. The first protocol required the cells to be suspended in a reduced volume of medium and contacted with a nucleic acid and transfection reagent. After an incubation period to allow uptake of the nucleic acid, additional medium was added to bring the culture volume up to a final volume.
  • The second protocol called for the introduction of a nucleic acid and transfection reagent into the culture at the final volume. Since the second protocol requires one less medium addition step, it requires less manipulation of the cells and less hands-on time.
  • For both protocols, 3×107 cells in a final volume of 30 ml medium was used. Nucleic acid:transfection reagent complexes were prepared as follows: 30 μg DNA was added to 1 ml of minimal medium (OPTIMEM, Invitrogen Corporation, Carlsbad, Calif.) 200 μl transfection reagent (LIPOFECTAMINE 2000, Invitrogen Corporation, Carlsbad, Calif.) was added to 1 ml minimal medium. The diluted DNA and transfection reagent were combined, mixed gently and incubated at room temperature for 20 minutes. The complexes were added to the cells and mixed. Cells were incubated at 37° C. on a shaking platform (125 rpm) in 8% CO2/92% air for a total of 48 hours. Aliquots were taken at 24 and 48 hours and assayed for β-galactosidase activity, cell concentration, cell viability and protein content. For protocol 1, cells were initially suspended in 10 ml medium and DNA complexes were added. The cells were incubated 4 hours and then an additional 18 ml medium were added. For protocol 2, cells were suspended in 28 ml medium and DNA complexes were added. The results are presented in Table 16.
    TABLE 16
    PROTOCOL 1 PROTOCOL 2
    24 hours
    mg protein/ 0.8 0.9
    106 cells
    μg β-gal/ 22.7 25.0
    106 cells
    μg β-gal/ 28.7 28.7
    mg protein
    expected mg 22.7 25
    β-gal/L cells
    48 hours
    mg protein/ 1.1 1.1
    106 cells
    μg β-gal/ 50.1 52.9
    106 cells
    μg β-gal/ 46.4 46.8
    mg protein
    expected mg 50.1 52.9
    β-gal/L cells
    mg β-gal/ 1.5 1.6
    30 ml culture
  • As seen in Table 16, initially culturing the cells in a reduced volume in order to facilitate uptake of nucleic acid by the cells did not increase the overall yield of β-galactosidase.
  • EXAMPLE 15 Effect of Transfection Reagent Concentration on Transfection Efficiency
  • Since no advantage was seen in incubating the nucleic acid complexes with cells in a reduced volume, protocol 2 was used in the following experiment. In order to assess the effects of varying the amount of transfection reagent transformations were conducted using 100 μl, 150 μl and 200 μl of LIPOFECTAMINE 2000. The indicated amount of transfection reagent was diluted in 1 ml minimal medium and 30 μg DNA was diluted in 1 ml medium as described above. The dilutions were combined and mixed gently and incubated 20 minutes at room temperature. The nucleic acid complexes were added to 3×107 cells in 28 ml of medium. Aliquots were taken at 25 hours and 49 hours and assayed above. The results are presented in Table 17.
    TABLE 17
    24 HOURS 48 HOURS
    live live
    cells/ml mg β-gal/ cells/ml mg β-gal/
    % viable 30 ml culture % viable 30 ml culture
    untransfected 2.3 × 106 0 2.4 × 106 0
    control 84% 69%
    100 μl 1.2 × 106 0.6 1.7 × 106 1.2
    LIPOFECTAMINE 79% 69%
    2000
    150 μl 1.1 × 106 0.5 1.6 × 106 1.3
    LIPOFECTAMINE 74% 65%
    2000
    150 μl 1.7 × 106 1.4 1.3 × 106 2.7
    LIPOFECTAMINE 82% 61%
    2000
    + butyrate
    200 μl 1.2 × 106 0.6 1.4 × 106 1.3
    LIPOFECTAMINE 70% 62%
    2000
  • The amount of transfection reagent did not seem to dramatically affect the expression levels of β-galactosidase (compare 1.2 mg/30 ml of culture at 100 μl transfection reagent to 1.3 mg/30 ml of culture at 200 μl of transfection reagent). In contrast, the presence of butyrate in the culture medium more than doubled the production of β-galactosidase from 1.3 mg/30 ml of culture to 2.7 mg/30 ml of culture.
  • EXAMPLE 16
  • The effect of PL-1, PL-2 and PL000458 media on cell growth and viability for 293 cells was determined. 293-F cells were seeded at 3×105 cell/ml initial density (in 30 ml cultures) in shaker flasks (8% CO2, 37° C., 125 rpm). Viability and cell density were measured approximately every 24 hours. The results are shown in FIGS. 4A-D.
  • Having now fully described the present invention in some detail by way of illustration and example for purposes of clarity of understanding, it will be obvious to one of ordinary skill in the art that the same can be performed by modifying or changing the invention within a wide and range of conditions, formulations and other parameters without affecting the scope of the invention or any specific embodiment thereof, and that such modifications or changes are intended to be encompassed within the scope of the appended claims.
  • All publications, patents and patent applications mentioned in this specification are indicative of the level of skill of those skilled in the art to which this invention pertains, and are herein incorporated by reference to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference.

Claims (20)

1. A cell culture medium, wherein said medium supports introduction of one or more macromolecules into at least one eukaryotic cell in culture and supports cultivation of said at least one cell subsequent to said introduction,
wherein it is not necessary to remove medium used during said introduction from the presence of said at least one cell to support growth of said at least one cell, and/or
wherein after said introduction growth can be accomplished in cultivation in a volume of medium that is about the same volume up to no more than about 10 times the volume of the medium in which said introduction occurred.
2. The cell culture medium of claim 1, wherein said medium also can support cultivation of said at least one cell prior to said introduction.
3. The cell culture medium of claim 1, wherein replenishing, replacing or supplementing said medium with fresh medium after said introduction does not improve said cultivation, relative to cultivation in said medium without said replenishing, replacing or supplementing.
4. The cell culture medium of claim 1, wherein said culture is a suspension culture.
5. The cell culture medium of claim 1, wherein said one or more macromolecules includes at least one nucleic acid molecule.
6. The cell culture medium of claim 1, wherein said cultivation includes the production of at least one protein product.
7. The cell culture medium of claim 1, said medium comprising at least one compound selected from the group consisting of a polyol, a hydroxypyridine derivative, 1,3,5-N,N′,N″-tris(2,3-dihydroxybenzoyl)aminomethylbenzene, ethylenedi-amine-N,N′-tetramethylenephosphonic acid, trisuccin, an acidic saccharide (e.g. ferrous gluconate), a glycosaminoglycan, diethylenetriaminepentaacetic acid, nitrilotriacetic acid mono-, bis-, or tris-substituted 2,2′-bipyridine, a hydroxamate derivative (e.g. acetohydroxamic acid), an amino acid, deferoxamine, ferrioxamine, iron basic porphine, porphyrin and derivatives thereof, DOTA-lysine, a texaphyrin, a sapphyrin, a polyaminocarboxylic acid, an α-hydroxycarboxylic acid, a polyethylenecarbamate, picolinic acid, 4-pyridoxic acid, 3-hydroxy-2-pyridinemaltol, ethyl maltol, Ustilago ferrichrome, nicotinic acid-N-oxide, IRC011, 2-hydroxypyridine-N-oxide, 3-hydroxy-4-pyrone, 3-hydroxypypyrid-2-one, 3-hydroxypyrid-2-one, 3-hydroxypyrid-4-one, 1-hydroxypyrid-2-one, 1,2-dimethyl-3-hydroxypyrid-4-one, 1-methyl-3-hydroxypyrid-2-one, 3-hydroxy-2(1H)-pyridinone, pyridoxal isonicotinyl hydrazone, nicotinic acid-N-oxide, and 2-hydroxy-nicotinic acid.
8. The cell culture medium of claim 1, wherein said eukaryotic cell is selected from the group consisting of a 293 cell, a PER-C6 cell, a CHO cell, a COS cell, a BHK cell, a HeLa cell, and a Sp2/0 cell.
9. The cell culture medium of claim 1, wherein said medium substantially lacks or does not contain transferrin.
10. The cell culture medium of claim 1, wherein said medium substantially lacks or does not contain insulin.
11. The cell culture medium of claim 1, wherein said medium substantially lacks or does not contain any polyanionic compounds.
12. The cell culture medium of claim 1, wherein said medium substantially lacks or does not contain dextran sulfate.
13. The medium of claim 1, wherein a cationic lipid is admixed to said medium for said introduction.
14. A method of making a medium comprising admixing water and at least one ingredient selected from the group consisting of an amino acid, a sugar, a fatty acid, a vitamin, a pH buffer, a surfactant, a trace metal or salt or hydrate thereof, an amine compound, a growth factor, an agent to control osmolarity and/or ionic strength and/or maintain membrane potential, a flavin, a compound that participates in or is a product of the glycolytic pathway, an alcoholamine, a cyclic alcohol, a phospholipid or portion thereof, a salt of selenious acid, a divalent or trivalent cation, a pteridine derivative, a valeric acid, and a coenzyme,
wherein said medium supports introduction of one or more macromolecules into at least one eukaryotic cell in culture and supports cultivation of said at least one cell subsequent to said introduction,
wherein it is not necessary to remove medium used during said introduction from the presence of said at least one cell to support growth of said at least one cell, and/or
wherein after said introduction growth is accomplished in cultivation in a volume of medium that is about the same volume up to no more than about 10 times the volume of the medium in which said introduction occurred.
15. A method of cultivating eukaryotic cells comprising:
(a) contacting said cells with the cell culture medium of claim 1; and
(b) maintaining said cells under conditions suitable to support cultivation of said cells in culture.
16. A method for introducing one or more macromolecules into at least one eukaryotic cell in culture, said method comprising:
(a) culturing at least one eukaryotic cell in said medium of claim 1 in culture;
(b) introducing at least one macromolecule into said culture under conditions sufficient to cause one or more of said at least one macromolecule to be introduced in said at least one cell; and
(c) cultivating said at least one cell in said medium to produce a product whose production is controlled by said at least one molecule,
wherein growth of said at least one cell continues in said medium in the absence of said medium being with fresh medium, wherein it is not necessary to remove medium used during said introducing from the presence of said at least one cell to support growth of said at least one cell, and/or
wherein after said introducing growth is accomplished in cultivation in a volume of medium that is about the same volume up to no more than about 10 times the volume of the medium in which said introducing occurred.
17. The method of claim 16, wherein said medium is not during said production for at least twelve hours subsequent to said introducing.
18. The method of claim 16, wherein said medium is not during said production for at least twenty-four hours subsequent to said introducing.
19. A kit for the cultivation and transfection of cells in vitro, said kit comprising the cell culture medium of claim 1.
20. A composition comprising the cell culture medium of claim 1 and at least one component selected from the group consisting of at least one eukaryotic cell, one or more agents for the introduction of at least one nucleic acid molecule into at least one cell, and one or more nucleic acid molecules.
US11/552,783 2001-03-27 2006-10-25 Culture medium for cell growth and transfection Abandoned US20070254358A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/552,783 US20070254358A1 (en) 2001-03-27 2006-10-25 Culture medium for cell growth and transfection
US13/968,157 US9879243B2 (en) 2001-03-27 2013-08-15 Culture medium for cell growth and transfection

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US27875401P 2001-03-27 2001-03-27
US10/105,937 US20030096414A1 (en) 2001-03-27 2002-03-26 Culture medium for cell growth and transfection
US11/552,783 US20070254358A1 (en) 2001-03-27 2006-10-25 Culture medium for cell growth and transfection

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/105,937 Continuation US20030096414A1 (en) 2001-03-27 2002-03-26 Culture medium for cell growth and transfection

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/968,157 Continuation US9879243B2 (en) 2001-03-27 2013-08-15 Culture medium for cell growth and transfection

Publications (1)

Publication Number Publication Date
US20070254358A1 true US20070254358A1 (en) 2007-11-01

Family

ID=23066207

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/105,937 Abandoned US20030096414A1 (en) 2001-03-27 2002-03-26 Culture medium for cell growth and transfection
US11/552,783 Abandoned US20070254358A1 (en) 2001-03-27 2006-10-25 Culture medium for cell growth and transfection
US13/968,157 Expired - Lifetime US9879243B2 (en) 2001-03-27 2013-08-15 Culture medium for cell growth and transfection

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/105,937 Abandoned US20030096414A1 (en) 2001-03-27 2002-03-26 Culture medium for cell growth and transfection

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/968,157 Expired - Lifetime US9879243B2 (en) 2001-03-27 2013-08-15 Culture medium for cell growth and transfection

Country Status (5)

Country Link
US (3) US20030096414A1 (en)
EP (2) EP2351827A1 (en)
JP (1) JP2005506048A (en)
CA (1) CA2441910A1 (en)
WO (1) WO2002077202A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009140701A2 (en) * 2008-05-16 2009-11-19 Synthetic Genomics, Inc. Delivery into cells using ultra-short pulse lasers
US20090311236A1 (en) * 2008-06-11 2009-12-17 Immune @Work, Inc. Therapeutic Peptide Compositions And Methods Of Making And Using Same
WO2010031074A2 (en) 2008-09-15 2010-03-18 Genentech, Inc. Compositions and methods for regulating cell osmolarity
WO2014110433A1 (en) * 2013-01-10 2014-07-17 Biogen Idec Ma Inc. Medium supplements for improved process performance
DE202011110859U1 (en) 2010-04-23 2016-12-20 Life Technologies Corporation Small peptides comprising cell culture medium
US9879243B2 (en) 2001-03-27 2018-01-30 Lifetechnologies Corporation Culture medium for cell growth and transfection
US10066000B2 (en) 2012-05-02 2018-09-04 Life Technologies Corporation High yield transient expression in mammalian cells using unique pairing of high density growth and transfection medium and expression enhancers
WO2019099891A1 (en) 2017-11-16 2019-05-23 Life Technologies Corporation Streamlined methods for making liquid media

Families Citing this family (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6767741B1 (en) * 1999-08-27 2004-07-27 Invitrogen Corporation Metal binding compounds and their use in cell culture medium compositions
US20040133938A1 (en) * 2002-11-19 2004-07-08 Yinghui Dan Use of lipoic acid in plant culture media
US7217566B2 (en) * 2003-03-24 2007-05-15 Invitrogen Corporation Attached cell lines
EP1698690B1 (en) * 2003-12-26 2010-04-28 Makoto Asashima Basal medium for es cell culturing
US7691607B2 (en) * 2004-02-09 2010-04-06 The Regents Of The University Of California Expression system of NELL peptide
ME01218B (en) 2004-03-01 2013-06-20 Ares Trading Sa Use of a serum-free cell culture medium for the production of il-18bp in mammalian cells
US20050287666A1 (en) * 2004-06-29 2005-12-29 Invitrogen Corporation Cell culture medium comprising transition metals or trace elements
UA92157C2 (en) 2004-11-02 2010-10-11 Арес Трейдинг С.А. Process for production of growth hormone
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
EP1917276B1 (en) 2005-08-26 2018-03-21 Ares Trading S.A. Process for the preparation of glycosylated interferon beta
TW200800235A (en) * 2005-10-18 2008-01-01 Otsuka Pharma Co Ltd Carrier composition for nucleic acid transport
PL3121266T3 (en) 2006-01-04 2020-07-13 Baxalta Incorporated Oligopeptide-free cell culture media
US8153429B2 (en) * 2006-02-23 2012-04-10 Viacyte, Inc. Compositions and methods useful for culturing differentiable cells
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
KR20140132017A (en) 2006-09-13 2014-11-14 애브비 인코포레이티드 Cell culture improvements
GB0720486D0 (en) * 2007-10-19 2007-11-28 Univ Edinburgh Cationic lipids
BRPI0920027A2 (en) * 2008-10-20 2015-10-06 Abbott Lab isolation and purification of antibodies using protein affinity chromatography
WO2010048192A2 (en) 2008-10-20 2010-04-29 Abbott Laboratories Viral inactivation during purification of antibodies
WO2011018472A2 (en) 2009-08-14 2011-02-17 Basf Se Methods in cell cultures, and related inventions, employing certain additives
KR101107022B1 (en) * 2009-09-11 2012-01-25 한림대학교 산학협력단 Method for preparing cryopreserved acellular dermal matrix and cryopreserved acellular dermal matrix prepared therefrom
KR101089614B1 (en) * 2010-02-26 2011-12-05 (주)시지바이오 Method for preparing acellular dermal matrix and acellular dermal matrix prepared therefrom
RU2563353C2 (en) 2010-04-26 2015-09-20 Новартис Аг Improved cell culture medium
CA2807607A1 (en) * 2010-08-20 2012-02-23 Wyeth Llc Cell culture of growth factor-free adapted cells
WO2012149197A2 (en) 2011-04-27 2012-11-01 Abbott Laboratories Methods for controlling the galactosylation profile of recombinantly-expressed proteins
CA2856455C (en) 2011-11-24 2022-08-23 Genethon Scalable lentiviral vector production system compatible with industrial pharmaceutical applications
KR101508323B1 (en) * 2012-02-28 2015-04-14 건국대학교 산학협력단 Fluid-based culture medium
WO2013158279A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Protein purification methods to reduce acidic species
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
US9181572B2 (en) 2012-04-20 2015-11-10 Abbvie, Inc. Methods to modulate lysine variant distribution
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
BR112015004467A2 (en) 2012-09-02 2017-03-21 Abbvie Inc method for controlling protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
EP2830651A4 (en) 2013-03-12 2015-09-02 Abbvie Inc Human antibodies that bind human tnf-alpha and methods of preparing the same
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
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
WO2014159579A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. MUTATED ANTI-TNFα ANTIBODIES AND METHODS OF THEIR USE
EP3052640A2 (en) 2013-10-04 2016-08-10 AbbVie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
WO2015138032A2 (en) 2013-12-20 2015-09-17 President And Fellows Of Harvard College Organomimetic devices and methods of use and manufacturing thereof
CN106715707A (en) 2014-06-18 2017-05-24 免疫医疗公司 Cell culture methods and media comprising n-acetylcysteine
CN106119186B (en) * 2016-06-24 2019-10-08 肇庆大华农生物药品有限公司 It is a kind of for the serum free medium and preparation method thereof for cultivating mdck cell that suspends entirely
JP7051820B2 (en) * 2016-09-12 2022-04-11 アレス トレーディング ソシエテ アノニム Methods for modifying the production profile of recombinant proteins
US10961500B1 (en) 2019-04-23 2021-03-30 Regeneron Pharmaceuticals, Inc. Cell culture medium for eukaryotic cells
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
CN110438066B (en) * 2019-08-19 2021-01-12 杭州百凌生物科技有限公司 Mammalian cell line 293C18P capable of being stably passaged and capable of being cultured in suspension, and preparation method and application thereof
WO2021035060A1 (en) * 2019-08-21 2021-02-25 Brain Chemistry Labs Compositions comprising a metal and l-serine, and uses thereof
WO2022187725A1 (en) * 2021-03-05 2022-09-09 Sensorium Bio Inc. Cell culture system and methods of using
CN115161262A (en) * 2022-08-11 2022-10-11 无锡多宁生物科技有限公司 Novel application of dextran sulfate in basic culture medium for culturing CHO cell line cells
CN115109801B (en) * 2022-08-25 2022-11-22 深圳市先康达生命科学有限公司 Slow virus transfection transfer aid and application thereof

Citations (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2695314A (en) * 1951-08-25 1954-11-23 Monsanto Chemicals Hydroxyethylated-n-keryl alkylenediamines
US2901461A (en) * 1955-05-05 1959-08-25 Union Carbide Corp Curable glycidyl polyether-polyamine compositions
US3152188A (en) * 1960-11-15 1964-10-06 Nalco Chemical Co Nu-substituted polyalkylene polyamines having 2-hydroxyethyl and mono-secondary-hydroxyl hydrocarbon substituents
US4673649A (en) * 1983-07-15 1987-06-16 University Patents, Inc. Process and defined medium for growth of human epidermal keratinocyte cells
US4897355A (en) * 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US4940666A (en) * 1983-07-15 1990-07-10 University Patents, Inc. Process and defined medium for growth of human epidermal keratinocyte cells
US5045454A (en) * 1987-01-09 1991-09-03 Medi-Cult A/S Serum-free growth medium and use thereof
US5118513A (en) * 1987-07-02 1992-06-02 The Procter & Gamble Company Method for enhancing bioavailability of iron-calcium mineral supplements
US5135866A (en) * 1989-03-03 1992-08-04 W. R. Grace & Co.-Conn. Very low protein nutrient medium for cell culture
US5166066A (en) * 1991-07-11 1992-11-24 The Upjohn Company Transformed cells comprising GABAA receptors
US5171678A (en) * 1989-04-17 1992-12-15 Centre National De La Recherche Scientifique Lipopolyamines, their preparation and their use
US5208036A (en) * 1985-01-07 1993-05-04 Syntex (U.S.A.) Inc. N-(ω, (ω-1)-dialkyloxy)- and N-(ω, (ω-1)-dialkenyloxy)-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US5232848A (en) * 1987-03-24 1993-08-03 W. R. Grace & Co.-Conn. Basal nutrient medium for cell culture
US5279833A (en) * 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
US5283185A (en) * 1991-08-28 1994-02-01 University Of Tennessee Research Corporation Method for delivering nucleic acids into cells
US5334761A (en) * 1992-08-28 1994-08-02 Life Technologies, Inc. Cationic lipids
US5455335A (en) * 1991-12-13 1995-10-03 The Trustees Of Princeton University Pharmaceutical compositions and methods of transporting compounds across biological membrane
US5459127A (en) * 1990-04-19 1995-10-17 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5474931A (en) * 1991-06-17 1995-12-12 Life Technologies, Inc. Media concentrate technology
US5545412A (en) * 1985-01-07 1996-08-13 Syntex (U.S.A.) Inc. N-[1, (1-1)-dialkyloxy]-and N-[1, (1-1)-dialkenyloxy]-alk-1-yl-n,n,n-tetrasubstituted ammonium lipids and uses therefor
US5589466A (en) * 1989-03-21 1996-12-31 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5667774A (en) * 1992-08-20 1997-09-16 E.I. Du Pont De Nemours And Company Crosslinked polymeric ammonium salts
US5674908A (en) * 1993-12-20 1997-10-07 Life Technologies, Inc. Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US5705385A (en) * 1995-06-07 1998-01-06 Inex Pharmaceuticals Corporation Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US5713055A (en) * 1996-11-25 1998-01-27 Eastman Kodak Company Ambient light sensitive automatic flash control circuit
US5719131A (en) * 1994-12-09 1998-02-17 Genzyme Corporation Cationic amphiphiles containing dialkylamine lipophilic groups for intracellular delivery of therapeutic molecules
US5736392A (en) * 1995-06-07 1998-04-07 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US5744335A (en) * 1995-09-19 1998-04-28 Mirus Corporation Process of transfecting a cell with a polynucleotide mixed with an amphipathic compound and a DNA-binding protein
US5753613A (en) * 1994-09-30 1998-05-19 Inex Pharmaceuticals Corporation Compositions for the introduction of polyanionic materials into cells
US5783566A (en) * 1996-05-10 1998-07-21 California Institute Of Technology Method for increasing or decreasing transfection efficiency
US5783565A (en) * 1994-12-09 1998-07-21 Genzyme Corporation Cationic amphiphiles containing spermine or spermidine cationic group for intracellular delivery of therapeutic molecules
US5785992A (en) * 1994-09-30 1998-07-28 Inex Pharmaceuticals Corp. Compositions for the introduction of polyanionic materials into cells
US5793566A (en) * 1994-01-10 1998-08-11 Micropolis Corporation Self securing compliant gasket for a disk drive assembly housing
US5795587A (en) * 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5830430A (en) * 1995-02-21 1998-11-03 Imarx Pharmaceutical Corp. Cationic lipids and the use thereof
US5830878A (en) * 1995-06-07 1998-11-03 Megabios Corporation Cationic lipid: DNA complexes for gene targeting
US5837283A (en) * 1997-03-12 1998-11-17 The Regents Of The University Of California Cationic lipid compositions targeting angiogenic endothelial cells
US5908635A (en) * 1994-08-05 1999-06-01 The United States Of America As Represented By The Department Of Health And Human Services Method for the liposomal delivery of nucleic acids
US5935936A (en) * 1996-06-03 1999-08-10 Genzyme Corporation Compositions comprising cationic amphiphiles and co-lipids for intracellular delivery of therapeutic molecules
US5948925A (en) * 1997-05-06 1999-09-07 Genzyme Corporation Cationic amphiphiles containing linkers derived from neutral or positively charged amino acids
US5948767A (en) * 1994-12-09 1999-09-07 Genzyme Corporation Cationic amphiphile/DNA complexes
US5962533A (en) * 1996-02-06 1999-10-05 University Of Florida Research Foundation, Inc. Hydroxy polyamines
US6093564A (en) * 1997-10-03 2000-07-25 V.I. Technologies, Inc. Methods and compositions for the selective modification of nucleic acids
US6103529A (en) * 1996-10-10 2000-08-15 Life Technologies, Inc. Animal cell culture media comprising peptides derived from rice
US6171862B1 (en) * 1998-03-31 2001-01-09 The Regents Of The University Of California Transfection in serum-containing media
US6506604B2 (en) * 1993-06-11 2003-01-14 Cell Genesys, Inc. Method for production of high titer virus and high efficiency retroviral mediated transduction of mammalian cells
US20030032203A1 (en) * 2001-07-10 2003-02-13 Sabatini David M. Small molecule microarrays
US6541225B1 (en) * 2000-01-26 2003-04-01 Raven Biotechnologies, Inc. Methods and compositions for generating human monoclonal antibodies
US6544790B1 (en) * 1999-09-17 2003-04-08 Whitehead Institute For Biomedical Research Reverse transfection method
US20040038862A1 (en) * 2001-07-30 2004-02-26 Goodwin Bryan James Identification of new therapeutic targets for modulating bile acid synthesis
US6767741B1 (en) * 1999-08-27 2004-07-27 Invitrogen Corporation Metal binding compounds and their use in cell culture medium compositions
US20050100940A1 (en) * 1998-03-02 2005-05-12 Millennium Pharmaceuticals, Inc. Novel FDRG protein and nucleic acid molecules and uses therefor
US20050118253A1 (en) * 1998-02-03 2005-06-02 Protiva Biotherapeutics, Inc. Systemic delivery of serum stable plasmid lipid particles for cancer therapy
US7026454B1 (en) * 1998-02-17 2006-04-11 University Of Kansas Medical Center Neuroendocrine marker of prostate cancer and method for producing same
US20070025962A1 (en) * 1998-06-25 2007-02-01 Eaton Dan L Interleukin-8 homologous polypeptides and therapeutic uses thereof

Family Cites Families (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB901673A (en) 1958-05-20 1962-07-25 Nat Res Dev Cell culture
AU610843B2 (en) 1986-10-20 1991-05-30 Life Technologies, Inc. Serum-free medium for the proliferation of lymphokine-activated killer cells
US4933281A (en) 1987-03-17 1990-06-12 The University Of Iowa Research Foundation Method for producing rhamnose
JPH0249579A (en) 1988-02-29 1990-02-19 Kyowa Hakko Kogyo Co Ltd Culture medium
WO1993000423A1 (en) * 1991-06-21 1993-01-07 Novo Nordisk A/S Iron chelate culture medium additive
WO1994027435A1 (en) 1993-06-01 1994-12-08 Life Technologies, Inc. Genetic immunization with cationic lipids
US5627159A (en) * 1994-10-27 1997-05-06 Life Technologies, Inc. Enhancement of lipid cationic transfections in the presence of serum
US6051429A (en) 1995-06-07 2000-04-18 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US6126964A (en) 1996-01-04 2000-10-03 Mirus Corporation Process of making a compound by forming a polymer from a template drug
WO1997042819A1 (en) 1996-05-13 1997-11-20 Alberto Haces Cationic lipids for transfection of negatively charged or neutral molecules into living cells
EP0953041A4 (en) * 1996-08-30 2003-01-29 Life Technologies Inc Serum-free mammalian cell culture medium, and uses thereof
CA2217550A1 (en) 1996-10-22 1998-04-22 F. Hoffmann-La Roche Ag Cationic lipids for gene therapy
DE69735382T2 (en) 1996-11-04 2006-11-30 Qiagen Gmbh CATIONIC REAGENTS FOR TRANSFECTION
AU1987197A (en) 1997-03-10 1998-09-29 Heather Lynn Davis Gene delivery to mucosal epithelium for immunization or therapeutic purpose
GB9726073D0 (en) 1997-12-09 1998-02-04 Smithkline Beecham Plc Novel compounds
US7091192B1 (en) * 1998-07-01 2006-08-15 California Institute Of Technology Linear cyclodextrin copolymers
EP1129064B1 (en) 1998-11-12 2008-01-09 Invitrogen Corporation Transfection reagents
JP4972261B2 (en) 1999-08-27 2012-07-11 ライフ テクノロジーズ コーポレーション Metal binding compounds and their use in cell culture media compositions
US20030096414A1 (en) 2001-03-27 2003-05-22 Invitrogen Corporation Culture medium for cell growth and transfection
AU2002336367A1 (en) * 2001-08-16 2003-03-03 The Regents Of The University Of Michigan Adamts13 genes and proteins and variants, and uses thereof
GB2459145A (en) 2008-04-10 2009-10-14 Spit N Polish Shoe Shine Compa Collapsible booth
US9718255B2 (en) 2011-11-03 2017-08-01 Barson Composites Corporation Corrosion-resistant diffusion coatings
US9926825B2 (en) 2016-04-19 2018-03-27 GM Global Technology Operations LLC Method and apparatus for exhaust purification for an internal combustion engine

Patent Citations (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2695314A (en) * 1951-08-25 1954-11-23 Monsanto Chemicals Hydroxyethylated-n-keryl alkylenediamines
US2901461A (en) * 1955-05-05 1959-08-25 Union Carbide Corp Curable glycidyl polyether-polyamine compositions
US3152188A (en) * 1960-11-15 1964-10-06 Nalco Chemical Co Nu-substituted polyalkylene polyamines having 2-hydroxyethyl and mono-secondary-hydroxyl hydrocarbon substituents
US4673649A (en) * 1983-07-15 1987-06-16 University Patents, Inc. Process and defined medium for growth of human epidermal keratinocyte cells
US4940666A (en) * 1983-07-15 1990-07-10 University Patents, Inc. Process and defined medium for growth of human epidermal keratinocyte cells
US5545412A (en) * 1985-01-07 1996-08-13 Syntex (U.S.A.) Inc. N-[1, (1-1)-dialkyloxy]-and N-[1, (1-1)-dialkenyloxy]-alk-1-yl-n,n,n-tetrasubstituted ammonium lipids and uses therefor
US4897355A (en) * 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US5208036A (en) * 1985-01-07 1993-05-04 Syntex (U.S.A.) Inc. N-(ω, (ω-1)-dialkyloxy)- and N-(ω, (ω-1)-dialkenyloxy)-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US5550289A (en) * 1985-01-07 1996-08-27 Syntex (U.S.A.) Inc. N-(1,(1-1)-dialkyloxy)-and N-(1,(1-1)-dialkenyloxy alk-1-yl-N-N,N-tetrasubstituted ammonium lipids and uses therefor
US5045454A (en) * 1987-01-09 1991-09-03 Medi-Cult A/S Serum-free growth medium and use thereof
US5232848A (en) * 1987-03-24 1993-08-03 W. R. Grace & Co.-Conn. Basal nutrient medium for cell culture
US5118513A (en) * 1987-07-02 1992-06-02 The Procter & Gamble Company Method for enhancing bioavailability of iron-calcium mineral supplements
US5135866A (en) * 1989-03-03 1992-08-04 W. R. Grace & Co.-Conn. Very low protein nutrient medium for cell culture
US5703055A (en) * 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5589466A (en) * 1989-03-21 1996-12-31 Vical Incorporated Induction of a protective immune response in a mammal by injecting a DNA sequence
US5171678A (en) * 1989-04-17 1992-12-15 Centre National De La Recherche Scientifique Lipopolyamines, their preparation and their use
US5279833A (en) * 1990-04-04 1994-01-18 Yale University Liposomal transfection of nucleic acids into animal cells
US5459127A (en) * 1990-04-19 1995-10-17 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5474931A (en) * 1991-06-17 1995-12-12 Life Technologies, Inc. Media concentrate technology
US5166066A (en) * 1991-07-11 1992-11-24 The Upjohn Company Transformed cells comprising GABAA receptors
US5283185A (en) * 1991-08-28 1994-02-01 University Of Tennessee Research Corporation Method for delivering nucleic acids into cells
US5455335A (en) * 1991-12-13 1995-10-03 The Trustees Of Princeton University Pharmaceutical compositions and methods of transporting compounds across biological membrane
US5667774A (en) * 1992-08-20 1997-09-16 E.I. Du Pont De Nemours And Company Crosslinked polymeric ammonium salts
US5334761A (en) * 1992-08-28 1994-08-02 Life Technologies, Inc. Cationic lipids
US6506604B2 (en) * 1993-06-11 2003-01-14 Cell Genesys, Inc. Method for production of high titer virus and high efficiency retroviral mediated transduction of mammalian cells
US5674908A (en) * 1993-12-20 1997-10-07 Life Technologies, Inc. Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US5834439A (en) * 1993-12-20 1998-11-10 Life Technologies, Inc. Highly packed polycationic ammonium, sulfonium and phosphonium lipids
US5793566A (en) * 1994-01-10 1998-08-11 Micropolis Corporation Self securing compliant gasket for a disk drive assembly housing
US5908635A (en) * 1994-08-05 1999-06-01 The United States Of America As Represented By The Department Of Health And Human Services Method for the liposomal delivery of nucleic acids
US5785992A (en) * 1994-09-30 1998-07-28 Inex Pharmaceuticals Corp. Compositions for the introduction of polyanionic materials into cells
US5753613A (en) * 1994-09-30 1998-05-19 Inex Pharmaceuticals Corporation Compositions for the introduction of polyanionic materials into cells
US5948767A (en) * 1994-12-09 1999-09-07 Genzyme Corporation Cationic amphiphile/DNA complexes
US5783565A (en) * 1994-12-09 1998-07-21 Genzyme Corporation Cationic amphiphiles containing spermine or spermidine cationic group for intracellular delivery of therapeutic molecules
US5840710A (en) * 1994-12-09 1998-11-24 Genzyme Corporation Cationic amphiphiles containing ester or ether-linked lipophilic groups for intracellular delivery of therapeutic molecules
US5719131A (en) * 1994-12-09 1998-02-17 Genzyme Corporation Cationic amphiphiles containing dialkylamine lipophilic groups for intracellular delivery of therapeutic molecules
US5795587A (en) * 1995-01-23 1998-08-18 University Of Pittsburgh Stable lipid-comprising drug delivery complexes and methods for their production
US5830430A (en) * 1995-02-21 1998-11-03 Imarx Pharmaceutical Corp. Cationic lipids and the use thereof
US5736392A (en) * 1995-06-07 1998-04-07 Life Technologies, Inc. Peptide-enhanced cationic lipid transfections
US5705385A (en) * 1995-06-07 1998-01-06 Inex Pharmaceuticals Corporation Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
US5830878A (en) * 1995-06-07 1998-11-03 Megabios Corporation Cationic lipid: DNA complexes for gene targeting
US5744335A (en) * 1995-09-19 1998-04-28 Mirus Corporation Process of transfecting a cell with a polynucleotide mixed with an amphipathic compound and a DNA-binding protein
US5962533A (en) * 1996-02-06 1999-10-05 University Of Florida Research Foundation, Inc. Hydroxy polyamines
US5783566A (en) * 1996-05-10 1998-07-21 California Institute Of Technology Method for increasing or decreasing transfection efficiency
US5935936A (en) * 1996-06-03 1999-08-10 Genzyme Corporation Compositions comprising cationic amphiphiles and co-lipids for intracellular delivery of therapeutic molecules
US6103529A (en) * 1996-10-10 2000-08-15 Life Technologies, Inc. Animal cell culture media comprising peptides derived from rice
US5713055A (en) * 1996-11-25 1998-01-27 Eastman Kodak Company Ambient light sensitive automatic flash control circuit
US5837283A (en) * 1997-03-12 1998-11-17 The Regents Of The University Of California Cationic lipid compositions targeting angiogenic endothelial cells
US5948925A (en) * 1997-05-06 1999-09-07 Genzyme Corporation Cationic amphiphiles containing linkers derived from neutral or positively charged amino acids
US6093564A (en) * 1997-10-03 2000-07-25 V.I. Technologies, Inc. Methods and compositions for the selective modification of nucleic acids
US20050118253A1 (en) * 1998-02-03 2005-06-02 Protiva Biotherapeutics, Inc. Systemic delivery of serum stable plasmid lipid particles for cancer therapy
US7026454B1 (en) * 1998-02-17 2006-04-11 University Of Kansas Medical Center Neuroendocrine marker of prostate cancer and method for producing same
US20050100940A1 (en) * 1998-03-02 2005-05-12 Millennium Pharmaceuticals, Inc. Novel FDRG protein and nucleic acid molecules and uses therefor
US6171862B1 (en) * 1998-03-31 2001-01-09 The Regents Of The University Of California Transfection in serum-containing media
US20070025962A1 (en) * 1998-06-25 2007-02-01 Eaton Dan L Interleukin-8 homologous polypeptides and therapeutic uses thereof
US6767741B1 (en) * 1999-08-27 2004-07-27 Invitrogen Corporation Metal binding compounds and their use in cell culture medium compositions
US6544790B1 (en) * 1999-09-17 2003-04-08 Whitehead Institute For Biomedical Research Reverse transfection method
US6541225B1 (en) * 2000-01-26 2003-04-01 Raven Biotechnologies, Inc. Methods and compositions for generating human monoclonal antibodies
US20030032203A1 (en) * 2001-07-10 2003-02-13 Sabatini David M. Small molecule microarrays
US20040038862A1 (en) * 2001-07-30 2004-02-26 Goodwin Bryan James Identification of new therapeutic targets for modulating bile acid synthesis

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Technical Resources - Media Formulations 11320 - DMEM/F-12, http://www.invitrogen.com/site/us/en/home/support/Product-Technical-Resources/media_formulation.55.html, downloaded 5/6/13 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9879243B2 (en) 2001-03-27 2018-01-30 Lifetechnologies Corporation Culture medium for cell growth and transfection
WO2009140701A2 (en) * 2008-05-16 2009-11-19 Synthetic Genomics, Inc. Delivery into cells using ultra-short pulse lasers
WO2009140701A3 (en) * 2008-05-16 2010-01-07 Synthetic Genomics, Inc. Delivery into cells using ultra-short pulse lasers
US20090311236A1 (en) * 2008-06-11 2009-12-17 Immune @Work, Inc. Therapeutic Peptide Compositions And Methods Of Making And Using Same
WO2010031074A2 (en) 2008-09-15 2010-03-18 Genentech, Inc. Compositions and methods for regulating cell osmolarity
EP3165600A1 (en) 2010-04-23 2017-05-10 Life Technologies Corporation Cell culture medium comprising small peptides
DE202011110859U1 (en) 2010-04-23 2016-12-20 Life Technologies Corporation Small peptides comprising cell culture medium
EP3168295A1 (en) 2010-04-23 2017-05-17 Life Technologies Corporation Cell culture medium comprising small peptides
EP4170016A1 (en) 2010-04-23 2023-04-26 Life Technologies Corporation Cell culture medium comprising small peptides
US10066000B2 (en) 2012-05-02 2018-09-04 Life Technologies Corporation High yield transient expression in mammalian cells using unique pairing of high density growth and transfection medium and expression enhancers
US11498949B2 (en) 2012-05-02 2022-11-15 Life Technologies Corporation System and method for high-yield transient expression in mammalian cells
WO2014110433A1 (en) * 2013-01-10 2014-07-17 Biogen Idec Ma Inc. Medium supplements for improved process performance
WO2019099891A1 (en) 2017-11-16 2019-05-23 Life Technologies Corporation Streamlined methods for making liquid media

Also Published As

Publication number Publication date
EP2351827A1 (en) 2011-08-03
US20030096414A1 (en) 2003-05-22
EP1385933B1 (en) 2012-07-11
US20140057335A1 (en) 2014-02-27
CA2441910A1 (en) 2002-10-03
US9879243B2 (en) 2018-01-30
WO2002077202A1 (en) 2002-10-03
EP1385933A1 (en) 2004-02-04
EP1385933A4 (en) 2004-05-19
JP2005506048A (en) 2005-03-03

Similar Documents

Publication Publication Date Title
US9879243B2 (en) Culture medium for cell growth and transfection
US20230130038A1 (en) System and method for high-yield transient expression in mammalian cells
US20220195478A1 (en) System and Method for Improved Transient Protein Expression in CHO Cells
WO1998008934A9 (en) Serum-free mammalian cell culture medium, and uses thereof
AU2002254372A1 (en) Culture medium for cell growth and transfection
DK2243827T3 (en) Serum-free culture medium for mammalian cells and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: BANK OF AMERICA, N.A., AS COLLATERAL AGENT, WASHIN

Free format text: SECURITY AGREEMENT;ASSIGNOR:LIFE TECHNOLOGIES CORPORATION;REEL/FRAME:021975/0467

Effective date: 20081121

Owner name: BANK OF AMERICA, N.A., AS COLLATERAL AGENT,WASHING

Free format text: SECURITY AGREEMENT;ASSIGNOR:LIFE TECHNOLOGIES CORPORATION;REEL/FRAME:021975/0467

Effective date: 20081121

AS Assignment

Owner name: LIFE TECHNOLOGIES CORPORATION, CALIFORNIA

Free format text: LIEN RELEASE;ASSIGNOR:BANK OF AMERICA, N.A.;REEL/FRAME:030182/0461

Effective date: 20100528

STCB Information on status: application discontinuation

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